1
|
Zhang Y, Ye X, Xu J, He J, Lu X. Identification and functional characterization of glycosyltransferase-related biomarkers for tuberculosis diagnosis. AMB Express 2025; 15:56. [PMID: 40153107 PMCID: PMC11953506 DOI: 10.1186/s13568-025-01870-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2024] [Accepted: 03/17/2025] [Indexed: 03/30/2025] Open
Abstract
Tuberculosis (TB) is an infectious disease that presents a serious risk to public health. Glycosyltransferase-related genes (GTRGs) are instrumental in assessing the risk of latent tuberculosis infection progressing to active TB. This study aims to develop novel, accurate, and effective diagnostic markers to enhance the early diagnosis and precision treatment of TB. We employed Weighted Gene Co-expression Network Analysis (WGCNA) to explore key genes that are notably linked toTB. In addition, we employed single-sample Gene Set Enrichment Analysis (ssGSEA) to examine the differences in immune cell infiltration between normal tissues and those affected by TB. The effectiveness of the potential biomarkers was evaluated through Receiver Operating Characteristic (ROC) curves and their expression patterns. We also conducted single-gene enrichment analysis to explore the biological functions and pathway activities linked to the characteristic genes. Finally, we constructed a competitive endogenous RNA (ceRNA) network to elucidate the potential regulatory mechanisms governing these genes. Through the screening of hub genes and differentially expressed genes from the GTRGs, we identified two potential biomarkers: B4GALT5 and KCNJ2. Evaluation results indicated that these characteristic genes displayed strong diagnostic performance in both the training and validation cohorts. Moreover, single-gene enrichment analysis revealed that these genes were primarily enriched in apoptosis pathways closely associated with TB treatment. Additionally, the construction of the mRNA-miRNA-lncRNA network identified 82 miRNAs and 65 lncRNAs. This study elucidates the roles of GTRGs in TB, identifies biomarkers associated with these groups, and establishes the lncRNA expression profile of characteristic genes. These findings provide a theoretical foundation for the early diagnosis of TB.
Collapse
Affiliation(s)
- Yibiao Zhang
- Laboratory Department, Zhejiang Jinhua Guangfu Tumor Hospital, No.1296, North Huancheng Road, Wucheng District, Jinhua, 321000, Zhejiang, China
| | - Xiaoyun Ye
- Laboratory Department, Zhejiang Jinhua Guangfu Tumor Hospital, No.1296, North Huancheng Road, Wucheng District, Jinhua, 321000, Zhejiang, China
| | - Jiancong Xu
- Laboratory Department, Zhejiang Jinhua Guangfu Tumor Hospital, No.1296, North Huancheng Road, Wucheng District, Jinhua, 321000, Zhejiang, China
| | - Jixiao He
- Lizhen Life Technology Co.Ltd, Hangzhou, 310000, Zhejiang, China
| | - Xiaobing Lu
- Laboratory Department, Zhejiang Jinhua Guangfu Tumor Hospital, No.1296, North Huancheng Road, Wucheng District, Jinhua, 321000, Zhejiang, China.
| |
Collapse
|
2
|
Xu Y, Yang L, Li G, Rao C. The Role of NF-κB/MIR155HG in Regulating the Stemness and Radioresistance in Breast Cancer Stem Cells. FRONT BIOSCI-LANDMRK 2025; 30:25810. [PMID: 39862080 DOI: 10.31083/fbl25810] [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: 07/23/2024] [Revised: 10/28/2024] [Accepted: 11/04/2024] [Indexed: 01/27/2025]
Abstract
BACKGROUND Breast cancer stem cells (BCSCs) are instrumental in treatment resistance, recurrence, and metastasis. The development of breast cancer and radiation sensitivity is intimately pertinent to long non-coding RNA (lncRNA). This work is formulated to investigate how the lncRNA MIR155HG affects the stemness and radioresistance of BCSCs. METHODS Effects of MIR155HG knockdown on BCSCs were gauged in MCF-7 and MDA-MB-231 cell lines. MIR155HG expression was manipulated in cells, followed by an assessment of stemness, DNA damage repair, apoptosis, cell cycle, and the Wnt signaling pathway under radiation conditions. The interaction between nuclear factor kappa B (NF-κB) subunit RelA and MIR155HG was examined using a dual-luciferase reporter assay. To examine the binding interaction between RelA and MIR155HG promoter, chromatin immunoprecipitation was performed. RESULTS Breast cancer-derived stem cells exhibited a high level of MIR155HG. Knockdown of MIR155HG reduced stemness, enhanced radiosensitivity, induced apoptosis, and arrested cells in the G1 phase. Mechanistically, MIR155HG knockdown repressed Wnt/β-catenin signaling and mediated apoptosis-related protein expressions. NF-κB subunit RelA transcriptionally activated MIR155HG, thereby contributing to radioresistance in BCSCs. CONCLUSION NF-κB regulates MIR155HG transcriptionally to activate the Wnt pathway, thus enhancing stemness and radioresistance in BCSCs. Targeting MIR155HG may enhance the susceptibility of cancer stem cells to radiation-induced cell death, potentially improving therapeutic outcomes. These findings underscore MIR155HG as a promising therapeutic target for breast cancer.
