Glycosylation represents one of the most abundant posttranslational modification of proteins. Glycosylation products are diverse and are regulated by the cooperative action of various glycosyltransferases, glycosidases, substrates thereof: nucleoside sugars and their transporters, and chaperons. In this article, we focus on a glycosyltransferase, α1,6-fucosyltransferase (Fut8) and its product, the core fucose structure on N-glycans, and summarize the potential protective functions of this structure against emphysema and chronic obstructive pulmonary disease (COPD). Studies of FUT8 and its enzymatic product, core fucose, are becoming an emerging area of interest in various fields of research including inflammation, cancer and therapeutics. This article discusses what we can learn from studies of Fut8 and core fucose by using knockout mice or in vitro studies that were conducted by our group as well as other groups. We also include a discussion of the potential protective functions of the keratan sulfate (KS) disaccharide, namely L4, against emphysema and COPD as a glycomimetic. Glycomimetics using glycan analogs is one of the more promising therapeutics that compensate for the usual therapeutic strategy that involves targeting the genome and the proteome. These typical glycans using KS derivatives as glycomimetics, will likely become a clue to the development of novel and effective therapeutic strategies.

In the human body, there are several types of protein modifications that involve glycans being attached to glycoproteins, such as N-glycosylation, O-glycosylation, glycosylphosphatidylinositol (GPI)-anchor and glycosaminoglycan chains and others attaching to protein molecules [1]. These glycan modifications are regulated by the tissue- or cell type-dependent expression pattern of multiple glycosyltransferases, which are required for glycoproteins to express tissue- or cell type-dependent functions. Our group focused on glycosyltransferases that are associated with branching enzymes of N-glycans [2] and major enzymes including N-acetylglucosaminyltransferases referred to as GnT-III, GnT-IV, GnT-V, GnT-VI and α1,6-Fucosyltransferase (N-acetyl-β-d-glucosamine α1,6 fucosyltransferase, FUT8). They were purified to homogeneity, and GnT-IX (Vb) was identified by molecular cloning. Among them the cDNA of GnT-III, GnT-V and FUT8 were cloned and their genes identified in our group [3–9].

FUT8 attaches a fucose residue onto N-linked-type complex glycoproteins (N-glycans) [8], is expressed ubiquitously in mammalian tissues and is the sole enzyme responsible for generating a core fucose structure. The core fucose structure has a variety of functions that have been demonstrated in studies of Fut8 knockout mice (Fut8−/−). Fut8−/− mice showed semi-lethal phenotypes with severe growth retardation and emphysematous phenotypic changes [9], aberrant B cell development [10], defects in T cell receptor signaling [11–14], long term potentiation [15] and enhancement of neuroinflammation with schizophrenia phenotypes [16]. In human patients with congenital disorders of glycosylation (CDG) caused by FUT8 mutations, lung dysfunction was reported [17,18] as was observed in Fut8 knockout mice [9]. These findings facilitated the development of therapeutics using cancer immunotherapy and tumor biomarkers. An excellent article on FUT8 was recently published [19]. Typical target proteins of FUT8 are the immunoglobulin G (IgG)-B receptor [20], μ-heavy chain [21], IgG1 [22,23], TGF-βreceptor [9], epidermal growth factor receptor (EGFR) [24,25], α4β1 integrin/VCAM-1 [10], α3β1integrin [26,27], T cell receptors (TCRs) [13,28], E-cadherin [29–32], Dectin-1 [33] and program cell death 1 (PD-1) [34].

It is well known that the aberrant glycan modifications of proteins are strongly related to the onset and progression of certain diseases [35–38]. This suggests that targeting those aberrant glycans is a potentially useful strategy for developing novel and effective therapies [39]. In fact, most of the cancer biomarkers in use today are glycan-based molecules, examples include, CEA, CA19-9, CA15-3, CA-125, PSA and α-fetoprotein L3 [40,41]. These glycans are not just biomarkers but are functional molecules involved in tumorigenesis and the malignant properties in some cancers [38,42]. The attachment of a core fucose to specific glycoproteins are likely candidates for various cancer biomarkers in non-small cell lung cancer (NSCLC) [43–45], castration-resistant and aggressive prostate cancer [46–48], papillary cancer of the thyroid [49], colorectal cancer [32,50], pancreatic ductal adenocarcinoma [50–52], ovarian cancer [53], breast cancer [54], hepatocellular carcinoma [44,55–60].

Research to identify unique disease-related glycans are ongoing concerning many other diseases as well because they could be biomarkers and may be available for developing targeting therapies [60,61]. Concerning this, we extensively investigated emphysema because our research group was the first in the world to generate Fut8 knockout mice and we found severe emphysema and chronic obstructive pulmonary disease (COPD)-like phenotypes in these model animals [9,62–64].

COPD is the third most common cause of death in the world between 1990 and 2010 [65]. In this disease, the development of chronic bronchitis leads to bronchial obstruction and respiratory failure that are not fully reversible [66]. Most COPD cases are associated with emphysema and chronic bronchitis, which is exacerbation by viral or bacterial infections [67] and have an increased risk of primary lung cancer [68]. In any case, the major cause of COPD is exposure to cigarette smoke, secondhand smoke and air pollutions [69]. Importantly, cigarette smoke can strongly activate a variety of immune cells such as macrophages, neutrophils, T-lymphocytes, B-lymphocytes and dendritic cells (DCs) in the bronchi. Those activated immune cells secrete many inflammatory mediators and proteolytic enzymes, which further damage the bronchi and alveoli.

In the first half of this review, we summarize the protective role of Fut8, as reported in our studies using Fut8−/− mice. Additionally, we also summarize our recent data regarding the potential therapeutic effects of the keratan sulfate (KS) disaccharide against emphysema and COPD, and discuss its potential use in the treatment of COPD as a candidate glycomimetic in the second half of this report.

FUT8 generates a core fucose structure by transferring a fucose moiety from GDP-fucose to the innermost GlcNAc residue in N-glycans [70] (Figure 1A). Regarding the molecular structure of human FUT8, the putative catalytic domain is located approximately in the middle of its protein primary structure, and also FUT8 contains other two characteristic domains: An N-terminal α-helical coiled-coil domain and a C-terminal Src homology 3 (SH3) domain [71,72]. The existing of SH3 domain is characteristic for FUT8, but a recent study reported that the SH3 domain is required for its enzymatic activity and identified candidate binding molecules that bind to the SH3 domain [73]. Our research group had generated Fut8−/− mice and analyzed them in order to address the physiological functions of Fut8 as well as its product: the core fucose structure, in in vivo.

Involvement of Fut8 in emphysema and COPD.

Figure 1.
Involvement of Fut8 in emphysema and COPD.

(A) Catalytic activity of α1,6 fucosyltransferase (FUT8) for the generation of the core fucose structure on N-glycans. (B) A scheme for the potential protective function of the core fucose structure on the TGF-β receptor-II (TβRII). On lung epithelium cells, TβRII forms heterodimer complexes with the TGR-β receptor-I (TβRI). The low binding affinity of TGF-β1 for those receptors is due to the deletion of the core fucose structure on TβRII which leads to a low level of phosphorylation of Smad2 and the up-regulation of Mmp9 and Mmp12. The strong expression of these proteolytic enzymes enhances the degradation of connective tissue compartments of the lungs, resulting in the development of severe emphysema-like phenotypes in Fut8−/− mice. P: phosphorylation of TGF-β receptors and Smad2.

Figure 1.
Involvement of Fut8 in emphysema and COPD.

(A) Catalytic activity of α1,6 fucosyltransferase (FUT8) for the generation of the core fucose structure on N-glycans. (B) A scheme for the potential protective function of the core fucose structure on the TGF-β receptor-II (TβRII). On lung epithelium cells, TβRII forms heterodimer complexes with the TGR-β receptor-I (TβRI). The low binding affinity of TGF-β1 for those receptors is due to the deletion of the core fucose structure on TβRII which leads to a low level of phosphorylation of Smad2 and the up-regulation of Mmp9 and Mmp12. The strong expression of these proteolytic enzymes enhances the degradation of connective tissue compartments of the lungs, resulting in the development of severe emphysema-like phenotypes in Fut8−/− mice. P: phosphorylation of TGF-β receptors and Smad2.

