101
|
Valk-Weeber RL, Nichols K, Dijkhuizen L, Bijl E, van Leeuwen SS. Variations in N-linked glycosylation of glycosylation-dependent cell adhesion molecule 1 (GlyCAM-1) whey protein: Intercow differences and dietary effects. J Dairy Sci 2021; 104:5056-5068. [PMID: 33551170 DOI: 10.3168/jds.2020-19297] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Accepted: 10/19/2020] [Indexed: 11/19/2022]
Abstract
In bovine milk serum, the whey proteins with the highest N-glycan contribution are lactoferrin, IgG, and glycosylation-dependent cellular adhesion molecule 1 (GlyCAM-1); GlyCAM-1 is the dominant N-linked glycoprotein in bovine whey protein products. Whey proteins are base ingredients in a range of food products, including infant formulas. Glycan monosaccharide composition and variation thereof may affect functionality, such as the interaction of glycans with the immune system via recognition receptors. It is therefore highly relevant to understand whether and how the glycosylation of whey proteins (and their functionality) can be modulated. We recently showed that the glycoprofile of GlyCAM-1 varies between cows and during early lactation, whereas the glycoprofile of lactoferrin was highly constant. In the current study, we evaluated intercow differences and the effects of macronutrient supply on the N-linked glycosylation profiles of the major whey proteins in milk samples of Holstein-Friesian cows. Overall, approximately 60% of the N-glycan pool in milk protein was sialylated, or fucosylated, or both; GlyCAM-1 contributed approximately 78% of the total number of glycans in the overall whey protein N-linked glycan pool. The degree of fucosylation ranged from 44.8 to 73.3% between cows, and this variation was mainly attributed to the glycans of GlyCAM-1. Dietary supplementation with fat or protein did not influence the overall milk serum glycoprofile. Postruminal infusion of palm olein, glucose, and essential AA resulted in shifts in the degree of GlyCAM-1 fucosylation within individual cows, ranging in some cases from 50 to 71% difference in degree of fucosylation, regardless of treatment. Overall, these data demonstrate that the glycosylation, and particularly fucosylation, of GlyCAM-1 was variable, although these shifts appear to be related more to individual cow variation than to nutrient supply. To our knowledge, this is the first report of variation in glycosylation of a milk glycoprotein in mature, noncolostral milk. The functional implications of variable GlyCAM-1 fucosylation remain to be investigated.
Collapse
Affiliation(s)
- Rivca L Valk-Weeber
- Microbial Physiology, Groningen Biomolecular Sciences and Biotechnology Institute (GBB), University of Groningen, Nijenborgh 7, 9747 AG Groningen, the Netherlands
| | - Kelly Nichols
- Animal Nutrition Group, Wageningen University and Research, PO Box 338, 6700 AH Wageningen, the Netherlands
| | - Lubbert Dijkhuizen
- Microbial Physiology, Groningen Biomolecular Sciences and Biotechnology Institute (GBB), University of Groningen, Nijenborgh 7, 9747 AG Groningen, the Netherlands; CarbExplore Research BV, Zernikepark 12, 9747 AN Groningen, the Netherlands
| | - Etske Bijl
- Dairy Science and Technology, Food Quality and Design Group, Wageningen University and Research, PO Box 17, 6700 AA Wageningen, the Netherlands
| | - Sander S van Leeuwen
- Microbial Physiology, Groningen Biomolecular Sciences and Biotechnology Institute (GBB), University of Groningen, Nijenborgh 7, 9747 AG Groningen, the Netherlands; Sector Human Nutrition and Health, Laboratory Medicine, University Medical Center Groningen (UMCG), Hanzeplein 1, 9713 GZ, Groningen, the Netherlands.
| |
Collapse
|
102
|
Gupta R, Kumar G, Jain BP, Chandra S, Goswami SK. Ectopic expression of 35 kDa and knocking down of 78 kDa SG2NAs induce cytoskeletal reorganization, alter membrane sialylation, and modulate the markers of EMT. Mol Cell Biochem 2021; 476:633-648. [PMID: 33083950 DOI: 10.1007/s11010-020-03932-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2020] [Accepted: 10/07/2020] [Indexed: 12/01/2022]
Abstract
SG2NA is a protein of the striatin family that organizes STRIPAK complexes. It has splice variants expressing differentially in tissues. Its 78 kDa isoform regulates cell cycle, maintains homeostasis in the endoplasmic reticulum, and prevents oxidative injuries. The 35 kDa variant is devoid of the signature WD-40 repeats in the carboxy terminal, and its function is unknown. We expressed it in NIH 3T3 cells that otherwise express 78 kDa variant only. These cells (35 EE) have altered morphology, faster rate of migration, and enhanced growth as measured by the MTT assay. Similar phenotypes were also seen in cells where the endogenous 78 kDa isoform was downregulated by siRNA (78 KD). Proteomic analyses showed that several cancer-associated proteins are modulated in both 35 EE and 78 KD cells. The 35 EE cells have diffused actin fibers, distinctive ultrastructure, reduced sialylation, and increased expression of MMP2 & 9. The 78 KD cells also had diffused actin fibers and an upregulated expression of MMP2. In both cells, markers epithelial to mesenchymal transition (EMT) viz, E- & N-cadherins, β-catenin, slug, vimentin, and ZO-1 were modulated partially in tune with the EMT process. Since NIH 3T3 cells are mesenchymal, we also expressed 35 kDa SG2NA in MCF-7 cells of epithelial origin. In these cells (MCF-7-35), the actin fibers were also diffused and the modulation of the markers was more in tune with the EMT process. However, unlike in 35 EE cells, in MCF-7-35 cells, membrane sialylation rather increased. We infer that ectopic expression of 35 kDa and downregulation of 78 kDa SG2NAs partially induce transformed phenotypes.
Collapse
Affiliation(s)
- Richa Gupta
- School of Life Sciences, Jawaharlal Nehru University, New Mehrauli Road, New Delhi, 110067, India
| | - Gaurav Kumar
- Peptide and Proteomics Division, Defense Institute of Physiology and Allied Sciences (DIPAS), DRDO, Delhi, 110054, India
| | - Buddhi Prakash Jain
- Department of Zoology, School of Life Sciences, Mahatma Gandhi Central University, Motihari, 845401, Bihar, India
| | - Sunandini Chandra
- Department of Cell Biology, Yale School of Medicine, New Haven, CT, USA
| | - Shyamal K Goswami
- School of Life Sciences, Jawaharlal Nehru University, New Mehrauli Road, New Delhi, 110067, India.
| |
Collapse
|
103
|
Satyavarapu EM, Nath S, Mandal C. Desialylation of Atg5 by sialidase (Neu2) enhances autophagosome formation to induce anchorage-dependent cell death in ovarian cancer cells. Cell Death Discov 2021; 7:26. [PMID: 33526785 PMCID: PMC7851153 DOI: 10.1038/s41420-020-00391-y] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2020] [Revised: 11/27/2020] [Accepted: 12/09/2020] [Indexed: 12/14/2022] Open
Abstract
Increased sialylation is one of the hallmarks of ovarian cancer (OC) but its relation with programmed cell death is not known. Here we explored the molecular interplay between autophagy, apoptosis/anoikis, and aberrant-expression of the PI3K-Akt/mTOR pathway in the context of sialidase. OC is accompanied by low expression of cytosolic sialidase (Neu2) and ~10-fold more α2,6- than α2,3-linked sialic acids found through qPCR, western blot, and flow cytometry. Interestingly, Neu2 overexpression cleaved α2,6- and α2,3-linked sialic acids and reduced cell viability. Several autophagy-related molecules like LC3B/Atg3/Atg5/Atg7/Atg12/Atg16L1/Beclin1 were upregulated upon Neu2 overexpression. Atg5, a crucial protein for autophagosome formation, was desialylated by overexpressed Neu2. Desialylated Atg5 now showed enhanced association both with Atg12 and Atg16L1 leading to more autophagosome formation. Neu2-overexpressing cells exhibited extrinsic pathway-mediated apoptosis as reflected the in activation of Fas/FasL/FADD/Bid/caspase 8/caspase 6/caspase 3/PARP cleavage. There was also increased Bax, reduced Bcl2, and several cell-cycle molecules (CDK2/CDK4/CDK6/cyclin-B1/cyclin-E). Inhibition of autophagy using bafilomycin A1 or Beclin1 siRNA leads to reversal of Neu2-induced apoptosis suggesting their possible relationship. Additionally, overexpressed Neu2 inhibited growth factor-mediated signaling molecules involved in the PI3K/Akt-mTOR pathway probably through their desialylation. Furthermore, overexpressed Neu2 inhibited epithelial (ZO-1/Claudin1), mesenchymal (snail/slug), and cell-adhesion (integrin-β3/focal-adhesion kinase) molecules suggesting anchorage-dependent cell death (anoikis). Such changes were absent in the presence of bafilomycin A1 indicating the involvement of autophagy in Neu2-induced anoikis. The physiological relevance of our in vitro observations was further confirmed in the OC xenograft model. Taken together, it is the first report demonstrating that Atg5 is a sialoglycoprotein having α2,6- and α2,3-linked sialic acids and its desialylation by overexpressed Neu2 leads to its activation for autophagosome formation, which induced apoptosis/anoikis in OC.
Collapse
Affiliation(s)
- Eswara Murali Satyavarapu
- Cancer Biology and Inflammatory Disorder Division, Council of Scientific and Industrial Research-Indian Institute of Chemical Biology, 4, Raja S.C. Mallick Road, Kolkata, 700032, India
| | - Shalini Nath
- Cancer Biology and Inflammatory Disorder Division, Council of Scientific and Industrial Research-Indian Institute of Chemical Biology, 4, Raja S.C. Mallick Road, Kolkata, 700032, India
| | - Chitra Mandal
- Cancer Biology and Inflammatory Disorder Division, Council of Scientific and Industrial Research-Indian Institute of Chemical Biology, 4, Raja S.C. Mallick Road, Kolkata, 700032, India.
| |
Collapse
|
104
|
Ferreira JA, Relvas-Santos M, Peixoto A, M N Silva A, Lara Santos L. Glycoproteogenomics: Setting the Course for Next-generation Cancer Neoantigen Discovery for Cancer Vaccines. GENOMICS, PROTEOMICS & BIOINFORMATICS 2021; 19:25-43. [PMID: 34118464 PMCID: PMC8498922 DOI: 10.1016/j.gpb.2021.03.005] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/11/2020] [Revised: 01/25/2021] [Accepted: 03/01/2021] [Indexed: 12/24/2022]
Abstract
Molecular-assisted precision oncology gained tremendous ground with high-throughput next-generation sequencing (NGS), supported by robust bioinformatics. The quest for genomics-based cancer medicine set the foundations for improved patient stratification, while unveiling a wide array of neoantigens for immunotherapy. Upfront pre-clinical and clinical studies have successfully used tumor-specific peptides in vaccines with minimal off-target effects. However, the low mutational burden presented by many lesions challenges the generalization of these solutions, requiring the diversification of neoantigen sources. Oncoproteogenomics utilizing customized databases for protein annotation by mass spectrometry (MS) is a powerful tool toward this end. Expanding the concept toward exploring proteoforms originated from post-translational modifications (PTMs) will be decisive to improve molecular subtyping and provide potentially targetable functional nodes with increased cancer specificity. Walking through the path of systems biology, we highlight that alterations in protein glycosylation at the cell surface not only have functional impact on cancer progression and dissemination but also originate unique molecular fingerprints for targeted therapeutics. Moreover, we discuss the outstanding challenges required to accommodate glycoproteomics in oncoproteogenomics platforms. We envisage that such rationale may flag a rather neglected research field, generating novel paradigms for precision oncology and immunotherapy.
Collapse
Affiliation(s)
- José Alexandre Ferreira
- Experimental Pathology and Therapeutics Group, Portuguese Institute of Oncology, Porto 4200-072, Portugal; Institute of Biomedical Sciences Abel Salazar, University of Porto, Porto 4050-313, Portugal; Porto Comprehensive Cancer Center (P.ccc), Porto 4200-072, Portugal.
| | - Marta Relvas-Santos
- Experimental Pathology and Therapeutics Group, Portuguese Institute of Oncology, Porto 4200-072, Portugal; Institute of Biomedical Sciences Abel Salazar, University of Porto, Porto 4050-313, Portugal; REQUIMTE-LAQV, Department of Chemistry and Biochemistry, Faculty of Sciences of the University of Porto, Porto 4169-007, Portugal
| | - Andreia Peixoto
- Experimental Pathology and Therapeutics Group, Portuguese Institute of Oncology, Porto 4200-072, Portugal; Institute of Biomedical Sciences Abel Salazar, University of Porto, Porto 4050-313, Portugal
| | - André M N Silva
- REQUIMTE-LAQV, Department of Chemistry and Biochemistry, Faculty of Sciences of the University of Porto, Porto 4169-007, Portugal
| | - Lúcio Lara Santos
- Experimental Pathology and Therapeutics Group, Portuguese Institute of Oncology, Porto 4200-072, Portugal; Institute of Biomedical Sciences Abel Salazar, University of Porto, Porto 4050-313, Portugal; Porto Comprehensive Cancer Center (P.ccc), Porto 4200-072, Portugal
| |
Collapse
|
105
|
Cai L, Meng L, Zeng J, Wan Q. Sequential activation of thioglycosides enables one-pot glycosylation. Org Chem Front 2021. [DOI: 10.1039/d0qo01414a] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
This review describes recent developments in relative reactivity value (RRV) controlled sequential glycosylation, pre-activation based iterative glycosylation, and sulfoxide activation initiated one-pot glycosylation.
