Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters










Database
Language
Publication year range
1.
iScience ; 26(10): 107883, 2023 Oct 20.
Article in English | MEDLINE | ID: mdl-37752945

ABSTRACT

The colonic mucosal barrier protects against infection, inflammation, and tissue ulceration. Composed primarily of Mucin-2, proteolytic erosion of this barrier is an invariant feature of colitis; however, the molecular mechanisms are not well understood. We have applied a recurrent food poisoning model of acquired inflammatory bowel disease using Salmonella enterica Typhimurium to investigate mucosal barrier erosion. Our findings reveal an innate Toll-like receptor 4-dependent mechanism activated by previous infection that induces Neu3 neuraminidase among colonic epithelial cells concurrent with increased Cathepsin-G protease secretion by Paneth cells. These anatomically separated host responses merge with the desialylation of nascent colonic Mucin-2 by Neu3 rendering the mucosal barrier susceptible to increased proteolytic breakdown by Cathepsin-G. Depletion of Cathepsin-G or Neu3 function using pharmacological inhibitors or genetic-null alleles protected against Mucin-2 proteolysis and barrier erosion and reduced the frequency and severity of colitis, revealing approaches to preserve and potentially restore the mucosal barrier.

2.
PNAS Nexus ; 1(3): pgac113, 2022 Jul.
Article in English | MEDLINE | ID: mdl-35967980

ABSTRACT

Glycosidases are hydrolytic enzymes studied principally in the context of intracellular catabolism within the lysosome. Therefore, glycosidase activities are classically measured in experimentally acidified assay conditions reflecting their low pH optima. However, glycosidases are also present in the bloodstream where they may retain sufficient activity to participate in the regulation of glycoprotein half-lives, proteostasis, and disease pathogenesis. We have, herein, established at physiological pH 7.4 in blood plasma and sera the normal ranges of four major glycosidase activities essential for blood glycoprotein remodeling in healthy mice and humans. These activities included ß-galactosidase, ß-N-acetylglucosaminidase, α-mannosidase, and α-fucosidase. We have identified their origins to include the mammalian genes Glb1, HexB, Man2a1, and Fuca1. In experimental sepsis, excursions of glycosidase activities occurred with differences in host responses to discrete bacterial pathogens. Among similar excursions in human sepsis, the elevation of ß-galactosidase activity was a prognostic indicator of increased likelihood of patient death.

3.
ACS Infect Dis ; 8(5): 1075-1085, 2022 05 13.
Article in English | MEDLINE | ID: mdl-35486714

ABSTRACT

Sepsis is an extreme inflammatory response to infection that occurs in the bloodstream and causes damage throughout the body. Glycosylation is known to play a role in immunity and inflammation, but the role of glycans in sepsis is not well-defined. Herein, we profiled the serum glycomes of experimental mouse sepsis models to identify changes induced by 4 different clinical bacterial pathogens (Gram-positive: Streptococcus pneumoniae and Staphylococcus aureus, Gram-negative: Escherichia coli and Salmonella Typhimurium) using our lectin microarray technology. We observed global shifts in the blood sera glycome that were conserved across all four species, regardless of whether they were Gram positive or negative. Bisecting GlcNAc was decreased upon sepsis and a strong increase in core 1/3 O-glycans was observed. Lectin blot analysis revealed a high molecular weight protein induced in sepsis by all four bacteria as the major cause of the core 1/3 O-glycan shift. Analysis of this band by mass spectrometry identified interalpha-trypsin inhibitor heavy chains (ITIHs) and fibronectin, both of which are associated with human sepsis. Shifts in the glycosylation of these proteins were observed. Overall, our work points toward a common mechanism for bacterially induced sepsis, marked by conserved changes in the glycome.


Subject(s)
Glycomics , Sepsis , Animals , Bacteria/metabolism , Glycomics/methods , Lectins , Mice , Polysaccharides
4.
Cell Host Microbe ; 24(4): 500-513.e5, 2018 10 10.
Article in English | MEDLINE | ID: mdl-30308156

ABSTRACT

Sepsis is a life-threatening inflammatory syndrome accompanying a bloodstream infection. Frequently secondary to pathogenic bacterial infections, sepsis remains difficult to treat as a singular disease mechanism. We compared the pathogenesis of murine sepsis experimentally elicited by five bacterial pathogens and report similarities among host responses to Gram-negative Salmonella and E. coli. We observed that a host protective mechanism involving de-toxification of lipopolysaccharide by circulating alkaline phosphatase (AP) isozymes was incapacitated during sepsis caused by Salmonella or E. coli through activation of host Toll-like receptor 4, which triggered Neu1 and Neu3 neuraminidase induction. Elevated neuraminidase activity accelerated the molecular aging and clearance of AP isozymes, thereby intensifying disease. Mice deficient in the sialyltransferase ST3Gal6 displayed increased disease severity, while deficiency of the endocytic lectin hepatic Ashwell-Morell receptor was protective. AP augmentation or neuraminidase inhibition diminished inflammation and promoted host survival. This study illuminates distinct routes of sepsis pathogenesis, which may inform therapeutic development.


Subject(s)
Alkaline Phosphatase/metabolism , Escherichia coli Infections/microbiology , Host-Pathogen Interactions , Lipopolysaccharides/metabolism , Neuraminidase/metabolism , Salmonella Infections/microbiology , Sepsis/microbiology , Alkaline Phosphatase/genetics , Animals , Disease Models, Animal , Escherichia coli/pathogenicity , Escherichia coli Infections/blood , Escherichia coli Infections/enzymology , Escherichia coli Infections/pathology , Humans , Inflammation/blood , Inflammation/enzymology , Inflammation/microbiology , Inflammation/pathology , Mice , Mice, Knockout , Neuraminidase/genetics , Salmonella Infections/blood , Salmonella Infections/enzymology , Salmonella Infections/pathology , Salmonella typhimurium/pathogenicity , Sepsis/blood , Sepsis/enzymology , Sepsis/pathology , Toll-Like Receptor 4/drug effects
5.
Science ; 358(6370)2017 12 22.
Article in English | MEDLINE | ID: mdl-29269445

ABSTRACT

Intestinal inflammation is the central pathological feature of colitis and the inflammatory bowel diseases. These syndromes arise from unidentified environmental factors. We found that recurrent nonlethal gastric infections of Gram-negative Salmonella enterica Typhimurium (ST), a major source of human food poisoning, caused inflammation of murine intestinal tissue, predominantly the colon, which persisted after pathogen clearance and irreversibly escalated in severity with repeated infections. ST progressively disabled a host mechanism of protection by inducing endogenous neuraminidase activity, which accelerated the molecular aging and clearance of intestinal alkaline phosphatase (IAP). Disease was linked to a Toll-like receptor 4 (TLR4)-dependent mechanism of IAP desialylation with accumulation of the IAP substrate and TLR4 ligand, lipopolysaccharide-phosphate. The administration of IAP or the antiviral neuraminidase inhibitor zanamivir was therapeutic by maintaining IAP abundance and function.


Subject(s)
Alkaline Phosphatase/deficiency , Colon/microbiology , Inflammatory Bowel Diseases/microbiology , Salmonella Food Poisoning/complications , Salmonella typhimurium , Toll-Like Receptor 4/metabolism , Alkaline Phosphatase/administration & dosage , Animals , Colon/immunology , Colon/pathology , Enzyme Inhibitors/administration & dosage , Inflammatory Bowel Diseases/drug therapy , Inflammatory Bowel Diseases/enzymology , Inflammatory Bowel Diseases/pathology , Lipopolysaccharides/immunology , Mice , Mice, Inbred C57BL , Neuraminidase/antagonists & inhibitors , Recurrence , Sialyltransferases/genetics , Sialyltransferases/metabolism , Toll-Like Receptor 4/genetics , Zanamivir/administration & dosage , beta-Galactoside alpha-2,3-Sialyltransferase
6.
Proc Natl Acad Sci U S A ; 112(44): 13657-62, 2015 Nov 03.
Article in English | MEDLINE | ID: mdl-26489654

ABSTRACT

The composition and functions of the secreted proteome are controlled by the life spans of different proteins. However, unlike intracellular protein fate, intrinsic factors determining secreted protein aging and turnover have not been identified and characterized. Almost all secreted proteins are posttranslationally modified with the covalent attachment of N-glycans. We have discovered an intrinsic mechanism of secreted protein aging and turnover linked to the stepwise elimination of saccharides attached to the termini of N-glycans. Endogenous glycosidases, including neuraminidase 1 (Neu1), neuraminidase 3 (Neu3), beta-galactosidase 1 (Glb1), and hexosaminidase B (HexB), possess hydrolytic activities that temporally remodel N-glycan structures, progressively exposing different saccharides with increased protein age. Subsequently, endocytic lectins with distinct binding specificities, including the Ashwell-Morell receptor, integrin αM, and macrophage mannose receptor, are engaged in N-glycan ligand recognition and the turnover of secreted proteins. Glycosidase inhibition and lectin deficiencies increased protein life spans and abundance, and the basal rate of N-glycan remodeling varied among distinct proteins, accounting for differences in their life spans. This intrinsic multifactorial mechanism of secreted protein aging and turnover contributes to health and the outcomes of disease.


Subject(s)
Proteins/metabolism , Glycosylation , Polysaccharides/metabolism , Protein Processing, Post-Translational
7.
Proc Natl Acad Sci U S A ; 110(50): 20218-23, 2013 Dec 10.
Article in English | MEDLINE | ID: mdl-24284176

ABSTRACT

The endocytic Ashwell-Morell receptor (AMR) of hepatocytes detects pathogen remodeling of host glycoproteins by neuraminidase in the bloodstream and mitigates the lethal coagulopathy of sepsis. We have investigated the mechanism of host protection by the AMR during the onset of sepsis and in response to the desialylation of blood glycoproteins by the NanA neuraminidase of Streptococcus pneumoniae. We find that the AMR selects among potential glycoprotein ligands unmasked by microbial neuraminidase activity in pneumococcal sepsis to eliminate from blood circulation host factors that contribute to coagulation and thrombosis. This protection is attributable in large part to the rapid induction of a moderate thrombocytopenia by the AMR. We further show that neuraminidase activity in the blood can be manipulated to induce the clearance of AMR ligands including platelets, thereby preactivating a protective response in pneumococcal sepsis that moderates the severity of disseminated intravascular coagulation and enables host survival.


Subject(s)
Asialoglycoprotein Receptor/immunology , Hepatocytes/immunology , Sepsis/prevention & control , Streptococcus pneumoniae/immunology , Analysis of Variance , Animals , Asialoglycoprotein Receptor/metabolism , Bleeding Time , Blood Platelets/metabolism , Humans , Mice , Mice, Inbred C57BL , Neuraminidase/administration & dosage , Neuraminidase/metabolism , Sepsis/immunology , Sepsis/microbiology
SELECTION OF CITATIONS
SEARCH DETAIL
...