1
|
Zhang TQ, Lv QY, Jin WL. The cellular-centered view of hypoxia tumor microenvironment: Molecular mechanisms and therapeutic interventions. Biochim Biophys Acta Rev Cancer 2024; 1879:189137. [PMID: 38880161 DOI: 10.1016/j.bbcan.2024.189137] [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: 12/11/2023] [Revised: 06/01/2024] [Accepted: 06/10/2024] [Indexed: 06/18/2024]
Abstract
Cancer is a profoundly dynamic, heterogeneous and aggressive systemic ailment, with a coordinated evolution of various types of tumor niches. Hypoxia plays an indispensable role in the tumor micro-ecosystem, drastically enhancing the plasticity of cancer cells, fibroblasts and immune cells and orchestrating intercellular communication. Hypoxia-induced signals, particularly hypoxia-inducible factor-1α (HIF-1α), drive the reprogramming of genetic, transcriptional, and proteomic profiles. This leads to a spectrum of interconnected processes, including augmented survival of cancer cells, evasion of immune surveillance, metabolic reprogramming, remodeling of the extracellular matrix, and the development of resistance to conventional therapeutic modalities like radiotherapy and chemotherapy. Here, we summarize the latest research on the multifaceted effects of hypoxia, where a multitude of cellular and non-cellular elements crosstalk with each other and co-evolve in a synergistic manner. Additionally, we investigate therapeutic approaches targeting hypoxic niche, encompassing hypoxia-activated prodrugs, HIF inhibitors, nanomedicines, and combination therapies. Finally, we discuss some of the issues to be addressed and highlight the potential of emerging technologies in the treatment of cancer.
Collapse
Affiliation(s)
- Tian-Qi Zhang
- Institute of Cancer Neuroscience, Medical Frontier Innovation Research Center, The First Hospital of Lanzhou University, The First Clinical Medical College of Lanzhou University, Lanzhou 730000, China; The Second Hospital of Jilin University, Changchun 130041, China
| | - Qian-Yu Lv
- The Second Hospital of Jilin University, Changchun 130041, China
| | - Wei-Lin Jin
- Institute of Cancer Neuroscience, Medical Frontier Innovation Research Center, The First Hospital of Lanzhou University, The First Clinical Medical College of Lanzhou University, Lanzhou 730000, China.
| |
Collapse
|
2
|
Shi X, Zeng X, Jiao R, Yang Y, Du X, Qian J, Liu J, Chen G. Expression, Prognostic Value, and Biological Function of CTHRC1 in Different Types of Gliomas: A Bioinformatic Analysis and Experiment Validation. Clin Med Insights Oncol 2024; 18:11795549241260576. [PMID: 38894702 PMCID: PMC11185027 DOI: 10.1177/11795549241260576] [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: 02/06/2024] [Accepted: 05/23/2024] [Indexed: 06/21/2024] Open
Abstract
Background In recent years, abnormal expression of collagen triple helix repeat containing 1 (CTHRC1) has been found in some tumors, closely related to the poor prognosis of cancer patients. However, the clinical significance of CTHRC1 in gliomas is not completely understood. Methods We investigated the expression, prognostic value, and potential biological function of CTHRC1 in different types of gliomas through bioinformatics analysis and experimental verification. Results Bioinformatics analysis revealed several key findings regarding the expression and clinical significance of CTHRC1 in gliomas. First, the analysis demonstrated a positive correlation between CTHRC1 expression and the World Health Organization (WHO) grading of gliomas, a relationship that was validated through immunohistochemistry experiments. In addition, a trend was observed in which CTHRC1 expression increased with the extent of glioma invasion, as supported by Western blot experiments. Subsequent bioinformatics analysis identified the mesenchymal subtype of gliomas as having the highest levels of CTHRC1 expression, a finding reinforced by immunohistochemical staining. Moreover, high CTHRC1 expression was associated with poor prognosis in gliomas and emerged as an independent prognostic factor, with varying impacts on prognosis between low-grade gliomas (LGGs) and glioblastoma (GBM) subgroups. Notably, comparative analysis unveiled distinct patterns of immune infiltration of CTHRC1 in LGG and GBM. Furthermore, alterations in copy number variations and DNA methylation were identified as potential mechanisms underlying elevated CTHRC1 levels in gliomas. Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analysis indicated that CTHRC1 and its associated genes mainly function in the extracellular matrix and participate in tumor-related signaling pathways. Conclusions The CTHRC1 has shown significant clinical utility as a prognostic marker and mesenchymal subtype marker of glioma.
Collapse
Affiliation(s)
- Xueping Shi
- Department of Neurosurgery, The Affiliated Hospital of Guizhou Medical University, Guiyang, P.R. China
| | - Xi Zeng
- Department of Neurosurgery, The Affiliated Hospital of Guizhou Medical University, Guiyang, P.R. China
| | - Rukai Jiao
- Department of Neurosurgery, The Jinyang Hospital Affiliated to Guizhou Medical University, Guiyang, P.R. China
| | - Yushi Yang
- Department of Pathology, The Affiliated Hospital of Guizhou Medical University, Guiyang, P.R. China
| | - Xiaolin Du
- Department of Neurosurgery, The Jinyang Hospital Affiliated to Guizhou Medical University, Guiyang, P.R. China
| | - Jiacai Qian
- Department of Neurosurgery, The Jinyang Hospital Affiliated to Guizhou Medical University, Guiyang, P.R. China
| | - Jian Liu
- Department of Neurosurgery, Guizhou Provincial People’s Hospital, Guiyang, P.R. China
| | - Guangtang Chen
- Department of Neurosurgery, The Affiliated Hospital of Guizhou Medical University, Guiyang, P.R. China
| |
Collapse
|
3
|
Xu C, Hou P, Li X, Xiao M, Zhang Z, Li Z, Xu J, Liu G, Tan Y, Fang C. Comprehensive understanding of glioblastoma molecular phenotypes: classification, characteristics, and transition. Cancer Biol Med 2024; 21:j.issn.2095-3941.2023.0510. [PMID: 38712813 PMCID: PMC11131044 DOI: 10.20892/j.issn.2095-3941.2023.0510] [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: 12/27/2023] [Accepted: 03/28/2024] [Indexed: 05/08/2024] Open
Abstract
Among central nervous system-associated malignancies, glioblastoma (GBM) is the most common and has the highest mortality rate. The high heterogeneity of GBM cell types and the complex tumor microenvironment frequently lead to tumor recurrence and sudden relapse in patients treated with temozolomide. In precision medicine, research on GBM treatment is increasingly focusing on molecular subtyping to precisely characterize the cellular and molecular heterogeneity, as well as the refractory nature of GBM toward therapy. Deep understanding of the different molecular expression patterns of GBM subtypes is critical. Researchers have recently proposed tetra fractional or tripartite methods for detecting GBM molecular subtypes. The various molecular subtypes of GBM show significant differences in gene expression patterns and biological behaviors. These subtypes also exhibit high plasticity in their regulatory pathways, oncogene expression, tumor microenvironment alterations, and differential responses to standard therapy. Herein, we summarize the current molecular typing scheme of GBM and the major molecular/genetic characteristics of each subtype. Furthermore, we review the mesenchymal transition mechanisms of GBM under various regulators.
Collapse
Affiliation(s)
- Can Xu
- School of Clinical Medicine, Hebei University, Department of Neurosurgery, Affiliated Hospital of Hebei University, Baoding 07100, China
- Hebei Key Laboratory of Precise Diagnosis and Treatment of Glioma, Baoding 071000, China
| | - Pengyu Hou
- Hebei Key Laboratory of Precise Diagnosis and Treatment of Glioma, Baoding 071000, China
- School of Basic Medical Sciences, Hebei University, Baoding 07100, China
| | - Xiang Li
- School of Basic Medical Sciences, Hebei University, Baoding 07100, China
| | - Menglin Xiao
- School of Clinical Medicine, Hebei University, Department of Neurosurgery, Affiliated Hospital of Hebei University, Baoding 07100, China
- Hebei Key Laboratory of Precise Diagnosis and Treatment of Glioma, Baoding 071000, China
| | - Ziqi Zhang
- School of Clinical Medicine, Hebei University, Department of Neurosurgery, Affiliated Hospital of Hebei University, Baoding 07100, China
- Hebei Key Laboratory of Precise Diagnosis and Treatment of Glioma, Baoding 071000, China
| | - Ziru Li
- Hebei Key Laboratory of Precise Diagnosis and Treatment of Glioma, Baoding 071000, China
- School of Basic Medical Sciences, Hebei University, Baoding 07100, China
| | - Jianglong Xu
- School of Clinical Medicine, Hebei University, Department of Neurosurgery, Affiliated Hospital of Hebei University, Baoding 07100, China
- Hebei Key Laboratory of Precise Diagnosis and Treatment of Glioma, Baoding 071000, China
| | - Guoming Liu
- School of Clinical Medicine, Hebei University, Department of Neurosurgery, Affiliated Hospital of Hebei University, Baoding 07100, China
- Hebei Key Laboratory of Precise Diagnosis and Treatment of Glioma, Baoding 071000, China
| | - Yanli Tan
- Hebei Key Laboratory of Precise Diagnosis and Treatment of Glioma, Baoding 071000, China
- School of Basic Medical Sciences, Hebei University, Baoding 07100, China
- Department of Pathology, Affiliated Hospital of Hebei University, Baoding 07100, China
| | - Chuan Fang
- School of Clinical Medicine, Hebei University, Department of Neurosurgery, Affiliated Hospital of Hebei University, Baoding 07100, China
- Hebei Key Laboratory of Precise Diagnosis and Treatment of Glioma, Baoding 071000, China
| |
Collapse
|
4
|
Dolgova N, Uhlemann EME, Boniecki MT, Vizeacoumar FS, Ara A, Nouri P, Ralle M, Tonelli M, Abbas SA, Patry J, Elhasasna H, Freywald A, Vizeacoumar FJ, Dmitriev OY. MEMO1 binds iron and modulates iron homeostasis in cancer cells. eLife 2024; 13:e86354. [PMID: 38640016 PMCID: PMC11081632 DOI: 10.7554/elife.86354] [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: 01/21/2023] [Accepted: 04/15/2024] [Indexed: 04/20/2024] Open
Abstract
Mediator of ERBB2-driven cell motility 1 (MEMO1) is an evolutionary conserved protein implicated in many biological processes; however, its primary molecular function remains unknown. Importantly, MEMO1 is overexpressed in many types of cancer and was shown to modulate breast cancer metastasis through altered cell motility. To better understand the function of MEMO1 in cancer cells, we analyzed genetic interactions of MEMO1 using gene essentiality data from 1028 cancer cell lines and found multiple iron-related genes exhibiting genetic relationships with MEMO1. We experimentally confirmed several interactions between MEMO1 and iron-related proteins in living cells, most notably, transferrin receptor 2 (TFR2), mitoferrin-2 (SLC25A28), and the global iron response regulator IRP1 (ACO1). These interactions indicate that cells with high-MEMO1 expression levels are hypersensitive to the disruptions in iron distribution. Our data also indicate that MEMO1 is involved in ferroptosis and is linked to iron supply to mitochondria. We have found that purified MEMO1 binds iron with high affinity under redox conditions mimicking intracellular environment and solved MEMO1 structures in complex with iron and copper. Our work reveals that the iron coordination mode in MEMO1 is very similar to that of iron-containing extradiol dioxygenases, which also display a similar structural fold. We conclude that MEMO1 is an iron-binding protein that modulates iron homeostasis in cancer cells.
