Characterization of a Novel C-type Lectin-like Gene, LSECtin
Wanli Liu,Li Tang,Ge Zhang,Wei Huang,Yan Cui,Lihai Guo,Zikuan Gou,Xiaoxiao Chen,Daifeng Jiang,Yunping Zhu,Ge-Fei Kang,Fuchu He
DOI: https://doi.org/10.1074/jbc.m311227200
2004-01-01
Abstract:A new C-type lectin-like gene encodes 293 amino acids and maps to chromosome 19p13.3 adjacent to the previously described C-type lectin genes, CD23, dendritic cell-specific intercellular adhesion molecule-3-grabbing nonintegrin (DC-SIGN), and DC-SIGN-related protein (DC-SIGNR). The four genes form a tight cluster in an insert size of 105 kb and have analogous genomic structures. The new C-type lectin-like molecule, designated liver and lymph node sinusoidal endothelial cell C-type lectin (LSECtin), is a type II integral membrane protein of ∼40 kDa in size with a single C-type lectin-like domain at the COOH terminus, closest in homology to DC-SIGNR, DC-SIGN, and CD23. LSECtin mRNA was only expressed in liver and lymph node among 15 human tissues tested, intriguingly neither expressed on hematopoietic cell lines nor on monocyte-derived dendritic cells (DCs). Moreover, LSECtin is expressed predominantly by sinusoidal endothelial cells of human liver and lymph node and co-expressed with DC-SIGNR. LSECtin binds to mannose, GlcNAc, and fucose in a Ca2+-dependent manner but not to galactose. Our results indicate that LSECtin is a novel member of a family of proteins comprising CD23, DC-SIGN, and DC-SIGNR and might function in vivo as a lectin receptor. A new C-type lectin-like gene encodes 293 amino acids and maps to chromosome 19p13.3 adjacent to the previously described C-type lectin genes, CD23, dendritic cell-specific intercellular adhesion molecule-3-grabbing nonintegrin (DC-SIGN), and DC-SIGN-related protein (DC-SIGNR). The four genes form a tight cluster in an insert size of 105 kb and have analogous genomic structures. The new C-type lectin-like molecule, designated liver and lymph node sinusoidal endothelial cell C-type lectin (LSECtin), is a type II integral membrane protein of ∼40 kDa in size with a single C-type lectin-like domain at the COOH terminus, closest in homology to DC-SIGNR, DC-SIGN, and CD23. LSECtin mRNA was only expressed in liver and lymph node among 15 human tissues tested, intriguingly neither expressed on hematopoietic cell lines nor on monocyte-derived dendritic cells (DCs). Moreover, LSECtin is expressed predominantly by sinusoidal endothelial cells of human liver and lymph node and co-expressed with DC-SIGNR. LSECtin binds to mannose, GlcNAc, and fucose in a Ca2+-dependent manner but not to galactose. Our results indicate that LSECtin is a novel member of a family of proteins comprising CD23, DC-SIGN, and DC-SIGNR and might function in vivo as a lectin receptor. Protein-carbohydrate interactions serve multiple functions in the immune system. A number of animal lectins (sugar-binding proteins) mediate both pathogen recognition and cell-cell interactions using structurally related Ca2+-dependent carbohydrate-recognition domains (C-type CRDs) 1The abbreviations used are: CRD, carbohydrate recognition domain; BSA, bovine serum albumin; CTLD, C-type lectin-like domain; DC, dendritic cell(s); DC-SIGN, DC-specific ICAM-3-grabbing nonintegrin; DC-SIGNR, DC-SIGN-related protein; EST, expressed sequence tag; FCS, fetal cattle serum; FACS, fluorescence-activated cell sorting; ICAM, intercellular adhesion molecule; ORF, open reading frame; PBMC, peripheral blood monocyte cell; RACE, rapid amplification of cDNA ends; CHO, Chinese hamster ovary; aa, amino acids; Ab, antibody; mAb, monoclonal antibody; contig, group of overlapping clones; LSECtin, liver and lymph node sinusoidal endothelial cell C-type lectin; sLSECtin, soluble LSECtin. (1Weis W.I. Taylor M.E. Drickamer K. Immunol. Rev. 1998; 163: 19-34Google Scholar). There are two groups of membrane-bound C-type lectins, type I and type II C-type lectins, which are distinguished from each other by the orientation of their cell surface NH2 terminus pointing outwards or into the cytoplasm of the cell, respectively. The type II C-type lectins identified so far have a single CRD at the extracellular carboxyl terminus (2Figdor C.G. van Kooyk Y. Adema G.J. Nat. Immunol. Rev. 2002; 2: 77-84Google Scholar). The genes coding for the type II C-type lectins are mainly localized in chromosome 12p12.3-p13.2 (3Trowsdale J. Barten R. Haude A. Stewart C.A. Beck S. Wilson M.J. Immunol. Rev. 2001; 181: 20-38Google Scholar) and 19p13.3 (4Soilleux E.J. Barten R. Trowsdale J. J. Immunol. 2000; 165: 2937-2942Google Scholar). The former is the natural killer gene complex, and the latter is the cluster of genes including CD23, dendritic cell-specific intercellular adhesion molecule-3-grabbing nonintegrin (DC-SIGN), and DC-SIGN-related protein (DC-SIGNR). In the last cluster, these three genes have analogous genomic structures and form a tight cluster in an insert size of 105 kb on human chromosome 19p13.3 (4Soilleux E.J. Barten R. Trowsdale J. J. Immunol. 2000; 165: 2937-2942Google Scholar). They display similar domain composition and organization containing a short NH2-terminal cytoplasmic tail, a transmembrane region, and an extracellular region that consists of a coiled-coil neck domain and a C-terminal CRD. The close linkage and similar genomic and protein structures suggest that these three genes may have arisen via duplication of an ancestral gene (5Soilleux E.J. Clin. Sci. 2003; 104: 437-446Google Scholar). Indeed, they share some common functions in vivo in that they serve as cell-cell adhesion molecules and play an important role in the immune system (6Bonnefoy J.Y. Lecoanet-Henchoz S. Gauchat J.F. Graber P. Aubry J.P. Jeannin P. Plater-Zyberk C. Int. Rev. Immunol. 1997; 16: 113-128Google Scholar, 7Van Kooyk Y. Geijtenbeek T.B. Immunol. Rev. 2002; 186: 47-56Google Scholar, 8Bashirova A.A. Geijtenbeek T.B. van Duijnhoven G.C.F. van Vliet S.J. Eilering J.B.G. Martin M.P. Wu L. Martin T.D. Viebig N. Knolle P.A. KewalRamani V.N. van Kooyk Y. Carrington M. J. Exp. Med. 2001; 193: 671-678Google Scholar). CD23, a low affinity receptor for IgE, was first identified as a type II C-type lectin in the latter gene cluster, which was expressed on a wide variety of human hematopoietic cell types including B and T cells, follicular dendritic cells, monocytes, platelets, Langerhans cells, esoinophils, and natural killer cells (9Conrad D.H. Annu. Rev. Immunol. 1990; 8: 623-645Google Scholar). CD23 specifically recognizes at least four different ligands, including IgE (10Yukawa K. Kikutani H. Howaki H. Yamasaki K. Yokota A. Nakamura H. Barsumian E.L. Hardy R.R. Suemura M. Kishimoto T. J. Immunol. 1987; 38: 2576-2580Google Scholar), CD21 (11Aubry J.P. Pochon S. Gauchat J.F. Nueda-Marin A. Holers V.M. Graber P. Siegfried C. Bonnefoy J.Y. J. Immunol. 1994; 152: 5806-5813Google Scholar), CD18/CD11b, and CD18/CD11c β2-integrins (12Lecoanet-Henchoz S. Gauchat J.F. Aubry J.P. Graber P. Life P. Paul-Eugene N. Ferrua B. Corbi A.L. Daugs B. Plater-Zyberk C. Bonnefoy J.Y. Immunity. 1995; 3: 119-125Google Scholar). The CD23-CD21 and CD23-β2-integrin interactions are lectin-like in that CD23 recognizes specific carbohydrate structures expressed on CD21 and CD11b/c, respectively (11Aubry J.P. Pochon S. Gauchat J.F. Nueda-Marin A. Holers V.M. Graber P. Siegfried C. Bonnefoy J.Y. J. Immunol. 1994; 152: 5806-5813Google Scholar, 12Lecoanet-Henchoz S. Gauchat J.F. Aubry J.P. Graber P. Life P. Paul-Eugene N. Ferrua B. Corbi A.L. Daugs B. Plater-Zyberk C. Bonnefoy J.Y. Immunity. 1995; 3: 119-125Google Scholar). In contrast, CD23 binds deglycosylated IgE, suggesting the non-lectin-like recognition of a protein (rather than a carbohydrate) epitope (13Delespesse G. Sarfati M. Wu C.Y. Fournier S. Letellier M. Immunol. Rev. 1992; 125: 77-97Google Scholar). CD23 plays an important role in a variety of biological activities such as cell-cell adhesions, survival of B cells in germinal centers, regulation of IgE synthesis, and antigen presentation (6Bonnefoy J.Y. Lecoanet-Henchoz S. Gauchat J.F. Graber P. Aubry J.P. Jeannin P. Plater-Zyberk C. Int. Rev. Immunol. 