Close modal

Embryogenesis of Fut8−/− mice does not appear to be impaired, hence they are born with an apparent normal pregnancy period. However, most newborn Fut8−/− mice died within a few days after birth. The survivors showed severe growth retardation. To determine the reason for this, we examined Fut8−/− mice and identified severe emphysema phenotypes that include the attenuation of the inhalation and exhalation activity, as well as the structural collapse of alveoli of the lung [9]. In a further study, we demonstrated that an impairment in TGF-β receptor-II/Smad signaling was one of the molecular mechanisms by which emphysema was spontaneously induced in Fut8−/− mice. TGF-β receptor-II is a transmembrane kinase and forms heteromeric complexes with the TGF-β receptor-I for ligand binding. Murine TGF-β receptor-II contains three N-linked glycans on its extracellular region, and a lack of a core fucose structure results in a lower binding affinity of the ligand: TGF-β1, to TGF-β receptor-II in the lungs of Fut8−/− mice. This weak ligand stimulation results in low levels of the phosphorylation of Smad2 and the up-regulation of the matrix metalloprotease (Mmp) family genes such as Mmp1, Mmp9 and Mmp12. The strong expression of these proteolytic enzymes effectively breaks down the connective tissue components of the bronchi, which induces severe emphysema phenotypes in the lungs of Fut8−/− mice (Figure 1B). These data demonstrate the protective roles of Fut8 and the core fucose structure on TGF-β receptor-II against emphysema and COPD. A study using mutant mice with a lung epithelium-specific deletion of the TGF-β receptor-II also demonstrated the protective roles of the TGF-β receptor-II/Smad signaling against emphysema [74].

In COPD patients, decreased levels of expression of TGF-β1 in the bronchiolar epithelium and alveolar macrophages and TGF-β receptor-II in bronchial glands as well as TGF-β receptor-I were observed [75–77]. While these studies indicate that the down-regulation of TGF-β receptor/Smad signaling is associated with the disease state in humans as well, these studies did not focus on the FUT8 gene and the core fucose structure. Notably, a Thr267Lys polymorphism in human FUT8 gene was also reported to be associated with the development of emphysema in humans. A significantly increased frequency of Thr267Lys polymorphisms in FUT8 in subjects with emphysema compared with subjects without emphysema was found (subjects with emphysema vs. subjects without emphysema = 18.7% vs. 14.2%) [78]. The Thr267Lys polymorphism is located in the proline-rich region of FUT8 and is conserved among glycosyltransferases [79,80], indicating that this amino acid change has an impact on its catalytic activity. Other recent studies reported that patients with a FUT8-CDG were found to show respiratory abnormalities [17,18]. In one allele, a substitution in the coding region of FUT8 (c.943C > T) contains a stop codon (p.Arg315*). As a result, the catalytic activity of FUT8 in this individual was completely deficient [18]. There have not been any studies showing the direct involvement of the core fucose structure on TGF-β receptor in the disease state of patients of emphysema and COPD. However, those studies strongly suggest FUT8 and/or core fucose structure in humans have potential protective roles in humans as well as in experimental animals.

Cigarette smoke is the major cause of emphysema and COPD, therefore, research to address how cigarette smoke is associated with the onset and progression of this disease is a particularly important subject. Interestingly, it was reported that cigarette smoke decreases the level of expression of Fut8 gene and its enzymatic activity in the lungs of mice that were exposed to cigarette smoke [64]. Depending on this Fut8 suppression, the lasting up-regulation of Mmp9 was observed in the lungs during the period of exposure to cigarette smoke, which is consistent with the results of a Fut8−/− mice study as described above. This indicates the potential of using Fut8 mutant mice as an emphysema model. Importantly, the emphysema-like phenotypes induced by exposure to cigarette smoke actually appeared in Fut8 heterozygous knockout (Fut8+/−) mice earlier than in wild type mice [64].

Instead of a cigarette smoke-exposure model, an elastase-induced model is also commonly used in the research field of emphysema and COPD [79]. Elastase is a proteolytic enzyme that digests elastin that is present in the extracellular matrix [81,82]. Thus, the administration of elastase to the mice by intratracheal spray induces the breakdown of connective tissue components in the lungs, which results in the development of emphysema-like phenotypes such as bronchial obstruction, respiratory failure and bronchitis. In addition to the above conventional model, we developed other models for exacerbation induced by viral and bacterial infections. During the pathogenesis and clinical course of COPD, a vicious circle of infection and inflammation is thought to lead to the exacerbation of the disease [69]. Therefore, our research group reported that the combined administration of a one shot administration of LPS and elastase to the murine bronchi induced more severe emphysema phenotypes [62] (Figure 2). Based on this and the protective roles of Fut8 gene, as outlined above, the intratracheal administration of elastase combined with LPS to the WT or Fut8+/- mice would be predicted to be an exacerbation model for emphysema and COPD (Figure 2). This model would be desirable in terms of developing effective therapies for the treatment of serious cases of emphysema and COPD patients with bacterial and viral infections.

Three types of mice model were used in this study.

Figure 2.
Three types of mice model were used in this study.

Model A: Cigarette smoke model. Exposure of wild type mice (WT) to cigarette smoke induces COPD-like phenotypes within approximately 6 months. When heterozygous Fut8 KO (Fut8+/−) mice are exposed to cigarette smoke, however, emphysematous changes are induced within 3 months indicating these mice are sensitive to cigarette smoke [62]. Model B: The conventional method for producing a porcine pancreatic elastase-induced emphysema model. When given a single dose of an elastase treatment, emphysema was produced after 3 weeks. Model C: Exacerbation model. Heterozygous Fut8 knockout (Fut8+/) mice or porcine pancreatic elastase-induced model mice plus the administration of one dose of an intratracheal administration of LPS. To the mice with emphysema caused by the cigarette exposure for 3 month or by the elastase treatment for 3 weeks, one shot LPS was administered intravenously.

Figure 2.
Three types of mice model were used in this study.

Model A: Cigarette smoke model. Exposure of wild type mice (WT) to cigarette smoke induces COPD-like phenotypes within approximately 6 months. When heterozygous Fut8 KO (Fut8+/−) mice are exposed to cigarette smoke, however, emphysematous changes are induced within 3 months indicating these mice are sensitive to cigarette smoke [62]. Model B: The conventional method for producing a porcine pancreatic elastase-induced emphysema model. When given a single dose of an elastase treatment, emphysema was produced after 3 weeks. Model C: Exacerbation model. Heterozygous Fut8 knockout (Fut8+/) mice or porcine pancreatic elastase-induced model mice plus the administration of one dose of an intratracheal administration of LPS. To the mice with emphysema caused by the cigarette exposure for 3 month or by the elastase treatment for 3 weeks, one shot LPS was administered intravenously.

Close modal

Unfortunately, there are still significant hurdles to the develop an effective therapy for emphysema and COPD targeting FUT8 and/or core fucose structure on TGF-β receptor-II. Thus, we studied other glycan candidates and identified KS proteoglycan because the expression levels of KS were significantly decreased in the lungs of mice that had been exposed to cigarette smoke [83]. This indicates that KS expression is down-regulated by cigarette smoke and is somehow involved in the onset and progression of emphysema. Based on this, our hypothesis was that KS exerted protective functions against emphysema and COPD and the supplementation of KS into the lungs would mitigate the clinical phenotypes of the disease.

The KS proteoglycan is one of the proteoglycans that was identified in corneal extracts in 1939 [26]. The KS is made up of a linear polymer of repeating N-acetyllactosamine (LacNAc) unit which is composed of β1.4 linked to a GlcNAc and is sulfated at different positions [80]. KS is now classified into three types, corneal KS-I, skeletal KS-II and brain KS-III. KS-I is attached to N-linked glycans, whereas KS-II and KS-III are attached to mucin type O-linked glycan chains and O-mannose type glycans, respectively [84,85]. The large and diverse structure of glycans of KS seemed to be unsuitable, hence our particular focus was on the simpler di-sulfated KS disaccharide called L4 which consists of a galactose-6-sulfate unit and a β1-4 linked GlcNAc-6-sulfate unit (Figure 3A). Xu et al. [86] reported that the treatment of model mice of autoimmune disease with L4 modulates Interleukin 12 (IL-12) production by macrophages. Shirato et al. [87] suggested that L4 specifically blocks the interaction of flagellin with the toll-like receptor 5, and subsequently suppresses IL-8 production in primary normal human bronchial epithelial cells . Taking these collective issues into account, L4 represents a potential molecule that could be used for the prevention and treatment of airway inflammatory responses to bacterial infections including COVID-19 [39], which play a critical role in the exacerbation of COPD [87]. L4 was obtained from KS of shark fin cartilage after a keratanase II treatment [86] and provided from the Seikagaku Corporation which is a collaborator of our group, and we identified it as a potential functional ligand of Langerin, a C-type lectin receptor that is expressed on DCs [88].

