1
|
Xiong WW, Ouyang ZJ, Tan Y, Xu LW, Peng XW. MUC4 O-GlcNAcylation Regulates the Epithelial Phenotype in Conjunctival Epithelial Cells. Biochem Genet 2025; 63:1780-1790. [PMID: 38627316 DOI: 10.1007/s10528-024-10791-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2023] [Accepted: 03/20/2024] [Indexed: 03/23/2025]
Abstract
In the present study, our aim was to explore the role of MUC4 in IL-4-stimulated conjunctival epithelial cells and the underlying mechanisms. Human recombinant IL-4 was employed in human conjunctival epithelial cells (HConEpic) cells, and MUC4 shRNA (sh-MUC4) was constructed to explore the functional role of MUC4. The protein level of MUC4, O-GlcNAc transferase (OGT), O-GlcNAc hydrolase (OGA), zonula occludens 1 (ZO-1), gap junction protein beta 2 (GJB2), claudin-8 (CLDN8), and E-cadherin were detected by Western blot in HConEpic cells, the interaction between MUC4 and OGT/OGA was assessed by co-immunoprecipitation (IP) and Western blot in 293T cells. Our results showed that IL-4 significantly up-regulated MUC4 and OGT protein levels in HConEpic cells, while down-regulated OGA protein level. Also, IL-4 down-regulated ZO-1, GJB2, CLDN8, and E-cadherin protein levels in HConEpic cells, while which was markedly reversed by sh-MUC4. Additionally, OGT inhibitor significantly reduced MUC4 protein level, and elevated ZO-1, GJB2, CLDN8, and E-cadherin protein levels in HConEpic cells, while OGA inhibitor resulted in the opposite results. Furthermore, in addition to the interaction between OGT/OGA and MUC4, Co-IP and Western blot also revealed the alteration of MUC4 O-GlcNAcylation in 293T cells treated with OGT/OGA inhibitor. Above findings suggested that OGT/OGA inhibitor regulated MUC4 protein level by affecting MUC4 O-GlcNAcylation to regulate ZO-1, GJB2, CLDN8, and E-cadherin protein levels in HConEpic cells, which was achieved via inhibiting the interaction between OGT/OGA and MUC4. This study may provide a better understanding of the pathogenesis of allergic conjunctivitis (AC).
Collapse
Affiliation(s)
- Wei-Wei Xiong
- Department of Ophthalmology, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, NO.1 Minde Road, Nanchang, 330006, Jiangxi, China
| | - Zi-Jing Ouyang
- Department of Ophthalmology, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, NO.1 Minde Road, Nanchang, 330006, Jiangxi, China
| | - Yi Tan
- Department of Ophthalmology, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, NO.1 Minde Road, Nanchang, 330006, Jiangxi, China
| | - Li-Wen Xu
- Department of Ophthalmology, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, NO.1 Minde Road, Nanchang, 330006, Jiangxi, China
| | - Xiao-Wei Peng
- Department of Ophthalmology, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, NO.1 Minde Road, Nanchang, 330006, Jiangxi, China.
| |
Collapse
|
2
|
Cheng SS, Mody AC, Woo CM. Opportunities for Therapeutic Modulation of O-GlcNAc. Chem Rev 2024; 124:12918-13019. [PMID: 39509538 DOI: 10.1021/acs.chemrev.4c00417] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2024]
Abstract
O-Linked β-N-acetylglucosamine (O-GlcNAc) is an essential, dynamic monosaccharide post-translational modification (PTM) found on serine and threonine residues of thousands of nucleocytoplasmic proteins. The installation and removal of O-GlcNAc is controlled by a single pair of enzymes, O-GlcNAc transferase (OGT) and O-GlcNAcase (OGA), respectively. Since its discovery four decades ago, O-GlcNAc has been found on diverse classes of proteins, playing important functional roles in many cellular processes. Dysregulation of O-GlcNAc homeostasis has been implicated in the pathogenesis of disease, including neurodegeneration, X-linked intellectual disability (XLID), cancer, diabetes, and immunological disorders. These foundational studies of O-GlcNAc in disease biology have motivated efforts to target O-GlcNAc therapeutically, with multiple clinical candidates under evaluation. In this review, we describe the characterization and biochemistry of OGT and OGA, cellular O-GlcNAc regulation, development of OGT and OGA inhibitors, O-GlcNAc in pathophysiology, clinical progress of O-GlcNAc modulators, and emerging opportunities for targeting O-GlcNAc. This comprehensive resource should motivate further study into O-GlcNAc function and inspire strategies for therapeutic modulation of O-GlcNAc.
Collapse
Affiliation(s)
- Steven S Cheng
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, Massachusetts 02138, United States
| | - Alison C Mody
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, Massachusetts 02138, United States
| | - Christina M Woo
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, Massachusetts 02138, United States
- Affiliate member of the Broad Institute, Cambridge, Massachusetts 02142, United States
| |
Collapse
|
3
|
Zhou Q, Yang M, Fu J, Sun X, Wang J, Zhang H, Hu J, Han B. KIF1A promotes neuroendocrine differentiation in prostate cancer by regulating the OGT-mediated O-GlcNAcylation. Cell Death Dis 2024; 15:796. [PMID: 39505875 PMCID: PMC11542072 DOI: 10.1038/s41419-024-07142-2] [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: 06/21/2024] [Revised: 09/27/2024] [Accepted: 10/07/2024] [Indexed: 11/08/2024]
Abstract
Neuroendocrine prostate cancer (NEPC) arises from prostate adenocarcinoma after endocrine treatment failure and implies lethality and limited therapeutic options. Deciphering the molecular mechanisms underlying transdifferentiation from adenocarcinoma to NEPC may provide valuable therapeutic strategies. We performed a pan-cancer differential mRNA abundance analysis and identified that Kinesin-like protein (KIF1A) was highly expressed in NEPC. KIF1A knockdown impaired neuroendocrine(NE) features, including NE marker gene expression, stemness, and epithelial-mesenchymal transition (EMT), whereas KIF1A overexpression promoted these processes. Targeting KIF1A inhibited the growth of NE differentiated prostate cancer (PCa) cells in vitro and in vivo. Mechanistically, KIF1A bound with O-linked N-acetylglucosamine transferase (OGT) and regulated its protein expression and activity. Nuclear accumulation of OGT induced by KIF1A overexpression promoted intranuclear O-GlcNAcylation of β-catenin and OCT4 in nucleus. More importantly, our data revealed that OGT was critical for KIF1A induced NE differentiation and aggressive tumor growth. An OGT inhibitor, OSMI-1, can significantly inhibited NE differentiated PCa cell proliferation in vitro and tumor growth in vivo. Our findings showed that KIF1A promotes NE differentiation to NEPC by regulating the OGT-mediated O-GlcNAcylation. Targeting O-GlcNAcylation may impede the development of NEPC for a group of PCa patients with elevated KIF1A expression.
Collapse
Affiliation(s)
- Qianqian Zhou
- The Key Laboratory of Experimental Teratology, Ministry of Education and Department of Pathology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250000, P R China
| | - Muyi Yang
- Department of Oncology-Pathology, Karolinska Institute, Stockholm, Sweden
| | - Jiawei Fu
- The Key Laboratory of Experimental Teratology, Ministry of Education and Department of Pathology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250000, P R China
| | - Xinyu Sun
- Jinan Central Hospital, Shandong University, Jinan, Shandong, 250000, P R China
| | - Jiajia Wang
- The Key Laboratory of Experimental Teratology, Ministry of Education and Department of Pathology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250000, P R China
| | - Hanwen Zhang
- The Key Laboratory of Experimental Teratology, Ministry of Education and Department of Pathology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250000, P R China
| | - Jing Hu
- Department of Pathology, Shandong University Qilu Hospital, Jinan, Shandong, 250000, P R China
| | - Bo Han
- The Key Laboratory of Experimental Teratology, Ministry of Education and Department of Pathology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250000, P R China.
- Department of Pathology, Shandong University Qilu Hospital, Jinan, Shandong, 250000, P R China.
| |
Collapse
|
4
|
Chen R, Hu J, Zhang Y, Liu Y, Cao L, He F, Wang Q, Chen Y, Zhang S, Tang S, Min B. Sodium aescinate alleviates neuropathic pain through suppressing OGT-mediated O-GlcNAc modification of TLR3 to inactivate MAPK signaling pathway. Brain Res Bull 2024; 217:111077. [PMID: 39265741 DOI: 10.1016/j.brainresbull.2024.111077] [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: 07/01/2024] [Revised: 09/08/2024] [Accepted: 09/10/2024] [Indexed: 09/14/2024]
Abstract
Neuropathic pain results from damage to nerves or the brain, and is characterized by symptoms such as allodynia, spontaneous pain, and hyperalgesia. The causes of this type of pain are intricate, which can make it difficult to treat. Sodium aescinate (SA), a natural extract from horse chestnut tree seeds, has been shown to act as a neuroprotector by inhibiting microglia activation. This study aims to explore the therapeutic potential of SA for neuropathic pain and the molecular mechanisms regulated by SA treatment. Through in vivo animal models and experiments, we found that SA treatment significantly reduced mechanical allodynia and heat hyperalgesia in neuropathic pain models. Additionally, SA inhibited O-GlcNAc-transferase (OGT)-induced O-GlcNAcylation (O-GlcNAc) modification in neuropathic pain mice. OGT overexpression could impede the therapeutic effects of SA on neuropathic pain. Further investigation revealed that Toll-like receptor 3 (TLR3), stabilized by OGT-induced O-GlcNAc modification, could activate the Mitogen activated protein kinase (MAPK) signaling pathway. Further in vivo experiments demonstrated that TLR3-mediated p38 mitogen-activated protein kinase (p38MAPK) activation is involved in SA-mediated relief of neuropathic pain. In conclusion, this study uncovers a novel molecular pathway deactivated by SA treatment in neuropathic pain.
Collapse
Affiliation(s)
- Rong Chen
- Department of Pain, The First Affiliated Hospital of Guizhou University of Traditional Chinese Medicine, Guiyang, Guizhou 550001, China
| | - Jiantao Hu
- Department of Respiratory Medicine, Bijie City Qixingguan District People's Hospital, Bijie, Guizhou 551700, China
| | - Yang Zhang
- Department of Anesthesiology, The First Affiliated Hospital of Guizhou University of Traditional Chinese Medicine, Guiyang, Guizhou 550001, China
| | - Yang Liu
- Department of Orthopedics, The First Affiliated Hospital of Guizhou University of Traditional Chinese Medicine, Guiyang, Guizhou 550001, China
| | - Liujian Cao
- Department of Anorectal Surgery, The First Affiliated Hospital of Guizhou University of Traditional Chinese Medicine, Guiyang, Guizhou 550001, China
| | - Fan He
- Department of Oncology, The First Affiliated Hospital of Guizhou University of Traditional Chinese Medicine, Guiyang, Guizhou 550001, China
| | - Qin Wang
- Department of Rheumatology, The First Affiliated Hospital of Guizhou University of Traditional Chinese Medicine, Guiyang, Guizhou 550001, China
| | - Ying Chen
- Department of Anesthesiology, The First Affiliated Hospital of Guizhou University of Traditional Chinese Medicine, Guiyang, Guizhou 550001, China
| | - Shengwei Zhang
- Graduate School, Guizhou University of Traditional Chinese Medicine, Guiyang, Guizhou 550001, China.
| | - Songjiang Tang
- Department of Anesthesiology, The First Affiliated Hospital of Guizhou University of Traditional Chinese Medicine, Guiyang, Guizhou 550001, China
| | - Baojun Min
- Department of Anesthesiology, Qianxi People's Hospital, Bijie, Guizhou 551500, China.
| |
Collapse
|
5
|
You C, Shen F, Yang P, Cui J, Ren Q, Liu M, Hu Y, Li B, Ye L, Shi Y. O-GlcNAcylation mediates Wnt-stimulated bone formation by rewiring aerobic glycolysis. EMBO Rep 2024; 25:4465-4487. [PMID: 39256595 PMCID: PMC11467389 DOI: 10.1038/s44319-024-00237-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2024] [Revised: 07/31/2024] [Accepted: 08/13/2024] [Indexed: 09/12/2024] Open
Abstract
Wnt signaling is an important target for anabolic therapies in osteoporosis. A sclerostin-neutralizing antibody (Scl-Ab), that blocks the Wnt signaling inhibitor (sclerostin), has been shown to promote bone mass in animal models and clinical studies. However, the cellular mechanisms by which Wnt signaling promotes osteogenesis remain to be further investigated. O-GlcNAcylation, a dynamic post-translational modification of proteins, controls multiple critical biological processes including transcription, translation, and cell fate determination. Here, we report that Wnt3a either induces O-GlcNAcylation rapidly via the Ca2+-PKA-Gfat1 axis, or increases it in a Wnt-β-catenin-dependent manner following prolonged stimulation. Importantly, we find O-GlcNAcylation indispensable for osteoblastogenesis both in vivo and in vitro. Genetic ablation of O-GlcNAcylation in the osteoblast-lineage diminishes bone formation and delays bone fracture healing in response to Wnt stimulation in vivo. Mechanistically, Wnt3a induces O-GlcNAcylation at Serine 174 of PDK1 to stabilize the protein, resulting in increased glycolysis and osteogenesis. These findings highlight O-GlcNAcylation as an important mechanism regulating Wnt-induced glucose metabolism and bone anabolism.
