1
|
Benjamin SV, Jégouzo SAF, Lieng C, Daniels C, Coispeau M, Lau RJ, Kim S, Metaxa Y, Philpott J, Li T, Dai C, Wang X, Newby ML, Pier GB, Crispin M, Clements A, Taylor ME, Drickamer K. A human lectin array for characterizing host-pathogen interactions. J Biol Chem 2024; 300:107869. [PMID: 39384043 DOI: 10.1016/j.jbc.2024.107869] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Revised: 09/21/2024] [Accepted: 10/01/2024] [Indexed: 10/11/2024] Open
Abstract
A human lectin array has been developed to probe the interactions of innate immune receptors with pathogenic and commensal microorganisms. Following the successful introduction of a lectin array containing all of the cow C-type carbohydrate-recognition domains (CRDs), a human array described here contains the C-type CRDs as well as CRDs from other classes of sugar-binding receptors, including galectins, siglecs, R-type CRDs, ficolins, intelectins, and chitinase-like lectins. The array is constructed with CRDs modified with single-site biotin tags, ensuring that the sugar-binding sites in CRDs are displayed on a streptavidin-coated surface in a defined orientation and are accessible to the surfaces of microbes. A common approach used for expression and display of CRDs from all of the different structural categories of glycan-binding receptors allows comparisons across lectin families. In addition to previously documented protocols for binding of fluorescently labeled bacteria, methods have been developed for detecting unlabeled bacteria bound to the array by counter-staining with DNA-binding dye. Screening has also been undertaken with viral glycoproteins and bacterial and fungal polysaccharides. The array provides an unbiased screen for sugar ligands that interact with receptors and many show binding not anticipated from earlier studies. For example, some of the galectins bind with high affinity to bacterial glycans that lack lactose or N-acetyllactosamine. The results demonstrate the utility of the human lectin array for providing a unique overview of the interactions of multiple classes of glycan-binding proteins in the innate immune system with different types of microorganisms.
Collapse
Affiliation(s)
- Stefi V Benjamin
- Department of Life Sciences, Imperial College London, London, United Kingdom
| | - Sabine A F Jégouzo
- Department of Life Sciences, Imperial College London, London, United Kingdom
| | - Chloe Lieng
- Department of Life Sciences, Imperial College London, London, United Kingdom
| | - Connor Daniels
- Department of Life Sciences, Imperial College London, London, United Kingdom
| | - Marine Coispeau
- Department of Life Sciences, Imperial College London, London, United Kingdom
| | - Rikin J Lau
- Department of Life Sciences, Imperial College London, London, United Kingdom
| | - Suyeon Kim
- Department of Life Sciences, Imperial College London, London, United Kingdom
| | - Yasmine Metaxa
- Department of Life Sciences, Imperial College London, London, United Kingdom
| | - James Philpott
- Department of Life Sciences, Imperial College London, London, United Kingdom
| | - Tiannuo Li
- Department of Life Sciences, Imperial College London, London, United Kingdom
| | - Chao Dai
- Department of Life Sciences, Imperial College London, London, United Kingdom
| | - Xin Wang
- Department of Life Sciences, Imperial College London, London, United Kingdom
| | - Maddy L Newby
- School of Biological Sciences, University of Southampton, United Kingdom
| | - Gerald B Pier
- Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Max Crispin
- School of Biological Sciences, University of Southampton, United Kingdom
| | - Abigail Clements
- Department of Life Sciences, Imperial College London, London, United Kingdom
| | - Maureen E Taylor
- Department of Life Sciences, Imperial College London, London, United Kingdom
| | - Kurt Drickamer
- Department of Life Sciences, Imperial College London, London, United Kingdom.
| |
Collapse
|
2
|
Zhang Z, Li X, Huang M, Huang Y, Tan X, Dong Y, Huang Y, Jian J. Siglec7 functions as an inhibitory receptor of non-specific cytotoxic cells and can regulate the innate immune responses in a primitive vertebrate (Oreochromis niloticus). Int J Biol Macromol 2024; 278:134851. [PMID: 39168212 DOI: 10.1016/j.ijbiomac.2024.134851] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2024] [Revised: 07/29/2024] [Accepted: 08/16/2024] [Indexed: 08/23/2024]
Abstract
In mammals, siglec7, an integral component of the siglecs, is principally found on the surface of natural killer (NK) cells, macrophages, and monocytes, where it interacts with various pathogens to perform immunological regulatory activities. Nonetheless, the immune defense and mechanism of siglec7 in early vertebrates remain unknown. In this study, we identified siglec7 from Oreochromis niloticus (OnSiglec7) and revealed its immune functions. Specifically, OnSiglec7 was abundantly expressed in immune-related tissues of healthy tilapia and its transcription level was strongly activated after being challenged with A. hydrophila, S. agalactiae, and Poly: IC. Meanwhile, OnSiglec7 protein was purified and analyzed, which could recognize multiple pathogens through binding and agglutinating activity. Moreover, OnSiglec7-positive cells were mainly distributed in non-specific cytotoxic cells (NCC) of tilapia HKLs and showed cell membrane localization. Furthermore, OnSiglec7 blockage affected multiple innate immune responses (inflammation, apoptosis, and pyroptosis process) by regulating the activation of MAPK, NF-κB, TLR, and JAK-STAT pathways. Finally, OnSiglec7 blockage also greatly enhanced the cytotoxic effect of tilapia NCC. Summarily, this study uncovers immune functions and mechanisms of siglec7 in primitive vertebrates, thereby enhancing our understanding of the systemic evolution and ancient functions of other siglecs within the host's innate immune system (to our knowledge).
Collapse
Affiliation(s)
- Zhiqiang Zhang
- College of Fishery, Guangdong Ocean University, Guangdong Provincial Key Laboratory of Aquatic Animal Disease Control and Healthy Culture, Zhanjiang, China; Laboratory for Marine Biology and Biotechnology, Qingdao National Laboratory for Marine Science and Technology, Qingdao, China
| | - Xing Li
- College of Fishery, Guangdong Ocean University, Guangdong Provincial Key Laboratory of Aquatic Animal Disease Control and Healthy Culture, Zhanjiang, China
| | - Meiling Huang
- College of Fishery, Guangdong Ocean University, Guangdong Provincial Key Laboratory of Aquatic Animal Disease Control and Healthy Culture, Zhanjiang, China
| | - Yongxiong Huang
- College of Fishery, Guangdong Ocean University, Guangdong Provincial Key Laboratory of Aquatic Animal Disease Control and Healthy Culture, Zhanjiang, China
| | - Xuyan Tan
- College of Fishery, Guangdong Ocean University, Guangdong Provincial Key Laboratory of Aquatic Animal Disease Control and Healthy Culture, Zhanjiang, China
| | - Yuhang Dong
- College of Fishery, Guangdong Ocean University, Guangdong Provincial Key Laboratory of Aquatic Animal Disease Control and Healthy Culture, Zhanjiang, China
| | - Yu Huang
- College of Fishery, Guangdong Ocean University, Guangdong Provincial Key Laboratory of Aquatic Animal Disease Control and Healthy Culture, Zhanjiang, China; Laboratory for Marine Biology and Biotechnology, Qingdao National Laboratory for Marine Science and Technology, Qingdao, China; Guangdong Provincial Engineering Research Center for Aquatic Animal Health Assessment, Shenzhen, China.
| | - Jichang Jian
- College of Fishery, Guangdong Ocean University, Guangdong Provincial Key Laboratory of Aquatic Animal Disease Control and Healthy Culture, Zhanjiang, China; Laboratory for Marine Biology and Biotechnology, Qingdao National Laboratory for Marine Science and Technology, Qingdao, China; Guangdong Provincial Engineering Research Center for Aquatic Animal Health Assessment, Shenzhen, China.
| |
Collapse
|
3
|
Kim B, Jang S, Jang H, Kim JS, Jeon TI, Park JG, Shin IS, Cho KO, Moon C. Lectin histochemistry in the small intestines of piglets naturally infected with porcine epidemic diarrhea virus. J Vet Sci 2024; 25:e66. [PMID: 39363654 PMCID: PMC11450395 DOI: 10.4142/jvs.24179] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Revised: 07/21/2024] [Accepted: 08/04/2024] [Indexed: 10/05/2024] Open
Abstract
IMPORTANCE Porcine epidemic diarrhea virus (PEDV) binds to particular cell surface receptors to penetrate cells. The virus specifically identifies certain carbohydrate structures present on the surface of the cell to facilitate the binding process. Nevertheless, the influence of viral infections on specific alterations of glycoconjugates in the small intestines remains unexplored. OBJECTIVE This work aimed to examine the alterations in glycoconjugates in the small intestines of piglets naturally infected with PEDV using lectin histochemistry. METHODS Six piglets including three PEDV-infected and three non-infected piglets were evaluated. Small intestinal samples were histopathologically examined, and lectin histochemistry was performed. RESULTS Piglets infected with PEDV had significant histological abnormalities in their small intestines, such as pronounced villous atrophy, varying degrees of villous fusion, and diverse mucosal alterations. Specific regions of the duodenum, jejunum, and ileum showed discernible variations in glycoconjugate distribution, as determined by lectin histochemistry. Compared with the controls, the PEDV-infected piglets showed significant changes in N-acetylglucosamine- and galactose-binding lectins (particularly wheat germ agglutinin and Arachis hypogaea (peanut) agglutinin) in multiple intestinal regions. CONCLUSIONS AND RELEVANCE These findings can enhance understanding of how viruses such as PEDV impact the glycoconjugate composition of the small intestines and emphasize the potential connection between the pathogenesis of PEDV and glycoconjugate.
Collapse
Affiliation(s)
- Bohye Kim
- College of Veterinary Medicine and Host-Directed Antiviral Research Center, Chonnam National University, Gwangju 61186, Korea
| | - Sungwoong Jang
- College of Veterinary Medicine and Host-Directed Antiviral Research Center, Chonnam National University, Gwangju 61186, Korea
| | - Hyewon Jang
- College of Veterinary Medicine and Host-Directed Antiviral Research Center, Chonnam National University, Gwangju 61186, Korea
| | - Joong-Sun Kim
- College of Veterinary Medicine and Host-Directed Antiviral Research Center, Chonnam National University, Gwangju 61186, Korea
| | - Tae-Il Jeon
- College of Veterinary Medicine and Host-Directed Antiviral Research Center, Chonnam National University, Gwangju 61186, Korea
| | - Jun-Gyu Park
- College of Veterinary Medicine and Host-Directed Antiviral Research Center, Chonnam National University, Gwangju 61186, Korea
| | - In-Sik Shin
- College of Veterinary Medicine and Host-Directed Antiviral Research Center, Chonnam National University, Gwangju 61186, Korea
| | - Kyoung-Oh Cho
- College of Veterinary Medicine and Host-Directed Antiviral Research Center, Chonnam National University, Gwangju 61186, Korea
| | - Changjong Moon
- College of Veterinary Medicine and Host-Directed Antiviral Research Center, Chonnam National University, Gwangju 61186, Korea.
| |
Collapse
|
4
|
Alvarado-Melendez EI, de Jong H, Hartman JEM, Ong JY, Wösten MMSM, Wennekes T. Glycoengineering with neuraminic acid analogs to label lipooligosaccharides and detect native sialyltransferase activity in gram-negative bacteria. Glycobiology 2024; 34:cwae071. [PMID: 39244665 DOI: 10.1093/glycob/cwae071] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2024] [Revised: 08/27/2024] [Accepted: 09/06/2024] [Indexed: 09/10/2024] Open
Abstract
Lipooligosaccharides are the most abundant cell surface glycoconjugates on the outer membrane of Gram-negative bacteria. They play important roles in host-microbe interactions. Certain Gram-negative pathogenic bacteria cap their lipooligosaccharides with the sialic acid, N-acetylneuraminic acid (Neu5Ac), to mimic host glycans that among others protects these bacteria from recognition by the hosts immune system. This process of molecular mimicry is not fully understood and remains under investigated. To explore the functional role of sialic acid-capped lipooligosaccharides at the molecular level, it is important to have tools readily available for the detection and manipulation of both Neu5Ac on glycoconjugates and the involved sialyltransferases, preferably in live bacteria. We and others have shown that the native sialyltransferases of some Gram-negative bacteria can incorporate extracellular unnatural sialic acid nucleotides onto their lipooligosaccharides. We here report on the expanded use of native bacterial sialyltransferases to incorporate neuraminic acids analogs with a reporter group into the lipooligosaccharides of a variety of Gram-negative bacteria. We show that this approach offers a quick strategy to screen bacteria for the expression of functional sialyltransferases and the ability to use exogenous CMP-Neu5Ac to decorate their glycoconjugates. For selected bacteria we also show this strategy complements two other glycoengineering techniques, Metabolic Oligosaccharide Engineering and Selective Exo-Enzymatic Labeling, and that together they provide tools to modify, label, detect and visualize sialylation of bacterial lipooligosaccharides.
Collapse
Affiliation(s)
- Erianna I Alvarado-Melendez
- Department of Chemical Biology and Drug Discovery, Utrecht Institute for Pharmaceutical Sciences and Bijvoet Center for Biomedical Research, Utrecht University, Universiteitsweg 99, 3584CG, Utrecht, The Netherlands
| | - Hanna de Jong
- Department of Chemical Biology and Drug Discovery, Utrecht Institute for Pharmaceutical Sciences and Bijvoet Center for Biomedical Research, Utrecht University, Universiteitsweg 99, 3584CG, Utrecht, The Netherlands
| | - Jet E M Hartman
- Department of Chemical Biology and Drug Discovery, Utrecht Institute for Pharmaceutical Sciences and Bijvoet Center for Biomedical Research, Utrecht University, Universiteitsweg 99, 3584CG, Utrecht, The Netherlands
| | - Jun Yang Ong
- Department of Chemical Biology and Drug Discovery, Utrecht Institute for Pharmaceutical Sciences and Bijvoet Center for Biomedical Research, Utrecht University, Universiteitsweg 99, 3584CG, Utrecht, The Netherlands
| | - Marc M S M Wösten
- Department of Biomolecular Health Sciences, Division Infectious Diseases and Immunology, Faculty of Veterinary Medicine, Utrecht University, Yalelaan 1, 3584CL, Utrecht, The Netherlands
| | - Tom Wennekes
- Department of Chemical Biology and Drug Discovery, Utrecht Institute for Pharmaceutical Sciences and Bijvoet Center for Biomedical Research, Utrecht University, Universiteitsweg 99, 3584CG, Utrecht, The Netherlands
| |
Collapse
|
5
|
Campanero-Rhodes MA, Martí S, Hernández-Ortiz N, Cubero M, Ereño-Orbea J, Ardá A, Jiménez-Barbero J, Ardanuy C, Solís D. Insights into the recognition of hypermucoviscous Klebsiella pneumoniae clinical isolates by innate immune lectins of the Siglec and galectin families. Front Immunol 2024; 15:1436039. [PMID: 39148735 PMCID: PMC11324429 DOI: 10.3389/fimmu.2024.1436039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2024] [Accepted: 07/16/2024] [Indexed: 08/17/2024] Open
Abstract
Klebsiella pneumoniae is an opportunistic bacterium that frequently colonizes the nasopharynx and gastrointestinal tract and can also cause severe infections when invading other tissues, particularly in immunocompromised individuals. Moreover, K. pneumoniae variants exhibiting a hypermucoviscous (HMV) phenotype are usually associated with hypervirulent strains that can produce invasive infections even in immunocompetent individuals. Major carbohydrate structures displayed on the K. pneumoniae surface are the polysaccharide capsule and the lipopolysaccharide, which presents an O-polysaccharide chain in its outermost part. Various capsular and O-chain structures have been described. Of note, production of a thick capsule is frequently observed in HMV variants. Here we examined the surface sugar epitopes of a collection of HMV and non-HMV K. pneumoniae clinical isolates and their recognition by several Siglecs and galectins, two lectin families of the innate immune system, using bacteria microarrays as main tool. No significant differences among isolates in sialic acid content or recognition by Siglecs were observed. In contrast, analysis of the binding of model lectins with diverse carbohydrate-binding specificities revealed striking differences in the recognition by galactose- and mannose-specific lectins, which correlated with the binding or lack of binding of galectins and pointed to the O-chain as the plausible ligand. Fluorescence microscopy and microarray analyses of galectin-9 binding to entire cells and outer membranes of two representative HMV isolates supported the bacteria microarray results. In addition, Western blot analysis of the binding of galectin-9 to outer membranes unveiled protein bands recognized by this galectin, and fingerprint analysis of these bands identified several proteins containing potential O-glycosylation sites, thus broadening the spectrum of possible galectin ligands on the K. pneumoniae surface. Moreover, Siglecs and galectins apparently target different structures on K. pneumoniae surfaces, thereby behaving as non-redundant complementary tools of the innate immune system.