Keratan sulfate in emphysema and COPD.

Figure 3.
Keratan sulfate in emphysema and COPD.

(A) The structure of keratan sulfate, KS-1, L4 consists of galactose-6-sulfate unit and β1-4 linked to GlcNAc-6-sulfate. (B) A scheme for the potential protective functions of the keratan sulfate disaccharide L4 and derivatives thereof: triangle-L4 and pendant-L4 against emphysema and COPD. L4 shows anti-proteolytic activity caused by the inhibition of Mmp-2 and Mmp-9 activities. Triangle-L4 and pendant-L4 bind to Langerin more strongly than the L4 monomer and may result in an enhanced down-regulation of TNFα in dendritic cells. These two functions of L4 shown by squares contribute to the mitigation of emphysema phenotypes in the elastase-induced mouse model. The suppressed signal transduction through Langerin is not currently understood. Langerin is required for Birbeck granule formation. 6S: the sulfate group on carbon number 6.

Figure 3.
Keratan sulfate in emphysema and COPD.

(A) The structure of keratan sulfate, KS-1, L4 consists of galactose-6-sulfate unit and β1-4 linked to GlcNAc-6-sulfate. (B) A scheme for the potential protective functions of the keratan sulfate disaccharide L4 and derivatives thereof: triangle-L4 and pendant-L4 against emphysema and COPD. L4 shows anti-proteolytic activity caused by the inhibition of Mmp-2 and Mmp-9 activities. Triangle-L4 and pendant-L4 bind to Langerin more strongly than the L4 monomer and may result in an enhanced down-regulation of TNFα in dendritic cells. These two functions of L4 shown by squares contribute to the mitigation of emphysema phenotypes in the elastase-induced mouse model. The suppressed signal transduction through Langerin is not currently understood. Langerin is required for Birbeck granule formation. 6S: the sulfate group on carbon number 6.

Close modal

Potential protective functions of keratan sulfate disaccharide L4 through Langerin in emphysema

Langerin, which was identified as a C-type lectin receptor in 2000 [89], is a type II membrane protein and has one C-terminal carbohydrate recognition domain (CRD) in its extracellular region. Physiologically, the expression of Langerin is selective and highly enriched in Langerhans cells (LCs), a subset of DCs that are localized in the dermis. DCs, including LCs, are antigen-presenting cells that function to initiate specific T cell immunity, and LCs are particularly characterized by the presence of a unique pentalamellar cytoplasmic organelle, the Birbeck granule, that mediates the intracellular transport of antigens. The precise roles of Birbeck granule have not been demonstrated, but importantly, Langerin is required for the formation of Birbeck granules and antigen presentation for nonpeptide antigens of Mycobacterium leprae in LCs [90]. However, the issue of how interactions between Langerin and its endogenous and exogenous sugar ligands modulate DC and LC functions and immune responses currently remain unclear.

Several studies have identified various candidates for sugar ligands of Langerin. Langerin recognizes a broad range of glycans that contain mannose, fucose or sulfated sugars, such as oligomannose type N-glycans, the blood group B epitope and glycosaminoglycans. Carbohydrates on gp120 of the human immunodeficiency virus (HIV) and the hemagglutinin glycoprotein of influenza A virus that were also reported to be recognized by Langerin [91,92], and the Langerin-mediated capture of those glycan ligands was proposed to function as a barrier against HIV and influenza A virus [91]. On the other hands, Langerin also binds the teichoic acid of staphylococcus aureus to enhance the inflammatory process [92] and heparin production. The binding of Langerin to glycosaminoglycans, especially heparin, has been extensively studied and the high-affinity binding of heparin to Langerin (dissociation constants ∼2.4 nM) as judged by surface plasmon resonance was reported [93].

Among many reports regarding the sugar-binding properties of Langerin, a comprehensive glycoconjugate microarray revealed that galactose-6-sulfate, a component of KS, was a unique sugar ligand of Langerin [94], suggesting that Langerin-KS interactions play important roles in the development of immunity. Interestingly, we previously characterized the interaction between L4 and recombinant trimeric Langerin in in vitro binding assays. Competitive enzyme-linked immunosorbent assay revealed that L4 binds to Langerin that was expressed and purified from Escherichia coli with an IC50 value of 3.5 mM. This high IC50 value indicates that the binding between Langerin and the L4 monomer is weak, as is the case of many lectin–sugar interactions, and we expected that the chemical oligomerization of L4 would overcome this weak affinity and lead to the development of high-affinity L4-related Langerin ligands for further functional analysis and clinical applications. Therefore, we synthesized two L4 oligomers: triangle-L4 (L4 trimer) and pendant-L4 (L4 polymer). As expected, the triangle-L4 and the pendant-L4 bound more strongly to recombinant Langerin than the L4 monomer. The IC50 value for pendant-L4 was 2.1 nM [88], which is a sufficiently low concentration to allow further in vivo analyses to be conducted.

We recently demonstrated that the administration of L4 mitigated the disease state of elastase-induced emphysema and the emphysema in the LPS-induced COPD exacerbation model [62].

In these studies, the intratracheal administration of L4 to the mice decreased the number of infiltrating immune cells, namely, neutrophils and macrophages and the amount of an inflammatory cytokine: tumor necrosis factor α (TNFα) in the bronchoalveolar lavage fluid (BALF). In addition, the activity of Mmp-9 and Mmp-12 in BALF of elastase- or LPS-treated mice was also attenuated by the administration of L4. These anti-inflammatory and anti-proteolytic effects of L4 attenuated severe bronchitis and the pathological collapse of alveoli (Figure 3B). These current pre-clinical data strongly indicate that L4 could serve as a drug for the treatment of emphysema and COPD. However, further investigations will be needed to determine whether Langerin is indeed a functional receptor of L4 in these mouse models as well as in humans.

Patients with COPD are at increased risk for the development of primary lung cancer [95–97]. Although the down-regulation of FUT8 is involved in the disease state of emphysema and COPD as described above, the overexpression of FUT8 and/or core fucose-containing glycans was reported in lung cancer and prostate cancer [45,98]. Moreover, a correlation of the expression of core fucose structure with poor survival outcome in some cancer has been reported [45,52,99]. Importantly, FUT8 was also identified as a driver of metastasis of melanoma cells [98].

Epithelial–mesenchymal transition (EMT) and its reversion process MET (mesenchymal–epithelial transition) are important in embryonal development, wound healing and cancer. The overexpression of FUT8 gene in breast cancer cells drives strong TGF-β signals and EMT due to excess levels of core fucose structures on TGF-β receptors in breast cancer cells [54]. In relation to EMT, our group summarized the role of glycosyltransferases involved in N-glycan branching and emphasized the roles of above glycosyltransferases including FUT8 in EMT and MET as transition state as well as biomarkers [99]. Moreover, the overexpression of FUT8 in cancer-associated fibroblasts (CAFs) can promote the malignancy of NSCLC through the activation of EGFR signaling in CAFs [100]. A knockdown study of FUT8 also revealed that weakened EGFR signaling in CAFs attenuated the malignancy of NSCLC [100].

Core fucose unit of N-linked glycan has a unique function in modulating anti-cancer immune responses, for instance, the core fucose structure of N-linked glycan on the TCR is required for the activation of TCR signaling [11–13]. Based on these reports, the inhibition of the core fucose structure considered to be one of the novel therapeutic strategies for cancer patients, but these trials are still in progress. TCR is required for the activation of TCR signaling, and the core fucose structure of PD-1 is essential for cancer immunotherapy [34]. Based on these reports, the inhibition of the core fucose structure is considered to be one of the novel therapeutic strategies for cancer patients [101], but these trials are still in progress.