Collapse
Affiliation(s)
- Yunbao Xu
- Department of Chemoradiotherapy, Ningbo No 2 Hospital, 315000 Ningbo, Zhejiang, China
| | - Lu Yang
- Department of Chemoradiotherapy, Ningbo No 2 Hospital, 315000 Ningbo, Zhejiang, China
| | - Guangming Li
- Department of Breast Surgery, Ningbo No 2 Hospital, 315000 Ningbo, Zhejiang, China
| | - Chuangzhou Rao
- Department of Chemoradiotherapy, Ningbo No 2 Hospital, 315000 Ningbo, Zhejiang, China
| |
Collapse
|
3
|
Li J, Jin H, Zhao X, Sun X, Zhong J, Zhao J, Yan M. Effect of β-1,4-GalTI on the biological function of astrocytes treated by LPS. BIOMOLECULES & BIOMEDICINE 2024; 25:226-239. [PMID: 39284278 PMCID: PMC11647251 DOI: 10.17305/bb.2024.11088] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/05/2024] [Revised: 09/02/2024] [Accepted: 09/02/2024] [Indexed: 12/14/2024]
Abstract
Inflammation of the central nervous system (CNS) is a common feature of neurological disorders and infections, playing a crucial role in the development of CNS-related conditions. CNS inflammation is primarily regulated by glial cells, with astrocytes being the most abundant type in the mammalian CNS. Numerous studies have demonstrated that astrocytes, as immunocompetent cells, perform diverse and complex functions in both health and disease. Glycosylation, a critical post-translational modification of proteins, regulates numerous biological functions. The expression and activity of glycosyltransferases, the enzymes responsible for glycosylation, are closely associated with the pathogenesis of various diseases. β-1,4-GalTI, a mammalian glycosyltransferase, plays a significant role in cell-cell interactions, adhesion, and migration. Although many studies have focused on β-1,4-GalTI, few have explored its effects on astrocyte function. In this study, we constructed lentiviral vectors for both interference and overexpression of β-1,4-GalTI and discovered that β-1,4-GalTI knockdown inhibited astrocyte migration and proliferation, while its overexpression promoted these processes. Concurrently, β-1,4-GalTI knockdown reduced the expression of TNF-α, IL-1β, and IL-6, whereas overexpression enhanced the expression of these cytokines. These findings suggest that modulating β-1,4-GalTI activity can influence the molecular functions of astrocytes and provide a theoretical foundation for further research into β-1,4-GalTI as a potential therapeutic target in astrocyte-mediated inflammation.
Collapse
Affiliation(s)
- Jiyu Li
- Department of Orthopedic Oncology, Second Affiliated Hospital of Naval Medical University, Shanghai, China
| | - Hui Jin
- The Jiangsu Key Laboratory of Neuroregeneration, Nantong University, Nantong, China
| | - Xinmin Zhao
- The Jiangsu Key Laboratory of Neuroregeneration, Nantong University, Nantong, China
| | - Xinran Sun
- The Jiangsu Key Laboratory of Neuroregeneration, Nantong University, Nantong, China
| | - Jiyuan Zhong
- The Jiangsu Key Laboratory of Neuroregeneration, Nantong University, Nantong, China
| | - Jian Zhao
- Department of Orthopedic Oncology, Second Affiliated Hospital of Naval Medical University, Shanghai, China
| | - Meijuan Yan
- The Jiangsu Key Laboratory of Neuroregeneration, Nantong University, Nantong, China
| |
Collapse
|
4
|
Jiang M, Yu H. Ginsenoside 20(S)-Rg3 Hinders Esophageal Squamous Cell Carcinoma Cells Malignant Behaviors by miR-210-3p/B4GALT5 Axis. Cell Biochem Biophys 2024:10.1007/s12013-024-01566-5. [PMID: 39422791 DOI: 10.1007/s12013-024-01566-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/17/2024] [Indexed: 10/19/2024]
Abstract
Ginsenoside 20(S)-Rg3 (20(S)-Rg3) belongs to a natural chemical with an anti-tumor function, but its potential function and underlying mechanism in esophageal squamous cell carcinoma (ESCC) are unknown. Several reports have manifested that microRNA (miRNA) miR-210-3p functions as a tumor repressor in tumors, but its biofunction in ESCC remains obscure. Herein, the role and interaction of 20(S)-Rg3 and miR-210-3p in ESCC cells were investigated. We performed a series of functional experiments to validate that 20(S)-Rg3 notably restrained ESCC cell proliferation and migration while promoting cell apoptosis. Besides, miR-210-3p was found to be lowly expressed in ESCC cells. Overexpressing miR-210-3p suppressed the malignant behaviors of ESCC cells. More importantly, 20(S)-Rg3 could upregulate miR-210-3p expression in ESCC cells. MiR-210-3p knockdown offset the inhibitive impacts of 20(S)-Rg3 treatment on ESCC cell growth and migration. Furthermore, through luciferase reporter assay, beta-1,4-galactosyltransferase 5 (B4GALT5) was certified to be targeted by miR-210-3p. B4GALT5 upregulation neutralized the suppressive function of 20(S)-Rg3 on ESCC progression. Overall, 20(S)-Rg3 attenuated malignant behaviors of ESCC cells by modulating miR-210-3p/B4GALT5 axis, indicating 20(S)-Rg3 has therapeutic potential for ESCC.
