1
|
Bai Y, Deng X, Chen D, Han S, Lin Z, Li Z, Tong W, Li J, Wang T, Liu X, Liu Z, Cui Z, Zhang Y. Integrative analysis based on ATAC-seq and RNA-seq reveals a novel oncogene PRPF3 in hepatocellular carcinoma. Clin Epigenetics 2024; 16:154. [PMID: 39501301 PMCID: PMC11539654 DOI: 10.1186/s13148-024-01769-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2024] [Accepted: 10/25/2024] [Indexed: 11/08/2024] Open
Abstract
BACKGROUND Assay of Transposase Accessible Chromatin Sequencing (ATAC-seq) is a high-throughput sequencing technique that detects open chromatin regions across the genome. These regions are critical in facilitating transcription factor binding and subsequent gene expression. Herein, we utilized ATAC-seq to identify key molecular targets regulating the development and progression of hepatocellular carcinoma (HCC) and elucidate the underlying mechanisms. METHODS We first compared chromatin accessibility profiles between HCC and normal tissues. Subsequently, RNA-seq data was employed to identify differentially expressed genes (DEGs). Integrating ATAC-seq and RNA-seq data allowed the identification of transcription factors and their putative target genes associated with differentially accessible regions (DARs). Finally, functional experiments were conducted to investigate the effects of the identified regulatory factors and corresponding targets on HCC cell proliferation and migration. RESULTS Enrichment analysis of DARs between HCC and adjacent normal tissues revealed distinct signaling pathways and regulatory factors. Upregulated DARs in HCC were enriched in genes related to the MAPK and FoxO signaling pathways and associated with transcription factor families like ETS and AP-1. Conversely, downregulated DARs were associated with the TGF-β, cAMP, and p53 signaling pathways and the CTCF family. Integration of the datasets revealed a positive correlation between specific DARs and DEGs. Notably, PRPF3 emerged as a gene associated with DARs in HCC, and functional assays demonstrated its ability to promote HCC cell proliferation and migration. To the best of our knowledge, this is the first report highlighting the oncogenic role of PRPF3 in HCC. Furthermore, ZNF93 expression positively correlated with PRPF3, and ChIP-seq data indicated its potential role as a transcription factor regulating PRPF3 by binding to its promoter region. CONCLUSION This study provides a comprehensive analysis of the epigenetic landscape in HCC, encompassing both chromatin accessibility and the transcriptome. Our findings reveal that ZNF93 promotes the proliferation and motility of HCC cells through transcriptional regulation of a novel oncogene, PRPF3.
Collapse
Affiliation(s)
- Yi Bai
- Department of Hepatobiliary Surgery, School of Medicine, Tianjin First Central Hospital, Nankai University, Tianjin, China
| | - Xiyue Deng
- Tianjin First Central Hospital Clinic Institute, Tianjin Medical University, Tianjin, 300192, China
| | - Dapeng Chen
- Tianjin First Central Hospital Clinic Institute, Tianjin Medical University, Tianjin, 300192, China
| | - Shuangqing Han
- Tianjin First Central Hospital Clinic Institute, Tianjin Medical University, Tianjin, 300192, China
| | - Zijie Lin
- Tianjin First Central Hospital Clinic Institute, Tianjin Medical University, Tianjin, 300192, China
| | - Zhongmin Li
- Department of Hepatobiliary and Pancreatic Surgery, Tianjin Nankai Hospital, Tianjin, China
| | - Wen Tong
- Tianjin First Central Hospital Clinic Institute, Tianjin Medical University, Tianjin, 300192, China
| | - Jinming Li
- Tianjin First Central Hospital Clinic Institute, Tianjin Medical University, Tianjin, 300192, China
| | - Tianze Wang
- School of Medicine, Nankai University, Tianjin, 300071, China
| | - Xiangyu Liu
- School of Medicine, Nankai University, Tianjin, 300071, China
| | - Zirong Liu
- Department of Hepatobiliary Surgery, School of Medicine, Tianjin First Central Hospital, Nankai University, Tianjin, China
| | - Zilin Cui
- Department of Hepatobiliary Surgery, School of Medicine, Tianjin First Central Hospital, Nankai University, Tianjin, China
| | - Yamin Zhang
- Department of Hepatobiliary Surgery, School of Medicine, Tianjin First Central Hospital, Nankai University, Tianjin, China.
| |
Collapse
|
2
|
Huang JZ, Li JD, Chen G, He RQ. Identification of the key genes and mechanisms associated with transcatheter arterial chemoembolisation refractoriness in hepatocellular carcinoma. World J Clin Oncol 2024; 15:62-88. [PMID: 38292662 PMCID: PMC10823944 DOI: 10.5306/wjco.v15.i1.62] [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: 10/23/2023] [Revised: 12/12/2023] [Accepted: 12/28/2023] [Indexed: 01/23/2024] Open
Abstract
BACKGROUND Transcatheter arterial embolisation (TACE) is the primary treatment for intermediate-stage hepatocellular carcinoma (HCC) patients while some HCC cases have shown resistance to TACE. AIM To investigate the key genes and potential mechanisms correlated with TACE refractoriness in HCC. METHODS The microarray datasets of TACE-treated HCC tissues, HCC and non-HCC tissues were collected by searching multiple public databases. The respective differentially expressed genes (DEGs) were attained via limma R package. Weighted gene co-expression network analysis was employed for identifying the significant modules related to TACE non-response. TACE refractoriness-related genes were obtained by intersecting up-regulated TACE-associated and HCC-associated DEGs together with the genes in significant modules related to TACE non-response. The key genes expression in the above two pairs of samples was compared respectively via Wilcoxon tests and standard mean differences model. The prognostic value of the key genes was evaluated by Kaplan-Meier curve. Multivariate analysis was utilised to investigate the independent prognostic factor in key genes. Single-cell RNA (scRNA) sequencing analysis was conducted to explore the cell types in HCC. TACE refractoriness-related genes activity was calculated via AUCell packages. The CellChat R package was used for the investigation of the cell-cell communication between the identified cell types. RESULTS HCC tissues of TACE non-responders (n = 66) and TACE responders (n = 81), HCC (n = 3941) and non-HCC (n = 3443) tissues were obtained. The five key genes, DLG associated protein 5 (DLGAP5), Kinesin family member 20A (KIF20A), Assembly factor for spindle microtubules (ASPM), Kinesin family member 11 (KIF11) and TPX2 microtubule nucleation factor (TPX2) in TACE refractoriness-related genes, were identified. The five key genes were all up-regulated in the TACE non-responders group and the HCC group. High expression of the five key genes predicted poor prognosis in HCC. Among the key genes, TPX2 was an independent prognostic factor. Four cell types, hepatocytes, embryonic stem cells, T cells and B cells, were identified in the HCC tissues. The TACE refractoriness-related genes expressed primarily in hepatocytes and embryonic stem cells. Hepatocytes, as the providers of ligands, had the strongest interaction with embryonic stem cells that provided receptors. CONCLUSION Five key genes (DLGAP5, KIF20A, ASPM, KIF11 and TPX2) were identified as promoting refractory TACE. Hepatocytes and embryonic stem cells were likely to boost TACE refractoriness.
