Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 149
Filter
1.
J Cell Physiol ; 236(8): 5698-5714, 2021 08.
Article in English | MEDLINE | ID: mdl-33393109

ABSTRACT

Our understanding of signaling pathways regulating the cell fate of human embryonic stem cells (hESCs) is limited. Calcineurin-NFAT signaling is associated with a wide range of biological processes and diseases. However, its role in controlling hESC fate remains unclear. Here, we report that calcineurin A gamma and the NFATc3/SRPX2 axis control the expression of lineage and epithelial-mesenchymal transition (EMT) markers in hESCs. Knockdown of PPP3CC, the gene encoding calcineurin A gamma, or NFATC3, downregulates certain markers both at the self-renewal state and during differentiation of hESCs. Furthermore, NFATc3 interacts with c-JUN and regulates the expression of SRPX2, the gene encoding a secreted glycoprotein known as a ligand of uPAR. We show that SRPX2 is a downstream target of NFATc3. Both SRPX2 and uPAR participate in controlling expression of lineage and EMT markers. Importantly, SRPX2 knockdown diminishes the upregulation of multiple lineage and EMT markers induced by co-overexpression of NFATc3 and c-JUN in hESCs. Together, this study uncovers a previously unknown role of calcineurin A gamma and the NFATc3/SRPX2 axis in modulating the fate determination of hESCs.


Subject(s)
Calcineurin/metabolism , Cell Differentiation/physiology , Human Embryonic Stem Cells/cytology , Membrane Proteins/metabolism , NFATC Transcription Factors/metabolism , Neoplasm Proteins/metabolism , Cell Differentiation/genetics , Epithelial-Mesenchymal Transition/physiology , Genes, jun/physiology , Humans , Nerve Tissue Proteins/metabolism
2.
Biochemistry ; 59(4): 530-540, 2020 02 04.
Article in English | MEDLINE | ID: mdl-31804811

ABSTRACT

Basic leucine-zipper (bZIP) proteins represent difficult, yet compelling, oncogenic targets since numerous cell-signaling cascades converge upon them, where they function to modulate the transcription of specific gene targets. bZIPs are widely recognized as important regulators of cellular processes that include cell proliferation, apoptosis, and differentiation. Once such validated transcriptional regulator, activator protein-1, is typically composed of heterodimers of Fos and Jun family members, with cFos-cJun being the best described. It has been shown to be key in the progression and development of a number of different diseases. As a proof-of-principle for our approach, we describe the first use of a novel combined in silico/in cellulo peptide-library screening platform that facilitates the derivation of a sequence that displays high selectivity for cJun relative to cFos, while also avoiding homodimerization. In particular, >60 million peptides were computationally screened and all potential on/off targets ranked according to predicted stability, leading to a reduced size library that was further refined by intracellular selection. The derived sequence is predicted to have limited cross-talk with a second previously derived peptide antagonist that is selective for cFos in the presence of cJun. The study provides new insight into the use of multistate screening with the ability to combine computational and intracellular approaches in evolving multiple cocompatible peptides that are capable of satisfying conflicting design requirements.


Subject(s)
Computational Biology/methods , Proto-Oncogene Proteins c-fos/antagonists & inhibitors , Proto-Oncogene Proteins c-jun/antagonists & inhibitors , Amino Acid Sequence , Basic-Leucine Zipper Transcription Factors/chemistry , Basic-Leucine Zipper Transcription Factors/metabolism , Cell Proliferation , Computer Simulation , Dimerization , Genes, fos/physiology , Genes, jun/physiology , Humans , Oncogenes , Peptide Library , Peptides/metabolism , Protein Binding , Proto-Oncogene Proteins c-fos/metabolism , Proto-Oncogene Proteins c-jun/metabolism , Signal Transduction , Transcription Factor AP-1/chemistry , Transcription Factor AP-1/metabolism
3.
Mol Vis ; 24: 647-666, 2018.
Article in English | MEDLINE | ID: mdl-30310263

ABSTRACT

Purpose: Systemic hypertension is a risk factor of neovascular age-related macular degeneration; consumption of dietary salt resulting in extracellular hyperosmolarity is a main cause of hypertension. Extracellular hyperosmolarity was shown to induce expression of angiogenic growth factors, such as vascular endothelial growth factor (VEGF) and placental growth factor (PlGF), in RPE cells. The aim of the present study was to determine whether the hyperosmotic expression of growth factor genes in RPE cells is mediated by activator protein-1 (AP-1), and whether c-Fos and c-Jun genes are regulated by extracellular osmolarity. Methods: Hyperosmotic media were made up with the addition of NaCl or sucrose. Gene expression was quantified with real-time reverse transcription (RT)-PCR, and protein secretion was investigated with enzyme-linked immunosorbent assay (ELISA). Nuclear factor of activated T cell 5 (NFAT5) was depleted with siRNA. DNA binding of AP-1 protein was evaluated with electrophoretic mobility shift assay (EMSA). Results: High NaCl and the addition of sucrose triggered expression of the c-Fos gene, but not of the c-Jun gene. High NaCl also increased the levels of c-Fos and phosphorylated c-Jun proteins and the level of DNA binding of AP-1. Hypoosmolarity decreased the expression of the c-Fos and c-Jun genes. NaCl-induced expression of the c-Fos gene was in part mediated by NFAT5. Autocrine/paracrine activation of fibroblast growth factor and adenosine A1 receptors is involved in mediating NaCl-induced expression of the c-Fos gene. Pharmacological inhibition of the AP-1 activity decreased the NaCl-induced expression of the HIF-1α, NFAT5, VEGF, PlGF, and TGF-ß2 genes, and prevented the NaCl-induced secretion of PlGF but not of VEGF. Conclusions: The data indicate that AP-1 is activated in RPE cells in response to extracellular hyperosmolarity and mediates in part via the NaCl-induced expression of VEGF and PlGF, and secretion of PlGF. It is suggested that high consumption of dietary salt may exacerbate the angiogenic response of RPE cells in part via activation of AP-1.


Subject(s)
Gene Expression Regulation/physiology , Placenta Growth Factor/genetics , Retinal Pigment Epithelium/drug effects , Sodium Chloride/pharmacology , Transcription Factor AP-1/physiology , Vascular Endothelial Growth Factor A/genetics , Blotting, Western , Cells, Cultured , Electrophoretic Mobility Shift Assay , Enzyme-Linked Immunosorbent Assay , Genes, fos/physiology , Genes, jun/physiology , Humans , Phosphorylation , Placenta Growth Factor/metabolism , RNA, Small Interfering/genetics , Real-Time Polymerase Chain Reaction , Retinal Pigment Epithelium/metabolism , Signal Transduction/drug effects , Transcription Factor AP-1/antagonists & inhibitors , Transfection , Vascular Endothelial Growth Factor A/metabolism
4.
Dev Dyn ; 245(5): 614-26, 2016 05.
Article in English | MEDLINE | ID: mdl-26930384

ABSTRACT

BACKGROUND: Digestion is facilitated by coordinated contractions of the intestinal muscularis externa, a bilayered smooth muscle structure that is composed of inner circular muscles (ICM) and outer longitudinal muscles (OLM). We performed transcriptome analysis of intestinal mesenchyme tissue at E14.5, when the ICM, but not the OLM, is present, to investigate the transcriptional program of the ICM. RESULTS: We identified 3967 genes enriched in E14.5 intestinal mesenchyme. The gene expression profiles were clustered and annotated to known muscle genes, identifying a muscle-enriched subcluster. Using publically available in situ data, 127 genes were verified as expressed in ICM. Examination of the promoter and regulatory regions for these co-expressed genes revealed enrichment for cJUN transcription factor binding sites, and cJUN protein was enriched in ICM. cJUN ChIP-seq, performed at E14.5, revealed that cJUN regulatory regions contain characteristics of muscle enhancers. Finally, we show that cJun is a target of Hedgehog (Hh), a signaling pathway known to be important in smooth muscle development, and identify a cJun genomic enhancer that is responsive to Hh. CONCLUSIONS: This work provides the first transcriptional catalog for the developing ICM and suggests that cJun regulates gene expression in the ICM downstream of Hh signaling. Developmental Dynamics 245:614-626, 2016. © 2016 Wiley Periodicals, Inc.


Subject(s)
Gene Expression Regulation, Developmental , Intestines/embryology , Muscle, Smooth/embryology , Transcriptome , Animals , Genes, jun/physiology , Hedgehog Proteins , Mice
5.
Glia ; 62(5): 736-50, 2014 May.
Article in English | MEDLINE | ID: mdl-24526449

ABSTRACT

We investigated three models for Charcot-Marie-Tooth type 1 (CMT1) neuropathy, comprising mice lacking connexin 32 (Cx32def), mice with reduced myelin protein zero (P0) expression (P0het) and transgenic mouse mutants overexpressing peripheral myelin protein 22 (PMP22tg), with regard of the expression of the developmentally regulated molecules NCAM, L1, the low-affinity NGF-receptor p75 (p75(NTR) ) and the transcription factor component c-Jun. We found that all molecules were uniformly expressed by myelin deficient and supernumerary Schwann cells. The mutant myelinating Schwann cells of PMP22tg mice showed a robust NCAM-immunoreactivity in Schmidt-Lanterman incisures (SLI) that accompanies other early onset abnormalities, such as the presence of supernumerary Schwann cells and impaired myelin formation in some fibers. In line with this, Cx32def and P0het mice, which represent demyelinating models, only rarely express NCAM in SLI. Surprisingly, c-Jun immunoreactivity displayed a mosaic-like pattern with mostly negative and some weakly or moderately positive nuclei both in myelinating Schwann cells and Remak cells of wildtype (wt), P0het and PMP22tg mice. However, c-Jun expression was substantially upregulated in myelinating Schwann cells of Cx32def mice and spatially associated with axon perturbation, a typical predemyelinating feature of Cx32 deficiency. Additionally, c-Jun upregulation was correlated with an elevated level of GDNF, possibly causally linked to the typical compensatory sprouting of axons in Cx32def mice and CMT1X patients. Our findings suggest that in myelinating Schwann cells of distinct models of CMT1, c-Jun upregulation is a marker for predemyelinating axonal perturbation while myelin-related NCAM expression is indicative for early Schwann cell abnormalities.


Subject(s)
Charcot-Marie-Tooth Disease/metabolism , Genes, jun/physiology , Nerve Fibers, Myelinated/metabolism , Neural Cell Adhesion Molecules/biosynthesis , Schwann Cells/metabolism , Up-Regulation/physiology , Animals , Axons/metabolism , Axons/pathology , Charcot-Marie-Tooth Disease/pathology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Nerve Fibers, Myelinated/pathology , Schwann Cells/pathology
6.
Glia ; 61(12): 2063-77, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24123203

ABSTRACT

Astrocyte activation is a hallmark of central nervous system injuries resulting in glial scar formation (astrogliosis). The activation of astrocytes involves metabolic and morphological changes with complex underlying mechanisms, which should be defined to provide targets for astrogliosis intervention. Astrogliosis is usually accompanied by an upregulation of glial fibrillary acidic protein (GFAP). Using an in vitro scratch injury model, we scratched primary cultures of cerebral cortical astrocytes and observed an influx of calcium in the form of waves spreading away from the wound through gap junctions. Using the calcium blocker BAPTA-AM and the JNK inhibitor SP600125, we demonstrated that the calcium wave triggered the activation of JNK, which then phosphorylated the transcription factor c-Jun to facilitate the binding of AP-1 to the GFAP gene promoter to switch on GFAP upregulation. Blocking calcium mobilization with BAPTA-AM in an in vivo stab wound model reduced GFAP expression and glial scar formation, showing that the calcium signal, and the subsequent regulation of downstream signaling molecules, plays an essential role in brain injury response. Our findings demonstrated that traumatic scratch injury to astrocytes triggered a calcium influx from the extracellular compartment and activated the JNK/c-Jun/AP-1 pathway to switch on GFAP expression, identifying a previously unreported signaling cascade that is important in astrogliosis and the physiological response following brain injury.


Subject(s)
Astrocytes/metabolism , Calcium/metabolism , Genes, jun/physiology , Glial Fibrillary Acidic Protein/metabolism , Gliosis/metabolism , MAP Kinase Signaling System/physiology , Transcription Factor AP-1/metabolism , Animals , Astrocytes/cytology , Calcium Signaling/genetics , Cells, Cultured , Cerebral Cortex/cytology , Cerebral Cortex/metabolism , Gliosis/genetics , Mice , Mice, Inbred ICR , Transcription Factor AP-1/genetics , Transcriptional Activation
7.
Gut ; 61(9): 1269-1278, 2012 Sep.
Article in English | MEDLINE | ID: mdl-21997551

ABSTRACT

OBJECTIVE: Inflammatory bowel diseases (IBDs) feature multiple cellular stress responses, including endoplasmic reticulum (ER) unfolded protein responses (UPRs). UPRs represent autoregulatory pathways that adjust organelle capacity to cellular demand. A similar mechanism, mitochondrial UPR (mtUPR), has been described for mitochondria. ER UPR in intestinal epithelial cells (IECs) contributes to the development of intestinal inflammation, and since mitochondrial alterations and dysfunction are implicated in the pathogenesis of IBDs, the authors characterised mtUPR in the context of intestinal inflammation. METHODS: Truncated ornithine transcarbamylase was used to selectively induce mtUPR in a murine IEC line. Dextran sodium sulphate (DSS) was administered to PKR (double-stranded-RNA-activated protein kinase) knockout mice to induce IEC stress in vivo and to test for their susceptibility to DSS-induced colitis. Expression levels of the mitochondrial chaperone chaperonin 60 (CPN60) and PKR were quantified in IECs from patients with IBDs and from murine models of colitis using immunohistochemistry and Western blot analysis. RESULTS: Selective mtUPR induction by truncated ornithine transcarbamylase transfection triggered the phosphorylation of eukaryotic translation initiation factor (eIF) 2α and cJun through the recruitment of PKR. Using pharmacological inhibitors and small inhibitory RNA, the authors identified mtUPR-induced eIF2α phosphorylation and transcription factor activation (cJun/AP1) as being dependent on the activities of the mitochondrial protease ClpP and the cytoplasmic kinase PKR. Pkr(-/-) mice failed to induce CPN60 in IECs upon DSS treatment at early time points and subsequently showed an almost complete resistance to DSS-induced colitis. Under inflammatory conditions, primary IECs from patients with IBDs and two murine models of colitis exhibited a strong induction of the mtUPR marker protein CPN60 associated with enhanced expression of PKR. CONCLUSION: PKR integrates mtUPR into the disease-relevant ER UPR via eIF2α phosphorylation and AP1 activation. Induction of mtUPR and PKR was observed in IECs from murine models and patients with IBDs. The authors' results indicate that PKR might link mitochondrial stress to intestinal inflammation.


Subject(s)
Colitis/enzymology , Colitis/pathology , Mitochondria/metabolism , Unfolded Protein Response/physiology , eIF-2 Kinase/biosynthesis , Animals , Blotting, Western , Cells, Cultured , Chaperonin 60/metabolism , Enzyme Activation , Epithelial Cells/enzymology , Eukaryotic Initiation Factor-2/metabolism , Genes, jun/physiology , Humans , Immunohistochemistry , Mice , Mice, Knockout , Phosphorylation , Signal Transduction/physiology , Transfection
8.
Am J Physiol Gastrointest Liver Physiol ; 302(6): G618-27, 2012 Mar 15.
Article in English | MEDLINE | ID: mdl-22159277

ABSTRACT

DRA (downregulated in adenoma) or SLC26A3 is the major apical anion exchanger mediating Cl(-) absorption in intestinal epithelial cells. Disturbances in DRA function and expression have been implicated in diarrheal conditions such as congenital chloride diarrhea and inflammatory bowel diseases. Previous studies have shown that DRA is subject to regulation by short-term and transcriptional mechanisms. In this regard, we have recently shown that short-term treatment by lysophosphatidic acid (LPA), an important bioactive phospholipid, stimulates Cl(-)/HCO(3)(-)(OH(-)) exchange activity via an increase in DRA surface levels in human intestinal epithelial cells. However, the long-term effects of LPA on DRA at the level of gene transcription have not been examined. The present studies were aimed at investigating the effects of LPA on DRA function and expression as well as elucidating the mechanisms underlying its transcriptional regulation. Long-term LPA treatment increased the Cl(-)/HCO(3)(-) exchange activity in Caco-2 cells. LPA treatment (50-100 µM) of Caco-2 cells significantly stimulated DRA mRNA levels and DRA promoter activity (-1183/+114). This increase in DRA promoter activity involved the LPA2 receptor and phosphatidylinositol 3-kinase (PI3K)/AKT pathways. Progressive deletions from -1183/+114 to -790/+114 abrogated the stimulatory effects of LPA, indicating that the -1183/-790 promoter region harbors LPA response elements. Utilizing EMSA and mutational studies, our results showed that LPA induced the DRA promoter activity in a c-Fos-dependent manner. LPA also increased the protein expression of c-Fos and c-Jun in Caco-2 cells. Furthermore, overexpression of c-Fos but not c-Jun enhanced the DRA promoter activity. This increase in DRA transcription in response to LPA indicates that LPA may act as an antidiarrheal agent and could be exploited for the treatment of diarrhea associated with inflammatory or infectious diseases of the gut.


Subject(s)
Chloride-Bicarbonate Antiporters/metabolism , Genes, fos/physiology , Lysophospholipids/pharmacology , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Receptors, Lysophosphatidic Acid/metabolism , Amino Acid Transport Systems/genetics , Amino Acid Transport Systems/metabolism , Caco-2 Cells , Chloride-Bicarbonate Antiporters/drug effects , Chloride-Bicarbonate Antiporters/genetics , Dose-Response Relationship, Drug , Gene Expression Regulation/drug effects , Genes, fos/drug effects , Genes, fos/genetics , Genes, jun/drug effects , Genes, jun/physiology , Humans , Phosphatidylinositol 3-Kinases/genetics , Promoter Regions, Genetic/drug effects , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-fos/drug effects , Proto-Oncogene Proteins c-fos/metabolism , Proto-Oncogene Proteins c-jun/drug effects , Proto-Oncogene Proteins c-jun/metabolism , Sulfate Transporters , Symporters/genetics , Symporters/metabolism , Transcription, Genetic/drug effects
9.
Mol Reprod Dev ; 79(2): 128-37, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22128093

ABSTRACT

Obesity increases the risk of female reproductive tract cancers, but the underlying mechanistic link between the two is ill-defined. Thus, the objective of the current study was to identify obesity-dependent changes in the expression of immediate early (IE) genes that contribute to cell proliferation and differentiation, and epithelial-mesenchymal transition (EMT) genes that promote cell migration. When HeLa cells were treated for 0-48 hr with IGF-1, leptin, TNFα, or IL-6, each individual adipocytokine altered the abundance of IE (cJUN, cFOS, and cMYC) and EMT (SNAI1, SNAI2, and TWIST1) mRNA abundance. For example, IGF-1 increased cJUN and cFOS and decreased cMYC; leptin increased cFOS; IL-6 increased cFOS and cMYC; and TNFα increased cJUN and cFOS mRNA abundance. Likewise, EMT gene expression was altered by IGF-1, TNFα, and IL-6. SNAI1 was increased by IGF-1 and IL-6; SNAI2 was increased by IGF-1 and TNFα; and TWIST1 was increased by TNFα and IL-6. Chronic exposure to adipocytokines also altered EMT gene expression in the whole uterus of obese compared to normal-weight mice. Specifically, there was no difference in cJun, cFos, or cMyc mRNA abundance between normal-weight and obese animals. Snai1, Snai2, and Twist1 mRNA abundance, however, was increased in the uterus of obese females and correlated with increased circulating IGF-1 levels. These data indicate that obesity-dependent alterations in adipocytokine levels regulate the expression of genes associated with cell proliferation and migration, and therefore may provide a plausible mechanism for obesity-dependent increases in cancers of the female reproductive tract.


Subject(s)
Adipokines/pharmacology , Epithelial-Mesenchymal Transition/drug effects , Epithelial-Mesenchymal Transition/genetics , Genes, Immediate-Early/physiology , Genitalia, Female/drug effects , Adipokines/genetics , Adipokines/metabolism , Animals , Cell Differentiation/genetics , Cell Movement/genetics , Cell Proliferation/drug effects , Female , Genes, fos/physiology , Genes, jun/physiology , Genes, myc/physiology , Genitalia, Female/metabolism , Genitalia, Female/physiology , HeLa Cells , Humans , Mice , Mice, Inbred C57BL
10.
J Biomed Sci ; 19: 96, 2012 Nov 17.
Article in English | MEDLINE | ID: mdl-23157661

ABSTRACT

BACKGROUND: Based on an experimental brain stem death model, we demonstrated previously that activation of the mitogen-activated protein kinase kinase 1/2 (MEK1/2)/extracellular signal-regulated kinase 1/2 (ERK1/2)/ mitogen-activated protein kinase signal-interacting kinase 1/2 (MNK1/2) cascade plays a pro-life role in the rostral ventrolateral medulla (RVLM), the origin of a life-and-death signal detected from systemic arterial pressure, which sequentially increases (pro-life) and decreases (pro-death) to reflect progressive dysfunction of central cardiovascular regulation during the advancement towards brain stem death in critically ill patients. The present study assessed the hypothesis that, in addition to ERK1/2, c-Jun NH2-terminal kinase (JNK) and p38 mitogen-activated protein kinase (p38MAPK), the other two mammalian members of MAPKs that are originally identified as stress-activated protein kinases, are activated specifically by MAPK kinase 4 (MAP2K4) or MAP2K6 and play a pro-life role in RVLM during experimental brain stem death. We further delineated the participation of phosphorylating activating transcriptional factor-2 (ATF-2) and c-Jun, the classical transcription factor activated by JNK or p38MAPK, in this process. RESULTS: An experimental model of brain stem death that employed microinjection of the organophosphate insecticide mevinphos (Mev; 10 nmol) bilaterally into RVLM of Sprague-Dawley rats was used, alongside cardiovascular, pharmacological and biochemical evaluations. Results from ELISA showed that whereas the total JNK, p38MAPK, MAP2K4 and MAP2K6 were not affected, augmented phosphorylation of JNK at Thr183 and Tyr185 and p38MAPK at Thr180 and Tyr182, accompanied by phosphorylation of their upstream activators MAP2K4 at Ser257 and Thr261 and MAP2K6 at Ser207 and Thr211 in RVLM occurred preferentially during the pro-life phase of experimental brain stem death. Moreover, the activity of transcription factors ATF-2 at Thr71 and c-Jun at Ser73, rather than Elk-1 at Ser383 in RVLM were also augmented during the pro-life phase. Furthermore, pretreatment by microinjection into the bilateral RVLM of specific JNK inhibitors, JNK inhibitor I (100 pmol) or SP600125 (5 pmol), or specific p38MAPK inhibitors, p38MAPK inhibitor III (500 pmol) or SB203580 (2 nmol), exacerbated the depressor effect and blunted the augmented life-and-death signal exhibited during the pro-life phase. On the other hand, pretreatment with the negative control for JNK or p38MAPK inhibitor, JNK inhibitor I negative control (100 pmol) or SB202474 (2 nmol), was ineffective in the vehicle-controls and Mev-treatment groups. CONCLUSIONS: Our results demonstrated that activation of JNK or p38MAPK in RVLM by their upstream activators MAP2K4 or MAP2K6 plays a preferential pro-life role by sustaining the central cardiovascular regulatory machinery during experimental brain stem death via phosphorylation and activation of nuclear transcription factor ATF-2 or c-Jun.


Subject(s)
Brain Death , Brain Stem , JNK Mitogen-Activated Protein Kinases/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism , Activating Transcription Factors/metabolism , Activating Transcription Factors/physiology , Animals , Brain Death/metabolism , Brain Death/physiopathology , Brain Stem/metabolism , Brain Stem/physiopathology , Genes, jun/physiology , Humans , Imidazoles/pharmacology , MAP Kinase Kinase 4/metabolism , MAP Kinase Kinase 6/metabolism , Male , Mevinphos/toxicity , Phosphorylation/drug effects , Rats , Rats, Sprague-Dawley , Signal Transduction/drug effects
11.
J Biol Chem ; 285(4): 2340-50, 2010 Jan 22.
Article in English | MEDLINE | ID: mdl-19910461

ABSTRACT

The SWI/SNF chromatin-remodeling complex has been implicated in the activation and proliferation of T cells. After T cell receptor signaling, the SWI/SNF complex rapidly associates with chromatin and controls gene expression in T cells. However, the process by which the SWI/SNF complex regulates peripheral T cell activation has not been elucidated. In this study, we show that the SWI/SNF complex regulates cytokine production and proliferation of T cells. During T cell activation, the SWI/SNF complex is recruited to the promoter of the transcription factor AP-1, and it increases the expression of AP-1. Increased expression of the SWI/SNF complex resulted in enhanced AP-1 activity, cytokine production, and proliferation of peripheral T cells, whereas knockdown of the SWI/SNF complex expression impaired the AP-1 expression and reduced the activation and proliferation of T cells. Moreover, mice that constitutively expressed the SWI/SNF complex in T cells were much more susceptible to experimentally induced autoimmune encephalomyelitis than the normal mice were. These results suggest that the SWI/SNF complex plays a critical role during T cell activation and subsequent immune responses.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental/physiopathology , Lymphocyte Activation/physiology , T-Lymphocytes/physiology , Transcription Factor AP-1/genetics , Transcription Factors/immunology , Animals , CD2 Antigens/genetics , CD2 Antigens/metabolism , Cell Division/immunology , Cell Line , Cytokines/metabolism , Encephalomyelitis, Autoimmune, Experimental/immunology , Female , Gene Expression Regulation/immunology , Genes, fos/physiology , Genes, jun/physiology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Promoter Regions, Genetic/physiology , T-Lymphocytes/cytology , Transcription Factors/genetics , Transcription Factors/metabolism
12.
Cells ; 10(9)2021 09 21.
Article in English | MEDLINE | ID: mdl-34572149

ABSTRACT

The inflammatory cytokine interleukin-26 (IL-26) is highly expressed in the serum and synovial fluid of patients with inflammatory arthritis. The effect of IL-26 on human articular chondrocytes (HACs) remains unclear. Obesity is associated with disability of patients with rheumatoid arthritis and disease activity in those with ankylosing spondylitis. The saturated free fatty acid palmitate with IL-1ß can synergistically induce catabolic effects in HACs. The aim of this study was to evaluate the effects of IL-26 and palmitate in HACs. In this study, palmitate markedly synergizes the IL-26-induced proinflammatory effects and matrix protease, including COX-2, IL-6, and MMP-1, in HACs via the toll-like receptor 4 (TLR4)-ERK1/2-c-Jun signal transduction pathway. The synergistic catabolic effects of palmitate and IL-26 were attenuated by inhibitors of TLR4 (TAK242), ERK1/2 (U0126), or c-Jun (SP600125) in HACs and cartilage matrix. In addition, metformin, a potential inhibitor of TLR4, also decreased expression of COX-2 and IL-6 induced by co-incubation with IL-26 and palmitate. IL-26 and palmitate synergistically induced expression of inflammatory and catabolic mediators, resulting in articular cartilage matrix breakdown. The present study also revealed a possible mechanism and therapeutic targets against articular cartilage degradation by increased saturated fatty acids in patients with inflammatory arthritis.


Subject(s)
Chondrocytes/metabolism , Interleukins/metabolism , Palmitates/metabolism , Arthritis/immunology , Arthritis/metabolism , Arthritis/physiopathology , Arthritis, Rheumatoid/metabolism , Cartilage, Articular/metabolism , Chondrocytes/physiology , Genes, jun/physiology , Humans , Interleukins/immunology , MAP Kinase Signaling System/physiology , Metabolism/physiology , Osteoarthritis/metabolism , Signal Transduction/genetics , Synovial Membrane/metabolism , Taiwan , Toll-Like Receptor 4/metabolism
13.
Mol Endocrinol ; 23(3): 402-11, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19131506

ABSTRACT

GnRH regulates gonadotrope function through a complex transcriptional network that includes three members of the immediate early gene family: Egr1, Jun, and Atf3. These DNA-binding proteins act alone or in pairs to confer hormonal responsiveness to Cga, Lhb, Fshb, and Gnrhr. Herein we suggest that the transcriptional response of Jun requires a functional interaction between the T-cell factor (TCF)/lymphoid enhancer factor (LEF) family of DNA-binding proteins and beta-catenin (officially CTNNB1), a coactivator of TCF/LEF. Supporting data include demonstration that GnRH increases activity of TOPflash, a TCF/LEF-dependent luciferase reporter, in LbetaT2 cells, a gonadotrope-derived cell line. Additional cotransfection experiments indicate that a dominant-negative form of TCF7L2 (TCFDN) that binds DNA, but not beta-catenin, blocks GnRH induction of TOPflash. Overexpression of AXIN, an inhibitor of beta-catenin, also reduces GnRH stimulation of TOPflash. Transduction of LbetaT2 cells with TCFDN adenoviruses diminishes GnRH stimulation of Jun mRNA without altering expression of Egr1 and Atf3, two other immediate early genes that confer GnRH responsiveness. Reduction of beta-catenin in LbetaT2 cells, through stable expression of short hairpin RNA, also selectively compromises GnRH regulation of Jun expression and levels of JUN protein. Finally, overexpression of TCFDN attenuates GnRH regulation of Cga promoter activity, a known downstream target of JUN. Together, these results indicate that GnRH regulation of Jun transcription requires a functional interaction between TCF/LEF and beta-catenin and that alteration of either impacts expression of JUN downstream targets such as Cga.


Subject(s)
Gene Regulatory Networks/drug effects , Genes, jun/drug effects , Gonadotrophs/drug effects , Gonadotropin-Releasing Hormone/pharmacology , TCF Transcription Factors/metabolism , beta Catenin/metabolism , Cells, Cultured , Gene Expression Regulation/drug effects , Genes, Dominant , Genes, jun/physiology , Glycoprotein Hormones, alpha Subunit/genetics , Gonadotrophs/metabolism , Humans , Promoter Regions, Genetic/drug effects , Protein Binding/drug effects , TCF Transcription Factors/genetics , TCF Transcription Factors/physiology , Transfection , beta Catenin/physiology
14.
Dev Cell ; 4(6): 865-77, 2003 Jun.
Article in English | MEDLINE | ID: mdl-12791271

ABSTRACT

The migration of epithelial layers requires specific and coordinated organization of the cells at the leading edge of the sheet. Mice that are conditionally deleted for the c-jun protooncogene in epidermis are born at expected frequencies, but with open eyes and with defects in epidermal wound healing. Keratinocytes lacking c-Jun are unable to migrate or elongate properly in culture at the border of scratch assays. Histological analyses in vitro and in vivo demonstrate an inability to activate EGF receptor at the leading edge of wounds, and we demonstrate that this can be rescued by supplementation with conditioned medium or the EGF receptor ligand HB-EGF. Lack of c-Jun prevents EGF-induced expression of HB-EGF, indicating that c-jun controls formation of the epidermal leading edge through its control of an EGF receptor autocrine loop.


Subject(s)
Epidermis/growth & development , ErbB Receptors/metabolism , Genes, jun/physiology , Animals , Cell Division , Cell Movement , Cells, Cultured , Epidermal Cells , Epidermal Growth Factor/metabolism , Epidermis/injuries , Eyelids/abnormalities , Eyelids/embryology , Gene Deletion , Heparin-binding EGF-like Growth Factor , Intercellular Signaling Peptides and Proteins , Keratinocytes/cytology , Keratinocytes/physiology , Keratins/metabolism , Mice , Mice, Transgenic , Mutation , Phosphoprotein Phosphatases , Proliferating Cell Nuclear Antigen/metabolism , Time Factors , Transcription Factor AP-1/metabolism
15.
J Cell Biol ; 131(4): 1095-103, 1995 Nov.
Article in English | MEDLINE | ID: mdl-7490285

ABSTRACT

Milk production during lactation is a consequence of the suckling stimulus and the presence of glucocorticoids, prolactin, and insulin. After weaning the glucocorticoid hormone level drops, secretory mammary epithelial cells die by programmed cell death and the gland is prepared for a new pregnancy. We studied the role of steroid hormones and prolactin on the mammary gland structure, milk protein synthesis, and on programmed cell death. Slow-release plastic pellets containing individual hormones were implanted into a single mammary gland at lactation. At the same time the pups were removed and the consequences of the release of hormones were investigated histologically and biochemically. We found a local inhibition of involution in the vicinity of deoxycorticosterone- and progesterone-release pellets while prolactin-release pellets were ineffective. Dexamethasone, a very stable and potent glucocorticoid hormone analogue, inhibited involution and programmed cell death in all the mammary glands. It led to an accumulation of milk in the glands and was accompanied by an induction of protein kinase A, AP-1 DNA binding activity and elevated c-fos, junB, and junD mRNA levels. Several potential target genes of AP-1 such as stromelysin-1, c-jun, and SGP-2 that are induced during normal involution were strongly inhibited in dexamethasone-treated animals. Our results suggest that the cross-talk between steroid hormone receptors and AP-1 previously described in cells in culture leads to an impairment of AP-1 activity and to an inhibition of involution in the mammary gland implying that programmed cell death in the postlactational mammary gland depends on functional AP-1.


Subject(s)
Apoptosis/physiology , Glucocorticoids/physiology , Mammary Glands, Animal/cytology , Molecular Chaperones , Progesterone/physiology , Animals , Base Sequence , Clusterin , Complement Inactivator Proteins/genetics , Dexamethasone/pharmacology , Endocrine Glands/drug effects , Epithelial Cells , Female , Genes, jun/physiology , Glycoproteins/genetics , Mammary Glands, Animal/physiology , Matrix Metalloproteinase 3 , Metalloendopeptidases/genetics , Mice , Mice, Inbred Strains , Molecular Sequence Data , Neoplasm Proteins/genetics , Transcription Factor AP-1/antagonists & inhibitors
16.
Science ; 255(5051): 1581-4, 1992 Mar 20.
Article in English | MEDLINE | ID: mdl-1549784

ABSTRACT

The suprachiasmatic nuclei (SCN) of the hypothalamus comprise the primary pacemaker responsible for generation of circadian rhythms in mammals. Light stimuli that synchronize this circadian clock induce expression of the c-fos gene in rodent SCN, which suggests a possible role for Fos in circadian entrainment. Appropriate light stimuli also induce the expression of jun-B messenger RNA in the SCN of golden hamsters but only slightly elevate c-jun messenger RNA levels. In addition, light increases the amount of a protein complex in the SCN that binds specifically to sites on DNA known to mediate regulation by the AP-1 transcription factor. The photic regulation of both jun-B messenger RNA expression and AP-1 binding activity is dependent on circadian phase: only light stimuli that shift behavioral rhythms induce jun-B and AP-1 expression. Thus, light and the circadian pacemaker interact to regulate a specific set of immediate-early genes in the SCN that may participate in entrainment of the circadian clock.


Subject(s)
Gene Expression Regulation , Genes, jun/physiology , Light , Periodicity , Proto-Oncogene Proteins c-jun/biosynthesis , RNA, Messenger/biosynthesis , Animals , Base Sequence , Cricetinae , Genes, fos/physiology , Molecular Sequence Data , Nucleic Acid Hybridization , RNA Probes , Suprachiasmatic Nucleus/physiology , Time Factors , Transcription, Genetic
17.
Toxicon ; 53(1): 169-75, 2009 Jan.
Article in English | MEDLINE | ID: mdl-19041662

ABSTRACT

Microcystins (MCs) are a potent liver tumor promoter, possessing potent tumor-promoting activity and weak initiating activity. Proto-oncogenes are known to be involved in the tumor-promoting mechanisms of microcystin-LR. However, few data are available on the effects of MCs on proto-oncogenes in the whole animal. To investigate the effects of MCs on the expression profile of the proto-oncogenes in different organs, male Wistar rats were injected intravenously with microcystin extracts at a dose of 86.7 mug MC-LR eq/kg bw (MC-LR eq, MC-LR equivalents). mRNA levels of three proto-oncogenes c-fos, c-jun and c-myc in liver, kidney and testis were analyzed using quantitative real-time PCR at several time points post-injection. Significant induction of these genes at transcriptional level was observed in the three organs. In addition, the increase of mRNA expression of all three genes was much higher in liver than in kidney and testis. Meanwhile, the protein levels of c-Fos and c-Jun were investigated by western blotting. Both proteins were induced in the three organs. However, elevations of protein levels were much lower than those of mRNA levels. These findings suggest that the expression of c-fos, c-jun and c-myc might be one possible mechanism for the tumor-promoting activity and initiating activity of microcystins.


Subject(s)
Microcystins/pharmacology , Proto-Oncogene Proteins c-fos/metabolism , Proto-Oncogene Proteins c-jun/metabolism , Proto-Oncogene Proteins c-myc/metabolism , Animals , Gene Expression Regulation, Neoplastic/drug effects , Genes, fos/drug effects , Genes, fos/genetics , Genes, fos/physiology , Genes, jun/drug effects , Genes, jun/genetics , Genes, jun/physiology , Genes, myc/drug effects , Genes, myc/genetics , Genes, myc/physiology , Kidney/drug effects , Kidney/metabolism , Liver/drug effects , Liver/metabolism , Male , Microcystins/chemistry , Proto-Oncogene Proteins c-fos/genetics , Proto-Oncogene Proteins c-jun/genetics , Proto-Oncogene Proteins c-myc/genetics , Rats , Rats, Wistar , Testis/drug effects , Testis/metabolism
18.
Clin Cancer Res ; 14(14): 4408-16, 2008 Jul 15.
Article in English | MEDLINE | ID: mdl-18628455

ABSTRACT

PURPOSE: Replicative senescence in cells acts as a barrier against excessive proliferation and carcinogenesis. Transient amplifying cells (TAC) are a subset of basal cell populations within the prostate from which cancers are thought to originate; therefore, we focused on prostate TAC to investigate the molecular mechanisms by which the TAC may be able to evade senescence. EXPERIMENTAL DESIGN: TAC clones were isolated from each zone within the whole prostate and analyzed in flow cytometry. Prostate cancer cells were transfected with junB small interfering RNA (siRNA) and examined by chorioallantoic membrane assay for cancer invasion. Immunohistochemical analysis was done in primary and metastatic prostate cancer specimens. RESULTS: TAC populations showed increased expression of p53, p21, p16, and pRb, resulting in senescence. TAC clones with reduced p16 expression successfully bypassed this phase. We further found close correlation between the levels of junB and p16 expression. Repeated transfection of junB siRNA in prostatic TAC allowed the cells to escape senescence presumably through inactivation of p16/pRb. The chorioallantoic membrane invasion assay showed much lower in invasive cancer cells with high expression of junB; conversely, silencing of junB by transfection with junB siRNA promoted invasion. We also found that metastatic prostate cancers, as well as cancers with high Gleason scores, showed significantly low junB immunopositivity. CONCLUSIONS: JunB is an essential upstream regulator of p16 and contributes to maintain cell senescence that blocks malignant transformation of TAC. JunB thus apparently plays an important role in controlling prostate carcinogenesis and may be a new target for cancer prevention and therapy.


Subject(s)
Cell Transformation, Neoplastic/metabolism , Cellular Senescence/physiology , Genes, jun/physiology , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Aged , Blotting, Western , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/pathology , Cyclin-Dependent Kinase Inhibitor p16/metabolism , Disease Progression , Flow Cytometry , Gene Expression , Humans , Immunohistochemistry , Male , Middle Aged , Prostatic Neoplasms/genetics , RNA, Small Interfering , Retinoblastoma Protein/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Transfection
19.
Mol Cancer Ther ; 7(4): 961-71, 2008 Apr.
Article in English | MEDLINE | ID: mdl-18413810

ABSTRACT

NAG-1 (nonsteroidal anti-inflammatory drug-activated gene), a member of the transforming growth factor-beta superfamily, is involved in many cellular processes, such as inflammation, apoptosis/survival, and tumorigenesis. Vitamin E succinate (VES) is the succinate derivative of alpha-tocopherol and has antitumorigenic activity in a variety of cell culture and animal models. In the current study, the regulation and role of NAG-1 expression in PC-3 human prostate carcinoma cells by VES was examined. VES treatment induced growth arrest and apoptosis as well as an increase in NAG-1 protein and mRNA levels in a time- and concentration-dependent manner. VES treatment induced nuclear translocation and activation of p38 kinase. Pretreatment with p38 kinase inhibitor blocked the VES-induced increase in NAG-1 protein and mRNA levels, whereas an inhibition of protein kinase C, Akt, c-Jun NH(2)-terminal kinase, or MEK activity had no effect on VES-induced NAG-1 levels. Forced expression of constitutively active MKK6, an upstream kinase for p38, induced an increase in NAG-1 promoter activity, whereas p38 kinase inhibitor blocked MKK6-induced increase in NAG-1 promoter activity. VES treatment resulted in >3-fold increase in the half-life of NAG-1 mRNA in a p38 kinase-dependent manner and transient transfection experiment showed that VES stabilizes NAG-1 mRNA through AU-rich elements in 3'-untranslated region of NAG-1 mRNA. The inhibition of NAG-1 expression by small interfering RNA significantly blocked VES-induced poly(ADP-ribose) polymerase cleavage, suggesting that NAG-1 may play an important role in VES-induced apoptosis. These results indicate that VES-induced expression of NAG-1 mRNA/protein is regulated by transcriptional/post-transcriptional mechanism in a p38 kinase-dependent manner and NAG-1 can be chemopreventive/therapeutic target in prostate cancer.


Subject(s)
Antioxidants/pharmacology , Cytokines/genetics , Prostatic Neoplasms/metabolism , Vitamin E/pharmacology , p38 Mitogen-Activated Protein Kinases/metabolism , 3' Untranslated Regions/genetics , Antioxidants/chemistry , Apoptosis/drug effects , Blotting, Northern , Blotting, Western , Cell Nucleus/metabolism , Cell Proliferation/drug effects , Cytokines/antagonists & inhibitors , Cytokines/metabolism , Fluorescent Antibody Technique , Genes, jun/physiology , Growth Differentiation Factor 15 , Humans , Luciferases , MAP Kinase Kinase 6/antagonists & inhibitors , MAP Kinase Kinase 6/metabolism , MAP Kinase Kinase Kinases/antagonists & inhibitors , MAP Kinase Kinase Kinases/metabolism , Male , Promoter Regions, Genetic/genetics , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/pathology , Protein Kinase C/antagonists & inhibitors , Protein Kinase C/metabolism , Protein Transport , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Proto-Oncogene Proteins c-akt/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Transcription, Genetic/drug effects , Transfection , Tumor Cells, Cultured , p38 Mitogen-Activated Protein Kinases/genetics
20.
Oncogene ; 26(20): 2885-93, 2007 May 03.
Article in English | MEDLINE | ID: mdl-17146445

ABSTRACT

Glioblastoma multiforme (GBM) is the highest grade of astrocytoma. GBM pathogenesis has been linked to receptor tyrosine kinases and kinases further down signal-transduction pathways - in particular, members of the protein kinase C (PKC) family. The expression and activity of various PKC isoforms are increased in malignant astrocytomas, but not in non-neoplastic astrocytes. This suggests that PKC activity contributes to tumor progression. The level of PKC-eta expressed correlates with the degree of phorbol-12-myristate-13-acetate (PMA)-induced proliferation of two glioblastoma cell lines, U-1242 MG and U-251 MG. Normally, U-1242 cells do not express PKC-eta, and PMA inhibits their proliferation. Conversely, PMA increases proliferation of U-1242 cells that are stably transfected with PKC-eta (U-1242-PKC-eta). PMA treatment also stimulates proliferation of U-251 cells, which express PKC-eta. Here, we determined that extracellular signal-regulated kinase (ERK) and Elk-1 are downstream targets of PKC-eta. Elk-1-mediated transcriptional activity correlates with the PKC-eta-mediated mitogenic response. Pretreatment of U-1242-PKC-eta cells with inhibitors of PKC or MAPK/ERK kinase (MEK) (bisindolyl maleimide (BIM) or U0126, respectively) blocked both PMA-induced Elk-1 transcriptional activity and PMA-stimulated proliferation. An overexpressed dominant-negative PKC-eta reduced the mitogenic response in U-251 cells, as did reduction of Elk-1 by small interfering RNA. Taken together, these results strongly suggest that PKC-eta-mediated glioblastoma proliferation involves MEK/mitogen-activated protein (MAP) kinase phosphorylation, activation of ERK and subsequently of Elk-1. Elk-1 target genes involved in GBM proliferative responses have yet to be identified.


Subject(s)
Cell Proliferation , Extracellular Signal-Regulated MAP Kinases/physiology , Glioblastoma/pathology , Protein Kinase C/physiology , ets-Domain Protein Elk-1/physiology , Cell Line, Tumor , Cell Proliferation/drug effects , Epidermal Growth Factor/pharmacology , Extracellular Signal-Regulated MAP Kinases/metabolism , Genes, fos/physiology , Genes, jun/physiology , Humans , Isoenzymes/physiology , Models, Biological , Phosphorylation , Signal Transduction , Tetradecanoylphorbol Acetate/pharmacology , Transcription Factor AP-1/genetics , Transcription, Genetic/genetics , Transfection
SELECTION OF CITATIONS
SEARCH DETAIL