Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 186
Filter
1.
Biomed Pharmacother ; 106: 532-542, 2018 Oct.
Article in English | MEDLINE | ID: mdl-29990841

ABSTRACT

The phenotype of multidrug resistance (MDR) is one of the main causes of chemotherapy failure. Our study investigated the effect of C-phycocyanin (C-PC) in three human erythroleukemia cell lines with or without the MDR phenotype: K562 (non-MDR; no overexpression of drug efflux proteins), K562-Lucena (MDR; overexpression of ATP-binding cassette, sub-family B/ABCB1), and FEPS (MDR; overexpression of ABCB1 and ATP-binding cassette, sub-family C/ABCC1). Using the 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay, we showed that 20 and 200 µg/mL C-PC decreased K562 viable cells after 24 h and 200 µg/mL C-PC decreased K562-Lucena cell proliferation after 48 h. C-PC did not decrease viable cells of FEPS cells. On the other hand, the MTT assay showed that exposure of 2, 20, and 200 µg/mL C-PC for 24 or 48 h was not cytotoxic to peritoneal macrophages. At 72 h, the trypan blue exclusion assay showed that 20 µg/mL C-PC decreased K562 and K562-Lucena cell proliferation and in FEPS cells, only 200 µg/mL C-PC decreased proliferation. In addition, protein-protein docking showed differences in energy and binding sites of ABCB1 and ABCC1 for C-PC, and these results were confirmed by the efflux protein activity assay. Only ABCC1 activity was altered in the presence of C-PC and FEPS cells showed lower C-PC accumulation, suggesting C-PC extrusion by ABCC1, conferring C-PC resistance. In combination with chemotherapy (vincristine [VCR] and daunorubicin [DNR]), the sensitivity of K562-Lucena cells for C-PC + VCR did not increase, whereas FEPS cell sensitivity for C-PC + DNR was increased. In molecular docking experiments, the estimated free energies of binding for C-PC associated with chemotherapy were similar (VCR: -6.9 kcal/mol and DNR: -7.2 kcal/mol) and these drugs were located within the C-PC cavity. However, C-PC exhibited specificity for tumor cells and K562 cells were more sensitive than K562-Lucena cells, followed by FEPS cells. Thus, C-PC is a possible chemotherapeutic agent for cells with the MDR phenotype, both alone in K562-Lucena cells (resistance due to ABCB1), or in combination with other drugs for cells similar to FEPS (resistance due to ABCC1). Moreover, C-PC did not damage healthy cells (peritoneal macrophages of Mus musculus).


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Daunorubicin/pharmacology , Drug Resistance, Multiple/drug effects , Drug Resistance, Neoplasm/drug effects , Leukemia, Erythroblastic, Acute/drug therapy , Multidrug Resistance-Associated Proteins/metabolism , Phycocyanin/pharmacology , Vincristine/pharmacology , ATP Binding Cassette Transporter, Subfamily B/genetics , ATP Binding Cassette Transporter, Subfamily B/metabolism , Animals , Antineoplastic Combined Chemotherapy Protocols/toxicity , Binding Sites , Cell Proliferation/drug effects , Cell Survival/drug effects , Dose-Response Relationship, Drug , Drug Resistance, Multiple/genetics , Drug Resistance, Neoplasm/genetics , Female , Gene Expression Regulation, Neoplastic , Humans , K562 Cells , Leukemia, Erythroblastic, Acute/genetics , Leukemia, Erythroblastic, Acute/metabolism , Leukemia, Erythroblastic, Acute/pathology , Macrophages, Peritoneal/drug effects , Macrophages, Peritoneal/pathology , Mice , Molecular Docking Simulation , Multidrug Resistance-Associated Proteins/genetics , Phycocyanin/metabolism , Phycocyanin/toxicity , Protein Binding , Protein Conformation , Time Factors
2.
Leukemia ; 31(7): 1570-1581, 2017 07.
Article in English | MEDLINE | ID: mdl-27890927

ABSTRACT

Despite therapeutic advances, multiple myeloma (MM) remains an incurable disease, predominantly because of the development of drug resistance. The activator protein-1 (AP-1) transcription factor family has been implicated in a multitude of physiologic processes and tumorigenesis; however, its role in MM is largely unknown. Here we demonstrate specific and rapid induction of the AP-1 family member JunB in MM cells when co-cultured with bone marrow stromal cells. Supporting a functional key role of JunB in MM pathogenesis, knockdown of JUNB significantly inhibited in vitro MM cell proliferation and survival. Consistently, induced silencing of JUNB markedly decreased tumor growth in a murine MM model of the microenvironment. Subsequent gene expression profiling revealed a role for genes associated with apoptosis, DNA replication and metabolism in driving the JunB-mediated phenotype in MM cells. Importantly, knockdown of JUNB restored the response to dexamethasone in dexamethasone-resistant MM cells. Moreover, 4-hydroxytamoxifen-induced activation of a JunB-ER fusion protein protected dexamethasone-sensitive MM cells against dexamethasone- and bortezomib-induced cytotoxicity. In summary, our results demonstrate for the first time a specific role for AP-1/JunB in MM cell proliferation, survival and drug resistance, thereby strongly supporting that this transcription factor is a promising new therapeutic target in MM.


