Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 1.381
Filter
Add more filters

Publication year range
1.
Nat Immunol ; 24(12): 2021-2031, 2023 Dec.
Article in English | MEDLINE | ID: mdl-37903858

ABSTRACT

S100A8/S100A9 is a proinflammatory mediator released by myeloid cells during many acute and chronic inflammatory disorders. However, the precise mechanism of its release from the cytosolic compartment of neutrophils is unclear. Here, we show that E-selectin-induced rapid S100A8/S100A9 release during inflammation occurs in an NLRP3 inflammasome-dependent fashion. Mechanistically, E-selectin engagement triggers Bruton's tyrosine kinase-dependent tyrosine phosphorylation of NLRP3. Concomitant potassium efflux via the voltage-gated potassium channel KV1.3 mediates ASC oligomerization. This is followed by caspase 1 cleavage and downstream activation of pore-forming gasdermin D, enabling cytosolic release of S100A8/S100A9. Strikingly, E-selectin-mediated gasdermin D pore formation does not result in cell death but is a transient process involving activation of the ESCRT III membrane repair machinery. These data clarify molecular mechanisms of controlled S100A8/S100A9 release from neutrophils and identify the NLRP3/gasdermin D axis as a rapid and reversible activation system in neutrophils during inflammation.


Subject(s)
Inflammasomes , NLR Family, Pyrin Domain-Containing 3 Protein , Humans , Inflammasomes/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Gasdermins , Neutrophils/metabolism , E-Selectin/metabolism , Calgranulin A/metabolism , Calgranulin B/metabolism , Inflammation/metabolism
2.
Nat Immunol ; 22(9): 1118-1126, 2021 09.
Article in English | MEDLINE | ID: mdl-34326534

ABSTRACT

Transcription factors specialized to limit the destructive potential of inflammatory immune cells remain ill-defined. We discovered loss-of-function variants in the X-linked ETS transcription factor gene ELF4 in multiple unrelated male patients with early onset mucosal autoinflammation and inflammatory bowel disease (IBD) characteristics, including fevers and ulcers that responded to interleukin-1 (IL-1), tumor necrosis factor or IL-12p40 blockade. Using cells from patients and newly generated mouse models, we uncovered ELF4-mutant macrophages having hyperinflammatory responses to a range of innate stimuli. In mouse macrophages, Elf4 both sustained the expression of anti-inflammatory genes, such as Il1rn, and limited the upregulation of inflammation amplifiers, including S100A8, Lcn2, Trem1 and neutrophil chemoattractants. Blockade of Trem1 reversed inflammation and intestine pathology after in vivo lipopolysaccharide challenge in mice carrying patient-derived variants in Elf4. Thus, ELF4 restrains inflammation and protects against mucosal disease, a discovery with broad translational relevance for human inflammatory disorders such as IBD.


Subject(s)
DNA-Binding Proteins/genetics , Hereditary Autoinflammatory Diseases/genetics , Inflammatory Bowel Diseases/genetics , Macrophages/immunology , Transcription Factors/genetics , Animals , Calgranulin A/metabolism , Female , Gene Expression Regulation/genetics , Hereditary Autoinflammatory Diseases/immunology , Hereditary Autoinflammatory Diseases/pathology , Humans , Inflammatory Bowel Diseases/immunology , Inflammatory Bowel Diseases/pathology , Interleukin 1 Receptor Antagonist Protein/immunology , Lipocalin-2/metabolism , Lipopolysaccharides/toxicity , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Th17 Cells/immunology , Transcription, Genetic/genetics , Triggering Receptor Expressed on Myeloid Cells-1/antagonists & inhibitors , Triggering Receptor Expressed on Myeloid Cells-1/metabolism
3.
Nat Immunol ; 18(6): 622-632, 2017 06.
Article in English | MEDLINE | ID: mdl-28459433

ABSTRACT

The high risk of neonatal death from sepsis is thought to result from impaired responses by innate immune cells; however, the clinical observation of hyperinflammatory courses of neonatal sepsis contradicts this concept. Using transcriptomic, epigenetic and immunological approaches, we demonstrated that high amounts of the perinatal alarmins S100A8 and S100A9 specifically altered MyD88-dependent proinflammatory gene programs. S100 programming prevented hyperinflammatory responses without impairing pathogen defense. TRIF-adaptor-dependent regulatory genes remained unaffected by perinatal S100 programming and responded strongly to lipopolysaccharide, but were barely expressed. Steady-state expression of TRIF-dependent genes increased only gradually during the first year of life in human neonates, shifting immune regulation toward the adult phenotype. Disruption of this critical sequence of transient alarmin programming and subsequent reprogramming of regulatory pathways increased the risk of hyperinflammation and sepsis. Collectively these data suggest that neonates are characterized by a selective, transient microbial unresponsiveness that prevents harmful hyperinflammation in the delicate neonate while allowing for sufficient immunological protection.


Subject(s)
Calgranulin A/immunology , Calgranulin B/immunology , Immunity, Innate/immunology , Monocytes/immunology , Neonatal Sepsis/immunology , Adaptor Proteins, Vesicular Transport/genetics , Adaptor Proteins, Vesicular Transport/immunology , Adaptor Proteins, Vesicular Transport/metabolism , Animals , Animals, Newborn , Calgranulin A/drug effects , Calgranulin B/drug effects , Epigenesis, Genetic , Fetal Blood , Flow Cytometry , Gene Expression Profiling , Gene Expression Regulation , Humans , Immunity, Innate/drug effects , Immunoblotting , Infant, Newborn , Inflammation , Lipopolysaccharides/pharmacology , Mice , Mice, Knockout , Monocytes/drug effects , Myeloid Differentiation Factor 88/genetics , Myeloid Differentiation Factor 88/immunology , Neonatal Sepsis/genetics , Real-Time Polymerase Chain Reaction , Toll-Like Receptor 4/immunology
4.
Cell ; 159(3): 623-34, 2014 Oct 23.
Article in English | MEDLINE | ID: mdl-25417112

ABSTRACT

S-nitrosylation is a ubiquitous protein modification emerging as a principal mechanism of nitric oxide (NO)-mediated signal transduction and cell function. S-nitrosylases can use NO synthase (NOS)-derived NO to modify selected cysteines in target proteins. Despite proteomic identification of over a thousand S-nitrosylated proteins, few S-nitrosylases have been identified. Moreover, mechanisms underlying site-selective S-nitrosylation and the potential role of specific sequence motifs remain largely unknown. Here, we describe a stimulus-inducible, heterotrimeric S-nitrosylase complex consisting of inducible NOS (iNOS), S100A8, and S100A9. S100A9 exhibits transnitrosylase activity, shuttling NO from iNOS to the target protein, whereas S100A8 and S100A9 coordinately direct site selection. A family of proteins S-nitrosylated by iNOS-S100A8/A9 were revealed by proteomic analysis. A conserved I/L-X-C-X2-D/E motif was necessary and sufficient for iNOS-S100A8/A9-mediated S-nitrosylation. These results reveal an elusive parallel between protein S-nitrosylation and phosphorylation, namely, stimulus-dependent posttranslational modification of selected targets by primary sequence motif recognition.


Subject(s)
Multiprotein Complexes/metabolism , Nitric Oxide Synthase Type II/metabolism , Nitric Oxide/metabolism , Protein Processing, Post-Translational , Proteins/chemistry , Proteins/metabolism , Amino Acid Motifs , Amino Acid Sequence , Animals , Calgranulin A/metabolism , Calgranulin B/metabolism , Glyceraldehyde-3-Phosphate Dehydrogenases/chemistry , Glyceraldehyde-3-Phosphate Dehydrogenases/metabolism , Humans , Interferon-gamma/metabolism , Lipoproteins, LDL/metabolism , Mice , Models, Molecular , Molecular Sequence Data , Sequence Alignment
5.
Cell ; 150(1): 165-78, 2012 Jul 06.
Article in English | MEDLINE | ID: mdl-22770218

ABSTRACT

Metastasis and chemoresistance in cancer are linked phenomena, but the molecular basis for this link is unknown. We uncovered a network of paracrine signals between carcinoma, myeloid, and endothelial cells that drives both processes in breast cancer. Cancer cells that overexpress CXCL1 and 2 by transcriptional hyperactivation or 4q21 amplification are primed for survival in metastatic sites. CXCL1/2 attract CD11b(+)Gr1(+) myeloid cells into the tumor, which produce chemokines including S100A8/9 that enhance cancer cell survival. Although chemotherapeutic agents kill cancer cells, these treatments trigger a parallel stromal reaction leading to TNF-α production by endothelial and other stromal cells. TNF-α via NF-kB heightens the CXCL1/2 expression in cancer cells, thus amplifying the CXCL1/2-S100A8/9 loop and causing chemoresistance. CXCR2 blockers break this cycle, augmenting the efficacy of chemotherapy against breast tumors and particularly against metastasis. This network of endothelial-carcinoma-myeloid signaling interactions provides a mechanism linking chemoresistance and metastasis, with opportunities for intervention.


Subject(s)
Breast Neoplasms/pathology , Carcinoma/pathology , Chemokine CXCL1/metabolism , Drug Resistance, Neoplasm , Neoplasm Metastasis , Paracrine Communication , Animals , Breast Neoplasms/metabolism , Calgranulin A/metabolism , Calgranulin B/metabolism , Carcinoma/metabolism , Chemokine CXCL1/genetics , Disease Models, Animal , Endothelial Cells/metabolism , Female , Gene Knockdown Techniques , Humans , Lung Neoplasms/secondary , Lymph Nodes/pathology , Lymphatic Metastasis , Mice , Mice, Inbred C57BL , Myeloid Cells/metabolism , Neoplasm Transplantation , Transplantation, Heterologous
6.
J Immunol ; 213(4): 456-468, 2024 Aug 15.
Article in English | MEDLINE | ID: mdl-38912868

ABSTRACT

Neutrophils play important roles in inflammatory airway diseases. In this study, we assessed whether apolipoprotein A-I modifies neutrophil heterogeneity as part of the mechanism by which it attenuates acute airway inflammation. Neutrophilic airway inflammation was induced by daily intranasal administration of LPS plus house dust mite (LPS+HDM) to Apoa1-/- and Apoa1+/+ mice for 3 d. Single-cell RNA sequencing was performed on cells recovered from bronchoalveolar lavage fluid on day 4. Unsupervised profiling identified 10 clusters of neutrophils in bronchoalveolar lavage fluid from Apoa1-/- and Apoa1+/+ mice. LPS+HDM-challenged Apoa1-/- mice had an increased proportion of the Neu4 neutrophil cluster that expressed S100a8, S100a9, and Mmp8 and had high maturation, aggregation, and TLR4 binding scores. There was also an increase in the Neu6 cluster of immature neutrophils, whereas neutrophil clusters expressing IFN-stimulated genes were decreased. An unsupervised trajectory analysis showed that Neu4 represented a distinct lineage in Apoa1-/- mice. LPS+HDM-challenged Apoa1-/- mice also had an increased proportion of recruited airspace macrophages, which was associated with a reciprocal reduction in resident airspace macrophages. Increased expression of a common set of proinflammatory genes, S100a8, S100a9, and Lcn2, was present in all neutrophils and airspace macrophages from LPS+HDM-challenged Apoa1-/- mice. These findings show that Apoa1-/- mice have increases in specific neutrophil and macrophage clusters in the lung during acute inflammation mediated by LPS+HDM, as well as enhanced expression of a common set of proinflammatory genes. This suggests that modifications in neutrophil and macrophage heterogeneity contribute to the mechanism by which apolipoprotein A-I attenuates acute airway inflammation.


Subject(s)
Apolipoprotein A-I , Mice, Knockout , Neutrophils , Pneumonia , Animals , Mice , Neutrophils/immunology , Pneumonia/immunology , Pneumonia/genetics , Apolipoprotein A-I/genetics , Mice, Inbred C57BL , Lipopolysaccharides/immunology , Bronchoalveolar Lavage Fluid/immunology , Bronchoalveolar Lavage Fluid/cytology , Lung/immunology , Lung/pathology , Calgranulin A , Calgranulin B
7.
J Immunol ; 212(10): 1523-1529, 2024 May 15.
Article in English | MEDLINE | ID: mdl-38709994

ABSTRACT

The study of S100A9 in viral infections has seen increased interest since the COVID-19 pandemic. S100A8/A9 levels were found to be correlated with the severity of COVID-19 disease, cytokine storm, and changes in myeloid cell subsets. These data led to the hypothesis that S100A8/A9 proteins might play an active role in COVID-19 pathogenesis. This review explores the structures and functions of S100A8/9 and the current knowledge on the involvement of S100A8/A9 and its constituents in viral infections. The potential roles of S100A9 in SARS-CoV-2 infections are also discussed.


Subject(s)
COVID-19 , Calgranulin A , Calgranulin B , Inflammation , SARS-CoV-2 , Humans , COVID-19/immunology , SARS-CoV-2/immunology , Inflammation/immunology , Cytokine Release Syndrome/immunology , Virus Diseases/immunology
8.
Proc Natl Acad Sci U S A ; 120(43): e2221859120, 2023 10 24.
Article in English | MEDLINE | ID: mdl-37844250

ABSTRACT

Metastatic cancer accounts for 90% of all cancer-related deaths and continues to be one of the toughest challenges in cancer treatment. A growing body of data indicates that S100A9, a major regulator of inflammation, plays a central role in cancer progression and metastasis, particularly in the lungs, where S100A9 forms a premetastatic niche. Thus, we developed a vaccine against S100A9 derived from plant viruses and virus-like particles. Using multiple tumor mouse models, we demonstrate the effectiveness of the S100A9 vaccine candidates in preventing tumor seeding within the lungs and outgrowth of metastatic disease. The elicited antibodies showed high specificity toward S100A9 without cross-reactivity toward S100A8, another member of the S100A family. When tested in metastatic mouse models of breast cancer and melanoma, the vaccines significantly reduced lung tumor nodules after intravenous challenge or postsurgical removal of the primary tumor. Mechanistically, the vaccines reduce the levels of S100A9 within the lungs and sera, thereby increasing the expression of immunostimulatory cytokines with antitumor function [(interleukin) IL-12 and interferonγ] while reducing levels of immunosuppressive cytokines (IL-10 and transforming growth factorß). This also correlated with decreased myeloid-derived suppressor cell populations within the lungs. This work has wide-ranging impact, as S100A9 is overexpressed in multiple cancers and linked with poor prognosis in cancer patients. The data presented lay the foundation for the development of therapies and vaccines targeting S100A9 to prevent metastasis.


Subject(s)
Lung Neoplasms , Viral Vaccines , Humans , Mice , Animals , Calgranulin B/metabolism , Lung Neoplasms/prevention & control , Calgranulin A/metabolism , Lung/pathology , Cytokines/metabolism
9.
PLoS Pathog ; 19(7): e1011493, 2023 07.
Article in English | MEDLINE | ID: mdl-37467233

ABSTRACT

S100A8/A9 has important immunomodulatory roles in antibacterial defense, but its relevance in focal pneumonia caused by Streptococcus pneumoniae (S. pneumoniae) is understudied. We show that S100A9 was significantly increased in BAL fluids of patients with bacterial but not viral pneumonia and correlated with procalcitonin and sequential organ failure assessment scores. Mice deficient in S100A9 exhibited drastically elevated Zn2+ levels in lungs, which led to bacterial outgrowth and significantly reduced survival. In addition, reduced survival of S100A9 KO mice was characterized by excessive release of neutrophil elastase, which resulted in degradation of opsonophagocytically important collectins surfactant proteins A and D. All of these features were attenuated in S. pneumoniae-challenged chimeric WT→S100A9 KO mice. Similarly, therapy of S. pneumoniae-infected S100A9 KO mice with a mutant S100A8/A9 protein showing increased half-life significantly decreased lung bacterial loads and lung injury. Collectively, S100A9 controls central antibacterial immune mechanisms of the lung with essential relevance to survival of pneumococcal pneumonia. Moreover, S100A9 appears to be a promising biomarker to distinguish patients with bacterial from those with viral pneumonia. Trial registration: Clinical Trials register (DRKS00000620).


Subject(s)
Pneumonia, Pneumococcal , Mice , Animals , Calgranulin B/genetics , Calgranulin B/metabolism , Lung , Streptococcus pneumoniae/metabolism , Calgranulin A/genetics , Calgranulin A/metabolism , Bacteria/metabolism , Mice, Knockout
10.
Circ Res ; 132(3): 267-289, 2023 02 03.
Article in English | MEDLINE | ID: mdl-36625265

ABSTRACT

BACKGROUND: The tyrosine kinase inhibitor ponatinib is the only treatment option for chronic myelogenous leukemia patients with T315I (gatekeeper) mutation. Pharmacovigilance analysis of Food and Drug Administration and World Health Organization datasets has revealed that ponatinib is the most cardiotoxic agent among all Food and Drug Administration-approved tyrosine kinase inhibitors in a real-world scenario. However, the mechanism of ponatinib-induced cardiotoxicity is unknown. METHODS: The lack of well-optimized mouse models has hampered the in vivo cardio-oncology studies. Here, we show that cardiovascular comorbidity mouse models evidence a robust cardiac pathological phenotype upon ponatinib treatment. A combination of multiple in vitro and in vivo models was employed to delineate the underlying molecular mechanisms. RESULTS: An unbiased RNA sequencing analysis identified the enrichment of dysregulated inflammatory genes, including a multifold upregulation of alarmins S100A8/A9, as a top hit in ponatinib-treated hearts. Mechanistically, we demonstrate that ponatinib activates the S100A8/A9-TLR4 (Toll-like receptor 4)-NLRP3 (NLR family pyrin domain-containing 3)-IL (interleukin)-1ß signaling pathway in cardiac and systemic myeloid cells, in vitro and in vivo, thereby leading to excessive myocardial and systemic inflammation. Excessive inflammation was central to the cardiac pathology because interventions with broad-spectrum immunosuppressive glucocorticoid dexamethasone or specific inhibitors of NLRP3 (CY-09) or S100A9 (paquinimod) nearly abolished the ponatinib-induced cardiac dysfunction. CONCLUSIONS: Taken together, these findings uncover a novel mechanism of ponatinib-induced cardiac inflammation leading to cardiac dysfunction. From a translational perspective, our results provide critical preclinical data and rationale for a clinical investigation into immunosuppressive interventions for managing ponatinib-induced cardiotoxicity.


Subject(s)
Cardiotoxicity , Heart Diseases , Mice , Animals , NLR Family, Pyrin Domain-Containing 3 Protein/genetics , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Calgranulin A/genetics , Inflammation/chemically induced
11.
Cell Mol Life Sci ; 81(1): 232, 2024 May 23.
Article in English | MEDLINE | ID: mdl-38780644

ABSTRACT

Ubiquitin-proteasome system dysfunction triggers α-synuclein aggregation, a hallmark of neurodegenerative diseases, such as Parkinson's disease (PD). However, the crosstalk between deubiquitinating enzyme (DUBs) and α-synuclein pathology remains unclear. In this study, we observed a decrease in the level of ubiquitin-specific protease 14 (USP14), a DUB, in the cerebrospinal fluid (CSF) of PD patients, particularly females. Moreover, CSF USP14 exhibited a dual correlation with α-synuclein in male and female PD patients. To investigate the impact of USP14 deficiency, we crossed USP14 heterozygous mouse (USP14+/-) with transgenic A53T PD mouse (A53T-Tg) or injected adeno-associated virus (AAV) carrying human α-synuclein (AAV-hα-Syn) in USP14+/- mice. We found that Usp14 deficiency improved the behavioral abnormities and pathological α-synuclein deposition in female A53T-Tg or AAV-hα-Syn mice. Additionally, Usp14 inactivation attenuates the pro-inflammatory response in female AAV-hα-Syn mice, whereas Usp14 inactivation demonstrated opposite effects in male AAV-hα-Syn mice. Mechanistically, the heterodimeric protein S100A8/A9 may be the downstream target of Usp14 deficiency in female mouse models of α-synucleinopathies. Furthermore, upregulated S100A8/A9 was responsible for α-synuclein degradation by autophagy and the suppression of the pro-inflammatory response in microglia after Usp14 knockdown. Consequently, our study suggests that USP14 could serve as a novel therapeutic target in PD.


Subject(s)
Calgranulin A , Calgranulin B , Mice, Transgenic , Parkinson Disease , Ubiquitin Thiolesterase , alpha-Synuclein , Animals , Female , Humans , Male , Mice , alpha-Synuclein/metabolism , alpha-Synuclein/genetics , Calgranulin A/metabolism , Calgranulin A/genetics , Calgranulin B/metabolism , Calgranulin B/genetics , Disease Models, Animal , Mice, Inbred C57BL , Parkinson Disease/metabolism , Parkinson Disease/genetics , Parkinson Disease/pathology , Ubiquitin Thiolesterase/metabolism , Ubiquitin Thiolesterase/genetics , Ubiquitin Thiolesterase/deficiency
12.
Cell Mol Life Sci ; 81(1): 110, 2024 Mar 01.
Article in English | MEDLINE | ID: mdl-38429401

ABSTRACT

Toll-like receptors (TLRs), especially TLR7, play an important role in systemic lupus erythematosus (SLE) pathogenesis. However, the regulatory mechanism underlying the abnormal activation of TLR pathways in patients with SLE has not been elucidated. Notably, accumulating evidence indicates that myeloid-derived suppressor cells (MDSCs) are important regulators of inflammation and autoimmune diseases. Compared with healthy control subjects, patients with SLE have a greater proportion of MDSCs among peripheral blood mononuclear cells (PBMCs); however, the effect of MDSCs on TLR7 pathway activation has not been determined. In the present study, lupus MDSCs significantly promoted TLR7 pathway activation in macrophages and dendritic cells (DCs), exacerbating the imiquimod-induced lupus model. RNA-sequencing analysis revealed significant overexpression of S100 calcium-binding protein A8 (S100A8) and S100A9 in MDSCs from diseased MRL/lpr mice. In vitro and in vivo studies demonstrated that S100A8/9 effectively promoted TLR7 pathway activation and that S100A8/9 deficiency reversed the promoting effect of MDSCs on TLR7 pathway activation in lupus. Mechanistically, MDSC-derived S100A8/9 upregulated interferon gamma (IFN-γ) secretion by macrophages and IFN-γ subsequently promoted TLR7 pathway activation in an autocrine manner. Taken together, these findings suggest that lupus MDSCs promote TLR7 pathway activation and lupus pathogenesis through the S100A8/9-IFN-γ axis. Our study identified an important target for SLE therapy.


Subject(s)
Calgranulin A , Calgranulin B , Lupus Erythematosus, Systemic , Myeloid-Derived Suppressor Cells , Animals , Mice , Dendritic Cells/metabolism , Leukocytes, Mononuclear/metabolism , Lupus Erythematosus, Systemic/metabolism , Lupus Erythematosus, Systemic/pathology , Macrophages/metabolism , Mice, Inbred MRL lpr , Toll-Like Receptor 7/genetics , Toll-Like Receptor 7/metabolism , Calgranulin A/metabolism , Calgranulin B/metabolism
13.
Cell Mol Life Sci ; 81(1): 345, 2024 Aug 12.
Article in English | MEDLINE | ID: mdl-39133305

ABSTRACT

BACKGROUND AND AIMS: Hepatitis B virus (HBV)-associated liver cirrhosis (LC), a common condition with high incidence and mortality rates, is often associated with diabetes mellitus (DM). However, the molecular mechanisms underlying impaired glucose regulation during HBV-associated LC remain unclear. METHODS: Data from 63 patients with LC and 62 patients with LC-associated DM were analysed. Co-culture of NK cells and islet ß cell lines were used to study the glucose regulation mechanism. A mouse model of LC was used to verify the effect of S100A8/A9 on the glucose regulation. RESULTS: Higher levels of interferon (IFN)-γ derived from natural killer (NK) cells and lower levels of insulin emerged in the peripheral blood of patients with both LC and DM compared with those from patients with LC only. IFN-γ derived from NK cells facilitated ß cell necroptosis and impaired insulin production. Furthermore, S100A8/A9 elevation in patients with both LC and DM was found to upregulate IFN-γ production in NK cells. Consistently, in the mouse model for LC, mice treated with carbon tetrachloride (CCL4) and S100A8/A9 exhibited increased blood glucose, impaired insulin production, increased IFN-γ, and increased ß cells necroptosis compared with those treated with CCL4. Mechanistically, S100A8/A9 activated the p38 MAPK pathway to increase IFN-γ production in NK cells. These effects were diminished after blocking RAGE. CONCLUSION: Together, the data indicate that IFN-γ produced by NK cells induces ß cell necroptosis via the S100A8/A9-RAGE-p38 MAPK axis in patients with LC and DM. Reduced levels of S100A8/A9, NK cells, and IFN-γ could be valuable for the treatment of LC with DM. Accumulation of S100A8/A9 in patients with LC may indicate the emergence of DM.


Subject(s)
Calgranulin A , Calgranulin B , Hepatitis B virus , Insulin-Secreting Cells , Interferon-gamma , Killer Cells, Natural , Liver Cirrhosis , Necroptosis , Killer Cells, Natural/immunology , Killer Cells, Natural/metabolism , Humans , Animals , Interferon-gamma/metabolism , Calgranulin B/metabolism , Liver Cirrhosis/pathology , Liver Cirrhosis/metabolism , Liver Cirrhosis/virology , Liver Cirrhosis/immunology , Mice , Male , Insulin-Secreting Cells/metabolism , Insulin-Secreting Cells/pathology , Insulin-Secreting Cells/virology , Calgranulin A/metabolism , Mice, Inbred C57BL , Female , Middle Aged , Hepatitis B/complications , Hepatitis B/pathology , Hepatitis B/metabolism , Disease Models, Animal , Carbon Tetrachloride
14.
Mol Cell Neurosci ; 130: 103957, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39111720

ABSTRACT

BACKGROUND: Microglia is the primary source of inflammatory factors during migraine attacks. This study aims to investigate the role of microglia related genes (MRGs) in migraine attacks. METHODS: The RNA sequencing results of migraineurs and the panglaodb database were used to obtain differentially expressed genes (DEGs) in migraine related to microglia. A migraine rat model was established for validating and localizing of the MRGs, and subsequent screening for target genes was conducted. A shRNA was designed to interference the expression of target genes and administered into the trigeminal ganglion (TG) of rats. Pain sensitivity in rats was evaluated via the hot water tail-flick (HWTF) and formalin-induced pain (FIP) experiments. ELISA was used to quantify the levels of inflammatory cytokines and CGRP. WB and immunofluorescence assays were applied to detect the activation of microglia. RESULTS: A total of five DEGs in migraine related to microglia were obtained from RNA sequencing and panglaodb database. Animal experiments showed that these genes expression were heightened in the TG and medulla oblongata (MO) of migraine rats. The gene S100A8 co-localized with microglia in both TG and MO. The HWTF and FIP experiments demonstrated that interference with S100A8 alleviated the sense of pain in migraine rats. Moreover, the levels of TNFα, IL-1ß, IL-6, and CGRP in the TG and MO of rats in the model rats were increased, and the expression of microglia markers IBA-1, M1 polarization markers CD86 and iNOS was upregulated. Significantly, interference with S100A8 reversed these indicators. CONCLUSION: Interference with S100A8 in microglia increased the pain threshold during migraine attacks, and inhibited neuroinflammation and microglia activation.


Subject(s)
Calgranulin A , Microglia , Migraine Disorders , Neuroinflammatory Diseases , Rats, Sprague-Dawley , Animals , Microglia/metabolism , Migraine Disorders/metabolism , Migraine Disorders/genetics , Rats , Male , Calgranulin A/metabolism , Calgranulin A/genetics , Neuroinflammatory Diseases/metabolism , Trigeminal Ganglion/metabolism , Disease Models, Animal
15.
J Biol Chem ; 299(4): 102952, 2023 04.
Article in English | MEDLINE | ID: mdl-36731796

ABSTRACT

S100A8 and S100A9 are small, human, Ca2+-binding proteins with multiple intracellular and extracellular functions in signaling, regulation, and defense. The two proteins are not detected as monomers but form various noncovalent homo- or hetero-oligomers related to specific activities in human physiology. Because of their significant roles in numerous medical conditions, there has been intense research on the conformational properties of various S100A8 and S100A9 proteoforms as essential targets of drug discovery. NMR or crystal structures are currently available only for mutated or truncated protein complexes, mainly with bound metal ions, that may well reflect the proteins' properties outside cells but not in other biological contexts in which they perform. Here, we used structural mass spectrometry methods combined with molecular dynamics simulations to compare the conformations of wildtype full-length S100A8 and S100A9 subunits in biologically relevant homo- and heterodimers and in higher oligomers formed in the presence of calcium or zinc ions. We provide, first, rationales for their functional response to changing environmental conditions, by elucidating differences between proteoforms in flexible protein regions that may provide the plasticity of the binding sites for the multiple targets, and second, the key factors contributing to the variable stability of the oligomers. The described methods and a systematic view of the conformational properties of S100A8 and S100A9 complexes provide a basis for further research to characterize and modulate their functions for basic science and therapies.


Subject(s)
Calgranulin A , Calgranulin B , Humans , Binding Sites , Calgranulin A/chemistry , Calgranulin B/chemistry , Protein Conformation , Molecular Dynamics Simulation , Mass Spectrometry
16.
Immunology ; 171(3): 365-376, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38013255

ABSTRACT

S100 proteins are small proteins that are only expressed in vertebrates. They are widely expressed in many different cell types and are involved in the regulation of calcium homeostasis, glucose metabolism, cell proliferation, apoptosis, inflammation and tumorigenesis. As members of the S100 protein subfamily of myeloid-related proteins, S100A8, S100A9 and S100A12 play a crucial role in resisting microbial infection and maintaining immune homeostasis. These proteins chelate the necessary metal nutrients of pathogens invading the host by means of 'nutritional immunity' and directly inhibit the growth of pathogens in the host. They interact with receptors on the cell surface to initiate inflammatory signal transduction, induce cytokine expression and participate in the inflammatory response and immune regulation. Furthermore, the increased content of these proteins during the pathological process makes them useful as disease markers for screening and detecting related diseases. This article summarizes the structure and function of the proteins S100A8, S100A9 and S100A12 and lays the foundation for further understanding their roles in infection, immunity and inflammation, as well as their potential applications in the prevention and treatment of infectious diseases.


Subject(s)
Inflammation , S100A12 Protein , Animals , Humans , Calgranulin B , Calgranulin A/metabolism , S100 Proteins/genetics , S100 Proteins/metabolism
17.
Immunology ; 173(2): 381-393, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39003642

ABSTRACT

Among several quantitative trait loci involved in tuberculosis (TB) control in mice, one was mapped within the chromosome 17 segment occupied by the H2 complex and another within the chromosome 3 segment comprising the S100A8/9 genes, which encode neutrophil inflammatory factor S100A8/9. Previously, we developed a panel of H2-congenic mouse strains differing by small segments of the major histocompatibility complex Class II (MHC-II) region from TB-susceptible H2j mice transferred onto the genetic background of the TB-resistant C57BL/6 (H2b) strain. Susceptible B6.I-9.3 mice differ from B6 progenitors by the alleles of their only classical MHC-II H2-Aß gene. The goals of the present study were to: (i) comprehensively characterise the differences in TB-related phenotypes between mice of the two strains and (ii) decipher interactions between the H2-Aß and S100A8/9 genes. Here, we describe the dynamics of TB-related phenotypes differentiating B6.I-9.3 and B6 mice (colony forming units counts, histopathology, lung immune cell infiltration and cytokine profiles). We show that disproportionally diminished CD4+ T-cell population, an enlarged S100A8/9-positive neutrophil population and higher S100A8/9 serum levels in B6.I-9.3 mice collectively form the 'susceptible' phenotype before infection. An increase in IL-17 and a decrease in intrferon-gamma production by CD4+ T-cells in these mice provide a mechanistic explanation of this phenotype. Using F2 segregation analysis, we show that the number of S100A8/9-producing neutrophils in lungs and spleens and the proportion of Th17 CD4+ T-cells in lungs are significantly lower in the presence of the MHC-II dominant 'resistant' b allele compared to the recessive 'susceptible' j/j genotype. This provides direct genetic evidence that MHC-II-regulated CD4+ T-cell landscapes determine neutrophil abundance before infection, an important pathogenic factor in TB immunity.


Subject(s)
Calgranulin A , Calgranulin B , Immunity, Innate , Mice, Inbred C57BL , Neutrophils , Tuberculosis , Animals , Calgranulin A/genetics , Calgranulin A/metabolism , Calgranulin B/genetics , Calgranulin B/metabolism , Neutrophils/immunology , Mice , Tuberculosis/immunology , Tuberculosis/genetics , Adaptive Immunity/genetics , Alleles , Histocompatibility Antigens Class II/genetics , Histocompatibility Antigens Class II/immunology , Histocompatibility Antigens Class II/metabolism , Quantitative Trait Loci , Mycobacterium tuberculosis/immunology , Lung/immunology , Lung/pathology , Cytokines/metabolism , Genetic Predisposition to Disease , Female
18.
Int J Cancer ; 155(11): 2080-2093, 2024 Dec 01.
Article in English | MEDLINE | ID: mdl-39129048

ABSTRACT

Bacteria are ideal anticancer agents and carriers due to their unique capabilities that are convenient in genetic manipulation, tumor-specific targeting, and deep-tissue penetration. However, the specific molecular mechanisms of bacteria-mediated cancer therapy (BMCT) have not been clarified. In this study, we found that TLR4 signaling pathway is critical for Salmonella-mediated tumor targeting, tumor suppression, and liver and spleen protection. TLR4 knockout in mice decreased the levels of cytokines and chemokines, such as S100a8, S100a9, TNF-α, and IL-1ß, in tumor microenvironments (TMEs) after Salmonella treatment, which inhibited tumor cell death and nutrient release, led to reduced bacterial contents in tumors and attenuated antitumor efficacy in a negative feedback manner. Importantly, we found that S100a8 and S100a9 played a leading role in Salmonella-mediated cancer therapy (SMCT). The antitumor efficacy was abrogated and liver damage was prominent when blocked with a specific inhibitor. These findings elucidated the mechanism of Salmonella-mediated tumor targeting, suppression, and host antibacterial defense, providing insights into clinical cancer therapeutics.


Subject(s)
Calgranulin A , Calgranulin B , Lipopolysaccharides , Toll-Like Receptor 4 , Animals , Toll-Like Receptor 4/metabolism , Calgranulin B/metabolism , Calgranulin B/genetics , Calgranulin A/metabolism , Mice , Mice, Knockout , Signal Transduction , Tumor Microenvironment , Humans , Mice, Inbred C57BL , Cell Line, Tumor , Salmonella/metabolism , Neoplasms/microbiology , Neoplasms/metabolism , Neoplasms/immunology , Neoplasms/pathology , Neoplasms/therapy
19.
J Neuroinflammation ; 21(1): 8, 2024 Jan 04.
Article in English | MEDLINE | ID: mdl-38178143

ABSTRACT

Progressive brain diseases create a huge social and economic burden on modern societies as a major cause of disability and death. Incidence of brain diseases has a significantly increasing trend and merits new therapeutic strategies. At the base of many progressive brain malfunctions is a process of unresolved, chronic inflammation. Macrophage migration inhibitory factor, MIF, is an inflammatory mediator that recently gained interest of neuro-researchers due to its varied effects on the CNS such as participation of nervous system development, neuroendocrine functions, and modulation of neuroinflammation. MIF appears to be a candidate as a new biomarker and target of novel therapeutics against numerous neurologic diseases ranging from cancer, autoimmune diseases, vascular diseases, neurodegenerative pathology to psychiatric disorders. In this review, we will focus on MIF's crucial role in neurological diseases such as multiple sclerosis (MS), Alzheimer's disease (AD) and glioblastoma (GBM).


Subject(s)
Brain Diseases , Macrophage Migration-Inhibitory Factors , Multiple Sclerosis , Nervous System Diseases , Humans , Macrophage Migration-Inhibitory Factors/genetics , Inflammation , Calgranulin A , Calgranulin B , Intramolecular Oxidoreductases
20.
Clin Exp Immunol ; 218(1): 93-100, 2024 Sep 16.
Article in English | MEDLINE | ID: mdl-38864482

ABSTRACT

Familial Mediterranean fever (FMF) is characterized by inflammatory attacks due to overactivation of pyrin inflammasome. This study aimed to investigate the reliability of S100A8/A9, neopterin, and matrix metalloproteinase 3 (MMP3) at monitoring subclinical inflammation and disease activity, and at differentiating FMF attacks from appendicitis, the most common misdiagnosis among FMF patients. Blood samples (n = 75), comprising from FMF patients during an attack (n = 20), the same FMF patients during the attack-free period (n = 14), patients with appendicitis (n = 24), and healthy volunteers (n = 17) were obtained. Duplicate determinations of S100A8/A9, neopterin, and MMP-3 levels were conducted using the enzyme-linked immunosorbent assay (ELISA). FMF patients with and without attack and patients with appendicitis had significantly elevated S100A8/A9 levels compared to healthy volunteers (P-values: < 0.001, 0.036, 0.002, respectively). Patients with appendicitis and FMF patients with and without attack had significantly increased serum neopterin levels compared to healthy volunteers (P-value: < 0.001). MMP3 levels were significantly higher among patients with appendicitis and FMF patients during attack compared to healthy controls (P-values: < 0.001, 0.001). Serum levels of S100A8/A9, neopterin, and MMP3 were increased significantly during attacks compared to attack-free periods among FMF patients (P-values: 0.03, 0.047, 0.007). S100A8/A9 emerges as a valuable marker for monitoring disease activity. Neopterin and S100A8/A9 might help physicians to monitor subclinical inflammation during the attack-free periods of FMF patients. MMP3 might aid in diagnosing FMF attacks when distinguishing between attack and attack-free periods is challenging.


Subject(s)
Calgranulin A , Calgranulin B , Familial Mediterranean Fever , Matrix Metalloproteinase 3 , Neopterin , Humans , Familial Mediterranean Fever/blood , Familial Mediterranean Fever/diagnosis , Calgranulin A/blood , Female , Male , Matrix Metalloproteinase 3/blood , Neopterin/blood , Adult , Calgranulin B/blood , Biomarkers/blood , Appendicitis/blood , Appendicitis/diagnosis , Young Adult , Diagnosis, Differential , Middle Aged , Adolescent
SELECTION OF CITATIONS
SEARCH DETAIL