Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 3 de 3
Filter
Add more filters











Database
Language
Publication year range
1.
Cell Mol Immunol ; 21(6): 533-545, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38532043

ABSTRACT

The skin is the most common site of Staphylococcus aureus infection, which can lead to various diseases, including invasive and life-threatening infections, through evasion of host defense. However, little is known about the host factors that facilitate the innate immune evasion of S. aureus in the skin. Chemerin, which is abundantly expressed in the skin and can be activated by proteases derived from S. aureus, has both direct bacteria-killing activity and immunomodulatory effects via interactions with its receptor CMKLR1. Here, we demonstrate that a lack of the chemerin/CMKLR1 axis increases the neutrophil-mediated host defense against S. aureus in a mouse model of cutaneous infection, whereas chemerin overexpression, which mimics high levels of chemerin in obese individuals, exacerbates S. aureus cutaneous infection. Mechanistically, we identified keratinocytes that express CMKLR1 as the main target of chemerin to suppress S. aureus-induced IL-33 expression, leading to impaired skin neutrophilia and bacterial clearance. CMKLR1 signaling specifically inhibits IL-33 expression induced by cell wall components but not secreted proteins of S. aureus by inhibiting Akt activation in mouse keratinocytes. Thus, our study revealed that the immunomodulatory effect of the chemerin/CMKLR1 axis mediates innate immune evasion of S. aureus in vivo and likely increases susceptibility to S. aureus infection in obese individuals.


Subject(s)
Chemokines , Immunity, Innate , Intercellular Signaling Peptides and Proteins , Keratinocytes , Receptors, Chemokine , Staphylococcus aureus , Animals , Keratinocytes/immunology , Keratinocytes/metabolism , Staphylococcus aureus/immunology , Chemokines/metabolism , Receptors, Chemokine/metabolism , Mice , Intercellular Signaling Peptides and Proteins/metabolism , Mice, Inbred C57BL , Humans , Signal Transduction , Staphylococcal Skin Infections/immunology , Staphylococcal Skin Infections/pathology , Staphylococcal Infections/immunology , Neutrophils/immunology , Neutrophils/metabolism , Skin/immunology , Skin/pathology , Skin/microbiology , Mice, Knockout
2.
Proc Natl Acad Sci U S A ; 119(29): e2205574119, 2022 07 19.
Article in English | MEDLINE | ID: mdl-35858331

ABSTRACT

Intestinal barrier immunity is essential for controlling gut microbiota without eliciting harmful immune responses, while its defect contributes to the breakdown of intestinal homeostasis and colitis development. Chemerin, which is abundantly expressed in barrier tissues, has been demonstrated to regulate tissue inflammation via CMKLR1, its functional receptor. Several studies have reported the association between increased expression of chemerin-CMKLR1 and disease severity and immunotherapy resistance in inflammatory bowel disease (IBD) patients. However, the pathophysiological role of endogenous chemerin-CMKLR1 signaling in intestinal homeostasis remains elusive. We herein demonstrated that deficiency of chemerin or intestinal epithelial cell (IEC)-specific CMKLR1 conferred high susceptibility to microbiota-driven neutrophilic colon inflammation and subsequent tumorigenesis in mice following epithelial injury. Unexpectedly, we found that lack of chemerin-CMKLR1 signaling specifically reduced expression of lactoperoxidase (LPO), a peroxidase that is predominantly expressed in colonic ECs and utilizes H2O2 to oxidize thiocyanates to the antibiotic compound, thereby leading to the outgrowth and mucosal invasion of gram-negative bacteria and dysregulated CXCL1/2-mediated neutrophilia. Importantly, decreased LPO expression was causally linked to aggravated microbiota-driven colitis and associated tumorigenesis, as LPO supplementation could completely rescue such phenotypes in mice deficient in epithelial chemerin-CMKLR1 signaling. Moreover, epithelial chemerin-CMKLR1 signaling is necessary for early host defense against bacterial infection in an LPO-dependent manner. Collectively, our study reveals that the chemerin-CMKLR1/LPO axis represents an unrecognized immune mechanism that potentiates epithelial antimicrobial defense and restricts harmful colonic neutrophilia and suggests that LPO supplementation may be beneficial for microbiota dysbiosis in IBD patients with a defective innate antimicrobial mechanism.


Subject(s)
Carcinogenesis , Chemokines , Colitis , Colon , Gastrointestinal Microbiome , Intercellular Signaling Peptides and Proteins , Lactoperoxidase , Receptors, Chemokine , Animals , Carcinogenesis/immunology , Cell Transformation, Neoplastic , Chemokines/genetics , Chemokines/metabolism , Colitis/immunology , Colitis/microbiology , Colon/immunology , Colon/microbiology , Hydrogen Peroxide/metabolism , Intercellular Signaling Peptides and Proteins/genetics , Intercellular Signaling Peptides and Proteins/metabolism , Intestinal Mucosa/metabolism , Intestinal Mucosa/microbiology , Lactoperoxidase/metabolism , Mice , Neutrophils/immunology , Receptors, Chemokine/genetics , Receptors, Chemokine/metabolism
3.
Peptides ; 135: 170422, 2021 01.
Article in English | MEDLINE | ID: mdl-33144092

ABSTRACT

Nonalcoholic steatohepatitis (NASH) is a global public health challenge. Overwhelmed oxidative stress and impaired autophagy play an important role in the progression of NASH. Chemerin is an adipokine that has attracted much attention in inflammation and metabolic diseases. This study aimed to examine the effects of chemerin in NASH and its association with oxidative stress and autophagy. In this study, chemerin was found to significantly ameliorate high-fat diet (HFD) induced NASH, marked by decreased serum levels of alanine aminotransferase (ALT), aspartate aminotransferase (AST), interleukin (IL)-1ß, IL-6, and tumor necrosis factor-α (TNF-α), decreased insulin resistance (IR) and leptin resistance (LR), and improved liver lesions. Besides, chemerin prevented enhanced oxidative stress in NASH mice by regulating the antioxidant defense system (MDA downregulation and upregulation of superoxide dismutase (SOD)). Moreover, chemerin contributed to the alleviation of NASH through autophagy activation (p62 downregulation, and upregulation of beclin-1 and LC3). Furthermore, these effects were related to increased phosphorylation of JAK2-STAT3 stimulated by chemerin, which could be inhibited by the CMKLR1 specific inhibitor α-NETA. In conclusion, excess chemerin highly probably ameliorated NASH by alleviating oxidative stress and promoting autophagy, the mechanism responsible for this process was related, at least in part, to the increased phosphorylation of JAK2-STAT3 stimulated by chemerin/CMKLR1. Rh-chemerin may represent promising therapeutic targets in the treatment of NASH.


Subject(s)
Autophagy/genetics , Chemokines/genetics , Intercellular Signaling Peptides and Proteins/genetics , Janus Kinase 2/genetics , Non-alcoholic Fatty Liver Disease/genetics , STAT3 Transcription Factor/genetics , Animals , Diet, High-Fat/adverse effects , Disease Models, Animal , Humans , Insulin Resistance/genetics , Leptin/metabolism , Liver/metabolism , Liver/pathology , Mice , Non-alcoholic Fatty Liver Disease/etiology , Non-alcoholic Fatty Liver Disease/metabolism , Non-alcoholic Fatty Liver Disease/pathology , Oxidative Stress/genetics , Signal Transduction/genetics
SELECTION OF CITATIONS
SEARCH DETAIL