1
|
Ma L, Zhao X, Lu X, Shen J, Huang J. KLF4 activates LATS2 to promote cisplatin sensitivity in ovarian cancer through DNA damage. Anticancer Drugs 2025; 36:49-61. [PMID: 39365847 DOI: 10.1097/cad.0000000000001662] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/06/2024]
Abstract
We aimed to investigate the role of large tumor suppressor kinase 2 (LATS2) in cisplatin (DDP) sensitivity in ovarian cancer. Bioinformatic analysis explored LATS2 expression, pathways, and regulators. Quantitative reverse transcription-PCR measured LATS2 and KLF4 mRNA levels. Dual-luciferase and chromatin immunoprecipitation assays confirmed their binding relationship. Cell viability, half maximal inhibitory concentration (IC 50 ) values, cell cycle, and DNA damage were assessed using CCK-8, flow cytometry, and comet assays. Western blot analyzed protein expression. The effect of LATS2 on the sensitivity of ovarian cancer to DDP was verified in vivo . LATS2 and KLF4 were downregulated in ovarian cancer, with LATS2 enriched in cell cycle, DNA replication, and mismatch repair pathways. KLF4, an upstream regulator of LATS2, bound to its promoter. Overexpressing LATS2 increased G1-phase cells, reduced cell viability and IC 50 values, and induced DNA damage. Silencing KLF4 alone showed the opposite effect on LATS2 overexpression. Knocking out LATS2 reversed the effects of KLF4 overexpression on cell viability, cell cycle, IC 50 values, and DNA damage in ovarian cancer cells. Inhibiting LATS2 inactivated the Hippo-YAP signaling pathway. In vivo experiments showed that overexpression of LATS2 enhanced the sensitivity of ovarian cancer to DDP. KLF4 activates LATS2 via DNA damage to enhance DDP sensitivity in ovarian cancer, providing a potential target for improving treatment outcomes.
Collapse
Affiliation(s)
| | | | | | | | - Jiankang Huang
- Department of Thyroid and Breast Surgery, Anhui No. 2 Provincial People's Hospital, Hefei, China
| |
Collapse
|
2
|
Wilczyński M, Wilczyński J, Nowak M. MiRNAs as Regulators of Immune Cells in the Tumor Microenvironment of Ovarian Cancer. Cells 2024; 13:1343. [PMID: 39195233 PMCID: PMC11352322 DOI: 10.3390/cells13161343] [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: 06/24/2024] [Revised: 07/25/2024] [Accepted: 08/06/2024] [Indexed: 08/29/2024] Open
Abstract
Ovarian cancer is one of the leading causes of cancer deaths among women. There is an ongoing need to develop new biomarkers and targeted therapies to improve patient outcomes. One of the most critical research areas in ovarian cancer is identifying tumor microenvironment (TME) functions. TME consists of tumor-infiltrating immune cells, matrix, endothelial cells, pericytes, fibroblasts, and other stromal cells. Tumor invasion and growth depend on the multifactorial crosstalk between tumor cells and immune cells belonging to the TME. MiRNAs, which belong to non-coding RNAs that post-transcriptionally control the expression of target genes, regulate immune responses within the TME, shaping the landscape of the intrinsic environment of tumor cells. Aberrant expression of miRNAs may lead to the pathological dysfunction of signaling pathways or cancer cell-regulatory factors. Cell-to-cell communication between infiltrating immune cells and the tumor may depend on exosomes containing multiple miRNAs. MiRNAs may exert both immunosuppressive and immunoreactive responses, which may cause cancer cell elimination or survival. In this review, we highlighted recent advances in the field of miRNAs shaping the landscape of immune cells in the TME.
Collapse
Affiliation(s)
- Miłosz Wilczyński
- Department of Operative Gynecology, Endoscopy and Gynecologic Oncology, Polish Mother’s Health Center-Research Institute, 281/289 Rzgowska Str., 93-338 Lodz, Poland
| | - Jacek Wilczyński
- Department of Gynecological Surgery and Gynecological Oncology, Medical University of Lodz, 4 Kosciuszki Str., 90-419 Lodz, Poland;
| | - Marek Nowak
- Department of Operative Gynecology and Gynecologic Oncology, Polish Mother’s Health Center-Research Institute, 281/289 Rzgowska Str., 93-338 Lodz, Poland;
| |
Collapse
|
3
|
Gou Z, Li J, Liu J, Yang N. The hidden messengers: cancer associated fibroblasts-derived exosomal miRNAs as key regulators of cancer malignancy. Front Cell Dev Biol 2024; 12:1378302. [PMID: 38694824 PMCID: PMC11061421 DOI: 10.3389/fcell.2024.1378302] [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: 01/29/2024] [Accepted: 04/08/2024] [Indexed: 05/04/2024] Open
Abstract
Cancer-associated fibroblasts (CAFs), a class of stromal cells in the tumor microenvironment (TME), play a key role in controlling cancer cell invasion and metastasis, immune evasion, angiogenesis, and resistance to chemotherapy. CAFs mediate their activities by secreting soluble chemicals, releasing exosomes, and altering the extracellular matrix (ECM). Exosomes contain various biomolecules, such as nucleic acids, lipids, and proteins. microRNA (miRNA), a 22-26 nucleotide non-coding RNA, can regulate the cellular transcription processes. Studies have shown that miRNA-loaded exosomes secreted by CAFs engage in various regulatory communication networks with other TME constituents. This study focused on the roles of CAF-derived exosomal miRNAs in generating cancer malignant characteristics, including immune modulation, tumor growth, migration and invasion, epithelial-mesenchymal transition (EMT), and treatment resistance. This study thoroughly examines miRNA's dual regulatory roles in promoting and suppressing cancer. Thus, changes in the CAF-derived exosomal miRNAs can be used as biomarkers for the diagnosis and prognosis of patients, and their specificity can be used to develop newer therapies. This review also discusses the pressing problems that require immediate attention, aiming to inspire researchers to explore more novel avenues in this field.