Glycomimetic using two C-type lectin receptors on dendritic cells as targets

Rational of Langerin as a glycomimetic target

DCs contain two major C-type lectin receptors, Langerin and ICAM-3 grabbing non-integrin (DC-SIGN) [102]. On the cell surface of DCs, both receptors can recognize pathogen- and danger-associated molecular patterns (PAMS and DMPS, respectively) [103] and function to regulate immune responses. However, if some receptor(s) are hijacked by viruses or bacteria, these functions are blocked. Therefore, the development of a specific ligand for binding to a receptor that would protect against this hijacking. This would be a candidate for a glycomimetic drug [39]. In HIV infections, DC-SIGN and Langerin have opposing effects, and DC-SIGN acts on the facilitating of viral survival transmission and infection, whereas Langerin acts on the promotion of viral uptake and degradation [104]. Wamhoff et al. [105] analyzed 275 sugar compounds using the bacterially produced and fluorescence-labeled monomeric carbohydrate recognition domain of DC-SIGN and Langerin, and found that the carbohydrate specificity for Langerin were a terminal mannose, a terminal N-acetylglucosamine and a 6-sulfogalactose residue. Portolabo et al. reported on a specific inhibitor against DC-SIGN, and Wen et al. [106] also reported on a highly specific inhibitor against DC-SIGN with a polyproline tetra helix macrocyte scaffold. Additionally, Pottelverge et al. [107] reported that there are two segregated subsets of tissue-resident pulmonary myeloid DCs and these cells were identified in subtle cell suspensions by flowcytometry. In those patients with COPD, Langerhans-type DCs were accumulated selectively in small airways. Therefore, as a target for COPD, Langerin would play a key role. In this respect, KS derivatives are specific ligands for Langerin and protect hijacking by viruses or bacteria as shown in Figure 4.

Proposed interaction between glycan-based PAMP and DAMP (virus and/or bacteria) and C-type lectins, Langerin and DC-SIGN.
Figure 4.
Proposed interaction between glycan-based PAMP and DAMP (virus and/or bacteria) and C-type lectins, Langerin and DC-SIGN.

PAMP and DAMP can bind to DC-SIGN and Langerin but L4 binds specifically to Langerin and blocks hijacking by PAMP and DAMP thus allowing Langerin to remain active in endocytosis and degradation of the pathogens. The structure information of DC-SIGN is cited from the Uniprot database.

Figure 4.
Proposed interaction between glycan-based PAMP and DAMP (virus and/or bacteria) and C-type lectins, Langerin and DC-SIGN.

PAMP and DAMP can bind to DC-SIGN and Langerin but L4 binds specifically to Langerin and blocks hijacking by PAMP and DAMP thus allowing Langerin to remain active in endocytosis and degradation of the pathogens. The structure information of DC-SIGN is cited from the Uniprot database.

Close modal
  • The molecular mechanisms responsible for the onset and progression of emphysema and COPD have not been completely revealed, and there is still no fundamental therapy for curing these diseases. This has already become one of the crucial issues for world health.

  • From the glycoscience point of view, the core fucose structure and KS are prospective targets for novel and effective drug discovery. It is particularly noteworthy that the KS disaccharide L4: a specific glycan ligand to Langerin, can be administered through the trachea as well as intravenously, and the protective functions against emphysema and COPD have been demonstrated in mouse models.

  • Although studies are needed to be continued to clarify the role of Langerin as well and to verify the anti-inflammatory effects and safety of L4 more substantially, we believe that L4 has the potential to be developed as a breakthrough glycan-based drug and vaccine for the subjects who are suffering from inflammatory diseases including COPD and cancer in near future.

The authors declare that there are no competing interests associated with the manuscript.

This work was supported for N.T. by Japan Agency for Medical Research and Development (AMED, 20ae0101070h0005) and Grants-in-Aid for Scientific Research (KAKENHI KIBAN C, 18K06914), and was partially supported for Y.O. by Grand-in-Aid for Scientific Research (KAKENHI KIBAN C, 20K07629).

Y.O. drafted this article. Y.H. and N.T. reviewed and revised. All of the authors have read and agree with the submission of this article.

N.T. acknowledges many colleagues who used to work with us at Osaka University Medical School, Osaka University, RIKEN and Osaka International Cancer Institute. We wish to thank Dr. Milton Feather for his English editing.

