Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 79
Filter
Add more filters

Publication year range
1.
Annu Rev Biochem ; 87: 621-643, 2018 06 20.
Article in English | MEDLINE | ID: mdl-29925260

ABSTRACT

In response to microbial infection, the human host deploys metal-sequestering host-defense proteins, which reduce nutrient availability and thereby inhibit microbial growth and virulence. Calprotectin (CP) is an abundant antimicrobial protein released from neutrophils and epithelial cells at sites of infection. CP sequesters divalent first-row transition metal ions to limit the availability of essential metal nutrients in the extracellular space. While functional and clinical studies of CP have been pursued for decades, advances in our understanding of its biological coordination chemistry, which is central to its role in the host-microbe interaction, have been made in more recent years. In this review, we focus on the coordination chemistry of CP and highlight studies of its metal-binding properties and contributions to the metal-withholding innate immune response. Taken together, these recent studies inform our current model of how CP participates in metal homeostasis and immunity, and they provide a foundation for further investigations of a remarkable metal-chelating protein at the host-microbe interface and beyond.


Subject(s)
Host Microbial Interactions/immunology , Host Microbial Interactions/physiology , Leukocyte L1 Antigen Complex/immunology , Leukocyte L1 Antigen Complex/metabolism , Transition Elements/metabolism , Amino Acid Sequence , Animals , Antimicrobial Cationic Peptides/genetics , Antimicrobial Cationic Peptides/immunology , Antimicrobial Cationic Peptides/metabolism , Humans , Immunity, Innate , Iron/immunology , Iron/metabolism , Leukocyte L1 Antigen Complex/genetics , Manganese/immunology , Manganese/metabolism , Models, Biological , Models, Molecular , Nickel/immunology , Nickel/metabolism , Protein Conformation , Sequence Homology, Amino Acid , Zinc/immunology , Zinc/metabolism
2.
PLoS Genet ; 18(9): e1010189, 2022 09.
Article in English | MEDLINE | ID: mdl-36155972

ABSTRACT

BACKGROUND: Genome wide association studies (GWAS) have identified and validated more than 200 genomic loci associated with the inflammatory bowel disease (IBD), although for most the causal gene remains unknown. Given the importance of myeloid cells in IBD pathogenesis, the current study aimed to uncover the role of genes within IBD genetic loci that are endogenously expressed in this cell lineage. METHODS: The open reading frames (ORF) of 42 genes from IBD-associated loci were expressed via lentiviral transfer in the THP-1 model of human monocytes and the impact of each of these on the cell's transcriptome was analyzed using a RNA sequencing-based approach. We used a combination of genetic and pharmacologic approaches to validate our findings in the THP-1 line with further validation in human induced pluripotent stem cell (hiPSC)-derived-monocytes. RESULTS: This functional genomics screen provided evidence that genes in four IBD GWAS loci (PTGIR, ZBTB40, SLC39A11 and NFKB1) are involved in controlling S100A8 and S100A9 gene expression, which encode the two subunits of calprotectin (CP). We demonstrated that increasing PTGIR expression and/or stimulating PTGIR signaling resulted in increased CP expression in THP-1. This was further validated in hiPSC-derived monocytes. Conversely, knocking-down PTGIR endogenous expression and/or inhibiting PTGIR signaling led to decreased CP expression. These analyses were extended to the known IBD gene PTGER4, whereby its specific agonist also led to increased CP expression. Furthermore, we demonstrated that the PTGIR and PTGER4 mediated control of CP expression was dependent on signaling via adenylate cyclase and STAT3. Finally, we demonstrated that LPS-mediated increases in CP expression could be potentiated by agonists of PTGIR and PTGER4, and diminished by their antagonists. CONCLUSION: Our results support a causal role for the PTGIR, PTGER4, ZBTB40, SLC39A11 and NFKB1 genes in IBD, with all five genes regulating the expression of CP in myeloid cells, as well as potential roles for the prostacyclin/prostaglandin biogenesis and signaling pathways in IBD susceptibility and pathogenesis.


Subject(s)
Induced Pluripotent Stem Cells , Inflammatory Bowel Diseases , Adenylyl Cyclases/genetics , Genome-Wide Association Study/methods , Humans , Inflammatory Bowel Diseases/genetics , Leukocyte L1 Antigen Complex/genetics , Lipopolysaccharides , Prostaglandins , Prostaglandins I
3.
J Immunol ; 208(4): 979-990, 2022 02 15.
Article in English | MEDLINE | ID: mdl-35046105

ABSTRACT

Calprotectin is released by activated neutrophils along with myeloperoxidase (MPO) and proteases. It plays numerous roles in inflammation and infection, and is used as an inflammatory biomarker. However, calprotectin is readily oxidized by MPO-derived hypohalous acids to form covalent dimers of its S100A8 and S100A9 subunits. The dimers are susceptible to degradation by proteases. We show that detection of human calprotectin by ELISA declines markedly because of its oxidation by hypochlorous acid and subsequent degradation. Also, proteolysis liberates specific peptides from oxidized calprotectin that is present at inflammatory sites. We identified six calprotectin-derived peptides by mass spectrometry and detected them in the bronchoalveolar lavage fluid of children with cystic fibrosis (CF). We assessed the peptides as biomarkers of neutrophilic inflammation and infection. The content of the calprotectin peptide ILVI was related to calprotectin (r = 0.72, p = 0.01, n = 10). Four of the peptides were correlated with the concentration of MPO (r > 0.7, p ≤ 0.01, n = 21), while three were higher (p < 0.05) in neutrophil elastase-positive (n = 14) than -negative samples (n = 7). Also, five of the peptides were higher (p < 0.05) in the bronchoalveolar lavage fluid from children with CF with infections (n = 21) than from non-CF children without infections (n = 6). The specific peptides liberated from calprotectin will signal uncontrolled activity of proteases and MPO during inflammation. They may prove useful in tracking inflammation in respiratory diseases dominated by neutrophils, including coronavirus disease 2019.


Subject(s)
Bronchoalveolar Lavage Fluid/immunology , Cystic Fibrosis/immunology , Inflammation/immunology , Leukocyte L1 Antigen Complex/metabolism , Neutrophils/immunology , Peptides/metabolism , Respiratory System/metabolism , Child , Child, Preschool , Cystic Fibrosis/diagnosis , Female , Humans , Inflammation/diagnosis , Leukocyte L1 Antigen Complex/genetics , Leukocyte L1 Antigen Complex/immunology , Male , Neutrophil Activation , Oxidation-Reduction , Peptides/genetics , Peptides/immunology , Proteolysis
4.
Lipids Health Dis ; 23(1): 216, 2024 Jul 13.
Article in English | MEDLINE | ID: mdl-39003477

ABSTRACT

BACKGROUND: The regulation of the circadian clock genes, which coordinate the activity of the immune system, is disturbed in inflammatory bowel disease (IBD). Emerging evidence suggests that butyrate, a short-chain fatty acid produced by the gut microbiota is involved in the regulation of inflammatory responses as well as circadian-clock genes. This study was conducted to investigate the effects of sodium-butyrate supplementation on the expression of circadian-clock genes, inflammation, sleep and life quality in active ulcerative colitis (UC) patients. METHODS: In the current randomized placebo-controlled trial, 36 active UC patients were randomly divided to receive sodium-butyrate (600 mg/kg) or placebo for 12-weeks. In this study the expression of circadian clock genes (CRY1, CRY2, PER1, PER2, BMAl1 and CLOCK) were assessed by real time polymerase chain reaction (qPCR) in whole blood. Gene expression changes were presented as fold changes in expression (2^-ΔΔCT) relative to the baseline. The faecal calprotectin and serum level of high-sensitivity C-reactive protein (hs-CRP) were assessed by enzyme-linked immunosorbent assay method (ELIZA). Moreover, the sleep quality and IBD quality of life (QoL) were assessed by Pittsburgh sleep quality index (PSQI) and inflammatory bowel disease questionnaire-9 (IBDQ-9) respectively before and after the intervention. RESULTS: The results showed that sodium-butyrate supplementation in comparison with placebo significantly decreased the level of calprotectin (-133.82 ± 155.62 vs. 51.58 ± 95.57, P-value < 0.001) and hs-CRP (-0.36 (-1.57, -0.05) vs. 0.48 (-0.09-4.77), P-value < 0.001) and upregulated the fold change expression of CRY1 (2.22 ± 1.59 vs. 0.63 ± 0.49, P-value < 0.001), CRY2 (2.15 ± 1.26 vs. 0.93 ± 0.80, P-value = 0.001), PER1 (1.86 ± 1.77 vs. 0.65 ± 0.48, P-value = 0.005), BMAL1 (1.85 ± 0.97 vs. 0.86 ± 0.63, P-value = 0.003). Also, sodium-butyrate caused an improvement in the sleep quality (PSQI score: -2.94 ± 3.50 vs. 1.16 ± 3.61, P-value < 0.001) and QoL (IBDQ-9: 17.00 ± 11.36 vs. -3.50 ± 6.87, P-value < 0.001). CONCLUSION: Butyrate may be an effective adjunct treatment for active UC patients by reducing biomarkers of inflammation, upregulation of circadian-clock genes and improving sleep quality and QoL.


Subject(s)
Colitis, Ulcerative , Dietary Supplements , Sleep Quality , Humans , Colitis, Ulcerative/drug therapy , Colitis, Ulcerative/genetics , Colitis, Ulcerative/metabolism , Male , Female , Adult , Double-Blind Method , Middle Aged , Inflammation/genetics , Inflammation/drug therapy , C-Reactive Protein/metabolism , C-Reactive Protein/genetics , Quality of Life , Circadian Clocks/genetics , Circadian Clocks/drug effects , Leukocyte L1 Antigen Complex/genetics , Leukocyte L1 Antigen Complex/metabolism , Gene Expression Regulation/drug effects , Butyrates , Butyric Acid
5.
Biometals ; 36(4): 817-828, 2023 08.
Article in English | MEDLINE | ID: mdl-36826733

ABSTRACT

Human calprotectin (CP, S100A8/S100A9 oligomer) is an abundant neutrophil protein that contributes to innate immunity by sequestering nutrient metal ions in the extracellular space. This process starves invading microbial pathogens of essential metal nutrients, which can inhibit growth and colonization. Over the past decade, fundamental and clinical studies have revealed that the S100A8 and S100A9 subunits of CP exhibit a variety of post-translational modifications (PTMs). This review summarizes PTMs on the CP subunits that have been detected and highlights two recent studies that evaluated the structural and functional consequences of methionine and cysteine oxidation on CP. Collectively, these investigations indicate that the molecular speciation of extracellular CP is complex and composed of multiple proteoforms. Moreover, PTMs may impact biological function and the lifetime of the protein. It is therefore important that post-translationally modified CP species receive consideration and integration into the current working model for how CP functions in nutritional immunity.


Subject(s)
Leukocyte L1 Antigen Complex , Metals , Humans , Leukocyte L1 Antigen Complex/genetics , Leukocyte L1 Antigen Complex/metabolism , Metals/metabolism , Protein Processing, Post-Translational , Iron/metabolism , Zinc/metabolism
6.
J Am Chem Soc ; 143(43): 18073-18090, 2021 11 03.
Article in English | MEDLINE | ID: mdl-34699194

ABSTRACT

Human calprotectin (CP, S100A8/S100A9 oligomer, MRP8/MRP14 oligomer) is an abundant innate immune protein that contributes to the host metal-withholding response. Its ability to sequester transition metal nutrients from microbial pathogens depends on a complex interplay of Ca(II) binding and self-association, which converts the αß heterodimeric apo protein into a Ca(II)-bound (αß)2 heterotetramer that displays enhanced transition metal affinities, antimicrobial activity, and protease stability. A paucity of structural data on the αß heterodimer has hampered molecular understanding of how Ca(II) binding enables CP to exert its metal-sequestering innate immune function. We report solution NMR data that reveal how Ca(II) binding affects the structure and dynamics of the CP αß heterodimer. These studies provide a structural model in which the apo αß heterodimer undergoes conformational exchange and switches between two states, a tetramerization-incompetent or "inactive" state and a tetramerization-competent or "active" state. Ca(II) binding to the EF-hands of the αß heterodimer causes the active state to predominate, resulting in self-association and formation of the (αß)2 heterotetramer. Moreover, Ca(II) binding causes local and allosteric ordering of the His3Asp and His6 metal-binding sites. Ca(II) binding to the noncanonical EF-hand of S100A9 positions (A9)D30 and organizes the His3Asp site. Remarkably, Ca(II) binding causes allosteric effects in the C-terminal region of helix αIV of S100A9, which stabilize the α-helicity at positions H91 and H95 and thereby organize the functionally versatile His6 site. Collectively, this study illuminates the molecular basis for how CP responds to high extracellular Ca(II) concentrations, which enables its metal-sequestering host-defense function.


Subject(s)
Calcium/metabolism , Leukocyte L1 Antigen Complex/metabolism , Protein Multimerization/drug effects , Transition Elements/metabolism , Calgranulin A/genetics , Calgranulin A/metabolism , Calgranulin B/genetics , Calgranulin B/metabolism , Histidine/chemistry , Humans , Leukocyte L1 Antigen Complex/genetics , Metals, Heavy/metabolism , Mutation , Nuclear Magnetic Resonance, Biomolecular , Protein Binding , Protein Conformation, alpha-Helical/drug effects , Protein Multimerization/genetics
7.
Infect Immun ; 88(8)2020 07 21.
Article in English | MEDLINE | ID: mdl-32393509

ABSTRACT

Colonization by pathogenic bacteria depends on their ability to overcome host nutritional defenses and acquire nutrients. The human pathogen group A streptococcus (GAS) encounters the host defense factor calprotectin (CP) during infection. CP inhibits GAS growth in vitro by imposing zinc (Zn) limitation. However, GAS counterstrategies to combat CP-mediated Zn limitation and the in vivo relevance of CP-GAS interactions to bacterial pathogenesis remain unknown. Here, we report that GAS upregulates the AdcR regulon in response to CP-mediated Zn limitation. The AdcR regulon includes genes encoding Zn import (adcABC), Zn sparing (rpsN.2), and Zn scavenging systems (adcAII, phtD, and phtY). Each gene in the AdcR regulon contributes to GAS Zn acquisition and CP resistance. The ΔadcC and ΔrpsN.2 mutant strains were the most susceptible to CP, whereas the ΔadcA, ΔadcAII, and ΔphtD mutant strains displayed less CP sensitivity during growth in vitro However, the ΔphtY mutant strain did not display an increased CP sensitivity. The varied sensitivity of the mutant strains to CP-mediated Zn limitation suggests distinct roles for individual AdcR regulon genes in GAS Zn acquisition. GAS upregulates the AdcR regulon during necrotizing fasciitis infection in WT mice but not in S100a9-/- mice lacking CP. This suggests that CP induces Zn deficiency in the host. Finally, consistent with the in vitro results, several of the AdcR regulon genes are critical for GAS virulence in WT mice, whereas they are dispensable for virulence in S100a9-/- mice, indicating the direct competition for Zn between CP and proteins encoded by the GAS AdcR regulon during infection.


Subject(s)
Bacterial Proteins/genetics , Host-Pathogen Interactions/immunology , Leukocyte L1 Antigen Complex/immunology , Regulon , Streptococcal Infections/immunology , Streptococcus pyogenes/pathogenicity , Zinc/metabolism , Animals , Bacterial Proteins/immunology , Binding Sites , Binding, Competitive , Calgranulin B/genetics , Calgranulin B/immunology , Gene Expression Regulation , Host-Pathogen Interactions/genetics , Humans , Ion Transport , Leukocyte L1 Antigen Complex/genetics , Mice , Mice, Knockout , Protein Binding , Streptococcal Infections/metabolism , Streptococcal Infections/microbiology , Streptococcal Infections/mortality , Streptococcus pyogenes/immunology , Streptococcus pyogenes/metabolism , Survival Analysis , Virulence , Zinc/immunology
8.
PLoS Pathog ; 14(5): e1007013, 2018 05.
Article in English | MEDLINE | ID: mdl-29727465

ABSTRACT

Nutritional immunity describes the host-driven manipulation of essential micronutrients, including iron, zinc and manganese. To withstand nutritional immunity and proliferate within their hosts, pathogenic microbes must express efficient micronutrient uptake and homeostatic systems. Here we have elucidated the pathway of cellular zinc assimilation in the major human fungal pathogen Candida albicans. Bioinformatics analysis identified nine putative zinc transporters: four cytoplasmic-import Zip proteins (Zrt1, Zrt2, Zrt3 and orf19.5428) and five cytoplasmic-export ZnT proteins (orf19.1536/Zrc1, orf19.3874, orf19.3769, orf19.3132 and orf19.52). Only Zrt1 and Zrt2 are predicted to localise to the plasma membrane and here we demonstrate that Zrt2 is essential for C. albicans zinc uptake and growth at acidic pH. In contrast, ZRT1 expression was found to be highly pH-dependent and could support growth of the ZRT2-null strain at pH 7 and above. This regulatory paradigm is analogous to the distantly related pathogenic mould, Aspergillus fumigatus, suggesting that pH-adaptation of zinc transport may be conserved in fungi and we propose that environmental pH has shaped the evolution of zinc import systems in fungi. Deletion of C. albicans ZRT2 reduced kidney fungal burden in wild type, but not in mice lacking the zinc-chelating antimicrobial protein calprotectin. Inhibition of zrt2Δ growth by neutrophil extracellular traps was calprotectin-dependent. This suggests that, within the kidney, C. albicans growth is determined by pathogen-Zrt2 and host-calprotectin. As well as serving as an essential micronutrient, zinc can also be highly toxic and we show that C. albicans deals with this potential threat by rapidly compartmentalising zinc within vesicular stores called zincosomes. In order to understand mechanistically how this process occurs, we created deletion mutants of all five ZnT-type transporters in C. albicans. Here we show that, unlike in Saccharomyces cerevisiae, C. albicans Zrc1 mediates zinc tolerance via zincosomal zinc compartmentalisation. This novel transporter was also essential for virulence and liver colonisation in vivo. In summary, we show that zinc homeostasis in a major human fungal pathogen is a multi-stage process initiated by Zrt1/Zrt2-cellular import, followed by Zrc1-dependent intracellular compartmentalisation.


Subject(s)
Candida albicans/metabolism , Candida albicans/pathogenicity , Zinc/metabolism , Adaptation, Physiological , Animals , Antimicrobial Cationic Peptides/genetics , Antimicrobial Cationic Peptides/metabolism , Calgranulin B/genetics , Calgranulin B/metabolism , Candida albicans/genetics , Candidiasis, Invasive/metabolism , Candidiasis, Invasive/microbiology , Carrier Proteins/genetics , Carrier Proteins/metabolism , Cation Transport Proteins/genetics , Cation Transport Proteins/metabolism , Cell Compartmentation , Female , Fungal Proteins/genetics , Fungal Proteins/metabolism , Gene Deletion , Genes, Fungal , Host-Pathogen Interactions/genetics , Host-Pathogen Interactions/physiology , Humans , Hydrogen-Ion Concentration , Leukocyte L1 Antigen Complex/genetics , Leukocyte L1 Antigen Complex/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Virulence/genetics , Virulence/physiology , Zinc/toxicity
9.
Clin Exp Immunol ; 191(2): 240-251, 2018 02.
Article in English | MEDLINE | ID: mdl-28984903

ABSTRACT

A child, 2 years with the 'hypercalprotectinaemia with hyperzincaemia' clinical syndrome, presented with atypical symptoms and signs, notably persistent fever of approximately 38°C, thrombocythaemia of > 700 × 109 /l and a predominance of persistent intestinal symptoms. In an effort to find a cure by identifying the dysregulated pathways we analysed whole-genome mRNA expression by the Affymetrix HG U133 Plus 2·0 array in blood on three occasions 3-5 months apart. Major up-regulation was demonstrated for the Janus kinase/signal transducer and activators of transcription (JAK/STAT) pathway including, in particular, CD177, S100A8, S100A9 and S100A12, accounting for the thrombocytosis; a large number of interleukins, their receptors and activators, accounting for the febrile apathic state; and the high mobility group box 1 (HMBG1) gene, possibly accounting for part of the intestinal symptoms. These results show that gene expression array technology may assist the clinician in the diagnostic work-up of individual patients with suspected syndromal states of unknown origin, and the expression data can guide the selection of optimal treatment directed at the identified target pathways.


Subject(s)
Blood Proteins/genetics , Leukocyte L1 Antigen Complex/metabolism , Metal Metabolism, Inborn Errors/diagnosis , Child, Preschool , Clinical Decision-Making , Female , Fever , Gastrointestinal Diseases , Genome-Wide Association Study , Humans , Leukocyte L1 Antigen Complex/genetics , Metal Metabolism, Inborn Errors/genetics , Microarray Analysis , Pathology, Molecular , Signal Transduction , Thrombocytosis , Whole Genome Sequencing
10.
J Immunol ; 196(1): 336-44, 2016 Jan 01.
Article in English | MEDLINE | ID: mdl-26582948

ABSTRACT

Calprotectin, a heterodimer of S100A8 and S100A9, is an abundant neutrophil protein that possesses antimicrobial activity primarily because of its ability to chelate zinc and manganese. In the current study, we showed that neutrophils from calprotectin-deficient S100A9(-/-) mice have an impaired ability to inhibit Aspergillus fumigatus hyphal growth in vitro and in infected corneas in a murine model of fungal keratitis; however, the ability to inhibit hyphal growth was restored in S100A9(-/-) mice by injecting recombinant calprotectin. Furthermore, using recombinant calprotectin with mutations in either the Zn and Mn binding sites or the Mn binding site alone, we show that both zinc and manganese binding are necessary for calprotectin's antihyphal activity. In contrast to hyphae, we found no role for neutrophil calprotectin in uptake or killing of intracellular A. fumigatus conidia either in vitro or in a murine model of pulmonary aspergillosis. We also found that an A. fumigatus ∆zafA mutant, which demonstrates deficient zinc transport, exhibits impaired growth in infected corneas and following incubation with neutrophils or calprotectin in vitro as compared with wild-type. Collectively, these studies demonstrate a novel stage-specific susceptibility of A. fumigatus to zinc and manganese chelation by neutrophil-derived calprotectin.


Subject(s)
Aspergillus fumigatus/growth & development , Keratitis/microbiology , Leukocyte L1 Antigen Complex/metabolism , Manganese/metabolism , Neutrophils/immunology , Pulmonary Aspergillosis/immunology , Zinc/metabolism , Adolescent , Adult , Aged , Animals , Aspergillus fumigatus/immunology , Biological Transport/genetics , Calgranulin A/genetics , Calgranulin A/metabolism , Calgranulin B/genetics , Calgranulin B/metabolism , Chelating Agents/metabolism , Cornea/immunology , Cornea/microbiology , Disease Models, Animal , Humans , Hyphae/growth & development , Keratitis/immunology , Leukocyte L1 Antigen Complex/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Middle Aged , Neutrophils/metabolism , Phagocytosis/immunology , Pulmonary Aspergillosis/microbiology , Spores, Fungal/growth & development , Spores, Fungal/immunology , Young Adult
11.
Nat Chem Biol ; 11(10): 765-71, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26302479

ABSTRACT

Human calprotectin (CP) is a metal-chelating antimicrobial protein of the innate immune response. The current working model states that CP sequesters manganese and zinc from pathogens. We report the discovery that CP chelates iron and deprives bacteria of this essential nutrient. Elemental analysis of CP-treated growth medium establishes that CP reduces the concentrations of manganese, iron and zinc. Microbial growth studies reveal that iron depletion by CP contributes to the growth inhibition of bacterial pathogens. Biochemical investigations demonstrate that CP coordinates Fe(II) at an unusual hexahistidine motif, and the Mössbauer spectrum of (57)Fe(II)-bound CP is consistent with coordination of high-spin Fe(II) at this site (δ = 1.20 mm/s, ΔEQ = 1.78 mm/s). In the presence of Ca(II), CP turns on its iron-sequestering function and exhibits subpicomolar affinity for Fe(II). Our findings expand the biological coordination chemistry of iron and support a previously unappreciated role for CP in mammalian iron homeostasis.


Subject(s)
Escherichia coli/metabolism , Host-Pathogen Interactions , Iron Chelating Agents/pharmacology , Iron/metabolism , Leukocyte L1 Antigen Complex/pharmacology , Staphylococcus aureus/metabolism , Calcium/chemistry , Calcium/metabolism , Electron Spin Resonance Spectroscopy , Escherichia coli/drug effects , Escherichia coli/growth & development , Host-Pathogen Interactions/physiology , Humans , Iron Chelating Agents/metabolism , Lactobacillus plantarum/drug effects , Lactobacillus plantarum/growth & development , Lactobacillus plantarum/metabolism , Leukocyte L1 Antigen Complex/genetics , Leukocyte L1 Antigen Complex/physiology , Microbial Sensitivity Tests , Spectroscopy, Mossbauer , Staphylococcus aureus/drug effects , Staphylococcus aureus/growth & development
12.
Scand J Clin Lab Invest ; 77(3): 210-215, 2017 May.
Article in English | MEDLINE | ID: mdl-28276729

ABSTRACT

Calprotectin is an inflammatory marker, which has been found elevated in patients suffering from cardiac conditions, e.g. myocardial infarction, unstable angina and chronic heart failure. Inflammation has further been linked to atrial fibrillation (AF). However, the association between calprotectin and AF is unknown. We aimed to compare calprotectin levels in patients suffering from AF with healthy adults. In addition, AF patients with and without heart failure were compared. Calprotectin was measured in patients undergoing elective direct current cardioversion for AF. Calprotectin was determined before, 4 hours and 3 months after cardioversion. Healthy blood donors were used to verify the reference interval for calprotectin. In total, 104 prospectively enrolled patients were included. The median serum calprotectin level for AF patients was 1.6 µg/mL before cardioversion. Calprotectin levels increased significantly 4 h (1.9 µg/mL) and 3 months (2.2 µg/mL) after cardioversion. Blood donors' median serum calprotectin (1.3 µg/mL) was significantly lower than AF patients. AF patients with heart failure had significantly higher calprotectin at baseline compared with AF patients without a history of heart failure (2.0 µg/mL vs. 1.5 µg/mL). The difference was not significant at 4 h (2.0 µg/mL vs. 1.7 µg/mL) or 3 months (2.5 µg/mL vs. 2.2 µg/mL). In conclusion, the calprotectin levels in patients with AF were significantly higher than healthy blood donors and were further increased after cardioversion. AF patients with heart failure had significantly higher levels of calprotectin than AF patients without heart failure.


Subject(s)
Atrial Fibrillation/blood , Electric Countershock , Heart Failure/blood , Leukocyte L1 Antigen Complex/genetics , Adolescent , Adult , Aged , Atrial Fibrillation/diagnosis , Atrial Fibrillation/physiopathology , Atrial Fibrillation/therapy , C-Reactive Protein/genetics , C-Reactive Protein/metabolism , Case-Control Studies , Female , Gene Expression , Heart Failure/diagnosis , Heart Failure/physiopathology , Heart Failure/therapy , Humans , Leukocyte L1 Antigen Complex/blood , Male , Middle Aged
13.
Allergol Int ; 66(2): 281-289, 2017 Apr.
Article in English | MEDLINE | ID: mdl-27475624

ABSTRACT

BACKGROUND: Calprotectin is a heterodimer complex of the S100A8 and S100A9 proteins, and has various functions as an innate mediator at the sites of inflammation. The aim of this study was to elucidate the roles of calprotectin in the eosinophilic chronic rhinosinusitis (ECRS). METHODS: Allergen-induced production of calprotectin was evaluated in cultured normal human bronchial epithelial (NHBE) cells by ELISA and RT-PCR. We then examined the roles of calprotectin on Alternaria alternata (Alternaria)-induced production of thymic stromal lymphopoietin (TSLP) and IL-25 in NHBE cells. The extracellular concentration and allergen-induced secretion of calprotectin in cultured primary nasal epithelial (PNE) cells were examined and compared between patients with ECRS and non-eosinophilic chronic rhinosinusitis (NECRS). RESULTS: Alternaria, house dust mites, protease from Staphylococcus aureus, papain, trypsin, polyinosinic:polycytidylic acid and lipopolysaccharide stimulated calprotectin production in the cultured NHBE cells. The combination of calprotectin and ATP stimulated the production of TSLP and IL-25 in NHBE cells, and calprotectin stimulated Alternaria-induced production of TSLP and IL-25, which was suppressed by blocking P2 purinergic receptors and by treatment with siRNA for S100A8, S100A9 or calprotectin receptors (Toll-like receptor 4 or receptor for advanced glycation end products). Allergen-induced calprotectin production was significantly stimulated in PNE cells from patients with ECRS. CONCLUSIONS: These results indicate that calprotectin enhances the allergen-induced Th2-type inflammatory responses in airway epithelial cells via the secretion of TSLP and IL-25, and that calprotectin secreted by the epithelial cells may be involved in the pathogenesis of ECRS.


Subject(s)
Cytokines/biosynthesis , Interleukin-17/biosynthesis , Leukocyte L1 Antigen Complex/metabolism , Respiratory Mucosa/metabolism , Adenosine Triphosphate/metabolism , Adult , Aged , Allergens/immunology , Animals , Biomarkers , Cells, Cultured , Eosinophilia/pathology , Epithelial Cells/metabolism , Female , Gene Expression , Humans , Leukocyte L1 Antigen Complex/genetics , Male , Middle Aged , Proteolysis , RNA, Messenger/genetics , RNA, Messenger/metabolism , Respiratory Mucosa/immunology , Rhinitis/immunology , Rhinitis/metabolism , Rhinitis/pathology , Sinusitis/immunology , Sinusitis/metabolism , Sinusitis/pathology , Th2 Cells/immunology , Th2 Cells/metabolism , Toll-Like Receptors/agonists , Toll-Like Receptors/metabolism , Young Adult , Thymic Stromal Lymphopoietin
14.
Proc Natl Acad Sci U S A ; 110(10): 3841-6, 2013 Mar 05.
Article in English | MEDLINE | ID: mdl-23431180

ABSTRACT

The S100A8/S100A9 heterodimer calprotectin (CP) functions in the host response to pathogens through a mechanism termed "nutritional immunity." CP binds Mn(2+) and Zn(2+) with high affinity and starves bacteria of these essential nutrients. Combining biophysical, structural, and microbiological analysis, we identified the molecular basis of Mn(2+) sequestration. The asymmetry of the CP heterodimer creates a single Mn(2+)-binding site from six histidine residues, which distinguishes CP from all other Mn(2+)-binding proteins. Analysis of CP mutants with altered metal-binding properties revealed that, despite both Mn(2+) and Zn(2+) being essential metals, maximal growth inhibition of multiple bacterial pathogens requires Mn(2+) sequestration. These data establish the importance of Mn(2+) sequestration in defense against infection, explain the broad-spectrum antimicrobial activity of CP relative to other S100 proteins, and clarify the impact of metal depletion on the innate immune response to infection.


Subject(s)
Immunity, Innate , Leukocyte L1 Antigen Complex/chemistry , Leukocyte L1 Antigen Complex/immunology , Manganese/metabolism , Amino Acid Substitution , Binding Sites , Calgranulin A/chemistry , Calgranulin A/genetics , Calgranulin A/immunology , Calgranulin B/chemistry , Calgranulin B/genetics , Calgranulin B/immunology , Crystallography, X-Ray , Histidine/chemistry , Host-Pathogen Interactions/immunology , Humans , Leukocyte L1 Antigen Complex/genetics , Leukocyte L1 Antigen Complex/pharmacology , Models, Molecular , Mutagenesis, Site-Directed , Protein Multimerization , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/immunology , Recombinant Proteins/pharmacology , Staphylococcus aureus/drug effects , Staphylococcus aureus/growth & development , Staphylococcus aureus/immunology , Zinc/metabolism
15.
J Allergy Clin Immunol ; 136(5): 1337-45, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26025129

ABSTRACT

BACKGROUND: Hyperzincemia and hypercalprotectinemia (Hz/Hc) is a distinct autoinflammatory entity involving extremely high serum concentrations of the proinflammatory alarmin myeloid-related protein (MRP) 8/14 (S100A8/S100A9 and calprotectin). OBJECTIVE: We sought to characterize the genetic cause and clinical spectrum of Hz/Hc. METHODS: Proline-serine-threonine phosphatase-interacting protein 1 (PSTPIP1) gene sequencing was performed in 14 patients with Hz/Hc, and their clinical phenotype was compared with that of 11 patients with pyogenic arthritis, pyoderma gangrenosum, and acne (PAPA) syndrome. PSTPIP1-pyrin interactions were analyzed by means of immunoprecipitation and Western blotting. A structural model of the PSTPIP1 dimer was generated. Cytokine profiles were analyzed by using the multiplex immunoassay, and MRP8/14 serum concentrations were analyzed by using an ELISA. RESULTS: Thirteen patients were heterozygous for a missense mutation in the PSTPIP1 gene, resulting in a p.E250K mutation, and 1 carried a mutation resulting in p.E257K. Both mutations substantially alter the electrostatic potential of the PSTPIP1 dimer model in a region critical for protein-protein interaction. Patients with Hz/Hc have extremely high MRP8/14 concentrations (2045 ± 1300 µg/mL) compared with those with PAPA syndrome (116 ± 74 µg/mL) and have a distinct clinical phenotype. A specific cytokine profile is associated with Hz/Hc. Hz/Hc mutations altered protein binding of PSTPIP1, increasing interaction with pyrin through phosphorylation of PSTPIP1. CONCLUSION: Mutations resulting in charge reversal in the y-domain of PSTPIP1 (E→K) and increased interaction with pyrin cause a distinct autoinflammatory disorder defined by clinical and biochemical features not found in patients with PAPA syndrome, indicating a unique genotype-phenotype correlation for mutations in the PSTPIP1 gene. This is the first inborn autoinflammatory syndrome in which inflammation is driven by uncontrolled release of members of the alarmin family.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Cytoskeletal Proteins/metabolism , Leukocyte L1 Antigen Complex/metabolism , Metal Metabolism, Inborn Errors/immunology , Adaptor Proteins, Signal Transducing/genetics , Adolescent , Alarmins/genetics , Alarmins/metabolism , Calgranulin A/genetics , Calgranulin A/metabolism , Child , Cytokines/metabolism , Cytoskeletal Proteins/genetics , Female , Genotype , Humans , Leukocyte L1 Antigen Complex/genetics , Male , Metal Metabolism, Inborn Errors/genetics , Mutation, Missense/genetics , Phenotype , Phosphorylation , Protein Binding/genetics , Protein Interaction Maps/genetics , Protein Multimerization , Pyrin , Young Adult
16.
J Am Chem Soc ; 137(8): 3004-16, 2015 Mar 04.
Article in English | MEDLINE | ID: mdl-25597447

ABSTRACT

The antimicrobial protein calprotectin (CP), a hetero-oligomer of the S100 family members S100A8 and S100A9, is the only identified mammalian Mn(II)-sequestering protein. Human CP uses Ca(II) ions to tune its Mn(II) affinity at a biologically unprecedented hexahistidine site that forms at the S100A8/S100A9 interface, and the molecular basis for this phenomenon requires elucidation. Herein, we investigate the remarkable Mn(II) coordination chemistry of human CP using X-ray crystallography as well as continuous-wave (CW) and pulse electron paramagnetic resonance (EPR) spectroscopies. An X-ray crystallographic structure of Mn(II)-CP containing one Mn(II), two Ca(II), and two Na(I) ions per CP heterodimer is reported. The CW EPR spectrum of Ca(II)- and Mn(II)-bound CP prepared with a 10:0.9:1 Ca(II):Mn(II):CP ratio is characterized by an unusually low zero-field splitting of 485 MHz (E/D = 0.30) for the S = 5/2 Mn(II) ion, consistent with the high symmetry of the His6 binding site observed crystallographically. Results from electron spin-echo envelope modulation and electron-nuclear double resonance experiments reveal that the six Mn(II)-coordinating histidine residues of Ca(II)- and Mn(II)-bound CP are spectroscopically equivalent. The observed (15)N (I = 1/2) hyperfine couplings (A) arise from two distinct classes of nitrogen atoms: the coordinating ε-nitrogen of the imidazole ring of each histidine ligand (A = [3.45, 3.71, 5.91] MHz) and the distal δ-nitrogen (A = [0.11, 0.18, 0.42] MHz). In the absence of Ca(II), the binding affinity of CP for Mn(II) drops by two to three orders of magnitude and coincides with Mn(II) binding at the His6 site as well as other sites. This study demonstrates the role of Ca(II) in enabling high-affinity and specific binding of Mn(II) to the His6 site of human calprotectin.


Subject(s)
Calcium/metabolism , Leukocyte L1 Antigen Complex/chemistry , Leukocyte L1 Antigen Complex/metabolism , Manganese/metabolism , Crystallography, X-Ray , Electron Spin Resonance Spectroscopy , Histidine/chemistry , Humans , Imidazoles/chemistry , Leukocyte L1 Antigen Complex/genetics , Models, Molecular , Mutation , Oligopeptides/chemistry , Protein Binding , Protein Structure, Secondary , Protons
17.
Arterioscler Thromb Vasc Biol ; 34(7): 1399-411, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24855059

ABSTRACT

OBJECTIVE: S100A12 and fibroblast growth factor 23 are biomarkers of cardiovascular morbidity and mortality in patients with chronic kidney disease (CKD). We tested the hypothesis that human S100/calgranulin would accelerate cardiovascular disease in mice subjected to CKD. APPROACH AND RESULTS: A bacterial artificial chromosome of the human S100/calgranulin gene cluster containing the genes and regulatory elements for S100A8, S100A9, and S100A12 was expressed in C57BL/6J mouse (hBAC-S100) to generate a novel humanized mouse model. CKD was induced by ureteral ligation, and hBAC-S100 mice and wild-type mice were studied after 10 weeks of chronic uremia. hBAC-S100 mice with CKD showed increased fibroblast growth factor 23 in the hearts, left ventricular hypertrophy, diastolic dysfunction, focal cartilaginous metaplasia, and calcification of the mitral and aortic valve annulus together with aortic valve sclerosis. This phenotype was not observed in wild-type mice with CKD or in hBAC-S100 mice lacking the receptor for advanced glycation end products with CKD, suggesting that the inflammatory milieu mediated by S100/receptor for advanced glycation end products promotes pathological cardiac hypertrophy in CKD. In vitro, inflammatory stimuli including interleukin-6, tumor necrosis factor-α, lipopolysaccarides, or serum from hBAC-S100 mice upregulated fibroblast growth factor 23 mRNA and protein in primary murine neonatal and adult cardiac fibroblasts. CONCLUSIONS: Myeloid-derived human S100/calgranulin is associated with the development of cardiac hypertrophy and ectopic cardiac calcification in a receptor for advanced glycation end products-dependent manner in a mouse model of CKD. We speculate that fibroblast growth factor 23 produced by cardiac fibroblasts in response to cytokines may act in a paracrine manner to accelerate left ventricular hypertrophy and diastolic dysfunction in hBAC-S100 mice with CKD.


Subject(s)
Aortic Valve/metabolism , Heart Valve Diseases/etiology , Hypertrophy, Left Ventricular/etiology , Inflammation/complications , Leukocyte L1 Antigen Complex/metabolism , Receptors, Immunologic/metabolism , Renal Insufficiency, Chronic/complications , Animals , Aortic Valve/pathology , Calcinosis/etiology , Calcinosis/metabolism , Calgranulin A/genetics , Calgranulin A/metabolism , Calgranulin B/genetics , Calgranulin B/metabolism , Cells, Cultured , Diastole , Disease Models, Animal , Fibroblast Growth Factor-23 , Fibroblast Growth Factors/metabolism , Fibroblasts/metabolism , Genotype , Heart Valve Diseases/genetics , Heart Valve Diseases/metabolism , Heart Valve Diseases/pathology , Humans , Hypertrophy, Left Ventricular/genetics , Hypertrophy, Left Ventricular/metabolism , Hypertrophy, Left Ventricular/physiopathology , Inflammation/genetics , Inflammation/metabolism , Inflammation Mediators/metabolism , Leukocyte L1 Antigen Complex/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Myocardium/metabolism , Phenotype , Receptor for Advanced Glycation End Products , Receptors, Immunologic/deficiency , Receptors, Immunologic/genetics , Renal Insufficiency, Chronic/genetics , Renal Insufficiency, Chronic/metabolism , S100 Proteins/genetics , S100 Proteins/metabolism , S100A12 Protein , Sclerosis , Signal Transduction , Time Factors , Ventricular Dysfunction, Left/etiology , Ventricular Dysfunction, Left/metabolism , Ventricular Dysfunction, Left/physiopathology , Ventricular Function, Left , Ventricular Remodeling
18.
Circ J ; 78(2): 271-80, 2014.
Article in English | MEDLINE | ID: mdl-24389598

ABSTRACT

S100A8, S100A9 and S100A12 are considered proinflammatory mediators of atherosclerosis. Known as calgranulins, they are major components of neutrophils and are upregulated in macrophages and foam cells. They influence leukocyte recruitment, and may propagate inflammation by binding TLR4 and/or receptor for advanced glycation endproducts (RAGE). However, the receptors for calgranulins remain an enigma; we have no evidence for TLR4 or RAGE activation by S100A8 or S100A12. Moreover, gene regulation studies suggest antiinflammatory functions for S100A8 and emerging reports indicate pleiotropic roles. Unlike S100A9, S100A8 effectively scavenges oxidants generated by the myeloperoxidase system in vivo, forming novel thiol modifications. S100A8 is also readily S-nitrosylated, stabilizing nitric oxide and transporting it to hemoglobin. S100A8-SNO reduces leukocyte transmigration in the vasculature. S-glutathionylation of S100A9 modifies its effects on leukocyte adhesion. Both S100A8 forms inhibit mast cell activation, at least partially by scavenging reactive oxygen species required for signaling. Conversely, S100A12 activates and sequesters mast cells. However S100A12 suppresses proinflammatory cytokine induction by SAA-activated monocytes and macrophages, and inhibits matrix metalloprotease activity. We propose that the abundance and types of cells expressing calgranulins in particular microenvironments, their relative concentrations and post-translational modifications may have distinct functional outcomes, including those that are protective, at different stages of atherogenesis.


Subject(s)
Atherosclerosis/metabolism , Free Radical Scavengers/metabolism , Leukocyte L1 Antigen Complex/metabolism , Signal Transduction , Atherosclerosis/genetics , Atherosclerosis/pathology , Foam Cells/metabolism , Foam Cells/pathology , Humans , Leukocyte L1 Antigen Complex/genetics , Oxidants/metabolism , Peroxidase/genetics , Peroxidase/metabolism , Toll-Like Receptor 4/genetics , Toll-Like Receptor 4/metabolism
19.
Infect Immun ; 81(11): 3975-83, 2013 Nov.
Article in English | MEDLINE | ID: mdl-23940207

ABSTRACT

To protect against invading bacteria, oral epithelial cells appear to use two effector antimicrobial peptides (AMPs): calprotectin (S100A8-S100A9 heterodimer [S100A8/A9]) in the cytosol and cathelicidin antimicrobial protein (CAMP) in endosomes. We sought to learn whether innate immunity might be augmented benignly to increase resistance against invasive bacteria. Epithelial cells were transiently transfected with mRNA constructs containing either the CAMP, S100A8, and S100A9 open reading frames, A8-IRES-A9 (fusion sequence), or A8-nIRES-A9 (fusion with native internal ribosome entry site [IRES] sequence). CAMP, S100A8, and S100A9 protein levels generally peaked between 16 and 44 h after mRNA transfection, depending on the construct; CAMP was processed to LL-37 over time. Following transfection with the respective mRNAs, CAMP and S100A8/A9 each independently increased resistance of epithelial cells to invasion by Listeria and Salmonella for up to 48 h; tandem S100A8/A9 constructs were also effective. Cotransfection to express S100A8/A9 and CAMP together augmented resistance, but synergy was not seen. Independent of the new proteins produced, transfection reduced cell viability after 48 h by 20%, with only 2% attributable to apoptosis. Taken together, these results suggest that epithelial cell resistance to invasive pathogens can be augmented by transient transfection of antimicrobial mRNAs into epithelial cells.


Subject(s)
Antimicrobial Cationic Peptides/immunology , Epithelial Cells/immunology , Epithelial Cells/microbiology , Leukocyte L1 Antigen Complex/immunology , Listeria/immunology , RNA, Messenger/metabolism , Salmonella/immunology , Antimicrobial Cationic Peptides/genetics , Cell Line , Gene Expression , Humans , Leukocyte L1 Antigen Complex/genetics , RNA, Messenger/genetics , Transfection , Cathelicidins
20.
J Am Chem Soc ; 135(47): 17804-17, 2013 Nov 27.
Article in English | MEDLINE | ID: mdl-24245608

ABSTRACT

Human calprotectin (CP) is an antimicrobial protein that coordinates Mn(II) with high affinity in a Ca(II)-dependent manner at an unusual histidine-rich site (site 2) formed at the S100A8/S100A9 dimer interface. We present a 16-member CP mutant family where mutations in the S100A9 C-terminal tail (residues 96-114) are employed to evaluate the contributions of this region, which houses three histidines and four acidic residues, to Mn(II) coordination at site 2. The results from analytical size-exclusion chromatography, Mn(II) competition titrations, and electron paramagnetic resonance spectroscopy establish that the C-terminal tail is essential for high-affinity Mn(II) coordination by CP in solution. The studies indicate that His103 and His105 (HXH motif) of the tail complete the Mn(II) coordination sphere in solution, affording an unprecedented biological His6 site. These solution studies are in agreement with a Mn(II)-CP crystal structure reported recently (Damo, S. M.; et al. Proc. Natl. Acad. Sci. U.S.A. 2013, 110, 3841). Remarkably high-affinity Mn(II) binding is retained when either H103 or H105 are mutated to Ala, when the HXH motif is shifted from positions 103-105 to 104-106, and when the human tail is substituted by the C-terminal tail of murine S100A9. Nevertheless, antibacterial activity assays employing human CP mutants reveal that the native disposition of His residues is important for conferring growth inhibition against Escherichia coli and Staphylococcus aureus. Within the S100 family, the S100A8/S100A9 heterooligomer is essential for providing high-affinity Mn(II) binding; the S100A7, S100A9(C3S), S100A12, and S100B homodimers do not exhibit such Mn(II)-binding capacity.


Subject(s)
Anti-Bacterial Agents/chemistry , Chelating Agents/chemistry , Leukocyte L1 Antigen Complex/chemistry , Manganese/metabolism , Amino Acid Sequence , Anti-Bacterial Agents/metabolism , Anti-Bacterial Agents/pharmacology , Bacteria/drug effects , Chelating Agents/metabolism , Chelating Agents/pharmacology , Humans , Leukocyte L1 Antigen Complex/genetics , Leukocyte L1 Antigen Complex/metabolism , Leukocyte L1 Antigen Complex/pharmacology , Models, Molecular , Molecular Sequence Data , Mutation , Protein Denaturation , Protein Multimerization
SELECTION OF CITATIONS
SEARCH DETAIL