Collapse
Affiliation(s)
- Zixuan Gou
- Bethune First Clinical School of Medicine, The First Hospital of Jilin University, Changchun, China
| | - Jiannan Li
- Department of General Surgery, The Second Hospital of Jilin University, Changchun, China
| | - Jianming Liu
- Department of Otolaryngology Head and Neck Surgery, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Na Yang
- Department of Clinical Pharmacy, The First Hospital of Jilin University, Changchun, China
| |
Collapse
|
4
|
Wilkinson AN, Chen R, Coleborn E, Neilson T, Le K, Bhavsar C, Wang Y, Atluri S, Irgam G, Wong K, Yang D, Steptoe R, Wu SY. Let-7i enhances anti-tumour immunity and suppresses ovarian tumour growth. Cancer Immunol Immunother 2024; 73:80. [PMID: 38554167 PMCID: PMC10981620 DOI: 10.1007/s00262-024-03674-w] [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: 01/31/2024] [Accepted: 03/08/2024] [Indexed: 04/01/2024]
Abstract
Cancer immunotherapy has seen significant success in the last decade for cancer management by enhancing endogenous cancer immunity. However, immunotherapies developed thus far have seen limited success in the majority of high-grade serous carcinoma (HGSC) ovarian cancer patients. This is largely due to the highly immunosuppressive tumour microenvironment of HGSC and late-stage identification. Thus, novel treatment interventions are needed to overcome this immunosuppression and complement existing immunotherapies. Here, we have identified through analysis of > 600 human HGSC tumours a critical role for Let-7i in modulating the tumoural immune network. Tumoural expression of Let-7i had high positive correlation with anti-cancer immune signatures in HGSC patients. Confirming this role, enforced Let-7i expression in murine HGSC tumours resulted in a significant decrease in tumour burden with a significant increase in tumour T cell numbers in tumours. In concert with the improved tumoural immunity, Let-7i treatment also significantly increased CD86 expression in antigen presenting cells (APCs) in the draining lymph nodes, indicating enhanced APC activity. Collectively, our findings highlight an important role of Let-7i in anti-tumour immunity and its potential use for inducing an anti-tumour effect in HGSC.
Collapse
Affiliation(s)
- Andrew N Wilkinson
- School of Biomedical Sciences, The University of Queensland, Brisbane, QLD, 4072, Australia
| | - Rui Chen
- School of Biomedical Sciences, The University of Queensland, Brisbane, QLD, 4072, Australia
| | - Elaina Coleborn
- School of Biomedical Sciences, The University of Queensland, Brisbane, QLD, 4072, Australia
| | - Trent Neilson
- School of Biomedical Sciences, The University of Queensland, Brisbane, QLD, 4072, Australia
| | - Khang Le
- School of Biomedical Sciences, The University of Queensland, Brisbane, QLD, 4072, Australia
| | - Chintan Bhavsar
- School of Biomedical Sciences, The University of Queensland, Brisbane, QLD, 4072, Australia
| | - Yue Wang
- Department of Pharmaceutical Sciences, University of Pittsburgh, Pittsburgh, PA, 15261, USA
| | - Sharat Atluri
- School of Biomedical Sciences, The University of Queensland, Brisbane, QLD, 4072, Australia
| | - Gowri Irgam
- School of Biomedical Sciences, The University of Queensland, Brisbane, QLD, 4072, Australia
| | - Kiefer Wong
- School of Biomedical Sciences, The University of Queensland, Brisbane, QLD, 4072, Australia
| | - Da Yang
- Department of Pharmaceutical Sciences, University of Pittsburgh, Pittsburgh, PA, 15261, USA
| | - Raymond Steptoe
- Frazer Institute, University of Queensland, Brisbane, QLD, 4102, Australia
| | - Sherry Y Wu
- School of Biomedical Sciences, The University of Queensland, Brisbane, QLD, 4072, Australia.
