1
|
Rahaman KA, Mukim MSI, Hasan ML, Kim H, Pan CH, Kwon OS, Song DG, Han HS. Protein to biomaterials: Unraveling the antiviral and proangiogenic activities of Ac-Tβ 1-17 peptide, a thymosin β4 metabolite, and its implications in peptide-scaffold preparation. Bioact Mater 2025; 49:437-455. [PMID: 40177110 PMCID: PMC11964602 DOI: 10.1016/j.bioactmat.2025.02.008] [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: 07/28/2024] [Revised: 01/20/2025] [Accepted: 02/04/2025] [Indexed: 04/05/2025] Open
Abstract
Peptide metabolites are emerging biomolecules with numerous possibilities in biomaterial-based regenerative medicine due to their inherent bioactivities. These small, naturally occurring compounds are intermediates or byproducts of larger proteins and peptides, and they can have profound effects, such as antiviral therapeutics, proangiogenic agents, and regenerative medicinal applications. This study is among the first to focus on using thymosin β4 protein-derived metabolites to pioneer novel applications for peptide metabolites in biomaterials. This study found that the novel peptide metabolite acetyl-thymosin β4 (amino acid 1-17) (Ac-Tβ1-17) exhibited significant protease inhibition activity against SARS-CoV-2, surpassing its precursor protein. Additionally, Ac-Tβ1-17 demonstrated beneficial effects, such as cell proliferation, wound healing, and scavenging of reactive oxygen species (ROS) in human umbilical vein endothelial cells (HUVEC). Integrating Ac-Tβ1-17 into a peptide-based scaffold facilitated cell growth and angiogenesis inside the scaffold and through gradual release into the surrounding environment. The Ac-Tβ1-17 peptide treatment induced significant biochemical responses in HUVEC, increasing Akt, ERK, PI3K, MEK, and Bcl-2 gene expression and proangiogenic proteins. Ac-Tβ1-17 peptide treatment showed similar results in ex vivo by enhancing mouse fetal metatarsal growth and angiogenesis. These findings highlight the potential of natural protein metabolites to generate biologically active peptides, offering a novel strategy for enhancing biomaterial compatibility. This approach holds promise for developing therapeutic biomaterials using peptide metabolites, presenting exciting prospects for future research and applications.
Collapse
Affiliation(s)
- Khandoker Asiqur Rahaman
- Center for Biomaterials, Korea Institute of Science and Technology, Seoul, 02792, Republic of Korea
| | - Md Sofequl Islam Mukim
- Center for Natural Product Systems Biology, KIST Gangneung Institute of Natural Products, Gangneung, 25451, Republic of Korea
- Natural Product Applied Science, KIST School, University of Science and Technology (UST), Gangneung, 25451, Republic of Korea
| | - Md Lemon Hasan
- Center for Biomaterials, Korea Institute of Science and Technology, Seoul, 02792, Republic of Korea
- Division of Bio-Medical Science & Technology, University of Science and Technology (UST), 113 Gwahangno, Yuseong-gu, Daejeon, 34113, Republic of Korea
| | - Hyeok Kim
- Center for Biomaterials, Korea Institute of Science and Technology, Seoul, 02792, Republic of Korea
| | - Cheol-Ho Pan
- Center for Natural Product Systems Biology, KIST Gangneung Institute of Natural Products, Gangneung, 25451, Republic of Korea
- Natural Product Applied Science, KIST School, University of Science and Technology (UST), Gangneung, 25451, Republic of Korea
| | - Oh-Seung Kwon
- Doping Control Center, Korea Institute of Science and Technology, Seoul, 02792, Republic of Korea
- Division of Bio-Medical Science & Technology, University of Science and Technology (UST), 113 Gwahangno, Yuseong-gu, Daejeon, 34113, Republic of Korea
| | - Dae-Geun Song
- Center for Natural Product Systems Biology, KIST Gangneung Institute of Natural Products, Gangneung, 25451, Republic of Korea
- Natural Product Applied Science, KIST School, University of Science and Technology (UST), Gangneung, 25451, Republic of Korea
| | - Hyung-Seop Han
- Center for Biomaterials, Korea Institute of Science and Technology, Seoul, 02792, Republic of Korea
- Division of Bio-Medical Science & Technology, University of Science and Technology (UST), 113 Gwahangno, Yuseong-gu, Daejeon, 34113, Republic of Korea
- Research and Development Center, Elecell Corporation, Seoul, 02455, Republic of Korea
| |
Collapse
|
2
|
Wang X, Su Y, Lan B, Li X, Zhang B, Zhang L, Wang Y, Zhang C, Xuan C. USP22 promotes the proliferation and Sorafenib resistance of hepatocellular carcinoma cells via its deubiquitinase activity. Clin Transl Med 2025; 15:e70324. [PMID: 40341781 PMCID: PMC12059209 DOI: 10.1002/ctm2.70324] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2024] [Revised: 02/21/2025] [Accepted: 04/17/2025] [Indexed: 05/11/2025] Open
Abstract
BACKGROUND Hepatocellular carcinoma remains one of the most lethal cancers, characterized by poor prognosis and low life expectancy. Unfortunately, there are very few molecular therapeutic options available for it. Sorafenib is a current standard first-line treatment for advanced hepatocellular carcinoma, however, drug resistance significantly limits its therapeutic efficacy. METHODS Ubiquitin-specific protease 22 (USP22) expression level and its prognostic significance in hepatocellular carcinoma were analyzed using The Cancer Genome Atlas (TCGA) database. A series of cellular experiments related to cell proliferation and ferroptosis, and mouse tumor-bearing experiments were performed to investigate the role of USP22 in hepatocellular carcinoma cell growth and Sorafenib resistance. Flag affinity purification coupled with mass spectrometry, co-immunoprecipitation, and ubiquitination assays were conducted to identify direct substrates of USP22. Spike-in chromatin-immunoprecipitation (ChIP)-seq, RNA-seq, and ChIP assays were employed to explore the transcriptional substrates of USP22 as an H2BK120ub deubiquitinase. RESULTS Analysis of TCGA database reveals that USP22 is highly expressed in hepatocellular carcinoma tissues, which is closely associated with poor patient prognosis. Our data further indicates that USP22 promotes the proliferation of hepatocellular carcinoma cells via deubiquitinating and stabilizing cyclin-dependent kinase 11B (CDK11B). Additionally, USP22 acts as a novel inducer of Sorafenib resistance and suppresses Sorafenib-triggered ferroptosis in hepatocellular carcinoma cells. It reduces the transcription of transferrin receptor (TFRC) by decreasing H2BK120ub occupancy at TFRC transcription start site (TSS) downstream region, thereby inhibiting ferroptosis upon Sorafenib treatment. Finally, animal experiments confirm the role of USP22 in promoting hepatocellular carcinoma cell growth and Sorafenib resistance in vivo. Taken together, this study demonstrates that USP22 promotes hepatocellular carcinoma growth and inhibits Sorafenib-induced ferroptosis by deubiquitinating non-histone substrate CDK11B and histone H2B, respectively. CONCLUSIONS Our findings suggest USP22 as a promising prognostic biomarker and therapeutic target for hepatocellular carcinoma patients, particularly those with Sorafenib resistance. KEY POINTS USP22 promotes the proliferation of hepatocellular carcinoma cells by deubiquitinating and stabilizing cyclin-dependent kinase CDK11B. USP22 enhances Sorafenib resistance of hepatocellular carcinoma cells by inhibiting ferroptosis through the USP22/H2BK120ub/TFRC axis.
