Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 53
Filter
1.
J Biol Chem ; 295(18): 6080-6091, 2020 05 01.
Article in English | MEDLINE | ID: mdl-32213596

ABSTRACT

Bacterial products such as lipopolysaccharides (or endotoxin) cause systemic inflammation, resulting in a substantial global health burden. The onset, progression, and resolution of the inflammatory response to endotoxin are usually tightly controlled to avoid chronic inflammation. Members of the NF-κB family of transcription factors are key drivers of inflammation that activate sets of genes in response to inflammatory signals. Such responses are typically short-lived and can be suppressed by proteins that act post-translationally, such as the SOCS (suppressor of cytokine signaling) family. Less is known about direct transcriptional regulation of these responses, however. Here, using a combination of in vitro approaches and in vivo animal models, we show that endotoxin treatment induced expression of the well-characterized transcriptional repressor Krüppel-like factor 3 (KLF3), which, in turn, directly repressed the expression of the NF-κB family member RELA/p65. We also observed that KLF3-deficient mice were hypersensitive to endotoxin and exhibited elevated levels of circulating Ly6C+ monocytes and macrophage-derived inflammatory cytokines. These findings reveal that KLF3 is a fundamental suppressor that operates as a feedback inhibitor of RELA/p65 and may be important in facilitating the resolution of inflammation.


Subject(s)
Kruppel-Like Transcription Factors/metabolism , Transcription Factor RelA/metabolism , Animals , Inflammation/genetics , Inflammation/immunology , Inflammation/metabolism , Kruppel-Like Transcription Factors/deficiency , Macrophages/metabolism , Mice , Transcription Factor RelA/genetics , Transcriptional Activation
2.
J Biol Chem ; 295(6): 1575-1586, 2020 02 07.
Article in English | MEDLINE | ID: mdl-31914403

ABSTRACT

Sterile alpha motif and HD domain-containing protein 1 (SAMHD1) is a deoxynucleoside triphosphohydrolase (dNTPase) with a nuclear localization signal (NLS). SAMHD1 suppresses innate immune responses to viral infection and inflammatory stimuli by inhibiting the NF-κB and type I interferon (IFN-I) pathways. However, whether the dNTPase activity and nuclear localization of SAMHD1 are required for its suppression of innate immunity remains unknown. Here, we report that the dNTPase activity, but not nuclear localization of SAMHD1, is important for its suppression of innate immune responses in differentiated monocytic cells. We generated monocytic U937 cell lines stably expressing WT SAMHD1 or mutated variants defective in dNTPase activity (HD/RN) or nuclear localization (mNLS). WT SAMHD1 in differentiated U937 cells significantly inhibited lipopolysaccharide-induced expression of tumor necrosis factor α (TNF-α) and interleukin-6 (IL-6) mRNAs, as well as IFN-α, IFN-ß, and TNF-α mRNA levels induced by Sendai virus infection. In contrast, the HD/RN mutant did not exhibit this inhibition in either U937 or THP-1 cells, indicating that the dNTPase activity of SAMHD1 is important for suppressing NF-κB activation. Of note, in lipopolysaccharide-treated or Sendai virus-infected U937 or THP-1 cells, the mNLS variant reduced TNF-α or IFN-ß mRNA expression to a similar extent as did WT SAMHD1, suggesting that SAMHD1-mediated inhibition of innate immune responses is independent of SAMHD1's nuclear localization. Moreover, WT and mutant SAMHD1 similarly interacted with key proteins in NF-κB and IFN-I pathways in cells. This study further defines the role and mechanisms of SAMHD1 in suppressing innate immunity.


Subject(s)
Immunity, Innate , Monocytes/immunology , SAM Domain and HD Domain-Containing Protein 1/immunology , Cell Nucleus/immunology , Humans , Respirovirus Infections/immunology , SAM Domain and HD Domain-Containing Protein 1/analysis , Sendai virus/immunology , THP-1 Cells , U937 Cells
3.
J Biol Chem ; 294(12): 4368-4380, 2019 03 22.
Article in English | MEDLINE | ID: mdl-30670595

ABSTRACT

Angiotensin-converting enzyme (ACE) can hydrolyze many peptides and plays a central role in controlling blood pressure. Moreover, ACE overexpression in monocytes and macrophages increases resistance of mice to tumor growth. ACE is composed of two independent catalytic domains. Here, to investigate the specific role of each domain in tumor resistance, we overexpressed either WT ACE (Tg-ACE mice) or ACE lacking N- or C-domain catalytic activity (Tg-NKO and Tg-CKO mice) in the myeloid cells of mice. Tg-ACE and Tg-NKO mice exhibited strongly suppressed growth of B16-F10 melanoma because of increased ACE expression in macrophages, whereas Tg-CKO mice resisted melanoma no better than WT animals. The effect of ACE overexpression reverted to that of the WT enzyme with an ACE inhibitor but not with an angiotensin II type 1 (AT1) receptor antagonist. ACE C-domain overexpression in macrophages drove them toward a pronounced M1 phenotype upon tumor stimulation, with increased activation of NF-κB and signal transducer and activator of transcription 1 (STAT1) and decreased STAT3 and STAT6 activation. Tumor necrosis factor α (TNFα) is important for M1 activation, and TNFα blockade reverted Tg-NKO macrophages to a WT phenotype. Increased ACE C-domain expression increased the levels of reactive oxygen species (ROS) and of the transcription factor C/EBPß in macrophages, important stimuli for TNFα expression, and decreased expression of several M2 markers, including interleukin-4Rα. Natural ACE C-domain-specific substrates are not well-described, and we propose that the peptide(s) responsible for the striking ACE-mediated enhancement of myeloid function are substrates/products of the ACE C-domain.


