1
|
Zhao Y, Yang K, Chen Y, Lv Z, Wang Q, Zhong Y, Chen X. Machine learning-based pan-cancer study of classification and mechanism of BRAF inhibitor resistance. Transl Cancer Res 2024; 13:6645-6660. [PMID: 39816555 PMCID: PMC11730697 DOI: 10.21037/tcr-24-961] [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: 06/11/2024] [Accepted: 10/25/2024] [Indexed: 01/18/2025]
Abstract
Background V-raf murine sarcoma viral oncogene homolog B1 (BRAF) inhibitor (BRAFi) therapy resistance affects approximately 15% of cancer patients, leading to disease recurrence and poor prognosis. The aim of the study was to develop a machine-learning based method to identify patients who are at high-risk of BRAFi resistance and potential biomarker. Methods From Cancer Cell Line Encyclopedia (CCLE) and Genomics of Drug Sensitivity in Cancer (GDSC) databases, we collected RNA sequencing and half maximal inhibitory concentration (IC50) data from 235 pan-cancer cell lines and then identified 37 significant differential expression genes associated with BRAFi resistance. Employing machine learning (ML) models, we successfully classified cell lines into resistant and sensitive groups, achieving robust performance in external validation datasets. Results AOX1 may play a vital part in BRAFi metabolism and resistance. Further, we found that higher mRNA expression of OXTR, H2AC13, and TBX2, and lower mRNA of SLC2A4, as detected by PCR in WM983B and SKMEL-5 cell lines, were independent risk factors for BRAFi resistance and were associated with poor prognosis. Conclusions We established a gene-expression model using ML methods, consisting of 37 variables based on RNA-seq database, which was externally validated and could be used to predict BRAFi resistance. Meanwhile, our findings provide valuable insights into the molecular mechanisms of BRAFi resistance, enabling the identification of high-risk patients.
Collapse
Affiliation(s)
- Yuhang Zhao
- Department of Integrative Medicine, Huashan Hospital, Fudan University, Shanghai, China
| | - Kai Yang
- Department of Integrative Medicine, Huashan Hospital, Fudan University, Shanghai, China
| | - Yujun Chen
- Department of Integrative Medicine, Huashan Hospital, Fudan University, Shanghai, China
| | - Zexi Lv
- Department of Integrative Medicine, Huashan Hospital, Fudan University, Shanghai, China
| | - Qing Wang
- Department of Integrative Medicine, Huashan Hospital, Fudan University, Shanghai, China
| | - Yuanyuan Zhong
- Department of Integrative Medicine, Huashan Hospital, Fudan University, Shanghai, China
| | - Xiqun Chen
- Department of Integrative Medicine, Huashan Hospital, Fudan University, Shanghai, China
| |
Collapse
|
2
|
Exposito F, Redrado M, Serrano D, Calabuig-Fariñas S, Bao-Caamano A, Gallach S, Jantus-Lewintre E, Diaz-Lagares A, Rodriguez-Casanova A, Sandoval J, San Jose-Eneriz E, Garcia J, Redin E, Senent Y, Leon S, Pio R, Lopez R, Oyarzabal J, Pineda-Lucena A, Agirre X, Montuenga LM, Prosper F, Calvo A. G9a/DNMT1 co-targeting inhibits non-small cell lung cancer growth and reprograms tumor cells to respond to cancer-drugs through SCARA5 and AOX1. Cell Death Dis 2024; 15:787. [PMID: 39488528 PMCID: PMC11531574 DOI: 10.1038/s41419-024-07156-w] [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: 02/21/2024] [Revised: 10/02/2024] [Accepted: 10/14/2024] [Indexed: 11/04/2024]
Abstract
The treatment of non-small cell lung cancer (NSCLC) patients has significantly improved with recent therapeutic strategies; however, many patients still do not benefit from them. As a result, new treatment approaches are urgently needed. In this study, we evaluated the antitumor efficacy of co-targeting G9a and DNMT1 enzymes and its potential as a cancer drug sensitizer. We observed co-expression and overexpression of G9a and DNMT1 in NSCLC, which were associated with poor prognosis. Co-targeting G9a/DNMT1 with the drug CM-272 reduced proliferation and induced cell death in a panel of human and murine NSCLC cell lines. Additionally, the transcriptomes of these cells were reprogrammed to become highly responsive to chemotherapy (cisplatin), targeted therapy (trametinib), and epigenetic therapy (vorinostat). In vivo, CM-272 reduced tumor volume in human and murine cell-derived cancer models, and this effect was synergistically enhanced by cisplatin. The expression of SCARA5 and AOX1 was induced by CM-272, and both proteins were found to be essential for the antiproliferative response, as gene silencing decreased cytotoxicity. Furthermore, the expression of SCARA5 and AOX1 was positively correlated with each other and inversely correlated with G9a and DNMT1 expression in NSCLC patients. SCARA5 and AOX1 DNA promoters were hypermethylated in NSCLC, and SCARA5 methylation was identified as an epigenetic biomarker in tumors and liquid biopsies from NSCLC patients. Thus, we demonstrate that co-targeting G9a/DNMT1 is a promising strategy to enhance the efficacy of cancer drugs, and SCARA5 methylation could serve as a non-invasive biomarker to monitor tumor progression.
