Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 36
Filter
Add more filters











Publication year range
1.
Clin Res Hepatol Gastroenterol ; : 102470, 2024 Sep 22.
Article in English | MEDLINE | ID: mdl-39317267

ABSTRACT

BACKGROUND AND AIM: Given the obesity epidemic, nonalcoholic fatty liver disease has become a public health concern; however, the progression mechanism remains obscure. Therefore, experimental nonalcoholic fatty liver disease/steatohepatitis (NASH) animal models that reflect human disease are necessary. In this study, we simulated the effects of gut microbiota imbalance on NASH and liver fibrosis. METHODS: Different bacterial sources of lipopolysaccharide, including Escherichia coli (GEC) and Salmonella abortus equi (GSE), were combined with a Gubra Amylin NASH (GAN) diet to induce NASH and liver fibrosis. RESULTS: Serum levels of alanine aminotransferase, hydroxyproline, CD68-positive cells, α-smooth muscle actin, and glial fibrillary acidic protein as well as TNF-α, COL1A1, TGF-ß, and NLRP3 expressions in the livers of the GSE group were markedly increased compared to those in the GAN group. The GAN diet with lipopolysaccharide (LPS) treatment resulted in a marked difference in microbiota α-diversity. The GSE group had higher numbers ofincreased Erysipelotrichaceae, Akkermansiaceae, and Bacteroidaceae families than the other groups. CONCLUSIONS: Based on these results, the GAN diet combined with LPS treatment successfully induced the progression of liver disease to NASH and fibrosis. With consistent histologically proven fibrosis, this obese NASH mouse model has relatively good clinical translatability and is highly applicable for preclinical drug testing for NASH.

2.
Int J Antimicrob Agents ; 64(4): 107319, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39233216

ABSTRACT

Nontypeable Haemophilus influenzae (NTHi), once considered a harmless commensal, has emerged as a significant concern due to the increased prevalence of multidrug-resistant (MDR) strains and their association with invasive infections. This study aimed to explore the epidemiology and molecular resistance mechanisms of 51 NTHi isolates collected from patients with invasive infections in northern Taiwan between 2011 and 2020. This investigation revealed substantial genetic diversity, encompassing 29 distinct sequence types and 18 clonal complexes. Notably, 68.6% of the isolates exhibited ampicillin resistance, with 28 categorised as MDR and four isolates were even resistant to up to six antibiotic classes. Among the MDR isolates, 18 pulsotypes were identified, indicating diverse genetic lineages. Elucidation of their resistance mechanisms revealed 18 ß-lactamase-producing amoxicillin-clavulanate-resistant (BLPACR) isolates, 12 ß-lactamase-producing ampicillin-resistant (BLPAR) isolates, and 5 ß-lactamase-nonproducing ampicillin-resistant (BLNAR) isolates. PBP3 analysis revealed 22 unique substitutions in BLPACR and BLNAR, potentially contributing to cephem resistance. Notably, novel transposons, Tn7736-Tn7739, which contain critical resistance genes, were discovered. Three strains harboured Tn7739, containing seven resistance genes [aph(3')-Ia, blaTEM-1, catA, sul2, strA, strB, and tet(B)], while four other strains carried Tn7736, Tn7737, and Tn7738, each containing three resistance genes [blaTEM-1, catA, and tet(B)]. The emergence of these novel transposons underscores the alarming threat posed by highly resistant NTHi strains. Our findings indicated that robust surveillance and comprehensive genomic studies are needed to address this growing public health challenge.


Subject(s)
Anti-Bacterial Agents , DNA Transposable Elements , Drug Resistance, Multiple, Bacterial , Haemophilus Infections , Haemophilus influenzae , Microbial Sensitivity Tests , beta-Lactamases , Humans , Haemophilus influenzae/genetics , Haemophilus influenzae/drug effects , Drug Resistance, Multiple, Bacterial/genetics , DNA Transposable Elements/genetics , Haemophilus Infections/microbiology , Haemophilus Infections/epidemiology , Anti-Bacterial Agents/pharmacology , Taiwan/epidemiology , Male , beta-Lactamases/genetics , Middle Aged , Female , Aged , Adult , Child , Child, Preschool , Adolescent , Young Adult , Aged, 80 and over , Infant , Genetic Variation
3.
NPJ Vaccines ; 9(1): 46, 2024 Feb 26.
Article in English | MEDLINE | ID: mdl-38409165

ABSTRACT

Group A Streptococcus (GAS) is a significant human pathogen that poses a global health concern. However, the development of a GAS vaccine has been challenging due to the multitude of diverse M-types and the risk of triggering cross-reactive immune responses. Our previous research has identified a critical role of PrsA1 and PrsA2, surface post-translational molecular chaperone proteins, in maintaining GAS proteome homeostasis and virulence traits. In this study, we aimed to further explore the potential of PrsA1 and PrsA2 as vaccine candidates for preventing GAS infection. We found that PrsA1 and PrsA2 are highly conserved among GAS isolates, demonstrating minimal amino acid variation. Antibodies specifically targeting PrsA1/A2 showed no cross-reactivity with human heart proteins and effectively enhanced neutrophil opsonophagocytic killing of various GAS serotypes. Additionally, passive transfer of PrsA1/A2-specific antibodies conferred protective immunity in infected mice. Compared to alum, immunization with CFA-adjuvanted PrsA1/A2 induced higher levels of Th1-associated IgG isotypes and complement activation and provided approximately 70% protection against invasive GAS challenge. These findings highlight the potential of PrsA1 and PrsA2 as universal vaccine candidates for the development of an effective GAS vaccine.

4.
J Exp Med ; 220(8)2023 08 07.
Article in English | MEDLINE | ID: mdl-37158982

ABSTRACT

Type I interferons are important antiviral cytokines, but prolonged interferon production is detrimental to the host. The TLR3-driven immune response is crucial for mammalian antiviral immunity, and its intracellular localization determines induction of type I interferons; however, the mechanism terminating TLR3 signaling remains obscure. Here, we show that the E3 ubiquitin ligase ZNRF1 controls TLR3 sorting into multivesicular bodies/lysosomes to terminate signaling and type I interferon production. Mechanistically, c-Src kinase activated by TLR3 engagement phosphorylates ZNRF1 at tyrosine 103, which mediates K63-linked ubiquitination of TLR3 at lysine 813 and promotes TLR3 lysosomal trafficking and degradation. ZNRF1-deficient mice and cells are resistant to infection by encephalomyocarditis virus and SARS-CoV-2 because of enhanced type I interferon production. However, Znrf1-/- mice have exacerbated lung barrier damage triggered by antiviral immunity, leading to enhanced susceptibility to respiratory bacterial superinfections. Our study highlights the c-Src-ZNRF1 axis as a negative feedback mechanism controlling TLR3 trafficking and the termination of TLR3 signaling.


Subject(s)
COVID-19 , Interferon Type I , Animals , Mice , Antiviral Agents , SARS-CoV-2 , Toll-Like Receptor 3 , Genes, src
5.
Inorg Chem ; 61(9): 3997-4008, 2022 Mar 07.
Article in English | MEDLINE | ID: mdl-35020371

ABSTRACT

In this study, aluminum complexes bearing ferrocene-based and arylthiomethylphenolate ligands were synthesized, and their catalytic activity for ε-caprolactone (CL) polymerization was investigated. The catalytic activity of the reduced form of Al complexes was higher than that of the oxidized form. The CL polymerization rate of the reduced form fcO2AlMe (75 min, conversion = 100%) was higher than that of the oxidized form fcoxO2AlMe (4320 min, conversion = 45%), and the CL polymerization rate of fc(OAlMe2)2 (40 min, conversion = 100%) was higher than that of fcox(OAlMe2)2 (60 min, conversion = 97%). Electron deficiency substituents on phenolate decreased the catalytic activity of Al complexes bearing arylthiomethylphenolate ligands. Density functional theory calculations revealed that thioether coordination stabilized the transition state (TS1) and that the oxidized form fcox(OAlMe2)2 exhibited weaker thioether coordination and higher activation energy in TS1 compared with those of the reduced form fcO2AlMe. In addition, our study determined that the thioether group is a suitable chelating group for Al catalysts in CL polymerization due to its labile nature.

6.
Virulence ; 12(1): 2633-2647, 2021 12.
Article in English | MEDLINE | ID: mdl-34592883

ABSTRACT

Streptococcus pyogenes (group A Streptococcus, GAS) is a strict human pathogen causing a broad spectrum of diseases and a variety of autoimmune sequelae. The pathogenesis of GAS infection mostly relies on the production of an extensive network of cell wall-associated and secreted virulence proteins, such as adhesins, toxins, and exoenzymes. PrsA, the only extracellular parvulin-type peptidyl-prolyl isomerase expressed ubiquitously in Gram-positive bacteria, has been suggested to assist the folding and maturation of newly exported proteins to acquire their native conformation and activity. Two PrsA proteins, PrsA1 and PrsA2, have been identified in GAS, but the respective contribution of each PrsA in GAS pathogenesis remains largely unknown. By combining comparative proteomic and phenotypic analysis approaches, we demonstrate that both PrsA isoforms are required to maintain GAS proteome homeostasis and virulence-associated traits in a unique and overlapping manner. The inactivation of both PrsA in GAS caused remarkable impairment in biofilm formation, host adherence, infection-induced cytotoxicity, and in vivo virulence in a murine soft tissue infection model. The concordance of proteomic and phenotypic data clearly features the essential role of PrsA in GAS full virulence.


Subject(s)
Streptococcal Infections , Streptococcus pyogenes , Animals , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Humans , Mice , Molecular Chaperones , Proteomics , Secretome , Streptococcus pyogenes/genetics , Virulence , Virulence Factors/genetics , Virulence Factors/metabolism
7.
Pharmaceutics ; 13(7)2021 Jul 11.
Article in English | MEDLINE | ID: mdl-34371756

ABSTRACT

Psoriasis, a complex inflammatory autoimmune skin disorder that affects 2-3% of the global population, is thought to be genetically predetermined and induced by environmental and immunological factors. In the past decades, basic and clinical studies have significantly expanded knowledge on the molecular, cellular, and immunological mechanisms underlying the pathogenesis of psoriasis. Based on these pathogenic mechanisms, the current disease model emphasizes the role of aberrant Th1 and Th17 responses. Th1 and Th17 immune responses are regulated by a complex network of different cytokines, including TNF-α, IL-17, and IL-23; signal transduction pathways downstream to the cytokine receptors; and various activated transcription factors, including NF-κB, interferon regulatory factors (IRFs), and signal transducer and activator of transcriptions (STATs). The biologics developed to specifically target the cytokines have achieved a better efficacy and safety for the systemic management of psoriasis compared with traditional treatments. Nevertheless, the current therapeutics can only alleviate the symptoms; there is still no cure for psoriasis. Therefore, the development of more effective, safe, and affordable therapeutics for psoriasis is important. In this review, we discussed the current trend of therapeutic development for psoriasis based on the recent discoveries in the immune modulation of the inflammatory response in psoriasis.

8.
J Antimicrob Chemother ; 76(8): 2049-2056, 2021 07 15.
Article in English | MEDLINE | ID: mdl-33855344

ABSTRACT

OBJECTIVES: In the past few decades, multiple-antibiotic-resistant Staphylococcus aureus has emerged and quickly spread in hospitals and communities worldwide. Additionally, the formation of antibiotic-tolerant persisters and biofilms further reduces treatment efficacy. Previously, we identified a sorafenib derivative, SC5005, with bactericidal activity against MRSA in vitro and in vivo. Here, we sought to elucidate the resistance status, mode of action and anti-persister activity of this compound. METHODS: The propensity of S. aureus to develop SC5005 resistance was evaluated by assessment of spontaneous resistance and by multi-passage selection. The mode of action of SC5005 was investigated using macromolecular synthesis, LIVE/DEAD and ATPlite assays and DiOC2(3) staining. The effect of SC5005 on the mammalian cytoplasmic membrane was measured using haemolytic and lactate dehydrogenase (LDH) assays and flow cytometry. RESULTS: SC5005 depolarized and permeabilized the bacterial cytoplasmic membrane, leading to reduced ATP production. Because of this mode of action, no resistance of S. aureus to SC5005 was observed after constant exposure to sub-lethal concentrations for 200 passages. The membrane-perturbing activity of SC5005 was specific to bacteria, as no significant haemolysis or release of LDH from human HT-29 cells was detected. Additionally, compared with other bactericidal antibiotics, SC5005 exhibited superior activity in eradicating both planktonic and biofilm-embedded S. aureus persisters. CONCLUSIONS: Because of its low propensity for resistance development and potent persister-eradicating activity, SC5005 is a promising lead compound for developing new therapies for biofilm-related infections caused by S. aureus.


Subject(s)
Methicillin-Resistant Staphylococcus aureus , Staphylococcal Infections , Animals , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/therapeutic use , Biofilms , Humans , Membrane Potentials , Microbial Sensitivity Tests , Staphylococcal Infections/drug therapy , Staphylococcus aureus
9.
Front Cell Infect Microbiol ; 11: 613195, 2021.
Article in English | MEDLINE | ID: mdl-33777832

ABSTRACT

Streptococcus pneumoniae is one of most deadly Gram-positive bacterium that causes significant mortality and morbidity worldwide. Intense inflammation and cytotoxicity is a hallmark of invasive pneumococcal disease. Pneumococcal NanA has been shown to exaggerate the production of inflammatory cytokines via unmasking of inhibitory Siglec-5 from its sialyl cis-ligands. To further investigate the mechanistic role of NanA and Siglec-5 in pneumococccal diseases, we systemically analyzed genes and signaling pathways differentially regulated in macrophages infected with wild type and NanA-deficient pneumococcus. We found that NanA-mediated desialylation impairs the Siglec-5-TLR-2 interaction and reduces the recruitment of phosphatase SHP-1 to Siglec-5. This dysregulated crosstalk between TLR-2 and inhibitory Siglec-5 exaggerated multiple inflammatory and death signaling pathways and consequently caused excessive inflammation and cytotoxicity in the infected macrophage. Collectively, our results reveal a novel virulence role of NanA in pneumococcal pathogenesis and suggest that targeting NanA activity may ameliorate the pneumococcus-mediated inflammation and cytotoxicity in severe invasive pneumococcal diseases.


Subject(s)
Pneumococcal Infections , Streptococcus pneumoniae , Caspases , Cell Death , Humans , Inflammasomes , Inflammation , Neuraminidase , Virulence
10.
mBio ; 11(4)2020 07 21.
Article in English | MEDLINE | ID: mdl-32694142

ABSTRACT

Streptococcus pyogenes (group A Streptococcus [GAS]) is an important human pathogen causing a broad spectrum of diseases and associated with significant global morbidity and mortality. Almost all GAS isolates express a surface hyaluronic acid capsule, a virulence determinant that facilitates host colonization and impedes phagocyte killing. However, recent epidemiologic surveillance has reported a sustained increase in both mucosal and invasive infections caused by nonencapsulated GAS, which questions the indispensable role of hyaluronic acid capsule in GAS pathogenesis. In this study, we found that pilus of M4 GAS not only significantly promotes biofilm formation, adherence, and cytotoxicity to human upper respiratory tract epithelial cells and keratinocytes, but also promotes survival in human whole blood and increased virulence in murine models of invasive infection. T4 antigen, the pilus backbone protein of M4 GAS, binds haptoglobin, an abundant human acute-phase protein upregulated upon infection and inflammation, on the bacterial surface. Haptoglobin sequestration reduces the susceptibility of nonencapsulated M4 GAS to antimicrobial peptides released from activated neutrophils and platelets. Our results reveal a previously unappreciated virulence-promoting role of M4 GAS pili, in part mediated by co-opting the biology of haptoglobin to mitigate host antimicrobial defenses.IMPORTANCE Group A Streptococcus (GAS) is a strict human pathogen causing more than 700 million infections globally each year. The majority of the disease-causing GAS are encapsulated, which greatly guarantees survival and dissemination in the host. Emergence of the capsule-negative GAS, such as M4 GAS, in recent epidemiologic surveillance alarms the necessity to elucidate the virulence determinants of these pathogens. Here, we found that M4 pili play an important role in promoting M4 GAS adherence and cytotoxicity to human pharyngeal epithelial cells and keratinocytes. The same molecule also significantly enhanced M4 GAS survival and replication in human whole blood and experimental murine infection. T4 antigen, which composes the backbone of M4 pili, was able to sequester the very abundant serum protein haptoglobin to further confer M4 GAS resistance to antibacterial substances released by neutrophils and platelets.


Subject(s)
Bacterial Proteins/metabolism , Fimbriae, Bacterial/immunology , Immune Evasion , Streptococcus pyogenes/immunology , Streptococcus pyogenes/pathogenicity , Animals , Bacterial Adhesion/immunology , Biofilms/growth & development , Blood Cells/microbiology , Female , Fimbriae, Bacterial/classification , HaCaT Cells , Haptoglobins/metabolism , Humans , Keratinocytes/microbiology , Mice , Mice, Inbred ICR , Neutrophils/microbiology , Phenotype , Streptococcal Infections/blood , Streptococcal Infections/immunology , Streptococcal Infections/microbiology , Streptococcus pyogenes/genetics , Virulence , Virulence Factors/metabolism
11.
Adv Exp Med Biol ; 1204: 197-214, 2020.
Article in English | MEDLINE | ID: mdl-32152948

ABSTRACT

Siglecs are sialic acid (Sia) recognizing immunoglobulin-like receptors expressed on the surface of all the major leukocyte lineages in mammals. Siglecs recognize ubiquitous Sia epitopes on various glycoconjugates in the cell glycocalyx and transduce signals to regulate immunological and inflammatory activities of these cells. The subset known as CD33-related Siglecs is principally inhibitory receptors that suppress leukocyte activation, and recent research has shown that a number of bacterial pathogens use Sia mimicry to engage these Siglecs as an immune evasion strategy. Conversely, Siglec-1 is a macrophage phagocytic receptor that engages GBS and other sialylated bacteria to promote effective phagocytosis and antigen presentation for the adaptive immune response, whereas certain viruses and parasites use Siglec-1 to gain entry to immune cells as a proximal step in the infectious process. Siglecs are positioned in crosstalk with other host innate immune sensing pathways to modulate the immune response to infection in complex ways. This chapter summarizes the current understanding of Siglecs at the host-pathogen interface, a field of study expanding in breadth and medical importance, and which provides potential targets for immune-based anti-infective strategies.


Subject(s)
Host-Pathogen Interactions/immunology , Sialic Acid Binding Immunoglobulin-like Lectins/immunology , Animals , Glycocalyx/immunology , Humans , Leukocytes/cytology , Leukocytes/immunology , Macrophages/immunology , Phagocytosis , Sialic Acid Binding Ig-like Lectin 3/immunology
12.
RSC Adv ; 10(67): 40690-40696, 2020 Nov 09.
Article in English | MEDLINE | ID: mdl-35519227

ABSTRACT

Using TiOiPr4 with a pyrazole ligand for one-pot LA polymerization improved catalytic activity compared with using TiOiPr4 only. At 60 °C, TiOiPr4 with furPz exhibited a higher catalytic activity (approximately 3-fold) than TiOiPr4. At room temperature, TiOiPr4 with BuPz exhibited a higher catalytic activity (approximately 17-fold) than TiOiPr4. High molecular mass PLA (M nGPC = 51 100, and D = 1.10) could be produced by using TiOiPr4 with furPz in melt polymerization ([TiOiPr4] : [furPz] = 1000 : 1 : 1 at 100 °C, 240 min). The crystal structure of MePz2Ti2OiPr7 revealed the cooperative activation between two Ti atoms during LA polymerization.

14.
Article in English | MEDLINE | ID: mdl-29616195

ABSTRACT

Clostridium difficile infection (CDI) is the leading cause of nosocomial infection in hospitalized patients receiving long-term antibiotic treatment. An excessive host inflammatory response is believed to be the major mechanism underlying the pathogenesis of C. difficile infection, and various proinflammatory cytokines such as IL-1ß are detected in patients with C. difficile infection. IL-1ß is known to be processed by caspase-1, a cysteine protease that is regulated by a protein complex called the inflammasome, which leads to a specialized form of cell death called pyroptosis. The function of inflammasome activation-induced pyroptosis is to clear or limit the spread of invading pathogens via infiltrated neutrophils. Here, we focused on inflammasome activation induced by intact C. difficile to re-evaluate the nature of inflammasome activation in CDI pathogenesis, which could provide information that leads to an alternative therapeutic strategy for the treatment of this condition in humans. First, we found that caspase-1-dependent IL-1ß production was induced by C. difficile pathogens in macrophages and increased in a time-dependent manner. Moreover, intracellular toxigenic C. difficile was essential for ATP-P2X7 pathway of inflammasome activation and subsequent caspase-1-dependent pyroptotic cell death, leading to the loss of membrane integrity and release of intracellular contents such as LDH. Notably, we also observed that bacterial components such as surface layer proteins (SLPs) were released from pyroptotic cells. In addition, pro-IL-1ß production was completely MyD88 and partially TLR2 dependent. Finally, to investigate the role of the caspase-1-dependent inflammasome in host defense, we found that colonic inflammasome activation was also induced by CDI and that caspase-1 inhibition by Ac-YVAD-CMK led to increased disease progression and C. difficile load. Taken together, the present results suggest that MyD88 and TLR2 are critical component in pro-IL-1ß production and intracellular C. difficile following the ATP-P2X7 pathway of inflammasome activation and pyroptosis, which play important roles in host defense through the utilization of inflammation-mediated bacterial clearance mechanisms during C. difficile infection.


Subject(s)
Adenosine Triphosphate/immunology , Clostridioides difficile/immunology , Clostridium Infections/immunology , Inflammasomes/immunology , Receptors, Purinergic P2X7/immunology , Animals , Caspase 1/genetics , Caspase 1/immunology , Clostridioides difficile/genetics , Clostridium Infections/genetics , Clostridium Infections/microbiology , Humans , Inflammasomes/genetics , Interleukin-1beta/genetics , Interleukin-1beta/immunology , Macrophages/immunology , Macrophages/microbiology , Mice , Mice, Inbred C57BL , Receptors, Purinergic P2X7/genetics , Signal Transduction , Toll-Like Receptor 2/genetics , Toll-Like Receptor 2/immunology
15.
Nat Commun ; 8: 15502, 2017 06 08.
Article in English | MEDLINE | ID: mdl-28593998

ABSTRACT

Caveolin-1 (CAV1), the major constituent of caveolae, plays a pivotal role in various cellular biological functions, including cancer and inflammation. The ubiquitin/proteasomal pathway is known to contribute to the regulation of CAV1 expression, but the ubiquitin ligase responsible for CAV1 protein stability remains unidentified. Here we reveal that E3 ubiquitin ligase ZNRF1 modulates CAV1 protein stability to regulate Toll-like receptor (TLR) 4-triggered immune responses. We demonstrate that ZNRF1 physically interacts with CAV1 in response to lipopolysaccharide and mediates ubiquitination and degradation of CAV1. The ZNRF1-CAV1 axis regulates Akt-GSK3ß activity upon TLR4 activation, resulting in enhanced production of pro-inflammatory cytokines and inhibition of anti-inflammatory cytokine IL-10. Mice with deletion of ZNRF1 in their hematopoietic cells display increased resistance to endotoxic and polymicrobial septic shock due to attenuated inflammation. Our study defines ZNRF1 as a regulator of TLR4-induced inflammatory responses and reveals another mechanism for the regulation of TLR4 signalling through CAV1.


Subject(s)
Caveolin 1/metabolism , Inflammation/metabolism , Inflammation/pathology , Proteolysis , Ubiquitin-Protein Ligases/metabolism , Ubiquitination , Amino Acid Sequence , Animals , Caveolin 1/chemistry , Cecum/pathology , Cyclic AMP Response Element-Binding Protein/metabolism , Gene Deletion , Glycogen Synthase Kinase 3 beta/metabolism , Inflammation Mediators/metabolism , Ligation , Lipopolysaccharides , Lysine/metabolism , Macrophages/metabolism , Mice , Mice, Inbred C57BL , Protein Binding , Protein Stability , Proto-Oncogene Proteins c-akt/metabolism , Punctures , RAW 264.7 Cells , Shock, Septic/immunology , Shock, Septic/metabolism , Shock, Septic/pathology , Signal Transduction , Toll-Like Receptor 4/metabolism , Ubiquitin-Protein Ligases/deficiency
16.
Infect Immun ; 83(9): 3648-56, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26150541

ABSTRACT

Recently, we showed that endothelial heparan sulfate facilitates entry of a bacterial pathogen into the central nervous system. Here, we show that normal bactericidal activity of neutrophils is influenced by the sulfation pattern of heparan sulfate. Inactivation of heparan sulfate uronyl 2-O-sulfotransferase (Hs2st) in neutrophils substantially reduced their bactericidal activity, and Hs2st deficiency rendered mice more susceptible to systemic infection with the pathogenic bacterium group B Streptococcus. Specifically, altered sulfation of heparan sulfate in mutant neutrophils affected formation of neutrophil extracellular traps while not influencing phagocytosis, production of reactive oxygen species, or secretion of granular proteases. Heparan sulfate proteoglycan(s) is present in neutrophil extracellular traps, modulates histone affinity, and modulates their microbial activity. Hs2st-deficient brain endothelial cells show enhanced binding to group B Streptococcus and are more susceptible to apoptosis, likely contributing to the observed increase in dissemination of group B Streptococcus into the brain of Hs2st-deficient mice following intravenous challenge. Taken together, our data provide strong evidence that heparan sulfate from both neutrophils and the endothelium plays important roles in modulating innate immunity.


Subject(s)
Endothelial Cells/immunology , Heparan Sulfate Proteoglycans/immunology , Immunity, Innate/immunology , Neutrophils/immunology , Animals , Blotting, Western , Cells, Cultured , Disease Models, Animal , Extracellular Traps/immunology , Heparan Sulfate Proteoglycans/metabolism , Mice , Microscopy, Electron, Scanning , Streptococcal Infections/immunology , Streptococcus agalactiae/immunology , Sulfotransferases/metabolism
17.
Glycobiology ; 24(9): 818-25, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24996821

ABSTRACT

Siglecs are mammalian sialic acid (Sia) recognizing immunoglobulin-like receptors expressed across the major leukocyte lineages, and function to recognize ubiquitous Sia epitopes on cell surface glycoconjugates and regulate immunological and inflammatory activities of these cells. A large subset referred to as CD33-related Siglecs are inhibitory receptors that limit leukocyte activation, and recent research has shown that the pathogen group B Streptococcus (GBS) binds to these Siglecs in Sia- and protein-dependent fashion to downregulate leukocyte bactericidal capacity. Conversely, sialoadhesin is a macrophage phagocytic receptor that engages GBS and other sialylated pathogens to promote effective phagocytosis and antigen presentation for the adaptive immune response. A variety of other important Siglec interactions with bacterial, viral and protozoan pathogens are beginning to be recognized. Siglec genes and binding specificities are rapidly evolving among primates, with key extant polymorphisms in human populations that may influence susceptibility to infection-associated disorders including chronic obstructive pulmonary disease and premature birth. This review summarizes current understanding of interactions between pathogens and Siglecs, a field of investigation that is likely to continue expanding in scope and medical importance.


Subject(s)
HIV/immunology , Sialic Acid Binding Immunoglobulin-like Lectins/immunology , Sialic Acids/metabolism , Streptococcus/immunology , Animals , Evolution, Molecular , Humans , Receptors, Immunologic/immunology , Receptors, Immunologic/metabolism , Sialic Acid Binding Immunoglobulin-like Lectins/metabolism
18.
J Mol Med (Berl) ; 92(9): 951-9, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24788876

ABSTRACT

UNLABELLED: Several bacterial pathogens decorate their surfaces with sialic acid (Sia) residues within cell wall components or capsular exopolysaccharides. Sialic acid expression can promote bacterial virulence by blocking complement activation or by engagement of inhibitory sialic acid-binding immunoglobulin-like lectins (Siglecs) on host leukocytes. Expressed at high levels on splenic and lymph node macrophages, sialoadhesin (Sn) is a unique Siglec with an elongated structure that lacks intracellular signaling motifs. Sialoadhesin allows macrophage to engage certain sialylated pathogens and stimulate inflammatory responses, but the in vivo significance of sialoadhesin in infection has not been shown. We demonstrate that macrophages phagocytose the sialylated pathogen group B Streptococcus (GBS) and increase bactericidal activity via sialoadhesin-sialic-acid-mediated recognition. Sialoadhesin expression on marginal zone metallophilic macrophages in the spleen trapped circulating GBS and restricted the spread of the GBS to distant organs, reducing mortality. Specific IgM antibody responses to GBS challenge were also impaired in sialoadhesin-deficient mice. Thus, sialoadhesin represents a key bridge to orchestrate innate and adaptive immune defenses against invasive sialylated bacterial pathogens. KEY MESSAGE: Sialoadhesin is critical for macrophages to phagocytose and clear GBS. Increased GBS organ dissemination in the sialoadhesin-deficient mice. Reduced anti-GBS IgM production in the sialoadhesin-deficient mice.


Subject(s)
Host-Pathogen Interactions , Macrophages/microbiology , Phagocytosis , Sialic Acid Binding Ig-like Lectin 1/immunology , Streptococcal Infections/immunology , Streptococcus agalactiae/physiology , Animals , Cells, Cultured , Gene Deletion , Immunoglobulin M/immunology , Macrophages/immunology , Mice , Sialic Acid Binding Ig-like Lectin 1/genetics , Streptococcus agalactiae/immunology
19.
PLoS Pathog ; 10(1): e1003846, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24391502

ABSTRACT

Group B Streptococcus (GBS) is a common agent of bacterial sepsis and meningitis in newborns. The GBS surface capsule contains sialic acids (Sia) that engage Sia-binding immunoglobulin-like lectins (Siglecs) on leukocytes. Here we use mice lacking Siglec-E, an inhibitory Siglec of myelomonocytic cells, to study the significance of GBS Siglec engagement during in vivo infection. We found GBS bound to Siglec-E in a Sia-specific fashion to blunt NF-κB and MAPK activation. As a consequence, Siglec-E-deficient macrophages had enhanced pro-inflammatory cytokine secretion, phagocytosis and bactericidal activity against the pathogen. Following pulmonary or low-dose intravenous GBS challenge, Siglec-E KO mice produced more pro-inflammatory cytokines and exhibited reduced GBS invasion of the central nervous system. In contrast, upon high dose lethal challenges, cytokine storm in Siglec-E KO mice was associated with accelerated mortality. We conclude that GBS Sia mimicry influences host innate immune and inflammatory responses in vivo through engagement of an inhibitory Siglec, with the ultimate outcome of the host response varying depending upon the site, stage and magnitude of infection.


Subject(s)
Antigens, CD/immunology , Antigens, Differentiation, B-Lymphocyte/immunology , Molecular Mimicry/immunology , N-Acetylneuraminic Acid/immunology , Pneumonia, Bacterial/immunology , Streptococcal Infections/immunology , Streptococcus agalactiae/immunology , Animals , Antigens, CD/genetics , Antigens, Differentiation, B-Lymphocyte/genetics , Cytokines/genetics , Cytokines/immunology , Humans , Inflammation/genetics , Inflammation/immunology , Inflammation/pathology , Macrophages/immunology , Macrophages/pathology , Mice , Mice, Knockout , N-Acetylneuraminic Acid/genetics , Pneumonia, Bacterial/genetics , Pneumonia, Bacterial/pathology , Streptococcal Infections/genetics , Streptococcal Infections/pathology
20.
FASEB J ; 28(3): 1280-93, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24308974

ABSTRACT

Siglecs are sialic acid-binding Ig-like lectins that recognize sialoglycans via amino-terminal V-set domains. CD33-related Siglecs (CD33rSiglecs) on innate immune cells recognize endogenous sialoglycans as "self-associated molecular patterns" (SAMPs), dampening immune responses via cytosolic immunoreceptor tyrosine-based inhibition motifs that recruit tyrosine phosphatases. However, sialic acid-expressing pathogens subvert this mechanism through molecular mimicry. Meanwhile, endogenous host SAMPs must continually evolve to evade other pathogens that exploit sialic acids as invasion targets. We hypothesized that these opposing selection forces have accelerated CD33rSiglec evolution. We address this by comparative analysis of major CD33rSiglec (Siglec-3, Siglec-5, and Siglec-9) orthologs in humans, chimpanzees, and baboons. Recombinant soluble molecules displaying ligand-binding domains show marked quantitative and qualitative interspecies differences in interactions with strains of the sialylated pathogen, group B Streptococcus, and with sialoglycans presented as gangliosides or in the form of sialoglycan microarrays, including variations such as N-glycolyl and O-acetyl groups. Primate Siglecs also show quantitative and qualitative intra- and interspecies variations in expression patterns on leukocytes, both in circulation and in tissues. Taken together our data explain why the CD33rSiglec-encoding gene cluster is undergoing rapid evolution via multiple mechanisms, driven by the need to maintain self-recognition by innate immune cells, while escaping 2 distinct mechanisms of pathogen subversion.


Subject(s)
Primates/immunology , Sialic Acid Binding Ig-like Lectin 3/immunology , Sialic Acid Binding Immunoglobulin-like Lectins/immunology , Animals , Antibody Specificity , Base Sequence , DNA Primers , Immunohistochemistry
SELECTION OF CITATIONS
SEARCH DETAIL