1
|
García-Hernández L, Dai L, Rodríguez-Ulloa A, Yi Y, González LJ, Besada V, Li W, Perea SE, Perera Y. Time- and dose-dependent effects of CIGB-300 on the proteome of lung squamous cell carcinoma. Biol Chem 2025:hsz-2024-0149. [PMID: 40261874 DOI: 10.1515/hsz-2024-0149] [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: 12/05/2024] [Accepted: 03/13/2025] [Indexed: 04/24/2025]
Abstract
Proteome-wide scale in a dose - and time-depending setting is crucial to fully understand the pharmacological mechanism of anticancer drugs as well as identification of candidates for drug response biomarkers. Here, we investigated the effect of the CIGB-300 anticancer peptide at IC50 and IC80 doses during 1 and 4 h of treatment on the squamous lung cancer cell (NCI-H226) proteome. An overwhelming dose-dependent inhibitory effect with minor up-regulated proteins was observed by increasing CIGB-300 dose level. Functional enrichment was also CIGB-300 dose-dependent with common or exclusively regulated proteins in each dose and time settings. A protein core involving small molecule biosynthesis, aldehyde metabolism and metabolism of nucleobases was regulated irrespectively to the dose or the treatment time. Importantly, a group of proteins linked to NSCLC tumor biology, poor clinical outcome and some Protein Kinase CK2 substrates, were significantly regulated by treating with both CIGB-300 doses. Likewise, we observed a consistent downregulation of different proteins that had been already reported to be inhibited by CIGB-300 in lung adenocarcinoma and acute myeloid leukemia. Overall, our proteomics-guided strategy based on time and drug dose served to uncover novel clues supporting the CIGB-300 cytotoxic effect and also to identify putative pharmacodynamic biomarkers in NSCLC.
Collapse
Affiliation(s)
- Liudy García-Hernández
- Department of System Biology, Center for Genetic Engineering & Biotechnology (CIGB), 31 Ave, Havana 10600, Cuba
| | - Lingfeng Dai
- Research Department, China-Cuba Biotechnology Joint Innovation Center (CCBJIC), Yongzhou Zhong Gu Biotechnology Co., Ltd, Yangjiaqiao Street, Lengshuitan District, Yongzhou City 425000, Hunan Province, China
| | - Arielis Rodríguez-Ulloa
- Department of System Biology, Center for Genetic Engineering & Biotechnology (CIGB), 31 Ave, Havana 10600, Cuba
| | - Ying Yi
- Research Department, China-Cuba Biotechnology Joint Innovation Center (CCBJIC), Yongzhou Zhong Gu Biotechnology Co., Ltd, Yangjiaqiao Street, Lengshuitan District, Yongzhou City 425000, Hunan Province, China
| | - Luis J González
- Department of System Biology, Center for Genetic Engineering & Biotechnology (CIGB), 31 Ave, Havana 10600, Cuba
| | - Vladimir Besada
- Department of System Biology, Center for Genetic Engineering & Biotechnology (CIGB), 31 Ave, Havana 10600, Cuba
| | - Wen Li
- Research Department, China-Cuba Biotechnology Joint Innovation Center (CCBJIC), Yongzhou Zhong Gu Biotechnology Co., Ltd, Yangjiaqiao Street, Lengshuitan District, Yongzhou City 425000, Hunan Province, China
| | - Silvio E Perea
- Department of Pharmaceuticals, Center for Genetic Engineering & Biotechnology (CIGB), 31 Ave, Playa, Havana 10600, Cuba
| | - Yasser Perera
- Research Department, China-Cuba Biotechnology Joint Innovation Center (CCBJIC), Yongzhou Zhong Gu Biotechnology Co., Ltd, Yangjiaqiao Street, Lengshuitan District, Yongzhou City 425000, Hunan Province, China
| |
Collapse
|
2
|
Jeon K, Park S, Pak ES, Kim J, Liu Y, Hwang S, Na Y, Kwon Y. Calpain 2 Isoform-Specific Cleavage of Filamin A Enhances HIF1α Nuclear Translocation, Promoting Metastasis in Triple-Negative Breast Cancer. MedComm (Beijing) 2025; 6:e70147. [PMID: 40151834 PMCID: PMC11949505 DOI: 10.1002/mco2.70147] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Revised: 01/14/2025] [Accepted: 01/20/2025] [Indexed: 03/29/2025] Open
Abstract
Triple-negative breast cancer (TNBC) remains a challenge due to its aggressive nature and limited therapeutic options. Calpain 2, a member of the calcium-dependent cysteine protease family, is particularly associated with poor prognosis in TNBC. This study explores the isoform-specific role of calpain 2 in TNBC, examining its correlation with prognosis and its mechanistic impact on metastasis. Bioinformatic analyses, including Kaplan-Meier survival plots, univariate Cox proportional analysis, and gene set enrichment analysis (GSEA), assessed CAPN2 expression and its association with mesenchymal genes in TNBC. Results of cell-based experiments with CAPN2 knockdown or overexpression indicate that elevated CAPN2 expression correlates with poor clinical outcomes and enhanced metastatic potential in TNBC. CAPN2 knockdown inhibited the epithelial-mesenchymal transition (EMT), reducing cancer cell proliferation, migration, and invasion. Calpain 2 downregulation reversed the EMT by reducing isoform-specific cleavage of filamin A, HIF1α nuclear localization and TWIST1 transcription. CNa 29, a calpain 2-specific inhibitor, reduced cancer cell proliferation, decreased filamin A cleavage, downregulated TWIST1 expression, and significantly retarded metastasis,. In conclusion, calpain 2 plays a critical role in TNBC progression by modulating HIF1α and TWIST1, to promote the EMT and metastasis. Isoform-selective inhibition of calpain 2 with CNa 29 presents a promising therapeutic strategy for managing TNBC.
Collapse
Affiliation(s)
- Kyung‐Hwa Jeon
- College of PharmacyGraduate School of Pharmaceutical SciencesEwha Womans UniversitySeoulRepublic of Korea
- Gradutate Program in Innovative Biomaterials ConvergenceEwha Womans UniversitySeoulRepublic of Korea
| | - Seojeong Park
- College of PharmacyGraduate School of Pharmaceutical SciencesEwha Womans UniversitySeoulRepublic of Korea
| | - Eun Seon Pak
- College of PharmacyGraduate School of Pharmaceutical SciencesEwha Womans UniversitySeoulRepublic of Korea
- Gradutate Program in Innovative Biomaterials ConvergenceEwha Womans UniversitySeoulRepublic of Korea
| | - Jeong‐Ahn Kim
- College of PharmacyGraduate School of Pharmaceutical SciencesEwha Womans UniversitySeoulRepublic of Korea
| | - Yi Liu
- College of PharmacyGraduate School of Pharmaceutical SciencesEwha Womans UniversitySeoulRepublic of Korea
| | - Soo‐Yeon Hwang
- College of PharmacyGraduate School of Pharmaceutical SciencesEwha Womans UniversitySeoulRepublic of Korea
| | - Younghwa Na
- College of PharmacyCHA UniversityGyeongghi‐doRepublic of Korea
| | - Youngjoo Kwon
- College of PharmacyGraduate School of Pharmaceutical SciencesEwha Womans UniversitySeoulRepublic of Korea
- Gradutate Program in Innovative Biomaterials ConvergenceEwha Womans UniversitySeoulRepublic of Korea
| |
Collapse
|
3
|
Hanitrarimalala V, Bednarska I, Murakami T, Papadakos KS, Blom AM. Intracellular cartilage oligomeric matrix protein augments breast cancer resistance to chemotherapy. Cell Death Dis 2024; 15:480. [PMID: 38965233 PMCID: PMC11224260 DOI: 10.1038/s41419-024-06872-7] [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: 11/13/2023] [Revised: 06/26/2024] [Accepted: 06/27/2024] [Indexed: 07/06/2024]
Abstract
Chemotherapy persists as the primary intervention for breast cancer, with chemoresistance posing the principal obstacle to successful treatment. Herein, we show that cartilage oligomeric matrix protein (COMP) expression leads to increased cancer cell survival and attenuated apoptosis under treatment with several chemotherapeutic drugs, anti-HER2 targeted treatment, and endocrine therapy in several breast cancer cell lines tested. The COMP-induced chemoresistance was independent of the breast cancer subtype. Extracellularly delivered recombinant COMP failed to rescue cells from apoptosis while endoplasmic reticulum (ER)-restricted COMP-KDEL conferred resistance to apoptosis, consistent with the localization of COMP in the ER, where it interacted with calpain. Calpain activation was reduced in COMP-expressing cells and maintained at a lower level of activation during treatment with epirubicin. Moreover, the downstream caspases of calpain, caspases -9, -7, and -3, exhibited significantly reduced activation in COMP-expressing cells under chemotherapy treatment. Chemotherapy, when combined with calpain activators, rendered the cells expressing COMP more chemosensitive. Also, the anti-apoptotic proteins phospho-Bcl2 and survivin were increased in COMP-expressing cells upon chemotherapy. Cells expressing a mutant COMP lacking thrombospondin repeats exhibited reduced chemoresistance compared to cells expressing full-length COMP. Evaluation of calcium levels in the ER, cytosol, and mitochondria revealed that COMP expression modulates intracellular calcium homeostasis. Furthermore, patients undergoing chemotherapy or endocrine therapy demonstrated significantly reduced overall survival time when tumors expressed high levels of COMP. This study identifies a novel role of COMP in chemoresistance and calpain inactivation in breast cancer, a discovery with potential implications for anti-cancer therapy.
