Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
1.
Mol Biol Rep ; 49(8): 7783-7792, 2022 Aug.
Article in English | MEDLINE | ID: mdl-35733067

ABSTRACT

BACKGROUND: Oral lichen planus (OLP) is a chronic inflammatory oral mucosal disease. Cytokines are closely associated with OLP development. In addition to immune cells, fibroblasts have been reported to induce regional inflammation. MicroRNA(miR)-155-5p is reportedly increased significantly in OLP and is known to regulate inflammation. This study aimed to investigate the role of miR-155-5p in fibroblasts of OLP lesions. METHODS AND RESULTS: Normal mucosal fibroblasts (NFs) and OLP associated-fibroblasts (OLP AFs) were isolated from the oral mucosa of 15 healthy controls and 30 OLP patients. We detected the expression of miR-155-5p and fibroblast activation protein alpha (FAP-α) using quantitative RT-PCR and analyzed their correlation. Interleukin (IL)-6 and IL-8 levels were determined using ELISA. Expression of suppressor of cytokine signaling (SOCS) 1 was analyzed by western blotting. A dual-luciferase reporter assay was performed to investigate the interaction between miR-155-5p and SOCS1. MiR-155-5p and FAP-α were significantly increased and positively correlated in OLP AFs. Overexpression of miR-155-5p in OLP AFs augmented IL-6 and IL-8 release and decreased SOCS1 expression, whereas knockdown of miR-155-5p in OLP AFs decreased IL-6 and IL-8 release. The expression of SOCS1 was downregulated in OLP AFs, and SOCS1 silencing augmented IL-6 and IL-8 production in OLP AFs. Furthermore, miR-155-5p inhibited SOCS1 expression by directly targeting its 3'-UTR in OLP AFs. CONCLUSIONS: MiR-155-5p regulates the secretion of IL-6 and IL-8 by downregulating the expression of SOCS1 in activated OLP AFs. Our results provide novel insights into the pathogenesis of OLP and identify a potential new target for OLP therapy.


Subject(s)
Inflammation/metabolism , Lichen Planus, Oral , MicroRNAs/metabolism , Suppressor of Cytokine Signaling 1 Protein/metabolism , Fibroblasts/metabolism , Humans , Interleukin-6/genetics , Interleukin-6/metabolism , Interleukin-8/genetics , Lichen Planus, Oral/metabolism , Lichen Planus, Oral/pathology , Phenotype , Suppressor of Cytokine Signaling 1 Protein/antagonists & inhibitors , Suppressor of Cytokine Signaling Proteins/genetics
2.
Eur J Med Chem ; 221: 113547, 2021 Oct 05.
Article in English | MEDLINE | ID: mdl-34023736

ABSTRACT

Herein we investigated the structural and cellular effects ensuing from the cyclization of a potent inhibitor of JAK2 as mimetic of SOCS1 protein, named PS5. The introduction of un-natural residues and a lactam internal bridge, within SOCS1-KIR motif, produced candidates that showed high affinity toward JAK2 catalytic domain. By combining CD, NMR and computational studies, we obtained valuable models of the interactions of two peptidomimetics of SOCS1 to deepen their functional behaviors. Notably, when assayed for their biological cell responses mimicking SOCS1 activity, the internal cyclic PS5 analogues demonstrated able to inhibit JAK-mediated tyrosine phosphorylation of STAT1 and to reduce cytokine-induced proinflammatory gene expression, oxidative stress generation and cell migration. The present study well inserts in the field of low-molecular-weight proteomimetics with improved longtime cellular effects and adds a new piece to the puzzled way for the conversion of bioactive peptides into drugs.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Peptidomimetics/pharmacology , Protein Kinase Inhibitors/pharmacology , Suppressor of Cytokine Signaling 1 Protein/antagonists & inhibitors , Animals , Anti-Inflammatory Agents/chemical synthesis , Anti-Inflammatory Agents/chemistry , Cells, Cultured , Dose-Response Relationship, Drug , Mice , Molecular Structure , Peptidomimetics/chemical synthesis , Peptidomimetics/chemistry , Protein Kinase Inhibitors/chemical synthesis , Protein Kinase Inhibitors/chemistry , Structure-Activity Relationship , Suppressor of Cytokine Signaling 1 Protein/metabolism
3.
Life Sci ; 271: 119197, 2021 Apr 15.
Article in English | MEDLINE | ID: mdl-33577847