Collapse
Affiliation(s)
- Natalia Dolgova
- Department of Biochemistry, Microbiology and Immunology, University of SaskatchewanSaskatoonCanada
| | - Eva-Maria E Uhlemann
- Department of Biochemistry, Microbiology and Immunology, University of SaskatchewanSaskatoonCanada
| | - Michal T Boniecki
- Protein Characterization and Crystallization Facility, University of SaskatchewanSaskatoonCanada
| | | | - Anjuman Ara
- Department of Biochemistry, Microbiology and Immunology, University of SaskatchewanSaskatoonCanada
| | - Paria Nouri
- Department of Biochemistry, Microbiology and Immunology, University of SaskatchewanSaskatoonCanada
| | - Martina Ralle
- Department of Molecular and Medical Genetics, Oregon Health and Sciences UniversityPortlandUnited States
| | - Marco Tonelli
- National Magnetic Resonance Facility at Madison (NMRFAM), University of WisconsinMadisonUnited States
| | - Syed A Abbas
- Department of Biochemistry, Microbiology and Immunology, University of SaskatchewanSaskatoonCanada
| | - Jaala Patry
- Department of Biochemistry, Microbiology and Immunology, University of SaskatchewanSaskatoonCanada
| | - Hussain Elhasasna
- Department of Pathology and Laboratory Medicine, University of SaskatchewanSaskatoonCanada
| | - Andrew Freywald
- Department of Pathology and Laboratory Medicine, University of SaskatchewanSaskatoonCanada
| | - Franco J Vizeacoumar
- Cancer Research Department, Saskatchewan Cancer AgencySaskatoonCanada
- Division of Oncology, University of SaskatchewanSaskatoonCanada
| | - Oleg Y Dmitriev
- Department of Biochemistry, Microbiology and Immunology, University of SaskatchewanSaskatoonCanada
| |
Collapse
|
5
|
Lapcik P, Stacey RG, Potesil D, Kulhanek P, Foster LJ, Bouchal P. Global Interactome Mapping Reveals Pro-tumorigenic Interactions of NF-κB in Breast Cancer. Mol Cell Proteomics 2024; 23:100744. [PMID: 38417630 PMCID: PMC10988130 DOI: 10.1016/j.mcpro.2024.100744] [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: 07/21/2023] [Revised: 02/01/2024] [Accepted: 02/23/2024] [Indexed: 03/01/2024] Open
Abstract
NF-κB pathway is involved in inflammation; however, recent data shows its role also in cancer development and progression, including metastasis. To understand the role of NF-κB interactome dynamics in cancer, we study the complexity of breast cancer interactome in luminal A breast cancer model and its rearrangement associated with NF-κB modulation. Liquid chromatography-mass spectrometry measurement of 160 size-exclusion chromatography fractions identifies 5460 protein groups. Seven thousand five hundred sixty eight interactions among these proteins have been reconstructed by PrInCE algorithm, of which 2564 have been validated in independent datasets. NF-κB modulation leads to rearrangement of protein complexes involved in NF-κB signaling and immune response, cell cycle regulation, and DNA replication. Central NF-κB transcription regulator RELA co-elutes with interactors of NF-κB activator PRMT5, and these complexes are confirmed by AlphaPulldown prediction. A complementary immunoprecipitation experiment recapitulates RELA interactions with other NF-κB factors, associating NF-κB inhibition with lower binding of NF-κB activators to RELA. This study describes a network of pro-tumorigenic protein interactions and their rearrangement upon NF-κB inhibition with potential therapeutic implications in tumors with high NF-κB activity.
Collapse
Affiliation(s)
- Petr Lapcik
- Department of Biochemistry, Faculty of Science, Masaryk University, Brno, Czech Republic
| | - R Greg Stacey
- Michael Smith Laboratories, University of British Columbia, Vancouver, Canada
| | - David Potesil
- Proteomics Core Facility, Central European Institute of Technology, Masaryk University, Brno, Czech Republic
| | - Petr Kulhanek
- National Centre for Biomolecular Research, Faculty of Science, Masaryk University, Brno, Czech Republic
| | - Leonard J Foster
- Michael Smith Laboratories, University of British Columbia, Vancouver, Canada; Department of Biochemistry and Molecular Biology, University of British Columbia, Vancouver, Canada
| | - Pavel Bouchal
- Department of Biochemistry, Faculty of Science, Masaryk University, Brno, Czech Republic.
| |
Collapse
|
6
|
Bigos KJA, Quiles CG, Lunj S, Smith DJ, Krause M, Troost EGC, West CM, Hoskin P, Choudhury A. Tumour response to hypoxia: understanding the hypoxic tumour microenvironment to improve treatment outcome in solid tumours. Front Oncol 2024; 14:1331355. [PMID: 38352889 PMCID: PMC10861654 DOI: 10.3389/fonc.2024.1331355] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Accepted: 01/08/2024] [Indexed: 02/16/2024] Open
Abstract
Hypoxia is a common feature of solid tumours affecting their biology and response to therapy. One of the main transcription factors activated by hypoxia is hypoxia-inducible factor (HIF), which regulates the expression of genes involved in various aspects of tumourigenesis including proliferative capacity, angiogenesis, immune evasion, metabolic reprogramming, extracellular matrix (ECM) remodelling, and cell migration. This can negatively impact patient outcomes by inducing therapeutic resistance. The importance of hypoxia is clearly demonstrated by continued research into finding clinically relevant hypoxia biomarkers, and hypoxia-targeting therapies. One of the problems is the lack of clinically applicable methods of hypoxia detection, and lack of standardisation. Additionally, a lot of the methods of detecting hypoxia do not take into consideration the complexity of the hypoxic tumour microenvironment (TME). Therefore, this needs further elucidation as approximately 50% of solid tumours are hypoxic. The ECM is important component of the hypoxic TME, and is developed by both cancer associated fibroblasts (CAFs) and tumour cells. However, it is important to distinguish the different roles to develop both biomarkers and novel compounds. Fibronectin (FN), collagen (COL) and hyaluronic acid (HA) are important components of the ECM that create ECM fibres. These fibres are crosslinked by specific enzymes including lysyl oxidase (LOX) which regulates the stiffness of tumours and induces fibrosis. This is partially regulated by HIFs. The review highlights the importance of understanding the role of matrix stiffness in different solid tumours as current data shows contradictory results on the impact on therapeutic resistance. The review also indicates that further research is needed into identifying different CAF subtypes and their exact roles; with some showing pro-tumorigenic capacity and others having anti-tumorigenic roles. This has made it difficult to fully elucidate the role of CAFs within the TME. However, it is clear that this is an important area of research that requires unravelling as current strategies to target CAFs have resulted in worsened prognosis. The role of immune cells within the tumour microenvironment is also discussed as hypoxia has been associated with modulating immune cells to create an anti-tumorigenic environment. Which has led to the development of immunotherapies including PD-L1. These hypoxia-induced changes can confer resistance to conventional therapies, such as chemotherapy, radiotherapy, and immunotherapy. This review summarizes the current knowledge on the impact of hypoxia on the TME and its implications for therapy resistance. It also discusses the potential of hypoxia biomarkers as prognostic and predictive indictors of treatment response, as well as the challenges and opportunities of targeting hypoxia in clinical trials.
Collapse
Affiliation(s)
- Kamilla JA. Bigos
- Division of Cancer Sciences, University of Manchester, Manchester, United Kingdom
| | - Conrado G. Quiles
- Division of Cancer Sciences, University of Manchester, Manchester, United Kingdom
| | - Sapna Lunj
- Division of Cancer Sciences, University of Manchester, Manchester, United Kingdom
| | - Danielle J. Smith
- Division of Cancer Sciences, University of Manchester, Manchester, United Kingdom
| | - Mechthild Krause
- German Cancer Consortium (DKTK), partner site Dresden and German Cancer Research Center (DKFZ), Heidelberg, Germany
- Department of Radiation Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
- OncoRay – National Center for Radiation Research in Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Helmholtz-Zentrum Dresden - Rossendorf, Dresden, Germany
- Translational Radiooncology and Clinical Radiotherapy, Helmholtz-Zentrum Dresden - Rossendorf, Dresden, Germany
- Translational Radiation Oncology, National Center for Tumor Diseases (NCT), Partner Site Dresden, Dresden, Germany
- Translational Radiooncology and Clinical Radiotherapy and Image-guided High Precision Radiotherapy, German Cancer Research Center (DKFZ), Heidelberg, Germany
- Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
- Translational Radiooncology and Clinical Radiotherapy and Image-guided High Precision Radiotherapy, Helmholtz Association / Helmholtz-Zentrum Dresden - Rossendorf (HZDR), Dresden, Germany
- School of Medicine, Technische Universitat Dresden, Dresden, Germany
| | - Esther GC. Troost
- OncoRay – National Center for Radiation Research in Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Helmholtz-Zentrum Dresden - Rossendorf, Dresden, Germany
- Translational Radiation Oncology, National Center for Tumor Diseases (NCT), Partner Site Dresden, Dresden, Germany
- Translational Radiooncology and Clinical Radiotherapy and Image-guided High Precision Radiotherapy, German Cancer Research Center (DKFZ), Heidelberg, Germany
- Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
- Translational Radiooncology and Clinical Radiotherapy and Image-guided High Precision Radiotherapy, Helmholtz Association / Helmholtz-Zentrum Dresden - Rossendorf (HZDR), Dresden, Germany
- School of Medicine, Technische Universitat Dresden, Dresden, Germany
- Department of Radiotherapy and Radiation Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
- Institute of Radiooncology – OncoRay, Helmholtz-Zentrum Dresden-Rossendorf, Rossendorf, Germany
| | - Catharine M. West
- Division of Cancer Sciences, University of Manchester, Manchester Academic Health Science Centre, Christie Hospital, Manchester, United Kingdom
| | - Peter Hoskin
- Division of Cancer Sciences, University of Manchester, Manchester, United Kingdom
- Mount Vernon Cancer Centre, Northwood, United Kingdom
| | - Ananya Choudhury
- Division of Cancer Sciences, University of Manchester, Manchester, United Kingdom
- Christie Hospital NHS Foundation Trust, Manchester, Germany
| |
Collapse
|
7
|
Shen C, Bi Y, Chai W, Zhang Z, Yang S, Liu Y, Wu Z, Peng F, Fan Z, Hu H. Construction and validation of a metabolism-associated gene signature for predicting the prognosis, immune landscape, and drug sensitivity in bladder cancer. BMC Med Genomics 2023; 16:264. [PMID: 37880682 PMCID: PMC10601123 DOI: 10.1186/s12920-023-01678-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2023] [Accepted: 09/30/2023] [Indexed: 10/27/2023] Open
Abstract
Tumor Metabolism is strongly correlated with prognosis. Nevertheless, the prognostic and therapeutic value of metabolic-associated genes in BCa patients has not been fully elucidated. First, in this study, metabolism-related differential expressed genes DEGs with prognostic value in BCa were determined. Through the consensus clustering algorithm, we identified two molecular clusters with significantly different clinicopathological features and survival prognosis. Next, a novel metabolism-related prognostic model was established. Its reliable predictive performance in BCa was verified by multiple external datasets. Multivariate Cox analysis exhibited that risk score were independent prognostic factors. Interestingly, GSEA enrichment analysis of GO, KEGG, and Hallmark gene sets showed that the biological processes and pathways associated with ECM and collagen binding in the high-risk group were significantly enriched. Notely, the model was also significantly correlated with drug sensitivity, immune cell infiltration, and immunotherapy efficacy prediction by the wilcox rank test and chi-square test. Based on the 7 immune infiltration algorithm, we found that Neutrophils, Myeloid dendritic cells, M2 macrophages, Cancer-associated fibroblasts, etc., were more concentrated in the high-risk group. Additionally, in the IMvigor210, GSE111636, GSE176307, or our Truce01 (registration number NCT04730219) cohorts, the expression levels of multiple model genes were significantly correlated with objective responses to anti-PD-1/anti-PD-L1 immunotherapy. Finally, the expression of interested model genes were verified in 10 pairs of BCa tissues and para-carcinoma tissues by the HPA and real-time fluorescent quantitative PCR. Altogether, the signature established and validated by us has high predictive power for the prognosis, immunotherapy responsiveness, and chemotherapy sensitivity of BCa.
Collapse
Affiliation(s)
- Chong Shen
- Department of Urology, The Second Hospital of Tianjin Medical University, 23 Pingjiang Road, Jianshan Street, Hexi, Tianjin, 300211, People's Republic of China
- Tianjin Key Laboratory of Urology, Tianjin Institute of Urology, Tianjin, 300211, China
| | - Yuxin Bi
- Department of Urology, The Second Hospital of Tianjin Medical University, 23 Pingjiang Road, Jianshan Street, Hexi, Tianjin, 300211, People's Republic of China
- Tianjin Key Laboratory of Urology, Tianjin Institute of Urology, Tianjin, 300211, China
| | - Wang Chai
- Department of Urology, The Second Hospital of Tianjin Medical University, 23 Pingjiang Road, Jianshan Street, Hexi, Tianjin, 300211, People's Republic of China
- Tianjin Key Laboratory of Urology, Tianjin Institute of Urology, Tianjin, 300211, China
| | - Zhe Zhang
- Department of Urology, The Second Hospital of Tianjin Medical University, 23 Pingjiang Road, Jianshan Street, Hexi, Tianjin, 300211, People's Republic of China
- Tianjin Key Laboratory of Urology, Tianjin Institute of Urology, Tianjin, 300211, China
| | - Shaobo Yang
- Department of Urology, The Second Hospital of Tianjin Medical University, 23 Pingjiang Road, Jianshan Street, Hexi, Tianjin, 300211, People's Republic of China
- Tianjin Key Laboratory of Urology, Tianjin Institute of Urology, Tianjin, 300211, China
| | - Yuejiao Liu
- Department of Pharmacy, Zhu Xianyi Memorial Hospital of Tianjin Medical University, Tianjin, China
| | - Zhouliang Wu
- Department of Urology, The Second Hospital of Tianjin Medical University, 23 Pingjiang Road, Jianshan Street, Hexi, Tianjin, 300211, People's Republic of China
- Tianjin Key Laboratory of Urology, Tianjin Institute of Urology, Tianjin, 300211, China
| | - Fei Peng
- Department of Critical Care Medicine, the Peoples Hospital of Yuxi City, Yunnan, China
| | - Zhenqian Fan
- Department of Endocrinology, The Second Hospital of Tianjin Medical University, 23 Pingjiang Road, Jianshan Street, Hexi, Tianjin, 300211, People's Republic of China.
| | - Hailong Hu
- Department of Urology, The Second Hospital of Tianjin Medical University, 23 Pingjiang Road, Jianshan Street, Hexi, Tianjin, 300211, People's Republic of China.
