Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 10 de 10
1.
Phytomedicine ; 118: 154950, 2023 Sep.
Article En | MEDLINE | ID: mdl-37441987

BACKGROUND: Sustained liver fibrosis may lead to cirrhosis. Activated hepatic stellate cells (HSCs) are crucial for liver fibrosis development. Ferroptosis, a newly iron-dependent regulated cell death, has been demonstrated to be involved in HSC inactivation. PURPOSE: Ginsenoside Rh2 (GRh2), a natural bioactive product derived from ginseng, has been shown to promote HSC inactivation. However, the effect of GRh2 on HSC ferroptosis remains unclear. METHODS: We explored the effects of GRh2 on liver fibrosis in vivo and in vitro. RNA-sequence analysis was performed in HSCs after GRh2 treatment. The crosstalk between ferroptotic HSCs and macrophages was also explored. RESULTS: GRh2 alleviated liver fibrosis in vivo. In vitro, GRh2 reduced HSC proliferation and activation via ferroptosis, with increased intracellular iron, reactive oxygen species, malondialdehyde and glutathione depletion. The expression of SLC7A11, a negative regulator of ferroptosis, was obviously reduced by GRh2. Interestingly, interferon regulatory factor 1 (IRF1), a transcription factor, was predicted to bind the promoter region of SCL7A11. The interaction between IRF1 and SCL7A11 was further confirmed by the results of chromatin immunoprecipitation and luciferase reporter assays. Furthermore, loss of IRF1 led to an increase in SCL7A11, which contributed to the suppression of HSC ferroptosis and the enhancement of HSC activation in GRh2-treated HSCs. Further studies revealed that GRh2-induced HSC ferroptosis contributed to the inhibition of macrophage recruitment via regulation of inflammation-related genes. Moreover, GRh2 caused a reduction in liver inflammation in vivo. CONCLUSION: Collectively, GRh2 up-regulates IRF1 expression, resulting in the suppression of SLC7A11, which contributes to HSC ferroptosis and inactivation. GRh2 ameliorates liver fibrosis through enhancing HSC ferroptosis and inhibiting liver inflammation. GRh2 may be a promising drug for treating liver fibrosis.


Ferroptosis , Hepatic Stellate Cells , Humans , Interferon Regulatory Factor-1/metabolism , Interferon Regulatory Factor-1/pharmacology , Liver Cirrhosis/metabolism , Fibrosis , Iron/metabolism , Inflammation/metabolism , Amino Acid Transport System y+/metabolism
2.
Clin Immunol ; 250: 109303, 2023 05.
Article En | MEDLINE | ID: mdl-36997038

Recombinant interferon-α2a (IFNα2a) has been widely used in the treatment of Behcet's uveitis (BU). However, the mechanism underlying its effects remains poorly understood. In this study, we investigated its effect on dendritic cells (DCs) and CD4+ T cells, which are essential for the development of BU. Our results showed that the expression of PDL1 and IRF1 was significantly decreased in DCs from active BU patients, and IFNα2a could significantly upregulate PDL1 expression in an IRF1-dependent manner. IFNα2a-treated DCs induced CD4+ T cells apoptosis and inhibited the Th1/Th17 immune response in association with reduced secretion of IFN-γ and IL-17. We also found that IFNα2a promoted Th1 cell differentiation and IL-10 secretion by CD4+ T cells. Finally, a comparison of patients before and after IFNα2a therapy revealed that the frequencies of Th1/Th17 cells significantly decreased in association with remission of uveitis after IFNα2a therapy. Collectively, these results show that IFNα2a could exert its effects by modulating the function of DCs and CD4+ T cells in BU.


Behcet Syndrome , Uveitis , Humans , Apoptosis , Dendritic Cells , Interferon alpha-2 , Interferon Regulatory Factor-1/genetics , Interferon Regulatory Factor-1/metabolism , Interferon Regulatory Factor-1/pharmacology , Th1 Cells , Th17 Cells , Uveitis/drug therapy , CD4-Positive T-Lymphocytes/immunology
3.
Tissue Cell ; 82: 102046, 2023 Jun.
Article En | MEDLINE | ID: mdl-36933274

BACKGROUND: Osteoarthritis is a main cause of deformity in aging people. The chondrogenesis of human adipose-derived stem cells (hADSCs) has a positive effect on the cure of osteoarthritis. However, the regulatory mechanism of hADSC chondrogenesis still needs further exploration. This research investigates the role of interferon regulatory factor 1 (IRF1) in the chondrogenesis of hADSCs. METHODS: hADSCs were purchased and cultured. The interaction between IRF1 and hypoxia inducible lipid droplet associated (HILPDA) was predicted by bioinformatics analysis, and verified through dual-luciferase reporter and chromatin immunoprecipitation assays. The expressions of IRF1 and HILPDA in osteoarthritis cartilage samples were measured through qRT-PCR. After hADSCs were transfected or further induced for chondrogenesis, the chondrogenesis was visualized by Alcian blue staining, and the expressions of IRF1, HILPDA and chondrogenesis-related factors (SOX9, Aggrecan, COL2A1, MMP13, MMP3) were determined through qRT-PCR or Western blot. RESULTS: HILPDA bound to IRF1 in hADSCs. IRF1 and HILPDA levels were up-regulated during the chondrogenesis of hADSCs. Overexpressions of IRF1 and HILPDA promoted the chondrogenesis of hADSCs with the up-regulation of SOX9, Aggrecan and COL2A1 and the down-regulation of MMP13 and MMP3; however, IRF1 silencing generated the opposite effects. Besides, HILPDA overexpression reversed the effects of IRF1 silencing on inhibiting chondrogenesis of hADSCs and regulating the expressions of chondrogenesis-related factors. CONCLUSION: IRF1 promotes the chondrogenesis of hADSCs through up-regulating HILPDA level, providing novel biomarkers for treating osteoarthritis.


Matrix Metalloproteinase 3 , Osteoarthritis , Humans , Matrix Metalloproteinase 13/metabolism , Matrix Metalloproteinase 13/pharmacology , Matrix Metalloproteinase 3/metabolism , Matrix Metalloproteinase 3/pharmacology , Aggrecans/genetics , Aggrecans/metabolism , Aggrecans/pharmacology , Chondrogenesis , Interferon Regulatory Factor-1/genetics , Interferon Regulatory Factor-1/metabolism , Interferon Regulatory Factor-1/pharmacology , Lipid Droplets/metabolism , Stem Cells/metabolism , Osteoarthritis/genetics , Osteoarthritis/metabolism , Cell Differentiation/genetics
4.
Antioxid Redox Signal ; 38(10-12): 731-746, 2023 04.
Article En | MEDLINE | ID: mdl-36242511

Aims: Calcium oxalate (CaOx) crystal deposition induces damage to the renal tubular epithelium, increases epithelial adhesion, and contributes to CaOx nephrocalcinosis. The long noncoding RNA (lncRNA) nuclear paraspeckle assembly transcript 1 (NEAT1) is thought to be involved in this process. In this study, we aimed to investigate the mechanism by which NEAT1 regulates renal tubular epithelium in response to inflammatory and oxidative injury triggered by CaOx crystals. Results: As CaOx crystals were deposited in mouse kidney tissue, the expression of NEAT1 was significantly elevated and positively correlated with interferon regulatory factor 1 (IRF1), Toll-like receptor 4 (TLR4), and NF-κB. NEAT1 targets and inhibits miR-130a-3p as a competitor to endogenous RNA. miR-130 binds to and exerts inhibitory effects on the 3'-untranslated region of IRF1. After transfected with silence-NEAT1, IRF1, TLR4, and NF-κB were also variously inhibited, and oxidative damage in renal calcinosis was subsequently attenuated. When we simultaneously inhibited NEAT1 and miR-130, renal tubular injury was exacerbated. Innovation and Conclusion: We found that the lncRNA NEAT1 can enhance IRF1 signaling through targeted repression of miR-130a-3p and activate TLR4/NF-κB pathways to promote oxidative damage during CaOx crystal deposition. This provides an explanation for the tubular epithelial damage caused by CaOx crystals and offers new ideas and drug targets for the prevention and treatment of CaOx nephrocalcinosis. Antioxid. Redox Signal. 38, 731-746.


Calcinosis , MicroRNAs , Nephrocalcinosis , RNA, Long Noncoding , Mice , Animals , Calcium Oxalate/chemistry , Calcium Oxalate/metabolism , Calcium Oxalate/pharmacology , Nephrocalcinosis/metabolism , Toll-Like Receptor 4/metabolism , RNA, Long Noncoding/genetics , NF-kappa B/metabolism , Interferon Regulatory Factor-1/metabolism , Interferon Regulatory Factor-1/pharmacology , Kidney/metabolism , Oxidative Stress , MicroRNAs/genetics , Oxidation-Reduction
5.
J Biomed Sci ; 29(1): 73, 2022 Sep 21.
Article En | MEDLINE | ID: mdl-36127734

BACKGROUND: Mesenchymal stem cells (MSCs) exhibit two bidirectional immunomodulatory abilities: proinflammatory and anti-inflammatory regulatory effects. Long noncoding RNAs (lncRNAs) have important functions in the immune system. Previously, we performed high-throughput sequencing comparing lncRNA expression profiles between MSCs cocultured with or without CD14+ monocytes and screened out a new lncRNA termed lncRNA MCP1 regulatory factor (MRF). However, the mechanism of MRF in MSCs is still unknown. METHODS: MRF expression was quantified via qRT-PCR. RNA interference and lentiviruses were used to regulate MRF expression. The immunomodulatory effects of MSCs on monocytes were evaluated via monocyte migration and macrophage polarization assays. RNA pull-down and mass spectrometry were utilized to identify downstream factors of MRF. A dual-luciferase reporter assay was applied to analyze the transcription factors regulating MRF. qRT-PCR, western blotting and ELISAs were used to assess MCP1 expression. A human monocyte adoptive transfer mouse model was applied to verify the function of MRF in vivo. RESULTS: MRF was upregulated in MSCs during coculture with CD14+ monocytes. MRF increased monocyte recruitment by upregulating the expression of monocyte chemotactic protein (MCP1). Knockdown of MRF enhanced the regulatory effect of MSCs on restraining M1 polarization and facilitating M2 polarization. Mechanistically, MRF bound to the downstream protein heterogeneous nuclear ribonucleoprotein D (HNRNPD) to upregulate MCP1 expression, and the transcription factor interferon regulatory factor 1 (IRF1) activated MRF transcription early during coculture. The human monocyte adoptive transfer model showed that MRF downregulation in MSCs inhibited monocyte chemotaxis and enhanced the effects of MSCs to inhibit M1 macrophage polarization and promote M2 polarization in vivo. CONCLUSION: We identified the new lncRNA MRF, which exhibits proinflammatory characteristics. MRF regulates the ability of MSCs to accelerate monocyte recruitment and modulate macrophage polarization through the HNRNPD-MCP1 axis and initiates the proinflammatory regulatory process in MSCs, suggesting that MRF is a potential target to improve the clinical effect of MSC-based therapy or correct MSC-related immunomodulatory dysfunction under pathological conditions.


Heterogeneous-Nuclear Ribonucleoprotein D , Mesenchymal Stem Cells , RNA, Long Noncoding , Animals , Anti-Inflammatory Agents/pharmacology , Heterogeneous-Nuclear Ribonucleoprotein D/metabolism , Heterogeneous-Nuclear Ribonucleoprotein D/pharmacology , Humans , Interferon Regulatory Factor-1/metabolism , Interferon Regulatory Factor-1/pharmacology , Mesenchymal Stem Cells/metabolism , Mice , Monocyte Chemoattractant Proteins/metabolism , Monocyte Chemoattractant Proteins/pharmacology , Monocytes/metabolism , RNA, Long Noncoding/metabolism
6.
Cytokine Growth Factor Rev ; 64: 1-6, 2022 04.
Article En | MEDLINE | ID: mdl-35090813

Many studies have been conducted over the last few decades to understand better the functions of IRF3 and IRF7 in antiviral immune responses. However, the precise underlying molecular mechanism of IRF1-mediated immune response remains largely unknown. Recent studies indicate that IRF1 exerts strong antiviral activities against several viral infections through diverse mechanisms, both in IFN-dependent and IFN-independent manners. Nevertheless, the efficacy and kinetics of inducing IFNs and ISGs remain unknown. Here we summarize the recent advances in IRF1 research and highlight its potential roles in initiating IFN immune responses and subsequent IRF1-triggering antiviral responses. Challenges regarding the IFN positive feedback mediated by IRF7 during infection will be discussed; this classical loop may also be mediated in part by IRF1. Therefore, we propose a revised model that may help decipher the functional roles of IRF1 in antiviral immunity.


Antiviral Agents , Virus Diseases , Antiviral Agents/pharmacology , Humans , Immunity, Innate , Interferon Regulatory Factor-1/pharmacology , Signal Transduction , Virus Replication/physiology
7.
Cancer Invest ; 40(1): 35-45, 2022 Jan.
Article En | MEDLINE | ID: mdl-34313498

IRF1 is a nuclear transcription factor that mediates interferon effects and appears to have anti-tumor activity. To determine the roles of IRF1 in colorectal cancer (CRC), we investigated the effects of IRF1 in CRC cells. We found that IRF1 inhibit cell proliferation and tumor growth. Under starvation conditions, IRF1 enhanced apoptosis and reduced autophagic flux. ATG13, an important factor of autophagy complex, was confirmed as a target of IRF1. These findings indicated that IRF1 function as a tumor suppressor in CRC and inhibit autophagy through ATG13, targeting this pathway may provide new insights into the molecular mechanisms of CRC progression.


Autophagy-Related Proteins/metabolism , Autophagy/drug effects , Colorectal Neoplasms/physiopathology , Interferon Regulatory Factor-1/therapeutic use , Animals , Cell Proliferation , Disease Models, Animal , Humans , Interferon Regulatory Factor-1/pharmacology , Mice , Mice, Nude , Middle Aged , Transfection
8.
Antiviral Res ; 158: 288-302, 2018 10.
Article En | MEDLINE | ID: mdl-30144461

Specific host pathways that may be targeted therapeutically to inhibit the replication of Ebola virus (EBOV) and other emerging viruses remain incompletely defined. A screen of 200,000 compounds for inhibition of an EBOV minigenome (MG) assay that measures the function of the viral polymerase complex identified as hits several compounds with an amino-tetrahydrocarbazole scaffold. This scaffold was structurally similar to GSK983, a compound previously described as having broad-spectrum antiviral activity due to its impairing de novo pyrimidine biosynthesis through inhibition of dihydroorotate dehydrogenase (DHODH). We generated compound SW835, the racemic version of GSK983 and demonstrated that SW835 and brequinar, another DHODH inhibitor, potently inhibit the MG assay and the replication of EBOV, vesicular stomatitis virus (VSV) and Zika (ZIKV) in vitro. Nucleoside and deoxynucleoside supplementation studies demonstrated that depletion of pyrimidine pools contributes to antiviral activity of these compounds. As reported for other DHODH inhibitors, SW835 and brequinar also induced expression of interferon stimulated genes (ISGs). ISG induction was demonstrated to occur without production of IFNα/ß and independently of the IFNα receptor and was not blocked by EBOV-encoded suppressors of IFN signaling pathways. Furthermore, we demonstrated that transcription factor IRF1 is required for this ISG induction, and that IRF1 induction requires the DNA damage response kinase ATM. Therefore, de novo pyrimidine biosynthesis is critical for the replication of EBOV and other RNA viruses and inhibition of this pathway activates an ATM and IRF1-dependent innate immune response that subverts EBOV immune evasion functions.


Ebolavirus/drug effects , Immunity, Innate/drug effects , Nucleosides/pharmacology , Pyrimidines/antagonists & inhibitors , Pyrimidines/biosynthesis , Virus Replication/drug effects , A549 Cells , Antiviral Agents/pharmacology , Biphenyl Compounds/chemistry , Biphenyl Compounds/pharmacology , Carbazoles/chemistry , Carbazoles/pharmacology , DNA Damage , Dihydroorotate Dehydrogenase , HEK293 Cells , Hemorrhagic Fever, Ebola/drug therapy , Hemorrhagic Fever, Ebola/virology , Host-Pathogen Interactions/drug effects , Humans , Immune Evasion , Immunity, Innate/genetics , Interferon Regulatory Factor-1/metabolism , Interferon Regulatory Factor-1/pharmacology , Interferon-alpha/metabolism , Interferon-beta/metabolism , Oxidoreductases Acting on CH-CH Group Donors/antagonists & inhibitors , RNA Viruses/drug effects , Signal Transduction/drug effects , Vesiculovirus/drug effects , Zika Virus/drug effects
9.
Am J Physiol Renal Physiol ; 305(2): F164-72, 2013 Jul 15.
Article En | MEDLINE | ID: mdl-23657854

We previously reported that expression of the transcription factor interferon regulatory factor 1 (IRF1) is an early, critical maladaptive signal expressed by renal tubules during murine ischemic acute kidney injury (AKI). We now show that IRF1 mediates signals from reactive oxygen species (ROS) generated during ischemic AKI and that these signals ultimately result in production of α-subtypes of type I interferons (IFNαs). We found that genetic knockout of the common type I IFN receptor (IFNARI-/-) improved kidney function and histology during AKI. There are major differences in the spatial-temporal production of the two major IFN subtypes, IFNß and IFNαs: IFNß expression peaks at 4 h, earlier than IFNαs, and continues at the same level at 24 h; expression of IFNαs also increases at 4 h but continues to increase through 24 h. The magnitude of the increase in IFNαs relative to baseline is much greater than that of IFNß. We show by immunohistology and study of isolated cells that IFNß is produced by renal leukocytes and IFNαs are produced by renal tubules. IRF1, IFNαs, and IFNARI were found on the same renal tubules during ischemic AKI. Furthermore, we found that ROS induced IFNα expression by renal tubules in vitro. This expression was inhibited by small interfering RNA knockdown of IRF1. Overexpression of IRF1 resulted in the production of IFNαs. Furthermore, we found that IFNα stimulated production of maladaptive proinflammatory CXCL2 by renal tubular cells. Altogether our data support the following autocrine pathway in renal tubular cells: ROS > IRF1 > IFNα > IFNARI > CXCL2.


Acute Kidney Injury/metabolism , Chemokine CXCL2/metabolism , Interferon Regulatory Factor-1/pharmacology , Interferon-alpha/biosynthesis , Reactive Oxygen Species/pharmacology , Reperfusion Injury/metabolism , Animals , Autocrine Communication , Disease Models, Animal , Kidney Tubules, Proximal/metabolism , Leukocytes/metabolism , Male , Mice , Mice, Knockout , Receptor, Interferon alpha-beta/metabolism
10.
Biochim Biophys Acta ; 1783(9): 1654-62, 2008 Sep.
Article En | MEDLINE | ID: mdl-18472010

Interferon regulatory factor-1 (IRF-1) is a tumor suppressor and transcriptional modulator that can regulate gene expression involved in cell growth control, induction of apoptosis, and post-translation modification. In this study, we found that IRF-1 inhibits endothelial cell angiogenesis using human umbilical vein endothelial cell (HUVECs) culture system. In addition, IRF-1 directly inhibited the tube formation of endothelial cells on Matrigel and reduced the expression of p-Akt, and p-eNOS, which play a significant role in angiogenesis when stimulated by VEGF. We also demonstrate that C-terminal region including transactivation domain (TA) of IRF-1 functions as a signal for its angiostatic activity, and is spliced in human tumor tissues. These findings indicate that splicing variant involving exons 7 of IRF-1 could potentially modulate anti-angiogenic effect of IRF-1. In overall, this study provides the first evidence for anti-angiogenic role of IRF-1, which may have therapeutic values for cancer and angiogenesis-associated diseases.


Angiogenesis Inhibitors/pharmacology , Endothelium, Vascular/drug effects , Interferon Regulatory Factor-1/pharmacology , Vascular Endothelial Growth Factor A/antagonists & inhibitors , Animals , Cell Movement/drug effects , Cell Proliferation/drug effects , Cells, Cultured , Chick Embryo , Endothelium, Vascular/growth & development , Endothelium, Vascular/metabolism , Exons , Humans , Interferon Regulatory Factor-1/genetics , Mice , Mice, Inbred BALB C , Neoplasms/blood supply , Neoplasms/pathology , Neovascularization, Pathologic , Neovascularization, Physiologic/drug effects , Phosphoinositide-3 Kinase Inhibitors , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Vascular Endothelial Growth Factor A/metabolism , Vascular Endothelial Growth Factor Receptor-2/metabolism
...