Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 92
1.
PLoS One ; 16(11): e0260188, 2021.
Article En | MEDLINE | ID: mdl-34793556

Chronic inflammation can cause oviduct mucosal damage and immune dysfunction, leading to infertility, early pregnancy loss, ectopic pregnancy, tumors, and a decrease in reproductive capacities in female animals. Estrogen can suppress immune responses in different tissues and oviducts, and regulate the oviduct immune balance; however, the underlying mechanisms remain unclear. The objective of this study was to explore the mechanism of estrogen-regulated oviduct mucosal immunity and discover new estrogen targets for regulating oviduct mucosal immune homeostasis. Sheep oviduct epithelial cells (SOECs) were treated with 17-ß estradiol (E2). Transcriptome sequencing and analysis showed differentially expressed S100 calcium-binding protein A (S100A) genes that may participate in the oviduct mucosa immunoregulation of estrogen. Quantitative polymerase chain reaction and immunocytochemistry analysis showed that S100A8 expression changed dynamically in E2-treated SOECs and peaked after 7 h of treatment. Estrogen nuclear receptors and G protein-coupled membrane receptors promoted E2-dependent S100A8 upregulation. The S100A8 gene was disrupted using the clustered regularly interspaced short palindromic repeats (CRISPR)/CRISPR-associated protein 9 method. Levels of inflammatory factors interleukin (IL)-1ß and IL-4 were significantly upregulated in S100A8-knockdown SOECs, whereas those of the anti-inflammatory factor IL-10 was downregulated. Following S100A8 knockdown in SOECs treated with E2 for 7 h, IL-10 levels increased significantly. Estrogen affected oviduct mucosa immune function and dynamically regulated S100A8 in SOECs. S100A8 knockdown caused an excessive immune response, indicating that S100A8 is beneficial for maintaining immune homeostasis in the oviduct mucosa. Moreover, estrogen can compensate for the effect of S100A8 knockdown by upregulating IL-10.


Calgranulin A/metabolism , Epithelial Cells/metabolism , Estrogens/metabolism , Homeostasis/immunology , Immunity/immunology , Mucous Membrane/metabolism , Oviducts/metabolism , Animals , Calgranulin A/immunology , Epithelial Cells/immunology , Estradiol/immunology , Estradiol/metabolism , Estrogens/immunology , Female , Mucous Membrane/immunology , Oviducts/immunology , Sheep/immunology , Sheep/metabolism , Up-Regulation/immunology
2.
Am J Physiol Lung Cell Mol Physiol ; 321(2): L451-L465, 2021 08 01.
Article En | MEDLINE | ID: mdl-34161747

Millions of people who survive sepsis each year are rehospitalized and die due to late pulmonary complications. To prevent and treat these complications, biomarkers and molecular mediators must be identified. Persistent immune reprogramming in the form of immunoparalysis and impaired host defense is proposed to mediate late pulmonary complications after sepsis, particularly new pulmonary infections. However, immune reprogramming may also involve enhanced/primed responses to secondary stimuli, although their contribution to long-term sepsis complications remains understudied. We hypothesize that enhanced/primed immune responses in the lungs of sepsis survivors are associated with late pulmonary complications. To this end, we developed a murine sepsis model using cecal ligation and puncture (CLP) followed 3 wk later by administration of intranasal lipopolysaccharide to induce inflammatory lung injury. Mice surviving sepsis exhibit enhanced lung injury with increased alveolar permeability, neutrophil recruitment, and enhanced Ly6Chi monocyte Tnf expression. To determine the mediators of enhanced lung injury, we performed flow cytometry and RNA sequencing of lungs 3 wk after CLP, prior to lipopolysaccharide. Sepsis survivor mice showed expanded Ly6Chi monocytes populations and increased expression of many inflammatory genes. Of these, S100A8/A9 was also elevated in the circulation of human sepsis survivors for months after sepsis, validating our model and identifying S100A8/A9 as a potential biomarker and therapeutic target for long-term pulmonary complications after sepsis. These data provide new insight into the importance of enhanced/primed immune responses in survivors of sepsis and establish a foundation for additional investigation into the mechanisms mediating this response.


Lipopolysaccharides/toxicity , Lung Injury/immunology , Sepsis/immunology , Animals , Calgranulin A/immunology , Calgranulin B/immunology , Female , Humans , Inflammation/chemically induced , Inflammation/immunology , Inflammation/pathology , Lung Injury/chemically induced , Lung Injury/pathology , Male , Mice , Monocytes/immunology , Monocytes/pathology , Sepsis/chemically induced , Sepsis/pathology , Tumor Necrosis Factor-alpha/immunology
3.
Cell Rep ; 35(3): 109012, 2021 04 20.
Article En | MEDLINE | ID: mdl-33882312

Caspase-11 sensing of intracellular lipopolysaccharide (LPS) plays critical roles during infections and sepsis. However, the key cell types that sense intracellular LPS and their contributions to the host responses at the organismal level are not completely clear. Here, we show that macrophage/monocyte-specific caspase-11 plays a dominant role in mediating the pathological manifestations of endotoxemia, including gasdermin D (GSDMD) activation, interleukin (IL)-1ß, IL-18, and damage-associated molecular pattern (DAMP) release, tissue damage, and death. Surprisingly, caspase-11 expression in CD11c+ cells and intestinal epithelial cells (IECs) plays minor detrimental roles in LPS shock. In contrast, caspase-11 expression in neutrophils is dispensable for LPS-induced lethality. Importantly, caspase-11 sensing of intracellular LPS in LyzM+ myeloid cells and MRP8+ neutrophils, but not CD11c+ cells and IECs, is necessary for bacterial clearance and host survival during intracellular bacterial infection. Thus, we reveal hierarchical cell-type-specific roles of caspase-11 that govern the host-protective and host-detrimental functions of the cytosolic LPS surveillance.