Collapse
Affiliation(s)
| | - Izabela Bednarska
- Department of Translational Medicine, Lund University, Malmö, S-214 28, Sweden
| | - Takashi Murakami
- Department of Microbiology, Saitama Medical University, Saitama, 350-0495, Japan
| | | | - Anna M Blom
- Department of Translational Medicine, Lund University, Malmö, S-214 28, Sweden
| |
Collapse
|
4
|
Wang Y, Chang Z, Ouyang M, Wang K, Gao X, Tang B. Advancing Nonsmall Cell Lung Cancer Diagnosis Accuracy via Dual Detection Fluorescent Nanoprobes. Anal Chem 2024; 96:6812-6818. [PMID: 38634576 DOI: 10.1021/acs.analchem.4c00794] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/19/2024]
Abstract
Among the primary threats to human health worldwide, nonsmall cell lung cancer (NSCLC) remains a significant factor and is a leading cause of cancer-related deaths. Due to subtle early symptoms, NSCLC patients are diagnosed at advanced stages, resulting in low survival rates. Herein, novel Au-Se bond nanoprobes (NPs) designed for the specific detection of Calpain-2 (CAPN2) and Human Neutrophil Elastase (HNE), pivotal biomarkers in NSCLC, were developed. The NPs demonstrated exceptional specificity and sensitivity toward CAPN2 and HNE, enabling dual-color fluorescence imaging to distinguish between NSCLC cells and normal lung cells effectively. The NPs' performance was consistent across a wide pH range (6.2 to 8.0), and it exhibited remarkable resistance to biological thiol interference, indicating its robustness in complex physiological environments. These findings suggest the nanoprobe is a promising tool for early NSCLC diagnosis, offering a novel approach for enhancing the accuracy of cancer detection.
Collapse
Affiliation(s)
- Yinian Wang
- College of Chemistry, Chemical Engineering and Materials Science, Collaborative Innovation Center of Functionalized Probes for Chemical Imaging in Universities of Shandong, Key Laboratory of Molecular and Nano Probes, Ministry of Edu-cation, Shandong Provincial Key Laboratory of Clean Production of Fine Chemicals, Shandong Normal University, Jinan 250014, P. R. China
| | - Zixuan Chang
- College of Chemistry, Chemical Engineering and Materials Science, Collaborative Innovation Center of Functionalized Probes for Chemical Imaging in Universities of Shandong, Key Laboratory of Molecular and Nano Probes, Ministry of Edu-cation, Shandong Provincial Key Laboratory of Clean Production of Fine Chemicals, Shandong Normal University, Jinan 250014, P. R. China
| | - Mingyi Ouyang
- College of Chemistry, Chemical Engineering and Materials Science, Collaborative Innovation Center of Functionalized Probes for Chemical Imaging in Universities of Shandong, Key Laboratory of Molecular and Nano Probes, Ministry of Edu-cation, Shandong Provincial Key Laboratory of Clean Production of Fine Chemicals, Shandong Normal University, Jinan 250014, P. R. China
| | - Keyi Wang
- College of Chemistry, Chemical Engineering and Materials Science, Collaborative Innovation Center of Functionalized Probes for Chemical Imaging in Universities of Shandong, Key Laboratory of Molecular and Nano Probes, Ministry of Edu-cation, Shandong Provincial Key Laboratory of Clean Production of Fine Chemicals, Shandong Normal University, Jinan 250014, P. R. China
| | - Xiaonan Gao
- College of Chemistry, Chemical Engineering and Materials Science, Collaborative Innovation Center of Functionalized Probes for Chemical Imaging in Universities of Shandong, Key Laboratory of Molecular and Nano Probes, Ministry of Edu-cation, Shandong Provincial Key Laboratory of Clean Production of Fine Chemicals, Shandong Normal University, Jinan 250014, P. R. China
| | - Bo Tang
- College of Chemistry, Chemical Engineering and Materials Science, Collaborative Innovation Center of Functionalized Probes for Chemical Imaging in Universities of Shandong, Key Laboratory of Molecular and Nano Probes, Ministry of Edu-cation, Shandong Provincial Key Laboratory of Clean Production of Fine Chemicals, Shandong Normal University, Jinan 250014, P. R. China
- Laoshan Laboratory, 168 Wenhai Middle Rd, Aoshanwei Jimo, Qingdao ,Shandong266237, P. R. China
| |
Collapse
|
5
|
Díaz-Campos MÁ, Vasquez-Arriaga J, Ochoa S, Hernández-Lemus E. Functional impact of multi-omic interactions in lung cancer. Front Genet 2024; 15:1282241. [PMID: 38389572 PMCID: PMC10881857 DOI: 10.3389/fgene.2024.1282241] [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: 08/23/2023] [Accepted: 01/23/2024] [Indexed: 02/24/2024] Open
Abstract
Lung tumors are a leading cause of cancer-related death worldwide. Lung cancers are highly heterogeneous on their phenotypes, both at the cellular and molecular levels. Efforts to better understand the biological origins and outcomes of lung cancer in terms of this enormous variability often require of high-throughput experimental techniques paired with advanced data analytics. Anticipated advancements in multi-omic methodologies hold potential to reveal a broader molecular perspective of these tumors. This study introduces a theoretical and computational framework for generating network models depicting regulatory constraints on biological functions in a semi-automated way. The approach successfully identifies enriched functions in analyzed omics data, focusing on Adenocarcinoma (LUAD) and Squamous cell carcinoma (LUSC, a type of NSCLC) in the lung. Valuable information about novel regulatory characteristics, supported by robust biological reasoning, is illustrated, for instance by considering the role of genes, miRNAs and CpG sites associated with NSCLC, both novel and previously reported. Utilizing multi-omic regulatory networks, we constructed robust models elucidating omics data interconnectedness, enabling systematic generation of mechanistic hypotheses. These findings offer insights into complex regulatory mechanisms underlying these cancer types, paving the way for further exploring their molecular complexity.
Collapse
Affiliation(s)
| | - Jorge Vasquez-Arriaga
- Computational Genomics Division, National Institute of Genomic Medicine, Mexico City, Mexico
| | - Soledad Ochoa
- Computational Genomics Division, National Institute of Genomic Medicine, Mexico City, Mexico
- Department of Obstetrics and Gynecology, Cedars-Sinai Medical Center, Los Angeles, CA, United States
| | - Enrique Hernández-Lemus
- Computational Genomics Division, National Institute of Genomic Medicine, Mexico City, Mexico
- Center for Complexity Sciences, Universidad Nacional Autónoma de México, Mexico City, Mexico
| |
Collapse
|
6
|
Zheng X, Song X, Zhu G, Pan D, Li H, Hu J, Xiao K, Gong Q, Gu Z, Luo K, Li W. Nanomedicine Combats Drug Resistance in Lung Cancer. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2308977. [PMID: 37968865 DOI: 10.1002/adma.202308977] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/02/2023] [Revised: 11/03/2023] [Indexed: 11/17/2023]
Abstract
Lung cancer is the second most prevalent cancer and the leading cause of cancer-related death worldwide. Surgery, chemotherapy, molecular targeted therapy, immunotherapy, and radiotherapy are currently available as treatment methods. However, drug resistance is a significant factor in the failure of lung cancer treatments. Novel therapeutics have been exploited to address complicated resistance mechanisms of lung cancer and the advancement of nanomedicine is extremely promising in terms of overcoming drug resistance. Nanomedicine equipped with multifunctional and tunable physiochemical properties in alignment with tumor genetic profiles can achieve precise, safe, and effective treatment while minimizing or eradicating drug resistance in cancer. Here, this work reviews the discovered resistance mechanisms for lung cancer chemotherapy, molecular targeted therapy, immunotherapy, and radiotherapy, and outlines novel strategies for the development of nanomedicine against drug resistance. This work focuses on engineering design, customized delivery, current challenges, and clinical translation of nanomedicine in the application of resistant lung cancer.
