1
|
López-Cortés R, Muinelo-Romay L, Fernández-Briera A, Gil Martín E. High-Throughput Mass Spectrometry Analysis of N-Glycans and Protein Markers after FUT8 Knockdown in the Syngeneic SW480/SW620 Colorectal Cancer Cell Model. J Proteome Res 2024; 23:1379-1398. [PMID: 38507902 PMCID: PMC11002942 DOI: 10.1021/acs.jproteome.3c00833] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2023] [Revised: 02/22/2024] [Accepted: 03/01/2024] [Indexed: 03/22/2024]
Abstract
Disruption of the glycosylation machinery is a common feature in many types of cancer, and colorectal cancer (CRC) is no exception. Core fucosylation is mediated by the enzyme fucosyltransferase 8 (FucT-8), which catalyzes the addition of α1,6-l-fucose to the innermost GlcNAc residue of N-glycans. We and others have documented the involvement of FucT-8 and core-fucosylated proteins in CRC progression, in which we addressed core fucosylation in the syngeneic CRC model formed by SW480 and SW620 tumor cell lines from the perspective of alterations in their N-glycosylation profile and protein expression as an effect of the knockdown of the FUT8 gene that encodes FucT-8. Using label-free, semiquantitative mass spectrometry (MS) analysis, we found noticeable differences in N-glycosylation patterns in FUT8-knockdown cells, affecting core fucosylation and sialylation, the Hex/HexNAc ratio, and antennarity. Furthermore, stable isotopic labeling of amino acids in cell culture (SILAC)-based proteomic screening detected the alteration of species involved in protein folding, endoplasmic reticulum (ER) and Golgi post-translational stabilization, epithelial polarity, and cellular response to damage and therapy. This data is available via ProteomeXchange with identifier PXD050012. Overall, the results obtained merit further investigation to validate their feasibility as biomarkers of progression and malignization in CRC, as well as their potential usefulness in clinical practice.
Collapse
Affiliation(s)
- Rubén López-Cortés
- Doctoral
Program in Methods and Applications in Life Sciences, Faculty of Biology, Universidade de Vigo, Campus Lagoas-Marcosende, 36310 Vigo, Pontevedra (Galicia), Spain
| | - Laura Muinelo-Romay
- Liquid
Biopsy Analysis Unit, Translational Medical Oncology (Oncomet), Health Research Institute of Santiago de Compostela
(IDIS), CIBERONC, Travesía da Choupana, 15706 Santiago de Compostela, A Coruña
(Galicia), Spain
| | - Almudena Fernández-Briera
- Molecular
Biomarkers, Biomedical Research Centre (CINBIO), Universidade de Vigo, Campus Lagoas-Marcosende, 36310 Vigo, Pontevedra (Galicia), Spain
| | - Emilio Gil Martín
- Nutrition
and Food Science Group, Department of Biochemistry, Genetics and Immunology,
Faculty of Biology, Universidade de Vigo, Campus Lagoas-Marcosende, 36310 Vigo, Pontevedra (Galicia), Spain
| |
Collapse
|
2
|
López-Cortés R, Correa Pardo I, Muinelo-Romay L, Fernández-Briera A, Gil-Martín E. Core Fucosylation Mediated by the FucT-8 Enzyme Affects TRAIL-Induced Apoptosis and Sensitivity to Chemotherapy in Human SW480 and SW620 Colorectal Cancer Cells. Int J Mol Sci 2023; 24:11879. [PMID: 37569254 PMCID: PMC10418920 DOI: 10.3390/ijms241511879] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2023] [Revised: 07/15/2023] [Accepted: 07/21/2023] [Indexed: 08/13/2023] Open
Abstract
Epithelial cells can undergo apoptosis by manipulating the balance between pro-survival and apoptotic signals. In this work, we show that TRAIL-induced apoptosis can be differentially regulated by the expression of α(1,6)fucosyltransferase (FucT-8), the only enzyme in mammals that transfers the α(1,6)fucose residue to the pentasaccharide core of complex N-glycans. Specifically, in the cellular model of colorectal cancer (CRC) progression formed using the human syngeneic lines SW480 and SW620, knockdown of the FucT-8-encoding FUT8 gene significantly enhanced TRAIL-induced apoptosis in SW480 cells. However, FUT8 repression did not affect SW620 cells, which suggests that core fucosylation differentiates TRAIL-sensitive premetastatic SW480 cells from TRAIL-resistant metastatic SW620 cells. In this regard, we provide evidence that phosphorylation of ERK1/2 kinases can dynamically regulate TRAIL-dependent apoptosis and that core fucosylation can control the ERK/MAPK pro-survival pathway in which SW480 and SW620 cells participate. Moreover, the depletion of core fucosylation sensitises primary tumour SW480 cells to the combination of TRAIL and low doses of 5-FU, oxaliplatin, irinotecan, or mitomycin C. In contrast, a combination of TRAIL and oxaliplatin, irinotecan, or bevacizumab reinforces resistance of FUT8-knockdown metastatic SW620 cells to apoptosis. Consequently, FucT-8 could be a plausible target for increasing apoptosis and drug response in early CRC.
Collapse
Affiliation(s)
- Rubén López-Cortés
- Doctoral Program in Methods and Applications in Life Sciences, Faculty of Biology, Universidade de Vigo, Campus Lagoas-Marcosende, ES36310 Vigo, Spain;
| | - Isabel Correa Pardo
- Master Program in Advanced Biotechnology, Faculty of Biology, Universidade de Vigo, Campus Lagoas-Marcosende, ES36310 Vigo, Spain;
| | - Laura Muinelo-Romay
- Liquid Biopsy Analysis Unit, Translational Medical Oncology (Oncomet), Health Research Institute of Santiago de Compostela (IDIS), CIBERONC, Travesía da Choupana, ES15706 Santiago de Compostela, Spain;
| | - Almudena Fernández-Briera
- Department of Biochemistry, Genetics and Immunology, Faculty of Biology, Universidade de Vigo, Campus Lagoas-Marcosende, ES36310 Vigo, Spain;
| | - Emilio Gil-Martín
- Department of Biochemistry, Genetics and Immunology, Faculty of Biology, Universidade de Vigo, Campus Lagoas-Marcosende, ES36310 Vigo, Spain;
| |
Collapse
|
3
|
Zhang NZ, Zhao LF, Zhang Q, Fang H, Song WL, Li WZ, Ge YS, Gao P. Core fucosylation and its roles in gastrointestinal glycoimmunology. World J Gastrointest Oncol 2023; 15:1119-1134. [PMID: 37546555 PMCID: PMC10401475 DOI: 10.4251/wjgo.v15.i7.1119] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/18/2023] [Revised: 02/28/2023] [Accepted: 05/08/2023] [Indexed: 07/12/2023] Open
Abstract
Glycosylation is a common post-translational modification in eukaryotic cells. It is involved in the production of many biologically active glycoproteins and the regulation of protein structure and function. Core fucosylation plays a vital role in the immune response. Most immune system molecules are core fucosylated glycoproteins such as complements, cluster differentiation antigens, immunoglobulins, cytokines, major histocompatibility complex molecules, adhesion molecules, and immune molecule synthesis-related transcription factors. These core fucosylated glycoproteins play important roles in antigen recognition and clearance, cell adhesion, lymphocyte activation, apoptosis, signal transduction, and endocytosis. Core fucosylation is dominated by fucosyltransferase 8 (Fut8), which catalyzes the addition of α-1,6-fucose to the innermost GlcNAc residue of N-glycans. Fut8 is involved in humoral, cellular, and mucosal immunity. Tumor immunology is associated with aberrant core fucosylation. Here, we summarize the roles and potential modulatory mechanisms of Fut8 in various immune processes of the gastrointestinal system.
