Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 491
1.
Endocrinology ; 163(2)2022 02 01.
Article En | MEDLINE | ID: mdl-34967898

Leydig cells produce androgens that are essential for male sex differentiation and reproductive function. Leydig cell function is regulated by several hormones and signaling molecules, including growth hormone (GH). Although GH is known to upregulate Star gene expression in Leydig cells, its molecular mechanism of action remains unknown. The STAT5B transcription factor is a downstream effector of GH signaling in other systems. While STAT5B is present in both primary and Leydig cell lines, its function in these cells has yet to be ascertained. Here we report that treatment of MA-10 Leydig cells with GH or overexpression of STAT5B induces Star messenger RNA levels and increases steroid hormone output. The mouse Star promoter contains a consensus STAT5B element (TTCnnnGAA) at -756 bp to which STAT5B binds in vitro (electrophoretic mobility shift assay and supershift) and in vivo (chromatin immunoprecipitation) in a GH-induced manner. In functional promoter assays, STAT5B was found to activate a -980 bp mouse Star reporter. Mutating the -756 bp element prevented STAT5B binding but did not abrogate STAT5B-responsiveness. STAT5B was found to functionally cooperate with DNA-bound cJUN. The STAT5B/cJUN cooperation was only observed in Leydig cells and not in Sertoli or fibroblast cells, indicating that additional Leydig cell-enriched transcription factors are required. The STAT5B/cJUN cooperation was lost only when both STAT5B and cJUN elements were mutated. In addition to identifying the Star gene as a novel target for STAT5B in Leydig cells, our data provide important new insights into the mechanism of GH and STAT5B action in the regulation of Leydig cell function.


Growth Hormone/pharmacology , Leydig Cells/metabolism , Phosphoproteins/genetics , Proto-Oncogene Proteins c-jun/physiology , STAT5 Transcription Factor/genetics , Animals , Base Sequence , Binding Sites , Cell Line , DNA/chemistry , DNA/metabolism , Gene Expression/drug effects , Leydig Cells/classification , Male , Mice , Phosphoproteins/analysis , Phosphoproteins/physiology , Promoter Regions, Genetic , RNA, Messenger/analysis , STAT5 Transcription Factor/analysis , STAT5 Transcription Factor/physiology , Up-Regulation/drug effects
2.
J Orthop Surg Res ; 16(1): 259, 2021 Apr 14.
Article En | MEDLINE | ID: mdl-33853636

BACKGROUND: Osteoarthritis (OA) is a worldwide musculoskeletal disorder. However, disease-modifying therapies for OA are not available. Here, we aimed to characterize the molecular signatures of OA and to identify novel therapeutic targets and strategies to improve the treatment of OA. METHODS: We collected genome-wide transcriptome data performed on 132 OA and 74 normal human cartilage or synovium tissues from 7 independent datasets. Differential gene expression analysis and functional enrichment were performed to identify genes and pathways that were dysregulated in OA. The computational drug repurposing method was used to uncover drugs that could be repurposed to treat OA. RESULTS: We identified several pathways associated with the development of OA, such as extracellular matrix organization, inflammation, bone development, and ossification. By protein-protein interaction (PPI) network analysis, we prioritized several hub genes, such as JUN, CDKN1A, VEGFA, and FOXO3. Moreover, we repurposed several FDA-approved drugs, such as cardiac glycosides, that could be used in the treatment of OA. CONCLUSIONS: We proposed that the hub genes we identified would play a role in cartilage homeostasis and could be important diagnostic and therapeutic targets. Drugs such as cardiac glycosides provided new possibilities for the treatment of OA.


Computational Biology/methods , Drug Repositioning/methods , Gene Expression/genetics , Osteoarthritis/genetics , Protein Interaction Maps/genetics , Proto-Oncogene Proteins c-jun/physiology , Cardiac Glycosides/therapeutic use , Cyclin-Dependent Kinase Inhibitor p21/physiology , Female , Forkhead Box Protein O3/physiology , Genome-Wide Association Study , Humans , Male , Molecular Targeted Therapy , Osteoarthritis/diagnosis , Osteoarthritis/drug therapy , Transcription Factors , Vascular Endothelial Growth Factor A/physiology
3.
Oncogene ; 39(22): 4436-4449, 2020 05.
Article En | MEDLINE | ID: mdl-32350443

