1
|
Wolters-Eisfeld G, Oliveira-Ferrer L. Glycan diversity in ovarian cancer: Unraveling the immune interplay and therapeutic prospects. Semin Immunopathol 2024; 46:16. [PMID: 39432076 PMCID: PMC11493797 DOI: 10.1007/s00281-024-01025-6] [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: 11/09/2023] [Accepted: 09/12/2024] [Indexed: 10/22/2024]
Abstract
Ovarian cancer remains a formidable challenge in oncology due to its late-stage diagnosis and limited treatment options. Recent research has revealed the intricate interplay between glycan diversity and the immune microenvironment within ovarian tumors, shedding new light on potential therapeutic strategies. This review seeks to investigate the complex role of glycans in ovarian cancer and their impact on the immune response. Glycans, complex sugar molecules decorating cell surfaces and secreted proteins, have emerged as key regulators of immune surveillance in ovarian cancer. Aberrant glycosylation patterns can promote immune evasion by shielding tumor cells from immune recognition, enabling disease progression. Conversely, certain glycan structures can modulate the immune response, leading to either antitumor immunity or immune tolerance. Understanding the intricate relationship between glycan diversity and immune interactions in ovarian cancer holds promise for the development of innovative therapeutic approaches. Immunotherapies that target glycan-mediated immune evasion, such as glycan-based vaccines or checkpoint inhibitors, are under investigation. Additionally, glycan profiling may serve as a diagnostic tool for patient stratification and treatment selection. This review underscores the emerging importance of glycan diversity in ovarian cancer, emphasizing the potential for unraveling immune interplay and advancing tailored therapeutic prospects for this devastating disease.
Collapse
Affiliation(s)
- Gerrit Wolters-Eisfeld
- Department of General, Visceral and Thoracic Surgery, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | | |
Collapse
|
2
|
Batara DC, Kim HJ, Phan LT, Kim M, Son YO, Lee S, Park SI, Choi YS, Beck S, Kim SH. Elevated α-1,2-mannosidase MAN1C1 in glioma stem cells and its implications for immunological changes and prognosis in glioma patients. Sci Rep 2024; 14:22159. [PMID: 39333557 PMCID: PMC11436702 DOI: 10.1038/s41598-024-72901-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Accepted: 09/11/2024] [Indexed: 09/29/2024] Open
Abstract
Glioblastoma multiforme (GBM) is the most aggressive type of primary brain tumor, and the presence of glioma stem cells (GSCs) has been linked to its resistance to treatments and recurrence. Additionally, aberrant glycosylation has been implicated in the aggressiveness of cancers. However, the influence and underlying mechanism of N-glycosylation on the GSC phenotype and GBM malignancy remain elusive. Here, we performed an in-silico analysis approach on publicly available datasets to examine the function of N-glycosylation-related genes in GSCs and gliomas, accompanied by a qRT-PCR validation experiment. We found that high α-1,2-mannosidase MAN1C1 is associated with immunological functions and worse survival of glioma patients. Differential gene expression analysis and qRT-PCR validation revealed that MAN1C1 is highly expressed in GSCs. Furthermore, higher MAN1C1 expression predicts worse outcomes in glioma patients. Also, MAN1C1 expression is increased in the perinecrotic region of GBM and is associated with immunological and inflammatory functions, a hallmark of the GBM mesenchymal subtype. Further analysis confirmed that MAN1C1 expression is closely associated with infiltrating immune cells and disrupted immune response in the GBM microenvironment. These suggest that MAN1C1 is a potential biomarker for gliomas and may be important as an immunotherapeutic target for GBM.
Collapse
Affiliation(s)
- Don Carlo Batara
- Animal Molecular Biochemistry Laboratory, Department of Animal Science, College of Agriculture and Life Sciences, Chonnam National University, Gwangju, 61186, Republic of Korea
| | - Hyun-Jin Kim
- Animal Molecular Biochemistry Laboratory, Department of Animal Science, College of Agriculture and Life Sciences, Chonnam National University, Gwangju, 61186, Republic of Korea
| | - Le Thi Phan
- Animal Molecular Biochemistry Laboratory, Department of Animal Science, College of Agriculture and Life Sciences, Chonnam National University, Gwangju, 61186, Republic of Korea
- Computational Biology and Bioinformatics Laboratory, Department of Integrative Biotechnology, Sungkyunkwan University, Suwon, Gyeonggi-do, 16419, Republic of Korea
| | - Minseo Kim
- Animal Molecular Biochemistry Laboratory, Department of Animal Science, College of Agriculture and Life Sciences, Chonnam National University, Gwangju, 61186, Republic of Korea
| | - Young-Ok Son
- Department of Animal Biotechnology, Faculty of Biotechnology, College of Applied Life Sciences, Interdisciplinary Graduate Program in Advanced Convergence Technology and Science, Jeju National University, Jeju, 63243, Republic of Korea
| | - Seongsoo Lee
- Gwangju Center, Korea Basic Science Institute (KBSI), 49, Dosicheomdansaneop-ro, Nam-gu, Gwangju, 61751, Republic of Korea
- Department of Systems Biotechnology, Chung-Ang University, Anseong-si, Gyeonggi-do, 17546, Republic of Korea
| | - Sang-Ik Park
- Laboratory of Veterinary Pathology, College of Veterinary Medicine and BK21 Plus Project Team, Chonnam National University, Gwangju, 61186, Republic of Korea
| | - Young Sun Choi
- Jeollanam-do Agriculture Research and Extension Services Livestock Research Institute, Naju-si, Jeollanam-do, 58213, Republic of Korea
| | - Samuel Beck
- Department of Dermatology, Center for Aging Research, Chobanian & Avedisian School of Medicine, Boston University, Boston, 02118, USA.
| | - Sung-Hak Kim
- Animal Molecular Biochemistry Laboratory, Department of Animal Science, College of Agriculture and Life Sciences, Chonnam National University, Gwangju, 61186, Republic of Korea.
| |
Collapse
|
3
|
Cooper TT, Dieters-Castator DZ, Liu J, Siegers GM, Pink D, Veliz L, Lewis JD, Lagugné-Labarthet F, Fu Y, Steed H, Lajoie GA, Postovit LM. Targeted proteomics of plasma extracellular vesicles uncovers MUC1 as combinatorial biomarker for the early detection of high-grade serous ovarian cancer. J Ovarian Res 2024; 17:149. [PMID: 39020428 PMCID: PMC11253408 DOI: 10.1186/s13048-024-01471-8] [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: 03/22/2024] [Accepted: 07/02/2024] [Indexed: 07/19/2024] Open
Abstract
BACKGROUND The five-year prognosis for patients with late-stage high-grade serous carcinoma (HGSC) remains dismal, underscoring the critical need for identifying early-stage biomarkers. This study explores the potential of extracellular vesicles (EVs) circulating in blood, which are believed to harbor proteomic cargo reflective of the HGSC microenvironment, as a source for biomarker discovery. RESULTS We conducted a comprehensive proteomic profiling of EVs isolated from blood plasma, ascites, and cell lines of patients, employing both data-dependent (DDA) and data-independent acquisition (DIA) methods to construct a spectral library tailored for targeted proteomics. Our investigation aimed at uncovering novel biomarkers for the early detection of HGSC by comparing the proteomic signatures of EVs from women with HGSC to those with benign gynecological conditions. The initial cohort, comprising 19 donors, utilized DDA proteomics for spectral library development. The subsequent cohort, involving 30 HGSC patients and 30 control subjects, employed DIA proteomics for a similar purpose. Support vector machine (SVM) classification was applied in both cohorts to identify combinatorial biomarkers with high specificity and sensitivity (ROC-AUC > 0.90). Notably, MUC1 emerged as a significant biomarker in both cohorts when used in combination with additional biomarkers. Validation through an ELISA assay on a subset of benign (n = 18), Stage I (n = 9), and stage II (n = 9) plasma samples corroborated the diagnostic utility of MUC1 in the early-stage detection of HGSC. CONCLUSIONS This study highlights the value of EV-based proteomic analysis in the discovery of combinatorial biomarkers for early ovarian cancer detection.
Collapse
Affiliation(s)
- Tyler T Cooper
- Department of Biochemistry, Western University, London, ON, Canada
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, ON, Canada
| | | | - Jiahui Liu
- Department of Oncology, University of Alberta, Edmonton, AB, Canada
| | | | - Desmond Pink
- Department of Oncology, University of Alberta, Edmonton, AB, Canada
| | - Lorena Veliz
- Department of Chemistry, Western University, London, ON, Canada
| | - John D Lewis
- Department of Oncology, University of Alberta, Edmonton, AB, Canada
| | | | - Yangxin Fu
- Department of Oncology, University of Alberta, Edmonton, AB, Canada
| | - Helen Steed
- Department of Obstetrics and Gynecology, University of Alberta, Edmonton, AB, Canada
| | - Gilles A Lajoie
- Department of Biochemistry, Western University, London, ON, Canada.
| | - Lynne-Marie Postovit
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, ON, Canada.
- Department of Oncology, University of Alberta, Edmonton, AB, Canada.
- Department of Obstetrics and Gynecology, University of Alberta, Edmonton, AB, Canada.
| |
Collapse
|
4
|
Di Gregorio J, Di Giuseppe L, Terreri S, Rossi M, Battafarano G, Pagliarosi O, Flati V, Del Fattore A. Protein Stability Regulation in Osteosarcoma: The Ubiquitin-like Modifications and Glycosylation as Mediators of Tumor Growth and as Targets for Therapy. Cells 2024; 13:537. [PMID: 38534381 PMCID: PMC10969184 DOI: 10.3390/cells13060537] [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: 02/14/2024] [Revised: 03/11/2024] [Accepted: 03/16/2024] [Indexed: 03/28/2024] Open
Abstract
The identification of new therapeutic targets and the development of innovative therapeutic approaches are the most important challenges for osteosarcoma treatment. In fact, despite being relatively rare, recurrence and metastatic potential, particularly to the lungs, make osteosarcoma a deadly form of cancer. In fact, although current treatments, including surgery and chemotherapy, have improved survival rates, the disease's recurrence and metastasis are still unresolved complications. Insights for analyzing the still unclear molecular mechanisms of osteosarcoma development, and for finding new therapeutic targets, may arise from the study of post-translational protein modifications. Indeed, they can influence and alter protein structure, stability and function, and cellular interactions. Among all the post-translational modifications, ubiquitin-like modifications (ubiquitination, deubiquitination, SUMOylation, and NEDDylation), as well as glycosylation, are the most important for regulating protein stability, which is frequently altered in cancers including osteosarcoma. This review summarizes the relevance of ubiquitin-like modifications and glycosylation in osteosarcoma progression, providing an overview of protein stability regulation, as well as highlighting the molecular mediators of these processes in the context of osteosarcoma and their possible targeting for much-needed novel therapy.
Collapse
Affiliation(s)
- Jacopo Di Gregorio
- Department of Biotechnological and Applied Clinical Sciences, University of L’Aquila, 67100 L’Aquila, Italy;
| | - Laura Di Giuseppe
- Department of Clinical, Internal, Anaesthesiological and Cardiovascular Sciences, Sapienza University, 00185 Rome, Italy;
| | - Sara Terreri
- Bone Physiopathology Research Unit, Translational Pediatrics and Clinical Genetics Research Division, Bambino Gesù Children’s Hospital, IRCCS, 00146 Rome, Italy; (S.T.); (M.R.); (G.B.); (O.P.); (A.D.F.)
| | - Michela Rossi
- Bone Physiopathology Research Unit, Translational Pediatrics and Clinical Genetics Research Division, Bambino Gesù Children’s Hospital, IRCCS, 00146 Rome, Italy; (S.T.); (M.R.); (G.B.); (O.P.); (A.D.F.)
| | - Giulia Battafarano
- Bone Physiopathology Research Unit, Translational Pediatrics and Clinical Genetics Research Division, Bambino Gesù Children’s Hospital, IRCCS, 00146 Rome, Italy; (S.T.); (M.R.); (G.B.); (O.P.); (A.D.F.)
| | - Olivia Pagliarosi
- Bone Physiopathology Research Unit, Translational Pediatrics and Clinical Genetics Research Division, Bambino Gesù Children’s Hospital, IRCCS, 00146 Rome, Italy; (S.T.); (M.R.); (G.B.); (O.P.); (A.D.F.)
| | - Vincenzo Flati
- Department of Biotechnological and Applied Clinical Sciences, University of L’Aquila, 67100 L’Aquila, Italy;
| | - Andrea Del Fattore
- Bone Physiopathology Research Unit, Translational Pediatrics and Clinical Genetics Research Division, Bambino Gesù Children’s Hospital, IRCCS, 00146 Rome, Italy; (S.T.); (M.R.); (G.B.); (O.P.); (A.D.F.)
| |
Collapse
|
5
|
Martins M, Vieira J, Pereira-Leite C, Saraiva N, Fernandes AS. The Golgi Apparatus as an Anticancer Therapeutic Target. BIOLOGY 2023; 13:1. [PMID: 38275722 PMCID: PMC10813373 DOI: 10.3390/biology13010001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/16/2023] [Revised: 12/12/2023] [Accepted: 12/15/2023] [Indexed: 01/27/2024]
Abstract
Although the discovery of the Golgi apparatus (GA) was made over 125 years ago, only a very limited number of therapeutic approaches have been developed to target this complex organelle. The GA serves as a modification and transport center for proteins and lipids and also has more recently emerged as an important store for some ions. The dysregulation of GA functions is implicated in many cellular processes associated with cancer and some GA proteins are indeed described as cancer biomarkers. This dysregulation can affect protein modification, localization, and secretion, but also cellular metabolism, redox status, extracellular pH, and the extracellular matrix structure. Consequently, it can directly or indirectly affect cancer progression. For these reasons, the GA is an appealing anticancer pharmacological target. Despite this, no anticancer drug specifically targeting the GA has reached the clinic and few have entered the clinical trial stage. Advances in nanodelivery approaches may help change this scenario by specifically targeting tumor cells and/or the GA through passive, active, or physical strategies. This article aims to examine the currently available anticancer GA-targeted drugs and the nanodelivery strategies explored for their administration. The potential benefits and challenges of modulating and specifically targeting the GA function in the context of cancer therapy are discussed.
