51
|
Alqualo NO, Campos-Fernandez E, Picolo BU, Ferreira EL, Henriques LM, Lorenti S, Moreira DC, Simião MPS, Oliveira LBT, Alonso-Goulart V. Molecular biomarkers in prostate cancer tumorigenesis and clinical relevance. Crit Rev Oncol Hematol 2024; 194:104232. [PMID: 38101717 DOI: 10.1016/j.critrevonc.2023.104232] [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: 09/22/2023] [Revised: 12/08/2023] [Accepted: 12/09/2023] [Indexed: 12/17/2023] Open
Abstract
Prostate cancer (PCa) is the second most frequent type of cancer in men and assessing circulating tumor cells (CTCs) by liquid biopsy is a promising tool to help in cancer early detection, staging, risk of recurrence evaluation, treatment prediction and monitoring. Blood-based liquid biopsy approaches enable the enrichment, detection and characterization of CTCs by biomarker analysis. Hence, comprehending the molecular markers, their role on each stage of cancer development and progression is essential to provide information that can help in future implementation of these biomarkers in clinical assistance. In this review, we studied the molecular markers most associated with PCa CTCs to better understand their function on tumorigenesis and metastatic cascade, the methodologies utilized to analyze these biomarkers and their clinical significance, in order to summarize the available information to guide researchers in their investigations, new hypothesis formulation and target choice for the development of new diagnostic and treatment tools.
Collapse
Affiliation(s)
- Nathalia Oliveira Alqualo
- Laboratory of Nanobiotechnology, Prof. Dr. Luiz Ricardo Goulart Filho, Institute of Biotechnology, Universidade Federal de Uberlândia, Uberlandia, MG 38400-902, Brazil
| | - Esther Campos-Fernandez
- Laboratory of Nanobiotechnology, Prof. Dr. Luiz Ricardo Goulart Filho, Institute of Biotechnology, Universidade Federal de Uberlândia, Uberlandia, MG 38400-902, Brazil
| | - Bianca Uliana Picolo
- Laboratory of Nanobiotechnology, Prof. Dr. Luiz Ricardo Goulart Filho, Institute of Biotechnology, Universidade Federal de Uberlândia, Uberlandia, MG 38400-902, Brazil
| | - Emanuelle Lorrayne Ferreira
- Laboratory of Nanobiotechnology, Prof. Dr. Luiz Ricardo Goulart Filho, Institute of Biotechnology, Universidade Federal de Uberlândia, Uberlandia, MG 38400-902, Brazil
| | - Laila Machado Henriques
- Laboratory of Nanobiotechnology, Prof. Dr. Luiz Ricardo Goulart Filho, Institute of Biotechnology, Universidade Federal de Uberlândia, Uberlandia, MG 38400-902, Brazil
| | - Sabrina Lorenti
- Laboratory of Nanobiotechnology, Prof. Dr. Luiz Ricardo Goulart Filho, Institute of Biotechnology, Universidade Federal de Uberlândia, Uberlandia, MG 38400-902, Brazil
| | - Danilo Caixeta Moreira
- Laboratory of Nanobiotechnology, Prof. Dr. Luiz Ricardo Goulart Filho, Institute of Biotechnology, Universidade Federal de Uberlândia, Uberlandia, MG 38400-902, Brazil
| | - Maria Paula Silva Simião
- Laboratory of Nanobiotechnology, Prof. Dr. Luiz Ricardo Goulart Filho, Institute of Biotechnology, Universidade Federal de Uberlândia, Uberlandia, MG 38400-902, Brazil
| | - Luciana Beatriz Tiago Oliveira
- Laboratory of Nanobiotechnology, Prof. Dr. Luiz Ricardo Goulart Filho, Institute of Biotechnology, Universidade Federal de Uberlândia, Uberlandia, MG 38400-902, Brazil
| | - Vivian Alonso-Goulart
- Laboratory of Nanobiotechnology, Prof. Dr. Luiz Ricardo Goulart Filho, Institute of Biotechnology, Universidade Federal de Uberlândia, Uberlandia, MG 38400-902, Brazil.
| |
Collapse
|
52
|
Takahashi H, Ito R, Matsumura Y, Sakai J. Environmental factor reversibly determines cellular identity through opposing Integrators that unify epigenetic and transcriptional pathways. Bioessays 2024; 46:e2300084. [PMID: 38013256 DOI: 10.1002/bies.202300084] [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: 05/15/2023] [Revised: 09/29/2023] [Accepted: 11/13/2023] [Indexed: 11/29/2023]
Abstract
Organisms must adapt to environmental stresses to ensure their survival and prosperity. Different types of stresses, including thermal, mechanical, and hypoxic stresses, can alter the cellular state that accompanies changes in gene expression but not the cellular identity determined by a chromatin state that remains stable throughout life. Some tissues, such as adipose tissue, demonstrate remarkable plasticity and adaptability in response to environmental cues, enabling reversible cellular identity changes; however, the mechanisms underlying these changes are not well understood. We hypothesized that positive and/or negative "Integrators" sense environmental cues and coordinate the epigenetic and transcriptional pathways required for changes in cellular identity. Adverse environmental factors such as pollution disrupt the coordinated control contributing to disease development. Further research based on this hypothesis will reveal how organisms adapt to fluctuating environmental conditions, such as temperature, extracellular matrix stiffness, oxygen, cytokines, and hormonal cues by changing their cellular identities.
Collapse
Grants
- JP20gm1310007 Japan Agency for Medical Research and Development
- JP16H06390 Ministry of Education, Culture, Sports, Science and Technology
- JP21H04826 Ministry of Education, Culture, Sports, Science and Technology
- JP20H04835 Ministry of Education, Culture, Sports, Science and Technology
- JP20K21747 Ministry of Education, Culture, Sports, Science and Technology
- JP22K18411 Ministry of Education, Culture, Sports, Science and Technology
- JP21K21211 Ministry of Education, Culture, Sports, Science and Technology
- JP19J11909 Ministry of Education, Culture, Sports, Science and Technology
- JPMJPF2013 Japan Science and Technology Agency
Collapse
Affiliation(s)
- Hiroki Takahashi
- Division of Molecular Physiology and Metabolism, Tohoku University Graduate School of Medicine, Sendai, Japan
- Division of Metabolic Medicine, Research Center for Advanced Science and Technology, The University of Tokyo, Tokyo, Japan
| | - Ryo Ito
- Division of Molecular Physiology and Metabolism, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Yoshihiro Matsumura
- Division of Molecular Physiology and Metabolism, Tohoku University Graduate School of Medicine, Sendai, Japan
- Division of Metabolic Medicine, Research Center for Advanced Science and Technology, The University of Tokyo, Tokyo, Japan
| | - Juro Sakai
- Division of Molecular Physiology and Metabolism, Tohoku University Graduate School of Medicine, Sendai, Japan
- Division of Metabolic Medicine, Research Center for Advanced Science and Technology, The University of Tokyo, Tokyo, Japan
| |
Collapse
|
53
|
Liu S, Zhi Y, Zhang R, You Y, You W, Xu Q, Li J, Li J. Cronkhite‒Canada syndrome as inflammatory hamartomatous polyposis: new evidence from whole transcriptome sequencing of colonic polyps. Orphanet J Rare Dis 2024; 19:35. [PMID: 38297356 PMCID: PMC10832113 DOI: 10.1186/s13023-024-03038-8] [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: 07/14/2023] [Accepted: 01/19/2024] [Indexed: 02/02/2024] Open
Abstract
BACKGROUND Cronkhite-Canada syndrome (CCS) is a rare, nonhereditary disease characterized by diffuse gastrointestinal polyposis and ectodermal abnormalities. Although it has been proposed to be a chronic inflammatory condition, direct evidence of its pathogenesis is lacking. This study aims to investigate the pathophysiology of CCS by analyzing transcriptomic changes in the colonic microenvironment. METHODS Next-generation sequencing-based genome-wide transcriptional profiling was performed on colonic hamartomatous polyps from four CCS patients and normal colonic mucosa from four healthy volunteers. Analyses of differential expression and multiple enrichment analyses were conducted from the molecular level to the cellular level. Quantitative real-time PCR (qRT-PCR) was carried out to validate the sequencing accuracy in samples from six CCS patients and six healthy volunteers. RESULTS A total of 543 differentially expressed genes were identified, including an abundance of CC- and CXC-chemokines. Innate immune response-related pathways and processes, such as leukocyte chemotaxis, cytokine production, IL-17, TNF, IL-1 and NF-kB signaling pathways, were prominently enhanced in CCS colonic polyps. Upregulation of wound healing, epithelial-mesenchymal transition, Wnt, and PI3K-Akt signaling pathways were also observed. Enrichment analyses at different levels identified extracellular structure disorganization, dysfunction of the gut mucosal barrier, and increased angiogenesis. Validation by qRT-PCR confirmed increased expression of the LCN2, IL1B, CXCL1, and CXCL3 genes in CCS colonic polyps. CONCLUSIONS This case-control whole transcriptome analysis of active CCS colonic hamartomatous polyps revealed intricate molecular pathways, emphasizing the role of the innate immune response, extracellular matrix disorganization, inflammatory cell infiltration, increased angiogenesis, and potential epithelial to mesenchymal transition. These findings supports CCS as a chronic inflammatory condition and sheds light on potential therapeutic targets, paving the way for more effective and personalized management of CCS in the future.
Collapse
Affiliation(s)
- Shuang Liu
- Department of Allergy, Chinese Academy of Medical Sciences, Peking Union Medical College Hospital, 100730, Beijing, People's Republic of China
| | - Yunfei Zhi
- Department of Gastroenterology, Chinese Academy of Medical Sciences, Peking Union Medical College Hospital, Beijing, 100730, People's Republic of China
| | - Runfeng Zhang
- Department of Internal Medicine, Chinese Academy of Medical Sciences, Peking Union Medical College Hospital, 100730, Beijing, China
| | - Yan You
- Department of Pathology, Chinese Academy of Medical Sciences, Peking Union Medical College Hospital, 100730, Beijing, People's Republic of China
| | - Wen You
- Department of Gastroenterology, First Affiliated Hospital of Nanjing Medical University, 210029, Nanjing, People's Republic of China
| | - Qiushi Xu
- Department of Gastroenterology, Chinese Academy of Medical Sciences, Peking Union Medical College Hospital, Beijing, 100730, People's Republic of China
| | - Jingnan Li
- Department of Gastroenterology, Chinese Academy of Medical Sciences, Peking Union Medical College Hospital, Beijing, 100730, People's Republic of China
| | - Ji Li
- Department of Gastroenterology, Chinese Academy of Medical Sciences, Peking Union Medical College Hospital, Beijing, 100730, People's Republic of China.
| |
Collapse
|
54
|
Cui Y, Zhang Y, Liu Y, Zhou Z, Zhu L, Zhou CX. EN1 promotes lung metastasis of salivary adenoid cystic carcinoma by regulating the PI3K-AKT pathway and epithelial-mesenchymal transition. Cancer Cell Int 2024; 24:51. [PMID: 38291456 PMCID: PMC10829235 DOI: 10.1186/s12935-024-03230-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Accepted: 01/16/2024] [Indexed: 02/01/2024] Open
Abstract
BACKGROUND Engrailed homeobox 1 (EN1) is a candidate oncogene that is epigenetically modified in salivary adenoid cystic carcinoma (SACC). We investigated the expression of EN1 in SACC tissues and cells, EN1 promoter methylation, and the role of EN1 in tumour progression in SACC. METHODS Thirty-five SACC samples were screened for key transcription factors that affect tumour progression. In vitro and in vivo assays were performed to determine the viability, tumorigenicity, and metastatic ability of SACC cells with modulated EN1 expression. Quantitative methylation-specific polymerase chain reaction analysis was performed on SACC samples. RESULTS EN1 was identified as a transcription factor that was highly overexpressed in SACC tissues, regardless of clinical stage and histology subtype, and its level of expression correlated with distant metastasis. EN1 promoted cell invasion and migration through epithelial-mesenchymal transition in vitro and enhanced SACC metastasis to the lung in vivo. RNA-seq combined with in vitro assays indicated that EN1 might play an oncogenic role in SACC through the PI3K-AKT pathway. EN1 mRNA levels were negatively correlated with promoter hypermethylation, and inhibition of DNA methylation by 5-aza-dC increased EN1 expression. CONCLUSIONS The transcription factor EN1 is overexpressed in SACC under methylation regulation and plays a pivotal role in SACC progression through the PI3K-AKT pathway. These results suggest that EN1 may be a diagnostic biomarker and a potential therapeutic target for SACC.
Collapse
Affiliation(s)
- Yajuan Cui
- Department of Oral Pathology, Peking University School and Hospital of Stomatology & National Center of Stomatology & National Clinical Research Center for Oral Disease & National Engineering Research Center of Oral Biomaterials and Digital Medicine Devices, 22 South Avenue Zhongguancun, Haidian District, Beijing, 100081, PR China
| | - Ye Zhang
- Department of Oral Pathology, Peking University School and Hospital of Stomatology & National Center of Stomatology & National Clinical Research Center for Oral Disease & National Engineering Research Center of Oral Biomaterials and Digital Medicine Devices, 22 South Avenue Zhongguancun, Haidian District, Beijing, 100081, PR China
| | - Yuping Liu
- Department of Oral Pathology, Peking University School and Hospital of Stomatology & National Center of Stomatology & National Clinical Research Center for Oral Disease & National Engineering Research Center of Oral Biomaterials and Digital Medicine Devices, 22 South Avenue Zhongguancun, Haidian District, Beijing, 100081, PR China
| | - Zheng Zhou
- Department of Oral Pathology, Peking University School and Hospital of Stomatology & National Center of Stomatology & National Clinical Research Center for Oral Disease & National Engineering Research Center of Oral Biomaterials and Digital Medicine Devices, 22 South Avenue Zhongguancun, Haidian District, Beijing, 100081, PR China
| | - Lijing Zhu
- Department of Oral Pathology, Peking University School and Hospital of Stomatology & National Center of Stomatology & National Clinical Research Center for Oral Disease & National Engineering Research Center of Oral Biomaterials and Digital Medicine Devices, 22 South Avenue Zhongguancun, Haidian District, Beijing, 100081, PR China
| | - Chuan-Xiang Zhou
- Department of Oral Pathology, Peking University School and Hospital of Stomatology & National Center of Stomatology & National Clinical Research Center for Oral Disease & National Engineering Research Center of Oral Biomaterials and Digital Medicine Devices, 22 South Avenue Zhongguancun, Haidian District, Beijing, 100081, PR China.
| |
Collapse
|
55
|
Liu P, Kong X, Yi S, Chen Y, Luo W. IFIT3 accelerates the progression of head and neck squamous cell carcinoma by targeting PD-L1 to activate PI3K/AKT signaling pathway. World J Surg Oncol 2024; 22:34. [PMID: 38273364 PMCID: PMC10809513 DOI: 10.1186/s12957-023-03274-5] [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: 08/23/2023] [Accepted: 12/08/2023] [Indexed: 01/27/2024] Open
Abstract
BACKGROUND Emerging evidence has shown interferon-induced protein with tetratricopeptide repeats 3 (IFIT3) may be predicted to be a candidate oncogene and involved in the onset and progression of cancer, but IFIT3's potential role in cancer, particularly in head and neck squamous cell carcinoma (HNSC), is not well recognized. This study aims to reveal the role of IFIT3 in HNSC and the underlying molecular mechanism. METHODS Bioinformatics analysis, immunohistochemical staining, RT-PCR, and Western blotting analysis were used to detect IFIT3 expression in HNSC. CCK-8 assays, colony formation assays, wound-healing assays, transwell assays, and sphere formation were used to explore proliferative, migratory, and invasive activities and cancer stemness of HNSC cells after IFIT3 knockdown and over-expressed. The alterations of EMT markers and PI3K/AKT pathway were detected by Western blotting. Animal studies were performed to analyze the effect of IFIT3 on tumor growth and metastasis of HNSC in vivo. RESULTS In this study, we observed that IFIT3 was highly expressed in HNSC, and its higher expression contributed to poorer survival of patients with clinical stage IV or grade 3. Function assay indicated that IFIT3 promoted malignant behaviors in vitro, as well as tumor growth and lung metastasis in vivo. Meanwhile, PD-L1 knockdown or over-expressed reversed cancer cell stemness, migration, invasion, and PI3K/AKT signaling pathway which were regulated by IFIT3. CONCLUSIONS Our results reveal that IFIT3 promotes EMT and cancer stemness by targeting PD-L1 to activate PI3K/AKT signaling pathway in HNSC, and targeting IFIT3 may be a novel strategy for the treatment of patients with HNSC.
Collapse
Affiliation(s)
- Peng Liu
- Department of Otolaryngology Head and Neck Surgery, the Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, China.
| | - Xin Kong
- Department of Infectious Diseases, Key Laboratory of Molecular Biology for Infectious Diseases, Institute for Viral Hepatitis, the Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Shijiang Yi
- Department of Otolaryngology Head and Neck Surgery, the Affiliated Hospital of Guilin Medical University, Guilin, China
| | - Ying Chen
- Department of Traditional Chinese Medicine, the Affiliated Hospital of Guilin Medical University, Guilin, China
| | - Wenlong Luo
- Department of Otolaryngology Head and Neck Surgery, the Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, China.
| |
Collapse
|
56
|
Hosseinalizadeh H, Hussain QM, Poshtchaman Z, Ahsan M, Amin AH, Naghavi S, Mahabady MK. Emerging insights into keratin 7 roles in tumor progression and metastasis of cancers. Front Oncol 2024; 13:1243871. [PMID: 38260844 PMCID: PMC10800941 DOI: 10.3389/fonc.2023.1243871] [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: 06/21/2023] [Accepted: 10/26/2023] [Indexed: 01/24/2024] Open
Abstract
Keratin 7 (KRT7), also known as cytokeratin-7 (CK-7) or K7, constitutes the principal constituent of the intermediate filament cytoskeleton and is primarily expressed in the simple epithelia lining the cavities of the internal organs, glandular ducts, and blood vessels. Various pathological conditions, including cancer, have been linked to the abnormal expression of KRT7. KRT7 overexpression promotes tumor progression and metastasis in different human cancers, although the mechanisms of these processes caused by KRT7 have yet to be established. Studies have indicated that the suppression of KRT7 leads to rapid regression of tumors, highlighting the potential of KRT7 as a novel candidate for therapeutic interventions. This review aims to delineate the various roles played by KRT7 in the progression and metastasis of different human malignancies and to investigate its prognostic significance in cancer treatment. Finally, the differential diagnosis of cancers based on the KRT7 is emphasized.
