158 results on '"Vasta GR"'
Search Results
2. F-type lectin from the sea bass (Dicentrarchus labrax): purification, cDNA cloning, tissue expression and localization, and opsonic activity
- Author
-
Daniela Parrinello, Gerardo R. Vasta, Maria Giovanna Parisi, Aiti Vizzini, Giuseppina Salerno, Gigliola Benenati, Matteo Cammarata, Mirella Vazzana, SALERNO G, PARISI MG, PARRINELLO D, BENENATI G, VIZZINI A, VAZZANA M, VASTA GR, CAMMARATA M, Salerno, G, Parisi, MG, Parrinello, D, Benenati, G, Vizzini, A, Vazzana, M, Vasta, GR, and Cammarata, M
- Subjects
food.ingredient ,DNA, Complementary ,Immunoblotting ,Aquatic Science ,Chromatography, Affinity ,Bass (fish) ,F-type lectin ,Dicentrarchus labrax ,teleost ,emaggluthinins, opsonin ,food ,Phagocytosis ,Opsonin Proteins ,Complementary DNA ,Lectins ,Environmental Chemistry ,Animals ,RNA, Messenger ,Sea bass ,Cloning, Molecular ,Opsonin ,Phylogeny ,biology ,Base Sequence ,Lectin ,General Medicine ,biology.organism_classification ,Molecular biology ,Gene Expression Regulation ,Immunology ,biology.protein ,Macrophages, Peritoneal ,F lectin, sea bass, inflammation ,Dicentrarchus ,Bass ,Electrophoresis, Polyacrylamide Gel ,Sequence motif - Abstract
Recently described biochemical and structural aspects of fucose-binding lectins from the European eel (Anguilla anguilla) and striped bass (Morone saxatilis) led to the identification of a novel lectin family ("F-type" lectins) characterized by a unique sequence motif and a characteristic structural fold. The F-type fold is shared not only with other members of this lectin family, but also with apparently unrelated proteins ranging from prokaryotes to vertebrates. Here we describe the purification, biochemical and molecular properties, and the opsonic activity of an F-type lectin (DlFBL) isolated from sea bass (Dicentrarchus labrax) serum. DlFBL exhibits two tandemly arranged carbohydrate-recognition domains that display the F-type sequence motif. In situ hybridization and immunohistochemical analysis revealed that DlFBL is specifically expressed and localized in hepatocytes and intestinal cells. Exposure of formalin-killed Escherichia coli to DlFBL enhanced their phagocytosis by D. labrax peritoneal macrophages relative to the unexposed controls, suggesting that DlFBL may function as an opsonin in plasma and intestinal mucus.
- Published
- 2008
3. Evidence of Heavy Methylation in the Galectin 3 Promoter in Early Stages of Prostate Adenocarcinoma: Development and Validation of a Methylated Marker for Early Diagnosis of Prostate Cancer
- Author
-
Francesco Cappello, Vito Rodolico, Hafiz Ahmed, Gerardo R. Vasta, Ahmed, H, Cappello, F, Rodolico, V, and Vasta, GR
- Subjects
Cancer Research ,Pathology ,medicine.medical_specialty ,Settore MED/08 - Anatomia Patologica ,Biology ,Metastasis ,03 medical and health sciences ,Prostate cancer ,0302 clinical medicine ,Prostate ,LNCaP ,medicine ,Prostate, adenocarcinoma,Galectin 3 ,030304 developmental biology ,Galectin ,0303 health sciences ,Methylation ,medicine.disease ,galectin 3, prostate adenocarcinoma ,3. Good health ,medicine.anatomical_structure ,Oncology ,030220 oncology & carcinogenesis ,DNA methylation ,Cancer research ,Adenocarcinoma ,Research Article - Abstract
Galectins, soluble intracellular and extracellular β-galactoside–binding proteins, are known to be involved in the progression and metastasis of various cancers, including prostate adenocarcinoma, but the detailed mechanism of their biological roles remains elusive. In the prostate cancer cell lines PC-3 and DU-145, galectin 3 (gal3) is present at normal levels, whereas in LNCaP, its expression is silenced. In LNCaP, the gal3 promoter was heavily methylated, whereas PC-3 or DU-145 cells showed negligible or no methylation in the gal3 promoter indicating a negative correlation between gal3 promoter methylation and its expression. On immunohistochemical analysis of normal and tumor prostate tissues, gal3 was found expressed both in nucleus and cytoplasm of benign prostatic hyperplasia, high-grade prostatic intraepithelial neoplasia, and stage I. The expression of the gal3 was found drastically downregulated in advanced stages and, interestingly, mostly in the cytoplasm. On methylation analysis, the gal3 promoter in stage II prostate adenocarcinoma (PCa) was found heavily methylated, whereas in stages III and IV, it was only lightly methylated. However, in stage I PCa, both heavy and light methylations were observed in the gal3 promoter. In normal and benign prostatic hyperplasia tissues, the gal3 promoter was almost unmethylated. The differential cytosine methylation in the gal3 promoter in stages I to IV PCa enabled us to develop and validate a methylation-specific polymerase chain reaction–based sensitive assay specific for stages I and II PCa. These stages are considered the critical stages for successful intervention, thus underscoring the significance of this diagnostic assay. Translational Oncology (2009) 2, 146–156 Galectins, soluble intracellular and extracellular β-galactoside- binding proteins, are known to be involved in the progression and metastasis of various cancers, including prostate adenocarcinoma, but the detailed mechanism of their biological roles remains elusive. In the prostate cancer cell lines PC-3 and DU-145, galectin 3 (gal3) is present at normal levels, whereas in LNCaP, its expression is silenced. In LNCaP, the gal3 promoter was heavily methylated, whereas PC-3 or DU-145 cells showed negligible or no methylation in the gal3 promoter indicating a negative correlation between gal3 promoter methylation and its expression. On immunohistochemical analysis of normal and tumor prostate tissues, gal3 was found expressed both in nucleus and cytoplasm of benign prostatic hyperplasia, high-grade prostatic intraepithelial neoplasia, and stage I. The expression of the gal3 was found drastically downregulated in advanced stages and, interestingly, mostly in the cytoplasm. On methylation analysis, the gal3 promoter in stage II prostate adenocarcinoma (PCa) was found heavily methylated, whereas in stages III and IV, it was only lightly methylated. However, in stage I PCa, both heavy and light methylations were observed in the gal3 promoter. In normal and benign prostatic hyperplasia tissues, the gal3 promoter was almost unmethylated. The differential cytosine methylation in the gal3 promoter in stages I to IV PCa enabled us to develop and validate a methylation-specific polymerase chain reaction-based sensitive assay specific for stages I and II PCa. These stages are considered the critical stages for successful intervention, thus underscoring the significance of this diagnostic assay.
- Published
- 2009
- Full Text
- View/download PDF
4. Structural and functional diversity of the lectin repertoire in teleost fish: Relevance to innate and adaptive immunity
- Author
-
Hafiz Ahmed, Barbara Giomarelli, Nicolò Parrinello, Gerardo R. Vasta, Matteo Cammarata, Mario A. Bianchet, Mihai Nita-Lazar, L. Mario Amzel, Shaojun Du, Vasta GR, Nita-Lazar M, Giomarelli B, Ahmed H, Du S, Cammarata M, Parrinello N, Bianchet MA, and Amzel LM
- Subjects
Fish Proteins ,Models, Molecular ,Immunology ,Settore BIO/05 - Zoologia ,Biology ,Adaptive Immunity ,Article ,Immune system ,Phagocytosis ,C-type lectin ,Antifreeze Proteins ,Lectins ,Animals ,Lectins, Innate immunity, Fish, Self/non-self recognition, Effector, Regulatory functions, Complement activation ,Protein Structure, Quaternary ,Antigens, Viral ,Complement Activation ,Mannan-binding lectin ,Antigens, Bacterial ,Innate immune system ,Bacteria ,Effector ,Fishes ,Lectin ,Complement System Proteins ,Opsonin Proteins ,Acquired immune system ,Invertebrates ,Immunity, Innate ,Complement system ,Cell biology ,Protein Structure, Tertiary ,Gene Expression Regulation ,Organ Specificity ,Vertebrates ,Viruses ,biology.protein ,Developmental Biology - Abstract
Protein–carbohydrate interactions mediated by lectins have been recognized as key components of innate immunity in vertebrates and invertebrates, not only for recognition of potential pathogens, but also for participating in downstream effector functions, such as their agglutination, immobilization, and complement-mediated opsonization and killing. More recently, lectins have been identified as critical regulators of mammalian adaptive immune responses. Fish are endowed with virtually all components of the mammalian adaptive immunity, and are equipped with a complex lectin repertoire. In this review, we discuss evidence suggesting that: (a) lectin repertoires in teleost fish are highly diversified, and include not only representatives of the lectin families described in mammals, but also members of lectin families described for the first time in fish species; (b) the tissue-specific expression and localization of the diverse lectin repertoires and their molecular partners is consistent with their distinct biological roles in innate and adaptive immunity; (c) although some lectins may bind endogenous ligands, others bind sugars on the surface of potential pathogens; (d) in addition to pathogen recognition and opsonization, some lectins display additional effector roles, such as complement activation and regulation of immune functions; (e) some lectins that recognize exogenous ligands mediate processes unrelated to immunity: they may act as anti-freeze proteins or prevent polyspermia during fertilization.
- Published
- 2011
5. Isolation and characterization of a fish F-type lectin from gilt head bream (Sparus aurata) serum
- Author
-
Gigliola Benenati, Eric W. Odom, Nicolò Parrinello, Aiti Vizzini, Gerardo R. Vasta, Matteo Cammarata, Giuseppina Salerno, CAMMARATA, M, BENENATI, G, ODOM EW, SALERNO, G, VIZZINI, A, VASTA, GR, and PARRINELLO, N
- Subjects
Serum hemagglutinins ,Teleost ,Molecular Sequence Data ,Biophysics ,Biochemistry ,Affinity chromatography ,Western blot ,Sparus aurata ,Lectins ,medicine ,Animals ,Dicentrarchus labrax ,Amino Acid Sequence ,Sea bass ,Molecular Biology ,Peptide sequence ,Polyacrylamide gel electrophoresis ,biology ,Molecular mass ,medicine.diagnostic_test ,Sequence Homology, Amino Acid ,Lectin ,F-type lectin ,biology.organism_classification ,Sea Bream ,biology.protein ,Chromatography, Gel ,Dicentrarchus ,Electrophoresis, Polyacrylamide Gel - Abstract
A novel fucose-binding lectin, designated SauFBP32, was purified by affinity chromatography on fucose-agarose, from the serum of the gilt head bream Sparus aurata. Electrophoretic mobility of the subunit revealed apparent molecular weights of 35 and 30 kDa under reducing and non-reducing conditions, respectively. Size exclusion analysis suggests that the native lectin is a monomer under the selected experimental conditions. Agglutinating activity towards rabbit erythrocytes was not significantly modified by addition of calcium or EDTA; activity was optimal at 37 degrees C, retained partial activity by treatment at 70 degrees C, and was fully inactivated at 90 degrees C. On western blot analysis, SauFBP showed intense cross-reactivity with antibodies specific for a sea bass (Dicentrarchus labrax) fucose-binding lectin. In addition, the similarity of the N-terminal sequence and a partial coding domain to teleost F-type lectins suggests that SauFBP32 is a member of this emerging family of lectins.
- Published
- 2006
6. Galectin-3 disrupts tight junctions of airway epithelial cell monolayers by inducing expression and release of matrix metalloproteinases upon influenza a infection.
- Author
-
Iqbal M, Feng C, Zong G, Wang LX, and Vasta GR
- Abstract
Galectins are β-galactosyl-binding lectins with key roles in early development, immune regulation, and infectious disease. Influenza A virus (IAV) infects the airway epithelia, and in severe cases may lead to bacterial superinfections and hypercytokinemia, and eventually, to acute respiratory distress syndrome (ARDS) through the breakdown of airway barriers. The detailed mechanisms involved, however, remain poorly understood. Our prior in vivo studies in a murine model system revealed that upon experimental IAV and pneumococcal primary and secondary challenges, respectively, galectin-1 and galectin-3 (Gal-3) are released into the airway and bind to the epithelium that has been desialylated by the viral neuraminidase, contributing to secondary bacterial infection and hypercytokinemia leading to the clinical decline and death of the animals. Here we report the results of in vitro studies that reveal the role of the extracellular Gal-3 in additional detrimental effects on the host by disrupting the integrity of the airway epithelial barrier. IAV infection of the human airway epithelia cell line A549 increased release of Gal-3 and its binding to the A549 desialylated cell surface, notably to the transmembrane signaling receptors CD147 and integrin-β1. Addition of recombinant Gal-3 to A549 monolayers resulted in enhanced expression and release of matrix metalloproteinases, leading to disruption of cell-cell tight junctions, and a significant increase in paracellular permeability. This study reveals a critical mechanism involving Gal-3 that may significantly contribute to the severity of IAV infections by promoting disruption of tight junctions and enhanced permeability of the airway epithelia, potentially leading to lung edema and ARDS., (© The Author(s) 2024. Published by Oxford University Press. All rights reserved. For permissions, please e-mail: journals.permissions@oup.com.)
- Published
- 2024
- Full Text
- View/download PDF
7. Aberrant GPA expression and regulatory function of red blood cells in sickle cell disease.
- Author
-
Marshall JN, Klein MN, Karki P, Promnares K, Setua S, Fan X, Buehler PW, Birukov KG, Vasta GR, and Fontaine MJ
- Subjects
- Humans, Endothelial Cells metabolism, Glycophorins metabolism, Erythrocytes metabolism, Sialic Acid Binding Immunoglobulin-like Lectins metabolism, Anemia, Sickle Cell, Vascular Diseases
- Abstract
Abstract: Glycophorin A (GPA), a red blood cell (RBC) surface glycoprotein, can maintain peripheral blood leukocyte quiescence through interaction with a sialic acid-binding Ig-like lectin (Siglec-9). Under inflammatory conditions such as sickle cell disease (SCD), the GPA of RBCs undergo structural changes that affect this interaction. Peripheral blood samples from patients with SCD before and after RBC transfusions were probed for neutrophil and monocyte activation markers and analyzed by fluorescence-activated cell sorting (FACS). RBCs were purified and tested by FACS for Siglec-9 binding and GPA expression, and incubated with cultured endothelial cells to evaluate their effect on barrier function. Activated leukocytes from healthy subjects (HS) were coincubated with healthy RBCs (RBCH), GPA-altered RBCs, or GPA-overexpressing (OE) cells and analyzed using FACS. Monocyte CD63 and neutrophil CD66b from patients with SCD at baseline were increased 47% and 27%, respectively, as compared with HS (P = .0017, P = .0162). After transfusion, these markers were suppressed by 22% and 17% (P = .0084, P = .0633). GPA expression in RBCSCD was 38% higher (P = .0291) with decreased Siglec-9 binding compared with RBCH (0.0266). Monocyte CD63 and neutrophil CD66b were suppressed after incubation with RBCH and GPA-OE cells, but not with GPA-altered RBCs. Endothelial barrier dysfunction after lipopolysaccharide challenge was restored fully with exposure to RBCH, but not with RBCSCD, from patients in pain crisis, or with RBCH with altered GPA. Pretransfusion RBCSCD do not effectively maintain the quiescence of leukocytes and endothelium, but quiescence is restored through RBC transfusion, likely by reestablished GPA-Siglec-9 interactions., (© 2024 by The American Society of Hematology. Licensed under Creative Commons Attribution-NonCommercial-NoDerivatives 4.0 International (CC BY-NC-ND 4.0), permitting only noncommercial, nonderivative use with attribution. All other rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
8. Scavenger receptor B2, a type III membrane pattern recognition receptor, senses LPS and activates the IMD pathway in crustaceans.
- Author
-
Shi XZ, Yang MC, Kang XL, Li YX, Hong PP, Zhao XF, Vasta GR, and Wang JX
- Subjects
- Animals, Drosophila melanogaster, Lipopolysaccharides, Receptors, Pattern Recognition genetics, Receptors, Pattern Recognition metabolism, Up-Regulation, Vibrio, Signal Transduction, Humans, Crustacea genetics, Crustacea immunology, Crustacea metabolism, Crustacea microbiology
- Abstract
The immune deficiency (IMD) pathway is critical for elevating host immunity in both insects and crustaceans. The IMD pathway activation in insects is mediated by peptidoglycan recognition proteins, which do not exist in crustaceans, suggesting a previously unidentified mechanism involved in crustacean IMD pathway activation. In this study, we identified a Marsupenaeus japonicus B class type III scavenger receptor, SRB2, as a receptor for activation of the IMD pathway. SRB2 is up-regulated upon bacterial challenge, while its depletion exacerbates bacterial proliferation and shrimp mortality via abolishing the expression of antimicrobial peptides. The extracellular domain of SRB2 recognizes bacterial lipopolysaccharide (LPS), while its C-terminal intracellular region containing a cryptic RHIM-like motif interacts with IMD, and activates the pathway by promoting nuclear translocation of RELISH. Overexpressing shrimp SRB2 in Drosophila melanogaster S2 cells potentiates LPS-induced IMD pathway activation and diptericin expression. These results unveil a previously unrecognized SRB2-IMD axis responsible for antimicrobial peptide induction and restriction of bacterial infection in crustaceans and provide evidence of biological diversity of IMD signaling in animals. A better understanding of the innate immunity of crustaceans will permit the optimization of prevention and treatment strategies against the arising shrimp diseases.
- Published
- 2023
- Full Text
- View/download PDF
9. Galectin-1 mediates interactions between polymorphonuclear leukocytes and vascular endothelial cells, and promotes their extravasation during lipopolysaccharide-induced acute lung injury.
- Author
-
Feng C, Cross AS, and Vasta GR
- Subjects
- Humans, Mice, Animals, Lipopolysaccharides pharmacology, Endothelial Cells, Galectin 1, Galectin 3, Cell Adhesion, Lung, Streptococcus pneumoniae, Intercellular Adhesion Molecule-1 metabolism, Neutrophils, Acute Lung Injury chemically induced, Acute Lung Injury metabolism
- Abstract
The lung airway epithelial surface is heavily covered with sialic acids as the terminal carbohydrate on most cell surface glycoconjugates and can be removed by microbial neuraminidases or endogenous sialidases. By desialylating the lung epithelial surface, neuraminidase acts as an important virulence factor in many mucosal pathogens, such as influenza and S. pneumoniae. Desialylation exposes the subterminal galactosyl moieties - the binding glycotopes for galectins, a family of carbohydrate-recognition proteins playing important roles in various aspects of immune responses. Galectin-1 and galectin-3 have been extensively studied in their roles related to host immune responses, but some questions about their role(s) in leukocyte recruitment during lung bacterial infection remain unanswered. In this study, we found that both galectin-1 and galectin-3 bind to polymorphonuclear leukocytes (PMNs) and enhance the interaction of endothelial intercellular adhesion molecule-1 (ICAM-1) with PMNs, which is further increased by PMN desialylation. In addition, we observed that in vitro galectin-1 mediates the binding of PMNs, particularly desialylated PMNs, onto the endothelial cells. Finally, in a murine model for LPS-mediated acute lung injury, we observed that galectin-1 modulates PMN infiltration to the lung without altering the expression of chemoattractant cytokines. We conclude that galectins, particularly galectin-1, may function as adhesion molecules that mediate PMN-endothelial cell interactions, and modulate PMN infiltration during acute lung injury., Competing Interests: Declaration of Competing Interest The authors declare that they have no known competing financial interests or personal relationships that could have appeared to influence the work reported in this paper., (Copyright © 2023 Elsevier Ltd. All rights reserved.)
- Published
- 2023
- Full Text
- View/download PDF
10. Editorial: The role of glycans in infectious disease, Volume II.
- Author
-
Martínez-Duncker I, Mora-Montes HM, Vasta GR, and Chiodo F
- Abstract
Competing Interests: The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.
- Published
- 2022
- Full Text
- View/download PDF
11. Synthesis, binding affinity, and inhibitory capacity of cyclodextrin-based multivalent glycan ligands for human galectin-3.
- Author
-
Ou C, Li C, Feng C, Tong X, Vasta GR, and Wang LX
- Subjects
- Galectin 3 metabolism, Humans, Ligands, Protein Binding, Cyclodextrins, beta-Cyclodextrins pharmacology
- Abstract
Human galectin 3 (Gal-3) has been implicated to play important roles in different biological recognition processes such as tumor growth and cancer metastasis. High-affinity Gal-3 ligands are desirable for functional studies and as inhibitors for potential therapeutic development. We report here a facile synthesis of β-cyclodextrin (CD)-based Tn and TF antigen-containing multivalent ligands via a click reaction. Binding studies indicated that the synthetic multivalent glycan ligands demonstrated a clear clustering effect in binding to human Gal-3, with up to 153-fold enhanced relative affinity in comparison with the monomeric glycan ligand. The GalNAc (Tn antigen) containing heptavalent ligand showed the highest affinity for human Gal-3 among the synthetic ligands tested, with an EC
50 of 1.4 μM in binding to human Gal-3. A cell-based assay revealed that the synthetic CD-based multivalent ligands could efficiently inhibit Gal-3 binding to human airway epithelial cells, with an inhibitory capacity consistent with their binding affinity measured by SPR. The synthetic cyclodextrin-based ligands described in this study should be valuable for functional studies of human Gal-3 and potentially for therapeutic applications., Competing Interests: Declaration of Competing Interest The authors declare that they have no known competing financial interests or personal relationships that could have appeared to influence the work reported in this paper., (Copyright © 2022 Elsevier Ltd. All rights reserved.)- Published
- 2022
- Full Text
- View/download PDF
12. Manipulating Galectin Expression in Zebrafish (Danio rerio).
- Author
-
Feng C, Nita-Lazar M, González-Montalbán N, Wang J, Mancini J, Wang S, Ravindran C, Ahmed H, and Vasta GR
- Subjects
- Animals, Gene Knockdown Techniques, Mammals genetics, Morpholinos genetics, Morpholinos metabolism, RNA metabolism, Galectins metabolism, Zebrafish metabolism
- Abstract
Techniques for disrupting gene expression are invaluable tools for the analysis of the biological role of a gene product. Because of its genetic tractability and multiple advantages over conventional mammalian models, the zebrafish (Danio rerio) is recognized as a powerful system for gaining new insight into diverse aspects of human health and disease. Among the multiple mammalian gene families for which the zebrafish has shown promise as an invaluable model for functional studies, the galectins have attracted great interest due to their participation in early development, regulation of immune homeostasis, and recognition of microbial pathogens. Galectins are β-galactosyl-binding lectins with a characteristic sequence motif in their carbohydrate recognition domains (CRDs), that constitute an evolutionary conserved family ubiquitous in eukaryotic taxa. Galectins are emerging as key players in the modulation of many important pathological processes, which include acute and chronic inflammatory diseases, autoimmunity and cancer, thus making them potential molecular targets for innovative drug discovery. Here, we provide a review of the current methods available for the manipulation of gene expression in the zebrafish, with a focus on gene knockdown [morpholino (MO)-derived antisense oligonucleotides] and knockout (CRISPR-Cas) technologies., (© 2022. Springer Science+Business Media, LLC, part of Springer Nature.)
- Published
- 2022
- Full Text
- View/download PDF
13. F-type lectin from serum of the Antarctic teleost fish Trematomus bernacchii (Boulenger, 1902): Purification, structural characterization, and bacterial agglutinating activity.
- Author
-
Dara M, Giulianini PG, Manfrin C, Parisi MG, Parrinello D, La Corte C, Vasta GR, and Cammarata M
- Subjects
- Amino Acid Sequence, Animals, Antarctic Regions, Base Sequence, Lectins isolation & purification, Lectins metabolism, Phylogeny, Bacteria metabolism, Fucose metabolism, Lectins blood, Lectins physiology, Perciformes physiology
- Abstract
The increasing availability of sequenced genomes has enabled a deeper understanding of the complexity of fish lectin repertoires involved in early development and immune recognition. The teleost fucose-type lectin (FTL) family includes proteins that preferentially bind fucose and display tandemly arrayed carbohydrate-recognition domains (CRDs) or are found in mosaic combinations with other domains. They function as opsonins, promoting phagocytosis and the clearance of microbial pathogens. The Antarctic fish Trematomus bernacchii is a Perciforme living at extremely low temperatures (-1.68 °C) which is considered a model for studying adaptability to the variability of environmental waters. Here, we isolated a Ca
++ -independent fucose-binding protein from the serum of T. bernacchii by affinity chromatography with apparent molecular weights of 32 and 30 kDa under reducing and non-reducing conditions, respectively. We have characterized its carbohydrate binding properties, thermal stability and potential ability to recognize bacterial pathogens. In western blot analysis, the protein showed intense cross-reactivity with antibodies specific for a sea bass (Dicentrarchus labrax) fucose-binding lectin. In addition, its molecular and structural aspects, showing that it contains two CRD-FTLs confirmed that T. bernacchii FTL (TbFTL) is a bona fide member of the FTL family, with binding activity at low temperatures and the ability to agglutinate bacteria, thereby suggesting it participates in host-pathogen interactions in low temperature environments., (Copyright © 2021 Elsevier Inc. All rights reserved.)- Published
- 2021
- Full Text
- View/download PDF
14. In Structural Glycobiology, Deuterium provides the Details.
- Author
-
Vasta GR and Amzel LM
- Subjects
- Deuterium, Lectins, Ligands, Fucose, Glycomics
- Abstract
In this issue of Structure, Gadjos et al. (2021b) determine the structure of a bacterial lectin in complex with L-fucose by neutron diffraction of both perdeuterated protein and carbohydrate ligand. The structure provides insight into lectin-ligand interactions, opening avenues for drug design targeting bacterial lectins for intervention in infectious disease., (Copyright © 2021 Elsevier Ltd. All rights reserved.)