Collapse
Affiliation(s)
- Marta Martins
- CBIOS—Universidade Lusófona’s Research Center for Biosciences & Health Technologies, Campo Grande 376, 1749-024 Lisboa, Portugal; (M.M.); (J.V.); (C.P.-L.)
- Department of Biomedical Sciences, University of Alcalá, Ctra. Madrid-Barcelona Km. 33.600, Alcalá de Henares, 28871 Madrid, Spain
| | - João Vieira
- CBIOS—Universidade Lusófona’s Research Center for Biosciences & Health Technologies, Campo Grande 376, 1749-024 Lisboa, Portugal; (M.M.); (J.V.); (C.P.-L.)
- Department of Biomedical Sciences, University of Alcalá, Ctra. Madrid-Barcelona Km. 33.600, Alcalá de Henares, 28871 Madrid, Spain
| | - Catarina Pereira-Leite
- CBIOS—Universidade Lusófona’s Research Center for Biosciences & Health Technologies, Campo Grande 376, 1749-024 Lisboa, Portugal; (M.M.); (J.V.); (C.P.-L.)
- LAQV, REQUIMTE, Departamento de Ciências Químicas, Faculdade de Farmácia, Universidade do Porto, Rua de Jorge Viterbo Ferreira 228, 4050-313 Porto, Portugal
| | - Nuno Saraiva
- CBIOS—Universidade Lusófona’s Research Center for Biosciences & Health Technologies, Campo Grande 376, 1749-024 Lisboa, Portugal; (M.M.); (J.V.); (C.P.-L.)
| | - Ana Sofia Fernandes
- CBIOS—Universidade Lusófona’s Research Center for Biosciences & Health Technologies, Campo Grande 376, 1749-024 Lisboa, Portugal; (M.M.); (J.V.); (C.P.-L.)
| |
Collapse
|
6
|
Senjor E, Pirro M, Švajger U, Prunk M, Sabotič J, Jewett A, Hensbergen PJ, Perišić Nanut M, Kos J. Different glycosylation profiles of cystatin F alter the cytotoxic potential of natural killer cells. Cell Mol Life Sci 2023; 81:8. [PMID: 38092995 PMCID: PMC10719177 DOI: 10.1007/s00018-023-05041-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2023] [Revised: 10/13/2023] [Accepted: 11/06/2023] [Indexed: 12/17/2023]
Abstract
Cystatin F, a cysteine peptidase inhibitor, is a potent modulator of NK cytotoxicity. By inhibiting granule-mediated cytotoxicity pathway, cystatin F induces formation of non-functional NK cell stage, called split-anergy. We show that N-glycosylation determines the localization and cellular function of cystatin F. Cystatin F mostly exhibited high-mannose glycosylation in U-937 cells, both high-mannose and complex glycosylation in NK-92 and primary NKs, and predominantly complex glycosylation in super-charged NKs. Manipulating N-glycosylation with kifunensine increased high-mannose glycosylation of cystatin F and lysosome localisation, which decreased cathepsin C activity and reduced NK cytotoxicity. Mannose-6-phosphate could significantly reduce the internalization of extracellular cystatin F. By comparing NK cells with different cytotoxic potentials, we found that high-mannose cystatin F was strongly associated with lysosomes and cathepsin C in NK-92 cell line. In contrast, in highly cytotoxic super-charged NKs, cystatin F with complex glycosylation was associated with the secretory pathway and less prone to inhibit cathepsin C. Modulating glycosylation to alter cystatin F localisation could increase the cytotoxicity of NK cells, thereby enhancing their therapeutic potential for treating cancer patients.
Collapse
Affiliation(s)
- Emanuela Senjor
- Department of Biotechnology, Jožef Stefan Institute, Ljubljana, Slovenia
- Faculty of Pharmacy, University of Ljubljana, Aškerčeva Cesta 7, 1000, Ljubljana, Slovenia
| | - Martina Pirro
- Center for Proteomics and Metabolomics, Leiden University Medical Center, Leiden, The Netherlands
| | - Urban Švajger
- Faculty of Pharmacy, University of Ljubljana, Aškerčeva Cesta 7, 1000, Ljubljana, Slovenia
- Blood Transfusion Centre of Slovenia, Ljubljana, Slovenia
| | - Mateja Prunk
- Department of Biotechnology, Jožef Stefan Institute, Ljubljana, Slovenia
| | - Jerica Sabotič
- Department of Biotechnology, Jožef Stefan Institute, Ljubljana, Slovenia
| | - Anahid Jewett
- Division of Oral Biology and Medicine, The Jane and Jerry Weintraub Center for Reconstructive Biotechnology, School of Dentistry, University of California Los Angeles, Los Angeles, USA
- The Jonsson Comprehensive Cancer Center, Los Angeles, USA
| | - Paul J Hensbergen
- Center for Proteomics and Metabolomics, Leiden University Medical Center, Leiden, The Netherlands
| | | | - Janko Kos
- Department of Biotechnology, Jožef Stefan Institute, Ljubljana, Slovenia.
- Faculty of Pharmacy, University of Ljubljana, Aškerčeva Cesta 7, 1000, Ljubljana, Slovenia.
| |
Collapse
|
7
|
Males A, Kok K, Nin-Hill A, de Koster N, van den Beukel S, Beenakker TJM, van der Marel GA, Codée JDC, Aerts JMFG, Overkleeft HS, Rovira C, Davies GJ, Artola M. Trans-cyclosulfamidate mannose-configured cyclitol allows isoform-dependent inhibition of GH47 α-d-mannosidases through a bump-hole strategy. Chem Sci 2023; 14:13581-13586. [PMID: 38033892 PMCID: PMC10685318 DOI: 10.1039/d3sc05016e] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2023] [Accepted: 10/29/2023] [Indexed: 12/02/2023] Open
Abstract
Class I inverting exo-acting α-1,2-mannosidases (CAZY family GH47) display an unusual catalytic itinerary featuring ring-flipped mannosides, 3S1 → 3H4‡ → 1C4. Conformationally locked 1C4 compounds, such as kifunensine, display nanomolar inhibition but large multigene GH47 mannosidase families render specific "isoform-dependent" inhibition impossible. Here we develop a bump-and-hole strategy in which a new mannose-configured 1,6-trans-cyclic sulfamidate inhibits α-d-mannosidases by virtue of its 1C4 conformation. This compound does not inhibit the wild-type GH47 model enzyme by virtue of a steric clash, a "bump", in the active site. An L310S (a conserved residue amongst human GH47 enzymes) mutant of the model Caulobacter GH47 awoke 574 nM inhibition of the previously dormant inhibitor, confirmed by structural analysis of a 0.97 Å structure. Considering that L310 is a conserved residue amongst human GH47 enzymes, this work provides a unique framework for future biotechnological studies on N-glycan maturation and ER associated degradation by isoform-specific GH47 α-d-mannosidase inhibition through a bump-and-hole approach.
Collapse
Affiliation(s)
- Alexandra Males
- York Structural Biology Laboratory, Department of Chemistry, The University of York York YO10 5DD UK
| | - Ken Kok
- Department of Medical Biochemistry, Leiden Institute of Chemistry (LIC), Leiden University P. O. Box 9502 2300 RA Leiden The Netherlands
| | - Alba Nin-Hill
- Departament de Química Inorgànica i Orgànica (Secció de Química Orgànica), Institut de Química Teòrica i Computacional (IQTCUB), Universitat de Barcelona Martí i Franquès 1 08028 Barcelona Spain
- Fundació Catalana de Recerca i Estudis Avançats (ICREA) Passeig Lluís Companys 23 08010 Barcelona Spain
| | - Nicky de Koster
- Department of Medical Biochemistry, Leiden Institute of Chemistry (LIC), Leiden University P. O. Box 9502 2300 RA Leiden The Netherlands
| | - Sija van den Beukel
- Department of Medical Biochemistry, Leiden Institute of Chemistry (LIC), Leiden University P. O. Box 9502 2300 RA Leiden The Netherlands
| | - Thomas J M Beenakker
- Department of Bio-organic Synthesis, Leiden Institute of Chemistry (LIC), Leiden University Einsteinweg 55 2333 CC Leiden The Netherlands
| | - Gijsbert A van der Marel
- Department of Bio-organic Synthesis, Leiden Institute of Chemistry (LIC), Leiden University Einsteinweg 55 2333 CC Leiden The Netherlands
| | - Jeroen D C Codée
- Department of Bio-organic Synthesis, Leiden Institute of Chemistry (LIC), Leiden University Einsteinweg 55 2333 CC Leiden The Netherlands
| | - Johannes M F G Aerts
- Department of Medical Biochemistry, Leiden Institute of Chemistry (LIC), Leiden University P. O. Box 9502 2300 RA Leiden The Netherlands
| | - Herman S Overkleeft
- Department of Bio-organic Synthesis, Leiden Institute of Chemistry (LIC), Leiden University Einsteinweg 55 2333 CC Leiden The Netherlands
| | - Carme Rovira
- Departament de Química Inorgànica i Orgànica (Secció de Química Orgànica), Institut de Química Teòrica i Computacional (IQTCUB), Universitat de Barcelona Martí i Franquès 1 08028 Barcelona Spain
- Fundació Catalana de Recerca i Estudis Avançats (ICREA) Passeig Lluís Companys 23 08010 Barcelona Spain
| | - Gideon J Davies
- York Structural Biology Laboratory, Department of Chemistry, The University of York York YO10 5DD UK
| | - Marta Artola
- Department of Medical Biochemistry, Leiden Institute of Chemistry (LIC), Leiden University P. O. Box 9502 2300 RA Leiden The Netherlands
| |
Collapse
|
8
|
Getmantseva L, Kolosova M, Fede K, Korobeinikova A, Kolosov A, Romanets E, Bakoev F, Romanets T, Yudin V, Keskinov A, Bakoev S. Finding Predictors of Leg Defects in Pigs Using CNV-GWAS. Genes (Basel) 2023; 14:2054. [PMID: 38002997 PMCID: PMC10671522 DOI: 10.3390/genes14112054] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2023] [Revised: 11/04/2023] [Accepted: 11/07/2023] [Indexed: 11/26/2023] Open
Abstract
One of the most important areas of modern genome research is the search for meaningful relationships between genetic variants and phenotypes. In the livestock field, there has been research demonstrating the influence of copy number variants (CNVs) on phenotypic variation. Despite the wide range in the number and size of detected CNVs, a significant proportion differ between breeds and their functional effects are underestimated in the pig industry. In this work, we focused on the problem of leg defects in pigs (lumps/growths in the area of the hock joint on the hind legs) and focused on searching for molecular genetic predictors associated with this trait for the selection of breeding stock. The study was conducted on Large White pigs using three CNV calling tools (PennCNV, QuantiSNP and R-GADA) and the CNVRanger association analysis tool (CNV-GWAS). As a result, the analysis identified three candidate CNVRs associated with the formation of limb defects. Subsequent functional analysis suggested that all identified CNVs may act as potential predictors of the hock joint phenotype of pigs. It should be noted that the results obtained indicate that all significant regions are localized in genes (CTH, SRSF11, MAN1A1 and LPIN1) responsible for the metabolism of amino acids, fatty acids, glycerolipids and glycerophospholipids, thereby related to the immune response, liver functions, content intramuscular fat and animal fatness. These results are consistent with previously published studies, according to which a predisposition to the formation of leg defects can be realized through genetic variants associated with the functions of the liver, kidneys and hematological characteristics.
Collapse
Affiliation(s)
- Lyubov Getmantseva
- Federal State Budgetary Educational Institution of Higher Education, Don State Agrarian University, 346493 Persianovsky, Russia; (L.G.); (A.K.)