Collapse
Affiliation(s)
- Jie-Zhuang Huang
- Department of Medical Oncology, The First Affiliated Hospital of Guangxi Medical University, Nanning 530021, Guangxi Zhuang Autonomous Region, China
| | - Jian-Di Li
- Department of Pathology, The First Affiliated Hospital of Guangxi Medical University, Nanning 530021, Guangxi Zhuang Autonomous Region, China
| | - Gang Chen
- Department of Pathology, The First Affiliated Hospital of Guangxi Medical University, Nanning 530021, Guangxi Zhuang Autonomous Region, China
| | - Rong-Quan He
- Department of Medical Oncology, The First Affiliated Hospital of Guangxi Medical University, Nanning 530021, Guangxi Zhuang Autonomous Region, China
| |
Collapse
|
3
|
Karagac MS, Ceylan H. Neuroprotective Potential of Tannic Acid Against Neurotoxic Outputs of Monosodium Glutamate in Rat Cerebral Cortex. Neurotox Res 2023; 41:670-680. [PMID: 37713032 DOI: 10.1007/s12640-023-00667-y] [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: 02/21/2023] [Revised: 08/29/2023] [Accepted: 09/08/2023] [Indexed: 09/16/2023]
Abstract
Glutamate in monosodium glutamate (MSG), which is widely used in the food industry, has an important role in major brain functions such as memory, learning, synapse formation, and stabilization. However, extensive use of MSG has been linked with neurotoxicity. Therefore, in addition to clarifying the underlying mechanisms of MSG-induced neurotoxicity, it is also important to determine safe agents that can diminish the damage caused by MSG. Tannic acid (TA) is a naturally occurring plant polyphenol that exhibits versatile physiological effects such as anti-inflammatory, anti-carcinogenic, antioxidant, and radical scavenging. This study was conducted to assess the neurotoxic and neuroprotective effects of these two dietary components in the rat cerebral cortex. Twenty-four Sprague Dawley rats were divided into 4 equal groups and were treated with MSG (2 g/kg) and TA (50 mg/kg) alone and in combination for 3 weeks. Alterations in oxidative stress indicators (MDA and GSH) were measured in the cortex tissues. In addition, changes in enzymatic activities and gene expression patterns of antioxidant system components (GST, GPx, CAT, and SOD) were investigated. Furthermore, mRNA expressions of FoxO transcription factors (Foxo1 and Foxo3) and apoptotic markers (Casp3 and Casp9) were assessed. Results revealed that dietary TA intake significantly rehabilitated MSG-induced dysregulation in cortical tissue by regulating redox balance, cellular homeostasis, and apoptosis. The present study proposes that MSG-induced detrimental effects on cortical tissue are potentially mitigated by TA via modulation of oxidative stress, cell metabolism, and programmed cell death.
Collapse
Affiliation(s)
- Medine Sibel Karagac
- Department of Molecular Biology and Genetics, Faculty of Science, Atatürk University, Erzurum, Turkey
| | - Hamid Ceylan
- Department of Molecular Biology and Genetics, Faculty of Science, Atatürk University, Erzurum, Turkey.
| |
Collapse
|
4
|
Tabnak P, Hasanzade Bashkandi A, Ebrahimnezhad M, Soleimani M. Forkhead box transcription factors (FOXOs and FOXM1) in glioma: from molecular mechanisms to therapeutics. Cancer Cell Int 2023; 23:238. [PMID: 37821870 PMCID: PMC10568859 DOI: 10.1186/s12935-023-03090-7] [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: 09/26/2022] [Accepted: 10/04/2023] [Indexed: 10/13/2023] Open
Abstract
Glioma is the most aggressive and malignant type of primary brain tumor, comprises the majority of central nervous system deaths, and is categorized into different subgroups according to its histological characteristics, including astrocytomas, oligodendrogliomas, glioblastoma multiforme (GBM), and mixed tumors. The forkhead box (FOX) transcription factors comprise a collection of proteins that play various roles in numerous complex molecular cascades and have been discovered to be differentially expressed in distinct glioma subtypes. FOXM1 and FOXOs have been recognized as crucial transcription factors in tumor cells, including glioma cells. Accumulating data indicates that FOXM1 acts as an oncogene in various types of cancers, and a significant part of studies has investigated its function in glioma. Although recent studies considered FOXO subgroups as tumor suppressors, there are pieces of evidence that they may have an oncogenic role. This review will discuss the subtle functions of FOXOs and FOXM1 in gliomas, dissecting their regulatory network with other proteins, microRNAs and their role in glioma progression, including stem cell differentiation and therapy resistance/sensitivity, alongside highlighting recent pharmacological progress for modulating their expression.
Collapse
Affiliation(s)
- Peyman Tabnak
- Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran.
- Imam Reza Hospital, Tabriz University of Medical Sciences, Tabriz, Iran.
| | | | - Mohammad Ebrahimnezhad
- Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
- Imam Reza Hospital, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mahdieh Soleimani
- Imam Reza Hospital, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
5
|
Akhlaghipour I, Fanoodi A, Zangouei AS, Taghehchian N, Khalili-Tanha G, Moghbeli M. MicroRNAs as the Critical Regulators of Forkhead Box Protein Family in Pancreatic, Thyroid, and Liver Cancers. Biochem Genet 2023; 61:1645-1674. [PMID: 36781813 DOI: 10.1007/s10528-023-10346-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2022] [Accepted: 02/02/2023] [Indexed: 02/15/2023]
Abstract
The metabolism of human body is mainly regulated by the pancreas, liver, and thyroid using the hormones or exocrine secretions that affect the metabolic processes from food digestion to intracellular metabolism. Therefore, metabolic organ disorders have wide clinical symptoms that severely affect the quality of patient's life. The pancreatic, liver, and thyroid cancers as the main malignancies of the metabolic system have always been considered as one of the serious health challenges worldwide. Despite the novel therapeutic modalities, there are still significant high mortality and recurrence rates, especially in liver and pancreatic cancer patients which are mainly related to the late diagnosis. Therefore, it is required to assess the molecular bases of tumor progressions to introduce novel early detection and therapeutic markers in these malignancies. Forkhead box (FOX) protein family is a group of transcription factors that have pivotal roles in regulation of cell proliferation, migration, and apoptosis. They function as oncogene or tumor suppressor during tumor progression. MicroRNAs (miRNAs) are also involved in regulation of cellular processes. Therefore, in the present review, we discussed the role of miRNAs during pancreatic, thyroid, and liver tumor progressions through FOX regulation. It has been shown that miRNAs were mainly involved in tumor progression via FOXM and FOXO targeting. This review paves the way for the introduction of miR/FOX axis as an efficient early detection marker and therapeutic target in pancreatic, thyroid, and liver tumors.
Collapse
Affiliation(s)
- Iman Akhlaghipour
- Student Research Committee, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Ali Fanoodi
- Student Research Committee, School of Medicine, Birjand University of Medical Sciences, Birjand, Iran
| | - Amir Sadra Zangouei
- Student Research Committee, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Negin Taghehchian
- Medical Genetics Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Ghazaleh Khalili-Tanha
- Department of Medical Genetics and Molecular Medicine, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Meysam Moghbeli
- Medical Genetics Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.