Caspases, Initiator/genetics , Lipopolysaccharides/toxicity , Macrophages, Peritoneal/immunology , Neutrophils/immunology , Shock, Septic/immunology , Spleen/immunology , Animals , Burkholderia/growth & development , Burkholderia/pathogenicity , CD11 Antigens/genetics , CD11 Antigens/immunology , Calgranulin A/genetics , Calgranulin A/immunology , Caspases, Initiator/immunology , Dendritic Cells/immunology , Dendritic Cells/microbiology , Epithelial Cells/immunology , Epithelial Cells/microbiology , Female , Gene Expression Regulation , Interleukin-18/genetics , Interleukin-18/immunology , Interleukin-1beta/genetics , Interleukin-1beta/immunology , Liver/immunology , Liver/microbiology , Macrophages, Peritoneal/microbiology , Male , Mice , Mice, Transgenic , Microfilament Proteins/genetics , Microfilament Proteins/immunology , Monocytes/immunology , Monocytes/microbiology , Neutrophils/microbiology , Phosphate-Binding Proteins/genetics , Phosphate-Binding Proteins/immunology , Pore Forming Cytotoxic Proteins/genetics , Pore Forming Cytotoxic Proteins/immunology , Shock, Septic/genetics , Shock, Septic/microbiology , Shock, Septic/mortality , Signal Transduction , Spleen/microbiology , Survival Analysis
4.
Expert Rev Clin Immunol ; 17(5): 431-443, 2021 05.
Article En | MEDLINE | ID: mdl-33750254

INTRODUCTION: Coronavirus disease 2019 (COVID-19) is an infectious disease caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection. Although demographic and clinical parameters such as sex, age, comorbidities, genetic background and various biomarkers have been identified as risk factors, there is an unmet need to predict the risk and onset of severe inflammatory disease leading to poor clinical outcomes. In addition, very few mechanistic biomarkers are available to inform targeted treatment of severe (auto)-inflammatory conditions associated with COVID-19. Calprotectin, also known as S100A8/S100A9, MRP8/14 (Myeloid-Related Protein) or L1, is a heterodimer involved in neutrophil-related inflammatory processes. In COVID-19 patients, calprotectin levels were reported to be associated with poor clinical outcomes such as significantly reduced survival time, especially in patients with severe pulmonary disease. AREAS COVERED: Pubmed was searched using the following keywords: Calprotectin + COVID19, S100A8/A9 + COVID19, S100A8 + COVID-19, S100A9 + COVID-19, MRP8/14 + COVID19; L1 + COVID-19 between May 2020 and 8 March 2021. The results summarized in this review provide supporting evidence and propose future directions that define calprotectin as an important biomarker in COVID-19. EXPERT OPINION: Calprotectin represents a promising serological biomarker for the risk assessment of COVID-19 patients.


ATP-Binding Cassette Transporters , COVID-19 , Calgranulin A , Calgranulin B , Severity of Illness Index , ATP-Binding Cassette Transporters/blood , ATP-Binding Cassette Transporters/immunology , Biomarkers/blood , COVID-19/blood , COVID-19/immunology , Calgranulin A/blood , Calgranulin A/immunology , Calgranulin B/blood , Calgranulin B/immunology , Humans
5.
J Immunol ; 206(7): 1597-1608, 2021 04 01.
Article En | MEDLINE | ID: mdl-33579725

Coronavirus disease 2019 (COVID-19) is associated with immune dysregulation and cytokine storm. Exploring the immune-inflammatory characteristics of COVID-19 patients is essential to reveal pathogenesis and predict progression. In this study, COVID-19 patients showed decreased CD3+, CD4+, and CD8+ T cells but increased neutrophils in circulation, exhibiting upregulated neutrophil-to-lymphocyte and neutrophil-to-CD8+ T cell ratio. IL-6, TNF-α, IL-1ß, IL-18, IL-12/IL-23p40, IL-10, Tim-3, IL-8, neutrophil extracellular trap-related proteinase 3, and S100A8/A9 were elevated, whereas IFN-γ and C-type lectin domain family 9 member A (clec9A) were decreased in COVID-19 patients compared with healthy controls. When compared with influenza patients, the expressions of TNF-α, IL-18, IL-12/IL-23p40, IL-8, S100A8/A9 and Tim-3 were significantly increased in critical COVID-19 patients, and carcinoembryonic Ag, IL-8, and S100A8/A9 could serve as clinically available hematologic indexes for identifying COVID-19 from influenza. Moreover, IL-6, IL-8, IL-1ß, TNF-α, proteinase 3, and S100A8/A9 were increased in bronchoalveolar lavage fluid of severe/critical patients compared with moderate patients, despite decreased CD4+ T cells, CD8+ T cells, B cells, and NK cells. Interestingly, bronchoalveolar IL-6, carcinoembryonic Ag, IL-8, S100A8/A9, and proteinase 3 were found to be predictive of COVID-19 severity and may serve as potential biomarkers for predicting COVID-19 progression and potential targets in therapeutic intervention of COVID-19.