- Published
- 2021
- Full Text
- View/download PDF
15. Glycan characterization of pregnancy-specific glycoprotein 1 and its identification as a novel Galectin-1 ligand.
- Author
-
Mendoza M, Lu D, Ballesteros A, Blois SM, Abernathy K, Feng C, Dimitroff CJ, Zmuda J, Panico M, Dell A, Vasta GR, Haslam SM, and Dveksler G
- Subjects
- Female, Galectin 1 chemistry, Humans, Ligands, Polysaccharides chemistry, Pregnancy, Pregnancy-Specific beta 1-Glycoproteins chemistry, Pregnancy-Specific beta 1-Glycoproteins isolation & purification, Galectin 1 metabolism, Polysaccharides metabolism, Pregnancy-Specific beta 1-Glycoproteins metabolism
- Abstract
Pregnancy-specific beta 1 glycoprotein (PSG1) is secreted from trophoblast cells of the human placenta in increasing concentrations as pregnancy progresses, becoming one of the most abundant proteins in maternal serum in the third trimester. PSG1 has seven potential N-linked glycosylation sites across its four domains. We carried out glycomic and glycoproteomic studies to characterize the glycan composition of PSG1 purified from serum of pregnant women and identified the presence of complex N-glycans containing poly LacNAc epitopes with α2,3 sialyation at four sites. Using different techniques, we explored whether PSG1 can bind to galectin-1 (Gal-1) as these two proteins were previously shown to participate in processes required for a successful pregnancy. We confirmed that PSG1 binds to Gal-1 in a carbohydrate-dependent manner with an affinity of the interaction of 0.13 μM. In addition, we determined that out of the three N-glycosylation-carrying domains, only the N and A2 domains of recombinant PSG1 interact with Gal-1. Lastly, we observed that the interaction between PSG1 and Gal-1 protects this lectin from oxidative inactivation and that PSG1 competes the ability of Gal-1 to bind to some but not all of its glycoprotein ligands., (Published by Oxford University Press 2020. This work is written by (a) US Government employee(s) and is in the public domain in the US.)
- Published
- 2020
- Full Text
- View/download PDF
16. Galectin-mediated immune recognition: Opsonic roles with contrasting outcomes in selected shrimp and bivalve mollusk species.
- Author
-
Vasta GR and Wang JX
- Subjects
- Animals, Arthropod Proteins genetics, Galectins genetics, Gene Expression Regulation, Host-Parasite Interactions, Immunity, Innate, Molecular Mimicry, Arthropod Proteins metabolism, Bacterial Infections immunology, Bivalvia immunology, Galectins metabolism, Opsonin Proteins metabolism, Penaeidae immunology, Receptors, Pattern Recognition metabolism
- Abstract
Galectins are a structurally conserved family of ß-galactoside-binding lectins characterized by a unique sequence motif in the carbohydrate recognition domain, and of wide taxonomic distribution, from fungi to mammals. Their biological functions, initially described as key to embryogenesis and early development via recognition of endogenous ("self") carbohydrate moieties, are currently understood as also encompassing tissue repair, cancer metastasis, angiogenesis, adipogenesis, and regulation of immune homeostasis. More recently, however, numerous studies have contributed to establish a new paradigm by revealing that galectins can also bind to exogenous ("non-self") glycans on the surface of potentially pathogenic virus, bacteria, and eukaryotic parasites, and function both as pathogen recognition receptors (PRRs) and effector factors in innate immunity. Our studies on a galectin from the kuruma shrimp Marsupenaeus japonicus (MjGal), revealed that it functions as a typical PRR. Expression of MjGal is upregulated by infectious challenge, and can recognize both Gram (+) and Gram (-) bacteria. MjGal also recognizes carbohydrates on the shrimp hemocyte surface, and can cross-link microbial pathogens to the hemocytes, promoting their phagocytosis and clearance from circulation. Therefore, MjGal contributes to the shrimp's immune defense against infectious challenge both as a PRR and effector factor. Our studies on galectins from the bivalve mollusks, however, have shown that although they can function in immune defense as MjGal, protistan parasites take advantage of the recognition roles of the host galectins, for successful attachment and host infection. We identified in the eastern oyster Crassostrea virginica two galectins (CvGal1 and CvGal2) that not only recognize a large variety of bacterial species, but also the protozoan parasite Perkinsus marinus. Like the shrimp MjGal, both oyster galectins function as opsonins, and promote parasite adhesion and phagocytosis. However, P. marinus survives intrahemocytic oxidative killing and proliferates, eventually causing systemic infection and death of the oyster host. In the softshell clam Mya arenaria we identified a galectin (MaGal1) that displays carbohydrate specificity and recognition properties for sympatric Perkinsus species (P. marinus and P. chesapeaki), that are different from CvGal1 and CvGal2. Our results suggest that although galectins from bivalves can function as PRRs, Perkinsus parasites have co-evolved with their hosts to subvert the galectins' immune functions for host infection by acquisition of carbohydrate-based mimicry., (Copyright © 2020. Published by Elsevier Ltd.)
- Published
- 2020
- Full Text
- View/download PDF
17. The functional relevance of shrimp C-type lectins in host-pathogen interactions.
- Author
-
Wang XW, Vasta GR, and Wang JX
- Subjects
- Animals, Arthropod Proteins genetics, Arthropod Proteins metabolism, Endosomes immunology, Endosomes metabolism, Endosomes virology, Hemocytes immunology, Hemocytes metabolism, Hemocytes virology, Host-Pathogen Interactions immunology, Immunity, Innate immunology, Lectins, C-Type genetics, Lectins, C-Type metabolism, Penaeidae genetics, Penaeidae virology, White spot syndrome virus 1 physiology, Arthropod Proteins immunology, Gene Expression immunology, Lectins, C-Type immunology, Penaeidae immunology, White spot syndrome virus 1 immunology
- Abstract
C-type lectins (CTLs) are key recognition proteins in shrimp immunity. A few years ago we reviewed sequence information, ligand specificity, expression profiles and specific functions of the shrimp CTLs. Since then, multiple integrated studies that implemented biochemical approaches using both the native and recombinant proteins, functional genetic approaches using RNA interference, and mechanistic studies by analyzing protein-protein interactions were carried out. Results from these rigorous studies revealed the functions and mechanisms of action of selected members of the shrimp CTL family. This review focuses on this new knowledge, that includes unique structural aspects, functions, and mechanisms in host-pathogen interactions, the functional relevance of regions other than the C-type lectin domain, and the regulation of transcription of shrimp CTLs. Thus, this review aims to provide a detailed update of recent studies that have contributed to our better understanding of the shrimp immune events that involve CTL functions., (Copyright © 2020 Elsevier Ltd. All rights reserved.)
- Published
- 2020
- Full Text
- View/download PDF
18. Introduction to special issue: Pattern recognition receptors and their roles in immunity in invertebrates.
- Author
-
Wang JX and Vasta GR
- Subjects
- Animals, Invertebrates microbiology, Invertebrates parasitology, Adaptive Immunity immunology, Immunity, Innate immunology, Invertebrates immunology, Receptors, Pattern Recognition immunology
- Published
- 2020
- Full Text
- View/download PDF
19. Biochemical Characterization of Oyster and Clam Galectins: Selective Recognition of Carbohydrate Ligands on Host Hemocytes and Perkinsus Parasites.
- Author
-
Vasta GR, Feng C, Tasumi S, Abernathy K, Bianchet MA, Wilson IBH, Paschinger K, Wang LX, Iqbal M, Ghosh A, Amin MN, Smith B, Brown S, and Vista A
- Abstract
Both vertebrates and invertebrates display active innate immune mechanisms for defense against microbial infection, including diversified repertoires of soluble and cell-associated lectins that can effect recognition and binding to potential pathogens, and trigger downstream effector pathways that clear them from the host internal milieu. Galectins are widely distributed and highly conserved lectins that have key regulatory effects on both innate and adaptive immune responses. In addition, galectins can bind to exogenous ("non-self") carbohydrates on the surface of bacteria, enveloped viruses, parasites, and fungi, and function as recognition receptors and effector factors in innate immunity. Like most invertebrates, eastern oysters ( Crassostrea virginica ) and softshell clams ( Mya arenaria ) can effectively respond to most immune challenges through soluble and hemocyte-associated lectins. The protozoan parasite Perkinsus marinus , however, can infect eastern oysters and cause "Dermo" disease, which is highly detrimental to both natural and farmed oyster populations. The sympatric Perkinsus chesapeaki , initially isolated from infected M. arenaria clams, can also be present in oysters, and there is little evidence of pathogenicity in either clams or oysters. In this review, we discuss selected observations from our studies on the mechanisms of Perkinsus recognition that are mediated by galectin-carbohydrate interactions. We identified in the oyster two galectins that we designated CvGal1 and CvGal2, which strongly recognize P. marinus trophozoites. In the clam we also identified galectin sequences, and focused on one (that we named MaGal1) that also recognizes Perkinsus species. Here we describe the biochemical characterization of CvGal1, CvGal2, and MaGal1 with focus on the detailed study of the carbohydrate specificity, and the glycosylated moieties on the surfaces of the oyster hemocytes and the two Perkinsus species ( P. marinus and P. chesapeaki ). Our goal is to gain further understanding of the biochemical basis for the interactions that lead to recognition and opsonization of the Perkinsus trophozoites by the bivalve hemocytes. These basic studies on the biology of host-parasite interactions may contribute to the development of novel intervention strategies for parasitic diseases of biomedical interest., (Copyright © 2020 Vasta, Feng, Tasumi, Abernathy, Bianchet, Wilson, Paschinger, Wang, Iqbal, Ghosh, Amin, Smith, Brown and Vista.)
- Published
- 2020
- Full Text
- View/download PDF
20. Galectins in Host-Pathogen Interactions: Structural, Functional and Evolutionary Aspects.
- Author
-
Vasta GR
- Subjects
- Animals, Humans, Phagocytosis, Polysaccharides immunology, Biological Evolution, Galectins chemistry, Galectins immunology, Host-Pathogen Interactions immunology, Immunity, Innate
- Abstract
Galectins are a family of ß-galactoside-binding lectins characterized by a unique sequence motif in the carbohydrate recognition domain, and evolutionary and structural conservation from fungi to invertebrates and vertebrates, including mammals. Their biological roles, initially understood as limited to recognition of endogenous ("self") carbohydrate ligands in embryogenesis and early development, dramatically expanded in later years by the discovery of their roles in tissue repair, cancer, adipogenesis, and regulation of immune homeostasis. In recent years, however, evidence has also accumulated to support the notion that galectins can bind ("non-self") glycans on the surface of potentially pathogenic microbes, and function as recognition and effector factors in innate immunity. Thus, this evidence has established a new paradigm by which galectins can function not only as pattern recognition receptors but also as effector factors, by binding to the microbial surface and inhibiting adhesion and/or entry into the host cell, directly killing the potential pathogen by disrupting its surface structures, or by promoting phagocytosis, encapsulation, autophagy, and pathogen clearance from circulation. Strikingly, some viruses, bacteria, and protistan parasites take advantage of the aforementioned recognition roles of the vector/host galectins, for successful attachment and invasion. These recent findings suggest that galectin-mediated innate immune recognition and effector mechanisms, which throughout evolution have remained effective for preventing or fighting viral, bacterial, and parasitic infection, have been "subverted" by certain pathogens by unique evolutionary adaptations of their surface glycome to gain host entry, and the acquisition of effective mechanisms to evade the host's immune responses.
