Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 937
Filter
Add more filters

Publication year range
1.
FASEB J ; 35(2): e21365, 2021 02.
Article in English | MEDLINE | ID: mdl-33496031

ABSTRACT

Besides its oncotic power, albumin exerts pleiotropic actions, including binding, transport, and detoxification of endogenous and exogenous molecules, antioxidant activity, and modulation of immune and inflammatory responses. In particular, recent studies have demonstrated that albumin reduces leukocyte cytokine production. Here, we investigated whether albumin also has the ability to protect tissues from the damaging actions of these inflammatory mediators. We circumscribed our investigation to tumor necrosis factor (TNF) α, which exemplifies the connection between immunity and tissue injury. In vivo experiments in analbuminemic mice showed that these mice exhibit a more pronounced response to a model of TNFα-mediated liver injury induced by the administration of lipopolysaccharide (LPS) and D-galactosamine (D-gal). A tissue protective action against LPS/D-gal liver injury was also observed during the administration of human albumin to humanized mice expressing the human genes for albumin and neonatal Fc receptor (hAlb+/+ /hFcRn+/+ ) with preestablished carbon tetrachloride (CCl4 )-induced early cirrhosis. The cytoprotective actions of albumin against TNFα-induced injury were confirmed ex vivo, in precision-cut liver slices, and in vitro, in primary hepatocytes in culture. Albumin protective actions were independent of its scavenging properties and were reproduced by recombinant human albumin expressed in Oryza sativa. Albumin cytoprotection against TNFα injury was related to inhibition of lysosomal cathepsin B leakage accompanied by reductions in mitochondrial cytochrome c release and caspase-3 activity. These data provide evidence that in addition to reducing cytokines, the albumin molecule also has the ability to protect tissues against inflammatory injury.


Subject(s)
Albumins/metabolism , Anti-Inflammatory Agents/pharmacology , Hepatocytes/metabolism , Liver Cirrhosis/metabolism , Tumor Necrosis Factor-alpha/toxicity , Albumins/pharmacology , Albumins/therapeutic use , Animals , Anti-Inflammatory Agents/therapeutic use , Carbon Tetrachloride/toxicity , Cells, Cultured , Hepatocytes/drug effects , Hepatocytes/pathology , Lipopolysaccharides/toxicity , Liver Cirrhosis/drug therapy , Liver Cirrhosis/etiology , Male , Mice , Mice, Inbred C57BL
2.
Proc Natl Acad Sci U S A ; 116(33): 16497-16506, 2019 08 13.
Article in English | MEDLINE | ID: mdl-31346084

ABSTRACT

Host inflammatory responses must be tightly regulated to ensure effective immunity while limiting tissue injury. IFN gamma (IFNγ) primes macrophages to mount robust inflammatory responses. However, IFNγ also induces cell death, and the pathways that regulate IFNγ-induced cell death are incompletely understood. Using genome-wide CRISPR/Cas9 screening, we identified autophagy genes as central mediators of myeloid cell survival during the IFNγ response. Hypersensitivity of autophagy gene-deficient cells to IFNγ was mediated by tumor necrosis factor (TNF) signaling via receptor interacting protein kinase 1 (RIPK1)- and caspase 8-mediated cell death. Mice with myeloid cell-specific autophagy gene deficiency exhibited marked hypersensitivity to fatal systemic TNF administration. This increased mortality in myeloid autophagy gene-deficient mice required the IFNγ receptor, and mortality was completely reversed by pharmacologic inhibition of RIPK1 kinase activity. These findings provide insight into the mechanism of IFNγ-induced cell death via TNF, demonstrate a critical function of autophagy genes in promoting cell viability in the presence of inflammatory cytokines, and implicate this cell survival function in protection against mortality during the systemic inflammatory response.


Subject(s)
Autophagy/genetics , Interferon-gamma/toxicity , Myeloid Cells/pathology , Tumor Necrosis Factor-alpha/toxicity , Animals , Autophagy/drug effects , Autophagy-Related Protein 5/metabolism , CRISPR-Cas Systems/genetics , Cell Line , Cell Survival/drug effects , Cell Survival/genetics , Cytoprotection/drug effects , Genome , Mice, Knockout , Myeloid Cells/drug effects , Myeloid Cells/metabolism , Myeloid Cells/ultrastructure , NF-kappa B/metabolism , Receptor-Interacting Protein Serine-Threonine Kinases/metabolism , Receptors, Tumor Necrosis Factor, Type I/metabolism , Signal Transduction/drug effects , Transcriptome/genetics
3.
Proteins ; 89(11): 1508-1521, 2021 11.
Article in English | MEDLINE | ID: mdl-34219271

ABSTRACT

Tumor necrosis factor-alpha (TNFα) inhibitors could prevent neurological disorders systemically, but their design generally relies on molecules unable to cross the blood-brain barrier (BBB). This research was aimed to design and characterize a novel TNFα inhibitor based on the angiopeptide-2 as a BBB shuttle molecule fused to the extracellular domain of human TNFα receptor 2 and a mutated vascular endothelial growth factor (VEGF) dimerization domain. This new chimeric protein (MTV) would be able to trigger receptor-mediated transcytosis across the BBB via low-density lipoprotein receptor-related protein-1 (LRP-1) and inhibit the cytotoxic effect of TNFα more efficiently because of its dimeric structure. Stably transformed CHO cells successfully expressed MTV, and its purification by Immobilized-Metal Affinity Chromatography (IMAC) rendered high purity degree. Mutated VEGF domain included in MTV did not show cell proliferation or angiogenic activities measured by scratch and aortic ring assays, which corroborate that the function of this domain is restricted to dimerization. The pairs MTV-TNFα (Kd 279 ± 40.9 nM) and MTV-LRP1 (Kd 399 ± 50.5 nM) showed high affinity by microscale thermophoresis, and a significant increase in cell survival was observed after blocking TNFα with MTV in a cell cytotoxicity assay. Also, the antibody staining in CHOK1 and bEnd3 cells demonstrated the adhesion of MTV to the LRP1 receptor located in the cell membrane. These results provide compelling evidence for the proper functioning of the three main domains of MTV individually, which encourage us to continue the research with this new molecule as a potential candidate for the systemic treatment of neurological disorders.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Endotoxins/antagonists & inhibitors , Low Density Lipoprotein Receptor-Related Protein-1/genetics , Peptides/genetics , Receptors, Tumor Necrosis Factor, Type II/genetics , Tumor Necrosis Factor-alpha/antagonists & inhibitors , Vascular Endothelial Growth Factor A/genetics , Animals , Anti-Inflammatory Agents/chemistry , Anti-Inflammatory Agents/metabolism , Blood-Brain Barrier/metabolism , CHO Cells , Cell Line , Cell Survival/drug effects , Cricetulus , Endothelial Cells/cytology , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Endotoxins/metabolism , Endotoxins/toxicity , Gene Expression , Humans , Low Density Lipoprotein Receptor-Related Protein-1/metabolism , Mice , Models, Biological , Models, Molecular , Peptides/metabolism , Protein Binding , Protein Conformation , Protein Engineering/methods , Receptors, Tumor Necrosis Factor, Type II/metabolism , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Recombinant Fusion Proteins/pharmacology , Tumor Necrosis Factor-alpha/metabolism , Tumor Necrosis Factor-alpha/toxicity , Vascular Endothelial Growth Factor A/metabolism
4.
Proteins ; 89(11): 1557-1564, 2021 11.
Article in English | MEDLINE | ID: mdl-34250652

ABSTRACT

The neutralization of tumor necrosis factor alpha (TNFα) with biopharmaceuticals is a successful therapy for inflammatory diseases. Currently, one of the main TNFα-antagonists is Etanercept, a dimeric TNF-R2 ectodomain. Considering that TNFα and its receptors are homotrimers, we proposed that a trimeric TNF-R2 ectodomain could be an innovative TNFα-antagonist. Here, the 3cTNFR2 protein was designed by the fusion of the TNF-R2 ectodomain with the collagen XV trimerization domain. 3cTNFR2 was produced in HEK293 cells and purified by immobilized metal affinity chromatography. Monomers, dimers, and trimers of 3cTNFR2 were detected. The interaction 3cTNFR2-TNFα was assessed. By microscale thermophoresis, the KD value for the interaction was 4.17 ± 0.88 nM, and complexes with different molecular weights were detected by size exclusion chromatography-high performance liquid chromatography. Moreover, 3cTNFR2 neutralized the TNFα-induced cytotoxicity totally in vitro. Although more studies are required to evaluate the anti-inflammatory effect, the results suggest that 3cTNFR2 could be a TNFα-antagonist agent.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Collagen/genetics , Endotoxins/antagonists & inhibitors , Etanercept/pharmacology , Receptors, Tumor Necrosis Factor, Type II/genetics , Tumor Necrosis Factor-alpha/antagonists & inhibitors , Anti-Inflammatory Agents/chemistry , Anti-Inflammatory Agents/metabolism , Cell Survival/drug effects , Collagen/metabolism , Endotoxins/metabolism , Endotoxins/toxicity , Etanercept/chemistry , Etanercept/metabolism , Gene Expression , HEK293 Cells , Humans , Models, Molecular , Protein Binding , Protein Conformation , Protein Domains , Protein Engineering/methods , Protein Multimerization , Receptors, Tumor Necrosis Factor, Type II/metabolism , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Recombinant Fusion Proteins/pharmacology , Tumor Necrosis Factor-alpha/metabolism , Tumor Necrosis Factor-alpha/toxicity
5.
Biochem Cell Biol ; 99(4): 488-498, 2021 08.
Article in English | MEDLINE | ID: mdl-34297624

ABSTRACT

Although tumor necrosis factor α (TNF-α)-mediated inflammation significantly impacts osteoporosis, the mechanisms underlying the osteogenic differentiation defects of bone marrow-derived mesenchymal stem cells (BM-MSCs) caused by TNF-α remain poorly understood. We found that TNF-α stimulation of murine BM-MSCs significantly upregulated the expression levels of several microRNAs (miRNAs), including let-7f-5p, but this increase was significantly reversed by treatment with the kinase inhibitor BAY 11-7082. To study gain- or loss of function, we transfected cells with an miRNA inhibitor or miRNA mimic. We then demonstrated that let-7f-5p impaired osteogenic differentiation of BM-MSCs in the absence and presence of TNF-α, as evidenced by alkaline phosphatase and alizarin red staining as well as quantitative assays of the mRNA levels of bone formation marker genes in differentiated BM-MSCs. Moreover, let-7f-5p targets the 3' untranslated region of Nucleoside diphosphate kinase 4 (Nme4) mRNA and negatively regulates Nme4 expression in mouse BM-MSCs. Ectopic expression of Nme4 completely reversed the inhibitory effects of the let-7f-5p mimic on osteogenic differentiation of mouse BM-MSCs. Furthermore, inhibition of let-7f-5p or overexpression of Nme4 in BM-MSCs restored in-vivo bone formation in an ovariectomized animal model. Collectively, our work indicates that let-7f-5p is involved in TNF-α-mediated reduction of BM-MSC osteogenesis via targeting Nme4.


Subject(s)
Bone Resorption/pathology , Cell Differentiation , Mesenchymal Stem Cells/pathology , MicroRNAs/genetics , Nucleoside Diphosphate Kinase D/metabolism , Osteogenesis , Tumor Necrosis Factor-alpha/toxicity , Animals , Bone Resorption/etiology , Bone Resorption/metabolism , Female , Mesenchymal Stem Cells/drug effects , Mesenchymal Stem Cells/metabolism , Mice , Mice, Inbred C57BL , Nucleoside Diphosphate Kinase D/genetics , Ovariectomy/adverse effects
6.
J Virol ; 94(21)2020 10 14.
Article in English | MEDLINE | ID: mdl-32847859

ABSTRACT

Interferon (IFN) family cytokines stimulate genes (interferon-stimulated genes [ISGs]) that are integral to antiviral host defense. Type I IFNs act systemically, whereas type III IFNs act preferentially at epithelial barriers. Among barrier cells, intestinal epithelial cells (IECs) are particularly dependent on type III IFN for the control and clearance of virus infection, but the physiological basis of this selective IFN response is not well understood. Here, we confirm that type III IFN treatment elicits robust and uniform ISG expression in neonatal mouse IECs and inhibits the replication of IEC-tropic rotavirus. In contrast, type I IFN elicits a marginal ISG response in neonatal mouse IECs and does not inhibit rotavirus replication. In vitro treatment of IEC organoids with type III IFN results in ISG expression that mirrors the in vivo type III IFN response. However, IEC organoids have increased expression of the type I IFN receptor relative to neonate IECs, and the response of IEC organoids to type I IFN is strikingly increased in magnitude and scope relative to type III IFN. The expanded type I IFN-specific response includes proapoptotic genes and potentiates toxicity triggered by tumor necrosis factor alpha (TNF-α). The ISGs stimulated in common by type I and III IFNs have strong interferon-stimulated response element (ISRE) promoter motifs, whereas the expanded set of type I IFN-specific ISGs, including proapoptotic genes, have weak ISRE motifs. Thus, the preferential responsiveness of IECs to type III IFN in vivo enables selective ISG expression during infection that confers antiviral protection but minimizes disruption of intestinal homeostasis.IMPORTANCE Enteric viral infections are a major cause of gastroenteritis worldwide and have the potential to trigger or exacerbate intestinal inflammatory diseases. Prior studies have identified specialized innate immune responses that are active in the intestinal epithelium following viral infection, but our understanding of the benefits of such an epithelium-specific response is incomplete. Here, we show that the intestinal epithelial antiviral response is programmed to enable protection while minimizing epithelial cytotoxicity that can often accompany an inflammatory response. Our findings offer new insight into the benefits of a tailored innate immune response at the intestinal barrier and suggest how dysregulation of this response could promote inflammatory disease.


Subject(s)
Cytokines/immunology , Intestinal Mucosa/immunology , Rotavirus Infections/immunology , STAT1 Transcription Factor/immunology , STAT2 Transcription Factor/immunology , Tumor Necrosis Factor-alpha/toxicity , Animals , Animals, Newborn , Cytokines/genetics , Epithelial Cells/drug effects , Epithelial Cells/immunology , Epithelial Cells/virology , Gene Expression Regulation , Humans , Interferon-Stimulated Gene Factor 3, gamma Subunit/genetics , Interferon-Stimulated Gene Factor 3, gamma Subunit/immunology , Intestinal Mucosa/drug effects , Intestinal Mucosa/virology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Organoids/drug effects , Organoids/immunology , Organoids/virology , Response Elements , Rotavirus/drug effects , Rotavirus/growth & development , Rotavirus/pathogenicity , Rotavirus Infections/genetics , Rotavirus Infections/pathology , Rotavirus Infections/virology , STAT1 Transcription Factor/genetics , STAT2 Transcription Factor/genetics , Signal Transduction , Virus Replication
7.
Microvasc Res ; 133: 104093, 2021 01.
Article in English | MEDLINE | ID: mdl-33007316

ABSTRACT

Acute respiratory distress syndrome (ARDS) is a rapidly progressive disease with unknown pathogenesis. Damage of pulmonary microvascular endothelial cells (PMVECs) caused by inflammatory storm caused by cytokines such as TNF-α is the potential pathogenesis of ARDS. In this study, we examined the role of ezrin and Rac1 in TNF-α-related pathways, which regulates the permeability of PMVECs. Primary rat pulmonary microvascular endothelial cells (RPMVECs) were isolated and cultured. RPMVECs were treated with rat TNF-α (0, 1, 10, 100 ng/ml), and the cell activity of each group was measured using a CCK8 kit. The integrity of endothelial barrier was measured by transendothelial resistance (TEER) and FITC-BSA flux across RPMVECs membranes. Pulldown assay and Western blot was used to detect the activity of RAS-associated C3 botulinum toxin substrate 1 (Rac1) and Ezrin phosphorylation. Short hairpin RNA (shRNA) targeting ezrin and Rac1 was utilized to evaluate the effect of RPMVECs permeability and related pathway. The effects of ezrin and Rac1 on cytoskeleton were confirmed by immunofluorescence. Our results revealed that active Rac1 was essential for protecting the RPMVEC barrier stimulated by TNF-α, while active ezrin could partially destroy the PMVEC barrier by reducing Rac1 activity and regulating the subcellular structure of the cytoskeleton. These findings may be used to create new therapeutic strategies for targeting Rac1 in the treatment of ARDS.


Subject(s)
Capillary Permeability/drug effects , Cytoskeletal Proteins/metabolism , Cytoskeleton/drug effects , Endothelial Cells/drug effects , Lung/blood supply , Microvessels/drug effects , Tumor Necrosis Factor-alpha/toxicity , Animals , Cells, Cultured , Cytoskeletal Proteins/genetics , Cytoskeleton/metabolism , Cytoskeleton/pathology , Electric Impedance , Endothelial Cells/metabolism , Endothelial Cells/pathology , Male , Microvessels/metabolism , Microvessels/pathology , Phosphorylation , Rats, Sprague-Dawley , rac1 GTP-Binding Protein/genetics , rac1 GTP-Binding Protein/metabolism
8.
J Cardiovasc Pharmacol ; 77(5): 570-577, 2021 05 01.
Article in English | MEDLINE | ID: mdl-33657050

ABSTRACT

ABSTRACT: This study aimed to evaluate the protective effect of quercetin and its in-depth mechanism in TNF-α-stimulated cardiomyocytes. The differential expression of TNF-alpha (TNF-α) and signal transducer and activator of transcription 1 (STAT1) was analyzed based on the GEO database. H9c2 cells were stimulated with TNF-α to simulate myocarditis. Cell counting kit-8 assay and flow cytometry assay were performed to detect the cell viability and apoptosis. ELISA was used to measure the levels of proinflammatory cytokines (IL-6 and IL-17A) and anti-inflammatory cytokine (IL-10). STAT1 expression was downregulated by transfection with si-STAT1, and its expression was detected using quantitative real-time polymerase chain reaction and Western blot. Western blot was also performed to assess the expression of the mitogen-activated protein kinase (MAPK) pathway-related factors. In this article, TNF-α was highly expressed in patients with myocarditis, and TNF-α (20 µg/mL) declined the viability of H9c2 cells. Quercetin pretreatment partially alleviated the decrease of cell viability, the increase of apoptosis, and the release of inflammatory cytokines (IL-10, IL-6, and IL-17A) induced by TNF-α. In addition, TNF-α increased STAT1 expression, but quercetin prevented the TNF-α-increased STAT1 level. Remarkably, knockdown of STAT1 enhanced the protective effect of quercetin on TNF-α-injured H9c2 cells. Moreover, quercetin restrained the TNF-α-induced activation of the MAPK pathway. Also, the inhibitory effect of quercetin on the pathway was aggravated by STAT1 lacking. In summing, quercetin plays a protective role in TNF-α-stimulated H9c2 cell injury, which may be related to the regulation of STAT1 and MAPK pathway.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Mitogen-Activated Protein Kinases/metabolism , Myocarditis/drug therapy , Myocytes, Cardiac/drug effects , Quercetin/pharmacology , STAT1 Transcription Factor/metabolism , Animals , Case-Control Studies , Cell Line , Humans , Interleukin-10/metabolism , Interleukin-17/metabolism , Interleukin-6/metabolism , Myocarditis/enzymology , Myocarditis/genetics , Myocarditis/pathology , Myocytes, Cardiac/enzymology , Myocytes, Cardiac/pathology , Rats , STAT1 Transcription Factor/genetics , Signal Transduction , Tumor Necrosis Factor-alpha/toxicity
9.
Biol Pharm Bull ; 44(1): 18-24, 2021.
Article in English | MEDLINE | ID: mdl-33390545

ABSTRACT

Deeper wrinkles and loss of elasticity are one of the skin-aging symptoms. Collagen breakdown by matrix metalloproteinase-1 (MMP-1), which is induced by reactive oxygen species (ROS) and pro-inflammatory cytokines, has been known to be responsible for these skin-aging symptoms. Therefore, much attention has been paid to chemicals to suppress the MMP-1 activity. Epigallocatechin-3-gallate (EGCG), catechin rich in green tea, has been reported to show antioxidant and protect skin from various stimuli such as UV and chemicals. In this study, we evaluated the inhibitory effect of EGCG on MMP-1 gene expression and secretion in tumor necrosis factor-α (TNF-α)-treated human dermal fibroblast cells (Hs68 cells). Pre-treatment with EGCG (10 and 20 µM) suppressed TNF-α-induced MMP-1 expression and secretion. EGCG also reduced the phosphorylation of extracellular signal regulated kinase (ERK) significantly but not that of p38 activation and c-Jun N-terminal kinase (JNK). Besides, EGCG (10 and 20 µM) showed the inhibitory effect on mitogen-activated protein extracellular kinase (MEK) and Src phosphorylation which is reported to be upstream signal proteins of ERK signal pathway. Based on these results, EGCG might have potential activity to slow down the skin-aging through inhibition of collagen breakdown, which remains to be elucidated.


Subject(s)
Antioxidants/pharmacology , Catechin/analogs & derivatives , Fibroblasts/drug effects , MAP Kinase Signaling System/drug effects , Matrix Metalloproteinase 1/biosynthesis , Tumor Necrosis Factor-alpha/toxicity , Catechin/pharmacology , Dose-Response Relationship, Drug , Fibroblasts/enzymology , Gene Expression Regulation, Enzymologic , Humans , MAP Kinase Signaling System/physiology , Reactive Oxygen Species/antagonists & inhibitors , Reactive Oxygen Species/metabolism , Skin Aging/drug effects , Skin Aging/physiology , Tumor Necrosis Factor-alpha/antagonists & inhibitors
10.
Int J Mol Sci ; 22(19)2021 Sep 23.
Article in English | MEDLINE | ID: mdl-34638561

ABSTRACT

Tumor necrosis factor (TNF) is a regulator of several chronic inflammatory diseases, such as rheumatoid arthritis. Although anti-TNF biologics have been used in clinic, they render several drawbacks, such as patients' progressive immunodeficiency and loss of response, high cost, and intravenous administration. In order to find new potential anti-TNF small molecule inhibitors, we employed an in silico approach, aiming to find natural products, analogs of Ampelopsin H, a compound that blocks the formation of TNF active trimer. Two out of nine commercially available compounds tested, Nepalensinol B and Miyabenol A, efficiently reduced TNF-induced cytotoxicity in L929 cells and production of chemokines in mice joints' synovial fibroblasts, while Nepalensinol B also abolished TNF-TNFR1 binding in non-toxic concentrations. The binding mode of the compounds was further investigated by molecular dynamics and free energy calculation studies, using and advancing the Enalos Asclepios pipeline. Conclusively, we propose that Nepalensinol B, characterized by the lowest free energy of binding and by a higher number of hydrogen bonds with TNF, qualifies as a potential lead compound for TNF inhibitors' drug development. Finally, the upgraded Enalos Asclepios pipeline can be used for improved identification of new therapeutics against TNF-mediated chronic inflammatory diseases, providing state-of-the-art insight on their binding mode.


Subject(s)
Anti-Inflammatory Agents/chemistry , Anti-Inflammatory Agents/pharmacology , Biological Products/chemistry , Biological Products/pharmacology , Drug Discovery/methods , Tumor Necrosis Factor Inhibitors/chemistry , Tumor Necrosis Factor Inhibitors/pharmacology , Animals , Binding Sites/drug effects , Cell Line , Cell Survival/drug effects , Computer Simulation , Drug Design , Fibroblasts/drug effects , Mice , Primary Cell Culture , Synovial Fluid/drug effects , Tumor Necrosis Factor-alpha/toxicity
11.
Biochem Biophys Res Commun ; 531(3): 383-389, 2020 10 20.
Article in English | MEDLINE | ID: mdl-32800547

ABSTRACT

Tumor necrosis factor-alpha (TNF-α), a major inflammatory factor released from activated retinal glial cells, is implicated in the pathogenesis of glaucoma. In this study, we investigated whether and how TNF-α may affect functional conditions of activated retinal Müller cells. Our results showed that in the group I metabotropic glutamate receptor (mGluR I) agonist DHPG-activated cultured Müller cells, TNF-α treatment aggravated cell gliosis, as evidenced by significantly increased expression of glial fibrillary acidic protein (GFAP). TNF-α treatment of the DHPG-activated Müller cells decreased cell proliferation and induced cell apoptosis. In normal Müller cells, TNF-α treatment increased the mRNA levels of leukocyte inhibitory factor (LIF), intercellular cell adhesion molecule (ICAM), vascular cell adhesion molecule (VCAM), and chemokine C-C-motif ligand 2 (CCL2), which could be significantly attenuated when Müller cells were pre-activated. However, TNF-α-induced elevation in mRNA levels of inflammatory factors, such as TNF-α, inducible nitric oxide synthase (iNOS), and interleukin-6 (IL-6), in normal Müller cells still kept higher levels when Müller cells were pre-activated. Furthermore, the TNF-α-induced changes of cytokines were partially mediated by NF-κB signaling pathway. Our results suggest that TNF-α may promote gliosis and inflammatory response of activated Müller cells, thus aggravating RGC injury in glaucoma.


Subject(s)
Ependymoglial Cells/pathology , Gliosis/pathology , Inflammation/pathology , Tumor Necrosis Factor-alpha/toxicity , Animals , Apoptosis/drug effects , Cell Proliferation/drug effects , Cell Survival/drug effects , Cells, Cultured , Cytokines/metabolism , Ependymoglial Cells/drug effects , Ependymoglial Cells/metabolism , Glial Fibrillary Acidic Protein/metabolism , Gliosis/complications , Inflammation/complications , Methoxyhydroxyphenylglycol/analogs & derivatives , Methoxyhydroxyphenylglycol/pharmacology , Mice, Inbred C57BL , NF-kappa B/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Signal Transduction/drug effects
12.
Mol Cell Probes ; 52: 101538, 2020 08.
Article in English | MEDLINE | ID: mdl-32084581

ABSTRACT

Tumor necrosis factor-alpha (TNF-α) has been shown to have an inhibitory effect on the osteogenic differentiation of mesenchymal stem cells. The metabolic switch from glycolysis to oxidative phosphorylation (OXPHOS) is vital for energy supply during osteogenic differentiation. However, the metabolic switch is inhibited under inflammatory stimulation. FGF2 has shown that it can improve osteogenic differentiation and promote autoimmune inflammation. In this study, we investigated whether FGF2 can ameliorate TNF-a-inhibited osteogenic damage by improving OXPHOS. Effects of TNF-α or FGF2 on the proliferation and osteogenic differentiation of hBMSCs were evaluated by MTT assay, qRT-PCR, and ALP activity tests. The function of FGF2 on the TNF-a-inhibited metabolic switch was determined by Mito Stress test. The results showed that TNF-α was able to inhibit the osteogenic differentiation and OXPHOS of hBMSCs. FGF2 has no obvious function in improving the osteogenic-related genes, but it can ameliorate the impaired osteogenesis and OCR value caused by TNF-α. These findings suggest that FGF2 can prevent the impaired osteogenic differentiation and metabolic switch of hBMSCs under inflammatory stimulation, which might enhance the regeneration capacity of hBMSCs.


Subject(s)
Fibroblast Growth Factor 2/pharmacology , Mesenchymal Stem Cells/metabolism , Mesenchymal Stem Cells/pathology , Osteogenesis/drug effects , Oxidative Phosphorylation/drug effects , Tumor Necrosis Factor-alpha/toxicity , Apoptosis/drug effects , Cell Differentiation/drug effects , Cell Proliferation/drug effects , Glycolysis/drug effects , Humans , Mesenchymal Stem Cells/drug effects
13.
Clin Exp Pharmacol Physiol ; 47(3): 432-438, 2020 03.
Article in English | MEDLINE | ID: mdl-31713877

ABSTRACT

Antiplatelet drugs are conventionally used as treatments because of their anti-coagulation functions. However, their pleiotropic effects are of great significance to the treatment of ischaemic cardiovascular diseases. Many studies have reported that an excessive amount of inflammation driven by tumour necrosis factor (TNF) is closely related to the prevalence of atherosclerosis. As the drug selection criteria and evaluation methods related to the anti-TNF activity of antiplatelet drugs remain limited, our investigation of these drugs should prove beneficial. In this study, we compared the anti-TNF activity of three antiplatelet agents, namely clopidogrel, sarpogrelate, and cilostazol, using the TNF-induced inflammatory mouse model. After the oral administration of these drugs, acute inflammation was induced via injection of lipopolysaccharide (LPS) or D-galactosamine (D-gal) and TNF. Serum TNF levels, and the mRNA and protein expression levels of TNF in mouse heart tissue, macrophage accumulation in aortic lesions, and mouse survival were analysed to compare the anti-TNF effects of the three antiplatelet agents. Of the three antiplatelet agents, cilostazol significantly reduced the different levels under the most effective observation. In addition, cilostazol was found to attenuate the TNF-stimulated phosphorylation of mitogen-activated protein kinase (MAPK) and nuclear factor kappa-light-chain-enhancer of activated B cell (NF-κB) p65 in the aortic vascular smooth muscle cell line, MOVAS-1 and the D-gal plus TNF-challenged heart tissue of mouse. Therefore, cilostazol is the most ideal of the three antiplatelet drugs for the treatment of TNF-mediated inflammatory disorders.


Subject(s)
Disease Models, Animal , Inflammation Mediators/antagonists & inhibitors , Inflammation Mediators/metabolism , Platelet Aggregation Inhibitors/therapeutic use , Tumor Necrosis Factor-alpha/toxicity , Animals , Cilostazol/pharmacology , Cilostazol/therapeutic use , Clopidogrel/pharmacology , Clopidogrel/therapeutic use , Inflammation/chemically induced , Inflammation/drug therapy , Inflammation/metabolism , Male , Mice , Mice, Inbred C57BL , Platelet Aggregation Inhibitors/pharmacology , Treatment Outcome
14.
J Neurosci ; 38(43): 9160-9174, 2018 10 24.
Article in English | MEDLINE | ID: mdl-30201771

ABSTRACT

To date, histone H2B monoubiquitination (H2Bub), a mark associated with transcriptional elongation and ongoing transcription, has not been linked to the development or maintenance of neuropathic pain states. Here, using male Sprague Dawley rats, we demonstrated spinal nerve ligation (SNL) induced behavioral allodynia and provoked ring finger protein 20 (RNF20)-dependent H2Bub in dorsal horn. Moreover, SNL provoked RNF20-mediated H2Bub phosphorylated RNA polymerase II (RNAPII) in the promoter fragments of mGluR5, thereby enhancing mGluR5 transcription/expression in the dorsal horn. Conversely, focal knockdown of spinal RNF20 expression reversed not only SNL-induced allodynia but also RNF20/H2Bub/RNAPII phosphorylation-associated spinal mGluR5 transcription/expression. Notably, TNF-α injection into naive rats and specific neutralizing antibody injection into SNL-induced allodynia rats revealed that TNF-α-associated allodynia involves the RNF20/H2Bub/RNAPII transcriptional axis to upregulate mGluR5 expression in the dorsal horn. Collectively, our findings indicated TNF-α induces RNF20-drived H2B monoubiquitination, which facilitates phosphorylated RNAPII-dependent mGluR5 transcription in the dorsal horn for the development of neuropathic allodynia.SIGNIFICANCE STATEMENT Histone H2B monoubiquitination (H2Bub), an epigenetic post-translational modification, positively correlated with gene expression. Here, TNF-α participated in neuropathic pain development by enhancing RNF20-mediated H2Bub, which facilitates phosphorylated RNAPII-dependent mGluR5 transcription in dorsal horn. Our finding potentially identified neuropathic allodynia pathophysiological processes underpinning abnormal nociception processing and opens a new avenue for the development of novel analgesics.


Subject(s)
Histones/metabolism , Neuralgia/metabolism , Posterior Horn Cells/metabolism , Receptor, Metabotropic Glutamate 5/metabolism , Ubiquitin-Protein Ligases/metabolism , Ubiquitination/physiology , Animals , Histones/genetics , Male , Neuralgia/chemically induced , Neuralgia/genetics , Posterior Horn Cells/drug effects , Rats , Rats, Sprague-Dawley , Receptor, Metabotropic Glutamate 5/genetics , Transcription, Genetic/drug effects , Transcription, Genetic/physiology , Tumor Necrosis Factor-alpha/toxicity , Ubiquitin-Protein Ligases/genetics , Ubiquitination/drug effects
15.
J Cell Mol Med ; 23(5): 3336-3344, 2019 05.
Article in English | MEDLINE | ID: mdl-30761739

ABSTRACT

The mechanisms of radiation-induced liver damage are poorly understood. We investigated if tumour necrosis factor (TNF)-α acts synergistically with irradiation, and how its activity is influenced by platelet endothelial cell adhesion molecule-1 (PECAM-1). We studied murine models of selective single-dose (25 Gy) liver irradiation with and without TNF-α application (2 µg/mouse; i.p.). In serum of wild-type (wt)-mice, irradiation induced a mild increase in hepatic damage marker aspartate aminotransferase (AST) in comparison to sham-irradiated controls. AST levels further increased in mice treated with both irradiation and TNF-α. Accordingly, elevated numbers of leucocytes and increased expression of the macrophage marker CD68 were observed in the liver of these mice. In parallel to hepatic damage, a consecutive decrease in expression of hepatic PECAM-1 was found in mice that received radiation or TNF-α treatment alone. The combination of radiation and TNF-α induced an additional significant decline of PECAM-1. Furthermore, increased expression of hepatic lipocalin-2 (LCN-2), a hepatoprotective protein, was detected at mRNA and protein levels after irradiation or TNF-α treatment alone and the combination of both. Signal transducer and activator of transcription-3 (STAT-3) seems to be involved in the signalling cascade. To study the involvement of PECAM-1 in hepatic damage more deeply, the liver of both wt- and PECAM-1-knock-out-mice were selectively irradiated (25 Gy). Thereby, ko-mice showed higher liver damage as revealed by elevated AST levels, but also increased hepatoprotective LCN-2 expression. Our studies show that TNF-α has a pivotal role in radiation-induced hepatic damage. It acts in concert with irradiation and its activity is modulated by PECAM-1, which mediates pro- and anti-inflammatory signalling.


Subject(s)
Liver/metabolism , Liver/pathology , Platelet Endothelial Cell Adhesion Molecule-1/metabolism , Radiation, Ionizing , Tumor Necrosis Factor-alpha/toxicity , Animals , Antigens, CD/metabolism , Antigens, Differentiation, Myelomonocytic/metabolism , Aspartate Aminotransferases/blood , Kinetics , Leukocytes/metabolism , Lipocalin-2/metabolism , Liver/radiation effects , Male , Mice, Inbred C57BL , Mice, Knockout , Phosphorylation/radiation effects , RNA, Messenger/genetics , RNA, Messenger/metabolism , STAT3 Transcription Factor/metabolism
16.
J Cell Mol Med ; 23(7): 4666-4678, 2019 07.
Article in English | MEDLINE | ID: mdl-31033175

ABSTRACT

The traditional Chinese herb Lonicerae Japonicae Flos has shown significant clinical benefits in the treatment of heart failure, but the mechanism remains unclear. As the main active ingredient found in the plasma after oral administration of Lonicerae Japonicae Flos, chlorogenic acid (CGA) has been reported to possess anti-inflammatory, anti-oxidant and anti-apoptosis function. We firstly confirmed the cardioprotective effects of CGA in transverse aortic constriction (TAC)-induced heart failure mouse model, through mitigating the TNF-α-induced toxicity. We further used TNF-α-induced cardiac injury in human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) to elucidate the underlying mechanisms. CGA pre-treatment could reverse TNF-α-induced cellular injuries, including improved cell viability, increased mitochondrial membrane potential and inhibited cardiomyocytes apoptosis. We then examined the NF-κB/p65 and major mitogen-activated protein kinases (MAPKs) signalling pathways involved in TNF-α-induced apoptosis of hiPSC-CMs. Importantly, CGA can directly inhibit NF-κB signal by suppressing the phosphorylation of NF-κB/p65. As for the MAPKs, CGA suppressed the activity of only c-Jun N-terminal kinase (JNK), but enhanced extracellular signal-regulated kinase1/2 (ERK1/2) and had no effect on p38. In summary, our study revealed that CGA has profound cardioprotective effects through inhibiting the activation of NF-κB and JNK pathway, providing a novel therapeutic alternative for prevention and treatment of heart failure.


Subject(s)
Chlorogenic Acid/pharmacology , Cytoprotection/drug effects , MAP Kinase Signaling System/drug effects , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , NF-kappa B/metabolism , Tumor Necrosis Factor-alpha/toxicity , Animals , Aorta/pathology , Apoptosis/drug effects , Cardiotonic Agents/pharmacology , Cell Survival/drug effects , Chlorogenic Acid/therapeutic use , Constriction, Pathologic , Disease Models, Animal , Heart Failure/drug therapy , Induced Pluripotent Stem Cells/cytology , Male , Mice, Inbred C57BL , Models, Biological , Myocytes, Cardiac/drug effects , Stroke Volume/drug effects
17.
J Cell Mol Med ; 23(11): 7641-7650, 2019 11.
Article in English | MEDLINE | ID: mdl-31496037

ABSTRACT

Tumour necrosis factor (TNF)-α induces cardiac metabolic disorder and mitochondrial dysfunction. Hydrogen sulphide (H2 S) contains anti-inflammatory and biological effects in cardiomyocytes. This study investigated whether H2 S modulates TNF-α-dysregulated mitochondrial function and metabolism in cardiomyocytes. HL-1 cells were incubated with TNF-α (25 ng/mL) with or without sodium hydrosulphide (NaHS, 0.1 mmol/L) for 24 hours. Cardiac peroxisome proliferator-activated receptor (PPAR) isoforms, pro-inflammatory cytokines, receptor for advanced glycation end products (RAGE) and fatty acid metabolism were evaluated through Western blotting. The mitochondrial oxygen consumption rate and adenosine triphosphate (ATP) production were investigated using Seahorse XF24 extracellular flux analyzer and bioluminescence assay. Fluorescence intensity using 2', 7'-dichlorodihydrofluorescein diacetate was used to evaluate mitochondrial oxidative stress. NaHS attenuated the impaired basal and maximal respiration, ATP production and ATP synthesis and enhanced mitochondrial oxidative stress in TNF-α-treated HL-1 cells. TNF-α-treated HL-1 cells exhibited lower expression of PPAR-α, PPAR-δ, phosphorylated 5' adenosine monophosphate-activated protein kinase-α2, phosphorylated acetyl CoA carboxylase, carnitine palmitoyltransferase-1, PPAR-γ coactivator 1-α and diacylglycerol acyltransferase 1 protein, but higher expression of PPAR-γ, interleukin-6 and RAGE protein than control or combined NaHS and TNF-α-treated HL-1 cells. NaHS modulates the effects of TNF-α on mitochondria and the cardiometabolic system, suggesting its therapeutic potential for inflammation-induced cardiac dysfunction.


Subject(s)
Mitochondria/metabolism , Mitochondria/pathology , Sulfides/pharmacology , Tumor Necrosis Factor-alpha/toxicity , Adenosine Triphosphate/biosynthesis , Animals , Cell Line , Cytokines/metabolism , Fatty Acids/metabolism , Glucose/metabolism , Inflammation Mediators/metabolism , Insulin/metabolism , Mice , Mitochondria/drug effects , Myocardium/metabolism , Oxidative Stress/drug effects , Peroxisome Proliferator-Activated Receptors/metabolism , Receptor for Advanced Glycation End Products/metabolism , Signal Transduction/drug effects
18.
J Cell Physiol ; 234(12): 22172-22182, 2019 12.
Article in English | MEDLINE | ID: mdl-31062359

ABSTRACT

Mitochondrial dysfunction has been found to be associated with neuronal inflammation; however, no effective drug is available to attenuate neuroinflammation via sustaining mitochondrial function. In the current study, experiments were performed to understand the beneficial effects of mitochonic acid 5 (MA-5) on tumor necrosis factor-α (TNF-α)-mediated neuronal injury and mitochondrial damage. Our data illustrated that MA-5 pretreatment reduced inflammation response induced by TNF-α in CATH.a cells. Molecular investigations demonstrated that MA-5 pretreatment repressed oxidative stress, inhibited endoplasmic reticulum stress, sustained cellular energy metabolism, and blocked cell apoptosis induced by TNF-α stress. Further, we found that MA-5 treatment elevated the expression of Sirtuin 3 (Sirt3) and this effect was dependent on the activation of AMP-activated protein kinase (AMPK) pathway. Blockade of AMPK abolished the promotive action of MA-5 on Sirt3 and thus mediated mitochondrial damage and cell death. Besides, we also found that MA-5 treatment augmented Parkin-related mitophagy and increased mitophagy promoted CATH.a cells survival via improving mitochondrial function. Knockdown of Parkin abolished the beneficial action of MA-5 on mitochondrial homeostasis and CATH.a cell survival. Altogether, our results confirm that MA-5 is an effective drug to attenuate neuroinflammation via sustaining mitochondrial damage and promoting CATH.a cell survival. The protective action of MA-5 on neuronal damage is associated with Parkin-related mitophagy and the activation of AMPK-Sirt3 pathways.


Subject(s)
Indoleacetic Acids/pharmacology , Inflammation/pathology , Mitophagy , Neurons/pathology , Phenylbutyrates/pharmacology , Signal Transduction , Sirtuin 3/metabolism , Tumor Necrosis Factor-alpha/toxicity , Ubiquitin-Protein Ligases/metabolism , Adenylate Kinase/metabolism , Animals , Antioxidants/metabolism , Apoptosis/drug effects , Cell Line , Energy Metabolism/drug effects , Mice , Mitochondria/drug effects , Mitochondria/metabolism , Mitophagy/drug effects , Neurons/drug effects , Neurons/metabolism , Oxidation-Reduction , Signal Transduction/drug effects , Stress, Physiological/drug effects , Up-Regulation/drug effects
19.
Glia ; 67(1): 78-90, 2019 01.
Article in English | MEDLINE | ID: mdl-30306657

ABSTRACT

Of the seven P2X receptor subtypes, P2X4 receptor (P2X4R) is widely distributed in the central nervous system, including in neurons, astrocytes, and microglia. Accumulating evidence supports roles for P2X4R in the central nervous system, including regulating cell excitability, synaptic transmission, and neuropathic pain. However, little information is available about the distribution and function of P2X4R in the peripheral nervous system. In this study, we find that P2X4R is mainly localized in the lysosomes of Schwann cells in the peripheral nervous system. In cultured Schwann cells, TNF-a not only enhances the synthesis of P2X4R protein but also promotes P2X4R trafficking to the surface of Schwann cells. TNF-a-induced BDNF secretion in Schwann cells is P2X4R dependent. in vivo experiments reveal that expression of P2X4R in Schwann cells of injured nerves is strikingly upregulated following nerve crush injury. Moreover, overexpression of P2X4R in Schwann cells by genetic manipulation promotes motor and sensory functional recovery and accelerates nerve remyelination via BDNF release following nerve injury. Our results suggest that enhancement of P2X4R expression in Schwann cells after nerve injury may be an effective approach to facilitate the regrowth and remyelination of injured nerves.


Subject(s)
Brain-Derived Neurotrophic Factor/metabolism , Peripheral Nerve Injuries/metabolism , Receptors, Purinergic P2X4/biosynthesis , Recovery of Function/physiology , Remyelination/physiology , Schwann Cells/metabolism , Animals , Animals, Newborn , Brain-Derived Neurotrophic Factor/agonists , Cells, Cultured , Gene Expression , Male , Mice , Mice, Inbred C57BL , Peripheral Nerve Injuries/pathology , Rats , Rats, Sprague-Dawley , Receptors, Purinergic P2X4/genetics , Recovery of Function/drug effects , Remyelination/drug effects , Schwann Cells/drug effects , Schwann Cells/pathology , Tumor Necrosis Factor-alpha/toxicity
20.
Toxicol Appl Pharmacol ; 381: 114729, 2019 10 15.
Article in English | MEDLINE | ID: mdl-31445927

ABSTRACT

The PI3K/AKT signaling pathway is one of the most frequently activated signaling networks in human cancers and has become a valuable target in anticancer therapy. However, accumulating reports suggest that adverse effects such as severe liver injury and inflammation may accompany treatment with pan-PI3K and pan-AKT inhibitors. Our prior work has demonstrated that activation of the PI3K/AKT pathway has a protective role in Fas- or TNFα-induced hepatocytic cell death and liver injury. We postulated that PI3K or AKT inhibitors may exacerbate liver damage via the death factor-mediated hepatocyte apoptosis. In this study we found that several drugs targeting PI3K/AKT either clinically used or in clinical trials sensitized hepatocytes to agonistic anti-Fas antibody- or TNFα-induced apoptosis and significantly shortened the survival of mice in in vivo liver damage models. The PI3K or AKT inhibitors promoted Fas aggregation, inhibited the expression of cellular FLICE-inhibitory protein S and L (FLIPL/S), and enhanced procaspase-8 activation. Conversely, cotreatment with the AKT specific activator SC79 reversed these effects. Taken together, these findings suggest that PI3K or AKT inhibitors may render hepatocytes hypersensitive to Fas- or TNFα-induced apoptosis and liver injury.


Subject(s)
Apoptosis/drug effects , Chemical and Drug Induced Liver Injury , Hepatocytes/drug effects , Phosphoinositide-3 Kinase Inhibitors/toxicity , Protein Kinase Inhibitors/toxicity , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Aminopyridines/toxicity , Animals , Antibodies/toxicity , CASP8 and FADD-Like Apoptosis Regulating Protein/metabolism , Chemical and Drug Induced Liver Injury/metabolism , Chemical and Drug Induced Liver Injury/pathology , Hep G2 Cells , Hepatocytes/metabolism , Humans , Imidazoles/toxicity , Liver/drug effects , Liver/pathology , Male , Mice, Inbred BALB C , Mice, Inbred C57BL , Purines/toxicity , Quinazolinones/toxicity , Tumor Necrosis Factor-alpha/toxicity
SELECTION OF CITATIONS
SEARCH DETAIL