- Federal State Budgetary Institution, “Center for Strategic Planning and Management of Medical and Biological Health Risks” of the Federal Medical and Biological Agency, 10/1 Pogodinskaya St., 119121 Moscow, Russia; (K.F.); (A.K.)
| | - Maria Kolosova
- Federal State Budgetary Educational Institution of Higher Education, Don State Agrarian University, 346493 Persianovsky, Russia; (L.G.); (A.K.)
| | - Kseniia Fede
- Federal State Budgetary Institution, “Center for Strategic Planning and Management of Medical and Biological Health Risks” of the Federal Medical and Biological Agency, 10/1 Pogodinskaya St., 119121 Moscow, Russia; (K.F.); (A.K.)
| | - Anna Korobeinikova
- Federal State Budgetary Institution, “Center for Strategic Planning and Management of Medical and Biological Health Risks” of the Federal Medical and Biological Agency, 10/1 Pogodinskaya St., 119121 Moscow, Russia; (K.F.); (A.K.)
| | - Anatoly Kolosov
- Federal State Budgetary Educational Institution of Higher Education, Don State Agrarian University, 346493 Persianovsky, Russia; (L.G.); (A.K.)
| | - Elena Romanets
- Federal State Budgetary Educational Institution of Higher Education, Don State Agrarian University, 346493 Persianovsky, Russia; (L.G.); (A.K.)
| | - Faridun Bakoev
- Federal State Budgetary Educational Institution of Higher Education, Don State Agrarian University, 346493 Persianovsky, Russia; (L.G.); (A.K.)
| | - Timofey Romanets
- Federal State Budgetary Educational Institution of Higher Education, Don State Agrarian University, 346493 Persianovsky, Russia; (L.G.); (A.K.)
| | - Vladimir Yudin
- Federal State Budgetary Institution, “Center for Strategic Planning and Management of Medical and Biological Health Risks” of the Federal Medical and Biological Agency, 10/1 Pogodinskaya St., 119121 Moscow, Russia; (K.F.); (A.K.)
| | - Anton Keskinov
- Federal State Budgetary Institution, “Center for Strategic Planning and Management of Medical and Biological Health Risks” of the Federal Medical and Biological Agency, 10/1 Pogodinskaya St., 119121 Moscow, Russia; (K.F.); (A.K.)
| | - Siroj Bakoev
- Federal State Budgetary Institution, “Center for Strategic Planning and Management of Medical and Biological Health Risks” of the Federal Medical and Biological Agency, 10/1 Pogodinskaya St., 119121 Moscow, Russia; (K.F.); (A.K.)
| |
Collapse
|
9
|
Cheng Y, Qu Z, Jiang Q, Xu T, Zheng H, Ye P, He M, Tong Y, Ma Y, Bao A. Functional Materials for Subcellular Targeting Strategies in Cancer Therapy: Progress and Prospects. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2023:e2305095. [PMID: 37665594 DOI: 10.1002/adma.202305095] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/29/2023] [Revised: 07/26/2023] [Indexed: 09/05/2023]
Abstract
Neoadjuvant and adjuvant therapies have made significant progress in cancer treatment. However, tumor adjuvant therapy still faces challenges due to the intrinsic heterogeneity of cancer, genomic instability, and the formation of an immunosuppressive tumor microenvironment. Functional materials possess unique biological properties such as long circulation times, tumor-specific targeting, and immunomodulation. The combination of functional materials with natural substances and nanotechnology has led to the development of smart biomaterials with multiple functions, high biocompatibilities, and negligible immunogenicities, which can be used for precise cancer treatment. Recently, subcellular structure-targeting functional materials have received particular attention in various biomedical applications including the diagnosis, sensing, and imaging of tumors and drug delivery. Subcellular organelle-targeting materials can precisely accumulate therapeutic agents in organelles, considerably reduce the threshold dosages of therapeutic agents, and minimize drug-related side effects. This review provides a systematic and comprehensive overview of the research progress in subcellular organelle-targeted cancer therapy based on functional nanomaterials. Moreover, it explains the challenges and prospects of subcellular organelle-targeting functional materials in precision oncology. The review will serve as an excellent cutting-edge guide for researchers in the field of subcellular organelle-targeted cancer therapy.
Collapse
Affiliation(s)
- Yanxiang Cheng
- Department of Gynecology, Renmin Hospital, Wuhan University, No.238 Jiefang Road, Wuchang, Wuhan, 430060, P. R. China
| | - Zhen Qu
- Department of Blood Transfusion Research, Wuhan Blood Center (WHBC), HUST-WHBC United Hematology Optical Imaging Center, No.8 Baofeng 1st Road, Wuhan, Hubei, 430030, P. R. China
| | - Qian Jiang
- Department of Blood Transfusion Research, Wuhan Blood Center (WHBC), HUST-WHBC United Hematology Optical Imaging Center, No.8 Baofeng 1st Road, Wuhan, Hubei, 430030, P. R. China
| | - Tingting Xu
- Department of Clinical Laboratory, Wuhan Blood Center (WHBC), No.8 Baofeng 1st Road, Wuhan, Hubei, 430030, P. R. China
| | - Hongyun Zheng
- Department of Clinical Laboratory, Renmin Hospital, Wuhan University, No.238 Jiefang Road, Wuchang, Wuhan, 430060, P. R. China
| | - Peng Ye
- Department of Pharmacy, Renmin Hospital, Wuhan University, No.238 Jiefang Road, Wuchang, Wuhan, 430060, P. R. China
| | - Mingdi He
- Department of Blood Transfusion Research, Wuhan Blood Center (WHBC), HUST-WHBC United Hematology Optical Imaging Center, No.8 Baofeng 1st Road, Wuhan, Hubei, 430030, P. R. China
| | - Yongqing Tong
- Department of Clinical Laboratory, Renmin Hospital, Wuhan University, No.238 Jiefang Road, Wuchang, Wuhan, 430060, P. R. China
| | - Yan Ma
- Department of Blood Transfusion Research, Wuhan Blood Center (WHBC), HUST-WHBC United Hematology Optical Imaging Center, No.8 Baofeng 1st Road, Wuhan, Hubei, 430030, P. R. China
| | - Anyu Bao
- Department of Clinical Laboratory, Renmin Hospital, Wuhan University, No.238 Jiefang Road, Wuchang, Wuhan, 430060, P. R. China
| |
Collapse
|
10
|
Bhosale S, Deen MC, Proceviat C, Hettle A, Winter DK, Brockerman J, Levene M, Bennet AJ, Spino C, Boraston AB, Vocadlo DJ. A Fluorogenic Disaccharide Substrate for α-Mannosidases Enables High-Throughput Screening and Identification of an Inhibitor of the GH92 Virulence Factor from Streptococcus pneumoniae. ACS Chem Biol 2023; 18:1730-1737. [PMID: 37531094 DOI: 10.1021/acschembio.3c00053] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/03/2023]
Abstract
Trimming of host glycans is a mechanism that is broadly employed by both commensal and pathogenic microflora to enable colonization. Host glycan trimming by the opportunistic Gram-positive bacterium Streptococcus pneumoniae has been demonstrated to be an important mechanism of virulence. While S. pneumoniae employs a multitude of glycan processing enzymes, the exo-mannosidase SpGH92 has been shown to be an important virulence factor. Accordingly, SpGH92 is hypothesized to be a target for much-needed new treatments of S. pneumoniae infection. Here we report the synthesis of 4-methylumbelliferyl α-d-mannopyranosyl-(1→2)-β-d-mannopyranoside (Manα1,2Manβ-4MU) as a fluorogenic disaccharide substrate and development of an assay for SpGH92 that overcomes its requirement for +1 binding site occupancy. We miniaturize our in vitro assay and apply it to a high-throughput screen of >65 000 compounds, identifying a single inhibitory chemotype, LIPS-343. We further show that Manα1,2Manβ-4MU is also a substrate of the human Golgi-localized α-mannosidase MAN1A1, suggesting that this substrate should be useful for assessing the activity of this and other mammalian α-mannosidases.
Collapse
Affiliation(s)
- Sandeep Bhosale
- Department of Chemistry, Simon Fraser University, Burnaby, British Columbia V5A 1S6, Canada
| | - Matthew C Deen
- Department of Chemistry, Simon Fraser University, Burnaby, British Columbia V5A 1S6, Canada
| | - Cameron Proceviat
- Department of Chemistry, Simon Fraser University, Burnaby, British Columbia V5A 1S6, Canada
| | - Andrew Hettle
- Department of Biochemistry & Microbiology, University of Victoria, P.O. Box 3055 STN CSC, Victoria, British Columbia V8W 3P6, Canada
| | - Dana K Winter
- Département de Chimie, Université de Sherbrooke, Sherbrooke, Québec J1K 2R1, Canada
| | - Jacob Brockerman
- Department of Molecular Biology and Biochemistry, Simon Fraser University, Burnaby, British Columbia V5A 1S6, Canada
| | - Marina Levene
- Department of Chemistry, Simon Fraser University, Burnaby, British Columbia V5A 1S6, Canada
| | - Andrew J Bennet
- Department of Chemistry, Simon Fraser University, Burnaby, British Columbia V5A 1S6, Canada
| | - Claude Spino
- Département de Chimie, Université de Sherbrooke, Sherbrooke, Québec J1K 2R1, Canada
| | - Alisdair B Boraston
- Department of Biochemistry & Microbiology, University of Victoria, P.O. Box 3055 STN CSC, Victoria, British Columbia V8W 3P6, Canada
| | - David J Vocadlo
- Department of Chemistry, Simon Fraser University, Burnaby, British Columbia V5A 1S6, Canada
- Department of Molecular Biology and Biochemistry, Simon Fraser University, Burnaby, British Columbia V5A 1S6, Canada
| |
Collapse
|
11
|
Kempska J, Oliveira-Ferrer L, Grottke A, Qi M, Alawi M, Meyer F, Borgmann K, Hamester F, Eylmann K, Rossberg M, Smit DJ, Jücker M, Laakmann E, Witzel I, Schmalfeldt B, Müller V, Legler K. Impact of AKT1 on cell invasion and radiosensitivity in a triple negative breast cancer cell line developing brain metastasis. Front Oncol 2023; 13:1129682. [PMID: 37483521 PMCID: PMC10358765 DOI: 10.3389/fonc.2023.1129682] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Accepted: 05/30/2023] [Indexed: 07/25/2023] Open
Abstract
Introduction The PI3K/AKT pathway is activated in 43-70% of breast cancer (BC)-patients and promotes the metastatic potential of BC cells by increasing cell proliferation, invasion and radioresistance. Therefore, AKT1-inhibition in combination with radiotherapy might be an effective treatment option for triple-negative breast cancer (TNBC)-patients with brain metastases. Methods The impact of AKT1-knockout (AKT1_KO) and AKT-inhibition using Ipatasertib on MDA-MB-231 BR cells was assessed using in vitro cell proliferation and migration assays. AKT1-knockout in MDA-MB-231BR cells was performed using CRISPR/Cas9. The effect of AKT1-knockout on radiosensitivity of MDA-MB-231BR cell lines was determined via colony formation assays after cell irradiation. To detect genomic variants in AKT1_KO MDA-MB-231BR cells, whole-genome sequencing (WGS) was performed. Results Pharmacological inhibition of AKT with the pan-AKT inhibitor Ipatasertib led to a significant reduction of cell viability but did not impact cell migration. Moreover, only MDA-MB-231BR cells were sensitized following Ipatasertib-treatment. Furthermore, specific AKT1-knockout in MDA-MB-231BR showed reduced cell viability in comparison to control cells, with significant effect in one of two analyzed clones. Unexpectedly, AKT1 knockout led to increased cell migration and clonogenic potential in both AKT1_KO clones. RNAseq-analysis revealed the deregulation of CTSO, CYBB, GPR68, CEBPA, ID1, ID4, METTL15, PBX1 and PTGFRN leading to the increased cell migration, higher clonogenic survival and decreased radiosensitivity as a consequence of the AKT1 knockout in MDA-MB-231BR. Discussion Collectively, our results demonstrate that Ipatasertib leads to radiosensitization and reduced cell proliferation of MDA-MB-231BR. AKT1-inhibition showed altered gene expression profile leading to modified cell migration, clonogenic survival and radioresistance in MDA-MB-231BR. We conclude, that AKT1-inhibition in combination with radiotherapy contribute to novel treatment strategies for breast cancer brain metastases.