Collapse
Affiliation(s)
- Chengjia You
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Fangyuan Shen
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Puying Yang
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Jingyao Cui
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
- Department of Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Qiaoyue Ren
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Moyu Liu
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Yujie Hu
- Department of Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Boer Li
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
- Department of Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Ling Ye
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China.
- Department of Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China.
| | - Yu Shi
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China.
| |
Collapse
|
6
|
Su H, Huang L, Zhou J, Yang G. Prostate cancer stem cells and their targeted therapies. Front Cell Dev Biol 2024; 12:1410102. [PMID: 39175878 PMCID: PMC11338935 DOI: 10.3389/fcell.2024.1410102] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2024] [Accepted: 07/23/2024] [Indexed: 08/24/2024] Open
Abstract
Prostate cancer (PCa) is the most common malignancy among men worldwide. Through androgen receptor signaling inhibitor (ARSI) treatment, patients eventually succumb to castration-resistant prostate cancer (CRPC). For this, the prostate cancer stem cells (PCSCs), as a minor population of tumor cells that can promote tumor relapse, ARSI resistance, and disease progression, are gaining attention. Therefore, specific therapy targeting PCSCs has momentum. This study reviewed the identification and characterization of PCSCs and PCSC-based putative biomarkers and summarized their mechanisms of action. We further discussed clinical trials of novel therapeutic interventions focused on PCSC-related pathways, the PCSC microenvironment, cutting-edge miRNA therapy, and immunotherapy approaches from a mechanistic standpoint. This review provides updated insights into PCSC plasticity, identifying new PCSC biomarkers and optimized treatments for patients with advanced PCa.
Collapse
Affiliation(s)
- Huilan Su
- Research Center for Translational Medicine, Cancer Stem Cell Institute, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Liqun Huang
- Department of Urology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Jianjun Zhou
- Research Center for Translational Medicine, Cancer Stem Cell Institute, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Guosheng Yang
- Department of Urology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| |
Collapse
|
7
|
Hu J, Xu H, Ma X, Bai M, Zhou Y, Miao R, Wang F, Li X, Cheng B. Modulating PCGF4/BMI1 Stability Is an Efficient Metastasis-Regulatory Strategy Used by Distinct Subtypes of Cancer-Associated Fibroblasts in Intrahepatic Cholangiocarcinoma. THE AMERICAN JOURNAL OF PATHOLOGY 2024; 194:1388-1404. [PMID: 38670529 DOI: 10.1016/j.ajpath.2024.03.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/16/2023] [Revised: 02/17/2024] [Accepted: 03/21/2024] [Indexed: 04/28/2024]
Abstract
Intrahepatic cholangiocarcinoma (ICC) is a highly malignant neoplasm prone to metastasis. Whether cancer-associated fibroblasts (CAFs) affect the metastasis of ICC is unclear. Herein, ICC patient-derived CAF lines and related cancerous cell lines were established and the effects of CAFs on the tumor progressive properties of the ICC cancerous cells were analyzed. CAFs could be classified into cancer-restraining or cancer-promoting categories based on distinct tumorigenic effects. The RNA-sequencing analyses of ICC cancerous cell lines identified polycomb group ring finger 4 (PCGF4; alias BMI1) as a potential metastasis regulator. The changes of PCGF4 levels in ICC cells mirrored the restraining or promoting effects of CAFs on ICC migration. Immunohistochemical analyses on the ICC tissue microarrays indicated that PCGF4 was negatively correlated with overall survival of ICC. The promoting effects of PCGF4 on cell migration, drug resistance activity, and stemness properties were confirmed. Mechanistically, cancer-restraining CAFs triggered the proteasome-dependent degradation of PCGF4, whereas cancer-promoting CAFs enhanced the stability of PCGF4 via activating the IL-6/phosphorylated STAT3 pathway. In summary, the current data identified the role of CAFs in ICC metastasis and revealed a new mechanism of the CAFs on ICC progression in which PCGF4 acted as the key effector by both categories of CAFs. These findings shed light on developing comprehensive therapeutic strategies for ICC.
Collapse
Affiliation(s)
- Jinjing Hu
- School of Life Sciences, Lanzhou University, Lanzhou, China; Key Laboratory Biotherapy and Regenerative Medicine of Gansu Province, Lanzhou, China
| | - Hao Xu
- The Fourth Department of General Surgery, The First Hospital of Lanzhou University, Lanzhou, China; The First School of Clinical Medicine, Lanzhou University, Lanzhou, China
| | - Xiaojun Ma
- School of Life Sciences, Lanzhou University, Lanzhou, China
| | - Mingzhen Bai
- The First School of Clinical Medicine, Lanzhou University, Lanzhou, China
| | - Yongqiang Zhou
- The First School of Clinical Medicine, Lanzhou University, Lanzhou, China
| | - Ruidong Miao
- School of Life Sciences, Lanzhou University, Lanzhou, China
| | - Fanghong Wang
- The Fourth Department of General Surgery, The First Hospital of Lanzhou University, Lanzhou, China; The First School of Clinical Medicine, Lanzhou University, Lanzhou, China
| | - Xun Li
- The First School of Clinical Medicine, Lanzhou University, Lanzhou, China; Department of General Surgery, The First Hospital of Lanzhou University, Lanzhou, China.
| | - Bo Cheng
- School of Life Sciences, Lanzhou University, Lanzhou, China; Key Laboratory of Cell Activities and Stress Adaptations, Ministry of Education, Lanzhou University, Lanzhou, China.
| |
Collapse
|
8
|
Cai Y, Chen M, Gong Y, Tang G, Shu Z, Chen J, Zhou H, He Y, Long Z, Gan Y. Androgen-repressed lncRNA LINC01126 drives castration-resistant prostate cancer by regulating the switch between O-GlcNAcylation and phosphorylation of androgen receptor. Clin Transl Med 2024; 14:e1531. [PMID: 38214432 PMCID: PMC10785194 DOI: 10.1002/ctm2.1531] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2023] [Revised: 12/16/2023] [Accepted: 12/23/2023] [Indexed: 01/13/2024] Open
Abstract
BACKGROUND Prostate cancer (PCa) initially shows satisfactory response to therapies targeting the androgen receptor (AR). However, progression to a castration-resistant stage indicates poor prognosis in PCa patients. AR signalling still plays a central role in most castration-resistant prostate cancers (CRPC). Therefore, unveiling the mechanisms of AR reactivation under androgen-deprived conditions is imperative to discover novel therapeutic targets for CRPC. METHODS Using an integrative analysis of the transcriptomics of three independent PCa cohorts and a published landscape of AR-regulated long non-coding RNA (lncRNA), lncRNA LINC01126 was selected as a candidate gene that could drive CRPC progression for further study. Quantitative reverse transcription polymerase chain reaction, in situ hybridisation (ISH) and fluorescent ISH were performed to detect LINC01126 in PCa tissues and cells. The functional role and mechanism of LINC01126 were further investigated using in vitro and in vivo gain and loss of function assays. RESULTS LINC01126, identified as an AR-repressed lncRNA, was significantly upregulated after AR-targeted therapies. In addition, we found that LINC01126 was upregulated in CRPC and was associated with poor prognosis. We also proved that LINC01126 stabilised AR protein and enhanced AR nuclear translocation and transactivation by promoting the transition from O-GlcNAcylation at threonine 80 to phosphorylation at serine 81 (S81) within the AR protein. Mechanism analysis revealed that LINC01126 facilitates the interaction of CDK9 with AR and impedes the binding of O-linked N-acetylglucosamine (O-GlcNAc) transferase to AR. Consequently, LINC01126 expression was sufficient to activate AR signalling without androgen. LINC01126 overexpression increased, whereas LINC01126 knockdown decreased castration resistance traits in PCa cells in vitro and in vivo. Furthermore, our data showed that LINC01126-targeting antisense oligonucleotides (ASO) substantially inhibited CRPC cells in vitro. CONCLUSIONS Our research expands the functions of AR-regulated lncRNA in sustaining androgen-independent AR activity and promoting CRPC progression and reveals that LINC01126 may be a new therapeutic target for PCa.
Collapse
Affiliation(s)
- Yi Cai
- Department of UrologyDisorders of Prostate Cancer Multidisciplinary TeamNational Clinical Research Center for Geriatric DisordersXiangya HospitalCentral South UniversityChangshaHunanP.R. China
| | - Minfeng Chen
- Department of UrologyDisorders of Prostate Cancer Multidisciplinary TeamNational Clinical Research Center for Geriatric DisordersXiangya HospitalCentral South UniversityChangshaHunanP.R. China
| | - Yuchen Gong
- Department of UrologyDisorders of Prostate Cancer Multidisciplinary TeamNational Clinical Research Center for Geriatric DisordersXiangya HospitalCentral South UniversityChangshaHunanP.R. China
| | - Guyu Tang
- Department of UrologyDisorders of Prostate Cancer Multidisciplinary TeamNational Clinical Research Center for Geriatric DisordersXiangya HospitalCentral South UniversityChangshaHunanP.R. China
| | - Zhiwei Shu
- Department of UrologyDisorders of Prostate Cancer Multidisciplinary TeamNational Clinical Research Center for Geriatric DisordersXiangya HospitalCentral South UniversityChangshaHunanP.R. China
| | - Jiaxian Chen
- Department of UrologyDisorders of Prostate Cancer Multidisciplinary TeamNational Clinical Research Center for Geriatric DisordersXiangya HospitalCentral South UniversityChangshaHunanP.R. China
| | - Hengfeng Zhou
- Andrology CenterDepartment of UrologyThe Third Xiangya HospitalCentral South UniversityChangshaHunanP.R. China
| | - Yao He
- Department of UrologyDisorders of Prostate Cancer Multidisciplinary TeamNational Clinical Research Center for Geriatric DisordersXiangya HospitalCentral South UniversityChangshaHunanP.R. China
| | - Zhi Long
- Andrology CenterDepartment of UrologyThe Third Xiangya HospitalCentral South UniversityChangshaHunanP.R. China
| | - Yu Gan
- Department of UrologyDisorders of Prostate Cancer Multidisciplinary TeamNational Clinical Research Center for Geriatric DisordersXiangya HospitalCentral South UniversityChangshaHunanP.R. China
| |
Collapse
|
9
|
Kohyanagi N, Kitamura N, Ikeda S, Shibutani S, Sato K, Ohama T. PP2A inhibitor SET promotes mTORC1 and Bmi1 signaling through Akt activation and maintains the colony-formation ability of cancer cells. J Biol Chem 2024; 300:105584. [PMID: 38141761 PMCID: PMC10826185 DOI: 10.1016/j.jbc.2023.105584] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2023] [Revised: 11/19/2023] [Accepted: 12/07/2023] [Indexed: 12/25/2023] Open
Abstract
Protein phosphatase 2A (PP2A) is an essential tumor suppressor, with its activity often hindered in cancer cells by endogenous PP2A inhibitory proteins like SE translocation (SET). SET/PP2A axis plays a pivotal role in the colony-formation ability of cancer cells and the stabilization of c-Myc and E2F1 proteins implicated in this process. However, in osteosarcoma cell line HOS, SET knock-down (KD) suppresses the colony-formation ability without affecting c-Myc and E2F1. This study aimed to unravel the molecular mechanism through which SET enhances the colony-formation ability of HOS cells and determine if it is generalized to other cancer cells. Transcriptome analysis unveiled that SET KD suppressed mTORC1 signaling. SET KD inhibited Akt phosphorylation, an upstream kinase for mTORC1. PP2A inhibitor blocked SET KD-mediated decrease in phosphorylation of Akt and a mTORC1 substrate p70S6K. A constitutively active Akt restored decreased colony-formation ability by SET KD, indicating the SET/PP2A/Akt/mTORC1 axis. Additionally, enrichment analysis highlighted that Bmi-1, a polycomb group protein, is affected by SET KD. SET KD decreased Bmi-1 protein by Akt inhibition but not by mTORC1 inhibition, and exogenous Bmi-1 expression rescued the reduced colony formation by SET KD. Four out of eight cancer cell lines exhibited decreased Bmi-1 by SET KD. Further analysis of these cell lines revealed that Myc activity plays a role in SET KD-mediated Bmi-1 degradation. These findings provide new insights into the molecular mechanism of SET-regulated colony-formation ability, which involved Akt-mediated activation of mTORC1/p70S6K and Bmi-1 signaling.
Collapse
Affiliation(s)
- Naoki Kohyanagi
- Laboratory of Veterinary Pharmacology, Yamaguchi University Joint Graduate School of Veterinary Medicine, Yamaguchi, Japan
| | - Nao Kitamura
- Laboratory of Veterinary Pharmacology, Yamaguchi University Joint Graduate School of Veterinary Medicine, Yamaguchi, Japan
| | - Shunta Ikeda
- Laboratory of Veterinary Pharmacology, Yamaguchi University Joint Graduate School of Veterinary Medicine, Yamaguchi, Japan
| | - Shusaku Shibutani
- Laboratory of Veterinary Hygiene, Yamaguchi University Joint Graduate School of Veterinary Medicine, Yamaguchi, Japan
| | - Koichi Sato
- Laboratory of Veterinary Pharmacology, Yamaguchi University Joint Graduate School of Veterinary Medicine, Yamaguchi, Japan
| | - Takashi Ohama
- Laboratory of Veterinary Pharmacology, Yamaguchi University Joint Graduate School of Veterinary Medicine, Yamaguchi, Japan.
| |
Collapse
|
10
|
Dupas T, Lauzier B, McGraw S. O-GlcNAcylation: the sweet side of epigenetics. Epigenetics Chromatin 2023; 16:49. [PMID: 38093337 PMCID: PMC10720106 DOI: 10.1186/s13072-023-00523-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2023] [Accepted: 11/24/2023] [Indexed: 12/17/2023] Open
Abstract
Histones display a wide variety of post-translational modifications, including acetylation, methylation, and phosphorylation. These epigenetic modifications can influence chromatin structure and function without altering the DNA sequence. Histones can also undergo post-translational O-GlcNAcylation, a rather understudied modification that plays critical roles in almost all biological processes and is added and removed by O-linked N-acetylglucosamine transferase and O-GlcNAcase, respectively. This review provides a current overview of our knowledge of how O-GlcNAcylation impacts the histone code both directly and by regulating other chromatin modifying enzymes. This highlights the pivotal emerging role of O-GlcNAcylation as an essential epigenetic marker.
