1
|
Yao J, Ning B, Ding J. The gut microbiota: an emerging modulator of drug resistance in hepatocellular carcinoma. Gut Microbes 2025; 17:2473504. [PMID: 40042184 PMCID: PMC11901387 DOI: 10.1080/19490976.2025.2473504] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Revised: 11/08/2024] [Accepted: 02/21/2025] [Indexed: 03/14/2025] Open
Abstract
Liver cancer is usually diagnosed at an advanced stage and is the third most common cause of cancer-related death worldwide. In addition to the lack of effective treatment options, resistance to therapeutic drugs is a major clinical challenge. The gut microbiota has recently been recognized as one of the key factors regulating host health. The microbiota and its metabolites can directly or indirectly regulate gene expression in the liver, leading to gut-liver axis dysregulation, which is closely related to liver cancer occurrence and the treatment response. Gut microbiota disturbance may participate in tumor progression and drug resistance through metabolite production, gene transfer, immune regulation, and other mechanisms. However, systematic reviews on the role of the gut microbiota in drug resistance in liver cancer are lacking. Herein, we review the relationships between the gut microbiota and the occurrence and drug resistance of hepatocellular carcinoma, summarize the emerging mechanisms underlying gut microbiota-mediated drug resistance, and propose new personalized treatment options to overcome this resistance.
Collapse
Affiliation(s)
- Jiali Yao
- Clinical Cancer Institute, Center for Translational Medicine, Naval Medical University, Shanghai, China
| | - Beifang Ning
- Department of Gastroenterology, Changzheng Hospital, Naval Medical University, Shanghai, China
| | - Jin Ding
- Clinical Cancer Institute, Center for Translational Medicine, Naval Medical University, Shanghai, China
| |
Collapse
|
2
|
Mohamed NM, Mohamed RH, Kennedy JF, Elhefnawi MM, Hamdy NM. A comprehensive review and in silico analysis of the role of survivin (BIRC5) in hepatocellular carcinoma hallmarks: A step toward precision. Int J Biol Macromol 2025; 311:143616. [PMID: 40306500 DOI: 10.1016/j.ijbiomac.2025.143616] [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: 02/13/2025] [Revised: 04/25/2025] [Accepted: 04/27/2025] [Indexed: 05/02/2025]
Abstract
Hepatocellular carcinoma (HCC) is a complex malignancy driven by the dysregulation of multiple cellular pathways. Survivin, a key member of the inhibitor of apoptosis (IAP) family, plays a central role in HCC tumorigenesis and progression. Despite significant research, a comprehensive understanding of the contributions of survivin to the hallmarks of cancer, its molecular network, and its potential as a therapeutic target remains incomplete. In this review, we integrated bioinformatics analysis with an extensive literature review to provide deeper insights into the role of survivin in HCC. Using bioinformatics tools such as the Human Protein Atlas, GEPIA, STRING, TIMER, and Metascape, we analyzed survivin expression and its functional associations and identified the top 20 coexpressed genes in HCC. These include TK1, SPC25, SGO2, PTTG1, PRR11, PLK1, NCAPH, KPNA2, KIF2C, KIF11, HJURP, GTSE1, FOXM1, CEP55, CENPA, CDCA3, CDC45, CCNB2, CCNB1 and CTD-2510F5.4. Our findings also revealed significant protein-protein interactions among these genes, which were enriched in pathways associated with the FOXM1 oncogenic signaling cascade, and biological processes such as cell cycle regulation, mitotic checkpoints, and diseases such as liver neoplasms. We also discussed the involvement of survivin in key oncogenic pathways, including the PI3K/AKT, WNT/β-catenin, Hippo, and JAK/STAT3 pathways, and its role in modulating cell cycle checkpoints, apoptosis, and autophagy. Furthermore, we explored its interactions with the tumor microenvironment, particularly its impact on immune modulation through myeloid-derived suppressor cells (MDSCs), tumor-associated macrophages, and natural killer cell function in HCC. Additionally, we highlighted its involvement in alkylglycerone phosphate synthase (AGPS)-mediated lipid reprogramming and identified important gaps in the survivin network that warrant further investigation. This review also examined the role of survivin in cancer stemness, inflammation, and virally mediated hepatocarcinogenesis. We evaluated its potential as a diagnostic, prognostic, predictive, and pharmacodynamic biomarker in HCC, emphasizing its relevance in precision medicine. Finally, we summarized emerging survivin-targeted therapeutics and ongoing clinical trials, underscoring the need for novel strategies to effectively target survivin in HCC.
Collapse
Affiliation(s)
- Nermin M Mohamed
- Department of Biochemistry, Faculty of Pharmacy, Ain Shams University, Abassia, 11566 Cairo, Egypt
| | - Rania Hassan Mohamed
- Department of Biochemistry, Faculty of Science, Ain Shams University, Abassia, 11566 Cairo, Egypt
| | - John F Kennedy
- Chembiotech Laboratories, Kyrewood House, Tenbury Wells, Worcestershire, United Kingdom
| | - Mahmoud M Elhefnawi
- Biomedical Informatics and Chemoinformatics Group, Informatics and Systems Department, National Research Centre, Cairo, Egypt.
| | - Nadia M Hamdy
- Department of Biochemistry, Faculty of Pharmacy, Ain Shams University, Abassia, 11566 Cairo, Egypt.
| |
Collapse
|
3
|
Tu L, Luo J, Yin Y, Yu H. Bioinformatics analysis across pan-cancer and experimental validation in hepatocellular carcinoma revealed the oncogenic role of SF3B6. Front Pharmacol 2025; 16:1516534. [PMID: 40356980 PMCID: PMC12067594 DOI: 10.3389/fphar.2025.1516534] [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: 03/06/2025] [Accepted: 04/15/2025] [Indexed: 05/15/2025] Open
Abstract
Background Splicing factor 3b subunit 6(SF3B6), a subunit of the SF3B complex, regulates the process of RNA splicing by recognizing the branch point adenosine in pre-mRNA and facilitating the interaction between U2 snRNA and the branch point sequence. Currently, there is no systematic multi-omics study exploring the diagnostic, prognostic, and immunotherapy predictive value of SF3B6 in pan-cancer, nor is its role in hepatocellular carcinoma (HCC) clear. Methods We utilized various databases to systematically examine the expression and genetic variation of SF3B6 across multiple cancer types, assessing its relationship with diagnosis, prognosis, immune infiltration, immunotherapy response, and associated signaling pathways. Additionally, we investigated the correlation between SF3B6 and prognosis, clinicopathological features, and treatment responses in HCC, as well as the roles of its related alternative splicing isoforms. Finally, we conducted in vitro experiments to validate the effects of SF3B6 on the proliferation, migration, invasion, apoptosis, and cell cycle progression of liver cancer cells. Results Results indicate that SF3B6 was highly expressed in various cancers and regulated by copy number variations and DNA methylation. The elevated expression of SF3B6 demonstrated predictive value for cancer diagnosis, prognosis, and responses to immunotherapy. Functional enrichment analysis suggests that SF3B6 was closely associated with pathways related to tumor immunity, tumor metabolism, and cell cycle. Additionally, high SF3B6 expression was an independent risk factor for overall survival and correlated with poor alpha-fetoprotein levels, pathological grading, clinical staging, and reduced responses to sorafenib and transarterial chemoembolization treatment in HCC. Interestingly, SF3B6 was associated with variant splicing isotypes of genes involved in the G2M checkpoint and DNA repair pathways, including NEIL3, NEK2, KIF4A, TROAP, and FANCD2. Moreover, SF3B6 was highly expressed in liver cancer cells, promoting the proliferation, migration, and invasion of cancer cells, inhibiting apoptosis, and regulating the transition from the S phase to the G2M phase of the cell cycle. Conclusion We emphasize that SF3B6 has the potential to serve as a biomarker for predicting cancer diagnosis, prognosis, and immunotherapy responses, especially in HCC. SF3B6 and its related alternative splicing isoforms promote the occurrence and progression of HCC and may serve as potential therapeutic targets.
Collapse
Affiliation(s)
- Linshuang Tu
- Department of Gastroenterology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Jiefu Luo
- Department of Hepatobiliary Surgery, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Ya Yin
- Department of Gastroenterology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Huihong Yu
- Department of Gastroenterology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| |
Collapse
|
4
|
He F, Liu H, Zhao F. The oncogenic role of FOXM1 in hepatocellular carcinoma: molecular mechanisms, clinical significance, and therapeutic potentials. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025:10.1007/s00210-025-04144-5. [PMID: 40266300 DOI: 10.1007/s00210-025-04144-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/10/2025] [Accepted: 04/03/2025] [Indexed: 04/24/2025]
Abstract
Hepatocellular carcinoma (HCC) remains a major global health challenge due to its aggressive nature and limited treatment options. This review aims to clarify the oncogenic role of FOXM1 in HCC and its potential as a therapeutic target. We examine how FOXM1 drives cancer development by regulating key cellular processes such as cell cycle progression, proliferation, metastasis, and therapy resistance. The review details mechanisms that control FOXM1 activity, including transcriptional regulation by upstream factors, post-transcriptional modulation via non-coding RNAs, and epigenetic modifications. We also explore how FOXM1 interacts with critical signaling pathways, such as AKT, p53, ERK, Hedgehog, STAT3, and Wnt/β-catenin, to promote metabolic reprogramming, angiogenesis, and the maintenance of cancer stem cell properties. In the therapeutic section, we assess emerging strategies that target FOXM1, including small-molecule inhibitors, proteasome inhibitors, and immunotherapeutic approaches, to improve treatment outcomes for HCC patients. This comprehensive review highlights the pivotal role of FOXM1 in HCC pathogenesis and provides novel avenues for targeted intervention.
Collapse
Affiliation(s)
- Fangyu He
- Department of Hematology, The Second Hospital of Dalian Medical University, Dalian, China
| | - He Liu
- Department of Infection Control and Prevention, The Second Hospital of Dalian Medical University, Dalian, China
| | - Fangcheng Zhao
- Department of Infectious Diseases, The Second Hospital of Dalian Medical University, Dalian, China.
| |
Collapse
|
5
|
Kinoshita H, Takenouchi K, Tsukamoto N, Ohnuki K, Suzuki T, Nakatsura T. Identification of 68 HLA-A24 and -A2-restricted cytotoxic T lymphocyte-inducing peptides derived from 10 common cancer-specific antigens frequently expressed in various solid cancers. Neoplasia 2025; 61:101135. [PMID: 39938154 PMCID: PMC11869973 DOI: 10.1016/j.neo.2025.101135] [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: 12/04/2024] [Revised: 01/31/2025] [Accepted: 02/03/2025] [Indexed: 02/14/2025]
Abstract
Targeting cancer antigens expressed in cancer cells is necessary to develop cancer-specific immunotherapy. We have performed immunohistochemical analysis of various solid cancer specimens, adding ROBO1, AFP, TGFBI, EphB4, CLDN1, and LAT1 to the previously studied glypican-3 (GPC3), HSP105α, FOXM1, and SPARC, and found that these 10 common cancer antigens are sufficient to cover most solid cancers. These antigens were frequently expressed in various solid cancers but shown to be rarely ex-pressed, with some exceptions, in non-cancerous normal organs adjacent to the cancer. In this study, we predicted 72 and 73 peptides that bind to HLA-A24 and -A2 in silico from the full-length amino acid sequences of these 10 common cancer antigens and immunized each HLA transgenic mouse with a cocktail of synthesized peptides together with the poly I:CLC three times weekly to analyze the antigen-specific immune response. As a result, 68 peptide sequences (30 and 38, respectively) were identified that had higher cytotoxic T lymphocyte (CTL) induction ability than GPC3 298-306 and GPC3 144-152 used in the clinical trials. Furthermore, experiments with cocktail peptide vaccines using mouse models expressing subcutaneous tumors of each antigen showed promising results in terms of safety and efficacy. These peptides identified in this study, derived from 10 common cancer antigens covering all solid cancers, are expected to be clinically applicable as cocktail peptide vaccines.
Collapse
Affiliation(s)
- Hiroki Kinoshita
- Graduate School of Biological Sciences, Tokyo University of Science, 2641 Yamazaki, Noda, Chiba 278-8510, Japan; Division of Cancer Immunotherapy, Exploratory Oncology Research and Clinical Trial Center, National Cancer Center, 6-5-1 Kashiwanoha, Kashiwa, Chiba 277-8577, Japan
| | - Kazumasa Takenouchi
- Division of Cancer Immunotherapy, Exploratory Oncology Research and Clinical Trial Center, National Cancer Center, 6-5-1 Kashiwanoha, Kashiwa, Chiba 277-8577, Japan
| | - Nobuo Tsukamoto
- Division of Cancer Immunotherapy, Exploratory Oncology Research and Clinical Trial Center, National Cancer Center, 6-5-1 Kashiwanoha, Kashiwa, Chiba 277-8577, Japan
| | - Kazunobu Ohnuki
- Division of Cancer Immunotherapy, Exploratory Oncology Research and Clinical Trial Center, National Cancer Center, 6-5-1 Kashiwanoha, Kashiwa, Chiba 277-8577, Japan
| | - Toshihiro Suzuki
- Division of Cancer Immunotherapy, Exploratory Oncology Research and Clinical Trial Center, National Cancer Center, 6-5-1 Kashiwanoha, Kashiwa, Chiba 277-8577, Japan
| | - Tetsuya Nakatsura
- Division of Cancer Immunotherapy, Exploratory Oncology Research and Clinical Trial Center, National Cancer Center, 6-5-1 Kashiwanoha, Kashiwa, Chiba 277-8577, Japan.
| |
Collapse
|
6
|
Hua X, Xiang K, Liang A, Chang M, Jia S, Shang Z, Jiang Y, Liu X, Mao C. A novel peptide CP29L, selected from the phage displayed cyclic random heptapeptide library, demonstrates its potent inhibitory effects to liver cancer HCCLM3 cells by targeting FOXM1. Eur J Pharmacol 2025; 989:177246. [PMID: 39755244 DOI: 10.1016/j.ejphar.2024.177246] [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: 08/09/2024] [Revised: 12/24/2024] [Accepted: 12/30/2024] [Indexed: 01/06/2025]
Abstract
FOXM1 is the "Achilles' heel" of cancers and hence the potential therapeutic target for anticancer drug discovery. In this work, we selected high affinity peptides against the protein of human DNA binding domain of FOXM1 (FOXM1-DBD) from the disulfide-constrained, phage displayed random cyclic heptapeptide library Ph.D.-C7C. We obtained a novel peptide, 9 R-CP29L, which was identified to be a potent anticancer peptide with IC50 values of 9.0 and 11.1 μM at 24 h for HCCLM3 and MD-MBA-231 cells respectively. Molecular docking, CETSA, ITC and immunoblot assays demonstrated that 9 R-CP29L can potentially specifically bind to FOXM1-DBD with a Kd value of 1.25 μM and reduced the expression of FOXM1. In addition, Annexin V/PI flow cytometry, AO/EB staining, PI flow cytometry, clone formation and Transwell assays revealed that 9 R-CP29L also induced cell apoptosis and cell cycle arrest while inhibited the proliferation and migration of HCCLM3 cells. The findings were further supported by the results of qRT-PCR and immunoblot assays for the associated gene (CMYC, CDC25B, BAX, CASPASE3 and MMP2, etc) expression in HCCLM3 cells. Finally, in vivo experiment showed that 9 R-CP29 significantly reduced the tumor growth and downregulated the expression of FOXM1 in HCCLM3 xenograft nude mouse models. Taking together, our work provides a novel FOXM1 targeted peptide which has potential in both anticancer drug development and scientific researches.
Collapse
Affiliation(s)
- Xinyi Hua
- School of Life Science and Engineering, Southwest Jiaotong University, Chengdu, 610031, Sichuan province, PR China; School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu, 610031, Sichuan, PR China
| | - Kun Xiang
- School of Life Science and Engineering, Southwest Jiaotong University, Chengdu, 610031, Sichuan province, PR China
| | - Anping Liang
- School of Life Science and Engineering, Southwest Jiaotong University, Chengdu, 610031, Sichuan province, PR China; School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu, 610031, Sichuan, PR China
| | - Miao Chang
- School of Life Science and Engineering, Southwest Jiaotong University, Chengdu, 610031, Sichuan province, PR China
| | - Shijie Jia
- School of Life Science and Engineering, Southwest Jiaotong University, Chengdu, 610031, Sichuan province, PR China
| | - Zhixian Shang
- School of Life Science and Engineering, Southwest Jiaotong University, Chengdu, 610031, Sichuan province, PR China
| | - Yuhong Jiang
- School of Life Science and Engineering, Southwest Jiaotong University, Chengdu, 610031, Sichuan province, PR China
| | - Xinrong Liu
- School of Life Science and Engineering, Southwest Jiaotong University, Chengdu, 610031, Sichuan province, PR China
| | - Canquan Mao
- School of Life Science and Engineering, Southwest Jiaotong University, Chengdu, 610031, Sichuan province, PR China.
| |
Collapse
|
7
|
Ebner LJA, Karademir D, Nötzli S, Wögenstein GM, Samardzija M, Grimm C. Oxygen-dependent alternative mRNA splicing and a cone-specific motor protein revealed by single-cell RNA sequencing in hypoxic retinas. Exp Eye Res 2025; 251:110190. [PMID: 39638278 DOI: 10.1016/j.exer.2024.110190] [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: 11/29/2024] [Accepted: 11/29/2024] [Indexed: 12/07/2024]
Abstract
Restricted oxygen supply in the aging eye may lead to hypoxic conditions in the outer retina and contribute not only to physiological aging but also to nonhereditary degenerative retinal diseases. To understand the hypoxic response of specific retinal cell types, we performed single-cell RNA sequencing of retinas isolated from mice exposed to hypoxia. Significantly upregulated expression of marker genes in hypoxic clusters confirmed a general transcriptional response to hypoxia. By focusing on the hypoxic response in photoreceptors, we identified and confirmed a kinesin motor protein (Kif4) that was specifically and strongly induced in hypoxic cones. In contrast, RNA-binding proteins Rbm3 and Cirbp were differentially expressed across clusters but demonstrated isoform switching in hypoxia. The resulting short variants of these gene transcripts are connected to epitranscriptomic regulation, a notion supported by the differential expression of writers, readers and erasers of m6A RNA methylations in the hypoxic retina. Our data indicate that retinal cells adapt to hypoxic conditions by adjusting their transcriptome at various levels including gene expression, alternative splicing and the epitranscriptome. Adaptational processes may be cell-type specific as exemplified by the cone-specific upregulation of Kif4 or general like alternative splicing of RNA binding proteins.
Collapse
Affiliation(s)
- Lynn J A Ebner
- Laboratory for Retinal Cell Biology, Department of Ophthalmology, University Hospital Zurich, University of Zurich, Wagistrasse 14, Schlieren, 8952, Zurich, Switzerland
| | - Duygu Karademir
- Laboratory for Retinal Cell Biology, Department of Ophthalmology, University Hospital Zurich, University of Zurich, Wagistrasse 14, Schlieren, 8952, Zurich, Switzerland
| | - Sarah Nötzli
- Laboratory for Retinal Cell Biology, Department of Ophthalmology, University Hospital Zurich, University of Zurich, Wagistrasse 14, Schlieren, 8952, Zurich, Switzerland
| | - Gabriele M Wögenstein
- Laboratory for Retinal Cell Biology, Department of Ophthalmology, University Hospital Zurich, University of Zurich, Wagistrasse 14, Schlieren, 8952, Zurich, Switzerland; Neuroscience Center Zurich (ZNZ), University of Zurich, Winterthurerstrasse 190, 8057, Zurich, Switzerland
| | - Marijana Samardzija
- Laboratory for Retinal Cell Biology, Department of Ophthalmology, University Hospital Zurich, University of Zurich, Wagistrasse 14, Schlieren, 8952, Zurich, Switzerland
| | - Christian Grimm
- Laboratory for Retinal Cell Biology, Department of Ophthalmology, University Hospital Zurich, University of Zurich, Wagistrasse 14, Schlieren, 8952, Zurich, Switzerland; Neuroscience Center Zurich (ZNZ), University of Zurich, Winterthurerstrasse 190, 8057, Zurich, Switzerland.
| |
Collapse
|
8
|
Ba L, Zhao Z, Zhang C, Chu Y, Wu C. Expression and prognostic impact of hypoxia- and immune escape-related genes in triple-negative breast cancer: A comprehensive analysis. Int Immunopharmacol 2025; 146:113810. [PMID: 39689602 DOI: 10.1016/j.intimp.2024.113810] [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: 09/19/2024] [Revised: 11/23/2024] [Accepted: 12/04/2024] [Indexed: 12/19/2024]
Abstract
Triple-negative breast cancer (TNBC) is an aggressive subtype of breast cancer that lacks effective therapeutic options. Hypoxia and immune escape are critical factors that contribute to the progression of and resistance to therapy in patients with TNBC. Nevertheless, few studies have comprehensively analyzed hypoxia and immune escape in patients with TNBC. This study aimed to examine the expression of hypoxia- and immune escape-related genes in TNBC and their influence on prognosis. TNBC datasets were downloaded and processed from The Cancer Genome Atlas and Gene Expression Omnibus. Differential expression analysis identified 4949 differentially expressed genes, between TNBC and normal tissues. The intersection yielded 116 hypoxia- and immune escape-related differentially expressed genes (H&IERDEGs), including KIF4A, BIRC5, and BUB1. Enrichment analyses indicated that H&IERDEGs were significantly enriched in biological processes, including cell chemotaxis, leukocyte migration, and cytokine-cytokine receptor interaction. Subsequently, weighted gene co-expression network analysis identified 43 module genes that were found to define two TNBC subtypes. We constructed a prognostic risk model consisting of eight signature genes, which demonstrated a high predictive performance to predict the overall survival (OS) of patients with TNBC with an area under the curve (AUC) exceeding 0.9 at 1 year survival. This indicates that the model effectively differentiates between outcomes, reflecting its robust performance. This study investigated the roles and potential mechanisms of hypoxia- and immune escape-related genes in TNBC and constructed a prognostic risk model with a high predictive performance. These findings offer novel molecular markers and potential therapeutic targets for TNBC.