Subject(s)
Cell Polarity , Macrophages/cytology , Melanoma, Experimental/pathology , Peptidyl-Dipeptidase A/metabolism , Animals , Catalysis , Cell Line, Tumor , Cell Survival , Gene Expression Regulation, Neoplastic , Macrophages/immunology , Melanoma, Experimental/enzymology , Melanoma, Experimental/genetics , Melanoma, Experimental/immunology , Mice , Mice, Transgenic , NF-kappa B/metabolism , Peptidyl-Dipeptidase A/chemistry , STAT1 Transcription Factor/metabolism , Tumor Necrosis Factor-alpha/biosynthesis
4.
J Biol Chem ; 294(21): 8480-8489, 2019 05 24.
Article in English | MEDLINE | ID: mdl-30940649

ABSTRACT

Among the repertoire of immunoregulatory proteins encoded by myxoma virus, M013 is a viral homologue of the viral pyrin domain-only protein (vPOP) family. In myeloid cells, M013 protein has been shown to inhibit both the inflammasome and NF-κB signaling pathways by direct binding to ASC1 and NF-κB1, respectively. In this study, a three-dimensional homology model of the M013 pyrin domain (PYD) was built based on similarities to known PYD structures. A distinctive feature of the deduced surface electrostatic map of the M013 PYD is the presence of a negatively region consisting of numerous aspartate and glutamate residues in close proximity. Single-site mutations of aspartate and glutamate residues reveal their role in interactions with ASC-1. The biological significance of charge complementarity in the M013-ASC-1 interaction was further confirmed by functional assays of caspase-1 activation and subsequent secretion of cytokines. M013 also has a unique 33-residue C-terminal tail that follows the N-terminal PYD, and it is enriched in positively charged residues. Deletion of the tail of M013 significantly inhibited the interactions between M013 and NF-κB1, thus compromising the ability of the viral protein to suppress the secretion of pro-inflammatory cytokines. These results demonstrate that vPOP M013 exploits distinct structural motifs to regulate both the inflammasome and NF-κB pathways.


Subject(s)
Myxoma virus , NF-kappa B/immunology , Signal Transduction/immunology , Viral Proteins , Amino Acid Motifs , Amino Acid Substitution , Caspase 1/genetics , Caspase 1/immunology , HeLa Cells , Humans , Inflammasomes/genetics , Mutagenesis, Site-Directed , Mutation, Missense , Myxoma virus/chemistry , Myxoma virus/genetics , Myxoma virus/immunology , NF-kappa B/genetics , Protein Domains , Signal Transduction/genetics , THP-1 Cells , Viral Proteins/chemistry , Viral Proteins/genetics , Viral Proteins/immunology
5.
J Biol Chem ; 294(38): 14135-14148, 2019 09 20.
Article in English | MEDLINE | ID: mdl-31366726

ABSTRACT

The tumor suppressor CYLD is a deubiquitinating enzyme that suppresses polyubiquitin-dependent signaling pathways, including the proinflammatory and cell growth-promoting NF-κB pathway. Missense mutations in the CYLD gene are present in individuals with syndromes such as multiple familial trichoepithelioma (MFT), but the pathogenic roles of these mutations remain unclear. Recent studies have shown that CYLD interacts with a RING finger domain protein, mind bomb homologue 2 (MIB2), in the regulation of NOTCH signaling. However, whether MIB2 is an E3 ubiquitin ligase that acts on CYLD is unknown. Here, using the cell-free-based AlphaScreen and pulldown assays to detect protein-protein interactions, along with immunofluorescence assays and murine Mib2 knockout cells and animals, we demonstrate that MIB2 promotes proteasomal degradation of CYLD and enhances NF-κB signaling. Of note, arthritic inflammation was suppressed in Mib2-deficient mice. We further observed that the ankyrin repeat in MIB2 interacts with the third CAP domain in CYLD and that MIB2 catalyzes Lys-48-linked polyubiquitination of CYLD at Lys-338 and Lys-530. MIB2-dependent CYLD degradation activated NF-κB signaling via tumor necrosis factor alpha (TNFα) stimulation and the linear ubiquitination assembly complex (LUBAC). Mib2-knockout mice had reduced serum interleukin-6 (IL-6) and exhibited suppressed inflammatory responses in the K/BxN serum-transfer arthritis model. Interestingly, MIB2 significantly enhanced the degradation of a CYLDP904L variant identified in an individual with MFT, although the molecular pathogenesis of the disease was not clarified here. Together, these results suggest that MIB2 enhances NF-κB signaling in inflammation by promoting the ubiquitin-dependent degradation of CYLD.


Subject(s)
Deubiquitinating Enzyme CYLD/metabolism , Ubiquitin-Protein Ligases/metabolism , Animals , Cysteine Endopeptidases/metabolism , Deubiquitinating Enzymes/metabolism , Female , HEK293 Cells , HeLa Cells , Humans , Inflammation/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , NF-kappa B/metabolism , Polyubiquitin/metabolism , Proteasome Endopeptidase Complex/metabolism , Signal Transduction/physiology , Transcription Factor RelA , Tumor Necrosis Factor-alpha/metabolism , Ubiquitin/metabolism , Ubiquitination
6.
J Biol Chem ; 293(7): 2452-2465, 2018 02 16.
Article in English | MEDLINE | ID: mdl-29279332

ABSTRACT

Nuclear factor-κB (NF-κB) is a family of transcription factors that play a key role in cell survival and proliferation in many hematological malignancies, including multiple myeloma (MM). Bortezomib, a proteasome inhibitor used in the management of MM, can inhibit both canonical and noncanonical activation of NF-κB in MM cells. However, we previously reported that a significant fraction of freshly isolated MM cells harbor bortezomib-resistant NF-κB activity. Here, we report that hyaluronan and proteoglycan link protein 1 (HAPLN1) is produced in bone marrow stromal cells from MM patients, is detected in patients' bone marrow plasma, and can activate an atypical bortezomib-resistant NF-κB pathway in MM cells. We found that this pathway involves bortezomib-resistant degradation of the inhibitor of NF-κB (IκBα), despite efficient bortezomib-mediated inhibition of proteasome activity. Moreover, HAPLN1 can also confer bortezomib-resistant survival of MM cells. We propose that HAPLN1 is a novel pathogenic factor in MM that induces an atypical NF-κB activation and thereby promotes bortezomib resistance in MM cells.


