1
|
Takasaki T, Hamabe Y, Touchi K, Khandakar GI, Ueda T, Okada H, Sakai K, Nishio K, Tanabe G, Sugiura R. ACA-28, an ERK MAPK Signaling Modulator, Exerts Anticancer Activity through ROS Induction in Melanoma and Pancreatic Cancer Cells. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2024; 2024:7683793. [PMID: 38500550 PMCID: PMC10948229 DOI: 10.1155/2024/7683793] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/05/2023] [Revised: 02/19/2024] [Accepted: 02/27/2024] [Indexed: 03/20/2024]
Abstract
The extracellular signal-regulated kinase (ERK) MAPK pathway is dysregulated in various human cancers and is considered an attractive therapeutic target for cancer. Therefore, several inhibitors of this pathway are being developed, and some are already used in the clinic. We have previously identified an anticancer compound, ACA-28, with a unique property to preferentially induce ERK-dependent apoptosis in melanoma cells. To comprehensively understand the biological cellular impact induced by ACA-28, we performed a global gene expression analysis of human melanoma SK-MEL-28 cells exposed to ACA-28 using a DNA microarray. The transcriptome analysis identified nuclear factor erythroid 2-related factor 2 (Nrf2), a master transcription factor that combats oxidative stress, as the most upregulated genetic pathway after ACA-28 treatment. Consistently, ACA-28 showed properties to increase the levels of reactive oxygen species (ROS) as well as Nrf2 protein, which is normally repressed by proteasomal degradation and activated in response to oxidative stresses. Furthermore, the ROS scavenger N-acetyl cysteine significantly attenuated the anticancer activity of ACA-28. Thus, ACA-28 activates Nrf2 signaling and exerts anticancer activity partly via its ROS-stimulating property. Interestingly, human A549 cancer cells with constitutively high levels of Nrf2 protein showed resistance to ACA-28, as compared with SK-MEL-28. Transient overexpression of Nrf2 also increased the resistance of cells to ACA-28, while knockdown of Nrf2 exerted the opposite effect. Thus, upregulation of Nrf2 signaling protects cancer cells from ACA-28-mediated cell death. Notably, the Nrf2 inhibitor ML385 substantially enhanced the cell death-inducing property of ACA-28 in pancreatic cancer cells, T3M4 and PANC-1. Our data suggest that Nrf2 plays a key role in determining cancer cell susceptibility to ACA-28 and provides a novel strategy for cancer therapy to combine the Nrf2 inhibitor and ACA-28.
Collapse
Affiliation(s)
- Teruaki Takasaki
- Laboratory of Molecular Pharmacogenomics, Department of Pharmaceutical Sciences, Faculty of Pharmacy, Kindai University, Osaka 577-8502, Japan
| | - Yasuyuki Hamabe
- Laboratory of Molecular Pharmacogenomics, Department of Pharmaceutical Sciences, Faculty of Pharmacy, Kindai University, Osaka 577-8502, Japan
| | - Kenta Touchi
- Laboratory of Molecular Pharmacogenomics, Department of Pharmaceutical Sciences, Faculty of Pharmacy, Kindai University, Osaka 577-8502, Japan
| | - Golam Iftakhar Khandakar
- Laboratory of Molecular Pharmacogenomics, Department of Pharmaceutical Sciences, Faculty of Pharmacy, Kindai University, Osaka 577-8502, Japan
| | - Takeshi Ueda
- Department of Biochemistry, Faculty of Medicine, Kindai University, Osaka 589-8511, Japan
- Anti-Aging Center, Kindai University, Osaka 577-8502, Japan
| | - Hitoshi Okada
- Department of Biochemistry, Faculty of Medicine, Kindai University, Osaka 589-8511, Japan
- Anti-Aging Center, Kindai University, Osaka 577-8502, Japan
| | - Kazuko Sakai
- Department of Genome Biology, Faculty of Medicine, Kindai University, Osaka 589-8511, Japan
| | - Kazuto Nishio
- Department of Genome Biology, Faculty of Medicine, Kindai University, Osaka 589-8511, Japan
| | - Genzoh Tanabe
- Laboratory of Organic Chemistry, Department of Pharmacy, Faculty of Pharmacy, Kindai University, Osaka 577-8502, Japan
| | - Reiko Sugiura
- Laboratory of Molecular Pharmacogenomics, Department of Pharmaceutical Sciences, Faculty of Pharmacy, Kindai University, Osaka 577-8502, Japan
- Anti-Aging Center, Kindai University, Osaka 577-8502, Japan
| |
Collapse
|
2
|
Fujii T, Nakano Y, Hagita D, Onishi N, Endo A, Nakagawa M, Yoshiura T, Otsuka Y, Takeuchi S, Suzuki M, Shimizu Y, Toyooka T, Matsushita Y, Hibiya Y, Tomura S, Kondo A, Wada K, Ichimura K, Tomiyama A. KLC1-ROS1 Fusion Exerts Oncogenic Properties of Glioma Cells via Specific Activation of JAK-STAT Pathway. Cancers (Basel) 2023; 16:9. [PMID: 38201436 PMCID: PMC10778328 DOI: 10.3390/cancers16010009] [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: 11/07/2023] [Revised: 12/12/2023] [Accepted: 12/13/2023] [Indexed: 01/12/2024] Open
Abstract
Here, we investigated the detailed molecular oncogenic mechanisms of a novel receptor tyrosine kinase (RTK) fusion, KLC1-ROS1, with an adapter molecule, KLC1, and an RTK, ROS1, discovered in pediatric glioma, and we explored a novel therapeutic target for glioma that possesses oncogenic RTK fusion. When wild-type ROS1 and KLC1-ROS1 fusions were stably expressed in the human glioma cell lines A172 and U343MG, immunoblotting revealed that KLC1-ROS1 fusion specifically activated the JAK2-STAT3 pathway, a major RTK downstream signaling pathway, when compared with wild-type ROS1. Immunoprecipitation of the fractionated cell lysates revealed a more abundant association of the KLC1-ROS1 fusion with JAK2 than that observed for wild-type ROS1 in the cytosolic fraction. A mutagenesis study of the KLC1-ROS1 fusion protein demonstrated the fundamental roles of both the KLC1 and ROS1 domains in the constitutive activation of KLC1-ROS1 fusion. Additionally, in vitro assays demonstrated that KLC1-ROS1 fusion upregulated cell proliferation, invasion, and chemoresistance when compared to wild-type ROS1. Combination treatment with the chemotherapeutic agent temozolomide and an inhibitor of ROS1, JAK2, or a downstream target of STAT3, demonstrated antitumor effects against KLC1-ROS1 fusion-expressing glioma cells. Our results demonstrate that KLC1-ROS1 fusion exerts oncogenic activity through serum-independent constitutive activation, resulting in specific activation of the JAK-STAT pathway. Our data suggested that molecules other than RTKs may serve as novel therapeutic targets for RTK fusion in gliomas.
Collapse
Affiliation(s)
- Takashi Fujii
- Department of Brain Disease Translational Research, Juntendo University Faculty of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo 113-8421, Japan; (T.F.); (D.H.); (Y.M.); (Y.H.); (K.I.)
- Department of Neurosurgery, National Defense Medical College, 3-2 Namiki, Tokorozawa 359-8513, Saitama, Japan; (A.E.); (M.N.); (T.Y.); (Y.O.); (S.T.); (T.T.); (K.W.)
- Department of Neurosurgery, Juntendo University School of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo 113-8421, Japan; (M.S.); (Y.S.); (A.K.)
| | - Yoshiko Nakano
- Department of Pediatrics, The University of Tokyo Hospital, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8655, Japan;
| | - Daichi Hagita
- Department of Brain Disease Translational Research, Juntendo University Faculty of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo 113-8421, Japan; (T.F.); (D.H.); (Y.M.); (Y.H.); (K.I.)
- Department of Neurosurgery, National Defense Medical College, 3-2 Namiki, Tokorozawa 359-8513, Saitama, Japan; (A.E.); (M.N.); (T.Y.); (Y.O.); (S.T.); (T.T.); (K.W.)
- Department of Neurosurgery, Juntendo University School of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo 113-8421, Japan; (M.S.); (Y.S.); (A.K.)
| | - Nobuyuki Onishi
- Department of Clinical Diagnostic Oncology, Clinical Research Institute for Clinical Pharmacology and Therapeutics, Showa University, 1-5-8 Hatanodai, Shinagawa-ku, Tokyo 142-8555, Japan;
| | - Arumu Endo
- Department of Neurosurgery, National Defense Medical College, 3-2 Namiki, Tokorozawa 359-8513, Saitama, Japan; (A.E.); (M.N.); (T.Y.); (Y.O.); (S.T.); (T.T.); (K.W.)
| | - Masaya Nakagawa
- Department of Neurosurgery, National Defense Medical College, 3-2 Namiki, Tokorozawa 359-8513, Saitama, Japan; (A.E.); (M.N.); (T.Y.); (Y.O.); (S.T.); (T.T.); (K.W.)
| | - Toru Yoshiura
- Department of Neurosurgery, National Defense Medical College, 3-2 Namiki, Tokorozawa 359-8513, Saitama, Japan; (A.E.); (M.N.); (T.Y.); (Y.O.); (S.T.); (T.T.); (K.W.)
| | - Yohei Otsuka
- Department of Neurosurgery, National Defense Medical College, 3-2 Namiki, Tokorozawa 359-8513, Saitama, Japan; (A.E.); (M.N.); (T.Y.); (Y.O.); (S.T.); (T.T.); (K.W.)
| | - Satoru Takeuchi
- Department of Neurosurgery, National Defense Medical College, 3-2 Namiki, Tokorozawa 359-8513, Saitama, Japan; (A.E.); (M.N.); (T.Y.); (Y.O.); (S.T.); (T.T.); (K.W.)
| | - Mario Suzuki
- Department of Neurosurgery, Juntendo University School of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo 113-8421, Japan; (M.S.); (Y.S.); (A.K.)
| | - Yuzaburo Shimizu
- Department of Neurosurgery, Juntendo University School of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo 113-8421, Japan; (M.S.); (Y.S.); (A.K.)
| | - Terushige Toyooka
- Department of Neurosurgery, National Defense Medical College, 3-2 Namiki, Tokorozawa 359-8513, Saitama, Japan; (A.E.); (M.N.); (T.Y.); (Y.O.); (S.T.); (T.T.); (K.W.)
| | - Yuko Matsushita
- Department of Brain Disease Translational Research, Juntendo University Faculty of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo 113-8421, Japan; (T.F.); (D.H.); (Y.M.); (Y.H.); (K.I.)
- Department of Neurosurgery, Juntendo University School of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo 113-8421, Japan; (M.S.); (Y.S.); (A.K.)
| | - Yuko Hibiya
- Department of Brain Disease Translational Research, Juntendo University Faculty of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo 113-8421, Japan; (T.F.); (D.H.); (Y.M.); (Y.H.); (K.I.)
