1
|
Naeem M, Ahmed K, Sultan A. Mutational and expression analysis of classical protein tyrosine phosphatase genes in pancreatic ductal adenocarcinoma. Comput Biol Med 2025; 193:110319. [PMID: 40403635 DOI: 10.1016/j.compbiomed.2025.110319] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2025] [Revised: 04/11/2025] [Accepted: 04/30/2025] [Indexed: 05/24/2025]
Abstract
BACKGROUND Pancreatic cancer is a highly lethal and aggressive malignancy with a minimal five-year survival rate (5 %) and a high mortality rate. The most common and fast-growing type of pancreatic cancer is PDAC, which constitutes 90 % of all cases. OBJECTIVE Numerous signaling pathways are disrupted in PDAC. We explored the mutational status and expression profiling of classical protein tyrosine phosphatase (PTP) genes, which are vital regulators of multiple significant signaling pathways. METHOD Through whole exome sequencing, we identified potentially pathogenic non-synonymous variants that were subsequently analyzed in-silico. To validate these findings, quantitative real-time PCR was performed on blood samples from PDAC patients to assess the expression of deleterious genes. RESULTS All the potential pathogenic variants were localized within the phosphatase domain 1, fibronectin type III domain, and the FERM domain of classical PTPs regions crucial for the proper functioning of the respective proteins. Among the analyzed genes, PTPN3, PTPN12, PTPRK, and PTPRZ1 were found statistically significant (p < 0.05), highlighting their potential as novel prognostic biomarkers and therapeutic targets for PDAC. CONCLUSION These findings hold particular relevance for the Pakistani population, offering valuable insights into the genetic landscape of this aggressive cancer.
Collapse
Affiliation(s)
- Maryam Naeem
- Cancer Genetics Lab, Department of Biochemistry, Quaid-i-Azam University, 45320, Islamabad, Pakistan
| | - Khursheed Ahmed
- Cancer Genetics Lab, Department of Biochemistry, Quaid-i-Azam University, 45320, Islamabad, Pakistan
| | - Aneesa Sultan
- Cancer Genetics Lab, Department of Biochemistry, Quaid-i-Azam University, 45320, Islamabad, Pakistan.
| |
Collapse
|
2
|
Wang JF, Wang MC, Jiang LL, Lin NM. The neuroscience in breast cancer: Current insights and clinical opportunities. Heliyon 2025; 11:e42293. [PMID: 39975839 PMCID: PMC11835589 DOI: 10.1016/j.heliyon.2025.e42293] [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: 08/06/2024] [Revised: 01/25/2025] [Accepted: 01/25/2025] [Indexed: 02/21/2025] Open
Abstract
The involvement of nerves in the development of breast cancer has emerged as a significant factor. Interaction between the nervous system and breast cancer can influence tumor initiation, growth, invasion, metastasis, reverse resistance to drugs, promote inflammation in tumors, and impair the immune system's ability to combat cancer. This review examined the intricate relationship linking the nervous system with breast cancer, emphasizing both central and peripheral aspects of the nervous system. Moreover, we reviewed neural cell factors and their impact on breast cancer progression, alongside the interactions between nerves and immunology, microbiota in breast cancer. Furthermore, the study discussed the potential of nerves as biomarkers for diagnosing and prognosticating breast cancer, and evaluated prospects for improving chemotherapy and immunotherapy therapeutic outcomes in breast cancer treatment. We hope to provide a deeper understanding of the neurobiological underpinnings of breast cancer and pave the way for the discovery of innovative therapeutic targets and prognostic markers.
Collapse
Affiliation(s)
- Jia-feng Wang
- Key Laboratory of Clinical Cancer Pharmacology and Toxicology Research of Zhejiang Province, Affiliated Hangzhou First People's Hospital, School of Medicine, Westlake University, Hangzhou, 310006, China
| | - Meng-chuan Wang
- Affiliated Cixi Hospital, Wenzhou Medical University, Ningbo, 315300, China
| | - Lei-lei Jiang
- The First Affiliated Hospital of Anhui University of Chinese Medicine,Hefei, 230031, China
| | - Neng-ming Lin
- Key Laboratory of Clinical Cancer Pharmacology and Toxicology Research of Zhejiang Province, Affiliated Hangzhou First People's Hospital, School of Medicine, Westlake University, Hangzhou, 310006, China
- Westlake Laboratory of Life Sciences and Biomedicine of Zhejiang Province, Hangzhou, 310024, China
| |
Collapse
|
3
|
Wang J, Zhao F, Zhang Q, Sun Z, Xiahou Z, Wang C, Liu Y, Yu Z. Unveiling the NEFH+ malignant cell subtype: Insights from single-cell RNA sequencing in prostate cancer progression and tumor microenvironment interactions. Front Immunol 2024; 15:1517679. [PMID: 39759507 PMCID: PMC11695424 DOI: 10.3389/fimmu.2024.1517679] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2024] [Accepted: 12/09/2024] [Indexed: 01/07/2025] Open
Abstract
Background Prostate cancer (PCa) is a multifactorial and heterogeneous disease, ranking among the most prevalent malignancies in men. In 2020, there were 1,414,259 new cases of PCa worldwide, accounting for 7.3% of all malignant tumors. The incidence rate of PCa ranks third, following breast cancer and lung cancer. Patients diagnosed with high-grade PCa frequently present with existing or developing metastases, complicating their treatment and resulting in poorer prognoses, particularly for those with bone metastases. Utilizing single-cell RNA sequencing (scRNA-seq), we identified specific malignant cell subtypes that are closely linked to high-grade PCa. By investigating the mechanisms that govern interactions within the tumor microenvironment (TME), we aim to offer new theoretical insights that can enhance the prevention, diagnosis, and treatment of PCa, ultimately striving to improve patient outcomes and quality of life. Methods Data on scRNA-seq was obtained from the GEO database. The gene ontology and gene set enrichment analysis were employed to analyze differential expression genes. Using inferCNV analysis to identify malignant epithelial cells. We subsequently employed Monocle, Cytotrace, and Slingshot packages to infer subtype differentiation trajectories. The cellular communication between malignant cell subtypes and other cells was predicted using the CellChat package. Furthermore, we employed pySCENIC to analyze and identify the regulatory networks of transcription factors (TFs) in malignant cell subtypes. The MDA PCa 2b and VCap cell lines were employed to validate the analysis results through cellular functional experiments. In addition, a risk scoring model was developed to assess the variation in clinical characteristics, prognosis, immune infiltration, immune checkpoint, and drug sensitivity. Results A malignant cell subtype in PCa with high expression of NEFH was identified through scRNA-seq analysis. This subtype was situated at the differentiation terminal, exhibited a higher level of malignancy, and exhibited characteristics that were more prone to advanced tumor lesions. In addition, our research underscored the intricate interactions that exist within the TME, particularly the interaction between PTN secreted by this subtype and fibroblasts via the NCL receptor. This interaction may be closely associated with cancer-associated fibroblasts and tumor progression. Subsequently, we determined that the NEFH+ malignant cell subtype was significantly correlated with the TF IRX4. This TF is linked to a worse prognosis in PCa and may affect disease progression by regulating gene transcription. Our conclusions were additionally verified through cellular experiments. Furthermore, the prognostic model we developed demonstrated satisfactory predictive performance, with gene sets from the high NmRS group facilitating tumor progression and deterioration. The analysis of immune infiltration was instrumental in the development of clinical intervention strategies and patient prognosis. Conclusion By examining the cellular heterogeneity of a unique NEFH+ malignant cell subtype within the PCa microenvironment, we were able to disclose their reciprocal interaction with disease progression. This offers a novel viewpoint on the diagnosis and treatment of PCa.
Collapse
Affiliation(s)
- Jie Wang
- Department of Urology, The Second People’s Hospital of Meishan City, Meishan, Sichuan, China
- Department of Urology, China-Japan Union Hospital of Jilin University, Changchun, Jilin, China
| | - Fu Zhao
- The First Clinical Medical College of Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Qiang Zhang
- Department of Urology, The Second People’s Hospital of Meishan City, Meishan, Sichuan, China
| | - Zhou Sun
- Department of Urology, China-Japan Union Hospital of Jilin University, Changchun, Jilin, China
| | - Zhikai Xiahou
- China Institute of Sport and Health Science, Beijing Sport University, Beijing, China
| | - Changzhong Wang
- Department of Urology, The First People’s Hospital of Jiangxia District, Wuhan, Hubei, China
| | - Yan Liu
- Department of Urology, Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science, Xiangyang, Hubei, China
| | - Zongze Yu
- Department of Urology, The Second People’s Hospital of Meishan City, Meishan, Sichuan, China
| |
Collapse
|
4
|
Chen M, Karimpour PA, Elliott A, He D, Knifley T, Liu J, Wang C, O’Connor KL. Integrin α6β4 Upregulates PTPRZ1 Through UCHL1-Mediated Hif-1α Nuclear Accumulation to Promote Triple-Negative Breast Cancer Cell Invasive Properties. Cancers (Basel) 2024; 16:3683. [PMID: 39518121 PMCID: PMC11545476 DOI: 10.3390/cancers16213683] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2024] [Revised: 10/26/2024] [Accepted: 10/28/2024] [Indexed: 11/16/2024] Open
Abstract
Integrin α6β4 drives triple-negative breast cancer (TNBC) aggressiveness through the transcriptional regulation of key genes. Here, we investigated how integrin α6β4 regulates protein tyrosine phosphatase receptor type Z1 (PTPRZ1). Using stable re-expression of integrin β4 (ITGB4) in cells naturally devoid of integrin α6β4 or knockdown or knockout (KO) of ITGB4, we found that integrin α6β4 regulates PTPRZ1 expression. To gain mechanistic insight, we focused on Hif-1α due to the impact of integrin α6β4 on a hypoxia-associated signature. We found that nuclear localization of Hif-1α, but not Hif-2α, was substantially enhanced with integrin α6β4 signaling. Hif-1α knockdown by shRNA or chemical inhibition decreased PTPRZ1 expression, while chemical activation of Hif-1α increased it. Upstream of Hif-1α, integrin α6β4 upregulates UCHL1 to stabilize Hif-1α and ultimately regulate PTPRZ1. Inhibition of UCHL1 and PTPRZ1 dramatically decreases integrin α6β4-mediated cell migration and three-dimensional invasive growth. Finally, public breast cancer database analyses demonstrated that ITGB4 correlates with PTPRZ1 and that high expression of ITGB4, UCHL1, HIF1A, and PTPRZ1 associated with decreased overall survival, distant metastasis free survival, post progression survival, and relapse-free survival. In summary, these findings provide a novel function of integrin α6β4 in promoting tumor invasive phenotypes through UCHL1-Hif-1α-mediated regulation of PTPRZ1.
Collapse
Affiliation(s)
- Min Chen
- Markey Cancer Center, University of Kentucky, Lexington, KY 40536, USA; (P.A.K.); (A.E.); (D.H.); (T.K.); (J.L.); (C.W.)
- Department of Toxicology and Cancer Biology, University of Kentucky, Lexington, KY 40536, USA
| | - Parvanee A. Karimpour
- Markey Cancer Center, University of Kentucky, Lexington, KY 40536, USA; (P.A.K.); (A.E.); (D.H.); (T.K.); (J.L.); (C.W.)
- Department of Molecular and Cellular Biochemistry, University of Kentucky, Lexington, KY 40536, USA
| | - Andrew Elliott
- Markey Cancer Center, University of Kentucky, Lexington, KY 40536, USA; (P.A.K.); (A.E.); (D.H.); (T.K.); (J.L.); (C.W.)
| | - Daheng He
- Markey Cancer Center, University of Kentucky, Lexington, KY 40536, USA; (P.A.K.); (A.E.); (D.H.); (T.K.); (J.L.); (C.W.)
- Division of Cancer Biostatistics, Department of Internal Medicine, University of Kentucky, Lexington, KY 40536, USA
| | - Teresa Knifley
- Markey Cancer Center, University of Kentucky, Lexington, KY 40536, USA; (P.A.K.); (A.E.); (D.H.); (T.K.); (J.L.); (C.W.)
| | - Jinpeng Liu
- Markey Cancer Center, University of Kentucky, Lexington, KY 40536, USA; (P.A.K.); (A.E.); (D.H.); (T.K.); (J.L.); (C.W.)
- Division of Cancer Biostatistics, Department of Internal Medicine, University of Kentucky, Lexington, KY 40536, USA
| | - Chi Wang
- Markey Cancer Center, University of Kentucky, Lexington, KY 40536, USA; (P.A.K.); (A.E.); (D.H.); (T.K.); (J.L.); (C.W.)
- Division of Cancer Biostatistics, Department of Internal Medicine, University of Kentucky, Lexington, KY 40536, USA
| | - Kathleen L. O’Connor
- Markey Cancer Center, University of Kentucky, Lexington, KY 40536, USA; (P.A.K.); (A.E.); (D.H.); (T.K.); (J.L.); (C.W.)