Collapse
Affiliation(s)
- Xiuli Zheng
- Department of Radiology, Department of Respiratory, Huaxi MR Research Center (HMRRC) and Critical Care Medicine, Institute of Respiratory Health, Precision Medicine Center, Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, No. 37 Guoxue Alley, Chengdu, 610041, China
| | - Xiaohai Song
- Department of General Surgery, Gastric Cancer Center and Laboratory of Gastric Cancer, West China Hospital, Sichuan University, No. 37 Guoxue Alley, Chengdu, 610041, China
| | - Guonian Zhu
- Department of Radiology, Department of Respiratory, Huaxi MR Research Center (HMRRC) and Critical Care Medicine, Institute of Respiratory Health, Precision Medicine Center, Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, No. 37 Guoxue Alley, Chengdu, 610041, China
| | - Dayi Pan
- Department of Radiology, Department of Respiratory, Huaxi MR Research Center (HMRRC) and Critical Care Medicine, Institute of Respiratory Health, Precision Medicine Center, Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, No. 37 Guoxue Alley, Chengdu, 610041, China
| | - Haonan Li
- Department of Radiology, Department of Respiratory, Huaxi MR Research Center (HMRRC) and Critical Care Medicine, Institute of Respiratory Health, Precision Medicine Center, Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, No. 37 Guoxue Alley, Chengdu, 610041, China
| | - Jiankun Hu
- Department of General Surgery, Gastric Cancer Center and Laboratory of Gastric Cancer, West China Hospital, Sichuan University, No. 37 Guoxue Alley, Chengdu, 610041, China
| | - Kai Xiao
- Department of Radiology, Department of Respiratory, Huaxi MR Research Center (HMRRC) and Critical Care Medicine, Institute of Respiratory Health, Precision Medicine Center, Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, No. 37 Guoxue Alley, Chengdu, 610041, China
| | - Qiyong Gong
- Department of Radiology, Department of Respiratory, Huaxi MR Research Center (HMRRC) and Critical Care Medicine, Institute of Respiratory Health, Precision Medicine Center, Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, No. 37 Guoxue Alley, Chengdu, 610041, China
- Precision Medicine Key Laboratory of Sichuan Province, Functional and Molecular Imaging Key Laboratory of Sichuan Province, and Research Unit of Psychoradiology, Chinese Academy of Medical Sciences, Chengdu, 610041, China
- Department of Radiology, West China Xiamen Hospital of Sichuan University, Xiamen, Fujian, 361000, China
| | - Zhongwei Gu
- Department of Radiology, Department of Respiratory, Huaxi MR Research Center (HMRRC) and Critical Care Medicine, Institute of Respiratory Health, Precision Medicine Center, Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, No. 37 Guoxue Alley, Chengdu, 610041, China
| | - Kui Luo
- Department of Radiology, Department of Respiratory, Huaxi MR Research Center (HMRRC) and Critical Care Medicine, Institute of Respiratory Health, Precision Medicine Center, Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, No. 37 Guoxue Alley, Chengdu, 610041, China
- Precision Medicine Key Laboratory of Sichuan Province, Functional and Molecular Imaging Key Laboratory of Sichuan Province, and Research Unit of Psychoradiology, Chinese Academy of Medical Sciences, Chengdu, 610041, China
| | - Weimin Li
- Department of Radiology, Department of Respiratory, Huaxi MR Research Center (HMRRC) and Critical Care Medicine, Institute of Respiratory Health, Precision Medicine Center, Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, No. 37 Guoxue Alley, Chengdu, 610041, China
- Precision Medicine Key Laboratory of Sichuan Province, Functional and Molecular Imaging Key Laboratory of Sichuan Province, and Research Unit of Psychoradiology, Chinese Academy of Medical Sciences, Chengdu, 610041, China
| |
Collapse
|
7
|
Li X, Liang Q, Zhou L, Deng G, Xiao Y, Gan Y, Han S, Liao J, Wang R, Qing X, Li W. Survivin degradation by bergenin overcomes pemetrexed resistance. Cell Oncol (Dordr) 2023; 46:1837-1853. [PMID: 37542022 DOI: 10.1007/s13402-023-00850-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/24/2023] [Indexed: 08/06/2023] Open
Abstract
PURPOSE Chemoresistance is a primary factor for treatment failure and tumor recurrence in non-small cell lung cancer (NSCLC) patients. The oncoprotein survivin is commonly upregulated in human malignancies and is associated with poor prognosis, but its effect on carcinogenesis and chemoresistance in NSCLC is not yet evident, and to explore an effective inhibitor targeting survivin expression is urgently needed. METHODS The protumor characteristics of survivin and antitumor activities of bergenin in NSCLC cells were examined by MTS, colony formation assays, immunoblot, immunohistochemistry, and in vivo xenograft development. RESULTS Survivin was upregulated in non-small cell lung cancer (NSCLC) tissues, while its depletion inhibited NSCLC tumorigenesis. The current study focused on bergenin, identifying its effective antitumor effect on NSCLC cells both in vivo and in vitro. The results showed that bergenin could inhibit cell proliferation and induce the intrinsic pathway of apoptosis via downregulating survivin. Mechanistically, bergenin reduced the phosphorylation of survivin via inhibiting the Akt/Wee1/CDK1 signaling pathway, thus resulting in enhanced interaction between survivin and E3 ligase Fbxl7 to promote survivin ubiquitination and degradation. Furthermore, bergenin promoted chemoresistance in NSCLC cells re-sensitized to pemetrexed treatment. CONCLUSIONS Survivin overexpression is required for maintaining multiple malignant phenotypes of NSCLC cells. Bergenin exerts a potent antitumor effect on NSCLC via targeting survivin, rendering it a promising agent for the treatment of NSCLC.
Collapse
Affiliation(s)
- Xiaoying Li
- Department of Radiology, The Third Xiangya Hospital of Central South University, Changsha, Hunan, 410013, People's Republic of China
- Cell Transplantation and Gene Therapy Institute, The Third Xiangya Hospital, Central South University, Changsha, Hunan, 410013, People's Republic of China
| | - Qi Liang
- Department of Radiology, The Third Xiangya Hospital of Central South University, Changsha, Hunan, 410013, People's Republic of China
- Cell Transplantation and Gene Therapy Institute, The Third Xiangya Hospital, Central South University, Changsha, Hunan, 410013, People's Republic of China
| | - Li Zhou
- Department of Pathology, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital of Central South University, Changsha, Hunan, 410008, People's Republic of China
| | - Gaoyan Deng
- Department of Thoracic Surgery, Hunan Chest Hospital, Changsha, Hunan, 410006, People's Republic of China
| | - Yeqing Xiao
- Department of Ultrasonography, Hunan Chest Hospital, Changsha, Hunan, 410006, People's Republic of China
| | - Yu Gan
- Department of Radiology, The Third Xiangya Hospital of Central South University, Changsha, Hunan, 410013, People's Republic of China
- Cell Transplantation and Gene Therapy Institute, The Third Xiangya Hospital, Central South University, Changsha, Hunan, 410013, People's Republic of China
| | - Shuangze Han
- Department of Radiology, The Third Xiangya Hospital of Central South University, Changsha, Hunan, 410013, People's Republic of China
- Department of Ultrasound, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430022, People's Republic of China
| | - Jinzhuang Liao
- Department of Radiology, The Third Xiangya Hospital of Central South University, Changsha, Hunan, 410013, People's Republic of China
- Cell Transplantation and Gene Therapy Institute, The Third Xiangya Hospital, Central South University, Changsha, Hunan, 410013, People's Republic of China
| | - Ruirui Wang
- Department of Radiology, The Third Xiangya Hospital of Central South University, Changsha, Hunan, 410013, People's Republic of China
- Cell Transplantation and Gene Therapy Institute, The Third Xiangya Hospital, Central South University, Changsha, Hunan, 410013, People's Republic of China
| | - Xiang Qing
- Department of Otolaryngology Head and Neck Surgery, The Third Xiangya Hospital, Central South University, Changsha, Hunan, 410013, People's Republic of China.
| | - Wei Li
- Department of Radiology, The Third Xiangya Hospital of Central South University, Changsha, Hunan, 410013, People's Republic of China.
- Cell Transplantation and Gene Therapy Institute, The Third Xiangya Hospital, Central South University, Changsha, Hunan, 410013, People's Republic of China.
| |
Collapse
|
8
|
Li J, Zheng K, Shen H, Wu H, Wan C, Zhang R, Liu Z. Calpain-2 protein influences chikungunya virus replication and regulates vimentin rearrangement caused by chikungunya virus infection. Front Microbiol 2023; 14:1229576. [PMID: 37928675 PMCID: PMC10620729 DOI: 10.3389/fmicb.2023.1229576] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2023] [Accepted: 10/06/2023] [Indexed: 11/07/2023] Open
Abstract
Chikungunya fever (CHIF), a vector-borne disease transmitted mainly by Aedes albopictus and Aedes aegypti, is caused by Chikungunya virus (CHIKV) infection. To date, it is estimated that 39% of the world's population is at risk of infection for living in countries and regions where CHIKV is endemic. However, at present, the cellular receptors of CHIKV remains not clear, and there are no specific drugs and vaccines for CHIF. Here, the cytotoxicity of calpain-2 protein activity inhibitor III and specific siRNA was detected by MTT assays. The replication of CHIKV was detected by qPCR amplification and plaque assay. Western blot was used to determine the level of the calpain-2 protein and vimentin protein. Immunofluorescence was also operated for detecting the rearrangement of vimentin protein. Our results indicated that calpain-2 protein activity inhibitor III and specific siRNA might suppress CHIKV replication. Furthermore, CHIKV infection led to vimentin remodeling and formation of cage-like structures, which could be inhibited by the inhibitor III. In summary, we confirmed that calpain-2 protein influenced chikungunya virus replication and regulated vimentin rearrangement caused by chikungunya virus infection, which could be important for understanding the biological significance of CHIKV replication and the future development of antiviral strategies.