Collapse
Affiliation(s)
- Li Ba
- Department of Ultrasound, First Affiliated Hospital of Harbin Medical University, Harbin 150001, PR China
| | - Zhiyu Zhao
- Department of Ultrasound, First Affiliated Hospital of Harbin Medical University, Harbin 150001, PR China; Laboratory of Medical Genetics, Harbin Medical University, Harbin 150001, PR China; Key Laboratory of Preservation of Human Genetic Resources and Disease Control in China (Harbin Medical University), Ministry of Education, Harbin 150001, PR China
| | - Chunmei Zhang
- Department of Ultrasound, First Affiliated Hospital of Harbin Medical University, Harbin 150001, PR China
| | - Yinzhu Chu
- Department of Ultrasound, First Affiliated Hospital of Harbin Medical University, Harbin 150001, PR China
| | - Changjun Wu
- Department of Ultrasound, First Affiliated Hospital of Harbin Medical University, Harbin 150001, PR China.
| |
Collapse
|
9
|
Zhang L, Wei F, Sun Q, Huang X, Zou Q, Jiang M, Su Y, Li S, Li X, Xie K, He J. FOXM1-Driven CKS1B Upregulation Promotes Pancreatic Cancer Progression and Therapeutic Resistance. Int J Biol Sci 2025; 21:1047-1064. [PMID: 39897042 PMCID: PMC11781179 DOI: 10.7150/ijbs.105289] [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: 10/16/2024] [Accepted: 11/25/2024] [Indexed: 02/04/2025] Open
Abstract
BACKGROUND Pancreatic ductal adenocarcinoma (PDAC) remains a highly lethal malignancy with limited treatment options. Investigating novel therapeutic targets and understanding mechanisms of chemoresistance are crucial for improving patient outcomes. This study investigated the role of CKS1B in PDAC carcinogenesis, stemness and chemoresistance, and explores the underlying mechanisms driving its upregulation. The findings may provide novel therapeutic insights and potential strategies for the treatment of PDAC. Methods: CKS1B expression was analyzed in PDAC tissues and cell lines, its impact on cell proliferation, migration, apoptosis, stemness and chemosensitivity were evaluated by using in vitro and in vivo models, and its underlying mechanistic connection to transcription factor FOXM1 was explored by using molecular biology methods. Results: CKS1B was significantly upregulated in PDAC tissues and correlated with poor patient survival. CKS1B promoted PDAC cell proliferation, migration, and inhibited apoptosis. Expression of CKS1B enhanced the stemness properties of pancreatic cancer. CKS1B knockdown sensitized PDAC cells to the treatment of gemcitabine and oxaliplatin. Mechanistically, CKS1B is transcriptionally regulated by FOXM1, establishing a novel FOXM1-CKS1B signaling axis that regulates carcinogenesis, proliferation, migration, stemness, apoptosis, and drug resistance in PDAC. Conclusions: Our findings strongly suggest that CKS1B plays a critical role in PDAC progression, stemness and chemoresistance. Targeting the FOXM1-CKS1B axis represents a promising therapeutic strategy for PDAC patients.
Collapse
Affiliation(s)
- Liuxi Zhang
- Guangzhou First People's Hospital and The Second Affiliated Hospital, South China University of Technology School of Medicine, #1 Panfu Road, Guangzhou, Guangdong 510180, P.R. China
- Center for Pancreatic Cancer Research, South China University of Technology College of Medicine, 382 Waihuan Road, Guangzhou, Guangdong 510006, P.R. China
| | - Fang Wei
- Guangzhou First People's Hospital and The Second Affiliated Hospital, South China University of Technology School of Medicine, #1 Panfu Road, Guangzhou, Guangdong 510180, P.R. China
- Center for Pancreatic Cancer Research, South China University of Technology College of Medicine, 382 Waihuan Road, Guangzhou, Guangdong 510006, P.R. China
| | - Qihui Sun
- Center for Pancreatic Cancer Research, South China University of Technology College of Medicine, 382 Waihuan Road, Guangzhou, Guangdong 510006, P.R. China
| | - Xinyan Huang
- Center for Pancreatic Cancer Research, South China University of Technology College of Medicine, 382 Waihuan Road, Guangzhou, Guangdong 510006, P.R. China
| | - Qi Zou
- Guangzhou First People's Hospital and The Second Affiliated Hospital, South China University of Technology School of Medicine, #1 Panfu Road, Guangzhou, Guangdong 510180, P.R. China
- Center for Pancreatic Cancer Research, South China University of Technology College of Medicine, 382 Waihuan Road, Guangzhou, Guangdong 510006, P.R. China
| | - Mengmeng Jiang
- Center for Pancreatic Cancer Research, South China University of Technology College of Medicine, 382 Waihuan Road, Guangzhou, Guangdong 510006, P.R. China
| | - Yuling Su
- Center for Pancreatic Cancer Research, South China University of Technology College of Medicine, 382 Waihuan Road, Guangzhou, Guangdong 510006, P.R. China
| | - Shu Li
- Center for Pancreatic Cancer Research, South China University of Technology College of Medicine, 382 Waihuan Road, Guangzhou, Guangdong 510006, P.R. China
| | - Xiaojia Li
- Center for Pancreatic Cancer Research, South China University of Technology College of Medicine, 382 Waihuan Road, Guangzhou, Guangdong 510006, P.R. China
| | - Keping Xie
- Center for Pancreatic Cancer Research, South China University of Technology College of Medicine, 382 Waihuan Road, Guangzhou, Guangdong 510006, P.R. China
| | - Jie He
- Guangzhou First People's Hospital and The Second Affiliated Hospital, South China University of Technology School of Medicine, #1 Panfu Road, Guangzhou, Guangdong 510180, P.R. China
- Center for Pancreatic Cancer Research, South China University of Technology College of Medicine, 382 Waihuan Road, Guangzhou, Guangdong 510006, P.R. China
| |
Collapse
|
10
|
Xu Y, Xue G, Zhou L, Wu G, Hu L, Ma S, Zhang J, Li X. KIF4A promotes epithelial-mesenchymal transition by activating the TGF-β/SMAD signaling pathway in glioma cells. Mol Cell Biochem 2025; 480:217-230. [PMID: 38411896 DOI: 10.1007/s11010-024-04943-z] [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: 09/09/2023] [Accepted: 01/14/2024] [Indexed: 02/28/2024]
Abstract
Gliomas are the most prevalent type of primary brain tumor, with poor prognosis reported in patients with high-grade glioma. Kinesin family member 4 A (KIF4A) stimulates the proliferation, migration, and invasion of tumor cells. However, its function in gliomas has not been clearly established. Therefore, this study aimed to investigate the effects of KIF4A on the epithelial-mesenchymal transition and invasion of glioma cells. We searched The Cancer Genome Atlas and Chinese Glioma Genome Atlas databases to identify KIF4A-related signaling pathways and downstream genes. We further validated them using western blotting, transwell migration and invasion, wound-healing scratch, and dual-luciferase reporter assays in U251 and U87 human glioblastoma cells. Our analysis of the Cancer Genome Atlas and Chinese Glioma Genome Atlas data showed elevated KIF4A expression in patients with gliomas and was associated with clinical grade. Here, KIF4A overexpression promoted the migration, invasion, and proliferation of glioma cells, whereas KIF4A knockdown showed contrasting results. Gene Ontology (GO) and Gene Set Enrichment Analysis (GSEA) analyses demonstrated that KIF4A positively controls TGF-β/SMAD signaling in glioma cells. Additionally, genetic correlation analysis revealed that KIF4A transcriptionally controls benzimidazoles-1 expression in glioma cells. KIF4A promotes the epithelial-mesenchymal transition by regulating the TGF-β/SMAD signaling pathway via benzimidazoles-1 in glioma cells.
Collapse
Affiliation(s)
- Yao Xu
- Department of Neurosurgery, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Guangren Xue
- Department of Neurosurgery, Dushu Lake Hospital Affiliated to Soochow University, Suzhou, China
| | - Lei Zhou
- Department of Neurosurgery, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Gaotian Wu
- Laboratory of Cancer Molecular Genetics, Soochow University, Medical College of Soochow University, Suzhou, China
| | - Lingji Hu
- Laboratory of Cancer Molecular Genetics, Soochow University, Medical College of Soochow University, Suzhou, China
| | - Shuchen Ma
- Department of Rehabilitation Medicine, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Jian Zhang
- Department of Neurosurgery, The First Affiliated Hospital of Soochow University, Suzhou, China.
| | - Xiangdong Li
- Department of Neurosurgery, The First Affiliated Hospital of Soochow University, Suzhou, China.
| |
Collapse
|
11
|
Lyu W, Qin H, Li Q, Lu D, Shi C, Zhao K, Zhang S, Yu R, Zhang H, Zhou X, Xia S, Zhang L, Wang X, Chi X, Liu Z. Novel mechanistic insights - A brand new Era for anti-HBV drugs. Eur J Med Chem 2024; 279:116854. [PMID: 39276582 DOI: 10.1016/j.ejmech.2024.116854] [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: 08/12/2024] [Revised: 09/03/2024] [Accepted: 09/04/2024] [Indexed: 09/17/2024]
Abstract
Hepatitis B Virus (HBV) remains a critical global health issue, with substantial morbidity and mortality. Current therapies, including interferons and nucleoside analogs, often fail to achieve complete cure or functional eradication. This review explores recent advances in anti-HBV agents, focusing on their innovative mechanisms of action. HBV entry inhibitors target the sodium taurocholate cotransporting polypeptide (NTCP) receptor, impeding viral entry, while nucleus translocation inhibitors disrupt key viral life cycle steps, preventing replication. Capsid assembly modulators inhibit covalently closed circular DNA (cccDNA) formation, aiming to eradicate the persistent viral reservoir. Transcription inhibitors targeting cccDNA and integrated DNA offer significant potential to suppress HBV replication. Immunomodulatory agents are highlighted for their ability to enhance host immune responses, facil-itating better control and possible eradication of HBV. These novel approaches represent significant advancements in HBV therapy, providing new strategies to overcome current treatment limitations. The development of cccDNA reducers is particularly critical, as they directly target the persistent viral reservoir, offering a promising pathway towards achieving a functional cure or complete viral eradication. Continued research in this area is essential to advance the effectiveness of anti-HBV therapies.
Collapse
Affiliation(s)
- Weiping Lyu
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, 100191, PR China
| | - Haoming Qin
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, 100191, PR China
| | - Qi Li
- Department of Medical Pharmacy, School of Basic Medicine, Qingdao University, Qingdao, 266071, Shandong, PR China
| | - Dehua Lu
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, 100191, PR China
| | - Cheng Shi
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, 100191, PR China
| | - Kangchen Zhao
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, 100191, PR China
| | - Shengran Zhang
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, 100191, PR China
| | - Ruohan Yu
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, 100191, PR China
| | - Huiying Zhang
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, 100191, PR China
| | - Xiaonan Zhou
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, 100191, PR China
| | - Sitian Xia
- Beijing National Day School, Beijing, 100089, PR China
| | - Liangren Zhang
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, 100191, PR China
| | - Xiaoqian Wang
- Beijing Tide Pharmaceutical Co., Ltd, No.8 East Rongjing Street, Beijing Economic-Technological Development Area (BDA), Beijing, 100176, PR China.
| | - Xiaowei Chi
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, 100191, PR China.
| | - Zhenming Liu
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, 100191, PR China.
| |
Collapse
|
12
|
Huang LH, Wu SC, Liu YW, Liu HT, Chien PC, Lin HP, Wu CJ, Hsieh TM, Hsieh CH. Identification of Crucial Cancer Stem Cell Genes Linked to Immune Cell Infiltration and Survival in Hepatocellular Carcinoma. Int J Mol Sci 2024; 25:11969. [PMID: 39596041 PMCID: PMC11593742 DOI: 10.3390/ijms252211969] [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: 09/07/2024] [Revised: 11/01/2024] [Accepted: 11/01/2024] [Indexed: 11/28/2024] Open
Abstract
Hepatocellular carcinoma is characterized by high recurrence rates and poor prognosis. Cancer stem cells contribute to tumor heterogeneity, treatment resistance, and recurrence. This study aims to identify key genes associated with stemness and immune cell infiltration in HCC. We analyzed RNA sequencing data from The Cancer Genome Atlas to calculate mRNA expression-based stemness index in HCC. A weighted gene co-expression network analysis was performed to identify stemness-related gene modules. A single-sample gene set enrichment analysis was used to evaluate immune cell infiltration. Key genes were validated using RT-qPCR. The mRNAsi was significantly higher in HCC tissues compared to adjacent normal tissues and correlated with poor overall survival. WGCNA and subsequent analyses identified 10 key genes, including minichromosome maintenance complex component 2, cell division cycle 6, forkhead box M1, NIMA-related kinase 2, Holliday junction recognition protein, DNA topoisomerase II alpha, denticleless E3 ubiquitin protein ligase homolog, maternal embryonic leucine zipper kinase, protein regulator of cytokinesis 1, and kinesin family member C1, associated with stemness and low immune cell infiltration. These genes were significantly upregulated in HCC tissues. A functional enrichment analysis revealed their involvement in cell cycle regulation. This study identified 10 key genes related to stemness and immune cell infiltration in HCC. These genes, primarily involved in cell cycle regulation, may serve as potential targets for developing more effective treatments to reduce HCC recurrence and improve patient outcomes.
Collapse
Affiliation(s)
- Lien-Hung Huang
- Department of Plastic Surgery, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung 833, Taiwan; (L.-H.H.); (P.-C.C.); (H.-P.L.); (C.-J.W.)
| | - Shao-Chun Wu
- Department of Anesthesiology, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung 833, Taiwan;
| | - Yueh-Wei Liu
- Department of General Surgery, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung 833, Taiwan;
| | - Hang-Tsung Liu
- Department of Trauma Surgery, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung 833, Taiwan;
| | - Peng-Chen Chien
- Department of Plastic Surgery, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung 833, Taiwan; (L.-H.H.); (P.-C.C.); (H.-P.L.); (C.-J.W.)
| | - Hui-Ping Lin
- Department of Plastic Surgery, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung 833, Taiwan; (L.-H.H.); (P.-C.C.); (H.-P.L.); (C.-J.W.)
| | - Chia-Jung Wu
- Department of Plastic Surgery, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung 833, Taiwan; (L.-H.H.); (P.-C.C.); (H.-P.L.); (C.-J.W.)
| | - Ting-Min Hsieh
- Department of Trauma Surgery, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung 833, Taiwan;
| | - Ching-Hua Hsieh
- Department of Plastic Surgery, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung 833, Taiwan; (L.-H.H.); (P.-C.C.); (H.-P.L.); (C.-J.W.)
| |
Collapse
|
13
|
Li LS, Chen PW, Zhao XJ, Cheng D, Liu BB, Tang XJ, Zhu WQ, Yang X, Zhao MX. Nuclear-targeted smart nanoplatforms featuring double-shell hollow mesoporous copper sulfide coated with manganese dioxide synergistically potentiate chemotherapy and immunotherapy in hepatocellular carcinoma cells. J Colloid Interface Sci 2024; 680:202-214. [PMID: 39504750 DOI: 10.1016/j.jcis.2024.11.003] [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: 09/06/2024] [Revised: 10/25/2024] [Accepted: 11/01/2024] [Indexed: 11/08/2024]
Abstract
Smart nanoplatforms designed for nuclear-targeted delivery of chemotherapeutic agents to tumor sites are pivotal in advancing tumor treatment and immunotherapy. Herein, we introduced a novel nuclear-targeting double-shell smart nanoplatform (HMCuS/Pt/ICG@MnO2@9R-P201 (HMCPIM9P)), which synergistically enhances chemotherapy, photodynamic therapy (PDT), photothermal therapy (PTT), immunotherapy and chemodynamic therapy (CDT). The core of this nanoplatform consists of double-shell multifunctional nanoparticles (HMCuS@MnO2) that enable targeted delivery of the photosensitizer Indocyanine Green (ICG) and the chemotherapeutic agent cisplatin (Pt). By effectively consuming glutathione (GSH), these nanoparticles boost the chemotherapeutic efficacy of Pt. Additionally, the manganese ion (Mn2+) present activate the cyclic GMP-AMP synthase (cGAS)-stimulator of interferon genes (STING) (cGAS-STING) pathway, bolstering adaptive immune responses against tumors and elevating the level of tumor-infiltrating CD8+ T cells. The incorporation of the hepatoma-targeting peptide (9R-P201 peptide) allows the system to exhibit FOXM1 receptor-mediated nuclear targeting properties specifically in hepatocellular carcinoma (HCC). Notably, when combined with near-infrared (NIR) light, HMCPIM9P demonstrated a remarkable tumor inhibition rate of 95.6 %, fostered a robust immune response, and significantly inhibited tumor growth and recurrence. Overall, the smart nanoplatform boasts active nuclear targeting capabilities, enabling the enrichment of chemotherapeutic agents at tumor sites, and holds great potential for synergistic applications in enhancing chemotherapy and immunotherapy for HCC.
Collapse
Affiliation(s)
- Lin-Song Li
- Henan Key Laboratory of Natural Medicine Innovation and Transformation, Henan University, Kaifeng 475004, China; State Key Laboratory of Antiviral Drugs, Henan University, Kaifeng 475004, China
| | - Peng-Wei Chen
- Henan Key Laboratory of Natural Medicine Innovation and Transformation, Henan University, Kaifeng 475004, China; State Key Laboratory of Antiviral Drugs, Henan University, Kaifeng 475004, China
| | - Xue-Jie Zhao
- Henan Key Laboratory of Natural Medicine Innovation and Transformation, Henan University, Kaifeng 475004, China; State Key Laboratory of Antiviral Drugs, Henan University, Kaifeng 475004, China
| | - Dong Cheng
- Henan Key Laboratory of Natural Medicine Innovation and Transformation, Henan University, Kaifeng 475004, China; State Key Laboratory of Antiviral Drugs, Henan University, Kaifeng 475004, China
| | - Bang-Bang Liu
- Henan Key Laboratory of Natural Medicine Innovation and Transformation, Henan University, Kaifeng 475004, China; State Key Laboratory of Antiviral Drugs, Henan University, Kaifeng 475004, China
| | - Xian-Jiao Tang
- Henan Key Laboratory of Natural Medicine Innovation and Transformation, Henan University, Kaifeng 475004, China; State Key Laboratory of Antiviral Drugs, Henan University, Kaifeng 475004, China
| | - Wen-Qi Zhu
- Henan Key Laboratory of Natural Medicine Innovation and Transformation, Henan University, Kaifeng 475004, China; State Key Laboratory of Antiviral Drugs, Henan University, Kaifeng 475004, China
| | - Xiaojing Yang
- Henan Key Laboratory of Natural Medicine Innovation and Transformation, Henan University, Kaifeng 475004, China; State Key Laboratory of Antiviral Drugs, Henan University, Kaifeng 475004, China
| | - Mei-Xia Zhao
- Henan Key Laboratory of Natural Medicine Innovation and Transformation, Henan University, Kaifeng 475004, China; State Key Laboratory of Antiviral Drugs, Henan University, Kaifeng 475004, China; The Zhongzhou Laboratory for Integrative Biology, Henan University, Kaifeng, China.
| |
Collapse
|
14
|
Yu W, Cai X, Wang C, Peng X, Xu L, Gao Y, Tian T, Zhu G, Pan Y, Chu H, Liang S, Chen C, Kim NH, Yuan B, Zhang J, Jiang H. FOXM1 affects oxidative stress, mitochondrial function, and the DNA damage response by regulating p21 in aging oocytes. Theriogenology 2024; 229:66-74. [PMID: 39163804 DOI: 10.1016/j.theriogenology.2024.08.010] [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: 05/30/2024] [Revised: 08/08/2024] [Accepted: 08/09/2024] [Indexed: 08/22/2024]
Abstract
Fertilization capacity and embryo survival rate are decreased in postovulatory aging oocytes, which results in a reduced reproductive rate in female animals. However, the key regulatory genes and related regulatory mechanisms involved in the process of postovulatory aging in oocytes remain unclear. In this study, RNA-Seq revealed that 3237 genes were differentially expressed in porcine oocytes between the MII and aging stages (MII + 24 h). The expression level of FOXM1 was increased at the aging stage, and FOXM1 was also observed to be enriched in many key biological processes, such as cell senescence, response to oxidative stress, and transcription, during porcine oocyte aging. Previous studies have shown that FOXM1 is involved in the regulation of various biological processes, such as oxidative stress, DNA damage repair, mitochondrial function, and cellular senescence, which suggests that FOXM1 may play a crucial role in the process of postovulatory aging. Therefore, in this study, we investigated the effects and mechanisms of FOXM1 on oxidative stress, mitochondrial function, DNA damage, and apoptosis during oocyte aging. Our study revealed that aging oocytes exhibited significantly increased ROS levels and significantly decreased GSH, SOD, T-AOC, and CAT levels than did oocytes at the MII stage and that FOXM1 inhibition exacerbated the changes in these levels in aging oocytes. In addition, FOXM1 inhibition increased the levels of DNA damage, apoptosis, and cell senescence in aging oocytes. A p21 inhibitor alleviated the effects of FOXM1 inhibition on oxidative stress, mitochondrial function, and DNA damage and thus alleviated the degree of senescence in aging oocytes. These results indicate that FOXM1 plays a crucial role in porcine oocyte aging. This study contributes to the understanding of the function and mechanism of FOXM1 during porcine oocyte aging and provides a theoretical basis for preventing oocyte aging and optimizing conditions for the in vitro culture of oocytes.
Collapse
Affiliation(s)
- Wenjie Yu
- College of Animal Sciences, Jilin University, Changchun, 130062, Jilin, China
| | - Xiaoshi Cai
- College of Animal Sciences, Jilin University, Changchun, 130062, Jilin, China
| | - Chen Wang
- College of Animal Sciences, Jilin University, Changchun, 130062, Jilin, China
| | - Xinyue Peng
- College of Animal Sciences, Jilin University, Changchun, 130062, Jilin, China
| | - Lingxia Xu
- College of Animal Sciences, Jilin University, Changchun, 130062, Jilin, China
| | - Yan Gao
- College of Animal Sciences, Jilin University, Changchun, 130062, Jilin, China
| | - Tian Tian
- College of Animal Sciences, Jilin University, Changchun, 130062, Jilin, China; Center of Reproductive Medicine & Center of Prenatal Diagnosis, First Hospital, Jilin University, Changchun, 130062, Jilin, China
| | - Guangying Zhu
- Department of Mental Health, First Hospital, Jilin University, Changchun, 130062, Jilin, China
| | - Yuan Pan
- Center of Reproductive Medicine & Center of Prenatal Diagnosis, First Hospital, Jilin University, Changchun, 130062, Jilin, China
| | - Hongzhong Chu
- General Animal Husbandry Center of Ili Kazakh Autonomous Prefecture, Yining, 835000, China
| | - Shuang Liang
- College of Animal Sciences, Jilin University, Changchun, 130062, Jilin, China
| | - Chengzhen Chen
- College of Animal Sciences, Jilin University, Changchun, 130062, Jilin, China
| | - Nam-Hyung Kim
- Guangdong Provincial Key Laboratory of Large Animal Models for Biomedicine, South China Institute of Large Animal Models for Biomedicine, School of Pharmacy and Food Engineering, Wuyi University, Jiangmen, 529000, China
| | - Bao Yuan
- College of Animal Sciences, Jilin University, Changchun, 130062, Jilin, China
| | - Jiabao Zhang
- College of Animal Sciences, Jilin University, Changchun, 130062, Jilin, China
| | - Hao Jiang
- College of Animal Sciences, Jilin University, Changchun, 130062, Jilin, China.
| |
Collapse
|
15
|
Natarajan SR, Krishnamoorthy R, Alshuniaber MA, Al-Anazi KM, Farah MA, Rajagopal P, Palanisamy CP, Veeraraghavan VP, Jayaraman S. Identification of FOXM1 as a novel protein biomarker and therapeutic target for colorectal cancer progression: Evidence from immune infiltration and bioinformatic analyses. Int J Biol Macromol 2024; 282:137201. [PMID: 39489237 DOI: 10.1016/j.ijbiomac.2024.137201] [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: 09/27/2024] [Revised: 10/23/2024] [Accepted: 10/31/2024] [Indexed: 11/05/2024]
Abstract
Colorectal cancer (CRC) remains a major global health challenge, with its underlying molecular mechanisms, particularly the role of FOXM1, not yet fully understood. This study employed an integrated approach combining bioinformatics along with experimental validation to explore the role of FOXM1 in CRC. Using advanced computational tools and experimental techniques, we aimed to clarify the biological significance of FOXM1 and its potential impact on CRC progression and treatment. Bioinformatic analyses, including pan-cancer views, mRNA expression analysis, immune infiltrations, pathway enrichment, and functional annotations, highlighted the oncogenic potential of FOXM1 in CRC. Protein and gene expression analyses (western blot and qPCR) were conducted in HCT-116 and HT-29 cells. Platforms like GEPIA and UALCAN confirmed the diagnostic relevance of FOXM1, showing upregulated mRNA expression across various stages and metastasis. The influence of FOXM1 on immune cells, particularly CD4+, CD8+, and B cells, was significant, as revealed by immunohistochemistry. Protein-protein interaction analysis through STRING and CYTOSCAPE identified genes closely linked to FOXM1 in CRC. KEGG pathway enrichment suggested FOXM1's involvement in the p53 pathway, reinforcing its role in oncogenesis. Experimental validation confirmed elevated FOXM1 expression in HCT-116 and HT-29 cells. In summary, this study indicates that targeting FOXM1 could be a promising therapeutic strategy in CRC, emphasizing its importance in the molecular landscape of cancer progression.
Collapse
Affiliation(s)
- Sathan Raj Natarajan
- Centre of Molecular Medicine and Diagnostics (COMManD), Department of Biochemistry, Saveetha Dental College & Hospitals, Saveetha Institute of Medical & Technical Sciences, Saveetha University, Chennai 600077, India.
| | - Rajapandiyan Krishnamoorthy
- Department of Food Science and Nutrition, College of Food and Agriculture Sciences, King Saud University, Riyadh 11451, Saudi Arabia.
| | - Mohammad A Alshuniaber
- Department of Food Science and Nutrition, College of Food and Agriculture Sciences, King Saud University, Riyadh 11451, Saudi Arabia.
| | - Khalid Mashay Al-Anazi
- Department of Zoology, College of Science, King Saud University, Riyadh 11451, Saudi Arabia.
| | - Mohammad Abul Farah
- Department of Zoology, College of Science, King Saud University, Riyadh 11451, Saudi Arabia.
| | - Ponnulakshmi Rajagopal
- Central Research Laboratory, Meenakshi Ammal Dental College and Hospital, Meenakshi Academy of Higher Education and Research (Deemed to be University), Chennai 600095, India.
| | - Chella Perumal Palanisamy
- Department of Chemical Technology, Faculty of Science, Chulalongkorn University, Bangkok 10330, Thailand.
| | - Vishnu Priya Veeraraghavan
- Centre of Molecular Medicine and Diagnostics (COMManD), Department of Biochemistry, Saveetha Dental College & Hospitals, Saveetha Institute of Medical & Technical Sciences, Saveetha University, Chennai 600077, India.
| | - Selvaraj Jayaraman
- Centre of Molecular Medicine and Diagnostics (COMManD), Department of Biochemistry, Saveetha Dental College & Hospitals, Saveetha Institute of Medical & Technical Sciences, Saveetha University, Chennai 600077, India.
| |
Collapse
|
16
|
Bao Z, Yang M, Guo Y, Ge Q, Zhang H. MTFR2 accelerates hepatocellular carcinoma mediated by metabolic reprogramming via the Akt signaling pathway. Cell Signal 2024; 123:111366. [PMID: 39182591 DOI: 10.1016/j.cellsig.2024.111366] [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: 04/28/2024] [Revised: 08/13/2024] [Accepted: 08/22/2024] [Indexed: 08/27/2024]
Abstract
Metabolic reprogramming has recently been identified as a hallmark of malignancies. The shift from oxidative phosphorylation to glycolysis in hepatocellular carcinoma (HCC) meets the demands of rapid cell growth and provides a microenvironment for tumor progression. This study sought to uncover the function and mechanism of MTFR2 in the metabolic reprogramming of HCC. Elevated MTFR2 expression was associated with poor patient prognosis. Downregulation of MTFR2 blocked malignant behaviors, epithelial-to-mesenchymal transition (EMT), and glycolysis in HCC cells. Nuclear transcription factor Y subunit gamma (NFYC) was also associated with poor patient prognosis, and NFYC bound to the promoter of MTFR2 to activate transcription and promote Akt signaling. The repressive effects of NFYC knockdown on EMT and glycolysis in HCC cells were compromised by MTFR2 overexpression, elicited through the activation of the Akt signaling. Knockdown of NFYC slowed the growth and intrahepatic metastasis in vivo, which was reversed by MTFR2 overexpression. In conclusion, our work shows that activation of MTFR2 by the transcription factor NFYC promotes Akt signaling, thereby potentiating metabolic reprogramming in HCC development. Targeting the NFYC/MTFR2/Akt axis may represent a therapeutic strategy for HCC.
Collapse
Affiliation(s)
- Zhongming Bao
- Department of Hepatobiliary Surgery, Huai'an Hospital Affiliated to Yangzhou University (The Fifth People's Hospital of Huai'an), Huaiyin 223300, Jiangsu, PR China
| | - Ming Yang
- Department of Hepatobiliary Surgery, Huai'an Hospital Affiliated to Yangzhou University (The Fifth People's Hospital of Huai'an), Huaiyin 223300, Jiangsu, PR China
| | - Yunhu Guo
- Department of Hepatobiliary Surgery, Huai'an Hospital Affiliated to Yangzhou University (The Fifth People's Hospital of Huai'an), Huaiyin 223300, Jiangsu, PR China
| | - Qi Ge
- School of Biological Science and Food Engineering, Chuzhou University, Chuzhou 239000, Anhui, PR China.
| | - Huaguo Zhang
- Department of Hepatobiliary Surgery, Huai'an Hospital Affiliated to Yangzhou University (The Fifth People's Hospital of Huai'an), Huaiyin 223300, Jiangsu, PR China.
| |
Collapse
|
17
|
Ghnim ZS, Mahdi MS, Ballal S, Chahar M, Verma R, Al-Nuaimi AMA, Kumar MR, Al-Hussein RKA, Adil M, Jawad MJ. The role of kinesin superfamily proteins in hepatocellular carcinoma. Med Oncol 2024; 41:271. [PMID: 39400594 DOI: 10.1007/s12032-024-02497-0] [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: 07/29/2024] [Accepted: 09/04/2024] [Indexed: 10/15/2024]
Abstract
The most prevalent form of primary liver cancer, hepatocellular carcinoma (HCC) poses a significant global health challenge due to its limited therapeutic options. Researchers are currently focused on the complex molecular landscape that governs the initiation and progression of HCC in order to identify new avenues for diagnosis, prognosis, and treatment. In the context of HCC, the Kinesin Superfamily Proteins (KIFs) have become critical regulators of cellular processes, prompting a growing interest in their function among the diverse array of molecular actors implicated in cancer. The KIFs, a family of microtubule-based molecular motors, are renowned for their essential roles in the dynamics of mitotic spindles and intracellular transport. Beyond their well-established functions in normal cellular physiology, emerging evidence indicates that dysregulation of KIFs significantly contributes to the pathogenesis of HCC. Novel therapeutic targets and diagnostic markers are revealed through the unique opportunity to comprehend the complex interplay between KIFs and the molecular events that drive HCC.
Collapse
Affiliation(s)
| | | | - Suhas Ballal
- Department of Chemistry and Biochemistry, School of Sciences, JAIN (Deemed to Be University), Bangalore, Karnataka, India.
| | - Mamata Chahar
- Department of Chemistry, NIMS Institute of Engineering & Technology, NIMS University, Jaipur, Rajasthan, India
| | - Rajni Verma
- Department of Applied Sciences, Chandigarh Engineering College, Chandigarh Group of Colleges, Jhanjeri, Mohali, Amritsar, Punjab, 140307, India
| | | | - M Ravi Kumar
- Department of Chemistry, Raghu Engineering College, Visakhapatnam, Andhra Pradesh, 531162, India
| | | | - Mohaned Adil
- Pharmacy College, Al-Farahidi University, Baghdad, Iraq
| | | |
Collapse
|
18
|
Zhao Z, Cheng J, Hou Q, Zhu J, Chen T, Lu S, Wu G, Lv H, Wu X. Role of FOXM1 and AURKB in regulating keratinocyte function in psoriasis. Open Med (Wars) 2024; 19:20241049. [PMID: 39381423 PMCID: PMC11459273 DOI: 10.1515/med-2024-1049] [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: 11/14/2023] [Revised: 09/04/2024] [Accepted: 09/05/2024] [Indexed: 10/10/2024] Open
Abstract
Objective This study investigated the effect of forkhead box M1 (FOXM1) and Aurora kinase B (AURKB) on the epidermal function of keratinocytes. Methods Bioinformatics analysis was used to analyze the co-expression network of FOXM1 and its correlation with AURKB. The expression of FOXM1 and AURKB in tissues and cells was detected by immunofluorescence and real-time quantitative polymerase chain reaction, respectively. HaCaT cells were transfected with si-FOXM1 to knock down FOXM1. Cell proliferation was detected by cell counting kit-8 assay. Cell migration was detected by scratch assay. Cell invasion was detected by the Transwell invasion assay. Cell apoptosis and cell cycle were detected by flow cytometry. Results FOXM1 and AURKB were positively correlated and highly expressed in psoriatic lesions. After transfection of si-FOXM1, the expression levels of FOXM1 and AURKB genes significantly decreased. The proliferation of HaCaT cells decreased, the apoptosis rate increased significantly, and the proportion of cells in the G1 phase increased significantly, while the proportion of cells in the S phase decreased significantly. The scratch closure of HaCaT cells was reduced, and the number of cell invasions decreased significantly. Conclusion FOXM1 and AURKB may affect the progression of psoriasis by regulating the proliferation, cell cycle, migration, and invasion of keratinocytes.
Collapse
Affiliation(s)
- Zhaofeng Zhao
- Central Laboratory, Shanghai Xuhui Central Hospital, Zhongshan-Xuhui Hospital, Fudan University, Shanghai, 200031, P.R. China
| | - Jie Cheng
- Department of Urology, Shanghai Xuhui Central Hospital, Zhongshan-Xuhui Hospital, Fudan University, Shanghai, 200031, P.R. China
| | - Qiang Hou
- Department of Dermatology, Xuhui District Dahua Hospital,
Shanghai, 200237, P.R. China
| | - Jian Zhu
- Department of Dermatology, Shanghai Xuhui Central Hospital, Zhongshan-Xuhui Hospital, Fudan University, Shanghai, 200031, P.R. China
| | - Tu Chen
- Department of Dermatology, Changqiao Street Community Health Service Center,
Shanghai, 200231, P.R. China
| | - Sheng Lu
- Department of Dermatology, Shanghai Xuhui Central Hospital, Zhongshan-Xuhui Hospital, Fudan University, Shanghai, 200031, P.R. China
| | - Guiju Wu
- Department of Dermatology, Xuhui District Dahua Hospital,
Shanghai, 200237, P.R. China
| | - Hongli Lv
- Department of Dermatology, Jia Ding Central Hospital,
No. 01, Dingcheng Road, Jiading District, Shanghai, 201899, P.R. China
| | - Xiujuan Wu
- Department of Dermatology, Shanghai Xuhui Central Hospital, Zhongshan-Xuhui Hospital, Fudan University, No. 366, Longchuan North Road, Xuhui District, Shanghai, 200031, P.R. China
| |
Collapse
|
19
|
Yang B, Lu L, Xiong T, Fan W, Wang J, Barbier-Torres L, Chhimwal J, Sinha S, Tsuchiya T, Mavila N, Tomasi ML, Cao D, Zhang J, Peng H, Mato JM, Liu T, Yang X, Kalinichenko VV, Ramani K, Han J, Seki E, Yang H, Lu SC. The role of forkhead box M1-methionine adenosyltransferase 2 A/2B axis in liver inflammation and fibrosis. Nat Commun 2024; 15:8388. [PMID: 39333125 PMCID: PMC11436801 DOI: 10.1038/s41467-024-52527-8] [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: 09/22/2023] [Accepted: 09/09/2024] [Indexed: 09/29/2024] Open
Abstract
Methionine adenosyltransferase 2 A (MAT2A) and MAT2B are essential for hepatic stellate cells (HSCs) activation. Forkhead box M1 (FOXM1) transgenic mice develop liver inflammation and fibrosis. Here we examine if they crosstalk in male mice. We found FOXM1/MAT2A/2B are upregulated after bile duct ligation (BDL) and carbon tetrachloride (CCl4) treatment in hepatocytes, HSCs and Kupffer cells (KCs). FDI-6, a FOXM1 inhibitor, attenuates the development and reverses the progression of CCl4-induced fibrosis while lowering the expression of FOXM1/MAT2A/2B, which exert reciprocal positive regulation on each other transcriptionally. Knocking down any of them lowers HSCs and KCs activation. Deletion of FOXM1 in hepatocytes, HSCs, and KCs protects from BDL-mediated inflammation and fibrosis comparably. Interestingly, HSCs from Foxm1Hep-/-, hepatocytes from Foxm1HSC-/-, and HSCs and hepatocytes from Foxm1KC-/- have lower FOXM1/MAT2A/2B after BDL. This may be partly due to transfer of extracellular vesicles between different cell types. Altogether, FOXM1/MAT2A/MAT2B axis drives liver inflammation and fibrosis.
Collapse
Affiliation(s)
- Bing Yang
- Karsh Division of Gastroenterology and Hepatology, Department of Medicine, Cedars-Sinai Medical Center, LA, Los Angeles, CA, 90048, USA
- Department of Geriatric Endocrinology and Metabolism, Guangxi Key Laboratory of Precision Medicine in Cardio-cerebrovascular Diseases Control and Prevention, Guangxi Clinical Research Center for Cardio-cerebrovascular Diseases, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, 530021, China
| | - Liqing Lu
- Karsh Division of Gastroenterology and Hepatology, Department of Medicine, Cedars-Sinai Medical Center, LA, Los Angeles, CA, 90048, USA
- Key Laboratory of Cancer Proteomics of Chinese Ministry of Health, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China
| | - Ting Xiong
- Karsh Division of Gastroenterology and Hepatology, Department of Medicine, Cedars-Sinai Medical Center, LA, Los Angeles, CA, 90048, USA
- Department of Pharmacy, The Third Hospital of Changsha, Changsha, Hunan, 410015, China
| | - Wei Fan
- Karsh Division of Gastroenterology and Hepatology, Department of Medicine, Cedars-Sinai Medical Center, LA, Los Angeles, CA, 90048, USA
| | - Jiaohong Wang
- Karsh Division of Gastroenterology and Hepatology, Department of Medicine, Cedars-Sinai Medical Center, LA, Los Angeles, CA, 90048, USA
| | - Lucía Barbier-Torres
- Karsh Division of Gastroenterology and Hepatology, Department of Medicine, Cedars-Sinai Medical Center, LA, Los Angeles, CA, 90048, USA
| | - Jyoti Chhimwal
- Karsh Division of Gastroenterology and Hepatology, Department of Medicine, Cedars-Sinai Medical Center, LA, Los Angeles, CA, 90048, USA
| | - Sonal Sinha
- Karsh Division of Gastroenterology and Hepatology, Department of Medicine, Cedars-Sinai Medical Center, LA, Los Angeles, CA, 90048, USA
| | - Takashi Tsuchiya
- Karsh Division of Gastroenterology and Hepatology, Department of Medicine, Cedars-Sinai Medical Center, LA, Los Angeles, CA, 90048, USA
| | - Nirmala Mavila
- Karsh Division of Gastroenterology and Hepatology, Department of Medicine, Cedars-Sinai Medical Center, LA, Los Angeles, CA, 90048, USA
| | - Maria Lauda Tomasi
- Karsh Division of Gastroenterology and Hepatology, Department of Medicine, Cedars-Sinai Medical Center, LA, Los Angeles, CA, 90048, USA
| | - DuoYao Cao
- Department of Biomedical Sciences, CSMC LA, Los Angeles, CA, 90048, USA
| | - Jing Zhang
- Karsh Division of Gastroenterology and Hepatology, Department of Medicine, Cedars-Sinai Medical Center, LA, Los Angeles, CA, 90048, USA
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Hui Peng
- Karsh Division of Gastroenterology and Hepatology, Department of Medicine, Cedars-Sinai Medical Center, LA, Los Angeles, CA, 90048, USA
| | - José M Mato
- CIC bioGUNE, Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (Ciberehd), Technology, Park of Bizkaia, 48120, Derio, Bizkaia, Spain
| | - Ting Liu
- Department of Gastroenterology, Xiangya Hospital, Key Laboratory of Cancer proteomics of Chinese Ministry of Health, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China
| | - Xi Yang
- Department of Geriatric Endocrinology and Metabolism, Guangxi Key Laboratory of Precision Medicine in Cardio-cerebrovascular Diseases Control and Prevention, Guangxi Clinical Research Center for Cardio-cerebrovascular Diseases, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, 530021, China
| | - Vladimir V Kalinichenko
- Phoenix Children's Research Institute, Department of Child Health, University of Arizona College of Medicine, Phoenix, AZ, 85004, USA
- Division of Neonatology, Phoenix Children's Hospital, Phoenix, AZ, 85016, USA
| | - Komal Ramani
- Karsh Division of Gastroenterology and Hepatology, Department of Medicine, Cedars-Sinai Medical Center, LA, Los Angeles, CA, 90048, USA
| | - Jenny Han
- Karsh Division of Gastroenterology and Hepatology, Department of Medicine, Cedars-Sinai Medical Center, LA, Los Angeles, CA, 90048, USA
- Department of Society and Genetics, UCLA LA, Los Angeles, CA, 92620, USA
| | - Ekihiro Seki
- Karsh Division of Gastroenterology and Hepatology, Department of Medicine, Cedars-Sinai Medical Center, LA, Los Angeles, CA, 90048, USA
| | - Heping Yang
- Karsh Division of Gastroenterology and Hepatology, Department of Medicine, Cedars-Sinai Medical Center, LA, Los Angeles, CA, 90048, USA.
| | - Shelly C Lu
- Karsh Division of Gastroenterology and Hepatology, Department of Medicine, Cedars-Sinai Medical Center, LA, Los Angeles, CA, 90048, USA.
| |
Collapse
|
20
|
Jiang Y, Zhu H, Wang T, Tong H, Liu J, Yang Y, Zhou X, Liu X. Hypermethylation and low expression of FOXM1 predisposes women to unexplained recurrent miscarriage by impairing trophoblast stem cell proliferation. Cell Signal 2024; 121:111259. [PMID: 38871040 DOI: 10.1016/j.cellsig.2024.111259] [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: 03/29/2024] [Revised: 05/26/2024] [Accepted: 06/10/2024] [Indexed: 06/15/2024]
Abstract
Recurrent miscarriage (RM) is a distressing pregnancy complication with an unknown etiology. Increasing evidence indicates the relevance of dysregulation of human trophoblast stem cells (hTSCs), which may play a role in the development of RM. However, the potential molecular regulatory mechanism underlying the initiation and maintenance of hTSCs is yet to be fully elucidated. In this study, we performed data analysis and identified Forkhead box M1 (FOXM1) as a potential factor associated with RM. FOXM1 is a typical transcription factor known for its involvement in various pathophysiological processes, while the precise function of FOXM1 functions in hTSCs and RM remains incompletely understood. Utilizing RNA-seq, CUT&Tag, ChIP-qPCR, and sodium bisulfite conversion methods for methylation analysis, we elucidate the underlying regulatory mechanisms of FOXM1 in hTSCs and its implications in RM. Our findings demonstrate the relative high expression of FOXM1 in proliferating cytotrophoblasts (CTBs) compared to differentiated extravillous cytotrophoblasts (EVTs) and syncytiotrophoblasts (STBs). Besides, we provide evidence supporting a significant correlation between FOXM1 downregulation and the incidence of RM. Furthermore, we demonstrate the significant role of FOXM1 in regulating hTSCs proliferation and cell cycle through the transcriptional regulation of CDKN3, CCNB2, CCNA2, MAD2L1 and CDC25C. Notably, we observed a correlation between the downregulation of FOXM1 in RM and hypermethylation in its promoter region. Collectively, these results provide insights into the impact of FOXM1 on trophoblast regulation and offer a novel perspective on RM.
Collapse
Affiliation(s)
- Youqing Jiang
- Reproductive Medicine Center, The First Affiliated Hospital of Chongqing Medical University, No. 1 Youyi Road, Yuzhong District, Chongqing 400016, China.; Chongqing Key Laboratory of Maternal and Fetal Medicine, Chongqing Medical University, Chongqing 400016, China
| | - Huimin Zhu
- Reproductive Medicine Center, The First Affiliated Hospital of Chongqing Medical University, No. 1 Youyi Road, Yuzhong District, Chongqing 400016, China
| | - Tingting Wang
- Department of Obstetrics, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Hai Tong
- Department of Obstetrics, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Jinkai Liu
- Department of Obstetrics, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Yi Yang
- Women and Children's Hospital of Chongqing Medical University, No. 120 Longshan Road, Yubei District, Chongqing 401147, China
| | - Xiaobo Zhou
- Reproductive Medicine Center, The First Affiliated Hospital of Chongqing Medical University, No. 1 Youyi Road, Yuzhong District, Chongqing 400016, China..
| | - Xiru Liu
- Reproductive Medicine Center, The First Affiliated Hospital of Chongqing Medical University, No. 1 Youyi Road, Yuzhong District, Chongqing 400016, China..
| |
Collapse
|
21
|
Wei T, Zeng C, Li Q, Xiao Z, Zhang L, Zhang Q, Ren L. FOXM1/DEPDC1 feedback loop promotes hepatocarcinogenesis and represents promising targets for cancer therapy. Cancer Sci 2024; 115:3041-3053. [PMID: 39004911 PMCID: PMC11463088 DOI: 10.1111/cas.16273] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2024] [Revised: 06/24/2024] [Accepted: 06/26/2024] [Indexed: 07/16/2024] Open
Abstract
Forkhead box M1 (FOXM1) is a key regulator of mitosis and is identified as an oncogene involved in several kinds of human malignancies. However, how it induces carcinogenesis and related therapeutic approaches remains not fully understood. In this study, we aimed to identify a regulatory axis involving FOXM1 and its target gene DEP domain containing 1 (DEPDC1) and investigate their biological functions. FOXM1 bound to the promoter and transcriptionally induced DEPDC1 expression, in turn, DEPDC1 physically interacted with FOXM1, promoted its nuclear translocation, and reinforced its transcriptional activities. The FOXM1/DEPDC1 axis was indispensable for cancer cells, as evidenced by the fact that DEPDC1 rescued cell growth inhibition caused by FOXM1 knockdown, and silencing DEPDC1 efficiently attenuated tumor growth in a murine hepatocellular carcinoma model. Furthermore, strong positive associations between FOXM1/DEPDC1 axis and poor clinical outcome were observed in human hepatocellular carcinoma samples, further indicating their significance for hepatocarcinogenesis. Finally, we attempted to exploit immunotherapy approaches to target the FOXM1/DEPDC1 axis. Several HLA-A24:02-restricted T-cell epitopes targeting FOXM1 or DEPDC1 were identified through bioinformatic analysis. Then, T cell receptor (TCR)-engineered T cells targeting FOXM1262-270 or DEPDC1294-302 were successfully established and proved to efficiently eradicate tumor cells. Our findings highlight the significance of the FOXM1/DEPDC1 axis in the process of oncogenesis and indicate their potential as immunotherapy targets.
Collapse
Affiliation(s)
- Teng Wei
- Cytotherapy LaboratoryShenzhen People's Hospital (The Second Clinical Medical College, Jinan University, The First Affiliated Hospital, Southern University of Science and Technology)ShenzhenGuangdongChina
| | - Chenquan Zeng
- Cytotherapy LaboratoryShenzhen People's Hospital (The Second Clinical Medical College, Jinan University, The First Affiliated Hospital, Southern University of Science and Technology)ShenzhenGuangdongChina
| | - Qineng Li
- Cytotherapy LaboratoryShenzhen People's Hospital (The Second Clinical Medical College, Jinan University, The First Affiliated Hospital, Southern University of Science and Technology)ShenzhenGuangdongChina
| | - Zhiyuan Xiao
- Department of PathologyShenzhen People's Hospital (The Second Clinical Medical College, Jinan University, The First Affiliated Hospital, Southern University of Science and Technology)ShenzhenGuangdongChina
| | - Leisheng Zhang
- Jinan Key Laboratory of Medical Cell Bioengineering, Science and Technology Innovation Center, The Fourth People's Hospital of JinanThe Teaching Hospital of Shandong First Medical UniversityJinanChina
- National Health Commission (NHC) Key Laboratory of Diagnosis and Therapy of Gastrointestinal TumorGansu Provincial HospitalLanzhouChina
| | - Qiangnu Zhang
- Biomedicine Research CenterThird Affiliated Hospital of Guangzhou Medical UniversityGuangzhouChina
| | - Lili Ren
- Cytotherapy LaboratoryShenzhen People's Hospital (The Second Clinical Medical College, Jinan University, The First Affiliated Hospital, Southern University of Science and Technology)ShenzhenGuangdongChina
| |
Collapse
|
22
|
Li M, Gao X, Su Y, Shan S, Qian W, Zhang Z, Zhu D. FOXM1 transcriptional regulation. Biol Cell 2024; 116:e2400012. [PMID: 38963053 DOI: 10.1111/boc.202400012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Revised: 05/09/2024] [Accepted: 05/13/2024] [Indexed: 07/05/2024]
Abstract
FOXM1 is a key transcriptional regulator involved in various biological processes in mammals, including carbohydrate and lipid metabolism, aging, immune regulation, development, and disease. Early studies have shown that FOXM1 acts as an oncogene by regulating cell proliferation, cell cycle, migration, metastasis, and apoptosis, as well as genes related to diagnosis, treatment, chemotherapy resistance, and prognosis. Researchers are increasingly focusing on FOXM1 functions in tumor microenvironment, epigenetics, and immune infiltration. However, researchers have not comprehensively described FOXM1's involvement in tumor microenvironment shaping, epigenetics, and immune cell infiltration. Here we review the role of FOXM1 in the formation and development of malignant tumors, and we will provide a comprehensive summary of the role of FOXM1 in transcriptional regulation, interacting proteins, tumor microenvironment, epigenetics, and immune infiltration, and suggest areas for further research.
Collapse
Affiliation(s)
- Mengxi Li
- Hubei Key Laboratory of Diabetes and Angiopathy, Xianning Medical College, Hubei University of Science and Technology, Xianning, Hubei Province, P. R. China
- School of Nuclear Technology and Chemistry & Biology, Hubei University of Science and Technology, Xianning, Hubei Province, P. R. China
| | - Xuzheng Gao
- Hubei Key Laboratory of Diabetes and Angiopathy, Xianning Medical College, Hubei University of Science and Technology, Xianning, Hubei Province, P. R. China
| | - Yanting Su
- School of Basic Medical Sciences, Xianning Medical College, Hubei University of Science and Technology, Hubei University of Science and Technology, Xianning, Hubei Province, P. R. China
| | - Shigang Shan
- School of Basic Medical Sciences, Xianning Medical College, Hubei University of Science and Technology, Hubei University of Science and Technology, Xianning, Hubei Province, P. R. China
| | - Wenbin Qian
- School of Basic Medical Sciences, Xianning Medical College, Hubei University of Science and Technology, Hubei University of Science and Technology, Xianning, Hubei Province, P. R. China
| | - Zhenwang Zhang
- Hubei Key Laboratory of Diabetes and Angiopathy, Xianning Medical College, Hubei University of Science and Technology, Xianning, Hubei Province, P. R. China
- School of Basic Medical Sciences, Xianning Medical College, Hubei University of Science and Technology, Hubei University of Science and Technology, Xianning, Hubei Province, P. R. China
| | - Dan Zhu
- Hubei Key Laboratory of Diabetes and Angiopathy, Xianning Medical College, Hubei University of Science and Technology, Xianning, Hubei Province, P. R. China
- School of Basic Medical Sciences, Xianning Medical College, Hubei University of Science and Technology, Hubei University of Science and Technology, Xianning, Hubei Province, P. R. China
| |
Collapse
|
23
|
Huang A, Cheng J, Zhan Y, Zhou F, Xuan Y, Wang Y, Chen Q, Wang H, Xu X, Luo S, Cheng M. Hedgehog ligand and receptor cooperatively regulate EGFR stability and activity in non-small cell lung cancer. Cell Oncol (Dordr) 2024; 47:1405-1423. [PMID: 38568419 DOI: 10.1007/s13402-024-00938-6] [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] [Accepted: 03/13/2024] [Indexed: 09/25/2024] Open
Abstract
PURPOSE The hyperactivation of epidermal growth factor receptor (EGFR) plays a crucial role in non-small cell lung cancer (NSCLC). Hedgehog (Hh) signaling has been implicated in the tumorigenesis and progression of various cancers, however, its function in NSCLC cells remains controversial. Herein, we present a novel finding that challenges the current understanding of Hh signaling in tumor growth. METHODS Expression of Hh ligands and receptor were assessed using TCGA datasets, immunoblotting and immunohistochemical. Biological function of Hh ligands and receptor in NSCLC were tested using colony formation, cell count kit-8 (CCK-8) and xenograft assays. Biochemical effect of Hh ligands and receptor on regulating EGFR stability and activity were checked via immunoblotting. RESULTS Expression of Hh ligands and receptor was suppressed in NSCLC tissues, and the lower expression levels of these genes were associated with poor prognosis. Ptch1 binds to EGFR and facilitates its poly-ubiquitylation and degradation independent of downstream transcriptional signaling. Moreover, Hh ligands cooperate with Ptch1 to regulate the protein stability and activity of EGFR. This unique mechanism leads to a suppressive effect on NSCLC tumor growth. CONCLUSION Non-canonical Hh signaling pathway, involving cooperation between Hh ligands and their receptor Ptch1, facilitates the degradation of EGFR and attenuates its activity in NSCLC. These findings provide novel insights into the regulation of EGFR protein stability and activity, offer new diagnostic indicators for molecular typing of NSCLC and identify potential targets for targeted therapy of this challenging disease.
Collapse
Affiliation(s)
- Aidi Huang
- Jiangxi Clinical Research Center for Respiratory Diseases, Jiangxi Institute of Respiratory Disease, The Department of Respiratory and Critical Care Medicine, the First Affiliated Hospital, Jiangxi Medical College, Nanchang University, 330006, Nanchang, Jiangxi, China
- Jiangxi Key Laboratory of Molecular Diagnostics and Precision Medicine, Center for Experimental Medicine, the First Affiliated Hospital, Jiangxi Medical College, Nanchang University, 330006, Nanchang, Jiangxi, China
| | - Junyao Cheng
- Jiangxi Clinical Research Center for Respiratory Diseases, Jiangxi Institute of Respiratory Disease, The Department of Respiratory and Critical Care Medicine, the First Affiliated Hospital, Jiangxi Medical College, Nanchang University, 330006, Nanchang, Jiangxi, China
- Jiangxi Key Laboratory of Molecular Diagnostics and Precision Medicine, Center for Experimental Medicine, the First Affiliated Hospital, Jiangxi Medical College, Nanchang University, 330006, Nanchang, Jiangxi, China
| | - Yuan Zhan
- Institute of Molecular Pathology, Department of Pathology, the First Affiliated Hospital, Jiangxi Medical College, Nanchang University, 330006, Nanchang, Jiangxi, China
| | - Feifei Zhou
- Jiangxi Key Laboratory of Molecular Diagnostics and Precision Medicine, Center for Experimental Medicine, the First Affiliated Hospital, Jiangxi Medical College, Nanchang University, 330006, Nanchang, Jiangxi, China
- Department of Gastroenterology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, 250021, Jinan, Shandong, China
| | - Yanlu Xuan
- Jiangxi Clinical Research Center for Respiratory Diseases, Jiangxi Institute of Respiratory Disease, The Department of Respiratory and Critical Care Medicine, the First Affiliated Hospital, Jiangxi Medical College, Nanchang University, 330006, Nanchang, Jiangxi, China
- Jiangxi Key Laboratory of Molecular Diagnostics and Precision Medicine, Center for Experimental Medicine, the First Affiliated Hospital, Jiangxi Medical College, Nanchang University, 330006, Nanchang, Jiangxi, China
| | - Yiting Wang
- Department of Oncology, the First Affiliated Hospital, Jiangxi Medical College, Nanchang University, 330006, Nanchang, Jiangxi, China
| | - Qingjie Chen
- Department of Nuclear Medicine, the First Affiliated Hospital, Jiangxi Medical College, Nanchang University, 330006, Nanchang, Jiangxi, China
| | - Hailong Wang
- Jiangxi Key Laboratory of Molecular Diagnostics and Precision Medicine, Center for Experimental Medicine, the First Affiliated Hospital, Jiangxi Medical College, Nanchang University, 330006, Nanchang, Jiangxi, China
| | - Xinping Xu
- Jiangxi Clinical Research Center for Respiratory Diseases, Jiangxi Institute of Respiratory Disease, The Department of Respiratory and Critical Care Medicine, the First Affiliated Hospital, Jiangxi Medical College, Nanchang University, 330006, Nanchang, Jiangxi, China
| | - Shiwen Luo
- Jiangxi Key Laboratory of Molecular Diagnostics and Precision Medicine, Center for Experimental Medicine, the First Affiliated Hospital, Jiangxi Medical College, Nanchang University, 330006, Nanchang, Jiangxi, China.
| | - Minzhang Cheng
- Jiangxi Clinical Research Center for Respiratory Diseases, Jiangxi Institute of Respiratory Disease, The Department of Respiratory and Critical Care Medicine, the First Affiliated Hospital, Jiangxi Medical College, Nanchang University, 330006, Nanchang, Jiangxi, China.
- Jiangxi Key Laboratory of Molecular Diagnostics and Precision Medicine, Center for Experimental Medicine, the First Affiliated Hospital, Jiangxi Medical College, Nanchang University, 330006, Nanchang, Jiangxi, China.
| |
Collapse
|
24
|
Yan T, Jiang Q, Ni G, Ma H, Meng Y, Kang G, Xu M, Peng F, Li H, Chen X, Wang M. WZ-3146 acts as a novel small molecule inhibitor of KIF4A to inhibit glioma progression by inducing apoptosis. Cancer Cell Int 2024; 24:221. [PMID: 38937742 PMCID: PMC11209999 DOI: 10.1186/s12935-024-03409-y] [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: 03/12/2024] [Accepted: 06/19/2024] [Indexed: 06/29/2024] Open
Abstract
BACKGROUND Glioma is considered the most common primary malignant tumor of the central nervous system. Although traditional treatments have not achieved satisfactory outcomes, recently, targeted therapies for glioma have shown promising efficacy. However, due to the single-target nature of targeted therapy, traditional targeted therapies are ineffective; thus, novel therapeutic targets are urgently needed. METHODS The gene expression data for glioma patients were derived from the GEO (GSE4290, GSE50161), TCGA and CGGA databases. Next, the upregulated genes obtained from the above databases were cross-analyzed, finally, 10 overlapping genes (BIRC5, FOXM1, EZH2, CDK1, KIF11, KIF4A, NDC80, PBK, RRM2, and TOP2A) were ultimately screened and only KIF4A expression has the strongest correlation with clinical characteristics in glioma patients. Futher, the TCGA and CGGA database were utilized to explore the correlation of KIF4A expression with glioma prognosis. Then, qRT-PCR and Western blot was used to detect the KIF4A mRNA and protein expression level in glioma cells, respectively. And WZ-3146, the small molecule inhibitor targeting KIF4A, were screened by Cmap analysis. Subsequently, the effect of KIF4A knockdown or WZ-3146 treatment on glioma was measured by the MTT, EdU, Colony formation assay and Transwell assay. Ultimately, GSEA enrichment analysis was performed to find that the apoptotic pathway could be regulated by KIF4A in glioma, in addition, the effect of WZ-3146 on glioma apoptosis was detected by flow cytometry and Western blot. RESULTS In the present study, we confirmed that KIF4A is abnormally overexpressed in glioma. In addition, KIF4A overexpression is a key indicator of glioma prognosis; moreover, suppressing KIF4A expression can inhibit glioma progression. We also discovered that WZ-3146, a small molecule inhibitor of KIF4A, can induce apoptosis in glioma cells and exhibit antiglioma effects. CONCLUSION In conclusion, these observations demonstrated that targeting KIF4A can inhibit glioma progression. With further research, WZ-3146, a small molecule inhibitor of KIF4A, could be combined with other molecular targeted drugs to cooperatively inhibit glioma progression.
Collapse
Affiliation(s)
- Tao Yan
- Central Laboratory, Linyi People's Hospital, Linyi, Shandong Province, 276000, China
- Linyi Key Laboratory of Neurophysiology, Linyi People's Hospital, Linyi, Shandong Province, 276000, China
| | - Qing Jiang
- Department of Neurosurgery, First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, 150001, China
- Key Laboratory of Neurosurgery of Colleges and Universities in Heilongjiang Province, Harbin, Heilongjiang Province, 150001, China
| | - Guangpu Ni
- Linyi Key Laboratory of Neurophysiology, Linyi People's Hospital, Linyi, Shandong Province, 276000, China
- Department of Neurosurgery, Linyi People's Hospital, Shandong Second Medical University, Linyi, Shandong Province, 276000, China
| | - Haofeng Ma
- Linyi Key Laboratory of Neurophysiology, Linyi People's Hospital, Linyi, Shandong Province, 276000, China
- Department of Neurosurgery, Linyi People's Hospital, Shandong Second Medical University, Linyi, Shandong Province, 276000, China
| | - Yun Meng
- Central Laboratory, Linyi People's Hospital, Linyi, Shandong Province, 276000, China
- Linyi Key Laboratory of Neurophysiology, Linyi People's Hospital, Linyi, Shandong Province, 276000, China
| | - Guiqiong Kang
- Central Laboratory, Linyi People's Hospital, Linyi, Shandong Province, 276000, China
- Linyi Key Laboratory of Neurophysiology, Linyi People's Hospital, Linyi, Shandong Province, 276000, China
| | - Meifang Xu
- Linyi Key Laboratory of Neurophysiology, Linyi People's Hospital, Linyi, Shandong Province, 276000, China
- Department of Neurology, Linyi People's Hospital, Shandong Second Medical University, Linyi, Shandong Province, 276000, China
| | - Fei Peng
- Department of Neurosurgery and Neurosurgical Disease Research Centre, the Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong Province, China
| | - Huadong Li
- Department of Neurosurgery, Linyi People's Hospital, Shandong Second Medical University, Linyi, Shandong Province, 276000, China.
| | - Xin Chen
- Department of Neurosurgery, First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, 150001, China.
- Key Laboratory of Neurosurgery of Colleges and Universities in Heilongjiang Province, Harbin, Heilongjiang Province, 150001, China.
| | - Mingguang Wang
- Department of Neurosurgery, Linyi People's Hospital, Shandong Second Medical University, Linyi, Shandong Province, 276000, China.
| |
Collapse
|
25
|
Li Q, Wang T, Wang X, Ge X, Yang T, Wang W. DDX56 promotes EMT and cancer stemness via MELK-FOXM1 axis in hepatocellular carcinoma. iScience 2024; 27:109827. [PMID: 38827395 PMCID: PMC11141150 DOI: 10.1016/j.isci.2024.109827] [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: 09/01/2023] [Revised: 03/06/2024] [Accepted: 04/24/2024] [Indexed: 06/04/2024] Open
Abstract
Hepatocellular carcinoma (HCC) is a major global cause of death, with epithelial-mesenchymal transition (EMT) and cancer stem cell (CSC)-like properties contributing to its metastasis. DEAD box helicase 56 (DDX56) is involved in carcinogenesis, but its role in EMT induction and stem phenotype maintenance is unclear. This study assessed the impact of DDX56 absence on HCC cell stemness and EMT. DDX56 was found to be overexpressed in HCC tissues, correlating with disease stage and prognosis. In vitro, DDX56 stimulated tumor cell proliferation, migration, invasion, EMT, and stemness. It also enhanced maternal embryonic leucine-zipper kinase (MELK)-mediated forkhead box protein M1 (FOXM1) expression, regulating cancer stemness and malignant traits. In vivo, DDX56 knockdown in tumor-bearing mice reduced tumorigenicity and lung metastasis by modulating the MELK-FOXM1 signaling pathway. Collectively, DDX56 initiates stem cell-like traits in HCC and promotes EMT via MELK-FOXM1 activation, shedding light on HCC pathogenesis and suggesting a potential anti-cancer therapeutic target.
Collapse
Affiliation(s)
- Qing Li
- Department of Internal Medicine, Third Affiliated Hospital of Jinzhou Medical University, Jinzhou 121000, Liaoning Province, China
| | - Tianyi Wang
- Department of General Surgery, First Affiliated Hospital of Jinzhou Medical University, Jinzhou 121001, Liaoning Province, China
| | - Ximin Wang
- Department of General Surgery, First Affiliated Hospital of Jinzhou Medical University, Jinzhou 121001, Liaoning Province, China
| | - XinYu Ge
- Department of General Surgery, First Affiliated Hospital of Jinzhou Medical University, Jinzhou 121001, Liaoning Province, China
| | - Tao Yang
- Department of General Surgery, First Affiliated Hospital of Jinzhou Medical University, Jinzhou 121001, Liaoning Province, China
| | - Wei Wang
- Department of General Surgery, First Affiliated Hospital of Jinzhou Medical University, Jinzhou 121001, Liaoning Province, China
| |
Collapse
|
26
|
Wei Y, Chen Z, Li Y, Song K. The splicing factor WBP11 mediates MCM7 intron retention to promote the malignant progression of ovarian cancer. Oncogene 2024; 43:1565-1578. [PMID: 38561505 DOI: 10.1038/s41388-024-03015-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2023] [Revised: 03/20/2024] [Accepted: 03/21/2024] [Indexed: 04/04/2024]
Abstract
Accumulating studies suggest that splicing factors play important roles in many diseases including human cancers. Our study revealed that WBP11, a core splicing factor, is highly expressed in ovarian cancer (OC) tissues and associated with a poor prognosis. WBP11 inhibition significantly impaired the proliferation and mobility of ovarian cancer cells in vitro and in vivo. Furthermore, FOXM1 transcriptionally activated WBP11 expression by directly binding to its promoter in OC cells. Importantly, RNA-seq and alternative splicing event analysis revealed that WBP11 silencing decreased the expression of MCM7 by regulating intron 4 retention. MCM7 inhibition attenuated the increase in malignant behaviors of WBP11-overexpressing OC cells. Overall, WBP11 was identified as an oncogenic splicing factor that contributes to malignant progression by repressing intron 4 retention of MCM7 in OC cells. Thus, WBP11 is an oncogenic splicing factor with potential therapeutic and prognostic implications in OC.
Collapse
Affiliation(s)
- Yuan Wei
- Department of Obstetrics and Gynecology, Qilu Hospital of Shandong University, 107 Wenhua Xi Road, Ji'nan, 250012, Shandong, China
| | - Zhongshao Chen
- Department of Obstetrics and Gynecology, Qilu Hospital of Shandong University, 107 Wenhua Xi Road, Ji'nan, 250012, Shandong, China
| | - Yingwei Li
- Department of Obstetrics and Gynecology, Qilu Hospital of Shandong University, Medical Integration and Practice Center, Cheeloo College of Medicine, Shandong University, Ji'nan, 250012, Shandong, China.
| | - Kun Song
- Department of Obstetrics and Gynecology, Qilu Hospital of Shandong University, 107 Wenhua Xi Road, Ji'nan, 250012, Shandong, China.
| |
Collapse
|
27
|
Zhang P, Cao J, Liang X, Su Z, Zhang B, Wang Z, Xie J, Chen G, Chen X, Zhang J, Feng Y, Xu Q, Song J, Hong A, Chen X, Zhang Y. Lian-Mei-Yin formula alleviates diet-induced hepatic steatosis by suppressing Yap1/FOXM1 pathway-dependent lipid synthesis. Acta Biochim Biophys Sin (Shanghai) 2024; 56:621-633. [PMID: 38516704 DOI: 10.3724/abbs.2024025] [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] [Indexed: 03/23/2024] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) is the most common chronic liver disease, with a global prevalence of 25%. Patients with NAFLD are more likely to suffer from advanced liver disease, cardiovascular disease, or type II diabetes. However, unfortunately, there is still a shortage of FDA-approved therapeutic agents for NAFLD. Lian-Mei-Yin (LMY) is a traditional Chinese medicine formula used for decades to treat liver disorders. It has recently been applied to type II diabetes which is closely related to insulin resistance. Given that NAFLD is another disease involved in insulin resistance, we hypothesize that LMY might be a promising formula for NAFLD therapy. Herein, we verify that the LMY formula effectively reduces hepatic steatosis in diet-induced zebrafish and NAFLD model mice in a time- and dose-dependent manner. Mechanistically, LMY suppresses Yap1-mediated Foxm1 activation, which is crucial for the occurrence and development of NAFLD. Consequently, lipogenesis is ameliorated by LMY administration. In summary, the LMY formula alleviates diet-induced NAFLD in zebrafish and mice by inhibiting Yap1/Foxm1 signaling-mediated NAFLD pathology.
Collapse
Affiliation(s)
- Peiguang Zhang
- Department of Cell Biology, College of Life Science and Technology, Jinan University; State Key Laboratory of Bioactive Molecules and Druggability Assessment, Jinan University; National Engineering Research Center of Genetic Medicine; Guangdong Provincial Key Laboratory of Bioengineering Medicine; Guangdong Provincial Biotechnology Drug & Engineering Technology Research Center, Jinan University, Guangzhou 510632, China
| | - Jieqiong Cao
- Department of Cell Biology, College of Life Science and Technology, Jinan University; State Key Laboratory of Bioactive Molecules and Druggability Assessment, Jinan University; National Engineering Research Center of Genetic Medicine; Guangdong Provincial Key Laboratory of Bioengineering Medicine; Guangdong Provincial Biotechnology Drug & Engineering Technology Research Center, Jinan University, Guangzhou 510632, China
| | - Xujing Liang
- Department of Infectious Disease, the First Affiliated Hospital of Jinan University, Guangzhou 510630, China
| | - Zijian Su
- Department of Cell Biology, College of Life Science and Technology, Jinan University; State Key Laboratory of Bioactive Molecules and Druggability Assessment, Jinan University; National Engineering Research Center of Genetic Medicine; Guangdong Provincial Key Laboratory of Bioengineering Medicine; Guangdong Provincial Biotechnology Drug & Engineering Technology Research Center, Jinan University, Guangzhou 510632, China
| | - Bihui Zhang
- Department of Cell Biology, College of Life Science and Technology, Jinan University; State Key Laboratory of Bioactive Molecules and Druggability Assessment, Jinan University; National Engineering Research Center of Genetic Medicine; Guangdong Provincial Key Laboratory of Bioengineering Medicine; Guangdong Provincial Biotechnology Drug & Engineering Technology Research Center, Jinan University, Guangzhou 510632, China
| | - Zhenyu Wang
- Department of Cell Biology, College of Life Science and Technology, Jinan University; State Key Laboratory of Bioactive Molecules and Druggability Assessment, Jinan University; National Engineering Research Center of Genetic Medicine; Guangdong Provincial Key Laboratory of Bioengineering Medicine; Guangdong Provincial Biotechnology Drug & Engineering Technology Research Center, Jinan University, Guangzhou 510632, China
| | - Junye Xie
- Department of Cell Biology, College of Life Science and Technology, Jinan University; State Key Laboratory of Bioactive Molecules and Druggability Assessment, Jinan University; National Engineering Research Center of Genetic Medicine; Guangdong Provincial Key Laboratory of Bioengineering Medicine; Guangdong Provincial Biotechnology Drug & Engineering Technology Research Center, Jinan University, Guangzhou 510632, China
| | - Gengrui Chen
- School of Biotechnology and Health Sciences, Wuyi University, Jiangmen 529020, China
| | - Xue Chen
- Department of Cell Biology, College of Life Science and Technology, Jinan University; State Key Laboratory of Bioactive Molecules and Druggability Assessment, Jinan University; National Engineering Research Center of Genetic Medicine; Guangdong Provincial Key Laboratory of Bioengineering Medicine; Guangdong Provincial Biotechnology Drug & Engineering Technology Research Center, Jinan University, Guangzhou 510632, China
| | - Jinting Zhang
- Department of Cell Biology, College of Life Science and Technology, Jinan University; State Key Laboratory of Bioactive Molecules and Druggability Assessment, Jinan University; National Engineering Research Center of Genetic Medicine; Guangdong Provincial Key Laboratory of Bioengineering Medicine; Guangdong Provincial Biotechnology Drug & Engineering Technology Research Center, Jinan University, Guangzhou 510632, China
| | - Yanxian Feng
- School of Biotechnology and Health Sciences, Wuyi University, Jiangmen 529020, China
| | - Qin Xu
- Guangzhou University of Traditional Chinese Medicine, Guangzhou 510006, China
| | - Jianping Song
- Guangzhou University of Traditional Chinese Medicine, Guangzhou 510006, China
| | - An Hong
- Department of Cell Biology, College of Life Science and Technology, Jinan University; State Key Laboratory of Bioactive Molecules and Druggability Assessment, Jinan University; National Engineering Research Center of Genetic Medicine; Guangdong Provincial Key Laboratory of Bioengineering Medicine; Guangdong Provincial Biotechnology Drug & Engineering Technology Research Center, Jinan University, Guangzhou 510632, China
| | - Xiaojia Chen
- Department of Cell Biology, College of Life Science and Technology, Jinan University; State Key Laboratory of Bioactive Molecules and Druggability Assessment, Jinan University; National Engineering Research Center of Genetic Medicine; Guangdong Provincial Key Laboratory of Bioengineering Medicine; Guangdong Provincial Biotechnology Drug & Engineering Technology Research Center, Jinan University, Guangzhou 510632, China
| | - Yibo Zhang
- Department of Cell Biology, College of Life Science and Technology, Jinan University; State Key Laboratory of Bioactive Molecules and Druggability Assessment, Jinan University; National Engineering Research Center of Genetic Medicine; Guangdong Provincial Key Laboratory of Bioengineering Medicine; Guangdong Provincial Biotechnology Drug & Engineering Technology Research Center, Jinan University, Guangzhou 510632, China
| |
Collapse
|
28
|
Andrade AAR, Pauli F, Pressete CG, Zavan B, Hanemann JAC, Miyazawa M, Fonseca R, Caixeta ES, Nacif JLM, Aissa AF, Barreiro EJ, Ionta M. Antiproliferative Activity of N-Acylhydrazone Derivative on Hepatocellular Carcinoma Cells Involves Transcriptional Regulation of Genes Required for G2/M Transition. Biomedicines 2024; 12:892. [PMID: 38672246 PMCID: PMC11048582 DOI: 10.3390/biomedicines12040892] [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: 02/26/2024] [Revised: 04/07/2024] [Accepted: 04/08/2024] [Indexed: 04/28/2024] Open
Abstract
Liver cancer is the second leading cause of cancer-related death in males. It is estimated that approximately one million deaths will occur by 2030 due to hepatic cancer. Hepatocellular carcinoma (HCC) is the most prevalent primary liver cancer subtype and is commonly diagnosed at an advanced stage. The drug arsenal used in systemic therapy for HCC is very limited. Multikinase inhibitors sorafenib (Nexavar®) and lenvatinib (Lenvima®) have been used as first-line drugs with modest therapeutic effects. In this scenario, it is imperative to search for new therapeutic strategies for HCC. Herein, the antiproliferative activity of N-acylhydrazone derivatives was evaluated on HCC cells (HepG2 and Hep3B), which were chemically planned on the ALL-993 scaffold, a potent inhibitor of vascular endothelial growth factor 2 (VEGFR2). The substances efficiently reduced the viability of HCC cells, and the LASSBio-2052 derivative was the most effective. Further, we demonstrated that LASSBio-2052 treatment induced FOXM1 downregulation, which compromises the transcriptional activation of genes required for G2/M transition, such as AURKA and AURKB, PLK1, and CDK1. In addition, LASSBio-2052 significantly reduced CCNB1 and CCND1 expression in HCC cells. Our findings indicate that LASSBio-2052 is a promising prototype for further in vivo studies.
Collapse
Affiliation(s)
| | - Fernanda Pauli
- Institute of Chemistry, Fluminense Federal University, Niterói 24020-140, RJ, Brazil
| | - Carolina Girotto Pressete
- Institute of Biomedical Sciences, Federal University of Alfenas, Alfenas 37130-001, MG, Brazil (A.F.A.)
| | - Bruno Zavan
- Institute of Biomedical Sciences, Federal University of Alfenas, Alfenas 37130-001, MG, Brazil (A.F.A.)
| | | | - Marta Miyazawa
- School of Dentistry, Federal University of Alfenas, Alfenas 37130-001, MG, Brazil
| | - Rafael Fonseca
- Institute of Biomedical Sciences, Federal University of Alfenas, Alfenas 37130-001, MG, Brazil (A.F.A.)
| | - Ester Siqueira Caixeta
- Institute of Biomedical Sciences, Federal University of Alfenas, Alfenas 37130-001, MG, Brazil (A.F.A.)
| | | | - Alexandre Ferro Aissa
- Institute of Biomedical Sciences, Federal University of Alfenas, Alfenas 37130-001, MG, Brazil (A.F.A.)
| | - Eliezer J. Barreiro
- Laboratory of Evaluation and Synthesis of Bioactive Substances (LASSBio), Institute of Biomedical Sciences, Federal University of Rio de Janeiro, Rio de Janeiro 21941-914, RJ, Brazil
| | - Marisa Ionta
- Institute of Biomedical Sciences, Federal University of Alfenas, Alfenas 37130-001, MG, Brazil (A.F.A.)
| |
Collapse
|
29
|
Sun M, Zhang S, Wang J, Du G, Ji T. Synthesis of Novel Acetyl-11-keto-β-boswellic Acid Derivatives as Potential Anti-GBM Agents. Chem Biodivers 2024; 21:e202301979. [PMID: 38302832 DOI: 10.1002/cbdv.202301979] [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: 12/16/2023] [Revised: 02/01/2024] [Accepted: 02/01/2024] [Indexed: 02/03/2024]
Abstract
Acetyl-11-keto-β-boswellic acid (AKBA) is known to inhibit the growth of glioblastoma (GBM) cells and subcutaneous GBM. A series of acetyl-11-keto-β-boswellic acid (AKBA) derivatives containing the oxime-ester functionality or amide side chains were synthesized, and their anti-GBM activities were evaluated. Some of these compounds exhibited significant inhibitory activity against cell proliferation in U87 and U251 GBM cell lines, with IC50 values in the micromolar concentration range. Cellular thermal shift analysis showed that A-01 and A-10 improved the thermal stability of FOXM1, indicating that these highly active compounds may directly bind to FOXM1 in cells. Docking studies of the two most active compounds, A-01 and A-10, revealed key interactions between these compounds and the active site of FOXM1, in which the amide moiety at the C-24 position was essential for improving the activity. These results suggested that A-10 is a suitable lead molecule for the development of FOXM1 inhibitors. Thus, the rational design of AKBA derivatives with amide side chains holds significant potential for discovering of a new class of triterpenoids capable of inhibiting GBM cell proliferation.
Collapse
Affiliation(s)
- Mingxia Sun
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, 100050, China
| | - Sen Zhang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, 100050, China
- Key Laboratory of Drug Target Research and Drug Screen, Institute of Materia Medica, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, 100050, China
| | - Jinhua Wang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, 100050, China
- Key Laboratory of Drug Target Research and Drug Screen, Institute of Materia Medica, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, 100050, China
| | - Guanhua Du
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, 100050, China
- Key Laboratory of Drug Target Research and Drug Screen, Institute of Materia Medica, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, 100050, China
| | - Tengfei Ji
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, 100050, China
| |
Collapse
|
30
|
Zhao K, Li X, Feng Y, Wang J, Yao W. The role of kinesin family members in hepatobiliary carcinomas: from bench to bedside. Biomark Res 2024; 12:30. [PMID: 38433242 PMCID: PMC10910842 DOI: 10.1186/s40364-024-00559-z] [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: 10/06/2023] [Accepted: 01/03/2024] [Indexed: 03/05/2024] Open
Abstract
As a major component of the digestive system malignancies, tumors originating from the hepatic and biliary ducts seriously endanger public health. The kinesins (KIFs) are molecular motors that enable the microtubule-dependent intracellular trafficking necessary for mitosis and meiosis. Normally, the stability of KIFs is essential to maintain cell proliferation and genetic homeostasis. However, aberrant KIFs activity may destroy this dynamic stability, leading to uncontrolled cell division and tumor initiation. In this work, we have made an integral summarization of the specific roles of KIFs in hepatocellular and biliary duct carcinogenesis, referring to aberrant signal transduction and the potential for prognostic evaluation. Additionally, current clinical applications of KIFs-targeted inhibitors have also been discussed, including their efficacy advantages, relationship with drug sensitivity or resistance, the feasibility of combination chemotherapy or other targeted agents, as well as the corresponding clinical trials. In conclusion, the abnormally activated KIFs participate in the regulation of tumor progression via a diverse range of mechanisms and are closely associated with tumor prognosis. Meanwhile, KIFs-aimed inhibitors also carry out a promising tumor-targeted therapeutic strategy that deserves to be further investigated in hepatobiliary carcinoma (HBC).
Collapse
Affiliation(s)
- Kai Zhao
- Department of Biliary and Pancreatic Surgery, Cancer Research Center Affiliated Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 430030, Wuhan, Hubei, China
| | - Xiangyu Li
- Department of Thoracic Surgery Affiliated Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 430030, Wuhan, Hubei, China
| | - Yunxiang Feng
- Department of Biliary and Pancreatic Surgery, Cancer Research Center Affiliated Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 430030, Wuhan, Hubei, China
| | - Jianming Wang
- Department of Biliary and Pancreatic Surgery, Cancer Research Center Affiliated Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 430030, Wuhan, Hubei, China.
- Affiliated Tianyou Hospital, Wuhan University of Science & Technology, 430064, Wuhan, China.
| | - Wei Yao
- Department of Oncology Affiliated Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 430030, Wuhan, Hubei, China.
| |
Collapse
|
31
|
Ren Y, Liang H, Huang Y, Miao Y, Li R, Qiang J, Wu L, Qi J, Li Y, Xia Y, Huang L, Wang S, Kong X, Zhou Y, Zhang Q, Zhu G. Key candidate genes and pathways in T lymphoblastic leukemia/lymphoma identified by bioinformatics and serological analyses. Front Immunol 2024; 15:1341255. [PMID: 38464517 PMCID: PMC10920334 DOI: 10.3389/fimmu.2024.1341255] [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: 11/20/2023] [Accepted: 02/08/2024] [Indexed: 03/12/2024] Open
Abstract
T-cell acute lymphoblastic leukemia (T-ALL)/T-cell lymphoblastic lymphoma (T-LBL) is an uncommon but highly aggressive hematological malignancy. It has high recurrence and mortality rates and is challenging to treat. This study conducted bioinformatics analyses, compared genetic expression profiles of healthy controls with patients having T-ALL/T-LBL, and verified the results through serological indicators. Data were acquired from the GSE48558 dataset from Gene Expression Omnibus (GEO). T-ALL patients and normal T cells-related differentially expressed genes (DEGs) were investigated using the online analysis tool GEO2R in GEO, identifying 78 upregulated and 130 downregulated genes. Gene Ontology (GO) and protein-protein interaction (PPI) network analyses of the top 10 DEGs showed enrichment in pathways linked to abnormal mitotic cell cycles, chromosomal instability, dysfunction of inflammatory mediators, and functional defects in T-cells, natural killer (NK) cells, and immune checkpoints. The DEGs were then validated by examining blood indices in samples obtained from patients, comparing the T-ALL/T-LBL group with the control group. Significant differences were observed in the levels of various blood components between T-ALL and T-LBL patients. These components include neutrophils, lymphocyte percentage, hemoglobin (HGB), total protein, globulin, erythropoietin (EPO) levels, thrombin time (TT), D-dimer (DD), and C-reactive protein (CRP). Additionally, there were significant differences in peripheral blood leukocyte count, absolute lymphocyte count, creatinine, cholesterol, low-density lipoprotein, folate, and thrombin times. The genes and pathways associated with T-LBL/T-ALL were identified, and peripheral blood HGB, EPO, TT, DD, and CRP were key molecular markers. This will assist the diagnosis of T-ALL/T-LBL, with applications for differential diagnosis, treatment, and prognosis.
Collapse
Affiliation(s)
- Yansong Ren
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
| | - Haoyue Liang
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
| | - Yali Huang
- Clinical Laboratory of Zhengning County People's Hospital, Qingyang, Gansu, China
| | - Yuyang Miao
- Department of Geriatrics, Tianjin Medical University General Hospital, Tianjin Geriatrics Institute, Tianjin, China
| | - Ruihua Li
- Department of Geriatrics, Tianjin Medical University General Hospital, Tianjin Geriatrics Institute, Tianjin, China
| | - Junlian Qiang
- Department of Geriatrics, Tianjin Medical University General Hospital, Tianjin Geriatrics Institute, Tianjin, China
| | - Lihong Wu
- Department of Geriatrics, Tianjin Medical University General Hospital, Tianjin Geriatrics Institute, Tianjin, China
| | - Jinfeng Qi
- Department of Geriatrics, Tianjin Medical University General Hospital, Tianjin Geriatrics Institute, Tianjin, China
| | - Ying Li
- Department of Geriatrics, Tianjin Medical University General Hospital, Tianjin Geriatrics Institute, Tianjin, China
| | - Yonghui Xia
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
| | - Lunhui Huang
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
| | - Shoulei Wang
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
| | - Xiaodong Kong
- Department of Geriatrics, Tianjin Medical University General Hospital, Tianjin Geriatrics Institute, Tianjin, China
| | - Yuan Zhou
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
| | - Qiang Zhang
- Department of Geriatrics, Tianjin Medical University General Hospital, Tianjin Geriatrics Institute, Tianjin, China
| | - Guoqing Zhu
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
| |
Collapse
|
32
|
Chen Y, Yang Y, Wang N, Liu R, Wu Q, Pei H, Li W. β-Sitosterol suppresses hepatocellular carcinoma growth and metastasis via FOXM1-regulated Wnt/β-catenin pathway. J Cell Mol Med 2024; 28:e18072. [PMID: 38063438 PMCID: PMC10844700 DOI: 10.1111/jcmm.18072] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2023] [Revised: 11/20/2023] [Accepted: 11/24/2023] [Indexed: 02/08/2024] Open
Abstract
β-Sitosterol is a natural compound with demonstrated anti-cancer properties against various cancers. However, its effects on hepatocellular carcinoma (HCC) and the underlying mechanisms are not well understood. This study aims to investigate the impact of β-sitosterol on HCC. In this study, we investigated the effects of β-sitosterol on HCC tumour growth and metastasis using a xenograft mouse model and a range of molecular analyses, including bioinformatics, real-time PCR, western blotting, lentivirus transfection, CCK8, scratch and transwell assays. The results found that β-sitosterol significantly inhibits HepG2 cell proliferation, migration and invasion both in vitro and in vivo. Bioinformatics analysis identifies forkhead box M1 (FOXM1) as a potential target for β-sitosterol in HCC treatment. FOXM1 is upregulated in HCC tissues and cell lines, correlating with poor prognosis in patients. β-Sitosterol downregulates FOXM1 expression in vitro and in vivo. FOXM1 overexpression mitigates β-sitosterol's inhibitory effects on HepG2 cells. Additionally, β-sitosterol suppresses epithelial-mesenchymal transition (EMT) in HepG2 cells, while FOXM1 overexpression promotes EMT. Mechanistically, β-sitosterol inhibits Wnt/β-catenin signalling by downregulating FOXM1, regulating target gene transcription related to HepG2 cell proliferation and metastasis. β-Sitosterol shows promising potential as a therapeutic candidate for inhibiting HCC growth and metastasis through FOXM1 downregulation and Wnt/β-catenin signalling inhibition.
Collapse
Affiliation(s)
- Yuankun Chen
- Department of Infectious and Tropical DiseasesThe Second Affiliated Hospital of Hainan Medical UniversityHaikouHainanChina
- Key Laboratory of Tropical Translational Medicine of Ministry of HealthHainan Medical UniversityHaikouHainanChina
- Department of Clinical LaboratoryThe Second Affiliated Hospital of Hainan Medical UniversityHaikouHainanChina
| | - Yijun Yang
- Department of Infectious and Tropical DiseasesThe Second Affiliated Hospital of Hainan Medical UniversityHaikouHainanChina
- Key Laboratory of Tropical Translational Medicine of Ministry of HealthHainan Medical UniversityHaikouHainanChina
| | - Nengyi Wang
- Department of Infectious and Tropical DiseasesThe Second Affiliated Hospital of Hainan Medical UniversityHaikouHainanChina
- Key Laboratory of Tropical Translational Medicine of Ministry of HealthHainan Medical UniversityHaikouHainanChina
| | - Rui Liu
- Department of Infectious and Tropical DiseasesThe Second Affiliated Hospital of Hainan Medical UniversityHaikouHainanChina
- Key Laboratory of Tropical Translational Medicine of Ministry of HealthHainan Medical UniversityHaikouHainanChina
| | - Qiuping Wu
- Department of Infectious and Tropical DiseasesThe Second Affiliated Hospital of Hainan Medical UniversityHaikouHainanChina
- Key Laboratory of Tropical Translational Medicine of Ministry of HealthHainan Medical UniversityHaikouHainanChina
| | - Hua Pei
- Department of Infectious and Tropical DiseasesThe Second Affiliated Hospital of Hainan Medical UniversityHaikouHainanChina
- Key Laboratory of Tropical Translational Medicine of Ministry of HealthHainan Medical UniversityHaikouHainanChina
- Department of Clinical LaboratoryThe Second Affiliated Hospital of Hainan Medical UniversityHaikouHainanChina
| | - Wenting Li
- Department of Infectious and Tropical DiseasesThe Second Affiliated Hospital of Hainan Medical UniversityHaikouHainanChina
- Key Laboratory of Tropical Translational Medicine of Ministry of HealthHainan Medical UniversityHaikouHainanChina
- Department of Infectious DiseasesThe First Affiliated Hospital of Anhui Medical UniversityHefeiAnhuiChina
| |
Collapse
|
33
|
Liu BW, Cao JL, Wang Y, Zhao X, Zeng Q, Liu WP, Zhang JH, Fan YZ, Dou J. GANT61, an inhibitor of Gli1, inhibits the proliferation and migration of hepatocellular carcinoma cells. J Investig Med 2024; 72:181-192. [PMID: 37724700 DOI: 10.1177/10815589231204056] [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] [Indexed: 09/21/2023]
Abstract
Constitutive activation of Hedgehog (Hh) signaling has been implicated in many cancers including hepatocellular carcinoma (HCC). Among them, the terminal glioma-associated oncogene homolog 1 (Gli1) regulates the expression of critical genes in the Hh pathway. The current study aims to evaluate the anti-HCC effect of the Gli1 inhibitor, GANT61. In vitro analysis including cell counting kit-8 (CCK-8) assay, flow cytometry, and migration and invasion assay were adopted to evaluate the effect of GANT61 on HCC cell lines. In vivo, xenograft studies were also performed to verify the effect of GANT61 on HCC. By CCK-8 assay, we found that GANT61 could significantly reduce the growth of HCC cell lines Huh7 and hemophagocytic lymphohistiocytosis (HLE), and their IC50 concentrations were 4.481 and 6.734 μM, respectively. Flow cytometry shows that GANT61 induced cell cycle arrest in the G2/M phase and accelerated apoptosis of both HLE and Huh7 cells. While migration and invasion assay shows that GANT61 weakens cells' migration and invasion ability. Besides that, GANT61 inhibits the expression of Gli1, FoxM1, CyclinD1, and Bcl-2, upregulates the level of Bax protein, and also reverses the epithelial-mesenchymal transition program by downregulating the expression of Vimentin and N-Cadherin and upregulating the expression of epithelial E-Cadherin expression. Furthermore, GANT61 inhibits the growth of subcutaneous xenografts of Huh7 cells in nude mice. Overall, this study suggests that Gli1 is a potential target for therapy and GANT61 shows promising therapeutic potential for future treatment in HCC.
Collapse
Affiliation(s)
- Bao-Wang Liu
- Department of Hepatobiliary Surgery, The Third Hospital of Hebei Medical University, Shijiazhuang, Hebei Province, China
| | - Jing-Lin Cao
- Department of Hepatobiliary Surgery, The Third Hospital of Hebei Medical University, Shijiazhuang, Hebei Province, China
| | - Yang Wang
- Department of Hepatobiliary Surgery, The Third Hospital of Hebei Medical University, Shijiazhuang, Hebei Province, China
| | - Xin Zhao
- Department of Hepatobiliary Surgery, The Third Hospital of Hebei Medical University, Shijiazhuang, Hebei Province, China
| | - Qiang Zeng
- Department of Hepatobiliary Surgery, The Third Hospital of Hebei Medical University, Shijiazhuang, Hebei Province, China
| | - Wen-Peng Liu
- Department of Hepatobiliary Surgery, The Third Hospital of Hebei Medical University, Shijiazhuang, Hebei Province, China
| | - Jun-Hong Zhang
- Department of Hepatobiliary Surgery, The Third Hospital of Hebei Medical University, Shijiazhuang, Hebei Province, China
| | - Yi-Ze Fan
- Department of Hepatobiliary Surgery, The Third Hospital of Hebei Medical University, Shijiazhuang, Hebei Province, China
| | - Jian Dou
- Department of Hepatobiliary Surgery, The Third Hospital of Hebei Medical University, Shijiazhuang, Hebei Province, China
| |
Collapse
|
34
|
Zheng H, Han X, Liu Q, Zhou L, Zhu Y, Wang J, Hu W, Zhu F, Liu R. Construction of immune-related molecular diagnostic and predictive models of hepatocellular carcinoma based on machine learning. Heliyon 2024; 10:e24854. [PMID: 38312556 PMCID: PMC10835357 DOI: 10.1016/j.heliyon.2024.e24854] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2023] [Revised: 01/10/2024] [Accepted: 01/16/2024] [Indexed: 02/06/2024] Open
Abstract
Background To exploit hepatocellular carcinoma (HCC) diagnostic substances, we identify potential predictive markers based on machine learning and to explore the significance of immune cell infiltration in this pathology. Method Three HCC gene expression datasets were used for weighted gene co-expression network analysis (WGCNA) and differential expression analysis. Least Absolute Shrinkage and Selection Operator (LASSO) and Random Forest were applied to identify candidate biomarkers. The diagnostic value of HCC diagnostic gene biomarkers was further assessed by the area under the ROC curve observed in the validation dataset. CIBERSORT was used to analyze 22 immune cell fractions from HCC patients and to analyze their correlation with diagnostic markers. In addition, the prognostic value of the markers and the sensitivity of the drugs were analyzed. Result WGCNA and differential expression analysis were used to screen 396 distinct gene signatures in HCC tissues. They were mostly engaged in cytoplasmic fusion and the cell division cycle, according to gene enrichment analyses. Five genes were shown to have a high diagnostic value for use as diagnostic biomarkers for HCC, including EFHD1 (AUC = 0.77), KIF4A (AUC = 0.97), UBE2C (AUC = 0.96), SMYD3 (AUC = 0.91), and MCM7 (AUC = 0.93). T cells, NK cells, macrophages, and dendritic cells were found to be related to diagnostic markers in HCC tissues by immune cell infiltration analysis, indicating that these cells are intimately linked to the onset and spread of HCC. Concurrently, these five genes and their constructed models have considerable prognostic value. Conclusion These five genes (EFHD1, KIF4A, UBE2C, SMYD3, and MCM7) may serve as new candidate molecular markers for HCC, providing new insights for future diagnosis, prognosis, and molecular therapy of HCC.
Collapse
Affiliation(s)
- Hui Zheng
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing, 210009, China
| | - Xu Han
- School of Public Health, Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Qian Liu
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing, 210009, China
| | - Li Zhou
- School of Public Health, Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Yawen Zhu
- School of Public Health, Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Jiaqi Wang
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing, 210009, China
| | - Wenjing Hu
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing, 210009, China
| | - Fengcai Zhu
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing, 210009, China
- National Health Commission Key Laboratory of Enteric Pathogenic Microbiology, Jiangsu Provincial Center for Disease Control and Prevention, Nanjing, Jiangsu Province, China
- School of Public Health, Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Ran Liu
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing, 210009, China
| |
Collapse
|
35
|
Guan X, Li Y, Guan X, Fan L, Ying J. XYA-2: a marine-derived compound targeting apoptosis and multiple signaling pathways in pancreatic cancer. PeerJ 2024; 12:e16805. [PMID: 38250721 PMCID: PMC10798151 DOI: 10.7717/peerj.16805] [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: 10/20/2023] [Accepted: 12/28/2023] [Indexed: 01/23/2024] Open
Abstract
Background Pancreatic cancer is a highly aggressive and fatal disease with limited treatment options and poor prognosis for patients. This study aimed to investigate the impact of XYA-2 {N-(3,7-dimethyl-2,6-octadienyl)-2-aza-2-deoxychaetoviridin A}, a nitrogenated azaphilon previously reported from a deep-sea-derived fungus on the progression of pancreatic cancer cells. Methods The inhibitory effects of XYA-2 on cell proliferation, clonogenic potential, cell cycle progression, apoptosis, migration, and invasion were assessed using various assays. The CCK-8 assay, clone formation assay, flow cytometry assay, wound healing assay, and transwell assay were employed to evaluate cell proliferation, clonogenic potential, cell cycle progression, apoptosis, migration, and invasion, respectively. Moreover, we employed RNA-seq and bioinformatics analyses to uncover the underlying mechanism by which XYA-2 influences pancreatic cancer cells. The revealed mechanism was subsequently validated through qRT-PCR. Results Our results demonstrated that XYA-2 dose-dependently inhibited the proliferation of pancreatic cancer cells and induced cell cycle arrest and apoptosis. Additionally, XYA-2 exerted a significant inhibitory effect on the invasion and migration of cancer cells. Moreover, XYA-2 was found to regulate the expression of genes involved in multiple cancer-related pathways based on our RNA-seq and bioinformatics analysis. Conclusion These findings highlight the potential of XYA-2 as a promising therapeutic option for the treatment of pancreatic cancer.
Collapse
Affiliation(s)
- Xiaoqing Guan
- Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Zhejiang, China
- Key Laboratory of Prevention, Diagnosis and Therapy of Upper Gastrointestinal Cancer of Zhejiang Province, Zhejiang, China
| | - Yun Li
- Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Zhejiang, China
| | - Xiaodan Guan
- Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Zhejiang, China
| | - Linfei Fan
- Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Zhejiang, China
| | - Jieer Ying
- Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Zhejiang, China
- Key Laboratory of Prevention, Diagnosis and Therapy of Upper Gastrointestinal Cancer of Zhejiang Province, Zhejiang, China
| |
Collapse
|
36
|
Xiao J, Wang L, Zhuang Y, Zhu Q, Li W, Liao H, Chen X, Liu Z. The deubiquitinase OTUB2 promotes cervical cancer growth through stabilizing FOXM1. Am J Transl Res 2024; 16:75-84. [PMID: 38322554 PMCID: PMC10839374] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2023] [Accepted: 12/06/2023] [Indexed: 02/08/2024]
Abstract
OBJECTIVES Ovarian tumor (OTU) domain-containing ubiquitin aldehyde-binding protein Otubain2 (OTUB2) is an important cysteine protease with deubiquitinase activity in the OTU family. However, the role of OTUB2 in cervical cancer (CC) has not been investigated. METHODS OTUB2 expression was analyzed employing the CC data from The Cancer Genome Atlas (TCGA) database. Western blot and qRT-PCR analysis were performed to identify OTUB2 expression in CC. The oncogenic function of OTUB2 was identified through a series of in vitro and in vivo experiments. Tandem Mass Tag™ Quantitative Proteomics examination was used to identify potential targets of OTUB2. RESULTS OTUB2 was overexpressed in CC and was related to poor prognosis of patients. In our in-house cohort, we also showed that OTUB2 was overexpressed in tumor tissues of CC compared to para-tumor. Knockdown of OTUB2 suppressed CC cell growth whereas OTUB2 upregulation fostered the proliferation of cancer cells. Forkhead box M1 (FOXM1) was found to be a target of OTUB2. FOXM1 can be positively regulated by OTUB2 in CC cells. In human CC tissues, protein level of FOXM1 was positively correlated with OTUB2. FOXM1 was found to play a critical role in OTUB2-mediated CC cell growth. Mechanistically, OTUB2 could bind FOXM1 and deubiquitinate FOXM1 to stabilize it. CONCLUSION OTUB2 promotes CC progression through deubiquitinating and stabilizing FOXM1.
Collapse
Affiliation(s)
- Jing Xiao
- Department of Reproductive Medicine Center, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang UniversityNanchang 330006, Jiangxi, PR China
- Department of Obstetrics and Gynecology, Jiangxi Maternal and Child Health HospitalNanchang 330006, Jiangxi, PR China
| | - Liqun Wang
- Department of Obstetrics and Gynecology, Jiangxi Maternal and Child Health HospitalNanchang 330006, Jiangxi, PR China
| | - Yuan Zhuang
- Department of Obstetrics and Gynecology, Jiangxi Maternal and Child Health HospitalNanchang 330006, Jiangxi, PR China
| | - Qizhou Zhu
- Department of Obstetrics and Gynecology, Jiangxi Maternal and Child Health HospitalNanchang 330006, Jiangxi, PR China
| | - Weimei Li
- Department of Obstetrics and Gynecology, Jiangxi Maternal and Child Health HospitalNanchang 330006, Jiangxi, PR China
| | - Hong Liao
- Department of Obstetrics and Gynecology, Jiangxi Maternal and Child Health HospitalNanchang 330006, Jiangxi, PR China
| | - Xiaofang Chen
- Department of Obstetrics and Gynecology, Jiangxi Maternal and Child Health HospitalNanchang 330006, Jiangxi, PR China
| | - Zhaoxia Liu
- Department of Reproductive Medicine Center, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang UniversityNanchang 330006, Jiangxi, PR China
| |
Collapse
|
37
|
Li Z, Li D, Yang T, Yao C. NAT10 promotes the tumorigenesis and progression of laryngeal squamous cell carcinoma through ac4C modification of FOXM1 mRNA. Cancer Biol Ther 2023; 24:2274143. [PMID: 37948132 PMCID: PMC10898813 DOI: 10.1080/15384047.2023.2274143] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2023] [Accepted: 10/18/2023] [Indexed: 11/12/2023] Open
Abstract
Laryngeal squamous cell carcinoma (LSCC), is a prevalent malignant tumor, belongs to the category of head and neck tumors. N-acetyltransferase 10 (NAT10) can alter mRNA stability through N4- acetylcytidine (ac4C) modification. This study aimed to make an investigation into the role of NAT10-mediated ac4C modification in the malignant processes of LSCC cells. The NAT10 expression in LSCC tissues and cells was detected RT-qPCR and western blot. The ac4C dot blot was performed to detect ac4C level. Besides, the cell viability, migration, and invasion abilities were detected by CCK-8 and transwell assays. AcRIP-qPCR was performed to measure the abundance of ac4C on FOXM1 mRNA. RIP and Luciferase reporter assays were performed to demonstrate the interaction between NAT10 and FOXM1. Finally, the xenograft model was established to explore the role of NAT10 in vivo. NAT1 levels were significantly increased in the LSCC tissues and cells. Knockdown of NAT10 could significantly suppress the proliferation, migration, and invasion of LSCC cells. Additionally, NAT10 recognized the ac4C-modified sites in the 3'-untranslated regions (3' UTR) of forkhead box M1 (FOXM1) to enhance the ability of FOXM1 mRNA. Furthermore, FOXM1 overexpression reversed the suppressing effects of NAT10 knockdown on the proliferation, migration, and invasion of LSCC cells, according to the results of rescue assays. Finally, results of animal experiments showed that NAT10 promoted in vivo tumorigenesis of LSCC cells through upregulating FOXM1. Our current study demonstrated that NAT10-mediated ac4C modification of FOXM1 mRNA promoted the malignant processes of LSCC cells.
Collapse
Affiliation(s)
- Zengpei Li
- Department of Otolaryngology, Nanyang First People’s Hospital, Nanyang, Henan, China
| | - Dajun Li
- Department of Otolaryngology, Nanyang First People’s Hospital, Nanyang, Henan, China
| | - Tianbin Yang
- Department of Otolaryngology, Nanyang First People’s Hospital, Nanyang, Henan, China
| | - Chen Yao
- Department of Otolaryngology, Nanyang First People’s Hospital, Nanyang, Henan, China
| |
Collapse
|
38
|
Wang P, Sun J, Sun C, Zhao H, Zhang Y, Chen J. BTF3 promotes proliferation and glycolysis in hepatocellular carcinoma by regulating GLUT1. Cancer Biol Ther 2023; 24:2225884. [PMID: 37382415 PMCID: PMC10312033 DOI: 10.1080/15384047.2023.2225884] [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: 01/08/2023] [Revised: 05/23/2023] [Accepted: 06/02/2023] [Indexed: 06/30/2023] Open
Abstract
Hepatocellular carcinoma (HCC) is a grievous tumor with an increasing incidence worldwide. Basic transcription factor 3 (BTF3) is discovered to regulate the expression of glucose transporter 1 (GLUT1), which benefits glycolysis, a momentous signature of tumors, through transactivation of the forkhead box M1 (FOXM1) expression. BTF3 is highly expressed in HCC. However, whether BTF3 promotes GLUT1 expression through FOXM1 to modulate glycolysis in HCC remains unclear. The expression profile of BTF3 were determined by online database, reverse transcription-quantitative polymerase chain reaction (RT-qPCR) and western blot. The role and mechanism of BTF3 in the proliferation and glycolysis of HCC cells were examined by cell counting kit-8 (CCK-8), 5-ethynyl-2'-deoxyuridine (EdU) incorporation, XF96 Extracellular Flux analyzer, spectrophotometry and western blot analysis. In addition, the direct interaction between BTF3 and FOXM1 was verified by dual-luciferase reporter and co-immunoprecipitation assays. Moreover, the role of BTF3 was also explored in a xenografted mice model. The expression of BTF3 was increased in HCC cells and tumor tissues. Knockdown of BTF3 reduced the cell viability, Edu positive cells, extracellular acidification rate (ECAR), glucose consumption and lactate production in both Huh7 and HCCLM3 cells. The expressions of FOXM1 and GLUT1 were increased in HCC tissues, which were positively correlated with the BTF3 expression. Moreover, a direct interaction existed between BTF3 and FOXM1 in HCC cells. Downregulation of BTF3 decreased the relative protein levels of FOXM1 and GLUT1, which were rescued with overexpression of FOXM1 in both cells. More importantly, overexpression of FOXM1 restored the cell viability, ECAR, glucose consumption and lactate production in both Huh7 and HCCLM3 cells transfected with siBTF3#1. Furthermore, inhibition of BTF3 decreased tumor weight and volume, and the relative level of BTF3, FOXM1, GLUT1 and Ki-67 in tumor tissues from mice xenografted with Huh7 cells. BTF3 enhanced the cell proliferation and glycolysis through FOXM1/GLUT1 axis in HCC.
Collapse
Affiliation(s)
- Peng Wang
- Department of Hepatobiliary and Pancreatic Surgery, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang, China
| | - Jianmin Sun
- Department of Hepatobiliary and Pancreatic Surgery, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang, China
| | - Chengming Sun
- Department of Hepatobiliary and Pancreatic Surgery, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang, China
| | - Haoran Zhao
- Department of Hepatobiliary and Pancreatic Surgery, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang, China
| | - YuBao Zhang
- Department of Hepatobiliary and Pancreatic Surgery, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang, China
| | - Jing Chen
- Department of Anesthesiology, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang, China
| |
Collapse
|
39
|
Fang K, Gong M, Liu D, Liang S, Li Y, Sang W, Zhu R. FOXM1/KIF20A axis promotes clear cell renal cell carcinoma progression via regulating EMT signaling and affects immunotherapy response. Heliyon 2023; 9:e22734. [PMID: 38125441 PMCID: PMC10730723 DOI: 10.1016/j.heliyon.2023.e22734] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2023] [Revised: 10/27/2023] [Accepted: 11/17/2023] [Indexed: 12/23/2023] Open
Abstract
Background The correlation between FOXM1 and KIF20A has not been revealed in clear cell renal cell carcinoma (ccRCC). Methods Public data was downloaded from The Cancer Genome Atlas (TCGA) database. R software was utilized for the execution of bioinformatic analysis. The expression levels of specific molecules (mRNA and protein) were detected using real-time quantitative PCR (qRT-PCR) and Western blot assays. The capacity of cell growth was assessed by employing CCK8 and colony formation assay. Cell invasion and migration ability were assessed using transwell assay. Results In our study, we illustrated the association between FOXM1 and KIF20A. Our results indicated that both FOXM1 and KIF20A were associated with poor prognosis and clinical performance. The malignant characteristics of ccRCC cells can be significantly suppressed by inhibiting FOXM1 and KIF20A, as demonstrated by in vitro experiments. Moreover, we found that FOXM1 can upregulate KIF20A. Then, EMT signaling was identified as the underlying pathway FOXM1 and KIF20A are involved. WB results indicated that FOXM1/KIF20A axis can activate EMT signaling. Moreover, we noticed that FOXM1 and KIF20A can affect the immunotherapy response and immune microenvironment of ccRCC patients. Conclusions Our results identified the role of the FOXM1/KIF20A axis in ccRCC progression and immunotherapy, making it the underlying target for ccRCC.
Collapse
Affiliation(s)
- Kai Fang
- Department of Urology, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai, China
| | - Min Gong
- Department of Urology, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai, China
| | - Dong Liu
- Department of Urology, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai, China
| | - Shengjie Liang
- Department of Urology, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai, China
| | - Yang Li
- Department of Urology, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai, China
| | - Weicong Sang
- Department of Urology, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai, China
| | - Rujian Zhu
- Department of Urology, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai, China
| |
Collapse
|
40
|
Liu ZY, Li YH, Li BW, Xin L. Development and validation of a vesicle-mediated transport-associated gene signature for predicting prognosis and immune therapy response in hepatocellular carcinoma. J Cancer Res Clin Oncol 2023; 149:13211-13230. [PMID: 37479759 DOI: 10.1007/s00432-023-05079-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2023] [Accepted: 06/29/2023] [Indexed: 07/23/2023]
Abstract
BACKGROUND Hepatocellular carcinoma (HCC) is a malignant tumor with a poor prognosis. The progression of numerous malignancies has been linked to abnormal vesicle-mediated transport-related gene (VMTRG) expression. The prognostic importance of VMTRGs in HCC is uncertain nonetheless. METHODS Utilizing HCC data from TCGA and ICGC, we employed univariate cox analysis, unsupervised clustering, and lasso analysis to construct molecular subtypes and prognostic signature of HCC based on the prognostic-associated VMTRGs expression levels. Subsequently, we validated the expression levels of the signature genes. We investigated the probable pathways using gene set variation analysis (GSVA) and gene set enrichment analysis (GSEA). Six methods were utilized to compare immune cell infiltration between two risk groups. Moreover, the "pRRophetic" algorithm was utilized to test the drug sensitivity of both groups. RESULTS We identified two distinct subtypes with divergent biological behaviors and immune functionality through unsupervised clustering. Subtype C1 demonstrated a poorer prognosis. A prognostic signature incorporating two VMTRGs (KIF2C and RAC1) was formulated. Immunohistochemistry and qRT-PCR analyses unveiled a significant upregulation of these pivotal genes within HCC tissues. The prognosis was worse for the high-risk group, which also had a higher clinicopathological grade, higher levels of tumor mutation burden (TMB), a higher immunological infiltration of CD8 + T cells, a higher expression of immune checkpoints, and enhanced immunotherapy efficacy. These two risk groups also have varied chemotherapy drug sensitivities. CONCLUSIONS Based on VMTRGs, we have developed a signature that assists in accurate prognosis prediction and formulating personalized treatment strategies for HCC patients.
Collapse
Affiliation(s)
- Zhi-Yang Liu
- Department of General Surgery, The Second Affiliated Hospital of Nanchang University, No. 1 Minde Road, Donghu District, Nanchang, 330006, Jiangxi, China
| | - Yi-He Li
- Department of Urology, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Bo-Wen Li
- Department of General Surgery, The Second Affiliated Hospital of Nanchang University, No. 1 Minde Road, Donghu District, Nanchang, 330006, Jiangxi, China
| | - Lin Xin
- Department of General Surgery, The Second Affiliated Hospital of Nanchang University, No. 1 Minde Road, Donghu District, Nanchang, 330006, Jiangxi, China.
| |
Collapse
|
41
|
Bai J, Li Y, Cai L. Clinical implications of forkhead box M1, cyclooxygenase-2 , and glucose-regulated protein 78 in breast invasive ductal carcinoma. World J Clin Cases 2023; 11:7284-7293. [PMID: 37969442 PMCID: PMC10643068 DOI: 10.12998/wjcc.v11.i30.7284] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/15/2023] [Revised: 09/11/2023] [Accepted: 09/18/2023] [Indexed: 10/25/2023] Open
Abstract
BACKGROUND Breast infiltrating ductal carcinoma (BIDC) represents the largest heterotypic tumor group, and an in-depth understanding of the pathogenesis of BIDC is key to improving its prognosis. AIM To analyze the expression profiles and clinical implications of forkhead box M1 (FOXM1), cyclooxygenase-2 (COX-2), and glucose-regulated protein 78 (GRP78) in BIDC. METHODS A total of 65 BIDC patients and 70 healthy controls who presented to our hospital between August 2019 and May 2021 were selected for analysis. The peripheral blood FOXM1, COX-2, and GRP78 levels in both groups were measured and the association between their expression profiles in BIDC was examined. Additionally, we investigated the diagnostic value of FOXM1, COX-2, and GRP78 in patients with BIDC and their correlations with clinicopathological features. Furthermore, BIDC patients were followed for 1 year to identify factors influencing patient prognosis. RESULTS The levels of FOXM1, COX-2, and GRP78 were significantly higher in BIDC patients compared to healthy controls (P < 0.05), and a positive correlation was observed among them (P < 0.05). Receiver operating characteristic analysis demonstrated that FOXM1, COX-2, and GRP78 had excellent diagnostic value in predicting the occurrence of BIDC (P < 0.05). Subsequently, we found significant differences in FOXM1, COX-2, and GRP78 levels among patients with different histological grades and metastasis statuses (with vs without) (P < 0.05). Cox analysis revealed that FOXM1, COX-2, GRP78, increased histological grade, and the presence of tumor metastasis were independent risk factors for prognostic death in BIDC (P < 0.001). CONCLUSION FOXM1, COX-2, and GRP78 exhibit abnormally high expression in BIDC, promoting malignant tumor development and closely correlating with prognosis. These findings hold significant research implications for the future diagnosis and treatment of BIDC.
Collapse
Affiliation(s)
- Jie Bai
- Department of Clinical Laboratory, Joint Logistics Support Unit 940 Hospital, Lanzhou 730030, Gansu Province, China
| | - Ying Li
- Department of Breast Surgery, The Fourth Hospital of Shijiazhuang, Shijiazhuang 050032, Hebei Province, China
| | - Li Cai
- Department of Pathology, Huai’an Maternal and Child Health Care Center, Huai’an 223002, Jiangsu Province, China
| |
Collapse
|
42
|
Kong J, Xu S, Deng Z, Wang Y, Zhang P. Transcription factor FOXM1 promotes hepatocellular carcinoma malignant progression through activation of the WNT pathway by binding to SETDB1. Tissue Cell 2023; 84:102186. [PMID: 37556918 DOI: 10.1016/j.tice.2023.102186] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Revised: 07/25/2023] [Accepted: 07/27/2023] [Indexed: 08/11/2023]
Abstract
BACKGROUND FOXM1 is a transcription factor confirmed by studies to promote the development of hepatocellular carcinoma (HCC) and various other cancers, yet the molecular mechanism remains rather enigmatic. This study attempted to unveil the function and regulatory mechanism of FOXM1 in the progression of HCC. METHODS Bioinformatics methods first analyzed the expression of FOXM1 in HCC tissues and then screened target genes downstream of FOXM1. Possible pathways of the target gene were specified through Gene Set Enrichment Analysis (GSEA). After using qRT-PCR to measure the expression of FOXM1 and its downstream regulatory gene SETDB1 in HCC tissues, ChIP and dual-luciferase assays were employed and verified the binding relationship between FOXM1 and the promoter of SETDB1. Then the effects of the FOXM1/SETDB1/Wnt pathway on the proliferation, migration, and invasion of HCC cells were profiled by CCK-8, colony formation, wound healing, and transwell assays. WNT and EMT-related protein expression levels were detected by western blot and immunofluorescence assay, respectively. RESULTS The bioinformatics prediction showed that SETDB1 was the target downstream of FOXM1, and their binding relationship was verified by ChIP and dual-luciferase assays. Cell experiments showed that FOXM1 could enhance the proliferative, migratory, and invasive abilities of HCC cells through binding to SETDB1. Rescue assay suggested that the activation of key genes of the WNT pathway and EMT-related genes were part of the regulatory mechanism that FOXM1 bound to SETDB1. CONCLUSION This study found that FOXM1 could bind with SETDB1 and hence activate the WNT signaling pathway to promote the malignant progression of HCC. It indicated that FOXM1 could be the possible target for treating HCC.
Collapse
Affiliation(s)
- Jianqiao Kong
- Department of General Surgery, Xiangyang No.1 People's Hospital, Hubei University of Medicine, Xiangyang City, China
| | - Song Xu
- Department of General Surgery, Xiangyang No.1 People's Hospital, Hubei University of Medicine, Xiangyang City, China
| | - Zhongming Deng
- Department of General Surgery, Xiangyang No.1 People's Hospital, Hubei University of Medicine, Xiangyang City, China
| | - Yi Wang
- Department of General Surgery, Xiangyang No.1 People's Hospital, Hubei University of Medicine, Xiangyang City, China.
| | - Peng Zhang
- Department of General Surgery, Xiangyang No.1 People's Hospital, Hubei University of Medicine, Xiangyang City, China.
| |
Collapse
|
43
|
Liu X, Zhao J, Dong P, Du X, Lu W, Feng Y, Wang L. TRIM6 silencing for inhibiting growth and angiogenesis of gliomas by regulating VEGFA. J Chem Neuroanat 2023; 132:102291. [PMID: 37236551 DOI: 10.1016/j.jchemneu.2023.102291] [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: 02/09/2023] [Revised: 05/22/2023] [Accepted: 05/23/2023] [Indexed: 05/28/2023]
Abstract
Gliomas are the highest prevalent primary central nervous system (CNS) cancers with poor overall survival rate. There is an urgent need to conduct more research into molecular therapies targeting critical elements of gliomas. This study herein targeted to assess the impact of tripartite motif protein 6 (TRIM6) on gliomas. Using public databases, we found the increased TRIM6 expression in tissues of glioma which was linked with worst overall survival. Silencing TRIM6 promoted glioma cell proliferation, migration and angiogenesis, suggesting the promoting effects of TRIM6 on gliomas. Knockdown of TRIM6 expression downregulated the expression levels of Forkhead box M1 (FOXM1) and vascular endothelial growth factor A (VEGFA) in glioma cells. Afterwards, impact of TRIM6 on VEGFA expression was regulated by FOXM1. VEGFA overexpression reversed the decreased abilities of glioma cell proliferation, migration and angiogenesis caused by silencing TRIM6. Furthermore, we also found that TRIM6 promoted the growth of gliomas in the xenograft mouse model. In summary, the expression of TRIM6 was increased which was related to poor prognosis of glioma patients. TRIM6 promoted glioma cell proliferation, migration and angiogenesis through the FOXM1-VEGFA pathway. Therefore, TRIM6 carries capacity to be explored as a novel therapeutic target in clinical.
Collapse
Affiliation(s)
- Xin Liu
- Department of Neurosurgery, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei 050000, China
| | - Junling Zhao
- Department of Neurosurgery, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei 050000, China
| | - PengFei Dong
- Department of Neurosurgery, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei 050000, China
| | - Xinyuan Du
- Department of Neurosurgery, JingXing Chinese Medicne Hospital, Shijiazhuang, Hebei 050000, China
| | - Wenpeng Lu
- Department of Neurosurgery, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei 050000, China
| | - Yan Feng
- Department of Neurosurgery, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei 050000, China
| | - Liqun Wang
- Department of Neurosurgery, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei 050000, China.
| |
Collapse
|
44
|
Aimaier R, Chung MH, Gu Y, Yu Q, Wei C, Li H, Guo Z, Long M, Li Y, Wang W, Li Q, Wang Z. FOXM1 promotes neurofibromatosis type 1-associated malignant peripheral nerve sheath tumor progression in a NUF2-dependent manner. Cancer Gene Ther 2023; 30:1390-1402. [PMID: 37488294 DOI: 10.1038/s41417-023-00645-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2022] [Revised: 06/23/2023] [Accepted: 07/04/2023] [Indexed: 07/26/2023]
Abstract
Malignant peripheral nerve sheath tumors (MPNSTs) are aggressive soft-tissue sarcomas characterized by poor prognosis and low drug response rates. Traditional chemo/radiotherapies show only mild benefits for patients with MPNSTs, and no targeted therapy is available in the clinic. A better understanding of the molecular background of MPNSTs is critical for the development of effective targeted therapies. Forkhead box M1 (FOXM1) has been implicated in the progression of many human malignancies, though its role in MPNSTs is unclear. In this study, using four Gene Expression Omnibus (GEO) datasets and a tissue microarray, we demonstrated that FOXM1 upregulation was associated with poor prognosis in patients with MPNSTs. FOXM1 overexpression and knockdown regulated the proliferation and colony formation of MPNST cells. Using bioinformatics analysis and luciferase reporter assays, we identified NUF2 as a direct downstream target of FOXM1. Both in vitro and in vivo experiments demonstrated that the induction of MPNST cell proliferation by FOXM1 was dependent on elevated NUF2 expression, as NUF2 knockdown abolished the FOXM1-induced proliferation of MPNST cells. Our study showed that the FOXM1-NUF2 axis mediates human MPNST progression and could be a potential therapeutic target.
Collapse
Affiliation(s)
- Rehanguli Aimaier
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Man-Hon Chung
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yihui Gu
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Qingxiong Yu
- Department of Facial Plastic and Reconstructive Surgery, Eye & ENT Hospital, Fudan University, Shanghai, China
| | - Chengjiang Wei
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Haibo Li
- Department of Plastic Surgery, The Third Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Zizhen Guo
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Manmei Long
- Department of Pathology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yuehua Li
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Wei Wang
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Qingfeng Li
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Zhichao Wang
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| |
Collapse
|
45
|
Tang S, Wu Z, Chen L, She L, Zuo W, Luo W, Zhang Y, Liang S, Liu G, He B, He J, Zhang N. Multi-omics analysis reveals the association between elevated KIF18B expression and unfavorable prognosis, immune evasion, and regulatory T cell activation in nasopharyngeal carcinoma. Front Immunol 2023; 14:1258344. [PMID: 37744335 PMCID: PMC10514916 DOI: 10.3389/fimmu.2023.1258344] [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: 07/13/2023] [Accepted: 08/25/2023] [Indexed: 09/26/2023] Open
Abstract
Background Nasopharyngeal carcinoma (NPC) is prevalent in Southern China. The expression profile and functions of kinesin family member 18B (KIF18B) remain unclear in NPC. Methods Bulk and single-cell transcriptome data for NPC were downloaded. KIF18B expression differences in NPC and normal tissues and its prognostic value were validated by immunohistochemistry and Cox model. We performed multi-faceted functional enrichment analysis on KIF18B. Immune infiltration was analyzed comprehensively by the CIBERSORT, EPIC, and quanTIseq algorithms and the BisqueRNA package and confirmed by immunofluorescence assay. The intercellular communication were investigated by the CellChat package. We explored the dynamics of KIF18B expression by pseudotime trajectory. M6A modification analysis rely on SRAMP platform. The treatment response were evaluated by Tumor Immune Dysfunction and Exclusion (TIDE) score, immunophenoscore and IC50 value. Results KIF18B overexpression in NPC led to unfavorable prognosis, and significantly associated with advanced T, N, and stage classifications. Functional analysis demonstrated that KIF18B was involved in immune suppression, epithelial-mesenchymal transition (EMT), N6-methyladenosine (m6A) modification and therapeutic responses. The deconvolution algorithm indicated that activated regulatory T cells (Tregs) had the strongest positive correlation with KIF18B among immune cells (R = 0.631). Validated by immunofluorescence assay, the high KIF18B expression group displayed a notable rise in Tregs infiltration, accompanied by a substantial decrease in the infiltration of CD8+ T cells and macrophages. In the intercellular communication network, malignant cells with high KIF18B expression implicated in more interactions, and activated and recruited Tregs by modulating cytokines, chemokines, and immune checkpoints. KIF18B was upregulated in more advanced malignant cells and influenced EMT by regulating ITGA6, VIM, and ZEB1/2. KIF18B expression was positively related to m6A "writer" and "reader" genes, and negatively related to "eraser" genes. The KIF18B high expression group exhibited a higher TIDE score and elevated IC50 values for the commonly used chemotherapy drugs, gemcitabine, oxaliplatin, and 5-fluorouracil. Conclusion KIF18B is a significant prognostic marker in NPC, and may modulate immune evasion and EMT. M6A modification may account for the aberrant overexpression of KIF18B in NPC. Furthermore, KIF18B may predict response to immunotherapy and chemotherapy.
Collapse
Affiliation(s)
- Siqi Tang
- Department of Radiation Oncology, First People’s Hospital of Foshan, Foshan, China
| | - Zhenyu Wu
- Department of Urology, First People’s Hospital of Foshan, Foshan, China
| | - Lusi Chen
- Department of Radiation Oncology, First People’s Hospital of Foshan, Foshan, China
| | - Longjiang She
- Department of Radiation Oncology, First People’s Hospital of Foshan, Foshan, China
| | - Weihan Zuo
- Department of Radiation Oncology, First People’s Hospital of Foshan, Foshan, China
| | - Weijun Luo
- Department of Radiation Oncology, First People’s Hospital of Foshan, Foshan, China
| | - Yang Zhang
- Department of Radiation Oncology, First People’s Hospital of Foshan, Foshan, China
| | - Shaoqiang Liang
- Department of Comprehensive (Head and Neck) Oncology and Hospice Ward, First People’s Hospital of Foshan, Foshan, China
| | - Guichao Liu
- Department of Radiation Oncology, First People’s Hospital of Foshan, Foshan, China
| | - Biyi He
- Department of Rhinology, First People’s Hospital of Foshan, Foshan, China
| | - Jinfeng He
- Department of Rhinology, First People’s Hospital of Foshan, Foshan, China
| | - Ning Zhang
- Department of Radiation Oncology, First People’s Hospital of Foshan, Foshan, China
| |
Collapse
|
46
|
He H, Zhou J, Cheng F, Li H, Quan Y. MiR-3677-3p promotes development and sorafenib resistance of hepatitis B-related hepatocellular carcinoma by inhibiting FOXM1 ubiquitination. Hum Cell 2023; 36:1773-1789. [PMID: 37402927 DOI: 10.1007/s13577-023-00945-z] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2022] [Accepted: 06/17/2023] [Indexed: 07/06/2023]
Abstract
Being encoded by hepatitis B, hepatitis B X (HBx) protein plays crucial roles in hepatitis B-related hepatocellular carcinoma (HCC) occurrence, development, and metastasis. miRNAs also function in the progression of hepatitis B-related HCC. Hence, the objective of this study was to explore the impacts of miR-3677-3p on tumor progression and sorafenib resistance in hepatitis B-related HCC and the related underlying mechanisms. Our research revealed that miR-3677-3p and FOXM1 was up-regulated and FBXO31 was down-regulated in HBV+ HCC cells and tumor tissues from nude mice. After miR-3677-3p overexpression, cell proliferative, invasive, and migrating potentials and stemness-related protein (CD133, EpCAM, and OCT4) levels were enhanced, and cell apoptosis was reduced in Huh7 + HBx/SR cells and HepG2.2.15/SR cells. Besides, miR-3677-3p promoted the drug resistance of Huh7 + HBx/SR cells and HepG2.2.15/SR cells to sorafenib and lifted IC50. In vivo experiments, miR-3677-3p down-regulation suppressed the tumor growth in the hepatitis B HCC nude mouse models. Mechanistically, miR-3677-3p targeted and negatively-regulated FBXO31 and FBXO31 could enrich FOXM1 protein. miR-3677-3p down-regulation or FBXO31 overexpression promoted the ubiquitylation of FOXM1. In a word, miR-3677-3p bound to FBXO31 and inhibited FBXO31 expression, thus curtailing the ubiquitylation degradation of FOXM1, contributing to HCC development and sorafenib resistance.
Collapse
Affiliation(s)
- Hengzheng He
- Department of Minimally Invasive Surgery, The Brain Hospital of Hunan Province, Changsha, 410007, Hunan, People's Republic of China
| | - Jian Zhou
- Department of Quality Control, The Brain Hospital of Hunan Province, Changsha, 410007, Hunan, People's Republic of China
| | - Fahui Cheng
- Department of Minimally Invasive Surgery, The Brain Hospital of Hunan Province, Changsha, 410007, Hunan, People's Republic of China
| | - Huijuan Li
- Department of Gastroenterology, The Brain Hospital of Hunan Province, No.427, Sec.3, Furong Mid Road, Changsha, 410007, Hunan, People's Republic of China
| | - Yangya Quan
- Department of Gastroenterology, The Brain Hospital of Hunan Province, No.427, Sec.3, Furong Mid Road, Changsha, 410007, Hunan, People's Republic of China.
| |
Collapse
|
47
|
Jia R, Che X, Jia J, Guo J. FOXM1a Isoform of Oncogene FOXM1 Is a Tumor Suppressor Suppressed by hnRNP C in Oral Squamous Cell Carcinoma. Biomolecules 2023; 13:1331. [PMID: 37759731 PMCID: PMC10526205 DOI: 10.3390/biom13091331] [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: 07/18/2023] [Revised: 08/24/2023] [Accepted: 08/29/2023] [Indexed: 09/29/2023] Open
Abstract
FOXM1 is an oncogenic transcriptional factor and includes several isoforms generated by alternative splicing. Inclusion of alternative exon 9 produces FOXM1a, a transcriptionally inactive isoform. However, the role of FOXM1a in tumorigenesis remains unknown. In addition, the regulatory mechanisms of exon 9 splicing are also unclear. In the present study, we found that overexpression of FOXM1a significantly reduced cell proliferation and colony formation of oral squamous cell carcinoma (OSCC) cell proliferation in vitro. Importantly, OSCC cells with FOXM1a overexpression showed significantly slower tumor formation in nude mice. Moreover, we identified a U-rich exonic splicing suppressor (ESS) which is responsible for exon 9 skipping. Splicing factor heterogeneous nuclear ribonucleoprotein C (hnRNP C) can bind to the ESS and suppress exon 9 inclusion and FOXM1a expression. Silence of hnRNP C also significantly suppresses OSCC cell proliferation. HnRNP C is significantly co-expressed with FOXM1 in cancers. Our study uncovered a novel regulatory mechanism of oncogene FOXM1 expression in OSCC.
Collapse
Affiliation(s)
- Rong Jia
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan 430072, China; (R.J.); (X.C.)
| | - Xiaoxuan Che
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan 430072, China; (R.J.); (X.C.)
| | - Jun Jia
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan 430072, China; (R.J.); (X.C.)
- Department of Oral and Maxillofacial Surgery, School & Hospital of Stomatology, Wuhan University, Wuhan 430072, China
| | - Jihua Guo
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan 430072, China; (R.J.); (X.C.)
- Department of Endodontics, School & Hospital of Stomatology, Wuhan University, Wuhan 430072, China
| |
Collapse
|
48
|
Gupta R, Kadhim MM, Turki Jalil A, Obayes AM, Aminov Z, Alsaikhan F, Ramírez-Coronel AA, Ramaiah P, Tayyib NA, Luo X. Multifaceted role of NF-κB in hepatocellular carcinoma therapy: Molecular landscape, therapeutic compounds and nanomaterial approaches. ENVIRONMENTAL RESEARCH 2023; 228:115767. [PMID: 36966991 DOI: 10.1016/j.envres.2023.115767] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/04/2023] [Revised: 03/20/2023] [Accepted: 03/22/2023] [Indexed: 05/16/2023]
Abstract
The predominant kind of liver cancer is hepatocellular carcinoma (HCC) that its treatment have been troublesome difficulties for physicians due to aggressive behavior of tumor cells in proliferation and metastasis. Moreover, stemness of HCC cells can result in tumor recurrence and angiogenesis occurs. Another problem is development of resistance to chemotherapy and radiotherapy in HCC cells. Genomic mutations participate in malignant behavior of HCC and nuclear factor-kappaB (NF-κB) has been one of the oncogenic factors in different human cancers that after nuclear translocation, it binds to promoter of genes in regulating their expression. Overexpression of NF-κB has been well-documented in increasing proliferation and invasion of tumor cells and notably, when its expression enhances, it induces chemoresistance and radio-resistance. Highlighting function of NF-κB in HCC can shed some light on the pathways regulating progression of tumor cells. The first aspect is proliferation acceleration and apoptosis inhibition in HCC cells mediated by enhancement in expression level of NF-κB. Moreover, NF-κB is able to enhance invasion of HCC cells via upregulation of MMPs and EMT, and it triggers angiogenesis as another step for increasing spread of tumor cells in tissues and organs. When NF-κB expression enhances, it stimulates chemoresistance and radio-resistance in HCC cells and by increasing stemness and population of cancer-stem cells, it can provide the way for recurrence of tumor. Overexpression of NF-κB mediates therapy resistance in HCC cells and it can be regulated by non-coding RNAs in HCC. Moreover, inhibition of NF-κB by anti-cancer and epigenetic drugs suppresses HCC tumorigenesis. More importantly, nanoparticles are considered for suppressing NF-κB axis in cancer and their prospectives and results can also be utilized for treatment of HCC. Nanomaterials are promising factors in treatment of HCC and by delivery of genes and drugs, they suppress HCC progression. Furthermore, nanomaterials provide phototherapy in HCC ablation.
Collapse
Affiliation(s)
- Reena Gupta
- Institute of Pharmaceutical Research, GLA University, District-Mathura, U. P., India
| | - Mustafa M Kadhim
- Department of Dentistry, Kut University College, Kut, Wasit, 52001, Iraq; Medical Laboratory Techniques Department, Al-Farahidi University, Baghdad, 10022, Iraq
| | - Abduladheem Turki Jalil
- Medical Laboratories Techniques Department, Al-Mustaqbal University College, Babylon, Hilla, 51001, Iraq.
| | | | - Zafar Aminov
- Department of Public Health and Healthcare Management, Samarkand State Medical University, 18 Amir Temur Street, Samarkand, Uzbekistan; Department of Scientific Affairs, Tashkent State Dental Institute, 103 Makhtumkuli Str., Tashkent, Uzbekistan
| | - Fahad Alsaikhan
- College of Pharmacy, Prince Sattam Bin Abdulaziz University, Alkharj, Saudi Arabia.
| | - Andrés Alexis Ramírez-Coronel
- Azogues Campus Nursing Career, Health and Behavior Research Group (HBR), Psychometry and Ethology Laboratory, Catholic University of Cuenca, Ecuador; Epidemiology and Biostatistics Research Group, CES University, Colombia; Educational Statistics Research Group (GIEE), National University of Education, Ecuador
| | | | - Nahla A Tayyib
- Faculty of Nursing, Umm al- Qura University, Makkah, Saudi Arabia
| | - Xuanming Luo
- Department of General Surgery, Shanghai Xuhui Central Hospital, Fudan University, Shanghai, 200031, China.
| |
Collapse
|
49
|
Asada H, Tani A, Sakuma H, Hirabayashi M, Matsumoto Y, Watanabe K, Tsuboi M, Yoshida S, Harada K, Uchikai T, Goto-Koshino Y, Chambers JK, Ishihara G, Kobayashi T, Irie M, Uchida K, Ohno K, Bonkobara M, Tsujimoto H, Tomiyasu H. Whole exome and transcriptome analysis revealed the activation of ERK and Akt signaling pathway in canine histiocytic sarcoma. Sci Rep 2023; 13:8512. [PMID: 37231193 DOI: 10.1038/s41598-023-35813-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2022] [Accepted: 05/24/2023] [Indexed: 05/27/2023] Open
Abstract
Histiocytic sarcoma (HS) is an incurable aggressive tumor, and no consensus has been made on the treatment due to its rare occurrence. Since dogs spontaneously develop the disease and several cell lines are available, they have been advocated as translational animal models. In the present study, therefore, we explored gene mutations and aberrant molecular pathways in canine HS by next generation sequencing to identify molecular targets for treatment. Whole exome sequencing and RNA-sequencing revealed gene mutations related to receptor tyrosine kinase pathways and activation of ERK1/2, PI3K-AKT, and STAT3 pathways. Analysis by quantitative PCR and immunohistochemistry revealed that fibroblast growth factor receptor 1 (FGFR1) is over-expressed. Moreover, activation of ERK and Akt signaling were confirmed in all HS cell lines, and FGFR1 inhibitors showed dose-dependent growth inhibitory effects in two of the twelve canine HS cell lines. The findings obtained in the present study indicated that ERK and Akt signaling were activated in canine HS and drugs targeting FGFR1 might be effective in part of the cases. The present study provides translational evidence that leads to establishment of novel therapeutic strategies targeting ERK and Akt signaling in HS patients.
Collapse
Affiliation(s)
- Hajime Asada
- Department of Veterinary Internal Medicine, the University of Tokyo, Bunkyo-ku, Tokyo, Japan
- Department of Obstetrics and Gynecology, The University of Chicago, Chicago, IL, 60637, USA
| | - Akiyoshi Tani
- Department of Veterinary Internal Medicine, the University of Tokyo, Bunkyo-ku, Tokyo, Japan
| | - Hiroki Sakuma
- Department of Veterinary Internal Medicine, the University of Tokyo, Bunkyo-ku, Tokyo, Japan
| | - Miyuki Hirabayashi
- Department of Veterinary Pathology, the University of Tokyo, Bunkyo-ku, Tokyo, Japan
| | - Yuki Matsumoto
- Anicom Specialty Medical Institute Inc., Shinjuku-ku, Tokyo, Japan
| | - Kei Watanabe
- Anicom Specialty Medical Institute Inc., Shinjuku-ku, Tokyo, Japan
| | - Masaya Tsuboi
- Veterinary Medical Center, the University of Tokyo, Bunkyo-ku, Tokyo, Japan
| | - Shino Yoshida
- Department of Veterinary Internal Medicine, the University of Tokyo, Bunkyo-ku, Tokyo, Japan
| | - Kei Harada
- Japan Small Animal Cancer Center, Tokorozawa, Saitama, Japan
| | - Takao Uchikai
- Anicom Specialty Medical Institute Inc., Shinjuku-ku, Tokyo, Japan
| | - Yuko Goto-Koshino
- Department of Veterinary Internal Medicine, the University of Tokyo, Bunkyo-ku, Tokyo, Japan
| | - James K Chambers
- Department of Veterinary Pathology, the University of Tokyo, Bunkyo-ku, Tokyo, Japan
| | - Genki Ishihara
- Anicom Specialty Medical Institute Inc., Shinjuku-ku, Tokyo, Japan
| | | | - Mitsuhiro Irie
- Shikoku Veterinary Medical Center, Kita-gun, Kagawa, Japan
| | - Kazuyuki Uchida
- Department of Veterinary Pathology, the University of Tokyo, Bunkyo-ku, Tokyo, Japan
| | - Koichi Ohno
- Department of Veterinary Internal Medicine, the University of Tokyo, Bunkyo-ku, Tokyo, Japan
| | - Makoto Bonkobara
- Department of Veterinary Clinical Pathology, Nippon Veterinary and Life Science University, Musashino, Tokyo, Japan
| | - Hajime Tsujimoto
- Department of Veterinary Internal Medicine, the University of Tokyo, Bunkyo-ku, Tokyo, Japan
| | - Hirotaka Tomiyasu
- Department of Veterinary Internal Medicine, the University of Tokyo, Bunkyo-ku, Tokyo, Japan.
| |
Collapse
|
50
|
Nong S, Han X, Xiang Y, Qian Y, Wei Y, Zhang T, Tian K, Shen K, Yang J, Ma X. Metabolic reprogramming in cancer: Mechanisms and therapeutics. MedComm (Beijing) 2023; 4:e218. [PMID: 36994237 PMCID: PMC10041388 DOI: 10.1002/mco2.218] [Citation(s) in RCA: 43] [Impact Index Per Article: 21.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Revised: 01/22/2023] [Accepted: 01/30/2023] [Indexed: 03/29/2023] Open
Abstract
Cancer cells characterized by uncontrolled growth and proliferation require altered metabolic processes to maintain this characteristic. Metabolic reprogramming is a process mediated by various factors, including oncogenes, tumor suppressor genes, changes in growth factors, and tumor-host cell interactions, which help to meet the needs of cancer cell anabolism and promote tumor development. Metabolic reprogramming in tumor cells is dynamically variable, depending on the tumor type and microenvironment, and reprogramming involves multiple metabolic pathways. These metabolic pathways have complex mechanisms and involve the coordination of various signaling molecules, proteins, and enzymes, which increases the resistance of tumor cells to traditional antitumor therapies. With the development of cancer therapies, metabolic reprogramming has been recognized as a new therapeutic target for metabolic changes in tumor cells. Therefore, understanding how multiple metabolic pathways in cancer cells change can provide a reference for the development of new therapies for tumor treatment. Here, we systemically reviewed the metabolic changes and their alteration factors, together with the current tumor regulation treatments and other possible treatments that are still under investigation. Continuous efforts are needed to further explore the mechanism of cancer metabolism reprogramming and corresponding metabolic treatments.
Collapse
Affiliation(s)
- Shiqi Nong
- State Key Laboratory of Oral DiseasesWest China Hospital of StomatologyWest China School of StomatologyNational Clinical Research Center for Oral DiseasesSichuan UniversityChengduSichuanChina
| | - Xiaoyue Han
- State Key Laboratory of Oral DiseasesWest China Hospital of StomatologyWest China School of StomatologyNational Clinical Research Center for Oral DiseasesSichuan UniversityChengduSichuanChina
| | - Yu Xiang
- Department of BiotherapyCancer CenterWest China HospitalSichuan UniversityChengduSichuanChina
| | - Yuran Qian
- State Key Laboratory of Oral DiseasesWest China Hospital of StomatologyWest China School of StomatologyNational Clinical Research Center for Oral DiseasesSichuan UniversityChengduSichuanChina
| | - Yuhao Wei
- Department of Clinical MedicineWest China School of MedicineWest China HospitalSichuan UniversityChengduSichuanChina
| | - Tingyue Zhang
- State Key Laboratory of Oral DiseasesWest China Hospital of StomatologyWest China School of StomatologyNational Clinical Research Center for Oral DiseasesSichuan UniversityChengduSichuanChina
| | - Keyue Tian
- State Key Laboratory of Oral DiseasesWest China Hospital of StomatologyWest China School of StomatologyNational Clinical Research Center for Oral DiseasesSichuan UniversityChengduSichuanChina
| | - Kai Shen
- Department of OncologyFirst Affiliated Hospital of Nanjing Medical UniversityNanjingChina
| | - Jing Yang
- State Key Laboratory of Oncology in South ChinaCollaborative Innovation Center for Cancer MedicineSun Yat‐sen University Cancer CenterGuangzhouChina
| | - Xuelei Ma
- State Key Laboratory of Oral DiseasesWest China Hospital of StomatologyWest China School of StomatologyNational Clinical Research Center for Oral DiseasesSichuan UniversityChengduSichuanChina
- Department of Biotherapy and Cancer CenterState Key Laboratory of BiotherapyCancer CenterWest China HospitalSichuan UniversityChengduSichuanChina
| |
Collapse
|