Subject(s)
Antineoplastic Agents/pharmacology , Bortezomib/pharmacology , Extracellular Matrix Proteins/metabolism , Multiple Myeloma/metabolism , NF-kappa B/metabolism , Proteoglycans/metabolism , Bone Marrow Cells/drug effects , Bone Marrow Cells/metabolism , Drug Resistance, Neoplasm , Extracellular Matrix Proteins/genetics , Humans , I-kappa B Proteins/genetics , I-kappa B Proteins/metabolism , Multiple Myeloma/drug therapy , Multiple Myeloma/genetics , NF-kappa B/genetics , Proteasome Endopeptidase Complex/genetics , Proteasome Endopeptidase Complex/metabolism , Proteoglycans/genetics , Proteolysis
7.
J Biol Chem ; 293(8): 2701-2710, 2018 02 23.
Article in English | MEDLINE | ID: mdl-29301940

ABSTRACT

Mammalian Nod-like receptor (NLR) proteins contribute to the regulation and induction of innate and adaptive immunity in mammals, although the function of about half of the currently identified NLR proteins remains poorly characterized. Here we analyzed the function of the primate-specific NLRP11 gene product. We show that NLRP11 is highly expressed in immune cells, including myeloid cells, B cells, and some B cell lymphoma lines. Overexpression of NLRP11 in human cells did not trigger key innate immune signaling pathways, including NF-κB and type I interferon responses. NLRP11 harbors a pyrin domain, which is responsible for inflammasome formation in related NLR proteins. However, NLRP11 did not interact with the inflammasome adaptor protein ASC, and it did not trigger caspase-1 activation. By contrast, expression of NLRP11 specifically repressed NF-κB and type I interferon responses, two key innate immune pathways involved in inflammation. This effect was independent of the pyrin domain and ATPase activity of NLRP11. siRNA-mediated knockdown of NLRP11 in human myeloid THP1 cells validated these findings and revealed enhanced lipopolysaccharide and Sendai virus-induced cytokine and interferon responses, respectively, in cells with reduced NLRP11 expression. In summary, our work identifies a novel role of NLRP11 in the regulation of inflammatory responses in human cells.


Subject(s)
B-Lymphocytes/metabolism , Down-Regulation , Gene Expression Regulation , Immunity, Innate , Intracellular Signaling Peptides and Proteins/metabolism , Myeloid Cells/metabolism , NLR Proteins/metabolism , Amino Acid Substitution , B-Lymphocytes/cytology , B-Lymphocytes/drug effects , B-Lymphocytes/immunology , Cell Line, Transformed , Cell Line, Tumor , Down-Regulation/drug effects , Female , Gene Expression Regulation/drug effects , Genes, Reporter/drug effects , Humans , Immunity, Innate/drug effects , Interferon Type I/agonists , Interferon Type I/antagonists & inhibitors , Interferon Type I/metabolism , Intracellular Signaling Peptides and Proteins/antagonists & inhibitors , Intracellular Signaling Peptides and Proteins/chemistry , Intracellular Signaling Peptides and Proteins/genetics , Lipopolysaccharides/toxicity , Male , Mutation , Myeloid Cells/cytology , Myeloid Cells/drug effects , Myeloid Cells/immunology , NF-kappa B/agonists , NF-kappa B/antagonists & inhibitors , NF-kappa B/metabolism , NLR Proteins/genetics , Neoplasm Proteins/antagonists & inhibitors , Neoplasm Proteins/chemistry , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Organ Specificity , Peptide Fragments/antagonists & inhibitors , Peptide Fragments/chemistry , Peptide Fragments/genetics , Peptide Fragments/metabolism , RNA Interference , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/metabolism
8.
J Biol Chem ; 293(41): 15790-15800, 2018 10 12.
Article in English | MEDLINE | ID: mdl-30093408

ABSTRACT

Interleukin (IL)-1ß plays a critical role in IL-6ß- and transforming growth factor ß (TGFß)-initiated Th17 differentiation and induction of Th17-mediated autoimmunity. However, the means by which IL-1 regulates various aspects of Th17 development remain poorly understood. We recently reported that IL-1ß enhances STAT3 phosphorylation via NF-κB-mediated repression of SOCS3 to facilitate Il17 transcription and Th17 differentiation, identifying an effect of IL-1 signaling on proximal events of STAT3 signaling. Here, we show that IL-1ß promotes STAT3 binding to key cis-elements that control IL-17 expression. Additionally, we demonstrate that the IL-1-induced NF-κB factor RelA directly regulates the Il17a/f loci in cooperation with STAT3. Our findings reveal that IL-1 impacts both proximal signaling events and downstream interactions between transcription factors and cis-regulatory elements to promote Il17a/f transcription and Th17 differentiation.


Subject(s)
Interleukin-17/metabolism , Receptors, Interleukin-1 Type II/metabolism , STAT3 Transcription Factor/metabolism , Transcription Factor RelA/metabolism , Animals , DNA/chemistry , DNA/genetics , Interleukin-17/genetics , Mice, Inbred C57BL , Regulatory Sequences, Nucleic Acid/genetics , STAT3 Transcription Factor/genetics , Signal Transduction/physiology , Th17 Cells , Transcription Factor RelA/genetics , Transcriptional Activation
9.
J Biol Chem ; 293(38): 14839-14849, 2018 09 21.
Article in English | MEDLINE | ID: mdl-30093411