- Department of Molecular and Cellular Biochemistry, University of Kentucky, Lexington, KY 40536, USA
| |
Collapse
|
5
|
Guo Q, Jin Y, Lin M, Zeng C, Zhang J. NF-κB signaling in therapy resistance of breast cancer: Mechanisms, approaches, and challenges. Life Sci 2024; 348:122684. [PMID: 38710275 DOI: 10.1016/j.lfs.2024.122684] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Revised: 04/19/2024] [Accepted: 05/02/2024] [Indexed: 05/08/2024]
Abstract
Breast cancer is the most common type of cancer and is the second leading cause of cancer-related mortality in women. Chemotherapy, targeted therapy, endocrine therapy, and radiotherapy are all effective in destroying tumor cells, but they also activate the defense and protection systems of cancer cells, leading to treatment resistance. Breast cancer is characterized by a highly inflammatory tumor microenvironment. The NF-κB pathway is essential for connecting inflammation and cancer, as well as for tumor growth and therapy resistance. An increase in NF-κB signaling boosts the growth potential of breast cancer cells and facilitates the spread of tumors to bone, lymph nodes, lungs, and liver. This review focuses on the mechanisms by which chemotherapy, targeted therapy, endocrine therapy, and radiotherapy induce breast cancer resistance through NF-κB signaling. Additionally, we investigate therapeutic regimens, including single agents or in combination with target inhibitors, plant extracts, nanomedicines, and miRNAs, that have been reported in clinical trials, in vivo, and in vitro to reverse resistance. In particular, NF-κB inhibitors combined with tamoxifen were shown to significantly increase the sensitivity of breast cancer cells to tamoxifen. Combination therapy of miRNA-34a with doxorubicin was also found to synergistically inhibit the progression of doxorubicin-resistant breast cancer by inhibiting Notch/NF-κB signaling.
Collapse
Affiliation(s)
- Qing Guo
- Department of Medical Oncology, Fudan University Shanghai Cancer Center, No. 270, Dong'an Road, Shanghai 200032, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Yizi Jin
- Department of Medical Oncology, Fudan University Shanghai Cancer Center, No. 270, Dong'an Road, Shanghai 200032, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Mingxi Lin
- Department of Medical Oncology, Fudan University Shanghai Cancer Center, No. 270, Dong'an Road, Shanghai 200032, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Cheng Zeng
- Department of Medical Oncology, Fudan University Shanghai Cancer Center, No. 270, Dong'an Road, Shanghai 200032, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Jian Zhang
- Department of Medical Oncology, Fudan University Shanghai Cancer Center, No. 270, Dong'an Road, Shanghai 200032, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China.
| |
Collapse
|
6
|
Chen WA, Williams TG, So L, Drew N, Fang J, Ochoa P, Nguyen N, Jawhar Y, Otiji J, Duerksen-Hughes PJ, Reeves ME, Casiano CA, Jin H, Dovat S, Yang J, Boyle KE, Francis-Boyle OL. Duocarmycin SA Reduces Proliferation and Increases Apoptosis in Acute Myeloid Leukemia Cells In Vitro. Int J Mol Sci 2024; 25:4342. [PMID: 38673926 PMCID: PMC11050052 DOI: 10.3390/ijms25084342] [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: 02/24/2024] [Revised: 04/09/2024] [Accepted: 04/10/2024] [Indexed: 04/28/2024] Open
Abstract
Acute myeloid leukemia (AML) is a hematological malignancy that is characterized by an expansion of immature myeloid precursors. Despite therapeutic advances, the prognosis of AML patients remains poor and there is a need for the evaluation of promising therapeutic candidates to treat the disease. The objective of this study was to evaluate the efficacy of duocarmycin Stable A (DSA) in AML cells in vitro. We hypothesized that DSA would induce DNA damage in the form of DNA double-strand breaks (DSBs) and exert cytotoxic effects on AML cells within the picomolar range. Human AML cell lines Molm-14 and HL-60 were used to perform 3-(4,5-dimethylthiazolyl-2)-2,5-diphenyltetrazolium bromide (MTT), DNA DSBs, cell cycle, 5-ethynyl-2-deoxyuridine (EdU), colony formation unit (CFU), Annexin V, RNA sequencing and other assays described in this study. Our results showed that DSA induced DNA DSBs, induced cell cycle arrest at the G2M phase, reduced proliferation and increased apoptosis in AML cells. Additionally, RNA sequencing results showed that DSA regulates genes that are associated with cellular processes such as DNA repair, G2M checkpoint and apoptosis. These results suggest that DSA is efficacious in AML cells and is therefore a promising potential therapeutic candidate that can be further evaluated for the treatment of AML.
Collapse
Affiliation(s)
- William A. Chen
- Department of Pharmaceutical Sciences, School of Pharmacy, Loma Linda University, Shryock Hall 24745 Stewart Street, Loma Linda, CA 92350, USA
| | - Terry G. Williams
- Department of Pharmaceutical Sciences, School of Pharmacy, Loma Linda University, Shryock Hall 24745 Stewart Street, Loma Linda, CA 92350, USA
| | - Leena So
- Department of Pharmaceutical Sciences, School of Pharmacy, Loma Linda University, Shryock Hall 24745 Stewart Street, Loma Linda, CA 92350, USA
| | - Natalie Drew
- Department of Pharmaceutical Sciences, School of Pharmacy, Loma Linda University, Shryock Hall 24745 Stewart Street, Loma Linda, CA 92350, USA
| | - Jie Fang
- Department of Surgery, St. Jude Children’s Research Hospital, 262 Danny Thomas Place, Memphis, TN 38105, USA
| | - Pedro Ochoa
- Department of Basic Sciences, School of Medicine, Loma Linda University, 11175 Campus Street, Loma Linda, CA 92350, USA
- Center for Health Disparities and Molecular Medicine, Loma Linda University, 11085 Campus Street, Loma Linda, CA 92350, USA
| | - Nhi Nguyen
- Department of Pharmaceutical Sciences, School of Pharmacy, Loma Linda University, Shryock Hall 24745 Stewart Street, Loma Linda, CA 92350, USA
| | - Yasmeen Jawhar
- Department of Pharmaceutical Sciences, School of Pharmacy, Loma Linda University, Shryock Hall 24745 Stewart Street, Loma Linda, CA 92350, USA
| | - Jide Otiji
- Department of Pharmaceutical Sciences, School of Pharmacy, Loma Linda University, Shryock Hall 24745 Stewart Street, Loma Linda, CA 92350, USA
| | - Penelope J. Duerksen-Hughes
- Department of Basic Sciences, School of Medicine, Loma Linda University, 11175 Campus Street, Loma Linda, CA 92350, USA
| | - Mark E. Reeves
- Department of Basic Sciences, School of Medicine, Loma Linda University, 11175 Campus Street, Loma Linda, CA 92350, USA
- Department of Surgery, School of Medicine, Loma Linda University, 11234 Anderson Street, Loma Linda, CA 92354, USA
| | - Carlos A. Casiano
- Department of Basic Sciences, School of Medicine, Loma Linda University, 11175 Campus Street, Loma Linda, CA 92350, USA
- Center for Health Disparities and Molecular Medicine, Loma Linda University, 11085 Campus Street, Loma Linda, CA 92350, USA
| | - Hongjian Jin
- Center for Applied Bioinformatics, St. Jude Children’s Research Hospital, 262 Danny Thomas Place, Memphis, TN 38105, USA
| | - Sinisa Dovat
- Departments of Pediatrics, Biochemistry and Molecular Biology, and Pharmacology, Penn State Cancer Institute, 400 University Drive, Hershey, PA 17033, USA
| | - Jun Yang
- Department of Surgery, St. Jude Children’s Research Hospital, 262 Danny Thomas Place, Memphis, TN 38105, USA
| | - Kristopher E. Boyle
- Department of Pharmaceutical Sciences, School of Pharmacy, Loma Linda University, Shryock Hall 24745 Stewart Street, Loma Linda, CA 92350, USA
| | - Olivia L. Francis-Boyle
- Department of Pharmaceutical Sciences, School of Pharmacy, Loma Linda University, Shryock Hall 24745 Stewart Street, Loma Linda, CA 92350, USA
- Department of Basic Sciences, School of Medicine, Loma Linda University, 11175 Campus Street, Loma Linda, CA 92350, USA
- Department of Pathology and Human Anatomy, Division of Anatomy, School of Medicine, Loma Linda University, 11175 Campus Street, Loma Linda, CA 92350, USA
| |
Collapse
|
7
|
Li JY, Hu CJ, Peng H, Chen EQ. A novel immune-related long noncoding RNA (lncRNA) pair model to predict the prognosis of triple-negative breast cancer. Transl Cancer Res 2024; 13:1252-1267. [PMID: 38617505 PMCID: PMC11009803 DOI: 10.21037/tcr-23-1975] [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: 10/24/2023] [Accepted: 02/08/2024] [Indexed: 04/16/2024]
Abstract
BACKGROUND Breast cancer (BC) is the most prevalent cancer type and is the principal cause of cancer-related death in women. Anti-programmed cell death protein 1/programmed cell death ligand 1 (PD-1/PD-L1) immunotherapy has shown promising effects in metastatic triple-negative breast cancer (TNBC), but the potential factors affecting its efficacy have not been elucidated. Immune-related long noncoding RNAs (irlncRNAs) have been reported to be involved in immune escape to influence the carcinogenic process through the PD-1/PD-L1 signaling pathway. Therefore, exploring the potential regulatory mechanism of irlncRNAs in PD-1/PD-L1 immunotherapy in TNBC is of great importance. METHODS We retrieved transcriptome profiling data from The Cancer Genome Atlas (TCGA) and identified differentially expressed irlncRNA (DEirlncRNA) pairs. Least absolute shrinkage and selection operator (LASSO) regression analysis was performed to construct a risk assessment model. RESULTS Receiver operating characteristic (ROC) curve analysis indicated that the risk model may serve as a potential prediction tool in TNBC patients. Clinical stage and risk score were proved to be independent prognostic predictors by univariate and multivariate Cox regression analyses. Subsequently, we investigated the correlation between the risk model and tumor-infiltrating immune cells and immune checkpoints. Finally, we identified USP30-AS1 through the StarBase and Multi Experiment Matrix (MEM) databases, predicted the potential target genes of USP30-AS1, and then discovered that these target genes were closely associated with immune responses. CONCLUSIONS Our study constructed a risk assessment model by irlncRNA pairs regardless of expression levels, which contributed to predicting the efficacy of immunotherapy in TNBC. Furthermore, the lncRNA USP30-AS1 in the model was positively correlated with the expression of PD-L1 and provided a potential therapeutic target for TNBC.
Collapse
Affiliation(s)
- Jing-Ying Li
- Department of Pharmacy, West China Hospital, Sichuan University, Chengdu, China
| | - Chen-Ji Hu
- Department of Pharmacy, West China Hospital, Sichuan University, Chengdu, China
| | - Hui Peng
- Department of Pharmacy, West China Hospital, Sichuan University, Chengdu, China
| | - En-Qiang Chen
- Center of Infectious Diseases, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
8
|
Li J, Liu T, Tang N, Lin S, Zhang F, Yuan W, Zhang T, Deng SH, Wu DM, Xu Y. Cyclin-dependent kinase inhibitor 1A inhibits pyroptosis to enhance human lung adenocarcinoma cell radioresistance by promoting DNA repair. Heliyon 2024; 10:e26975. [PMID: 38468925 PMCID: PMC10926078 DOI: 10.1016/j.heliyon.2024.e26975] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2023] [Revised: 01/26/2024] [Accepted: 02/22/2024] [Indexed: 03/13/2024] Open
Abstract
Purpose One of the best anticancer treatments available is radiotherapy, which can be used either alone or in conjunction with other forms of treatment including chemotherapy and surgery. Nevertheless, a number of biochemical and physiological processes that react to ionizing radiation might provide tumor cells radioresistance, which makes radiotherapy ineffective. It has been found that CDKN1A regulates DNA damage repair, which contributes to tumor radioresistance. However, the precise mechanism is still unknown. Therefore, this study aimed to explore the mechanisms underlying CDKN1A-enhanced radioresistance in tumor cells. Methods Cells were irradiated with 4 Gy after CDKN1A overexpression or knockdown. CDKN1A expression was measured using real-time PCR, cell viability was evaluated using cell counting kit-8 and colony formation assays, and cytotoxicity was assessed using a lactate dehydrogenase assay. Pyroptosis in cells was analyzed using caspase-1 activity assay, enzyme-linked immunosorbent assay, and flow cytometry. Inflammation activation was detected through a co-immunoprecipitation assay. Activation of pyroptosis-related proteins was analyzed using immunohistochemistry, Western blot, and immunofluorescence. Tumor radioresistance in vivo was evaluated in a mouse xenograft model. Results Radiotherapy upregulated CDKN1A expression, which promoted lung adenocarcinoma cell survival. CDKN1A influenced radiation-induced pyroptosis in A549, which mainly depended on inhibiting the activation of the AIM2 inflammasome by promoting DNA repair. Additionally, CDKN1A upregulation enhanced A549 xenograft tumor radioresistance by inhibiting radiation-induced pyroptosis in vivo. Conclusions CDKN1A inhibits pyroptosis to enhance the radioresistance of lung adenocarcinoma cells by promoting DNA repair. This study may serve as a reference for developing novel targeted therapies against cancer.