1997; 16: 113-128Google Scholar). DC-SIGN, originally described in 1992 as a C-type lectin being able to bind the human immunodeficiency virus surface protein, gp120 (14Curtis B.M. Scharnowske S. Watson A.J. Proc. Natl. Acad. Sci. U. S. A. 1992; 89: 8356-8360Google Scholar), is expressed on dendritic cells and some subsets of macrophages (15Geijtenbeek T.B. Torensma R. van Vliet S.J. van Duijnhoven G.C. Adema G.J. van Kooyk Y. Figdor C.G. Cell. 2000; 100: 575-585Google Scholar, 16Soilleux E.J. Morris L.S. Leslie G. Chehimi J. Luo Q. Levroney E. Trowsdale J. Montaner L.J. Doms R.W. Weissman D. Coleman N. Lee B. J. Leukocyte Biol. 2002; 71: 445-457Google Scholar). DC-SIGNR, which shows 77% identity to DC-SIGN at the nuclear acid level, is highly expressed on human liver and lymph node sinusoidal and placental endothelial cells (8Bashirova A.A. Geijtenbeek T.B. van Duijnhoven G.C.F. van Vliet S.J. Eilering J.B.G. Martin M.P. Wu L. Martin T.D. Viebig N. Knolle P.A. KewalRamani V.N. van Kooyk Y. Carrington M. J. Exp. Med. 2001; 193: 671-678Google Scholar, 17Pohlmann S. Soilleux E.J. Baribaud F. Leslie G.J. Morris L.S. Trowsdale J. Lee B. Coleman N. Doms R.W. Proc. Natl. Acad. Sci. U. S. A. 2001; 98: 2670-2675Google Scholar). Both of DC-SIGN and DC-SIGNR are typical C-type lectins in that they bind ligands bearing mannose and related sugar through their CRDs (18Mitchell D.A. Fadden A.J. Drickamer K. J. Biol. Chem. 2001; 276: 28939-28945Google Scholar). DC-SIGN regulates the migration of dendritic cells and their interaction with lymphocytes through interactions with ICAM-2 on endothelium and ICAM-3 on T cells, respectively (15Geijtenbeek T.B. Torensma R. van Vliet S.J. van Duijnhoven G.C. Adema G.J. van Kooyk Y. Figdor C.G. Cell. 2000; 100: 575-585Google Scholar, 19Geijtenbeek T.B. Krooshoop D.J. Bleijs D.A. van Vliet S.J. van Duijnhoven G.C. Grabovsky V. Alon R. Figdor C.G. van Kooyk Y. Nat. Immunol. 2000; 1: 353-357Google Scholar). DC-SIGNR also binds to ICAM-3 on T cells and is involved in liver sinusoidal endothelial cells (LSEC) interactions with passing leukocytes (8Bashirova A.A. Geijtenbeek T.B. van Duijnhoven G.C.F. van Vliet S.J. Eilering J.B.G. Martin M.P. Wu L. Martin T.D. Viebig N. Knolle P.A. KewalRamani V.N. van Kooyk Y. Carrington M. J. Exp. Med. 2001; 193: 671-678Google Scholar, 17Pohlmann S. Soilleux E.J. Baribaud F. Leslie G.J. Morris L.S. Trowsdale J. Lee B. Coleman N. Doms R.W. Proc. Natl. Acad. Sci. U. S. A. 2001; 98: 2670-2675Google Scholar). To date, there have been many studies suggesting that DC-SIGN and DC-SIGNR can also facilitate infections of viral and bacterial pathogens including human immunodeficiency virus (20Geijtenbeek T.B. Kwon D.S. Torensma R. van Vliet S.J. van Duijnhoven G.C. Middel J. Cornelissen I.L. Nottet H.S. KewalRamani V.N. Littman D.R. Figdor C.G. van Kooyk Y. Cell. 2000; 100: 587-597Google Scholar), Ebola virus (21Alvarez C.P. Lasala F. Carrillo J. Muniz O. Corbi A.L. Delgado R. J. Virol. 2002; 76: 6841-6844Google Scholar), cytomegalovirus (22Halary F. Amara A. Lortat-Jacob H. Messerle M. Delaunay T. Houles C. Fieschi F. Arenzana-Seisdedos F. Moreau J.F. Dechanet-Merville J. Immunity. 2002; 17: 653-664Google Scholar), hepatitis C virus (23Lozach P.Y. Lortat-Jacob H. De Lacroix De Lavalette A. Staropoli I. Foung S. Amara A. Houles C. Fieschi F. Schwartz O. Virelizier J.L. Arenzana-Seisdedos F. Altmeyer R. J. Biol. Chem. 2003; 278: 20358-20366Google Scholar), and Mycobacterium tuberculosis (24Tailleux L. Schwartz O. Herrmann J.L. Pivert E. Jackson M. Amara A. Legres L. Dreher D. Nicod L.P. Gluckman J.C. Lagrange P.H. Gicquel B. Neyrolles O. J. Exp. Med. 2003; 97: 121-127Google Scholar, 25Geijtenbeek T.B. Van Vliet S.J. Koppel E.A. Sanchez-Hernandez M. Vandenbroucke-Grauls C.M. Appelmelk B. Van Kooyk Y. J. Exp. Med. 2003; 197: 7-17Google Scholar), in which mannose and related sugar on envelope glycoproteins of these pathogens play a key role in their interactions. Recently, many new genes with important biological functions have been detected based on large scale sequencing of human fetal liver cDNA library in our laboratory (26Qu X.H. Zhang C.G. Zhai Y. Xing G.C. Wei H.D. Yu Y.T. Wu S.F. He F.C. Gene (Amst.). 2001; 264: 37-44Google Scholar, 27Qu X. Wei H. Zhai Y. Que H. Chen Q. Tang F. Wu Y. Xing G. Zhu Y. Liu S. Fan M. He F. J. Biol. Chem. 2002; 277: 35574-35585Google Scholar, 28Zhang C. Yu Y. Zhang S. Liu M. Xing G. Wei H. Bi J. Liu X. Zhou G. Dong C. Hu Z. Zhang Y. Luo L. Wu C. Zhao S. He F. Genomics. 2000; 63: 400-408Google Scholar). To identify potential novel adhesion molecules, more than 14,000 expressed sequence tags (ESTs) of human fetal liver cDNA libraries (29Yu Y. Zhang C. Zhou G. Wu S. Qu X. Wei H. Xing G. Dong C. Zhai Y. Wan J. Ouyang S. Li L. Zhang S. Zhou K. Zhang Y. Wu C. He F. Genome Res. 2001; 11: 1392-1403Google Scholar) have been analyzed to search homologues of CD23, DC-SIGN, and DC-SIGNR, which were selected as baits, because all of them are important adhesion molecules involved in host defense. Among these ESTs, an insert clone D1210 with moderate homology to human DC-SIGNR, DC-SIGN, and CD23 at the amino acid level was selected for further study. This novel C-type lectin-like gene, termed LSECtin, is localized between CD23 and DC-SIGN, which is followed by DC-SIGNR, on chromosome 19p13.3, and these four structurally related genes form a tight cluster. In this study, we report on the molecular cloning, mapping, expression of LSECtin, and characterization of its sugar-binding activities. Cell Lines—The promyelomonocytic cell line HL60; the chronic myelogenous leukemia K562; the monocyte cell lines U937 and THP-1; the B cell lines Raji, Daudi, and Namalwa; the T cell lines Jurkat, Hut-78, and Molt-4; the natural killer cell line, NKL; and liver cancer cell lines HepG2 and BEL-7402 were all obtained from the Cell Bank (Shanghai Institute of Cell Biology, Chinese Academy of Sciences). The CHO cell line was obtained from the ATCC. Hematopoietic cell lines were cultured in RPMI 1640 (Invitrogen) supplemented with 2 mm glutamine, 10% heat-inactivated FCS (Hyclone), and 100 units/ml penicillin/streptomycin. HepG2, BEL-7402, and CHO cells were cultured in Dulbecco's modified Eagle's medium with 2 mm glutamine, 10% heat-inactivated FCS, and 100 units/ml penicillin/streptomycin. Human umbilical vein endothelial cells were prepared as previously reported (30Whelan J. Ghersa P. Hooft van Huijsduijnen R. Gray J. Chandra G. Talabot F. DeLamarter J.F. Nucleic Acids Res. 1991; 19: 2645-2653Google Scholar). Peripheral blood mononuclear cells (PBMCs) were isolated from healthy adult volunteers by Ficoll centrifugation (Amersham Biosciences). An aliquot of these PBMCs were stimulated for 72 h with phytohemagglutinin (5 μg/ml; Sigma). Peripheral blood granulocytes were purified from erythrocytes by hypotonic lysis (155 mm NH4Cl). DC cultures were obtained from peripheral blood monocytes as described elsewhere (31Romani N. Gruner S. Braug D. Lenz A. Trockenbacher B. Konwalinka G. Fritsch P.O. Steinman R.M. Schuler G. J. Exp. Med. 1994; 180: 83-93Google Scholar). In brief, PBMCs in RPMI 1640, 10% FCS were allowed to adhere to 6-well plates (Costar). After 1 h at 37 °C, the nonadherent cells were washed off by extensive washing with phosphate-buffered saline, and the adherent monocytes were cultured in RPMI 1640, 10% FCS with 1000 units/ml GM-CSF and 1000 units/ml IL-4 (R & D Systems). On day 4, the cultures were refreshed by adding culture medium supplemented with granulocyte-macrophage colony-stimulating factor and interleukin-4 (both at 500 units/ml). At day 7, nonadherent and loosely proliferating DC aggregates were collected and tested by flow cytometry for CD1α, CD86, CD83, and HLA-DR (fluorescein isothiocyanate-conjugated mAbs; Becton Dickinson) expression. Identification and Cloning of Human LSECtin—To identify novel adhesion molecules homologous to CD23, DC-SIGN, and DC-SIGNR, a cDNA library of human fetal liver of 22 weeks in our laboratory (29Yu Y. Zhang C. Zhou G. Wu S. Qu X. Wei H. Xing G. Dong C. Zhai Y. Wan J. Ouyang S. Li L. Zhang S. Zhou K. Zhang Y. Wu C. He F. Genome Res. 2001; 11: 1392-1403Google Scholar) was searched using the tBlastn algorithm with their CRD sequences. A novel human cDNA clone (D1210) homologous to human DC-SIGNR, DC-SIGN, and CD23 was isolated and characterized. An online BLAST search was performed against the public EST and nonredundant GenBank™ data base using the D1210 sequence, and then the human clones homologous to D1210 were picked for further analysis. Based on the results of ESTs, we designed an antisense primer (GSP1, 5′-GGA ACA GCT CAG TGC GGA CGT CCT CAC-3′) and a sense primer (GSP2, 5′-CCT GCG GGA ACT GCG TGA GCG CGT GAC-3′) to perform 5′-RACE and 3′-RACE, respectively. 