Collapse
Affiliation(s)
- Joanna Kempska
- Department of Gynecology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | | | - Astrid Grottke
- Department of Gynecology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Minyue Qi
- Bioinformatics Core, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Malik Alawi
- Bioinformatics Core, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Felix Meyer
- Laboratory of Radiobiology & Experimental Radio Oncology, Department of Radiotherapy and Radiation Oncology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Kerstin Borgmann
- Laboratory of Radiobiology & Experimental Radio Oncology, Department of Radiotherapy and Radiation Oncology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Fabienne Hamester
- Department of Gynecology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Kathrin Eylmann
- Department of Gynecology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Maila Rossberg
- Department of Gynecology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Daniel J. Smit
- Institute of Biochemistry and Signal Transduction, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Manfred Jücker
- Institute of Biochemistry and Signal Transduction, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Elena Laakmann
- Department of Gynecology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Isabell Witzel
- Department of Gynecology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Barbara Schmalfeldt
- Department of Gynecology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Volkmar Müller
- Department of Gynecology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Karen Legler
- Department of Gynecology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| |
Collapse
|
12
|
Sohn EJ, Oh SO. P-Element-Induced Wimpy Testis Proteins and P-Element-Induced Wimpy Testis-Interacting RNAs Expression in Ovarian Cancer Stem Cells. Genet Test Mol Biomarkers 2023; 27:56-64. [PMID: 36853842 DOI: 10.1089/gtmb.2022.0113] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/01/2023] Open
Abstract
Background: P-element-induced wimpy testis (PIWI)-interacting RNAs (piRNAs) are a type of noncoding RNA and are predominantly expressed in germline cells. piRNAs function as gene regulators and potential biomarkers for the development of a number of malignancies. The biological importance of piRNAs in ovarian cancer is still unknown. In this study, we investigated the expression of piRNAs in ovarian cancer stem cells and compared it with that in adherent cells. Methods: To assess changes in the expression levels of PIWIL1/HIWI, PIWIL2/HILI, PIWIL3, and PIWIL4/HIWI2, we used quantitative reverse-transcription polymerase chain reaction (RT-qPCR) analysis. Changes in piRNA expression levels in ovarian cancer stem cells were analyzed using Arraystar piRNA microarray screening. Gene Ontology (GO) enrichment analysis was conducted to determine the potential functions of piRNAs. Results: Using microarray analysis, we identified a cohort of differentially expressed piRNAs. Fifteen piRNAs, including DQ570763 and DQ597396, were downregulated, and 58 piRNAs were upregulated when compared with those in adherent A2780 and SKOV3 cells (p > 0.05, >2.0, respectively). GO functions of the downregulated piRNAs (DQ570763 and DQ570797) suggest that their roles are commonly associated with the Golgi apparatus. In addition, A2780-SP and SKOV3-SP cells had higher PIWIL3 and PIWIL4 mRNA levels than adherent cells (A2780 and SKOV3). Moreover, we determined, using receiver operating characteristic plot, that the expression level of PIWIL4 was lower in responders than in nonresponders after treatment with platins in patients with ovarian cancer. Finally, in ovarian cancer, PIWIL4 expression was associated with somatic mutations of dynein axonemal heavy chain 2, signal induced proliferation associated 1 like 2, YTH N6-methyladenosine RNA-binding protein 1, TBC1 domain family member 8, and LPS responsive Beige-like anchor protein. Conclusion: Our study showed that PIWI proteins and piRNAs are potential diagnostic and prognostic biomarkers for ovarian cancer.
Collapse
Affiliation(s)
- Eun Jung Sohn
- College of Medicine, Pusan National University, Yangsan, Korea
| | - Sae-Ock Oh
- Department of Anatomy, School of Medicine, Pusan National University, Yangsan, Korea
| |
Collapse
|
13
|
Huang A, Kurhade SE, Ross P, Apley KD, Griffin JD, Berkland CJ, Farrell MP. Disrupting N-Glycosylation Using Type I Mannosidase Inhibitors Alters B-Cell Receptor Signaling. ACS Pharmacol Transl Sci 2022; 5:1062-1069. [PMID: 36407961 PMCID: PMC9667535 DOI: 10.1021/acsptsci.2c00153] [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: 07/29/2022] [Indexed: 11/29/2022]
Abstract
Kifunensine is a known inhibitor of type I α-mannosidase enzymes and has been shown to have therapeutic potential for a variety of diseases and application in the expression of high-mannose N-glycan bearing glycoproteins; however, the compound's hydrophilic nature limits its efficacy. We previously synthesized two hydrophobic acylated derivatives of kifunensine, namely, JDW-II-004 and JDW-II-010, and found that these compounds were over 75-fold more potent than kifunensine. Here we explored the effects of these compounds on different mice and human B cells, and we demonstrate that they affected the cells in a similar fashion to kifunensine, further demonstrating their functional equivalence to kifunensine in assays utilizing primary cells. Specifically, a dose-dependent increase in the formation of high-mannose N-glycans decorated glycoproteins were observed upon treatment with kifunensine, JDW-II-004, and JDW-II-010, but greater potency was observed with the acylated derivatives. Treatment with kifunensine or the acylated derivatives also resulted in impaired B-cell receptor (BCR) signaling of the primary mouse B cells; however, primary human B cells treated with kifunensine or JDW-II-004 did not affect BCR signaling, while a modest increase in BCR signaling was observed upon treatment with JDW-010. Nevertheless, these findings demonstrate that the hydrophobic acylated derivatives of kifunensine can help overcome the mass-transfer limitations of the parent compound, and they may have applications for the treatment of ERAD-related diseases or prove to be more cost-effective alternatives for the generation and production of high-mannose N-glycan bearing glycoproteins.
Collapse
Affiliation(s)
- Aric Huang
- Department
of Pharmaceutical Chemistry, The University
of Kansas, Lawrence, Kansas 66047, United States
| | - Suresh E. Kurhade
- Department
of Medicinal Chemistry, The University of
Kansas, Lawrence, Kansas 66047, United
States
| | - Patrick Ross
- Department
of Medicinal Chemistry, The University of
Kansas, Lawrence, Kansas 66047, United
States
| | - Kyle D. Apley
- Department
of Pharmaceutical Chemistry, The University
of Kansas, Lawrence, Kansas 66047, United States
| | | | - Cory J. Berkland
- Department
of Pharmaceutical Chemistry, The University
of Kansas, Lawrence, Kansas 66047, United States
- Bioengineering
Program, The University of Kansas, Lawrence, Kansas 66045, United States
- Department
of Chemical and Petroleum Engineering, University
of Kansas, Lawrence, Kansas 66045, United
States
| | - Mark P. Farrell
- Department
of Medicinal Chemistry, The University of
Kansas, Lawrence, Kansas 66047, United
States
| |
Collapse
|
14
|
Identification of MAN1B1 as a Novel Marker for Bladder Cancer and Its Relationship with Immune Cell Infiltration. JOURNAL OF ONCOLOGY 2022; 2022:3387671. [PMID: 36016584 PMCID: PMC9398811 DOI: 10.1155/2022/3387671] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/13/2022] [Revised: 07/10/2022] [Accepted: 07/12/2022] [Indexed: 11/25/2022]
Abstract
Bladder cancer (BC) is a common malignant tumor of the genitourinary system, and there are not enough tumor biomarker tests that are specific, trustworthy, and noninvasive for the diagnosis and prognosis. The purpose of this study is to investigate the clinical relevance, prognostic value, and immunological signature of Mannosidase alpha class 1B member 1(MAN1B1) expressions in BC. The Cancer Genome Atlas (TCGA) and Genotype-Tissue Expression (GTEx) databases provided the raw information that was used to analyze the expression of MAN1B1 in tumor patients. Then, a statistical study was carried out to assess the correlations of MAN1B1 expression with the clinical characteristics and the prognosis of BC. The correlation between MAN1B1 expression and tumor immune infiltration was explored via single-sample gene set enrichment analysis (ssGSEA). In human cancers, MAN1B1 expressions were shown to be generally higher in tumors than in normal specimens. We confirmed that MAN1B1 expression was distinctly increased in BC specimens compared with nontumor specimens. BC specimens with advanced T stage and M stage showed a higher level of MAN1B1. Survival analysis revealed that the overall survival (OS), disease-specific survival (DSS), and progression-free interval (PFI) of patients with high MAN1B1 expressions were distinctly worse than those with low MAN1B1 expressions. Importantly, multivariate analyses only confirmed that MAN1B1 expression was an independent prognostic factor for OS of the patients with BC. Furthermore, we observed that MAN1B1 expression level was significantly correlated with abundance of multiple immune infiltrates including Th2 cells, macrophages, Th1 cells, neutrophils, T helper cells, and NK CD56 bright cells. In conjunction with all of these findings, elevated MAN1B1 expression is associated with a poor prognosis and a higher number of immune cells in BC. MAN1B1 has the potential to act as a biomarker that can evaluate both the patient's prognosis and the degree of immune infiltration in BC.
Collapse
|
15
|
Cheng H, Jin S, Huang S, Hu T, Zhao M, Li D, Wu B. Serum Proteomic Analysis by Tandem Mass Tag-Based Quantitative Proteomics in Pediatric Obstructive Sleep Apnea. Front Mol Biosci 2022; 9:762336. [PMID: 35480887 PMCID: PMC9035643 DOI: 10.3389/fmolb.2022.762336] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2021] [Accepted: 03/04/2022] [Indexed: 12/02/2022] Open
Abstract
Pediatric obstructive sleep apnea (OSA) is a frequent respiratory disorder with an estimated prevalence of 3–6% in the general population. However, the underlying pathophysiology of OSA remains unclear. Recently, proteomic analysis using high-resolution and high-throughput mass spectrometry has been widely used in the field of medical sciences. In the present study, tandem mass tag (TMT)-based proteomic analysis was performed in the serum of patients with OSA. The proteomic analysis revealed a set of differentially expressed proteins that may be associated with the pathophysiology of OSA. The differentially expressed proteins in patients with OSA were enriched in pathways including phagosome and glycan synthesis/degradation, immune response, and the hedgehog signaling pathway, indicating that such functions are key targets of OSA. Moreover, the experimental validation studies revealed that four proteins including ANTXR1, COLEC10, NCAM1, and VNN1 were reduced in the serum from patients with moderate and severe OSA, while MAN1A1 and CSPG4 protein levels were elevated in the serum from patients with severe OSA. The protein levels of ANTXR1, COLEC10, NCAM1, and VNN1 were inversely correlated with apnea-hypopnea index (AHI) in the recruited subjects, while the protein level of MAN1A1 was positively correlated with AHI, and no significant correlation was detected between CSPG4 protein and AHI. In summary, the present study for the first time identified differentially expressed proteins in the serum from OSA patients with different severities by using TMT-based proteomic analysis. The functional enrichment studies suggested that several signaling pathways may be associated with the pathophysiology of OSA. The experimental validation results indicated that six proteins including ANTXR1, COLEC10, NCAM1, VNN1, CGPG4, and MAN1A1 may play important roles in the pathophysiology of OSA, which requires further mechanistic investigation.
Collapse
Affiliation(s)
- Hanrong Cheng
- Institute of Respiratory Diseases, Shenzhen People’s Hospital, The Second Clinical Medical College of Jinan University, The First Affiliated Hospital of Southern University of Science and Technology, Shenzhen, China
| | - Shoumei Jin
- Longgang ENT Hospital, Institute of ENT and Shenzhen Key Laboratory of ENT, Shenzhen, China
| | - Simin Huang
- Longgang ENT Hospital, Institute of ENT and Shenzhen Key Laboratory of ENT, Shenzhen, China
| | - Tianyong Hu
- Longgang ENT Hospital, Institute of ENT and Shenzhen Key Laboratory of ENT, Shenzhen, China
| | - Miao Zhao
- Longgang ENT Hospital, Institute of ENT and Shenzhen Key Laboratory of ENT, Shenzhen, China
| | - Dongcai Li
- Longgang ENT Hospital, Institute of ENT and Shenzhen Key Laboratory of ENT, Shenzhen, China
- *Correspondence: Dongcai Li, ; Benqing Wu,
| | - Benqing Wu
- Department of Neonatology, University of Chinese Academy of Science-Shenzhen Hospital, Shenzhen, China
- *Correspondence: Dongcai Li, ; Benqing Wu,
| |
Collapse
|
16
|
In Situ N-glycosylation Signatures of Epithelial Ovarian Cancer Tissue as Defined by MALDI Mass Spectrometry Imaging. Cancers (Basel) 2022; 14:cancers14041021. [PMID: 35205768 PMCID: PMC8870006 DOI: 10.3390/cancers14041021] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2021] [Accepted: 01/14/2022] [Indexed: 12/31/2022] Open
Abstract
The particularly high mortality of epithelial ovarian cancer (EOC) is in part linked to limited understanding of its molecular signatures. Although there are data available on in situ N-glycosylation in EOC tissue, previous studies focused primarily on neutral N-glycan species and, hence, still little is known regarding EOC tissue-specific sialylation. In this proof-of-concept study, we implemented MALDI mass spectrometry imaging (MALDI-MSI) in combination with sialic acid derivatization to simultaneously investigate neutral and sialylated N-glycans in formalin-fixed paraffin-embedded tissue microarray specimens of less common EOC histotypes and non-malignant borderline ovarian tumor (BOT). The applied protocol allowed detecting over 50 m/z species, many of which showed differential tissue distribution. Most importantly, it could be demonstrated that α2,6- and α2,3-sialylated N-glycans are enriched in tissue regions corresponding to tumor and adjacent tumor-stroma, respectively. Interestingly, analogous N-glycosylation patterns were observed in tissue cores of BOT, suggesting that regio-specific N-glycan distribution might occur already in non-malignant ovarian pathologies. All in all, our data provide proof that the combination of MALDI-MSI and sialic acid derivatization is suitable for delineating regio-specific N-glycan distribution in EOC and BOT tissues and might serve as a promising strategy for future glycosylation-based biomarker discovery studies.
Collapse
|
17
|
Rosa-Fernandes L, Oba-Shinjo SM, Macedo-da-Silva J, Marie SKN, Palmisano G. Aberrant Protein Glycosylation in Brain Cancers, with Emphasis on Glioblastoma. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2022; 1382:39-70. [DOI: 10.1007/978-3-031-05460-0_4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/09/2022]
|
18
|
Yang Y, Sanders AJ, Dou QP, Jiang DG, Li AX, Jiang WG. The Clinical and Theranostic Values of Activated Leukocyte Cell Adhesion Molecule (ALCAM)/CD166 in Human Solid Cancers. Cancers (Basel) 2021; 13:cancers13205187. [PMID: 34680335 PMCID: PMC8533996 DOI: 10.3390/cancers13205187] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2021] [Revised: 10/13/2021] [Accepted: 10/14/2021] [Indexed: 02/08/2023] Open
Abstract
Simple Summary ALCAM (activated leukocyte cell adhesion molecule) is an important regulator in human cancers, particularly solid tumours. Its expression in cancer tissues has prognostic values depending on cancer types and is also linked to distant metastases. A truncated form, soluble form of ALCAM (sALCAM) in circulation has been suggested to be a prognostic indicator and a potential therapeutic tool. This article summarises recent findings and progress in ALCAM and its involvement in cancer, with a primary focus on its clinical connections and therapeutic values. Abstract Activated leukocyte cell adhesion molecule (ALCAM), also known as CD166, is a cell adhesion protein that is found in multiple cell types. ALCAM has multiple and diverse roles in various physiological and pathological conditions, including inflammation and cancer. There has been compelling evidence of ALCAM’s prognostic value in solid cancers, indicating that it is a potential therapeutic target. The present article overviews the recent findings and progress in ALCAM and its involvement in cancer, with a primary focus on its clinical connections in cancer and therapeutic values.
Collapse
Affiliation(s)
- Yiming Yang
- School of Medicine, Cardiff University, Henry Wellcome Building, Cardiff CF14 4XN, UK; (Y.Y.); (Q.P.D.); (D.G.J.); (A.X.L.)
| | - Andrew J. Sanders
- School of Medicine, Cardiff University, Henry Wellcome Building, Cardiff CF14 4XN, UK; (Y.Y.); (Q.P.D.); (D.G.J.); (A.X.L.)
- Correspondence: (A.J.S.); (W.G.J.)
| | - Q. Ping Dou
- School of Medicine, Cardiff University, Henry Wellcome Building, Cardiff CF14 4XN, UK; (Y.Y.); (Q.P.D.); (D.G.J.); (A.X.L.)
- Departments of Oncology, Pharmacology and Pathology School of Medicine, Barbara Ann Karmanos Cancer Institute, Wayne State University, Detroit, MI 48201-2013, USA
| | - David G. Jiang
- School of Medicine, Cardiff University, Henry Wellcome Building, Cardiff CF14 4XN, UK; (Y.Y.); (Q.P.D.); (D.G.J.); (A.X.L.)
- Stoke Mandeville Hospital, Buckinghamshire Healthcare NHS Trust, Aylesbury HP21 8AL, UK
| | - Amber Xinyu Li
- School of Medicine, Cardiff University, Henry Wellcome Building, Cardiff CF14 4XN, UK; (Y.Y.); (Q.P.D.); (D.G.J.); (A.X.L.)
| | - Wen G. Jiang
- School of Medicine, Cardiff University, Henry Wellcome Building, Cardiff CF14 4XN, UK; (Y.Y.); (Q.P.D.); (D.G.J.); (A.X.L.)
- Correspondence: (A.J.S.); (W.G.J.)
| |
Collapse
|
19
|
Chatterjee S, Ugonotti J, Lee LY, Everest-Dass A, Kawahara R, Thaysen-Andersen M. Trends in oligomannosylation and α1,2-mannosidase expression in human cancers. Oncotarget 2021; 12:2188-2205. [PMID: 34676051 PMCID: PMC8522845 DOI: 10.18632/oncotarget.28064] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2021] [Accepted: 08/18/2021] [Indexed: 02/05/2023] Open
Abstract
Aberrant protein glycosylation is a prominent cancer feature. While many tumour-associated glycoepitopes have been reported, advances in glycoanalytics continue to uncover new associations between glycosylation and cancer. Guided by a comprehensive literature survey suggesting that oligomannosylation (Man5–9 GlcNAc2) is a widespread and often regulated glycosignature in human cancers, we here revisit a valuable compilation of nearly 500 porous graphitized carbon LC-MS/MS N-glycomics datasets acquired across 11 human cancer types to systematically test for oligomannose-cancer associations. Firstly, the quantitative glycomics data obtained across 34 cancerous cell lines demonstrated that oligomannosylation is a pan-cancer feature spanning in a wide abundance range. In keeping with literature, our quantitative glycomics data of tumour and matching control tissues and new MALDI-MS imaging data of tissue microarrays showed a strong cancer-associated elevation of oligomannosylation in both basal cell (p = 1.78 × 10–12) and squamous cell (p = 1.23 × 10–11) skin cancer and colorectal cancer (p = 8.0 × 10–4). The glycomics data also indicated that some cancer types including gastric and liver cancer exhibit unchanged or reduced oligomannose levels, observations also supported by literature and MALDI-MS imaging data. Finally, expression data from public cancer repositories indicated that several α1,2-mannosidases are regulated in tumour tissues suggesting that these glycan-processing enzymes may contribute to the cancer-associated modulation of oligomannosylation. This omics-centric study has compiled robust glycomics and enzyme expression data revealing interesting molecular trends that open avenues to better understand the role of oligomannosylation in human cancers.
Collapse
Affiliation(s)
| | - Julian Ugonotti
- Department of Molecular Sciences, Macquarie University, Sydney, Australia
| | - Ling Y Lee
- Department of Molecular Sciences, Macquarie University, Sydney, Australia
| | | | - Rebeca Kawahara
- Department of Molecular Sciences, Macquarie University, Sydney, Australia.,Joint senior authors
| | - Morten Thaysen-Andersen
- Department of Molecular Sciences, Macquarie University, Sydney, Australia.,Biomolecular Discovery Research Centre (BDRC), Macquarie University, Sydney, Australia.,Joint senior authors
| |
Collapse
|
20
|
Ferragut F, Vachetta VS, Troncoso MF, Rabinovich GA, Elola MT. ALCAM/CD166: A pleiotropic mediator of cell adhesion, stemness and cancer progression. Cytokine Growth Factor Rev 2021; 61:27-37. [PMID: 34272152 DOI: 10.1016/j.cytogfr.2021.07.001] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2021] [Accepted: 07/05/2021] [Indexed: 12/14/2022]
Abstract
Activated Leukocyte Cell Adhesion Molecule (ALCAM/CD166) is a glycoprotein involved in homotypic and heterotypic cell adhesion. ALCAM can be proteolytically cleaved at the cell surface by metalloproteases, which generate shedding of its ectodomain. In various tumors, ALCAM is overexpressed and serves as a valuable prognostic marker of disease progression. Moreover, CD166 has been identified as a putative cancer stem cell marker in particular cancers. Herein, we summarize biochemical aspects of ALCAM, including structure, proteolytic shedding, alternative splicing, and specific ligands, and integrate this information with biological functions of this glycoprotein including cell adhesion, migration and invasion. In addition, we discuss different patterns of ALCAM expression in distinct tumor types and its contribution to tumor progression. Finally, we highlight the role of ALCAM as a cancer stem cell marker and introduce current clinical trials associated with this molecule. Future studies are needed to define the value of shed ALCAM in biofluids or ALCAM isoform expression as prognostic biomarkers in tumor progression.
Collapse
Affiliation(s)
- Fátima Ferragut
- Universidad de Buenos Aires, Facultad de Farmacia y Bioquímica, Departamento de Química Biológica, Buenos Aires, Argentina; CONICET-Universidad de Buenos Aires, Instituto de Química y Fisicoquímica Biológicas (IQUIFIB) Prof. Alejandro C. Paladini, Buenos Aires, Argentina
| | - Vanina S Vachetta
- Universidad de Buenos Aires, Facultad de Farmacia y Bioquímica, Departamento de Química Biológica, Buenos Aires, Argentina; CONICET-Universidad de Buenos Aires, Instituto de Química y Fisicoquímica Biológicas (IQUIFIB) Prof. Alejandro C. Paladini, Buenos Aires, Argentina
| | - María F Troncoso
- Universidad de Buenos Aires, Facultad de Farmacia y Bioquímica, Departamento de Química Biológica, Buenos Aires, Argentina; CONICET-Universidad de Buenos Aires, Instituto de Química y Fisicoquímica Biológicas (IQUIFIB) Prof. Alejandro C. Paladini, Buenos Aires, Argentina
| | - Gabriel A Rabinovich
- Laboratorio de Inmunopatología, Instituto de Biología y Medicina Experimental (IBYME-CONICET), Buenos Aires, Argentina; Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - María T Elola
- Universidad de Buenos Aires, Facultad de Farmacia y Bioquímica, Departamento de Química Biológica, Buenos Aires, Argentina; CONICET-Universidad de Buenos Aires, Instituto de Química y Fisicoquímica Biológicas (IQUIFIB) Prof. Alejandro C. Paladini, Buenos Aires, Argentina.
| |
Collapse
|
21
|
Nagakawa S, Shiota M, Fujimoto N, Yamamoto Y, Blas L, Tsukahara S, Matsumoto T, Kashiwagi E, Takeuchi A, Inokuchi J, Uchiumi T, Matsuyama H, Eto M. The impact of single-nucleotide polymorphisms on intravesical recurrence after bacillus Calmette-Guérin therapy for non-muscle invasive bladder cancer in a genome-wide association study. Urol Oncol 2021; 39:733.e17-733.e24. [PMID: 34215507 DOI: 10.1016/j.urolonc.2021.05.034] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2021] [Revised: 05/25/2021] [Accepted: 05/27/2021] [Indexed: 11/15/2022]
Abstract
OBJECTIVE Bacillus Calmette-Guérin (BCG) instillation therapy is widely used to reduce intravesical recurrence in non-muscle invasive bladder cancer (NMIBC). In this study, we aimed to reveal the genetic variations associated with intravesical recurrence after BCG therapy for NMIBC in a genome-wide association study (GWAS). MATERIALS AND METHODS This study included Japanese patients with NMIBC, in whom genomic DNA was obtained from whole blood samples. The association between genetic variation and treatment failure was analyzed by GWAS in 44 patients treated with BCG instillation as a discovery cohort. Candidate single-nucleotide polymorphisms (SNPs) were examined separately in 47 patients treated with BCG instillation and in 62 patients treated with chemotherapeutic agent instillation as validation studies. RESULTS Among the 44 patients in the discovery cohort, 14 cases experienced intravesical recurrent diseases. GWAS identified 12 candidate SNPs (rs9374832, rs35176001, rs363765, rs2127120, rs4277759, rs73664140, rs1607282, rs12141654, rs4541358, rs6986852, rs12373386, and rs17637903). In the validation study, a genetic risk stratification model using the number of risk alleles in rs363765 and rs6986852 discriminated the risk of intravesical recurrence after BCG therapy, but not after non-BCG therapy. CONCLUSION This study suggested that several SNPs were associated with intravesical recurrence after BCG therapy for NMIBC. A genetic risk model may be useful to predict intravesical recurrence after BCG therapy, warranting further research and development for clinical application.