BALF

bronchoalveolar lavage fluid

CAFs

cancer-associated fibroblasts

CDG

congenital disorders of glycosylation

COPD

chronic obstructive pulmonary disease

DCs

dendritic cells

EGFR

epidermal growth factor receptor

EMT

epithelial-mesenchymal transition

HIV

human immunodeficiency virus

IgG

immunoglobulin G

KS

keratan sulfate

LCs

Langerhans cells

NSCLC

non-small cell lung cancer

PD-1

program cell death 1

SH3

Src homology 3

TCR

T cell receptor

1
Varki
,
A.
(
2017
)
Biological roles of glycans
.
Glycobiology
27
,
3
49
2
Stanley
,
P.
,
Taniguchi
,
N.
and
Aebi
,
M
. (
2015
) N-Glycans. In
Essentials of Glycobiology
, 3rd edn (
Varki
,
A
.,
Cummings
,
R.D.
,
Esko
,
J.D.
,
Stanley
,
P.
,
Hart
,
G.W.
,
Aebi
,
M.
et al
, eds),
Cold Spring Harbor Laboratory Press
,
Cold Spring Harbor, NY
3
Taniguchi
,
N.
,
Miyoshi
,
E.
,
Jianguo
,
G.
,
Honke
,
K.
and
Matsumoto
,
A.
(
2006
)
Decoding sugar functions by identifying target glycoproteins
.
Curr. Opin. Struct. Biol.
16
,
561
566
4
Taniguchi
,
N.
and
Korekane
,
H.
(
2011
)
Branched N-glycans and their implications for cell adhesion, signaling and clinical applications for cancer biomarkers and in therapeutics
.
BMB Rep.
44
,
772
781
5
Taniguchi
,
N.
and
Kizuka
,
Y.
(
2015
)
Glycans and cancer: role of N-glycans in cancer biomarker, progression and metastasis, and therapeutics
.
Adv. Cancer Res.
126
,
11
51
6
Uozumi
,
N.
,
Yanagidani
,
S.
,
Miyoshi
,
E.
,
Ihara
,
Y.
,
Sakuma
,
T.
,
Gao
,
C.-X.
et al (
1996
)
Purification and cDNA cloning of porcine brain GDP-L-Fuc:N-acetyl-β-D-glucosaminide α1→6fucosyltransferase
.
J. Biol. Chem.
271
,
27810
27817
7
Yanagidani
,
S.
,
Uozumi
,
N.
,
Ihara
,
Y.
,
Miyoshi
,
E.
,
Yamaguchi
,
N.
and
Taniguchi
,
N.
(
1997
)
Purification and cDNA cloning of GDP-L-Fuc:N-acetyl-β-D-glucosaminide:α1-6 fucosyltransferase (α1-6 FucT) from human gastric cancer MKN45 cells
.
J. Biochem.
121
,
626
632
8
Brockhausen
,
I.
,
Carver
,
J.P.
and
Schachter
,
H.
(
1988
)
Control of glycoprotein synthesis. The use of oligosaccharide substrates and HPLC to study the sequential pathway for N-acetylglucosaminyltransferases I, II, III, IV, V, and VI in the biosynthesis of highly branched N-glycans by hen oviduct membranes
.
Biochem. Cell Biol.
66
,
1134
1151
9
Wang
,
X.
,
Inoue
,
S.
,
Gu
,
J.
,
Miyoshi
,
E.
,
Noda
,
K.
,
Li
,
W.
et al (
2005
)
Dysregulation of TGF-beta1 receptor activation leads to abnormal lung development and emphysema-like phenotype in core fucose-deficient mice
.
Proc. Natl Acad. Sci. U.S.A.
102
,
15791
15796
10
Li
,
W.
,
Nakagawa
,
T.
,
Koyama
,
N.
,
Wang
,
X.
,
Jin
,
J.
,
Mizuno-Horikawa
,
Y.
et al (
2006
)
Down-regulation of trypsinogen expression is associated with growth retardation in alpha1,6-fucosyltransferase-deficient mice: attenuation of proteinase-activated receptor 2 activity
.
Glycobiology
16
,
1007
1019
11
Fujii
,
H.
,
Shinzaki
,
S.
,
Iijima
,
H.
,
Wakamatsu
,
K.
,
Iwamoto
,
C.
,
Sobajima
,
T.
et al (
2016
)
Core fucosylation on T cells, required for activation of T-cell receptor signaling and induction of colitis in mice, is increased in patients with inflammatory bowel disease
.
Gastroenterology
150
,
1620
1632
12
Liang
,
W.
,
Mao
,
S.
,
Li
,
M.
,
Zhang
,
N.
,
Sun
,
S.
,
Fang
,
H.
et al (
2019
)
Ablation of core fucosylation attenuates the signal transduction via T cell receptor to suppress the T cell development
.
Mol. Immunol.
112
,
312
321
13
Liang
,
W.
,
Mao
,
S.
,
Sun
,
S.
,
Li
,
M.
,
Li
,
Z.
,
Yu
,
R.
et al (
2018
)
Core fucosylation of the T cell receptor is required for T cell activation
.
Front. Immunol.
9
,
78
14
Dias
,
A.M.
,
Correia
,
A.
,
Pereira
,
M.S.
,
Almeida
,
C.R.
,
Alves
,
I.
,
Pinto
,
V.
et al (
2018
)
Metabolic control of T cell immune response through glycans in inflammatory bowel disease
.
Proc. Natl Acad. Sci. U.S.A.
115
,
E4651
E4660
15
Gu
,
W.
,
Fukuda
,
T.
,
Isaji
,
T.
,
Hang
,
Q.
,
Lee
,
H.
,
Sakai
,
S.
et al (
2015
)
Loss of α1,6-fucosyltransferase decreases hippocampal long term potentiation: implications for core fucosylation in the regulation of AMPA receptor heteromerization and cellular signaling
.
J. Biol. Chem.
290
,
17566
17575
16
Lu
,
X.
,
Zhang
,
D.
,
Shoji
,
H.
,
Duan
,
C.
,
Zhang
,
G.
,
Isaji
,
T.
et al (
2019
)
Deficiency of α1,6-fucosyltransferase promotes neuroinflammation by increasing the sensitivity of glial cells to inflammatory mediators
.
Biochim. Biophys. Acta
1863
,
598
608
17
Ng
,
B.G.
,
Dastsooz
,
H.
,
Silawi
,
M.
,
Habibzadeh
,
P.
,
Jahan
,
S.B.
,
Fard
,
M.A.F.
et al (
2020
)
Expanding the molecular and clinical phenotypes of FUT8-CDG
.
J. Inherit. Metab Dis.
43
,
871
879
18
Ng
,
B.G.
,
Xu
,
G.
,
Chandy
,
N.
,
Steyermark
,
J.
,
Shinde
,
D.N.
,
Radtke
,
K.
et al (
2018
)
Biallelic mutations in FUT8 cause a congenital disorder of glycosylation with defective fucosylation
.
Am. J. Hum. Genet.
102
,
188
195
19
Boruah
,
B.M.
,
Kadirvelraj
,
R.
,
Liu
,
L.
,
Ramiah
,
A.
,
Li
,
C.
,
Zong
,
G.
et al (
2020
)
Characterizing human α-1,6-fucosyltransferase (FUT8) substrate specificity and structural similarities with related fucosyltransferases
.
J. Biol. Chem.
295
,
17027
17045
20
Li
,
W.
,
Yu
,
R.
,
Ma
,
B.
,
Yang
,
Y.
,
Jiao
,
X.
,
Liu
,
Y.
et al (
2015
)
Core fucosylation of IgG B cell receptor is required for antigen recognition and antibody production
.
J. Immunol.
194
,
2596
2606
21
Li
,
W.
,
Liu
,
Q.
,
Pang
,
Y.
,
Jin
,
J.
,
Wang
,
H.
,
Cao
,
H.
et al (
2012
)
Core fucosylation of μ heavy chains regulates assembly and intracellular signaling of precursor B cell receptors
.
J. Biol. Chem.
287
,
2500
2508
22
Shields
,
R.L.
,
Lai
,
J.
,
Keck
,
R.
,
O'Connell
,
L.Y.
,
Hong
,
K.
,
Meng
,
Y.G.
et al (
2002
)
Lack of fucose on human IgG1 N-linked oligosaccharide improves binding to human Fcgamma RIII and antibody-dependent cellular toxicity
.
J. Biol. Chem.
277
,
26733
26740
23
Shinkawa
,
T.
,
Nakamura
,
K.
,
Yamane
,
N.
,
Shoji-Hosaka
,
E.
,
Kanda
,
Y.
,
Sakurada
,
M.
et al (
2003
)
The absence of fucose but not the presence of galactose or bisecting N-acetylglucosamine of human IgG1 complex-type oligosaccharides shows the critical role of enhancing antibody-dependent cellular cytotoxicity
.
J. Biol. Chem.
278
,
3466
3473
24
Wang
,
X.
,
Gu
,
J.
,
Ihara
,
H.
,
Miyoshi
,
E.
,
Honke
,
K.
and
Taniguchi
,
N.
(
2006
)