5′-RACE and 3′-RACE were carried out using a SMARTTM RACE cDNA amplification kit (Clontech) on adapter-ligated cDNA, synthesized from 1 μg of poly(A)+ RNA from fetal liver (Clontech). 5′-RACE and 3′-RACE reactions were performed by touchdown PCR program as described by the manufacturer. In both cases, a nested PCR was performed with an inner primer, NGSP1 (5′-GCT CTC CTG CTC CAT CAG CTT CGC-3′), for 5′-RACE and NGSP2 (5′-GTG AGG ACG TCC GCA CTG AGC TGT TCC-3′) for 3′-RACE, respectively. PCRs were carried out at 95 °C for 5 min followed by 35 cycles at 94 °C for 1 min, 58 °C for 1 min, and 72 °C for 2 min. The predominant fragments were cloned into pGEM-T vector (Promega) for sequencing. DNA sequencing was carried out on an ABI Prism 377-XL DNA sequencer (PerkinElmer Life Sciences). Chromosomal Localization and Sequence Analysis of Human LSEC-tin—The full-length cDNA sequence of LSECtin was used to search the human genome and picked up its corresponding genomic sequence. The alignment analysis between the cDNA and its genomic sequence was performed to determine the boundaries of exons and introns and the intronic lengths of the LSECtin gene. The genomic sequences of DC-SIGN, DC-SIGNR, and CD23 were also obtained using their cDNA for comparison. The sequence alignment among the type II C-type lectins homologous to LSECtin was performed using the program ClustalW. Protein subsequence motif was identified using the network service SMART (available on the World Wide Web at smart.embl-heidelberg.de). Prosite information of the protein was analyzed with the TMpred program that was used to make a prediction of membrane-spanning regions and their orientation. Northern Blot and RT-PCR Analysis—Two human multiple tissue Northern Blots containing ∼2 μg of poly(RNA)+ per lane (MTN; Clontech) were hybridized to the specific probes of LSECtin and β-actin, respectively, following the manufacturer's protocol. Briefly, members were hybridized in ExpressHyb™ hybridization solution (Clontech) containing 1 × 106 cpm/ml 32P-labeled cDNA for LSECtin or β-actin. The probe for LSECtin corresponded to the open reading frame (ORF) of LSECtin and was generated with the Klenow fragment of DNA polymerase I and [α-32P]dNTP using the Primer-a-Gene® Labeling system (Promega). After being washed, the blots were exposed to x-ray film at -70 °C with an intensifying screen. Total RNAs were extracted from human peripheral blood cells, cultured cell lines, placenta, and fetal liver using Trizol reagent (Invitrogen). First cDNA was synthesized using random hexameric primers (Promega) and Moloney murine leukemia virus reverse transcriptase (Promega), as recommended by the manufacturer. Primers flanking the ORF of LSECtin were used for PCRs (forward, 5′-GCC ATG GAC ACC ACC AGG TAC AGC-3′; reverse, 5′-GAC TCA GCA GTT GTG CCT TTT CTC-3′). The PCRs were performed under the following conditions: initial denaturation at 95 °C for 5 min and then 35 cycles at 94 °C for 45 s, annealing at 58 °C for 45 s, and extension at 72 °C for 1 min. Specific primers for glyceraldehyde-3-phosphate dehydrogenase were used as an internal control for cDNA quantity (5′-ACC ACA GTC CAT GCC ATC AC-3′ as sense primer, 5′-TCC ACC ACC CTG TTG CTG TA-3′ as antisense primer). The products were visualized on ethidium bromide gels. Preparation of Anti-LSECtin Polyclonal Antibody—cDNA coding the extracellular LSECtin (aa 55–293) was generated by PCR with the primers (forward, 5′-GGA TTC AAG GCC TCC ACG GAG CGC G-3′; reverse, 5′-GTC GAC TCA GCA GTT GTG CCT TTT CTC-3′), cloned into pGEM-T vector (Promega), and sequenced. The fragment encoding the neck and CTLD domain of LSECtin was ligated immediately downstream of the hexahistidine sequence in pET-28a vector (Novagen). This recombinant vector was introduced into BL21 (DE3) cells. His-LSECtin protein was extracted from isopropyl-1-thio-β-d-galactopyranoside-treated BL21 cells in 8 m urea, 100 mm sodium phosphate, 10 mm Tris-HCl (pH 8.0) buffer and then purified under denatured conditions using Ni2+-nitrilotriacetic acid-agarose (Qiagen). Purified His-LSECtin fusion protein was subjected to preparative SDS-PAGE separation, and then the His-LSECtin protein was excised from the gel, pulverized, and emulsified with an equal volume of Freund's complete adjuvant (Sigma) (32Sambrook J. Fritsch E.F. Maniatis T. Molecular Cloning: A Laboratory Manual.2nd Ed. Cold Spring Harbor Laboratory, Cold Spring Harbor, NY1989: 852-861Google Scholar). The LSECtin antigen was injected subcutaneously into adult New Zealand White rabbits. After four rounds of booster injections with Freund's incomplete adjuvant (Sigma), serum was collected, and then contaminant rabbit Abs were immunoabsorbed from the antiserum. For these purposes, antisera were first absorbed with dried BL21 cells powder, which was prepared by precipitation of BL21 cells with cold acetone for 15 min on ice (32Sambrook J. Fritsch E.F. Maniatis T. Molecular Cloning: A Laboratory Manual.2nd Ed. Cold Spring Harbor Laboratory, Cold Spring Harbor, NY1989: 852-861Google Scholar), and then with CHO cells, which had been fixed with 4% paraformaldehyde and permeabilized with 0.2% (v/v) Triton X-100. The IgG fraction of this absorbed antiserum was purified by an rProtein A FF column (Amersham Biosciences). The IgG fraction of preimmune serum was also purified and served as rabbit control IgG. Immunoblotting and Immunofluorescence Analysis—The whole coding region of LSECtin was initially obtained by RT-PCR on the fetal liver total RNA as described above. PCR products were cloned into pGEM-T vector (Promega) and sequenced. The entire coding sequence of LSECtin was subcloned in-frame into pcDNA3.1A vector (Invitrogen) to produce the construct allowing the expression of C-terminal tagged Myc-His fusion protein. CHO cells were transfected with 1 μg of plasmid using LipofectAMINE™ 2000 (Invitrogen) and selected for 10–14 days in presence of 800 μg/ml G418 (Invitrogen). Resistant populations were enriched for cells expressing LSECtin and maintained in Dulbecco's modified Eagle's medium, 10% FCS supplemented with 300 μg/ml G418. CHO cells stably expressing LSECtin (CHO-LSECtin) and CHO cells transfected with an empty vector (mock cells) were treated in a lysis buffer containing 20 m Tris-HCl, pH 8.0, 150 mm NaCl, 0.15% Triton X-100, 1 mm MgCl2, 2 mm CaCl2, and a mixture of protease inhibitors (Roche Applied Science). Samples were separated by 12% SDS-PAGE under reducing conditions or by 10% SDS-PAGE under nonreducing conditions and transferred onto polyvinylidene fluoride membranes (Millipore Corp.), which were blotted either with rabbit anti-LSECtin serum (1.5 μg/ml) or anti-Myc mAb (0.5 μg/ml). After an extensive wash, membranes were further blotted with horseradish peroxidase-conjugated goat anti-rabbit or anti-mouse IgG (Santa Cruz Biotechnology, Inc., Santa Cruz, CA) and developed with the ECL system (Amersham Biosciences). For detecting LSECtin protein in fetal liver, the membrane proteins were extracted from fetal liver tissue using an aqueous two-phase partitioning system according to previously reported methods (33Morre D.J. Morre D.M. BioTechniques. 