Collapse
Affiliation(s)
- Xiaochen Wang
- Key Laboratory of Breast Cancer Prevention and Therapy (Ministry of Education); The Province and Ministry Co‐sponsored Collaborative Innovation Center for Medical Epigenetics; Key Laboratory of Immune Microenvironment and Disease (Ministry of Education); Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Tianjin Medical UniversityTianjinChina
| | - Yijie Su
- Key Laboratory of Breast Cancer Prevention and Therapy (Ministry of Education); The Province and Ministry Co‐sponsored Collaborative Innovation Center for Medical Epigenetics; Key Laboratory of Immune Microenvironment and Disease (Ministry of Education); Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Tianjin Medical UniversityTianjinChina
| | - Bei Lan
- Key Laboratory of Breast Cancer Prevention and Therapy (Ministry of Education); The Province and Ministry Co‐sponsored Collaborative Innovation Center for Medical Epigenetics; Key Laboratory of Immune Microenvironment and Disease (Ministry of Education); Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Tianjin Medical UniversityTianjinChina
| | - Xuanyuan Li
- Key Laboratory of Breast Cancer Prevention and Therapy (Ministry of Education); The Province and Ministry Co‐sponsored Collaborative Innovation Center for Medical Epigenetics; Key Laboratory of Immune Microenvironment and Disease (Ministry of Education); Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Tianjin Medical UniversityTianjinChina
| | - Bodi Zhang
- Key Laboratory of Breast Cancer Prevention and Therapy (Ministry of Education); The Province and Ministry Co‐sponsored Collaborative Innovation Center for Medical Epigenetics; Key Laboratory of Immune Microenvironment and Disease (Ministry of Education); Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Tianjin Medical UniversityTianjinChina
| | - Liang Zhang
- Research Center of Translational MedicineJinan Central Hospital Affiliated to Shandong First Medical UniversityJinanChina
| | - Yingmei Wang
- Department of Gynecology and ObstetricsTianjin Medical University General HospitalTianjinChina
| | - Chunze Zhang
- Department of Colorectal SurgeryTianjin Union Medical CenterTianjinChina
| | - Chenghao Xuan
- Key Laboratory of Breast Cancer Prevention and Therapy (Ministry of Education); The Province and Ministry Co‐sponsored Collaborative Innovation Center for Medical Epigenetics; Key Laboratory of Immune Microenvironment and Disease (Ministry of Education); Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Tianjin Medical UniversityTianjinChina
| |
Collapse
|
3
|
Zhang X, Gao Y, Zhang S, Wang Y, Du Y, Hao S, Ni T. The Regulation of Cellular Senescence in Cancer. Biomolecules 2025; 15:448. [PMID: 40149983 PMCID: PMC11940315 DOI: 10.3390/biom15030448] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2025] [Revised: 03/10/2025] [Accepted: 03/18/2025] [Indexed: 03/29/2025] Open
Abstract
Cellular senescence is a stable state of cell cycle arrest caused by telomere shortening or various stresses. After senescence, cells cease dividing and exhibit many age-related characteristics. Unlike the halted proliferation of senescence cells, cancer cells are considered to have unlimited growth potential. When cells display senescence-related features, such as telomere loss or stem cell failure, they can inhibit tumor development. Therefore, inducing cells to enter a senescence state can serve as a barrier to tumor cell development. However, many recent studies have found that sustained senescence of tumor cells or normal cells under certain circumstances can exert environment-dependent effects of tumor promotion and inhibition by producing various cytokines. In this review, we first introduce the causes and characteristics of induced cellular senescence, analyze the senescence process of immune cells and cancer cells, and then discuss the dual regulatory role of cell senescence on tumor growth and senescence-induced therapies targeting cancer cells. Finally, we discuss the role of senescence in tumor progression and treatment opportunities, and propose further studies on cellular senescence and cancer therapy.
Collapse
Affiliation(s)
- Xianhong Zhang
- State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock, Institutes of Biomedical Sciences, School of Life Sciences, Inner Mongolia University, Hohhot 010070, China; (X.Z.); (Y.G.); (Y.W.); (Y.D.)
| | - Yue Gao
- State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock, Institutes of Biomedical Sciences, School of Life Sciences, Inner Mongolia University, Hohhot 010070, China; (X.Z.); (Y.G.); (Y.W.); (Y.D.)
| | - Siyu Zhang
- Key Lab of Ministry of Education for Protection and Utilization of Special Biological Resources in Western China, School of Life Sciences, Ningxia University, Yinchuan 750021, China;
| | - Yixiong Wang
- State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock, Institutes of Biomedical Sciences, School of Life Sciences, Inner Mongolia University, Hohhot 010070, China; (X.Z.); (Y.G.); (Y.W.); (Y.D.)
| | - Yitian Du
- State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock, Institutes of Biomedical Sciences, School of Life Sciences, Inner Mongolia University, Hohhot 010070, China; (X.Z.); (Y.G.); (Y.W.); (Y.D.)
| | - Shuailin Hao
- State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock, Institutes of Biomedical Sciences, School of Life Sciences, Inner Mongolia University, Hohhot 010070, China; (X.Z.); (Y.G.); (Y.W.); (Y.D.)
| | - Ting Ni
- State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock, Institutes of Biomedical Sciences, School of Life Sciences, Inner Mongolia University, Hohhot 010070, China; (X.Z.); (Y.G.); (Y.W.); (Y.D.)