Collapse
Affiliation(s)
- Francisco Exposito
- Program in Solid Tumors, Cima-Universidad de Navarra, Cancer Center Clinica Universidad de Navarra (CCUN), Pamplona, Spain
- CIBERONC, ISCIII, Madrid, Spain
- IDISNA, Pamplona, Spain
- Department of Pathology, Anatomy and Physiology, School of Medicine, University of Navarra, Pamplona, Spain
- Yale Cancer Center, New Haven, CT, USA
| | - Miriam Redrado
- Program in Solid Tumors, Cima-Universidad de Navarra, Cancer Center Clinica Universidad de Navarra (CCUN), Pamplona, Spain
- IDISNA, Pamplona, Spain
| | - Diego Serrano
- Program in Solid Tumors, Cima-Universidad de Navarra, Cancer Center Clinica Universidad de Navarra (CCUN), Pamplona, Spain
- CIBERONC, ISCIII, Madrid, Spain
- IDISNA, Pamplona, Spain
- Department of Pathology, Anatomy and Physiology, School of Medicine, University of Navarra, Pamplona, Spain
| | - Silvia Calabuig-Fariñas
- CIBERONC, ISCIII, Madrid, Spain
- Molecular Oncology Laboratory, Fundación Hospital General Universitario de Valencia, 46014, Valencia, Spain
- TRIAL Mixed Unit, Centro de Investigación Príncipe Felipe-Fundación para la Investigación del Hospital General Universitario de Valencia, 46014, Valencia, Spain
- Department of Pathology, Universitat de València, 46010, Valencia, Spain
| | - Aida Bao-Caamano
- Epigenomics Units, Cancer Epigenomics, Translational Medical Oncology Group (ONCOGAL), Health Research Institute of Santiago de Compostela (IDIS), and Department of Clinical Analysis, University Hospital Complex of Santiago de Compostela (CHUS), Roche-CHUS Joint Unit (ONCOMET), Health Research Institute of Santiago (IDIS), 15706, Santiago de Compostela, Spain, 15706, Santiago de Compostela, Spain
| | - Sandra Gallach
- CIBERONC, ISCIII, Madrid, Spain
- Molecular Oncology Laboratory, Fundación Hospital General Universitario de Valencia, 46014, Valencia, Spain
- TRIAL Mixed Unit, Centro de Investigación Príncipe Felipe-Fundación para la Investigación del Hospital General Universitario de Valencia, 46014, Valencia, Spain
| | - Eloisa Jantus-Lewintre
- CIBERONC, ISCIII, Madrid, Spain
- Molecular Oncology Laboratory, Fundación Hospital General Universitario de Valencia, 46014, Valencia, Spain
- TRIAL Mixed Unit, Centro de Investigación Príncipe Felipe-Fundación para la Investigación del Hospital General Universitario de Valencia, 46014, Valencia, Spain
- Department of Biotechnology, Universitat Politècnica de València, 46022, Valencia, Spain
| | - Angel Diaz-Lagares
- CIBERONC, ISCIII, Madrid, Spain
- Epigenomics Units, Cancer Epigenomics, Translational Medical Oncology Group (ONCOGAL), Health Research Institute of Santiago de Compostela (IDIS), and Department of Clinical Analysis, University Hospital Complex of Santiago de Compostela (CHUS), Roche-CHUS Joint Unit (ONCOMET), Health Research Institute of Santiago (IDIS), 15706, Santiago de Compostela, Spain, 15706, Santiago de Compostela, Spain
| | - Aitor Rodriguez-Casanova
- Epigenomics Units, Cancer Epigenomics, Translational Medical Oncology Group (ONCOGAL), Health Research Institute of Santiago de Compostela (IDIS), and Department of Clinical Analysis, University Hospital Complex of Santiago de Compostela (CHUS), Roche-CHUS Joint Unit (ONCOMET), Health Research Institute of Santiago (IDIS), 15706, Santiago de Compostela, Spain, 15706, Santiago de Compostela, Spain
