1
|
Pilling D, Martinez TC, Gomer RH. Inhibition of CCl4-induced liver inflammation and fibrosis by a NEU3 inhibitor. PLoS One 2024; 19:e0308060. [PMID: 39570922 PMCID: PMC11581222 DOI: 10.1371/journal.pone.0308060] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2024] [Accepted: 09/26/2024] [Indexed: 11/24/2024] Open
Abstract
Sialic acids are located on the ends of many glycoconjugates and are cleaved off by enzymes called sialidases (neuraminidases). Upregulation of neuraminidase 3 (NEU3) is associated with intestinal inflammation and colitis, neuroinflammation, and lung fibrosis. Genetic ablation of NEU3 or pharmacological inhibition of NEU3 reduces lung fibrosis in mice. To determine if inhibiting NEU3 can inhibit liver fibrosis in the commonly-used CCl4 model, in this report, we examined the effects of injections of the NEU3 inhibitor 2-acetyl pyridine (2AP). 2AP inhibited CCl4-induced weight loss in female but not male mice. 2AP attenuated CCl4-induced liver inflammation and fibrosis in male and female mice, but did not affect CCl4-induced steatosis. After CCl4 treatment, female but not male mice had significant increases in liver neutrophils, and 2AP attenuated this response. 2AP also reversed CCl4-induced liver desialylation and CCl4-induced increased expression of NEU3. Patients with pulmonary fibrosis have increased desialylation of some serum proteins, and elevated serum levels of NEU3. We find that sera from patients with nonalcoholic fatty liver disease (NAFLD) and nonalcoholic steatohepatitis (NASH) have elevated desialylation of a serum protein and patients with NAFLD have increased levels of NEU3. These data suggest that elevated levels of NEU3 may be associated with liver inflammation and fibrosis, and that in mice this is ameliorated by injections of a NEU3 inhibitor.
Collapse
Affiliation(s)
- Darrell Pilling
- Department of Biology, Texas A&M University, College Station, Texas, United States of America
| | - Trevor C. Martinez
- Department of Biology, Texas A&M University, College Station, Texas, United States of America
| | - Richard H. Gomer
- Department of Biology, Texas A&M University, College Station, Texas, United States of America
| |
Collapse
|
2
|
Lillehoj EP, Yu Y, Verceles AC, Imamura A, Ishida H, Piepenbrink KH, Goldblum SE. Stenotrophomonas maltophilia provokes NEU1-mediated release of a flagellin-binding decoy receptor that protects against lethal infection. iScience 2024; 27:110866. [PMID: 39314239 PMCID: PMC11418149 DOI: 10.1016/j.isci.2024.110866] [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] [Received: 04/10/2024] [Revised: 08/03/2024] [Accepted: 08/29/2024] [Indexed: 09/25/2024] Open
Abstract
Stenotrophomonas maltophilia (Sm), a multidrug-resistant pathogen often isolated from immunocompromised individuals, presents its flagellin to multimeric tandem repeats within the ectodomain of mucin-1 (MUC1-ED), expressed on airway epithelia. Flagellated Sm increases neuraminidase-1 (NEU1) sialidase association with and desialylation of MUC1-ED. This NEU1-mediated MUC1-ED desialylation unmasks cryptic binding sites for Sm flagellin, increasing flagellin and Sm binding to airway epithelia. MUC1 overexpression increases receptor number whereas NEU1 overexpression elevates receptor binding affinity. Silencing of either MUC1 or NEU1 reduces the flagellin-MUC1 interaction. Sm/flagellin provokes MUC1-ED autoproteolysis at a juxtamembranous glycine-serine peptide bond. MUC1-ED shedding from the epithelium not only occurs in vitro, but in the bronchoalveolar compartments of Sm/flagellin-challenged mice and patients with ventilator-associated Sm pneumonia. Finally, the soluble flagellin-targeting, MUC1-ED decoy receptor dose-dependently inhibits multiple Sm flagellin-driven pathogenic processes, in vitro, including motility, biofilm formation, adhesion, and proinflammatory cytokine production, and protects against lethal Sm lung infection, in vivo.
Collapse
Affiliation(s)
- Erik P. Lillehoj
- Department of Pediatrics, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Yafan Yu
- Department of Biochemistry, University of Nebraska, Lincoln, NE, USA
| | - Avelino C. Verceles
- Department of Medicine, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Akihiro Imamura
- Institute for Glyco-core Research (iGCORE), Gifu University, Gifu, Japan
| | - Hideharu Ishida
- Institute for Glyco-core Research (iGCORE), Gifu University, Gifu, Japan
| | - Kurt H. Piepenbrink
- Department of Biochemistry, University of Nebraska, Lincoln, NE, USA
- Department of Food Science and Technology, University of Nebraska, Lincoln, NE, USA
- Department of Chemistry, University of Nebraska, Lincoln, NE, USA
| | - Simeon E. Goldblum
- Department of Medicine, University of Maryland School of Medicine, Baltimore, MD, USA
| |
Collapse
|
3
|
Irons EE, Sajina GC, Lau JT. Sialic acid in the regulation of blood cell production, differentiation and turnover. Immunology 2024; 172:517-532. [PMID: 38503445 PMCID: PMC11223974 DOI: 10.1111/imm.13780] [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: 11/27/2023] [Accepted: 03/04/2024] [Indexed: 03/21/2024] Open
Abstract
Sialic acid is a unique sugar moiety that resides in the distal and most accessible position of the glycans on mammalian cell surface and extracellular glycoproteins and glycolipids. The potential for sialic acid to obscure underlying structures has long been postulated, but the means by which such structural changes directly affect biological processes continues to be elucidated. Here, we appraise the growing body of literature detailing the importance of sialic acid for the generation, differentiation, function and death of haematopoietic cells. We conclude that sialylation is a critical post-translational modification utilized in haematopoiesis to meet the dynamic needs of the organism by enforcing rapid changes in availability of lineage-specific cell types. Though long thought to be generated only cell-autonomously within the intracellular ER-Golgi secretory apparatus, emerging data also demonstrate previously unexpected diversity in the mechanisms of sialylation. Emphasis is afforded to the mechanism of extrinsic sialylation, whereby extracellular enzymes remodel cell surface and extracellular glycans, supported by charged sugar donor molecules from activated platelets.
Collapse
Affiliation(s)
| | | | - Joseph T.Y. Lau
- Department of Molecular and Cellular Biology, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14203 USA
| |
Collapse
|
4
|
Zamanian MY, Alsaab HO, Golmohammadi M, Yumashev A, Jabba AM, Abid MK, Joshi A, Alawadi AH, Jafer NS, Kianifar F, Obakiro SB. NF-κB pathway as a molecular target for curcumin in diabetes mellitus treatment: Focusing on oxidative stress and inflammation. Cell Biochem Funct 2024; 42:e4030. [PMID: 38720663 DOI: 10.1002/cbf.4030] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2024] [Revised: 04/05/2024] [Accepted: 04/25/2024] [Indexed: 08/03/2024]
Abstract
Diabetes mellitus (DM) is a collection of metabolic disorder that is characterized by chronic hyperglycemia. Recent studies have demonstrated the crucial involvement of oxidative stress (OS) and inflammatory reactions in the development of DM. Curcumin (CUR), a natural compound derived from turmeric, exerts beneficial effects on diabetes mellitus through its interaction with the nuclear factor kappa B (NF-κB) pathway. Research indicates that CUR targets inflammatory mediators in diabetes, including tumor necrosis factor α (TNF-α) and interleukin-6 (IL-6), by modulating the NF-κB signaling pathway. By reducing the expression of these inflammatory factors, CUR demonstrates protective effects in DM by improving pancreatic β-cells function, normalizing inflammatory cytokines, reducing OS and enhancing insulin sensitivity. The findings reveal that CUR administration effectively lowered blood glucose elevation, reinstated diminished serum insulin levels, and enhanced body weight in Streptozotocin -induced diabetic rats. CUR exerts its beneficial effects in management of diabetic complications through regulation of signaling pathways, such as calcium-calmodulin (CaM)-dependent protein kinase II (CaMKII), peroxisome proliferator-activated receptor gamma (PPAR-γ), NF-κB, and transforming growth factor β1 (TGFB1). Moreover, CUR reversed the heightened expression of inflammatory cytokines (TNF-α, Interleukin-1 beta (IL-1β), IL-6) and chemokines like MCP-1 in diabetic specimens, vindicating its anti-inflammatory potency in counteracting hyperglycemia-induced alterations. CUR diminishes OS, avert structural kidney damage linked to diabetic nephropathy, and suppress NF-κB activity. Furthermore, CUR exhibited a protective effect against diabetic cardiomyopathy, lung injury, and diabetic gastroparesis. Conclusively, the study posits that CUR could potentially offer therapeutic benefits in relieving diabetic complications through its influence on the NF-κB pathway.
Collapse
Affiliation(s)
- Mohammad Yasin Zamanian
- Department of Physiology, Hamadan University of Medical Sciences, Hamadan, Iran
- Department of Pharmacology and Toxicology, School of Pharmacy, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Hashem O Alsaab
- Department of Pharmaceutics and Pharmaceutical Technology, Taif University, Taif, Saudi Arabia
| | - Maryam Golmohammadi
- School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Alexey Yumashev
- Department of Prosthetic Dentistry, Sechenov First Moscow State Medical University, Moscow, Russia
| | - Abeer Mhussan Jabba
- Colleges of Pharmacy, National University of Science and Technology, Dhi Qar, Iraq
| | - Mohammed Kadhem Abid
- Department of Anesthesia, College of Health & Medical Technology, Al-Ayen University, Nasiriyah, Iraq
| | - Abhishek Joshi
- Department of Liberal Arts School of Liberal Arts, Uttaranchal University, Dehradun, India
| | - Ahmed Hussien Alawadi
- College of Technical Engineering, The Islamic University, Najaf, Iraq
- College of Technical Engineering, The Islamic University of Al Diwaniyah, Al Diwaniyah, Iraq
- College of Technical Engineering, The Islamic University of Babylon, Babylon, Iraq
| | - Noor S Jafer
- Department of Medical Laboratory Technologies, Al Rafidain University College, Bagdad, Iraq
| | - Farzaneh Kianifar
- School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Samuel Baker Obakiro
- Department of Pharmacology and Therapeutics, Faculty of Health Sciences, Busitema University, Mbale, Uganda
| |
Collapse
|
5
|
Cao Y, Rische CH, Bochner BS, O’Sullivan JA. Interactions between Siglec-8 and endogenous sialylated cis ligands restrain cell death induction in human eosinophils and mast cells. Front Immunol 2023; 14:1283370. [PMID: 37928558 PMCID: PMC10623328 DOI: 10.3389/fimmu.2023.1283370] [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: 08/25/2023] [Accepted: 10/06/2023] [Indexed: 11/07/2023] Open
Abstract
Sialic acid-binding immunoglobulin-like lectin (Siglec)-8 is a sialoside-binding receptor expressed by eosinophils and mast cells that exhibits priming status- and cell type-dependent inhibitory activity. On eosinophils that have been primed with IL-5, GM-CSF, or IL-33, antibody ligation of Siglec-8 induces cell death through a pathway involving the β2 integrin-dependent generation of reactive oxygen species (ROS) via NADPH oxidase. In contrast, Siglec-8 engagement on mast cells inhibits cellular activation and mediator release but reportedly does not impact cell viability. The differences in responses between cytokine-primed and unprimed eosinophils, and between eosinophils and mast cells, to Siglec-8 ligation are not understood. We previously found that Siglec-8 binds to sialylated ligands present on the surface of the same cell (so-called cis ligands), preventing Siglec-8 ligand binding in trans. However, the functional relevance of these cis ligands has not been elucidated. We therefore explored the potential influence of cis ligands of Siglec-8 on both eosinophils and mast cells. De-sialylation using exogenous sialidase profoundly altered the consequences of Siglec-8 antibody engagement on both cell types, eliminating the need for cytokine priming of eosinophils to facilitate cell death and enabling Siglec-8-dependent mast cell death without impacting anti-Siglec-8 antibody binding. The cell death process licensed by de-sialylation resembled that characterized in IL-5-primed eosinophils, including CD11b upregulation, ROS production, and the activities of Syk, PI3K, and PLC. These results implicate cis ligands in restraining Siglec-8 function on eosinophils and mast cells and reveal a promising approach to the selective depletion of mast cells in patients with mast cell-mediated diseases.
Collapse
Affiliation(s)
- Yun Cao
- Division of Allergy and Immunology, Northwestern University Feinberg School of Medicine, Chicago, IL, United States
| | - Clayton H. Rische
- Division of Allergy and Immunology, Northwestern University Feinberg School of Medicine, Chicago, IL, United States
- Department of Biomedical Engineering, Northwestern University McCormick School of Engineering, Evanston, IL, United States
| | - Bruce S. Bochner
- Division of Allergy and Immunology, Northwestern University Feinberg School of Medicine, Chicago, IL, United States
| | - Jeremy A. O’Sullivan
- Division of Allergy and Immunology, Northwestern University Feinberg School of Medicine, Chicago, IL, United States
| |
Collapse
|
6
|
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: 6] [Impact Index Per Article: 3.0] [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
|
7
|
Conley HE, Sheats MK. Targeting Neutrophil β 2-Integrins: A Review of Relevant Resources, Tools, and Methods. Biomolecules 2023; 13:892. [PMID: 37371473 DOI: 10.3390/biom13060892] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2023] [Revised: 05/15/2023] [Accepted: 05/23/2023] [Indexed: 06/29/2023] Open
Abstract
Neutrophils are important innate immune cells that respond during inflammation and infection. These migratory cells utilize β2-integrin cell surface receptors to move out of the vasculature into inflamed tissues and to perform various anti-inflammatory responses. Although critical for fighting off infection, neutrophil responses can also become dysregulated and contribute to disease pathophysiology. In order to limit neutrophil-mediated damage, investigators have focused on β2-integrins as potential therapeutic targets, but so far these strategies have failed in clinical trials. As the field continues to move forward, a better understanding of β2-integrin function and signaling will aid the design of future therapeutics. Here, we provide a detailed review of resources, tools, experimental methods, and in vivo models that have been and will continue to be utilized to investigate the vitally important cell surface receptors, neutrophil β2-integrins.
Collapse
Affiliation(s)
- Haleigh E Conley
- Department of Clinical Sciences, College of Veterinary Medicine, North Carolina State University, Raleigh, NC 27607, USA
- Comparative Medicine Institute, North Carolina State University, Raleigh, NC 27607, USA
| | - M Katie Sheats
- Department of Clinical Sciences, College of Veterinary Medicine, North Carolina State University, Raleigh, NC 27607, USA
- Comparative Medicine Institute, North Carolina State University, Raleigh, NC 27607, USA
| |
Collapse
|
8
|
Feng C, Cross AS, Vasta GR. Galectin-1 mediates interactions between polymorphonuclear leukocytes and vascular endothelial cells, and promotes their extravasation during lipopolysaccharide-induced acute lung injury. Mol Immunol 2023; 156:127-135. [PMID: 36921487 PMCID: PMC10154945 DOI: 10.1016/j.molimm.2023.02.011] [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: 10/17/2022] [Revised: 01/29/2023] [Accepted: 02/26/2023] [Indexed: 03/14/2023]
Abstract
The lung airway epithelial surface is heavily covered with sialic acids as the terminal carbohydrate on most cell surface glycoconjugates and can be removed by microbial neuraminidases or endogenous sialidases. By desialylating the lung epithelial surface, neuraminidase acts as an important virulence factor in many mucosal pathogens, such as influenza and S. pneumoniae. Desialylation exposes the subterminal galactosyl moieties - the binding glycotopes for galectins, a family of carbohydrate-recognition proteins playing important roles in various aspects of immune responses. Galectin-1 and galectin-3 have been extensively studied in their roles related to host immune responses, but some questions about their role(s) in leukocyte recruitment during lung bacterial infection remain unanswered. In this study, we found that both galectin-1 and galectin-3 bind to polymorphonuclear leukocytes (PMNs) and enhance the interaction of endothelial intercellular adhesion molecule-1 (ICAM-1) with PMNs, which is further increased by PMN desialylation. In addition, we observed that in vitro galectin-1 mediates the binding of PMNs, particularly desialylated PMNs, onto the endothelial cells. Finally, in a murine model for LPS-mediated acute lung injury, we observed that galectin-1 modulates PMN infiltration to the lung without altering the expression of chemoattractant cytokines. We conclude that galectins, particularly galectin-1, may function as adhesion molecules that mediate PMN-endothelial cell interactions, and modulate PMN infiltration during acute lung injury.
Collapse
Affiliation(s)
- Chiguang Feng
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, MD, USA; Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore, MD, USA.
| | - Alan S Cross
- Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore, MD, USA; Department of Medicine, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Gerardo R Vasta
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, MD, USA; Institute of Marine and Environmental Technology, University of Maryland, Baltimore, MD, USA
| |
Collapse
|
9
|
Azcutia V, Kelm M, Fink D, Cummings RD, Nusrat A, Parkos CA, Brazil JC. Sialylation regulates neutrophil transepithelial migration, CD11b/CD18 activation, and intestinal mucosal inflammatory function. JCI Insight 2023; 8:e167151. [PMID: 36719745 PMCID: PMC10077474 DOI: 10.1172/jci.insight.167151] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2022] [Accepted: 01/25/2023] [Indexed: 02/01/2023] Open
Abstract
Polymorphonuclear neutrophils (PMNs) play a critical role in clearing invading microbes and promoting tissue repair following infection/injury. However, dysregulated PMN trafficking and associated tissue damage is pathognomonic of numerous inflammatory mucosal diseases. The final step in PMN influx into mucosal lined organs (including the lungs, kidneys, skin, and gut) involves transepithelial migration (TEpM). The β2-integrin CD11b/CD18 plays an important role in mediating PMN intestinal trafficking, with recent studies highlighting that terminal fucose and GlcNAc glycans on CD11b/CD18 can be targeted to reduce TEpM. However, the role of the most abundant terminal glycan, sialic acid (Sia), in regulating PMN epithelial influx and mucosal inflammatory function is not well understood. Here we demonstrate that inhibiting sialidase-mediated removal of α2-3-linked Sia from CD11b/CD18 inhibits PMN migration across intestinal epithelium in vitro and in vivo. Sialylation was also found to regulate critical PMN inflammatory effector functions, including degranulation and superoxide release. Finally, we demonstrate that sialidase inhibition reduces bacterial peptide-mediated CD11b/CD18 activation in PMN and blocks downstream intracellular signaling mediated by spleen tyrosine kinase (Syk) and p38 MAPK. These findings suggest that sialylated glycans on CD11b/CD18 represent potentially novel targets for ameliorating PMN-mediated tissue destruction in inflammatory mucosal diseases.
Collapse
Affiliation(s)
- Veronica Azcutia
- Department of Pathology, University of Michigan, Ann Arbor, Michigan, USA
| | - Matthias Kelm
- Department of Pathology, University of Michigan, Ann Arbor, Michigan, USA
| | - Dylan Fink
- Department of Pathology, University of Michigan, Ann Arbor, Michigan, USA
| | - Richard D. Cummings
- Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA
| | - Asma Nusrat
- Department of Pathology, University of Michigan, Ann Arbor, Michigan, USA
| | - Charles A. Parkos
- Department of Pathology, University of Michigan, Ann Arbor, Michigan, USA
| | - Jennifer C. Brazil
- Department of Pathology, University of Michigan, Ann Arbor, Michigan, USA
| |
Collapse
|
10
|
Kirolos SA, Pilling D, Gomer RH. The extracellular sialidase NEU3 primes neutrophils. J Leukoc Biol 2022; 112:1399-1411. [PMID: 35899930 PMCID: PMC9701152 DOI: 10.1002/jlb.3a0422-217rr] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2022] [Revised: 06/28/2022] [Indexed: 01/04/2023] Open
Abstract
Some extracellular glycoconjugates have sialic acid as the terminal sugar, and sialidases are enzymes that remove this sugar. Mammals have 4 sialidases and can be elevated in inflammation and fibrosis. In this report, we show that incubation of human neutrophils with the extracellular human sialidase NEU3, but not NEU1, NEU2 or NEU4, induces human male and female neutrophils to change from a round to a more amoeboid morphology, causes the primed human neutrophil markers CD11b, CD18, and CD66a to localize to the cell cortex, and decreases the localization of the unprimed human neutrophil markers CD43 and CD62-L at the cell cortex. NEU3, but not the other 3 sialidases, also causes human male and female neutrophils to increase their F-actin content. Human neutrophils treated with NEU3 show a decrease in cortical levels of Sambucus nigra lectin staining and an increase in cortical levels of peanut agglutinin staining, indicating a NEU3-induced desialylation. The inhibition of NEU3 by the NEU3 inhibitor 2-acetylpyridine attenuated the NEU3 effect on neutrophil morphology, indicating that the effect of NEU3 is dependent on its enzymatic activity. Together, these results indicate that NEU3 can prime human male and female neutrophils, and that NEU3 is a potential regulator of inflammation.
Collapse
Affiliation(s)
- Sara A Kirolos
- Department of Biology, Texas A&M University, College Station, Texas, USA
| | - Darrell Pilling
- Department of Biology, Texas A&M University, College Station, Texas, USA
| | - Richard H Gomer
- Department of Biology, Texas A&M University, College Station, Texas, USA
| |
Collapse
|
11
|
Keil J, Rafn GR, Turan IM, Aljohani MA, Sahebjam-Atabaki R, Sun XL. Sialidase Inhibitors with Different Mechanisms. J Med Chem 2022; 65:13574-13593. [PMID: 36252951 PMCID: PMC9620260 DOI: 10.1021/acs.jmedchem.2c01258] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Indexed: 11/28/2022]
Abstract
Sialidases, or neuraminidases, are enzymes that catalyze the hydrolysis of sialic acid (Sia)-containing molecules, mostly removal of the terminal Sia (desialylation). By desialylation, sialidase can modulate the functionality of the target compound and is thus often involved in biological pathways. Inhibition of sialidases with inhibitors is an important approach for understanding sialidase function and the underlying mechanisms and could serve as a therapeutic approach as well. Transition-state analogues, such as anti-influenza drugs oseltamivir and zanamivir, are major sialidase inhibitors. In addition, difluoro-sialic acids were developed as mechanism-based sialidase inhibitors. Further, fluorinated quinone methide-based suicide substrates were reported. Sialidase product analogue inhibitors were also explored. Finally, natural products have shown competitive inhibiton against viral, bacterial, and human sialidases. This Perspective describes sialidase inhibitors with different mechanisms and their activities and future potential, which include transition-state analogue inhibitors, mechanism-based inhibitors, suicide substrate inhibitors, product analogue inhibitors, and natural product inhibitors.
Collapse
Affiliation(s)
- Joseph
M. Keil
- Department of Chemistry, Chemical and
Biomedical Engineering and Center for Gene Regulation in Health and
Disease (GRHD), Cleveland State University, Cleveland, Ohio 44115, United States
| | - Garrett R. Rafn
- Department of Chemistry, Chemical and
Biomedical Engineering and Center for Gene Regulation in Health and
Disease (GRHD), Cleveland State University, Cleveland, Ohio 44115, United States
| | - Isaac M. Turan
- Department of Chemistry, Chemical and
Biomedical Engineering and Center for Gene Regulation in Health and
Disease (GRHD), Cleveland State University, Cleveland, Ohio 44115, United States
| | - Majdi A. Aljohani
- Department of Chemistry, Chemical and
Biomedical Engineering and Center for Gene Regulation in Health and
Disease (GRHD), Cleveland State University, Cleveland, Ohio 44115, United States
| | - Reza Sahebjam-Atabaki
- Department of Chemistry, Chemical and
Biomedical Engineering and Center for Gene Regulation in Health and
Disease (GRHD), Cleveland State University, Cleveland, Ohio 44115, United States
| | - Xue-Long Sun
- Department of Chemistry, Chemical and
Biomedical Engineering and Center for Gene Regulation in Health and
Disease (GRHD), Cleveland State University, Cleveland, Ohio 44115, United States
| |
Collapse
|
12
|
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: 1.3] [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
|
13
|
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
|
14
|
Myeloid-Specific Pyruvate-Kinase-Type-M2-Deficient Mice Are Resistant to Acute Lung Injury. Biomedicines 2022; 10:biomedicines10051193. [PMID: 35625931 PMCID: PMC9138865 DOI: 10.3390/biomedicines10051193] [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: 03/05/2022] [Revised: 05/17/2022] [Accepted: 05/19/2022] [Indexed: 02/05/2023] Open
Abstract
Infiltration of polymorphonuclear neutrophils (PMNs) plays a central role in acute lung injury (ALI). The mechanisms governing PMN inflammatory responses, however, remain incompletely understood. Based on our recent study showing a non-metabolic role of pyruvate kinase type M2 (PKM2) in controlling PMN degranulation of secondary and tertiary granules and consequent chemotaxis, here we tested a hypothesis that Pkm2-deficient mice may resist ALI due to impaired PMN inflammatory responses. We found that PMN aerobic glycolysis controlled the degranulation of secondary and tertiary granules induced by fMLP and PMA. Compared to WT PMNs, Pkm2-deficient (Pkm2-/-) PMNs displayed significantly less capacity for fMLP- or PMA-induced degranulation of secondary and tertiary granules, ROS production, and transfilter migration. In line with this, myeloid-specific Pkm2-/- mice exhibited impaired zymosan-induced PMN infiltration in the peritoneal cavity. Employing an LPS-induced ALI mouse model, LPS-treated Pkm2-/- mice displayed significantly less infiltration of inflammatory PMNs in the alveolar space and a strong resistance to LPS-induced ALI. Our results thus reveal that PKM2 is required for PMN inflammatory responses and deletion of PKM2 in PMN leads to an impaired PMN function but protection against LPS-induced ALI.
Collapse
|
15
|
Lillehoj EP, Luzina IG, Atamas SP. Mammalian Neuraminidases in Immune-Mediated Diseases: Mucins and Beyond. Front Immunol 2022; 13:883079. [PMID: 35479093 PMCID: PMC9035539 DOI: 10.3389/fimmu.2022.883079] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Accepted: 03/21/2022] [Indexed: 12/28/2022] Open
Abstract
Mammalian neuraminidases (NEUs), also known as sialidases, are enzymes that cleave off the terminal neuraminic, or sialic, acid resides from the carbohydrate moieties of glycolipids and glycoproteins. A rapidly growing body of literature indicates that in addition to their metabolic functions, NEUs also regulate the activity of their glycoprotein targets. The simple post-translational modification of NEU protein targets-removal of the highly electronegative sialic acid-affects protein folding, alters protein interactions with their ligands, and exposes or covers proteolytic sites. Through such effects, NEUs regulate the downstream processes in which their glycoprotein targets participate. A major target of desialylation by NEUs are mucins (MUCs), and such post-translational modification contributes to regulation of disease processes. In this review, we focus on the regulatory roles of NEU-modified MUCs as coordinators of disease pathogenesis in fibrotic, inflammatory, infectious, and autoimmune diseases. Special attention is placed on the most abundant and best studied NEU1, and its recently discovered important target, mucin-1 (MUC1). The role of the NEU1 - MUC1 axis in disease pathogenesis is discussed, along with regulatory contributions from other MUCs and other pathophysiologically important NEU targets.
Collapse
Affiliation(s)
- Erik P. Lillehoj
- Department of Pediatrics, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Irina G. Luzina
- Department of Medicine, University of Maryland School of Medicine, Baltimore, MD, United States
- Research Service, Baltimore Veterans Affairs (VA) Medical Center, Baltimore, MD, United States
| | - Sergei P. Atamas
- Department of Medicine, University of Maryland School of Medicine, Baltimore, MD, United States
| |
Collapse
|
16
|
Abstract
Coronary atherosclerosis is a chronic inflammatory disease that can lead to varying degrees of blood flow obstruction and a common pathophysiological basis of cardiovascular disease. Inflammatory factors run through the whole process of atherosclerotic lesions. Macrophages, T cells, and neutrophils play important roles in the process of atherosclerotic inflammation. Considering the evolutionary characteristics, atherosclerosis can be divided into different stages as early atherosclerotic plaque, plaque formation stage, and plaque rupture stage. In this paper, the changes in inflammatory cells at different stages of lesions and their related mechanisms are discussed, which can provide new insights from a clinical to bench perspective for atherosclerosis me chanism.
Collapse
|
17
|
Howlader MA, Demina EP, Samarani S, Guo T, Caillon A, Ahmad A, Pshezhetsky AV, Cairo CW. The Janus-like role of neuraminidase isoenzymes in inflammation. FASEB J 2022; 36:e22285. [PMID: 35363389 PMCID: PMC9323473 DOI: 10.1096/fj.202101218r] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2021] [Revised: 02/09/2022] [Accepted: 03/17/2022] [Indexed: 01/13/2023]
Abstract
The processes of activation, extravasation, and migration of immune cells to a site are early and essential steps in the induction of an acute inflammatory response. These events are an essential part of the inflammatory cascade, which involves multiple regulatory steps. Using a murine air pouch model of inflammation with LPS as an inflammation inducer, we demonstrate that isoenzymes of the neuraminidase family (NEU1, 3, and 4) play essential roles in these processes by acting as positive or negative regulators of leukocyte infiltration. In genetically knocked‐out (KO) mice for different NEU genes (Neu1 KO, Neu3 KO, Neu4 KO, and Neu3/4 double KO mice) with LPS‐induced air pouch inflammation, leukocytes at the site of inflammation were counted, and the inflamed tissue was analyzed using immunohistochemistry. Our data show that leukocyte recruitment was decreased in NEU1‐ and NEU3‐deficient mice, while it was increased in NEU4‐deficient animals. Consistent with these results, systemic as well as pouch exudate levels of pro‐inflammatory cytokines were reduced in Neu1 and increased in Neu4 KO mice. Pharmacological inhibitors specific for NEU1, NEU3, and NEU4 isoforms also affected leukocyte recruitment. Together our data demonstrate that NEU isoenzymes have distinct—and even opposing—effects on leukocyte recruitment, and therefore warrant further investigation to determine their mechanisms and importance as regulators of the inflammatory cascade.
Collapse
Affiliation(s)
- Md Amran Howlader
- Department of Chemistry, University of Alberta, Edmonton, Alberta, Canada
| | - Ekaterina P Demina
- Division of Medical Genetics, Sainte-Justine Hospital Research Center, University of Montreal, Montreal, Quebec, Canada
| | - Suzanne Samarani
- Department of Microbiology, Infectious Diseases & Immunology, Sainte-Justine Hospital Research Center, University of Montreal, Montreal, Quebec, Canada
| | - Tianlin Guo
- Department of Chemistry, University of Alberta, Edmonton, Alberta, Canada
| | - Antoine Caillon
- Division of Medical Genetics, Sainte-Justine Hospital Research Center, University of Montreal, Montreal, Quebec, Canada
| | - Ali Ahmad
- Department of Microbiology, Infectious Diseases & Immunology, Sainte-Justine Hospital Research Center, University of Montreal, Montreal, Quebec, Canada
| | - Alexey V Pshezhetsky
- Division of Medical Genetics, Sainte-Justine Hospital Research Center, University of Montreal, Montreal, Quebec, Canada.,Department of Anatomy and Cell Biology, McGill University, Montreal, Quebec, Canada
| | | |
Collapse
|
18
|
Miura S, Habibabady ZA, Pollok F, Connolly M, Pratts S, Dandro A, Sorrells L, Karavi K, Phelps C, Eyestone W, Ayares D, Burdorf L, Azimzadeh A, Pierson RN. Effects of human TFPI and CD47 expression and selectin and integrin inhibition during GalTKO.hCD46 pig lung perfusion with human blood. Xenotransplantation 2022; 29:e12725. [PMID: 35234315 PMCID: PMC10207735 DOI: 10.1111/xen.12725] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2021] [Revised: 10/30/2021] [Accepted: 12/17/2021] [Indexed: 01/07/2023]
Abstract
BACKGROUND Loss of barrier function when GalTKO.hCD46 porcine lungs are perfused with human blood is associated with coagulation pathway dysregulation, innate immune system activation, and rapid sequestration of human formed blood elements. Here, we evaluate whether genetic expression of human tissue factor pathway inhibitor (hTFPI) and human CD47 (hCD47), alone or with combined selectin and integrin adhesion pathway inhibitors, delays GalTKO.hCD46 porcine lung injury or modulates neutrophil and platelet sequestration. METHODS In a well-established paired ex vivo lung perfusion model, GalTKO.hCD46.hTFPI.hCD47 transgenic porcine lungs (hTFPI.hCD47, n = 7) were compared to GalTKO.hCD46 lungs (reference, n = 5). All lung donor pigs were treated with a thromboxane synthase inhibitor, anti-histamine, and anti-GPIb integrin-blocking Fab, and were pre-treated with Desmopressin. In both genotypes, one lung of each pair was additionally treated with PSGL-1 and GMI-1271 (P- and E-selectin) and IB4 (CD11b/18 integrin) adhesion inhibitors (n = 6 hTFPI.hCD47, n = 3 reference). RESULTS All except for two reference lungs did not fail within 480 min when experiments were electively terminated. Selectin and integrin adhesion inhibitors moderately attenuated initial pulmonary vascular resistance (PVR) elevation in hTFPI.hCD47 lungs. Neutrophil sequestration was significantly delayed during the early time points following reperfusion and terminal platelet activation was attenuated in association with lungs expressing hTFPI.hCD47, but additional adhesion pathway inhibitors did not show further effects with either lung genotype. CONCLUSION Expression of hTFPI.hCD47 on porcine lung may be useful as part of an integrated strategy to prevent neutrophil adhesion and platelet activation that are associated with xenograft injury. Additionally, targeting canonical selectin and integrin adhesion pathways reduced PVR elevation associated with hTFPI.hCD47 expression, but did not significantly attenuate neutrophil or platelet sequestration. We conclude that other adhesive mechanisms mediate the residual sequestration of human formed blood elements to pig endothelium that occurs even in the context of the multiple genetic modifications and drug treatments tested here.
Collapse
Affiliation(s)
- Shuhei Miura
- Center for Transplantation Sciences, Department of Surgery, Massachusetts General Hospital, Boston, Massachusetts, USA
- Department of Cardiovascular Surgery, Teine Keijinkai Hospital, Sapporo, Japan
| | - Zahra A. Habibabady
- Center for Transplantation Sciences, Department of Surgery, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Franziska Pollok
- Center for Transplantation Sciences, Department of Surgery, Massachusetts General Hospital, Boston, Massachusetts, USA
- Department of Anesthesiology, University Hospital Hamburg-Eppendorf, Hamburg, Germany
| | - Margaret Connolly
- Center for Transplantation Sciences, Department of Surgery, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Shannon Pratts
- Center for Transplantation Sciences, Department of Surgery, Massachusetts General Hospital, Boston, Massachusetts, USA
| | | | | | | | | | | | | | - Lars Burdorf
- Center for Transplantation Sciences, Department of Surgery, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Agnes Azimzadeh
- Center for Transplantation Sciences, Department of Surgery, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Richard N. Pierson
- Center for Transplantation Sciences, Department of Surgery, Massachusetts General Hospital, Boston, Massachusetts, USA
| |
Collapse
|
19
|
Yu L, Peng J, Mineo C. Lipoprotein sialylation in atherosclerosis: Lessons from mice. Front Endocrinol (Lausanne) 2022; 13:953165. [PMID: 36157440 PMCID: PMC9498574 DOI: 10.3389/fendo.2022.953165] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Accepted: 08/15/2022] [Indexed: 11/22/2022] Open
Abstract
Sialylation is a dynamically regulated modification, which commonly occurs at the terminal of glycan chains in glycoproteins and glycolipids in eukaryotic cells. Sialylation plays a key role in a wide array of biological processes through the regulation of protein-protein interactions, intracellular localization, vesicular trafficking, and signal transduction. A majority of the proteins involved in lipoprotein metabolism and atherogenesis, such as apolipoproteins and lipoprotein receptors, are sialylated in their glycan structures. Earlier studies in humans and in preclinical models found a positive correlation between low sialylation of lipoproteins and atherosclerosis. More recent works using loss- and gain-of-function approaches in mice have revealed molecular and cellular mechanisms by which protein sialylation modulates causally the process of atherosclerosis. The purpose of this concise review is to summarize these findings in mouse models and to provide mechanistic insights into lipoprotein sialylation and atherosclerosis.
Collapse
Affiliation(s)
- Liming Yu
- Center for Pulmonary and Vascular Biology, Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX, United States
| | - Jun Peng
- Center for Pulmonary and Vascular Biology, Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX, United States
| | - Chieko Mineo
- Center for Pulmonary and Vascular Biology, Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX, United States
- Department of Cell Biology, University of Texas Southwestern Medical Center, Dallas, TX, United States
- *Correspondence: Chieko Mineo,
| |
Collapse
|
20
|
Bocquet O, Tembely D, Rioult D, Terryn C, Romier B, Bennasroune A, Blaise S, Sartelet H, Martiny L, Duca L, Maurice P. Characterization of novel interactions with membrane NEU1 highlights new regulatory functions for the Elastin Receptor Complex in monocyte interaction with endothelial cells. Cell Biosci 2021; 11:206. [PMID: 34903296 PMCID: PMC8670255 DOI: 10.1186/s13578-021-00718-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2021] [Accepted: 11/20/2021] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Vascular aging is associated with remodeling of elastin, one of the main extracellular matrix component of the arterial wall, and production of elastin-derived peptides (EDP). These extracellular matrix degradation products have been shown to trigger biological activities through the elastin receptor complex (ERC) and data from the last decade have brought significant insights on the critical role played by its NEU1 subunit in the biological effects mediated by EDP and the ERC in vascular and metabolic diseases. RESULTS Using a proteomic approach, we previously identified new potential interaction partners of membrane NEU1. Here, we validated the interaction between NEU1 and the β2 integrin in human monocytes and show that binding of EDP to the ERC leads to desialylation of β2 integrin through NEU1. A similar action mechanism was identified in human umbilical vein endothelial cells (HUVEC) for intercellular cell adhesion molecule-1 (ICAM-1). Importantly, these effects were associated with a significant increase in monocyte adhesion to endothelial cells and monocyte transendothelial migration. CONCLUSIONS These results demonstrate that membrane NEU1 sialidase interacts and modulates the sialylation levels of the β2 integrin and ICAM-1 through the ERC in monocytes and endothelial cells, respectively, and suggest that EDP and the ERC, through this newly identified common mode of action governed by NEU1, may be important regulators of circulating monocyte recruitment to inflamed vascular sites. Moreover, by its ability to interact with and to modulate the sialylation of key membrane glycoproteins through NEU1, new biological functions are anticipated for EDP and the ERC in elastin remodeling-associated disorders.
Collapse
Affiliation(s)
- Olivier Bocquet
- UMR CNRS 7369 Matrice Extracellulaire Et Dynamique Cellulaire (MEDyC), Université de Reims Champagne Ardenne (URCA), UFR Sciences Exactes Et Naturelles, Moulin de La Housse, BP1039, 51687, Reims cedex 2, France
| | - Dignê Tembely
- UMR CNRS 7369 Matrice Extracellulaire Et Dynamique Cellulaire (MEDyC), Université de Reims Champagne Ardenne (URCA), UFR Sciences Exactes Et Naturelles, Moulin de La Housse, BP1039, 51687, Reims cedex 2, France
| | - Damien Rioult
- Plateau Technique Mobile de Cytométrie Environnementale MOBICYTE, URCA/INERIS, Université de Reims Champagne Ardenne (URCA), UFR Sciences Exactes Et Naturelles, Reims, France
| | - Christine Terryn
- Plate-Forme Imagerie Cellulaire Et Tissulaire (PICT), Université de Reims Champagne Ardenne (URCA), UFR Médecine, Reims, France
| | - Béatrice Romier
- UMR CNRS 7369 Matrice Extracellulaire Et Dynamique Cellulaire (MEDyC), Université de Reims Champagne Ardenne (URCA), UFR Sciences Exactes Et Naturelles, Moulin de La Housse, BP1039, 51687, Reims cedex 2, France
| | - Amar Bennasroune
- UMR CNRS 7369 Matrice Extracellulaire Et Dynamique Cellulaire (MEDyC), Université de Reims Champagne Ardenne (URCA), UFR Sciences Exactes Et Naturelles, Moulin de La Housse, BP1039, 51687, Reims cedex 2, France
| | - Sébastien Blaise
- UMR CNRS 7369 Matrice Extracellulaire Et Dynamique Cellulaire (MEDyC), Université de Reims Champagne Ardenne (URCA), UFR Sciences Exactes Et Naturelles, Moulin de La Housse, BP1039, 51687, Reims cedex 2, France
| | - Hervé Sartelet
- UMR CNRS 7369 Matrice Extracellulaire Et Dynamique Cellulaire (MEDyC), Université de Reims Champagne Ardenne (URCA), UFR Sciences Exactes Et Naturelles, Moulin de La Housse, BP1039, 51687, Reims cedex 2, France
| | - Laurent Martiny
- UMR CNRS 7369 Matrice Extracellulaire Et Dynamique Cellulaire (MEDyC), Université de Reims Champagne Ardenne (URCA), UFR Sciences Exactes Et Naturelles, Moulin de La Housse, BP1039, 51687, Reims cedex 2, France
| | - Laurent Duca
- UMR CNRS 7369 Matrice Extracellulaire Et Dynamique Cellulaire (MEDyC), Université de Reims Champagne Ardenne (URCA), UFR Sciences Exactes Et Naturelles, Moulin de La Housse, BP1039, 51687, Reims cedex 2, France
| | - Pascal Maurice
- UMR CNRS 7369 Matrice Extracellulaire Et Dynamique Cellulaire (MEDyC), Université de Reims Champagne Ardenne (URCA), UFR Sciences Exactes Et Naturelles, Moulin de La Housse, BP1039, 51687, Reims cedex 2, France.
| |
Collapse
|
21
|
Hyun SW, Imamura A, Ishida H, Piepenbrink KH, Goldblum SE, Lillehoj EP. The sialidase NEU1 directly interacts with the juxtamembranous segment of the cytoplasmic domain of mucin-1 to inhibit downstream PI3K-Akt signaling. J Biol Chem 2021; 297:101337. [PMID: 34688655 PMCID: PMC8591358 DOI: 10.1016/j.jbc.2021.101337] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2021] [Revised: 10/14/2021] [Accepted: 10/15/2021] [Indexed: 12/04/2022] Open
Abstract
The extracellular domain (ED) of the membrane-spanning sialoglycoprotein, mucin-1 (MUC1), is an in vivo substrate for the lysosomal sialidase, neuraminidase-1 (NEU1). Engagement of the MUC1-ED by its cognate ligand, Pseudomonas aeruginosa-expressed flagellin, increases NEU1-MUC1 association and NEU1-mediated MUC1-ED desialylation to unmask cryptic binding sites for its ligand. However, the mechanism(s) through which intracellular NEU1 might physically interact with its surface-expressed MUC1-ED substrate are unclear. Using reciprocal coimmunoprecipitation and in vitro binding assays in a human airway epithelial cell system, we show here that NEU1 associates with the MUC1-cytoplasmic domain (CD) but not with the MUC1-ED. Prior pharmacologic inhibition of the NEU1 catalytic activity using the NEU1-selective sialidase inhibitor, C9-butyl amide-2-deoxy-2,3-dehydro-N-acetylneuraminic acid, did not diminish NEU1-MUC1-CD association. In addition, glutathione-S-transferase (GST) pull-down assays using the deletion mutants of the MUC1-CD mapped the NEU1-binding site to the membrane-proximal 36 aa of the MUC1-CD. In a cell-free system, we found that the purified NEU1 interacted with the immobilized GST-MUC1-CD and the purified MUC1-CD associated with the immobilized 6XHis-NEU1, indicating that the NEU1-MUC1-CD interaction was direct and independent of its chaperone protein, protective protein/cathepsin A. However, the NEU1-MUC1-CD interaction was not required for the NEU1-mediated MUC1-ED desialylation. Finally, we demonstrated that overexpression of either WT NEU1 or a catalytically dead NEU1 G68V mutant diminished the association of the established MUC1-CD binding partner, PI3K, to MUC1-CD and reduced downstream Akt kinase phosphorylation. These results indicate that NEU1 associates with the juxtamembranous region of the MUC1-CD to inhibit PI3K-Akt signaling independent of NEU1 catalytic activity.
Collapse
Affiliation(s)
- Sang W Hyun
- US Department of Veterans Affairs, Veterans Affairs Medical Center, University of Maryland School of Medicine, Baltimore, Maryland, USA; Department of Medicine, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Akihiro Imamura
- Department of Applied Bio-organic Chemistry, Gifu University, Gifu, Japan
| | - Hideharu Ishida
- Department of Applied Bio-organic Chemistry, Gifu University, Gifu, Japan
| | - Kurt H Piepenbrink
- Food Science and Technology Department, University of Nebraska, Lincoln, Nebraska, USA
| | - Simeon E Goldblum
- US Department of Veterans Affairs, Veterans Affairs Medical Center, University of Maryland School of Medicine, Baltimore, Maryland, USA; Department of Medicine, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Erik P Lillehoj
- Department of Pediatrics, University of Maryland School of Medicine, Baltimore, Maryland, USA.
| |
Collapse
|
22
|
Dewi IM, Cunha C, Jaeger M, Gresnigt MS, Gkountzinopoulou ME, Garishah FM, Duarte-Oliveira C, Campos CF, Vanderbeke L, Sharpe AR, Brüggemann RJ, Verweij PE, Lagrou K, Vande Velde G, de Mast Q, Joosten LA, Netea MG, van der Ven AJ, Wauters J, Carvalho A, van de Veerdonk FL. Neuraminidase and SIGLEC15 modulate the host defense against pulmonary aspergillosis. Cell Rep Med 2021; 2:100289. [PMID: 34095887 PMCID: PMC8149467 DOI: 10.1016/j.xcrm.2021.100289] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2019] [Revised: 09/01/2020] [Accepted: 04/23/2021] [Indexed: 11/30/2022]
Abstract
Influenza-associated pulmonary aspergillosis (IAPA) has been reported increasingly since the advent of use of neuraminidase (NA) inhibitors following the 2009 influenza pandemic. We hypothesize that blocking host NA modulates the immune response against Aspergillus fumigatus. We demonstrate that NA influences the host response against A. fumigatus in vitro and that oseltamivir increases the susceptibility of mice to pulmonary aspergillosis. Oseltamivir impairs the mouse splenocyte and human peripheral blood mononuclear cell (PBMC) killing capacity of A. fumigatus, and adding NA restores this defect in PBMCs. Furthermore, the sialic acid-binding receptor SIGLEC15 is upregulated in PBMCs stimulated with A. fumigatus. Silencing of SIGLEC15 decrease PBMC killing of A. fumigatus. We provide evidence that host NA activity and sialic acid recognition are important for anti-Aspergillus defense. NA inhibitors might predispose individuals with severe influenza to invasive aspergillosis. These data shed light on the pathogenesis of invasive fungal infections and may identify potential therapeutic targets.
Collapse
Affiliation(s)
- Intan M.W. Dewi
- Department of Internal Medicine, Radboud University Medical Center, Nijmegen, the Netherlands
- Microbiology Division, Department of Biomedical Sciences, Faculty of Medicine, Universitas Padjadjaran, Bandung, Indonesia
| | - Cristina Cunha
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal
- ICVS/3B’s – PT Government Associate Laboratory, Guimarães/Braga, Portugal
| | - Martin Jaeger
- Department of Internal Medicine, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Mark S. Gresnigt
- Department of Microbial Pathogenicity Mechanisms, Leibniz Institute for Natural Product Research and Infection Biology-Hans Knoll Institute, Jena, Germany
| | | | - Fadel M. Garishah
- Department of Internal Medicine, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Cláudio Duarte-Oliveira
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal
- ICVS/3B’s – PT Government Associate Laboratory, Guimarães/Braga, Portugal
| | - Cláudia F. Campos
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal
- ICVS/3B’s – PT Government Associate Laboratory, Guimarães/Braga, Portugal
| | - Lore Vanderbeke
- Department of Microbiology and Immunology, KU Leuven, Leuven, Belgium
| | | | - Roger J. Brüggemann
- Department of Pharmacy, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Paul E. Verweij
- Department of Medical Microbiology, Radboud University Medical Centre, Nijmegen, the Netherlands
| | - Katrien Lagrou
- Department of Microbiology and Immunology, KU Leuven, Leuven, Belgium
| | - Greetje Vande Velde
- Biomedical MRI/Molecular Small Animal Imaging Center, Department of Imaging and Pathology, KU Leuven, Belgium
| | - Quirijn de Mast
- Department of Internal Medicine, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Leo A.B. Joosten
- Department of Internal Medicine, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Mihai G. Netea
- Department of Internal Medicine, Radboud University Medical Center, Nijmegen, the Netherlands
| | | | - Joost Wauters
- Department of Microbiology and Immunology, KU Leuven, Leuven, Belgium
| | - Agostinho Carvalho
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal
- ICVS/3B’s – PT Government Associate Laboratory, Guimarães/Braga, Portugal
| | | |
Collapse
|
23
|
Lorenz L, Amann B, Hirmer S, Degroote RL, Hauck SM, Deeg CA. NEU1 is more abundant in uveitic retina with concomitant desialylation of retinal cells. Glycobiology 2021; 31:873-883. [PMID: 33677598 DOI: 10.1093/glycob/cwab014] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2020] [Revised: 02/05/2021] [Accepted: 02/05/2021] [Indexed: 12/16/2022] Open
Abstract
Desialylation of cell surface glycoproteins carried out by sialidases affects various immunological processes. However, the role of neuraminidase 1 (NEU1), one of four mammalian sialidases, in inflammation and autoimmune disease is not completely unraveled to date. In this study, we analyzed retinal expression of NEU1 in equine recurrent uveitis (ERU), a spontaneous animal model for autoimmune uveitis. Mass spectrometry revealed significantly higher abundance of NEU1 in retinal Müller glial cells (RMG) of ERU-diseased horses compared to healthy controls. Immunohistochemistry uncovered NEU1 expression along the whole Müller cell body in healthy and uveitic state and confirmed higher abundance in inflamed retina. Müller glial cells are the principal macroglial cells of the retina and play a crucial role in uveitis pathogenesis. To determine whether higher expression levels of NEU1 in uveitic RMG correlate with desialylation of retinal cells, we performed lectin binding assays with sialic acid-specific lectins. Through these experiments we could demonstrate a profound loss of both α2-3- and α2-6-linked terminal sialic acids in uveitis. Hence, we hypothesize that higher abundance of NEU1 in uveitic RMG plays an important role in the pathogenesis of uveitis by desialylation of retinal cells. As RMG become activated in the course of uveitis and actively promote inflammation, we propose that NEU1 might represent a novel activation marker for inflammatory RMG. Our data provide novel insights in the expression and implication of NEU1 in inflammation and autoimmune disease.
Collapse
Affiliation(s)
- Lea Lorenz
- Chair of Physiology, Department of Veterinary Sciences, LMU Munich, 82152 Martinsried, Germany
| | - Barbara Amann
- Chair of Physiology, Department of Veterinary Sciences, LMU Munich, 82152 Martinsried, Germany
| | - Sieglinde Hirmer
- Chair of Physiology, Department of Veterinary Sciences, LMU Munich, 82152 Martinsried, Germany
| | - Roxane L Degroote
- Chair of Physiology, Department of Veterinary Sciences, LMU Munich, 82152 Martinsried, Germany
| | - Stefanie M Hauck
- Research Unit Protein Science, Helmholtz Center Munich, German Research Center for Environmental Health (GmbH), 80939 Munich, Germany
| | - Cornelia A Deeg
- Chair of Physiology, Department of Veterinary Sciences, LMU Munich, 82152 Martinsried, Germany
| |
Collapse
|
24
|
Demina EP, Smutova V, Pan X, Fougerat A, Guo T, Zou C, Chakraberty R, Snarr BD, Shiao TC, Roy R, Orekhov AN, Miyagi T, Laffargue M, Sheppard DC, Cairo CW, Pshezhetsky AV. Neuraminidases 1 and 3 Trigger Atherosclerosis by Desialylating Low-Density Lipoproteins and Increasing Their Uptake by Macrophages. J Am Heart Assoc 2021; 10:e018756. [PMID: 33554615 PMCID: PMC7955353 DOI: 10.1161/jaha.120.018756] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Background Chronic vascular disease atherosclerosis starts with an uptake of atherogenic modified low-density lipoproteins (LDLs) by resident macrophages, resulting in formation of arterial fatty streaks and eventually atheromatous plaques. Increased plasma sialic acid levels, increased neuraminidase activity, and reduced sialic acid LDL content have been previously associated with atherosclerosis and coronary artery disease in human patients, but the mechanism underlying this association has not been explored. Methods and Results We tested the hypothesis that neuraminidases contribute to development of atherosclerosis by removing sialic acid residues from glycan chains of the LDL glycoprotein and glycolipids. Atherosclerosis progression was investigated in apolipoprotein E and LDL receptor knockout mice with genetic deficiency of neuraminidases 1, 3, and 4 or those treated with specific neuraminidase inhibitors. We show that desialylation of the LDL glycoprotein, apolipoprotein B 100, by human neuraminidases 1 and 3 increases the uptake of human LDL by human cultured macrophages and by macrophages in aortic root lesions in Apoe-/- mice via asialoglycoprotein receptor 1. Genetic inactivation or pharmacological inhibition of neuraminidases 1 and 3 significantly delays formation of fatty streaks in the aortic root without affecting the plasma cholesterol and LDL levels in Apoe-/- and Ldlr-/- mouse models of atherosclerosis. Conclusions Together, our results suggest that neuraminidases 1 and 3 trigger the initial phase of atherosclerosis and formation of aortic fatty streaks by desialylating LDL and increasing their uptake by resident macrophages.
Collapse
Affiliation(s)
- Ekaterina P Demina
- Departments of Pediatrics and Biochemistry Sainte-Justine University Hospital Research CenterUniversity of Montreal Quebec Canada
| | - Victoria Smutova
- Departments of Pediatrics and Biochemistry Sainte-Justine University Hospital Research CenterUniversity of Montreal Quebec Canada
| | - Xuefang Pan
- Departments of Pediatrics and Biochemistry Sainte-Justine University Hospital Research CenterUniversity of Montreal Quebec Canada
| | - Anne Fougerat
- Departments of Pediatrics and Biochemistry Sainte-Justine University Hospital Research CenterUniversity of Montreal Quebec Canada
| | - Tianlin Guo
- Department of Chemistry University of Alberta Edmonton Alberta Canada
| | - Chunxia Zou
- Department of Chemistry University of Alberta Edmonton Alberta Canada
| | | | - Brendan D Snarr
- Departments of Medicine, Microbiology and Immunology McGill University Montreal Quebec Canada
| | - Tze C Shiao
- Department of Chemistry Université du Québec à Montréal Montreal Quebec Canada
| | - Rene Roy
- Department of Chemistry Université du Québec à Montréal Montreal Quebec Canada
| | | | - Taeko Miyagi
- Miyagi Cancer Center Research Institute Natori Miyagi Japan
| | - Muriel Laffargue
- Institut National de la Santé et de la Recherche MédicaleUMR 1048Institute of Metabolic and Cardiovascular Diseases Toulouse France
| | - Donald C Sheppard
- Departments of Medicine, Microbiology and Immunology McGill University Montreal Quebec Canada
| | | | - Alexey V Pshezhetsky
- Departments of Pediatrics and Biochemistry Sainte-Justine University Hospital Research CenterUniversity of Montreal Quebec Canada
| |
Collapse
|
25
|
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: 0.8] [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
|
26
|
Klaus C, Liao H, Allendorf DH, Brown GC, Neumann H. Sialylation acts as a checkpoint for innate immune responses in the central nervous system. Glia 2020; 69:1619-1636. [PMID: 33340149 DOI: 10.1002/glia.23945] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Revised: 11/23/2020] [Accepted: 11/26/2020] [Indexed: 12/28/2022]
Abstract
Sialic acids are monosaccharides that normally terminate the glycan chains of cell surface glyco-proteins and -lipids in mammals, and are highly enriched in the central nervous tissue. Sialic acids are conjugated to proteins and lipids (termed "sialylation") by specific sialyltransferases, and are removed ("desialylation") by neuraminidases. Cell surface sialic acids are sensed by complement factor H (FH) to inhibit complement activation or by sialic acid-binding immunoglobulin-like lectin (SIGLEC) receptors to inhibit microglial activation, phagocytosis, and oxidative burst. In contrast, desialylation of cells enables binding of the opsonins C1, calreticulin, galectin-3, and collectins, stimulating phagocytosis of such cells. Hypersialylation is used by bacteria and cancers as camouflage to escape immune recognition, while polysialylation of neurons protects synapses and neurogenesis. Insufficient lysosomal cleavage of sialylated molecules can lead to lysosomal accumulation of lipids and aggregated proteins, which if excessive may be expelled into the extracellular space. On the other hand, desialylation of immune receptors can activate them or trigger removal of proteins. Loss of inhibitory SIGLECs or FH triggers reduced clearance of aggregates, oxidative brain damage and complement-mediated retinal damage. Thus, cell surface sialylation recognized by FH, SIGLEC, and other immune-related receptors acts as a major checkpoint inhibitor of innate immune responses in the central nervous system, while excessive cleavage of sialic acid residues and consequently removing this checkpoint inhibitor may trigger lipid accumulation, protein aggregation, inflammation, and neurodegeneration.
Collapse
Affiliation(s)
- Christine Klaus
- Institute of Reconstructive Neurobiology, Medical Faculty and University Hospital of Bonn, University of Bonn, Bonn, Germany
| | - Huan Liao
- Institute of Reconstructive Neurobiology, Medical Faculty and University Hospital of Bonn, University of Bonn, Bonn, Germany
| | | | - Guy C Brown
- Department of Biochemistry, University of Cambridge, Cambridge, UK
| | - Harald Neumann
- Institute of Reconstructive Neurobiology, Medical Faculty and University Hospital of Bonn, University of Bonn, Bonn, Germany
| |
Collapse
|
27
|
Circulating Neutrophils of Nonalcoholic Steatohepatitis Patients Show an Activated Phenotype and Suppress T Lymphocytes Activity. J Immunol Res 2020; 2020:4570219. [PMID: 32671116 PMCID: PMC7346102 DOI: 10.1155/2020/4570219] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2020] [Revised: 04/24/2020] [Accepted: 06/02/2020] [Indexed: 02/07/2023] Open
Abstract
Neutrophils or PolyMorphonuclear Neutrophils (PMNs) are key effector cells of the innate immune system and thanks to their remarkable plasticity, establish a cross talk with T cells modulating their survival and effector functions. During Nonalcoholic Steatohepatitis (NASH), the advanced form of hepatic steatosis or NAFL, PMNs infiltrate liver tissue, becoming a histological feature of NASH. Our aim was to evaluate the frequency of PMNs in NAFL and NASH patients in order to understand how they modulate the activity of circulating CD4+ and CD8+ T cells. In our cohort of patients, NASH patients displayed a higher frequency of circulating PMNs that was strongly correlated to liver enzymes, grade of steatosis, inflammation and fibrosis, the hepatocellular ballooning, and NAFLD Activity Score (NAS). Furthermore, even if ex vivo, in both groups of patients, PMNs shared the same phenotype of resting cells, after 24 hours of coculture with autologous CD4+ and CD8+ T cells, PMNs of NASH patients acquired a more active phenotype, becoming able to strongly inhibit proliferation and activation of CD4+ and CD8+ T cells. The higher ability of PMNs of NASH patients in suppressing CD4+ and CD8+ T cells, over time, might contribute in reducing the immunological defense of liver tissue against damages thus taking part in the progression of the NAFL disease toward NASH.
Collapse
|
28
|
Allendorf DH, Franssen EH, Brown GC. Lipopolysaccharide activates microglia via neuraminidase 1 desialylation of Toll‐like Receptor 4. J Neurochem 2020; 155:403-416. [DOI: 10.1111/jnc.15024] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2020] [Revised: 03/31/2020] [Accepted: 03/31/2020] [Indexed: 12/21/2022]
|
29
|
Silva Z, Ferro T, Almeida D, Soares H, Ferreira JA, Deschepper FM, Hensbergen PJ, Pirro M, van Vliet SJ, Springer S, Videira PA. MHC Class I Stability is Modulated by Cell Surface Sialylation in Human Dendritic Cells. Pharmaceutics 2020; 12:pharmaceutics12030249. [PMID: 32164343 PMCID: PMC7150992 DOI: 10.3390/pharmaceutics12030249] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2020] [Revised: 03/04/2020] [Accepted: 03/06/2020] [Indexed: 02/07/2023] Open
Abstract
Maturation of human Dendritic Cells (DCs) is characterized by increased expression of antigen presentation molecules, and overall decreased levels of sialic acid at cell surface. Here, we aimed to identify sialylated proteins at DC surface and comprehend their role and modulation. Mass spectrometry analysis of DC’s proteins, pulled down by a sialic acid binding lectin, identified molecules of the major human histocompatibility complex class I (MHC-I), known as human leucocyte antigen (HLA). After desialylation, DCs showed significantly higher reactivity with antibodies specific for properly folded MHC-I-β2-microglobulin complex and for β2-microglobulin but showed significant lower reactivity with an antibody specific for free MHC-I heavy chain. Similar results for antibody reactivities were observed for TAP2-deficient lymphoblastoid T2 cells, which express HLA-A*02:01. Using fluorescent peptide specifically fitting the groove of HLA-A*02:01, instead of antibody staining, also showed higher peptide binding on desialylated cells, confirming higher surface expression of MHC-I complex. A decay assay showed that desialylation doubled the half-life of MHC-I molecules at cell surface in both DCs and T2 cells. The biological impact of DC´s desialylation was evaluated in co-cultures with autologous T cells, showing higher number and earlier immunological synapses, and consequent significantly increased production of IFN-γ by T cells. In summary, sialic acid content modulates the expression and stability of complex MHC-I, which may account for the improved DC-T synapses.
Collapse
Affiliation(s)
- Zélia Silva
- UCIBIO, Departamento Ciências da Vida, Faculdade de Ciências e Tecnologia, Universidade Nova de Lisboa, 2829-516 Caparica, Portugal; (Z.S.); (T.F.); (D.A.); (F.M.D.)
| | - Tiago Ferro
- UCIBIO, Departamento Ciências da Vida, Faculdade de Ciências e Tecnologia, Universidade Nova de Lisboa, 2829-516 Caparica, Portugal; (Z.S.); (T.F.); (D.A.); (F.M.D.)
- CDG & Allies – PPAIN- Congenital Disorders of Glycosylation & Allies - Professionals and Patient Associations International Network, 2829-516 Caparica, Portugal
| | - Danielle Almeida
- UCIBIO, Departamento Ciências da Vida, Faculdade de Ciências e Tecnologia, Universidade Nova de Lisboa, 2829-516 Caparica, Portugal; (Z.S.); (T.F.); (D.A.); (F.M.D.)
| | - Helena Soares
- Human Immunobiology and Pathogenesis, CEDOC-Chronic Diseases Research Centre, NOVA Medical School, Faculdade de Ciências Médicas, Universidade Nova de Lisboa, 1150-082 Lisbon, Portugal;
| | - José Alexandre Ferreira
- Experimental Pathology and Therapeutics Group, Portuguese Institute of Oncology, 4200-162 Porto, Portugal;
- Porto Comprehensive Cancer Center (P.ccc), 4200-072 Porto, Portugal
| | - Fanny M. Deschepper
- UCIBIO, Departamento Ciências da Vida, Faculdade de Ciências e Tecnologia, Universidade Nova de Lisboa, 2829-516 Caparica, Portugal; (Z.S.); (T.F.); (D.A.); (F.M.D.)
| | - Paul J. Hensbergen
- Center for Proteomics and Metabolomics, Leiden University Medical Center, 2300 RC Leiden, The Netherlands; (P.J.H.); (M.P.)
| | - Martina Pirro
- Center for Proteomics and Metabolomics, Leiden University Medical Center, 2300 RC Leiden, The Netherlands; (P.J.H.); (M.P.)
| | - Sandra J. van Vliet
- Amsterdam UMC, Vrije Universiteit Amsterdam, Department of Molecular Cell Biology and Immunology, Cancer Center Amsterdam, Amsterdam Infection and Immunity Institute, De Boelelaan 1117, 1081 HzAmsterdam, The Netherlands;
| | - Sebastian Springer
- Department of Life Sciences and Chemistry, Jacobs University, 28759 Bremen, Germany;
| | - Paula A. Videira
- UCIBIO, Departamento Ciências da Vida, Faculdade de Ciências e Tecnologia, Universidade Nova de Lisboa, 2829-516 Caparica, Portugal; (Z.S.); (T.F.); (D.A.); (F.M.D.)
- CDG & Allies – PPAIN- Congenital Disorders of Glycosylation & Allies - Professionals and Patient Associations International Network, 2829-516 Caparica, Portugal
- Correspondence: ; Tel.: +351-212948530
| |
Collapse
|
30
|
Allendorf DH, Puigdellívol M, Brown GC. Activated microglia desialylate their surface, stimulating complement receptor 3-mediated phagocytosis of neurons. Glia 2019; 68:989-998. [PMID: 31774586 PMCID: PMC7079032 DOI: 10.1002/glia.23757] [Citation(s) in RCA: 58] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2019] [Revised: 11/13/2019] [Accepted: 11/15/2019] [Indexed: 12/30/2022]
Abstract
The glycoproteins and glycolipids of the cell surface have sugar chains that normally terminate in a sialic acid residue, but inflammatory activation of myeloid cells can cause sialidase enzymes to remove these residues, resulting in desialylation and altered activity of surface receptors, such as the phagocytic complement receptor 3 (CR3). We found that activation of microglia with lipopolysaccharide (LPS), fibrillar amyloid beta (Aβ), Tau or phorbol myristate acetate resulted in increased surface sialidase activity and desialylation of the microglial surface. Desialylation of microglia by adding sialidase, stimulated microglial phagocytosis of beads, but this was prevented by siRNA knockdown of CD11b or a blocking antibody to CD11b (a component of CR3). Desialylation of microglia by a sialyl-transferase inhibitor (3FAx-peracetyl-Neu5Ac) also stimulated microglial phagocytosis of beads. Desialylation of primary glial-neuronal co-cultures by adding sialidase or the sialyl-transferase inhibitor resulted in neuronal loss that was prevented by inhibiting phagocytosis with cytochalasin D or the blocking antibody to CD11b. Adding desialylated microglia to glial-neuronal cultures, in the absence of neuronal desialylation, also caused neuronal loss prevented by CD11b blocking antibody. Adding LPS or Aβ to primary glial-neuronal co-cultures caused neuronal loss, and this was prevented by inhibiting endogenous sialidase activity with N-acetyl-2,3-dehydro-2-deoxyneuraminic acid or blockage of CD11b. Thus, activated microglia release a sialidase activity that desialylates the cell surface, stimulating CR3-mediated phagocytosis of neurons, making extracellular sialidase and CR3 potential treatment targets to prevent inflammatory loss of neurons.
Collapse
Affiliation(s)
| | - Mar Puigdellívol
- Department of Biochemistry, University of Cambridge, Cambridge, UK
| | - Guy C Brown
- Department of Biochemistry, University of Cambridge, Cambridge, UK
| |
Collapse
|
31
|
Howlader MA, Li C, Zou C, Chakraberty R, Ebesoh N, Cairo CW. Neuraminidase-3 Is a Negative Regulator of LFA-1 Adhesion. Front Chem 2019; 7:791. [PMID: 31824923 PMCID: PMC6882948 DOI: 10.3389/fchem.2019.00791] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2019] [Accepted: 11/04/2019] [Indexed: 01/13/2023] Open
Abstract
Within the plasma membrane environment, glycoconjugate-receptor interactions play an important role in the regulation of cell-cell interactions. We have investigated the mechanism and activity of the human neuraminidase (NEU) isoenzyme, NEU3, on T cell adhesion receptors. The enzyme is known to prefer glycolipid substrates, and we confirmed that exogenous enzyme altered the glycolipid composition of cells. NEU3 was able to modify the sialic acid content of purified LFA-1 in vitro. Enzymatic activity of NEU3 resulted in re-organization of LFA-1 into large clusters on the membrane. This change was facilitated by an increase in the lateral mobility of LFA-1 upon NEU3 treatment. Changes to the lateral mobility of LFA-1 were specific for NEU3 activity, and we observed no significant change in diffusion when cells were treated with a bacterial NEU (NanI). Furthermore, we found that NEU3 treatment of cells increased surface expression levels of LFA-1. We observed that NEU3-treated cells had suppressed LFA-1 adhesion to an ICAM-1 coated surface using an in vitro static adhesion assay. These results establish that NEU3 can modulate glycoconjugate composition and contribute to the regulation of integrin activity. We propose that NEU3 should be investigated to determine its role on LFA-1 within the inflammatory cascade.
Collapse
Affiliation(s)
- Md Amran Howlader
- Department of Chemistry, University of Alberta, Edmonton, AB, Canada
| | - Caishun Li
- Department of Chemistry, University of Alberta, Edmonton, AB, Canada
| | - Chunxia Zou
- Department of Chemistry, University of Alberta, Edmonton, AB, Canada
| | | | - Njuacha Ebesoh
- Department of Chemistry, University of Alberta, Edmonton, AB, Canada
| | | |
Collapse
|
32
|
Murakami Y, Ishikawa K, Nakao S, Sonoda KH. Innate immune response in retinal homeostasis and inflammatory disorders. Prog Retin Eye Res 2019; 74:100778. [PMID: 31505218 DOI: 10.1016/j.preteyeres.2019.100778] [Citation(s) in RCA: 75] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2019] [Revised: 08/12/2019] [Accepted: 09/02/2019] [Indexed: 01/03/2023]
Abstract
Innate immune cells such as neutrophils, monocyte-macrophages and microglial cells are pivotal for the health and disease of the retina. For the maintenance of retinal homeostasis, these cells and immunosuppressive molecules in the eye actively regulate the induction and the expression of inflammation in order to prevent excessive activation and subsequent tissue damage. In the disease context, these regulatory mechanisms are modulated genetically and/or by environmental stimuli such as damage-associated molecular patterns (DAMPs), and a chronic innate immune response regulates or contributes to the formation of diverse retinal disorders such as uveitis, retinitis pigmentosa, retinal vascular diseases and retinal fibrosis. Here we summarize the recent knowledge regarding the innate immune response in both ocular immune regulation and inflammatory retinal diseases, and we describe the potential of the innate immune response as a biomarker and therapeutic target.
Collapse
Affiliation(s)
- Yusuke Murakami
- Department of Ophthalmology, Graduate School of Medical Science, Kyushu University, Fukuoka, 812-8582, Japan
| | - Keijiro Ishikawa
- Department of Ophthalmology, Graduate School of Medical Science, Kyushu University, Fukuoka, 812-8582, Japan
| | - Shintaro Nakao
- Department of Ophthalmology, Graduate School of Medical Science, Kyushu University, Fukuoka, 812-8582, Japan
| | - Koh-Hei Sonoda
- Department of Ophthalmology, Graduate School of Medical Science, Kyushu University, Fukuoka, 812-8582, Japan.
| |
Collapse
|
33
|
Bennasroune A, Romier-Crouzet B, Blaise S, Laffargue M, Efremov RG, Martiny L, Maurice P, Duca L. Elastic fibers and elastin receptor complex: Neuraminidase-1 takes the center stage. Matrix Biol 2019; 84:57-67. [PMID: 31226402 DOI: 10.1016/j.matbio.2019.06.007] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2019] [Revised: 06/11/2019] [Accepted: 06/17/2019] [Indexed: 01/02/2023]
Abstract
Extracellular matrix (ECM) has for a long time being considered as a simple architectural support for cells. It is now clear that ECM presents a fundamental influence on cells driving their phenotype and fate. This complex network is highly specialized and the different classes of macromolecules that comprise the ECM determine its biological functions. For instance, collagens are responsible for the tensile strength of tissues, proteoglycans and glycosaminoglycans are essential for hydration and resistance to compression, and glycoproteins such as laminins facilitate cell attachment. The largest structures of the ECM are the elastic fibers found in abundance in tissues suffering high mechanical constraints such as skin, lungs or arteries. These structures present a very complex composition whose core is composed of elastin surrounded by a microfibrils mantle. Elastogenesis is a tightly regulated process involving the sialidase activity of the Neuraminidase-1 (Neu-1) sub-unit of the Elastin Receptor Complex. Interestingly, Neu-1 subunit also serves as a sensor of elastin degradation via its ability to transmit elastin-derived peptides signaling. Finally, reports showing that neuraminidase activity is able to regulate TGF-β activation raises questions about a possible role for Neu-1 in elastic fibers remodeling. In this mini review, we develop the concept of the regulation of the whole life of elastic fibers through an original scope, the key role of Neu-1 sialidase enzymatic activity.
Collapse
Affiliation(s)
- Amar Bennasroune
- UMR CNRS 7369 MEDyC, SFR CAP-Santé, Université de Reims Champagne-Ardenne, Reims, France
| | | | - Sébastien Blaise
- UMR CNRS 7369 MEDyC, SFR CAP-Santé, Université de Reims Champagne-Ardenne, Reims, France
| | - Muriel Laffargue
- UMR INSERM 1048 I2MC, Université Paul Sabatier, Toulouse, France
| | - Roman G Efremov
- M. M. Shemyakin and Yu. A. Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, Russia; Higher School of Economics, Myasnitskaya ul. 20, 101000 Moscow, Russia
| | - Laurent Martiny
- UMR CNRS 7369 MEDyC, SFR CAP-Santé, Université de Reims Champagne-Ardenne, Reims, France
| | - Pascal Maurice
- UMR CNRS 7369 MEDyC, SFR CAP-Santé, Université de Reims Champagne-Ardenne, Reims, France
| | - Laurent Duca
- UMR CNRS 7369 MEDyC, SFR CAP-Santé, Université de Reims Champagne-Ardenne, Reims, France.
| |
Collapse
|
34
|
Kijimoto-Ochiai S, Kamimura K, Koda T. Neu-medullocytes, sialidase-positive B cells in the thymus, express autoimmune regulator (AIRE). Sci Rep 2019; 9:858. [PMID: 30696872 PMCID: PMC6351566 DOI: 10.1038/s41598-018-37225-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2018] [Accepted: 12/04/2018] [Indexed: 01/18/2023] Open
Abstract
Neu-medullocytes, which were previously identified and named by our group, are sialidase (neuraminidase)-positive B cells that express immunoglobulin and Mac-1 in the mouse thymus. Recently, B cells that migrated into the thymus were reported to express autoimmune regulator (AIRE) and to contribute to self-tolerance. We sought to determine whether Neu-medullocytes also express AIRE. We obtained positive results by triple staining Neu-medullocytes for in situ sialidase activity, anti-AIRE, and either anti-IgG or anti-IgM antibodies and observing the staining with confocal microscopy. Additional molecules including CD5, IgM, major histocompatibility complex (MHC) Class II, and neuraminidase 1 (NEU1) were found in sialidase-positive cells independently. The real-time PCR results suggest that the primary sialidase in AIRE-positive cells is neuraminidase 2 (NEU2). Furthermore, some of the AIRE-positive medullary thymic epithelial cells also clearly showed sialidase activity when a triple staining of sialidase activity, anti-AIRE, and Ulex europaeus agglutinin-1 (UEA-1) was performed. Neu-medullocytes may present Aire-dependent antigens for negative selection. We discuss the negative selection steps in consideration of sialidases and sialic acids.
Collapse
Affiliation(s)
- Shigeko Kijimoto-Ochiai
- Faculty of Advanced Life Science, Hokkaido University, N21 W11, Kitaku, Sapporo, 001-0021, Japan.
- Life Space COSMOS, Hirosaki, 036-8222, Japan.
| | - Keiko Kamimura
- Faculty of Advanced Life Science, Hokkaido University, N21 W11, Kitaku, Sapporo, 001-0021, Japan
| | - Toshiaki Koda
- Faculty of Advanced Life Science, Hokkaido University, N21 W11, Kitaku, Sapporo, 001-0021, Japan.
| |
Collapse
|
35
|
Wei M, Wang PG. Desialylation in physiological and pathological processes: New target for diagnostic and therapeutic development. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2019; 162:25-57. [PMID: 30905454 DOI: 10.1016/bs.pmbts.2018.12.001] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Desialylation is a pivotal part of sialic acid metabolism, which initiates the catabolism of glycans by removing the terminal sialic acid residues on glycans, thereby modulating the structure and functions of glycans, glycoproteins, or glycolipids. The functions of sialic acids have been well recognized, whereas the function of desialylation process is underappreciated or largely ignored. However, accumulating evidence demonstrates that desialylation plays an important role in a variety of physiological and pathological processes. This chapter summarizes the current knowledge pertaining to desialylation in a variety of physiological and pathological processes, with a focus on the underlying molecular mechanisms. The potential of targeting desialylation process for diagnostic and therapeutic development is also discussed.
Collapse
Affiliation(s)
- Mohui Wei
- Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, United States.
| | - Peng George Wang
- Department of Chemistry, Center for Diagnostics and Therapeutics, Georgia State University, Atlanta, GA, United States
| |
Collapse
|
36
|
French BM, Sendil S, Pierson RN, Azimzadeh AM. The role of sialic acids in the immune recognition of xenografts. Xenotransplantation 2017; 24. [PMID: 29057592 PMCID: PMC10167934 DOI: 10.1111/xen.12345] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2016] [Revised: 07/05/2017] [Accepted: 08/14/2017] [Indexed: 12/11/2022]
Abstract
Presentation of sialic acid (Sia) varies among different tissues and organs within each species, and between species. This diversity has biologically important consequences regarding the recognition of cells by "xeno" antibodies (Neu5Gc vs Neu5Ac). Sia also plays a central role in inflammation by influencing binding of the asialoglycoprotein receptor 1 (ASGR-1), Siglec-1 (Sialoadhesin), and cellular interactions mediated by the selectin, integrin, and galectin receptor families. This review will focus on what is known about basic Sia structure and function in association with xenotransplantation, how changes in sialylation may occur in this context (through desialylation or changes in sialyltransferases), and how this fundamental pathway modulates adhesive and cell activation pathways that appear to be particularly crucial to homeostasis and inflammation for xenografts.
Collapse
Affiliation(s)
- Beth M French
- Division of Cardiac Surgery, University of Maryland Baltimore, School of Medicine, and VAMC, Baltimore, MD, USA
| | - Selin Sendil
- Division of Cardiac Surgery, University of Maryland Baltimore, School of Medicine, and VAMC, Baltimore, MD, USA
| | - Richard N Pierson
- Division of Cardiac Surgery, University of Maryland Baltimore, School of Medicine, and VAMC, Baltimore, MD, USA
| | - Agnes M Azimzadeh
- Division of Cardiac Surgery, University of Maryland Baltimore, School of Medicine, and VAMC, Baltimore, MD, USA
| |
Collapse
|
37
|
Antibody against Microbial Neuraminidases Recognizes Human Sialidase 3 (NEU3): the Neuraminidase/Sialidase Superfamily Revisited. mBio 2017; 8:mBio.00078-17. [PMID: 28655817 PMCID: PMC5487728 DOI: 10.1128/mbio.00078-17] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Neuraminidases (NAs) are critical virulence factors for several microbial pathogens. With a highly conserved catalytic domain, a microbial NA "superfamily" has been proposed. We previously reported that murine polymorphonuclear leukocyte (PMN) sialidase activity was important in leukocyte trafficking to inflamed sites and that antibodies to Clostridium perfringens NA recognized a cell surface molecule(s), presumed to be a sialidase of eukaryotic origin on interleukin-8-stimulated human and murine PMNs. These antibodies also inhibited cell sialidase activity both in vitro and, in the latter instance, in vivo We therefore hypothesized that mammalian sialidases share structural homology and epitopes with microbial NAs. We now report that antibodies to one of the isoforms of C. perfringens NA, as well as anti-influenza virus NA serum, recognize human NEU3 but not NEU1 and that antibodies to C. perfringens NA inhibit NEU3 enzymatic activity. We conclude that the previously described microbial NA superfamily extends to human sialidases. Strategies designed to therapeutically inhibit microbial NA may need to consider potential compromising effects on human sialidases, particularly those expressed in cells of the immune system.IMPORTANCE We previously reported that sialidase activity of human neutrophils plays a critical role in the host inflammatory response. Since the catalytic domains of microbial neuraminidases are highly conserved, we hypothesized that antibodies against Clostridium perfringens neuraminidase might inhibit mammalian sialidase activity. Before the recognition of four mammalian sialidase (Neu) isoforms, we demonstrated that anti-C. perfringens neuraminidase antibodies inhibited human and murine sialidase activity in vivo and in vitro We now show that the antibodies to microbial neuraminidase (C. perfringens and influenza virus) recognize human NEU3, which is important for neural development and cell signaling. Since many microbes that infect mucosal surfaces express neuraminidase, it is possible that the use of sialidase inhibitors (e.g., zanamivir), might also compromise human sialidase activity critical to the human immune response. Alternatively, sialidase inhibitors may prove useful in the treatment of hyperinflammatory conditions.
Collapse
|
38
|
Nomura K, Vilalta A, Allendorf DH, Hornik TC, Brown GC. Activated Microglia Desialylate and Phagocytose Cells via Neuraminidase, Galectin-3, and Mer Tyrosine Kinase. THE JOURNAL OF IMMUNOLOGY 2017; 198:4792-4801. [PMID: 28500071 PMCID: PMC5458330 DOI: 10.4049/jimmunol.1502532] [Citation(s) in RCA: 68] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/03/2015] [Accepted: 04/12/2017] [Indexed: 12/31/2022]
Abstract
Activated microglia can phagocytose dying, stressed, or excess neurons and synapses via the phagocytic receptor Mer tyrosine kinase (MerTK). Galectin-3 (Gal-3) can cross-link surface glycoproteins by binding galactose residues that are normally hidden below terminal sialic acid residues. Gal-3 was recently reported to opsonize cells via activating MerTK. We found that LPS-activated BV-2 microglia rapidly released Gal-3, which was blocked by calcineurin inhibitors. Gal-3 bound to MerTK on microglia and to stressed PC12 (neuron-like) cells, and it increased microglial phagocytosis of PC12 cells or primary neurons, which was blocked by inhibition of MerTK. LPS-activated microglia exhibited a sialidase activity that desialylated PC12 cells and could be inhibited by Tamiflu, a neuraminidase (sialidase) inhibitor. Sialidase treatment of PC12 cells enabled Gal-3 to bind and opsonize the live cells for phagocytosis by microglia. LPS-induced microglial phagocytosis of PC12 was prevented by small interfering RNA knockdown of Gal-3 in microglia, lactose inhibition of Gal-3 binding, inhibition of neuraminidase with Tamiflu, or inhibition of MerTK by UNC569. LPS-induced phagocytosis of primary neurons by primary microglia was also blocked by inhibition of MerTK. We conclude that activated microglia release Gal-3 and a neuraminidase that desialylates microglial and PC12 surfaces, enabling Gal-3 binding to PC12 cells and their phagocytosis via MerTK. Thus, Gal-3 acts as an opsonin of desialylated surfaces, and inflammatory loss of neurons or synapses may potentially be blocked by inhibiting neuraminidases, Gal-3, or MerTK.
Collapse
Affiliation(s)
- Koji Nomura
- Department of Biochemistry, University of Cambridge, Cambridge CB2 1QW, United Kingdom
| | - Anna Vilalta
- Department of Biochemistry, University of Cambridge, Cambridge CB2 1QW, United Kingdom
| | - David H Allendorf
- Department of Biochemistry, University of Cambridge, Cambridge CB2 1QW, United Kingdom
| | - Tamara C Hornik
- Department of Biochemistry, University of Cambridge, Cambridge CB2 1QW, United Kingdom
| | - Guy C Brown
- Department of Biochemistry, University of Cambridge, Cambridge CB2 1QW, United Kingdom
| |
Collapse
|
39
|
Abstract
Oseltamivir is recommended for the treatment and prophylaxis of influenza in persons at higher risk for influenza complications such as individuals with diabetes, neuropsychiatric illnesses, and respiratory, cardiac, renal, hepatic or haematological diseases. However, a recent Cochrane review reported that reduction of antibody production, renal disorders, hyperglycaemia, psychiatric disorders, and QT prolongation may be related to oseltamivir use. The underlying mechanisms are reviewed. There is decisive evidence that administration of a clinically compatible dose of oseltamivir in mice challenged by a respiratory syncytial virus (RSV) that lacks a neuraminidase gene showed symptom-relieving effects and inhibition of viral clearance. These effects were accompanied by decreased level of T cell surface sialoglycosphingolipid (ganglioside) GM1 that is regulated by the endogenous neuraminidase in response to viral challenge. Clinical and non-clinical evidence supports the view that the usual dose of oseltamivir suppresses pro-inflammatory cytokines such as interferon-gamma, interleukin-6, and tumour necrosis factor-alpha almost completely with partial suppression of viral shedding in human influenza virus infection experiment. Animal toxicity tests support the clinical evidence with regard to renal and cardiac disorders (bradycardia and QT prolongation) and do not disprove the metabolic effect. Reduction of antibody production and cytokine induction and renal, metabolic, cardiac, and prolonged psychiatric disorders after oseltamivir use may be related to inhibition of the host’s endogenous neuraminidase. While the usual clinical dose of zanamivir may not have this effect, a higher dose or prolonged administration of zanamivir and other neuraminidase inhibitors may induce similar delayed reactions, including reduction of the antibody and/or cytokine production.
Collapse
Affiliation(s)
- Rokuro Hama
- a Non-Profit Organization, Japan Institute of Pharmacovigilance , Osaka , Japan
| |
Collapse
|
40
|
Targeting of Neutrophil Lewis X Blocks Transepithelial Migration and Increases Phagocytosis and Degranulation. THE AMERICAN JOURNAL OF PATHOLOGY 2015; 186:297-311. [PMID: 26687991 DOI: 10.1016/j.ajpath.2015.10.015] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/15/2015] [Revised: 08/28/2015] [Accepted: 10/02/2015] [Indexed: 12/21/2022]
Abstract
Polymorphonuclear leukocytes (PMNs) are innate immune cells whose principal function is to migrate from the blood to sites of inflammation, where they exert crucial anti-infectious and immunomodulatory effects. However, dysregulated migration of PMNs into mucosal epithelial tissues is characteristic of chronic inflammatory disorders, including inflammatory bowel disease. Carbohydrate-mediated binding interactions between PMN Lewis glycans and endothelial glycan-binding proteins are critical for initial migration of PMN out of the vasculature. However, the role of Lewis glycans during transepithelial migration (TEM) has not been well characterized. Herein, we show that antibody blockade of Lewis X (Le(x)) displayed as terminal glycan residues on the PMN surface blocks chemotaxis and TEM while enhancing PMN-adhesive interactions with intestinal epithelia. Unexpectedly, targeting of subterminal Le(x) residues within glycan chains had no effect on PMN migration or adhesive interactions. There was increased surface expression of Le(x) on PMN after TEM, and blockade of terminal Le(x) regulated post-migratory PMN functions, increasing PMN phagocytosis and the surface mobilization of azurophilic (CD63, myeloperoxidase, and neutrophil elastase) and specific (CD66b and lactoferrin) granule markers. These findings suggest that terminal Le(x) represents a potential target for regulating PMN trafficking and function in inflamed mucosa. Furthermore, given its abundant expression on migrating PMN, Le(x) may be a rational target for modulating inflammation in diseases where dysregulated PMN influx is associated with host tissue damage.
Collapse
|
41
|
Zhang PP, Meng ZT, Wang LC, Guo LM, Li K. Astragalus polysaccharide promotes the release of mature granulocytes through the L-selectin signaling pathway. Chin Med 2015; 10:17. [PMID: 26161135 PMCID: PMC4497426 DOI: 10.1186/s13020-015-0043-z] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2014] [Accepted: 06/23/2015] [Indexed: 01/03/2023] Open
Abstract
Background This study aims to investigate the leukogenic effect of astragalus polysaccharide (APS), to compare its effect of increasing the numbers of mature granulocytes with that of granulocyte colony-stimulating factor (G-CSF), and to investigate the mechanism. Methods Rats were arbitrarily grouped into four groups (control, cyclophosphamide (CTX), CTX + APS, and CTX + G-CSF groups), and each group was then arbitrarily divided into five subgroups according to the time period since CTX infusion (0, 4, 7, 10, and 14 days). The expression of leukocyte selectin (L-selectin), its ligand, and shedding-related protease on granulocytes was analyzed. Leukocyte counts were obtained. Chemotactic capacity of polymorphonuclear leukocytes (PMNLs) was assessed. Results Both APS and G-CSF restored the expression of L-selectin, P-selectin glycoprotein ligand-1 (PSGL-1), CD11b/CD18, and ADAM17 to normal levels (P > 0.05 vs. control group on each time point), with APS eliciting a greater effect than G-CSF (P = 0.005 on day 7, P < 0.001 on day 10 and 14 for L-selectin; P = 0.038 on day 7, P = 0.001 on day 10, P < 0.001 on day 14 for PSGL-1; P < 0.001 on day 7, 10 and 14 for ADAM17; P < 0.001 on day 7, 10, and 14 for CD11b/CD18). The percentages of the bands and segmented bone marrow (BM) cells in myeloid neutrophils were higher in the CTX + APS group than in the CTX group on day 7 (P = 0.030) and reached normal levels on day 10 (P = 0.547) and 14 (P = 0.431) vs. control group. The ability of APS to increase numbers of PMNLs in peripheral blood after chemotherapy was significantly superior to that of G-CSF 7 days after chemotherapy (P = 0.029 on day 10, P = 0.006 on day 14). Moreover, APS more significantly improved the chemotactic ability of PMNLs among mature BM granulocytes and peripheral blood neutrophils after chemotherapy than did G-CSF (P < 0.001 on day 7, P = 0.001 on day 10 and P = 0.005 on day 14). Conclusions APS promoted the differentiation and chemotactic ability of BM granulocytes via the L-selectin signaling pathway. Electronic supplementary material The online version of this article (doi:10.1186/s13020-015-0043-z) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Ping-Ping Zhang
- Department of Thoracic Medical Oncology, Lung Cancer Diagnosis and Treatment Center, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer; Key Laboratory of Cancer Prevention and Therapy, Tianjin, 300060 China.,Department of Radiotherapy, Hubei Cancer Hospital, Wuhan, 430079 China
| | - Zhao-Ting Meng
- Department of Thoracic Medical Oncology, Lung Cancer Diagnosis and Treatment Center, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer; Key Laboratory of Cancer Prevention and Therapy, Tianjin, 300060 China
| | - Liu-Chun Wang
- Department of Thoracic Medical Oncology, Lung Cancer Diagnosis and Treatment Center, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer; Key Laboratory of Cancer Prevention and Therapy, Tianjin, 300060 China
| | - Lei-Ming Guo
- Tianjin Institute for Biomedicinal Research, Tianjin, 300050 China
| | - Kai Li
- Department of Thoracic Medical Oncology, Lung Cancer Diagnosis and Treatment Center, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer; Key Laboratory of Cancer Prevention and Therapy, Tianjin, 300060 China
| |
Collapse
|
42
|
Kabanov DS, Grachev SV, Prokhorenko IR. Role of CD11b/CD18 in priming of human leukocytes by endotoxin glycoforms from Escherichia coli. BIOCHEMISTRY (MOSCOW) 2015; 79:812-9. [PMID: 25365491 DOI: 10.1134/s0006297914080094] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
The primary objective of this study was to determine the role of β2 integrin α-subunit (CD11b) in the mechanism of human polymorphonuclear leukocyte (PML) priming by S or Re endotoxin glycoforms from Escherichia coli for fMLP-induced respiratory burst. Similar priming activity of S and Re endotoxin glycoforms for fMLP-induced reactive oxygen species (ROS) generation from primed PML was found. Anti-CD11b antibodies (clone ICRF 44) as well as isotype-matched immunoglobulin G1 (clone MOPC-21) do not influence the fMLP-induced ROS generation from unprimed PML. Antibodies against CD11b do not change fMLP-induced ROS generation from endotoxin-primed PML as well. The involvement of different isoforms of Fcγ receptors in fMLP-induced ROS generation from activated PML is proposed.
Collapse
Affiliation(s)
- D S Kabanov
- Institute of Basic Biological Problems, Russian Academy of Sciences, Pushchino, Moscow Region, 142290, Russia.
| | | | | |
Collapse
|
43
|
Neves JDC, Rizzato VR, Fappi A, Garcia MM, Chadi G, van de Vlekkert D, d'Azzo A, Zanoteli E. Neuraminidase-1 mediates skeletal muscle regeneration. Biochim Biophys Acta Mol Basis Dis 2015; 1852:1755-64. [PMID: 26001931 PMCID: PMC5617636 DOI: 10.1016/j.bbadis.2015.05.006] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2014] [Revised: 05/06/2015] [Accepted: 05/11/2015] [Indexed: 12/31/2022]
Abstract
Neuraminidase-1 (NEU1) is the sialidase responsible for the catabolism of sialoglycoconjugates in lysosomes. Congenital NEU1 deficiency causes sialidosis, a severe lysosomal storage disease associated with a broad spectrum of clinical manifestations, which also include skeletal deformities, skeletal muscle hypotonia and weakness. Neu1(-/-) mice, a model of sialidosis, develop an atypical form of muscle degeneration caused by progressive expansion of the connective tissue that infiltrates the muscle bed, leading to fiber degeneration and atrophy. Here we investigated the role of Neu1 in the myogenic process that ensues during muscle regeneration after cardiotoxin-induced injury of limb muscles. A comparative analysis of cardiotoxin-treated muscles from Neu1(-/-) mice and Neu1(+/+) mice showed increased inflammatory and proliferative responses in the absence of Neu1 during the early stages of muscle regeneration. This was accompanied by significant and sequential upregulation of Pax7, MyoD, and myogenin mRNAs. The levels of both MyoD and myogenin proteins decreased during the late stages of regeneration, which most likely reflected an increased rate of degradation of the myogenic factors in the Neu1(-/-) muscle. We also observed a delay in muscle cell differentiation, which was characterized by prolonged expression of embryonic myosin heavy chain, as well as reduced myofiber cross-sectional area. At the end of the regenerative process, collagen type III deposition was increased compared to wild-type muscles and internal controls, indicating the initiation of fibrosis. Overall, these results point to a role of Neu1 throughout muscle regeneration.
Collapse
Affiliation(s)
| | | | - Alan Fappi
- Department of Neurology, University of São Paulo, São Paulo, SP 01246-903, Brazil
| | | | - Gerson Chadi
- Department of Neurology, University of São Paulo, São Paulo, SP 01246-903, Brazil
| | | | - Alessandra d'Azzo
- Department of Genetics, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Edmar Zanoteli
- Department of Neurology, University of São Paulo, São Paulo, SP 01246-903, Brazil.
| |
Collapse
|
44
|
Pshezhetsky AV, Ashmarina LI. Desialylation of surface receptors as a new dimension in cell signaling. BIOCHEMISTRY (MOSCOW) 2014; 78:736-45. [PMID: 24010837 DOI: 10.1134/s0006297913070067] [Citation(s) in RCA: 68] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Terminal sialic acid residues are found in abundance in glycan chains of glycoproteins and glycolipids on the surface of all live cells forming an outer layer of the cell originally known as glycocalyx. Their presence affects the molecular properties and structure of glycoconjugates, modifying their function and interactions with other molecules. Consequently, the sialylation state of glycoproteins and glycolipids has been recognized as a critical factor modulating molecular recognitions inside the cell, between the cells, between the cells and the extracellular matrix, and between the cells and certain exogenous pathogens. Until recently sialyltransferases that catalyze transfer of sialic acid residues to the glycan chains in the process of their biosynthesis were thought to be mainly responsible for the creation and maintenance of a temporal and spatial diversity of sialylated moieties. However, the growing evidence suggests that in mammalian cells, at least equally important roles belong to sialidases/neuraminidases, which are located on the cell surface and in intracellular compartments, and may either initiate the catabolism of sialoglycoconjugates or just cleave their sialic acid residues, and thereby contribute to temporal changes in their structure and functions. The current review summarizes emerging data demonstrating that mammalian neuraminidase 1, well known for its lysosomal catabolic function, is also targeted to the cell surface and assumes the previously unrecognized role as a structural and functional modulator of cellular receptors.
Collapse
Affiliation(s)
- A V Pshezhetsky
- Department of Medical Genetics, CHU Sainte-Justine Research Center, Montreal, Qc, H3T1C5, Canada.
| | | |
Collapse
|
45
|
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: 27] [Impact Index Per Article: 2.3] [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
|
46
|
Pei Y, Chen X, Aboutouk D, Fuller MT, Dadoo O, Yu P, White EJ, Igdoura SA, Trigatti BL. SR-BI in bone marrow derived cells protects mice from diet induced coronary artery atherosclerosis and myocardial infarction. PLoS One 2013; 8:e72492. [PMID: 23967310 PMCID: PMC3742605 DOI: 10.1371/journal.pone.0072492] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2011] [Accepted: 07/17/2013] [Indexed: 01/01/2023] Open
Abstract
SR-BI deficient mice that are also hypomorphic for apolipoprotein E expression develop diet induced occlusive coronary artery atherosclerosis, myocardial infarction and early death. To test the role of SR-BI in bone marrow derived cells, we used bone marrow transplantation to generate SR-BI-null; apoE-hypomorphic mice in which SR-BI expression was restored solely in bone marrow derived cells. SR-BI-null; apoE-hypomorphic mice were transplanted with SR-BI+/+apoE-hypomorphic, or control, autologous SR-BI-null; apoE-hypomorphic bone marrow. Four weeks later, mice were fed a high-fat, high-cholesterol, cholate-containing diet to induce coronary artery atherosclerosis. Mice transplanted with autologous bone marrow developed extensive aortic atherosclerosis and severe occlusive coronary artery atherosclerosis after 4 weeks of feeding. This was accompanied by myocardial fibrosis and increased heart weights. In contrast, restoration of SR-BI expression in bone marrow derived-cells reduced diet induced aortic and coronary artery atherosclerosis, myocardial fibrosis and the increase in heart weights in SR-BI-null; apoE-hypomorphic mice. Restoration of SR-BI in bone marrow derived cells did not, however, affect steady state lipoprotein cholesterol levels, but did reduce plasma levels of IL-6. Monocytes from SR-BI-null mice exhibited a greater capacity to bind to VCAM-1 and ICAM-1 than those from SR-BI+/+ mice. Furthermore, restoration of SR-BI expression in bone marrow derived cells attenuated monocyte recruitment into atherosclerotic plaques in mice fed high fat, high cholesterol cholate containing diet. These data demonstrate directly that SR-BI in bone marrow-derived cells protects against both aortic and CA atherosclerosis.
Collapse
Affiliation(s)
- Ying Pei
- Department of Biochemistry and Biomedical Sciences and Thrombosis and Atherosclerosis Research Institute, McMaster University, Hamilton, Ontario, Canada
| | - Xing Chen
- Department of Biochemistry and Biomedical Sciences and Thrombosis and Atherosclerosis Research Institute, McMaster University, Hamilton, Ontario, Canada
| | - Dina Aboutouk
- Department of Biochemistry and Biomedical Sciences and Thrombosis and Atherosclerosis Research Institute, McMaster University, Hamilton, Ontario, Canada
| | - Mark T. Fuller
- Department of Biochemistry and Biomedical Sciences and Thrombosis and Atherosclerosis Research Institute, McMaster University, Hamilton, Ontario, Canada
| | - Omid Dadoo
- Department of Biochemistry and Biomedical Sciences and Thrombosis and Atherosclerosis Research Institute, McMaster University, Hamilton, Ontario, Canada
| | - Pei Yu
- Department of Biochemistry and Biomedical Sciences and Thrombosis and Atherosclerosis Research Institute, McMaster University, Hamilton, Ontario, Canada
| | - Elizabeth J. White
- Department of Biology and Pathology and Molecular Medicine, McMaster University, Hamilton, Ontario, Canada
| | - Suleiman A. Igdoura
- Department of Biology and Pathology and Molecular Medicine, McMaster University, Hamilton, Ontario, Canada
| | - Bernardo L. Trigatti
- Department of Biochemistry and Biomedical Sciences and Thrombosis and Atherosclerosis Research Institute, McMaster University, Hamilton, Ontario, Canada
- * E-mail:
| |
Collapse
|
47
|
Zhao ZG, Niu CY, Zhang LL, Zhang J, Han R, Zhang YP, Hou YL. Exogenous normal lymph alleviates lipopolysaccharide-induced acute kidney injury in rats. Ren Fail 2013; 35:806-11. [PMID: 23713704 DOI: 10.3109/0886022x.2013.794680] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Acute kidney injury (AKI) is a common pathological process which occurs in hemorrhage, intoxication, etc. It has been shown that the lymphatic circulation plays an important regulatory role in the pathogenesis of hemorrhage shock, and that exogenous normal lymph (ENL) has a beneficial effect on multiple organ injuries. In the present study, we investigated the effect of ENL on lipopolysaccharide (LPS)-induced AKI in rats. METHODS The AKI was induced by the jugular vein injection of LPS (iv, 15 mg/kg). After 15 min of LPS injection, saline or ENL without cell components (5 mL/kg) was iv infused at the speed of 0.5 mL per minute. Then, the renal function indices in plasma and renal histomorphology, and the levels of P-selectin, intercellular adhesion molecule-1 (ICAM-1), myeloperoxidase (MPO) and Na(+)-K(+)-ATPase in renal tissue were assessed at 3 or 6 h after LPS injection. RESULTS LPS induced a severe kidney injury including increased levels of urea, creatinine in plasma, aggrandized activities of ICAM-1 and MPO in renal tissue, and decreased the Na(+)-K(+)-ATPase activity in renal cells. These deleterious effects of LPS were significantly ameliorated by ENL treatment. CONCLUSION The present results indicate that ENL protect against LPS-induced AKI, suggesting an alternative therapeutic strategy for treatment of kidney injury accompanied with severe infection or sepsis.
Collapse
Affiliation(s)
- Zi-Gang Zhao
- Institute of Microcirculation, Hebei North University, Zhangjiakou, Hebei, PR China
| | | | | | | | | | | | | |
Collapse
|
48
|
Zhang Y, Albohy A, Zou Y, Smutova V, Pshezhetsky AV, Cairo CW. Identification of selective inhibitors for human neuraminidase isoenzymes using C4,C7-modified 2-deoxy-2,3-didehydro-N-acetylneuraminic acid (DANA) analogues. J Med Chem 2013; 56:2948-58. [PMID: 23530623 DOI: 10.1021/jm301892f] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
In the past two decades, human neuraminidases (human sialidases, hNEUs) have been found to be involved in numerous pathways in biology. The development of selective and potent inhibitors of these enzymes will provide critical tools for glycobiology, help to avoid undesired side effects of antivirals, and may reveal new small-molecule therapeutic targets for human cancers. However, because of the high active site homology of the hNEU isoenzymes, little progress in the design and synthesis of selective inhibitors has been realized. Guided by our previous studies of human NEU3 inhibitors, we designed a series of C4,C7-modified analogues of 2-deoxy-2,3-didehydro-N-acetylneuraminic acid (DANA) and tested them against the full panel of hNEU isoenzymes (NEU1, NEU2, NEU3, NEU4). We identified inhibitors with up to 38-fold selectivity for NEU3 and 12-fold selectivity for NEU2 over all other isoenzymes. We also identified compounds that targeted NEU2 and NEU3 with similar potency.
Collapse
Affiliation(s)
- Yi Zhang
- Alberta Glycomics Center, Department of Chemistry, University of Alberta, Edmonton, Alberta T6G 2G2, Canada
| | | | | | | | | | | |
Collapse
|
49
|
Wang J, Shiratori I, Uehori J, Ikawa M, Arase H. Neutrophil infiltration during inflammation is regulated by PILRα via modulation of integrin activation. Nat Immunol 2012; 14:34-40. [DOI: 10.1038/ni.2456] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2012] [Accepted: 09/21/2012] [Indexed: 12/15/2022]
|
50
|
Yang A, Gyulay G, Mitchell M, White E, Trigatti BL, Igdoura SA. Hypomorphic sialidase expression decreases serum cholesterol by downregulation of VLDL production in mice. J Lipid Res 2012; 53:2573-85. [PMID: 22984145 DOI: 10.1194/jlr.m027300] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Lipoprotein metabolism is an important contributing factor in the development and progression of atherosclerosis. Plasma lipoproteins and their receptors are heavily glycosylated and sialylated, and levels of sialic acids modulate their biological functions. Sialylation is controlled by the activities of sialyltranferases and sialidases. To address the impact of sialidase (neu1) activity on lipoprotein metabolism, we have generated a mouse model with a hypomorphic neu1 allele (B6.SM) that displays reduced sialidase expression and sialidase activity. The objectives of this study are to determine the impact of sialidase on the rate of hepatic lipoprotein secretion and lipoprotein uptake. Our results indicate that hepatic levels of cholesterol and triglycerides are significantly higher in B6.SM mice compared with C57Bl/6 mice; however, VLDL-triglyceride production rate is lower. In addition, B6.SM mice show significantly lower levels of hepatic microsomal triglyceride transfer protein (MTP) and active sterol-regulatory element binding protein (SREBP)-2 but higher levels of diglyceride acyltransferase (DGAT)2; these are all indicative of increased hepatic lipid storage. Rescue of sialidase activity in hypomorphic sialidase mice using helper-dependent adenovirus resulted in increased VLDL production and an increase in MTP levels. Furthermore, hypomorphic sialidase expression results in stabilization of hepatic LDL receptor (LDLR) protein expression, which enhances LDL uptake. These findings provide novel evidence for a central role of sialidase in the cross talk between the uptake and production of lipoproteins.
Collapse
Affiliation(s)
- Abraham Yang
- Department of Biology, McMaster University, Hamilton, Ontario, Canada
| | | | | | | | | | | |
Collapse
|