| |
Collapse
|
4
|
Yi SA, Cho D, Kim S, Kim H, Choi MK, Choi HS, Shin S, Yun S, Lim A, Jeong JK, Yoon DE, Cha HJ, Kim K, Han J, Cho H, Cho J. Functional loss of ERBB receptor feedback inhibitor 1 (MIG6) promotes glioblastoma tumorigenesis by aberrant activation of epidermal growth factor receptor (EGFR). Mol Oncol 2025; 19:937-953. [PMID: 39129344 PMCID: PMC11887669 DOI: 10.1002/1878-0261.13717] [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: 12/05/2023] [Revised: 06/07/2024] [Accepted: 07/30/2024] [Indexed: 08/13/2024] Open
Abstract
Dysregulation of epidermal growth factor receptor (EGFR) is one of the most common mechanisms associated with the pathogenesis of various cancers. Mitogen-inducible gene 6 [MIG6; also known as ERBB receptor feedback inhibitor 1 (ERRFI1)], identified as a feedback inhibitor of EGFR, negatively regulates EGFR by directly inhibiting its kinase activity and facilitating its internalization, subsequently leading to degradation. Despite its proposed role as an EGFR-dependent tumor suppressor, the functional consequences and clinical relevance in cancer etiology remain incompletely understood. Here, we identify that the stoichiometric balance between MIG6 and EGFR is crucial in promoting EGFR-dependent oncogenic growth in various experimental model systems. In addition, a subset of ERRFI1 (the official gene symbol of MIG6) mutations exhibit impaired ability to suppress the enzymatic activation of EGFR at multiple levels. In summary, our data suggest that decreased or loss of MIG6 activity can lead to abnormal activation of EGFR, potentially contributing to cellular transformation. We propose that the mutation status of ERRFI1 and the expression levels of MIG6 can serve as additional biomarkers for guiding EGFR-targeted cancer therapies, including glioblastoma.
Collapse
Affiliation(s)
- Sang Ah Yi
- Epigenome Dynamics Control Research Center, School of PharmacySungkyunkwan UniversitySuwonKorea
- Present address:
Chemical Biology ProgramMemorial Sloan Kettering Cancer CenterNew YorkNYUSA
| | - Daseul Cho
- Department of Biomedical Science & EngineeringDankook UniversityCheonanKorea
- Department of Nanobiomedical ScienceDankook UniversityCheonanKorea
| | - Sujin Kim
- Department of Biomedical Science & EngineeringDankook UniversityCheonanKorea
- Department of Nanobiomedical ScienceDankook UniversityCheonanKorea
| | - Hyunjin Kim
- Department of Nanobiomedical ScienceDankook UniversityCheonanKorea
| | - Myung Kyung Choi
- Department of Systems Biology, College of Life Science and BiotechnologyYonsei UniversitySeoulKorea
| | - Hee Seong Choi
- Department of Systems Biology, College of Life Science and BiotechnologyYonsei UniversitySeoulKorea
| | - Sukjin Shin
- Department of Biomedical Science & EngineeringDankook UniversityCheonanKorea
- Department of Nanobiomedical ScienceDankook UniversityCheonanKorea
| | - Sujin Yun
- Department of Biomedical Science & EngineeringDankook UniversityCheonanKorea
- Department of Nanobiomedical ScienceDankook UniversityCheonanKorea
| | - Ahjin Lim
- Department of Biomedical Science & EngineeringDankook UniversityCheonanKorea
- Department of Nanobiomedical ScienceDankook UniversityCheonanKorea
| | - Jae Kyun Jeong
- Department of Nanobiomedical ScienceDankook UniversityCheonanKorea
| | - Da Eun Yoon
- Department of Biomedical SciencesKorea University College of MedicineSeoulKorea
- Department of PhysiologyKorea University College of MedicineSeoulKorea
| | - Hye Ji Cha
- Department of Biomedical Science & EngineeringDankook UniversityCheonanKorea
| | - Kyoungmi Kim
- Department of Biomedical SciencesKorea University College of MedicineSeoulKorea
- Department of PhysiologyKorea University College of MedicineSeoulKorea
| | - Jeung‐Whan Han
- Epigenome Dynamics Control Research Center, School of PharmacySungkyunkwan UniversitySuwonKorea
| | - Hyun‐Soo Cho
- Department of Systems Biology, College of Life Science and BiotechnologyYonsei UniversitySeoulKorea
| | - Jeonghee Cho
- Department of Biomedical Science & EngineeringDankook UniversityCheonanKorea
- Department of Nanobiomedical ScienceDankook UniversityCheonanKorea
| |
Collapse
|
5
|
Pei D, Zhang D, Guo Y, Chang H, Cui H. Long Non-Coding RNAs in Malignant Human Brain Tumors: Driving Forces Behind Progression and Therapy. Int J Mol Sci 2025; 26:694. [PMID: 39859408 PMCID: PMC11766336 DOI: 10.3390/ijms26020694] [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: 12/08/2024] [Revised: 01/12/2025] [Accepted: 01/13/2025] [Indexed: 01/27/2025] Open
Abstract
Long non-coding RNAs (lncRNAs) play a pivotal role in regulating gene expression and are critically involved in the progression of malignant brain tumors, including glioblastoma, medulloblastoma, and meningioma. These lncRNAs interact with microRNAs (miRNAs), proteins, and DNA, influencing key processes such as cell proliferation, migration, and invasion. This review highlights the multifaceted impact of lncRNA dysregulation on tumor progression and underscores their potential as therapeutic targets to enhance the efficacy of chemotherapy, radiotherapy, and immunotherapy. The insights provided offer new directions for advancing basic research and clinical applications in malignant brain tumors.