Collapse
Affiliation(s)
- Hamed Hosseinalizadeh
- Department of Medical Biotechnology, Faculty of Paramedicine, Guilan University of Medical Sciences, Rasht, Iran
| | | | - Zahra Poshtchaman
- Department of Nursing, Esfarayen Faculty of Medical Sciences, Esfarayen, Iran
| | | | - Ali H. Amin
- Zoology Department, Faculty of Science, Mansoura University, Mansoura, Egypt
| | - Soroush Naghavi
- Student Research Committee, Iran University of Medical Sciences, Tehran, Iran
| | - Mahmood Khaksary Mahabady
- Anatomical Sciences Research Center, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, Iran
| |
Collapse
|
57
|
Zhou J, Luo J, Gan R, Zhi L, Zhou H, Lv M, Huang Y, Liang G. SSPH I, A Novel Anti-cancer Saponin, Inhibits EMT and Invasion and Migration of NSCLC by Suppressing MAPK/ERK1/2 and PI3K/AKT/ mTOR Signaling Pathways. Recent Pat Anticancer Drug Discov 2024; 19:543-555. [PMID: 38305308 DOI: 10.2174/0115748928283132240103073039] [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: 10/21/2023] [Revised: 11/30/2023] [Accepted: 12/07/2023] [Indexed: 02/03/2024]
Abstract
BACKGROUND Saponin of Schizocapsa plantaginea Hance I (SSPH I).a bioactive saponin found in Schizocapsa plantaginea, exhibits significant anti-proliferation and antimetastasis in lung cancer. OBJECTIVE To explore the anti-metastatic effects of SSPH I on non-small cell lung cancer (NSCLC) with emphasis on epithelial-mesenchymal transition (EMT) both in vitro and in vivo. METHODS The effects of SSPH I at the concentrations of 0, 0.875,1.75, and 3.5 μM on A549 and PC9 lung cancer cells were evaluated using colony formation assay, CCK-8 assay, transwell assay and wound-healing assay. The actin cytoskeleton reorganization of PC9 and A549 cells was detected using the FITC-phalloidin fluorescence staining assay. The proteins related to EMT (N-cadherin, E-cadherin and vimentin), p- PI3K, p- AKT, p- mTOR and p- ERK1/2 were detected by Western blotting. A mouse model of lung cancer metastasis was established by utilizing 95-D cells, and the mice were treated with SSPH I by gavage. RESULTS The results suggested that SSPH I significantly inhibited the migration and invasion of NSCLC cells under a non-cytotoxic concentration. Furthermore, SSPH I at a non-toxic concentration of 0.875 μM inhibited F-actin cytoskeleton organization. Importantly, attenuation of EMT was observed in A549 cells with upregulation in the expression of epithelial cell marker E-cadherin and downregulation of the mesenchymal cell markers vimentin as well as Ncadherin. Mechanistic studies revealed that SSPH I inhibited MAPK/ERK1/2 and PI3K/AKT/mTOR signaling pathways. CONCLUSION SSPH I inhibited EMT, migration, and invasion of NSCLC cells by suppressing MAPK/ERK1/2 and PI3K/AKT/mTOR signaling pathways, suggesting that the natural compound SSPH I could be used for inhibiting metastasis of NSCLC.
Collapse
Affiliation(s)
- Jinling Zhou
- College of Pharmacy, Guangxi University of Chinese Medicine, Nanning, China
| | - Jian Luo
- Department of Pharmacy, Guangxi University of Chinese Medicine, Nanning, China
| | - Rizhi Gan
- College of Pharmacy, Guangxi Medical University, Nanning, China
| | - Limin Zhi
- College of Pharmacy, Guangxi Medical University, Nanning, China
| | - Huan Zhou
- Department of Pharmacy, Guangxi Medical University Cancer Hospital, Nanning, China
| | - Meixian Lv
- College of Pharmacy, Guangxi Medical University, Nanning, China
| | - Yinmei Huang
- College of Pharmacy, Guangxi Medical University, Nanning, China
| | - Gang Liang
- College of Pharmacy, Guangxi Medical University, Nanning, China
- Key Laboratory of Longevity and Aging-related Diseases of Chinese Ministry of Education, Center for Translational Medicine, Guangxi Medical University, Nanning, Guangxi 530021, China
| |
Collapse
|
58
|
Zhao Z, Song Z, Wang Z, Zhang F, Ding Z, Zhao Z, Liu L, Fan T. Retinol dehydrogenase 10 promotes epithelial-mesenchymal transition in spinal cord gliomas via PI3K/AKT pathway. Int J Immunopathol Pharmacol 2024; 38:3946320241276336. [PMID: 39180753 PMCID: PMC11344904 DOI: 10.1177/03946320241276336] [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: 03/19/2024] [Accepted: 07/30/2024] [Indexed: 08/26/2024] Open
Abstract
Background: Spinal cord glioma (SCG), a rare subset of central nervous system (CNS) glioma, represents a complex challenge in neuro-oncology. There has been research showing that Retinol Dehydrogenase 10 (RDH10) may be a tumor promoting factor in brain glioma, but the biological effects of RDH10 remain undefined in SCG. Methods: We performed gene set enrichment analysis (GSEA) and unsupervised clustering analysis to investigate the roles of EMT (epithelial-mesenchymal transition) in glioma. DEG (differently expressed gene) screening and correlation analysis were conducted to filter the candidate genes which were closely associated with EMT process in SCG. Enrichment analysis and GSVA (Gene Set Variation Analysis) were conducted to investigate the potential mechanism of RDH10 for SCG. Trans-well and healing assay were performed to explore the role of RDH10 in the invasion of SCG. Western blotting was performed to evaluate the levels of markers in PI3K-AKT and EMT pathway. In vivo tests were conducted to verify the role of RDH10 in EMT process. Results: Bioinformatic analysis demonstrated the EMT pathway was associated with dismal prognosis of glioma. Further analysis demonstrated that RDH10 showed the strongest correlation with the EMT process. Retinol Dehydrogenase 10 expression was significantly increased in SCG tissues, correlating with advanced tumor grade and unfavorable prognosis. Functional analysis indicated that decreasing RDH10 levels impeded the invasive and migratory abilities of SCG cells, whereas increasing RDH10 levels augmented them. Enrichment analysis and western blot revealed that RDH10 regulated EMT process of SCG by PI3K-AKT pathway. We observed that the enhanced invasion ability and increased EMT-related protein induced by RDH10 overexpression can be suppressed by PI3K-AKT pathway inhibitor (LY294002). Conclusion: Our research found that RDH10 was an effective biomarker associated with tumor grade and prognosis of SCG. RDH10 could regulate EMT process of SCG through PI3K-AKT pathway.
Collapse
Affiliation(s)
- Zijun Zhao
- Spine Center, Sanbo Brain Hospital, Capital Medical University, Beijing, China
| | - Zihan Song
- Department of Neurosurgery, The Second Hospital of Hebei Medical University, Shijiazhuang, China
| | - Zairan Wang
- Department of Neurosurgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Fan Zhang
- Spine Center, Sanbo Brain Hospital, Capital Medical University, Beijing, China
| | - Ze Ding
- Spine Center, Sanbo Brain Hospital, Capital Medical University, Beijing, China
| | - Zongmao Zhao
- Department of Neurosurgery, The Second Hospital of Hebei Medical University, Shijiazhuang, China
| | - Liqiang Liu
- Department of Neurosurgery, The Second Hospital of Hebei Medical University, Shijiazhuang, China
| | - Tao Fan
- Spine Center, Sanbo Brain Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
59
|
Tekin C, Ercelik M, Dunaev P, Galembikova A, Tezcan G, Aksoy SA, Budak F, Isık O, Ugras N, Boichuk S, Tunca B. Leaf Extract from European Olive (Olea europaea L.) Post-Transcriptionally Suppresses the Epithelial-Mesenchymal Transition and Sensitizes Gastric Cancer Cells to Chemotherapy. BIOCHEMISTRY. BIOKHIMIIA 2024; 89:97-115. [PMID: 38467548 DOI: 10.1134/s0006297924010061] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/10/2023] [Revised: 11/21/2023] [Accepted: 11/30/2023] [Indexed: 03/13/2024]
Abstract
The overall survival of patients with the advanced and recurrent gastric cancer (GC) remains unfavorable. In particular, this is due to cancer spreading and resistance to chemotherapy associated with the epithelial-mesenchymal transition (EMT) of tumor cells. EMT can be identified by the transcriptome profiling of GC for EMT markers. Indeed, analysis of the TCGA and GTEx databases (n = 408) and a cohort of GC patients (n = 43) revealed that expression of the CDH2 gene was significantly decreased in the tumors vs. non-tumor tissues and correlated with the overall survival of GC patients. Expression of the EMT-promoting transcription factors SNAIL and ZEB1 was significantly increased in GC. These data suggest that targeting the EMT might be an attractive therapeutic approach for patients with GC. Previously, we demonstrated a potent anti-cancer activity of the olive leaf extract (OLE). However, its effect on the EMT regulation in GC remained unknown. Here, we showed that OLE efficiently potentiated the inhibitory effect of the chemotherapeutic agents 5-fluorouracil (5-FU) and cisplatin (Cis) on the EMT and their pro-apoptotic activity, as was demonstrated by changes in the expression of the EMT markers (E- and N-cadherins, vimentin, claudin-1) in GC cells treated with the aforementioned chemotherapeutic agents in the presence of OLE. Thus, culturing GC cells with 5-FU + OLE or Cis + OLE attenuated the invasive properties of cancer cells. Importantly, upregulation of expression of the apoptotic markers (PARP cleaved form) and increase in the number of cells undergoing apoptosis (annexin V-positive) were observed for GC cells treated with a combination of OLE and 5-FU or Cis. Collectively, our data illustrate that OLE efficiently interferes with the EMT in GC cells and potentiates the pro-apoptotic activity of certain chemotherapeutic agents used for GC therapy.
Collapse
Affiliation(s)
- Cagla Tekin
- Department of Medical Biology, Faculty of Medicine, Bursa Uludag University, Bursa, Turkey
| | - Melis Ercelik
- Department of Medical Biology, Faculty of Medicine, Bursa Uludag University, Bursa, Turkey
| | - Pavel Dunaev
- Department of Pathology, Kazan State Medical University, Kazan, Russia
| | - Aigul Galembikova
- Department of Pathology, Kazan State Medical University, Kazan, Russia
| | - Gulcin Tezcan
- Department of Fundamental Sciences, Faculty of Dentistry, Bursa Uludag University, Bursa, Turkey
| | - Secil Ak Aksoy
- Inegol Vocation School, Bursa Uludag University, Bursa, Turkey
- Experimental Animal Breeding and Research Unit, Faculty of Medicine, Bursa Uludag University, Bursa, Turkey
| | - Ferah Budak
- Department of Immunology, Medical Faculty, Bursa Uludag University Bursa, Turkey
| | - Ozgen Isık
- Department of General Surgery, Medical Faculty, Bursa Uludag University Bursa, Turkey
| | - Nesrin Ugras
- Department of Pathology, Medical Faculty, Bursa Uludag University, Bursa, Turkey
| | - Sergei Boichuk
- Department of Pathology, Kazan State Medical University, Kazan, Russia.
- Department of Radiotherapy and Radiology, Russian Medical Academy of Continuous Professional Education, Moscow, Russia
- "Biomarker" Research Laboratory, Institute of Fundamental Medicine and Biology, Kazan Federal University, Kazan, Russia
| | - Berrin Tunca
- Department of Medical Biology, Faculty of Medicine, Bursa Uludag University, Bursa, Turkey.
| |
Collapse
|
60
|
Zhang Q, Tang X, Zhou Y, Chen X, Peng K, Jiang R, Liu Z, Song X, Xia H. LINC01060 knockdown inhibits osteosarcoma cell malignant behaviors in vitro and tumor growth and metastasis in vivo through the PI3K/Akt signaling. Cancer Biol Ther 2023; 24:2198904. [PMID: 37211864 PMCID: PMC10208149 DOI: 10.1080/15384047.2023.2198904] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2022] [Revised: 02/07/2023] [Accepted: 03/28/2023] [Indexed: 05/23/2023] Open
Abstract
Despite its low frequency, osteosarcoma is one of the deadliest malignancies in children and adolescents. The phosphatidylinositol 3-kinase (PI3K)/Akt signaling activation and epithelial-to-mesenchymal transition (EMT) are critical issues during osteosarcoma development. This study found long intergenic non-protein coding RNA 1060 (LINC01060) to be an EMT-related long non-coding RNA (lncRNA) up-regulated in osteosarcoma; higher LINC01060 expression was linked to a worse prognosis in osteosarcoma patients. In vitro, knocking down LINC01060 significantly inhibits osteosarcoma cell malignant behaviors, including hyperproliferation, invasion, migration, and EMT. In vivo, LINC01060 knockdown inhibited tumor growth and metastasis, and suppressed PI3K and Akt phosphorylation. In osteosarcoma cells, Akt agonist SC79 exerted opposite effects to those of LINC01060 knockdown through the promotion of cell viability, cell migration, and cell invasion. Moreover, the Akt agonist SC79 partially eliminated LINC01060 knockdown effects on osteosarcoma cells, suggesting that LINC01060 exerts its effects through the PI3K/Akt signaling. Therefore, it is deduced that LINC01060 is overexpressed in osteosarcoma. In vitro, LINC01060 knockdown inhibits cancer cell malignant behaviors; in vivo, LINC01060 knockdown inhibits tumor development and metastasis. The PI3K/Akt signaling is involved in LINC01060 functions in osteosarcoma.
Collapse
Affiliation(s)
- Qiang Zhang
- Department of Orthopedics, Xiangtan Central Hospital, Xiangtan, Hunan, China
| | - Xinqiao Tang
- Department of Orthopedics, Xiangtan Central Hospital, Xiangtan, Hunan, China
| | - Yi Zhou
- Department of Orthopedics, Xiangtan Central Hospital, Xiangtan, Hunan, China
| | - Xiaoming Chen
- Department of Orthopedics, Xiangtan Central Hospital, Xiangtan, Hunan, China
| | - Ke Peng
- Department of Spine Surgery, Xiangtan Central Hospital, Xiangtan, Hunan, China
| | - Ruizhong Jiang
- Department of Orthopedics, Xiangtan Central Hospital, Xiangtan, Hunan, China
| | - Zhong Liu
- Department of Orthopedics, Xiangtan Central Hospital, Xiangtan, Hunan, China
| | - Xiaoxia Song
- Department of Respiratory Medicine, Xiangtan Central Hospital, Xiangtan, Hunan, China
| | - Hong Xia
- Department of Orthopedics, Xiangtan Central Hospital, Xiangtan, Hunan, China
| |
Collapse
|
61
|
Loeffler E, Ancel J, Dalstein V, Deslée G, Polette M, Nawrocki-Raby B. HER2 Alterations in Non-Small Cell Lung Cancer: Biologico-Clinical Consequences and Interest in Therapeutic Strategies. Life (Basel) 2023; 14:64. [PMID: 38255679 PMCID: PMC10820545 DOI: 10.3390/life14010064] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Revised: 12/20/2023] [Accepted: 12/21/2023] [Indexed: 01/24/2024] Open
Abstract
Lung cancer stands as the first cause of death by cancer in the world. Despite the improvement in patients' outcomes in the past decades through the development of personalized medicine approaches, a substantial portion of patients remains ineligible for targeted therapies due to the lack of a "druggable" molecular target. HER2, a receptor tyrosine kinase member of the EGFR/ErbB family, is known to show oncogenic properties. In this review, we focus on the different HER2 dysregulation mechanisms that have been observed in non-small cell lung cancer (NSCLC): gene mutation, gene amplification, protein overexpression and protein hyper-phosphorylation, the latter suggesting that HER2 dysregulation can occur independently of any molecular aberration. These HER2 alterations inevitably have consequences on tumor biology. Here, we discuss how they are not only involved in abnormal proliferation and survival of cancer cells but also potentially in increased angiogenic properties, mesenchymal features and tumor immune escape. Finally, we review the impact of these HER2 alterations in various therapeutic approaches. While standard chemotherapy and groundbreaking immunotherapy seem rather ineffective for HER2-altered NSCLCs, the development of HER2-targeted therapies such as tyrosine kinase inhibitors, anti-HER2 antibodies and especially antibody-drug conjugates could provide new hopes for patients.
Collapse
Affiliation(s)
- Emma Loeffler
- Université de Reims Champagne Ardenne, Inserm, UMR-S 1250 P3Cell, SFR CAP Santé, 51092 Reims, France; (E.L.); (J.A.); (V.D.); (G.D.); (M.P.)
| | - Julien Ancel
- Université de Reims Champagne Ardenne, Inserm, UMR-S 1250 P3Cell, SFR CAP Santé, 51092 Reims, France; (E.L.); (J.A.); (V.D.); (G.D.); (M.P.)
- CHU de Reims, Hôpital Maison-Blanche, Service de Pneumologie, 51092 Reims, France
| | - Véronique Dalstein
- Université de Reims Champagne Ardenne, Inserm, UMR-S 1250 P3Cell, SFR CAP Santé, 51092 Reims, France; (E.L.); (J.A.); (V.D.); (G.D.); (M.P.)
- CHU de Reims, Pôle de Biologie Territoriale, Service de Pathologie, 51092 Reims, France
| | - Gaëtan Deslée
- Université de Reims Champagne Ardenne, Inserm, UMR-S 1250 P3Cell, SFR CAP Santé, 51092 Reims, France; (E.L.); (J.A.); (V.D.); (G.D.); (M.P.)
- CHU de Reims, Hôpital Maison-Blanche, Service de Pneumologie, 51092 Reims, France
| | - Myriam Polette
- Université de Reims Champagne Ardenne, Inserm, UMR-S 1250 P3Cell, SFR CAP Santé, 51092 Reims, France; (E.L.); (J.A.); (V.D.); (G.D.); (M.P.)
- CHU de Reims, Pôle de Biologie Territoriale, Service de Pathologie, 51092 Reims, France
| | - Béatrice Nawrocki-Raby
- Université de Reims Champagne Ardenne, Inserm, UMR-S 1250 P3Cell, SFR CAP Santé, 51092 Reims, France; (E.L.); (J.A.); (V.D.); (G.D.); (M.P.)
| |
Collapse
|
62
|
Bea-Mascato B, Gómez-Castañeda E, Sánchez-Corrales YE, Castellano S, Valverde D. Loss of the centrosomal protein ALMS1 alters lipid metabolism and the regulation of extracellular matrix-related processes. Biol Direct 2023; 18:84. [PMID: 38062477 PMCID: PMC10704752 DOI: 10.1186/s13062-023-00441-2] [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: 10/07/2023] [Accepted: 11/27/2023] [Indexed: 12/18/2023] Open
Abstract
BACKGROUND Alström syndrome (ALMS) is a rare autosomal recessive disease that is associated with mutations in ALMS1 gene. The main clinical manifestations of ALMS are retinal dystrophy, obesity, type 2 diabetes mellitus, dilated cardiomyopathy and multi-organ fibrosis, characteristic in kidneys and liver. Depletion of the protein encoded by ALMS1 has been associated with the alteration of different processes regulated via the primary cilium, such as the NOTCH or TGF-β signalling pathways. However, the cellular impact of these deregulated pathways in the absence of ALMS1 remains unknown. METHODS In this study, we integrated RNA-seq and proteomic analysis to determine the gene expression profile of hTERT-BJ-5ta ALMS1 knockout fibroblasts after TGF-β stimulation. In addition, we studied alterations in cross-signalling between the TGF-β pathway and the AKT pathway in this cell line. RESULTS We found that ALMS1 depletion affects the TGF-β pathway and its cross-signalling with other pathways such as PI3K/AKT, EGFR1 or p53. In addition, alterations associated with ALMS1 depletion clustered around the processes of extracellular matrix regulation and lipid metabolism in both the transcriptome and proteome. By studying the enriched pathways of common genes differentially expressed in the transcriptome and proteome, collagen fibril organisation, β-oxidation of fatty acids and eicosanoid metabolism emerged as key processes altered by the absence of ALMS1. Finally, an overactivation of the AKT pathway was determined in the absence of ALMS1 that could be explained by a decrease in PTEN gene expression. CONCLUSION ALMS1 deficiency disrupts cross-signalling between the TGF-β pathway and other dependent pathways in hTERT-BJ-5ta cells. Furthermore, altered cross-signalling impacts the regulation of extracellular matrix-related processes and fatty acid metabolism, and leads to over-activation of the AKT pathway.
