1
|
Pero JE, Mueller EA, Adams AM, Adolph RS, Bagchi P, Balce D, Bantscheff M, Barauskas O, Bartha I, Bohan D, Cai H, Carabajal E, Cassidy J, Cato M, Chaudhary KW, Chen D, Chen YP, Colas C, Darwech I, Eberl HC, Fernandez B, Gordon E, Grosse J, Hansen J, Hetzler B, Hwang S, Jeyasingh S, Kowalski B, Lehmann S, Lo G, McAllaster M, McHugh C, Momont C, Newby Z, Nigro M, Oladunni F, Pannirselvam M, Park A, Pearson N, Peat AJ, Plastridge B, Ranjan R, Safabakhsh P, Shapiro ND, Soriaga L, Stokes N, Sweeney D, Talecki L, Telenti A, Terrell A, Tse W, Wang L, Wang S, Wedel L, Werner T, Dalmas Wilk D, Yim S, Zhou J. Discovery of Potent STT3A/B Inhibitors and Assessment of Their Multipathogen Antiviral Potential and Safety. J Med Chem 2024; 67:14586-14608. [PMID: 39136957 DOI: 10.1021/acs.jmedchem.4c01402] [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: 08/23/2024]
Abstract
In the aftermath of the COVID-19 pandemic, opportunities to modulate biological pathways common to the lifecycles of viruses need to be carefully considered. N-linked glycosylation in humans is mediated exclusively by the oligosaccharyltransferase complex and is frequently hijacked by viruses to facilitate infection. As such, STT3A/B, the catalytic domain of the OST complex, became an intriguing drug target with broad-spectrum antiviral potential. However, due to the critical role N-linked glycosylation plays in a number of fundamental human processes, the toxicological ramifications of STT3A/B inhibition required attention commensurate to that given to antiviral efficacy. Herein, we describe how known STT3A/B inhibitor NGI-1 inspired the discovery of superior tool compounds which were evaluated in in vitro efficacy and translational safety (e.g., CNS, cardiovascular, liver) studies. The described learnings will appeal to those interested in the therapeutic utility of modulating N-linked glycosylation as well as the broader scientific community.
Collapse
Affiliation(s)
- Joseph E Pero
- GSK, 1250 South Collegeville Road, Collegeville, Pennsylvania 19426, United States
| | - Elizabeth A Mueller
- Vir Biotechnology, Inc., 1800 Owens St., San Francisco, California 94158, United States
| | - Ashley M Adams
- GSK, 1250 South Collegeville Road, Collegeville, Pennsylvania 19426, United States
| | - Ramona S Adolph
- Cellzome GmbH, a GSK company, Meyerhofstrasse 1, 69117 Heidelberg, Germany
| | - Parikshit Bagchi
- Vir Biotechnology, Inc., 1800 Owens St., San Francisco, California 94158, United States
| | - Dale Balce
- Vir Biotechnology, Inc., 1800 Owens St., San Francisco, California 94158, United States
| | - Marcus Bantscheff
- Cellzome GmbH, a GSK company, Meyerhofstrasse 1, 69117 Heidelberg, Germany
| | - Ona Barauskas
- Vir Biotechnology, Inc., 1800 Owens St., San Francisco, California 94158, United States
| | - Istvan Bartha
- Vir Biotechnology, Inc., 1800 Owens St., San Francisco, California 94158, United States
| | - Dana Bohan
- Vir Biotechnology, Inc., 1800 Owens St., San Francisco, California 94158, United States
| | - Haiying Cai
- Vir Biotechnology, Inc., 1800 Owens St., San Francisco, California 94158, United States
| | - Esteban Carabajal
- Vir Biotechnology, Inc., 1800 Owens St., San Francisco, California 94158, United States
| | - James Cassidy
- Vir Biotechnology, Inc., 1800 Owens St., San Francisco, California 94158, United States
| | - Matthew Cato
- GSK, 1250 South Collegeville Road, Collegeville, Pennsylvania 19426, United States
| | - Khuram W Chaudhary
- GSK, 1250 South Collegeville Road, Collegeville, Pennsylvania 19426, United States
| | - Dingjun Chen
- Vir Biotechnology, Inc., 1800 Owens St., San Francisco, California 94158, United States
| | - Yi-Pei Chen
- Vir Biotechnology, Inc., 1800 Owens St., San Francisco, California 94158, United States
| | - Christophe Colas
- Vir Biotechnology, Inc., 1800 Owens St., San Francisco, California 94158, United States
| | - Isra Darwech
- Vir Biotechnology, Inc., 1800 Owens St., San Francisco, California 94158, United States
| | - H Christian Eberl
- Cellzome GmbH, a GSK company, Meyerhofstrasse 1, 69117 Heidelberg, Germany
| | - Beth Fernandez
- Vir Biotechnology, Inc., 1800 Owens St., San Francisco, California 94158, United States
| | - Earl Gordon
- GSK, 1250 South Collegeville Road, Collegeville, Pennsylvania 19426, United States
| | - Johannes Grosse
- Vir Biotechnology, Inc., 1800 Owens St., San Francisco, California 94158, United States
| | - Justin Hansen
- Vir Biotechnology, Inc., 1800 Owens St., San Francisco, California 94158, United States
| | - Belinda Hetzler
- Vir Biotechnology, Inc., 1800 Owens St., San Francisco, California 94158, United States
| | - Seungmin Hwang
- Vir Biotechnology, Inc., 1800 Owens St., San Francisco, California 94158, United States
| | - Sam Jeyasingh
- Vir Biotechnology, Inc., 1800 Owens St., San Francisco, California 94158, United States
| | - Beatriz Kowalski
- Vir Biotechnology, Inc., 1800 Owens St., San Francisco, California 94158, United States
| | - Stephanie Lehmann
- Cellzome GmbH, a GSK company, Meyerhofstrasse 1, 69117 Heidelberg, Germany
| | - Gary Lo
- Vir Biotechnology, Inc., 1800 Owens St., San Francisco, California 94158, United States
| | - Michael McAllaster
- Vir Biotechnology, Inc., 1800 Owens St., San Francisco, California 94158, United States
| | - Charles McHugh
- GSK, 1250 South Collegeville Road, Collegeville, Pennsylvania 19426, United States
| | - Corey Momont
- Vir Biotechnology, Inc., 1800 Owens St., San Francisco, California 94158, United States
| | - Zachary Newby
- Vir Biotechnology, Inc., 1800 Owens St., San Francisco, California 94158, United States
| | - Maria Nigro
- Vir Biotechnology, Inc., 1800 Owens St., San Francisco, California 94158, United States
| | - Fatai Oladunni
- Vir Biotechnology, Inc., 1800 Owens St., San Francisco, California 94158, United States
| | - Malar Pannirselvam
- GSK, 1250 South Collegeville Road, Collegeville, Pennsylvania 19426, United States
| | - Arnold Park
- Vir Biotechnology, Inc., 1800 Owens St., San Francisco, California 94158, United States
| | - Neil Pearson
- GSK, 1250 South Collegeville Road, Collegeville, Pennsylvania 19426, United States
| | - Andrew J Peat
- GSK, 1250 South Collegeville Road, Collegeville, Pennsylvania 19426, United States
| | - Bob Plastridge
- Vir Biotechnology, Inc., 1800 Owens St., San Francisco, California 94158, United States
| | - Rohit Ranjan
- Vir Biotechnology, Inc., 1800 Owens St., San Francisco, California 94158, United States
| | - Pegah Safabakhsh
- Vir Biotechnology, Inc., 1800 Owens St., San Francisco, California 94158, United States
| | - Nathan D Shapiro
- Vir Biotechnology, Inc., 1800 Owens St., San Francisco, California 94158, United States
| | - Leah Soriaga
- Vir Biotechnology, Inc., 1800 Owens St., San Francisco, California 94158, United States
| | - Neil Stokes
- GSK, 1250 South Collegeville Road, Collegeville, Pennsylvania 19426, United States
| | - David Sweeney
- Vir Biotechnology, Inc., 1800 Owens St., San Francisco, California 94158, United States
| | - Lindsey Talecki
- GSK, 1250 South Collegeville Road, Collegeville, Pennsylvania 19426, United States
| | - Amalio Telenti
- Vir Biotechnology, Inc., 1800 Owens St., San Francisco, California 94158, United States
| | - Ashley Terrell
- Vir Biotechnology, Inc., 1800 Owens St., San Francisco, California 94158, United States
| | - Winston Tse
- Vir Biotechnology, Inc., 1800 Owens St., San Francisco, California 94158, United States
| | - Lisha Wang
- Vir Biotechnology, Inc., 1800 Owens St., San Francisco, California 94158, United States
| | - Shuya Wang
- GSK, 1250 South Collegeville Road, Collegeville, Pennsylvania 19426, United States
| | - Laura Wedel
- Vir Biotechnology, Inc., 1800 Owens St., San Francisco, California 94158, United States
| | - Thilo Werner
- Cellzome GmbH, a GSK company, Meyerhofstrasse 1, 69117 Heidelberg, Germany
| | - Deidre Dalmas Wilk
- GSK, 1250 South Collegeville Road, Collegeville, Pennsylvania 19426, United States
| | - Samantha Yim
- Vir Biotechnology, Inc., 1800 Owens St., San Francisco, California 94158, United States
| | - Jiayi Zhou
- Vir Biotechnology, Inc., 1800 Owens St., San Francisco, California 94158, United States
| |
Collapse
|
2
|
Yue J, Huang R, Lan Z, Xiao B, Luo Z. Abnormal glycosylation in glioma: related changes in biology, biomarkers and targeted therapy. Biomark Res 2023; 11:54. [PMID: 37231524 DOI: 10.1186/s40364-023-00491-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2022] [Accepted: 04/26/2023] [Indexed: 05/27/2023] Open
Abstract
Glioma is a rapidly growing and aggressive primary malignant tumor of the central nervous system that can diffusely invade the brain tissue around, and the prognosis of patients is not significantly improved by traditional treatments. One of the most general posttranslational modifications of proteins is glycosylation, and the abnormal distribution of this modification in gliomas may shed light on how it affects biological behaviors of glioma cells, including proliferation, migration, and invasion, which may be produced by regulating protein function, cell-matrix and cell‒cell interactions, and affecting receptor downstream pathways. In this paper, from the perspective of regulating protein glycosylation changes and abnormal expression of glycosylation-related proteins (such as glycosyltransferases in gliomas), we summarize how glycosylation may play a crucial role in the discovery of novel biomarkers and new targeted treatment options for gliomas. Overall, the mechanistic basis of abnormal glycosylation affecting glioma progression remains to be more widely and deeply explored, which not only helps to inspire researchers to further explore related diagnostic and prognostic markers but also provides ideas for discovering effective treatment strategies and improving glioma patient survival and prognosis.
Collapse
Affiliation(s)
- Juan Yue
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
- Xiangya School of Medicine, Central South University, Changsha, 410013, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, 87 Xiangya road of Kaifu district, 410008, Changsha, Hunan, China
| | - Roujie Huang
- Department of Obstetrics and Gynecology, Peking Union Medical College, Peking Union Medical College Hospital, Chinese Academy of Medical Science, Shuaifuyuan No. 1, Dongcheng District, 100730, Beijing, China
| | - Zehao Lan
- Xiangya School of Medicine, Central South University, Changsha, 410013, Hunan, China
| | - Bo Xiao
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, 87 Xiangya road of Kaifu district, 410008, Changsha, Hunan, China
- Clinical Research Center for Epileptic disease of Hunan Province, Central South University, 410008, Changsha, Hunan, P.R. China
| | - Zhaohui Luo
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China.
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, 87 Xiangya road of Kaifu district, 410008, Changsha, Hunan, China.
- Clinical Research Center for Epileptic disease of Hunan Province, Central South University, 410008, Changsha, Hunan, P.R. China.
| |
Collapse
|
3
|
Lecaille F, Chazeirat T, Saidi A, Lalmanach G. Cathepsin V: Molecular characteristics and significance in health and disease. Mol Aspects Med 2022; 88:101086. [PMID: 35305807 DOI: 10.1016/j.mam.2022.101086] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Revised: 02/23/2022] [Accepted: 02/26/2022] [Indexed: 12/31/2022]
Abstract
Human cysteine cathepsins form a family of eleven proteases (B, C, F, H, K, L, O, S, V, W, X/Z) that play important roles in a considerable number of biological and pathophysiological processes. Among them, cathepsin V, also known as cathepsin L2, is a lysosomal enzyme, which is mainly expressed in cornea, thymus, heart, brain, and skin. Cathepsin V is a multifunctional endopeptidase that is involved in both the release of antigenic peptides and the maturation of MHC class II molecules and participates in the turnover of elastin fibrils as well in the cleavage of intra- and extra-cellular substrates. Moreover, there is increasing evidence that cathepsin V may contribute to the progression of diverse diseases, due to the dysregulation of its expression and/or its activity. For instance, increased expression of cathepsin V is closely correlated with malignancies (breast cancer, squamous cell carcinoma, or colorectal cancer) as well vascular disorders (atherosclerosis, aortic aneurysm, hypertension) being the most prominent examples. This review aims to shed light on current knowledge on molecular aspects of cathepsin V (genomic organization, protein structure, substrate specificity), its regulation by protein and non-protein inhibitors as well to summarize its expression (tissue and cellular distribution). Then the core biological and pathophysiological roles of cathepsin V will be depicted, raising the question of its interest as a valuable target that can open up pioneering therapeutic avenues.
Collapse
Affiliation(s)
- Fabien Lecaille
- Université de Tours, Tours, France; INSERM, UMR 1100, Centre d'Etude des Pathologies Respiratoires (CEPR), Team "Mécanismes protéolytiques dans l'inflammation", Tours, France.
| | - Thibault Chazeirat
- Université de Tours, Tours, France; INSERM, UMR 1100, Centre d'Etude des Pathologies Respiratoires (CEPR), Team "Mécanismes protéolytiques dans l'inflammation", Tours, France
| | - Ahlame Saidi
- Université de Tours, Tours, France; INSERM, UMR 1100, Centre d'Etude des Pathologies Respiratoires (CEPR), Team "Mécanismes protéolytiques dans l'inflammation", Tours, France
| | - Gilles Lalmanach
- Université de Tours, Tours, France; INSERM, UMR 1100, Centre d'Etude des Pathologies Respiratoires (CEPR), Team "Mécanismes protéolytiques dans l'inflammation", Tours, France.
| |
Collapse
|
4
|
Li X, Loh TJ, Lim JJ, Er Saw P, Liao Y. Glycan-RNA: a new class of non-coding RNA. BIO INTEGRATION 2022. [DOI: 10.15212/bioi-2021-0032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Affiliation(s)
- Xiuling Li
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Tiing Jen Loh
- Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton, Victoria 3800, Australia
| | - Jia Jia Lim
- Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton, Victoria 3800, Australia
| | - Phei Er Saw
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Yong Liao
- Key Laboratory of Molecular Biology for Infectious Diseases, Ministry of Education, Institute for Viral Hepatitis, and Department of Infectious Diseases, the Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
| |
Collapse
|
5
|
Xi X, Wang J, Qin Y, Huang W, You Y, Zhan J. Glycosylated modification of MUC1 maybe a new target to promote drug sensitivity and efficacy for breast cancer chemotherapy. Cell Death Dis 2022; 13:708. [PMID: 35970845 PMCID: PMC9378678 DOI: 10.1038/s41419-022-05110-2] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2022] [Revised: 07/13/2022] [Accepted: 07/18/2022] [Indexed: 01/21/2023]
Abstract
Breast cancer, the most common cancer in women, usually exhibits intrinsic insensitivity to drugs, even without drug resistance. MUC1 is a highly glycosylated transmembrane protein, overexpressed in breast cancer, contributing to tumorigenesis and worse prognosis. However, the molecular mechanism between MUC1 and drug sensitivity still remains unclear. Here, natural flavonoid apigenin was used as objective due to the antitumor activity and wide availability. MUC1 knockout (KO) markedly sensitized breast cancer cells to apigenin cytotoxicity in vitro and in vivo. Both genetical and pharmacological inhibition significantly enhanced the chemosensitivity to apigenin and clinical drugs whereas MUC1 overexpression conversely aggravated such drug resistance. Constitutively re-expressing wild type MUC1 in KO cells restored the drug resistance; however, the transmembrane domain deletant could not rescue the phenotype. Notably, further investigation discovered that membrane-dependent drug resistance relied on the extracellular glycosylated modification since removing O-glycosylation via inhibitor, enzyme digestion, or GCNT3 (MUC1 related O-glycosyltransferase) knockout markedly reinvigorated the chemosensitivity in WT cells, but had no effect on KO cells. Conversely, inserting O-glycosylated sites to MUC1-N increased the drug tolerance whereas the O-glycosylated deletant (Ser/Thr to Ala) maintained high susceptibility to drugs. Importantly, the intracellular concentration of apigenin measured by UPLC and fluorescence distribution firmly revealed the increased drug permeation in MUC1 KO and BAG-pretreated cells. Multiple clinical chemotherapeutics with small molecular were tested and obtained the similar conclusion. Our findings uncover a critical role of the extracellular O-glycosylation of MUC1-N in weakening drug sensitivity through acting as a barrier, highlighting a new perspective that targeting MUC1 O-glycosylation has great potential to promote drug sensitivity and efficacy.