Collapse
Affiliation(s)
- Nian-Zhu Zhang
- Clinical Laboratory, The Second Hospital of Dalian Medical University, Dalian 116023, Liaoning Province, China
| | - Li-Fen Zhao
- Clinical Laboratory, The Second Hospital of Dalian Medical University, Dalian 116023, Liaoning Province, China
| | - Qian Zhang
- Department of Cell Therapy, Shanghai Tianze Yuntai Biomedical Co., Ltd., Shanghai 200100, China
| | - Hui Fang
- Graduate School of Comprehensive Human Sciences, University of Tsukuba, Tsukuba 305-0005, Ibaraki, Japan
| | - Wan-Li Song
- Guangdong Provincial Key Laboratory of Infectious Diseases and Molecular Immunopathology, Shantou University Medical College, Shantou 515041, Guangdong Province, China
| | - Wen-Zhe Li
- Guangdong Provincial Key Laboratory of Infectious Diseases and Molecular Immunopathology, Shantou University Medical College, Shantou 515041, Guangdong Province, China
| | - Yu-Song Ge
- Department of Neurology, The Second Hospital of Dalian Medical University, Dalian 116023, Liaoning Province, China
| | - Peng Gao
- Clinical Laboratory, The Second Hospital of Dalian Medical University, Dalian 116023, Liaoning Province, China
| |
Collapse
|
4
|
López-Cortés R, Muinelo-Romay L, Fernández-Briera A, Gil-Martín E. Inhibition of α(1,6)fucosyltransferase: Effects on Cell Proliferation, Migration, and Adhesion in an SW480/SW620 Syngeneic Colorectal Cancer Model. Int J Mol Sci 2022; 23:ijms23158463. [PMID: 35955598 PMCID: PMC9369121 DOI: 10.3390/ijms23158463] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Revised: 07/27/2022] [Accepted: 07/28/2022] [Indexed: 02/01/2023] Open
Abstract
The present study explored the impact of inhibiting α(1,6)fucosylation (core fucosylation) on the functional phenotype of a cellular model of colorectal cancer (CRC) malignization formed by the syngeneic SW480 and SW620 CRC lines. Expression of the FUT8 gene encoding α(1,6)fucosyltransferase was inhibited in tumor line SW480 by a combination of shRNA-based antisense knockdown and Lens culinaris agglutinin (LCA) selection. LCA-resistant clones were subsequently assayed in vitro for proliferation, migration, and adhesion. The α(1,6)FT-inhibited SW480 cells showed enhanced proliferation in adherent conditions, unlike their α(1,6)FT-depleted SW620 counterparts, which displayed reduced proliferation. Under non-adherent conditions, α(1,6)FT-inhibited SW480 cells also showed greater growth capacity than their respective non-targeted control (NTC) cells. However, cell migration decreased in SW480 after FUT8 knockdown, while adhesion to EA.hy926 cells was significantly enhanced. The reported results indicate that the FUT8 knockdown strategy with subsequent selection for LCA-resistant clones was effective in greatly reducing α(1,6)FT expression in SW480 and SW620 CRC lines. In addition, α(1,6)FT impairment affected the proliferation, migration, and adhesion of α(1,6)FT-deficient clones SW480 and SW620 in a tumor stage-dependent manner, suggesting that core fucosylation has a dynamic role in the evolution of CRC.
Collapse
Affiliation(s)
- Rubén López-Cortés
- Doctoral Program in Methods and Applications in Life Sciences, Faculty of Biology, Campus Lagoas-Marcosende, Universidade de Vigo, 36310 Vigo, Spain;
| | - Laura Muinelo-Romay
- Liquid Biopsy Analysis Unit, Translational Medical Oncology (Oncomet), Health Research Institute of Santiago de Compostela (IDIS), CIBERONC, Travesía da Choupana, 15706 Santiago de Compostela, Spain;
| | - Almudena Fernández-Briera
- Department of Biochemistry, Genetics and Immunology, Faculty of Biology, Campus Lagoas-Marcosende, Universidade de Vigo, 36310 Vigo, Spain;
| | - Emilio Gil-Martín
- Department of Biochemistry, Genetics and Immunology, Faculty of Biology, Campus Lagoas-Marcosende, Universidade de Vigo, 36310 Vigo, Spain;
- Correspondence: ; Tel.: +34-(986)-812-570
| |
Collapse
|
5
|
Gao Y, Zhang G, Liu J, Li H. Tissue-specific transplantation antigen P35B functions as an oncogene and is regulated by microRNA-125a-5p in lung cancer. Oncol Rep 2021; 45:72. [PMID: 33760213 PMCID: PMC8020207 DOI: 10.3892/or.2021.8023] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2020] [Accepted: 09/16/2020] [Indexed: 12/21/2022] Open
Abstract
Tissue‑specific transplantation antigen P35B (TSTA3) expression is upregulated in esophageal squamous cell carcinoma and breast cancer, and functions as an oncogene in breast cancer. However, the roles and underlying mechanisms of TSTA3 in lung cancer have not been fully elucidated. The current study aimed to reveal the role of TSTA3 in lung cancer and explore whether TSTA3 may be modulated by microRNA (miR)‑125a‑5p to activate β‑catenin signaling. Immunohistochemical staining and western blotting were used to analyze TSTA3 expression in lung cancer tissues and cells. Cell functions were assessed via Cell Counting Kit‑8, flow cytometry, wound‑healing, Transwell and in vivo tumor formation assays. The effect of TSTA3 on the activation of β‑catenin signaling was determined using western blot and immunofluorescence analyses. The association between miR‑125a‑5p and TSTA3 was determined by western blotting and luciferase gene reporter assay. The present study revealed that, compared with normal tissues and cells, TSTA3 expression was significantly increased in lung cancer tissues and cell lines, and high TSTA3 expression predicted a poor prognosis and more malignant clinical features in patients with lung cancer. TSTA3 upregulation significantly enhanced β‑catenin expression and promoted its nuclear accumulation. In addition, TSTA3 expression was negatively regulated by miR‑125a‑5p, which was downregulated in lung cancer. Furthermore, TSTA3 overexpression markedly promoted cell proliferation, migration, invasion and tumorigenesis, and suppressed cell apoptosis. TSTA3 downregulation abolished the effects of miR‑125a‑5p downregulation on promoting lung cancer cell malignant transformation. Overall, the current study demonstrates that TSTA3 is regulated by miR‑125a‑5p and functions as an oncogene in lung cancer via promoting the activation of β‑catenin signaling.
Collapse
Affiliation(s)
- Yingjie Gao
- Department of Oncology, Liaocheng People's Hospital, Liaocheng, Shandong 252000, P.R. China
| | - Guangliang Zhang
- Department of Oncology, Liaocheng People's Hospital, Liaocheng, Shandong 252000, P.R. China
| | - Jinlong Liu
- Department of Orthopedics, Liaocheng People's Hospital, Liaocheng, Shandong 252000, P.R. China
| | - Huimin Li
- Department of Oncology, Liaocheng People's Hospital, Liaocheng, Shandong 252000, P.R. China
| |
Collapse
|
6
|
Bastian K, Scott E, Elliott DJ, Munkley J. FUT8 Alpha-(1,6)-Fucosyltransferase in Cancer. Int J Mol Sci 2021; 22:E455. [PMID: 33466384 PMCID: PMC7795606 DOI: 10.3390/ijms22010455] [Citation(s) in RCA: 75] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2020] [Revised: 12/21/2020] [Accepted: 12/24/2020] [Indexed: 12/15/2022] Open
Abstract
Aberrant glycosylation is a universal feature of cancer cells that can impact all steps in tumour progression from malignant transformation to metastasis and immune evasion. One key change in tumour glycosylation is altered core fucosylation. Core fucosylation is driven by fucosyltransferase 8 (FUT8), which catalyses the addition of α1,6-fucose to the innermost GlcNAc residue of N-glycans. FUT8 is frequently upregulated in cancer, and plays a critical role in immune evasion, antibody-dependent cellular cytotoxicity (ADCC), and the regulation of TGF-β, EGF, α3β1 integrin and E-Cadherin. Here, we summarise the role of FUT8 in various cancers (including lung, liver, colorectal, ovarian, prostate, breast, melanoma, thyroid, and pancreatic), discuss the potential mechanisms involved, and outline opportunities to exploit FUT8 as a critical factor in cancer therapeutics in the future.