Collapse
Affiliation(s)
- Brais Bea-Mascato
- CINBIO Facultad de Biología, Universidad de Vigo, Campus As Lagoas-Marcosende s/n, Vigo, 36310, Spain
- Grupo de Investigación en Enfermedades Raras y Medicina Pediátrica, Instituto de Investigación Sanitaria Galicia Sur (IIS Galicia Sur), SERGAS-UVIGO, Vigo, Spain
| | - Eduardo Gómez-Castañeda
- Molecular and Cellular Immunology Section, Great Ormond Street Institute of Child Health, University College London, London, UK
| | - Yara E Sánchez-Corrales
- Genetics and Genomic Medicine Department, Great Ormond Street Institute of Child Health, University College London, London, UK
| | - Sergi Castellano
- Genetics and Genomic Medicine Department, Great Ormond Street Institute of Child Health, University College London, London, UK
- Zayed Centre for Research into Rare Disease in Children, UCL Genomics, University College London, London, UK
| | - Diana Valverde
- CINBIO Facultad de Biología, Universidad de Vigo, Campus As Lagoas-Marcosende s/n, Vigo, 36310, Spain.
- Grupo de Investigación en Enfermedades Raras y Medicina Pediátrica, Instituto de Investigación Sanitaria Galicia Sur (IIS Galicia Sur), SERGAS-UVIGO, Vigo, Spain.
| |
Collapse
|
63
|
Proença C, Freitas M, Ribeiro D, Rufino AT, Fernandes E, Ferreira de Oliveira JMP. The role of flavonoids in the regulation of epithelial-mesenchymal transition in cancer: A review on targeting signaling pathways and metastasis. Med Res Rev 2023; 43:1878-1945. [PMID: 37147865 DOI: 10.1002/med.21966] [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: 07/22/2022] [Revised: 03/20/2023] [Accepted: 04/12/2023] [Indexed: 05/07/2023]
Abstract
One of the hallmarks of cancer is metastasis, a process that entails the spread of cancer cells to distant regions in the body, culminating in tumor formation in secondary organs. Importantly, the proinflammatory environment surrounding cancer cells further contributes to cancer cell transformation and extracellular matrix destruction. During metastasis, front-rear polarity and emergence of migratory and invasive features are manifestations of epithelial-mesenchymal transition (EMT). A variety of transcription factors (TFs) are implicated in the execution of EMT, the most prominent belonging to the Snail Family Transcriptional Repressor (SNAI) and Zinc Finger E-Box Binding Homeobox (ZEB) families of TFs. These TFs are regulated by interaction with specific microRNAs (miRNAs), as miR34 and miR200. Among the several secondary metabolites produced in plants, flavonoids constitute a major group of bioactive molecules, with several described effects including antioxidant, antiinflammatory, antidiabetic, antiobesogenic, and anticancer effects. This review scrutinizes the modulatory role of flavonoids on the activity of SNAI/ZEB TFs and on their regulatory miRNAs, miR-34, and miR-200. The modulatory role of flavonoids can attenuate mesenchymal features and stimulate epithelial features, thereby inhibiting and reversing EMT. Moreover, this modulation is concomitant with the attenuation of signaling pathways involved in diverse processes as cell proliferation, cell growth, cell cycle progression, apoptosis inhibition, morphogenesis, cell fate, cell migration, cell polarity, and wound healing. The antimetastatic potential of these versatile compounds is emerging and represents an opportunity for the synthesis of more specific and potent agents.
Collapse
Affiliation(s)
- Carina Proença
- LAQV, REQUIMTE, Laboratory of Applied Chemistry, Department of Chemical Sciences, Faculty of Pharmacy, University of Porto, Porto, Portugal
| | - Marisa Freitas
- LAQV, REQUIMTE, Laboratory of Applied Chemistry, Department of Chemical Sciences, Faculty of Pharmacy, University of Porto, Porto, Portugal
| | - Daniela Ribeiro
- LAQV, REQUIMTE, Laboratory of Applied Chemistry, Department of Chemical Sciences, Faculty of Pharmacy, University of Porto, Porto, Portugal
| | - Ana T Rufino
- LAQV, REQUIMTE, Laboratory of Applied Chemistry, Department of Chemical Sciences, Faculty of Pharmacy, University of Porto, Porto, Portugal
| | - Eduarda Fernandes
- LAQV, REQUIMTE, Laboratory of Applied Chemistry, Department of Chemical Sciences, Faculty of Pharmacy, University of Porto, Porto, Portugal
| | - José Miguel P Ferreira de Oliveira
- LAQV, REQUIMTE, Laboratory of Applied Chemistry, Department of Chemical Sciences, Faculty of Pharmacy, University of Porto, Porto, Portugal
| |
Collapse
|
64
|
Luo Y, Cao H, Lei C, Liu J. ST6GALNAC1 promotes the invasion and migration of breast cancer cells via the EMT pathway. Genes Genomics 2023; 45:1367-1376. [PMID: 37747641 DOI: 10.1007/s13258-023-01445-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2023] [Accepted: 08/28/2023] [Indexed: 09/26/2023]
Abstract
BACKGROUND A specific sialyl-transferases called ST6GALNAC1 has been proven to up-regulate abnormal O-glycosylation, which is strongly associated with tumorigenesis and cancer progression. However, the precise pathological outcome of ST6GALNAC1 expression in breast cancer cells remains unknown. Therefore, our study aims to investigate the functional role of ST6GALNAC1 and its impact on the epithelial-mesenchymal transition (EMT) pathway in breast cancer cells. METHODS Plasmids with siRNA were used to construct ST6GALNAC1 knockoff (si-ST6GALNAC1) MDA-MB-231 and MDA-MB-453 cells, while lentiviruses were used to construct ST6GALNAC1 over-expression (oe-ST6GALNAC1) MCF-7 and BT474 cells. Transfer efficiency was verified by Western Blot. Then we selected transfected cells and assessed the changes in cell proliferation, invasion, migration, and EMT markers. RESULTS The expression of ST6GALNAC1 significantly enhanced cell migration and invasion, which was confirmed by Wound Scratch Assay and Transwell Assay. Particularly, ST6GALNAC1 expression directly induced the EMT signaling pathway. E-cadherin was markedly decreased in oe-ST6GALNAC1 cells, accompanied by an up-regulation of mesenchymal markers including N-cadherin, snail, and ZEB1. However, no significant correlation was found between ST6GALNAC1 expression and cell proliferation. All of the outcomes were reversely validated in si-ST6GALNAC1 cells. CONCLUSIONS The expression of ST6GALNAC1 promotes cell migration and invasion probably by triggering the molecular process of the EMT pathway in breast cancer cells, which may provide new clues for designing novel molecular targeted drugs in breast cancer treatment.
Collapse
Affiliation(s)
- Yunzhao Luo
- Department of Breast Surgery, Beijing Chaoyang Hospital of Capital Medical University, No. 8 Workers' Stadium South Road, Beijing, 100020, China
| | - Heng Cao
- Department of Breast Surgery, Cancer Institute and Hospital, Chinese Academy of Medical Sciences, Beijing, 100021, China
| | - Chuqi Lei
- Department of Breast Surgery, Cancer Institute and Hospital, Chinese Academy of Medical Sciences, Beijing, 100021, China
| | - Jun Liu
- Department of Breast Surgery, Beijing Chaoyang Hospital of Capital Medical University, No. 8 Workers' Stadium South Road, Beijing, 100020, China.
| |
Collapse
|
65
|
Liu Y, Bai Y, Zhang J, Silva-Filho R, Zhu Q, Lei Z. Utilizing network pharmacology and experimental validation to explore the potential molecular mechanisms of raw Pinellia ternate in treating esophageal cancer. J Gastrointest Oncol 2023; 14:2006-2017. [PMID: 37969842 PMCID: PMC10643601 DOI: 10.21037/jgo-23-684] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/15/2023] [Accepted: 10/20/2023] [Indexed: 11/17/2023] Open
Abstract
Background Esophageal cancer (EC) is a highly lethal malignancy with a grim prognosis and high mortality rates, primarily treated through surgery and radiotherapy. Herbal remedies are emerging as complementary approaches in cancer therapy. Here, we explore the potential therapeutic benefits of Chinese medicine raw Pinellia ternata (RP) in EC using web-based pharmacological methods and cellular experiments. Methods The chemical components of RP were obtained by data mining via searches of the systematic pharmacology database, analysis platform, and literature on traditional Chinese medicine (TCM). The properties of the main components of RP were calculated using Traditional Chinese Medicine Systems Pharmacology Database and Analysis Platform (TCMSP). The potential targets of the components were mined and collected through multiple databases, and the relevant potential targets of efficacy were imported into Search Tool for the Retrieval of Interacting Genes/Proteins (STRING) database to obtain protein interactions. Gene Ontology (GO) enrichment analysis and Kyoto Encyclopedia of Genes and Genomes (KEGG) metabolic pathway enrichment analysis of the potential targets were performed through Metascape. A target-pathway network was established using Cytoscape, and topological analysis was performed on the network so as to obtain the relevant targets and pathways of RP in the treatment of EC. The inhibitory effect of RP on human EC cells was verified by cell experiments. Results Thirteen bioactive components of RP were screened, 87 related targets were obtained by construction, and 68 co-targets were obtained after taking intersection with EC related genes. The results of the protein-protein interaction (PPI) network analysis of the targets showed that the pharmacodynamic targets of hemicellulose might be closely related to the signaling pathways such as PI3K-Akt, FOS/JUN, and HIF-1. Meanwhile, GO and KEGG enrichment analysis showed that PI3K-Akt was also significantly enriched. The in vitro cellular experiments further indicated that raw hemicrania could inhibit EC through the PI3K-Akt signaling pathway. Conclusions The pharmacodynamic mechanism of RP in the treatment of esophageal carcinoma was preliminarily revealed, which provided ideas and the basis for further experimental study of RP in the treatment of esophageal carcinoma.
Collapse
Affiliation(s)
- Yanxin Liu
- Department of Pharmacy, Henan Provincial Hospital of Traditional Chinese Medicine (The Second Affiliated Hospital of Henan University of Traditional Chinese Medicine), Zhengzhou, China
| | - Yunfei Bai
- Department of Pharmacy, Henan Provincial Hospital of Traditional Chinese Medicine (The Second Affiliated Hospital of Henan University of Traditional Chinese Medicine), Zhengzhou, China
| | - Jinbang Zhang
- Department of Pharmacy, Henan Provincial Hospital of Traditional Chinese Medicine (The Second Affiliated Hospital of Henan University of Traditional Chinese Medicine), Zhengzhou, China
| | | | - Qingchun Zhu
- Department of Pharmacy, Henan Provincial Hospital of Traditional Chinese Medicine (The Second Affiliated Hospital of Henan University of Traditional Chinese Medicine), Zhengzhou, China
| | - Zhen Lei
- Central Laboratory, Henan Provincial Hospital of Traditional Chinese Medicine (The Second Affiliated Hospital of Henan University of Traditional Chinese Medicine), Zhengzhou, China
| |
Collapse
|
66
|
Han X, Yang L, Tian H, Ji Y. Machine learning developed a PI3K/Akt pathway-related signature for predicting prognosis and drug sensitivity in ovarian cancer. Aging (Albany NY) 2023; 15:11162-11183. [PMID: 37851341 PMCID: PMC10637788 DOI: 10.18632/aging.205119] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Accepted: 09/18/2023] [Indexed: 10/19/2023]
Abstract
BACKGROUND Ovarian cancer is one of the deadliest malignancies among females, generally having a poor prognosis. The PI3K/Akt pathway plays a vital role in the oncogenesis and progression of many types of cancer. Limited studies have fully clarified the role of PI3K/Akt pathway in the prognosis of ovarian cancer and its correlation with drug sensitivity. METHODS A prognostic PI3K/Akt pathway related signature (PRS) was constructed with 10 machine learning algorithms using TCGA, GSE14764, GSE26193, GSE26712, GSE63885 and GSE140082 datasets. Gaussian mixture and logistic regression were performed to identify the optimal models for classifying lymphatic and venous invasion. RESULTS The optimal prognostic PRS developed by Lasso + survivalSVM algorithm acted as an independent risk factor for overall survival (OS) of ovarian cancer patients and had a good performance in evaluating OS rate of ovarian cancer patients. Significant correlation was obtained between PRS-based risk score and Immune score, ESTIMATE score, immune cells and cancer-related hallmarks. Low risk score indicated a lower immune escape score, TIDE score, and higher PD1&CTLA4 immunophenoscore in ovarian cancer. Moreover, PRS-based risk score acted as an indicator for drug sensitivity in the immunotherapy and chemotherapy of ovarian cancer patients. CONCLUSIONS All in all, our study developed a prognostic PRS showing powerful and good performance in predicting clinical outcome of ovarian cancer patients. PRS could serve as an indicator for drug sensitivity in the chemotherapy and immunotherapy.
Collapse
Affiliation(s)
- Xiaofang Han
- Core Laboratory, Shanxi Provincial People's Hospital (Fifth Hospital) of Shanxi Medical University, Taiyuan 030012, China
| | - Liu Yang
- Core Laboratory, Shanxi Provincial People's Hospital (Fifth Hospital) of Shanxi Medical University, Taiyuan 030012, China
| | - Hui Tian
- Core Laboratory, Shanxi Provincial People's Hospital (Fifth Hospital) of Shanxi Medical University, Taiyuan 030012, China
| | - Yuanyuan Ji
- Core Laboratory, Shanxi Provincial People's Hospital (Fifth Hospital) of Shanxi Medical University, Taiyuan 030012, China
| |
Collapse
|
67
|
Semik-Gurgul E, Szmatoła T, Gurgul A, Pawlina-Tyszko K, Gałuszka A, Pędziwiatr R, Witkowski M, Ząbek T. Methylome and transcriptome data integration reveals aberrantly regulated genes in equine sarcoids. Biochimie 2023; 213:100-113. [PMID: 37211255 DOI: 10.1016/j.biochi.2023.05.008] [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: 03/27/2023] [Revised: 05/08/2023] [Accepted: 05/15/2023] [Indexed: 05/23/2023]
Abstract
DNA methylation is a key mechanism in transcription regulation, and aberrant methylation is a common and important mechanism in tumor initiation, maintenance, and progression. To find genes that are aberrantly regulated by altered methylation in horse sarcoids, we used reduced representation bisulfite sequencing (RRBS) accompanied by RNA sequencing (RNA-Seq) for methylome (whole genome DNA methylation sequencing) and transcriptome profiling, respectively. We found that the DNA methylation level was generally lower in lesion samples than in controls. In the analyzed samples, a total of 14,692 differentially methylated sites (DMSs) in the context of CpG (where cytosine and guanine are separated by a phosphate), and 11,712 differentially expressed genes (DEGs) were identified. The integration of the methylome and transcriptome data suggests that aberrant DNA methylation may be involved in the deregulation of expression of the 493 genes in equine sarcoid. Furthermore, enrichment analysis of the genes demonstrated the activation of multiple molecular pathways related to extracellular matrix (ECM), oxidative phosphorylation (OXPHOS), immune response, and disease processes that can be related to tumor progression. The results provide further insight into the epigenetic alterations in equine sarcoids and provide a valuable resource for follow-up studies to identify biomarkers for predicting susceptibility to this common condition in horses.
Collapse
Affiliation(s)
- Ewelina Semik-Gurgul
- Department of Animal Molecular Biology, National Research Institute of Animal Production, Krakowska 1 St., 32-083, Balice, Poland.
| | - Tomasz Szmatoła
- Department of Animal Molecular Biology, National Research Institute of Animal Production, Krakowska 1 St., 32-083, Balice, Poland; Center for Experimental and Innovative Medicine, University of Agriculture in Krakow, Redzina 1c, 30-248, Krakow, Poland
| | - Artur Gurgul
- Center for Experimental and Innovative Medicine, University of Agriculture in Krakow, Redzina 1c, 30-248, Krakow, Poland
| | - Klaudia Pawlina-Tyszko
- Department of Animal Molecular Biology, National Research Institute of Animal Production, Krakowska 1 St., 32-083, Balice, Poland
| | - Anna Gałuszka
- Department of Animal Anatomy and Preclinical Sciences, University Centre of Veterinary Medicine JU-UA, University of Agriculture in Krakow, Mickiewicza 24/28, 30-059, Krakow, Poland
| | - Rafał Pędziwiatr
- University Centre of Veterinary Medicine JU-UA, University of Agriculture in Krakow, Mickiewicza 24/28, 30-059, Krakow, Poland; Equine Vet Clinic EQUI-VET, Stogniowice 55A St., 32-100 Stogniowice, Poland
| | - Maciej Witkowski
- University Centre of Veterinary Medicine JU-UA, University of Agriculture in Krakow, Mickiewicza 24/28, 30-059, Krakow, Poland; Equine Hospital on the Racing Truck, Sluzewiec, Pulawska 266, 02-684, Warszawa, Poland
| | - Tomasz Ząbek
- Department of Animal Molecular Biology, National Research Institute of Animal Production, Krakowska 1 St., 32-083, Balice, Poland
| |
Collapse
|
68
|
Vinita N, Devan U, Durgadevi S, Anitha S, Govarthanan M, Antony Joseph Velanganni A, Jeyakanthan J, Arul Prakash P, Mohamed Jaabir MS, Kumar P. Impact of Surface Charge-Tailored Gold Nanorods for Selective Targeting of Mitochondria in Breast Cancer Cells Using Photodynamic Therapy. ACS OMEGA 2023; 8:33229-33241. [PMID: 37744785 PMCID: PMC10515365 DOI: 10.1021/acsomega.2c06731] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/19/2022] [Accepted: 03/10/2023] [Indexed: 09/26/2023]
Abstract
Herein, the impact of surface charge tailored of gold nanorods (GNRs) on breast cancer cells (MCF-7 and MDA-MB-231) upon conjugation with triphenylphosphonium (TPP) for improved photodynamic therapy (PDT) targeting mitochondria was studied. The salient features of the study are as follows: (i) positive (CTAB@GNRs) and negative (PSS-CTAB@GNRs) surface-charged gold nanorods were developed and characterized; (ii) the mitochondrial targeting efficiency of gold nanorods was improved by conjugating TPP molecules; (iii) the conjugated nanoprobes (TPP-CTAB@GNRs and TPP-PSS-CTAB@GNRs) were evaluated for PDT in the presence of photosensitizer (PS), 5-aminolevulinic acid (5-ALA) in breast cancer cells; (iv) both nanoprobes (TPP-CTAB@GNRs and TPP-PSS-CTAB@GNRs) induce apoptosis, damage DNA, generate reactive oxygen species, and decrease mitochondrial membrane potential upon 5-ALA-based PDT; and (v) 5-ALA-PDT of two nanoprobes (TPP-CTAB@GNRs and TPP-PSS-CTAB@GNRs) impact cell signaling (PI3K/AKT) pathway by upregulating proapoptotic genes and proteins. Based on the results, we confirm that the positively charged (rapid) nanoprobes are more advantageous than their negatively (slow) charged nanoprobes. However, depending on the kind and degree of cancer, both nanoprobes can serve as efficient agents for delivering anticancer therapy.
Collapse
Affiliation(s)
- Nadar
Manimaran Vinita
- Food
Chemistry and Molecular Cancer Biology Lab, Department of Animal Health
and Management, Alagappa University, Karaikudi 630 003, Tamil Nadu, India
| | - Umapathy Devan
- Molecular
Oncology Laboratory, Department of Biochemistry, Bharathidasan University, Tiruchirappalli 620 024, Tamil Nadu, India
| | - Sabapathi Durgadevi
- Food
Chemistry and Molecular Cancer Biology Lab, Department of Animal Health
and Management, Alagappa University, Karaikudi 630 003, Tamil Nadu, India
| | - Selvaraj Anitha
- Food
Chemistry and Molecular Cancer Biology Lab, Department of Animal Health
and Management, Alagappa University, Karaikudi 630 003, Tamil Nadu, India
| | - Muthusamy Govarthanan
- Department
of Environmental Engineering, Kyungpook
National University, Deagu 41566, Republic
of Korea
- Department
of Biomaterials, Saveetha Dental College and Hospital, Saveetha Institute of Medical and Technical Sciences, Chennai 600077, Tamil Nadu, India
| | | | - Jeyaraman Jeyakanthan
- Department
of Bioinformatics, Alagappa University, Karaikudi 630 003, Tamil Nadu, India
| | - Pitchan Arul Prakash
- PG
and Research
Department of Biotechnology and Microbiology, The National College, Tiruchirappalli 620001, Tamil Nadu, India
| | - Mohamed Sultan Mohamed Jaabir
- PG
and Research
Department of Biotechnology and Microbiology, The National College, Tiruchirappalli 620001, Tamil Nadu, India
| | - Ponnuchamy Kumar
- Food
Chemistry and Molecular Cancer Biology Lab, Department of Animal Health
and Management, Alagappa University, Karaikudi 630 003, Tamil Nadu, India
| |
Collapse
|
69
|
Fasoulakis Z, Psarommati MZ, Papapanagiotou A, Pergialiotis V, Koutras A, Douligeris A, Mortaki A, Mihail A, Theodora M, Stavros S, Karakalpakis D, Papamihail M, Kontomanolis EN, Daskalakis G, Antsaklis P. MicroRNAs Can Influence Ovarian Cancer Progression by Dysregulating Integrin Activity. Cancers (Basel) 2023; 15:4449. [PMID: 37760437 PMCID: PMC10526761 DOI: 10.3390/cancers15184449] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2023] [Revised: 08/26/2023] [Accepted: 08/30/2023] [Indexed: 09/29/2023] Open
Abstract
Ovarian cancer is a deadly disease that affects thousands of women worldwide. Integrins, transmembrane receptors that mediate cell adhesion and signaling, play important roles in ovarian cancer progression, metastasis, and drug resistance. Dysregulated expression of integrins is implicated in various cellular processes, such as cell migration, invasion, and proliferation. Emerging evidence suggests that microRNAs (miRNAs) can regulate integrin expression and function, thus affecting various physiological and pathological processes, including ovarian cancer. In this article, we review the current understanding of integrin-mediated cellular processes in ovarian cancer and the roles of miRNAs in regulating integrins. We also discuss the therapeutic potential of targeting miRNAs that regulate integrins for the treatment of ovarian cancer. Targeting miRNAs that regulate integrins or downstream signaling pathways of integrins may provide novel therapeutic strategies for inhibiting integrin-mediated ovarian cancer progression.
Collapse
Affiliation(s)
- Zacharias Fasoulakis
- 1st Department of Obstetrics and Gynecology, National and Kapodistrian University of Athens, 115 28 Athens, Greece; (V.P.); (A.K.); (A.D.); (A.M.); (A.M.); (M.T.); (D.K.); (M.P.)
| | - Michaela-Zoi Psarommati
- Department of Obstetrics and Gynecology, Democritus University of Thrace, 681 00 Alexandroupolis, Greece; (M.-Z.P.); (E.N.K.)
| | - Angeliki Papapanagiotou
- Laboratory of Chemistry Biology, National and Kapodistrian University of Athens, 115 28 Athens, Greece
| | - Vasilios Pergialiotis
- 1st Department of Obstetrics and Gynecology, National and Kapodistrian University of Athens, 115 28 Athens, Greece; (V.P.); (A.K.); (A.D.); (A.M.); (A.M.); (M.T.); (D.K.); (M.P.)
| | - Antonios Koutras
- 1st Department of Obstetrics and Gynecology, National and Kapodistrian University of Athens, 115 28 Athens, Greece; (V.P.); (A.K.); (A.D.); (A.M.); (A.M.); (M.T.); (D.K.); (M.P.)
| | - Athanasios Douligeris
- 1st Department of Obstetrics and Gynecology, National and Kapodistrian University of Athens, 115 28 Athens, Greece; (V.P.); (A.K.); (A.D.); (A.M.); (A.M.); (M.T.); (D.K.); (M.P.)
| | - Anastasia Mortaki
- 1st Department of Obstetrics and Gynecology, National and Kapodistrian University of Athens, 115 28 Athens, Greece; (V.P.); (A.K.); (A.D.); (A.M.); (A.M.); (M.T.); (D.K.); (M.P.)
| | - Antonios Mihail
- 1st Department of Obstetrics and Gynecology, National and Kapodistrian University of Athens, 115 28 Athens, Greece; (V.P.); (A.K.); (A.D.); (A.M.); (A.M.); (M.T.); (D.K.); (M.P.)
| | - Marianna Theodora
- 1st Department of Obstetrics and Gynecology, National and Kapodistrian University of Athens, 115 28 Athens, Greece; (V.P.); (A.K.); (A.D.); (A.M.); (A.M.); (M.T.); (D.K.); (M.P.)
| | - Sofoklis Stavros
- 3rd Department of Obstetrics and Gynecology, National and Kapodistrian University of Athens, Medical School, Attikon Hospital, 124 62 Athens, Greece;
| | - Defkalion Karakalpakis
- 1st Department of Obstetrics and Gynecology, National and Kapodistrian University of Athens, 115 28 Athens, Greece; (V.P.); (A.K.); (A.D.); (A.M.); (A.M.); (M.T.); (D.K.); (M.P.)
| | - Maria Papamihail
- 1st Department of Obstetrics and Gynecology, National and Kapodistrian University of Athens, 115 28 Athens, Greece; (V.P.); (A.K.); (A.D.); (A.M.); (A.M.); (M.T.); (D.K.); (M.P.)
| | - Emmanuel N. Kontomanolis
- Department of Obstetrics and Gynecology, Democritus University of Thrace, 681 00 Alexandroupolis, Greece; (M.-Z.P.); (E.N.K.)
| | - George Daskalakis
- 1st Department of Obstetrics and Gynecology, National and Kapodistrian University of Athens, 106 76 Athens, Greece; (G.D.); (P.A.)
| | - Panos Antsaklis
- 1st Department of Obstetrics and Gynecology, National and Kapodistrian University of Athens, 106 76 Athens, Greece; (G.D.); (P.A.)
| |
Collapse
|
70
|
Trasierras AM, Luna JM, Ventura S. A contrast set mining based approach for cancer subtype analysis. Artif Intell Med 2023; 143:102590. [PMID: 37673572 DOI: 10.1016/j.artmed.2023.102590] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2022] [Revised: 05/24/2023] [Accepted: 05/30/2023] [Indexed: 09/08/2023]
Abstract
The task of detecting common and unique characteristics among different cancer subtypes is an important focus of research that aims to improve personalized therapies. Unlike current approaches mainly based on predictive techniques, our study aims to improve the knowledge about the molecular mechanisms that descriptively led to cancer, thus not requiring previous knowledge to be validated. Here, we propose an approach based on contrast set mining to capture high-order relationships in cancer transcriptomic data. In this way, we were able to extract valuable insights from several cancer subtypes in the form of highly specific genetic relationships related to functional pathways affected by the disease. To this end, we have divided several cancer gene expression databases by the subtype associated with each sample to detect which gene groups are related to each cancer subtype. To demonstrate the potential and usefulness of the proposed approach we have extensively analysed RNA-Seq gene expression data from breast, kidney, and colon cancer subtypes. The possible role of the obtained genetic relationships was further evaluated through extensive literature research, while its prognosis was assessed via survival analysis, finding gene expression patterns related to survival in various cancer subtypes. Some gene associations were described in the literature as potential cancer biomarkers while other results have been not described yet and could be a starting point for future research.
Collapse
Affiliation(s)
- A M Trasierras
- Department of Computer Science and Numerical Analysis, Andalusian Research Institute in Data Science and Computational Intelligence (DaSCI), Spain; Maimonides Biomedical Research Institute of Cordoba, IMIBIC, University of Cordoba, Córdoba, 14071, Spain; Phytoplant Research S.L.U, Departamento Tecnología y Control, Rabanales 21-Parque Científico Tecnológico de Córdoba, Calle Astrónoma Cecilia Payne, Córdoba, Spain
| | - J M Luna
- Department of Computer Science and Numerical Analysis, Andalusian Research Institute in Data Science and Computational Intelligence (DaSCI), Spain; Maimonides Biomedical Research Institute of Cordoba, IMIBIC, University of Cordoba, Córdoba, 14071, Spain
| | - S Ventura
- Department of Computer Science and Numerical Analysis, Andalusian Research Institute in Data Science and Computational Intelligence (DaSCI), Spain; Maimonides Biomedical Research Institute of Cordoba, IMIBIC, University of Cordoba, Córdoba, 14071, Spain.
| |
Collapse
|
71
|
Yang YC, Lin YW, Lee WJ, Lai FR, Ho KH, Chu CY, Hua KT, Chen JQ, Tung MC, Hsiao M, Wen YC, Chien MH. The RNA-binding protein KSRP aggravates malignant progression of clear cell renal cell carcinoma through transcriptional inhibition and post-transcriptional destabilization of the NEDD4L ubiquitin ligase. J Biomed Sci 2023; 30:68. [PMID: 37580757 PMCID: PMC10424398 DOI: 10.1186/s12929-023-00949-9] [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: 03/07/2023] [Accepted: 07/16/2023] [Indexed: 08/16/2023] Open
Abstract
BACKGROUND KH-type splicing regulatory protein (KHSRP, also called KSRP), a versatile RNA-binding protein, plays a critical role in various physiological and pathological conditions through modulating gene expressions at multiple levels. However, the role of KSRP in clear cell renal cell carcinoma (ccRCC) remains poorly understood. METHODS KSRP expression was detected by a ccRCC tissue microarray and evaluated by an in silico analysis. Cell loss-of-function and gain-of-function, colony-formation, anoikis, and transwell assays, and an orthotopic bioluminescent xenograft model were conducted to determine the functional role of KRSP in ccRCC progression. Micro (mi)RNA and complementary (c)DNA microarrays were used to identify downstream targets of KSRP. Western blotting, quantitative real-time polymerase chain reaction, and promoter- and 3-untranslated region (3'UTR)-luciferase reporter assays were employed to validate the underlying mechanisms of KSRP which aggravate progression of ccRCC. RESULTS Our results showed that dysregulated high levels of KSRP were correlated with advanced clinical stages, larger tumor sizes, recurrence, and poor prognoses of ccRCC. Neural precursor cell-expressed developmentally downregulated 4 like (NEDD4L) was identified as a novel target of KSRP, which can reverse the protumorigenic and prometastatic characteristics as well as epithelial-mesenchymal transition (EMT) promotion by KSRP in vitro and in vivo. Molecular studies revealed that KSRP can decrease NEDD4L messenger (m)RNA stability via inducing mir-629-5p upregulation and directly targeting the AU-rich elements (AREs) of the 3'UTR. Moreover, KSRP was shown to transcriptionally suppress NEDD4L via inducing the transcriptional repressor, Wilm's tumor 1 (WT1). In the clinic, ccRCC samples revealed a positive correlation between KSRP and mesenchymal-related genes, and patients expressing high KSRP and low NEDD4L had the worst prognoses. CONCLUSION The current findings unveil novel mechanisms of KSRP which promote malignant progression of ccRCC through transcriptional inhibition and post-transcriptional destabilization of NEDD4L transcripts. Targeting KSRP and its pathways may be a novel pharmaceutical intervention for ccRCC.
Collapse
Affiliation(s)
- Yi-Chieh Yang
- Department of Medical Research, Tungs' Taichung MetroHarbor Hospital, Taichung, Taiwan
- Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, 250 Wu Hsing Street, Taipei, 11031, Taiwan
| | - Yung-Wei Lin
- International Master/PhD Program in Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
- Department of Urology, Wan Fang Hospital, Taipei Medical University, 111, Section 3, Hsing Long Road, Taipei, 11696, Taiwan
- Department of Urology, School of Medicine, College of Medicine and TMU Research Center of Urology and Kidney (TMU-RCUK), Taipei Medical University, Taipei, Taiwan
| | - Wei-Jiunn Lee
- Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, 250 Wu Hsing Street, Taipei, 11031, Taiwan
- Department of Urology, School of Medicine, College of Medicine and TMU Research Center of Urology and Kidney (TMU-RCUK), Taipei Medical University, Taipei, Taiwan
- Department of Medical Education and Research, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan
| | - Feng-Ru Lai
- Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, 250 Wu Hsing Street, Taipei, 11031, Taiwan
| | - Kuo-Hao Ho
- Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, 250 Wu Hsing Street, Taipei, 11031, Taiwan
| | - Chih-Ying Chu
- Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, 250 Wu Hsing Street, Taipei, 11031, Taiwan
| | - Kuo-Tai Hua
- Graduate Institute of Toxicology, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Ji-Qing Chen
- Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, 250 Wu Hsing Street, Taipei, 11031, Taiwan
- Department of Cancer Biology, Geisel School of Medicine at Dartmouth, Lebanon, NH, USA
| | - Min-Che Tung
- Department of Surgery, Tungs' Taichung Metro Harbor Hospital, Taichung, Taiwan
| | - Michael Hsiao
- Genomics Research Center, Academia Sinica, Taipei, Taiwan
| | - Yu-Ching Wen
- Department of Urology, Wan Fang Hospital, Taipei Medical University, 111, Section 3, Hsing Long Road, Taipei, 11696, Taiwan.
- Department of Urology, School of Medicine, College of Medicine and TMU Research Center of Urology and Kidney (TMU-RCUK), Taipei Medical University, Taipei, Taiwan.
| | - Ming-Hsien Chien
- Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, 250 Wu Hsing Street, Taipei, 11031, Taiwan.
- TMU Research Center of Cancer Translational Medicine, Taipei Medical University, Taipei, Taiwan.
- Pulmonary Research Center, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan.
- Traditional Herbal Medicine Research Center, Taipei Medical University Hospital, Taipei, Taiwan.
| |
Collapse
|
72
|
Wang Y, Xie M, Lin F, Sheng X, Zhao X, Zhu X, Wang Y, Lu B, Chen J, Zhang T, Wan X, Liu W, Sun X. Nomogram of uveal melanoma as prediction model of metastasis risk. Heliyon 2023; 9:e18956. [PMID: 37609406 PMCID: PMC10440531 DOI: 10.1016/j.heliyon.2023.e18956] [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: 01/21/2023] [Revised: 08/01/2023] [Accepted: 08/03/2023] [Indexed: 08/24/2023] Open
Abstract
Background Since the poor prognosis of uveal melanoma with distant metastasis, we intended to screen out possible biomarkers for uveal melanoma metastasis risk and establish a nomogram model for predicting the risk of uveal melanoma (UVM) metastasis. Methods Two datasets of UVM (GSE84976, GSE22138) were selected. Data was analyzed by R language, CTD database and GEPIA. Results The co-upregulated genes of two datasets, HTR2B, CHAC1, AHNAK2, and PTP4A3 were identified using a Venn diagram. These biomarkers are combined with clinical characteristics, and Lasso regression was conducted to filter the metastasis-related biomarkers. HTR2B, CHAC1, AHNAK2, PTP4A3, tumor thickness, and retinal detachment (RD) were selected to establish the nomogram. Conclusion Our study provides a comprehensive predictive model and personalized risk estimation tool for assessment of 3-year metastasis risk of UVM with a better accuracy.
Collapse
Affiliation(s)
- Yimin Wang
- Shanghai General Hospital,Shanghai Jiao Tong University School of Medicine, China
- National Clinical Research Center for Eye Disease, China
- Shanghai Key Laboratory of Ocular Fundus Diseases, China
- Shanghai Engineering Center for Visual Science and Photomedicine, China
- Department of Ophthalmology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Minyue Xie
- Beijing Tongren Hospital, Capital Medical University, China
| | - Feng Lin
- Eye Institute and Department of Ophthalmology, Eye & ENT Hospital, Fudan University, China
| | - Xiaonan Sheng
- Department of Breast Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, PR China
| | - Xiaohuan Zhao
- Shanghai General Hospital,Shanghai Jiao Tong University School of Medicine, China
- National Clinical Research Center for Eye Disease, China
- Shanghai Key Laboratory of Ocular Fundus Diseases, China
- Shanghai Engineering Center for Visual Science and Photomedicine, China
| | - Xinyue Zhu
- Shanghai General Hospital,Shanghai Jiao Tong University School of Medicine, China
- National Clinical Research Center for Eye Disease, China
- Shanghai Key Laboratory of Ocular Fundus Diseases, China
- Shanghai Engineering Center for Visual Science and Photomedicine, China
| | - Yuwei Wang
- Shanghai General Hospital,Shanghai Jiao Tong University School of Medicine, China
- National Clinical Research Center for Eye Disease, China
- Shanghai Key Laboratory of Ocular Fundus Diseases, China
- Shanghai Engineering Center for Visual Science and Photomedicine, China
| | - Bing Lu
- Shanghai General Hospital,Shanghai Jiao Tong University School of Medicine, China
- National Clinical Research Center for Eye Disease, China
- Shanghai Key Laboratory of Ocular Fundus Diseases, China
- Shanghai Engineering Center for Visual Science and Photomedicine, China
| | - Jieqiong Chen
- Shanghai General Hospital,Shanghai Jiao Tong University School of Medicine, China
- National Clinical Research Center for Eye Disease, China
- Shanghai Key Laboratory of Ocular Fundus Diseases, China
- Shanghai Engineering Center for Visual Science and Photomedicine, China
| | - Ting Zhang
- Shanghai General Hospital,Shanghai Jiao Tong University School of Medicine, China
- National Clinical Research Center for Eye Disease, China
- Shanghai Key Laboratory of Ocular Fundus Diseases, China
| | - Xiaoling Wan
- Shanghai General Hospital,Shanghai Jiao Tong University School of Medicine, China
- National Clinical Research Center for Eye Disease, China
- Shanghai Key Laboratory of Ocular Fundus Diseases, China
| | - Wenjia Liu
- Shanghai General Hospital,Shanghai Jiao Tong University School of Medicine, China
- National Clinical Research Center for Eye Disease, China
- Shanghai Key Laboratory of Ocular Fundus Diseases, China
- Shanghai Engineering Center for Visual Science and Photomedicine, China
| | - Xiaodong Sun
- Shanghai General Hospital,Shanghai Jiao Tong University School of Medicine, China
- National Clinical Research Center for Eye Disease, China
- Shanghai Key Laboratory of Ocular Fundus Diseases, China
- Shanghai Engineering Center for Visual Science and Photomedicine, China
| |
Collapse
|
73
|
Fang Y, Zhang Q, Chen C, Chen Z, Zheng R, She C, Zhang R, Wu J. Identification and comprehensive analysis of epithelial-mesenchymal transition related target genes of miR-222-3p in breast cancer. Front Oncol 2023; 13:1189635. [PMID: 37546414 PMCID: PMC10400091 DOI: 10.3389/fonc.2023.1189635] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2023] [Accepted: 06/16/2023] [Indexed: 08/08/2023] Open
Abstract
Background Epithelial-mesenchymal transition (EMT) is a crucial mechanism that microRNA-222-3p (miR-222-3p) promotes breast cancer (BC) progression. Our study aimed to identify EMT-associated target genes (ETGs) of miR-222-3p for further analysis of their roles in BC based on bioinformatics tools. Methods Based on bioinformatics analysis, we identified 10 core ETGs of miR-222-3p. Then, we performed a comprehensive analysis of 10 ETGs and miR-222-3p, including pathway enrichment analysis of ETGs, differential expression, clinical significance, correlation with immune cell infiltration, immune checkpoint genes (ICGs) expression, tumor mutational burden (TMB), microsatellite instability (MSI), stemness, drug sensitivity, and genetic alteration. Results The expression of miR222-3p in basal-like BC was significantly higher than in other subtypes of BC and the normal adjacent tissue. Pathway analysis suggested that the ETGs might regulate the EMT process via the PI3K-Akt and HIF-1 signaling pathway. Six of the 10 core ETGs of miR-222-3p identified were down-expressed in BC, which were EGFR, IL6, NRP1, NTRK2, LAMC2, and PIK3R1, and SERPINE1, MUC1, MMP11, and BIRC5 were up-expressed in BC, which also showed potential diagnostic values in BC. Prognosis analysis revealed that higher NTRK2 and PIK3R1 expressions were related to a better prognosis, and higher BIRC5 and miR-222-3p expressions were related to a worse prognosis. Most ETGs and miR-222-3p were positively correlated with various infiltration of various immune cells and ICGs expression. Lower TMB scores were correlated with higher expression of MUC1 and NTRK2, and higher BIRC5 was related to a higher TMB score. Lower expression of MUC1, NTRK2, and PIK3R1 were associated with higher MSI scores. Higher expression of ETGs was associated with lower mRNAsi scores, except BIRC5 and miR-222-3p conversely. Most ETGs and miR-222-3p expression were negatively correlated with the drug IC50 values. The analysis of the genetic alteration of the ETGs suggested that amplification was the main genetic alteration of eight ETGs except for NTRK2 and PIK3R1. Conclusion MiR-222-3p might be a specific biomarker of basal-like BC. We successfully identify 10 core ETGs of miR-222-3p, some might be useful diagnostic and prognostic biomarkers. The comprehensive analysis of 10 ETGs and miR-222-3p indicated that they might be involved in the development of BC, which might be novel therapeutic targets for the treatment of BC.
Collapse
Affiliation(s)
- Yutong Fang
- The Breast Center, Cancer Hospital of Shantou University Medical College, Shantou, Guangdong, China
- The Department of Central Laboratory, Cancer Hospital of Shantou University Medical College, Shantou, Guangdong, China
| | - Qunchen Zhang
- The Breast Center, Cancer Hospital of Shantou University Medical College, Shantou, Guangdong, China
- The Department of Central Laboratory, Cancer Hospital of Shantou University Medical College, Shantou, Guangdong, China
| | - Chunfa Chen
- The Breast Center, Cancer Hospital of Shantou University Medical College, Shantou, Guangdong, China
- The Department of Central Laboratory, Cancer Hospital of Shantou University Medical College, Shantou, Guangdong, China
| | - Zexiao Chen
- The Breast Center, Cancer Hospital of Shantou University Medical College, Shantou, Guangdong, China
- The Department of Central Laboratory, Cancer Hospital of Shantou University Medical College, Shantou, Guangdong, China
| | - Rongji Zheng
- The Breast Center, Cancer Hospital of Shantou University Medical College, Shantou, Guangdong, China
- The Department of Central Laboratory, Cancer Hospital of Shantou University Medical College, Shantou, Guangdong, China
| | - Chuanghong She
- The Breast Center, Cancer Hospital of Shantou University Medical College, Shantou, Guangdong, China
- The Department of Central Laboratory, Cancer Hospital of Shantou University Medical College, Shantou, Guangdong, China
| | - Rendong Zhang
- The Breast Center, Cancer Hospital of Shantou University Medical College, Shantou, Guangdong, China
- The Department of Central Laboratory, Cancer Hospital of Shantou University Medical College, Shantou, Guangdong, China
| | - Jundong Wu
- The Breast Center, Cancer Hospital of Shantou University Medical College, Shantou, Guangdong, China
- The Department of Central Laboratory, Cancer Hospital of Shantou University Medical College, Shantou, Guangdong, China
| |
Collapse
|
74
|
Wang Y, Shi K, Tu J, Ke C, Chen N, Wang B, Liu Y, Zhou Z. Atractylenolide III Ameliorates Bile Duct Ligation-Induced Liver Fibrosis by Inhibiting the PI3K/AKT Pathway and Regulating Glutamine Metabolism. Molecules 2023; 28:5504. [PMID: 37513376 PMCID: PMC10383814 DOI: 10.3390/molecules28145504] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2023] [Revised: 07/13/2023] [Accepted: 07/17/2023] [Indexed: 07/30/2023] Open
Abstract
Liver fibrosis is one of the leading causes of hepatic sclerosis and hepatocellular carcinoma worldwide. However, the complex pathophysiological mechanisms of liver fibrosis are unknown, and no specific drugs are available to treat liver fibrosis. Atractylenolide III (ATL III) is a natural compound isolated from the plant Atractylodes lancea (Thunb.) DC. that possesses antioxidant properties and the ability to inhibit inflammatory responses. In this study, cholestatic hepatic fibrosis was induced in mice using a bile duct ligation (BDL) model and treated with 10 mg/kg and 50 mg/kg of ATL III via gavage for 14 days. ATL III significantly reduced the liver index, lowered serum ALT and AST levels, and reduced liver injury in bile-duct-ligated mice. In addition, ATL III significantly attenuated histopathological changes and reduced collagen deposition. ATL III reduced the expression of fibrosis-related genes α-smooth muscle actin (α-SMA), Collagen I (col1a1), Collagen IV (col4a2), and fibrosis-related proteins α-SMA and col1a1 in liver tissue. Using RNA sequencing (RNA-seq) to screen molecular targets and pathways, ATL III was found to affect the PI3K/AKT singling pathway by inhibiting the phosphorylation of PI3K and AKT, thereby ameliorating BDL-induced liver fibrosis. Gas chromatography-mass spectrometry (GC-MS) was used to evaluate the effect of ATL III on liver metabolites in BDL mice. ATL III further affected glutamine metabolism by down-regulating the activity of glutamine (GLS1) and glutamine metabolism. ATL III further affected glutamine metabolism by down-regulating the activity of glutaminase (GLS1), as well as glutamine metabolism. Therefore, we conclude that ATL III attenuates liver fibrosis by inhibiting the PI3K/AKT pathway and glutamine metabolism, suggesting that ATL III is a potential drug candidate for treating liver fibrosis.
Collapse
Affiliation(s)
- Yan Wang
- College of Pharmacy, Hubei University of Chinese Medicine, Wuhan 430065, China
| | - Kun Shi
- College of Pharmacy, Hubei University of Chinese Medicine, Wuhan 430065, China
| | - Jiyuan Tu
- College of Pharmacy, Hubei University of Chinese Medicine, Wuhan 430065, China
- Center for Hubei TCM Processing Technology Engineering, Wuhan 430065, China
| | - Chang Ke
- College of Pharmacy, Hubei University of Chinese Medicine, Wuhan 430065, China
| | - Niping Chen
- College of Pharmacy, Hubei University of Chinese Medicine, Wuhan 430065, China
| | - Bo Wang
- Hubei Institute for Drug Control, NMPA Key Laboratory of Quality Control of Chinese Medicine, Hubei Engineering Research Center for Drug Quality Control, Wuhan 430075, China
| | - Yanju Liu
- College of Pharmacy, Hubei University of Chinese Medicine, Wuhan 430065, China
- Center for Hubei TCM Processing Technology Engineering, Wuhan 430065, China
| | - Zhongshi Zhou
- College of Pharmacy, Hubei University of Chinese Medicine, Wuhan 430065, China
- Center for Hubei TCM Processing Technology Engineering, Wuhan 430065, China
| |
Collapse
|
75
|
Yuan M, Barefoot ME, Peterson K, Campbell MJ, Blancato JK, Chen M, Schmidt MO, Kiliti AJ, Fang HB, Wellstein A, Riegel AT, Sharif GM. Loss of ANCO1 Expression Regulates Chromatin Accessibility and Drives Progression of Early-Stage Triple-Negative Breast Cancer. Int J Mol Sci 2023; 24:11505. [PMID: 37511268 PMCID: PMC10380654 DOI: 10.3390/ijms241411505] [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: 06/09/2023] [Revised: 06/30/2023] [Accepted: 07/10/2023] [Indexed: 07/30/2023] Open
Abstract
Mutations in the gene ankyrin repeat domain containing 11 (ANKRD11/ANCO1) play a role in neurodegenerative disorders, and its loss of heterozygosity and low expression are seen in some cancers. Here, we show that low ANCO1 mRNA and protein expression levels are prognostic markers for poor clinical outcomes in breast cancer and that loss of nuclear ANCO1 protein expression predicts lower overall survival of patients with triple-negative breast cancer (TNBC). Knockdown of ANCO1 in early-stage TNBC cells led to aneuploidy, cellular senescence, and enhanced invasion in a 3D matrix. The presence of a subpopulation of ANCO1-depleted cells enabled invasion of the overall cell population in vitro and they converted more rapidly to invasive lesions in a xenograft mouse model. In ANCO1-depleted cells, ChIP-seq analysis showed a global increase in H3K27Ac signals that were enriched for AP-1, TEAD, STAT3, and NFκB motifs. ANCO1-regulated H3K27Ac peaks had a significantly higher overlap with known breast cancer enhancers compared to ANCO1-independent ones. H3K27Ac engagement was associated with transcriptional activation of genes in the PI3K-AKT, epithelial-mesenchymal transition (EMT), and senescence pathways. In conclusion, ANCO1 has hallmarks of a tumor suppressor whose loss of expression activates breast-cancer-specific enhancers and oncogenic pathways that can accelerate the early-stage progression of breast cancer.
Collapse
Affiliation(s)
- Meng Yuan
- Lombardi Comprehensive Cancer Center, Georgetown University, Washington, DC 20057, USA
| | - Megan E. Barefoot
- Lombardi Comprehensive Cancer Center, Georgetown University, Washington, DC 20057, USA
| | - Kendell Peterson
- Lombardi Comprehensive Cancer Center, Georgetown University, Washington, DC 20057, USA
| | - Moray J. Campbell
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Jan K. Blancato
- Lombardi Comprehensive Cancer Center, Georgetown University, Washington, DC 20057, USA
| | - Manjing Chen
- Department of Biostatistics, Bioinformatics and Biomathematics, Georgetown University, Washington, DC 20057, USA
| | - Marcel O. Schmidt
- Lombardi Comprehensive Cancer Center, Georgetown University, Washington, DC 20057, USA
| | - Amber J. Kiliti
- Lombardi Comprehensive Cancer Center, Georgetown University, Washington, DC 20057, USA
| | - Hong-Bin Fang
- Department of Biostatistics, Bioinformatics and Biomathematics, Georgetown University, Washington, DC 20057, USA
| | - Anton Wellstein
- Lombardi Comprehensive Cancer Center, Georgetown University, Washington, DC 20057, USA
| | - Anna T. Riegel
- Lombardi Comprehensive Cancer Center, Georgetown University, Washington, DC 20057, USA
| | - Ghada M. Sharif
- Lombardi Comprehensive Cancer Center, Georgetown University, Washington, DC 20057, USA
| |
Collapse
|
76
|
Liu D, Lu X, Huang W, Zhuang W. Long non-coding RNAs in non-small cell lung cancer: implications for EGFR-TKI resistance. Front Genet 2023; 14:1222059. [PMID: 37456663 PMCID: PMC10349551 DOI: 10.3389/fgene.2023.1222059] [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: 05/13/2023] [Accepted: 06/07/2023] [Indexed: 07/18/2023] Open
Abstract
Non-small cell lung cancer (NSCLC) is one of the most common types of malignant tumors as well as the leading cause of cancer-related deaths in the world. The application of epidermal growth factor receptor (EGFR)-tyrosine kinase inhibitors (TKIs) has dramatically improved the prognosis of NSCLC patients who harbor EGFR mutations. However, despite an excellent initial response, NSCLC inevitably becomes resistant to EGFR-TKIs, leading to irreversible disease progression. Hence, it is of great significance to shed light on the molecular mechanisms underlying the EGFR-TKI resistance in NSCLC. Long non-coding RNAs (lncRNAs) are critical gene modulators that are able to act as oncogenes or tumor suppressors that modulate tumorigenesis, invasion, and metastasis. Recently, extensive evidence demonstrates that lncRNAs also have a significant function in modulating EGFR-TKI resistance in NSCLC. In this review, we present a comprehensive summary of the lncRNAs involved in EGFR-TKI resistance in NSCLC and focus on their detailed mechanisms of action, including activation of alternative bypass signaling pathways, phenotypic transformation, intercellular communication in the tumor microenvironment, competing endogenous RNAs (ceRNAs) networks, and epigenetic modifications. In addition, we briefly discuss the limitations and the clinical implications of current lncRNAs research in this field.
Collapse
Affiliation(s)
- Detian Liu
- Department of Thoracic Surgery, Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Xiaolin Lu
- The Second Clinical Medical College of Nanchang University, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Wentao Huang
- Department of Thoracic Surgery, Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Wei Zhuang
- Department of Thoracic Surgery, Xiangya Hospital of Central South University, Changsha, Hunan, China
| |
Collapse
|
77
|
Mehdizadeh R, Ansari AM, Forouzesh F, Ghadirian R, Shahriari F, Shariatpanahi SP, Javidi MA. Cross-talk between non-ionizing electromagnetic fields and metastasis; EMT and hybrid E/M may explain the anticancer role of EMFs. PROGRESS IN BIOPHYSICS AND MOLECULAR BIOLOGY 2023:S0079-6107(23)00060-3. [PMID: 37302516 DOI: 10.1016/j.pbiomolbio.2023.06.003] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/17/2023] [Revised: 05/06/2023] [Accepted: 06/09/2023] [Indexed: 06/13/2023]
Abstract
Recent studies have shown that non-ionizing electromagnetic fields (NIEMFs) in a specific frequency, intensity, and exposure time can have anti-cancer effects on various cancer cells; however, the underlying precise mechanism of action is not transparent. Most cancer deaths are due to metastasis. This important phenomenon plays an inevitable role in different steps of cancer including progression and development. It has different stages including invasion, intravasation, migration, extravasation, and homing. Epithelial-mesenchymal transition (EMT), as well as hybrid E/M state, are biological processes, that involve both natural embryogenesis and tissue regeneration, and abnormal conditions including organ fibrosis or metastasis. In this context, some evidence reveals possible footprints of the important EMT-related pathways which may be affected in different EMFs treatments. In this article, critical EMT molecules and/or pathways which can be potentially affected by EMFs (e.g., VEGFR, ROS, P53, PI3K/AKT, MAPK, Cyclin B1, and NF-кB) are discussed to shed light on the mechanism of EMFs anti-cancer effect.
Collapse
Affiliation(s)
- Romina Mehdizadeh
- Department of Genetics, Faculty of Advanced Science, and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Alireza Madjid Ansari
- Department of Integrative Oncology, Breast Cancer Research Center, Motamed Cancer Institute, ACECR, Tehran, Iran
| | - Flora Forouzesh
- Department of Genetics, Faculty of Advanced Science, and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Reyhane Ghadirian
- Department of Integrative Oncology, Breast Cancer Research Center, Motamed Cancer Institute, ACECR, Tehran, Iran
| | - Fatemeh Shahriari
- Department of Molecular Genetics, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran
| | | | - Mohammad Amin Javidi
- Department of Integrative Oncology, Breast Cancer Research Center, Motamed Cancer Institute, ACECR, Tehran, Iran; Department of Genetics, Faculty of Advanced Science, and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran.
| |
Collapse
|
78
|
Zhong Y, Li X, Xie J, Zhang Y, Li H, Zheng D. Network pharmacology combined with molecular docking and experimental validation to reveal the pharmacological mechanism of naringin against renal fibrosis. Open Med (Wars) 2023; 18:20230736. [PMID: 37305520 PMCID: PMC10251165 DOI: 10.1515/med-2023-0736] [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: 03/14/2023] [Revised: 05/18/2023] [Accepted: 05/22/2023] [Indexed: 06/13/2023] Open
Abstract
To explore the pharmacological mechanism of naringin (NRG) in renal fibrosis (RF) based on network pharmacology combined with molecular docking and experimental validation. We used databases to screen for the targets of NRG and RF. The "drug-disease network" was established using Cytoscape. Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) analyses of targets were performed using Metascape, and molecular docking was performed using Schrödinger. We established an RF model in both mice and cells to validate the results of network pharmacology. After screening the database, we identified 222 common targets of NRG and RF and established a target network. Molecular docking showed that the target AKT had a good interaction with NRG. We found that the phosphatidylinositol 3-kinase (PI3K)/AKT signaling pathway was enriched by multiple targets and served as a target for experimental validation through GO and KEGG. The results revealed that NRG ameliorated renal dysfunction, reduced the release of inflammatory cytokines, decreased the expression of α-SMA, collagen I, and Fn, and recovered the expression of E-cad by inhibiting the PI3K/AKT signaling pathway. Our study used pharmacological analysis to predict the targets and mechanisms of NRG against RF. Furthermore, experiments proved that NRG inhibited RF effectively by targeting the PI3K/AKT signaling pathway.
Collapse
Affiliation(s)
- Yanan Zhong
- Department of Nephrology, The Affiliated Huai’an Hospital of Xuzhou Medical University and Huai’an Second People’s Hospital, Huai’an, China
| | - Xiang Li
- Department of Nephrology, The Affiliated Huai’an Hospital of Xuzhou Medical University and Huai’an Second People’s Hospital, Huai’an, China
| | - Juan Xie
- Department of Nephrology, The Affiliated Huai’an Hospital of Xuzhou Medical University and Huai’an Second People’s Hospital, Huai’an, China
| | - Yiyuan Zhang
- Department of Nephrology, The Affiliated Huai’an Hospital of Xuzhou Medical University and Huai’an Second People’s Hospital, Huai’an, China
| | - Hailun Li
- Department of Nephrology, The Affiliated Huai’an Hospital of Xuzhou Medical University and Huai’an Second People’s Hospital, Huai’an, China
| | - Donghui Zheng
- Department of Nephrology, The Affiliated Huai’an Hospital of Xuzhou Medical University and Huai’an Second People’s Hospital, Huai’an, China
| |
Collapse
|
79
|
Janta S, Pranweerapaiboon K, Vivithanaporn P, Plubrukarn A, Chairoungdua A, Prasertsuksri P, Apisawetakan S, Chaithirayanon K. Holothurin A Inhibits RUNX1-Enhanced EMT in Metastasis Prostate Cancer via the Akt/JNK and P38 MAPK Signaling Pathway. Mar Drugs 2023; 21:345. [PMID: 37367670 DOI: 10.3390/md21060345] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2023] [Revised: 06/01/2023] [Accepted: 06/02/2023] [Indexed: 06/28/2023] Open
Abstract
Due to the challenge of prostate cancer (PCa) management, there has been a surge in efforts to identify more safe and effective compounds that can modulate the epithelial-mesenchymal transition (EMT) for driving metastasis. Holothurin A (HA), a triterpenoid saponin isolated from Holothuria scabra, has now been characterized for its diverse biological activities. However, the mechanisms of HA in EMT-driven metastasis of human PCa cell lines has not yet been investigated. Moreover, runt-related transcription factor 1 (RUNX1) acts as an oncogene in prostate cancer, but little is known about its role in the EMT. Thus, the purpose of this study was to determine how RUNX1 influences EMT-mediated metastasis, as well as the potential effect of HA on EMT-mediated metastasis in endogenous and exogenous RUNX1 expressions of PCa cell lines. The results demonstrated that RUNX1 overexpression could promote the EMT phenotype with increased EMT markers, consequently driving metastatic migration and invasion in PC3 cell line through the activation of Akt/MAPK signaling pathways. Intriguingly, HA treatment could antagonize the EMT program in endogenous and exogenous RUNX1-expressing PCa cell lines. A decreasing metastasis of both HA-treated cell lines was evidenced through a downregulation of MMP2 and MMP9 via the Akt/P38/JNK-MAPK signaling pathway. Overall, our approach first demonstrated that RUNX1 enhanced EMT-driven prostate cancer metastasis and that HA was capable of inhibiting the EMT and metastatic processes and should probably be considered as a candidate for metastasis PCa treatment.
Collapse
Affiliation(s)
- Sirorat Janta
- Department of Anatomy, Faculty of Science, Mahidol University, Bangkok 10400, Thailand
| | - Kanta Pranweerapaiboon
- Department of Anatomy, Faculty of Science, Mahidol University, Bangkok 10400, Thailand
- Chulabhorn International College of Medicine, Thammasat University, Pathumthani 12120, Thailand
| | - Pornpun Vivithanaporn
- Chakri Naruebodindra Medical Institute, Faculty of Medicine, Ramathibodi Hospital, Mahidol University, Bangkok 10540, Thailand
| | - Anuchit Plubrukarn
- Department of Pharmacognosy and Pharmaceutical Botany, Faculty of Pharmaceutical Sciences, Prince of Songkla University, Songkhla 09112, Thailand
| | - Arthit Chairoungdua
- Department of Physiology, Faculty of Science, Mahidol University, Bangkok 10400, Thailand
| | | | - Somjai Apisawetakan
- Department of Anatomy, Faculty of Medicine, Srinakharinwirot University, Wattana, Bangkok 10110, Thailand
| | | |
Collapse
|
80
|
Wang Y, Zhang H, La Ferlita A, Sp N, Goryunova M, Sarchet P, Hu Z, Sorkin M, Kim A, Huang H, Zhu H, Tsung A, Pollock RE, Beane JD. Phosphorylation of IWS1 by AKT maintains liposarcoma tumor heterogeneity through preservation of cancer stem cell phenotypes and mesenchymal-epithelial plasticity. Oncogenesis 2023; 12:30. [PMID: 37237004 DOI: 10.1038/s41389-023-00469-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2022] [Revised: 04/07/2023] [Accepted: 04/17/2023] [Indexed: 05/28/2023] Open
Abstract
Chemotherapy remains the mainstay of treatment for patients with advanced liposarcoma (LPS), but response rates are only 25% and the overall survival at 5 years is dismal at 20-34%. Translation of other therapies have not been successful and there has been no significant improvement in prognosis for nearly 20 years. The aberrant activation of the phosphatidylinositol 3-kinase (PI3K)/AKT pathway has been implicated in the aggressive clinical behavior LPS and in resistance to chemotherapy, but the precise mechanism remains elusive and efforts to target AKT clinically have failed. Here we show that the AKT-mediated phosphorylation of the transcription elongation factor IWS1, promotes the maintenance of cancer stem cells in both cell and xenograft models of LPS. In addition, phosphorylation of IWS1 by AKT contributes to a "metastable" cell phenotype, characterized by mesenchymal/epithelial plasticity. The expression of phosphorylated IWS1 also promotes anchorage-dependent and independent growth, cell migration, invasion, and tumor metastasis. In patients with LPS, IWS1 expression is associated with reduced overall survival, increased frequency of recurrence, and shorter time to relapse after resection. These findings indicate that IWS1-mediated transcription elongation is an important regulator of human LPS pathobiology in an AKT-dependent manner and implicate IWS1 as an important molecular target to treat LPS.
Collapse
Affiliation(s)
- Yu Wang
- Department of Surgery, Division of Surgical Oncology, James Cancer Center, The Ohio State University, Columbus, OH, USA
- Institute of Pathology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
| | - Hongji Zhang
- Department of Surgery, Division of Surgical Oncology, James Cancer Center, The Ohio State University, Columbus, OH, USA
| | - Alessandro La Ferlita
- Department of Cancer Biology and Genetics, James Cancer Center, The Ohio State University, Columbus, OH, USA
| | - Nipin Sp
- Department of Surgery, Division of Surgical Oncology, James Cancer Center, The Ohio State University, Columbus, OH, USA
| | - Marina Goryunova
- Department of Surgery, Division of Surgical Oncology, James Cancer Center, The Ohio State University, Columbus, OH, USA
| | - Patricia Sarchet
- Department of Surgery, Division of Surgical Oncology, James Cancer Center, The Ohio State University, Columbus, OH, USA
| | - Zhiwei Hu
- Department of Surgery, Division of Surgical Oncology, James Cancer Center, The Ohio State University, Columbus, OH, USA
| | - Michael Sorkin
- Department of Plastic and Reconstructive Surgery, James Cancer Center, The Ohio State University, Columbus, OH, USA
| | - Alex Kim
- Department of Surgery, Division of Surgical Oncology, James Cancer Center, The Ohio State University, Columbus, OH, USA
| | - Hai Huang
- Department of Surgery, Division of Surgical Oncology, James Cancer Center, The Ohio State University, Columbus, OH, USA
| | - Hua Zhu
- Department of Surgery, Division of Cardiac Surgery, Wexner Medical Center, The Ohio State University, Columbus, OH, USA
| | - Allan Tsung
- Department of Surgery, Division of Surgical Oncology, James Cancer Center, The Ohio State University, Columbus, OH, USA
| | - Raphael E Pollock
- Department of Surgery, Division of Surgical Oncology, James Cancer Center, The Ohio State University, Columbus, OH, USA
| | - Joal D Beane
- Department of Surgery, Division of Surgical Oncology, James Cancer Center, The Ohio State University, Columbus, OH, USA.
| |
Collapse
|
81
|
Chu X, Lou J, Yi Y, Zhong L, Huang O. Knockdown of ARHGAP30 inhibits ovarian cancer cell proliferation, migration, and invasiveness by suppressing the PI3K/AKT/mTOR signaling pathway. Eur J Histochem 2023; 67. [PMID: 37170915 DOI: 10.4081/ejh.2023.3653] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2023] [Accepted: 04/22/2023] [Indexed: 05/13/2023] Open
Abstract
The mortality and morbidity rates of ovarian cancer (OC) are high, but the underlying mechanisms of OC have not been characterized. In this study, we determined the role of Rho GTPase Activating Protein 30 (ARHGAP30) in OC progression. We measured ARHGAP30 abundance in OC tissue samples and cells using immunohistochemistry (IHC) and RT-qPCR. EdU, transwell, and annexin V/PI apoptosis assays were used to evaluate proliferation, invasiveness, and apoptosis of OC cells, respectively. The results showed that ARHGAP30 was overexpressed in OC tissue samples and cells. Inhibition of ARHGAP30 suppressed growth and metastasis of OC cells, and enhanced apoptosis. Knockdown of ARHGAP30 in OC cells significantly inhibited the PI3K/AKT/mTOR pathway. Treatment with the PI3K/AKT/mTOR pathway inhibitor buparlisib simulated the effects of ARHGAP30 knockdown on growth, invasiveness, and apoptosis of OC cells. Following buparlisib treatment, the expression levels of p-PI3K, p-AKT, and p-mTOR were significantly decreased. Furthermore, buparlisib inhibited the effects of ARHGAP30 upregulation on OC cell growth and invasiveness. In conclusion, ARHGAP30 regulated the PI3K/AKT/mTOR pathway to promote progression of OC.
Collapse
Affiliation(s)
- Xiaoyan Chu
- College of Medicine, Nanchang University, Nanchang; Department of Gynecological Oncology, Jiangxi Cancer Hospital, Nanchang, Jiangxi.
| | - Jun Lou
- Department of Gynecological Oncology, Jiangxi Cancer Hospital, Nanchang, Jiangxi.
| | - Yun Yi
- Department of Gynecological Oncology, Jiangxi Cancer Hospital, Nanchang, Jiangxi.
| | - Linlin Zhong
- Department of Gynecological Oncology, Jiangxi Cancer Hospital, Nanchang, Jiangxi.
| | - Ouping Huang
- College of Medicine, Nanchang University, Nanchang; Key Laboratory of Women's Reproductive Health of Jiangxi Province, Jiangxi Provincial Maternal and Child Health Hospital, Nanchang, Jiangxi.
| |
Collapse
|
82
|
Jiang X, Yin H, Su W, Quan H, Yuan X, Feng X, Li P, He Y, Xiao J, Li R. Trifolirhizin inhibits proliferation, migration and invasion in nasopharyngeal carcinoma cells via PI3K/Akt signaling pathway suppression. Biochem Biophys Res Commun 2023; 667:111-119. [PMID: 37216826 DOI: 10.1016/j.bbrc.2023.05.030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2023] [Revised: 04/30/2023] [Accepted: 05/09/2023] [Indexed: 05/24/2023]
Abstract
Nasopharyngeal carcinoma (NPC) is a highly recurrent and metastatic malignant tumor affecting a large number of individuals in southern China. Traditional Chinese herbal medicine has been found to be a rich source of natural compounds with mild therapeutic effects and minimal side effects, making them increasingly popular for treating various diseases. Trifolirhizin, a natural flavonoid derived from leguminous plants, has gained significant attention for its therapeutic potential. In this study, we confirmed that trifolirhizin could effectively inhibit the proliferation, migration and invasion of nasopharyngeal carcinoma 6-10B and HK1 cells. Furthermore, our findings demonstrated that trifolirhizin achieves this by suppressing the PI3K/Akt signaling pathway. The findings of the present study provides a valuable perspective on the potential therapeutic applications of trifolirhizin for the treatment of nasopharyngeal carcinoma.
Collapse
Affiliation(s)
- Xing Jiang
- Department of Pharmacy, The Second Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
| | - Haihui Yin
- Department of Medical Ultrasound, The Second Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
| | - Wenqing Su
- Youjiang Medical University for Nationalities, Baise, Guangxi, 533000, China
| | - Haiyan Quan
- Hunan Polytechnic of Environment and Biology, Hengyang, Hunan, 421001, China
| | - Xinye Yuan
- Department of Pharmacy, The Second Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
| | - Xu Feng
- Department of Pharmacy, The Second Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
| | - Pei Li
- Department of Pharmacy, The Second Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
| | - Yan He
- Department of Pharmacy, The Second Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
| | - Junhui Xiao
- Department of Pharmacy, The Second Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China.
| | - Rong Li
- Department of Pharmacy, The Second Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China.
| |
Collapse
|
83
|
Neuendorf HM, Simmons JL, Boyle GM. Therapeutic targeting of anoikis resistance in cutaneous melanoma metastasis. Front Cell Dev Biol 2023; 11:1183328. [PMID: 37181747 PMCID: PMC10169659 DOI: 10.3389/fcell.2023.1183328] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2023] [Accepted: 04/14/2023] [Indexed: 05/16/2023] Open
Abstract
The acquisition of resistance to anoikis, the cell death induced by loss of adhesion to the extracellular matrix, is an absolute requirement for the survival of disseminating and circulating tumour cells (CTCs), and for the seeding of metastatic lesions. In melanoma, a range of intracellular signalling cascades have been identified as potential drivers of anoikis resistance, however a full understanding of the process is yet to be attained. Mechanisms of anoikis resistance pose an attractive target for the therapeutic treatment of disseminating and circulating melanoma cells. This review explores the range of small molecule, peptide and antibody inhibitors targeting molecules involved in anoikis resistance in melanoma, and may be repurposed to prevent metastatic melanoma prior to its initiation, potentially improving the prognosis for patients.
Collapse
Affiliation(s)
- Hannah M. Neuendorf
- Cancer Drug Mechanisms Group, QIMR Berghofer Medical Research Institute, Herston, QLD, Australia
- School of Biomedical Sciences, Faculty of Health, Queensland University of Technology, Brisbane, QLD, Australia
| | - Jacinta L. Simmons
- Cancer Drug Mechanisms Group, QIMR Berghofer Medical Research Institute, Herston, QLD, Australia
- School of Biomedical Sciences, Faculty of Health, Queensland University of Technology, Brisbane, QLD, Australia
- School of Biomedical Sciences, Faculty of Medicine, University of Queensland, Brisbane, QLD, Australia
| | - Glen M. Boyle
- Cancer Drug Mechanisms Group, QIMR Berghofer Medical Research Institute, Herston, QLD, Australia
- School of Biomedical Sciences, Faculty of Health, Queensland University of Technology, Brisbane, QLD, Australia
- School of Biomedical Sciences, Faculty of Medicine, University of Queensland, Brisbane, QLD, Australia
| |
Collapse
|
84
|
Monti N, Dinicola S, Querqui A, Fabrizi G, Fedeli V, Gesualdi L, Catizone A, Unfer V, Bizzarri M. Myo-Inositol Reverses TGF-β1-Induced EMT in MCF-10A Non-Tumorigenic Breast Cells. Cancers (Basel) 2023; 15:cancers15082317. [PMID: 37190245 DOI: 10.3390/cancers15082317] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2023] [Revised: 04/12/2023] [Accepted: 04/13/2023] [Indexed: 05/17/2023] Open
Abstract
Epithelial-Mesenchymal Transition (EMT), triggered by external and internal cues in several physiological and pathological conditions, elicits the transformation of epithelial cells into a mesenchymal-like phenotype. During EMT, epithelial cells lose cell-to-cell contact and acquire unusual motility/invasive capabilities. The associated architectural and functional changes destabilize the epithelial layer consistency, allowing cells to migrate and invade the surrounding tissues. EMT is a critical step in the progression of inflammation and cancer, often sustained by a main driving factor as the transforming growth factor-β1 (TGF-β1). Antagonizing EMT has recently gained momentum as an attractive issue in cancer treatment and metastasis prevention. Herein, we demonstrate the capability of myo-inositol (myo-Ins) to revert the EMT process induced by TGF-β1 on MCF-10A breast cells. Upon TGF-β1 addition, cells underwent a dramatic phenotypic transformation, as witnessed by structural (disappearance of the E-cadherin-β-catenin complexes and the emergence of a mesenchymal shape) and molecular modifications (increase in N-cadherin, Snai1, and vimentin), including the release of increased collagen and fibronectin. However, following myo-Ins, those changes were almost completely reverted. Inositol promotes the reconstitution of E-cadherin-β-catenin complexes, decreasing the expression of genes involved in EMT, while promoting the re-expression of epithelial genes (keratin-18 and E-cadherin). Noticeably, myo-Ins efficiently inhibits the invasiveness and migrating capability of TGF-β1 treated cells, also reducing the release of metalloproteinase (MMP-9) altogether with collagen synthesis, allowing for the re-establishment of appropriate cell-to-cell junctions, ultimately leading the cell layer back towards a more compact state. Inositol effects were nullified by previous treatment with an siRNA construct to inhibit CDH1 transcripts and, hence, E-cadherin synthesis. This finding suggests that the reconstitution of E-cadherin complexes is an irreplaceable step in the inositol-induced reversion of EMT. Overall, such a result advocates for the useful role of myo-Ins in cancer treatment.
Collapse
Affiliation(s)
- Noemi Monti
- Department of Experimental Medicine, Sapienza University of Rome, Viale Regina Elena 324, 00161 Rome, Italy
- Systems Biology Group Laboratory, Sapienza University, 00161 Rome, Italy
| | - Simona Dinicola
- Systems Biology Group Laboratory, Sapienza University, 00161 Rome, Italy
| | - Alessandro Querqui
- Department of Experimental Medicine, Sapienza University of Rome, Viale Regina Elena 324, 00161 Rome, Italy
- Systems Biology Group Laboratory, Sapienza University, 00161 Rome, Italy
| | - Gianmarco Fabrizi
- Systems Biology Group Laboratory, Sapienza University, 00161 Rome, Italy
| | - Valeria Fedeli
- Department of Experimental Medicine, Sapienza University of Rome, Viale Regina Elena 324, 00161 Rome, Italy
- Systems Biology Group Laboratory, Sapienza University, 00161 Rome, Italy
| | - Luisa Gesualdi
- Section of Histology and Embryology, Department of Anatomy, Histology, Forensic Medicine and Orthopedics, Sapienza University of Rome, Viale Regina Elena 336, 00161 Rome, Italy
| | - Angela Catizone
- Section of Histology and Embryology, Department of Anatomy, Histology, Forensic Medicine and Orthopedics, Sapienza University of Rome, Viale Regina Elena 336, 00161 Rome, Italy
| | - Vittorio Unfer
- Systems Biology Group Laboratory, Sapienza University, 00161 Rome, Italy
- The Experts Group on Inositol in Basic and Clinical Research (EGOI), 00161 Rome, Italy
- Gynecology Department, UniCamillus-Saint Camillus International University of Health and Medical Sciences, 00161 Rome, Italy
| | - Mariano Bizzarri
- Department of Experimental Medicine, Sapienza University of Rome, Viale Regina Elena 324, 00161 Rome, Italy
- Systems Biology Group Laboratory, Sapienza University, 00161 Rome, Italy
- The Experts Group on Inositol in Basic and Clinical Research (EGOI), 00161 Rome, Italy
| |
Collapse
|
85
|
Hohmann T, Hohmann U, Dehghani F. MACC1-induced migration in tumors: Current state and perspective. Front Oncol 2023; 13:1165676. [PMID: 37051546 PMCID: PMC10084939 DOI: 10.3389/fonc.2023.1165676] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Accepted: 03/14/2023] [Indexed: 03/29/2023] Open
Abstract
Malignant tumors are still a global, heavy health burden. Many tumor types cannot be treated curatively, underlining the need for new treatment targets. In recent years, metastasis associated in colon cancer 1 (MACC1) was identified as a promising biomarker and drug target, as it is promoting tumor migration, initiation, proliferation, and others in a multitude of solid cancers. Here, we will summarize the current knowledge about MACC1-induced tumor cell migration with a special focus on the cytoskeletal and adhesive systems. In addition, a brief overview of several in vitro models used for the analysis of cell migration is given. In this context, we will point to issues with the currently most prevalent models used to study MACC1-dependent migration. Lastly, open questions about MACC1-dependent effects on tumor cell migration will be addressed.
Collapse
|
86
|
Shi ZD, Pang K, Wu ZX, Dong Y, Hao L, Qin JX, Wang W, Chen ZS, Han CH. Tumor cell plasticity in targeted therapy-induced resistance: mechanisms and new strategies. Signal Transduct Target Ther 2023; 8:113. [PMID: 36906600 PMCID: PMC10008648 DOI: 10.1038/s41392-023-01383-x] [Citation(s) in RCA: 49] [Impact Index Per Article: 49.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Revised: 12/07/2022] [Accepted: 02/20/2023] [Indexed: 03/13/2023] Open
Abstract
Despite the success of targeted therapies in cancer treatment, therapy-induced resistance remains a major obstacle to a complete cure. Tumor cells evade treatments and relapse via phenotypic switching driven by intrinsic or induced cell plasticity. Several reversible mechanisms have been proposed to circumvent tumor cell plasticity, including epigenetic modifications, regulation of transcription factors, activation or suppression of key signaling pathways, as well as modification of the tumor environment. Epithelial-to-mesenchymal transition, tumor cell and cancer stem cell formation also serve as roads towards tumor cell plasticity. Corresponding treatment strategies have recently been developed that either target plasticity-related mechanisms or employ combination treatments. In this review, we delineate the formation of tumor cell plasticity and its manipulation of tumor evasion from targeted therapy. We discuss the non-genetic mechanisms of targeted drug-induced tumor cell plasticity in various types of tumors and provide insights into the contribution of tumor cell plasticity to acquired drug resistance. New therapeutic strategies such as inhibition or reversal of tumor cell plasticity are also presented. We also discuss the multitude of clinical trials that are ongoing worldwide with the intention of improving clinical outcomes. These advances provide a direction for developing novel therapeutic strategies and combination therapy regimens that target tumor cell plasticity.
Collapse
Affiliation(s)
- Zhen-Duo Shi
- Department of Urology, Xuzhou Clinical School of Xuzhou Medical University, Jiangsu, China.,Department of Urology, Xuzhou Central Hospital, Xuzhou, Jiangsu, China.,School of Life Sciences, Jiangsu Normal University, Jiangsu, China.,Department of Urology, Heilongjiang Provincial Hospital, Heilongjiang, China
| | - Kun Pang
- Department of Urology, Xuzhou Clinical School of Xuzhou Medical University, Jiangsu, China.,Department of Urology, Xuzhou Central Hospital, Xuzhou, Jiangsu, China
| | - Zhuo-Xun Wu
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, Queens, NY, 11439, USA
| | - Yang Dong
- Department of Urology, Xuzhou Clinical School of Xuzhou Medical University, Jiangsu, China.,Department of Urology, Xuzhou Central Hospital, Xuzhou, Jiangsu, China
| | - Lin Hao
- Department of Urology, Xuzhou Clinical School of Xuzhou Medical University, Jiangsu, China.,Department of Urology, Xuzhou Central Hospital, Xuzhou, Jiangsu, China
| | - Jia-Xin Qin
- Department of Urology, Xuzhou Clinical School of Xuzhou Medical University, Jiangsu, China.,Department of Urology, Xuzhou Central Hospital, Xuzhou, Jiangsu, China
| | - Wei Wang
- Department of Medical College, Southeast University, Nanjing, China
| | - Zhe-Sheng Chen
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, Queens, NY, 11439, USA.
| | - Cong-Hui Han
- Department of Urology, Xuzhou Clinical School of Xuzhou Medical University, Jiangsu, China. .,Department of Urology, Xuzhou Central Hospital, Xuzhou, Jiangsu, China. .,School of Life Sciences, Jiangsu Normal University, Jiangsu, China. .,Department of Urology, Heilongjiang Provincial Hospital, Heilongjiang, China.
| |
Collapse
|
87
|
Chen J, Liu F, Wu J, Yang Y, He J, Wu F, Yang K, Li J, Jiang Z, Jiang Z. Effect of STK3 on proliferation and apoptosis of pancreatic cancer cells via PI3K/AKT/mTOR pathway. Cell Signal 2023; 106:110642. [PMID: 36871796 DOI: 10.1016/j.cellsig.2023.110642] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2022] [Revised: 02/12/2023] [Accepted: 02/28/2023] [Indexed: 03/07/2023]
Abstract
Pancreatic cancer, as a malignant tumor with a very poor prognosis, has a high mortality. It is imperative to clarify the mechanism of pancreatic cancer development and find suitable targets for diagnosis and treatment. Serine/threonine kinase 3 (STK3) is one of the core kinases of the Hippo pathway and has the ability to inhibit tumor growth. But the biological function of STK3 in pancreatic cancer remains unknown. Here, we confirmed that STK3 has an impact on the growth, apoptosis, and metastasis of pancreatic cancer cells and investigated the related molecular mechanisms. In our research, we found that STK3 is reduced in pancreatic cancer by RT-qPCR, IHC and IF, its expression level is correlated with the clinicopathological features. CCK-8 assay, colony formation assay and flow cytometry were used to detect the effect of STK3 on the proliferation and apoptosis of pancreatic cancer cells. In addition, the Transwell assay was used to detect the ability of cell migration and invasion. The results showed that STK3 promoted apoptosis and inhibited cell migration, invasion and proliferation in pancreatic cancer. Gene set enrichment analysis (GSEA) and western blotting are used to predict and verify the pathways related to STK3. Subsequently, we found that the effect of STK3 on proliferation and apoptosis is closely related to the PI3K/AKT/mTOR pathway. Moreover, the assistance of RASSF1 plays a significant role in the regulation of PI3K/AKT/mTOR pathway by STK3. The nude mouse xenograft experiment demonstrated the tumor suppressive ability of STK3 in vivo. Collectively, this study found that STK3 regulates pancreatic cancer cell proliferation and apoptosis by suppressing the PI3K/AKT/mTOR pathway with the assistance of RASSF1.
Collapse
Affiliation(s)
- Jun Chen
- Department of Gastroenterology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Fuqiang Liu
- Department of Gastroenterology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Jiao Wu
- Department of Gastroenterology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Yichun Yang
- Department of Gastroenterology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Jin He
- Department of Gastroenterology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Fan Wu
- Department of Gastroenterology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Kun Yang
- Department of Gastroenterology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Junfeng Li
- Department of Gastroenterology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Zhongxiang Jiang
- Department of Gastroenterology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Zheng Jiang
- Department of Gastroenterology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China.
| |
Collapse
|
88
|
Jiang S, Wan F, Lian H, Lu Z, Li X, Cao D, Jiang Y, Li J. Friend or foe? The dual role of triptolide in the liver, kidney, and heart. Biomed Pharmacother 2023; 161:114470. [PMID: 36868013 DOI: 10.1016/j.biopha.2023.114470] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2023] [Revised: 02/23/2023] [Accepted: 02/28/2023] [Indexed: 03/05/2023] Open
Abstract
Triptolide, a controversial natural compound due to its significant pharmacological activities and multiorgan toxicity, has gained much attention since it was isolated from the traditional Chinese herb Tripterygium wilfordii Hook F. However, in addition to its severe toxicity, triptolide also presents powerful therapeutic potency in the same organs, such as the liver, kidney, and heart, which corresponds to the Chinese medicine theory of You Gu Wu Yun (anti-fire with fire) and deeply interested us. To determine the possible mechanisms involved in the dual role of triptolide, we reviewed related articles about the application of triptolide in both physiological and pathological conditions. Inflammation and oxidative stress are the two main ways triptolide exerts different roles, and the cross-talk between NF-κB and Nrf2 may be one of the mechanisms responsible for the dual role of triptolide and may represent the scientific connotation of You Gu Wu Yun. For the first time, we present a review of the dual role of triptolide in the same organ and propose the possible scientific connotation of the Chinese medicine theory of You Gu Wu Yun, hoping to promote the safe and efficient use of triptolide and other controversial medicines.
Collapse
Affiliation(s)
- Shiyuan Jiang
- Department of Histology and Embryology, School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Feng Wan
- Department of Anatomy, School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Hui Lian
- Department of Histology and Embryology, School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Zhihao Lu
- Department of Histology and Embryology, School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Xueming Li
- Department of Histology and Embryology, School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Dan Cao
- Department of Histology and Embryology, School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Yangyu Jiang
- Department of Histology and Embryology, School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Jian Li
- Department of Histology and Embryology, School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing 102488, China.
| |
Collapse
|
89
|
Lin CH, Lin WD, Huang YC, Chen YC, Loh ZJ, Ger LP, Lin FC, Li HY, Cheng HC, Lee KH, Hsiao M, Lu PJ. Carboxyl-terminal modulator protein facilitates tumor metastasis in triple-negative breast cancer. Cancer Gene Ther 2023; 30:404-413. [PMID: 36400965 PMCID: PMC10014580 DOI: 10.1038/s41417-022-00559-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Revised: 10/18/2022] [Accepted: 10/28/2022] [Indexed: 11/19/2022]
Abstract
Currently, the survival rate for breast cancer is more than 90%, but once the cancer cells metastasize to distal organs, the survival rate is dramatically reduced, to less than 30%. Triple-negative breast cancer accounts for 15-20% of all breast cancers. Triple-negative breast cancer (TNBC) is associated with poor prognostic and diagnostic outcomes due to the limiting therapeutic strategies, relative to non-TNBC breast cancers. Therefore, the development of targeted therapy for TNBC metastasis remains an urgent issue. In this study, high Carboxyl-terminal modulator protein (CTMP) is significantly associated with recurrence and disease-free survival rate in TNBC patients. Overexpression of CTMP promotes migration and invasion abilities in BT549 cells. Down-regulating of CTMP expression inhibits migration and invasion abilities in MDA-MB-231 cells. In vivo inoculation of high-CTMP cells enhances distant metastasis in mice. The metastasis incidence rate is decreased in mice injected with CTMP-downregulating MDA-MB-231 cells. Gene expression microarray analysis indicates the Akt-dependent pathway is significantly enhanced in CTMP overexpressing cells compared to the parental cells. Blocking Akt activation via Akt inhibitor treatment or co-expression of the dominant-negative form of Akt proteins successfully abolishes the CTMP mediating invasion in TNBC cells. Our findings suggest that CTMP is a potential diagnostic marker for recurrence and poor disease-free survival in TNBC patients. CTMP promotes TNBC metastasis via the Akt-activation-dependent pathway.
Collapse
Affiliation(s)
- Cheng-Han Lin
- Institute of Clinical Medicine, College of Medicine, National Cheng Kung University, Tainan, 70401, Taiwan
| | - Wen-Der Lin
- Institute of Clinical Medicine, College of Medicine, National Cheng Kung University, Tainan, 70401, Taiwan
| | - Yun-Chin Huang
- Institute of Clinical Medicine, College of Medicine, National Cheng Kung University, Tainan, 70401, Taiwan
| | - Yu-Chia Chen
- Division of General Surgery, Department of Surgery, Kaohsiung Veterans General Hospital, Kaohsiung, 813414, Taiwan
| | - Zhu-Jun Loh
- Department of Surgery, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, 70401, Taiwan
| | - Luo-Ping Ger
- Department of Medical Education and Research, Kaohsiung Veterans General Hospital, Kaohsiung, 813414, Taiwan
| | - Forn-Chia Lin
- Department of Radiation Oncology, National Cheng Kung University Hospital, Tainan, 70401, Taiwan
| | - Hao-Yi Li
- Institute of Clinical Medicine, College of Medicine, National Cheng Kung University, Tainan, 70401, Taiwan
| | - Hui-Chuan Cheng
- Institute of Clinical Medicine, College of Medicine, National Cheng Kung University, Tainan, 70401, Taiwan
| | - Kuen-Haur Lee
- Graduate Institute of Cancer Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei, 11031, Taiwan
| | - Michael Hsiao
- Genomics Research Center, Academia Sinica, Taipei, 115, Taiwan
| | - Pei-Jung Lu
- Institute of Clinical Medicine, College of Medicine, National Cheng Kung University, Tainan, 70401, Taiwan. .,Department of Clinical Medicine Research, National Cheng Kung University Hospital, Tainan, 70401, Taiwan.
| |
Collapse
|
90
|
Hu Q, Chen Y, Deng X, Li Y, Ma X, Zeng J, Zhao Y. Diabetic nephropathy: Focusing on pathological signals, clinical treatment, and dietary regulation. Biomed Pharmacother 2023; 159:114252. [PMID: 36641921 DOI: 10.1016/j.biopha.2023.114252] [Citation(s) in RCA: 42] [Impact Index Per Article: 42.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2022] [Revised: 01/04/2023] [Accepted: 01/12/2023] [Indexed: 01/15/2023] Open
Abstract
Diabetic nephropathy (DN) is one of the most severe complications of diabetes. However, due to its complex pathological mechanisms, no effective therapeutic methods (other than ACEIs and ARBs) have been applied, which have been used for many years in clinical practice. Recent studies have shown that emerging therapeutics, including novel target-based pharmacotherapy, cell therapies, and dietary regulation, are leading to new hopes for DN management. This review aims to shed new light on the treatment of DN by describing the important pathological mechanisms of DN and by analysing recent advances in clinical treatment, including drug therapy, cell therapy, and dietary regulation. In pathological mechanisms, RAAS activation, AGE accumulation, and EMT are involved in inflammation, cellular stress, apoptosis, pyroptosis, and autophagy. In pharmacotherapy, several new therapeutics, including SGLT2 inhibitors, GLP-1 agonists, and MRAs, are receiving public attention. In addition, stem cell therapies and dietary regulation are also being emphasized. Herein, we highlight the importance of combining therapy and dietary regulation in the treatment of DN and anticipate more basic research or clinical trials to verify novel strategies.
Collapse
Affiliation(s)
- Qichao Hu
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China; Department of Pharmacy, the Fifth Medical Center of PLA General Hospital, Beijing 100039, China.
| | - Yuan Chen
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China.
| | - Xinyu Deng
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China.
| | - Yubing Li
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China.
| | - Xiao Ma
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China.
| | - Jinhao Zeng
- TCM Regulating Metabolic Diseases Key Laboratory of Sichuan Province, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China.
| | - Yanling Zhao
- Department of Pharmacy, the Fifth Medical Center of PLA General Hospital, Beijing 100039, China.
| |
Collapse
|
91
|
The Transcription Factor Twist1 Has a Significant Role in Mycosis Fungoides (MF) Cell Biology: An RNA Sequencing Study of 40 MF Cases. Cancers (Basel) 2023; 15:cancers15051527. [PMID: 36900319 PMCID: PMC10000433 DOI: 10.3390/cancers15051527] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2023] [Revised: 02/20/2023] [Accepted: 02/24/2023] [Indexed: 03/05/2023] Open
Abstract
The purpose of this RNA sequencing study was to investigate the biological mechanism underlying how the transcription factors (TFs) Twist1 and Zeb1 influence the prognosis of mycosis fungoides (MF). We used laser-captured microdissection to dissect malignant T-cells obtained from 40 skin biopsies from 40 MF patients with stage I-IV disease. Immunohistochemistry (IHC) was used to determinate the protein expression levels of Twist1 and Zeb1. Based on RNA sequencing, principal component analysis (PCA), differential expression (DE) analysis, ingenuity pathway analysis (IPA), and hub gene analysis were performed between the high and low Twist1 IHC expression cases. The DNA from 28 samples was used to analyze the TWIST1 promoter methylation level. In the PCA, Twist1 IHC expression seemed to classify cases into different groups. The DE analysis yielded 321 significant genes. In the IPA, 228 significant upstream regulators and 177 significant master regulators/causal networks were identified. In the hub gene analysis, 28 hub genes were found. The methylation level of TWIST1 promoter regions did not correlate with Twist1 protein expression. Zeb1 protein expression did not show any major correlation with global RNA expression in the PCA. Many of the observed genes and pathways associated with high Twist1 expression are known to be involved in immunoregulation, lymphocyte differentiation, and aggressive tumor biology. In conclusion, Twist1 might be an important regulator in the disease progression of MF.
Collapse
|
92
|
Ghaemi Z, Mowla SJ, Soltani BM. Novel splice variants of LINC00963 suppress colorectal cancer cell proliferation via miR-10a/miR-143/miR-217/miR-512-mediated regulation of PI3K/AKT and Wnt/β-catenin signaling pathways. BIOCHIMICA ET BIOPHYSICA ACTA. GENE REGULATORY MECHANISMS 2023; 1866:194921. [PMID: 36804476 DOI: 10.1016/j.bbagrm.2023.194921] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/21/2022] [Revised: 01/28/2023] [Accepted: 02/10/2023] [Indexed: 02/18/2023]
Abstract
Emerging evidence has shown lncRNAs play important roles in signaling pathways involved in colorectal cancer (CRC) carcinogenesis. However, only a few functional lncRNAs have been extensively researched, especially in CRC-related signaling pathways. Looking for novel candidate regulators of CRC incidence and progression, using available RNA-seq and microarray datasets, LINC00963 was introduced as a bona fide oncogenic-lncRNA. Consistently, RT-qPCR results showed that LINC00963 was up-regulated in CRC tissues. However, our attempt to amplify the full-length lncRNA from cDNA resulted in the discovery of two novel variants (LINC00963-v2 & LINC00963-v3) that surprisingly, were downregulated in CRC tissues, detected by RT-qPCR. Overexpression of LINC00963-v2/-v3 in HCT116 and SW480 cells resulted in downregulation of the major oncogenes and upregulation of the main tumor suppressor genes involved in PI3K and Wnt signaling, verified through RT-qPCR, western blotting, and TOPFlash assays. Mechanistic studies revealed that LINC00963-v2/-v3 exert their effect on PI3K and Wnt signaling through sponging miR-10a-5p, miR-143-3p, miR-217, and miR-512-3p, which in turn these miRNAs are fine-regulators of PTEN, APC1, and Axin1 tumor suppressor genes verified by dual-luciferase assay and RT-qPCR. At cellular levels, LINC00963-v2/-v3 overexpression suppressed cell proliferation, viability, and migration while increasing the apoptosis of CRC cell lines, detected by PI flow cytometry, colony formation, MTT, RT-qPCR, wound-healing, Transwell, AnnexinV-PE/7AAD, caspase3/7 activity assays, and Hoechst/PI-AO/EB staining. Overall, our results indicate that LINC00963-v2 & -v3 are novel tumor suppressor ceRNAs that attenuate the PI3K and Wnt pathways during CRC incidence and these lncRNAs may serve as potential targets for CRC therapy.
Collapse
Affiliation(s)
- Zahra Ghaemi
- Department of Genetics, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran.
| | - Seyed Javad Mowla
- Department of Genetics, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran.
| | - Bahram Mohammad Soltani
- Department of Genetics, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran.
| |
Collapse
|
93
|
Paccosi E, Balzerano A, Proietti-De-Santis L. Interfering with the Ubiquitin-Mediated Regulation of Akt as a Strategy for Cancer Treatment. Int J Mol Sci 2023; 24:ijms24032809. [PMID: 36769122 PMCID: PMC9917864 DOI: 10.3390/ijms24032809] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2022] [Revised: 01/26/2023] [Accepted: 01/30/2023] [Indexed: 02/05/2023] Open
Abstract
The serine/threonine kinase Akt modulates the functions of numerous substrates, many of them being involved in cell proliferation and growth, metabolism, angiogenesis, resistance to hypoxia and migration. Akt is frequently deregulated in many types of human cancers, its overexpression or abnormal activation being associated with the increased proliferation and survival of cancer cells. A promising avenue for turning off the functionality of Akt is to either interfere with the K63-linked ubiquitination that is necessary for Akt membrane recruitment and activation or increase the K48-linked polyubiquitination that aims to target Akt to the proteasome for its degradation. Recent evidence indicates that targeting the ubiquitin proteasome system is effective for certain cancer treatments. In this review, the functions and roles of Akt in human cancer will be discussed, with a main focus on molecules and compounds that target various elements of the ubiquitination processes that regulate the activation and inactivation of Akt. Moreover, their possible and attractive implications for cancer therapy will be discussed.
Collapse
|
94
|
Velez BC, Petrella CP, DiSalvo KH, Cheng K, Kravtsov R, Krasniqi D, Krucher NA. Combined inhibition of ACLY and CDK4/6 reduces cancer cell growth and invasion. Oncol Rep 2023; 49:32. [PMID: 36562384 PMCID: PMC9827262 DOI: 10.3892/or.2022.8469] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2022] [Accepted: 11/23/2022] [Indexed: 12/24/2022] Open
Abstract
The use of small molecule kinase inhibitors, which target specific enzymes that are overactive in cancer cells, has revolutionized cancer patient treatment. To treat some types of breast cancer, CDK4/6 inhibitors, such as palbociclib, have been developed that target the phosphorylation of the retinoblastoma tumor suppressor gene. Acquired resistance to CDK4/6 inhibitors may be due to activation of the AKT pro‑survival signaling pathway that stimulates several processes, such as growth, metastasis and changes in metabolism that support rapid cell proliferation. The aim of the present study was to investigate whether targeting ATP citrate lyase (ACLY), a downstream target of AKT, may combine with CDK4/6 inhibition to inhibit tumorigenesis. The present study determined that ACLY is activated in breast and pancreatic cancer cells in response to palbociclib treatment and AKT mediates this effect. Inhibition of ACLY using bempedoic acid used in combination with palbociclib reduced cell viability in a panel of breast and pancreatic cancer cell lines. This effect was also observed using breast cancer cells grown in 3D cell culture. Mechanistically, palbociclib inhibited cell proliferation, whereas bempedoic acid stimulated apoptosis. Finally, using Transwell invasion assays and immunoblotting, the present study demonstrated that ACLY inhibition blocked cell invasion, when used alone or in combination with palbociclib. These data may yield useful information that could guide the development of future therapies aimed at the reduction of acquired resistance observed clinically.
Collapse
Affiliation(s)
| | | | | | - Keyi Cheng
- Department of Biology, Pace University, Pleasantville, NY 10570, USA
| | - Rebecca Kravtsov
- Department of Biology, Pace University, Pleasantville, NY 10570, USA
| | - Dorina Krasniqi
- Department of Biology, Pace University, Pleasantville, NY 10570, USA
| | - Nancy Ann Krucher
- Department of Biology, Pace University, Pleasantville, NY 10570, USA
| |
Collapse
|
95
|
Xu J, Wang Y, Jiang J, Yin C, Shi B. ADAM12 promotes clear cell renal cell carcinoma progression and triggers EMT via EGFR/ERK signaling pathway. J Transl Med 2023; 21:56. [PMID: 36717944 PMCID: PMC9885678 DOI: 10.1186/s12967-023-03913-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Accepted: 01/22/2023] [Indexed: 01/31/2023] Open
Abstract
BACKGROUND Clear cell renal cell carcinoma (ccRCC) is a major worldwide health problem due to its high prevalence and mortality rate. A disintegrin and metalloproteinase 12 (ADAM12) is aberrantly expressed in various cancers and plays an important role in tumor progression. However, its explicit effect and molecular mechanism in ccRCC remain unclear. METHODS We investigated the dysregulation of ADAM12 in ccRCC through public databases and bioinformatics analyses. The expression of ADAM12 was further verified in ccRCC tissues by RT-qPCR and immunohistochemistry (IHC). The relationship between ADAM12 expression and clinicopathological characteristics was analyzed statistically. The effects of ADAM12 on the proliferation, migration and invasion of ccRCC cells were examined by in vitro and in vivo experiments. RESULTS ADAM12 was significantly upregulated in ccRCC tissues and associated with poor prognosis in ccRCC patients. ADAM12 promoted ccRCC cell proliferation, migration and invasion in vitro and the growth of subcutaneous tumors in vivo. Knockdown of ADAM12 successfully suppressed its oncogenic function. Mechanistically, its overexpression induced epithelial-mesenchymal transition (EMT) by downregulating E-cadherin and upregulating N-cadherin and Snail. Moreover, ADAM12 participated in the epidermal growth factor receptor (EGFR) pathway and activated the downstream signal ERK1/2 by shedding the EGFR ligand, thereby upregulating target genes including c-Myc, enhancing cell survival and invasion ability, and promoting tumor progression, metastasis and the induction of EMT. CONCLUSIONS High expression of ADAM12 induced EMT and promoted cell proliferation, migration, and invasion by activating the EGFR/ERK signaling pathway in ccRCC.
Collapse
Affiliation(s)
- Jinming Xu
- Department of Urology, Shenzhen Second People's Hospital/First Affiliated Hospital of Shenzhen University, Shenzhen, 518035, Guangdong, China
- Shantou University Medical College, Shantou, 515041, Guangdong, China
| | - Yan Wang
- Department of Urology, Peking University Shenzhen Hospital, Shenzhen, 518036, Guangdong, China
| | - Jiahao Jiang
- Department of Urology, Shenzhen Second People's Hospital/First Affiliated Hospital of Shenzhen University, Shenzhen, 518035, Guangdong, China
| | - Cong Yin
- Department of Urology, Shenzhen Second People's Hospital/First Affiliated Hospital of Shenzhen University, Shenzhen, 518035, Guangdong, China
| | - Bentao Shi
- Department of Urology, Shenzhen Second People's Hospital/First Affiliated Hospital of Shenzhen University, Shenzhen, 518035, Guangdong, China.
| |
Collapse
|
96
|
丁 虹, 李 肖, 周 璐, 崔 智, 蒙 海, 王 娟. [Silenced ANP32A inhibits the growth, invasion and migration of colorectal cancer in vitro via the inactivation of AKT pathway]. NAN FANG YI KE DA XUE XUE BAO = JOURNAL OF SOUTHERN MEDICAL UNIVERSITY 2023; 43:52-59. [PMID: 36856210 PMCID: PMC9978733 DOI: 10.12122/j.issn.1673-4254.2023.01.07] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 05/23/2022] [Indexed: 03/02/2023]
Abstract
OBJECTIVE To investigate the effect of ANP32A silencing on invasion and migration of colon cancer cells and the influence of the activity of AKT signaling pathway on this effect. METHODS Colorectal cancer HCT116 and SW480 were transfected with a small interfering RNA targeting ANP32A via a lentiviral vector. At 24, 48 and 72 h after the transfection, the changes in cell proliferation and AKT activity in the cells were detected using MTT assay and Western blotting, respectively. HCT116 and SW480 cells were treated with the AKT agonist SC79 or its inhibitor MK2206 for 24, 48, 72 and 96 h, and the changes in cell migration and invasion ability were analyzed using Transwell chamber assay and cell proliferation was assessed using MTT assay. The effects of SC79 and MK2206 on migration and invasion abilities of HCT116 and SW480 cells with or without ANP32A silencing were examined using wound healing and Transwell chamber assays, and the changes in the expression of metadherin (MTDH), a factor associated with cells invasion and migration, was detected with Western blotting. RESULTS Lentivirus-mediated ANP32A silencing significantly down-regulated the activity of AKT and inhibited the proliferation of both HCT116 and SW480 cells (P < 0.01). The application of AKT inhibitor MK2206 obviously inhibited the proliferation, invasion and migration of the colorectal cancer cells (P < 0.05), while the AKT agonist SC79 significantly promoted the invasion and migration of the cells (P < 0.01). In HCT116 and SW480 cells with ANP32A silencing, treatment with MK2206 strongly enhanced the inhibitory effects of ANP32A silencing on cell invasion and migration (P < 0.05) and the expression of MTDH, while SC79 partially reversed these inhibitory effects (P < 0.01). CONCLUSION ANP32A silencing inhibits invasion and migration of colorectal cancer cells possibly by inhibiting the activation of the AKT signaling pathway.
Collapse
Affiliation(s)
- 虹芳 丁
- 桂林医学院基础医学院,广西 桂林 541199Faculty of Basic Medical Sciences, of Guilin Medical College, Guilin 541199, China
| | - 肖娟 李
- 桂林医学院药学院,广西 桂林 541199School of Pharmacy, of Guilin Medical College, Guilin 541199, China
| | - 璐炜 周
- 桂林医学院药学院,广西 桂林 541199School of Pharmacy, of Guilin Medical College, Guilin 541199, China
| | - 智 崔
- 桂林医学院基础医学院,广西 桂林 541199Faculty of Basic Medical Sciences, of Guilin Medical College, Guilin 541199, China
| | - 海德 蒙
- 桂林医学院生物技术学院,广西 桂林 541199School of Intelligent Medicine and Biotechnology, of Guilin Medical College, Guilin 541199, China
| | - 娟 王
- 桂林医学院基础医学院,广西 桂林 541199Faculty of Basic Medical Sciences, of Guilin Medical College, Guilin 541199, China
| |
Collapse
|
97
|
Namiki T, Terakawa J, Karakama H, Noguchi M, Murakami H, Hasegawa Y, Ohara O, Daikoku T, Ito J, Kashiwazaki N. Uterine epithelial Gp130 orchestrates hormone response and epithelial remodeling for successful embryo attachment in mice. Sci Rep 2023; 13:854. [PMID: 36646738 PMCID: PMC9842754 DOI: 10.1038/s41598-023-27859-y] [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: 08/03/2022] [Accepted: 01/09/2023] [Indexed: 01/18/2023] Open
Abstract
Leukemia inhibitory factor (LIF) receptor, an interleukin 6 cytokine family signal transducer (Il6st, also known as Gp130) that is expressed in the uterine epithelium and stroma, has been recognized to play an essential role in embryo implantation. However, the molecular mechanism underlying Gp130-mediated LIF signaling in the uterine epithelium during embryo implantation has not been elucidated. In this study, we generated mice with uterine epithelium specific deletion of Gp130 (Gp130 ecKO). Gp130 ecKO females were infertile due to the failure of embryo attachment and decidualization. Histomorphological observation revealed that the endometrial shape and embryo position from Gp130 ecKO were comparable to those of the control, and uterine epithelial cell proliferation, whose attenuation is essential for embryo implantation, was controlled in Gp130 ecKO. Comprehensive gene expression analysis using RNA-seq indicates that epithelial Gp130 regulates the expression of estrogen- and progesterone-responsive genes in conjunction with immune response during embryo implantation. We also found that an epithelial remodeling factor, snail family transcriptional repressor 1 (Snai1), was markedly reduced in the pre-implantation uterus from Gp130 ecKO. These results suggest that not only the suppression of uterine epithelial cell proliferation, but also Gp130-mediated epithelial remodeling is required for successful implantation in mice.
Collapse
Affiliation(s)
- Takafumi Namiki
- Laboratory of Animal Reproduction, School of Veterinary Medicine, Azabu University, 1-17-71 Fuchinobe, Chuo-Ku, Sagamihara, Kanagawa, 252-5201, Japan.,Graduate School of Veterinary Science, Azabu University, Sagamihara, Japan.,Department of Life Science Frontiers, Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto, Japan
| | - Jumpei Terakawa
- Graduate School of Veterinary Science, Azabu University, Sagamihara, Japan. .,Laboratory of Toxicology, School of Veterinary Medicine, Azabu University, 1-17-71 Fuchinobe, Chuo-Ku, Sagamihara, Kanagawa, 252-5201, Japan.
| | - Harumi Karakama
- Laboratory of Animal Reproduction, School of Veterinary Medicine, Azabu University, 1-17-71 Fuchinobe, Chuo-Ku, Sagamihara, Kanagawa, 252-5201, Japan
| | - Michiko Noguchi
- Graduate School of Veterinary Science, Azabu University, Sagamihara, Japan.,Laboratory of Theriogenology, School of Veterinary Medicine, Azabu University, Sagamihara, Japan
| | - Hironobu Murakami
- Graduate School of Veterinary Science, Azabu University, Sagamihara, Japan.,Laboratory of Infectious Diseases, School of Veterinary Medicine, Azabu University, Sagamihara, Japan
| | - Yoshinori Hasegawa
- Department of Applied Genomics, Kazusa DNA Research Institute, Kisarazu, Japan
| | - Osamu Ohara
- Department of Applied Genomics, Kazusa DNA Research Institute, Kisarazu, Japan
| | - Takiko Daikoku
- Research Center for Experimental Modeling of Human Disease, Institute for Experimental Animals, Kanazawa University, Kanazawa, Japan
| | - Junya Ito
- Laboratory of Animal Reproduction, School of Veterinary Medicine, Azabu University, 1-17-71 Fuchinobe, Chuo-Ku, Sagamihara, Kanagawa, 252-5201, Japan. .,Graduate School of Veterinary Science, Azabu University, Sagamihara, Japan. .,Center for Human and Animal Symbiosis Science, Azabu University, Sagamihara, Japan.
| | - Naomi Kashiwazaki
- Laboratory of Animal Reproduction, School of Veterinary Medicine, Azabu University, 1-17-71 Fuchinobe, Chuo-Ku, Sagamihara, Kanagawa, 252-5201, Japan.,Graduate School of Veterinary Science, Azabu University, Sagamihara, Japan
| |
Collapse
|
98
|
Davenport BN, Jones HN, Wilson RL. Placental treatment with insulin-like growth factor 1 via nanoparticle differentially impacts vascular remodeling factors in guinea pig sub-placenta/decidua. Front Physiol 2023; 13:1055234. [PMID: 36685211 PMCID: PMC9845775 DOI: 10.3389/fphys.2022.1055234] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Accepted: 12/13/2022] [Indexed: 01/06/2023] Open
Abstract
Clinically, fetal growth restriction (FGR) is only detectable in later gestation, despite pathophysiological establishment likely earlier in pregnancy. Additionally, there are no effective in utero treatment options for FGR. We have developed a nanoparticle to deliver human insulin-like 1 growth factor (hIGF-1) in a trophoblast-specific manner which results in increased expression of hIGF-1. IGF-1 signaling in the placenta regulates multiple developmental processes including trophoblast invasion and maternal vascular remodeling, both of which can be diminished in the FGR placenta. We aimed to determine the effects of short-term hIGF-1 nanoparticle treatment on sub-placenta/decidua trophoblast signaling mechanisms in FGR and under normal growth conditions. Using the guinea pig maternal nutrient restriction (MNR) model of FGR, ultrasound-guided, intra-placenta injections of hIGF-1 nanoparticle were performed at gestational day 30-33, and dams sacrificed 5 days later. Sub-placenta/decidua tissue was separated from placenta for further analyses. Western blot was used to analyze protein expression of ERK/AKT/mTOR signaling proteins (phospho-Erk (pERK), phospho-Akt (pAKT), raptor, rictor and deptor). qPCR was used to analyze gene expression of vascular/remodeling factors [vascular endothelial growth factor (Vegf), placenta growth factor (Pgf), platelet-derived growth factor (Pdgf)) and tight junction/adhesion proteins (claudin 5 (Cldn5), p-glycoprotein (Abcb1), occludin (Ocln) and tight junction protein 1 (Zo1)]. MNR reduced expression of pERK, PdgfB and Cldn5, and increased expression of Ocln and Zo1 in the sub-placenta/decidua. In MNR + hIGF1 nanoparticle sub-placenta/decidua, expression of PdgfB, Ocln and Zo1 was normalized, whilst pAkt, VegfB, Vegf receptor 1 and PdgfB receptor were increased compared to MNR. In contrast, hIGF-1 nanoparticle treatment of normal placentas reduced expression of pERK, raptor and increased expression of the mTOR inhibitor deptor. This was associated with reduced expression of VegfA, Plgf, and PdgfB. Here we have shown that the impact of hIGF-1 nanoparticle treatment is dependent on pregnancy environment. Under MNR/FGR, hIGF-1 nanoparticle treatment triggers increased expression of growth factors and normalization of EMT factors. However, under normal conditions, the response of the placenta is to decrease AKT/mTOR signaling and growth factor expression to achieve homeostasis.
Collapse
Affiliation(s)
- Baylea N. Davenport
- Center for Research in Perinatal Outcomes, University of Florida College of Medicine, Gainesville, FL, United States
- Department of Physiology and Aging, University of Florida College of Medicine, Gainesville, FL, United States
| | - Helen N. Jones
- Center for Research in Perinatal Outcomes, University of Florida College of Medicine, Gainesville, FL, United States
- Department of Physiology and Aging, University of Florida College of Medicine, Gainesville, FL, United States
| | - Rebecca L. Wilson
- Center for Research in Perinatal Outcomes, University of Florida College of Medicine, Gainesville, FL, United States
- Department of Physiology and Aging, University of Florida College of Medicine, Gainesville, FL, United States
| |
Collapse
|
99
|
Liang L, Li Y, Ying B, Huang X, Liao S, Yang J, Liao G. Mutation-associated transcripts reconstruct the prognostic features of oral tongue squamous cell carcinoma. Int J Oral Sci 2023; 15:1. [PMID: 36593250 PMCID: PMC9807648 DOI: 10.1038/s41368-022-00210-3] [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: 08/14/2022] [Revised: 11/02/2022] [Accepted: 11/06/2022] [Indexed: 01/04/2023] Open
Abstract
Tongue squamous cell carcinoma is highly malignant and has a poor prognosis. In this study, we aimed to combine whole-genome sequencing, whole-genome methylation, and whole-transcriptome analyses to understand the molecular mechanisms of tongue squamous cell carcinoma better. Oral tongue squamous cell carcinoma and adjacent normal tissues from five patients with tongue squamous cell carcinoma were included as five paired samples. After multi-omics sequencing, differentially methylated intervals, methylated loop sites, methylated promoters, and transcripts were screened for variation in all paired samples. Correlations were analyzed to determine biological processes in tongue squamous cell carcinoma. We found five mutated methylation promoters that were significantly associated with mRNA and lncRNA expression levels. Functional annotation of these transcripts revealed their involvement in triggering the mitogen-activated protein kinase cascade, which is associated with cancer progression and the development of drug resistance during treatment. The prognostic signature models constructed based on WDR81 and HNRNPH1 and combined clinical phenotype-gene prognostic signature models showed high predictive efficacy and can be applied to predict patient prognostic risk in clinical settings. We identified biological processes in tongue squamous cell carcinoma that are initiated by mutations in the methylation promoter and are associated with the expression levels of specific mRNAs and lncRNAs. Collectively, changes in transcript levels affect the prognosis of tongue squamous cell carcinoma patients.
Collapse
Affiliation(s)
- Libo Liang
- grid.13291.380000 0001 0807 1581General Practice Medical Center, West China Hospital, Sichuan University, Chengdu, China
| | - Yi Li
- grid.13291.380000 0001 0807 1581State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Binwu Ying
- grid.13291.380000 0001 0807 1581Department of Laboratory Medicine, West China Hospital, Sichuan University, Chengdu, China
| | - Xinyan Huang
- grid.13291.380000 0001 0807 1581West China School/Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Shenling Liao
- grid.13291.380000 0001 0807 1581Department of Laboratory Medicine, West China Hospital, Sichuan University, Chengdu, China
| | - Jiajin Yang
- grid.13291.380000 0001 0807 1581West China School/Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Ga Liao
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China. .,Department of Information Management, Department of Stomatology Informatics, West China Hospital of Stomatology, Sichuan University, Chengdu, China.
| |
Collapse
|
100
|
Functional roles of long noncoding RNA MALAT1 in gynecologic cancers. CLINICAL & TRANSLATIONAL ONCOLOGY : OFFICIAL PUBLICATION OF THE FEDERATION OF SPANISH ONCOLOGY SOCIETIES AND OF THE NATIONAL CANCER INSTITUTE OF MEXICO 2023; 25:48-65. [PMID: 36042115 DOI: 10.1007/s12094-022-02914-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/11/2022] [Accepted: 07/25/2022] [Indexed: 02/06/2023]
Abstract
Gynecologic cancers are reproductive disorders characterized by pelvic pain and infertility. The identification of new predictive markers and therapeutic targets for the treatment of gynecologic cancers is urgently necessary. One of the recent successes in gynecologic cancers research is identifying the role of signaling pathways in the pathogenesis of the disease. Recent experiments showed long noncoding RNAs (lncRNA) can be novel therapeutic approaches for the diagnosis and treatment of gynecologic cancers. LncRNA are transcribed RNA molecules that play pivotal roles in multiple biological processes by regulating the different steps of gene expression. Metastasis-associated lung adenocarcinoma transcript-1 (MALAT1) is a well-known lncRNA that plays functional roles in gene expression, RNA processing, and epigenetic regulation. High expression of MALAT1 is closely related to numerous human diseases. It is generally believed that MALAT1 expression is associated with cancer cell growth, autophagy, invasion, and metastasis. MALAT1 by targeting multiple signaling pathways and microRNAs (miRNAs) could contribute to the pathogenesis of gynecologic cancers. In this review, we will summarize functional roles of MALAT1 in the most common gynecologic cancers, including endometrium, breast, ovary, and cervix.
Collapse
|