| |
Collapse
|
5
|
Gao Y, Gong Y, Lu J, Hao H, Shi X. Targeting YAP1 to improve the efficacy of immune checkpoint inhibitors in liver cancer: mechanism and strategy. Front Immunol 2024; 15:1377722. [PMID: 38550587 PMCID: PMC10972981 DOI: 10.3389/fimmu.2024.1377722] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2024] [Accepted: 03/04/2024] [Indexed: 04/02/2024] Open
Abstract
Liver cancer is the third leading of tumor death, including hepatocellular carcinoma (HCC) and intrahepatic cholangiocarcinoma (ICC). Immune checkpoint inhibitors (ICIs) are yielding much for sufferers to hope for patients, but only some patients with advanced liver tumor respond. Recent research showed that tumor microenvironment (TME) is critical for the effectiveness of ICIs in advanced liver tumor. Meanwhile, metabolic reprogramming of liver tumor leads to immunosuppression in TME. These suggest that regulating the abnormal metabolism of liver tumor cells and firing up TME to turn "cold tumor" into "hot tumor" are potential strategies to improve the therapeutic effect of ICIs in liver tumor. Previous studies have found that YAP1 is a potential target to improve the efficacy of anti-PD-1 in HCC. Here, we review that YAP1 promotes immunosuppression of TME, mainly due to the overstimulation of cytokines in TME by YAP1. Subsequently, we studied the effects of YAP1 on metabolic reprogramming in liver tumor cells, including glycolysis, gluconeogenesis, lipid metabolism, arachidonic acid metabolism, and amino acid metabolism. Lastly, we summarized the existing drugs targeting YAP1 in the treatment of liver tumor, including some medicines from natural sources, which have the potential to improve the efficacy of ICIs in the treatment of liver tumor. This review contributed to the application of targeted YAP1 for combined therapy with ICIs in liver tumor patients.
Collapse
Affiliation(s)
- Yuting Gao
- Laboratory of Integrated Medicine Tumor Immunology, Shanxi University of Chinese Medicine, Taiyuan, China
| | - Yi Gong
- Laboratory of Integrated Medicine Tumor Immunology, Shanxi University of Chinese Medicine, Taiyuan, China
| | - Junlan Lu
- Laboratory of Integrated Medicine Tumor Immunology, Shanxi University of Chinese Medicine, Taiyuan, China
| | - Huiqin Hao
- Chinese Medicine Gene Expression Regulation Laboratory, State Administration of Traditional Chinese Medicine, Shanxi University of Chinese Medicine, Taiyuan, China
- Basic Laboratory of Integrated Traditional Chinese and Western, Shanxi University of Chinese Medicine, Taiyuan, China
| | - Xinli Shi
- Laboratory of Integrated Medicine Tumor Immunology, Shanxi University of Chinese Medicine, Taiyuan, China
| |
Collapse
|
6
|
Hou Y, Zhao X, Nie X. Enhancing the therapeutic efficacy of NK cells in the treatment of ovarian cancer (Review). Oncol Rep 2024; 51:50. [PMID: 38299257 PMCID: PMC10851334 DOI: 10.3892/or.2024.8709] [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] [Accepted: 01/03/2024] [Indexed: 02/02/2024] Open
Abstract
Ovarian cancer is a prevalent gynecological malignancy associated with a high mortality rate and a low 5‑year survival rate. Typically, >70% of patients present with an advanced stage of the disease, resulting in a high number of ovarian cancer‑associated deaths worldwide. Over the past decade, adoptive cellular immunotherapy has been investigated in clinical trials, and the results have led to the increased use in cancer treatment. Natural killer (NK) cells are cytotoxic lymphoid cells that recognize and lyse transformed cells, thereby impeding tumor growth. Thus, NK cells exhibit potential as a form of immunotherapy in the treatment of cancer. However, some patients with ovarian cancer treated with NK cells have experienced unsatisfactory outcomes. Therefore, further optimization of NK cells is required to increase the number of patients achieving long‑term remission. In the present review article, studies focusing on improving NK cell function were systematically summarized, and innovative strategies that augment the anticancer properties of NK cells were proposed.
Collapse
Affiliation(s)
- Yuzhu Hou
- Department of Gynecology, Qingdao Eighth People's Hospital, Qingdao, Shandong 266000, P.R. China
| | - Xiujun Zhao
- Department of Gynecology, Qingdao Eighth People's Hospital, Qingdao, Shandong 266000, P.R. China
| | - Xiaoqian Nie
- Department of Gynecology, Qingdao Eighth People's Hospital, Qingdao, Shandong 266000, P.R. China
| |
Collapse
|
7
|
Cao Y, Li J, Zhang G, Fang H, Du Y, Liang Y. KLF15 transcriptionally activates LINC00689 to inhibit colorectal cancer development. Commun Biol 2024; 7:130. [PMID: 38273088 PMCID: PMC10810960 DOI: 10.1038/s42003-023-05757-3] [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: 04/21/2023] [Accepted: 12/29/2023] [Indexed: 01/27/2024] Open
Abstract
Colorectal cancer is a grievous health concern, we have proved long non-coding RNA LINC00689 is considered as a potential diagnosis biomarker for colorectal cancer, and it is necessary to further investigate its upstream and downstream mechanisms. Here, we show that KLF15, a transcription factor, exhibits the reduced expression in colorectal cancer. KLF15 suppresses the proliferative and metastatic capacities of colorectal cancer cells both in vitro and in vivo by transcriptionally activating LINC00689. Subsequently, LINC00689 recruits PTBP1 protein to enhance the stability of LATS2 mRNA in the cytoplasm. This stabilization causes the suppression of the YAP1/β-catenin pathway and its target downstream genes. Our findings highlight a regulatory network involving KLF15, LINC00689, PTBP1, LATS2, and the YAP1/β-catenin pathway in colorectal cancer, shedding light on potential therapeutic targets for colorectal cancer therapy.