Core fucosylation regulates epidermal growth factor receptor-mediated intracellular signaling
.
J. Biol. Chem.
281
,
2572
2577
25
Matsumoto
,
K.
,
Yokote
,
H.
,
Arao
,
T.
,
Maegawa
,
M.
,
Tanaka
,
K.
,
Fujita
,
Y.
et al (
2008
)
N-Glycan fucosylation of epidermal growth factor receptor modulates receptor activity and sensitivity to epidermal growth factor receptor tyrosine kinase inhibitor
.
Cancer Sci.
99
,
1611
1617
26
M
,
S.U.Z.U.K.I.
(
1941
)
BIOCHEMICAL STUDIES ON CARBOHYDRATES: LX. Anticoagulant from “Hunori” (gloiopeltis furcata): preliminary report
.
J. Biochem.
33
,
215
218
27
Zhao
,
Y.
,
Sato
,
Y.
,
Isaji
,
T.
,
Fukuda
,
T.
,
Matsumoto
,
A.
,
Miyoshi
,
E.
et al (
2008
)
Branched N-glycans regulate the biological functions of integrins and cadherins
.
FEBS J.
275
,
1939
1948
28
Takahashi
,
M.
,
Kizuka
,
Y.
,
Ohtsubo
,
K.
,
Gu
,
J.
and
Taniguchi
,
N.
(
2016
)
Disease-associated glycans on cell surface proteins
.
Mol. Asp. Med.
51
,
56
70
29
Pinho
,
S.S.
,
Figueiredo
,
J.
,
Cabral
,
J.
,
Carvalho
,
S.
,
Dourado
,
J.
,
Magalhães
,
A.
et al (
2013
)
E-cadherin and adherens-junctions stability in gastric carcinoma: functional implications of glycosyltransferases involving N-glycan branching biosynthesis, N-acetylglucosaminyltransferases III and V
.
Biochim. Biophys. Acta
1830
,
2690
2700
30
Yoshimura
,
M.
,
Nishikawa
,
A.
,
Ihara
,
Y.
,
Taniguchi
,
S.
and
Taniguchi
,
N.
(
1995
)
Suppression of lung metastasis of B16 mouse melanoma by N-acetylglucosaminyltransferase III gene transfection
.
Proc. Natl Acad. Sci. U.S.A.
92
,
8754
8758
31
Kitada
,
T.
,
Miyoshi
,
E.
,
Noda
,
K.
,
Higashiyama
,
S.
,
Ihara
,
H.
,
Matsuura
,
N.
et al (
2001
)
The addition of bisecting N-acetylglucosamine residues to E-cadherin down-regulates the tyrosine phosphorylation of beta-catenin
.
J. Biol. Chem.
276
,
475
480
32
Osumi
,
D.
,
Takahashi
,
M.
,
Miyoshi
,
E.
,
Yokoe
,
S.
,
Lee
,
S.H.
,
Noda
,
K.
et al (
2009
)
Core fucosylation of E-cadherin enhances cell-cell adhesion in human colon carcinoma WiDr cells
.
Cancer Sci.
100
,
888
895
33
Manabe
,
Y.
,
Marchetti
,
R.
,
Takakura
,
Y.
,
Nagasaki
,
M.
,
Nihei
,
W.
,
Takebe
,
T.
et al (
2019
)
The core fucose on an IgG antibody is an endogenous ligand of dectin-1
.
Angew. Chem. Int. Ed. Engl.
58
,
18697
18702
34
Okada
,
M.
,
Chikuma
,
S.
,
Kondo
,
T.
,
Hibino
,
S.
,
Machiyama
,
H.
,
Yokosuka
,
T.
et al (
2017
)
Blockage of core fucosylation reduces cell-surface expression of PD-1 and promotes anti-tumor immune responses of T cells
.
Cell Rep.
20
,
1017
1028
35
Freeze
,
H. H.
,
Kinoshita
,
T.
, and
Varki
,
A
. (
2015
)
Glycans in acquired human diseases
. In
Essentials of Glycobiology
, 3rd edn (
Varki
,
A.
,
Cummings
,
R.D.
,
Esko
,
J.D.
,
Stanley
,
P.
,
Hart
,
G.W.
,
Aebi
,
M.
et al
, eds),
Cold Spring Harbor Laboratory Press
,
Cold Spring Harbor, NY
36
Freeze
,
H.H.
(
2013
)
Understanding human glycosylation disorders: biochemistry leads the charge
.
J Biol Chem.
288
,
6936
6945
37
Mereiter
,
S.
,
Balmaña
,
M.
,
Campos
,
D.
,
Gomes
,
J.
and
Reis
,
C.A.
(
2019
)
Glycosylation in the era of cancer-targeted therapy: where are we heading?
Cancer Cell
36
,
6
16
38
Hakomori
,
S.
(
2002
)
Glycosylation defining cancer malignancy: new wine in an old bottle
.
Proc. Natl Acad. Sci. U.S.A.
99
,
10231
10233
39
Ernst
,
B.
and
Magnani
,
J.L.
(
2009
)
From carbohydrate leads to glycomimetic drugs
.
Nat. Rev. Drug Discov.
8
,
661
677
40
Adamczyk
,
B.
,
Tharmalingam
,
T.
and
Rudd
,
P.M.
(
2012
)
Glycans as cancer biomarkers
.
Biochim. Biophys. Acta
1820
,
1347
1353
41
Kirwan
,
A.
,
Utratna
,
M.
,
O'Dwyer
,
M.E.
,
Joshi
,
L.
and
Kilcoyne
,
M.
(
2015
)
Glycosylation-based serum biomarkers for cancer diagnostics and prognostics
.
BioMed Res. Int.
2015
,
e490531
42
Engle
,
D.D.
,
Tiriac
,
H.
,
Rivera
,
K.D.
,
Pommier
,
A.
,
Whalen
,
S.
,
Oni
,
T.E.
et al (
2019
)
The glycan CA19-9 promotes pancreatitis and pancreatic cancer in mice
.
Science
364
,
1156
1162
43
Chen
,
C.-Y.
,
Jan
,
Y.-H.
,
Juan
,
Y.-H.
,
Yang
,
C.-J.
,
Huang
,
M.-S.
,
Yu
,
C.-J.
et al (
2013
)
Fucosyltransferase 8 as a functional regulator of nonsmall cell lung cancer
.
Proc. Natl Acad. Sci. U.S.A.
110
,
630
635
44
Leng
,
Q.
,
Tsou
,
J.-H.
,
Zhan
,
M.
and
Jiang
,
F.
(
2018
)
Fucosylation genes as circulating biomarkers for lung cancer
.
J Cancer Res. Clin. Oncol.
144
,
2109
2115
45
Honma
,
R.
,
Kinoshita
,
I.
,
Miyoshi
,
E.
,
Tomaru
,
U.
,
Matsuno
,
Y.
,
Shimizu
,
Y.
et al (
2015
)
Expression of fucosyltransferase 8 is associated with an unfavorable clinical outcome in non-small cell lung cancers
.
Oncology
88
,
298
308
46
Wang
,
X.
,
Chen
,
J.
,
Li
,
Q.K.
,
Peskoe
,
S.B.
,
Zhang
,
B.
,
Choi
,
C.
et al (
2014
)
Overexpression of α (1,6) fucosyltransferase associated with aggressive prostate cancer
.
Glycobiology
24
,
935
944
47
Höti
,
N.
,
Yang
,
S.
,
Hu
,
Y.
,
Shah
,
P.
,
Haffner
,
M.C.
and
Zhang
,
H.
(
2018
)
Overexpression of α (1,6) fucosyltransferase in the development of castration-resistant prostate cancer cells
.
Prostate Cancer and Prostatic Dis.
21
,
137
146
48
Fujita
,
K.
,
Shimomura
,
M.
,
Uemura
,
M.
,
Nakata
,
W.
,
Sato
,
M.
,
Nagahara
,
A.
et al (
2014
)
Serum fucosylated haptoglobin as a novel prognostic biomarker predicting high-Gleason prostate cancer
.
Prostate
74
,
1052
1058
49
Ito
,
Y.
,
Miyauchi
,
A.
,
Yoshida
,
H.
,
Uruno
,
T.
,
Nakano
,
K.
,
Takamura
,
Y.
et al (
2003
)
Expression of alpha1,6-fucosyltransferase (FUT8) in papillary carcinoma of the thyroid: its linkage to biological aggressiveness and anaplastic transformation
.
Cancer Lett.
200
,
167
172
50
Noda
,
M.
,
Okayama
,
H.
,
Kofunato
,
Y.
,
Chida
,
S.
,
Saito
,
K.
,
Tada
,
T.
et al (
2018
)
Prognostic role of FUT8 expression in relation to p53 status in stage II and III colorectal cancer
.
PLoS One
13
,
e0200315
51
Kumar
,
S.
,
Das
,
S.
,
Rachagani
,
S.
,
Kaur
,
S.
,
Joshi
,
S.
,
Johansson
,
S.L.
et al (
2015
)
NCOA3-mediated upregulation of mucin expression via transcriptional and post-translational changes during the development of pancreatic cancer
.
Oncogene
34
,
4879
4889
52
Tada
,
K.
,
Ohta
,
M.
,
Hidano
,
S.
,
Watanabe
,
K.
,
Hirashita
,
T.
,
Oshima
,
Y.
et al (
2020
)
Fucosyltransferase 8 plays a crucial role in the invasion and metastasis of pancreatic ductal adenocarcinoma
.
Surg. Today
50
,
767
777
53
Takahashi
,
T.
,
Ikeda
,
Y.
,
Miyoshi
,
