1
|
Wu Z, Wang Z, Hua Z, Ji Y, Ye Q, Zhang H, Yan W. Prognostic signature and immunotherapeutic relevance of Focal adhesion signaling pathway-related genes in osteosarcoma. Heliyon 2024; 10:e38523. [PMID: 39524888 PMCID: PMC11550747 DOI: 10.1016/j.heliyon.2024.e38523] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Revised: 09/22/2024] [Accepted: 09/25/2024] [Indexed: 11/16/2024] Open
Abstract
Background As the most common primary malignant bone tumor in children and adolescents, osteosarcoma currently lacks an effective clinical cure. Focal adhesion plays a crucial role in tumor invasion, migration, and drug resistance by mediating communication between the extracellular matrix and tumor cells. This study investigated the prognostic features and immunotherapeutic relevance of focal adhesion pathway-related genes in osteosarcoma to aid in the development of new therapeutic options. Methods We obtained mutational, transcriptomic, gene expression, and clinical data of osteosarcoma patients from the Gene Expression Omnibus (GEO) and Therapeutically Applicable Research to Generate Effective (TARGET) databases. Differentially expressed genes were screened, followed by the Kyoto Encyclopedia of Genes and Genomes (KEGG) and Gene Ontology (GO) analyses. Kaplan-Meier survival analysis was performed for genes related to the focal adhesion pathway, and multivariate Cox regression analysis was employed to construct a prognostic signature model. Genes such as SIGLEC15, TIGIT, CD274, HAVCR2, PDCD1, CTLA4, and LAG3 were extracted from the TARGET and CCLE databases for osteosarcoma patients and osteosarcoma cell lines, respectively,to observe the expression of immune checkpoint-related genes. Finally, qRT-PCR was used to verify the expression of these immune checkpoint-related genes in osteosarcoma cell lines. Results In our study, 376 samples were analyzed, including 369 osteosarcoma samples and 7 normal tissue samples. We identified 50 up-regulated and 28 down-regulated differentially expressed genes. Among these, 10 Candidate genes relative to focal Adhesion were selected, and CAV1, ZYX, and ITGA5 were found to have a significant prognostic role based on survival analysis of osteosarcoma samples from the TARGET database. A predictive signature model related to the focal adhesion signaling pathway was constructed using these genes, and the AUCs of the 1-year, 3-year, and 5-year ROC curves were 0. 647, 0. 712, and 0. 717, respectively. The overall survival (OS) rate of osteosarcoma patients with high-risk scores was poorer than those with low-risk scores. Then, samples were divided into two subgroups based on the expression of the three genes, revealing significant differences in the expression of certain immune checkpoint-related genes between the subgroups. Additionally, above three genes and immune checkpoint-related genes in osteosarcoma cell lines were extracted from the CCLE database, showing high expression levels in eight osteosarcoma cell lines. We observed that CD274 and PDCD1LG2 were highly expressed in some osteosarcoma cell lines. Finally, the expression of CAV1, ZYX, ITGA5, CD80, CD274, and PDCD1LG2 in osteosarcoma cell lines was verified by qRT-PCR. Conclusions Our study validated the prognostic role of three focal adhesion pathway-related genes (ZYX, CAV1, and ITGA5) in patients with osteosarcoma and constructed a prognostic signature model associated with the focal adhesion signaling pathway. We identified significant differences in the expression of multiple immune checkpoint-related genes among subgroups defined by the three genes. Additionally, CD274 and PDCD1LG2 showed higher expression in osteosarcoma cell lines characterized by these genes. These findings may aid in the selection of effective immunotherapy for specific osteosarcoma patients.
Collapse
Affiliation(s)
- Zhiqiang Wu
- Department of Musculoskeletal Oncology, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Zhiqing Wang
- Zhabei Central Hospital, No. 619, Zhonghuaxin Road, Jing'an District, Shanghai, 200070, China
| | - Zhanqiang Hua
- Department of Orthopedics, Shanghai Electric Power Hospital, Shanghai, 200050, China
| | - Yingzheng Ji
- Department of Orthopedics, Naval Medical Center of PLA, Second Military Medical University, China
| | - Qingrong Ye
- Department of Musculoskeletal Oncology, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Hao Zhang
- Department of Orthopedics, Naval Medical Center of PLA, Second Military Medical University, China
| | - Wangjun Yan
- Department of Musculoskeletal Oncology, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| |
Collapse
|
2
|
Wang L, Wang X, Zhu X, Zhong L, Jiang Q, Wang Y, Tang Q, Li Q, Zhang C, Wang H, Zou D. Drug resistance in ovarian cancer: from mechanism to clinical trial. Mol Cancer 2024; 23:66. [PMID: 38539161 PMCID: PMC10976737 DOI: 10.1186/s12943-024-01967-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2023] [Accepted: 02/22/2024] [Indexed: 04/05/2024] Open
Abstract
Ovarian cancer is the leading cause of gynecological cancer-related death. Drug resistance is the bottleneck in ovarian cancer treatment. The increasing use of novel drugs in clinical practice poses challenges for the treatment of drug-resistant ovarian cancer. Continuing to classify drug resistance according to drug type without understanding the underlying mechanisms is unsuitable for current clinical practice. We reviewed the literature regarding various drug resistance mechanisms in ovarian cancer and found that the main resistance mechanisms are as follows: abnormalities in transmembrane transport, alterations in DNA damage repair, dysregulation of cancer-associated signaling pathways, and epigenetic modifications. DNA methylation, histone modifications and noncoding RNA activity, three key classes of epigenetic modifications, constitute pivotal mechanisms of drug resistance. One drug can have multiple resistance mechanisms. Moreover, common chemotherapies and targeted drugs may have cross (overlapping) resistance mechanisms. MicroRNAs (miRNAs) can interfere with and thus regulate the abovementioned pathways. A subclass of miRNAs, "epi-miRNAs", can modulate epigenetic regulators to impact therapeutic responses. Thus, we also reviewed the regulatory influence of miRNAs on resistance mechanisms. Moreover, we summarized recent phase I/II clinical trials of novel drugs for ovarian cancer based on the abovementioned resistance mechanisms. A multitude of new therapies are under evaluation, and the preliminary results are encouraging. This review provides new insight into the classification of drug resistance mechanisms in ovarian cancer and may facilitate in the successful treatment of resistant ovarian cancer.
Collapse
Affiliation(s)
- Ling Wang
- Department of Gynecologic Oncology, Chongqing University Cancer Hospital & Chongqing Cancer Institute & Chongqing Cancer Hospital, Chongqing, China
- Chongqing Specialized Medical Research Center of Ovarian Cancer, Chongqing, China
- Organoid Transformational Research Center, Chongqing Key Laboratory of Translational Research for Cancer Metastasis and Individualized Treatment, Chongqing University Cancer Hospital, Chongqing, China
| | - Xin Wang
- Department of Gynecologic Oncology, Chongqing University Cancer Hospital & Chongqing Cancer Institute & Chongqing Cancer Hospital, Chongqing, China
- Chongqing Specialized Medical Research Center of Ovarian Cancer, Chongqing, China
- Organoid Transformational Research Center, Chongqing Key Laboratory of Translational Research for Cancer Metastasis and Individualized Treatment, Chongqing University Cancer Hospital, Chongqing, China
| | - Xueping Zhu
- Department of Gynecologic Oncology, Chongqing University Cancer Hospital & Chongqing Cancer Institute & Chongqing Cancer Hospital, Chongqing, China
- Chongqing Specialized Medical Research Center of Ovarian Cancer, Chongqing, China
- Organoid Transformational Research Center, Chongqing Key Laboratory of Translational Research for Cancer Metastasis and Individualized Treatment, Chongqing University Cancer Hospital, Chongqing, China
| | - Lin Zhong
- Department of Gynecologic Oncology, Chongqing University Cancer Hospital & Chongqing Cancer Institute & Chongqing Cancer Hospital, Chongqing, China
- Chongqing Specialized Medical Research Center of Ovarian Cancer, Chongqing, China
- Organoid Transformational Research Center, Chongqing Key Laboratory of Translational Research for Cancer Metastasis and Individualized Treatment, Chongqing University Cancer Hospital, Chongqing, China
| | - Qingxiu Jiang
- Department of Gynecologic Oncology, Chongqing University Cancer Hospital & Chongqing Cancer Institute & Chongqing Cancer Hospital, Chongqing, China
- Chongqing Specialized Medical Research Center of Ovarian Cancer, Chongqing, China
- Organoid Transformational Research Center, Chongqing Key Laboratory of Translational Research for Cancer Metastasis and Individualized Treatment, Chongqing University Cancer Hospital, Chongqing, China
| | - Ya Wang
- Department of Gynecologic Oncology, Chongqing University Cancer Hospital & Chongqing Cancer Institute & Chongqing Cancer Hospital, Chongqing, China
- Chongqing Specialized Medical Research Center of Ovarian Cancer, Chongqing, China
- Organoid Transformational Research Center, Chongqing Key Laboratory of Translational Research for Cancer Metastasis and Individualized Treatment, Chongqing University Cancer Hospital, Chongqing, China
| | - Qin Tang
- Department of Gynecologic Oncology, Chongqing University Cancer Hospital & Chongqing Cancer Institute & Chongqing Cancer Hospital, Chongqing, China
- Chongqing Specialized Medical Research Center of Ovarian Cancer, Chongqing, China
- Organoid Transformational Research Center, Chongqing Key Laboratory of Translational Research for Cancer Metastasis and Individualized Treatment, Chongqing University Cancer Hospital, Chongqing, China
| | - Qiaoling Li
- Department of Gynecologic Oncology, Chongqing University Cancer Hospital & Chongqing Cancer Institute & Chongqing Cancer Hospital, Chongqing, China
- Chongqing Specialized Medical Research Center of Ovarian Cancer, Chongqing, China
- Organoid Transformational Research Center, Chongqing Key Laboratory of Translational Research for Cancer Metastasis and Individualized Treatment, Chongqing University Cancer Hospital, Chongqing, China
| | - Cong Zhang
- Chongqing Specialized Medical Research Center of Ovarian Cancer, Chongqing, China
- Organoid Transformational Research Center, Chongqing Key Laboratory of Translational Research for Cancer Metastasis and Individualized Treatment, Chongqing University Cancer Hospital, Chongqing, China
- Biological and Pharmaceutical Engineering, School of Medicine, Chongqing University, Chongqing, China
| | - Haixia Wang
- Department of Gynecologic Oncology, Chongqing University Cancer Hospital & Chongqing Cancer Institute & Chongqing Cancer Hospital, Chongqing, China.
- Chongqing Specialized Medical Research Center of Ovarian Cancer, Chongqing, China.
- Organoid Transformational Research Center, Chongqing Key Laboratory of Translational Research for Cancer Metastasis and Individualized Treatment, Chongqing University Cancer Hospital, Chongqing, China.
| | - Dongling Zou
- Department of Gynecologic Oncology, Chongqing University Cancer Hospital & Chongqing Cancer Institute & Chongqing Cancer Hospital, Chongqing, China.
- Chongqing Specialized Medical Research Center of Ovarian Cancer, Chongqing, China.
- Organoid Transformational Research Center, Chongqing Key Laboratory of Translational Research for Cancer Metastasis and Individualized Treatment, Chongqing University Cancer Hospital, Chongqing, China.
| |
Collapse
|
3
|
Chen X, Yu Y, Su Y, Shi L, Xie S, Hong Y, Liu X, Yin F. Low FHL1 expression indicates a good prognosis and drug sensitivity in ovarian cancer. Funct Integr Genomics 2024; 24:25. [PMID: 38324167 DOI: 10.1007/s10142-024-01294-2] [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: 11/22/2023] [Revised: 01/01/2024] [Accepted: 01/06/2024] [Indexed: 02/08/2024]
Abstract
Chemotherapy resistance is the main reason for the poor prognosis of ovarian cancer (OC). FHL1 is an important tumour regulator, but its relationship with the prognosis, drug resistance, and tumour microenvironment of OC is unknown. Immunohistochemistry was used to determine FHL1 expression in OC. Kaplan‒Meier plotter was used for survival analysis. The value of gene expression in predicting drug resistance was estimated using the area under the curve (AUC). Bivariate correlation was used to determine the coexpression of two genes. Functional cluster and pathway enrichment were used to uncover hidden signalling pathways. The relationship between gene levels and the tumour microenvironment was visualised through the ggstatsplot and pheatmap packages. The mRNA and protein levels of FHL1 were downregulated in 426 and 100 OC tissues, respectively. Low FHL1 expression was correlated with good progression-free survival (PFS), postprogression survival, and overall survival (OS) in 1815 OC patients, and was further confirmed to be associated with good OS by immunohistochemistry in 152 OC tissues. Furthermore, FHL1 was downregulated in drug-sensitive tissues, while its high expression predicted drug resistance (AUC > 0.65). Mechanistically, FHL1 was coexpressed with FLNC, CAV1, PPP1R12B, and FLNA at the mRNA and protein levels in 558 and 174 OC tissues, respectively, and their expression was downregulated in OC. Additionally, very strong coexpression of FHL1 with the four genes was identified in at least 23 different tumours. Low expression of the four genes was associated with good PFS, and the combination of FHL1 with the four genes provided better prognostic power. Meanwhile, the expression of all five genes was strongly and positively associated with the abundance of macrophages. Low FHL1 expression acts as a favourable factor in OC, probably via positive coexpression with FLNC, CAV1, PPP1R12B, and FLNA.
Collapse
Affiliation(s)
- Xiaoying Chen
- Key Laboratory of Longevity and Ageing-Related Disease of Chinese Ministry of Education, Centre for Translational Medicine and School of Basic Medical Sciences, Guangxi Medical University, Nanning, 530021, Guangxi, China
| | - Yue Yu
- Life Sciences Institute, Guangxi Medical University, Nanning, 530021, Guangxi, China
| | - Yuting Su
- Life Sciences Institute, Guangxi Medical University, Nanning, 530021, Guangxi, China
| | - Lizhou Shi
- Life Sciences Institute, Guangxi Medical University, Nanning, 530021, Guangxi, China
| | - Shanzhou Xie
- Key Laboratory of Longevity and Ageing-Related Disease of Chinese Ministry of Education, Centre for Translational Medicine and School of Basic Medical Sciences, Guangxi Medical University, Nanning, 530021, Guangxi, China
| | - Yi Hong
- Key Laboratory of Longevity and Ageing-Related Disease of Chinese Ministry of Education, Centre for Translational Medicine and School of Basic Medical Sciences, Guangxi Medical University, Nanning, 530021, Guangxi, China
| | - Xia Liu
- Key Laboratory of Longevity and Ageing-Related Disease of Chinese Ministry of Education, Centre for Translational Medicine and School of Basic Medical Sciences, Guangxi Medical University, Nanning, 530021, Guangxi, China.
- Key Laboratory of Human Development and Disease Research (Guangxi Medical University), Education Department of Guangxi Zhang Autonomous Region, Nanning, 530021, Guangxi, China.
| | - Fuqiang Yin
- Life Sciences Institute, Guangxi Medical University, Nanning, 530021, Guangxi, China.
- Key Laboratory of High-Incidence-Tumor Prevention and Treatment (Guangxi Medical University), Ministry of Education, Nanning, 530021, Guangxi, China.
| |
Collapse
|
4
|
Mazarei M, Shahabi Rabori V, Ghasemi N, Salehi M, Rayatpisheh N, Jahangiri N, Saberiyan M. LncRNA MALAT1 signaling pathway and clinical applications in overcome on cancers metastasis. Clin Exp Med 2023; 23:4457-4472. [PMID: 37695391 DOI: 10.1007/s10238-023-01179-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2023] [Accepted: 08/27/2023] [Indexed: 09/12/2023]
Abstract
In spite of its high mortality rate and difficulty in finding a cure, scientific advancements have contributed to a reduction in cancer-related fatalities. Aberrant gene expression during carcinogenesis emphasizes the importance of targeting the signaling networks that control gene expression in cancer treatment. Long noncoding RNAs (lncRNAs), which are transcribed RNA molecules that play a role in gene expression regulation, are a recent innovative therapeutic approach for diagnosing and treating malignancies. MALAT1, a well-known lncRNA, functions in gene expression, RNA processing, and epigenetic control. High expression levels of MALAT1 are associated with several human disorders, including metastasis, invasion, autophagy, and proliferation of cancer cells. MALAT1 affects various signaling pathways and microRNAs (miRNAs), and this study aims to outline its functional roles in cancer metastasis and its interactions with cellular signaling pathways. Moreover, MALAT1 and its interactions with signaling pathways can be promising target for cancer treatment.