1989; 7: 946-958Google Scholar). The membrane and cytosolic fractions and crude lysates of fetal liver tissues were separated by 12% SDS-PAGE under reduced conditions and then immunoblotted as described above. For immunofluorescence, CHO-LSECtin cultured on glass coverslips were fixed in 4% paraformaldehyde, blocked with 5% bovine serum albumin (BSA) for 2 h at 37 °C, and stained with rabbit anti-LSECtin (5 μg/ml), anti-Myc mAb (2.5 μg/ml), or preimmune rabbit IgG (5 μg/ml) according to the standard procedure (34Harlow E. Lane D. Antibodies: A Laboratory Manual. Cold Spring Harbor Laboratory, Cold Spring Harbor, NY1998: 72-79Google Scholar). All cell samples were viewed on an epifluorescence microscope (Olympus). For flow cytometric analysis, CHO-LSECtin and mock cells were freed from culture plates using 0.02% EDTA in phosphate-buffered saline. For staining, 2 × 105 cells were incubated in ice-cold phosphate-buffered saline containing 2% BSA, 0.02% sodium azide (FACS buffer), and rabbit anti-LSECtin (5 μg/ml) or anti-Myc mAb (2.5 μg/ml). After 30 min at 4 °C, the cells were washed with the FACS buffer and recovered in 100 μl of FACS buffer containing 2 μg of fluorescein isothiocyanate-conjugated antibody against rabbit or mouse IgG (Santa Cruz) per ml. Cells were incubated for 30 min at 4 °C, washed with FACS buffer, and analyzed by a FACSCalibur apparatus (Becton Dickinson). Immunohistochemistry—5-μm paraffin sections of normal human liver and lymph node tissues were obtained from Dr. Zhao at the Center of Cancer Medicine, Beijing. Rehydrated paraffin sections were pressure-cooked for 3 min in 10 mm sodium citrate buffer, pH 6.0. After washing, the sections were preblocked in 0.5% hydrogen peroxide for 30 min and then blocked in 10% normal goat serum for 2 h. Then the sections were incubated with rabbit anti-LSECtin serum (5 μg/ml), rabbit anti-von Willerbrand factor (1:50 dilution), and preimmunization rabbit IgG (5 μg/ml) for 60 min at 37 °C, respectively. Subsequently, sections were incubated with biotinylated goat anti-rabbit IgG (Santa Cruz Biotechnology) for 30 min and finally with streptavidin coupled to peroxidase (Santa Cruz Biotechnology) for 30 min. After washing, enzyme activity was developed using 3,3-diaminobenzidine (Santa Cruz Biotechnology), and the sections were counterstained with hematoxylin. Double immunofluorescence staining was performed on formalin-fixed liver and lymph node tissues using rabbit anti-LSECtin serum and goat anti-DC-SIGNR. Antigen unmasking was done as described above. After blocking in 1% bovine serum albumin, 10% house serum, 10% chicken serum, both primary antibodies (5 μg/ml) were added overnight at 4 °C. Sections were washed with TBS and incubated for 30 min at 37 °C either with red-conjugated chicken anti-goat antibody (Santa Cruz Biotechnology) or biotinylated horse anti-rabbit IgG antibody (Santa Cruz Biotechnology) detected with fluorescein isothiocyanate-conjugated streptavidin. The stained slides were analyzed by using a conventional fluorescence microscope (Olympus). Preparation of LSECtin-Fc Fusion Protein—Recombinant chimeras containing the entire extracellular region of LSECtin fused to the Fc region of human IgG1 (LSECtin-Fc) were generated by PCR using fetal liver cDNA. The forward and reverse primers were used (5′-GC AAG CTT AAG GCC TCC ACG GAG CGC-3′ and 5′-GGC TCT AGA GCA GTT GTG CCT TTT CTC-3′). The PCR product was cloned in-frame downstream of the signal sequence of CD33 in Signal pIgplus vector (35Freeman S.D. Kelm S. Barber E.K. Crocker P.R. Blood. 1995; 85: 2005-2012Google Scholar). Plasmids were transfected into CHO cells by LipofectAMINE™ 2000. The cells stably expressing LSECtin-Fc were selected by G418 as described above. After 5 days of incubation in a non-protein-containing CD CHO medium (Invitrogen) with 300 μg/ml G418, the culture supernatant was harvested, followed by affinity chromatography with rProtein A FF column, and then examined by immunoblotting with anti-Fc mAb (Sigma). Sugar Binding Assays—Sugar binding assays were performed as previously reported (36East L. Rushton S. Taylor M.E. Isacke C.M. J. Biol. Chem. 2002; 277: 50469-50475Google Scholar). For LSECtin-Fc chimeras, 200 μl of CHO tissue culture supernatant in 800 μl of loading buffer (150 mm NaCl, 25 mm Tris-HCl pH 8.0, 25 mm CaCl2, and 1 mm MgCl2) was loaded onto 1-ml columns of mannose-, GlcNAc-, fucose-, or galactose-agarose (Sigma). The flow-through was collected, and the columns were washed with 8 × 1 ml of loading buffer followed by 8 × 1 ml of elution buffer (150 mm NaCl, 25 mm Tris-HCl, pH 8.0, and 10 mm EDTA). For CHOLSECtin cells, cells were lysed in detergent extraction solution (150 mm NaCl, 10 mm Tris-HCl, pH 8.0, 0.15% Triton X-100, 1 mm MgCl2, and 10 mm CaCl2). Lysate was incubated on ice for 30 min before centrifugation at 4 °C and 14,000 × g, and the supernatant was collected. 400 μl of supernatant was added to 600 μl of loading buffer (150 mm NaCl, 25 mm Tris-HCl, pH 8.0, 25 mm CaCl2, 1 mm MgCl2, and 0.15% Triton X-100) and applied to the columns. The columns were washed in loading buffer and eluted in elution buffer (150 mm NaCl, 25 mm Tris-HCl, pH 8.0, 0.15% Triton X-100, and 10 mm EDTA). Fractions were precipitated by incubation with 20 μg of BSA and 0.5 ml of 30% trichloroacetic acid for 30 min on ice and then centrifuged for 30 min at 4 °C, 15,000 × g. The pellets were washed twice in 1:1 ethanol ether, air-dried, and resuspended in 40 μl of reducing sample buffer. Samples were resolved by 12% SDS-PAGE and analyzed by Western blotting with anti-LSECtin antibody. Sugar Competition Assays—First, Man30-BSA (E.-Y. Laboratories) was labeled with biotin using EZ-Link TFP-PEO-Biotin (Pierce) according to the protocol recommended by the manufacturer. Purified sLSEC-tin-Fc fusion proteins were immobilized on enzyme-linked immunosorbent assay plates (NUNC, Denmark) by overnight incubation of 200 μg/ml stock solution at 4 °C (50 μl/well). After blocking of the wells usi
What problem does this paper attempt to address?
-
Characterization Of A Novel C-Type Lectin-Like Gene, Lsectin - Demonstration Of Carbohydrate Node
Wanli Liu,Li Tang,Ge Zhang,Handong Wei,Yufang Cui,Lihai Guo,Zikuan Gou,Xiaoxiao Chen,Daifeng Jiang,Yunping Zhu,Gefei Kang,Fuchu He
DOI: https://doi.org/10.1074/jbc.M311227200
IF: 5.485
2004-01-01
Journal of Biological Chemistry
Abstract:A new C-type lectin-like gene encodes 293 amino acids and maps to chromosome 19p13.3 adjacent to the previously described C-type lectin genes, CD23, dendritic cell-specific intercellular adhesion molecule-3-grabbing nonintegrin (DC-SIGN), and DC-SIGN-related protein (DC-SIGNR). The four genes form a tight cluster in an insert size of 105 kb and have analogous genomic structures. The new C-type lectin-like molecule, designated liver and lymph node sinusoidal endothelial cell C-type lectin (LSECtin), is a type II integral membrane protein of similar to 40 kDa in size with a single C-type lectin-like domain at the COOH terminus, closest in homology to DC-SIGNR, DC-SIGN, and CD23. LSECtin mRNA was only expressed in liver and lymph node among 15 human tissues tested, intriguingly neither expressed on hematopoietic cell lines nor on monocyte-derived dendritic cells (DCs). Moreover, LSECtin is expressed predominantly by sinusoidal endothelial cells of human liver and lymph node and co-expressed with DC-SIGNR. LSECtin binds to mannose, GlcNAc, and fucose in a Ca2+-dependent manner but not to galactose. Our results indicate that LSECtin is a novel member of a family of proteins comprising CD23, DC-SIGN, and DC-SIGNR and might function in vivo as a lectin receptor.
-
The Dc-Sign Family Member Lsectin is A Novel Ligand of Cd44 on Activated T Cells
Li Tang,Juntao Yang,Xiaoming Tang,Wantao Ying,Xiaohong Qian,Fuchu He
DOI: https://doi.org/10.1002/eji.200939936
2010-01-01
European Journal of Immunology
Abstract:LSECtin, a novel member of the C-type lectin DC-SIGN family, not only acts as an attachment factor for pathogens, but also recognizes "endogenous" activated T cells. The endogenous ligands of LSECtin, however, have remained unclear. In this study, we identified CD44 on Jurkat T cells as a candidate ligand of LSECtin, and confirmed the specific interaction between LSECtin and CD44. Moreover, we showed that LSECtin selectively bound CD44s, CD44v4 and CD44v8-10 by screening a series of typical CD44 isoforms. By deletion of the carbohydrate-recognition domain region and mutation of crucial amino acids involved in carbohydrate-recognition of LSECtin and by inhibition of the N-linked glycosylation of CD44, we further demonstrated that the interaction between CD44 and LSECtin is dependent on protein-glycan recognition. Our findings indicate that CD44 is the first identified endogenous ligand of LSECtin, and similarly, that LSECtin is a novel ligand of CD44. These findings provide important new perspectives on the biology of both LSECtin and CD44 in the immune system.
-
The human and mouse dendritic cell receptor DCIR binds to LRP1 N-glycans containing terminal galactoses, including the α-Gal antigen
Benjamin B.A. Raymond,Tamara Sneperger,Stella Rousset,Nelly Gilles,Aurélie Bally,Alexandre Stella,Agnes L. Hipgrave Ederveen,Julien Marcoux,Giulia Trimaglio,Michel Thépaut,Frederic Lagarrigue,Odile Burlet-Schiltz,Manfred Wuhrer,Franck Fieschi,Pascal Demange,Olivier Neyrolles,Yoann Rombouts
DOI: https://doi.org/10.1101/2024.05.20.594848
2024-05-21
Abstract:The dendritic cell immunoreceptor (DCIR) is a C-type lectin receptor expressed by myeloid cells that plays a key immunoregulatory role in a wide range of diseases, from inflammation to cancer. However, the ligand(s) of DCIR remain(s) unknown, hampering our understanding of the exact function of this immune receptor. Here, we found that both human DCIR and mouse DCIR1 bind specifically to the low-density lipoprotein receptor-related protein 1 (LRP1), a heavily glycosylated receptor mediating the clearance of various molecules from the extracellular matrix and apoptotic cells. This interaction is mediated by galactose-terminated biantennary complex-type N-glycans, including those carrying the immunogenic α-Gal epitope. Our study provides a deeper understanding of the role of DCIR in immune regulation and its potential impact on a range of immune disorders, highlighting its specificity in ligand recognition which is crucial for developing therapeutic strategies.
Biology
-
A Cellular Model for the Expression of the C-type Lectin Dendritic Cell-Specific Intercellular Adhesion Molecule-3-grabbing Non-Integrin: Construction and Functional Analysis
Zhang Yu,Yao Xu,Gu Hanyan,Wang Baoxi,Liu Jun
DOI: https://doi.org/10.3760/cma.j.issn.0412-4030.2014.07.002
2014-01-01
Chinese Journal of Dermatology
Abstract:Objective To establish a cellular model for the expression of the C-type lectin dendritic cellspecific intercellular adhesion molecule-3-grabbing non-integrin (DC-SIGN),and to provide a basis for the functional analysis of DC-SIGN.Methods The cDNA of DC-SIGN was obtained via PCR,and cloned into the eukaryotic expression vector porcine cytomegalovirus-enhanced green fluorescent protein (PCMV-EGFP) with EGFP at the N terminal of DC-SIGN.Then,the recombinant PCMV-EGFP-DC-SIGN plasmid was transfected into HEK293T cells followed by the detection of DC-SIGN expression using PCR,Western blot and flow cytometry.Confocal microscopy was performed to localize the expression of DC-SIGN-EGFP and visualize the recognization and internalization of the Derp2 allergen by DC-SIGN.Results The recombinant fluorescent fusion protein-expressing plasmid was successfully constructed.Both PCR and Western blot confirmed the expression of DC-SIGN.Flow cytometry showed that the expression of DC-SIGN was increased by approximately 50% in HEK293T cells transfected with the recombinant expression plasmid compared with those untransfected.As confocal microscopy showed,the green fluorescence-labelled DC-SIGN was located on the cell membrane,which could bind to the red fluorescence-labelled antigen Derp2 and internalize it into the cells.Conclusions The recombinant DC-SIGN-EGFP fusion protein is characteristically located on the surface of 293T cells,which can be recognized by the DC-SIGN-specific antibody and is capable of internalizing the allergen Derp2,and may serve as an ideal cell model for further studies on molecular function of DC-SIGN.
-
Regulation Role of C-type Lectin-Like Receptor DC-SIGN in Immune Responses
WANG Jingxue,LI Hong,HE Lixiong,WU Yuzhang
DOI: https://doi.org/10.13431/j.cnki.immunol.j.20100220
2010-01-01
Abstract:DC-SIGN is one of lectin-like receptors specially expressed on dendritic cells,which can recognize and capture many kinds of glycosylated antigens.It is suggested that different carbohydrate ligands and different pathogen maybe result in different immune responses via interaction with DC-SIGN,according to reported research works.In the present paper,we reviewed immunophenotypic and signal difference after DC-SIGN interacted with different carbohydrate types.
-
Identification of a novel type I cytokine receptor CRL2 preferentially expressed by human dendritic cells and activated monocytes
W Zhang,J Wang,Q Wang,G Chen,J Zhang,T Chen,T Wan,Y Zhang,X Cao
DOI: https://doi.org/10.1006/bbrc.2001.4432
2001-03-09
Abstract:From a human dendritic cell (DC) cDNA library, we identified a novel type I cytokine receptor, designated as cytokine receptor-like molecule 2 (CRL2). CRL2 cDNA encoded a 371-residue type I transmembrane protein with an extracellular domain of 210 residues and an intracellular domain of 119 residues. Its extracellular domain contains conserved cysteine residues and WAS-like motif in place of the hallmark of WSXWS motif present in other type I cytokine receptors. The intracellular domain contained a membrane-proximal "box 1" motif and conserved tyrosine residue potentially as a binding site for signal transducing molecules. CRL2 protein shares significant homology with common cytokine receptor (gammac) and interleukin-13 receptor alpha1 chain. Northern blot analysis showed that CRL2 was restrictedly expressed by spleen and peripheral blood leukocytes, and abundantly expressed by HL-60 cells. RT-PCR analysis demonstrated that CRL2 was preferentially expressed by DC and monocytes. Interestingly, CRL2 expression was up-regulated when monocytes were activated by LPS. These indicate that CRL2 may be involved in the biological functions of DC and monocytes. The Ba/F3 transfectants of CRL2 was retrovirally established with the expressed FLAG-tagged CRL2 in the size of approximately 48 kD, which could be efficiently immunoprecipitated. We also prepared a CRL2Ig fusion protein. The identification of its ligand and involvement of signal transduction will help to elucidate its potential function.
-
DC-SIGN and L-SIGN Are High Affinity Binding Receptors for Hepatitis C Virus Glycoprotein E2*
Py Lozach,H Lortat-Jacob,Ad De Lavalette,I Staropoli,S Foung,A Amara,C Houles,F Fieschi,O Schwartz,Jl Virelizier,F Arenzana-Seisdedos,R Altmeyer
DOI: https://doi.org/10.1074/jbc.m301284200
IF: 5.485
2003-01-01
Journal of Biological Chemistry
Abstract:The hepatitis C virus (HCV) genome codes for highly mannosylated envelope proteins, which are naturally retained in the endoplasmic reticulum. We found that the HCV envelope glycoprotein E2 binds the dendritic cell-specific intercellular adhesion molecule 3-grabbing nonintegrin (DC-SIGN) and the related liver endothelial cell lectin L-SIGN through high-mannose N-glycans. Competing ligands such as mannan and an antibody directed against the carbohydrate recognition domains (CRD) abrogated binding. While no E2 interaction with distant monomeric CRDs on biosensor chips could be detected, binding is observed if CRDs are closely seeded (Kd = 48 nm) and if the CRD is part of the oligomeric-soluble extracellular domain of DC-SIGN (Kd = 30 nm). The highest affinity is seen for plasma membrane-expressed DC-SIGN and L-SIGN (Kd = 3 and 6 nm, respectively). These results indicate that several high-mannose N-glycans in a structurally defined cluster on E2 bind to several subunits of the oligomeric lectin CRD. High affinity interaction of viral glycoproteins with oligomeric lectins might represent a strategy by which HCV targets to and concentrates in the liver and infects dendritic cells.
-
Cloning and Characterization of Siglec-10, a Novel Sialic Acid Binding Member of the Ig Superfamily, from Human Dendritic Cells.
N Li,WP Zhang,T Wan,J Zhang,TY Chen,YZ Yu,JL Wang,XT Cao
DOI: https://doi.org/10.1074/jbc.m100467200
IF: 5.485
2001-01-01
Journal of Biological Chemistry
Abstract:The Siglecs (sialic acid-binding Ig-like lectins) are a subfamily of I-type lectins, which specifically recognize sialic acids. Nine members of the family have been identified thus far. We have obtained a novel cDNA clone from a human dendritic cell cDNA library encoding a protein with sequence and structural features of the Siglec family, hence designated as Siglec-10. The full-length Siglec-10 cDNA encodes a type 1 transmembrane protein containing four extracellular immunoglobulin-like domains, a transmembrane region, and a cytoplasmic tail with two classical immunoreceptor tyrosine-based inhibitory motifs. The N-terminal V-set Ig domain has most of the amino acid residues typical of the Siglecs. Siglec-10 shows the closest homology to Siglec-5 and Siglec-3/CD33. Various cells and cell lines including monocytes and dendritic cells express Siglec-10. High levels of mRNA expression were seen in peripheral blood leukocytes, spleen, and liver. When expressed on COS-7 cells, Siglec-10 was able to bind human red blood cells and soluble sialoglycoconjugates in a sialic acid-dependent manner. The identification of Siglec-10 as a new Siglec family member and its expression profile, together with its sialic acid-dependent binding capacity, suggest that it may be involved in cell-cell recognition by interacting with sialylated ligands expressed on specific cell populations.
-
Variations of Dendritic Cell-Specific Intercellualar Adhesion Molecule-3-grabing Nonintegrin Neck Region in HIV Infected Individuals.