| | - Juan Sandoval
- Biomarkers and Precision Medicine (UBMP) and Epigenomics Unit, IIS, La Fe, 46026, Valencia, Spain
| | - Edurne San Jose-Eneriz
- CIBERONC, ISCIII, Madrid, Spain
- IDISNA, Pamplona, Spain
- Division of Hemato-Oncology, Cima-Universidad de Navarra, Cancer Center Clinica Universidad de Navarra (CCUN), Pamplona, Spain
| | - Javier Garcia
- Department of Pathology, Anatomy and Physiology, School of Medicine, University of Navarra, Pamplona, Spain
| | - Esther Redin
- Program in Solid Tumors, Cima-Universidad de Navarra, Cancer Center Clinica Universidad de Navarra (CCUN), Pamplona, Spain
- CIBERONC, ISCIII, Madrid, Spain
- IDISNA, Pamplona, Spain
- Department of Pathology, Anatomy and Physiology, School of Medicine, University of Navarra, Pamplona, Spain
| | - Yaiza Senent
- Program in Solid Tumors, Cima-Universidad de Navarra, Cancer Center Clinica Universidad de Navarra (CCUN), Pamplona, Spain
- Department of Biochemistry and Genetics, School of Sciences, University of Navarra, Pamplona, Spain
| | - Sergio Leon
- Program in Solid Tumors, Cima-Universidad de Navarra, Cancer Center Clinica Universidad de Navarra (CCUN), Pamplona, Spain
- CIBERONC, ISCIII, Madrid, Spain
| | - Ruben Pio
- Program in Solid Tumors, Cima-Universidad de Navarra, Cancer Center Clinica Universidad de Navarra (CCUN), Pamplona, Spain
- CIBERONC, ISCIII, Madrid, Spain
- Department of Biochemistry and Genetics, School of Sciences, University of Navarra, Pamplona, Spain
| | - Rafael Lopez
- CIBERONC, ISCIII, Madrid, Spain
- Epigenomics Units, Cancer Epigenomics, Translational Medical Oncology Group (ONCOGAL), Health Research Institute of Santiago de Compostela (IDIS), and Department of Clinical Analysis, University Hospital Complex of Santiago de Compostela (CHUS), Roche-CHUS Joint Unit (ONCOMET), Health Research Institute of Santiago (IDIS), 15706, Santiago de Compostela, Spain, 15706, Santiago de Compostela, Spain
| | - Julen Oyarzabal
- Molecular Therapeutics Program, CIMA, CCUN, University of Navarra, Pamplona, Spain
| | | | - Xabier Agirre
- CIBERONC, ISCIII, Madrid, Spain
- IDISNA, Pamplona, Spain
- Division of Hemato-Oncology, Cima-Universidad de Navarra, Cancer Center Clinica Universidad de Navarra (CCUN), Pamplona, Spain
| | - Luis M Montuenga
- Program in Solid Tumors, Cima-Universidad de Navarra, Cancer Center Clinica Universidad de Navarra (CCUN), Pamplona, Spain
- CIBERONC, ISCIII, Madrid, Spain
- IDISNA, Pamplona, Spain
- Department of Pathology, Anatomy and Physiology, School of Medicine, University of Navarra, Pamplona, Spain
| | - Felipe Prosper
- CIBERONC, ISCIII, Madrid, Spain
- IDISNA, Pamplona, Spain
- Hematology and Cell Therapy Service, Cancer Center Clinica Universidad de Navarra (CCUN), Pamplona, Spain
| | - Alfonso Calvo
- Program in Solid Tumors, Cima-Universidad de Navarra, Cancer Center Clinica Universidad de Navarra (CCUN), Pamplona, Spain.
- CIBERONC, ISCIII, Madrid, Spain.
- IDISNA, Pamplona, Spain.