Collapse
Affiliation(s)
- Lei Cai
- Hubei Key Laboratory of Natural Medicinal Chemistry and Resource Evaluation
- School of Pharmacy
- Huazhong University of Science and Technology
- Wuhan
- China
| | - Lingkui Meng
- Hubei Key Laboratory of Natural Medicinal Chemistry and Resource Evaluation
- School of Pharmacy
- Huazhong University of Science and Technology
- Wuhan
- China
| | - Jing Zeng
- Hubei Key Laboratory of Natural Medicinal Chemistry and Resource Evaluation
- School of Pharmacy
- Huazhong University of Science and Technology
- Wuhan
- China
| | - Qian Wan
- Hubei Key Laboratory of Natural Medicinal Chemistry and Resource Evaluation
- School of Pharmacy
- Huazhong University of Science and Technology
- Wuhan
- China
| |
Collapse
|
106
|
Britain CM, Bhalerao N, Silva AD, Chakraborty A, Buchsbaum DJ, Crowley MR, Crossman DK, Edwards YJK, Bellis SL. Glycosyltransferase ST6Gal-I promotes the epithelial to mesenchymal transition in pancreatic cancer cells. J Biol Chem 2021; 296:100034. [PMID: 33148698 PMCID: PMC7949065 DOI: 10.1074/jbc.ra120.014126] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Revised: 10/05/2020] [Accepted: 11/04/2020] [Indexed: 12/18/2022] Open
Abstract
ST6Gal-I, an enzyme upregulated in numerous malignancies, adds α2-6-linked sialic acids to select membrane receptors, thereby modulating receptor signaling and cell phenotype. In this study, we investigated ST6Gal-I's role in epithelial to mesenchymal transition (EMT) using the Suit2 pancreatic cancer cell line, which has low endogenous ST6Gal-I and limited metastatic potential, along with two metastatic Suit2-derived subclones, S2-013 and S2-LM7AA, which have upregulated ST6Gal-I. RNA-Seq results suggested that the metastatic subclones had greater activation of EMT-related gene networks than parental Suit2 cells, and forced overexpression of ST6Gal-I in the Suit2 line was sufficient to activate EMT pathways. Accordingly, we evaluated expression of EMT markers and cell invasiveness (a key phenotypic feature of EMT) in Suit2 cells with or without ST6Gal-I overexpression, as well as S2-013 and S2-LM7AA cells with or without ST6Gal-I knockdown. Cells with high ST6Gal-I expression displayed enrichment in mesenchymal markers (N-cadherin, slug, snail, fibronectin) and cell invasiveness, relative to ST6Gal-I-low cells. Contrarily, epithelial markers (E-cadherin, occludin) were suppressed in ST6Gal-I-high cells. To gain mechanistic insight into ST6Gal-I's role in EMT, we examined the activity of epidermal growth factor receptor (EGFR), a known EMT driver. ST6Gal-I-high cells had greater α2-6 sialylation and activation of EGFR than ST6Gal-I-low cells. The EGFR inhibitor, erlotinib, neutralized ST6Gal-I-dependent differences in EGFR activation, mesenchymal marker expression, and invasiveness in Suit2 and S2-LM7AA, but not S2-013, lines. Collectively, these results advance our understanding of ST6Gal-I's tumor-promoting function by highlighting a role for ST6Gal-I in EMT, which may be mediated, at least in part, by α2-6-sialylated EGFR.
Collapse
Affiliation(s)
- Colleen M Britain
- Department of Cell, Developmental, and Integrative Biology, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Nikita Bhalerao
- Department of Cell, Developmental, and Integrative Biology, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Austin D Silva
- Department of Cell, Developmental, and Integrative Biology, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Asmi Chakraborty
- Department of Cell, Developmental, and Integrative Biology, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Donald J Buchsbaum
- Department of Radiation Oncology, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Michael R Crowley
- Department of Genetics, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - David K Crossman
- Department of Genetics, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Yvonne J K Edwards
- Department of Cell, Developmental, and Integrative Biology, University of Alabama at Birmingham, Birmingham, Alabama, USA; Department of Biochemistry and Molecular Genetics, University of Alabama at Birmingham, Birmingham, Alabama, USA.
| | - Susan L Bellis
- Department of Cell, Developmental, and Integrative Biology, University of Alabama at Birmingham, Birmingham, Alabama, USA.
| |
Collapse
|
107
|
Skropeta D, Dobie C, Montgomery AP, Steele H, Szabo R, Yu H. Sialyltransferase Inhibitors as Potential Anti-Cancer Agents. Aust J Chem 2021. [DOI: 10.1071/ch21195] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Sialic acid occupies a privileged position at the terminus of the glycan chain of many cell-surface glycoconjugates. Owing to both their structure and location, charged sialic acid residues mediate numerous critical interactions in cell–cell communication including cell recognition, invasion, migration, receptor binding, and immunological responses. Sialyltransferases (STs) are the enzymes involved in the biosynthesis of sialylated glycans and are highly upregulated, up to 40–60 %, in a range of cancers, with tumour hypersialylation strongly correlated with both tumour progression and treatment resistance. Accordingly, inhibiting sialylation is currently being explored by several research groups worldwide as a potential new cancer treatment strategy. However, to progress small molecule ST inhibitors into the clinic, issues around selectivity, synthetic accessibility, and cell permeability need to be addressed. Using computationally guided design principles, we produced a leading series of ST inhibitors by replacing the cytidine nucleoside with uridine and substituting the charged phosphodiester linker with a carbamate or triazole moiety. Biological evaluation of the newly developed inhibitors was performed using commercially available human ST enzymes, with the Ki inhibition values of the lead compounds ranging from 1 to 20 µM. Compared with earlier generations of sialylation inhibitors, our inhibitors are non-toxic in a range of cell studies, with improved synthetic accessibility.
Collapse
|
108
|
Ene CD, Penescu MN, Georgescu SR, Tampa M, Nicolae I. Posttranslational Modifications Pattern in Clear Cell Renal Cell Carcinoma. Metabolites 2020; 11:10. [PMID: 33375435 PMCID: PMC7824589 DOI: 10.3390/metabo11010010] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2020] [Revised: 12/18/2020] [Accepted: 12/21/2020] [Indexed: 02/07/2023] Open
Abstract
Posttranslational modifications are dynamic enzymatic-mediated processes, regulated in time and space, associated with cancer development. We aimed to evaluate the significance of posttranslational modifications in the pathogenesis of clear cell renal cell carcinoma. The authors developed a prospective, observational study during a period of three years and included 55 patients with localized renal cell carcinoma and 30 heathy subjects. Glycosylation, nitration and carbonylation, thiol-disulfide homeostasis, methylation, phosphorylation and proteolytic cleavage were evaluated in the serum of the evaluated subjects in the present study. Our results showed some characteristics for early ccRCC: high production of cytokines, substrate hypersialylation, induced nitrosative and carbonylic stress, arginine hypermethylation, thiol/disulfide homeostasis (TDH) alteration, the regulatory role of soluble receptors (sRAGE, sIL-6R) in RAGE and IL-6 signaling, the modulatory effect of TK-1and TuM2-PK in controlling the level of phosphometabolites in neoplastic cells. These data could be the initial point for development of a panel of biomarkers such as total sialic acid, orosomucoids, nitrotyrosine, carbonylic metabolites, ADMA, SDMA, and thiol-disulfide equilibrium for early diagnosis of ccRCC. Moreover, they could be considered a specific disease PTM signature which underlines the transition from early to advanced stages in this neoplasia, and of a therapeutic target in kidney oncogenesis.
Collapse
Affiliation(s)
- Corina Daniela Ene
- Faculty of General Medicine, Carol Davila University of Medicine and Pharmacy, 020021 Bucharest, Romania; (M.N.P.); (S.R.G.); (M.T.)
- Carol Davila Clinical Hospital of Nephrology, 010731 Bucharest, Romania
| | - Mircea Nicolae Penescu
- Faculty of General Medicine, Carol Davila University of Medicine and Pharmacy, 020021 Bucharest, Romania; (M.N.P.); (S.R.G.); (M.T.)
- Carol Davila Clinical Hospital of Nephrology, 010731 Bucharest, Romania
| | - Simona Roxana Georgescu
- Faculty of General Medicine, Carol Davila University of Medicine and Pharmacy, 020021 Bucharest, Romania; (M.N.P.); (S.R.G.); (M.T.)
- Victor Babes Clinical Hospital of Tropical and Infectious Diseases, 030303 Bucharest, Romania;
| | - Mircea Tampa
- Faculty of General Medicine, Carol Davila University of Medicine and Pharmacy, 020021 Bucharest, Romania; (M.N.P.); (S.R.G.); (M.T.)
- Victor Babes Clinical Hospital of Tropical and Infectious Diseases, 030303 Bucharest, Romania;
| | - Ilinca Nicolae
- Victor Babes Clinical Hospital of Tropical and Infectious Diseases, 030303 Bucharest, Romania;
| |
Collapse
|
109
|
Dasgupta D, Pally D, Saini DK, Bhat R, Ghosh A. Nanomotors Sense Local Physicochemical Heterogeneities in Tumor Microenvironments*. Angew Chem Int Ed Engl 2020; 59:23690-23696. [PMID: 32918839 PMCID: PMC7756332 DOI: 10.1002/anie.202008681] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2020] [Revised: 08/16/2020] [Indexed: 12/11/2022]
Abstract
The invasion of cancer is brought about by continuous interaction of malignant cells with their surrounding tissue microenvironment. Investigating the remodeling of local extracellular matrix (ECM) by invading cells can thus provide fundamental insights into the dynamics of cancer progression. In this paper, we use an active untethered nanomechanical tool, realized as magnetically driven nanomotors, to locally probe a 3D tissue culture environment. We observed that nanomotors preferentially adhere to the cancer-proximal ECM and magnitude of the adhesive force increased with cell lines of higher metastatic ability. We experimentally confirmed that sialic acid linkage specific to cancer-secreted ECM makes it differently charged, which causes this adhesion. In an assay consisting of both cancerous and non-cancerous epithelia, that mimics the in vivo histopathological milieu of a malignant breast tumor, we find that nanomotors preferentially decorate the region around the cancer cells.
Collapse
Affiliation(s)
- Debayan Dasgupta
- Centre for Nano Science and EngineeringIndian Institute of ScienceBangalore560012India
| | - Dharma Pally
- Department of Molecular Reproduction, Development and GeneticsIndian Institute of ScienceBangalore560012India
| | - Deepak K. Saini
- Department of Molecular Reproduction, Development and GeneticsIndian Institute of ScienceBangalore560012India
- Centre for Biosystems Science and Engineering, IIScBangalore560012India
| | - Ramray Bhat
- Department of Molecular Reproduction, Development and GeneticsIndian Institute of ScienceBangalore560012India
| | - Ambarish Ghosh
- Centre for Nano Science and EngineeringIndian Institute of ScienceBangalore560012India
- Department of PhysicsIndian Institute of ScienceBangalore560012India
| |
Collapse
|
110
|
Klaus C, Liao H, Allendorf DH, Brown GC, Neumann H. Sialylation acts as a checkpoint for innate immune responses in the central nervous system. Glia 2020; 69:1619-1636. [PMID: 33340149 DOI: 10.1002/glia.23945] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Revised: 11/23/2020] [Accepted: 11/26/2020] [Indexed: 12/28/2022]
Abstract
Sialic acids are monosaccharides that normally terminate the glycan chains of cell surface glyco-proteins and -lipids in mammals, and are highly enriched in the central nervous tissue. Sialic acids are conjugated to proteins and lipids (termed "sialylation") by specific sialyltransferases, and are removed ("desialylation") by neuraminidases. Cell surface sialic acids are sensed by complement factor H (FH) to inhibit complement activation or by sialic acid-binding immunoglobulin-like lectin (SIGLEC) receptors to inhibit microglial activation, phagocytosis, and oxidative burst. In contrast, desialylation of cells enables binding of the opsonins C1, calreticulin, galectin-3, and collectins, stimulating phagocytosis of such cells. Hypersialylation is used by bacteria and cancers as camouflage to escape immune recognition, while polysialylation of neurons protects synapses and neurogenesis. Insufficient lysosomal cleavage of sialylated molecules can lead to lysosomal accumulation of lipids and aggregated proteins, which if excessive may be expelled into the extracellular space. On the other hand, desialylation of immune receptors can activate them or trigger removal of proteins. Loss of inhibitory SIGLECs or FH triggers reduced clearance of aggregates, oxidative brain damage and complement-mediated retinal damage. Thus, cell surface sialylation recognized by FH, SIGLEC, and other immune-related receptors acts as a major checkpoint inhibitor of innate immune responses in the central nervous system, while excessive cleavage of sialic acid residues and consequently removing this checkpoint inhibitor may trigger lipid accumulation, protein aggregation, inflammation, and neurodegeneration.