ABSTRACT

AIMS: To investigate the improvement and mechanisms of silymarin on renal injury in mouse podocytes and streptozotocin (STZ)-induced diabetic nephropathy model (DN) rats. MAIN METHODS: Firstly, the effects of silymarin on the cell viability and cellular injury-related indicators of high-glucose incubated mouse podocytes MPC-5 were assessed by CCK-8 and western blotting (WB) methods, respectively. The STZ-induced diabetic rats with DN were treated with silymarin nanoliposomes at three doses for consecutive 8-week. General metabolic indicators, renal functions and lipid accumulation-related factors were all measured. The renal tissue sections were stained and observed via hematoxylin-eosin (H&E) staining method. Real-time RT-PCR and WB methods were utilized to measure the expression of JAK2/STAT3/SOCS1 and TGF-ß/Smad signaling pathway related factors. KEY FINDINGS: Silymarin significantly improve the high-glucose induced up-regulation of podoxin and nephrin, as well as the expression of inflammatory cytokines IL-6, ICAM-1 and TNF-α, and the cell survival rates were also significantly increased in a dose-dependent manner. Significant improvement on body weight/kidney ratio, renal functions and lipid profiles in renal tissues were observed in STZ-induced diabetic rats after chronic silymarin treatment. The H&E staining exhibited that the pathological damages in renal tissues were obviously improved. Moreover, silymarin nanoliposomes treatment notably suppressed expression levels of inflammation-related proteins as well as IL-6 and ICAM-1, and regulated JAK2/STAT3/SOCS1 and TGF-ß/Smad signaling pathway, thereby exhibited protective effects on kidney of DN model rats. SIGNIFICANCE: Silymarin nanoliposomes ameliorate STZ-induced kidney injury by improving oxidative stress, renal fibrosis, and co-inhibiting JAK2/STAT3/SOCS1 and TGF-ß/Smad signaling pathways in diabetic rats.


Subject(s)
Diabetic Nephropathies/drug therapy , Janus Kinase 2/antagonists & inhibitors , Nanoparticles/administration & dosage , STAT3 Transcription Factor/antagonists & inhibitors , Silymarin/administration & dosage , Suppressor of Cytokine Signaling 1 Protein/antagonists & inhibitors , Transforming Growth Factor beta/antagonists & inhibitors , Animals , Antioxidants/administration & dosage , Cell Line , Diabetes Mellitus, Experimental/drug therapy , Diabetes Mellitus, Experimental/metabolism , Diabetic Nephropathies/metabolism , Dose-Response Relationship, Drug , Female , Janus Kinase 2/metabolism , Liposomes , Male , Rats , Rats, Sprague-Dawley , STAT3 Transcription Factor/metabolism , Signal Transduction/drug effects , Signal Transduction/physiology , Smad Proteins/antagonists & inhibitors , Smad Proteins/metabolism , Suppressor of Cytokine Signaling 1 Protein/metabolism , Transforming Growth Factor beta/metabolism
4.
Virol J ; 17(1): 187, 2020 11 25.
Article in English | MEDLINE | ID: mdl-33239033

ABSTRACT

BACKGROUND: Respiratory syncytial virus (RSV) is a major cause of acute lower respiratory tract infection in infants, children, immunocompromised adults, and elderly individuals. Currently, there are few therapeutic options available to prevent RSV infection. The present study aimed to investigate the effects of luteolin on RSV replication and the related mechanisms. MATERIAL AND METHODS: We pretreated cells and mice with luteolin before infection with RSV, the virus titer, expressions of RSV-F, interferon (IFN)-stimulated genes (ISGs), and production of IFN-α and IFN-ß were determined by plaque assay, RT-qPCR, and ELISA, respectively. The activation of Janus kinase (JAK)-signal transducer and activator of transcription 1 (STAT1) signaling pathway was detected by Western blotting and luciferase assay. Proteins which negatively regulate STAT1 were determined by Western blotting. Then cells were transfected with suppressor of cytokine signaling 1 (SOCS1) plasmid and virus replication and ISGs expression were determined. Luciferase reporter assay and Western blotting were performed to detect the relationship between SOCS1 and miR-155. RESULTS: Luteolin inhibited RSV replication, as shown by the decreased viral titer and RSV-F mRNA expression both in vitro and in vivo. The antiviral activity of luteolin was attributed to the enhanced phosphorylation of STAT1, resulting in the increased production of ISGs. Further study showed that SOCS1 was downregulated by luteolin and SOCS1 is a direct target of microRNA-155 (miR-155). Inhibition of miR-155 rescued luteolin-mediated SOCS1 downregulation, whereas upregulation of miR-155 enhanced the inhibitory effect of luteolin. CONCLUSION: Luteolin inhibits RSV replication by regulating the miR-155/SOCS1/STAT1 signaling pathway.


Subject(s)
Antiviral Agents/pharmacology , Luteolin/pharmacology , MicroRNAs/antagonists & inhibitors , Respiratory Syncytial Virus, Human/drug effects , STAT1 Transcription Factor/antagonists & inhibitors , Signal Transduction/drug effects , Suppressor of Cytokine Signaling Proteins/antagonists & inhibitors , Virus Replication/drug effects , A549 Cells , Animals , Cell Line , Down-Regulation , Female , Fibroblasts/drug effects , Fibroblasts/virology , Gene Expression Regulation , Humans , Mice , Mice, Inbred BALB C , Phosphorylation/drug effects , Respiratory Syncytial Virus, Human/genetics , Respiratory Syncytial Virus, Human/metabolism , STAT1 Transcription Factor/genetics , Suppressor of Cytokine Signaling 1 Protein/antagonists & inhibitors
5.
Pak J Pharm Sci ; 33(1(Special)): 403-408, 2020 Jan.
Article in English | MEDLINE | ID: mdl-32173634

ABSTRACT

Inflammatory response that occur post-ischemia is a serious problem in the treatment of ischemic brain disease. MicroRNA-155 is a brain-specific or brain-enriched miRNA, which mediates inflammatory reactions in cerebral ischemic tissue by regulating inflammatory signal and the expression level of SOCS1. The present study was aimed to assess the effect of GuaLou GuiZhi Decoction (GLGZD) on miR-155 expression in activated microglia following inflammation and further explore the role of GLGZD on expression of the inflammation-related gene. BV2 cells were used to simulated by LPS to make the inflammatory model. Expression level of miR-155 was detected by Real-Time PCR. BV2 cells after simulated by LPS were then transfected with miR-155 mimic and its negative controls. Cytokines release were measured by corresponding purchased ELISA kits, respectively. Then target protein expression of miR-155 were detected by western blotting assay. After miRNA over expression transfections, expressions of inflammation-related factors, SOCS-1 and SAMD in BV2 cells after activation were measured by Western blot assay. Results showed that in BV2 cells after simulated by LPS, miR-155 was upregulated. The elevated miR-155 expression enhanced the inflammatory cytokine release. miR-155 directly target and negatively regulated SOCS-1 and SMAD-1 expression. Over expression of SOCS-1 and SMAD reduced inflammatory action that was enhanced by miR-155 mimic transfection. miR-155 was positively related with activation of NF-Ï°B signal pathways via SOCS-1 and SMAD. In conclusion, GuaLou GuiZhi Decoction (GLGZD) might exert its anti-inflammatory action by inhibiting the expression of miR-155, indicating that miR-155 may be used as a treatment target in clinical treatment with GuaLou GuiZhi Decoction (GLGZD) in ischemic brain.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Drugs, Chinese Herbal/pharmacology , Lipopolysaccharides/pharmacology , MicroRNAs/physiology , Microglia/drug effects , Animals , Brain Ischemia/drug therapy , Cells, Cultured , Cytokines/biosynthesis , Mice , MicroRNAs/antagonists & inhibitors , Microglia/physiology , Smad1 Protein/antagonists & inhibitors , Suppressor of Cytokine Signaling 1 Protein/antagonists & inhibitors
6.
J Immunol ; 203(1): 188-197, 2019 07 01.
Article in English | MEDLINE | ID: mdl-31101668