Collapse
Affiliation(s)
- Thomas Dupas
- Centre Hospitalier Universitaire Sainte-Justine Research Center, Montréal, Canada.
- Department of Obstetrics and Gynecology, Université de Montréal, 2900 Boulevard Edouard‑Montpetit, Montréal, QC, H3T 1J4, Canada.
| | - Benjamin Lauzier
- Centre Hospitalier Universitaire Sainte-Justine Research Center, Montréal, Canada
- Nantes Université, CNRS, INSERM, L'institut du Thorax, 44000, Nantes, France
| | - Serge McGraw
- Centre Hospitalier Universitaire Sainte-Justine Research Center, Montréal, Canada.
- Department of Obstetrics and Gynecology, Université de Montréal, 2900 Boulevard Edouard‑Montpetit, Montréal, QC, H3T 1J4, Canada.
| |
Collapse
|
11
|
Li H, Wang Y, Feng S, Chang K, Yu X, Yang F, Huang H, Wang Y, Li X, Guan F. Reciprocal regulation of TWIST1 and OGT determines the decitabine efficacy in MDS/AML. Cell Commun Signal 2023; 21:255. [PMID: 37736724 PMCID: PMC10514931 DOI: 10.1186/s12964-023-01278-y] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Accepted: 08/14/2023] [Indexed: 09/23/2023] Open
Abstract
Chemoresistance poses a significant impediment to effective treatment strategies for myelodysplastic syndromes (MDS) and acute myeloid leukemia (AML). Our previous study unveiled that oncogene TWIST1 interacted with DNA methyltransferase 3a (DNMT3a) to regulate the decitabine (DAC) resistance in MDS/AML. However, the underlying mechanism of TWIST1 dysregulation in DAC resistance remained enigmatic. Here, we found that O-GlcNAc modification was upregulated in CD34+ cells from MDS/AML patients who do not respond to DAC treatment. Functional study revealed that O-GlcNAcylation could stabilize TWIST1 by impeding its interaction with ubiquitin E3 ligase CBLC. In addition, as one typical transcription factor, TWIST1 could bind to the promoter of O-GlcNAc transferase (OGT) gene and activate its transcription. Collectively, we highlighted the crucial role of the O-GlcNAcylated TWIST1 in the chemoresistance capacity of MDS/AML clonal cells, which may pave the way for the development of a new therapeutic strategy targeting O-GlcNAcylated proteins and reducing the ratio of MDS/AML relapse. Video Abstract.
Collapse
Affiliation(s)
- Hongjiao Li
- Key Laboratory of Resource Biology and Biotechnology Western China, Ministry of Education; Provincial Key Laboratory of Biotechnology, College of Life Sciences, Northwest University, Xi'an, 710069, China
| | - Yi Wang
- Department of Hematology, Provincial People's Hospital, Xi'an, 710068, China
| | - Shuang Feng
- Key Laboratory of Resource Biology and Biotechnology Western China, Ministry of Education; Provincial Key Laboratory of Biotechnology, College of Life Sciences, Northwest University, Xi'an, 710069, China
| | - Kaijing Chang
- Key Laboratory of Resource Biology and Biotechnology Western China, Ministry of Education; Provincial Key Laboratory of Biotechnology, College of Life Sciences, Northwest University, Xi'an, 710069, China
| | - Xinwen Yu
- Key Laboratory of Resource Biology and Biotechnology Western China, Ministry of Education; Provincial Key Laboratory of Biotechnology, College of Life Sciences, Northwest University, Xi'an, 710069, China
| | - Fenfang Yang
- Key Laboratory of Resource Biology and Biotechnology Western China, Ministry of Education; Provincial Key Laboratory of Biotechnology, College of Life Sciences, Northwest University, Xi'an, 710069, China
| | - Haozhe Huang
- Key Laboratory of Resource Biology and Biotechnology Western China, Ministry of Education; Provincial Key Laboratory of Biotechnology, College of Life Sciences, Northwest University, Xi'an, 710069, China
| | - Yuanbo Wang
- Key Laboratory of Resource Biology and Biotechnology Western China, Ministry of Education; Provincial Key Laboratory of Biotechnology, College of Life Sciences, Northwest University, Xi'an, 710069, China
| | - Xiang Li
- Institute of Hematology, School of Medicine, Northwest University, Xi'an, 710069, China.
- College of Life Science, Northwest University, 229 Taibai North Road, Xi'an, 710069, Shaanxi, China.
| | - Feng Guan
- Key Laboratory of Resource Biology and Biotechnology Western China, Ministry of Education; Provincial Key Laboratory of Biotechnology, College of Life Sciences, Northwest University, Xi'an, 710069, China.
- College of Life Science, Northwest University, 229 Taibai North Road, Xi'an, 710069, Shaanxi, China.
| |
Collapse
|
12
|
He XF, Hu X, Wen GJ, Wang Z, Lin WJ. O-GlcNAcylation in cancer development and immunotherapy. Cancer Lett 2023; 566:216258. [PMID: 37279852 DOI: 10.1016/j.canlet.2023.216258] [Citation(s) in RCA: 36] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Revised: 05/03/2023] [Accepted: 05/30/2023] [Indexed: 06/08/2023]
Abstract
O-linked β-D-N-acetylglucosamine (O-GlcNAc), as a posttranslational modification (PTM), is a reversible reaction that attaches β-N-GlcNAc to Ser/Thr residues on specific proteins by O-GlcNAc transferase (OGT). O-GlcNAcase (OGA) removes the O-GlcNAc from O-GlcNAcylated proteins. O-GlcNAcylation regulates numerous cellular processes, including signal transduction, the cell cycle, metabolism, and energy homeostasis. Dysregulation of O-GlcNAcylation contributes to the development of various diseases, including cancers. Accumulating evidence has revealed that higher expression levels of OGT and hyper-O-GlcNAcylation are detected in many cancer types and governs glucose metabolism, proliferation, metastasis, invasion, angiogenesis, migration and drug resistance. In this review, we describe the biological functions and molecular mechanisms of OGT- or O-GlcNAcylation-mediated tumorigenesis. Moreover, we discuss the potential role of O-GlcNAcylation in tumor immunotherapy. Furthermore, we highlight that compounds can target O-GlcNAcylation by regulating OGT to suppress oncogenesis. Taken together, targeting protein O-GlcNAcylation might be a promising strategy for the treatment of human malignancies.
Collapse
Affiliation(s)
- Xue-Fen He
- Department of Obstetrics and Gynecology, Wenzhou Third Clinical Institute Affiliated to Wenzhou Medical University, Wenzhou People's Hospital, Wenzhou, 325000, Zhejiang, China
| | - Xiaoli Hu
- Department of Gynecology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Gao-Jing Wen
- Department of Obstetrics and Gynecology, Wenzhou Third Clinical Institute Affiliated to Wenzhou Medical University, Wenzhou People's Hospital, Wenzhou, 325000, Zhejiang, China
| | - Zhiwei Wang
- Department of Biochemistry and Molecular Biology, School of Laboratory Medicine, Bengbu Medical College, Anhui, China; Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China.
| | - Wen-Jing Lin
- Department of Obstetrics and Gynecology, Wenzhou Third Clinical Institute Affiliated to Wenzhou Medical University, Wenzhou People's Hospital, Wenzhou, 325000, Zhejiang, China.
| |
Collapse
|
13
|
Zhou Y, Li T, Jia M, Dai R, Wang R. The Molecular Biology of Prostate Cancer Stem Cells: From the Past to the Future. Int J Mol Sci 2023; 24:ijms24087482. [PMID: 37108647 PMCID: PMC10140972 DOI: 10.3390/ijms24087482] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2023] [Revised: 04/03/2023] [Accepted: 04/11/2023] [Indexed: 04/29/2023] Open
Abstract
Prostate cancer (PCa) continues to rank as the second leading cause of cancer-related mortality in western countries, despite the golden treatment using androgen deprivation therapy (ADT) or anti-androgen therapy. With decades of research, scientists have gradually realized that the existence of prostate cancer stem cells (PCSCs) successfully explains tumor recurrence, metastasis and therapeutic failure of PCa. Theoretically, eradication of this small population may improve the efficacy of current therapeutic approaches and prolong PCa survival. However, several characteristics of PCSCs make their diminishment extremely challenging: inherent resistance to anti-androgen and chemotherapy treatment, over-activation of the survival pathway, adaptation to tumor micro-environments, escape from immune attack and being easier to metastasize. For this end, a better understanding of PCSC biology at the molecular level will definitely inspire us to develop PCSC targeted approaches. In this review, we comprehensively summarize signaling pathways responsible for homeostatic regulation of PCSCs and discuss how to eliminate these fractional cells in clinical practice. Overall, this study deeply pinpoints PCSC biology at the molecular level and provides us some research perspectives.
Collapse
Affiliation(s)
- Yong Zhou
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Southwest Medical University, Luzhou 646000, China
| | - Tian Li
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Southwest Medical University, Luzhou 646000, China
| | - Man Jia
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Southwest Medical University, Luzhou 646000, China
| | - Rongyang Dai
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Southwest Medical University, Luzhou 646000, China
| | - Ronghao Wang
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Southwest Medical University, Luzhou 646000, China
| |
Collapse
|
14
|
Harvey DJ. Analysis of carbohydrates and glycoconjugates by matrix-assisted laser desorption/ionization mass spectrometry: An update for 2017-2018. MASS SPECTROMETRY REVIEWS 2023; 42:227-431. [PMID: 34719822 DOI: 10.1002/mas.21721] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/14/2021] [Revised: 07/26/2021] [Accepted: 07/26/2021] [Indexed: 06/13/2023]
Abstract
This review is the tenth update of the original article published in 1999 on the application of matrix-assisted laser desorption/ionization mass spectrometry (MALDI) mass spectrometry to the analysis of carbohydrates and glycoconjugates and brings coverage of the literature to the end of 2018. Also included are papers that describe methods appropriate to glycan and glycoprotein analysis by MALDI, such as sample preparation techniques, even though the ionization method is not MALDI. Topics covered in the first part of the review include general aspects such as theory of the MALDI process, new methods, matrices, derivatization, MALDI imaging, fragmentation and the use of arrays. The second part of the review is devoted to applications to various structural types such as oligo- and poly-saccharides, glycoproteins, glycolipids, glycosides, and biopharmaceuticals. Most of the applications are presented in tabular form. The third part of the review covers medical and industrial applications of the technique, studies of enzyme reactions, and applications to chemical synthesis. The reported work shows increasing use of combined new techniques such as ion mobility and highlights the impact that MALDI imaging is having across a range of diciplines. MALDI is still an ideal technique for carbohydrate analysis and advancements in the technique and the range of applications continue steady progress.
Collapse
Affiliation(s)
- David J Harvey
- Nuffield Department of Medicine, Target Discovery Institute, University of Oxford, Oxford, UK
| |
Collapse
|
15
|
Lu Q, Zhang X, Liang T, Bai X. O-GlcNAcylation: an important post-translational modification and a potential therapeutic target for cancer therapy. Mol Med 2022; 28:115. [PMID: 36104770 PMCID: PMC9476278 DOI: 10.1186/s10020-022-00544-y] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2022] [Accepted: 09/07/2022] [Indexed: 02/07/2023] Open
Abstract
O-linked β-d-N-acetylglucosamine (O-GlcNAc) is an important post-translational modification of serine or threonine residues on thousands of proteins in the nucleus and cytoplasm of all animals and plants. In eukaryotes, only two conserved enzymes are involved in this process. O-GlcNAc transferase is responsible for adding O-GlcNAc to proteins, while O-GlcNAcase is responsible for removing it. Aberrant O-GlcNAcylation is associated with a variety of human diseases, such as diabetes, cancer, neurodegenerative diseases, and cardiovascular diseases. Numerous studies have confirmed that O-GlcNAcylation is involved in the occurrence and progression of cancers in multiple systems throughout the body. It is also involved in regulating multiple cancer hallmarks, such as metabolic reprogramming, proliferation, invasion, metastasis, and angiogenesis. In this review, we first describe the process of O-GlcNAcylation and the structure and function of O-GlcNAc cycling enzymes. In addition, we detail the occurrence of O-GlcNAc in various cancers and the role it plays. Finally, we discuss the potential of O-GlcNAc as a promising biomarker and novel therapeutic target for cancer diagnosis, treatment, and prognosis.
Collapse
|
16
|
He X, Li Y, Chen Q, Zheng L, Lou J, Lin C, Gong J, Zhu Y, Wu Y. O-GlcNAcylation and stablization of SIRT7 promote pancreatic cancer progression by blocking the SIRT7-REGγ interaction. Cell Death Differ 2022; 29:1970-1981. [PMID: 35422493 PMCID: PMC9525610 DOI: 10.1038/s41418-022-00984-3] [Citation(s) in RCA: 41] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2021] [Revised: 03/13/2022] [Accepted: 03/14/2022] [Indexed: 01/05/2023] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is one of the most lethal cancers and its dismal prognosis indicates the urgent need to elucidate the potential oncogenic mechanisms. SIRT7 is a classic NAD+-dependent deacetylase that stabilizes the transformed state of cancer cells. However, its functional roles in PDAC are still unclear. Here, we found that SIRT7 expression is upregulated and predicts poor prognosis in PDAC. Then we screened the new interacting proteins of SIRT7 by mass spectrometry and the results showed that SIRT7 can interact with O-GlcNAc transferase (OGT). O-GlcNAcylation stabilizes the SIRT7 protein by inhibiting its interaction with REGγ to prevent degradation, and hyper-O-GlcNAcylation in pancreatic cancer cells leads to hypoacetylation of H3K18 via SIRT7, which promotes transcriptional repression of several tumour suppressor genes. In addition, SIRT7 O-GlcNAcylation at the serine 136 residue (S136) is required to maintain its protein stability and deacetylation ability. In vivo and in vitro experiments showed that blocking SIRT7 O-GlcNAcylation at S136 attenuates tumour progression. Collectively, we demonstrate that O-GlcNAcylation is an important post-translational modification of SIRT7 in pancreatic cancer cells, and elucidating this mechanism of SIRT7 is expected to pave the way for the development of novel therapeutic methods in the future.
Collapse
Affiliation(s)
- Xiaoman He
- Department of Hepato-Pancreato-Biliary Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, People's Republic of China
- Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education, Cancer Institute, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, People's Republic of China
| | - Yongzhou Li
- Department of Hepato-Pancreato-Biliary Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, People's Republic of China
- Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education, Cancer Institute, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, People's Republic of China
| | - Qing Chen
- Department of Hepato-Pancreato-Biliary Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, People's Republic of China
- Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education, Cancer Institute, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, People's Republic of China
| | - Lei Zheng
- The Affiliated Hospital of Qingdao University, Qingdao, People's Republic of China
| | - Jianyao Lou
- Department of Hepato-Pancreato-Biliary Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, People's Republic of China
- Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education, Cancer Institute, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, People's Republic of China
| | - Chuanshuai Lin
- Department of Hepato-Pancreato-Biliary Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, People's Republic of China
- Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education, Cancer Institute, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, People's Republic of China
| | - Jiali Gong
- Department of Hepato-Pancreato-Biliary Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, People's Republic of China
- Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education, Cancer Institute, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, People's Republic of China
| | - Yi Zhu
- Department of Hepato-Pancreato-Biliary Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, People's Republic of China.
- Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education, Cancer Institute, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, People's Republic of China.
| | - Yulian Wu
- Department of Hepato-Pancreato-Biliary Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, People's Republic of China.
- Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education, Cancer Institute, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, People's Republic of China.
| |
Collapse
|
17
|
Critical Roles of Polycomb Repressive Complexes in Transcription and Cancer. Int J Mol Sci 2022; 23:ijms23179574. [PMID: 36076977 PMCID: PMC9455514 DOI: 10.3390/ijms23179574] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2022] [Revised: 08/16/2022] [Accepted: 08/18/2022] [Indexed: 11/17/2022] Open
Abstract
Polycomp group (PcG) proteins are members of highly conserved multiprotein complexes, recognized as gene transcriptional repressors during development and shown to play a role in various physiological and pathological processes. PcG proteins consist of two Polycomb repressive complexes (PRCs) with different enzymatic activities: Polycomb repressive complexes 1 (PRC1), a ubiquitin ligase, and Polycomb repressive complexes 2 (PRC2), a histone methyltransferase. Traditionally, PRCs have been described to be associated with transcriptional repression of homeotic genes, as well as gene transcription activating effects. Particularly in cancer, PRCs have been found to misregulate gene expression, not only depending on the function of the whole PRCs, but also through their separate subunits. In this review, we focused especially on the recent findings in the transcriptional regulation of PRCs, the oncogenic and tumor-suppressive roles of PcG proteins, and the research progress of inhibitors targeting PRCs.
Collapse
|
18
|
Xu J, Li L, Shi P, Cui H, Yang L. The Crucial Roles of Bmi-1 in Cancer: Implications in Pathogenesis, Metastasis, Drug Resistance, and Targeted Therapies. Int J Mol Sci 2022; 23:ijms23158231. [PMID: 35897796 PMCID: PMC9367737 DOI: 10.3390/ijms23158231] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2022] [Revised: 07/22/2022] [Accepted: 07/23/2022] [Indexed: 12/01/2022] Open
Abstract
B-cell-specific Moloney murine leukemia virus integration region 1 (Bmi-1, also known as RNF51 or PCGF4) is one of the important members of the PcG gene family, and is involved in regulating cell proliferation, differentiation and senescence, and maintaining the self-renewal of stem cells. Many studies in recent years have emphasized the role of Bmi-1 in the occurrence and development of tumors. In fact, Bmi-1 has multiple functions in cancer biology and is closely related to many classical molecules, including Akt, c-MYC, Pten, etc. This review summarizes the regulatory mechanisms of Bmi-1 in multiple pathways, and the interaction of Bmi-1 with noncoding RNAs. In particular, we focus on the pathological processes of Bmi-1 in cancer, and explore the clinical relevance of Bmi-1 in cancer biomarkers and prognosis, as well as its implications for chemoresistance and radioresistance. In conclusion, we summarize the role of Bmi-1 in tumor progression, reveal the pathophysiological process and molecular mechanism of Bmi-1 in tumors, and provide useful information for tumor diagnosis, treatment, and prognosis.
Collapse
Affiliation(s)
- Jie Xu
- State Key Laboratory of Silkworm Genome Biology, Southwest University, Chongqing 400716, China; (J.X.); (L.L.); (P.S.)
- Cancer Center, Medical Research Institute, Southwest University, Chongqing 400716, China
| | - Lin Li
- State Key Laboratory of Silkworm Genome Biology, Southwest University, Chongqing 400716, China; (J.X.); (L.L.); (P.S.)
| | - Pengfei Shi
- State Key Laboratory of Silkworm Genome Biology, Southwest University, Chongqing 400716, China; (J.X.); (L.L.); (P.S.)
- Cancer Center, Medical Research Institute, Southwest University, Chongqing 400716, China
| | - Hongjuan Cui
- State Key Laboratory of Silkworm Genome Biology, Southwest University, Chongqing 400716, China; (J.X.); (L.L.); (P.S.)
- Cancer Center, Medical Research Institute, Southwest University, Chongqing 400716, China
- Correspondence: (H.C.); (L.Y.)
| | - Liqun Yang
- State Key Laboratory of Silkworm Genome Biology, Southwest University, Chongqing 400716, China; (J.X.); (L.L.); (P.S.)
- Cancer Center, Medical Research Institute, Southwest University, Chongqing 400716, China
- Correspondence: (H.C.); (L.Y.)
| |
Collapse
|
19
|
GIPC2 interacts with Fzd7 to promote prostate cancer metastasis by activating WNT signaling. Oncogene 2022; 41:2609-2623. [PMID: 35347223 PMCID: PMC9054671 DOI: 10.1038/s41388-022-02255-4] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2021] [Revised: 02/08/2022] [Accepted: 02/17/2022] [Indexed: 12/21/2022]
Abstract
Prostate cancer (PCa) causes significant mortality and morbidity, with advanced metastasis. WNT signaling is a promising therapeutic target for metastatic PCa. GIPC2 is a GIPC1 paralog involved in WNT signaling pathways associated with tumor progression, but its role in PCa metastasis remains unclear. Herein, we demonstrated that high GIPC2 expression in PCa tissues was significantly associated with distant metastasis and poor prognosis. Functional studies demonstrated that high GIPC2 expression due to CpG-island demethylation promoted increased metastatic capabilities of PCa cells. Conversely, silencing GIPC2 expression significantly inhibited PCa metastasis in vitro and in vivo. Furthermore, GIPC2 directly bound the WNT co-receptor Fzd7 through its PDZ domain, which enabled activation of WNT-β-catenin cascades, thereby stimulating PCa metastasis. Interestingly, GIPC2 protein was also identified as a component of exosomes and that it robustly stimulated PCa adhesion, invasion, and migration. The presence of GIPC2 in tumor-derived exosomes and ability to impact the behavior of tumor cells suggest that GIPC2 is a novel epigenetic oncogene involved in PCa metastasis. Our findings identified GIPC2 as a novel exosomal molecule associated with WNT signaling and may represent a potential therapeutic target and biomarker for metastatic PCa.
Collapse
|
20
|
Abstract
Post-translational modification with O-linked β-N-acetylglucosamine (O-GlcNAc), a process referred to as O-GlcNAcylation, occurs on a vast variety of proteins. Mounting evidence in the past several decades has clearly demonstrated that O-GlcNAcylation is a unique and ubiquitous modification. Reminiscent of a code, protein O-GlcNAcylation functions as a crucial regulator of nearly all cellular processes studied. The primary aim of this review is to summarize the developments in our understanding of myriad protein substrates modified by O-GlcNAcylation from a systems perspective. Specifically, we provide a comprehensive survey of O-GlcNAcylation in multiple species studied, including eukaryotes (e.g., protists, fungi, plants, Caenorhabditis elegans, Drosophila melanogaster, murine, and human), prokaryotes, and some viruses. We evaluate features (e.g., structural properties and sequence motifs) of O-GlcNAc modification on proteins across species. Given that O-GlcNAcylation functions in a species-, tissue-/cell-, protein-, and site-specific manner, we discuss the functional roles of O-GlcNAcylation on human proteins. We focus particularly on several classes of relatively well-characterized human proteins (including transcription factors, protein kinases, protein phosphatases, and E3 ubiquitin-ligases), with representative O-GlcNAc site-specific functions presented. We hope the systems view of the great endeavor in the past 35 years will help demystify the O-GlcNAc code and lead to more fascinating studies in the years to come.
Collapse
Affiliation(s)
- Junfeng Ma
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Georgetown University, Washington, DC 20057, United States
| | - Chunyan Hou
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Georgetown University, Washington, DC 20057, United States
| | - Ci Wu
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Georgetown University, Washington, DC 20057, United States
| |
Collapse
|
21
|
Parreno V, Martinez AM, Cavalli G. Mechanisms of Polycomb group protein function in cancer. Cell Res 2022; 32:231-253. [PMID: 35046519 PMCID: PMC8888700 DOI: 10.1038/s41422-021-00606-6] [Citation(s) in RCA: 73] [Impact Index Per Article: 24.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2021] [Accepted: 12/10/2021] [Indexed: 02/01/2023] Open
Abstract
Cancer arises from a multitude of disorders resulting in loss of differentiation and a stem cell-like phenotype characterized by uncontrolled growth. Polycomb Group (PcG) proteins are members of multiprotein complexes that are highly conserved throughout evolution. Historically, they have been described as essential for maintaining epigenetic cellular memory by locking homeotic genes in a transcriptionally repressed state. What was initially thought to be a function restricted to a few target genes, subsequently turned out to be of much broader relevance, since the main role of PcG complexes is to ensure a dynamically choregraphed spatio-temporal regulation of their numerous target genes during development. Their ability to modify chromatin landscapes and refine the expression of master genes controlling major switches in cellular decisions under physiological conditions is often misregulated in tumors. Surprisingly, their functional implication in the initiation and progression of cancer may be either dependent on Polycomb complexes, or specific for a subunit that acts independently of other PcG members. In this review, we describe how misregulated Polycomb proteins play a pleiotropic role in cancer by altering a broad spectrum of biological processes such as the proliferation-differentiation balance, metabolism and the immune response, all of which are crucial in tumor progression. We also illustrate how interfering with PcG functions can provide a powerful strategy to counter tumor progression.
Collapse
Affiliation(s)
- Victoria Parreno
- Institute of Human Genetics, UMR 9002, CNRS-University of Montpellier, Montpellier, France
| | - Anne-Marie Martinez
- Institute of Human Genetics, UMR 9002, CNRS-University of Montpellier, Montpellier, France.
| | - Giacomo Cavalli
- Institute of Human Genetics, UMR 9002, CNRS-University of Montpellier, Montpellier, France.
| |
Collapse
|
22
|
Spaner DE. O-GlcNAcylation in Chronic Lymphocytic Leukemia and Other Blood Cancers. Front Immunol 2021; 12:772304. [PMID: 34868034 PMCID: PMC8639227 DOI: 10.3389/fimmu.2021.772304] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Accepted: 11/02/2021] [Indexed: 12/17/2022] Open
Abstract
In the past decade, aberrant O-GlcNAcylation has emerged as a new hallmark of cancer. O-GlcNAcylation is a post-translational modification that results when the amino-sugar β-D-N-acetylglucosamine (GlcNAc) is made in the hexosamine biosynthesis pathway (HBP) and covalently attached to serine and threonine residues in intracellular proteins by the glycosyltransferase O-GlcNAc transferase (OGT). O-GlcNAc moieties reflect the metabolic state of a cell and are removed by O-GlcNAcase (OGA). O-GlcNAcylation affects signaling pathways and protein expression by cross-talk with kinases and proteasomes and changes gene expression by altering protein interactions, localization, and complex formation. The HBP and O-GlcNAcylation are also recognized to mediate survival of cells in harsh conditions. Consequently, O-GlcNAcylation can affect many of the cellular processes that are relevant for cancer and is generally thought to promote tumor growth, disease progression, and immune escape. However, recent studies suggest a more nuanced view with O-GlcNAcylation acting as a tumor promoter or suppressor depending on the stage of disease or the genetic abnormalities, proliferative status, and state of the p53 axis in the cancer cell. Clinically relevant HBP and OGA inhibitors are already available and OGT inhibitors are in development to modulate O-GlcNAcylation as a potentially novel cancer treatment. Here recent studies that implicate O-GlcNAcylation in oncogenic properties of blood cancers are reviewed, focusing on chronic lymphocytic leukemia and effects on signal transduction and stress resistance in the cancer microenvironment. Therapeutic strategies for targeting the HBP and O-GlcNAcylation are also discussed.