Collapse
Affiliation(s)
- Jing Li
- School of Clinical Medicine, Chengdu Medical College, Chengdu, Sichuan, 610500, PR China
- The First Affiliated Hospital of Chengdu Medical College, Chengdu, Sichuan, 610500, PR China
| | - Teng Liu
- School of Clinical Medicine, Chengdu Medical College, Chengdu, Sichuan, 610500, PR China
- The First Affiliated Hospital of Chengdu Medical College, Chengdu, Sichuan, 610500, PR China
| | - Ning Tang
- School of Clinical Medicine, Chengdu Medical College, Chengdu, Sichuan, 610500, PR China
- The First Affiliated Hospital of Chengdu Medical College, Chengdu, Sichuan, 610500, PR China
| | - Sheng Lin
- The First People's Hospital of Ziyang City, Ziyang, Sichuan, PR China
| | - Feng Zhang
- School of Clinical Medicine, Chengdu Medical College, Chengdu, Sichuan, 610500, PR China
- The First Affiliated Hospital of Chengdu Medical College, Chengdu, Sichuan, 610500, PR China
| | - Wei Yuan
- School of Clinical Medicine, Chengdu Medical College, Chengdu, Sichuan, 610500, PR China
- The First Affiliated Hospital of Chengdu Medical College, Chengdu, Sichuan, 610500, PR China
| | - Ting Zhang
- School of Clinical Medicine, Chengdu Medical College, Chengdu, Sichuan, 610500, PR China
- The First Affiliated Hospital of Chengdu Medical College, Chengdu, Sichuan, 610500, PR China
| | - Shi-hua Deng
- School of Clinical Medicine, Chengdu Medical College, Chengdu, Sichuan, 610500, PR China
- The First Affiliated Hospital of Chengdu Medical College, Chengdu, Sichuan, 610500, PR China
| | - Dong-ming Wu
- School of Clinical Medicine, Chengdu Medical College, Chengdu, Sichuan, 610500, PR China
- The First Affiliated Hospital of Chengdu Medical College, Chengdu, Sichuan, 610500, PR China
| | - Ying Xu
- School of Clinical Medicine, Chengdu Medical College, Chengdu, Sichuan, 610500, PR China
- The First Affiliated Hospital of Chengdu Medical College, Chengdu, Sichuan, 610500, PR China
| |
Collapse
|
9
|
Ganguly D, Schmidt MO, Coleman M, Ngo TVC, Sorrelle N, Dominguez AT, Murimwa GZ, Toombs JE, Lewis C, Fang YV, Valdes-Mora F, Gallego-Ortega D, Wellstein A, Brekken RA. Pleiotrophin drives a prometastatic immune niche in breast cancer. J Exp Med 2023; 220:e20220610. [PMID: 36828390 PMCID: PMC9998964 DOI: 10.1084/jem.20220610] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2022] [Revised: 11/21/2022] [Accepted: 01/09/2023] [Indexed: 11/04/2022] Open
Abstract
Metastatic cancer cells adapt to thrive in secondary organs. To investigate metastatic adaptation, we performed transcriptomic analysis of metastatic and non-metastatic murine breast cancer cells. We found that pleiotrophin (PTN), a neurotrophic cytokine, is a metastasis-associated factor that is expressed highly by aggressive breast cancers. Moreover, elevated PTN in plasma correlated significantly with metastasis and reduced survival of breast cancer patients. Mechanistically, we find that PTN activates NF-κB in cancer cells leading to altered cytokine production, subsequent neutrophil recruitment, and an immune suppressive microenvironment. Consequently, inhibition of PTN, pharmacologically or genetically, reduces the accumulation of tumor-associated neutrophils and reverts local immune suppression, resulting in increased T cell activation and attenuated metastasis. Furthermore, inhibition of PTN significantly enhanced the efficacy of immune checkpoint blockade and chemotherapy in reducing metastatic burden in mice. These findings establish PTN as a previously unrecognized driver of a prometastatic immune niche and thus represents a promising therapeutic target for the treatment of metastatic breast cancer.
Collapse
Affiliation(s)
- Debolina Ganguly
- Department of Surgery, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Hamon Center for Therapeutic Oncology Research, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Harold C. Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Cancer Biology Graduate Program, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Marcel O. Schmidt
- Lombardi Comprehensive Cancer Center, Georgetown University, Washington, DC, USA
| | - Morgan Coleman
- Department of Surgery, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Hamon Center for Therapeutic Oncology Research, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Tuong-Vi Cindy Ngo
- Hamon Center for Therapeutic Oncology Research, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Noah Sorrelle
- Hamon Center for Therapeutic Oncology Research, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Adrian T.A. Dominguez
- Hamon Center for Therapeutic Oncology Research, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Gilbert Z. Murimwa
- Department of Surgery, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Hamon Center for Therapeutic Oncology Research, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Jason E. Toombs
- Department of Surgery, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Hamon Center for Therapeutic Oncology Research, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Cheryl Lewis
- Harold C. Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Department of Pathology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Yisheng V. Fang
- Harold C. Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Department of Pathology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Fatima Valdes-Mora
- Cancer Epigenetic Biology and Therapeutics group, Precision Medicine Theme, Children’s Cancer Institute, Sydney, Australia
- School of Clinical Medicine, University of NSW Sydney, Sydney, Australia
| | - David Gallego-Ortega
- School of Biomedical Engineering, Faculty of Engineering and Information Technology, University of Technology Sydney, Sydney, Australia
- Garvan Institute of Medical Research and The Kinghorn Cancer Centre, Sydney, Australia
- School of Clinical Medicine, St Vincent’s Healthcare Clinical Campus, Faculty of Medicine and Health, UNSW Sydney, Sydney, Australia
| | - Anton Wellstein
- Lombardi Comprehensive Cancer Center, Georgetown University, Washington, DC, USA
| | - Rolf A. Brekken
- Department of Surgery, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Hamon Center for Therapeutic Oncology Research, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Harold C. Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Cancer Biology Graduate Program, University of Texas Southwestern Medical Center, Dallas, TX, USA
| |
Collapse
|
10
|
Papadimitriou E, Kanellopoulou VK. Protein Tyrosine Phosphatase Receptor Zeta 1 as a Potential Target in Cancer Therapy and Diagnosis. Int J Mol Sci 2023; 24:ijms24098093. [PMID: 37175798 PMCID: PMC10178973 DOI: 10.3390/ijms24098093] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Revised: 04/25/2023] [Accepted: 04/27/2023] [Indexed: 05/15/2023] Open
Abstract
Protein tyrosine phosphatase receptor zeta 1 (PTPRZ1) is a type V transmembrane tyrosine phosphatase that is highly expressed during embryonic development, while its expression during adulthood is limited. PTPRZ1 is highly detected in the central nervous system, affecting oligodendrocytes' survival and maturation. In gliomas, PTPRZ1 expression is significantly upregulated and is being studied as a potential cancer driver and as a target for therapy. PTPRZ1 expression is also increased in other cancer types, but there are no data on the potential functional significance of this finding. On the other hand, low PTPRZ1 expression seems to be related to a worse prognosis in some cancer types, suggesting that in some cases, it may act as a tumor-suppressor gene. These discrepancies may be due to our limited understanding of PTPRZ1 signaling and tumor microenvironments. In this review, we present evidence on the role of PTPRZ1 in angiogenesis and cancer and discuss the phenomenal differences among the different types of cancer, depending on the regulation of its tyrosine phosphatase activity or ligand binding. Clarifying the involved signaling pathways will lead to its efficient exploitation as a novel therapeutic target or as a biomarker, and the development of proper therapeutic approaches.
Collapse
Affiliation(s)
- Evangelia Papadimitriou
- Laboratory of Molecular Pharmacology, Department of Pharmacy, University of Patras, 26504 Patras, Greece
| | - Vasiliki K Kanellopoulou
- Laboratory of Molecular Pharmacology, Department of Pharmacy, University of Patras, 26504 Patras, Greece
| |
Collapse
|
11
|
Azari M, Bahreini F, Uversky VN, Rezaei N. Current therapeutic approaches and promising perspectives of using bioengineered peptides in fighting chemoresistance in triple-negative breast cancer. Biochem Pharmacol 2023; 210:115459. [PMID: 36813121 DOI: 10.1016/j.bcp.2023.115459] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2022] [Revised: 02/11/2023] [Accepted: 02/14/2023] [Indexed: 02/22/2023]
Abstract
Breast cancer is a collation of malignancies that manifest in the mammary glands at the early stages. Among breast cancer subtypes, triple-negative breast cancer (TNBC) shows the most aggressive behavior, with apparent stemness features. Owing to the lack of response to hormone therapy and specific targeted therapies, chemotherapy remains the first line of the TNBC treatment. However, the acquisition of resistance to chemotherapeutic agents increase therapy failure, and promotes cancer recurrence and distant metastasis. Invasive primary tumors are the birthplace of cancer burden, though metastasis is a key attribute of TNBC-associated morbidity and mortality. Targeting the chemoresistant metastases-initiating cells via specific therapeutic agents with affinity to the upregulated molecular targets is a promising step in the TNBC clinical management. Exploring the capacity of peptides as biocompatible entities with the specificity of action, low immunogenicity, and robust efficacy provides a principle for designing peptide-based drugs capable of increasing the efficacy of current chemotherapy agents for selective targeting of the drug-tolerant TNBC cells. Here, we first focus on the resistance mechanisms that TNBC cells acquire to evade the effect of chemotherapeutic agents. Next, the novel therapeutic approaches employing tumor-targeting peptides to exploit the mechanisms of drug resistance in chemorefractory TNBC are described.
Collapse
Affiliation(s)
- Mandana Azari
- School of Chemical Engineering-Biotechnology, College of Engineering, University of Tehran, Iran; Network of Immunity in Infection, Malignancy and Autoimmunity (NIIMA), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Farbod Bahreini
- Network of Immunity in Infection, Malignancy and Autoimmunity (NIIMA), Universal Scientific Education and Research Network (USERN), Tehran, Iran; Department of Biochemistry, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran
| | - Vladimir N Uversky
- Department of Molecular Medicine and USF Health Byrd Alzheimer's Research Institute, Morsani College of Medicine, University of South Florida, Tampa, USA
| | - Nima Rezaei
- Network of Immunity in Infection, Malignancy and Autoimmunity (NIIMA), Universal Scientific Education and Research Network (USERN), Tehran, Iran; Research Center for Immunodeficiencies (RCID), Children's Medical Center, Tehran University of Medical Sciences, Tehran, Iran; Department of Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
12
|
Lima APB, da Silva GN. Long Non-Coding RNA and Chemoresistance in Bladder Cancer - A Mini Review. Cancer Invest 2023; 41:164-172. [PMID: 36373675 DOI: 10.1080/07357907.2022.2146703] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Bladder cancer is the 10th most common cancer worldwide. It is a heterogeneous disease, comprising several tumor subtypes with differences in histology, genomic aberrations, prognosis and sensitivity to anti-cancer treatments. Although the treatment of bladder cancer is based tumor classifications and gradings, patients have different clinical response. In recent years, long non-coding RNAs (lncRNAs) were associated with bladder cancer chemoresistance. Thus, lncRNAs seem to be promising targets in treatment of bladder cancer. This review highlights the recent findings concerning lncRNAs and their relevance to the chemoresistance of bladder cancer. This may provide a basis for exploiting more robust therapeutic approaches in the future.
Collapse
Affiliation(s)
- Ana Paula Braga Lima
- Programa de Pós-graduação em Ciências Farmacêuticas (CIPHARMA), Universidade Federal de Ouro Preto, Ouro Preto, Minas Gerais, Brazil
| | - Glenda Nicioli da Silva
- Programa de Pós-graduação em Ciências Farmacêuticas (CIPHARMA), Universidade Federal de Ouro Preto, Ouro Preto, Minas Gerais, Brazil.,Programa de Pós-graduação em Ciência Biológicas (CBIOL), Universidade Federal de Ouro Preto, Ouro Preto, Minas Gerais, Brazil.,Departamento de Análises Clínicas (DEACL), Escola de Farmácia, Universidade Federal de Ouro Preto, Ouro Preto, Minas Gerais, Brazil
| |
Collapse
|
13
|
Zheng K, Hou Y, Zhang Y, Wang F, Sun A, Yang D. Molecular features and predictive models identify the most lethal subtype and a therapeutic target for osteosarcoma. Front Oncol 2023; 13:1111570. [PMID: 36874110 PMCID: PMC9980341 DOI: 10.3389/fonc.2023.1111570] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Accepted: 01/23/2023] [Indexed: 02/18/2023] Open
Abstract
Background Osteosarcoma is the most common primary malignant bone tumor. The existing treatment regimens remained essentially unchanged over the past 30 years; hence the prognosis has plateaued at a poor level. Precise and personalized therapy is yet to be exploited. Methods One discovery cohort (n=98) and two validation cohorts (n=53 & n=48) were collected from public data sources. We performed a non-negative matrix factorization (NMF) method on the discovery cohort to stratify osteosarcoma. Survival analysis and transcriptomic profiling characterized each subtype. Then, a drug target was screened based on subtypes' features and hazard ratios. We also used specific siRNAs and added a cholesterol pathway inhibitor to osteosarcoma cell lines (U2OS and Saos-2) to verify the target. Moreover, PermFIT and ProMS, two support vector machine (SVM) tools, and the least absolute shrinkage and selection operator (LASSO) method, were employed to establish predictive models. Results We herein divided osteosarcoma patients into four subtypes (S-I ~ S-IV). Patients of S- I were found probable to live longer. S-II was characterized by the highest immune infiltration. Cancer cells proliferated most in S-III. Notably, S-IV held the most unfavorable outcome and active cholesterol metabolism. SQLE, a rate-limiting enzyme for cholesterol biosynthesis, was identified as a potential drug target for S-IV patients. This finding was further validated in two external independent osteosarcoma cohorts. The function of SQLE to promote proliferation and migration was confirmed by cell phenotypic assays after the specific gene knockdown or addition of terbinafine, an inhibitor of SQLE. We further employed two machine learning tools based on SVM algorithms to develop a subtype diagnostic model and used the LASSO method to establish a 4-gene model for predicting prognosis. These two models were also verified in a validation cohort. Conclusion The molecular classification enhanced our understanding of osteosarcoma; the novel predicting models served as robust prognostic biomarkers; the therapeutic target SQLE opened a new way for treatment. Our results served as valuable hints for future biological studies and clinical trials of osteosarcoma.
Collapse
Affiliation(s)
- Kun Zheng
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing, China.,Department of Orthopedics, General Hospital of Southern Theater Command, Guangzhou, China
| | - Yushan Hou
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing, China.,Research Unit of Proteomics Driven Cancer Precision Medicine, Chinese Academy of Medical Sciences, Beijing, China
| | - Yiming Zhang
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing, China.,Research Unit of Proteomics Driven Cancer Precision Medicine, Chinese Academy of Medical Sciences, Beijing, China
| | - Fei Wang
- Department of Orthopedics, General Hospital of Southern Theater Command, Guangzhou, China
| | - Aihua Sun
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing, China.,Research Unit of Proteomics Driven Cancer Precision Medicine, Chinese Academy of Medical Sciences, Beijing, China
| | - Dong Yang
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing, China
| |
Collapse
|
14
|
Zhu S, Li Z, Zheng D, Yu Y, Xiang J, Ma X, Xu D, Qiu J, Yang Z, Wang Z, Li J, Sun H, Chen W, Meng X, Lu Y, Ren Q. A cancer cell membrane coated, doxorubicin and microRNA co-encapsulated nanoplatform for colorectal cancer theranostics. Mol Ther Oncolytics 2022; 28:182-196. [PMID: 36820302 PMCID: PMC9937835 DOI: 10.1016/j.omto.2022.12.002] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2022] [Accepted: 12/05/2022] [Indexed: 12/13/2022] Open
Abstract
Endogenous microRNAs (miRNA) in tumors are currently under exhaustive investigation as potential therapeutic agents for cancer treatment. Nevertheless, RNase degradation, inefficient and untargeted delivery, limited biological effect, and currently unclear side effects remain unsettled issues that frustrate clinical application. To address this, a versatile targeted delivery system for multiple therapeutic and diagnostic agents should be adapted for miRNA. In this study, we developed membrane-coated PLGA-b-PEG DC-chol nanoparticles (m-PPDCNPs) co-encapsulating doxorubicin (Dox) and miRNA-190-Cy7. Such a system showed low biotoxicity, high loading efficiency, and superior targeting ability. Systematic delivery of m-PPDCNPs in mouse models showed exceptionally specific tumor accumulation. Sustained release of miR-190 inhibited tumor angiogenesis, tumor growth, and migration by regulating a large group of angiogenic effectors. Moreover, m-PPDCNPs also enhanced the sensitivity of Dox by suppressing TGF-β signal in colorectal cancer cell lines and mouse models. Together, our results demonstrate a stimulating and promising m-PPDCNPs nanoplatform for colorectal cancer theranostics.
Collapse
Affiliation(s)
- Sihao Zhu
- Department of Biomedical Engineering, College of Future Technology, Peking University, Beijing 100871, China,Institute of Medical Technology, Peking University Health Science Center, Peking University, Beijing 100191, China,National Biomedical Imaging Center, Peking University, Beijing 100871, China,Institute of Biomedical Engineering, Shenzhen Bay Laboratory, Shenzhen 5181071, China,Institute of Biomedical Engineering, Peking University Shenzhen Graduate School, Shenzhen 518055, China
| | - Ziyuan Li
- Department of Biomedical Engineering, College of Future Technology, Peking University, Beijing 100871, China,Institute of Medical Technology, Peking University Health Science Center, Peking University, Beijing 100191, China,National Biomedical Imaging Center, Peking University, Beijing 100871, China,Institute of Biomedical Engineering, Shenzhen Bay Laboratory, Shenzhen 5181071, China,Institute of Biomedical Engineering, Peking University Shenzhen Graduate School, Shenzhen 518055, China
| | - Dongye Zheng
- Department of Biomedical Engineering, College of Future Technology, Peking University, Beijing 100871, China,Institute of Medical Technology, Peking University Health Science Center, Peking University, Beijing 100191, China,National Biomedical Imaging Center, Peking University, Beijing 100871, China,Institute of Biomedical Engineering, Shenzhen Bay Laboratory, Shenzhen 5181071, China,Institute of Biomedical Engineering, Peking University Shenzhen Graduate School, Shenzhen 518055, China
| | - Yue Yu
- Beijing National Laboratory for Molecular Sciences (BNLMS), Key Laboratory of Bioorganic Chemistry and Molecular Engineering of Ministry of Education, College of Chemistry and Molecular Engineering, Peking University, Beijing 100871, China
| | - Jing Xiang
- Department of Biomedical Engineering, College of Future Technology, Peking University, Beijing 100871, China,Institute of Medical Technology, Peking University Health Science Center, Peking University, Beijing 100191, China,National Biomedical Imaging Center, Peking University, Beijing 100871, China,Institute of Biomedical Engineering, Shenzhen Bay Laboratory, Shenzhen 5181071, China,Institute of Biomedical Engineering, Peking University Shenzhen Graduate School, Shenzhen 518055, China
| | - Xiao Ma
- Research Group Signal Transduction, Department of Psychiatry, Ludwig Maximilian University of Munich, Nussbaumstr.7, 80336 Munich, Germany
| | - Dongqing Xu
- Department of Pediatric Hematology/Oncology, Xin Hua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China
| | - Jiajun Qiu
- Department of Otolaryngology Head and Neck Surgery, the Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China
| | - Ziyu Yang
- College of Physics and Optoelectronic Engineering, Shenzhen University, Shenzhen 518060, China
| | - Zhiyi Wang
- School of Chemistry and Chemical Engineering, South China University of Technology, Guangzhou 510640, China
| | - Jun Li
- Laboratory Animal Center, Peking University, Beijing 100871, China
| | - Hongfang Sun
- Department of Biomedical Engineering, College of Future Technology, Peking University, Beijing 100871, China
| | - Weiqiang Chen
- Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou 730000, Gansu Province, China,Key Laboratory of Heavy Ion Radiation Biology and Medicine of Chinese Academy of Sciences, Key Laboratory of Basic Research on Heavy Ion Radiation Application in Medicine, Lanzhou 730000, Gansu Province, China
| | - Xiangxi Meng
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Nuclear Medicine, Peking University Cancer Hospital & Institute, Beijing 100142, China,NMPA Key Laboratory for Research and Evaluation of Radiopharmaceuticals, Beijing 100142, China,Corresponding author.
| | - Yanye Lu
- Department of Biomedical Engineering, College of Future Technology, Peking University, Beijing 100871, China,Institute of Medical Technology, Peking University Health Science Center, Peking University, Beijing 100191, China,National Biomedical Imaging Center, Peking University, Beijing 100871, China,Institute of Biomedical Engineering, Peking University Shenzhen Graduate School, Shenzhen 518055, China,Corresponding author.
| | - Qiushi Ren
- Department of Biomedical Engineering, College of Future Technology, Peking University, Beijing 100871, China,National Biomedical Imaging Center, Peking University, Beijing 100871, China,Institute of Biomedical Engineering, Shenzhen Bay Laboratory, Shenzhen 5181071, China,Institute of Biomedical Engineering, Peking University Shenzhen Graduate School, Shenzhen 518055, China,Corresponding author.
| |
Collapse
|
15
|
Wen Y, Ouyang D, Zou Q, Chen Q, Luo N, He H, Anwar M, Yi W. A literature review of the promising future of TROP2: a potential drug therapy target. ANNALS OF TRANSLATIONAL MEDICINE 2022; 10:1403. [PMID: 36660684 PMCID: PMC9843409 DOI: 10.21037/atm-22-5976] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/21/2022] [Accepted: 12/19/2022] [Indexed: 12/29/2022]
Abstract
Background and Objective Previous studies have demonstrated that the oncogene trophoblast cell surface antigen 2 (TROP2) has great application prospects as a therapeutic target. However, few literature reviews have systematically summarized and evaluated its role in cancer therapy. This study aims to summarize the molecular structure, functions, signal transduction pathways, and prognostic value of TROP2, and explore therapeutic agents that target TROP2. Methods A total of 1,376 published literatures from PubMed and 614 published literatures from EMBASE were retrieved by searching "TROP2" or "Trophoblast cell surface antigen 2". The search was conducted on December 12, 2020, and updated on November 20, 2022. The cBioportal and GEPIA (Gene Expression Profiling Interactive Analysis) databases were used to analyze the expression, mutation, and prognostic value of TROP2 in different types of cancer. Key Content and Findings TROP2 is overexpressed in different tumor tissues and plays roles in cell proliferation, invasion, migration, apoptosis, and treatment resistance by binding to or interacting with several molecules. As a therapeutic target, TROP2 is particularly suitable for antibody-based therapies. Monoclonal antibodies, bispecific antibodies, antibody-drug conjugates (ADCs), virus-like particles, and antibody drugs in combination with traditional chemotherapy, immunotherapy, radioimmunotherapy, photoimmunotherapy, and nanoparticles that target TROP2 have thus far been rapidly developed. For example, sacituzumab govitecan (IMMU-132), a TROP2-targeting ADC, was granted accelerated approval for the treatment of metastatic triple-negative breast cancer (TNBC). Anti-TROP2 antibody-conjugated nanoparticles (ST-NPs) are a promising vehicle for delivering doxorubicin in targeted TNBC therapy. Conclusions The availability of TROP2-targeting ADCs makes TROP2 an accessible and promising therapeutic target for advanced metastatic cancers. The present review describes the important role of TROP2 in tumorigenesis and its potential applications as a promising biomarker and therapeutic target that is capable of reversing resistance.
Collapse
Affiliation(s)
- Ying Wen
- Department of General Surgery, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Dengjie Ouyang
- Department of General Surgery, Xiangya Hospital, Central South University, Changsha, China
| | - Qiongyan Zou
- Department of General Surgery, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Qitong Chen
- Department of General Surgery, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Na Luo
- Department of General Surgery, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Hongye He
- Department of General Surgery, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Munawar Anwar
- Department of General Surgery, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Wenjun Yi
- Department of General Surgery, The Second Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
16
|
Leveraging diverse cell-death patterns to predict the prognosis and drug sensitivity of triple-negative breast cancer patients after surgery. Int J Surg 2022; 107:106936. [PMID: 36341760 DOI: 10.1016/j.ijsu.2022.106936] [Citation(s) in RCA: 230] [Impact Index Per Article: 76.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Revised: 08/08/2022] [Accepted: 09/09/2022] [Indexed: 10/14/2022]
Abstract
BACKGROUND Postoperative progression and chemotherapy resistance is the major cause of treatment failure in patients with triple-negative breast cancer (TNBC). Currently, there is a lack of an ideal predictive model for the progression and drug sensitivity of postoperative TNBC patients. Diverse programmed cell death (PCD) patterns play an important role in tumor progression, which has the potential to be a prognostic and drug sensitivity indicator for TNBC after surgery. MATERIALS AND METHODS Twelve PCD patterns (apoptosis, necroptosis, pyroptosis, ferroptosis, cuproptosis, entotic cell death, netotic cell death, parthanatos, lysosome-dependent cell death, autophagy-dependent cell death, alkaliptosis, and oxeiptosis) were analyzed for model construction. Bulk transcriptome, single-cell transcriptome, genomics, and clinical information were collected from TCGA-BRCA, METABRIC, GSE58812, GSE21653, GSE176078, GSE75688, and KM-plotter cohorts to validate the model. RESULTS The machine learning algorithm established a cell death index (CDI) with a 12-gene signature. Validated in five independent datasets, TNBC patients with high CDI had a worse prognosis after surgery. Two molecular subtypes of TNBC with distinct vital biological processes were identified by an unsupervised clustering model. A nomogram with high predictive performance was constructed by incorporating CDI with clinical features. Furthermore, CDI was associated with immune checkpoint genes and key tumor microenvironment components by integrated analysis of bulk and single-cell transcriptome. TNBC patients with high CDI are resistant to standard adjuvant chemotherapy regimens (docetaxel, oxaliplatin, etc.); however, they might be sensitive to palbociclib (an FDA-approved drug for luminal breast cancer). CONCLUSION Generally, we established a novel CDI model by comprehensively analyzing diverse cell death patterns, which can accurately predict clinical prognosis and drug sensitivity of TNBC after surgery. A user-friendly website was created to facilitate the application of this prediction model (https://tnbc.shinyapps.io/CDI_Model/).
Collapse
|
17
|
Benchama O, Tyukhtenko S, Malamas MS, Williams MK, Makriyannis A, Avraham HK. Inhibition of triple negative breast cancer-associated inflammation, tumor growth and brain colonization by targeting monoacylglycerol lipase. Sci Rep 2022; 12:5328. [PMID: 35351947 PMCID: PMC8964799 DOI: 10.1038/s41598-022-09358-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2021] [Accepted: 03/22/2022] [Indexed: 12/15/2022] Open
Abstract
While the prevalence of breast cancer metastasis in the brain is significantly higher in triple negative breast cancers (TNBCs), there is a lack of novel and/or improved therapies for these patients. Monoacylglycerol lipase (MAGL) is a hydrolase involved in lipid metabolism that catalyzes the degradation of 2-arachidonoylglycerol (2-AG) linked to generation of pro- and anti-inflammatory molecules. Here, we targeted MAGL in TNBCs, using a potent carbamate-based inhibitor AM9928 (hMAGL IC50 = 9 nM) with prolonged pharmacodynamic effects (46 h of target residence time). AM9928 blocked TNBC cell adhesion and transmigration across human brain microvascular endothelial cells (HBMECs) in 3D co-cultures. In addition, AM9928 inhibited the secretion of IL-6, IL-8, and VEGF-A from TNBC cells. TNBC-derived exosomes activated HBMECs resulting in secretion of elevated levels of IL-8 and VEGF, which were inhibited by AM9928. Using in vivo studies of syngeneic GFP-4T1-BrM5 mammary tumor cells, AM9928 inhibited tumor growth in the mammary fat pads and attenuated blood brain barrier (BBB) permeability changes, resulting in reduced TNBC colonization in brain. Together, these results support the potential clinical application of MAGL inhibitors as novel treatments for TNBC.
Collapse
Affiliation(s)
- Othman Benchama
- The Center for Drug Discovery, Northeastern University, 360 Huntington Avenue, Boston, MA, 02115, USA
| | - Sergiy Tyukhtenko
- The Center for Drug Discovery, Northeastern University, 360 Huntington Avenue, Boston, MA, 02115, USA
| | - Michael S Malamas
- The Center for Drug Discovery, Northeastern University, 360 Huntington Avenue, Boston, MA, 02115, USA
| | | | - Alexandros Makriyannis
- The Center for Drug Discovery, Northeastern University, 360 Huntington Avenue, Boston, MA, 02115, USA
| | - Hava Karsenty Avraham
- The Center for Drug Discovery, Northeastern University, 360 Huntington Avenue, Boston, MA, 02115, USA.
| |
Collapse
|
18
|
Gu L, Jiang C, Xu C, Liu Y, Zhou H. Based on Molecular Subtypes, Immune Characteristics and Genomic Variation to Constructing and Verifying Multi-Gene Prognostic Characteristics of Colorectal Cancer. Front Cell Dev Biol 2022; 10:828415. [PMID: 35281077 PMCID: PMC8905350 DOI: 10.3389/fcell.2022.828415] [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: 12/03/2021] [Accepted: 01/31/2022] [Indexed: 11/25/2022] Open
Abstract
Background: Colon cancer (COAD) has been identified as being among the most prevalent tumors globally and ranked the third major contributor to cancer-related mortality. COAD is a molecularly heterogeneous disease. There are great differences in clinical manifestations and prognosis among different molecular subtypes. Methods:379 TCGA-COAD samples were divided into four subtypes: primary proliferative, with collective, crypt-like, and EMT invasion. The differences among the four subtypes were analyzed from the multidimensional perspectives of immunity, genomic variation, and prognosis. The limma package was utilized to identify differentially expressed genes (DEGs) amongst different molecular subtypes. Phenotype-related coexpressed gene modules were identified using WGCNA. The polygenic prognosis model was created utilizing the lasso Cox analysis and verified by time-dependent subject operating characteristics (ROC). Results: There are some differences in prognosis, TMB and common gene variation, immune score, and immunotherapy/chemotherapy between proliferative and three invasive molecular subtypes. 846 differential genes (DEGs) were obtained by limma packet analysis. Differential gene analysis was utilized to screen the DEGs among distinct subtypes, which were significantly enriched in the pathways related to tumorigenesis and development. Co-expression network analysis found 46 co-expressed genes correlated with proliferative and three invasive phenotypes. Based on differentially co-expressed genes, we developed a prognostic risk model of 8-genes signature, which exhibited strong stability regardless of external and internal validation. RT-PCR experiments proved the expression of eight genes in tumor and normal samples. Conclusion: We have developed an eight-gene signature prognostic stratification system. Furthermore, we proposed that this classifier can serve as a molecular diagnostic tool to assess the prognosis of colon cancer patients.
Collapse
Affiliation(s)
- Lei Gu
- Department of Gastrointestinal Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Chunhui Jiang
- Department of Gastrointestinal Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Chunjie Xu
- Department of Gastrointestinal Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Ye Liu
- Department of Gastrointestinal Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Hong Zhou
- Department of Gastrointestinal Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| |
Collapse
|
19
|
Negishi R, Yamakawa H, Kobayashi T, Horikawa M, Shimoyama T, Koizumi F, Sawada T, Oboki K, Omuro Y, Funasaka C, Kageyama A, Kanemasa Y, Tanaka T, Matsunaga T, Yoshino T. Transcriptomic profiling of single circulating tumor cells provides insight into human metastatic gastric cancer. Commun Biol 2022; 5:20. [PMID: 35017627 PMCID: PMC8752828 DOI: 10.1038/s42003-021-02937-x] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Accepted: 12/01/2021] [Indexed: 12/24/2022] Open
Abstract
Transcriptome analysis of circulating tumor cells (CTCs), which migrate into blood vessels from primary tumor tissues, at the single-cell level offers critical insights into the biology of metastasis and contributes to drug discovery. However, transcriptome analysis of single CTCs has only been reported for a limited number of cancer types, such as multiple myeloma, breast, hepatocellular, and prostate cancer. Herein, we report the transcriptome analysis of gastric cancer single-CTCs. We utilized an antigen-independent strategy for CTC isolation from metastatic gastric cancer patients involving a size-dependent recovery of CTCs and a single cell isolation technique. The transcriptomic profile of single-CTCs revealed that a majority of gastric CTCs had undergone epithelial-mesenchymal transition (EMT), and indicated the contribution of platelet adhesion toward EMT progression and acquisition of chemoresistance. Taken together, this study serves to employ CTC characterization to elucidate the mechanisms of chemoresistance and metastasis in gastric cancer.
Collapse
Affiliation(s)
- Ryo Negishi
- Division of Biotechnology and Life science, Institute of Engineering, Tokyo University of Agriculture and Technology, 2-24-16, Naka-cho, Koganei, Tokyo, 184-8588, Japan
| | - Hitomi Yamakawa
- Division of Biotechnology and Life science, Institute of Engineering, Tokyo University of Agriculture and Technology, 2-24-16, Naka-cho, Koganei, Tokyo, 184-8588, Japan
| | - Takeru Kobayashi
- Division of Biotechnology and Life science, Institute of Engineering, Tokyo University of Agriculture and Technology, 2-24-16, Naka-cho, Koganei, Tokyo, 184-8588, Japan
| | - Mayuko Horikawa
- Division of Biotechnology and Life science, Institute of Engineering, Tokyo University of Agriculture and Technology, 2-24-16, Naka-cho, Koganei, Tokyo, 184-8588, Japan
| | - Tatsu Shimoyama
- Department of Medical Oncology, Tokyo Metropolitan Cancer and Infectious Diseases Center Komagome Hospital, Tokyo, Japan
| | - Fumiaki Koizumi
- Department of Laboratory Medicine, Tokyo Metropolitan Cancer and Infectious Diseases Center Komagome Hospital, Tokyo, Japan
| | - Takeshi Sawada
- Department of Medical Oncology, Tokyo Metropolitan Cancer and Infectious Diseases Center Komagome Hospital, Tokyo, Japan
| | - Keisuke Oboki
- Center for Medical Research Cooperation, Tokyo Metropolitan Institute of Medical Science, Setagaya-ku, Japan
| | - Yasushi Omuro
- Department of Medical Oncology, Tokyo Metropolitan Cancer and Infectious Diseases Center Komagome Hospital, Tokyo, Japan
| | - Chikako Funasaka
- Department of Medical Oncology, Tokyo Metropolitan Cancer and Infectious Diseases Center Komagome Hospital, Tokyo, Japan
| | - Akihiko Kageyama
- Department of Medical Oncology, Tokyo Metropolitan Cancer and Infectious Diseases Center Komagome Hospital, Tokyo, Japan
| | - Yusuke Kanemasa
- Department of Medical Oncology, Tokyo Metropolitan Cancer and Infectious Diseases Center Komagome Hospital, Tokyo, Japan
| | - Tsuyoshi Tanaka
- Division of Biotechnology and Life science, Institute of Engineering, Tokyo University of Agriculture and Technology, 2-24-16, Naka-cho, Koganei, Tokyo, 184-8588, Japan
| | - Tadashi Matsunaga
- Division of Biotechnology and Life science, Institute of Engineering, Tokyo University of Agriculture and Technology, 2-24-16, Naka-cho, Koganei, Tokyo, 184-8588, Japan
| | - Tomoko Yoshino
- Division of Biotechnology and Life science, Institute of Engineering, Tokyo University of Agriculture and Technology, 2-24-16, Naka-cho, Koganei, Tokyo, 184-8588, Japan.
| |
Collapse
|
20
|
Wang Y, Yang L, Fan C, Mu H, Han M, Liu T, Xie L, Gao Q. miR-130b Expression Level Changes Promote Cervical Cancer Cell Proliferation But Inhibit its Apoptosis by Targeting CDKN1A Gene. Curr Cancer Drug Targets 2022; 22:153-168. [PMID: 35016595 PMCID: PMC9413419 DOI: 10.2174/1568009622666220111090715] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2021] [Revised: 10/21/2021] [Accepted: 11/12/2021] [Indexed: 12/05/2022]
Abstract
Background:
Dysregulation of miR-130b expression is associated with the development of different cancers. However, the description of the biological roles of miR-130b in the growth and survival of cervical cancer cells is limited. Methods:
The miR-130b levels in cervical cancer cells during different stages of growth were determined using reverse transcription-quantitative PCR. The methylation level of DNA sequences upstream of the miR-130b gene was measured using an SYBR Green-based quantitative methylation-specific PCR. Reverse transcription-quantitative PCR, Western blotting, and fluorescence report assays were used to identify the miR-130b-targeted gene. Cell counting kit-8 and comet assays were used to determine cell viability and DNA damage levels in cells, respectively. EdU Apopllo488 in vitro Flow Cytometry kit, propidium iodide staining, anti-γ-H2AX antibody staining, and Annexin-V apoptosis kit were subsequently used to determine DNA synthesis rates, cell cycle distribution, count of DNA double-strand breaks, and levels of apoptotic cells. Results:
miR-130b levels increased at exponential phases of the growth of cervical cancer cells but reduced at stationary phases. The methylation of a prominent CpG island near the transcript start site suppressed the miR-130b gene expression. MiR-130b increased cell viability, promoted both DNA synthesis and G1 to S phase transition of the cells at exponential phases, but reduced cell viability accompanied by accumulations of DNA breaks and augmentations in apoptosis rates of the cells in stationary phases by targeting cyclin-dependent kinase inhibitor 1A mRNA. Conclusion:
miR-130b promoted the growth of cervical cancer cells during the exponential phase, whereas it impaired the survival of cells during stationary phases.
Collapse
Affiliation(s)
- Yanli Wang
- Department of Clinical Laboratory, Tianjin Hospital of ITCWM Nankai Hospital, Tianjin, China
| | - Lei Yang
- Department of Clinical Laboratory, Tianjin Hospital of ITCWM Nankai Hospital, Tianjin, China
| | - Caihong Fan
- The First Central Clinical College of Tianjin Medical University, Tianjin Medical University, Tianjin, China
| | - Hong Mu
- Department of Clinical Laboratory, Tianjin First Center Hospital, Tianjin, China
| | - Munan Han
- First Clinical Medical College, Nanjing Medical University, Nanjing, China
| | - Tao Liu
- a; eKey Laboratory for Critical Care Medicine of the Ministry of Health, Tianjin, China
| | - Lili Xie
- The First Central Clinical College of Tianjin Medical University, Tianjin Medical University, Tianjin, China
| | - Qiang Gao
- Department of Clinical Laboratory, Tianjin First Center Hospital, Tianjin, China
| |
Collapse
|
21
|
Wang P, Hu Y, Qu P, Zhao Y, Liu J, Zhao J, Kong B. Protein tyrosine phosphatase receptor type Z1 inhibits the cisplatin resistance of ovarian cancer by regulating PI3K/AKT/mTOR signal pathway. Bioengineered 2022; 13:1931-1941. [PMID: 35001804 PMCID: PMC8805848 DOI: 10.1080/21655979.2021.2022268] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Accepted: 12/20/2021] [Indexed: 12/30/2022] Open
Abstract
Most patients with ovarian cancer (OC) get remission after undergoing cytoreductive surgery and platinum-based standard chemotherapy, but more than 50% of patients with advanced OC relapse within the first 5 years after treatment and develop resistance to standard chemotherapy. The production of medicinal properties is the main reason for the poor prognosis and high mortality of OC patients. Cisplatin (DDP) resistance is a major cause for poor prognosis of OC patients. PTPRZ1 can regulate the growth and apoptosis of ovarian cancer cells, while the molecular mechanism remains unknown. This study was designed to investigate the roles of PTPRZ1 in DDP-resistant OC cells and possible mechanism. PTPRZ1 expression in OC tissues and normal tissues was analyzed by GEPIA database and verified by Real-time Quantitative Reverse Transcription PCR (RT-PCR) assay. PTPRZ1 expression in normal ovarian cancer cells and DDP-resistant OC cells was also analyzed. Subsequently, RT-PCR, Western blot, MTT experiment and flow cytometry were used to assess the effects of PTPRZ1-PI3K/AKT/mTOR regulating axis on DDP resistance of OC. PTPRZ1 expression was abnormally low in OC tissues, and notably reduced in DDP-resistant OC cells. MTT experiment and flow cytometer indicated that overexpression of PTPRZ1 enhanced the DDP sensitivity of OC cells and promoted the cell apoptosis. Moreover, the results of our research showed that PTPRZ1 might exert its biological effects through blocking PI3K/AKT/mTOR pathway. PTPRZ1 overexpression inhibitied OC tumor growth and resistance to DDP in vivo. Overall, PTPRZ1 might suppress the DDP resistance of OC and induce the cytotoxicity by blocking PI3K/AKT/mTOR pathway.
Collapse
Affiliation(s)
- Peng Wang
- Department of Gynecology Oncology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
- Department of Gynecology Oncology, Tianjin Central Hospital of Obstetrics and Gynecology, Tianjin, China
| | - Yuanjing Hu
- Department of Gynecology Oncology, Tianjin Central Hospital of Obstetrics and Gynecology, Tianjin, China
| | - Pengpeng Qu
- Department of Gynecology Oncology, Tianjin Central Hospital of Obstetrics and Gynecology, Tianjin, China
| | - Ying Zhao
- Department of Gynecology Oncology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Jing Liu
- Department of Gynecology Oncology, Tianjin Central Hospital of Obstetrics and Gynecology, Tianjin, China
| | - Jianguo Zhao
- Department of Gynecology Oncology, Tianjin Central Hospital of Obstetrics and Gynecology, Tianjin, China
| | - Beihua Kong
- Department of Gynecology Oncology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| |
Collapse
|
22
|
Park J, Park JS, Huang CH, Jo A, Cook K, Wang R, Lin HY, Deun JV, Li H, Min J, Wang L, Yoon G, Carter BS, Balaj L, Choi GS, Castro CM, Weissleder R, Lee H. An integrated magneto-electrochemical device for the rapid profiling of tumour extracellular vesicles from blood plasma. Nat Biomed Eng 2021; 5:678-689. [PMID: 34183802 PMCID: PMC8437135 DOI: 10.1038/s41551-021-00752-7] [Citation(s) in RCA: 108] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2019] [Accepted: 05/18/2021] [Indexed: 02/02/2023]
Abstract
Assays for cancer diagnosis via the analysis of biomarkers on circulating extracellular vesicles (EVs) typically have lengthy sample workups, limited throughput or insufficient sensitivity, or do not use clinically validated biomarkers. Here we report the development and performance of a 96-well assay that integrates the enrichment of EVs by antibody-coated magnetic beads and the electrochemical detection, in less than one hour of total assay time, of EV-bound proteins after enzymatic amplification. By using the assay with a combination of antibodies for clinically relevant tumour biomarkers (EGFR, EpCAM, CD24 and GPA33) of colorectal cancer (CRC), we classified plasma samples from 102 patients with CRC and 40 non-CRC controls with accuracies of more than 96%, prospectively assessed a cohort of 90 patients, for whom the burden of tumour EVs was predictive of five-year disease-free survival, and longitudinally analysed plasma from 11 patients, for whom the EV burden declined after surgery and increased on relapse. Rapid assays for the detection of combinations of tumour biomarkers in plasma EVs may aid cancer detection and patient monitoring.
Collapse
Affiliation(s)
- Jongmin Park
- Center for Systems Biology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA,Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA,Department of Chemistry, Kangwon National University, Chuncheon 24341, Korea
| | - Jun Seok Park
- Center for Systems Biology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA,Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA,Department of Surgery, Kyungpook National University Hospital, Kyungpook National University School of Medicine, Daegu, Korea
| | - Chen-Han Huang
- Center for Systems Biology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA,Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA,Department of Biomedical Sciences and Engineering, National Central University, Taoyuan 32001, Taiwan
| | - Ala Jo
- Center for Systems Biology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA,Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - Kaitlyn Cook
- Department of Biostatistics, Harvard T. H. Chan School of Public Health, Boston, MA 02115, USA
| | - Rui Wang
- Department of Biostatistics, Harvard T. H. Chan School of Public Health, Boston, MA 02115, USA,Department of Population Medicine, Harvard Pilgrim Health Care Institute, Harvard Medical School, Boston, MA 02215, USA
| | - Hsing-Ying Lin
- Center for Systems Biology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA,Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - Jan Van Deun
- Center for Systems Biology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA,Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - Huiyan Li
- Center for Systems Biology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA,Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - Jouha Min
- Center for Systems Biology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA,Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - Lan Wang
- Department of Neurosurgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - Ghilsuk Yoon
- Department of Pathology, School of Medicine, Kyungpook National University, Daegu 41944, Korea
| | - Bob S. Carter
- Department of Neurosurgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - Leonora Balaj
- Department of Neurosurgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - Gyu-Seog Choi
- Department of Surgery, Kyungpook National University Hospital, Kyungpook National University School of Medicine, Daegu, Korea
| | - Cesar M. Castro
- Center for Systems Biology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA,Cancer Center, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - Ralph Weissleder
- Center for Systems Biology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA,Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA,Department of Systems Biology, Harvard Medical School, Boston, MA 02115, USA,Correspondence and requests for materials should be addressed to ;
| | - Hakho Lee
- Center for Systems Biology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA,Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA,Correspondence and requests for materials should be addressed to ;
| |
Collapse
|
23
|
Identification of a 5-Gene-Based Scoring System by WGCNA and LASSO to Predict Prognosis for Rectal Cancer Patients. ACTA ACUST UNITED AC 2021; 2021:6697407. [PMID: 33833937 PMCID: PMC8012151 DOI: 10.1155/2021/6697407] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2020] [Revised: 01/29/2021] [Accepted: 02/27/2021] [Indexed: 12/17/2022]
Abstract
Background Although accumulating evidence suggested that a molecular signature panel may be more effective for the prognosis prediction than routine clinical characteristics, current studies mainly focused on colorectal or colon cancers. No reports specifically focused on the signature panel for rectal cancers (RC). Our present study was aimed at developing a novel prognostic signature panel for RC. Methods Sequencing (or microarray) data and clinicopathological details of patients with RC were retrieved from The Cancer Genome Atlas (TCGA-READ) or the Gene Expression Omnibus (GSE123390, GSE56699) database. A weighted gene coexpression network was used to identify RC-related modules. The least absolute shrinkage and selection operator analysis was performed to screen the prognostic signature panel. The prognostic performance of the risk score was evaluated by survival curve analyses. Functions of prognostic genes were predicted based on the interaction proteins and the correlation with tumor-infiltrating immune cells. The Human Protein Atlas (HPA) tool was utilized to validate the protein expression levels. Results A total of 247 differentially expressed genes (DEGs) were commonly identified using TCGA and GSE123390 datasets. Brown and yellow modules (including 77 DEGs) were identified to be preserved for RC. Five DEGs (ASB2, GPR15, PRPH, RNASE7, and TCL1A) in these two modules constituted the optimal prognosis signature panel. Kaplan-Meier curve analysis showed that patients in the high-risk group had a poorer prognosis than those in the low-risk group. Receiver operating characteristic (ROC) curve analysis demonstrated that this risk score had high predictive accuracy for unfavorable prognosis, with the area under the ROC curve of 0.915 and 0.827 for TCGA and GSE56699 datasets, respectively. This five-mRNA classifier was an independent prognostic factor. Its predictive accuracy was also higher than all clinical factor models. A prognostic nomogram was developed by integrating the risk score and clinical factors, which showed the highest prognostic power. ASB2, PRPH, and GPR15/TCL1A were predicted to function by interacting with CASQ2/PDK4/EPHA67, PTN, and CXCL12, respectively. TCL1A and GPR15 influenced the infiltration levels of B cells and dendritic cells, while the expression of PRPH was positively associated with the abundance of macrophages. HPA analysis supported the downregulation of PRPH, RNASE7, CASQ2, EPHA6, and PDK4 in RC compared with normal controls. Conclusion Our immune-related signature panel may be a promising prognostic indicator for RC.
Collapse
|
24
|
Huang P, Tu B, Liao HJ, Huang FZ, Li ZZ, Zhu KY, Dai F, Liu HZ, Zhang TY, Sun CZ. Elevation of plasma tRNA fragments as a promising biomarker for liver fibrosis in nonalcoholic fatty liver disease. Sci Rep 2021; 11:5886. [PMID: 33723340 PMCID: PMC7961013 DOI: 10.1038/s41598-021-85421-0] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Accepted: 03/02/2021] [Indexed: 02/07/2023] Open
Abstract
Fibrotic tissue remodelling in nonalcoholic fatty liver disease (NAFLD) will probably emerge as the leading cause of end-stage liver disease in the coming decades, but the ability to diagnose liver fibrosis in NAFLD patients noninvasively is limited. The abnormal expression of tRNA-derived small RNA (tsRNA) in plasma provides a novel idea for noninvasive diagnosis of various diseases, however, the relationship between tsRNAs and NAFLD is still unknown. Here, we took advantage of small RNA-Seq technology to profile tsRNAs in NAFLD patients and found the ubiquitous presence of hepatic tsRNAs secreted into circulating blood. Verification in a cohort of 114 patients with NAFLD and 42 patients without NAFLD revealed that three tsRNAs (tRF-Val-CAC-005, tiRNA-His-GTG-001, and tRF-Ala-CGC-006) were significantly elevated in the plasma of NAFLD patients, and the expression level are associated with NAFLD activity score (calculated from 0 to 8) and fibrosis stage (scored from 0 to 4). In mouse models, we further found that increased plasma levels of these three tsRNAs were positively correlated with the degree of liver fibrosis. Our study potentially identifies a new class of NAFLD biomarkers and reveal the possible existence of tsRNAs in the blood that can be used to predict fibrogenesis risk in patients diagnosed with NAFLD.
Collapse
Affiliation(s)
- Peng Huang
- Department of General Surgery, Xiangya Hospital Central South University, No. 87 Xiangya Road, Changsha, 410008, Hunan, People's Republic of China
| | - Biao Tu
- Department of General Surgery, Central South University Third Xiangya Hospital, No. 138 Tongzipo Road, Changsha, 410013, Hunan, People's Republic of China
| | - Hui-Jun Liao
- Department of General Surgery, Chenzhou No. 1 People's Hospital, No. 102 Luojiajing Road, Chenzhou, 423000, Hunan, People's Republic of China
| | - Fei-Zhou Huang
- Department of General Surgery, Central South University Third Xiangya Hospital, No. 138 Tongzipo Road, Changsha, 410013, Hunan, People's Republic of China
| | - Zhen-Zhou Li
- Emergency Department, Central South University Third Xiangya Hospital, No. 138 Tongzipo Road, Changsha, 410013, Hunan, People's Republic of China
| | - Kuang-Ye Zhu
- Department of General Surgery, Central South University Third Xiangya Hospital, No. 138 Tongzipo Road, Changsha, 410013, Hunan, People's Republic of China
| | - Feng Dai
- Department of General Surgery, Central South University Third Xiangya Hospital, No. 138 Tongzipo Road, Changsha, 410013, Hunan, People's Republic of China
| | - Huai-Zheng Liu
- Emergency Department, Central South University Third Xiangya Hospital, No. 138 Tongzipo Road, Changsha, 410013, Hunan, People's Republic of China
| | - Tian-Yi Zhang
- Emergency Department, Central South University Third Xiangya Hospital, No. 138 Tongzipo Road, Changsha, 410013, Hunan, People's Republic of China
| | - Chuan-Zheng Sun
- Emergency Department, Central South University Third Xiangya Hospital, No. 138 Tongzipo Road, Changsha, 410013, Hunan, People's Republic of China.
| |
Collapse
|
25
|
Kalhori MR, Khodayari H, Khodayari S, Vesovic M, Jackson G, Farzaei MH, Bishayee A. Regulation of Long Non-Coding RNAs by Plant Secondary Metabolites: A Novel Anticancer Therapeutic Approach. Cancers (Basel) 2021; 13:cancers13061274. [PMID: 33805687 PMCID: PMC8001769 DOI: 10.3390/cancers13061274] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2021] [Revised: 03/05/2021] [Accepted: 03/09/2021] [Indexed: 02/07/2023] Open
Abstract
Simple Summary Cancer is caused by the rapid and uncontrolled growth of cells that eventually lead to tumor formation. Genetic and epigenetic alterations are among the most critical factors in the onset of carcinoma. Phytochemicals are a group of natural compounds that play an essential role in cancer prevention and treatment. Long non-coding RNAs (lncRNAs) are potential therapeutic targets of bioactive phytochemicals, and these compounds could regulate the expression of lncRNAs directly and indirectly. Here, we critically evaluate in vitro and in vivo anticancer effects of phytochemicals in numerous human cancers via regulation of lncRNA expression and their downstream target genes. Abstract Long non-coding RNAs (lncRNAs) are a class of non-coding RNAs that play an essential role in various cellular activities, such as differentiation, proliferation, and apoptosis. Dysregulation of lncRNAs serves a fundamental role in the progression and initiation of various diseases, including cancer. Precision medicine is a suitable and optimal treatment method for cancer so that based on each patient’s genetic content, a specific treatment or drug is prescribed. The rapid advancement of science and technology in recent years has led to many successes in this particular treatment. Phytochemicals are a group of natural compounds extracted from fruits, vegetables, and plants. Through the downregulation of oncogenic lncRNAs or upregulation of tumor suppressor lncRNAs, these bioactive compounds can inhibit metastasis, proliferation, invasion, migration, and cancer cells. These natural products can be a novel and alternative strategy for cancer treatment and improve tumor cells’ sensitivity to standard adjuvant therapies. This review will discuss the antineoplastic effects of bioactive plant secondary metabolites (phytochemicals) via regulation of expression of lncRNAs in various human cancers and their potential for the treatment and prevention of human cancers.
Collapse
Affiliation(s)
- Mohammad Reza Kalhori
- Medical Biology Research Center, Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah 6714415185, Iran;
| | - Hamid Khodayari
- International Center for Personalized Medicine, 40235 Düsseldorf, Germany; (H.K.); (S.K.)
- Breast Disease Research Center, Tehran University of Medical Sciences, Tehran 1419733141, Iran
| | - Saeed Khodayari
- International Center for Personalized Medicine, 40235 Düsseldorf, Germany; (H.K.); (S.K.)
- Breast Disease Research Center, Tehran University of Medical Sciences, Tehran 1419733141, Iran
| | - Miko Vesovic
- Department of Mathematics, Statistics, and Computer Science, University of Illinois at Chicago, Chicago, IL 60607, USA;
| | - Gloria Jackson
- Lake Erie College of Osteopathic Medicine, Bradenton, FL 34211, USA;
| | - Mohammad Hosein Farzaei
- Medical Technology Research Center, Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah 6718874414, Iran
- Correspondence: (M.H.F.); or (A.B.)
| | - Anupam Bishayee
- Lake Erie College of Osteopathic Medicine, Bradenton, FL 34211, USA;
- Correspondence: (M.H.F.); or (A.B.)
| |
Collapse
|
26
|
Dai H, Sheng X, Sha R, Peng J, Yang F, Zhou L, Lin Y, Xu Y, Zhang S, Yin W, Lu J. Linc00665 Can Predict the Response to Cisplatin-Paclitaxel Neoadjuvant Chemotherapy for Breast Cancer Patients. Front Oncol 2021; 11:604319. [PMID: 33738251 PMCID: PMC7961084 DOI: 10.3389/fonc.2021.604319] [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: 09/09/2020] [Accepted: 01/15/2021] [Indexed: 11/29/2022] Open
Abstract
Objective Linc00665 is a novel long non-coding RNA that can promote the progression of breast cancer, but its value in predicting the efficacy of neoadjuvant chemotherapy (NAC) for breast cancer has not been reported. We aim to analyze the correlation between Linc00665 expression and pathological complete response (pCR) in breast cancer patients. Materials and Methods The present study examined the predictive role of Linc00665 expression in pCR after NAC using both univariate and multivariate logistic regression analyses. Receiver operating characteristic (ROC) curve and area under curve (AUC) were utilized to evaluate the performance of Linc00665 in predicting pCR. The Kyoto Encyclopedia of Gene and Genome (KEGG) analysis and Gene Set Enrichment Analysis (GSEA) were also conducted to determine the biological processes where Linc00665 may participate in. Results The present study study totally enrolled 102 breast cancer patients. The univariate analysis showed that Linc00665 level, human epidermal growth factor receptor 2 (HER2) status and hormone receptor (HR) status were correlated with pCR. The multivariate analysis showed that Linc00665 expression was an independent predictor of pCR (OR = 0.351, 95% CI: 0.125–0.936, P = 0.040), especially in patients with HR-positive/HER2-negative subtype (OR = 0.272, 95% CI: 0.104–0.664, P = 0.005). The KEGG analysis indicated that Linc00665 may be involved in drug metabolism. The GSEA analysis revealed that Linc00665 is correlated to DNA damage repair. Conclusion Linc00665 may be a potential novel predictive biomarker for breast cancer in NAC, especially for HR-positive/HER2-negative patients.
Collapse
Affiliation(s)
- Huijuan Dai
- Department of Breast Surgery, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Xiaonan Sheng
- Department of Breast Surgery, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Rui Sha
- Department of Breast Surgery, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Jing Peng
- Department of Breast Surgery, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Fan Yang
- Department of Breast Surgery, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Liheng Zhou
- Department of Breast Surgery, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Yanping Lin
- Department of Breast Surgery, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Yaqian Xu
- Department of Breast Surgery, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Shan Zhang
- Department of Breast Surgery, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Wenjin Yin
- Department of Breast Surgery, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Jinsong Lu
- Department of Breast Surgery, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| |
Collapse
|
27
|
Kang TZE, Zhu L, Yang D, Ding D, Zhu X, Wan YCE, Liu J, Ramakrishnan S, Chan LL, Chan SY, Wang X, Gan H, Han J, Ishibashi T, Li Q, Chan KM. The elevated transcription of ADAM19 by the oncohistone H2BE76K contributes to oncogenic properties in breast cancer. J Biol Chem 2021; 296:100374. [PMID: 33548228 PMCID: PMC7949156 DOI: 10.1016/j.jbc.2021.100374] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2020] [Revised: 01/26/2021] [Accepted: 02/02/2021] [Indexed: 02/05/2023] Open
Abstract
The recent discovery of the cancer-associated E76K mutation in histone H2B (H2BE76-to-K) in several types of cancers revealed a new class of oncohistone. H2BE76K weakens the stability of histone octamers, alters gene expression, and promotes colony formation. However, the mechanism linking the H2BE76K mutation to cancer development remains largely unknown. In this study, we knock in the H2BE76K mutation in MDA-MB-231 breast cancer cells using CRISPR/Cas9 and show that the E76K mutant histone H2B preferentially localizes to genic regions. Interestingly, genes upregulated in the H2BE76K mutant cells are enriched for the E76K mutant H2B and are involved in cell adhesion and proliferation pathways. We focused on one H2BE76K target gene, ADAM19 (a disintegrin and metalloproteinase-domain-containing protein 19), a gene highly expressed in various human cancers including breast invasive carcinoma, and demonstrate that H2BE76K directly promotes ADAM19 transcription by facilitating efficient transcription along the gene body. ADAM19 depletion reduced the colony formation ability of the H2BE76K mutant cells, whereas wild-type MDA-MB-231 cells overexpressing ADAM19 mimics the colony formation phenotype of the H2BE76K mutant cells. Collectively, our data demonstrate the mechanism by which H2BE76K deregulates the expression of genes that control oncogenic properties through a combined effect of its specific genomic localization and nucleosome destabilization effect.
Collapse
Affiliation(s)
- Tze Zhen Evangeline Kang
- Department of Biomedical Sciences, City University of Hong Kong, Hong Kong, China; Key Laboratory of Biochip Technology, Biotech and Health Centre, Shenzhen Research Institute of City University of Hong Kong, Shenzhen, China
| | - Lina Zhu
- Department of Biomedical Sciences, City University of Hong Kong, Hong Kong, China; Key Laboratory of Biochip Technology, Biotech and Health Centre, Shenzhen Research Institute of City University of Hong Kong, Shenzhen, China
| | - Du Yang
- State Key Laboratory of Protein and Plant Gene Research, School of Life Sciences and Peking-Tsinghua Center for Life Sciences, Peking University, Peking, China
| | - Dongbo Ding
- Division of Life Science, Hong Kong University of Science and Technology, Hong Kong, China
| | - Xiaoxuan Zhu
- Department of Biomedical Sciences, City University of Hong Kong, Hong Kong, China; Key Laboratory of Biochip Technology, Biotech and Health Centre, Shenzhen Research Institute of City University of Hong Kong, Shenzhen, China
| | - Yi Ching Esther Wan
- Department of Biomedical Sciences, City University of Hong Kong, Hong Kong, China; Key Laboratory of Biochip Technology, Biotech and Health Centre, Shenzhen Research Institute of City University of Hong Kong, Shenzhen, China
| | - Jiaxian Liu
- Department of Biomedical Sciences, City University of Hong Kong, Hong Kong, China; Key Laboratory of Biochip Technology, Biotech and Health Centre, Shenzhen Research Institute of City University of Hong Kong, Shenzhen, China
| | - Saravanan Ramakrishnan
- Department of Biomedical Sciences, City University of Hong Kong, Hong Kong, China; Key Laboratory of Biochip Technology, Biotech and Health Centre, Shenzhen Research Institute of City University of Hong Kong, Shenzhen, China
| | - Landon Long Chan
- Department of Oncology, Princess Margaret Hospital, Hong Kong, China
| | - Siu Yuen Chan
- Department of Paediatrics and Adolescent Medicine, The University of Hong Kong, Hong Kong, China
| | - Xin Wang
- Department of Biomedical Sciences, City University of Hong Kong, Hong Kong, China; Key Laboratory of Biochip Technology, Biotech and Health Centre, Shenzhen Research Institute of City University of Hong Kong, Shenzhen, China
| | - Haiyun Gan
- Guangdong Provincial Key Laboratory of Synthetic Genomics, CAS Key Laboratory of Quantitative Engineering Biology and Shenzhen Key Laboratory of Synthetic Genomics, Shenzhen Institute of Synthetic Biology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Junhong Han
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University, Sichuan, China
| | - Toyotaka Ishibashi
- Division of Life Science, Hong Kong University of Science and Technology, Hong Kong, China
| | - Qing Li
- State Key Laboratory of Protein and Plant Gene Research, School of Life Sciences and Peking-Tsinghua Center for Life Sciences, Peking University, Peking, China
| | - Kui Ming Chan
- Department of Biomedical Sciences, City University of Hong Kong, Hong Kong, China; Key Laboratory of Biochip Technology, Biotech and Health Centre, Shenzhen Research Institute of City University of Hong Kong, Shenzhen, China.
| |
Collapse
|
28
|
Sun X, Tian C, Zhang H, Han K, Zhou M, Gan Z, Zhu H, Min D. Long noncoding RNA OIP5-AS1 mediates resistance to doxorubicin by regulating miR-137-3p/PTN axis in osteosarcoma. Biomed Pharmacother 2020; 128:110201. [PMID: 32460190 DOI: 10.1016/j.biopha.2020.110201] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2019] [Revised: 04/24/2020] [Accepted: 04/27/2020] [Indexed: 12/18/2022] Open
Abstract
Opa-interacting protein 5 antisense RNA1 (OIP5-AS1) has been demonstrated to facilitate proliferation, metastasis and resistance to treatments in various types of cancers. Nevertheless, the exact mechanisms underlying the roles of OIP5-AS1 in osteosarcoma(OS) drug resistance have not yet been clearly elucidated. Therefore, we sought to investigate the functional involvement of OIP5-AS1 in osteosarcoma. Our results indicated that OIP5-AS1 was dramatically up-regulated in osteosarcoma drug-resistant tissues and cells in comparison with drug-sensitive tissues and cells. Also, the knockdown of OIP5-AS1 was found to have decreased doxorubicin resistance of OS cells. Further analyses revealed that OIP5-AS1 operated as a competitor for endogenous RNA of miR-137-3p as well as regulated pleiotrophin(PTN) expression, which has been reported to be an oncogene in OS in previous research. Furthermore, the loss of miR-137-3p or alternatively, the gain of PTN, both resulted in the abolishment of the inhibitory role of OIP5-AS1 silencing the proliferative activity. Our analyses indicated and helped to determine the role of OIP5-AS1 in contributing to tumorigenesis of osteosarcoma via the miR-137-3p/PTN axis and, therefore outlining its potential for use as a therapeutic target against this cancer.
Collapse
Affiliation(s)
- Xingxing Sun
- Department of Oncology, Sixth People's Hospital East Campus Affiliated to Shanghai Jiao Tong University, Shanghai 201306, China; Shanghai University of Medicine & Health Sciences Affiliated Sixth People's Hospital East Campus, Shanghai 201306, China
| | - Cong Tian
- Department of Oncology, Sixth People's Hospital East Campus Affiliated to Shanghai Jiao Tong University, Shanghai 201306, China; Shanghai University of Medicine & Health Sciences Affiliated Sixth People's Hospital East Campus, Shanghai 201306, China
| | - Hui Zhang
- Department of Oncology, Sixth People's Hospital East Campus Affiliated to Shanghai Jiao Tong University, Shanghai 201306, China; Shanghai University of Medicine & Health Sciences Affiliated Sixth People's Hospital East Campus, Shanghai 201306, China
| | - Kun Han
- Department of Oncology, Sixth People's Hospital East Campus Affiliated to Shanghai Jiao Tong University, Shanghai 201306, China; Shanghai University of Medicine & Health Sciences Affiliated Sixth People's Hospital East Campus, Shanghai 201306, China
| | - Meixiang Zhou
- Department of Oncology, Sixth People's Hospital East Campus Affiliated to Shanghai Jiao Tong University, Shanghai 201306, China; Shanghai University of Medicine & Health Sciences Affiliated Sixth People's Hospital East Campus, Shanghai 201306, China
| | - Zhihua Gan
- Department of Oncology, Sixth People's Hospital East Campus Affiliated to Shanghai Jiao Tong University, Shanghai 201306, China; Shanghai University of Medicine & Health Sciences Affiliated Sixth People's Hospital East Campus, Shanghai 201306, China
| | - Hongling Zhu
- Department of Oncology, Sixth People's Hospital East Campus Affiliated to Shanghai Jiao Tong University, Shanghai 201306, China; Shanghai University of Medicine & Health Sciences Affiliated Sixth People's Hospital East Campus, Shanghai 201306, China
| | - Daliu Min
- Department of Oncology, Sixth People's Hospital East Campus Affiliated to Shanghai Jiao Tong University, Shanghai 201306, China; Shanghai University of Medicine & Health Sciences Affiliated Sixth People's Hospital East Campus, Shanghai 201306, China.
| |
Collapse
|
29
|
Jiang W, Xia J, Xie S, Zou R, Pan S, Wang ZW, Assaraf YG, Zhu X. Long non-coding RNAs as a determinant of cancer drug resistance: Towards the overcoming of chemoresistance via modulation of lncRNAs. Drug Resist Updat 2020; 50:100683. [DOI: 10.1016/j.drup.2020.100683] [Citation(s) in RCA: 56] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2019] [Revised: 02/18/2020] [Accepted: 02/21/2020] [Indexed: 12/11/2022]
|
30
|
Abstract
Pleiotrophin (PTN) is a potent mitogenic cytokine with a high affinity for the polysaccharide glycosaminoglycan (GAG). Although it is most strongly associated with neural development during embryogenesis and the neonatal period, its expression has also been linked to a plethora of other physiological events including cancer metastasis, angiogenesis, bone development, and inflammation. A considerable amount of research has been carried out to understand the mechanisms by which PTN regulates these events. In particular, PTN has now been shown to bind a diverse collection of receptors including many GAG-containing proteoglycans. These interactions lead to the activation of many intracellular kinases and, ultimately, activation and transformation of cells. Structural studies of PTN in complex with both GAG and domains from its non-proteoglycan receptors reveal a binding mechanism that relies on electrostatic interactions and points to PTN-induced receptor oligomerization as one of the possible ways PTN uses to control cellular functions.
Collapse
|
31
|
Xu R, Gong CX, Duan CM, Huang JC, Yang GQ, Yuan JJ, Zhang Q, Xiong XY, Yang QW. Age-Dependent Changes in the Plasma Proteome of Healthy Adults. J Nutr Health Aging 2020; 24:846-856. [PMID: 33009535 DOI: 10.1007/s12603-020-1392-6] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
OBJECTIVE Human blood plasma is a complex that communicates with most parts of the body and reflects the changes in the state of an organism. Identifying age-related biomarkers can help predict and monitor age-related physiological decline and diseases and identify new treatments for diseases. METHODS AND PARTICIPANTS In this study, TMT-LC-MS/MS was utilized to screen differentially expressed plasma proteins in 118 healthy adults of different ages. Participants were divided into three groups: 21-30 years of age (Young), 41-50 years of age (Middle) and ≥60 years of age (Old). RESULTS The number of differentially expressed proteins in the comparisons of Young vs Middle, Middle vs Old and Young vs Old were 82, 22 and 99, respectively. These proteins were involved in numerous physiological processes, such as "negative regulation of smooth muscle cell proliferation" and "blood coagulation". Moreover, when Young was compared with Middle or Old, "complement and coagulation cascades" was the top enriched pathway by KEGG pathway enrichment analysis. Functional phenotyping of the proteome demonstrated that the plasma proteomic profiles of young adults were strikingly dissimilar to those of the middle-aged or older adults. CONCLUSIONS The results of this study mapped the variation in the expression of plasma proteins and provided information about possible biomarkers/treatments for different age-related functional disorders.
Collapse
Affiliation(s)
- R Xu
- Xiaoyi Xiong and Qingwu Yang, No.183, Xinqiaozheng Street, Shapingba District, Chongqing 400037, China, Fax number: +86 23 6877 4413, (Xiaoyi Xiong) and (Qingwu Yang)
| | | | | | | | | | | | | | | | | |
Collapse
|
32
|
Mehrgou A, Ebadollahi S, Jameie B, Teimourian S. Analysis of subtype-specific and common Gene/MiRNA expression profiles of four main breast cancer subtypes using bioinformatic approach; Characterization of four genes, and two MicroRNAs with possible diagnostic and prognostic values. INFORMATICS IN MEDICINE UNLOCKED 2020. [DOI: 10.1016/j.imu.2020.100425] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
|
33
|
Liu AS, Yu HY, Yang YL, Xue FY, Chen X, Zhang Y, Zhou ZY, Zhang B, Li L, Sun CZ, Huang P, Huang JF. A Chemotherapy-Driven Increase in Mcl-1 Mediates the Effect of miR-375 on Cisplatin Resistance in Osteosarcoma Cells. Onco Targets Ther 2019; 12:11667-11677. [PMID: 32021245 PMCID: PMC6942534 DOI: 10.2147/ott.s231125] [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: 09/15/2019] [Accepted: 12/13/2019] [Indexed: 12/12/2022] Open
Abstract
Background Osteosarcoma (OS) is one of the most difficult cancers to treat due to its resistance to chemotherapy. The essential role played by Mcl-1 in promoting chemoresistance has been observed in a variety of cancers, including OS, while the underlying mechanism remains unclear. Methods We investigated the expression of Mcl-1 in 42 paired OS specimens obtained before and after adjuvant chemotherapy, and its correlation with clinicopathological characteristics. Loss and gain of function studies were performed to analyze the effects of Mcl-1 modulations on the chemosensitivity, and the mechanism involved in the deregulation of Mcl-1 in OS cells. Results In OS specimens, the expression of Mcl-1 was significantly upregulated after chemotherapy, and high Mcl-1 expression was associated with poorer overall survival and an increased recurrence rate. Furthermore, we demonstrated that chemotherapy-driven increased Mcl-1 decreased chemosensitivity by promoting tumour proliferation and inhibiting DNA damage. Moreover, Mcl-1 was found to be a direct target of miR-375 in OS cells. The knockdown of Mcl-1 phenocopied miR-375 downregulation, and the overexpression of miR-375 rescued the effects of cisplatin-induced DNA damage mediated by Mcl-1. Conclusion Our data indicated that chemotherapy-driven increase in the expression of Mcl-1 plays a critical role in chemoresistance, and the intervention of the miR-375/Mcl-1 axis may offer a novel strategy to enhance chemosensitivity in OS treatment.
Collapse
Affiliation(s)
- An-Song Liu
- Department of Anatomy and Neurobiology, School of Basic Medical Sciences, Central South University, Changsha, People's Republic of China
| | - Hai-Yang Yu
- Department of Anatomy and Neurobiology, School of Basic Medical Sciences, Central South University, Changsha, People's Republic of China
| | - Yan-Lin Yang
- Department of Oncology, Affiliated Nanhua Hospital, University of South China, Hengyang, People's Republic of China
| | - Fu-Yao Xue
- Department of Anatomy and Neurobiology, School of Basic Medical Sciences, Central South University, Changsha, People's Republic of China
| | - Xia Chen
- Department of Orthopaedics, The Second Xiangya Hospital of Central South University, Changsha, People's Republic of China
| | - Yun Zhang
- Department of Orthopaedics, The Second Xiangya Hospital of Central South University, Changsha, People's Republic of China
| | - Zi-Yu Zhou
- Department of Anatomy and Neurobiology, School of Basic Medical Sciences, Central South University, Changsha, People's Republic of China
| | - Bin Zhang
- Department of Anatomy and Neurobiology, School of Basic Medical Sciences, Central South University, Changsha, People's Republic of China
| | - Lan Li
- Department of Pathology, The Second Xiangya Hospital of Central South University, Changsha, People's Republic of China
| | - Chuan-Zheng Sun
- Emergency Department, The Third Xiangya Hospital, Central South University, Changsha, People's Republic of China
| | - Peng Huang
- Department of General Surgery, Xiangya Hospital, Central South University, Changsha, People's Republic of China
| | - Ju-Fang Huang
- Department of Anatomy and Neurobiology, School of Basic Medical Sciences, Central South University, Changsha, People's Republic of China
| |
Collapse
|
34
|
Xiao B, Cao ZY, He AY. Synthesis, characterization and anticancer activity on human osteosarcoma cells of a pentavalent antimony complex. MAIN GROUP CHEMISTRY 2019. [DOI: 10.3233/mgc-190783] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Affiliation(s)
- Bo Xiao
- Department of Orthopedics, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Zhi-Yuan Cao
- Department of Orthopedics, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Ai-Yong He
- Department of Orthopedics, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| |
Collapse
|
35
|
Lavogina D, Samuel K, Lavrits A, Meltsov A, Sõritsa D, Kadastik Ü, Peters M, Rinken A, Salumets A. Chemosensitivity and chemoresistance in endometriosis – differences for ectopic versus eutopic cells. Reprod Biomed Online 2019; 39:556-568. [DOI: 10.1016/j.rbmo.2019.05.019] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2019] [Revised: 05/22/2019] [Accepted: 05/28/2019] [Indexed: 01/19/2023]
|
36
|
Paço A, Freitas R. HOX genes as transcriptional and epigenetic regulators during tumorigenesis and their value as therapeutic targets. Epigenomics 2019; 11:1539-1552. [PMID: 31556724 DOI: 10.2217/epi-2019-0090] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Several HOX genes are aberrantly expressed in a wide range of cancers interfering with their development and resistance to treatment. This seems to be often caused by alterations in the methylation profiles of their promoters. The role of HOX gene products in cancer is highly 'tissue specific', relying ultimately on their ability to regulate oncogenes or tumor-suppressor genes, directly as transcriptional regulators or indirectly interfering with the levels of epigenetic regulators. Nowadays, different strategies have been tested the use of HOX genes as therapeutic targets for cancer diagnosis and treatment. Here, we trace the history of the research concerning the involvement of HOX genes in cancer, their connection with epigenetic regulation and their potential use as therapeutic targets.
Collapse
Affiliation(s)
- Ana Paço
- Laboratório de Microbiologia do Solo, Instituto de Ciências Agrárias e Ambientais Mediterrânicas (ICAAM), Instituto de Investigação e Formação Avançada (IIFA), Universidade de Évora, 7006-554 Évora, Portugal
| | - Renata Freitas
- I3S - Institute for Innovation & Health Research, University of Porto, 4200-135 Porto, Portugal.,IBMC - Institute for Molecular & Cell Biology, University of Porto, 4200-135 Porto, Portugal.,ICBAS - Institute of Biomedical Sciences Abel Salazar, University of Porto, 4050-313 Porto, Portugal
| |
Collapse
|
37
|
Generation of stable reporter breast and lung cancer cell lines for NF-κB activation studies. J Biotechnol 2019; 301:79-87. [DOI: 10.1016/j.jbiotec.2019.05.014] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2019] [Revised: 05/16/2019] [Accepted: 05/26/2019] [Indexed: 01/09/2023]
|
38
|
Zhao WS, Yan WP, Chen DB, Dai L, Yang YB, Kang XZ, Fu H, Chen P, Deng KJ, Wang XY, Xie XW, Chen HS, Chen KN. Genome-scale CRISPR activation screening identifies a role of ELAVL2-CDKN1A axis in paclitaxel resistance in esophageal squamous cell carcinoma. Am J Cancer Res 2019; 9:1183-1200. [PMID: 31285951 PMCID: PMC6610048] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2019] [Accepted: 05/07/2019] [Indexed: 06/09/2023] Open
Abstract
Neoadjuvant chemotherapy (NAC) may provide survival benefits for patients with advanced esophageal squamous cell carcinoma. However, tumor cells can display primary or secondary resistance to paclitaxel (PTX), a primary component of induction chemotherapy regimen. To identify genes capable of conveying PTX resistance, we performed a genome-wide CRISPR transcriptional activation library in human KYSE-180 cells. High throughput next generation sequencing was further applied to establish the phenotype-to-genotype relationship. Our highest-ranking hits are CDKN1A, TSPAN4, ELAVL2, JUNB and PAAF1. We generated evidence that esophageal tumors with high CDKN1A, ELAVL2 and TSPAN4 levels, quantified using qRT-PCR and Western blot assays, showed poorer chemotherapy response. Higher expression levels of TSPAN4 and ELAVL2 protein are independent risk factors for poor chemotherapy response in ESCC patients. We then found that overexpression of CDKN1A, ELAVL2 or TSPAN4 in ESCC cell lines significantly promoted the resistance to PTX by inhibiting cell apoptosis. Interestingly, ESCC cells overexpressed CDKN1A, ELAVL2 or TSPAN4 also acquired resistance to cisplatin (DDP). This phenomenon may be explained by cross-resistance of chemotherapy. We additionally found an association between ELAVL2 and CDKN1A, which may be regarded as the upstream and downstream factors that synergistically involved in the regulation of chemo-resistance in ESCC. Therefore, our study demonstrated that the genome-wide CRISPR activation library is a powerful strategy for the discovery of chemo-resistant genes critical for ESCC and we reported the first evidence that the ELAVL2-CDKN1A axis may be an important mechanism involved in chemo-resistance in ESCC.
Collapse
Affiliation(s)
- Wen-Si Zhao
- Department of Thoracic Surgery I, Peking University Cancer Hospital and InstituteBeijing 100142, China
| | - Wan-Pu Yan
- Department of Thoracic Surgery I, Peking University Cancer Hospital and InstituteBeijing 100142, China
| | - Dong-Bo Chen
- Peking University People’s Hospital, Peking University Hepatology Institute, Beijing Key Laboratory of Hepatitis C and Immunotherapy for Liver DiseaseBeijing 100044, China
| | - Liang Dai
- Department of Thoracic Surgery I, Peking University Cancer Hospital and InstituteBeijing 100142, China
| | - Yong-Bo Yang
- Department of Thoracic Surgery I, Peking University Cancer Hospital and InstituteBeijing 100142, China
| | - Xiao-Zheng Kang
- Department of Thoracic Surgery I, Peking University Cancer Hospital and InstituteBeijing 100142, China
| | - Hao Fu
- Department of Thoracic Surgery I, Peking University Cancer Hospital and InstituteBeijing 100142, China
| | - Pu Chen
- Peking University People’s Hospital, Peking University Hepatology Institute, Beijing Key Laboratory of Hepatitis C and Immunotherapy for Liver DiseaseBeijing 100044, China
| | - Kang-Jian Deng
- Peking University People’s Hospital, Peking University Hepatology Institute, Beijing Key Laboratory of Hepatitis C and Immunotherapy for Liver DiseaseBeijing 100044, China
| | - Xue-Yan Wang
- Peking University People’s Hospital, Peking University Hepatology Institute, Beijing Key Laboratory of Hepatitis C and Immunotherapy for Liver DiseaseBeijing 100044, China
| | - Xing-Wang Xie
- Peking University People’s Hospital, Peking University Hepatology Institute, Beijing Key Laboratory of Hepatitis C and Immunotherapy for Liver DiseaseBeijing 100044, China
| | - Hong-Song Chen
- Peking University People’s Hospital, Peking University Hepatology Institute, Beijing Key Laboratory of Hepatitis C and Immunotherapy for Liver DiseaseBeijing 100044, China
| | - Ke-Neng Chen
- Department of Thoracic Surgery I, Peking University Cancer Hospital and InstituteBeijing 100142, China
| |
Collapse
|