ABSTRACT

Suppressor of cytokine signaling 1 (SOCS1) plays a key role in the negative regulation of JAK/STAT signaling, which is involved in innate immunity and subsequent adaptive immunity. Bacillus Calmette-Guérin (BCG) induces upregulation of SOCS1 expression in host cells, which may lead to the suppression of immune responses by BCG via inhibition of the JAK/STAT signaling pathway. This might cause A reduction in the protective effect of a BCG vaccine. In the current study, we assessed the immune responses to and the protective efficacy of a recombinant BCG secreting a dominant negative mutant of the SOCS1 molecule (rBCG-SOCS1DN). C57BL/6 mice were immunized with rBCG-SOCS1DN or parental BCG Tokyo vaccine strain harboring an empty plasmid vector (rBCG-pSO). rBCG-SOCS1DN enhanced the activation of bone marrow-derived dendritic cells and the activation of T cells compared with those with rBCG-pSO. The amounts of IFN-γ, TNF-α, and IL-6 produced by splenocytes of rBCG-SOCS1DN-immunized mice were larger than those produced by splenocytes of rBCG-pSO-immunized mice. Moreover, the rBCG-SOCS1DN-immunized mice showed a substantial reduction in the number of CFU of Mycobacterium tuberculosis in the lungs and spleens compared with that in control BCG-immunized mice when the immunized mice were infected with a highly pathogenic M. tuberculosis strain by inhalation. These findings provide evidence for the possibility of rBCG-SOCS1DN being an effective M. tuberculosis vaccine with a novel concept of rBCG as a tool for immunomodulation in host cells.


Subject(s)
BCG Vaccine/immunology , Dendritic Cells/immunology , Mutation/genetics , Mycobacterium tuberculosis/physiology , Suppressor of Cytokine Signaling 1 Protein/genetics , T-Lymphocytes/immunology , Tuberculosis/immunology , Animals , BCG Vaccine/genetics , Colony Count, Microbial , Cytokines/metabolism , Disease Models, Animal , Female , Humans , Immunization , Lymphocyte Activation , Mice , Mice, Inbred C57BL , RAW 264.7 Cells , Signal Transduction , Suppressor of Cytokine Signaling 1 Protein/antagonists & inhibitors , Tuberculosis/prevention & control , Vaccines, Synthetic/genetics
7.
Oncol Rep ; 39(3): 1306-1312, 2018 Mar.
Article in English | MEDLINE | ID: mdl-29286121

ABSTRACT

The aim of the present study was to observe the effects of cytokine signaling suppressor 1 (SOCS1)-silenced dendritic cells (DCs) pulsed with epidermal growth factor receptor (EGFR) fusion protein on the activation of T lymphocyte and cytotoxic T-lymphocyte (CTL) activity against Hep-2 cells. DCs were derived from the medullary cells of mice and authenticated by flow cytometry (FCM). Recombinant glutathione-S-transferase (GST)-EGFR fusion protein was produced and purified. After being pulsed with it, DCs were modified by recombinant SOCS1-siRNA adenoviral to silence SOCS1 gene expression. The maturation of DCs was evaluated by FCM. The effects of modified DCs on T-cell proliferation were assessed by MTT assay. The killing effects against Hep-2 cells of CTL were assessed by lactate dehydrogenase (LDH) release assay. High-purity DCs from the medullary cells of mice were obtained. Compared with the control, EGFR-pulsed DCs displayed higher expression of cell surface molecules, including CD83, CD860 and HLA-DR. The MTT assay revealed that all of the EGFR-pulsed, SOCS1­silenced and EGFR-pulsed plus SOCS1-silenced DCs had an enhanced capacity to stimulate T-lymphocyte proliferation. As expected, EGFR-pulsed plus SOCS1-silenced DCs had the strongest effects on T-cell proliferation. The splenic T cells isolated from both EGFR-pulsed DC-immunized mice and EGFR-pulsed plus SOCS1-silenced DC-immunized mice enhanced the cytotoxicity against Hep-2 cells, while T cells from EGFR­pulsed plus SOCS1-silenced DC-immunized mice exhibited significantly higher cytotoxicity than those from EGFR-DC-immunized mice. The EGFR-pulsed SOCS1­siRNA-silenced DCs had the strongest effects on activation of T-cell proliferation and the CTL activity against Hep-2 cells.


Subject(s)
Cytotoxicity, Immunologic/immunology , Dendritic Cells/immunology , ErbB Receptors/metabolism , Laryngeal Neoplasms/pathology , Recombinant Fusion Proteins/metabolism , Suppressor of Cytokine Signaling 1 Protein/antagonists & inhibitors , T-Lymphocytes, Cytotoxic/immunology , Adenoviridae , Animals , Apoptosis , Cell Proliferation , Dendritic Cells/metabolism , Dendritic Cells/pathology , ErbB Receptors/genetics , Humans , In Vitro Techniques , Laryngeal Neoplasms/genetics , Laryngeal Neoplasms/immunology , Male , Mice , Mice, SCID , RNA, Small Interfering/genetics , Recombinant Fusion Proteins/genetics , Suppressor of Cytokine Signaling 1 Protein/genetics , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
8.
Mol Med Rep ; 16(5): 6405-6411, 2017 Nov.
Article in English | MEDLINE | ID: mdl-28901399

ABSTRACT

Macrophage polarization is flexible, and involves in different signaling pathways and various transcription factors. Suppressor of cytokine signaling (SOCS) is an important inhibitor of cytokine signaling pathways and also a key physiological regulator for natural and acquired immunity systems. Following transfection of SOCS1 short hairpin (sh)RNA into mouse macrophage cells, reverse transcription­quantitative polymerase chain reaction demonstrated that the mRNA levels of Janus kinase (JAK)1 and signal transducer and activator of transcription (STAT)1 increased significantly. In addition, western blotting indicated that JAK1, STAT1 and p­STAT1 expression was significantly enhanced. Fludarabine can inhibit phosphorylation of STAT1 and SOCS1 expression. When fludarabine was added and SOCS1 shRNA was transfected, the inhibition of fludarabine was weakened, and p­STAT1 expression was upregulated. Flow cytometry detection indicated that, following the downregulation of SOCS1 expression, M1­type cells significantly increased, but the proportion of M2­type cells did not change significantly. Fludarabine can reduce the effect of SOCS1 shRNA on promoting M1­type cell polarization, and macrophages can polarize into both M1 and M2 phenotypes. Further ELISA results presented that, when downregulating SOCS1 expression, interleukin (IL)­4 and IL­10 expression was both downregulated, and tumor necrosis factor (TNF)­α and interferon (IFN)­Î³ expression was significantly upregulated. When adding fludarabine or injecting with the traditional Chinese medicine Xuebijing, IL­4 and IL­10 expression was both significantly upregulated, and TNF­α and IFN­Î³ expression was significantly downregulated. When adding fludarabine and downregulating SOCS1, IL­4, IL­10, TNF­α and IFN­Î³ expression presented no significant changes. The above results indicated that, when SOCS1 expression is downregulated, it will activate the JAK1/STAT1 pathway, and thereby promote the polarization of macrophages into M1 type. The findings are of great importance for understanding occurrence, development and treatment of various immune­related diseases.


Subject(s)
Janus Kinase 1/immunology , Macrophages, Peritoneal/immunology , STAT1 Transcription Factor/immunology , Signal Transduction/immunology , Suppressor of Cytokine Signaling 1 Protein/immunology , Animals , Anti-Inflammatory Agents/pharmacology , Cell Differentiation , Drugs, Chinese Herbal/pharmacology , Enzyme Inhibitors/pharmacology , Gene Expression Regulation , Interferon-gamma/genetics , Interferon-gamma/immunology , Interleukin-10/genetics , Interleukin-10/immunology , Interleukin-4/genetics , Interleukin-4/immunology , Janus Kinase 1/genetics , Macrophages, Peritoneal/cytology , Macrophages, Peritoneal/drug effects , Mice , Phosphorylation , Primary Cell Culture , RNA, Small Interfering/genetics , RNA, Small Interfering/immunology , STAT1 Transcription Factor/agonists , STAT1 Transcription Factor/genetics , Suppressor of Cytokine Signaling 1 Protein/antagonists & inhibitors , Suppressor of Cytokine Signaling 1 Protein/genetics , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/immunology , Vidarabine/analogs & derivatives , Vidarabine/antagonists & inhibitors , Vidarabine/pharmacology
9.
Tumour Biol ; 39(5): 1010428317694315, 2017 May.
Article in English | MEDLINE | ID: mdl-28466787

ABSTRACT

This study aimed to investigate the effect of SOCS1 silencing on the proliferation and apoptosis of melanoma cells by in vivo and in vitro studies. Immunohistochemical staining was used to detect SOCS1 expression in melanoma tissues and pigmented nevi. Quantitative real-time polymerase chain reaction and western blotting were applied to detect the messenger RNA and protein expressions of SOCS1 in primary human melanocytes and malignant melanoma cell lines (A375, SK-MEL-5, M14, and MV3). Melanoma cells were assigned into mock, negative small interfering RNA, and SOCS1-small interfering RNA groups. The proliferation, cell cycle and apoptosis, and messenger RNA expression of SOCS1 in MV3 and A375 cells were detected using MTT assay, flow cytometry, and quantitative real-time polymerase chain reaction, respectively. The expressions of SOCS1 protein, extracellular signal-regulated kinase, and janus kinase signal transduction and activators of transcription signaling pathways-related proteins were detected using western blotting. After the establishment of subcutaneous xenograft tumor models in nude mice, the latent period, size, volume and growth speed of xenograft tumors in the mock, negative small interfering RNA, and SOCS1-small interfering RNA groups were examined and compared. The results indicated that positive expression rate of SOCS1 was higher in malignant melanoma tissues than in pigmented nevi. MV3 cells had the highest messenger RNA and protein expressions of SOCS1, followed by A357 cells. Compared with the mock and negative small interfering RNA groups, SOCS1-small interfering RNA group showed lower cell viability, elevated cell apoptosis, more cells in G0/G1 phase and less cells in S and G2/M phases, and decreased messenger RNA and protein expressions of SOCS1, p-ERK1/2, p-JAK2, p-STAT1, and p-STAT3. Compared with the mock and negative small interfering RNA groups, the SOCS1-small interfering RNA group showed longer latent period of tumor, smaller tumor size and volume, and smoother tumor growth curve. To conclude, SOCS1 silencing can inhibit proliferation and induce apoptosis of MV3 and A357 melanoma cells in vivo and in vitro by inhibiting extracellular signal-regulated kinase and janus kinase signal transduction and activators of transcription signaling pathways.


Subject(s)
Apoptosis/genetics , Cell Proliferation/genetics , Melanoma/genetics , Suppressor of Cytokine Signaling 1 Protein/genetics , Adult , Animals , Cell Line, Tumor , Gene Expression Regulation, Neoplastic/genetics , Gene Silencing , Humans , Melanoma/pathology , Mice , Middle Aged , Signal Transduction/genetics , Suppressor of Cytokine Signaling 1 Protein/antagonists & inhibitors , Xenograft Model Antitumor Assays
SELECTION OF CITATIONS
SEARCH DETAIL
...