Xu Li-Jun,Yao Hang-Ping,Li Dan,Wang Zhi-Gang,Chen Liang,Wu Nan-Ping
DOI: https://doi.org/10.1097/00029330-200802010-00017
2008-01-01
Abstract:Dendritic cells (DCs) play a critical role in initiating the immune response by virtue of their ability to capture and present antigens to T cells.1 Although the precise mechanism by which DCs acquire human immunodeficiency virus (HIV-1) is not completely understood, migration of DCs from the periphery to the draining lymph nodes may enable CD4+ T cells to become infected.2 DC-specific intercellular adhesion molecule 3 grabbing nonintegrin (DC-SIGN, CD209), a mannose specific C-type lectin receptor on DCs, plays a vital role in this process by binding HIV-gp120 and helping DCs transport HIV from the infection site to the secondary lymph nodes.3 DC-SIGN related lectin (DC-SIGNR, or L-SIGN, CD209R) shares 77% amino acid identity with DC-SIGN, and is expressed on endothelial cells in the liver, lymph nodes and placental capillaries.4 Both DC-SIGN and DC-SIGNR are HIV receptors.5 Peptide sequence and structural analyses indicate that DC-SIGN consists of four regions: carbohydrate recognition domain (CRD), consisting of 110 to 140 amino acid residues; neck region (also named repeat region), normally including seven 23-amino acid residue tandem repeat alleles; transmembrane region; and an endocellular domain. The CRD and neck regions have the greatest influence on pathogen infection. CRD is necessary for binding and transmission of HIV-1 and other pathogens. The neck region forms a tetramer which stabilizes the DC-SIGN structure. So the number changes of repeat alleles may have a potential impact on the function of CRD.6,7 Recently, associations between host gene polymorphisms and HIV infection have been investigated. A study of 1716 American individuals suggests that heterozygous 7/5R DC-SIGNR tend to decrease the risk of HIV infection;8 however this conclusion is not supported by the investigation of a German population conducted by Lichterfels et al,9 whose research indicates the polymorphisms in the DC-SIGNR neck region does not effect HIV infection and HIV/AIDS progress. Usually, polymorphisms of host genes are related with ethnic background and heredity. In fact, the distribution of DC-SIGN differs widely in populations from industrialized and developing countries as well as between different ethnic populations. The reasons for this diversity are presumably related to geographically determined selection pressures and long-term selected pressures of pathogens.10,11 Little is known about DC-SIGN variations and HIV infection in Chinese populations. For the purpose of exploring this association in our present study we investigate a potential correlation between variations of the DC-SIGN neck region and susceptibility to HIV in Chinese Han population. METHODS Samples Samples from one hundred and nineteen HIV-1 seropositive Chinese patients were collected during 2004 to 2006 from Chinese Center for Disease Control and Prevention (Beijing), Hangzhou Infectious Hospital, Qingchun Hospital (Hangzhou) and the First Affiliated Hospital of Zhejiang University. All HIV-1 infected subjects were HIV-1 primary infected individuals with an average age of (42±10) years old and did not receive any anti-virus therapies. One hundred and twenty HIV-1 seronegative individuals were sampled as controls. They were outpatients in the First Affiliated Hospital of Zhejiang University for routine health examination and were unrelated to the HIV infected individuals. The average age of the controls was (40±15) years old. All the subjects were males from the Han population except for one female. With the subjects' authorization, five milliliter of blood was taken from a vein and preserved in EDTA-anticoagulant tubes. Isolation of DC-SIGN neck region Peripheral blood mononuclear cells (PBMCs) were isolated from the blood samples by density gradient centrifugation and washed twice with PBS. PBMC was treated with Trizol (Invitrogen, USA) for total RNA extraction according to standard protocol. The total RNA was resuspended in 50 μl DEPC-treated deionized water, then preserved at -80°C for further use. The DC-SIGN repeat region was analyzed by RT-PCR. We amplified the DC-SIGN repeat region with a sense primer 5′-AACAATCCAGGCAAGACG-3 and an antisense primer 5′-TGCTCAGGCAGGGTCAGT-3′. PCR products were analyzed by 2% agarose gel electrophoresis. The products were then purified with a PCR purification kit (Takara, Japan), cloned into a PMD-18T vector (Takara, Japan) by a T-A reaction, followed by a two-way sequencing. For the heterozygous DC-SIGN, every DNA strip in the gel was separated by gel purification (Takara, Japan) and sequenced. The numbers of repeat allele within DC-SIGN neck region were determined based on the results of agarose gel electrophoresis and nucleotide sequences. Statistical analysis The two-sided Pearson χ2 test was used to determine whether genotypes or repeat alleles of DC-SIGN had differential discrepancies between HIV infected individuals and the controls. Heterozygous and homozygous DC-SIGN were categorized into two groups respectively for analysis. A 2×2 contingency table was used to examine whether the distributions of homozygous/heterozygous genotypes were independent. Yates' continuity correction was used when necessary. A P value less than 0.05 was considered statistically significant. All analysis was performed using the software SPSS11.5. RESULTS Variations of DC-SIGN neck region The numbers and frequencies of DC-SIGN variations among HIV infected individuals and controls are summarized in Table 1. The percentage of 7/7 repeat allele (7/7R) genotype was 91.41% in HIV infected Chinese, which was significantly lower than the frequencies among control individuals (91.41% vs 97.50%, P=0.038). The dominance of the 7/7R genotype among the HIV-1 seropositive and control patients suggested that the 7/7R genotype was the wild type gene in the population. In order to analyze whether heterozygous DC-SIGN reduced the susceptibility to HIV infection or not, homozygous genotypes (8/8R, 7/7R, 6/6R, 4/R and 3/3R) and heterozygous genotypes (8/7R, 8/6R, 7/6R and 7/5R) present in the Chinese population were categorized into two groups as in Table 2. There were no significant discrepancies of genotypes for DC-SIGN between the two groups (P=0.561).Table 1: Distributions of genotypes of DC-SIGN repeat alleles (n (%))Table 2: Distributions of heterozygous and homozygous DC-SIGNGene sketch map for DC-SIGN neck region Though homozygous 7/7R was the dominant genotype in our research subjects although many different DC-SIGN allelic combinations were found in our research. In order to illustrate the missing fragments of each repeat region, and to identify the genotype of repeat alleles within the neck region, we draw a sketch map of the DC-SIGN gene, as described previously.6,7 The map shows that the first, second, and eighth repeat alleles were conserved among all samples and were rarely absent (Figure).Figure.: Sketch map of repeat units within repeat region. The first, second, and eighth repeat units are conserved in all samples.DISCUSSION The associations of D-SIGN/R variation and susceptibility to HIV infection have been studied in recent years. These studies showed that the variations in the DC-SIGN promoter region had potential effects on pathogen infection. For example, individuals with the -336C variant are more susceptible to HIV infection than individuals with the -336T variant.12 A study including 316 HIV-1-seropositive and 425 HIV-1-seronegative individuals suggest that heterozygous DC-SIGN reduces the risk of HIV-1 infection.13 This conclusion is draw from the fact that the occurrence of heterozygous DC-SIGN is 0 in HIV-seropositive populations, but 3.2% in the HIV-1 seronegative individuals. Another important factor is that DC-SIGN translated from mRNA rather than from DNA may have great significance on HIV infection. Different DC-SIGN structure from different recombination of DC-SIGN mRNA may directly interact with HIV and could have a potential effect on HIV infection. In our present study, we determined variants of the DC-SIGN neck region from HIV-1 infected and uninfected individuals, respectively. We show that variations within DC-SIGN are significantly more common in the HIV infected cohort than in the control group. Differing from previous report that heterozygous DC-SIGN is not available in HIV-1(+) individuals,13 our date suggest heterozygous DC-SIGN in the Chinese Han population is detectable and heterozygous DC-SIGN does not have an impact on HIV infection. The reasons for the variations of DC-SIGN detected in HIV-1(+) individuals might be as follows. First, interleukin-4 (IL-4) is a pivotal regulator for DC-SIGN expression in in vitro experiments, and is responsible for allelic DC-SIGN mRNA scission, recombination and rearrangement.7,14 One character of HIV infection is gradual T lymphocyte subpopulation switching from a Th1 population to a Th2 population. In vivo the Th1 population mainly secrets interferon γ (INF-γ) not IL-4, whereas, Th2 population secret mainly IL-4 but not INF-γ. So variation of DC-SIGN in HIV infected people maybe result from a variety of levels of IL-4 secreted by Th2 cells at different stages of HIV infection. Second, mutations of IL-4 receptor (IL-4R) are closely associated with susceptibility to HIV-1 infection and its progression to AIDS.20 So the mutation of the IL-4R maybe also be a factor associated with DC-SIGN variations. Third, because the genotype of DC-SIGN does not impact HIV infection the variations of DC-SIGN that appeared in HIV infected persons is the result from HIV infection, not the result of HIV infection of individuals with different DC-SIGN. It should be pointed that further research is urgently required to explore potential the significance for the presence of these mutations in the HIV-1 seropositive populations. For example, whether variations of DC-SIGN are related to HIV/AIDS progress or not will require a detailed study. Our results provide a starting point to understanding why mutation rates in DC-SIGN are increased in HIV infected individuals. In summary, our data reveal that there are more genotypes of DC-SIGN in HIV-infected individuals than in non-infected people, and heterozygous DC-SIGN does not reduce the risk to HIV infection in Chinese Han population.
-
L-C1qDC-1, a Novel C1q Domain-Containing Protein from Lethenteron Camtschaticum That is Involved in the Immune Response.