- Department of Neurosurgery, Juntendo University School of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo 113-8421, Japan; (M.S.); (Y.S.); (A.K.)
| | - Satoshi Tomura
- Division of Traumatology, Research Institute, National Defense Medical College, 3-2 Namiki, Tokorozawa 359-8513, Saitama, Japan;
| | - Akihide Kondo
- Department of Neurosurgery, Juntendo University School of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo 113-8421, Japan; (M.S.); (Y.S.); (A.K.)
| | - Kojiro Wada
- Department of Neurosurgery, National Defense Medical College, 3-2 Namiki, Tokorozawa 359-8513, Saitama, Japan; (A.E.); (M.N.); (T.Y.); (Y.O.); (S.T.); (T.T.); (K.W.)
| | - Koichi Ichimura
- Department of Brain Disease Translational Research, Juntendo University Faculty of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo 113-8421, Japan; (T.F.); (D.H.); (Y.M.); (Y.H.); (K.I.)
- Department of Neurosurgery, Juntendo University School of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo 113-8421, Japan; (M.S.); (Y.S.); (A.K.)
| | - Arata Tomiyama
- Department of Brain Disease Translational Research, Juntendo University Faculty of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo 113-8421, Japan; (T.F.); (D.H.); (Y.M.); (Y.H.); (K.I.)
- Department of Neurosurgery, National Defense Medical College, 3-2 Namiki, Tokorozawa 359-8513, Saitama, Japan; (A.E.); (M.N.); (T.Y.); (Y.O.); (S.T.); (T.T.); (K.W.)
- Department of Neurosurgery, Juntendo University School of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo 113-8421, Japan; (M.S.); (Y.S.); (A.K.)
| |
Collapse
|
3
|
Guo T, Wu C, Zhang J, Yu J, Li G, Jiang H, Zhang X, Yu R, Liu X. Dual blockade of EGFR and PI3K signaling pathways offers a therapeutic strategy for glioblastoma. Cell Commun Signal 2023; 21:363. [PMID: 38115126 PMCID: PMC10729576 DOI: 10.1186/s12964-023-01400-0] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2023] [Accepted: 11/14/2023] [Indexed: 12/21/2023] Open
Abstract
BACKGROUND Glioblastoma multiforme (GBM) is a devastating disease that lacks effective drugs for targeted therapy. Previously, we found that the third-generation epidermal growth factor receptor (EGFR) inhibitor AZD-9291 persistently blocked the activation of the ERK pathway but had no inhibitory effect on the phosphoinositide 3-kinase (PI3K)/Akt pathway. Given that the PI3K inhibitor GDC-0084 is being evaluated in phase I/II clinical trials of GBM treatment, we hypothesized that combined inhibition of the EGFR/ERK and PI3K/Akt pathways may have a synergistic effect in the treatment of GBM. METHODS The synergistic effects of cotreatment with AZD-9291 and GDC-0084 were validated using cell viability assays in GBM and primary GBM cell lines. Moreover, the underlying inhibitory mechanisms were assessed through colony formation, EdU proliferation, and cell cycle assays, as well as RNA-seq analyses and western blot. The therapeutic effects of the drug combination on tumor growth and survival were investigated in mice bearing tumors using subcutaneously or intracranially injected LN229 xenografts. RESULTS Combined treatment with AZD-9291 and GDC-0084 synergistically inhibited the proliferation and clonogenic survival, as well as induced cell cycle arrest of GBM cells and primary GBM cells, compared to monotherapy. Moreover, AZD-9291 plus GDC-0084 combination therapy significantly inhibited the growth of subcutaneous tumors and orthotopic brain tumor xenografts, thus prolonging the survival of tumor-bearing mice. More importantly, the combination of AZD-9291 and GDC-0084 simultaneously blocked the activation of the EGFR/MEK/ERK and PI3K/AKT/mTOR signaling pathways, thereby exerting significant antitumor activity. CONCLUSION Our findings demonstrate that the combined blockade of the EGFR/MEK/ERK and PI3K/AKT/mTOR pathways is more effective against GBM than inhibition of each pathway alone, both in vitro and in vivo. Our results suggest that AZD-9291 combined with GDC-0084 may be considered as a potential treatment strategy in future clinical trials. Video Abstract.
Collapse
Affiliation(s)
- Tongxuan Guo
- Insititute of Nervous System Diseases, Xuzhou Medical University, Xuzhou, Jiangsu, China
- Department of Neurosurgery, Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Changyong Wu
- Insititute of Nervous System Diseases, Xuzhou Medical University, Xuzhou, Jiangsu, China
- Department of Neurosurgery, Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Junhao Zhang
- Insititute of Nervous System Diseases, Xuzhou Medical University, Xuzhou, Jiangsu, China
- Department of Neurosurgery, Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Jiefeng Yu
- Insititute of Nervous System Diseases, Xuzhou Medical University, Xuzhou, Jiangsu, China
- Department of Neurosurgery, Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Guoxi Li
- Insititute of Nervous System Diseases, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Hongyan Jiang
- Insititute of Nervous System Diseases, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Xu Zhang
- Insititute of Nervous System Diseases, Xuzhou Medical University, Xuzhou, Jiangsu, China.
- Department of Neurosurgery, Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China.
| | - Rutong Yu
- Insititute of Nervous System Diseases, Xuzhou Medical University, Xuzhou, Jiangsu, China.
- Department of Neurosurgery, Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China.
| | - Xuejiao Liu
- Insititute of Nervous System Diseases, Xuzhou Medical University, Xuzhou, Jiangsu, China.
- Department of Neurosurgery, Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China.
| |
Collapse
|
4
|
Hong J, Zheng W, Cai X. Small-molecule high-throughput screening identifies a MEK inhibitor PD1938306 that enhances sorafenib efficacy via MCL-1 and BIM in hepatocellular carcinoma cells. Comb Chem High Throughput Screen 2022; 26:1364-1374. [PMID: 36043792 PMCID: PMC9971357 DOI: 10.2174/1386207325666220830145026] [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: 02/06/2022] [Revised: 07/03/2022] [Accepted: 07/07/2022] [Indexed: 11/22/2022]
Abstract
Background Sorafenib is the most widely used systematic therapy drug for treating unresectable hepatocellular carcinoma (HCC) but showed dissatisfactory efficacy in clinical applications. Objective We conducted a combinational quantitative small-molecule high-throughput screening (qHTS) to identify potential candidates to enhance the treatment effectiveness of sorafenib. Methods First, using a Hep3B human HCC cell line, 7051 approved drugs and bioactive compounds were screened, then the primary hits were tested with/ without 0.5 μM sorafenib respectively, the compound has the half maximal inhibitory concentration (IC50) shift value greater than 1.5 was thought to have the synergistic effect with sorafenib. Furthermore, the MEK inhibitor PD198306 was selected for further mechanistic study. Results 12 effective compounds were identified, including kinase inhibitors that target MEK, AURKB, CAMK, ROCK2, BRAF, PI3K, AKT and EGFR, as well as a μ-opioid receptor agonist and a L-type calcium channel blocker. The mechanistic research of the combination of sorafenib plus PD198306 showed that the two compounds synergistically inhibited MEK-ERK and mTORC1-4EBP1, and induced apoptosis in HCC cells, which can be attributed to the transcriptional and posttranslational regulation of MCL-1 and BIM. Conclusion Small-molecule qHTS identifies MEK inhibitor PD1938306 as a potent sorafenib enhancer, together with several novel combination strategies that are valuable for further studies.
Collapse
Affiliation(s)
- Junjie Hong
- Department of General Surgery, Key Laboratory of Laparoscopic Technique Research of Zhejiang Province, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou 310016, China,National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, MD 20850, USA
| | - Wei Zheng
- National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, MD 20850, USA
| | - Xiujun Cai
- Department of General Surgery, Key Laboratory of Laparoscopic Technique Research of Zhejiang Province, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou 310016, China,Correspondence to: Xiujun Cai, 3 East Qingchun Road, Jianggan District, Hangzhou 310000, China. Tel: +86-0571-8600-6617; Fax: +86-0571-8604-4817;
| |
Collapse
|
5
|
Wu W, Klockow JL, Zhang M, Lafortune F, Chang E, Jin L, Wu Y, Daldrup-Link HE. Glioblastoma multiforme (GBM): An overview of current therapies and mechanisms of resistance. Pharmacol Res 2021; 171:105780. [PMID: 34302977 PMCID: PMC8384724 DOI: 10.1016/j.phrs.2021.105780] [Citation(s) in RCA: 342] [Impact Index Per Article: 85.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/16/2021] [Revised: 07/18/2021] [Accepted: 07/19/2021] [Indexed: 12/21/2022]
Abstract
Glioblastoma multiforme (GBM) is a WHO grade IV glioma and the most common malignant, primary brain tumor with a 5-year survival of 7.2%. Its highly infiltrative nature, genetic heterogeneity, and protection by the blood brain barrier (BBB) have posed great treatment challenges. The standard treatment for GBMs is surgical resection followed by chemoradiotherapy. The robust DNA repair and self-renewing capabilities of glioblastoma cells and glioma initiating cells (GICs), respectively, promote resistance against all current treatment modalities. Thus, durable GBM management will require the invention of innovative treatment strategies. In this review, we will describe biological and molecular targets for GBM therapy, the current status of pharmacologic therapy, prominent mechanisms of resistance, and new treatment approaches. To date, medical imaging is primarily used to determine the location, size and macroscopic morphology of GBM before, during, and after therapy. In the future, molecular and cellular imaging approaches will more dynamically monitor the expression of molecular targets and/or immune responses in the tumor, thereby enabling more immediate adaptation of tumor-tailored, targeted therapies.
Collapse
Affiliation(s)
- Wei Wu
- Department of Radiology, Molecular Imaging Program at Stanford, Stanford University, Stanford, CA 94305, USA
| | - Jessica L Klockow
- Department of Radiation Oncology, Stanford University, Stanford, CA 94305, USA
| | - Michael Zhang
- Department of Radiology, Molecular Imaging Program at Stanford, Stanford University, Stanford, CA 94305, USA; Department of Neurosurgery, Stanford University, Stanford, CA 94305, USA
| | - Famyrah Lafortune
- Department of Radiology, Molecular Imaging Program at Stanford, Stanford University, Stanford, CA 94305, USA
| | - Edwin Chang
- Department of Radiology, Molecular Imaging Program at Stanford, Stanford University, Stanford, CA 94305, USA
| | - Linchun Jin
- Lillian S. Wells Department of Neurosurgery, University of Florida, Gainesville, FL 32611, USA
| | - Yang Wu
- Department of Neuropathology, Institute of Pathology, Technical University of Munich, Munich, Bayern 81675, Germany
| | - Heike E Daldrup-Link
- Department of Radiology, Molecular Imaging Program at Stanford, Stanford University, Stanford, CA 94305, USA.
| |
Collapse
|
6
|
Karthikkeyan G, Pervaje R, Pervaje SK, Prasad TSK, Modi PK. Prevention of MEK-ERK-1/2 hyper-activation underlines the neuroprotective effect of Glycyrrhiza glabra L. (Yashtimadhu) against rotenone-induced cellular and molecular aberrations. JOURNAL OF ETHNOPHARMACOLOGY 2021; 274:114025. [PMID: 33775804 DOI: 10.1016/j.jep.2021.114025] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/13/2020] [Revised: 12/07/2020] [Accepted: 03/10/2021] [Indexed: 06/12/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Yashtimadhu choorna (powder) is prepared from the dried root of Glycyrrhiza glabra L., commonly known as licorice. The Indian Ayurvedic system classifies Yashtimadhu as a Medhya Rasayana that can enhance brain function, improves memory, and possess neuroprotective functions, which can be used against neurodegenerative diseases like Parkinson's disease (PD). AIM OF THE STUDY We aimed to decipher the neuroprotective effects of G. glabra L., i.e., Yashtimadhu, in a rotenone-induced PD model. MATERIALS AND METHODS Retinoic acid-differentiated IMR-32 cells were treated with rotenone (PD model) and Yashtimadhu, and were assessed for cellular toxicity, live-dead staining, cell cycle, oxidative stress, protein abundance, and kinase phosphorylation. RESULTS Yashtimadhu conferred protection against rotenone-induced cytotoxicity, countered cell death, reduced expression of pro-apoptotic proteins (cleaved-caspases-9, and 3, cleaved-PARP, BAX, and BAK) and increased anti-apoptotic protein, BCL-2. Rotenone-induced cell cycle re-entry (G2/M transition), was negated by Yashtimadhu and was confirmed with PCNA levels. Yashtimadhu countered rotenone-mediated activation of mitochondrial proteins involved in oxidative stress, cytochrome-C, PDHA1, and HSP60. Inhibition of rotenone-induced ERK-1/2 hyperphosphorylation prevented activation of apoptosis, which was confirmed with MEK-inhibitor, highlighted the action of Yashtimadhu via ERK-1/2 modulation. CONCLUSIONS We provide the evidence for neuroprotection conferred by G. glabra L. (Yashtimadhu) and its mechanism via inhibiting MEK-ERK-1/2 hyper-phosphorylation, prevention of mitochondrial stress, and subsequent prevention of apoptosis. The study highlights Yashtimadhu as a promising candidate with neuroprotective effects, the potential of which can be harnessed for identifying novel therapeutic targets.
Collapse
Affiliation(s)
- Gayathree Karthikkeyan
- Center for Systems Biology and Molecular Medicine, Yenepoya Research Centre, Yenepoya (Deemed to be University), Mangalore 575018, India.
| | | | - Sameera Krishna Pervaje
- Yenepoya Medical College and Hospital, Yenepoya (Deemed to be University), Mangalore 575018, India.
| | | | - Prashant Kumar Modi
- Center for Systems Biology and Molecular Medicine, Yenepoya Research Centre, Yenepoya (Deemed to be University), Mangalore 575018, India.
| |
Collapse
|
7
|
Ferrari B, Roda E, Priori EC, De Luca F, Facoetti A, Ravera M, Brandalise F, Locatelli CA, Rossi P, Bottone MG. A New Platinum-Based Prodrug Candidate for Chemotherapy and Its Synergistic Effect With Hadrontherapy: Novel Strategy to Treat Glioblastoma. Front Neurosci 2021; 15:589906. [PMID: 33828444 PMCID: PMC8019820 DOI: 10.3389/fnins.2021.589906] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Accepted: 02/08/2021] [Indexed: 12/18/2022] Open
Abstract
Glioblastoma (GBM) is the most common tumor of the central nervous system. Current therapies, often associated with severe side effects, are inefficacious to contrast the GBM relapsing forms. In trying to overcome these drawbacks, (OC-6-44)-acetatodiamminedichlorido(2-(2-propynyl)octanoato)platinum(IV), also called Pt(IV)Ac-POA, has been recently synthesized. This new prodrug bearing as axial ligand (2-propynyl)octanoic acid (POA), a histone deacetylase inhibitor, has a higher activity due to (i) its high cellular accumulation by virtue of its high lipophilicity and (ii) the inhibition of histone deacetylase, which leads to the increased exposure of nuclear DNA, permitting higher platination and promoting cancer cell death. In the present study, we investigated the effects induced by Pt(IV)Ac-POA and its potential antitumor activity in human U251 glioblastoma cell line using a battery of complementary techniques, i.e., flow cytometry, immunocytochemistry, TEM, and Western blotting analyses. In addition, the synergistic effect of Pt(IV)Ac-POA associated with the innovative oncological hadrontherapy with carbon ions was investigated, with the aim to identify the most efficient anticancer treatment combination. Our in vitro data demonstrated that Pt(IV)Ac-POA is able to induce cell death, through different pathways, at concentrations lower than those tested for other platinum analogs. In particular, an enduring Pt(IV)Ac-POA antitumor effect, persisting in long-term treatment, was demonstrated. Interestingly, this effect was further amplified by the combined exposure to carbon ion radiation. In conclusion, Pt(IV)Ac-POA represents a promising prodrug to be incorporated into the treatment regimen for GBM. Moreover, the synergistic efficacy of the combined protocol using chemotherapeutic Pt(IV)Ac-POA followed by carbon ion radiation may represent a promising approach, which may overcome some typical limitations of conventional therapeutic protocols for GBM treatment.
Collapse
Affiliation(s)
- Beatrice Ferrari
- Department of Biology and Biotechnology "L. Spallanzani", University of Pavia, Pavia, Italy
| | - Elisa Roda
- Department of Biology and Biotechnology "L. Spallanzani", University of Pavia, Pavia, Italy.,Laboratory of Clinical & Experimental Toxicology, Pavia Poison Centre, National Toxicology Information Centre, Toxicology Unit, Istituti Clinici Scientifici Maugeri IRCCS, Pavia, Italy
| | - Erica Cecilia Priori
- Department of Biology and Biotechnology "L. Spallanzani", University of Pavia, Pavia, Italy
| | - Fabrizio De Luca
- Department of Biology and Biotechnology "L. Spallanzani", University of Pavia, Pavia, Italy
| | - Angelica Facoetti
- National Center of Oncological Hadrontherapy (Fondazione CNAO), Pavia, Italy
| | - Mauro Ravera
- Department of Sciences and Technological Innovation (DiSIT), University of Piemonte Orientale "A. Avogadro", Alessandria, Italy
| | - Federico Brandalise
- Department of Fundamental Neurosciences (NEUFO), University of Geneva, Geneva, Switzerland
| | - Carlo Alessandro Locatelli
- Laboratory of Clinical & Experimental Toxicology, Pavia Poison Centre, National Toxicology Information Centre, Toxicology Unit, Istituti Clinici Scientifici Maugeri IRCCS, Pavia, Italy
| | - Paola Rossi
- Department of Biology and Biotechnology "L. Spallanzani", University of Pavia, Pavia, Italy
| | - Maria Grazia Bottone
- Department of Biology and Biotechnology "L. Spallanzani", University of Pavia, Pavia, Italy
| |
Collapse
|
8
|
Enhanced Malignant Phenotypes of Glioblastoma Cells Surviving NPe6-Mediated Photodynamic Therapy are Regulated via ERK1/2 Activation. Cancers (Basel) 2020; 12:cancers12123641. [PMID: 33291680 PMCID: PMC7761910 DOI: 10.3390/cancers12123641] [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: 10/26/2020] [Revised: 11/27/2020] [Accepted: 11/29/2020] [Indexed: 12/12/2022] Open
Abstract
To manage refractory and invasive glioblastomas (GBM)s, photodynamic therapy (PDT) using talaporfin sodium (NPe6) (NPe6-PDT) was recently approved in clinical practice. However, the molecular machineries regulating resistance against NPe6-PDT in GBMs and mechanisms underlying the changes in GBM phenotypes following NPe6-PDT remain unknown. Herein, we established an in vitro NPe6-mediated PDT model using human GBM cell lines. NPe6-PDT induced GBM cell death in a NPe6 dose-dependent manner. However, this NPe6-PDT-induced GBM cell death was not completely blocked by the pan-caspase inhibitor, suggesting NPe6-PDT induces both caspase-dependent and -independent cell death. Moreover, treatment with poly (ADP-ribose) polymerase inhibitor blocked NPe6-PDT-triggered caspase-independent GBM cell death. Next, it was also revealed resistance to re-NPe6-PDT of GBM cells and GBM stem cells survived following NPe6-PDT (NPe6-PDT-R cells), as well as migration and invasion of NPe6-PDT-R cells were enhanced. Immunoblotting of NPe6-PDT-R cells to assess the behavior of the proteins that are known to be stress-induced revealed that only ERK1/2 activation exhibited the same trend as migration. Importantly, treatment with the MEK1/2 inhibitor trametinib reversed resistance against re-NPe6-PDT and suppressed the enhanced migration and invasion of NPe6-PDT-R cells. Overall, enhanced ERK1/2 activation is suggested as a key regulator of elevated malignant phenotypes of GBM cells surviving NPe6-PDT and is therefore considered as a potential therapeutic target against GBM.
Collapse
|
9
|
Pu J, Shen J, Zhong Z, Yanling M, Gao J. KANK1 regulates paclitaxel resistance in lung adenocarcinoma A549 cells. ARTIFICIAL CELLS NANOMEDICINE AND BIOTECHNOLOGY 2020; 48:639-647. [PMID: 32064933 DOI: 10.1080/21691401.2020.1728287] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Paclitaxel (PTX), a tubulin-binding agent, is widely used and has shown good efficacy in the initial period of treatment for non-small cell lung cancer (NSCLC). However, the relatively rapid acquisition of resistance to PTX treatments that is observed in virtually all cases significantly limits its utility and remains a substantial challenge to the clinical management of NSCLC. The aim of this study was to identify candidate genes and mechanisms that might mediate acquired paclitaxel resistance. In this work, we established paclitaxel-resistant cells (A549-T) from parental cell lines by step-dose selection in vitro. Using methylation chip analysis and transcriptome sequencing, 43,426 differentially methylated genes and 2,870 differentially expressed genes are identified. Six genes (KANK1, ALDH3A1, GALNT14, PIK3R3, LRG1, WEE2), which may be related to paclitaxel resistance in lung adenocarcinoma, were identified. Among these genes, KANK1 exhibited significant differences in methylation and expression between cell lines. Since KANK1 plays an important role in the development of renal cancer and gastric cancer, we hypothesised that it may also play a role in acquired resistance in lung adenocarcinoma. Transient transfection of SiKANK1 significantly reduced the expression of KANK1, reducing apoptosis, increasing cell migration, and enhancing the tolerance of A549 cells to paclitaxel. KANK1 acts as a tumour suppressor gene, mediating the resistance of lung adenocarcinoma A549 to paclitaxel. The reduction of KANK1 expression can increase the paclitaxel resistance of non-small cell lung cancer and increase the difficulty of clinical treatment.
Collapse
Affiliation(s)
- Junyi Pu
- School of Life Sciences, Northwest University, Xi'an, China.,Department of Cardiovascular Medicine, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Jianfeng Shen
- School of Life Sciences, Northwest University, Xi'an, China
| | - Zihua Zhong
- School of Life Sciences, Northwest University, Xi'an, China
| | - Ma Yanling
- School of Life Sciences, Northwest University, Xi'an, China
| | - Jie Gao
- Department of Cardiovascular Medicine, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| |
Collapse
|
10
|
Impact of Hypoxia on Carbon Ion Therapy in Glioblastoma Cells: Modulation by LET and Hypoxia-Dependent Genes. Cancers (Basel) 2020; 12:cancers12082019. [PMID: 32718037 PMCID: PMC7464439 DOI: 10.3390/cancers12082019] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2020] [Revised: 07/15/2020] [Accepted: 07/19/2020] [Indexed: 12/26/2022] Open
Abstract
Tumor hypoxia is known to limit the efficacy of ionizing radiations, a concept called oxygen enhancement ratio (OER). OER depends on physical factors such as pO2 and linear energy transfer (LET). Biological pathways, such as the hypoxia-inducible transcription factors (HIF), might also modulate the influence of LET on OER. Glioblastoma (GB) is resistant to low-LET radiation (X-rays), due in part to the hypoxic environment in this brain tumor. Here, we aim to evaluate in vitro whether high-LET particles, especially carbon ion radiotherapy (CIRT), can overcome the contribution of hypoxia to radioresistance, and whether HIF-dependent genes, such as erythropoietin (EPO), influence GB sensitivity to CIRT. Hypoxia-induced radioresistance was studied in two human GB cells (U251, GL15) exposed to X-rays or to carbon ion beams with various LET (28, 50, 100 keV/µm), and in genetically-modified GB cells with downregulated EPO signaling. Cell survival, radiobiological parameters, cell cycle, and ERK activation were assessed under those conditions. The results demonstrate that, although CIRT is more efficient than X-rays in GB cells, hypoxia can limit CIRT efficacy in a cell-type manner that may involve differences in ERK activation. Using high-LET carbon beams, or targeting hypoxia-dependent genes such as EPO might reduce the effects of hypoxia.
Collapse
|
11
|
Polyphenolic Composition and Anti-Melanoma Activity of White Forsythia ( Abeliophyllum distichum Nakai) Organ Extracts. PLANTS 2020; 9:plants9060757. [PMID: 32560393 PMCID: PMC7356668 DOI: 10.3390/plants9060757] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/06/2020] [Revised: 06/15/2020] [Accepted: 06/15/2020] [Indexed: 12/13/2022]
Abstract
Abeliophyllum distichum Nakai, commonly called white forsythia, is a monotypic genus endemic to Korea. Although A. distichum is mainly used as an ornamental plant because of its horticultural value, recent studies have demonstrated its bioactivities, including antioxidant and anti-inflammatory activities, prompting us to investigate the potential anticancer effect of A. distichum organ extracts (leaves, fruit, and branches) against human melanoma SK-MEL-2 cells. The methanol extract of A. distichum leaves (AL) exhibited dose- and time-dependent cytotoxicities against SK-MEL-2 cells but not against HDFa human dermal fibroblasts. Based on high-performance liquid chromatography analysis, we identified 18 polyphenolic compounds from A. distichum organ extracts and suggest that differences in anticancer activity between organ extracts should be caused by different compositions of polyphenolic compounds. Additionally, the Annexin V/propidium iodide staining assay and analysis of caspase activity and expression indicated that AL induced cell death, including early and late apoptosis, as well as necrosis, by inducing the extrinsic pathway. Furthermore, we analyzed the differentially expressed genes between mock- and AL-treated cells using RNA-seq technology, suggesting that the anti-melanoma action of AL is mediated by down-regulation of the phosphoinositide 3-kinase (PI3K)/Akt signaling pathway. Taken together, these results shed light on the potential use of A. distichum as a green resource with potent anti-melanoma activity.
Collapse
|
12
|
Vališ K, Novák P. Targeting ERK-Hippo Interplay in Cancer Therapy. Int J Mol Sci 2020; 21:ijms21093236. [PMID: 32375238 PMCID: PMC7247570 DOI: 10.3390/ijms21093236] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2020] [Revised: 04/26/2020] [Accepted: 04/27/2020] [Indexed: 02/06/2023] Open
Abstract
Extracellular signal-regulated kinase (ERK) is a part of the mitogen-activated protein kinase (MAPK) signaling pathway which allows the transduction of various cellular signals to final effectors and regulation of elementary cellular processes. Deregulation of the MAPK signaling occurs under many pathological conditions including neurodegenerative disorders, metabolic syndromes and cancers. Targeted inhibition of individual kinases of the MAPK signaling pathway using synthetic compounds represents a promising way to effective anti-cancer therapy. Cross-talk of the MAPK signaling pathway with other proteins and signaling pathways have a crucial impact on clinical outcomes of targeted therapies and plays important role during development of drug resistance in cancers. We discuss cross-talk of the MAPK/ERK signaling pathway with other signaling pathways, in particular interplay with the Hippo/MST pathway. We demonstrate the mechanism of cell death induction shared between MAPK/ERK and Hippo/MST signaling pathways and discuss the potential of combination targeting of these pathways in the development of more effective anti-cancer therapies.
Collapse
Affiliation(s)
- Karel Vališ
- Correspondence: (K.V.); (P.N.); Tel.: +420-325873610 (P.N.)
| | - Petr Novák
- Correspondence: (K.V.); (P.N.); Tel.: +420-325873610 (P.N.)
| |
Collapse
|
13
|
Peng C, Sun Z, Wang L, Shu Y, He M, Ding H, Li Y, Wang X, Feng S, Li J, Wu J. Soybean antigen protein induces caspase-3/mitochondrion-regulated apoptosis in IPEC-J2 cells. FOOD AGR IMMUNOL 2019. [DOI: 10.1080/09540105.2019.1702926] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023] Open
Affiliation(s)
- Chenglu Peng
- College of Animal Science and Technology, Anhui Agricultural University, Hefei, People’s Republic of China
| | - Zhifeng Sun
- College of Animal Science and Technology, Anhui Agricultural University, Hefei, People’s Republic of China
| | - Lei Wang
- College of Animal Science and Technology, Anhui Agricultural University, Hefei, People’s Republic of China
| | - Yingshuang Shu
- College of Animal Science and Technology, Anhui Agricultural University, Hefei, People’s Republic of China
| | - Mengchu He
- College of Animal Science and Technology, Anhui Agricultural University, Hefei, People’s Republic of China
| | - Hongyan Ding
- College of Animal Science and Technology, Anhui Agricultural University, Hefei, People’s Republic of China
| | - Yu Li
- College of Animal Science and Technology, Anhui Agricultural University, Hefei, People’s Republic of China
| | - Xichun Wang
- College of Animal Science and Technology, Anhui Agricultural University, Hefei, People’s Republic of China
| | - Shibin Feng
- College of Animal Science and Technology, Anhui Agricultural University, Hefei, People’s Republic of China
| | - Jinchun Li
- College of Animal Science and Technology, Anhui Agricultural University, Hefei, People’s Republic of China
| | - Jinjie Wu
- College of Animal Science and Technology, Anhui Agricultural University, Hefei, People’s Republic of China
| |
Collapse
|
14
|
Wang G, Xiao L, Wang F, Yang J, Yang L, Zhao Y, Jin W. Hypoxia inducible factor-1α/B-cell lymphoma 2 signaling impacts radiosensitivity of H1299 non-small cell lung cancer cells in a normoxic environment. RADIATION AND ENVIRONMENTAL BIOPHYSICS 2019; 58:439-448. [PMID: 31203382 DOI: 10.1007/s00411-019-00802-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/09/2018] [Accepted: 06/10/2019] [Indexed: 06/09/2023]
Abstract
Hypoxia inducible factor-1α (HIF-1α) is a critical transcriptional factor for the response of cells to hypoxic microenvironment and its expression induces resistance of hypoxic non-small-cell lung cancer (NSCLC) cells to radiotherapy. This study investigated how the activation of HIF-1α/B-cell lymphoma 2 (BCL-2) signaling under normoxic conditions impacted radiosensitivity of NSCLC cells. The recombinant pcDNA3.0-EGFP plasmids with wild-type or mutant HIF-1α complementary DNA (cDNA) were transfected into H1299 cells, an NSCLC cell line, establishing two H1299 sublines with high expression of HIF-1α. Compared with the levels of HIF-1α and BCL-2 proteins in non-transfected cells, increased levels of both proteins were found in transfected cells. Moreover, the expression of HIF-1α in non-transfected cells induced by chloride cobalt (CoCl2), a commonly used mimetic hypoxia reagent, was concomitant with the enhancement of BCL-2 expression. Conversely, reduction of HIF-1α expression by an inhibitor decreased the levels of BCL-2 proteins. The results revealed that the stabilization and expression of HIF-1α promoted the accumulation of BCL-2 proteins in H1299 cells. Subsequent experiments showed that intracellular HIF-1α/BCL-2 signaling was triggered in a normoxic environment after H1299 cells were exposed to irradiation, causing an elevated radioresistance. In contrast, blockage of HIF-1α/BCL-2 signaling leads to an elevated radiosensitivity. Proliferation of cells assay showed that, under normoxic conditions, population doubling times (PDTs) of irradiated cells were prolonged by suppression of HIF-1α/BCL-2 signaling. It is, therefore, indicated that HIF-1α/BCL-2 signaling activated by ionizing radiation reduces the radiosensitivity of H1299 cells independent of the hypoxic environment.
Collapse
Affiliation(s)
- Gang Wang
- Teaching and Research Section of Nuclear Medicine, Anhui Medical University, 81 Meishan Road, Hefei, 230032, Anhui, People's Republic of China
| | - Liang Xiao
- Teaching and Research Section of Nuclear Medicine, Anhui Medical University, 81 Meishan Road, Hefei, 230032, Anhui, People's Republic of China
- Department of Radiation Oncology, First Affiliated Hospital, Anhui Medical University, 218 Jixi Road, Hefei, 230022, Anhui, People's Republic of China
| | - Fen Wang
- Teaching and Research Section of Nuclear Medicine, Anhui Medical University, 81 Meishan Road, Hefei, 230032, Anhui, People's Republic of China
| | - Jing Yang
- Teaching and Research Section of Nuclear Medicine, Anhui Medical University, 81 Meishan Road, Hefei, 230032, Anhui, People's Republic of China
- Department of Radiation Oncology, Anhui Provincial Cancer Hospital, 107 Huanhu East Road, Hefei, 230031, Anhui, People's Republic of China
| | - Li Yang
- Teaching and Research Section of Nuclear Medicine, Anhui Medical University, 81 Meishan Road, Hefei, 230032, Anhui, People's Republic of China
| | - Ye Zhao
- Teaching and Research Section of Nuclear Medicine, Anhui Medical University, 81 Meishan Road, Hefei, 230032, Anhui, People's Republic of China.
| | - Wensen Jin
- Teaching and Research Section of Nuclear Medicine, Anhui Medical University, 81 Meishan Road, Hefei, 230032, Anhui, People's Republic of China.
