1
|
Li M, Wang Y, Lin X, Yang H, Zhang X, Bai Y, Li X, Zhang L, Cheng F, Cao C, Zhou Q. Evaluation of antitumor potential of an anti-glypican-1 monoclonal antibody in preclinical lung cancer models reveals a distinct mechanism of action. EXPLORATION OF TARGETED ANTI-TUMOR THERAPY 2024; 5:600-626. [PMID: 38966167 PMCID: PMC11220310 DOI: 10.37349/etat.2024.00238] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2023] [Accepted: 02/26/2024] [Indexed: 07/06/2024] Open
Abstract
Aim The main objective of this study was to investigate the antitumor effect of a mouse anti-human glypican-1 (GPC1) monoclonal antibody (mAb) on non-small cell lung carcinoma (NSCLC) and associated molecular mechanisms. Methods The anti-proliferative and anti-migratory activities of anti-GPC1 mAb were examined in A549 and H460 NSCLC cells and LL97A lung fibroblasts. The inhibitory effect of anti-GPC1 mAb on tumor growth was evaluated in an orthotopic lung tumor model. Results The in vitro study showed that anti-GPC1 mAb profoundly inhibited the anchorage-independent growth of A549 and H460 NSCLC cells and exhibited relatively high cytotoxic activities towards LL97A lung fibroblasts, A549/LL97A and H460/LL97A coculture spheroids. Moreover, anti-GPC1 mAb significantly decreased the expression of phospho-Src (p-Src; Tyr416), p-Akt (Ser473) and β-catenin in the co-cultured LL97A lung fibroblasts, and the expression of phospho-mitogen-activated protein kinase kinase (p-MEK; Ser217/221) and phospho-90 kDa ribosomal s6 kinase (p-p90RSK; Ser380) in co-cultured A549 cells. When anti-GPC1 mAb was administered to tumor-bearing mice, the inhibitory effect of anti-GPC1 mAb on the orthotopic lung tumor growth was not statistically significant. Nonetheless, results of Western blot analysis showed significant decrease in the phosphorylation of fibroblast growth factor receptor 1 (FGFR1) at Tyr766, Src at Tyr416, extracellular signal-regulated kinase (ERK) at Thr202/Tyr204, 90 kDa ribosomal S6 kinase (RSK) at Ser380, glycogen synthase kinases 3α (GSK3α) at Ser21 and GSK3β at Ser9 in tumor tissues. These data implicate that anti-GPC1 mAb treatment impairs the interaction between tumor cells and tumor associated fibroblasts by attenuating the paracrine FGFR signal transduction. Conclusions The relatively potent cytotoxicity of anti-GPC1 mAb in lung fibroblasts and its potential inhibitory effect on the paracrine FGFR signal transduction warrant further studies on the combined use of this mAb with targeted therapeutics to improve therapeutic outcomes in lung cancer.
Collapse
Affiliation(s)
- Minghua Li
- Department of Pharmaceutical Sciences, Taneja College of Pharmacy, University of South Florida, Tampa, FL 33612, USA
| | - Yanhong Wang
- Department of Pharmaceutical Sciences, Taneja College of Pharmacy, University of South Florida, Tampa, FL 33612, USA
| | - Xiaoyang Lin
- Department of Pharmaceutical Sciences, Taneja College of Pharmacy, University of South Florida, Tampa, FL 33612, USA
| | - Haiqiang Yang
- Department of Pharmaceutical Sciences, Taneja College of Pharmacy, University of South Florida, Tampa, FL 33612, USA
| | - Xiaolin Zhang
- Department of Pharmaceutical Sciences, Taneja College of Pharmacy, University of South Florida, Tampa, FL 33612, USA
| | - Yun Bai
- MegaNano Biotech Inc., Tampa, FL 33612, USA
| | - Xiankun Li
- Zhengzhou Molecular Diagnosis Engineering Technology Research Center, Zhengzhou 450001, Henan Province, China
| | - Lulu Zhang
- Zhengzhou Molecular Diagnosis Engineering Technology Research Center, Zhengzhou 450001, Henan Province, China
| | - Feng Cheng
- Department of Pharmaceutical Sciences, Taneja College of Pharmacy, University of South Florida, Tampa, FL 33612, USA
| | - Chuanhai Cao
- Department of Pharmaceutical Sciences, Taneja College of Pharmacy, University of South Florida, Tampa, FL 33612, USA
| | - Qingyu Zhou
- Department of Pharmaceutical Sciences, Taneja College of Pharmacy, University of South Florida, Tampa, FL 33612, USA
| |
Collapse
|
2
|
Chen HC, Kuo CY, Chang Y, Tsai DL, Lee MH, Lee JY, Lee HM, Su YC. 5-Methoxytryptophan enhances the sensitivity of sorafenib on the inhibition of proliferation and metastasis for lung cancer cells. BMC Cancer 2024; 24:248. [PMID: 38388902 PMCID: PMC10885375 DOI: 10.1186/s12885-024-11986-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2023] [Accepted: 02/09/2024] [Indexed: 02/24/2024] Open
Abstract
BACKGROUND Lung cancer is a leading cause of cancer-related mortality worldwide, and effective therapies are limited. Lung cancer is a leading cause of cancer-related mortality worldwide with limited effective therapy. Sorafenib is a multi-tyrosine kinase inhibitor frequently used to treat numerous types of malignant tumors. However, it has been demonstrated that sorafenib showed moderate antitumor activity and is associated with several side effects in lung cancer, which restricted its clinical application. This study aimed to examine the antitumor effect of the combination treatment of sorafenib and 5-methoxytryptophan (5-MTP) on cell growth and metastasis of Lewis lung carcinoma (LLC) cells. METHOD The anticancer effect of the combination treatment of sorafenib and 5-MTP was determined through cytotoxicity assay and colony forming assays. The mechanism was elucidated using flow cytometry and western blotting. Wound healing and Transwell assays were conducted to evaluate the impact of the combination treatment on migration and invasion abilities. An in vivo model was employed to analyze the effect of the combination treatment on the tumorigenic ability of LLC cells. RESULT Our results demonstrated that the sorafenib and 5-MTP combination synergistically reduced viability and proliferation compared to sorafenib or 5-MTP treatment alone. Reduction of cyclin D1 expression was observed in the sorafenib alone or combination treatments, leading to cell cycle arrest. Furthermore, the sorafenib-5-MTP combination significantly increased the inhibitory effect on migration and invasion of LLC cells compared to the single treatments. The combination also significantly downregulated vimentin and MMP9 levels, contributing to the inhibition of metastasis. The reduction of phosphorylated Akt and STAT3 expression may further contribute to the inhibitory effect on proliferation and metastasis. In vivo, the sorafenib-5-MTP combination further reduced tumor growth and metastasis compared to the treatment of sorafenib alone. CONCLUSIONS In conclusion, our data indicate that 5-MTP sensitizes the antitumor activity of sorafenib in LLC cells in vitro and in vivo, suggesting that sorafenib-5-MTP has the potential to serve as a therapeutic option for patients with lung cancer.
Collapse
Affiliation(s)
- Huang-Chi Chen
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Kaohsiung Municipal Siaogang Hospital, Kaohsiung, Taiwan
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Chia-Yu Kuo
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Kaohsiung Municipal Siaogang Hospital, Kaohsiung, Taiwan
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Yu Chang
- Department of Obstetrics and Gynecology, E-Da Hospital, I-Shou University, Kaohsiung, Taiwan
- School of Medicine for International Students, College of Medicine, I-Shou University, Kaohsiung, Taiwan
| | - Dong-Lin Tsai
- Department of Surgery, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan
- Division of Chest Surgery, Department of Surgery, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Mei-Hsuan Lee
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Jui-Ying Lee
- Division of Chest Surgery, Department of Surgery, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan
- Graduate Institute of Clinical Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Hui-Ming Lee
- Division of General Surgery, Department of Surgery, E-Da Cancer Hospital, Kaohsiung, Taiwan
- School of Medicine, College of Medicine, I-Shou University, Kaohsiung, Taiwan
| | - Yu-Chieh Su
- School of Medicine, College of Medicine, I-Shou University, Kaohsiung, Taiwan.
- Division of Hematology-Oncology, Department of Internal Medicine, E-Da Hospital, Kaohsiung, Taiwan.
| |
Collapse
|
3
|
Limayem A, Mehta M, Kondos N, Kaushal D, Azam FB, Chellappan S, Qin N, Zhou Q. Evaluation of bactericidal effects of silver hydrosol nanotherapeutics against Enterococcus faecium 1449 drug resistant biofilms. Front Cell Infect Microbiol 2023; 12:1095156. [PMID: 36710982 PMCID: PMC9875038 DOI: 10.3389/fcimb.2022.1095156] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2022] [Accepted: 12/20/2022] [Indexed: 01/13/2023] Open
Abstract
Introduction Silver (Ag) nanoparticles (NPs) are well documented for their broad-spectrum bactericidal effects. This study aimed to test the effect of bioactive Ag-hydrosol NPs on drug-resistant E. faecium 1449 strain and explore the use of artificial intelligence (AI) for automated detection of the bacteria. Methods The formation of E. faecium 1449 biofilms in the absence and presence of Ag-hydrosol NPs at different concentrations ranging from 12.4 mg/L to 123 mg/L was evaluated using a 3-dimentional culture system. The biofilm reduction was evaluated using the confocal microscopy in addition to the Transmission Electronic Microscopy (TEM) visualization and spectrofluorimetric quantification using a Biotek Synergy Neo2 microplate reader. The cytotoxicity of the NPs was evaluated in human nasal epithelial cells using the MTT assay. The AI technique based on Fast Regional Convolutional Neural Network architecture was used for the automated detection of the bacteria. Results Treatment with Ag-hydrosol NPs at concentrations ranging from 12.4 mg/L to 123 mg/L resulted in 78.09% to 95.20% of biofilm reduction. No statistically significant difference in biofilm reduction was found among different batches of Ag-hydrosol NPs. Quantitative concentration-response relationship analysis indicated that Ag-hydrosol NPs exhibited a relative high anti-biofilm activity and low cytotoxicity with an average EC50 and TC50 values of 0.0333 and 6.55 mg/L, respectively, yielding an average therapeutic index value of 197. The AI-assisted TEM image analysis allowed automated detection of E. faecium 1449 with 97% ~ 99% accuracy. Discussion Conclusively, the bioactive Ag-hydrosol NP is a promising nanotherapeutic agent against drug-resistant pathogens. The AI-assisted TEM image analysis was developed with the potential to assess its treatment effect.
Collapse
Affiliation(s)
- Alya Limayem
- Department of Biology, College of Arts & Sciences, University of North Florida, Jacksonville, FL, United States,Department of Pharmaceutical Sciences, Graduate Program, Taneja College of Pharmacy, University of South Florida, Tampa, FL, United States,*Correspondence: Alya Limayem, ; Qingyu Zhou,
| | - Mausam Mehta
- Department of Pharmaceutical Sciences, Graduate Program, Taneja College of Pharmacy, University of South Florida, Tampa, FL, United States,Morsani College of Medicine, University of South Florida, Tampa, FL, United States
| | - Natalie Kondos
- Department of Pharmaceutical Sciences, Graduate Program, Taneja College of Pharmacy, University of South Florida, Tampa, FL, United States,Morsani College of Medicine, University of South Florida, Tampa, FL, United States
| | - Divya Kaushal
- Department of Pharmaceutical Sciences, Graduate Program, Taneja College of Pharmacy, University of South Florida, Tampa, FL, United States
| | - Farhat Binte Azam
- Department of Computer Science & Engineering, College of Engineering, University of South Florida, Tampa, FL, United States
| | - Sriram Chellappan
- Department of Computer Science & Engineering, College of Engineering, University of South Florida, Tampa, FL, United States
| | - Nan Qin
- Department of R&D and Analytical Services, Natural Immunogenics Corporation, Sarasota, FL, United States
| | - Qingyu Zhou
- Department of Pharmaceutical Sciences, Graduate Program, Taneja College of Pharmacy, University of South Florida, Tampa, FL, United States,*Correspondence: Alya Limayem, ; Qingyu Zhou,
| |
Collapse
|
4
|
Kim N, Kim S, Song Y, Choi I, Lee SY, Kim KM, Rhu HC, Lee JY, Seo HR. Chromenopyrimidinone exhibit antitumor effects through inhibition of CAP1 (Adenylyl cyclase-associated protein 1) expression in hepatocellular carcinoma. Chem Biol Interact 2022; 365:110066. [PMID: 35931200 DOI: 10.1016/j.cbi.2022.110066] [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: 03/29/2022] [Revised: 07/05/2022] [Accepted: 07/19/2022] [Indexed: 11/03/2022]
Abstract
Hepatocellular carcinoma (HCC) is one of the most malignant human cancers, with a high mortality rate worldwide. Within an HCC tumor, cancer stem cells (CSCs) are responsible for tumor maintenance and progression and may contribute to resistance to standard HCC treatments. Previously, we characterized CD133+ cells as CSCs in primary HCC and identified chromenopyrimidinone (CPO) as a novel therapeutic for the effective treatment of CD133+ HCC. However, the biological function and molecular mechanism of CD133 remain unclear. Epigenetic alterations of CSCs have impacts on tumor initiation, progression, and therapeutic response. Here, we found that pharmacological and genetic depletion of CD133 in HCC attenuated the activity of DNA methyltransferases via control of DNMT3B stabilization. Genes were ranked by degree of promoter hypo/hyper methylation and significantly differential expression to create an "epigenetically activated by CPO" ranked genes list. Through this epigenetic analysis, we found that CPO treatment altered DNA methylation-mediated oncogenic signaling in HCCs. Specifically, CPO treatment inhibited Adenylyl cyclase-associated protein 1 (CAP1) expression, thereby reducing FAK/ERK activity and EMT-related proteins in HCC. Moreover, CPO improved the efficacy of sorafenib by inhibiting CAP1 expression and FAK/ERK activation in sorafenib-resistant HCC. These novel mechanistic insights may ultimately open up avenues for strategies targeting DNA methylation in liver cancer stem cells and provides novel therapeutic function of CPO for the effective treatment of sorafenib-resistant HCC.
Collapse
Affiliation(s)
- Namjeong Kim
- Advanced Biomedical Research Laboratory, Institut Pasteur Korea, 16, Daewangpangyo-ro 712 Beon-gil, Bundang-gu, Seongnam-si, Gyeonggi-do, 463-400, South Korea
| | - Sanghwa Kim
- Advanced Biomedical Research Laboratory, Institut Pasteur Korea, 16, Daewangpangyo-ro 712 Beon-gil, Bundang-gu, Seongnam-si, Gyeonggi-do, 463-400, South Korea
| | - Yeonhwa Song
- Advanced Biomedical Research Laboratory, Institut Pasteur Korea, 16, Daewangpangyo-ro 712 Beon-gil, Bundang-gu, Seongnam-si, Gyeonggi-do, 463-400, South Korea
| | - Inhee Choi
- Medicinal Chemistry, Institut Pasteur Korea, 16, Daewangpangyo-ro 712 Beon-gil, Bundang-gu, Seongnam-si, Gyeonggi-do, 463-400, South Korea
| | - Su-Yeon Lee
- Advanced Biomedical Research Laboratory, Institut Pasteur Korea, 16, Daewangpangyo-ro 712 Beon-gil, Bundang-gu, Seongnam-si, Gyeonggi-do, 463-400, South Korea
| | - Kang Mo Kim
- Department of Gastroenterology, Asan Liver Center, Asan Medical Center, University of Ulsan College of Medicine, Olympic-ro 43-gil, Songpa-gu, Seoul, 05505, South Korea
| | - Hyung Chul Rhu
- R&D Center, J2H Biotech, Saneop-ro 156 Beon-gil, Gwonseon-gu, Suwon-si, Gyeonggi-do, 16648, Republic of Korea
| | - Ju Young Lee
- R&D Center, J2H Biotech, Saneop-ro 156 Beon-gil, Gwonseon-gu, Suwon-si, Gyeonggi-do, 16648, Republic of Korea
| | - Haeng Ran Seo
- Advanced Biomedical Research Laboratory, Institut Pasteur Korea, 16, Daewangpangyo-ro 712 Beon-gil, Bundang-gu, Seongnam-si, Gyeonggi-do, 463-400, South Korea.
| |
Collapse
|
5
|
Ye H, Sun M, Huang S, Xu F, Wang J, Liu H, Zhang L, Luo W, Guo W, Wu Z, Zhu J, Li H. Gene Network Analysis of Hepatocellular Carcinoma Identifies Modules Associated with Disease Progression, Survival, and Chemo Drug Resistance. Int J Gen Med 2021; 14:9333-9347. [PMID: 34898998 PMCID: PMC8654693 DOI: 10.2147/ijgm.s336729] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Accepted: 11/08/2021] [Indexed: 12/11/2022] Open
Abstract
Background Hepatocellular carcinoma (HCC) is the second leading cause of cancer-related mortality worldwide. HCC transcriptome has been extensively studied; however, the progress in disease mechanisms, prognosis, and treatment is still slow. Methods A rank-based module-centric workflow was introduced to analyze important modules associated with HCC development, prognosis, and drug resistance. The currently largest HCC cell line RNA-Seq dataset from the LIMORE database was used to construct the reference modules by weighted gene co-expression network analysis. Results Thirteen reference modules were identified with validated reproducibility. These modules were all associated with specific biological functions. Differentially expressed module analysis revealed the crucial modules during HCC development. Modules and hub genes are indicative of patient survival. Modules can differentiate patients in different HCC stages. Furthermore, drug resistance was revealed by drug-module association analysis. Based on differentially expressed modules and hub genes, six candidate drugs were screened. The hub genes of those modules merit further investigation. Conclusion We proposed a reference module-based analysis of the HCC transcriptome. The identified modules are associated with HCC development, survival, and drug resistance. M3 and M6 may play important roles during HCV to HCC development. M1, M3, M5, and M7 are associated with HCC survival. High M4, high M9, low M1, and low M3 may be associated with dasatinib, doxorubicin, CD532, and simvastatin resistance. Our analysis provides useful information for HCC diagnosis and treatment.
Collapse
Affiliation(s)
- Hua Ye
- Department of Gastroenterology, Ningbo Medical Treatment Center Lihuili Hospital, Medical School of Ningbo University, Ningbo, Zhejiang, 315040, People's Republic of China
| | - Mengxia Sun
- Department of Clinical Medicine, Medical School of Ningbo University, Ningbo, Zhejiang, 315211, People's Republic of China
| | - Shiliang Huang
- Department of Gastroenterology, Ningbo Medical Treatment Center Lihuili Hospital, Medical School of Ningbo University, Ningbo, Zhejiang, 315040, People's Republic of China
| | - Feng Xu
- Department of Gastroenterology, Ningbo Medical Treatment Center Lihuili Hospital, Medical School of Ningbo University, Ningbo, Zhejiang, 315040, People's Republic of China
| | - Jian Wang
- Department of Dermatology, Ningbo Medical Treatment Center Lihuili Hospital, Medical School of Ningbo University, Ningbo, Zhejiang, 315040, People's Republic of China
| | - Huiwei Liu
- Department of Gastroenterology, Ningbo Medical Treatment Center Lihuili Hospital, Medical School of Ningbo University, Ningbo, Zhejiang, 315040, People's Republic of China
| | - Liangshun Zhang
- Department of Gastroenterology, Ningbo Medical Treatment Center Lihuili Hospital, Medical School of Ningbo University, Ningbo, Zhejiang, 315040, People's Republic of China
| | - Wenjing Luo
- Department of Gastroenterology, Ningbo Medical Treatment Center Lihuili Hospital, Medical School of Ningbo University, Ningbo, Zhejiang, 315040, People's Republic of China
| | - Wenying Guo
- Department of Gastroenterology, Ningbo Medical Treatment Center Lihuili Hospital, Medical School of Ningbo University, Ningbo, Zhejiang, 315040, People's Republic of China
| | - Zhe Wu
- Department of Gastroenterology, Ningbo Medical Treatment Center Lihuili Hospital, Medical School of Ningbo University, Ningbo, Zhejiang, 315040, People's Republic of China
| | - Jie Zhu
- Department of Hepatobiliary Surgery, Ningbo Medical Treatment Center Lihuili Hospital, Medical School of Ningbo University, Ningbo, Zhejiang, 315040, People's Republic of China
| | - Hong Li
- Department of Hepatobiliary Surgery, Ningbo Medical Treatment Center Lihuili Hospital, Medical School of Ningbo University, Ningbo, Zhejiang, 315040, People's Republic of China
| |
Collapse
|
6
|
Lue HW, Derrick DS, Rao S, Van Gaest A, Cheng L, Podolak J, Lawson S, Xue C, Garg D, White R, Ryan CW, Drake JM, Ritz A, Heiser LM, Thomas GV. Cabozantinib and dasatinib synergize to induce tumor regression in non-clear cell renal cell carcinoma. Cell Rep Med 2021; 2:100267. [PMID: 34095877 PMCID: PMC8149375 DOI: 10.1016/j.xcrm.2021.100267] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2020] [Revised: 01/18/2021] [Accepted: 04/13/2021] [Indexed: 12/24/2022]
Abstract
The lack of effective treatment options for advanced non-clear cell renal cell carcinoma (NCCRCC) is a critical unmet clinical need. Applying a high-throughput drug screen to multiple human kidney cancer cells, we identify the combination of the VEGFR-MET inhibitor cabozantinib and the SRC inhibitor dasatinib acts synergistically in cells to markedly reduce cell viability. Importantly, the combination is well tolerated and causes tumor regression in vivo. Transcriptional and phosphoproteomic profiling reveals that the combination converges to downregulate the MAPK-ERK signaling pathway, a result not predicted by single-agent analysis alone. Correspondingly, the addition of a MEK inhibitor synergizes with either dasatinib or cabozantinib to increase its efficacy. This study, by using approved, clinically relevant drugs, provides the rationale for the design of effective combination treatments in NCCRCC that can be rapidly translated to the clinic.
Collapse
Affiliation(s)
- Hui-wen Lue
- Knight Cancer Institute, Oregon Health and Science University, Portland, OR, USA
| | - Daniel S. Derrick
- Department of Biomedical Engineering, Oregon Health and Science University Center for Spatial Systems Biomedicine, Portland, OR, USA
| | - Soumya Rao
- Knight Cancer Institute, Oregon Health and Science University, Portland, OR, USA
| | - Ahna Van Gaest
- Knight Cancer Institute, Oregon Health and Science University, Portland, OR, USA
| | - Larry Cheng
- Graduate Program in Quantitative Biomedicine, Rutgers University, Piscataway, NJ, USA
| | - Jennifer Podolak
- Knight Cancer Institute, Oregon Health and Science University, Portland, OR, USA
| | - Samantha Lawson
- Knight Cancer Institute, Oregon Health and Science University, Portland, OR, USA
| | - Changhui Xue
- Knight Cancer Institute, Oregon Health and Science University, Portland, OR, USA
| | - Devin Garg
- Knight Cancer Institute, Oregon Health and Science University, Portland, OR, USA
| | - Ralph White
- Department of Pharmacology, University of Minnesota, Minneapolis, MN, USA
| | - Christopher W. Ryan
- Knight Cancer Institute, Oregon Health and Science University, Portland, OR, USA
- Division of Hematology and Oncology, Oregon Health and Science University, Portland, OR, USA
| | - Justin M. Drake
- Department of Pharmacology, University of Minnesota, Minneapolis, MN, USA
- Department of Urology, University of Minnesota, Minneapolis, MN, USA
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN, USA
| | - Anna Ritz
- Department of Biology, Reed College, Portland, OR, USA
| | - Laura M. Heiser
- Knight Cancer Institute, Oregon Health and Science University, Portland, OR, USA
- Department of Biomedical Engineering, Oregon Health and Science University Center for Spatial Systems Biomedicine, Portland, OR, USA
| | - George V. Thomas
- Knight Cancer Institute, Oregon Health and Science University, Portland, OR, USA
- Department of Pathology and Laboratory Medicine, Oregon Health and Science University, Portland, OR, USA
| |
Collapse
|
7
|
Suppressing Cdk5 Activity by Luteolin Inhibits MPP +-Induced Apoptotic of Neuroblastoma through Erk/Drp1 and Fak/Akt/GSK3β Pathways. Molecules 2021; 26:molecules26051307. [PMID: 33671094 PMCID: PMC7957557 DOI: 10.3390/molecules26051307] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Revised: 02/23/2021] [Accepted: 02/23/2021] [Indexed: 02/07/2023] Open
Abstract
Parkinson’s disease (PD) is characterized by the progressive degeneration of dopaminergic neurons. The cause of PD is still unclear. Oxidative stress and mitochondrial dysfunction have been linked to the development of PD. Luteolin, a non-toxic flavonoid, has become interested in an alternative medicine, according to its effects on anti-oxidative stress and anti-apoptosis, although the underlying mechanism of luteolin on PD has not been fully elucidated. This study aims to investigate whether luteolin prevents neurotoxicity induction by 1-methyl-4-phenylpyridinium iodide (MPP+), a neurotoxin in neuroblastoma SH-SY5Y cells. The results reveal that luteolin significantly improved cell viability and reduced apoptosis in MPP+-treated cells. Increasing lipid peroxidation and superoxide anion (O2−), including mitochondrial membrane potential (Δψm) disruption, is ameliorated by luteolin treatment. In addition, luteolin attenuated MPP+-induced neurite damage via GAP43 and synapsin-1. Furthermore, Cdk5 is found to be overactivated and correlated with elevation of cleaved caspase-3 activity in MPP+-exposed cells, while phosphorylation of Erk1/2, Drp1, Fak, Akt and GSK3β are inhibited. In contrast, luteolin attenuated Cdk5 overactivation and supported phosphorylated level of Erk1/2, Drp1, Fak, Akt and GSK3β with reducing in cleaved caspase-3 activity. Results indicate that luteolin exerts neuroprotective effects via Cdk5-mediated Erk1/2/Drp1 and Fak/Akt/GSK3β pathways, possibly representing a potential preventive agent for neuronal disorder.
Collapse
|
8
|
Li Q, Zhang Z, Fan Y, Zhang Q. Epigenetic Alterations in Renal Cell Cancer With TKIs Resistance: From Mechanisms to Clinical Applications. Front Genet 2021; 11:562868. [PMID: 33510766 PMCID: PMC7835797 DOI: 10.3389/fgene.2020.562868] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Accepted: 12/10/2020] [Indexed: 12/13/2022] Open
Abstract
The appearance of tyrosine kinase inhibitors (TKIs) has been a major breakthrough in renal cell carcinoma (RCC) therapy. Unfortunately, a portion of patients with TKIs resistance experience disease progression after TKIs therapy. Epigenetic alterations play an important role in the development of TKIs resistance. Current evidence suggests that epigenetic alterations occur frequently in RCC patients with poor response to TKIs therapy, and modulation of them could enhance the cytotoxic effect of antitumor therapy. In this review, we summarize the currently known epigenetic alterations relating to TKIs resistance in RCC, focusing on DNA methylation, non-coding RNAs (ncRNAs), histone modifications, and their interactions with TKIs treatment. In addition, we discuss application of epigenetic alteration analyses in the clinical setting to predict prognosis of patients with TKIs treatment, and the potential use of epigenetics-based therapies to surmount TKIs resistance.
Collapse
Affiliation(s)
- Qinhan Li
- Department of Urology, Peking University First Hospital, Institute of Urology, National Research Center for Genitourinary Oncology, Peking University, Beijing, China
| | - Zhenan Zhang
- Department of Urology, Peking University First Hospital, Institute of Urology, National Research Center for Genitourinary Oncology, Peking University, Beijing, China
| | - Yu Fan
- Department of Urology, Peking University First Hospital, Institute of Urology, National Research Center for Genitourinary Oncology, Peking University, Beijing, China
| | - Qian Zhang
- Department of Urology, Peking University First Hospital, Institute of Urology, National Research Center for Genitourinary Oncology, Peking University, Beijing, China
| |
Collapse
|
9
|
Li M, Li J, Guo X, Pan H, Zhou Q. Absence of HTATIP2 Expression in A549 Lung Adenocarcinoma Cells Promotes Tumor Plasticity in Response to Hypoxic Stress. Cancers (Basel) 2020; 12:cancers12061538. [PMID: 32545251 PMCID: PMC7352940 DOI: 10.3390/cancers12061538] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2020] [Revised: 06/05/2020] [Accepted: 06/08/2020] [Indexed: 12/15/2022] Open
Abstract
HIV-1 Tat Interactive Protein 2 (HTATIP2) is a tumor suppressor, of which reduced or absent expression is associated with increased susceptibility to tumorigenesis and enhanced tumor invasion and metastasis. However, whether the absent expression of HTATIP2 is a tumor-promoting factor that acts through improving tumor adaptation to hypoxia is unclear. Here, we established a stable HTATIP2-knockdown A549 human lung adenocarcinoma cell line (A549shHTATIP2) using lentiviral-delivered HTATIP2-targeting short hairpin RNA (shRNA), employed a double subcutaneous xenograft model and incorporated photoacoustic imaging and metabolomics approaches to elucidate the impact of the absent HTATIP2 expression on tumor response to hypoxic stress. Results from the in vivo study showed that A549shHTATIP2 tumors exhibited accelerated growth but decreased intratumoral oxygenation and angiogenesis and reduced sensitivity to sorafenib treatment as compared with their parental counterparts. Moreover, results of the immunoblot and real-time PCR analyses revealed that the HIF2α protein and mRNA levels in vehicle-treated A549shHTATIP2 tumors were significantly increased (p < 0.01 compared with the parental control tumors). Despite the strong HIF2α-c-Myc protein interaction indicated by our co-immunoprecipitation data, the increase in the c-Myc protein and mRNA levels was not significant in the A549shHTATIP2 tumors. Nonetheless, MCL-1 and β-catenin protein levels in A549shHTATIP2 tumors were significantly increased (p < 0.05 compared with the parental control tumors), suggesting an enhanced β-catenin/c-Myc/MCL-1 pathway in the absence of HTATIP2 expression. The finding of significantly decreased E-cadherin (p < 0.01 compared with vehicle-treated A549shHTATIP2 tumors) and increased vimentin (p < 0.05 compared with sorafenib-treated A549 tumors) protein levels in A549shHTATIP2 tumors implicates that the absence of HTATIP2 expression increases the susceptibility of A549 tumors to sorafenib-activated epithelial-mesenchymal transition (EMT) process. Comparison of the metabolomic profiles between A549 and A549shHTATIP2 tumors demonstrated that the absence of HTATIP2 expression resulted in increased tumor metabolic plasticity that enabled tumor cells to exploit alternative metabolic pathways for survival and proliferation rather than relying on glutamine and fatty acids as a carbon source to replenish TCA cycle intermediates. Our data suggest a mechanism by which the absent HTATIP2 expression modulates tumor adaptation to hypoxia and promotes an aggressive tumor phenotype by enhancing the HIF2α-regulated β-catenin/c-Myc/MCL-1 signaling, increasing the susceptibility of tumors to sorafenib treatment-activated EMT process, and improving tumor metabolic plasticity.
Collapse
Affiliation(s)
- Minghua Li
- Department of Pharmaceutical Sciences, Taneja College of Pharmacy, University of South Florida, Tampa, FL 33612, USA; (M.L.); (X.G.)
| | - Jing Li
- Karmanos Cancer Institute, Wayne State University School of Medicine, Detroit, MI 48201, USA;
| | - Xiaofang Guo
- Department of Pharmaceutical Sciences, Taneja College of Pharmacy, University of South Florida, Tampa, FL 33612, USA; (M.L.); (X.G.)
| | - Hua Pan
- Division of Cardiovascular Sciences, Morsani College of Medicine, University of South Florida, Tampa, FL 33612, USA;
| | - Qingyu Zhou
- Department of Pharmaceutical Sciences, Taneja College of Pharmacy, University of South Florida, Tampa, FL 33612, USA; (M.L.); (X.G.)
- Correspondence: ; Tel.: +1-813-974-7081
| |
Collapse
|
10
|
Ma X, Qiu Y, Zhu L, Zhao Y, Lin Y, Ma D, Qin Z, Sun C, Shen X, Li T, Han L. NOD1 inhibits proliferation and enhances response to chemotherapy via suppressing SRC-MAPK pathway in hepatocellular carcinoma. J Mol Med (Berl) 2019; 98:221-232. [PMID: 31872284 DOI: 10.1007/s00109-019-01868-9] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2019] [Revised: 11/25/2019] [Accepted: 12/10/2019] [Indexed: 12/13/2022]
Abstract
NOD1 is an innate immune sensor playing an important role in fighting against infection. However, its role in cancer is far from being clarified, and whether NOD1 plays a role in the progression of hepatocellular carcinoma (HCC) has never been reported. Here, we found that NOD1 expression was significantly decreased in hepatocellular carcinoma tissues and overexpression of NOD1 significantly inhibited tumorigenesis in vivo. In vitro experiments demonstrated that NOD1 inhibited proliferation of HCC cells by directly targeting proto-oncogene SRC and inducing cell cycle arrest at G1 phase. Further investigation showed that NOD1 exerted its antitumor effect by inhibiting SRC activation and further suppressing SRC/MAPK axis in hepatocellular carcinoma cells. Moreover, NOD1 dramatically enhanced the response of HCC cells to chemotherapy via inhibition of SRC-MAPK axis both in vitro and in vivo. Collectively, these data indicated that NOD1 suppressed proliferation and enhanced response to sorafenib or 5-FU treatment through inhibiting SRC-MAPK axis in hepatocellular carcinoma. KEY MESSAGES: NOD1 significantly inhibited tumorigenesis of HCC in cellular and animal models. NOD1 inhibited proliferation of HCC cells by inducing cell cycle arrest. NOD1 exerted its antitumor effect on HCC by directly interacting with SRC and inhibiting SRC-MAPK axis. NOD1 significantly enhanced the chemosensitivity of HCC cells to chemotherapeutic drugs.