Collapse
Affiliation(s)
| | | | | | | | - Hongjuan Cui
- State Key Laboratory of Resource Insects, Medical Research Institute, Southwest University, Chongqing 400715, China; (D.P.); (D.Z.); (Y.G.); (H.C.)
| |
Collapse
|
6
|
Zhang M, Hu T, Ma T, Huang W, Wang Y. Epigenetics and environmental health. Front Med 2024; 18:571-596. [PMID: 38806988 DOI: 10.1007/s11684-023-1038-2] [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: 07/03/2023] [Accepted: 10/15/2023] [Indexed: 05/30/2024]
Abstract
Epigenetic modifications including DNA methylation, histone modifications, chromatin remodeling, and RNA modifications complicate gene regulation and heredity and profoundly impact various physiological and pathological processes. In recent years, accumulating evidence indicates that epigenetics is vulnerable to environmental changes and regulates the growth, development, and diseases of individuals by affecting chromatin activity and regulating gene expression. Environmental exposure or induced epigenetic changes can regulate the state of development and lead to developmental disorders, aging, cardiovascular disease, Alzheimer's disease, cancers, and so on. However, epigenetic modifications are reversible. The use of specific epigenetic inhibitors targeting epigenetic changes in response to environmental exposure is useful in disease therapy. Here, we provide an overview of the role of epigenetics in various diseases. Furthermore, we summarize the mechanism of epigenetic alterations induced by different environmental exposures, the influence of different environmental exposures, and the crosstalk between environmental variation epigenetics, and genes that are implicated in the body's health. However, the interaction of multiple factors and epigenetics in regulating the initiation and progression of various diseases complicates clinical treatments. We discuss some commonly used epigenetic drugs targeting epigenetic modifications and methods to prevent or relieve various diseases regulated by environmental exposure and epigenetics through diet.
Collapse
Affiliation(s)
- Min Zhang
- Key Laboratory of Cancer and Microbiome, State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Ting Hu
- Key Laboratory of Cancer and Microbiome, State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Tianyu Ma
- Key Laboratory of Cancer and Microbiome, State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
- Beijing Key Laboratory of Cancer Invasion and Metastasis Research, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Capital Medical University, Beijing, 100069, China
| | - Wei Huang
- Beijing Key Laboratory of Cancer Invasion and Metastasis Research, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Capital Medical University, Beijing, 100069, China.
| | - Yan Wang
- Key Laboratory of Cancer and Microbiome, State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China.
- Beijing Key Laboratory of Cancer Invasion and Metastasis Research, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Capital Medical University, Beijing, 100069, China.
| |
Collapse
|
7
|
Kong Y, Jiang R, Zhou H, Ge M, Lin H, Wang Y, Yao R, Wang Q, Liang X, Li J, Zhou X. PHF12 regulates HDAC1 to promote tumorigenesis via EGFR/AKT signaling pathway in non-small cell lung cancer. J Transl Med 2024; 22:689. [PMID: 39075515 PMCID: PMC11287983 DOI: 10.1186/s12967-024-05488-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2023] [Accepted: 07/03/2024] [Indexed: 07/31/2024] Open
Abstract
BACKGROUND Lung cancer stands as the second most prevalent malignant neoplasm worldwide. Addressing the underlying mechanisms propelling the progression of non-small cell lung cancer is of paramount importance. In this study, we have elucidated the pivotal role of PHF12 in this context. MATERIALS AND METHODS We harnessed clinical lung cancer tissue samples and non-small cell lung cancer cell lines to discern the expression pattern of PHF12. In vitro assays probing cell proliferation were conducted to substantiate the functional impact of PHF12. Furthermore, an in vivo Xenograft model was employed to dissect the role of PHF12. Employing ChIP assays and qRT-PCR, we delved into the intricate binding dynamics between PHF12 and HDAC1. Mechanistic insights into the PHF12-HDAC1 axis in lung cancer progression were pursued via RNA-seq and GSEA analyses. RESULTS Notably, PHF12 exhibited a substantial upregulation within tumor tissue, concomitant with its correlation to HDAC1. The trilogy of cell proliferation assays, transwell assays, and the Xenograft model collectively underscored the promoting influence of PHF12 on lung cancer proliferation, both in vitro and in vivo. The ChIP assay unveiled the transcriptional regulatory role of PHF12 in governing HDAC1 expression. This correlation extended to both mRNA and protein levels. PHF12 promotes NSCLC progression through regulating HDCA1 expression. Intriguingly, the rescue of function within NSCLC cell lines post PHF12 knockdown was achievable through HDAC1 overexpression. Additionally, our findings unveiled the capacity of the PHF12-HDAC1 axis to activate the EGFR/AKT signaling pathway, thereby further corroborating its significance in lung cancer progression. CONCLUSION Our study identified PHF12 as an oncogenic role in lung cancer proliferation and migration for the first time. PHF12 transcriptionally regulate HDAC1 and activate EGFR/AKT signaling pathway in NSCLC progression. PHF12 may serve as an important target in lung cancer therapy.
Collapse
Affiliation(s)
- Yiru Kong
- Department of Oncology, Huashan Hospital Fudan University, 12 Middle Urumqi Road, Shanghai, 200000, China
- Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Rongrong Jiang
- Department of Cardiothoracic Surgery, Huashan Hospital Fudan University, 12 Middle Urumqi Road, Shanghai, 200000, China
| | - Hui Zhou
- Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Mengxi Ge
- Department of Oncology, Huashan Hospital Fudan University, 12 Middle Urumqi Road, Shanghai, 200000, China
| | - Hao Lin
- Department of Oncology, Huashan Hospital Fudan University, 12 Middle Urumqi Road, Shanghai, 200000, China
| | - Yu Wang
- Department of Oncology, Huashan Hospital Fudan University, 12 Middle Urumqi Road, Shanghai, 200000, China
| | - Rongrong Yao
- Department of Oncology, Huashan Hospital Fudan University, 12 Middle Urumqi Road, Shanghai, 200000, China
| | - Qing Wang
- Department of Oncology, Huashan Hospital Fudan University, 12 Middle Urumqi Road, Shanghai, 200000, China
| | - Xiaohua Liang
- Department of Oncology, Huashan Hospital Fudan University, 12 Middle Urumqi Road, Shanghai, 200000, China
| | - Jing Li
- Department of Oncology, Huashan Hospital Fudan University, 12 Middle Urumqi Road, Shanghai, 200000, China.
| | - Xinli Zhou
- Department of Oncology, Huashan Hospital Fudan University, 12 Middle Urumqi Road, Shanghai, 200000, China.