| |
Collapse
|
15
|
Tau binding protein CAPON induces tau aggregation and neurodegeneration. Nat Commun 2019; 10:2394. [PMID: 31160584 PMCID: PMC6546774 DOI: 10.1038/s41467-019-10278-x] [Citation(s) in RCA: 52] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2018] [Accepted: 04/24/2019] [Indexed: 02/07/2023] Open
Abstract
To understand the molecular processes that link Aβ amyloidosis, tauopathy and neurodegeneration, we screened for tau-interacting proteins by immunoprecipitation/LC-MS. We identified the carboxy-terminal PDZ ligand of nNOS (CAPON) as a novel tau-binding protein. CAPON is an adaptor protein of neuronal nitric oxide synthase (nNOS), and activated by the N-methyl-D-aspartate receptor. We observed accumulation of CAPON in the hippocampal pyramidal cell layer in the AppNL-G-F -knock-in (KI) brain. To investigate the effect of CAPON accumulation on Alzheimer’s disease (AD) pathogenesis, CAPON was overexpressed in the brain of AppNL-G-F mice crossbred with MAPT (human tau)-KI mice. This produced significant hippocampal atrophy and caspase3-dependent neuronal cell death in the CAPON-expressing hippocampus, suggesting that CAPON accumulation increases neurodegeneration. CAPON expression also induced significantly higher levels of phosphorylated, oligomerized and insoluble tau. In contrast, CAPON deficiency ameliorated the AD-related pathological phenotypes in tauopathy model. These findings suggest that CAPON could be a druggable AD target. To understand the molecular processes that link Aβ amyloidosis, tauopathy and neurodegeneration, the authors screened for tau-interacting proteins. They demonstrated that a novel tau binding protein CAPON accelerates tau pathology and neuronal cell death in an Alzheimer’s disease mouse model.
Collapse
|
16
|
Biological Rationale for Targeting MEK/ERK Pathways in Anti-Cancer Therapy and to Potentiate Tumour Responses to Radiation. Int J Mol Sci 2019; 20:ijms20102530. [PMID: 31126017 PMCID: PMC6567863 DOI: 10.3390/ijms20102530] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2019] [Revised: 05/16/2019] [Accepted: 05/21/2019] [Indexed: 02/07/2023] Open
Abstract
ERK1 and ERK2 (ERKs), two extracellular regulated kinases (ERK1/2), are evolutionary-conserved and ubiquitous serine-threonine kinases involved in regulating cell signalling in normal and pathological tissues. The expression levels of these kinases are almost always different, with ERK2 being the more prominent. ERK1/2 activation is fundamental for the development and progression of cancer. Since their discovery, much research has been dedicated to their role in mitogen-activated protein kinases (MAPK) pathway signalling and in their activation by mitogens and mutated RAF or RAS in cancer cells. In order to gain a better understanding of the role of ERK1/2 in MAPK pathway signalling, many studies have been aimed at characterizing ERK1/2 splicing isoforms, mutants, substrates and partners. In this review, we highlight the differences between ERK1 and ERK2 without completely discarding the hypothesis that ERK1 and ERK2 exhibit functional redundancy. The main goal of this review is to shed light on the role of ERK1/2 in targeted therapy and radiotherapy and highlight the importance of identifying ERK inhibitors that may overcome acquired resistance. This is a highly relevant therapeutic issue that needs to be addressed to combat tumours that rely on constitutively active RAF and RAS mutants and the MAPK pathway.
Collapse
|
17
|
Zang YQ, Feng YY, Luo YH, Zhai YQ, Ju XY, Feng YC, Wang JR, Yu CQ, Jin CH. Glycitein induces reactive oxygen species-dependent apoptosis and G0/G1 cell cycle arrest through the MAPK/STAT3/NF-κB pathway in human gastric cancer cells. Drug Dev Res 2019; 80:573-584. [PMID: 30916421 DOI: 10.1002/ddr.21534] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2019] [Revised: 02/26/2019] [Accepted: 03/18/2019] [Indexed: 02/06/2023]
Abstract
Glycitein is an isoflavone that reportedly inhibits the proliferation of human breast cancer and prostate cancer cells. However, its anti-cancer molecular mechanisms in human gastric cancer remain to be defined. This study evaluated the antitumor effects of glycitein on human gastric cancer cells and investigated the underlying mechanisms. We used MTT assay, flow cytometry and western blotting to investigate its molecular mechanisms with focus on reactive oxygen species (ROS) production. Our results showed that glycitein had significant cytotoxic effects on human gastric cancer cells. Glycitein markedly decreased mitochondrial transmembrane potential (ΔΨm) and increased AGS cells mitochondrial-related apoptosis, and caused G0/G1 cell cycle arrest by regulating cycle-related protein. Mechanistically, accompanying ROS, glycitein can activate mitogen-activated protein kinase (MAPK) and inhibited the signal transducer and activator of transcription 3 (STAT3) and nuclear factor-kappaB (NF-κB) signaling pathways. Furthermore, the MAPK signaling pathway regulated the expression levels of STAT3 and NF-κB upon treatment with MAPK inhibitor and N-acetyl-L-cysteine (NAC). These findings suggested that glycitein induced AGS cell apoptosis and G0/G1 phase cell cycle arrest via ROS-related MAPK/STAT3/NF-κB signaling pathways. Thus, glycitein has the potential to a novel targeted therapeutic agent for human gastric cancer.
Collapse
Affiliation(s)
- Yan-Qing Zang
- Department of Food Science and Engineering, College of Food Science, Heilongjiang Bayi Agricultural University, Daqing, Heilongjiang, China
| | - Yan-Yu Feng
- Department of Food Science and Engineering, College of Food Science, Heilongjiang Bayi Agricultural University, Daqing, Heilongjiang, China
| | - Ying-Hua Luo
- Department of Grass Science, College of Animal Science & Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing, Heilongjiang, China
| | - Yu-Qing Zhai
- Department of Food Science and Engineering, College of Food Science, Heilongjiang Bayi Agricultural University, Daqing, Heilongjiang, China
| | - Xue-Ying Ju
- Department of Food Science and Engineering, College of Food Science, Heilongjiang Bayi Agricultural University, Daqing, Heilongjiang, China
| | - Yu-Chao Feng
- Department of Food Science and Engineering, College of Food Science, Heilongjiang Bayi Agricultural University, Daqing, Heilongjiang, China
| | - Jia-Ru Wang
- Department of Biochemistry and Molecular Biology, College of Life Science & Technology, Heilongjiang Bayi Agricultural University, Daqing, Heilongjiang, China
| | - Chang-Qing Yu
- Department of Food Science and Engineering, College of Food Science, Heilongjiang Bayi Agricultural University, Daqing, Heilongjiang, China
| | - Cheng-Hao Jin
- Department of Food Science and Engineering, College of Food Science, Heilongjiang Bayi Agricultural University, Daqing, Heilongjiang, China.,Department of Biochemistry and Molecular Biology, College of Life Science & Technology, Heilongjiang Bayi Agricultural University, Daqing, Heilongjiang, China
| |
Collapse
|
18
|
Soto C, Bergado G, Blanco R, Griñán T, Rodríguez H, Ros U, Pazos F, Lanio ME, Hernández AM, Álvarez C. Sticholysin II-mediated cytotoxicity involves the activation of regulated intracellular responses that anticipates cell death. Biochimie 2018; 148:18-35. [DOI: 10.1016/j.biochi.2018.02.006] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2017] [Accepted: 02/07/2018] [Indexed: 12/12/2022]
|
19
|
Xing Z, Zeng M, Hu H, Zhang H, Hao Z, Long Y, Chen S, Su H, Yuan Z, Xu M, Chen J. Fragile X mental retardation protein promotes astrocytoma proliferation via the MEK/ERK signaling pathway. Oncotarget 2018; 7:75394-75406. [PMID: 27683117 PMCID: PMC5342749 DOI: 10.18632/oncotarget.12215] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2016] [Accepted: 09/12/2016] [Indexed: 12/16/2022] Open
Abstract
Objective To examine the association between fragile X mental retardation protein (FMRP) expression and astrocytoma characteristics. Methods Pathologic grade and expressions of glial fibrillary acidic protein (GFAP), Ki67 (proliferation marker), and FMRP were determined in astrocytoma specimens from 74 patients. Kaplan-Meier survival analysis was undertaken. Pathologic grade and protein levels of FMRP were determined in 24 additional patients with astrocytoma and 6 controls (cerebral trauma). In cultured U251 and U87 cell lines, the effects of FMRP knock-down on cell proliferation, AKT/mTOR/GSK-3β and MEK/ERK signaling were studied. The effects of FMRP knock-down on the volumes and weights of U251 cell-derived orthotopic tumors in mice were investigated. Results In patients, FMRP expression was increased in grade IV (5.1-fold, P<0.01) and grade III (3.2-fold, P<0.05) astrocytoma, compared with controls. FMRP and Ki67 expressions were positively correlated (R2=0.877, P<0.001). Up-regulation of FMRP was associated with poorer survival among patients with FMRP integrated optical density >30 (P<0.01). In astrocytoma cell lines, FMRP knock-down slowed proliferation (P<0.05), inhibited total MEK levels P<0.05, and reduced phosphorylation of MEK (Ser217/221) and ERK (Thr202/Tyr204) (P<0.05). In mice with orthotopic tumors, FMRP knock-down decreased FMRP and Ki67 expressions, and reduced tumor volume and weight (36.3% or 61.5% on day 15, both P<0.01). Also, phosphorylation of MEK (Ser217/221) and ERK (Thr202/Tyr204), and total MEK in xenografts were decreased in sh-FMRP xenografts compared with non-transfected ones (all P<0.05). Conclusion Enhanced FMRP expression in astrocytoma may promote proliferation through activation of MEK/ERK signaling.
Collapse
Affiliation(s)
- Zhou Xing
- Department of Oncology, The First Affiliated Hospital, Jinan University, Guangzhou 510632, People's Republic of China.,Department of Neurosurgery, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou 510260, People's Republic of China
| | - Minling Zeng
- Department of Neurosurgery, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou 510260, People's Republic of China
| | - Huixian Hu
- Translational Medicine Center, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou 510260, People's Republic of China
| | - Hui Zhang
- Translational Medicine Center, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou 510260, People's Republic of China
| | - Zhuofang Hao
- Department of Pathology, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou 510260, People's Republic of China
| | - Yuesheng Long
- Institute of Neuroscience, Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and Ministry of Education of China, Guangzhou Medical University, Guangzhou 510260, People's Republic of China
| | - Shengqiang Chen
- Institute of Neuroscience, Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and Ministry of Education of China, Guangzhou Medical University, Guangzhou 510260, People's Republic of China
| | - Hang Su
- Translational Medicine Center, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou 510260, People's Republic of China
| | - Zhongmin Yuan
- Department of Neurosurgery, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou 510260, People's Republic of China.,Institute of Neuroscience, Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and Ministry of Education of China, Guangzhou Medical University, Guangzhou 510260, People's Republic of China
| | - Meng Xu
- Department of Oncology, The First Affiliated Hospital, Jinan University, Guangzhou 510632, People's Republic of China
| | - Jingqi Chen
- Translational Medicine Center, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou 510260, People's Republic of China.,Department of Medical Oncology, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou 510260, People's Republic of China
| |
Collapse
|
20
|
Carbon Ion-Irradiated Hepatoma Cells Exhibit Coupling Interplay between Apoptotic Signaling and Morphological and Mechanical Remodeling. Sci Rep 2016; 6:35131. [PMID: 27731354 PMCID: PMC5059721 DOI: 10.1038/srep35131] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2016] [Accepted: 09/22/2016] [Indexed: 12/27/2022] Open
Abstract
A apoptotic model was established based on the results of five hepatocellular carcinoma cell (HCC) lines irradiated with carbon ions to investigate the coupling interplay between apoptotic signaling and morphological and mechanical cellular remodeling. The expression levels of key apoptotic proteins and the changes in morphological characteristics and mechanical properties were systematically examined in the irradiated HCC lines. We observed that caspase-3 was activated and that the Bax/Bcl-2 ratio was significantly increased over time. Cellular morphology and mechanics analyses indicated monotonic decreases in spatial sizes, an increase in surface roughness, a considerable reduction in stiffness, and disassembly of the cytoskeletal architecture. A theoretical model of apoptosis revealed that mechanical changes in cells induce the characteristic cellular budding of apoptotic bodies. Statistical analysis indicated that the projected area, stiffness, and cytoskeletal density of the irradiated cells were positively correlated, whereas stiffness and caspase-3 expression were negatively correlated, suggesting a tight coupling interplay between the cellular structures, mechanical properties, and apoptotic protein levels. These results help to clarify a novel arbitration mechanism of cellular demise induced by carbon ions. This biomechanics strategy for evaluating apoptosis contributes to our understanding of cancer-killing mechanisms in the context of carbon ion radiotherapy.
Collapse
|
21
|
Ghorai A, Sarma A, Bhattacharyya NP, Ghosh U. Carbon ion beam triggers both caspase-dependent and caspase-independent pathway of apoptosis in HeLa and status of PARP-1 controls intensity of apoptosis. Apoptosis 2016; 20:562-80. [PMID: 25670618 DOI: 10.1007/s10495-015-1107-3] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
High linear energy transfer (LET) carbon ion beam (CIB) is becoming very promising tool for various cancer treatments and is more efficient than conventional low LET gamma or X-rays to kill malignant or radio-resistant cells, although detailed mechanism of cell death is still unknown. Poly (ADP-ribose) polymerase-1 (PARP-1) is a key player in DNA repair and its inhibitors are well-known as radio-sensitizer for low LET radiation. The objective of our study was to find mechanism(s) of induction of apoptosis by CIB and role of PARP-1 in CIB-induced apoptosis. We observed overall higher apoptosis in PARP-1 knocked down HeLa cells (HsiI) compared with negative control H-vector cells after irradiation with CIB (0-4 Gy). CIB activated both intrinsic and extrinsic pathways of apoptosis via caspase-9 and caspase-8 activation respectively, followed by caspase-3 activation, apoptotic body, nucleosomal ladder formation and sub-G1 accumulation. Apoptosis inducing factor translocation into nucleus in H-vector but not in HsiI cells after CIB irradiation contributed caspase-independent apoptosis. Higher p53 expression was observed in HsiI cells compared with H-vector after exposure with CIB. Notably, we observed about 37 % fall of mitochondrial membrane potential, activation of caspase-9 and caspase-3 and mild activation of caspase-8 without any detectable apoptotic body formation in un-irradiated HsiI cells. We conclude that reduction of PARP-1 expression activates apoptotic signals via intrinsic and extrinsic pathways in un-irradiated cells. CIB irradiation further intensified both intrinsic and extrinsic pathways of apoptosis synergistically along with up-regulation of p53 in HsiI cells resulting overall higher apoptosis in HsiI than H-vector.
Collapse
Affiliation(s)
- Atanu Ghorai
- Department of Biochemistry & Biophysics, University of Kalyani, Kalyani, 741235, India
| | | | | | | |
Collapse
|
22
|
Wang M, Lu X, Dong X, Hao F, Liu Z, Ni G, Chen D. pERK1/2 silencing sensitizes pancreatic cancer BXPC-3 cell to gemcitabine-induced apoptosis via regulating Bax and Bcl-2 expression. World J Surg Oncol 2015; 13:66. [PMID: 25880226 PMCID: PMC4337256 DOI: 10.1186/s12957-015-0451-7] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2014] [Accepted: 01/08/2015] [Indexed: 11/10/2022] Open
Abstract
Background Our previous study has demonstrated that knockdown of activated ERK1/2(pERK1/2) sensitizes pancreatic cancer cells to chemotherapeutic drug gemcitabine (Gem) treatment. However, the details of this survival mechanism remain undefined. It has also shown that Bcl-2 confers resistance and Bax sensitizes to gemcitabine-induced apoptosis in pancreatic cancer cells. Furthermore, the extracellular signaling-regulated kinase (ERK) signaling pathway regulates Bcl-2/Bax expression ratio. We therefore tested the hypothesis that pancreatic cancer cells are resistant to gemcitabine and this resistance is due to activation of ERK1/2 and subsequent upregulation of Bcl-2 and downregulation of Bax. Methods Pancreatic cancer cell BXPC-3 was used in the study. The effect of pharmacological inhibition of ERK1/2 on resistance of pancreatic cancer cells to apoptosis induced by treatment with gemcitabine was analyzed. The following methods were utilized: TUNEL and ELISA were used to detect apoptosis. Western blot was used to detect the protein expression. Results Gemcitabine treatment enhanced the activity of ERK1/2 in the BXPC-3 cells. Inhibition of the ERK1/2 by PD98059 could downregulate Bcl-2 and upregulate Bax and was associated with restoration of sensitivity to gemcitabine in BXPC-3 cells. Depletion of endogenous Bcl-2 expression by specific small interfering RNA transfection significantly increased gemcitabine-induced cell apoptosis. Combined treatment with PD98059 and Bax siRNA transfection could decrease gemcitabine-induced ERK1/2 and Bax activation, which subsequently resulted in decreased apoptosis. Conclusions The upregulation of ERK1/2-dependent Bcl-2 and downregulation of ERK1/2-dependent Bax can protect human pancreatic cancer cells from gemcitabine-induced apoptosis. Targeting the ERK1/2-Bax/Bcl-2 pathway may in part lead to sensitization of pancreatic cancer to gemcitabine.
Collapse
Affiliation(s)
- Min Wang
- Department of Clinical Laboratory, People's Hospital of Laiwu, Laiwu, Shangdong, China.
| | - Xingjiao Lu
- Department of Internal Neurology, People's Hospital of Zhangqiu, ZhangQiu, Shangdong, China.
| | - Xueguang Dong
- Department of Clinical Laboratory, People's Hospital of Laiwu, Laiwu, Shangdong, China.
| | - Fengyun Hao
- Department of General Surgery, The Affiliated Hospital of Qingdao University, Qingdao, China. .,Department of Clinical Laboratory, People's Hospital of Weifang, Weifang, Shangdong, China.
| | - Zimin Liu
- Department of Clinical Laboratory, People's Hospital of Laiwu, Laiwu, Shangdong, China. .,Department of General Surgery, The Affiliated Hospital of Qingdao University, Qingdao, China.
| | - Guangzhen Ni
- Department of Clinical Laboratory, People's Hospital of Weifang, Weifang, Shangdong, China.
| | - Dong Chen
- Department of General Surgery, The Affiliated Hospital of Qingdao University, Qingdao, China.
| |
Collapse
|
23
|
Kovarova M, Koller BH. PGE₂ promotes apoptosis induced by cytokine deprivation through EP3 receptor and induces Bim in mouse mast cells. PLoS One 2014; 9:e102948. [PMID: 25054560 PMCID: PMC4108439 DOI: 10.1371/journal.pone.0102948] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2014] [Accepted: 06/24/2014] [Indexed: 12/18/2022] Open
Abstract
Increased mast cell numbers are observed at sites of allergic inflammation and restoration of normal mast cell numbers is critical to the resolution of these responses. Early studies showed that cytokines protect mast cells from apoptosis, suggesting a simple model in which diminished cytokine levels during resolution leads to cell death. The report that prostaglandins can contribute both to recruitment and to the resolution of inflammation together with the demonstration that mast cells express all four PGE2 receptors raises the question of whether a single PGE2 receptor mediates the ability of PGE2 to regulate mast cell survival and apoptosis. We report here that PGE2 through the EP3 receptor promotes cell death of mast cells initiated by cytokine withdrawal. Furthermore, the ability of PGE2 to limit reconstitution of tissues with cultured mast cells is lost in cell lacking the EP3 receptor. Apoptosis is accompanied by higher dissipation of mitochondrial potential (ΔΨm), increased caspase-3 activation, chromatin condensation, and low molecular weight DNA cleavage. PGE2 augmented cell death is dependent on an increase in intracellular calcium release, calmodulin dependent kinase II and MAPK activation. Synergy between the EP3 pathway and the intrinsic mitochondrial apoptotic pathway results in increased Bim expression and higher sensitivity of mast cells to cytokine deprivation. This supports a model in which PGE2 can contribute to the resolution of inflammation in part by augmenting the removal of inflammatory cells in this case, mast cells.
Collapse
Affiliation(s)
- Martina Kovarova
- Department of Medicine, Pulmonary Division, University of North Carolina, Chapel Hill, Chapel Hill, North Carolina, United States of America
| | - Beverly H. Koller
- Department of Medicine, Pulmonary Division, University of North Carolina, Chapel Hill, Chapel Hill, North Carolina, United States of America
- Department of Genetics, University of North Carolina, Chapel Hill, Chapel Hill, North Carolina, United States of America
- * E-mail:
| |
Collapse
|
24
|
Hocsak E, Cseh A, Szabo A, Bellyei S, Pozsgai E, Kalai T, Hideg K, Sumegi B, Boronkai A. PARP inhibitor attenuated colony formation can be restored by MAP kinase inhibitors in different irradiated cancer cell lines. Int J Radiat Biol 2014; 90:1152-61. [PMID: 24937370 DOI: 10.3109/09553002.2014.934927] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
UNLABELLED Abstract Purpose: Sensitizing cancer cells to irradiation is a major challenge in clinical oncology. We aimed to define the signal transduction pathways involved in poly(ADP-ribose) polymerase (PARP) inhibitor-induced radiosensitization in various mammalian cancer lines. MATERIALS AND METHODS Clonogenic survival assays and Western blot examinations were performed following telecobalt irradiation of cancer cells in the presence or absence of various combinations of PARP- and selective mitogen-activated protein kinase (MAPK) inhibitors. RESULTS HO3089 resulted in significant cytotoxicity when combined with irradiation. In human U251 glioblastoma and A549 lung cancer cell lines, Erk1/2 and JNK/SAPK were found to mediate this effect of HO3089 since inhibitors of these kinases ameliorated it. In murine 4T1 breast cancer cell line, p38 MAPK rather than Erk1/2 or JNK/SAPK was identified as the main mediator of HO3089's radiosensitizing effect. Besides the aforementioned changes in kinase signaling, we detected increased p53, unchanged Bax and decreased Bcl-2 expression in the A549 cell line. CONCLUSIONS HO3089 sensitizes cancer cells to photon irradiation via proapoptotic processes where p53 plays a crucial role. Activation of MAPK pathways is regarded the consequence of irradiation-induced DNA damage, thus their inhibition can counteract the radiosenzitizing effect of the PARP inhibitor.
Collapse
Affiliation(s)
- Eniko Hocsak
- Department of Biochemistry and Medical Chemistry, Medical School, University of Pecs , Hungary
| | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Lu S, Yu L, Mu Y, Ma J, Tian J, Xu W, Wang H. Role and mechanism of Twist1 in modulating the chemosensitivity of FaDu cells. Mol Med Rep 2014; 10:53-60. [PMID: 24805866 PMCID: PMC4068721 DOI: 10.3892/mmr.2014.2212] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2013] [Accepted: 04/02/2014] [Indexed: 12/14/2022] Open
Abstract
Multidrug resistance (MDR) is one of the most important obstacles affecting the efficacy of chemotherapy treatments for numerous types of cancer. In the present study, we have demonstrated the possible function of Twist1 in the chemosensitivity of head and neck squamous cell carcinoma (HNSCC) and have identified that its mechanism maybe associated with MDR1/P-gp regulation. To investigate this, the hypopharyngeal cancer cell line, FaDu, and its MDR cell line induced by taxol, FaDu/T, were employed. Stable transfectants targeted to Twist1 overexpression and Twist1 silencing based on FaDu were also conducted. Morphological observation, flow cytometry, reverse transcription-polymerase chain reaction (RT-PCR), western blotting and laser scanning confocal microscope detection were utilized to detect the associations between Twist1 and the chemosensitivity of FaDu cells. Our results demonstrated that Twist1 and MDR1/P-gp were upregulated in FaDu/T cells in a MDR dose-dependent manner. The anti-apoptotic capabilities of FaDu/T cells were enhanced during MDR progression, with apoptosis-related proteins (Bcl-2, Bax, activated caspase-3 and caspase-9) changing to resist apoptosis. Twist1 overexpression decreased the sensitivity of cells to taxol as revealed by a significant increase in MDR1/P-gp and IC50 (P<0.05). This overexpression also enhanced the resistance to apoptosis, with apoptotic proteins changing to resist cell death, and inhibited Ca2+ release induced by taxol (P<0.05). Detections in Twist1 silencing cells also confirmed this result. This study provided evidence that alterations of Twist1 expression modulates the chemosensitivity of FaDu cells to taxol. Therefore, Twist1 knockdown may be a promising treatment regimen for advanced hypopharyngeal carcinoma patients with MDR.
Collapse
Affiliation(s)
- Sumei Lu
- Department of Otorhinolaryngology Head and Neck Surgery, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, Shandong 250022, P.R. China
| | - Liang Yu
- Department of Otorhinolaryngology Head and Neck Surgery, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, Shandong 250022, P.R. China
| | - Yakui Mu
- Department of Otorhinolaryngology Head and Neck Surgery, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, Shandong 250022, P.R. China
| | - Juke Ma
- Department of Otorhinolaryngology Head and Neck Surgery, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, Shandong 250022, P.R. China
| | - Jiajun Tian
- Department of Otorhinolaryngology Head and Neck Surgery, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, Shandong 250022, P.R. China
| | - Wei Xu
- Department of Otorhinolaryngology Head and Neck Surgery, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, Shandong 250022, P.R. China
| | - Haibo Wang
- Department of Otorhinolaryngology Head and Neck Surgery, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, Shandong 250022, P.R. China
| |
Collapse
|
26
|
Wang Y, Liu D, Zhao H, Jiang H, Luo C, Wang M, Yin H. Cordyceps sinensis polysaccharide CPS-2 protects human mesangial cells from PDGF-BB-induced proliferation through the PDGF/ERK and TGF-β1/Smad pathways. Mol Cell Endocrinol 2014; 382:979-88. [PMID: 24309234 DOI: 10.1016/j.mce.2013.11.018] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/26/2013] [Revised: 10/29/2013] [Accepted: 11/25/2013] [Indexed: 12/18/2022]
Abstract
CPS-2, a Cordyceps sinensis polysaccharide, has been demonstrated to have significant therapeutic activity against chronic renal failure. However, little is known about the underlying molecular mechanism. In this study, we found that CPS-2 could inhibit PDGF-BB-induced human mesangial cells (HMCs) proliferation in a dose-dependent manner. In addition, CPS-2 notably suppressed the expression of α-SMA, PDGF receptor-beta (PDGFRβ), TGF-β1, and Smad 3 in PDGF-BB-treated HMCs. Furthermore, PDGF-BB-stimulated ERK activation was significantly inhibited by CPS-2, and this inhibitory effect was synergistically potentiated by U0126. CPS-2 could prevent the PDGFRβ promoter activity induced by PDGF-BB, and return expression of PDGFRβ, TGF-β1, and TGFβRI to normal levels while cells were under PDGFRβ and ERK silencing conditions and transfected with DN-ERK. Taken together, these findings demonstrated that CPS-2 reduces PDGF-BB-induced cell proliferation through the PDGF/ERK and TGF-β1/Smad pathways, and it may have bi-directional regulatory effects on the PDGF/ERK cellular signaling pathway.
Collapse
Affiliation(s)
- Ying Wang
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, People's Republic of China; School of Life Science and Technology, China Pharmaceutical University, Nanjing 210009, Jiangsu, People's Republic of China
| | - Dan Liu
- School of Life Science and Technology, China Pharmaceutical University, Nanjing 210009, Jiangsu, People's Republic of China
| | - Huan Zhao
- School of Life Science and Technology, China Pharmaceutical University, Nanjing 210009, Jiangsu, People's Republic of China
| | - Huixing Jiang
- First Clinical Medical College, Nanjing University of Traditional Chinese Medicine, Nanjing Traditional Chinese Medicine Hospital, Nanjing 210010, Jiangsu, People's Republic of China
| | - Chen Luo
- School of Life Science and Technology, China Pharmaceutical University, Nanjing 210009, Jiangsu, People's Republic of China
| | - Min Wang
- School of Life Science and Technology, China Pharmaceutical University, Nanjing 210009, Jiangsu, People's Republic of China.
| | - Hongping Yin
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, People's Republic of China; School of Life Science and Technology, China Pharmaceutical University, Nanjing 210009, Jiangsu, People's Republic of China.
| |
Collapse
|
27
|
Guo X, Fan W, Bian X, Ma D. Upregulation of the Kank1 gene-induced brain glioma apoptosis and blockade of the cell cycle in G0/G1 phase. Int J Oncol 2014; 44:797-804. [PMID: 24399197 DOI: 10.3892/ijo.2014.2247] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2013] [Accepted: 12/19/2013] [Indexed: 11/06/2022] Open
Abstract
The Kank1 gene is one of the important members of the Kank gene family. As an important adaptor protein, Kank1 plays a significant role in the genesis and development of many malignant tumors. It was recently discovered that the Kank1 gene is a new cancer suppressor, and its expression is significantly downregulated or it is not expressed in kidney cancer, bladder cancer, prostate cancer, lung cancer and breast cancer. However, no report on the role of Kank1 in the genesis of brain glioma is available to date. In this study, we found significantly lower expression of the Kank1 gene in human brain glioma cells compared to the other cells evaluated. We used RNA interference techniques to silence Kank1 gene expression and found acceleration of tumor cell proliferation. However, when the Kank1 gene was upregulated, cell apoptosis occurred and the cell cycle was blocked in the G0/G1 phase. Also, we found that upregulating the Kank1 gene may result in the change of mitochondrial membrane potential, and the regulation of Bax and Bcl-2 may promote the mitochondria to release cytochrome C so as to activate Caspase-9 and -3. Thus, the human brain glioma apoptosis induced by upregulation of the Kank1 gene is closely relevant to the mitochondrial pathway.
Collapse
Affiliation(s)
- Xiaohang Guo
- Department of Neurology, The First Hospital of Jilin University, Changchun 130021, P.R. China
| | - Wenhai Fan
- Department of Neurosurgery, The Affiliated Zhongshan Hospital of Dalian University, Dalian 116001, P.R. China
| | - Xinchao Bian
- Department of Neurosurgery, Zibo Central Hospital, Zibo 255000, P.R. China
| | - Dihui Ma
- Department of Neurology, The First Hospital of Jilin University, Changchun 130021, P.R. China
| |
Collapse
|
28
|
Debus J, Abdollahi A. For the next trick: new discoveries in radiobiology applied to glioblastoma. Am Soc Clin Oncol Educ Book 2014:e95-e99. [PMID: 24857153 DOI: 10.14694/edbook_am.2014.34.e95] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/03/2023]
Abstract
Glioblastoma (GBM) is the most common malignant brain tumor. Radiotherapy post surgical resection remained the mainstay of the management of GBM for decades until the addition of temozolomide was shown to prolong the median overall survival (OS) by 2.5 months to 14.6 months in 2005. Infiltrative growth to surrounding normal brain tissue and cooption of vascular niches, peripheral microvasuclar hyperplasia, and central hypoxic regions with pseudopalisading necrosis are characteristics of GBM and are causally linked to their exceptional radio- and chemo-resistant phenotype. An intratumoral hierarchy is postulated consisting of tumor stem cells in the apex with high DNA-repair proficiency resisting radiotherapy. It is conceivable that the stem cell property is more dynamic than originally anticipated. Niche effects such as exposure to hypoxia and intercellular communication in proximities to endothelial or bone marrow-derived cells (BMDC), for example, may activate such "stem cell" programs. GBM are exceptionally stroma-rich tumors and may consist of more than 70% stroma components, such as microglia and BMDC. It becomes increasingly apparent that treatment of GBM needs to integrate therapies targeting all above-mentioned distinct pathophysiological features. Accordingly, recent approaches in GBM therapy include inhibition of invasion (e.g., integrin, EGFR, CD95, and mTOR inhibition), antiangiogenesis and stroma modulators (TGFbeta, VEGF, angiopoetin, cMET inhibitors) and activation of immune response (vaccination and blockage of negative co-stimulatory signals). In addition, high LET-radiotherapy, for example with carbon ions, is postulated to ablate tumor stem cell and hypoxic cells more efficiently as compared with conventional low-LET photon irradiation. We discuss current key concepts, their limitations, and potentials to improve the outcome in this rapidly progressive and devastating disease.
Collapse
Affiliation(s)
- Juergen Debus
- From the German Cancer Consortium (DKTK), Heidelberg, Germany; Department of Radiation Oncology, Heidelberg Ion Therapy Center (HIT), Heidelberg Institute of Radiation Oncology (HIRO), University of Heidelberg Medical School; Molecular and Translational Radiation Oncology, National Center for Tumor Diseases (NCT), German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Amir Abdollahi
- From the German Cancer Consortium (DKTK), Heidelberg, Germany; Department of Radiation Oncology, Heidelberg Ion Therapy Center (HIT), Heidelberg Institute of Radiation Oncology (HIRO), University of Heidelberg Medical School; Molecular and Translational Radiation Oncology, National Center for Tumor Diseases (NCT), German Cancer Research Center (DKFZ), Heidelberg, Germany
| |
Collapse
|
29
|
Signalling mechanisms involved in renal pathological changes during cisplatin-induced nephropathy. Eur J Clin Pharmacol 2013; 69:1863-74. [PMID: 23929259 DOI: 10.1007/s00228-013-1568-7] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2013] [Accepted: 07/24/2013] [Indexed: 12/20/2022]
Abstract
CONTEXT Cisplatin, a coordination platinum complex, is used as a potential anti-neoplastic agent, having well recognized DNA-damaging property that triggers cell-cycle arrest and cell death in cancer therapy. Beneficial chemotherapeutic actions of cisplatin can be detrimental for kidneys. BACKGROUND Unbound cisplatin gets accumulated in renal tubular cells, leading to cell injury and death. This liable action of cisplatin on kidneys is mediated by altered intracellular signalling pathways such as mitogen-activated protein kinase (MAPK), extracellular regulated kinase (ERK), or C- Jun N terminal kinase/stress-activated protein kinase (JNK/SAPK). Further, these signalling alterations are responsible for release and activation of tumour necrosis factor (TNF-α), mitochondrial dysfunction, and apoptosis, which ultimately cause the renal pathogenic process. Cisplatin itself enhances the generation of reactive oxygen species (ROS) and activation of nuclear factor-κB (NF-κB), inflammation, and mitochondrial dysfunction, which further leads to renal apoptosis. Cisplatin-induced nephropathy is also mediated through the p53 and protein kinase-Cδ (PKCδ) signalling pathways. OBJECTIVE This review explores these signalling alterations and their possible role in the pathogenesis of cisplatin-induced renal injury.
Collapse
|
30
|
ZHANG RUIJIAN, WANG RUIJUN, CHANG HONG, WU FEI, LIU CHUNTAO, DENG DONGFENG, FAN WENHAI. Downregulation of Ezh2 expression by RNA interference induces cell cycle arrest in the G0/G1 phase and apoptosis in U87 human glioma cells. Oncol Rep 2012; 28:2278-84. [DOI: 10.3892/or.2012.2033] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2012] [Accepted: 08/17/2012] [Indexed: 11/05/2022] Open
|
31
|
Musha A, Yoshida Y, Takahashi T, Ando K, Funayama T, Kobayashi Y, Negishi A, Yokoo S, Nakano T. Synergistic effect of heat shock protein 90 inhibitor, 17-allylamino-17-demethoxygeldanamycin and X-rays, but not carbon-ion beams, on lethality in human oral squamous cell carcinoma cells. JOURNAL OF RADIATION RESEARCH 2012; 53:545-50. [PMID: 22843619 PMCID: PMC3393353 DOI: 10.1093/jrr/rrs012] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/08/2023]
Abstract
The purpose of this study is to clarify the effect of a heat shock protein 90 inhibitor, 17-allylamino-17-demethoxygeldanamycin (17-AAG), in combination with X-rays or carbon-ion beams on cell killing in human oral squamous cell carcinoma LMF4 cells. Cell survival was measured by colony formation assay. Cell-cycle distribution was analyzed by flow cytometry. Expression of DNA repair-related proteins was investigated by western blotting. The results showed 17-AAG to have synergistic effects on cell lethality with X-rays, but not with carbon-ion beams. The 17-AAG decreased G(2)/M arrest induced by X-rays, but not by carbon-ion beams. Both X-ray and carbon-ion irradiation up-regulated expression of non-homologous end-joining-associated proteins, Ku70 and Ku80, but 17-AAG inhibited only X-ray-induced up-regulation of these proteins. These results show that 17-AAG with X-rays releases G(2)/M phase arrest; cells carrying misrepaired DNA damage then move on to the G(1) phase. We demonstrate, for the first time, that the radiosensitization effect of 17-AAG is not seen with carbon-ion beams because 17-AAG does not affect these changes.
Collapse
Affiliation(s)
- Atsushi Musha
- Department of Radiation Oncology, Gunma University Graduate School of Medicine, Gunma, Japan
- Department of Stomatology and Oral Surgery, Gunma University Graduate School of Medicine, Gunma, Japan
| | | | - Takeo Takahashi
- Department of Radiation Oncology, Gunma University Graduate School of Medicine, Gunma, Japan
- Department of Radiology, Saitama Medical Center, Saitama Medical University, Saitama, Japan
| | - Koichi Ando
- Gunma University Heavy Ion Medical Center, Gunma, Japan
| | - Tomoo Funayama
- Microbeam Radiation Biology Group, Japan Atomic Energy Agency, Gunma, Japan
| | - Yasuhiko Kobayashi
- Microbeam Radiation Biology Group, Japan Atomic Energy Agency, Gunma, Japan
| | - Akihide Negishi
- Department of Stomatology and Oral Surgery, Gunma University Graduate School of Medicine, Gunma, Japan
| | - Satoshi Yokoo
- Department of Stomatology and Oral Surgery, Gunma University Graduate School of Medicine, Gunma, Japan
| | - Takashi Nakano
- Department of Radiation Oncology, Gunma University Graduate School of Medicine, Gunma, Japan
- Gunma University Heavy Ion Medical Center, Gunma, Japan
- Corresponding author: Department of Radiation Oncology, Gunma University Graduate School of Medicine, 3-39-22, Showa-machi, Maebashi, Gunma 371-8511, Japan; Tel: +81(27)220-8383; Fax: +81(27)220-8397;
| |
Collapse
|
32
|
Dilmanian FA, Rusek A, Fois GR, Olschowka J, Desnoyers NR, Park JY, Dioszegi I, Dane B, Wang R, Tomasi D, Lee H, Hurley SD, Coyle PK, Meek AG, O'Banion MK. Interleaved carbon minibeams: an experimental radiosurgery method with clinical potential. Int J Radiat Oncol Biol Phys 2012; 84:514-9. [PMID: 22342299 DOI: 10.1016/j.ijrobp.2011.12.025] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2011] [Revised: 10/17/2011] [Accepted: 12/05/2011] [Indexed: 12/18/2022]
Abstract
PURPOSE To evaluate the efficacy of "interleaved carbon minibeams" for ablating a 6.5-mm target in a rabbit brain with little damage to the surrounding brain. The method is based on the well-established tissue-sparing effect of arrays of thin planes of radiation. METHODS AND MATERIALS Broad carbon beams from the National Aeronautics and Space Agency Space Radiation Facility at Brookhaven National Laboratory were segmented into arrays of parallel, horizontal, 0.3-mm-thick planar beams (minibeams). The minibeams' gradual broadening in tissues resulted in 0.525-mm beam thickness at the target's proximal side in the spread-out Bragg peak. Interleaving was therefore implemented by choosing a 1.05 mm beam spacing on-center. The anesthetized rabbit, positioned vertically on a stage capable of rotating about a vertical axis, was exposed to arrays from four 90° angles, with the stage moving up by 0.525 mm in between. This produced a solid radiation field at the target while exposing the nontargeted tissues to single minibeam arrays. The target "physical" absorbed dose was 40.2 Gy. RESULTS The rabbit behaved normally during the 6-month observation period. Contrast magnetic resonance imaging and hematoxylin and eosin histology at 6 months showed substantial focal target damage with little damage to the surrounding brain. CONCLUSION We plan to evaluate the method's therapeutic efficacy by comparing it with broad-beam carbon therapy in animal models. The method's merits would combine those of carbon therapy (i.e., tight target dose because of the carbon's Bragg-peak, sharp dose falloff, and high relative biological effectiveness at the target), together with the method's low impact on the nontargeted tissues. The method's smaller impact on the nontargeted brain might allow carbon therapy at higher target doses and/or lower normal tissue impact, thus leading to a more effective treatment of radioresistant tumors. It should also make the method more amenable to administration in either a single dose fraction or in a small number of fractions.
Collapse
Affiliation(s)
- F Avraham Dilmanian
- Medical Department, Brookhaven National Laboratory, Upton, NY 11973-5000, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Abstract
Frequently, low doses of toxins and other stressors not only are harmless but also activate an adaptive stress response that raise the resistance of the organism against high doses of the same agent. This phenomenon, which is known as "hormesis", is best represented by ischemic preconditioning, the situation in which short ischemic episodes protect the brain and the heart against prolonged shortage of oxygen and nutrients. Many molecules that cause cell death also elicit autophagy, a cytoprotective mechanism relying on the digestion of potentially harmful intracellular structures, notably mitochondria. When high doses of these agents are employed, cells undergo mitochondrial outer membrane permeabilization and die. In contrast, low doses of such cytotoxic agents can activate hormesis in several paradigms, and this may explain the lifespan-prolonging potential of autophagy inducers including resveratrol and caloric restriction.
Collapse
|
34
|
Bucur O, Stancu AL, Khosravi-Far R, Almasan A. Analysis of apoptosis methods recently used in Cancer Research and Cell Death & Disease publications. Cell Death Dis 2012; 3:e263. [PMID: 22297295 PMCID: PMC3288344 DOI: 10.1038/cddis.2012.2] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
35
|
Abstract
The promyelocytic leukaemia gene PML was originally identified at the t(15;17) translocation of acute promyelocytic leukaemia, which generates the oncogene PML-retinoic acid receptor α. PML epitomises a subnuclear structure called PML nuclear body. Current models propose that PML through its scaffold properties is able to control cell growth and survival at many different levels. Here we discuss the current literature and propose new avenues for investigation.
Collapse
|
36
|
Rufini A, Melino G. Cell death pathology: the war against cancer. Biochem Biophys Res Commun 2011; 414:445-50. [PMID: 21971555 DOI: 10.1016/j.bbrc.2011.09.110] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2011] [Accepted: 09/21/2011] [Indexed: 12/25/2022]
Abstract
Programmed cell death was a fundamental discovery, awarded with the Nobel price in 2002 to Sulston, Brenner and Horvitz. Since then it has been clear that alteration of apoptotic pathways is a common feature of tumors, enabling cancer cells to survive chemotherapeutic interventions. Thus, apoptosis is an attractive target in cancer therapy, with the aim to revert the cancer-related alterations of the cell death machinery. Here, we overview the fundamental apoptotic pathways and summarize the attempts to target apoptosis to restore cell death in cancer cells with a special focus on the p53-family and autophagy.
Collapse
|
37
|
Abstract
The strong interest in cell death, and the shift in emphasis from basic mechanisms to translational aspects fostered the launch last year of the new sister journal of Cell Death and Differentiation, named Cell Death and Disease, to reflect its stronger focus towards clinical applications. Here, we review that first year of activity, which reflects an enthusiastic response by the scientific community. On the basis of this, we now launch two novel initiatives, the start of a new section dedicated to cancer metabolism and the opening of a new editorial office in Shanghai.
Collapse
|