- Department of Pathology, Anatomy and Physiology, School of Medicine, University of Navarra, Pamplona, Spain.
| |
Collapse
|
3
|
Wen H, Mi Y, Li F, Xue X, Sun X, Zheng P, Liu S. Identifying the signature of NAD+ metabolism-related genes for immunotherapy of gastric cancer. Heliyon 2024; 10:e38823. [PMID: 39640811 PMCID: PMC11620085 DOI: 10.1016/j.heliyon.2024.e38823] [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: 06/29/2023] [Revised: 09/03/2024] [Accepted: 09/30/2024] [Indexed: 12/07/2024] Open
Abstract
NAD (Nicotinamide Adenine Dinucleotide) -related metabolic reprogramming in tumor cells involves multiple vital cellular processes. However, the role of NAD metabolism in immunity and the prognosis of gastric cancer (GC) remains not elucidated. Here we identified and clustered 33 NAD + metabolism-related genes (NMRGs) based on 808 GC samples from the Cancer Genome Atlas (TCGA) and the Gene Expression Omnibus (GEO) databases. Survival analysis between different groups found a poor prognosis in the GC patients with high NMRGs expression. Gene SGCE, APOD, and PPP1R14A were identified and performed high expression in GC samples, while the qRT-PCR results further confirmed that their expression levels in GC cell lines were significantly higher than those from normal human gastric mucosa epithelial cells. Based on the single-cell analysis, Gene SGCE, APOD, and PPP1R14A can potentially be novel biomarkers of tumor-associated fibroblasts (CAFs). In parallel, the proliferation and migration of GC cells were significantly hampered following the knockdown of SGCE, APOD, and PPP1R14A, particularly APOD, we confirmed that APOD knockdown can inhibit β-catenin and N-cadherin expression, while promote E-cadherin expression. This study unveils a novel NMRGs-related gene signature, highlighting APOD as a prognostic biomarker linked to the tumor microenvironment. APOD drives GC cell proliferation and metastasis through the Wnt/β-catenin/EMT signaling pathway, establishing it as a promising therapeutic target for GC patients.
Collapse
Affiliation(s)
- Huijuan Wen
- Henan Key Laboratory of Helicobacter pylori & Microbiota and Gastrointestinal Cancer, Marshall Medical Research Center, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
- Academy of medical science, Zhengzhou University, Zhengzhou, 450052, China
| | - Yang Mi
- Henan Key Laboratory of Helicobacter pylori & Microbiota and Gastrointestinal Cancer, Marshall Medical Research Center, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
- Department of Gastroenterology, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, China
| | - Fazhan Li
- Henan Key Laboratory of Helicobacter pylori & Microbiota and Gastrointestinal Cancer, Marshall Medical Research Center, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
- Academy of medical science, Zhengzhou University, Zhengzhou, 450052, China
| | - Xia Xue
- Henan Key Laboratory of Helicobacter pylori & Microbiota and Gastrointestinal Cancer, Marshall Medical Research Center, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
- Department of Gastroenterology, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, China
| | - Xiangdong Sun
- Henan Key Laboratory of Helicobacter pylori & Microbiota and Gastrointestinal Cancer, Marshall Medical Research Center, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
- Academy of medical science, Zhengzhou University, Zhengzhou, 450052, China
| | - Pengyuan Zheng
- Henan Key Laboratory of Helicobacter pylori & Microbiota and Gastrointestinal Cancer, Marshall Medical Research Center, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
- Academy of medical science, Zhengzhou University, Zhengzhou, 450052, China
- Department of Gastroenterology, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, China
| | - Simeng Liu
- Henan Key Laboratory of Helicobacter pylori & Microbiota and Gastrointestinal Cancer, Marshall Medical Research Center, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
- Department of Gastroenterology, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, China
| |
Collapse
|
4
|
Xuan M, Gu X, Xing H. Multi-omic analysis identifies the molecular mechanism of hepatocellular carcinoma with cirrhosis. Sci Rep 2024; 14:23832. [PMID: 39394373 PMCID: PMC11470084 DOI: 10.1038/s41598-024-75609-5] [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: 07/16/2024] [Accepted: 10/07/2024] [Indexed: 10/13/2024] Open
Abstract
Hepatocellular carcinoma with cirrhosis promotes the advancement of malignancy and the development of fibrosis in normal liver tissues. Understanding the pathological mechanisms underlying the development of HCC with cirrhosis is important for developing effective therapeutic strategies. Herein, the RNA-sequencing (RNA-seq) data and corresponding clinical features of patients with HCC were extracted from The Cancer Genome Atlas (TCGA) database using the University of California Santa Cruz (UCSC) Xena platform. The enrichment degree of hallmarkers for each TCGA-LIHC cohort was quantified by ssGSEA algorithm. Weighted gene co-expression network analysis (WGCNA) revealed two gene module eigengenes (MEs) associated with cirrhosis, namely, MEbrown and MEgreen. Analysis of these modules using AUCell showed that MEbrown had higher enrichment scores in all immune cells, whereas MEgreen had higher enrichment scores in malignant cells. The CellChat package revealed that both immune and malignant cells contributed to the fibrotic activity of myofibroblasts through diverse signaling pathways. Additionally, spatial transcriptomic data showed that hepatocytes, proliferating hepatocytes, macrophages, and myofibroblasts were located in closer proximity in HCC tissues. These cells may potentially participate in the process of stimulating myofibroblast fibrotic activity, which may be related to the development of liver fibrosis. In summary, we made full use of multi-omics data to explore gene networks and cell types that may be involved in the development and progression of cirrhosis in HCC.
Collapse
Affiliation(s)
- Mengjuan Xuan
- Department of Infectious Diseases, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, China
| | - Xinyu Gu
- Department of Oncology, The First Affiliated Hospital, College of Clinical Medicine, Henan University of Science and Technology, Luoyang, 471000, Henan, China
| | - Huiwu Xing
- Department of Pediatric Surgery, The First Affiliated Hospital of Zhengzhou University, No. 1 Jianshe East Road, Erqi District, Zhengzhou, 450052, Henan, China.
| |
Collapse
|
5
|
Paskeh MDA, Ghadyani F, Hashemi M, Abbaspour A, Zabolian A, Javanshir S, Razzazan M, Mirzaei S, Entezari M, Goharrizi MASB, Salimimoghadam S, Aref AR, Kalbasi A, Rajabi R, Rashidi M, Taheriazam A, Sethi G. Biological impact and therapeutic perspective of targeting PI3K/Akt signaling in hepatocellular carcinoma: Promises and Challenges. Pharmacol Res 2023; 187:106553. [PMID: 36400343 DOI: 10.1016/j.phrs.2022.106553] [Citation(s) in RCA: 66] [Impact Index Per Article: 33.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/12/2022] [Revised: 11/09/2022] [Accepted: 11/10/2022] [Indexed: 11/17/2022]
Abstract
Cancer progression results from activation of various signaling networks. Among these, PI3K/Akt signaling contributes to proliferation, invasion, and inhibition of apoptosis. Hepatocellular carcinoma (HCC) is a primary liver cancer with high incidence rate, especially in regions with high prevalence of viral hepatitis infection. Autoimmune disorders, diabetes mellitus, obesity, alcohol consumption, and inflammation can also lead to initiation and development of HCC. The treatment of HCC depends on the identification of oncogenic factors that lead tumor cells to develop resistance to therapy. The present review article focuses on the role of PI3K/Akt signaling in HCC progression. Activation of PI3K/Akt signaling promotes glucose uptake, favors glycolysis and increases tumor cell proliferation. It inhibits both apoptosis and autophagy while promoting HCC cell survival. PI3K/Akt stimulates epithelial-to-mesenchymal transition (EMT) and increases matrix-metalloproteinase (MMP) expression during HCC metastasis. In addition to increasing colony formation capacity and facilitating the spread of tumor cells, PI3K/Akt signaling stimulates angiogenesis. Therefore, silencing PI3K/Akt signaling prevents aggressive HCC cell behavior. Activation of PI3K/Akt signaling can confer drug resistance, particularly to sorafenib, and decreases the radio-sensitivity of HCC cells. Anti-cancer agents, like phytochemicals and small molecules can suppress PI3K/Akt signaling by limiting HCC progression. Being upregulated in tumor tissues and clinical samples, PI3K/Akt can also be used as a biomarker to predict patients' response to therapy.
Collapse
Affiliation(s)
- Mahshid Deldar Abad Paskeh
- Department of Genetics, Faculty of Advanced Science and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran; Farhikhtegan Medical Convergence sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Fatemeh Ghadyani
- Farhikhtegan Medical Convergence sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Mehrdad Hashemi
- Department of Genetics, Faculty of Advanced Science and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran; Farhikhtegan Medical Convergence sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Alireza Abbaspour
- Cellular and Molecular Research Center,Qazvin University of Medical Sciences, Qazvin, Iran
| | - Amirhossein Zabolian
- Resident of department of Orthopedics, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Salar Javanshir
- Young Researchers and Elite Club, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Mehrnaz Razzazan
- Medical Student, Student Research Committee, Golestan University of Medical Sciences, Gorgan, Iran
| | - Sepideh Mirzaei
- Department of Biology, Faculty of Science, Islamic Azad University, Science and Research Branch, Tehran, Iran
| | - Maliheh Entezari
- Department of Genetics, Faculty of Advanced Science and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran; Farhikhtegan Medical Convergence sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | | | - Shokooh Salimimoghadam
- Department of Biochemistry and Molecular Biology, Faculty of Veterinary Medicine, Shahid Chamran University of Ahvaz, Ahvaz, Iran
| | - Amir Reza Aref
- Belfer Center for Applied Cancer Science, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA; Translational Sciences, Xsphera Biosciences Inc. 6, Tide Street, Boston, MA 02210, USA
| | - Alireza Kalbasi
- Department of Pharmacy, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Romina Rajabi
- Faculty of Veterinary Medicine, Islamic Azad University, Science and Research Branch, Tehran, Iran.
| | - Mohsen Rashidi
- Department Pharmacology, Faculty of Medicine, Mazandaran University of Medical Sciences, Sari, Iran; The Health of Plant and Livestock Products Research Center, Mazandaran University of Medical Sciences, Sari, Iran.
| | - Afshin Taheriazam
- Farhikhtegan Medical Convergence sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran; Department of Orthopedics, Faculty of medicine, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran.
| | - Gautam Sethi
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117600, Singapore; NUS Centre for Cancer Research, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117599, Singapore.
| |
Collapse
|
6
|
Antitumor Effect of Pseudolaric Acid B Involving Regulation of Notch1/Akt Signaling Response in Human Hepatoma Cell In Vitro. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2022; 2022:5353686. [PMID: 35747382 PMCID: PMC9213129 DOI: 10.1155/2022/5353686] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/10/2022] [Revised: 05/04/2022] [Accepted: 05/06/2022] [Indexed: 12/11/2022]
Abstract
Background Liver cancer, particularly hepatocellular carcinoma (HCC), is the fourth leading cause of cancer-related death worldwide. Sorafenib is a crucial drug for the treatment of advanced HCC, but it is difficult to meet the challenge of increasing clinical demands due to its severe side effects and drug resistance. Hence, development of novel antitumor drugs is urged. Previous studies showed that pseudolaric acid B (PAB) could reduce the expression of protein kinase B (PKB/Akt), a downstream effector of Notch signaling, facilitating cell apoptosis in HCC. The disruption of Notch signaling was verified to exacerbate malignant progression and drug resistance, however, the antitumor effect of PAB on Notch signaling in HCC remains unclear. Thus, this study aims to investigate the anti-HCC effect of PAB in association with the regulation of Notch1/Akt signaling. Methods CCK-8 assay and transwell assay were used to examine the cell proliferation and invasion in Huh7 cells after treatment with PAB and a Notch inhibitor DAPT. Moreover, the cell cycle of Huh7 cells after treatment with PAB was analyzed using flow cytometry. Finally, the changes of Notch1, Jagged1, Hes1, and Akt expression at the protein and mRNA level in Notch1/Akt signaling in Huh7 cells after treatment with PAB and DAPT were analyzed using immunofluorescence assay and real-time qPCR. Results The proliferation rate of Huh7 cells exposed to PAB of 0.5, 1, 2, 4, 8, 10, 20, 40, 80, 100, and 200 μmol/L revealed a time-and dose-dependent decrease in vitro, showing cell cycle arrest at G2/M phase (P < 0.05). Furthermore, compared with the untreated group, at the concentration of 40 μmol/L, the proliferation rate and invasion rate of Huh7 cells in PAB, DAPT, and PAB-DAPT combination (PAB + DAPT) group were significantly decreased (P < 0.05), but the PAB + DAPT showed no synergistic antiproliferation and anti-invasion effect in comparison with PAB treatment alone (P > 0.05). In addition, compared with the untreated group, PAB and DAPT alone significantly downregulated the expression of Notch1, Jagged1, Hes1, Akt mRNA, or/and protein in Huh7 cells (P < 0.05), but there was no significant difference in synergistic downregulated effect between the PAB + DAPT group and the PAB group (P > 0.05). Conclusion PAB can suppress proliferation and invasion of HCC cells through downregulating the expression of Notch1/Akt signaling protein and mRNA, and may be a potential novel antitumor drug candidate for the clinical treatment of HCC in the future.
Collapse
|