E.
,
Yaginuma
,
Y.
,
Ishikawa
,
M.
and
Taniguchi
,
N.
(
2000
)
Alpha1,6fucosyltransferase is highly and specifically expressed in human ovarian serous adenocarcinomas
.
Int. J. Cancer
88
,
914
919
54
Tu
,
C.-F.
,
Wu
,
M.-Y.
,
Lin
,
Y.-C.
,
Kannagi
,
R.
and
Yang
,
R.-B.
(
2017
)
FUT8 promotes breast cancer cell invasiveness by remodeling TGF-β receptor core fucosylation
.
Breast Cancer Res.
19
,
111
55
Cheng
,
L.
,
Gao
,
S.
,
Song
,
X.
,
Dong
,
W.
,
Zhou
,
H.
,
Zhao
,
L.
et al (
2016
)
Comprehensive N-glycan profiles of hepatocellular carcinoma reveal association of fucosylation with tumor progression and regulation of FUT8 by microRNAs
.
Oncotarget
7
,
61199
61214
56
Lee
,
H.-S.
,
Park
,
M.-H.
,
Yang
,
S.-J.
,
Park
,
K.C.
,
Kim
,
N.-S.
,
Kim
,
Y.-S.
et al (
2007
)
Novel candidate targets of Wnt/beta-catenin signaling in hepatoma cells
.
Life Sci.
80
,
690
698
57
Wang
,
Y.
,
Fukuda
,
T.
,
Isaji
,
T.
,
Lu
,
J.
,
Im
,
S.
,
Hang
,
Q.
et al (
2015
)
Loss of α1,6-fucosyltransferase inhibits chemical-induced hepatocellular carcinoma and tumorigenesis by down-regulating several cell signaling pathways
.
FASEB J.
29
,
3217
3227
58
Norton
,
P.A.
and
Mehta
,
A.S.
(
2019
)
Expression of genes that control core fucosylation in hepatocellular carcinoma: systematic review
.
World J. Gastroenterol.
25
,
2947
2960
59
Nakagawa
,
T.
,
Uozumi
,
N.
,
Nakano
,
M.
,
Mizuno-Horikawa
,
Y.
,
Okuyama
,
N.
,
Taguchi
,
T.
et al (
2006
)
Fucosylation of N-glycans regulates the secretion of hepatic glycoproteins into bile ducts
.
J. Biol. Chem.
281
,
29797
29806
60
Lv
,
X.
,
Song
,
J.
,
Xue
,
K.
,
Li
,
Z.
,
Li
,
M.
,
Zahid
,
D.
et al (
2019
)
Core fucosylation of copper transporter 1 plays a crucial role in cisplatin-resistance of epithelial ovarian cancer by regulating drug uptake
.
Mol. Carcinog.
58
,
794
807
61
Yue
,
L.
,
Han
,
C.
,
Li
,
Z.
,
Li
,
X.
,
Liu
,
D.
,
Liu
,
S.
et al (
2016
)
Fucosyltransferase 8 expression in breast cancer patients: a high throughput tissue microarray analysis
.
Histol. Histopathol.
31
,
547
555
62
Kobayashi
,
S.
,
Fujinawa
,
R.
,
Ota
,
F.
,
Kobayashi
,
S.
,
Angata
,
T.
,
Ueno
,
M.
et al (
2013
)
A single dose of lipopolysaccharide into mice with emphysema mimics human chronic obstructive pulmonary disease exacerbation as assessed by micro-computed tomography
.
Am. J. Respir. Cell Mol. Biol.
49
,
971
977
63
Kamio
,
K.
,
Yoshida
,
T.
,
Gao
,
C.
,
Ishii
,
T.
,
Ota
,
F.
,
Motegi
,
T.
et al (
2012
)
Α1,6-Fucosyltransferase (Fut8) is implicated in vulnerability to elastase-induced emphysema in mice and a possible non-invasive predictive marker for disease progression and exacerbations in chronic obstructive pulmonary disease (COPD)
.
Biochem. Biophys. Res. Commun.
424
,
112
117
64
Gao
,
C.
,
Maeno
,
T.
,
Ota
,
F.
,
Ueno
,
M.
,
Korekane
,
H.
,
Takamatsu
,
S.
et al (
2012
)
Sensitivity of heterozygous α1,6-fucosyltransferase knock-out mice to cigarette smoke-induced emphysema: Implication of aberrant transforming growth factor -β signaling and matrix metalloproteinase gene expression
.
J. Biol. Chem.
287
,
16699
16708
65
Burney
,
P.
,
Patel
,
J.
,
Newson
,
R.
,
Minelli
,
C.
and
Naghavi
,
M.
(
2015
)
Global and regional trends in chronic obstructive pulmonary disease mortality 1990-2010
.
Eur. Respir. J.
45
,
1239
1247
66
Barnes
,
P.J.
,
Burney
,
P.G.J.
,
Silverman
,
E.K.
,
Celli
,
B.R.
,
Vestbo
,
J.
,
Wedzicha
,
J.A.
et al (
2015
)
Chronic obstructive pulmonary disease
.
Nat. Rev. Dis. Primers
1
,
1
21
67
De Serres
,
G.
,
Lampron
,
N.
,
La Forge
,
J.
,
Rouleau
,
I.
,
Bourbeau
,
J.
,
Weiss
,
K.
et al (
2009
)
Importance of viral and bacterial infections in chronic obstructive pulmonary disease exacerbations
.
J. Clin. Virol.
46
,
129
133
68
Durham
,
A.L.
and
Adcock
,
I.M.
(
2015
)
The relationship between COPD and lung cancer
.
Lung Cancer
90
,
121
127
69
Sethi
,
S.
(
2004
)
Bacteria in exacerbations of chronic obstructive pulmonary disease
.
Proc. Am. Thorac. Soc.
1
,
109
114
70
Taniguchi-Ikeda
,
M.
,
Morioka
,
I.
,
Iijima
,
K.
and
Toda
,
T.
(
2016
)
Mechanistic aspects of the formation of α-dystroglycan and therapeutic research for the treatment of α-dystroglycanopathy: a review
.
Mol. Aspects Med.
51
,
115
124
71
Ihara
,
H.
,
Ikeda
,
Y.
,
Toma
,
S.
,
Wang
,
X.
,
Suzuki
,
T.
,
Gu
,
J.
et al (
2007
)
Crystal structure of mammalian α1,6-fucosyltransferase, FUT8
.
Glycobiology
17
,
455
466
72
Ihara
,
H.
,
Okada
,
T.
,
Taniguchi
,
N.
and
Ikeda
,
Y.
(
2020
)
Involvement of the α-helical and Src homology 3 domains in the molecular assembly and enzymatic activity of human α1,6-fucosyltransferase, FUT8
.
Biochim. Biophys. Acta Gen. Subj.
1864
,
129596
73
Tomida
,
S.
,
Takata
,
M.
,
Hirata
,
T.
,
Nagae
,
M.
,
Nakano
,
M.
and
Kizuka
,
Y.
(
2020
)
The SH3 domain in the fucosyltransferase FUT8 controls FUT8 activity and localization and is essential for core fucosylation
.
J. Biol. Chem.
295
,
7992
8004
74
Li
,
M.
,
Krishnaveni
,
M.S.
,
Li
,
C.
,
Zhou
,
B.
,
Xing
,
Y.
,
Banfalvi
,
A.
et al (
2011
)
Epithelium-specific deletion of TGF-β receptor type II protects mice from bleomycin-induced pulmonary fibrosis
.
J. Clin. Invest.
121
,
277
287
75
Di Stefano
,
A.
,
Sangiorgi
,
C.
,
Gnemmi
,
I.
,
Casolari
,
P.
,
Brun
,
P.
,
Ricciardolo
,
F.L.M.
et al (
2018
)
TGF-β signaling pathways in different compartments of the lower airways of patients with stable COPD
.
Chest
153
,
851
862
76
Baraldo
,
S.
(
2005
)
Decreased expression of TGF- type II receptor in bronchial glands of smokers with COPD
.
Thorax
60
,
998
1002
77
Zandvoort
,
A.
,
Postma
,
D.S.
,
Jonker
,
M.R.
,
Noordhoek
,
J.A.
,
Vos
,
J.T.W.M.
,
van der Geld
,
Y.M.
et al (
2006
)
Altered expression of the Smad signalling pathway: implications for COPD pathogenesis
.
Eur. Respir. J.
28
,
533
541
78
Yamada
,
M.
,
Ishii
,
T.
,
Ikeda
,
S.
,
Naka-Mieno
,
M.
,
Tanaka
,
N.
,
Arai
,
T.
et al (
2011
)
Association of fucosyltransferase 8 (FUT8) polymorphism Thr267Lys with pulmonary emphysema
.
J. Hum. Genet.
56
,
857
860
79
Suki
,
B.
,
Bartolák-Suki
,
E.
and
Rocco
,
P.R.M.
(
2017
)
Elastase-induced lung emphysema models in mice
.
Methods Mol. Biol.
1639
,
67
75
80
Meyer
,
K.
,
Linker
,
A.
,
Davidson
,
E.A.
and
Weissmann
,
B.
(
1953
)
The mucopolysaccharides of bovine cornea
.
J. Biol. Chem.
205
,
611
616
81
Kuhn
,
C.
and
Senior
,
R.M.
(
1978
)
