1
|
Zhang F, Wang S, Yang J, Fraser K, Gibson JM, Wang C, Dordick JS, Tomatsidou A, Linhardt RJ, Wang L, Sun X. Characterization of heparin interactions with Clostridioides difficile toxins and its potential as anti-CDI therapeutics. Carbohydr Polym 2025; 351:123143. [PMID: 39779041 PMCID: PMC11783924 DOI: 10.1016/j.carbpol.2024.123143] [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: 07/26/2024] [Revised: 11/25/2024] [Accepted: 12/11/2024] [Indexed: 01/11/2025]
Abstract
Clostridioides difficile (C. difficile) infection (CDI) is a life-threatening healthcare-associated infection occurring worldwide. C. difficile toxins (toxin A and toxin B) are the major virulence factors, causing CDI-related diarrhea and complications. Recent studies have shown that sulfated glycosaminoglcans (GAGs) are involved in mediating the cellular entry of these toxins. Although interactions between GAGs and toxins were reported, their binding kinetics and the structure features of glycans that facilitate toxin interaction have not been thoroughly studied. This research utilized surface plasmon resonance (SPR) to directly measure the kinetics of interactions between heparin and various toxins. Both toxin A and toxin B bind to heparin with high affinity (KD = 3.3 nM and 13.5 nM, respectively). SPR competition assay showed that both toxin A and B prefer binding to longer heparin chains and that all sulfation on the heparin chain is crucial for the heparin-toxin interaction. Finally, an in vitro assay showed that heparin and non-anticoagulant heparin inhibit the cell rounding caused by toxin A in HeLa cells.
Collapse
Affiliation(s)
- Fuming Zhang
- Department of Chemical and Biological Engineering, Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, Troy, NY 12180, USA.
| | - Shaohui Wang
- Department of Molecular Medicine, Morsani College of Medicine, University of South Florida Tampa, FL 33620, USA
| | - Jiyuan Yang
- Department of Chemical and Biological Engineering, Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, Troy, NY 12180, USA
| | - Keith Fraser
- Department of Biological Science, Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, Troy, NY 12180, USA
| | - James M Gibson
- Department of Biological Science, Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, Troy, NY 12180, USA
| | - Chunyu Wang
- Department of Biological Science, Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, Troy, NY 12180, USA
| | - Jonathan S Dordick
- Department of Chemical and Biological Engineering, Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, Troy, NY 12180, USA
| | - Anastasia Tomatsidou
- Department of Molecular Medicine, Morsani College of Medicine, University of South Florida Tampa, FL 33620, USA
| | - Robert J Linhardt
- Department of Chemical and Biological Engineering, Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, Troy, NY 12180, USA
| | - Lianchun Wang
- Department of Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida, Tampa, FL 33620, USA
| | - Xingmin Sun
- Department of Molecular Medicine, Morsani College of Medicine, University of South Florida Tampa, FL 33620, USA.
| |
Collapse
|
2
|
Roberts JM, Milo S, Metcalf DG. Harnessing the Power of Our Immune System: The Antimicrobial and Antibiofilm Properties of Nitric Oxide. Microorganisms 2024; 12:2543. [PMID: 39770746 PMCID: PMC11677572 DOI: 10.3390/microorganisms12122543] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2024] [Revised: 12/04/2024] [Accepted: 12/09/2024] [Indexed: 01/11/2025] Open
Abstract
Nitric oxide (NO) is a free radical of the human innate immune response to invading pathogens. NO, produced by nitric oxide synthases (NOSs), is used by the immune system to kill microorganisms encapsulated within phagosomes via protein and DNA disruption. Owing to its ability to disperse biofilm-bound microorganisms, penetrate the biofilm matrix, and act as a signal molecule, NO may also be effective as an antibiofilm agent. NO can be considered an underappreciated antimicrobial that could be levied against infected, at-risk, and hard-to-heal wounds due to the inherent lack of bacterial resistance, and tolerance by human tissues. NO produced within a wound dressing may be an effective method of disrupting biofilms and killing microorganisms in hard-to-heal wounds such as diabetic foot ulcers, venous leg ulcers, and pressure injuries. We have conducted a narrative review of the evidence underlying the key antimicrobial and antibiofilm mechanisms of action of NO for it to serve as an exogenously-produced antimicrobial agent in dressings used in the treatment of hard-to-heal wounds.
Collapse
Affiliation(s)
| | | | - Daniel Gary Metcalf
- Advanced Wound Care Research & Development, Convatec, Deeside Industrial Park, Deeside CH5 2NU, UK; (J.M.R.); (S.M.)
| |
Collapse
|
3
|
Yadav M, Chaudhary PP, Ratley G, D’Souza B, Kaur M, Ganesan S, Kabat J, Myles IA. TRPA1 Influences Staphylococcus aureus Skin Infection in Mice and Associates with HIF-1a and MAPK Pathway Modulation. Int J Mol Sci 2024; 25:9933. [PMID: 39337422 PMCID: PMC11432213 DOI: 10.3390/ijms25189933] [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: 08/01/2024] [Revised: 09/05/2024] [Accepted: 09/10/2024] [Indexed: 09/30/2024] Open
Abstract
Infections caused by methicillin-resistant Staphylococcus aureus (MRSA) are a major public health burden. Emerging antibiotic resistance has heightened the need for new treatment approaches for MRSA infection such as developing novel antimicrobial agents and enhancing the host's defense response. The thermo-ion channels Transient Receptor Potential (TRP-) A1 and V1 have been identified as modulators of S. aureus quorum sensing in cell culture models. However, their effects on in vivo infection control are unknown. In this study, we investigated the therapeutic effect of natural TRP ion channel inhibitors on MRSA skin infection in mice. While deletion of TRPV1 did not affect lesion size or inflammatory markers, TRPA1-/- mice demonstrated significantly reduced infection severity and abscess size. Treatment with natural inhibitors of TRPA1 with or without blockade of TRPV1 also reduced abscess size. Tissue transcriptomic data coupled with immunohistochemistry revealed that TRPA1 inhibition impacted heat shock protein expression (HSP), modulated the HIF-1a and MAPK pathways, and reduced IL4 expression. Additionally, metabolomics data showed an impact on purine and glycosaminoglycan pathways. Multi-omic integration of transcriptomic and metabolic data revealed that diacylglycerol metabolism was the likely bridge between metabolic and immunological impacts. Our findings suggest that TRPA1 antagonism could provide a promising and cost-effective therapeutic approach for reducing the severity of MRSA infection, and presents a novel underlying molecular mechanism.
Collapse
Affiliation(s)
- Manoj Yadav
- Epithelial Therapeutic Unit, National Institute of Allergy, and Infectious Disease, National Institutes of Health, Bethesda, MD 20892, USA; (P.P.C.); (G.R.); (B.D.); (M.K.); (I.A.M.)
| | - Prem Prashant Chaudhary
- Epithelial Therapeutic Unit, National Institute of Allergy, and Infectious Disease, National Institutes of Health, Bethesda, MD 20892, USA; (P.P.C.); (G.R.); (B.D.); (M.K.); (I.A.M.)
| | - Grace Ratley
- Epithelial Therapeutic Unit, National Institute of Allergy, and Infectious Disease, National Institutes of Health, Bethesda, MD 20892, USA; (P.P.C.); (G.R.); (B.D.); (M.K.); (I.A.M.)
| | - Brandon D’Souza
- Epithelial Therapeutic Unit, National Institute of Allergy, and Infectious Disease, National Institutes of Health, Bethesda, MD 20892, USA; (P.P.C.); (G.R.); (B.D.); (M.K.); (I.A.M.)
| | - Mahaldeep Kaur
- Epithelial Therapeutic Unit, National Institute of Allergy, and Infectious Disease, National Institutes of Health, Bethesda, MD 20892, USA; (P.P.C.); (G.R.); (B.D.); (M.K.); (I.A.M.)
| | - Sundar Ganesan
- Biological Imaging Section, Research Technologies Branch, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA; (S.G.); (J.K.)
| | - Juraj Kabat
- Biological Imaging Section, Research Technologies Branch, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA; (S.G.); (J.K.)
| | - Ian A. Myles
- Epithelial Therapeutic Unit, National Institute of Allergy, and Infectious Disease, National Institutes of Health, Bethesda, MD 20892, USA; (P.P.C.); (G.R.); (B.D.); (M.K.); (I.A.M.)
| |
Collapse
|
4
|
Rehman S, Antonovic AK, McIntire IE, Zheng H, Cleaver L, Baczynska M, Adams CO, Portlock T, Richardson K, Shaw R, Oregioni A, Mastroianni G, Whittaker SBM, Kelly G, Lorenz CD, Fornili A, Cianciotto NP, Garnett JA. The Legionella collagen-like protein employs a distinct binding mechanism for the recognition of host glycosaminoglycans. Nat Commun 2024; 15:4912. [PMID: 38851738 PMCID: PMC11162425 DOI: 10.1038/s41467-024-49255-4] [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: 09/16/2023] [Accepted: 05/30/2024] [Indexed: 06/10/2024] Open
Abstract
Bacterial adhesion is a fundamental process which enables colonisation of niche environments and is key for infection. However, in Legionella pneumophila, the causative agent of Legionnaires' disease, these processes are not well understood. The Legionella collagen-like protein (Lcl) is an extracellular peripheral membrane protein that recognises sulphated glycosaminoglycans on the surface of eukaryotic cells, but also stimulates bacterial aggregation in response to divalent cations. Here we report the crystal structure of the Lcl C-terminal domain (Lcl-CTD) and present a model for intact Lcl. Our data reveal that Lcl-CTD forms an unusual trimer arrangement with a positively charged external surface and negatively charged solvent exposed internal cavity. Through molecular dynamics simulations, we show how the glycosaminoglycan chondroitin-4-sulphate associates with the Lcl-CTD surface via distinct binding modes. Our findings show that Lcl homologs are present across both the Pseudomonadota and Fibrobacterota-Chlorobiota-Bacteroidota phyla and suggest that Lcl may represent a versatile carbohydrate-binding mechanism.
Collapse
Affiliation(s)
- Saima Rehman
- Centre for Host-Microbiome Interactions, Faculty of Dental, Oral & Craniofacial Sciences, King's College London, London, UK
| | - Anna Katarina Antonovic
- Department of Chemistry, School of Physical and Chemical Sciences, Queen Mary University of London, London, UK
| | - Ian E McIntire
- Department of Microbiology and Immunology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Huaixin Zheng
- Department of Microbiology and Immunology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Leanne Cleaver
- Centre for Host-Microbiome Interactions, Faculty of Dental, Oral & Craniofacial Sciences, King's College London, London, UK
| | - Maria Baczynska
- Centre for Host-Microbiome Interactions, Faculty of Dental, Oral & Craniofacial Sciences, King's College London, London, UK
- Biological Physics & Soft Matter Research Group, Department of Physics, King's College London, London, UK
| | - Carlton O Adams
- Department of Microbiology and Immunology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Theo Portlock
- Centre for Host-Microbiome Interactions, Faculty of Dental, Oral & Craniofacial Sciences, King's College London, London, UK
- School of Biological and Behavioural Sciences, Queen Mary University of London, London, UK
| | - Katherine Richardson
- School of Biological and Behavioural Sciences, Queen Mary University of London, London, UK
| | - Rosie Shaw
- School of Biological and Behavioural Sciences, Queen Mary University of London, London, UK
| | - Alain Oregioni
- The Medical Research Council Biomedical NMR Centre, The Francis Crick Institute, 1 Midland Road, London, NW1 1AT, UK
| | - Giulia Mastroianni
- Department of Chemistry, School of Physical and Chemical Sciences, Queen Mary University of London, London, UK
| | - Sara B-M Whittaker
- School of Cancer Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
| | - Geoff Kelly
- The Medical Research Council Biomedical NMR Centre, The Francis Crick Institute, 1 Midland Road, London, NW1 1AT, UK
| | - Christian D Lorenz
- Biological Physics & Soft Matter Research Group, Department of Physics, King's College London, London, UK
| | - Arianna Fornili
- Department of Chemistry, School of Physical and Chemical Sciences, Queen Mary University of London, London, UK.
| | - Nicholas P Cianciotto
- Department of Microbiology and Immunology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA.
| | - James A Garnett
- Centre for Host-Microbiome Interactions, Faculty of Dental, Oral & Craniofacial Sciences, King's College London, London, UK.
| |
Collapse
|
5
|
Xie Y, Butler M. N-glycomic profiling of capsid proteins from Adeno-Associated Virus serotypes. Glycobiology 2024; 34:cwad074. [PMID: 37774344 PMCID: PMC10950483 DOI: 10.1093/glycob/cwad074] [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: 05/01/2023] [Revised: 09/14/2023] [Accepted: 09/23/2023] [Indexed: 10/01/2023] Open
Abstract
Adeno-associated virus (AAV) vector has become the leading platform for gene delivery. Each serotype exhibits a different tissue tropism, immunogenicity, and in vivo transduction performance. Therefore, selecting the most suitable AAV serotype is critical for efficient gene delivery to target cells or tissues. Genome divergence among different serotypes is due mainly to the hypervariable regions of the AAV capsid proteins. However, the heterogeneity of capsid glycosylation is largely unexplored. In the present study, the N-glycosylation profiles of capsid proteins of AAV serotypes 1 to 9 have been systemically characterized and compared using a previously developed high-throughput and high-sensitivity N-glycan profiling platform. The results showed that all 9 investigated AAV serotypes were glycosylated, with comparable profiles. The most conspicuous feature was the high abundance mannosylated N-glycans, including FM3, M5, M6, M7, M8, and M9, that dominated the chromatograms within a range of 74 to 83%. Another feature was the relatively lower abundance of fucosylated and sialylated N-glycan structures, in the range of 23%-40% and 10%-17%, respectively. However, the exact N-glycan composition differed. These differences may be utilized to identify potential structural relationships between the 9 AAV serotypes. The current research lays the foundation for gaining better understanding of the importance of N-glycans on the AAV capsid surface that may play a significant role in tissue tropism, interaction with cell surface receptors, cellular uptake, and intracellular processing.
Collapse
Affiliation(s)
- Yongjing Xie
- National Institute for Bioprocessing Research and Training, Foster Avenue, Mount Merrion, Blackrock, Co. Dublin, A94 X099, Ireland
| | - Michael Butler
- National Institute for Bioprocessing Research and Training, Foster Avenue, Mount Merrion, Blackrock, Co. Dublin, A94 X099, Ireland
- School of Chemical and Bioprocess Engineering, University College Dublin (UCD), Belfield, Dublin 4, D04 V1W8, Ireland
| |
Collapse
|
6
|
Ouyang Y, Nauwynck HJ. Molecular basis for the different PCV2 susceptibility of T-lymphoblasts in Landrace and Piétrain pigs. Vet Res 2024; 55:22. [PMID: 38374131 PMCID: PMC10875804 DOI: 10.1186/s13567-024-01275-0] [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: 11/09/2023] [Accepted: 01/29/2024] [Indexed: 02/21/2024] Open
Abstract
Clinically, Landrace pigs are more susceptible to porcine circovirus-associated diseases (PCVADs) than Piétrain pigs. We previously found that porcine circovirus type 2 (PCV2) can infect T-lymphoblasts. The present study examined the replication kinetics of six PCV2 strains in the lymphoblasts of Landrace and Piétrain pigs. The results showed that T-lymphoblasts from Landrace pigs are much more susceptible to PCV2 infection than those from Piétrain pigs. In addition, PCV2 replication was strain-dependent. PCV2 binding to T-lymphoblasts was partially mediated by chondroitin sulfate (CS) and dermatan sulfate (DS). Phosphacan, an effective internalization mediator in monocytes that contains several CS chains, was also demonstrated to be involved in PCV2 internalization. Viral binding and internalization were not different between the two breeds, however, the subsequent step, the disassembly was. Although inhibition of serine proteases blocked PCV2 replication in both Landrace and Piétrain pigs, this only occurred at a neutral pH in Piétrain pigs, whereas this occurred also at a low pH in Landrace. This suggested that more proteases can cleave PCV2 in Landrace lymphoblasts than in Piétrain lymphoblasts, explaining the better replication. Through co-localization studies of viral particles with endo-lysosomal markers, and quantitative analysis of organelle sizes during viral internalization, it was observed that PCV2 may exhibit a higher propensity for viral escape from late endosomes in Landrace pigs (smaller) compared to Piétrain pigs. These results provide new understandings of the different PCV2 susceptibility in Landrace and Piétrain pigs.
Collapse
Affiliation(s)
- Yueling Ouyang
- Laboratory of Virology, Department of Translational Physiology, Infectiology and Public Health, Faculty of Veterinary Medicine, Ghent University, Ghent, Belgium.
| | - Hans J Nauwynck
- Laboratory of Virology, Department of Translational Physiology, Infectiology and Public Health, Faculty of Veterinary Medicine, Ghent University, Ghent, Belgium
| |
Collapse
|
7
|
Tan WS, Rong E, Dry I, Lillico S, Law A, Digard P, Whitelaw B, Dalziel RG. Validation of Candidate Host Cell Entry Factors for Bovine Herpes Virus Type-1 Based on a Genome-Wide CRISPR Knockout Screen. Viruses 2024; 16:297. [PMID: 38400072 PMCID: PMC10893506 DOI: 10.3390/v16020297] [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: 12/08/2023] [Revised: 02/09/2024] [Accepted: 02/13/2024] [Indexed: 02/25/2024] Open
Abstract
To identify host factors that affect Bovine Herpes Virus Type 1 (BoHV-1) infection we previously applied a genome wide CRISPR knockout screen targeting all bovine protein coding genes. By doing so we compiled a list of both pro-viral and anti-viral proteins involved in BoHV-1 replication. Here we provide further analysis of those that are potentially involved in viral entry into the host cell. We first generated single cell knockout clones deficient in some of the candidate genes for validation. We provide evidence that Polio Virus Receptor-related protein (PVRL2) serves as a receptor for BoHV-1, mediating more efficient entry than the previously identified Polio Virus Receptor (PVR). By knocking out two enzymes that catalyze HSPG chain elongation, HST2ST1 and GLCE, we further demonstrate the significance of HSPG in BoHV-1 entry. Another intriguing cluster of candidate genes, COG1, COG2 and COG4-7 encode six subunits of the Conserved Oligomeric Golgi (COG) complex. MDBK cells lacking COG6 produced fewer but bigger plaques compared to control cells, suggesting more efficient release of newly produced virions from these COG6 knockout cells, due to impaired HSPG biosynthesis. We further observed that viruses produced by the COG6 knockout cells consist of protein(s) with reduced N-glycosylation, potentially explaining their lower infectivity. To facilitate candidate validation, we also detailed a one-step multiplex CRISPR interference (CRISPRi) system, an orthogonal method to KO that enables quick and simultaneous deployment of three CRISPRs for efficient gene inactivation. Using CRISPR3i, we verified eight candidates that have been implicated in the synthesis of surface heparan sulfate proteoglycans (HSPGs). In summary, our experiments confirmed the two receptors PVR and PVRL2 for BoHV-1 entry into the host cell and other factors that affect this process, likely through the direct or indirect roles they play during HSPG synthesis and glycosylation of viral proteins.
Collapse
Affiliation(s)
- Wenfang Spring Tan
- Division of Infection and Immunity, the Roslin Institute, Easter Bush Campus, University of Edinburgh, Edinburgh EH259RG, UK (I.D.); (P.D.); (R.G.D.)
| | - Enguang Rong
- Division of Infection and Immunity, the Roslin Institute, Easter Bush Campus, University of Edinburgh, Edinburgh EH259RG, UK (I.D.); (P.D.); (R.G.D.)
| | - Inga Dry
- Division of Infection and Immunity, the Roslin Institute, Easter Bush Campus, University of Edinburgh, Edinburgh EH259RG, UK (I.D.); (P.D.); (R.G.D.)
| | - Simon Lillico
- Division of Functional Genetics and Development, the Roslin Institute, Easter Bush Campus, University of Edinburgh, Edinburgh EH259RG, UK; (S.L.); (B.W.)
- Centre for Tropical Livestock Genetics and Health, the Roslin Institute, Easter Bush Campus, University of Edinburgh, Edinburgh EH259RG, UK
| | - Andy Law
- Division of Genetics and Genomics, the Roslin Institute, Easter Bush Campus, University of Edinburgh, Edinburgh EH259RG, UK;
| | - Paul Digard
- Division of Infection and Immunity, the Roslin Institute, Easter Bush Campus, University of Edinburgh, Edinburgh EH259RG, UK (I.D.); (P.D.); (R.G.D.)
| | - Bruce Whitelaw
- Division of Functional Genetics and Development, the Roslin Institute, Easter Bush Campus, University of Edinburgh, Edinburgh EH259RG, UK; (S.L.); (B.W.)
- Division of Genetics and Genomics, the Roslin Institute, Easter Bush Campus, University of Edinburgh, Edinburgh EH259RG, UK;
| | - Robert G. Dalziel
- Division of Infection and Immunity, the Roslin Institute, Easter Bush Campus, University of Edinburgh, Edinburgh EH259RG, UK (I.D.); (P.D.); (R.G.D.)
| |
Collapse
|
8
|
Chienwichai P, Tipthara P, Tarning J, Limpanont Y, Chusongsang P, Chusongsang Y, Kiangkoo N, Adisakwattana P, Reamtong O. Identification of trans-genus biomarkers for early diagnosis of intestinal schistosomiasis and progression of gut pathology in a mouse model using metabolomics. PLoS Negl Trop Dis 2024; 18:e0011966. [PMID: 38381759 PMCID: PMC10880994 DOI: 10.1371/journal.pntd.0011966] [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: 11/12/2023] [Accepted: 02/05/2024] [Indexed: 02/23/2024] Open
Abstract
Schistosomiasis is one of the most devastating human diseases worldwide. The disease is caused by six species of Schistosoma blood fluke; five of which cause intestinal granulomatous inflammation and bleeding. The current diagnostic method is inaccurate and delayed, hence, biomarker identification using metabolomics has been applied. However, previous studies only investigated infection caused by one Schistosoma spp., leaving a gap in the use of biomarkers for other species. No study focused on understanding the progression of intestinal disease. Therefore, we aimed to identify early gut biomarkers of infection with three Schistosoma spp. and progression of intestinal pathology. We infected 3 groups of mice, 3 mice each, with Schistosoma mansoni, Schistosoma japonicum or Schistosoma mekongi and collected their feces before and 1, 2, 4 and 8 weeks after infection. Metabolites in feces were extracted and identified using mass spectrometer-based metabolomics. Metabolites were annotated and analyzed with XCMS bioinformatics tool and Metaboanalyst platform. From >36,000 features in all conditions, multivariate analysis found a distinct pattern at each time point for all species. Pathway analysis reported alteration of several lipid metabolism pathways as infection progressed. Disturbance of the glycosaminoglycan degradation pathway was found with the presence of parasite eggs, indicating involvement of this pathway in disease progression. Biomarkers were discovered using a combination of variable importance for projection score cut-off and receiver operating characteristic curve analysis. Five molecules met our criteria and were present in all three species: 25-hydroxyvitamin D2, 1α-hydroxy-2β-(3-hydroxypropoxy) vitamin D3, Ganoderic acid Md, unidentified feature with m/z 455.3483, and unidentified feature with m/z 456.3516. These molecules were proposed as trans-genus biomarkers of early schistosomiasis. Our findings provide evidence for disease progression in intestinal schistosomiasis and potential biomarkers, which could be beneficial for early detection of this disease.
Collapse
Affiliation(s)
- Peerut Chienwichai
- Princess Srisavangavadhana College of Medicine, Chulabhorn Royal Academy, Bangkok, Thailand
| | - Phornpimon Tipthara
- Mahidol Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
| | - Joel Tarning
- Mahidol Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
- Centre for Tropical Medicine and Global Health, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, United Kingdom
| | - Yanin Limpanont
- Department of Social and Environmental Medicine, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
| | - Phiraphol Chusongsang
- Department of Social and Environmental Medicine, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
| | - Yupa Chusongsang
- Department of Social and Environmental Medicine, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
| | - Nuttapohn Kiangkoo
- Department of Social and Environmental Medicine, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
| | - Poom Adisakwattana
- Department of Helminthology, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
| | - Onrapak Reamtong
- Department of Molecular Tropical Medicine and Genetics, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
| |
Collapse
|
9
|
Merchán A, Ramírez-López P, Martínez C, Suárez JR, Perona A, Hernáiz MJ. Exploring Rigid and Flexible Scaffolds to Develop Potent Glucuronic Acid Glycodendrimers for Dengue Virus Inhibition. Bioconjug Chem 2024; 35:34-42. [PMID: 37964742 PMCID: PMC10797590 DOI: 10.1021/acs.bioconjchem.3c00309] [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: 07/12/2023] [Revised: 10/27/2023] [Accepted: 10/27/2023] [Indexed: 11/16/2023]
Abstract
Multivalent glycodendrimers are valuable tools for studying carbohydrate-protein interactions, and their scaffolds represent important components to increase specificity and affinity. Previous work by our group described the preparation of a tetravalent glucuronic acid rigid dendron that binds with good affinity to the dengue virus envelope protein (KD = 22 μM). Herein, the chemical synthesis and binding analysis of three new sets of rigid, semirigid, and flexible glucuronic acid-based dendrimers bearing different levels of multivalency and their interactions with the dengue virus envelope protein are described. The different oligoalkynyl scaffolds were coupled to glucuronic acid azides by a copper-catalyzed azide-alkyne cycloaddition reaction through optimized synthetic strategies to afford the desired glycodendrimers with good yields. Surface plasmon resonance studies have demonstrated that glycodendrimers 12b and 12c, with flexible scaffolds, give the best binding interactions with the dengue virus envelope protein (12b: KD = 0.487 μM and 12c: KD = 0.624 μM). Their binding constant values were 45 and 35 times higher than the one obtained in previous studies with a rigid tetravalent glucuronic acid dendron (KD = 22 μM), respectively. Molecular modeling studies were carried out in order to understand the difference in behavior observed for 12b and 12c. This work reports an efficient glycodendrimer chemical synthesis process that provides an appropriate scaffold that offers an easy and versatile strategy to find new active compounds against the dengue virus.
Collapse
Affiliation(s)
- Alejandro Merchán
- Departamento de Química
en Ciencias Farmacéuticas, Facultad de Farmacia, Universidad Complutense de Madrid, Plz. Ramón y Cajal s/n, Madrid, C.P. 28040, España
| | - Pedro Ramírez-López
- Departamento de Química
en Ciencias Farmacéuticas, Facultad de Farmacia, Universidad Complutense de Madrid, Plz. Ramón y Cajal s/n, Madrid, C.P. 28040, España
| | - Carlos Martínez
- Departamento de Química
en Ciencias Farmacéuticas, Facultad de Farmacia, Universidad Complutense de Madrid, Plz. Ramón y Cajal s/n, Madrid, C.P. 28040, España
| | - José Ramón Suárez
- Departamento de Química
en Ciencias Farmacéuticas, Facultad de Farmacia, Universidad Complutense de Madrid, Plz. Ramón y Cajal s/n, Madrid, C.P. 28040, España
| | - Almudena Perona
- Departamento de Química
en Ciencias Farmacéuticas, Facultad de Farmacia, Universidad Complutense de Madrid, Plz. Ramón y Cajal s/n, Madrid, C.P. 28040, España
| | - María J. Hernáiz
- Departamento de Química
en Ciencias Farmacéuticas, Facultad de Farmacia, Universidad Complutense de Madrid, Plz. Ramón y Cajal s/n, Madrid, C.P. 28040, España
| |
Collapse
|
10
|
Rehman S, Antonovic AK, McIntire IE, Zheng H, Cleaver L, Adams CO, Portlock T, Richardson K, Shaw R, Oregioni A, Mastroianni G, Whittaker SBM, Kelly G, Fornili A, Cianciotto NP, Garnett JA. The Legionella collagen-like protein employs a unique binding mechanism for the recognition of host glycosaminoglycans. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.12.10.570962. [PMID: 38106198 PMCID: PMC10723406 DOI: 10.1101/2023.12.10.570962] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2023]
Abstract
Bacterial adhesion is a fundamental process which enables colonisation of niche environments and is key for infection. However, in Legionella pneumophila, the causative agent of Legionnaires' disease, these processes are not well understood. The Legionella collagen-like protein (Lcl) is an extracellular peripheral membrane protein that recognises sulphated glycosaminoglycans (GAGs) on the surface of eukaryotic cells, but also stimulates bacterial aggregation in response to divalent cations. Here we report the crystal structure of the Lcl C-terminal domain (Lcl-CTD) and present a model for intact Lcl. Our data reveal that Lcl-CTD forms an unusual dynamic trimer arrangement with a positively charged external surface and a negatively charged solvent exposed internal cavity. Through Molecular Dynamics (MD) simulations, we show how the GAG chondroitin-4-sulphate associates with the Lcl-CTD surface via unique binding modes. Our findings show that Lcl homologs are present across both the Pseudomonadota and Fibrobacterota-Chlorobiota-Bacteroidota phyla and suggest that Lcl may represent a versatile carbohydrate binding mechanism.
Collapse
Affiliation(s)
- Saima Rehman
- Centre for Host-Microbiome Interactions, Faculty of Dental, Oral & Craniofacial Sciences, King’s College London, London, UK
| | - Anna K. Antonovic
- School of Physical and Chemical Sciences, Queen Mary University of London, London, UK
| | - Ian E. McIntire
- Department of Microbiology and Immunology, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Huaixin Zheng
- Department of Microbiology and Immunology, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Leanne Cleaver
- Centre for Host-Microbiome Interactions, Faculty of Dental, Oral & Craniofacial Sciences, King’s College London, London, UK
| | - Carlton O. Adams
- Department of Microbiology and Immunology, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Theo Portlock
- Centre for Host-Microbiome Interactions, Faculty of Dental, Oral & Craniofacial Sciences, King’s College London, London, UK
- School of Biological and Behavioural Sciences, Queen Mary University of London, London, UK
| | - Katherine Richardson
- School of Biological and Behavioural Sciences, Queen Mary University of London, London, UK
| | - Rosie Shaw
- School of Biological and Behavioural Sciences, Queen Mary University of London, London, UK
| | - Alain Oregioni
- The Medical Research Council Biomedical NMR Centre, the Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK
| | - Giulia Mastroianni
- School of Physical and Chemical Sciences, Queen Mary University of London, London, UK
| | - Sara B-M. Whittaker
- School of Cancer Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
| | - Geoff Kelly
- The Medical Research Council Biomedical NMR Centre, the Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK
| | - Arianna Fornili
- School of Physical and Chemical Sciences, Queen Mary University of London, London, UK
| | - Nicholas P. Cianciotto
- Department of Microbiology and Immunology, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - James A. Garnett
- Centre for Host-Microbiome Interactions, Faculty of Dental, Oral & Craniofacial Sciences, King’s College London, London, UK
| |
Collapse
|
11
|
Ouyang Y, Nauwynck HJ. PCV2 Uptake by Porcine Monocytes Is Strain-Dependent and Is Associated with Amino Acid Characteristics on the Capsid Surface. Microbiol Spectr 2023; 11:e0380522. [PMID: 36719220 PMCID: PMC10100887 DOI: 10.1128/spectrum.03805-22] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2022] [Accepted: 01/13/2023] [Indexed: 02/01/2023] Open
Abstract
Porcine circovirus type 2 (PCV2) is associated with several economically important diseases that are described as PCV2-associated diseases (PCVADs). PCV2 is replicating in lymphoblasts, and PCV2 particles are taken up by monocytes without effective replication or complete degradation. Glycosaminoglycans (GAGs) have been demonstrated to be important receptors for PCV2 binding and entry in T-lymphocytes and continuous cell lines. The objective of this study was to determine whether differences exist in viral uptake and outcome among six PCV2 strains from different disease outbreaks in primary porcine monocytes: Stoon-1010 (PCV2a; PMWS), 1121 (PCV2a; abortion), 1147 (PCV2b; PDNS), 09V448 (PCV2d-1; PCVAD with high viral load in lymphoid tissues [PCVADhigh]), DE222-13 (PCV2d-2; PCVADhigh), and 19V245 (PCV2d-2; PCVADhigh). The uptake of PCV2 in peripheral blood monocytes was different among the PCV2 strains. A large number of PCV2 particles were found in the monocytes for Stoon-1010, DE222-13, and 19V245, while a low number was found for 1121, 1147, and 09V448. Competition with, and removal of GAGs on the cell surface, demonstrated an important role of chondroitin sulfate (CS) and dermatan sulfate (DS) in PCV2 entry into monocytes. The mapping of positively/negatively charged amino acids exposed on the surface of PCV2 capsids revealed that their number and distribution could have an impact on the binding of the capsids to GAGs, and the internalization into monocytes. Based on the distribution of positively charged amino acids on PCV2 capsids, phosphacan was hypothesized, and further demonstrated, as an effective candidate to mediate virus attachment to, and internalization in, monocytes. IMPORTANCE PCV2 is present on almost every pig farm in the world and is associated with a high number of diseases (PCV2-associated diseases [PCVADs]). It causes severe economic losses. Although vaccination is successfully applied in the field, there are still a lot of unanswered questions on the pathogenesis of PCV2 infections. This article reports on the uptake difference of various PCV2 strains by peripheral blood monocytes, and reveals the mechanism of the strong viral uptake ability of monocytes of Piétrain pigs. We further demonstrated that: (i) GAGs mediate the uptake of PCV2 particles by monocytes, (ii) positively charged three-wings-windmill-like amino acid patterns on the capsid outer surface are activating PCV2 uptake, and (iii) phosphacan is one of the potential candidates for PCV2 internalization. These results provide new insights into the mechanisms involved in PCVAD and contribute to a better understanding of PCV2 evolution. This may lead to the development of resistant pigs.
Collapse
Affiliation(s)
- Yueling Ouyang
- Laboratory of Virology, Department of Translational Physiology, Infectiology and Public Health, Faculty of Veterinary Medicine, Ghent University, Ghent, Belgium
| | - Hans J. Nauwynck
- Laboratory of Virology, Department of Translational Physiology, Infectiology and Public Health, Faculty of Veterinary Medicine, Ghent University, Ghent, Belgium
| |
Collapse
|
12
|
Kantarcioglu B, Mehrotra S, Papineni C, Siddiqui F, Kouta A, Hoppensteadt D, Bansal V, Darki A, Van Thiel DH, Fareed J. Endogenous Glycosaminoglycans in Various Pathologic Plasma Samples as Measured by a Fluorescent Quenching Method. Clin Appl Thromb Hemost 2022; 28:10760296221144047. [PMID: 36474353 PMCID: PMC9732799 DOI: 10.1177/10760296221144047] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Endogenous glycosaminoglycans (GAGs) with a similar structure to heparin are widely distributed in various tissues. A fluorescence probe, namely Heparin Red, can detect polyanionic GAGs in plasma samples. The purpose of this study is to measure endogenous GAGs in various plasma samples obtained from different pathologic states in comparison to healthy controls utilizing this method. Plasma samples were obtained from patient groups including atrial fibrillation (AF), end-stage-renal-disease (ESRD), diabetes mellitus (DM), sepsis, cancer, liver disease (LD), and pulmonary embolism (PE). Normal human plasma (NHP) was used as healthy controls. The Heparin Red kit from Red Probes (Münster, Germany) was used for the quantification of endogenous GAGs in each sample before and after heparinase I degradation. All results were compiled as group means ± SD for comparison. NHP was found to have relatively low levels of endogenous GAGs with a mean concentration of 0.06 μg/mL. The AF, ESRD, DM, and sepsis patient samples had a mean endogenous GAG concentration of 0.55, 0.72, 0.92, and 0.94 μg/mL, respectively. The levels of endogenous GAGs were highest in cancer, LD, and PE patient plasma samples with a mean concentration of 1.95, 2.78, and 2.83 μg/mL, respectively. Heparinase I degradation resulted in a decline in GAG levels in plasma samples. These results clearly show that detectable Heparin Red sensitive endogenous GAGs are present in circulating plasma at varying levels in various patient groups. Additional studies are necessary to understand this complex pathophysiology.
Collapse
Affiliation(s)
- Bulent Kantarcioglu
- Department of Pathology and Laboratory Medicine, Cardiovascular
Research Institute, Loyola
University Chicago, Health Sciences
Division, Maywood, IL, USA,Bulent Kantarcioglu, Department of
Pathology and Laboratory Medicine, Cardiovascular Research Institute, Loyola
University Chicago, Health Sciences Division, Maywood, IL 60153, USA.
| | - Siddharth Mehrotra
- Department of Pathology and Laboratory Medicine, Cardiovascular
Research Institute, Loyola
University Chicago, Health Sciences
Division, Maywood, IL, USA
| | - Charulatha Papineni
- Department of Pathology and Laboratory Medicine, Cardiovascular
Research Institute, Loyola
University Chicago, Health Sciences
Division, Maywood, IL, USA,Department of Molecular Pharmacology and Neuroscience,
Loyola
University Chicago, Maywood, IL, USA
| | - Fakiha Siddiqui
- Department of Pathology and Laboratory Medicine, Cardiovascular
Research Institute, Loyola
University Chicago, Health Sciences
Division, Maywood, IL, USA,Program in Health Sciences, UCAM - Universidad Católica San Antonio de
Murcia, Murcia, Spain
| | - Ahmed Kouta
- Department of Pathology and Laboratory Medicine, Cardiovascular
Research Institute, Loyola
University Chicago, Health Sciences
Division, Maywood, IL, USA,Department of Molecular Pharmacology and Neuroscience,
Loyola
University Chicago, Maywood, IL, USA
| | - Debra Hoppensteadt
- Department of Pathology and Laboratory Medicine, Cardiovascular
Research Institute, Loyola
University Chicago, Health Sciences
Division, Maywood, IL, USA
| | - Vinod Bansal
- Department of Internal Medicine, Nephrology,
Loyola
University Medical Center, Maywood, IL,
USA
| | - Amir Darki
- Cardiology Department, Loyola University Medical
Center, Maywood, IL, USA
| | - David H. Van Thiel
- Division of Gastroenterology and Hepatology,
Rush Oak Park
Hospital, Oak Park, Illinois and Rush
University Medical Center, Chicago, IL, USA
| | - Jawed Fareed
- Department of Pathology and Laboratory Medicine, Cardiovascular
Research Institute, Loyola
University Chicago, Health Sciences
Division, Maywood, IL, USA
| |
Collapse
|
13
|
Ramarajan MG, Saraswat M, Budhraja R, Garapati K, Raymond K, Pandey A. Mass spectrometric analysis of chondroitin sulfate-linked peptides. JOURNAL OF PROTEINS AND PROTEOMICS 2022; 13:187-203. [PMID: 36213313 PMCID: PMC9526814 DOI: 10.1007/s42485-022-00092-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Revised: 06/08/2022] [Accepted: 06/14/2022] [Indexed: 11/26/2022]
Abstract
Chondroitin sulfate proteoglycans (CSPGs) are extracellular matrix components composed of linear glycosaminoglycan (GAG) side chains attached to a core protein. CSPGs play a vital role in neurodevelopment, signal transduction, cellular proliferation and differentiation and tumor metastasis through interaction with growth factors and signaling proteins. These pleiotropic functions of proteoglycans are regulated spatiotemporally by the GAG chains attached to the core protein. There are over 70 chondroitin sulfate-linked proteoglycans reported in cells, cerebrospinal fluid and urine. A core glycan linker of 3-6 monosaccharides attached to specific serine residues can be extended by 20-200 disaccharide repeating units making intact CSPGs very large and impractical to analyze. The current paradigm of CSPG analysis involves digesting the GAG chains by chondroitinase enzymes and analyzing either the protein part, the disaccharide repeats, or both by mass spectrometry. This method, however, provides no information about the site of attachment or the composition of linker oligosaccharides and the degree of sulfation and/or phosphorylation. Further, the analysis by mass spectrometry and subsequent identification of novel CSPGs is hampered by technical challenges in their isolation, less optimal ionization and data analysis. Unknown identity of the linker oligosaccharide also makes it more difficult to identify the glycan composition using database searching approaches. Following chondroitinase digestion of long GAG chains linked to tryptic peptides, we identified intact GAG-linked peptides in clinically relevant samples including plasma, urine and dermal fibroblasts. These intact glycopeptides including their core linker glycans were identified by mass spectrometry using optimized stepped higher energy collision dissociation and electron-transfer/higher energy collision dissociation combined with hybrid database search/de novo glycan composition search. We identified 25 CSPGs including three novel CSPGs that have not been described earlier. Our findings demonstrate the utility of combining enrichment strategies and optimized high-resolution mass spectrometry analysis including alternative fragmentation methods for the characterization of CSPGs. Supplementary Information The online version contains supplementary material available at 10.1007/s42485-022-00092-3.
Collapse
Affiliation(s)
- Madan Gopal Ramarajan
- Department of Laboratory Medicine and Pathology, Mayo Clinic, 200 First ST SW, Rochester, MN 55905 USA
- Institute of Bioinformatics, International Technology Park, Bangalore, 560066 India
- Manipal Academy of Higher Education (MAHE), Manipal, 576104 Karnataka India
- Center for Molecular Medicine, National Institute of Mental Health and Neurosciences (NIMHANS), Hosur Road, Bangalore, 560 029 India
| | - Mayank Saraswat
- Department of Laboratory Medicine and Pathology, Mayo Clinic, 200 First ST SW, Rochester, MN 55905 USA
- Institute of Bioinformatics, International Technology Park, Bangalore, 560066 India
- Manipal Academy of Higher Education (MAHE), Manipal, 576104 Karnataka India
| | - Rohit Budhraja
- Department of Laboratory Medicine and Pathology, Mayo Clinic, 200 First ST SW, Rochester, MN 55905 USA
| | - Kishore Garapati
- Department of Laboratory Medicine and Pathology, Mayo Clinic, 200 First ST SW, Rochester, MN 55905 USA
- Institute of Bioinformatics, International Technology Park, Bangalore, 560066 India
- Manipal Academy of Higher Education (MAHE), Manipal, 576104 Karnataka India
- Center for Molecular Medicine, National Institute of Mental Health and Neurosciences (NIMHANS), Hosur Road, Bangalore, 560 029 India
| | - Kimiyo Raymond
- Biochemical Genetics Laboratory, Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN 55905 USA
| | - Akhilesh Pandey
- Department of Laboratory Medicine and Pathology, Mayo Clinic, 200 First ST SW, Rochester, MN 55905 USA
- Center for Individualized Medicine, Mayo Clinic, Rochester, MN 55905 USA
| |
Collapse
|
14
|
Nissen EJ, Saeger M, Nölle B, Roider J. Clinicopathological Correlation of Microbial Keratitis and Ahead: Is There a Corneal Sepsis? Klin Monbl Augenheilkd 2022; 239:857-866. [PMID: 35858596 DOI: 10.1055/a-1811-7171] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/17/2022]
Abstract
Microbial, infectious keratitis is a relevant indication for penetrating keratoplasty. The requirement for transplantation results in histopathological examination of the entire thickness of the cornea. Although the clinical diagnosis is not always possible to confirm, pathology can support diagnostic evidence of clinical presentation and pathogenesis. This is achieved with multiple methods from cytology, histochemistry, immunohistology, molecular pathology and in rare cases electron microscopy. These allow tissue-based detection of previous and parallel diseases and the responsible pathogens. The failure of satisfactory clinicopathological correlation raises the question whether a suspected pathogen was not ultimately responsible for destroyed corneal tissue. The pathogenesis of keratitis requiring transplantation is not yet completely understood, also on the experimental level. The development of such a keratitis can lead to a clinical symptomatology which can be described as "threatening organ dysfunction", a term used in sepsis research. Considering recent literature, possible correlations between sepsis and microbial keratitis and their relation to histopathology are discussed.
Collapse
Affiliation(s)
- Ebba J Nissen
- Klinik für Ophthalmologie, Universitätsklinikum Schleswig-Holstein, Campus Kiel, Deutschland
| | - Mark Saeger
- Klinik für Ophthalmologie, Universitätsklinikum Schleswig-Holstein, Campus Kiel, Deutschland
| | - Bernhard Nölle
- Klinik für Ophthalmologie, Universitätsklinikum Schleswig-Holstein, Campus Kiel, Deutschland
| | - Johann Roider
- Klinik für Ophthalmologie, Universitätsklinikum Schleswig-Holstein, Campus Kiel, Deutschland
| |
Collapse
|
15
|
Mechanical Forces Govern Interactions of Host Cells with Intracellular Bacterial Pathogens. Microbiol Mol Biol Rev 2022; 86:e0009420. [PMID: 35285720 PMCID: PMC9199418 DOI: 10.1128/mmbr.00094-20] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
To combat infectious diseases, it is important to understand how host cells interact with bacterial pathogens. Signals conveyed from pathogen to host, and vice versa, may be either chemical or mechanical. While the molecular and biochemical basis of host-pathogen interactions has been extensively explored, relatively less is known about mechanical signals and responses in the context of those interactions. Nevertheless, a wide variety of bacterial pathogens appear to have developed mechanisms to alter the cellular biomechanics of their hosts in order to promote their survival and dissemination, and in turn many host responses to infection rely on mechanical alterations in host cells and tissues to limit the spread of infection. In this review, we present recent findings on how mechanical forces generated by host cells can promote or obstruct the dissemination of intracellular bacterial pathogens. In addition, we discuss how in vivo extracellular mechanical signals influence interactions between host cells and intracellular bacterial pathogens. Examples of such signals include shear stresses caused by fluid flow over the surface of cells and variable stiffness of the extracellular matrix on which cells are anchored. We highlight bioengineering-inspired tools and techniques that can be used to measure host cell mechanics during infection. These allow for the interrogation of how mechanical signals can modulate infection alongside biochemical signals. We hope that this review will inspire the microbiology community to embrace those tools in future studies so that host cell biomechanics can be more readily explored in the context of infection studies.
Collapse
|
16
|
Martín C, Ordiales H, Vázquez F, Pevida M, Rodríguez D, Merayo J, Vázquez F, García B, Quirós LM. Bacteria associated with acne use glycosaminoglycans as cell adhesion receptors and promote changes in the expression of the genes involved in their biosynthesis. BMC Microbiol 2022; 22:65. [PMID: 35219289 PMCID: PMC8881830 DOI: 10.1186/s12866-022-02477-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2021] [Accepted: 02/16/2022] [Indexed: 11/14/2022] Open
Abstract
Background Cell surface glycosaminoglycans (GAGs) participate in many physiological and pathological processes, including infections and inflammatory response. Acne is a common chronic inflammatory skin disorder that affects the pilosebaceous unit and has a multifactorial etiology, including bacterial colonization of the hair follicle. This study aimed to investigate the participation of GAG in the adhesion of Propionibacterium acnes, Staphylococcus aureus and Staphylococcus epidermidis to keratinocytes and fibroblasts of the skin by competition experiments and cell surface removal using specific liases. The alteration in the transcription of the genes responsible for the synthesis of GAG induced by the adhesion of these bacteria was also analyzed by qRT-PCR. Results GAGs are involved in bacterial adherence to skin cells, especially fibroblasts, where chondroitin sulfate displayed the higher effect. Bacterial adherence produced different alterations in the transcription of the genes responsible for GAG structures. P. acnes induced mostly changes in keratinocytes, while S. epidermidis was the main cause of alterations in fibroblasts. These variations in gene expression affected all the stages in the biosynthesis of the main species of GAGs, heparan and chondroitin sulphate. Conclusions GAGs species are involved in the adhesion of acne-related bacteria to skin cells in a differential manner depending on each microorganism and cellular type, although other receptors seem to exist. Bacterial adherence led to variations on gene expression in skin cells affecting GAG chains structure what, consequently, should alter their interactions with different ligands, affecting the development of acne disease. Supplementary Information The online version contains supplementary material available at 10.1186/s12866-022-02477-2.
Collapse
|
17
|
Fedotov VD, Zhestkov A, Lyamin AV, Zaslavskaya M, Dobrotina I, Tulichev A. Microbiota in the pathogenesis of COPD and its impact on the course of the disease. CLINICAL MICROBIOLOGY AND ANTIMICROBIAL CHEMOTHERAPY 2022. [DOI: 10.36488/cmac.2022.3.202-212] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Chronic obstructive pulmonary disease (COPD) is a serious problem for global health. Infectious agents play a main role in the development of COPD exacerbations. Bacterial colonization of the lower respiratory tract is common in patients with stable COPD. The role of microbiota and host immune response to potential pathogens is not well studied. Microbiota composition disorders in respiratory tract are found in patients with COPD and associated with maladaptive changes in the immune system of the lungs and increased level of inflammation. This review investigates role of microbiota in the pathogenesis of COPD and its impact on the course of the disease. Some important issues such as pneumococcal vaccination and antimicrobial resistance of respiratory pathogens are also discussed.
Collapse
Affiliation(s)
| | | | | | - M.I. Zaslavskaya
- Privolzhskiy Research Medical University (Nizhny Novgorod, Russia)
| | - I.S. Dobrotina
- Privolzhskiy Research Medical University (Nizhny Novgorod, Russia)
| | - A.A. Tulichev
- Privolzhskiy Research Medical University (Nizhny Novgorod, Russia)
| |
Collapse
|
18
|
Orts DJB, Arcisio-Miranda M. Cell glycosaminoglycans content modulates human voltage-gated proton channel (H V 1) gating. FEBS J 2021; 289:2593-2612. [PMID: 34800064 DOI: 10.1111/febs.16290] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2021] [Revised: 11/01/2021] [Accepted: 11/19/2021] [Indexed: 11/28/2022]
Abstract
Voltage-gated proton channels (HV 1) have been found in many mammalian cells and play a crucial role in the immune system, male fertility, and cancer progression. Glycosaminoglycans play a significant role in various aspects of cell physiology, including the modulation of membrane receptors and ion channel function. We present here evidence that mechanosensitivity of the dimeric HV 1 channel transduce changes on cell membrane fluidity related to the defective biosynthesis of chondroitin sulfate and heparan sulfate in Chinese Hamster Ovary (CHO-745) cells into a leftward shift in the activation voltage dependence. This effect was accompanied by an increase in the H+ current, and an acceleration of the activation kinetics, under symmetrical or asymmetrical pH gradient (ΔpH) conditions. Similar gating alterations were evoked by two naturally occurring HV 1 N-terminal truncated isoforms expressed in wild-type CHO-K1 and CHO-745 cells. On three different monomeric HV 1 constructs, no alterations in the biophysical parameters were observed. Moreover, we have shown that HV 1 gating can be modulated by manipulating CHO-K1 cell membrane fluidity. Our results suggest that the defective biosynthesis of chondroitin sulfate and heparan sulfate on CHO-745 cell increases membrane fluidity and allosterically modulates the coupling between voltage- and ΔpH-sensing through the dimeric HV 1 channel.
Collapse
Affiliation(s)
- Diego J B Orts
- Departamento de Biofísica, Laboratório de Neurobiologia Estrutural e Funcional (LaNEF), Universidade Federal de São Paulo - UNIFESP, Brasil
| | - Manoel Arcisio-Miranda
- Departamento de Biofísica, Laboratório de Neurobiologia Estrutural e Funcional (LaNEF), Universidade Federal de São Paulo - UNIFESP, Brasil
| |
Collapse
|
19
|
Motta JP, Wallace JL, Buret AG, Deraison C, Vergnolle N. Gastrointestinal biofilms in health and disease. Nat Rev Gastroenterol Hepatol 2021; 18:314-334. [PMID: 33510461 DOI: 10.1038/s41575-020-00397-y] [Citation(s) in RCA: 160] [Impact Index Per Article: 40.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 11/25/2020] [Indexed: 01/30/2023]
Abstract
Microorganisms colonize various ecological niches in the human habitat, as they do in nature. Predominant forms of multicellular communities called biofilms colonize human tissue surfaces. The gastrointestinal tract is home to a profusion of microorganisms with intertwined, but not identical, lifestyles: as isolated planktonic cells, as biofilms and in biofilm-dispersed form. It is therefore of major importance in understanding homeostatic and altered host-microorganism interactions to consider not only the planktonic lifestyle, but also biofilms and biofilm-dispersed forms. In this Review, we discuss the natural organization of microorganisms at gastrointestinal surfaces, stratification of microbiota taxonomy, biogeographical localization and trans-kingdom interactions occurring within the biofilm habitat. We also discuss existing models used to study biofilms. We assess the contribution of the host-mucosa biofilm relationship to gut homeostasis and to diseases. In addition, we describe how host factors can shape the organization, structure and composition of mucosal biofilms, and how biofilms themselves are implicated in a variety of homeostatic and pathological processes in the gut. Future studies characterizing biofilm nature, physical properties, composition and intrinsic communication could shed new light on gut physiology and lead to potential novel therapeutic options for gastrointestinal diseases.
Collapse
Affiliation(s)
- Jean-Paul Motta
- Institute of Digestive Health Research, IRSD, INSERM U1220, Toulouse, France.
| | - John L Wallace
- Department of Physiology & Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada.,Antibe Therapeutics Inc., Toronto, ON, Canada
| | - André G Buret
- Department of Biological Sciences, University of Calgary, Calgary, AB, Canada
| | - Céline Deraison
- Institute of Digestive Health Research, IRSD, INSERM U1220, Toulouse, France
| | - Nathalie Vergnolle
- Institute of Digestive Health Research, IRSD, INSERM U1220, Toulouse, France. .,Department of Physiology & Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada.
| |
Collapse
|
20
|
Antiviral strategies should focus on stimulating the biosynthesis of heparan sulfates, not their inhibition. Life Sci 2021; 277:119508. [PMID: 33865880 PMCID: PMC8046744 DOI: 10.1016/j.lfs.2021.119508] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2021] [Revised: 03/31/2021] [Accepted: 04/06/2021] [Indexed: 12/23/2022]
Abstract
Antiviral strategies for viruses that utilize proteoglycan core proteins (syndecans and glypicans) as receptors should focus on heparan sulfate (HS) biosynthesis rather than on inhibition of these sugar chains. Here, we show that heparin and certain xylosides, which exhibit in vitro viral entry inhibitory properties against HSV-1, HSV-2, HPV-16, HPV-31, HVB, HVC, HIV-1, HTLV-1, SARS-CoV-2, HCMV, DENV-1, and DENV-2, stimulated HS biosynthesis at the cell surface 2- to 3-fold for heparin and up to 10-fold for such xylosides. This is consistent with the hypothesis from a previous study that for core protein attachment, viruses are glycosylated at HS attachment sites (i.e., serine residues intended to receive the D-xylose molecule for initiating HS chains). Heparanase overexpression, endocytic entry, and syndecan shedding enhancement, all of which are observed during viral infection, lead to glycocalyx deregulation and appear to be direct consequences of this hypothesis. In addition to the appearance of type 2 diabetes and the degradation of HS observed during viral infection, we linked this hypothesis to that proposed in a previous publication.
Collapse
|
21
|
Important Roles of Oligo- and Polysaccharides against SARS-CoV-2: Recent Advances. APPLIED SCIENCES-BASEL 2021. [DOI: 10.3390/app11083512] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
The severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2)-initiated outbreak of COVID-19 has spread rapidly around the world, posing a huge threat to public health. Natural oligo- and polysaccharides with low toxicity, good sustainability, high biocompatibility, respectable safety, immune regulation, and antiviral activity can be employed as promising candidates for the prevention and inhibition of viral infections, especially COVID-19. Glycosaminoglycans, marine polysaccharides, terrestrial plant polysaccharides, and some others have exhibited potential antiviral activity against pathogenic viruses, in the format of polysaccharide-centered vaccine adjuvants, nano-based structures, drug conveyance platforms, etc. In this review, significant recent advancements pertaining to the antiviral applications of oligo- and polysaccharides against SARS-CoV-2 are highlighted, including important challenges and future perspectives.
Collapse
|
22
|
Aragona P, Baudouin C, Benitez Del Castillo JM, Messmer E, Barabino S, Merayo-Lloves J, Brignole-Baudouin F, Inferrera L, Rolando M, Mencucci R, Rescigno M, Bonini S, Labetoulle M. The ocular microbiome and microbiota and their effects on ocular surface pathophysiology and disorders. Surv Ophthalmol 2021; 66:907-925. [PMID: 33819460 DOI: 10.1016/j.survophthal.2021.03.010] [Citation(s) in RCA: 64] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2020] [Revised: 03/25/2021] [Accepted: 03/26/2021] [Indexed: 12/17/2022]
Abstract
The ocular surface flora perform an important role in the defense mechanisms of the ocular surface system. Its regulation of the immunological activity and the barrier effect against pathogen invasion are remarkable. Composition of the flora differs according to the methods of investigation, because the microbiome, composed of the genetic material of bacteria, fungi, viruses, protozoa, and eukaryotes on the ocular surface, differs from the microbiota, which are the community of microorganisms that colonize the ocular surface. The observed composition of the ocular surface flora depends on harvesting and examining methods, whether with traditional culture or with more refined genetic analysis based on rRNA and DNA sequencing. Environment, diet, sex, and age influence the microbial flora composition, thus complicating the analysis of the baseline status. Moreover, potentially pathogenic organisms can affect its composition, as do various disorders, including chronic inflammation, and therapies applied to the ocular surface. A better understanding of the composition and function of microbial communities at the ocular surface could bring new insights and clarify the epidemiology and pathology of ocular surface dynamics in health and disease. The purpose of this review is to provide an up-to-date overview of knowledge about this topic.
Collapse
Affiliation(s)
- Pasquale Aragona
- Department of Biomedical Sciences, Ophthalmology Clinic, University of Messina, Messina, Italy.
| | - Christophe Baudouin
- Quinze-Vingts National Eye Hospital, IHU ForeSight, Paris Saclay University, Paris, France
| | - Jose M Benitez Del Castillo
- Departamento de Oftalmología, Hospital Clínico San Carlos, Clínica Rementeria, Instituto Investigaciones Oftalmologicas Ramon Castroviejo, Universidad Complutense, Madrid, Spain
| | - Elisabeth Messmer
- Department of Ophthalmology, Ludwig-Maximilians-University, Munich, Germany
| | - Stefano Barabino
- Ocular Surface and Dry Eye Center, Ospedale L. Sacco, University of Milan, Milan, Italy
| | - Jesus Merayo-Lloves
- Instituto Universitario Fernández-Vega, Universidad de Oviedo, Oviedo, Spain
| | - Francoise Brignole-Baudouin
- Sorbonne Université, INSERM UMR_S968, CNRS UMR7210, Institut de la Vision, Paris, France; CHNO des Quinze-Vingts, INSERM-DGOS CIC 1423, Laboratoire de Biologie Médicale, Paris, France; Université de Paris, Faculté de Pharmacie de Paris, Département de Chimie-Toxicologie Analytique et Cellulaire, Paris, France
| | - Leandro Inferrera
- Department of Biomedical Sciences, Ophthalmology Clinic, University of Messina, Messina, Italy
| | - Maurizio Rolando
- Ocular Surface and Dry Eye Center, ISPRE Ophthalmics, Genoa, Italy
| | - Rita Mencucci
- Department of Neuroscience, Psychology, Pharmacology and Child Health (NEUROFARBA), Eye Clinic, University of Florence, Florence, Italy
| | - Maria Rescigno
- Humanitas Clinical and Research Center - IRCCS, Humanitas University Department of Biomedical Sciences, Milan, Italy
| | - Stefano Bonini
- Department of Ophthalmology, University of Rome Campus Biomedico, Rome, Italy
| | - Marc Labetoulle
- Ophthalmology Départment, Hôpitaux Universitaires Paris-Sud, APHP, Université Paris-Saclay, IDMIT Infrastructure, Fontenay-aux-Roses Cedex, France
| |
Collapse
|
23
|
Song N, Chen L, Ren X, Waterfield NR, Yang J, Yang G. N-Glycans and sulfated glycosaminoglycans contribute to the action of diverse Tc toxins on mammalian cells. PLoS Pathog 2021; 17:e1009244. [PMID: 33539469 PMCID: PMC7861375 DOI: 10.1371/journal.ppat.1009244] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2020] [Accepted: 12/18/2020] [Indexed: 01/11/2023] Open
Abstract
Tc toxin is an exotoxin composed of three subunits named TcA, TcB and TcC. Structural analysis revealed that TcA can form homopentamer that mediates the cellular recognition and delivery processes, thus contributing to the host tropism of Tc toxin. N-glycans and heparan sulfates have been shown to act as receptors for several Tc toxins. Here, we performed two independent genome-wide CRISPR-Cas9 screens, and have validated glycans and sulfated glycosaminoglycans (sGAGs) as Tc toxin receptors also for previously uncharacterized Tc toxins. We found that TcdA1 form Photorhabdus luminescens W14 (TcdA1W14) can recognize N-glycans via the RBD-D domain, corroborating previous findings. Knockout of N-glycan processing enzymes specifically blocks the intoxication of TcdA1W14-assembled Tc toxin. On the other hand, our results showed that sGAG biosynthesis pathway is involved in the cell surface binding of TcdA2TT01 (TcdA2 from P. luminescens TT01). Competition assays and biolayer interferometry demonstrated that the sulfation group in sGAGs is required for the binding of TcdA2TT01. Finally, based on the conserved domains of representative TcA proteins, we have identified 1,189 putative TcAs from 1,039 bacterial genomes. These TcAs are categorized into five subfamilies. Each subfamily shows a good correlation with both genetic organization of the TcA protein(s) and taxonomic origin of the genomes, suggesting these subfamilies may utilize different mechanisms for cellular recognition. Taken together, our results support the previously described two different binding modalities of Tc toxins, leading to unique host targeting properties. We also present the bioinformatics data and receptor screening strategies for TcA proteins, provide new insights into understanding host specificity and biomedical applications of Tc toxins. The Toxin complexes, also referred to as Tc toxins, are a family of A5BC exotoxins widely distributed among Gram-negative and positive bacteria. First identified in Entomopathogenic bacteria as key virulence factors to combat insect hosts, putative Tc toxin loci are also encoded by a range of human pathogens such as Salmonella and Yersinia. Previous studies indicated that several Tc toxins can target invertebrate and vertebrate cells via binding with N-glycans and heparan sulfates. Here our genome-wide CRISPR-Cas9 screens validated that different Tc toxins utilized distinct receptors for the adhesion to their targets, which is determined by TcA homopentamer. For example, TcdA1 from Photorhabdus luminescens W14 (TcdA1W14) relies on N-glycan binding to exert its toxic effects, while sulfate groups of sulfated glycosaminoglycans are critical for the cell targeting of other TcAs such as TcdA2TT01 (TcdA2 from P. luminescens TT01). Consistent with the previously described different binding modalities of Tc toxins, our results confirm that the receptor selectivity of TcAs contribute to the cellular tropism of Tc toxins. Furthermore we has also identified 1,189 TcA homologues and categorized them into five subfamilies. Each TcA subfamily shows a good correlation with the taxonomic origin of the genomes, suggesting these subfamilies are linked to diverse host tropisms via different binding modalities. Together, our findings provide mechanistic insights into understanding host specificity of distinct Tc toxins and the development of therapeutics for Tc toxin-related infections, as well as the adaptation of Tc-injectisomes as potential biotechnology tools and pest-control weapons.
Collapse
Affiliation(s)
- Nan Song
- Beijing Institute of Tropical Medicine, Beijing Friendship Hospital, Capital Medical University, Beijing, China
- Emergency and Critical Care Center, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Lihong Chen
- NHC Key Laboratory of Systems Biology of Pathogens, Institute of Pathogen Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Xingmei Ren
- Beijing Institute of Tropical Medicine, Beijing Friendship Hospital, Capital Medical University, Beijing, China
- Emergency and Critical Care Center, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | | | - Jian Yang
- NHC Key Laboratory of Systems Biology of Pathogens, Institute of Pathogen Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Guowei Yang
- Beijing Institute of Tropical Medicine, Beijing Friendship Hospital, Capital Medical University, Beijing, China
- Emergency and Critical Care Center, Beijing Friendship Hospital, Capital Medical University, Beijing, China
- * E-mail:
| |
Collapse
|
24
|
GAG-DB, the New Interface of the Three-Dimensional Landscape of Glycosaminoglycans. Biomolecules 2020; 10:biom10121660. [PMID: 33322545 PMCID: PMC7763844 DOI: 10.3390/biom10121660] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2020] [Revised: 12/03/2020] [Accepted: 12/09/2020] [Indexed: 12/18/2022] Open
Abstract
Glycosaminoglycans (GAGs) are complex linear polysaccharides. GAG-DB is a curated database that classifies the three-dimensional features of the six mammalian GAGs (chondroitin sulfate, dermatan sulfate, heparin, heparan sulfate, hyaluronan, and keratan sulfate) and their oligosaccharides complexed with proteins. The entries are structures of GAG and GAG-protein complexes determined by X-ray single-crystal diffraction methods, X-ray fiber diffractometry, solution NMR spectroscopy, and scattering data often associated with molecular modeling. We designed the database architecture and the navigation tools to query the database with the Protein Data Bank (PDB), UniProtKB, and GlyTouCan (universal glycan repository) identifiers. Special attention was devoted to the description of the bound glycan ligands using simple graphical representation and numerical format for cross-referencing to other databases in glycoscience and functional data. GAG-DB provides detailed information on GAGs, their bound protein ligands, and features their interactions using several open access applications. Binding covers interactions between monosaccharides and protein monosaccharide units and the evaluation of quaternary structure. GAG-DB is freely available.
Collapse
|
25
|
De Caluwé L, Ariën KK, Bartholomeeusen K. Host Factors and Pathways Involved in the Entry of Mosquito-Borne Alphaviruses. Trends Microbiol 2020; 29:634-647. [PMID: 33208275 DOI: 10.1016/j.tim.2020.10.011] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2020] [Revised: 10/22/2020] [Accepted: 10/23/2020] [Indexed: 11/17/2022]
Abstract
Chikungunya virus (CHIKV) is an arthropod-borne virus that has re-emerged recently and has spread to previously unaffected regions, resulting in millions of infections worldwide. The genus Alphavirus, in the family Togaviridae, contains several members with a similar potential for epidemic emergence. In order for CHIKV to replicate in targeted cell types it is essential for the virus to enter these cells. In this review, we summarize our current understanding of the versatile and promiscuous steps in CHIKV binding and entry into human and mosquito host cells. We describe the different entry pathways, receptors, and attachment factors so far described for CHIKV and other mosquito-borne alphaviruses and discuss them in the context of tissue tropism and potential therapeutic targeting.
Collapse
Affiliation(s)
- Lien De Caluwé
- Virology Unit, Biomedical Sciences, Institute of Tropical Medicine Antwerp, Antwerp, Belgium
| | - Kevin K Ariën
- Virology Unit, Biomedical Sciences, Institute of Tropical Medicine Antwerp, Antwerp, Belgium; Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium.
| | - Koen Bartholomeeusen
- Virology Unit, Biomedical Sciences, Institute of Tropical Medicine Antwerp, Antwerp, Belgium.
| |
Collapse
|
26
|
Hyatt JG, Prévost S, Devos JM, Mycroft-West CJ, Skidmore MA, Winter A. Molecular Changes in Dengue Envelope Protein Domain III upon Interaction with Glycosaminoglycans. Pathogens 2020; 9:pathogens9110935. [PMID: 33187224 PMCID: PMC7697694 DOI: 10.3390/pathogens9110935] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2020] [Revised: 11/06/2020] [Accepted: 11/08/2020] [Indexed: 11/16/2022] Open
Abstract
Dengue fever is a rapidly emerging vector-borne viral disease with a growing global burden of approximately 390 million new infections per annum. The Dengue virus (DENV) is a flavivirus spread by female mosquitos of the aedes genus, but the mechanism of viral endocytosis is poorly understood at a molecular level, preventing the development of effective transmission blocking vaccines (TBVs). Recently, glycosaminoglycans (GAGs) have been identified as playing a role during initial viral attachment through interaction with the third domain of the viral envelope protein (EDIII). Here, we report a systematic study investigating the effect of a range of biologically relevant GAGs on the structure and oligomeric state of recombinantly generated EDIII. We provide novel in situ biophysical evidence that heparin and chondroitin sulphate C induce conformational changes in EDIII at the secondary structure level. Furthermore, we report the ability of chondroitin sulphate C to bind EDIII and induce higher-order dynamic molecular changes at the tertiary and quaternary structure levels which are dependent on pH, GAG species, and the GAG sulphation state. Lastly, we conducted ab initio modelling of Small Angle Neutron Scattering (SANS) data to visualise the induced oligomeric state of EDIII caused by interaction with chondroitin sulphate C, which may aid in TBV development.
Collapse
Affiliation(s)
- James G. Hyatt
- School of Life Sciences, Keele University, Huxley Building, Keele, Staffordshire ST5 5BG, UK; (J.G.H.); (C.J.M.-W.); (M.A.S.)
| | - Sylvain Prévost
- Large Scale Structures Group, Institut Laue-Langevin, 71 avenue des Martyrs, CS 20156, 38042 Grenoble CEDEX 9, France;
| | - Juliette M. Devos
- Life Sciences Group, Institut Laue-Langevin, 71 avenue des Martyrs, CS 20156, 38042 Grenoble CEDEX 9, France;
| | - Courtney J. Mycroft-West
- School of Life Sciences, Keele University, Huxley Building, Keele, Staffordshire ST5 5BG, UK; (J.G.H.); (C.J.M.-W.); (M.A.S.)
| | - Mark A. Skidmore
- School of Life Sciences, Keele University, Huxley Building, Keele, Staffordshire ST5 5BG, UK; (J.G.H.); (C.J.M.-W.); (M.A.S.)
| | - Anja Winter
- School of Life Sciences, Keele University, Huxley Building, Keele, Staffordshire ST5 5BG, UK; (J.G.H.); (C.J.M.-W.); (M.A.S.)
- Life Sciences Group, Institut Laue-Langevin, 71 avenue des Martyrs, CS 20156, 38042 Grenoble CEDEX 9, France;
- Correspondence: ; Tel.: +44-01782-7-33117
| |
Collapse
|
27
|
Vallet SD, Clerc O, Ricard-Blum S. Glycosaminoglycan-Protein Interactions: The First Draft of the Glycosaminoglycan Interactome. J Histochem Cytochem 2020; 69:93-104. [PMID: 32757871 DOI: 10.1369/0022155420946403] [Citation(s) in RCA: 120] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
The six mammalian glycosaminoglycans (GAGs), chondroitin sulfate, dermatan sulfate, heparin, heparan sulfate, hyaluronan, and keratan sulfate, are linear polysaccharides. Except for hyaluronan, they are sulfated to various extent, and covalently attached to proteins to form proteoglycans. GAGs interact with growth factors, morphogens, chemokines, extracellular matrix proteins and their bioactive fragments, receptors, lipoproteins, and pathogens. These interactions mediate their functions, from embryonic development to extracellular matrix assembly and regulation of cell signaling in various physiological and pathological contexts such as angiogenesis, cancer, neurodegenerative diseases, and infections. We give an overview of GAG-protein interactions (i.e., specificity and chemical features of GAG- and protein-binding sequences), and review the available GAG-protein interaction networks. We also provide the first comprehensive draft of the GAG interactome composed of 832 biomolecules (827 proteins and five GAGs) and 932 protein-GAG interactions. This network is a scaffold, which in the future should integrate structures of GAG-protein complexes, quantitative data of the abundance of GAGs in tissues to build tissue-specific interactomes, and GAG interactions with metal ions such as calcium, which plays a major role in the assembly of the extracellular matrix and its interactions with cells. This contextualized interactome will be useful to identify druggable GAG-protein interactions for therapeutic purpose.
Collapse
Affiliation(s)
- Sylvain D Vallet
- Univ Lyon, University Claude Bernard Lyon 1, CNRS, INSA Lyon, CPE, Institute of Molecular and Supramolecular Chemistry and Biochemistry, UMR 5246, Villeurbanne Cedex, France
| | - Olivier Clerc
- Univ Lyon, University Claude Bernard Lyon 1, CNRS, INSA Lyon, CPE, Institute of Molecular and Supramolecular Chemistry and Biochemistry, UMR 5246, Villeurbanne Cedex, France
| | - Sylvie Ricard-Blum
- Univ Lyon, University Claude Bernard Lyon 1, CNRS, INSA Lyon, CPE, Institute of Molecular and Supramolecular Chemistry and Biochemistry, UMR 5246, Villeurbanne Cedex, France
| |
Collapse
|
28
|
García-Sánchez M, Jiménez-Pelayo L, Horcajo P, Collantes-Fernández E, Ortega-Mora LM, Regidor-Cerrillo J. Neospora caninum infection induces an isolate virulence-dependent pro-inflammatory gene expression profile in bovine monocyte-derived macrophages. Parasit Vectors 2020; 13:374. [PMID: 32711550 PMCID: PMC7382829 DOI: 10.1186/s13071-020-04239-3] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2020] [Accepted: 07/15/2020] [Indexed: 01/22/2023] Open
Abstract
Background Neospora caninum is an obligate intracellular parasite, and its ability to survive inside host immune cells may be a key mechanism for the establishment of infection in cattle. In vitro studies carried out by our group have shown that N. caninum is able to replicate in bovine macrophages (MØs), alter their microbicidal mechanisms and exploit their motility. Furthermore, host-cell control seems to be isolate virulence-dependent. Methods To investigate the molecular basis underlying the innate responses in MØs against N. caninum and the mechanisms of parasite manipulation of the host cell environment, the transcriptome profile of bovine monocyte-derived MØs infected with high-virulence (Nc-Spain7) or low-virulence (Nc-Spain1H) N. caninum isolates was studied. Results Functional enrichment revealed upregulation of genes involved in chemokine signalling, inflammation, cell survival, and inhibition of genes related with metabolism and phagolysosome formation. MØs activation was characterized by the induction of a predominantly M1 phenotype with expression of TLR2, TLR3 and TLR9 and activation of the NF-ƙB signalling pathway. Heat-killed N. caninum tachyzoites failed to activate NF-ƙB, and to inhibit lysosomal activity and apoptosis, which indicates active modulation by the parasite. The FoxO signalling pathway, Th1-Th2 differentiation, glycosaminoglycan degradation and apoptosis were pathways enriched only for low virulent Nc-Spain1H infection. In addition, Nc-Spain1H infection upregulated the IL12A and IL8 pro-inflammatory cytokines, whereas IL23 was downregulated by high virulent Nc-Spain7. Conclusions This study revealed mechanisms implicated in the recognition of N. caninum by bovine MØs and in the development of the subsequent immune response. NF-ƙB seems to be the main signalling pathway implicated in the pro-inflammatory bovine MØs response against this pathogen. Apoptosis and phagolysosome maturation are processes repressed by N. caninum infection, which may guarantee its intracellular survival. The results also indicate that Nc-Spain7 may be able to partially circumvent the pro-inflammatory response whereas Nc-Spain1H induces a protective response to infection, which may explain the more efficient transmission of the high-virulence Nc-Spain7 isolate observed in vivo.![]()
Collapse
Affiliation(s)
- Marta García-Sánchez
- Saluvet, Animal Health Department, Faculty of Veterinary Sciences, Complutense University of Madrid, Ciudad Universitaria s/n, 28040, Madrid, Spain
| | - Laura Jiménez-Pelayo
- Saluvet, Animal Health Department, Faculty of Veterinary Sciences, Complutense University of Madrid, Ciudad Universitaria s/n, 28040, Madrid, Spain
| | - Pilar Horcajo
- Saluvet, Animal Health Department, Faculty of Veterinary Sciences, Complutense University of Madrid, Ciudad Universitaria s/n, 28040, Madrid, Spain
| | - Esther Collantes-Fernández
- Saluvet, Animal Health Department, Faculty of Veterinary Sciences, Complutense University of Madrid, Ciudad Universitaria s/n, 28040, Madrid, Spain
| | - Luis Miguel Ortega-Mora
- Saluvet, Animal Health Department, Faculty of Veterinary Sciences, Complutense University of Madrid, Ciudad Universitaria s/n, 28040, Madrid, Spain.
| | - Javier Regidor-Cerrillo
- Saluvet, Animal Health Department, Faculty of Veterinary Sciences, Complutense University of Madrid, Ciudad Universitaria s/n, 28040, Madrid, Spain. .,Saluvet-Innova, Faculty of Veterinary Sciences, Complutense University of Madrid, Ciudad Universitaria s/n, 28040, Madrid, Spain.
| |
Collapse
|
29
|
Vignovich WP, Pomin VH. Saturation Transfer Difference in Characterization of Glycosaminoglycan-Protein Interactions. SLAS Technol 2020; 25:307-319. [PMID: 32452261 DOI: 10.1177/2472630320921130] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Novel methods in nuclear magnetic resonance (NMR) spectroscopy have recently been developed to investigate the binding properties of intermolecular complexes endowed with biomedical functions. Among these methods is the saturation transfer difference (STD), which enables the mapping of specific binding motifs of functional ligands. STD can efficiently uncover the specific and preferential binding sites of these ligands in their intermolecular complexes. This is particularly useful in the case of glycosaminoglycans (GAGs), a group of sulfated polysaccharides that play pivotal roles in various biological and pathological processes. The activity of GAGs is ultimately mediated through molecular interactions with key functional proteins, namely, GAG-binding proteins (GBPs). The quality of the GAG-GBP interactions depends on sulfation patterns, oligosaccharide length, and the composing monosaccharides of GAGs. Through STD NMR, information about the atoms of the GAG ligands involved in the complexes is provided. Here we highlight the latest achievements of the literature using STD NMR on GAG oligosaccharide-GBP complexes. Interestingly, most of the GBPs studied so far by STD NMR belong to one of the three major classes: coagulation factors, growth factors, or chemokine/cytokines. Unveiling the structural requirements of GAG ligands in bindings with their protein partners is a crucial step to understand the biochemical and medical actions of GAGs. This process is also a requirement in GAG-based drug discovery and development.
Collapse
Affiliation(s)
- William P Vignovich
- BioMolecular Sciences Department, School of Pharmacy, the University of Mississippi, Oxford, MS, USA
| | - Vitor H Pomin
- BioMolecular Sciences Department, School of Pharmacy, the University of Mississippi, Oxford, MS, USA.,Research Institute of Pharmaceutical Sciences, School of Pharmacy, the University of Mississippi, Oxford, MS, USA
| |
Collapse
|
30
|
Afosah DK, Al-Horani RA. Sulfated Non-Saccharide Glycosaminoglycan Mimetics as Novel Drug Discovery Platform for Various Pathologies. Curr Med Chem 2020; 27:3412-3447. [PMID: 30457046 PMCID: PMC6551317 DOI: 10.2174/0929867325666181120101147] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2018] [Revised: 11/06/2018] [Accepted: 11/13/2018] [Indexed: 01/14/2023]
Abstract
Glycosaminoglycans (GAGs) are very complex, natural anionic polysaccharides. They are polymers of repeating disaccharide units of uronic acid and hexosamine residues. Owing to their template-free, spatiotemporally-controlled, and enzyme-mediated biosyntheses, GAGs possess enormous polydispersity, heterogeneity, and structural diversity which often translate into multiple biological roles. It is well documented that GAGs contribute to physiological and pathological processes by binding to proteins including serine proteases, serpins, chemokines, growth factors, and microbial proteins. Despite advances in the GAG field, the GAG-protein interface remains largely unexploited by drug discovery programs. Thus, Non-Saccharide Glycosaminoglycan Mimetics (NSGMs) have been rationally developed as a novel class of sulfated molecules that modulate GAG-protein interface to promote various biological outcomes of substantial benefit to human health. In this review, we describe the chemical, biochemical, and pharmacological aspects of recently reported NSGMs and highlight their therapeutic potentials as structurally and mechanistically novel anti-coagulants, anti-cancer agents, anti-emphysema agents, and anti-viral agents. We also describe the challenges that complicate their advancement and describe ongoing efforts to overcome these challenges with the aim of advancing the novel platform of NSGMs to clinical use.
Collapse
Affiliation(s)
- Daniel K. Afosah
- Department of Medicinal Chemistry and Institute for Structural Biology, Drug Discovery and Development, Virginia Commonwealth University, Richmond, Virginia 23219
| | - Rami A. Al-Horani
- Division of Basic Pharmaceutical Sciences, College of Pharmacy, Xavier University of Louisiana, New Orleans, Louisiana 70125
| |
Collapse
|
31
|
Cerezo-Magaña M, Bång-Rudenstam A, Belting M. The pleiotropic role of proteoglycans in extracellular vesicle mediated communication in the tumor microenvironment. Semin Cancer Biol 2019; 62:99-107. [PMID: 31276785 DOI: 10.1016/j.semcancer.2019.07.001] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2019] [Revised: 07/01/2019] [Accepted: 07/02/2019] [Indexed: 12/21/2022]
Abstract
Compartmental exchange between cells through extracellular vesicles (EVs), including exosomes and microvesicles, has emerged as a central mechanism that coordinates the complex communication between malignant and stromal cells during tumor initiation and evolution. Some of the most critical processes of EV-mediated communication, including EV biogenesis and EV uptake, can be mediated by heparan sulfate proteoglycans (HSPGs) that reside on the surface of producer and recipient cells as well as on EVs. With interestingly similar, HSPG-dependent, pathways as the ones exploited by some viruses, EVs may, in an evolutionary perspective, be viewed as endogenous counterparts of viral particles. Cancer cell-derived EVs exert their protumorigenic effects by direct interactions of biologically active surface molecules, by transfer of proteins and nucleic acids into recipient cells or by transfer of metabolites that can be utilized as an energy source by the recipient cell. Here, we discuss the pleiotropic role of the HSPG family in these different contexts of EV communication with a specific focus on tumor development. We propose EV-associated PGs as dynamic reservoirs and chaperones of signaling molecules with potential implications in ligand exchange between EVs and tumor target cells. The protumorigenic consequences of EV mediated communication through HSPG should motivate the development of therapeutic approaches targeting EV-HSPG interactions as a novel strategy in cancer treatment.
Collapse
Affiliation(s)
- M Cerezo-Magaña
- Department of Clinical Sciences, Lund, Section of Oncology and Pathology, Lund University, Lund, Sweden
| | - A Bång-Rudenstam
- Department of Clinical Sciences, Lund, Section of Oncology and Pathology, Lund University, Lund, Sweden
| | - M Belting
- Department of Clinical Sciences, Lund, Section of Oncology and Pathology, Lund University, Lund, Sweden; Department of Hematology, Oncology and Radiophysics, Skåne University Hospital, Lund, Sweden; Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden.
| |
Collapse
|
32
|
A comparative analysis of secreted protein disulfide isomerases from the tropical co-endemic parasites Schistosoma mansoni and Leishmania major. Sci Rep 2019; 9:9568. [PMID: 31267027 PMCID: PMC6606611 DOI: 10.1038/s41598-019-45709-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2019] [Accepted: 06/10/2019] [Indexed: 12/19/2022] Open
Abstract
The human parasites Schistosoma mansoni and Leishmania major are co-endemic and a major threat to human health. Though displaying different tissue tropisms, they excrete/secrete similar subsets of intracellular proteins that, interacting with the host extracellular matrix (ECM), help the parasites invading the host. We selected one of the most abundant proteins found in the secretomes of both parasites, protein disulfide isomerase (PDI), and performed a comparative screening with surface plasmon resonance imaging (SPRi), looking for ECM binding partners. Both PDIs bind heparan sulfate; none of them binds collagens; each of them binds further ECM components, possibly linked to the different tropisms. We investigated by small-angle X-ray scattering both PDIs structures and those of a few complexes with host partners, in order to better understand the differences within this conserved family fold. Furthermore, we highlighted a previously undisclosed moonlighting behaviour of both PDIs, namely a concentration-dependent switch of function from thiol-oxidoreductase to holdase. Finally, we have tried to exploit the differences to look for possible compounds able to interfere with the redox activity of both PDI.
Collapse
|
33
|
A Bioinformatics View of Glycan⁻Virus Interactions. Viruses 2019; 11:v11040374. [PMID: 31018588 PMCID: PMC6521074 DOI: 10.3390/v11040374] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2019] [Revised: 04/05/2019] [Accepted: 04/15/2019] [Indexed: 02/06/2023] Open
Abstract
Evidence of the mediation of glycan molecules in the interaction between viruses and their hosts is accumulating and is now partially reflected in several online databases. Bioinformatics provides convenient and efficient means of searching, visualizing, comparing, and sometimes predicting, interactions in numerous and diverse molecular biology applications related to the -omics fields. As viromics is gaining momentum, bioinformatics support is increasingly needed. We propose a survey of the current resources for searching, visualizing, comparing, and possibly predicting host–virus interactions that integrate the presence and role of glycans. To the best of our knowledge, we have mapped the specialized and general-purpose databases with the appropriate focus. With an illustration of their potential usage, we also discuss the strong and weak points of the current bioinformatics landscape in the context of understanding viral infection and the immune response to it.
Collapse
|
34
|
Kalaska B, Miklosz J, Kamiński K, Musielak B, Yusa SI, Pawlak D, Nowakowska M, Szczubiałka K, Mogielnicki A. The neutralization of heparan sulfate by heparin-binding copolymer as a potential therapeutic target. RSC Adv 2019; 9:3020-3029. [PMID: 35518950 PMCID: PMC9059929 DOI: 10.1039/c8ra09724k] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2018] [Accepted: 01/15/2019] [Indexed: 12/12/2022] Open
Abstract
Besides regulating ligand–receptor and cell–cell interactions, heparan sulfate (HS) may participate in the development of many diseases, such as cancer, bacterial or viral infections, and their complications, like bleeding or inflammation. In these cases, the neutralization of HS could be a potential therapeutic target. The heparin-binding copolymer (HBC, PEG41-PMAPTAC53) was previously reported by us as a fully synthetic compound for efficient and safe neutralization of heparins and synthetic anticoagulants. In a search for molecular antagonists of HS, we examined the activity of HBC as an HS inhibitor both in vitro and in vivo and characterized HBC/HS complexes. Using a colorimetric Azure A method, isothermal titration calorimetry and dynamic light scattering techniques we found that HBC binds HS by forming complexes below 200 nm with less than 1 : 1 stoichiometry. We confirmed the HBC inhibitory effect in rats by measuring activated partial thromboplastin time, prothrombin time, anti-factor Xa activity, anti-factor IIa activity, and platelet aggregation. HBC reversed the enhancement of all tested parameters caused by HS demonstrating that cationic synthetic block copolymers may have a therapeutic value in various disorders involving overproduction of HS. The neutralization of heparan sulfate (HS) by a heparin-binding copolymer (HBC) could be a promising treating option for bacterial or viral infections or bleeding related to overproduction of HS in cancer or other diseases.![]()
Collapse
Affiliation(s)
- Bartlomiej Kalaska
- Department of Pharmacodynamics
- Medical University of Bialystok
- 15-089 Bialystok
- Poland
| | - Joanna Miklosz
- Department of Pharmacodynamics
- Medical University of Bialystok
- 15-089 Bialystok
- Poland
| | - Kamil Kamiński
- Faculty of Chemistry
- Jagiellonian University
- 30-387 Krakow
- Poland
| | - Bogdan Musielak
- Faculty of Chemistry
- Jagiellonian University
- 30-387 Krakow
- Poland
| | - Shin-Ichi Yusa
- Department of Applied Chemistry
- Graduate School of Engineering
- University of Hyogo
- Himeji
- Japan
| | - Dariusz Pawlak
- Department of Pharmacodynamics
- Medical University of Bialystok
- 15-089 Bialystok
- Poland
| | | | | | - Andrzej Mogielnicki
- Department of Pharmacodynamics
- Medical University of Bialystok
- 15-089 Bialystok
- Poland
| |
Collapse
|
35
|
Kaltenbach DD, Jaishankar D, Hao M, Beer JC, Volin MV, Desai UR, Tiwari V. Sulfotransferase and Heparanase: Remodeling Engines in Promoting Virus Infection and Disease Development. Front Pharmacol 2018; 9:1315. [PMID: 30555321 PMCID: PMC6282075 DOI: 10.3389/fphar.2018.01315] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2018] [Accepted: 10/29/2018] [Indexed: 01/08/2023] Open
Abstract
An extraordinary binding site generated in heparan sulfate (HS) structures, during its biosynthesis, provides a unique opportunity to interact with multiple protein ligands including viral proteins, and therefore adds tremendous value to this master molecule. An example of such a moiety is the sulfation at the C3 position of glucosamine residues in HS chain via 3-O sulfotransferase (3-OST) enzymes, which generates a unique virus-cell fusion receptor during herpes simplex virus (HSV) entry and spread. Emerging evidence now suggests that the unique patterns in HS sulfation assist multiple viruses in invading host cells at various steps of their life cycles. In addition, sulfated-HS structures are known to assist in invading host defense mechanisms and initiating multiple inflammatory processes; a critical event in the disease development. All these processes are detrimental for the host and therefore raise the question of how HS-sulfation is regulated. Epigenetic modulations have been shown to be implicated in these reactions during HSV infection as well as in HS modifying enzyme sulfotransferases, and therefore pose a critical component in answering it. Interestingly, heparanase (HPSE) activity is shown to be upregulated during virus infection and multiple other diseases assisting in virus replication to promote cell and tissue damage. These phenomena suggest that sulfotransferases and HPSE serve as key players in extracellular matrix remodeling and possibly generating unique signatures in a given disease. Therefore, identifying the epigenetic regulation of OST genes, and HPSE resulting in altered yet specific sulfation patterns in HS chain during virus infection, will be a significant a step toward developing potential diagnostic markers and designing novel therapies.
Collapse
Affiliation(s)
- Dominik D Kaltenbach
- Department of Biomedical Sciences, College of Graduate Studies, Midwestern University, Downers Grove, IL, United States
| | - Dinesh Jaishankar
- Department of Ophthalmology & Visual Sciences, University of Illinois at Chicago, Chicago, IL, United States
| | - Meng Hao
- Chicago College of Pharmacy, Midwestern University, Downers Grove, IL, United States
| | - Jacob C Beer
- Chicago College of Osteopathic Medicine, Midwestern University, Downers Grove, IL, United States
| | - Michael V Volin
- Department of Microbiology & Immunology, College of Graduate Studies, Midwestern University, Downers Grove, IL, United States
| | - Umesh R Desai
- Department of Medicinal Chemistry and Institute for Structural Biology, Drug Discovery and Development, Virginia Commonwealth University, Richmond, VA, United States
| | - Vaibhav Tiwari
- Department of Microbiology & Immunology, College of Graduate Studies, Midwestern University, Downers Grove, IL, United States
| |
Collapse
|
36
|
Lentz CS, Sheldon JR, Crawford LA, Cooper R, Garland M, Amieva MR, Weerapana E, Skaar EP, Bogyo M. Identification of a S. aureus virulence factor by activity-based protein profiling (ABPP). Nat Chem Biol 2018; 14:609-617. [PMID: 29769740 PMCID: PMC6202179 DOI: 10.1038/s41589-018-0060-1] [Citation(s) in RCA: 73] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2017] [Accepted: 03/27/2018] [Indexed: 12/22/2022]
Abstract
Serine hydrolases play diverse roles in regulating host-pathogen interactions in a number of organisms, yet few have been characterized in the human pathogen Staphylococcus aureus. Here we describe a chemical proteomic screen that identified ten previously uncharacterized S. aureus serine hydrolases that mostly lack human homologs. We termed these enzymes fluorophosphonate-binding hydrolases (FphA-J). One hydrolase, FphB, can process short fatty acid esters, exhibits increased activity in response to host cell factors, is located predominantly on the bacterial cell surface in a subset of cells, and is concentrated in the division septum. Genetic disruption of fphB confirmed that the enzyme is dispensable for bacterial growth in culture but crucial for establishing infection in distinct sites in vivo. A selective small molecule inhibitor of FphB effectively reduced infectivity in vivo, suggesting that it may be a viable therapeutic target for the treatment or management of Staphylococcus infections.
Collapse
Affiliation(s)
- Christian S Lentz
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
| | - Jessica R Sheldon
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Lisa A Crawford
- Department of Chemistry, Boston College, Chestnut Hill, MA, USA
| | - Rachel Cooper
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, USA
| | - Megan Garland
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
| | - Manuel R Amieva
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, USA
- Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA, USA
| | | | - Eric P Skaar
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Matthew Bogyo
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA.
- Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA, USA.
| |
Collapse
|
37
|
Heparinase Is Essential for Pseudomonas aeruginosa Virulence during Thermal Injury and Infection. Infect Immun 2017; 86:IAI.00755-17. [PMID: 29061710 DOI: 10.1128/iai.00755-17] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2017] [Accepted: 10/18/2017] [Indexed: 01/07/2023] Open
Abstract
The opportunistic pathogen Pseudomonas aeruginosa is a major cause of sepsis in severely burned patients. If it is not eradicated from the wound, it translocates to the bloodstream, causing sepsis, multiorgan failure, and death. We recently described the P. aeruginosa heparinase-encoding gene, hepP, whose expression was significantly enhanced when P. aeruginosa strain UCBPP_PA14 (PA14) was grown in whole blood from severely burned patients. Further analysis demonstrated that hepP contributed to the in vivo virulence of PA14 in the Caenorhabditis elegans model. In this study, we utilized the murine model of thermal injury to examine the contribution of hepP to the pathogenesis of P. aeruginosa during burn wound infection. Mutation of hepP reduced the rate of mortality from 100% for mice infected with PA14 to 7% for mice infected with PA14::hepP While comparable numbers of PA14 and PA14::hepP bacteria were recovered from infected skin, only PA14 was recovered from the livers and spleens of infected mice. Despite its inability to spread systemically, PA14::hepP formed perivascular cuffs around the blood vessels within the skin of the thermally injured/infected mice. Intraperitoneal inoculation of the thermally injured mice, bypassing the need for translocation, produced similar results. The rate of mortality for mice infected with PA14::hepP was 0%, whereas it was 66% for mice infected with PA14. As before, only PA14 was recovered from the livers and spleens of infected mice. These results suggest that hepP plays a crucial role in the pathogenesis of PA14 during burn wound infection, most likely by contributing to PA14 survival in the bloodstream of the thermally injured mouse during sepsis.
Collapse
|
38
|
Abstract
Syndecans comprise a major family of cell surface heparan sulfate proteoglycans (HSPGs). Syndecans are composed of sulfated glycosaminoglycans (GAGs), heparan sulfate (HS) or both HS and chondroitin sulfate (CS), attached covalently to core proteins. Syndecans regulate many cellular processes, such as adhesion, proliferation, and migration. Syndecans bind and regulate molecules primarily through their HS chains, but do not bind to all HS/heparin-binding molecules. Furthermore, mice ablated for the syndecan-1 or -4 gene do not show major developmental abnormalities, but they do show striking pathological phenotypes when challenged with infectious or inflammatory stimuli and conditions, suggesting that certain functions of syndecans are specific and cannot be compensated for by other syndecans or other HSPGs. These observations underscore the physiological importance of syndecans and indicate a need to study the activities of isolated native syndecans to define their molecular and cellular functions, and to establish their biological significance. Here we describe methods to isolate syndecans and several assays to analyze their functions.
Collapse
Affiliation(s)
- Pyong Woo Park
- Boston Children's Hospital, Harvard Medical School, Boston, MA, United States.
| |
Collapse
|
39
|
Abstract
Heparin is one of the oldest drugs, which nevertheless remains in widespread clinical use as an inhibitor of blood coagulation. The history of its identification a century ago unfolded amid one of the most fascinating scientific controversies turning around the distribution of credit for its discovery. The composition, purification and structure-function relationship of this naturally occurring glycosaminoglycan regarding its classical role as anticoagulant will be dealt with before proceeding to discuss its therapeutic potential in, among other, inflammatory and infectious disease, cancer treatment, cystic fibrosis and Alzheimer's disease. The first bibliographic reference hit using the words 'nanomedicine' and 'heparin' is as recent as 2008. Since then, nanomedical applications of heparin have experienced an exponential growth that will be discussed in detail, with particular emphasis on its antimalarial activity. Some of the most intriguing potential applications of heparin nanomedicines will be exposed, such as those contemplating the delivery of drugs to the mosquito stages of malaria parasites.
Collapse
Affiliation(s)
| | - Elena Lantero
- Nanomalaria Group, Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology, Baldiri Reixac 10-12, ES-08028 Barcelona, Spain.,Barcelona Institute for Global Health (ISGlobal), Barcelona Center for International Health Research (CRESIB, Hospital Clínic-Universitat de Barcelona), Rosselló 149-153, ES-08036 Barcelona, Spain
| | - Xavier Fernàndez-Busquets
- Nanomalaria Group, Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology, Baldiri Reixac 10-12, ES-08028 Barcelona, Spain.,Barcelona Institute for Global Health (ISGlobal), Barcelona Center for International Health Research (CRESIB, Hospital Clínic-Universitat de Barcelona), Rosselló 149-153, ES-08036 Barcelona, Spain.,Nanoscience & Nanotechnology Institute (IN2UB), University of Barcelona, Martí i Franquès 1, ES-08028 Barcelona, Spain
| |
Collapse
|
40
|
Kubaski F, Osago H, Mason RW, Yamaguchi S, Kobayashi H, Tsuchiya M, Orii T, Tomatsu S. Glycosaminoglycans detection methods: Applications of mass spectrometry. Mol Genet Metab 2017; 120:67-77. [PMID: 27746032 PMCID: PMC5477676 DOI: 10.1016/j.ymgme.2016.09.005] [Citation(s) in RCA: 89] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/23/2016] [Accepted: 09/25/2016] [Indexed: 12/26/2022]
Abstract
Glycosaminoglycans (GAGs) are long blocks of negatively charged polysaccharides. They are one of the major components of the extracellular matrix and play multiple roles in different tissues and organs. The accumulation of undegraded GAGs causes mucopolysaccharidoses (MPS). GAGs are associated with other pathological conditions such as osteoarthritis, inflammation, diabetes mellitus, spinal cord injury, and cancer. The need for further understanding of GAG functions and mechanisms of action boosted the development of qualitative and quantitative (alcian blue, toluidine blue, paper and thin layer chromatography, gas chromatography, high pressure liquid chromatography, capillary electrophoresis, 1,9-dimethylmethylene blue, enzyme linked-immunosorbent assay, mass spectrometry) techniques. The availability of quantitative techniques has facilitated translational research on GAGs into the medical field for: 1) diagnosis, monitoring, and screening for MPS; 2) analysis of GAG synthetic and degradation pathways; and 3) determination of physiological and pathological roles of GAGs. This review provides a history of development of GAG assays and insights about the use of tandem mass spectrometry and its applications for GAG analysis.
Collapse
Affiliation(s)
- Francyne Kubaski
- Nemours/Alfred I. duPont Hospital for Children, Wilmington, DE, USA; Department of Biological Sciences, University of Delaware, Newark, DE, USA
| | - Harumi Osago
- Department of Biochemistry, Shimane University, Shimane, Japan
| | - Robert W Mason
- Nemours/Alfred I. duPont Hospital for Children, Wilmington, DE, USA; Department of Biological Sciences, University of Delaware, Newark, DE, USA
| | - Seiji Yamaguchi
- Department of Pediatrics, Shimane University, Shimane, Japan
| | | | - Mikako Tsuchiya
- Department of Biochemistry, Shimane University, Shimane, Japan.
| | - Tadao Orii
- Department of Pediatrics, Gifu University, Gifu, Japan
| | - Shunji Tomatsu
- Nemours/Alfred I. duPont Hospital for Children, Wilmington, DE, USA; Department of Biological Sciences, University of Delaware, Newark, DE, USA; Department of Pediatrics, Shimane University, Shimane, Japan; Department of Pediatrics, Gifu University, Gifu, Japan.
| |
Collapse
|
41
|
García B, Merayo-Lloves J, Rodríguez D, Alcalde I, García-Suárez O, Alfonso JF, Baamonde B, Fernández-Vega A, Vazquez F, Quirós LM. Different Use of Cell Surface Glycosaminoglycans As Adherence Receptors to Corneal Cells by Gram Positive and Gram Negative Pathogens. Front Cell Infect Microbiol 2016; 6:173. [PMID: 27965938 PMCID: PMC5127826 DOI: 10.3389/fcimb.2016.00173] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2016] [Accepted: 11/17/2016] [Indexed: 12/13/2022] Open
Abstract
The epithelium of the cornea is continuously exposed to pathogens, and adhesion to epithelial cells is regarded as an essential first step in bacterial pathogenesis. In this article, the involvement of glycosaminoglycans in the adhesion of various pathogenic bacteria to corneal epithelial cells is analyzed. All microorganisms use glycosaminoglycans as receptors, but arranged in different patterns depending on the Gram-type of the bacterium. The heparan sulfate chains of syndecans are the main receptors, though other molecular species also seem to be involved, particularly in Gram-negative bacteria. Adherence is inhibited differentially by peptides, including heparin binding sequences, indicating the participation of various groups of Gram-positive, and -negative adhesins. The length of the saccharides produces a major effect, and low molecular weight chains inhibit the binding of Gram-negative microorganisms but increase the adherence of Gram-positives. Pathogen adhesion appears to occur preferentially through sulfated domains, and is very dependent on N- and 6-O-sulfation of the glucosamine residue and, to a lesser extent, 2-O sulfation of uronic acid. These data show the differential use of corneal receptors, which could facilitate the development of new anti-infective strategies.
Collapse
Affiliation(s)
- Beatriz García
- Ophthalmology, Vision Sciences and Advanced Therapies Research Group, Instituto Universitario Fernández-Vega, Universidad de OviedoOviedo, Spain; Departmento de Biología Funcional, Universidad de OviedoOviedo, Spain
| | - Jesús Merayo-Lloves
- Ophthalmology, Vision Sciences and Advanced Therapies Research Group, Instituto Universitario Fernández-Vega, Universidad de Oviedo Oviedo, Spain
| | - David Rodríguez
- Departamento de Bioquímica y Biología Molecular, Universidad de OviedoOviedo, Spain; Instituto Universitario de Oncología del Principado de Asturias, Universidad de OviedoOviedo, Spain
| | - Ignacio Alcalde
- Ophthalmology, Vision Sciences and Advanced Therapies Research Group, Instituto Universitario Fernández-Vega, Universidad de Oviedo Oviedo, Spain
| | - Olivia García-Suárez
- Ophthalmology, Vision Sciences and Advanced Therapies Research Group, Instituto Universitario Fernández-Vega, Universidad de OviedoOviedo, Spain; Departmento de Morfología y Biología Celular, Universidad de OviedoOviedo, Spain
| | - José F Alfonso
- Fundación de Investigación Oftalmológica, Instituto Oftalmológico Fernández-Vega Oviedo, Spain
| | - Begoña Baamonde
- Ophthalmology, Vision Sciences and Advanced Therapies Research Group, Instituto Universitario Fernández-Vega, Universidad de Oviedo Oviedo, Spain
| | - Andrés Fernández-Vega
- Fundación de Investigación Oftalmológica, Instituto Oftalmológico Fernández-Vega Oviedo, Spain
| | - Fernando Vazquez
- Ophthalmology, Vision Sciences and Advanced Therapies Research Group, Instituto Universitario Fernández-Vega, Universidad de OviedoOviedo, Spain; Departmento de Biología Funcional, Universidad de OviedoOviedo, Spain; Departmento de Microbiología, Hospital Universitario Central de AsturiasOviedo, Spain
| | - Luis M Quirós
- Ophthalmology, Vision Sciences and Advanced Therapies Research Group, Instituto Universitario Fernández-Vega, Universidad de OviedoOviedo, Spain; Departmento de Biología Funcional, Universidad de OviedoOviedo, Spain
| |
Collapse
|
42
|
Dual and antagonic therapeutic effects of sulfated glycans. Bioorg Med Chem 2016; 24:3965-3971. [DOI: 10.1016/j.bmc.2016.07.037] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2016] [Revised: 07/15/2016] [Accepted: 07/16/2016] [Indexed: 11/24/2022]
|
43
|
Chung H, Multhaupt HAB, Oh ES, Couchman JR. Minireview: Syndecans and their crucial roles during tissue regeneration. FEBS Lett 2016; 590:2408-17. [PMID: 27383370 DOI: 10.1002/1873-3468.12280] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2016] [Revised: 06/27/2016] [Accepted: 07/01/2016] [Indexed: 12/30/2022]
Abstract
Syndecans are transmembrane heparan sulfate proteoglycans, with roles in development, tumorigenesis and inflammation, and growing evidence for involvement in tissue regeneration. This is a fast developing field with the prospect of utilizing tissue engineering and biomaterials in novel therapies. Syndecan receptors are not only ubiquitous in mammalian tissues, regulating cell adhesion, migration, proliferation, and differentiation through independent signaling but also working alongside other receptors. Their importance is highlighted by an ability to interact with a diverse array of ligands, including extracellular matrix glycoproteins, growth factors, morphogens, and cytokines that are important regulators of regeneration. We also discuss the potential for syndecans to regulate stem cell properties, and suggest that understanding these proteoglycans is relevant to exploiting cell, tissue, and materials technologies.
Collapse
Affiliation(s)
- Heesung Chung
- Department of Life Sciences and the Research Center for Cellular Homeostasis, Ewha Womans University, Seoul, Korea
| | - Hinke A B Multhaupt
- Department of Biomedical Sciences and Biotech Research & Innovation Center, University of Copenhagen, Denmark
| | - Eok-Soo Oh
- Department of Life Sciences and the Research Center for Cellular Homeostasis, Ewha Womans University, Seoul, Korea
| | - John R Couchman
- Department of Biomedical Sciences and Biotech Research & Innovation Center, University of Copenhagen, Denmark
| |
Collapse
|
44
|
Ucakturk E, Akman O, Sun X, Baydar DE, Dolgun A, Zhang F, Linhardt RJ. Changes in composition and sulfation patterns of glycoaminoglycans in renal cell carcinoma. Glycoconj J 2015; 33:103-12. [PMID: 26662466 DOI: 10.1007/s10719-015-9643-1] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2015] [Revised: 11/23/2015] [Accepted: 11/24/2015] [Indexed: 01/08/2023]
Abstract
Glycosaminoglycans (GAGs) are heterogeneous, linear, highly charged, anionic polysaccharides consisting of repeating disaccharides units. GAGs have some biological significance in cancer progression (invasion and metastasis) and cell signaling. In different cancer types, GAGs undergo specific structural changes. In the present study, in depth investigation of changes in sulfation pattern and composition of GAGs, heparan sulfate (HS)/heparin (HP), chondroitin sulfate (CS)/dermatan sulfate and hyaluronan (HA) in normal renal tissue (NRT) and renal cell carcinoma tissue (RCCT) were evaluated. The statistical evaluation showed that alteration of the HS (HSNRT = 415.1 ± 115.3; HSRCCT = 277.5 ± 134.3), and CS (CSNRT = 35.3 ± 12.3; CSRCCT = 166.7 ± 108.8) amounts (in ng/mg dry tissue) were statistically significant (p < 0.05). Sulfation pattern in NRT and RCCT was evaluated to reveal disaccharide profiles. Statistical analyses showed that RCCT samples contain significantly increased amounts (in units of ng/mg dry tissue) of 4SCS (NRT = 25.7 ± 9.4; RCCT = 117.1 ± 73.9), SECS (NRT = 0.7 ± 0.3; RCCT = 4.7 ± 4.5), 6SCS (NRT = 6.1 ± 2.7; RCCT = 39.4 ± 34.7) and significantly decreased amounts (in units of ng/mg dry tissue) of NS6SHS (RCCT = 28.6 ± 6.5, RCCT = 10.2 ± 8.0), NS2SHS (RCCT = 44.2 ± 13.8; RCCT = 27.2 ± 15.0), NSHS (NRT = 68.4 ± 15.8; RCCT = 50.4 ± 21.2), 2S6SHS (NRT = 1.0 ± 0.4; RCCT = 0.4 ± 0.3), and 6SHS (NRT = 60.6 ± 17.5; RCCT = 24.9 ± 12.3). If these changes in GAGs are proven to be specific and sensitive, they may serve as potential biomarkers in RCC. Our findings are likely to help us to show the direction for further investigations to be able to bring different diagnostic and prognostic approaches in renal tumors.
Collapse
Affiliation(s)
- Ebru Ucakturk
- Department of Analytical Chemistry, Faculty of Pharmacy, Hacettepe University, 06100, Sıhhıye, Ankara, Turkey.
| | - Orkun Akman
- Department of Pathology, Hacettepe University School of Medicine, 06100, Sıhhıye, Ankara, Turkey
| | - Xiaojun Sun
- Department of Chemistry and Chemical Biology, Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, 110 Eighth Street, Troy, NY, 12180, USA
- Department of Biology, Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, 110 Eighth Street, Troy, NY, 12180, USA
- Department of Chemical and Biological Engineering, Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, 110 Eighth Street, Troy, NY, 12180, USA
- Department of Biomedical Engineering, Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, 110 Eighth Street, Troy, NY, 12180, USA
| | - Dilek Ertoy Baydar
- Department of Pathology, Hacettepe University School of Medicine, 06100, Sıhhıye, Ankara, Turkey
| | - Anil Dolgun
- Department of Biostatistics, Faculty of Medicine, Hacettepe University, 06100, Sıhhıye, Ankara, Turkey
| | - Fuming Zhang
- Department of Chemistry and Chemical Biology, Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, 110 Eighth Street, Troy, NY, 12180, USA
- Department of Biology, Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, 110 Eighth Street, Troy, NY, 12180, USA
- Department of Chemical and Biological Engineering, Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, 110 Eighth Street, Troy, NY, 12180, USA
- Department of Biomedical Engineering, Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, 110 Eighth Street, Troy, NY, 12180, USA
| | - Robert J Linhardt
- Department of Chemistry and Chemical Biology, Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, 110 Eighth Street, Troy, NY, 12180, USA.
- Department of Biology, Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, 110 Eighth Street, Troy, NY, 12180, USA.
- Department of Chemical and Biological Engineering, Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, 110 Eighth Street, Troy, NY, 12180, USA.
- Department of Biomedical Engineering, Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, 110 Eighth Street, Troy, NY, 12180, USA.
| |
Collapse
|
45
|
Pomin VH. Marine Non-Glycosaminoglycan Sulfated Glycans as Potential Pharmaceuticals. Pharmaceuticals (Basel) 2015; 8:848-64. [PMID: 26690451 PMCID: PMC4695813 DOI: 10.3390/ph8040848] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2015] [Revised: 11/30/2015] [Accepted: 12/08/2015] [Indexed: 12/18/2022] Open
Abstract
Sulfated fucans (SFs) and sulfated galactans (SGs) are currently the marine non-glycosaminoglycan (GAG) sulfated glycans most studied in glycomics. These compounds exhibit therapeutic effects in several pathophysiological systems such as blood coagulation, thrombosis, neovascularization, cancer, inflammation, and microbial infections. As analogs of the largely employed GAGs and due to some limitations of the GAG-based therapies, SFs and SGs comprise new carbohydrate-based therapeutics available for clinical studies. Here, the principal structural features and the major mechanisms of action of the SFs and SGs in the above-mentioned pathophysiological systems are presented. Discussion is also given on the current challenges and the future perspectives in drug development of these marine glycans.
Collapse
Affiliation(s)
- Vitor H Pomin
- Program of Glycobiology, Institute of Medical Biochemistry Leopoldo de Meis, University Hospital Clementino Fraga Filho, Federal University of Rio de Janeiro, Rio de Janeiro, RJ 21941-913, Brazil.
| |
Collapse
|