| |
Collapse
|
8
|
Xiao F, Zhu H, Xiong Y, Guo Y, Zhang Z, Zeng J, Xiao Y, Liao B, Shang X, Zhao S, Hu G, Huang K, Guo H. Positive feedback loop of c-myc/XTP6/NDH2/NF-κB to promote malignant progression in glioblastoma. J Exp Clin Cancer Res 2024; 43:187. [PMID: 38965580 PMCID: PMC11225266 DOI: 10.1186/s13046-024-03109-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2024] [Accepted: 06/24/2024] [Indexed: 07/06/2024] Open
Abstract
BACKGROUND Recent studies have highlighted the significant role of the NF-κB signaling pathway in the initiation and progression of cancer. Furthermore, long noncoding RNAs (lncRNAs) have been identified as pivotal regulators in sustaining the NF-κB signaling pathway's functionality. Despite these findings, the underlying molecular mechanisms through which lncRNAs influence the NF-κB pathway remain largely unexplored. METHODS Bioinformatic analyses were utilized to investigate the differential expression and prognostic significance of XTP6. The functional roles of XTP6 were further elucidated through both in vitro and in vivo experimental approaches. To estimate the interaction between XTP6 and NDH2, RNA pulldown and RNA Immunoprecipitation (RIP) assays were conducted. The connection between XTP6 and the IκBα promoter was examined using Chromatin Isolation by RNA Purification (ChIRP) assays. Additionally, Chromatin Immunoprecipitation (ChIP) assays were implemented to analyze the binding affinity of c-myc to the XTP6 promoter, providing insights into the regulatory mechanisms at play. RESULTS XTP6 was remarkedly upregulated in glioblastoma multiforme (GBM) tissues and was connected with adverse prognosis in GBM patients. Our investigations revealed that XTP6 can facilitate the malignant progression of GBM both in vitro and in vivo. Additionally, XTP6 downregulated IκBα expression by recruiting NDH2 to the IκBα promoter, which resulted in elevated levels of H3K27me3, thereby reducing the transcriptional activity of IκBα. Moreover, the progression of GBM was further driven by the c-myc-mediated upregulation of XTP6, establishing a positive feedback loop with IκBα that perpetuated the activation of the NF-κB signaling pathway. Notably, the application of an inhibitor targeting the NF-κB signaling pathway effectively inhibited the continuous activation induced by XTP6, leading to a significant reduction in tumor formation in vivo. CONCLUSION The results reveal that XTP6 unveils an innovative epigenetic mechanism instrumental in the sustained activation of the NF-κB signaling pathway, suggesting a promising therapeutic target for the treatment of GBM.
Collapse
Affiliation(s)
- Feng Xiao
- Department of Neurosurgery, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, 330006, China
- Jiangxi Key Laboratory of Neurological Tumors and Cerebrovascular Diseases, Nanchang University, Nanchang, Jiangxi, 330006, China
- JXHC key Laboratory of Neurological medicine, Nanchang University, Nanchang, Jiangxi, 330006, China
- Institute of Neuroscience, Nanchang University, Nanchang, Jiangxi, 330006, China
- Jiangxi Province Key Laboratory of Neurological Diseases, Nanchang University, Nanchang, Jiangxi, 330006, China
| | - Hong Zhu
- Department of Neurosurgery, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, 330006, China
- Jiangxi Key Laboratory of Neurological Tumors and Cerebrovascular Diseases, Nanchang University, Nanchang, Jiangxi, 330006, China
- JXHC key Laboratory of Neurological medicine, Nanchang University, Nanchang, Jiangxi, 330006, China
- Institute of Neuroscience, Nanchang University, Nanchang, Jiangxi, 330006, China
- Jiangxi Province Key Laboratory of Neurological Diseases, Nanchang University, Nanchang, Jiangxi, 330006, China
| | - Yaping Xiong
- Departments of Anesthesiology, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, 330006, China
| | - Yun Guo
- Department of Neurosurgery, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, 330006, China
- Jiangxi Key Laboratory of Neurological Tumors and Cerebrovascular Diseases, Nanchang University, Nanchang, Jiangxi, 330006, China
- JXHC key Laboratory of Neurological medicine, Nanchang University, Nanchang, Jiangxi, 330006, China
- Institute of Neuroscience, Nanchang University, Nanchang, Jiangxi, 330006, China
- Jiangxi Province Key Laboratory of Neurological Diseases, Nanchang University, Nanchang, Jiangxi, 330006, China
| | - Zhe Zhang
- Department of Neurosurgery, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, 330006, China
- Jiangxi Key Laboratory of Neurological Tumors and Cerebrovascular Diseases, Nanchang University, Nanchang, Jiangxi, 330006, China
- JXHC key Laboratory of Neurological medicine, Nanchang University, Nanchang, Jiangxi, 330006, China
- Institute of Neuroscience, Nanchang University, Nanchang, Jiangxi, 330006, China
- Jiangxi Province Key Laboratory of Neurological Diseases, Nanchang University, Nanchang, Jiangxi, 330006, China
| | - Jie Zeng
- Department of Neurosurgery, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, 330006, China
- Jiangxi Key Laboratory of Neurological Tumors and Cerebrovascular Diseases, Nanchang University, Nanchang, Jiangxi, 330006, China
- JXHC key Laboratory of Neurological medicine, Nanchang University, Nanchang, Jiangxi, 330006, China
- Institute of Neuroscience, Nanchang University, Nanchang, Jiangxi, 330006, China
- Jiangxi Province Key Laboratory of Neurological Diseases, Nanchang University, Nanchang, Jiangxi, 330006, China
| | - Yao Xiao
- Department of Neurosurgery, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, 330006, China
- Jiangxi Key Laboratory of Neurological Tumors and Cerebrovascular Diseases, Nanchang University, Nanchang, Jiangxi, 330006, China
- JXHC key Laboratory of Neurological medicine, Nanchang University, Nanchang, Jiangxi, 330006, China
- Institute of Neuroscience, Nanchang University, Nanchang, Jiangxi, 330006, China
- Jiangxi Province Key Laboratory of Neurological Diseases, Nanchang University, Nanchang, Jiangxi, 330006, China
| | - Bin Liao
- Department of Neurosurgery, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, 330006, China
- Jiangxi Key Laboratory of Neurological Tumors and Cerebrovascular Diseases, Nanchang University, Nanchang, Jiangxi, 330006, China
- JXHC key Laboratory of Neurological medicine, Nanchang University, Nanchang, Jiangxi, 330006, China
- Institute of Neuroscience, Nanchang University, Nanchang, Jiangxi, 330006, China
- Jiangxi Province Key Laboratory of Neurological Diseases, Nanchang University, Nanchang, Jiangxi, 330006, China
| | - Xuesong Shang
- Department of Neurosurgery, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, 330006, China
- Jiangxi Key Laboratory of Neurological Tumors and Cerebrovascular Diseases, Nanchang University, Nanchang, Jiangxi, 330006, China
- JXHC key Laboratory of Neurological medicine, Nanchang University, Nanchang, Jiangxi, 330006, China
- Institute of Neuroscience, Nanchang University, Nanchang, Jiangxi, 330006, China
- Jiangxi Province Key Laboratory of Neurological Diseases, Nanchang University, Nanchang, Jiangxi, 330006, China
| | - Siyi Zhao
- Department of Neurosurgery, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, 330006, China
- Jiangxi Key Laboratory of Neurological Tumors and Cerebrovascular Diseases, Nanchang University, Nanchang, Jiangxi, 330006, China
- JXHC key Laboratory of Neurological medicine, Nanchang University, Nanchang, Jiangxi, 330006, China
- Institute of Neuroscience, Nanchang University, Nanchang, Jiangxi, 330006, China
- Jiangxi Province Key Laboratory of Neurological Diseases, Nanchang University, Nanchang, Jiangxi, 330006, China
| | - Guowen Hu
- Department of Neurosurgery, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, 330006, China
| | - Kai Huang
- Department of Neurosurgery, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, 330006, China
- Jiangxi Key Laboratory of Neurological Tumors and Cerebrovascular Diseases, Nanchang University, Nanchang, Jiangxi, 330006, China
- JXHC key Laboratory of Neurological medicine, Nanchang University, Nanchang, Jiangxi, 330006, China
- Institute of Neuroscience, Nanchang University, Nanchang, Jiangxi, 330006, China
- Jiangxi Province Key Laboratory of Neurological Diseases, Nanchang University, Nanchang, Jiangxi, 330006, China
| | - Hua Guo
- Department of Neurosurgery, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, 330006, China.
- Jiangxi Key Laboratory of Neurological Tumors and Cerebrovascular Diseases, Nanchang University, Nanchang, Jiangxi, 330006, China.
- JXHC key Laboratory of Neurological medicine, Nanchang University, Nanchang, Jiangxi, 330006, China.
- Institute of Neuroscience, Nanchang University, Nanchang, Jiangxi, 330006, China.
- Jiangxi Province Key Laboratory of Neurological Diseases, Nanchang University, Nanchang, Jiangxi, 330006, China.
| |
Collapse
|
9
|
Zhao J, Hua J, Zhan Y, Chen C, Liu Y, Yang L, Wang H, Wang H, Li J. O-GlcNAcylation stimulates the deubiquitination activity of USP16 and regulates cell cycle progression. J Biol Chem 2024; 300:107150. [PMID: 38462164 PMCID: PMC10998217 DOI: 10.1016/j.jbc.2024.107150] [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: 10/16/2023] [Revised: 02/14/2024] [Accepted: 02/26/2024] [Indexed: 03/12/2024] Open
Abstract
Histone 2A monoubiquitination (uH2A) underscores a key epigenetic regulation of gene expression. In this report, we show that the deubiquitinase for uH2A, ubiquitin-specific peptidase 16 (USP16), is modified by O-linked N-acetylglucosamine (O-GlcNAc). O-GlcNAcylation involves the installation of the O-GlcNAc moiety to Ser/Thr residues. It crosstalks with Ser/Thr phosphorylation, affects protein-protein interaction, alters enzyme activity or protein folding, and changes protein subcellular localization. In our study, we first confirmed that USP16 is glycosylated on Thr203 and Ser214, as reported in a previous chemoenzymatic screen. We then discovered that mutation of the O-GlcNAcylation site Thr203, which is adjacent to deubiquitination-required Cys204, reduces the deubiquitination activity toward H2AK119ub in vitro and in cells, while mutation on Ser214 had the opposite effects. Using USP16 Ser552 phosphorylation-specific antibodies, we demonstrated that O-GlcNAcylation antagonizes cyclin-dependent kinase 1-mediated phosphorylation and promotes USP16 nuclear export. O-GlcNAcylation of USP16 is also required for deubiquitination of Polo-like kinase 1, a mitotic master kinase, and the subsequent chromosome segregation and cytokinesis. In summary, our study revealed that O-GlcNAcylation of USP16 at Thr203 and Ser214 coordinates deubiquitination of uH2A and Polo-like kinase 1, thus ensuring proper cell cycle progression.
Collapse
Affiliation(s)
- Jianxin Zhao
- Beijing Key Laboratory of DNA Damage Response and College of Life Science, Capital Normal University, Beijing, China
| | - Jie Hua
- Department of Biochemistry and Molecular Genetics, University of Alabama at Birmingham, Birmingham, Alabama, USA; School of Life Sciences, Fudan University, Shanghai, China
| | - Yahui Zhan
- Beijing Key Laboratory of DNA Damage Response and College of Life Science, Capital Normal University, Beijing, China
| | - Chunxu Chen
- Department of Biochemistry and Molecular Genetics, University of Alabama at Birmingham, Birmingham, Alabama, USA; Massey Cancer Center, Virginia Commonwealth University, Richmond, Virginia, USA; Department of Bioengineering, Virginia Commonwealth University, Richmond, Virginia, USA
| | - Yue Liu
- Beijing Key Laboratory of DNA Damage Response and College of Life Science, Capital Normal University, Beijing, China
| | - Liqian Yang
- Beijing Key Laboratory of Protein Posttranslational Modifications and Cell Function, Department of Biochemistry and Biophysics, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China
| | - Haiying Wang
- Beijing Key Laboratory of Protein Posttranslational Modifications and Cell Function, Department of Biochemistry and Biophysics, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China.
| | - Hengbin Wang
- Department of Biochemistry and Molecular Genetics, University of Alabama at Birmingham, Birmingham, Alabama, USA; Massey Cancer Center, Virginia Commonwealth University, Richmond, Virginia, USA; Division of Hematology, Oncology, and Palliative Care, Department of Internal Medicine, Virginia Commonwealth University, Richmond, Virginia, USA.
| | - Jing Li
- Beijing Key Laboratory of DNA Damage Response and College of Life Science, Capital Normal University, Beijing, China.