Collapse
Affiliation(s)
- Jia Li
- Department of Clinical Laboratory, Affiliated Hengyang Hospital of Southern Medical University, Hengyang Central Hospital, Hengyang, China
- Institute of Pathogenic Organisms, Shenzhen Center for Disease Control and Prevention, Shenzhen, China
- Biosafety Level 3 Laboratory, School of Public Health, Southern Medical University, Guangzhou, China
| | - Kang Zheng
- Department of Clinical Laboratory, Affiliated Hengyang Hospital of Southern Medical University, Hengyang Central Hospital, Hengyang, China
| | - Huilong Shen
- Department of Clinical Laboratory, Affiliated Hengyang Hospital of Southern Medical University, Hengyang Central Hospital, Hengyang, China
| | - Hua Wu
- Department of Clinical Laboratory, Affiliated Hengyang Hospital of Southern Medical University, Hengyang Central Hospital, Hengyang, China
| | - Chengsong Wan
- Institute of Pathogenic Organisms, Shenzhen Center for Disease Control and Prevention, Shenzhen, China
| | - Renli Zhang
- Biosafety Level 3 Laboratory, School of Public Health, Southern Medical University, Guangzhou, China
| | - Zhimin Liu
- Department of Clinical Laboratory, Affiliated Hengyang Hospital of Southern Medical University, Hengyang Central Hospital, Hengyang, China
| |
Collapse
|
9
|
Hua T, Robitaille M, Roberts-Thomson SJ, Monteith GR. The intersection between cysteine proteases, Ca 2+ signalling and cancer cell apoptosis. BIOCHIMICA ET BIOPHYSICA ACTA. MOLECULAR CELL RESEARCH 2023; 1870:119532. [PMID: 37393017 DOI: 10.1016/j.bbamcr.2023.119532] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Revised: 06/19/2023] [Accepted: 06/26/2023] [Indexed: 07/03/2023]
Abstract
Apoptosis is a highly complex and regulated cell death pathway that safeguards the physiological balance between life and death. Over the past decade, the role of Ca2+ signalling in apoptosis and the mechanisms involved have become clearer. The initiation and execution of apoptosis is coordinated by three distinct groups of cysteines proteases: the caspase, calpain and cathepsin families. Beyond its physiological importance, the ability to evade apoptosis is a prominent hallmark of cancer cells. In this review, we will explore the involvement of Ca2+ in the regulation of caspase, calpain and cathepsin activity, and how the actions of these cysteine proteases alter intracellular Ca2+ handling during apoptosis. We will also explore how apoptosis resistance can be achieved in cancer cells through deregulation of cysteine proteases and remodelling of the Ca2+ signalling toolkit.
Collapse
Affiliation(s)
- Trinh Hua
- School of Pharmacy, The University of Queensland, Brisbane, QLD 4102, Australia.
| | - Mélanie Robitaille
- School of Pharmacy, The University of Queensland, Brisbane, QLD 4102, Australia.
| | | | - Gregory R Monteith
- School of Pharmacy, The University of Queensland, Brisbane, QLD 4102, Australia; Mater Research Institute, Translational Research Institute, The University of Queensland, Brisbane, QLD, Australia.
| |
Collapse
|
10
|
Batts TL, Sasaki E, Miller M, Sparago J, Bauer RW, Paulsen D, Boudreaux B, Liu CC, Byrum SD, Johnston AN. Neoplastic signatures: Comparative proteomics of canine hepatobiliary neuroendocrine tumors to normal niche tissue. PLoS One 2023; 18:e0280928. [PMID: 36696389 PMCID: PMC9876354 DOI: 10.1371/journal.pone.0280928] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2022] [Accepted: 01/11/2023] [Indexed: 01/26/2023] Open
Abstract
Hepatobiliary neuroendocrine neoplasms are rare cancers in humans and dogs. To date, no large-scale primary hepatobiliary neoplasm omics analyses exist in any species. This limits the development of diagnostic biomarkers and targeted therapeutics. Neuroendocrine cancers are a heterogenous group of neoplasms categorized by their tissue-of-origin. Because the anatomic niche of neuroendocrine neoplasms shapes tumor phenotype, we sought to compare the proteomes of 3 canine hepatobiliary neoplasms to normal hepatobiliary tissue and adrenal glands with the objective of identifying unique protein signatures. Protein was extracted from formalin-fixed paraffin-embedded samples and submitted for tandem mass spectroscopy. Thirty-two upregulated and 126 downregulated differentially expressed proteins were identified. Remarkably, 6 (19%) of the upregulated proteins are correlated to non-hepatobiliary neuroendocrine neoplasia and 16 (50%) are functionally annotated within the exosome cellular compartment key to neuroendocrine signaling. Twenty-six (21%) downregulated proteins are enriched in metabolic pathways consistent with alterations in cancer. These results suggests that characteristic neoplastic protein signatures can be gleaned from small data sets using a comparative proteomics approach.
Collapse
Affiliation(s)
- Tifini L. Batts
- Veterinary Clinical Sciences Department, Louisiana State University School of Veterinary Medicine, Baton Rouge, Louisiana, United States of America
| | - Emi Sasaki
- Department of Pathobiological Sciences and Louisiana Animal Disease Diagnostic Laboratory, Baton Rouge, Louisiana, United States of America
| | - Mayzie Miller
- Veterinary Clinical Sciences Department, Louisiana State University School of Veterinary Medicine, Baton Rouge, Louisiana, United States of America
| | - Joshua Sparago
- Veterinary Clinical Sciences Department, Louisiana State University School of Veterinary Medicine, Baton Rouge, Louisiana, United States of America
| | - Rudy W. Bauer
- Department of Pathobiological Sciences and Louisiana Animal Disease Diagnostic Laboratory, Baton Rouge, Louisiana, United States of America
| | - Daniel Paulsen
- Department of Pathobiological Sciences and Louisiana Animal Disease Diagnostic Laboratory, Baton Rouge, Louisiana, United States of America
| | - Bonnie Boudreaux
- Veterinary Clinical Sciences Department, Louisiana State University School of Veterinary Medicine, Baton Rouge, Louisiana, United States of America
| | - Chin-Chi Liu
- Veterinary Clinical Sciences Department, Louisiana State University School of Veterinary Medicine, Baton Rouge, Louisiana, United States of America
| | - Stephanie D. Byrum
- Department of Biochemistry and Molecular Biology, University of Arkansas for Medical Sciences, Little Rock, AR, United States of America
- Arkansas Children’s Research Institute, Little Rock, AR, United States of America
| | - Andrea N. Johnston
- Veterinary Clinical Sciences Department, Louisiana State University School of Veterinary Medicine, Baton Rouge, Louisiana, United States of America
- * E-mail:
| |
Collapse
|
11
|
Ezhilarasan D, Lakshmi T, Mallineni SK. Nano-based targeted drug delivery for lung cancer: therapeutic avenues and challenges. Nanomedicine (Lond) 2022; 17:1855-1869. [PMID: 35311343 DOI: 10.2217/nnm-2021-0364] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Accepted: 03/04/2022] [Indexed: 12/24/2022] Open
Abstract
Most anticancer drugs often fail in clinical trials due to poor solubility, poor bioavailability, lack of targeted delivery and several off-target effects. Polymeric nanoparticles such as poly(lactide), poly(lactic-co-glycolic acid), ALB-loading paclitaxel (Abraxane® ABI-007), lomustine-loaded chitosan, gelatin (decorated with EGF receptor-targeted biotinylated EGF) and so on offer controlled and sustained drug-release properties, biocompatibility and promising anticancer effects. EGF, folic acid, transferrin, sigma and urokinase plasminogen activator receptors-targeting nano preparations improve bioavailability and accumulate drugs on the lung tumor cell surface. However, route of administration, size, pharmacokinetic properties, immune clearance and so on hamper nanomedicines' clinical uses. This review focuses on the benefits, avenues and challenges of nanoparticle-based drug-delivery systems for lung cancer treatment.
Collapse
Affiliation(s)
- Devaraj Ezhilarasan
- Department of Pharmacology, Gold Lab, Saveetha Dental College, Saveetha Institute of Medical & Technical Sciences (SIMATS), Chennai, Tamil Nadu, 600077, India
| | - Thangavelu Lakshmi
- Department of Pharmacology, Gold Lab, Saveetha Dental College, Saveetha Institute of Medical & Technical Sciences (SIMATS), Chennai, Tamil Nadu, 600077, India
| | - Sreekanth Kumar Mallineni
- Department of Preventive Dental Sciences, College of Dentistry, Majmaah University, Almajmaah, 11952, Saudi Arabia
| |
Collapse
|
12
|
Luo L, Yao X, Xiang J. Pyroptosis-Related Gene Model Predicts Prognosis and Immune Microenvironment for Non-Small-Cell Lung Cancer. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:1749111. [PMID: 36092153 PMCID: PMC9453043 DOI: 10.1155/2022/1749111] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/02/2022] [Revised: 07/12/2022] [Accepted: 08/09/2022] [Indexed: 12/13/2022]
Abstract
Non-small-cell lung cancer (NSCLC) has a high incidence and mortality worldwide. Moreover, it needs more accurate means for predicting prognosis and treatments. Pyroptosis is a novel form of cell death about inflammation which was highly related to the occurrence and development of tumors. Despite having some studies about pyroptosis-related genes (PRGs) and cancer, the correlation has not been explored enough between PRGs and immune in NSCLC. In this study, we constructed a PRG model by WGCNA to access the prognosis value PRGs have. The testing cohort (n = 464) with four datasets from the GEO database conducted a survival analysis to confirm the stability of the prognostic model. The risk score and age are examined as independent prognostic factors. Based on the PRGs, we found multiple pathways enriched in immune in NSCLC. Separating samples into three subtypes by consensus cluster analysis, Cluster 3 was identified as immune-inflamed phenotype with an optimistic prognostic outcome. A three-gene PRG signature (BNIP3, CASP9, and CAPN1) was identified, and BNIP3 was identified as the core gene. Knockdown of BNIP3 significantly inhibited the growth of H358 cells and induced pyroptosis. In conclusion, the model construction based on PRGs provides novel insights into the prediction of NSCLC prognosis, and BNIP3 can serve as a diagnostic biomarker for NSCLC.
Collapse
Affiliation(s)
- Lianxiang Luo
- The Marine Biomedical Research Institute, Guangdong Medical University, Zhanjiang, Guangdong, China 524023
- The Marine Biomedical Research Institute of Guangdong Zhanjiang, Zhanjiang, Guangdong, China 524023
| | - Xinyue Yao
- The First Clinical College, Guangdong Medical University, Zhanjiang, Guangdong, China 524023
| | - Jing Xiang
- The Marine Biomedical Research Institute, Guangdong Medical University, Zhanjiang, Guangdong, China 524023
| |
Collapse
|
13
|
Ma XL, Zhu KY, Chen YD, Tang WG, Xie SH, Zheng H, Tong Y, Wang YC, Ren N, Guo L, Lu RQ. Identification of a novel Calpain-2-SRC feed-back loop as necessity for β-Catenin accumulation and signaling activation in hepatocellular carcinoma. Oncogene 2022; 41:3554-3569. [PMID: 35697802 DOI: 10.1038/s41388-022-02367-x] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Revised: 05/17/2022] [Accepted: 05/30/2022] [Indexed: 11/09/2022]
Abstract
Rapid progression is the major cause of the poor prognosis of hepatocellular carcinoma (HCC); however, the underlying mechanism remained unclear. Here, we found Calpain-2 (CAPN2), a well-established protease that accelerates tumor progression in several malignancies, is overexpressed in HCC and acts as an independent predictor for poor outcomes. Furthermore, CAPN2 promoted the proliferation and invasion of HCC, and showed a positive correlation with the levels of invasion-related markers. Mechanistically, a novel CAPN2-SRC positive regulatory loop was identified upstream of β-catenin to prevent its ubiquitination and degradation, and subsequently promoted HCC progression: CAPN2 could proteolyze PTP1B to form a truncation of approximately 42 kDa with increased phosphatase activity, resulting in reduced SRC Y530 phosphorylation and increased SRC kinase activity; meanwhile, CAPN2 itself was a bone fide substrate of SRC that was primarily phosphorylated at Y625 by SRC and exhibited increased proteolysis activity upon phosphorylation. Interestingly, the CAPN2-SRC loop could not only restrain most of cytoplasmic β-catenin degradation by inhibiting GSK3β pathway, but also prevented TRIM33-induced nuclear β-catenin degradation even in β-catenin-mutant cells. Present study identified a CAPN2-SRC positive loop responsible for intracellular β-catenin accumulation and signaling activation, and targeting CAPN2 protease activity might be a promising approach for preventing HCC progression.
Collapse
Affiliation(s)
- Xiao-Lu Ma
- Department of clinical laboratory, Shanghai Cancer center, Fudan University, Shanghai, 200032, China.,Department of Oncology, Shanghai Medical School, Fudan University, Shanghai, 200032, China
| | - Ke-Yu Zhu
- Department of clinical laboratory, Shanghai Cancer center, Fudan University, Shanghai, 200032, China.,Department of Oncology, Shanghai Medical School, Fudan University, Shanghai, 200032, China
| | - Yue-Da Chen
- Department of general surgery, Xiamen Branch, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Wei-Guo Tang
- Liver cancer institute, Zhongshan hospital, Fudan university, Shanghai, 200032, China.,Department of Hepatobiliary and Pancreatic Surgery, Minhang Hospital, Fudan University, Shanghai, 201100, China
| | - Su-Hong Xie
- Department of clinical laboratory, Shanghai Cancer center, Fudan University, Shanghai, 200032, China.,Department of Oncology, Shanghai Medical School, Fudan University, Shanghai, 200032, China
| | - Hui Zheng
- Department of clinical laboratory, Shanghai Cancer center, Fudan University, Shanghai, 200032, China.,Department of Oncology, Shanghai Medical School, Fudan University, Shanghai, 200032, China
| | - Ying Tong
- Department of clinical laboratory, Shanghai Cancer center, Fudan University, Shanghai, 200032, China.,Department of Oncology, Shanghai Medical School, Fudan University, Shanghai, 200032, China
| | - Yan-Chun Wang
- Department of clinical laboratory, Shanghai Cancer center, Fudan University, Shanghai, 200032, China.,Department of Oncology, Shanghai Medical School, Fudan University, Shanghai, 200032, China
| | - Ning Ren
- Liver cancer institute, Zhongshan hospital, Fudan university, Shanghai, 200032, China. .,Department of Hepatobiliary and Pancreatic Surgery, Minhang Hospital, Fudan University, Shanghai, 201100, China.
| | - Lin Guo
- Department of clinical laboratory, Shanghai Cancer center, Fudan University, Shanghai, 200032, China. .,Department of Oncology, Shanghai Medical School, Fudan University, Shanghai, 200032, China.
| | - Ren-Quan Lu
- Department of clinical laboratory, Shanghai Cancer center, Fudan University, Shanghai, 200032, China. .,Department of Oncology, Shanghai Medical School, Fudan University, Shanghai, 200032, China.
| |
Collapse
|
14
|
Peng X, Yang R, Song J, Wang X, Dong W. Calpain2 Upregulation Regulates EMT-Mediated Pancreatic Cancer Metastasis via the Wnt/β-Catenin Signaling Pathway. Front Med (Lausanne) 2022; 9:783592. [PMID: 35707527 PMCID: PMC9189366 DOI: 10.3389/fmed.2022.783592] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2021] [Accepted: 04/25/2022] [Indexed: 12/24/2022] Open
Abstract
Calpains2 (CAPN2) is a calcium-dependent, non-lysosomal cysteine protease that plays critical roles in normal cellular functions and pathological processes, including tumorigenesis, cancer progression, and metastasis. However, the role and underlying regulatory mechanisms of CAPN2 in pancreatic cancer (PC) are still unknown. We found that CAPN2 is highly expressed in PC tissues and associated with poor PC prognosis by using The Cancer Genome Atlas (TCGA) datasets, Gene Expression Omnibus (GEO) datasets, and PC tissue arrays. CAPN2 downregulation significantly inhibited cell proliferation, migration, and invasion and regulated Wnt/β-catenin signaling pathway-mediated epithelial-mesenchymal transition (EMT) in PC cells. Our findings highlight the significance of CAPN2 in tumor regression and, thus, indicate that CAPN2 could be a promising target for PC treatment.
Collapse
Affiliation(s)
- Xiulan Peng
- Department of Oncology, The Second Affiliated Hospital of Jianghan University, Wuhan, China
- *Correspondence: Xiulan Peng
| | - Rui Yang
- Department of Vascular Surgery, The Second Affiliated Hospital of Jianghan University, Wuhan, China
| | - Jia Song
- Departments of Institute, The Third Affiliated Teaching Hospital of Xinjiang Medical University, Affiliated Cancer Hospital, Ürümqi, China
| | - Xia Wang
- Department of Pharmacy, The Second Affiliated Hospital of Jianghan University, Wuhan, China
| | - Weiguo Dong
- Department of Gastroenterology, Renmin Hospital of Wuhan University, Wuhan, China
- Weiguo Dong
| |
Collapse
|
15
|
Peng J, Shi S, Yu J, Liu J, Wei H, Song H, Wang S, Li Z, He S, Li L, Zhang H, Yan Z, Zhao R, Liu Y, Liu Y, Li J, Zhang R, Wang W. miR-378d suppresses malignant phenotype of ESCC cells through AKT signaling. Cancer Cell Int 2021; 21:702. [PMID: 34937563 PMCID: PMC8697470 DOI: 10.1186/s12935-021-02403-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2021] [Accepted: 12/08/2021] [Indexed: 01/12/2023] Open
Abstract
Background Post-resistance progress in paclitaxel (PTX) treatment remains a major challenge in tumor treatment. A high dose of PTX was used for cell lines to analyze the changes in molecular expression. The miR-378d was sharply reduced in surviving cells, but the role of miR-378d in Esophageal squamous cell carcinoma (ESCC) remained unclear. Methods We analyzed the relationship between miR-378d expression and the clinicopathological features of ESCC. We constructed miR-378d silent expression cell lines to study phenotypes and molecular mechanisms. Results The miR-378d expression was associated with good prognosis in patients with ESCC. miR-378d inhibition promoted chemo-resistance, monoclonal formation, EMT, migration, invasion, stemness, and metastasis of ESCC cells. miR-378d can target downregulated AKT1. Conclusions Therefore, miR-378d expression is a good prognostic factor of patients with ESCC and regulates the malignant phenotype of tumor cells through AKT. Supplementary Information The online version contains supplementary material available at 10.1186/s12935-021-02403-y.
Collapse
|
16
|
Genetic variants in histone modification regions are associated with the prognosis of lung adenocarcinoma. Sci Rep 2021; 11:21520. [PMID: 34728688 PMCID: PMC8563968 DOI: 10.1038/s41598-021-00909-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2021] [Accepted: 09/20/2021] [Indexed: 11/25/2022] Open
Abstract
We investigated the association between genetic variants in the histone modification regions and the prognosis of lung adenocarcinoma after curative surgery. Potentially functional SNPs were selected using integrated analysis of ChIP-seq and RNA-seq. The SNPs were analyzed in a discovery set (n = 166) and a validation set (n = 238). The associations of the SNPs with overall survival (OS) and disease-free survival (DFS) were analyzed. A total of 279 SNPs were selected for genotyping. Among these, CAPN1 rs17583C>T was significantly associated with better OS and DFS (P = 0.001 and P = 0.007, respectively), and LINC00959 rs4751162A>G was significantly associated with worse DFS (P = 0.008). Luciferase assays showed a significantly lower promoter activity of CAPN1 in the rs17583 T allele than C allele (P = 0.008), and consistently the CT + TT genotypes had significantly lower CAPN1 expression than CC genotype (P = 0.01) in clinical samples. The rs4751162 G allele had higher promoter activity of GLRX3 than A allele (P = 0.05). The motif analyses and ChIP-qPCR confirmed that the variants are located in the active promoter/enhancer regions where transcription factor binding occurs. This study showed that genetic variants in the histone modification regions could predict the prognosis of lung adenocarcinoma after surgery.
Collapse
|
17
|
Yang J, Yao Y, Tong L, Zhu Z, Wang L, Yang J. CD47 is highly expressed in gliomas and targeting CD47 is a promising therapeutic strategy. EUR J INFLAMM 2021. [DOI: 10.1177/20587392211000899] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Gliomas are very malignant brain tumors that are difficult to treat. CD47 is an antiphagocytic molecule that binds to SIPRα on phagocytes. It is overexpressed on the plasma membranes of multiple human tumor cell types and is an important diagnostic and prognostic biomarker in many types of cancer. However, the association between CD47 protein expression in glioma tissue and clinicopathological stage has not been investigated in detail. A total of 80 surgical glioma specimens were stained with anti-CD47 antibody to assess the relationship between CD47 protein expression and clinicopathological stage of the glioma. Wound healing assays were performed to analyze the influence of CD47 on the migration and invasion of glioma cells, and near-infrared fluorescence localization assays in a U-87 MG-bearing xenograft model were used to determine the distribution of anti-CD47 antibody in vivo. MTT assays and administration of anti-CD47 to a U251-bearing xenograft model were used to analyze the inhibitory effects of the antibody on gliomas. CD47 expression was higher in high-grade gliomas than in low-grade gliomas, and high CD47 expression was positively correlated with histology and tumor clinicopathological stage. CD47 over-expression promoted the growth and motility of two glioma cell lines (U-87 MG and U251) and a laboratory-developed anti-CD47 antibody accumulated at the glioma site. Proliferation of U251 and U-87 MG cells was not significantly inhibited by the anti-CD47 antibody in vitro, but the antibody significantly inhibited U251 growth in vivo. It also enhanced inhibition capacity by Taxol. Our results suggest that CD47 plays a critical role in the progression of gliomas from stage I to IV and may be a potential target for the treatment of gliomas. CD47 appears to play a critical role in the progression of gliomas from stage I to IV and an anti-CD47 antibody prepared in the laboratory may inhibit the growth of gliomas.
Collapse
Affiliation(s)
- Jiaying Yang
- Department of Neurosurgery, Lanling People’s Hospital, Shandong, P.R. China
| | - Yongjun Yao
- Department of Neurosurgery, Lanling People’s Hospital, Shandong, P.R. China
- Department of Pathology, Lanling People’s Hospital, Shandong, P.R. China
| | - Li Tong
- Department of Biochemistry and Molecular Biology, Beijing Normal University, Gene Engineering and Biotechnology Beijing Key Laboratory, Beijing, P.R. China
| | - Ziwei Zhu
- Department of Biochemistry and Molecular Biology, Beijing Normal University, Gene Engineering and Biotechnology Beijing Key Laboratory, Beijing, P.R. China
| | - Lei Wang
- Department of Neurosurgery, Lanling People’s Hospital, Shandong, P.R. China
| | - Jinju Yang
- Department of Biochemistry and Molecular Biology, Beijing Normal University, Gene Engineering and Biotechnology Beijing Key Laboratory, Beijing, P.R. China
- National Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing, China
| |
Collapse
|
18
|
Mechanisms of resistance to chemotherapy in non-small cell lung cancer. Arch Pharm Res 2021; 44:146-164. [PMID: 33608812 DOI: 10.1007/s12272-021-01312-y] [Citation(s) in RCA: 84] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2020] [Accepted: 01/16/2021] [Indexed: 02/07/2023]
Abstract
Non-small cell lung cancer (NSCLC), which represents 80-85% of lung cancer cases, is one of the leading causes of human death worldwide. The majority of patients undergo an intensive and invasive treatment regimen, which may include radiotherapy, chemotherapy, targeted therapy, immunotherapy, or a combination of these, depending on disease stage and performance status. Despite advances in therapeutic regimens, the 5-year survival of NSCLC is approximately 20-30%, largely due to diagnosis at advanced stages. Conventional chemotherapy is still the standard treatment option for patients with NSCLC, especially those with advanced disease. However, the emergence of resistance to chemotherapeutic agents (chemoresistance) poses a significant obstacle to the management of patients with NSCLC. Therefore, to develop efficacious chemotherapeutic approaches for NSCLC, it is necessary to understand the mechanisms underlying chemoresistance. Several mechanisms are known to mediate chemoresistance. These include altered cellular targets for chemotherapy, decreased cellular drug concentrations, blockade of chemotherapy-induced cell cycle arrest and apoptosis, acquisition of epithelial-mesenchymal transition and cancer stem cell-like phenotypes, deregulated expression of microRNAs, epigenetic modulation, and the interaction with tumor microenvironments. In this review, we summarize the mechanisms underlying chemoresistance and tumor recurrence in NSCLC and discuss potential strategies to avoid or overcome chemoresistance.
Collapse
|
19
|
Nian H, Ma B. Calpain-calpastatin system and cancer progression. Biol Rev Camb Philos Soc 2021; 96:961-975. [PMID: 33470511 DOI: 10.1111/brv.12686] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2020] [Revised: 12/26/2020] [Accepted: 01/06/2021] [Indexed: 12/19/2022]
Abstract
The calpain system is required by many important physiological processes, including the cell cycle, cytoskeleton remodelling, cellular proliferation, migration, cancer cell invasion, metastasis, survival, autophagy, apoptosis and signalling, as well as the pathogenesis of a wide range of disorders, in which it may function to promote tumorigenesis. Calpains are intracellular conserved calcium-activated neutral cysteine proteinases that are involved in mediating cancer progression via catalysing and regulating the proteolysis of their specific substrates, which are important signalling molecules during cancer progression. μ-calpain, m-calpain, and their specific inhibitor calpastatin are the three molecules originally identified as comprising the calpain system and they contain several crucial domains, specific motifs, and functional sites. A large amount of data supports the roles of the calpain-calpastatin system in cancer progression via regulation of cellular adhesion, proliferation, invasion, metastasis, and cellular survival and death, as well as inflammation and angiogenesis during tumorigenesis, implying that the inhibition of calpain activity may be a potential anti-cancer intervention strategy targeting cancer cell survival, invasion and chemotherapy resistance.
Collapse
Affiliation(s)
- Hong Nian
- Tianjin Key Laboratory of Retinal Functions and Diseases, Tianjin International Joint Research and Development Centre of Ophthalmology and Vision Science, Eye Institute and School of Optometry, Tianjin Medical University Eye Hospital, Tianjin, 300384, China
| | - Binyun Ma
- Department of Medicine/Hematology, Keck School of Medicine of the University of Southern California, Los Angeles, CA, 90033, U.S.A
| |
Collapse
|
20
|
Nuclear Localization of Heme Oxygenase-1 in Pathophysiological Conditions: Does It Explain the Dual Role in Cancer? Antioxidants (Basel) 2021; 10:antiox10010087. [PMID: 33440611 PMCID: PMC7826503 DOI: 10.3390/antiox10010087] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2020] [Revised: 12/11/2020] [Accepted: 12/12/2020] [Indexed: 12/12/2022] Open
Abstract
Heme Oxygenase-1 (HO-1) is a type II detoxifying enzyme that catalyzes the rate-limiting step in heme degradation leading to the formation of equimolar quantities of carbon monoxide (CO), free iron and biliverdin. HO-1 was originally shown to localize at the smooth endoplasmic reticulum membrane (sER), although increasing evidence demonstrates that the protein translocates to other subcellular compartments including the nucleus. The nuclear translocation occurs after proteolytic cleavage by proteases including signal peptide peptidase and some cysteine proteases. In addition, nuclear translocation has been demonstrated to be involved in several cellular processes leading to cancer progression, including induction of resistance to therapy and enhanced metastatic activity. In this review, we focus on nuclear HO-1 implication in pathophysiological conditions with special emphasis on malignant processes. We provide a brief background on the current understanding of the mechanisms underlying how HO-1 leaves the sER membrane and migrates to the nucleus, the circumstances under which it does so and, maybe the most important and unknown aspect, what the function of HO-1 in the nucleus is.
Collapse
|
21
|
Park YI, Kwon SH, Lee G, Motoyama K, Kim MW, Lin M, Niidome T, Choi JH, Lee R. pH-sensitive multi-drug liposomes targeting folate receptor β for efficient treatment of non-small cell lung cancer. J Control Release 2020; 330:1-14. [PMID: 33321157 DOI: 10.1016/j.jconrel.2020.12.011] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2020] [Revised: 11/16/2020] [Accepted: 12/09/2020] [Indexed: 02/07/2023]
Abstract
Non-small cell lung cancer (NSCLC) is the leading cause of lung cancer-related deaths worldwide. Tumor-associated macrophages (TAMs), which can be polarized into tumor-promoting M2 phenotype, overexpress folate receptor beta (FRβ) and are associated with poor prognosis in NSCLC. In addition, calpain-2 (CAPN2) is overexpressed in NSCLC and is involved in tumor growth. To improve the anticancer efficacy of drugs and reduce their side effects in the treatment of NSCLC, it is important to develop smart drug delivery systems with specific targeting ability and controlled release mechanisms. In this study, FRβ-targeted pH-sensitive liposomes were designed as carriers to ensure efficient drug delivery and acid-responsive release in NSCLC cells. Folate-mediated targeting of FRβ in M2 TAMs and NSCLC cells effectively inhibited tumor growth and the stimulus-responsive drug release reduced the toxic side effects of the drug. The combination of doxycycline (anti-CAPN2) and docetaxel (anticancer drug) showed a synergistic inhibitory effect on tumor growth by suppressing CAPN2 expression.
Collapse
Affiliation(s)
- Yong Il Park
- School of Chemical Engineering, Chonnam National University, Gwangju 61186, Republic of Korea
| | - Seung-Hae Kwon
- Korea Basic Science Institute, Seoul 02841, Republic of Korea
| | - Gibok Lee
- School of Chemical Engineering, Chonnam National University, Gwangju 61186, Republic of Korea
| | - Keiichi Motoyama
- Graduate School of Pharmaceutical Sciences, Kumamoto University, Kumamoto 862-0973, Japan
| | - Min Woo Kim
- International Research Organization for Advanced Science and Technology (IROAST), Kumamoto University, Kumamoto 860-8555, Japan
| | - Min Lin
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an 710049, China
| | - Takuro Niidome
- Faculty of Advanced Science and Technology, Kumamoto University, Kumamoto 860-8555, Japan
| | - Jung Hoon Choi
- Department of Anatomy & Institute of Veterinary Science, College of Veterinary Medicine, Kangwon National University, Chuncheon 24341, Republic of Korea.
| | - Ruda Lee
- International Research Organization for Advanced Science and Technology (IROAST), Kumamoto University, Kumamoto 860-8555, Japan.
| |
Collapse
|
22
|
Chen Y, Tang J, Lu T, Liu F. CAPN1 promotes malignant behavior and erlotinib resistance mediated by phosphorylation of c-Met and PIK3R2 via degrading PTPN1 in lung adenocarcinoma. Thorac Cancer 2020; 11:1848-1860. [PMID: 32395869 PMCID: PMC7327690 DOI: 10.1111/1759-7714.13465] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2020] [Revised: 04/14/2020] [Accepted: 04/17/2020] [Indexed: 01/04/2023] Open
Abstract
BACKGROUND Calpain 1 (CAPN1) has been found to be a promoter of cancer progression. PTPN1 as a physiological target molecule of CAPN1 plays a dephosphorylated role on multiple receptor tyrosine kinases. This study aimed to reveal the effects of CAPN1/PTPN1 on malignant phenotype and EGFR-TKI resistance of lung adenocarcinoma (LUAD) cells. METHODS A total of 84 primary LUAD tissues and paired paracancerous normal tissues were collected. Quantitative real-time PCR (qRT-PCR) and immunohistochemical (IHC) methods were used to measure the expression of CAPN1 and PTPN1 in tissues. qRT-PCR and western blot were used to detect the expressions of CAPN1, PTPN1, c-Met and PIK3R2 in cell lines. Cell counting kit-8 (CCK-8), colony formation and transwell assay were carried out to evaluate cell erlotinib resistance, proliferation, migration and invasion. Co-IP assay was used to verify the interaction between proteins. Cycloheximide (CHX) was applied to block protein synthesis. RESULTS CAPN1, c-Met and PIK3R2 were significantly upregulated and the correlation was positive in LUAD, while PTPN1 was decreased. EGFR-sensitive mutation was related to CAPN1/PTPN1. in vitro studies showed that PTPN1 can mediate dephosphorylation of c-Met and PIK3R2 by binding with both, thereby weakening cell proliferation, metastasis and erlotinib resistance, while CAPN1 could enhance the degradation of PTPN1 protein as a cancer promoter. CONCLUSIONS CAPN1 enhances the malignant behavior and erlotinib resistance of LUAD cells via degrading PTPN1 and then activating c-Met/PIK3R2, which suggests CAPN1/PTPN1 may serve as tumor markers or potential targets for diagnosis and treatment of LUAD. KEY POINTS Significant findings of the study Superior CAPN1 and inferior PTPN1 were related to activation of c-Met/PIK3R2 in lung adenocarcinoma. Moreover, regulations of CAPN1 and PTPN1 induced the changes of malignant behavior and erlotinib resistance. What this study adds Our findings confirmed that CAPN1/PTPN1 play crucial roles on proliferation, metastasis and erlotinib resistance of LUAD cells as c-Met/PIK3R2 regulators, and validated the regulatory mechanism of CAPN1 on PTPN1 in tumor model for the first time.
Collapse
Affiliation(s)
- Yichuan Chen
- Department of Cardiovascular Surgery, The Second Xiangya HospitalCentral South UniversityChangshaChina
| | - Jingqun Tang
- Department of Thoracic Surgery, The Second Xiangya HospitalCentral South UniversityChangshaChina
| | - Ting Lu
- Department of Cardiovascular Surgery, The Second Xiangya HospitalCentral South UniversityChangshaChina
| | - Fang Liu
- Clinic Nursing Teaching and Research Section, The Second Xiangya HospitalCentral South UniversityChangshaChina
| |
Collapse
|
23
|
Deng XZ, Geng SS, Luo M, Chai JJ, Xu Y, Chen CL, Qiu L, Ke Q, Duan QW, Song SM, Shen L, Luo ZG. Curcumin potentiates laryngeal squamous carcinoma radiosensitivity via NF-ΚB inhibition by suppressing IKKγ expression. J Recept Signal Transduct Res 2020; 40:541-549. [PMID: 32515250 DOI: 10.1080/10799893.2020.1767649] [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] [Indexed: 01/07/2023]
Abstract
Context: Curcumin has shown efficacy in promoting radiosensitivity combined with radiotherapy. However, the role and mechanism of curcumin on radiosensitivity in laryngeal squamous cell cancer (LSCC) is largely unknown.Objective: The aim of our study is to explore the role of IKKγ-NF-κB signaling in curcumin enhancing LSCC cell radiosensitivity in vitro.Materials and methods: Curcumin and X-ray were used to induce cell DNA damage and apoptosis, or inhibit cell clone formation. IKKγ siRNA and plasmid were used to change IKKγ expression. The CCK8 assay was used to detect cell viability. Clone formation ability was analyzed using a clonogenic assay, cell apoptosis was examined using flow cytometry, an immunofluorescence assay was used to detect DNA damage, while mRNA and protein levels were assayed using real time PCR and western blotting, respectively.Results: Curcumin significantly enhanced irradiation-induced DNA damage and apoptosis, while weakening clone-forming abilities of LSCC cell line Hep2 and Hep2-max. Compared to Hep2 cells, Hep2-max cells are more sensitive to curcumin post-irradiation. Curcumin suppressed irradiation-induced NF-κB activation by suppressing IKKγ expression, but not IKKα and IKKβ. Overexpression of IKKγ decreased irradiation-induced DNA damage and apoptosis, while promoting clone-forming abilities of Hep2 and Hep2-max cells. IKKγ overexpression further increased expression of NF-κB downstream genes, Bcl-XL, Bcl-2, and cyclin D1. Conversely, IKKγ silencing enhanced irradiation-induced DNA damage and apoptosis, but promoted clone formation in Hep2 and Hep2-max cells. Additionally, IKKγ silencing inhibited expression of Bcl-XL, Bcl-2, and cyclin D1.Conclusions: Curcumin enhances LSCC radiosensitivity via NF-ΚB inhibition by suppressing IKKγ expression.
Collapse
Affiliation(s)
- Xin-Zhou Deng
- Department of Clinical Oncology, Taihe Hospital, Hubei University of Medicine, Shiyan, Hubei, PR China.,HubeiKey Laboratory of Embryonic Stem Cell Research, Taihe Hospital, Hubei University of Medicine, Shiyan, Hubei, PR China
| | - Shan-Shan Geng
- Department of Clinical Oncology, Taihe Hospital, Hubei University of Medicine, Shiyan, Hubei, PR China.,Postgraduate Training Base, Shiyan Taihe Hospital, Jinzhou Medical University, Shiyan, Hubei, PR China
| | - Ming Luo
- Department of Clinical Oncology, Taihe Hospital, Hubei University of Medicine, Shiyan, Hubei, PR China
| | - Jing-Jing Chai
- Department of Clinical Oncology, Taihe Hospital, Hubei University of Medicine, Shiyan, Hubei, PR China
| | - Ying Xu
- Department of Clinical Oncology, Taihe Hospital, Hubei University of Medicine, Shiyan, Hubei, PR China
| | - Chun-Li Chen
- Department of Clinical Oncology, Taihe Hospital, Hubei University of Medicine, Shiyan, Hubei, PR China
| | - Li Qiu
- Department of Clinical Oncology, Taihe Hospital, Hubei University of Medicine, Shiyan, Hubei, PR China
| | - Qing Ke
- Department of Clinical Oncology, Taihe Hospital, Hubei University of Medicine, Shiyan, Hubei, PR China
| | - Qi-Wen Duan
- Department of Clinical Oncology, Taihe Hospital, Hubei University of Medicine, Shiyan, Hubei, PR China
| | - Shi-Mao Song
- Department of Clinical Oncology, Taihe Hospital, Hubei University of Medicine, Shiyan, Hubei, PR China
| | - Li Shen
- Department of Biochemistry, School of Basic Medical Sciences, Hubei University of Medicine, Shiyan, Hubei, PR China
| | - Zhi-Guo Luo
- Department of Clinical Oncology, Taihe Hospital, Hubei University of Medicine, Shiyan, Hubei, PR China
| |
Collapse
|
24
|
Li J, Song Y, Yu B, Yu Y. TNFAIP2 Promotes Non-Small Cell Lung Cancer Cells and Targeted by miR-145-5p. DNA Cell Biol 2020; 39:1256-1263. [PMID: 32456459 DOI: 10.1089/dna.2020.5415] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Tumor necrosis factor-alpha (TNFα) is an inflammatory cytokine that regulates inflammation and tumor progression in non-small cell lung cancer (NSCLC). The higher levels of TNF α are known to induce expression of several genes such as TNFα-induced protein 2 (TNFAIP2) with a largely unknown role in NSCLC. We provide the preliminary evidence for the role of TNFAIP2 in NSCLC progression and its epigenetic regulation mediated by microRNA, miR-145-5p. The expression of TNFAIP2 was confirmed using quantitative real-time PCR, immunohistochemistry, and Western blot in NSCLC tissue and paired adjacent normal tissue. All in vitro assays were undertaken in A549 and H23 cells and chemoresistance assays were undertaken in A549/Cisplatin (DDP) and H23/DDP cell types. TNFAIP2 silencing was undertaken using lipofectamine transfection of specific siRNA. Cells were co-transfected with miR-145-5p, and TNFAIP2-3' untranslated region (UTR) or TNFAIP2 with mutated 3'UTR using the luciferase vector pGL. Cell viability, transwell migration, and invasion were assessed. The role of caspase 3 proteins in cell viability was ascertained using Western blot. The tumor tissues (and cisplatin-resistant cell lines A549/DDP and H23/DDP) expressed significantly higher levels of TNAIP2 mRNA and protein. Silencing of TNFAIP2 resulted in reduced cell viability, reduced invasion, and migration in vitro. Silencing of TNFAIP2 in A549/DDP and H23/DDP had higher expression of TNFAIP2, reduced cell viability, and increased induction of caspase 3. MiR-145-5p binds to the 3'UTR of TNFAIP2. Overexpression of MiR-145-5p reduced expression of TNFAIP2, decreased cell viability, reduced cell migration and invasion, and significantly reduced expression of caspase 3 protein. TNFAIP2 mediates tumorigenesis in NSCLC through, not completely known pathways. miR-145-5p negatively regulates TNFAIP2 expression. miR-145-5p-mediated therapies may be explored in NSCLC.
Collapse
Affiliation(s)
- Jianing Li
- Department of Respiratory, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Yongfeng Song
- Department of Respiratory, The 10th Hospital of Harbin, Harbin, China
| | - Baiquan Yu
- Department of Respiratory, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Yao Yu
- Department of Respiratory, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| |
Collapse
|
25
|
Zhu Q, Luo R, Gu J, Hou Y, Chen Z, Xu F, Wang L, Mao W, Lu C, Ge D. High CXCR4 Expression Predicts a Poor Prognosis in Resected Lung Adenosquamous Carcinoma. J Cancer 2020; 11:810-818. [PMID: 31949484 PMCID: PMC6959020 DOI: 10.7150/jca.36498] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2019] [Accepted: 10/28/2019] [Indexed: 12/29/2022] Open
Abstract
Background: Primary adenosquamous carcinoma (ASC) is a rare malignant tumor in the lung and its biological behavior has not yet been thoroughly described. In this study, we aimed to explore the clinical and biological role of CXCR4 in patients with resected lung ASC. Methods: We retrospectively reviewed the clinical records of patients with histologically confirmed lung ASC who underwent surgical resection with systematic lymph node dissection. Immunohistochemical staining was performed to detect the expression of CXCR4 in tumor tissues. The correlation between CXCR4 expression and clinicopathological characteristics were evaluated. The association between CXCR4 expression and patients' prognosis was analyzed by Kaplan-Meier and Cox regression. Moreover, we performed in vitro studies including CCK8, transwell and cell apoptosis to explore the potential role of CXCR4 in lung ASC. Results: A total of 78 patients with resected lung ASC were reviewed. Seventy (89.7%) patient tumors expressed CXCR4, with high level of CXCR4 expression observed in 45 (57.7%) cases. In vitro, CXCR4 conferred no difference in proliferative capacity but increased invasive potential, enhanced chemoresistance and inhibited apoptosis of lung ASC. Clinically, high CXCR4 expression was significantly associated with solid ASC, lymph node metastasis and advanced TNM stage. Patients with high CXCR4 expression and solid ASC had decreased disease-free survival and overall survival.Conclusions: CXCR4 was commonly expressed in lung ASC tumors. High CXCR4 expression might be a novel marker in predicting a poor prognosis in resected lung ASC and might serve as a potential therapeutic target.
Collapse
Affiliation(s)
- Qiaoliang Zhu
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, Shanghai, P. R. China
| | - Rongkui Luo
- Department of Pathology, Zhongshan Hospital, Fudan University, Shanghai, P. R. China
| | - Jie Gu
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, Shanghai, P. R. China
| | - Yingyong Hou
- Department of Pathology, Zhongshan Hospital, Fudan University, Shanghai, P. R. China
| | - Zongwei Chen
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, Shanghai, P. R. China
| | - Fengkai Xu
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, Shanghai, P. R. China
| | - Lin Wang
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, Shanghai, P. R. China
| | - Wei Mao
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, Shanghai, P. R. China
| | - Chunlai Lu
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, Shanghai, P. R. China
| | - Di Ge
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, Shanghai, P. R. China.,Shanghai Respiratory Research Institute, Zhongshan Hospital, Fudan University, Shanghai, P.R. China
| |
Collapse
|
26
|
Li J, Gu Y, Zhang W, Bao CY, Li CR, Zhang JY, Liu T, Li S, Huang JX, Xie ZG, Hua SC, Wan Y. Molecular Mechanism for Selective Cytotoxicity towards Cancer Cells of Diselenide-Containing Paclitaxel Nanoparticles. Int J Biol Sci 2019; 15:1755-1770. [PMID: 31360117 PMCID: PMC6643224 DOI: 10.7150/ijbs.34878] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2019] [Accepted: 05/30/2019] [Indexed: 12/14/2022] Open
Abstract
Diselenide-containing paclitaxel nanoparticles (SePTX NPs) indicated selectivity of cytotoxicity between cancerous and normal cells in our previous work. Herein, the mechanism is revealed by molecular biology in detail. Cancer cells and normal cells were treated with the SePTX NPs and cell proliferation was measured using 3-(4, 5-dimethyl-2-thiazolyl)-2, 5-diphenyl-2-H-tetrazolium bromide (MTT) assay and cell morphology. Measurement of reactive oxygen species (ROS) levels and biochemical parameters were employed to monitor oxidative stress of the cells. JC-1 assay was used to detect the mitochondrial dysfunction of the cells. Terminal deoxynucleotidyl transferase-mediated dUTP nick end labeling (TUNEL) analysis was used to detect apoptosis of the cells. Immunofluorescence analysis and western blotting were employed to monitor changes in signaling pathway-related proteins. Compared with PTX, SePTX NPs has a good selectivity to cancer cells and can obviously induce the proliferation damage of cancer cells, but has no significant toxicity to normal cells, indicating that SePTX NPs has a specific killing effect on cancer cells. The results of mechanism research show that SePTX NPs can successfully inhibit the depolymerization of microtubules and induce cell cycle arrest, which is related to the upregulation of p53 and CyclinB1. Simultaneously, SePTX NPs can successfully induce oxidative stress, cause mitochondrial dysfunction, resulting in mitochondrial pathway-mediated apoptosis, which is related to the upregulation of autophagy-related protein LC3-II. On the other hand, lewis lung cancer C57BL/6 mice were used to evaluate the anti-tumor effects of SePTX NPs in vivo. Our data show that SePTX NPs exhibited high inhibiting efficiency against the growth of tumors and were able to reduce the side effects. Collectively, these data indicate that the high antitumor effect and selective cytotoxicities of SePTX NPs is promising in future cancer therapy.
Collapse
Affiliation(s)
- Jing Li
- Hubei Province Key Laboratory on Cardiovascular, Cerebrovascular, and Metabolic Disorders, Hubei University of Science and Technology, Xianning, Hubei 437100, P. R. China
| | - Yue Gu
- Department of Reparatory and Critical Care Medicine, the First Affiliated Hospital of Jilin University, Changchun 130021, P. R. China
| | - Wei Zhang
- State Key Laboratory of Polymer Physics and Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences5625 Renmin Street, Changchun, Jilin 130022, P. R. China
| | - Cui-Yu Bao
- Hubei Province Key Laboratory on Cardiovascular, Cerebrovascular, and Metabolic Disorders, Hubei University of Science and Technology, Xianning, Hubei 437100, P. R. China
| | - Cai-Rong Li
- Hubei Province Key Laboratory on Cardiovascular, Cerebrovascular, and Metabolic Disorders, Hubei University of Science and Technology, Xianning, Hubei 437100, P. R. China
| | - Jing-Yi Zhang
- Hubei Province Key Laboratory on Cardiovascular, Cerebrovascular, and Metabolic Disorders, Hubei University of Science and Technology, Xianning, Hubei 437100, P. R. China
| | - Tao Liu
- Hubei Province Key Laboratory on Cardiovascular, Cerebrovascular, and Metabolic Disorders, Hubei University of Science and Technology, Xianning, Hubei 437100, P. R. China
| | - Shuai Li
- Hubei Province Key Laboratory on Cardiovascular, Cerebrovascular, and Metabolic Disorders, Hubei University of Science and Technology, Xianning, Hubei 437100, P. R. China
| | - Jia-Xi Huang
- Hubei Province Key Laboratory on Cardiovascular, Cerebrovascular, and Metabolic Disorders, Hubei University of Science and Technology, Xianning, Hubei 437100, P. R. China
| | - Zhi-Gang Xie
- State Key Laboratory of Polymer Physics and Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences5625 Renmin Street, Changchun, Jilin 130022, P. R. China
| | - Shu-Cheng Hua
- Department of Reparatory and Critical Care Medicine, the First Affiliated Hospital of Jilin University, Changchun 130021, P. R. China
| | - Ying Wan
- College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, P. R. China
| |
Collapse
|