Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 1.686
Filtrer
1.
Cancer Rep (Hoboken) ; 7(9): e2161, 2024 Sep.
Article de Anglais | MEDLINE | ID: mdl-39307915

RÉSUMÉ

BACKGROUND: Hepatocellular carcinoma (HCC) is a prevalent malignancy with a high mortality rate worldwide. Suppressor of cytokine signaling (SOCS) family members play important roles in the proliferation, metabolism, and immunity of HCC cells by regulating cytokines and growth factors. However, it remains uncertain whether the level of SOCS family members can affect the prognosis of HCC patients. AIMS: This study aimed to comprehensively assess the role and mechanisms of SOCS family members in the development of HCC and to guide clinical selection. METHODS: We investigated the expression levels of SOCS family genes in HCC patients and their associations with various clinicopathological characteristics. We also utilized a public database to analyze the changes in the expression, potential functions, transcription factors, and immune invasion of SOCS family members. Additionally, we examined the prognostic value of the SOC family for HCC and its correlation with the SOC family and ferroptosis-related genes. RESULTS: This study revealed that the expression of SOCS2-7, and CISH was downregulated in HCC. The SOCS4, SOCS5, and SOCS7 genes were associated with the clinicopathological features of HCC patients. SOCS family genes are mainly related to the PIK3R3, GHR, and TNS4 pathways. Additionally, this study revealed that STAT3, PPAR-gamma 2, and IRF-2 are important transcription factors that regulate SOCS family members. The expression levels of SOCS family members are closely related to immune infiltration in liver cancer. The study also indicated that SOCS2 and SOCS4 are risk-related genes for predicting the prognosis of patients with liver cancer. Finally, this study suggested that the SOCS2 gene may be involved in the development and progression of HCC. CONCLUSION: Our study enhances the current understanding of SOCS gene function in liver cancer and can help clinicians select appropriate drugs and predict the prognosis of HCC patients.


Sujet(s)
Carcinome hépatocellulaire , Régulation de l'expression des gènes tumoraux , Tumeurs du foie , Protéines SOCS , Humains , Tumeurs du foie/génétique , Tumeurs du foie/immunologie , Tumeurs du foie/mortalité , Tumeurs du foie/anatomopathologie , Protéines SOCS/génétique , Pronostic , Carcinome hépatocellulaire/génétique , Carcinome hépatocellulaire/immunologie , Carcinome hépatocellulaire/mortalité , Carcinome hépatocellulaire/anatomopathologie , Marqueurs biologiques tumoraux/génétique , Marqueurs biologiques tumoraux/métabolisme , Mâle , Femelle , Ferroptose/génétique , Adulte d'âge moyen , Analyse de profil d'expression de gènes , Multi-omique
2.
Trop Anim Health Prod ; 56(8): 272, 2024 Sep 23.
Article de Anglais | MEDLINE | ID: mdl-39312078

RÉSUMÉ

BACKGROUND: Livestock farmers face financial losses every year because milk yield and components are severely affected by udder diseases. These udder infections attract the immune response from the host and lead to the influx of neutrophils into milk to fight infection and thus the number of somatic cell count (SCC) is increased. The SCC value of milk could be used as an important indicator in detecting clinical mastitis in dairy animals. Also, the milk yield and milk quality (e.g. fat) are negatively affected by the increased SCC. The SCC is used to estimate the somatic cell score (SCS)of the milk, which is used as an indirect measure to detect subclinical mastitis. Therefore, the purpose of this study was to investigate the presence of a significant SNP rs868996547, on the suppressor of cytokine signaling 2 gene (SOCS2) which is related to milk yield and milk quality in Awassi sheep. METHODS: In this study, milk production data was obtained from 210 healthy Awassi ewes with different parties and ages. The general linear model (GLM) process analysis of variance (ANOVA) was used to determine fixed effects on milk traits. The DNA extraction was done using a blood DNA extraction kit from Qiagen. To validate the presence of SNP a customized SNP detection developed by Thermofisher Scientific was used. The presence of the SNP in the SOCS2 gene was detected with genotypes (C/T, T/T, and C/C) and T being the mutated allele and it had a significant (p < 0.015) effect on the milk yield (p < 0,015;0.091), fat (p < 0,001;0,003), fat/protein ratio (p < 0.001;0,037) and log10SCC value (p < 0,006;0,015) of Awassi ewes. However, the protein, total solid, and lactose percentages in the wild type and the mutated ewes found having no significant difference (P > 0.05). CONCLUSION: Our result showed the increase in SCC or SCS of the milk significantly affected the milk yield and composition. Parity and age had significant effects on ewes' milk yield (p < 0.001). In conclusion, we investigated the presence of SOCS2 gene of Awassi ewes in the study flock and its effect on milk yield, fat, and somatic cell count, and the change in milk composition and milk yield because of SCC.


Sujet(s)
Lait , Polymorphisme de nucléotide simple , Protéines SOCS , Animaux , Lait/composition chimique , Lait/métabolisme , Lait/cytologie , Femelle , Numération cellulaire/médecine vétérinaire , Protéines SOCS/génétique , Lactation , Protéines de lait/analyse , Ovis aries/génétique , Matières grasses/analyse , Matières grasses/métabolisme
3.
Biomolecules ; 14(9)2024 Aug 26.
Article de Anglais | MEDLINE | ID: mdl-39334830

RÉSUMÉ

The suppressor of cytokine signaling (SOCS) family of proteins were named after their defining role as negative feedback regulators of signaling initiated by numerous cytokine receptors. However, multiple members of the SOCS family likely function outside of this paradigm, including SOCS4. Zebrafish possess two SOCS4 paralogues, with socs4a previously shown to participate in central nervous system development and function. This study examined the role of the other paralogue, socs4b, through expression analysis and functional investigations in vivo and in vitro. This revealed maternal deposition of socs4b mRNA, specific zygotic expression during late embryogenesis, including in the brain, eye and intestine, and broad adult expression that was highest in the brain. A mutant allele, socs4bΔ18, was generated by genome editing, in which the start codon was deleted. Fish homozygous for this likely hypomorphic allele showed no overt developmental phenotypes. However, in vitro studies suggested the Socs4b protein may be able to regulate EGFR signaling.


Sujet(s)
Récepteurs ErbB , Transduction du signal , Protéines SOCS , Protéines de poisson-zèbre , Danio zébré , Animaux , Danio zébré/génétique , Danio zébré/métabolisme , Protéines SOCS/métabolisme , Protéines SOCS/génétique , Protéines de poisson-zèbre/génétique , Protéines de poisson-zèbre/métabolisme , Récepteurs ErbB/métabolisme , Récepteurs ErbB/génétique , Régulation de l'expression des gènes au cours du développement
4.
Int J Mol Sci ; 25(16)2024 Aug 20.
Article de Anglais | MEDLINE | ID: mdl-39201703

RÉSUMÉ

Preeclampsia is a complex pregnancy-related hypertensive disorder which poses significant risks for both maternal and fetal health. Preeclampsia affects 5-8% of pregnancies in the United States, causing a significant public health and economic burden. Despite extensive research, the etiology and pathogenesis of preeclampsia remain elusive, but have been correlated with maternal conditions such as obesity. In recent decades, the incidence of preeclampsia increased along with the prevalence of obesity among women of reproductive age. Maternal obesity has been shown to negatively affect pregnancy in almost all aspects. However, the precise mechanisms by which obesity influences preeclampsia are unclear. Ankyrin repeat and SOCS Box Containing protein 4 (ASB4) is an E3 ubiquitin ligase that can promote the degradation of a wide range of target proteins. ASB4-null mice display a full spectrum of preeclampsia-like phenotypes during pregnancy including hypertension, proteinuria, and decreased litter size. Furthermore, maternal obesity induced by a high-fat diet aggravates preeclampsia-like phenotypes in pregnant mice lacking ASB4. Variants in the ASB4 gene have been associated with obesity in humans, and a functional connection between the ASB4 gene and obesity has been established in mice. This review discusses the connections between preeclampsia, obesity, and ASB4.


Sujet(s)
Obésité , Pré-éclampsie , Protéines SOCS , Animaux , Femelle , Humains , Souris , Grossesse , Obésité/métabolisme , Obésité/génétique , Obésité/étiologie , Pré-éclampsie/étiologie , Pré-éclampsie/métabolisme , Pré-éclampsie/génétique , Protéines SOCS/génétique , Protéines SOCS/métabolisme
5.
Mol Immunol ; 174: 18-31, 2024 Oct.
Article de Anglais | MEDLINE | ID: mdl-39142007

RÉSUMÉ

PURPOSE: Nonalcoholic steatohepatitis (NASH) has been an increasingly significant contributor to hepatocellular carcinoma (HCC). Understanding the progression from NASH to HCC is critical to early diagnosis and elucidating the underlying mechanisms. RESULTS: 5 significant prognostic genes related to NASH-HCC transformation were identified through algorithm selection, which were ME1, TP53I3, SOCS2, GADD45G and CYP7A1. A diagnostic model for NASH prediction was established (AUC=0.988). TP53I3 and SOCS2 were selected as potential critical genes in the progression of NASH-HCC by external dataset validation and in vitro experiments on NASH and HCC cell lines. Immune infiltration analysis illustrated the correlation between 5 significant prognostic genes and immune cells. Single-cell analysis identified hepatocytes related to NASH-HCC transformation markers, revealing their promoting role in the transformation from NASH to HCC. CONCLUSION: With bulk-seq analysis and single-cell analysis, 5 significant prognostic genes related to NASH-HCC transformation were identified and validated at both dataset and in vitro experiment level. Among them, TP53I3 and SOCS2 might be potential critical genes in NASH-HCC progression. Single-cell analysis identified and revealed the critical role that NASH-HCC related hepatocytes play in NASH-HCC tansformation. Our research may introduce a new perspective to the diagnosis, treatment of NASH-related HCC.


Sujet(s)
Carcinome hépatocellulaire , Biologie informatique , Tumeurs du foie , Apprentissage machine , Stéatose hépatique non alcoolique , Humains , Carcinome hépatocellulaire/génétique , Tumeurs du foie/génétique , Tumeurs du foie/anatomopathologie , Stéatose hépatique non alcoolique/génétique , Biologie informatique/méthodes , Pronostic , Transformation cellulaire néoplasique/génétique , Évolution de la maladie , Régulation de l'expression des gènes tumoraux , Protéines SOCS/génétique , Lignée cellulaire tumorale , Analyse sur cellule unique/méthodes
6.
mBio ; 15(9): e0204324, 2024 Sep 11.
Article de Anglais | MEDLINE | ID: mdl-39162488

RÉSUMÉ

E3 ubiquitin ligase (E3) plays a vital role in regulating inflammatory responses by mediating ubiquitination. Previous studies have shown that ankyrin repeat and SOCS box-containing protein 3 (ASB3) is involved in immunomodulatory functions associated with cancer. However, the impact of ASB3 on the dynamic interplay of microbiota and inflammatory responses in inflammatory bowel disease (IBD) is unclear. Here, we systematically identify the E3 ligase ASB3 as a facilitative regulator in the development and progression of IBD. We observed that ASB3 exhibited significant upregulation in the lesions of patients with IBD. ASB3-/- mice are resistant to dextran sodium sulfate-induced colitis. IκBα phosphorylation levels and production of proinflammatory factors IL-1ß, IL-6, and TNF-α were reduced in the colonic tissues of ASB3-/- mice compared to WT mice. This colitis-resistant phenotype was suppressed after coprophagic microbial transfer and reversed after combined antibiotics removed the gut commensal microbiome. Mechanistically, ASB3 specifically catalyzes K48-linked polyubiquitination of TRAF6 in intestinal epithelial cells. In contrast, in ASB3-deficient organoids, the integrity of the TRAF6 protein is shielded, consequently decelerating the onset of intestinal inflammation. ASB3 is associated with dysregulation of the colitis microbiota and promotes proinflammatory factors' production by disrupting TRAF6 stability. Strategies to limit the protein level of ASB3 in intestinal epithelial cells may help in the treatment of colitis. IMPORTANCE: Ubiquitination is a key process that controls protein stability. We determined the ubiquitination of TRAF6 by ASB3 in intestinal epithelial cells during colonic inflammation. Inflammatory bowel disease patients exhibit upregulated ASB3 expression at focal sites, supporting the involvement of degradation of TRAF6, which promotes TLR-Myd88/TRIF-independent NF-κB aberrant activation and intestinal microbiota imbalance. Sustained inflammatory signaling in intestinal epithelial cells and dysregulated protective probiotic immune responses mediated by ASB3 collectively contribute to the exacerbation of inflammatory bowel disease. These findings provide insights into the pathogenesis of inflammatory bowel disease and suggest a novel mechanism by which ASB3 increases the risk of colitis. Our results suggest that future inhibition of ASB3 in intestinal epithelial cells may be a novel clinical strategy.


Sujet(s)
Microbiome gastro-intestinal , Maladies inflammatoires intestinales , Souris knockout , Facteur-6 associé aux récepteurs de TNF , Animaux , Humains , Souris , Colite/microbiologie , Colite/induit chimiquement , Colite/génétique , Colite/métabolisme , Modèles animaux de maladie humaine , Maladies inflammatoires intestinales/microbiologie , Maladies inflammatoires intestinales/métabolisme , Maladies inflammatoires intestinales/génétique , Maladies inflammatoires intestinales/immunologie , Muqueuse intestinale/métabolisme , Muqueuse intestinale/microbiologie , Muqueuse intestinale/immunologie , Souris de lignée C57BL , Stabilité protéique , Protéines SOCS/génétique , Protéines SOCS/métabolisme , Facteur-6 associé aux récepteurs de TNF/métabolisme , Facteur-6 associé aux récepteurs de TNF/génétique , Ubiquitination
7.
Front Immunol ; 15: 1397330, 2024.
Article de Anglais | MEDLINE | ID: mdl-39185412

RÉSUMÉ

Introduction: The role of suppressor of cytokine signaling (SOCS)2 in anti-infective bacterial immunity has been poorly investigated compared to other members of the SOCS family. Methods: We characterized the previously identified loss of function R96C point mutation of SOCS2 using a genome-edited mouse model that resumes the phenotype of Socs2 knockout mice. The response of macrophages to TLR-ligands and Staphylococcus aureus was examined. Results and discussion: Conversely to previously published data using human monocyte-derived macrophages, the stimulation of bone-marrow-derived macrophages with various TLR ligands did not show any difference according to the SOCS2 variant. Upregulation of IL-6 and TNF-α pro-inflammatory cytokines production was only seen when the SOCS2 expression was promoted by the culture of macrophages in the presence of GM-CSF. Furthermore, we showed that the SOCS2 point mutation is associated with heightened STAT5 phosphorylation in a short time frame upon GM-CSF incubation. In mice, recruitment of neutrophil and F4/80int Ly6C+ inflammatory macrophage, as well as IFN-γ and IL-10 concentrations, are significantly increased upon S. aureus peritoneal infection. Altogether, these data support the idea that by lowering the pro-inflammatory environment, SOCS2 favors better control of bacterial burden during a systemic infection caused by S. aureus.


Sujet(s)
Macrophages , Souris knockout , Infections à staphylocoques , Staphylococcus aureus , Protéines SOCS , Animaux , Staphylococcus aureus/immunologie , Souris , Protéines SOCS/génétique , Protéines SOCS/métabolisme , Macrophages/immunologie , Macrophages/métabolisme , Infections à staphylocoques/immunologie , Infections à staphylocoques/génétique , Récepteurs de type Toll/métabolisme , Récepteurs de type Toll/génétique , Cytokines/métabolisme , Mutation perte de fonction , Inflammation/immunologie , Inflammation/génétique , Souris de lignée C57BL , Ligands , Humains
8.
Pathol Res Pract ; 262: 155558, 2024 Oct.
Article de Anglais | MEDLINE | ID: mdl-39213689

RÉSUMÉ

Different types of cytokines, growth factors, or hormones present within the tumor microenvironment that can activate the JAK-STAT signaling pathway by binding to their specific cell surface receptors. The constitutive activation of the JAK-STAT pathway can promote uncontrolled cell proliferation and prevent apoptosis contributing to tumor development. Activation of the JAK-STAT pathway is controlled by several regulatory molecules, particularly the suppressor of cytokine signaling (SOCS) family consisting of eight members, which include SOCS1-SOCS7 and the cytokine-inducible SH2-containing (CIS) proteins. In prostate cancer cells, the irregular expression of the SOCS1-SOCS3, SOCS5-SOCS7 as well as CIS can similarly and differentially result in the initiation of various cellular signaling pathways (in particular JAK-STAT3, MAPK, ERK) that promote cell proliferation, migration, invasion and viability; cell cycle progression; epithelial-mesenchymal transition; angiogenesis; resistance to therapy; immune evasion; and chronic inflammation within the tumor microenvironment which lead to tumor progression, metastasis and poor prognosis. Epigenetic modifications, mainly due to DNA methylation, microRNAs, pro-inflammatory cytokines, growth factors and androgens can influence the expression of the SOCS molecules in prostate cancer cells. Using strategies to modulate, restore or enhance the expression of SOCS proteins, may help overcome treatment resistance and improve the efficacy of existing therapies. In this review, we provide a comprehensive explanation regarding SOCS dysregulation in prostate cancer to provide insights into the mechanisms underlying the dysregulation of SOCS proteins. This knowledge may pave the way for the development of novel therapeutic strategies to manage prostate cancer by restoring and modulating the expression of SOCS molecules.


Sujet(s)
Tumeurs de la prostate , Transduction du signal , Protéines SOCS , Humains , Mâle , Tumeurs de la prostate/anatomopathologie , Tumeurs de la prostate/métabolisme , Tumeurs de la prostate/génétique , Protéines SOCS/métabolisme , Protéines SOCS/génétique , Transduction du signal/physiologie , Régulation de l'expression des gènes tumoraux , Microenvironnement tumoral
9.
Biochim Biophys Acta Mol Cell Res ; 1871(7): 119804, 2024 Oct.
Article de Anglais | MEDLINE | ID: mdl-39084528

RÉSUMÉ

BACKGROUND: Myocardial fibrosis is an important pathological feature of dilated cardiomyopathy (DCM). The roles of SOCS2 in fibrosis of different organs are controversial. Herein, we investigated the function and potential mechanism of SOCS2 in myocardial fibrosis. METHODS: Bioinformatics, immunohistochemistry (IHC), immunofluorescence (IF), western blot (WB), real-time fluorescence quantitative PCR (qPCR), rat primary myocardial fibroblasts (rCFs) culture, doxorubicin (DOX) induced mouse dilated cardiomyopathy (DCM) model, and in vivo adeno-associated virus (AAV) infection were used to explore the role of SOCS2 in DCM. RESULTS: Bioinformatics analysis showed that SOCS2 was positively correlated with fibrosis related factors. SOCS2 was significantly upregulated in patients and mice with DCM. In vivo experiments showed that targeted inhibition of cardiac SOCS2 could improve mouse cardiac function and alleviate myocardial fibrosis. Further research demonstrated that SOCS2 promoted the transformation of myofibroblasts. Knockdown of SOCS2 reduced the nuclear localization of ß-catenin, which inhibited the fibrogenic effect of Wnt/ß-catenin pathway. In addition, bioinformatics analysis suggested that lymphoid enhancer binding factor 1 (LEF1) was significantly positively correlated with SOCS2. Finally, dual luciferase assays demonstrated that LEF1 could bind to the promoter region of SOCS2, thereby mediating its transcriptional activation. CONCLUSION: SOCS2 could activate the Wnt/ß-catenin by regulating the nuclear translocation of ß-catenin, which induces the transcriptional activation of SOCS2. Overall, these results indicated a positive feedback activation phenomenon between SOCS2, ß-catenin and LEF1 in DCM. These results suggested that inhibition of SOCS2 could effectively alleviate the progression of myocardial fibrosis and improve cardiac function.


Sujet(s)
Fibrose , Myocarde , Protéines SOCS , bêta-Caténine , Animaux , bêta-Caténine/métabolisme , bêta-Caténine/génétique , Souris , Fibrose/métabolisme , Humains , Rats , Myocarde/métabolisme , Myocarde/anatomopathologie , Mâle , Protéines SOCS/métabolisme , Protéines SOCS/génétique , Cardiomyopathie dilatée/métabolisme , Cardiomyopathie dilatée/anatomopathologie , Cardiomyopathie dilatée/génétique , Voie de signalisation Wnt , Modèles animaux de maladie humaine , Noyau de la cellule/métabolisme , Myofibroblastes/métabolisme , Myofibroblastes/anatomopathologie , Souris de lignée C57BL
10.
Nat Commun ; 15(1): 6177, 2024 Jul 22.
Article de Anglais | MEDLINE | ID: mdl-39039081

RÉSUMÉ

The ankyrin (ANK) SOCS box (ASB) family, encompassing ASB1-18, is the largest group of substrate receptors of cullin 5 Ring E3 ubiquitin ligase. Nonetheless, the mechanism of substrate recognition by ASB family proteins has remained largely elusive. Here we present the crystal structure of ASB7-Elongin B-Elongin C ternary complex bound to a conserved helical degron. ASB7 employs its ANK3-6 to form an extended groove, effectively interacting with the internal α-helix-degron through a network of side-chain-mediated electrostatic and hydrophobic interactions. Our structural findings, combined with biochemical and cellular analyses, identify the key residues of the degron motif and ASB7 required for their recognition. This will facilitate the identification of additional physiological substrates of ASB7 by providing a defined degron motif for screening. Furthermore, the structural insights provide a basis for the rational design of compounds that can specifically target ASB7 by disrupting its interaction with its cognate degron.


Sujet(s)
Liaison aux protéines , Protéines SOCS , Humains , Cristallographie aux rayons X , Protéines SOCS/métabolisme , Protéines SOCS/composition chimique , Protéines SOCS/génétique , Ubiquitin-protein ligases/métabolisme , Ubiquitin-protein ligases/composition chimique , Ubiquitin-protein ligases/génétique , Ankyrines/métabolisme , Ankyrines/composition chimique , Ankyrines/génétique , Modèles moléculaires , Élongine/métabolisme , Élongine/génétique , Élongine/composition chimique , Cellules HEK293 , Motifs d'acides aminés , Degrons
11.
Helicobacter ; 29(3): e13105, 2024.
Article de Anglais | MEDLINE | ID: mdl-38924222

RÉSUMÉ

Helicobacter pylori (H. pylori) colonizes the stomach and leads to the secretion of a vast range of cytokines by infiltrated leukocytes directing immune/inflammatory response against the bacterium. To regulate immune/inflammatory responses, suppressors of cytokine signaling (SOCS) proteins bind to multiple signaling components located downstream of cytokine receptors, such as Janus kinase (JAK), signal transducers and activators of transcription (STAT). Dysfunctional SOCS proteins in immune cells may facilitate the immune evasion of H. pylori, allowing the bacteria to induce chronic inflammation. Dysregulation of SOCS expression and function can contribute to the sustained H. pylori-mediated gastric inflammation which can lead to gastric cancer (GC) development. Among SOCS molecules, dysregulated expression of SOCS1, SOCS2, SOCS3, and SOCS6 were indicated in H. pylori-infected individuals as well as in GC tissues and cells. H. pylori-induced SOCS1, SOCS2, SOCS3, and SOCS6 dysregulation can contribute to the GC development. The expression of SOCS molecules can be influenced by various factors, such as epigenetic DNA methylation, noncoding RNAs, and gene polymorphisms. Modulation of the expression of SOCS molecules in gastric epithelial cells and immune cells can be considered to control gastric carcinogenesis as well as regulate antitumor immune responses, respectively. This review aimed to explain the interplay between H. pylori and SOCS molecules in GC development and immune response induction as well as to provide insights regarding potential therapeutic strategies modulating SOCS molecules.


Sujet(s)
Infections à Helicobacter , Helicobacter pylori , Tumeurs de l'estomac , Protéines SOCS , Humains , Tumeurs de l'estomac/immunologie , Tumeurs de l'estomac/microbiologie , Helicobacter pylori/immunologie , Helicobacter pylori/pathogénicité , Infections à Helicobacter/immunologie , Infections à Helicobacter/microbiologie , Protéines SOCS/métabolisme , Protéines SOCS/génétique , Interactions hôte-pathogène/immunologie , Transduction du signal
12.
Fish Shellfish Immunol ; 151: 109669, 2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-38849106

RÉSUMÉ

The suppressor of cytokine signaling (SOCS) gene family is a group of genes involved in the negative regulation of cytokine signal transduction. The members of this family play a crucial role in regulating immune and inflammatory processes. However, comprehensive investigations of these genes have not yet been conducted in the economically significant fish large yellow croaker (Larimichthys crocea). In this study, a total of 13 SOCS genes (LcSOCS1a, LcSOCS1b, LcSOCS2, LcSOCS3a, LcSOCS3b, LcSOCS4, LcSOCS5a, LcSOCS5b, LcSOCS6, LcSOCS7a, LcSOCS7b, LcCISHa and LcCISHb) were identified and analyzed in L. crocea. The phylogenetic tree revealed a high conservation of SOCS genes in evolution, and the gene structure and motif analysis indicated a high similarity in the structure of LcSOCSs in the same subfamily. In addition, the expression patterns of LcSOCSs showed that LcSOCS1b was significantly down-regulated in all time under acute hypoxia stress, but it was markedly up-regulated throughout the entire process after P. plecoglossicida infection, revealing its different immune effects to two stresses. Besides, LcSOCS2a, LcSOCS6 and LcSOCS7a only participated in acute hypoxic stress, while LcSOCS5a was more sensitive to P. plecoglossicida infection. In summary, these results indicated that SOCS genes were involved in stress responses to both biological and non-biological stimuli, setting the foundation for deeper study on the functions of SOCS genes.


Sujet(s)
Maladies des poissons , Protéines de poisson , Régulation de l'expression des gènes , Immunité innée , Perciformes , Phylogenèse , Infections à Pseudomonas , Pseudomonas , Protéines SOCS , Animaux , Perciformes/immunologie , Perciformes/génétique , Maladies des poissons/immunologie , Protéines de poisson/génétique , Protéines de poisson/immunologie , Protéines de poisson/composition chimique , Protéines SOCS/génétique , Protéines SOCS/immunologie , Protéines SOCS/composition chimique , Immunité innée/génétique , Infections à Pseudomonas/immunologie , Infections à Pseudomonas/médecine vétérinaire , Infections à Pseudomonas/génétique , Pseudomonas/physiologie , Régulation de l'expression des gènes/immunologie , Analyse de profil d'expression de gènes/médecine vétérinaire , Stress physiologique/immunologie , Stress physiologique/génétique , Alignement de séquences/médecine vétérinaire , Hypoxie/génétique , Hypoxie/immunologie , Hypoxie/médecine vétérinaire
13.
Histol Histopathol ; 39(11): 1493-1503, 2024 Nov.
Article de Anglais | MEDLINE | ID: mdl-38591191

RÉSUMÉ

Chondroitin polymerizing factor (CHPF) has been reported to play a pivotal role in the progression of multiple cancers, however, the relationship between CHPF and colorectal cancer (CRC) progression has not been fully understood. The current study revealed that CHPF expression was upregulated in patients with CRC and correlated with an unfavorable prognosis. Also, CHPF knockdown effectively suppressed the viability and mobility of CRC cells and the growth of xenograft tumors. Additionally, SMAD9 was identified as a downstream target of CHPF. SMAD9 knockdown successfully abrogated the promotion of CHPF overexpression in CRC progression, indicating that CHPF regulated the development of CRC through SMAD9. Mechanistically, SMAD9 is ubiquitinated by ASB2, and the regulatory effect of CHPF on SMAD9 activity was exerted via its mediation of ASB2. Collectively, CHPF functioned as a promising prognostic biomarker and tumor-promoter of CRC by regulating the ASB2-mediated ubiquitination of SMAD9.


Sujet(s)
Tumeurs colorectales , Évolution de la maladie , Souris nude , Ubiquitination , Humains , Tumeurs colorectales/anatomopathologie , Tumeurs colorectales/métabolisme , Tumeurs colorectales/génétique , Animaux , Femelle , Mâle , Souris , N-acetylgalactosaminyltransferase/métabolisme , N-acetylgalactosaminyltransferase/génétique , Lignée cellulaire tumorale , Souris de lignée BALB C , Régulation de l'expression des gènes tumoraux , Adulte d'âge moyen , Mouvement cellulaire , Protéines SOCS/métabolisme , Protéines SOCS/génétique , Marqueurs biologiques tumoraux/métabolisme , Marqueurs biologiques tumoraux/génétique , Pronostic
14.
Poult Sci ; 103(6): 103672, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38564834

RÉSUMÉ

The development of the avian wing pattern has been the subject of heated debate due to its special shape. The Suppressor of cytokine signaling 2 (SOCS2) gene encodes a negative regulator of growth hormone (GH) signaling and bone growth and is known to be strongly expressed in the third digit of chicken forelimbs. These observations suggest that SOCS2 might regulate the morphology of the avian wing, however, the function of SOCS2 in avian limb development remains unknown. Here, we reexamined SOCS2 expression in successive developmental stages of chicken limb development by in situ hybridization (ISH) and describe extended expression from the posterior of the stypolod to the third digit of the forelimbs. We used the RCAS avian retrovirus to overexpress SOCS2 in the developing chicken limb buds, which resulted in reduced or malformed chicken wings while hindlimbs developed normally. Transcriptome sequencing (mRNA-Seq) revealed changes in expression of genes known to be associated with growth and development in forelimbs with overexpressed SOCS2. This study highlights a pivotal role for SOCS2 during the development of the wing in the chicken and provides new insight into molecular mechanisms regulating avian limb development.


Sujet(s)
Protéines aviaires , Poulets , Protéines SOCS , Ailes d'animaux , Animaux , Protéines SOCS/génétique , Protéines SOCS/métabolisme , Embryon de poulet , Ailes d'animaux/croissance et développement , Protéines aviaires/génétique , Protéines aviaires/métabolisme , Poulets/croissance et développement , Poulets/génétique , Membre thoracique , Bourgeons de membre/métabolisme , Régulation de l'expression des gènes au cours du développement
15.
Cells ; 13(6)2024 Mar 13.
Article de Anglais | MEDLINE | ID: mdl-38534350

RÉSUMÉ

Inflammatory skin diseases include a series of disorders characterized by a strong activation of the innate and adaptive immune system in which proinflammatory cytokines play a fundamental role in supporting inflammation. Skin inflammation is a complex process influenced by various factors, including genetic and environmental factors, characterized by the dysfunction of both immune and non-immune cells. Psoriasis (PS) and atopic dermatitis (AD) are the most common chronic inflammatory conditions of the skin whose pathogeneses are very complex and multifactorial. Both diseases are characterized by an immunological dysfunction involving a predominance of Th1 and Th17 cells in PS and of Th2 cells in AD. Suppressor of cytokine signaling (SOCS) proteins are intracellular proteins that control inflammatory responses by regulating various signaling pathways activated by proinflammatory cytokines. SOCS signaling is involved in the regulation and progression of inflammatory responses in skin-resident and non-resident immune cells, and recent data suggest that these negative modulators are dysregulated in inflammatory skin diseases such as PS and AD. This review focuses on the current understanding about the role of SOCS proteins in modulating the activity of inflammatory mediators implicated in the pathogenesis of inflammatory skin diseases such as PS and AD.


Sujet(s)
Eczéma atopique , Psoriasis , Humains , Protéines SOCS/génétique , Protéines SOCS/métabolisme , Transduction du signal/génétique , Cytokines/métabolisme , Inflammation
16.
mBio ; 15(3): e0321323, 2024 Mar 13.
Article de Anglais | MEDLINE | ID: mdl-38376239

RÉSUMÉ

DEAD-box helicase (DDX) family members play differential roles in regulating innate antiviral immune response. However, the physiological roles played by DDX4 in antiviral innate immunity remain unclear. In this study, we unveiled that DDX4 acts as a positive regulatory molecule of Type-I interferon (IFN-I)-mediated antiviral activity. Our findings demonstrate that IFN-I upregulates DDX4 protein levels, and subsequently, overexpression of DDX4 enhances the IFN-I-mediated signaling pathway. This creates a positive feedback loop that amplifies the antiviral response. DDX4 was found to bind with deubiquitinase ubiquitin-specific protease 7 (USP7), leading to the disruption of the interaction between USP7 and suppressor of cytokine signaling 1 (SOCS1) and the subsequent degradation of SOCS1. This process enhances the antiviral function of IFN-I. Our findings provide new insights into the regulatory role of DDX4 in the IFN-I response.IMPORTANCEDDX4, identified as a putative RNA helicase that modulates RNA secondary structure through RNA binding, is primarily acknowledged for its role in regulating mRNA translation within the germline. Nevertheless, the extent of DDX4's involvement in the antiviral innate immune response remains largely unexplored. This study presents evidence of a previously unrecognized positive feedback loop between DDX4 and the antiviral response, suggesting that disruption of this loop may serve as a novel mechanism for viral evasion. Furthermore, our findings elucidate a positive regulatory mechanism by which the DDX4/USP7/SOCS1 axis mediates the antiviral activity of Type-I interferon, which provides new insight into strategies for improving the efficacy of IFN-based antiviral therapy.


Sujet(s)
Interféron de type I , Ubiquitin-specific peptidase 7/génétique , Ubiquitin-specific peptidase 7/métabolisme , Protéine-1 suppressive de la signalisation des cytokines/métabolisme , Protéines SOCS/génétique , Protéines SOCS/métabolisme , Immunité innée , ARN
17.
Cell Biochem Biophys ; 82(2): 715-727, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38319584

RÉSUMÉ

The ubiquitin proteasome system is a highly specific and selective protein regulatory system that plays an essential role in the regulation of the cell cycle. Despite its significance, the role of ubiquitination in cardiomyocyte proliferation remains largely unclear. This study aimed to investigate the potential impact of E3 ubiquitin ligase ASB14 (Ankyrin Repeat And SOCS Box Containing 14) on cardiac regeneration. We conducted a microarray analysis of apical resection ventricle tissues, and our findings revealed that ASB14 was down-regulated during the cardiac regenerative response. Subsequently, we examined the effect of ASB14 silencing on cardiomyocyte nuclear proliferation both in vitro and in vivo. Our results indicated that ASB14 silencing promoted cardiomyocyte nuclear proliferation, suggesting that ASB14 may play a role in regulating cardiac regeneration. To further investigate the potential therapeutic implications of ASB14 deficiency, we examined the cardiac function of mice with ASB14 deficiency in response to ischemic injury. Our findings showed that mice with ASB14 deficiency exhibited preserved cardiac function and a therapeutic effect in response to ischemic injury, which was attributed to the enhancement of cardiomyocyte nuclear proliferation. To elucidate the underlying mechanisms, we investigated the effect of ASB14 on microtubule-associated protein RP/EB family member 2 (MAPRE2) protein degradation. Our results indicated that the loss of ASB14 decreased the degradation of MAPRE2 protein, subsequently promoting cardiomyocyte nuclear proliferation and enhancing cardiac repair after myocardial infarction (MI). In conclusion, our study provides evidence that inhibition of ASB14-mediated MAPRE2 ubiquitination promotes cardiomyocyte nuclear proliferation, which may serve as a potential target for treating heart failure induced by MI injury.


Sujet(s)
Prolifération cellulaire , Protéines associées aux microtubules , Myocytes cardiaques , Ubiquitin-protein ligases , Ubiquitination , Animaux , Mâle , Souris , Rats , Souris de lignée C57BL , Protéines associées aux microtubules/métabolisme , Protéines associées aux microtubules/génétique , Infarctus du myocarde/métabolisme , Infarctus du myocarde/anatomopathologie , Myocytes cardiaques/métabolisme , Myocytes cardiaques/cytologie , Protéines SOCS/métabolisme , Protéines SOCS/génétique , Ubiquitin-protein ligases/métabolisme , Ubiquitin-protein ligases/génétique
18.
Int J Biol Macromol ; 262(Pt 1): 129876, 2024 Mar.
Article de Anglais | MEDLINE | ID: mdl-38310055

RÉSUMÉ

Impaired polarization of M1 to M2 macrophages has been reported in diabetic wounds. We aimed to improve this polarization by down-regulation of expression of the "Suppressor of Cytokine Signaling 3" (SOCS3) gene in macrophages. Two oligodeoxynucleotide (ASO) sequences were designed against SOC3 mRNA and were loaded to mannosylated-polyethyleneimine (Man-PEI). The optimum N/P ratio for Man-PEI-ASO was determined to be 8 based on loading efficiency, particle size, zeta potential, cellular uptake and cytotoxicity assay. pH stability of ASO in Man-PEI-ASO and its protection from DNase I was confirmed. After in vitro treatment of macrophages with Man-PEI-ASO, SOCS3 was downregulated, SOCS1 upregulated, and SOCS1/SOCS3 ratio increased. Also, expressions of macrophage markers of M2 (IL-10, Arg1, CD206) increased and those of M1 (IL-1ß, NOS2, CD68) decreased, and secretion of pro-inflammatory cytokines (TNF-α and IL-1ß) decreased while that of anti-inflammatory cytokine IL-4 increased. All suggested a polarization into M2 phenotype. Finally, the Man-PEI-ASO was loaded in hydrogel and applied to a diabetic wound model in mice. It improved the healing to the level observed in non-diabetic wounds. We show that using antisense sequences against SOC3 mRNA, macrophage polarization could be directed into the M2 phenotype and healing of diabetic wound could be highly improved.


Sujet(s)
Diabète , Macrophages , Protéines SOCS , Cicatrisation de plaie , Animaux , Humains , Souris , Cytokines/génétique , Cytokines/métabolisme , Diabète/métabolisme , Macrophages/métabolisme , ARN messager/métabolisme , Protéine-3 suppressive de la signalisation des cytokine/génétique , Protéine-3 suppressive de la signalisation des cytokine/métabolisme , Protéines SOCS/génétique , Protéines SOCS/métabolisme
19.
Eur J Haematol ; 112(3): 439-449, 2024 Mar.
Article de Anglais | MEDLINE | ID: mdl-37950514

RÉSUMÉ

OBJECTIVES: As a tumor suppressor gene, SOCS3 inhibits the growth of tumor cells by regulating JAK/STAT signaling pathway through negative feedback. This study aimed to investigate the biological function and mechanism of SOCS3 methylation mediated by DNMTs in the development of AML. METHODS: Bone marrow samples were collected from 70 AML patients and 20 healthy volunteers. The expression and methylation status of each gene were detected by RT-qPCR, western blot and MS-PCR, and the growth and apoptosis rate of leukemia cell lines were detected by CCK-8 and flow cytometry. The effects of changes in SOCS3 gene expression and methylation status of AML cell lines were observed by gene transfection and gene knockdown. RESULTS: The methylation rate of SOCS3 in AML initial treatment group was significantly higher than that in the remission group and the normal control group (60% vs. 0%, 0%). The expression of SOCS3 in the SOCS3 methylation group was significantly lower than that in the non-methylated group and control group, while the expression of DNMT1, DNMT3a, p-JAK2, p-STAT3 and p-STAT5 were significantly higher than those in the non-methylated group and control group. Demethylation treatment, SOCS3 transfection and DNMT3a knockdown could up-regulate the expression of SOCS3, which decreased the proliferation and increased the apoptosis of leukemia cell lines. CONCLUSION: SOCS3 methylation mediated by DNMTs promotes the occurrence and development of AML and can be used as a potential biomarker for the diagnosis and efficacy evaluation of AML.


Sujet(s)
Leucémie aigüe myéloïde , Transduction du signal , Humains , Lignée cellulaire tumorale , Protéines SOCS/génétique , Méthylation de l'ADN , Leucémie aigüe myéloïde/génétique , Protéine-3 suppressive de la signalisation des cytokine/génétique , Protéine-3 suppressive de la signalisation des cytokine/métabolisme
20.
Microbes Infect ; 26(3): 105282, 2024.
Article de Anglais | MEDLINE | ID: mdl-38135025

RÉSUMÉ

Mycobacterium tuberculosis (Mtb) infection leads to upregulation of Suppressors of Cytokine signaling (SOCS) expression in host macrophages (Mϕ). SOCS proteins inhibit cytokine signaling by negatively regulating JAK/STAT. We investigated this host-pathogen dialectic at the level of transcription. We used phorbol-differentiated THP-1 Mϕ infected with Mtb to investigate preferential upregulation of some SOCS isoforms that are known to inhibit signaling by IFN-γ, IL-12, and IL-6. We examined time kinetics of likely transcription factors and signaling molecules upstream of SOCS transcription, and survival of intracellular Mtb following SOCS upregulation. Our results suggest a plausible mechanism that involves PGE2 secretion during infection to induce the PKA/CREB axis, culminating in nuclear translocation of C/EBPß to induce expression of SOCS1. Mtb-infected Mϕ secreted IL-10, suggesting a mechanism of induction of STAT3, which may subsequently induce SOCS3. We provide evidence of temporal variation in SOCS isoform exspression and decay. Small-interfering RNA-mediated knockdown of SOCS1 and SOCS3 restored the pro-inflammatory milieu and reduced Mtb viability. In mice infected with Mtb, SOCS isoforms persisted across Days 28-85 post infection. Our results suggest that differential temporal regulation of SOCS isoforms by Mtb drives the host immune response towards a phenotype that facilitates the pathogen's survival.


Sujet(s)
Mycobacterium tuberculosis , Humains , Animaux , Souris , Protéine-1 suppressive de la signalisation des cytokines/génétique , Protéine-1 suppressive de la signalisation des cytokines/métabolisme , Protéine-3 suppressive de la signalisation des cytokine/génétique , Protéine-3 suppressive de la signalisation des cytokine/métabolisme , Protéines SOCS/génétique , Protéines SOCS/métabolisme , Macrophages/microbiologie , Interleukine-12 , Isoformes de protéines/métabolisme
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE