Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 2.269
Filtrer
1.
Front Immunol ; 15: 1381802, 2024.
Article de Anglais | MEDLINE | ID: mdl-38966637

RÉSUMÉ

Background: Yishen-Tongbi Decoction (YSTB), a traditional Chinese prescription, has been used to improve syndromes of rheumatoid arthritis (RA) for many years. Previous research has shown that YSTB has anti-inflammatory and analgesic properties. However, the underlying molecular mechanism of the anti-RA effects of YSTB remains unclear. Purpose and study design: The purpose of this research was to investigate how YSTB affected mice with collagen-induced arthritis (CIA) and RAW264.7 cells induced with lipopolysaccharide (LPS). Results: The findings show that YSTB could significantly improve the clinical arthritic symptoms of CIA mice (mitigate paw swelling, arthritis score, thymus and spleen indices, augment body weight), downregulated expression of pro-inflammatory cytokines like tumor necrosis factor-alpha (TNF-α), interleukin-1ß (IL-1ß), IL-6 and IL-17, while upregulated the level of anti-inflammatory like IL-10 and transforming growth factor-ß (TGF-ß). Meanwhile, YSTB inhibits bone erosion and reduces inflammatory cell infiltration, synovial proliferation, and joint destruction in CIA mice. In addition, we found that YSTB was able to suppress the LPS-induced inflammation of RAW264.7 cells, which was ascribed to the suppression of nitric oxide (NO) production and reactive oxygen species formation (ROS). YSTB also inhibited the production of inducible nitric oxide synthase and reduced the releases of pro-inflammatory cytokines TNF-α, IL-1ß, and IL-6 in LPS-induced RAW264.7 cells. Furthermore, the phosphorylation expression of JAK2, JAK3, STAT3, p38, ERK and p65 protein could be suppressed by YSTB, while the expression of SOCS3 could be activated. Conclusion: Taken together, YSTB possesses anti-inflammatory and prevention bone destruction effects in RA disease by regulating the JAK/STAT3/SOCS3 signaling pathway.


Sujet(s)
Arthrite expérimentale , Polyarthrite rhumatoïde , Médicaments issus de plantes chinoises , Janus kinases , Facteur de transcription STAT-3 , Transduction du signal , Protéine-3 suppressive de la signalisation des cytokine , Animaux , Souris , Médicaments issus de plantes chinoises/pharmacologie , Médicaments issus de plantes chinoises/usage thérapeutique , Polyarthrite rhumatoïde/traitement médicamenteux , Polyarthrite rhumatoïde/métabolisme , Cellules RAW 264.7 , Facteur de transcription STAT-3/métabolisme , Protéine-3 suppressive de la signalisation des cytokine/métabolisme , Protéine-3 suppressive de la signalisation des cytokine/génétique , Arthrite expérimentale/traitement médicamenteux , Arthrite expérimentale/anatomopathologie , Arthrite expérimentale/métabolisme , Transduction du signal/effets des médicaments et des substances chimiques , Janus kinases/métabolisme , Mâle , Cytokines/métabolisme , Anti-inflammatoires/pharmacologie , Anti-inflammatoires/usage thérapeutique , Inflammation/traitement médicamenteux , Souris de lignée DBA , Modèles animaux de maladie humaine
2.
Front Immunol ; 15: 1393799, 2024.
Article de Anglais | MEDLINE | ID: mdl-38975347

RÉSUMÉ

SOCS are a family of negative inhibitors of the molecular cascades induced by cytokines, growth factors and hormones. At molecular level, SOCS proteins inhibit the kinase activity of specific sets of receptor-associated Janus Activated Kinases (JAKs), thereby suppressing the propagation of intracellular signals. Of the eight known members, SOCS1 and SOCS3 inhibit activity of JAKs mainly induced by cytokines and can play key roles in regulation of inflammatory and immune responses. SOCS1 and SOCS3 are the most well-characterized SOCS members in skin inflammatory diseases, where their inhibitory activity on cytokine activated JAKs and consequent anti-inflammatory action has been widely investigated in epidermal keratinocytes. Structurally, SOCS1 and SOCS3 share the presence of a N-terminal domain containing a kinase inhibitory region (KIR) motif able to act as a pseudo-substrate for JAK and to inhibit its activity. During the last decades, the design and employment of SOCS1 and SOCS3-derived peptides mimicking KIR domains in experimental models of dermatoses definitively established a strong anti-inflammatory and ameliorative impact of JAK inhibition on skin inflammatory responses. Herein, we discuss the importance of the findings collected in the past on SOCS1 and SOCS3 function in the inflammatory responses associated to skin immune-mediated diseases and malignancies, for the development of the JAK inhibitor drugs. Among them, different JAK inhibitors have been introduced in the clinical practice for treatment of atopic dermatitis and psoriasis, and others are being investigated for skin diseases like alopecia areata and vitiligo.


Sujet(s)
Transformation cellulaire néoplasique , Protéine-1 suppressive de la signalisation des cytokines , Protéine-3 suppressive de la signalisation des cytokine , Humains , Protéine-1 suppressive de la signalisation des cytokines/métabolisme , Animaux , Protéine-3 suppressive de la signalisation des cytokine/métabolisme , Transformation cellulaire néoplasique/immunologie , Transformation cellulaire néoplasique/métabolisme , Transduction du signal , Tumeurs cutanées/immunologie , Tumeurs cutanées/métabolisme , Tumeurs cutanées/anatomopathologie , Dermatite/immunologie , Dermatite/métabolisme , Janus kinases/métabolisme , Peau/immunologie , Peau/anatomopathologie , Peau/métabolisme
3.
Yale J Biol Med ; 97(2): 165-177, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38947108

RÉSUMÉ

Background: Chronic rhinosinusitis (CRS) is an inflammatory condition classified into chronic rhinosinusitis with nasal polyps (CRSwNP) and chronic rhinosinusitis without nasal polyps (CRSsNP). Th cells manage inflammatory cells in CRS. Suppressor of Cytokine Signaling (SOCS) proteins regulate Janus kinase (JAK)-signal transducer and activator of transcription (STAT) pathway in Th cells by polarizing toward Th1, Th2, and Th17 cells. This study evaluated the levels of SOCS1,3,5 in CRS patients to find associations with Th cells. Methods: In this cross-sectional study, 20 CRSwNP patients, 12 CRSsNP patients, and 12 controls participated. The infiltration of CD4+ T cells was determined using immunohistochemistry. The expression of specific transcription factors and SOCS proteins was assessed using real-time PCR. Cytokine levels were evaluated using ELISA. SOCS protein levels were investigated using western blot analysis. Results: The expression of SOCS3 increased in the CRSwNP group compared to CRSsNP and control groups (p <0.001). SOCS3 protein levels increased in the CRSwNP group compared to CRSsNP (p <0.05) and control (p <0.001) groups. Although there was a significant difference in SOCS5 expression between CRSsNP and control groups, SOCS5 protein levels were significantly different between CRSsNP and control (p <0.001) and CRSwNP (p <0.05) groups. Conclusions: Targeted therapies may be suggested for CRS by modulating SOCS3 and SOCS5 proteins that are responsible for polarization of Th cells toward Th2 or Th1 cells, respectively. JAK-STAT pathway targeting, which encompasses numerous cells, can be limited to SOCS proteins to more effectively orchestrate Th cell differentiation.


Sujet(s)
Rhinite , Sinusite , Protéine-3 suppressive de la signalisation des cytokine , Protéines SOCS , Humains , Sinusite/métabolisme , Sinusite/immunologie , Protéines SOCS/métabolisme , Maladie chronique , Mâle , Protéine-3 suppressive de la signalisation des cytokine/métabolisme , Rhinite/métabolisme , Rhinite/immunologie , Femelle , Adulte , Adulte d'âge moyen , Lymphocytes T auxiliaires/métabolisme , Lymphocytes T auxiliaires/immunologie , Études transversales , Polypes du nez/métabolisme , Cytokines/métabolisme , Protéine-1 suppressive de la signalisation des cytokines/métabolisme , Protéine-1 suppressive de la signalisation des cytokines/génétique , Transduction du signal ,
4.
Int J Mol Sci ; 25(12)2024 Jun 11.
Article de Anglais | MEDLINE | ID: mdl-38928125

RÉSUMÉ

Leptin regulates lipid metabolism, maximizing insulin sensitivity; however, peripheral leptin resistance is not fully understood, and its contribution to metabolic dysfunction-associated steatotic liver disease (MASLD) is unclear. This study evaluated the contribution of the leptin axis to MASLD in humans. Forty-three participants, mostly female (86.04%), who underwent cholecystectomy were biopsied. Of the participants, 24 were healthy controls, 8 had MASLD, and 11 had metabolic dysfunction-associated steatohepatitis (MASH). Clinical and biochemical data and the gene expression of leptin, leptin receptor (LEPR), suppressor of cytokine signaling 3 (SOCS3), sterol regulatory element-binding transcription factor 1 (SREBF1), stearoyl-CoA desaturase-1 (SCD1), and patatin-like phospholipase domain-containing protein 2 (PNPLA2), were determined from liver and adipose tissue. Higher serum leptin and LEPR levels in the omental adipose tissue (OAT) and liver with MASH were found. In the liver, LEPR was positively correlated with leptin expression in adipose tissue, and SOCS3 was correlated with SREBF1-SCD1. In OAT, SOCS3 was correlated with insulin resistance and transaminase enzymes (p < 0.05 for all. In conclusion, we evidenced the correlation between the peripheral leptin resistance axis in OAT-liver crosstalk and the complications of MASLD in humans.


Sujet(s)
Tissu adipeux , Stéatose hépatique , Leptine , Foie , Omentum , Humains , Leptine/métabolisme , Femelle , Mâle , Foie/métabolisme , Adulte d'âge moyen , Omentum/métabolisme , Omentum/anatomopathologie , Tissu adipeux/métabolisme , Adulte , Stéatose hépatique/métabolisme , Stéatose hépatique/anatomopathologie , Récepteurs à la leptine/métabolisme , Récepteurs à la leptine/génétique , Protéine-3 suppressive de la signalisation des cytokine/métabolisme , Protéine-3 suppressive de la signalisation des cytokine/génétique , Insulinorésistance , Protéine-1 de liaison à l'élément de régulation des stérols/métabolisme , Protéine-1 de liaison à l'élément de régulation des stérols/génétique , Acyl-(acyl-carrier-protein)desaturase/métabolisme , Acyl-(acyl-carrier-protein)desaturase/génétique
5.
Mol Med ; 30(1): 78, 2024 Jun 06.
Article de Anglais | MEDLINE | ID: mdl-38844873

RÉSUMÉ

BACKGROUND: Diabetic nephropathy (DN) is a life-threatening renal disease and needs urgent therapies. Wogonin is renoprotective in DN. This study aimed to explore the mechanism of how wogonin regulated high glucose (HG)-induced renal cell injury. METHODS: Diabetic mice (db/db), control db/m mice, and normal glucose (NG)- or HG-treated human tubule epithelial cells (HK-2) were used to evaluate the levels of suppressor of cytokine signaling 3 (SOCS3), Toll-like receptor 4 (TLR4), inflammation and fibrosis. Lentivirus was used to regulate SOCS3 and TLR4 expressions. After oral gavage of wogonin (10 mg/kg) or vehicle in db/db mice, histological morphologies, blood glucose, urinary protein, serum creatinine values (Scr), blood urea nitrogen (BUN), superoxide dismutase (SOD), glutathione (GSH), and reactive oxygen species (ROS) were assessed. RT-qPCR and Western blot evaluated inflammation and fibrosis-related molecules. RESULTS: HG exposure induced high blood glucose, severe renal injuries, high serumal Src and BUN, low SOD and GSH, and increased ROS. HG downregulated SOCS3 but upregulated TLR4 and JAK/STAT, fibrosis, and inflammasome-related proteins. Wogonin alleviated HG-induced renal injuries by decreasing cytokines, ROS, Src, and MDA and increasing SOD and GSH. Meanwhile, wogonin upregulated SOCS3 and downregulated TLR4 under HG conditions. Wogonin-induced SOCS3 overexpression directly decreased TLR4 levels and attenuated JAK/STAT signaling pathway-related inflammation and fibrosis, but SOCS3 knockdown significantly antagonized the protective effects of wogonin. However, TLR4 knockdown diminished SOCS3 knockdown-induced renal injuries. CONCLUSION: Wogonin attenuates renal inflammation and fibrosis by upregulating SOCS3 to inhibit TLR4 and JAK/STAT pathway.


Sujet(s)
Néphropathies diabétiques , Flavanones , Transduction du signal , Protéine-3 suppressive de la signalisation des cytokine , Récepteur de type Toll-4 , Flavanones/pharmacologie , Flavanones/usage thérapeutique , Récepteur de type Toll-4/métabolisme , Protéine-3 suppressive de la signalisation des cytokine/métabolisme , Protéine-3 suppressive de la signalisation des cytokine/génétique , Néphropathies diabétiques/métabolisme , Néphropathies diabétiques/traitement médicamenteux , Néphropathies diabétiques/étiologie , Animaux , Transduction du signal/effets des médicaments et des substances chimiques , Souris , Humains , Mâle , Janus kinases/métabolisme , Facteurs de transcription STAT/métabolisme , Lignée cellulaire , Diabète expérimental/métabolisme , Diabète expérimental/complications , Diabète expérimental/traitement médicamenteux , Modèles animaux de maladie humaine
6.
Nat Commun ; 15(1): 4711, 2024 Jun 03.
Article de Anglais | MEDLINE | ID: mdl-38830841

RÉSUMÉ

The fetal development of organs and functions is vulnerable to perturbation by maternal inflammation which may increase susceptibility to disorders after birth. Because it is not well understood how the placenta and fetus respond to acute lung- inflammation, we characterize the response to maternal pulmonary lipopolysaccharide exposure across 24 h in maternal and fetal organs using multi-omics, imaging and integrative analyses. Unlike maternal organs, which mount strong inflammatory immune responses, the placenta upregulates immuno-modulatory genes, in particular the IL-6 signaling suppressor Socs3. Similarly, we observe no immune response in the fetal liver, which instead displays metabolic changes, including increases in lipids containing docosahexaenoic acid, crucial for fetal brain development. The maternal liver and plasma display similar metabolic alterations, potentially increasing bioavailability of docosahexaenoic acid for the mother and fetus. Thus, our integrated temporal analysis shows that systemic inflammation in the mother leads to a metabolic perturbation in the fetus.


Sujet(s)
Foetus , Lipopolysaccharides , Foie , Poumon , Placenta , Femelle , Grossesse , Placenta/métabolisme , Placenta/immunologie , Animaux , Foetus/immunologie , Foetus/métabolisme , Poumon/immunologie , Poumon/métabolisme , Foie/métabolisme , Foie/immunologie , Acide docosahexaénoïque/métabolisme , Protéine-3 suppressive de la signalisation des cytokine/métabolisme , Protéine-3 suppressive de la signalisation des cytokine/génétique , Souris , Inflammation/immunologie , Inflammation/métabolisme , Souris de lignée C57BL , Adaptation physiologique/immunologie , Développement foetal/immunologie , Échange foetomaternel/immunologie , Interleukine-6/métabolisme , Interleukine-6/immunologie
7.
Emerg Microbes Infect ; 13(1): 2366359, 2024 Dec.
Article de Anglais | MEDLINE | ID: mdl-38855910

RÉSUMÉ

Tuberculosis (TB) remains a leading cause of mortality among individuals coinfected with HIV, characterized by progressive pulmonary inflammation. Despite TB's hallmark being focal granulomatous lung lesions, our understanding of the histopathological features and regulation of inflammation in HIV & TB coinfection remains incomplete. In this study, we aimed to elucidate these histopathological features through an immunohistochemistry analysis of HIV & TB co-infected and TB patients, revealing marked differences. Notably, HIV & TB granulomas exhibited aggregation of CD68 + macrophage (Mφ), while TB lesions predominantly featured aggregation of CD20+ B cells, highlighting distinct immune responses in coinfection. Spatial transcriptome profiling further elucidated CD68+ Mφ aggregation in HIV & TB, accompanied by activation of IL6 pathway, potentially exacerbating inflammation. Through multiplex immunostaining, we validated two granuloma types in HIV & TB versus three in TB, distinguished by cell architecture. Remarkably, in the two types of HIV & TB granulomas, CD68 + Mφ highly co-expressed IL6R/pSTAT3, contrasting TB granulomas' high IFNGRA/SOCS3 expression, indicating different signaling pathways at play. Thus, activation of IL6 pathway may intensify inflammation in HIV & TB-lungs, while SOCS3-enriched immune microenvironment suppresses IL6-induced over-inflammation in TB. These findings provide crucial insights into HIV & TB granuloma formation, shedding light on potential therapeutic targets, particularly for granulomatous pulmonary under HIV & TB co-infection. Our study emphasizes the importance of a comprehensive understanding of the immunopathogenesis of HIV & TB coinfection and suggests potential avenues for targeting IL6 signaling with SOCS3 activators or anti-IL6R agents to mitigate lung inflammation in HIV & TB coinfected individuals.


Sujet(s)
Co-infection , Granulome , Infections à VIH , Poumon , Macrophages , Récepteurs à l'interleukine-6 , Facteur de transcription STAT-3 , Humains , Co-infection/virologie , Co-infection/immunologie , Co-infection/microbiologie , Infections à VIH/complications , Infections à VIH/immunologie , Macrophages/immunologie , Facteur de transcription STAT-3/métabolisme , Facteur de transcription STAT-3/génétique , Granulome/immunologie , Poumon/anatomopathologie , Poumon/immunologie , Récepteurs à l'interleukine-6/métabolisme , Récepteurs à l'interleukine-6/génétique , Protéine-3 suppressive de la signalisation des cytokine/métabolisme , Protéine-3 suppressive de la signalisation des cytokine/génétique , Antigènes de différenciation des myélomonocytes/métabolisme , Antigènes de différenciation des myélomonocytes/génétique , Antigènes CD/métabolisme , Antigènes CD/génétique , Transduction du signal , Tuberculose pulmonaire/immunologie , Tuberculose pulmonaire/complications , Mâle , Tuberculose/immunologie , Tuberculose/microbiologie , Tuberculose/complications , Femelle , Adulte , Interleukine-6/métabolisme , Interleukine-6/génétique ,
8.
Cell Stem Cell ; 31(7): 1020-1037.e9, 2024 Jul 05.
Article de Anglais | MEDLINE | ID: mdl-38754428

RÉSUMÉ

Autophagy is central to the benefits of longevity signaling programs and to hematopoietic stem cell (HSC) response to nutrient stress. With age, a subset of HSCs increases autophagy flux and preserves regenerative capacity, but the signals triggering autophagy and maintaining the functionality of autophagy-activated old HSCs (oHSCs) remain unknown. Here, we demonstrate that autophagy is an adaptive cytoprotective response to chronic inflammation in the aging murine bone marrow (BM) niche. We find that inflammation impairs glucose uptake and suppresses glycolysis in oHSCs through Socs3-mediated inhibition of AKT/FoxO-dependent signaling, with inflammation-mediated autophagy engagement preserving functional quiescence by enabling metabolic adaptation to glycolytic impairment. Moreover, we show that transient autophagy induction via a short-term fasting/refeeding paradigm normalizes glycolytic flux and significantly boosts oHSC regenerative potential. Our results identify inflammation-driven glucose hypometabolism as a key driver of HSC dysfunction with age and establish autophagy as a targetable node to reset oHSC regenerative capacity.


Sujet(s)
Autophagie , Glycolyse , Cellules souches hématopoïétiques , Inflammation , Animaux , Cellules souches hématopoïétiques/métabolisme , Inflammation/anatomopathologie , Inflammation/métabolisme , Souris , Souris de lignée C57BL , Vieillissement/anatomopathologie , Vieillissement/métabolisme , Vieillissement de la cellule , Transduction du signal , Protéine-3 suppressive de la signalisation des cytokine/métabolisme , Glucose/métabolisme
9.
Exp Neurol ; 377: 114809, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38714285

RÉSUMÉ

Neurogenesis as a potential strategy to improve the consequences of intracerebral hemorrhage (ICH). The current study investigates the effects of withaferin A (WFA) in combination with leptin (LEP) on ICH and neurogenesis mechanisms. LEP levels were dramatically reduced on days 7 and 14 following ICH insults in mice, but continuous WFA therapy significantly improved the potency of intrinsic LEP on day 14 after ICH. Furthermore, WFA combined with LEP enhances intrinsic neurogenesis and lessen motor deficits and long-term cognitive outcomes after ICH. In parallel, leptin deficiency in ob/ob mice limits enhancement of neurogenesis following ICH in response to WFA combined with LEP treatment. Importantly, the functional recovery conferred by WFA combined with LEP after ICH was inhibited by neurogenesis suppression. Mechanistically, this study unveiled that the signal transducer and activator of transcription-3 (STAT3) / suppressor of cytokine signaling-3 (SOCS3) pathway is a critical signaling pathway through which WFA combined with LEP treatment promotes intrinsic neurogenesis after ICH. Collectively, the results of this study elucidate the neuroprotective effects of WFA and LEP in ICH, and highlight a potential approach for ICH cell therapy.


Sujet(s)
Hémorragie cérébrale , Leptine , Souris de lignée C57BL , Neurogenèse , Facteur de transcription STAT-3 , Transduction du signal , Protéine-3 suppressive de la signalisation des cytokine , Withanolides , Animaux , Withanolides/pharmacologie , Neurogenèse/effets des médicaments et des substances chimiques , Facteur de transcription STAT-3/métabolisme , Souris , Protéine-3 suppressive de la signalisation des cytokine/métabolisme , Leptine/pharmacologie , Mâle , Transduction du signal/effets des médicaments et des substances chimiques , Hémorragie cérébrale/traitement médicamenteux , Neuroprotecteurs/pharmacologie , Association de médicaments
10.
Int J Mol Sci ; 25(10)2024 May 18.
Article de Anglais | MEDLINE | ID: mdl-38791551

RÉSUMÉ

Rotavirus is the main cause of acute diarrhea in children up to five years of age. In this regard, probiotics are commonly used to treat or prevent gastroenteritis including viral infections. The anti-rotavirus effect of Bifidobacterium longum and Chlorella sorokiniana, by reducing viral infectivity and improving IFN-type I response, has been previously reported. The present study aimed to study the effect of B. longum and/or C. sorokiniana on modulating the antiviral cellular immune response mediated by IFN-γ, IL-10, SOCS3, STAT1, and STAT2 genes in rotavirus-infected cells. To determine the mRNA relative expression of these genes, HT-29 cells were treated with B. longum and C. sorokiniana alone or in combination, followed by rotavirus infection. In addition, infected cells were treated with B. longum and/or C. sorokiniana. Cellular RNA was purified, used for cDNA synthesis, and amplified by qPCR. Our results demonstrated that the combination of B. longum and C. sorokiniana stimulates the antiviral cellular immune response by upregulating IFN-γ and may block pro-inflammatory cytokines by upregulating IL-10 and SOCS3. The results of our study indicated that B. longum, C. sorokiniana, or their combination improve antiviral cellular immune response and might modulate pro-inflammatory responses.


Sujet(s)
Bifidobacterium longum , Chlorella , Interféron gamma , Interleukine-10 , Probiotiques , Infections à rotavirus , Protéine-3 suppressive de la signalisation des cytokine , Humains , Cellules HT29 , Interféron gamma/métabolisme , Interleukine-10/métabolisme , Probiotiques/pharmacologie , Rotavirus/physiologie , Infections à rotavirus/immunologie , Infections à rotavirus/virologie , Facteur de transcription STAT-1/métabolisme , Protéine-3 suppressive de la signalisation des cytokine/métabolisme
11.
Mol Genet Genomics ; 299(1): 60, 2024 May 27.
Article de Anglais | MEDLINE | ID: mdl-38801463

RÉSUMÉ

Type 2 diabetes (DM2) is an increasingly prevalent disease that challenges tuberculosis (TB) control strategies worldwide. It is significant that DM2 patients with poor glycemic control (PDM2) are prone to developing tuberculosis. Furthermore, elucidating the molecular mechanisms that govern this susceptibility is imperative to address this problem. Therefore, a pilot transcriptomic study was performed. Human blood samples from healthy controls (CTRL, HbA1c < 6.5%), tuberculosis (TB), comorbidity TB-DM2, DM2 (HbA1c 6.5-8.9%), and PDM2 (HbA1c > 10%) groups (n = 4 each) were analyzed by differential expression using microarrays. We use a network strategy to identify potential molecular patterns linking the differentially expressed genes (DEGs) specific for TB-DM2 and PDM2 (p-value < 0.05, fold change > 2). We define OSM, PRKCD, and SOCS3 as key regulatory genes (KRGs) that modulate the immune system and related pathways. RT-qPCR assays confirmed upregulation of OSM, PRKCD, and SOCS3 genes (p < 0.05) in TB-DM2 patients (n = 18) compared to CTRL, DM2, PDM2, or TB groups (n = 17, 19, 15, and 9, respectively). Furthermore, OSM, PRKCD, and SOCS3 were associated with PDM2 susceptibility pathways toward TB-DM2 and formed a putative protein-protein interaction confirmed in STRING. Our results reveal potential molecular patterns where OSM, PRKCD, and SOCS3 are KRGs underlying the compromised immune response and susceptibility of patients with PDM2 to develop tuberculosis. Therefore, this work paved the way for fundamental research of new molecular targets in TB-DM2. Addressing their cellular implications, and the impact on the diagnosis, treatment, and clinical management of TB-DM2 could help improve the strategy to end tuberculosis for this vulnerable population.


Sujet(s)
Diabète de type 2 , Protéine-3 suppressive de la signalisation des cytokine , Tuberculose , Humains , Diabète de type 2/génétique , Diabète de type 2/sang , Diabète de type 2/complications , Projets pilotes , Tuberculose/génétique , Tuberculose/sang , Mâle , Femelle , Adulte d'âge moyen , Protéine-3 suppressive de la signalisation des cytokine/génétique , Protéine-3 suppressive de la signalisation des cytokine/métabolisme , Régulation de la glycémie , Analyse de profil d'expression de gènes , Sujet âgé , Adulte , Réseaux de régulation génique , Études cas-témoins , Transcriptome/génétique , Prédisposition aux maladies
12.
J Matern Fetal Neonatal Med ; 37(1): 2337723, 2024 Dec.
Article de Anglais | MEDLINE | ID: mdl-38637274

RÉSUMÉ

OBJECTIVE: The objective of this study is to explore the functions and mechanisms of the LncRNA-KCNQ1OT1/miR-29a-3p/SOCS3 molecular pathway in the context of unexplained recurrent spontaneous abortion (URSA). METHODS: We conducted qRT-PCR to assess the levels of LncRNA-KCNQ1OT1, miR-29a-3p, and SOCS3 in both abortion tissues from women who experienced URSA and healthy early pregnant women. A dual-luciferase assay was employed to investigate whether miR-29a-3p targets SOCS3. Furthermore, RNA IP and RNA Pull-Down assays were employed to confirm the interaction between KCNQ1OT1 and SOCS3 with miR-29a-3p. RNA FISH was used to determine the cellular localization of KCNQ1OT1. Additionally, trophoblast cells (HTR8/SVneo) were cultured and the CCK-8 assay was utilized to assess cell proliferation, while flow cytometry was employed to analyze cell apoptosis. RESULTS: Compared to abortion tissues obtained from healthy early pregnant individuals, those from women who experienced URSA displayed a notable downregulation of KCNQ1OT1 and SOCS3, accompanied by an upregulation of miR-29a-3p. Suppression of KCNQ1OT1 resulted in the inhibition of cell proliferation and the facilitation of apoptosis in HTR8/SVneo cells. Our findings suggest that KCNQ1OT1 may exert a regulatory influence on SOCS3 through a competitive binding mechanism with miR-29a-3p. Notably, KCNQ1OT1 exhibited expression in both the cytoplasm and nucleus, with a predominant localization in the cytoplasm. Furthermore, we observed a negative regulatory relationship between miR-29a-3p and SOCS3, as the miR-29a-3p mimic group demonstrated significantly reduced cell proliferation and an increased rate of apoptosis when compared to the negative control (NC mimic) group. Additionally, the SOCS3 Vector group exhibited a substantial improvement in proliferation capability and a marked reduction in the apoptosis rate in comparison to the NC Vector group. The miR-29a-3p mimic + SOCS3 Vector group demonstrated a remarkable enhancement in proliferation and a reduction in apoptosis when compared to the miR-29a-3p mimic group. CONCLUSION: The competitive binding of miR-29a-3p to LncRNA-KCNQ1OT1 appears to result in the elevation of SOCS3 expression, consequently fostering the proliferation of trophoblast cells while concomitantly suppressing apoptosis.


Sujet(s)
Avortements à répétition , microARN , ARN long non codant , Femelle , Humains , Grossesse , Avortements à répétition/génétique , Apoptose/génétique , Lignée cellulaire tumorale , Prolifération cellulaire/génétique , microARN/génétique , microARN/métabolisme , ARN long non codant/génétique , ARN long non codant/métabolisme , Protéine-3 suppressive de la signalisation des cytokine/génétique , Protéine-3 suppressive de la signalisation des cytokine/métabolisme
13.
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
14.
Acta Pharmacol Sin ; 45(7): 1466-1476, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38514862

RÉSUMÉ

Disturbances in intestinal immune homeostasis predispose susceptible individuals to type 1 diabetes (T1D). G-protein-coupled receptor 41 (GPR41) is a receptor for short-chain fatty acids (SCFAs) mainly produced by gut microbiota, which plays key roles in maintaining intestinal homeostasis. In this study, we investigated the role of GPR41 in the progression of T1D. In non-obese diabetic (NOD) mice, we found that aberrant reduction of GPR41 expression in the pancreas and colons was associated with the development of T1D. GPR41-deficient (Gpr41-/-) mice displayed significantly exacerbated streptozotocin (STZ)-induced T1D compared to wild-type mice. Furthermore, Gpr41-/- mice showed enhanced gut immune dysregulation and increased migration of gut-primed IFN-γ+ T cells to the pancreas. In bone marrow-derived dendritic cells from Gpr41-/- mice, the expression of suppressor of cytokine signaling 3 (SOCS) was significantly inhibited, while the phosphorylation of STAT3 was significantly increased, thus promoting dendritic cell (DC) maturation. Furthermore, adoptive transfer of bone marrow-derived dendritic cells (BMDC) from Gpr41-/- mice accelerated T1D in irradiated NOD mice. We conclude that GPR41 is essential for maintaining intestinal and pancreatic immune homeostasis and acts as a negative regulator of DC maturation in T1D. GPR41 may be a potential therapeutic target for T1D.


Sujet(s)
Cellules dendritiques , Diabète expérimental , Diabète de type 1 , Souris de lignée NOD , Souris knockout , Récepteurs couplés aux protéines G , Streptozocine , Animaux , Diabète de type 1/immunologie , Diabète de type 1/métabolisme , Récepteurs couplés aux protéines G/déficit , Récepteurs couplés aux protéines G/génétique , Récepteurs couplés aux protéines G/métabolisme , Cellules dendritiques/immunologie , Cellules dendritiques/métabolisme , Souris , Diabète expérimental/métabolisme , Diabète expérimental/immunologie , Souris de lignée C57BL , Facteur de transcription STAT-3/métabolisme , Protéine-3 suppressive de la signalisation des cytokine/métabolisme , Protéine-3 suppressive de la signalisation des cytokine/génétique , Interféron gamma/métabolisme , Pancréas/métabolisme , Pancréas/anatomopathologie , Pancréas/immunologie , Mâle , Femelle , Microbiome gastro-intestinal
15.
Mol Ther ; 32(5): 1425-1444, 2024 May 01.
Article de Anglais | MEDLINE | ID: mdl-38504518

RÉSUMÉ

Pathological ocular angiogenesis has long been associated with myeloid cell activation. However, the precise cellular and molecular mechanisms governing the intricate crosstalk between the immune system and vascular changes during ocular neovascularization formation remain elusive. In this study, we demonstrated that the absence of the suppressor of cytokine signaling 3 (SOCS3) in myeloid cells led to a substantial accumulation of microglia and macrophage subsets during the neovascularization process. Our single-cell RNA sequencing data analysis revealed a remarkable increase in the expression of the secreted phosphoprotein 1 (Spp1) gene within these microglia and macrophages, identifying subsets of Spp1-expressing microglia and macrophages during neovascularization formation in angiogenesis mouse models. Notably, the number of Spp1-expressing microglia and macrophages exhibited further elevation during neovascularization in mice lacking myeloid SOCS3. Moreover, our investigation unveiled the Spp1 gene as a direct transcriptional target gene of signal transducer and activator of transcription 3. Importantly, pharmaceutical activation of SOCS3 or blocking of SPP1 resulted in a significant reduction in pathological neovascularization. In conclusion, our study highlights the pivotal role of the SOCS3/STAT3/SPP1 axis in the regulation of pathological retinal angiogenesis.


Sujet(s)
Modèles animaux de maladie humaine , Macrophages , Microglie , Ostéopontine , Néovascularisation rétinienne , Facteur de transcription STAT-3 , Protéine-3 suppressive de la signalisation des cytokine , Animaux , Protéine-3 suppressive de la signalisation des cytokine/métabolisme , Protéine-3 suppressive de la signalisation des cytokine/génétique , Macrophages/métabolisme , Souris , Microglie/métabolisme , Néovascularisation rétinienne/métabolisme , Néovascularisation rétinienne/anatomopathologie , Néovascularisation rétinienne/génétique , Néovascularisation rétinienne/étiologie , Ostéopontine/métabolisme , Ostéopontine/génétique , Facteur de transcription STAT-3/métabolisme , Facteur de transcription STAT-3/génétique , Régulation de l'expression des gènes , Transduction du signal , Souris knockout , Néovascularisation pathologique/métabolisme , Néovascularisation pathologique/génétique ,
16.
Exp Mol Med ; 56(3): 711-720, 2024 Mar.
Article de Anglais | MEDLINE | ID: mdl-38486105

RÉSUMÉ

Protein arginine methyltransferases (PRMTs) modulate diverse cellular processes, including stress responses. The present study explored the role of Prmt7 in protecting against menopause-associated cardiomyopathy. Mice with cardiac-specific Prmt7 ablation (cKO) exhibited sex-specific cardiomyopathy. Male cKO mice exhibited impaired cardiac function, myocardial hypertrophy, and interstitial fibrosis associated with increased oxidative stress. Interestingly, female cKO mice predominantly exhibited comparable phenotypes only after menopause or ovariectomy (OVX). Prmt7 inhibition in cardiomyocytes exacerbated doxorubicin (DOX)-induced oxidative stress and DNA double-strand breaks, along with apoptosis-related protein expression. Treatment with 17ß-estradiol (E2) attenuated the DOX-induced decrease in Prmt7 expression in cardiomyocytes, and Prmt7 depletion abrogated the protective effect of E2 against DOX-induced cardiotoxicity. Transcriptome analysis of ovariectomized wild-type (WT) or cKO hearts and mechanical analysis of Prmt7-deficient cardiomyocytes demonstrated that Prmt7 is required for the control of the JAK/STAT signaling pathway by regulating the expression of suppressor of cytokine signaling 3 (Socs3), which is a negative feedback inhibitor of the JAK/STAT signaling pathway. These data indicate that Prmt7 has a sex-specific cardioprotective effect by regulating the JAK/STAT signaling pathway and, ultimately, may be a potential therapeutic tool for heart failure treatment depending on sex.


Sujet(s)
Cardiomyopathies , Post-ménopause , Protein-arginine N-methyltransferases , Animaux , Femelle , Mâle , Souris , Apoptose/génétique , Cardiomyopathies/génétique , Cardiomyopathies/métabolisme , Doxorubicine/pharmacologie , Myocytes cardiaques/métabolisme , Post-ménopause/génétique , Transduction du signal , Protéine-3 suppressive de la signalisation des cytokine/métabolisme , Protein-arginine N-methyltransferases/génétique , Protein-arginine N-methyltransferases/métabolisme , Janus kinases/métabolisme , Facteurs de transcription STAT/métabolisme
17.
Int Immunopharmacol ; 129: 111576, 2024 Mar 10.
Article de Anglais | MEDLINE | ID: mdl-38350353

RÉSUMÉ

Cyanogramide (AC14), a novel alkaloid, isolated from the fermentation broth of the marine-derived Actinoalloteichus cyanogriseus. However, the exact role of AC14 in inflammatory bowel disease (IBD) is poorly understood. Our results demonstrated that AC14 exhibited significant inhibition of IL-6 release in THP-1 cells and a "Caco-2/THP-1" coculture system after stimulation with LPS for 24 h. However, no significant effect on TNF-α production was observed. Furthermore, in 2.5 % DSS-induced colitis mice, AC14 treatment led to improvement in body weight, colon length, and intestine mucosal barrier integrity. AC14 also suppressed serum IL-6 production and modulated dysregulated microbiota in the mice. Mechanistically, AC14 was found to inhibit the phosphorylation of Janus kinase (JAK) 2 and signal transducers and activators of transcription (STAT) 3, while simultaneously elevating the expression of suppressor of cytokine signaling (SOCS) 3, both in vivo and in vitro. These findings suggest that AC14 exerts its suppressive effects on IL-6 production in DSS-induced IBD mice through the JAK2-STAT3-SOCS3 signaling pathway. Our study highlights the potential of AC14 as a therapeutic agent for the treatment of IBD.


Sujet(s)
Alcaloïdes , Antinéoplasiques , Maladies inflammatoires intestinales , Porifera , Humains , Souris , Animaux , Interleukine-6/métabolisme , Protéine-3 suppressive de la signalisation des cytokine/génétique , Protéine-3 suppressive de la signalisation des cytokine/métabolisme , Cellules Caco-2 , Protéines SOCS/métabolisme , Maladies inflammatoires intestinales/induit chimiquement , Maladies inflammatoires intestinales/traitement médicamenteux , Kinase Janus-2/métabolisme , Porifera/métabolisme , Alcaloïdes/usage thérapeutique , Facteur de transcription STAT-3/métabolisme
18.
J Ovarian Res ; 17(1): 29, 2024 Feb 01.
Article de Anglais | MEDLINE | ID: mdl-38302986

RÉSUMÉ

BACKGROUND: Polycystic ovary syndrome (PCOS) is a frequent and complicated endocrine disease that remains a major reason for infertility. Bushenhuoluo Decotion (BSHLD) has been validated to exhibit curative effects on PCOS. This study was aimed to explore the potential mechanism underlying the therapeutic action of BSHLD. METHODS: PCOS rat model was induced by dehydroepiandrosterone (DHEA). Serum hormone and cytokines levels and ovarian pathological alterations were measured to assess ovarian function. Exosomes (Exos) were identified by Transmission electron microscopy and Nanoparticle Tracking Analysis. RT-qPCR, Western blotting, immunohistochemical staining, and immunofluorescence staining were performed to detect molecule expressions. Proliferation and pyroptosis of granulosa cells (GCs) were evaluated by CCK-8 and flow cytometry, respectively. The binding relationship between miR-30a-5p and suppressor of cytokine signaling 3 (SOCS3) was verified by dual luciferase reporter and RIP assays. RESULTS: BSHLD treatment improved serum hormone abnormality, insulin sensitivity, and ovarian morphologic changes of PCOS rats. Moreover, BSHLD treatment restrained the excessive autophagy and pyroptosis in ovarian tissues of PCOS rats. Moreover, BSHLD reduced the expression of miR-30a-5p in serum, serum-derived Exos, and ovarian tissues, thus inhibiting autophagy and NLRP3-mediated pyroptosis in GCs. Mechanistically, SOCS3 was proved as a target of miR-30a-5p and could activate mTOR/P70S6K pathway to repress autophagy. The inhibitory effect of miR-30a-5p deficiency on autophagy and pyroptosis of GCs was attenuated by rapamycin. CONCLUSION: Collectively, BSHLD suppressed autophagy and pyroptosis to improve POCS by regulating exosomal miR-30a-5p/SOCS3/mTOR signaling.


Sujet(s)
Médicaments issus de plantes chinoises , microARN , Extraits de plantes , Syndrome des ovaires polykystiques , Animaux , Femelle , Humains , Rats , Autophagie , Hormones , microARN/génétique , microARN/métabolisme , Protéine-3 de la famille des NLR contenant un domaine pyrine/métabolisme , Syndrome des ovaires polykystiques/anatomopathologie , Pyroptose , Protéine-3 suppressive de la signalisation des cytokine/génétique , Protéine-3 suppressive de la signalisation des cytokine/métabolisme , Sérine-thréonine kinases TOR/métabolisme , Extraits de plantes/usage thérapeutique , Médicaments issus de plantes chinoises/usage thérapeutique
19.
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 , Protéines SOCS , Humains , Souris , Animaux , Protéines SOCS/génétique , Protéines SOCS/métabolisme , Cytokines/génétique , Cytokines/métabolisme , Cicatrisation de plaie , 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
20.
Joint Bone Spine ; 91(3): 105698, 2024 May.
Article de Anglais | MEDLINE | ID: mdl-38309518

RÉSUMÉ

OBJECTIVE: Hyperuricaemia is necessary for gout. High urate concentrations have been linked to inflammation in mononuclear cells. Here, we explore the role of the suppressor of cytokine signaling 3 (SOCS3) in urate-induced inflammation. METHODS: Peripheral blood mononuclear cells (PBMCs) from gout patients, hyperuricemic and normouricemic individuals were cultured for 24h with varying concentrations of soluble urate, followed by 24h restimulation with lipopolysaccharides (LPS)±monosodium urate (MSU) crystals. Transcriptomic profiling was performed using RNA-Sequencing. DNA methylation was assessed using Illumina Infinium® MethylationEPIC BeadChip system (EPIC array). Phosphorylation of signal transducer and activator of transcription 3 (STAT3) was determined by flow cytometry. Cytokine responses were also assessed in PBMCs from patients with JAK2 V617F tyrosine kinase mutation. RESULTS: PBMCs pre-treated with urate produced more interleukin-1beta (IL-1ß) and interleukin-6 (IL-6) and less interleukin-1 receptor anatagonist (IL-1Ra) after LPS simulation. In vitro, urate treatment enhanced SOCS3 expression in control monocytes but no DNA methylation changes were observed at the SOCS3 gene. A dose-dependent reduction in phosphorylated STAT3 concomitant with a decrease in IL-1Ra was observed with increasing concentrations of urate. PBMCs with constitutively activated STAT3 (JAK2 V617F mutation) could not be primed by urate. CONCLUSION: In vitro, urate exposure increased SOCS3 expression, while urate priming, and subsequent stimulation resulted in decreased STAT3 phosphorylation and IL-1Ra production. There was no evidence that DNA methylation constitutes a regulatory mechanism of SOCS3. Elevated SOCS3 and reduced pSTAT3 could play a role in urate-induced hyperinflammation since urate priming had no effect in PBMCs from patients with constitutively activated STAT3.


Sujet(s)
Cytokines , Goutte , Facteur de transcription STAT-3 , Protéine-3 suppressive de la signalisation des cytokine , Acide urique , Humains , Protéine-3 suppressive de la signalisation des cytokine/métabolisme , Protéine-3 suppressive de la signalisation des cytokine/génétique , Acide urique/pharmacologie , Facteur de transcription STAT-3/métabolisme , Cytokines/métabolisme , Goutte/génétique , Goutte/métabolisme , Cellules cultivées , Mâle , Cellules myéloïdes/métabolisme , Cellules myéloïdes/effets des médicaments et des substances chimiques , Agranulocytes/métabolisme , Agranulocytes/effets des médicaments et des substances chimiques , Hyperuricémie/métabolisme , Femelle , Adulte d'âge moyen , Méthylation de l'ADN , Kinase Janus-2/métabolisme
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...