- Department of Medical Genetics and Molecular Medicine, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
6
|
Xie W, Shi L, Quan H, Xiao H, Chen J, Liu J, de Dieu Habimana J, Huang R, Luo J, Chen P, Li Z. SYVN1 ubiquitinates FoxO1 to induce β-catenin nuclear translocation, PD-L1-mediated metastasis, and immune evasion of hepatocellular carcinoma. Cell Oncol (Dordr) 2023; 46:1285-1299. [PMID: 37099251 PMCID: PMC10618324 DOI: 10.1007/s13402-023-00811-y] [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] [Accepted: 04/04/2023] [Indexed: 04/27/2023] Open
Abstract
BACKGROUND A high incidence of hepatocellular carcinoma (HCC), the most frequently diagnosed form of liver cancer, is observed in Africa and Asia. SYVN1 is upregulated in HCC; however, the biological roles of SYVN1 in immune evasion remain unclear. METHODS RT-qPCR and western blot were employed to detect the expression levels of SYVN1 and the key molecules in HCC cells and tissues. Flow cytometry was used to determine the proportion of T cells, and an ELISA assay was used to determine the amount of IFN-γ secreted. Cell viability was monitored by CCK-8 and colony formation assays. The metastatic properties of HCC cells were detected by Transwell assays. Bioinformatics analysis, ChIP, and luciferase assays were used to study the transcriptional regulation of PD-L1. Co-IP was used to detect direct interaction between SYVN1 and FoxO1, as well as the ubiquitination of FoxO1. The in vitro findings were validated in xenograft and lung metastasis models. RESULTS In HCC cells and tissues, SYVN1 was upregulated while FoxO1 was downregulated. SYVN1 knockdown or FoxO1 overexpression reduced PD-L1 expression, and inhibited immune evasion, cell growth, and metastasis in HCC cells. Mechanistically, FoxO1 regulated PD-L1 transcription in a β-catenin-independent or -dependent manner. Functional studies further showed that SYVN1 promoted immune evasion, cell proliferation, migration and invasion via facilitating ubiquitin-proteasome-dependent degradation of FoxO1. In vivo investigations showed that silencing of SYVN1 inhibited immune evasion and metastasis of HCC cells, possible via the FoxO1/PD-L1 axis. CONCLUSION SYVN1 regulates FoxO1 ubiquitination to stimulate β-catenin nuclear translocation and promotes PD-L1-mediated metastasis and immune evasion in HCC.
Collapse
Affiliation(s)
- Wei Xie
- Department of Anatomy and Neurobiology, School of Basic Medical Sciences, Central South University, Changsha, 410031, Hunan Province, P.R. China
- Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, No. 283, Tongzipo Road, Yuelu Distirct, Changsha, 410031, Hunan Province, P.R. China
| | - Lei Shi
- Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, No. 283, Tongzipo Road, Yuelu Distirct, Changsha, 410031, Hunan Province, P.R. China
| | - Hu Quan
- Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, No. 283, Tongzipo Road, Yuelu Distirct, Changsha, 410031, Hunan Province, P.R. China
| | - Hua Xiao
- Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, No. 283, Tongzipo Road, Yuelu Distirct, Changsha, 410031, Hunan Province, P.R. China
| | - Jie Chen
- Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, No. 283, Tongzipo Road, Yuelu Distirct, Changsha, 410031, Hunan Province, P.R. China
| | - Jia Liu
- Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, No. 283, Tongzipo Road, Yuelu Distirct, Changsha, 410031, Hunan Province, P.R. China
| | - Jean de Dieu Habimana
- CAS Key Laboratory of Regenerative Biology, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530, Guangdong Province, P.R. China
| | - Rongqi Huang
- CAS Key Laboratory of Regenerative Biology, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530, Guangdong Province, P.R. China
| | - Jia Luo
- Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, No. 283, Tongzipo Road, Yuelu Distirct, Changsha, 410031, Hunan Province, P.R. China
| | - Pan Chen
- Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, No. 283, Tongzipo Road, Yuelu Distirct, Changsha, 410031, Hunan Province, P.R. China.
| | - Zhiyuan Li
- Department of Anatomy and Neurobiology, School of Basic Medical Sciences, Central South University, Changsha, 410031, Hunan Province, P.R. China
- CAS Key Laboratory of Regenerative Biology, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530, Guangdong Province, P.R. China
| |
Collapse
|
7
|
Rajan PK, Udoh UAS, Nakafuku Y, Pierre SV, Sanabria J. Normalization of the ATP1A1 Signalosome Rescinds Epigenetic Modifications and Induces Cell Autophagy in Hepatocellular Carcinoma. Cells 2023; 12:2367. [PMID: 37830582 PMCID: PMC10572209 DOI: 10.3390/cells12192367] [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: 08/10/2023] [Revised: 09/22/2023] [Accepted: 09/23/2023] [Indexed: 10/14/2023] Open
Abstract
Hepatocellular carcinoma (HCC) is the third leading cause of cancer-related death worldwide. In metabolic dysfunction-associated steatohepatitis (MASH)-related HCC, cellular redox imbalance from metabolic disturbances leads to dysregulation of the α1-subunit of the Na/K-ATPase (ATP1A1) signalosome. We have recently reported that the normalization of this pathway exhibited tumor suppressor activity in MASH-HCC. We hypothesized that dysregulated signaling from the ATP1A1, mediated by cellular metabolic stress, promotes aberrant epigenetic modifications including abnormal post-translational histone modifications and dysfunctional autophagic activity, leading to HCC development and progression. Increased H3K9 acetylation (H3K9ac) and H3K9 tri-methylation (H3K9me3) were observed in human HCC cell lines, HCC-xenograft and MASH-HCC mouse models, and epigenetic changes were associated with decreased cell autophagy in HCC cell lines. Inhibition of the pro-autophagic transcription factor FoxO1 was associated with elevated protein carbonylation and decreased levels of reduced glutathione (GSH). In contrast, normalization of the ATP1A1 signaling significantly decreased H3K9ac and H3K9me3, in vitro and in vivo, with concomitant nuclear localization of FoxO1, heightening cell autophagy and cancer-cell apoptotic activities in treated HCC cell lines. Our results showed the critical role of the ATP1A1 signalosome in HCC development and progression through epigenetic modifications and impaired cell autophagy activity, highlighting the importance of the ATP1A1 pathway as a potential therapeutic target for HCC.
Collapse
Affiliation(s)
- Pradeep Kumar Rajan
- Department of Surgery, Marshall Institute for Interdisciplinary Research, Marshall University School of Medicine, Huntington, WV 25701, USA; (P.K.R.); (U.-A.S.U.); (Y.N.); (S.V.P.)
| | - Utibe-Abasi S. Udoh
- Department of Surgery, Marshall Institute for Interdisciplinary Research, Marshall University School of Medicine, Huntington, WV 25701, USA; (P.K.R.); (U.-A.S.U.); (Y.N.); (S.V.P.)
| | - Yuto Nakafuku
- Department of Surgery, Marshall Institute for Interdisciplinary Research, Marshall University School of Medicine, Huntington, WV 25701, USA; (P.K.R.); (U.-A.S.U.); (Y.N.); (S.V.P.)
| | - Sandrine V. Pierre
- Department of Surgery, Marshall Institute for Interdisciplinary Research, Marshall University School of Medicine, Huntington, WV 25701, USA; (P.K.R.); (U.-A.S.U.); (Y.N.); (S.V.P.)
| | - Juan Sanabria
- Department of Surgery, Marshall Institute for Interdisciplinary Research, Marshall University School of Medicine, Huntington, WV 25701, USA; (P.K.R.); (U.-A.S.U.); (Y.N.); (S.V.P.)
- Department of Nutrition and Metabolomic Core Facility, Case Western Reserve University School of Medicine, Cleveland, OH 44100, USA
| |
Collapse
|
8
|
Tu S, Qiu Y. Molecular subtypes and scoring tools related to Foxo signaling pathway for assessing hepatocellular carcinoma prognosis and treatment responsiveness. Front Pharmacol 2023; 14:1213506. [PMID: 37693891 PMCID: PMC10483071 DOI: 10.3389/fphar.2023.1213506] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Accepted: 08/11/2023] [Indexed: 09/12/2023] Open
Abstract
Background: Transcription factors in Foxo signaling pathway influence hepatocellular carcinoma metastasis through epithelial mesenchymal transition-related pathways. Prognostic factors in the Foxo signaling pathway are feasible for HCC prognosis and therapeutic management. Methods: Based on the differentially expressed genes and Foxo signaling pathway genes in HCC, the ConsensusClusterPlus package was conducted to identify Foxo signaling pathway-related molecular subtypes in HCC. Based on the DEGs in the FMSs, the optimal prognostic factors in HCC were screened by cox and least absolute shrinkage and selection operator (LASSO) cox analysis to form the Foxo prognosis score (FPS). The prognostic predictive effectiveness of FPS was assessed by Kaplan Meier (K-M) analysis and Receiver Operating Characteristic (ROC) analysis. Additionally, tumor microenvironment (TME) score, tumor mutation burden (TMB) and treatment sensitivity differences in FMSs and FPS groups were also evaluated. Results: There were low, medium and high Foxo signaling pathway activity molecular subtypes in HCC named FMS 1, FMS 2 and FMS 3, respectively. FMS 1 with lowest Foxo signaling pathway activity presented an excellent survival advantage, while FMS 3 with highest Foxo signaling pathway activity exhibited an inhibitory TME status. According to FPS grouping, low FPS exhibited favorable survival, low TMB and anti-tumor activity. Patients in the low FPS group were mostly in the early stage of cancer. Moreover, we found that patients with high and low FPS exhibited different sensitivity to chemotherapy, and patients with low FPS were more sensitive to immunotherapy. Conclusion: We revealed a novel molecular subtype and prognostic tool based on Foxo signaling pathway signature, which could potentially provide a direction for accurate and effective assessment of potential personalized treatment options and prognostic management for HCC patients.
Collapse
Affiliation(s)
| | - Yunqing Qiu
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| |
Collapse
|
9
|
Guan J, Zhang ZY, Sun JH, Wang XP, Zhou ZQ, Qin L. LITAF inhibits colorectal cancer stemness and metastatic behavior by regulating FOXO1-mediated SIRT1 expression. Clin Exp Metastasis 2023:10.1007/s10585-023-10213-x. [PMID: 37266842 DOI: 10.1007/s10585-023-10213-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Accepted: 05/20/2023] [Indexed: 06/03/2023]
Abstract
Lipopolysaccharide-induced tumor necrosis factor alpha factor (LITAF) is a transcription factor that activates the transcription of TNF-α and regulates the inflammatory response. LITAF has been found to have potential anti-cancer effects of in several tumors. However, the role of LITAF in colorectal cancer (CRC) remains unclear. Through a comprehensive pan-cancer analysis of the Cancer Genome Atlas (TCGA), LITAF was identified as a differentially downregulated gene in CRC. We hypothesized that LITAF may participate in the modulation of CRC progression. The present study was aimed to investigate the expression profile of LITAF in CRC and its effect on metastatic behavior and stemness as well as the underlying molecular mechanism. The expression profile of LITAF in CRC, and its relationship with the prognosis of CRC were explored using public databases. LITAF expression was detected by quantitative real-time PCR (qRT-PCR), western blot, and immunohistochemistry. Furthermore, the effects of overexpression or knockdown of LITAF on cell proliferation, apoptosis, migration, invasion, and stemness of CRC cells were investigated in vitro. The regulatory effect of LITAF on forkhead Box O 1 (FOXO1)-sirtuin 1 (SIRT1) signaling axis was also explored. In addition, a xenograft mouse model was used to investigate the in-vivo role of LITAF. LITAF was downregulated in tumor tissues and its expression was associated with the prognosis, pathological stage and liver metastasis. In-vitro experiments confirmed that LITAF inhibited tumor cell proliferation, migration, invasion and stemness, and induced cell apoptosis. In vivo experiments demonstrated that LITAF inhibited the tumorigenicity and liver metastasis in tumor-bearing mice. Additionally, LITAF promoted FOXO1-mediated SIRT1 inhibition, thus regulating cancer stemness and malignant phenotypes. LITAF was silenced in CRC and it participated in the progression of CRC by inhibiting CRC cell stemness, and malignant phenotypes. Therefore, LITAF may serve as a novel biomarker of CRC prognosis.
Collapse
Affiliation(s)
- Jiao Guan
- Department of Surgery, The First Affiliated Hospital of Soochow University, Suzhou, 215006, China
- Department of Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
| | - Zheng-Yun Zhang
- Department of Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
| | - Jian-Hua Sun
- Department of Emergency, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
| | - Xin-Ping Wang
- Department of Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
| | - Zun-Qiang Zhou
- Department of Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China.
| | - Lei Qin
- Department of Surgery, The First Affiliated Hospital of Soochow University, Suzhou, 215006, China.
| |
Collapse
|
10
|
Xing B, Pu C, Chen Y, Sheng Y, Zhang B, Cui J, Wu G, Zhao Y. Insights into the characteristics of primary radioresistant cervical cancer using single-cell transcriptomics. Hum Cell 2023; 36:1135-1146. [PMID: 36867313 PMCID: PMC10110719 DOI: 10.1007/s13577-023-00882-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2022] [Accepted: 02/11/2023] [Indexed: 03/04/2023]
Abstract
Radioresistance is a major cause of radiotherapy failure among patients with cervical cancer (CC), the fourth most common cause of cancer mortality in women worldwide. Traditional CC cell lines lose intra-tumoral heterogeneity, posing a challenge for radioresistance research. Meanwhile, conditional reprogramming (CR) maintains intra-tumoral heterogeneity and complexity, as well as the genomic and clinical characteristics of original cells and tissues. Three radioresistant and two radiosensitive primary CC cell lines were developed under CR conditions from patient specimens, and their characteristics were verified via immunofluorescence, growth kinetics, clone forming assay, xenografting, and immunohistochemistry. The CR cell lines had homogenous characteristics with original tumor tissues and maintained radiosensitivity in vitro and in vivo, while also maintaining intra-tumoral heterogeneity according to single-cell RNA sequencing analysis. Upon further investigation, 20.83% of cells in radioresistant CR cell lines aggregated in the G2/M cell cycle phase, which is sensitive to radiation, compared to 38.1% of cells in radiosensitive CR cell lines. This study established three radioresistant and two radiosensitive CC cell lines through CR, which will benefit further research investigating radiosensitivity in CC. Our present study may provide an ideal model for research on development of radioresistance and potential therapeutic targets in CC.
Collapse
Affiliation(s)
- Biyuan Xing
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.,Institute of Radiation Oncology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Congli Pu
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.,Institute of Radiation Oncology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Yunshang Chen
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.,Institute of Radiation Oncology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Yuhan Sheng
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.,Institute of Radiation Oncology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Baofang Zhang
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.,Institute of Radiation Oncology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Jie Cui
- CAS Key Laboratory of Molecular Virology & Immunology, Center for Biosafety Mega-Science, Institut Pasteur of Shanghai, Chinese Academy of Sciences, Shanghai, China.
| | - Gang Wu
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China. .,Institute of Radiation Oncology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
| | - Yingchao Zhao
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China. .,Institute of Radiation Oncology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
| |
Collapse
|
11
|
Li S, Li Q, Ren Y, Yi J, Guo J, Kong X. HSV: The scout and assault for digestive system tumors. Front Mol Biosci 2023; 10:1142498. [PMID: 36926680 PMCID: PMC10011716 DOI: 10.3389/fmolb.2023.1142498] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2023] [Accepted: 02/15/2023] [Indexed: 03/08/2023] Open
Abstract
More than 25% of all malignant tumors are digestive system tumors (DSTs), which mostly include esophageal cancer, gastric cancer, pancreatic cancer, liver cancer, gallbladder cancer and cholangiocarcinoma, and colorectal cancer. DSTs have emerged as one of the prominent reasons of morbidity and death in many nations and areas around the world, posing a serious threat to human life and health. General treatments such as radiotherapy, chemotherapy, and surgical resection can poorly cure the patients and have a bad prognosis. A type of immunotherapy known as oncolytic virus therapy, have recently shown extraordinary anti-tumor effectiveness. One of the viruses that has been the subject of the greatest research in this field, the herpes simplex virus (HSV), has shown excellent potential in DSTs. With a discussion of HSV-1 based on recent studies, we outline the therapeutic effects of HSV on a number of DSTs in this review. Additionally, the critical function of HSV in the detection of cancers is discussed, and some HSV future possibilities are shown.
Collapse
Affiliation(s)
- Sheng Li
- College of Traditional Chinese medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Qingbo Li
- College of Traditional Chinese medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Yi Ren
- College of Traditional Chinese medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Jia Yi
- College of Traditional Chinese medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Jinhe Guo
- College of Traditional Chinese medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Xianbin Kong
- College of Traditional Chinese medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| |
Collapse
|
12
|
Zhao R, Ge Y, Gong Y, Li B, Xiao B, Zuo S. NAP1L5 targeting combined with MYH9 Inhibit HCC progression through PI3K/AKT/mTOR signaling pathway. Aging (Albany NY) 2022; 14:9000-9019. [PMID: 36374212 PMCID: PMC9740361 DOI: 10.18632/aging.204377] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2022] [Accepted: 10/29/2022] [Indexed: 11/13/2022]
Abstract
Hepatocellular carcinoma (HCC) is one of the leading causes of cancer death worldwide. Nucleosome assembly protein 1-like 5 (NAP1L5) is a protein-coding gene that encodes a protein similar to nucleosome assembly protein 1 (NAP1). It is a histone chaperone that plays an important role in gene transcription in organisms. However, the role of NAP1L5 in the pathogenesis of hepatocellular carcinoma remains to be elucidated. In this study, low expression of NAP1L5 was found in hepatocellular carcinoma, and the downregulation of NAP1L5 was related to shorter survival and disease-free survival. In addition, its expression is also related to the tumor size and recurrence of hepatocellular carcinoma. The overexpression and knockdown of NAP1L5 by plasmid and siRNA showed that NAP1L5 inhibited the proliferation, migration and invasion and induced apoptosis of hepatoma cells. In vivo experiments confirmed that NAP1L5 can inhibit the growth and metastasis of hepatocellular carcinoma cells. In the mechanistic study, we found that NAP1L5 affects the occurrence and development of hepatocellular carcinoma by regulating MYH9 to inhibit the PI3K/AKT/mTOR signaling pathway. As a functional tumor suppressor, NAP1L5 is expressed at low levels in HCC. NAP1L5 inhibits the PI3K/AKT/mTOR signaling pathway in hepatocellular carcinoma by regulating MYH9. It may be a new potential target for liver cancer treatment.
Collapse
Affiliation(s)
- Rui Zhao
- Guizhou Medical University, Guiyang, Guizhou 550001, China,Department of Hepatobiliary Surgery, The Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou 550001, China
| | - Yuzhen Ge
- Guizhou Medical University, Guiyang, Guizhou 550001, China
| | - Yongjun Gong
- Department of Hepatobiliary Surgery, The Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou 550001, China
| | - Bo Li
- Guizhou Medical University, Guiyang, Guizhou 550001, China
| | - Benli Xiao
- Guizhou Medical University, Guiyang, Guizhou 550001, China
| | - Shi Zuo
- Guizhou Medical University, Guiyang, Guizhou 550001, China,Department of Hepatobiliary Surgery, The Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou 550001, China
| |
Collapse
|
13
|
Scheffschick A, Babel J, Sperling S, Nerusch J, Herzog N, Seehofer D, Damm G. Primary-like Human Hepatocytes Genetically Engineered to Obtain Proliferation Competence as a Capable Application for Energy Metabolism Experiments in In Vitro Oncologic Liver Models. BIOLOGY 2022; 11:biology11081195. [PMID: 36009822 PMCID: PMC9405410 DOI: 10.3390/biology11081195] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/08/2022] [Revised: 07/21/2022] [Accepted: 08/03/2022] [Indexed: 11/17/2022]
Abstract
Simple Summary Fatty liver disease is an increasing health concern in Westernized countries. A fatty liver can lead to hepatocellular carcinoma (HCC), a type of liver cancer arising from hepatocytes, the major cells of the liver. How HCC may develop from the fatty liver is not known, and good cellular systems to investigate this are lacking. Recently, hepatocytes that can multiply continuously have been generated and suggested for hepatocyte research. In this study, we compared these continuously multiplying human hepatocytes to normal human hepatocytes and liver cancer cells, both within the state of fatty liver or not. We identified that these multiplying hepatocytes displayed many similarities to the liver cancer cells in terms of energy metabolism and concluded that these hepatocytes could be a pre-cancer model for liver cancer research and would be a valuable tool for HCC research. Abstract Non-alcoholic fatty liver disease (NAFLD), characterized by lipid accumulation in the liver, is the most common cause of liver diseases in Western countries. NAFLD is a major risk factor for developing hepatocellular carcinoma (HCC); however, in vitro evaluation of hepatic cancerogenesis fails due to a lack of liver models displaying a proliferation of hepatocytes. Originally designed to overcome primary human hepatocyte (PHH) shortages, upcyte hepatocytes were engineered to obtain continuous proliferation and, therefore, could be a suitable tool for HCC research. We generated upcyte hepatocytes, termed HepaFH3 cells, and compared their metabolic characteristics to HepG2 hepatoma cells and PHHs isolated from resected livers. For displaying NAFLD-related HCCs, we induced steatosis in all liver models. Lipid accumulation, lipotoxicity and energy metabolism were characterized using biochemical assays and Western blot analysis. We showed that proliferating HepaFH3 cells resemble HepG2, both showing a higher glucose uptake rate, lactate levels and metabolic rate compared to PHHs. Confluent HepaFH3 cells displayed some similarities to PHHs, including higher levels of the transaminases AST and ALT compared to proliferating HepaFH3 cells. We recommend proliferating HepaFH3 cells as a pre-malignant cellular model for HCC research, while confluent HepaFH3 cells could serve as PHH surrogates for energy metabolism studies.
Collapse
Affiliation(s)
- Andrea Scheffschick
- Department of Hepatobiliary Surgery and Visceral Transplantation, University Hospital, Leipzig University, 04103 Leipzig, Germany
- Saxonian Incubator for Clinical Translation (SIKT), Leipzig University, 04103 Leipzig, Germany
| | - Jonas Babel
- Department of Hepatobiliary Surgery and Visceral Transplantation, University Hospital, Leipzig University, 04103 Leipzig, Germany
| | - Sebastian Sperling
- Department of General, Visceral and Transplantation Surgery, Charité University Medicine Berlin, 13353 Berlin, Germany
| | - Julia Nerusch
- Department of Hepatobiliary Surgery and Visceral Transplantation, University Hospital, Leipzig University, 04103 Leipzig, Germany
- Saxonian Incubator for Clinical Translation (SIKT), Leipzig University, 04103 Leipzig, Germany
| | - Natalie Herzog
- Faculty of Science, Brandenburg University of Technology Cottbus-Senftenberg, 01968 Senftenberg, Germany
| | - Daniel Seehofer
- Department of Hepatobiliary Surgery and Visceral Transplantation, University Hospital, Leipzig University, 04103 Leipzig, Germany
- Department of General, Visceral and Transplantation Surgery, Charité University Medicine Berlin, 13353 Berlin, Germany
| | - Georg Damm
- Department of Hepatobiliary Surgery and Visceral Transplantation, University Hospital, Leipzig University, 04103 Leipzig, Germany
- Saxonian Incubator for Clinical Translation (SIKT), Leipzig University, 04103 Leipzig, Germany
- Department of General, Visceral and Transplantation Surgery, Charité University Medicine Berlin, 13353 Berlin, Germany
- Correspondence: ; Tel.: +49-341-97-39656
| |
Collapse
|
14
|
Udoh UAS, Banerjee M, Rajan PK, Sanabria JD, Smith G, Schade M, Sanabria JA, Nakafuku Y, Sodhi K, Pierre SV, Shapiro JI, Sanabria JR. Tumor-Suppressor Role of the α1-Na/K-ATPase Signalosome in NASH Related Hepatocellular Carcinoma †. Int J Mol Sci 2022; 23:ijms23137359. [PMID: 35806364 PMCID: PMC9266688 DOI: 10.3390/ijms23137359] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Revised: 06/24/2022] [Accepted: 06/29/2022] [Indexed: 02/07/2023] Open
Abstract
Hepatocellular carcinoma (HCC) is the second leading cause of cancer-related mortality worldwide, with an estimate of 0.84 million cases every year. In Western countries, because of the obesity epidemic, non-alcoholic steatohepatitis (NASH) has become the major cause of HCC. Intriguingly, the molecular mechanisms underlying tumorigenesis of HCC from NASH are largely unknown. We hypothesized that the growing uncoupled metabolism during NASH progression to HCC, manifested by lower cell redox status and an apoptotic ‘switch’ activity, follows a dysregulation of α1-Na/K-ATPase (NKA)/Src signalosome. Our results suggested that in NASH-related malignancy, α1-NKA signaling causes upregulation of the anti-apoptotic protein survivin and downregulation of the pro-apoptotic protein Smac/DIABLO via the activation of the PI3K → Akt pro-survival pathway with concomitant inhibition of the FoxO3 circuit, favoring cell division and primary liver carcinogenesis. Signalosome normalization using an inhibitory peptide resets apoptotic activity in malignant cells, with a significant decrease in tumor burden in vivo. Therefore, α1-NKA signalosome exercises in HCC the characteristic of a tumor suppressor, suggesting α1-NKA as a putative target for clinical therapy.
Collapse
Affiliation(s)
- Utibe-Abasi S. Udoh
- Department of Surgery, Marshall University Joan C. Edwards School of Medicine, Huntington, WV 25701, USA; (U.-A.S.U.); (M.B.); (P.K.R.); (J.D.S.); (G.S.); (M.S.); (J.A.S.); (Y.N.); (K.S.); (J.I.S.)
- Marshall Institute for Interdisciplinary Research, Marshall University Joan C. Edwards School of Medicine, Huntington, WV 25703, USA;
| | - Moumita Banerjee
- Department of Surgery, Marshall University Joan C. Edwards School of Medicine, Huntington, WV 25701, USA; (U.-A.S.U.); (M.B.); (P.K.R.); (J.D.S.); (G.S.); (M.S.); (J.A.S.); (Y.N.); (K.S.); (J.I.S.)
- Marshall Institute for Interdisciplinary Research, Marshall University Joan C. Edwards School of Medicine, Huntington, WV 25703, USA;
| | - Pradeep K. Rajan
- Department of Surgery, Marshall University Joan C. Edwards School of Medicine, Huntington, WV 25701, USA; (U.-A.S.U.); (M.B.); (P.K.R.); (J.D.S.); (G.S.); (M.S.); (J.A.S.); (Y.N.); (K.S.); (J.I.S.)
- Marshall Institute for Interdisciplinary Research, Marshall University Joan C. Edwards School of Medicine, Huntington, WV 25703, USA;
| | - Juan D. Sanabria
- Department of Surgery, Marshall University Joan C. Edwards School of Medicine, Huntington, WV 25701, USA; (U.-A.S.U.); (M.B.); (P.K.R.); (J.D.S.); (G.S.); (M.S.); (J.A.S.); (Y.N.); (K.S.); (J.I.S.)
- Marshall Institute for Interdisciplinary Research, Marshall University Joan C. Edwards School of Medicine, Huntington, WV 25703, USA;
| | - Gary Smith
- Department of Surgery, Marshall University Joan C. Edwards School of Medicine, Huntington, WV 25701, USA; (U.-A.S.U.); (M.B.); (P.K.R.); (J.D.S.); (G.S.); (M.S.); (J.A.S.); (Y.N.); (K.S.); (J.I.S.)
- Marshall Institute for Interdisciplinary Research, Marshall University Joan C. Edwards School of Medicine, Huntington, WV 25703, USA;
| | - Mathew Schade
- Department of Surgery, Marshall University Joan C. Edwards School of Medicine, Huntington, WV 25701, USA; (U.-A.S.U.); (M.B.); (P.K.R.); (J.D.S.); (G.S.); (M.S.); (J.A.S.); (Y.N.); (K.S.); (J.I.S.)
- Marshall Institute for Interdisciplinary Research, Marshall University Joan C. Edwards School of Medicine, Huntington, WV 25703, USA;
| | - Jacqueline A. Sanabria
- Department of Surgery, Marshall University Joan C. Edwards School of Medicine, Huntington, WV 25701, USA; (U.-A.S.U.); (M.B.); (P.K.R.); (J.D.S.); (G.S.); (M.S.); (J.A.S.); (Y.N.); (K.S.); (J.I.S.)
- Marshall Institute for Interdisciplinary Research, Marshall University Joan C. Edwards School of Medicine, Huntington, WV 25703, USA;
| | - Yuto Nakafuku
- Department of Surgery, Marshall University Joan C. Edwards School of Medicine, Huntington, WV 25701, USA; (U.-A.S.U.); (M.B.); (P.K.R.); (J.D.S.); (G.S.); (M.S.); (J.A.S.); (Y.N.); (K.S.); (J.I.S.)
- Marshall Institute for Interdisciplinary Research, Marshall University Joan C. Edwards School of Medicine, Huntington, WV 25703, USA;
| | - Komal Sodhi
- Department of Surgery, Marshall University Joan C. Edwards School of Medicine, Huntington, WV 25701, USA; (U.-A.S.U.); (M.B.); (P.K.R.); (J.D.S.); (G.S.); (M.S.); (J.A.S.); (Y.N.); (K.S.); (J.I.S.)
- Marshall Institute for Interdisciplinary Research, Marshall University Joan C. Edwards School of Medicine, Huntington, WV 25703, USA;
| | - Sandrine V. Pierre
- Marshall Institute for Interdisciplinary Research, Marshall University Joan C. Edwards School of Medicine, Huntington, WV 25703, USA;
| | - Joseph I. Shapiro
- Department of Surgery, Marshall University Joan C. Edwards School of Medicine, Huntington, WV 25701, USA; (U.-A.S.U.); (M.B.); (P.K.R.); (J.D.S.); (G.S.); (M.S.); (J.A.S.); (Y.N.); (K.S.); (J.I.S.)
- Marshall Institute for Interdisciplinary Research, Marshall University Joan C. Edwards School of Medicine, Huntington, WV 25703, USA;
| | - Juan R. Sanabria
- Department of Surgery, Marshall University Joan C. Edwards School of Medicine, Huntington, WV 25701, USA; (U.-A.S.U.); (M.B.); (P.K.R.); (J.D.S.); (G.S.); (M.S.); (J.A.S.); (Y.N.); (K.S.); (J.I.S.)
- Marshall Institute for Interdisciplinary Research, Marshall University Joan C. Edwards School of Medicine, Huntington, WV 25703, USA;
- Department of Nutrition and Metabolomic Core Facility, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA
- Correspondence: or
| |
Collapse
|
15
|
The potential role of FNDC5/irisin in various liver diseases: awakening the sleeping beauties. Expert Rev Mol Med 2022; 24:e23. [PMID: 35695040 DOI: 10.1017/erm.2022.19] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Fibronectin type III domain-containing protein 5 (FNDC5) is a transmembrane protein and the precursor of irisin, which serves as a systemic exerkine/myokine with multiple origins. Since its discovery in 2012, this hormone-like polypeptide has rapidly evolved to a component significantly involved in a gamut of metabolic dysregulations and various liver diseases. After a decade of extensive investigation on FNDC5/irisin, we are still surrounded by lots of open questions regarding its diagnostic and therapeutic values. In this review, we first concentrated on the structure-function relationship of FNDC5/irisin. Next, we comprehensively summarised the current knowledge and research findings regarding pathogenic roles/therapeutic applications of FNDC5/irisin in the context of non-alcoholic fatty liver disease, fibrosis, liver injury due to multiple detrimental insults, hepatic malignancy and intrahepatic cholestasis of pregnancy. Moreover, the prominent molecules involved in the underlying mechanisms and signalling pathways were highlighted. As a result, emerging evidence reveals FNDC5/irisin may act as a proxy for diagnosing liver disease pathology, a sensitive biomarker for assessing damage severity, a predisposing factor for surveilling illness progression and a treatment option with protective/preventive impact, all of which are highly dependent on disease grading and contextually pathological features.
Collapse
|
16
|
Wang J, Wang Y, Wang J, Zhang S, Yu Z, Zheng K, Fu Z, Wang C, Huang W, Chen J. DEAD-box helicase 56 functions as an oncogene promote cell proliferation and invasion in gastric cancer via the FOXO1/p21 Cip1/c-Myc signaling pathway. Bioengineered 2022; 13:13970-13985. [PMID: 35723050 PMCID: PMC9275944 DOI: 10.1080/21655979.2022.2084235] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022] Open
Abstract
DEAD-box helicase (DDX) family exerts a critical effect on cancer initiation and progression through alternative splicing, transcription and ribosome biogenesis. Increasing evidence has demonstrated that DEAD-box helicase 56 (DDX56) is over-expressed in several cancers, which plays an oncogenic role. Till the present, the impact of DDX56 on gastric cancer (GC) remains unclear. We conducted high-throughput sequencing (RNA-seq) to demonstrate aberrant DDX56 levels within 10 GC and matched non-carcinoma tissue samples. DDX56 levels were detected through qRT-PCR, western blotting (WB) and immunochemical staining in GC patients. We conducted gain- and loss-of-function studies to examine DDX56's biological role in GC development. In vitro, we carried out 5‑Ethynyl‑2‑deoxyuridine (EdU), scratch, Transwell, and flow cytometry (FCM) assays for detecting GC cell growth, invasion, migration and apoptosis. Additionally, gene set enrichment analysis (GSEA), WB assay, and Encyclopedia of RNA Interactomes (ENCORI) were carried out for analyzing DDX56-regulated downstream genes and signaling pathways. In vivo, tumor xenograft experiment was performed for investigating how DDX56 affected GC development within BALB/c nude mice. Functionally, DDX56 knockdown arrested cell cycle at G1 phase, invasion and migration of AGS and MKN28 cells, and enhanced their apoptosis. Ectopic DDX56 expression enhanced the cell growth, migration and invasion, and inhibited apoptosis. Knockdown of DDX56 suppressed GC growth in the tumor models of BALB/c nude mice. Mechanistically, DDX56 post-transcriptionally suppressed FOXO1/p21 Cip1 protein expression, which could activate its downstream cyclin E1/CDK2/c-Myc signaling pathways. This sheds lights on the GC pathogenic mechanism and offers a potential anti-cancer therapeutic target.
Collapse
Affiliation(s)
- Jiancheng Wang
- Department of Gastrointestinal Gland Surgery, the First Affiliated Hospital of Guangxi Medical University, Guangxi Medical University, Nanning, Guangxi, China.,Guangxi Key Laboratory of Enhanced Recovery after Surgery for Gastrointestinal Cancer, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
| | - Ye Wang
- Department of Gastrointestinal Gland Surgery, the First Affiliated Hospital of Guangxi Medical University, Guangxi Medical University, Nanning, Guangxi, China.,Guangxi Key Laboratory of Enhanced Recovery after Surgery for Gastrointestinal Cancer, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
| | - Junfu Wang
- Department of General Surgery, The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Siwen Zhang
- Department of Gastrointestinal Gland Surgery, the First Affiliated Hospital of Guangxi Medical University, Guangxi Medical University, Nanning, Guangxi, China.,Guangxi Key Laboratory of Enhanced Recovery after Surgery for Gastrointestinal Cancer, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
| | - Zhu Yu
- Department of Gastrointestinal Gland Surgery, the First Affiliated Hospital of Guangxi Medical University, Guangxi Medical University, Nanning, Guangxi, China.,Guangxi Key Laboratory of Enhanced Recovery after Surgery for Gastrointestinal Cancer, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
| | - Kaitian Zheng
- Department of Gastrointestinal Gland Surgery, the First Affiliated Hospital of Guangxi Medical University, Guangxi Medical University, Nanning, Guangxi, China.,Guangxi Key Laboratory of Enhanced Recovery after Surgery for Gastrointestinal Cancer, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
| | - Zhao Fu
- Department of Gastrointestinal Gland Surgery, the First Affiliated Hospital of Guangxi Medical University, Guangxi Medical University, Nanning, Guangxi, China.,Guangxi Key Laboratory of Enhanced Recovery after Surgery for Gastrointestinal Cancer, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
| | - Congjun Wang
- Department of Gastrointestinal Gland Surgery, the First Affiliated Hospital of Guangxi Medical University, Guangxi Medical University, Nanning, Guangxi, China.,Guangxi Key Laboratory of Enhanced Recovery after Surgery for Gastrointestinal Cancer, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
| | - Weijia Huang
- Department of Gastrointestinal Gland Surgery, the First Affiliated Hospital of Guangxi Medical University, Guangxi Medical University, Nanning, Guangxi, China.,Guangxi Key Laboratory of Enhanced Recovery after Surgery for Gastrointestinal Cancer, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
| | - Junqiang Chen
- Department of Gastrointestinal Gland Surgery, the First Affiliated Hospital of Guangxi Medical University, Guangxi Medical University, Nanning, Guangxi, China.,Guangxi Key Laboratory of Enhanced Recovery after Surgery for Gastrointestinal Cancer, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
| |
Collapse
|
17
|
The Bright and the Dark Side of TGF-β Signaling in Hepatocellular Carcinoma: Mechanisms, Dysregulation, and Therapeutic Implications. Cancers (Basel) 2022; 14:cancers14040940. [PMID: 35205692 PMCID: PMC8870127 DOI: 10.3390/cancers14040940] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2021] [Revised: 02/02/2022] [Accepted: 02/03/2022] [Indexed: 01/18/2023] Open
Abstract
Simple Summary Transforming growth factor β (TGF-β) signaling is a preeminent regulator of diverse cellular and physiological processes. Frequent dysregulation of TGF-β signaling has been implicated in cancer. In hepatocellular carcinoma (HCC), the most prevalent form of primary liver cancer, the autocrine and paracrine effects of TGF-β have paradoxical implications. While acting as a potent tumor suppressor pathway in the early stages of malignancy, TGF-β diverts to a promoter of tumor progression in the late stages, reflecting its bright and dark natures, respectively. Within this context, targeting TGF-β represents a promising therapeutic option for HCC treatment. We discuss here the molecular properties of TGF-β signaling in HCC, attempting to provide an overview of its effects on tumor cells and the stroma. We also seek to evaluate the dysregulation mechanisms that mediate the functional switch of TGF-β from a tumor suppressor to a pro-tumorigenic signal. Finally, we reconcile its biphasic nature with the therapeutic implications. Abstract Hepatocellular carcinoma (HCC) is associated with genetic and nongenetic aberrations that impact multiple genes and pathways, including the frequently dysregulated transforming growth factor β (TGF-β) signaling pathway. The regulatory cytokine TGF-β and its signaling effectors govern a broad spectrum of spatiotemporally regulated molecular and cellular responses, yet paradoxically have dual and opposing roles in HCC progression. In the early stages of tumorigenesis, TGF-β signaling enforces profound tumor-suppressive effects, primarily by inducing cell cycle arrest, cellular senescence, autophagy, and apoptosis. However, as the tumor advances in malignant progression, TGF-β functionally switches to a pro-tumorigenic signal, eliciting aggressive tumor traits, such as epithelial–mesenchymal transition, tumor microenvironment remodeling, and immune evasion of cancer cells. On this account, the inhibition of TGF-β signaling is recognized as a promising therapeutic strategy for advanced HCC. In this review, we evaluate the functions and mechanisms of TGF-β signaling and relate its complex and pleiotropic biology to HCC pathophysiology, attempting to provide a detailed perspective on the molecular determinants underlying its functional diversion. We also address the therapeutic implications of the dichotomous nature of TGF-β signaling and highlight the rationale for targeting this pathway for HCC treatment, alone or in combination with other agents.
Collapse
|
18
|
Sun EJ, Wankell M, Palamuthusingam P, McFarlane C, Hebbard L. Targeting the PI3K/Akt/mTOR Pathway in Hepatocellular Carcinoma. Biomedicines 2021; 9:biomedicines9111639. [PMID: 34829868 PMCID: PMC8615614 DOI: 10.3390/biomedicines9111639] [Citation(s) in RCA: 97] [Impact Index Per Article: 32.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2021] [Revised: 11/05/2021] [Accepted: 11/05/2021] [Indexed: 12/24/2022] Open
Abstract
Despite advances in the treatment of cancers through surgical procedures and new pharmaceuticals, the treatment of hepatocellular carcinoma (HCC) remains challenging as reflected by low survival rates. The PI3K/Akt/mTOR pathway is an important signaling mechanism that regulates the cell cycle, proliferation, apoptosis, and metabolism. Importantly, deregulation of the PI3K/Akt/mTOR pathway leading to activation is common in HCC and is hence the subject of intense investigation and the focus of current therapeutics. In this review article, we consider the role of this pathway in the pathogenesis of HCC, focusing on its downstream effectors such as glycogen synthase kinase-3 (GSK-3), cAMP-response element-binding protein (CREB), forkhead box O protein (FOXO), murine double minute 2 (MDM2), p53, and nuclear factor-κB (NF-κB), and the cellular processes of lipogenesis and autophagy. In addition, we provide an update on the current ongoing clinical development of agents targeting this pathway for HCC treatments.
Collapse
Affiliation(s)
- Eun Jin Sun
- Centre for Molecular Therapeutics, Department of Molecular and Cell Biology, Australian Institute of Tropical Medicine and Health, College of Public Health, Medical and Veterinary Sciences, James Cook University, Townsville, QLD 4811, Australia; (E.J.S.); (M.W.); (C.M.)
- College of Medicine and Dentistry, James Cook University, Townsville, QLD 4811, Australia
| | - Miriam Wankell
- Centre for Molecular Therapeutics, Department of Molecular and Cell Biology, Australian Institute of Tropical Medicine and Health, College of Public Health, Medical and Veterinary Sciences, James Cook University, Townsville, QLD 4811, Australia; (E.J.S.); (M.W.); (C.M.)
| | - Pranavan Palamuthusingam
- Institute of Surgery, The Townsville University Hospital, Townsville, QLD 4811, Australia;
- Mater Hospital, Townsville, QLD 4811, Australia
| | - Craig McFarlane
- Centre for Molecular Therapeutics, Department of Molecular and Cell Biology, Australian Institute of Tropical Medicine and Health, College of Public Health, Medical and Veterinary Sciences, James Cook University, Townsville, QLD 4811, Australia; (E.J.S.); (M.W.); (C.M.)
| | - Lionel Hebbard
- Centre for Molecular Therapeutics, Department of Molecular and Cell Biology, Australian Institute of Tropical Medicine and Health, College of Public Health, Medical and Veterinary Sciences, James Cook University, Townsville, QLD 4811, Australia; (E.J.S.); (M.W.); (C.M.)
- Storr Liver Centre, Westmead Institute for Medical Research, Westmead Hospital and University of Sydney, Sydney, NSW 2145, Australia
- Correspondence:
| |
Collapse
|
19
|
Autophagy-Related Chemoprotection against Sorafenib in Human Hepatocarcinoma: Role of FOXO3 Upregulation and Modulation by Regorafenib. Int J Mol Sci 2021; 22:ijms222111770. [PMID: 34769197 PMCID: PMC8583804 DOI: 10.3390/ijms222111770] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2021] [Revised: 10/20/2021] [Accepted: 10/26/2021] [Indexed: 02/07/2023] Open
Abstract
Early acquisition of sorafenib resistance is responsible for the dismal prognosis of advanced hepatocarcinoma (HCC). Autophagy, a catabolic process involved in liver homeostasis, has been associated with chemosensitivity modulation. Forkhead box O3 (FOXO3) is a transcription factor linked to HCC pathogenesis whose role on autophagy-related sorafenib resistance remains controversial. Here, we unraveled the linkage between autophagy and sorafenib resistance in HCC, focusing on the implication of FOXO3 and its potential modulation by regorafenib. We worked with two HepG2-derived sorafenib-resistant HCC in vitro models (HepG2S1 and HepG2S3) and checked HCC patient data from the UALCAN database. Resistant cells displayed an enhanced basal autophagic flux compared to HepG2, showing higher autophagolysosome content and autophagy markers levels. Pharmacological inhibition of autophagy boosted HepG2S1 and HepG2S3 apoptosis and subG1 cells, but reduced viability, indicating the cytoprotective role of autophagy. HCC samples displayed higher FOXO3 levels, being associated with shorter survival and autophagic genes expression. Consistently, chemoresistant in vitro models showed significant FOXO3 upregulation. FOXO3 knockdown suppressed autophagy and caused resistant cell death, demonstrating that overactivation of such pro-survival autophagy during sorafenib resistance is FOXO3-dependent; a cytoprotective mechanism that the second-line drug regorafenib successfully abolished. Therefore, targeting FOXO3-mediated autophagy could significantly improve the clinical efficacy of sorafenib.
Collapse
|