COVID-19 , Inflammation Mediators , SARS-CoV-2 , Severity of Illness Index , Aged , COVID-19/blood , COVID-19/immunology , Calgranulin A/blood , Calgranulin A/immunology , Calgranulin B/blood , Calgranulin B/immunology , Cytokines/blood , Cytokines/immunology , Disease Progression , Female , Hepatitis A Virus Cellular Receptor 2/blood , Hepatitis A Virus Cellular Receptor 2/immunology , Humans , Inflammation Mediators/blood , Inflammation Mediators/immunology , Leukocyte Count , Male , Middle Aged , Myeloblastin/blood , Myeloblastin/immunology , Retrospective Studies , SARS-CoV-2/immunology , SARS-CoV-2/metabolism
6.
J Leukoc Biol ; 109(1): 67-72, 2021 01.
Article En | MEDLINE | ID: mdl-32869342

Severe cases of coronavirus disease 2019 (COVID-19) are regularly complicated by respiratory failure. Although it has been suggested that elevated levels of blood neutrophils associate with worsening oxygenation in COVID-19, it is unknown whether neutrophils are drivers of the thrombo-inflammatory storm or simple bystanders. To better understand the potential role of neutrophils in COVID-19, we measured levels of the neutrophil activation marker S100A8/A9 (calprotectin) in hospitalized patients and determined its relationship to severity of illness and respiratory status. Patients with COVID-19 (n = 172) had markedly elevated levels of calprotectin in their blood. Calprotectin tracked with other acute phase reactants including C-reactive protein, ferritin, lactate dehydrogenase, and absolute neutrophil count, but was superior in identifying patients requiring mechanical ventilation. In longitudinal samples, calprotectin rose as oxygenation worsened. When tested on day 1 or 2 of hospitalization (n = 94 patients), calprotectin levels were significantly higher in patients who progressed to severe COVID-19 requiring mechanical ventilation (8039 ± 7031 ng/ml, n = 32) as compared to those who remained free of intubation (3365 ± 3146, P < 0.0001). In summary, serum calprotectin levels track closely with current and future COVID-19 severity, implicating neutrophils as potential perpetuators of inflammation and respiratory compromise in COVID-19.


COVID-19 , Calgranulin A , Calgranulin B , Neutrophil Activation , Neutrophils , SARS-CoV-2 , COVID-19/blood , COVID-19/immunology , COVID-19/pathology , COVID-19/therapy , Calgranulin A/blood , Calgranulin A/immunology , Calgranulin B/blood , Calgranulin B/immunology , Female , Hospitalization , Humans , Male , Middle Aged , Neutrophils/immunology , Neutrophils/metabolism , Neutrophils/pathology , SARS-CoV-2/immunology , SARS-CoV-2/metabolism , Severity of Illness Index , Time Factors
7.
Elife ; 92020 04 07.
Article En | MEDLINE | ID: mdl-32255429

Multifunctional proteins are evolutionary puzzles: how do proteins evolve to satisfy multiple functional constraints? S100A9 is one such multifunctional protein. It potently amplifies inflammation via Toll-like receptor four and is antimicrobial as part of a heterocomplex with S100A8. These two functions are seemingly regulated by proteolysis: S100A9 is readily degraded, while S100A8/S100A9 is resistant. We take an evolutionary biochemical approach to show that S100A9 evolved both functions and lost proteolytic resistance from a weakly proinflammatory, proteolytically resistant amniote ancestor. We identify a historical substitution that has pleiotropic effects on S100A9 proinflammatory activity and proteolytic resistance but has little effect on S100A8/S100A9 antimicrobial activity. We thus propose that mammals evolved S100A8/S100A9 antimicrobial and S100A9 proinflammatory activities concomitantly with a proteolytic 'timer' to selectively regulate S100A9. This highlights how the same mutation can have pleiotropic effects on one functional state of a protein but not another, thus facilitating the evolution of multifunctionality.


A single protein sometimes does multiple jobs. For instance, our immune system uses a small number of multipurpose proteins to respond quickly to a large number of threats. One example is the protein S100A9. It acts as an antimicrobial by preventing microbes from getting the nutrients they need, while also stimulating inflammation by inducing the release of molecules that recruit white blood cells. S100A9, like all proteins, is made up of a chain of small building blocks. These building blocks interact with each other and with other molecules in the environment. The sequence of the building blocks thus determines what jobs the protein can do. Therefore, a single change to the sequence of building blocks can have a dramatic effect: one change might render the protein faulty, while another change might allow it to do a new job. Proteins face similar challenges humans do when trying to do several things at once. A person driving a car while using their phone will not do either task well. Likewise, a protein that does two jobs faces challenges a single-purpose protein does not. Harman et al. were interested in how S100A9 was able to evolve and maintain its dual functionality, despite this potential problem. They started by asking when S100A9 acquired its two purposes. They measured the antimicrobial and inflammatory activity of S100A9 proteins from humans, mice and opossums. The activities of S100A9 in these species was similar, suggesting that S100A9 acquired its different jobs in the ancestor of mammals, some 160 million years ago. Next, Harman et al. computationally reconstructed ancestral forms of S100A9 by comparing hundreds of similar proteins and building an evolutionary tree. They then measured the antimicrobial and inflammatory activity of these ancestral proteins. By comparing the last ancestor that did not have these activities to the first ancestor that did, they identified the sequence changes that gave S100A9 its dual activity. Importantly, these changes are located in separate regions of the protein, meaning they could occur independently, without affecting each other. Further, the same sequence change that converted S100A9 into an inflammatory signal also introduced a mechanism to regulate this activity. The results suggest that a small number of sequence changes ­ or even a single change ­ can make a protein more versatile. This means that evolving multipurpose proteins may not be as difficult as is often thought.