Collapse
Affiliation(s)
- Xiaomin Ma
- Shandong Provincial Key Laboratory of Infection & Immunology, Department of Immunology, Shandong University School of Basic Medical Sciences, 44 Wenhua Xi Road, Jinan, 250012, China
| | - Yumin Qiu
- Shandong Provincial Key Laboratory of Infection & Immunology, Department of Immunology, Shandong University School of Basic Medical Sciences, 44 Wenhua Xi Road, Jinan, 250012, China
| | - Lihui Zhu
- Shandong Provincial Key Laboratory of Infection & Immunology, Department of Immunology, Shandong University School of Basic Medical Sciences, 44 Wenhua Xi Road, Jinan, 250012, China
| | - Yunxue Zhao
- Department of Pharmacology, Shandong University School of Basic Medical Sciences, Jinan, 250012, China
| | - Yueke Lin
- Shandong Provincial Key Laboratory of Infection & Immunology, Department of Immunology, Shandong University School of Basic Medical Sciences, 44 Wenhua Xi Road, Jinan, 250012, China
| | - Dapeng Ma
- Shandong Provincial Key Laboratory of Infection & Immunology, Department of Immunology, Shandong University School of Basic Medical Sciences, 44 Wenhua Xi Road, Jinan, 250012, China
| | - Zhenzhi Qin
- Shandong Provincial Key Laboratory of Infection & Immunology, Department of Immunology, Shandong University School of Basic Medical Sciences, 44 Wenhua Xi Road, Jinan, 250012, China
| | - Caiyu Sun
- Shandong Provincial Key Laboratory of Infection & Immunology, Department of Immunology, Shandong University School of Basic Medical Sciences, 44 Wenhua Xi Road, Jinan, 250012, China
| | - Xuecheng Shen
- Shandong Provincial Key Laboratory of Infection & Immunology, Department of Immunology, Shandong University School of Basic Medical Sciences, 44 Wenhua Xi Road, Jinan, 250012, China
| | - Tao Li
- Department of Gastroenterology, Provincial Hospital Affiliated with Shandong University, Jinan, 250021, China
| | - Lihui Han
- Shandong Provincial Key Laboratory of Infection & Immunology, Department of Immunology, Shandong University School of Basic Medical Sciences, 44 Wenhua Xi Road, Jinan, 250012, China.
| |
Collapse
|
11
|
Yan D, Dong W, He Q, Yang M, Huang L, Kong J, Qin H, Lin T, Huang J. Circular RNA circPICALM sponges miR-1265 to inhibit bladder cancer metastasis and influence FAK phosphorylation. EBioMedicine 2019; 48:316-331. [PMID: 31648990 PMCID: PMC6838432 DOI: 10.1016/j.ebiom.2019.08.074] [Citation(s) in RCA: 55] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2019] [Revised: 08/30/2019] [Accepted: 08/30/2019] [Indexed: 12/12/2022] Open
Abstract
Background Metastasis is a major obstacle in the treatment of bladder cancer (BC). Circular RNAs exert various functions in the aggressive biological behaviour of cancers. In this study, we aimed to elucidate how circPICALM influences BC metastasis. Methods The expression of circPICALM was analysed by real-time PCR. The tumourigenic properties of BC cells were evaluated using in vitro migration, invasion, and wound healing assays and an in vivo footpad model. The interaction between circPICALM and miR-1265 was confirmed by pull-down and dual-luciferase reporter assays and biotin-labelled miRNA capture. The interaction of STEAP4 and focal adhesion kinase (FAK) was confirmed by co-immunoprecipitation. Findings CircPICALM was downregulated in BC tissues, and low circPICALM expression was related to advanced T stage, high grade, lymph node positivity and poor overall survival. Overexpression of circPICALM inhibited the metastasis of BC cells, and DHX9 negatively regulated circPICALM levels. CircPICALM colocalized with miR-1265 and acted as a sponge for this miRNA, and the pro-invasion effect of miR-1265 was abolished by circPICALM overexpression. STEAP4, a target of miR-1265, suppressed metastasis; it bound to FAK to prevent autophosphorylation at Y397 and influenced EMT in BC cells. Interpretation CircPICALM can inhibit BC metastasis and bind to miR-1265 to block its pro-invasion activity. STEAP4 is a target of miR-1265 and is related to FAK phosphorylation and EMT. Fund This research was supported by National Natural Science Foundation of China, No.81772728, National Natural Science Foundation of China, No.81772719, National Natural Science Foundation of China No.81572514.
Collapse
Affiliation(s)
- Dong Yan
- Department of Urology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China; Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Wei Dong
- Department of Urology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China; Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Qingqing He
- Department of Urology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China; Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Meihua Yang
- Department of Urology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China; Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Lifang Huang
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Jianqiu Kong
- Department of Urology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China; Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Haide Qin
- Department of Urology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China; Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Tianxin Lin
- Department of Urology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China; Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China.
| | - Jian Huang
- Department of Urology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China; Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China.
| |
Collapse
|
12
|
Wei Z, Zhongqiu T, Lu S, Zhang F, Xie W, Wang Y. Gene coexpression analysis offers important modules and pathway of human lung adenocarcinomas. J Cell Physiol 2019; 235:454-464. [PMID: 31264215 DOI: 10.1002/jcp.28985] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2019] [Accepted: 05/07/2019] [Indexed: 12/24/2022]
Abstract
Lung adenocarcinomas injured greatly on the people worldwide. Although clinic experiments and gene profiling analyses had been well performed, to our knowledge, systemic coexpression analysis of human genes for this cancer is still limited to date. Here, using the published data GSE75037, we built the coexpression modules of genes by Weighted Gene Co-Expression Network Analysis (WGCNA), and investigated function and protein-protein interaction network of coexpression genes by Database for Annotation, visualization, and Integrated Discovery (DAVID) and String database, respectively. First, 11 coexpression modules were conducted for 5,000 genes in the 83 samples recently. Number of genes for each module ranged from 90 to 1,260, with the mean of 454. Second, interaction relationships of hub-genes between pairwise modules showed great differences, suggesting relatively high scale independence of the modules. Third, functional enrichment of the coexpression modules showed great differences. We found that genes in modules 8 significantly enriched in the biological process and/or pathways of cell adhesion, extracellular matrix (ECM)-receptor interaction, focal adhesion, and PI3K-Akt signaling pathway, and so forth. It was inferred as the key module underlying lung adenocarcinomas. Furthermore, PPI analysis revealed that the genes COL1A1, COL1A2, COL3A1, CTGF, and BGN owned the largest number of adjacency genes, unveiling that they may functioned importantly during the occurrence of lung adenocarcinomas. To summary, genes involved in cell adhesion, ECM-receptor interaction, focal adhesion, and PI3K-Akt signaling pathway play crucial roles in human lung adenocarcinomas.
Collapse
Affiliation(s)
- Zhongheng Wei
- Department of Oncology, Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, Guangxi, China
| | - Tan Zhongqiu
- Department of Oncology, Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, Guangxi, China
| | - Shuxiong Lu
- Department of Pathology, Huai'an Maternal and Child Health Care Center of Jiangsu Province Affiliated Hospital of Yangzhou University, Huai'an, China
| | - Fang Zhang
- School of Medicine, Fudan University, Shanghai, China
| | - Wei Xie
- Department of Radiology, Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ying Wang
- Respiratory Medicine Department, The First People's Hospital of Tianmen, Tianmen, Hubei, China
| |
Collapse
|
13
|
Liu X, Su C, Xu J, Zhou D, Yan H, Li W, Chen G, Zhang N, Xu D, Hu H. Immunohistochemical analysis of matrix metalloproteinase-9 predicts papillary thyroid carcinoma prognosis. Oncol Lett 2019; 17:2308-2316. [PMID: 30675296 PMCID: PMC6341782 DOI: 10.3892/ol.2018.9850] [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: 03/24/2018] [Accepted: 11/14/2018] [Indexed: 02/06/2023] Open
Abstract
The present study aimed to explore the association between immunohistochemical matrix metalloproteinase-9 (MMP-9) expression and the clinicopathological characteristics of patients with papillary thyroid carcinoma (PTC), and to determine whether it may be used as a diagnostic or prognostic tool for PTC. Immunohistochemical staining of MMP-9 was performed in thyroid tissues obtained from 112 patients with PTC and 42 subjects with benign thyroid nodules (BTNs). The receiver operating characteristic curve was used to evaluate the legitimacy of MMP-9 as a diagnostic tool for PTC, and a predictor for structurally persistent/recurrent disease (SPRD) and disease status. Cox regression was applied to identify the risk factors of disease status and SPRD. The present study revealed that MMP-9 was overexpressed in PTC tissues, compared with in BTN tissues. Furthermore, MMP-9 scores yielded an area under the curve (AUC) of 0.842 (95% CI, 0.776-0.908) for differentially diagnosing PTC from BTN. In addition, the MMP-9 score was greater if patients previously had central lymph node metastasis, lateral lymph node metastasis or an advanced tumor-node-metastasis stage (III+IV). When MMP-9 was employed to predict disease status and SPRD, an AUC of 0.811 (95% CI, 0.706-0.917) and 0.806 (95% CI, 0.620-0.992) was obtained, respectively. A tumor size of >2 cm and an MMP-9 staining score of ≥6 were independent risk factors for predicting disease status, whereas vascular invasion and an MMP-9 staining score of ≥8 were risk factors for predicting SPRD. Furthermore, an MMP-9 staining score of ≥6 and ≥8 indicated shortened disease-free survival and survival without SPRD, respectively. In conclusion, the assessment of MMP-9 expression in thyroid carcinoma samples may represent a potential and supplementary tool for the diagnosis and prognostic prediction of PTC.
Collapse
Affiliation(s)
- Xingkai Liu
- Department of Hepatobiliary and Pancreatic Surgery, The First Hospital of Jilin University, Changchun, Jilin 130021, P.R. China
| | - Chang Su
- Department of Thyroid Surgery, The First Hospital of Jilin University, Changchun, Jilin 130021, P.R. China
| | - Jing Xu
- Cardiovascular Disease Center, The First Hospital of Jilin University, Changchun, Jilin 130021, P.R. China
| | - Dan Zhou
- Department of Pediatrics, The Second Hospital of Jilin University, Changchun, Jilin 130041, P.R. China
| | - He Yan
- Department of Emergency, The First Hospital of Jilin University, Changchun, Jilin 130021, P.R. China
| | - Wei Li
- Department of Emergency, The First Hospital of Jilin University, Changchun, Jilin 130021, P.R. China
| | - Guihui Chen
- Department of Pathology, Jilin City People's Hospital, Jilin, Jilin 132000, P.R. China
| | - Nan Zhang
- Department of Emergency, The First Hospital of Jilin University, Changchun, Jilin 130021, P.R. China
| | - Dahai Xu
- Department of Emergency, The First Hospital of Jilin University, Changchun, Jilin 130021, P.R. China
| | - Haixia Hu
- Department of Emergency, The First Hospital of Jilin University, Changchun, Jilin 130021, P.R. China
| |
Collapse
|
14
|
Cucurbitacin B induces mitochondrial-mediated apoptosis pathway in cholangiocarcinoma cells via suppressing focal adhesion kinase signaling. Naunyn Schmiedebergs Arch Pharmacol 2018; 392:271-278. [DOI: 10.1007/s00210-018-1584-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2018] [Accepted: 11/12/2018] [Indexed: 01/08/2023]
|
15
|
Das TK, Esernio J, Cagan RL. Restraining Network Response to Targeted Cancer Therapies Improves Efficacy and Reduces Cellular Resistance. Cancer Res 2018; 78:4344-4359. [DOI: 10.1158/0008-5472.can-17-2001] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2017] [Revised: 10/04/2017] [Accepted: 05/21/2018] [Indexed: 11/16/2022]
|
16
|
Taylor KN, Schlaepfer DD. Adaptive Resistance to Chemotherapy, A Multi-FAK-torial Linkage. Mol Cancer Ther 2018; 17:719-723. [PMID: 29610281 PMCID: PMC6538033 DOI: 10.1158/1535-7163.mct-17-1177] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2017] [Revised: 01/17/2018] [Accepted: 01/23/2018] [Indexed: 01/08/2023]
Abstract
Oncogenes provide tumor cells with a growth and survival advantage. Directed therapies targeted to oncogenic mutations (such as BRAF V600E) are part of effective late-stage melanoma treatment. However, tumors with BRAF V600E mutations, in approximately 10% of colorectal cancer, are generally treatment-insensitive. Research has identified various "feedback" mechanisms that result in BRAF signal pathway reactivation in response to BRAF inhibition. Herein, we highlight key findings from Chen and colleagues (this issue) showing that integrin-associated focal adhesion kinase (FAK) activation selectively occurs in BRAF V600E-mutant colorectal cancer cells in response to pharmacological BRAF inhibition. FAK activation results in elevated β-catenin protein levels, β-catenin nuclear localization, and increased gene transcription. Small-molecule inhibitors of β-catenin or FAK synergize with vemurafenib BRAF inhibitor to prevent BRAF V600E colorectal cancer cell proliferation in vitro and xenograft tumor growth in mice. This study complements findings linking FAK to β-catenin in intestinal tumorigenesis, resistance to radiotherapy, and cancer stem cell survival. Thus, FAK activation may occur as a frequent tumor cell "adaptive resistance" mechanism. Although FAK (PTK2) is not mutated in most cancers, targeting FAK activity in combinational approaches may limit tumor cell escape mechanisms and enhance durable responses to treatment. Mol Cancer Ther; 17(4); 719-23. ©2018 AACR.
Collapse
Affiliation(s)
- Kristin N Taylor
- Department of Reproductive Medicine, University of California, San Diego, School of Medicine, Moores Cancer Center, La Jolla, California
| | - David D Schlaepfer
- Department of Reproductive Medicine, University of California, San Diego, School of Medicine, Moores Cancer Center, La Jolla, California.
| |
Collapse
|
17
|
Fu L, Guo L, Zheng Y, Zhu Z, Zhang M, Zhao X, Cui H. Synergistic antitumor activity of low-dose c-Met tyrosine kinase inhibitor and sorafenib on human non-small cell lung cancer cells. Oncol Lett 2018; 15:5081-5086. [PMID: 29552141 DOI: 10.3892/ol.2018.7933] [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: 04/12/2017] [Accepted: 12/08/2017] [Indexed: 11/06/2022] Open
Abstract
Sorafenib is a multikinase inhibitor that is frequently used to treat various types of malignant tumors. However, it has been demonstrated that Sorafenib only has a moderate antitumor efficacy and is associated with numerous side effects in non-small cell lung cancer (NSCLC), which greatly limits its clinical application. The present study aimed to examine the effects of a combination of Sorafenib and low-dose PF-2341066, a selective c-Met tyrosine kinase inhibitor, on the proliferation, apoptosis and migration of the NSCLC cell line NCI-H1993. The data indicated that treatment with a combination of Sorafenib and low-dose PF-2341066 was able to significantly inhibit the proliferation and migration as well as promote the apoptosis, of NCI-H1993 cells, compared with treatment with Sorafenib or low-dose PF-2341066 alone. Further experiments indicated that the levels of phosphorylated epidermal growth factor receptor and c-Met were significantly decreased following the combined treatment of Sorafenib and PF-2341066, compared with the treatment with Sorafenib or PF-2341066 alone. The findings of the present study indicated that using a low-dose c-Met inhibitor enhances the antitumor activity of Sorafenib in NSCLC and may provide a novel strategy for the treatment of NSCLC.
Collapse
Affiliation(s)
- Ling Fu
- Department of Oncology, Affiliated Hospital of Weifang Medical University, Weifang, Shandong 261031, P.R. China
| | - Liang Guo
- Department of Cardio-Thoracic Surgery, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200092, P.R. China
| | - Yi Zheng
- Department of Oncology, Affiliated Hospital of Weifang Medical University, Weifang, Shandong 261031, P.R. China
| | - Zhenyu Zhu
- Surgical Ward 4, Shandong Tumor Hospital, Jinan, Shandong 250117, P.R. China
| | - Mingyue Zhang
- Department of Thoracic Surgery, Affiliated Hospital of Weifang Medical University, Weifang, Shandong 261031, P.R. China
| | - Xiaohua Zhao
- Department of Thoracic Surgery, Affiliated Hospital of Weifang Medical University, Weifang, Shandong 261031, P.R. China
| | - Hongxue Cui
- Department of Thoracic Surgery, Affiliated Hospital of Weifang Medical University, Weifang, Shandong 261031, P.R. China
| |
Collapse
|
18
|
Heavey S, Dowling P, Moore G, Barr MP, Kelly N, Maher SG, Cuffe S, Finn SP, O'Byrne KJ, Gately K. Development and characterisation of a panel of phosphatidylinositide 3-kinase - mammalian target of rapamycin inhibitor resistant lung cancer cell lines. Sci Rep 2018; 8:1652. [PMID: 29374181 PMCID: PMC5786033 DOI: 10.1038/s41598-018-19688-1] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2017] [Accepted: 01/05/2018] [Indexed: 12/19/2022] Open
Abstract
The PI3K-mTOR pathway is involved in regulating all hallmarks of cancer, and is often dysregulated in NSCLC, making it an attractive therapeutic target in this setting. Acquired resistance to PI3K-mTOR inhibition is a major hurdle to overcome in the success of PI3K-mTOR targeted agents. H460, A549, and H1975 resistant cells were generated by prolonged treatment in culture with Apitolisib (GDC-0980), a dual PI3K-mTOR inhibitor over a period of several months, from age-matched parent cells. Resistance was deemed to have developed when a log fold difference in IC50 had been achieved. Resistant cell lines also exhibited resistance to another widely investigated PI3K-mTOR dual inhibitor; Dactolisib (BEZ235). Cell lines were characterised at the level of mRNA (expression array profiling expression of >150 genes), miRNA (expression array profiling of 2100 miRNAs), protein (bottoms-up label-free mass spectrometry) and phosphoprotein (expression array profiling of 84 phospho/total proteins). Key alterations were validated by qPCR and Western blot. H1975 cells were initially most sensitive to Apitolisib (GDC-0980), but developed resistance more quickly than the other cell lines, perhaps due to increased selective pressure from the impressive initial effect. In-depth molecular profiling suggested epithelial-mesenchymal transition (EMT) may play a role in resistance to PI3K-mTOR dual inhibition in NSCLC.
Collapse
Affiliation(s)
- Susan Heavey
- Thoracic Oncology Research Group, Trinity Translational Medicine Institute, Trinity College Dublin, Ireland.
| | | | - Gillian Moore
- Thoracic Oncology Research Group, Trinity Translational Medicine Institute, Trinity College Dublin, Ireland
| | - Martin P Barr
- Thoracic Oncology Research Group, Trinity Translational Medicine Institute, Trinity College Dublin, Ireland
| | - Niamh Kelly
- Thoracic Oncology Research Group, Trinity Translational Medicine Institute, Trinity College Dublin, Ireland
| | - Stephen G Maher
- Department of Surgery, Trinity Translational Medicine Institute, Trinity College Dublin, Ireland
| | - Sinead Cuffe
- Thoracic Oncology Research Group, Trinity Translational Medicine Institute, Trinity College Dublin, Ireland
| | - Stephen P Finn
- Thoracic Oncology Research Group, Trinity Translational Medicine Institute, Trinity College Dublin, Ireland
| | | | - Kathy Gately
- Thoracic Oncology Research Group, Trinity Translational Medicine Institute, Trinity College Dublin, Ireland
| |
Collapse
|