Activator protein (AP)-1 transcription factors are essential elements of the pro-oncogenic functions of transforming growth factor-ß (TGFß)-SMAD signaling. Here we show that in multiple HER2+ and/or EGFR+ breast cancer cell lines these AP-1-dependent tumorigenic properties of TGFß critically rely on epidermal growth factor receptor (EGFR) activation and expression of the ΔN isoform of transcriptional regulator p63. EGFR and ΔNp63 enabled and/or potentiated the activation of a subset of TGFß-inducible invasion/migration-associated genes, e.g., ITGA2, LAMB3, and WNT7A/B, and enhanced the recruitment of SMAD2/3 to these genes. The TGFß- and EGF-induced binding of SMAD2/3 and JUNB to these gene loci was accompanied by p63-SMAD2/3 and p63-JUNB complex formation. p63 and EGFR were also found to strongly potentiate TGFß induction of AP-1 proteins and, in particular, FOS family members. Ectopic overexpression of FOS could counteract the decrease in TGFß-induced gene activation after p63 depletion. p63 is also involved in the transcriptional regulation of heparin binding (HB)-EGF and EGFR genes, thereby establishing a self-amplification loop that facilitates and empowers the pro-invasive functions of TGFß. These cooperative pro-oncogenic functions of EGFR, AP-1, p63, and TGFß were efficiently inhibited by clinically relevant chemical inhibitors. Our findings may, therefore, be of importance for therapy of patients with breast cancers with an activated EGFR-RAS-RAF pathway.


Breast Neoplasms/pathology , Epidermal Growth Factor/physiology , Gene Expression Regulation, Neoplastic , Neoplasm Invasiveness/genetics , Neoplasm Proteins/physiology , Signal Transduction , Transcription Factor AP-1/genetics , Transcription Factors/genetics , Transcription, Genetic , Transforming Growth Factor beta1/physiology , Tumor Suppressor Proteins/genetics , Breast Neoplasms/chemistry , Breast Neoplasms/genetics , Cell Line, Tumor , Cell Movement , ErbB Receptors/physiology , Female , Humans , MAP Kinase Signaling System , Neoplasm Proteins/genetics , Neoplasms, Hormone-Dependent/genetics , Neoplasms, Hormone-Dependent/pathology , Phosphorylation , Protein Kinase Inhibitors/pharmacology , Protein Processing, Post-Translational/drug effects , Proto-Oncogene Proteins c-fos/physiology , Proto-Oncogene Proteins c-jun/physiology , Receptor, ErbB-2/physiology , Receptor, Transforming Growth Factor-beta Type I/physiology , Smad Proteins/physiology
5.
Hepatology ; 72(2): 626-641, 2020 08.
Article En | MEDLINE | ID: mdl-30053321

Clinical data have provided evidence that schistosomiasis can promote hepatocellular carcinogenesis. c-Jun and STAT3 are critical regulators of liver cancer development and progression. The aim of the present study was to investigate the hepatocellular activation of c-Jun and STAT3 by Schistosoma mansoni infection. Expression and function of c-Jun and STAT3 as well as proliferation and DNA repair were analyzed by western blotting, electrophoretic mobility-shift assay, and immunohistochemistry in liver of S. mansoni-infected hamsters, Huh7 cells, primary hepatocytes, and human liver biopsies. Hepatocellular activation of c-Jun was demonstrated by nuclear translocation of c-Jun, enhanced phosphorylation (Ser73), and AP-1/DNA-binding in response to S. mansoni infection. Nuclear c-Jun staining pattern around lodged eggs without ambient immune reaction, and directionally from granuloma to the central veins, suggested that substances released from schistosome eggs were responsible for the observed effects. In addition, hepatocytes with c-Jun activation show cell activation and DNA double-strand breaks. These findings from the hamster model were confirmed by analyses of human biopsies from patients with schistosomiasis. Cell culture experiments finally demonstrated that activation of c-Jun and STAT3 as well as DNA repair were induced by an extract from schistosome eggs (soluble egg antigens) and culture supernatants of live schistosome egg (egg-conditioned medium), and in particular by IPSE/alpha-1, the major component secreted by live schistosome eggs. The permanent activation of hepatocellular carcinoma-associated proto-oncogenes such as c-Jun and associated transcription factors including STAT3 by substances released from tissue-trapped schistosome eggs may be important factors contributing to the development of liver cancer in S. mansoni-infected patients. Therefore, identification and therapeutic targeting of the underlying pathways is a useful strategy to prevent schistosomiasis-associated carcinogenesis.


Antigens, Helminth/physiology , Carcinoma, Hepatocellular , Hepatocytes , Liver Neoplasms , Ovum/immunology , Proto-Oncogene Proteins c-jun/physiology , STAT3 Transcription Factor/physiology , Schistosoma mansoni/immunology , Animals , Antigens, Helminth/metabolism , Carcinoma, Hepatocellular/genetics , Cricetinae , Female , Humans , Liver Neoplasms/genetics , Ovum/metabolism
6.
J Physiol Biochem ; 75(4): 433-441, 2019 Nov.
Article En | MEDLINE | ID: mdl-31376136

Ischemia and hypoxia are common pathophysiological characteristics in cardiovascular diseases. c-Jun expression could be induced by extra- or intracellular stimuli and plays a pivotal role in regulating cell survival in response to the stress. However, previous studies of c-Jun in cell proliferation and apoptosis showed conflicting results. In the present study, we demonstrated that the expression of c-Jun was induced by hypoxia in H9c2 cells. Loss of function of c-Jun was investigated by CCK-8, LDH, and TUNEL assays in low oxygen (1% O2) conditions. We revealed that c-Jun could promote cell survival and inhibit cell apoptosis under hypoxia. Knockdown of c-Jun also promoted the expression of apoptosis-related proteins under hypoxia, such as cleaved caspase-3, cleaved caspase-9, Bax, and Bim. Furthermore, we demonstrated that the knockdown of c-Jun inhibited the PTEN/Akt signaling pathway under hypoxia. Our findings suggested that c-Jun protected H9c2 cells from apoptosis and promoted the survival of H9c2 cells under hypoxia via PTEN/Akt signaling pathway.