Calgranulin B/genetics , Calgranulin B/immunology , Evolution, Molecular , Immunity, Innate/genetics , Animals , Calgranulin A/genetics , Calgranulin A/immunology , Humans , Inflammation , Mammals/immunology , Proteolysis , Toll-Like Receptor 4/immunology
8.
J Clin Invest ; 130(6): 3098-3112, 2020 06 01.
Article En | MEDLINE | ID: mdl-32134742

Neutrophil accumulation is associated with lung pathology during active tuberculosis (ATB). However, the molecular mechanism or mechanisms by which neutrophils accumulate in the lung and contribute to TB immunopathology are not fully delineated. Using the well-established mouse model of TB, our new data provide evidence that the alarmin S100A8/A9 mediates neutrophil accumulation during progression to chronic TB. Depletion of neutrophils or S100A8/A9 deficiency resulted in improved Mycobacterium tuberculosis (Mtb) control during chronic but not acute TB. Mechanistically, we demonstrate that, following Mtb infection, S100A8/A9 expression is required for upregulation of the integrin molecule CD11b specifically on neutrophils, mediating their accumulation during chronic TB disease. These findings are further substantiated by increased expression of S100A8 and S100A9 mRNA in whole blood in human TB progressors when compared with nonprogressors and rapidly decreased S100A8/A9 protein levels in the serum upon TB treatment. Furthermore, we demonstrate that S100A8/A9 serum levels along with chemokines are useful in distinguishing between ATB and asymptomatic Mtb-infected latent individuals. Thus, our results support targeting S100A8/A9 pathways as host-directed therapy for TB.


CD11b Antigen/immunology , Calgranulin A/immunology , Calgranulin B/immunology , Mycobacterium tuberculosis/immunology , Neutrophil Infiltration/immunology , Neutrophils/immunology , Tuberculosis/immunology , Animals , CD11b Antigen/genetics , Calgranulin A/genetics , Calgranulin B/genetics , Mice , Mice, Knockout , Neutrophils/pathology , Tuberculosis/genetics , Tuberculosis/pathology , Tuberculosis/therapy
9.
J Immunol ; 204(9): 2589-2599, 2020 05 01.
Article En | MEDLINE | ID: mdl-32198140

S100A8 is a damage-associated molecular pattern protein released by monocytes, playing a decisive role in the development of inflammation. Nonresolving inflammation is viewed as a driving force in tumorigenesis, and its role in tumor immune escape also attracted attentions. PD-1/PD-L1 axis is a critical determinant of physiological immune homeostasis, and anti-PD-1 or PD-L1 therapy has becoming the most exciting field of oncology. Multiple regulation mechanisms have been contributed to PD-L1 expression modulation including inflammatory mediators. In this study we reported that S100A8 significantly induced PD-L1 expression in monocytes/macrophages but not in tumor cells. S100A8 induced PD-L1 transcription through the TLR4 receptor and multiple crucial pathways of inflammation process. S100A8 modulated the histone modification of the PD-L1 promoter in monocytes/macrophages. S100A8-pretreated macrophages had immunosuppressive function and attenuated the antitumor ability of CTLs both in vitro and in vivo. A highly positive correlation existed between S100A8 expression and PD-L1 expression in human cancer specimens. To our knowledge, our study uncovers a novel molecular mechanism for regulating PD-L1 transcription by an inflammatory mediator S100A8, and reveals the importance of comprehensive understanding the role of inflammation in tumorigenesis as well as in tumor immune escape.


B7-H1 Antigen/immunology , Calgranulin A/immunology , Inflammation/immunology , Macrophages/immunology , Neoplasms/genetics , Tumor Escape/immunology , Animals , Carcinogenesis/immunology , Cell Line , Cell Line, Tumor , Humans , Male , Mice , Mice, Inbred BALB C , Mice, Inbred NOD , Mice, SCID , Monocytes/immunology , Promoter Regions, Genetic/immunology , RAW 264.7 Cells , THP-1 Cells , Toll-Like Receptor 4/immunology , Transcription, Genetic/immunology
10.
Front Immunol ; 11: 599947, 2020.
Article En | MEDLINE | ID: mdl-33643287

Psoriasis is one of the most common immune-mediated inflammatory skin diseases. Expression and secretion of two pro-inflammatory molecules of the S100-alarmin family, S100A8 and S100A9, in keratinocytes is a hallmark of psoriasis, which is also characterized by an altered differentiation of keratinocytes. Dimers of S100A8/S100A9 (calprotectin) bind to Toll-like receptor 4 and induce an inflammatory response in target cells. Targeted deletion of S100A9 reduced the inflammatory phenotype of psoriasis-like inflammation in mice. A role of S100-alarmins in differentiation and activation of keratinocytes was suggested but has been never shown in primary keratinocytes. We now confirm that induction of S100-alarmins in an imiquimod-induced murine model of psoriasis-like skin inflammation was associated with an increased expression of interleukin (IL)-1α, IL-6, IL-17A, or TNFα. This association was confirmed in transcriptome data obtained from controls, lesional and non-lesional skin of psoriasis patients, and a down-regulation of S100-alarmin expression after IL-17 directed therapy. However, analyzing primary S100A9-/- keratinocytes we found that expression of S100A8/S100A9 has no significant role for the maturation and inflammatory response pattern of keratinocytes. Moreover, keratinocytes are no target cells for the pro-inflammatory effects of S100A8/S100A9. However, different cytokines, especially IL-17A and F, highly abundant in psoriasis, strongly induced expression of S100-alarmins preferentially during early maturation stages of keratinocytes. Our data indicate that expression of S100A8 and S100A9 does not primarily influence maturation or activation of keratinocytes but rather represents the inflammatory response of these cells during psoriasis.


Calgranulin A/immunology , Calgranulin B/immunology , Interleukin-17/immunology , Keratinocytes/immunology , Psoriasis/immunology , Animals , Calgranulin A/genetics , Calgranulin B/genetics , Inflammation/genetics , Inflammation/immunology , Inflammation/pathology , Interleukin-17/genetics , Keratinocytes/pathology , Mice , Mice, Inbred BALB C , Mice, Knockout , Psoriasis/genetics , Psoriasis/pathology
11.
J Oral Pathol Med ; 49(4): 320-327, 2020 Apr.
Article En | MEDLINE | ID: mdl-31145494

BACKGROUND: Candida albicans (C albicans) is the most common fungal pathogen causing opportunistic infections. IL17 (IL17A) is a vital mediator of antifungal immunity. The aim of the study was to investigate the effect of recombinant human interleukin 17A (rhIL17A) on human oral mucosal epithelial cells (hOMECs) defending against C albicans infection. METHODS: Human oral mucosal epithelial cells were divided into four groups: C albicans+ (MOI = 0.1), rhIL17A+ (100 µg/L), rhIL17A + C albicans+ (MOI = 0.1, rhIL17A:100 µg/L) and blank control. Then, C albicans growth was observed after 24 hours. Human beta-2 defensin (hBD-2), S100A8 and LL-37 in supernatants and their mRNAs in cells were measured by enzyme-linked immunosorbent assay and reverse transcription-polymerase chain reaction, respectively. RESULTS: In C albicans+ group, C albicans hyphae formation and the death of infected hOMECs were observed. However, in the rhIL17A + C albicans+ group, IL17 inhibited both hypha formation, and C albicans from infecting hOMECs and its further growth. There was no statistical significance in adhesion rates of C albicans to hOMECs. Compared with the control group, the level of hBD-2 mRNA has increased, while hBD-2 and hBD-2 mRNA levels in the rhIL17A + C albicans+ group were the highest. Both hBD-2 and hBD-2 mRNA levels were higher in the rhIL17A+ group than in the C albicans+ group. S100A8 and LL-37 mRNAs have similar trend, and both upregulated after treatment with rhIL17A; however, protein levels were undetectable. CONCLUSION: Recombinant human interleukin 17A may inhibit C albicans from infecting hOMECs by affecting the growth and reproduction of C albicans as well as the formation of hyphae. Besides, rhIL17A might induce hBD-2, S100A8 and LL-37 secretion from hOMECs to strengthen their anti-infective ability.


Candida albicans/growth & development , Epithelial Cells/immunology , Interleukin-17/pharmacology , beta-Defensins/immunology , Anti-Infective Agents , Antimicrobial Cationic Peptides/immunology , Calgranulin A/immunology , Candida albicans/drug effects , Cells, Cultured , Epithelial Cells/microbiology , Humans , Mouth Mucosa/cytology , Recombinant Proteins/pharmacology , Cathelicidins
12.
Mol Med Rep ; 20(3): 2476-2483, 2019 Sep.
Article En | MEDLINE | ID: mdl-31322196

Atopic dermatitis (AD) is an inflammatory skin disorder caused by immunological dysregulation and genetic factors. Whether the expression levels of cytokine and skin barrier protein were altered by S100 calcium binding protein A8 (S100A8) and S100A9 in human keratinocytic HaCaT cells was examined in the present study. Alterations of cytokine expression were examined by ELISA following treatment with S100A8/9 and various signal protein­specific inhibitors. Activation of the mitogen activated protein kinase (MAPK) pathway and nuclear factor (NF)­κB was evaluated by using western blotting and an NF­κB activity test, respectively. The expression levels of interleukin (IL)­6, IL­8 and monocyte chemoattractant protein­1 increased following treatment with S100A8 and S100A9, and the increase was significantly blocked by specific signaling pathway inhibitors, including toll­like receptor 4 inhibitor (TLR4i), rottlerin, PD98059, SB203580 and BAY­11­7085. Extracellular signal­regulated kinase (ERK) and p38 MAPK pathways were activated in a time­dependent manner following treatment with S100A8 and S100A9. Phosphorylation of ERK and p38 MAPK were blocked by TLR4i and rottlerin. S100A8 and S100A9 induced translocation of NF­κB in a time­dependent manner, and the activation of NF­κB was inhibited by TLR4i, rottlerin, PD98059 and SB203580. In addition, S100A8 and S100A9 decreased the expression of skin barrier proteins, filaggrin and loricrin. These results may help to elucidate the pathogenic mechanisms of AD and develop clinical strategies for controlling AD.


Calgranulin A/immunology , Calgranulin B/immunology , Cytokines/immunology , Keratinocytes/immunology , Membrane Proteins/immunology , S100 Proteins/immunology , Cell Line , Cytokines/analysis , Dermatitis, Atopic/immunology , Filaggrin Proteins , Humans , MAP Kinase Signaling System , Membrane Proteins/analysis , NF-kappa B/analysis , NF-kappa B/immunology , S100 Proteins/analysis
13.
Nat Commun ; 10(1): 1685, 2019 04 11.
Article En | MEDLINE | ID: mdl-30976008

Neonatal sepsis is characterized by hyperinflammation causing enhanced morbidity and mortality compared to adults. This suggests differences in the response towards invading threats. Here we investigate activated cord blood macrophages (CBMΦ) in comparison to adult macrophages (PBMΦ), indicating incomplete interferon gamma (IFN-γ) and interleukin 10 (IL-10)-induced activation of CBMΦ. CBMΦ show reduced expression of phagocytosis receptors and cytokine expression in addition to altered energy metabolism. In particular, IFN-γ as well as IL-10-activated CBMΦ completely fail to increase glycolysis and furthermore show reduced activation of the mTOR pathway, which is important for survival in sepsis. MTOR inhibition by rapamycin equalizes cytokine production in CBMΦ and PBMΦ. Finally, incubation of PBMΦ with cord blood serum or S100A8/A9, which is highly expressed in neonates, suppresses mTOR activation, prevents glycolysis and the expression of an PBMΦ phenotype. Thus, a metabolic alteration is apparent in CBMΦ, which might be dependent on S100A8/A9 expression.


Cytotoxicity, Immunologic , Energy Metabolism/immunology , Macrophages/metabolism , Adult , Age Factors , Calgranulin A/immunology , Calgranulin A/metabolism , Calgranulin B/immunology , Calgranulin B/metabolism , Cell Differentiation/immunology , Cells, Cultured , Fetal Blood/cytology , Glycolysis/immunology , Healthy Volunteers , Humans , Infant, Newborn , Interferon-gamma/immunology , Interferon-gamma/metabolism , Interleukin-10/immunology , Interleukin-10/metabolism , Macrophages/immunology , Primary Cell Culture , Sepsis/immunology , Signal Transduction/drug effects , Signal Transduction/immunology , Sirolimus/pharmacology , TOR Serine-Threonine Kinases/immunology , TOR Serine-Threonine Kinases/metabolism
14.
Int J Cancer ; 145(2): 569-575, 2019 07 15.
Article En | MEDLINE | ID: mdl-30414170

The metastatic dissemination of cancer cells to remote areas of the body is the most problematic aspect in cancer patients. Among cancers, melanomas are notoriously difficult to treat due to their significantly high metastatic potential even during early stages. Hence, the establishment of advanced therapeutic approaches to regulate metastasis is required to overcome the melanoma disease. An accumulating mass of evidence has indicated a critical role of extracellular S100A8/A9 in melanoma distant metastasis. Lung S100A8/A9 is induced by melanoma cells from distant organs and it attracts these cells to its enriched lung environment since melanoma cells possess several receptors that sense the S100A8/A9 ligand. We hence aimed to develop a neutralizing antibody against S100A8/A9 that would efficiently block melanoma lung metastasis. Our protocol provided us with one prominent antibody, Ab45 that efficiently suppressed not only S100A8/A9-mediated melanoma mobility but also lung tropic melanoma metastasis in a mouse model. This prompted us to make chimeric Ab45, a chimera antibody consisting of mouse Ab45-Fab and human IgG2-Fc. Chimeric Ab45 also showed significant inhibition of the lung metastasis of melanoma. From these results, we have high hopes that the newly produced antibody will become a potential biological tool to block melanoma metastasis in future clinical settings.


Antibodies, Monoclonal/administration & dosage , Lung Neoplasms/drug therapy , Lung Neoplasms/secondary , Melanoma/drug therapy , Animals , Antibodies, Monoclonal/pharmacology , Antibodies, Neutralizing/administration & dosage , Antibodies, Neutralizing/pharmacology , Calgranulin A/immunology , Calgranulin B/immunology , Cell Line, Tumor , Gene Expression Regulation, Neoplastic/drug effects , Humans , Lung Neoplasms/metabolism , Melanoma/metabolism , Mice , Treatment Outcome , Tumor Microenvironment/drug effects , Xenograft Model Antitumor Assays
15.
Cell Rep ; 24(12): 3296-3311.e6, 2018 09 18.
Article En | MEDLINE | ID: mdl-30232010

Inflammatory bowel disease (IBD) is prevalent, but the mechanisms underlying disease development remain elusive. We identify a role for the E3 ubiquitin ligase RNF5 in IBD. Intestinal epithelial cells (IECs) express a high level of RNF5, while the colon of Rnf5-/- mice exhibits activated dendritic cells and intrinsic inflammation. Rnf5-/- mice exhibit severe acute colitis following dextran sodium sulfate (DSS) treatment. S100A8 is identified as an RNF5 substrate, resulting in S100A8 ubiquitination and proteasomal-dependent degradation that is attenuated upon inflammatory stimuli. Loss of RNF5 from IECs leads to enhanced S100A8 secretion, which induces mucosal CD4+ T cells, resulting in Th1 pro-inflammatory responses. Administration of S100A8-neutralizing antibodies to DSS-treated Rnf5-/- mice attenuates acute colitis development and increases survival. An inverse correlation between RNF5 and S100A8 protein expression in IECs of IBD patients coincides with disease severity. Collectively, RNF5-mediated regulation of S100A8 stability in IECs is required for the maintenance of intestinal homeostasis.


Calgranulin A/metabolism , Colitis, Ulcerative/metabolism , Enterocytes/metabolism , Membrane Proteins/genetics , Ubiquitin-Protein Ligases/genetics , Animals , Antibodies, Neutralizing/immunology , Antibodies, Neutralizing/therapeutic use , CD4-Positive T-Lymphocytes/immunology , Calgranulin A/immunology , Cell Line , Cells, Cultured , Colitis, Ulcerative/drug therapy , HEK293 Cells , Humans , Membrane Proteins/metabolism , Mice , Mice, Inbred C57BL , Protein Stability , Ubiquitin-Protein Ligases/metabolism
16.
Int J Mol Sci ; 19(7)2018 06 21.
Article En | MEDLINE | ID: mdl-29933628

High expression levels of the calcium-binding proteins S100A8 and S100A9 in myeloid cells in kidney transplant rejections are associated with a favorable outcome. Here we investigated the myeloid cell subset expressing these molecules, and their function in inflammatory reactions. Different monocyte subsets were sorted from buffy coats of healthy donors and investigated for S100A8 and S100A9 expression. To characterize S100A9high and S100A9low subsets within the CD14+ classical monocyte subset, intracellular S100A9 staining was combined with flow cytometry (FACS) and qPCR profiling. Furthermore, S100A8 and S100A9 were overexpressed by transfection in primary monocyte-derived macrophages and the THP-1 macrophage cell line to investigate the functional relevance. Expression of S100A8 and S100A9 was primarily found in classical monocytes and to a much lower extent in intermediate and non-classical monocytes. All S100A9+ cells expressed human leukocyte antigen—antigen D related (HLA-DR) on their surface. A small population (<3%) of CD14+ CD11b+ CD33+ HLA-DR− cells, characterized as myeloid derived suppressor cells (MDSCs), also expressed S100A9 to high extent. Overexpression of S100A8 and S00A9 in macrophages led to enhanced extracellular reactive oxygen species (ROS) production, as well as elevated mRNA expression of anti-inflammatory IL-10. The results suggest that the calcium-binding proteins S100A8 and S100A9 in myeloid cells have an immune regulatory effect.


Calgranulin A/immunology , Calgranulin B/immunology , Interleukin-10/immunology , Macrophages/immunology , Myeloid-Derived Suppressor Cells/immunology , Calcium/immunology , Calcium/metabolism , Calgranulin A/genetics , Calgranulin B/genetics , Cell Lineage/genetics , Cell Lineage/immunology , Flow Cytometry , Gene Expression Regulation , HLA-DR Antigens/genetics , HLA-DR Antigens/immunology , Healthy Volunteers , Humans , Immunophenotyping , Interleukin-10/genetics , Macrophages/cytology , Myeloid-Derived Suppressor Cells/cytology , Primary Cell Culture , RNA, Messenger/genetics , RNA, Messenger/immunology , Reactive Oxygen Species/immunology , Reactive Oxygen Species/metabolism , THP-1 Cells
17.
Arthritis Res Ther ; 20(1): 80, 2018 05 02.
Article En | MEDLINE | ID: mdl-29720243

BACKGROUND: Osteoclast-mediated bone erosion is a central feature of rheumatoid arthritis (RA). Immune complexes, present in a large percentage of patients, bind to Fcγ receptors (FcγRs), thereby modulating the activity of immune cells. In this study, we investigated the contribution of FcγRs, and FcγRIV in particular, during antigen-induced arthritis (AIA). METHODS: AIA was induced in knee joints of wild-type (WT), FcγRI,II,III-/-, and FcγRI,II,III,IV-/- mice. Bone destruction, numbers of tartrate-resistant acid phosphatase-positive (TRAP+) osteoclasts, and inflammation were evaluated using histology; expression of the macrophage marker F4/80, neutrophil marker NIMPR14, and alarmin S100A8 was evaluated using immunohistochemistry. The percentage of osteoclast precursors in the bone marrow was determined using flow cytometry. In vitro osteoclastogenesis was evaluated with TRAP staining, and gene expression was assessed using real-time PCR. RESULTS: FcγRI,II,III,IV-/- mice showed decreased bone erosion compared with WT mice during AIA, whereas both the humoral and cellular immune responses against methylated bovine serum albumin were not impaired in FcγRI,II,III,IV-/- mice. The percentage of osteoclast precursors in the bone marrow of arthritic mice and their ability to differentiate into osteoclasts in vitro were comparable between FcγRI,II,III,IV-/- and WT mice. In line with these observations, numbers of TRAP+ osteoclasts on the bone surface during AIA were comparable between the two groups. Inflammation, a process that strongly activates osteoclast activity, was reduced in FcγRI,II,III,IV-/- mice, and of note, mainly decreased numbers of neutrophils were present in the joint. In contrast to FcγRI,II,III,IV-/- mice, AIA induction in knee joints of FcγRI,II,III-/- mice resulted in increased bone erosion, inflammation, and numbers of neutrophils, suggesting a crucial role for FcγRIV in the joint pathology by the recruitment of neutrophils. Finally, significant correlations were found between bone erosion and the number of neutrophils present in the joint as well as between bone erosion and the number of S100A8-positive cells, with S100A8 being an alarmin strongly produced by neutrophils that stimulates osteoclast resorbing activity. CONCLUSIONS: FcγRs play a crucial role in the development of bone erosion during AIA by inducing inflammation. In particular, FcγRIV mediates bone erosion in AIA by inducing the influx of S100A8/A9-producing neutrophils into the arthritic joint.


Arthritis, Experimental/immunology , Bone and Bones/immunology , Calgranulin A/immunology , Calgranulin B/immunology , Neutrophils/immunology , Receptors, IgG/immunology , Animals , Arthritis, Experimental/genetics , Arthritis, Experimental/metabolism , Bone and Bones/metabolism , Bone and Bones/pathology , Calgranulin A/metabolism , Calgranulin B/metabolism , Knee Joint/immunology , Knee Joint/metabolism , Knee Joint/pathology , Macrophages/immunology , Macrophages/metabolism , Mice, Inbred C57BL , Mice, Knockout , Neutrophils/metabolism , Osteoclasts/immunology , Osteoclasts/metabolism , Receptors, IgG/genetics , Receptors, IgG/metabolism , Tartrate-Resistant Acid Phosphatase/immunology , Tartrate-Resistant Acid Phosphatase/metabolism
18.
J Clin Invest ; 128(5): 1852-1866, 2018 05 01.
Article En | MEDLINE | ID: mdl-29611822

Autoimmune diseases, such as psoriasis and arthritis, show a patchy distribution of inflammation despite systemic dysregulation of adaptive immunity. Thus, additional tissue-derived signals, such as danger-associated molecular patterns (DAMPs), are indispensable for manifestation of local inflammation. S100A8/S100A9 complexes are the most abundant DAMPs in many autoimmune diseases. However, regulatory mechanisms locally restricting DAMP activities are barely understood. We now unravel for the first time, to our knowledge, a mechanism of autoinhibition in mice and humans restricting S100-DAMP activity to local sites of inflammation. Combining protease degradation, pull-down assays, mass spectrometry, and targeted mutations, we identified specific peptide sequences within the second calcium-binding EF-hands triggering TLR4/MD2-dependent inflammation. These binding sites are free when S100A8/S100A9 heterodimers are released at sites of inflammation. Subsequently, S100A8/S100A9 activities are locally restricted by calcium-induced (S100A8/S100A9)2 tetramer formation hiding the TLR4/MD2-binding site within the tetramer interphase, thus preventing undesirable systemic effects. Loss of this autoinhibitory mechanism in vivo results in TNF-α-driven fatal inflammation, as shown by lack of tetramer formation in crossing S100A9-/- mice with 2 independent TNF-α-transgene mouse strains. Since S100A8/S100A9 is the most abundant DAMP in many inflammatory diseases, specifically blocking the TLR4-binding site of active S100 dimers may represent a promising approach for local suppression of inflammatory diseases, avoiding systemic side effects.


Alarmins/immunology , Calgranulin A/immunology , Calgranulin B/immunology , Alarmins/chemistry , Alarmins/genetics , Animals , Arthritis/genetics , Arthritis/immunology , Arthritis/pathology , Binding Sites , Calgranulin A/chemistry , Calgranulin A/genetics , Calgranulin B/chemistry , Calgranulin B/genetics , Inflammation/genetics , Inflammation/immunology , Inflammation/pathology , Mice , Mice, Knockout , Proto-Oncogene Proteins c-mdm2/genetics , Proto-Oncogene Proteins c-mdm2/immunology , Psoriasis/genetics , Psoriasis/immunology , Psoriasis/pathology , Toll-Like Receptor 4/genetics , Toll-Like Receptor 4/immunology
19.
Inflamm Res ; 67(7): 609-616, 2018 Jul.
Article En | MEDLINE | ID: mdl-29605872

The pathophysiology of psoriasis is very complex and involves an interplay between immune cells and keratinocytes. The keratinocyte production of calprotectin (S100A8/A9), induced by the inflammatory psoriatic milieu, may be involved in initiating immune cell invasion, as well as in propagating inflammation. However, the exact role of calprotectin in psoriasis remains unclear. Therapeutic approaches utilizing adalimumab, etanercept and ustekinumab are widely used in psoriatic treatment, but their anti-inflammatory mechanisms are not fully understood. The aim of this study was to investigate, by immunohistochemical analysis, the expression of the heterocomplex S100A8/A9 in lesional skin from psoriatic patients undergoing biological therapy with adalimumab, etanercept or ustekinumab. Our results showed that S100A8/A9, absent or present at very low level in skin biopsies from healthy subjects, is dramatically upregulated in each epidermal layer from psoriatic patients. Interestingly, calprotectin was mainly localized in keratinocyte nuclei from psoriatic patients, suggesting a role of S100A8/A9 in keratinocyte nuclear function. Furthermore, we have shown that the biological treatment induced a drastic reduction of S100A8/A9 expression in skin biopsies from treated patients, correlating with PASI reduction. Our results suggest that calprotectin may play a crucial role as a significant marker of inflammation in psoriasis, and that its reduction of expression may be considered a favourable prognostic marker in psoriasis.


Adalimumab , Anti-Inflammatory Agents, Non-Steroidal , Calgranulin A/immunology , Calgranulin B/immunology , Dermatologic Agents , Etanercept , Psoriasis/immunology , Ustekinumab , Adalimumab/pharmacology , Adalimumab/therapeutic use , Adult , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Anti-Inflammatory Agents, Non-Steroidal/therapeutic use , Biological Therapy , Dermatologic Agents/pharmacology , Dermatologic Agents/therapeutic use , Down-Regulation , Etanercept/pharmacology , Etanercept/therapeutic use , Female , Humans , Male , Middle Aged , Psoriasis/drug therapy , Skin/drug effects , Skin/immunology , Ustekinumab/pharmacology , Ustekinumab/therapeutic use
20.
J Biol Chem ; 293(16): 5934-5946, 2018 04 20.
Article En | MEDLINE | ID: mdl-29496993

Chronic low-grade inflammation in the pancreatic islets is observed in individuals with type 2 diabetes, and macrophage levels are elevated in the islets of these individuals. However, the molecular mechanisms underlying the interactions between the pancreatic ß cells and macrophages and their involvement in inflammation are not fully understood. Here, we investigated the role of S100 calcium-binding protein A8 (S100A8), a member of the damage-associated molecular pattern molecules (DAMPs), in ß-cell inflammation. Co-cultivation of pancreatic islets with unstimulated peritoneal macrophages in the presence of palmitate (to induce lipotoxicity) and high glucose (to induce glucotoxicity) synergistically increased the expression and release of islet-produced S100A8 in a Toll-like receptor 4 (TLR4)-independent manner. Consistently, a significant increase in the expression of the S100a8 gene was observed in the islets of diabetic db/db mice. Furthermore, the islet-derived S100A8 induced TLR4-mediated inflammatory cytokine production by migrating macrophages. When human islet cells were co-cultured with U937 human monocyte cells, the palmitate treatment up-regulated S100A8 expression. This S100A8-mediated interaction between islets and macrophages evoked ß-cell apoptosis, which was ameliorated by TLR4 inhibition in the macrophages or S100A8 neutralization in the pancreatic islets. Of note, both glucotoxicity and lipotoxicity triggered S100A8 secretion from the pancreatic islets, which in turn promoted macrophage infiltration of the islets. Taken together, a positive feedback loop between islet-derived S100A8 and macrophages drives ß-cell apoptosis and pancreatic islet inflammation. We conclude that developing therapeutic approaches to inhibit S100A8 may serve to prevent ß-cell loss in patients with diabetes.


Apoptosis , Calgranulin A/immunology , Inflammation/immunology , Insulin-Secreting Cells/immunology , Macrophages/immunology , Animals , Cell Line , Cells, Cultured , Glucose/immunology , Humans , Insulin-Secreting Cells/cytology , Macrophages/cytology , Male , Mice, Inbred C57BL , Palmitates/immunology , Signal Transduction , Toll-Like Receptor 4/immunology
...