Collapse
Affiliation(s)
- Madineh Mazarei
- Cellular and Molecular Research Center, Basic Health Sciences Institute, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | | | - Nazila Ghasemi
- Department of Biology, Jahrom Branch, Islamic Azad University, Jahrom, Iran
| | - Mehrnaz Salehi
- School of Medicine, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | - Najmeh Rayatpisheh
- School of Medicine, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | - Negin Jahangiri
- Department of Biology, Faculty of Basic Sciences and Engineering, Gonbad Kavous University, Gonbad-e Kavus, Iran
| | - Mohammadreza Saberiyan
- Department of Medical Genetics, Faculty of Medicine, School of Medical Sciences, Hormozgan University of Medical Sciences, Bandar Abbas, Iran.
| |
Collapse
|
5
|
Surendra Panikar S, Shmuel S, Lewis JS, Pereira PMR. PET and Optical Imaging of Caveolin-1 in Gastric Tumors. ACS OMEGA 2023; 8:35884-35892. [PMID: 37810678 PMCID: PMC10552508 DOI: 10.1021/acsomega.3c03614] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/23/2023] [Accepted: 09/05/2023] [Indexed: 10/10/2023]
Abstract
Previous studies have suggested tumoral caveolin-1 (CAV1) as a predictive biomarker for the response to anti-HER2 antibody drug therapies in gastric tumors. In this study, radiolabeled and fluorescently labeled anti-CAV1 antibodies were developed and tested as an immunoPET or optical imaging agent to detect CAV1 in HER2-positive/CAV1-high NCIN87 gastric tumors. The expression of CAV1 receptors in NCIN87 gastric tumors and nontumor murine organs was determined by Western blot. Binding assays were performed to validate the anti-CAV1 antibody specificity for CAV1-expressing NCIN87 cancer cells. Subcutaneous and orthotopic NCIN87 xenografts were used for PET imaging and ex vivo biodistribution of the radioimmunoconjugate. Additional HER2-PET and CAV1-optical imaging was also performed to determine CAV1 in the HER2-positive tumors. 89Zr-labeled anti-CAV1 antibody was able to bind to CAV1-expressing NCIN87 cells with a Bmax value of 2.7 × 103 CAV1 receptors/cell in vitro. ImmunoPET images demonstrated the localization of the antibody in subcutaneous NCIN87 xenografts. In the orthotopic model, CAV1 expression was also observed by optical imaging in the HER2-positive tumors previously imaged with HER2-PET. Ex vivo biodistribution analysis further confirmed these imaging results. The preclinical data from this study demonstrate the potential of using CAV1-PET and optical imaging for detecting gastric tumors.
Collapse
Affiliation(s)
- Sandeep Surendra Panikar
- Department
of Radiology, Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, Missouri 63110, United States
| | - Shayla Shmuel
- Department
of Radiology, Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, Missouri 63110, United States
| | - Jason S. Lewis
- Department
of Radiology, Memorial Sloan Kettering Cancer
Center, New York, New York 10065, United States
- Department of Pharmacology, Weill Cornell Medical College, New York, New York 10065, United States
- Molecular
Pharmacology Program, Memorial Sloan Kettering
Cancer Center, New York, New York 10065, United States
- Department
of Radiology, Weill Cornell Medical College, New York, New York 10065, United States
- Radiochemistry
and Molecular Imaging Probes Core, Memorial
Sloan Kettering Cancer Center, New York, New York 10065, United States
| | - Patrícia M. R. Pereira
- Department
of Radiology, Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, Missouri 63110, United States
| |
Collapse
|
6
|
Wang W, Liu R, Liao W, Ji L, Mei J, Su D. NOTCH2 gene mutation and gamma-secretase inhibitor in mediating the malignancy of ovarian cancer. Aging (Albany NY) 2023; 15:9743-9758. [PMID: 37728427 PMCID: PMC10564443 DOI: 10.18632/aging.205045] [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: 04/10/2023] [Accepted: 08/25/2023] [Indexed: 09/21/2023]
Abstract
The carcinogenic mechanisms by which serous ovarian cancer (OC) occurs remain to be explored. Currently, we have conducted whole-exome sequencing (WES) and targeted deep sequencing to validate new molecular markers, including NOTCH2, that impede the progression of cell malignancy in ovarian cancer (OC). Following NOTCH2 P2113S mutation and NOTCH signaling pathway inhibitor N-[N-(3,5-difluorophenacetyl)-L-alanyl]-S-phenylglycine t-butyl ester (DAPT) treatment, the cell proliferation, migration, and invasion of A2780 and SKOV3 OC cells were examined in vitro. WES identified the P2113S point mutation in NOTCH2. The NOTCH2 mutation rate was 26.67 % among the 75 OC cases. The NOTCH2 P2113S mutation and DAPT treatment downregulated Notch-2 protein levels in the two OC cells. Functionally, interfering with NOTCH2 expression promoted the migrative, proliferative, and invasive capacities of OC cells. Western blotting further confirmed that NOTCH2-mediated tumorigenesis lies in reducing apoptosis through dysregulation of Bax/Bcl2, affecting repair of DNA damage through reducing DNA-PK and blocking the transcription factor Hes1 along with increasing immune regulator p65. Furthermore, the NOTCH2-mediated tumorigenesis was mostly reversed after NF-κB inhibitor Bay11-7082 treatment. These findings identified the NOTCH2 P2113S mutation in ovarian carcinogenesis, and NOTCH2 P2113S is a potential target in treating OC.
Collapse
Affiliation(s)
- Wenjing Wang
- School of Medicine, University of Electronic Science and Technology of China, Chengdu 610054, Sichuan, China
- Department of Gynecology and Obstetrics, Sichuan Academy of Medical Sciences and Sichuan Provincial People’s Hospital, University of Electronic Science and Technology of China, Chengdu 610054, Sichuan, China
| | - Ruiqian Liu
- Deyang People’s Hospital, Deyang 618099, Sichuan, China
| | - Wei Liao
- Department of Gynecology and Obstetrics, Sichuan Academy of Medical Sciences and Sichuan Provincial People’s Hospital, University of Electronic Science and Technology of China, Chengdu 610054, Sichuan, China
| | - Landie Ji
- School of Medicine, University of Electronic Science and Technology of China, Chengdu 610054, Sichuan, China
- Department of Gynecology and Obstetrics, Sichuan Academy of Medical Sciences and Sichuan Provincial People’s Hospital, University of Electronic Science and Technology of China, Chengdu 610054, Sichuan, China
| | - Jie Mei
- Department of Gynecology and Obstetrics, Sichuan Academy of Medical Sciences and Sichuan Provincial People’s Hospital, University of Electronic Science and Technology of China, Chengdu 610054, Sichuan, China
- Chinese Academy of Sciences Sichuan Translational Medicine Research Hospital, Chengdu 610054, Sichuan, China
| | - Dan Su
- Department of Gynecology and Obstetrics, Sichuan Academy of Medical Sciences and Sichuan Provincial People’s Hospital, University of Electronic Science and Technology of China, Chengdu 610054, Sichuan, China
- Chinese Academy of Sciences Sichuan Translational Medicine Research Hospital, Chengdu 610054, Sichuan, China
| |
Collapse
|
7
|
Nagappan A, Kim KH, Moon Y. Caveolin-1-ACE2 axis modulates xenobiotic metabolism-linked chemoresistance in ovarian clear cell carcinoma. Cell Biol Toxicol 2023; 39:1181-1201. [PMID: 35622184 PMCID: PMC9136213 DOI: 10.1007/s10565-022-09733-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2022] [Accepted: 05/17/2022] [Indexed: 11/06/2022]
Abstract
Among epithelial ovarian cancers, ovarian clear cell carcinoma (OCCC) remains markedly resistant to platinum-based chemotherapy, leading to poor clinical outcomes. In response to xenobiotic insults, caveolar platforms play crucial roles in modulating stress signaling responses in cancer cells. It has been hypothesized that caveolin-1 (Cav-1), a main component of the lipid raft, may regulate the response to platinum-based treatment in OCCC. The clinical transcriptomic evaluation demonstrated that high Cav-1 expression was positively associated with a favorable prognosis in patients with ovarian cancer. Cav-1 overexpression enhanced sensitivity to cisplatin (CDDP) treatment, whereas Cav-1 deficiency promoted chemoresistance in OCCC cells. Mechanistically, although Cav-1 counteracted angiotensin-converting enzyme 2 (ACE2) expression, ACE2 positively facilitated resistance to CDDP in OCCC cells. Furthermore, ACE2 restricted aryl hydrocarbon receptor expression and subsequent transcription of drug-metabolizing enzymes. Of note, ACE2 positively regulated the expression of the platinum-clearing enzyme CYP3A4. These findings suggest that the Cav-1-ACE2 axis modulates xenobiotic metabolism-linked chemoresistance in OCCC, predicting potential roles for the stress sentinel networks in oncogenic processes.
Collapse
Affiliation(s)
- Arulkumar Nagappan
- Laboratory of Mucosal Exposome and Biomodulation, Department of Integrative Biomedical Sciences, Pusan National University, Yangsan, 50612, Korea
| | - Ki-Hyung Kim
- Laboratory of Mucosal Exposome and Biomodulation, Department of Integrative Biomedical Sciences, Pusan National University, Yangsan, 50612, Korea
- Department of Obstetrics and Gynecology, College of Medicine, Pusan National University, Busan, Korea
- Biomedical Research Institute, Pusan National University Hospital, Busan, Korea
| | - Yuseok Moon
- Laboratory of Mucosal Exposome and Biomodulation, Department of Integrative Biomedical Sciences, Pusan National University, Yangsan, 50612, Korea.
- Biomedical Research Institute, Pusan National University Hospital, Busan, Korea.
- Graduate Program of Genomic Data Sciences, Pusan National University, Yangsan, 50612, Korea.
| |
Collapse
|
8
|
Rahimian N, Sheida A, Rajabi M, Heidari MM, Tobeiha M, Esfahani PV, Ahmadi Asouri S, Hamblin MR, Mohamadzadeh O, Motamedzadeh A, Khaksary Mahabady M. Non-coding RNAs and exosomal non-coding RNAs in pituitary adenoma. Pathol Res Pract 2023; 248:154649. [PMID: 37453360 DOI: 10.1016/j.prp.2023.154649] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/17/2023] [Revised: 06/25/2023] [Accepted: 06/25/2023] [Indexed: 07/18/2023]
Abstract
Pituitary adenoma (PA) is the third most common primary intracranial tumor in terms of overall disease incidence. Although they are benign tumors, they can have a variety of clinical symptoms, but are mostly asymptomatic, which often leads to diagnosis at an advanced stage when surgical intervention is ineffective. Earlier identification of PA could reduce morbidity and allow better clinical management of the affected patients. Non-coding RNAs (ncRNAs) do not generally code for proteins, but can modulate biological processes at the post-transcriptional level through a variety of molecular mechanisms. An increased number of ncRNA expression profiles have been found in PAs. Therefore, understanding the expression patterns of different ncRNAs could be a promising method for developing non-invasive biomarkers. This review summarizes the expression patterns of dysregulated ncRNAs (microRNAs, long non-coding RNAs, and circular RNAs) involved in PA, which could one day serve as innovative biomarkers or therapeutic targets for the treatment of this neoplasia. We also discuss the potential molecular pathways by which the dysregulated ncRNAs could cause PA and affect its progression.
Collapse
Affiliation(s)
- Neda Rahimian
- Endocrine Research Center, Institute of Endocrinology and Metabolism, Iran University of Medical Sciences (IUMS), Tehran, Iran; Department of Internal Medicine, School of Medicine, Firoozgar Hospital, Iran University of Medical Sciences, Tehran, Iran
| | - Amirhossein Sheida
- Student Research Committee, Kashan University of Medical Sciences, Kashan, Iran; School of Medicine, Kashan University of Medical Sciences, Kashan, Iran
| | - Mohammadreza Rajabi
- Department of Pathology, Transplant Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Mohammad Mahdi Heidari
- School of Medicine, Kashan University of Medical Sciences, Kashan, Iran; Department of Pediatric, Faculty of Medicine, Kashan University of Medical Sciences, Kashan, Iran
| | - Mohammad Tobeiha
- Student Research Committee, Kashan University of Medical Sciences, Kashan, Iran; School of Medicine, Kashan University of Medical Sciences, Kashan, Iran
| | - Pegah Veradi Esfahani
- Student Research Committee, Kashan University of Medical Sciences, Kashan, Iran; School of Medicine, Kashan University of Medical Sciences, Kashan, Iran
| | - Sahar Ahmadi Asouri
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, Iran
| | - Michael R Hamblin
- Laser Research Centre, Faculty of Health Science, University of Johannesburg, Doornfontein, South Africa
| | - Omid Mohamadzadeh
- Department of Neurological Surgery, Imam Khomeini Hospital Complex, Tehran University of Medical Sciences, Tehran, Iran.
| | - Alireza Motamedzadeh
- Department of Internal Medicine, Faculty of Medicine, Kashan University of Medical Sciences, Kashan, Iran.
| | - Mahmood Khaksary Mahabady
- Anatomical Sciences Research Center, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, Iran.
| |
Collapse
|
9
|
Rajendran NK, Liu W, Cahill PA, Redmond EM. Caveolin-1 inhibition mediates the opposing effects of alcohol on γ-secretase activity in arterial endothelial and smooth muscle cells. Physiol Rep 2023; 11:e15544. [PMID: 36635975 PMCID: PMC9837422 DOI: 10.14814/phy2.15544] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Revised: 12/01/2022] [Accepted: 12/04/2022] [Indexed: 04/18/2023] Open
Abstract
Notch is important to vessel homeostasis. We investigated the mechanistic role of caveolin-1 (Cav-1) in mediating the effects of alcohol (Ethanol/EtOH) on the γ-secretase proteolytic activity necessary for Notch signaling in vascular cells. Human coronary artery endothelial cells (HCAEC) were treated with EtOH (0-50 mM), Notch ligand delta-like ligand 4 (Dll4), and the γ-secretase inhibitor DAPT. EtOH stimulated Notch signaling in HCAEC as evidenced by increased Notch receptor (N1, N4) and target gene (hrt2, hrt3) mRNA levels with the most robust response achieved at 25 mM EtOH. Ethanol (25 mM) stimulated γ-secretase proteolytic activity, to the same extent as Dll4, in HCAEC membranes. Ethanol inhibited Cav-1 mRNA and protein levels in HCAEC. Caveolin-1 negatively regulated γ-secretase activity in HCAEC as Cav-1 knockdown stimulated it, while Cav-1 overexpression inhibited it. Moreover, Cav-1 overexpression blocked the stimulatory effect of EtOH on γ-secretase activity in HCAEC. Although EtOH also inhibited Cav-1 expression in human coronary artery smooth muscle cells (HCASMC), EtOH inhibited γ-secretase activity in HCASMC in contrast to its effect in HCAEC. The inhibitory effect of EtOH on γ-secretase in HCASMC was mimicked by Cav-1 knockdown and prevented by Cav-1 overexpression, suggesting that in these cells Cav-1 positively regulates γ-secretase activity. In conclusion, EtOH differentially regulates γ-secretase activity in arterial EC and SMC, being stimulatory and inhibitory, respectively. These effects are both mediated by caveolin-1 inhibition which itself has opposite effects on γ-secretase in the two cell types. This mechanism may underlie, in part, the effects of moderate drinking on atherosclerosis.
Collapse
Affiliation(s)
- Naresh K. Rajendran
- Department of SurgeryUniversity of Rochester Medical CenterRochesterNew YorkUSA
| | - Weimin Liu
- Department of SurgeryUniversity of Rochester Medical CenterRochesterNew YorkUSA
| | - Paul A. Cahill
- Vascular Biology and Therapeutics Laboratory, School of BiotechnologyDublin City UniversityDublinIreland
| | - Eileen M. Redmond
- Department of SurgeryUniversity of Rochester Medical CenterRochesterNew YorkUSA
| |
Collapse
|
10
|
Low JY, Laiho M. Caveolae-Associated Molecules, Tumor Stroma, and Cancer Drug Resistance: Current Findings and Future Perspectives. Cancers (Basel) 2022; 14:cancers14030589. [PMID: 35158857 PMCID: PMC8833326 DOI: 10.3390/cancers14030589] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Revised: 01/21/2022] [Accepted: 01/22/2022] [Indexed: 02/04/2023] Open
Abstract
Simple Summary Cell membranes contain small invaginations called caveolae. They are a specialized lipid domain and orchestrate cellular signaling events, mechanoprotection, and lipid homeostasis. Formation of the caveolae depends on two classes of proteins, the caveolins and cavins, which form large complexes that allow their self-assembly into caveolae. Loss of either of these two proteins leads to distortion of the caveolae structure and disruption of many physiological processes that affect diseases of the muscle, metabolic states governing lipids, and the glucose balance as well as cancers. In cancers, the expression of caveolins and cavins is heterogenous, and they undergo alterations both in the tumors and the surrounding tumor microenvironment stromal cells. Remarkably, their expression and function has been associated with resistance to many cancer drugs. Here, we summarize the current knowledge of the resistance mechanisms and how this knowledge could be applied into the clinic in future. Abstract The discovery of small, “cave-like” invaginations at the plasma membrane, called caveola, has opened up a new and exciting research area in health and diseases revolving around this cellular ultrastructure. Caveolae are rich in cholesterol and orchestrate cellular signaling events. Within caveola, the caveola-associated proteins, caveolins and cavins, are critical components for the formation of these lipid rafts, their dynamics, and cellular pathophysiology. Their alterations underlie human diseases such as lipodystrophy, muscular dystrophy, cardiovascular disease, and diabetes. The expression of caveolins and cavins is modulated in tumors and in tumor stroma, and their alterations are connected with cancer progression and treatment resistance. To date, although substantial breakthroughs in cancer drug development have been made, drug resistance remains a problem leading to treatment failures and challenging translation and bench-to-bedside research. Here, we summarize the current progress in understanding cancer drug resistance in the context of caveola-associated molecules and tumor stroma and discuss how we can potentially design therapeutic avenues to target these molecules in order to overcome treatment resistance.
Collapse
Affiliation(s)
- Jin-Yih Low
- Department of Radiation Oncology and Molecular Radiation Sciences, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA;
- Correspondence: ; Tel.: +1-410-502-9748; Fax: +1-410-502-2821
| | - Marikki Laiho
- Department of Radiation Oncology and Molecular Radiation Sciences, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA;
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| |
Collapse
|
11
|
Leiser D, Samanta S, Eley J, Strauss J, Creed M, Kingsbury T, Staats PN, Bhandary B, Chen M, Dukic T, Roy S, Mahmood J, Vujaskovic Z, Shukla HD. Role of caveolin-1 as a biomarker for radiation resistance and tumor aggression in lung cancer. PLoS One 2021; 16:e0258951. [PMID: 34762666 PMCID: PMC8584669 DOI: 10.1371/journal.pone.0258951] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2021] [Accepted: 10/10/2021] [Indexed: 01/14/2023] Open
Abstract
Radiation therapy plays a major role in the treatment of lung cancer patients. However, cancer cells develop resistance to radiation. Tumor radioresistance is a complex multifactorial mechanism which may be dependent on DNA damage and repair, hypoxic conditions inside tumor microenvironment, and the clonal selection of radioresistant cells from the heterogeneous tumor site, and it is a major cause of treatment failure in non-small cell lung cancer (NSCLC). In the present investigation caveolin-1 (CAV-1) has been observed to be highly expressed in radiation resistant A549 lung cancer cells. CRISPR-Cas9 knockout of CAV-1 reverted the cells to a radio sensitive phenotype. In addition, CAV-1 overexpression in parental A549 cells, led to radiation resistance. Further, gene expression analysis of A549 parental, radiation resistant, and caveolin-1 overexpressed cells, exhibited overexpression of DNA repair genes RAD51B, RAD18, SOX2 cancer stem cell marker, MMPs, mucins and cytoskeleton proteins in resistant and caveolin-1 over expressed A549 cells, as compared to parental A549 cells. Bioinformatic analysis shows upregulation of BRCA1, Nuclear Excision DNA repair, TGFB and JAK/STAT signaling pathways in radioresistant and caveolin-1 overexpressed cells, which may functionally mediate radiation resistance. Immunohistochemistry data demonstrated heterogeneous expression of CAV-1 gene in human lung cancer tissues, which was analogous to its enhanced expression in human lung cancer cell line model and mouse orthotopic xenograft lung cancer model. Also, TCGA PanCancer clinical studies have demonstrated amplification, deletions and missense mutation in CAV-1 gene in lung cancer patients, and that CAV-1 alteration has been linked to poor prognosis, and poor survival in lung cancer patients. Interestingly, we have also optimized ELISA assay to measure caveolin-1 protein in the blood of A549 radiation resistant human xenograft preclinical mouse model and discovered higher level of caveolin-1 (950 pg/ml) in tumor bearing animals treated with radiation, as compared to xenograft with radiosensitive lung cancer cells (450 pg/ml). Thus, we conclude that caveolin-1 is involved in radio-resistance and contributes to tumor aggression, and it has potential to be used as prognostic biomarker for radiation treatment response, and tumor progression for precision medicine in lung cancer patients.
Collapse
Affiliation(s)
- Dominic Leiser
- Division of Translational Radiation Sciences (DTRS), Department of Radiation Oncology, University of Maryland School of Medicine, Baltimore, MD, United States of America
| | - Santanu Samanta
- Division of Translational Radiation Sciences (DTRS), Department of Radiation Oncology, University of Maryland School of Medicine, Baltimore, MD, United States of America
| | - John Eley
- Department of Radiation Oncology, School of Medicine, Vanderbilt University, Nashville, TN, United States of America
| | - Josh Strauss
- Department of Radiation Oncology, School of Medicine, Vanderbilt University, Nashville, TN, United States of America
| | - Michael Creed
- Department of Physiology, University of Maryland School of Medicine, Baltimore, MD, United States of America
| | - Tami Kingsbury
- Department of Physiology, University of Maryland School of Medicine, Baltimore, MD, United States of America
| | - Paul N. Staats
- Department of Pathology, University of Maryland, School of Medicine, Baltimore, MD, United States of America
| | - Binny Bhandary
- Division of Translational Radiation Sciences (DTRS), Department of Radiation Oncology, University of Maryland School of Medicine, Baltimore, MD, United States of America
| | - Minjie Chen
- Division of Translational Radiation Sciences (DTRS), Department of Radiation Oncology, University of Maryland School of Medicine, Baltimore, MD, United States of America
| | - Tijana Dukic
- Division of Translational Radiation Sciences (DTRS), Department of Radiation Oncology, University of Maryland School of Medicine, Baltimore, MD, United States of America
| | - Sanjit Roy
- Division of Translational Radiation Sciences (DTRS), Department of Radiation Oncology, University of Maryland School of Medicine, Baltimore, MD, United States of America
| | - Javed Mahmood
- Division of Translational Radiation Sciences (DTRS), Department of Radiation Oncology, University of Maryland School of Medicine, Baltimore, MD, United States of America
| | - Zeljko Vujaskovic
- Division of Translational Radiation Sciences (DTRS), Department of Radiation Oncology, University of Maryland School of Medicine, Baltimore, MD, United States of America
| | - Hem D. Shukla
- Division of Translational Radiation Sciences (DTRS), Department of Radiation Oncology, University of Maryland School of Medicine, Baltimore, MD, United States of America
| |
Collapse
|
12
|
Cheng D, Fan J, Qin K, Zhou Y, Yang J, Ma Y, Shi M, Jin J. LncRNA SNHG7 Regulates Mesenchymal Stem Cell Through the Notch1/Jagged1/Hes-1 Signaling Pathway and Influences Folfirinox Resistance in Pancreatic Cancer. Front Oncol 2021; 11:719855. [PMID: 34631547 PMCID: PMC8494469 DOI: 10.3389/fonc.2021.719855] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2021] [Accepted: 07/30/2021] [Indexed: 01/05/2023] Open
Abstract
Pancreatic cancer (PC) is one of the deadliest gastrointestinal cancers, accounting for the fourth highest number of cancer-related fatalities. Increasing data suggests that mesenchymal stem cells (MSCs) might influence the drug resistance of GC cells in the tumor microenvironment and play essential roles in drug resistance development. However, the precise underlying process remains a mystery. The purpose of this study was to look at the control of MSC-induced SNHG7 in pancreatic cancer. In vitro and in vivo sphere formation, colony formation, and flow cytometry investigations revealed the stemness and Folfirinox resistance in pancreatic cancer cells. To confirm the direct connections between SNHG7 and other related targets, RNA pulldown and immunoprecipitation tests were performed. MSC co-culture enhanced the stemness and Folfirinox resistance in pancreatic cancer cells according to the findings. MSC co-culture increased SNHG7 expression in pancreatic cancer cells, contributing to the stemness and Folfirinox resistance. We demonstrated that Notch1 interacted with SNHG7 and could reverse the facilitative effect of SNHG7 on the stemness and Folfirinox resistance in pancreatic cancer cells. Finally, our findings showed that MSCs increased SNHG7 expression in pancreatic cancer cells, promoting the stemness and Folfirinox resistance via the Notch1/Jagged1/Hes-1 signaling pathway. These findings could provide a novel approach and therapeutic target for pancreatic cancer patients.
Collapse
Affiliation(s)
- Dongfeng Cheng
- Pancreatic Disease Center, Department of General Surgery, Rui Jin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Juanjuan Fan
- Department of General Surgery, Yichuan Community Health, Shanghai, China
| | - Kai Qin
- Pancreatic Disease Center, Department of General Surgery, Rui Jin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yiran Zhou
- Pancreatic Disease Center, Department of General Surgery, Rui Jin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jingrui Yang
- Pancreatic Disease Center, Department of General Surgery, Rui Jin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yang Ma
- Pancreatic Disease Center, Department of General Surgery, Rui Jin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Minmin Shi
- Department of Surgery, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China.,Research Institute of Digestive Surgery, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Jiabin Jin
- Pancreatic Disease Center, Department of General Surgery, Rui Jin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
13
|
Cao RZ, Min L, Liu S, Tian RY, Jiang HY, Liu J, Shao LL, Cheng R, Zhu ST, Guo SL, Li P. Rictor Activates Cav 1 Through the Akt Signaling Pathway to Inhibit the Apoptosis of Gastric Cancer Cells. Front Oncol 2021; 11:641453. [PMID: 34540654 PMCID: PMC8442624 DOI: 10.3389/fonc.2021.641453] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Accepted: 03/08/2021] [Indexed: 01/01/2023] Open
Abstract
Background Rapamycin-insensitive companion of mammalian target of rapamycin (Rictor) protein is a core subunit of mammalian target of rapamycin complex 2, and is associated with cancer progression. However, the biological function of Rictor in cancer, particularly its clinical relevance in gastric cancer (GC) remains largely unknown. Methods Rictor expression and its association with clinicopathologic characteristics in GC were analyzed by immunohistochemistry. Effect of Rictor and Caveolin-1 (Cav 1) on GC cells apoptosis was evaluated via overexpression experiment in vitro. Mechanisms of Rictor and Cav 1 in GC were explored through overexpression and knockdown, by immunofluorescence and western blot analyses. Results Rictor was upregulated in GC, and mainly located in the cytoplasm of cancer cells. Moreover, higher Rictor levels were associated with worse prognosis. Rictor could inhibit GC cell apoptosis and promote cell growth in vitro. The results of immunofluorescence revealed that Cav 1 localized in GC cell membrane but did not co-localize with Rictor. Further, Rictor regulated apoptosis-related proteins, long non-coding RNAs and also activated cellular signaling, thereby positively regulating Cav 1 expression. This effect was attenuated by the Akt inhibitor ly294002. Cav 1 did not significantly affect the ability of Rictor to inhibit tumor cell apoptosis. Conclusions Rictor is upregulated in GC and associated with worse prognosis. It inhibits tumor apoptosis and activates Cav 1 through the Akt signaling pathway to inhibit the apoptosis of GC cells. Rictor is, therefore, a promising prognostic biomarker and possible therapeutic target in GC patients.
Collapse
Affiliation(s)
- Rui-Zhen Cao
- Department of Gastroenterology, Beijing Friendship Hospital, Capital Medical University, National Clinical Research Center for Digestive Disease, Beijing Digestive Disease Center, Beijing Key Laboratory for Precancerous Lesion of Digestive Disease, Beijing, China.,Department of Gastroenterology, Ordos Central Hospital, National Clinical Research Center for Digestive Disease-Ordos Subcenter, Ordos, China
| | - Li Min
- Department of Gastroenterology, Beijing Friendship Hospital, Capital Medical University, National Clinical Research Center for Digestive Disease, Beijing Digestive Disease Center, Beijing Key Laboratory for Precancerous Lesion of Digestive Disease, Beijing, China
| | - Si Liu
- Department of Gastroenterology, Beijing Friendship Hospital, Capital Medical University, National Clinical Research Center for Digestive Disease, Beijing Digestive Disease Center, Beijing Key Laboratory for Precancerous Lesion of Digestive Disease, Beijing, China
| | - Ru-Yue Tian
- Department of Gastroenterology, Beijing Friendship Hospital, Capital Medical University, National Clinical Research Center for Digestive Disease, Beijing Digestive Disease Center, Beijing Key Laboratory for Precancerous Lesion of Digestive Disease, Beijing, China
| | - Hai-Yan Jiang
- Department of Gastroenterology, Beijing Friendship Hospital, Capital Medical University, National Clinical Research Center for Digestive Disease, Beijing Digestive Disease Center, Beijing Key Laboratory for Precancerous Lesion of Digestive Disease, Beijing, China.,Department of Gastroenterology, Beijing University of Chinese Medicine Third Affiliated Hospital, Beijing, China
| | - Juan Liu
- Department of Gastroenterology, Beijing Friendship Hospital, Capital Medical University, National Clinical Research Center for Digestive Disease, Beijing Digestive Disease Center, Beijing Key Laboratory for Precancerous Lesion of Digestive Disease, Beijing, China.,Department of Gastroenterology, Shanxi Province Cancer Hospital, Shanxi Medical University, Taiyuan, China
| | - Lin-Lin Shao
- Department of Gastroenterology, Beijing Friendship Hospital, Capital Medical University, National Clinical Research Center for Digestive Disease, Beijing Digestive Disease Center, Beijing Key Laboratory for Precancerous Lesion of Digestive Disease, Beijing, China
| | - Rui Cheng
- Department of Gastroenterology, Beijing Friendship Hospital, Capital Medical University, National Clinical Research Center for Digestive Disease, Beijing Digestive Disease Center, Beijing Key Laboratory for Precancerous Lesion of Digestive Disease, Beijing, China
| | - Sheng-Tao Zhu
- Department of Gastroenterology, Beijing Friendship Hospital, Capital Medical University, National Clinical Research Center for Digestive Disease, Beijing Digestive Disease Center, Beijing Key Laboratory for Precancerous Lesion of Digestive Disease, Beijing, China
| | - Shui-Long Guo
- Department of Gastroenterology, Beijing Friendship Hospital, Capital Medical University, National Clinical Research Center for Digestive Disease, Beijing Digestive Disease Center, Beijing Key Laboratory for Precancerous Lesion of Digestive Disease, Beijing, China
| | - Peng Li
- Department of Gastroenterology, Beijing Friendship Hospital, Capital Medical University, National Clinical Research Center for Digestive Disease, Beijing Digestive Disease Center, Beijing Key Laboratory for Precancerous Lesion of Digestive Disease, Beijing, China
| |
Collapse
|
14
|
Glycyrrhizic Acid Alleviates Lipopolysaccharide (LPS)-Induced Acute Lung Injury by Regulating Angiotensin-Converting Enzyme-2 (ACE2) and Caveolin-1 Signaling Pathway. Inflammation 2021; 45:253-266. [PMID: 34427852 DOI: 10.1007/s10753-021-01542-8] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2020] [Revised: 08/08/2021] [Accepted: 08/10/2021] [Indexed: 10/20/2022]
Abstract
Acute lung injury (ALI) is mainly caused by severe infection, shock, trauma, and burn, which causes the extensive release of inflammatory factors and other mediators. As a major bioactive constituent of traditional Chinese herb licorice, glycyrrhizic acid (GA) plays an important effect on inflammatory regulation. Nevertheless, the exact mechanism of this effect remains unclear. The present study aims to explore the potential protective effect of GA on LPS-induced ALI. Our results showed that GA significantly attenuated LPS-induced ALI and decreased the production of inflammatory factors, including IL-1β, MCP-1, COX2, HMGB1, and adhesion molecules, such as E-selectin, VCAM-1, and modulated expression of angiotensin-converting enzyme 2 (ACE2). Moreover, treatment of ACE2 inhibitor (MLN-4760) reversed the effects of GA on the secretion of pro-inflammatory factors in ALI. Additionally, GA exerts its protective effect by regulating the ACE2 and caveolin-1/NF-κB signaling pathway. In conclusion, this study showed that GA alleviated LPS-induced ALI by upregulating ACE2 and inhibiting the caveolin-1/NF-κB signaling pathway.
Collapse
|
15
|
Yang C, He B, Dai W, Zhang H, Zheng Y, Wang X, Zhang Q. The role of caveolin-1 in the biofate and efficacy of anti-tumor drugs and their nano-drug delivery systems. Acta Pharm Sin B 2021; 11:961-977. [PMID: 33996409 PMCID: PMC8105775 DOI: 10.1016/j.apsb.2020.11.020] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2020] [Revised: 07/24/2020] [Accepted: 08/07/2020] [Indexed: 12/13/2022] Open
Abstract
As one of the most important components of caveolae, caveolin-1 is involved in caveolae-mediated endocytosis and transcytosis pathways, and also plays a role in regulating the cell membrane cholesterol homeostasis and mediating signal transduction. In recent years, the relationship between the expression level of caveolin-1 in the tumor microenvironment and the prognostic effect of tumor treatment and drug treatment resistance has also been widely explored. In addition, the interplay between caveolin-1 and nano-drugs is bidirectional. Caveolin-1 could determine the intracellular biofate of specific nano-drugs, preventing from lysosomal degradation, and facilitate them penetrate into deeper site of tumors by transcytosis; while some nanocarriers could also affect caveolin-1 levels in tumor cells, thereby changing certain biophysical function of cells. This article reviews the role of caveolin-1 in tumor prognosis, chemotherapeutic drug resistance, antibody drug sensitivity, and nano-drug delivery, providing a reference for the further application of caveolin-1 in nano-drug delivery systems.
Collapse
Key Words
- 5-FU, 5-fluorouracil
- ADC, antibody drug conjugates
- BBB, blood–brain barrier
- Biofate
- CAFs, cancer-associated fibroblasts
- CPT, camptothecin
- CSD, caveolin scaffolding domain
- CTB, cholera toxins B
- Cancer
- Caveolin-1
- Drug resistance
- ECM, extracellular matrix
- EGF, epidermal growth factor
- EGFR, epidermal growth factor receptor
- ER, endoplasmic reticulum
- ERK, extracellular regulated protein kinases
- FGF2, fibroblast growth factor 2
- GGT, γ-glutamyl transpeptidase
- GPI, glycosylphosphatidylinositol
- HER2, human epidermal growth factor receptor 2
- HMG-CoA, 3-hydroxy-3-methylglutaryl-coenzyme A
- HSA, human serum albumin
- IBC, infiltrating breast cancer
- IR, insulin receptor
- MAPK, mitogen-activated protein kinase
- MDR, multidrug resistance
- MSV, multistage nanovectors
- NPs, nanoparticles
- Nano-drug delivery systems
- PC, prostate cancer
- PDGF, platelet-derived growth factor
- PFS, progression free survival
- ROS, reactive oxygen species
- SCLC, small cell lung cancer
- SV40, simian virus 40
- Transcytosis
- cell SMA, styrene maleic acid
Collapse
|
16
|
Li Q, Wang C, Dong W, Su Y, Ma Z. WTAP facilitates progression of endometrial cancer via CAV-1/NF-κB axis. Cell Biol Int 2021; 45:1269-1277. [PMID: 33559954 DOI: 10.1002/cbin.11570] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2020] [Revised: 12/26/2020] [Accepted: 02/07/2021] [Indexed: 11/10/2022]
Abstract
The N6 -methyladenosine (m6 A) modification is one of the most prevalent methylations in eukaryotic messenger RNA (mRNA), and it is essential for the development of many important biological processes such as multiple types of tumors. One of the most important enzymes catalyzing generation of m6 A on mRNA is Wilms' tumor 1-associating protein (WTAP); however, the potential role of WTAP in endometrial cancer (EC) still remains unknown. Here, we investigated WTAP expression level in cancer tissue and paracancerous tissue from an EC patient. Subsequently, WTAP was knocked down by small interfering RNA in EC cell line of Ishikawa and HEC-1A, respectively. Cell proliferation, migration, and invasion were studied. The expression of caveolin-1 (CAV-1) was detected by quantitative polymerase chain reaction (qPCR). The enrichments of m6 A and METTL3 on CAV-1 were detected using RNA immunoprecipitation-qPCR. The activity of nuclear factor-κB (NF-κB) was studied using Western blot. We observed that WTAP was dramatically upregulated in the cancer tissue, and there was an enhancement in cell proliferation, migration, and invasion and a decrease in EC apoptosis in vivo and in vitro, which indicated higher tumor malignancy and worse survival outcome. After WTAP was knocked down in EC cells, CAV-1 was significantly upregulated and the enrichments of m6 A and METTL3 at 3'-untranslated region (UTR) region of CAV-1 were decreased. Moreover, the activity of NF-κB signaling pathway was inhibited by its regulator CAV-1. Taken together, we concluded that WTAP could methylate 3'-UTR of CAV-1 and downregulate CAV-1 expression to activate NF-κB signaling pathway in EC, which promoted EC progression.
Collapse
Affiliation(s)
- Qin Li
- Department of Clinical Laboratory, Yantai Yuhuangding Hospital, Yantai, Shandong, China
| | - Chenyu Wang
- Department of Clinical Laboratory, Yantai Yuhuangding Hospital, Yantai, Shandong, China
| | - Wei Dong
- Department of Radiation Oncology, Yantai Yuhuangding Hospital, Yantai, Shandong, China
| | - Yi Su
- Department of Radiation Oncology, Yantai Yuhuangding Hospital, Yantai, Shandong, China
| | - Zhao Ma
- Department of Radiation Oncology, Yantai Yuhuangding Hospital, Yantai, Shandong, China
| |
Collapse
|
17
|
Xiong J, Guo G, Guo L, Wang Z, Chen Z, Nan Y, Cao Y, Li R, Yang X, Dong J, Jin X, Yang W, Huang Q. Amlexanox Enhances Temozolomide-Induced Antitumor Effects in Human Glioblastoma Cells by Inhibiting IKBKE and the Akt-mTOR Signaling Pathway. ACS OMEGA 2021; 6:4289-4299. [PMID: 33644550 PMCID: PMC7906592 DOI: 10.1021/acsomega.0c05399] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/08/2020] [Accepted: 01/04/2021] [Indexed: 05/05/2023]
Abstract
Temozolomide (TMZ), as the first-line chemotherapeutic agent for the treatment of glioblastoma multiforme (GBM), often fails to improve the prognosis of GBM patients due to the quick development of resistance. The need for more effective management of GBM is urgent. The aim of this study is to evaluate the efficacy of combined therapy with TMZ and amlexanox, a selective inhibitor of IKBKE, for GBM. We found that the combined treatment resulted in significant induction of cellular apoptosis and the inhibition of cell viability, migration, and invasion in primary glioma cells and in the human glioma cell line, U87 MG. As expected, TMZ enhanced the expression of p-AMPK and amlexanox led to the reduction of IKBKE, with no impact on p-AMPK. Furthermore, we demonstrated that compared to other groups treated with each component alone, TMZ combined with amlexanox effectively reversed the TMZ-induced activation of Akt and inhibited the phosphorylation of mTOR. In addition, the combination treatment also clearly reduced in vivo tumor volume and prolonged median survival time in the xenograft mouse model. These results suggest that amlexanox sensitized the primary glioma cells and U87 MG cells to TMZ at least partially through the suppression of IKBKE activation and the attenuation of TMZ-induced Akt activation. Overall, combined treatment with TMZ and amlexanox may provide a promising possibility for improving the prognosis of glioblastoma patients in clinical practice.
Collapse
Affiliation(s)
- Jinbiao Xiong
- Department
of Neurosurgery, Tianjin Medical University
General Hospital, Tianjin 300052, China
- Key
Laboratory of Post-Trauma Neuro-Repair and Regeneration in Central
Nervous System, Ministry of Education and
Tianjin City, Tianjin 300052, China
| | - Gaochao Guo
- Department
of Neurosurgery, Tianjin Medical University
General Hospital, Tianjin 300052, China
- Key
Laboratory of Post-Trauma Neuro-Repair and Regeneration in Central
Nervous System, Ministry of Education and
Tianjin City, Tianjin 300052, China
- Tianjin
Key Laboratory of Injuries, Variations and
Regeneration of Nervous System, Tianjin 300052, China
| | - Lianmei Guo
- Department
of Neurosurgery, Tianjin Medical University
General Hospital, Tianjin 300052, China
- Key
Laboratory of Post-Trauma Neuro-Repair and Regeneration in Central
Nervous System, Ministry of Education and
Tianjin City, Tianjin 300052, China
- Tianjin
Key Laboratory of Injuries, Variations and
Regeneration of Nervous System, Tianjin 300052, China
| | - Zengguang Wang
- Department
of Neurosurgery, Tianjin Medical University
General Hospital, Tianjin 300052, China
- Key
Laboratory of Post-Trauma Neuro-Repair and Regeneration in Central
Nervous System, Ministry of Education and
Tianjin City, Tianjin 300052, China
- Tianjin
Key Laboratory of Injuries, Variations and
Regeneration of Nervous System, Tianjin 300052, China
| | - Zhijuan Chen
- Department
of Neurosurgery, Tianjin Medical University
General Hospital, Tianjin 300052, China
- Key
Laboratory of Post-Trauma Neuro-Repair and Regeneration in Central
Nervous System, Ministry of Education and
Tianjin City, Tianjin 300052, China
- Tianjin
Key Laboratory of Injuries, Variations and
Regeneration of Nervous System, Tianjin 300052, China
| | - Yang Nan
- Department
of Neurosurgery, Tianjin Medical University
General Hospital, Tianjin 300052, China
- Key
Laboratory of Post-Trauma Neuro-Repair and Regeneration in Central
Nervous System, Ministry of Education and
Tianjin City, Tianjin 300052, China
- Tianjin
Key Laboratory of Injuries, Variations and
Regeneration of Nervous System, Tianjin 300052, China
| | - Yiyao Cao
- Department
of Neurosurgery, Tianjin Medical University
General Hospital, Tianjin 300052, China
- Key
Laboratory of Post-Trauma Neuro-Repair and Regeneration in Central
Nervous System, Ministry of Education and
Tianjin City, Tianjin 300052, China
| | - Ruilong Li
- Department
of Neurosurgery, Tianjin Medical University
General Hospital, Tianjin 300052, China
- Key
Laboratory of Post-Trauma Neuro-Repair and Regeneration in Central
Nervous System, Ministry of Education and
Tianjin City, Tianjin 300052, China
- Tianjin
Key Laboratory of Injuries, Variations and
Regeneration of Nervous System, Tianjin 300052, China
| | - Xuejun Yang
- Department
of Neurosurgery, Tianjin Medical University
General Hospital, Tianjin 300052, China
- Key
Laboratory of Post-Trauma Neuro-Repair and Regeneration in Central
Nervous System, Ministry of Education and
Tianjin City, Tianjin 300052, China
- Tianjin
Key Laboratory of Injuries, Variations and
Regeneration of Nervous System, Tianjin 300052, China
| | - Jun Dong
- Department
of Neurosurgery, The Second Affiliated Hospital
of Soochow University, Suzhou 215004, China
| | - Xun Jin
- Tianjin
Medical University Cancer Institute and Hospital, Tianjin 300052, China
- National
Clinical Research Center for Cancer, Tianjin 300052, China
- Key
Laboratory of Cancer Prevention and Therapy, Tianjin 300052, China
- Tianjin’s
Clinical Research Center for Cancer, Tianjin 300052, China
| | - Weidong Yang
- Department
of Neurosurgery, Tianjin Medical University
General Hospital, Tianjin 300052, China
- Key
Laboratory of Post-Trauma Neuro-Repair and Regeneration in Central
Nervous System, Ministry of Education and
Tianjin City, Tianjin 300052, China
- . Tel: (+86)13820763396
| | - Qiang Huang
- Department
of Neurosurgery, Tianjin Medical University
General Hospital, Tianjin 300052, China
- Key
Laboratory of Post-Trauma Neuro-Repair and Regeneration in Central
Nervous System, Ministry of Education and
Tianjin City, Tianjin 300052, China
- Tianjin
Key Laboratory of Injuries, Variations and
Regeneration of Nervous System, Tianjin 300052, China
- . Tel: (+86)13820689221
| |
Collapse
|
18
|
Ma H, Zhang Z, Wang Y, Shang F, Du B, Wang Y, Cheng Z. Caveolin-1 regulates the expression of miR-183 and inhibits the invasion and migration of invasive pituitary adenomas by affecting early growth response 1 (EGR1)/Krueppel-like factor 5 (KLF5) interaction. Gland Surg 2021; 9:2144-2154. [PMID: 33447565 DOI: 10.21037/gs-20-823] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Background This study aims to investigate the mechanism through which Caveolin-1 (CAV-1) regulates the expression of micro ribonucleic acid (miR)-183 in invasive pituitary adenoma (IPA) tissues and GH3 cells, and explore the effects of CAV-1 and miR-183 on the invasion and migration ability of GH3 cells. Methods Western blotting was used to detect the expression level of CAV-1, early growth response 1 (EGR1) and Krueppel-like factor 5 (KLF5). Quantitative real-time polymerase chain reaction (qRT-PCR) was performed to detect the expression of miR-183. The mechanisms of interaction between CAV-1, EGR1, and KLF5 were studied by immunoprecipitation experiments. Transwell and cell scratch tests were used to determine the invasion and migration ability of GH3 cells. The dual-luciferase reporter gene experiment was used to detect the effects of EGR1 and KLF5 on miR-183 luciferase activity and verify the targeting relationship between miR-183 and ezrin. Results The expression of CAV-1 was up-regulated. However, following the knockdown of CAV-1, the invasion and migration ability of GH3 cells was significantly inhibited (P<0.05). The expression of miR-183 was down-regulated, but the expression level of miR-183 was markedly increased following the knockdown of CAV-1 (P<0.05). The knockdown of CAV-1 inhibited the nuclear ectopic of the EGR1 protein in GH3 cells. At the same time, the interaction between EGR1 and KLF5 in GH3 cells was significantly inhibited (P<0.05). The luciferase activity of miR-183 increased significantly after overexpression of KLF5 while overexpression of EGR1 and KLF5 had no significant effect on intracellular luciferase activity. Overexpression of miR-183 markedly inhibited the luciferase activity of wild-type EZR and the expression of the EZR protein in GH3 cells. Furthermore, the overexpression of miR-183 or the inhibition of EZR can reduce the invasion and migration ability of GH3 cells. The simultaneous overexpression or inhibition of miR-183 and EZR expression has no obvious effect on the invasion and migration ability of GH3 cells. Conclusions CAV-1 up-regulates the expression of miR-183 by inhibiting the nuclear ectopic of EGR1 and the interaction between EGR1 and KLF5 in GH3 cells. Also, miR-183 negatively regulates the expression of EZR and inhibits the invasion and migration of GH3 cells.
Collapse
Affiliation(s)
- Huan Ma
- Department of Neurosurgery, Xinxiang Central Hospital, Xinxiang, China
| | | | - Yang Wang
- Department of Neurosurgery, Xinxiang Central Hospital, Xinxiang, China
| | - Fei Shang
- Department of Neurosurgery, Xinxiang Central Hospital, Xinxiang, China
| | - Baoshun Du
- Department of Neurosurgery, Xinxiang Central Hospital, Xinxiang, China
| | - Yungang Wang
- Department of Neurosurgery, Xinxiang Central Hospital, Xinxiang, China
| | - Zhenguo Cheng
- Department of Neurosurgery, Xinxiang Central Hospital, Xinxiang, China
| |
Collapse
|
19
|
Zakaria S, Nawaya R, Abdel-Hamid NM, Eldomany RA, El-Shishtawy MM. Targeting the HIF-1α/Cav-1 Pathway with a Chicory Extract/Daidzein Combination Plays a Potential Role in Retarding Hepatocellular Carcinoma. Curr Cancer Drug Targets 2021; 21:881-896. [PMID: 34382525 DOI: 10.2174/1568009621666210811121120] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2021] [Revised: 05/17/2021] [Accepted: 05/17/2021] [Indexed: 11/22/2022]
Abstract
BACKGROUND Hepatocellular carcinoma (HCC) is one of the most rapidly growing solid cancers, that is characterized by hypoxia. Hypoxia-inducible factor-1α (HIF-1α) is a transcription factor that regulates tumor proliferation and metastasis. It induces caveolin-1 (Cav-1) expression, a glycoprotein found on the membrane surface, then Cav-1 triggers angiogenesis and metastasis in HCC. OBJECTIVE We hypothesize that targeting HIF-1α and consequently, Cav-1 using the antioxidant natural compound such as chicoric acid and a Cav-1 inhibitor daidzein (DAZ) could be a useful approach in the management of HCC. This study was conducted to investigate the possible therapeutic efficacy of standardized chicory leaf extract (SCLE) and DAZ via modulation of HIF-1α and Cav-1 in HCC rats. METHODS Diethyl nitrosamine (DENA) was used for HCC induction. After the induction period, four groups (10 rats for each) were treated with SCLE, DAZ, a combination of both, as well as sorafenib, all compared to the non-treated control. We assessed hepatic HIF-1α protein expression, Cav-1 gene expression, serum level of AFP, hepatic tissue content of VEGF, MMP-9, oxidative stress markers MDA and SOD. RESULTS DAZ, SCLE, and their combination, significantly down-regulated the expression of HIF-1α, Cav-1, and consequently dampened MMP-9, VEGF, hepatic content. It has been observed that the combination treatment showed a synergistic effect compared to either treatment alone. Importantly, the combination treatment exhibited a significantly more potent effect than sorafenib. CONCLUSION This study showed the potential role of the HIF-1α/Cav-1 pathway in HCC progression, moreover, SCLE and DAZ showed a potent efficacy in retarding HCC via modulation of this pathway.
Collapse
Affiliation(s)
- Sherin Zakaria
- Department of Pharmacology and Toxicology, Kafrelsheikh University, Kafrelsheikh, 33516, Egypt
| | - Reem Nawaya
- Department of Biochemistry, Kafrelsheikh University, Kafrelsheikh, 33516, Egypt
| | - Nabil M Abdel-Hamid
- Department of Biochemistry, Kafrelsheikh University, Kafrelsheikh, 33516, Egypt
| | - Ramadan A Eldomany
- Department of Microbiology and Immunology, Faculty of Pharmacy, Kafrelsheikh University, Kafrelsheikh, 33516, Egypt
| | - Mamdouh M El-Shishtawy
- Department of Biochemistry, Faculty of Pharmacy, Mansoura University, Mansoura, 35516, Egypt
| |
Collapse
|
20
|
Ye JH, Shi JJ, Yin X, Wu HY, Xu XY, Yao YZ, Zhang WJ. Elevated Expression of CAV1 is Associated with Unfavorable Prognosis of Patients with Breast Cancer Who Undergo Surgery and Neoadjuvant Chemotherapy. Cancer Manag Res 2020; 12:8887-8892. [PMID: 33061584 PMCID: PMC7519866 DOI: 10.2147/cmar.s264673] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2020] [Accepted: 09/08/2020] [Indexed: 12/24/2022] Open
Abstract
INTRODUCTION Neoadjuvant chemotherapy (NACT), which is standard treatment for locally advanced breast cancer, improves the resectability of patients with early breast cancer and reduces the extent of breast and axillary surgery. Caveolin-1 (CAV1) is implicated in human cancers, although its utility for cancer prognosis is unknown. Here, we investigated the expression of CAV1 in breast cancer tissues to evaluate its prognostic significance on patients with breast cancer administered NACT. METHODS CAV1 expression in 80 breast cancer tissue samples was evaluated using immunohistochemistry (IHC). The association between CAV1 levels and clinical factors was analyzed using the chi-square test and that between CAV1 and prognosis was evaluated using multivariate Cox regression and Kaplan-Meier analyses. RESULTS High levels of CAV1 were significantly associated with survival, and patients with overexpression of CAV1 had a poor prognosis. Adjusted multivariate Cox regression analyses revealed that a high level of CAV1 expression was an independent, significant prognostic factor for patients with breast cancer treated with NACT. DISCUSSION Overexpression of CAV1 in patients with breast cancer administered NACT was associated with shorter disease-free survival and overall survival. Therefore, high levels of CAV1 may serve as a prognostic biomarker for such patients.
Collapse
Affiliation(s)
- Jia-Hui Ye
- General Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, Jiangsu, People’s Republic of China
| | - Jia-Jun Shi
- General Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, Jiangsu, People’s Republic of China
| | - Xi Yin
- General Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, Jiangsu, People’s Republic of China
| | - Hong-Yan Wu
- Pathology Department, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, Jiangsu, People’s Republic of China
| | - Xin-Yun Xu
- Pathology Department, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, Jiangsu, People’s Republic of China
| | - Yong-Zhong Yao
- General Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, Jiangsu, People’s Republic of China
| | - Wei-Jie Zhang
- General Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, Jiangsu, People’s Republic of China
| |
Collapse
|
21
|
Targeting Notch signaling pathway as an effective strategy in overcoming drug resistance in ovarian cancer. Pathol Res Pract 2020; 216:153158. [PMID: 32829107 DOI: 10.1016/j.prp.2020.153158] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/25/2020] [Revised: 07/30/2020] [Accepted: 08/01/2020] [Indexed: 12/21/2022]
Abstract
Ovarian cancer, as one of the most common types of gynecological malignancies, has an increasing rate of incidence worldwide. Despite huge amounts of recent efforts in designing novel therapeutic strategies for complete removal of tumors and increasing overall survival of patients, chemotherapy is still the preferred therapy for ovarian cancer. However, chemotherapy is also challenged by development of drug resistance. Therefore, elucidating the underlying mechanisms of drug reissuance is an urgent need in ovarian cancer. Numerous studies have shown the implication of the Notch signaling pathway in the development of various human malignancies. Therefore, this study will provide a brief overview of the published evidence in support of Notch targeting in reverting multidrug resistance as a safer and novel approach for the improvement of ovarian cancer treatment.
Collapse
|
22
|
Dukaew N, Chairatvit K, Pitchakarn P, Imsumran A, Karinchai J, Tuntiwechapikul W, Wongnoppavich A. Inactivation of AKT/NF‑κB signaling by eurycomalactone decreases human NSCLC cell viability and improves the chemosensitivity to cisplatin. Oncol Rep 2020; 44:1441-1454. [PMID: 32945500 PMCID: PMC7448543 DOI: 10.3892/or.2020.7710] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2020] [Accepted: 07/03/2020] [Indexed: 01/03/2023] Open
Abstract
The high activation of protein kinase B (AKT)/nuclear factor-κB (NF-κB) signaling has often been associated with the induction of non-small cell lung cancer (NSCLC) cell survival and resistance to cisplatin, which is one of the most widely used chemotherapeutic drugs in the treatment of NSCLC. The inhibition of AKT/NF-κB can potentially be used as a molecular target for cancer therapy. Eurycomalactone (ECL), a quassinoid from Eurycoma longifolia Jack, has previously been revealed to exhibit strong cytotoxic activity against the human NSCLC A549 cell line, and can inhibit NF-κB activity in TNF-α-activated 293 cells stably transfected with an NF-κB luciferase reporter. The present study was the first to investigate whether ECL inhibits the activation of AKT/NF-κB signaling, induces apoptosis and enhances chemosensitivity to cisplatin in human NSCLC cells. The anticancer activity of ECL was evaluated in two NSCLC cell lines, A549 and Calu-1. ECL decreased the viability and colony formation ability of both cell lines by inducing cell cycle arrest and apoptosis through the activation of pro-apoptotic caspase-3 and poly (ADP-ribose) polymerase, as well as the reduction of anti-apoptotic proteins Bcl-xL and survivin. In addition, ECL treatment suppressed the levels of AKT (phospho Ser473) and NF-κB (phospho Ser536). Notably, ECL significantly enhanced cisplatin sensitivity in both assessed NSCLC cell lines. The combination treatment of cisplatin and ECL promoted cell apoptosis more effectively than cisplatin alone, as revealed by the increased cleaved caspase-3, but decreased Bcl-xL and survivin levels. Exposure to cisplatin alone induced the levels of phosphorylated-AKT and phosphorylated-NF-κB, whereas co-treatment with ECL inhibited the cisplatin-induced phosphorylation of AKT and NF-κB, leading to an increased sensitization effect on cisplatin-induced apoptosis. In conclusion, ECL exhibited an anticancer effect and sensitized NSCLC cells to cisplatin through the inactivation of AKT/NF-κB signaling. This finding provides a rationale for the combined use of chemotherapy drugs with ECL to improve their efficacy in NSCLC treatment.
Collapse
Affiliation(s)
- Nahathai Dukaew
- Graduate/PhD Degree Program in Biochemistry, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Kongthawat Chairatvit
- Department of Oral Biology, Faculty of Dentistry, Mahidol University, Bangkok 10400, Thailand
| | - Pornsiri Pitchakarn
- Department of Biochemistry, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Arisa Imsumran
- Department of Biochemistry, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Jirarat Karinchai
- Department of Biochemistry, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Wirote Tuntiwechapikul
- Department of Biochemistry, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Ariyaphong Wongnoppavich
- Department of Biochemistry, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand
| |
Collapse
|
23
|
Lin CJ, Lo UG, Hsieh JT. The regulatory pathways leading to stem-like cells underlie prostate cancer progression. Asian J Androl 2020; 21:233-240. [PMID: 30178777 PMCID: PMC6498735 DOI: 10.4103/aja.aja_72_18] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Prostate cancer (PCa) is the most common cause of malignancy in males and the third leading cause of cancer mortality in the United States. The standard care for primary PCa with local invasive disease mainly is surgery and radiation. For patients with distant metastases, androgen deprivation therapy (ADT) is a gold standard. Regardless of a favorable outcome of ADT, patients inevitably relapse to an end-stage castration-resistant prostate cancer (CRPC) leading to mortality. Therefore, revealing the mechanism and identifying cellular components driving aggressive PCa is critical for prognosis and therapeutic intervention. Cancer stem cell (CSC) phenotypes characterized as poor differentiation, cancer initiation with self-renewal capabilities, and therapeutic resistance are proposed to contribute to the onset of CRPC. In this review, we discuss the role of CSC in CRPC with the evidence of CSC phenotypes and the possible underlying mechanisms.
Collapse
Affiliation(s)
- Chun-Jung Lin
- Department of Urology, UT Southwestern Medical Center, Dallas, TX 75390, USA
| | - U-Ging Lo
- Department of Urology, UT Southwestern Medical Center, Dallas, TX 75390, USA
| | - Jer-Tsong Hsieh
- Department of Urology, UT Southwestern Medical Center, Dallas, TX 75390, USA
| |
Collapse
|
24
|
Wang X, Lu B, Dai C, Fu Y, Hao K, Zhao B, Chen Z, Fu L. Caveolin-1 Promotes Chemoresistance of Gastric Cancer Cells to Cisplatin by Activating WNT/β-Catenin Pathway. Front Oncol 2020; 10:46. [PMID: 32117718 PMCID: PMC7008851 DOI: 10.3389/fonc.2020.00046] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2019] [Accepted: 01/10/2020] [Indexed: 12/13/2022] Open
Abstract
Drug resistance is a major challenge for chemotherapy in treating human gastric cancer (GC), as the underlying molecular mechanism of chemoresistance in GC remains unknown. Caveolin-1 (Cav-1) is a scaffold protein of plasma membrane caveolae that acts as a tumor modulator by interacting with several cell signals. In this research, we showed that the survival rate of GC cells to cisplatin (CDDP) increased in the presence of Cav-1. Moreover, Cav-1 overexpression inhibited cisplatin-induced apoptosis and improved the survival rate of GC cells. Cav-1 overexpression and knock-down experiments indicated that Cav-1 expression stimulated wingless-type MMTV integration site (WNTs) pathway through the phosphorylation of LRP6 and dephosphorylation of β-catenin. Cav-1 was positively associated with the increase of WNT downstream target gene Met, which led to the activation of HER2 signaling. Moreover, our results demonstrated that the expression of Cav-1 and Met were positively associated with the resistance of GC cells to cisplatin. Collectively, Cav-1 enhances the cisplatin-resistance of GC cells by activating the WNT signaling pathway and Met-HER2 crosstalk. Understanding the role of Cav-1 in the chemoresistance of GC would help to develop novel therapies for a better treatment outcome of GC patients.
Collapse
Affiliation(s)
- Xi Wang
- Key Laboratory of Digestive Pathophysiology of Zhejiang Province, The First Affiliated Hospital of Zhejiang Chinese Medicine, Zhejiang Chinese Medical University, Hangzhou, China
| | - Bin Lu
- Key Laboratory of Digestive Pathophysiology of Zhejiang Province, The First Affiliated Hospital of Zhejiang Chinese Medicine, Zhejiang Chinese Medical University, Hangzhou, China.,Department of Gastroenterology, First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, China
| | - Chunyan Dai
- Key Laboratory of Digestive Pathophysiology of Zhejiang Province, The First Affiliated Hospital of Zhejiang Chinese Medicine, Zhejiang Chinese Medical University, Hangzhou, China
| | - Yufei Fu
- Key Laboratory of Digestive Pathophysiology of Zhejiang Province, The First Affiliated Hospital of Zhejiang Chinese Medicine, Zhejiang Chinese Medical University, Hangzhou, China
| | - Ke Hao
- Research Center of Blood Transfusion Medicine, Ministry of Education Key Laboratory of Laboratory Medicine, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, Hangzhou, China
| | - Bing Zhao
- Department of Anesthesiology, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Zhe Chen
- Key Laboratory of Digestive Pathophysiology of Zhejiang Province, The First Affiliated Hospital of Zhejiang Chinese Medicine, Zhejiang Chinese Medical University, Hangzhou, China
| | - Li Fu
- Guangdong Provincial Key Laboratory of Regional Immunity and Diseases, Department of Pharmacology and Shenzhen University International Cancer Center, Shenzhen University School of Medicine, Shenzhen, China
| |
Collapse
|
25
|
Lin CJ, Yun EJ, Lo UG, Tai YL, Deng S, Hernandez E, Dang A, Chen YA, Saha D, Mu P, Lin H, Li TK, Shen TL, Lai CH, Hsieh JT. The paracrine induction of prostate cancer progression by caveolin-1. Cell Death Dis 2019; 10:834. [PMID: 31685812 PMCID: PMC6828728 DOI: 10.1038/s41419-019-2066-3] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2019] [Revised: 10/02/2019] [Accepted: 10/16/2019] [Indexed: 12/21/2022]
Abstract
A subpopulation of cancer stem cells (CSCs) plays a critical role of cancer progression, recurrence, and therapeutic resistance. Many studies have indicated that castration-resistant prostate cancer (CRPC) is associated with stem cell phenotypes, which could further promote neuroendocrine transdifferentiation. Although only a small subset of genetically pre-programmed cells in each organ has stem cell capability, CSCs appear to be inducible among a heterogeneous cancer cell population. However, the inductive mechanism(s) leading to the emergence of these CSCs are not fully understood in CRPC. Tumor cells actively produce, release, and utilize exosomes to promote cancer development and metastasis, cancer immune evasion as well as chemotherapeutic resistance; the impact of tumor-derived exosomes (TDE) and its cargo on prostate cancer (PCa) development is still unclear. In this study, we demonstrate that the presence of Cav-1 in TDE acts as a potent driver to induce CSC phenotypes and epithelial-mesenchymal transition in PCa undergoing neuroendocrine differentiation through NFκB signaling pathway. Furthermore, Cav-1 in mCRPC-derived exosomes is capable of inducing radio- and chemo-resistance in recipient cells. Collectively, these data support Cav-1 as a critical driver for mCRPC progression.
Collapse
Affiliation(s)
- Chun-Jung Lin
- Department of Urology, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Eun-Jin Yun
- Department of Urology, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
- Division of Integrative Bioscience and Biotechnology, POSTECH, Pohang, 37673, Republic of Korea
| | - U-Ging Lo
- Department of Urology, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Yu-Ling Tai
- Department of Urology, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
- Department of Plant Pathology and Microbiology, National Taiwan University, Taipei, Taiwan
| | - Su Deng
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Elizabeth Hernandez
- Department of Urology, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Andrew Dang
- Department of Urology, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Yu-An Chen
- Department of Urology, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Debabrata Saha
- Department of Radiation Oncology, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Ping Mu
- Department of Plant Pathology and Microbiology, National Taiwan University, Taipei, Taiwan
| | - Ho Lin
- Department of Life Sciences, National Chung Hsing University, Taichung, Taiwan
| | - Tsai-Kun Li
- Department and Graduate Institute of Microbiology, National Taiwan University, Taipei, Taiwan
| | - Tang-Long Shen
- Department of Plant Pathology and Microbiology, National Taiwan University, Taipei, Taiwan
| | - Chih-Ho Lai
- Department of Microbiology and Immunology, Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Jer-Tsong Hsieh
- Department of Urology, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA.
- Department of Biotechnology, Kaohsiung Medical University, Kaohsiung, Taiwan.
| |
Collapse
|
26
|
Gong Y, Yang Y, Tian S, Chen H. Different Role of Caveolin-1 Gene in the Progression of Gynecological Tumors. Asian Pac J Cancer Prev 2019; 20:3259-3268. [PMID: 31759347 PMCID: PMC7062999 DOI: 10.31557/apjcp.2019.20.11.3259] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2019] [Indexed: 12/13/2022] Open
Abstract
Caveolin-1 (Cav-1), an integral membrane protein, is a principal component of caveolae and has been reported to play a promoting or inhibiting role in cancer progression. Gynecologic tumor is a group of tumors that affect the tissue and organs of the female reproductive system, especially cervical cancer. Cervical cancer, as one of the most common cancers, severely affects female health in developing countries in particular because of its high morbidity and mortality. This review summarizes some mechanisms of Cav-1 in the development and progression of gynecological tumors. The role of Cav-1 in tumorigenesis, including dysregulation of cell cycle, apoptosis and autophagy, adhesion, invasion, and metastasis, such as the formation of invadopodia and matrix metalloproteinase degradation are presented in detail. In addition, Cav-1 modulates autophagy and the formation of invadopodia and target regulated by miRNAs to affect tumor progress. Taken together, we find that, no matter Cav-1 expression in the tumor or stromal cells , Cav-1 has paradoxical role in different types of gynecological tumors in vivo or in vitro and even in the same tumor from the same organ.
Collapse
Affiliation(s)
- Yan Gong
- Department of Pathology, Zhongnan Hospital of Wuhan University, Wuhan, P. R. China
| | - Yuhan Yang
- Department of Pathology, School of Basic Medical Science, Wuhan University, Wuhan, P. R. China
| | - Sufang Tian
- Department of Pathology, Zhongnan Hospital of Wuhan University, Wuhan, P. R. China
| | - Honglei Chen
- Department of Pathology, Zhongnan Hospital of Wuhan University, Wuhan, P. R. China
| |
Collapse
|
27
|
Harrington BS, Annunziata CM. NF-κB Signaling in Ovarian Cancer. Cancers (Basel) 2019; 11:cancers11081182. [PMID: 31443240 PMCID: PMC6721592 DOI: 10.3390/cancers11081182] [Citation(s) in RCA: 81] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2019] [Revised: 08/05/2019] [Accepted: 08/12/2019] [Indexed: 12/13/2022] Open
Abstract
The NF-κB signaling pathway is a master and commander in ovarian cancer (OC) that promotes chemoresistance, cancer stem cell maintenance, metastasis and immune evasion. Many signaling pathways are dysregulated in OC and can activate NF-κB signaling through canonical or non-canonical pathways which have both overlapping and distinct roles in tumor progression. The activation of canonical NF-κB signaling has been well established for anti-apoptotic and immunomodulatory functions in response to the tumor microenvironment and the non-canonical pathway in cancer stem cell maintenance and tumor re-initiation. NF-κB activity in OC cells helps to create an immune-evasive environment and to attract infiltrating immune cells with tumor-promoting phenotypes, which in turn, drive constitutive NF-κB activation in OC cells to promote cell survival and metastasis. For these reasons, NF-κB is an attractive target in OC, but current strategies are limited and broad inhibition of this major signaling pathway in normal physiological and immunological functions may produce unwanted side effects. There are some promising pre-clinical outcomes from developing research to target and inhibit NF-κB only in the tumor-reinitiating cancer cell population of OC and concurrently activate canonical NF-κB signaling in immune cells to promote anti-tumor immunity.
Collapse
|
28
|
Liu J, Huang Y, Liu Y, Chen Y. Irisin Enhances Doxorubicin-Induced Cell Apoptosis in Pancreatic Cancer by Inhibiting the PI3K/AKT/NF-κB Pathway. Med Sci Monit 2019; 25:6085-6096. [PMID: 31412018 PMCID: PMC6705179 DOI: 10.12659/msm.917625] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
Background Irisin, a myokine released from skeletal muscle following exercise, has been shown to affect the proliferation of some cancer cells and chemosensitivity of anticancer drugs like doxorubicin (DOX). However, the effects of irisin on chemosensitivity in pancreatic cancer (PC) cells have not been studied. Material/Methods In this study, the effects of irisin co-treatment with DOX or gemcitabine (GEM) on MIA PaCa-2, BxPC-3 PC cells, and H9c2 cardiomyocytes were investigated. MTT (3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide) assay, flow cytometry, and TUNEL (TdT-mediated dUTP nick-end labeling) assays were conducted to evaluate cytotoxicity induced by DOX or GEM. Fluorescence microscopy and flow cytometry experiments were performed to assess the intracellular accumulation of DOX. Cellular levels of apoptosis-related protein expression and protein phosphorylation were determined by Western blot analyses. Results The results showed that irisin can increase the chemosensitivity of PC cells to DOX or GEM. The analyses of apoptosis indicated that irisin enhances DOX-induced cellular apoptosis by increasing the expression of cleaved PARP (poly ADP-ribose polymerase) and cleaved caspase-3, and reducing the expression of B cell lymphoma/lewkmia-2 (BCL-2) and B cell lymphoma-extra large (BCL-xL) in PC cells but not in H9c2 cells. Irisin attenuated serine/threonine kinase AKT (protein kinase B/PKB) phosphorylation and inhibited the activation of nuclear factor κB (NF-κB) signaling in PC cells. Conclusions Irisin can potentiate the cytotoxicity of doxorubicin in PC cells without increasing cardiotoxicity, possibly through inactivating the PI3K/AKT/NF-κB signaling pathway.
Collapse
Affiliation(s)
- Jiayu Liu
- Key Laboratory for Molecular Enzymology and Engineering of The Ministry of Education, Jilin University, Changchun, Jilin, China (mainland).,School of Life Sciences, Jilin University, Changchun, Jilin, China (mainland)
| | - Yibing Huang
- Key Laboratory for Molecular Enzymology and Engineering of The Ministry of Education, Jilin University, Changchun, Jilin, China (mainland).,School of Life Sciences, Jilin University, Changchun, Jilin, China (mainland)
| | - Yu Liu
- Department of Endocrinology, Sir Run Run Hospital, Nanjing Medical University, Nanjing, Jiangsu, China (mainland)
| | - Yuxin Chen
- Key Laboratory for Molecular Enzymology and Engineering of The Ministry of Education, Jilin University, Changchun, Jilin, China (mainland).,School of Life Sciences, Jilin University, Changchun, Jilin, China (mainland)
| |
Collapse
|
29
|
Liu B, Zhang J, Yang D. miR-96-5p promotes the proliferation and migration of ovarian cancer cells by suppressing Caveolae1. J Ovarian Res 2019; 12:57. [PMID: 31228941 PMCID: PMC6588920 DOI: 10.1186/s13048-019-0533-1] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2019] [Accepted: 06/12/2019] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Ovarian cancer (OC) is the second most common gynaecological malignancy. MicroRNAs (miRNAs) have been found to be aberrantly expressed in OC tissue and have been proposed as biomarkers and therapeutic targets for OC. RESULTS In this study, we found that miR-96-5p was up-regulated in OC tissues and OC cells compared to normal ovarian tissues and epithelial cell line. And, miR-96-5p was also up-regulated in the serum samples from OC patients compared to health participants. In addition, there was a positive correlation of miR-96-5p levels between OC tissues and serum samples. At the cellular level, overexpression of miR-96-5p promoted cell proliferation and migration in OC cells. Moreover, we further validated Caveolae1 (CAV1) as the direct target of miR-96-5p in OC cells through luciferase activity assays and western blot. CAV1 was obvious low expression in OC tissues. The overexpression of CAV1 abrogated the promotion of miR-96-5p on the OC cells proliferation and migration. Finally, we found that AKT signaling pathway was involved in this process. MiR-96-5p inhibited the phosphorylation of AKT and expression of down-stream proteins Cyclin D1 and P70 by targeting CAV1. CONCLUSIONS The above findings suggested that targeting miR-96-5p may be a promising strategy for OC treatment.
Collapse
Affiliation(s)
- Bo Liu
- Department of gynaecology & obstetrics, Jinan Maternal & Children Health Care Hospital, No 2 Jianguo Xiaojing Three road, Jinan, 250001, Shandong, People's Republic of China
| | - Jinglu Zhang
- Department of gynaecology & obstetrics, Jinan Maternal & Children Health Care Hospital, No 2 Jianguo Xiaojing Three road, Jinan, 250001, Shandong, People's Republic of China
| | - Dongxia Yang
- Department of gynaecology & obstetrics, Jinan Maternal & Children Health Care Hospital, No 2 Jianguo Xiaojing Three road, Jinan, 250001, Shandong, People's Republic of China.
| |
Collapse
|
30
|
Teng H, Wang D, Lu J, Zhou Y, Pang Y, Li Q. Novel insights into the evolution of the caveolin superfamily and mechanisms of antiapoptotic effects and cell proliferation in lamprey. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2019; 95:118-128. [PMID: 30742851 DOI: 10.1016/j.dci.2019.01.005] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/21/2018] [Revised: 01/10/2019] [Accepted: 01/10/2019] [Indexed: 06/09/2023]
Abstract
Caveolin-1 is the main structural and functional component of caveolin, and it is involved in the regulation of cholesterol transport, endocytosis, and signal transduction. Moreover, changes in caveolin-1 play an important role in tumorigenesis and inflammatory processes. Previous studies have demonstrated that human caveolin-1 is mainly located in the cell membrane and exhibits cell type- and stage-dependent functional differences during cancer development and inflammatory responses. However, the role of Lamprey-caveolin-like (L-caveolin-like) in lamprey remained unknown. Here, we demonstrated that L-caveolin-like performs anti-inflammation and oncogenic functions and the function of caveolin-1 diverged during vertebrate evolution. Moreover, the results reveal the mechanism underlying the antiapoptotic effects of L-caveolin-like. An L-caveolin-like gene from Lampetra japonica (L. japonica) was identified and characterized. L-Caveolin-like was primarily distributed in the leukocytes, intestines and supraneural bodies (Sp-bodies) immune organs as indicated by Q-PCR and immunohistochemistry assays. The mRNA and protein expression levels of L-caveolin exhibited consistent increases in expression at 2 and 72 h in adult tissues after exposure to lipopolysaccharide (LPS) and in leukocytes stimulated by Vibrio anguillarum (V. anguillarum), Staphylococcus aureus (S. aureus), and Poly I:C. Furthermore, the overexpression of pEGFP-N1-L-caveolin-like was associated with a distinct localization in mitochondria, with decreased cytochrome C (Cyt C) and mitochondrial Cyt C oxidase subunit I (CO I) expression. In addition, increased cellular ATP levels suggested that this protein prevented mitochondrial damage. The overexpression of pEGFP-N1-L-caveolin-like led to the altered expression of factors related to apoptosis, such as decreased Caspase-9, Caspase-3, p53, and Bax expression and increased Bcl-2 expression. In addition, the overexpression of pEGFP-N1-L-caveolin-like promoted cell proliferation associated with upregulated EGF, bFGF, and PDGFB expression. Together, these findings indicated that the L-caveolin-like protein from L. japonica induced the activation of antiapoptotic effects via the mitochondrial Cyt C-mediated Caspase-3 signaling pathway. Our analysis further suggests that L-caveolin-like is an oncogene protein product and anti-inflammatory molecule from lamprey that evolved early in vertebrate evolution.
Collapse
Affiliation(s)
- Hongming Teng
- College of Life Sciences, Liaoning Normal University, Dalian, 116081, China; Lamprey Research Center, Liaoning Normal University, Dalian, 116081, China.
| | - Dayu Wang
- College of Life Sciences, Liaoning Normal University, Dalian, 116081, China; Lamprey Research Center, Liaoning Normal University, Dalian, 116081, China.
| | - Jiali Lu
- College of Life Sciences, Liaoning Normal University, Dalian, 116081, China; Lamprey Research Center, Liaoning Normal University, Dalian, 116081, China.
| | - Ying Zhou
- College of Life Sciences, Liaoning Normal University, Dalian, 116081, China; Lamprey Research Center, Liaoning Normal University, Dalian, 116081, China.
| | - Yue Pang
- College of Life Sciences, Liaoning Normal University, Dalian, 116081, China; Lamprey Research Center, Liaoning Normal University, Dalian, 116081, China.
| | - Qingwei Li
- College of Life Sciences, Liaoning Normal University, Dalian, 116081, China; Lamprey Research Center, Liaoning Normal University, Dalian, 116081, China.
| |
Collapse
|
31
|
Glutathione Peroxidase 1 Promotes NSCLC Resistance to Cisplatin via ROS-Induced Activation of PI3K/AKT Pathway. BIOMED RESEARCH INTERNATIONAL 2019; 2019:7640547. [PMID: 31032363 PMCID: PMC6457285 DOI: 10.1155/2019/7640547] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/02/2018] [Revised: 02/07/2019] [Accepted: 02/19/2019] [Indexed: 01/15/2023]
Abstract
Purpose Reactive oxygen species (ROS)-induced cytotoxicity is an important mechanism by which cisplatin kills tumor cells. Glutathione peroxidase family (GPXs) is an important member of antioxidant system which metabolizes intracellular ROS and maintains homeostasis of cells. Altered expressions of GPXs enzymes, especially GPX1, have been described in a variety of human cancers. However, their functional roles in cisplatin-based chemoresistance in human malignancies including non-small cell lung cancer have never been explored. Methods A panel of NSCLC cell lines were selected for this study. GPX1 expression was detected using quantitative RT-PCR and Western blot. Cisplatin-induced cell killing was analyzed by CCK8 assay. Intracellular ROS levels were detected by fluorescence-based flow cytometry analysis. In vitro overexpression and knockdown of GPX1 expression were performed using GPX1 expression vector and siRNA approaches. Protein levels of PTEN, NF-κB, BCL2, Bax, and phosphorylated AKT were detected with western blot analysis using specific antibodies. Results GPX1 expression was upregulated in a subset of NSCLC cell lines resistant to cisplatin treatment. Expression vector-mediated forced overexpression of GPX1 significantly increased cisplatin resistance in NSCLC cell lines, whereas RNA inference-mediated downregulation of GPX1 could restore sensitivity to cisplatin. Overexpression of GPX1 significantly suppressed elevation of intracellular ROS and activation of AKT pathway when NSCLC cell lines were exposed to different concentrations of cisplatin. Activation of the AKT pathway inhibited proapoptotic cascade and subsequently led to cisplatin resistance in NSCLC cells. Inhibition of NF-κB by its chemical inhibitor BAY can significantly downregulate GPX1 expression and restore the cisplatin sensitivity of the cell lines resistant to cisplatin. Conclusions Our findings suggested that overexpression of GPX1 is a novel molecular mechanism for cisplatin-based chemoresistance in NSCLC. GPX1 overexpression blocks cisplatin-induced ROS intracellular accumulation, activates PI3K-AKT pathway by increased AKT phosphorylation, and further leads to cisplatin resistance in NSCLC cells. Inhibition of NF-κB signaling may be an alternative approach for restoring cisplatin sensitivity for NSCLC cells resistant to cisplatin-based chemotherapy.
Collapse
|
32
|
Sun Y, Xiaoyan H, Yun L, Chaoqun L, Jialing W, Liu Y, Yingqi Z, Peipei Y, Junjun P, Yuanming L. Identification of Key Candidate Genes and Pathways for Relationship between Ovarian Cancer and Diabetes Mellitus Using Bioinformatical Analysis. Asian Pac J Cancer Prev 2019; 20:145-155. [PMID: 30678426 PMCID: PMC6485580 DOI: 10.31557/apjcp.2019.20.1.145] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
Ovarian cancer is one of the three major gynecologic cancers in the world. The aim of this study is to find the
relationship between ovarian cancer and diabetes mellitus by using the genetic screening technique. By GEO database
query and related online tools of analysis, we analyzed 185 cases of ovarian cancer and 10 control samples from
GSE26712, and a total of 379 different genes were identified, including 104 up-regulated genes and 275 down-regulated
genes. The up-regulated genes were mainly enriched in biological processes, including cell adhesion, transcription of
nucleic acid and biosynthesis, and negative regulation of cell metabolism. The down-regulated genes were enriched in
cell proliferation, migration, angiogenesis and macromolecular metabolism. Protein-protein interaction was analyzed
by network diagram and module synthesis analysis. The top ten hub genes (CDC20, H2AFX, ENO1, ACTB, ISG15,
KAT2B, HNRNPD, YWHAE, GJA1 and CAV1) were identified, which play important roles in critical signaling
pathways that regulate the process of oxidation-reduction reaction and carboxylic acid metabolism. CTD analysis
showed that the hub genes were involved in 1,128 distinct diseases (bonferroni-corrected P<0.05). Further analysis by
drawing the Kaplan-Meier survival curve indicated that CDC20 and ISG15 were statistically significant (P<0.05). In
conclusion, glycometabolism was related to ovarian cancer and genes and proteins in glycometabolism could serve as
potential targets in ovarian cancer treatment.
Collapse
Affiliation(s)
- Yi Sun
- Department of Toxicology, Guilin Medical University School of Public Health, Guilin, China.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Zhong W, Huang Q, Zeng L, Hu Z, Tang X. Caveolin-1 and MLRs: A potential target for neuronal growth and neuroplasticity after ischemic stroke. Int J Med Sci 2019; 16:1492-1503. [PMID: 31673241 PMCID: PMC6818210 DOI: 10.7150/ijms.35158] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/23/2019] [Accepted: 09/03/2019] [Indexed: 12/22/2022] Open
Abstract
Ischemic stroke is a leading cause of morbidity and mortality worldwide. Thrombolytic therapy, the only established treatment to reduce the neurological deficits caused by ischemic stroke, is limited by time window and potential complications. Therefore, it is necessary to develop new therapeutic strategies to improve neuronal growth and neurological function following ischemic stroke. Membrane lipid rafts (MLRs) are crucial structures for neuron survival and growth signaling pathways. Caveolin-1 (Cav-1), the main scaffold protein present in MLRs, targets many neural growth proteins and promotes growth of neurons and dendrites. Targeting Cav-1 may be a promising therapeutic strategy to enhance neuroplasticity after cerebral ischemia. This review addresses the role of Cav-1 and MLRs in neuronal growth after ischemic stroke, with an emphasis on the mechanisms by which Cav-1/MLRs modulate neuroplasticity via related receptors, signaling pathways, and gene expression. We further discuss how Cav-1/MLRs may be exploited as a potential therapeutic target to restore neuroplasticity after ischemic stroke. Finally, several representative pharmacological agents known to enhance neuroplasticity are discussed in this review.
Collapse
Affiliation(s)
- Wei Zhong
- Department of Neurology, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, China
| | - Qianyi Huang
- Department of Neurology, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, China
| | - Liuwang Zeng
- Department of Neurology, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, China
| | - Zhiping Hu
- Department of Neurology, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, China
| | - Xiangqi Tang
- Department of Neurology, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, China
| |
Collapse
|
34
|
Granulin A Synergizes with Cisplatin to Inhibit the Growth of Human Hepatocellular Carcinoma. Int J Mol Sci 2018; 19:ijms19103060. [PMID: 30301274 PMCID: PMC6213591 DOI: 10.3390/ijms19103060] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2018] [Revised: 10/02/2018] [Accepted: 10/02/2018] [Indexed: 01/16/2023] Open
Abstract
Cisplatin is one of the most potent chemotherapy drugs widely used for cancer treatment. However, due to resistance and toxicity, the application of cisplatin for the treatment of hepatocellular carcinoma (HCC) is limited. Our previous study has shown that granulin A (GRN A), an anticancer peptide, is able to interact with enolase1 (ENO1) and inhibit the growth of HCC in vitro. In the present study, we studied the synergistic effect of the combination of cisplatin and GRN A for the inhibitory effect on HCC. An 3-(4,5-dimethylthiazol-2-yl)-5-(3-carboxymethoxyphenyl)-2-(4-sulfophenyl)-2H-tetrazolium (MTS) assay and Chou-Talalay approaches revealed that the combination of GRN A and cisplatin displayed potent synergistic effect. The colony formation and cell viability of HCC cells were inhibited significantly in cells treated with the combination of cisplatin and GRN A, compared with cells treated with cisplatin or GRN A alone. Overexpression of ENO1 diminished the synergistic effect of GRN A and cisplatin in HCC cells. The combination of the two drugs exhibited a more obvious inhibitory effect on cancer cell apoptosis, as analyzed by the cytometry flow, mitochondrial membrane potential (MMP) and western blot analysis. An in vivo study confirmed that the combined use of the two drugs displayed more potent antitumor activity compared to mice treated with cisplatin and GRN A alone; the inhibitory rate of tumor growth was 65.46% and 68.94%, respectively, in mice treated with GRN A and cisplatin. However, the inhibitory rate increased to 86.63% in mice treated with the combination of the two drugs. This study provides evidence that the combination of GRN A and cisplatin is able to sensitize the liver cancer to cisplatin, and that targeting ENO1 is a promising approach for enhancing the antitumor activity of cisplatin.
Collapse
|
35
|
Zheng Y, Dai Y, Liu W, Wang N, Cai Y, Wang S, Zhang F, Liu P, Chen Q, Wang Z. Astragaloside IV enhances taxol chemosensitivity of breast cancer via caveolin-1-targeting oxidant damage. J Cell Physiol 2018; 234:4277-4290. [PMID: 30146689 DOI: 10.1002/jcp.27196] [Citation(s) in RCA: 50] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2017] [Accepted: 07/17/2018] [Indexed: 01/09/2023]
Abstract
Accumulating evidence suggests that caveolin-1 (CAV-1) is a stress-related oncotarget and closely correlated to chemoresistance. Targeting CAV-1 might be a promising strategy to improve chemosensitivity for breast cancer treatment. Astragaloside IV (AS-IV), a bioactive compound purified from Astragalus membranaceus, has been shown to exhibit multiple bioactivities, including anticancer. However, the involved molecular targets are still ambiguous. In this study, we investigated the critical role of CAV-1 in mediating the chemosensitizing effects of AS-IV to Taxol on breast cancer. We found that AS-IV could enhance the chemosensitivity of Taxol with minimal direct cytotoxicity on breast cancer cell lines MCF-7 and MDA-MB-231, as well as the nontumor mammary epithelial cell line MCF-10A. AS-IV was further demonstrated to aggravate Taxol-induced apoptosis and G2/M checkpoint arrest. The phosphorylation of mitogen-activated protein kinase (MAPK) signaling extracellular signal-regulated kinase (ERK) and c-Jun N-terminal Kinase (JNK), except p38, was also abrogated by a synergistic interaction between AS-IV and Taxol. Moreover, AS-IV inhibited CAV-1 expression in a dose-dependent manner and reversed CAV-1 upregulation induced by Taxol administration. Mechanism study further demonstrated that AS-IV treatment triggered the eNOS/NO/ONOO- pathway via inhibiting CAV-1, which led to intense oxidant damage. CAV-1 overexpression abolished the chemosensitizing effects of AS-IV to Taxol by inhibiting oxidative stress. In vivo experiments further validated that AS-IV increased Taxol chemosensitivity on breast cancer via inhibiting CAV-1 expression, followed by activation of the eNOS/NO/ONOO- pathway. Taken together, our findings not only suggested the potential of AS-IV as a promising candidate to enhance chemosensitivity, but also highlighted the significance of CAV-1 as the target to reverse cancer drug resistance.
Collapse
Affiliation(s)
- Yifeng Zheng
- Guangzhou University of Chinese Medicine, Integrative Research Laboratory of Breast Cancer, The Research Centre of Integrative Medicine, Discipline of Integrated Chinese and Western Medicine & The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China.,Department of Mammary Disease, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, China.,Translational Research Laboratory of Chinese Medicine & Cancer Stress Signaling, College of Basic Medicine, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Yan Dai
- Department of Mammary Disease, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, China
| | - Weiping Liu
- Guangzhou University of Chinese Medicine, Integrative Research Laboratory of Breast Cancer, The Research Centre of Integrative Medicine, Discipline of Integrated Chinese and Western Medicine & The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Neng Wang
- Guangzhou University of Chinese Medicine, Integrative Research Laboratory of Breast Cancer, The Research Centre of Integrative Medicine, Discipline of Integrated Chinese and Western Medicine & The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China.,Translational Research Laboratory of Chinese Medicine & Cancer Stress Signaling, College of Basic Medicine, Guangzhou University of Chinese Medicine, Guangzhou, China.,Discipline of Integrated Chinese and Western Medicine, Post-Doctoral Research Center, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Youli Cai
- Guangzhou University of Chinese Medicine, Integrative Research Laboratory of Breast Cancer, The Research Centre of Integrative Medicine, Discipline of Integrated Chinese and Western Medicine & The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China.,Discipline of Integrated Chinese and Western Medicine, Post-Doctoral Research Center, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Shengqi Wang
- Guangzhou University of Chinese Medicine, Integrative Research Laboratory of Breast Cancer, The Research Centre of Integrative Medicine, Discipline of Integrated Chinese and Western Medicine & The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China.,Department of Mammary Disease, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, China.,Translational Research Laboratory of Chinese Medicine & Cancer Stress Signaling, College of Basic Medicine, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Fengxue Zhang
- Guangzhou University of Chinese Medicine, Integrative Research Laboratory of Breast Cancer, The Research Centre of Integrative Medicine, Discipline of Integrated Chinese and Western Medicine & The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China.,Translational Research Laboratory of Chinese Medicine & Cancer Stress Signaling, College of Basic Medicine, Guangzhou University of Chinese Medicine, Guangzhou, China.,Discipline of Integrated Chinese and Western Medicine, Post-Doctoral Research Center, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Pengxi Liu
- Department of Mammary Disease, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, China.,Translational Research Laboratory of Chinese Medicine & Cancer Stress Signaling, College of Basic Medicine, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Qianjun Chen
- Department of Mammary Disease, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, China
| | - Zhiyu Wang
- Guangzhou University of Chinese Medicine, Integrative Research Laboratory of Breast Cancer, The Research Centre of Integrative Medicine, Discipline of Integrated Chinese and Western Medicine & The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China.,Department of Mammary Disease, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, China.,Translational Research Laboratory of Chinese Medicine & Cancer Stress Signaling, College of Basic Medicine, Guangzhou University of Chinese Medicine, Guangzhou, China.,Discipline of Integrated Chinese and Western Medicine, Post-Doctoral Research Center, Guangzhou University of Chinese Medicine, Guangzhou, China
| |
Collapse
|
36
|
Zhang C, Wang M, Shi C, Shi F, Pei C. Long non-coding RNA Linc00312 modulates the sensitivity of ovarian cancer to cisplatin via the Bcl-2/Caspase-3 signaling pathway. Biosci Trends 2018; 12:309-316. [PMID: 29952351 DOI: 10.5582/bst.2018.01052] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
Chemotherapy is one of the main treatments for ovarian cancer (OC). Cisplatin combined with paclitaxel is a commonly used chemotherapy regimen. However, effective cancer therapy is hindered by a patient's resistance to cisplatin. The mechanism that potentially leads to that resistance is unclear. The current study examined the mechanism by which Linc00312 is involved in resistance to cisplatin in OC. Quantitative real-time PCR (RT-qPCR) was used to test for expression of Linc00312 in freshly frozen tissue samples of OC and in SKOV3 and SKOV3/DDP cells. In situ hybridization was performed to examine the distribution of Linc00312 expression in paraffin-embedded histological sections that were sensitive or resistant to cisplatin. The cell counting kit-8 assay was used to detect cell viability. Flow cytometry was used to measure cell apoptosis. RT-qPCR was performed to confirm changes in expression of MDR1, MRP1, Bcl-2, Bax, Caspase-3, and Caspase-9 mRNA. Levels of MDR1, Bcl-2, Bax, Caspase-3, and Caspase-9 protein were detected with Western blotting. Experiments indicated that the expression of Linc00312 decreased significantly in SKOV3/DDP cells compared to that in SKOV3 cells. Upregulation of Linc00312 can considerably increase the sensitivity of SKOV3/DDP cells to cisplatin, while down-regulation of Linc00312 has the exact opposite effect in SKOV3 cells. Linc00312 enhanced the sensitivity of SKOV3/DDP cells to cisplatin by promoting cell apoptosis via the Bcl-2/Caspase-3 signaling pathway. These findings suggest that Linc00312 may be a promising clinical strategy for the treatment of drug-resistant OC.
Collapse
Affiliation(s)
- Chuanqi Zhang
- Department of Gynecology and Obstetrics, Shengjing Hospital Affiliated of China Medical University
| | - Min Wang
- Department of Gynecology and Obstetrics, Shengjing Hospital Affiliated of China Medical University
| | - Cong Shi
- Department of Gynecology and Obstetrics, Shengjing Hospital Affiliated of China Medical University
| | - Fanli Shi
- Department of Gynecology and Obstetrics, Shengjing Hospital Affiliated of China Medical University
| | - Cheng Pei
- Department of Gynecology and Obstetrics, Shengjing Hospital Affiliated of China Medical University
| |
Collapse
|
37
|
Bai L, Wang A, Zhang Y, Xu X, Zhang X. Knockdown of MALAT1 enhances chemosensitivity of ovarian cancer cells to cisplatin through inhibiting the Notch1 signaling pathway. Exp Cell Res 2018; 366:161-171. [PMID: 29548748 DOI: 10.1016/j.yexcr.2018.03.014] [Citation(s) in RCA: 73] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2017] [Revised: 03/09/2018] [Accepted: 03/12/2018] [Indexed: 12/14/2022]
Abstract
Long non-coding RNAs (lncRNAs) are critical regulators in chemoresistance of various tumors including ovarian cancer. Metastasis-associated lung adenocarcinoma transcript 1 (MALAT1) has been reported to be upregulated and contributed to ovarian cancer tumorigenesis. The aim of this study was to explore the roles of MALAT1 and the underlying molecular regulatory mechanism in the chemoresistance of ovarian cancer cells. Our data demonstrated that MALAT1 and Notch1 mRNA were upregulated in ovarian cancer tissues, as well as cisplatin (CDDP)-resistant ovarian cancer cells. A positive correlation between MALAT1 and Notch1 mRNA expression was observed. MALAT1 knockdown significantly attenuated CDDP resistance, and enhanced CDDP-induced apoptosis in CDDP-resistant ovarian cancer cells. MALAT1 knockdown enhanced CDDP-induced apoptosis in vivo, as indicated by upregulation of Bax protein expression and downregulation of Bcl-2 protein expression. Additionally, MALAT1 knockdown inhibited the Notch1 pathway and ABCC1 expression in CDDP-resistant ovarian cancer cells. MALAT1 was demonstrated to interact with Notch1. Notch1 knockdown attenuated CDDP resistance, and downregulated the protein expression of ABCC1 in ovarian cancer cells. Taken together, our findings suggested that knockdown of MALAT-1 enhanced chemosensitivity of ovarian cancer cells to CDDP through inhibiting Notch1 signaling pathway.
Collapse
MESH Headings
- Adenocarcinoma, Mucinous/drug therapy
- Adenocarcinoma, Mucinous/metabolism
- Adenocarcinoma, Mucinous/secondary
- Animals
- Antineoplastic Agents/pharmacology
- Apoptosis
- Biomarkers, Tumor/genetics
- Biomarkers, Tumor/metabolism
- Cell Proliferation
- Cisplatin/pharmacology
- Cystadenocarcinoma, Serous/drug therapy
- Cystadenocarcinoma, Serous/metabolism
- Cystadenocarcinoma, Serous/secondary
- Drug Resistance, Neoplasm
- Endometrial Neoplasms/drug therapy
- Endometrial Neoplasms/metabolism
- Endometrial Neoplasms/secondary
- Female
- Follow-Up Studies
- Humans
- Lymphatic Metastasis
- Mice
- Mice, Inbred BALB C
- Mice, Nude
- Middle Aged
- Ovarian Neoplasms/drug therapy
- Ovarian Neoplasms/metabolism
- Ovarian Neoplasms/pathology
- Prognosis
- RNA, Long Noncoding/genetics
- Receptor, Notch1/genetics
- Receptor, Notch1/metabolism
- Tumor Cells, Cultured
- Xenograft Model Antitumor Assays
Collapse
Affiliation(s)
- Lin Bai
- Department of Obstetrics and Gynecology, The First People's Hospital of Shangqiu, No. 292 Kaixuan Nan Road, Suiyang District, Shangqiu 476100, China.
| | - Aihua Wang
- Department of Obstetrics and Gynecology, The First People's Hospital of Shangqiu, No. 292 Kaixuan Nan Road, Suiyang District, Shangqiu 476100, China
| | - Yali Zhang
- Department of Obstetrics and Gynecology, The First People's Hospital of Shangqiu, No. 292 Kaixuan Nan Road, Suiyang District, Shangqiu 476100, China
| | - Xiaofeng Xu
- Department of Obstetrics and Gynecology, The First People's Hospital of Shangqiu, No. 292 Kaixuan Nan Road, Suiyang District, Shangqiu 476100, China
| | - Xiao Zhang
- Department of Obstetrics and Gynecology, The First People's Hospital of Shangqiu, No. 292 Kaixuan Nan Road, Suiyang District, Shangqiu 476100, China
| |
Collapse
|
38
|
Abstract
Resistance of solid tumors to chemo- and radiotherapy remains a major obstacle in anti-cancer treatment. Herein, the membrane protein caveolin-1 (CAV1) came into focus as it is highly expressed in many tumors and high CAV1 levels were correlated with tumor progression, invasion and metastasis, and thus a worse clinical outcome. Increasing evidence further indicates that the heterogeneous tumor microenvironment, also known as the tumor stroma, contributes to therapy resistance resulting in poor clinical outcome. Again, CAV1 seems to play an important role in modulating tumor host interactions by promoting tumor growth, metastasis, therapy resistance and cell survival. However, the mechanisms driving stroma-mediated tumor growth and radiation resistance remain to be clarified. Understanding these interactions and thus, targeting CAV1 may offer a novel strategy for preventing cancer therapy resistance and improving clinical outcomes. In this review, we will summarize the resistance-promoting effects of CAV1 in tumors, and emphasize its role in the tumor-stroma communication as well as the resulting malignant phenotype of epithelial tumors.
Collapse
Affiliation(s)
- Julia Ketteler
- Institute for Cell Biology (Cancer Research), University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Diana Klein
- Institute for Cell Biology (Cancer Research), University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| |
Collapse
|
39
|
AP-2α reverses vincristine-induced multidrug resistance of SGC7901 gastric cancer cells by inhibiting the Notch pathway. Apoptosis 2018; 22:933-941. [PMID: 28439677 DOI: 10.1007/s10495-017-1379-x] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Multidrug resistance (MDR) remains a major clinical obstacle in the treatment of gastric cancer (GC) since it causes tumor recurrence and metastasis. The transcription factor activator protein-2α (AP-2α) has been implicated in drug-resistance in breast cancer; however, its effects on MDR of gastric cancer are far from understood. In this study, we aimed to explore the effects of AP-2α on the MDR in gastric cancer cells selected by vincristine (VCR). Decreased AP-2α levels were markedly detected by RT-PCR and Western blot in gastric cancer cell lines (BGC-823, SGC-7901, AGS, MKN-45) compared with that in the gastric epithelial cell line (GES-1). Furthermore, we found that the expression of AP-2α in SGC7901/VCR or SGC7901/adriamycin (ADR) cells was lower than in SGC7901 cells. Thus, a vector overexpressing AP-2α was constructed and used to perform AP-2α gain-of-function studies in SGC7901/VCR cells. The decreased IC50 values of the anti-cancer drugs in sensitive and resistant cells after transfect with pcDNA3.1/AP-2α were determined in SGC7901/VCR cells by MTT assay. Moreover, flow cytometry analysis indicated that overexpressed AP-2α induced cell cycle arrest in the G0/G1 phase and promoted cell apoptosis of VCR-selected SGC7901/VCR cells. RT-PCR and Western blot demonstrated that overexpressed AP-2α can significantly induce the down-regulation of Notch1, Hes-1, P-gp and MRP1 in SGC7901/VCR cells. Similar effects can be observed when Numb (Notch inhibitor) was introduced. In addition, the intracellular ADR accumulation was markedly detected in AP-2α overexpressed or Numb cells. In conclusion, our results indicate that AP-2α can reverse the MDR of gastric cancer cells, which may be realized by inhibiting the Notch signaling pathway.
Collapse
|
40
|
Xu L, Li H, Su L, Lu Q, Liu Z. MicroRNA-455 inhibits cell proliferation and invasion of epithelial ovarian cancer by directly targeting Notch1. Mol Med Rep 2017; 16:9777-9785. [DOI: 10.3892/mmr.2017.7790] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2017] [Accepted: 09/05/2017] [Indexed: 11/05/2022] Open
|
41
|
Qian Z, Zhou S, Zhou Z, Yang X, Que S, Lan J, Qiu Y, Lin Y. miR-146b-5p suppresses glioblastoma cell resistance to temozolomide through targeting TRAF6. Oncol Rep 2017; 38:2941-2950. [DOI: 10.3892/or.2017.5970] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2017] [Accepted: 08/08/2017] [Indexed: 11/05/2022] Open
|
42
|
Liver X receptors agonist GW3965 re-sensitizes gefitinib-resistant human non-small cell lung cancer cell to gefitinib treatment by inhibiting NF-κB in vitro. Oncotarget 2017; 8:15802-15814. [PMID: 28178657 PMCID: PMC5362524 DOI: 10.18632/oncotarget.15007] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2016] [Accepted: 01/06/2017] [Indexed: 01/16/2023] Open
Abstract
The recent research shows that the inhibition of the nuclear factor-κB (NF-κB) pathway is a promising therapeutic option for patients who progress after treatment with the novel mutant-selective EGFR-TKIs. For propose to find a nontoxic drug to reverse the acquired gefitinib resistance, we examined whether the Liver X Receptors agonist GW3965 affect gefitinib resistance of HCC827/GR-8-2 cells. Cell viability was measured by CCK-8 assay. Levels of NF-κB, p-AKT and caspases were detected by Western blot analysis. Immunocytochemical analysis was used to detect the expression of NF-κB, p-AKT intracellularly. Induction of apoptosis and cell cycle arrest was measured by Flow cytometry assay. And results revealed that more than 90% of HCC827/GR-8-2 cells lived upon treatment with gefitinib at a dose of 5μM for 48h. However, when under the combine treatment of GW3965 (5μM) & gefitinib(5μM), cell death rate was increased observably. Co-administration of gefitinib & GW3965 induced cell apoptosis and cell cycle arrest. Additionally, we observed a dose-dependent- down-regulation of NF-κB in HCC827/GR-8-2 cells treated with gefitinib & GW3965. GW3965 and gefitinib synergistically decreased cell proliferation and induced apoptosis by inhibiting NF-κB signaling pathway in gefitinib resistant cells. These findings support our hypothesis that GW3965 could act as a useful drug to reverse the gefitinib resistance.
Collapse
|
43
|
Ruan H, Li X, Yang H, Song Z, Tong J, Cao Q, Wang K, Xiao W, Xiao H, Chen X, Xu G, Bao L, Xiong Z, Yuan C, Liu L, Qu Y, Hu W, Gao Y, Ru Z, Chen K, Zhang X. Enhanced expression of caveolin-1 possesses diagnostic and prognostic value and promotes cell migration, invasion and sunitinib resistance in the clear cell renal cell carcinoma. Exp Cell Res 2017; 358:269-278. [PMID: 28684115 DOI: 10.1016/j.yexcr.2017.07.004] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2017] [Revised: 06/29/2017] [Accepted: 07/01/2017] [Indexed: 01/01/2023]
Abstract
Caveolin-1 (CAV1) has been identified to be up-regulated in many cancers, including clear cell renal cell carcinoma (ccRCC). However, its potential function is still unclear in ccRCC. In this study, we demonstrated that CAV1 was frequently overexpressed in renal cell carcinoma tissues and cells, and was significantly associated with various clinicopathological parameters. In addition, high CAV1 expression was associated with poor disease-free survival (DFS) rate and could serve as a useful diagnostic indicator in ccRCC patients with different clinicopathological stages. Functional experiments demonstrated that CAV1 knockdown inhibited cell migration and invasion, whereas overexpression of CAV1 promoted cell migration and invasion in ccRCC. Moreover, CAV1 expression was up-regulated in sunitinib-resistant renal cancer cell lines, and its overexpression promoted sunitinib resistance. In general, our results confirm that CAV1 plays an important role in the metastasis of kidney cancer and induces sunitinib resistance, so CAV1 function suppression may become a promising clinical treatment strategy during renal cell carcinoma metastasis and sunitinib resistance.
Collapse
Affiliation(s)
- HaiLong Ruan
- Department of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan 430022, Hubei Province, China
| | - Xiang Li
- Department of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan 430022, Hubei Province, China
| | - HongMei Yang
- Department of Pathogenic Biology, School of Basic Medicine, Huazhong University of Science and Technology, 13 Hangkong Road, Wuhan 430030, Hubei Province, China
| | - ZhengShuai Song
- Department of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan 430022, Hubei Province, China
| | - JunWei Tong
- Department of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan 430022, Hubei Province, China
| | - Qi Cao
- Department of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan 430022, Hubei Province, China
| | - KeShan Wang
- Department of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan 430022, Hubei Province, China
| | - Wen Xiao
- Department of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan 430022, Hubei Province, China
| | - HaiBin Xiao
- Department of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan 430022, Hubei Province, China
| | - XuanYu Chen
- Department of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan 430022, Hubei Province, China; Department of Urology, Zhejiang Provincial People's Hospital, Hangzhou 310014, China; Department of Neuroscience and Regenerative Medicine, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA
| | - GuangHua Xu
- Department of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan 430022, Hubei Province, China
| | - Lin Bao
- Department of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan 430022, Hubei Province, China
| | - ZhiYong Xiong
- Department of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan 430022, Hubei Province, China
| | - ChangFei Yuan
- Department of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan 430022, Hubei Province, China
| | - Lei Liu
- Department of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan 430022, Hubei Province, China
| | - Yan Qu
- Department of Pathogenic Biology, School of Basic Medicine, Huazhong University of Science and Technology, 13 Hangkong Road, Wuhan 430030, Hubei Province, China
| | - WenJun Hu
- Department of Pathogenic Biology, School of Basic Medicine, Huazhong University of Science and Technology, 13 Hangkong Road, Wuhan 430030, Hubei Province, China
| | - YaoYing Gao
- Department of Pathogenic Biology, School of Basic Medicine, Huazhong University of Science and Technology, 13 Hangkong Road, Wuhan 430030, Hubei Province, China
| | - ZeYuan Ru
- Department of Pathogenic Biology, School of Basic Medicine, Huazhong University of Science and Technology, 13 Hangkong Road, Wuhan 430030, Hubei Province, China
| | - Ke Chen
- Department of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan 430022, Hubei Province, China
| | - XiaoPing Zhang
- Department of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan 430022, Hubei Province, China.
| |
Collapse
|
44
|
Caveolin-1: An Oxidative Stress-Related Target for Cancer Prevention. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2017; 2017:7454031. [PMID: 28546853 PMCID: PMC5436035 DOI: 10.1155/2017/7454031] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/06/2016] [Revised: 01/23/2017] [Accepted: 03/07/2017] [Indexed: 01/19/2023]
Abstract
Aberrant oxidative metabolism is one of the hallmarks of cancer. Reactive species overproduction could promote carcinogenesis via inducing genetic mutations and activating oncogenic pathways, and thus, antioxidant therapy was considered as an important strategy for cancer prevention and treatment. Caveolin-1 (Cav-1), a constituent protein of caveolae, has been shown to mediate tumorigenesis and progression through oxidative stress modulation recently. Reactive species could modulate the expression, degradation, posttranslational modifications, and membrane trafficking of Cav-1, while Cav-1-targeted treatments could scavenge the reactive species. More importantly, emerging evidences have indicated that multiple antioxidants could exert antitumor activities in cancer cells and protective activities in normal cells by modulating the Cav-1 pathway. Altogether, these findings indicate that Cav-1 may be a promising oxidative stress-related target for cancer antioxidant prevention. Elucidating the underlying interaction mechanisms between oxidative stress and Cav-1 is helpful for enhancing the preventive effects of antioxidants on cancer, for improving clinical outcomes of antioxidant-related therapeutics in cancer patients, and for developing Cav-1 targeted drugs. Herein, we summarize the available evidence of the roles of Cav-1 and oxidative stress in tumorigenesis and development and shed novel light on designing strategies for cancer prevention or treatment by utilizing the interaction mode between Cav-1 and oxidative stress.
Collapse
|
45
|
Reversal effects of local anesthetics on P-glycoprotein-mediated cancer multidrug resistance. Anticancer Drugs 2017; 28:243-249. [DOI: 10.1097/cad.0000000000000455] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
|
46
|
Demirci NS, Dogan M, Erdem GU, Kacar S, Turhan T, Kilickap S, Cigirgan LC, Kayacetin E, Bozkaya Y, Zengin N. Is plasma caveolin-1 level a prognostic biomarker in metastatic pancreatic cancer? Saudi J Gastroenterol 2017; 23:183-189. [PMID: 28611342 PMCID: PMC5470378 DOI: 10.4103/sjg.sjg_483_16] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
BACKGROUND/AIMS To evaluate the prognostic significance of plasma caveolin (CAV)-1 and its association with survival and treatment response rates in metastatic pancreatic cancer (MPC). PATIENTS AND METHODS Plasma samples were prospectively collected from 41 patients with newly diagnosed MPC. Moreover, plasma samples were collected from 48 patients with chronic pancreatitis and 41 healthy individuals (control groups) for assessing Cav-1 levels. Plasma Cav-1 levels were evaluated at baseline and after three cycles of chemotherapy in the patients with MPC. RESULTS The median Cav-1 level was 13.8 ng/mL for the patients with MPC and 12.2 ng/mL for healthy individuals (P = 0.009). The Cav-1 cut-off level was calculated as 11.6 ng/mL by using the receiver operating characteristic curve. The median overall survival and progression-free survival rates were 5 and 2.4 months, respectively, for participants with a high basal plasma Cav-1 level; the corresponding values were 10.5 and 9.4 months for participants with a low plasma Cav-1 level (P = 0.011 and P= 0.003, respectively). Of the 41 patients with MPC, 23 completed at least three cycles of chemotherapy. The median Cav-1 level was 13 ng/mL for post-treatment MPC (r2: 0.917; P= 0.001). High basal plasma caveolin-1 level have continued to remain at high levels even after chemotherapy, showing a trend toward worse response rates (P = 0.086). CONCLUSION High basal plasma Cav-1 levels seem to be associated with poor survival and tend to yield worse therapeutic outcomes in patients with MPC. This study is the first to evaluate the prognostic significance of plasma Cav-1 levels as a prognostic factor in patients with MPC. However, larger prospective clinical trials are warranted.
Collapse
Affiliation(s)
- Nebi S. Demirci
- Department of Medical Oncology, Ankara Numune Training and Research Hospital, Ankara, Turkey,Address for correspondence: Dr. Nebi S. Demirci, Department of Medical Oncology, Ankara Numune Training and Research Hospital, Ankara, Turkey. E-mail:
| | - Mutlu Dogan
- Department of Medical Oncology, Ankara Numune Training and Research Hospital, Ankara, Turkey
| | - Gokmen U. Erdem
- Department of Medical Oncology, Ankara Numune Training and Research Hospital, Ankara, Turkey
| | - Sabite Kacar
- Department of Gastroenterology, Turkiye Yuksek Ihtisas Training and Research Hospital, Ankara, Turkey
| | - Turan Turhan
- Department of Biochemistry, Ankara Numune Training and Research Hospital, Ankara, Turkey
| | - Saadettin Kilickap
- Department of Medical Oncology, Hacettepe University Medical Faculty, Ankara, Turkey
| | - Lutfi C. Cigirgan
- Department of Biochemistry, Ankara Numune Training and Research Hospital, Ankara, Turkey
| | - Ertugrul Kayacetin
- Department of Gastroenterology, Turkiye Yuksek Ihtisas Training and Research Hospital, Ankara, Turkey
| | - Yakup Bozkaya
- Department of Medical Oncology, Ankara Numune Training and Research Hospital, Ankara, Turkey
| | - Nurullah Zengin
- Department of Medical Oncology, Ankara Numune Training and Research Hospital, Ankara, Turkey
| |
Collapse
|
47
|
Li L, Wang S, Zheng F, Wu W, Hann SS. Chinese herbal medicine Fuzheng Kang-Ai decoction sensitized the effect of gefitinib on inhibition of human lung cancer cells through inactivating PI3-K/Akt -mediated suppressing MUC1 expression. JOURNAL OF ETHNOPHARMACOLOGY 2016; 194:918-929. [PMID: 27989877 DOI: 10.1016/j.jep.2016.10.077] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/15/2016] [Revised: 10/17/2016] [Accepted: 10/24/2016] [Indexed: 06/06/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Chinese herbal medicine (CHM) Fuzheng Kang-Ai (FZKA for short) decoction has been used as adjuvant treatment strategies in lung cancer patients for decades. However, the molecular mechanism underlying the therapeutic potential especially in sensitizing the effect of EGFR-TKI gefitinib has not been well elucidated. MATERIALS AND METHODS Cell viability was detected by MTT assay. Cell cycle distribution was detected by flow cytometry. Western blot were used to examine phosphorylation and protein levels of Akt, p65, p50 and MUC1. The mRNA level of MUC1 was measured by qRT-PCR. Transient transfection experiments were used to overexpression of Akt, p65 and MUC1. Tumor xenograft and bioluminescent imaging experiments were carried out to confirm the in vitro findings. RESULTS Cell viability was inhibited by FZKA treatment and showed more significant when treated with FZKA and gefitinib in combine in lung cancer cells. FZKA induced the cell arrest at G0/G1 phase. Mechanistically, we showed that the phosphorylation of Akt, protein expressions of p65 and MUC1 were suppressed by FZKA and even more responses were observed in the FZKA and gefitinib combining. Overexpressed Akt overcame the effect of FZKA on p65 protein, and exogenously expressed p65 resisted the inhibitory effect of MUC1 protein expression by FZKA. On the contrary, while overexpressed MUC1 had no effect on p65 expression, it feedback increased phosphorylation of Akt, and more importantly, reversed the cell growth inhibition affected by FZKA. In line with the above, our results confirmed the synergistic effects of FZKA and gefitinib combination on tumor growth, the phosphorylation of Akt, and protein expression of p65 and MUC1 in vivo. CONCLUSION This study shows that FZKA decoction inhibits the growth of NSCLC cells through Akt-mediated inhibition of p65, followed by reducing the expression of MUC1. More importantly, there is a synergistic effect of FZKA decoction and gefitinib combination with greater suppression. The positive feedback regulatory loop of MUC1 to Akt signaling pathway further added the important role of MUC1 in mediating the overall responses of FZKA decoction in this process. The in vitro and in vivo study provides an additional and a novel mechanism by which the FZKA decoction enhances the growth inhibition of gefitinib in gefitinib-resistant NSCLC cells.
Collapse
Affiliation(s)
- Longmei Li
- Laboratory of Tumor Molecular Biology and Targeted Therapies of TCM, China; Department of Medical Oncology, Guangdong Provincial Hospital of Chinese Medicine, The Second Clinical Medical Collage, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong Province 510120, China
| | - SuMei Wang
- Department of Medical Oncology, Guangdong Provincial Hospital of Chinese Medicine, The Second Clinical Medical Collage, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong Province 510120, China
| | - Fang Zheng
- Laboratory of Tumor Molecular Biology and Targeted Therapies of TCM, China
| | - WanYin Wu
- Department of Medical Oncology, Guangdong Provincial Hospital of Chinese Medicine, The Second Clinical Medical Collage, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong Province 510120, China.
| | - Swei Sunny Hann
- Laboratory of Tumor Molecular Biology and Targeted Therapies of TCM, China; Department of Medical Oncology, Guangdong Provincial Hospital of Chinese Medicine, The Second Clinical Medical Collage, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong Province 510120, China.
| |
Collapse
|
48
|
Zhang F, Sun H, Zhang S, Yang X, Zhang G, Su T. Overexpression of PER3 Inhibits Self-Renewal Capability and Chemoresistance of Colorectal Cancer Stem-Like Cells via Inhibition of Notch and β-Catenin Signaling. Oncol Res 2016; 25:709-719. [PMID: 27983919 PMCID: PMC7841129 DOI: 10.3727/096504016x14772331883976] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
PER3, a circadian clock gene, plays an important role in colorectal cancer, but its action and underlying mechanism in colorectal cancer stem-like cells (CSCs) remain unclear. In this study, the colorectal CSCs were enriched in colorectal HCT-116 sphere-forming cells, expressing lower levels of stem cell markers CD133, CD44, LGR5, and SOX2 compared with HCT-116 cells. A drug-resistant strain from HCT-116 was established. Western blot and qRT-PCR analysis showed that PER3 was downregulated in colorectal CSCs and drug-resistant HCT-116. Overexpression of PER3 could strengthen 5-FU-induced inhibitory effects on colorectal CSCs, but knockdown of PER3 decreased its inhibition of colorectal CSCs. In addition, overexpression of PER3 in colorectal CSCs resulted in reduced colony formation efficiency in a soft agar medium and self-renewal efficiency. Inversely, knockdown of PER3 enhanced self-renewal of colorectal CSCs. Overexpression of PER3 decreased stemness markers and Notch1, Jagged1, β-catenin, c-Myc, and LGR5 in colorectal CSCs. When Notch or β-catenin signaling was inhibited, the chemoresistance and self-renewal capability of colorectal CSCs were decreased. It was confirmed that PER3 can reduce chemoresistance and self-renewal capability of colorectal CSCs via inhibition of Notch and β-catenin signaling. Our results reveal that PER3 plays a critical role in maintaining the stemness of colorectal CSCs and may be a promising target for elimination of CSCs.
Collapse
|
49
|
Zhang Z, Huang K, Gu C, Zhao L, Wang N, Wang X, Zhao D, Zhang C, Lu Y, Meng Y. Molecular Subtyping of Serous Ovarian Cancer Based on Multi-omics Data. Sci Rep 2016; 6:26001. [PMID: 27184229 PMCID: PMC4868982 DOI: 10.1038/srep26001] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2016] [Accepted: 04/25/2016] [Indexed: 01/22/2023] Open
Abstract
Classification of ovarian cancer by morphologic features has a limited effect on serous ovarian cancer (SOC) treatment and prognosis. Here, we proposed a new system for SOC subtyping based on the molecular categories from the Cancer Genome Atlas project. We analyzed the DNA methylation, protein, microRNA, and gene expression of 1203 samples from 599 serous ovarian cancer patients. These samples were divided into nine subtypes based on RNA-seq data, and each subtype was found to be associated with the activation and/or suppression of the following four biological processes: immunoactivity, hormone metabolic, mesenchymal development and the MAPK signaling pathway. We also identified four DNA methylation, two protein expression, six microRNA sequencing and four pathway subtypes. By integrating the subtyping results across different omics platforms, we found that most RNA-seq subtypes overlapped with one or two subtypes from other omics data. Our study sheds light on the molecular mechanisms of SOC and provides a new perspective for the more accurate stratification of its subtypes.
Collapse
Affiliation(s)
- Zhe Zhang
- Department of Gynecologic Oncology, Chinese PLA General Hospital, Beijing 100853, China
| | - Ke Huang
- Department of Gynecologic Oncology, Chinese PLA General Hospital, Beijing 100853, China
| | - Chenglei Gu
- Department of Gynecologic Oncology, Chinese PLA General Hospital, Beijing 100853, China
| | - Luyang Zhao
- Department of Gynecologic Oncology, Chinese PLA General Hospital, Beijing 100853, China
| | - Nan Wang
- Department of Gynecologic Oncology, Chinese PLA General Hospital, Beijing 100853, China
| | - Xiaolei Wang
- Beijing Institute of Health Service and Medical Information, Beijing 100850, China
| | - Dongsheng Zhao
- Beijing Institute of Health Service and Medical Information, Beijing 100850, China
| | - Chenggang Zhang
- Beijing Institute of Radiation Medicine, State Key Laboratory of Proteomics, Cognitive and Mental Health Research Center, Beijing 100850, China
| | - Yiming Lu
- Beijing Institute of Radiation Medicine, State Key Laboratory of Proteomics, Cognitive and Mental Health Research Center, Beijing 100850, China
| | - Yuanguang Meng
- Department of Gynecologic Oncology, Chinese PLA General Hospital, Beijing 100853, China
| |
Collapse
|
50
|
Zhou X, Teng L, Wang M. Distinct prognostic values of four-Notch-receptor mRNA expression in ovarian cancer. Tumour Biol 2015; 37:6979-85. [PMID: 26662955 DOI: 10.1007/s13277-015-4594-5] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2015] [Accepted: 12/03/2015] [Indexed: 12/18/2022] Open
Abstract
Notch signaling pathway includes ligands and Notch receptors, which are frequently deregulated in several human malignancies including ovarian cancer. Aberrant activation of Notch signaling has been linked to ovarian carcinogenesis and progression. In the current study, we used the "Kaplan-Meier plotter" (KM plotter) database, in which updated gene expression data and survival information from a total of 1306 ovarian cancer patients were used to access the prognostic value of four Notch receptors in ovarian cancer patients. Hazard ratio (HR), 95 % confidence intervals, and log-rank P were calculated. Notch1 messenger RNA (mRNA) high expression was not found to be correlated to overall survival (OS) for all ovarian cancer, as well as in serous and endometrioid cancer patients followed for 20 years. However, Notch1 mRNA high expression is significantly associated with worsen OS in TP53 wild-type ovarian cancer patients, while it is significantly associated with better OS in TP53 mutation-type ovarian cancer patients. Notch2 mRNA high expression was found to be significantly correlated to worsen OS for all ovarian cancer patients, as well as in grade II ovarian cancer patients. Notch3 mRNA high expression was found to be significantly correlated to better OS for all ovarian cancer patients, but not in serous cancer patients and endometrioid cancer patients. Notch4 mRNA high expression was not found to be significantly correlated to OS for all ovarian cancer patients, serous cancer patients, and endometrioid cancer patients. These results indicate that there are distinct prognostic values of four Notch receptors in ovarian cancer. This information will be useful for better understanding of the heterogeneity and complexity in the molecular biology of ovarian cancer and for developing tools to more accurately predict their prognosis. Based on our results, Notch1 could be a potential drug target of TP53 wild-type ovarian cancer and Notch2 could be a potential drug target of ovarian cancer.
Collapse
Affiliation(s)
- Xinling Zhou
- Department of Obstetrics and Gynecology, The Second People's Hospital of Liaocheng Affiliated to Taishan Medical College, No. 306 Jiankang Rd, Linqing, 252601, Shandong Province, People's Republic of China.
| | - Lingling Teng
- Department of Obstetrics and Gynecology, The Second People's Hospital of Liaocheng Affiliated to Taishan Medical College, No. 306 Jiankang Rd, Linqing, 252601, Shandong Province, People's Republic of China
| | - Min Wang
- Department of Pathology, The Second People's Hospital of Liaocheng Affiliated to Taishan Medical College, Linqing, 252601, Shandong Province, People's Republic of China
| |
Collapse
|