Myocytes, Cardiac/metabolism , PTEN Phosphohydrolase/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Proto-Oncogene Proteins c-jun/physiology , Animals , Apoptosis , Cell Hypoxia , Cell Line , Cell Survival , Myocytes, Cardiac/cytology
7.
Cancer Lett ; 448: 155-167, 2019 04 28.
Article En | MEDLINE | ID: mdl-30763715

JunD, a member of the AP-1 family, is essential for cell proliferation in prostate cancer (PCa) cells. We recently demonstrated that JunD knock-down (KD) in PCa cells results in cell cycle arrest in G1-phase concomitant with a decrease in cyclin D1, Ki67, and c-MYC, but an increase in p21 levels. Furthermore, the over-expression of JunD significantly increased proliferation suggesting JunD regulation of genes required for cell cycle progression. Here, employing gene expression profiling, quantitative proteomics, and validation approaches, we demonstrate that JunD KD is associated with distinct gene and protein expression patterns. Comparative integrative analysis by Ingenuity Pathway Analysis (IPA) identified 1) cell cycle control/regulation as the top canonical pathway whose members exhibited a significant decrease in their expression following JunD KD including PRDX3, PEA15, KIF2C, and CDK2, and 2) JunD dependent genes are associated with cell proliferation, with MYC as the critical downstream regulator. Conversely, JunD over-expression induced the expression of the above genes including c-MYC. We conclude that JunD is a crucial regulator of cell cycle progression and inhibiting its target genes may be an effective approach to block prostate carcinogenesis.


Cell Proliferation/physiology , Prostatic Neoplasms/metabolism , Proto-Oncogene Proteins c-jun/physiology , Proto-Oncogene Proteins c-myc/physiology , Cell Cycle/physiology , Cell Line, Tumor , Gene Expression Regulation, Neoplastic , Humans , Male , Microarray Analysis , Signal Transduction/physiology
8.
Mol Biol Rep ; 46(1): 27-39, 2019 Feb.
Article En | MEDLINE | ID: mdl-30515697

Although NFE2L2 transcription factor is considered to make the most significant contribution to the NFE2L2/AP-1-pathway-dependent antioxidants regulation in the human cell, AP-1 has the potential to provide significant backup and even play an equal role in the cell. Considering this, the present study is focused on revealing how JUN, an AP-1 component, and NFE2L2 contribute to regulation of four target genes containing AREs with embedded TREs-SQSTM1, FTH1, HMOX1 and CBR3 and to cellular oxidative status in general in basal conditions and under pro-oxidative influence. NFE2L2 and JUN were down-regulated in HeLa cells using siRNA-mediated knockdown approach. These cells were subsequently exposed to 400 µM hydrogen peroxide in the medium or equal volume of sterile water. They revealed some evidence of both backup functioning and competing between the two factors. Importantly, JUN demonstrated a high level of participation (inc. as a negative regulator) in functioning of the classic NFE2L2 targets and in cellular oxidative status establishment in general. One of the key findings was a dramatic increase in JUN expression following NFE2L2 knockdown in basal conditions. The both AP-1 and NFE2L2 sub-pathways equally determine the outcome of the NFE2L2/AP-1 pathway activation induced by various stimuli, and the outcome is stimulus type- and stimulus-intensity-specific and results from either of the two eventually dominating sub-pathways.


NF-E2-Related Factor 2/physiology , Oxidative Stress/genetics , Proto-Oncogene Proteins c-jun/physiology , Antioxidants/metabolism , Down-Regulation/drug effects , Epithelial Cells/metabolism , Gene Expression Regulation/drug effects , Gene Knockdown Techniques/methods , HeLa Cells , Heme Oxygenase-1/genetics , Humans , Hydrogen Peroxide/metabolism , NF-E2-Related Factor 2/genetics , Oxidation-Reduction , Oxidative Stress/drug effects , Proto-Oncogene Proteins c-jun/genetics , Transcription Factor AP-1/genetics , Transcription Factor AP-1/physiology
9.
Biochem Biophys Res Commun ; 505(2): 593-599, 2018 10 28.
Article En | MEDLINE | ID: mdl-30274780

ATP-binding cassette sub-family G member 2 (ABCG2) confers to the major phenotypes of side population (SP) cells, the cancer stem-like cells. In this study, the SP cells displayed a distinctly higher ABCG2 expression level, sphere formation efficiency (SFE) and growth rate even under hypoxia condition. CXCR4 overexpression by pcDNA-CXCR4 transfection robustly increased ABCG2 expression, and promoted SFE and growth of hypoxic SP cells, while CXCR4 inhibitor AMD3100 could suppress the promotion. Additionally, we found that CXCR4 promoted the expression of c-Jun, a major gene in the oncogenic JNK/c-Jun pathway. Our data on electrophoretic mobility shift assay (EMSA) and chromatin immunoprecipitation (ChIP) assays both showed that c-Jun directly bound with the ABCG2 promoter sequence. Moreover, overexpression of JNK/c-Jun promoted ABCG2 expression, SFE, and growth of hypoxic SP cells and the promotion could be rescued by c-Jun inhibitor SP600125. In conclusion, CXCR4 increases the growth and SFE of breast cancer SP cells under hypoxia through c-Jun-mediated transcriptional activation of ABCG2.


ATP Binding Cassette Transporter, Subfamily G, Member 2/genetics , Breast Neoplasms/genetics , Gene Expression Regulation, Neoplastic , Neoplasm Proteins/genetics , Proto-Oncogene Proteins c-jun/metabolism , Receptors, CXCR4/metabolism , ATP Binding Cassette Transporter, Subfamily G, Member 2/biosynthesis , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Hypoxia , Cell Proliferation , Chemokine CXCL12/metabolism , Female , Humans , JNK Mitogen-Activated Protein Kinases/physiology , MCF-7 Cells , Neoplasm Proteins/biosynthesis , Promoter Regions, Genetic , Proto-Oncogene Proteins c-jun/physiology , Signal Transduction , Spheroids, Cellular , Transcriptional Activation
10.
J Neurosci ; 37(50): 12297-12313, 2017 12 13.
Article En | MEDLINE | ID: mdl-29109239

Schwann cell c-Jun is implicated in adaptive and maladaptive functions in peripheral nerves. In injured nerves, this transcription factor promotes the repair Schwann cell phenotype and regeneration and promotes Schwann-cell-mediated neurotrophic support in models of peripheral neuropathies. However, c-Jun is associated with tumor formation in some systems, potentially suppresses myelin genes, and has been implicated in demyelinating neuropathies. To clarify these issues and to determine how c-Jun levels determine its function, we have generated c-Jun OE/+ and c-Jun OE/OE mice with graded expression of c-Jun in Schwann cells and examined these lines during development, in adulthood, and after injury using RNA sequencing analysis, quantitative electron microscopic morphometry, Western blotting, and functional tests. Schwann cells are remarkably tolerant of elevated c-Jun because the nerves of c-Jun OE/+ mice, in which c-Jun is elevated ∼6-fold, are normal with the exception of modestly reduced myelin thickness. The stronger elevation of c-Jun in c-Jun OE/OE mice is, however, sufficient to induce significant hypomyelination pathology, implicating c-Jun as a potential player in demyelinating neuropathies. The tumor suppressor P19ARF is strongly activated in the nerves of these mice and, even in aged c-Jun OE/OE mice, there is no evidence of tumors. This is consistent with the fact that tumors do not form in injured nerves, although they contain proliferating Schwann cells with strikingly elevated c-Jun. Furthermore, in crushed nerves of c-Jun OE/+ mice, where c-Jun levels are overexpressed sufficiently to accelerate axonal regeneration, myelination and function are restored after injury.SIGNIFICANCE STATEMENT In injured and diseased nerves, the transcription factor c-Jun in Schwann cells is elevated and variously implicated in controlling beneficial or adverse functions, including trophic Schwann cell support for neurons, promotion of regeneration, tumorigenesis, and suppression of myelination. To analyze the functions of c-Jun, we have used transgenic mice with graded elevation of Schwann cell c-Jun. We show that high c-Jun elevation is a potential pathogenic mechanism because it inhibits myelination. Conversely, we did not find a link between c-Jun elevation and tumorigenesis. Modest c-Jun elevation, which is beneficial for regeneration, is well tolerated during Schwann cell development and in the adult and is compatible with restoration of myelination and nerve function after injury.


Gene Dosage , Myelin Sheath/physiology , Nerve Regeneration/physiology , Nerve Tissue Proteins/physiology , Proto-Oncogene Proteins c-jun/physiology , Schwann Cells/metabolism , Animals , Axons/pathology , Cell Nucleus/metabolism , Cell Transformation, Neoplastic , Female , Gene Expression Profiling , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Myelin Proteins/biosynthesis , Myelin Proteins/genetics , Myelin Sheath/ultrastructure , Nerve Crush , Nerve Tissue Proteins/biosynthesis , Nerve Tissue Proteins/genetics , Proto-Oncogene Proteins c-jun/biosynthesis , Proto-Oncogene Proteins c-jun/genetics , RNA, Messenger/biosynthesis , Recovery of Function , Sciatic Nerve/injuries , Sciatic Nerve/pathology
11.
Leukemia ; 31(10): 2235-2243, 2017 10.
Article En | MEDLINE | ID: mdl-28260789

Human T-cell leukemia virus type 1 (HTLV-1) basic-leucine zipper (bZIP) factor (HBZ) is a key player in proliferation and transformation of HTLV-1-infected cells, thus contributing to adult T-cell leukemia (ATL) development. HBZ deregulates gene expression within the host cell by interacting with several cellular partners. Through its C-terminal ZIP domain, HBZ is able to contact and activate JunD, a transcription factor of the AP-1 family. JunD mRNA is intronless but can generate two protein isoforms by alternative translation initiation: JunD full-length and Δ JunD, an N-terminal truncated form unresponsive to the tumor suppressor menin. Using various cell lines and primary T-lymphocytes, we show that after serum deprivation HBZ induces the expression of Δ JunD isoform. We demonstrate that, unlike JunD, Δ JunD induces proliferation and transformation of cells. To decipher the mechanisms for Δ JunD production, we looked into the translational machinery and observed that HBZ induces nuclear retention of RPS25 mRNA and loss of RPS25 protein expression, a component of the small ribosomal subunit. Therefore, HBZ bypasses translational control of JunD uORF and favors the expression of Δ JunD. In conclusion, we provide strong evidences that HBZ induces Δ JunD expression through alteration of the cellular translational machinery and that the truncated isoform Δ JunD has a central role in the oncogenic process leading to ATL.


Basic-Leucine Zipper Transcription Factors/physiology , Cell Transformation, Viral/genetics , Gene Expression Regulation, Leukemic/genetics , Gene Expression Regulation, Viral/genetics , Protein Biosynthesis/genetics , Proto-Oncogene Proteins c-jun/physiology , Retroviridae Proteins/physiology , Ribosomal Proteins/antagonists & inhibitors , Biological Transport , Cell Line , Cell Nucleus/metabolism , Culture Media, Serum-Free , HEK293 Cells , HTLV-I Infections/blood , Humans , Protein Isoforms/genetics , Protein Isoforms/physiology , Proto-Oncogene Proteins/physiology , Proto-Oncogene Proteins c-jun/genetics , RNA, Messenger/metabolism , Ribosomal Proteins/genetics , Ribosomes/metabolism , T-Lymphocytes/pathology , T-Lymphocytes/virology , Transfection
12.
J Exp Med ; 214(5): 1313-1331, 2017 05 01.
Article En | MEDLINE | ID: mdl-28356392

Inappropriate IL-17 responses are implicated in chronic tissue inflammation. IL-23 contributes to Trypanosoma cruzi-specific IL-17 production, but the molecular mechanisms underlying regulation of the IL-23-IL-17 axis during T. cruzi infection are poorly understood. Here, we demonstrate a novel function of BATF2 as a negative regulator of Il23a in innate immune cells. IL-17, but not IFN-γ, was more highly produced by CD4+ T cells from spleens and livers of T. cruzi-infected Batf2-/- mice than by those of wild-type mice. In this context, Batf2-/- mice showed severe multiorgan pathology despite reduced parasite burden. T. cruzi-induced IL-23 production was increased in Batf2-/- innate immune cells. The T. cruzi-induced enhanced Th17 response was abrogated in Batf2-/-Il23a-/- mice. The interaction of BATF2 with c-JUN prevented c-JUN-ATF-2 complex formation, inhibiting Il23a expression. These results demonstrate that IFN-γ-inducible BATF2 in innate immune cells controls Th17-mediated immunopathology by suppressing IL-23 production during T. cruzi infection.


Basic-Leucine Zipper Transcription Factors/physiology , Chagas Disease/immunology , Interleukin-23 Subunit p19/metabolism , Th17 Cells/physiology , Trypanosoma cruzi/immunology , Animals , Chagas Disease/pathology , Female , Immunity, Innate/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Proto-Oncogene Proteins c-jun/physiology , Th17 Cells/immunology
13.
Leukemia ; 31(5): 1196-1205, 2017 05.
Article En | MEDLINE | ID: mdl-27840425

The transcription factor JUN is frequently overexpressed in multiple genetic subtypes of acute myeloid leukemia (AML); however, the functional role of JUN in AML is not well defined. Here we report that short hairpin RNA (shRNA)-mediated inhibition of JUN decreases AML cell survival and propagation in vivo. By performing RNA sequencing analysis, we discovered that JUN inhibition reduces the transcriptional output of the unfolded protein response (UPR), an intracellular signaling transduction network activated by endoplasmic reticulum (ER) stress. Specifically, we found that JUN is activated by MEK signaling in response to ER stress, and that JUN binds to the promoters of several key UPR effectors, such as XBP1 and ATF4, to activate their transcription and allow AML cells to properly negotiate ER stress. In addition, we observed that shRNA-mediated inhibition of XBP1 or ATF4 induces AML cell apoptosis and significantly extends disease latency in vivo tying the reduced survival mediated by JUN inhibition to the loss of pro-survival UPR signaling. These data uncover a previously unrecognized role of JUN as a regulator of the UPR as well as provide key new insights into the how ER stress responses contribute to AML and identify JUN and the UPR as promising therapeutic targets in this disease.


Leukemia, Myeloid, Acute/metabolism , Proto-Oncogene Proteins c-jun/physiology , Unfolded Protein Response , Animals , Apoptosis , Cell Proliferation , Cell Survival , Endoplasmic Reticulum Stress , Humans , Leukemia, Myeloid, Acute/pathology , Mice , Proto-Oncogene Proteins c-jun/antagonists & inhibitors , RNA, Small Interfering/pharmacology , Tumor Cells, Cultured
14.
J Clin Invest ; 126(1): 151-68, 2016 Jan.
Article En | MEDLINE | ID: mdl-26619121

Satellite cells are a stem cell population within adult muscle and are responsible for myofiber regeneration upon injury. Satellite cell dysfunction has been shown to underlie the loss of skeletal muscle mass in many acquired and genetic muscle disorders. The transcription factor paired box-protein-7 (PAX7) is indispensable for supplementing the reservoir of satellite cells and driving regeneration in normal and diseased muscle. TNF receptor-associated factor 6 (TRAF6) is an adaptor protein and an E3 ubiquitin ligase that mediates the activation of multiple cell signaling pathways in a context-dependent manner. Here, we demonstrated that TRAF6-mediated signaling is critical for homeostasis of satellite cells and their function during regenerative myogenesis. Selective deletion of Traf6 in satellite cells of adult mice led to profound muscle regeneration defects and dramatically reduced levels of PAX7 and late myogenesis markers. TRAF6 was required for the activation of MAPKs ERK1/2 and JNK1/2, which in turn activated the transcription factor c-JUN, which binds the Pax7 promoter and augments Pax7 expression. Moreover, TRAF6/c-JUN signaling repressed the levels of the microRNAs miR-1 and miR-206, which promote differentiation, to maintain PAX7 levels in satellite cells. We also determined that satellite cell-specific deletion of Traf6 exaggerates the dystrophic phenotype in the mdx (a mouse model of Duchenne muscular dystrophy) mouse by blunting the regeneration of injured myofibers. Collectively, our study reveals an essential role for TRAF6 in satellite stem cell function.


Cell Self Renewal , Muscle Development , Regeneration/physiology , Satellite Cells, Skeletal Muscle/physiology , Stem Cells/physiology , TNF Receptor-Associated Factor 6/physiology , Animals , MAP Kinase Signaling System , Mice , MicroRNAs/analysis , PAX7 Transcription Factor/analysis , PAX7 Transcription Factor/physiology , Proto-Oncogene Proteins c-jun/physiology
15.
Am J Respir Crit Care Med ; 193(2): 143-53, 2016 Jan 15.
Article En | MEDLINE | ID: mdl-26426522

RATIONALE: Corticosteroid resistance is a major barrier to the effective treatment of chronic obstructive pulmonary disease (COPD). Several molecular mechanisms have been proposed, such as activations of the phosphoinositide-3-kinase/Akt pathway and p38 mitogen-activated protein kinase. However, the mechanism for corticosteroid resistance is still not fully elucidated. OBJECTIVES: To investigate the role of mammalian target of rapamycin (mTOR) in corticosteroid sensitivity in COPD. METHODS: The corticosteroid sensitivity of peripheral blood mononuclear cells collected from patients with COPD, smokers, and nonsmoking control subjects, or of human monocytic U937 cells exposed to cigarette smoke extract (CSE), was quantified as the dexamethasone concentration required to achieve 30% inhibition of tumor necrosis factor-α-induced CXCL8 production in the presence or absence of the mTOR inhibitor rapamycin. mTOR activity was determined as the phosphorylation of p70 S6 kinase, using Western blotting. MEASUREMENTS AND MAIN RESULTS: mTOR activity was increased in peripheral blood mononuclear cells from patients with COPD, and treatment with rapamycin inhibited this as well as restoring corticosteroid sensitivity. In U937 cells, CSE stimulated mTOR activity and c-Jun expression, but pretreatment with rapamycin inhibited both and also reversed CSE-induced corticosteroid insensitivity. CONCLUSIONS: mTOR inhibition by rapamycin restores corticosteroid sensitivity via inhibition of c-Jun expression, and thus mTOR is a potential novel therapeutic target for COPD.


Adrenal Cortex Hormones/pharmacology , Drug Resistance/drug effects , Oxidative Stress/drug effects , Proto-Oncogene Proteins c-jun/drug effects , Pulmonary Disease, Chronic Obstructive/drug therapy , Sirolimus/pharmacology , TOR Serine-Threonine Kinases/drug effects , Adrenal Cortex Hormones/therapeutic use , Aged , Drug Resistance/immunology , Female , Histone Deacetylase 2/drug effects , Histone Deacetylase 2/physiology , Humans , Immunosuppressive Agents/immunology , Immunosuppressive Agents/pharmacology , Immunosuppressive Agents/therapeutic use , Male , Middle Aged , Oxidative Stress/physiology , Phosphatidylinositol 3-Kinases/drug effects , Phosphatidylinositol 3-Kinases/physiology , Proto-Oncogene Proteins c-akt/drug effects , Proto-Oncogene Proteins c-akt/physiology , Proto-Oncogene Proteins c-jun/physiology , Pulmonary Disease, Chronic Obstructive/immunology , Pulmonary Disease, Chronic Obstructive/physiopathology , Sirolimus/immunology , Sirolimus/therapeutic use , Smoking/adverse effects , Smoking/physiopathology , TOR Serine-Threonine Kinases/physiology , U937 Cells/drug effects , p38 Mitogen-Activated Protein Kinases/drug effects , p38 Mitogen-Activated Protein Kinases/physiology
16.
Oncogene ; 35(31): 4080-90, 2016 08 04.
Article En | MEDLINE | ID: mdl-26686085

Emerging evidence from The Cancer Genome Atlas has revealed that nuclear factor κB2 (nfκb2) gene encoding p100 is genetically deleted or mutated in human cancers, implicating NFκB2 as a potential tumor suppressor. However, the molecular mechanism underlying the antitumorigenic action of p100 remains poorly understood. Here we report that p100 inhibits cancer cell anchorage-independent growth, a hallmark of cellular malignancy, by stabilizing the tumor-suppressor phosphatase and tensin homolog (PTEN) mRNA via a mechanism that is independent of p100's inhibitory role in NFκB activation. We further demonstrate that the regulatory effect of p100 on PTEN expression is mediated by its downregulation of miR-494 as a result of the inactivation of extracellular signal-regulated kinase 2 (ERK2), in turn leading to inhibition of c-Jun/activator protein-1-dependent transcriptional activity. Furthermore, we identify that p100 specifically interacts with non-phosphorylated ERK2 and prevents ERK2 phosphorylation and nuclear translocation. Moreover, the death domain at C-terminal of p100 is identified as being crucial and sufficient for its interaction with ERK2. Taken together, our findings provide novel mechanistic insights into the understanding of the tumor-suppressive role for NFκB2 p100.


MicroRNAs/antagonists & inhibitors , Mitogen-Activated Protein Kinase 1/physiology , NF-kappa B p52 Subunit/physiology , PTEN Phosphohydrolase/genetics , RNA Stability , Tumor Suppressor Proteins/physiology , Active Transport, Cell Nucleus , HCT116 Cells , Humans , MicroRNAs/physiology , PTEN Phosphohydrolase/antagonists & inhibitors , Phosphorylation , Proto-Oncogene Proteins c-jun/physiology , Signal Transduction
17.
Eur Heart J ; 36(48): 3392-403, 2015 Dec 21.
Article En | MEDLINE | ID: mdl-26543043

Western societies are aging due to an increasing life span, decreased birth rates, and improving social and health conditions. On the other hand, the prevalence of cardiovascular (CV) and cerebrovascular (CBV) diseases rises with age. Thus, in view of the ongoing aging pandemic, it is appropriate to better understand the molecular pathways of aging as well as age-associated CV and CBV diseases. Oxidative stress contributes to aging of organs and the whole body by an accumulation of reactive oxygen species promoting oxidative damage. Indeed, increased oxidative stress produced in the mitochondria and cytosol of heart and brain is a common denominator to almost all CV and CBV diseases. The mitochondrial adaptor protein p66(Shc) and the family of deacetylase enzymes, the sirtuins, regulate the aging process, determine lifespan of many species and are involved in CV diseases. GDF11, a member of TGFß superfamily with homology to myostatin also retards the aging process via yet unknown mechanisms. Recent evidence points towards a promising role of this novel 'rejuvenation' factor in reducing age-related heart disease. Finally, telomere length is also involved in aging and the development of age-related CV dysfunction. This review focuses on the latest scientific advances in understanding age-related changes of the CV and CBV system, as well as delineating potential novel therapeutic targets derived from aging research for CV and CBV diseases.


Aging/physiology , Cardiovascular Diseases/etiology , Aging/genetics , Animals , Bone Morphogenetic Proteins/physiology , Caloric Restriction , Free Radicals/metabolism , Growth Differentiation Factors/physiology , Humans , Longevity/physiology , Mitochondria/physiology , Models, Biological , Oxidative Stress/physiology , Proto-Oncogene Proteins c-jun/physiology , Risk Reduction Behavior , Shc Signaling Adaptor Proteins/physiology , Sirtuins/physiology , Src Homology 2 Domain-Containing, Transforming Protein 1 , Telomere Shortening/physiology , Vascular Stiffness/physiology
18.
Leukemia ; 29(11): 2162-72, 2015 Nov.
Article En | MEDLINE | ID: mdl-25987255

Anaplastic lymphoma kinase-positive (ALK+) anaplastic large-cell lymphoma (ALCL) is an aggressive T-cell non-Hodgkin lymphoma characterized by the t(2;5), resulting in the overexpression of nucleophosmin (NPM)-ALK, which is known to activate the phosphatidylinositol-3-kinase (PI3K)/AKT/mammalian target of rapamycin (mTOR) pathway, resulting in cell cycle and apoptosis deregulation. ALK+ ALCL is also characterized by strong activator protein-1 (AP-1) activity and overexpression of two AP-1 transcription factors, CJUN and JUNB. Here, we hypothesized that a biologic link between AP-1 and AKT kinase may exist, thus contributing to ALCL oncogenesis. We show that JUNB and CJUN bind directly to the AKT1 promoter, inducing AKT1 transcription in ALK+ ALCL. Knockdown of JUNB and CJUN in ALK+ ALCL cell lines downregulated AKT1 mRNA and promoter activity and was associated with lower AKT1 protein expression and activation. We provide evidence that this is a transcriptional control mechanism shared by other cell types even though it may operate in a way that is cell context-specific. In addition, STAT3 (signal transducer and activator of transcription 3)-induced control of AKT1 transcription was functional in ALK+ ALCL and blocking of STAT3 and AP-1 signaling synergistically affected cell proliferation and colony formation. Our findings uncover a novel transcriptional crosstalk mechanism that links AP-1 and AKT kinase, which coordinate uncontrolled cell proliferation and survival in ALK+ ALCL.


Gene Expression Regulation, Neoplastic , Lymphoma, Large-Cell, Anaplastic/genetics , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-jun/physiology , Receptor Protein-Tyrosine Kinases/analysis , Transcription Factors/physiology , Anaplastic Lymphoma Kinase , Cell Line, Tumor , Humans , Promoter Regions, Genetic , STAT3 Transcription Factor/physiology , Transcription Factor AP-1/physiology
19.
Leuk Res ; 39(5): 544-52, 2015 May.
Article En | MEDLINE | ID: mdl-25828744

There are few studies on the correlation between heme oxygenase-1 (HO-1) and acute myeloid leukemia (AML). We found that HO-1 was aberrantly overexpressed in the majority of AML patients, especially in patients with acute monocytic leukemia (M5) and leukocytosis, and inhibited the apoptosis of HL-60 and U937 cells. Moreover, silencing HO-1 prolonged the survival of xenograft mouse models. Further studies demonstrated that HO-1 suppressed the apoptosis of AML cells through activating the JNK/c-JUN signaling pathway. These data indicate a molecular role of HO-1 in inhibiting cell apoptosis, allowing it to be a potential target for treating AML.


Apoptosis/genetics , Heme Oxygenase-1/physiology , Leukemia, Myeloid, Acute/genetics , MAP Kinase Signaling System/physiology , Proto-Oncogene Proteins c-jun/physiology , Adult , Aged , Aged, 80 and over , Animals , Down-Regulation , Female , HL-60 Cells , Humans , Male , Mice , Mice, SCID , Middle Aged , U937 Cells , Young Adult
20.
PLoS One ; 9(5): e96610, 2014.
Article En | MEDLINE | ID: mdl-24796531

Transcription factor c-Jun plays a key role in controlling epithelium cell proliferation, apoptosis and differentiation. However, molecular mechanism and biological functions of c-Jun in squamous differentiation and the progression of esophageal squamous cell carcinoma (ESCC) remain elusive. In this study, we found that c-Jun bound directly to the promoter region, and activated the transcription of differentiation-associated genes including cystatin A, involucrin and SPRR3 in vivo. Ectopic expression of c-Jun enhanced SPRR3 transactivation in KYSE450 cells. Conversely, TAM67, a dominant negative mutant of c-Jun, inhibited SPRR3 transactivation. c-Jun increased expression of SPPR3 mainly via a PKC/JNK pathway in response to TPA in KYSE450 cells. Furthermore, c-Jun was remarkably reduced in esophageal cancer. Interestingly, cystatin A, involucrin and SPRR3 were significantly downregulated as well, and associated with differentiation grade. Expression of c-Jun was correlated with the expression of these genes in normal epithelium and ESCC. Importantly, the expression of these genes was remarkably decreased during the malignant transformation from normal epithelium to low-grade intraepithelial neoplasia (LGIN) or high-grade intraepithelial neoplasia (HGIN). The expression of cystatin A and involucrin was significantly reduced from LGIN to HGIN. These results suggest c-Jun was involved in the regulation of differentiation-associated genes in ESCC. These genes might serve as the potential markers in distinguishing normal epithelium from esophageal squamous intraepithelial neoplasia.


Carcinoma, Squamous Cell/genetics , Carcinoma, Squamous Cell/metabolism , Esophageal Neoplasms/genetics , Esophageal Neoplasms/metabolism , Gene Expression Regulation, Neoplastic , Genes, jun , Proto-Oncogene Proteins c-jun/genetics , Proto-Oncogene Proteins c-jun/physiology , Aged , Carcinoma in Situ/metabolism , Cell Differentiation , Cell Line , Cornified Envelope Proline-Rich Proteins/metabolism , Cystatin A/metabolism , Disease Progression , Epithelium/metabolism , Esophageal Squamous Cell Carcinoma , Female , Genes, Dominant , Humans , Male , Middle Aged , Mutation , Oligonucleotide Array Sequence Analysis , Protein Kinase C/metabolism , Protein Precursors/metabolism , Transcriptional Activation
...