Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 10.930
Filtrer
1.
Bull Exp Biol Med ; 177(1): 68-73, 2024 May.
Article de Anglais | MEDLINE | ID: mdl-38955855

RÉSUMÉ

Substances of silver nanoparticles dialyzed through a 13 kDa membrane, synthesized in a medium of humic ligands modified with hydroquinone and 2-hydroxynaphthoquinone from PowHumus brown coal, specifically enhance the M2 properties of peritoneal macrophages due to inhibition of NO synthase and significant activation of arginase, thus enhancing anti-inflammatory properties of cells. In small, but effective concentrations, they do not have cytotoxic properties and do not contain pyrogenic impurities. The studied humates are able to influence the mechanisms of immune response formation and are an effective means for correcting inflammation and regeneration.


Sujet(s)
Arginase , Arginine , Substances humiques , Macrophages péritonéaux , Argent , Animaux , Macrophages péritonéaux/effets des médicaments et des substances chimiques , Macrophages péritonéaux/métabolisme , Souris , Arginine/pharmacologie , Arginine/composition chimique , Arginase/métabolisme , Argent/composition chimique , Argent/pharmacologie , Nanoparticules métalliques/composition chimique , Hydroquinones/pharmacologie , Hydroquinones/composition chimique , Mâle , Monoxyde d'azote/métabolisme , Nitric oxide synthase/métabolisme , Naphtoquinones/pharmacologie , Naphtoquinones/composition chimique
2.
Chem Biol Drug Des ; 103(6): e14572, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38923686

RÉSUMÉ

The environmental factor aryl hydrocarbon receptor (AhR), a key protein connecting the external environmental signals (e.g., environmental endocrine disruptor TCDD) to internal cellular processes, is involved in the activation of peripheral macrophages and inflammatory response in human body. Thus, there is widespread interest in finding compounds to anti-inflammatory response in macrophages by targeting human AhR. Here, ensemble docking based-virtual screening was first used to screen a library (~200,000 compounds) against human AhR ligand binding domain (LBD) and 25 compounds were identified as potential inhibitors. Then, 9 out of the 25 ligands were found to down-regulate the mRNA expression of CYP1A1 (a downstream gene of AhR signaling) in AhR overexpressing macrophages. The most potent compound AE-411/41415610 was selected for further study and found to reduce both mRNA and protein expressions level of CYP1A1 in mouse peritoneal macrophage. Moreover, protein chip signal pathway analysis indicated that AE-411/41415610 play a role in regulating JAK-STAT and AKT-mTOR pathways. In sum, the discovered hits with novel scaffolds provided a starting point for future design of more effective AhR-targeted lead compounds to regulate CYP1A1 expression of inflammatory peritoneal macrophages.


Sujet(s)
Cytochrome P-450 CYP1A1 , Simulation de docking moléculaire , Récepteurs à hydrocarbure aromatique , Transduction du signal , Récepteurs à hydrocarbure aromatique/métabolisme , Cytochrome P-450 CYP1A1/métabolisme , Cytochrome P-450 CYP1A1/génétique , Animaux , Ligands , Souris , Humains , Transduction du signal/effets des médicaments et des substances chimiques , Macrophages/métabolisme , Macrophages/effets des médicaments et des substances chimiques , Macrophages péritonéaux/métabolisme , Macrophages péritonéaux/effets des médicaments et des substances chimiques , Inflammation/métabolisme , Inflammation/traitement médicamenteux , Protéines proto-oncogènes c-akt/métabolisme , Sérine-thréonine kinases TOR/métabolisme , Sites de fixation
3.
J Surg Res ; 300: 211-220, 2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-38824851

RÉSUMÉ

INTRODUCTION: A dysregulated immune system is a major driver of the mortality and long-term morbidity from sepsis. With respect to macrophages, it has been shown that phenotypic changes are critical to effector function in response to acute infections, including intra-abdominal sepsis. Invariant natural killer T cells (iNKT cells) have emerged as potential central regulators of the immune response to a variety of infectious insults. Specifically, various iNKT cell:macrophage interactions have been noted across a spectrum of diseases, including acute events such as sepsis. However, the potential for iNKT cells to affect peritoneal macrophages during an abdominal septic event is as yet unknown. METHODS: Cecal ligation and puncture (CLP) was performed in both wild type (WT) and invariant natural killer T cell knockout (iNKT-/-) mice. 24 h following CLP or sham operation, peritoneal macrophages were collected for analysis. Analysis of macrophage phenotype and function was undertaken to include analysis of bactericidal activity and cytokine or superoxide production. RESULTS: Within iNKT-/- mice, a greater degree of intraperitoneal macrophages in response to the sepsis was noted. Compared to WT mice, within iNKT-/- mice, CLP did induce an increase in CD86+ and CD206+, but no difference in CD11b+. Unlike WT mice, intra-abdominal sepsis within iNKT-/- mice induced an increase in Ly6C-int (5.2% versus 14.9%; P < 0.05) and a decrease in Ly6C-high on peritoneal macrophages. Unlike phagocytosis, iNKT cells did not affect macrophage bactericidal activity. Although iNKT cells did not affect interleukin-6 production, iNKT cells did affect IL-10 production and both nitrite and superoxide production from peritoneal macrophages. CONCLUSIONS: The observations indicate that iNKT cells affect specific phenotypic and functional aspects of peritoneal macrophages during polymicrobial sepsis. Given that pharmacologic agents that affect iNKT cell functioning are currently in clinical trial, these findings may have the potential for translation to critically ill surgical patients with abdominal sepsis.


Sujet(s)
Macrophages péritonéaux , Souris de lignée C57BL , Souris knockout , Cellules T tueuses naturelles , Sepsie , Animaux , Macrophages péritonéaux/immunologie , Macrophages péritonéaux/métabolisme , Sepsie/immunologie , Sepsie/microbiologie , Cellules T tueuses naturelles/immunologie , Souris , Mâle , Superoxydes/métabolisme , Cytokines/métabolisme , Modèles animaux de maladie humaine
4.
Int Immunopharmacol ; 137: 112470, 2024 Aug 20.
Article de Anglais | MEDLINE | ID: mdl-38908085

RÉSUMÉ

BACKGROUND: The surplus cytokines remaining after use in the early stages of the inflammatory response stimulate immune cells even after the response is over, causing a secondary inflammatory response and ultimately damaging the host, which is called a cytokine storm. Inhibiting heat shock protein 90 (Hsp90), which has recently been shown to play an important role in regulating inflammation in various cell types, may help control excessive inflammatory responses and cytokine storms. METHODS: We discovered an anti-inflammatory compound by measuring the inhibitory effect of CD86 expression on spleen DCs (sDCs) using the chemical compounds library of Hsp90 inhibitors. Subsequently, to select the hit compound, the production of cytokines and expression of surface molecules were measured on the bone marrow-derived DCs (BMDCs) and peritoneal macrophages. Then, we analyzed the response of antigen-specific Th1 cells. Finally, we confirmed the effect of the compound using acute lung injury (ALI) and delayed-type hypersensitivity (DTH) models. RESULTS: We identified Be01 as the hit compound, which reduced CD86 expression the most in sDCs. Treatment with Be01 decreased the production of pro-inflammatory cytokines (IL-6, TNF-α, and IL-1ß) in BMDC and peritoneal macrophages stimulated by LPS. Under the DTH model, Be01 treatment reduced ear swelling and pro-inflammatory cytokines in the spleen. Similarly, Be01 treatment in the ALI model decreased neutrophil infiltration and lower levels of secreted cytokines (IL-6, TNF-α). CONCLUSIONS: Reduction of CD80 and CD86 expression on DCs by Be01 indicates reduced secondary inflammatory response by Th1 cells, and reduced release of pro-inflammatory cytokines by peritoneal macrophages may initially control the cytokine storm.


Sujet(s)
Anti-inflammatoires , Cytokines , Cellules dendritiques , Protéines du choc thermique HSP90 , Macrophages péritonéaux , Souris de lignée C57BL , Animaux , Cellules dendritiques/effets des médicaments et des substances chimiques , Cellules dendritiques/immunologie , Protéines du choc thermique HSP90/antagonistes et inhibiteurs , Protéines du choc thermique HSP90/métabolisme , Cytokines/métabolisme , Anti-inflammatoires/pharmacologie , Anti-inflammatoires/usage thérapeutique , Souris , Macrophages péritonéaux/effets des médicaments et des substances chimiques , Macrophages péritonéaux/immunologie , Macrophages péritonéaux/métabolisme , Hypersensibilité retardée/traitement médicamenteux , Hypersensibilité retardée/immunologie , Antigène CD86/métabolisme , Lésion pulmonaire aigüe/traitement médicamenteux , Lésion pulmonaire aigüe/immunologie , Cellules cultivées , Syndrome de libération de cytokines/traitement médicamenteux , Syndrome de libération de cytokines/immunologie , Lymphocytes auxiliaires Th1/immunologie , Lymphocytes auxiliaires Th1/effets des médicaments et des substances chimiques , Inflammation/traitement médicamenteux , Inflammation/immunologie , Femelle , Modèles animaux de maladie humaine , Rate/immunologie , Rate/effets des médicaments et des substances chimiques
5.
Proc Natl Acad Sci U S A ; 121(27): e2402143121, 2024 Jul 02.
Article de Anglais | MEDLINE | ID: mdl-38923993

RÉSUMÉ

The non-neural cholinergic system plays a critical role in regulating immune equilibrium and tissue homeostasis. While the expression of choline acetyltransferase (ChAT), the enzyme catalyzing acetylcholine biosynthesis, has been well documented in lymphocytes, its role in the myeloid compartment is less understood. Here, we identify a significant population of macrophages (Mϕs) expressing ChAT and synthesizing acetylcholine in the resolution phase of acute peritonitis. Using Chat-GFP reporter mice, we observed marked upregulation of ChAT in monocyte-derived small peritoneal Mϕs (SmPMs) in response to Toll-like receptor agonists and bacterial infections. These SmPMs, phenotypically and transcriptionally distinct from tissue-resident large peritoneal macrophages, up-regulated ChAT expression through a MyD88-dependent pathway involving MAPK signaling. Notably, this process was attenuated by the TRIF-dependent TLR signaling pathway, and our tests with a range of neurotransmitters and cytokines failed to induce a similar response. Functionally, Chat deficiency in Mϕs led to significantly decreased peritoneal acetylcholine levels, reduced efferocytosis of apoptotic neutrophils, and a delayed resolution of peritonitis, which were reversible with exogenous ACh supplementation. Intriguingly, despite B lymphocytes being a notable ChAT-expressing population within the peritoneal cavity, Chat deletion in B cells did not significantly alter the resolution process. Collectively, these findings underscore the crucial role of Mϕ-derived acetylcholine in the resolution of inflammation and highlight the importance of the non-neuronal cholinergic system in immune regulation.


Sujet(s)
Acétylcholine , Choline O-acetyltransferase , Macrophages péritonéaux , Péritonite , Animaux , Choline O-acetyltransferase/métabolisme , Choline O-acetyltransferase/génétique , Péritonite/immunologie , Péritonite/métabolisme , Souris , Macrophages péritonéaux/métabolisme , Macrophages péritonéaux/immunologie , Acétylcholine/métabolisme , Facteur de différenciation myéloïde-88/métabolisme , Facteur de différenciation myéloïde-88/génétique , Souris de lignée C57BL , Transduction du signal , Inflammation/métabolisme , Inflammation/anatomopathologie , Lymphocytes B/immunologie , Lymphocytes B/métabolisme , Récepteurs de type Toll/métabolisme , Phagocytose , Macrophages/métabolisme , Macrophages/immunologie , Souris knockout
6.
Exp Oncol ; 46(1): 30-37, 2024 May 31.
Article de Anglais | MEDLINE | ID: mdl-38852055

RÉSUMÉ

AIM: To study the activity of antitumor immunity effectors and to analyze possible mechanisms of peritoneal Mph M1/M2 repolarization of Balb/c mice under the influence of lectin from B. subtilis IMV B-7724 in the dynamics of the model tumor growth. MATERIALS AND METHODS: Studies were performed on Balb/c mice; Ehrlich adenocarcinoma (АСЕ) was used as an experimental tumor. Lectin from B. subtilis IMV B-7724 was administered to ACE-bearing mice at a dose of 1 mg/kg of body weight, 10 times. Immunological testing was performed on days 21 and 28 after tumor grafting. The functional activity of peritoneal macrophages (Mph), natural killer (NK) cells, cytotoxic lymphocytes (CTL), and cytokine levels (IFN-γ, IL-4) were studied by the standard methods. mRNA expression levels of transcription factors STAT-1, STAT-6, IRF5, and IRF4 in Mph were evaluated. RESULTS: The administration of lectin from B. subtilis IMV B-7724 to mice with solid ACE led to the preservation of the initial functional state of peritoneal Mph M1 during the experiment. The bacterial lectin ensured the preservation of the cytotoxic activity of CD8+ T-lymphocytes and a significant (p < 0.05) increase in the NK activity (by 2.7 times compared to the intact animals and by 12.9 times compared to the untreated mice). A strong positive correlation was noted between the levels of the functional activity of Mph and CD8+ T-lymphocytes of animals with tumors and the indices of the antitumor effectiveness of bacterial lectin. The indirect polarization of Mph was evidenced by a strong positive correlation between the level of the NO/Arg ratio (which characterizes the direction of Mph polarization) and the cytotoxic activity of CD8+ T-lymphocytes, NK cells, and the expression of STAT1/STAT6 (the 21st day) and IRF5/IRF4 (the 28th day). CONCLUSION: In ACE-bearing mice, repolarization of the peritoneal Mph toward M1 can occur not only due to the direct action of bacterial lectin on the cellular receptors but also with the involvement of other effectors of antitumor immunity (NK cells, T-lymphocytes). The transcription factors of the STAT and IRF signaling pathways are involved in the polarization process.


Sujet(s)
Cellules tueuses naturelles , Macrophages péritonéaux , Souris de lignée BALB C , Animaux , Souris , Macrophages péritonéaux/immunologie , Macrophages péritonéaux/métabolisme , Macrophages péritonéaux/effets des médicaments et des substances chimiques , Cellules tueuses naturelles/immunologie , Cellules tueuses naturelles/métabolisme , Carcinome d'Ehrlich/immunologie , Carcinome d'Ehrlich/anatomopathologie , Carcinome d'Ehrlich/métabolisme , Bacillus subtilis , Cytokines/métabolisme , Lymphocytes T cytotoxiques/immunologie , Lymphocytes T cytotoxiques/métabolisme
7.
Front Immunol ; 15: 1426682, 2024.
Article de Anglais | MEDLINE | ID: mdl-38938563

RÉSUMÉ

Background: The disruption of the circadian clock is associated with inflammatory and immunological disorders. BMAL2, a critical circadian protein, forms a dimer with CLOCK, activating transcription. Extracellular cold-inducible RNA-binding protein (eCIRP), released during sepsis, can induce macrophage endotoxin tolerance. We hypothesized that eCIRP induces BMAL2 expression and promotes macrophage endotoxin tolerance through triggering receptor expressed on myeloid cells-1 (TREM-1). Methods: C57BL/6 wild-type (WT) male mice were subjected to sepsis by cecal ligation and puncture (CLP). Serum levels of eCIRP 20 h post-CLP were assessed by ELISA. Peritoneal macrophages (PerM) were treated with recombinant mouse (rm) CIRP (eCIRP) at various doses for 24 h. The cells were then stimulated with LPS for 5 h. The levels of TNF-α and IL-6 in the culture supernatants were assessed by ELISA. PerM were treated with eCIRP for 24 h, and the expression of PD-L1, IL-10, STAT3, TREM-1 and circadian genes such as BMAL2, CRY1, and PER2 was assessed by qPCR. Effect of TREM-1 on eCIRP-induced PerM endotoxin tolerance and PD-L1, IL-10, and STAT3 expression was determined by qPCR using PerM from TREM-1-/- mice. Circadian gene expression profiles in eCIRP-treated macrophages were determined by PCR array and confirmed by qPCR. Induction of BMAL2 activation in bone marrow-derived macrophages was performed by transfection of BMAL2 CRISPR activation plasmid. The interaction of BMAL2 in the PD-L1 promoter was determined by computational modeling and confirmed by the BIAcore assay. Results: Serum levels of eCIRP were increased in septic mice compared to sham mice. Macrophages pre-treated with eCIRP exhibited reduced TNFα and IL-6 release upon LPS challenge, indicating macrophage endotoxin tolerance. Additionally, eCIRP increased the expression of PD-L1, IL-10, and STAT3, markers of immune tolerance. Interestingly, TREM-1 deficiency reversed eCIRP-induced macrophage endotoxin tolerance and significantly decreased PD-L1, IL-10, and STAT3 expression. PCR array screening of circadian clock genes in peritoneal macrophages treated with eCIRP revealed the elevated expression of BMAL2, CRY1, and PER2. In eCIRP-treated macrophages, TREM-1 deficiency prevented the upregulation of these circadian genes. In macrophages, inducible BMAL2 expression correlated with increased PD-L1 expression. In septic human patients, blood monocytes exhibited increased expression of BMAL2 and PD-L1 in comparison to healthy subjects. Computational modeling and BIAcore assay identified a putative binding region of BMAL2 in the PD-L1 promoter, suggesting BMAL2 positively regulates PD-L1 expression in macrophages. Conclusion: eCIRP upregulates BMAL2 expression via TREM-1, leading to macrophage endotoxin tolerance in sepsis. Targeting eCIRP to maintain circadian rhythm may correct endotoxin tolerance and enhance host resistance to bacterial infection.


Sujet(s)
Protéines de liaison à l'ARN , Sepsie , Animaux , Humains , Mâle , Souris , Facteurs de transcription ARNTL/génétique , Modèles animaux de maladie humaine , Endotoxines/immunologie , Tolérance immunitaire , Lipopolysaccharides/immunologie , Macrophages/immunologie , Macrophages/métabolisme , Macrophages péritonéaux/immunologie , Macrophages péritonéaux/métabolisme , Souris de lignée C57BL , Souris knockout , Protéines de liaison à l'ARN/génétique , Protéines de liaison à l'ARN/métabolisme , Sepsie/immunologie , Sepsie/métabolisme , Récepteur de déclenchement de type-1 exprimé sur les cellules myéloïdes/immunologie , Récepteur de déclenchement de type-1 exprimé sur les cellules myéloïdes/génétique , Récepteur de déclenchement de type-1 exprimé sur les cellules myéloïdes/métabolisme
8.
Toxicol Appl Pharmacol ; 487: 116958, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38735591

RÉSUMÉ

Acute lung injury (ALI) remains a significant clinical challenge due to the absence of effective treatment alternatives. This study presents a new method that employs a screening platform focusing on MyD88 affinity, anti-inflammatory properties, and toxicity. This platform was used to evaluate a 300-compound library known for its anti-inflammatory potential. Among the screened compounds, Bicyclol emerged as a standout, exhibiting MyD88 binding and a significant reduction in LPS-stimulated pro-inflammatory factors production in mouse primary peritoneal macrophages. By targeting MyD88, Bicyclol disrupts the MyD88/TLR4 complex and MyD88 polymer formation, thereby mitigating the MAPKs and NF-κB signaling pathways. In vivo experiments further confirmed Bicyclol's efficacy, demonstrating alleviated ALI symptoms, decreased inflammatory cytokines level, and reduced inflammatory cells presence in lung tissues. These findings were associated with a decrease in mortality in LPS-challenged mice. Overall, Bicyclol represents a promising treatment option for ALI by specifically targeting MyD88 and limiting inflammatory responses.


Sujet(s)
Lésion pulmonaire aigüe , Dérivés du biphényle , Lipopolysaccharides , Souris de lignée C57BL , Facteur de différenciation myéloïde-88 , Animaux , Lésion pulmonaire aigüe/induit chimiquement , Lésion pulmonaire aigüe/prévention et contrôle , Lésion pulmonaire aigüe/métabolisme , Lésion pulmonaire aigüe/anatomopathologie , Lipopolysaccharides/toxicité , Facteur de différenciation myéloïde-88/métabolisme , Souris , Mâle , Dérivés du biphényle/pharmacologie , Anti-inflammatoires/pharmacologie , Macrophages péritonéaux/effets des médicaments et des substances chimiques , Macrophages péritonéaux/métabolisme , Facteur de transcription NF-kappa B/métabolisme , Transduction du signal/effets des médicaments et des substances chimiques , Récepteur de type Toll-4/métabolisme , Cytokines/métabolisme , Poumon/effets des médicaments et des substances chimiques , Poumon/anatomopathologie , Poumon/métabolisme
9.
Pancreas ; 53(7): e617-e626, 2024 Aug 01.
Article de Anglais | MEDLINE | ID: mdl-38696351

RÉSUMÉ

BACKGROUND: Although the involvement of intestinal microbiota in innate immunity has been reported recently, the pathogenicity of autoimmune pancreatitis (AIP) remains unclear. This study aimed to investigate whether probiotics ameliorate inflammation in AIP through interactions with innate immunity. MATERIALS AND METHODS: The AIP mouse model was generated by intraperitoneal administration of Escherichia coli to C56BL/6 female mice. Alterations in the intestinal microbiota in the AIP group were evaluated using high-throughput sequencing. Peritoneal macrophages (PMs) were collected and cocultured in vitro with Lactobacillus gasseri (LG) or ligands of Toll-like receptors (TLRs). LG was administered intraperitoneally to AIP model mice, and pancreatitis activity was evaluated to examine the ameliorative effects of LG. RESULTS: In the AIP model mice, inflammation was significantly induced in the pancreas, and the intestinal microbiota was altered with decreased LG. Antimicrobial treatment suppressed pancreatitis. In vitro, E. coli stimulation increased inflammatory cytokine expression, which was significantly decreased when the LG or TLR7 ligand was cocultured with PMs. Intraperitoneal administration of LG to AIP model mice significantly suppressed pancreatitis. CONCLUSION: The mouse model demonstrated the involvement of intestinal microbiota in pancreatitis, and LG administration suppressed pancreatitis, possibly through TLR7 signaling in PMs. LG may be a helpful probiotic for treating AIP.


Sujet(s)
Pancréatite auto-immune , Modèles animaux de maladie humaine , Microbiome gastro-intestinal , Immunité innée , Macrophages péritonéaux , Souris de lignée C57BL , Probiotiques , Animaux , Microbiome gastro-intestinal/immunologie , Probiotiques/administration et posologie , Probiotiques/pharmacologie , Femelle , Pancréatite auto-immune/immunologie , Macrophages péritonéaux/immunologie , Macrophages péritonéaux/microbiologie , Cytokines/métabolisme , Souris , Escherichia coli/immunologie , Récepteur de type Toll-7/immunologie , Pancréas/immunologie , Pancréas/anatomopathologie , Lactobacillus/immunologie
10.
Drug Dev Res ; 85(3): e22194, 2024 May.
Article de Anglais | MEDLINE | ID: mdl-38704828

RÉSUMÉ

The aim the present study was to investigate the impact of novel pentavalent organobismuth and organoantimony complexes on membrane integrity and their interaction with DNA, activity against Sb(III)-sensitive and -resistant Leishmania strains and toxicity in mammalian peritoneal macrophages. Ph3M(L)2 type complexes were synthesized, where M = Sb(V) or Bi(V) and L = deprotonated 3-(dimethylamino)benzoic acid or 2-acetylbenzoic acid. Both organobismuth(V) and organoantimony(V) complexes exhibited efficacy at micromolar concentrations against Leishmania amazonensis and L. infantum but only the later ones demonstrated biocompatibility. Ph3Sb(L1)2 and Ph3Bi(L1)2 demonstrated distinct susceptibility profiles compared to inorganic Sb(III)-resistant strains of MRPA-overexpressing L. amazonensis and AQP1-mutated L. guyanensis. These complexes were able to permeate the cell membrane and interact with the Leishmania DNA, suggesting that this effect may contribute to the parasite growth inhibition via apoptosis. Taken altogether, our data substantiate the notion of a distinct mechanism of uptake pathway and action in Leishmania for these organometallic complexes, distinguishing them from the conventional inorganic antimonial drugs.


Sujet(s)
Antimoine , Antiprotozoaires , Membrane cellulaire , Résistance aux substances , Composés organométalliques , Antimoine/pharmacologie , Antimoine/composition chimique , Animaux , Composés organométalliques/pharmacologie , Souris , Membrane cellulaire/effets des médicaments et des substances chimiques , Antiprotozoaires/pharmacologie , Macrophages péritonéaux/effets des médicaments et des substances chimiques , Macrophages péritonéaux/parasitologie , Leishmania/effets des médicaments et des substances chimiques , ADN des protozoaires , Leishmania infantum/effets des médicaments et des substances chimiques , Leishmania infantum/génétique , Souris de lignée BALB C
11.
Stem Cell Res Ther ; 15(1): 127, 2024 May 01.
Article de Anglais | MEDLINE | ID: mdl-38693589

RÉSUMÉ

BACKGROUND: Endometria are one of the important components of the uterus, which is located in the peritoneal cavity. Endometrial injury usually leads to intrauterine adhesions (IUA), accompanied by inflammation and cell death. We previously reported that both the endometrial ferroptosis was increased and monocytes/macrophages were involved in endometrial injury of IUA. Large peritoneal macrophages (LPMs) are recently reported to migrate into the injured tissues and phagocytose dead cells to repair the tissues. We previously demonstrated that mesenchymal stromal cells (MSCs) had made excellent progress in the repair of endometrial injury. However, it is unclear whether MSCs regulate the LPM efferocytosis against ferroptotic monocytes/macrophages in the injured endometria. METHODS: Here, endometrial injury in IUA mouse model was conducted by uterine curettage and LPS injection surgery and the samples were collected at different times to detect the changes of LPMs and ferroptotic monocytes/macrophages. We conducted LPMs depletion assay in vivo and LPMs and Erastin-induced ferroptotic THP-1 cells coculture systems in vitro to detect the LPM efferocytosis against ferroptotic monocytes/macrophages. The IUA model was treated with MSCs, and their effects on LPMs and endometrial repair were analyzed. Flow cytometry, western blotting, quantitative real-time PCR, immunohistochemical analysis, ELISA, and RNA-sequencing were performed. RESULTS: We found that LPMs migrated to the injured uteri in response to the damage in early phase (3 h), and sustained to a later stage (7 days). Astonishingly, we found that ferroptotic monocytes/macrophages were significantly increased in the injured uteri since 12 h after injury. Moreover, LPMs cocultured with Erastin-induced ferroptotic THP-1 cells in vitro, efferocytosis of LPMs against ferroptotic monocytes/macrophages was emerged. The mRNA expression profiles revealed that LPM efferocytosis against ferroptotic monocytes/macrophages was an induction of glycolysis program and depended on the PPARγ-HK2 pathway. Importantly, we validated that MSCs promoted the efferocytic capability and migration of LPMs to the injured uteri via secreting stanniocalcin-1 (STC-1). CONCLUSION: The data collectively demonstrated first the roles of LPMs via removal of ferroptotic monocytes/macrophages and provided a novel mechanism of MSCs in repairing the endometrial injury.


Sujet(s)
Macrophages péritonéaux , Cellules souches mésenchymateuses , Monocytes , Femelle , Animaux , Souris , Cellules souches mésenchymateuses/métabolisme , Cellules souches mésenchymateuses/cytologie , Monocytes/métabolisme , Monocytes/cytologie , Humains , Macrophages péritonéaux/métabolisme , Endomètre/traumatismes , Endomètre/métabolisme , Endomètre/cytologie , Endomètre/anatomopathologie , Phagocytose , Souris de lignée C57BL , Modèles animaux de maladie humaine ,
12.
Sci Rep ; 14(1): 11079, 2024 05 14.
Article de Anglais | MEDLINE | ID: mdl-38745047

RÉSUMÉ

N-acetyl glucosamine (NAG) is a natural amino sugar found in various human tissues with previously described anti-inflammatory effects. Various chemical modifications of NAG have been made to promote its biomedical applications. In this study, we synthesized two bi-deoxygenated NAG, BNAG1 and BNAG2 and investigated their anti-inflammatory properties, using an in vivo and in vitro inflammation mouse model induced by lipopolysaccharide (LPS). Among the parent molecule NAG, BNAG1 and BNAG2, BNAG1 showed the highest inhibition against serum levels of IL-6 and TNF α and the leukocyte migration to lungs and peritoneal cavity in LPS challenged mice, as well as IL-6 and TNF α production in LPS-stimulated primary peritoneal macrophages. BNAG2 displayed an anti-inflammatory effect which was comparable to NAG. These findings implied potential application of these novel NAG derivatives, especially BNAG1, in treatment of certain inflammation-related diseases.


Sujet(s)
Acétyl-glucosamine , Anti-inflammatoires , Lipopolysaccharides , Macrophages péritonéaux , Facteur de nécrose tumorale alpha , Animaux , Acétyl-glucosamine/pharmacologie , Souris , Anti-inflammatoires/pharmacologie , Anti-inflammatoires/synthèse chimique , Macrophages péritonéaux/effets des médicaments et des substances chimiques , Macrophages péritonéaux/métabolisme , Facteur de nécrose tumorale alpha/métabolisme , Interleukine-6/métabolisme , Interleukine-6/sang , Inflammation/traitement médicamenteux , Mâle , Modèles animaux de maladie humaine
13.
Mol Immunol ; 170: 110-118, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38653076

RÉSUMÉ

Sepsis is a systemic inflammatory response syndrome caused by trauma or infection, which can lead to multiple organ dysfunction. In severe cases, sepsis can also progress to septic shock and even death. Effective treatments for sepsis are still under development. This study aimed to determine if targeting the PI3K/Akt signaling with CAL-101, a PI3K p110δ inhibitor, could alleviate lipopolysaccharide (LPS)-induced sepsis and contribute to immune tolerance. Our findings indicated that CAL-101 treatment improved survival rates and alleviated the progression of LPS-induced sepsis. Compared to antibiotics, CAL-101 not only restored the Th17/regulatory T cells (Treg) balance but also enhanced Treg cell function. Additionally, CAL-101 promoted type 2 macrophage (M2) polarization, inhibited TNF-α secretion, and increased IL-10 secretion. Moreover, CAL-101 treatment reduced pyroptosis in peritoneal macrophages by inhibiting caspase-1/gasdermin D (GSDMD) activation. This study provides a mechanistic basis for future clinical exploration of targeted therapeutics and immunomodulatory strategies in the treatment of sepsis.


Sujet(s)
Phosphatidylinositol 3-kinases de classe I , Souris de lignée C57BL , Pyroptose , Sepsie , Lymphocytes T régulateurs , Cellules Th17 , Animaux , Souris , Phosphatidylinositol 3-kinases de classe I/antagonistes et inhibiteurs , Phosphatidylinositol 3-kinases de classe I/métabolisme , Macrophages/immunologie , Macrophages/effets des médicaments et des substances chimiques , Macrophages/métabolisme , Macrophages péritonéaux/immunologie , Macrophages péritonéaux/effets des médicaments et des substances chimiques , Macrophages péritonéaux/métabolisme , Inhibiteurs des phosphoinositide-3 kinases/pharmacologie , Pyroptose/effets des médicaments et des substances chimiques , Sepsie/immunologie , Sepsie/traitement médicamenteux , Transduction du signal/effets des médicaments et des substances chimiques , Lymphocytes T régulateurs/immunologie , Lymphocytes T régulateurs/effets des médicaments et des substances chimiques , Cellules Th17/immunologie , Cellules Th17/effets des médicaments et des substances chimiques
14.
Int Immunopharmacol ; 133: 112123, 2024 May 30.
Article de Anglais | MEDLINE | ID: mdl-38663314

RÉSUMÉ

The NOD-like receptor family protein 3 (NLRP3) inflammasome is a crucial complex for the host to establish inflammatory immune responses and plays vital roles in a series of disorders, including Alzheimer's disease and acute peritonitis. However, its regulatory mechanism remains largely unclear. Zinc finger antiviral protein (ZAP), also known as zinc finger CCCH-type antiviral protein 1 (ZC3HAV1), promotes viral RNA degradation and plays vital roles in host antiviral immune responses. However, the role of ZAP in inflammation, especially in NLRP3 activation, is unclear. Here, we show that ZAP interacts with NLRP3 and promotes NLRP3 oligomerization, thus facilitating NLRP3 inflammasome activation in peritoneal macrophages of C57BL/6 mice. The shorter isoform of ZAP (ZAPS) appears to play a greater role than the full-length isoform (ZAPL) in HEK293T cells. Congruously, Zap-deficient C57BL/6 mice may be less susceptible to alum-induced peritonitis and lipopolysaccharide-induced sepsis in vivo. Therefore, we propose that ZAP is a positive regulator of NLRP3 activation and a potential therapeutic target for NLRP3-related inflammatory disorders.


Sujet(s)
Inflammasomes , Protéine-3 de la famille des NLR contenant un domaine pyrine , Péritonite , Animaux , Humains , Mâle , Souris , Cellules HEK293 , Inflammasomes/métabolisme , Inflammasomes/immunologie , Lipopolysaccharides/immunologie , Macrophages péritonéaux/immunologie , Macrophages péritonéaux/métabolisme , Souris de lignée C57BL , Souris knockout , Protéine-3 de la famille des NLR contenant un domaine pyrine/métabolisme , Protéine-3 de la famille des NLR contenant un domaine pyrine/génétique , Péritonite/immunologie , Péritonite/induit chimiquement , Multimérisation de protéines , Protéines de liaison à l'ARN/métabolisme , Protéines de liaison à l'ARN/génétique , Sepsie/immunologie , Sepsie/métabolisme
15.
Int Immunopharmacol ; 133: 112153, 2024 May 30.
Article de Anglais | MEDLINE | ID: mdl-38678669

RÉSUMÉ

LPS induced sepsis is a complex process involving various immune cells and signaling molecules. Dysregulation of macrophage polarization and ROS production contributed to the pathogenesis of sepsis. PGP is a transmembrane transporter responsible for the efflux of a number of drugs and also expressed in murine macrophages. Natural products have been shown to decrease inflammation and expression of efflux transporters. However, no treatment is currently available to treat LPS induced sepsis. Verapamil and Tangeretin also reported to attenuate lipopolysaccharide-induced inflammation. However, the effects of verapamil or tangeretin on lipopolysaccharide (LPS)-induced sepsis and its detailed anti-inflammatory mechanism have not been reported. Here, we have determined that verapamil and tangeretin protects against LPS-induced sepsis by suppressing M1 macrophages populations and also through the inhibition of P-glycoprotein expression via downregulating STAT1/STAT3 and upregulating SOCS3 expression in macrophages. An hour before LPS (10 mg/kg) was administered; mice were given intraperitoneal injections of either verapamil (5 mg/kg) or tangeretin (5 mg/kg). The peritoneal macrophages from different experimental groups of mice were isolated. Hepatic, pulmonary and splenic morphometric analyses revealed that verapamil and tangeretin decreased the infiltration of neutrophils into the tissues. Verapamil and tangeritin also enhanced the activity of SOD, CAT, GRX and GSH level in all the tissues tested. verapamil or tangeretin pre-treated mice shifted M1 macrophages to M2 type possibly through the inhibition of P-glycoprotein expression via downregulating STAT1/STAT3 and upregulating SOCS3 expression. Hence, both these drugs have shown protective effects in sepsis via suppressing iNOS, COX-2, oxidative stress and NF-κB signaling in macrophages. Therefore, in our study we can summarize that mice were treated with either Vera or Tan before LPS administration cause an elevated IL-10 by the macrophages which enhances the SOCS3 expression, and thereby able to limits STAT1/STAT3 inter-conversion in the macrophages. As a result, NF-κB activity is also getting down regulated and ultimately mitigating the adverse effect of inflammation caused by LPS in resident macrophages. Whether verapamil or tangeretin offers such protection possibly through the inhibition of P-glycoprotein expression in macrophages needs clarification with the bio availability of these drugs under PGP inhibited conditions is a limitation of this study.


Sujet(s)
Flavones , Lipopolysaccharides , Facteur de transcription STAT-1 , Facteur de transcription STAT-3 , Protéine-3 suppressive de la signalisation des cytokine , Vérapamil , Animaux , Vérapamil/pharmacologie , Facteur de transcription STAT-1/métabolisme , Protéine-3 suppressive de la signalisation des cytokine/métabolisme , Protéine-3 suppressive de la signalisation des cytokine/génétique , Flavones/pharmacologie , Flavones/usage thérapeutique , Souris , Facteur de transcription STAT-3/métabolisme , Mâle , Sepsie/traitement médicamenteux , Sepsie/immunologie , Sepsie/métabolisme , Glycoprotéine P/métabolisme , Anti-inflammatoires/pharmacologie , Anti-inflammatoires/usage thérapeutique , Régulation négative/effets des médicaments et des substances chimiques , Macrophages péritonéaux/effets des médicaments et des substances chimiques , Macrophages péritonéaux/métabolisme , Macrophages péritonéaux/immunologie , Cellules cultivées , Macrophages/effets des médicaments et des substances chimiques , Macrophages/métabolisme , Macrophages/immunologie , Transduction du signal/effets des médicaments et des substances chimiques , Régulation positive/effets des médicaments et des substances chimiques
16.
Theranostics ; 14(6): 2526-2543, 2024.
Article de Anglais | MEDLINE | ID: mdl-38646640

RÉSUMÉ

Rationale: A mature tissue resident macrophage (TRM) population residing in the peritoneal cavity has been known for its unique ability to migrate to peritoneally located injured tissues and impart wound healing properties. Here, we sought to expand on this unique ability of large peritoneal macrophages (LPMs) by investigating whether these GATA6+ LPMs could also intravasate into systemic circulation and migrate to extra-peritoneally located lungs upon ablating lung-resident alveolar macrophages (AMs) by intranasally administered clodronate liposomes in mice. Methods: C12-200 cationic lipidoid-based nanoparticles were employed to selectively deliver a small interfering RNA (siRNA)-targeting CD-45 labeled with a cyanine 5.5 (Cy5.5) dye to LPMs in vivo via intraperitoneal injection. We utilized a non-invasive optical technique called Diffuse In Vivo Flow Cytometry (DiFC) to then systemically track these LPMs in real time and paired it with more conventional techniques like flow cytometry and immunocytochemistry to initially confirm uptake of C12-200 encapsulated siRNA-Cy5.5 (siRNA-Cy5.5 (C12-200)) into LPMs, and further track them from the peritoneal cavity to the lungs in a mouse model of AM depletion incited by intranasally administered clodronate liposomes. Also, we stained for LPM-specific marker zinc-finger transcription factor GATA6 in harvested cells from biofluids like broncho-alveolar lavage as well as whole blood to probe for Cy5.5-labeled LPMs in the lungs as well as in systemic circulation. Results: siRNA-Cy5.5 (C12-200) was robustly taken up by LPMs. Upon depletion of lung-resident AMs, these siRNA-Cy5.5 (C12-200) labeled LPMs rapidly migrated to the lungs via systemic circulation within 12-24 h. DiFC results showed that these LPMs intravasated from the peritoneal cavity and utilized a systemic route of migration. Moreover, immunocytochemical staining of zinc-finger transcription factor GATA6 further confirmed results from DiFC and flow cytometry, confirming the presence of siRNA-Cy5.5 (C12-200)-labeled LPMs in the peritoneum, whole blood and BALF only upon clodronate-administration. Conclusion: Our results indicate for the very first time that selective tropism, migration, and infiltration of LPMs into extra-peritoneally located lungs was dependent on clodronate-mediated AM depletion. These results further open the possibility of therapeutically utilizing LPMs as delivery vehicles to carry nanoparticle-encapsulated oligonucleotide modalities to potentially address inflammatory diseases, infectious diseases and even cancer.


Sujet(s)
Acide clodronique , Poumon , Macrophages péritonéaux , Nanoparticules , Animaux , Acide clodronique/pharmacologie , Acide clodronique/administration et posologie , Nanoparticules/composition chimique , Nanoparticules/administration et posologie , Souris , Poumon/métabolisme , Macrophages péritonéaux/métabolisme , Macrophages péritonéaux/immunologie , Macrophages péritonéaux/effets des médicaments et des substances chimiques , Macrophages alvéolaires/métabolisme , Petit ARN interférent/administration et posologie , Facteur de transcription GATA-6/métabolisme , Liposomes , Souris de lignée C57BL , Carbocyanines/composition chimique , Mouvement cellulaire/effets des médicaments et des substances chimiques , Cytométrie en flux
17.
Molecules ; 29(8)2024 Apr 16.
Article de Anglais | MEDLINE | ID: mdl-38675622

RÉSUMÉ

IRAK4 is a critical mediator in NF-κB-regulated inflammatory signaling and has emerged as a promising therapeutic target for the treatment of autoimmune diseases; however, none of its inhibitors have received FDA approval. In this study, we identified a novel small-molecule IRAK4 kinase inhibitor, DW18134, with an IC50 value of 11.2 nM. DW18134 dose-dependently inhibited the phosphorylation of IRAK4 and IKK in primary peritoneal macrophages and RAW264.7 cells, inhibiting the secretion of TNF-α and IL-6 in both cell lines. The in vivo study demonstrated the efficacy of DW18134, significantly attenuating behavioral scores in an LPS-induced peritonitis model. Mechanistically, DW18134 reduced serum TNF-α and IL-6 levels and attenuated inflammatory tissue injury. By directly blocking IRAK4 activation, DW18134 diminished liver macrophage infiltration and the expression of related inflammatory cytokines in peritonitis mice. Additionally, in the DSS-induced colitis model, DW18134 significantly reduced the disease activity index (DAI) and normalized food and water intake and body weight. Furthermore, DW18134 restored intestinal damage and reduced inflammatory cytokine expression in mice by blocking the IRAK4 signaling pathway. Notably, DW18134 protected DSS-threatened intestinal barrier function by upregulating tight junction gene expression. In conclusion, our findings reported a novel IRAK4 inhibitor, DW18134, as a promising candidate for treating inflammatory diseases, including peritonitis and IBD.


Sujet(s)
Maladies inflammatoires intestinales , Interleukin-1 Receptor-Associated Kinases , Péritonite , Animaux , Interleukin-1 Receptor-Associated Kinases/antagonistes et inhibiteurs , Interleukin-1 Receptor-Associated Kinases/métabolisme , Souris , Péritonite/traitement médicamenteux , Péritonite/induit chimiquement , Cellules RAW 264.7 , Maladies inflammatoires intestinales/traitement médicamenteux , Maladies inflammatoires intestinales/métabolisme , Inhibiteurs de protéines kinases/pharmacologie , Inhibiteurs de protéines kinases/composition chimique , Modèles animaux de maladie humaine , Transduction du signal/effets des médicaments et des substances chimiques , Macrophages péritonéaux/effets des médicaments et des substances chimiques , Macrophages péritonéaux/métabolisme , Humains , Mâle , Phosphorylation/effets des médicaments et des substances chimiques , Cytokines/métabolisme , Facteur de transcription NF-kappa B/métabolisme , Souris de lignée C57BL
18.
Cell Rep ; 43(4): 114096, 2024 Apr 23.
Article de Anglais | MEDLINE | ID: mdl-38607919

RÉSUMÉ

Receptors controlling the cross-presentation of tumor antigens by macrophage subsets in cancer tissues are poorly explored. Here, we show that TIM4+ large peritoneal macrophages efficiently capture and cross-present tumor-associated antigens at early stages of peritoneal infiltration by ovarian cancer cells. The phosphatidylserine (PS) receptor TIM4 promotes maximal uptake of dead cells or PS-coated artificial targets and triggers inflammatory and metabolic gene programs in combination with cytoskeletal remodeling and upregulation of transcriptional signatures related to antigen processing. At the cellular level, TIM4-mediated engulfment induces nucleation of F-actin around nascent phagosomes, delaying the recruitment of vacuolar ATPase, acidification, and cargo degradation. In vivo, TIM4 deletion blunts induction of early anti-tumoral effector CD8 T cells and accelerates the progression of ovarian tumors. We conclude that TIM4-mediated uptake drives the formation of specialized phagosomes that prolong the integrity of ingested antigens and facilitate cross-presentation, contributing to immune surveillance of the peritoneum.


Sujet(s)
Antigènes néoplasiques , Carcinogenèse , Macrophages péritonéaux , Animaux , Macrophages péritonéaux/métabolisme , Macrophages péritonéaux/immunologie , Femelle , Souris , Carcinogenèse/anatomopathologie , Carcinogenèse/immunologie , Carcinogenèse/métabolisme , Humains , Antigènes néoplasiques/métabolisme , Antigènes néoplasiques/immunologie , Tumeurs de l'ovaire/immunologie , Tumeurs de l'ovaire/anatomopathologie , Tumeurs de l'ovaire/métabolisme , Tumeurs de l'ovaire/génétique , Protéines membranaires/métabolisme , Souris de lignée C57BL , Cross-priming/immunologie , Lignée cellulaire tumorale , Phagosomes/métabolisme , Présentation d'antigène/immunologie , Lymphocytes T CD8+/immunologie , Lymphocytes T CD8+/métabolisme , Actines/métabolisme
19.
J Ethnopharmacol ; 331: 118210, 2024 Sep 15.
Article de Anglais | MEDLINE | ID: mdl-38641074

RÉSUMÉ

ETHNOPHARMACOLOGICAL RELEVANCE: Long-term chronic inflammation often leads to chronic diseases. Although Sophora flavescens has been shown to have anti-inflammatory properties, its detailed molecular mechanism is still unknown. AIM OF STUDY: This study investigated the effect of Radix Sophorae Flavescentis on the LPS-induced inflammatory response in macrophages. MATERIALS AND METHODS: LPS was used to induce the peritoneal macrophages to simulate the inflammatory environment in vitro. Different concentrations of Radix Sophorae Flavescentis-containing (medicated) serum were used for intervention. The peritoneal macrophages were identified by using hematoxylin-eosin and immunofluorescence staining. ELISA was used to measure the TNF-α and IL-6 expression to determine the concentration of LPS. ELISA and Western blot (WB) were used to detect the PGE2 and CFHR2 expression in each group, respectively. The lentiviral vector for interference and overexpression of the CFHR2 gene was constructed, packaged, and transfected into LPS-induced macrophages. The transfection efficiency was verified by WB. Then, ELISA was used to detect the TNF-α, PGE2, and IL-6 expression. WB was used to detect the CFHR2, iNOS, COX-2, TLR2, TLR4, IFN-γ, STAT1, and p-STAT1 expression. RESULTS: The primary isolated cells were identified as macrophages. The LPS-treated macrophages exhibited significantly higher expression of PGE2 and CFHR2, and the inflammatory factors TNF-α and IL-6, as well as iNOS, COX-2, TLR2, TLR4, IFN-γ, STAT1, and p-STAT1 expression compared with the control group (P < 0.05). The TNF-α, PGE2, and IL-6 levels, as well as CFHR2, iNOS, COX-2, TLR2, TLR4, IFN-γ, STAT1, and p-STAT1 expression were considerably lower in the LPS-induced+10% medicated-serum group, LPS-induced+20% medicated-serum group, and shCFHR interference group compared with the LPS group (P < 0.05). CONCLUSION: Radix Sophorae Flavescentis might mediate CFHR2 expression and play an important role in inhibiting the LPS-induced pro-inflammatory response of macrophages. Radix Sophorae Flavescentis could be a potential treatment for LPS-induced related inflammatory diseases.


Sujet(s)
Anti-inflammatoires , Lipopolysaccharides , Sophora , Animaux , Sophora/composition chimique , Anti-inflammatoires/pharmacologie , Souris , Macrophages péritonéaux/effets des médicaments et des substances chimiques , Macrophages péritonéaux/métabolisme , Interleukine-6/métabolisme , Interleukine-6/génétique , Facteur de nécrose tumorale alpha/métabolisme , Dinoprostone/métabolisme , Extraits de plantes/pharmacologie , Inflammation/traitement médicamenteux , Inflammation/métabolisme , Inflammation/induit chimiquement , Récepteur de type Toll-2/métabolisme , Récepteur de type Toll-2/génétique , Mâle , Facteur de transcription STAT-1/métabolisme , Racines de plante , Cellules cultivées , Macrophages/effets des médicaments et des substances chimiques , Macrophages/métabolisme , Récepteur de type Toll-4/métabolisme , Récepteur de type Toll-4/génétique , Sophora flavescens
20.
Int J Mol Sci ; 25(7)2024 Apr 08.
Article de Anglais | MEDLINE | ID: mdl-38612926

RÉSUMÉ

A spectrum of immune states resulting from tumor resident macrophages and T-lymphocytes in the solid tumor microenvironment correlates with patient outcomes. We hypothesized that in gastric cancer (GC), macrophages in a polarized immunosuppressive transcriptional state would be prognostic of poor survival. We derived transcriptomic signatures for M2 (M2TS, MRC1; MS4A4A; CD36; CCL13; CCL18; CCL23; SLC38A6; FGL2; FN1; MAF) and M1 (M1TS, CCR7; IL2RA; CXCL11; CCL19; CXCL10; PLA1A; PTX3) macrophages, and cytolytic T-lymphocytes (CTLTS, GZMA; GZMB; GZMH; GZMM; PRF1). Primary GC in a TCGA stomach cancer dataset was evaluated for signature expressions, and a log-rank test determined overall survival (OS) and the disease-free interval (DFI). In 341 TCGA GC entries, high M2TS expression was associated with histological types and later stages. Low M2TS expression was associated with significantly better 5-year OS and DFI. We validated M2TS in prospectively collected peritoneal fluid of a GC patient cohort (n = 28). Single-cell RNA sequencing was used for signature expression in CD68+CD163+ cells and the log-rank test compared OS. GC patients with high M2TS in CD68+CD163+ cells in their peritoneal fluid had significantly worse OS than those with low expression. Multivariate analyses confirmed M2TS was significantly and independently associated with survival. As an independent predictor of poor survival, M2TS may be prognostic in primary tumors and peritoneal fluid of GC patients.


Sujet(s)
Tumeurs de l'estomac , Humains , Tumeurs de l'estomac/génétique , Péritoine , Macrophages péritonéaux , Marqueurs biologiques , Macrophages , Microenvironnement tumoral/génétique , Fibrinogène
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...