Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 124
Filter
1.
Aging Cell ; 21(6): e13646, 2022 06.
Article in English | MEDLINE | ID: mdl-35645319

ABSTRACT

Older age and underlying conditions such as diabetes/obesity or immunosuppression are leading host risk factors for developing severe complications from COVID-19 infection. The pathogenesis of COVID-19-related cytokine storm, tissue damage, and fibrosis may be interconnected with fundamental aging processes, including dysregulated immune responses and cellular senescence. Here, we examined effects of key cytokines linked to cellular senescence on expression of SARS-CoV-2 viral entry receptors. We found exposure of human umbilical vein endothelial cells (HUVECs) to the inflammatory cytokines, TNF-α + IFN-γ or a cocktail of TNF-α + IFN-γ + IL-6, increased expression of ACE2/DPP4, accentuated the pro-inflammatory senescence-associated secretory phenotype (SASP), and decreased cellular proliferative capacity, consistent with progression towards a cellular senescence-like state. IL-6 by itself failed to induce substantial effects on viral entry receptors or SASP-related genes, while synergy between TNF-α and IFN-γ initiated a positive feedback loop via hyper-activation of the JAK/STAT1 pathway, causing SASP amplification. Breaking the interactive loop between senescence and cytokine secretion with JAK inhibitor ruxolitinib or antiviral drug remdesivir prevented hyper-inflammation, normalized SARS-CoV-2 entry receptor expression, and restored HUVECs proliferative capacity. This loop appears to underlie cytokine-mediated viral entry receptor activation and links with senescence and hyper-inflammation.


Subject(s)
COVID-19 Drug Treatment , COVID-19 , Interferon-gamma , SARS-CoV-2 , Tumor Necrosis Factor-alpha , COVID-19/virology , Cytokines/immunology , Drug Synergism , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Inflammation/drug therapy , Inflammation/virology , Interferon-gamma/pharmacology , Interleukin-6/metabolism , Receptors, Virus/metabolism , SARS-CoV-2/drug effects , SARS-CoV-2/metabolism , STAT1 Transcription Factor/biosynthesis , STAT1 Transcription Factor/immunology , Tumor Necrosis Factor-alpha/pharmacology
2.
Cell Rep ; 38(4): 110302, 2022 01 25.
Article in English | MEDLINE | ID: mdl-35081346

ABSTRACT

It is well known that interferon (IFN)-α/-ß activates the JAK/STAT signaling pathway and suppresses viral replication through the induction of IFN stimulated genes (ISGs). Here, we report that knockout of HDAC3 from macrophages results in the decreased expression of STAT1 and STAT2, leading to defective antiviral immunity in cells and mice. Further studies show that HDAC3 interacts with a conserved transcription factor Forkhead Box K1 (FOXK1), co-localizes with FOXK1 at the promoter of STAT1 and STAT2, and is required for protecting FOXK1 from lysosomal system-mediated degradation. FOXK1-deficient macrophages also show low STAT1 and STAT2 expression with defective responses to viruses. Thus, our studies uncover the biological importance of HDAC3 in regulating the antiviral immunity of macrophages through interacting with FOXK1 to regulate the expression of STAT1 and STAT2.


Subject(s)
Gene Expression Regulation/immunology , Histone Deacetylases/immunology , Immunity, Innate/immunology , Macrophages/immunology , Virus Diseases/immunology , Animals , Forkhead Transcription Factors/immunology , Mice , STAT1 Transcription Factor/biosynthesis , STAT1 Transcription Factor/immunology , STAT2 Transcription Factor/biosynthesis , STAT2 Transcription Factor/immunology , Transcription, Genetic
3.
Int J Biol Sci ; 17(7): 1795-1807, 2021.
Article in English | MEDLINE | ID: mdl-33994863

ABSTRACT

Emerging evidence has suggested the functions of exosomes in allergic diseases including asthma. By using a mouse model with asthma induced by ovalbumin (OVA), we explored the roles of M2 macrophage-derived exosomes (M2Φ-Exos) in asthma progression. M2Φ-Exos significantly alleviated OVA-induced fibrosis and inflammatory responses in mouse lung tissues, as well as inhibited abnormal proliferation, invasion, and fibrosis-related protein production in platelet derived growth factor (PDGF-BB) treated primary mouse airway smooth muscle cells (ASMCs). The OVA administration in mice or the PDGF-BB treatment in ASMCs reduced the expression of miR-370, which was detected in M2Φ-Exos by miRNA sequencing. However, treating the mice or ASMCs with M2Φ-Exos reversed the inhibitory effect of OVA or PDGF-BB on miR-370 expression. We identified that the target of miR-370 was fibroblast growth factor 1 (FGF1). Downregulation of miR-370 by Lv-miR-370 inhibitor or overexpression of FGF1 by Lv-FGF1 blocked the protective roles of M2Φ-Exos in asthma-like mouse and cell models. M2Φ-Exos were found to inactivate the MAPK signaling pathway, which was recovered by miR-370 inhibition or FGF1 overexpression. Collectively, we conclude that M2Φ-Exos carry miR-370 to alleviate asthma progression through downregulating FGF1 expression and the MAPK/STAT1 signaling pathway. Our study may offer a novel insight into asthma treatment.


Subject(s)
Asthma/genetics , Fibroblast Growth Factor 1/genetics , Gene Expression Regulation , Macrophages, Alveolar/metabolism , MicroRNAs/genetics , Mitogen-Activated Protein Kinase Kinases/genetics , STAT1 Transcription Factor/genetics , Animals , Asthma/metabolism , Asthma/pathology , Cell Proliferation , Cells, Cultured , Disease Models, Animal , Down-Regulation , Exosomes/genetics , Exosomes/metabolism , Fibroblast Growth Factor 1/biosynthesis , Macrophages, Alveolar/pathology , Mice , Mice, Inbred C57BL , MicroRNAs/biosynthesis , Mitogen-Activated Protein Kinase Kinases/biosynthesis , Myocytes, Smooth Muscle/metabolism , Myocytes, Smooth Muscle/pathology , STAT1 Transcription Factor/biosynthesis , Signal Transduction
4.
Inflammation ; 44(1): 206-216, 2021 Feb.
Article in English | MEDLINE | ID: mdl-32772240

ABSTRACT

Sjögren's syndrome (SS) is a chronic autoimmune disease targeting salivary and lacrimal glands. C-X-C motif chemokine ligand 10 (CXCL10) expression is upregulated in lip salivary glands (LSGs) of primary SS (pSS) patients, and CXCL10 involved in SS pathogenesis via immune-cell accumulation. Moreover, interferon (IFN)-γ enhances CXCL10 production via the Janus kinase (JAK)/signal transducer and activator of transcription (STAT) pathway. We investigated the effects of baricitinib, a selective JAK1/2 inhibitor, on both IFN-γ-induced CXCL10 production and immune-cell chemotaxis. We used immunohistochemical staining to determine the expression levels and localization of JAK1 and JAK2 in LSGs of SS patients (n = 12) and healthy controls (n = 3). We then evaluated the effect of baricitinib in an immortalized normal human salivary gland ductal (NS-SV-DC) cell line. Immunohistochemical analysis of LSGs from pSS patients revealed strong JAK1 and JAK2 expression in ductal and acinar cells, respectively. Baricitinib significantly inhibited IFN-γ-induced CXCL10 expression as well as the protein levels in an immortalized human salivary gland ductal-cell clone in a dose-dependent manner. Additionally, western blot analysis showed that baricitinib suppressed the IFN-γ-induced phosphorylation of STAT1 and STAT3, with a stronger effect observed in the case of STAT1. It also inhibited IFN-γ-mediated chemotaxis of Jurkat T cells. These results suggested that baricitinib suppressed IFN-γ-induced CXCL10 expression and attenuated immune-cell chemotaxis by inhibiting JAK/STAT signaling, suggesting its potential as a therapeutic strategy for pSS.


Subject(s)
Azetidines/pharmacology , Chemokine CXCL10/antagonists & inhibitors , Interferon-gamma/pharmacology , Janus Kinase 1/antagonists & inhibitors , Janus Kinase 2/antagonists & inhibitors , Purines/pharmacology , Pyrazoles/pharmacology , STAT1 Transcription Factor/antagonists & inhibitors , Salivary Ducts/metabolism , Sulfonamides/pharmacology , Azetidines/therapeutic use , Cell Line, Transformed , Chemokine CXCL10/biosynthesis , Female , Humans , Janus Kinase 1/biosynthesis , Janus Kinase 2/biosynthesis , Jurkat Cells , Purines/therapeutic use , Pyrazoles/therapeutic use , STAT1 Transcription Factor/biosynthesis , Salivary Ducts/drug effects , Signal Transduction/drug effects , Signal Transduction/physiology , Sjogren's Syndrome/drug therapy , Sjogren's Syndrome/metabolism , Sulfonamides/therapeutic use
5.
Cytokine ; 138: 155359, 2021 02.
Article in English | MEDLINE | ID: mdl-33160814

ABSTRACT

Cardiac fibroblasts (CF) play a key role in the homeostasis of the extracellular matrix in cardiac tissue and are newly recognized as inflammatory supporter cells. Besides, CF-to-Cardiac myofibroblast differentiation is commanded by TGF-b, through SMAD signaling pathways, and these last cells are strongly implicated in cardiac fibrosis. In the heart IFN-ß is produced by CF; however, the role of IFN-ß, STAT proteins, and STAT-homo or heterodimers in the regulation of CF function with or without a fibrotic environment is unknown. CF were isolated from hearts of adult rats, and by western blot analysis we studied STAT1, STAT2, and STAT3 phosphorylation and through specific siRNA against these proteins we analyzed their role in CF functions such as differentiation (α-SMA expression); and pro-collagen type-I synthesis and secretion expression levels; collagen gels contraction and CF migration. In cultured adult rats CF, IFN-ß increases phosphorylation of STAT1, STAT2, and STAT3. Both STAT1 and STAT2 were involved in decreasing α-SMA and CF migration induced by TGF-ß1. Also, IFN-ß through STAT1 regulated pro-collagen type-I protein expression levels, and collagen gels contraction induced by TGF-ß1. STAT3 was not involved in any effects of IFN-ß studied. In conclusion, IFN-ß through STAT1 and STAT2 shows antifibrotic effects on CF TGF-ß1-treated, whereas STAT3 did not participate in such effect.


Subject(s)
Collagen/metabolism , Fibroblasts/metabolism , Gene Expression Regulation , Interferon-beta/metabolism , Transforming Growth Factor beta1/metabolism , Animals , Cell Differentiation , Cell Movement , Collagen/chemistry , Extracellular Matrix/metabolism , Gene Silencing , Male , Myofibroblasts/drug effects , Phosphorylation , RNA, Small Interfering/metabolism , Rats , Rats, Sprague-Dawley , STAT1 Transcription Factor/biosynthesis , STAT2 Transcription Factor/biosynthesis , Signal Transduction/drug effects
6.
Cancer Biol Ther ; 21(10): 963-971, 2020 10 02.
Article in English | MEDLINE | ID: mdl-33043814

ABSTRACT

Signal transducer and activator of transcription 1 (STAT1) is related to the immune microenvironment of tumorigenesis. The programmed cell death 1 (PD-1) and its ligand (PD-L1) have been reported to be important in immunotherapy by mediating tumor immune evasion. Blocking the PD-1/PD-L1 pathway can restore the endogenous anti-tumor immune response. This study aimed to examine the expression of STAT1, PD-1, and PD-L1 and the correlation between selected markers in human epithelial ovarian cancer (EOC). The results showed that malignant tumors contained more STAT1, PD-1, and PD-L1 positive cells. The expression of STAT1 and PD-L1 was associated with age, whereas PD-1 and PD-L1 associated with histopathological type, in patients with ovarian tumors. Moreover, the expression of STAT1 was found to be associated with disease stages and the grade of serous carcinoma. STAT1 expression was higher in OC cells than normal ovarian surface epithelial cells and was positively correlated with PD-L1 expression. The knockdown of STAT1 decreased PD-L1 expression, whereas overexpression of STAT1 increased PD-L1 expression. Furthermore, the expression of STAT1, PD-1, and PD-L1 was lower in paclitaxel-resistant cells than sensitive cells. Finally, STAT1 affected the overall survival and progression-free survival of patients with EOC. These findings suggest that STAT1, PD-1, and PD-L1 are the tissue markers of EOC and imply the possibility that the high level of STAT1, PD-1, and PD-L1 may favor the checkpoint immunotherapy in patients with EOC, but may have a limit in paclitaxel-resistant patients because of the low expression of STAT1, PD-1, and PD-L1 in paclitaxel-resistant cells.


Subject(s)
B7-H1 Antigen/metabolism , Ovarian Neoplasms/metabolism , STAT1 Transcription Factor/biosynthesis , B7-H1 Antigen/immunology , Female , Humans , Ovarian Neoplasms/genetics , Ovarian Neoplasms/immunology , Ovarian Neoplasms/pathology , STAT1 Transcription Factor/genetics , STAT1 Transcription Factor/immunology , Tumor Escape
7.
Oncogene ; 39(22): 4344-4357, 2020 05.
Article in English | MEDLINE | ID: mdl-32335582

ABSTRACT

We explore a novel strategy of activating immune signaling through increased micronuclei formation utilizing a cell cycle checkpoint inhibitor to drive cell cycle progression following ionizing radiation. The Chk1/2 inhibitor AZD7762 is used to abrogate radiation therapy (RT)-induced G2/M cell cycle arrest in multiple cell lines and, we find that this therapeutic combination promotes increased micronuclei formation in vitro and subsequently drives increased type I interferon signaling and cytotoxic T-cell activation. In vivo studies using B16-F10 melanoma cancer cells implanted in C57/BL6 mice demonstrate improved rates of tumor control at the abscopal (unirradiated) site, located outside of the radiation field, only in the AZD7762 + RT group, with a corresponding reduction in mean tumor volume, increase in the CD8 T-cell population, and immune activated gene signaling. Our results demonstrate that targeted inhibition of cell cycle checkpoint activation following ionizing radiation drives increased production of immunogenic micronuclei, leading to systemic tumor response with potential future clinical benefit.


Subject(s)
Cell Nucleus/drug effects , Cell Nucleus/radiation effects , Checkpoint Kinase 1/antagonists & inhibitors , Checkpoint Kinase 2/antagonists & inhibitors , G2 Phase Cell Cycle Checkpoints/drug effects , G2 Phase Cell Cycle Checkpoints/radiation effects , Melanoma, Experimental/immunology , Neoplasm Proteins/antagonists & inhibitors , Thiophenes/pharmacology , Urea/analogs & derivatives , Animals , Cell Line, Tumor , Female , Humans , Interferon-beta/biosynthesis , Interferon-beta/genetics , Melanoma, Experimental/drug therapy , Melanoma, Experimental/pathology , Melanoma, Experimental/radiotherapy , Membrane Proteins/biosynthesis , Membrane Proteins/genetics , Mice, Inbred C57BL , Micronucleus Tests , Neoplasm Proteins/biosynthesis , Neoplasm Proteins/genetics , RNA Interference , RNA, Small Interfering/genetics , RNA, Small Interfering/pharmacology , STAT1 Transcription Factor/biosynthesis , STAT1 Transcription Factor/genetics , Tumor Burden/drug effects , Tumor Burden/radiation effects , Urea/pharmacology , Xenograft Model Antitumor Assays
8.
Sci Rep ; 9(1): 17620, 2019 11 26.
Article in English | MEDLINE | ID: mdl-31772273

ABSTRACT

Inactivation of the protein complex 'mechanistic target of rapamycin complex 1' (mTORC1) can increase the nuclear content of transcriptional regulators of metabolism and apoptosis. Previous studies established that nuclear import of signal transducer and activator of transcription-1 (STAT1) requires the mTORC1-associated adaptor karyopherin-α1 (KPNA1) when mTORC1 activity is reduced. However, the role of other mTORC1-interacting proteins in the complex, including 'protein kinase C delta' (PKCδ), have not been well characterized. In this study, we demonstrate that PKCδ, a STAT1 kinase, contains a functional 'target of rapamycin signaling' (TOS) motif that directs its interaction with mTORC1. Depletion of KPNA1 by RNAi prevented the nuclear import of PKCδ in cells exposed to the mTORC1 inhibitor rapamycin or amino acid restriction. Mutation of the TOS motif in PKCδ led to its loss of regulation by mTORC1 or karyopherin-α1, resulting in increased constitutive nuclear content. In cells expressing wild-type PKCδ, STAT1 activity and apoptosis were increased by rapamycin or interferon-ß. Those expressing the PKCδ TOS mutant exhibited increased STAT1 activity and apoptosis; further enhancement by rapamycin or interferon-ß, however, was lost. Therefore, the TOS motif in PKCδ is a novel structural mechanism by which mTORC1 prevents PKCδ and STAT1 nuclear import, and apoptosis.


Subject(s)
Cell Nucleus/metabolism , Mechanistic Target of Rapamycin Complex 1/metabolism , Protein Kinase C-delta/metabolism , Active Transport, Cell Nucleus/drug effects , Amino Acid Motifs , Apoptosis/drug effects , Apoptosis/physiology , Cell Line , Humans , Models, Molecular , Mutation, Missense , Point Mutation , Protein Conformation , Protein Interaction Mapping , Protein Kinase C-delta/chemistry , Protein Kinase C-delta/genetics , RNA Interference , RNA, Small Interfering/genetics , Recombinant Proteins/metabolism , Regulatory-Associated Protein of mTOR/metabolism , STAT1 Transcription Factor/biosynthesis , Sequence Alignment , Sirolimus/pharmacology , alpha Karyopherins/antagonists & inhibitors , alpha Karyopherins/metabolism
9.
Respir Res ; 20(1): 184, 2019 Aug 15.
Article in English | MEDLINE | ID: mdl-31416461

ABSTRACT

BACKGROUND: Influenza attacks the epithelium of the lung, causing cell death and disruption of the epithelial barrier leading to fluid buildup in the lung and impairment of gas exchange. Limited treatment options for severe influenza pneumonia prioritize the need for the discovery of effective therapies. IL-22 is a cytokine that promotes tissue integrity and has strong promise as a treatment option. While research has been focused on the cytokine itself, there is limited understanding of the regulation of the IL-22 receptor (IL-22Ra1) at the epithelial surface during infection. METHODS: IL-22Ra1 levels were measured by qRT-PCR, western blot and immunofluorescence following H1N1 influenza infection (A/PR/8/34 H1N1) or synthetic TLR3 mimetic, Poly (I:C). Regulation of the receptor was determined using STAT inhibitors (STAT1, STAT3 and PanSTAT inhibitors), TLR3 inhibition, and neutralization of interferon alpha receptor 2 (IFNAR2). Significance was determined by a p-value of greater than 0.05. Significance between two groups was measured using unpaired t-test and significance between more than two groups was measured using one-way ANOVA with Tukey Multiple Comparison Test. RESULTS: Here we show both in vivo and in vitro that IL-22Ra1 was induced as early as 24 h after influenza (H1N1 PR8) infection. This induction was triggered by toll-like receptor 3 (TLR3) as a TLR3 mimetic [Poly (I:C)] also induced IL-22Ra1 and inhibition of endosomal formation required for TLR3 function inhibited this process. This upregulation was dependent upon IFNß signaling through STAT1. Importantly, induction of IL-22Ra1 significantly increased IL-22 signaling as evidenced by pSTAT3 levels following IL-22 treatment. CONCLUSION: Collectively, these data suggest epithelial cells may optimize the beneficial effects of IL-22 through the induction of the IL-22 receptor during viral infection in the lung.


Subject(s)
Influenza, Human/metabolism , Receptors, Interleukin/biosynthesis , STAT1 Transcription Factor/biosynthesis , Toll-Like Receptor 3/biosynthesis , A549 Cells , Animals , Chloroquine/pharmacology , Humans , Influenza, Human/pathology , Interferons/pharmacology , Male , Mice , Mice, Inbred C57BL , Poly I-C/pharmacology
10.
Dermatol Ther ; 32(5): e13029, 2019 09.
Article in English | MEDLINE | ID: mdl-31330078

ABSTRACT

The aim of this study was to assess changes in the expression of STAT1, STAT3, STAT4, SOCS2, and IL17 in psoriatic patients under ustekinumab treatment and in healthy volunteers. The study group consisted of 14 patients suffering from psoriasis vulgaris qualified for ustekinumab therapy (4 women, 10 men) The control group consisted of 14 healthy volunteers (7 women, 7 men), their whole blood was used as a material in this study. A quantitative reverse transcription polymerase chain assay was used to amplify analyzed genes. To indicate the differences in expression of selected genes in the test and control groups, the Kruskal-Wallis and the post hoc Dunn's test was carried out. After 40 weeks of observation of the effectiveness of ustekinumab in patients with psoriasis, the expression of STAT1, STAT3, STAT4, IL17, and SOCS2 was silenced. Statistic differences in expression were observed for STAT3 (40 vs. 0 weeks, p < .05; 0 week vs. C, p < .05) and SOCS2 (0 week vs. C, p < .05). Patients with psoriasis vulgaris have higher levels of STAT1, STAT3, STAT4, SOCS2, and IL17 expression compared to healthy individuals. On the other hand, the treatment of ustekinumab lasting 40 weeks caused a decrease in the transcriptional activity of the analyzed genes.


Subject(s)
Gene Expression Regulation/drug effects , Psoriasis/genetics , STAT1 Transcription Factor/genetics , STAT3 Transcription Factor/genetics , STAT4 Transcription Factor/genetics , Suppressor of Cytokine Signaling Proteins/genetics , Ustekinumab/pharmacology , Adult , Dermatologic Agents/pharmacology , Female , Humans , Interleukin-17/biosynthesis , Interleukin-17/genetics , Male , Middle Aged , Psoriasis/drug therapy , Psoriasis/metabolism , RNA/genetics , STAT1 Transcription Factor/biosynthesis , STAT4 Transcription Factor/biosynthesis , Signal Transduction , Suppressor of Cytokine Signaling Proteins/biosynthesis , Young Adult
11.
J Neuroimmunol ; 331: 46-57, 2019 06 15.
Article in English | MEDLINE | ID: mdl-29113698

ABSTRACT

Glycolysis and mitochondrial respiration are essential for oligodendrocyte metabolism in both the developing and adult CNS. Based on recent reports on the effects of the proinflammatory cytokine IFN-γ on metabolism and on oligodendrocytes, we addressed whether IFN-γ may affect oligodendrocyte bioenergetics in ways relevant to CNS disease. Oligodendrocytes of mice treated with IFN-γ showed significant reductions in aerobic glycolysis and mitochondrial respiration. As expected, IFN-γ treatment led to the induction of STAT1 in oligodendrocytes indicating active signaling into these cells. To determine the direct effects of IFN-γ on oligodendrocyte metabolism, cultured oligodendrocytes were treated with IFN-γ in vitro, which resulted in suppression of glycolysis similar to oligodendrocytes of animals treated with IFN-γ in vivo. Mice lacking SHP-1, a key regulator of IFN-γ and STAT1 signaling in CNS glia, had high constitutive levels of STAT1 and decreased aerobic glycolysis and mitochondrial respiration rates relative to wild type mouse oligodendrocytes. Together, these data show that IFN-γ and SHP-1 control oligodendrocyte bioenergetics in ways that may relate to the role of this cytokine in CNS disease.


Subject(s)
Energy Metabolism/drug effects , Interferon-gamma/pharmacology , Oligodendroglia/drug effects , Protein Tyrosine Phosphatase, Non-Receptor Type 6/physiology , Animals , Cells, Cultured , Central Nervous System/pathology , Enzyme Activation/drug effects , Enzyme Induction/physiology , Female , Glycolysis/drug effects , Male , Mice , Mice, Inbred C3H , Mice, Neurologic Mutants , Oligodendroglia/metabolism , Oxidative Phosphorylation/drug effects , Protein Tyrosine Phosphatase, Non-Receptor Type 6/deficiency , STAT1 Transcription Factor/biosynthesis , STAT1 Transcription Factor/genetics , Signal Transduction/drug effects
12.
J Pak Med Assoc ; 68(11): 1446-1450, 2018 Nov.
Article in English | MEDLINE | ID: mdl-30410131

ABSTRACT

OBJECTIVE: : To determine the expression of interferon alpha receptors 1 and 2 along with signal transducer and activator of transcription-1 in peripheral blood mononuclear cells of both hepatitis C mono-infected and hepatitis C and B co-infected patients, and to assess whether these targeted genes predict sustained virological response to interferon therapy. METHODS: This cross-sectional study was carried out at the Army Medical College, Rawalpindi, Pakistan, from January 2012 to December 2015, and comprised hepatitis C mono-infected and hepatitis C and B co-infected patients. The patients were divided into groups 1 and 2. Group-1a and group-2a consisted of mono-infected and co-infected sustained responders, while group-1b and group-2b had mono-infected and co-infected non sustained responders. Peripheral blood mononuclear cells from healthy controls were also quantified for these subunits. Target gene expressions were studied by retro-transcription of respective messenger ribonuclieic acid extracted from the cells followed by polymerase chain reaction amplification. RESULTS: Of the 191 subjects, there were 20(10.5%) in group-1a, 35(18.3%) in group-2a, 65(34%) in group-1b and 51(26.7%) in group-2b. The remaining 20(10.5%) were controls. Overall, 106 (55.5%) were males and 85 (44.5%) were females. Interferon alpha receptor 1 expression in groups 1a and 2a was significantly higher compared to groups 1b (p=0.018) and 2b (p 0.031). Signal transducer and activator of transcription-1 protein expression showed no significant difference (p=0.062 and p=0.519). No difference in expression was measured between the two sets of groups with regard to interferon alpha receptor 2 expression (p=0.278 and p=0.590). CONCLUSIONS: Our results show that levels of IFNAR-1 mRNA expression may be a good predictor for IFN-related anti-viral activity in both HCV mono infected and HCV/HBV co-infected patients.


Subject(s)
Gene Expression Regulation , Hepatitis B, Chronic/drug therapy , Hepatitis C, Chronic/drug therapy , Interferon alpha-2/therapeutic use , Interferon-alpha/therapeutic use , Polyethylene Glycols/therapeutic use , Receptor, Interferon alpha-beta/genetics , Ribavirin/therapeutic use , STAT1 Transcription Factor/genetics , Adult , Antibodies, Viral/analysis , Coinfection/drug therapy , Coinfection/metabolism , Coinfection/virology , Cross-Sectional Studies , DNA, Viral/analysis , Enzyme-Linked Immunosorbent Assay , Female , Hepacivirus/genetics , Hepacivirus/immunology , Hepatitis B virus/genetics , Hepatitis B virus/immunology , Hepatitis B, Chronic/metabolism , Hepatitis B, Chronic/virology , Hepatitis C, Chronic/metabolism , Hepatitis C, Chronic/virology , Humans , Male , Polymerase Chain Reaction , Receptor, Interferon alpha-beta/biosynthesis , Recombinant Proteins/therapeutic use , Retrospective Studies , STAT1 Transcription Factor/biosynthesis
13.
Eur Rev Med Pharmacol Sci ; 22(3): 582-589, 2018 02.
Article in English | MEDLINE | ID: mdl-29461585

ABSTRACT

OBJECTIVE: Spinal cord injury (SCI) is a severe trauma to the central nervous system. Long non-coding RNAs have been reported to play essential roles in spinal cord injury. This study mainly explored the role of micro-125 in the regulation of spinal cord injury by regulating STAT3. MATERIALS AND METHODS: The stable mouse model of cervical spinal cord contusion was established by Infinite Horizon spinal cord striker, and the model mice' motor function was analyzed. Bioinformatics databases were used to screen the target mRNAs of micro-125b. qRT-PCR was performed to detect the expression of micro-125b and its target genes in injury area of mice' spinal cord. Western Blot and ELISA were introduced to detect the expression of inflammation and apoptosis-related proteins in each group. The recovery status of spinal cord after SCI was assessed by motor function scores and axon counts of mice in each group. RESULTS: Micro-125b appeared to be significantly down-regulated over-time after SCI. JAK1 and STAT1, two important neuregulin proteins, were predicted to be the target genes of micro-125b, and overexpression of micro-125b induced the decrease of phosphorylated JAK1 and STAT1. Enhanced micro-125b expression also allowed axons from the injury area of spinal cord to extend into the outer periphery of the damaged area, thus improving the motor function of the injured rats. Besides, overexpression of micro-125b demonstrated significant neuronal protective effects by reducing apoptosis and inflammatory responses in neurons. CONCLUSIONS: Our data revealed that micro-125b was down-regulated in injured spinal cord, and overexpression of micro-125b promoted the repair and regeneration following spinal cord injury through the regulation of the JAK/STAT pathway.


Subject(s)
Apoptosis , Janus Kinase 1/metabolism , MicroRNAs/biosynthesis , STAT1 Transcription Factor/biosynthesis , Spinal Cord Injuries/metabolism , Animals , Axons/physiology , Down-Regulation , Inflammation/metabolism , Mice , Neurons/metabolism , Phosphorylation , Recovery of Function , Regeneration/physiology
14.
Int J Gynecol Cancer ; 28(3): 459-465, 2018 03.
Article in English | MEDLINE | ID: mdl-29303938

ABSTRACT

OBJECTIVE: Recently it has been demonstrated that constitutively activated signal transducer and activator of transcription 1 (STAT1) gene expression may act as a biomarker of ovarian cancer chemotherapy response. In this study, our objective was to validate the use of STAT1 immunohistochemistry as a prognostic biomarker for disease outcome using a cohort derived from Latin America. METHODS: We evaluated a cohort of Brazilian high-grade serous ovarian cancer, comprising 65 patients with outcome data covering more than 5 years to determine the prognostic and predictive value of STAT1 expression levels. High-grade serous ovarian cancer tumors were used to construct a tissue microarray. Exploratory analyses were conducted on clinical, histopathological, and STAT1 expression data that included descriptive statistics and Pearson correlative analyses. Survival curves for disease-free survival and overall survival were obtained by the Kaplan-Meier method, and the significance of homogeneity between the classes was assessed by log-rank statistics (Mantel-Cox). RESULTS: High expression of STAT1 in tumors was significantly associated with improved disease-free survival (P = 0.0256) and overall survival (P = 0.0193). Proportional hazards regression analysis showed STAT1 expression had an independent effect on both disease-free survival (P = 0.0358) and overall survival (P = 0.0469). CONCLUSIONS: These findings from a Brazilian cohort of patients with ovarian cancer reinforce the association of high STAT1 expression with better response to chemotherapy, providing additional validation of this protein as both a prognostic and predictive biomarker. Collectively, these results together with other recently published studies increase the feasibility of using the STAT1 pathway for the development of novel immunomodulator drugs that could enhance response to treatment.


Subject(s)
Cystadenocarcinoma, Serous/metabolism , Ovarian Neoplasms/metabolism , STAT1 Transcription Factor/biosynthesis , Adult , Aged , Biomarkers, Tumor/biosynthesis , Biomarkers, Tumor/genetics , Cohort Studies , Cystadenocarcinoma, Serous/genetics , Cystadenocarcinoma, Serous/pathology , Disease-Free Survival , Female , Humans , Immunohistochemistry , Middle Aged , Neoplasm Grading , Ovarian Neoplasms/genetics , Ovarian Neoplasms/pathology , Predictive Value of Tests , STAT1 Transcription Factor/genetics , Young Adult
15.
Int J Mol Med ; 40(3): 907-912, 2017 Sep.
Article in English | MEDLINE | ID: mdl-28677733

ABSTRACT

Sulforaphane (SFN) is a dietary isothiocyanate abundantly available in cruciferous vegetables and has been shown to possess anti-inflammatory and immunomodulatory activities. Chemokines are important mediators of inflammation and immune responses due to their ability to recruit and activate macrophages and leukocytes. To date, little is known about the SFN-mediated regulation of chemokine expression in pancreatic ß-cells. In this study, we investigated the inhibitory effects and mechanisms of SFN on the interferon-γ (IFN-γ)-induced expression of a subset of chemokines, including monokine induced by IFN-γ (MIG), IFN-inducible protein of 10 kDa (IP-10) and IFN-inducible T­cell alpha chemoattractant (I-TAC), in INS­1 cells, a rat pancreatic ß-cell line. Notably, IFN-γ treatment led to an increase in the mRNA expression levels of MIG, IP-10 and I-TAC in the INS­1 cells. However, SFN strongly blocked the mRNA expressions of MIG, IP-10 and I-TAC induced by IFN-γ in INS­1 cells. On the mechanistic level, SFN significanlty decreased not only the mRNA expression levels of interferon regulatory factor-1 (IRF-1), but also the phosphorylation levels of signal transducer and activator of transcription-1 (STAT-1) and protein kinase B (PKB) which were induced by IFN-γ in the INS­1 cells. Pharmacological inhibition experiments further revealed that treatment with JAK inhibitor I weakly inhibited the IFN-γ-induced expression of IP-10, whereas it strongly suppressed the IFN-γ-induced expression of MIG and I-TAC in the INS­1 cells. Moreover, treatment with LY294002, a PI3K/PKB inhibitor, was able to slightly repress IFN­Î³­induced expressions of MIG and I-TAC, but not IP-10, in INS­1 cells. Importantly, the IFN-γ-induced increase in the expression levels of MIG, IP-10 and I-TAC in the INS-1 cells was strongly inhibited by SFN, but not by other natural substances, such as curcumin, sanguinarine, resveratrol, triptolide and epigallocatechin gallate (EGCG), suggesting the specificity of SFN in downregulating the levels of these chemokines. To the best of our knowledge, these results collectively demonstrate for the first time that SFN strongly inhibits the IFN-γ-induced expression of MIG, IP-10 and I-TAC in INS­1 cells and this inhibition is, at least in part, mediated through the reduced expression and phosphorylation levels of IRF-1, STAT-1 and PKB.


Subject(s)
Chemokine CXCL10/biosynthesis , Chemokine CXCL11/biosynthesis , Chemokine CXCL9/biosynthesis , Gene Expression Regulation/drug effects , Insulin-Secreting Cells/metabolism , Interferon Regulatory Factor-1/biosynthesis , Interferon-gamma/pharmacology , Isothiocyanates/pharmacology , Proto-Oncogene Proteins c-akt/biosynthesis , STAT1 Transcription Factor/biosynthesis , Animals , Cell Line, Transformed , Insulin-Secreting Cells/cytology , Rats , Sulfoxides
16.
J Vector Borne Dis ; 54(2): 131-138, 2017.
Article in English | MEDLINE | ID: mdl-28748833

ABSTRACT

BACKGROUND & OBJECTIVES: Limited studies have been reported on Japanese encephalitis (JE) with reference to microarray data analysis. The present study involved an in silico approach for identification and characterization of differentially expressed genes in human microglial cell (CHME3) samples, infected with P20778 strain of Japanese encephalitis virus (JEV). METHODS: Gene expression data (GSE57330) belonging to mRNA expression profile of CHME3 cells infected with JEV, was downloaded from the gene expression omnibus (GEO) database, processed and normalized by robust multichip averaging (RMA) method using affy packages of R. The Bayes method was used to correct multiple testing. The log fold change (logFC > 1) and p< 0.05 were used as cut-off to identify differentially expressed genes (DEGs). The newly identified hub genes were set at the centre for construction of protein-protein interaction network using search tool for the retrieval of interacting genes/proteins (STRING) database considering human genome as reference. Gene ontology and pathway enrichment analysis of the hub gene and its associated genes were performed using STRING and DAVID tool. RESULTS: Microarray data analysis revealed that STAT1 gene was down-regulated during JEV infection. STAT1 gene was found to interact with tyrosine protein kinase family members, and showed strong interaction with JAK1 and JAK2 genes. INTERPRETATION & CONCLUSION: The identified transcription factors and the binding sites in the promoter region of STAT1 gene might act as potential drug targets in near future.


Subject(s)
Encephalitis Virus, Japanese/growth & development , Gene Expression Profiling , Host-Pathogen Interactions , Microglia/virology , Neuroglia/virology , Down-Regulation , Humans , Microarray Analysis , STAT1 Transcription Factor/biosynthesis
17.
Arch Oral Biol ; 82: 11-18, 2017 Oct.
Article in English | MEDLINE | ID: mdl-28578027

ABSTRACT

OBJECTIVE: This study was performed in an attempt to examine the influence of agomelatine in mitigating the generation of proinflammatory mediators in RAW264.7 murine macrophages exposed to lipopolysaccharide (LPS) obtained from Prevotella intermedia, a gram-negative anaerobic bacterium that is related with various types of periodontal diseases, and the molecular mechanisms behind its effects. DESIGN: LPS from P. intermedia strain ATCC 25611 was prepared employing the conventional phenol-water procedure. Conditioned culture media were analyzed for the levels of nitric oxide (NO), interleukin-1ß (IL-1ß) and IL-6. Real-time PCR analysis was carried out to determine the mRNA levels of inducible NO synthase (iNOS), IL-1ß, IL-6 and SOCS1. Protein expression levels were evaluated by immunoblot test. NF-κB-dependent SEAP reporter assay was performed using a reporter cell line. DNA-binding activities of NF-κB subunits were analyzed utilizing the ELISA-based kits. RESULTS: Agomelatine was found to down-regulate significantly the generation of iNOS-derived NO, IL-1ß and IL-6 as well as the expression of their mRNAs in cells activated with P. intermedia LPS. Agomelatine decreased NF-κB-dependent SEAP release caused by P. intermedia LPS. Agomelatine did not inhibit NF-κB transcription induced by LPS at the level of IκB-α degradation. Instead, LPS-induced nuclear translocation and DNA binding of NF-κB p50 subunit was blocked by agomelatine. P. intermedia LPS-elicited activation of STAT1 and STAT3 was reduced notably by co-treatment with agomelatine. Agomelatine showed a tendency to enhance mRNA level of SOCS1 in LPS-activated cells as well. CONCLUSIONS: Agomelatine merits further evaluation to reveal its usefulness on the host modulation of periodontal disease.


Subject(s)
Acetamides/pharmacology , Inflammation Mediators/antagonists & inhibitors , Macrophages/drug effects , Prevotella intermedia/drug effects , Serotonin 5-HT2 Receptor Antagonists/pharmacology , Animals , Cell Line , Enzyme-Linked Immunosorbent Assay , Immunoblotting , Interleukin-1beta/biosynthesis , Interleukin-6/biosynthesis , Lipopolysaccharides/pharmacology , Mice , NF-kappa B/biosynthesis , Nitric Oxide/biosynthesis , Nitric Oxide Synthase Type II/biosynthesis , RNA, Messenger/biosynthesis , Real-Time Polymerase Chain Reaction , STAT1 Transcription Factor/biosynthesis , STAT3 Transcription Factor/biosynthesis , Suppressor of Cytokine Signaling 1 Protein/biosynthesis
18.
Cell Physiol Biochem ; 42(2): 519-529, 2017.
Article in English | MEDLINE | ID: mdl-28578326

ABSTRACT

BACKGROUND/AIMS: Adult T-cell leukemia/lymphoma (ATL) is a very aggressive T cell malignancy that carries a poor prognosis, primarily due to its resistance to chemotherapy and to life-threatening infectious complications. Interferon-alpha (IFNα) has been used in combination with the anti-retroviral drug zidovudine to treat patients with ATL. However, the efficacy of long-term therapy is significantly limited due to the systemic toxicity of IFNα. METHODS: We utilized phage display library screening to identify short peptides that specifically bind to Jurkat T lymphocyte leukemia cells. By fusing the Jurkat-binding peptide to the C-terminus of IFNα, we constructed an engineered chimeric IFNα molecule (IFNP) for the treatment of ATL. RESULTS: We found that IFNP exhibited significantly higher activity than wild type IFNα in inhibiting the growth of leukemia cells and inducing cell blockage at the G0/G1 phase. The synthetic IFNP molecule exerted its antitumor activity by upregulating the downstream genes involved in the STAT1 pathway and in apoptosis. Using a cell receptor binding assay, we showed that this Jurkat-binding peptide facilitated the binding affinity of IFNα to the cell surface type I IFN receptor. CONCLUSION: The isolated Jurkat-binding peptide significantly potentiates the therapeutic activity of IFNα in T lymphocyte leukemia cells. The engineered IFNP molecule may prove to a novel antitumor approach in the treatment of patients with ATL.


Subject(s)
Interferon-alpha/genetics , Leukemia-Lymphoma, Adult T-Cell/genetics , Peptides/genetics , Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Apoptosis/drug effects , Cell Line, Tumor , Genes, Synthetic/genetics , Genetic Engineering , Humans , Interferon-alpha/administration & dosage , Interferon-alpha/adverse effects , Jurkat Cells , Leukemia-Lymphoma, Adult T-Cell/pathology , Leukemia-Lymphoma, Adult T-Cell/therapy , Peptide Library , Peptides/administration & dosage , STAT1 Transcription Factor/biosynthesis , STAT1 Transcription Factor/genetics , T-Lymphocytes/drug effects , T-Lymphocytes/pathology , Zidovudine/administration & dosage
19.
Oncotarget ; 8(28): 45249-45258, 2017 Jul 11.
Article in English | MEDLINE | ID: mdl-28431406

ABSTRACT

BACKGROUND: Esophageal squamous cell carcinoma is one of leading causes of cancer-related deaths in Chaoshan region a high-risk region for esophageal cancer. Extracellular regulated protein kinases (ERK) usually play an important role in cell proliferation and differentiation. However, accumulating evidence has shown that the ERK was aberrantly expressed in cancers and correlated with STAT1 depression. RESULTS: The activated ERK downregulates STAT1 expression in ESCC cell lines and U0126 increases expression of STAT1. Our immunohistochemistry result also confirms that the expression of ERK inversely correlated with that of STAT1 in ESCC tumors. In addition, a significantly higher expression of ERK/p-ERK was found in ESCC tissues in comparison with case-matched normal esophageal tissues (p < 0.05). Moreover, the immunohistochemical analysis demonstrated that ERK expression was paralleled with the differentiation and clinical stage. In 74 patients with follow-up data, those with ERKlow tumors survived significantly longer than those with ERKhigh tumors (p = 0.04); patients with ERKlow/STAT1high tumors had the longest survival (p = 0.001). MATERIALS AND METHODS: To investigate whether ERK can mediated STAT1 expression in ESCC, we used the MEK plasmid and U0126, a MEK inhibitor, to treat the cell. To further confirm our in-vitro study, we detected the ERK, p-ERK and STAT1 expression in 131 ESCC cases and 22 case-matched normal esophageal tissues adjacent to the tumors specimens. CONCLUSIONS: These findings provide pathological evidence that ERK/p-ERK is negatively correlated with STAT1 in ESCC. Our data suggests that inhibition of ERK and/or restoration of STAT1 expression maybe useful therapeutic strategies for ESCC.


Subject(s)
Carcinoma, Squamous Cell/metabolism , Esophageal Neoplasms/metabolism , Extracellular Signal-Regulated MAP Kinases/biosynthesis , STAT1 Transcription Factor/biosynthesis , Adult , Aged , Animals , Carcinoma, Squamous Cell/enzymology , Carcinoma, Squamous Cell/genetics , Carcinoma, Squamous Cell/pathology , Cell Line, Tumor , Down-Regulation , Enzyme Activation , Esophageal Neoplasms/enzymology , Esophageal Neoplasms/genetics , Esophageal Neoplasms/pathology , Esophageal Squamous Cell Carcinoma , Extracellular Signal-Regulated MAP Kinases/genetics , Extracellular Signal-Regulated MAP Kinases/metabolism , Female , Humans , Immunohistochemistry , MAP Kinase Signaling System , Male , Mice , Middle Aged , STAT1 Transcription Factor/genetics , STAT1 Transcription Factor/metabolism
20.
Sci Rep ; 7: 43958, 2017 03 07.
Article in English | MEDLINE | ID: mdl-28266659

ABSTRACT

We have analyzed the pathway networks of ischemia-affected and remote myocardial areas after repetitive ischemia/reperfusion (r-I/R) injury without ensuing myocardial infarction (MI) to elaborate a spatial- and chronologic model of cardioprotective gene networks to prevent left ventricular (LV) adverse remodeling. Domestic pigs underwent three cycles of 10/10 min r-I/R by percutaneous intracoronary balloon inflation/deflation in the mid left anterior descending artery, without consecutive MI. Sham interventions (n = 8) served as controls. Hearts were explanted at 5 h (n = 6) and 24 h (n = 6), and transcriptomic profiling of the distal (ischemia-affected) and proximal (non-affected) anterior myocardial regions were analyzed by next generation sequencing (NGS) and post-processing with signaling pathway impact and pathway network analyses. In ischemic region, r-I/R induced early activation of Ca-, adipocytokine and insulin signaling pathways with key regulator STAT3, which was also upregulated in the remote areas together with clusterin (CLU) and TNF-alpha. During the late phase of cardioprotection, antigen immunomodulatory pathways were activated with upregulation of STAT1 and CASP3 and downregulation of neprilysin in both zones, suggesting r-I/R induced intrinsic remote conditioning. The temporo-spatially differently activated pathways revealed a global myocardial response, and neprilysin and the STAT family as key regulators of intrinsic remote conditioning for prevention of adverse remodeling.


Subject(s)
Gene Regulatory Networks , Ischemia/pathology , Myocardial Reperfusion Injury/pathology , Physical Conditioning, Animal/methods , Signal Transduction , Ventricular Remodeling , Animals , Computational Biology , Disease Models, Animal , Gene Expression Profiling , High-Throughput Nucleotide Sequencing , Neprilysin/biosynthesis , STAT1 Transcription Factor/biosynthesis , STAT3 Transcription Factor/biosynthesis , Sus scrofa
SELECTION OF CITATIONS
SEARCH DETAIL
...