Collapse
Affiliation(s)
- María Asunción Campanero-Rhodes
- Department of Biological Physical Chemistry, Instituto de Química Física Blas Cabrera, Consejo Superior de Investigaciones Científicas, Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Respiratorias (CIBERES), Instituto de Salud Carlos III, Madrid, Spain
| | - Sara Martí
- Centro de Investigación Biomédica en Red de Enfermedades Respiratorias (CIBERES), Instituto de Salud Carlos III, Madrid, Spain
- Microbiology Department, Hospital Universitari Bellvitge, University of Barcelona-Fundación Instituto de Investigación Biomédica de Bellvitge, L’Hospitalet de Llobregat, Spain
| | - Noelia Hernández-Ortiz
- Department of Biological Physical Chemistry, Instituto de Química Física Blas Cabrera, Consejo Superior de Investigaciones Científicas, Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Respiratorias (CIBERES), Instituto de Salud Carlos III, Madrid, Spain
| | - Meritxell Cubero
- Centro de Investigación Biomédica en Red de Enfermedades Respiratorias (CIBERES), Instituto de Salud Carlos III, Madrid, Spain
- Microbiology Department, Hospital Universitari Bellvitge, University of Barcelona-Fundación Instituto de Investigación Biomédica de Bellvitge, L’Hospitalet de Llobregat, Spain
| | - June Ereño-Orbea
- CIC bioGUNE - Center for Cooperative Research in Biosciences, Basque Research & Technology Alliance (BRTA), Bizkaia Technology Park, Derio, Spain
- Ikerbasque, Basque Foundation for Science, Bilbao, Spain
| | - Ana Ardá
- CIC bioGUNE - Center for Cooperative Research in Biosciences, Basque Research & Technology Alliance (BRTA), Bizkaia Technology Park, Derio, Spain
- Ikerbasque, Basque Foundation for Science, Bilbao, Spain
| | - Jesús Jiménez-Barbero
- Centro de Investigación Biomédica en Red de Enfermedades Respiratorias (CIBERES), Instituto de Salud Carlos III, Madrid, Spain
- CIC bioGUNE - Center for Cooperative Research in Biosciences, Basque Research & Technology Alliance (BRTA), Bizkaia Technology Park, Derio, Spain
- Ikerbasque, Basque Foundation for Science, Bilbao, Spain
- Department of Organic Chemistry, II Faculty of Science and Technology University of the Basque Country, EHU/UPV, Leioa, Spain
| | - Carmen Ardanuy
- Centro de Investigación Biomédica en Red de Enfermedades Respiratorias (CIBERES), Instituto de Salud Carlos III, Madrid, Spain
- Microbiology Department, Hospital Universitari Bellvitge, University of Barcelona-Fundación Instituto de Investigación Biomédica de Bellvitge, L’Hospitalet de Llobregat, Spain
- Department of Pathology and Experimental Therapeutics, University of Barcelona, Barcelona, Spain
| | - Dolores Solís
- Department of Biological Physical Chemistry, Instituto de Química Física Blas Cabrera, Consejo Superior de Investigaciones Científicas, Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Respiratorias (CIBERES), Instituto de Salud Carlos III, Madrid, Spain
| |
Collapse
|
6
|
Lund SJ, Del Rosario PGB, Honda A, Caoili KJ, Hoeksema MA, Nizet V, Patras KA, Prince LS. Sialic Acid-Siglec-E Interactions Regulate the Response of Neonatal Macrophages to Group B Streptococcus. Immunohorizons 2024; 8:384-396. [PMID: 38809232 PMCID: PMC11150127 DOI: 10.4049/immunohorizons.2300076] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2023] [Accepted: 04/24/2024] [Indexed: 05/30/2024] Open
Abstract
The mammalian Siglec receptor sialoadhesin (Siglec1, CD169) confers innate immunity against the encapsulated pathogen group B Streptococcus (GBS). Newborn lung macrophages have lower expression levels of sialoadhesin at birth compared with the postnatal period, increasing their susceptibility to GBS infection. In this study, we investigate the mechanisms regulating sialoadhesin expression in the newborn mouse lung. In both neonatal and adult mice, GBS lung infection reduced Siglec1 expression, potentially delaying acquisition of immunity in neonates. Suppression of Siglec1 expression required interactions between sialic acid on the GBS capsule and the inhibitory host receptor Siglec-E. The Siglec1 gene contains multiple STAT binding motifs, which could regulate expression of sialoadhesin downstream of innate immune signals. Although GBS infection reduced STAT1 expression in the lungs of wild-type newborn mice, we observed increased numbers of STAT1+ cells in Siglece-/- lungs. To test if innate immune activation could increase sialoadhesin at birth, we first demonstrated that treatment of neonatal lung macrophages ex vivo with inflammatory activators increased sialoadhesin expression. However, overcoming the low sialoadhesin expression at birth using in vivo prenatal exposures or treatments with inflammatory stimuli were not successful. The suppression of sialoadhesin expression by GBS-Siglec-E engagement may therefore contribute to disease pathogenesis in newborns and represent a challenging but potentially appealing therapeutic opportunity to augment immunity at birth.
Collapse
MESH Headings
- Animals
- Mice
- Streptococcus agalactiae/immunology
- Animals, Newborn
- N-Acetylneuraminic Acid/metabolism
- Sialic Acid Binding Ig-like Lectin 1/metabolism
- Streptococcal Infections/immunology
- Streptococcal Infections/microbiology
- STAT1 Transcription Factor/metabolism
- STAT1 Transcription Factor/genetics
- Mice, Knockout
- Immunity, Innate
- Mice, Inbred C57BL
- Lung/immunology
- Lung/microbiology
- Lung/metabolism
- Macrophages, Alveolar/immunology
- Macrophages, Alveolar/metabolism
- Female
- Macrophages/immunology
- Macrophages/metabolism
- Lectins/metabolism
- Lectins/genetics
- Sialic Acid Binding Immunoglobulin-like Lectins/metabolism
- Sialic Acid Binding Immunoglobulin-like Lectins/genetics
- Antigens, CD/metabolism
- Antigens, CD/genetics
- Antigens, Differentiation, B-Lymphocyte
Collapse
Affiliation(s)
- Sean J. Lund
- Department of Pediatrics, University of California, San Diego, La Jolla, CA
| | - Pamela G. B. Del Rosario
- Department of Pediatrics, University of California, San Diego, La Jolla, CA
- Rady Children’s Hospital, San Diego, CA
| | - Asami Honda
- Department of Pediatrics, University of California, San Diego, La Jolla, CA
| | | | - Marten A. Hoeksema
- Department of Medical Biochemistry, Amsterdam University Medical Center, Amsterdam Zuidoost, the Netherlands
| | - Victor Nizet
- Department of Pediatrics, University of California, San Diego, La Jolla, CA
| | - Kathryn A. Patras
- Department of Molecular Virology and Microbiology, Alkek Center for Metagenomics and Microbiome Research, Baylor College of Medicine, Houston, TX
| | | |
Collapse
|
7
|
Lee IM, Wu HY, Angata T, Wu SH. Bacterial pseudaminic acid binding to Siglec-10 induces a macrophage interleukin-10 response and suppresses phagocytosis. Chem Commun (Camb) 2024; 60:2930-2933. [PMID: 38372418 DOI: 10.1039/d4cc00077c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/20/2024]
Abstract
Pseudaminic acid (Pse) on pathogenic bacteria exopolysaccharide engages with the sialic acid-binding immunoglobulin-type lectin (Siglec)-10 receptor on macrophages via the critical 7-N-acetyl group. This binding stimulates macrophages to secrete interleukin 10 that suppresses phagocytosis against bacteria, but can be reverted by blocking Pse-Siglec-10 interaction with Pse-binding protein as a promising therapy.
Collapse
Affiliation(s)
- I-Ming Lee
- Department of Marine Biotechnology and Resources, National Sun Yat-sen University, No. 70 Lien-hai Road, Kaohsiung, 804201, Taiwan
| | - Hsing-Yu Wu
- Institute of Biological Chemistry, Academia Sinica, 128 Academia Road, Section 2, Nankang, 11529, Taipei, Taiwan.
| | - Takashi Angata
- Institute of Biological Chemistry, Academia Sinica, 128 Academia Road, Section 2, Nankang, 11529, Taipei, Taiwan.
| | - Shih-Hsiung Wu
- Institute of Biological Chemistry, Academia Sinica, 128 Academia Road, Section 2, Nankang, 11529, Taipei, Taiwan.
| |
Collapse
|
8
|
Dedola S, Ahmadipour S, de Andrade P, Baker AN, Boshra AN, Chessa S, Gibson MI, Hernando PJ, Ivanova IM, Lloyd JE, Marín MJ, Munro-Clark AJ, Pergolizzi G, Richards SJ, Ttofi I, Wagstaff BA, Field RA. Sialic acids in infection and their potential use in detection and protection against pathogens. RSC Chem Biol 2024; 5:167-188. [PMID: 38456038 PMCID: PMC10915975 DOI: 10.1039/d3cb00155e] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2023] [Accepted: 12/12/2023] [Indexed: 03/09/2024] Open
Abstract
In structural terms, the sialic acids are a large family of nine carbon sugars based around an alpha-keto acid core. They are widely spread in nature, where they are often found to be involved in molecular recognition processes, including in development, immunology, health and disease. The prominence of sialic acids in infection is a result of their exposure at the non-reducing terminus of glycans in diverse glycolipids and glycoproteins. Herein, we survey representative aspects of sialic acid structure, recognition and exploitation in relation to infectious diseases, their diagnosis and prevention or treatment. Examples covered span influenza virus and Covid-19, Leishmania and Trypanosoma, algal viruses, Campylobacter, Streptococci and Helicobacter, and commensal Ruminococci.
Collapse
Affiliation(s)
- Simone Dedola
- Department of Chemistry and Manchester Institute of Biotechnology, University of Manchester 131 Princess Street Manchester M1 7DN UK
- Iceni Glycoscience Ltd, Norwich Research Park Norwich NR4 7TJ UK
| | - Sanaz Ahmadipour
- Department of Chemistry and Manchester Institute of Biotechnology, University of Manchester 131 Princess Street Manchester M1 7DN UK
| | - Peterson de Andrade
- Department of Chemistry and Manchester Institute of Biotechnology, University of Manchester 131 Princess Street Manchester M1 7DN UK
| | - Alexander N Baker
- Department of Chemistry, University of Warwick Gibbet Hill Road Coventry CV4 7AL UK
| | - Andrew N Boshra
- Department of Chemistry and Manchester Institute of Biotechnology, University of Manchester 131 Princess Street Manchester M1 7DN UK
- Department of Pharmaceutical Organic Chemistry, Faculty of Pharmacy, Assiut University Assiut 71526 Egypt
| | - Simona Chessa
- Iceni Glycoscience Ltd, Norwich Research Park Norwich NR4 7TJ UK
| | - Matthew I Gibson
- Department of Chemistry and Manchester Institute of Biotechnology, University of Manchester 131 Princess Street Manchester M1 7DN UK
- Department of Chemistry, University of Warwick Gibbet Hill Road Coventry CV4 7AL UK
- Division of Biomedical Sciences, Warwick Medical School Coventry CV4 7AL UK
| | - Pedro J Hernando
- Iceni Glycoscience Ltd, Norwich Research Park Norwich NR4 7TJ UK
| | - Irina M Ivanova
- Iceni Glycoscience Ltd, Norwich Research Park Norwich NR4 7TJ UK
| | - Jessica E Lloyd
- Department of Chemistry and Manchester Institute of Biotechnology, University of Manchester 131 Princess Street Manchester M1 7DN UK
| | - María J Marín
- School of Chemistry, University of East Anglia, Norwich Research Park Norwich NR4 7TJ UK
| | - Alexandra J Munro-Clark
- Department of Chemistry and Manchester Institute of Biotechnology, University of Manchester 131 Princess Street Manchester M1 7DN UK
| | | | - Sarah-Jane Richards
- Department of Chemistry and Manchester Institute of Biotechnology, University of Manchester 131 Princess Street Manchester M1 7DN UK
- Department of Chemistry, University of Warwick Gibbet Hill Road Coventry CV4 7AL UK
| | - Iakovia Ttofi
- Department of Chemistry and Manchester Institute of Biotechnology, University of Manchester 131 Princess Street Manchester M1 7DN UK
- Iceni Glycoscience Ltd, Norwich Research Park Norwich NR4 7TJ UK
| | - Ben A Wagstaff
- Department of Chemistry and Manchester Institute of Biotechnology, University of Manchester 131 Princess Street Manchester M1 7DN UK
| | - Robert A Field
- Department of Chemistry and Manchester Institute of Biotechnology, University of Manchester 131 Princess Street Manchester M1 7DN UK
- Iceni Glycoscience Ltd, Norwich Research Park Norwich NR4 7TJ UK
| |
Collapse
|
9
|
Hamrangsekachaee M, Wen K, Yazdani N, Willits RK, Bencherif SA, Ebong EE. Endothelial glycocalyx sensitivity to chemical and mechanical sub-endothelial substrate properties. Front Bioeng Biotechnol 2023; 11:1250348. [PMID: 38026846 PMCID: PMC10643223 DOI: 10.3389/fbioe.2023.1250348] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Accepted: 10/13/2023] [Indexed: 12/01/2023] Open
Abstract
Glycocalyx (GCX) is a carbohydrate-rich structure that coats the surface of endothelial cells (ECs) and lines the blood vessel lumen. Mechanical perturbations in the vascular environment, such as blood vessel stiffness, can be transduced and sent to ECs through mechanosensors such as GCX. Adverse stiffness alters GCX-mediated mechanotransduction and leads to EC dysfunction and eventually atherosclerotic cardiovascular diseases. To understand GCX-regulated mechanotransduction events, an in vitro model emulating in vivo vessel conditions is needed. To this end, we investigated the impact of matrix chemical and mechanical properties on GCX expression via fabricating a tunable non-swelling matrix based on the collagen-derived polypeptide, gelatin. To study the effect of matrix composition, we conducted a comparative analysis of GCX expression using different concentrations (60-25,000 μg/mL) of gelatin and gelatin methacrylate (GelMA) in comparison to fibronectin (60 μg/mL), a standard coating material for GCX-related studies. Using immunocytochemistry analysis, we showed for the first time that different substrate compositions and concentrations altered the overall GCX expression on human umbilical vein ECs (HUVECs). Subsequently, GelMA hydrogels were fabricated with stiffnesses of 2.5 and 5 kPa, representing healthy vessel tissues, and 10 kPa, corresponding to diseased vessel tissues. Immunocytochemistry analysis showed that on hydrogels with different levels of stiffness, the GCX expression in HUVECs remained unchanged, while its major polysaccharide components exhibited dysregulation in distinct patterns. For example, there was a significant decrease in heparan sulfate expression on pathological substrates (10 kPa), while sialic acid expression increased with increased matrix stiffness. This study suggests the specific mechanisms through which GCX may influence ECs in modulating barrier function, immune cell adhesion, and mechanotransduction function under distinct chemical and mechanical conditions of both healthy and diseased substrates.
Collapse
Affiliation(s)
| | - Ke Wen
- Chemical Engineering Department, Northeastern University, Boston, MA, United States
| | - Narges Yazdani
- Bioengineering Department, Northeastern University, Boston, MA, United States
| | - Rebecca K. Willits
- Chemical Engineering Department, Northeastern University, Boston, MA, United States
- Bioengineering Department, Northeastern University, Boston, MA, United States
| | - Sidi A. Bencherif
- Chemical Engineering Department, Northeastern University, Boston, MA, United States
- Bioengineering Department, Northeastern University, Boston, MA, United States
- Laboratoire de BioMécanique et BioIngénierie (BMBI), UMR CNRS, Sorbonne Universités, Université de Technologie of Compiègne (UTC), Compiègne, France
- Harvard John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA, United States
| | - Eno E. Ebong
- Chemical Engineering Department, Northeastern University, Boston, MA, United States
- Bioengineering Department, Northeastern University, Boston, MA, United States
- Neuroscience Department, Albert Einstein College of Medicine, New York, NY, United States
| |
Collapse
|
10
|
Pinho SS, Alves I, Gaifem J, Rabinovich GA. Immune regulatory networks coordinated by glycans and glycan-binding proteins in autoimmunity and infection. Cell Mol Immunol 2023; 20:1101-1113. [PMID: 37582971 PMCID: PMC10541879 DOI: 10.1038/s41423-023-01074-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Accepted: 07/26/2023] [Indexed: 08/17/2023] Open
Abstract
The immune system is coordinated by an intricate network of stimulatory and inhibitory circuits that regulate host responses against endogenous and exogenous insults. Disruption of these safeguard and homeostatic mechanisms can lead to unpredictable inflammatory and autoimmune responses, whereas deficiency of immune stimulatory pathways may orchestrate immunosuppressive programs that contribute to perpetuate chronic infections, but also influence cancer development and progression. Glycans have emerged as essential components of homeostatic circuits, acting as fine-tuners of immunological responses and potential molecular targets for manipulation of immune tolerance and activation in a wide range of pathologic settings. Cell surface glycans, present in cells, tissues and the extracellular matrix, have been proposed to serve as "self-associated molecular patterns" that store structurally relevant biological data. The responsibility of deciphering this information relies on different families of glycan-binding proteins (including galectins, siglecs and C-type lectins) which, upon recognition of specific carbohydrate structures, can recalibrate the magnitude, nature and fate of immune responses. This process is tightly regulated by the diversity of glycan structures and the establishment of multivalent interactions on cell surface receptors and the extracellular matrix. Here we review the spatiotemporal regulation of selected glycan-modifying processes including mannosylation, complex N-glycan branching, core 2 O-glycan elongation, LacNAc extension, as well as terminal sialylation and fucosylation. Moreover, we illustrate examples that highlight the contribution of these processes to the control of immune responses and their integration with canonical tolerogenic pathways. Finally, we discuss the power of glycans and glycan-binding proteins as a source of immunomodulatory signals that could be leveraged for the treatment of autoimmune inflammation and chronic infection.
Collapse
Affiliation(s)
- Salomé S Pinho
- i3S - Institute for Research and Innovation in Health, University of Porto, 4200-135, Porto, Portugal.
- ICBAS-School of Medicine and Biomedical Sciences, University of Porto, 4050-313, Porto, Portugal.
- Faculty of Medicine, University of Porto, 4200-319, Porto, Portugal.
| | - Inês Alves
- i3S - Institute for Research and Innovation in Health, University of Porto, 4200-135, Porto, Portugal
| | - Joana Gaifem
- i3S - Institute for Research and Innovation in Health, University of Porto, 4200-135, Porto, Portugal
| | - Gabriel A Rabinovich
- Laboratorio de Glicomedicina, Instituto de Biología y Medicina Experimental (IBYME), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), C1428, Ciudad de Buenos Aires, Argentina.
- Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, C1428, Ciudad de Buenos Aires, Argentina.
| |
Collapse
|
11
|
Chen Y, Chen H, Zheng Q. Siglecs family used by pathogens for immune escape may engaged in immune tolerance in pregnancy. J Reprod Immunol 2023; 159:104127. [PMID: 37572430 DOI: 10.1016/j.jri.2023.104127] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2023] [Revised: 07/18/2023] [Accepted: 08/02/2023] [Indexed: 08/14/2023]
Abstract
The Siglecs family is a group of type I sialic acid-binding immunoglobulin-like receptors that regulate cellular signaling by recognizing sialic acid epitopes. Siglecs are predominantly expressed on the surface of leukocytes, where they play a crucial role in regulating immune activity. Pathogens can exploit inhibitory Siglecs by utilizing their sialic acid components to promote invasion or suppress immune functions, facilitating immune evasion. The establishing of an immune-balanced maternal-fetal interface microenvironment is essential for a successful pregnancy. Dysfunctional immune cells may lead to adverse pregnancy outcomes. Siglecs are important for inducing a phenotypic switch in leukocytes at the maternal-fetal interface toward a less toxic and more tolerant phenotype. Recent discoveries regarding Siglecs in the reproductive system have drawn further attention to their potential roles in reproduction. In this review, we primarily discuss the latest advances in understanding the impact of Siglecs as immune regulators on infections and pregnancy.
Collapse
Affiliation(s)
- Ying Chen
- Prenatal Diagnosis Center, The Eighth Affiliated Hospital, Sun Yat-sen University, 3025# Shennan Road, Shenzhen 518033, PR China
| | - Huan Chen
- Prenatal Diagnosis Center, The Eighth Affiliated Hospital, Sun Yat-sen University, 3025# Shennan Road, Shenzhen 518033, PR China
| | - Qingliang Zheng
- Prenatal Diagnosis Center, The Eighth Affiliated Hospital, Sun Yat-sen University, 3025# Shennan Road, Shenzhen 518033, PR China.
| |
Collapse
|
12
|
Tsubata T. Siglec cis-ligands and their roles in the immune system. Glycobiology 2023; 33:532-544. [PMID: 37154567 DOI: 10.1093/glycob/cwad038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2022] [Revised: 04/14/2023] [Indexed: 05/10/2023] Open
Abstract
Sialic acid-binding immunoglobulin-like lectins are a family of membrane molecules primarily expressed in immune cells. Most of them are inhibitory receptors containing immunoreceptor tyrosine-based inhibition motifs in the cytoplasmic tail. On the cell surface, sialic acid-binding immunoglobulin-like lectins are mostly bound by sialylated glycans on membrane molecules expressed in the same cell (cis-ligands). Although ligands of sialic acid-binding immunoglobulin-like lectins are not efficiently identified by conventional methods such as immunoprecipitation, in situ labeling including proximity labeling is useful in identifying both cis-ligands and the sialylated ligands expressed by other cells (trans-ligands) of sialic acid-binding immunoglobulin-like lectins. Interaction of the inhibitory sialic acid-binding immunoglobulin-like lectins with cis-ligands including both those with and without signaling function modulates the inhibitory activity of sialic acid-binding immunoglobulin-like lectins by multiple different ways. This interaction also modulates signaling function of the cis-ligands. So far, little is known about the role of the interaction between sialic acid-binding immunoglobulin-like lectins and the cis-ligands. Nonetheless, recent studies showed that the inhibitory activity of CD22 (also known as Siglec-2) is regulated by endogenous ligands, most likely cis-ligands, differentially in resting B cells and those in which B-cell antigen receptor is ligated. This differential regulation plays a role in quality control of signaling-competent B cells and also partial restoration of B-cell antigen receptor signaling in immunodeficient B cells.
Collapse
Affiliation(s)
- Takeshi Tsubata
- Department of Pathology, Nihon University School of Dentistry, Tokyo 101-8310, Japan
| |
Collapse
|
13
|
Pillsbury CE, Dougan J, Rabe JL, Fonseca JA, Zhou C, Evans AN, Abukharma H, Ichoku O, Gonzalez-Flamenco G, Park SI, Aljudi A, DeRyckere D, Castellino SM, Rafiq S, Langermann S, Liu LN, Henry CJ, Porter CC. Siglec-15 Promotes Evasion of Adaptive Immunity in B-cell Acute Lymphoblastic Leukemia. CANCER RESEARCH COMMUNICATIONS 2023; 3:1248-1259. [PMID: 37465593 PMCID: PMC10351425 DOI: 10.1158/2767-9764.crc-23-0056] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/27/2023] [Revised: 04/28/2023] [Accepted: 06/20/2023] [Indexed: 07/20/2023]
Abstract
Siglec-15 (Sig15) has been implicated as an immune checkpoint expressed in solid tumor-infiltrating macrophages and is being targeted in clinical trials with mAbs to normalize the tumor immune microenvironment and stimulate antitumor immunity. However, the role of Sig15 in hematologic malignancies remains undefined. Sig15 mRNA and protein expression levels in hematologic malignancies were determined from publicly available databases, cell lines, and primary patient samples. Human B-cell acute lymphoblastic leukemia (B-ALL) cell lines were used to identify signaling pathways involved in the regulation of Sig15 expression. Secreted/soluble Sig15 and cytokine levels were measured from the plasma of children with leukemia and healthy controls. Knockdown and knockout of Siglec15 in a murine model of B-ALL was used to evaluate the effect of leukemia-derived Sig15 on the immune response to leukemia. We observed pathologic overexpression of Sig15 in a variety of hematologic malignancies, including primary B-ALL samples. This overexpression was driven by NFκB activation, which also increased the surface localization of Sig15. Secreted/soluble Sig15 was found to circulate at elevated levels in the plasma of children with B-ALL and correlated with an immune-suppressive cytokine milieu. Genetic inhibition of Sig15 in murine B-ALL promoted clearance of the leukemia by the immune system and a marked reversal of the immune-privileged leukemia bone marrow niche, including expanded early effector CD8+ T cells and reduction of immunosuppressive cytokines. Thus, Sig15 is a novel, potent immunosuppressive molecule active in leukemia that may be targeted therapeutically to activate T lymphocytes against leukemia cells. Significance We demonstrate that Sig15 is overexpressed in hematologic malignancies driven by NFκB, is required for immune evasion in a mouse model of leukemia, and, for the first time, that it circulates at high levels in the plasma of children with leukemia.
Collapse
Affiliation(s)
- Claire E. Pillsbury
- Cancer Biology Program, Laney Graduate School, Emory University, Atlanta, Georgia
| | - Jodi Dougan
- Department of Pediatrics, Emory University School of Medicine, Atlanta, Georgia
| | - Jennifer L. Rabe
- Molecular Biology Program, University of Colorado Denver, Aurora, Colorado
| | - Jairo A. Fonseca
- Department of Pediatrics, Emory University School of Medicine, Atlanta, Georgia
| | - Chengjing Zhou
- Department of Pediatrics, Emory University School of Medicine, Atlanta, Georgia
| | - Alyssa N. Evans
- Winship Cancer Institute, Emory University, Atlanta, Georgia
| | | | | | | | - Sunita I. Park
- Clinical Laboratory, Children's Healthcare of Atlanta, Atlanta, Georgia
- Department of Pathology, Emory University School of Medicine, Atlanta, Georgia
| | - Ahmed Aljudi
- Clinical Laboratory, Children's Healthcare of Atlanta, Atlanta, Georgia
- Department of Pathology, Emory University School of Medicine, Atlanta, Georgia
| | - Deborah DeRyckere
- Department of Pediatrics, Emory University School of Medicine, Atlanta, Georgia
- Winship Cancer Institute, Emory University, Atlanta, Georgia
- Aflac Cancer and Blood Disorders Center, Children's Healthcare of Atlanta, Atlanta, Georgia
| | - Sharon M. Castellino
- Department of Pediatrics, Emory University School of Medicine, Atlanta, Georgia
- Winship Cancer Institute, Emory University, Atlanta, Georgia
- Aflac Cancer and Blood Disorders Center, Children's Healthcare of Atlanta, Atlanta, Georgia
| | - Sarwish Rafiq
- Winship Cancer Institute, Emory University, Atlanta, Georgia
| | | | | | - Curtis J. Henry
- Department of Pediatrics, Emory University School of Medicine, Atlanta, Georgia
- Winship Cancer Institute, Emory University, Atlanta, Georgia
- Aflac Cancer and Blood Disorders Center, Children's Healthcare of Atlanta, Atlanta, Georgia
| | - Christopher C. Porter
- Department of Pediatrics, Emory University School of Medicine, Atlanta, Georgia
- Winship Cancer Institute, Emory University, Atlanta, Georgia
- Aflac Cancer and Blood Disorders Center, Children's Healthcare of Atlanta, Atlanta, Georgia
| |
Collapse
|
14
|
Shinde P, Kiepas A, Zhang L, Sudhir S, Konstantopoulos K, Stamatos NM. Polysialylation controls immune function of myeloid cells in murine model of pneumococcal pneumonia. Cell Rep 2023; 42:112648. [PMID: 37339052 PMCID: PMC10592499 DOI: 10.1016/j.celrep.2023.112648] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Revised: 04/13/2023] [Accepted: 05/31/2023] [Indexed: 06/22/2023] Open
Abstract
Polysialic acid (polySia) is a post-translational modification of a select group of cell-surface proteins that guides cellular interactions. As the overall impact of changes in expression of this glycan on leukocytes during infection is not known, we evaluate the immune response of polySia-deficient ST8SiaIV-/- mice infected with Streptococcus pneumoniae (Spn). Compared with wild-type (WT) mice, ST8SiaIV-/- mice are less susceptible to infection and clear Spn from airways faster, with alveolar macrophages demonstrating greater viability and phagocytic activity. Leukocyte pulmonary recruitment, paradoxically, is diminished in infected ST8SiaIV-/- mice, corroborated by adoptive cell transfer, microfluidic migration experiments, and intravital microscopy, and possibly explained by dysregulated ERK1/2 signaling. PolySia is progressively lost from neutrophils and monocytes migrating from bone marrow to alveoli in Spn-infected WT mice, consistent with changing cellular functions. These data highlight multidimensional effects of polySia on leukocytes during an immune response and suggest therapeutic interventions for optimizing immunity.
Collapse
Affiliation(s)
- Prajakta Shinde
- Institute of Human Virology, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Alexander Kiepas
- Department of Chemical and Biomolecular Engineering, Whiting School of Engineering, Johns Hopkins University, Baltimore, MD 21218, USA
| | - Lei Zhang
- Institute of Human Virology, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Shreya Sudhir
- Institute of Human Virology, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Konstantinos Konstantopoulos
- Department of Chemical and Biomolecular Engineering, Whiting School of Engineering, Johns Hopkins University, Baltimore, MD 21218, USA
| | - Nicholas M Stamatos
- Institute of Human Virology, University of Maryland School of Medicine, Baltimore, MD 21201, USA.
| |
Collapse
|
15
|
Muñoz-Provencio D, Yebra MJ. Gut Microbial Sialidases and Their Role in the Metabolism of Human Milk Sialylated Glycans. Int J Mol Sci 2023; 24:9994. [PMID: 37373145 DOI: 10.3390/ijms24129994] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2023] [Revised: 05/26/2023] [Accepted: 06/08/2023] [Indexed: 06/29/2023] Open
Abstract
Sialic acids (SAs) are α-keto-acid sugars with a nine-carbon backbone present at the non-reducing end of human milk oligosaccharides and the glycan moiety of glycoconjugates. SAs displayed on cell surfaces participate in the regulation of many physiologically important cellular and molecular processes, including signaling and adhesion. Additionally, sialyl-oligosaccharides from human milk act as prebiotics in the colon by promoting the settling and proliferation of specific bacteria with SA metabolism capabilities. Sialidases are glycosyl hydrolases that release α-2,3-, α-2,6- and α-2,8-glycosidic linkages of terminal SA residues from oligosaccharides, glycoproteins and glycolipids. The research on sialidases has been traditionally focused on pathogenic microorganisms, where these enzymes are considered virulence factors. There is now a growing interest in sialidases from commensal and probiotic bacteria and their potential transglycosylation activity for the production of functional mimics of human milk oligosaccharides to complement infant formulas. This review provides an overview of exo-alpha-sialidases of bacteria present in the human gastrointestinal tract and some insights into their biological role and biotechnological applications.
Collapse
Affiliation(s)
- Diego Muñoz-Provencio
- Department of Food Biotechnology, Instituto de Agroquímica y Tecnología de Alimentos (IATA-CSIC), Av. Agustín Escardino 7, 46980 Paterna, Spain
| | - María J Yebra
- Department of Food Biotechnology, Instituto de Agroquímica y Tecnología de Alimentos (IATA-CSIC), Av. Agustín Escardino 7, 46980 Paterna, Spain
| |
Collapse
|
16
|
Lustig M, Chan C, Jansen JHM, Bräutigam M, Kölling MA, Gehlert CL, Baumann N, Mester S, Foss S, Andersen JT, Bastian L, Sondermann P, Peipp M, Burger R, Leusen JHW, Valerius T. Disruption of the sialic acid/Siglec-9 axis improves antibody-mediated neutrophil cytotoxicity towards tumor cells. Front Immunol 2023; 14:1178817. [PMID: 37346044 PMCID: PMC10279866 DOI: 10.3389/fimmu.2023.1178817] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2023] [Accepted: 05/22/2023] [Indexed: 06/23/2023] Open
Abstract
Upregulation of surface expressed sialoglycans on tumor cells is one of the mechanisms which promote tumor growth and progression. Specifically, the interactions of sialic acids with sialic acid-binding immunoglobulin-like lectins (Siglecs) on lymphoid or myeloid cells transmit inhibitory signals and lead to suppression of anti-tumor responses. Here, we show that neutrophils express among others Siglec-9, and that EGFR and HER2 positive breast tumor cells express ligands for Siglec-9. Treatment of tumor cells with neuraminidases or a sialyl transferase inhibitor significantly reduced binding of a soluble recombinant Siglec-9-Fc fusion protein, while EGFR and HER2 expression remained unchanged. Importantly, the cytotoxic activity of neutrophils driven by therapeutic EGFR or HER2 antibodies in vitro was increased by blocking the sialic acid/Siglec interaction, either by reducing tumor cell sialylation or by a Siglec-9 blocking antibody containing an effector silenced Fc domain. In vivo a short-term xenograft mouse model confirmed the improved therapeutic efficacy of EGFR antibodies against sialic acid depleted, by a sialyltransferase inhibitor, tumor cells compared to untreated cells. Our studies demonstrate that sialic acid/Siglec interactions between tumor cells and myeloid cells can impair antibody dependent tumor cell killing, and that Siglec-9 on polymorphonuclear cells (PMN) is critically involved. Considering that PMN are often a highly abundant cell population in the tumor microenvironment, Siglec-9 constitutes a promising target for myeloid checkpoint blockade to improve antibody-based tumor immunotherapy.
Collapse
Affiliation(s)
- Marta Lustig
- Division of Stem Cell Transplantation and Immunotherapy, Department of Medicine II, Christian-Albrechts-University Kiel and University Medical Center Schleswig-Holstein Campus Kiel, Kiel, Germany
| | - Chilam Chan
- Center for Translational Immunology, University Medical Center Utrecht, Utrecht, Netherlands
| | - J. H. Marco Jansen
- Center for Translational Immunology, University Medical Center Utrecht, Utrecht, Netherlands
| | | | - Max A. Kölling
- Division of Stem Cell Transplantation and Immunotherapy, Department of Medicine II, Christian-Albrechts-University Kiel and University Medical Center Schleswig-Holstein Campus Kiel, Kiel, Germany
| | - Carina Lynn Gehlert
- Division of Antibody-Based Immunotherapy, Department of Medicine II, Christian-Albrechts-University Kiel and University Medical Center Schleswig-Holstein Campus Kiel, Kiel, Germany
| | - Niklas Baumann
- Division of Stem Cell Transplantation and Immunotherapy, Department of Medicine II, Christian-Albrechts-University Kiel and University Medical Center Schleswig-Holstein Campus Kiel, Kiel, Germany
| | - Simone Mester
- Institute for Clinical Medicine, Department of Pharmacology, University of Oslo and Oslo University Hospital, Oslo, Norway
- Institute for Clinical Medicine, Department of Immunology, University of Oslo and Oslo University Hospital, Oslo, Norway
| | - Stian Foss
- Institute for Clinical Medicine, Department of Pharmacology, University of Oslo and Oslo University Hospital, Oslo, Norway
- Institute for Clinical Medicine, Department of Immunology, University of Oslo and Oslo University Hospital, Oslo, Norway
| | - Jan Terje Andersen
- Institute for Clinical Medicine, Department of Pharmacology, University of Oslo and Oslo University Hospital, Oslo, Norway
- Institute for Clinical Medicine, Department of Immunology, University of Oslo and Oslo University Hospital, Oslo, Norway
| | - Lorenz Bastian
- Department of Medicine II, Christian-Albrechts-University Kiel and University Medical Center Schleswig-Holstein, Kiel, Germany
| | | | - Matthias Peipp
- Division of Antibody-Based Immunotherapy, Department of Medicine II, Christian-Albrechts-University Kiel and University Medical Center Schleswig-Holstein Campus Kiel, Kiel, Germany
| | - Renate Burger
- Division of Stem Cell Transplantation and Immunotherapy, Department of Medicine II, Christian-Albrechts-University Kiel and University Medical Center Schleswig-Holstein Campus Kiel, Kiel, Germany
| | - Jeanette H. W. Leusen
- Center for Translational Immunology, University Medical Center Utrecht, Utrecht, Netherlands
| | - Thomas Valerius
- Division of Stem Cell Transplantation and Immunotherapy, Department of Medicine II, Christian-Albrechts-University Kiel and University Medical Center Schleswig-Holstein Campus Kiel, Kiel, Germany
| |
Collapse
|
17
|
Sun X, Cao J, Sun P, Yang H, Li H, Ma W, Wu X, He X, Li J, Li Z, Huang J. Pretreatment soluble Siglec-5 protein predicts early progression and R-CHOP efficacy in diffuse large B-cell lymphoma. Biomark Med 2023; 17:143-158. [PMID: 37097021 DOI: 10.2217/bmm-2022-0764] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/26/2023] Open
Abstract
Aims: To explore the clinical association between soluble Siglec-5/CD163 and clinical feature and prognosis in peripheral blood samples of patients with diffuse large B-cell lymphoma. Method: Significantly elevated cytokines in peripheral blood were characterized by cytokines array and validated by ELISA. Results: Compared with CD163, Siglec-5 exhibited superiority in discriminating patients into low- and high-risk subgroups based on overall survival and progression-free survival. In addition, Siglec-5 was an indicator of rituximab plus cyclophosphamide, doxorubicin, vincristine and prednisone (R-CHOP) treatment efficacy. Conclusion: Siglec-5 may be applied as a reliable independent immune indicator for overall survival and progression-free survival. It may also predict R-CHOP efficacy in diffuse large B-cell lymphoma.
Collapse
Affiliation(s)
- Xiaoqing Sun
- Department of Intensive Care Unit(ICU), State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center, Guangzhou, 510060, PR China
| | - Jianghua Cao
- Department of Medical Oncology, State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center, Guangzhou, 510060, PR China
| | - Peng Sun
- Department of Medical Oncology, State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center, Guangzhou, 510060, PR China
| | - Hang Yang
- Department of Medical Oncology, State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center, Guangzhou, 510060, PR China
| | - Huan Li
- Department of Intensive Care Unit(ICU), State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center, Guangzhou, 510060, PR China
| | - Wenjuan Ma
- Department of Intensive Care Unit(ICU), State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center, Guangzhou, 510060, PR China
| | - Xianqiu Wu
- Department of Hepatobiliary Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Xiaohua He
- Department of Medical Oncology, State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center, Guangzhou, 510060, PR China
| | - Jing Li
- Department of Medical Oncology, State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center, Guangzhou, 510060, PR China
| | - Zhiming Li
- Department of Medical Oncology, State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center, Guangzhou, 510060, PR China
| | - Jiajia Huang
- Department of Medical Oncology, State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center, Guangzhou, 510060, PR China
| |
Collapse
|
18
|
Schmidt EN, Lamprinaki D, McCord KA, Joe M, Sojitra M, Waldow A, Nguyen J, Monyror J, Kitova EN, Mozaneh F, Guo XY, Jung J, Enterina JR, Daskhan GC, Han L, Krysler AR, Cromwell CR, Hubbard BP, West LJ, Kulka M, Sipione S, Klassen JS, Derda R, Lowary TL, Mahal LK, Riddell MR, Macauley MS. Siglec-6 mediates the uptake of extracellular vesicles through a noncanonical glycolipid binding pocket. Nat Commun 2023; 14:2327. [PMID: 37087495 PMCID: PMC10122656 DOI: 10.1038/s41467-023-38030-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2022] [Accepted: 04/12/2023] [Indexed: 04/24/2023] Open
Abstract
Immunomodulatory Siglecs are controlled by their glycoprotein and glycolipid ligands. Siglec-glycolipid interactions are often studied outside the context of a lipid bilayer, missing the complex behaviors of glycolipids in a membrane. Through optimizing a liposomal formulation to dissect Siglec-glycolipid interactions, it is shown that Siglec-6 can recognize glycolipids independent of its canonical binding pocket, suggesting that Siglec-6 possesses a secondary binding pocket tailored for recognizing glycolipids in a bilayer. A panel of synthetic neoglycolipids is used to probe the specificity of this glycolipid binding pocket on Siglec-6, leading to the development of a neoglycolipid with higher avidity for Siglec-6 compared to natural glycolipids. This neoglycolipid facilitates the delivery of liposomes to Siglec-6 on human mast cells, memory B-cells and placental syncytiotrophoblasts. A physiological relevance for glycolipid recognition by Siglec-6 is revealed for the binding and internalization of extracellular vesicles. These results demonstrate a unique and physiologically relevant ability of Siglec-6 to recognize glycolipids in a membrane.
Collapse
Affiliation(s)
- Edward N Schmidt
- Department of Chemistry, University of Alberta, Edmonton, AB, Canada
| | | | - Kelli A McCord
- Department of Chemistry, University of Alberta, Edmonton, AB, Canada
| | - Maju Joe
- Department of Chemistry, University of Alberta, Edmonton, AB, Canada
| | - Mirat Sojitra
- Department of Chemistry, University of Alberta, Edmonton, AB, Canada
| | - Ayk Waldow
- Department of Chemistry, University of Alberta, Edmonton, AB, Canada
| | - Jasmine Nguyen
- Department of Obstetrics & Gynaecology and Physiology University of Alberta, Edmonton, AB, Canada
| | - John Monyror
- Department of Pharmacology, University of Alberta, Edmonton, AB, Canada
- Neuroscience and Mental Health Institute, University of Alberta, Edmonton, AB, Canada
| | - Elena N Kitova
- Department of Chemistry, University of Alberta, Edmonton, AB, Canada
| | - Fahima Mozaneh
- Department of Chemistry, University of Alberta, Edmonton, AB, Canada
| | - Xue Yan Guo
- Department of Chemistry, University of Alberta, Edmonton, AB, Canada
| | - Jaesoo Jung
- Department of Chemistry, University of Alberta, Edmonton, AB, Canada
| | - Jhon R Enterina
- Department of Medical Microbiology and Immunology, University of Alberta, Edmonton, AB, Canada
| | - Gour C Daskhan
- Department of Chemistry, University of Alberta, Edmonton, AB, Canada
| | - Ling Han
- Department of Chemistry, University of Alberta, Edmonton, AB, Canada
| | - Amanda R Krysler
- Department of Pharmacology, University of Alberta, Edmonton, AB, Canada
| | | | - Basil P Hubbard
- Department of Pharmacology, University of Alberta, Edmonton, AB, Canada
- Department of Pharmacology and Toxicology, University of Toronto, Toronto, ON, Canada
| | - Lori J West
- Department of Medical Microbiology and Immunology, University of Alberta, Edmonton, AB, Canada
- Department of Pediatrics, University of Alberta, Edmonton, AB, Canada
- Department of Surgery, University of Alberta, Edmonton, AB, Canada
| | - Marianne Kulka
- Department of Medical Microbiology and Immunology, University of Alberta, Edmonton, AB, Canada
- National Research Council, Edmonton, AB, Canada
| | - Simonetta Sipione
- Department of Pharmacology, University of Alberta, Edmonton, AB, Canada
- Neuroscience and Mental Health Institute, University of Alberta, Edmonton, AB, Canada
| | - John S Klassen
- Department of Chemistry, University of Alberta, Edmonton, AB, Canada
| | - Ratmir Derda
- Department of Chemistry, University of Alberta, Edmonton, AB, Canada
| | - Todd L Lowary
- Department of Chemistry, University of Alberta, Edmonton, AB, Canada
- Institute of Biological Chemistry, Academia Sinica, Nangang, Taipei, Taiwan
- Institute of Biochemical Sciences, National Taiwan University, Taipei, Taiwan
| | - Lara K Mahal
- Department of Chemistry, University of Alberta, Edmonton, AB, Canada
| | - Meghan R Riddell
- Department of Obstetrics & Gynaecology and Physiology University of Alberta, Edmonton, AB, Canada
| | - Matthew S Macauley
- Department of Chemistry, University of Alberta, Edmonton, AB, Canada.
- Neuroscience and Mental Health Institute, University of Alberta, Edmonton, AB, Canada.
- Department of Medical Microbiology and Immunology, University of Alberta, Edmonton, AB, Canada.
| |
Collapse
|
19
|
Vargas RA, Soto-Aguilera S, Parra M, Herrera S, Santibañez A, Kossack C, Saavedra CP, Mora O, Pineda M, Gonzalez O, Gonzalez A, Maisey K, Torres-Maravilla E, Bermúdez-Humarán LG, Suárez-Villota EY, Tello M. Analysis of microbiota-host communication mediated by butyrate in Atlantic Salmon. Comput Struct Biotechnol J 2023; 21:2558-2578. [PMID: 37122632 PMCID: PMC10130356 DOI: 10.1016/j.csbj.2023.03.050] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2022] [Revised: 03/28/2023] [Accepted: 03/29/2023] [Indexed: 04/03/2023] Open
Abstract
Butyrate is a microbiota-produced metabolite, sensed by host short-chain fatty acid receptors FFAR2 (Gpr43), FFAR3 (Gpr41), HCAR2 (Gpr109A), and Histone deacetylase (HDAC) that promotes microbiota-host crosstalk. Butyrate influences energy uptake, developmental and immune response in mammals. This microbial metabolite is produced by around 79 anaerobic genera present in the mammalian gut, yet little is known about the role of butyrate in the host-microbiota interaction in salmonid fish. To further our knowledge of this interaction, we analyzed the intestinal microbiota and genome of Atlantic salmon (Salmo salar), searching for butyrate-producing genera and host butyrate receptors. We identified Firmicutes, Proteobacteria, and Actinobacteria as the main butyrate-producing bacteria in the salmon gut microbiota. In the Atlantic salmon genome, we identified an expansion of genes orthologous to FFAR2 and HCAR2 receptors, and class I and IIa HDACs that are sensitive to butyrate. In addition, we determined the expression levels of orthologous of HCAR2 in the gut, spleen, and head-kidney, and FFAR2 in RTgutGC cells. The effect of butyrate on the Atlantic salmon immune response was evaluated by analyzing the pro and anti-inflammatory cytokines response in vitro in SHK-1 cells by RT-qPCR. Butyrate decreased the expression of the pro-inflammatory cytokine IL-1β and increased anti-inflammatory IL-10 and TGF-β cytokines. Butyrate also reduced the expression of interferon-alpha, Mx, and PKR, and decreased the viral load at a higher concentration (4 mM) in cells treated with this molecule before the infection with Infectious Pancreatic Necrosis Virus (IPNV) by mechanisms independent of FFAR2, FFAR3 and HCAR2 expression that probably inhibit HDAC. Moreover, butyrate modified phosphorylation of cytoplasmic proteins in RTgutGC cells. Our data allow us to infer that Atlantic salmon have the ability to sense butyrate produced by their gut microbiota via different specific targets, through which butyrate modulates the immune response of pro and anti-inflammatory cytokines and the antiviral response.
Collapse
|
20
|
Prenzler S, Rudrawar S, Waespy M, Kelm S, Anoopkumar-Dukie S, Haselhorst T. The role of sialic acid-binding immunoglobulin-like-lectin-1 (siglec-1) in immunology and infectious disease. Int Rev Immunol 2023; 42:113-138. [PMID: 34494938 DOI: 10.1080/08830185.2021.1931171] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
Siglec-1, also known as Sialoadhesin (Sn) and CD169 is highly conserved among vertebrates and with 17 immunoglobulin-like domains is Siglec-1 the largest member of the Siglec family. Expression of Siglec-1 is found primarily on dendritic cells (DCs), macrophages and interferon induced monocyte. The structure of Siglec-1 is unique among siglecs and its function as a receptor is also different compared to other receptors in this class as it contains the most extracellular domains out of all the siglecs. However, the ability of Siglec-1 to internalize antigens and to pass them on to lymphocytes by allowing dendritic cells and macrophages to act as antigen presenting cells, is the main reason that has granted Siglec-1's key role in multiple human disease states including atherosclerosis, coronary artery disease, autoimmune diseases, cell-cell signaling, immunology, and more importantly bacterial and viral infections. Enveloped viruses for example have been shown to manipulate Siglec-1 to increase their virulence by binding to sialic acids present on the virus glycoproteins allowing them to spread or evade immune response. Siglec-1 mediates dissemination of HIV-1 in activated tissues enhancing viral spread via infection of DC/T-cell synapses. Overall, the ability of Siglec-1 to bind a variety of target cells within the immune system such as erythrocytes, B-cells, CD8+ granulocytes and NK cells, highlights that Siglec-1 is a unique player in these essential processes.
Collapse
Affiliation(s)
- Shane Prenzler
- School of Pharmacy and Medical Sciences, Griffith University, Gold Coast, Queensland, Australia.,Menzies Health Institute Queensland, Griffith University, Gold Coast, Queensland, Australia
| | - Santosh Rudrawar
- School of Pharmacy and Medical Sciences, Griffith University, Gold Coast, Queensland, Australia.,Menzies Health Institute Queensland, Griffith University, Gold Coast, Queensland, Australia
| | - Mario Waespy
- Centre for Biomolecular Interactions Bremen, Department of Biology and Chemistry, University of Bremen, Bremen, Germany
| | - Sørge Kelm
- Centre for Biomolecular Interactions Bremen, Department of Biology and Chemistry, University of Bremen, Bremen, Germany
| | - Shailendra Anoopkumar-Dukie
- School of Pharmacy and Medical Sciences, Griffith University, Gold Coast, Queensland, Australia.,Menzies Health Institute Queensland, Griffith University, Gold Coast, Queensland, Australia
| | - Thomas Haselhorst
- Institute for Glycomics, Griffith University, Gold Coast, Queensland, Australia
| |
Collapse
|
21
|
The Blessed Union of Glycobiology and Immunology: A Marriage That Worked. MEDICINES (BASEL, SWITZERLAND) 2023; 10:medicines10020015. [PMID: 36827215 PMCID: PMC9967969 DOI: 10.3390/medicines10020015] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/08/2022] [Revised: 01/03/2023] [Accepted: 01/17/2023] [Indexed: 01/21/2023]
Abstract
In this article, we discuss the main aspects regarding the recognition of cell surface glycoconjugates and the immunomodulation of responses against the progression of certain pathologies, such as cancer and infectious diseases. In the first part, we talk about different aspects of glycoconjugates and delve deeper into the importance of N-glycans in cancer immunotherapy. Then, we describe two important lectin families that have been very well studied in the last 20 years. Examples include the sialic acid-binding immunoglobulin (Ig)-like lectins (siglecs), and galectins. Finally, we discuss a topic that needs to be better addressed in the field of glycoimmunology: the impact of oncofetal antigens on the cells of the immune system. New findings in this area are of great importance for advancement, especially in the field of oncology, since it is already known that cellular interactions mediated by carbohydrate-carbohydrate and/or carbohydrate proteins are able to modulate the progression of different types of cancer in events that compromise the functionality of the immune responses.
Collapse
|
22
|
Huang R, Zheng J, Shao Y, Zhu L, Yang T. Siglec-15 as multifunctional molecule involved in osteoclast differentiation, cancer immunity and microbial infection. PROGRESS IN BIOPHYSICS AND MOLECULAR BIOLOGY 2023; 177:34-41. [PMID: 36265694 DOI: 10.1016/j.pbiomolbio.2022.10.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/28/2021] [Revised: 09/19/2022] [Accepted: 10/13/2022] [Indexed: 11/06/2022]
Abstract
Siglec-15 is a highly conserved member of the Siglec family, expressed on osteoclasts, a subset of myeloid cells and some cancer cells. Except for regulating osteoclast differentiation, Siglec-15 engages in immunoregulation as an immune suppressor. Siglec-15 functions as an immunosuppressive molecule in tumor-associated macrophage-mediated T cell immunity in the tumor microenvironment (TME), which makes Siglec-15 to be an emerging and promising target for normalization cancer immunotherapy. Besides, Siglec-15 interacts with sialylated pathogens and modulates host immune response against microbial pathogens by altering cytokine production and/or phagocytosis, which further broadens the underlying pathophysiological roles of Siglec-15. The fact that N-glycosylation and sialylation of Siglec-15 play a pivotal role in Siglec-15 biological function indicates that targeting certain post-translational modification may be an effective strategy for targeting Siglec-15 therapy. In-depth exploring Siglec-15 biology function is crucial for better design of Siglec-15-based therapy according to different clinical indications.
Collapse
Affiliation(s)
- Rui Huang
- Department of Biochemistry & Molecular Biology, Shanxi Medical University, Taiyuan, China; Department of Clinical Laboratory, Children's Hospital and Women Health Center of Shanxi, Taiyuan, China
| | - Jinxiu Zheng
- Department of Biochemistry & Molecular Biology, Shanxi Medical University, Taiyuan, China; Key Laboratory of Cellular Physiology (Shanxi Medical University), Ministry of Education, Taiyuan, China
| | - Ying Shao
- Department of Pathophysiology, Shanxi Medical University, Taiyuan, China
| | - Lei Zhu
- Department of Clinical Laboratory, Children's Hospital and Women Health Center of Shanxi, Taiyuan, China
| | - Tao Yang
- Department of Biochemistry & Molecular Biology, Shanxi Medical University, Taiyuan, China; Key Laboratory of Cellular Physiology (Shanxi Medical University), Ministry of Education, Taiyuan, China.
| |
Collapse
|
23
|
Dudek B, Rybka J, Bugla-Płoskońska G, Korzeniowska-Kowal A, Futoma-Kołoch B, Pawlak A, Gamian A. Biological functions of sialic acid as a component of bacterial endotoxin. Front Microbiol 2022; 13:1028796. [PMID: 36338080 PMCID: PMC9631793 DOI: 10.3389/fmicb.2022.1028796] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2022] [Accepted: 10/05/2022] [Indexed: 11/28/2022] Open
Abstract
Lipopolysaccharide (endotoxin, LPS) is an important Gram-negative bacteria antigen. LPS of some bacteria contains sialic acid (Neu5Ac) as a component of O-antigen (O-Ag), in this review we present an overview of bacteria in which the presence of Neu5Ac has been confirmed in their outer envelope and the possible ways that bacteria can acquire Neu5Ac. We explain the role of Neu5Ac in bacterial pathogenesis, and also involvement of Neu5Ac in bacterial evading the host innate immunity response and molecular mimicry phenomenon. We also highlight the role of sialic acid in the mechanism of bacterial resistance to action of serum complement. Despite a number of studies on involvement of Neu5Ac in bacterial pathogenesis many aspects of this phenomenon are still not understood.
Collapse
Affiliation(s)
- Bartłomiej Dudek
- Department of Microbiology, University of Wrocław, Wrocław, Poland
- *Correspondence: Bartłomiej Dudek,
| | - Jacek Rybka
- Department of Immunology of Infectious Diseases, Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, Wrocław, Poland
| | | | - Agnieszka Korzeniowska-Kowal
- Department of Immunology of Infectious Diseases, Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, Wrocław, Poland
| | | | | | - Andrzej Gamian
- Department of Immunology of Infectious Diseases, Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, Wrocław, Poland
- Andrzej Gamian,
| |
Collapse
|
24
|
Almeida‐Marrero V, Bethlehem F, Longo S, Bertolino MC, Torres T, Huskens J, de la Escosura A. Tailored Multivalent Targeting of Siglecs with Photosensitizing Liposome Nanocarriers. Angew Chem Int Ed Engl 2022; 61:e202206900. [PMID: 35652453 PMCID: PMC9401027 DOI: 10.1002/anie.202206900] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2022] [Indexed: 11/18/2022]
Abstract
The modification of surfaces with multiple ligands allows the formation of platforms for the study of multivalency in diverse processes. Herein we use this approach for the implementation of a photosensitizer (PS)-nanocarrier system that binds efficiently to siglec-10, a member of the CD33 family of siglecs (sialic acid (SA)-binding immunoglobulin-like lectins). In particular, a zinc phthalocyanine derivative bearing three SA moieties (PcSA) has been incorporated in the membrane of small unilamellar vesicles (SUVs), retaining its photophysical properties upon insertion into the SUV's membrane. The interaction of these biohybrid systems with human siglec-10-displaying supported lipid bilayers (SLBs) has shown the occurrence of weakly multivalent, superselective interactions between vesicle and SLB. The SLB therefore acts as an excellent cell membrane mimic, while the binding with PS-loaded SUVs shows the potential for targeting siglec-expressing cells with photosensitizing nanocarriers.
Collapse
Affiliation(s)
- Verónica Almeida‐Marrero
- Department of Organic ChemistryUniversidad Autónoma de MadridCampus de Cantoblanco28049MadridSpain
| | - Fleur Bethlehem
- Department of Molecules & MaterialsMESA+ Institute for NanotechnologyFaculty of Science and TechnologyUniversity of TwenteP.O. Box 2177500 AEEnschedeThe Netherlands
| | - Sara Longo
- Department of Molecules & MaterialsMESA+ Institute for NanotechnologyFaculty of Science and TechnologyUniversity of TwenteP.O. Box 2177500 AEEnschedeThe Netherlands
| | - M. Candelaria Bertolino
- Department of Molecules & MaterialsMESA+ Institute for NanotechnologyFaculty of Science and TechnologyUniversity of TwenteP.O. Box 2177500 AEEnschedeThe Netherlands
| | - Tomás Torres
- Department of Organic ChemistryUniversidad Autónoma de MadridCampus de Cantoblanco28049MadridSpain
- Institute for Advanced Research in Chemistry (IAdChem)Campus de Cantoblanco28049MadridSpain
- Tomás TorresIMDEA NanoscienceCampus de Cantoblanco28049MadridSpain
| | - Jurriaan Huskens
- Department of Molecules & MaterialsMESA+ Institute for NanotechnologyFaculty of Science and TechnologyUniversity of TwenteP.O. Box 2177500 AEEnschedeThe Netherlands
| | - Andrés de la Escosura
- Department of Organic ChemistryUniversidad Autónoma de MadridCampus de Cantoblanco28049MadridSpain
- Institute for Advanced Research in Chemistry (IAdChem)Campus de Cantoblanco28049MadridSpain
| |
Collapse
|
25
|
Lende SSF, Pahus MH, Monrad I, Olesen R, Mahr AR, Vibholm LK, Østergaard L, Søgaard OS, Andersen AHF, Denton PW, Tolstrup M. CD169 (Siglec-1) as a Robust Human Cell Biomarker of Toll-Like Receptor 9 Agonist Immunotherapy. Front Cell Infect Microbiol 2022; 12:919097. [PMID: 35865810 PMCID: PMC9294151 DOI: 10.3389/fcimb.2022.919097] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2022] [Accepted: 06/14/2022] [Indexed: 11/13/2022] Open
Abstract
Immunotherapy is a promising therapeutic area in cancer and chronic viral infections. An important component of immunotherapy in these contexts is the activation of innate immunity. Here we investigate the potential for CD169 (Siglec 1) expression on monocytes to serve as a robust biomarker for activation of innate immunity and, particular, as a proxy for IFN-α production. Specifically, we investigated the effects of Toll-like receptor 9 agonism with MGN1703 (lefitolimod) across experimental conditions ex vivo, in humanized mice, and in clinical trial participants. Ex vivo we observed that the percentage of classical monocytes expressing CD169 increased dramatically from 10% pre-stimulation to 97% 24 hrs after MGN1703 stimulation (p<0.0001). In humanized NOG mice, we observed prominent upregulation of the proportions of monocytes expressing CD169 after two doses of MGN1703 where 73% of classical monocytes were CD169 positive in bone marrow following MGN1703 treatment vs 19% in vehicle treated mice (p=0.0159). Finally, in a clinical trial in HIV-infected individuals receiving immunotherapy treatment with MGN1703, we observed a uniform upregulation of CD169 on monocytes after dosing with 97% of classical monocytes positive for CD169 (p=0.002). Hence, in this comprehensive evaluation ex vivo, in an animal model, and in a clinical trial, we find increases in the percentage of CD169 positive monocytes to be a reliable and robust biomarker of immune activation following TLR9 agonist treatment.
Collapse
Affiliation(s)
| | - Marie Høst Pahus
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Ida Monrad
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Rikke Olesen
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
- Department of Infectious Diseases, Aarhus University Hospital, Aarhus, Denmark
| | - Anna R. Mahr
- Department of Biology, University of Nebraska at Omaha, Omaha, NE, United States
| | - Line K. Vibholm
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
- Department of Infectious Diseases, Aarhus University Hospital, Aarhus, Denmark
| | - Lars Østergaard
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
- Department of Infectious Diseases, Aarhus University Hospital, Aarhus, Denmark
| | - Ole Schmeltz Søgaard
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
- Department of Infectious Diseases, Aarhus University Hospital, Aarhus, Denmark
| | | | - Paul W. Denton
- Department of Biology, University of Nebraska at Omaha, Omaha, NE, United States
| | - Martin Tolstrup
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
- Department of Infectious Diseases, Aarhus University Hospital, Aarhus, Denmark
- *Correspondence: Martin Tolstrup,
| |
Collapse
|
26
|
Wu J, Peng J, Zhou Y, Zhang R, Wang Z, Hu N, Zhang D, Quan G, Wu Y, Feng J, Shen B, Zhao J, Zhang Y, Yang K, Luo L. Screening and Identification of A Novel Anti-Siglec-15 Human Antibody 3F1 and Relevant Antitumor Activity. Mol Pharmacol 2022; 102:161-171. [PMID: 35764384 DOI: 10.1124/molpharm.121.000470] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Accepted: 06/19/2022] [Indexed: 11/22/2022] Open
Abstract
Siglec-15 is an important immunosuppressive molecule considered to be a key target in next generation tumor immunotherapy. In this study, we screened 22 high affinity antibodies that specifically recognize human siglec-15 by using a large human phage antibody library and 5 representative sequences were selected for further study. The results showed the binding activity of 5 antibodies to siglec-15 (EC50 ranged from 0.02368μg /mL to 0.07949 μg /mL) and in 2 siglec-15 overexpressed cell lines, 3 antibodies had the strongest binding activity, so the 2 clones were discarded for further study. Subsequently the affinity of 3 antibodies were measured by Bio-layer interferometry technology (5-9×10E-09M). As the reported ligands of siglec-15, the binding activity of siglec-15 and sialyl-Tn, CD44, MAG and LRRC4C can be blocked by 3 of the antibodies. Among these, 3F1 had a competitive advantage. Then, the antibody 3F1 showed an obvious ADCC effect (EC50 were 0.85 μg/mL). Further, antibody 3F1 can reverse the inhibitory effect of siglec-15 on lymphocyte proliferation (especially CD4+T, CD8+T) and cytokine release (IFN-γ). Given the above results, 3F1 was selected as a candidate for the in vivo pharmacodynamics study. In the tumor model of Balb/c Nude mice, 3F1 (10 mg/kg) showed certain anti-tumor effects (TGI was 31.5%) while the combination of 3F1 (5 mg/kg) and ERBITUX (5 mg/kg) showed significant antitumor effects (TGI was 48.7%) compared with the PBS group. In conclusion, novel human antibody 3F1 has anti-tumor activity and is expected to be an innovative candidate drug targeting siglec-15 for tumor immunotherapy Significance Statement Siglec-15 is considered as an important target in the next generation of tumor immunotherapy. 3F1 is expected to be the most promising potential candidate for targeting siglec-15 for cancer treatment, and could provide a reference for the development of anti-tumor drugs.
Collapse
Affiliation(s)
- Jiaguo Wu
- Dali University; Institute of Pharmacology and Toxicology, China
| | - Jingyi Peng
- Institute of Pharmacology and Toxicology, China
| | | | - Ran Zhang
- Hunan Normal University School of Medicine, China
| | - Zhihong Wang
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, China
| | - Naijing Hu
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, China
| | - Dingmu Zhang
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, China
| | - Guiqi Quan
- Institute of Pharmacology and Toxicology, China
| | - Yuanyu Wu
- Institute of Pharmacology and Toxicology, China
| | - Jiannan Feng
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, China
| | - Beifen Shen
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, China
| | | | - Yan Zhang
- Department of Obstetrics and Gynecology, First Medical Center, General Hospital of Chinese PLA, China
| | - Kaiming Yang
- School of Basic Medical Sciences of Dali University, China
| | | |
Collapse
|
27
|
Almeida-Marrero V, Bethlehem F, Longo S, Bertolino MC, Torres T, Huskens J, de la Escosura A. Tailored Multivalent Targeting of Siglecs with Photosensitizing Liposome Nanocarriers. Angew Chem Int Ed Engl 2022. [DOI: 10.1002/ange.202206900] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Affiliation(s)
- Verónica Almeida-Marrero
- Universidad Autonoma de Madrid - Campus de Cantoblanco: Universidad Autonoma de Madrid Organic Chemistry SPAIN
| | - Fleur Bethlehem
- University of Twente Institute for Nanotechnology: Universiteit Twente MESA+ Institute for Nanotechnology MESA+ NETHERLANDS
| | - Sara Longo
- University of Twente Institute for Nanotechnology: Universiteit Twente MESA+ Institute for Nanotechnology MESA+ NETHERLANDS
| | - M. Candelaria Bertolino
- University of Twente Institute for Nanotechnology: Universiteit Twente MESA+ Institute for Nanotechnology MESA+ NETHERLANDS
| | - Tomas Torres
- Universidad Autonoma de Madrid - Campus de Cantoblanco: Universidad Autonoma de Madrid Departmento de Química Orgánica Cantoblanco 28049 Madrid SPAIN
| | - Jurriaan Huskens
- University of Twente Institute for Nanotechnology: Universiteit Twente MESA+ Institute for Nanotechnology MESA+ NETHERLANDS
| | - Andrés de la Escosura
- Universidad Autonoma de Madrid - Campus de Cantoblanco: Universidad Autonoma de Madrid Organic Chemistry C/ Francisco Tomás y Valiente 7, Facultad de CienciasMódulo 01, Planta 4, L-401 28049 Madrid SPAIN
| |
Collapse
|
28
|
Wielgat P, Narejko K, Car H. SARS-CoV-2 Attacks in the Brain: Focus on the Sialome. Cells 2022; 11:1458. [PMID: 35563764 PMCID: PMC9104523 DOI: 10.3390/cells11091458] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Revised: 04/20/2022] [Accepted: 04/24/2022] [Indexed: 12/16/2022] Open
Abstract
The epidemiological observations suggest that respiratory and gastrointestinal symptoms caused by severe acute respiratory coronavirus 2 (SARS-CoV-2) are accompanied by short- and long-term neurological manifestations. There is increasing evidence that the neuroinvasive potential of SARS-CoV-2 is closely related to its capacity to interact with cell membrane sialome. Given the wide expression of sialylated compounds of cell membranes in the brain, the interplay between cell membrane sialoglycans and the virus is crucial for its attachment and cell entry, transport, neuronal damage and brain immunity. Here, we focus on the significance of the brain sialome in the progress of coronavirus disease 2019 (COVID-19) and SARS-CoV-2-induced neuropathology.
Collapse
Affiliation(s)
- Przemyslaw Wielgat
- Department of Clinical Pharmacology, Medical University of Bialystok, Waszyngtona 15A, 15-274 Bialystok, Poland; (K.N.); (H.C.)
| | - Karolina Narejko
- Department of Clinical Pharmacology, Medical University of Bialystok, Waszyngtona 15A, 15-274 Bialystok, Poland; (K.N.); (H.C.)
| | - Halina Car
- Department of Clinical Pharmacology, Medical University of Bialystok, Waszyngtona 15A, 15-274 Bialystok, Poland; (K.N.); (H.C.)
- Department of Experimental Pharmacology, Medical University of Bialystok, Szpitalna 37, 15-265 Bialystok, Poland
| |
Collapse
|
29
|
Ozdilek A, Avci FY. Glycosylation as a key parameter in the design of nucleic acid vaccines. Curr Opin Struct Biol 2022; 73:102348. [PMID: 35255387 PMCID: PMC8957583 DOI: 10.1016/j.sbi.2022.102348] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2021] [Revised: 01/25/2022] [Accepted: 01/30/2022] [Indexed: 01/21/2023]
Abstract
Vaccine-induced immunity is expected to target the native antigens expressed by the pathogens. Therefore, it is highly important to generate vaccine antigens that are immunologically indistinguishable from the native antigens. Nucleic acid vaccines, comprised of DNA, mRNA, or recombinant viral vector vaccines, introduce the genetic material encoding the antigenic protein for the host to express. Because these proteins will undergo host posttranslational modifications, host glycosylation can potentially alter the structure and immunological efficacy of the antigen. In this review, we discuss the potential impact of host protein glycosylation on the immune responses generated by nucleic acid vaccines against bacterial and viral pathogens.
Collapse
Affiliation(s)
- Ahmet Ozdilek
- Department of Biochemistry and Molecular Biology, Center for Molecular Medicine, The University of Georgia, Athens, Georgia, USA
| | - Fikri Y Avci
- Department of Biochemistry and Molecular Biology, Center for Molecular Medicine, The University of Georgia, Athens, Georgia, USA.
| |
Collapse
|
30
|
Saad AA. Targeting cancer-associated glycans as a therapeutic strategy in leukemia. ALL LIFE 2022. [DOI: 10.1080/26895293.2022.2049901] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022] Open
Affiliation(s)
- Ashraf Abdullah Saad
- Unit of Pediatric Hematologic Oncology and BMT, Sultan Qaboos University Hospital, Muscat, Oman
| |
Collapse
|
31
|
Systemic Lectin-Glycan Interaction of Pathogenic Enteric Bacteria in the Gastrointestinal Tract. Int J Mol Sci 2022; 23:ijms23031451. [PMID: 35163392 PMCID: PMC8835900 DOI: 10.3390/ijms23031451] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Revised: 01/20/2022] [Accepted: 01/25/2022] [Indexed: 01/27/2023] Open
Abstract
Microorganisms, such as bacteria, viruses, and fungi, and host cells, such as plants and animals, have carbohydrate chains and lectins that reciprocally recognize one another. In hosts, the defense system is activated upon non-self-pattern recognition of microbial pathogen-associated molecular patterns. These are present in Gram-negative and Gram-positive bacteria and fungi. Glycan-based PAMPs are bound to a class of lectins that are widely distributed among eukaryotes. The first step of bacterial infection in humans is the adhesion of the pathogen's lectin-like proteins to the outer membrane surfaces of host cells, which are composed of glycans. Microbes and hosts binding to each other specifically is of critical importance. The adhesion factors used between pathogens and hosts remain unknown; therefore, research is needed to identify these factors to prevent intestinal infection or treat it in its early stages. This review aims to present a vision for the prevention and treatment of infectious diseases by identifying the role of the host glycans in the immune response against pathogenic intestinal bacteria through studies on the lectin-glycan interaction.
Collapse
|
32
|
Wu Y, Yang D, Chen GY. The role of the Siglec-G ITIM domain during bacterial infection. Cell Mol Biol (Noisy-le-grand) 2022; 67:163-169. [PMID: 35809291 PMCID: PMC11397909 DOI: 10.14715/cmb/2021.67.4.18] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2022] [Indexed: 01/16/2023]
Abstract
Siglecs, membrane-bound lectins of the sialic acid-binding immunoglobulin superfamily, inhibit immune responses by recruiting tyrosine phosphatases (e.g., SHP-1 and SHP-2) through their cytoplasmic immunoreceptor tyrosine-based inhibition motif (ITIM) domain. The role of Siglecs in infection has been extensively studied, but downstream signaling through the ITIM domain remains unclear. Here, we used a GST pull-down assay to identify additional proteins associated with the ITIM domain during bacterial infection. Gdi2 bound to ITIM under normal homeostasis, but Rab1a was recruited to ITIM during bacterial infection. Western blot analysis confirmed the presence of SHP-1 and SHP-2 in eluted ITIM-associated proteins under normal homeostasis. We confirmed the association of ITIM with Gdi2 or Rab1a by transfection of corresponding expression vectors in 293T cells followed by immunoprecipitation-western blot assay. Thus, ITIM's role in the inhibition of the immune response during bacterial infection may be regulated by interaction with Gdi2 and Rab1a in addition to SHP-1 and SHP-2.
Collapse
Affiliation(s)
- Yin Wu
- Children's Foundation Research Institute at Le Bonheur Children's Hospital, Department of Pediatrics, University of Tennessee Health Science Center, Memphis, TN 38103, United States.
| | - Darong Yang
- Children's Foundation Research Institute at Le Bonheur Children's Hospital, Department of Pediatrics, University of Tennessee Health Science Center, Memphis, TN 38103, United States.
| | - Guo-Yun Chen
- Children's Foundation Research Institute at Le Bonheur Children's Hospital, Department of Pediatrics, University of Tennessee Health Science Center, Memphis, TN 38103, United States.
| |
Collapse
|
33
|
Malik A, Brudvig JM, Gadsden BJ, Ethridge AD, Mansfield LS. Campylobacter jejuni induces autoimmune peripheral neuropathy via Sialoadhesin and Interleukin-4 axes. Gut Microbes 2022; 14:2064706. [PMID: 35442154 PMCID: PMC9037470 DOI: 10.1080/19490976.2022.2064706] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Accepted: 03/29/2022] [Indexed: 02/04/2023] Open
Abstract
Campylobacter jejuni is a leading cause of gastroenteritis that has been causally linked with development of the autoimmune peripheral neuropathy Guillain Barré Syndrome (GBS). Previously, we showed that C. jejuni isolates from human enteritis patients induced Type1/17-cytokine dependent colitis in interleukin-10 (IL-10)-/- mice, while isolates from GBS patients colonized these mice without colitis but instead induced autoantibodies that cross-reacted with the sialylated oligosaccharide motifs on the LOS of GBS-associated C. jejuni and the peripheral nerve gangliosides. We show here that infection of IL-10-/- mice with the GBS but not the colitis isolate led to sciatic nerve inflammation and abnormal gait and hind limb movements, with character and timing consistent with this syndrome in humans. Autoantibody responses and associated nerve histologic changes were dependent on IL-4 production by CD4 T cells. We further show that Siglec-1 served as a central antigen presenting cell receptor mediating the uptake of the GBS isolates via interaction with the sialylated oligosaccharide motifs found specifically on the LOS of GBS-associated C. jejuni, and the ensuing T cell differentiation and autoantibody elicitation. Sialylated oligosaccharide motifs on the LOS of GBS-associated C. jejuni therefore acted as both the Siglec-1-ligand for phagocytosis, as well as the epitope for autoimmunity. Overall, we present a mouse model of an autoimmune disease induced directly by a bacterium that is dependent upon Siglec-1 and IL-4. We also demonstrate the negative regulatory role of IL-10 in C. jejuni induced autoimmunity and provide IL-4 and Siglec-1 blockade as potential therapeutic interventions against GBS.
Collapse
Affiliation(s)
- Ankit Malik
- Department of Large Animal Clinical Sciences, Michigan State University, East Lansing, MI, USA
- Department of Microbiology and Molecular Genetics, Michigan State University, East Lansing, MI, USA
| | - Jean M. Brudvig
- Department of Large Animal Clinical Sciences, Michigan State University, East Lansing, MI, USA
- Comparative Medicine and Integrative Biology Program, Michigan State University, East Lansing, MIUSA
| | - Barbie J. Gadsden
- Department of Large Animal Clinical Sciences, Michigan State University, East Lansing, MI, USA
- Comparative Medicine and Integrative Biology Program, Michigan State University, East Lansing, MIUSA
| | - Alexander D. Ethridge
- Department of Large Animal Clinical Sciences, Michigan State University, East Lansing, MI, USA
- Department of Microbiology and Molecular Genetics, Michigan State University, East Lansing, MI, USA
| | - Linda S. Mansfield
- Department of Large Animal Clinical Sciences, Michigan State University, East Lansing, MI, USA
- Department of Microbiology and Molecular Genetics, Michigan State University, East Lansing, MI, USA
| |
Collapse
|
34
|
O-Acetylation of Capsular Polysialic Acid Enables Escherichia coli K1 Escaping from Siglec-Mediated Innate Immunity and Lysosomal Degradation of E. coli-Containing Vacuoles in Macrophage-Like Cells. Microbiol Spectr 2021; 9:e0039921. [PMID: 34878295 PMCID: PMC8653822 DOI: 10.1128/spectrum.00399-21] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Escherichia coli K1 causes bacteremia and meningitis in human neonates. The K1 capsule, an α2,8-linked polysialic acid (PSA) homopolymer, is its essential virulence factor. PSA is usually partially modified by O-acetyl groups. It is known that O-acetylation alters the antigenicity of PSA, but its impact on the interactions between E. coli K1 and host cells is unclear. In this study, a phase variant was obtained by passage of E. coli K1 parent strain, which expressed a capsule with 44% O-acetylation whereas the capsule of the parent strain has only 3%. The variant strain showed significantly reduced adherence and invasion to macrophage-like cells in comparison to the parent strain. Furthermore, we found that O-acetylation of PSA enhanced the modulation of trafficking of E. coli-containing vacuoles (ECV), enabling them to avoid fusing with lysosomes in these cells. Intriguingly, by using quartz crystal microbalance, we demonstrated that the PSA purified from the parent strain interacted with human sialic acid-binding immunoglobulin-like lectins (Siglecs), including Siglec-5, Siglec-7, Siglec-11, and Siglec-14. However, O-acetylated PSA from the variant interacted much less and also suppressed the production of Siglec-mediated proinflammatory cytokines. The adherence of the parent strain to human macrophage-like cells was significantly blocked by monoclonal antibodies against Siglec-11 and Siglec-14. Furthermore, the variant strain caused increased bacteremia and higher lethality in neonatal mice compared to the parent strain. These data elucidate that O-acetylation of K1 capsule enables E. coli to escape from Siglec-mediated innate immunity and lysosomal degradation; therefore, it is a strategy used by E. coli K1 to regulate its virulence. IMPORTANCEEscherichia coli K1 is a leading cause of neonatal meningitis. The mortality and morbidity of this disease remain significantly high despite antibiotic therapy. One major limitation on advances in prevention and therapy for meningitis is an incomplete understanding of its pathogenesis. E. coli K1 is surrounded by PSA, which is observed to have high-frequency variation of O-acetyl modification. Here, we present an in-depth study of the function of O-acetylation in PSA at each stage of host-pathogen interaction. We found that a high level of O-acetylation significantly interfered with Siglec-mediated bacterial adherence to macrophage-like cells, and blunted the proinflammatory response. Furthermore, the O-acetylation of PSA modulated the trafficking of ECVs to prevent them from fusing with lysosomes, enabling them to escape degradation by lysozymes within these cells. Elucidating how subtle modification of the capsule enhances bacterial defenses against host innate immunity will enable the future development of effective drugs or vaccines against infection by E. coli K1.
Collapse
|
35
|
Rana R, Rani S, Kumar V, Nakhate KT, Ajazuddin, Gupta U. Sialic Acid Conjugated Chitosan Nanoparticles: Modulation to Target Tumour Cells and Therapeutic Opportunities. AAPS PharmSciTech 2021; 23:10. [PMID: 34862568 DOI: 10.1208/s12249-021-02170-z] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2021] [Accepted: 10/22/2021] [Indexed: 12/24/2022] Open
Abstract
Targeted delivery of therapeutics forestalls the dreadful delocalized effects, drug toxicities and needless immunosuppression. Cancer cells are bounteous with sialic acid and the differential expression of glycosyl transferase, glycosidase and monosaccharide transporter compared to healthy tissues. The current study entails the development and characterisation of sialic acid (SA)-labelled chitosan nanoparticles encapsulating gemcitabine (GEM). Chitosan (CS) was conjugated with SA using coupling reaction and characterised spectroscopically. Furthermore, different concentrations of chitosan and tripolyphosphate (TPP) were optimised to fabricate surface modified chitosan nanoparticles. SA conjugated chitosan nanoparticles encapsulating GEM (SA-CS_GEM NPs) of 232 ± 9.69 nm with narrow distribution (PDI < 0.5) and zeta potential of - 19 ± 0.97 mV was fabricated. GEM was successfully loaded in the SA-CS NPs, depicting prolonged and biphasic drug release pattern more elated at low pH. Pronounced cellular uptake (FITC tagged) and cytotoxicity (IC50 487.4 nM) was observed in SA-CS_GEM NPs against A549 cells. IC50 for SA-CS_GEM NPs plunged with an increase in the time points from 24 to 72 h. Concentration-dependent haemolytic study confirmed significant haemocompatibility of SA-CS_GEM NPs. Pharmacokinetic study was performed on Sprague-Dawley rats and the kinetic parameters were calculated using PKSolver 2.0. Results demonstrated a consequential refinement of 2.98 times in modified SA-CS_GEM NPs with a significant increase in retention time, bioavailability and elimination half-life, and decrease in elimination rate constant and volume of distribution in comparison to CS_GEM NPs. Therefore, SA-CS shell core nanoparticles could be a beneficial approach to target and treat NSCLC (non-small cell lung cancer) and direct for research possibilities to target the other tumour cells.
Collapse
|
36
|
He F, Wang N, Li J, He L, Yang Z, Lu J, Xiong G, Yu C, Wang S. High affinity monoclonal antibody targeting Siglec-15 for cancer immunotherapy. J Clin Transl Res 2021; 7:739-749. [PMID: 34988324 PMCID: PMC8710358] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2021] [Revised: 06/11/2021] [Accepted: 09/29/2021] [Indexed: 11/29/2022] Open
Abstract
BACKGROUND AND AIM Recently, Siglec-15 has been proved as a novel immune suppressor and a potential target for normalization cancer immunotherapy, which is non-redundant to the well-known PD-L1/PD-1 pathway. Herein, anti-Siglec-15 mAb, a monoclonal antibody (mAb) with a high affinity against Siglec-15, was prepared. METHODS The engineered CHO-K1 Siglec-15 cell line was constructed to heterologously expressed Siglec-15 for the affinity test with the mAb. Antigens Siglec-15-mIgG and Siglec-15-his were recombinantly expressed by 293F cells and purified by high-performance liquid chromatography (HPLC). Hybridoma cell line against Siglec-15 was prepared and validated by enzyme-linked immunoabsorbant assay (ELISA) and fluorescent-activated cell sorting (FACS). Finally, the anti-Siglec-15 mAb was produced, purified, and confirmed by SDS-PAGE, ELISA, and FACS. RESULTS The EC50 of the anti-Siglec-15 mAb with Siglec-15 is 76.65 ng/mL, lower than that of the positive control 5G12 (90.7 ng/mL), indicating a high affinity of the anti-Siglec-15 mAb. In vitro and in vivo studies verified that the anti-Siglec-15 mAb blocks the Siglec-15-mediated suppression of T cell and moderately prevents the tumor growth. CONCLUSIONS The anti-Siglec-15 mAb can be considered as an effective immunotherapy for tumor suppression. RELEVANCE FOR PATIENTS The anti-Siglec-15 mAb prepared in this study is useful as an immune checkpoint inhibitor against Siglec-15 for normalization cancer immunotherapy. This immunotherapy provides an alternative treatment for cancer patients who are refractory to the well-known PD-L1/PD-1-targeting therapies.
Collapse
Affiliation(s)
- Fei He
- 1College of Life Science and Technology, Beijing University of Chemical Technology, Beijing 100029, China
| | - Na Wang
- 1College of Life Science and Technology, Beijing University of Chemical Technology, Beijing 100029, China
| | - Jiangwei Li
- 1College of Life Science and Technology, Beijing University of Chemical Technology, Beijing 100029, China
| | - Luanying He
- 1College of Life Science and Technology, Beijing University of Chemical Technology, Beijing 100029, China
| | - Zhao Yang
- 1College of Life Science and Technology, Beijing University of Chemical Technology, Beijing 100029, China,2College of Life Science, Key Laboratory of Protection and Utilization of Biological Resources in Tarim Basin of Xinjiang Production and Construction Corps, Tarim University, Alar 843300, Xinjiang, China
| | - Jiandong Lu
- 3Shenzhen Traditional Chinese Medicine Hospital, Guangzhou University of Chinese Medicine, Shenzhen, 518033, Guangdong, China
| | - Guoliang Xiong
- 3Shenzhen Traditional Chinese Medicine Hospital, Guangzhou University of Chinese Medicine, Shenzhen, 518033, Guangdong, China
| | - Changyuan Yu
- 1College of Life Science and Technology, Beijing University of Chemical Technology, Beijing 100029, China,Corresponding authors: Changyuan Yu and Shihui Wang College of Life Science and Technology, Beijing University of Chemical Technology, Beijing 100029, China Tel: +86-10-64421335 Fax: +86-10-64421335 E-mail: ;
| | - Shihui Wang
- 1College of Life Science and Technology, Beijing University of Chemical Technology, Beijing 100029, China,Corresponding authors: Changyuan Yu and Shihui Wang College of Life Science and Technology, Beijing University of Chemical Technology, Beijing 100029, China Tel: +86-10-64421335 Fax: +86-10-64421335 E-mail: ;
| |
Collapse
|
37
|
Visser EA, Moons SJ, Timmermans SBPE, de Jong H, Boltje TJ, Büll C. Sialic acid O-acetylation: From biosynthesis to roles in health and disease. J Biol Chem 2021; 297:100906. [PMID: 34157283 PMCID: PMC8319020 DOI: 10.1016/j.jbc.2021.100906] [Citation(s) in RCA: 41] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2021] [Revised: 06/16/2021] [Accepted: 06/18/2021] [Indexed: 02/06/2023] Open
Abstract
Sialic acids are nine-carbon sugars that frequently cap glycans at the cell surface in cells of vertebrates as well as cells of certain types of invertebrates and bacteria. The nine-carbon backbone of sialic acids can undergo extensive enzymatic modification in nature and O-acetylation at the C-4/7/8/9 position in particular is widely observed. In recent years, the detection and analysis of O-acetylated sialic acids have advanced, and sialic acid-specific O-acetyltransferases (SOATs) and O-acetylesterases (SIAEs) that add and remove O-acetyl groups, respectively, have been identified and characterized in mammalian cells, invertebrates, bacteria, and viruses. These advances now allow us to draw a more complete picture of the biosynthetic pathway of the diverse O-acetylated sialic acids to drive the generation of genetically and biochemically engineered model cell lines and organisms with altered expression of O-acetylated sialic acids for dissection of their roles in glycoprotein stability, development, and immune recognition, as well as discovery of novel functions. Furthermore, a growing number of studies associate sialic acid O-acetylation with cancer, autoimmunity, and infection, providing rationale for the development of selective probes and inhibitors of SOATs and SIAEs. Here, we discuss the current insights into the biosynthesis and biological functions of O-acetylated sialic acids and review the evidence linking this modification to disease. Furthermore, we discuss emerging strategies for the design, synthesis, and potential application of unnatural O-acetylated sialic acids and inhibitors of SOATs and SIAEs that may enable therapeutic targeting of this versatile sialic acid modification.
Collapse
Affiliation(s)
- Eline A Visser
- Institute for Molecules and Materials, Department of Synthetic Organic Chemistry, Radboud University Nijmegen, Nijmegen, the Netherlands
| | - Sam J Moons
- Institute for Molecules and Materials, Department of Synthetic Organic Chemistry, Radboud University Nijmegen, Nijmegen, the Netherlands
| | - Suzanne B P E Timmermans
- Institute for Molecules and Materials, Department of Synthetic Organic Chemistry, Radboud University Nijmegen, Nijmegen, the Netherlands
| | - Heleen de Jong
- Institute for Molecules and Materials, Department of Synthetic Organic Chemistry, Radboud University Nijmegen, Nijmegen, the Netherlands
| | - Thomas J Boltje
- Institute for Molecules and Materials, Department of Synthetic Organic Chemistry, Radboud University Nijmegen, Nijmegen, the Netherlands.
| | - Christian Büll
- Copenhagen Center for Glycomics, Departments of Cellular and Molecular Medicine, Faculty of Health Sciences, University of Copenhagen, Copenhagen, Denmark; Hubrecht Institute, Utrecht, the Netherlands.
| |
Collapse
|
38
|
Dewi IM, Cunha C, Jaeger M, Gresnigt MS, Gkountzinopoulou ME, Garishah FM, Duarte-Oliveira C, Campos CF, Vanderbeke L, Sharpe AR, Brüggemann RJ, Verweij PE, Lagrou K, Vande Velde G, de Mast Q, Joosten LA, Netea MG, van der Ven AJ, Wauters J, Carvalho A, van de Veerdonk FL. Neuraminidase and SIGLEC15 modulate the host defense against pulmonary aspergillosis. Cell Rep Med 2021; 2:100289. [PMID: 34095887 PMCID: PMC8149467 DOI: 10.1016/j.xcrm.2021.100289] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2019] [Revised: 09/01/2020] [Accepted: 04/23/2021] [Indexed: 11/30/2022]
Abstract
Influenza-associated pulmonary aspergillosis (IAPA) has been reported increasingly since the advent of use of neuraminidase (NA) inhibitors following the 2009 influenza pandemic. We hypothesize that blocking host NA modulates the immune response against Aspergillus fumigatus. We demonstrate that NA influences the host response against A. fumigatus in vitro and that oseltamivir increases the susceptibility of mice to pulmonary aspergillosis. Oseltamivir impairs the mouse splenocyte and human peripheral blood mononuclear cell (PBMC) killing capacity of A. fumigatus, and adding NA restores this defect in PBMCs. Furthermore, the sialic acid-binding receptor SIGLEC15 is upregulated in PBMCs stimulated with A. fumigatus. Silencing of SIGLEC15 decrease PBMC killing of A. fumigatus. We provide evidence that host NA activity and sialic acid recognition are important for anti-Aspergillus defense. NA inhibitors might predispose individuals with severe influenza to invasive aspergillosis. These data shed light on the pathogenesis of invasive fungal infections and may identify potential therapeutic targets.
Collapse
Affiliation(s)
- Intan M.W. Dewi
- Department of Internal Medicine, Radboud University Medical Center, Nijmegen, the Netherlands
- Microbiology Division, Department of Biomedical Sciences, Faculty of Medicine, Universitas Padjadjaran, Bandung, Indonesia
| | - Cristina Cunha
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal
- ICVS/3B’s – PT Government Associate Laboratory, Guimarães/Braga, Portugal
| | - Martin Jaeger
- Department of Internal Medicine, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Mark S. Gresnigt
- Department of Microbial Pathogenicity Mechanisms, Leibniz Institute for Natural Product Research and Infection Biology-Hans Knoll Institute, Jena, Germany
| | | | - Fadel M. Garishah
- Department of Internal Medicine, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Cláudio Duarte-Oliveira
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal
- ICVS/3B’s – PT Government Associate Laboratory, Guimarães/Braga, Portugal
| | - Cláudia F. Campos
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal
- ICVS/3B’s – PT Government Associate Laboratory, Guimarães/Braga, Portugal
| | - Lore Vanderbeke
- Department of Microbiology and Immunology, KU Leuven, Leuven, Belgium
| | | | - Roger J. Brüggemann
- Department of Pharmacy, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Paul E. Verweij
- Department of Medical Microbiology, Radboud University Medical Centre, Nijmegen, the Netherlands
| | - Katrien Lagrou
- Department of Microbiology and Immunology, KU Leuven, Leuven, Belgium
| | - Greetje Vande Velde
- Biomedical MRI/Molecular Small Animal Imaging Center, Department of Imaging and Pathology, KU Leuven, Belgium
| | - Quirijn de Mast
- Department of Internal Medicine, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Leo A.B. Joosten
- Department of Internal Medicine, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Mihai G. Netea
- Department of Internal Medicine, Radboud University Medical Center, Nijmegen, the Netherlands
| | | | - Joost Wauters
- Department of Microbiology and Immunology, KU Leuven, Leuven, Belgium
| | - Agostinho Carvalho
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal
- ICVS/3B’s – PT Government Associate Laboratory, Guimarães/Braga, Portugal
| | | |
Collapse
|
39
|
Han Z, Thuy-Boun PS, Pfeiffer W, Vartabedian VF, Torkamani A, Teijaro JR, Wolan DW. Identification of an N-acetylneuraminic acid-presenting bacteria isolated from a human microbiome. Sci Rep 2021; 11:4763. [PMID: 33637779 PMCID: PMC7910532 DOI: 10.1038/s41598-021-83875-w] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2020] [Accepted: 02/01/2021] [Indexed: 01/31/2023] Open
Abstract
N-Acetylneuraminic acid is the most abundant sialic acid (SA) in humans and is expressed as the terminal sugar on intestinal mucus glycans. Several pathogenic bacteria harvest and display host SA on their own surfaces to evade Siglec-mediated host immunity. While previous studies have identified bacterial enzymes associated with SA catabolism, no reported methods permit the selective labeling, tracking, and quantitation of SA-presenting microbes within complex multi-microbial systems. We combined metabolic labeling, click chemistry, 16S rRNA gene, and whole-genome sequencing to track and identify SA-presenting microbes from a cultured human fecal microbiome. We isolated a new strain of Escherichia coli that incorporates SA onto its own surface and encodes for the nanT, neuA, and neuS genes necessary for harvesting and presenting SA. Our method is applicable to the identification of SA-presenting bacteria from human, animal, and environmental microbiomes, as well as providing an entry point for the investigation of surface-expressed SA-associated structures.
Collapse
Affiliation(s)
- Zhen Han
- grid.214007.00000000122199231Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA 92037 USA
| | - Peter S. Thuy-Boun
- grid.214007.00000000122199231Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA 92037 USA
| | - Wayne Pfeiffer
- grid.266100.30000 0001 2107 4242San Diego Supercomputer Center, University of California, San Diego, La Jolla, CA 92093 USA
| | - Vincent F. Vartabedian
- grid.214007.00000000122199231Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA 92037 USA
| | - Ali Torkamani
- grid.214007.00000000122199231Scripps Research Translational Institute, The Scripps Research Institute, La Jolla, CA 92037 USA ,grid.214007.00000000122199231Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA 92037 USA
| | - John R. Teijaro
- grid.214007.00000000122199231Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA 92037 USA
| | - Dennis W. Wolan
- grid.214007.00000000122199231Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA 92037 USA ,grid.214007.00000000122199231Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA 92037 USA
| |
Collapse
|
40
|
DeRogatis JM, Viramontes KM, Neubert EN, Tinoco R. PSGL-1 Immune Checkpoint Inhibition for CD4 + T Cell Cancer Immunotherapy. Front Immunol 2021; 12:636238. [PMID: 33708224 PMCID: PMC7940186 DOI: 10.3389/fimmu.2021.636238] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2020] [Accepted: 01/04/2021] [Indexed: 01/15/2023] Open
Abstract
Immune checkpoint inhibition targeting T cells has shown tremendous promise in the treatment of many cancer types and are now standard therapies for patients. While standard therapies have focused on PD-1 and CTLA-4 blockade, additional immune checkpoints have shown promise in promoting anti-tumor immunity. PSGL-1, primarily known for its role in cellular migration, has also been shown to function as a negative regulator of CD4+ T cells in numerous disease settings including cancer. PSGL-1 is highly expressed on T cells and can engage numerous ligands that impact signaling pathways, which may modulate CD4+ T cell differentiation and function. PSGL-1 engagement in the tumor microenvironment may promote CD4+ T cell exhaustion pathways that favor tumor growth. Here we highlight that blocking the PSGL-1 pathway on CD4+ T cells may represent a new cancer therapy approach to eradicate tumors.
Collapse
Affiliation(s)
| | | | | | - Roberto Tinoco
- Department of Molecular Biology and Biochemistry, University of California, Irvine, Irvine, CA, United States
| |
Collapse
|
41
|
Sun J, Lu Q, Sanmamed MF, Wang J. Siglec-15 as an Emerging Target for Next-generation Cancer Immunotherapy. Clin Cancer Res 2021; 27:680-688. [PMID: 32958700 PMCID: PMC9942711 DOI: 10.1158/1078-0432.ccr-19-2925] [Citation(s) in RCA: 80] [Impact Index Per Article: 26.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2020] [Revised: 08/07/2020] [Accepted: 09/16/2020] [Indexed: 01/21/2023]
Abstract
Immunomodulatory agents blocking the PD-1/PD-L1 pathway have shown a new way to treat cancer. The explanation underlying the success of these agents may be the selective expression of PD-L1 with dominant immune-suppressive activities in the tumor microenvironment (TME), supporting a more favorable tumor response-to-toxicity ratio. However, despite the big success of these drugs, most patients with cancer show primary or acquired resistance, calling for the identification of new immune modulators in the TME. Using a genome-scale T-cell activity array in combination with bioinformatic analysis of human cancer databases, we identified Siglec-15 as a critical immune suppressor with broad upregulation on various cancer types and a potential target for cancer immunotherapy. Siglec-15 has unique molecular features compared with many other known checkpoint inhibitory ligands. It shows prominent expression on macrophages and cancer cells and a mutually exclusive expression with PD-L1, suggesting that it may be a critical immune evasion mechanism in PD-L1-negative patients. Interestingly, Siglec-15 has also been identified as a key regulator for osteoclast differentiation and may have potential implications in bone disorders not limited to osteoporosis. Here, we provide an overview of Siglec-15 biology, its role in cancer immune regulation, the preliminary and encouraging clinical data related to the first-in-class Siglec-15 targeting mAb, as well as many unsolved questions in this pathway. As a new player in the cancer immunotherapeutic arena, Siglec-15 may represent a novel class of immune inhibitors with tumor-associated expression and divergent mechanisms of action to PD-L1, with potential implications in anti-PD-1/PD-L1-resistant patients.
Collapse
Affiliation(s)
- Jingwei Sun
- Department of Immunobiology, Yale University School of Medicine, New Haven, Connecticut
| | - Qiao Lu
- Department of Pathology, New York University Grossman School of Medicine, New York, New York
- The Laura and Isaac Perlmutter Cancer Center, New York University Langone Health, New York, New York
| | - Miguel F Sanmamed
- Program of Immunology and Immunotherapy, CIMA, University of Navarra, Pamplona, Spain
| | - Jun Wang
- Department of Pathology, New York University Grossman School of Medicine, New York, New York.
- The Laura and Isaac Perlmutter Cancer Center, New York University Langone Health, New York, New York
| |
Collapse
|
42
|
Functions and therapeutic targets of Siglec-mediated infections, inflammations and cancers. J Formos Med Assoc 2021; 120:5-24. [DOI: 10.1016/j.jfma.2019.10.019] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2019] [Revised: 06/11/2019] [Accepted: 10/28/2019] [Indexed: 12/20/2022] Open
|
43
|
Wu Y, Yang D, Liu R, Wang L, Chen GY. Selective Response to Bacterial Infection by Regulating Siglec-E Expression. iScience 2020; 23:101473. [PMID: 32889432 PMCID: PMC7479279 DOI: 10.1016/j.isci.2020.101473] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2020] [Revised: 07/10/2020] [Accepted: 08/11/2020] [Indexed: 12/24/2022] Open
Abstract
Interactions between microbes and hosts can be a benign, deleterious, or even fatal, resulting in death of the host, the microbe, or both. Sialic acid-binding immunoglobulin-like lectins (Siglecs) suppress infection responses to sialylated pathogens. However, most pathogens are nonsialylated. Here we determined Siglecs respond to nonsialylated Gram-negative bacteria (Escherichia coli 25922 and DH5α) and Gram-positive bacteria (Staphylococcus aureus and Listeria monocytogenes). We found that Siglece-/- mice had higher mortality than wild-type mice following Gram-negative but not Gram-positive bacterial infection. Better survival in wild-type mice depended on more efficient clearance of Gram-negative than Gram-positive bacteria. Gram-negative bacteria upregulated Siglec-E, thus increasing reactive oxygen species (ROS); Tyr432 in the ITIM domain of Siglec-E was required to increase ROS. Moreover, Gram-negative bacteria upregulated Siglec-E via TLR4/MyD88/JNK/NF-κB/AP-1, whereas Gram-positive bacteria downregulated Siglec-E via TLR2/RANKL/TRAF6/Syk. Thus, our study describes a fundamentally new role for Siglec-E during infection.
Collapse
Affiliation(s)
- Yin Wu
- Children's Foundation Research Institute at Le Bonheur Children's Hospital, Department of Pediatrics, University of Tennessee Health Science Center, Memphis, TN 38103, USA
| | - Darong Yang
- Children's Foundation Research Institute at Le Bonheur Children's Hospital, Department of Pediatrics, University of Tennessee Health Science Center, Memphis, TN 38103, USA
| | - Runhua Liu
- Department of Genetics, University of Alabama at Birmingham, Birmingham, AL, USA
- Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Lizhong Wang
- Department of Genetics, University of Alabama at Birmingham, Birmingham, AL, USA
- Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Guo-Yun Chen
- Children's Foundation Research Institute at Le Bonheur Children's Hospital, Department of Pediatrics, University of Tennessee Health Science Center, Memphis, TN 38103, USA
| |
Collapse
|
44
|
Furumaya C, Martinez-Sanz P, Bouti P, Kuijpers TW, Matlung HL. Plasticity in Pro- and Anti-tumor Activity of Neutrophils: Shifting the Balance. Front Immunol 2020; 11:2100. [PMID: 32983165 PMCID: PMC7492657 DOI: 10.3389/fimmu.2020.02100] [Citation(s) in RCA: 61] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2020] [Accepted: 08/03/2020] [Indexed: 12/11/2022] Open
Abstract
Over the last decades, cancer immunotherapies such as checkpoint blockade and adoptive T cell transfer have been a game changer in many aspects and have improved the treatment for various malignancies considerably. Despite the clinical success of harnessing the adaptive immunity to combat the tumor, the benefits of immunotherapy are still limited to a subset of patients and cancer types. In recent years, neutrophils, the most abundant circulating leukocytes, have emerged as promising targets for anti-cancer therapies. Traditionally regarded as the first line of defense against infections, neutrophils are increasingly recognized as critical players during cancer progression. Evidence shows the functional plasticity of neutrophils in the tumor microenvironment, allowing neutrophils to exert either pro-tumor or anti-tumor effects. This review describes the tumor-promoting roles of neutrophils, focusing on their myeloid-derived suppressor cell activity, as well as their role in tumor elimination, exerted mainly via antibody-dependent cellular cytotoxicity. We will discuss potential approaches to therapeutically target neutrophils in cancer. These include strategies in humans to either silence the pro-tumor activity of neutrophils, or to activate or enhance their anti-tumor functions. Redirecting neutrophils seems a promising approach to harness innate immunity to improve treatment for cancer patients.
Collapse
Affiliation(s)
- Charita Furumaya
- Department of Blood Cell Research, Sanquin Research, Amsterdam University Medical Center, University of Amsterdam, Amsterdam, Netherlands
| | - Paula Martinez-Sanz
- Department of Blood Cell Research, Sanquin Research, Amsterdam University Medical Center, University of Amsterdam, Amsterdam, Netherlands
| | - Panagiota Bouti
- Department of Blood Cell Research, Sanquin Research, Amsterdam University Medical Center, University of Amsterdam, Amsterdam, Netherlands
| | - Taco W Kuijpers
- Department of Blood Cell Research, Sanquin Research, Amsterdam University Medical Center, University of Amsterdam, Amsterdam, Netherlands.,Department of Pediatric Immunology, Rheumatology and Infectious Diseases, Emma Children's Hospital, Amsterdam University Medical Center, University of Amsterdam, Amsterdam, Netherlands
| | - Hanke L Matlung
- Department of Blood Cell Research, Sanquin Research, Amsterdam University Medical Center, University of Amsterdam, Amsterdam, Netherlands
| |
Collapse
|
45
|
Wielgat P, Rogowski K, Godlewska K, Car H. Coronaviruses: Is Sialic Acid a Gate to the Eye of Cytokine Storm? From the Entry to the Effects. Cells 2020; 9:E1963. [PMID: 32854433 PMCID: PMC7564400 DOI: 10.3390/cells9091963] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2020] [Accepted: 08/24/2020] [Indexed: 12/16/2022] Open
Abstract
Coronaviruses (CoVs) are a diverse family of the enveloped human and animal viruses reported as causative agents for respiratory and intestinal infections. The high pathogenic potential of human CoVs, including SARS-CoV, MERS-CoV and SARS-CoV-2, is closely related to the invasion mechanisms underlying the attachment and entry of viral particles to the host cells. There is increasing evidence that sialylated compounds of cellular glycocalyx can serve as an important factor in the mechanism of CoVs infection. Additionally, the sialic acid-mediated cross-reactivity with the host immune lectins is known to exert the immune response of different intensity in selected pathological stages. Here, we focus on the last findings in the field of glycobiology in the context of the role of sialic acid in tissue tropism, viral entry kinetics and immune regulation in the CoVs infections.
Collapse
Affiliation(s)
- Przemyslaw Wielgat
- Department of Clinical Pharmacology, Medical University of Bialystok, Waszyngtona 15A, 15274 Bialystok, Poland;
| | - Karol Rogowski
- Department of Experimental Pharmacology, Medical University of Bialystok, Szpitalna 37, 15295 Bialystok, Poland;
| | - Katarzyna Godlewska
- Department of Haematology, Medical University of Bialystok, M. Sklodowskiej-Curie 24A, 15276 Bialystok, Poland;
| | - Halina Car
- Department of Clinical Pharmacology, Medical University of Bialystok, Waszyngtona 15A, 15274 Bialystok, Poland;
- Department of Experimental Pharmacology, Medical University of Bialystok, Szpitalna 37, 15295 Bialystok, Poland;
| |
Collapse
|
46
|
Cipollo JF, Parsons LM. Glycomics and glycoproteomics of viruses: Mass spectrometry applications and insights toward structure-function relationships. MASS SPECTROMETRY REVIEWS 2020; 39:371-409. [PMID: 32350911 PMCID: PMC7318305 DOI: 10.1002/mas.21629] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/15/2019] [Revised: 04/01/2020] [Accepted: 04/05/2020] [Indexed: 05/21/2023]
Abstract
The advancement of viral glycomics has paralleled that of the mass spectrometry glycomics toolbox. In some regard the glycoproteins studied have provided the impetus for this advancement. Viral proteins are often highly glycosylated, especially those targeted by the host immune system. Glycosylation tends to be dynamic over time as viruses propagate in host populations leading to increased number of and/or "movement" of glycosylation sites in response to the immune system and other pressures. This relationship can lead to highly glycosylated, difficult to analyze glycoproteins that challenge the capabilities of modern mass spectrometry. In this review, we briefly discuss five general areas where glycosylation is important in the viral niche and how mass spectrometry has been used to reveal key information regarding structure-function relationships between viral glycoproteins and host cells. We describe the recent past and current glycomics toolbox used in these analyses and give examples of how the requirement to analyze these complex glycoproteins has provided the incentive for some advances seen in glycomics mass spectrometry. A general overview of viral glycomics, special cases, mass spectrometry methods and work-flows, informatics and complementary chemical techniques currently used are discussed. © 2020 The Authors. Mass Spectrometry Reviews published by John Wiley & Sons Ltd. Mass Spec Rev.
Collapse
Affiliation(s)
- John F. Cipollo
- Center for Biologics Evaluation and Research, Food and Drug AdministrationSilver SpringMaryland
| | - Lisa M. Parsons
- Center for Biologics Evaluation and Research, Food and Drug AdministrationSilver SpringMaryland
| |
Collapse
|
47
|
Kong L, Wu L, Guo Z, Mu L, Yang Y, Bian X, Li B, Pan X, Fu S, Ye J. A Siglec-1-like lectin from Nile tilapia (Oreochromis niloticus) possesses functions of agglutination and mediation of macrophage phagocytic activity. FISH & SHELLFISH IMMUNOLOGY 2020; 102:203-210. [PMID: 32330627 DOI: 10.1016/j.fsi.2020.04.034] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/07/2019] [Revised: 04/14/2020] [Accepted: 04/16/2020] [Indexed: 06/11/2023]
Abstract
Siglec-1, one of the sialic acid-binding immunoglobulin-type lectins, is closely related to the recognition of host-pathogen and cell-cell interactions in the adaptive and innate immune systems. In this communication, a Siglec-1-like gene (OnSiglec-1-like) from Nile tilapia (Oreochromis niloticus) was analyzed. Relative expression revealed that the OnSiglec-1-like was expressed in all tested tissues, and the highest expression was found in the anterior kidney. Upon Streptococcus agalactiae (S. agalactiae) infection, the expression of OnSiglec-1-like was up-regulated in anterior kidney and spleen significantly in vivo. Additionally, the same phenomenon was observed in anterior kidney leukocytes upon LPS and S. agalactiae challenges as well in vitro. Western-blotting and ELISA analyses revealed that recombinant OnSiglec-1-like protein possessed high binding activity to LTA, LPS and S. agalactiae. Further, the recombinant OnSiglec-1-like was able to agglutinate S. agalactiae. Moreover, with the digestion of specific sialidase, the phagocytic ability of macrophages to S. agalactiae was greatly enhanced. Taken together, these results indicated that the Siglec-1-like possesses conserved functions of agglutination and promotion of macrophage phagocytic activity in Nile tilapia.
Collapse
Affiliation(s)
- Linghe Kong
- Institute of Modern Aquaculture Science and Engineering, School of Life Sciences, South China Normal University, Guangdong Provincial Key Laboratory for Healthy and Safe Aquaculture, Guangdong, 510631, PR China
| | - Liting Wu
- Institute of Modern Aquaculture Science and Engineering, School of Life Sciences, South China Normal University, Guangdong Provincial Key Laboratory for Healthy and Safe Aquaculture, Guangdong, 510631, PR China
| | - Zheng Guo
- Institute of Modern Aquaculture Science and Engineering, School of Life Sciences, South China Normal University, Guangdong Provincial Key Laboratory for Healthy and Safe Aquaculture, Guangdong, 510631, PR China.
| | - Liangliang Mu
- Institute of Modern Aquaculture Science and Engineering, School of Life Sciences, South China Normal University, Guangdong Provincial Key Laboratory for Healthy and Safe Aquaculture, Guangdong, 510631, PR China
| | - Yanjian Yang
- Institute of Modern Aquaculture Science and Engineering, School of Life Sciences, South China Normal University, Guangdong Provincial Key Laboratory for Healthy and Safe Aquaculture, Guangdong, 510631, PR China
| | - Xia Bian
- Institute of Modern Aquaculture Science and Engineering, School of Life Sciences, South China Normal University, Guangdong Provincial Key Laboratory for Healthy and Safe Aquaculture, Guangdong, 510631, PR China
| | - Bingxi Li
- Institute of Modern Aquaculture Science and Engineering, School of Life Sciences, South China Normal University, Guangdong Provincial Key Laboratory for Healthy and Safe Aquaculture, Guangdong, 510631, PR China
| | - Xunbin Pan
- Institute of Modern Aquaculture Science and Engineering, School of Life Sciences, South China Normal University, Guangdong Provincial Key Laboratory for Healthy and Safe Aquaculture, Guangdong, 510631, PR China
| | - Shengli Fu
- Institute of Modern Aquaculture Science and Engineering, School of Life Sciences, South China Normal University, Guangdong Provincial Key Laboratory for Healthy and Safe Aquaculture, Guangdong, 510631, PR China
| | - Jianmin Ye
- Institute of Modern Aquaculture Science and Engineering, School of Life Sciences, South China Normal University, Guangdong Provincial Key Laboratory for Healthy and Safe Aquaculture, Guangdong, 510631, PR China.
| |
Collapse
|
48
|
Jaeger M, Pinelli M, Borghi M, Constantini C, Dindo M, van Emst L, Puccetti M, Pariano M, Ricaño-Ponce I, Büll C, Gresnigt MS, Wang X, Gutierrez Achury J, Jacobs CWM, Xu N, Oosting M, Arts P, Joosten LAB, van de Veerdonk FL, Veltman JA, Ten Oever J, Kullberg BJ, Feng M, Adema GJ, Wijmenga C, Kumar V, Sobel J, Gilissen C, Romani L, Netea MG. A systems genomics approach identifies SIGLEC15 as a susceptibility factor in recurrent vulvovaginal candidiasis. Sci Transl Med 2020; 11:11/496/eaar3558. [PMID: 31189718 DOI: 10.1126/scitranslmed.aar3558] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2017] [Revised: 08/13/2018] [Accepted: 05/14/2019] [Indexed: 12/30/2022]
Abstract
Candida vaginitis is a frequent clinical diagnosis with up to 8% of women experiencing recurrent vulvovaginal candidiasis (RVVC) globally. RVVC is characterized by at least three episodes per year. Most patients with RVVC lack known risk factors, suggesting a role for genetic risk factors in this condition. Through integration of genomic approaches and immunological studies in two independent cohorts of patients with RVVC and healthy individuals, we identified genes and cellular processes that contribute to the pathogenesis of RVVC, including cellular morphogenesis and metabolism, and cellular adhesion. We further identified SIGLEC15, a lectin expressed by various immune cells that binds sialic acid-containing structures, as a candidate gene involved in RVVC susceptibility. Candida stimulation induced SIGLEC15 expression in human peripheral blood mononuclear cells (PBMCs) and a polymorphism in the SIGLEC15 gene that was associated with RVVC in the patient cohorts led to an altered cytokine profile after PBMC stimulation. The same polymorphism led to an increase in IL1B and NLRP3 expression after Candida stimulation in HeLa cells in vitro. Last, Siglec15 expression was induced by Candida at the vaginal surface of mice, where in vivo silencing of Siglec15 led to an increase in the fungal burden. Siglec15 silencing was additionally accompanied by an increase in polymorphonuclear leukocytes during the course of infection. Identification of these pathways and cellular processes contributes to a better understanding of RVVC and may open new therapeutic avenues.
Collapse
Affiliation(s)
- M Jaeger
- Department of Internal Medicine and Radboud Centre for Infectious Diseases (RCI), Radboud University Medical Center, Nijmegen, 6525GA, Netherlands.,Radboud Institute for Molecular Life Sciences (RIMLS), Radboud University Nijmegen Medical Center (Radboudumc), Nijmegen, 6525GA, Netherlands
| | - M Pinelli
- Telethon Institute of Genetics and Medicine (TIGEM), Pozzuoli, Naples, 80078, Italy.,Department of Human Genetics, Donders Center for Neuroscience, Radboud University Medical Center, Nijmegen, 6525HR, Netherlands
| | - M Borghi
- Department of Experimental Medicine, University of Perugia, Polo Unico Sant'Andrea delle Fratte, Perugia, 06123, Italy
| | - C Constantini
- Department of Experimental Medicine, University of Perugia, Polo Unico Sant'Andrea delle Fratte, Perugia, 06123, Italy
| | - M Dindo
- Department of Experimental Medicine, University of Perugia, Polo Unico Sant'Andrea delle Fratte, Perugia, 06123, Italy
| | - L van Emst
- Department of Internal Medicine and Radboud Centre for Infectious Diseases (RCI), Radboud University Medical Center, Nijmegen, 6525GA, Netherlands
| | - M Puccetti
- Department of Experimental Medicine, University of Perugia, Polo Unico Sant'Andrea delle Fratte, Perugia, 06123, Italy
| | - M Pariano
- Department of Experimental Medicine, University of Perugia, Polo Unico Sant'Andrea delle Fratte, Perugia, 06123, Italy
| | - I Ricaño-Ponce
- University of Groningen, University Medical Centre Groningen, Department of Genetics, Groningen, 9713GZ, Netherlands
| | - C Büll
- Department of Radiation Oncology, Radiotherapy & OncoImmunology Laboratory, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Geert Grooteplein Zuid 32, Nijmegen, 6525GA, Netherlands
| | - M S Gresnigt
- Department of Internal Medicine and Radboud Centre for Infectious Diseases (RCI), Radboud University Medical Center, Nijmegen, 6525GA, Netherlands.,Radboud Institute for Molecular Life Sciences (RIMLS), Radboud University Nijmegen Medical Center (Radboudumc), Nijmegen, 6525GA, Netherlands.,Department of Microbial Pathogenicity Mechanisms, Leibniz Institute for Natural Product Research and Infection Biology-Hans Knöll Institute, Beutenbergstraße 11a, Jena, 07745, Germany
| | - X Wang
- Department of Internal Medicine and Radboud Centre for Infectious Diseases (RCI), Radboud University Medical Center, Nijmegen, 6525GA, Netherlands.,College of Computer, Qinghai Normal University, 810008 Xining, China
| | - J Gutierrez Achury
- University of Groningen, University Medical Centre Groningen, Department of Genetics, Groningen, 9713GZ, Netherlands
| | - C W M Jacobs
- Department of Internal Medicine and Radboud Centre for Infectious Diseases (RCI), Radboud University Medical Center, Nijmegen, 6525GA, Netherlands
| | - N Xu
- BGI-Shenzhen, Shenzhen 518083, China
| | - M Oosting
- Department of Internal Medicine and Radboud Centre for Infectious Diseases (RCI), Radboud University Medical Center, Nijmegen, 6525GA, Netherlands
| | - P Arts
- Department of Human Genetics, Donders Center for Neuroscience, Radboud University Medical Center, Nijmegen, 6525HR, Netherlands
| | - L A B Joosten
- Department of Internal Medicine and Radboud Centre for Infectious Diseases (RCI), Radboud University Medical Center, Nijmegen, 6525GA, Netherlands
| | - F L van de Veerdonk
- Department of Internal Medicine and Radboud Centre for Infectious Diseases (RCI), Radboud University Medical Center, Nijmegen, 6525GA, Netherlands
| | - J A Veltman
- Department of Human Genetics, Donders Center for Neuroscience, Radboud University Medical Center, Nijmegen, 6525HR, Netherlands.,Department of Clinical Genetics, Maastricht University Medical Centre, Maastricht, 6229HX, Netherlands
| | - J Ten Oever
- Department of Internal Medicine and Radboud Centre for Infectious Diseases (RCI), Radboud University Medical Center, Nijmegen, 6525GA, Netherlands
| | - B J Kullberg
- Department of Internal Medicine and Radboud Centre for Infectious Diseases (RCI), Radboud University Medical Center, Nijmegen, 6525GA, Netherlands
| | - M Feng
- BGI-Shenzhen, Shenzhen 518083, China
| | - G J Adema
- Department of Radiation Oncology, Radiotherapy & OncoImmunology Laboratory, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Geert Grooteplein Zuid 32, Nijmegen, 6525GA, Netherlands
| | - C Wijmenga
- University of Groningen, University Medical Centre Groningen, Department of Genetics, Groningen, 9713GZ, Netherlands
| | - V Kumar
- University of Groningen, University Medical Centre Groningen, Department of Genetics, Groningen, 9713GZ, Netherlands
| | - J Sobel
- Infectious Diseases, Wayne State University School of Medicine, Detroit, MI 48201, USA
| | - C Gilissen
- Department of Human Genetics, Donders Center for Neuroscience, Radboud University Medical Center, Nijmegen, 6525HR, Netherlands
| | - L Romani
- Department of Experimental Medicine, University of Perugia, Polo Unico Sant'Andrea delle Fratte, Perugia, 06123, Italy
| | - M G Netea
- Department of Internal Medicine and Radboud Centre for Infectious Diseases (RCI), Radboud University Medical Center, Nijmegen, 6525GA, Netherlands. .,Human Genomics Laboratory, Craiova University of Medicine and Pharmacy, Craiova, 200349
| |
Collapse
|
49
|
Wielgat P, Rogowski K, Niemirowicz-Laskowska K, Car H. Sialic Acid-Siglec Axis as Molecular Checkpoints Targeting of Immune System: Smart Players in Pathology and Conventional Therapy. Int J Mol Sci 2020; 21:ijms21124361. [PMID: 32575400 PMCID: PMC7352527 DOI: 10.3390/ijms21124361] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Revised: 06/17/2020] [Accepted: 06/17/2020] [Indexed: 12/11/2022] Open
Abstract
The sialic acid-based molecular mimicry in pathogens and malignant cells is a regulatory mechanism that leads to cross-reactivity with host antigens resulting in suppression and tolerance in the immune system. The interplay between sialoglycans and immunoregulatory Siglec receptors promotes foreign antigens hiding and immunosurveillance impairment. Therefore, molecular targeting of immune checkpoints, including sialic acid-Siglec axis, is a promising new field of inflammatory disorders and cancer therapy. However, the conventional drugs used in regular management can interfere with glycome machinery and exert a divergent effect on immune controlling systems. Here, we focus on the known effects of standard therapies on the sialoglycan-Siglec checkpoint and their importance in diagnosis, prediction, and clinical outcomes.
Collapse
Affiliation(s)
- Przemyslaw Wielgat
- Department of Clinical Pharmacology, Medical University of Bialystok, Waszyngtona 15A, 15-274 Bialystok, Poland;
- Correspondence: ; Tel.: +48-85-7450-647
| | - Karol Rogowski
- Department of Experimental Pharmacology, Medical University of Bialystok, Szpitalna 37, 15-295 Bialystok, Poland; (K.R.); (K.N.-L.)
| | - Katarzyna Niemirowicz-Laskowska
- Department of Experimental Pharmacology, Medical University of Bialystok, Szpitalna 37, 15-295 Bialystok, Poland; (K.R.); (K.N.-L.)
| | - Halina Car
- Department of Clinical Pharmacology, Medical University of Bialystok, Waszyngtona 15A, 15-274 Bialystok, Poland;
- Department of Experimental Pharmacology, Medical University of Bialystok, Szpitalna 37, 15-295 Bialystok, Poland; (K.R.); (K.N.-L.)
| |
Collapse
|
50
|
Van Bockstal L, Bulté D, Van den Kerkhof M, Dirkx L, Mabille D, Hendrickx S, Delputte P, Maes L, Caljon G. Interferon Alpha Favors Macrophage Infection by Visceral Leishmania Species Through Upregulation of Sialoadhesin Expression. Front Immunol 2020; 11:1113. [PMID: 32582193 PMCID: PMC7296180 DOI: 10.3389/fimmu.2020.01113] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2019] [Accepted: 05/07/2020] [Indexed: 12/24/2022] Open
Abstract
Type I interferons (IFNs) induced by an endogenous Leishmania RNA virus or exogenous viral infections have been shown to exacerbate infections with New World Cutaneous Leishmania parasites, however, the impact of type I IFNs in visceral Leishmania infections and implicated mechanisms remain to be unraveled. This study assessed the impact of type I IFN on macrophage infection with L. infantum and L. donovani and the implication of sialoadhesin (Siglec-1/CD169, Sn) as an IFN-inducible surface receptor. Stimulation of bone marrow-derived macrophages with type I IFN (IFN-α) significantly enhanced susceptibility to infection of reference laboratory strains and a set of recent clinical isolates. IFN-α particularly enhanced promastigote uptake. Enhanced macrophage susceptibility was linked to upregulated Sn surface expression as a major contributing factor to the infection exacerbating effect of IFN-α. Stimulation experiments in Sn-deficient macrophages, macrophage pretreatment with a monoclonal anti-Sn antibody or a novel bivalent anti-Sn nanobody and blocking of parasites with soluble Sn restored normal susceptibility levels. Infection of Sn-deficient mice with bioluminescent L. infantum promastigotes revealed a moderate, strain-dependent role for Sn during visceral infection under the used experimental conditions. These data indicate that IFN-responsive Sn expression can enhance the susceptibility of macrophages to infection with visceral Leishmania promastigotes and that targeting of Sn may have some protective effects in early infection.
Collapse
Affiliation(s)
- Lieselotte Van Bockstal
- Laboratory of Microbiology, Parasitology and Hygiene (LMPH), Faculty of Pharmaceutical, Biomedical and Veterinary Sciences, University of Antwerp, Wilrijk, Belgium
| | - Dimitri Bulté
- Laboratory of Microbiology, Parasitology and Hygiene (LMPH), Faculty of Pharmaceutical, Biomedical and Veterinary Sciences, University of Antwerp, Wilrijk, Belgium
| | - Magali Van den Kerkhof
- Laboratory of Microbiology, Parasitology and Hygiene (LMPH), Faculty of Pharmaceutical, Biomedical and Veterinary Sciences, University of Antwerp, Wilrijk, Belgium
| | - Laura Dirkx
- Laboratory of Microbiology, Parasitology and Hygiene (LMPH), Faculty of Pharmaceutical, Biomedical and Veterinary Sciences, University of Antwerp, Wilrijk, Belgium
| | - Dorien Mabille
- Laboratory of Microbiology, Parasitology and Hygiene (LMPH), Faculty of Pharmaceutical, Biomedical and Veterinary Sciences, University of Antwerp, Wilrijk, Belgium
| | - Sarah Hendrickx
- Laboratory of Microbiology, Parasitology and Hygiene (LMPH), Faculty of Pharmaceutical, Biomedical and Veterinary Sciences, University of Antwerp, Wilrijk, Belgium
| | - Peter Delputte
- Laboratory of Microbiology, Parasitology and Hygiene (LMPH), Faculty of Pharmaceutical, Biomedical and Veterinary Sciences, University of Antwerp, Wilrijk, Belgium
| | - Louis Maes
- Laboratory of Microbiology, Parasitology and Hygiene (LMPH), Faculty of Pharmaceutical, Biomedical and Veterinary Sciences, University of Antwerp, Wilrijk, Belgium
| | - Guy Caljon
- Laboratory of Microbiology, Parasitology and Hygiene (LMPH), Faculty of Pharmaceutical, Biomedical and Veterinary Sciences, University of Antwerp, Wilrijk, Belgium
| |
Collapse
|