Collapse
Affiliation(s)
- Yan Cao
- Department of Nuclear Medicine, Xiangya Third Hospital, Central South University, Changsha, 410013, Hunan Province, PR China
| | - Jian Li
- Department of Nuclear Medicine, Xiangya Third Hospital, Central South University, Changsha, 410013, Hunan Province, PR China
| | - Gang Zhang
- Department 2 of Gastrointestinal Surgery, Haikou Hospital Affiliated to Xiangya Medical College of Central South University, Haikou People's Hospital, Haikou, 570208, Hainan Province, PR China
| | - Hao Fang
- Department of Nuclear Medicine, Xiangya Third Hospital, Central South University, Changsha, 410013, Hunan Province, PR China
| | - Yongliang Du
- Department of Nuclear Medicine, Xiangya Third Hospital, Central South University, Changsha, 410013, Hunan Province, PR China
| | - Yan Liang
- Department of Nuclear Medicine, Xiangya Third Hospital, Central South University, Changsha, 410013, Hunan Province, PR China.
| |
Collapse
|
8
|
Brown JS. Comparison of Oncogenes, Tumor Suppressors, and MicroRNAs Between Schizophrenia and Glioma: The Balance of Power. Neurosci Biobehav Rev 2023; 151:105206. [PMID: 37178944 DOI: 10.1016/j.neubiorev.2023.105206] [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] [Received: 11/29/2022] [Revised: 04/25/2023] [Accepted: 04/30/2023] [Indexed: 05/15/2023]
Abstract
The risk of cancer in schizophrenia has been controversial. Confounders of the issue are cigarette smoking in schizophrenia, and antiproliferative effects of antipsychotic medications. The author has previously suggested comparison of a specific cancer like glioma to schizophrenia might help determine a more accurate relationship between cancer and schizophrenia. To accomplish this goal, the author performed three comparisons of data; the first a comparison of conventional tumor suppressors and oncogenes between schizophrenia and cancer including glioma. This comparison determined schizophrenia has both tumor-suppressive and tumor-promoting characteristics. A second, larger comparison between brain-expressed microRNAs in schizophrenia with their expression in glioma was then performed. This identified a core carcinogenic group of miRNAs in schizophrenia offset by a larger group of tumor-suppressive miRNAs. This proposed "balance of power" between oncogenes and tumor suppressors could cause neuroinflammation. This was assessed by a third comparison between schizophrenia, glioma and inflammation in asbestos-related lung cancer and mesothelioma (ALRCM). This revealed that schizophrenia shares more oncogenic similarity to ALRCM than glioma.
Collapse
|
9
|
Zhang P, Zhao L, Li H, Shen J, Li H, Xing Y. Novel diagnostic biomarkers related to immune infiltration in Parkinson's disease by bioinformatics analysis. Front Neurosci 2023; 17:1083928. [PMID: 36777638 PMCID: PMC9909419 DOI: 10.3389/fnins.2023.1083928] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2022] [Accepted: 01/09/2023] [Indexed: 01/27/2023] Open
Abstract
Background Parkinson's disease (PD) is Pengfei Zhang Liwen Zhao Pengfei Zhang Liwen Zhao a common neurological disorder involving a complex relationship with immune infiltration. Therefore, we aimed to explore PD immune infiltration patterns and identify novel immune-related diagnostic biomarkers. Materials and methods Three substantia nigra expression microarray datasets were integrated with elimination of batch effects. Differentially expressed genes (DEGs) were screened using the "limma" package, and functional enrichment was analyzed. Weighted gene co-expression network analysis (WGCNA) was performed to explore the key module most significantly associated with PD; the intersection of DEGs and the key module in WGCNA were considered common genes (CGs). The CG protein-protein interaction (PPI) network was constructed to identify candidate hub genes by cytoscape. Candidate hub genes were verified by another two datasets. Receiver operating characteristic curve analysis was used to evaluate the hub gene diagnostic ability, with further gene set enrichment analysis (GSEA). The immune infiltration level was evaluated by ssGSEA and CIBERSORT methods. Spearman correlation analysis was used to evaluate the hub genes association with immune cells. Finally, a nomogram model and microRNA-TF-mRNA network were constructed based on immune-related biomarkers. Results A total of 263 CGs were identified by the intersection of 319 DEGs and 1539 genes in the key turquoise module. Eleven candidate hub genes were screened by the R package "UpSet." We verified the candidate hub genes based on two validation sets and identified six (SYT1, NEFM, NEFL, SNAP25, GAP43, and GRIA1) that distinguish the PD group from healthy controls. Both CIBERSORT and ssGSEA revealed a significantly increased proportion of neutrophils in the PD group. Correlation between immune cells and hub genes showed SYT1, NEFM, GAP43, and GRIA1 to be significantly related to immune cells. Moreover, the microRNA-TFs-mRNA network revealed that the microRNA-92a family targets all four immune-related genes in PD pathogenesis. Finally, a nomogram exhibited a reliable capability of predicting PD based on the four immune-related genes (AUC = 0.905). Conclusion By affecting immune infiltration, SYT1, NEFM, GAP43, and GRIA1, which are regulated by the microRNA-92a family, were identified as diagnostic biomarkers of PD. The correlation of these four genes with neutrophils and the microRNA-92a family in PD needs further investigation.
Collapse
Affiliation(s)
- Pengfei Zhang
- Department of Neurosurgery, Beichen Traditional Chinese Medical Hospital Tianjin, Tianjin, China
| | - Liwen Zhao
- Department of Neurosurgery, Tianjin Medical University General Hospital Airport Site, Tianjin, China
| | - Hongbin Li
- Department of Neurology, Beichen Traditional Chinese Medical Hospital Tianjin, Tianjin, China
| | - Jie Shen
- Department of Neurology, Beichen Traditional Chinese Medical Hospital Tianjin, Tianjin, China
| | - Hui Li
- Department of Neurosurgery, Beichen Traditional Chinese Medical Hospital Tianjin, Tianjin, China
| | - Yongguo Xing
- Department of Neurosurgery, Beichen Traditional Chinese Medical Hospital Tianjin, Tianjin, China,*Correspondence: Yongguo Xing,
| |
Collapse
|
10
|
Ghafouri-Fard S, Shoorei H, Hussen BM, Poornajaf Y, Taheri M, Sharifi G. Interplay between programmed death-ligand 1 and non-coding RNAs. Front Immunol 2022; 13:982902. [PMID: 36405753 PMCID: PMC9667550 DOI: 10.3389/fimmu.2022.982902] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Accepted: 10/21/2022] [Indexed: 01/25/2023] Open
Abstract
Programmed death-ligand 1 (PD-L1) is a transmembrane protein with essential roles in the suppression of adaptive immune responses. As an immune checkpoint molecule, PD-L1 can be exploited by cancer cells to evade the anti-tumor attacks initiated by the immune system. Thus, blockade of the PD1/PD-L1 axis can eliminate the suppressive signals and release the antitumor immune responses. Identification of the underlying mechanisms of modulation of the activity of the PD1/PD-L1 axis would facilitate the design of more efficacious therapeutic options and better assignment of patients for each option. Recent studies have confirmed the interactions between miRNAs/lncRNAs/circ-RNAs and the PD1/PD-L1 axis. In the current review, we give a summary of interactions between these transcripts and PD-L1 in the context of cancer. We also overview the consequences of these interactions in the determination of the response of patients to anti-cancer drugs.
Collapse
Affiliation(s)
- Soudeh Ghafouri-Fard
- Department of Medical Genetics, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Hamed Shoorei
- Clinical Research Development Unit of Tabriz Valiasr Hospital, Tabriz University of Medical Sciences, Tabriz, Iran,Department of Anatomical Sciences, Faculty of Medicine, Birjand University of Medical Sciences, Birjand, Iran
| | - Bashdar Mahmud Hussen
- Department of Pharmacognosy, College of Pharmacy, Hawler Medical University, Erbil, Kurdistan, Iraq,Center of Research and Strategic Studies, Lebanese French University, Erbil, Kurdistan, Iraq
| | - Yadollah Poornajaf
- Faculty of Medicine, Birjand University of Medical Sciences, Birjand, Iran
| | - Mohammad Taheri
- Urology and Nephrology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran,Institute of Human Genetics, Jena University Hospital, Jena, Germany,*Correspondence: Mohammad Taheri, ; Guive Sharifi,
| | - Guive Sharifi
- Skull Base Research Center, Loghman Hakim Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran,*Correspondence: Mohammad Taheri, ; Guive Sharifi,
| |
Collapse
|
11
|
Kandettu A, Adiga D, Devi V, Suresh PS, Chakrabarty S, Radhakrishnan R, Kabekkodu SP. Deregulated miRNA clusters in ovarian cancer: Imperative implications in personalized medicine. Genes Dis 2022; 9:1443-1465. [PMID: 36157483 PMCID: PMC9485269 DOI: 10.1016/j.gendis.2021.12.026] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Revised: 12/04/2021] [Accepted: 12/31/2021] [Indexed: 11/25/2022] Open
Abstract
Ovarian cancer (OC) is one of the most common and fatal types of gynecological cancer. OC is usually detected at the advanced stages of the disease, making it highly lethal. miRNAs are single-stranded, small non-coding RNAs with an approximate size ranging around 22 nt. Interestingly, a considerable proportion of miRNAs are organized in clusters with miRNA genes placed adjacent to one another, getting transcribed together to result in miRNA clusters (MCs). MCs comprise two or more miRNAs that follow the same orientation during transcription. Abnormal expression of the miRNA cluster has been identified as one of the key drivers in OC. MC exists both as tumor-suppressive and oncogenic clusters and has a significant role in OC pathogenesis by facilitating cancer cells to acquire various hallmarks. The present review summarizes the regulation and biological function of MCs in OC. The review also highlights the utility of abnormally expressed MCs in the clinical management of OC.
Collapse
Affiliation(s)
- Amoolya Kandettu
- Department of Cell and Molecular Biology, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal, Karnataka 576104, India
| | - Divya Adiga
- Department of Cell and Molecular Biology, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal, Karnataka 576104, India
| | - Vasudha Devi
- Department of Pharmacology, Centre for Cardiovascular Pharmacology, Melaka Manipal Medical College, Manipal Academy of Higher Education, Manipal Campus, Manipal, Karnataka 576104, India
| | - Padmanaban S. Suresh
- School of Biotechnology, National Institute of Technology, Calicut, Kerala 673601, India
| | - Sanjiban Chakrabarty
- Department of Cell and Molecular Biology, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal, Karnataka 576104, India
- Center for DNA Repair and Genome Stability (CDRGS), Manipal Academy of Higher Education, Manipal, Karnataka 576104, India
| | - Raghu Radhakrishnan
- Department of Oral Pathology, Manipal College of Dental Sciences, Manipal Academy of Higher Education, Manipal, Karnataka 576104, India
| | - Shama Prasada Kabekkodu
- Department of Cell and Molecular Biology, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal, Karnataka 576104, India
- Center for DNA Repair and Genome Stability (CDRGS), Manipal Academy of Higher Education, Manipal, Karnataka 576104, India
| |
Collapse
|
12
|
Zhou P, Gao S, Hu B. Exploration of Potential Biomarkers and Immune Landscape for Hepatoblastoma: Evidence from Machine Learning Algorithm. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE : ECAM 2022; 2022:2417134. [PMID: 35958911 PMCID: PMC9357682 DOI: 10.1155/2022/2417134] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/03/2022] [Accepted: 06/02/2022] [Indexed: 11/17/2022]
Abstract
This study aimed to investigate the immune landscape in hepatoblastoma (HB) based on deconvolution methods and identify a biomarkers panel for diagnosis based on a machine learning algorithm. Firstly, we identified 277 differentially expressed genes (DEGs) and differentiated and functionally identified the modules in DEGs. The Kyoto Encyclopedia of Genes and Genomes (KEGG) analysis and GO (gene ontology) were used to annotate these DEGs, and the results suggested that the occurrence of HB was related to DNA adducts, bile secretion, and metabolism of xenobiotics by cytochrome P450. We selected the top 10 genes for our final diagnostic panel based on the random forest tree method. Interestingly, TNFRSF19 and TOP2A were significantly down-regulated in normal samples, while other genes (TRIB1, MAT1A, SAA2-SAA4, NAT2, HABP2, CYP2CB, APOF, and CFHR3) were significantly down-regulated in HB samples. Finally, we constructed a neural network model based on the above hub genes for diagnosis. After cross-validation, the area under the ROC curve was close to 1 (AUC = 0.972), and the AUC of the validation set was 0.870. In addition, the results of single-sample gene-set enrichment analysis (ssGSEA) and deconvolution methods revealed a more active immune responses in the HB tissue. In conclusion, we have developed a robust biomarkers panel for HB patients.
Collapse
Affiliation(s)
- Peng Zhou
- Department of Pediatric, Maternal and Child Health Hospital, Zibo, China
| | - Shanshan Gao
- Department of Ultrasound, Zibo Forth People's Hospital, Zibo, China
| | - Bin Hu
- Department of Pediatric, Maternal and Child Health Hospital, Zibo, China
| |
Collapse
|
13
|
Zhang Y, Wang Y, Ji H, Ding J, Wang K. The interplay between noncoding RNA and YAP/TAZ signaling in cancers: molecular functions and mechanisms. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2022; 41:202. [PMID: 35701841 PMCID: PMC9199231 DOI: 10.1186/s13046-022-02403-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/12/2022] [Accepted: 05/25/2022] [Indexed: 12/13/2022]
Abstract
The Hippo signaling pathway was found coordinately modulates cell regeneration and organ size. Its dysregulation contributes to uncontrolled cell proliferation and malignant transformation. YAP/TAZ are two critical effectors of the Hippo pathway and have been demonstrated essential for the initiation or growth of most tumors. Noncoding RNAs (ncRNAs), including miRNAs, lncRNAs, and circRNAs, have been shown to play critical roles in the development of many cancers. In the past few decades, a growing number of studies have revealed that ncRNAs can directly or indirectly regulate YAP/TAZ signaling. YAP/TAZ also regulate ncRNAs expression in return. This review summarizes the interactions between YAP/TAZ signaling and noncoding RNAs together with their biological functions on cancer progression. We also try to describe the complex feedback loop existing between these components.