The role of elastases in the development of emphysema
.
Lung
155
,
185
197
82
Demkow
,
U.
and
van Overveld
,
F.
(
2010
)
Role of elastases in the pathogenesis of chronic obstructive pulmonary disease: implications for treatment
.
Eur. J. Med. Res.
15
,
27
35
83
Gao
,
C.
,
Fujinawa
,
R.
,
Yoshida
,
T.
,
Ueno
,
M.
,
Ota
,
F.
,
Kizuka
,
Y.
et al (
2017
)
A keratan sulfate disaccharide prevents inflammation and the progression of emphysema in murine models
.
Am. J. Physiol. Lung Cell Mol.
312
,
L268
L276
84
Caterson
,
B.
and
Melrose
,
J.
(
2018
)
Keratan sulfate, a complex glycosaminoglycan with unique functional capability
.
Glycobiology
28
,
182
206
85
Uchimura
,
K.
(
2015
)
Keratan sulfate: biosynthesis, structures, and biological functions
.
Methods Mol. Biol.
1229
,
389
400
86
Xu
,
H.
,
Kurihara
,
H.
,
Ito
,
T.
,
Kikuchi
,
H.
,
Yoshida
,
K.
,
Yamanokuchi
,
H.
et al (
2005
)
The keratan sulfate disaccharide Gal(6S03) beta1,4-GlcNAc(6S03) modulates interleukin 12 production by macrophages in murine Thy-1 type autoimmune disease
.
J. Biol. Chem.
280
,
20879
20886
87
Shirato
,
K.
,
Gao
,
C.
,
Ota
,
F.
,
Angata
,
T.
,
Shogomori
,
H.
,
Ohtsubo
,
K.
et al (
2013
)
Flagellin/Toll-like receptor 5 response was specifically attenuated by keratan sulfate disaccharide via decreased EGFR phosphorylation in normal human bronchial epithelial cells
.
Biochem. Biophys. Res. Commun.
435
,
460
465
88
Ota
,
F.
,
Hirayama
,
T.
,
Kizuka
,
Y.
,
Yamaguchi
,
Y.
,
Fujinawa
,
R.
,
Nagata
,
M.
et al (
2018
)
High affinity sugar ligands of C-type lectin receptor langerin
.
Biochim. Biophys. Acta Gen. Subj.
1862
,
1592
1601
89
Valladeau
,
J.
,
Ravel
,
O.
,
Dezutter-Dambuyant
,
C.
,
Moore
,
K.
,
Kleijmeer
,
M.
,
Liu
,
Y.
et al (
2000
)
Langerin, a novel C-type lectin specific to Langerhans cells, is an endocytic receptor that induces the formation of Birbeck granules
.
Immunity
12
,
71
81
90
Hunger
,
R.E.
,
Sieling
,
P.A.
,
Ochoa
,
M.T.
,
Sugaya
,
M.
,
Burdick
,
A.E.
,
Rea
,
T.H.
et al (
2004
)
Langerhans cells utilize CD1a and langerin to efficiently present nonpeptide antigens to T cells
.
J. Clin. Invest.
113
,
701
708
91
Epaulard
,
O.
,
Adam
,
L.
,
Poux
,
C.
,
Zurawski
,
G.
,
Salabert
,
N.
,
Rosenbaum
,
P.
et al (
2014
)
Macrophage- and neutrophil-derived TNF-α instructs skin langerhans cells to prime antiviral immune responses
.
J. Immunol.
193
,
2416
2426
92
van Dalen
,
R.
,
De La Cruz Diaz
,
J.S.
,
Rumpret
,
M.
,
Fuchsberger
,
F.F.
,
van Teijlingen
,
N.H.
,
Hanske
,
J.
et al (
2019
)
Langerhans cells sense staphylococcus aureus wall teichoic acid through langerin to induce inflammatory responses
.
mBio
10
,
e00330-19
93
Zhao
,
J.
,
Liu
,
X.
,
Kao
,
C.
,
Zhang
,
E.
,
Li
,
Q.
,
Zhang
,
F.
et al (
2016
)
Kinetic and structural studies of interactions between glycosaminoglycans and langerin
.
Biochemistry
55
,
4552
4559
94
Tateno
,
H.
,
Ohnishi
,
K.
,
Yabe
,
R.
,
Hayatsu
,
N.
,
Sato
,
T.
,
Takeya
,
M.
et al (
2010
)
Dual specificity of Langerin to sulfated and mannosylated glycans via a single C-type carbohydrate recognition domain
.
J. Biol. Chem.
285
,
6390
6400
95
Shin
,
J.I.
and
Brusselle
,
G.G.
(
2014
)
Mechanistic links between COPD and lung cancer: a role of microRNA let-7?
Nat. Rev. Cancer
14
,
70
70
96
Cruz
,
D.
,
Tanoue
,
C.S.
,
and Matthay
,
L.T.
and
A
,
R.
(
2011
)
Lung cancer: epidemiology, etiology, and prevention
.
Clin. Chest Med.
32
,
605
644
97
Mouronte-Roibás
,
C.
,
Leiro-Fernández
,
V.
,
Fernández-Villar
,
A.
,
Botana-Rial
,
M.
,
Ramos-Hernández
,
C.
and
Ruano-Ravina
,
A.
(
2016
)
COPD, emphysema and the onset of lung cancer. A systematic review
.
Cancer Lett.
382
,
240
244
98
Agrawal
,
P.
,
Fontanals-Cirera
,
B.
,
Sokolova
,
E.
,
Jacob
,
S.
,
Vaiana
,
C.A.
,
Argibay
,
D.
et al (
2017
)
A systems biology approach identifies FUT8 as a driver of melanoma metastasis
.
Cancer Cell
31
,
804
819.e7
99
Taniguchi
,
N.
,
Ohkawa
,
Y.
,
Maeda
,
K.
,
Harada
,
Y.
,
Nagae
,
M.
,
Kizuka
,
Y.
et al (
2020
)
True significance of N-acetylglucosaminyltransferases GnT-III, V and α1,6 fucosyltransferase in epithelial-mesenchymal transition and cancer
.
Mol Aspects Med.
100905
100
Li
,
F.
,
Zhao
,
S.
,
Cui
,
Y.
,
Guo
,
T.
,
Qiang
,
J.
,
Xie
,
Q.
et al (
2020
)
Α1,6-Fucosyltransferase (FUT8) regulates the cancer-promoting capacity of cancer-associated fibroblasts (CAFs) by modifying EGFR core fucosylation (CF) in non-small cell lung cancer (NSCLC)
.
Am. J. Cancer Res.
10
,
816
837
PMID:
[PubMed]
101
Zhou
,
Y.
,
Fukuda
,
T.
,
Hang
,
Q.
,
Hou
,
S.
,
Isaji
,
T.
,
Kameyama
,
A.
et al (
2017
)
Inhibition of fucosylation by 2-fluorofucose suppresses human liver cancer HepG2 cell proliferation and migration as well as tumor formation
.
Sci. Rep.
7
,
11563
102
Fehres
,
C.M.
,
Kalay
,
H.
,
Bruijns
,
S.C.M.
,
Musaafir
,
S.A.M.
,
Ambrosini
,
M.
,
van Bloois
,
L.
et al (
2015
)
Cross-presentation through langerin and DC-SIGN targeting requires different formulations of glycan-modified antigens
.
J. Control Release
203
,
67
76
103
Porkolab
,
V.
,
Chabrol
,
E.
,
Varga
,
N.
,
Ordanini
,
S.
,
Sutkevičiu Tė
,
I.
,
Thépaut
,
M.
et al (
2018
)
Rational-differential design of highly specific glycomimetic ligands: targeting DC-SIGN and excluding langerin recognition
.
ACS Chem. Biol.
13
,
600
608
104
Holla
,
A.
and
Skerra
,
A.
(
2011
)
Comparative analysis reveals selective recognition of glycans by the dendritic cell receptors DC-SIGN and Langerin
.
Protein Eng. Des. Sel.
24
,
659
669
105
Wamhoff
,
E.-C.
,
Schulze
,
J.
,
Bellmann
,
L.
,
Rentzsch
,
M.
,
Bachem
,
G.
,
Fuchsberger
,
F.F.
et al (
2019
)
A specific, glycomimetic langerin ligand for human langerhans cell targeting
.
ACS Cent. Sci.
5
,
808
820
106
Wen
,
H.-C.
,
Lin
,
C.-H.
,
Huang
,
J.-S.
,
Tsai
,
C.-L.
,
Chen
,
T.-F.
and
Wang
,
S.-K.
(
2019
)
Selective targeting of DC-SIGN by controlling the oligomannose pattern on a polyproline tetra-helix macrocycle scaffold
.
Chem. Commun.
55
,
9124
9127
107
Van Pottelberge
,
G.R.
,
Bracke
,
K.R.
,
Joos
,
G.F.
and
Brusselle
,
G.G.
(
2009
)
The role of dendritic cells in the pathogenesis of COPD: liaison officers in the front line
.
J. Chronic Obstr. Pulm. Dis.
6
,
284
290
This is an open access article published by Portland Press Limited on behalf of the Biochemical Society and distributed under the Creative Commons Attribution License 4.0 (CC BY-NC-ND).