1
|
Cardenas AJ, Thomas KS, Broden MW, Ferraro NJ, Pires MM, John CM, Jarvis GA, Criss AK. Neisseria gonorrhoeae scavenges host sialic acid for Siglec-mediated, complement-independent suppression of neutrophil activation. mBio 2024; 15:e0011924. [PMID: 38587424 PMCID: PMC11078009 DOI: 10.1128/mbio.00119-24] [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: 01/14/2024] [Accepted: 03/12/2024] [Indexed: 04/09/2024] Open
Abstract
Gonorrhea, caused by the bacterium Neisseria gonorrhoeae (Gc), is characterized by neutrophilic influx to infection sites. Gc has developed mechanisms to resist killing by neutrophils that include modifications to its surface lipooligosaccharide (LOS). One such LOS modification is sialylation: Gc sialylates its terminal LOS sugars with cytidine-5'-monophosphate-N-acetylneuraminic acid, which is scavenged from the host using LOS sialyltransferase (Lst) since Gc cannot make its sialic acid. Sialylation enables sensitive strains of Gc to resist complement-mediated killing in a serum-dependent manner. However, little is known about the contribution of sialylation to complement-independent, direct Gc-neutrophil interactions. In the absence of complement, we found sialylated Gc expressing opacity-associated (Opa) proteins decreased the oxidative burst and granule exocytosis from primary human neutrophils. In addition, sialylated Opa+ Gc survived better than vehicle treated or Δlst Gc when challenged with neutrophils. However, Gc sialylation did not significantly affect Opa-dependent association with or internalization of Gc by neutrophils. Previous studies have implicated sialic acid-binding immunoglobulin-type lectins (Siglecs) in modulating neutrophil interactions with sialylated Gc. Blocking neutrophil Siglecs with antibodies that bind to their extracellular domains eliminated the ability of sialylated Opa+ Gc to suppress the oxidative burst and resist neutrophil killing. These findings highlight a new role for sialylation in Gc evasion of human innate immunity, with implications for the development of vaccines and therapeutics for gonorrhea. IMPORTANCE Neisseria gonorrhoeae, the bacterium that causes gonorrhea, is an urgent global health concern due to increasing infection rates, widespread antibiotic resistance, and its ability to thwart protective immune responses. The mechanisms by which Gc subverts protective immune responses remain poorly characterized. One way N. gonorrhoeae evades human immunity is by adding sialic acid that is scavenged from the host onto its lipooligosaccharide, using the sialyltransferase Lst. Here, we found that sialylation enhances N. gonorrhoeae survival from neutrophil assault and inhibits neutrophil activation, independently of the complement system. Our results implicate bacterial binding of sialic acid-binding lectins (Siglecs) on the neutrophil surface, which dampens neutrophil antimicrobial responses. This work identifies a new role for sialylation in protecting N. gonorrhoeae from cellular innate immunity, which can be targeted to enhance the human immune response in gonorrhea.
Collapse
Affiliation(s)
- Amaris J. Cardenas
- Department of Microbiology, Immunology, and Cancer Biology, University of Virginia, Charlottesville, Virginia, USA
| | - Keena S. Thomas
- Department of Microbiology, Immunology, and Cancer Biology, University of Virginia, Charlottesville, Virginia, USA
| | - Mary W. Broden
- Department of Microbiology, Immunology, and Cancer Biology, University of Virginia, Charlottesville, Virginia, USA
| | - Noel J. Ferraro
- Department of Chemistry, University of Virginia, Charlottesville, Virginia, USA
| | - Marcos M. Pires
- Department of Chemistry, University of Virginia, Charlottesville, Virginia, USA
| | - Constance M. John
- VA Medical Center and University of California, San Francisco, San Francisco, California, USA
| | - Gary A. Jarvis
- VA Medical Center and University of California, San Francisco, San Francisco, California, USA
| | - Alison K. Criss
- Department of Microbiology, Immunology, and Cancer Biology, University of Virginia, Charlottesville, Virginia, USA
| |
Collapse
|
2
|
Cardenas AJ, Thomas KS, Broden MW, Ferraro NJ, John CM, Pires MM, Jarvis GA, Criss AK. Neisseria gonorrhoeae scavenges host sialic acid for Siglec-mediated, complement-independent suppression of neutrophil activation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.01.17.576097. [PMID: 38293026 PMCID: PMC10827150 DOI: 10.1101/2024.01.17.576097] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/01/2024]
Abstract
Gonorrhea, caused by the bacterium Neisseria gonorrhoeae (Gc), is characterized by neutrophil influx to infection sites. Gc has developed mechanisms to resist killing by neutrophils that include modifications to its surface lipooligosaccharide (LOS). One such LOS modification is sialylation: Gc sialylates its terminal LOS sugars with cytidine-5'-monophosphate-N-acetylneuraminic acid (CMP-NANA) scavenged from the host using LOS sialyltransferase (Lst), since Gc cannot make its own sialic acid. Sialylation enables sensitive strains of Gc to resist complement-mediated killing in a serum-dependent manner. However, little is known about the contribution of sialylation to complement-independent, direct Gc-neutrophil interactions. In the absence of complement, we found sialylated Gc expressing opacity-associated (Opa) proteins decreased the oxidative burst and granule exocytosis from primary human neutrophils. In addition, sialylated Opa+ Gc survived better than vehicle treated or Δlst Gc when challenged with neutrophils. However, Gc sialylation did not significantly affect Opa-dependent association with or internalization of Gc by neutrophils. Previous studies have implicated sialic acid-binding immunoglobulin-type lectins (Siglecs) in modulating neutrophil interactions with sialylated Gc. Blocking neutrophil Siglecs with antibodies that bind to their extracellular domains eliminated the ability of sialylated Opa+ Gc to suppress oxidative burst and resist neutrophil killing. These findings highlight a new role for sialylation in Gc evasion of human innate immunity, with implications for the development of vaccines and therapeutics for gonorrhea.
Collapse
Affiliation(s)
- Amaris J Cardenas
- Department of Microbiology, Immunology, and Cancer Biology, University of Virginia, Charlottesville, VA, USA
| | - Keena S. Thomas
- Department of Microbiology, Immunology, and Cancer Biology, University of Virginia, Charlottesville, VA, USA
| | - Mary W. Broden
- Department of Microbiology, Immunology, and Cancer Biology, University of Virginia, Charlottesville, VA, USA
| | - Noel J. Ferraro
- Department of Chemistry, University of Virginia, Charlottesville, VA, USA
| | - Constance M. John
- VA Medical Center and University of California, San Francisco, San Francisco, CA, USA
| | - Marcos M. Pires
- Department of Chemistry, University of Virginia, Charlottesville, VA, USA
| | - Gary A. Jarvis
- VA Medical Center and University of California, San Francisco, San Francisco, CA, USA
| | - Alison K. Criss
- Department of Microbiology, Immunology, and Cancer Biology, University of Virginia, Charlottesville, VA, USA
| |
Collapse
|
3
|
de Oliveira Formiga R, Amaral FC, Souza CF, Mendes DAGB, Wanderley CWS, Lorenzini CB, Santos AA, Antônia J, Faria LF, Natale CC, Paula NM, Silva PCS, Fonseca FR, Aires L, Heck N, Starick MR, Queiroz‐Junior CM, Santos FRS, de Souza FRO, Costa VV, Barroso SPC, Morrot A, Van Weyenbergh J, Sordi R, Alisson‐Silva F, Cunha FQ, Rocha EL, Chollet‐Martin S, Hurtado‐Nedelec MM, Martin C, Burgel P, Mansur DS, Maurici R, Macauley MS, Báfica A, Witko‐Sarsat V, Spiller F. Neuraminidase is a host-directed approach to regulate neutrophil responses in sepsis and COVID-19. Br J Pharmacol 2023; 180:1460-1481. [PMID: 36526272 PMCID: PMC9877938 DOI: 10.1111/bph.16013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2022] [Revised: 07/29/2022] [Accepted: 08/16/2022] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND AND PURPOSE Neutrophil overstimulation plays a crucial role in tissue damage during severe infections. Because pathogen-derived neuraminidase (NEU) stimulates neutrophils, we investigated whether host NEU can be targeted to regulate the neutrophil dysregulation observed in severe infections. EXPERIMENTAL APPROACH The effects of NEU inhibitors on lipopolysaccharide (LPS)-stimulated neutrophils from healthy donors or COVID-19 patients were determined by evaluating the shedding of surface sialic acids, cell activation, and reactive oxygen species (ROS) production. Re-analysis of single-cell RNA sequencing of respiratory tract samples from COVID-19 patients also was carried out. The effects of oseltamivir on sepsis and betacoronavirus-induced acute lung injury were evaluated in murine models. KEY RESULTS Oseltamivir and zanamivir constrained host NEU activity, surface sialic acid release, cell activation, and ROS production by LPS-activated human neutrophils. Mechanistically, LPS increased the interaction of NEU1 with matrix metalloproteinase 9 (MMP-9). Inhibition of MMP-9 prevented LPS-induced NEU activity and neutrophil response. In vivo, treatment with oseltamivir fine-tuned neutrophil migration and improved infection control as well as host survival in peritonitis and pneumonia sepsis. NEU1 also is highly expressed in neutrophils from COVID-19 patients, and treatment of whole-blood samples from these patients with either oseltamivir or zanamivir reduced neutrophil overactivation. Oseltamivir treatment of intranasally infected mice with the mouse hepatitis coronavirus 3 (MHV-3) decreased lung neutrophil infiltration, viral load, and tissue damage. CONCLUSION AND IMPLICATIONS These findings suggest that interplay of NEU1-MMP-9 induces neutrophil overactivation. In vivo, NEU may serve as a host-directed target to dampen neutrophil dysfunction during severe infections.
Collapse
Affiliation(s)
- Rodrigo de Oliveira Formiga
- Department of PharmacologyFederal University of Santa CatarinaFlorianópolisBrazil
- Université de Paris, Institut Cochin, INSERM U1016, CNRSParisFrance
- Laboratory of Immunobiology, Department of Microbiology, Immunology and ParasitologyFederal University of Santa CatarinaFlorianópolisBrazil
| | - Flávia C. Amaral
- Department of PharmacologyFederal University of Santa CatarinaFlorianópolisBrazil
- Laboratory of Immunobiology, Department of Microbiology, Immunology and ParasitologyFederal University of Santa CatarinaFlorianópolisBrazil
| | - Camila F. Souza
- Department of PharmacologyFederal University of Santa CatarinaFlorianópolisBrazil
| | - Daniel A. G. B. Mendes
- Department of PharmacologyFederal University of Santa CatarinaFlorianópolisBrazil
- Laboratory of Immunobiology, Department of Microbiology, Immunology and ParasitologyFederal University of Santa CatarinaFlorianópolisBrazil
| | - Carlos W. S. Wanderley
- Department of Pharmacology, School of Medicine of Ribeirão PretoUniversity of São PauloRibeirão PretoBrazil
| | - Cristina B. Lorenzini
- Department of PharmacologyFederal University of Santa CatarinaFlorianópolisBrazil
- Laboratory of Immunobiology, Department of Microbiology, Immunology and ParasitologyFederal University of Santa CatarinaFlorianópolisBrazil
| | - Adara A. Santos
- Department of PharmacologyFederal University of Santa CatarinaFlorianópolisBrazil
- Laboratory of Immunobiology, Department of Microbiology, Immunology and ParasitologyFederal University of Santa CatarinaFlorianópolisBrazil
| | - Juliana Antônia
- Department of PharmacologyFederal University of Santa CatarinaFlorianópolisBrazil
| | - Lucas F. Faria
- Department of PharmacologyFederal University of Santa CatarinaFlorianópolisBrazil
| | - Caio C. Natale
- Department of PharmacologyFederal University of Santa CatarinaFlorianópolisBrazil
- Laboratory of Immunobiology, Department of Microbiology, Immunology and ParasitologyFederal University of Santa CatarinaFlorianópolisBrazil
| | - Nicholas M. Paula
- Department of PharmacologyFederal University of Santa CatarinaFlorianópolisBrazil
- Laboratory of Immunobiology, Department of Microbiology, Immunology and ParasitologyFederal University of Santa CatarinaFlorianópolisBrazil
| | - Priscila C. S. Silva
- Department of PharmacologyFederal University of Santa CatarinaFlorianópolisBrazil
| | - Fernanda R. Fonseca
- Department of Clinical MedicineFederal University of Santa CatarinaFlorianópolisBrazil
| | - Luan Aires
- Department of PharmacologyFederal University of Santa CatarinaFlorianópolisBrazil
- Laboratory of Immunobiology, Department of Microbiology, Immunology and ParasitologyFederal University of Santa CatarinaFlorianópolisBrazil
| | - Nicoli Heck
- Department of PharmacologyFederal University of Santa CatarinaFlorianópolisBrazil
- Laboratory of Immunobiology, Department of Microbiology, Immunology and ParasitologyFederal University of Santa CatarinaFlorianópolisBrazil
| | - Márick R. Starick
- Department of PharmacologyFederal University of Santa CatarinaFlorianópolisBrazil
- Laboratory of Immunobiology, Department of Microbiology, Immunology and ParasitologyFederal University of Santa CatarinaFlorianópolisBrazil
| | - Celso M. Queiroz‐Junior
- Department of Morphology, Institute of Biological SciencesFederal University of Minas GeraisBelo HorizonteBrazil
| | - Felipe R. S. Santos
- Department of Biochemistry and Immunology, Institute of Biological SciencesFederal University of Minas GeraisBelo HorizonteBrazil
| | - Filipe R. O. de Souza
- Department of Morphology, Institute of Biological SciencesFederal University of Minas GeraisBelo HorizonteBrazil
| | - Vivian V. Costa
- Department of Morphology, Institute of Biological SciencesFederal University of Minas GeraisBelo HorizonteBrazil
| | - Shana P. C. Barroso
- Molecular Biology Laboratory, Institute of Biomedical ResearchMarcilio Dias Naval Hospital, Navy of BrazilRio de JaneiroBrazil
| | - Alexandre Morrot
- Tuberculosis Research Laboratory, Faculty of MedicineFederal University of Rio de JaneiroRio de JaneiroBrazil
- Immunoparasitology LaboratoryOswaldo Cruz Foundation (FIOCRUZ)Rio de JaneiroBrazil
| | - Johan Van Weyenbergh
- Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, Laboratory for Clinical and Epidemiological VirologyKU LeuvenLeuvenBelgium
| | - Regina Sordi
- Department of PharmacologyFederal University of Santa CatarinaFlorianópolisBrazil
| | - Frederico Alisson‐Silva
- Department of Immunology, Paulo de Goes Institute of MicrobiologyFederal University of Rio de JaneiroRio de JaneiroBrazil
| | - Fernando Q. Cunha
- Department of Pharmacology, School of Medicine of Ribeirão PretoUniversity of São PauloRibeirão PretoBrazil
| | - Edroaldo L. Rocha
- Department of PharmacologyFederal University of Santa CatarinaFlorianópolisBrazil
- Laboratory of Immunobiology, Department of Microbiology, Immunology and ParasitologyFederal University of Santa CatarinaFlorianópolisBrazil
| | - Sylvie Chollet‐Martin
- INSERM UMR 996, ‘Infammation, Microbiome and Immunosurveillance’, Faculty of PharmacyUniversité Paris‐SaclayChâtenay‐MalabryFrance
| | | | - Clémence Martin
- Université de Paris, Institut Cochin, INSERM U1016, CNRSParisFrance
- Department of PneumologyAP‐HP, Hôpital CochinParisFrance
| | - Pierre‐Régis Burgel
- Université de Paris, Institut Cochin, INSERM U1016, CNRSParisFrance
- Department of PneumologyAP‐HP, Hôpital CochinParisFrance
| | - Daniel S. Mansur
- Laboratory of Immunobiology, Department of Microbiology, Immunology and ParasitologyFederal University of Santa CatarinaFlorianópolisBrazil
| | - Rosemeri Maurici
- Department of Clinical MedicineFederal University of Santa CatarinaFlorianópolisBrazil
| | - Matthew S. Macauley
- Department of Chemistry, Department of Medical Microbiology and ImmunologyUniversity of AlbertaEdmontonAlbertaCanada
| | - André Báfica
- Laboratory of Immunobiology, Department of Microbiology, Immunology and ParasitologyFederal University of Santa CatarinaFlorianópolisBrazil
| | | | - Fernando Spiller
- Department of PharmacologyFederal University of Santa CatarinaFlorianópolisBrazil
- Laboratory of Immunobiology, Department of Microbiology, Immunology and ParasitologyFederal University of Santa CatarinaFlorianópolisBrazil
| |
Collapse
|
4
|
Makarava N, Baskakov IV. Role of sialylation of N-linked glycans in prion pathogenesis. Cell Tissue Res 2023; 392:201-214. [PMID: 35088180 PMCID: PMC9329487 DOI: 10.1007/s00441-022-03584-2] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2021] [Accepted: 01/12/2022] [Indexed: 01/10/2023]
Abstract
Mammalian prion or PrPSc is a proteinaceous infectious agent that consists of a misfolded, self-replicating state of the prion protein or PrPC. PrPC and PrPSc are posttranslationally modified with N-linked glycans, which are sialylated at the terminal positions. More than 30 years have passed since the first characterization of the composition and structural diversity of N-linked glycans associated with the prion protein, yet the role of carbohydrate groups that constitute N-glycans and, in particular, their terminal sialic acid residues in prion disease pathogenesis remains poorly understood. A number of recent studies shed a light on the role of sialylation in the biology of prion diseases. This review article discusses several mechanisms by which terminal sialylation dictates the spread of PrPSc across brain regions and the outcomes of prion infection in an organism. In particular, relationships between the sialylation status of PrPSc and important strain-specific features including lymphotropism, neurotropism, and neuroinflammation are discussed. Moreover, emerging evidence pointing out the roles of sialic acid residues in prion replication, cross-species transmission, strain competition, and strain adaptation are reviewed. A hypothesis according to which selective, strain-specified recruitment of PrPC sialoglycoforms dictates unique strain-specific disease phenotypes is examined. Finally, the current article proposes that prion strains evolve as a result of a delicate balance between recruiting highly sialylated glycoforms to avoid an "eat-me" response by glia and limiting heavily sialylated glycoforms for enabling rapid prion replication.
Collapse
Affiliation(s)
- Natallia Makarava
- Center for Biomedical Engineering and Technology and Department of Anatomy and Neurobiology, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
| | - Ilia V Baskakov
- Center for Biomedical Engineering and Technology and Department of Anatomy and Neurobiology, University of Maryland School of Medicine, Baltimore, MD, 21201, USA.
| |
Collapse
|
5
|
de Oliveira Formiga R, Amaral FC, Souza CF, Mendes DAGB, Wanderley CWS, Lorenzini CB, Santos AA, Antônia J, Faria LF, Natale CC, Paula NM, Silva PCS, Fonseca FR, Aires L, Heck N, Starick MR, Queiroz-Junior CM, Santos FRS, de Souza FRO, Costa VV, Barroso SPC, Morrot A, Van Weyenbergh J, Sordi R, Alisson-Silva F, Cunha FQ, Rocha EL, Chollet-Martin S, Hurtado-Nedelec MM, Martin C, Burgel PR, Mansur DS, Maurici R, Macauley MS, Báfica A, Witko-Sarsat V, Spiller F. Neuraminidase inhibitors rewire neutrophil function in vivo in murine sepsis and ex vivo in COVID-19. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2022:2020.11.12.379115. [PMID: 33200130 PMCID: PMC7668734 DOI: 10.1101/2020.11.12.379115] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Neutrophil overstimulation plays a crucial role in tissue damage during severe infections. Neuraminidase (NEU)-mediated cleavage of surface sialic acid has been demonstrated to regulate leukocyte responses. Here, we report that antiviral NEU inhibitors constrain host NEU activity, surface sialic acid release, ROS production, and NETs released by microbial-activated human neutrophils. In vivo, treatment with Oseltamivir results in infection control and host survival in peritonitis and pneumonia models of sepsis. Single-cell RNA sequencing re-analysis of publicly data sets of respiratory tract samples from critical COVID-19 patients revealed an overexpression of NEU1 in infiltrated neutrophils. Moreover, Oseltamivir or Zanamivir treatment of whole blood cells from severe COVID-19 patients reduces host NEU-mediated shedding of cell surface sialic acid and neutrophil overactivation. These findings suggest that neuraminidase inhibitors can serve as host-directed interventions to dampen neutrophil dysfunction in severe infections.
Collapse
Affiliation(s)
- Rodrigo de Oliveira Formiga
- Department of Pharmacology, Federal University of Santa Catarina, Florianopolis, SC, Brazil
- Université de Paris, Institut Cochin, INSERM U1016, CNRS, Paris, France
- Laboratory of Immunobiology, Department of Microbiology, Immunology and Parasitology, Federal University of Santa Catarina, Florianopolis, SC, Brazil
| | - Flávia C. Amaral
- Department of Pharmacology, Federal University of Santa Catarina, Florianopolis, SC, Brazil
- Laboratory of Immunobiology, Department of Microbiology, Immunology and Parasitology, Federal University of Santa Catarina, Florianopolis, SC, Brazil
| | - Camila F. Souza
- Department of Pharmacology, Federal University of Santa Catarina, Florianopolis, SC, Brazil
| | - Daniel A. G. B. Mendes
- Department of Pharmacology, Federal University of Santa Catarina, Florianopolis, SC, Brazil
- Laboratory of Immunobiology, Department of Microbiology, Immunology and Parasitology, Federal University of Santa Catarina, Florianopolis, SC, Brazil
| | - Carlos W. S. Wanderley
- Department of Pharmacology, School of Medicine of Ribeirao Preto, University of Sao Paulo, Ribeirao Preto, SP, Brazil
| | - Cristina B. Lorenzini
- Department of Pharmacology, Federal University of Santa Catarina, Florianopolis, SC, Brazil
- Laboratory of Immunobiology, Department of Microbiology, Immunology and Parasitology, Federal University of Santa Catarina, Florianopolis, SC, Brazil
| | - Adara A. Santos
- Department of Pharmacology, Federal University of Santa Catarina, Florianopolis, SC, Brazil
- Laboratory of Immunobiology, Department of Microbiology, Immunology and Parasitology, Federal University of Santa Catarina, Florianopolis, SC, Brazil
| | - Juliana Antônia
- Department of Pharmacology, Federal University of Santa Catarina, Florianopolis, SC, Brazil
| | - Lucas F. Faria
- Department of Pharmacology, Federal University of Santa Catarina, Florianopolis, SC, Brazil
| | - Caio C. Natale
- Department of Pharmacology, Federal University of Santa Catarina, Florianopolis, SC, Brazil
- Laboratory of Immunobiology, Department of Microbiology, Immunology and Parasitology, Federal University of Santa Catarina, Florianopolis, SC, Brazil
| | - Nicholas M. Paula
- Department of Pharmacology, Federal University of Santa Catarina, Florianopolis, SC, Brazil
- Laboratory of Immunobiology, Department of Microbiology, Immunology and Parasitology, Federal University of Santa Catarina, Florianopolis, SC, Brazil
| | - Priscila C. S. Silva
- Department of Pharmacology, Federal University of Santa Catarina, Florianopolis, SC, Brazil
| | - Fernanda R. Fonseca
- Department of Clinical Medicine, Federal University of Santa Catarina, Florianopolis, SC, Brazil
| | - Luan Aires
- Department of Pharmacology, Federal University of Santa Catarina, Florianopolis, SC, Brazil
- Laboratory of Immunobiology, Department of Microbiology, Immunology and Parasitology, Federal University of Santa Catarina, Florianopolis, SC, Brazil
| | - Nicoli Heck
- Department of Pharmacology, Federal University of Santa Catarina, Florianopolis, SC, Brazil
- Laboratory of Immunobiology, Department of Microbiology, Immunology and Parasitology, Federal University of Santa Catarina, Florianopolis, SC, Brazil
| | - Márick R. Starick
- Department of Pharmacology, Federal University of Santa Catarina, Florianopolis, SC, Brazil
- Laboratory of Immunobiology, Department of Microbiology, Immunology and Parasitology, Federal University of Santa Catarina, Florianopolis, SC, Brazil
| | - Celso M. Queiroz-Junior
- Department of Morphology, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | - Felipe R. S. Santos
- Department of Biochemistry and Immunology, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | - Filipe R. O. de Souza
- Department of Morphology, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | - Vivian V. Costa
- Department of Morphology, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | - Shana P. C. Barroso
- Molecular Biology Laboratory, Institute of Biomedical Research, Marcilio Dias Naval Hospital, Navy of Brazil, RJ, Brazil
| | - Alexandre Morrot
- Tuberculosis Research Laboratory, Faculty of Medicine, Federal University of Rio de Janeiro
- Immunoparasitology Laboratory, Oswaldo Cruz Foundation, FIOCRUZ, Rio de Janeiro, Brazil
| | - Johan Van Weyenbergh
- Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, Laboratory for Clinical and Epidemiological Virology, KU Leuven, Leuven, Belgium
| | - Regina Sordi
- Department of Pharmacology, Federal University of Santa Catarina, Florianopolis, SC, Brazil
| | - Frederico Alisson-Silva
- Department of Immunology, Paulo de Goes Institute of Microbiology, Federal University of Rio de Janeiro (UFRJ), Rio de Janeiro, Brazil
| | - Fernando Q. Cunha
- Department of Pharmacology, School of Medicine of Ribeirao Preto, University of Sao Paulo, Ribeirao Preto, SP, Brazil
| | - Edroaldo L. Rocha
- Department of Pharmacology, Federal University of Santa Catarina, Florianopolis, SC, Brazil
- Laboratory of Immunobiology, Department of Microbiology, Immunology and Parasitology, Federal University of Santa Catarina, Florianopolis, SC, Brazil
| | - Sylvie Chollet-Martin
- INSERM UMR 996, “Infammation, Microbiome and Immunosurveillance”, Faculty of Pharmacy, Université Paris-Saclay, Châtenay-Malabry, France
| | | | - Clémence Martin
- Université de Paris, Institut Cochin, INSERM U1016, CNRS, Paris, France
- Department of Pneumology, AP-HP, Hôpital Cochin, Paris, France
| | - Pierre-Régis Burgel
- Université de Paris, Institut Cochin, INSERM U1016, CNRS, Paris, France
- Department of Pneumology, AP-HP, Hôpital Cochin, Paris, France
| | - Daniel S. Mansur
- Laboratory of Immunobiology, Department of Microbiology, Immunology and Parasitology, Federal University of Santa Catarina, Florianopolis, SC, Brazil
| | - Rosemeri Maurici
- Department of Clinical Medicine, Federal University of Santa Catarina, Florianopolis, SC, Brazil
| | - Matthew S. Macauley
- Department of Chemistry, Department of Medical Microbiology and Immunology, University of Alberta, Edmonton, Alberta, Canada
| | - André Báfica
- Laboratory of Immunobiology, Department of Microbiology, Immunology and Parasitology, Federal University of Santa Catarina, Florianopolis, SC, Brazil
| | | | - Fernando Spiller
- Department of Pharmacology, Federal University of Santa Catarina, Florianopolis, SC, Brazil
- Laboratory of Immunobiology, Department of Microbiology, Immunology and Parasitology, Federal University of Santa Catarina, Florianopolis, SC, Brazil
| |
Collapse
|
6
|
Hyun SW, Feng C, Liu A, Lillehoj EP, Trotta R, Kingsbury TJ, Passaniti A, Lugkey KN, Chauhan S, Cipollo JF, Luzina IG, Atamas SP, Cross AS, Goldblum SE. Altered sialidase expression in human myeloid cells undergoing apoptosis and differentiation. Sci Rep 2022; 12:14173. [PMID: 35986080 PMCID: PMC9390117 DOI: 10.1038/s41598-022-18448-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2021] [Accepted: 08/11/2022] [Indexed: 11/10/2022] Open
Abstract
To gain insight into sialic acid biology and sialidase/neuraminidase (NEU) expression in mature human neutrophil (PMN)s, we studied NEU activity and expression in PMNs and the HL60 promyelocytic leukemic cell line, and changes that might occur in PMNs undergoing apoptosis and HL60 cells during their differentiation into PMN-like cells. Mature human PMNs contained NEU activity and expressed NEU2, but not NEU1, the NEU1 chaperone, protective protein/cathepsin A(PPCA), NEU3, and NEU4 proteins. In proapoptotic PMNs, NEU2 protein expression increased > 30.0-fold. Granulocyte colony-stimulating factor protected against NEU2 protein upregulation, PMN surface desialylation and apoptosis. In response to 3 distinct differentiating agents, dimethylformamide, dimethylsulfoxide, and retinoic acid, total NEU activity in differentiated HL60 (dHL60) cells was dramatically reduced compared to that of nondifferentiated cells. With differentiation, NEU1 protein levels decreased > 85%, PPCA and NEU2 proteins increased > 12.0-fold, and 3.0-fold, respectively, NEU3 remained unchanged, and NEU4 increased 1.7-fold by day 3, and then returned to baseline. In dHL60 cells, lectin blotting revealed decreased α2,3-linked and increased α2,6-linked sialylation. dHL60 cells displayed increased adhesion to and migration across human bone marrow-derived endothelium and increased bacterial phagocytosis. Therefore, myeloid apoptosis and differentiation provoke changes in NEU catalytic activity and protein expression, surface sialylation, and functional responsiveness.
Collapse
|
7
|
Neuraminidase Inhibitor Zanamivir Ameliorates Collagen-Induced Arthritis. Int J Mol Sci 2021; 22:ijms22031428. [PMID: 33572654 PMCID: PMC7867009 DOI: 10.3390/ijms22031428] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2020] [Revised: 01/27/2021] [Accepted: 01/27/2021] [Indexed: 01/20/2023] Open
Abstract
Altered sialylation patterns play a role in chronic autoimmune diseases such as rheumatoid arthritis (RA). Recent studies have shown the pro-inflammatory activities of immunoglobulins (Igs) with desialylated sugar moieties. The role of neuraminidases (NEUs), enzymes which are responsible for the cleavage of terminal sialic acids (SA) from sialoglycoconjugates, is not fully understood in RA. We investigated the impact of zanamivir, an inhibitor of the influenza virus neuraminidase, and mammalian NEU2/3 on clinical outcomes in experimental arthritides studies. The severity of arthritis was monitored and IgG titers were measured by ELISA. (2,6)-linked SA was determined on IgG by ELISA and on cell surfaces by flow cytometry. Zanamivir at a dose of 100 mg/kg (zana-100) significantly ameliorated collagen-induced arthritis (CIA), whereas zana-100 was ineffective in serum transfer-induced arthritis. Systemic zana-100 treatment reduced the number of splenic CD138+/TACI+ plasma cells and CD19+ B cells, which was associated with lower IgG levels and an increased sialylation status of IgG compared to controls. Our data reveal the contribution of NEU2/3 in CIA. Zanamivir down-modulated the T and B cell-dependent humoral immune response and induced an anti-inflammatory milieu by inhibiting sialic acid degradation. We suggest that neuraminidases might represent a promising therapeutic target for RA and possibly also for other antibody-mediated autoimmune diseases.
Collapse
|
8
|
Baskakov IV. Role of sialylation in prion disease pathogenesis and prion structure. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2020; 175:31-52. [PMID: 32958238 DOI: 10.1016/bs.pmbts.2020.07.004] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Mammalian prion or PrPSc is a proteinaceous infectious agent that consists of a misfolded, self-replicating state of a sialoglycoprotein called the prion protein or PrPC. Sialylation of the prion protein, a terminal modification of N-linked glycans, was discovered more than 30 years ago, yet the role of sialylation in prion pathogenesis is not well understood. This chapter summarizes current knowledge on the role of sialylation of the prion protein in prion diseases. First, we discuss recent data suggesting that sialylation of PrPSc N-linked glycans determines the fate of prion infection in an organism and control prion lymphotropism. Second, emerging evidence pointing out at the role N-glycans in neuroinflammation are discussed. Thirds, this chapter reviews a mechanism postulating that sialylated N-linked glycans are important players in defining strain-specific structures. A new hypothesis according to which individual strain-specific PrPSc structures govern selection of PrPC sialoglycoforms is discussed. Finally, this chapter explain how N-glycan sialylation control the prion replication and strain interference. In summary, comprehensive review of our knowledge on N-linked glycans and their sialylation provided in this chapter helps to answer important questions of prion biology that have been puzzling for years.
Collapse
Affiliation(s)
- Ilia V Baskakov
- Department of Anatomy and Neurobiology, and Center for Biomedical Engineering and Technology, University of Maryland School of Medicine, Baltimore, MD, United States.
| |
Collapse
|
9
|
Vilcaes AA, Garbarino-Pico E, Torres Demichelis V, Daniotti JL. Ganglioside Synthesis by Plasma Membrane-Associated Sialyltransferase in Macrophages. Int J Mol Sci 2020; 21:ijms21031063. [PMID: 32033474 PMCID: PMC7043224 DOI: 10.3390/ijms21031063] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2019] [Revised: 02/03/2020] [Accepted: 02/03/2020] [Indexed: 12/21/2022] Open
Abstract
Gangliosides are constituents of the mammalian cell membranes and participate in the inflammatory response. However, little is known about the presence and enzymatic activity of ganglioside sialyltransferases at the cell surface of macrophages, one of the most important immune cells involved in the innate inflammatory process. In the present study, using biochemical and fluorescent microscopy approaches, we found that endogenous ST8Sia-I is present at the plasma membrane (ecto-ST8Sia-I) of murine macrophage RAW264.7 cells. Moreover, ecto-ST8Sia-I can synthetize GD3 ganglioside at the cell surface in lipopolysaccharide (LPS)-stimulated macrophages even when LPS-stimulated macrophages reduced the total ST8Sia-I expression levels. Besides, cotreatment of LPS with an inhibitor of nitric oxide (NO) synthase recovered the ecto-ST8Sia-I expression, suggesting that NO production is involved in the reduction of ST8Sia-I expression. The diminution of ST8Sia-I expression in LPS-stimulated macrophages correlated with a reduction of GD3 and GM1 gangliosides and with an increment of GD1a. Taken together, the data supports the presence and activity of sialyltransferases at the plasma membrane of RAW264.7 cells. The variations of ecto-ST8Sia-I and ganglioside levels in stimulated macrophages constitutes a promissory pathway to further explore the physiological role of this and others ganglioside metabolism-related enzymes at the cell surface during the immune response.
Collapse
Affiliation(s)
- Aldo A. Vilcaes
- Centro de Investigaciones en Química Biológica de Córdoba (CIQUIBIC), CONICET. Universidad Nacional de Córdoba, Córdoba X5000HUA, Argentina; (E.G.-P.); (V.T.D.)
- Departamento de Química Biológica Ranwel Caputto, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba X5000HUA, Argentina
- Correspondence: (A.A.V.); (J.L.D.)
| | - Eduardo Garbarino-Pico
- Centro de Investigaciones en Química Biológica de Córdoba (CIQUIBIC), CONICET. Universidad Nacional de Córdoba, Córdoba X5000HUA, Argentina; (E.G.-P.); (V.T.D.)
- Departamento de Química Biológica Ranwel Caputto, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba X5000HUA, Argentina
| | - Vanina Torres Demichelis
- Centro de Investigaciones en Química Biológica de Córdoba (CIQUIBIC), CONICET. Universidad Nacional de Córdoba, Córdoba X5000HUA, Argentina; (E.G.-P.); (V.T.D.)
- Departamento de Química Biológica Ranwel Caputto, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba X5000HUA, Argentina
| | - Jose L. Daniotti
- Centro de Investigaciones en Química Biológica de Córdoba (CIQUIBIC), CONICET. Universidad Nacional de Córdoba, Córdoba X5000HUA, Argentina; (E.G.-P.); (V.T.D.)
- Departamento de Química Biológica Ranwel Caputto, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba X5000HUA, Argentina
- Correspondence: (A.A.V.); (J.L.D.)
| |
Collapse
|
10
|
Szabo R, Skropeta D. Advancement of Sialyltransferase Inhibitors: Therapeutic Challenges and Opportunities. Med Res Rev 2016; 37:219-270. [DOI: 10.1002/med.21407] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2016] [Revised: 07/14/2016] [Accepted: 08/03/2016] [Indexed: 01/06/2023]
Affiliation(s)
- Rémi Szabo
- School of Chemistry; University of Wollongong; Wollongong NSW 2522 Australia
| | - Danielle Skropeta
- School of Chemistry; University of Wollongong; Wollongong NSW 2522 Australia
- Centre for Medical & Molecular Bioscience; University of Wollongong; Wollongong NSW 2522 Australia
| |
Collapse
|
11
|
Katorcha E, Srivastava S, Klimova N, Baskakov IV. Sialylation of Glycosylphosphatidylinositol (GPI) Anchors of Mammalian Prions Is Regulated in a Host-, Tissue-, and Cell-specific Manner. J Biol Chem 2016; 291:17009-19. [PMID: 27317661 PMCID: PMC5016106 DOI: 10.1074/jbc.m116.732040] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2016] [Revised: 06/09/2016] [Indexed: 01/08/2023] Open
Abstract
Prions or PrP(Sc) are proteinaceous infectious agents that consist of misfolded, self-replicating states of the prion protein or PrP(C) PrP(C) is posttranslationally modified with N-linked glycans and a sialylated glycosylphosphatidylinositol (GPI) anchor. Conformational conversion of PrP(C) gives rise to glycosylated and GPI-anchored PrP(Sc) The question of the sialylation status of GPIs within PrP(Sc) has been controversial. Previous studies that examined scrapie brains reported that both sialo- and asialo-GPIs were present in PrP(Sc), with the majority being asialo-GPIs. In contrast, recent work that employed cultured cells claimed that only PrP(C) with sialylo-GPIs could be recruited into PrP(Sc), whereas PrP(C) with asialo-GPIs inhibited conversion. To resolve this controversy, we analyzed the sialylation status of GPIs within PrP(Sc) generated in the brain, spleen, or cultured N2a or C2C12 myotube cells. We found that recruiting PrP(C) with both sialo- and asialo-GPIs is a common feature of PrP(Sc) The mixtures of sialo- and asialo-GPIs were observed in PrP(Sc) universally regardless of prion strain as well as host, tissue, or type of cells that produced PrP(Sc) Remarkably, the proportion of sialo- versus asialo-GPIs was found to be controlled by host, tissue, and cell type but not prion strain. In summary, this study found no strain-specific preferences for selecting PrP(C) with sialo- versus asialo-GPIs. Instead, this work suggests that the sialylation status of GPIs within PrP(Sc) is regulated in a cell-, tissue-, or host-specific manner and is likely to be determined by the specifics of GPI biosynthesis.
Collapse
Affiliation(s)
- Elizaveta Katorcha
- From the Center for Biomedical Engineering and Technology and the Department of Anatomy and Neurobiology, University of Maryland School of Medicine, Baltimore, Maryland 21201
| | - Saurabh Srivastava
- From the Center for Biomedical Engineering and Technology and the Department of Anatomy and Neurobiology, University of Maryland School of Medicine, Baltimore, Maryland 21201
| | - Nina Klimova
- From the Center for Biomedical Engineering and Technology and the Department of Anatomy and Neurobiology, University of Maryland School of Medicine, Baltimore, Maryland 21201
| | - Ilia V Baskakov
- From the Center for Biomedical Engineering and Technology and the Department of Anatomy and Neurobiology, University of Maryland School of Medicine, Baltimore, Maryland 21201
| |
Collapse
|
12
|
Baskakov IV, Katorcha E. Multifaceted Role of Sialylation in Prion Diseases. Front Neurosci 2016; 10:358. [PMID: 27551257 PMCID: PMC4976111 DOI: 10.3389/fnins.2016.00358] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2016] [Accepted: 07/18/2016] [Indexed: 11/13/2022] Open
Abstract
Mammalian prion or PrP(Sc) is a proteinaceous infectious agent that consists of a misfolded, self-replicating state of a sialoglycoprotein called the prion protein, or PrP(C). Sialylation of the prion protein N-linked glycans was discovered more than 30 years ago, yet the role of sialylation in prion pathogenesis remains poorly understood. Recent years have witnessed extraordinary growth in interest in sialylation and established a critical role for sialic acids in host invasion and host-pathogen interactions. This review article summarizes current knowledge on the role of sialylation of the prion protein in prion diseases. First, we discuss the correlation between sialylation of PrP(Sc) glycans and prion infectivity and describe the factors that control sialylation of PrP(Sc). Second, we explain how glycan sialylation contributes to the prion replication barrier, defines strain-specific glycoform ratios, and imposes constraints for PrP(Sc) structure. Third, several topics, including a possible role for sialylation in animal-to-human prion transmission, prion lymphotropism, toxicity, strain interference, and normal function of PrP(C), are critically reviewed. Finally, a metabolic hypothesis on the role of sialylation in the etiology of sporadic prion diseases is proposed.
Collapse
Affiliation(s)
- Ilia V. Baskakov
- Department of Anatomy and Neurobiology, Center for Biomedical Engineering and Technology, University of Maryland School of MedicineBaltimore, MD, USA
| | | |
Collapse
|
13
|
Wratil PR, Horstkorte R, Reutter W. Metabolic Glycoengineering with N-Acyl Side Chain Modified Mannosamines. Angew Chem Int Ed Engl 2016; 55:9482-512. [PMID: 27435524 DOI: 10.1002/anie.201601123] [Citation(s) in RCA: 96] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2016] [Indexed: 12/14/2022]
Abstract
In metabolic glycoengineering (MGE), cells or animals are treated with unnatural derivatives of monosaccharides. After entering the cytosol, these sugar analogues are metabolized and subsequently expressed on newly synthesized glycoconjugates. The feasibility of MGE was first discovered for sialylated glycans, by using N-acyl-modified mannosamines as precursor molecules for unnatural sialic acids. Prerequisite is the promiscuity of the enzymes of the Roseman-Warren biosynthetic pathway. These enzymes were shown to tolerate specific modifications of the N-acyl side chain of mannosamine analogues, for example, elongation by one or more methylene groups (aliphatic modifications) or by insertion of reactive groups (bioorthogonal modifications). Unnatural sialic acids are incorporated into glycoconjugates of cells and organs. MGE has intriguing biological consequences for treated cells (aliphatic MGE) and offers the opportunity to visualize the topography and dynamics of sialylated glycans in vitro, ex vivo, and in vivo (bioorthogonal MGE).
Collapse
Affiliation(s)
- Paul R Wratil
- Institut für Laboratoriumsmedizin, Klinische Chemie und Pathobiochemie, Charité-Universitätsmedizin Berlin, Arnimallee 22, 14195, Berlin, Germany.
| | - Rüdiger Horstkorte
- Institut für Physiologische Chemie, Martin-Luther-Universität Halle-Wittenberg, Hollystrasse 1, 06114, Halle, Germany.
| | - Werner Reutter
- Institut für Laboratoriumsmedizin, Klinische Chemie und Pathobiochemie, Charité-Universitätsmedizin Berlin, Arnimallee 22, 14195, Berlin, Germany
| |
Collapse
|
14
|
Wratil PR, Horstkorte R, Reutter W. Metabolisches Glykoengineering mitN-Acyl-Seiten- ketten-modifizierten Mannosaminen. Angew Chem Int Ed Engl 2016. [DOI: 10.1002/ange.201601123] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Affiliation(s)
- Paul R. Wratil
- Institut für Laboratoriumsmedizin, Klinische Chemie und Pathobiochemie; Charité - Universitätsmedizin Berlin; Arnimallee 22 14195 Berlin Deutschland
| | - Rüdiger Horstkorte
- Institut für Physiologische Chemie; Martin-Luther-Universität Halle-Wittenberg; Hollystraße 1 06114 Halle Deutschland
| | - Werner Reutter
- Institut für Laboratoriumsmedizin, Klinische Chemie und Pathobiochemie; Charité - Universitätsmedizin Berlin; Arnimallee 22 14195 Berlin Deutschland
| |
Collapse
|
15
|
Srivastava S, Makarava N, Katorcha E, Savtchenko R, Brossmer R, Baskakov IV. Post-conversion sialylation of prions in lymphoid tissues. Proc Natl Acad Sci U S A 2015; 112:E6654-62. [PMID: 26627256 PMCID: PMC4672809 DOI: 10.1073/pnas.1517993112] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Sialylated glycans on the surface of mammalian cells act as part of a "self-associated molecular pattern," helping the immune system to recognize "self" from "altered self" or "nonself." To escape the host immune system, some bacterial pathogens have evolved biosynthetic pathways for host-like sialic acids, whereas others recruited host sialic acids for decorating their surfaces. Prions lack nucleic acids and are not conventional pathogens. Nevertheless, prions might use a similar strategy for invading and colonizing the lymphoreticular system. Here we show that the sialylation status of the infectious, disease-associated state of the prion protein (PrP(Sc)) changes with colonization of secondary lymphoid organs (SLOs). As a result, spleen-derived PrP(Sc) is more sialylated than brain-derived PrP(Sc). Enhanced sialylation of PrP(Sc) is recapitulated in vitro by incubating brain-derived PrP(Sc) with primary splenocytes or cultured macrophage RAW 264.7 cells. General inhibitors of sialyltranserases (STs), the enzymes that transfer sialic acid residues onto terminal positions of glycans, suppressed extrasialylation of PrP(Sc). A fluorescently labeled precursor of sialic acid revealed ST activity associated with RAW macrophages. This study illustrates that, upon colonization of SLOs, the sialylation status of prions changes by host STs. We propose that this mechanism is responsible for camouflaging prions in SLOs and has broad implications.
Collapse
Affiliation(s)
- Saurabh Srivastava
- Center for Biomedical Engineering and Technology, University of Maryland School of Medicine, Baltimore, MD 21201; Department of Anatomy and Neurobiology, University of Maryland School of Medicine, Baltimore, MD 21201
| | - Natallia Makarava
- Center for Biomedical Engineering and Technology, University of Maryland School of Medicine, Baltimore, MD 21201; Department of Anatomy and Neurobiology, University of Maryland School of Medicine, Baltimore, MD 21201
| | - Elizaveta Katorcha
- Center for Biomedical Engineering and Technology, University of Maryland School of Medicine, Baltimore, MD 21201; Department of Anatomy and Neurobiology, University of Maryland School of Medicine, Baltimore, MD 21201
| | - Regina Savtchenko
- Center for Biomedical Engineering and Technology, University of Maryland School of Medicine, Baltimore, MD 21201; Department of Anatomy and Neurobiology, University of Maryland School of Medicine, Baltimore, MD 21201
| | - Reinhard Brossmer
- Biochemistry Center, University of Heidelberg, 69120 Heidelberg, Germany
| | - Ilia V Baskakov
- Center for Biomedical Engineering and Technology, University of Maryland School of Medicine, Baltimore, MD 21201; Department of Anatomy and Neurobiology, University of Maryland School of Medicine, Baltimore, MD 21201;
| |
Collapse
|
16
|
Feng C, Zhang L, Nguyen C, Vogel SN, Goldblum SE, Blackwelder WC, Cross AS. Neuraminidase reprograms lung tissue and potentiates lipopolysaccharide-induced acute lung injury in mice. THE JOURNAL OF IMMUNOLOGY 2013; 191:4828-37. [PMID: 24068662 DOI: 10.4049/jimmunol.1202673] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
We previously reported that removal of sialyl residues primed PBMCs to respond to bacterial LPS stimulation in vitro. Therefore, we speculated that prior desialylation can sensitize the host to generate an enhanced inflammatory response upon exposure to a TLR ligand, such as LPS, in a murine model of acute lung injury. Intratracheal instillation of neuraminidase (NA) 30 min prior to intratracheal administration of LPS increased polymorphonuclear leukocytes (PMNs) in the bronchoalveolar lavage fluid and the wet-to-dry lung weight ratio, a measure of pulmonary edema, compared with mice that received LPS alone. Administration of NA alone resulted in desialylation of bronchiolar and alveolar surfaces and induction of TNF-α, IL-1β, and chemokines in lung homogenates and bronchoalveolar lavage fluid; however, PMN recruitment in mice treated with NA alone did not differ from that of PBS-administered controls. NA pretreatment alone induced apoptosis and markedly enhanced LPS-induced endothelial apoptosis. Administration of recombinant Bcl-2, an antiapoptotic molecule, abolished the effect of NA treatment on LPS-induced PMN recruitment and pulmonary edema formation. We conclude that NA pretreatment potentiates LPS-induced lung injury through enhanced PMN recruitment, pulmonary edema formation, and endothelial and myeloid cell apoptosis. A similar "reprogramming" of immune responses with desialylation may occur during respiratory infection with NA-expressing microbes and contribute to severe lung injury.
Collapse
Affiliation(s)
- Chiguang Feng
- Center for Vaccine Development, University of Maryland School of Medicine, Baltimore, MD 21201
| | | | | | | | | | | | | |
Collapse
|
17
|
Abstract
Tumor cells exhibit striking changes in cell surface glycosylation as a consequence of dysregulated glycosyltransferases and glycosidases. In particular, an increase in the expression of certain sialylated glycans is a prominent feature of many transformed cells. Altered sialylation has long been associated with metastatic cell behaviors including invasion and enhanced cell survival; however, there is limited information regarding the molecular details of how distinct sialylated structures or sialylated carrier proteins regulate cell signaling to control responses such as adhesion/migration or resistance to specific apoptotic pathways. The goal of this review is to highlight selected examples of sialylated glycans for which there is some knowledge of molecular mechanisms linking aberrant sialylation to critical processes involved in metastasis.
Collapse
Affiliation(s)
- Matthew J Schultz
- Department of Cell, Developmental and Integrative Biology, University of Alabama at Birmingham, MCLM 982A 1918 University Boulevard, Birmingham, AL 35294-0005, USA
| | | | | |
Collapse
|
18
|
Feng C, Stamatos NM, Dragan AI, Medvedev A, Whitford M, Zhang L, Song C, Rallabhandi P, Cole L, Nhu QM, Vogel SN, Geddes CD, Cross AS. Sialyl residues modulate LPS-mediated signaling through the Toll-like receptor 4 complex. PLoS One 2012; 7:e32359. [PMID: 22496731 PMCID: PMC3322133 DOI: 10.1371/journal.pone.0032359] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2011] [Accepted: 01/26/2012] [Indexed: 11/19/2022] Open
Abstract
We previously reported that neuraminidase (NA) pretreatment of human PBMCs markedly increased their cytokine response to lipopolysaccharide (LPS). To study the mechanisms by which this occurs, we transfected HEK293T cells with plasmids encoding TLR4, CD14, and MD2 (three components of the LPS receptor complex), as well as a NFκB luciferase reporting system. Both TLR4 and MD2 encoded by the plasmids are α-2,6 sialylated. HEK293T cells transfected with TLR4/MD2/CD14 responded robustly to the addition of LPS; however, omission of the MD2 plasmid abrogated this response. Addition of culture supernatants from MD2 (sMD2)-transfected HEK293T cells, but not recombinant, non-glycosylated MD2 reconstituted this response. NA treatment of sMD2 enhanced the LPS response as did NA treatment of the TLR4/CD14-transfected cell supplemented with untreated sMD2, but optimal LPS-initiated responses were observed with NA-treated TLR4/CD14-transfected cells supplemented with NA-treated sMD2. We hypothesized that removal of negatively charged sialyl residues from glycans on the TLR4 complex would hasten the dimerization of TLR4 monomers required for signaling. Co-transfection of HEK293T cells with separate plasmids encoding either YFP- or FLAG-tagged TLR4, followed by treatment with NA and stimulation with LPS, led to an earlier and more robust time-dependent dimerization of TLR4 monomers on co-immunoprecipitation, compared to untreated cells. These findings were confirmed by fluorescence resonance energy transfer (FRET) analysis. Overexpression of human Neu1 increased LPS-initiated TLR4-mediated NFκB activation and a NA inhibitor suppressed its activation. We conclude that (1) sialyl residues on TLR4 modulate LPS responsiveness, perhaps by facilitating clustering of the homodimers, and that (2) sialic acid, and perhaps other glycosyl species, regulate MD2 activity required for LPS-mediated signaling. We speculate that endogenous sialidase activity mobilized during cell activation may play a role in this regulation.
Collapse
Affiliation(s)
- Chiguang Feng
- Center for Vaccine Development, University of Maryland School of Medicine, Baltimore, Maryland, United States of America
| | - Nicholas M. Stamatos
- Institute of Human Virology and Department of Medicine, University of Maryland School of Medicine, Baltimore, Maryland, United States of America
| | - Anatoliy I. Dragan
- Institute of Fluorescence, University of Maryland, Baltimore County, Baltimore, Maryland, United States of America
| | - Andrei Medvedev
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, Maryland, United States of America
| | - Melissa Whitford
- Center for Vaccine Development, University of Maryland School of Medicine, Baltimore, Maryland, United States of America
| | - Lei Zhang
- Center for Vaccine Development, University of Maryland School of Medicine, Baltimore, Maryland, United States of America
| | - Chang Song
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, Maryland, United States of America
| | - Prasad Rallabhandi
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, Maryland, United States of America
| | - Leah Cole
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, Maryland, United States of America
| | - Quan M. Nhu
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, Maryland, United States of America
| | - Stefanie N. Vogel
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, Maryland, United States of America
| | - Chris D. Geddes
- Institute of Fluorescence, University of Maryland, Baltimore County, Baltimore, Maryland, United States of America
| | - Alan S. Cross
- Center for Vaccine Development, University of Maryland School of Medicine, Baltimore, Maryland, United States of America
- * E-mail:
| |
Collapse
|
19
|
Cross AS, Hyun SW, Miranda-Ribera A, Feng C, Liu A, Nguyen C, Zhang L, Luzina IG, Atamas SP, Twaddell WS, Guang W, Lillehoj EP, Puché AC, Huang W, Wang LX, Passaniti A, Goldblum SE. NEU1 and NEU3 sialidase activity expressed in human lung microvascular endothelia: NEU1 restrains endothelial cell migration, whereas NEU3 does not. J Biol Chem 2012; 287:15966-80. [PMID: 22403397 DOI: 10.1074/jbc.m112.346817] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
The microvascular endothelial surface expresses multiple molecules whose sialylation state regulates multiple aspects of endothelial function. To better regulate these sialoproteins, we asked whether endothelial cells (ECs) might express one or more catalytically active sialidases. Human lung microvascular EC lysates contained heat-labile sialidase activity for a fluorogenic substrate, 2'-(4-methylumbelliferyl)-α-D-N-acetylneuraminic acid (4-MU-NANA), that was dose-dependently inhibited by the competitive sialidase inhibitor, 2,3-dehydro-2-deoxy-N-acetylneuraminic acid but not its negative control. The EC lysates also contained sialidase activity for a ganglioside mixture. Using real time RT-PCR to detect mRNAs for the four known mammalian sialidases, NEU1, -2, -3, and -4, NEU1 mRNA was expressed at levels 2700-fold higher that those found for NEU2, -3, or -4. Western analyses indicated NEU1 and -3 protein expression. Using confocal microscopy and flow cytometry, NEU1 was immunolocalized to both the plasma membrane and the perinuclear region. NEU3 was detected both in the cytosol and nucleus. Prior siRNA-mediated knockdown of NEU1 and NEU3 each decreased EC sialidase activity for 4-MU-NANA by >65 and >17%, respectively, and for the ganglioside mixture by 0 and 40%, respectively. NEU1 overexpression in ECs reduced their migration into a wound by >40%, whereas NEU3 overexpression did not. Immunohistochemical studies of normal human tissues immunolocalized NEU1 and NEU3 proteins to both pulmonary and extrapulmonary vascular endothelia. These combined data indicate that human lung microvascular ECs as well as other endothelia express catalytically active NEU1 and NEU3. NEU1 restrains EC migration, whereas NEU3 does not.
Collapse
Affiliation(s)
- Alan S Cross
- Center for Vaccine Development, University of Maryland, School of Medicine, Baltimore, Maryland 21201, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
20
|
Vilcaes AA, Demichelis VT, Daniotti JL. Trans-activity of plasma membrane-associated ganglioside sialyltransferase in mammalian cells. J Biol Chem 2011; 286:31437-46. [PMID: 21768099 DOI: 10.1074/jbc.m111.257196] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Gangliosides are acidic glycosphingolipids that contain sialic acid residues and are expressed in nearly all vertebrate cells. They are synthesized at the Golgi complex by a combination of glycosyltransferase activities followed by vesicular delivery to the plasma membrane, where they participate in a variety of physiological as well as pathological processes. Recently, a number of enzymes of ganglioside anabolism and catabolism have been shown to be associated with the plasma membrane. In particular, it was observed that CMP-NeuAc:GM3 sialyltransferase (Sial-T2) is able to sialylate GM3 at the plasma membrane (cis-catalytic activity). In this work, we demonstrated that plasma membrane-integrated ecto-Sial-T2 also displays a trans-catalytic activity at the cell surface of epithelial and melanoma cells. By using a highly sensitive enzyme-linked immunosorbent assay combined with confocal fluorescence microscopy, we observed that ecto-Sial-T2 was able to sialylate hydrophobically or covalently immobilized GM3 onto a solid surface. More interestingly, we observed that ecto-Sial-T2 was able to sialylate GM3 exposed on the membrane of neighboring cells by using both the exogenous and endogenous donor substrate (CMP-N-acetylneuraminic acid) available at the extracellular milieu. In addition, the trans-activity of ecto-Sial-T2 was considerably reduced when the expression of the acceptor substrate was inhibited by using a specific inhibitor of biosynthesis of glycolipids, indicating the lipidic nature of the acceptor. Our findings provide the first direct evidence that an ecto-sialyltransferase is able to trans-sialylate substrates exposed in the plasma membrane from mammalian cells, which represents a novel insight into the molecular events that regulate the local glycosphingolipid composition.
Collapse
Affiliation(s)
- Aldo A Vilcaes
- Centro de Investigaciones en Química Biológica de Córdoba (CIQUIBIC, UNC-CONICET), Departamento de Química Biológica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina
| | | | | |
Collapse
|
21
|
Feng C, Zhang L, Almulki L, Faez S, Whitford M, Hafezi-Moghadam A, Cross AS. Endogenous PMN sialidase activity exposes activation epitope on CD11b/CD18 which enhances its binding interaction with ICAM-1. J Leukoc Biol 2011; 90:313-21. [PMID: 21551251 DOI: 10.1189/jlb.1210708] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023] Open
Abstract
Diapedesis is a dynamic, highly regulated process by which leukocytes are recruited to inflammatory sites. We reported previously that removal of sialyl residues from PMNs enables these cells to become more adherent to EC monolayers and that sialidase activity within intracellular compartments of resting PMNs translocates to the plasma membrane following activation. We did not identify which surface adhesion molecules were targeted by endogenous sialidase. Upon activation, β2 integrin (CD11b/CD18) on the PMN surface undergoes conformational change, which allows it to bind more tightly to the ICAM-1 and ICAM-2 on the EC surface. Removal of sialyl residues from CD18 and CD11b, by exogenous neuraminidase or mobilization of PMN sialidase, unmasked activation epitopes, as detected by flow cytometry and enhanced binding to ICAM-1. One sialidase isoform, Neu1, colocalized with CD18 on confocal microscopy. Using an autoperfused microflow chamber, desialylation of immobilized ICAM-1 enhanced leukocyte arrest in vivo. Further, treatment with a sialidase inhibitor in vivo reversed endotoxin-induced binding of leukocytes to ICAM-1, thereby suggesting a role for leukocyte sialidase in the cellular arrest. These data suggest that PMN sialidase could be a physiologic source of the enzymatic activity that removes sialyl residues on β2 integrin and ICAM-1, resulting in their enhanced interaction. Thus, PMN sialidase may be an important regulator of the recruitment of these cells to inflamed sites.
Collapse
Affiliation(s)
- Chiguang Feng
- Center for Vaccine Development, Department of Medicine, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | | | | | | | | | | | | |
Collapse
|
22
|
Thiemann S, Baum LG. The road less traveled: regulation of leukocyte migration across vascular and lymphatic endothelium by galectins. J Clin Immunol 2010; 31:2-9. [PMID: 20859666 PMCID: PMC3064902 DOI: 10.1007/s10875-010-9460-z] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2010] [Accepted: 09/07/2010] [Indexed: 12/30/2022]
Abstract
Leukocyte entry from the blood into inflamed tissues, exit into the lymphatics, and migration to regional lymph nodes are all crucial processes for mounting an effective adaptive immune response. Leukocytes must cross two endothelial cell layers, the vascular and the lymphatic endothelial cell layers, during the journey from the blood to the lymph node. The proteins and cellular interactions which regulate leukocyte migration across the vascular endothelium are well studied; however, little is known about the factors that regulate leukocyte migration across the lymphatic endothelium. Here, we will summarize evidence for a role for galectins, a family of carbohydrate-binding proteins, in regulating leukocyte migration across the vascular endothelium and propose that galectins are also involved in leukocyte migration across the lymphatic endothelium.
Collapse
Affiliation(s)
- Sandra Thiemann
- Department of Pathology and Laboratory Medicine, UCLA School of Medicine, University of California, 10833 Le Conte Ave, Los Angeles, CA 90095, USA
| | | |
Collapse
|
23
|
Cabral MG, Piteira AR, Silva Z, Ligeiro D, Brossmer R, Videira PA. Human dendritic cells contain cell surface sialyltransferase activity. Immunol Lett 2010; 131:89-96. [PMID: 20206207 DOI: 10.1016/j.imlet.2010.02.009] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2009] [Revised: 02/22/2010] [Accepted: 02/24/2010] [Indexed: 11/17/2022]
Abstract
Human monocyte-derived dendritic cells (mo-DCs) express highly sialylated structures, with recognized but poorly understood function in maturation, immunogenicity and endocytosis capacity. We have previously shown that mo-DCs surface sialylation is changeable upon different stimuli, which led us to hypothesise the existence of cell surface (non-intracellular) sialyltransferases, rapidly restoring or altering mo-DC surface sialylation, thus modulating specific functions. Here, we demonstrate that, in the presence of exogenous CMP-Neu5Ac, mo-DCs incorporate considerable amounts of sialic acids into cell surface, predominantly when mo-DCs were previously desialylated or matured. This is a genuine sialyltransferase activity, confirmed by specific inhibition assays, which is not influenced by secreted enzymes. Functionally, the ecto-sialyltransferase activity causes a significant down-regulation of mo-DCs endocytic capacity, without affecting the maturation state. These findings suggest that ecto-sialyltransferases participate in a dynamic control of mo-DC sialylation, with functional repercussions. This activity is possibly related with specific physiological and pathological conditions, as inflammation and infection, contributing to protection and homeostasis regulation.
Collapse
Affiliation(s)
- M Guadalupe Cabral
- CEDOC, Departamento de Imunologia, Faculdade de Ciências Médicas, FCM, Universidade Nova de Lisboa, Campo Mártires da Pátria 130, Lisbon, Portugal
| | | | | | | | | | | |
Collapse
|