ABSTRACT

Initiation of expression of fibroblast growth factor receptor 1 (FGFR1) concurrent with loss of FGFR2 expression is a well-documented event in the progression of prostate cancer (PCa). Although it is known that some FGFR isoforms confer advantages in cell proliferation and survival, the mechanism by which the subversion of different FGFR isoforms contributes to PCa progression is incompletely understood. Here, we report that fibroblast growth factor (FGF) promotes NF-κB signaling in PCa cells and that this increase is associated with FGFR1 expression. Disruption of FGFR1 kinase activity abrogated both FGF activity and NF-κB signaling in PCa cells. Of note, the three common signaling pathways downstream of FGFR1 kinase, extracellular signal-regulated kinase 1/2 (ERK1/2), phosphoinositide 3-kinase (PI3K/AKT), and phosphoinositide phospholipase Cγ (PLCγ), were not required for FGF-mediated NF-κB signaling. Instead, transforming growth factor ß-activating kinase 1 (TAK1), a central regulator of the NF-κB pathway, was required for FGFR1 to stimulate NF-κB signaling. Moreover, we found that FGFR1 promotes NF-κB signaling in PCa cells by reducing TAK1 degradation and thereby supporting sustained NF-κB activation. Consistently, Fgfr1 ablation in the transgenic adenocarcinoma of the mouse prostate (TRAMP) model reduced inflammation in the tumor microenvironment. In contrast, activation of the FGFR1 kinase in the juxtaposition of chemical-induced dimerization (CID) and kinase 1 (JOCK1) mouse model increased inflammation. As inflammation plays an important role in PCa initiation and progression, these findings suggest that ectopically expressed FGFR1 promotes PCa progression, at least in part, by increasing inflammation in the tumor microenvironment.


Subject(s)
Inflammation/metabolism , NF-kappa B/metabolism , Prostatic Neoplasms/metabolism , Receptor, Fibroblast Growth Factor, Type 1/metabolism , Signal Transduction , Animals , Cell Line, Tumor , HEK293 Cells , Humans , MAP Kinase Kinase Kinases/metabolism , MAP Kinase Signaling System , Male , Mice , Phosphatidylinositol 3-Kinases/metabolism , Phospholipase C gamma/metabolism , Phosphorylation , Prostatic Neoplasms/pathology , Proto-Oncogene Proteins c-akt/metabolism , Tumor Microenvironment
10.
J Biol Chem ; 292(20): 8136-8148, 2017 05 19.
Article in English | MEDLINE | ID: mdl-28270510

ABSTRACT

The selective estrogen receptor (ER) modulator tamoxifen inhibits ER signaling in breast cancer cells, and it is used for the treatment of ER-positive breast cancer. However, this type of cancer often acquires resistance to tamoxifen, and a better understanding of the molecular mechanisms underlying tamoxifen resistance is required. In this study, we established tamoxifen-resistant (TAM-R) breast cancer cells by long-term tamoxifen treatment of ER-positive breast cancer MCF7 cells. In TAM-R cells, expression of not only ERα, a major form of ER in breast cancer, but also its transcriptional partner forkhead box protein A1 (FOXA1) was found to be reduced. In contrast, activation of the transcription factor nuclear factor-κB (NF-κB) and expression of its target IL6 were increased in these cells. Stable expression of FOXA1, but not ERα, reduced the expression of IL6 in the FOXA1- and ERα-negative breast cancer MDA-MB-231 cells and TAM-R cells, without affecting the activation of the NF-κB signaling pathways. Conversely, FOXA1 knockdown induced IL6 expression in MCF7 cells. Chromatin immunoprecipitation assays revealed that FOXA1 bound to the promoter region of IL6 and repressed recruitment of the NF-κB complex to this region. TAM-R cells were found to have high mammosphere-forming activity, characteristics of cancer stem cells, and this activity was suppressed by NF-κB and IL6 signaling inhibitors. Taken together, these results suggest that FOXA1 suppresses expression of IL6 through inhibition of NF-κB recruitment to the IL6 promoter in an ERα-independent manner and that reduction in FOXA1 expression induces IL6 expression and contributes to cancer stem cell-like properties in TAM-R cells.


Subject(s)
Breast Neoplasms/metabolism , Drug Resistance, Neoplasm , Hepatocyte Nuclear Factor 3-alpha/metabolism , Interleukin-6/biosynthesis , Neoplasm Proteins/metabolism , Neoplastic Stem Cells/metabolism , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Estrogen Receptor alpha/genetics , Estrogen Receptor alpha/metabolism , Female , Gene Expression Regulation, Neoplastic/drug effects , Gene Knockdown Techniques , Hepatocyte Nuclear Factor 3-alpha/genetics , Humans , Interleukin-6/genetics , MCF-7 Cells , NF-kappa B/genetics , NF-kappa B/metabolism , Neoplasm Proteins/genetics , Response Elements , Tamoxifen/pharmacology
11.
J Biol Chem ; 291(38): 19900-12, 2016 09 16.
Article in English | MEDLINE | ID: mdl-27481940

ABSTRACT

Systemic lupus erythematosus (SLE) is a chronic inflammatory autoimmune disease affecting multiple organs. Glucocorticoids (GCs), the potent anti-inflammatory drugs, remain as a cornerstone in the treatment for SLE; nevertheless, their clinical efficacy is compromised by the side effects of long term treatment and resistance. To improve the therapeutic efficacy of GCs in SLE, it is important to further decipher the molecular mechanisms of how GCs exert their anti-inflammatory effects. In this investigation, FOXO3a was identified as a molecule that was down-regulated in the course of SLE. Of interest, GC treatment was found to rescue FOXO3a expression both in SLE mice and in SLE patients. Gain- and loss-of-function studies demonstrated that FOXO3a played a crucial role in GC treatment of SLE via inhibiting inflammatory responses. Further studies showed that the up-regulation of FOXO3a by GCs relied on the suppression of pI3K/AKT-mediated FOXO3a phosphorylation and the arrest of FOXO3a in the nucleus. Finally, our data revealed that FOXO3a was critical for GC-mediated inhibition of NF-κB activity, which might involve its interaction with NF-κB p65 protein. Collectively, these data indicated that FOXO3a played an important role in GC treatment of SLE by suppressing pro-inflammatory response, and targeting FOXO3a might provide a novel therapeutic strategy against SLE.