| |
Collapse
|
10
|
He Y, Huang X, Ma Y, Yang G, Cui Y, Lv X, Zhao R, Jin H, Tong Y, Zhang X, Li J, Peng M. A novel aging-associated lncRNA signature for predicting prognosis in osteosarcoma. Sci Rep 2024; 14:1386. [PMID: 38228673 PMCID: PMC10791644 DOI: 10.1038/s41598-024-51732-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2023] [Accepted: 01/09/2024] [Indexed: 01/18/2024] Open
Abstract
Osteosarcoma (OS) is one of the most prevalent bone tumors in adolescents, and the correlation between aging and OS remains unclear. Currently, few accurate and reliable biomarkers have been determined for OS prognosis. To address this issue, we carried out a detailed bioinformatics analysis based on OS with data from the Cancer Genome Atlas data portal and Human Aging Genomic Resources database, as well as in vitro experiments. A total of 88 OS samples with gene expression profiles and corresponding clinical characteristics were obtained. Through univariate Cox regression analysis and survival analysis, 10 aging-associated survival lncRNAs (AASRs) were identified to be associated with the overall survival of OS patients. Based on the expression levels of the 10 AASRs, the OS patients were classified into two clusters (Cluster A and Cluster B). Cluster A had a worse prognosis, while Cluster B had a better prognosis. Then, 5 AASRs were ultimately included in the signature through least absolute shrinkage and selection operator-Cox regression analysis. Kaplan‒Meier survival analysis verified that the high-risk group exhibited a worse prognosis than the low-risk group. Furthermore, univariate and multivariate Cox regression analyses confirmed that the riskScore was an independent prognostic factor for OS patients. Subsequently, we discovered that the risk signature was correlated with the properties of the tumor microenvironment and immune cell infiltration. Specifically, there was a positive association between the risk model and naïve B cells, resting dendritic cells and gamma delta T cells, while it was negatively related to CD8+ T cells. Finally, in vitro experiments, we found that UNC5B-AS1 inhibited OS cells from undergoing cellular senescence and apoptosis, thereby promoting OS cells proliferation. In conclusion, we constructed and verified a 5 AASR-based signature, that exhibited excellent performance in evaluating the overall survival of OS patients. In addition, we found that UNC5B-AS1 might inhibit the senescence process, thus leading to the development and progression of OS. Our findings may provide novel insights into the treatment of OS patients.
Collapse
Affiliation(s)
- Yi He
- Department of Mini-Invasive Spinal Surgery, The Third People's Hospital of Henan Province, Zhengzhou, 450006, Henan, China
| | - Xiao Huang
- Department of Clinical Laboratory, Luohe Central Hospital, Luohe, 462300, Henan, China
| | - Yajie Ma
- Department of Medical Affair, The Third People's Hospital of Henan Province, Zhengzhou, 450006, Henan, China
| | - Guohui Yang
- Department of Emergency Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, China
| | - Yuqing Cui
- General ICU, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, China
| | - Xuefeng Lv
- Department of Clinical Laboratory, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, China
| | - Rongling Zhao
- Department of Clinical Laboratory, The Third People's Hospital of Henan Province, Zhengzhou, 450006, Henan, China
| | - Huifang Jin
- Department of Blood Transfusion, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, China
| | - Yalin Tong
- Department of Digestion, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, China
| | - Xinyu Zhang
- Department of Medical Affair, The Third People's Hospital of Henan Province, Zhengzhou, 450006, Henan, China
| | - Jitian Li
- Henan Luoyang Orthopedic Hospital (Henan Provincial Orthopedic Hospital), Henan Provincial Orthopedic Institute, Henan University of Chinese Medicine, 100 Yongping Road, Zhengzhou, 450000, Henan, China.
| | - Mengle Peng
- Department of Clinical Laboratory, The Third People's Hospital of Henan Province, Zhengzhou, 450006, Henan, China.
| |
Collapse
|
11
|
Zeng K, Zeng Y, Zhan H, Zhan Z, Wang L, Xie Y, Tang Y, Li C, Chen Y, Li S, Liu M, Chen X, Liang L, Deng F, Song Y, Zhou A. SEC61G assists EGFR-amplified glioblastoma to evade immune elimination. Proc Natl Acad Sci U S A 2023; 120:e2303400120. [PMID: 37523556 PMCID: PMC10410745 DOI: 10.1073/pnas.2303400120] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2023] [Accepted: 07/06/2023] [Indexed: 08/02/2023] Open
Abstract
Amplification of chromosome 7p11 (7p11) is the most common alteration in primary glioblastoma (GBM), resulting in gains of epidermal growth factor receptor (EGFR) copy number in 50 to 60% of GBM tumors. However, treatment strategies targeting EGFR have thus far failed in clinical trials, and the underlying mechanism remains largely unclear. We here demonstrate that EGFR amplification at the 7p11 locus frequently encompasses its neighboring genes and identifies SEC61G as a critical regulator facilitating GBM immune evasion and tumor growth. We found that SEC61G is always coamplified with EGFR and is highly expressed in GBM. As an essential subunit of the SEC61 translocon complex, SEC61G promotes translocation of newly translated immune checkpoint ligands (ICLs, including PD-L1, PVR, and PD-L2) into the endoplasmic reticulum and promotes their glycosylation, stabilization, and membrane presentation. Depletion of SEC61G promotes the infiltration and cytolytic activity of CD8+ T cells and thus inhibits GBM occurrence. Further, SEC61G inhibition augments the therapeutic efficiency of EGFR tyrosine kinase inhibitors in mice. Our study demonstrates a critical role of SEC61G in GBM immune evasion, which provides a compelling rationale for combination therapy of EGFR-amplified GBMs.
Collapse
Affiliation(s)
- Kunlin Zeng
- Department of Cell Biology, School of Basic Medical Science, Southern Medical University, Guangzhou510515, China
| | - Yu Zeng
- Department of Cell Biology, School of Basic Medical Science, Southern Medical University, Guangzhou510515, China
| | - Hongchao Zhan
- Department of Cell Biology, School of Basic Medical Science, Southern Medical University, Guangzhou510515, China
| | - Ziling Zhan
- Department of Cell Biology, School of Basic Medical Science, Southern Medical University, Guangzhou510515, China
| | - Li Wang
- Department of Cell Biology, School of Basic Medical Science, Southern Medical University, Guangzhou510515, China
| | - Yuxin Xie
- Department of Cell Biology, School of Basic Medical Science, Southern Medical University, Guangzhou510515, China
| | - Yanqing Tang
- Department of Cell Biology, School of Basic Medical Science, Southern Medical University, Guangzhou510515, China
| | - Cuiying Li
- Department of Cell Biology, School of Basic Medical Science, Southern Medical University, Guangzhou510515, China
| | - Yanwen Chen
- Department of Cell Biology, School of Basic Medical Science, Southern Medical University, Guangzhou510515, China
| | - Shangbiao Li
- Department of Cell Biology, School of Basic Medical Science, Southern Medical University, Guangzhou510515, China
- Department of Radiation Oncology, Zhujiang Hospital, Southern Medical University, Guangzhou510285, China
| | - Ming Liu
- Department of Cell Biology, School of Basic Medical Science, Southern Medical University, Guangzhou510515, China
| | - Xiaoxia Chen
- Department of Cell Biology, School of Basic Medical Science, Southern Medical University, Guangzhou510515, China
| | - Li Liang
- Guangdong Province Key Laboratory of Molecular Tumor Pathology, School of Basic Medical Science, Southern Medical University, Guangzhou510515, China
| | - Fan Deng
- Department of Cell Biology, School of Basic Medical Science, Southern Medical University, Guangzhou510515, China
| | - Ye Song
- Department of Neurosurgery, Nanfang Hospital, Southern Medical University, Guangzhou510515, China
| | - Aidong Zhou
- Department of Cell Biology, School of Basic Medical Science, Southern Medical University, Guangzhou510515, China
- Department of Radiation Oncology, Zhujiang Hospital, Southern Medical University, Guangzhou510285, China
- Guangdong Province Key Laboratory of Molecular Tumor Pathology, School of Basic Medical Science, Southern Medical University, Guangzhou510515, China
| |
Collapse
|
12
|
Pathania AS. Crosstalk between Noncoding RNAs and the Epigenetics Machinery in Pediatric Tumors and Their Microenvironment. Cancers (Basel) 2023; 15:2833. [PMID: 37345170 DOI: 10.3390/cancers15102833] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2023] [Revised: 05/08/2023] [Accepted: 05/10/2023] [Indexed: 06/23/2023] Open
Abstract
According to the World Health Organization, every year, an estimated 400,000+ new cancer cases affect children under the age of 20 worldwide. Unlike adult cancers, pediatric cancers develop very early in life due to alterations in signaling pathways that regulate embryonic development, and environmental factors do not contribute much to cancer development. The highly organized complex microenvironment controlled by synchronized gene expression patterns plays an essential role in the embryonic stages of development. Dysregulated development can lead to tumor initiation and growth. The low mutational burden in pediatric tumors suggests the predominant role of epigenetic changes in driving the cancer phenotype. However, one more upstream layer of regulation driven by ncRNAs regulates gene expression and signaling pathways involved in the development. Deregulation of ncRNAs can alter the epigenetic machinery of a cell, affecting the transcription and translation profiles of gene regulatory networks required for cellular proliferation and differentiation during embryonic development. Therefore, it is essential to understand the role of ncRNAs in pediatric tumor development to accelerate translational research to discover new treatments for childhood cancers. This review focuses on the role of ncRNA in regulating the epigenetics of pediatric tumors and their tumor microenvironment, the impact of their deregulation on driving pediatric tumor progress, and their potential as effective therapeutic targets.
Collapse
Affiliation(s)
- Anup S Pathania
- Department of Biochemistry and Molecular Biology & The Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE 68198, USA
| |
Collapse
|
13
|
Zheng J, Chen C, Guo C, Caba C, Tong Y, Wang H. The Pleiotropic Ubiquitin-Specific Peptidase 16 and Its Many Substrates. Cells 2023; 12:886. [PMID: 36980227 PMCID: PMC10047310 DOI: 10.3390/cells12060886] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2023] [Revised: 02/22/2023] [Accepted: 03/09/2023] [Indexed: 03/16/2023] Open
Abstract
Ubiquitin-specific peptidase 16 (USP16) is a deubiquitinase that plays a role in the regulation of gene expression, cell cycle progression, and various other functions. It was originally identified as the major deubiquitinase for histone H2A and has since been found to deubiquitinate a range of other substrates, including proteins from both the cytoplasm and nucleus. USP16 is phosphorylated when cells enter mitosis and dephosphorylated during the metaphase/anaphase transition. While much of USP16 is localized in the cytoplasm, separating the enzyme from its substrates is considered an important regulatory mechanism. Some of the functions that USP16 has been linked to include DNA damage repair, immune disease, tumorigenesis, protein synthesis, coronary artery health, and male infertility. The strong connection to immune response and the fact that multiple oncogene products are substrates of USP16 suggests that USP16 may be a potential therapeutic target for the treatment of certain human diseases.
Collapse
Affiliation(s)
- Jiahuan Zheng
- Department of Internal Medicine, Division of Hematology, Oncology, and Palliative Care, Massey Cancer Center, School of Medicine, Virginia Commonwealth University, Richmond, VA 23298, USA
- Department of Obstetrics and Gynecology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong 999077, China
| | - Chunxu Chen
- Department of Bioengineering, School of Engineering, Virginia Commonwealth University, Richmond, VA 23298, USA
| | - Chunqing Guo
- Department of Human and Molecular Genetics, School of Medicine, Virginia Commonwealth University, Richmond, VA 23298, USA
| | - Cody Caba
- Department of Chemistry and Biochemistry, University of Windsor, Windsor, ON N9B 3P4, Canada
| | - Yufeng Tong
- Department of Chemistry and Biochemistry, University of Windsor, Windsor, ON N9B 3P4, Canada
| | - Hengbin Wang
- Department of Internal Medicine, Division of Hematology, Oncology, and Palliative Care, Massey Cancer Center, School of Medicine, Virginia Commonwealth University, Richmond, VA 23298, USA
| |
Collapse
|
14
|
Epigenetics and Metabolism Reprogramming Interplay into Glioblastoma: Novel Insights on Immunosuppressive Mechanisms. Antioxidants (Basel) 2023; 12:antiox12020220. [PMID: 36829778 PMCID: PMC9952003 DOI: 10.3390/antiox12020220] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2022] [Revised: 01/09/2023] [Accepted: 01/16/2023] [Indexed: 01/19/2023] Open
Abstract
The central nervous system represents a complex environment in which glioblastoma adapts skillfully, unleashing a series of mechanisms suitable for its efficient development and diffusion. In particular, changes in gene expression and mutational events that fall within the domain of epigenetics interact complexly with metabolic reprogramming and stress responses enacted in the tumor microenvironment, which in turn fuel genomic instability by providing substrates for DNA modifications. The aim of this review is to analyze this complex interaction that consolidates several conditions that confer a state of immunosuppression and immunoevasion, making glioblastoma capable of escaping attack and elimination by immune cells and therefore invincible against current therapies. The progressive knowledge of the cellular mechanisms that underlie the resistance of the glioblastoma represents, in fact, the only weapon to unmask its weak points to be exploited to plan successful therapeutic strategies.
Collapse
|