- Tianjin Key Laboratory of Urology, Tianjin Institute of Urology, Tianjin, 300211, China.
| |
Collapse
|
8
|
Zheng L, Yang S, Xu R, Yang Y, Quan J, Lin Z, Quan C. NQO1 drives glioblastoma cell aggressiveness through EMT induction via the PI3K/Akt/mTOR/Snail pathway. Int J Oncol 2023; 63:110. [PMID: 37594082 PMCID: PMC10552716 DOI: 10.3892/ijo.2023.5558] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2022] [Accepted: 06/20/2023] [Indexed: 08/19/2023] Open
Abstract
Glioblastoma multiforme (GBM) is the most frequent and lethal cancer derived from the central nervous system, of which the mesenchymal (MES) subtype seriously influences the survival and prognosis of patients. NAD(P)H: quinone acceptor oxidoreductase 1 (NQO1) serves an important role in the carcinogenesis and progression of various types of cancer; however, the specific mechanism underlying the regulatory effects of NQO1 on GBM is unclear. Thus, the present study aimed to explore the role and mechanism of NQO1 in GBM progression. The results of bioinformatics analysis and immunohistochemistry showed that high expression of NQO1 was significantly related to the MES phenotype of GBM and shorter survival. In addition, MTT, colony formation, immunofluorescence and western blot analyses, and lung metastasis model experiments suggested that silencing NQO1 inhibited the proliferation and metastasis of GBM cells in vitro and in vivo. Furthermore, western blotting showed that the activity of the PI3K/Akt/mTOR signaling pathway was revealed to be inhibited by downregulation of NQO1 expression, whereas it was enhanced by overexpression of NQO1. Notably, co‑immunoprecipitation and ubiquitination experiments suggested that Snail was considered an important downstream target of NQO1 in GBM cells. Snail knockdown could eliminate the promoting effect of ectopic NQO1 on the proliferation and invasion of GBM cells, and reduce its effects on the activity of PI3K/Akt/mTOR signaling pathway. These results indicated that NQO1 could promote GBM aggressiveness by activating the PI3K/Akt/mTOR signaling pathway in a Snail‑dependent manner, and NQO1 and its relevant pathways may be considered novel targets for GBM therapy.
Collapse
Affiliation(s)
- Lan Zheng
- Central Laboratory, Affiliated Hospital of Yanbian University, Yanji, Jilin 133002
- Department of Obstetrics and Gynecology, Taizhou Hospital of Zhejiang Province Affiliated with Wenzhou Medical University, Linhai, Zhejiang 317000
- Key Laboratory of Pathobiology, State Ethnic Affairs Commission, Yanbian University, Yanji, Jilin 133000
| | - Shipeng Yang
- Central Laboratory, Affiliated Hospital of Yanbian University, Yanji, Jilin 133002
- Department of Pathology, Yanbian University Medical College, Yanji, Jilin 133000, P.R. China
| | - Ran Xu
- Key Laboratory of Pathobiology, State Ethnic Affairs Commission, Yanbian University, Yanji, Jilin 133000
- Department of Pathology, Yanbian University Medical College, Yanji, Jilin 133000, P.R. China
| | - Yang Yang
- Key Laboratory of Pathobiology, State Ethnic Affairs Commission, Yanbian University, Yanji, Jilin 133000
- Department of Pathology, Yanbian University Medical College, Yanji, Jilin 133000, P.R. China
| | - Jishu Quan
- Department of Pathology, Yanbian University Medical College, Yanji, Jilin 133000, P.R. China
| | - Zhenhua Lin
- Central Laboratory, Affiliated Hospital of Yanbian University, Yanji, Jilin 133002
- Key Laboratory of Pathobiology, State Ethnic Affairs Commission, Yanbian University, Yanji, Jilin 133000
- Department of Pathology, Yanbian University Medical College, Yanji, Jilin 133000, P.R. China
| | - Chunhua Quan
- Central Laboratory, Affiliated Hospital of Yanbian University, Yanji, Jilin 133002
- Key Laboratory of Pathobiology, State Ethnic Affairs Commission, Yanbian University, Yanji, Jilin 133000
| |
Collapse
|
9
|
Kumari S, Kumar P. Identification and characterization of putative biomarkers and therapeutic axis in Glioblastoma multiforme microenvironment. Front Cell Dev Biol 2023; 11:1236271. [PMID: 37538397 PMCID: PMC10395518 DOI: 10.3389/fcell.2023.1236271] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2023] [Accepted: 06/23/2023] [Indexed: 08/05/2023] Open
Abstract
Non-cellular secretory components, including chemokines, cytokines, and growth factors in the tumor microenvironment, are often dysregulated, impacting tumorigenesis in Glioblastoma multiforme (GBM) microenvironment, where the prognostic significance of the current treatment remains unsatisfactory. Recent studies have demonstrated the potential of post-translational modifications (PTM) and their respective enzymes, such as acetylation and ubiquitination in GBM etiology through modulating signaling events. However, the relationship between non-cellular secretory components and post-translational modifications will create a research void in GBM therapeutics. Therefore, we aim to bridge the gap between non-cellular secretory components and PTM modifications through machine learning and computational biology approaches. Herein, we highlighted the importance of BMP1, CTSB, LOX, LOXL1, PLOD1, MMP9, SERPINE1, and SERPING1 in GBM etiology. Further, we demonstrated the positive relationship between the E2 conjugating enzymes (Ube2E1, Ube2H, Ube2J2, Ube2C, Ube2J2, and Ube2S), E3 ligases (VHL and GNB2L1) and substrate (HIF1A). Additionally, we reported the novel HAT1-induced acetylation sites of Ube2S (K211) and Ube2H (K8, K52). Structural and functional characterization of Ube2S (8) and Ube2H (1) have identified their association with protein kinases. Lastly, our results found a putative therapeutic axis HAT1-Ube2S(K211)-GNB2L1-HIF1A and potential predictive biomarkers (CTSB, HAT1, Ube2H, VHL, and GNB2L1) that play a critical role in GBM pathogenesis.
Collapse
|
10
|
Shi P, Xu J, Cui H. The Recent Research Progress of NF-κB Signaling on the Proliferation, Migration, Invasion, Immune Escape and Drug Resistance of Glioblastoma. Int J Mol Sci 2023; 24:10337. [PMID: 37373484 PMCID: PMC10298967 DOI: 10.3390/ijms241210337] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2023] [Revised: 06/09/2023] [Accepted: 06/16/2023] [Indexed: 06/29/2023] Open
Abstract
Glioblastoma multiforme (GBM) is the most common and invasive primary central nervous system tumor in humans, accounting for approximately 45-50% of all primary brain tumors. How to conduct early diagnosis, targeted intervention, and prognostic evaluation of GBM, in order to improve the survival rate of glioblastoma patients, has always been an urgent clinical problem to be solved. Therefore, a deeper understanding of the molecular mechanisms underlying the occurrence and development of GBM is also needed. Like many other cancers, NF-κB signaling plays a crucial role in tumor growth and therapeutic resistance in GBM. However, the molecular mechanism underlying the high activity of NF-κB in GBM remains to be elucidated. This review aims to identify and summarize the NF-κB signaling involved in the recent pathogenesis of GBM, as well as basic therapy for GBM via NF-κB signaling.
Collapse
Affiliation(s)
- Pengfei Shi
- Cancer Center, Medical Research Institute, Southwest University, Chongqing 400716, China; (P.S.); (J.X.)
- Jinfeng Laboratory, Chongqing 401329, China
| | - Jie Xu
- Cancer Center, Medical Research Institute, Southwest University, Chongqing 400716, China; (P.S.); (J.X.)
- Jinfeng Laboratory, Chongqing 401329, China
| | - Hongjuan Cui
- Cancer Center, Medical Research Institute, Southwest University, Chongqing 400716, China; (P.S.); (J.X.)
- Jinfeng Laboratory, Chongqing 401329, China
- State Key Laboratory of Resource Insects, Southwest University, Chongqing 400716, China
| |
Collapse
|
11
|
Cao ZR, Zheng WX, Jiang YX, Chai H, Gong JH, Zhao MJ, Yan P, Liu YY, Liu XY, Huang ZT, Yang H, Peng DD, Zong KZ, Wu ZJ. miR-449a ameliorates acute rejection after liver transplantation via targeting procollagen-lysine1,2-oxoglutarate5-dioxygenase 1 in macrophages. Am J Transplant 2023; 23:336-352. [PMID: 36695693 DOI: 10.1016/j.ajt.2022.12.009] [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: 05/17/2022] [Revised: 12/06/2022] [Accepted: 12/07/2022] [Indexed: 01/13/2023]
Abstract
Acute rejection (AR) is an important factor that leads to poor prognosis after liver transplantation (LT). Macrophage M1-polarization is an important mechanism in AR development. MicroRNAs play vital roles in disease regulation; however, their effects on macrophages and AR remain unclear. In this study, rat models of AR were established following LT, and macrophages and peripheral blood mononuclear cells were isolated from rats and humans, respectively. We found miR-449a expression to be significantly reduced in macrophages and peripheral blood mononuclear cells. Overexpression of miR-449a not only inhibited the M1-polarization of macrophages in vitro but also improved the AR of transplant in vivo. The mechanism involved inhibiting the noncanonical nuclear factor-kappaB (NF-κB) pathway. We identified procollagen-lysine1,2-oxoglutarate5-dioxygenase 1 (PLOD1) as a target gene of miR-449a, which could reverse miR-449a's inhibition of macrophage M1-polarization, amelioration of AR, and inhibition of the NF-κB pathway. Overall, miR-449a inhibited the NF-κB pathway in macrophages through PLOD1 and also inhibited the M1-polarization of macrophages, thus attenuating AR after LT. In conclusion, miR-449a and PLOD1 may be new targets for the prevention and mitigation of AR.
Collapse
Affiliation(s)
- Zhen-Rui Cao
- Department of Hepatobiliary Surgery, the First Affiliated Hospital of Chongqing Medical University, Chongqing, People's Republic of China.
| | - Wei-Xiong Zheng
- Department of Hepatobiliary Surgery, the First Affiliated Hospital of Chongqing Medical University, Chongqing, People's Republic of China.
| | - Yu-Xin Jiang
- Department of Dermatology, the First Affiliated Hospital of Chongqing Medical University, Chongqing, People's Republic of China.
| | - Hao Chai
- Department of Hepatobiliary Surgery, the First Affiliated Hospital of Chongqing Medical University, Chongqing, People's Republic of China.
| | - Jun-Hua Gong
- Department of Hepatobiliary Surgery, the Second Affiliated Hospital of Chongqing Medical University, Chongqing, People's Republic of China.
| | - Min-Jie Zhao
- Department of Hepatobiliary Surgery, the Second Affiliated Hospital of Chongqing Medical University, Chongqing, People's Republic of China.
| | - Ping Yan
- Department of Hepatobiliary Surgery, the First Affiliated Hospital of Chongqing Medical University, Chongqing, People's Republic of China.
| | - Yan-Yao Liu
- Department of Hepatobiliary Surgery, the First Affiliated Hospital of Chongqing Medical University, Chongqing, People's Republic of China.
| | - Xiao-Ya Liu
- Department of Oncology, the First Affiliated Hospital of Chongqing Medical University, Chongqing, People's Republic of China.
| | - Zuo-Tian Huang
- Department of Hepatobiliary Surgery, the First Affiliated Hospital of Chongqing Medical University, Chongqing, People's Republic of China.
| | - Hang Yang
- Department of Hepatobiliary Surgery, the First Affiliated Hospital of Chongqing Medical University, Chongqing, People's Republic of China.
| | - Da-Di Peng
- Department of Hepatobiliary Surgery, the First Affiliated Hospital of Chongqing Medical University, Chongqing, People's Republic of China.
| | - Ke-Zhen Zong
- Department of Hepatobiliary Surgery, the First Affiliated Hospital of Chongqing Medical University, Chongqing, People's Republic of China.
| | - Zhong-Jun Wu
- Department of Hepatobiliary Surgery, the First Affiliated Hospital of Chongqing Medical University, Chongqing, People's Republic of China.
| |
Collapse
|
12
|
Yang Z, Zhou L, Si T, Chen S, Liu C, Ng KK, Wang Z, Chen Z, Qiu C, Liu G, Wang Q, Zhou X, Zhang L, Yao Z, He S, Yang M, Zhou Z. Lysyl hydroxylase LH1 promotes confined migration and metastasis of cancer cells by stabilizing Septin2 to enhance actin network. Mol Cancer 2023; 22:21. [PMID: 36721170 PMCID: PMC9887875 DOI: 10.1186/s12943-023-01727-9] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2022] [Accepted: 01/22/2023] [Indexed: 02/02/2023] Open
Abstract
BACKGROUND Excessive extracellular matrix deposition and increased stiffness are typical features of solid tumors such as hepatocellular carcinoma (HCC) and pancreatic ductal adenocarcinoma (PDAC). These conditions create confined spaces for tumor cell migration and metastasis. The regulatory mechanism of confined migration remains unclear. METHODS LC-MS was applied to determine the differentially expressed proteins between HCC tissues and corresponding adjacent tissue. Collective migration and single cell migration microfluidic devices with 6 μm-high confined channels were designed and fabricated to mimic the in vivo confined space. 3D invasion assay was created by Matrigel and Collagen I mixture treat to adherent cells. 3D spheroid formation under various stiffness environment was developed by different substitution percentage GelMA. Immunoprecipitation was performed to pull down the LH1-binding proteins, which were identified by LC-MS. Immunofluorescent staining, FRET, RT-PCR, Western blotting, FRAP, CCK-8, transwell cell migration, wound healing, orthotopic liver injection mouse model and in vivo imaging were used to evaluate the target expression and cellular phenotype. RESULTS Lysyl hydroxylase 1 (LH1) promoted the confined migration of cancer cells at both collective and single cell levels. In addition, LH1 enhanced cell invasion in a 3D biomimetic model and spheroid formation in stiffer environments. High LH1 expression correlated with poor prognosis of both HCC and PDAC patients, while it also promoted in vivo metastasis. Mechanistically, LH1 bound and stabilized Septin2 (SEPT2) to enhance actin polymerization, depending on the hydroxylase domain. Finally, the subpopulation with high expression of both LH1 and SEPT2 had the poorest prognosis. CONCLUSIONS LH1 promotes the confined migration and metastasis of cancer cells by stabilizing SEPT2 and thus facilitating actin polymerization.
Collapse
Affiliation(s)
- Zihan Yang
- grid.35030.350000 0004 1792 6846Department of Biomedical Sciences, and Tung Biomedical Sciences Center, City University of Hong Kong, 83 Tat Chee Avenue, Kowloon, Hong Kong SAR, People’s Republic of China ,grid.35030.350000 0004 1792 6846Department of Precision Diagnostic and Therapeutic Technology, City University of Hong Kong Futian Research Institute, Shenzhen, Guangdong China
| | - Li Zhou
- grid.35030.350000 0004 1792 6846Department of Biomedical Sciences, and Tung Biomedical Sciences Center, City University of Hong Kong, 83 Tat Chee Avenue, Kowloon, Hong Kong SAR, People’s Republic of China ,grid.412461.40000 0004 9334 6536Department of Gastroenterology, the Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Tongxu Si
- grid.35030.350000 0004 1792 6846Department of Biomedical Sciences, and Tung Biomedical Sciences Center, City University of Hong Kong, 83 Tat Chee Avenue, Kowloon, Hong Kong SAR, People’s Republic of China ,grid.35030.350000 0004 1792 6846Department of Precision Diagnostic and Therapeutic Technology, City University of Hong Kong Futian Research Institute, Shenzhen, Guangdong China
| | - Siyuan Chen
- grid.412461.40000 0004 9334 6536Department of Gastroenterology, the Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Chengxi Liu
- grid.16890.360000 0004 1764 6123State Key Laboratory of Chemical Biology and Drug Discovery, Research Institute for Future Food and Department of Applied Biology and Chemical Technology, The Hong Kong Polytechnic University, Hung Hom, Kowloon, Hong Kong SAR, China
| | - Kelvin Kaki Ng
- grid.35030.350000 0004 1792 6846Department of Biomedical Sciences, and Tung Biomedical Sciences Center, City University of Hong Kong, 83 Tat Chee Avenue, Kowloon, Hong Kong SAR, People’s Republic of China ,grid.35030.350000 0004 1792 6846Department of Precision Diagnostic and Therapeutic Technology, City University of Hong Kong Futian Research Institute, Shenzhen, Guangdong China
| | - Zesheng Wang
- grid.35030.350000 0004 1792 6846Department of Biomedical Sciences, and Tung Biomedical Sciences Center, City University of Hong Kong, 83 Tat Chee Avenue, Kowloon, Hong Kong SAR, People’s Republic of China ,grid.35030.350000 0004 1792 6846Department of Precision Diagnostic and Therapeutic Technology, City University of Hong Kong Futian Research Institute, Shenzhen, Guangdong China
| | - Zhiji Chen
- grid.412461.40000 0004 9334 6536Department of Gastroenterology, the Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Chan Qiu
- grid.412461.40000 0004 9334 6536Department of Gastroenterology, the Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Guopan Liu
- grid.35030.350000 0004 1792 6846Department of Biomedical Sciences, and Tung Biomedical Sciences Center, City University of Hong Kong, 83 Tat Chee Avenue, Kowloon, Hong Kong SAR, People’s Republic of China ,grid.35030.350000 0004 1792 6846Department of Precision Diagnostic and Therapeutic Technology, City University of Hong Kong Futian Research Institute, Shenzhen, Guangdong China
| | - Qingliang Wang
- grid.412461.40000 0004 9334 6536Department of Pathology, the Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Xiaoyu Zhou
- grid.35030.350000 0004 1792 6846Department of Biomedical Sciences, and Tung Biomedical Sciences Center, City University of Hong Kong, 83 Tat Chee Avenue, Kowloon, Hong Kong SAR, People’s Republic of China ,grid.35030.350000 0004 1792 6846Department of Precision Diagnostic and Therapeutic Technology, City University of Hong Kong Futian Research Institute, Shenzhen, Guangdong China
| | - Liang Zhang
- grid.35030.350000 0004 1792 6846Department of Biomedical Sciences, and Tung Biomedical Sciences Center, City University of Hong Kong, 83 Tat Chee Avenue, Kowloon, Hong Kong SAR, People’s Republic of China ,grid.35030.350000 0004 1792 6846Department of Precision Diagnostic and Therapeutic Technology, City University of Hong Kong Futian Research Institute, Shenzhen, Guangdong China
| | - Zhongping Yao
- grid.16890.360000 0004 1764 6123State Key Laboratory of Chemical Biology and Drug Discovery, Research Institute for Future Food and Department of Applied Biology and Chemical Technology, The Hong Kong Polytechnic University, Hung Hom, Kowloon, Hong Kong SAR, China
| | - Song He
- grid.412461.40000 0004 9334 6536Department of Gastroenterology, the Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Mengsu Yang
- grid.35030.350000 0004 1792 6846Department of Biomedical Sciences, and Tung Biomedical Sciences Center, City University of Hong Kong, 83 Tat Chee Avenue, Kowloon, Hong Kong SAR, People’s Republic of China ,grid.35030.350000 0004 1792 6846Department of Precision Diagnostic and Therapeutic Technology, City University of Hong Kong Futian Research Institute, Shenzhen, Guangdong China
| | - Zhihang Zhou
- grid.35030.350000 0004 1792 6846Department of Biomedical Sciences, and Tung Biomedical Sciences Center, City University of Hong Kong, 83 Tat Chee Avenue, Kowloon, Hong Kong SAR, People’s Republic of China ,grid.412461.40000 0004 9334 6536Department of Gastroenterology, the Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| |
Collapse
|
13
|
Hypoxia-induced ROS aggravate tumor progression through HIF-1α-SERPINE1 signaling in glioblastoma. J Zhejiang Univ Sci B 2023; 24:32-49. [PMID: 36632749 PMCID: PMC9837376 DOI: 10.1631/jzus.b2200269] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Hypoxia, as an important hallmark of the tumor microenvironment, is a major cause of oxidative stress and plays a central role in various malignant tumors, including glioblastoma. Elevated reactive oxygen species (ROS) in a hypoxic microenvironment promote glioblastoma progression; however, the underlying mechanism has not been clarified. Herein, we found that hypoxia promoted ROS production, and the proliferation, migration, and invasion of glioblastoma cells, while this promotion was restrained by ROS scavengers N-acetyl-L-cysteine (NAC) and diphenyleneiodonium chloride (DPI). Hypoxia-induced ROS activated hypoxia-inducible factor-1α (HIF-1α) signaling, which enhanced cell migration and invasion by epithelial-mesenchymal transition (EMT). Furthermore, the induction of serine protease inhibitor family E member 1 (SERPINE1) was ROS-dependent under hypoxia, and HIF-1α mediated SERPINE1 increase induced by ROS via binding to the SERPINE1 promoter region, thereby facilitating glioblastoma migration and invasion. Taken together, our data revealed that hypoxia-induced ROS reinforce the hypoxic adaptation of glioblastoma by driving the HIF-1α-SERPINE1 signaling pathway, and that targeting ROS may be a promising therapeutic strategy for glioblastoma.
Collapse
|
14
|
Miao Z, Zhao X, Liu X. Hypoxia induced β-catenin lactylation promotes the cell proliferation and stemness of colorectal cancer through the wnt signaling pathway. Exp Cell Res 2023; 422:113439. [PMID: 36464122 DOI: 10.1016/j.yexcr.2022.113439] [Citation(s) in RCA: 23] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2022] [Revised: 10/21/2022] [Accepted: 11/27/2022] [Indexed: 12/03/2022]
Abstract
Colorectal cancer (CRC) is a common malignant tumor of digestive system. Its incidence rate and mortality rate ranks the third among all the malignant tumors. The objective of this study was to explore the role of β-catenin in the CRC progression. The CRC tissues were collected to analyze the β-catenin levels. The CRC cells (SW620 and RRKO) were treated with hypoxia to simulate the hypoxic microenvironment of tumor in vitro. The β-catenin levels in the CRC cells were assessed with RT-qPCR, Western blot and Immunofluorescence. The cell biological behaviors were determined with CCK-8, flow cytometry and sphere formation assays. Besides, the glucose uptake, lactate production, ECAR and OCR was detected by seahorse. For the β-catenin lactylation determination, the IP and Western blot assay was performed. Then the protein stability of β-catenin was measured after cycloheximide treatment. The results showed that β-catenin was highly expressed in the CRC tissues and cells. Hypoxia treatment dramatically increased the protein levels and lactylation of β-catenin in the CRC cells. In addition, β-catenin knockdown dramatically inhibited the cell growth and stemness of the CRC cells. Besides, activation of Wnt signaling pathway neutralized the role of sh-β-catenin in the hypoxia treated CRC cells. In conclusion, this study confirmed that hypoxia induced the glycolysis promoted the β-catenin lactylation, which further enhanced the protein stability and expression of β-catenin, thus aggravating the malignant behaviors of CRC cells.
Collapse
Affiliation(s)
- Zhi Miao
- Frontier Science Center for Synthetic Biology and Key Laboratory of Systems Bioengineering (Ministry of Education), Tianjin University, Tianjin, 300072, China; Collaborative Innovation Center of Chemical Science and Engineering (Tianjin), School of Chemical Engineering and Technology, Tianjin University, Tianjin, 300072, China.
| | - Xiaomeng Zhao
- Frontier Science Center for Synthetic Biology and Key Laboratory of Systems Bioengineering (Ministry of Education), Tianjin University, Tianjin, 300072, China; Collaborative Innovation Center of Chemical Science and Engineering (Tianjin), School of Chemical Engineering and Technology, Tianjin University, Tianjin, 300072, China
| | - Xiang Liu
- Department of Laboratory Medicine, Wuhan Children's Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430016, China; The Key Laboratory for Biomedical Photonics of MOE at Wuhan National Laboratory for Optoelectronics-Hubei Bioinformatics & Molecular Imaging Key Laboratory, Systems Biology Theme, Department of Biomedical Engineering, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, 430074, China
| |
Collapse
|
15
|
Circular RNA circPTPRF promotes the progression of GBM via sponging miR-1208 to up-regulate YY1. Cancer Cell Int 2022; 22:359. [DOI: 10.1186/s12935-022-02753-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2022] [Accepted: 10/07/2022] [Indexed: 11/18/2022] Open
Abstract
AbstractGlioblastoma (GBM) is the most common primary malignant tumor in the brain, and its robust proliferation and invasion abilities reduce the survival time of patients. Circular RNAs (circRNAs) play an essential role in various tumors, such as regulating tumor cell proliferation, apoptosis, invasion, metastasis, and other progressive phenotypes through different mechanisms. Finding novel circRNAs may significantly contribute to the prognosis of GBM and provide the basis for the targeted therapy of GBM. In this study, we found circPTPRF is a novel circRNA that has never been studied, which was highly expressed in GBM and is closely related to poor patient prognoses. After knockdown or overexpression in glioma cell lines (U87 and LN229) and glioma stem cells (GSCs), we identified that circPTPRF could promote proliferation, invasion, and neurospheres formation abilities of GBM via in vitro and in vivo experiments. Mechanisms, miR-1208 was confirmed as a target of circPTPRF, and miR-1208 can also target the 3’UTR of YY1, and they were proved by luciferase reporter, western blotting (WB), qPCR and RNA immunoprecipitation (RIP) assays. The following rescue experiments demonstrated that circPTPRF was a miR-1208 sponge for upregulating YY1 expression to promote proliferation, invasion and neurosphere formation abilities of GBM in vitro. In conclusion, the circPTPRF/miR-1208/YY1 axis can regulate GBM progression. CircPTPRF may play an essential role in GBM diagnosis and prognostic prediction and be an important molecular target for GBM therapy.
Collapse
|
16
|
Gao Z, Xu J, Fan Y, Zhang Z, Wang H, Qian M, Zhang P, Deng L, Shen J, Xue H, Zhao R, Zhou T, Guo X, Li G. ARPC1B promotes mesenchymal phenotype maintenance and radiotherapy resistance by blocking TRIM21-mediated degradation of IFI16 and HuR in glioma stem cells. J Exp Clin Cancer Res 2022; 41:323. [PMID: 36380368 PMCID: PMC9667586 DOI: 10.1186/s13046-022-02526-8] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2022] [Accepted: 10/25/2022] [Indexed: 11/17/2022] Open
Abstract
Background Intratumoral heterogeneity is the primary challenge in the treatment of glioblastoma (GBM). The presence of glioma stem cells (GSCs) and their conversion between different molecular phenotypes contribute to the complexity of heterogeneity, culminating in preferential resistance to radiotherapy. ARP2/3 (actin-related protein-2/3) complexes (ARPs) are associated with cancer migration, invasion and differentiation, while the implications of ARPs in the phenotype and resistance to radiotherapy of GSCs remain unclear. Methods We screened the expression of ARPs in TCGA-GBM and CGGA-GBM databases. Tumor sphere formation assays and limiting dilution assays were applied to assess the implications of ARPC1B in tumorigenesis. Apoptosis, comet, γ-H2AX immunofluorescence (IF), and cell cycle distribution assays were used to evaluate the effect of ARPC1B on radiotherapy resistance. Immunoprecipitation (IP) and mass spectrometry analysis were used to detect ARPC1B-interacting proteins. Immune blot assays were performed to evaluate protein ubiquitination, and deletion mutant constructs were designed to determine the binding sites of protein interactions. The Spearman correlation algorithm was performed to screen for drugs that indicated cell sensitivity by the expression of ARPC1B. An intracranial xenograft GSC mouse model was used to investigate the role of ARPC1B in vivo. Results We concluded that ARPC1B was significantly upregulated in MES-GBM/GSCs and was correlated with a poor prognosis. Both in vitro and in vivo assays indicated that knockdown of ARPC1B in MES-GSCs reduced tumorigenicity and resistance to IR treatment, whereas overexpression of ARPC1B in PN-GSCs exhibited the opposite effects. Mechanistically, ARPC1B interacted with IFI16 and HuR to maintain protein stability. In detail, the Pyrin of IFI16 and RRM2 of HuR were implicated in binding to ARPC1B, which counteracted TRIM21-mediated degradation of ubiquitination to IFI16 and HuR. Additionally, the function of ARPC1B was dependent on IFI16-induced activation of NF-κB pathway and HuR-induced activation of STAT3 pathway. Finally, we screened AZD6738, an ataxia telangiectasia mutated and rad3-related (ATR) inhibitor, based on the expression of ARPC1B. In addition to ARPC1B expression reflecting cellular sensitivity to AZD6738, the combination of AZD6738 and radiotherapy exhibited potent antitumor effects both in vitro and in vivo. Conclusion ARPC1B promoted MES phenotype maintenance and radiotherapy resistance by inhibiting TRIM21-mediated degradation of IFI16 and HuR, thereby activating the NF-κB and STAT3 signaling pathways, respectively. AZD6738, identified based on ARPC1B expression, exhibited excellent anti-GSC activity in combination with radiotherapy. Supplementary Information The online version contains supplementary material available at 10.1186/s13046-022-02526-8.
Collapse
|
17
|
Fan Y, Gao Z, Xu J, Wang H, Guo Q, Xue H, Zhao R, Guo X, Li G. Identification and validation of SNHG gene signature to predict malignant behaviors and therapeutic responses in glioblastoma. Front Immunol 2022; 13:986615. [PMID: 36159816 PMCID: PMC9493242 DOI: 10.3389/fimmu.2022.986615] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2022] [Accepted: 08/22/2022] [Indexed: 11/13/2022] Open
Abstract
Glioblastoma (GBM) patients exhibit high mortality and recurrence rates despite multimodal therapy. Small nucleolar RNA host genes (SNHGs) are a group of long noncoding RNAs that perform a wide range of biological functions. We aimed to reveal the role of SNHGs in GBM subtypes, cell infiltration into the tumor microenvironment (TME), and stemness characteristics. SNHG interaction patterns were determined based on 25 SNHGs and systematically correlated with GBM subtypes, TME and stemness characteristics. The SNHG interaction score (SNHGscore) model was generated to quantify SNHG interaction patterns. The high SNHGscore group was characterized by a poor prognosis, the mesenchymal (MES) subtype, the infiltration of suppressive immune cells and a differentiated phenotype. Further analysis indicated that high SNHGscore was associated with a weaker response to anti-PD-1/L1 immunotherapy. Tumor cells with high SNHG scores were more sensitive to drugs targeting the EGFR and ERK-MAPK signaling pathways. Finally, we assessed SNHG interaction patterns in multiple cancers to verify their universality. This is a novel and comprehensive study that provides targeted therapeutic strategies based on SNHG interactions. Our work highlights the crosstalk and potential clinical utility of SNHG interactions in cancer therapy.
Collapse
Affiliation(s)
- Yang Fan
- Department of Neurosurgery, Qilu Hospital, Cheeloo College of Medicine and Institute of Brain and Brain-Inspired Science, Shandong University, Jinan, China
- Shandong Key Laboratory of Brain Function Remodeling, Jinan, China
| | - Zijie Gao
- Department of Neurosurgery, Qilu Hospital, Cheeloo College of Medicine and Institute of Brain and Brain-Inspired Science, Shandong University, Jinan, China
- Shandong Key Laboratory of Brain Function Remodeling, Jinan, China
| | - Jianye Xu
- Department of Neurosurgery, Qilu Hospital, Cheeloo College of Medicine and Institute of Brain and Brain-Inspired Science, Shandong University, Jinan, China
- Shandong Key Laboratory of Brain Function Remodeling, Jinan, China
| | - Huizhi Wang
- Department of Neurosurgery, Qilu Hospital, Cheeloo College of Medicine and Institute of Brain and Brain-Inspired Science, Shandong University, Jinan, China
- Shandong Key Laboratory of Brain Function Remodeling, Jinan, China
| | - Qindong Guo
- Department of Neurosurgery, Qilu Hospital, Cheeloo College of Medicine and Institute of Brain and Brain-Inspired Science, Shandong University, Jinan, China
- Shandong Key Laboratory of Brain Function Remodeling, Jinan, China
| | - Hao Xue
- Department of Neurosurgery, Qilu Hospital, Cheeloo College of Medicine and Institute of Brain and Brain-Inspired Science, Shandong University, Jinan, China
- Shandong Key Laboratory of Brain Function Remodeling, Jinan, China
| | - Rongrong Zhao
- Department of Neurosurgery, Qilu Hospital, Cheeloo College of Medicine and Institute of Brain and Brain-Inspired Science, Shandong University, Jinan, China
- Shandong Key Laboratory of Brain Function Remodeling, Jinan, China
| | - Xing Guo
- Department of Neurosurgery, Qilu Hospital, Cheeloo College of Medicine and Institute of Brain and Brain-Inspired Science, Shandong University, Jinan, China
- Shandong Key Laboratory of Brain Function Remodeling, Jinan, China
- *Correspondence: Xing Guo, ; Gang Li,
| | - Gang Li
- Department of Neurosurgery, Qilu Hospital, Cheeloo College of Medicine and Institute of Brain and Brain-Inspired Science, Shandong University, Jinan, China
- Shandong Key Laboratory of Brain Function Remodeling, Jinan, China
- *Correspondence: Xing Guo, ; Gang Li,
| |
Collapse
|
18
|
Loers JU, Vermeirssen V. SUBATOMIC: a SUbgraph BAsed mulTi-OMIcs clustering framework to analyze integrated multi-edge networks. BMC Bioinformatics 2022; 23:363. [PMID: 36064320 PMCID: PMC9442970 DOI: 10.1186/s12859-022-04908-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2022] [Accepted: 08/24/2022] [Indexed: 11/02/2022] Open
Abstract
BACKGROUND Representing the complex interplay between different types of biomolecules across different omics layers in multi-omics networks bears great potential to gain a deep mechanistic understanding of gene regulation and disease. However, multi-omics networks easily grow into giant hairball structures that hamper biological interpretation. Module detection methods can decompose these networks into smaller interpretable modules. However, these methods are not adapted to deal with multi-omics data nor consider topological features. When deriving very large modules or ignoring the broader network context, interpretability remains limited. To address these issues, we developed a SUbgraph BAsed mulTi-OMIcs Clustering framework (SUBATOMIC), which infers small and interpretable modules with a specific topology while keeping track of connections to other modules and regulators. RESULTS SUBATOMIC groups specific molecular interactions in composite network subgraphs of two and three nodes and clusters them into topological modules. These are functionally annotated, visualized and overlaid with expression profiles to go from static to dynamic modules. To preserve the larger network context, SUBATOMIC investigates statistically the connections in between modules as well as between modules and regulators such as miRNAs and transcription factors. We applied SUBATOMIC to analyze a composite Homo sapiens network containing transcription factor-target gene, miRNA-target gene, protein-protein, homologous and co-functional interactions from different databases. We derived and annotated 5586 modules with diverse topological, functional and regulatory properties. We created novel functional hypotheses for unannotated genes. Furthermore, we integrated modules with condition specific expression data to study the influence of hypoxia in three cancer cell lines. We developed two prioritization strategies to identify the most relevant modules in specific biological contexts: one considering GO term enrichments and one calculating an activity score reflecting the degree of differential expression. Both strategies yielded modules specifically reacting to low oxygen levels. CONCLUSIONS We developed the SUBATOMIC framework that generates interpretable modules from integrated multi-omics networks and applied it to hypoxia in cancer. SUBATOMIC can infer and contextualize modules, explore condition or disease specific modules, identify regulators and functionally related modules, and derive novel gene functions for uncharacterized genes. The software is available at https://github.com/CBIGR/SUBATOMIC .
Collapse
Affiliation(s)
- Jens Uwe Loers
- Lab for Computational Biology, Integromics and Gene Regulation (CBIGR), Cancer Research Institute Ghent (CRIG), Ghent, Belgium.,Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium.,Department of Biomolecular Medicine, Ghent University, Ghent, Belgium
| | - Vanessa Vermeirssen
- Lab for Computational Biology, Integromics and Gene Regulation (CBIGR), Cancer Research Institute Ghent (CRIG), Ghent, Belgium. .,Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium. .,Department of Biomolecular Medicine, Ghent University, Ghent, Belgium.
| |
Collapse
|
19
|
Liu Y, Zhang X, Jiang T, Du N. Hypoxia-Induced Nestin Regulates Viability and Metabolism of Lung Cancer by Targeting Transcriptional Factor Nrf2, STAT3, and SOX2. COMPUTATIONAL INTELLIGENCE AND NEUROSCIENCE 2022; 2022:9811905. [PMID: 36082356 PMCID: PMC9448566 DOI: 10.1155/2022/9811905] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Revised: 08/13/2022] [Accepted: 08/16/2022] [Indexed: 11/29/2022]
Abstract
Objective To investigate hypoxia-induced Nestin regulates lung cancer viability and metabolism by targeting transcription factors Nrf2, STAT3, and SOX2. Methods Eighty-four cases of nonsmall cell lung cancer (nonsmall cell lung cancer, NSCLC), which had been treated from June 2020 to February 2021, were randomly selected from our clinicopathology database. Immunohistochemical staining of collected tissue cells was performed to assess the expression patterns of Nestin, STAT3, Nrf2, and SOX2. Data were quantified and statistically analyzed using one-way and two-way ANOVA tests with P < 0.05. Results Clinicopathological findings showed significant differences in lymph node metastasis, tissue differentiation, and histology on induction of Nestin expression; Nestin expression correlated with STAT3, Nrf2, and SOX2 expression.Nestin/STAT3/SOX2/Nrf2 are involved in angiogenesis and lung cancer development. Conclusion Hypoxia-induced Nestin promotes the progression of nonsmall lung cancer cells by targeting the downstream transcription factors STAT3, Nrf2, and SOX2.
Collapse
Affiliation(s)
- Yongshi Liu
- Department of Thoracic Surgery, Tangdu Hospital, Air Force Medical University, Xi'an, Shaanxi, China
| | - Xinglin Zhang
- Department of Oncology, Qingdao Municipal Hospital, Qingdao, Shandong, China
| | - Tao Jiang
- Department of Thoracic Surgery, Tangdu Hospital, Air Force Medical University, Xi'an, Shaanxi, China
| | - Ning Du
- Department of Thoracic Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
| |
Collapse
|
20
|
Scietti L, Moroni E, Mattoteia D, Fumagalli M, De Marco M, Negro L, Chiapparino A, Serapian SA, De Giorgi F, Faravelli S, Colombo G, Forneris F. A Fe2+-dependent self-inhibited state influences the druggability of human collagen lysyl hydroxylase (LH/PLOD) enzymes. Front Mol Biosci 2022; 9:876352. [PMID: 36090047 PMCID: PMC9453210 DOI: 10.3389/fmolb.2022.876352] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Accepted: 07/11/2022] [Indexed: 11/13/2022] Open
Abstract
Multifunctional human collagen lysyl hydroxylase (LH/PLOD) enzymes catalyze post-translational hydroxylation and subsequent glycosylation of collagens, enabling their maturation and supramolecular organization in the extracellular matrix (ECM). Recently, the overexpression of LH/PLODs in the tumor microenvironment results in abnormal accumulation of these collagen post-translational modifications, which has been correlated with increased metastatic progression of a wide variety of solid tumors. These observations make LH/PLODs excellent candidates for prospective treatment of aggressive cancers. The recent years have witnessed significant research efforts to facilitate drug discovery on LH/PLODs, including molecular structure characterizations and development of reliable high-throughput enzymatic assays. Using a combination of biochemistry and in silico studies, we characterized the dual role of Fe2+ as simultaneous cofactor and inhibitor of lysyl hydroxylase activity and studied the effect of a promiscuous Fe2+ chelating agent, 2,2’-bipyridil, broadly considered a lysyl hydroxylase inhibitor. We found that at low concentrations, 2,2’-bipyridil unexpectedly enhances the LH enzymatic activity by reducing the inhibitory effect of excess Fe2+. Together, our results show a fine balance between Fe2+-dependent enzymatic activity and Fe2+-induced self-inhibited states, highlighting exquisite differences between LH/PLODs and related Fe2+, 2-oxoglutarate dioxygenases and suggesting that conventional structure-based approaches may not be suited for successful inhibitor development. These insights address outstanding questions regarding druggability of LH/PLOD lysyl hydroxylase catalytic site and provide a solid ground for upcoming drug discovery and screening campaigns.
Collapse
Affiliation(s)
- Luigi Scietti
- The Armenise-Harvard Laboratory of Structural Biology, Department of Biology and Biotechnology, University of Pavia, Pavia, Italy
- *Correspondence: Luigi Scietti, ; Federico Forneris,
| | - Elisabetta Moroni
- Consiglio Nazionale delle Ricerche, Istituto di Scienze e Tecnologie Chimiche “Giulio Natta” (SCITEC-CNR), Milano, Italy
| | - Daiana Mattoteia
- The Armenise-Harvard Laboratory of Structural Biology, Department of Biology and Biotechnology, University of Pavia, Pavia, Italy
| | - Marco Fumagalli
- The Armenise-Harvard Laboratory of Structural Biology, Department of Biology and Biotechnology, University of Pavia, Pavia, Italy
| | - Matteo De Marco
- The Armenise-Harvard Laboratory of Structural Biology, Department of Biology and Biotechnology, University of Pavia, Pavia, Italy
| | - Lisa Negro
- The Armenise-Harvard Laboratory of Structural Biology, Department of Biology and Biotechnology, University of Pavia, Pavia, Italy
| | - Antonella Chiapparino
- The Armenise-Harvard Laboratory of Structural Biology, Department of Biology and Biotechnology, University of Pavia, Pavia, Italy
| | | | - Francesca De Giorgi
- The Armenise-Harvard Laboratory of Structural Biology, Department of Biology and Biotechnology, University of Pavia, Pavia, Italy
| | - Silvia Faravelli
- The Armenise-Harvard Laboratory of Structural Biology, Department of Biology and Biotechnology, University of Pavia, Pavia, Italy
| | | | - Federico Forneris
- The Armenise-Harvard Laboratory of Structural Biology, Department of Biology and Biotechnology, University of Pavia, Pavia, Italy
- *Correspondence: Luigi Scietti, ; Federico Forneris,
| |
Collapse
|
21
|
Lv X, Li Q, Liu H, Gong M, Zhao Y, Hu J, Wu F, Wu X. JUN
activation modulates chromatin accessibility to drive
TNFα
‐induced mesenchymal transition in glioblastoma. J Cell Mol Med 2022; 26:4602-4612. [PMID: 35851726 PMCID: PMC9357637 DOI: 10.1111/jcmm.17490] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2022] [Revised: 06/13/2022] [Accepted: 07/05/2022] [Indexed: 11/27/2022] Open
Abstract
Chromatin dynamics as well as genetic evolution underlies the adaptability of tumour cells to environmental cues. Three subtypes of tumour cells have been identified in glioblastoma, one of the commonest malignant brain tumours in adults. During tumour progression or under therapeutic pressure, the non‐mesenchymal subtypes may progress to the mesenchymal subtype, leading to unfavourable prognosis. However, the molecular mechanisms for this transition remain poorly understood. Here taking a TNFα‐induced cellular model, we profile the chromatin accessibility dynamics during mesenchymal transition. Moreover, we identify the JUN family as one of the key driving transcription factors for the gained chromatin accessibility. Accordingly, inhibition of JUN phosphorylation and therefore its transcription activity successfully impedes TNFα‐induced chromatin remodelling and mesenchymal transition. In line with these findings based on experimental models, JUN activity is positively correlated with mesenchymal features in clinical glioblastoma specimens. Together, this study unveils a deregulated transcription regulatory network in glioblastoma progression and hopefully provides a rationale for anti‐glioblastoma therapy.
Collapse
Affiliation(s)
- Xuejiao Lv
- State Key Laboratory of Experimental Hematology, The Province and Ministry Co‐sponsored Collaborative Innovation Center for Medical Epigenetics, Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), Department of Cell Biology Tianjin Medical University Tianjin China
| | - Qian Li
- State Key Laboratory of Experimental Hematology, The Province and Ministry Co‐sponsored Collaborative Innovation Center for Medical Epigenetics, Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), Department of Cell Biology Tianjin Medical University Tianjin China
| | - Hang Liu
- State Key Laboratory of Experimental Hematology, The Province and Ministry Co‐sponsored Collaborative Innovation Center for Medical Epigenetics, Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), Department of Cell Biology Tianjin Medical University Tianjin China
| | - Meihan Gong
- State Key Laboratory of Experimental Hematology, The Province and Ministry Co‐sponsored Collaborative Innovation Center for Medical Epigenetics, Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), Department of Cell Biology Tianjin Medical University Tianjin China
| | - Yingying Zhao
- State Key Laboratory of Experimental Hematology, The Province and Ministry Co‐sponsored Collaborative Innovation Center for Medical Epigenetics, Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), Department of Cell Biology Tianjin Medical University Tianjin China
| | - Jinyang Hu
- Department of Neurosurgery Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology Wuhan China
| | - Fan Wu
- Department of Molecular Neuropathology Beijing Neurosurgical Institute, Capital Medical University Beijing China
| | - Xudong Wu
- State Key Laboratory of Experimental Hematology, The Province and Ministry Co‐sponsored Collaborative Innovation Center for Medical Epigenetics, Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), Department of Cell Biology Tianjin Medical University Tianjin China
- Department of Neurosurgery Tianjin Medical University General Hospital Tianjin China
| |
Collapse
|
22
|
Mirzaei S, Saghari S, Bassiri F, Raesi R, Zarrabi A, Hushmandi K, Sethi G, Tergaonkar V. NF-κB as a regulator of cancer metastasis and therapy response: A focus on epithelial-mesenchymal transition. J Cell Physiol 2022; 237:2770-2795. [PMID: 35561232 DOI: 10.1002/jcp.30759] [Citation(s) in RCA: 82] [Impact Index Per Article: 41.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2022] [Revised: 03/24/2022] [Accepted: 04/19/2022] [Indexed: 12/13/2022]
Abstract
Metastasis of tumor cells is a complex challenge and significantly diminishes the overall survival and prognosis of cancer patients. The epithelial-to-mesenchymal transition (EMT) is a well-known mechanism responsible for the invasiveness of tumor cells. A number of molecular pathways can regulate the EMT mechanism in cancer cells and nuclear factor-kappaB (NF-κB) is one of them. The nuclear translocation of NF-κB p65 can induce the transcription of several genes involved in EMT induction. The present review describes NF-κB and EMT interaction in cancer cells and their association in cancer progression. Due to the oncogenic role NF-κB signaling, its activation enhances metastasis of tumor cells via EMT induction. This has been confirmed in various cancers including brain, breast, lung and gastric cancers, among others. The ZEB1/2, transforming growth factor-β, and Slug as inducers of EMT undergo upregulation by NF-κB to promote metastasis of tumor cells. After EMT induction driven by NF-κB, a significant decrease occurs in E-cadherin levels, while N-cadherin and vimentin levels undergo an increase. The noncoding RNAs can potentially also function as upstream mediators and modulate NF-κB/EMT axis in cancers. Moreover, NF-κB/EMT axis is involved in mediating drug resistance in tumor cells. Thus, suppressing NF-κB/EMT axis can also promote the sensitivity of cancer cells to chemotherapeutic agents.
Collapse
Affiliation(s)
- Sepideh Mirzaei
- Department of Biology, Faculty of Science, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | - Sam Saghari
- Department of Health Services Management, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Farzaneh Bassiri
- Department of Biology, Fars Science and Research Branch, Islamic Azad University, Fars, Iran.,Department of Biology, Shiraz Branch, Islamic Azad University, Shiraz, Iran
| | - Rasoul Raesi
- PhD in Health Services Management, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Ali Zarrabi
- Department of Biomedical Engineering, Faculty of Engineering and Natural Sciences, Istinye University, Istanbul, Turkey
| | - Kiavash Hushmandi
- Department of Food Hygiene and Quality Control, Division of Epidemiology and Zoonoses, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran
| | - Gautam Sethi
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.,Yong Loo Lin School of Medicine, NUS Centre for Cancer Research (N2CR), National University of Singapore, Singapore, Singapore
| | - Vinay Tergaonkar
- Laboratory of NF-κB Signaling, Institute of Molecular and Cell Biology (IMCB), 61 Biopolis Drive, Proteos, Singapore, Singapore.,Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| |
Collapse
|
23
|
Gong S, Schopow N, Duan Y, Wu C, Kallendrusch S, Osterhoff G. PLOD Family: A Novel Biomarker for Prognosis and Personalized Treatment in Soft Tissue Sarcoma. Genes (Basel) 2022; 13:genes13050787. [PMID: 35627171 PMCID: PMC9141206 DOI: 10.3390/genes13050787] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2022] [Revised: 04/26/2022] [Accepted: 04/27/2022] [Indexed: 11/16/2022] Open
Abstract
Despite various treatment attempts, the heterogenous group of soft tissue sarcomata (STS) with more than 100 subtypes still shows poor outcomes. Therefore, effective biomarkers for prognosis prediction and personalized treatment are of high importance. The Procollagen-Lysine, 2-Oxoglutarate 5-Dioxygenase (PLOD) gene family, which is related to multiple cancer entities, consists of three members which encode important enzymes for the formation of connective tissue. The relation to STS, however, has not yet been explored. In this study, data from The Cancer Genome Atlas (TCGA) and Genotype-Tissue Expression (GTEx) databases were used to analyze the role of PLOD1–3 in STS. It was found that an overexpression of PLOD family members correlates with poor prognosis, which might be due to an increased infiltration of immune-related cells in the tumor microenvironment. In STS, the expression of PLOD genes could be a novel biomarker for prognosis and a personalized, more aggressive treatment in these patients.
Collapse
Affiliation(s)
- Siming Gong
- Institute of Anatomy, University of Leipzig, Liebigstraße 13, 04103 Leipzig, Germany; (S.G.); (N.S.); (S.K.)
| | - Nikolas Schopow
- Institute of Anatomy, University of Leipzig, Liebigstraße 13, 04103 Leipzig, Germany; (S.G.); (N.S.); (S.K.)
- Sarcoma Center, Department for Orthopedics, Trauma Surgery and Reconstructive Surgery, University Hospital Leipzig, Liebigstraße 20, 04103 Leipzig, Germany;
| | - Yingjuan Duan
- Faculty of Chemistry and Mineralogy, University of Leipzig, Johannisallee 29, 04103 Leipzig, Germany;
| | - Changwu Wu
- Institute of Anatomy, University of Leipzig, Liebigstraße 13, 04103 Leipzig, Germany; (S.G.); (N.S.); (S.K.)
- Correspondence: or
| | - Sonja Kallendrusch
- Institute of Anatomy, University of Leipzig, Liebigstraße 13, 04103 Leipzig, Germany; (S.G.); (N.S.); (S.K.)
- Department of Medicine, Health and Medical University Potsdam, Olympischer Weg 1, 14471 Potsdam, Germany
| | - Georg Osterhoff
- Sarcoma Center, Department for Orthopedics, Trauma Surgery and Reconstructive Surgery, University Hospital Leipzig, Liebigstraße 20, 04103 Leipzig, Germany;
| |
Collapse
|
24
|
Li B, Xian X, Lin X, Huang L, Liang A, Jiang H, Gong Q. Hypoxia Alters the Proteome Profile and Enhances the Angiogenic Potential of Dental Pulp Stem Cell-Derived Exosomes. Biomolecules 2022; 12:biom12040575. [PMID: 35454164 PMCID: PMC9029684 DOI: 10.3390/biom12040575] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Revised: 04/12/2022] [Accepted: 04/12/2022] [Indexed: 12/12/2022] Open
Abstract
Dental pulp stem cells (DPSCs) and their exosomes (Exos) are effective treatments for regenerative medicine. Hypoxia was confirmed to improve the angiogenic potential of stem cells. However, the angiogenic effect and mechanism of hypoxia-preconditioned DPSC-Exos are poorly understood. We isolated exosomes from DPSCs under normoxia (Nor-Exos) and hypoxia (Hypo-Exos) and added them to human umbilical vein endothelial cells (HUVECs). HUVEC proliferation, migration and angiogenic capacity were assessed by CCK-8, transwell, tube formation assays, qRT-PCR and Western blot. iTRAQ-based proteomics and bioinformatic analysis were performed to investigate proteome profile differences between Nor-Exos and Hypo-Exos. Western blot, immunofluorescence and immunohistochemistry were used to detect the expression of lysyl oxidase-like 2 (LOXL2) in vitro and in vivo. Finally, we silenced LOXL2 in HUVECs and rescued tube formation with Hypo-Exos. Hypo-Exos enhanced HUVEC proliferation, migration and tube formation in vitro superior to Nor-Exos. The proteomics analysis identified 79 proteins with significantly different expression in Hypo-Exos, among which LOXL2 was verified as being upregulated in hypoxia-preconditioned DPSCs, Hypo-Exos, and inflamed dental pulp. Hypo-Exos partially rescued the inhibitory influence of LOXL2 silence on HUVEC tube formation. In conclusion, hypoxia enhanced the angiogenic potential of DPSCs-Exos and partially altered their proteome profile. LOXL2 is likely involved in Hypo-Exos mediated angiogenesis.
Collapse
Affiliation(s)
- Baoyu Li
- Hospital of Stomatology, Sun Yat-Sen University, Guangzhou 510055, China; (B.L.); (X.L.); (L.H.); (A.L.)
- Guangdong Provincial Key Laboratory of Stomatology, Guangzhou 510080, China
- Guanghua School of Stomatology, Sun Yat-Sen University, Guangzhou 510055, China
| | - Xuehong Xian
- Department of Stomatology, The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou 510655, China;
- Foshan Stomatological Hospital, Foshan University, Foshan 528000, China
| | - Xinwei Lin
- Hospital of Stomatology, Sun Yat-Sen University, Guangzhou 510055, China; (B.L.); (X.L.); (L.H.); (A.L.)
- Guangdong Provincial Key Laboratory of Stomatology, Guangzhou 510080, China
- Guanghua School of Stomatology, Sun Yat-Sen University, Guangzhou 510055, China
| | - Luo Huang
- Hospital of Stomatology, Sun Yat-Sen University, Guangzhou 510055, China; (B.L.); (X.L.); (L.H.); (A.L.)
- Guangdong Provincial Key Laboratory of Stomatology, Guangzhou 510080, China
- Guanghua School of Stomatology, Sun Yat-Sen University, Guangzhou 510055, China
| | - Ailin Liang
- Hospital of Stomatology, Sun Yat-Sen University, Guangzhou 510055, China; (B.L.); (X.L.); (L.H.); (A.L.)
- Guangdong Provincial Key Laboratory of Stomatology, Guangzhou 510080, China
- Guanghua School of Stomatology, Sun Yat-Sen University, Guangzhou 510055, China
| | - Hongwei Jiang
- Hospital of Stomatology, Sun Yat-Sen University, Guangzhou 510055, China; (B.L.); (X.L.); (L.H.); (A.L.)
- Guangdong Provincial Key Laboratory of Stomatology, Guangzhou 510080, China
- Guanghua School of Stomatology, Sun Yat-Sen University, Guangzhou 510055, China
- Correspondence: (H.J.); (Q.G.)
| | - Qimei Gong
- Hospital of Stomatology, Sun Yat-Sen University, Guangzhou 510055, China; (B.L.); (X.L.); (L.H.); (A.L.)
- Guangdong Provincial Key Laboratory of Stomatology, Guangzhou 510080, China
- Guanghua School of Stomatology, Sun Yat-Sen University, Guangzhou 510055, China
- Correspondence: (H.J.); (Q.G.)
| |
Collapse
|
25
|
Markwell SM, Ross JL, Olson CL, Brat DJ. Necrotic reshaping of the glioma microenvironment drives disease progression. Acta Neuropathol 2022; 143:291-310. [PMID: 35039931 DOI: 10.1007/s00401-021-02401-4] [Citation(s) in RCA: 29] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2021] [Revised: 12/20/2021] [Accepted: 12/21/2021] [Indexed: 12/13/2022]
Abstract
Glioblastoma is the most common primary brain tumor and has a dismal prognosis. The development of central necrosis represents a tipping point in the evolution of these tumors that foreshadows aggressive expansion, swiftly leading to mortality. The onset of necrosis, severe hypoxia and associated radial glioma expansion correlates with dramatic tumor microenvironment (TME) alterations that accelerate tumor growth. In the past, most have concluded that hypoxia and necrosis must arise due to "cancer outgrowing its blood supply" when rapid tumor growth outpaces metabolic supply, leading to diffusion-limited hypoxia. However, growing evidence suggests that microscopic intravascular thrombosis driven by the neoplastic overexpression of pro-coagulants attenuates glioma blood supply (perfusion-limited hypoxia), leading to TME restructuring that includes breakdown of the blood-brain barrier, immunosuppressive immune cell accumulation, microvascular hyperproliferation, glioma stem cell enrichment and tumor cell migration outward. Cumulatively, these adaptations result in rapid tumor expansion, resistance to therapeutic interventions and clinical progression. To inform future translational investigations, the complex interplay among environmental cues and myriad cell types that contribute to this aggressive phenotype requires better understanding. This review focuses on contributions from intratumoral thrombosis, the effects of hypoxia and necrosis, the adaptive and innate immune responses, and the current state of targeted therapeutic interventions.
Collapse
Affiliation(s)
- Steven M Markwell
- Department of Pathology, Northwestern Medicine Malnati Brain Tumor Institute of the Robert H. Lurie Comprehensive Cancer Center, Northwestern University Feinberg School of Medicine, 303 E. Chicago Ave. Ward 3-140, Chicago, IL, USA
| | - James L Ross
- Department of Microbiology and Immunology, Emory University, Atlanta, GA, USA
| | - Cheryl L Olson
- Department of Pathology, Northwestern Medicine Malnati Brain Tumor Institute of the Robert H. Lurie Comprehensive Cancer Center, Northwestern University Feinberg School of Medicine, 303 E. Chicago Ave. Ward 3-140, Chicago, IL, USA
| | - Daniel J Brat
- Department of Pathology, Northwestern Medicine Malnati Brain Tumor Institute of the Robert H. Lurie Comprehensive Cancer Center, Northwestern University Feinberg School of Medicine, 303 E. Chicago Ave. Ward 3-140, Chicago, IL, USA.
| |
Collapse
|
26
|
Meng Y, Sun J, Zhang G, Yu T, Piao H. Clinical Prognostic Value of the PLOD Gene Family in Lung Adenocarcinoma. Front Mol Biosci 2022; 8:770729. [PMID: 35265665 PMCID: PMC8899219 DOI: 10.3389/fmolb.2021.770729] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2021] [Accepted: 12/30/2021] [Indexed: 01/09/2023] Open
Abstract
Accumulating evidence has implicated members of the procollagen-lysine, 2-oxoglutarate 5-dioxygenase (PLOD) gene family, PLOD1, PLOD2, and PLOD3, in cancer progression and metastasis. However, their expression, prognostic value, and mechanisms underlying their roles in lung adenocarcinoma (LUAD) have not yet been reported. We downloaded PLOD data for LUAD and normal tissues from The Cancer Genome Atlas (TCGA). PLOD1-3 protein expression was evaluated using the Clinical Proteomics Tumor Analysis Consortium and Human Protein Atlas. Survival analysis was performed using the Kaplan–Meier method. A protein–protein interaction network was constructed using STRING software. The “ClusterProfiler” package was used for functional-enrichment analysis. The relationship between PLOD mRNA expression and immune infiltration was analyzed using the Tumor Immunity Assessment Resource and Tumor Immune System Interaction Database. The expression of PLODs in LUAD tissues was significantly upregulated compared with that in adjacent normal tissues. PLOD mRNA overexpression is associated with lymph node metastasis and high TNM staging. Receiver operating characteristic curve analysis showed that when the cut-off level was 6.073, the accuracy, sensitivity, and specificity of PLOD1 in distinguishing LUAD from adjacent controls were 84.4, 79.7, and 82.6%, respectively. The accuracy, sensitivity, and specificity of PLOD2 in distinguishing LUAD from adjacent controls were 81.0, 98.3, and 68.0%, respectively, at a cut-off value of 4.360. The accuracy, sensitivity, and specificity of PLOD3 in distinguishing LUAD from adjacent controls were 69.0, 86.4, and 52.0%, respectively, with a cut-off value of 5.499. Kaplan–Meier survival analysis demonstrated that LUAD patients with high PLODs had a worse prognosis than those with low PLODs. Correlation analysis showed that PLOD mRNA expression was related to immune infiltration and tumor purity. Upregulation of PLOD expression was significantly associated with poor survival and immune cell infiltration in LUAD. Our research shows that PLOD family members have potential as novel biomarkers for poor prognosis and as potential immunotherapy targets for LUAD.
Collapse
Affiliation(s)
- Yiming Meng
- Department of Central Laboratory, Cancer Hospital of China Medical University, Liaoning Cancer Hospital and Institute, Shenyang, China
| | - Jing Sun
- Department of Biobank, Cancer Hospital of China Medical University, Liaoning Cancer Hospital and Institute, Shenyang, China
| | - Guirong Zhang
- Department of Central Laboratory, Cancer Hospital of China Medical University, Liaoning Cancer Hospital and Institute, Shenyang, China
| | - Tao Yu
- Department of Medical Imaging, Cancer Hospital of China Medical University, Liaoning Cancer Hospital and Institute, Shenyang, China
- *Correspondence: Tao Yu, ; Haozhe Piao,
| | - Haozhe Piao
- Department of Central Laboratory, Cancer Hospital of China Medical University, Liaoning Cancer Hospital and Institute, Shenyang, China
- Department of Neurosurgery, Cancer Hospital of China Medical University, Liaoning Cancer Hospital and Institute, Shenyang, China
- *Correspondence: Tao Yu, ; Haozhe Piao,
| |
Collapse
|
27
|
Zhang J, Dong Y, Shi Z, He H, Chen J, Zhang S, Wu W, Zhang Q, Han C, Hao L. P3H4 and PLOD1 expression associates with poor prognosis in bladder cancer. Clin Transl Oncol 2022; 24:1524-1532. [PMID: 35149972 DOI: 10.1007/s12094-022-02791-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2021] [Accepted: 01/21/2022] [Indexed: 12/24/2022]
Abstract
PURPOSE The prolyl 3-hydroxylase family member 4 gene (P3H4) is involved in the development of human cancers. The association of P3H4 with bladder cancer (BC) prognosis is unclear. This study aimed to analyze the association of P3H4 with BC prognosis. METHODS RNA-Seq data were downloaded from The Cancer Genome Atlas project and BC microarray datasets (GSE13507, GSE31684, and GSE32548) were downloaded from the Gene Expression Omnibus database. We analyzed the differences in P3H4 expression levels between BC tumors and non-tumor tissues and between samples with different clinical information. The association of P3H4 and P3H4-related genes with BC prognosis and the possibility of using P3H4 expression as a prognostic biomarker in BC patients were also analyzed. RevMan was used to perform the meta-analysis. RESULTS P3H4 was upregulated in BC tissues compared with the adjacent non-tumor tissues (p = 4.06e-08). Univariate Cox regression analysis and meta-analysis showed that high P3H4 expression level contributed to a poor BC prognosis (Hazard ratio, HR = 1.348, 95% CI 1.140-1.594, p = 4.89e-04; meta-analysis: HR = 1.45, 95% CI 1.10-1.91; p = 9.00e-03). Among the genes related to P3H4, the PLOD1 gene was closely associated with P3H4 expression (r = 0.620, p = 2.49e-44). Also, a meta-analysis showed that PLOD1 expression was associated with a poor prognosis in BC patients (HR = 1.77, 95% CI 1.31-2.38; p = 2.00e-04). CONCLUSIONS The P3H4 and PLOD1 genes might be used as reliable prognostic biomarkers for BC.
Collapse
Affiliation(s)
- Junjie Zhang
- Medical College of Soochow University, Suzhou, 215123, Jiangsu, China.,Department of Urology, Xuzhou Central Hospital, 199 Jiefang South Road, Xuzhou, 221009, Jiangsu, China
| | - Yang Dong
- Medical College of Soochow University, Suzhou, 215123, Jiangsu, China.,Department of Urology, Xuzhou Central Hospital, 199 Jiefang South Road, Xuzhou, 221009, Jiangsu, China
| | - Zhenduo Shi
- Department of Urology, Xuzhou Central Hospital, 199 Jiefang South Road, Xuzhou, 221009, Jiangsu, China
| | - Houguang He
- Department of Urology, Xuzhou Central Hospital, 199 Jiefang South Road, Xuzhou, 221009, Jiangsu, China
| | - Jiangang Chen
- Medical College of Soochow University, Suzhou, 215123, Jiangsu, China
| | - Shaoqi Zhang
- Medical College of Soochow University, Suzhou, 215123, Jiangsu, China
| | - Wei Wu
- Medical College of Soochow University, Suzhou, 215123, Jiangsu, China
| | - Qianjin Zhang
- Medical College of Soochow University, Suzhou, 215123, Jiangsu, China
| | - Conghui Han
- Medical College of Soochow University, Suzhou, 215123, Jiangsu, China.,Department of Urology, Xuzhou Central Hospital, 199 Jiefang South Road, Xuzhou, 221009, Jiangsu, China
| | - Lin Hao
- Medical College of Soochow University, Suzhou, 215123, Jiangsu, China. .,Department of Urology, Xuzhou Central Hospital, 199 Jiefang South Road, Xuzhou, 221009, Jiangsu, China.
| |
Collapse
|
28
|
Xu M, Fang S, Xie A. Posttranscriptional control of PLOD1 in adipose-derived stem cells regulates scar formation through altering macrophage polarization. ANNALS OF TRANSLATIONAL MEDICINE 2021; 9:1573. [PMID: 34790779 PMCID: PMC8576667 DOI: 10.21037/atm-21-4978] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Accepted: 10/16/2021] [Indexed: 11/06/2022]
Abstract
Background The level of cutaneous scar formation is a critical parameter to determine the success of skin wound healing. Adipose-derived mesenchymal stem cells (AMSCs) have been applied to improve treatment of cutaneous injury with the purpose of reducing scar formation. Methods The levels of procollagen-lysine 1,2-oxoglutarate 5-dioxygenase 1 (PLOD1) were assessed at scar sites. Then, PLOD1 in AMSCs was depleted by either expression of a PLOD1-specific short-hair interfering RNA (shPLOD1) or by expression of microRNA-449 (miR-449) that targets and suppresses protein translation of PLOD1 through 3 prime untranslated region (3'-UTR) interfering. For induction of skin injury, a blade cut of 1.5-cm long and 2-mm thick was made on the middle back of the mice. Transplantation of either AMSCs-shPLOD1 or AMSCs-miR-449 into the injured region of the mice was performed via tail vein injection. The fibrosis as well as underlying mechanisms were assessed. Results The AMSCs expressed high levels of PLOD1, a potent stimulator of fibrosis. We knocked down PLOD1 in AMSCs by expression of either shPLOD1 or miR-449. Transplantation of either AMSCs-shPLOD1 or AMSCs-miR-449 significantly reduced the fibrotic process in the injured region of the mice to a similar degree. Mechanistically, transplantation of either AMSCs-shPLOD1 or AMSCs-miR-449 shifted macrophage polarization from M2 to M1-like and reduced both reactive oxygen species (ROS) and activation of myofibroblasts from fibroblasts. Conclusions Suppression of PLOD1 levels in AMSCs either directly by shPLOD1 or indirectly by miR-449 may substantially improve the anti-fibrotic potential of AMSCs during wound healing, likely through altering macrophage polarization.
Collapse
Affiliation(s)
- Miao Xu
- Department of Plastic and Reconstructive Surgery, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Shuo Fang
- Department of Plastic and Reconstructive Surgery, Shanghai Changhai Hospital, Naval Medical University, Shanghai, China
| | - Aiguo Xie
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| |
Collapse
|
29
|
Jiang W, Zheng F, Yao T, Gong F, Zheng W, Yao N. IFI30 as a prognostic biomarker and correlation with immune infiltrates in glioma. ANNALS OF TRANSLATIONAL MEDICINE 2021; 9:1686. [PMID: 34988195 PMCID: PMC8667103 DOI: 10.21037/atm-21-5569] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/24/2021] [Accepted: 11/12/2021] [Indexed: 12/25/2022]
Abstract
Background Increased evidence indicates that the tumour microenvironment (TME) plays an essential role in the development, treatment and prognosis of glioma. High expression of interferon-gamma-inducible protein 30 (IFI30) is associated with the malignant phenotype, but the effect of IFI30 on the tumour immune microenvironment and its potential role in the carcinogenesis of glioma remain unknown. Methods The RNA sequencing (RNA-seq) data of 33 types of human cancer were obtained from The Cancer Genome Atlas (TCGA) Genomic Data Commons (GDC). R software was used to perform analyses, such as the expression of IFI30 in pan-cancer, evaluation of IFI30 as a prognostic biomarker in glioma, the relationship between IFI30 expression and clinical characteristics, and immune checkpoint. TIMER was used to analyse the correlation of IFI30 expression level with immune cell infiltration, and also to conduct survival analysis for immune cells and IFI30 in low grade glioma (LGG). DAVID was used for Gene Ontology (GO) functional annotations and Kyoto Encyclopedia of Genes and Genomes (KEGG) for pathway analysis of the genes similar to IFI30 in glioma. The differentially expressed genes (DEGs) between the high- and low-IFI30 expression groups were determined by DESeq2. Gene set enrichment analysis (GSEA) was then conducted to identify IFI30-related functional significance based on the hallmark gene set. Results Dysregulated expression of IFI30 was associated with human cancers. High IFI30 expression was associated with poor overall survival (OS), disease-specific survival (DSS) and progression-free interval (PFI). Univariate and multivariate analyses identified IFI30 as an independent predictor for glioma. Meanwhile, IFI30 overexpression significantly correlated with high-grade tumours, poor OS, and immune infiltration. In addition, IFI30-associated genes significantly enriched the hallmark tumour progression-related clusters and cancer pathways. Conclusions IFI30 is a prognostic biomarker correlated with immune infiltrates and acts as an oncogene in glioma.
Collapse
Affiliation(s)
- Wei Jiang
- Department of Neurology, the Second People's Hospital of Wuxi, Wuxi, China
| | - Feifei Zheng
- Department of Laboratory Medicine, Affiliated Hospital of Jiangnan University, Wuxi, China
| | - Taotao Yao
- Rehabilitation Center, the First Affiliated Hospital of Guangzhou University of Traditional Chinese Medicine, Guangzhou, China
| | - Fang Gong
- Department of Laboratory Medicine, Affiliated Hospital of Jiangnan University, Wuxi, China
| | - Wenjie Zheng
- Research Center of Clinical Medicine, Affiliated Hospital of Nantong University, Nantong, China
| | - Ninghua Yao
- Department of Oncology, Affiliated Hospital of Nantong University, Nantong, China
| |
Collapse
|
30
|
Shire K, Marcon E, Greenblatt J, Frappier L. Characterization of a cancer-associated Epstein-Barr virus EBNA1 variant reveals a novel interaction with PLOD1 and PLOD3. Virology 2021; 562:103-109. [PMID: 34304093 DOI: 10.1016/j.virol.2021.07.009] [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: 05/05/2021] [Revised: 07/18/2021] [Accepted: 07/19/2021] [Indexed: 10/20/2022]
Abstract
Whole genome sequence analysis of Epstein-Barr virus genomes from tumours and healthy individuals identified three amino acid changes in EBNA1 that are strongly associated with gastric carcinoma and nasopharyngeal carcinoma. Here we show that, while these mutations do not impact EBNA1 plasmid maintenance function, one of them (Thr85Ala) decreases transcriptional activation and results in a gain of function interaction with PLOD1 and PLOD3. PLOD family proteins are strongly linked to multiple cancers, and PLOD1 is recognized as a prognostic marker of gastric carcinoma. We identified the PLOD1 binding site in EBNA1as the N-terminal transactivation domain and show that lysine 83 is critical for this interaction. The results provide a novel link between EBV infection and the cancer-associated PLOD proteins.
Collapse
Affiliation(s)
- Kathy Shire
- Department of Molecular Genetics, University of Toronto, 661 University Ave, Suite 1600, Toronto, ON, M5G 1M1, Canada
| | - Edyta Marcon
- Donnelly Centre, University of Toronto, Toronto, M5S 3E1, Canada
| | - Jack Greenblatt
- Department of Molecular Genetics, University of Toronto, 661 University Ave, Suite 1600, Toronto, ON, M5G 1M1, Canada; Donnelly Centre, University of Toronto, Toronto, M5S 3E1, Canada
| | - Lori Frappier
- Department of Molecular Genetics, University of Toronto, 661 University Ave, Suite 1600, Toronto, ON, M5G 1M1, Canada.
| |
Collapse
|
31
|
FMR1/circCHAF1A/miR-211-5p/HOXC8 feedback loop regulates proliferation and tumorigenesis via MDM2-dependent p53 signaling in GSCs. Oncogene 2021; 40:4094-4110. [PMID: 34017077 DOI: 10.1038/s41388-021-01833-2] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2020] [Revised: 04/12/2021] [Accepted: 05/07/2021] [Indexed: 02/04/2023]
Abstract
Glioma is the most common and fatal primary malignant brain tumor. Glioma stem cells (GSCs) may be an important factor in glioma cell proliferation, invasion, chemoradiotherapy tolerance, and recurrence. Therefore, discovering novel GSCs related circular RNAs (circRNAs) may finds out a prospective target for the treatment of glioma. A novel circRNA-CHAF1A (circCHAF1A) was first found in our study. CircCHAF1A was overexpressed in glioma and related to the low survival rate. Functionally, it was found that no matter in vitro or in vivo, circCHAF1A can facilitate the proliferation and tumorigenesis of TP53wt GSCs. Mechanistically, circCHAF1A upregulated transcription factor HOXC8 expression in GSCs through miR-211-5p sponging. Then, HOXC8 can transcriptionally upregulate MDM2 expression and inhibited the antitumor effect of p53. Furtherly, the RNA binding protein FMR1 can bind to and promoted the expression of circCHAF1A via maintaining its stability, while HOXC8 also transcribed the FMR1 expression to form a feedback loop, which may be involved in the malignant transformation of glioma. The novel feedback loop among FMR1, circCHAF1A, miR-211-5p, and HOXC8 in GSCs can facilitate the proliferation and tumorigenesis of glioma and GSCs. It also provided a helpful biomarker for diagnosis and prognostic evaluation of glioma and may be applied to molecular targeted therapy.
Collapse
|
32
|
Chang XS, Zhu J, Yang T, Gao Y. MiR-524 suppressed the progression of oral squamous cell carcinoma by suppressing Metadherin and NF-κB signaling pathway in OSCC cell lines. Arch Oral Biol 2021; 125:105090. [PMID: 33676362 DOI: 10.1016/j.archoralbio.2021.105090] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2020] [Revised: 02/07/2021] [Accepted: 02/18/2021] [Indexed: 11/25/2022]
Abstract
OBJECTIVES The aim of the present study was to explore the functional role of miR-524 in oral squamous cell carcinoma (OSCC) and determine its underlying mechanism. MATERIALS AND METHODS Tumor tissues and adjacent tissues were obtained from 55 patients with OSCC (20 females and 35 males) with a mean age of 54 years (range from 24 to 72 years). Additionally, OSCC cell lines culture was used and Reverse transcription‑quantitative PCR (RT-qPCR) was applied to measure the expression of miR-524 in OSCC tissues and cells. The protein density of Metadherin (MTDH) in OSCC tissues was detected by Immunohistochemistry (IHC) assay. MiR-524 mimic was employed to investigate the impact of miR-524 on proliferation, migration, and invasion using 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyl tetrazolium bromide (MTT) assay and transwell assays. The dual luciferase reporter assay was utilized to investigate the interaction between MTDH and miR-524 expression. Cells transfected with miR-524 mimic and pcDNA-MTDH were subjected to western blot to investigate the role of NF-κB signaling in miR-524/MTDH axis mediated cell proliferation, migration, and invasion. RESULTS MiR-524 expression was decreased significantly in OSCC tissues compared to adjacent tissues, and closely related to clinical stage, tumor size, and lymph node metastasis. Over-expression of miR-524 suppressed the proliferation, migration, and invasion of OSCC cells. Luciferase reporter assay results demonstrated that MTDH was the target gene of miR-524. Over-expression of miR-524 reduced MTDH expression and inhibited NF-κB signaling pathway. Rescue experiments revealed that over-expression of MTDH partially reversed the efficacy of miR-524 mimic on OSCC cells. CONCLUSIONS These results indicated that miR-524 inhibits the activation of NF-κB signaling pathway via inhibiting MTDH, resulting in the suppression of cell proliferation, migration, and invasion.
Collapse
Affiliation(s)
- Xiang-Shuang Chang
- Department of Stomatology, The 964st Hospital, Changchun City, Jilin Province, China
| | - Jing Zhu
- Department of Nursing, The 964st Hospital, Changchun City, Jilin Province, China
| | - Tao Yang
- Department of Health Team, The 93313 Army, Changchun City, Jilin Province, China
| | - Ying Gao
- Department of Stomatology, The 964st Hospital, Changchun City, Jilin Province, China.
| |
Collapse
|