Collapse
Affiliation(s)
- Min Jiang
- Department of Pathology, The Affiliated Taizhou People's Hospital of Nanjing Medical University, Taizhou School of Clinical Medicine, Nanjing Medical University, Taizhou, Jiangsu, China
| | - Hong Yu
- Department of Pathology, The Affiliated Taizhou People's Hospital of Nanjing Medical University, Taizhou School of Clinical Medicine, Nanjing Medical University, Taizhou, Jiangsu, China.
| |
Collapse
|
5
|
He M, Zhou X, Wang X. Glycosylation: mechanisms, biological functions and clinical implications. Signal Transduct Target Ther 2024; 9:194. [PMID: 39098853 PMCID: PMC11298558 DOI: 10.1038/s41392-024-01886-1] [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: 10/21/2023] [Revised: 05/25/2024] [Accepted: 06/07/2024] [Indexed: 08/06/2024] Open
Abstract
Protein post-translational modification (PTM) is a covalent process that occurs in proteins during or after translation through the addition or removal of one or more functional groups, and has a profound effect on protein function. Glycosylation is one of the most common PTMs, in which polysaccharides are transferred to specific amino acid residues in proteins by glycosyltransferases. A growing body of evidence suggests that glycosylation is essential for the unfolding of various functional activities in organisms, such as playing a key role in the regulation of protein function, cell adhesion and immune escape. Aberrant glycosylation is also closely associated with the development of various diseases. Abnormal glycosylation patterns are closely linked to the emergence of various health conditions, including cancer, inflammation, autoimmune disorders, and several other diseases. However, the underlying composition and structure of the glycosylated residues have not been determined. It is imperative to fully understand the internal structure and differential expression of glycosylation, and to incorporate advanced detection technologies to keep the knowledge advancing. Investigations on the clinical applications of glycosylation focused on sensitive and promising biomarkers, development of more effective small molecule targeted drugs and emerging vaccines. These studies provide a new area for novel therapeutic strategies based on glycosylation.
Collapse
Affiliation(s)
- Mengyuan He
- Department of Hematology, Shandong Provincial Hospital, Shandong University, Jinan, Shandong, 250021, China
| | - Xiangxiang Zhou
- Department of Hematology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, 250021, China.
- National Clinical Research Center for Hematologic Diseases, the First Affiliated Hospital of Soochow University, Suzhou, 251006, China.
| | - Xin Wang
- Department of Hematology, Shandong Provincial Hospital, Shandong University, Jinan, Shandong, 250021, China.
- Department of Hematology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, 250021, China.
- National Clinical Research Center for Hematologic Diseases, the First Affiliated Hospital of Soochow University, Suzhou, 251006, China.
- Taishan Scholars Program of Shandong Province, Jinan, Shandong, 250021, China.
- Branch of National Clinical Research Center for Hematologic Diseases, Jinan, Shandong, 250021, China.
| |
Collapse
|
6
|
Wei H, Naruse C, Takakura D, Sugihara K, Pan X, Ikeda A, Kawasaki N, Asano M. Beta-1,4-galactosyltransferase-3 deficiency suppresses the growth of immunogenic tumors in mice. Front Immunol 2023; 14:1272537. [PMID: 37901252 PMCID: PMC10600447 DOI: 10.3389/fimmu.2023.1272537] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2023] [Accepted: 09/18/2023] [Indexed: 10/31/2023] Open
Abstract
Background Beta-1,4-galactosyltransferase-3 (B4GALT3) belongs to the family of beta-1,4-galactosyltransferases (B4GALTs) and is responsible for the transfer of UDP-galactose to terminal N-acetylglucosamine. B4GALT3 is differentially expressed in tumors and adjacent normal tissues, and is correlated with clinical prognosis in several cancers, including neuroblastoma, cervical cancer, and bladder cancer. However, the exact role of B4GALT3 in the tumor immune microenvironment (TIME) remains unclear. Here, we aimed to elucidate the function of B4GALT3 in the TIME. Methods To study the functions of B4GALT3 in cancer immunity, either weakly or strongly immunogenic tumor cells were subcutaneously transplanted into wild-type (WT) and B4galt3 knockout (KO) mice. Bone marrow transplantation and CD8+ T cell depletion experiments were conducted to elucidate the role of immune cells in suppressing tumor growth in B4galt3 KO mice. The cell types and gene expression in the tumor region and infiltrating CD8+ T cells were analyzed using flow cytometry and RNA sequencing. N-glycosylated proteins from WT and B4galt3 KO mice were compared using the liquid chromatography tandem mass spectrometry (LC-MS/MS)-based glycoproteomic approach. Results B4galt3 KO mice exhibited suppressed growth of strongly immunogenic tumors with a notable increase in CD8+ T cell infiltration within tumors. Notably, B4galt3 deficiency led to changes in N-glycan modification of several proteins, including integrin alpha L (ITGAL), involved in T cell activity and proliferation. In vitro experiments suggested that B4galt3 KO CD8+ T cells were more susceptible to activation and displayed increased downstream phosphorylation of FAK linked to ITGAL. Conclusion Our study demonstrates that B4galt3 deficiency can potentially boost anti-tumor immune responses, largely through enhancing the influx of CD8+ T cells. B4GALT3 might be suppressing cancer immunity by synthesizing the glycan structure of molecules on the CD8+ T cell surface, as evidenced by the changes in the glycan structure of ITGAL in immune cells. Importantly, B4galt3 KO mice showed no adverse effects on growth, development, or reproduction, underscoring the potential of B4GALT3 as a promising and safe therapeutic target for cancer treatment.
Collapse
Affiliation(s)
- Heng Wei
- Institute of Laboratory Animals, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Chie Naruse
- Institute of Laboratory Animals, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Daisuke Takakura
- Graduate School of Medical Life Science, Yokohama City University, Yokohama, Japan
| | - Kazushi Sugihara
- Institute of Laboratory Animals, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Xuchi Pan
- Institute of Laboratory Animals, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Aki Ikeda
- Institute of Laboratory Animals, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Nana Kawasaki
- Graduate School of Medical Life Science, Yokohama City University, Yokohama, Japan
| | - Masahide Asano
- Institute of Laboratory Animals, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| |
Collapse
|
7
|
Cui S, Zhang X, Li Y, Hu S, Wu B, Fang Z, Gao J, Li M, Wu H, Tao B, Xia H, Xu L. UGCG modulates heart hypertrophy through B4GalT5-mediated mitochondrial oxidative stress and the ERK signaling pathway. Cell Mol Biol Lett 2023; 28:71. [PMID: 37658291 PMCID: PMC10472674 DOI: 10.1186/s11658-023-00484-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2023] [Accepted: 08/17/2023] [Indexed: 09/03/2023] Open
Abstract
Mechanical pressure overload and other stimuli often contribute to heart hypertrophy, a significant factor in the induction of heart failure. The UDP-glucose ceramide glycosyltransferase (UGCG) enzyme plays a crucial role in the metabolism of sphingolipids through the production of glucosylceramide. However, its role in heart hypertrophy remains unknown. In this study, UGCG was induced in response to pressure overload in vivo and phenylephrine stimulation in vitro. Additionally, UGCG downregulation ameliorated cardiomyocyte hypertrophy, improved cardiomyocyte mitochondrial oxidative stress, and reduced the ERK signaling pathway. Conversely, UGCG overexpression in cardiomyocytes promoted heart hypertrophy development, aggravated mitochondrial oxidative stress, and stimulated ERK signaling. Furthermore, the interaction between beta-1,4-galactosyltransferase 5 (B4GalT5), which catalyses the synthesis of lactosylceramide, and UGCG was identified, which also functions as a synergistic molecule of UGCG. Notably, limiting the expression of B4GalT5 impaired the capacity of UGCG to promote myocardial hypertrophy, suggesting that B4GalT5 acts as an intermediary for UGCG. Overall, this study highlights the potential of UGCG as a modulator of heart hypertrophy, rendering it a potential target for combating heart hypertrophy.
Collapse
Affiliation(s)
- Shengyu Cui
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, 430060, China
- Cardiovascular Research Institute, Wuhan University, Wuhan, 430060, China
- Hubei Key Laboratory of Cardiology, Wuhan, 430060, China
| | - Xutao Zhang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, 430060, China
- Cardiovascular Research Institute, Wuhan University, Wuhan, 430060, China
- Hubei Key Laboratory of Cardiology, Wuhan, 430060, China
| | - Yuhua Li
- Intensive Care Unit, Wuhan Children's Hospital (Wuhan Maternal and Child Healthcare Hospital), Tongji Medical College, Huazhong University of Science & Technology, Wuhan, China
| | - Shan Hu
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, 430060, China
- Cardiovascular Research Institute, Wuhan University, Wuhan, 430060, China
- Hubei Key Laboratory of Cardiology, Wuhan, 430060, China
| | - Bing Wu
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, 430060, China
- Cardiovascular Research Institute, Wuhan University, Wuhan, 430060, China
- Hubei Key Laboratory of Cardiology, Wuhan, 430060, China
| | - Zhao Fang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, 430060, China
- Cardiovascular Research Institute, Wuhan University, Wuhan, 430060, China
- Hubei Key Laboratory of Cardiology, Wuhan, 430060, China
| | - Jixian Gao
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, 430060, China
- Cardiovascular Research Institute, Wuhan University, Wuhan, 430060, China
- Hubei Key Laboratory of Cardiology, Wuhan, 430060, China
| | - Ming Li
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, 430060, China
- Cardiovascular Research Institute, Wuhan University, Wuhan, 430060, China
- Hubei Key Laboratory of Cardiology, Wuhan, 430060, China
| | - Haoliang Wu
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, 430060, China
- Cardiovascular Research Institute, Wuhan University, Wuhan, 430060, China
- Hubei Key Laboratory of Cardiology, Wuhan, 430060, China
| | - Bo Tao
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, 430060, China
- Cardiovascular Research Institute, Wuhan University, Wuhan, 430060, China
- Hubei Key Laboratory of Cardiology, Wuhan, 430060, China
| | - Hao Xia
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, 430060, China.
- Cardiovascular Research Institute, Wuhan University, Wuhan, 430060, China.
- Hubei Key Laboratory of Cardiology, Wuhan, 430060, China.
| | - Lin Xu
- Department of Geriatrics, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei, China.
| |
Collapse
|
8
|
Ordaz-Ramos A, Tellez-Jimenez O, Vazquez-Santillan K. Signaling pathways governing the maintenance of breast cancer stem cells and their therapeutic implications. Front Cell Dev Biol 2023; 11:1221175. [PMID: 37492224 PMCID: PMC10363614 DOI: 10.3389/fcell.2023.1221175] [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: 05/11/2023] [Accepted: 06/28/2023] [Indexed: 07/27/2023] Open
Abstract
Breast cancer stem cells (BCSCs) represent a distinct subpopulation of cells with the ability to self-renewal and differentiate into phenotypically diverse tumor cells. The involvement of CSC in treatment resistance and cancer recurrence has been well established. Numerous studies have provided compelling evidence that the self-renewal ability of cancer stem cells is tightly regulated by specific signaling pathways, which exert critical roles to maintain an undifferentiated phenotype and prevent the differentiation of CSCs. Signaling pathways such as Wnt/β-catenin, NF-κB, Notch, Hedgehog, TGF-β, and Hippo have been implicated in the promotion of self-renewal of many normal and cancer stem cells. Given the pivotal role of BCSCs in driving breast cancer aggressiveness, targeting self-renewal signaling pathways holds promise as a viable therapeutic strategy for combating this disease. In this review, we will discuss the main signaling pathways involved in the maintenance of the self-renewal ability of BCSC, while also highlighting current strategies employed to disrupt the signaling molecules associated with stemness.
Collapse
Affiliation(s)
- Alejandro Ordaz-Ramos
- Innovation in Precision Medicine Laboratory, Instituto Nacional de Medicina Genómica, Mexico City, México
- Posgrado en Ciencias Biológicas, Unidad de Posgrado, Circuito de Posgrados, Ciudad Universitaria, Coyoacán, México
| | - Olivia Tellez-Jimenez
- Innovation in Precision Medicine Laboratory, Instituto Nacional de Medicina Genómica, Mexico City, México
- Posgrado en Ciencias Biológicas, Unidad de Posgrado, Circuito de Posgrados, Ciudad Universitaria, Coyoacán, México
| | - Karla Vazquez-Santillan
- Innovation in Precision Medicine Laboratory, Instituto Nacional de Medicina Genómica, Mexico City, México
| |
Collapse
|
9
|
Wei Y, Li Y, Chen Y, Liu P, Huang S, Zhang Y, Sun Y, Wu Z, Hu M, Wu Q, Wu H, Liu F, She T, Ning Z. ALDH1: A potential therapeutic target for cancer stem cells in solid tumors. Front Oncol 2022; 12:1026278. [PMID: 36387165 PMCID: PMC9650078 DOI: 10.3389/fonc.2022.1026278] [Citation(s) in RCA: 46] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Accepted: 10/12/2022] [Indexed: 12/02/2022] Open
Abstract
Solid tumors can be divided into benign solid tumors and solid malignant tumors in the academic community, among which malignant solid tumors are called cancers. Cancer is the second leading cause of death in the world, and the global incidence of cancer is increasing yearly New cancer patients in China are always the first. After the concept of stem cells was introduced in the tumor community, the CSC markers represented by ALDH1 have been widely studied due to their strong CSC cell characteristics and potential to be the driving force of tumor metastasis. In the research results in the past five years, it has been found that ALDH1 is highly expressed in various solid cancers such as breast cancer, lung cancer, colorectal cancer, liver cancer, gastric cancer, cervical cancer, esophageal cancer, ovarian cancer, head,and neck cancer. ALDH1 can activate and transform various pathways (such as the USP28/MYC signaling pathway, ALDH1A1/HIF-1α/VEGF axis, wnt/β-catenin signaling pathway), as well as change the intracellular pH value to promote formation and maintenance, resulting in drug resistance in tumors. By targeting and inhibiting ALDH1 in tumor stem cells, it can enhance the sensitivity of drugs and inhibit the proliferation, differentiation, and metastasis of solid tumor stem cells to some extent. This review discusses the relationship and pathway of ALDH1 with various solid tumors. It proposes that ALDH1 may serve as a diagnosis and therapeutic target for CSC, providing new insights and new strategies for reliable tumor treatment.
Collapse
Affiliation(s)
- Yaolu Wei
- School of Basic Medicine Sciences, Xianning Medical College, Hubei University of Science and Technology, Xianning, China
| | - Yan Li
- School of Basic Medicine Sciences, Xianning Medical College, Hubei University of Science and Technology, Xianning, China
| | - Yenan Chen
- Xianning Medical College, Hubei University of Science and Technology, Xianning, China
| | - Pei Liu
- Xianning Medical College, Hubei University of Science and Technology, Xianning, China
| | - Sheng Huang
- Xianning Medical College, Hubei University of Science and Technology, Xianning, China
| | - Yuping Zhang
- School of Basic Medicine Sciences, Xianning Medical College, Hubei University of Science and Technology, Xianning, China
| | - Yanling Sun
- School of Basic Medicine Sciences, Xianning Medical College, Hubei University of Science and Technology, Xianning, China
| | - Zhe Wu
- School of Basic Medicine Sciences, Xianning Medical College, Hubei University of Science and Technology, Xianning, China
| | - Meichun Hu
- School of Basic Medicine Sciences, Xianning Medical College, Hubei University of Science and Technology, Xianning, China
| | - Qian Wu
- School of Basic Medicine Sciences, Xianning Medical College, Hubei University of Science and Technology, Xianning, China
| | - Hongnian Wu
- School of Basic Medicine Sciences, Xianning Medical College, Hubei University of Science and Technology, Xianning, China
| | - Fuxing Liu
- School of Basic Medicine Sciences, Xianning Medical College, Hubei University of Science and Technology, Xianning, China
- *Correspondence: Fuxing Liu, ; Tonghui She, ; Zhifeng Ning,
| | - Tonghui She
- School of Basic Medicine Sciences, Xianning Medical College, Hubei University of Science and Technology, Xianning, China
- *Correspondence: Fuxing Liu, ; Tonghui She, ; Zhifeng Ning,
| | - Zhifeng Ning
- School of Basic Medicine Sciences, Xianning Medical College, Hubei University of Science and Technology, Xianning, China
- *Correspondence: Fuxing Liu, ; Tonghui She, ; Zhifeng Ning,
| |
Collapse
|
10
|
Guindolet D, Woodward AM, Gabison EE, Argüeso P. Glycogene Expression Profile of Human Limbal Epithelial Cells with Distinct Clonogenic Potential. Cells 2022; 11:cells11091575. [PMID: 35563881 PMCID: PMC9102009 DOI: 10.3390/cells11091575] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2022] [Revised: 05/05/2022] [Accepted: 05/06/2022] [Indexed: 12/13/2022] Open
Abstract
Glycans function as valuable markers of stem cells but also regulate the ability of these cells to self-renew and differentiate. Approximately 2% of the human genome encodes for proteins that are involved in the biosynthesis and recognition of glycans. In the present study, we evaluated the expression of a small subset of glycogenes in human limbal epithelial cells with distinct clonogenic potential. Individual clones were classified as abortive or clonogenic, based on the fraction of the terminal colonies produced; clones leading exclusively to terminal colonies were referred to as abortive while those with half or fewer terminal colonies were referred to as clonogenic. An analysis of glycogene expression in clonogenic cultures revealed a high content of transcripts regulating the galactose and mannose metabolic pathways. Abortive clones were characterized by increased levels of GCNT4 and FUCA2, genes that are responsible for the branching of mucin-type O-glycans and the hydrolysis of fucose residues on N-glycans, respectively. The expansion of primary cultures of human limbal epithelial cells for 10 days resulted in stratification and a concomitant increase in MUC16, GCNT4 and FUCA2 expression. These data indicate that the clonogenic potential of human limbal epithelial cells is associated with specific glycosylation pathways. Mucin-type O-glycan branching and increased fucose metabolism are linked to limbal epithelial cell differentiation.
Collapse
Affiliation(s)
- Damien Guindolet
- Schepens Eye Research Institute of Mass. Eye and Ear, Department of Ophthalmology, Harvard Medical School, 20 Staniford Street, Boston, MA 02114, USA; (D.G.); (A.M.W.)
- Hôpital Fondation A. de Rothschild, Department of Ophthalmology, 25 rue Manin, 75019 Paris, France
| | - Ashley M. Woodward
- Schepens Eye Research Institute of Mass. Eye and Ear, Department of Ophthalmology, Harvard Medical School, 20 Staniford Street, Boston, MA 02114, USA; (D.G.); (A.M.W.)
| | - Eric E. Gabison
- Hôpital Fondation A. de Rothschild, Department of Ophthalmology, 25 rue Manin, 75019 Paris, France
- Correspondence: (E.E.G.); (P.A.)
| | - Pablo Argüeso
- Schepens Eye Research Institute of Mass. Eye and Ear, Department of Ophthalmology, Harvard Medical School, 20 Staniford Street, Boston, MA 02114, USA; (D.G.); (A.M.W.)
- Correspondence: (E.E.G.); (P.A.)
| |
Collapse
|
11
|
Glycosyltransferases in Cancer: Prognostic Biomarkers of Survival in Patient Cohorts and Impact on Malignancy in Experimental Models. Cancers (Basel) 2022; 14:cancers14092128. [PMID: 35565254 PMCID: PMC9100214 DOI: 10.3390/cancers14092128] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2022] [Revised: 04/19/2022] [Accepted: 04/21/2022] [Indexed: 02/04/2023] Open
Abstract
Background: Glycosylation changes are a main feature of cancer. Some carbohydrate epitopes and expression levels of glycosyltransferases have been used or proposed as prognostic markers, while many experimental works have investigated the role of glycosyltransferases in malignancy. Using the transcriptomic data of the 21 TCGA cohorts, we correlated the expression level of 114 glycosyltransferases with the overall survival of patients. Methods: Using the Oncolnc website, we determined the Kaplan−Meier survival curves for the patients falling in the 15% upper or lower percentile of mRNA expression of each glycosyltransferase. Results: Seventeen glycosyltransferases involved in initial steps of N- or O-glycosylation and of glycolipid biosynthesis, in chain extension and sialylation were unequivocally associated with bad prognosis in a majority of cohorts. Four glycosyltransferases were associated with good prognosis. Other glycosyltransferases displayed an extremely high predictive value in only one or a few cohorts. The top were GALNT3, ALG6 and B3GNT7, which displayed a p < 1 × 10−9 in the low-grade glioma (LGG) cohort. Comparison with published experimental data points to ALG3, GALNT2, B4GALNT1, POFUT1, B4GALT5, B3GNT5 and ST3GAL2 as the most consistently malignancy-associated enzymes. Conclusions: We identified several cancer-associated glycosyltransferases as potential prognostic markers and therapeutic targets.
Collapse
|
12
|
Sha Y, Han L, Sun B, Zhao Q. Identification of a Glycosyltransferase Signature for Predicting Prognosis and Immune Microenvironment in Neuroblastoma. Front Cell Dev Biol 2022; 9:769580. [PMID: 35071226 PMCID: PMC8773256 DOI: 10.3389/fcell.2021.769580] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2021] [Accepted: 11/30/2021] [Indexed: 01/17/2023] Open
Abstract
Neuroblastoma (NB) is one of the most common solid tumors in children. Glycosyltransferases (GTs) play a crucial role in tumor development and immune escape and have been used as prognostic biomarkers in various tumors. However, the biological functions and prognostic significance of GTs in NB remain poorly understood. The expression data from Gene Expression Omnibus (GEO) and Therapeutically Applicable Research to Generate Effective Treatments (TARGET) were collected as training and testing data. Based on a progression status, differentially expressed GTs were identified. We constructed a GTscore through support vector machine, least absolute shrinkage and selection operator, and Cox regression in NB, which included four prognostic GTs and was an independent prognostic risk factor for NB. Patients in the high GTscore group had an older age, MYCN amplification, advanced International Neuroblastoma Staging System stage, and high risk. Samples with high GTscores revealed high disialoganglioside (GD2) and neuron-specific enolase expression levels. In addition, a lack of immune cell infiltration was observed in the high GTscore group. This GTscore was also associated with the expression of chemokines (CCL2, CXCL9, and CXCL10) and immune checkpoint genes (cytotoxic T-lymphocyte–associated protein 4, granzyme H, and granzyme K). A low GTscore was also linked to an enhanced response to anti–PD-1 immunotherapy in melanoma patients, and one type of tumor was also derived from neuroectodermal cells such as NB. In conclusion, the constructed GTscore revealed the relationship between GT expression and the NB outcome, GD2 phenotype, and immune infiltration and provided novel clues for the prediction of prognosis and immunotherapy response in NB.
Collapse
Affiliation(s)
- Yongliang Sha
- Department of Pediatric Oncology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China.,National Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China.,Key Laboratory of Cancer Prevention and Therapy, Tianjin, China.,Tianjin's Clinical Research Center for Cancer, Tianjin, China
| | - Lei Han
- National Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China.,Key Laboratory of Cancer Prevention and Therapy, Tianjin, China.,Tianjin's Clinical Research Center for Cancer, Tianjin, China.,Cancer Molecular Diagnostics Core, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Bei Sun
- National Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China.,Key Laboratory of Cancer Prevention and Therapy, Tianjin, China.,Tianjin's Clinical Research Center for Cancer, Tianjin, China.,Department of Outpatient Office, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Qiang Zhao
- Department of Pediatric Oncology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China.,National Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China.,Key Laboratory of Cancer Prevention and Therapy, Tianjin, China.,Tianjin's Clinical Research Center for Cancer, Tianjin, China
| |
Collapse
|
13
|
Wang L. A Novel Glycosyltransferase-Related Gene Signature for Overall Survival Prediction in Patients with Ovarian Cancer. Int J Gen Med 2022; 14:10337-10350. [PMID: 34992448 PMCID: PMC8717217 DOI: 10.2147/ijgm.s332945] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2021] [Accepted: 12/14/2021] [Indexed: 12/21/2022] Open
Abstract
Background Ovarian cancer is a highly malignant epithelial tumor. Recently, it has been reported the role of glycosyltransferases (GTs) in various cancers. However, the prognostic value of GTs-related genes in ovarian cancer remained largely unknown. Methods RNA-sequencing (RNA-seq) data and corresponding clinical characteristics of patients with ovarian cancer were extracted from the public database of the Cancer Genome Atlas (TCGA) and Genotype-Tissue Expression (GTEx). We constructed the least absolute shrinkage and selection operator (LASSO) Cox regression model to explore a multigene signature comprising GTs-related genes in the TCGA and GTEx cohort. Patients with ovarian cancer in International Cancer Genome Consortium (ICGC) database were applied for further validation. We also performed functional analysis on the differentially expressed genes (DEGs) of high-risk and low-risk groups in the TCGA cohort. Additionally, the immune status between the two risk groups was assessed, respectively. Results Our results showed that 64 GTs-related genes were differentially expressed between tumor tissues and normal tissues in the TCGA and GTEx cohort. A prognostic model of 15 candidate genes was constructed, which classified patients into high- and low-risk groups. Compared with low-risk patients, high-risk patients had an obvious worse overall survival (OS) (P < 0.0001 in the TCGA and GTEx cohort and P = 0.042 in the ICGC cohort). Multivariate Cox regression analysis revealed that the risk score was an independent factor for OS of ovarian cancer. Functional analysis indicated that these DEGs were also enriched in immune-related pathways, and the immune status was significantly different between the two risk groups in TCGA cohort. Conclusion In conclusion, a novel GTs-related gene signature may be used for the prognosis of ovarian cancer. Targeting GTs-related gene can act as a therapeutic alternative for ovarian cancer.
Collapse
Affiliation(s)
- Liang Wang
- Department of Gynecology and Obstetrics, Tianjin NanKai Hospital, Tianjin, 300100, People's Republic of China
| |
Collapse
|
14
|
Rizzo R, Russo D, Kurokawa K, Sahu P, Lombardi B, Supino D, Zhukovsky MA, Vocat A, Pothukuchi P, Kunnathully V, Capolupo L, Boncompain G, Vitagliano C, Zito Marino F, Aquino G, Montariello D, Henklein P, Mandrich L, Botti G, Clausen H, Mandel U, Yamaji T, Hanada K, Budillon A, Perez F, Parashuraman S, Hannun YA, Nakano A, Corda D, D'Angelo G, Luini A. Golgi maturation-dependent glycoenzyme recycling controls glycosphingolipid biosynthesis and cell growth via GOLPH3. EMBO J 2021; 40:e107238. [PMID: 33749896 DOI: 10.15252/embj.2020107238] [Citation(s) in RCA: 63] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2020] [Revised: 01/24/2021] [Accepted: 02/10/2021] [Indexed: 01/08/2023] Open
Abstract
Glycosphingolipids are important components of the plasma membrane where they modulate the activities of membrane proteins including signalling receptors. Glycosphingolipid synthesis relies on competing reactions catalysed by Golgi-resident enzymes during the passage of substrates through the Golgi cisternae. The glycosphingolipid metabolic output is determined by the position and levels of the enzymes within the Golgi stack, but the mechanisms that coordinate the intra-Golgi localisation of the enzymes are poorly understood. Here, we show that a group of sequentially-acting enzymes operating at the branchpoint among glycosphingolipid synthetic pathways binds the Golgi-localised oncoprotein GOLPH3. GOLPH3 sorts these enzymes into vesicles for intra-Golgi retro-transport, acting as a component of the cisternal maturation mechanism. Through these effects, GOLPH3 controls the sub-Golgi localisation and the lysosomal degradation rate of specific enzymes. Increased GOLPH3 levels, as those observed in tumours, alter glycosphingolipid synthesis and plasma membrane composition thereby promoting mitogenic signalling and cell proliferation. These data have medical implications as they outline a novel oncogenic mechanism of action for GOLPH3 based on glycosphingolipid metabolism.
Collapse
Affiliation(s)
- Riccardo Rizzo
- Institute of Biochemistry and Cell Biology, National Research Council, Naples, Italy.,Institute of Nanotechnology, National Research Council (CNR-NANOTEC), Lecce, Italy
| | - Domenico Russo
- Institute of Biochemistry and Cell Biology, National Research Council, Naples, Italy
| | - Kazuo Kurokawa
- Live Cell Super-Resolution Imaging Research Team, RIKEN Center for Advanced Photonics, Saitama, Japan
| | - Pranoy Sahu
- Institute of Biochemistry and Cell Biology, National Research Council, Naples, Italy
| | - Bernadette Lombardi
- Institute of Biochemistry and Cell Biology, National Research Council, Naples, Italy
| | - Domenico Supino
- Institute of Biochemistry and Cell Biology, National Research Council, Naples, Italy
| | - Mikhail A Zhukovsky
- Institute of Biochemistry and Cell Biology, National Research Council, Naples, Italy
| | - Anthony Vocat
- École Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - Prathyush Pothukuchi
- Institute of Biochemistry and Cell Biology, National Research Council, Naples, Italy
| | - Vidya Kunnathully
- Institute of Biochemistry and Cell Biology, National Research Council, Naples, Italy
| | - Laura Capolupo
- École Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | | | - Carlo Vitagliano
- Istituto Nazionale Tumori Fondazione G. Pascale-IRCCS, Naples, Italy
| | | | - Gabriella Aquino
- Istituto Nazionale Tumori Fondazione G. Pascale-IRCCS, Naples, Italy
| | - Daniela Montariello
- Institute of Biochemistry and Cell Biology, National Research Council, Naples, Italy
| | - Petra Henklein
- Humboldt-Universität zu Berlin, and Berlin Institute of Health, Institute of Biochemistry, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Berlin, Germany
| | - Luigi Mandrich
- Institute of Biochemistry and Cell Biology, National Research Council, Naples, Italy
| | - Gerardo Botti
- Istituto Nazionale Tumori Fondazione G. Pascale-IRCCS, Naples, Italy
| | - Henrik Clausen
- Faculty of Health Sciences, Centre for Glycomics, Department of Cellular and Molecular Medicine Nørre Alle 20, University of Copenhagen, Copenhagen N, Denmark
| | - Ulla Mandel
- Faculty of Health Sciences, Centre for Glycomics, Department of Cellular and Molecular Medicine Nørre Alle 20, University of Copenhagen, Copenhagen N, Denmark
| | - Toshiyuki Yamaji
- Department of Biochemistry & Cell Biology, National Institute of Infectious Diseases, Tokyo, Japan
| | - Kentaro Hanada
- Department of Biochemistry & Cell Biology, National Institute of Infectious Diseases, Tokyo, Japan
| | - Alfredo Budillon
- Istituto Nazionale Tumori Fondazione G. Pascale-IRCCS, Naples, Italy
| | - Franck Perez
- Institute Curie - CNRS UMR1 44, Research Center, Paris, France
| | | | - Yusuf A Hannun
- Stony Brook University Medical Center, New York, NY, USA
| | - Akihiko Nakano
- Live Cell Super-Resolution Imaging Research Team, RIKEN Center for Advanced Photonics, Saitama, Japan
| | - Daniela Corda
- Institute of Biochemistry and Cell Biology, National Research Council, Naples, Italy
| | - Giovanni D'Angelo
- Institute of Biochemistry and Cell Biology, National Research Council, Naples, Italy.,École Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - Alberto Luini
- Institute of Biochemistry and Cell Biology, National Research Council, Naples, Italy
| |
Collapse
|
15
|
Heparan Sulfate Proteoglycan Signaling in Tumor Microenvironment. Int J Mol Sci 2020; 21:ijms21186588. [PMID: 32916872 PMCID: PMC7554799 DOI: 10.3390/ijms21186588] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2020] [Revised: 09/04/2020] [Accepted: 09/08/2020] [Indexed: 12/18/2022] Open
Abstract
In the last few decades, heparan sulfate (HS) proteoglycans (HSPGs) have been an intriguing subject of study for their complex structural characteristics, their finely regulated biosynthetic machinery, and the wide range of functions they perform in living organisms from development to adulthood. From these studies, key roles of HSPGs in tumor initiation and progression have emerged, so that they are currently being explored as potential biomarkers and therapeutic targets for cancers. The multifaceted nature of HSPG structure/activity translates in their capacity to act either as inhibitors or promoters of tumor growth and invasion depending on the tumor type. Deregulation of HSPGs resulting in malignancy may be due to either their abnormal expression levels or changes in their structure and functions as a result of the altered activity of their biosynthetic or remodeling enzymes. Indeed, in the tumor microenvironment, HSPGs undergo structural alterations, through the shedding of proteoglycan ectodomain from the cell surface or the fragmentation and/or desulfation of HS chains, affecting HSPG function with significant impact on the molecular interactions between cancer cells and their microenvironment, and tumor cell behavior. Here, we overview the structural and functional features of HSPGs and their signaling in the tumor environment which contributes to tumorigenesis and cancer progression.
Collapse
|