- Published
- 2020
- Full Text
- View/download PDF
21. F-Type Lectins: Structure, Function, and Evolution.
- Author
-
Vasta GR and Feng C
- Subjects
- Binding Sites, Carbohydrate Sequence, Evolution, Molecular, Lectins genetics, Models, Molecular, Multigene Family, Protein Binding, Protein Conformation, Protein Domains, Fucose metabolism, Lectins chemistry, Lectins metabolism
- Abstract
F-type lectins (FTLs) are characterized by a fucose recognition domain (F-type lectin domain; FTLD) that displays a novel jellyroll fold ("F-type" fold) and unique carbohydrate- and calcium-binding sequence motifs. This novel lectin family comprises widely distributed proteins exhibiting single, double, or greater multiples of the FTLD, either tandemly arrayed or combined with other structurally and functionally distinct domains. Further, differences in carbohydrate specificity among tandemly arrayed FTLDs present in any FTL polypeptide subunit, together with the expression of multiple FTL isoforms in a single individual supports a striking diversity in ligand recognition. Functions of FTLs in self/nonself recognition include innate immunity, fertilization, microbial adhesion, and pathogenesis, among others, revealing an extensive structural/functional diversification. The taxonomic distribution of FTLDs is surprisingly discontinuous, suggesting that this lectin family has been subject to secondary loss, lateral transfer, and functional co-option along evolutionary lineages.
- Published
- 2020
- Full Text
- View/download PDF
22. Purification and Biochemical Characterization of Selected F-Type Lectins.
- Author
-
Feng C and Vasta GR
- Subjects
- Animals, Bass genetics, Chromatography, Affinity, Chromatography, Gel, Fish Proteins metabolism, Gene Expression Profiling, Gene Expression Regulation, Lectins metabolism, Multigene Family, Recombinant Proteins metabolism, Bass metabolism, Lectins genetics, Lectins isolation & purification, Liver metabolism
- Abstract
The purification of fucose-binding lectins from the liver of striped bass (Morone saxatilis), a teleost fish, and the identification of a novel lectin sequence motif led to the identification of a new family of lectins, the F-type lectins (FTLs) (see overview of the FTL family in Chapter 23 ). Isolation and purification of these proteins from liver extracts of striped bass was accomplished by affinity chromatography and size exclusion, and their identification as FTLs, by direct Edman sequencing, and protein, transcript, and gene sequence analysis. The development of specific antibodies against the M. saxatilis FTL provided an additional tool for the identification of FTLs. These methods have been successfully used for the purification of the FTL family members from tissues and body fluids of various animal species. Production and characterization of FTLs has been facilitated by the expression of the recombinant proteins. In this chapter, the biochemical characterization of FTLs is focused on the analysis of their carbohydrate specificity.
- Published
- 2020
- Full Text
- View/download PDF
23. Lacking catalase, a protistan parasite draws on its photosynthetic ancestry to complete an antioxidant repertoire with ascorbate peroxidase.
- Author
-
Schott EJ, Di Lella S, Bachvaroff TR, Amzel LM, and Vasta GR
- Subjects
- Amino Acid Sequence, Animals, Ascorbate Peroxidases chemistry, Ascorbate Peroxidases genetics, Ascorbate Peroxidases isolation & purification, Hydrogen Peroxide metabolism, Kinetics, Models, Molecular, Parasites genetics, Phylogeny, Structural Homology, Protein, Subcellular Fractions metabolism, Antioxidants metabolism, Ascorbate Peroxidases metabolism, Catalase metabolism, Parasites enzymology, Photosynthesis
- Abstract
Background: Antioxidative enzymes contribute to a parasite's ability to counteract the host's intracellular killing mechanisms. The facultative intracellular oyster parasite, Perkinsus marinus, a sister taxon to dinoflagellates and apicomplexans, is responsible for mortalities of oysters along the Atlantic coast of North America. Parasite trophozoites enter molluscan hemocytes by subverting the phagocytic response while inhibiting the typical respiratory burst. Because P. marinus lacks catalase, the mechanism(s) by which the parasite evade the toxic effects of hydrogen peroxide had remained unclear. We previously found that P. marinus displays an ascorbate-dependent peroxidase (APX) activity typical of photosynthetic eukaryotes. Like other alveolates, the evolutionary history of P. marinus includes multiple endosymbiotic events. The discovery of APX in P. marinus raised the questions: From which ancestral lineage is this APX derived, and what role does it play in the parasite's life history?, Results: Purification of P. marinus cytosolic APX activity identified a 32 kDa protein. Amplification of parasite cDNA with oligonucleotides corresponding to peptides of the purified protein revealed two putative APX-encoding genes, designated PmAPX1 and PmAPX2. The predicted proteins are 93% identical, and PmAPX2 carries a 30 amino acid N-terminal extension relative to PmAPX1. The P. marinus APX proteins are similar to predicted APX proteins of dinoflagellates, and they more closely resemble chloroplastic than cytosolic APX enzymes of plants. Immunofluorescence for PmAPX1 and PmAPX2 shows that PmAPX1 is cytoplasmic, while PmAPX2 is localized to the periphery of the central vacuole. Three-dimensional modeling of the predicted proteins shows pronounced differences in surface charge of PmAPX1 and PmAPX2 in the vicinity of the aperture that provides access to the heme and active site., Conclusions: PmAPX1 and PmAPX2 phylogenetic analysis suggests that they are derived from a plant ancestor. Plant ancestry is further supported by the presence of ascorbate synthesis genes in the P. marinus genome that are similar to those in plants. The localizations and 3D structures of the two APX isoforms suggest that APX fulfills multiple functions in P. marinus within two compartments. The possible role of APX in free-living and parasitic stages of the life history of P. marinus is discussed.
- Published
- 2019
- Full Text
- View/download PDF
24. Pregnancy Galectinology: Insights Into a Complex Network of Glycan Binding Proteins.
- Author
-
Blois SM, Dveksler G, Vasta GR, Freitag N, Blanchard V, and Barrientos G
- Subjects
- Animals, Carbohydrate Sequence, Chromosome Mapping, Dimerization, Embryo, Nonmammalian metabolism, Embryonic Development physiology, Endothelial Cells metabolism, Extracellular Vesicles metabolism, Female, Galectins chemistry, Galectins genetics, Glycosylation, Humans, Maternal-Fetal Exchange physiology, Neovascularization, Physiologic physiology, Placentation physiology, Polysaccharides chemistry, Pre-Eclampsia metabolism, Pregnancy, Protein Processing, Post-Translational, Structure-Activity Relationship, Substrate Specificity, Trophoblasts metabolism, Galectins physiology, Polysaccharides metabolism, Pregnancy Proteins physiology
- Abstract
Galectins are a phylogenetically conserved family of soluble β-galactoside binding proteins, consisting of 15 different types, each with a specific function. Galectins contribute to placentation by regulating trophoblast development, migration, and invasion during early pregnancy. In addition, galectins are critical players regulating maternal immune tolerance to the embedded embryo. Recently, the role of galectins in angiogenesis during decidualization and in placenta formation has gained attention. Altered expression of galectins is associated with abnormal pregnancies and infertility. This review focuses on the role of galectins in pregnancy-associated processes and discusses the relevance of galectin-glycan interactions as potential therapeutic targets in pregnancy disorders.
- Published
- 2019
- Full Text
- View/download PDF
25. Structure of the zebrafish galectin-1-L2 and model of its interaction with the infectious hematopoietic necrosis virus (IHNV) envelope glycoprotein.
- Author
-
Ghosh A, Banerjee A, Amzel LM, Vasta GR, and Bianchet MA
- Subjects
- Amino Acid Sequence, Animals, Glycoproteins metabolism, Infectious hematopoietic necrosis virus metabolism, Models, Molecular, Sequence Alignment, Zebrafish, Galectins chemistry, Galectins metabolism, Glycoproteins chemistry, Infectious hematopoietic necrosis virus chemistry, Zebrafish Proteins chemistry, Zebrafish Proteins metabolism
- Abstract
Galectins, highly conserved β-galactoside-binding lectins, have diverse regulatory roles in development and immune homeostasis and can mediate protective functions during microbial infection. In recent years, the role of galectins in viral infection has generated considerable interest. Studies on highly pathogenic viruses have provided invaluable insight into the participation of galectins in various stages of viral infection, including attachment and entry. Detailed mechanistic and structural aspects of these processes remain undetermined. To address some of these gaps in knowledge, we used Zebrafish as a model system to examine the role of galectins in infection by infectious hematopoietic necrosis virus (IHNV), a rhabdovirus that is responsible for significant losses in both farmed and wild salmonid fish. Like other rhabdoviruses, IHNV is characterized by an envelope consisting of trimers of a glycoprotein that display multiple N-linked oligosaccharides and play an integral role in viral infection by mediating the virus attachment and fusion. Zebrafish's proto-typical galectin Drgal1-L2 and the chimeric-type galectin Drgal3-L1 interact directly with the glycosylated envelope of IHNV, and significantly reduce viral attachment. In this study, we report the structure of the complex of Drgal1-L2 with N-acetyl-d-lactosamine at 2.0 Å resolution. To gain structural insight into the inhibitory effect of these galectins on IHNV attachment to the zebrafish epithelial cells, we modeled Drgal3-L1 based on human galectin-3, as well as, the ectodomain of the IHNV glycoprotein. These models suggest mechanisms for which the binding of these galectins to the IHNV glycoprotein hinders with different potencies the viral attachment required for infection., (© The Author(s) 2019. Published by Oxford University Press. All rights reserved. For permissions, please e-mail: journals.permissions@oup.com.)
- Published
- 2019
- Full Text
- View/download PDF
26. F-Type Lectins: A Highly Diversified Family of Fucose-Binding Proteins with a Unique Sequence Motif and Structural Fold, Involved in Self/Non-Self-Recognition.
- Author
-
Vasta GR, Amzel LM, Bianchet MA, Cammarata M, Feng C, and Saito K
- Abstract
The F-type lectin (FTL) family is one of the most recent to be identified and structurally characterized. Members of the FTL family are characterized by a fucose recognition domain [F-type lectin domain (FTLD)] that displays a novel jellyroll fold ("F-type" fold) and unique carbohydrate- and calcium-binding sequence motifs. This novel lectin family comprises widely distributed proteins exhibiting single, double, or greater multiples of the FTLD, either tandemly arrayed or combined with other structurally and functionally distinct domains, yielding lectin subunits of pleiotropic properties even within a single species. Furthermore, the extraordinary variability of FTL sequences (isoforms) that are expressed in a single individual has revealed genetic mechanisms of diversification in ligand recognition that are unique to FTLs. Functions of FTLs in self/non-self-recognition include innate immunity, fertilization, microbial adhesion, and pathogenesis, among others. In addition, although the F-type fold is distinctive for FTLs, a structure-based search revealed apparently unrelated proteins with minor sequence similarity to FTLs that displayed the FTLD fold. In general, the phylogenetic analysis of FTLD sequences from viruses to mammals reveals clades that are consistent with the currently accepted taxonomy of extant species. However, the surprisingly discontinuous distribution of FTLDs within each taxonomic category suggests not only an extensive structural/functional diversification of the FTLs along evolutionary lineages but also that this intriguing lectin family has been subject to frequent gene duplication, secondary loss, lateral transfer, and functional co-option.
- Published
- 2017
- Full Text
- View/download PDF
27. Binding of a C-type lectin's coiled-coil domain to the Domeless receptor directly activates the JAK/STAT pathway in the shrimp immune response to bacterial infection.
- Author
-
Sun JJ, Lan JF, Zhao XF, Vasta GR, and Wang JX
- Subjects
- Animals, Arthropod Proteins immunology, Crustacea microbiology, DNA, Complementary genetics, Janus Kinases metabolism, Lectins, C-Type chemistry, Receptors, Pattern Recognition metabolism, STAT Transcription Factors metabolism, Up-Regulation, Arthropod Proteins metabolism, Crustacea metabolism, Lectins, C-Type metabolism, Signal Transduction
- Abstract
C-type lectins (CTLs) are characterized by the presence of a C-type carbohydrate recognition domain (CTLD) that by recognizing microbial glycans, is responsible for their roles as pattern recognition receptors in the immune response to bacterial infection. In addition to the CTLD, however, some CTLs display additional domains that can carry out effector functions, such as the collagenous domain of the mannose-binding lectin. While in vertebrates, the mechanisms involved in these effector functions have been characterized in considerable detail, in invertebrates they remain poorly understood. In this study, we identified in the kuruma shrimp (Marsupenaeus japonicus) a structurally novel CTL (MjCC-CL) that in addition to the canonical CTLD, contains a coiled-coil domain (CCD) responsible for the effector functions that are key to the shrimp's antibacterial response mediated by antimicrobial peptides (AMPs). By the use of in vitro and in vivo experimental approaches we elucidated the mechanism by which the recognition of bacterial glycans by the CTLD of MjCC-CL leads to activation of the JAK/STAT pathway via interaction of the CCD with the surface receptor Domeless, and upregulation of AMP expression. Thus, our study of the shrimp MjCC-CL revealed a striking functional difference with vertebrates, in which the JAK/STAT pathway is indirectly activated by cell death and stress signals through cytokines or growth factors. Instead, by cross-linking microbial pathogens with the cell surface receptor Domeless, a lectin directly activates the JAK/STAT pathway, which plays a central role in the shrimp antibacterial immune responses by upregulating expression of selected AMPs.