Subject(s)
Forkhead Box Protein O3/immunology , Gene Expression Regulation/drug effects , Glucocorticoids/pharmacology , Lupus Erythematosus, Systemic/drug therapy , Signal Transduction/drug effects , Adult , Animals , Autoantigens/immunology , Cell Nucleus/immunology , Female , Gene Expression Regulation/immunology , Humans , Lupus Erythematosus, Systemic/immunology , Lupus Erythematosus, Systemic/pathology , Male , Mice , Mice, Inbred BALB C , Phosphatidylinositol 3-Kinases/immunology , Phosphorylation/drug effects , Phosphorylation/immunology , Proto-Oncogene Proteins c-akt/immunology , Signal Transduction/immunology , Transcription Factor RelA/immunology
12.
J Biol Chem ; 291(46): 24017-24028, 2016 Nov 11.
Article in English | MEDLINE | ID: mdl-27650497

ABSTRACT

Activation of the blood vessel endothelium is a critical step during inflammation. Endothelial cells stimulated by pro-inflammatory cytokines play an essential part in the adhesion and extravasation of circulating leukocytes into inflamed tissues. The endothelial egfl7 gene (VE-statin) represses endothelial cell activation in tumors, and prior observations suggested that it could also participate in the regulation of endothelial cell activation during inflammation. We show here that Egfl7 expression is strongly repressed in mouse lung endothelial cells during LPS- and TNFα-induced inflammation in vivo LPS have a limited effect on Egfl7 expression by endothelial cells in vitro, whereas the pro-inflammatory cytokine TNFα strongly represses Egfl7 expression in endothelial cells. TNFα regulates the egfl7 gene promoter through regions located between -7585 and -5550 bp ahead of the main transcription start site and via an NF-κB-dependent mechanism. Conversely, Egfl7 regulates the response of endothelial cells to TNFα by restraining the induced expression of intercellular adhesion molecule-1 (ICAM-1), vascular cell adhesion molecule-1 (VCAM-1), and E-selectin, resulting in a decreased adhesion of leukocytes onto endothelial cells stimulated by TNFα. Egfl7 regulates the expression of these adhesion molecules through the NF-κB and MEK/Erk pathways, in particular by preventing the proteasome-mediated degradation of IkBα both in non-activated endothelial cells and during activation. Egfl7 is thus an endogenous and constitutive repressor of blood vessel endothelial cell activation in normal and inflammatory conditions and participates in a loop of regulation of activation of these cells by pro-inflammatory cytokines.


Subject(s)
Endothelial Growth Factors/biosynthesis , Gene Expression Regulation , MAP Kinase Signaling System , Response Elements , Animals , Calcium-Binding Proteins , EGF Family of Proteins , Endothelial Growth Factors/genetics , Human Umbilical Vein Endothelial Cells , Humans , Inflammation/genetics , Inflammation/metabolism , Inflammation/pathology , Intercellular Adhesion Molecule-1/genetics , Intercellular Adhesion Molecule-1/metabolism , Jurkat Cells , Mice , NF-kappa B/genetics , NF-kappa B/metabolism , Proteasome Endopeptidase Complex/genetics , Proteasome Endopeptidase Complex/metabolism , Proteolysis , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/metabolism , Vascular Cell Adhesion Molecule-1/genetics , Vascular Cell Adhesion Molecule-1/metabolism
13.
J Biol Chem ; 291(21): 11105-13, 2016 May 20.
Article in English | MEDLINE | ID: mdl-27033704

ABSTRACT

Water accumulation in the interstitium (edema) and the peritoneum (ascites) of nephrotic patients is classically thought to stem from the prevailing low plasma albumin concentration and the decreased transcapillary oncotic pressure gradient. However, several clinical and experimental observations suggest that it might also stem from changes in capillary permeability. We addressed this hypothesis by studying the peritoneum permeability of rats with puromycin aminonucleoside-induced nephrotic syndrome. The peritoneum of puromycin aminonucleoside rats displayed an increase in the water filtration coefficient of paracellular and transcellular pathways, and a decrease in the reflection coefficient to proteins. It also displayed oxidative stress and subsequent activation of NF-κB. Scavenging of reactive oxygen species and inhibition of NF-κB prevented the changes in the water permeability and reflection coefficient to proteins and reduced the volume of ascites by over 50%. Changes in water permeability were associated with the overexpression of the water channel aquaporin 1, which was prevented by reactive oxygen species scavenging and inhibition of NF-κB. In conclusion, nephrotic syndrome is associated with an increased filtration coefficient of the peritoneum and a decreased reflection coefficient to proteins. These changes, which account for over half of ascite volume, are triggered by oxidative stress and subsequent activation of NF-κB.


Subject(s)
Ascites , NF-kappa B/metabolism , Nephrotic Syndrome , Oxidative Stress/drug effects , Peritoneum , Puromycin Aminonucleoside/adverse effects , Animals , Aquaporin 1/metabolism , Ascites/chemically induced , Ascites/metabolism , Ascites/pathology , Humans , Male , Nephrotic Syndrome/chemically induced , Nephrotic Syndrome/metabolism , Nephrotic Syndrome/pathology , Peritoneum/metabolism , Peritoneum/pathology , Puromycin Aminonucleoside/pharmacology , Rats , Rats, Sprague-Dawley
14.
J Biol Chem ; 291(24): 12586-12600, 2016 Jun 10.
Article in English | MEDLINE | ID: mdl-27053111

ABSTRACT

Phospholipase Cϵ (PLCϵ), an effector of Ras and Rap small GTPases, plays a crucial role in inflammation by augmenting proinflammatory cytokine expression. This proinflammatory function of PLCϵ is implicated in its facilitative role in tumor promotion and progression during skin and colorectal carcinogenesis, although their direct link remains to be established. Moreover, the molecular mechanism underlying these functions of PLCϵ remains unknown except that PKD works downstream of PLCϵ. Here we show by employing the colitis-induced colorectal carcinogenesis model, where Apc(Min) (/+) mice are administered with dextran sulfate sodium, that PLCϵ knock-out alleviates the colitis and suppresses the following tumorigenesis concomitant with marked attenuation of proinflammatory cytokine expression. In human colon epithelial Caco2 cells, TNF-α induces sustained expression of proinflammatory molecules and sustained activation of nuclear factor-κB (NF-κB) and PKD, the late phases of which are suppressed by not only siRNA-mediated PLCϵ knockdown but also treatment with a lysophosphatidic acid (LPA) receptor antagonist. Also, LPA stimulation induces these events in an early time course, suggesting that LPA mediates TNF-α signaling in an autocrine manner. Moreover, PLCϵ knockdown results in inhibition of phosphorylation of IκB by ribosomal S6 kinase (RSK) but not by IκB kinases. Subcellular fractionation suggests that enhanced phosphorylation of a scaffolding protein, PEA15 (phosphoprotein enriched in astrocytes 15), downstream of the PLCϵ-PKD axis causes sustained cytoplasmic localization of phosphorylated RSK, thereby facilitating IκB phosphorylation in the cytoplasm. These results suggest the crucial role of the TNF-α-LPA-LPA receptor-PLCϵ-PKD-PEA15-RSK-IκB-NF-κB pathway in facilitating inflammation and inflammation-associated carcinogenesis in the colon.


Subject(s)
Epithelial Cells/metabolism , NF-kappa B/metabolism , Phosphoinositide Phospholipase C/metabolism , Ribosomal Protein S6 Kinases/metabolism , Signal Transduction , Adenomatous Polyposis Coli Protein/genetics , Adenomatous Polyposis Coli Protein/metabolism , Animals , Apoptosis Regulatory Proteins , Caco-2 Cells , Colitis/genetics , Colitis/metabolism , Colon/metabolism , Colon/pathology , Colorectal Neoplasms/genetics , Colorectal Neoplasms/metabolism , Cytoplasm/enzymology , Humans , I-kappa B Proteins/metabolism , Immunoblotting , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Lysophospholipids/pharmacology , Mice, Inbred C57BL , Mice, Knockout , NF-KappaB Inhibitor alpha , Phosphoinositide Phospholipase C/genetics , Phosphoproteins/genetics , Phosphoproteins/metabolism , Phosphorylation/drug effects , Protein Kinase C/metabolism , RNA Interference , Receptors, Lysophosphatidic Acid/genetics , Receptors, Lysophosphatidic Acid/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Ribosomal Protein S6 Kinases/genetics , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/metabolism , Tumor Necrosis Factor-alpha/pharmacology
15.
J Biol Chem ; 291(30): 15614-27, 2016 07 22.
Article in English | MEDLINE | ID: mdl-27231343

ABSTRACT

Cryptococcus neoformans (Cn) is a common facultative intracellular pathogen that can cause life-threatening fungal meningitis in immunocompromised individuals. Shortly after infection, Cn is detectable as both extra- and intracellular yeast particles, with Cn being capable of establishing long-lasting latent infections within host macrophages. Although recent studies have shown that shed capsular polysaccharides and intact extracellular Cn can compromise macrophage function through modulation of NF-κB signaling, it is currently unclear whether intracellular Cn also affects NF-κB signaling. Utilizing live cell imaging and computational modeling, we find that extra- and intracellular Cn support distinct modes of NF-κB signaling in cultured murine macrophages. Specifically, in RAW 264.7 murine macrophages treated with extracellular glucuronoxylomannan (GXM), the major Cn capsular polysaccharide, LPS-induced nuclear translocation of p65 is inhibited, whereas in cells with intracellular Cn, LPS-induced nuclear translocation of p65 is both amplified and sustained. Mathematical simulations and quantification of nascent protein expression indicate that this is a possible consequence of Cn-induced "translational interference," impeding IκBα resynthesis. We also show that long term Cn infection induces stable nuclear localization of p65 and IκBα proteins in the absence of additional pro-inflammatory stimuli. In this case, nuclear localization of p65 is not accompanied by TNFα or inducible NOS (iNOS) expression. These results demonstrate that capsular polysaccharides and intact intracellular yeast manipulate NF-κB via multiple distinct mechanisms and provide new insights into how Cn might modulate cellular signaling at different stages of an infection.


Subject(s)
Cell Nucleus/metabolism , Cryptococcosis/metabolism , Cryptococcus neoformans/metabolism , Macrophages/metabolism , Models, Biological , Transcription Factor RelA/metabolism , Active Transport, Cell Nucleus , Animals , Cell Nucleus/pathology , Cryptococcosis/pathology , Lipopolysaccharides/toxicity , Macrophages/microbiology , Macrophages/pathology , Mice , NF-KappaB Inhibitor alpha/metabolism , Nitric Oxide Synthase Type II/biosynthesis , RAW 264.7 Cells , Tumor Necrosis Factor-alpha/metabolism
16.
J Biol Chem ; 291(52): 26686-26697, 2016 Dec 23.
Article in English | MEDLINE | ID: mdl-27875309

ABSTRACT

Transcription factor tonicity-responsive enhancer-binding protein (TonEBP/NFAT5) is critical for osmo-adaptation and extracellular matrix homeostasis of nucleus pulposus (NP) cells in their hypertonic tissue niche. Recent studies implicate TonEBP signaling in inflammatory disease and rheumatoid arthritis pathogenesis. However, broader functions of TonEBP in the disc remain unknown. RNA sequencing was performed on NP cells with TonEBP knockdown under hypertonic conditions. 1140 TonEBP-dependent genes were identified and categorized using Ingenuity Pathway Analysis. Bioinformatic analysis showed enrichment of matrix homeostasis and cytokine/chemokine signaling pathways. C-C motif chemokine ligand 2 (CCL2), interleukin 6 (IL6), tumor necrosis factor (TNF), and nitric oxide synthase 2 (NOS2) were studied further. Knockdown experiments showed that TonEBP was necessary to maintain expression levels of these genes. Gain- and loss-of-function experiments and site-directed mutagenesis demonstrated that TonEBP binding to a specific site in the CCL2 promoter is required for hypertonic inducibility. Despite inhibition by dominant-negative TonEBP, IL6 and NOS2 promoters were not hypertonicity-inducible. Whole-disc response to hypertonicity was studied in an ex vivo organ culture model, using wild-type and haploinsufficient TonEBP mice. Pro-inflammatory targets were induced by hypertonicity in discs from wild-type but not TonEBP-haploinsufficient mice. Mechanistically, NF-κB activity increased with hypertonicity and was necessary for hypertonic induction of target genes IL6, TNF, and NOS2 but not CCL2 Although TonEBP maintains transcription of genes traditionally considered pro-inflammatory, it is important to note that some of these genes also serve anabolic and pro-survival roles. Therefore, in NP cells, this phenomenon may reflect a physiological adaptation to diurnal osmotic loading of the intervertebral disc.


Subject(s)
Gene Expression Regulation , High-Throughput Nucleotide Sequencing/methods , Homeostasis , Inflammation Mediators/metabolism , NFATC Transcription Factors/metabolism , Nucleus Pulposus/metabolism , Osmosis/physiology , Animals , Intervertebral Disc , Mice , Mice, Knockout , Mutagenesis, Site-Directed , NF-kappa B/genetics , NF-kappa B/metabolism , NFATC Transcription Factors/genetics , Organ Culture Techniques , Promoter Regions, Genetic/genetics , Rats , Signal Transduction
17.
J Biol Chem ; 291(50): 26098-26108, 2016 Dec 09.
Article in English | MEDLINE | ID: mdl-27803164

ABSTRACT

Recent work has demonstrated pro-oncogenic functions of the transcription factor CCAAT box/enhancer-binding protein ß (C/EBPß) in various tumors, implicating C/EBPß as an interesting target for the development of small-molecule inhibitors. We have previously discovered that the sesquiterpene lactone helenalin acetate, a natural compound known to inhibit NF-κB, is a potent C/EBPß inhibitor. We have now examined the inhibitory mechanism of helenalin acetate in more detail. We demonstrate that helenalin acetate is a significantly more potent inhibitor of C/EBPß than of NF-κB. Our work shows that helenalin acetate inhibits C/EBPß by binding to the N-terminal part of C/EBPß, thereby disrupting the cooperation of C/EBPß with the co-activator p300. C/EBPß is expressed in several isoforms from alternative translational start codons. We have previously demonstrated that helenalin acetate selectively inhibits only the full-length (liver-enriched activating protein* (LAP*)) isoform but not the slightly shorter (LAP) isoform. Consistent with this, helenalin acetate binds to the LAP* but not to the LAP isoform, explaining why its inhibitory activity is selective for LAP*. Although helenalin acetate contains reactive groups that are able to interact covalently with cysteine residues, as exemplified by its effect on NF-κB, the inhibition of C/EBPß by helenalin acetate is not due to irreversible reaction with cysteine residues of C/EBPß. In summary, helenalin acetate is the first highly active small-molecule C/EBPß inhibitor that inhibits C/EBPß by a direct binding mechanism. Its selectivity for the LAP* isoform also makes helenalin acetate an interesting tool to dissect the functions of the LAP* and LAP isoforms.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Antineoplastic Agents, Phytogenic/pharmacology , CCAAT-Enhancer-Binding Protein-beta/antagonists & inhibitors , Sesquiterpenes/pharmacology , p300-CBP Transcription Factors/antagonists & inhibitors , 3T3-L1 Cells , Animals , Anti-Inflammatory Agents, Non-Steroidal/pharmacokinetics , Antineoplastic Agents, Phytogenic/pharmacokinetics , CCAAT-Enhancer-Binding Protein-beta/genetics , CCAAT-Enhancer-Binding Protein-beta/metabolism , Mice , NF-kappa B/genetics , NF-kappa B/metabolism , Protein Isoforms/antagonists & inhibitors , Protein Isoforms/genetics , Protein Isoforms/metabolism , Sesquiterpenes/pharmacokinetics , Sesquiterpenes, Guaiane , p300-CBP Transcription Factors/genetics , p300-CBP Transcription Factors/metabolism
18.
J Biol Chem ; 291(47): 24517-24527, 2016 Nov 18.
Article in English | MEDLINE | ID: mdl-27733681

ABSTRACT

Vitamin B6 includes six water-soluble vitamers: pyridoxal (PL), pyridoxamine (PM), pyridoxine (PN), and their phosphorylated forms. Pyridoxal 5'-phosphate (PLP) is an important cofactor for many metabolic enzymes. Several lines of evidence demonstrate that blood levels of PLP are significantly lower in patients with inflammation than in control subjects and that vitamin B6 has anti-inflammatory effects, with therapeutic potential for a variety of inflammatory diseases. Although one of our group previously demonstrated that PL inhibits the NF-κB pathway, the molecular mechanism by which vitamin B6 suppresses inflammation is not well understood. Here, we showed that both PL and PLP suppressed the expression of cytokine genes in macrophages by inhibiting Toll-like receptor (TLR)-mediated TAK1 phosphorylation and the subsequent NF-κB and JNK activation. Furthermore, PL and PLP abolished NLRP3-dependent caspase-1 processing and the subsequent secretion of mature IL-1ß and IL-18 in LPS-primed macrophages. In contrast, PM and PN had little effect on IL-1ß production. PLP, but not PL, markedly reduced the production of mitochondrial reactive oxygen species (ROS) in peritoneal macrophages. Importantly, PL and PLP reduced IL-1ß production induced by LPS and ATP, or by LPS alone, in mice. Moreover, PL and PLP protected mice from lethal endotoxic shock. Collectively, these findings reveal novel anti-inflammatory activities for vitamin B6 and suggest its potential for preventing inflammatory diseases driven by the NLRP3 inflammasome.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Inflammasomes/metabolism , Interleukin-1beta/biosynthesis , Macrophages/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Vitamin B 6/pharmacology , Animals , Interleukin-18/biosynthesis , Lipopolysaccharides/toxicity , MAP Kinase Kinase Kinases/metabolism , Mice , Mice, Inbred ICR , NF-kappa B/metabolism , Phosphorylation/drug effects , Shock, Septic/chemically induced , Shock, Septic/metabolism , Shock, Septic/prevention & control
19.
J Biol Chem ; 291(9): 4754-62, 2016 Feb 26.
Article in English | MEDLINE | ID: mdl-26694617

ABSTRACT

Here, we identify ATP1B3 and fibrillin-1 as novel BST-2-binding proteins. ATP1B3 depletion in HeLa cells (BST-2-positive cells), but not 293T cells (BST-2-negative cells), induced the restriction of HIV-1 production in a BST-2-dependent manner. In contrast, fibrillin-1 knockdown reduced HIV-1 production in 293T and HeLa cells in a BST-2-independent manner. Moreover, NF-κB activation was enhanced by siATP1B3 treatment in HIV-1- and HIV-1ΔVpu-infected HeLa cells. In addition, ATP1B3 silencing induced high level BST-2 expression on the surface of HeLa cells. These results indicate that ATP1B3 is a co-factor that accelerates BST-2 degradation and reduces BST-2-mediated restriction of HIV-1 production and NF-κB activation.


Subject(s)
Antigens, CD/metabolism , HIV-1/physiology , Microfilament Proteins/metabolism , NF-kappa B/antagonists & inhibitors , Sodium-Potassium-Exchanging ATPase/metabolism , Antigens, CD/chemistry , Antigens, CD/genetics , Cell Line , Fibrillin-1 , Fibrillins , GPI-Linked Proteins/antagonists & inhibitors , GPI-Linked Proteins/chemistry , GPI-Linked Proteins/genetics , GPI-Linked Proteins/metabolism , Gene Deletion , Gene Expression Regulation , HEK293 Cells , HeLa Cells , Humans , Microfilament Proteins/antagonists & inhibitors , Microfilament Proteins/chemistry , Microfilament Proteins/genetics , NF-kappa B/agonists , NF-kappa B/genetics , NF-kappa B/metabolism , Protein Interaction Domains and Motifs , Protein Stability , Proteolysis , RNA Interference , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/metabolism , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Sodium-Potassium-Exchanging ATPase/antagonists & inhibitors , Sodium-Potassium-Exchanging ATPase/chemistry , Sodium-Potassium-Exchanging ATPase/genetics , Two-Hybrid System Techniques , Viral Tropism , Virus Replication
20.
J Biol Chem ; 291(14): 7373-85, 2016 Apr 01.
Article in English | MEDLINE | ID: mdl-26839315

ABSTRACT

Choroidal neovascularization (CNV) is a pathogenic process of age-related macular degeneration, a vision-threatening disease. The retinal pigment epithelium and macrophages both influence CNV development. However, the underlying mechanisms remain obscure. Here, we focus on Angptl2 (angiopoietin-like protein 2), a cytokine involved in age-related systemic diseases. Angptl2 was originally identified as an adipocytokine and is also expressed in the eye. Using a laser-induced CNV model, we found thatAngptl2KO mice exhibited suppressed CNV development with reduced macrophage recruitment and inflammatory mediator induction. The mediators monocyte chemotactic protein-1, interleukin-1ß (Il-1ß),Il-6, matrix metalloprotease-9 (Mmp-9), and transforming growth factor-ß1 (Tgf-ß1) that were up-regulated during CNV development were all suppressed in the retinal pigment epithelium-choroid of CNV models generated in theAngptl2KO mice. Bone marrow transplantation using wild-type and KO mice suggested that both bone marrow-derived and host-derived Angptl2 were responsible for macrophage recruitment and CNV development. Peritoneal macrophages derived fromAngptl2KO mice expressed lower levels of the inflammatory mediators. In the wild-type peritoneal macrophages and RAW264.7 cells, Angptl2 induced the mediators via integrins α4 and ß2, followed by the downstream activation of NF-κB and ERK. The activation of NF-κB and ERK by Angptl2 also promoted macrophage migration. Therefore, Angptl2 from focal tissue might trigger macrophage recruitment, and that from recruited macrophages might promote expression of inflammatory mediators including Angptl2 in an autocrine and/or paracrine fashion to facilitate CNV development. Angptl2 might therefore represent a multistep regulator of CNV pathogenesis and serve as a new therapeutic target for age-related macular degeneration.


Subject(s)
Angiopoietins/biosynthesis , Choroidal Neovascularization/metabolism , Macrophages/metabolism , Macular Degeneration/metabolism , Angiopoietin-Like Protein 2 , Angiopoietin-like Proteins , Angiopoietins/genetics , Animals , CD18 Antigens/genetics , CD18 Antigens/metabolism , Cell Line , Chemokine CCL2/genetics , Chemokine CCL2/metabolism , Choroidal Neovascularization/genetics , Choroidal Neovascularization/pathology , Disease Models, Animal , Humans , Inflammation/genetics , Inflammation/metabolism , Inflammation/pathology , Inflammation Mediators/metabolism , Integrin alpha4/genetics , Integrin alpha4/metabolism , Interleukin-1beta/genetics , Interleukin-1beta/metabolism , Interleukin-6/genetics , Interleukin-6/metabolism , Macrophages/pathology , Macular Degeneration/genetics , Macular Degeneration/pathology , Matrix Metalloproteinase 9/genetics , Matrix Metalloproteinase 9/metabolism , Mice , Transforming Growth Factor beta1/genetics , Transforming Growth Factor beta1/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL