Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 3.616
Filtrar
1.
Eur Rev Med Pharmacol Sci ; 28(9): 3290, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38766781

RESUMEN

Correction to: Eur Rev Med Pharmacol Sci 2015; 19 (17): 3208-3217. PMID: 26400524-published online on September 14, 2015. After publication, a reader brought to our attention a mistake in Figure 4. The journal found that Figure 3 was mistakenly inserted twice in the galley proof, resulting in the publication of the same figure for Figures 3 and 4. The publisher is, therefore, substituting Figure 4 with the correct figure provided at the time of submission as follows: There are amendments to this paper. The Publisher apologizes for any inconvenience this may cause. https://www.europeanreview.org/article/9429.


Asunto(s)
Carcinoma Hepatocelular , Neoplasias Hepáticas , MicroARNs , Proteínas Supresoras de la Señalización de Citocinas , MicroARNs/metabolismo , MicroARNs/genética , Carcinoma Hepatocelular/patología , Carcinoma Hepatocelular/metabolismo , Humanos , Neoplasias Hepáticas/patología , Neoplasias Hepáticas/metabolismo , Neoplasias Hepáticas/genética , Proteínas Supresoras de la Señalización de Citocinas/metabolismo , Proteínas Supresoras de la Señalización de Citocinas/genética , Proliferación Celular/efectos de los fármacos , Línea Celular Tumoral , Invasividad Neoplásica
2.
Redox Biol ; 71: 103100, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38484644

RESUMEN

Th2-high asthma is characterized by elevated levels of type 2 cytokines, such as interleukin 13 (IL-13), and its prevalence has been increasing worldwide. Ferroptosis, a recently discovered type of programmed cell death, is involved in the pathological process of Th2-high asthma; however, the underlying mechanisms remain incompletely understood. In this study, we demonstrated that the serum level of malondialdehyde (MDA), an index of lipid peroxidation, positively correlated with IL-13 level and negatively correlated with the predicted forced expiratory volume in 1 s (FEV1%) in asthmatics. Furthermore, we showed that IL-13 facilitates ferroptosis by upregulating of suppressor of cytokine signaling 1 (SOCS1) through analyzing immortalized airway epithelial cells, human airway organoids, and the ovalbumin (OVA)-challenged asthma model. We identified that signal transducer and activator of transcription 6 (STAT6) promotes the transcription of SOCS1 upon IL-13 stimulation. Moreover, SOCS1, an E3 ubiquitin ligase, was found to bind to solute carrier family 7 member 11 (SLC7A11) and catalyze its ubiquitinated degradation, thereby promoting ferroptosis in airway epithelial cells. Last, we found that inhibiting SOCS1 can decrease ferroptosis in airway epithelial cells and alleviate airway hyperresponsiveness (AHR) in OVA-challenged wide-type mice, while SOCS1 overexpression exacerbated the above in OVA-challenged IL-13-knockout mice. Our findings reveal that the IL-13/STAT6/SOCS1/SLC7A11 pathway is a novel molecular mechanism for ferroptosis in Th2-high asthma, confirming that targeting ferroptosis in airway epithelial cells is a potential therapeutic strategy for Th2-high asthma.


Asunto(s)
Asma , Interleucina-13 , Animales , Humanos , Ratones , Sistema de Transporte de Aminoácidos y+ , Asma/genética , Asma/metabolismo , Modelos Animales de Enfermedad , Células Epiteliales/metabolismo , Pulmón/metabolismo , Ratones Endogámicos BALB C , Ovalbúmina/metabolismo , Ovalbúmina/uso terapéutico , Proteína 1 Supresora de la Señalización de Citocinas/genética , Proteína 1 Supresora de la Señalización de Citocinas/metabolismo , Proteína 1 Supresora de la Señalización de Citocinas/uso terapéutico , Proteínas Supresoras de la Señalización de Citocinas/metabolismo , Células Th2/metabolismo , Células Th2/patología
3.
Cells ; 13(6)2024 Mar 13.
Artículo en Inglés | MEDLINE | ID: mdl-38534350

RESUMEN

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.


Asunto(s)
Dermatitis Atópica , Psoriasis , Humanos , Proteínas Supresoras de la Señalización de Citocinas/genética , Proteínas Supresoras de la Señalización de Citocinas/metabolismo , Transducción de Señal/genética , Citocinas/metabolismo , Inflamación
4.
Front Immunol ; 15: 1320077, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38533493

RESUMEN

Background: The family of Suppressor of Cytokine Signaling (SOCS) acts as a controller of the duration and intensity of cytokine function by negatively regulating the JAK-STAT signaling pathway. SOCS' role in inflammatory diseases in animal models is well demonstrated. However, its role in the development of human disease is still under investigation. SOCS3 plays an important role in tumor development where its downregulation has been implicated in the pathogenesis of various solid tumors such as triple-negative breast cancer. Aim: The aim of this work was to study (1) the expression of SOCS3 in smokers' lungs and its relation to the degree of inflammation and (2) SOCS3 regulation by microRNA (miRNA) in alveolar-macrophage (AM)-derived extracellular vesicles (EVs) in bronchoalveolar lavage (BAL). Methods: Group A: 35 smokers' [19 with COPD (SC) and 16 without COPD (S)] and 9 nonsmokers (NS); SOCS3, TNFα in AM, and CD8+ T cells were quantified by immunohistochemistry, in lung tissue. Group B: additional 9 SC, 11 S, and 5 NS; AM-EVs expressing SOCS3 (CD14+SOCS3+) and SOCS3 suppressors miRNA-19a-3p and 221-3p in EVs were quantified by flow cytometry and PCR, in BAL. Results: The percentage of SOCS3+ AM was higher in SC [68 (6.6-99)%] and S [48 (8-100)%] than in NS [9.6 (1.9-61)%; p = 0.002; p = 0.03] and correlated with % of TNFα+AM (r = 0.48; p = 0.0009) and CD8+ T cells (r = 0.44; p = 0.0029). In BAL, the CD14+SOCS3+ EVs/µL were increased in SC [33 (21-74)] compared to S [16 (8-37); p = 0.03] and NS [9 (7-21); p = 0.003]. Conversely, miRNA-19a-3p and miRNA-221-3p expression were increased in S when compared to SC [19 (2-53) vs. 3 (0.6-8); p = 0.03 and 3 (0.005-9.6) vs. 0.2 (0.08-0.7); p = 0.05]. Conclusions: The suppressor function of SOCS3 in COPD seems to be overridden by other factors and does not follow the animal-model paradigm. Expression of SOCS3 in BAL macrophage-derived EVs might be useful to assess the degree of inflammation and possible progression of COPD. Downregulation of SOCS3, by miRNA, in smokers without COPD might contribute to the risk of developing cancer in these patients.


Asunto(s)
MicroARNs , Enfermedad Pulmonar Obstructiva Crónica , Animales , Humanos , Líquido del Lavado Bronquioalveolar , Linfocitos T CD8-positivos/metabolismo , Citocinas/metabolismo , Inflamación , Enfermedad Pulmonar Obstructiva Crónica/metabolismo , Proteínas Supresoras de la Señalización de Citocinas/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo
5.
Front Immunol ; 15: 1362224, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38415248

RESUMEN

Suppressor of cytokine signaling 1 (SOCS1) is a potent regulator immune cell responses and a proven tumor suppressor. Inhibition of SOCS1 in T cells can boost antitumor immunity, whereas its loss in tumor cells increases tumor aggressivity. Investigations into the tumor suppression mechanisms so far focused on tumor cell-intrinsic functions of SOCS1. However, it is possible that SOCS1 expression in tumor cells also regulate antitumor immune responses in a cell-extrinsic manner via direct and indirect mechanisms. Here, we discuss the evidence supporting the latter, and its implications for antitumor immunity.


Asunto(s)
Neoplasias , Proteínas Supresoras de la Señalización de Citocinas , Humanos , Proteína 1 Supresora de la Señalización de Citocinas/genética , Proteína 1 Supresora de la Señalización de Citocinas/metabolismo , Proteínas Supresoras de la Señalización de Citocinas/metabolismo , Linfocitos T/metabolismo
6.
Int J Mol Sci ; 25(4)2024 Feb 14.
Artículo en Inglés | MEDLINE | ID: mdl-38396957

RESUMEN

This study aimed to identify and evaluate drug candidates targeting the kinase inhibitory region of suppressor of cytokine signaling (SOCS) 3 for the treatment of allergic rhinitis (AR). Utilizing an artificial intelligence (AI)-based new drug development platform, virtual screening was conducted to identify compounds inhibiting the SH2 domain binding of SOCS3. Luminescence assays assessed the ability of these compounds to restore JAK-2 activity diminished by SOCS3. Jurkat T and BEAS-2B cells were utilized to investigate changes in SOCS3 and STAT3 expression, along with STAT3 phosphorylation in response to the identified compounds. In an OVA-induced allergic rhinitis mouse model, we measured serum levels of total IgE and OVA-specific IgE, performed real-time PCR on nasal mucosa samples to quantify Th2 cytokines and IFN-γ expression, and conducted immunohistochemistry to analyze eosinophil levels. Screening identified 20 hit compounds with robust binding affinities. As the concentration of SOCS3 increased, a corresponding decrease in JAK2 activity was observed. Compounds 5 and 8 exhibited significant efficacy in restoring JAK2 activity without toxicity. Treatment with these compounds resulted in reduced SOCS3 expression and the reinstatement of STAT3 phosphorylation in Jurkat T and BEAS-2B cells. In the OVA-induced allergic rhinitis mouse model, compounds 5 and 8 effectively alleviated nasal symptoms and demonstrated lower levels of immune markers compared to the allergy group. This study underscores the promising nonclinical efficacy of compounds identified through the AI-based drug development platform. These findings introduce innovative strategies for the treatment of AR and highlight the potential therapeutic value of targeting SOCS3 in managing AR.


Asunto(s)
Inteligencia Artificial , Rinitis Alérgica , Ratones , Animales , Ovalbúmina , Mucosa Nasal/metabolismo , Citocinas/metabolismo , Proteínas Supresoras de la Señalización de Citocinas/metabolismo , Inmunoglobulina E/metabolismo , Ratones Endogámicos BALB C , Modelos Animales de Enfermedad
7.
Int Immunopharmacol ; 129: 111576, 2024 Mar 10.
Artículo en Inglés | MEDLINE | ID: mdl-38350353

RESUMEN

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.


Asunto(s)
Alcaloides , Antineoplásicos , Enfermedades Inflamatorias del Intestino , Poríferos , Humanos , Ratones , Animales , Interleucina-6/metabolismo , Proteína 3 Supresora de la Señalización de Citocinas/genética , Proteína 3 Supresora de la Señalización de Citocinas/metabolismo , Células CACO-2 , Proteínas Supresoras de la Señalización de Citocinas/metabolismo , Enfermedades Inflamatorias del Intestino/inducido químicamente , Enfermedades Inflamatorias del Intestino/tratamiento farmacológico , Janus Quinasa 2/metabolismo , Poríferos/metabolismo , Alcaloides/uso terapéutico , Factor de Transcripción STAT3/metabolismo
8.
Int J Biol Macromol ; 262(Pt 1): 129876, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-38310055

RESUMEN

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.


Asunto(s)
Diabetes Mellitus , Proteínas Supresoras de la Señalización de Citocinas , Humanos , Ratones , Animales , Proteínas Supresoras de la Señalización de Citocinas/genética , Proteínas Supresoras de la Señalización de Citocinas/metabolismo , Citocinas/genética , Citocinas/metabolismo , Cicatrización de Heridas , Diabetes Mellitus/metabolismo , Macrófagos/metabolismo , ARN Mensajero/metabolismo , Proteína 3 Supresora de la Señalización de Citocinas/genética , Proteína 3 Supresora de la Señalización de Citocinas/metabolismo
9.
Int Immunopharmacol ; 130: 111638, 2024 Mar 30.
Artículo en Inglés | MEDLINE | ID: mdl-38373387

RESUMEN

L-arginine, as an essential substance of the immune system, plays a vital role in innate immunity. MiR155, a multi-functional microRNA, has gained importance as a regulator of homeostasis in immune cells. However, the immunoregulatory mechanism between L-arginine and miR155 in bacterial infections is unknown. Here, we investigated the potential role of miR155 in inflammation and the molecular regulatory mechanisms of L-arginine in Streptococcus uberis (S. uberis) infections. And we observed that miR155 was up-regulated after infection, accompanying the depletion of L-arginine, leading to metabolic disorders of amino acids and severe tissue damage. Mechanically, the upregulated miR155 mediated by the p65 protein played a pro-inflammatory role by suppressing the suppressor of cytokine signaling 6 (SOCS6)-mediated p65 ubiquitination and degradation. This culminated in a violently inflammatory response and tissue damage. Interestingly, a significant anti-inflammatory effect was revealed in L-arginine supplementation by reducing miR155 production via inhibiting p65. This work firstly uncovers the pro-inflammatory role of miR155 and an anti-inflammatory mechanism of L-arginine in S.uberis infection with a mouse mastitis model. Collectively, we provide new insights and strategies for the prevention and control of this important pathogen, which is of great significance for ensuring human food health and safety.


Asunto(s)
Arginina , Mastitis , MicroARNs , Infecciones Estreptocócicas , Animales , Femenino , Humanos , Ratones , Arginina/metabolismo , Inflamación/metabolismo , MicroARNs/genética , Infecciones Estreptocócicas/metabolismo , Streptococcus/fisiología , Proteínas Supresoras de la Señalización de Citocinas/metabolismo , Mastitis/inmunología , Mastitis/metabolismo
10.
mBio ; 15(3): e0321323, 2024 Mar 13.
Artículo en Inglés | MEDLINE | ID: mdl-38376239

RESUMEN

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.


Asunto(s)
Interferón Tipo I , Peptidasa Específica de Ubiquitina 7/genética , Peptidasa Específica de Ubiquitina 7/metabolismo , Proteína 1 Supresora de la Señalización de Citocinas/metabolismo , Proteínas Supresoras de la Señalización de Citocinas/genética , Proteínas Supresoras de la Señalización de Citocinas/metabolismo , Inmunidad Innata , ARN
11.
Cytokine ; 176: 156507, 2024 04.
Artículo en Inglés | MEDLINE | ID: mdl-38244240

RESUMEN

Endothelial cell injury and mitochondrial dysfunction are crucial events during coronary artery disease (CAD). Suppressor of cytokine signaling-1 (SOCS1) is a negative mediator for inflammation, but there are few reports regarding histone acetylation of SOCS1 in CAD. The aim of the current study is to examine the impact of SOCS1 in CAD patients and human umbilical vein endothelial cells (HUVECs). We enrolled patients with CAD and healthy volunteers. HUVECs treated with ox-LDL were used as in vitro model. This study showed that SOCS1 expression was decreased in patients with CAD and ox-LDL-stimulated HUVECs. Overexpressing SOCS1 ameliorated endothelial cell injury and mitochondrial dysfunction induced by ox-LDL in vitro. Moreover, EP300 promoted SOCS1 transcription through increasing the acetylation of SOCS1 and recruiting H3K27ac to the SOCS1 gene promoter in HUVECs induced by ox-LDL. Additionally, SOCS1 repressed JAK/STAT cascade in ox-LDL-stimulated HUVECs. Silencing of EP300 reversed endothelial cell injury and mitochondrial dysfunction ameliorated by overexpression of SOCS1 in ox-LDL-induced HUVECs. In summary, SOCS1 alleviated endothelial injury and mitochondrial dysfunction via enhancing H3K27ac acetylation by recruiting EP300 to promoter region and inhibiting JAK/STAT pathway. These results contribute to discover underlying diagnostic biomarkers and therapeutic targets for CAD.


Asunto(s)
Enfermedad de la Arteria Coronaria , Enfermedades Mitocondriales , Humanos , Histonas , Quinasas Janus , Enfermedad de la Arteria Coronaria/genética , Acetilación , Transducción de Señal , Factores de Transcripción STAT , Proteínas Supresoras de la Señalización de Citocinas , Células Endoteliales de la Vena Umbilical Humana , Regiones Promotoras Genéticas/genética , Proteína 1 Supresora de la Señalización de Citocinas/genética , Proteína p300 Asociada a E1A
12.
Cell Biol Toxicol ; 40(1): 3, 2024 01 25.
Artículo en Inglés | MEDLINE | ID: mdl-38267746

RESUMEN

OBJECTIVE: This study investigated the effect and mechanism of POU6F1 and lncRNA-CASC2 on ferroptosis of gastric cancer (GC) cells. METHODS: GC cells treated with erastin and RSL3 were detected for ferroptosis, reactive oxygen species (ROS) level, and cell viability. The expression levels of POU6F1, lncRNA-CASC2, SOCS2, and ferroptosis-related molecules (GPX4 and SLC7A11) were also measured. The regulations among POU6F1, lncRNA-CASC2, FMR1, SOCS2, and SLC7A11 were determined. Subcutaneous tumor models were established, in which the expressions of Ki-67, SOCS2, and GPX4 were detected by immunohistochemistry. RESULTS: GC patients with decreased expressions of POU6F1 and lncRNA-CASC2 had lower survival rate. Overexpression of POU6F1 or lncRNA-CASC2 decreased cell proliferation and GSH levels in GC cells, in addition to increasing total iron, Fe2+, MDA, and ROS levels. POU6F1 directly binds to the lncRNA-CASC2 promoter to promote its transcription. LncRNA-CASC2 can target FMR1 and increase SOCS2 mRNA stability to promote SLC7A11 ubiquitination degradation and activate ferroptosis signaling. Knockdown of SOCS2 inhibited the ferroptosis sensitivity of GC cells and reversed the effects of POU6F1 and lncRNA-CASC2 overexpression on ferroptosis in GC cells. CONCLUSION: Transcription factor POU6F1 binds directly to the lncRNA-CASC2 promoter to promote its expression, while upregulated lncRNA-CASC2 increases SOCS2 stability and expression by targeting FMR1, thereby inhibiting SLC7A11 signaling to promote ferroptosis in GC cells and inhibit GC progression.


Asunto(s)
Ferroptosis , ARN Largo no Codificante , Neoplasias Gástricas , Humanos , Sistema de Transporte de Aminoácidos y+/genética , Proteína de la Discapacidad Intelectual del Síndrome del Cromosoma X Frágil , Factores del Dominio POU , Especies Reactivas de Oxígeno , ARN Largo no Codificante/genética , Transducción de Señal , Neoplasias Gástricas/genética , Proteínas Supresoras de la Señalización de Citocinas
13.
Nat Commun ; 15(1): 603, 2024 Jan 19.
Artículo en Inglés | MEDLINE | ID: mdl-38242867

RESUMEN

CD8+ T cells play an important role in anti-tumor immunity. Better understanding of their regulation could advance cancer immunotherapies. Here we identify, via stepwise CRISPR-based screening, that CUL5 is a negative regulator of the core signaling pathways of CD8+ T cells. Knocking out CUL5 in mouse CD8+ T cells significantly improves their tumor growth inhibiting ability, with significant proteomic alterations that broadly enhance TCR and cytokine signaling and their effector functions. Chemical inhibition of neddylation required by CUL5 activation, also enhances CD8 effector activities with CUL5 validated as a major target. Mechanistically, CUL5, which is upregulated by TCR stimulation, interacts with the SOCS-box-containing protein PCMTD2 and inhibits TCR and IL2 signaling. Additionally, CTLA4 is markedly upregulated by CUL5 knockout, and its inactivation further enhances the anti-tumor effect of CUL5 KO. These results together reveal a negative regulatory mechanism for CD8+ T cells and have strong translational implications in cancer immunotherapy.


Asunto(s)
Linfocitos T CD8-positivos , Proteínas Cullin , Ubiquitina-Proteína Ligasas , Animales , Ratones , Linfocitos T CD8-positivos/metabolismo , Proteínas Cullin/genética , Proteínas Cullin/metabolismo , Neoplasias/tratamiento farmacológico , Neoplasias/metabolismo , Proteómica , Receptores de Antígenos de Linfocitos T/metabolismo , Transducción de Señal , Proteínas Supresoras de la Señalización de Citocinas/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo
14.
Biol Trace Elem Res ; 202(1): 258-267, 2024 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-36988786

RESUMEN

The exact molecular mechanism of arsenic-induced liver injury has not been fully elucidated. The aim of the study was to investigate the potential mechanism of NaAsO2-induced cytotoxicity in BRL-3A cells and to provide a basis for the mechanism of arsenic poisoning. BRL-3A cells were treated with different doses of NaAsO2, DNMT1 inhibitor (DC_517), TLR4 inhibitor (TAK-242), and transfection of SOCS1 plasmid. Cell activity, apoptosis, inflammation and protein expression of DNMT1, SOCS1, TLR4, MyD88, and NF-κB were detected by CCK8 assay, Annexin V-FITC and Western blot, respectively. With increasing NaAsO2 doses, BAX and caspase-3 expression increased, Bcl-2 expression decreased, pro-inflammatory factors TNF-α, IL-1ß, and IL-6 increased, and cell activity decreased causing increased apoptosis. When BRL-3A was intervened with 10, and 20 µmol/L NaAsO2, DNMT1 expression was elevated, SOCS1 expression was decreased, and TLR4, MyD88, p-IκBα/IκBα, and p-p65/p65 expression were elevated. After the combination of NaAsO2 and DC_517, compared to the NaAsO2 group, apoptosis and inflammation were attenuated, SOCS1 expression was elevated and TLR4, MyD88, p-IκBα/IκBα and p-p65/p65 expression was decreased. Apoptosis and inflammation were attenuated after co-treatment of SOCS1 high expression with NaAsO2 compared to the NaAsO2 group. In addition, TLR4, MyD88, p-IκBα/IκBα and p-p65/p65 expression was reduced. When NaAsO2 and TAK-242 were combined, apoptosis and inflammation were attenuated. Besides MyD88, p-IκBα/IκBα and p-p65/p65 expression was reduced compared to the NaAsO2 group. We found that NaAsO2 induce apoptosis and inflammation in BLR-3A cells, which may be related to inhibit SOCS1 through regulation of DNMT1 and thus activating the TLR4/MyD88/NF-κB signaling pathway.


Asunto(s)
Factor 88 de Diferenciación Mieloide , FN-kappa B , Humanos , FN-kappa B/metabolismo , Inhibidor NF-kappaB alfa/metabolismo , Factor 88 de Diferenciación Mieloide/genética , Factor 88 de Diferenciación Mieloide/metabolismo , Receptor Toll-Like 4/genética , Receptor Toll-Like 4/metabolismo , Transducción de Señal , Apoptosis , Proteínas Supresoras de la Señalización de Citocinas , Inflamación/inducido químicamente , Proteína 1 Supresora de la Señalización de Citocinas/genética , Proteína 1 Supresora de la Señalización de Citocinas/metabolismo
15.
Eur J Haematol ; 112(3): 439-449, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-37950514

RESUMEN

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.


Asunto(s)
Leucemia Mieloide Aguda , Transducción de Señal , Humanos , Línea Celular Tumoral , Proteínas Supresoras de la Señalización de Citocinas/genética , Metilación de ADN , Leucemia Mieloide Aguda/genética , Proteína 3 Supresora de la Señalización de Citocinas/genética , Proteína 3 Supresora de la Señalización de Citocinas/metabolismo
16.
Environ Toxicol ; 39(3): 1283-1293, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-37948135

RESUMEN

BACKGROUND: Nonsmall-cell lung cancer (NSCLC) has emerged as one of the dreadful lung cancers globally due to its increased mortality rates. Concerning chemotherapy, gefitinib has been employed as an effective first-line treatment drug for NSCLC. Nonetheless, the acquired resistance to gefitinib has remained one of the treatment obstacles of NSCLC, requiring improvement in the therapeutic effect of gefitinib. METHODS: Initially, reverse transcription-quantitative polymerase chain reaction (RT-qPCR), and Western blotting (WB) analyses were conducted to measure micro-ribose nucleic acid (miRNA, specifically miR-578) and suppressor of cytokine signaling 2 (SOCS2) levels in the clinical samples. Further, NSCLC cell lines resistance to gefitinib, established in vitro, were transfected by miR-578 inhibitor, miR-578 mimic, and si-SOCS2. Similarly, the xenograft mouse model in vivo was constructed to validate the reversing effect of miR-578. RESULTS: Our findings indicated the increased miR-578 expression levels in the gefitinib resistance group. Further, inhibiting the miR-578 expression substantially reversed the gefitinib resistance. In addition, the miR-578 effect was modulated via the SOCS2 expression level. The decreased gefitinib resistance effect of miR-578 was weakened by inhibiting the SOCS2 expression. CONCLUSION: These findings demonstrated that miR-578 effectively abolished gefitinib resistance by regulating the SOCS2 expression within NSCLC cells in vitro and in vivo. Together, these results will undoubtedly support a reference to provide potential molecular therapeutic targets and clinical treatments for treating NSCLC patients.


Asunto(s)
Antineoplásicos , Carcinoma de Pulmón de Células no Pequeñas , Neoplasias Pulmonares , MicroARNs , Humanos , Animales , Ratones , Gefitinib/farmacología , Gefitinib/uso terapéutico , Resistencia a Antineoplásicos , Línea Celular Tumoral , Carcinoma de Pulmón de Células no Pequeñas/genética , Neoplasias Pulmonares/genética , MicroARNs/genética , Antineoplásicos/farmacología , Proliferación Celular , Proteínas Supresoras de la Señalización de Citocinas
17.
Cytokine ; 174: 156461, 2024 02.
Artículo en Inglés | MEDLINE | ID: mdl-38065046

RESUMEN

Establishing a balance between Th1 and Th2 subsets and M1- and M2-type macrophages is essential for the control of Leishmania infection. The suppressors of cytokine secretion (SOCS) proteins, particularly SOCS1 and SOCS3, play a significant role in regulating cytokine-triggered signaling pathways, thereby impacting the macrophage-and effector T-cell mediated antileishmanial immune response. In addition to the pro-inflammatory cytokines, Leishmania-derived lipophosphoglycan (LPG) and CpG-DNA interact with TLR2 and TLR9 to trigger SOCS expression. The aberrant levels of SOCS1 and SOCS3 expression in Leishmania-infected macrophages impair macrophage-T-cell interaction perturbing the balance in macrophage subsets polarization. This hinders macrophage apoptosis and macrophage-mediated leishmanicidal activity, both support the establishment of infection and parasite replication. Furthermore, aberrant SOCS3 levels in T-cells disrupt Th1 differentiation and aid in parasite replication, lesion development, and pathological immune responses. Strategically, selective modulation of SOCS expression and function in immune effector cells may reduce parasite survival and prevent disease progression.


Asunto(s)
Leishmania , Proteínas Supresoras de la Señalización de Citocinas , Proteína 1 Supresora de la Señalización de Citocinas/metabolismo , Proteína 3 Supresora de la Señalización de Citocinas/metabolismo , Proteínas Supresoras de la Señalización de Citocinas/metabolismo , Citocinas/metabolismo , Inmunidad
18.
Cancer Sci ; 115(1): 155-169, 2024 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-37972389

RESUMEN

Cancer-associated fibroblasts (CAFs), as important components of the tumor microenvironment, can regulate intercellular communication and tumor development by secreting extracellular vesicles (EVs). However, the role of CAF-derived EVs in ovarian cancer has not been fully elucidated. Here, using an EV-microRNA sequencing analysis, we reveal specific overexpression of microRNA (miR)-296-3p in activated CAF-derived EVs, which can be transferred to tumor cells to regulate the malignant phenotypes of ovarian cancer cells. Moreover, overexpression of miR-296-3p significantly promotes the proliferation, migration, invasion, and drug resistance of ovarian cancer cells in vitro, as well as tumor growth in vivo, while its inhibition has the opposite effects. Further mechanistic studies reveal that miR-296-3p promotes ovarian cancer progression by directly targeting PTEN and SOCS6 and activating AKT and STAT3 signaling pathways. Importantly, increased expression of miR-296-3p encapsulated in plasma EVs is closely correlated with tumorigenesis and chemoresistance in patients with ovarian cancer. Our results highlight the cancer-promoting role of CAF-derived EVs carrying miR-296-3p in ovarian cancer progression for the first time, and suggest that miR-296-3p encapsulated in CAF-derived EVs could be a diagnostic biomarker and therapeutic target for ovarian cancer.


Asunto(s)
Fibroblastos Asociados al Cáncer , Vesículas Extracelulares , MicroARNs , Neoplasias Ováricas , Humanos , Femenino , Proteínas Proto-Oncogénicas c-akt , Neoplasias Ováricas/genética , Vesículas Extracelulares/genética , MicroARNs/genética , Proliferación Celular/genética , Microambiente Tumoral/genética , Proteínas Supresoras de la Señalización de Citocinas , Fosfohidrolasa PTEN/genética , Factor de Transcripción STAT3/genética
19.
J Nutr Biochem ; 123: 109512, 2024 01.
Artículo en Inglés | MEDLINE | ID: mdl-37907171

RESUMEN

Long-term consumption of a high-fat diet (HFD) disrupts energy homeostasis and leads to weight gain. The fat mass and obesity-associated (FTO) gene has been consistently identified to be associated with HFD-induced obesity. The hypothalamus is crucial for regulating energy balance, and HFD-induced hypothalamic leptin resistance contributes to obesity. FTO, an N6-methyladenosine (m6A) RNA methylation regulator, may be a key mediator of leptin resistance. However, the exact mechanisms remain unclear. Therefore, the present study aims to investigate the association between FTO and leptin resistance. After HFD or standard diet (SD) feeding in male mice for 22 weeks, m6A-sequencing and western blotting assays were used to identify target genes and assess protein level, and molecular interaction changes. CRISPR/Cas9 gene knockout system was employed to investigate the potential function of FTO in leptin resistance and obesity. Our data showed that chemokine (C-X3-C motif) ligand 1 (CX3CL1) was a direct downstream target of FTO-mediated m6A modification. Furthermore, upregulation of FTO/CX3CL1 and suppressor of cytokine signaling 3 (SOCS3) in the hypothalamus impaired leptin-signal transducer and activator of transcription 3 signaling, resulting in leptin resistance and obesity. Compared to wild-type (WT) mice, FTO deficiency in leptin receptor-expressing neurons of the hypothalamus significantly inhibited the upregulation of CX3CL1 and SOCS3, and partially ameliorating leptin resistance under HFD conditions. Our findings reveal that FTO involved in the hypothalamic leptin resistance and provides novel insight into the function of FTO in the contribution to hypothalamic leptin resistance and obesity.


Asunto(s)
Dieta Alta en Grasa , Leptina , Animales , Masculino , Ratones , Dioxigenasa FTO Dependiente de Alfa-Cetoglutarato/genética , Dioxigenasa FTO Dependiente de Alfa-Cetoglutarato/metabolismo , Quimiocina CX3CL1/metabolismo , Dieta Alta en Grasa/efectos adversos , Hipotálamo/metabolismo , Leptina/metabolismo , Ratones Endogámicos C57BL , Obesidad/genética , Obesidad/metabolismo , Proteínas Supresoras de la Señalización de Citocinas/genética
20.
Geroscience ; 46(2): 2583-2604, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38103096

RESUMEN

DNA methylation (DNAm) clocks hold promise for measuring biological age, useful for guiding clinical interventions and forensic identification. This study compared the commonly used DNAm clocks, using DNA methylation and SNP data generated from nearly 1000 human blood or buccal swab samples. We evaluated different preprocessing methods for age estimation, investigated the association of epigenetic age acceleration (EAA) with various lifestyle and sociodemographic factors, and undertook a series of novel genome-wide association analyses for different EAA measures to find associated genetic variants. Our results highlighted the Skin&Blood clock with ssNoob normalization as the most accurate predictor of chronological age. We provided novel evidence for an association between the practice of yoga and a reduction in the pace of aging (DunedinPACE). Increased sleep and physical activity were associated with lower mortality risk score (MRS) in our dataset. University degree, vegetable consumption, and coffee intake were associated with reduced levels of epigenetic aging, whereas smoking, higher BMI, meat consumption, and manual occupation correlated well with faster epigenetic aging, with FitAge, GrimAge, and DunedinPACE clocks showing the most robust associations. In addition, we found a novel association signal for SOCS2 rs73218878 (p = 2.87 × 10-8) and accelerated GrimAge. Our study emphasizes the importance of an optimized DNAm analysis workflow for accurate estimation of epigenetic age, which may influence downstream analyses. The results support the influence of genetic background on EAA. The associated SOCS2 is a member of the suppressor of cytokine signaling family known for its role in human longevity. The reported association between various risk factors and EAA has practical implications for the development of health programs to improve quality of life and reduce premature mortality associated with age-related diseases.


Asunto(s)
Yoga , Humanos , Café , Estudio de Asociación del Genoma Completo , Calidad de Vida , Envejecimiento/genética , Sueño/genética , Carne , Epigénesis Genética , Proteínas Supresoras de la Señalización de Citocinas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...