Collapse
Affiliation(s)
- Christine Klaus
- Institute of Reconstructive Neurobiology, Medical Faculty and University Hospital of Bonn, University of Bonn, Bonn, Germany
| | - Huan Liao
- Institute of Reconstructive Neurobiology, Medical Faculty and University Hospital of Bonn, University of Bonn, Bonn, Germany
| | | | - Guy C Brown
- Department of Biochemistry, University of Cambridge, Cambridge, UK
| | - Harald Neumann
- Institute of Reconstructive Neurobiology, Medical Faculty and University Hospital of Bonn, University of Bonn, Bonn, Germany
| |
Collapse
|
111
|
Current Status on Therapeutic Molecules Targeting Siglec Receptors. Cells 2020; 9:cells9122691. [PMID: 33333862 PMCID: PMC7765293 DOI: 10.3390/cells9122691] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2020] [Revised: 12/09/2020] [Accepted: 12/12/2020] [Indexed: 12/15/2022] Open
Abstract
The sialic acid-binding immunoglobulin-type of lectins (Siglecs) are receptors that recognize sialic acid-containing glycans. In the majority of the cases, Siglecs are expressed on immune cells and play a critical role in regulating immune cell signaling. Over the years, it has been shown that the sialic acid-Siglec axis participates in immunological homeostasis, and that any imbalance can trigger different pathologies, such as autoimmune diseases or cancer. For all this, different therapeutics have been developed that bind to Siglecs, either based on antibodies or being smaller molecules. In this review, we briefly introduce the Siglec family and we compile a description of glycan-based molecules and antibody-based therapies (including CAR-T and bispecific antibodies) that have been designed to therapeutically targeting Siglecs.
Collapse
|
112
|
Dorsett KA, Marciel MP, Hwang J, Ankenbauer KE, Bhalerao N, Bellis SL. Regulation of ST6GAL1 sialyltransferase expression in cancer cells. Glycobiology 2020; 31:530-539. [PMID: 33320246 DOI: 10.1093/glycob/cwaa110] [Citation(s) in RCA: 52] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2020] [Revised: 11/07/2020] [Accepted: 11/25/2020] [Indexed: 02/06/2023] Open
Abstract
The ST6GAL1 sialyltransferase, which adds α2-6 linked sialic acids to N-glycosylated proteins, is overexpressed in a wide range of human malignancies. Recent studies have established the importance of ST6GAL1 in promoting tumor cell behaviors such as invasion, resistance to cell stress and chemoresistance. Furthermore, ST6GAL1 activity has been implicated in imparting cancer stem cell characteristics. However, despite the burgeoning interest in the role of ST6GAL1 in the phenotypic features of tumor cells, insufficient attention has been paid to the molecular mechanisms responsible for ST6GAL1 upregulation during neoplastic transformation. Evidence suggests that these mechanisms are multifactorial, encompassing genetic, epigenetic, transcriptional and posttranslational regulation. The purpose of this review is to summarize current knowledge regarding the molecular events that drive enriched ST6GAL1 expression in cancer cells.
Collapse
Affiliation(s)
- Kaitlyn A Dorsett
- Department of Cell, Developmental and Integrative Biology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Michael P Marciel
- Department of Cell, Developmental and Integrative Biology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Jihye Hwang
- Department of Cell, Developmental and Integrative Biology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Katherine E Ankenbauer
- Department of Cell, Developmental and Integrative Biology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Nikita Bhalerao
- Department of Cell, Developmental and Integrative Biology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Susan L Bellis
- Department of Cell, Developmental and Integrative Biology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| |
Collapse
|
113
|
Yu X, Shi H, Li Y, Guo Y, Zhang P, Wang G, Li L, Chen X, Ding L, Ju H. Thermally Triggered, Cell-Specific Enzymatic Glyco-Editing: In Situ Regulation of Lectin Recognition and Immune Response on Target Cells. ACS APPLIED MATERIALS & INTERFACES 2020; 12:54387-54398. [PMID: 33236873 DOI: 10.1021/acsami.0c15212] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
In situ glyco-editing on the cell surface can endow cellular glycoforms with new structures and properties; however, the lack of cell specificity and dependence on cells' endogenous functions plague the revelation of cellular glycan recognition properties and hamper the application of glyco-editing in complicated authentic biosystems. Herein, we develop a thermally triggered, cell-specific glyco-editing method for regulation of lectin recognition on target live cells in both single- and cocultured settings. The method relies on the aptamer-mediated anchoring of microgel-encapsulated neuraminidase on target cells and subsequent thermally triggered enzyme release for localized sialic acid (Sia) trimming. This temperature-based enzyme accessibility modulation strategy exempts genetic or metabolic engineering operations and, thus for the first time, enables tumor-specific desialylation on complicated tissue slices. The proposed method also provides an unprecedented opportunity to potentiate the innate immune response of natural killer cells toward target tumor cells through thermally triggered cell-specific desialylation, which paves the way for in vivo glycoimmune-checkpoint-targeted cancer therapeutic intervention.
Collapse
Affiliation(s)
- Xiaofei Yu
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing 210023, China
| | - Huifang Shi
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing 210023, China
| | - Yiran Li
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing 210023, China
| | - Yuna Guo
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing 210023, China
| | - Peiwen Zhang
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing 210023, China
| | - Guyu Wang
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing 210023, China
| | - Lei Li
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing 210023, China
| | - Xian Chen
- Jiangsu Province Blood Center, Nanjing 210008, China
| | - Lin Ding
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing 210023, China
- Chemistry and Biomedicine Innovation Center (ChemBIC), Nanjing University, Nanjing 210023, China
| | - Huangxian Ju
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing 210023, China
| |
Collapse
|
114
|
Läubli H, Kawanishi K, George Vazhappilly C, Matar R, Merheb M, Sarwar Siddiqui S. Tools to study and target the Siglec-sialic acid axis in cancer. FEBS J 2020; 288:6206-6225. [PMID: 33251699 DOI: 10.1111/febs.15647] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2020] [Revised: 11/18/2020] [Accepted: 11/23/2020] [Indexed: 12/16/2022]
Abstract
Siglecs are widely expressed on leucocytes and bind to ubiquitously presented glycans containing sialic acids (sialoglycans). Most Siglecs carry an immunoreceptor tyrosine-based inhibition motif (ITIM) and elicit an inhibitory intracellular signal upon ligand binding. A few Siglec receptors can, however, recruit immunoreceptor tyrosine-based activation motif (ITAM)-containing factors, which activate cells. The role of hypersialylation (the enhanced expression of sialoglycans) has recently been explored in cancer progression. Mechanistic studies have shown that hypersialylation on cancer cells can engage inhibitory Siglecs on the surface of immune cells and induce immunosuppression. These recent studies strongly suggest that the Siglec-sialic acid axis can act as a potential target for cancer immunotherapy. Moreover, the use of new tools and techniques is facilitating these studies. In this review, we summarise techniques used to study Siglecs, including different mouse models, monoclonal antibodies, Siglec fusion proteins, and sialoglycan arrays. Furthermore, we discuss the recent major developments in the study of Siglecs in cancer immunosuppression, tools, and techniques used in targeting the Siglec-sialic acid axis and the possibility of clinical intervention.
Collapse
Affiliation(s)
- Heinz Läubli
- Laboratory for Cancer Immunotherapy, Department of Biomedicine, and Medical Oncology, Department of Internal Medicine, University Hospital Basel, Switzerland
| | - Kunio Kawanishi
- Kidney and Vascular Pathology, University of Tsukuba, Ibaraki, Japan
| | | | - Rachel Matar
- Department of Biotechnology, American University of Ras Al Khaimah (AURAK), UAE
| | - Maxime Merheb
- Department of Biotechnology, American University of Ras Al Khaimah (AURAK), UAE
| | | |
Collapse
|
115
|
Dobie C, Skropeta D. Insights into the role of sialylation in cancer progression and metastasis. Br J Cancer 2020; 124:76-90. [PMID: 33144696 PMCID: PMC7782833 DOI: 10.1038/s41416-020-01126-7] [Citation(s) in RCA: 179] [Impact Index Per Article: 35.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2020] [Revised: 09/11/2020] [Accepted: 10/08/2020] [Indexed: 02/07/2023] Open
Abstract
Upregulation of sialyltransferases—the enzymes responsible for the addition of sialic acid to growing glycoconjugate chains—and the resultant hypersialylation of up to 40–60% of tumour cell surfaces are established hallmarks of several cancers, including lung, breast, ovarian, pancreatic and prostate cancer. Hypersialylation promotes tumour metastasis by several routes, including enhancing immune evasion and tumour cell survival, and stimulating tumour invasion and migration. The critical role of enzymes that regulate sialic acid in tumour cell growth and metastasis points towards targeting sialylation as a potential new anti-metastatic cancer treatment strategy. Herein, we explore insights into the mechanisms by which hypersialylation plays a role in promoting metastasis, and explore the current state of sialyltransferase inhibitor development.
Collapse
Affiliation(s)
- Christopher Dobie
- School of Chemistry & Molecular Bioscience, Faculty of Science, Medicine & Health; and Molecular Horizons, University of Wollongong, NSW, 2522, Wollongong, Australia
| | - Danielle Skropeta
- School of Chemistry & Molecular Bioscience, Faculty of Science, Medicine & Health; and Molecular Horizons, University of Wollongong, NSW, 2522, Wollongong, Australia. .,Illawarra Health & Medical Research Institute, Wollongong, NSW, 2522, Australia.
| |
Collapse
|
116
|
Unliganded and CMP-Neu5Ac bound structures of human α-2,6-sialyltransferase ST6Gal I at high resolution. J Struct Biol 2020; 212:107628. [DOI: 10.1016/j.jsb.2020.107628] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2020] [Revised: 08/31/2020] [Accepted: 09/16/2020] [Indexed: 01/12/2023]
|
117
|
Dasgupta D, Pally D, Saini DK, Bhat R, Ghosh A. Nanomotors Sense Local Physicochemical Heterogeneities in Tumor Microenvironments**. Angew Chem Int Ed Engl 2020. [DOI: 10.1002/ange.202008681] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Affiliation(s)
- Debayan Dasgupta
- Centre for Nano Science and Engineering Indian Institute of Science Bangalore 560012 India
| | - Dharma Pally
- Department of Molecular Reproduction, Development and Genetics Indian Institute of Science Bangalore 560012 India
| | - Deepak K. Saini
- Department of Molecular Reproduction, Development and Genetics Indian Institute of Science Bangalore 560012 India
- Centre for Biosystems Science and Engineering, IISc Bangalore 560012 India
| | - Ramray Bhat
- Department of Molecular Reproduction, Development and Genetics Indian Institute of Science Bangalore 560012 India
| | - Ambarish Ghosh
- Centre for Nano Science and Engineering Indian Institute of Science Bangalore 560012 India
- Department of Physics Indian Institute of Science Bangalore 560012 India
| |
Collapse
|
118
|
A versatile soluble siglec scaffold for sensitive and quantitative detection of glycan ligands. Nat Commun 2020; 11:5091. [PMID: 33037195 PMCID: PMC7547722 DOI: 10.1038/s41467-020-18907-6] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2020] [Accepted: 09/21/2020] [Indexed: 12/15/2022] Open
Abstract
Sialic acid-binding immunoglobulin-type lectins (Siglecs) are immunomodulatory receptors that are regulated by their glycan ligands. The connections between Siglecs and human disease motivate improved methods to detect Siglec ligands. Here, we describe a new versatile set of Siglec-Fc proteins for glycan ligand detection. Enhanced sensitivity and selectivity are enabled through multimerization and avoiding Fc receptors, respectively. Using these Siglec-Fc proteins, Siglec ligands are systematically profiled on healthy and cancerous cells and tissues, revealing many unique patterns. Additional features enable the production of small, homogenous Siglec fragments and development of a quantitative ligand-binding mass spectrometry assay. Using this assay, the ligand specificities of several Siglecs are clarified. For CD33 (Siglec-3), we demonstrate that it recognizes both α2-3 and α2-6 sialosides in solution and on cells, which has implications for its link to Alzheimer’s disease susceptibility. These soluble Siglecs reveal the abundance of their glycan ligands on host cells as self-associated molecular patterns. Sialic acid-binding immunoglobulin-type lectins (Siglecs) are a family of immunomodulatory receptors expressed on cells of the hematopoietic lineage. Here the authors demonstrate an approach for the identification of the glycan ligands of Siglecs, which is also applicable to other families of glycan-binding proteins.
Collapse
|
119
|
Rodrigues Mantuano N, Natoli M, Zippelius A, Läubli H. Tumor-associated carbohydrates and immunomodulatory lectins as targets for cancer immunotherapy. J Immunother Cancer 2020; 8:jitc-2020-001222. [PMID: 33020245 PMCID: PMC7537339 DOI: 10.1136/jitc-2020-001222] [Citation(s) in RCA: 55] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/28/2020] [Indexed: 12/17/2022] Open
Abstract
During oncogenesis, tumor cells present specific carbohydrate chains that are new targets for cancer immunotherapy. Whereas these tumor-associated carbohydrates (TACA) can be targeted with antibodies and vaccination approaches, TACA including sialic acid-containing glycans are able to inhibit anticancer immune responses by engagement of immune receptors on leukocytes. A family of immune-modulating receptors are sialic acid-binding Siglec receptors that have been recently described to inhibit antitumor activity mediated by myeloid cells, natural killer cells and T cells. Other TACA-binding receptors including selectins have been linked to cancer progression. Recent studies have shown that glycan-lectin interactions can be targeted to improve cancer immunotherapy. For example, interactions between the immune checkpoint T cell immunoglobulin and mucin-domain containing-3 and the lectin galectin-9 are targeted in clinical trials. In addition, an antibody against the lectin Siglec-15 is being tested in an early clinical trial. In this review, we summarize the previous and current efforts to target TACA and to inhibit inhibitory immune receptors binding to TACA including the Siglec-sialoglycan axis.
Collapse
Affiliation(s)
| | - Marina Natoli
- Department of Biomedicine, Universitätsspital Basel, Basel, Switzerland
| | - Alfred Zippelius
- Department of Biomedicine, Universitätsspital Basel, Basel, Switzerland
| | - Heinz Läubli
- Department of Biomedicine, Universitätsspital Basel, Basel, Switzerland
| |
Collapse
|
120
|
Campesato LF, Weng CH, Merghoub T. Innate immune checkpoints for cancer immunotherapy: expanding the scope of non T cell targets. ANNALS OF TRANSLATIONAL MEDICINE 2020; 8:1031. [PMID: 32953831 PMCID: PMC7475486 DOI: 10.21037/atm-20-1816] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Affiliation(s)
- Luis F Campesato
- Swim Across America and Ludwig Collaborative Laboratory, Immunology Program, Parker Institute for Cancer Immunotherapy, Memorial Sloan Kettering Cancer Center, New York, NY, USA.,Immuno-Oncology Service, Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Chien-Huan Weng
- Swim Across America and Ludwig Collaborative Laboratory, Immunology Program, Parker Institute for Cancer Immunotherapy, Memorial Sloan Kettering Cancer Center, New York, NY, USA.,Immuno-Oncology Service, Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Taha Merghoub
- Swim Across America and Ludwig Collaborative Laboratory, Immunology Program, Parker Institute for Cancer Immunotherapy, Memorial Sloan Kettering Cancer Center, New York, NY, USA.,Immuno-Oncology Service, Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| |
Collapse
|
121
|
Adenovirus Receptor Expression in Cancer and Its Multifaceted Role in Oncolytic Adenovirus Therapy. Int J Mol Sci 2020; 21:ijms21186828. [PMID: 32957644 PMCID: PMC7554712 DOI: 10.3390/ijms21186828] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2020] [Revised: 09/14/2020] [Accepted: 09/15/2020] [Indexed: 02/06/2023] Open
Abstract
Oncolytic adenovirus therapy is believed to be a promising way to treat cancer patients. To be able to target tumor cells with an oncolytic adenovirus, expression of the adenovirus receptor on the tumor cell is essential. Different adenovirus types bind to different receptors on the cell, of which the expression can vary between tumor types. Pre-existing neutralizing immunity to human adenovirus species C type 5 (HAdV-C5) has hampered its therapeutic efficacy in clinical trials, hence several adenoviral vectors from different species are currently being developed as a means to evade pre-existing immunity. Therefore, knowledge on the expression of appropriate adenovirus receptors on tumor cells is important. This could aid in determining which tumor types would benefit most from treatment with a certain oncolytic adenovirus type. This review provides an overview of the known receptors for human adenoviruses and how their expression on tumor cells might be differentially regulated compared to healthy tissue, before and after standardized anticancer treatments. Mechanisms behind the up- or downregulation of adenovirus receptor expression are discussed, which could be used to find new targets for combination therapy to enhance the efficacy of oncolytic adenovirus therapy. Additionally, the utility of the adenovirus receptors in oncolytic virotherapy is examined, including their role in viral spread, which might even surpass their function as primary entry receptors. Finally, future directions are offered regarding the selection of adenovirus types to be used in oncolytic adenovirus therapy in the fight against cancer.
Collapse
|
122
|
Macpherson AM, Barry SC, Ricciardelli C, Oehler MK. Epithelial Ovarian Cancer and the Immune System: Biology, Interactions, Challenges and Potential Advances for Immunotherapy. J Clin Med 2020; 9:E2967. [PMID: 32937961 PMCID: PMC7564553 DOI: 10.3390/jcm9092967] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2020] [Revised: 08/28/2020] [Accepted: 09/03/2020] [Indexed: 12/11/2022] Open
Abstract
Recent advances in the understanding of immune function and the interactions with tumour cells have led to the development of various cancer immunotherapies and strategies for specific cancer types. However, despite some stunning successes with some malignancies such as melanomas and lung cancer, most patients receive little or no benefit from immunotherapy, which has been attributed to the tumour microenvironment and immune evasion. Although the US Food and Drug Administration have approved immunotherapies for some cancers, to date, only the anti-angiogenic antibody bevacizumab is approved for the treatment of epithelial ovarian cancer. Immunotherapeutic strategies for ovarian cancer are still under development and being tested in numerous clinical trials. A detailed understanding of the interactions between cancer and the immune system is vital for optimisation of immunotherapies either alone or when combined with chemotherapy and other therapies. This article, in two main parts, provides an overview of: (1) components of the normal immune system and current knowledge regarding tumour immunology, biology and their interactions; (2) strategies, and targets, together with challenges and potential innovative approaches for cancer immunotherapy, with attention given to epithelial ovarian cancer.
Collapse
Affiliation(s)
- Anne M. Macpherson
- Discipline of Obstetrics and Gynaecology, Adelaide Medical School, Robinson Research Institute, University of Adelaide, Adelaide 5000, Australia; (A.M.M.); (C.R.)
| | - Simon C. Barry
- Molecular Immunology, Robinson Research Institute, University of Adelaide, Adelaide 5005, Australia;
| | - Carmela Ricciardelli
- Discipline of Obstetrics and Gynaecology, Adelaide Medical School, Robinson Research Institute, University of Adelaide, Adelaide 5000, Australia; (A.M.M.); (C.R.)
| | - Martin K. Oehler
- Discipline of Obstetrics and Gynaecology, Adelaide Medical School, Robinson Research Institute, University of Adelaide, Adelaide 5000, Australia; (A.M.M.); (C.R.)
- Department of Gynaecological Oncology, Royal Adelaide Hospital, Adelaide 5000, Australia
| |
Collapse
|
123
|
Movsisyan LD, Macauley MS. Structural advances of Siglecs: insight into synthetic glycan ligands for immunomodulation. Org Biomol Chem 2020; 18:5784-5797. [PMID: 32756649 DOI: 10.1039/d0ob01116a] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Sialic acid-binding immunoglobulin-like lectins (Siglecs) are transmembrane proteins of the immunoglobulin (Ig) superfamily predominantly expressed on the cells of our immune system. Siglecs recognize sialic acid via their terminal V-set domain. In mammals, sialic acid-terminated glycolipids and glycoproteins are the ligands of Siglecs, and the monomeric affinity of Siglecs for their sialic acid-containing ligands is weak. Significant efforts have been devoted toward the development of chemically modified sialoside ligands to target Siglecs with higher affinity and selectivity. In this review we discuss natural and synthetic sialoside ligands for each human Siglec, emphasizing the ligand binding determinants uncovered from recent advances in protein structural information. Potential therapeutic applications of these ligands are also discussed.
Collapse
Affiliation(s)
- Levon D Movsisyan
- Department of Chemistry, University of Alberta, Edmonton, Alberta, Canada
| | - Matthew S Macauley
- Department of Chemistry, University of Alberta, Edmonton, Alberta, Canada and Department of Medical Microbiology and Immunology, University of Alberta, Edmonton, Alberta, Canada.
| |
Collapse
|
124
|
Wang S, Qin H, Dong J, Hu L, Ye M. Multi-histidine functionalized material for the specific enrichment of sialylated glycopeptides. J Chromatogr A 2020; 1627:461422. [PMID: 32823117 DOI: 10.1016/j.chroma.2020.461422] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2020] [Revised: 06/30/2020] [Accepted: 07/19/2020] [Indexed: 12/26/2022]
Abstract
Sialylation, an important form of glycosylation, is involved in many biological processes and plays an important role in the development of diseases. However, due to the low abundance among various glycosylation and lack of efficient enrichment method with high specificity, the study of sialylation remains a challenge. Herein, multi-histidine modified microspheres (MHM) were synthesized to enrich sialylated glycopeptides. It was found that MHM could selectively enrich sialylated glycopeptides from over 100 times of non-sialylated glycopeptides, which indicated MHM possessed good enrichment specificity towards sialylated glycopeptides. Furthermore, MHM were utilized to the large-scale analysis of protein sialylation, and 510 intact glycopeptides were identified with over 94.5% sialylated glycopeptide specificity from 4 μL human serum. The good specificity could be attributed to the synergistic effect by the electrostatic interaction and hydrophilic interaction. Hence, MHM could provide an alternative approach for the analysis of site-specific sialylation at proteome level from complex biological samples.
Collapse
Affiliation(s)
- Shuyue Wang
- Key Laboratory Molecular Enzymology and Engineering, the Ministry of Education, National Engineering Laboratory of AIDS Vaccine, School of Life Sciences, Jilin University, Changchun 130023, China; CAS Key Lab of Separation Sciences for Analytical Chemistry, National Chromatographic Research and Analysis Center, 457 Zhongshan Road, Dalian 116023, China
| | - Hongqiang Qin
- CAS Key Lab of Separation Sciences for Analytical Chemistry, National Chromatographic Research and Analysis Center, 457 Zhongshan Road, Dalian 116023, China
| | - Jing Dong
- CAS Key Lab of Separation Sciences for Analytical Chemistry, National Chromatographic Research and Analysis Center, 457 Zhongshan Road, Dalian 116023, China
| | - Lianghai Hu
- Key Laboratory Molecular Enzymology and Engineering, the Ministry of Education, National Engineering Laboratory of AIDS Vaccine, School of Life Sciences, Jilin University, Changchun 130023, China.
| | - Mingliang Ye
- CAS Key Lab of Separation Sciences for Analytical Chemistry, National Chromatographic Research and Analysis Center, 457 Zhongshan Road, Dalian 116023, China.
| |
Collapse
|
125
|
Ono K, Sanada Y, Kimura Y, Aoyama S, Ueda N, Katayama T, Nagahama K. A thin hydrogel barrier linked onto cell surface sialic acids through covalent bonds induces cancer cell death in vivo. Biomater Sci 2020; 8:577-585. [PMID: 31872195 DOI: 10.1039/c9bm01758e] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Hypersialylation is the aberrant expression of sialic acid in cell surface glycans and is pervasive in cancer cells. Recent studies have shown that hypersialylation provides a microenvironment conducive to cancer progression, mediated by the interaction between sialic acid and sialic acid-binding receptors. Therefore, a technique to block the interaction between the overexpressed sialic acid on cancer cell surfaces and its receptors is a promising approach to develop new cancer therapies. We focused on hydrogels as an artificial barrier to block this interaction and present here the development of a novel technique for selectively covalently binding a thin hydrogel barrier on sialic acid residues on cancer cell surfaces. This technique effectively inhibited cancer cell adhesion, motility and growth, caused cancer cell death in vitro, and completely suppressed tumor growth in vivo, thereby clearly demonstrating a potent antitumor effect.
Collapse
Affiliation(s)
- Kimika Ono
- Department of Nanobiochemistry, Frontiers of Innovative Research in Science and Technology (FIRST), Konan University, 7-1-20 Minatojima-Minamimachi, Chuo-ku, Kobe 650-0047, Japan.
| | | | | | | | | | | | | |
Collapse
|
126
|
Wielgat P, Rogowski K, Niemirowicz-Laskowska K, Car H. Sialic Acid-Siglec Axis as Molecular Checkpoints Targeting of Immune System: Smart Players in Pathology and Conventional Therapy. Int J Mol Sci 2020; 21:ijms21124361. [PMID: 32575400 PMCID: PMC7352527 DOI: 10.3390/ijms21124361] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Revised: 06/17/2020] [Accepted: 06/17/2020] [Indexed: 12/11/2022] Open
Abstract
The sialic acid-based molecular mimicry in pathogens and malignant cells is a regulatory mechanism that leads to cross-reactivity with host antigens resulting in suppression and tolerance in the immune system. The interplay between sialoglycans and immunoregulatory Siglec receptors promotes foreign antigens hiding and immunosurveillance impairment. Therefore, molecular targeting of immune checkpoints, including sialic acid-Siglec axis, is a promising new field of inflammatory disorders and cancer therapy. However, the conventional drugs used in regular management can interfere with glycome machinery and exert a divergent effect on immune controlling systems. Here, we focus on the known effects of standard therapies on the sialoglycan-Siglec checkpoint and their importance in diagnosis, prediction, and clinical outcomes.