Collapse
Affiliation(s)
- Xiaomin Xi
- grid.22935.3f0000 0004 0530 8290Beijing Key Laboratory of Viticulture and Enology, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, People’s Republic of China
| | - Jiting Wang
- grid.22935.3f0000 0004 0530 8290Beijing Key Laboratory of Viticulture and Enology, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, People’s Republic of China
| | - Yue Qin
- grid.22935.3f0000 0004 0530 8290Beijing Key Laboratory of Viticulture and Enology, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, People’s Republic of China
| | - Weidong Huang
- grid.22935.3f0000 0004 0530 8290Beijing Key Laboratory of Viticulture and Enology, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, People’s Republic of China
| | - Yilin You
- grid.22935.3f0000 0004 0530 8290Beijing Key Laboratory of Viticulture and Enology, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, People’s Republic of China
| | - Jicheng Zhan
- grid.22935.3f0000 0004 0530 8290Beijing Key Laboratory of Viticulture and Enology, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, People’s Republic of China
| |
Collapse
|
6
|
Cao X, Meng P, Shao Y, Yan G, Yao J, Zhou X, Liu C, Zhang L, Shu H, Lu H. Nascent Glycoproteome Reveals That N-Linked Glycosylation Inhibitor-1 Suppresses Expression of Glycosylated Lysosome-Associated Membrane Protein-2. Front Mol Biosci 2022; 9:899192. [PMID: 35573732 PMCID: PMC9092021 DOI: 10.3389/fmolb.2022.899192] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2022] [Accepted: 04/12/2022] [Indexed: 11/15/2022] Open
Abstract
Glycosylation inhibition has great potential in cancer treatment. However, the corresponding cellular response, protein expression and glycosylation changes remain unclear. As a cell-permeable small-molecule inhibitor with reduced cellular toxicity, N-linked glycosylation inhibitor-1 (NGI-1) has become a great approach to regulate glycosylation in mammalian cells. Here for the first time, we applied a nascent proteomic method to investigate the effect of NGI-1 in hepatocellular carcinoma (HCC) cell line. Besides, hydrophilic interaction liquid chromatography (HILIC) was adopted for the enrichment of glycosylated peptides. Glycoproteomic analysis revealed the abundance of glycopeptides from LAMP2, NICA, and CEIP2 was significantly changed during NGI-1 treatment. Moreover, the alterations of LAMP2 site-specific intact N-glycopeptides were comprehensively assessed. NGI-1 treatment also led to the inhibition of Cathepsin D maturation and the induction of autophagy. In summary, we provided evidence that NGI-1 repressed the expression of glycosylated LAMP2 accompanied with the occurrence of lysosomal defects and autophagy.
Collapse
Affiliation(s)
- Xinyi Cao
- Institutes of Biomedical Sciences and Shanghai Cancer Center, Fudan University, Shanghai, China
| | - Peiyi Meng
- Department of Chemistry, Fudan University, Shanghai, China
| | - Yuyin Shao
- Institutes of Biomedical Sciences and Shanghai Cancer Center, Fudan University, Shanghai, China
| | - Guoquan Yan
- Institutes of Biomedical Sciences and Shanghai Cancer Center, Fudan University, Shanghai, China
| | - Jun Yao
- Institutes of Biomedical Sciences and Shanghai Cancer Center, Fudan University, Shanghai, China
| | - Xinwen Zhou
- Institutes of Biomedical Sciences and Shanghai Cancer Center, Fudan University, Shanghai, China
| | - Chao Liu
- Beijing Advanced Innovation Center for Precision Medicine, Beihang University, Beijing, China
| | - Lei Zhang
- Institutes of Biomedical Sciences and Shanghai Cancer Center, Fudan University, Shanghai, China
| | - Hong Shu
- Department of Clinical Laboratory, Guangxi Medical University Cancer Hospital, Nanning, China
- *Correspondence: Hong Shu, ; Haojie Lu,
| | - Haojie Lu
- Institutes of Biomedical Sciences and Shanghai Cancer Center, Fudan University, Shanghai, China
- Department of Chemistry, Fudan University, Shanghai, China
- NHC Key Laboratory of Glycoconjugates Research, Fudan University, Shanghai, China
- *Correspondence: Hong Shu, ; Haojie Lu,
| |
Collapse
|
7
|
Dragic H, Barthelaix A, Duret C, Le Goupil S, Laprade H, Martin S, Brugière S, Couté Y, Machon C, Guitton J, Rudewicz J, Hofman P, Lebecque S, Chaveroux C, Ferraro-Peyret C, Renno T, Manié SN. The hexosamine pathway and coat complex II promote malignant adaptation to nutrient scarcity. Life Sci Alliance 2022; 5:5/7/e202101334. [PMID: 35396334 PMCID: PMC9008580 DOI: 10.26508/lsa.202101334] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Revised: 03/23/2022] [Accepted: 03/24/2022] [Indexed: 11/24/2022] Open
Abstract
We present adaptive mechanisms of resistance of lung adenocarcinoma to their harsh microenvironment, which typically contains a lower glucose concentration compared with normal tissue. The glucose-requiring hexosamine biosynthetic pathway (HBP), which produces UDP-N-acetylglucosamine for glycosylation reactions, promotes lung adenocarcinoma (LUAD) progression. However, lung tumor cells often reside in low-nutrient microenvironments, and whether the HBP is involved in the adaptation of LUAD to nutrient stress is unknown. Here, we show that the HBP and the coat complex II (COPII) play a key role in cell survival during glucose shortage. HBP up-regulation withstood low glucose-induced production of proteins bearing truncated N-glycans, in the endoplasmic reticulum. This function for the HBP, alongside COPII up-regulation, rescued cell surface expression of a subset of glycoproteins. Those included the epidermal growth factor receptor (EGFR), allowing an EGFR-dependent cell survival under low glucose in anchorage-independent growth. Accordingly, high expression of the HBP rate-limiting enzyme GFAT1 was associated with wild-type EGFR activation in LUAD patient samples. Notably, HBP and COPII up-regulation distinguished LUAD from the lung squamous-cell carcinoma subtype, thus uncovering adaptive mechanisms of LUAD to their harsh microenvironment.
Collapse
Affiliation(s)
- Helena Dragic
- Centre de Recherche en Cancérologie de Lyon, INSERM U1052, Centre National de la Recherche Scientifique (CNRS) 5286, Centre Léon Bérard, Université de Lyon, Université Claude Bernard Lyon 1, Lyon, France
| | - Audrey Barthelaix
- Institute for Regenerative Medecine and Biotherapy (IRBM), Université de Montpellier, INSERM, Montpellier, France
| | - Cédric Duret
- Centre de Recherche en Cancérologie de Lyon, INSERM U1052, Centre National de la Recherche Scientifique (CNRS) 5286, Centre Léon Bérard, Université de Lyon, Université Claude Bernard Lyon 1, Lyon, France
| | - Simon Le Goupil
- Inserm U1242, Centre de Lutte Contre le Cancer Eugène Marquis, Université de Rennes, Rennes, France
| | - Hadrien Laprade
- Inserm U1242, Centre de Lutte Contre le Cancer Eugène Marquis, Université de Rennes, Rennes, France
| | - Sophie Martin
- Inserm U1242, Centre de Lutte Contre le Cancer Eugène Marquis, Université de Rennes, Rennes, France
| | - Sabine Brugière
- Université Grenoble Alpes, INSERM, Commissariat à l'Energie Atomique (CEA), Unite Mixte de Recherche (UMR) BioSanté U1292, CNRS, CEA, FR2048, Grenoble, France
| | - Yohann Couté
- Université Grenoble Alpes, INSERM, Commissariat à l'Energie Atomique (CEA), Unite Mixte de Recherche (UMR) BioSanté U1292, CNRS, CEA, FR2048, Grenoble, France
| | - Christelle Machon
- Centre de Recherche en Cancérologie de Lyon, INSERM U1052, Centre National de la Recherche Scientifique (CNRS) 5286, Centre Léon Bérard, Université de Lyon, Université Claude Bernard Lyon 1, Lyon, France.,U Hospices Civils of Lyon, Biochemistry and Pharmaco-toxicology Laboratory, Lyon Sud Hospital, Lyon, France
| | - Jerome Guitton
- Centre de Recherche en Cancérologie de Lyon, INSERM U1052, Centre National de la Recherche Scientifique (CNRS) 5286, Centre Léon Bérard, Université de Lyon, Université Claude Bernard Lyon 1, Lyon, France.,U Hospices Civils of Lyon, Biochemistry and Pharmaco-toxicology Laboratory, Lyon Sud Hospital, Lyon, France
| | - Justine Rudewicz
- Bordeaux Bioinformatics Center, CBiB, University of Bordeaux, Bordeaux, France
| | - Paul Hofman
- Laboratory of Clinical and Experimental Pathology, Federation Hospitalo-Universitaire (FHU) OncoAge and BB-0033-00025, Nice University Hospital, IRCAN Antoine Lacassagne Center, Côte d'Azur University, Nice, France
| | - Serge Lebecque
- Centre de Recherche en Cancérologie de Lyon, INSERM U1052, Centre National de la Recherche Scientifique (CNRS) 5286, Centre Léon Bérard, Université de Lyon, Université Claude Bernard Lyon 1, Lyon, France
| | - Cedric Chaveroux
- Centre de Recherche en Cancérologie de Lyon, INSERM U1052, Centre National de la Recherche Scientifique (CNRS) 5286, Centre Léon Bérard, Université de Lyon, Université Claude Bernard Lyon 1, Lyon, France
| | - Carole Ferraro-Peyret
- Centre de Recherche en Cancérologie de Lyon, INSERM U1052, Centre National de la Recherche Scientifique (CNRS) 5286, Centre Léon Bérard, Université de Lyon, Université Claude Bernard Lyon 1, Lyon, France.,Hospices Civils de Lyon, Biopathology of Tumours, GHE Hospital, Bron, France
| | - Toufic Renno
- Centre de Recherche en Cancérologie de Lyon, INSERM U1052, Centre National de la Recherche Scientifique (CNRS) 5286, Centre Léon Bérard, Université de Lyon, Université Claude Bernard Lyon 1, Lyon, France
| | - Serge N Manié
- Centre de Recherche en Cancérologie de Lyon, INSERM U1052, Centre National de la Recherche Scientifique (CNRS) 5286, Centre Léon Bérard, Université de Lyon, Université Claude Bernard Lyon 1, Lyon, France .,Inserm U1242, Centre de Lutte Contre le Cancer Eugène Marquis, Université de Rennes, Rennes, France
| |
Collapse
|
8
|
Almahayni K, Spiekermann M, Fiore A, Yu G, Pedram K, Möckl L. Small molecule inhibitors of mammalian glycosylation. Matrix Biol Plus 2022; 16:100108. [PMID: 36467541 PMCID: PMC9713294 DOI: 10.1016/j.mbplus.2022.100108] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2021] [Revised: 02/10/2022] [Accepted: 03/10/2022] [Indexed: 01/06/2023] Open
Abstract
Glycans are one of the fundamental biopolymers encountered in living systems. Compared to polynucleotide and polypeptide biosynthesis, polysaccharide biosynthesis is a uniquely combinatorial process to which interdependent enzymes with seemingly broad specificities contribute. The resulting intracellular cell surface, and secreted glycans play key roles in health and disease, from embryogenesis to cancer progression. The study and modulation of glycans in cell and organismal biology is aided by small molecule inhibitors of the enzymes involved in glycan biosynthesis. In this review, we survey the arsenal of currently available inhibitors, focusing on agents which have been independently validated in diverse systems. We highlight the utility of these inhibitors and drawbacks to their use, emphasizing the need for innovation for basic research as well as for therapeutic applications.
Collapse
Affiliation(s)
- Karim Almahayni
- Max Planck Institute for the Science of Light, 91058 Erlangen, Germany
| | - Malte Spiekermann
- Max Planck Institute for the Science of Light, 91058 Erlangen, Germany
| | - Antonio Fiore
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA 20147, USA
| | - Guoqiang Yu
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA 20147, USA
| | - Kayvon Pedram
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA 20147, USA,Corresponding authors.
| | - Leonhard Möckl
- Max Planck Institute for the Science of Light, 91058 Erlangen, Germany,Corresponding authors.
| |
Collapse
|
9
|
Saraon P, Snider J, Schormann W, Rai A, Radulovich N, Sánchez-Osuna M, Coulombe-Huntington J, Huard C, Mohammed M, Lima-Fernandes E, Thériault B, Halabelian L, Chan M, Joshi D, Drecun L, Yao Z, Pathmanathan S, Wong V, Lyakisheva A, Aboualizadeh F, Niu L, Li F, Kiyota T, Subramanian R, Joseph B, Aman A, Prakesch M, Isaac M, Mamai A, Poda G, Vedadi M, Marcellus R, Uehling D, Leighl N, Sacher A, Samaržija M, Jakopović M, Arrowsmith C, Tyers M, Tsao MS, Andrews D, Al-Awar R, Stagljar I. Chemical Genetics Screen Identifies COPB2 Tool Compounds That Alters ER Stress Response and Induces RTK Dysregulation in Lung Cancer Cells. J Mol Biol 2021; 433:167294. [PMID: 34662547 DOI: 10.1016/j.jmb.2021.167294] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Revised: 09/27/2021] [Accepted: 09/27/2021] [Indexed: 12/12/2022]
Abstract
Activating mutations in the epidermal growth factor receptor (EGFR) are common driver mutations in non-small cell lung cancer (NSCLC). First, second and third generation EGFR tyrosine kinase inhibitors (TKIs) are effective at inhibiting mutant EGFR NSCLC, however, acquired resistance is a major issue, leading to disease relapse. Here, we characterize a small molecule, EMI66, an analog of a small molecule which we previously identified to inhibit mutant EGFR signalling via a novel mechanism of action. We show that EMI66 attenuates receptor tyrosine kinase (RTK) expression and signalling and alters the electrophoretic mobility of Coatomer Protein Complex Beta 2 (COPB2) protein in mutant EGFR NSCLC cells. Moreover, we demonstrate that EMI66 can alter the subcellular localization of EGFR and COPB2 within the early secretory pathway. Furthermore, we find that COPB2 knockdown reduces the growth of mutant EGFR lung cancer cells, alters the post-translational processing of RTKs, and alters the endoplasmic reticulum (ER) stress response pathway. Lastly, we show that EMI66 treatment also alters the ER stress response pathway and inhibits the growth of mutant EGFR lung cancer cells and organoids. Our results demonstrate that targeting of COPB2 with EMI66 presents a viable approach to attenuate mutant EGFR signalling and growth in NSCLC.
Collapse
Affiliation(s)
- Punit Saraon
- Drug Discovery Program, Ontario Institute for Cancer Research, Ontario, Canada.
| | - Jamie Snider
- Donnelly Centre, University of Toronto, Ontario, Canada
| | - Wiebke Schormann
- Biological Sciences, Sunnybrook Research Institute, Toronto, Ontario, Canada
| | - Ankit Rai
- Cell Biology, Department of Biology, Faculty of Science, Utrecht University, 3548CH Utrecht, the Netherlands
| | - Nikolina Radulovich
- Princess Margaret Cancer Centre, University Health Network, University of Toronto, Toronto, Ontario, Canada
| | - Maria Sánchez-Osuna
- Institute for Research in Immunology and Cancer, Université de Montréal, PO Box 6128, Downtown Station, Montreal, QC H3C 3J7, Canada
| | - Jasmin Coulombe-Huntington
- Institute for Research in Immunology and Cancer, Université de Montréal, PO Box 6128, Downtown Station, Montreal, QC H3C 3J7, Canada
| | - Caroline Huard
- Institute for Research in Immunology and Cancer, Université de Montréal, PO Box 6128, Downtown Station, Montreal, QC H3C 3J7, Canada
| | - Mohammed Mohammed
- Drug Discovery Program, Ontario Institute for Cancer Research, Ontario, Canada
| | | | - Brigitte Thériault
- Drug Discovery Program, Ontario Institute for Cancer Research, Ontario, Canada
| | - Levon Halabelian
- Structural Genomics Consortium, University of Toronto, Toronto, Ontario, Canada
| | - Manuel Chan
- Drug Discovery Program, Ontario Institute for Cancer Research, Ontario, Canada
| | - Dhananjay Joshi
- Drug Discovery Program, Ontario Institute for Cancer Research, Ontario, Canada
| | - Luka Drecun
- Donnelly Centre, University of Toronto, Ontario, Canada; Department of Molecular Genetics, University of Toronto, Ontario, Canada
| | - Zhong Yao
- Donnelly Centre, University of Toronto, Ontario, Canada
| | - Shivanthy Pathmanathan
- Donnelly Centre, University of Toronto, Ontario, Canada; Department of Molecular Genetics, University of Toronto, Ontario, Canada
| | - Victoria Wong
- Donnelly Centre, University of Toronto, Ontario, Canada
| | | | | | - Li Niu
- Princess Margaret Cancer Centre, University Health Network, University of Toronto, Toronto, Ontario, Canada
| | - Fengling Li
- Structural Genomics Consortium, University of Toronto, Toronto, Ontario, Canada
| | - Taira Kiyota
- Drug Discovery Program, Ontario Institute for Cancer Research, Ontario, Canada
| | | | - Babu Joseph
- Drug Discovery Program, Ontario Institute for Cancer Research, Ontario, Canada
| | - Ahmed Aman
- Drug Discovery Program, Ontario Institute for Cancer Research, Ontario, Canada
| | - Michael Prakesch
- Drug Discovery Program, Ontario Institute for Cancer Research, Ontario, Canada
| | - Methvin Isaac
- Drug Discovery Program, Ontario Institute for Cancer Research, Ontario, Canada
| | - Ahmed Mamai
- Drug Discovery Program, Ontario Institute for Cancer Research, Ontario, Canada
| | - Gennady Poda
- Drug Discovery Program, Ontario Institute for Cancer Research, Ontario, Canada; University of Toronto, Leslie Dan Faculty of Pharmacy, Toronto, Ontario, Canada
| | - Masoud Vedadi
- Structural Genomics Consortium, University of Toronto, Toronto, Ontario, Canada; Department of Pharmacology and Toxicology, University of Toronto, Ontario, Canada
| | - Richard Marcellus
- Drug Discovery Program, Ontario Institute for Cancer Research, Ontario, Canada
| | - David Uehling
- Drug Discovery Program, Ontario Institute for Cancer Research, Ontario, Canada
| | - Natasha Leighl
- Princess Margaret Cancer Centre, University Health Network, University of Toronto, Toronto, Ontario, Canada
| | - Adrian Sacher
- Princess Margaret Cancer Centre, University Health Network, University of Toronto, Toronto, Ontario, Canada
| | - Miroslav Samaržija
- Department for Lung Diseases Jordanovac, Clinical Hospital Centre Zagreb, University of Zagreb, Zagreb, Croatia
| | - Marko Jakopović
- Department for Lung Diseases Jordanovac, Clinical Hospital Centre Zagreb, University of Zagreb, Zagreb, Croatia
| | - Cheryl Arrowsmith
- Structural Genomics Consortium, University of Toronto, Toronto, Ontario, Canada; Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada
| | - Mike Tyers
- Institute for Research in Immunology and Cancer, Université de Montréal, PO Box 6128, Downtown Station, Montreal, QC H3C 3J7, Canada
| | - Ming-Sound Tsao
- Princess Margaret Cancer Centre, University Health Network, University of Toronto, Toronto, Ontario, Canada; Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada; Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada
| | - David Andrews
- Biological Sciences, Sunnybrook Research Institute, Toronto, Ontario, Canada
| | - Rima Al-Awar
- Drug Discovery Program, Ontario Institute for Cancer Research, Ontario, Canada; Department of Pharmacology and Toxicology, University of Toronto, Ontario, Canada.
| | - Igor Stagljar
- Donnelly Centre, University of Toronto, Ontario, Canada; Department of Molecular Genetics, University of Toronto, Ontario, Canada; Department of Biochemistry, University of Toronto, Ontario, Canada; Mediterranean Institute for Life Sciences, Split, Croatia; School of Medicine, University of Split, Split, Croatia.
| |
Collapse
|
10
|
CKAP2L, as an Independent Risk Factor, Closely Related to the Prognosis of Glioma. BIOMED RESEARCH INTERNATIONAL 2021; 2021:5486131. [PMID: 34631884 PMCID: PMC8494202 DOI: 10.1155/2021/5486131] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/24/2020] [Revised: 08/02/2021] [Accepted: 08/05/2021] [Indexed: 11/17/2022]
Abstract
Recent studies have found that cytoskeleton-associated protein 2 like (CKAP2L), an important oncogene, is involved in the biological behavior of many malignant tumors, but its function in the malignant course of glioma has not been confirmed. The main purpose of this study was to clarify the relationship between prognostic clinical characteristics of glioma patients and CKAP2L expression using data collected from the GEPIA, HPA, CGGA, TCGA, and GEO databases. CKAP2L expression was significantly increased in glioma. Further, Kaplan-Meier plots revealed that increased expression of CKAP2L was associated with shorter survival time of glioma patients in datasets retrieved from multiple databases. Cox regression analysis indicated that CKAP2L can serve as an independent risk factor but also has relatively reliable diagnostic value for the prognosis of glioma patients. The results of gene set enrichment analysis suggested that CKAP2L may play a regulatory role through the cell cycle, homologous recombination, and N-glycan biosynthesis cell signaling pathways. Several drugs with potential inhibitory effects on CKAP2L were identified in the CMap database that may have therapeutic effects on glioma. Finally, knockdown of CKAP2L inhibited the proliferation and invasion of cells by reducing the expression level of cell cycle-related proteins. This is the first study to demonstrate that high CKAP2L expression leads to poor prognosis in glioma patients, providing a novel target for diagnosis and treatment of glioma.
Collapse
|
11
|
Receptor tyrosine kinases and cancer: oncogenic mechanisms and therapeutic approaches. Oncogene 2021; 40:4079-4093. [PMID: 34079087 DOI: 10.1038/s41388-021-01841-2] [Citation(s) in RCA: 63] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2021] [Revised: 04/29/2021] [Accepted: 05/13/2021] [Indexed: 02/05/2023]
Abstract
Receptor tyrosine kinases (RTKs) are transmembrane receptors of great clinical interest due to their role in disease, notably cancer. Since their discovery, several mechanisms of RTK dysregulation have been identified, resulting in multiple cancer types displaying 'oncogenic addiction' to RTKs. As a result, RTKs have represented a major class for targeted therapeutics over the past two decades, with numerous small molecule-based tyrosine kinase inhibitor (TKI) therapeutics having been developed and clinically approved for several cancers. However, many of the current RTK inhibitor treatments eventually result in the rapid development of acquired resistance and subsequent tumor relapse. Recent technological advances and tools are being generated for the identification of novel RTK small molecule therapeutics. These newer technologies will be important for the identification of diverse types of RTK inhibitors, targeting both the receptors themselves as well as key cellular factors that play important roles in the RTK signaling cascade.
Collapse
|
12
|
Role of Glycans on Key Cell Surface Receptors That Regulate Cell Proliferation and Cell Death. Cells 2021; 10:cells10051252. [PMID: 34069424 PMCID: PMC8159107 DOI: 10.3390/cells10051252] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2021] [Revised: 05/13/2021] [Accepted: 05/13/2021] [Indexed: 12/12/2022] Open
Abstract
Cells undergo proliferation and apoptosis, migration and differentiation via a number of cell surface receptors, most of which are heavily glycosylated. This review discusses receptor glycosylation and the known roles of glycans on the functions of receptors expressed in diverse cell types. We included growth factor receptors that have an intracellular tyrosine kinase domain, growth factor receptors that have a serine/threonine kinase domain, and cell-death-inducing receptors. N- and O-glycans have a wide range of functions including roles in receptor conformation, ligand binding, oligomerization, and activation of signaling cascades. A better understanding of these functions will enable control of cell survival and cell death in diseases such as cancer and in immune responses.
Collapse
|
13
|
Yan P, Patel HJ, Sharma S, Corben A, Wang T, Panchal P, Yang C, Sun W, Araujo TL, Rodina A, Joshi S, Robzyk K, Gandu S, White JR, de Stanchina E, Modi S, Janjigian YY, Hill EG, Liu B, Erdjument-Bromage H, Neubert TA, Que NLS, Li Z, Gewirth DT, Taldone T, Chiosis G. Molecular Stressors Engender Protein Connectivity Dysfunction through Aberrant N-Glycosylation of a Chaperone. Cell Rep 2021; 31:107840. [PMID: 32610141 PMCID: PMC7372946 DOI: 10.1016/j.celrep.2020.107840] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2020] [Revised: 05/04/2020] [Accepted: 06/09/2020] [Indexed: 01/08/2023] Open
Abstract
Stresses associated with disease may pathologically remodel the proteome by both increasing interaction strength and altering interaction partners, resulting in proteome-wide connectivity dysfunctions. Chaperones play an important role in these alterations, but how these changes are executed remains largely unknown. Our study unveils a specific N-glycosylation pattern used by a chaperone, Glucose-regulated protein 94 (GRP94), to alter its conformational fitness and stabilize a state most permissive for stable interactions with proteins at the plasma membrane. This "protein assembly mutation' remodels protein networks and properties of the cell. We show in cells, human specimens, and mouse xenografts that proteome connectivity is restorable by inhibition of the N-glycosylated GRP94 variant. In summary, we provide biochemical evidence for stressor-induced chaperone-mediated protein mis-assemblies and demonstrate how these alterations are actionable in disease.
Collapse
Affiliation(s)
- Pengrong Yan
- Chemical Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Hardik J Patel
- Chemical Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Sahil Sharma
- Chemical Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Adriana Corben
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; Currently at Mount Sinai Hospital, New York, NY 10029, USA
| | - Tai Wang
- Chemical Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Palak Panchal
- Chemical Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Chenghua Yang
- Chemical Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; Currently at Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Shanghai 200031, China
| | - Weilin Sun
- Chemical Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Thais L Araujo
- Chemical Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Anna Rodina
- Chemical Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Suhasini Joshi
- Chemical Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Kenneth Robzyk
- Chemical Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Srinivasa Gandu
- Chemical Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Julie R White
- Comparative Pathology Laboratory, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Elisa de Stanchina
- Molecular Pharmacology Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Shanu Modi
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Yelena Y Janjigian
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Elizabeth G Hill
- Hollings Cancer Center, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Bei Liu
- Pelotonia Institute for Immuno-Oncology, The Ohio State University, Columbus, OH 43210, USA
| | - Hediye Erdjument-Bromage
- Department of Cell Biology and Kimmel Center for Biology and Medicine of the Skirball Institute, New York University School of Medicine, New York, NY 10016, USA
| | - Thomas A Neubert
- Department of Cell Biology and Kimmel Center for Biology and Medicine of the Skirball Institute, New York University School of Medicine, New York, NY 10016, USA
| | - Nanette L S Que
- Hauptman-Woodward Medical Research Institute, Buffalo, NY 14203, USA
| | - Zihai Li
- Pelotonia Institute for Immuno-Oncology, The Ohio State University, Columbus, OH 43210, USA
| | - Daniel T Gewirth
- Hauptman-Woodward Medical Research Institute, Buffalo, NY 14203, USA
| | - Tony Taldone
- Chemical Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Gabriela Chiosis
- Chemical Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA.
| |
Collapse
|
14
|
Phoomak C, Cui W, Hayman TJ, Yu SH, Zhao P, Wells L, Steet R, Contessa JN. The translocon-associated protein (TRAP) complex regulates quality control of N-linked glycosylation during ER stress. SCIENCE ADVANCES 2021; 7:eabc6364. [PMID: 33523898 PMCID: PMC7810369 DOI: 10.1126/sciadv.abc6364] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/05/2020] [Accepted: 11/24/2020] [Indexed: 05/04/2023]
Abstract
Asparagine (N)-linked glycosylation is required for endoplasmic reticulum (ER) homeostasis, but how this co- and posttranslational modification is maintained during ER stress is unknown. Here, we introduce a fluorescence-based strategy to detect aberrant N-glycosylation in individual cells and identify a regulatory role for the heterotetrameric translocon-associated protein (TRAP) complex. Unexpectedly, cells with knockout of SSR3 or SSR4 subunits restore N-glycosylation over time concurrent with a diminished ER stress transcriptional signature. Activation of ER stress or silencing of the ER chaperone BiP exacerbates or rescues the glycosylation defects, respectively, indicating that SSR3 and SSR4 enable N-glycosylation during ER stress. Protein levels of the SSR3 subunit are ER stress and UBE2J1 dependent, revealing a mechanism that coordinates upstream N-glycosylation proficiency with downstream ER-associated degradation and proteostasis. The fidelity of N-glycosylation is not static in both nontransformed and tumor cells, and the TRAP complex regulates ER glycoprotein quality control under conditions of stress.
Collapse
Affiliation(s)
- Chatchai Phoomak
- Department of Therapeutic Radiology, Yale University School of Medicine, New Haven, CT 06510, USA
| | - Wei Cui
- Department of Therapeutic Radiology, Yale University School of Medicine, New Haven, CT 06510, USA
| | - Thomas J Hayman
- Department of Therapeutic Radiology, Yale University School of Medicine, New Haven, CT 06510, USA
| | - Seok-Ho Yu
- Greenwood Genetic Center, Greenwood, SC 29646, USA
| | - Peng Zhao
- Complex Carbohydrate Research Center, Department of Biochemistry and Molecular Biology, University of Georgia, Athens, GA 30601, USA
| | - Lance Wells
- Complex Carbohydrate Research Center, Department of Biochemistry and Molecular Biology, University of Georgia, Athens, GA 30601, USA
| | | | - Joseph N Contessa
- Department of Therapeutic Radiology, Yale University School of Medicine, New Haven, CT 06510, USA.
- Department of Pharmacology, Yale University School of Medicine, New Haven, CT 06510, USA
| |
Collapse
|
15
|
Wu Y, Peng Y, Guan B, He A, Yang K, He S, Gong Y, Li X, Zhou L. P4HB: A novel diagnostic and prognostic biomarker for bladder carcinoma. Oncol Lett 2020; 21:95. [PMID: 33376528 PMCID: PMC7751343 DOI: 10.3892/ol.2020.12356] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2020] [Accepted: 10/23/2020] [Indexed: 12/15/2022] Open
Abstract
Prolyl 4-hydroxylase, beta polypeptide (P4HB) protein is an endoplasmic reticulum (ER) molecular chaperone protein and has been reported to be overexpressed in multiple tumor types. However, the role of P4HB in bladder cancer (BLCA) has not yet been elucidated. The aim of the present study was to investigate the prognostic value of P4HB and the association between clinicopathological characteristics and P4HB in BLCA. P4HB expression levels were assessed through The Cancer Genome Atlas (TCGA) and Gene Expression Omnibus (GEO) databases, and validated by reverse transcription-quantitative polymerase chain reaction (RT-qPCR) and western blot analysis in BLCA tissues and cells. A total of 69 pairs of tumor and normal samples were used to analyze the expression of P4HB via immunohistochemical staining. A co-expression network and functional enrichment analyses were conducted to investigate the biological function of P4HB in BLCA. The protein-protein interaction (PPI) network was constructed by Search Tool for the Retrieval of Interacting Genes. The results showed that P4HB was highly expressed in BLCA cells and tissues. The area under the curve value for P4HB expression to discriminate between tumor and normal tissues was up to 0.888 (95% CI: 0.801–0.975; P<0.001) and 0.881 (95% CI: 0.825–0.937; P<0.001) in TCGA database and our database, respectively. Furthermore, the expression level of P4HB was an independent risk factor for overall survival (OS) and recurrence-free survival (RFS) by univariate and multivariate analyses. Kaplan-Meier survival analysis demonstrated that high P4HB expression was associated with low OS and RFS. Pathway enrichment analysis suggested that P4HB was involved in protein processing in the endoplasmic reticulum (ER), including N-glycan modification and protein metabolic processes responding to ER stress. PPI analysis revealed that the potential targets of P4HB were mainly involved in posttranslational protein modification and response to ER stress. In conclusion, the expression level of P4HB aid in identifying patients with early-stage BLCA and predicting the prognosis of BLCA. Therefore, P4HB may be a novel diagnostic and prognostic biomarker for BLCA.
Collapse
Affiliation(s)
- Yucai Wu
- Department of Urology, Peking University First Hospital, Xicheng, Beijing 100034, P.R. China.,Institute of Urology, Peking University, Xicheng, Beijing 100034, P.R. China.,National Urological Cancer Center, Peking University, Xicheng, Beijing 100034, P.R. China.,Urogenital Diseases (Male) Molecular Diagnosis and Treatment Center, Peking University, Xicheng, Beijing 100034, P.R. China
| | - Yiji Peng
- Department of Urology, Peking University First Hospital, Xicheng, Beijing 100034, P.R. China.,Institute of Urology, Peking University, Xicheng, Beijing 100034, P.R. China.,National Urological Cancer Center, Peking University, Xicheng, Beijing 100034, P.R. China.,Urogenital Diseases (Male) Molecular Diagnosis and Treatment Center, Peking University, Xicheng, Beijing 100034, P.R. China
| | - Bao Guan
- Department of Urology, Peking University First Hospital, Xicheng, Beijing 100034, P.R. China.,Institute of Urology, Peking University, Xicheng, Beijing 100034, P.R. China.,National Urological Cancer Center, Peking University, Xicheng, Beijing 100034, P.R. China.,Urogenital Diseases (Male) Molecular Diagnosis and Treatment Center, Peking University, Xicheng, Beijing 100034, P.R. China
| | - Anbang He
- Department of Urology, Peking University First Hospital, Xicheng, Beijing 100034, P.R. China.,Institute of Urology, Peking University, Xicheng, Beijing 100034, P.R. China.,National Urological Cancer Center, Peking University, Xicheng, Beijing 100034, P.R. China.,Urogenital Diseases (Male) Molecular Diagnosis and Treatment Center, Peking University, Xicheng, Beijing 100034, P.R. China
| | - Kunlin Yang
- Department of Urology, Peking University First Hospital, Xicheng, Beijing 100034, P.R. China.,Institute of Urology, Peking University, Xicheng, Beijing 100034, P.R. China.,National Urological Cancer Center, Peking University, Xicheng, Beijing 100034, P.R. China.,Urogenital Diseases (Male) Molecular Diagnosis and Treatment Center, Peking University, Xicheng, Beijing 100034, P.R. China
| | - Shiming He
- Department of Urology, Peking University First Hospital, Xicheng, Beijing 100034, P.R. China.,Institute of Urology, Peking University, Xicheng, Beijing 100034, P.R. China.,National Urological Cancer Center, Peking University, Xicheng, Beijing 100034, P.R. China.,Urogenital Diseases (Male) Molecular Diagnosis and Treatment Center, Peking University, Xicheng, Beijing 100034, P.R. China
| | - Yanqing Gong
- Department of Urology, Peking University First Hospital, Xicheng, Beijing 100034, P.R. China.,Institute of Urology, Peking University, Xicheng, Beijing 100034, P.R. China.,National Urological Cancer Center, Peking University, Xicheng, Beijing 100034, P.R. China.,Urogenital Diseases (Male) Molecular Diagnosis and Treatment Center, Peking University, Xicheng, Beijing 100034, P.R. China
| | - Xuesong Li
- Department of Urology, Peking University First Hospital, Xicheng, Beijing 100034, P.R. China.,Institute of Urology, Peking University, Xicheng, Beijing 100034, P.R. China.,National Urological Cancer Center, Peking University, Xicheng, Beijing 100034, P.R. China.,Urogenital Diseases (Male) Molecular Diagnosis and Treatment Center, Peking University, Xicheng, Beijing 100034, P.R. China
| | - Liqun Zhou
- Department of Urology, Peking University First Hospital, Xicheng, Beijing 100034, P.R. China.,Institute of Urology, Peking University, Xicheng, Beijing 100034, P.R. China.,National Urological Cancer Center, Peking University, Xicheng, Beijing 100034, P.R. China.,Urogenital Diseases (Male) Molecular Diagnosis and Treatment Center, Peking University, Xicheng, Beijing 100034, P.R. China
| |
Collapse
|
16
|
Pérez AG, Andrade-Da-Costa J, De Souza WF, De Souza Ferreira M, Boroni M, De Oliveira IM, Freire-Neto CA, Fernandes PV, De Lanna CA, Souza-Santos PT, Morgado-Díaz JA, De-Freitas-Junior JCM. N‑glycosylation and receptor tyrosine kinase signaling affect claudin‑3 levels in colorectal cancer cells. Oncol Rep 2020; 44:1649-1661. [PMID: 32945502 PMCID: PMC7448416 DOI: 10.3892/or.2020.7727] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2019] [Accepted: 07/14/2020] [Indexed: 12/14/2022] Open
Abstract
Changes in protein levels in different components of the apical junctional complex occur in colorectal cancer (CRC). Claudin-3 is one of the main constituents of tight junctions, and its overexpression can increase the paracellular flux of macromolecules, as well as the malignant potential of CRC cells. The aim of this study was to investigate the molecular mechanisms involved in the regulation of claudin-3 and its prognostic value in CRC. In silico evaluation in each of the CRC consensus molecular subtypes (CMSs) revealed that high expression levels of CLDN3 (gene encoding claudin-3) in CMS2 and CMS3 worsened the patients' long-term survival, whereas a decrease in claudin-3 levels concomitant with a reduction in phosphorylation levels of epidermal growth factor receptor (EGFR) and insulin-like growth factor 1 receptor (IGF1R) could be achieved by inhibiting N-glycan biosynthesis in CRC cells. We also observed that specific inactivation of these receptor tyrosine kinases (RTKs) led to a decrease in claudin-3 levels, and this regulation seems to be mediated by phospholipase C (PLC) and signal transducer and activator of transcription 3 (STAT3) in CRC cells. RTKs are modulated by their N-linked glycans, and inhibition of N-glycan biosynthesis decreased the claudin-3 levels; therefore, we evaluated the correlation between N-glycogenes and CLDN3 expression levels in each of the CRC molecular subtypes. The CMS1 (MSI immune) subtype concomitantly exhibited low expression levels of CLDN3 and N-glycogenes (MGAT5, ST6GAL1, and B3GNT8), whereas CMS2 (canonical) exhibited high gene expression levels of CLDN3 and N-glycogenes (ST6GAL1 and B3GNT8). A robust positive correlation was also observed between CLDN3 and B3GNT8 expression levels in all CMSs. These results support the hypothesis of a mechanism integrating RTK signaling and N-glycosylation for the regulation of claudin-3 levels in CRC, and they suggest that CLDN3 expression can be used to predict the prognosis of patients identified as CMS2 or CMS3.
Collapse
Affiliation(s)
- Amelia G Pérez
- Cellular and Molecular Oncobiology Program, National Cancer Institute (INCA), Rio de Janeiro, RJ 20231‑050, Brazil
| | - Jéssica Andrade-Da-Costa
- Cellular and Molecular Oncobiology Program, National Cancer Institute (INCA), Rio de Janeiro, RJ 20231‑050, Brazil
| | - Waldemir F De Souza
- Cellular and Molecular Oncobiology Program, National Cancer Institute (INCA), Rio de Janeiro, RJ 20231‑050, Brazil
| | - Michelle De Souza Ferreira
- Cellular and Molecular Oncobiology Program, National Cancer Institute (INCA), Rio de Janeiro, RJ 20231‑050, Brazil
| | - Mariana Boroni
- Bioinformatics and Computational Biology Laboratory, National Cancer Institute (INCA), Rio de Janeiro, RJ 20231‑050, Brazil
| | - Ivanir M De Oliveira
- Pathology Division, National Cancer Institute (INCA), Rio de Janeiro, RJ 20231‑050, Brazil
| | - Carlos A Freire-Neto
- Cellular and Molecular Oncobiology Program, National Cancer Institute (INCA), Rio de Janeiro, RJ 20231‑050, Brazil
| | - Priscila V Fernandes
- Pathology Division, National Cancer Institute (INCA), Rio de Janeiro, RJ 20231‑050, Brazil
| | - Cristóvão A De Lanna
- Bioinformatics and Computational Biology Laboratory, National Cancer Institute (INCA), Rio de Janeiro, RJ 20231‑050, Brazil
| | | | - José A Morgado-Díaz
- Cellular and Molecular Oncobiology Program, National Cancer Institute (INCA), Rio de Janeiro, RJ 20231‑050, Brazil
| | | |
Collapse
|
17
|
Alam SMD, Tsukamoto Y, Ogawa M, Senoo Y, Ikeda K, Tashima Y, Takeuchi H, Okajima T. N-Glycans on EGF domain-specific O-GlcNAc transferase (EOGT) facilitate EOGT maturation and peripheral endoplasmic reticulum localization. J Biol Chem 2020; 295:8560-8574. [PMID: 32376684 DOI: 10.1074/jbc.ra119.012280] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2019] [Revised: 04/30/2020] [Indexed: 02/03/2023] Open
Abstract
Epidermal growth factor (EGF) domain-specific O-GlcNAc transferase (EOGT) is an endoplasmic reticulum (ER)-resident protein that modifies EGF repeats of Notch receptors and thereby regulates Delta-like ligand-mediated Notch signaling. Several EOGT mutations that may affect putative N-glycosylation consensus sites are recorded in the cancer database, but the presence and function of N-glycans in EOGT have not yet been characterized. Here, we identified N-glycosylation sites in mouse EOGT and elucidated their molecular functions. Three predicted N-glycosylation consensus sequences on EOGT are highly conserved among mammalian species. Within these sites, we found that Asn-263 and Asn-354, but not Asn-493, are modified with N-glycans. Lectin blotting, endoglycosidase H digestion, and MS analysis revealed that both residues are modified with oligomannose N-glycans. Loss of an individual N-glycan on EOGT did not affect its endoplasmic reticulum (ER) localization, enzyme activity, and ability to O-GlcNAcylate Notch1 in HEK293T cells. However, simultaneous substitution of both N-glycosylation sites affected both EOGT maturation and expression levels without an apparent change in enzymatic activity, suggesting that N-glycosylation at a single site is sufficient for EOGT maturation and expression. Accordingly, a decrease in O-GlcNAc stoichiometry was observed in Notch1 co-expressed with an N263Q/N354Q variant compared with WT EOGT. Moreover, the N263Q/N354Q variant exhibited altered subcellular distribution within the ER in HEK293T cells, indicating that N-glycosylation of EOGT is required for its ER localization at the cell periphery. These results suggest critical roles of N-glycans in sustaining O-GlcNAc transferase function both by maintaining EOGT levels and by ensuring its proper subcellular localization in the ER.
Collapse
Affiliation(s)
- Sayad Md Didarul Alam
- Department of Molecular Biochemistry, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Yohei Tsukamoto
- Department of Molecular Biochemistry, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Mitsutaka Ogawa
- Department of Molecular Biochemistry, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Yuya Senoo
- Department of Molecular Biochemistry, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Kazutaka Ikeda
- Department of Molecular Biochemistry, Nagoya University Graduate School of Medicine, Nagoya, Japan.,RIKEN, Center for Integrative Medical Sciences, Suehiro-cho, Tsurumi, Yokohama, Japan
| | - Yuko Tashima
- Department of Molecular Biochemistry, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Hideyuki Takeuchi
- Department of Molecular Biochemistry, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Tetsuya Okajima
- Department of Molecular Biochemistry, Nagoya University Graduate School of Medicine, Nagoya, Japan
| |
Collapse
|
18
|
Markouli M, Strepkos D, Papavassiliou AG, Piperi C. Targeting of endoplasmic reticulum (ER) stress in gliomas. Pharmacol Res 2020; 157:104823. [PMID: 32305494 DOI: 10.1016/j.phrs.2020.104823] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/31/2020] [Revised: 03/29/2020] [Accepted: 04/06/2020] [Indexed: 12/11/2022]
Abstract
Gliomas remain a group of malignant brain tumors with dismal prognosis and limited treatment options with molecular mechanisms being constantly investigated. The past decade, extracellular stress and intracellular DNA damage have been shown to disturb proteostasis leading to Endoplasmic Reticulum (ER) stress that is implicated in the regulation of gene expression and the pathogenesis of several tumor types, including gliomas. Upon ER stress induction, neoplastic cells activate the adaptive mechanism of unfolded protein response (UPR), an integrated signaling system that either restores ER homeostasis or induces cell apoptosis. Recently, the manipulation of the UPR has emerged as a new therapeutic target in glioma treatment. General UPR activators or selective GRP78, ATF6 and PERK inducers have been detected to modulate cell proliferation and induce apoptosis of glioma cells. At the same time, target-specific UPR inhibitors and small molecule proteostasis disruptors, work in reverse to increase misfolded proteins and cause a dysregulation in protein maturation and sorting, thus preventing the growth of neoplastic cells. Herein, we discuss the pathogenic implication of ER stress in gliomas onset and progression, providing an update on the current UPR modifying agents that can be potentially used in glioma treatment.
Collapse
Affiliation(s)
- Mariam Markouli
- Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece
| | - Dimitrios Strepkos
- Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece
| | - Athanasios G Papavassiliou
- Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece.
| | - Christina Piperi
- Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece.
| |
Collapse
|
19
|
Abstract
Objective: To summarize the abnormal location of FLT3 caused by different glycosylation status which further leads to the distinguishing signaling pathways and discuss targeting on FLT3 glycosylation by drugs reported in recent literatures. Methods: We review FLT3 glycosylation in endoplasmic reticulum. The abnormal signal of mutant FLT3 with different glycosylation status is discussed. We also address potential FLT3 glycosylation-targeting strategies for the treatment. Results: Inhibition of FLT3 mutant cells by drugs reported in recent literatures involves the influence of glycosylation of FLT3: 2-deoxy-D-glucose, Tunicamycin and Fluvastatin are reported to inhibit N-glycosylation of FLT3; Pim-1 inhibitors are proved to block the inhibition of Pim-1 on FLT3 Oglycosylation; HSP90 inhibitors and Tyrosine Kinase Inhibitors are shown to increase fully glycosylated form of FLT3. Discussion: The FMS-like tyrosine kinase 3 (FLT3) gene expressed only in CD34+ progenitor cells in bone marrow is located on chromosome 13q12 encoding FLT3 protein. FLT3 is initially synthesized as a 110 KD protein, which glycosylated in the endoplasmic reticulum to a 130 KD immature protein rich in mannose, and further processed into a mature 160 KD protein in the Golgi apparatus, which could be transferred to the cell surface. Therapy targeting on FLT3 glycosylation is a promising direction for AML treatment. Conclusions: The abnormal location of FLT3 caused by different glycosylation status leads to the distinguishing signaling pathways. Targeting on FLT3 glycosylation may provide a new perspective for therapeutic strategies. Abbreviations: ABCG2: ATP-binding cassette transporter breast cancer resistance protein; ATF: activating transcription factor; AML: acute myeloid leukemia; CHOP: CCAAT-enhancer-binding protein homologous protein; 2-DG: 2-deoxy-D-glucose; EFS: event free survival; EPO: erythropoietin; EPOR: erythropoietin receptor; ERS: endoplasmic reticulum stress; FLT3: FMS-like tyrosine kinase 3; GPI: glycosylphosphatidylinositol; HSP: heat shock protein; ITD: internal tandem duplication; IRE1a: inositol-requiring enzyme 1 alpha; JNK: c-Jun N-terminal kinase; JMD: juxtamembrane domain; JAK: janus kinase; MAPK/ERK: mitogen activated protein kinase/extracellular signal-regulated protein kinase; OS: overall survival; PI3K/AKT: phosphatidylinositide 3-kinases/protein kinase B; PERK: RNA-activated protein kinase-like endoplasmic reticulum kinase; Pgp: P-glycoprotein; PTX3: human pentraxin-3; STAT: signal transducer and activator of transcriptions; TKD: tyrosine-kinase domain; TKI: tyrosine kinase inhibitor; TM: Tunicamycin; UPR: unfolded protein reaction.
Collapse
Affiliation(s)
- Xiaoli Hu
- Department of Hematology, RenJi Hospital, School of Medicine, Shanghai Jiao Tong University , Shanghai , People's Republic of China
| | - Fangyuan Chen
- Department of Hematology, RenJi Hospital, School of Medicine, Shanghai Jiao Tong University , Shanghai , People's Republic of China
| |
Collapse
|
20
|
Takahashi S. Mutations of FLT3 receptor affect its surface glycosylation, intracellular localization, and downstream signaling. Leuk Res Rep 2019; 13:100187. [PMID: 31853441 PMCID: PMC6911968 DOI: 10.1016/j.lrr.2019.100187] [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] [Received: 07/30/2019] [Revised: 10/29/2019] [Accepted: 11/23/2019] [Indexed: 11/29/2022] Open
Abstract
This review describes the effects of FLT3 mutations that alter its intracellular localization and modify its glycosylation, leading to differences in downstream signaling pathways. The most common type of FLT3 mutation, internal tandem duplication (FLT3-ITD), leads to localization in the endoplasmic reticulum and constitutive strong activation of STAT5. In contrast, the ligand-activated FLT3-wild type is mainly expressed on the cell surface and activates MAP kinases. Based on these backgrounds, several reports have demonstrated that glycosylation inhibitors are effective for inhibition of FLT3-ITD expression and intracellular localization. The general subcellular localization regulatory mechanisms for receptor tyrosine kinases are also discussed.
Collapse
Affiliation(s)
- Shinichiro Takahashi
- Division of Laboratory Medicine, Faculty of Medicine, Tohoku Medical and Pharmaceutical University, 1-15-1 Fukumuro, Miyagino-ku, Sendai 983-8536, Japan
| |
Collapse
|
21
|
Lee N, Jang WJ, Seo JH, Lee S, Jeong CH. 2-Deoxy-d-Glucose-Induced Metabolic Alteration in Human Oral Squamous SCC15 Cells: Involvement of N-Glycosylation of Axl and Met. Metabolites 2019; 9:E188. [PMID: 31533338 PMCID: PMC6780519 DOI: 10.3390/metabo9090188] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2019] [Revised: 09/07/2019] [Accepted: 09/12/2019] [Indexed: 12/20/2022] Open
Abstract
One of the most prominent hallmarks of cancer cells is their dependency on the glycolytic pathway for energy production. As a potent inhibitor of glycolysis, 2-deoxy-d-glucose (2DG) has been proposed for cancer treatment and extensively investigated in clinical studies. Moreover, 2DG has been reported to interfere with other biological processes including glycosylation. To further understand the overall effect of and metabolic alteration by 2DG, we performed biochemical and metabolomics analyses on oral squamous cell carcinoma cell lines. In this study, we found that 2DG more effectively reduced glucose consumption and lactate level in SCC15 cells than in SCC4 cells, which are less dependent on glycolysis. Coincidentally, 2DG impaired N-linked glycosylation of the key oncogenic receptors Axl and Met in SCC15 cells, thereby reducing the cell viability and colony formation ability. The impaired processes of glycolysis and N-linked glycosylation were restored by exogenous addition of pyruvate and mannose, respectively. Additionally, our targeted metabolomics analysis revealed significant alterations in the metabolites, including amino acids, biogenic amines, glycerophospholipids, and sphingolipids, caused by the impairment of glycolysis and N-linked glycosylation. These observations suggest that alterations of these metabolites may be responsible for the phenotypic and metabolic changes in SCC15 cells induced by 2DG. Moreover, our data suggest that N-linked glycosylation of Axl and Met may contribute to the maintenance of cancer properties in SCC15 cells. Further studies are needed to elucidate the roles of these altered metabolites to provide novel therapeutic targets for treating human oral cancer.
Collapse
Affiliation(s)
- Naeun Lee
- College of Pharmacy, Keimyung University, Daegu 42601, Korea.
| | - Won-Jun Jang
- College of Pharmacy, Keimyung University, Daegu 42601, Korea.
| | - Ji Hae Seo
- Department of Biochemistry, Keimyung University School of Medicine, Daegu 42601, Korea.
| | - Sooyeun Lee
- College of Pharmacy, Keimyung University, Daegu 42601, Korea.
| | - Chul-Ho Jeong
- College of Pharmacy, Keimyung University, Daegu 42601, Korea.
| |
Collapse
|
22
|
Cole DW, Svider PF, Shenouda KG, Lee PB, Yoo NG, McLeod TM, Mutchnick SA, Yoo GH, Kaufman RJ, Callaghan MU, Fribley AM. Targeting the unfolded protein response in head and neck and oral cavity cancers. Exp Cell Res 2019; 382:111386. [PMID: 31075256 DOI: 10.1016/j.yexcr.2019.04.007] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2018] [Revised: 03/25/2019] [Accepted: 04/05/2019] [Indexed: 12/18/2022]
Abstract
Many FDA-approved anti-cancer therapies, targeted toward a wide array of molecular targets and signaling networks, have been demonstrated to activate the unfolded protein response (UPR). Despite a critical role for UPR signaling in the apoptotic execution of cancer cells by many of these compounds, the authors are currently unaware of any instance whereby a cancer drug was developed with the UPR as the intended target. With the essential role of the UPR as a driving force in the genesis and maintenance of the malignant phenotype, a great number of pre-clinical studies have surged into the medical literature describing the ability of dozens of compounds to induce UPR signaling in a myriad of cancer models. The focus of the current work is to review the literature and explore the role of the UPR as a mediator of chemotherapy-induced cell death in squamous cell carcinomas of the head and neck (HNSCC) and oral cavity (OCSCC), with an emphasis on preclinical studies.
Collapse
Affiliation(s)
- Daniel W Cole
- Department of Otolaryngology - Head and Neck Surgery, Wayne State University School of Medicine, Detroit, MI, USA
| | - Peter F Svider
- Department of Otolaryngology - Head and Neck Surgery, Wayne State University School of Medicine, Detroit, MI, USA
| | - Kerolos G Shenouda
- Department of Otolaryngology - Head and Neck Surgery, Wayne State University School of Medicine, Detroit, MI, USA
| | - Paul B Lee
- Oakland University William Beaumont School of Medicine, Rochester Hills, Michigan, USA
| | - Nicholas G Yoo
- Department of Otolaryngology - Head and Neck Surgery, Wayne State University School of Medicine, Detroit, MI, USA
| | - Thomas M McLeod
- Department of Otolaryngology - Head and Neck Surgery, Wayne State University School of Medicine, Detroit, MI, USA
| | - Sean A Mutchnick
- Department of Otolaryngology - Head and Neck Surgery, Wayne State University School of Medicine, Detroit, MI, USA
| | - George H Yoo
- Department of Otolaryngology - Head and Neck Surgery, Wayne State University School of Medicine, Detroit, MI, USA; Barbara Ann Karmanos Cancer Institute, Detroit, MI, USA
| | - Randal J Kaufman
- Degenerative Diseases Program, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, USA
| | - Michael U Callaghan
- Carman and Ann Adams Department of Pediatrics, Wayne State University School of Medicine, Detroit, MI, USA; Children's Hospital of Michigan, Detroit Medical Center, Detroit, MI, USA
| | - Andrew M Fribley
- Department of Otolaryngology - Head and Neck Surgery, Wayne State University School of Medicine, Detroit, MI, USA; Carman and Ann Adams Department of Pediatrics, Wayne State University School of Medicine, Detroit, MI, USA; Barbara Ann Karmanos Cancer Institute, Detroit, MI, USA; Children's Hospital of Michigan, Detroit Medical Center, Detroit, MI, USA; Molecular Therapeutics Program, Barbara Ann Karmanos Cancer Institute, Detroit, MI, USA.
| |
Collapse
|
23
|
Li S, Cao L, Wang X, Wang F, Wang L, Jiang R. Neuron-Specific Enolase Is an Independent Prognostic Factor in Resected Lung Adenocarcinoma Patients with Anaplastic Lymphoma Kinase Gene Rearrangements. Med Sci Monit 2019; 25:675-690. [PMID: 30673691 PMCID: PMC6353286 DOI: 10.12659/msm.913054] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/02/2022] Open
Abstract
Background An extensive body of research reveals the clinical value of serum tumor markers in lung cancer patients, including carcinoembryonic antigen (CEA), squamous cell carcinoma antigen (SCCA), cytokeratin-19 fragments (Cyfra21-1), and neuron-specific enolase (NSE), but little is known about the clinical properties of these serum tumor markers in anaplastic lymphoma kinase (ALK)-positive lung cancer patients. Matreial/Methods We retrospectively analyzed 54 patients harboring ALK rearrangements and 520 patients without ALK rearrangements, and all these patients were treated exclusively by surgery between 2011 and 2016. Results NSE level (P=0.007 for OS) was identified as an independent prognostic factor among patients with resected ALK-positive adenocarcinoma of the lung. Conclusions A high level of NSE is associated with worse outcome among resected lung adenocarcinoma patients harboring ALK rearrangements.
Collapse
Affiliation(s)
- Shouying Li
- Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, China (mainland).,Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin, China (mainland).,Department of Thoracic Oncology, Tianjin Lung Cancer Center, Tianjin Cancer Institute and Hospital, Tianjin Medical University, Tianjin, China (mainland)
| | - Lianjing Cao
- Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, China (mainland).,Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin, China (mainland).,Department of Thoracic Oncology, Tianjin Lung Cancer Center, Tianjin Cancer Institute and Hospital, Tianjin Medical University, Tianjin, China (mainland)
| | - Xinyue Wang
- Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, China (mainland).,Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin, China (mainland).,Department of Thoracic Oncology, Tianjin Lung Cancer Center, Tianjin Cancer Institute and Hospital, Tianjin Medical University, Tianjin, China (mainland)
| | - Fan Wang
- Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, China (mainland).,Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin, China (mainland).,Department of Thoracic Oncology, Tianjin Lung Cancer Center, Tianjin Cancer Institute and Hospital, Tianjin Medical University, Tianjin, China (mainland)
| | - Liuchun Wang
- Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, China (mainland).,Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin, China (mainland).,Department of Thoracic Oncology, Tianjin Lung Cancer Center, Tianjin Cancer Institute and Hospital, Tianjin Medical University, Tianjin, China (mainland)
| | - Richeng Jiang
- Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, China (mainland).,Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin, China (mainland).,Department of Thoracic Oncology, Tianjin Lung Cancer Center, Tianjin Cancer Institute and Hospital, Tianjin Medical University, Tianjin, China (mainland)
| |
Collapse
|
24
|
Lopez Sambrooks C, Baro M, Quijano A, Narayan A, Cui W, Greninger P, Egan R, Patel A, Benes CH, Saltzman WM, Contessa JN. Oligosaccharyltransferase Inhibition Overcomes Therapeutic Resistance to EGFR Tyrosine Kinase Inhibitors. Cancer Res 2018; 78:5094-5106. [PMID: 30026325 PMCID: PMC6125176 DOI: 10.1158/0008-5472.can-18-0505] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2018] [Revised: 05/14/2018] [Accepted: 07/09/2018] [Indexed: 12/24/2022]
Abstract
Asparagine (N)-linked glycosylation is a posttranslational modification essential for the function of complex transmembrane proteins. However, targeting glycosylation for cancer therapy has not been feasible due to generalized effects on all glycoproteins. Here, we perform sensitivity screening of 94 lung cancer cell lines using NGI-1, a small-molecule inhibitor of the oligosaccharyltransferase (OST) that partially disrupts N-linked glycosylation, and demonstrate a selective loss of tumor cell viability. This screen revealed NGI-1 sensitivity in just 11 of 94 (12%) cell lines, with a significant correlation between OST and EGFR inhibitors. In EGFR-mutant non-small cell lung cancer with EGFR tyrosine kinase inhibitor (TKI) resistance (PC9-GR, HCC827-GR, and H1975-OR), OST inhibition maintained its ability to induce cell-cycle arrest and a proliferative block. Addition of NGI-1 to EGFR TKI treatment was synthetic lethal in cells resistant to gefitinib, erlotinib, or osimertinib. OST inhibition invariably disrupted EGFR N-linked glycosylation and reduced activation of receptors either with or without the T790M TKI resistance mutation. OST inhibition also dissociated EGFR signaling from other coexpressed receptors like MET via altered receptor compartmentalization. Translation of this approach to preclinical models was accomplished through synthesis and delivery of NGI-1 nanoparticles, confirmation of in vivo activity through molecular imaging, and demonstration of significant tumor growth delay in TKI-resistant HCC827 and H1975 xenografts. This therapeutic strategy breaks from kinase-targeted approaches and validates N-linked glycosylation as an effective target in tumors driven by glycoprotein signaling.Significance:EGFR-mutant NSCLC is incurable despite the marked sensitivity of these tumors to EGFR TKIs. These findings identify N-linked glycosylation, a posttranslational modification common to EGFR and other oncogenic signaling proteins, as an effective therapeutic target that enhances tumor responses for EGFR-mutant NSCLC. Cancer Res; 78(17); 5094-106. ©2018 AACR.
Collapse
Affiliation(s)
| | - Marta Baro
- Department of Therapeutic Radiology, Yale University, New Haven, Connecticut
| | - Amanda Quijano
- Department of Biomedical Engineering, Yale University, New Haven, Connecticut
| | - Azeet Narayan
- Department of Therapeutic Radiology, Yale University, New Haven, Connecticut
| | - Wei Cui
- Department of Therapeutic Radiology, Yale University, New Haven, Connecticut
| | - Patricia Greninger
- The Center for Molecular Therapeutics, Massachusetts General Hospital, Boston, Massachusetts
- Department of Medicine, Harvard Medical School, Boston, Massachusetts
| | - Regina Egan
- The Center for Molecular Therapeutics, Massachusetts General Hospital, Boston, Massachusetts
- Department of Medicine, Harvard Medical School, Boston, Massachusetts
| | - Abhijit Patel
- Department of Therapeutic Radiology, Yale University, New Haven, Connecticut
| | - Cyril H Benes
- The Center for Molecular Therapeutics, Massachusetts General Hospital, Boston, Massachusetts
- Department of Medicine, Harvard Medical School, Boston, Massachusetts
| | - W Mark Saltzman
- Department of Biomedical Engineering, Yale University, New Haven, Connecticut
| | - Joseph N Contessa
- Department of Therapeutic Radiology, Yale University, New Haven, Connecticut.
- Department of Pharmacology, Yale University, New Haven, Connecticut
| |
Collapse
|
25
|
Editing N-Glycan Site Occupancy with Small-Molecule Oligosaccharyltransferase Inhibitors. Cell Chem Biol 2018; 25:1231-1241.e4. [PMID: 30078634 DOI: 10.1016/j.chembiol.2018.07.005] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2018] [Revised: 04/12/2018] [Accepted: 07/03/2018] [Indexed: 12/22/2022]
Abstract
The oligosaccharyltransferase (OST) is a multisubunit enzyme complex that N-glycosylates proteins in the secretory pathway and is considered to be constitutive and unregulated. However, small-molecule OST inhibitors such as NGI-1 provide a pharmacological approach for regulating N-linked glycosylation. Herein we design cell models with knockout of each OST catalytic subunit (STT3A or STT3B) to screen the activity of NGI-1 and its analogs. We show that NGI-1 targets the function of both STT3A and STT3B and use structure-activity relationships to guide synthesis of catalytic subunit-specific inhibitors. Using this approach, pharmacophores that increase STT3B selectivity are characterized and an STT3B-specific inhibitor is identified. This inhibitor has discrete biological effects on endogenous STT3B target proteins such as COX2 but does not activate the cellular unfolded protein response. Together this work demonstrates that subsets of glycoproteins can be regulated through pharmacologic inhibition of N-linked glycosylation.
Collapse
|
26
|
Baro M, Lopez Sambrooks C, Quijano A, Saltzman WM, Contessa J. Oligosaccharyltransferase Inhibition Reduces Receptor Tyrosine Kinase Activation and Enhances Glioma Radiosensitivity. Clin Cancer Res 2018; 25:784-795. [PMID: 29967251 DOI: 10.1158/1078-0432.ccr-18-0792] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2018] [Revised: 05/21/2018] [Accepted: 06/27/2018] [Indexed: 12/31/2022]
Abstract
PURPOSE Parallel signaling reduces the effects of receptor tyrosine kinase (RTK)-targeted therapies in glioma. We hypothesized that inhibition of protein N-linked glycosylation, an endoplasmic reticulum co- and posttranslational modification crucial for RTK maturation and activation, could provide a new therapeutic approach for glioma radiosensitization.Experimental Design: We investigated the effects of a small-molecule inhibitor of the oligosaccharyltransferase (NGI-1) on EGFR family receptors, MET, PDGFR, and FGFR1. The influence of glycosylation state on tumor cell radiosensitivity, chemotherapy-induced cell toxicity, DNA damage, and cell-cycle arrest were determined and correlated with glioma cell receptor expression profiles. The effects of NGI-1 on xenograft tumor growth were tested using a nanoparticle formulation validated by in vivo molecular imaging. A mechanistic role for RTK signaling was evaluated through the expression of a glycosylation-independent CD8-EGFR chimera. RESULTS NGI-1 reduced glycosylation, protein levels, and activation of most RTKs. NGI-1 also enhanced the radiosensitivity and cytotoxic effects of chemotherapy in those glioma cells with elevated ErbB family activation, but not in cells without high levels of RTK activation. NGI-1 radiosensitization was associated with increases in both DNA damage and G1 cell-cycle arrest. Combined treatment of glioma xenografts with fractionated radiotherapy and NGI-1 significantly reduced tumor growth compared with controls. Expression of the CD8-EGFR eliminated the effects of NGI-1 on G1 arrest, DNA damage, and cellular radiosensitivity, identifying RTK inhibition as the principal mechanism for the NGI-1 effect. CONCLUSIONS This study suggests that oligosaccharyltransferase inhibition with NGI-1 is a novel approach to radiosensitize malignant gliomas with enhanced RTK signaling.See related commentary by Wahl and Lawrence, p. 455.
Collapse
Affiliation(s)
- Marta Baro
- Department of Therapeutic Radiology, Yale University, New Haven, Connecticut
| | | | - Amanda Quijano
- Department of Biomedical Engineering, Yale University, New Haven, Connecticut
| | - W Mark Saltzman
- Department of Biomedical Engineering, Yale University, New Haven, Connecticut
| | - Joseph Contessa
- Department of Therapeutic Radiology, Yale University, New Haven, Connecticut. .,Department of Pharmacology, Yale University, New Haven, Connecticut
| |
Collapse
|
27
|
Bennett DC, Cazet A, Charest J, Contessa JN. MPDU1 regulates CEACAM1 and cell adhesion in vitro and in vivo. Glycoconj J 2018; 35:265-274. [PMID: 29671116 DOI: 10.1007/s10719-018-9819-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2018] [Revised: 03/21/2018] [Accepted: 03/22/2018] [Indexed: 11/27/2022]
Abstract
N-linked glycosylation (NLG) is a co-translational modification that is essential for the folding, stability, and trafficking of transmembrane (TM) and secretory glycoproteins. Efficient NLG requires the stepwise synthesis and en bloc transfer of a 14-sugar carbohydrate known as a lipid-linked oligosaccharide (LLO). The genetics of LLO biosynthesis have been established in yeast and Chinese hamster systems, but human models of LLO biosynthesis are lacking. In this study we report that Kato III human gastric cancer cells represent a model of deficient LLO synthesis, possessing a homozygous deletion of the LLO biosynthesis factor, MPDU1. Kato III cells lacking MPDU1 have all the hallmarks of a glycosylation-deficient cell line, including altered sensitivity to lectins and the formation of truncated LLOs. Analysis of transcription using an expression microarray and protein levels using a proteome antibody array reveal changes in the expression of several membrane proteins, including the metalloprotease ADAM-15 and the cell adhesion molecule CEACAM1. Surprisingly, the restoration of MPDU1 expression in Kato III cells demonstrated a clear phenotype of increased cell-cell adhesion, a finding that was confirmed in vivo through analysis of tumor xenografts. These experiments also confirmed that protein levels of CEACAM-1, which functions in cell adhesion, is dependent on LLO biosynthesis in vivo. Kato III cells and the MPDU1-rescued Kato IIIM cells therefore provide a novel model to examine the consequences of defective LLO biosynthesis both in vitro and in vivo.
Collapse
Affiliation(s)
- Daniel C Bennett
- Department of Therapeutic Radiology, Yale School of Medicine, New Haven, CT, 06510, USA
| | - Aurelie Cazet
- Department of Therapeutic Radiology, Yale School of Medicine, New Haven, CT, 06510, USA
| | - Jon Charest
- Department of Therapeutic Radiology, Yale School of Medicine, New Haven, CT, 06510, USA
| | - Joseph N Contessa
- Department of Therapeutic Radiology, Yale School of Medicine, New Haven, CT, 06510, USA.
| |
Collapse
|
28
|
Tsitsipatis D, Jayavelu AK, Müller JP, Bauer R, Schmidt-Arras D, Mahboobi S, Schnöder TM, Heidel F, Böhmer FD. Synergistic killing of FLT3ITD-positive AML cells by combined inhibition of tyrosine-kinase activity and N-glycosylation. Oncotarget 2018; 8:26613-26624. [PMID: 28460451 PMCID: PMC5432283 DOI: 10.18632/oncotarget.15772] [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] [Subscribe] [Scholar Register] [Received: 05/11/2016] [Accepted: 02/16/2017] [Indexed: 01/05/2023] Open
Abstract
Fms-like tyrosine kinase 3 (FLT3) with internal tandem duplications (ITD) is a major oncoprotein in acute myeloid leukemia (AML), and confers an unfavorable prognosis. Interference with FLT3ITD signaling is therefore pursued as a promising therapeutic strategy. In this study we show that abrogation of FLT3ITD glycoprotein maturation using low doses of the N-glycosylation inhibitor tunicamycin has anti-proliferative and pro-apoptotic effects on FLT3ITD-expressing human and murine cell lines. This effect is mediated in part by arresting FLT3ITD in an underglycosylated state and thereby attenuating FLT3ITD-driven AKT and ERK signaling. In addition, tunicamycin caused pronounced endoplasmatic reticulum stress and apoptosis through activation of protein kinase RNA-like endoplasmic reticulum kinase (PERK) and activation of the gene encoding CCAAT-enhancer-binding protein homologous protein (CHOP). PERK inhibition with a small molecule attenuated CHOP induction and partially rescued cells from apoptosis. Combination of tunicamycin with potent FLT3ITD kinase inhibitors caused synergistic cell killing, which was highly selective for cell lines and primary AML cells expressing FLT3ITD. Although tunicamycin is currently not a clinically applicable drug, we propose that mild inhibition of N-glycosylation may have therapeutic potential in combination with FLT3 kinase inhibitors for FLT3ITD-positive AML.
Collapse
Affiliation(s)
- Dimitrios Tsitsipatis
- Institute of Molecular Cell Biology, CMB, Jena University Hospital, Jena, Germany.,Current address: Institute of Nutrition, Department of Nutrigenomics, Friedrich-Schiller-University, Jena, Germany
| | - Ashok Kumar Jayavelu
- Institute of Molecular Cell Biology, CMB, Jena University Hospital, Jena, Germany.,Current address: Max-Planck Institute of Biochemistry, Department of Proteomics and Signal Transduction, Martinsried, Germany
| | - Jörg P Müller
- Institute of Molecular Cell Biology, CMB, Jena University Hospital, Jena, Germany
| | - Reinhard Bauer
- Institute of Molecular Cell Biology, CMB, Jena University Hospital, Jena, Germany
| | - Dirk Schmidt-Arras
- Institute of Biochemistry, Christian-Albrechts-University Kiel, Kiel, Germany
| | - Siavosh Mahboobi
- Institute of Pharmacy, Faculty of Chemistry and Pharmacy, University of Regensburg, Regensburg, Germany
| | - Tina M Schnöder
- Innere Medizin II, Hämatologie und Onkologie, Universitätsklinikum Jena, Jena, Germany.,Leibniz Institute on Aging, Fritz-Lipmann-Institute, Jena, Germany
| | - Florian Heidel
- Innere Medizin II, Hämatologie und Onkologie, Universitätsklinikum Jena, Jena, Germany.,Leibniz Institute on Aging, Fritz-Lipmann-Institute, Jena, Germany
| | - Frank-D Böhmer
- Institute of Molecular Cell Biology, CMB, Jena University Hospital, Jena, Germany
| |
Collapse
|
29
|
Vašíčková K, Horak P, Vaňhara P. TUSC3: functional duality of a cancer gene. Cell Mol Life Sci 2018; 75:849-857. [PMID: 28929175 PMCID: PMC11105401 DOI: 10.1007/s00018-017-2660-4] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2017] [Accepted: 09/13/2017] [Indexed: 02/06/2023]
Abstract
Two decades ago, following a systematic screening of LOH regions on chromosome 8p22, TUSC3 has been identified as a candidate tumor suppressor gene in ovarian, prostate and pancreatic cancers. Since then, a growing body of evidence documented its clinical importance in various other types of cancers, and first initial insights into its molecular function and phenotypic effects have been gained, though the precise role of TUSC3 in different cancers remains unclear. As a part of the oligosaccharyltransferase complex, TUSC3 localizes to the endoplasmic reticulum and functions in final steps of N-glycosylation of proteins, while its loss evokes the unfolded protein response. We are still trying to figure out how this mechanistic function is reconcilable with its varied effects on cancer promotion. In this review, we focus on cancer-related effects of TUSC3 and envisage a possible role of TUSC3 beyond endoplasmic reticulum.
Collapse
Affiliation(s)
- Kateřina Vašíčková
- Department of Histology and Embryology, Faculty of Medicine, Masaryk University, Kamenice 126/3, 625 00, Brno, Czech Republic
- International Clinical Research Center, St. Anne's University Hospital Brno, Pekařská 53, 65691, Brno, Czech Republic
| | - Peter Horak
- Department of Translational Oncology, National Center for Tumor Diseases (NCT), Im Neuenheimer Feld 460, 69120, Heidelberg, Germany
- German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Petr Vaňhara
- Department of Histology and Embryology, Faculty of Medicine, Masaryk University, Kamenice 126/3, 625 00, Brno, Czech Republic.
- International Clinical Research Center, St. Anne's University Hospital Brno, Pekařská 53, 65691, Brno, Czech Republic.
| |
Collapse
|
30
|
Ferreira IG, Pucci M, Venturi G, Malagolini N, Chiricolo M, Dall'Olio F. Glycosylation as a Main Regulator of Growth and Death Factor Receptors Signaling. Int J Mol Sci 2018; 19:ijms19020580. [PMID: 29462882 PMCID: PMC5855802 DOI: 10.3390/ijms19020580] [Citation(s) in RCA: 72] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2017] [Revised: 02/12/2018] [Accepted: 02/14/2018] [Indexed: 12/22/2022] Open
Abstract
Glycosylation is a very frequent and functionally important post-translational protein modification that undergoes profound changes in cancer. Growth and death factor receptors and plasma membrane glycoproteins, which upon activation by extracellular ligands trigger a signal transduction cascade, are targets of several molecular anti-cancer drugs. In this review, we provide a thorough picture of the mechanisms bywhich glycosylation affects the activity of growth and death factor receptors in normal and pathological conditions. Glycosylation affects receptor activity through three non-mutually exclusive basic mechanisms: (1) by directly regulating intracellular transport, ligand binding, oligomerization and signaling of receptors; (2) through the binding of receptor carbohydrate structures to galectins, forming a lattice thatregulates receptor turnover on the plasma membrane; and (3) by receptor interaction with gangliosides inside membrane microdomains. Some carbohydrate chains, for example core fucose and β1,6-branching, exert a stimulatory effect on all receptors, while other structures exert opposite effects on different receptors or in different cellular contexts. In light of the crucial role played by glycosylation in the regulation of receptor activity, the development of next-generation drugs targeting glyco-epitopes of growth factor receptors should be considered a therapeutically interesting goal.
Collapse
Affiliation(s)
- Inês Gomes Ferreira
- Department of Experimental, Diagnostic and Specialty Medicine (DIMES), General Pathology Building, University of Bologna, 40126 Bologna, Italy.
| | - Michela Pucci
- Department of Experimental, Diagnostic and Specialty Medicine (DIMES), General Pathology Building, University of Bologna, 40126 Bologna, Italy.
| | - Giulia Venturi
- Department of Experimental, Diagnostic and Specialty Medicine (DIMES), General Pathology Building, University of Bologna, 40126 Bologna, Italy.
| | - Nadia Malagolini
- Department of Experimental, Diagnostic and Specialty Medicine (DIMES), General Pathology Building, University of Bologna, 40126 Bologna, Italy.
| | - Mariella Chiricolo
- Department of Experimental, Diagnostic and Specialty Medicine (DIMES), General Pathology Building, University of Bologna, 40126 Bologna, Italy.
| | - Fabio Dall'Olio
- Department of Experimental, Diagnostic and Specialty Medicine (DIMES), General Pathology Building, University of Bologna, 40126 Bologna, Italy.
| |
Collapse
|
31
|
Xing Y, Ge Y, Liu C, Zhang X, Jiang J, Wei Y. ER stress inducer tunicamycin suppresses the self-renewal of glioma-initiating cell partly through inhibiting Sox2 translation. Oncotarget 2017; 7:36395-36406. [PMID: 27119230 PMCID: PMC5095008 DOI: 10.18632/oncotarget.8954] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2015] [Accepted: 04/11/2016] [Indexed: 11/25/2022] Open
Abstract
Glioma-initiating cells possess tumor-initiating potential and are relatively resistant to conventional chemotherapy and irradiation. Therefore, their elimination is an essential factor for the development of efficient therapy. Here, we report that endoplasmic reticulum (ER) stress inducer tunicamycin inhibits glioma-initiating cell self-renewal as determined by neurosphere formation assay. Moreover, tunicamycin decreases the efficiency of glioma-initiating cell to initiate tumor formation. Although tunicamycin induces glioma-initiating cell apoptosis, apoptosis inhibitor z-VAD-fmk only partly abrogates the reduction in glioma-initiating cell self-renewal induced by tunicamycin. Indeed, tunicamycin reduces the expression of self-renewal regulator Sox2 at translation level. Overexpression of Sox2 obviously abrogates the reduction in glioma-initiating cell self-renewal induced by tunicamycin. Taken together, tunicamycin suppresses the self-renewal and tumorigenic potential of glioma-initiating cell partly through reducing Sox2 translation. This finding provides a cue to potential effective treatment of glioblastoma through controlling stem cells.
Collapse
Affiliation(s)
- Yang Xing
- Key Laboratory of Glycoconjuates Research, Ministry of Public Health, Department of Biochemistry and Molecular Biology, Shanghai Medical College of Fudan University, Shanghai, People's Republic of China
| | - Yuqing Ge
- Key Laboratory of Glycoconjuates Research, Ministry of Public Health, Department of Biochemistry and Molecular Biology, Shanghai Medical College of Fudan University, Shanghai, People's Republic of China
| | - Chanjuan Liu
- Key Laboratory of Glycoconjuates Research, Ministry of Public Health, Department of Biochemistry and Molecular Biology, Shanghai Medical College of Fudan University, Shanghai, People's Republic of China
| | - Xiaobiao Zhang
- Division of Neurosurgery, Zhongshan Hospital, Fudan University, Shanghai, People's Republic of China
| | - Jianhai Jiang
- Key Laboratory of Glycoconjuates Research, Ministry of Public Health, Department of Biochemistry and Molecular Biology, Shanghai Medical College of Fudan University, Shanghai, People's Republic of China
| | - Yuanyan Wei
- Key Laboratory of Glycoconjuates Research, Ministry of Public Health, Department of Biochemistry and Molecular Biology, Shanghai Medical College of Fudan University, Shanghai, People's Republic of China
| |
Collapse
|
32
|
de Freitas Junior JCM, Morgado-Díaz JA. The role of N-glycans in colorectal cancer progression: potential biomarkers and therapeutic applications. Oncotarget 2017; 7:19395-413. [PMID: 26539643 PMCID: PMC4991391 DOI: 10.18632/oncotarget.6283] [Citation(s) in RCA: 58] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2015] [Accepted: 10/22/2015] [Indexed: 12/12/2022] Open
Abstract
Changes in glycosylation, which is one of the most common protein post-translational modifications, are considered to be a hallmark of cancer. N-glycans can modulate cell migration, cell-cell adhesion, cell signaling, growth and metastasis. The colorectal cancer (CRC) is a leading cause of cancer-related mortality and the correlation between CRC progression and changes in the pattern of expression of N-glycans is being considered in the search for new biomarkers. Here, we review the role of N-glycans in CRC cell biology. The perspectives on emerging N-glycan-related anticancer therapies, along with new insights and challenges, are also discussed.
Collapse
Affiliation(s)
| | - José Andrés Morgado-Díaz
- Cellular Biology Program, Structural Biology Group, Brazilian National Cancer Institute (INCA), Rio de Janeiro, RJ, Brazil
| |
Collapse
|
33
|
Wang D, Fang M, Shen N, Li L, Wang W, Wang L, Lin H. Inhibiting post-translational core fucosylation protects against albumin-induced proximal tubular epithelial cell injury. Am J Transl Res 2017; 9:4415-4427. [PMID: 29118904 PMCID: PMC5666051] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2017] [Accepted: 09/10/2017] [Indexed: 06/07/2023]
Abstract
Albuminuria is an independent risk factor for renal interstitial fibrosis (RIF). Glomerular-filtered albumin in endocytic and non-endocytic pathways may injure proximal tubular epithelial cells (PTECs) via megalin and TGFβRII, respectively. Since megalin and TGFβRII are both modified by post-translational core fucosylation, which plays a critical role in RIF. Thus, we sought to identify whether core fucosylation is a potential target for reducing albumin-induced injury to PTECs. We constructed a human PTEC-derived cell line (HK-2 cells) and established an in vitro model of bovine serum albumin (BSA) injury. RNAi was used to inhibit the expression of megalin, TGFβRII, and Fut8. Western blotting, immunostaining, ELISA, lectin blotting, and fluorescence-activated cell sorting were used to identify BSA-induced endocytic and non-endocytic damage in HK-2 cells. Fut8 is a core fucosylation-related gene, which is significantly increased in HK-2 cells following an incubation with BSA. Fut8 siRNA significantly reduced the core fucosylation of megalin and TGFβRII and also inhibited the activation of the TGFβ/TGFβRII/Smad2/3 signaling pathway. Furthermore, Fut8 siRNA could reduce monocyte chemotactic protein-1, reactive oxygen species, and apoptosis, as well as significantly decrease the fibronectin and collagen I levels in BSA-overloaded HK-2 cells. Core fucosylation inhibition was more effective than inhibiting either megalin or TGFβRII for the prevention of albumin-induced injury to PTECs. Our findings indicate that post-translational core fucosylation is essential for the albumin-induced injury to PTECs. Thus, the inhibition of core fucosylation could effectively alleviate albumin-induced endocytic and non-endocytic injury to PTECs. Our study provides a potential therapeutic target for albuminuria-induced injury.
Collapse
Affiliation(s)
- Dapeng Wang
- Department of Nephrology, The First Affiliated Hospital, Institute for Nephrology Research of Dalian Medical University, Center for Kidney Diseases Translational Medicine of Liaoning ProvinceDalian, China
| | - Ming Fang
- Postgraduate School of Dalian Medical UniversityDalian, China
| | - Nan Shen
- Department of Nephrology, The First Affiliated Hospital, Institute for Nephrology Research of Dalian Medical University, Center for Kidney Diseases Translational Medicine of Liaoning ProvinceDalian, China
| | - Longkai Li
- Department of Nephrology, The First Affiliated Hospital, Institute for Nephrology Research of Dalian Medical University, Center for Kidney Diseases Translational Medicine of Liaoning ProvinceDalian, China
| | - Weidong Wang
- Department of Nephrology, The First Affiliated Hospital, Institute for Nephrology Research of Dalian Medical University, Center for Kidney Diseases Translational Medicine of Liaoning ProvinceDalian, China
| | - Lingyu Wang
- Department of Nephrology, The First Affiliated Hospital, Institute for Nephrology Research of Dalian Medical University, Center for Kidney Diseases Translational Medicine of Liaoning ProvinceDalian, China
| | - Hongli Lin
- Department of Nephrology, The First Affiliated Hospital, Institute for Nephrology Research of Dalian Medical University, Center for Kidney Diseases Translational Medicine of Liaoning ProvinceDalian, China
| |
Collapse
|
34
|
Glycans as Regulatory Elements of the Insulin/IGF System: Impact in Cancer Progression. Int J Mol Sci 2017; 18:ijms18091921. [PMID: 28880250 PMCID: PMC5618570 DOI: 10.3390/ijms18091921] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2017] [Revised: 08/30/2017] [Accepted: 09/02/2017] [Indexed: 12/12/2022] Open
Abstract
The insulin/insulin-like growth factor (IGF) system in mammals comprises a dynamic network of proteins that modulate several biological processes such as development, cell growth, metabolism, and aging. Dysregulation of the insulin/IGF system has major implications for several pathological conditions such as diabetes and cancer. Metabolic changes also culminate in aberrant glycosylation, which has been highlighted as a hallmark of cancer. Changes in glycosylation regulate every pathophysiological step of cancer progression including tumour cell-cell dissociation, cell migration, cell signaling and metastasis. This review discusses how the insulin/IGF system integrates with glycosylation alterations and impacts on cell behaviour, metabolism and drug resistance in cancer.
Collapse
|
35
|
The directed migration of gonadal distal tip cells in Caenorhabditis elegans requires NGAT-1, a ß1,4-N-acetylgalactosaminyltransferase enzyme. PLoS One 2017; 12:e0183049. [PMID: 28817611 PMCID: PMC5560668 DOI: 10.1371/journal.pone.0183049] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2017] [Accepted: 07/30/2017] [Indexed: 01/01/2023] Open
Abstract
Glycoproteins such as growth factor receptors and extracellular matrix have well-known functions in development and cancer progression, however, the glycans at sites of modification are often heterogeneous molecular populations which makes their functional characterization challenging. Here we provide evidence for a specific, discrete, well-defined glycan modification and regulation of a stage-specific cell migration in Caenorhabditis elegans. We show that a chain-terminating, putative null mutation in the gene encoding a predicted β1,4-N-acetylgalactosaminyltransferase, named ngat-1, causes a maternally rescued temperature sensitive (ts) defect in the second phase of the three phase migration pattern of the posterior, but not the anterior, hermaphrodite Distal Tip Cell (DTC). An amino-terminal partial deletion of ngat-1 causes a similar but lower penetrance ts phenotype. The existence of multiple ts alleles with distinctly different molecular DNA lesions, neither of which is likely to encode a ts protein, indicates that NGAT-1 normally prevents innate temperature sensitivity for phase 2 DTC pathfinding. Temperature shift analyses indicate that the ts period for the ngat-1 mutant defect ends by the beginning of post-embryonic development-nearly 3 full larval stages prior to the defective phase 2 migration affected by ngat-1 mutations. NGAT-1 homologs generate glycan-terminal GalNAc-β1-4GlcNAc, referred to as LacdiNAc modifications, on glycoproteins and glycolipids. We also found that the absence of the GnT1/Mgat1 activity [UDP-N-acetyl-D-glucosamine:α-3-D-mannoside β-1,2-N-acetylglucosaminyltransferase 1 (encoded by C. elegans gly-12, gly-13, and gly-14 and homologous to vertebrate GnT1/Mgat1)], causes a similar spectrum of DTC phenotypes as ngat-1 mutations-primarily affecting posterior DTC phase 2 migration and preventing manifestation of the same innate ts period as ngat-1. GnT1/Mgat1 is a medial Golgi enzyme known to modify mannose residues and initiate N-glycan branching, an essential step in the biosynthesis of hybrid, paucimannose and complex-type N-glycans. Quadruple mutant animals bearing putative null mutations in ngat-1 and the three GnT genes (gly-12, gly-13, gly-14) were not enhanced for DTC migration defects, suggesting NGAT-1 and GnT1 act in the same pathway. These findings suggest that GnTI generates an N-glycan substrate for NGAT-1 modification, which is required at restrictive temperature (25°C) to prevent, stabilize, reverse or compensate a perinatal thermo-labile process (or structure) causing late larval stage DTC phase 2 migration errors.
Collapse
|
36
|
Chen R, Lai LA, Sullivan Y, Wong M, Wang L, Riddell J, Jung L, Pillarisetty VG, Brentnall TA, Pan S. Disrupting glutamine metabolic pathways to sensitize gemcitabine-resistant pancreatic cancer. Sci Rep 2017; 7:7950. [PMID: 28801576 PMCID: PMC5554139 DOI: 10.1038/s41598-017-08436-6] [Citation(s) in RCA: 62] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2017] [Accepted: 07/11/2017] [Indexed: 12/12/2022] Open
Abstract
Pancreatic cancer is a lethal disease with poor prognosis. Gemcitabine has been the first line systemic treatment for pancreatic cancer. However, the rapid development of drug resistance has been a major hurdle in gemcitabine therapy leading to unsatisfactory patient outcomes. With the recent renewed understanding of glutamine metabolism involvement in drug resistance and immuno-response, we investigated the anti-tumor effect of a glutamine analog (6-diazo-5-oxo-L-norleucine) as an adjuvant treatment to sensitize chemoresistant pancreatic cancer cells. We demonstrate that disruption of glutamine metabolic pathways improves the efficacy of gemcitabine treatment. Such a disruption induces a cascade of events which impacts glycan biosynthesis through Hexosamine Biosynthesis Pathway (HBP), as well as cellular redox homeostasis, resulting in global changes in protein glycosylation, expression and functional effects. The proteome alterations induced in the resistant cancer cells and the secreted exosomes are intricately associated with the reduction in cell proliferation and the enhancement of cancer cell chemosensitivity. Proteins associated with EGFR signaling, including downstream AKT-mTOR pathways, MAPK pathway, as well as redox enzymes were downregulated in response to disruption of glutamine metabolic pathways.
Collapse
Affiliation(s)
- Ru Chen
- Department of Medicine, University of Washington, Seattle, WA, 98195, USA.
| | - Lisa A Lai
- Department of Medicine, University of Washington, Seattle, WA, 98195, USA
| | - Yumi Sullivan
- Department of Medicine, University of Washington, Seattle, WA, 98195, USA
| | - Melissa Wong
- Department of Medicine, University of Washington, Seattle, WA, 98195, USA
| | - Lei Wang
- Department of Medicine, University of Washington, Seattle, WA, 98195, USA
| | - Jonah Riddell
- Cell Signaling Technology, Inc, Danvers, MA, 01923, USA
| | - Linda Jung
- Cell Signaling Technology, Inc, Danvers, MA, 01923, USA
| | | | - Teresa A Brentnall
- Department of Medicine, University of Washington, Seattle, WA, 98195, USA
| | - Sheng Pan
- Department of Medicine, University of Washington, Seattle, WA, 98195, USA.
| |
Collapse
|
37
|
Huang S, Wang D, Zhang S, Huang X, Wang D, Ijaz M, Shi Y. Tunicamycin potentiates paclitaxel-induced apoptosis through inhibition of PI3K/AKT and MAPK pathways in breast cancer. Cancer Chemother Pharmacol 2017; 80:685-696. [DOI: 10.1007/s00280-017-3393-7] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2017] [Accepted: 07/13/2017] [Indexed: 10/19/2022]
|
38
|
Xie YZ, Ma WL, Meng JM, Ren XQ. Knockdown of ZFPL1 results in increased autophagy and autophagy‑related cell death in NCI‑N87 and BGC‑823 human gastric carcinoma cell lines. Mol Med Rep 2017; 15:2633-2642. [PMID: 28447717 DOI: 10.3892/mmr.2017.6300] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2015] [Accepted: 09/28/2016] [Indexed: 11/06/2022] Open
Abstract
Macroautophagy, which will hereafter be referred to as autophagy, is an evolutionarily conserved process, during which cells recycle and remove damaged organelles and proteins in response to cellular stress. However, the mechanisms underlying the regulation of autophagy remain to be fully elucidated. The present study demonstrated that knockdown of zinc finger protein like 1 (ZFPL1) induces autophagy and increases autophagic cell death in NCI‑N87 and BGC‑823 human gastric carcinoma cell lines. To examine the role of ZFPL1 in gastric carcinoma cells, ZFPL1 expression was downregulated by lentiviral infection. Zinc finger domain‑FLAG was used to compete with ZFPL1 for golgin A2/GM130 binding. Autophagy was analyzed by red fluorescent protein‑microtubule‑associated protein 1A/1B‑light chain 3 (LC3) puncta, LC3I to LC3II conversion, and p62 expression. The results demonstrated that knockdown of ZFPL1 was able to significantly increase cell death rate. However, ZFPL1 knockdown exerted almost no effect on the expression of apoptosis‑associated markers, including B cell lymphoma 2 (Bcl‑2), Bcl‑x, Bcl‑2‑associated X protein, BH3 interacting domain death agonist, p53, and the classical caspase family members, caspase‑3, caspase‑8 and caspase‑9. An endogenous ZFPL1‑GM130 association was identified in NCI‑N87 cells and BGC‑823 cells by co‑immunoprecipitation. Furthermore, cell death was restricted following treatment of ZFPL1 knockdown cells with an autophagy inhibitor. Therefore, knockdown of ZFPL1 expression may induce cell death via autophagy, rather than apoptosis. These results suggest that ZFPL1 may serve an important role in regulating autophagy in NCI‑N87 and BGC‑823 cells.
Collapse
Affiliation(s)
- Yong-Zheng Xie
- Department of General Surgery, Huaihe Hospital of Henan University, Kaifeng, Henan 475000, P.R. China
| | - Wan-Li Ma
- Department of General Surgery, Huaihe Hospital of Henan University, Kaifeng, Henan 475000, P.R. China
| | - Ji-Ming Meng
- Department of General Surgery, Huaihe Hospital of Henan University, Kaifeng, Henan 475000, P.R. China
| | - Xue-Qun Ren
- Department of General Surgery, Huaihe Hospital of Henan University, Kaifeng, Henan 475000, P.R. China
| |
Collapse
|
39
|
Han X, Zhang X, Li H, Huang S, Zhang S, Wang F, Shi Y. Tunicamycin enhances the antitumor activity of trastuzumab on breast cancer in vitro and in vivo. Oncotarget 2016; 6:38912-25. [PMID: 26498681 PMCID: PMC4770746 DOI: 10.18632/oncotarget.5334] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2015] [Accepted: 08/26/2015] [Indexed: 12/21/2022] Open
Abstract
Trastuzumab, a humanized monoclonal antibody targeting HER2, has demonstrated clinical benefits for women with HER2-positive breast cancer; however, trastuzumab resistance remains the biggest clinical challenge. In this study, results showed that tunicamycin, an inhibitor of N-glycosylation, synergistically enhanced the antitumor activity of trastuzumab against HER2-overexpressing breast cancer cells through induction of cell cycle arrest and apoptosis. Combined treatment of tunicamycin with trastuzumab dramatically decreased the expression of EGFR family and its down signaling pathway in SKBR3 and MCF-7/HER2 cells. Tunicamycin dose-dependently inhibited tumor growth in both of SKBR3 xenografts and MCF-7/HER2 xenografts. Optimal tunicamycin without inducing ER stress in liver tissue significantly increased the antitumor effect of trastuzumab in MCF-7/HER2 xenografts. Combinations of trastuzumab with N-glycosylation inhibitors tunicamycin may be a promising approach for improving clinical efficacy of trastuzumab.
Collapse
Affiliation(s)
- Xiqian Han
- National Glycoengineering Research Center, School of Pharmaceutical Science, Shandong University, Jinan, China
| | - Xiaobing Zhang
- National Glycoengineering Research Center, School of Pharmaceutical Science, Shandong University, Jinan, China
| | - Hui Li
- National Glycoengineering Research Center, School of Pharmaceutical Science, Shandong University, Jinan, China
| | - Shengshi Huang
- National Glycoengineering Research Center, School of Pharmaceutical Science, Shandong University, Jinan, China
| | - Shu Zhang
- National Glycoengineering Research Center, School of Pharmaceutical Science, Shandong University, Jinan, China
| | - Fengshan Wang
- National Glycoengineering Research Center, School of Pharmaceutical Science, Shandong University, Jinan, China
| | - Yikang Shi
- National Glycoengineering Research Center, School of Pharmaceutical Science, Shandong University, Jinan, China
| |
Collapse
|
40
|
Oligosaccharyltransferase inhibition induces senescence in RTK-driven tumor cells. Nat Chem Biol 2016; 12:1023-1030. [PMID: 27694802 DOI: 10.1038/nchembio.2194] [Citation(s) in RCA: 75] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2016] [Accepted: 07/29/2016] [Indexed: 12/21/2022]
Abstract
Asparagine (N)-linked glycosylation is a protein modification critical for glycoprotein folding, stability, and cellular localization. To identify small molecules that inhibit new targets in this biosynthetic pathway, we initiated a cell-based high-throughput screen and lead-compound-optimization campaign that delivered a cell-permeable inhibitor, NGI-1. NGI-1 targets oligosaccharyltransferase (OST), a hetero-oligomeric enzyme that exists in multiple isoforms and transfers oligosaccharides to recipient proteins. In non-small-cell lung cancer cells, NGI-1 blocks cell-surface localization and signaling of the epidermal growth factor receptor (EGFR) glycoprotein, but selectively arrests proliferation in only those cell lines that are dependent on EGFR (or fibroblast growth factor, FGFR) for survival. In these cell lines, OST inhibition causes cell-cycle arrest accompanied by induction of p21, autofluorescence, and cell morphology changes, all hallmarks of senescence. These results identify OST inhibition as a potential therapeutic approach for treating receptor-tyrosine-kinase-dependent tumors and provides a chemical probe for reversibly regulating N-linked glycosylation in mammalian cells.
Collapse
|
41
|
Hennigan K, Conroy PJ, Walsh MT, Amin M, O'Kennedy R, Ramasamy P, Gleich GJ, Siddiqui Z, Glynn S, McCabe O, Mooney C, Harvey BJ, Costello RW, McBryan J. Eosinophil peroxidase activates cells by HER2 receptor engagement and β1-integrin clustering with downstream MAPK cell signaling. Clin Immunol 2016; 171:1-11. [PMID: 27519953 PMCID: PMC5070911 DOI: 10.1016/j.clim.2016.08.009] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2016] [Revised: 08/02/2016] [Accepted: 08/06/2016] [Indexed: 01/21/2023]
Abstract
Eosinophils account for 1–3% of peripheral blood leukocytes and accumulate at sites of allergic inflammation, where they play a pathogenic role. Studies have shown that treatment with mepolizumab (an anti-IL-5 monoclonal antibody) is beneficial to patients with severe eosinophilic asthma, however, the mechanism of precisely how eosinophils mediate these pathogenic effects is uncertain. Eosinophils contain several cationic granule proteins, including Eosinophil Peroxidase (EPO). The main significance of this work is the discovery of EPO as a novel ligand for the HER2 receptor. Following HER2 activation, EPO induces activation of FAK and subsequent activation of β1-integrin, via inside-out signaling. This complex results in downstream activation of ERK1/2 and a sustained up regulation of both MUC4 and the HER2 receptor. These data identify a receptor for one of the eosinophil granule proteins and demonstrate a potential explanation of the proliferative effects of eosinophils. Eosinophil peroxidase (EPO) is confirmed as a ligand for the HER2 receptor. EPO activation of HER2 leads to activation of FAK, ERK and β1 integrin. EPO induces a sustained upregulation of MUC4 and HER2. Possible mechanism for the proliferative effects of eosinophils is uncovered.
Collapse
Affiliation(s)
- Kerrie Hennigan
- Department of Medicine Respiratory Research Division, Royal College of Surgeons in Ireland, Beaumont Hospital, Dublin 9, Ireland
| | - Paul J Conroy
- Biomedical Diagnostics Institute, Dublin City University, Dublin 9, Ireland
| | - Marie-Therese Walsh
- Department of Medicine Respiratory Research Division, Royal College of Surgeons in Ireland, Beaumont Hospital, Dublin 9, Ireland
| | - Mohamed Amin
- Department of Medicine Respiratory Research Division, Royal College of Surgeons in Ireland, Beaumont Hospital, Dublin 9, Ireland
| | - Richard O'Kennedy
- Biomedical Diagnostics Institute, Dublin City University, Dublin 9, Ireland
| | - Patmapriya Ramasamy
- Department of Medicine Respiratory Research Division, Royal College of Surgeons in Ireland, Beaumont Hospital, Dublin 9, Ireland
| | - Gerald J Gleich
- Department of Dermatology, University of Utah, Salt Lake City, USA
| | - Zeshan Siddiqui
- Graduate Entry Medical School, University of Limerick, Ireland
| | - Senan Glynn
- Department of Medicine Respiratory Research Division, Royal College of Surgeons in Ireland, Beaumont Hospital, Dublin 9, Ireland
| | - Olive McCabe
- Department of Molecular Medicine, Royal College of Surgeons in Ireland, Beaumont Hospital, Dublin 9, Ireland
| | - Catherine Mooney
- Department of Physiology and Medical Physics, Royal College of Surgeons in Ireland, Dublin 2, Ireland
| | - Brian J Harvey
- Department of Molecular Medicine, Royal College of Surgeons in Ireland, Beaumont Hospital, Dublin 9, Ireland
| | - Richard W Costello
- Department of Medicine Respiratory Research Division, Royal College of Surgeons in Ireland, Beaumont Hospital, Dublin 9, Ireland.
| | - Jean McBryan
- Department of Molecular Medicine, Royal College of Surgeons in Ireland, Beaumont Hospital, Dublin 9, Ireland
| |
Collapse
|
42
|
Peñaranda Fajardo NM, Meijer C, Kruyt FAE. The endoplasmic reticulum stress/unfolded protein response in gliomagenesis, tumor progression and as a therapeutic target in glioblastoma. Biochem Pharmacol 2016; 118:1-8. [PMID: 27106078 DOI: 10.1016/j.bcp.2016.04.008] [Citation(s) in RCA: 80] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2016] [Accepted: 04/12/2016] [Indexed: 11/16/2022]
Abstract
Endoplasmic reticulum (ER) stress disrupts among others protein homeostasis in cells leading to the activation of the unfolded protein response (UPR) that is crucial for restoring this balance and cell survival. Hypoxia, reactive oxygen species and nutrient deprivation, conditions commonly present in the tumor microenvironment, are well-known triggers of the UPR. Apart from being an adaptive response, recently the UPR has been implicated in oncogenesis. Here we review the current understanding of the UPR in the most life threatening brain tumor in adults, glioblastoma multiforme (GBM). The UPR is controlled by BiP/GRP78 and three different sensors, PERK, IRE1 and ATF6. In orthotopic GBM mouse models IRE1 was reported to control angiogenesis, invasion and mesenchymal differentiation. Furthermore, PERK also was found to stimulate GBM growth. However, a direct role of the UPR in gliomagenesis remains to be demonstrated. Patient samples display chronic activation of the UPR and in vitro standard chemo- and radiotherapy partially act by aggravating ER stress leading to cell death. The UPR has been linked to enhanced sensitivity for apoptosis-inducing agents such as TRAIL and MDA-7. A number of agents such as proteasome inhibitors and several natural products were reported to exert cytotoxicity by enhancing ER stress in GBM cells, and some demonstrated activity in clinical studies. Finally, ER stress was suggested to be implicated in the maintenance of homeostasis in GBM stem cells. Taken together, the UPR appears to play an important role in GBM tumor progression and is a promising target for developing novel therapeutic interventions.
Collapse
Affiliation(s)
- Natalia M Peñaranda Fajardo
- Department of Medical Oncology, University of Groningen, University Medical Center Groningen, The Netherlands
| | - Coby Meijer
- Department of Medical Oncology, University of Groningen, University Medical Center Groningen, The Netherlands
| | - Frank A E Kruyt
- Department of Medical Oncology, University of Groningen, University Medical Center Groningen, The Netherlands.
| |
Collapse
|
43
|
Zito Marino F, Rocco G, Morabito A, Mignogna C, Intartaglia M, Liguori G, Botti G, Franco R. A new look at the ALK gene in cancer: copy number gain and amplification. Expert Rev Anticancer Ther 2016; 16:493-502. [PMID: 26943457 DOI: 10.1586/14737140.2016.1162098] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
To date, ALK-rearrangement is a molecular target in several cancers, i.e. NSCLC. The dramatic benefits of crizotinib have prompted research into identifying other possible patients carrying ALK gene alterations with possible clinical significance. The ALK gene is involved not only in several rearrangements but also in other alterations such as amplification. ALK-amplification (ALK-A) is a common genetic event in several cancers, generally associated with poor outcome and more aggressive behaviour. Here we review the role of ALK-A in cancer as a prognostic and predictive biomarker. Furthermore, several critical issues regarding ALK-A in relation to; methods of detection, acquired resistance and ALK second generation inhibitors are analyzed. We conclude that ALK-A could be an intriguing alteration in the context of targeted therapy.
Collapse
Affiliation(s)
- Federica Zito Marino
- a Pathology Unit , Istituto Nazionale Tumori 'Fondazione G. Pascale'-IRCCS , Naples , Italy
| | - Gaetano Rocco
- b Division of Thoracic Surgery, Department of Thoracic Surgical and Medical Oncology , Istituto Nazionale Tumori 'Fondazione G. Pascale'-IRCCS , Naples , Italy
| | - Alessandro Morabito
- c Medical Oncology Unit, Department of Thoracic Surgical and Medical Oncology , Istituto Nazionale Tumori 'Fondazione G. Pascale'-IRCCS , Naples , Italy
| | - Chiara Mignogna
- d Department of Heath Science, Pathology Unit , University 'Magna Graecia' of Catanzaro , Catanzaro , Italy
| | - Martina Intartaglia
- a Pathology Unit , Istituto Nazionale Tumori 'Fondazione G. Pascale'-IRCCS , Naples , Italy
| | - Giuseppina Liguori
- a Pathology Unit , Istituto Nazionale Tumori 'Fondazione G. Pascale'-IRCCS , Naples , Italy
| | - Gerardo Botti
- a Pathology Unit , Istituto Nazionale Tumori 'Fondazione G. Pascale'-IRCCS , Naples , Italy
| | - Renato Franco
- a Pathology Unit , Istituto Nazionale Tumori 'Fondazione G. Pascale'-IRCCS , Naples , Italy.,e Pathology Unit , Second University of Naples - SUN , Naples , Italy
| |
Collapse
|
44
|
Gim J, Cho YB, Hong HK, Kim HC, Yun SH, Wu HG, Jeong SY, Joung JG, Park T, Park WY, Lee WY. Predicting multi-class responses to preoperative chemoradiotherapy in rectal cancer patients. Radiat Oncol 2016; 11:50. [PMID: 27005571 PMCID: PMC4804643 DOI: 10.1186/s13014-016-0623-9] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2015] [Accepted: 03/15/2016] [Indexed: 02/06/2023] Open
Abstract
Background Preoperative chemoradiotherapy (CRT) has become a widely used treatment for improving local control of disease and increasing survival rates of rectal cancer patients. We aimed to identify a set of genes that can be used to predict responses to CRT in patients with rectal cancer. Methods Gene expression profiles of pre-therapeutic biopsy specimens obtained from 77 rectal cancer patients were analyzed using DNA microarrays. The response to CRT was determined using the Dworak tumor regression grade: grade 1 (minimal, MI), grade 2 (moderate, MO), grade 3 (near total, NT), or grade 4 (total, TO). Results Top ranked genes for three different feature scores such as a p-value (pval), a rank product (rank), and a normalized product (norm) were selected to distinguish pre-defined groups such as complete responders (TO) from the MI, MO, and NT groups. Among five different classification algorithms, supporting vector machine (SVM) with the top 65 norm features performed at the highest accuracy for predicting MI using a 5-fold cross validation strategy. On the other hand, 98 pval features were selected for predicting TO by elastic net (EN). Finally we combined TO- and MI-finder models to build a three-class classification model and validated it using an independent dataset of rectal cancer mRNA expression. Conclusions We identified MI- and TO-finders for predicting preoperative CRT responses, and validated these data using an independent public dataset. This stepwise prediction model requires further evaluation in clinical studies in order to develop personalized preoperative CRT in patients with rectal cancer. Electronic supplementary material The online version of this article (doi:10.1186/s13014-016-0623-9) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Jungsoo Gim
- Institute of Health and Environment, Seoul National University, Seoul, 151-742, Korea
| | - Yong Beom Cho
- Department of Surgery, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, 135-710, Korea
| | - Hye Kyung Hong
- Department of Surgery, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, 135-710, Korea
| | - Hee Cheol Kim
- Department of Surgery, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, 135-710, Korea
| | - Seong Hyeon Yun
- Department of Surgery, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, 135-710, Korea
| | - Hong-Gyun Wu
- Department of Radiation Oncology, College of Medicine, Seoul National University, Seoul, 110-799, Korea
| | - Seung-Yong Jeong
- Department of Surgery, College of Medicine, Seoul National University, Seoul, 110-799, Korea
| | - Je-Gun Joung
- Samsung Medical Center, Samsung Genome Institute, Seoul, 135-710, Korea
| | - Taesung Park
- Institute of Health and Environment, Seoul National University, Seoul, 151-742, Korea. .,Department of Statistics, College of Natural Science, Seoul National University, Seoul, 151-742, Korea.
| | - Woong-Yang Park
- Department of Molecular Cell Biology, Sungkyunkwan University School of Medicine, Seoul, 135-710, Korea. .,Samsung Medical Center, Samsung Genome Institute, Seoul, 135-710, Korea.
| | - Woo Yong Lee
- Department of Surgery, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, 135-710, Korea.
| |
Collapse
|
45
|
Kanev MO, Bakar E. Kanserde Glikokonjugatlar. KOCAELI ÜNIVERSITESI SAĞLIK BILIMLERI DERGISI 2016. [DOI: 10.30934/kusbed.358473] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
|
46
|
Olivares S, Henkel AS. Hepatic Xbp1 Gene Deletion Promotes Endoplasmic Reticulum Stress-induced Liver Injury and Apoptosis. J Biol Chem 2015; 290:30142-51. [PMID: 26504083 DOI: 10.1074/jbc.m115.676239] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2015] [Indexed: 12/12/2022] Open
Abstract
Endoplasmic reticulum (ER) stress activates the unfolded protein response (UPR), a highly conserved signaling cascade that functions to alleviate stress and promote cell survival. If, however, the cell is unable to adapt and restore homeostasis, then the UPR activates pathways that promote apoptotic cell death. The molecular mechanisms governing the critical transition from adaptation and survival to initiation of apoptosis remain poorly understood. We aim to determine the role of hepatic Xbp1, a key mediator of the UPR, in controlling the adaptive response to ER stress in the liver. Liver-specific Xbp1 knockout mice (Xbp1(LKO)) and Xbp1(fl/fl) control mice were subjected to varying levels and durations of pharmacologic ER stress. Xbp1(LKO) and Xbp1(fl/fl) mice showed robust and equal activation of the UPR acutely after induction of ER stress. By 24 h, Xbp1(fl/fl) controls showed complete resolution of UPR activation and no liver injury, indicating successful adaptation to the stress. Conversely, Xbp1(LKO) mice showed ongoing UPR activation associated with progressive liver injury, apoptosis, and, ultimately, fibrosis by day 7 after induction of ER stress. These data indicate that hepatic XBP1 controls the adaptive response of the UPR and is critical to restoring homeostasis in the liver in response to ER stress.
Collapse
Affiliation(s)
- Shantel Olivares
- From the Division of Gastroenterology and Hepatology, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois 60611
| | - Anne S Henkel
- From the Division of Gastroenterology and Hepatology, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois 60611
| |
Collapse
|
47
|
Abstract
This review presents principles of glycosylation, describes the relevant glycosylation pathways and their related disorders, and highlights some of the neurological aspects and issues that continue to challenge researchers. More than 100 rare human genetic disorders that result from deficiencies in the different glycosylation pathways are known today. Most of these disorders impact the central and/or peripheral nervous systems. Patients typically have developmental delays/intellectual disabilities, hypotonia, seizures, neuropathy, and metabolic abnormalities in multiple organ systems. Among these disorders there is great clinical diversity because all cell types differentially glycosylate proteins and lipids. The patients have hundreds of misglycosylated products, which afflict a myriad of processes, including cell signaling, cell-cell interaction, and cell migration. This vast complexity in glycan composition and function, along with the limited availability of analytic tools, has impeded the identification of key glycosylated molecules that cause pathologies. To date, few critical target proteins have been pinpointed.
Collapse
|
48
|
Vasconcelos-Dos-Santos A, Oliveira IA, Lucena MC, Mantuano NR, Whelan SA, Dias WB, Todeschini AR. Biosynthetic Machinery Involved in Aberrant Glycosylation: Promising Targets for Developing of Drugs Against Cancer. Front Oncol 2015; 5:138. [PMID: 26161361 PMCID: PMC4479729 DOI: 10.3389/fonc.2015.00138] [Citation(s) in RCA: 100] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2015] [Accepted: 06/02/2015] [Indexed: 12/22/2022] Open
Abstract
Cancer cells depend on altered metabolism and nutrient uptake to generate and keep the malignant phenotype. The hexosamine biosynthetic pathway is a branch of glucose metabolism that produces UDP-GlcNAc and its derivatives, UDP-GalNAc and CMP-Neu5Ac and donor substrates used in the production of glycoproteins and glycolipids. Growing evidence demonstrates that alteration of the pool of activated substrates might lead to different glycosylation and cell signaling. It is already well established that aberrant glycosylation can modulate tumor growth and malignant transformation in different cancer types. Therefore, biosynthetic machinery involved in the assembly of aberrant glycans are becoming prominent targets for anti-tumor drugs. This review describes three classes of glycosylation, O-GlcNAcylation, N-linked, and mucin type O-linked glycosylation, involved in tumor progression, their biosynthesis and highlights the available inhibitors as potential anti-tumor drugs.
Collapse
Affiliation(s)
| | - Isadora A Oliveira
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro , Rio de Janeiro , Brasil
| | - Miguel Clodomiro Lucena
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro , Rio de Janeiro , Brasil
| | - Natalia Rodrigues Mantuano
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro , Rio de Janeiro , Brasil
| | - Stephen A Whelan
- Department of Biochemistry, Cardiovascular Proteomics Center, Boston University School of Medicine , Boston, MA , USA
| | - Wagner Barbosa Dias
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro , Rio de Janeiro , Brasil
| | - Adriane Regina Todeschini
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro , Rio de Janeiro , Brasil
| |
Collapse
|
49
|
Wang Y, Fukuda T, Isaji T, Lu J, Im S, Hang Q, Gu W, Hou S, Ohtsubo K, Gu J. Loss of α1,6-fucosyltransferase inhibits chemical-induced hepatocellular carcinoma and tumorigenesis by down-regulating several cell signaling pathways. FASEB J 2015; 29:3217-27. [PMID: 25873065 DOI: 10.1096/fj.15-270710] [Citation(s) in RCA: 65] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2015] [Accepted: 03/31/2015] [Indexed: 12/28/2022]
Abstract
Up-regulation of core fucosylation catalyzed by α1,6-fucosyltransferase (Fut8) has been observed in hepatocellular carcinoma (HCC). Here, to explore the role of Fut8 expression in hepatocarcinogensis, we established the chemical-induced HCC models in the male wild-type (WT; Fut8(+/+)), hetero (Fut8(+/-)), and knockout (KO; Fut8(-/-)) mice by use of diethylnitrosamine (DEN) and pentobarbital (PB). In the Fut8(+/+) and Fut8(+/-) mice, multiple large and vascularized nodules were induced with an increased expression of Fut8 after DEN and PB treatment. However, the formation of HCC in Fut8(-/-) mice was suppressed almost completely. This potent inhibitory effect of Fut8 deficiency on tumorigenesis was also confirmed by the abolished tumor formation of Fut8 KO human hepatoma cell line cells by use of a xenograft tumor model. Furthermore, loss of the Fut8 gene resulted in attenuated responses to epidermal growth factor (EGF) and hepatocyte growth factor (HGF) in the HepG2 cell line, which provides the possible mechanisms for the contribution of Fut8 to hepatocarcinogensis. Taken together, our study clearly demonstrated that core fucosylation acts as a critical functional modulator in the liver and implicated Fut8 as a prognostic marker, as well as a novel, therapeutic target for HCC.
Collapse
Affiliation(s)
- Yuqin Wang
- *Division of Regulatory Glycobiology, Tohoku Pharmaceutical University, Sendai, Miyagi, Japan; and Department of Analytical Biochemistry, Faculty of Life Sciences, Kumamoto University, Japan
| | - Tomohiko Fukuda
- *Division of Regulatory Glycobiology, Tohoku Pharmaceutical University, Sendai, Miyagi, Japan; and Department of Analytical Biochemistry, Faculty of Life Sciences, Kumamoto University, Japan
| | - Tomoya Isaji
- *Division of Regulatory Glycobiology, Tohoku Pharmaceutical University, Sendai, Miyagi, Japan; and Department of Analytical Biochemistry, Faculty of Life Sciences, Kumamoto University, Japan
| | - Jishun Lu
- *Division of Regulatory Glycobiology, Tohoku Pharmaceutical University, Sendai, Miyagi, Japan; and Department of Analytical Biochemistry, Faculty of Life Sciences, Kumamoto University, Japan
| | - Sanghun Im
- *Division of Regulatory Glycobiology, Tohoku Pharmaceutical University, Sendai, Miyagi, Japan; and Department of Analytical Biochemistry, Faculty of Life Sciences, Kumamoto University, Japan
| | - Qinglei Hang
- *Division of Regulatory Glycobiology, Tohoku Pharmaceutical University, Sendai, Miyagi, Japan; and Department of Analytical Biochemistry, Faculty of Life Sciences, Kumamoto University, Japan
| | - Wei Gu
- *Division of Regulatory Glycobiology, Tohoku Pharmaceutical University, Sendai, Miyagi, Japan; and Department of Analytical Biochemistry, Faculty of Life Sciences, Kumamoto University, Japan
| | - Sicong Hou
- *Division of Regulatory Glycobiology, Tohoku Pharmaceutical University, Sendai, Miyagi, Japan; and Department of Analytical Biochemistry, Faculty of Life Sciences, Kumamoto University, Japan
| | - Kazuaki Ohtsubo
- *Division of Regulatory Glycobiology, Tohoku Pharmaceutical University, Sendai, Miyagi, Japan; and Department of Analytical Biochemistry, Faculty of Life Sciences, Kumamoto University, Japan
| | - Jianguo Gu
- *Division of Regulatory Glycobiology, Tohoku Pharmaceutical University, Sendai, Miyagi, Japan; and Department of Analytical Biochemistry, Faculty of Life Sciences, Kumamoto University, Japan
| |
Collapse
|
50
|
Cazet A, Charest J, Bennett DC, Sambrooks CL, Contessa JN. Mannose phosphate isomerase regulates fibroblast growth factor receptor family signaling and glioma radiosensitivity. PLoS One 2014; 9:e110345. [PMID: 25314669 PMCID: PMC4196966 DOI: 10.1371/journal.pone.0110345] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2014] [Accepted: 09/18/2014] [Indexed: 12/11/2022] Open
Abstract
Asparagine-linked glycosylation is an endoplasmic reticulum co- and post-translational modification that enables the transit and function of receptor tyrosine kinase (RTK) glycoproteins. To gain insight into the regulatory role of glycosylation enzymes on RTK function, we investigated shRNA and siRNA knockdown of mannose phosphate isomerase (MPI), an enzyme required for mature glycan precursor biosynthesis. Loss of MPI activity reduced phosphorylation of FGFR family receptors in U-251 and SKMG-3 malignant glioma cell lines and also resulted in significant decreases in FRS2, Akt, and MAPK signaling. However, MPI knockdown did not affect ligand-induced activation or signaling of EGFR or MET RTKs, suggesting that FGFRs are more susceptible to MPI inhibition. The reductions in FGFR signaling were not caused by loss of FGF ligands or receptors, but instead were caused by interference with receptor dimerization. Investigations into the cellular consequences of MPI knockdown showed that cellular programs driven by FGFR signaling, and integral to the clinical progression of malignant glioma, were impaired. In addition to a blockade of cellular migration, MPI knockdown also significantly reduced glioma cell clonogenic survival following ionizing radiation. Therefore our results suggest that targeted inhibition of enzymes required for cell surface receptor glycosylation can be manipulated to produce discrete and limited consequences for critical client glycoproteins expressed by tumor cells. Furthermore, this work identifies MPI as a potential enzymatic target for disrupting cell surface receptor-dependent survival signaling and as a novel approach for therapeutic radiosensitization.
Collapse
MESH Headings
- Cell Line, Tumor
- Cell Membrane/metabolism
- Cell Movement/genetics
- Cell Proliferation
- Gene Knockdown Techniques
- Glioma/genetics
- Glioma/metabolism
- Glioma/radiotherapy
- Humans
- Mannose-6-Phosphate Isomerase/genetics
- Mannose-6-Phosphate Isomerase/metabolism
- Protein Multimerization
- Protein Transport
- RNA, Small Interfering
- Radiation Tolerance/genetics
- Receptor Protein-Tyrosine Kinases/metabolism
- Receptor, Fibroblast Growth Factor, Type 2/chemistry
- Receptor, Fibroblast Growth Factor, Type 2/genetics
- Receptor, Fibroblast Growth Factor, Type 2/metabolism
- Receptors, Fibroblast Growth Factor/chemistry
- Receptors, Fibroblast Growth Factor/genetics
- Receptors, Fibroblast Growth Factor/metabolism
- Signal Transduction
Collapse
Affiliation(s)
- Aurélie Cazet
- Department of Therapeutic Radiology, Yale School of Medicine, New Haven, Connecticut, United States of America
| | - Jonathan Charest
- Department of Therapeutic Radiology, Yale School of Medicine, New Haven, Connecticut, United States of America
| | - Daniel C. Bennett
- Department of Therapeutic Radiology, Yale School of Medicine, New Haven, Connecticut, United States of America
| | - Cecilia Lopez Sambrooks
- Department of Therapeutic Radiology, Yale School of Medicine, New Haven, Connecticut, United States of America
| | - Joseph N. Contessa
- Department of Therapeutic Radiology, Yale School of Medicine, New Haven, Connecticut, United States of America
| |
Collapse
|