Collapse
Affiliation(s)
- David E Spaner
- Biology Platform, Sunnybrook Research Institute, Toronto, ON, Canada.,Department of Immunology, University of Toronto, Toronto, ON, Canada.,Department of Medical Biophysics, University of Toronto, Toronto, ON, Canada.,Department of Medical Oncology, Sunnybrook Odette Cancer Center, Toronto, ON, Canada.,Department of Medicine, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
23
|
Sun L, Lv S, Song T. O-GlcNAcylation links oncogenic signals and cancer epigenetics. Discov Oncol 2021; 12:54. [PMID: 35201498 PMCID: PMC8777512 DOI: 10.1007/s12672-021-00450-5] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/18/2021] [Accepted: 11/11/2021] [Indexed: 12/19/2022] Open
Abstract
Prevalent dysregulation of epigenetic modifications plays a pivotal role in cancer. Targeting epigenetic abnormality is a new strategy for cancer therapy. Understanding how conventional oncogenic factors cause epigenetic abnormality is of great basic and translational value. O-GlcNAcylation is a protein modification which affects physiology and pathophysiology. In mammals, O-GlcNAcylation is catalyzed by one single enzyme OGT and removed by one single enzyme OGA. O-GlcNAcylation is affected by the availability of the donor, UDP-GlcNAc, generated by the serial enzymatic reactions in the hexoamine biogenesis pathway (HBP). O-GlcNAcylation regulates a wide spectrum of substrates including many proteins involved in epigenetic modification. Like epigenetic modifications, abnormality of O-GlcNAcylation is also common in cancer. Studies have revealed substantial impact on HBP enzymes and OGT/OGA by oncogenic signals. In this review, we will first summarize how oncogenic signals regulate HBP enzymes, OGT and OGA in cancer. We will then integrate this knowledge with the up to date understanding how O-GlcNAcylation regulates epigenetic machinery. With this, we propose a signal axis from oncogenic signals through O-GlcNAcylation dysregulation to epigenetic abnormality in cancer. Further elucidation of this axis will not only advance our understanding of cancer biology but also provide new revenues towards cancer therapy.
Collapse
Affiliation(s)
- Lidong Sun
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, 13 Hangkong Road, Wuhan, 430030, China.
| | - Suli Lv
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, 13 Hangkong Road, Wuhan, 430030, China
| | - Tanjing Song
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, 13 Hangkong Road, Wuhan, 430030, China.
| |
Collapse
|
24
|
Zhang Z, Oh M, Sasaki JI, Nör JE. Inverse and reciprocal regulation of p53/p21 and Bmi-1 modulates vasculogenic differentiation of dental pulp stem cells. Cell Death Dis 2021; 12:644. [PMID: 34168122 PMCID: PMC8225874 DOI: 10.1038/s41419-021-03925-z] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Revised: 06/10/2021] [Accepted: 06/11/2021] [Indexed: 12/20/2022]
Abstract
Dental pulp stem cells (DPSC) are capable of differentiating into vascular endothelial cells. Although the capacity of vascular endothelial growth factor (VEGF) to induce endothelial differentiation of stem cells is well established, mechanisms that maintain stemness and prevent vasculogenic differentiation remain unclear. Here, we tested the hypothesis that p53 signaling through p21 and Bmi-1 maintains stemness and inhibits vasculogenic differentiation. To address this hypothesis, we used primary human DPSC from permanent teeth and Stem cells from Human Exfoliated Deciduous (SHED) teeth as models of postnatal mesenchymal stem cells. DPSC seeded in biodegradable scaffolds and transplanted into immunodeficient mice generated mature human blood vessels invested with smooth muscle actin-positive mural cells. Knockdown of p53 was sufficient to induce vasculogenic differentiation of DPSC (without vasculogenic differentiation medium containing VEGF), as shown by increased expression of endothelial markers (VEGFR2, Tie-2, CD31, VE-cadherin), increased capillary sprouting in vitro; and increased DPSC-derived blood vessel density in vivo. Conversely, induction of p53 expression with small molecule inhibitors of the p53-MDM2 binding (MI-773, APG-115) was sufficient to inhibit VEGF-induced vasculogenic differentiation. Considering that p21 is a major downstream effector of p53, we knocked down p21 in DPSC and observed an increase in capillary sprouting that mimicked results observed when p53 was knocked down. Stabilization of ubiquitin activity was sufficient to induce p53 and p21 expression and reduce capillary sprouting. Interestingly, we observed an inverse and reciprocal correlation between p53/p21 and the expression of Bmi-1, a major regulator of stem cell self-renewal. Further, direct inhibition of Bmi-1 with PTC-209 resulted in blockade of capillary-like sprout formation. Collectively, these data demonstrate that p53/p21 functions through Bmi-1 to prevent the vasculogenic differentiation of DPSC.
Collapse
Affiliation(s)
- Zhaocheng Zhang
- Angiogenesis Research Laboratory, Department of Cariology, Restorative Sciences and Endodontics, University of Michigan School of Dentistry, Ann Arbor, MI, 48109, USA
| | - Min Oh
- Angiogenesis Research Laboratory, Department of Cariology, Restorative Sciences and Endodontics, University of Michigan School of Dentistry, Ann Arbor, MI, 48109, USA
| | - Jun-Ichi Sasaki
- Angiogenesis Research Laboratory, Department of Cariology, Restorative Sciences and Endodontics, University of Michigan School of Dentistry, Ann Arbor, MI, 48109, USA
| | - Jacques E Nör
- Angiogenesis Research Laboratory, Department of Cariology, Restorative Sciences and Endodontics, University of Michigan School of Dentistry, Ann Arbor, MI, 48109, USA.
- Department of Biomedical Engineering, University of Michigan College of Engineering, Ann Arbor, MI, USA.
- Department of Otolaryngology, University of Michigan School of Medicine, Ann Arbor, MI, USA.
| |
Collapse
|
25
|
Alantolactone inhibits cervical cancer progression by downregulating BMI1. Sci Rep 2021; 11:9251. [PMID: 33927214 PMCID: PMC8085045 DOI: 10.1038/s41598-021-87781-z] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2020] [Accepted: 03/10/2021] [Indexed: 01/31/2023] Open
Abstract
Cervical cancer is the second most common cancer in women. Despite advances in cervical cancer therapy, tumor recurrence and metastasis remain the leading causes of mortality. High expression of BMI1 is significantly associated with poor tumor differentiation, high clinical grade, and poor prognosis of cervical cancer, and is an independent prognostic factor in cervical carcinoma. Alantolactone (AL), a sesquiterpene lactone, exhibits potent anti-inflammatory and anticancer activities. In this paper, we investigated the mechanism of AL in reducing the proliferation, migration, and invasion of HeLa and SiHa cervical cancer cells as well as its promotion of mitochondrial damage and autophagy. BMI1 silencing decreased epithelial-mesenchymal transformation-associated proteins and increased autophagy-associated proteins in HeLa cells. These effects were reversed by overexpression of BMI1 in HeLa cells. Thus, BMI1 expression is positively correlated with invasion and negatively correlated with autophagy in HeLa cells. Importantly, AL decreased the weight, volume, and BMI1 expression in HeLa xenograft tumors. Furthermore, the structure of BMI1 and target interaction of AL were virtually screened using the molecular docking program Autodock Vina; AL decreased the expression of N-cadherin, vimentin, and P62 and increased the expression of LC3B and Beclin-1 in xenograft tumors. Finally, expression of BMI1 increased the phosphorylation of STAT3, which is important for cell proliferation, survival, migration, and invasion. Therefore, we suggest that AL plays a pivotal role in inhibiting BMI1 in the tumorigenesis of cervical cancer and is a potential therapeutic agent for cervical cancer.
Collapse
|
26
|
Makwana V, Rudrawar S, Anoopkumar-Dukie S. Signalling transduction of O-GlcNAcylation and PI3K/AKT/mTOR-axis in prostate cancer. Biochim Biophys Acta Mol Basis Dis 2021; 1867:166129. [PMID: 33744394 DOI: 10.1016/j.bbadis.2021.166129] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2020] [Revised: 02/26/2021] [Accepted: 03/14/2021] [Indexed: 12/23/2022]
Abstract
Hexosamine biosynthetic (HBP) and PI3K/AKT/mTOR pathways are found to predominate the proliferation and survival of prostate cancer cells. Both these pathways have their own specific intermediates to propagate the secondary signals in down-stream cascades and besides having their own structured network, also have shared interconnecting branches. These interconnections are either competitive or co-operative in nature depending on the microenvironmental conditions. Specifically, in prostate cancer HBP and mTOR pathways increases the expression and protein level of androgen receptor in order to support cancer cell proliferation, advancement and metastasis. Pharmacological inhibition of a single pathway is therefore insufficient to stop disease progression as the cancer cells manage to alter the signalling channel. This is one of the primary reasons for the therapeutic failure in prostate cancer and emergence of chemoresistance. Inhibition of these multiple pathways at their common junctures might prove to be of benefit in men suffering from an advanced disease state. Hence, a thorough understanding of these cellular intersecting points and their significance with respect to signal transduction mechanisms might assist in the rational designing of combinations for effective management of prostate cancer.
Collapse
Affiliation(s)
- Vivek Makwana
- School of Pharmacy and Pharmacology, Griffith University, Gold Coast, QLD 4222, Australia
| | - Santosh Rudrawar
- School of Pharmacy and Pharmacology, Griffith University, Gold Coast, QLD 4222, Australia; Menzies Health Institute Queensland, Griffith University, Gold Coast, QLD 4222, Australia; Quality Use of Medicines Network, Griffith University, Gold Coast, QLD 4222, Australia.
| | - Shailendra Anoopkumar-Dukie
- School of Pharmacy and Pharmacology, Griffith University, Gold Coast, QLD 4222, Australia; Menzies Health Institute Queensland, Griffith University, Gold Coast, QLD 4222, Australia; Quality Use of Medicines Network, Griffith University, Gold Coast, QLD 4222, Australia.
| |
Collapse
|
27
|
Chen Y, Liu J, Zhang W, Kadier A, Wang R, Zhang H, Yao X. O-GlcNAcylation Enhances NUSAP1 Stability and Promotes Bladder Cancer Aggressiveness. Onco Targets Ther 2021; 14:445-454. [PMID: 33488099 PMCID: PMC7815093 DOI: 10.2147/ott.s258175] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2020] [Accepted: 12/24/2020] [Indexed: 11/23/2022] Open
Abstract
Objective NUSAPl and O-GlcNAcylation were reported to be hyper-activated in many kinds of cancers and involved in the advanced progression of cancers. In bladder cancer, O-GlcNAc transferase (OGT) expresses in patients' urine samples, with no expression in healthy individuals, indicating O-GlcNAcylation might involve in the occurrence and development of bladder cancer. Therefore, the present study aims to investigate the effects of O-GlcNAcylation in bladder cancer and if it can regulate NUSAP1 protein. Materials and Methods Western blot, immunohistochemistry, and PCR were used to evaluate the protein expression and mRNA level of NUSAP1; CCK-8 and flow cytometry used to evaluate the proliferation and inhibited the apoptosis of bladder cancer. Results The results showed that NUSAP1 was highly expressed in bladder cancer cells and tissue samples. NUSAP1 up-regulation significantly promoted the proliferation and inhibited the apoptosis of bladder cancer HT-1376 and T24 cells. Besides, the expression of O-GlcNAc was elevated in bladder cancer tissues and cells, and up-regulation of O-GlcNAc with GlcNAc and PuGNAc obviously increased NUSAP1 protein expression and stability. Moreover, knockdown OGT significantly inhibited the proliferation and tumorigenesis and promoted the apoptosis of bladder cancer cells, confirmed by CCK-8, in vivo xenotransplantation, and flow cytometry, whereas these roles were impaired when NUSAP1 was up-regulated. Conclusion Overall, our study makes clear that hyper-O-GlcNAcylation accelerates bladder cancer progression through promotion of NUSAP1 expression and its stability.