Guangying Pei,Ge Liu,Xiong Pan,Yue Pang,Qingwei Li
DOI: https://doi.org/10.1016/j.dci.2015.08.011
IF: 3.605
2016-01-01
Developmental & Comparative Immunology
Abstract:The C1q domain-containing (C1qDC) proteins are a family of proteins characterized by a globular C1q (gC1q) domain at their C-terminus. These proteins are involved in various processes in vertebrates and are assumed to serve as important pattern recognition receptors in innate immunity in invertebrates. Here, a novel C1qDC protein from Lethenteron camtschaticum was identified and characterized (designated as L-C1qDC-1). After a partial cDNA sequence of L-C1qDC-1 was identified in a L. camtschaticum liver cDNA library, the full-length cDNA was obtained using 3'- and 5'-rapid amplification of cDNA ends (RACE). L-C1qDC-1 encodes 236 amino acids and contains a signal peptide, a collagen-like sequence with Gly-Xaa-Yaa repeats, and a C-terminal gC1q domain. The L-C1qDC-1 protein was primarily distributed in the gut, liver and supraneural body of L. camtschaticum and was also marginally detectable in leukocytes via real-time PCR and immunofluorescence assays. Furthermore, both immunoprecipitation and immunofluorescence results showed that in L camtschaticum serum, L-C1qDC-1 could interact with variable lymphocyte receptor (VLR) B and displayed strong colocalization with cancer cell immune responses. These results indicated that the L-C1qDC-1 gene encodes a novel C1qDC protein that may play an important role in the immune responses of L. camtschaticum, providing clues for understanding the universal functions of C1qDC proteins in other species and suggesting that these proteins could serve as pattern recognition molecules in immunotherapy. (C) 2015 Elsevier Ltd. All rights reserved.
-
A novel C-type lectin with two CRD domains from Chinese shrimp Fenneropenaeus chinensis functions as a pattern recognition protein.
Xiao-Wen Zhang,Wen-Teng Xu,Xian-Wei Wang,Yi Mu,Xiao-Fan Zhao,Xiao-Qiang Yu,Jin-Xing Wang
DOI: https://doi.org/10.1016/j.molimm.2009.02.029
IF: 4.174
2009-01-01
Molecular Immunology
Abstract:Lectins are regarded as potential immune recognition proteins. In this study, a novel C-type lectin (Fc-Lec2) was cloned from the hepatopancreas of Chinese shrimp, Fenneropenaeus chinensis. The cDNA of Fc-Lec2 is 1219bp with an open reading frame (ORF) of 1002bp that encodes a protein of 333 amino acids. Fc-Lec2 contains a signal peptide and two different carbohydrate recognition domains (CRDs) arranged in tandem. The first CRD contains a QPD (Gln-Pro-Asp) motif that has a predicted binding specificity for galactose and the second CRD contains a EPN (Glu-Pro-Asn) motif for mannose. Fc-Lec2 was constitutively expressed in the hepatopancreas of normal shrimp, and its expression was up-regulated in the hepatopancreas of shrimp challenged with bacteria or viruses. Recombinant mature Fc-Lec2 and its two individual CRDs (CRD1 and 2) did not have hemagglutinating activity against animal red blood cells, but agglutinated some Gram-positive and Gram-negative bacteria in a calcium-dependent manner. The three recombinant proteins also bound to bacteria in the absence of calcium. Fc-Lec2 seems to have broader specificity and higher affinity for bacteria and polysaccharides (peptidoglycan, lipoteichoic acid and lipopolysaccharide) than each of the two individual CRDs. These data suggest that the two CRDs have synergistic effect, and the intact lectin may be more effective in response to bacterial infection, the Fc-Lec2 performs its pattern recognition function by binding to polysaccharides of pathogen cells.
-
DC-SIGN: escape mechanism for pathogens
Yvette van Kooyk,Teunis B. H. Geijtenbeek
DOI: https://doi.org/10.1038/nri1182
IF: 108.555
2003-09-01
Nature Reviews Immunology
Abstract:Key PointsDendritic cells (DCs) express Toll-like receptors (TLRs) and C-type lectins that interact with pathogens.C-type lectins interact with specific carbohydrate structures on pathogens to internalize pathogens for degradation in lysosomal compartments to enhance antigen processing and presentation.TLRs recognize molecular patterns on pathogens that leads to DC activation, expression of co-stimulatory molecules and the production of inflammatory cytokines.Different DC-subsets express different sets of TLRs and C-type lectins to orchestrate specific immune responses.C-type lectins recognize self and non-self antigens, and can function as antigen receptors, adhesion receptors and signalling receptors. So far, only little is known about the antigen specificity of C-type lectins with the exception of the DC-specific C-type lectin DC-SIGN.Cell- and tissue-specific glycosylation of a glycoprotein regulates the expression of carbohydrate residues and recognition by specific C-type lectins.The C-type lectin DC-SIGN recognizes a wide variety of pathogens such as viruses, bacteria, yeast and parasites through the binding of mannose or Lewis-x carbohydrate structures.DC-SIGN also functions as an adhesion receptor through binding of intercellular adhesion molecule 2 (ICAM2) and ICAM3.Pathogens such as HIV-1 and Mycobacteria tuberculosis target DC-SIGN to escape immune surveillance.HIV-1 targets DC-SIGN to hide in DCs and for efficient transmission to T cells, as DC-SIGN functions as a trans-receptor that efficiently presents infectious virus to target T cells.Mycobacteria tuberculosis subverts DCs by targeting DC-SIGN, leading to altered signalling events that inhibit TLR signalling and suppress DC maturation.The balance between TLRs and C-type lectins triggering by pathogens might be instrumental in the final immune response — immune activation or immune suppression.
immunology
-
Molecular recognition of Escherichia coli R1-type core lipooligosaccharide by DC-SIGN
Ferran Nieto-Fabregat,Angela Marseglia,Michel Thépaut,Jean-Philippe Kleman,Massilia Abbas,Aline Le Roy,Christine Ebel,Meriem Maalej,Jean-Pierre Simorre,Cedric Laguri,Antonio Molinaro,Alba Silipo,Franck Fieschi,Roberta Marchetti
DOI: https://doi.org/10.1016/j.isci.2024.108792
IF: 5.8
2024-01-04
iScience
Abstract:Due to their ability to recognize carbohydrate structures, lectins emerged as potential receptors for bacterial lipopolysaccharides (LPS). Despite growing interest in investigating the association between host receptor lectins and exogenous glycan ligands, the molecular mechanisms underlying bacterial recognition by human lectins are still not fully understood. We contributed to fill this gap by unveiling the molecular basis of the interaction between the lipooligosaccharide of Escherichia coli and the dendritic cell-specific intracellular adhesion molecules (ICAM)-3 grabbing non-integrin (DC-SIGN). Specifically, a combination of different techniques, including fluorescence microscopy, surface plasmon resonance, NMR spectroscopy, and computational studies, demonstrated that DC-SIGN binds to the purified deacylated R1 lipooligosaccharide mainly through the recognition of its outer core pentasaccharide, which acts as a crosslinker between two different tetrameric units of DC-SIGN. Our results contribute to a better understanding of DC-SIGN-LPS interaction and may support the development of pharmacological and immunostimulatory strategies for bacterial infections, prevention, and therapy.
-
Siglecs: sialic-acid-binding immunoglobulin-like lectins in cell–cell interactions and signalling
Paul R Crocker,P Crocker
DOI: https://doi.org/10.1016/s0959-440x(02)00375-5
IF: 7.786
2002-10-01
Current Opinion in Structural Biology
Abstract:Siglecs are sialic-acid-binding immunoglobulin-like lectins involved in cell-cell interactions and signalling functions in the haemopoietic, immune and nervous systems. Significant advances have been made in our understanding of the link between carbohydrate recognition and signalling for two well-characterised siglecs, CD22 and myelin-associated glycoprotein. Over the past few years, several novel siglecs have been discovered through genomics and functional screens. These "CD33-related" siglecs have molecular features of inhibitory receptors and may be important in regulating leucocyte activation during immune and inflammatory responses.
cell biology,biochemistry & molecular biology
-
Secondary sites of the C‐type lectin‐like fold
Jonathan Lefebre,Torben Falk,Yunzhan Ning,Christoph Rademacher
DOI: https://doi.org/10.1002/chem.202400660
2024-03-27
Chemistry - A European Journal
Abstract:C‐type lectins are a large superfamily of proteins involved in a multitude of biological processes. In particular, their involvement in immunity and homeostasis has rendered them attractive targets for diverse therapeutic interventions. They share a characteristic C‐type lectin‐like domain whose adaptability enables them to bind a broad spectrum of ligands beyond the originally defined canonical Ca2+‐dependent carbohydrate binding. Together with variable domain architecture and high‐level conformational plasticity, this enables C‐type lectins to meet diverse functional demands. Secondary sites provide another layer of regulation and are often intricately linked to functional diversity. Located remote from the canonical primary binding site, secondary sites can accommodate ligands with other physicochemical properties and alter protein dynamics, thus enhancing selectivity and enabling fine‐tuning of the biological response. In this review, we outline the structural determinants allowing C‐type lectins to perform a large variety of tasks and to accommodate the ligands associated with it. Using the six well‐characterized Ca2+‐dependent and Ca2+‐independent C‐type lectin receptors DC‐SIGN, langerin, MGL, dectin‐1, CLEC‐2 and NKG2D as examples, we focus on the characteristics of non‐canonical interactions and secondary sites and their potential use in drug discovery endeavors.
chemistry, multidisciplinary
-
IgSF13, a novel human inhibitory receptor of the immunoglobulin superfamily, is preferentially expressed in dendritic cells and monocytes
Lili Sui,Nan Li,Qiuyan Liu,Weiping Zhang,Tao Wan,Baomei Wang,Kun Luo,Hongying Sun,Xuetao Cao
DOI: https://doi.org/10.1016/j.bbrc.2004.05.065
IF: 3.1
2005-01-01
Biochemical and Biophysical Research Communications
Abstract:A novel inhibitory receptor of immunoglobin superfamily (IgSF), IgSF member 13 (IgSF13), has been identified from human dendritic cells (DC). IgSF13 is a type I transmembrane protein containing an N-terminal signal peptide, a extracellular region with a single Ig V-like domain, a transmembrane region, and a cytoplasmic tail with two classical immunoreceptor tyrosine-based inhibitory motifs (ITIM), suggesting its inhibitory function. IgSF13 shows significant homology to human CMRF35 and pIgR. IgSF13 gene is mapped to chromosome 17q25.2, very close to that of CMRF35. IgSF13 is preferentially expressed in myelo-monocytic cells, including monocytes, monocyte-derived DC, and monocyte-related cell lines. Upon pervanadate treatment, IgSF13 was hyper-phosphorylated and associated with Src homology-2 domain-containing phosphatases SHP-1 and SHIP, but not SHP-2. The identification of IgSF13 as a novel ITIM-bearing receptor selectively expressed by DC and monocytes suggests that it may be potentially involved in the negative regulation of specific leukocyte population.