Collapse
Affiliation(s)
- Shohei Nagakawa
- Department of Urology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Masaki Shiota
- Department of Urology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan.
| | - Naohiro Fujimoto
- Department of Urology, School of Medicine, University of Occupational and Environmental Health, Kitakyushu, Japan
| | - Yoshiaki Yamamoto
- Department of Urology, Graduate School of Medicine, Yamaguchi University, Ube, Japan
| | - Leandro Blas
- Department of Urology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Shigehiro Tsukahara
- Department of Urology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan; Department of Clinical Chemistry and Laboratory Medicine, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Takashi Matsumoto
- Department of Urology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Eiji Kashiwagi
- Department of Urology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Ario Takeuchi
- Department of Urology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Junichi Inokuchi
- Department of Urology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Takeshi Uchiumi
- Department of Clinical Chemistry and Laboratory Medicine, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Hideyasu Matsuyama
- Department of Urology, Graduate School of Medicine, Yamaguchi University, Ube, Japan
| | - Masatoshi Eto
- Department of Urology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| |
Collapse
|
22
|
Kurhade SE, Weiner JD, Gao FP, Farrell MP. Functionalized High Mannose-Specific Lectins for the Discovery of Type I Mannosidase Inhibitors. Angew Chem Int Ed Engl 2021; 60:12313-12318. [PMID: 33728787 PMCID: PMC8131250 DOI: 10.1002/anie.202101249] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2021] [Revised: 02/27/2021] [Indexed: 01/01/2023]
Abstract
An engineered cyanovirin-N homologue that exhibits specificity for high mannose N-glycans has been constructed to aid type I α 1,2-mannosidase inhibitor discovery and development. Engineering the lectins C-terminus permitted facile functionalization with fluorophores via a sortase and click strategy. The resulting lectin constructs exhibit specificity for cells presenting high mannose N-glycans. Importantly, these lectin constructs can also be applied to specifically assess changes in cell surface glycosylation induced by type I mannosidase inhibitors. Testing the utility of these lectin constructs led to the discovery of type I mannosidase inhibitors with nanomolar potency. Cumulatively, these findings reveal the specificity and utility of the functionalized cyanovirin-N homologue constructs, and highlight their potential in analytical contexts that require high mannose-specific lectins.
Collapse
Affiliation(s)
- Suresh E Kurhade
- Department of Medicinal Chemistry, The University of Kansas, 2034 Becker Drive, Lawrence, KS, 66047, USA
| | - Jack D Weiner
- Department of Medicinal Chemistry, The University of Kansas, 2034 Becker Drive, Lawrence, KS, 66047, USA
| | - Fei Philip Gao
- Protein Production Group, The University of Kansas, 2034 Becker Drive, Lawrence, KS, 66047, USA
| | - Mark P Farrell
- Department of Medicinal Chemistry, The University of Kansas, 2034 Becker Drive, Lawrence, KS, 66047, USA
| |
Collapse
|
23
|
Kurhade SE, Weiner JD, Gao FP, Farrell MP. Functionalized High Mannose‐Specific Lectins for the Discovery of Type I Mannosidase Inhibitors. Angew Chem Int Ed Engl 2021. [DOI: 10.1002/ange.202101249] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Affiliation(s)
- Suresh E. Kurhade
- Department of Medicinal Chemistry The University of Kansas 2034 Becker Drive Lawrence KS 66047 USA
| | - Jack D. Weiner
- Department of Medicinal Chemistry The University of Kansas 2034 Becker Drive Lawrence KS 66047 USA
| | - Fei Philip Gao
- Protein Production Group The University of Kansas 2034 Becker Drive Lawrence KS 66047 USA
| | - Mark P. Farrell
- Department of Medicinal Chemistry The University of Kansas 2034 Becker Drive Lawrence KS 66047 USA
| |
Collapse
|
24
|
Zhang GZ, Wu ZL, Li CY, Ren EH, Yuan WH, Deng YJ, Xie QQ. Development of a Machine Learning-Based Autophagy-Related lncRNA Signature to Improve Prognosis Prediction in Osteosarcoma Patients. Front Mol Biosci 2021; 8:615084. [PMID: 34095215 PMCID: PMC8176230 DOI: 10.3389/fmolb.2021.615084] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2020] [Accepted: 03/29/2021] [Indexed: 12/15/2022] Open
Abstract
Background Osteosarcoma is a frequent bone malignancy in children and young adults. Despite the availability of some prognostic biomarkers, most of them fail to accurately predict prognosis in osteosarcoma patients. In this study, we used bioinformatics tools and machine learning algorithms to establish an autophagy-related long non-coding RNA (lncRNA) signature to predict the prognosis of osteosarcoma patients. Methods We obtained expression and clinical data from osteosarcoma patients in the Therapeutically Applicable Research to Generate Effective Treatments (TARGET) and Gene Expression Omnibus (GEO) databases. We acquired an autophagy gene list from the Human Autophagy Database (HADb) and identified autophagy-related lncRNAs by co-expression analyses. Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analyses of the autophagy-related lncRNAs were conducted. Univariate and multivariate Cox regression analyses were performed to assess the prognostic value of the autophagy-related lncRNA signature and validate the relationship between the signature and osteosarcoma patient survival in an independent cohort. We also investigated the relationship between the signature and immune cell infiltration. Results We initially identified 69 autophagy-related lncRNAs, 13 of which were significant predictors of overall survival in osteosarcoma patients. Kaplan-Meier analyses revealed that the 13 autophagy-related lncRNAs could stratify patients based on their outcomes. Receiver operating characteristic curve analyses confirmed the superior prognostic value of the lncRNA signature compared to clinically used prognostic biomarkers. Importantly, the autophagy-related lncRNA signature predicted patient prognosis independently of clinicopathological characteristics. Furthermore, we found that the expression levels of the autophagy-related lncRNA signature were significantly associated with the infiltration levels of different immune cell subsets, including T cells, NK cells, and dendritic cells. Conclusion The autophagy-related lncRNA signature established here is an independent and robust predictor of osteosarcoma patient survival. Our findings also suggest that the expression of these 13 autophagy-related lncRNAs may promote osteosarcoma progression by regulating immune cell infiltration in the tumor microenvironment.
Collapse
Affiliation(s)
- Guang-Zhi Zhang
- The Second Clinical Medical College, Lanzhou University, Lanzhou, China.,Department of Orthopedics, Lanzhou University Second Hospital, Lanzhou, China.,Lintao County Traditional Chinese Medicine Hospital of Gansu Province, Lintao, China
| | - Zuo-Long Wu
- The Second Clinical Medical College, Lanzhou University, Lanzhou, China.,Department of Orthopedics, Lanzhou University Second Hospital, Lanzhou, China
| | - Chun-Ying Li
- The Fourth People's Hospital of Qinghai Province, Xining, China
| | - En-Hui Ren
- The Second Clinical Medical College, Lanzhou University, Lanzhou, China.,Department of Orthopedics, Lanzhou University Second Hospital, Lanzhou, China.,Department of Orthopaedics, Xining First People's Hospital, Xining, China
| | - Wen-Hua Yuan
- The Second Clinical Medical College, Lanzhou University, Lanzhou, China.,Department of Orthopedics, Lanzhou University Second Hospital, Lanzhou, China
| | - Ya-Jun Deng
- The Second Clinical Medical College, Lanzhou University, Lanzhou, China.,Department of Orthopedics, Lanzhou University Second Hospital, Lanzhou, China
| | - Qi-Qi Xie
- Affiliated Hospital of Qinghai University, Xining, China.,Affiliated Cancer Hospital of Qinghai University, Xining, China.,Breast Disease Diagnosis and Treatment Center, Affiliated Hospital of Qinghai University & Affiliated Cancer Hospital of Qinghai University, Xining, China
| |
Collapse
|
25
|
Suehara Y, Kohsaka S, Hayashi T, Akaike K, Kurisaki-Arakawa A, Sato S, Kobayashi E, Mizuno S, Ueno T, Morii T, Okuma T, Kurihara T, Hasegawa N, Sano K, Sasa K, Okubo T, Kim Y, Mano H, Saito T. Identification of a Novel MAN1A1-ROS1 Fusion Gene Through mRNA-based Screening for Tyrosine Kinase Gene Aberrations in a Patient with Leiomyosarcoma. Clin Orthop Relat Res 2021; 479:838-852. [PMID: 33196586 PMCID: PMC8083907 DOI: 10.1097/corr.0000000000001548] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/30/2020] [Accepted: 10/06/2020] [Indexed: 02/06/2023]
Abstract
BACKGROUND Soft tissue sarcomas are a heterogeneous group of rare malignant tumors. Advanced soft tissue sarcomas have a poor prognosis, and effective systemic therapies have not been established. Tyrosine kinases are increasingly being used as therapeutic targets for a variety of cancers and soft tissue sarcomas. Although complex karyotype sarcomas typically tend to carry more potentially actionable genetic alterations than do translocation-associated sarcomas (fusion gene sarcomas), based on our database review, we found that leiomyosarcoma and malignant peripheral nerve sheath tumors have lower frequencies of potential targets than other nontranslocation soft tissue sarcomas. We theorized that both leiomyosarcoma and malignant peripheral nerve sheath tumors might be included in any unique translocations. Furthermore, if tyrosine kinase imbalances, especially fusion genes, occur in patients with leiomyosarcomas and malignant peripheral nerve sheath tumors, tyrosine kinase inhibitors might be a drug development target for this sarcoma. In this study, we used a tyrosine kinase screening system that could detect an imbalance in mRNA between 5'- and 3'-sides in tyrosine kinase genes to identify potential novel therapeutic tyrosine kinase targets for soft tissue sarcomas. QUESTIONS/PURPOSES (1) Are there novel therapeutic tyrosine kinase targets in tumors from patients with soft tissue sarcomas that are detectable using mRNA screening focusing on imbalance expressions between the 5' and 3' end of the kinase domain? (2) Can potential targets be verified by RNA sequencing and reverse transcription PCR (RT-PCR)? (3) Will potential fusion gene(s) transform cells in in vitro assays? (4) Will tumors in mice that have an identified fusion gene respond to treatment with a therapeutic drug directed at that target? METHODS We used mRNA screening to look for novel tyrosine kinase targets that might be of therapeutic potential. Using functional assays, we verified whether the identified fusion genes would be good therapeutic candidates for soft tissue sarcomas. Additionally, using in vivo assays, we assessed whether suppressing the fusion's kinase activity has therapeutic potential. Study eligibility was based on a patient having high-grade spindle cell and nontranslocation sarcomas, including leiomyosarcoma, malignant peripheral nerve sheath tumor, and high-grade myxofibrosarcoma. Between 2015 and 2019, of the 172 patients with soft tissue sarcomas treated with surgical resection at Juntendo University Hospital, 72 patients had high-grade nontranslocation sarcomas. The analysis was primarily for leiomyosarcoma and malignant peripheral nerve sheath tumors, and there was a limitation of analysis size (reagent limitations) totaling 24 samples at the start of the study. We collected additional samples from a sample bank at the Tokyo Medical and Dental University to increase the number of sarcomas to study. Therefore, in this study, a total of 15 leiomyosarcoma samples, five malignant peripheral nerve sheath tumors samples, and four high-grade myxofibrosarcoma samples were collected to achieve the sample size of 24 patients. To identify tyrosine kinase fusion genes, we designed a NanoString-based assay (NanoString Technologies Inc, Seattle, WA, USA) to query the expression balances regarding transcripts of 90 tyrosine kinases at two points: the 5' end of the kinase domain and within the kinase domain or 3' end of the kinase domain. The tumor's RNA was hybridized to the NanoString probes and analyzed for the expression ratios of outliers from the 3' to 5' end of the kinase domain. Presumed novel fusion events in these positive tumors that were defined by NanoString-based assays were confirmed tyrosine kinase fusion genes by RNA sequencing and confirmatory RT-PCR. Functional analyses consisting of in vitro and in vivo assays were also performed to elucidate whether the identified tyrosine kinase gene fusions were associated with oncogenic abilities and drug responses. RESULTS We identified aberrant expression ratios regarding the 3' to 5' end of the kinase domain ratios in ROS1 transcripts in a leiomyosarcoma in a 90-year-old woman. A novel MAN1A1-ROS1 fusion gene was identified from her thigh tumor through RNA sequencing, which was confirmed with real-time PCR. In functional assays, MAN1A1-ROS1 rearrangement revealed strong transforming potential in 3T3 cells. Moreover, in an in vivo assay, crizotinib, a ROS1 inhibitor, markedly inhibited the growth of MAN1A1-ROS1 rearrangement-induced transformed cells in a dose-dependent manner. CONCLUSION We conducted tyrosine kinase screening to identify new therapeutic targets in soft tissue sarcomas. We found a novel MAN1A1-ROS1 fusion gene that may be a therapeutic target in patients with leiomyosarcoma. This study demonstrates that the mRNA screening system may aid in the development of useful therapeutic options for soft tissue sarcomas. CLINICAL RELEVANCE If novel tyrosine fusions such as MAN1A1-ROS1 fusion can be found in sarcomas from other patients, they could offer avenues for new molecular target therapies for sarcomas that currently do not have effective chemotherapeutic options. Therefore, the establishment of a screening system that includes both genomic and transcript analyses in the clinical setting is needed to verify our discoveries and take the developmental process of treatment to the next step.
Collapse
Affiliation(s)
- Yoshiyuki Suehara
- Y. Suehara, K. Akaike, T. Kurihara, N. Hasegawa, K. Sano, K. Sasa, T. Okubo, Y. Kim, Department of Orthopedic Surgery, Juntendo University School of Medicine, Tokyo, Japan
- S. Kohsaka, S. Mizuno, T. Ueno, N. Hasegawa, H. Mano, Division of Cellular Signaling, National Cancer Center Research Institute, Tokyo, Japan
- T. Hayashi, A. Kurisaki-Arakawa, T. Saito, Department of Human Pathology, Juntendo University School of Medicine, Tokyo, Japan
- S. Sato, Center for Innovative Cancer Treatment, Tokyo Medical and Dental University Hospital, Tokyo, Japan
- E. Kobayashi, Division of Musculoskeletal Oncology, National Cancer Center Hospital, Tokyo, Japan
- T. Morii, Department of Orthopedic Surgery, Kyorin University, Faculty of Medicine, Tokyo, Japan
- T. Okuma, Department of Musculoskeletal Oncology, Tokyo Metropolitan Cancer and Infectious Disease Center Komagome Hospital, Tokyo, Japan
| | - Shinji Kohsaka
- Y. Suehara, K. Akaike, T. Kurihara, N. Hasegawa, K. Sano, K. Sasa, T. Okubo, Y. Kim, Department of Orthopedic Surgery, Juntendo University School of Medicine, Tokyo, Japan
- S. Kohsaka, S. Mizuno, T. Ueno, N. Hasegawa, H. Mano, Division of Cellular Signaling, National Cancer Center Research Institute, Tokyo, Japan
- T. Hayashi, A. Kurisaki-Arakawa, T. Saito, Department of Human Pathology, Juntendo University School of Medicine, Tokyo, Japan
- S. Sato, Center for Innovative Cancer Treatment, Tokyo Medical and Dental University Hospital, Tokyo, Japan
- E. Kobayashi, Division of Musculoskeletal Oncology, National Cancer Center Hospital, Tokyo, Japan
- T. Morii, Department of Orthopedic Surgery, Kyorin University, Faculty of Medicine, Tokyo, Japan
- T. Okuma, Department of Musculoskeletal Oncology, Tokyo Metropolitan Cancer and Infectious Disease Center Komagome Hospital, Tokyo, Japan
| | - Takuo Hayashi
- Y. Suehara, K. Akaike, T. Kurihara, N. Hasegawa, K. Sano, K. Sasa, T. Okubo, Y. Kim, Department of Orthopedic Surgery, Juntendo University School of Medicine, Tokyo, Japan
- S. Kohsaka, S. Mizuno, T. Ueno, N. Hasegawa, H. Mano, Division of Cellular Signaling, National Cancer Center Research Institute, Tokyo, Japan
- T. Hayashi, A. Kurisaki-Arakawa, T. Saito, Department of Human Pathology, Juntendo University School of Medicine, Tokyo, Japan
- S. Sato, Center for Innovative Cancer Treatment, Tokyo Medical and Dental University Hospital, Tokyo, Japan
- E. Kobayashi, Division of Musculoskeletal Oncology, National Cancer Center Hospital, Tokyo, Japan
- T. Morii, Department of Orthopedic Surgery, Kyorin University, Faculty of Medicine, Tokyo, Japan
- T. Okuma, Department of Musculoskeletal Oncology, Tokyo Metropolitan Cancer and Infectious Disease Center Komagome Hospital, Tokyo, Japan
| | - Keisuke Akaike
- Y. Suehara, K. Akaike, T. Kurihara, N. Hasegawa, K. Sano, K. Sasa, T. Okubo, Y. Kim, Department of Orthopedic Surgery, Juntendo University School of Medicine, Tokyo, Japan
- S. Kohsaka, S. Mizuno, T. Ueno, N. Hasegawa, H. Mano, Division of Cellular Signaling, National Cancer Center Research Institute, Tokyo, Japan
- T. Hayashi, A. Kurisaki-Arakawa, T. Saito, Department of Human Pathology, Juntendo University School of Medicine, Tokyo, Japan
- S. Sato, Center for Innovative Cancer Treatment, Tokyo Medical and Dental University Hospital, Tokyo, Japan
- E. Kobayashi, Division of Musculoskeletal Oncology, National Cancer Center Hospital, Tokyo, Japan
- T. Morii, Department of Orthopedic Surgery, Kyorin University, Faculty of Medicine, Tokyo, Japan
- T. Okuma, Department of Musculoskeletal Oncology, Tokyo Metropolitan Cancer and Infectious Disease Center Komagome Hospital, Tokyo, Japan
| | - Aiko Kurisaki-Arakawa
- Y. Suehara, K. Akaike, T. Kurihara, N. Hasegawa, K. Sano, K. Sasa, T. Okubo, Y. Kim, Department of Orthopedic Surgery, Juntendo University School of Medicine, Tokyo, Japan
- S. Kohsaka, S. Mizuno, T. Ueno, N. Hasegawa, H. Mano, Division of Cellular Signaling, National Cancer Center Research Institute, Tokyo, Japan
- T. Hayashi, A. Kurisaki-Arakawa, T. Saito, Department of Human Pathology, Juntendo University School of Medicine, Tokyo, Japan
- S. Sato, Center for Innovative Cancer Treatment, Tokyo Medical and Dental University Hospital, Tokyo, Japan
- E. Kobayashi, Division of Musculoskeletal Oncology, National Cancer Center Hospital, Tokyo, Japan
- T. Morii, Department of Orthopedic Surgery, Kyorin University, Faculty of Medicine, Tokyo, Japan
- T. Okuma, Department of Musculoskeletal Oncology, Tokyo Metropolitan Cancer and Infectious Disease Center Komagome Hospital, Tokyo, Japan
| | - Shingo Sato
- Y. Suehara, K. Akaike, T. Kurihara, N. Hasegawa, K. Sano, K. Sasa, T. Okubo, Y. Kim, Department of Orthopedic Surgery, Juntendo University School of Medicine, Tokyo, Japan
- S. Kohsaka, S. Mizuno, T. Ueno, N. Hasegawa, H. Mano, Division of Cellular Signaling, National Cancer Center Research Institute, Tokyo, Japan
- T. Hayashi, A. Kurisaki-Arakawa, T. Saito, Department of Human Pathology, Juntendo University School of Medicine, Tokyo, Japan
- S. Sato, Center for Innovative Cancer Treatment, Tokyo Medical and Dental University Hospital, Tokyo, Japan
- E. Kobayashi, Division of Musculoskeletal Oncology, National Cancer Center Hospital, Tokyo, Japan
- T. Morii, Department of Orthopedic Surgery, Kyorin University, Faculty of Medicine, Tokyo, Japan
- T. Okuma, Department of Musculoskeletal Oncology, Tokyo Metropolitan Cancer and Infectious Disease Center Komagome Hospital, Tokyo, Japan
| | - Eisuke Kobayashi
- Y. Suehara, K. Akaike, T. Kurihara, N. Hasegawa, K. Sano, K. Sasa, T. Okubo, Y. Kim, Department of Orthopedic Surgery, Juntendo University School of Medicine, Tokyo, Japan
- S. Kohsaka, S. Mizuno, T. Ueno, N. Hasegawa, H. Mano, Division of Cellular Signaling, National Cancer Center Research Institute, Tokyo, Japan
- T. Hayashi, A. Kurisaki-Arakawa, T. Saito, Department of Human Pathology, Juntendo University School of Medicine, Tokyo, Japan
- S. Sato, Center for Innovative Cancer Treatment, Tokyo Medical and Dental University Hospital, Tokyo, Japan
- E. Kobayashi, Division of Musculoskeletal Oncology, National Cancer Center Hospital, Tokyo, Japan
- T. Morii, Department of Orthopedic Surgery, Kyorin University, Faculty of Medicine, Tokyo, Japan
- T. Okuma, Department of Musculoskeletal Oncology, Tokyo Metropolitan Cancer and Infectious Disease Center Komagome Hospital, Tokyo, Japan
| | - Sho Mizuno
- Y. Suehara, K. Akaike, T. Kurihara, N. Hasegawa, K. Sano, K. Sasa, T. Okubo, Y. Kim, Department of Orthopedic Surgery, Juntendo University School of Medicine, Tokyo, Japan
- S. Kohsaka, S. Mizuno, T. Ueno, N. Hasegawa, H. Mano, Division of Cellular Signaling, National Cancer Center Research Institute, Tokyo, Japan
- T. Hayashi, A. Kurisaki-Arakawa, T. Saito, Department of Human Pathology, Juntendo University School of Medicine, Tokyo, Japan
- S. Sato, Center for Innovative Cancer Treatment, Tokyo Medical and Dental University Hospital, Tokyo, Japan
- E. Kobayashi, Division of Musculoskeletal Oncology, National Cancer Center Hospital, Tokyo, Japan
- T. Morii, Department of Orthopedic Surgery, Kyorin University, Faculty of Medicine, Tokyo, Japan
- T. Okuma, Department of Musculoskeletal Oncology, Tokyo Metropolitan Cancer and Infectious Disease Center Komagome Hospital, Tokyo, Japan
| | - Toshihide Ueno
- Y. Suehara, K. Akaike, T. Kurihara, N. Hasegawa, K. Sano, K. Sasa, T. Okubo, Y. Kim, Department of Orthopedic Surgery, Juntendo University School of Medicine, Tokyo, Japan
- S. Kohsaka, S. Mizuno, T. Ueno, N. Hasegawa, H. Mano, Division of Cellular Signaling, National Cancer Center Research Institute, Tokyo, Japan
- T. Hayashi, A. Kurisaki-Arakawa, T. Saito, Department of Human Pathology, Juntendo University School of Medicine, Tokyo, Japan
- S. Sato, Center for Innovative Cancer Treatment, Tokyo Medical and Dental University Hospital, Tokyo, Japan
- E. Kobayashi, Division of Musculoskeletal Oncology, National Cancer Center Hospital, Tokyo, Japan
- T. Morii, Department of Orthopedic Surgery, Kyorin University, Faculty of Medicine, Tokyo, Japan
- T. Okuma, Department of Musculoskeletal Oncology, Tokyo Metropolitan Cancer and Infectious Disease Center Komagome Hospital, Tokyo, Japan
| | - Takeshi Morii
- Y. Suehara, K. Akaike, T. Kurihara, N. Hasegawa, K. Sano, K. Sasa, T. Okubo, Y. Kim, Department of Orthopedic Surgery, Juntendo University School of Medicine, Tokyo, Japan
- S. Kohsaka, S. Mizuno, T. Ueno, N. Hasegawa, H. Mano, Division of Cellular Signaling, National Cancer Center Research Institute, Tokyo, Japan
- T. Hayashi, A. Kurisaki-Arakawa, T. Saito, Department of Human Pathology, Juntendo University School of Medicine, Tokyo, Japan
- S. Sato, Center for Innovative Cancer Treatment, Tokyo Medical and Dental University Hospital, Tokyo, Japan
- E. Kobayashi, Division of Musculoskeletal Oncology, National Cancer Center Hospital, Tokyo, Japan
- T. Morii, Department of Orthopedic Surgery, Kyorin University, Faculty of Medicine, Tokyo, Japan
- T. Okuma, Department of Musculoskeletal Oncology, Tokyo Metropolitan Cancer and Infectious Disease Center Komagome Hospital, Tokyo, Japan
| | - Tomotake Okuma
- Y. Suehara, K. Akaike, T. Kurihara, N. Hasegawa, K. Sano, K. Sasa, T. Okubo, Y. Kim, Department of Orthopedic Surgery, Juntendo University School of Medicine, Tokyo, Japan
- S. Kohsaka, S. Mizuno, T. Ueno, N. Hasegawa, H. Mano, Division of Cellular Signaling, National Cancer Center Research Institute, Tokyo, Japan
- T. Hayashi, A. Kurisaki-Arakawa, T. Saito, Department of Human Pathology, Juntendo University School of Medicine, Tokyo, Japan
- S. Sato, Center for Innovative Cancer Treatment, Tokyo Medical and Dental University Hospital, Tokyo, Japan
- E. Kobayashi, Division of Musculoskeletal Oncology, National Cancer Center Hospital, Tokyo, Japan
- T. Morii, Department of Orthopedic Surgery, Kyorin University, Faculty of Medicine, Tokyo, Japan
- T. Okuma, Department of Musculoskeletal Oncology, Tokyo Metropolitan Cancer and Infectious Disease Center Komagome Hospital, Tokyo, Japan
| | - Taisei Kurihara
- Y. Suehara, K. Akaike, T. Kurihara, N. Hasegawa, K. Sano, K. Sasa, T. Okubo, Y. Kim, Department of Orthopedic Surgery, Juntendo University School of Medicine, Tokyo, Japan
- S. Kohsaka, S. Mizuno, T. Ueno, N. Hasegawa, H. Mano, Division of Cellular Signaling, National Cancer Center Research Institute, Tokyo, Japan
- T. Hayashi, A. Kurisaki-Arakawa, T. Saito, Department of Human Pathology, Juntendo University School of Medicine, Tokyo, Japan
- S. Sato, Center for Innovative Cancer Treatment, Tokyo Medical and Dental University Hospital, Tokyo, Japan
- E. Kobayashi, Division of Musculoskeletal Oncology, National Cancer Center Hospital, Tokyo, Japan
- T. Morii, Department of Orthopedic Surgery, Kyorin University, Faculty of Medicine, Tokyo, Japan
- T. Okuma, Department of Musculoskeletal Oncology, Tokyo Metropolitan Cancer and Infectious Disease Center Komagome Hospital, Tokyo, Japan
| | - Nobuhiko Hasegawa
- Y. Suehara, K. Akaike, T. Kurihara, N. Hasegawa, K. Sano, K. Sasa, T. Okubo, Y. Kim, Department of Orthopedic Surgery, Juntendo University School of Medicine, Tokyo, Japan
- S. Kohsaka, S. Mizuno, T. Ueno, N. Hasegawa, H. Mano, Division of Cellular Signaling, National Cancer Center Research Institute, Tokyo, Japan
- T. Hayashi, A. Kurisaki-Arakawa, T. Saito, Department of Human Pathology, Juntendo University School of Medicine, Tokyo, Japan