- Published
- 2017
- Full Text
- View/download PDF
28. Functions of galectins as 'self/non-self'-recognition and effector factors.
- Author
-
Vasta GR, Feng C, González-Montalbán N, Mancini J, Yang L, Abernathy K, Frost G, and Palm C
- Subjects
- Animals, Humans, Galectins metabolism, Host-Pathogen Interactions, Immune Evasion, Immunity, Innate, Receptors, Immunologic metabolism
- Abstract
Carbohydrate structures on the cell surface encode complex information that through specific recognition by carbohydrate-binding proteins (lectins) modulates interactions between cells, cells and the extracellular matrix, or mediates recognition of potential microbial pathogens. Galectins are a family of ß-galactoside-binding lectins, which are evolutionary conserved and have been identified in most organisms, from fungi to invertebrates and vertebrates, including mammals. Since their discovery in the 1970s, their biological roles, initially understood as limited to recognition of endogenous carbohydrate ligands in embryogenesis and development, have expanded in recent years by the discovery of their roles in tissue repair and regulation of immune homeostasis. More recently, evidence has accumulated to support the notion that galectins can also bind glycans on the surface of potentially pathogenic microbes, and function as recognition and effector factors in innate immunity, thus establishing a new paradigm. Furthermore, some parasites 'subvert' the recognition roles of the vector/host galectins for successful attachment or invasion. These recent findings have revealed a striking functional diversification in this structurally conserved lectin family., (© FEMS 2017. All rights reserved. For permissions, please e-mail: journals.permissions@oup.com.)
- Published
- 2017
- Full Text
- View/download PDF
29. Comparison of proteomic profiles in the zebrafish retina during experimental degeneration and regeneration.
- Author
-
Eastlake K, Heywood WE, Tracey-White D, Aquino E, Bliss E, Vasta GR, Mills K, Khaw PT, Moosajee M, and Limb GA
- Subjects
- Animals, Apolipoproteins metabolism, Cell Membrane drug effects, Cell Membrane metabolism, Cytoskeletal Proteins metabolism, Eye Proteins metabolism, Fibrin metabolism, Gene Ontology, Histones metabolism, Injections, Ouabain administration & dosage, Ouabain pharmacology, Reproducibility of Results, Retina drug effects, Retina pathology, Retinal Degeneration pathology, Zebrafish Proteins metabolism, Proteomics methods, Regeneration drug effects, Retina metabolism, Retinal Degeneration metabolism, Zebrafish metabolism
- Abstract
Zebrafish spontaneously regenerate the retina after injury. Although the gene expression profile has been extensively studied in this species during regeneration, this does not reflect protein function. To further understand the regenerative process in the zebrafish, we compared the proteomic profile of the retina during injury and upon regeneration. Using two-dimensional difference gel electrophoresis (2D-DIGE) and label-free quantitative proteomics (quadrupole time of flight LC-MS/MS), we analysed the retina of adult longfin wildtype zebrafish at 0, 3 and 18 days after Ouabain injection. Gene ontology analysis indicates reduced metabolic processing, and increase in fibrin clot formation, with significant upregulation of fibrinogen gamma polypeptide, apolipoproteins A-Ib and A-II, galectin-1, and vitellogenin-6 during degeneration when compared to normal retina. In addition, cytoskeleton and membrane transport proteins were considerably altered during regeneration, with the highest fold upregulation observed for tubulin beta 2 A, histone H2B and brain type fatty acid binding protein. Key proteins identified in this study may play an important role in the regeneration of the zebrafish retina and investigations on the potential regulation of these proteins may lead to the design of protocols to promote endogenous regeneration of the mammalian retina following retinal degenerative disease.
- Published
- 2017
- Full Text
- View/download PDF
30. Transient Expression of Plasmodium berghei MSP8 and HAP2 in the Marine Protozoan Parasite Perkinsus marinus.
- Author
-
Cold ER, Vasta GR, and Robledo JA
- Subjects
- Alveolata genetics, Animals, Antigens, Protozoan immunology, Female, Gene Expression Regulation, Genetic Vectors, Malaria prevention & control, Male, Plasmids, Plasmodium berghei genetics, Protozoan Proteins immunology, Transfection, Alveolata immunology, Antigens, Protozoan metabolism, Malaria Vaccines genetics, Malaria Vaccines immunology, Plasmodium berghei immunology, Protozoan Proteins metabolism
- Abstract
Perkinsus marinus is a protozoan parasite of molluscs that can be propagated in vitro in a defined culture medium, in the absence of host cells. We previously reported that P. marinus trophozoites can be transfected with high efficiency by electroporation using a plasmid based on MOE, a highly expressed gene, and proposed its potential use as a "pseudoparasite." This is a novel gene expression platform for parasites of medical relevance for which the choice of the surrogate organism is based on phylogenetic affinity to the parasite of interest, while taking advantage of the whole engineered surrogate organism as a vaccination adjuvant. Here we improved the original transfection plasmid by incorporating a multicloning site, an enterokinase recognition sequence upstream of GFP, and a His-tag and demonstrate its potential suitability for the heterologous expression of Plasmodium sp. genes relevant to the development of anti-malarial vaccines. Plasmodium berghei HAP2 and MSP8, currently considered candidate genes for a malaria vaccine, were cloned into p[MOE]:GFP, and the constructs were used to transfect P. marinus trophozoites. Within 48 hr of transfection we observed fluorescent cells indicating that the P. berghei genes fused to GFP were expressed. The expression appeared to be transient for both P. berghei genes, as florescence of the transfectants diminished gradually over time. Although this heterologous expression system will require optimization for integration and constitutive expression of Plasmodium genes, our results represent attainment of proof for the "pseudoparasite" concept we previously proposed, as we show that the engineered P. marinus system has the potential to become a surrogate system suitable for expression of Plasmodium spp. genes of interest, which could eventually be used as a malaria vaccine delivery platform. The aim of the present study was to test the ability of marine protozoan parasite P. marinus to express genes of P. berghei .
- Published
- 2017
- Full Text
- View/download PDF
31. The zebrafish galectins Drgal1-L2 and Drgal3-L1 bind in vitro to the infectious hematopoietic necrosis virus (IHNV) glycoprotein and reduce viral adhesion to fish epithelial cells.
- Author
-
Nita-Lazar M, Mancini J, Feng C, González-Montalbán N, Ravindran C, Jackson S, de Las Heras-Sánchez A, Giomarelli B, Ahmed H, Haslam SM, Wu G, Dell A, Ammayappan A, Vakharia VN, and Vasta GR
- Subjects
- Animals, Cells, Cultured, Epithelial Cells virology, Galectins genetics, Host-Pathogen Interactions, Infectious hematopoietic necrosis virus pathogenicity, Recombinant Fusion Proteins genetics, Rhabdoviridae Infections transmission, Virulence, Virus Attachment, Zebrafish Proteins genetics, Epithelial Cells physiology, Galectins metabolism, Infectious hematopoietic necrosis virus immunology, Recombinant Fusion Proteins metabolism, Rhabdoviridae Infections immunology, Viral Envelope Proteins metabolism, Zebrafish immunology, Zebrafish Proteins metabolism
- Abstract
The infectious hematopoietic necrosis virus (IHNV; Rhabdoviridae, Novirhabdovirus) infects teleost fish, such as salmon and trout, and is responsible for significant losses in the aquaculture industry and in wild fish populations. Although IHNV enters the host through the skin at the base of the fins, the viral adhesion and entry mechanisms are not fully understood. In recent years, evidence has accumulated in support of the key roles played by protein-carbohydrate interactions between host lectins secreted to the extracellular space and virion envelope glycoproteins in modulating viral adhesion and infectivity. In this study, we assessed in vitro the potential role(s) of zebrafish (Danio rerio) proto type galectin-1 (Drgal1-L2) and a chimera galectin-3 (Drgal3-L1) in IHNV adhesion to epithelial cells. Our results suggest that the extracellular Drgal1-L2 and Drgal3-L1 interact directly and in a carbohydrate-dependent manner with the IHNV glycosylated envelope and glycans on the epithelial cell surface, significantly reducing viral adhesion., (Copyright © 2015 Elsevier Ltd. All rights reserved.)
- Published
- 2016
- Full Text
- View/download PDF
32. Galectins regulate the inflammatory response in airway epithelial cells exposed to microbial neuraminidase by modulating the expression of SOCS1 and RIG1.
- Author
-
Nita-Lazar M, Banerjee A, Feng C, and Vasta GR
- Subjects
- Animals, Bacterial Proteins pharmacology, Cell Line, Tumor, Cytokines biosynthesis, Cytokines metabolism, DEAD Box Protein 58, DEAD-box RNA Helicases immunology, DEAD-box RNA Helicases pharmacology, Epithelial Cells cytology, Epithelial Cells drug effects, Escherichia coli genetics, Escherichia coli metabolism, Extracellular Signal-Regulated MAP Kinases genetics, Extracellular Signal-Regulated MAP Kinases immunology, Galectin 1 biosynthesis, Galectin 1 immunology, Galectin 3 biosynthesis, Galectin 3 immunology, Gene Expression Regulation, Humans, Inflammation, Influenza A virus immunology, Janus Kinases genetics, Janus Kinases immunology, Mice, Neuraminidase pharmacology, Proto-Oncogene Proteins c-akt genetics, Proto-Oncogene Proteins c-akt immunology, Receptors, Immunologic, Recombinant Proteins biosynthesis, Recombinant Proteins immunology, Recombinant Proteins pharmacology, Respiratory Mucosa cytology, Respiratory Mucosa drug effects, Respiratory Mucosa immunology, STAT1 Transcription Factor genetics, STAT1 Transcription Factor immunology, Signal Transduction, Suppressor of Cytokine Signaling 1 Protein, Suppressor of Cytokine Signaling Proteins immunology, Suppressor of Cytokine Signaling Proteins pharmacology, DEAD-box RNA Helicases genetics, Epithelial Cells immunology, Galectin 1 pharmacology, Galectin 3 pharmacology, Suppressor of Cytokine Signaling Proteins genetics
- Abstract
Influenza patients frequently display increased susceptibility to Streptococcus pneumoniae co-infection and sepsis, the prevalent cause of mortality during influenza pandemics. However, the detailed mechanisms by which an influenza infection predisposes patients to suffer pneumococcal pneumonia are not fully understood. A murine model for influenza infection closely reflects the observations in human patients, since if the animals that have recovered from influenza A virus (IAV) sublethal infection are challenged with S. pneumoniae, they undergo a usually fatal uncontrolled cytokine response. We have previously demonstrated both in vitro and in vivo that the expression and secretion of galectin-1 (Gal1) and galectin-3 (Gal3) are modulated during IAV infection, and that the viral neuraminidase unmasks galactosyl moieties in the airway epithelia. In this study we demonstrate in vitro that the binding of secreted Gal1 and Gal3 to the epithelial cell surface modulates the expression of SOCS1 and RIG1, and activation of ERK, AKT or JAK/STAT1 signaling pathways, leading to a disregulated expression and release of pro-inflammatory cytokines. Our results suggest that the activity of the viral and pneumococcal neuraminidases on the surface of the airway epithelial cells function as a "danger signal" that leads to rapid upregulation of SOCS1 expression to prevent an uncontrolled inflammatory response. The binding of extracellular Gal1 or Gal3 to the galactosyl moieties unmasked on the surface of airway epithelial cells can either "fine-tune" or severely disregulate this process, respectively, the latter potentially leading to hypercytokinemia., (Copyright © 2015 Elsevier Ltd. All rights reserved.)
- Published
- 2015
- Full Text
- View/download PDF
33. Structural, functional, and evolutionary aspects of galectins in aquatic mollusks: From a sweet tooth to the Trojan horse.
- Author
-
Vasta GR, Feng C, Bianchet MA, Bachvaroff TR, and Tasumi S
- Subjects
- Animals, Galectins metabolism, Mollusca metabolism, Evolution, Molecular, Galectins genetics, Mollusca genetics, Mollusca immunology
- Abstract
Galectins constitute a conserved and widely distributed lectin family characterized by their binding affinity for β-galactosides and a unique binding site sequence motif in the carbohydrate recognition domain (CRD). In spite of their structural conservation, galectins display a remarkable functional diversity, by participating in developmental processes, cell adhesion and motility, regulation of immune homeostasis, and recognition of glycans on the surface of viruses, bacteria and protozoan parasites. In contrast with mammals, and other vertebrate and invertebrate taxa, the identification and characterization of bona fide galectins in aquatic mollusks has been relatively recent. Most of the studies have focused on the identification and domain organization of galectin-like transcripts or proteins in diverse tissues and cell types, including hemocytes, and their expression upon environmental or infectious challenge. Lectins from the eastern oyster Crassostrea virginica, however, have been characterized in their molecular, structural and functional aspects and some notable features have become apparent in the galectin repertoire of aquatic mollusks. These including less diversified galectin repertoires and different domain organizations relative to those observed in vertebrates, carbohydrate specificity for blood group oligosaccharides, and up regulation of galectin expression by infectious challenge, a feature that supports their proposed role(s) in innate immune responses. Although galectins from some aquatic mollusks have been shown to recognize microbial pathogens and parasites and promote their phagocytosis, they can also selectively bind to phytoplankton components, suggesting that they also participate in uptake and intracellular digestion of microalgae. In addition, the experimental evidence suggests that the protozoan parasite Perkinsus marinus has co-evolved with the oyster host to be selectively recognized by the oyster hemocyte galectins over algal food or bacterial pathogens, thereby subverting the oyster's innate immune/feeding recognition mechanisms to gain entry into the host cells., (Copyright © 2015 Elsevier Ltd. All rights reserved.)
- Published
- 2015
- Full Text
- View/download PDF
34. Galectin CvGal2 from the Eastern Oyster (Crassostrea virginica) Displays Unique Specificity for ABH Blood Group Oligosaccharides and Differentially Recognizes Sympatric Perkinsus Species.
- Author
-
Feng C, Ghosh A, Amin MN, Bachvaroff TR, Tasumi S, Pasek M, Banerjee A, Shridhar S, Wang LX, Bianchet MA, and Vasta GR
- Subjects
- Animals, Hemocytes chemistry, Hemocytes metabolism, Hemocytes parasitology, Alveolata chemistry, Alveolata genetics, Alveolata metabolism, Blood Group Antigens chemistry, Blood Group Antigens genetics, Blood Group Antigens metabolism, Crassostrea chemistry, Crassostrea genetics, Crassostrea metabolism, Crassostrea parasitology, Galectins chemistry, Galectins genetics, Galectins metabolism, Oligosaccharides chemistry, Oligosaccharides genetics, Oligosaccharides metabolism, Phylogeny
- Abstract
Galectins are highly conserved lectins that are key to multiple biological functions, including pathogen recognition and regulation of immune responses. We previously reported that CvGal1, a galectin expressed in phagocytic cells (hemocytes) of the eastern oyster (Crassostrea virginica), is hijacked by the parasite Perkinsus marinus to enter the host, where it causes systemic infection and death. Screening of an oyster hemocyte cDNA library revealed a novel galectin, which we designated CvGal2, with four tandemly arrayed carbohydrate recognition domains (CRDs). Phylogentic analysis of the CvGal2 CRDs suggests close relationships with homologous CRDs from CvGal1. Glycan array analysis, however, revealed that, unlike CvGal1 which preferentially binds to the blood group A tetrasaccharide, CvGal2 recognizes both blood group A and B tetrasaccharides and related structures, suggesting that CvGal2 has broader binding specificity. Furthermore, SPR analysis demonstrated significant differences in the binding kinetics of CvGal1 and CvGal2, and structural modeling revealed substantial differences in their interactions with the oligosaccharide ligands. CvGal2 is homogeneously distributed in the hemocyte cytoplasm, is released to the extracellular space, and binds to the hemocyte surface. CvGal2 binds to P. marinus trophozoites in a dose-dependent and β-galactoside-specific manner. Strikingly, negligible binding of CvGal2 was observed for Perkinsus chesapeaki, a sympatric parasite species mostly prevalent in the clams Mya arenaria and Macoma balthica. The differential recognition of Perkinsus species by the oyster galectins is consistent with their relative prevalence in oyster and clam species and supports their role in facilitating parasite entry and infectivity in a host-preferential manner.
- Published
- 2015
- Full Text
- View/download PDF
35. Desialylation of airway epithelial cells during influenza virus infection enhances pneumococcal adhesion via galectin binding.
- Author
-
Nita-Lazar M, Banerjee A, Feng C, Amin MN, Frieman MB, Chen WH, Cross AS, Wang LX, and Vasta GR
- Subjects
- Adhesins, Bacterial, Animals, Apoptosis, Bacterial Adhesion physiology, Cell Line, Disease Susceptibility, Epithelial Cells metabolism, Galectin 1 biosynthesis, Galectin 3 biosynthesis, Hemagglutinin Glycoproteins, Influenza Virus, Humans, Influenza A virus pathogenicity, Mice, Mice, Inbred C57BL, Neuraminidase pharmacology, Protein Binding drug effects, Respiratory Mucosa microbiology, Respiratory Mucosa virology, Streptococcus pneumoniae pathogenicity, Galectin 1 metabolism, Galectin 3 metabolism, Orthomyxoviridae Infections pathology, Pneumococcal Infections pathology, Respiratory Mucosa cytology
- Abstract
The continued threat of worldwide influenza pandemics, together with the yearly emergence of antigenically drifted influenza A virus (IAV) strains, underscore the urgent need to elucidate not only the mechanisms of influenza virulence, but also those mechanisms that predispose influenza patients to increased susceptibility to subsequent infection with Streptococcus pneumoniae. Glycans displayed on the surface of epithelia that are exposed to the external environment play important roles in microbial recognition, adhesion, and invasion. It is well established that the IAV hemagglutinin and pneumococcal adhesins enable their attachment to the host epithelia. Reciprocally, the recognition of microbial glycans by host carbohydrate-binding proteins (lectins) can initiate innate immune responses, but their relevance in influenza or pneumococcal infections is poorly understood. Galectins are evolutionarily conserved lectins characterized by affinity for β-galactosides and a unique sequence motif, with critical regulatory roles in development and immune homeostasis. In this study, we examined the possibility that galectins expressed in the airway epithelial cells might play a significant role in viral or pneumococcal adhesion to airway epithelial cells. Our results in a mouse model for influenza and pneumococcal infection revealed that the murine lung expresses a diverse galectin repertoire, from which selected galectins, including galectin 1 (Gal1) and galectin 3 (Gal3), are released to the bronchoalveolar space. Further, the results showed that influenza and subsequent S. pneumoniae infections significantly alter the glycosylation patterns of the airway epithelial surface and modulate galectin expression. In vitro studies on the human airway epithelial cell line A549 were consistent with the observations made in the mouse model, and further revealed that both Gal1 and Gal3 bind strongly to IAV and S. pneumoniae, and that exposure of the cells to viral neuraminidase or influenza infection increased galectin-mediated S. pneumoniae adhesion to the cell surface. Our results suggest that upon influenza infection, pneumococcal adhesion to the airway epithelial surface is enhanced by an interplay among the host galectins and viral and pneumococcal neuraminidases. The observed enhancement of pneumococcal adhesion may be a contributing factor to the observed hypersusceptibility to pneumonia of influenza patients., (Copyright © 2014 Elsevier Ltd. All rights reserved.)
- Published
- 2015
- Full Text
- View/download PDF
36. Manipulating galectin expression in zebrafish (Danio rerio).
- Author
-
Feng C, Nita-Lazar M, González-Montalbán N, Wang J, Mancini J, Ravindran C, Ahmed H, and Vasta GR
- Subjects
- Animals, Base Sequence, Embryo, Nonmammalian, Female, Galectins deficiency, Injections, Male, Morpholinos genetics, Phenotype, RNA genetics, Zebrafish embryology, Galectins genetics, Gene Expression Regulation genetics, Gene Knockdown Techniques methods, Gene Knockout Techniques methods, Zebrafish genetics
- Abstract
Techniques for disrupting gene expression are invaluable tools for the analysis of the biological role(s) of a gene product. Because of its genetic tractability and multiple advantages over conventional mammalian models, the zebrafish (Danio rerio) is recognized as a powerful system for gaining new insight into diverse aspects of human health and disease. Among the multiple mammalian gene families for which the zebrafish has shown promise as an invaluable model for functional studies, the galectins have attracted great interest due to their participation in early development, regulation of immune homeostasis, and recognition of microbial pathogens. Galectins are β-galactosyl-binding lectins with a characteristic sequence motif in their carbohydrate recognition domains (CRDs), which comprise an evolutionary conserved family ubiquitous in eukaryotic taxa. Galectins are emerging as key players in the modulation of many important pathological processes, which include acute and chronic inflammatory diseases, autoimmunity and cancer, thus making them potential molecular targets for innovative drug discovery. Here, we provide a review of the current methods available for the manipulation of gene expression in the zebrafish, with a focus on gene knockdown [morpholino (MO)-derived antisense oligonucleotides] and knockout (CRISPR-Cas) technologies.
- Published
- 2015
- Full Text
- View/download PDF
37. Protozoan parasites of bivalve molluscs: literature follows culture.
- Author
-
Fernández Robledo JA, Vasta GR, and Record NR
- Subjects
- Animals, Databases as Topic, Bivalvia parasitology, Parasites physiology, Publications
- Abstract
Bivalve molluscs are key components of the estuarine environments as contributors to the trophic chain, and as filter -feeders, for maintaining ecosystem integrity. Further, clams, oysters, and scallops are commercially exploited around the world both as traditional local shellfisheries, and as intensive or semi-intensive farming systems. During the past decades, populations of those species deemed of environmental or commercial interest have been subject to close monitoring given the realization that these can suffer significant decline, sometimes irreversible, due to overharvesting, environmental pollution, or disease. Protozoans of the genera Perkinsus, Haplosporidium, Marteilia, and Bonamia are currently recognized as major threats for natural and farmed bivalve populations. Since their identification, however, the variable publication rates of research studies addressing these parasitic diseases do not always appear to reflect their highly significant environmental and economic impact. Here we analyzed the peer- reviewed literature since the initial description of these parasites with the goal of identifying potential milestone discoveries or achievements that may have driven the intensity of the research in subsequent years, and significantly increased publication rates. Our analysis revealed that after initial description of the parasite as the etiological agent of a given disease, there is a time lag before a maximal number of yearly publications are reached. This has already taken place for most of them and has been followed by a decrease in publication rates over the last decade (20- to 30- year lifetime in the literature). Autocorrelation analyses, however, suggested that advances in parasite purification and culture methodologies positively drive publication rates, most likely because they usually lead to novel molecular tools and resources, promoting mechanistic studies. Understanding these trends should help researchers in prioritizing research efforts for these and other protozoan parasites, together with their development as model systems for further basic and translational research in parasitic diseases.
- Published
- 2014
- Full Text
- View/download PDF
38. A rhamnose-binding lectin from sea bass (Dicentrarchus labrax) plasma agglutinates and opsonizes pathogenic bacteria.
- Author
-
Cammarata M, Parisi MG, Benenati G, Vasta GR, and Parrinello N
- Subjects
- Agglutination, Amino Acid Motifs genetics, Amino Acid Sequence, Animals, Immunity, Innate, Lectins genetics, Lectins isolation & purification, Molecular Sequence Data, Phagocytosis, Phylogeny, Protein Binding, Protein Multimerization, Protein Structure, Tertiary genetics, Rabbits, Rhamnose metabolism, Bacterial Infections immunology, Bass immunology, Erythrocytes metabolism, Escherichia coli immunology, Lectins metabolism, Macrophages, Peritoneal immunology, Plasma metabolism
- Abstract
The discovery of rhamnose-binding lectins (RBLs) in teleost fish eggs led to the identification of a novel lectin family characterized by a unique sequence motif and a structural fold, and initially proposed to modulate fertilization. Further studies of the RBL tissue localization and gene organization were also suggestive of role(s) in innate immunity. Here we describe the purification, and biochemical and functional characterization of a novel RBL (DlRBL) from sea bass (Dicentrarchus labrax) serum. The purified DlRBL had electrophoretic mobilities corresponding to 24 kDa and 100 kDa under reducing and non-reducing conditions, respectively, suggesting that in plasma the DlRBL is present as a physiological homotetramer. DlRBL subunit transcripts revealed an open reading frame encoding 212 amino acid residues that included two tandemly-arrayed carbohydrate-recognition domains, and an 18-residue signal sequence at the N-terminus. The deduced size of 24.1 kDa for the mature protein was in good agreement with the subunit size of the isolated lectin. Binding activity of DlRBL for rabbit erythrocytes could be inhibited in the presence of rhamnose or galactose, did not require calcium, and was optimal at around 20°C and within the pH 6.5-8.0 range. DlRBL agglutinated Gram positive and Gram negative bacteria, and exposure of formalin-killed Escherichia coli to DlRBL enhanced their phagocytosis by D. labrax peritoneal macrophages relative to the unexposed controls. Taken together, the results suggest that plasma DlRBL may play a role in immune recognition of microbial pathogens and facilitate their clearance by phagocytosis., (Copyright © 2014 Elsevier Ltd. All rights reserved.)
- Published
- 2014
- Full Text
- View/download PDF
39. A shrimp C-type lectin inhibits proliferation of the hemolymph microbiota by maintaining the expression of antimicrobial peptides.
- Author
-
Wang XW, Xu JD, Zhao XF, Vasta GR, and Wang JX
- Subjects
- Animals, Base Sequence, Crustacea, DNA Primers, Anti-Infective Agents pharmacology, Hemolymph microbiology, Lectins, C-Type metabolism, Microbiota drug effects, Peptides pharmacology
- Abstract
Some aquatic invertebrates such as shrimp contain low albeit stable numbers of bacteria in the circulating hemolymph. The proliferation of this hemolymph microbiota in such a nutrient-rich environment is tightly controlled in healthy animals, but the mechanisms responsible had remained elusive. In the present study, we report a C-type lectin (MjHeCL) from the kuruma shrimp (Marsupenaeus japonicus) that participates in restraining the hemolymph microbiota. Although the expression of MjHeCL did not seem to be modulated by bacterial challenge, the down-regulation of its expression by RNA interference led to proliferation of the hemolymph microbiota, ultimately resulting in shrimp death. This phenotype was rescued by the injection of recombinant MjHeCL, which restored the healthy status of the knockdown shrimp. A mechanistic analysis revealed that MjHeCL inhibited bacterial proliferation by modulating the expression of antimicrobial peptides. The key function of MjHeCL in the shrimp immune homeostasis might be related to its broader recognition spectrum of the hemolymph microbiota components than other lectins. Our study demonstrates the role of MjHeCL in maintaining the healthy status of shrimp and provides new insight into the biological significance of C-type lectins, a diversified and abundant lectin family in invertebrate species.
- Published
- 2014
- Full Text
- View/download PDF
40. A galectin from the kuruma shrimp (Marsupenaeus japonicus) functions as an opsonin and promotes bacterial clearance from hemolymph.
- Author
-
Shi XZ, Wang L, Xu S, Zhang XW, Zhao XF, Vasta GR, and Wang JX
- Subjects
- Agglutination, Animals, Cloning, Molecular, DNA, Complementary, Decapoda microbiology, Disease Resistance genetics, Disease Resistance immunology, Galectins metabolism, Gene Expression Regulation, Gene Silencing, Hemocytes immunology, Hemocytes metabolism, Hemolymph metabolism, Host-Pathogen Interactions genetics, Host-Pathogen Interactions immunology, Lipopolysaccharides metabolism, Opsonin Proteins metabolism, Phagocytosis genetics, Phagocytosis immunology, Phylogeny, Protein Binding, Recombinant Proteins genetics, Recombinant Proteins metabolism, Bacteria immunology, Decapoda genetics, Decapoda immunology, Galectins genetics, Hemolymph microbiology, Opsonin Proteins genetics
- Abstract
Galectins are a lectin family characterized by a conserved sequence motif in the carbohydrate recognition domain, which preferential binds to galactosyl moieties. However, few studies about the biological roles of galectins in invertebrates have been reported except for the galectin (CvGal1) from the eastern oyster Crassostrea virginica. Furthermore, galectins have been described in only a few crustacean species, and no functional studies have been reported so far. In this study, we identified and functionally characterized a galectin from the kuruma shrimp Marsupenaeus japonicus, which we designated MjGal. Upon Vibrio anguillarum challenge, expression of MjGal was up-regulated mostly in hemocytes and hepatopancreas, and the protein bound to both Gram-positive and Gram-negative bacteria through the recognition of lipoteichoic acid (LTA) or lipopolysaccharide (LPS), respectively. By also binding to the shrimp hemocyte surface, MjGal functions as an opsonin for microbial pathogens, promoting their phagocytosis. Further, as shown by RNA interference, MjGal participates in clearance of bacteria from circulation, and thereby contributes to the shrimp's immune defense against infectious challenge. Elucidation of functional and mechanistic aspects of shrimp immunity will enable the development of novel strategies for intervention in infectious diseases currently affecting the shrimp farming industry worldwide.
- Published
- 2014
- Full Text
- View/download PDF
41. Humanized HLA-DR4 mice fed with the protozoan pathogen of oysters Perkinsus marinus (Dermo) do not develop noticeable pathology but elicit systemic immunity.
- Author
-
Wijayalath W, Majji S, Kleschenko Y, Pow-Sang L, Brumeanu TD, Villasante EF, Vasta GR, Fernández-Robledo JA, and Casares S
- Subjects
- Animals, HLA-DR4 Antigen genetics, Humans, Interferon-gamma immunology, Mice, Mice, Transgenic, Alveolata immunology, HLA-DR4 Antigen immunology, Ostreidae parasitology, Shellfish parasitology
- Abstract
Perkinsus marinus (Phylum Perkinsozoa) is a marine protozoan parasite responsible for "Dermo" disease in oysters, which has caused extensive damage to the shellfish industry and estuarine environment. The infection prevalence has been estimated in some areas to be as high as 100%, often causing death of infected oysters within 1-2 years post-infection. Human consumption of the parasites via infected oysters is thus likely to occur, but to our knowledge the effect of oral consumption of P. marinus has not been investigated in humans or other mammals. To address the question we used humanized mice expressing HLA-DR4 molecules and lacking expression of mouse MHC-class II molecules (DR4.EA(0)) in such a way that CD4 T cell responses are solely restricted by the human HLA-DR4 molecule. The DR4.EA(0) mice did not develop diarrhea or any detectable pathology in the gastrointestinal tract or lungs following single or repeated feedings with live P. marinus parasites. Furthermore, lymphocyte populations in the gut associated lymphoid tissue and spleen were unaltered in the parasite-fed mice ruling out local or systemic inflammation. Notably, naïve DR4.EA(0) mice had antibodies (IgM and IgG) reacting against P. marinus parasites whereas parasite specific T cell responses were undetectable. Feeding with P. marinus boosted the antibody responses and stimulated specific cellular (IFNγ) immunity to the oyster parasite. Our data indicate the ability of P. marinus parasites to induce systemic immunity in DR4.EA(0) mice without causing noticeable pathology, and support rationale grounds for using genetically engineered P. marinus as a new oral vaccine platform to induce systemic immunity against infectious agents.
- Published
- 2014
- Full Text
- View/download PDF
42. Hemocytes and plasma of the eastern oyster (Crassostrea virginica) display a diverse repertoire of sulfated and blood group A-modified N-glycans.
- Author
-
Kurz S, Jin C, Hykollari A, Gregorich D, Giomarelli B, Vasta GR, Wilson IB, and Paschinger K
- Subjects
- ABO Blood-Group System chemistry, Alveolata physiology, Animals, Blood Proteins chemistry, Crassostrea chemistry, Crassostrea parasitology, Epitopes chemistry, Epitopes metabolism, Galectins chemistry, Hemocytes chemistry, Hemocytes parasitology, Host-Parasite Interactions physiology, Polysaccharides chemistry, ABO Blood-Group System metabolism, Blood Proteins metabolism, Crassostrea metabolism, Galectins metabolism, Hemocytes metabolism, Polysaccharides metabolism
- Abstract
The eastern oyster (Crassostrea virginica) has become a useful model system for glycan-dependent host-parasite interactions due to the hijacking of the oyster galectin CvGal1 for host entry by the protozoan parasite Perkinsus marinus, the causative agent of Dermo disease. In this study, we examined the N-glycans of both the hemocytes, which via CvGal1 are the target of the parasite, and the plasma of the oyster. In combination with HPLC fractionation, exoglycosidase digestion, and fragmentation of the glycans, mass spectrometry revealed that the major N-glycans of plasma are simple hybrid structures, sometimes methylated and core α1,6-fucosylated, with terminal β1,3-linked galactose; a remarkable high degree of sulfation of such glycans was observed. Hemocytes express a larger range of glycans, including core-difucosylated paucimannosidic forms, whereas bi- and triantennary glycans were found in both sources, including structures carrying sulfated and methylated variants of the histo-blood group A epitope. The primary features of the oyster whole hemocyte N-glycome were also found in dominin, the major plasma glycoprotein, which had also been identified as a CvGal1 glycoprotein ligand associated with hemocytes. The occurrence of terminal blood group moieties on oyster dominin and on hemocyte surfaces can account in part for their affinity for the endogenous CvGal1.
- Published
- 2013
- Full Text
- View/download PDF
43. The galectin CvGal1 from the eastern oyster (Crassostrea virginica) binds to blood group A oligosaccharides on the hemocyte surface.
- Author
-
Feng C, Ghosh A, Amin MN, Giomarelli B, Shridhar S, Banerjee A, Fernández-Robledo JA, Bianchet MA, Wang LX, Wilson IB, and Vasta GR
- Subjects
- ABO Blood-Group System genetics, ABO Blood-Group System metabolism, Animals, Crassostrea genetics, Crassostrea metabolism, Crassostrea parasitology, Galectins genetics, Galectins metabolism, Hemocytes metabolism, Hemocytes parasitology, Oligosaccharides genetics, Oligosaccharides metabolism, Protein Binding, Proteomics methods, ABO Blood-Group System chemistry, Crassostrea chemistry, Galectins chemistry, Hemocytes chemistry, Oligosaccharides chemistry
- Abstract
The galectin CvGal1 from the eastern oyster (Crassostrea virginica), which possesses four tandemly arrayed carbohydrate recognition domains, was previously shown to display stronger binding to galactosamine and N-acetylgalactosamine relative to d-galactose. CvGal1 expressed by phagocytic cells is "hijacked" by the parasite Perkinsus marinus to enter the host, where it proliferates and causes systemic infection and death. In this study, a detailed glycan array analysis revealed that CvGal1 preferentially recognizes type 2 blood group A oligosaccharides. Homology modeling of the protein and its oligosaccharide ligands supported this preference over type 1 blood group A and B oligosaccharides. The CvGal ligand models were further validated by binding, inhibition, and competitive binding studies of CvGal1 and ABH-specific monoclonal antibodies with intact and deglycosylated glycoproteins, hemocyte extracts, and intact hemocytes and by surface plasmon resonance analysis. A parallel glycomic study carried out on oyster hemocytes (Kurz, S., Jin, C., Hykollari, A., Gregorich, D., Giomarelli, B., Vasta, G. R., Wilson, I. B. H., and Paschinger, K. (2013) J. Biol. Chem. 288) determined the structures of oligosaccharides recognized by CvGal1. Proteomic analysis of the hemocyte glycoproteins identified β-integrin and dominin as CvGal1 "self"-ligands. Despite strong CvGal1 binding to P. marinus trophozoites, no binding of ABH blood group antibodies was observed. Thus, parasite glycans structurally distinct from the blood group A oligosaccharides on the hemocyte surface may function as potentially effective ligands for CvGal1. We hypothesize that carbohydrate-based mimicry resulting from the host/parasite co-evolution facilitates CvGal1-mediated cross-linking to β-integrin, located on the hemocyte surface, leading to cell activation, phagocytosis, and host infection.
- Published
- 2013
- Full Text
- View/download PDF
44. Quantitative assessment of the proliferation of the protozoan parasite Perkinsus marinus using a bioluminescence assay for ATP content.
- Author
-
Shridhar S, Hassan K, Sullivan DJ, Vasta GR, and Fernández Robledo JA
- Abstract
Perkinsus marinus is a protozoan parasite that causes "Dermo" disease in the eastern oyster Crasssostrea virginica in coastal areas of the USA. Until now, intervention strategies against the parasite have found limited success, and Dermo still remains one of the main hurdles for the restoration of oyster populations. We adapted a commercial adenosine tri-phosphate (ATP) content-based assay to assess the in vitro proliferation of P. marinus in a 96-well plate format, and validated the method by measuring the effects of potential anti-proliferative compounds. The sensitivity (1.5-3.1 × 10(4) cells/well), linearity (R (2) = 0.983), and signal stability (60 min) support the reliability of the assay for assessing cell proliferation. Validation of the assay by culturing P. marinus in the presence of increasing concentrations of triclosan showed a dose-response profile. The IC50 value obtained was higher than that reported earlier, possibly due to the use of different viability assay methods and a different P. marinus strain. The antibiotics G418 and tetracycline and the herbicide fluridone were active against P. marinus proliferation; the IC50 of chloramphenicol, ciprofloxacin, and atrazine was relatively high suggesting either off-target effects or inability to reach the targets. The validation of the ATP-based assay, together with significant advantages of the Perkinsus culture methodology (homogeneity, reproducibility, and high cell densities), underscores the value of this assay for developing high-throughput screens for the identification of novel leader compounds against Perkinsus species, and most importantly, for the closely-related apicomplexan parasites.
- Published
- 2013
- Full Text
- View/download PDF
45. Design and synthesis of glycoprotein-based multivalent glyco-ligands for influenza hemagglutinin and human galectin-3.
- Author
-
Wang H, Huang W, Orwenyo J, Banerjee A, Vasta GR, and Wang LX
- Subjects
- Animals, Carbohydrate Sequence, Cell Line, Tumor, Chickens, Click Chemistry, Drug Design, Glycoconjugates chemical synthesis, Humans, Ligands, Models, Molecular, Molecular Sequence Data, Serum Albumin chemical synthesis, Serum Albumin chemistry, Serum Albumin pharmacology, Galectin 3 metabolism, Glycoconjugates chemistry, Glycoconjugates pharmacology, Hemagglutinin Glycoproteins, Influenza Virus metabolism, Influenza, Human virology, Orthomyxoviridae metabolism
- Abstract
We report a facile synthesis of glycoprotein-based glyco-ligands and their binding with influenza hemagglutinin and human galectin-3. Human serum albumin (HSA) was used as the scaffold and an Asn-linked complex type N-glycan prepared from chicken eggs was used as the glycan building block. It was found that Cu(I)-catalyzed alkyne-azide cycloaddition reaction (click chemistry) between the alkyne-labeled glycan and the azide-tagged HSA led to an efficient formation of the glycoconjugates. The density of glycan ligands on the protein scaffold was readily varied by changing the molar ratios of the two reactants. Binding studies indicated that the sialylated and desialylated multivalent glycoligands could selectively bind to influenza hemagglutinin and human galectin-3, respectively, with high affinity. In the two glycan-lectin interactions, a clear multivalent effect was observed. Moreover, a cell-based assay showed that the synthetic multivalent glyco-ligands could efficiently inhibit the attachment of galectin-3 to human prostate cancer and lung cancer cell lines. This study suggests that the synthetic glycoprotein-based glyco-ligands can be useful for different applications, including blocking the function of galectin-3 in cancer metastasis., (Copyright © 2013 Elsevier Ltd. All rights reserved.)
- Published
- 2013
- Full Text
- View/download PDF
46. Cod glycopeptide with picomolar affinity to galectin-3 suppresses T-cell apoptosis and prostate cancer metastasis.
- Author
-
Guha P, Kaptan E, Bandyopadhyaya G, Kaczanowska S, Davila E, Thompson K, Martin SS, Kalvakolanu DV, Vasta GR, and Ahmed H
- Subjects
- Adenocarcinoma metabolism, Adenocarcinoma pathology, Animals, Antifreeze Proteins chemistry, Antifreeze Proteins isolation & purification, Antigens, Tumor-Associated, Carbohydrate chemistry, Antigens, Tumor-Associated, Carbohydrate metabolism, Antineoplastic Agents chemistry, Antineoplastic Agents isolation & purification, Fish Proteins chemistry, Fish Proteins isolation & purification, Human Umbilical Vein Endothelial Cells, Humans, Jurkat Cells, Male, Mice, Neoplasm Metastasis, Neovascularization, Pathologic drug therapy, Neovascularization, Pathologic metabolism, Neovascularization, Pathologic pathology, Prostatic Neoplasms metabolism, Prostatic Neoplasms pathology, Adenocarcinoma drug therapy, Antifreeze Proteins pharmacology, Antineoplastic Agents pharmacology, Apoptosis drug effects, Fish Proteins pharmacology, Gadus morhua, Galectin 3 metabolism, Neoplasm Proteins metabolism, Prostatic Neoplasms drug therapy, T-Lymphocytes metabolism
- Abstract
Cancer metastasis and immune suppression are critical issues in cancer therapy. Here, we show that a β-galactoside-binding lectin [galectin-3 (gal3)] that recognizes the Thomsen-Friedenreich disaccharide (TFD, Galβ1,3GalNAc) present on the surface of most cancer cells is involved in promoting angiogenesis, tumor-endothelial cell adhesion, and metastasis of prostate cancer cells, as well as evading immune surveillance through killing of activated T cells. To block gal3-mediated interactions, we purified a glycopeptide from cod (designated TFD100) that binds gal3 with picomolar affinity. TFD100 blocks gal3-mediated angiogenesis, tumor-endothelial cell interactions, and metastasis of prostate cancer cells in mice at nanomolar levels. Moreover, apoptosis of activated T cells induced by either recombinant gal3 or prostate cancer patient serum-associated gal3 was inhibited at nanomolar concentration of TFD100. Because the gal3-TFD interaction is a key factor driving metastasis in most epithelial cancers, this high-affinity TFD100 should be a promising antimetastatic agent for the treatment of various cancers, including prostate adenocarcinoma.
- Published
- 2013
- Full Text
- View/download PDF
47. Galectins as self/non-self recognition receptors in innate and adaptive immunity: an unresolved paradox.
- Author
-
Vasta GR, Ahmed H, Nita-Lazar M, Banerjee A, Pasek M, Shridhar S, Guha P, and Fernández-Robledo JA
- Abstract
Galectins are characterized by their binding affinity for β-galactosides, a unique binding site sequence motif, and wide taxonomic distribution and structural conservation in vertebrates, invertebrates, protista, and fungi. Since their initial description, galectins were considered to bind endogenous ("self") glycans and mediate developmental processes and cancer. In the past few years, however, numerous studies have described the diverse effects of galectins on cells involved in both innate and adaptive immune responses, and the mechanistic aspects of their regulatory roles in immune homeostasis. More recently, however, evidence has accumulated to suggest that galectins also bind exogenous ("non-self") glycans on the surface of potentially pathogenic microbes, parasites, and fungi, suggesting that galectins can function as pattern recognition receptors (PRRs) in innate immunity. Thus, a perplexing paradox arises by the fact that galectins also recognize lactosamine-containing glycans on the host cell surface during developmental processes and regulation of immune responses. According to the currently accepted model for non-self recognition, PRRs recognize pathogens via highly conserved microbial surface molecules of wide distribution such as LPS or peptidoglycan (pathogen-associated molecular patterns; PAMPs), which are absent in the host. Hence, this would not apply to galectins, which apparently bind similar self/non-self molecular patterns on host and microbial cells. This paradox underscores first, an oversimplification in the use of the PRR/PAMP terminology. Second, and most importantly, it reveals significant gaps in our knowledge about the diversity of the host galectin repertoire, and the subcellular targeting, localization, and secretion. Furthermore, our knowledge about the structural and biophysical aspects of their interactions with the host and microbial carbohydrate moieties is fragmentary, and warrants further investigation.
- Published
- 2012
- Full Text
- View/download PDF
48. Diversity in recognition of glycans by F-type lectins and galectins: molecular, structural, and biophysical aspects.
- Author
-
Vasta GR, Ahmed H, Bianchet MA, Fernández-Robledo JA, and Amzel LM
- Subjects
- Animals, Carbohydrate Sequence, Fucose chemistry, Fucose metabolism, Fucose physiology, Galectins metabolism, Humans, Lectins metabolism, Molecular Sequence Data, Polysaccharides metabolism, Structure-Activity Relationship, Biodiversity, Galectins chemistry, Galectins physiology, Lectins chemistry, Lectins physiology, Polysaccharides chemistry, Polysaccharides physiology
- Abstract
Although lectins are "hard-wired" in the germline, the presence of tandemly arrayed carbohydrate recognition domains (CRDs), of chimeric structures displaying distinct CRDs, of polymorphic genes resulting in multiple isoforms, and in some cases, of a considerable recognition plasticity of their carbohydrate binding sites, significantly expand the lectin ligand-recognition spectrum and lectin functional diversification. Analysis of structural/functional aspects of galectins and F-lectins-the most recently identified lectin family characterized by a unique CRD sequence motif (a distinctive structural fold) and nominal specificity for l-Fuc-has led to a greater understanding of self/nonself recognition by proteins with tandemly arrayed CRDs. For lectins with a single CRD, however, recognition of self and nonself glycans can only be rationalized in terms of protein oligomerization and ligand clustering and presentation. Spatial and temporal changes in lectin expression, secretion, and local concentrations in extracellular microenvironments, as well as structural diversity and spatial display of their carbohydrate ligands on the host or microbial cell surface, are suggestive of a dynamic interplay of their recognition and effector functions in development and immunity., (© 2012 New York Academy of Sciences.)
- Published
- 2012
- Full Text
- View/download PDF
49. Galectins as pattern recognition receptors: structure, function, and evolution.
- Author
-
Vasta GR
- Subjects
- Animals, Galectins chemistry, Galectins genetics, Humans, Receptors, Pattern Recognition chemistry, Receptors, Pattern Recognition genetics, Structure-Activity Relationship, Evolution, Molecular, Galectins immunology, Immunity, Innate immunology, Receptors, Pattern Recognition immunology
- Abstract
Galectins constitute an evolutionary conserved family of ß-galactoside-binding proteins, ubiquitous in mammals and other vertebrate taxa, invertebrates, and fungi. Since their discovery in the 1970s, their biological roles, initially understood as limited to recognition of carbohydrate ligands in embryogenesis and development, have expanded in recent years by the discovery of their immunoregulatory activities. A gradual paradigm shift has taken place in the past few years through the recognition that galectins also bind glycans on the surface of potentially pathogenic microbes, and function as recognition and effector factors in innate immunity. Further, an additional level of functional complexity has emerged with the most recent findings that some parasites "subvert" the recognition roles of the vector/host galectins for successful attachment or invasion.
- Published
- 2012
- Full Text
- View/download PDF
50. Structural and functional diversity of the lectin repertoire in teleost fish: relevance to innate and adaptive immunity.
- Author
-
Vasta GR, Nita-Lazar M, Giomarelli B, Ahmed H, Du S, Cammarata M, Parrinello N, Bianchet MA, and Amzel LM
- Subjects
- Animals, Antifreeze Proteins genetics, Antifreeze Proteins immunology, Antigens, Bacterial immunology, Antigens, Bacterial metabolism, Antigens, Viral immunology, Antigens, Viral metabolism, Bacteria immunology, Complement System Proteins immunology, Fish Proteins chemistry, Fish Proteins classification, Fish Proteins genetics, Fish Proteins metabolism, Fishes immunology, Gene Expression Regulation immunology, Invertebrates immunology, Lectins chemistry, Lectins classification, Lectins genetics, Lectins metabolism, Models, Molecular, Opsonin Proteins genetics, Opsonin Proteins immunology, Organ Specificity, Phagocytosis immunology, Protein Structure, Quaternary, Protein Structure, Tertiary, Vertebrates immunology, Viruses immunology, Adaptive Immunity, Complement Activation immunology, Fish Proteins immunology, Immunity, Innate, Lectins immunology
- Abstract
Protein-carbohydrate interactions mediated by lectins have been recognized as key components of innate immunity in vertebrates and invertebrates, not only for recognition of potential pathogens, but also for participating in downstream effector functions, such as their agglutination, immobilization, and complement-mediated opsonization and killing. More recently, lectins have been identified as critical regulators of mammalian adaptive immune responses. Fish are endowed with virtually all components of the mammalian adaptive immunity, and are equipped with a complex lectin repertoire. In this review, we discuss evidence suggesting that: (a) lectin repertoires in teleost fish are highly diversified, and include not only representatives of the lectin families described in mammals, but also members of lectin families described for the first time in fish species; (b) the tissue-specific expression and localization of the diverse lectin repertoires and their molecular partners is consistent with their distinct biological roles in innate and adaptive immunity; (c) although some lectins may bind endogenous ligands, others bind sugars on the surface of potential pathogens; (d) in addition to pathogen recognition and opsonization, some lectins display additional effector roles, such as complement activation and regulation of immune functions; (e) some lectins that recognize exogenous ligands mediate processes unrelated to immunity: they may act as anti-freeze proteins or prevent polyspermia during fertilization., (Copyright © 2011. Published by Elsevier Ltd.)
- Published
- 2011
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.