-
C-type Lectin LSECtin Interacts with DC-SIGNR and is Involved in Hepatitis C Virus Binding
Yi Li,Bingtao Hao,Xuezhang Kuai,Guichun Xing,Juntao Yang,Jie Chen,Li Tang,Lingqiang Zhang,Fuchu He
DOI: https://doi.org/10.1007/s11010-009-0056-y
IF: 3.842
2009-01-01
Molecular and Cellular Biochemistry
Abstract:Hepatitis C virus (HCV) is a major cause of liver disease. However, the detailed mechanism underlying hepatocyte infection with HCV is not yet completely understood. We previously identified a novel C-type lectin--LSECtin predominantly expressed on liver sinusoidal endothelial cells. Here we demonstrate that LSECtin can interact with two HCV receptors, DC-SIGNR and CD81, through its central ectodomain. Furthermore, cells expressing LSECtin specifically can be attached by the naturally occurring HCV in the sera of infected individuals. This binding was found to be mediated by the HCV E2 glycoprotein and could be efficiently inhibited by EGTA but not by mannan treatment. The present study suggests that LSECtin interaction with DC-SIGNR might contribute to HCV binding to liver sinusoidal endothelial cells.
-
An ancient C-type lectin in Chlamys farreri (CfLec-2) that mediate pathogen recognition and cellular adhesion.
Jialong Yang,Limei Qiu,Xiumei Wei,Leilei Wang,Lingling Wang,Zhi Zhou,Huan Zhang,Lin Liu,Linsheng Song
DOI: https://doi.org/10.1016/j.dci.2010.07.004
IF: 3.605
2010-01-01
Developmental & Comparative Immunology
Abstract:C-type lectins are a superfamily of Ca2+ dependent carbohydrate-recognition proteins which play significant diverse roles in nonself-recognition and clearance of invaders. In the present study, a C-type lectin (CfLec-2) from Zhikong scallop Chlamys farreri was selected to investigate its functions in innate immunity. The mRNA expression of CfLec-2 in hemocytes was significantly up-regulated (P<0.01) after scallops were stimulated by LPS, PGN or β-glucan, and reached the highest expression level at 12h post-stimulation, which was 72.5-, 23.6- or 43.8-fold compared with blank group, respectively. The recombinant CfLec-2 (designated as rCfLec-2) could bind LPS, PGN, mannan and zymosan in vitro, but it could not bind β-glucan. Immunofluorescence assay with polyclonal antibody specific for CfLec-2 revealed that CfLec-2 was mainly located in the mantle, kidney and gonad. Furthermore, rCfLec-2 could bind to the surface of scallop hemocytes, and then initiated cellular adhesion and recruited hemocytes to enhance their encapsulation in vitro, and this process could be specifically blocked by anti-rCfLec-2 serum. These results collectively suggested that CfLec-2 from the primitive deuterostome C. farreri could perform two distinct immune functions, pathogen recognition and cellular adhesion synchronously, while these functions were performed by collectins and selectins in vertebrates, respectively. The synchronous functions of pathogen recognition and cellular adhesion performed by CfLec-2 tempted us to suspect that CfLec-2 was an ancient form of C-type lectin, and apparently the differentiation of these two functions mediated by C-type lectins occurred after mollusk in phylogeny.
-
Unprecedented selectivity for homologous lectin targets: differential targeting of the viral receptors L-SIGN and DC-SIGN
Franck FIESCHI,Clara Delaunay,Sara Pollastri,Michel Thépaut,Gianluca Cavazzoli,Laura Belvisi,Clémentine Bouchikri,Nuria Labiod,Ana Gimeno,Antonio Franconetti,Jesús Jiménez-Barbero,Ana Ardá,Rafael Delgado,Anna Bernardi
DOI: https://doi.org/10.26434/chemrxiv-2023-tf1fg-v2
2024-04-17
Abstract:DC-SIGN (CD209) and L-SIGN (CD209L) are two C-type lectin receptors (CLRs) that facilitate SARS-CoV-2 infections as viral co-receptors. SARS-CoV-2 manipulates both DC-SIGN and L-SIGN for enhanced infection, leading to interest in developing receptor antagonists. Despite their structural similarity (82% sequence identity), they function differently. DC-SIGN, found in dendritic cells, shapes the immune response by recognizing pathogen-associated carbohydrate patterns. In contrast, L-SIGN, expressed in airway epithelial endothelial cells, is not directly involved in immunity. COVID-19’s primary threat is the hyperactivation of the immune system, potentially reinforced if DC-SIGN engages with exogenous ligands. Therefore, L-SIGN, co-localized with ACE2-expressing cells in the respiratory tract, is a more suitable target for anti-adhesion therapy. However, designing a selective ligand for L-SIGN is challenging due to the high sequence identity of the Carbohydrate Recognition Domains (CRDs) of the two lectins. We here present Man84, a mannose ring modified with a methylene guanidine triazole at position 2. It binds L-SIGN with a KD of 12.7 μM ± 1 μM (ITC) and is the first known L-SIGN selective ligand, showing 50-fold selectivity over DC-SIGN (SPR). The X-ray structure of the L-SIGN CRD/Man84 complex reveals the guanidinium group’s role in achieving steric and electrostatic complementarity with L-SIGN. This allows us to trace the source of selectivity to a single amino acid difference between the two CRDs. NMR analysis confirms the binding mode in solution, highlighting Man84’s conformational selection upon complex formation. Dimeric versions of Man84 achieve additional selectivity and avidity in the low nanomolar range. These compounds selectively inhibit L-SIGN dependent trans-infection by SARS-CoV-2 and Ebola virus. Man84 and its dimeric constructs display the best affinity and avidity reported to date for low-valency glycomimetics targeting CLRs. They are promising tools for competing with SARS-CoV-2 anchoring in the respiratory tract and have potential for other medical applications.
Chemistry
-
Functional Interaction of Common Allergens and a C-type Lectin Receptor, Dendritic Cell-specific ICAM3-grabbing Non-integrin (DC-SIGN), on Human Dendritic Cells*
Shih-Chang Hsu,Chien-Ho Chen,Shih-Han Tsai,Hirokazu Kawasaki,Chih-Hsing Hung,Yu-Te Chu,Hui-Wen Chang,Yufeng Zhou,Jinrong Fu,Beverly Plunkett,Song-Nan Su,Stefan Vieths,Reiko T. Lee,Yuan C. Lee,Shau-Ku Huang
DOI: https://doi.org/10.1074/jbc.m109.058370
IF: 5.485
2010-01-01
Journal of Biological Chemistry
Abstract:Fucosylated glycans on pathogens are known to shape the immune response through their interaction with pattern recognition receptors, such as C-type lectin receptors (CLRs), on dendritic cells (DCs). Similar fucosylated structures are also commonly found in a variety of allergens, but their functional significance remains unclear. To test a hypothesis that allergen-associated glycans serve as the molecular patterns in functional interaction with CLRs, an enzyme-linked immunosorbent assay-based binding assay was performed to determine the binding activity of purified allergens and allergen extracts. THP-1 cells and monocyte-derived DCs (MDDCs) were investigated as a model for testing the functional effects of allergen-CLR interaction using enzyme-linked immunosorbent assay, Western blotting, and flow cytometry. Significant and saturable bindings of allergens and allergen extracts with variable binding activities to DC- specific ICAM3-grabbing non-integrin (DCSIGN) and its related receptor, L-SIGN, were found. These include bovine serum albumin coupled with a common glycoform (fucosylated glycan lacking the alpha 1,3-linked mannose) of allergens and a panel of purified allergens, including BG60 (Cyn dBG-60; Bermuda grass pollen) and Der p2 (house dust mite). The binding activity was calcium-dependent and inhibitable by fucose and Lewis-x trisaccharides (Lex). In THP-1 cells and human MDDCs, BG60-DC-SIGN interaction led to the activation of Raf-1 and ERK kinases and the induction of tumor necrosis factor-alpha expression. This effect could be blocked, in part, by Raf-1 inhibitor or anti-DC-SIGN antibodies and was significantly reduced in cells with DC-SIGN knockdown. These results suggest that allergens are able to interact with DC- SIGN and induce tumor necrosis factor-alpha expression in MDDCs via, in part, Raf-1 signaling pathways.