Collapse
Affiliation(s)
- Yirao Zhang
- Department of Oncology, Second Affiliated Hospital of Nanjing Medical University, Nanjing, 210011, Jiangsu, China
| | - Yang Wang
- Department of Oncology, Second Affiliated Hospital of Nanjing Medical University, Nanjing, 210011, Jiangsu, China
| | - Hao Ji
- Department of Liver Surgery and Liver Transplantation Center, School of Medicine, Renji Hospital, Shanghai Jiao Tong University, Shanghai, 200127, China
| | - Jie Ding
- Department of Oncology, Second Affiliated Hospital of Nanjing Medical University, Nanjing, 210011, Jiangsu, China.
| | - Keming Wang
- Department of Oncology, Second Affiliated Hospital of Nanjing Medical University, Nanjing, 210011, Jiangsu, China.
| |
Collapse
|
14
|
Identification and Validation of a GPX4-Related Immune Prognostic Signature for Lung Adenocarcinoma. JOURNAL OF ONCOLOGY 2022; 2022:9054983. [PMID: 35620733 PMCID: PMC9130018 DOI: 10.1155/2022/9054983] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/20/2022] [Revised: 03/31/2022] [Accepted: 04/05/2022] [Indexed: 12/24/2022]
Abstract
Lung adenocarcinoma (LUAD) is a commonly occurring histological subtype of lung cancer. Glutathione peroxidase 4 (GPX4) is an important regulatory factor of ferroptosis and is involved in the development of many cancers, but its prognostic significance has not been systematically described in LUAD. In this study, we focused on developing a robust GPX4-related prognostic signature (GPS) for LUAD. Data for the training cohort was extracted from The Cancer Genome Atlas, and that for the validation cohort was sourced from the GSE72094 dataset including 863 LUAD patients. GPX4-related genes were screened out by weighted gene coexpression network analysis and Spearman’s correlation analysis. Then, Cox regression and least absolute shrinkage and selection operator regression analyses were employed to construct a GPS. The ESTIMATE algorithm, single-sample gene set enrichment analysis (ssGSEA), and GSEA were utilized to evaluate the relationship between GPS and the tumor microenvironment (TME). We constructed and validated a GPS premised on four GPX4-related genes (KIF14, LATS2, PRKCE, and TM6SF1), which could classify LUAD patients into low- and high-score cohorts. The high-risk cohort presented noticeably poorer overall survival (OS) as opposed to the low-risk cohort, meaning that the GPS may be utilized as an independent predictor of the OS of LUAD. The GPS was also adversely correlated with multiple tumor-infiltrating immune cells and immune-related processes and pathways in TME. Furthermore, greater sensitivity to erlotinib and lapatinib were identified in the low-risk cohort based on the GDSC database. Our findings suggest that the GPS can effectively forecast the prognosis of LUAD patients and may possibly regulate the TME of LUAD.
Collapse
|
15
|
Liu X, Chen B, Chen J, Su Z, Sun S. Deubiquitinase ubiquitin-specific peptidase 10 maintains cysteine rich angiogenic inducer 61 expression via Yes1 associated transcriptional regulator to augment immune escape and metastasis of pancreatic adenocarcinoma. Cancer Sci 2022; 113:1868-1879. [PMID: 35271750 PMCID: PMC9128165 DOI: 10.1111/cas.15326] [Citation(s) in RCA: 12] [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: 11/23/2021] [Revised: 03/01/2022] [Accepted: 03/03/2022] [Indexed: 12/24/2022] Open
Abstract
Pancreatic adenocarcinoma (PAAD) remains an extremely fatal malignancy with a high mortality rate worldwide. This study focuses on the roles of ubiquitin-specific peptidase 10 (USP10) and cysteine rich angiogenic inducer 61 (Cyr61) in macrophage polarization, immune escape, and metastasis of PAAD. USP10 showed a positive correlation with Yes1 associated transcriptional regulator (YAP1), which, according to the TCGA-PAAD database, is highly expressed in PAAD and indicates poor patient prognosis. USP10 knockdown increased ubiquitination and degradation of YAP1, which further decreased the programmed cell death ligand 1 (PD-L1) and Galectin-9 expression, suppressed immune escape, and reduced the proliferation and metastasis of PAAD cells in vitro and in vivo. Cyr61, a downstream factor of YAP1, was overexpressed in PAAD cells after USP10 silencing for rescue experiments. Overexpression of Cyr61 restored the PD-L1 and Galectin-9 expression in cells and triggered M2 polarization of macrophages, which enhanced the immune escape and maintained the proliferation and metastasis ability of PAAD cells. In conclusion, this work demonstrates that USP10 inhibits YAP1 ubiquitination and degradation to promote Cyr61 expression, which induces immune escape and promotes growth and metastasis of PAAD.
Collapse
Affiliation(s)
- Xun Liu
- Department of General SurgeryShengjing Hospital of China Medical UniversityShenyangChina
| | - Bobo Chen
- Department of General SurgeryShengjing Hospital of China Medical UniversityShenyangChina
| | - Jiahui Chen
- Department of General SurgeryShengjing Hospital of China Medical UniversityShenyangChina
| | - Zuoyuan Su
- Department of General SurgeryShengjing Hospital of China Medical UniversityShenyangChina
| | - Shaolong Sun
- Department of General SurgeryShengjing Hospital of China Medical UniversityShenyangChina
| |
Collapse
|
16
|
Zhao C, Chen J, Liu Y, Ju S, Wang G, Wang X. Large tumor suppressor 2 is a prognostic biomarker and correlated with immune infiltrates in colorectal cancer. Bioengineered 2021; 12:11648-11661. [PMID: 34699318 PMCID: PMC8810027 DOI: 10.1080/21655979.2021.1996513] [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] [Indexed: 11/18/2022] Open
Abstract
Colorectal cancer (CRC) is a common malignancy that has both low 5-year survival and high prevalence. Immunotherapy has achieved impressive progress for treatment of CRC, but still faces huge challenges. Although large tumor suppressor 2 (LATS2) is well accepted to be related to cancer progression, the prognostic potential and immune response role of LATS2 expression in CRC remain unclear. To investigate the value of LATS2 for prognosis and immune infiltration, a retrospective study of 213 CRC patients was carried out. We determined the expression of LATS2 in tumor tissues by immunohistochemistry. The results indicated that LATS2 expression was down-regulated in CRC tissues and clearly related to tumor differentiation (P = 0.002) and TNM stage (P = 0.002). Low LATS2 expression and TNM stage were subsequently identified as significant independent predictors of prognosis in CRC by univariate and multivariate analyses. In Kaplan–Meier survival analyses, CRC patients with elevated LATS2 expression and early TNM stage had better overall survival. We further found that LATS2 was involved in the regulation of immune-related pathways and that its expression was positively related to tumor-infiltrating immune cells by GSEA, TIMER, and ssGSEA analyses. In summary, our data imply that LATS2 may act as a cancer suppressor gene and be correlated with clinical prognosis and immune infiltration in CRC. Thus, LATS2 may be applied as a novel biomarker for predicting clinical outcomes and immune infiltration levels in CRC.
Collapse
Affiliation(s)
- Chengwen Zhao
- Department of Laboratory Medicine, Affiliated Hospital of Nantong University, Nantong, Jiangsu, China
| | - Jianping Chen
- Department of Laboratory Medicine, Affiliated Hospital of Nantong University, Nantong, Jiangsu, China
| | - Yonghui Liu
- Department of Laboratory Medicine, Affiliated Hospital of Nantong University, Nantong, Jiangsu, China
| | - Shaoqing Ju
- Department of Laboratory Medicine, Affiliated Hospital of Nantong University, Nantong, Jiangsu, China.,School of Public Health, Nantong University, Nantong, Jiangsu, China
| | - Guihua Wang
- Department of Laboratory Medicine, Affiliated Hospital of Nantong University, Nantong, Jiangsu, China
| | - Xudong Wang
- Department of Laboratory Medicine, Affiliated Hospital of Nantong University, Nantong, Jiangsu, China.,School of Public Health, Nantong University, Nantong, Jiangsu, China
| |
Collapse
|
17
|
Li M, Xue L, Xu W, Liu P, Li F. rno-miR-128-3p promotes apoptosis in rat granulosa cells (GCs) induced by norepinephrine through Wilms tumor 1 (WT1). In Vitro Cell Dev Biol Anim 2021; 57:775-785. [PMID: 34554376 PMCID: PMC8585816 DOI: 10.1007/s11626-021-00609-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2021] [Accepted: 07/07/2021] [Indexed: 11/24/2022]
Abstract
The mechanism related to ovarian follicular is complex, which has not been fully elucidated. Abundant reports have confirmed that the ovarian function development is closely related to sympathetic innervation. As one of the major neurotransmitters, norepinephrine (NE) is considered an effective regulator of ovarian functions like granulosa cell (GC) apoptosis. However, the mechanism between NE and GC apoptosis in rat is still unclear. In our study, GCs were isolated and cultured in vitro with NE treatment. The apoptosis of GCs was facilitated by NE. Wilms tumor 1 (WT1) was found to be significantly downregulated in GCs after NE treatment, and overexpression of WT1 repressed apoptosis in rat GCs induced by NE. rno-miR-128-3p was found to be significantly enhanced by NE in GCs, and inhibition of rno-miR-128-3p repressed apoptosis in rat GCs induced by NE. Mechanistically, rno-miR-128-3p interacted with WT1 and repressed its expression. In summary, inhibition of rno-miR-128-3p may enhance WT1 expression, and then repress NE-induced apoptosis in rat GCs. Our research may provide a new insight for the improvement of ovarian follicular development.
Collapse
Affiliation(s)
- Ming Li
- Department of Pharmacy, Lian Yungang Hospital of Traditional Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Jiang Su Province, Lianyungang, 222000, People's Republic of China
| | - Ling Xue
- Pharmacy Department, Shandong Qingdao Hospital of Integrated Traditional Chinese and Western Medicine, No. 3 Jiaxiang Road, South District, Qingdao City, Shandong Province, 266002, China
| | - Weibin Xu
- Pharmacy Department, Gaoqing County People's Hospital, No.11 Qingcheng Road, Gaoqing County, Zibo City, 256300, Shandong Province, China
| | - Pingping Liu
- Pharmacy Department, Liaocheng Chiping District People's Hospital, No. 1057 Culture Road, Chiping County, Liaocheng City, 252100, Shandong Province, China
| | - Feng Li
- Drug Dispensing Department, Zibo Central Hospital, No. 54 The Communist Youth League West Road, Zhangdian District, Zibo City, 255000, Shandong Province, China.
| |
Collapse
|