Collapse
Affiliation(s)
- Przemyslaw Wielgat
- Department of Clinical Pharmacology, Medical University of Bialystok, Waszyngtona 15A, 15-274 Bialystok, Poland;
- Correspondence: ; Tel.: +48-85-7450-647
| | - Karol Rogowski
- Department of Experimental Pharmacology, Medical University of Bialystok, Szpitalna 37, 15-295 Bialystok, Poland; (K.R.); (K.N.-L.)
| | - Katarzyna Niemirowicz-Laskowska
- Department of Experimental Pharmacology, Medical University of Bialystok, Szpitalna 37, 15-295 Bialystok, Poland; (K.R.); (K.N.-L.)
| | - Halina Car
- Department of Clinical Pharmacology, Medical University of Bialystok, Waszyngtona 15A, 15-274 Bialystok, Poland;
- Department of Experimental Pharmacology, Medical University of Bialystok, Szpitalna 37, 15-295 Bialystok, Poland; (K.R.); (K.N.-L.)
| |
Collapse
|
127
|
Gebri E, Kovács Z, Mészáros B, Tóth F, Simon Á, Jankovics H, Vonderviszt F, Kiss A, Guttman A, Hortobágyi T. N-Glycosylation Alteration of Serum and Salivary Immunoglobulin a Is a Possible Biomarker in Oral Mucositis. J Clin Med 2020; 9:jcm9061747. [PMID: 32516910 PMCID: PMC7355945 DOI: 10.3390/jcm9061747] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2020] [Revised: 05/30/2020] [Accepted: 06/03/2020] [Indexed: 02/07/2023] Open
Abstract
Background: Oral and enteral mucositis due to high-dose cytostatic treatment administered during autologous and allogeneic stem-cell transplantation increases mortality. Salivary secretory immunoglobulin A (sIgA) is a basic pillar of local immunity in the first line of defense. Altered salivary sialoglycoprotein carbohydrates are important in the pathologies in the oral cavity including inflammation, infection and neoplasia. Therefore, we assessed whether changes in the salivary and serum IgA glycosylation correlated with development and severity of oral mucositis. Methods: Using capillary electrophoresis, comparative analysis of serum and salivary IgA total N-glycans was conducted in 8 patients with autologous peripheral stem-cell transplantation (APSCT) at four different stages of transplantation (day −3/−7, 0, +7, +14) and in 10 healthy controls. Results: Fourteen out of the 31 structures identified in serum and 6 out of 38 in saliva showed significant changes upon transplantation compared with the control group. Only serum core fucosylated, sialylated bisecting biantennary glycan (FA2BG2S2) showed significant differences between any two stages of transplantation (day −3/−7 and day +14; p = 0.0279). Conclusion: Our results suggest that changes in the serum IgA total N-glycan profile could serve as a disease-specific biomarker in patients undergoing APSCT, while analysis of salivary IgA N-glycan reflects the effect of APSCT on local immunity.
Collapse
Affiliation(s)
- Enikő Gebri
- Department of Dentoalveolar Surgery and Dental Outpatient Care, Faculty of Dentistry, University of Debrecen, Nagyerdei krt. 98., H-4032 Debrecen, Hungary;
| | - Zsuzsanna Kovács
- Horváth Csaba Laboratory of Bioseparation Sciences, Research Center for Molecular Medicine, Doctoral School of Molecular Medicine, Faculty of Medicine, University of Debrecen, Nagyerdei krt. 98., H-4032 Debrecen, Hungary; (Z.K.); (B.M.); (Á.S.); (A.G.)
- Research Institute of Biomolecular and Chemical Engineering, University of Pannonia, Egyetem u 10., H-8200 Veszprém, Hungary; (H.J.); (F.V.)
| | - Brigitta Mészáros
- Horváth Csaba Laboratory of Bioseparation Sciences, Research Center for Molecular Medicine, Doctoral School of Molecular Medicine, Faculty of Medicine, University of Debrecen, Nagyerdei krt. 98., H-4032 Debrecen, Hungary; (Z.K.); (B.M.); (Á.S.); (A.G.)
- Research Institute of Biomolecular and Chemical Engineering, University of Pannonia, Egyetem u 10., H-8200 Veszprém, Hungary; (H.J.); (F.V.)
| | - Ferenc Tóth
- Department of Biomaterials and Prosthetic Dentistry, Faculty of Dentistry, University of Debrecen, Nagyerdei krt. 98., H-4032 Debrecen, Hungary;
| | - Ádám Simon
- Horváth Csaba Laboratory of Bioseparation Sciences, Research Center for Molecular Medicine, Doctoral School of Molecular Medicine, Faculty of Medicine, University of Debrecen, Nagyerdei krt. 98., H-4032 Debrecen, Hungary; (Z.K.); (B.M.); (Á.S.); (A.G.)
| | - Hajnalka Jankovics
- Research Institute of Biomolecular and Chemical Engineering, University of Pannonia, Egyetem u 10., H-8200 Veszprém, Hungary; (H.J.); (F.V.)
| | - Ferenc Vonderviszt
- Research Institute of Biomolecular and Chemical Engineering, University of Pannonia, Egyetem u 10., H-8200 Veszprém, Hungary; (H.J.); (F.V.)
| | - Attila Kiss
- Department of Hematopoietic Transplantation Centre, Faculty of Medicine, University of Debrecen, Nagyerdei krt. 98., H-4032 Debrecen, Hungary;
| | - András Guttman
- Horváth Csaba Laboratory of Bioseparation Sciences, Research Center for Molecular Medicine, Doctoral School of Molecular Medicine, Faculty of Medicine, University of Debrecen, Nagyerdei krt. 98., H-4032 Debrecen, Hungary; (Z.K.); (B.M.); (Á.S.); (A.G.)
- Research Institute of Biomolecular and Chemical Engineering, University of Pannonia, Egyetem u 10., H-8200 Veszprém, Hungary; (H.J.); (F.V.)
| | - Tibor Hortobágyi
- Institute of Pathology, Faculty of Medicine, University of Szeged, Állomás utca 1., H-6725 Szeged, Hungary
- MTA-DE Cerebrovascular and Neurodegenerative Research Group, Department of Neurology, Faculty of Medicine, University of Debrecen, Nagyerdei krt. 98., H-4032 Debrecen, Hungary
- Institute of Psychiatry Psychology and Neuroscience, King’s College London, De Crespigny Park, London SE5 8AF, UK
- Correspondence: ; Tel.: +36-30-687-5983
| |
Collapse
|
128
|
Tumor microenvironment, immune response and post-radiotherapy tumor clearance. Clin Transl Oncol 2020; 22:2196-2205. [PMID: 32445035 DOI: 10.1007/s12094-020-02378-8] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2020] [Accepted: 05/07/2020] [Indexed: 02/06/2023]
Abstract
Radiotherapy is the treatment of choice for many cancer patients. Residual tumor leads to local recurrence after a period of an equilibrium created between proliferating, quiescent and dying cancer cells. The tumor microenvironment is a main obstacle for the efficacy of radiotherapy, as impaired blood flow leads to hypoxia, acidity and reduced accessibility of radiosensitizers. Eradication of remnant disease is an intractable clinical quest. After more than a century of research, anti-tumor immunity has gained a dominant position in oncology research and therapy. Immune cells play a significant role in the eradication of tumors during and after the completion of radiotherapy. The tumor equilibrium reached in the irradiated tumor may shift towards cancer cell eradication if the immune response is appropriately modulated. In the modern immunotherapy era, clinical trials are urged to standardize immunotherapy schemes that could be safely applied to improve clearance of the post-radiotherapy remnant disease.
Collapse
|
129
|
Meril S, Harush O, Reboh Y, Matikhina T, Barliya T, Cohen CJ. Targeting glycosylated antigens on cancer cells using siglec‐7/9‐based CAR T‐cells. Mol Carcinog 2020; 59:713-723. [DOI: 10.1002/mc.23213] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2020] [Revised: 04/29/2020] [Accepted: 04/30/2020] [Indexed: 01/05/2023]
Affiliation(s)
- Sara Meril
- Division of Molecular, Cellular and Medical BiologyThe Laboratory of Tumor Immunology and Immunotherapy, The Mina and Everard Goodman Faculty of Life Sciences, Bar‐Ilan University Ramat Gan Israel
| | - Ortal Harush
- Division of Molecular, Cellular and Medical BiologyThe Laboratory of Tumor Immunology and Immunotherapy, The Mina and Everard Goodman Faculty of Life Sciences, Bar‐Ilan University Ramat Gan Israel
| | - Yishai Reboh
- Division of Molecular, Cellular and Medical BiologyThe Laboratory of Tumor Immunology and Immunotherapy, The Mina and Everard Goodman Faculty of Life Sciences, Bar‐Ilan University Ramat Gan Israel
| | - Tatyana Matikhina
- Division of Molecular, Cellular and Medical BiologyThe Laboratory of Tumor Immunology and Immunotherapy, The Mina and Everard Goodman Faculty of Life Sciences, Bar‐Ilan University Ramat Gan Israel
| | - Tilda Barliya
- Division of Molecular, Cellular and Medical BiologyThe Laboratory of Tumor Immunology and Immunotherapy, The Mina and Everard Goodman Faculty of Life Sciences, Bar‐Ilan University Ramat Gan Israel
| | - Cyrille J. Cohen
- Division of Molecular, Cellular and Medical BiologyThe Laboratory of Tumor Immunology and Immunotherapy, The Mina and Everard Goodman Faculty of Life Sciences, Bar‐Ilan University Ramat Gan Israel
| |
Collapse
|
130
|
Marini M, Tani A, Manetti M, Sgambati E. Characterization and distribution of sialic acids in human testicular seminoma. Acta Histochem 2020; 122:151532. [PMID: 32143917 DOI: 10.1016/j.acthis.2020.151532] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2020] [Revised: 02/21/2020] [Accepted: 02/27/2020] [Indexed: 12/15/2022]
Abstract
Aberrant content of sialic acids (Sias) has been observed in various human cancer types in different organs. Sias have been implicated in cancerous transformation, invasiveness and metastasis, and in the escaping of cancer cells from immune surveillance. Indeed, Sias are commonly regarded as important biomarkers to distinguish cancer cells from their healthy counterparts. However, scarce and not exhaustive investigations have been performed on Sia content in testicular cancers and, in particular, in seminoma, one of the most common malignant testicular tumors. Hence, the aim of this study was to investigate the content and distribution of Sias with different glycosidic linkage, namely α2,3 and α2,6 galactose- or N-acetyl-galactosamine-linked Sias and polymeric Sia (polySia), in the germinal and stromal components of human testes affected by seminoma compared to normal testicular tissue. Structural changes in seminoma tissue were examined using hematoxylin-eosin staining. α2,3 and α2,6 linked Sias were evaluated by lectin histochemistry (Maackia amurensis agglutinin (MAA) and Sambucus nigra agglutinin (SNA)), while confocal immunofluorescence was used for polySia detection. Histopathological findings in seminoma tissue included loss of seminiferous tubules replaced by clusters of uniform polygonal cells with a clear cytoplasm, bundles of fibrotic tissue, numerous microvessels and some atrophic tubules. The content of α2,3 and α2,6 linked Sias was lost in almost all seminoma components respect to normal tissue, with the exception of microvessels in which it was higher. On the contrary, polySia level was increased in all the seminoma components compared to normal testicular tissue. Our findings suggest that an aberrant content of different Sias might have important and differential roles in seminoma development and progression. In particular, polySia might be implicated in seminoma progression by promoting cancer invasiveness and regulating the cross-talk between cancer cells, reactive stroma and vessels. Thus, the possibility that polySia might represent an important biomarker for seminoma deserves further investigation.
Collapse
Affiliation(s)
- Mirca Marini
- Department of Experimental and Clinical Medicine, Section of Anatomy and Histology, University of Florence, Largo Brambilla 3, 50134, Florence, Italy.
| | - Alessia Tani
- Department of Experimental and Clinical Medicine, Section of Anatomy and Histology, University of Florence, Largo Brambilla 3, 50134, Florence, Italy.
| | - Mirko Manetti
- Department of Experimental and Clinical Medicine, Section of Anatomy and Histology, University of Florence, Largo Brambilla 3, 50134, Florence, Italy.
| | - Eleonora Sgambati
- Department of Biosciences and Territory, University of Molise, Contrada Fonte Lappone, 86090 Pesche (Isernia), Italy.
| |
Collapse
|
131
|
Abstract
Sialylation (the covalent addition of sialic acid to the terminal end of glycoproteins or glycans), tightly regulated cell- and microenvironment-specific process and orchestrated by sialyltransferases and sialidases (neuraminidases) family, is one of the posttranslational modifications, which plays an important biological role in the maintenance of normal physiology and involves many pathological dysfunctions. Glycans have roles in all the cancer hallmarks, referring to capabilities acquired during all steps of cancer development to initiate malignant transformation (a driver of a malignant genotype), enable cancer cells to survive, proliferate, and metastasize (a consequence of a malignant phenotype), which includes sustaining proliferative signaling, evading growth suppressor, resisting cell apoptosis, enabling replicative immortality, inducing angiogenesis, reprogramming of energy metabolism, evading tumor destruction, accumulating inflammatory microenvironment, and activating invasion and accelerating metastases. Regarding the important role of altered sialylation of cancers, further knowledge about the initiation and the consequences of altered sialylation pattern in tumor cells is needed, because all may offer a better chance for developing novel therapeutic strategy. In this review, we would like to update alteration of sialylation in ovarian cancers.
Collapse
Affiliation(s)
- Wen-Ling Lee
- Department of Medicine, Cheng-Hsin General Hospital, Taipei, Taiwan, ROC
- Department of Nursing, Oriental Institute of Technology, New Taipei City, Taiwan, ROC
- Institute of Clinical Medicine, National Yang-Ming University, Taipei, Taiwan, ROC
| | - Peng-Hui Wang
- Institute of Clinical Medicine, National Yang-Ming University, Taipei, Taiwan, ROC
- Department of Obstetrics and Gynecology, Taipei Veterans General Hospital, Taipei, Taiwan, ROC
- Department of Medical Research, China Medical University Hospital, Taichung, Taiwan, ROC
| |
Collapse
|
132
|
Bauer TJ, Gombocz E, Wehland M, Bauer J, Infanger M, Grimm D. Insight in Adhesion Protein Sialylation and Microgravity Dependent Cell Adhesion-An Omics Network Approach. Int J Mol Sci 2020; 21:ijms21051749. [PMID: 32143440 PMCID: PMC7084616 DOI: 10.3390/ijms21051749] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2020] [Revised: 02/28/2020] [Accepted: 03/02/2020] [Indexed: 12/18/2022] Open
Abstract
The adhesion behavior of human tissue cells changes in vitro, when gravity forces affecting these cells are modified. To understand the mechanisms underlying these changes, proteins involved in cell-cell or cell-extracellular matrix adhesion, their expression, accumulation, localization, and posttranslational modification (PTM) regarding changes during exposure to microgravity were investigated. As the sialylation of adhesion proteins is influencing cell adhesion on Earth in vitro and in vivo, we analyzed the sialylation of cell adhesion molecules detected by omics studies on cells, which change their adhesion behavior when exposed to microgravity. Using a knowledge graph created from experimental omics data and semantic searches across several reference databases, we studied the sialylation of adhesion proteins glycosylated at their extracellular domains with regards to its sensitivity to microgravity. This way, experimental omics data networked with the current knowledge about the binding of sialic acids to cell adhesion proteins, its regulation, and interactions in between those proteins provided insights into the mechanisms behind our experimental findings, suggesting that balancing the sialylation against the de-sialylation of the terminal ends of the adhesion proteins' glycans influences their binding activity. This sheds light on the transition from two- to three-dimensional growth observed in microgravity, mirroring cell migration and cancer metastasis in vivo.
Collapse
Affiliation(s)
- Thomas J. Bauer
- Clinic for Plastic, Aesthetic and Hand Surgery, Otto-von-Guericke-University Magdeburg, D-39120 Magdeburg, Germany; (T.J.B.); (M.W.); (M.I.); (D.G.)
| | - Erich Gombocz
- Melissa Informatics, 2550 Ninth Street, Suite 114, Berkeley, CA 94710, USA;
| | - Markus Wehland
- Clinic for Plastic, Aesthetic and Hand Surgery, Otto-von-Guericke-University Magdeburg, D-39120 Magdeburg, Germany; (T.J.B.); (M.W.); (M.I.); (D.G.)
| | - Johann Bauer
- Max Planck Institute of Biochemistry, D-82152 Martinsried, Germany
- Correspondence: ; Tel.: +49-89-85783803
| | - Manfred Infanger
- Clinic for Plastic, Aesthetic and Hand Surgery, Otto-von-Guericke-University Magdeburg, D-39120 Magdeburg, Germany; (T.J.B.); (M.W.); (M.I.); (D.G.)
| | - Daniela Grimm
- Clinic for Plastic, Aesthetic and Hand Surgery, Otto-von-Guericke-University Magdeburg, D-39120 Magdeburg, Germany; (T.J.B.); (M.W.); (M.I.); (D.G.)
- Department of Biomedicine, Aarhus University Hospital, DK-8000 Aarhus C, Denmark
- Department of Microgravity and Translational Regenerative Medicine, Otto von Guericke University, Pfälzer Platz, 39106 Magdeburg, Germany
| |
Collapse
|
133
|
Ashdown CP, Johns SC, Aminov E, Unanian M, Connacher W, Friend J, Fuster MM. Pulsed Low-Frequency Magnetic Fields Induce Tumor Membrane Disruption and Altered Cell Viability. Biophys J 2020; 118:1552-1563. [PMID: 32142642 PMCID: PMC7136334 DOI: 10.1016/j.bpj.2020.02.013] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2019] [Revised: 12/12/2019] [Accepted: 02/10/2020] [Indexed: 12/03/2022] Open
Abstract
Tumor cells express a unique cell surface glycocalyx with upregulation of sulfated glycosaminoglycans and charged glycoproteins. Little is known about how electromagnetic fields interact with this layer, particularly with regard to harnessing unique properties for therapeutic benefit. We applied a pulsed 20-millitesla (mT) magnetic field with rate of rise (dB/dt) in the msec range to cultured tumor cells to assess whether this affects membrane integrity as measured using cytolytic assays. A 10-min exposure of A549 human lung cancer cells to sequential 50- and 385-Hz oscillating magnetic fields was sufficient to induce intracellular protease release, suggesting altered membrane integrity after the field exposure. Heparinase treatment, which digests anionic sulfated glycan polymers, before exposure rendered cells insensitive to this effect. We further examined a non-neoplastic human primary cell line (lung lymphatic endothelial cells) as a typical normal host cell from the lung cancer microenvironment and found no effect of field exposure on membrane integrity. The field exposure was also sufficient to alter proliferation of tumor cells in culture, but not that of normal lymphatic cells. Pulsed magnetic field exposure of human breast cancer cells that express a sialic-acid rich glycocalyx also induced protease release, and this was partially abrogated by sialidase pretreatment, which removes cell surface anionic sialic acid. Scanning electron microscopy showed that field exposure may induce unique membrane “rippling” along with nanoscale pores on A549 cells. These effects were caused by a short exposure to pulsed 20-mT magnetic fields, and future work may examine greater magnitude effects. The proof of concept herein points to a mechanistic basis for possible applications of pulsed magnetic fields in novel anticancer strategies.
Collapse
Affiliation(s)
- Christopher P Ashdown
- VA San Diego Healthcare System, San Diego, California; Division of Biological Sciences, University of California, San Diego, La Jolla, California
| | - Scott C Johns
- VA San Diego Healthcare System, San Diego, California; Veterans Medical Research Foundation, San Diego, California
| | - Edward Aminov
- Department of Mechanical and Aerospace Engineering, University of California, San Diego, La Jolla, California
| | - Michael Unanian
- School of Electrical Engineering, Columbia University, New York, New York
| | - William Connacher
- Department of Mechanical and Aerospace Engineering, University of California, San Diego, La Jolla, California
| | - James Friend
- Department of Mechanical and Aerospace Engineering, University of California, San Diego, La Jolla, California
| | - Mark M Fuster
- VA San Diego Healthcare System, San Diego, California; Veterans Medical Research Foundation, San Diego, California; Department of Medicine, Division of Pulmonary & Critical Care, University of California, San Diego, La Jolla, California; Glycobiology Research and Training Center, University of California, San Diego, La Jolla, California.
| |
Collapse
|
134
|
Ding Y, Vara Prasad CVNS, Wang B. Glycosylation on Unprotected or Partially Protected Acceptors. European J Org Chem 2020. [DOI: 10.1002/ejoc.201901675] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Affiliation(s)
- Yili Ding
- Life Science Department; Foshan University; 528000 Foshan Guangdong China
| | | | - Bingyun Wang
- Life Science Department; Foshan University; 528000 Foshan Guangdong China
| |
Collapse
|
135
|
Busold S, Nagy NA, Tas SW, van Ree R, de Jong EC, Geijtenbeek TBH. Various Tastes of Sugar: The Potential of Glycosylation in Targeting and Modulating Human Immunity via C-Type Lectin Receptors. Front Immunol 2020; 11:134. [PMID: 32117281 PMCID: PMC7019010 DOI: 10.3389/fimmu.2020.00134] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2019] [Accepted: 01/20/2020] [Indexed: 12/31/2022] Open
Abstract
C-type lectin receptors (CLRs) are important in several immune regulatory processes. These receptors recognize glycans expressed by host cells or by pathogens. Whereas pathogens are recognized through their glycans, which leads to protective immunity, aberrant cellular glycans are now increasingly recognized as disease-driving factors in cancer, auto-immunity, and allergy. The vast variety of glycan structures translates into a wide spectrum of effects on the immune system ranging from immune suppression to hyper-inflammatory responses. CLRs have distinct expression patterns on antigen presenting cells (APCs) controlling their role in immunity. CLRs can also be exploited to selectively target specific APCs, modulate immune responses and enhance antigen presentation. Here we will discuss the role of glycans and their receptors in immunity as well as potential strategies for immune modulation. A special focus will be given to different dendritic cell subsets as these APCs are crucial orchestrators of immune responses in infections, cancer, auto-immunity and allergies. Furthermore, we will highlight the potential use of nanoscale lipid bi-layer structures (liposomes) in targeted immunotherapy.
Collapse
Affiliation(s)
- Stefanie Busold
- Department of Experimental Immunology, Amsterdam University Medical Centers, Amsterdam Institute for Infection and Immunity, University of Amsterdam, Amsterdam, Netherlands
| | - Noémi A Nagy
- Department of Experimental Immunology, Amsterdam University Medical Centers, Amsterdam Institute for Infection and Immunity, University of Amsterdam, Amsterdam, Netherlands
| | - Sander W Tas
- Department of Experimental Immunology, Amsterdam University Medical Centers, Amsterdam Institute for Infection and Immunity, University of Amsterdam, Amsterdam, Netherlands.,Department of Rheumatology and Clinical Immunology, Amsterdam University Medical Centers, Amsterdam Rheumatology and Immunology Center, University of Amsterdam, Amsterdam, Netherlands
| | - Ronald van Ree
- Department of Experimental Immunology, Amsterdam University Medical Centers, Amsterdam Institute for Infection and Immunity, University of Amsterdam, Amsterdam, Netherlands.,Department of Otorhinolaryngology, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, Netherlands
| | - Esther C de Jong
- Department of Experimental Immunology, Amsterdam University Medical Centers, Amsterdam Institute for Infection and Immunity, University of Amsterdam, Amsterdam, Netherlands
| | - Teunis B H Geijtenbeek
- Department of Experimental Immunology, Amsterdam University Medical Centers, Amsterdam Institute for Infection and Immunity, University of Amsterdam, Amsterdam, Netherlands
| |
Collapse
|
136
|
Natoni A, Farrell ML, Harris S, Falank C, Kirkham-McCarthy L, Macauley MS, Reagan MR, O’Dwyer M. Sialyltransferase inhibition leads to inhibition of tumor cell interactions with E-selectin, VCAM1, and MADCAM1, and improves survival in a human multiple myeloma mouse model. Haematologica 2020; 105:457-467. [PMID: 31101754 PMCID: PMC7012485 DOI: 10.3324/haematol.2018.212266] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2018] [Accepted: 05/16/2019] [Indexed: 12/21/2022] Open
Abstract
Aberrant glycosylation resulting from altered expression of sialyltransferases, such as ST3 β-galactoside α2-3-sialyltransferase 6, plays an important role in disease progression in multiple myeloma (MM). Hypersialylation can lead to increased immune evasion, drug resistance, tumor invasiveness, and disseminated disease. In this study, we explore the in vitro and in vivo effects of global sialyltransferase inhibition on myeloma cells using the pan-sialyltransferase inhibitor 3Fax-Neu5Ac delivered as a per-acetylated methyl ester pro-drug. Specifically, we show in vivo that 3Fax-Neu5Ac improves survival by enhancing bortezomib sensitivity in an aggressive mouse model of MM. However, 3Fax-Neu5Ac treatment of MM cells in vitro did not reverse bortezomib resistance conferred by bone marrow (BM) stromal cells. Instead, 3Fax-Neu5Ac significantly reduced interactions of myeloma cells with E-selectin, MADCAM1 and VCAM1, suggesting that reduced sialylation impairs extravasation and retention of myeloma cells in the BM. Finally, we showed that 3Fax-Neu5Ac alters the post-translational modification of the α4 integrin, which may explain the reduced affinity of α4β1/α4β7 integrins for their counter-receptors. We propose that inhibiting sialylation may represent a valuable strategy to restrict myeloma cells from entering the protective BM microenvironment, a niche in which they are normally protected from chemotherapeutic agents such as bortezomib. Thus, our work demonstrates that targeting sialylation to increase the ratio of circulating to BM-resident MM cells represents a new avenue that could increase the efficacy of other anti-myeloma therapies and holds great promise for future clinical applications.
Collapse
Affiliation(s)
- Alessandro Natoni
- Apoptosis Research Center, National University of Ireland, Galway, Ireland
| | - Mariah L. Farrell
- Maine Medical Center Research Institute, Scarborough, ME, USA,Tufts University School of Medicine, Boston, MA, USA,Graduate School of Biomedical Science and Engineering, University of Maine, Orono, ME, USA
| | - Sophie Harris
- Maine Medical Center Research Institute, Scarborough, ME, USA,Tufts University School of Medicine, Boston, MA, USA,Graduate School of Biomedical Science and Engineering, University of Maine, Orono, ME, USA
| | - Carolyne Falank
- Maine Medical Center Research Institute, Scarborough, ME, USA,Tufts University School of Medicine, Boston, MA, USA,Graduate School of Biomedical Science and Engineering, University of Maine, Orono, ME, USA
| | | | - Matthew S. Macauley
- Department of Chemistry and Department of Medical Microbiology and Immunology, University of Alberta, Alberta, Canada
| | - Michaela R. Reagan
- Maine Medical Center Research Institute, Scarborough, ME, USA,Tufts University School of Medicine, Boston, MA, USA,Graduate School of Biomedical Science and Engineering, University of Maine, Orono, ME, USA
| | - Michael O’Dwyer
- Apoptosis Research Center, National University of Ireland, Galway, Ireland,Correspondence: MICHAEL O’DWYER,
| |
Collapse
|
137
|
Rossi GR, Trindade ES, Souza-Fonseca-Guimaraes F. Tumor Microenvironment-Associated Extracellular Matrix Components Regulate NK Cell Function. Front Immunol 2020; 11:73. [PMID: 32063906 PMCID: PMC7000552 DOI: 10.3389/fimmu.2020.00073] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2019] [Accepted: 01/13/2020] [Indexed: 12/22/2022] Open
Abstract
The tumor microenvironment (TME) is composed of multiple infiltrating host cells (e.g., endothelial cells, fibroblasts, lymphocytes, and myeloid cells), extracellular matrix, and various secreted or cell membrane-presented molecules. Group 1 innate lymphoid cells (ILCs), which includes natural killer (NK) cells and ILC1, contribute to protecting the host against cancer and infection. Both subsets are able to quickly produce cytokines such as interferon gamma (IFN-γ), chemokines, and other growth factors in response to activating signals. However, the TME provides many molecules that can prevent the potential effector function of these cells, thereby protecting the tumor. For example, TME-derived tumor growth factor (TGF)-β and associated members of the superfamily downregulate NK cell cytotoxicity, cytokine secretion, metabolism, proliferation, and induce effector NK cells to upregulate ILC1-like characteristics. In concert, a family of carbohydrate-binding proteins called galectins, which can be produced by different cells composing the TME, can downregulate NK cell function. Matrix metalloproteinase (MMP) and a disintegrin and metalloproteinase (ADAM) are also enzymes that can remodel the extracellular matrix and shred receptors from the tumor cell surface, impairing the activation of NK cells and leading to less effective effector functions. Gaining a better understanding of the characteristics of the TME and its associated factors, such as infiltrating cells and extracellular matrix, could lead to tailoring of new personalized immunotherapy approaches. This review provides an overview of our current knowledge on the impact of the TME and extracellular matrix-associated components on differentiation, impairment, and function of NK cells.
Collapse
Affiliation(s)
| | - Edvaldo S Trindade
- Cellular Biology Department, Federal University of Paraná, Curitiba, Brazil
| | | |
Collapse
|
138
|
Sialic acid and biology of life: An introduction. SIALIC ACIDS AND SIALOGLYCOCONJUGATES IN THE BIOLOGY OF LIFE, HEALTH AND DISEASE 2020. [PMCID: PMC7153325 DOI: 10.1016/b978-0-12-816126-5.00001-9] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Sialic acids are important molecule with high structural diversity. They are known to occur in higher animals such as Echinoderms, Hemichordata, Cephalochorda, and Vertebrata and also in other animals such as Platyhelminthes, Cephalopoda, and Crustaceae. Plants are known to lack sialic acid. But they are reported to occur in viruses, bacteria, protozoa, and fungi. Deaminated neuraminic acid although occurs in vertebrates and bacteria, is reported to occur in abundance in the lower vertebrates. Sialic acids are mostly located in terminal ends of glycoproteins and glycolipids, capsular and tissue polysialic acids, bacterial lipooligosaccharides/polysaccharides, and in different forms that dictate their role in biology. Sialic acid play important roles in human physiology of cell-cell interaction, communication, cell-cell signaling, carbohydrate-protein interactions, cellular aggregation, development processes, immune reactions, reproduction, and in neurobiology and human diseases in enabling the infection process by bacteria and virus, tumor growth and metastasis, microbiome biology, and pathology. It enables molecular mimicry in pathogens that allows them to escape host immune responses. Recently sialic acid has found role in therapeutics. In this chapter we have highlighted the (i) diversity of sialic acid, (ii) their occurrence in the diverse life forms, (iii) sialylation and disease, and (iv) sialic acid and therapeutics.
Collapse
|
139
|
Wellington N, Macklai S, Britz-McKibbin P. Elucidating the Anomalous Binding Enhancement of Isoquinoline Boronic Acid for Sialic Acid Under Acidic Conditions: Expanding Biorecognition Beyond Vicinal Diols. Chemistry 2019; 25:15277-15280. [PMID: 31596002 DOI: 10.1002/chem.201904442] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2019] [Indexed: 11/06/2022]
Abstract
A zwitterionic heterocyclic boronic acid based on 4-isoquinolineboronic acid (IQBA) exhibits the highest reported binding affinity for sialic acid or N-acetylneuraminic acid (Neu5Ac, K=5390±190 m-1 ) through the formation of a cyclic boronate ester complex under acidic conditions (pH 3). This anomalous pH-dependent binding enhancement does not occur with common neutral saccharides (e.g., glucose, fructose, sorbitiol), because it is mediated via selective complexation to a α-hydroxycarboxylate moiety forming a stable ion pair and ternary complex with Neu5Ac in phosphate buffer. IQBA expands biorecognition beyond classical vicinal diols under neutral or alkaline buffer conditions, which enables the direct analysis of Neu5Ac by native fluorescence with sub-micromolar detection limits.
Collapse
Affiliation(s)
- Nadine Wellington
- Department of Chemistry and Chemical Biology, McMaster University, 1280 Main St. W., Hamilton, ON, Canada
| | - Sabrina Macklai
- Department of Chemistry and Chemical Biology, McMaster University, 1280 Main St. W., Hamilton, ON, Canada
| | - Philip Britz-McKibbin
- Department of Chemistry and Chemical Biology, McMaster University, 1280 Main St. W., Hamilton, ON, Canada
| |
Collapse
|
140
|
Ortiz-Soto ME, Reising S, Schlosser A, Seibel J. Structural and functional role of disulphide bonds and substrate binding residues of the human beta-galactoside alpha-2,3-sialyltransferase 1 (hST3Gal1). Sci Rep 2019; 9:17993. [PMID: 31784620 PMCID: PMC6884586 DOI: 10.1038/s41598-019-54384-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2019] [Accepted: 11/11/2019] [Indexed: 12/27/2022] Open
Abstract
Overexpression of hST3Gal1 leads to hypersialylation of cell-surface glycoconjugates, a cancer-associated condition that promotes cell growth, migration and invasion. Upregulation of this enzyme in ovarian cancer is linked to cancer progression and metastasis, contributing also to chemotherapy resistance. Strategies for preventing metastasis include the inhibition of hST3Gal1, which demands structure-based studies on its strict regioselectivity and substrate/donor preference. Herein we describe the contribution of various residues constituting donor CMP-Neu5Ac and acceptor Galβ1-3GalNAc-R binding sites to catalysis. Removal of hydrogen bonds and/or stacking interactions among substrates and residues Y191, Y230, N147, S148 and N170 affected the enzyme’s activity to a different extent, revealing the fine control needed for an optimal catalytic performance. To gain further understanding of the correlation among structure, activity and stability, the in vitro role of hST3Gal1 disulphide bonds was analysed. As expected, disruption of the Glycosyltransferase family 29 (GT29) invariant bond C142-C281, as well as the ST3Gal1 subfamily conserved disulphide C61-C139 inactivates the enzyme. While disulphide C59-C64 is not essential for function, its absence reduces the activity (kcat) for donor and acceptor substrates to about 67 and 72%, respectively, and diminishes the enzyme’s melting temperature (Tm) by 7 °C.
Collapse
Affiliation(s)
- Maria Elena Ortiz-Soto
- Institut für Organische Chemie, Universität Würzburg, Am Hubland, 97074, Würzburg, Germany
| | - Sabine Reising
- Institut für Organische Chemie, Universität Würzburg, Am Hubland, 97074, Würzburg, Germany
| | - Andreas Schlosser
- Rudolf-Virchow-Zentrum für Experimentelle Biomedizin, Universität Würzburg, Josef-Schneider Str. 2, Haus D15, 97080, Würzburg, Germany
| | - Jürgen Seibel
- Institut für Organische Chemie, Universität Würzburg, Am Hubland, 97074, Würzburg, Germany.
| |
Collapse
|
141
|
Ferreira HJ, de Almeida EM, da Silva WMB, Teixeira EH, do Nascimento Neto LG. Molecular Mechanisms Involved in the Antitumor Activity of Isolated Lectins from Marine Organisms: A Systematic Review. Curr Drug Targets 2019; 21:616-625. [PMID: 31763966 DOI: 10.2174/1389450120666191122113850] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2019] [Revised: 09/27/2019] [Accepted: 11/04/2019] [Indexed: 11/22/2022]
Abstract
INTRODUCTION Tumor cells may present several molecular alterations that favor their malignancy, among which there is the expression of tumor-related antigens, such as truncated T-glycans, Thomsen-nouvelle, sialyl-Lewis X and sialyl Tn, which may help in the diagnosis and treatment using specific target molecules. Lectins are ubiquitous proteins capable of interacting with specific carbohydrates. Lectins isolated from marine organisms have important characteristics such as low immunogenicity and can bind to complex glycans compared to plant lectins. OBJECTIVE This work evaluated, through a systematic review, the molecular mechanisms of antitumor activity of lectins isolated from marine organisms. METHODOLOGY The Pubmed, Lilacs, Science Direct, Wiley and Scopus databases were reviewed using the descriptors: marine lectin and cancer. Articles in English, published between January 2008 and December 2018, which proposed the molecular mechanisms of anticancer activity of lectins from marine organisms were eligible for the study. RESULTS 17 articles were eligible. The lectins showed promising performance against cancer cells, presenting specific cytotoxicity for some types of malignant cells. The articles presented several lectins specific to different carbohydrates, modulating: pro and anti-apoptotic proteins, transcription factor E2F-1, via mitogen-activated protein kinase. In addition, exogenous lectin expression in cancer cells has been shown to be a promising way to treat cancer. CONCLUSION This review showed the various studies that described the molecular mechanisms caused by marine lectins with antineoplastic potential. This knowledge is relevant for the development and use of the next generations of lectins isolated from marine organisms, supporting their potential in cancer treatment.
Collapse
Affiliation(s)
- Hugo Jefferson Ferreira
- Biomedicine College, Christus University Center, Fortaleza - CE, Brazil.,Integrated Laboratory of Biomolecules (LIBS), Federal University of Ceara, Department of Pathology and Forensic Medicine, 60430-160, Fortaleza, CE, Brazil
| | - Evandro Moreira de Almeida
- Biomedicine College, Christus University Center, Fortaleza - CE, Brazil.,Integrated Laboratory of Biomolecules (LIBS), Federal University of Ceara, Department of Pathology and Forensic Medicine, 60430-160, Fortaleza, CE, Brazil
| | | | - Edson Holanda Teixeira
- Integrated Laboratory of Biomolecules (LIBS), Federal University of Ceara, Department of Pathology and Forensic Medicine, 60430-160, Fortaleza, CE, Brazil
| | - Luiz Gonzaga do Nascimento Neto
- Integrated Laboratory of Biomolecules (LIBS), Federal University of Ceara, Department of Pathology and Forensic Medicine, 60430-160, Fortaleza, CE, Brazil.,Federal Institute of Education, Science and Technology of Ceará, Limoeiro do Norte Campus, 62930-000, Limoeiro do Norte, Ceara, Brazil
| |
Collapse
|
142
|
Cancer-derived sialylated IgG promotes tumor immune escape by binding to Siglecs on effector T cells. Cell Mol Immunol 2019; 17:1148-1162. [PMID: 31754235 DOI: 10.1038/s41423-019-0327-9] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2019] [Accepted: 10/21/2019] [Indexed: 12/11/2022] Open
Abstract
To date, IgG in the tumor microenvironment (TME) has been considered a product of B cells and serves as an antitumor antibody. However, in this study, using a monoclonal antibody against cancer-derived IgG (Cancer-IgG), we found that cancer cells could secrete IgG into the TME. Furthermore, Cancer-IgG, which carries an abnormal sialic acid modification in the CH1 domain, directly inhibited effector T-cell proliferation and significantly promoted tumor growth by reducing CD4+ and CD8+ T-cell infiltration into tumor tissues. Mechanistic studies showed that the immunosuppressive effect of sialylated Cancer-IgG is dependent on its sialylation and binding to sialic acid-binding immunoglobulin-type lectins (Siglecs) on effector CD4+ and CD8+ T cells. Importantly, we show that several Siglecs are overexpressed on effector T cells from cancer patients, but not those from healthy donors. These findings suggest that sialylated Cancer-IgG may be a ligand for Siglecs, which may serve as potential checkpoint proteins and mediate tumor immune evasion.
Collapse
|
143
|
Acharya S, Jin C, Bylund J, Shen Q, Kamali-Moghaddam M, Jontell M, Carlén A, Karlsson NG. Reduced sialyl-Lewis x on salivary MUC7 from patients with burning mouth syndrome. Mol Omics 2019; 15:331-339. [PMID: 31414088 DOI: 10.1039/c9mo00061e] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
We analysed and compared MUC7 O-glycosylation and inflammatory biomarkers in saliva from female patients with burning mouth syndrome (BMS) and gender/age-matched controls. Oligosaccharides from salivary MUC7 from BMS and controls were released. Inflammatory mediators were measured by multiplex proximity extension assay. Presence of sialyl-Lewisx (Si-Lex) epitope on MUC7 was confirmed using Western blot. MUC7 O-glycans and measured inflammatory biomarkers were found to be similar between BMS and controls. However, oligosaccharides sialyl-Lewisx (Si-Lex) was found to be reduced in samples from BMS patients. Positive correlation (combined patients and controls) was found between levels of C-C motif chemokine 19 (CCL-19) and the amount of core-2 oligosaccharides on MUC7 as well as fractalkine (CX3CL1) and level of sialylation. Patients with BMS were shown to represent a heterogeneous group in terms of inflammatory biomarkers. This indicates that BMS patients could be further stratified on the basis of low-level inflammation. The results furthermore indicate that reduced sialylation of MUC7, particularly Si-Lex, may be an important feature in patients with BMS. However, the functional aspects and potential involvement in immune regulation of Si-Lex remains unclear. Our data suggests a chemokine driven alteration of MUC7 glycosylation.
Collapse
Affiliation(s)
- Shikha Acharya
- Department of Oral Microbiology and Immunology, Institute of Odontology, The Sahlgrenska Academy, University of Gothenburg, PO 450, 40530, Gothenburg, Sweden.
| | | | | | | | | | | | | | | |
Collapse
|
144
|
Natoni A, Bohara R, Pandit A, O'Dwyer M. Targeted Approaches to Inhibit Sialylation of Multiple Myeloma in the Bone Marrow Microenvironment. Front Bioeng Biotechnol 2019; 7:252. [PMID: 31637237 PMCID: PMC6787837 DOI: 10.3389/fbioe.2019.00252] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2019] [Accepted: 09/17/2019] [Indexed: 11/13/2022] Open
Abstract
Aberrant glycosylation modulates different aspects of tumor biology, and it has long been recognized as a hallmark of cancer. Among the different forms of glycosylation, sialylation, the addition of sialic acid to underlying oligosaccharides, is often dysregulated in cancer. Increased expression of sialylated glycans has been observed in many types of cancer, including multiple myeloma, and often correlates with aggressive metastatic behavior. Myeloma, a cancer of plasma cells, develops in the bone marrow, and colonizes multiple sites of the skeleton including the skull. In myeloma, the bone marrow represents an essential niche where the malignant cells are nurtured by the microenvironment and protected from chemotherapy. Here, we discuss the role of hypersialylation in the metastatic process focusing on multiple myeloma. In particular, we examine how increased sialylation modulates homing of malignant plasma cells into the bone marrow by regulating the activity of molecules important in bone marrow cellular trafficking including selectins and integrins. We also propose that inhibiting sialylation may represent a new therapeutic strategy to overcome bone marrow-mediated chemotherapy resistance and describe different targeted approaches to specifically deliver sialylation inhibitors to the bone marrow microenvironment.
Collapse
Affiliation(s)
- Alessandro Natoni
- Apoptosis Research Centre, School of Medicine, National University of Ireland, Galway, Ireland
| | - Raghvendra Bohara
- Centre for Research in Medical Devices (CÚRAM), National University of Ireland, Galway, Ireland
| | - Abhay Pandit
- Centre for Research in Medical Devices (CÚRAM), National University of Ireland, Galway, Ireland
| | - Michael O'Dwyer
- Apoptosis Research Centre, School of Medicine, National University of Ireland, Galway, Ireland
| |
Collapse
|
145
|
Läubli H, Borsig L. Altered Cell Adhesion and Glycosylation Promote Cancer Immune Suppression and Metastasis. Front Immunol 2019; 10:2120. [PMID: 31552050 PMCID: PMC6743365 DOI: 10.3389/fimmu.2019.02120] [Citation(s) in RCA: 172] [Impact Index Per Article: 28.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2019] [Accepted: 08/23/2019] [Indexed: 12/14/2022] Open
Abstract
Cell-cell interactions and cell adhesion are key mediators of cancer progression and facilitate hallmarks of cancer including immune evasion and metastatic dissemination. Many cell adhesion molecules within the tumor microenvironment are changed and significant alterations of glycosylation are observed. These changes in cell adhesion molecules alter the ability of tumor cells to interact with other cells and extracellular matrix proteins. Three families of cell-cell interaction molecules selectins, Siglecs, and integrins have been associated with cancer progression in many pre-clinical studies, yet inhibition of cell adhesion as a therapeutic target is just beginning to be explored. We review how cell-cell interactions mediated by integrins and the glycan-binding receptors selectins and Siglec receptors support cancer progression. The discussion focuses on mechanisms during immune evasion and metastasis that can be therapeutically targeted by blocking these cell-cell interactions.
Collapse
Affiliation(s)
- Heinz Läubli
- Laboratory for Cancer Immunotherapy, Department of Biomedicine and Medical Oncology, Department of Internal Medicine, University Hospital, Basel, Switzerland
| | - Lubor Borsig
- Department of Physiology, University of Zurich, Zurich, Switzerland.,Comprehensive Cancer Center, Zurich, Switzerland
| |
Collapse
|
146
|
Kellokumpu S. Golgi pH, Ion and Redox Homeostasis: How Much Do They Really Matter? Front Cell Dev Biol 2019; 7:93. [PMID: 31263697 PMCID: PMC6584808 DOI: 10.3389/fcell.2019.00093] [Citation(s) in RCA: 76] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2019] [Accepted: 05/16/2019] [Indexed: 02/06/2023] Open
Abstract
Exocytic and endocytic compartments each have their own unique luminal ion and pH environment that is important for their normal functioning. A failure to maintain this environment - the loss of homeostasis - is not uncommon. In the worst case, all the main Golgi functions, including glycosylation, membrane trafficking and protein sorting, can be perturbed. Several factors contribute to Golgi homeostasis. These include not only ions such as H+, Ca2+, Mg2+, Mn2+, but also Golgi redox state and nitric oxide (NO) levels, both of which are dependent on the oxygen levels in the cells. Changes to any one of these factors have consequences on Golgi functions, the nature of which can be dissimilar or similar depending upon the defects themselves. For example, altered Golgi pH homeostasis gives rise to Cutis laxa disease, in which glycosylation and membrane trafficking are both affected, while altered Ca2+ homeostasis due to the mutated SCPA1 gene in Hailey-Hailey disease, perturbs various protein sorting, proteolytic cleavage and membrane trafficking events in the Golgi. This review gives an overview of the molecular machineries involved in the maintenance of Golgi ion, pH and redox homeostasis, followed by a discussion of the organelle dysfunction and disease that frequently result from their breakdown. Congenital disorders of glycosylation (CDGs) are discussed only when they contribute directly to Golgi pH, ion or redox homeostasis. Current evidence emphasizes that, rather than being mere supporting factors, Golgi pH, ion and redox homeostasis are in fact key players that orchestrate and maintain all Golgi functions.
Collapse
Affiliation(s)
- Sakari Kellokumpu
- Faculty of Biochemistry and Molecular Medicine, University of Oulu, Oulu, Finland
| |
Collapse
|
147
|
Structural basis for the delivery of activated sialic acid into Golgi for sialyation. Nat Struct Mol Biol 2019; 26:415-423. [PMID: 31133698 DOI: 10.1038/s41594-019-0225-y] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2018] [Accepted: 04/09/2019] [Indexed: 01/08/2023]
Abstract
The decoration of secretory glycoproteins and glycolipids with sialic acid is critical to many physiological and pathological processes. Sialyation is dependent on a continuous supply of sialic acid into Golgi organelles in the form of CMP-sialic acid. Translocation of CMP-sialic acid into Golgi is carried out by the CMP-sialic acid transporter (CST). Mutations in human CST are linked to glycosylation disorders, and CST is important for glycopathway engineering, as it is critical for sialyation efficiency of therapeutic glycoproteins. The mechanism of how CMP-sialic acid is recognized and translocated across Golgi membranes in exchange for CMP is poorly understood. Here we have determined the crystal structure of a Zea mays CST in complex with CMP. We conclude that the specificity of CST for CMP-sialic acid is established by the recognition of the nucleotide CMP to such an extent that they are mechanistically capable of both passive and coupled antiporter activity.
Collapse
|
148
|
Bornhöfft KF, Galuska SP. Glycans as Modulators for the Formation and Functional Properties of Neutrophil Extracellular Traps: Used by the Forces of Good and Evil. Front Immunol 2019; 10:959. [PMID: 31134066 PMCID: PMC6514094 DOI: 10.3389/fimmu.2019.00959] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2019] [Accepted: 04/15/2019] [Indexed: 12/13/2022] Open
Abstract
A very common mechanism to trap pathogens is the release of DNA. Like flies in a spider's web, pathogens are enclosed in a sticky chromatin meshwork. Interestingly, plants already use this mechanism to catch bacteria. In mammals, especially neutrophils release their DNA to prevent an invasion of bacteria. These neutrophil extracellular traps (NETs) are equipped with antimicrobial molecules, including, for instance, histones, antimicrobial peptides, lactoferrin, and neutrophil elastase. Thus, in a defined area, pathogens and toxic molecules are directly adjacent. However, several of these antimicrobial substances are also cytotoxic for endogenous cells. It is, therefore, not surprising that distinct control mechanisms exist to prevent an exaggerated NETosis. Nevertheless, despite these endogenous control instruments, an extraordinary NET release is characteristic for several pathologies. Consequently, NETs are a novel target for developing therapeutic strategies. In this review, we summarize the roles of glycans in the biology of NETs; on the one hand, we focus on the glycan-dependent strategies of endogenous cells to control NET formation or to inactivate its cytotoxic effects, and, on the other hand, the “sweet” tricks of pathogens to inhibit the release of NETs or to prevent NET-mediated killing mechanisms are examined. Understanding both, the forces of good and evil, allows the development of novel glycan-based approaches to combat the harmful side of NETs during distinct pathologies.
Collapse
Affiliation(s)
- Kim F Bornhöfft
- Institute of Reproductive Biology, Leibniz Institute for Farm Animal Biology, Dummerstorf, Germany
| | - Sebastian P Galuska
- Institute of Reproductive Biology, Leibniz Institute for Farm Animal Biology, Dummerstorf, Germany
| |
Collapse
|
149
|
Hassinen A, Khoder-Agha F, Khosrowabadi E, Mennerich D, Harrus D, Noel M, Dimova EY, Glumoff T, Harduin-Lepers A, Kietzmann T, Kellokumpu S. A Golgi-associated redox switch regulates catalytic activation and cooperative functioning of ST6Gal-I with B4GalT-I. Redox Biol 2019; 24:101182. [PMID: 30959459 PMCID: PMC6454061 DOI: 10.1016/j.redox.2019.101182] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2019] [Revised: 03/22/2019] [Accepted: 03/28/2019] [Indexed: 01/10/2023] Open
Abstract
Glycosylation, a common modification of cellular proteins and lipids, is often altered in diseases and pathophysiological states such as hypoxia, yet the underlying molecular causes remain poorly understood. By utilizing lectin microarray glycan profiling, Golgi pH and redox screens, we show here that hypoxia inhibits terminal sialylation of N- and O-linked glycans in a HIF- independent manner by lowering Golgi oxidative potential. This redox state change was accompanied by loss of two surface-exposed disulfide bonds in the catalytic domain of the α-2,6-sialyltransferase (ST6Gal-I) and its ability to functionally interact with B4GalT-I, an enzyme adding the preceding galactose to complex N-glycans. Mutagenesis of selected cysteine residues in ST6Gal-I mimicked these effects, and also rendered the enzyme inactive. Cells expressing the inactive mutant, but not those expressing the wild type ST6Gal-I, were able to proliferate and migrate normally, supporting the view that inactivation of the ST6Gal-I help cells to adapt to hypoxic environment. Structure comparisons revealed similar disulfide bonds also in ST3Gal-I, suggesting that this O-glycan and glycolipid modifying sialyltransferase is also sensitive to hypoxia and thereby contribute to attenuated sialylation of O-linked glycans in hypoxic cells. Collectively, these findings unveil a previously unknown redox switch in the Golgi apparatus that is responsible for the catalytic activation and cooperative functioning of ST6Gal-I with B4GalT-I.
Collapse
Affiliation(s)
- Antti Hassinen
- University of Oulu, Faculty of Biochemistry and Molecular Medicine, Oulu, Finland
| | - Fawzi Khoder-Agha
- University of Oulu, Faculty of Biochemistry and Molecular Medicine, Oulu, Finland
| | - Elham Khosrowabadi
- University of Oulu, Faculty of Biochemistry and Molecular Medicine, Oulu, Finland
| | - Daniela Mennerich
- University of Oulu, Faculty of Biochemistry and Molecular Medicine, Oulu, Finland
| | - Deborah Harrus
- University of Oulu, Faculty of Biochemistry and Molecular Medicine, Oulu, Finland
| | - Maxence Noel
- Université de Lille, CNRS, UMR 8576, UGSF - Unité de Glycobiologie Structurale et Fonctionnelle, F-59000 Lille, France
| | - Elitsa Y Dimova
- University of Oulu, Faculty of Biochemistry and Molecular Medicine, Oulu, Finland
| | - Tuomo Glumoff
- University of Oulu, Faculty of Biochemistry and Molecular Medicine, Oulu, Finland
| | - Anne Harduin-Lepers
- Université de Lille, CNRS, UMR 8576, UGSF - Unité de Glycobiologie Structurale et Fonctionnelle, F-59000 Lille, France
| | - Thomas Kietzmann
- University of Oulu, Faculty of Biochemistry and Molecular Medicine, Oulu, Finland
| | - Sakari Kellokumpu
- University of Oulu, Faculty of Biochemistry and Molecular Medicine, Oulu, Finland.
| |
Collapse
|
150
|
Patel A, Tiwari S, Jha PK. Molecular interaction between bi-antennary phenylboronic acid and sialic acid using density functional theory and multi-time scale trajectories. J Biomol Struct Dyn 2019; 38:1242-1247. [PMID: 30885094 DOI: 10.1080/07391102.2019.1596839] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Anjali Patel
- Department of Physics, Faculty of Science, The Maharaja Sayajirao University of Baroda, Vadodara, Gujarat, India
| | - Sanjay Tiwari
- Maliba Pharmacy College, UKA Tarsadia University, Surat, Gujarat, India
| | - Prafulla K Jha
- Department of Physics, Faculty of Science, The Maharaja Sayajirao University of Baroda, Vadodara, Gujarat, India
| |
Collapse
|