Collapse
Affiliation(s)
- Kayla Bastian
- Institute of Biosciences, Newcastle University, Newcastle Upon Tyne NE1 3BZ, UK; (E.S.); (D.J.E.); (J.M.)
| | | | | | | |
Collapse
|
7
|
Kitaguchi D, Oda T, Enomoto T, Ohara Y, Owada Y, Akashi Y, Furuta T, Yu Y, Kimura S, Kuroda Y, Kurimori K, Miyazaki Y, Furuya K, Shimomura O, Tateno H. Lectin drug conjugate therapy for colorectal cancer. Cancer Sci 2020; 111:4548-4557. [PMID: 33058342 PMCID: PMC7734164 DOI: 10.1111/cas.14687] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2020] [Revised: 10/06/2020] [Accepted: 10/08/2020] [Indexed: 01/05/2023] Open
Abstract
Drug resistance represents an obstacle in colorectal cancer (CRC) treatment because of its association with poor prognosis. rBC2LCN is a lectin isolated from Burkholderia that binds cell surface glycans that have fucose moieties. Because fucosylation is enhanced in many types of cancers, this lectin could be an efficient drug carrier if CRC cells specifically present such glycans. Therefore, we examined the therapeutic efficacy and toxicity of lectin drug conjugate therapy in CRC mouse xenograft models. The affinity of rBC2LCN for human CRC cell lines HT-29, LoVo, LS174T, and DLD-1 was assessed in vitro. The cytocidal efficacy of a lectin drug conjugate, rBC2LCN-38 kDa domain of pseudomonas exotoxin A (PE38) was evaluated by MTT assay. The therapeutic effects and toxicity for each CRC cell line-derived mouse xenograft model were compared between the intervention and control groups. LS174T and DLD-1 cell lines showed a strong affinity for rBC2LCN. In the xenograft model, the tumor volume in the rBC2LCN-PE38 group was significantly reduced compared with that using control treatment alone. However, the HT-29 cell line showed weak affinity and poor therapeutic efficacy. No significant toxicities or adverse responses were observed. In conclusion, we demonstrated that rBC2LCN lectin binds CRC cells and that rBC2LCN-PE38 significantly suppresses tumor growth in vivo. In addition, the efficacy of the drug conjugate correlated with its binding affinity for each CRC cell line. These results suggest that lectin drug conjugate therapy has potential as a novel targeted therapy for CRC cell surface glycans.
Collapse
Affiliation(s)
- Daichi Kitaguchi
- Department of Gastrointestinal and Hepato‐Biliary‐Pancreatic SurgeryFaculty of MedicineUniversity of TsukubaTsukubaJapan
| | - Tatsuya Oda
- Department of Gastrointestinal and Hepato‐Biliary‐Pancreatic SurgeryFaculty of MedicineUniversity of TsukubaTsukubaJapan
| | - Tsuyoshi Enomoto
- Department of Gastrointestinal and Hepato‐Biliary‐Pancreatic SurgeryFaculty of MedicineUniversity of TsukubaTsukubaJapan
| | - Yusuke Ohara
- Department of Gastrointestinal and Hepato‐Biliary‐Pancreatic SurgeryFaculty of MedicineUniversity of TsukubaTsukubaJapan
| | - Yohei Owada
- Department of Gastrointestinal and Hepato‐Biliary‐Pancreatic SurgeryFaculty of MedicineUniversity of TsukubaTsukubaJapan
| | - Yoshimasa Akashi
- Department of Gastrointestinal and Hepato‐Biliary‐Pancreatic SurgeryFaculty of MedicineUniversity of TsukubaTsukubaJapan
| | - Tomoaki Furuta
- Department of Gastrointestinal and Hepato‐Biliary‐Pancreatic SurgeryFaculty of MedicineUniversity of TsukubaTsukubaJapan
| | - Yang Yu
- Department of Gastrointestinal and Hepato‐Biliary‐Pancreatic SurgeryFaculty of MedicineUniversity of TsukubaTsukubaJapan
| | - Sota Kimura
- Department of Gastrointestinal and Hepato‐Biliary‐Pancreatic SurgeryFaculty of MedicineUniversity of TsukubaTsukubaJapan
| | - Yukihito Kuroda
- Department of Gastrointestinal and Hepato‐Biliary‐Pancreatic SurgeryFaculty of MedicineUniversity of TsukubaTsukubaJapan
| | - Ko Kurimori
- Department of Gastrointestinal and Hepato‐Biliary‐Pancreatic SurgeryFaculty of MedicineUniversity of TsukubaTsukubaJapan
| | - Yoshihiro Miyazaki
- Department of Gastrointestinal and Hepato‐Biliary‐Pancreatic SurgeryFaculty of MedicineUniversity of TsukubaTsukubaJapan
| | - Kinji Furuya
- Department of Gastrointestinal and Hepato‐Biliary‐Pancreatic SurgeryFaculty of MedicineUniversity of TsukubaTsukubaJapan
| | - Osamu Shimomura
- Department of Gastrointestinal and Hepato‐Biliary‐Pancreatic SurgeryFaculty of MedicineUniversity of TsukubaTsukubaJapan
| | - Hiroaki Tateno
- Biotechnology Research Institute for Drug DiscoveryNational Institute of Advanced Industrial Science and TechnologyTsukubaJapan
| |
Collapse
|
8
|
Rubén LC, Laura MR, Almudena FB, Emilio GM. Glycan array analysis of Pholiota squarrosa lectin and other fucose-oriented lectins. Glycobiology 2020; 31:459-476. [PMID: 33021632 DOI: 10.1093/glycob/cwaa093] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2020] [Revised: 09/23/2020] [Accepted: 09/23/2020] [Indexed: 12/14/2022] Open
Abstract
The α(1,6)fucose residue attached to the N-glycoprotein core is suspected to play an essential role in the progression of several types of cancer. Lectins remain the first choice for probing glycan modifications, although they may lack specificity. Thus, efforts have been made to identify new lectins with a narrower core fucose (CF) detection profile. Here, we present a comparison of the classical Aleuria aurantia lectin (AAL), Lens culinaris agglutinin (LCA) and Aspergillus oryzae lectin (AOL) with the newer Pholiota squarrosa lectin (PhoSL), which has been described as being specific for core fucosylated N-glycans. To this end, we studied the binding profiles of the four lectins using mammalian glycan arrays from the Consortium of Functional Glycomics. To validate their glycan specificity, we probed AOL, LCA and PhoSL in western-blot assays using protein extracts from eight common colorectal cancer (CRC) lines and colorectal biopsies from a small cohort of patients with CRC. The results showed that (i) LCA and PhoSL were the most specific lectins for detecting the presence of CF in a concentration-dependent manner; (ii) PhoSL exhibited the highest N-glycan sequence restriction, with preferential binding to core fucosylated paucimannosidic-type N-glycans, (iii) the recognition ability of PhoSL was highly influenced by the presence of terminal N-acetyl-lactosamine; (iv) LCA bound to paucimannosidic, bi-antennary and tri-antennary core fucosylated N-glycans and (v) AOL and AAL exhibited broader specificity towards fucosylation. Together, our results support the choice of LCA as the most appropriate lectin for CF detection, as validated in protein extracts from CRC cell lines and tissue specimens from patients with CRC.
Collapse
Affiliation(s)
- López-Cortés Rubén
- Doctoral Program in Methods and Applications in Life Sciences, Faculty of Biology, Universidade de Vigo, Campus Lagoas-Marcosende, Vigo, Pontevedra, Galicia ES36310, Spain
| | - Muinelo-Romay Laura
- Liquid Biopsy Analysis Unit, Translational Medical Oncology (Oncomet), Health Research Institute of Santiago de Compostela (IDIS), CIBERONC, Travesía da Choupana, Santiago de Compostela, A Coruña, Galicia ES15706, Spain
| | - Fernández-Briera Almudena
- Molecular Biomarkers, Biomedical Research Centre (CINBIO), Universidade de Vigo, Campus Lagoas-Marcosende, Vigo, Pontevedra, Galicia ES36310, Spain
| | - Gil Martín Emilio
- Nutrition and Food Science Group, Department of Biochemistry, Genetics and Immunology, Faculty of Biology, Universidade de Vigo. Campus Lagoas-Marcosende, Vigo, Pontevedra, Galicia ES36310, Spain
| |
Collapse
|
9
|
Yang J, Kong P, Yang J, Jia Z, Hu X, Wang Z, Cui H, Bi Y, Qian Y, Li H, Wang F, Yang B, Yan T, Ma Y, Zhang L, Cheng C, Song B, Li Y, Xu E, Liu H, Gao W, Wang J, Liu Y, Zhai Y, Chang L, Wang Y, Zhang Y, Shi R, Liu J, Wang Q, Cheng X, Cui Y. High TSTA3 Expression as a Candidate Biomarker for Poor Prognosis of Patients With ESCC. Technol Cancer Res Treat 2018; 17:1533033818781405. [PMID: 29950151 PMCID: PMC6048620 DOI: 10.1177/1533033818781405] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
Esophageal squamous cell carcinoma is the sixth most lethal cancer worldwide and the
fourth most lethal cancer in China. Tissue-specific transplantation antigen P35B codifies
the enzyme GDP-d-mannose-4,6-dehydratase, which participates in the biosynthesis
of GDP-l-fucose. GDP-l-fucose is an important substrate involved in the
biosynthesis of many glycoproteins. Cancer cells are often accompanied by the changes in
glycoprotein structure, which affects the adhesion, invasion, and metastasis of cells. It
is not clear whether tissue-specific transplantation antigen P35B has any effect on the
development of esophageal squamous cell carcinoma. We used an immunohistochemical method
to assess the expression of tissue-specific transplantation antigen P35B in 104 esophageal
squamous cell carcinoma samples. The results showed tissue-specific transplantation
antigen P35B expression was associated with some clinical features in patients, such as
age (P = .017), clinical stage (P = .010), and lymph
node metastasis (P = .043). Kaplan-Meier analysis and log-rank test
showed that patients with esophageal squamous cell carcinoma having high tissue-specific
transplantation antigen P35B expression had a worse prognosis compared to the patients
with low expression (P = .048). Multivariate Cox proportional hazards
regression model showed that high expression of tissue-specific transplantation antigen
P35B could predict poor prognosis for patients with esophageal squamous cell carcinoma
independently. In conclusion, abnormal fucosylation might participate in the progress of
esophageal squamous cell carcinoma and tissue-specific transplantation antigen P35B may
serve as a novel biomarker for prognosis of patients with esophageal squamous cell
carcinoma.
Collapse
Affiliation(s)
- Jie Yang
- 1 Translational Medicine Research Center, Shanxi Medical University, Taiyuan, Shanxi, PR China.,2 Key Laboratory of Cellular Physiology, Ministry of Education, Shanxi Medical University, Taiyuan, Shanxi, PR China.,3 Department of Gastroenterology, The Second Hospital, Shanxi Medical University, Taiyuan, Shanxi, PR China
| | - Pengzhou Kong
- 1 Translational Medicine Research Center, Shanxi Medical University, Taiyuan, Shanxi, PR China.,2 Key Laboratory of Cellular Physiology, Ministry of Education, Shanxi Medical University, Taiyuan, Shanxi, PR China
| | - Jian Yang
- 1 Translational Medicine Research Center, Shanxi Medical University, Taiyuan, Shanxi, PR China.,2 Key Laboratory of Cellular Physiology, Ministry of Education, Shanxi Medical University, Taiyuan, Shanxi, PR China
| | - Zhiwu Jia
- 1 Translational Medicine Research Center, Shanxi Medical University, Taiyuan, Shanxi, PR China.,2 Key Laboratory of Cellular Physiology, Ministry of Education, Shanxi Medical University, Taiyuan, Shanxi, PR China
| | - Xiaoling Hu
- 1 Translational Medicine Research Center, Shanxi Medical University, Taiyuan, Shanxi, PR China.,2 Key Laboratory of Cellular Physiology, Ministry of Education, Shanxi Medical University, Taiyuan, Shanxi, PR China.,4 Department of Pharmacology, Shanxi Medical University, Taiyuan, Shanxi, PR China
| | - Zianyi Wang
- 1 Translational Medicine Research Center, Shanxi Medical University, Taiyuan, Shanxi, PR China.,5 Taiyuan Lingde Secondary School, Taiyuan, Shanxi, PR China
| | - Heyang Cui
- 1 Translational Medicine Research Center, Shanxi Medical University, Taiyuan, Shanxi, PR China.,2 Key Laboratory of Cellular Physiology, Ministry of Education, Shanxi Medical University, Taiyuan, Shanxi, PR China
| | - Yanghui Bi
- 1 Translational Medicine Research Center, Shanxi Medical University, Taiyuan, Shanxi, PR China.,2 Key Laboratory of Cellular Physiology, Ministry of Education, Shanxi Medical University, Taiyuan, Shanxi, PR China
| | - Yu Qian
- 1 Translational Medicine Research Center, Shanxi Medical University, Taiyuan, Shanxi, PR China.,2 Key Laboratory of Cellular Physiology, Ministry of Education, Shanxi Medical University, Taiyuan, Shanxi, PR China
| | - Hongyi Li
- 1 Translational Medicine Research Center, Shanxi Medical University, Taiyuan, Shanxi, PR China.,2 Key Laboratory of Cellular Physiology, Ministry of Education, Shanxi Medical University, Taiyuan, Shanxi, PR China
| | - Fang Wang
- 1 Translational Medicine Research Center, Shanxi Medical University, Taiyuan, Shanxi, PR China.,2 Key Laboratory of Cellular Physiology, Ministry of Education, Shanxi Medical University, Taiyuan, Shanxi, PR China
| | - Bin Yang
- 1 Translational Medicine Research Center, Shanxi Medical University, Taiyuan, Shanxi, PR China.,2 Key Laboratory of Cellular Physiology, Ministry of Education, Shanxi Medical University, Taiyuan, Shanxi, PR China.,6 Department of General Surgery, Shanxi Cancer Hospital, Taiyuan, Shanxi, PR China
| | - Ting Yan
- 1 Translational Medicine Research Center, Shanxi Medical University, Taiyuan, Shanxi, PR China.,2 Key Laboratory of Cellular Physiology, Ministry of Education, Shanxi Medical University, Taiyuan, Shanxi, PR China
| | - Yanchun Ma
- 1 Translational Medicine Research Center, Shanxi Medical University, Taiyuan, Shanxi, PR China.,2 Key Laboratory of Cellular Physiology, Ministry of Education, Shanxi Medical University, Taiyuan, Shanxi, PR China
| | - Ling Zhang
- 1 Translational Medicine Research Center, Shanxi Medical University, Taiyuan, Shanxi, PR China.,2 Key Laboratory of Cellular Physiology, Ministry of Education, Shanxi Medical University, Taiyuan, Shanxi, PR China
| | - Caixia Cheng
- 1 Translational Medicine Research Center, Shanxi Medical University, Taiyuan, Shanxi, PR China.,2 Key Laboratory of Cellular Physiology, Ministry of Education, Shanxi Medical University, Taiyuan, Shanxi, PR China.,7 Department of Pathology, The First Hospital, Shanxi Medical University, Taiyuan, Shanxi, PR China
| | - Bin Song
- 1 Translational Medicine Research Center, Shanxi Medical University, Taiyuan, Shanxi, PR China.,2 Key Laboratory of Cellular Physiology, Ministry of Education, Shanxi Medical University, Taiyuan, Shanxi, PR China.,8 Department of Oncology, The First Hospital, Shanxi Medical University, Taiyuan, Shanxi, PR China
| | - Yaoping Li
- 1 Translational Medicine Research Center, Shanxi Medical University, Taiyuan, Shanxi, PR China.,2 Key Laboratory of Cellular Physiology, Ministry of Education, Shanxi Medical University, Taiyuan, Shanxi, PR China.,9 Department of Anorectum, Shanxi Provincial People's Hospital, Taiyuan, Shanxi, China
| | - Enwei Xu
- 1 Translational Medicine Research Center, Shanxi Medical University, Taiyuan, Shanxi, PR China.,2 Key Laboratory of Cellular Physiology, Ministry of Education, Shanxi Medical University, Taiyuan, Shanxi, PR China.,10 Department of Pathology, Shanxi Cancer Hospital, Taiyuan, Shanxi, PR China
| | - Haiyan Liu
- 1 Translational Medicine Research Center, Shanxi Medical University, Taiyuan, Shanxi, PR China.,2 Key Laboratory of Cellular Physiology, Ministry of Education, Shanxi Medical University, Taiyuan, Shanxi, PR China
| | - Wei Gao
- 1 Translational Medicine Research Center, Shanxi Medical University, Taiyuan, Shanxi, PR China.,2 Key Laboratory of Cellular Physiology, Ministry of Education, Shanxi Medical University, Taiyuan, Shanxi, PR China.,11 Shanxi Key Laboratory of Otorhinolaryngology Head and Neck Cancer, Taiyuan, Shanxi, PR China.,12 Department of Otolaryngology Head & Neck Surgery, The First Hospital, Shanxi Medical University, Taiyuan, Shanxi, PR China
| | - Juan Wang
- 1 Translational Medicine Research Center, Shanxi Medical University, Taiyuan, Shanxi, PR China.,2 Key Laboratory of Cellular Physiology, Ministry of Education, Shanxi Medical University, Taiyuan, Shanxi, PR China
| | - Yiqian Liu
- 1 Translational Medicine Research Center, Shanxi Medical University, Taiyuan, Shanxi, PR China.,2 Key Laboratory of Cellular Physiology, Ministry of Education, Shanxi Medical University, Taiyuan, Shanxi, PR China
| | - Yuanfang Zhai
- 1 Translational Medicine Research Center, Shanxi Medical University, Taiyuan, Shanxi, PR China.,2 Key Laboratory of Cellular Physiology, Ministry of Education, Shanxi Medical University, Taiyuan, Shanxi, PR China.,13 Department of Anatomy, Shanxi Medical University, Taiyuan, Shanxi, PR China
| | - Lu Chang
- 1 Translational Medicine Research Center, Shanxi Medical University, Taiyuan, Shanxi, PR China.,2 Key Laboratory of Cellular Physiology, Ministry of Education, Shanxi Medical University, Taiyuan, Shanxi, PR China
| | - Yi Wang
- 1 Translational Medicine Research Center, Shanxi Medical University, Taiyuan, Shanxi, PR China.,2 Key Laboratory of Cellular Physiology, Ministry of Education, Shanxi Medical University, Taiyuan, Shanxi, PR China
| | - Yingchun Zhang
- 1 Translational Medicine Research Center, Shanxi Medical University, Taiyuan, Shanxi, PR China.,2 Key Laboratory of Cellular Physiology, Ministry of Education, Shanxi Medical University, Taiyuan, Shanxi, PR China.,13 Department of Anatomy, Shanxi Medical University, Taiyuan, Shanxi, PR China
| | - Ruyi Shi
- 1 Translational Medicine Research Center, Shanxi Medical University, Taiyuan, Shanxi, PR China.,2 Key Laboratory of Cellular Physiology, Ministry of Education, Shanxi Medical University, Taiyuan, Shanxi, PR China
| | - Jing Liu
- 1 Translational Medicine Research Center, Shanxi Medical University, Taiyuan, Shanxi, PR China.,2 Key Laboratory of Cellular Physiology, Ministry of Education, Shanxi Medical University, Taiyuan, Shanxi, PR China.,14 Department of General Surgery, The First Hospital, Shanxi Medical University, Taiyuan, Shanxi, PR China
| | - Qi Wang
- 3 Department of Gastroenterology, The Second Hospital, Shanxi Medical University, Taiyuan, Shanxi, PR China
| | - Xiaolong Cheng
- 1 Translational Medicine Research Center, Shanxi Medical University, Taiyuan, Shanxi, PR China.,2 Key Laboratory of Cellular Physiology, Ministry of Education, Shanxi Medical University, Taiyuan, Shanxi, PR China.,13 Department of Anatomy, Shanxi Medical University, Taiyuan, Shanxi, PR China
| | - Yongping Cui
- 1 Translational Medicine Research Center, Shanxi Medical University, Taiyuan, Shanxi, PR China.,2 Key Laboratory of Cellular Physiology, Ministry of Education, Shanxi Medical University, Taiyuan, Shanxi, PR China
| |
Collapse
|
10
|
Noda M, Okayama H, Tachibana K, Sakamoto W, Saito K, Thar Min AK, Ashizawa M, Nakajima T, Aoto K, Momma T, Katakura K, Ohki S, Kono K. Glycosyltransferase Gene Expression Identifies a Poor Prognostic Colorectal Cancer Subtype Associated with Mismatch Repair Deficiency and Incomplete Glycan Synthesis. Clin Cancer Res 2018; 24:4468-4481. [PMID: 29844132 DOI: 10.1158/1078-0432.ccr-17-3533] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2017] [Revised: 03/18/2018] [Accepted: 05/22/2018] [Indexed: 11/16/2022]
Abstract
Purpose: We aimed to discover glycosyltransferase gene (glycogene)-derived molecular subtypes of colorectal cancer associated with patient outcomes.Experimental Design: Transcriptomic and epigenomic datasets of nontumor, precancerous, cancerous tissues, and cell lines with somatic mutations, mismatch repair status, clinicopathologic and survival information were assembled (n = 4,223) and glycogene profiles were analyzed. IHC for a glycogene, GALNT6, was conducted in adenoma and carcinoma specimens (n = 403). The functional role and cell surface glycan profiles were further investigated by in vitro loss-of-function assays and lectin microarray analysis.Results: We initially developed and validated a 15-glycogene signature that can identify a poor-prognostic subtype, which closely related to deficient mismatch repair (dMMR) and GALNT6 downregulation. The association of decreased GALNT6 with dMMR was confirmed in multiple datasets of tumors and cell lines, and was further recapitulated by IHC, where approximately 15% tumors exhibited loss of GALNT6 protein. GALNT6 mRNA and protein was expressed in premalignant/preinvasive lesions but was subsequently downregulated in a subset of carcinomas, possibly through epigenetic silencing. Decreased GALNT6 was independently associated with poor prognosis in the IHC cohort and an additional microarray meta-cohort, by multivariate analyses, and its discriminative power of survival was particularly remarkable in stage III patients. GALNT6 silencing in SW480 cells promoted invasion, migration, chemoresistance, and increased cell surface expression of a cancer-associated truncated O-glycan, Tn-antigen.Conclusions: The 15-glycogene signature and the expression levels of GALNT6 mRNA and protein each serve as a novel prognostic biomarker, highlighting the role of dysregulated glycogenes in cancer-associated glycan synthesis and poor prognosis. Clin Cancer Res; 24(18); 4468-81. ©2018 AACR.
Collapse
Affiliation(s)
- Masaru Noda
- Department of Gastrointestinal Tract Surgery, Fukushima Medical University School of Medicine, Fukushima City, Japan
- Department of Breast Surgery, Fukushima Medical University School of Medicine, Fukushima City, Japan
| | - Hirokazu Okayama
- Department of Gastrointestinal Tract Surgery, Fukushima Medical University School of Medicine, Fukushima City, Japan.
| | - Kazunoshin Tachibana
- Department of Breast Surgery, Fukushima Medical University School of Medicine, Fukushima City, Japan
| | - Wataru Sakamoto
- Department of Gastrointestinal Tract Surgery, Fukushima Medical University School of Medicine, Fukushima City, Japan
| | - Katsuharu Saito
- Department of Gastrointestinal Tract Surgery, Fukushima Medical University School of Medicine, Fukushima City, Japan
| | - Aung Kyi Thar Min
- Department of Gastrointestinal Tract Surgery, Fukushima Medical University School of Medicine, Fukushima City, Japan
| | - Mai Ashizawa
- Department of Gastrointestinal Tract Surgery, Fukushima Medical University School of Medicine, Fukushima City, Japan
| | - Takahiro Nakajima
- Department of Gastrointestinal Tract Surgery, Fukushima Medical University School of Medicine, Fukushima City, Japan
| | - Keita Aoto
- Department of Gastrointestinal Tract Surgery, Fukushima Medical University School of Medicine, Fukushima City, Japan
| | - Tomoyuki Momma
- Department of Gastrointestinal Tract Surgery, Fukushima Medical University School of Medicine, Fukushima City, Japan
| | - Kyoko Katakura
- Department of Gastroenterology, Fukushima Medical University School of Medicine, Fukushima City, Japan
| | - Shinji Ohki
- Department of Gastrointestinal Tract Surgery, Fukushima Medical University School of Medicine, Fukushima City, Japan
| | - Koji Kono
- Department of Gastrointestinal Tract Surgery, Fukushima Medical University School of Medicine, Fukushima City, Japan
| |
Collapse
|
11
|
Saarinen L, Nummela P, Leinonen H, Heiskanen A, Thiel A, Haglund C, Lepistö A, Satomaa T, Hautaniemi S, Ristimäki A. Glycomic Profiling Highlights Increased Fucosylation in Pseudomyxoma Peritonei. Mol Cell Proteomics 2018; 17:2107-2118. [PMID: 30072579 DOI: 10.1074/mcp.ra118.000615] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2018] [Revised: 07/31/2018] [Indexed: 12/23/2022] Open
Abstract
Pseudomyxoma peritonei (PMP) is a subtype of mucinous adenocarcinoma that most often originates from the appendix, and grows in the peritoneal cavity filling it with mucinous ascites. KRAS and GNAS mutations are frequently found in PMP, but other common driver mutations are infrequent. As altered glycosylation can promote carcinogenesis, we compared N-linked glycan profiles of PMP tissues to those of normal appendix. Glycan profiles of eight normal appendix samples and eight low-grade and eight high-grade PMP specimens were analyzed by mass spectrometry. Our results show differences in glycan profiles between PMP and the controls, especially in those of neutral glycans, and the most prominent alteration was increased fucosylation. We further demonstrate up-regulated mRNA expression of four fucosylation-related enzymes, the core fucosylation performing fucosyltransferase 8 and three GDP-fucose biosynthetic enzymes in PMP tissues when compared with the controls. Up-regulated protein expression of the latter three enzymes was further observed in PMP cells by immunohistochemistry. We also demonstrate that restoration of fucosylation either by salvage pathway or by introduction of an expression of intact GDP-mannose 4,6-dehydratase enhance expression of MUC2, which is the predominant mucin molecule secreted by the PMP cells, in an intestinal-derived adenocarcinoma cell line with defective fucosylation because of deletion in the GDP-mannose 4,6-dehydratase gene. Thus, altered glycosylation especially in the form of fucosylation is linked to the characteristic mucin production of PMP. Glycomic data are available via ProteomeXchange with identifier PXD010086.
Collapse
Affiliation(s)
- Lilli Saarinen
- From the ‡Genome-Scale Biology Research Program, Research Programs Unit, University of Helsinki, P.O. Box 63, FI-00014 University of Helsinki, Finland
| | - Pirjo Nummela
- From the ‡Genome-Scale Biology Research Program, Research Programs Unit, University of Helsinki, P.O. Box 63, FI-00014 University of Helsinki, Finland
| | - Hannele Leinonen
- From the ‡Genome-Scale Biology Research Program, Research Programs Unit, University of Helsinki, P.O. Box 63, FI-00014 University of Helsinki, Finland
| | | | - Alexandra Thiel
- From the ‡Genome-Scale Biology Research Program, Research Programs Unit, University of Helsinki, P.O. Box 63, FI-00014 University of Helsinki, Finland
| | - Caj Haglund
- ¶Department of Surgery, University of Helsinki and Helsinki University Hospital, P.O. Box 440, FI-00029 HUS, Finland.,‖Translational Cancer Biology, Research Programs Unit, University of Helsinki, P.O. Box 63, FI-00014 University of Helsinki, Finland
| | - Anna Lepistö
- ¶Department of Surgery, University of Helsinki and Helsinki University Hospital, P.O. Box 440, FI-00029 HUS, Finland
| | - Tero Satomaa
- §Glykos Finland Ltd, Viikinkaari 6, FI-00790 Helsinki, Finland
| | - Sampsa Hautaniemi
- From the ‡Genome-Scale Biology Research Program, Research Programs Unit, University of Helsinki, P.O. Box 63, FI-00014 University of Helsinki, Finland
| | - Ari Ristimäki
- From the ‡Genome-Scale Biology Research Program, Research Programs Unit, University of Helsinki, P.O. Box 63, FI-00014 University of Helsinki, Finland; .,**Department of Pathology, HUSLAB, University of Helsinki and Helsinki University Hospital, P.O. Box 400, FI-00029 HUS, Finland
| |
Collapse
|
12
|
Noda M, Okayama H, Kofunato Y, Chida S, Saito K, Tada T, Ashizawa M, Nakajima T, Aoto K, Kikuchi T, Sakamoto W, Endo H, Fujita S, Saito M, Momma T, Ohki S, Kono K. Prognostic role of FUT8 expression in relation to p53 status in stage II and III colorectal cancer. PLoS One 2018; 13:e0200315. [PMID: 29975776 PMCID: PMC6033451 DOI: 10.1371/journal.pone.0200315] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2018] [Accepted: 06/22/2018] [Indexed: 12/29/2022] Open
Abstract
The expression of fucosyltransferase 8, an enzyme responsible for core fucosylation encoded by FUT8, influences tumor biology and correlates with patient prognosis in several solid cancers. We hypothesized that p53 alteration modifies prognostic associations of FUT8 expression in colorectal cancer (CRC), since FUT8 has recently been identified as a direct transcriptional target of wild-type p53. Utilizing multiple datasets of microarray and RNA sequence of CRC, FUT8 mRNA was found to be highly expressed in wild-type p53 tumors (n = 382) compared to those of mutant p53 (n = 437). Prognostic values of FUT8 expression in conjunction with the p53 status for disease-free survival (DFS) were analyzed using two independent cohorts of stage II and III CRC after curative surgery, including the immunohistochemistry (IHC) cohort (n = 123) and the microarray cohort (n = 357). In both cohorts, neither FUT8 expression nor the p53 status was associated with DFS. Strikingly, positive expression of FUT8 protein was significantly associated with better DFS only in tumors with negative p53, while it had no prognostic impact in tumors with positive p53 in the IHC cohort. Although not statistically significant, a similar prognostic trend was observed in the microarray cohort when patients were stratified by the p53 status. Our results suggest that the prognostic values of FUT8 expression on DFS may be modified by the p53 status, and the expression of FUT8 protein can be a prognostic biomarker for patients with stage II and III CRC.
Collapse
Affiliation(s)
- Masaru Noda
- Department of Gastrointestinal Tract Surgery, Fukushima Medical University School of Medicine, Fukushima, Japan.,Department of Breast Surgery, Fukushima Medical University School of Medicine, Fukushima, Japan
| | - Hirokazu Okayama
- Department of Gastrointestinal Tract Surgery, Fukushima Medical University School of Medicine, Fukushima, Japan
| | - Yasuhide Kofunato
- Department of Hepato-Biliary-Pancreatic and Transplant Surgery, Fukushima Medical University School of Medicine, Fukushima, Japan
| | - Shun Chida
- Department of Gastrointestinal Tract Surgery, Fukushima Medical University School of Medicine, Fukushima, Japan
| | - Katsuharu Saito
- Department of Gastrointestinal Tract Surgery, Fukushima Medical University School of Medicine, Fukushima, Japan
| | - Takeshi Tada
- Department of Gastrointestinal Tract Surgery, Fukushima Medical University School of Medicine, Fukushima, Japan
| | - Mai Ashizawa
- Department of Gastrointestinal Tract Surgery, Fukushima Medical University School of Medicine, Fukushima, Japan
| | - Takahiro Nakajima
- Department of Gastrointestinal Tract Surgery, Fukushima Medical University School of Medicine, Fukushima, Japan
| | - Keita Aoto
- Department of Gastrointestinal Tract Surgery, Fukushima Medical University School of Medicine, Fukushima, Japan
| | - Tomohiro Kikuchi
- Department of Gastrointestinal Tract Surgery, Fukushima Medical University School of Medicine, Fukushima, Japan
| | - Wataru Sakamoto
- Department of Gastrointestinal Tract Surgery, Fukushima Medical University School of Medicine, Fukushima, Japan
| | - Hisahito Endo
- Department of Gastrointestinal Tract Surgery, Fukushima Medical University School of Medicine, Fukushima, Japan
| | - Shotaro Fujita
- Department of Gastrointestinal Tract Surgery, Fukushima Medical University School of Medicine, Fukushima, Japan
| | - Motonobu Saito
- Department of Gastrointestinal Tract Surgery, Fukushima Medical University School of Medicine, Fukushima, Japan
| | - Tomoyuki Momma
- Department of Gastrointestinal Tract Surgery, Fukushima Medical University School of Medicine, Fukushima, Japan
| | - Shinji Ohki
- Department of Gastrointestinal Tract Surgery, Fukushima Medical University School of Medicine, Fukushima, Japan
| | - Koji Kono
- Department of Gastrointestinal Tract Surgery, Fukushima Medical University School of Medicine, Fukushima, Japan
| |
Collapse
|
13
|
Calderon AD, Li L, Wang PG. FUT8: from biochemistry to synthesis of core-fucosylated N-glycans. PURE APPL CHEM 2017. [DOI: 10.1515/pac-2016-0923] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Abstract
Glycosylation is a major posttranslational modification of proteins. Modification in structure on N-glycans leads to many diseases. One of such modifications is core α-1,6 fucosylation, which is only found in eukaryotes. For this reason, lots of research has been done on approaches to synthesize core-fucosylated N-glycans both chemically and enzymatically, in order to have well defined structures that can be used as probes for glycan analysis and identifying functions of glycan-binding proteins. This review will focus on FUT8, the enzyme responsible for core fucosylation in mammals and the strategies that have been developed for the synthesis of core fucosylated N-glycans have been synthesized so far.
Collapse
Affiliation(s)
- Angie D. Calderon
- Department of Chemistry and Center for Diagnostics and Therapeutics , Georgia State University , Atlanta , GA 30303 , USA
| | - Lei Li
- Department of Chemistry and Center for Diagnostics and Therapeutics , Georgia State University , Atlanta , GA 30303 , USA
| | - Peng G. Wang
- Department of Chemistry and Center for Diagnostics and Therapeutics , Georgia State University , Atlanta , GA 30303 , USA
| |
Collapse
|
14
|
Fucosylation is associated with the malignant transformation of intraductal papillary mucinous neoplasms: a lectin microarray-based study. Surg Today 2016; 46:1217-23. [PMID: 26754572 DOI: 10.1007/s00595-015-1299-8] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2015] [Accepted: 11/26/2015] [Indexed: 02/08/2023]
Abstract
PURPOSE Intraductal papillary mucinous neoplasm (IPMN) is an intraductal mucin-producing pancreatic neoplasm with the potential for malignant transformation. Changes in glycans expressed on the cell surface and glycotransferases play important roles in malignant transformation. We conducted this study to analyze glycan alterations in IPMNs by using a lectin microarray and to identify the factors associated with altered glycans and their relationships with malignant transformation. METHODS Using a lectin microarray, we evaluated glycan expression in 22 samples of IPMN with carcinoma, obtained from curative resections performed in our department. We also used immunohistochemistry to investigate fucosyltransferase 8 (Fut 8) protein expression, which is associated with glycan alterations in IPMNs. RESULTS The lectin microarray demonstrated that only two lectins, Aleuria aurantia lectin (AAL) and Aspergillus oryzae L-fucose-specific lectin (AOL), which bind to fucose, exhibited significant sequential increases from normal pancreatic duct to adenoma and carcinoma. Similarly, Fut 8 protein expression, which is associated with AAL and AOL, sequentially and significantly increased from the normal pancreatic duct to adenoma and carcinoma. CONCLUSIONS Lectin microarray analysis suggested that fucosylation is associated with the malignant transformation of IPMNs.
Collapse
|
15
|
Swierczynski S, Klieser E, Illig R, Alinger-Scharinger B, Kiesslich T, Neureiter D. Histone deacetylation meets miRNA: epigenetics and post-transcriptional regulation in cancer and chronic diseases. Expert Opin Biol Ther 2015; 15:651-64. [PMID: 25766312 DOI: 10.1517/14712598.2015.1025047] [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] [Indexed: 12/16/2022]
Abstract
INTRODUCTION Epigenetic regulation via DNA methylation, histone acetylation, as well as by microRNAs (miRNAs) is currently in the scientific focus due to its role in carcinogenesis and its involvement in initiation, progression and metastasis. While many target genes of DNA methylation, histone acetylation and miRNAs are known, even less information exists as to how these mechanisms cooperate and how they may regulate each other in a specific pathological context. For further development of therapeutic approaches, this review presents the current status of the crosstalk of histone acetylation and miRNAs in human carcinogenesis and chronic diseases. AREAS COVERED This article reviews information from comprehensive PubMed searches to evaluate relevant literature with a focus on possible association between histone acetylation, miRNAs and their targets. Our analysis identified specific miRNAs which collaborate with histone deacetylases (HDACs) and cooperatively regulate several relevant target genes. EXPERT OPINION Fourteen miRNAs could be linked to the expression of eight HDACs influencing the α-(1,6)-fucosyltransferase, polycystin-2 and the fibroblast-growth-factor 2 pathways. Focusing on the complex linkage of miRNA and HDAC expression could give deeper insights in new 'druggable' targets and might provide possible novel therapeutic approaches in future.
Collapse
Affiliation(s)
- Stefan Swierczynski
- Paracelsus Medical University, Salzburger Landeskliniken, Department of Surgery , Salzburg , Austria
| | | | | | | | | | | |
Collapse
|
16
|
Honma R, Kinoshita I, Miyoshi E, Tomaru U, Matsuno Y, Shimizu Y, Takeuchi S, Kobayashi Y, Kaga K, Taniguchi N, Dosaka-Akita H. Expression of fucosyltransferase 8 is associated with an unfavorable clinical outcome in non-small cell lung cancers. Oncology 2015; 88:298-308. [PMID: 25572677 DOI: 10.1159/000369495] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2014] [Accepted: 10/30/2014] [Indexed: 11/19/2022]
Abstract
UNLABELLED Objecitive: Fucosyltransferase 8 (FUT8), the only enzyme responsible for the core α1,6-fucosylation of asparagine-linked oligosaccharides of glycoproteins, is a vital enzyme in cancer development and progression. We examined FUT8 expression in non-small cell lung cancers (NSCLCs) to analyze its clinical significance. We also examined the expression of guanosine diphosphate-mannose-4,6-dehydratase (GMD), which is imperative for the synthesis of fucosylated oligosaccharides. METHODS Using immunohistochemistry, we evaluated the expression of FUT8 and GMD in relation to patient survival and prognosis in potentially curatively resected NSCLCs. RESULTS High expression of FUT8 was found in 67 of 129 NSCLCs (51.9%) and was significantly found in non-squamous cell carcinomas (p = 0.008). High expression of FUT8 was associated with poor survival (p = 0.03) and was also a significant and independent unfavorable prognostic factor in patients with potentially curatively resected NSCLCs (p = 0.047). High expression of GMD was significantly associated with high FUT8 expression (p = 0.04). CONCLUSIONS High expression of FUT8 is associated with an unfavorable clinical outcome in patients with potentially curatively resected NSCLCs, suggesting that FUT8 can be a prognostic factor. The analysis of FUT8 expression and its core fucosylated products may provide new insights for the therapeutic targets of NSCLCs.
Collapse
Affiliation(s)
- Rio Honma
- Department of Medical Oncology, Hokkaido University Graduate School of Medicine, Sapporo, Japan
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
17
|
Fucosylated TGF-β receptors transduces a signal for epithelial-mesenchymal transition in colorectal cancer cells. Br J Cancer 2013; 110:156-63. [PMID: 24253505 PMCID: PMC3887298 DOI: 10.1038/bjc.2013.699] [Citation(s) in RCA: 66] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Revised: 09/11/2013] [Accepted: 10/08/2013] [Indexed: 02/06/2023] Open
Abstract
Background: Transforming growth factor-β (TGF-β) is a major inducer of epithelial–mesenchymal transition (EMT) in different cell types. TGF-β-mediated EMT is thought to contribute to tumour cell spread and metastasis. Sialyl Lewis antigens synthesised by fucosyltransferase (FUT) 3 and FUT6 are highly expressed in patients with metastatic colorectal cancer (CRC) and are utilised as tumour markers for cancer detection and evaluation of treatment efficacy. However, the role of FUT3 and FUT6 in augmenting the malignant potential of CRC induced by TGF-β is unclear. Methods: Colorectal cancer cell lines were transfected with siRNAs for FUT3/6 and were examined by cell proliferation, invasion and migration assays. The expression and phosphorylation status of TGF-β downstream molecules were analysed by western blot. Fucosylation of TGF-β receptor (TβR) was examined by lectin blot analysis. Results: Inhibition of FUT3/6 expression by siRNAs suppressed the fucosylation of type I TβR and phosphorylation of the downstream molecules, thereby inhibiting the invasion and migration of CRC cells by EMT. Conclusion: Fucosyltransferase 3/6 has an essential role in cancer cell adhesion to endothelial cells by upregulation of sialyl Lewis antigens and also by enhancement of cancer cell migration through TGF-β-mediated EMT.
Collapse
|
18
|
Mol BM, Massink MPG, van der Hout AH, Dommering CJ, Zaman JMA, Bosscha MI, Kors WA, Meijers-Heijboer HE, Kaspers GJL, Riele HT, Moll AC, Cloos J, Dorsman JC. High resolution SNP array profiling identifies variability in retinoblastoma genome stability. Genes Chromosomes Cancer 2013; 53:1-14. [PMID: 24249257 DOI: 10.1002/gcc.22111] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2013] [Accepted: 08/21/2013] [Indexed: 12/18/2022] Open
Abstract
Both hereditary and nonhereditary retinoblastoma (Rb) are commonly initiated by loss of both copies of the retinoblastoma tumor suppressor gene (RB1), while additional genomic changes are required for tumor initiation and progression. Our aim was to determine whether there is genomic heterogeneity between different clinical Rb subtypes. Therefore, 21 Rb tumors from 11 hereditary patients and 10 nonhereditary Rb patients were analyzed using high-resolution single nucleotide polymorphism (SNP) arrays and gene losses and gains were validated with Multiplex Ligation-dependent Probe Amplification. In these tumors only a few focal aberrations were detected. The most frequent was a focal gain on chromosome 2p24.3, the minimal region of gain encompassing the oncogene MYCN. The genes BAZ1A, OTX2, FUT8, and AKT1 were detected in four focal regions on chromosome 14 in one nonhereditary Rb. There was a large difference in number of copy number aberrations between tumors. A subset of nonhereditary Rbs turned out to be the most genomic unstable, while especially very young patients with hereditary Rb display stable genomes. Established Rb copy number aberrations, including gain of chromosome arm 1q and loss of chromosome arm 16q, turned out to be preferentially associated with the nonhereditary Rbs with later age of diagnosis. In contrast, copy number neutral loss of heterozygosity was detected mainly on chromosome 13, where RB1 resides, irrespective of hereditary status or age. Focal amplifications and deletions and copy number neutral loss of heterozygosity besides chromosome 13 appear to be rare events in retinoblastoma.
Collapse
Affiliation(s)
- Berber M Mol
- Department of Clinical Genetics, VU University Medical Center, Amsterdam, The Netherlands
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
19
|
Villar-Portela S, Muinelo-Romay L, Cuevas E, Gil-Martín E, Fernández-Briera A. FX enzyme and GDP-L-Fuc transporter expression in colorectal cancer. Histopathology 2013; 63:174-86. [PMID: 23730929 DOI: 10.1111/his.12157] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2013] [Accepted: 03/29/2013] [Indexed: 11/28/2022]
Abstract
AIMS Fucosylation is regulated by fucosyltransferases, the guanosine diphosphate-L-fucose (GDP-L-Fuc) synthetic pathway, and the GDP-L-fucose transporter (GDP-L-Fuc Tr). We have reported previously an increased level of α(1,6)fucosyltransferase activity and expression in colorectal cancer (CRC). The present study aimed to analyse the expression profiles of the FX enzyme and GDP-L-Fuc Tr in a cohort of operated CRC patients to elucidate their role in α(1,6)fucosylation in this neoplasm. METHODS AND RESULTS We assessed the immunohistochemical expression of FX and GDP-L-Fuc Tr in a series of tumour samples and healthy tissues from CRC specimens. FX expression was observed in 58 of 91 (63.7%) tumours and 23 of 28 (82.1%) corresponding healthy samples. GDP-L-Fuc Tr expression was detected in 86 of 102 (84.3%) colorectal tumours, and 13 of 27 (48.1%) healthy tissue specimens. The expression of GDP-L-Fuc Tr was statistically higher in tumours than in healthy tissues (P < 0.001). A correlation was found between FX and GDP-L-Fuc Tr expression in tumour samples (P = 0.003). CONCLUSION GDP-L-Fuc Tr overexpression in the tumour tissue of CRC patients suggests that GDP-L-Fuc transport to the Golgi apparatus may be an important factor associated with increased α(1,6)fucosylation in CRC.
Collapse
Affiliation(s)
- Susana Villar-Portela
- Department of Biochemistry, Genetics and Immunology, Faculty of Biology, University of Vigo, Vigo, Spain
| | | | | | | | | |
Collapse
|
20
|
Expression of alpha 1,6-fucosyltransferase 8 in hepatitis B virus-related hepatocellular carcinoma influences tumour progression. Dig Liver Dis 2013; 45:414-21. [PMID: 23352314 DOI: 10.1016/j.dld.2012.12.007] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/04/2012] [Revised: 11/23/2012] [Accepted: 12/03/2012] [Indexed: 02/07/2023]
Abstract
BACKGROUND Alpha 1,6-fucosyltransferase 8 expression was demonstrated to be enhanced during hepatocarcinogenesis. AIMS Our study aimed to find out the clinical significance and biological function of alpha 1,6-fucosyltransferase 8 in hepatitis B virus-related hepatocellular carcinoma. METHODS Alpha 1,6-fucosyltransferase 8 expression levels were determined in 52 pairs of tissues to compare its expression between tumour tissues [with/without portal vein tumour thrombus] and adjacent noncancerous liver tissues. Relationship between alpha 1,6-fucosyltransferase 8 expression and clinical indicators was also investigated. An alpha 1,6-fucosyltransferase 8-knockdown (by RNAi) cell line MHCC97-H/siFUT8 was established to reveal functional impact of alpha 1,6-fucosyltransferase 8 on cell growth, migration and invasion in hepatocellular carcinoma, respectively using Cell Counting Kit-8, wound healing migration assay, transwell assay and gelatin zymography. RESULTS We observed a higher alpha 1,6-fucosyltransferase 8 expression level in tumour tissues than adjacent noncancerous liver tissues. In portal vein tumour thrombus group, alpha 1,6-fucosyltransferase 8 protein expressed more in portal vein tumour thrombus tissues than that in adjacent noncancerous liver tissues. The expression level in tumour tissues was highly correlated with tumour size and presence of satellite nodules (P<0.05). Furthermore, alpha 1,6-fucosyltransferase 8-knockdown suppressed the tumour proliferation, migration and invasion of MHCC97-H cells. CONCLUSION These findings suggest that alpha 1,6-fucosyltransferase 8 expression might be a good indicator of poor prognosis in hepatocellular carcinoma. High alpha 1,6-fucosyltransferase 8 expression may play an important role in hepatitis B virus-related hepatocellular carcinoma progression.
Collapse
|
21
|
Chen YT, Chong YM, Cheng CW, Ho CL, Tsai HW, Kasten FH, Chen YL, Chang CF. Identification of novel tumor markers for oral squamous cell carcinoma using glycoproteomic analysis. Clin Chim Acta 2012; 420:45-53. [PMID: 23078850 DOI: 10.1016/j.cca.2012.10.019] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2012] [Accepted: 10/10/2012] [Indexed: 01/22/2023]
Abstract
BACKGROUND Oral cancer, the largest subset of head and neck cancer, has become one of the most lethal malignancies during the last two decades. Although several diagnostic tools have been applied for the early detection of oral malignancies, it is still urgent to identify novel tumor markers. In this study, we explored the cell surface N-glycomes of primary cultured human oral keratinocytes (HOK), immortalized human gingival keratinocytes (SG cells), and oral squamous cell carcinoma (OC2). METHODS Enzymatically hydrolyzed cell surface N-glycans were analyzed by MALDI-TOF mass spectrometry. RESULTS High levels of fucosylated N-glycans, especially core-fucosylated N-glycans, were observed on the OC2 cell surface whereas the major N-glycans on SG and HOK cells were high mannose type. In addition, the mRNA expression level of fucosyltransferase 8 was elevated significantly in OC2 cells than in SG and HOK cells. Core-fucosylated glycoproteins of OC2 cells were then purified with lectin affinity chromatography and a key adhesion molecule in cancer cells, CD147, was identified. Finally, overexpression of cell surface CD147 was confirmed on OC2 cells and oral cancer tissues (tissue array). CONCLUSIONS CD147 was discovered by glycoproteomic approaches and suggested to be a potential novel tumor marker for oral cancer diagnosis.
Collapse
Affiliation(s)
- Yi-Ting Chen
- Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan 70101, Taiwan
| | | | | | | | | | | | | | | |
Collapse
|
22
|
Muinelo-Romay L, Villar-Portela S, Cuevas E, Gil-Martín E, Fernández-Briera A. Identification of α(1,6)fucosylated proteins differentially expressed in human colorectal cancer. BMC Cancer 2011; 11:508. [PMID: 22152070 PMCID: PMC3297542 DOI: 10.1186/1471-2407-11-508] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2011] [Accepted: 12/07/2011] [Indexed: 12/30/2022] Open
Abstract
Summary
Collapse
Affiliation(s)
- Laura Muinelo-Romay
- Department of Biochemistry, Genetics and Immunology, Faculty of Biology, University of Vigo, Campus As Lagoas-Marcosende S/N, 36310 Vigo, Spain
| | | | | | | | | |
Collapse
|