- S. Sato, Center for Innovative Cancer Treatment, Tokyo Medical and Dental University Hospital, Tokyo, Japan
- E. Kobayashi, Division of Musculoskeletal Oncology, National Cancer Center Hospital, Tokyo, Japan
- T. Morii, Department of Orthopedic Surgery, Kyorin University, Faculty of Medicine, Tokyo, Japan
- T. Okuma, Department of Musculoskeletal Oncology, Tokyo Metropolitan Cancer and Infectious Disease Center Komagome Hospital, Tokyo, Japan
| | - Kei Sano
- Y. Suehara, K. Akaike, T. Kurihara, N. Hasegawa, K. Sano, K. Sasa, T. Okubo, Y. Kim, Department of Orthopedic Surgery, Juntendo University School of Medicine, Tokyo, Japan
- S. Kohsaka, S. Mizuno, T. Ueno, N. Hasegawa, H. Mano, Division of Cellular Signaling, National Cancer Center Research Institute, Tokyo, Japan
- T. Hayashi, A. Kurisaki-Arakawa, T. Saito, Department of Human Pathology, Juntendo University School of Medicine, Tokyo, Japan
- S. Sato, Center for Innovative Cancer Treatment, Tokyo Medical and Dental University Hospital, Tokyo, Japan
- E. Kobayashi, Division of Musculoskeletal Oncology, National Cancer Center Hospital, Tokyo, Japan
- T. Morii, Department of Orthopedic Surgery, Kyorin University, Faculty of Medicine, Tokyo, Japan
- T. Okuma, Department of Musculoskeletal Oncology, Tokyo Metropolitan Cancer and Infectious Disease Center Komagome Hospital, Tokyo, Japan
| | - Keita Sasa
- Y. Suehara, K. Akaike, T. Kurihara, N. Hasegawa, K. Sano, K. Sasa, T. Okubo, Y. Kim, Department of Orthopedic Surgery, Juntendo University School of Medicine, Tokyo, Japan
- S. Kohsaka, S. Mizuno, T. Ueno, N. Hasegawa, H. Mano, Division of Cellular Signaling, National Cancer Center Research Institute, Tokyo, Japan
- T. Hayashi, A. Kurisaki-Arakawa, T. Saito, Department of Human Pathology, Juntendo University School of Medicine, Tokyo, Japan
- S. Sato, Center for Innovative Cancer Treatment, Tokyo Medical and Dental University Hospital, Tokyo, Japan
- E. Kobayashi, Division of Musculoskeletal Oncology, National Cancer Center Hospital, Tokyo, Japan
- T. Morii, Department of Orthopedic Surgery, Kyorin University, Faculty of Medicine, Tokyo, Japan
- T. Okuma, Department of Musculoskeletal Oncology, Tokyo Metropolitan Cancer and Infectious Disease Center Komagome Hospital, Tokyo, Japan
| | - Taketo Okubo
- Y. Suehara, K. Akaike, T. Kurihara, N. Hasegawa, K. Sano, K. Sasa, T. Okubo, Y. Kim, Department of Orthopedic Surgery, Juntendo University School of Medicine, Tokyo, Japan
- S. Kohsaka, S. Mizuno, T. Ueno, N. Hasegawa, H. Mano, Division of Cellular Signaling, National Cancer Center Research Institute, Tokyo, Japan
- T. Hayashi, A. Kurisaki-Arakawa, T. Saito, Department of Human Pathology, Juntendo University School of Medicine, Tokyo, Japan
- S. Sato, Center for Innovative Cancer Treatment, Tokyo Medical and Dental University Hospital, Tokyo, Japan
- E. Kobayashi, Division of Musculoskeletal Oncology, National Cancer Center Hospital, Tokyo, Japan
- T. Morii, Department of Orthopedic Surgery, Kyorin University, Faculty of Medicine, Tokyo, Japan
- T. Okuma, Department of Musculoskeletal Oncology, Tokyo Metropolitan Cancer and Infectious Disease Center Komagome Hospital, Tokyo, Japan
| | - Youngji Kim
- Y. Suehara, K. Akaike, T. Kurihara, N. Hasegawa, K. Sano, K. Sasa, T. Okubo, Y. Kim, Department of Orthopedic Surgery, Juntendo University School of Medicine, Tokyo, Japan
- S. Kohsaka, S. Mizuno, T. Ueno, N. Hasegawa, H. Mano, Division of Cellular Signaling, National Cancer Center Research Institute, Tokyo, Japan
- T. Hayashi, A. Kurisaki-Arakawa, T. Saito, Department of Human Pathology, Juntendo University School of Medicine, Tokyo, Japan
- S. Sato, Center for Innovative Cancer Treatment, Tokyo Medical and Dental University Hospital, Tokyo, Japan
- E. Kobayashi, Division of Musculoskeletal Oncology, National Cancer Center Hospital, Tokyo, Japan
- T. Morii, Department of Orthopedic Surgery, Kyorin University, Faculty of Medicine, Tokyo, Japan
- T. Okuma, Department of Musculoskeletal Oncology, Tokyo Metropolitan Cancer and Infectious Disease Center Komagome Hospital, Tokyo, Japan
| | - Hiroyuki Mano
- Y. Suehara, K. Akaike, T. Kurihara, N. Hasegawa, K. Sano, K. Sasa, T. Okubo, Y. Kim, Department of Orthopedic Surgery, Juntendo University School of Medicine, Tokyo, Japan
- S. Kohsaka, S. Mizuno, T. Ueno, N. Hasegawa, H. Mano, Division of Cellular Signaling, National Cancer Center Research Institute, Tokyo, Japan
- T. Hayashi, A. Kurisaki-Arakawa, T. Saito, Department of Human Pathology, Juntendo University School of Medicine, Tokyo, Japan
- S. Sato, Center for Innovative Cancer Treatment, Tokyo Medical and Dental University Hospital, Tokyo, Japan
- E. Kobayashi, Division of Musculoskeletal Oncology, National Cancer Center Hospital, Tokyo, Japan
- T. Morii, Department of Orthopedic Surgery, Kyorin University, Faculty of Medicine, Tokyo, Japan
- T. Okuma, Department of Musculoskeletal Oncology, Tokyo Metropolitan Cancer and Infectious Disease Center Komagome Hospital, Tokyo, Japan
| | - Tsuyoshi Saito
- Y. Suehara, K. Akaike, T. Kurihara, N. Hasegawa, K. Sano, K. Sasa, T. Okubo, Y. Kim, Department of Orthopedic Surgery, Juntendo University School of Medicine, Tokyo, Japan
- S. Kohsaka, S. Mizuno, T. Ueno, N. Hasegawa, H. Mano, Division of Cellular Signaling, National Cancer Center Research Institute, Tokyo, Japan
- T. Hayashi, A. Kurisaki-Arakawa, T. Saito, Department of Human Pathology, Juntendo University School of Medicine, Tokyo, Japan
- S. Sato, Center for Innovative Cancer Treatment, Tokyo Medical and Dental University Hospital, Tokyo, Japan
- E. Kobayashi, Division of Musculoskeletal Oncology, National Cancer Center Hospital, Tokyo, Japan
- T. Morii, Department of Orthopedic Surgery, Kyorin University, Faculty of Medicine, Tokyo, Japan
- T. Okuma, Department of Musculoskeletal Oncology, Tokyo Metropolitan Cancer and Infectious Disease Center Komagome Hospital, Tokyo, Japan
| |
Collapse
|
26
|
Progress in research into the role of abnormal glycosylation modification in tumor immunity. Immunol Lett 2020; 229:8-17. [PMID: 33186635 DOI: 10.1016/j.imlet.2020.11.003] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2020] [Revised: 10/25/2020] [Accepted: 11/07/2020] [Indexed: 12/22/2022]
Abstract
In abnormal glycosylation, molecules of glucose or other carbohydrates in living organisms are inappropriately attached to proteins, which causes protein denaturation. Abnormal glycosylation modification is known to directly or indirectly affect the tumor escape process, but very few studies have been performed on whether protein glycosylation changes the structure and function of immune cells and immune molecules and thereby regulates the occurrence and development of tumor escape. Therefore, this article summarizes the effect of the immune system on tumor escape in association with the abnormal glycosylation process from an immunological perspective.
Collapse
|
27
|
Thomas D, Rathinavel AK, Radhakrishnan P. Altered glycosylation in cancer: A promising target for biomarkers and therapeutics. Biochim Biophys Acta Rev Cancer 2020; 1875:188464. [PMID: 33157161 DOI: 10.1016/j.bbcan.2020.188464] [Citation(s) in RCA: 161] [Impact Index Per Article: 32.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2020] [Revised: 10/08/2020] [Accepted: 10/28/2020] [Indexed: 12/13/2022]
Abstract
Glycosylation is a well-regulated cell and microenvironment specific post-translational modification. Several glycosyltransferases and glycosidases orchestrate the addition of defined glycan structures on the proteins and lipids. Recent advances and systemic approaches in glycomics have significantly contributed to a better understanding of instrumental roles of glycans in health and diseases. Emerging research evidence recognized aberrantly glycosylated proteins as the modulators of the malignant phenotype of cancer cells. The Cancer Genome Atlas has identified alterations in the expressions of glycosylation-specific genes that are correlated with cancer progression. However, the mechanistic basis remains poorly explored. Recent researches have shown that specific changes in the glycan structures are associated with 'stemness' and epithelial-to-mesenchymal transition of cancer cells. Moreover, epigenetic changes in the glycosylation pattern make the tumor cells capable of escaping immunosurveillance mechanisms. The deciphering roles of glycans in cancer emphasize that glycans can serve as a source for the development of novel clinical biomarkers. The ability of glycans in intervening various stages of tumor progression and the biosynthetic pathways involved in glycan structures constitute a promising target for cancer therapy. Advances in the knowledge of innovative strategies for identifying the mechanisms of glycan-binding proteins are hoped to hold great potential in cancer therapy. This review discusses the fundamental role of glycans in regulating tumorigenesis and tumor progression and provides insights into the influence of glycans in the current tactics of targeted therapies in the clinical setting.
Collapse
Affiliation(s)
- Divya Thomas
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, NE 68198, USA; Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Ashok Kumar Rathinavel
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, NE 68198, USA; Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Prakash Radhakrishnan
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, NE 68198, USA; Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE 68198, USA; Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE 68198, USA; Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE 68198, USA; Department of Genetics, Cell Biology and Anatomy, University of Nebraska Medical Center, Omaha, NE 68198, USA.
| |
Collapse
|
28
|
A novel Golgi mannosidase inhibitor: Molecular design, synthesis, enzyme inhibition, and inhibition of spheroid formation. Bioorg Med Chem 2020; 28:115492. [PMID: 32291147 DOI: 10.1016/j.bmc.2020.115492] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2020] [Revised: 04/01/2020] [Accepted: 04/03/2020] [Indexed: 11/23/2022]
Abstract
Effective chemotherapy for solid cancers is challenging due to a limitation in permeation that prevents anticancer drugs from reaching the center of the tumor, therefore unable to limit cancer cell growth. To circumvent this issue, we planned to apply the drugs directly at the center by first collapsing the outer structure. For this, we focused on cell-cell communication (CCC) between N-glycans and proteins at the tumor cell surface. Mature N-glycans establish CCC; however, CCC is hindered when numerous immature N-glycans are present at the cell surface. Inhibition of Golgi mannosidases (GMs) results in the transport of immature N-glycans to the cell surface. This can be employed to disrupt CCC. Here, we describe the molecular design and synthesis of an improved GM inhibitor with a non-sugar mimic scaffold that was screened from a compound library. The synthesized compounds were tested for enzyme inhibition ability and inhibition of spheroid formation using cell-based methods. Most of the compounds designed and synthesized exhibited GM inhibition at the cellular level. Of those, AR524 had higher inhibitory activity than a known GM inhibitor, kifunensine. Moreover, AR524 inhibited spheroid formation of human malignant cells at low concentration (10 µM), based on the disruption of CCC by GM inhibition.
Collapse
|