Collapse
Affiliation(s)
- Yifan Chen
- Department of Urology, Shanghai Tenth People's Hospital, Tongji University of Medicine, Shanghai, 200072, People's Republic of China
| | - Ji Liu
- Department of Urology, Shanghai Tenth People's Hospital, Tongji University of Medicine, Shanghai, 200072, People's Republic of China
| | - Wentao Zhang
- Department of Urology, Shanghai Tenth People's Hospital, Tongji University of Medicine, Shanghai, 200072, People's Republic of China
| | - Aimaitiaji Kadier
- Department of Urology, Shanghai Tenth People's Hospital, Tongji University of Medicine, Shanghai, 200072, People's Republic of China
| | - Ruiliang Wang
- Department of Urology, Shanghai Tenth People's Hospital, Tongji University of Medicine, Shanghai, 200072, People's Republic of China
| | - Haimin Zhang
- Department of Urology, Shanghai Tenth People's Hospital, Tongji University of Medicine, Shanghai, 200072, People's Republic of China
| | - Xudong Yao
- Department of Urology, Shanghai Tenth People's Hospital, Tongji University of Medicine, Shanghai, 200072, People's Republic of China
| |
Collapse
|
28
|
O-GlcNAc stabilizes SMAD4 by inhibiting GSK-3β-mediated proteasomal degradation. Sci Rep 2020; 10:19908. [PMID: 33199824 PMCID: PMC7670456 DOI: 10.1038/s41598-020-76862-0] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2020] [Accepted: 10/28/2020] [Indexed: 12/29/2022] Open
Abstract
O-linked β-N-acetylglucosamine (O-GlcNAc) is a post-translational modification which occurs on the hydroxyl group of serine or threonine residues of nucleocytoplasmic proteins. It has been reported that the presence of this single sugar motif regulates various biological events by altering the fate of target proteins, such as their function, localization, and degradation. This study identified SMAD4 as a novel O-GlcNAc-modified protein. SMAD4 is a component of the SMAD transcriptional complex, a major regulator of the signaling pathway for the transforming growth factor-β (TGF-β). TGF-β is a powerful promoter of cancer EMT and metastasis. This study showed that the amount of SMAD4 proteins changes according to cellular O-GlcNAc levels in human lung cancer cells. This observation was made based on the prolonged half-life of SMAD4 proteins. The mechanism behind this interaction was that O-GlcNAc impeded interactions between SMAD4 and GSK-3β which promote proteasomal degradation of SMAD4. In addition, O-GlcNAc modification on SMAD4 Thr63 was responsible for stabilization. As a result, defects in O-GlcNAcylation on SMAD4 Thr63 attenuated the reporter activity of luciferase, the TGF-β-responsive SMAD binding element (SBE). This study’s findings imply that cellular O-GlcNAc may regulate the TGF-β/SMAD signaling pathway by stabilizing SMAD4.
Collapse
|
29
|
Sarfraz M, Afzal A, Khattak S, Saddozai UAK, Li HM, Zhang QQ, Madni A, Haleem KS, Duan SF, Wu DD, Ji SP, Ji XY. Multifaceted behavior of PEST sequence enriched nuclear proteins in cancer biology and role in gene therapy. J Cell Physiol 2020; 236:1658-1676. [PMID: 32841373 DOI: 10.1002/jcp.30011] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2020] [Revised: 07/18/2020] [Accepted: 08/04/2020] [Indexed: 01/12/2023]
Abstract
The amino acid sequence enriched with proline (P), glutamic acid (E), serine (S), and threonine (T) (PEST) is a signal-transducing agent providing unique features to its substrate nuclear proteins (PEST-NPs). The PEST motif is responsible for particular posttranslational modifications (PTMs). These PTMs impart distinct properties to PEST-NPs that are responsible for their activation/inhibition, intracellular localization, and stability/degradation. PEST-NPs participate in cancer metabolism, immunity, and protein transcription as oncogenes or as tumor suppressors. Gene-based therapeutics are getting the attention of researchers because of their cell specificity. PEST-NPs are good targets to explore as cancer therapeutics. Insights into PTMs of PEST-NPs demonstrate that these proteins not only interact with each other but also recruit other proteins to/from their active site to promote/inhibit tumors. Thus, the role of PEST-NPs in cancer biology is multivariate. It is hard to obtain therapeutic objectives with single gene therapy. An especially designed combination gene therapy might be a promising strategy in cancer treatment. This review highlights the multifaceted behavior of PEST-NPs in cancer biology. We have summarized a number of studies to address the influence of structure and PEST-mediated PTMs on activation, localization, stability, and protein-protein interactions of PEST-NPs. We also recommend researchers to adopt a pragmatic approach in gene-based cancer therapy.
Collapse
Affiliation(s)
- Muhammad Sarfraz
- Henan International Joint Laboratory for Nuclear Protein Regulation & Kaifeng Key Laboratory of Infectious Diseases and Bio-safety, School of Basic Medical Sciences, Henan University College of Medicine, Kaifeng, Henan, China.,Faculty of Pharmacy, The University of Lahore, Lahore, Punjab, Pakistan.,Kaifeng Municipal Key Laboratory of Cell Signal Transduction, Henan Provincial Engineering Centre for Tumor Molecular Medicine, Henan University, Kaifeng, Henan, China
| | - Attia Afzal
- Henan International Joint Laboratory for Nuclear Protein Regulation & Kaifeng Key Laboratory of Infectious Diseases and Bio-safety, School of Basic Medical Sciences, Henan University College of Medicine, Kaifeng, Henan, China.,Faculty of Pharmacy, The University of Lahore, Lahore, Punjab, Pakistan
| | - Saadullah Khattak
- Henan International Joint Laboratory for Nuclear Protein Regulation & Kaifeng Key Laboratory of Infectious Diseases and Bio-safety, School of Basic Medical Sciences, Henan University College of Medicine, Kaifeng, Henan, China
| | - Umair A K Saddozai
- Henan International Joint Laboratory for Nuclear Protein Regulation & Kaifeng Key Laboratory of Infectious Diseases and Bio-safety, School of Basic Medical Sciences, Henan University College of Medicine, Kaifeng, Henan, China
| | - Hui-Min Li
- Henan International Joint Laboratory for Nuclear Protein Regulation & Kaifeng Key Laboratory of Infectious Diseases and Bio-safety, School of Basic Medical Sciences, Henan University College of Medicine, Kaifeng, Henan, China.,Department of Histology and Embryology, Cell Signal Transduction Laboratory, School of Basic Medical Sciences, Bioinformatics Centre, Institute of Biomedical Informatics, Henan University, Kaifeng, Henan, China
| | - Qian-Qian Zhang
- Henan International Joint Laboratory for Nuclear Protein Regulation & Kaifeng Key Laboratory of Infectious Diseases and Bio-safety, School of Basic Medical Sciences, Henan University College of Medicine, Kaifeng, Henan, China
| | - Asadullah Madni
- Faculty of Pharmacy and Alternative Medicine, The Islamia University of Bahawalpur, Bahawalpur, Punjab, Pakistan
| | - Kashif S Haleem
- Department of Microbiology, Hazara University, Mansehra, Pakistan
| | - Shao-Feng Duan
- Henan International Joint Laboratory for Nuclear Protein Regulation & Kaifeng Key Laboratory of Infectious Diseases and Bio-safety, School of Basic Medical Sciences, Henan University College of Medicine, Kaifeng, Henan, China.,School of Pharmacy, Institute for Innovative Drug Design and Evaluation, Henan University, Kaifeng, Henan, China
| | - Dong-Dong Wu
- Henan International Joint Laboratory for Nuclear Protein Regulation & Kaifeng Key Laboratory of Infectious Diseases and Bio-safety, School of Basic Medical Sciences, Henan University College of Medicine, Kaifeng, Henan, China.,School of Stomatology, Henan University, Kaifeng, Henan, China
| | - Shao-Ping Ji
- Kaifeng Municipal Key Laboratory of Cell Signal Transduction, Henan Provincial Engineering Centre for Tumor Molecular Medicine, Henan University, Kaifeng, Henan, China
| | - Xin-Ying Ji
- Henan International Joint Laboratory for Nuclear Protein Regulation & Kaifeng Key Laboratory of Infectious Diseases and Bio-safety, School of Basic Medical Sciences, Henan University College of Medicine, Kaifeng, Henan, China
| |
Collapse
|
30
|
Li Y, Tian Z, Tan Y, Lian G, Chen S, Chen S, Li J, Li X, Huang K, Chen Y. Bmi-1-induced miR-27a and miR-155 promote tumor metastasis and chemoresistance by targeting RKIP in gastric cancer. Mol Cancer 2020; 19:109. [PMID: 32580736 PMCID: PMC7315508 DOI: 10.1186/s12943-020-01229-y] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2020] [Accepted: 06/17/2020] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND We previously reported an inverse relationship between B cell-specific Moloney murine leukemia virus integration site 1 (Bmi-1) and Raf kinase inhibitory protein (RKIP), which is associated with the prognosis of gastric cancer (GC). In this study, we further explored the microRNA (miRNA) regulatory mechanism between Bmi-1 and RKIP. METHODS Microarray analysis was first carried out to identify miRNA profiles that were differentially expressed in cells overexpressing Bmi-1. Then, miRNAs that could regulate RKIP were identified. Quantitative real-time PCR (qRT-PCR) and Western blotting were performed to measure the expression of Bmi-1, miR-155, miR-27a and RKIP. RKIP was confirmed as a target of miR-27a and miR-155 through luciferase reporter assays, qRT-PCR and Western blotting. The effects of the Bmi-1/miR-27a/RKIP and Bmi-1/miR-155/RKIP axes on tumor growth, proliferation, migration, invasion, colony-formation ability, metastasis and chemoresistance were investigated both in vitro and in vivo. RESULTS The downregulation of RKIP by Bmi-1 occurred at the protein but not mRNA level. This indicates probable posttranscriptional regulation. miRNA expression profiles of cells with ectopic expression of Bmi-1 were analyzed and compared to those of control cells by microarray analysis. A total of 51 upregulated and 72 downregulated miRNAs were identified. Based on publicly available algorithms, miR-27a and miR-155 were predicted, selected and demonstrated to target RKIP. Bmi-1, miR-27a and miR-155 are elevated in human GC and associated with poor prognosis of GC, while RKIP is expressed at lower levels in GC and correlated with good prognosis. Then, in vitro tests shown that in addition to regulating RKIP expression via miR-27a and miR-155, Bmi-1 was also able to regulate the migration, invasion, proliferation, colony-formation ability and chemosensitivity of GC cells through the same pathway. Finally, the in vivo test showed similar results, whereby the knockdown of the Bmi-1 gene led to the inhibition of tumor growth, metastasis and chemoresistance through miR-27a and miR-155. CONCLUSIONS Bmi-1 was proven to induce the expression of miR-27a and miR-155 and thus promote tumor metastasis and chemoresistance by targeting RKIP in GC. Overall, miR-27a and miR-155 might be promising targets for the screening, diagnosis, prognosis, treatment and disease monitoring of GC.
Collapse
Affiliation(s)
- Yaqing Li
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510120, P. R. China
- Department of Gastroenterology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510120, P. R. China
| | - Zhenfeng Tian
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510120, P. R. China
- Department of Gastroenterology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510120, P. R. China
| | - Ying Tan
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510120, P. R. China
- Department of Gastroenterology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510120, P. R. China
| | - Guoda Lian
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510120, P. R. China
- Department of Gastroenterology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510120, P. R. China
| | - Shangxiang Chen
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510120, P. R. China
- Department of Gastroenterology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510120, P. R. China
| | - Shaojie Chen
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510120, P. R. China
- Department of Gastroenterology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510120, P. R. China
| | - Jiajia Li
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510120, P. R. China
- Department of Nephrology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510120, P. R. China
| | - Xuanna Li
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510120, P. R. China
- Department of Gastroenterology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510120, P. R. China
| | - Kaihong Huang
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510120, P. R. China.
- Department of Gastroenterology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510120, P. R. China.
| | - Yinting Chen
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510120, P. R. China.
- Department of Gastroenterology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510120, P. R. China.
| |
Collapse
|
31
|
Althobiti M, Muftah AA, Aleskandarany MA, Joseph C, Toss MS, Green A, Rakha E. The prognostic significance of BMI1 expression in invasive breast cancer is dependent on its molecular subtypes. Breast Cancer Res Treat 2020; 182:581-589. [PMID: 32524353 PMCID: PMC7320923 DOI: 10.1007/s10549-020-05719-x] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2019] [Accepted: 06/01/2020] [Indexed: 02/07/2023]
Abstract
Purpose BMI1, which is a major component of the polycomb group complex 1, is an essential epigenetic repressor of multiple regulatory genes and has been identified as a cancer stem cell (CSC) marker in several cancers. However, its role in breast cancer (BC) remains to be defined. In this study, we have evaluated the prognostic significance of BMI1 among the different molecular subtypes and assessed its association with other breast CSC markers (BCSC). Material and method BMI1 copy number and mRNA was assessed in large and well-characterised cohorts of early-stage BC patients [METABRIC (n = 1980) and the Bc-GenExMiner (n = 9616) databases]. BMI1 protein expression was assessed using tissue microarray and immunohistochemistry in a cohort of 870 invasive BC patients with long-term outcome data and the expression of a panel of BCSC markers was monitored. Result BMI1 expression, prognostic significance and its association with BCSC markers were differed between molecular classes. In the luminal oestrogen receptor-positive (ER+) BC, BMI1 showed significantly higher expression compared to ER− tumours. BMI1 showed positive correlation with favourable prognostic features and it was negatively associated with the expression of key BCSC markers (ALDH1A1, CD24, CD44, CD133, SOX10 and SOX9). High expression of BMI1 was associated with longer breast cancer-specific survival (BCSS) independent of other prognostic variables. In the basal triple negative BC subtype, BMI1 expression showed positive association with CD133 and SOX10 and it was significantly associated with shorter BCSS. Conclusion High BMI1 expression is associated with clinicopathological variables and outcome in BC. However, this association is dependent on the molecular subtypes. Further functional assessment to detect its underlying mechanistic roles in BC subtypes is warranted. Electronic supplementary material The online version of this article (10.1007/s10549-020-05719-x) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Maryam Althobiti
- Division of Cancer and Stem Cells, Nottingham Breast Cancer Research Centre, University of Nottingham Biodiscovery Institute, University Park, Nottingham, NG7 2RD, UK.,Department of Clinical Laboratory Science, College of Applied Medical Science, Shaqra University 33, Shaqra, 11961, Saudi Arabia
| | - Abir A Muftah
- Department of Pathology, Faculty of Medicine, Faculty of Medicine and Health Science, University of Benghazi, Benghazi, Libya
| | - Mohammed A Aleskandarany
- Division of Cancer and Stem Cells, Nottingham Breast Cancer Research Centre, University of Nottingham Biodiscovery Institute, University Park, Nottingham, NG7 2RD, UK
| | - Chitra Joseph
- Division of Cancer and Stem Cells, Nottingham Breast Cancer Research Centre, University of Nottingham Biodiscovery Institute, University Park, Nottingham, NG7 2RD, UK
| | - Michael S Toss
- Division of Cancer and Stem Cells, Nottingham Breast Cancer Research Centre, University of Nottingham Biodiscovery Institute, University Park, Nottingham, NG7 2RD, UK
| | - Andrew Green
- Division of Cancer and Stem Cells, Nottingham Breast Cancer Research Centre, University of Nottingham Biodiscovery Institute, University Park, Nottingham, NG7 2RD, UK
| | - Emad Rakha
- Division of Cancer and Stem Cells, Nottingham Breast Cancer Research Centre, University of Nottingham Biodiscovery Institute, University Park, Nottingham, NG7 2RD, UK. .,Department of Histopathology, Nottingham University Hospital NHS Trust, City Hospital Campus, Hucknall Road, Nottingham, NG5 1PB, UK.
| |
Collapse
|
32
|
Liu Q, Li Q, Zhu S, Yi Y, Cao Q. B lymphoma Moloney murine leukemia virus insertion region 1: An oncogenic mediator in prostate cancer. Asian J Androl 2020; 21:224-232. [PMID: 29862993 PMCID: PMC6498728 DOI: 10.4103/aja.aja_38_18] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
B lymphoma Moloney murine leukemia virus insertion region 1 (BMI1), a core member of polycomb repressive complex 1 (PRC1), has been intensely investigated in the field of cancer epigenetics for decades. Widely known as a critical regulator in cellular physiology, BMI1 is essential in self-renewal and differentiation in different lineages of stem cells. BMI1 also plays a significant role in cancer etiology for its involvement in pathological progress such as epithelial–mesenchymal transition (EMT) and cancer stem cell maintenance, propagation, and differentiation. Importantly, overexpression of BMI1 is predictive for drug resistance, tumor recurrence, and eventual therapy failure of various cancer subtypes, which renders the pharmacological targeting at BMI1 as a novel and promising therapeutic approach. The study on prostate cancer, a prevalent hormone-related cancer among men, has promoted enormous research advancements in cancer genetics and epigenetics. This review summarizes the role of BMI1 as an oncogenic and epigenetic regulator in tumor initiation, progression, and relapse of prostate cancer.
Collapse
Affiliation(s)
- Qipeng Liu
- Center for Inflammation and Epigenetics, Houston Methodist Research Institute, Houston, TX 77030, USA.,Xiangya School of Medicine, Central South University, Changsha 410008, China
| | - Qiaqia Li
- Center for Inflammation and Epigenetics, Houston Methodist Research Institute, Houston, TX 77030, USA.,Xiangya School of Medicine, Central South University, Changsha 410008, China
| | - Sen Zhu
- Center for Inflammation and Epigenetics, Houston Methodist Research Institute, Houston, TX 77030, USA
| | - Yang Yi
- Center for Inflammation and Epigenetics, Houston Methodist Research Institute, Houston, TX 77030, USA.,Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Sun Yat-Sen University, Guangzhou 510080, China.,Department of Histology and Embryology, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou 510080, China
| | - Qi Cao
- Center for Inflammation and Epigenetics, Houston Methodist Research Institute, Houston, TX 77030, USA.,Houston Methodist Cancer Center, Houston Methodist Research Institute, Houston, TX 77030, USA.,Department of Microbiology and Immunology, Weill Cornell Medicine, Cornell University, New York, NY 10065, USA
| |
Collapse
|
33
|
de Oliveira VM, Caetano DLZ, da Silva FB, Mouro PR, de Oliveira AB, de Carvalho SJ, Leite VBP. pH and Charged Mutations Modulate Cold Shock Protein Folding and Stability: A Constant pH Monte Carlo Study. J Chem Theory Comput 2020; 16:765-772. [PMID: 31756296 DOI: 10.1021/acs.jctc.9b00894] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
The folding and stability of proteins is a fundamental problem in several research fields. In the present paper, we have used different computational approaches to study the effects caused by changes in pH and for charged mutations in cold shock proteins from Bacillus subtilis (Bs-CspB). First, we have investigated the contribution of each ionizable residue for these proteins to their thermal stability using the TKSA-MC, a Web server for rational mutation via optimizing the protein charge interactions. Based on these results, we have proposed a new mutation in an already optimized Bs-CspB variant. We have evaluated the effects of this new mutation in the folding energy landscape using structure-based models in Monte Carlo simulation at constant pH, SBM-CpHMC. Our results using this approach have indicated that the charge rearrangements already in the unfolded state are critical to the thermal stability of Bs-CspB. Furthermore, the conjunction of these simplified methods was able not only to predict stabilizing mutations in different pHs but also to provide essential information about their effects in each stage of protein folding.
Collapse
Affiliation(s)
- Vinícius M de Oliveira
- Brazilian Biosciences National Laboratory, National Center for Research in Energy and Materials, LNBio/CNPEM , Campinas , São Paulo , 13083-970 , Brazil
| | - Daniel L Z Caetano
- Department of Physics , São Paulo State University (UNESP), Institute of Biosciences, Humanities and Exact Sciences , São José do Rio Preto , São Paulo , 15054-000 , Brazil
| | - Fernando B da Silva
- Department of Physics , São Paulo State University (UNESP), Institute of Biosciences, Humanities and Exact Sciences , São José do Rio Preto , São Paulo , 15054-000 , Brazil
| | - Paulo R Mouro
- Department of Physics , São Paulo State University (UNESP), Institute of Biosciences, Humanities and Exact Sciences , São José do Rio Preto , São Paulo , 15054-000 , Brazil
| | - Antonio B de Oliveira
- Department of Physics , São Paulo State University (UNESP), Institute of Biosciences, Humanities and Exact Sciences , São José do Rio Preto , São Paulo , 15054-000 , Brazil
| | - Sidney J de Carvalho
- Department of Physics , São Paulo State University (UNESP), Institute of Biosciences, Humanities and Exact Sciences , São José do Rio Preto , São Paulo , 15054-000 , Brazil
| | - Vitor B P Leite
- Department of Physics , São Paulo State University (UNESP), Institute of Biosciences, Humanities and Exact Sciences , São José do Rio Preto , São Paulo , 15054-000 , Brazil.,Center for Theoretical Biological Physics , Rice University , Houston , Texas 77005 , United States
| |
Collapse
|
34
|
Nagy T, Fisi V, Frank D, Kátai E, Nagy Z, Miseta A. Hyperglycemia-Induced Aberrant Cell Proliferation; A Metabolic Challenge Mediated by Protein O-GlcNAc Modification. Cells 2019; 8:E999. [PMID: 31466420 PMCID: PMC6769692 DOI: 10.3390/cells8090999] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2019] [Revised: 08/26/2019] [Accepted: 08/26/2019] [Indexed: 12/13/2022] Open
Abstract
Chronic hyperglycemia has been associated with an increased prevalence of pathological conditions including cardiovascular disease, cancer, or various disorders of the immune system. In some cases, these associations may be traced back to a common underlying cause, but more often, hyperglycemia and the disturbance in metabolic balance directly facilitate pathological changes in the regular cellular functions. One such cellular function crucial for every living organism is cell cycle regulation/mitotic activity. Although metabolic challenges have long been recognized to influence cell proliferation, the direct impact of diabetes on cell cycle regulatory elements is a relatively uncharted territory. Among other "nutrient sensing" mechanisms, protein O-linked β-N-acetylglucosamine (O-GlcNAc) modification emerged in recent years as a major contributor to the deleterious effects of hyperglycemia. An increasing amount of evidence suggest that O-GlcNAc may significantly influence the cell cycle and cellular proliferation. In our present review, we summarize the current data available on the direct impact of metabolic changes caused by hyperglycemia in pathological conditions associated with cell cycle disorders. We also review published experimental evidence supporting the hypothesis that O-GlcNAc modification may be one of the missing links between metabolic regulation and cellular proliferation.
Collapse
Affiliation(s)
- Tamás Nagy
- Department of Laboratory Medicine, Medical School, University of Pécs, H-7624 Pécs, Hungary.
| | - Viktória Fisi
- Department of Laboratory Medicine, Medical School, University of Pécs, H-7624 Pécs, Hungary
| | - Dorottya Frank
- Department of Dentistry, Oral and Maxillofacial Surgery, Medical School, University of Pécs, H-7621 Pécs, Hungary
| | - Emese Kátai
- Department of Laboratory Medicine, Medical School, University of Pécs, H-7624 Pécs, Hungary
| | - Zsófia Nagy
- Department of Laboratory Medicine, Medical School, University of Pécs, H-7624 Pécs, Hungary
| | - Attila Miseta
- Department of Laboratory Medicine, Medical School, University of Pécs, H-7624 Pécs, Hungary
| |
Collapse
|
35
|
Akella NM, Ciraku L, Reginato MJ. Fueling the fire: emerging role of the hexosamine biosynthetic pathway in cancer. BMC Biol 2019; 17:52. [PMID: 31272438 PMCID: PMC6610925 DOI: 10.1186/s12915-019-0671-3] [Citation(s) in RCA: 258] [Impact Index Per Article: 43.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/10/2023] Open
Abstract
Altered metabolism and deregulated cellular energetics are now considered a hallmark of all cancers. Glucose, glutamine, fatty acids, and amino acids are the primary drivers of tumor growth and act as substrates for the hexosamine biosynthetic pathway (HBP). The HBP culminates in the production of an amino sugar uridine diphosphate N-acetylglucosamine (UDP-GlcNAc) that, along with other charged nucleotide sugars, serves as the basis for biosynthesis of glycoproteins and other glycoconjugates. These nutrient-driven post-translational modifications are highly altered in cancer and regulate protein functions in various cancer-associated processes. In this review, we discuss recent progress in understanding the mechanistic relationship between the HBP and cancer.
Collapse
Affiliation(s)
- Neha M Akella
- Department of Biochemistry and Molecular Biology, Drexel University College of Medicine, Philadelphia, PA, 19102, USA
| | - Lorela Ciraku
- Department of Biochemistry and Molecular Biology, Drexel University College of Medicine, Philadelphia, PA, 19102, USA
| | - Mauricio J Reginato
- Department of Biochemistry and Molecular Biology, Drexel University College of Medicine, Philadelphia, PA, 19102, USA.
| |
Collapse
|
36
|
Sharma NS, Gupta VK, Dauer P, Kesh K, Hadad R, Giri B, Chandra A, Dudeja V, Slawson C, Banerjee S, Vickers SM, Saluja A, Banerjee S. O-GlcNAc modification of Sox2 regulates self-renewal in pancreatic cancer by promoting its stability. Theranostics 2019; 9:3410-3424. [PMID: 31281487 PMCID: PMC6587167 DOI: 10.7150/thno.32615] [Citation(s) in RCA: 48] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2018] [Accepted: 04/24/2019] [Indexed: 12/16/2022] Open
Abstract
Pancreatic adenocarcinoma (PDAC) claims more than 90% of the patients diagnosed with the disease owing to its aggressive biology that is manifested by high rate of tumor recurrence. Aberrant upregulation in the transcriptional activity of proteins involved in self-renewal like Sox2, Oct4 and Nanog is instrumental in these recurrence phenomena. In cancer, Sox2 is aberrantly "turned-on" leading to activation of downstream genes those results in relapse of the tumor. Molecular mechanisms that regulate the activity of Sox2 in PDAC are not known. In the current study, we have studied the how glycosylation of Sox2 by O-GlcNAc transferase (OGT) can affect its transcriptional activity and thus regulate self-renewal in cancer. Methods: RNA-Seq analysis of CRISPR-OGTi PDAC cells indicated a deregulation of differentiation and self-renewal pathways in PDAC. Pancreatic tumor burden following inhibition of OGT in vivo was done by using small molecule inhibitor, OSMI, on subcutaneous implantation of PDAC cells. Sox2 activity assay was performed by Dual Luciferase Reporter Assay kit. Results: Our study shows for the first time that in PDAC, glycosylation of Sox2 by OGT stabilizes it in the nucleus. Site directed mutagenesis of this site (S246A) prevents this modification. We further show that inhibition of OGT delayed initiation of pancreatic tumors by inhibition of Sox2. We also show that targeting OGT in vivo with a small molecule-inhibitor OSMI, results in decreased tumor burden in PDAC. Conclusion: Understanding this mechanism of SOX2 regulation by its glycosylation is expected to pave the way for development of novel therapy that has the potential to eradicate the cells responsible for tumor-recurrence.
Collapse
Affiliation(s)
| | | | - Patricia Dauer
- Department of Pharmacology, University of Minnesota, Minneapolis Minnesota
| | - Kousik Kesh
- Department of Surgery, University of Miami, Miami, FL
| | - Roey Hadad
- Department of Surgery, University of Miami, Miami, FL
| | - Bhuwan Giri
- Department of Surgery, University of Miami, Miami, FL
| | | | - Vikas Dudeja
- Department of Surgery, University of Miami, Miami, FL
- Sylvester Comprehensive Cancer Center, Miami, FL
| | - Chad Slawson
- University of Kansas Medical Center, Kansas City, KS
| | - Santanu Banerjee
- Department of Surgery, University of Miami, Miami, FL
- Sylvester Comprehensive Cancer Center, Miami, FL
| | - Selwyn M Vickers
- School of Medicine Dean's Office, University of Alabama at Birmingham
| | - Ashok Saluja
- Department of Surgery, University of Miami, Miami, FL
- Sylvester Comprehensive Cancer Center, Miami, FL
| | - Sulagna Banerjee
- Department of Surgery, University of Miami, Miami, FL
- Sylvester Comprehensive Cancer Center, Miami, FL
| |
Collapse
|
37
|
Efimova EV, Appelbe OK, Ricco N, Lee SSY, Liu Y, Wolfgeher DJ, Collins TN, Flor AC, Ramamurthy A, Warrington S, Bindokas VP, Kron SJ. O-GlcNAcylation Enhances Double-Strand Break Repair, Promotes Cancer Cell Proliferation, and Prevents Therapy-Induced Senescence in Irradiated Tumors. Mol Cancer Res 2019; 17:1338-1350. [PMID: 30885991 DOI: 10.1158/1541-7786.mcr-18-1025] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2018] [Revised: 02/04/2019] [Accepted: 03/14/2019] [Indexed: 12/16/2022]
Abstract
The metabolic reprogramming associated with characteristic increases in glucose and glutamine metabolism in advanced cancer is often ascribed to answering a higher demand for metabolic intermediates required for rapid tumor cell growth. Instead, recent discoveries have pointed to an alternative role for glucose and glutamine metabolites as cofactors for chromatin modifiers and other protein posttranslational modification enzymes in cancer cells. Beyond epigenetic mechanisms regulating gene expression, many chromatin modifiers also modulate DNA repair, raising the question whether cancer metabolic reprogramming may mediate resistance to genotoxic therapy and genomic instability. Our prior work had implicated N-acetyl-glucosamine (GlcNAc) formation by the hexosamine biosynthetic pathway (HBP) and resulting protein O-GlcNAcylation as a common means by which increased glucose and glutamine metabolism can drive double-strand break (DSB) repair and resistance to therapy-induced senescence in cancer cells. We have examined the effects of modulating O-GlcNAcylation on the DNA damage response (DDR) in MCF7 human mammary carcinoma in vitro and in xenograft tumors. Proteomic profiling revealed deregulated DDR pathways in cells with altered O-GlcNAcylation. Promoting protein O-GlcNAc modification by targeting O-GlcNAcase or simply treating animals with GlcNAc protected tumor xenografts against radiation. In turn, suppressing protein O-GlcNAcylation by blocking O-GlcNAc transferase activity led to delayed DSB repair, reduced cell proliferation, and increased cell senescence in vivo. Taken together, these findings confirm critical connections between cancer metabolic reprogramming, DDR, and senescence and provide a rationale to evaluate agents targeting O-GlcNAcylation in patients as a means to restore tumor sensitivity to radiotherapy. IMPLICATIONS: The finding that the HBP, via its impact on protein O-GlcNAcylation, is a key determinant of the DDR in cancer provides a mechanistic link between metabolic reprogramming, genomic instability, and therapeutic response and suggests novel therapeutic approaches for tumor radiosensitization.
Collapse
Affiliation(s)
- Elena V Efimova
- Department of Molecular Genetics and Cell Biology, The University of Chicago, Chicago, IL
| | - Oliver K Appelbe
- Department of Molecular Genetics and Cell Biology, The University of Chicago, Chicago, IL
| | - Natalia Ricco
- Department of Molecular Genetics and Cell Biology, The University of Chicago, Chicago, IL
| | - Steve S-Y Lee
- Department of Molecular Genetics and Cell Biology, The University of Chicago, Chicago, IL
| | - Yue Liu
- Department of Molecular Genetics and Cell Biology, The University of Chicago, Chicago, IL
| | - Donald J Wolfgeher
- Department of Molecular Genetics and Cell Biology, The University of Chicago, Chicago, IL
| | - Tamica N Collins
- Department of Molecular Genetics and Cell Biology, The University of Chicago, Chicago, IL
| | - Amy C Flor
- Department of Molecular Genetics and Cell Biology, The University of Chicago, Chicago, IL
| | - Aishwarya Ramamurthy
- Department of Molecular Genetics and Cell Biology, The University of Chicago, Chicago, IL
| | - Sara Warrington
- Department of Molecular Genetics and Cell Biology, The University of Chicago, Chicago, IL
| | - Vytautas P Bindokas
- Integrated Light Microscopy Facility, The University of Chicago, Chicago, IL
| | - Stephen J Kron
- Department of Molecular Genetics and Cell Biology, The University of Chicago, Chicago, IL. .,Ludwig Center for Metastasis Research, The University of Chicago, Chicago, IL
| |
Collapse
|
38
|
Decourcelle A, Leprince D, Dehennaut V. Regulation of Polycomb Repression by O-GlcNAcylation: Linking Nutrition to Epigenetic Reprogramming in Embryonic Development and Cancer. Front Endocrinol (Lausanne) 2019; 10:117. [PMID: 30873122 PMCID: PMC6400832 DOI: 10.3389/fendo.2019.00117] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/10/2018] [Accepted: 02/08/2019] [Indexed: 11/25/2022] Open
Abstract
Epigenetic modifications are major actors of early embryogenesis and carcinogenesis and are sensitive to nutritional environment. In recent years, the nutritional sensor O-GlcNAcylation has been recognized as a key regulator of chromatin remodeling. In this review, we summarize and discuss recent clues that OGT and O-GlcNAcylation intimately regulate the functions of the Polycomb group proteins at different levels especially during Drosophila melanogaster embryonic development and in human cancer cell lines. These observations define an additional connection between nutrition and epigenetic reprogramming associated to embryonic development and cancer.
Collapse
|
39
|
Chai Y, Du Y, Zhang S, Xiao J, Luo Z, He F, Huang K. MicroRNA-485-5p reduces O-GlcNAcylation of Bmi-1 and inhibits colorectal cancer proliferation. Exp Cell Res 2018; 368:111-118. [PMID: 29680296 DOI: 10.1016/j.yexcr.2018.04.020] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2018] [Revised: 04/16/2018] [Accepted: 04/17/2018] [Indexed: 12/13/2022]
Abstract
Emerging evidences showed that miRNAs are involved in the oncogenesis of many cancers. Here, miRNA microarray analysis was performed to screen the significant miRNAs involved in the progression of colorectal cancer (CRC), miR-485-5p was chosen for further study. We found that the expression of miR-485-5p was significantly lower in CRC specimens and cell lines. In addition, low expression level of miR-485-5p is correlated with tumor progression and poor survival in CRC patients. Based on in vitro and in vivo assays, we found that miR-485-5p significantly inhibits CRC proliferation. Moreover, our results showed that miR-485-5p inhibits cell proliferation by reducing Bmi-1 protein expression, which has been reported to control the proliferation of many cancers. Mechanistically, OGT is a direct target of miR-485-5p, and miR-485-5p could inhibit the O-GlcNAcylation level of Bmi-1 by OGT. Overall, these results suggested that as a tumor suppressor, miR-485-5p may regulate CRC cells proliferation, which could regulate the O-GlcNAcylation and the stability of Bmi-1 through targeting OGT. This may give insight into a novel mechanism and therapy of CRC growth.
Collapse
Affiliation(s)
- Yong Chai
- Department of Ophthalmology, Jiangxi Children's Hospital, Nanchang, Jiangxi Province 330006, China
| | - Yunyan Du
- Department of Otorhinolaryngology, Jiangxi Provincial People's Hospital, Nanchang, Jiangxi Province 330006, China
| | - Shouhua Zhang
- Department of Ophthalmology, Jiangxi Children's Hospital, Nanchang, Jiangxi Province 330006, China
| | - Juhua Xiao
- Department of Ultrasound, Jiangxi Provincial Maternal and Child Health Hospital, Nanchang, Jiangxi Province 330006, China
| | - Zhipeng Luo
- Department of Gastrointestinal Surgery, Jiangxi Provincial Cancer Hospital, Nanchang 330029, China
| | - Fei He
- Department of Ophthalmology, Jiangxi Children's Hospital, Nanchang, Jiangxi Province 330006, China
| | - Kai Huang
- Department of Gastrointestinal Surgery, Jiangxi Provincial Cancer Hospital, Nanchang 330029, China.
| |
Collapse
|
40
|
Lambert M, Bastide B, Cieniewski-Bernard C. Involvement of O-GlcNAcylation in the Skeletal Muscle Physiology and Physiopathology: Focus on Muscle Metabolism. Front Endocrinol (Lausanne) 2018; 9:578. [PMID: 30459708 PMCID: PMC6232757 DOI: 10.3389/fendo.2018.00578] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/21/2018] [Accepted: 09/11/2018] [Indexed: 12/13/2022] Open
Abstract
Skeletal muscle represents around 40% of whole body mass. The principal function of skeletal muscle is the conversion of chemical energy toward mechanic energy to ensure the development of force, provide movement and locomotion, and maintain posture. This crucial energy dependence is maintained by the faculty of the skeletal muscle for being a central place as a "reservoir" of amino acids and carbohydrates in the whole body. A fundamental post-translational modification, named O-GlcNAcylation, depends, inter alia, on these nutrients; it consists to the transfer or the removal of a unique monosaccharide (N-acetyl-D-glucosamine) to a serine or threonine hydroxyl group of nucleocytoplasmic and mitochondrial proteins in a dynamic process by the O-GlcNAc Transferase (OGT) and the O-GlcNAcase (OGA), respectively. O-GlcNAcylation has been shown to be strongly involved in crucial intracellular mechanisms through the modulation of signaling pathways, gene expression, or cytoskeletal functions in various organs and tissues, such as the brain, liver, kidney or pancreas, and linked to the etiology of associated diseases. In recent years, several studies were also focused on the role of O-GlcNAcylation in the physiology and the physiopathology of skeletal muscle. These studies were mostly interested in O-GlcNAcylation during muscle exercise or muscle-wasting conditions. Major findings pointed out a different "O-GlcNAc signature" depending on muscle type metabolism at resting, wasting and exercise conditions, as well as depending on acute or long-term exhausting exercise protocol. First insights showed some differential OGT/OGA expression and/or activity associated with some differential stress cellular responses through Reactive Oxygen Species and/or Heat-Shock Proteins. Robust data displayed that these O-GlcNAc changes could lead to (i) a differential modulation of the carbohydrates metabolism, since the majority of enzymes are known to be O-GlcNAcylated, and to (ii) a differential modulation of the protein synthesis/degradation balance since O-GlcNAcylation regulates some key signaling pathways such as Akt/GSK3β, Akt/mTOR, Myogenin/Atrogin-1, Myogenin/Mef2D, Mrf4 and PGC-1α in the skeletal muscle. Finally, such involvement of O-GlcNAcylation in some metabolic processes of the skeletal muscle might be linked to some associated diseases such as type 2 diabetes or neuromuscular diseases showing a critical increase of the global O-GlcNAcylation level.
Collapse
|
41
|
"Nutrient-sensing" and self-renewal: O-GlcNAc in a new role. J Bioenerg Biomembr 2017; 50:205-211. [PMID: 29204729 DOI: 10.1007/s10863-017-9735-7] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2017] [Accepted: 11/21/2017] [Indexed: 12/14/2022]
Abstract
Whether embryonic, hematopoietic or cancer stem cells, this metabolic reprogramming is dependent on the nutrient-status and bioenergetic pathways that is influenced by the micro-environmental niches like hypoxia. Thus, the microenvironment plays a vital role in determining the stem cell fate by inducing metabolic reprogramming. Under the influence of the microenvironment, like hypoxia, the stem cells have increased glucose and glutamine uptake which result in activation of hexosamine biosynthesis pathway (HBP) and increased O-GlcNAc Transferase (OGT). The current review is focused on understanding how HBP, a nutrient-sensing pathway (that leads to increased OGT activity) is instrumental in regulating self-renewal not only in embryonic and hematopoietic stem cells (ESC/HSC) but also in cancer stem cells.
Collapse
|
42
|
Tarbet HJ, Toleman CA, Boyce M. A Sweet Embrace: Control of Protein-Protein Interactions by O-Linked β-N-Acetylglucosamine. Biochemistry 2017; 57:13-21. [PMID: 29099585 DOI: 10.1021/acs.biochem.7b00871] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
O-Linked β-N-acetylglucosamine (O-GlcNAc) is a critical post-translational modification (PTM) of thousands of intracellular proteins. Reversible O-GlcNAcylation governs many aspects of cell physiology and is dysregulated in numerous human diseases. Despite this broad pathophysiological significance, major aspects of O-GlcNAc signaling remain poorly understood, including the biochemical mechanisms through which O-GlcNAc transduces information. Recent work from many laboratories, including our own, has revealed that O-GlcNAc, like other intracellular PTMs, can control its substrates' functions by inhibiting or inducing protein-protein interactions. This dynamic regulation of multiprotein complexes exerts diverse downstream signaling effects in a range of processes, cell types, and organisms. Here, we review the literature about O-GlcNAc-regulated protein-protein interactions and suggest important questions for future studies in the field.
Collapse
Affiliation(s)
- Heather J Tarbet
- Department of Biochemistry, Duke University School of Medicine , Durham, North Carolina 27710, United States
| | - Clifford A Toleman
- Department of Biochemistry, Duke University School of Medicine , Durham, North Carolina 27710, United States
| | - Michael Boyce
- Department of Biochemistry, Duke University School of Medicine , Durham, North Carolina 27710, United States
| |
Collapse
|