Subject(s)
Bone Marrow/pathology , Multiple Myeloma/pathology , Transcription Factors/physiology , Tumor Microenvironment , Animals , Bortezomib/pharmacology , Cell Proliferation , Dexamethasone/pharmacology , Drug Resistance, Neoplasm , Female , Humans , Mice , Multiple Myeloma/drug therapy , Multiple Myeloma/mortality , NF-kappa B/physiology
3.
Cell Death Dis ; 7(11): e2461, 2016 11 10.
Article in English | MEDLINE | ID: mdl-27831556

ABSTRACT

CD44, a large family of transmembrane glycoproteins, plays decisive roles in physiological and pathological conditions. CD44 isoforms are involved in several signaling pathways essential for life such as growth factor-induced signaling by EGF, HGF or VEGF. CD44 is also the main hyaluronan (HA) receptor and as such is involved in HA-dependent processes. To allow a genetic dissection of CD44 functions in homeostasis and disease, we generated a Cd44 floxed allele allowing tissue- and time-specific inactivation of all CD44 isoforms in vivo. As a proof of principle, we inactivated Cd44 in the skin epidermis using the K14Cre allele. Although the skin of such Cd44Δker mutants appeared morphologically normal, epidermal stiffness was reduced, wound healing delayed and TPA induced epidermal thickening decreased. These phenotypes might be caused by cell autonomous defects in differentiation and HA production as well as impaired adhesion and migration on HA by Cd44Δker keratinocytes. These findings support the usefulness of the conditional Cd44 allele in unraveling essential physiological and pathological functions of CD44 isoforms.


Subject(s)
Epidermis/metabolism , Gene Deletion , Hyaluronan Receptors/metabolism , Keratinocytes/metabolism , Stress, Mechanical , Animals , Cell Adhesion/drug effects , Cell Differentiation/drug effects , Cell Movement/drug effects , Cell Proliferation/drug effects , Homeostasis/drug effects , Hyaluronic Acid/pharmacology , Keratinocytes/drug effects , Keratins/metabolism , Mice, Knockout , Organ Specificity/drug effects , Skin/metabolism , Wound Healing/drug effects
4.
Cell Death Differ ; 23(4): 576-82, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26470729

ABSTRACT

Hepatocellular carcinoma (HCC) develops as a consequence of chronic inflammatory liver diseases such as chronic hepatitis B virus (HBV) infection. The transcription factor c-Jun/activator protein 1 (AP-1) is strongly expressed in response to inflammatory stimuli, promotes hepatocyte survival during acute hepatitis and acts as an oncogene during chemically induced liver carcinogenesis in mice. Here, we therefore aimed to characterize the functions of c-Jun during HBV-related liver tumorigenesis. To this end, transgenic mice expressing all HBV envelope proteins (HBV(+)), an established model of HBV-related HCC, were crossed with knockout mice lacking c-Jun specifically in hepatocytes and tumorigenesis was analyzed. Hepatic expression of c-Jun was strongly induced at several time points during tumorigenesis in HBV(+) mice, whereas expression of other AP-1 components remained unchanged. Importantly, formation of premalignant foci and tumors was strongly reduced in HBV(+) mice lacking c-Jun. This phenotype correlated with impaired hepatocyte proliferation and increased expression of the cell cycle inhibitor p21, whereas hepatocyte survival was not affected. Progression and prognosis of HBV-related HCC correlates with the expression of the cytokine osteopontin (Opn), an established AP-1 target gene. Opn expression was strongly reduced in HBV(+) livers and primary mouse hepatocytes lacking c-Jun, demonstrating that c-Jun regulates hepatic Opn expression in a cell-autonomous manner. These findings indicate that c-Jun has important functions during HBV-associated tumorigenesis by promoting hepatocyte proliferation as well as progression of dysplasia. Therefore, targeting c-Jun may be a useful strategy to prevent hepatitis-associated tumorigenesis.


Subject(s)
Carcinoma, Hepatocellular/metabolism , Cell Transformation, Viral , Hepatitis B virus/metabolism , Liver Neoplasms/metabolism , Liver/metabolism , Proto-Oncogene Proteins c-jun/metabolism , Transcription Factor AP-1/metabolism , Animals , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/pathology , Carcinoma, Hepatocellular/virology , Cell Proliferation/genetics , Hepatitis B virus/genetics , Hepatocytes/metabolism , Hepatocytes/pathology , Hepatocytes/virology , Liver/pathology , Liver/virology , Liver Neoplasms/genetics , Liver Neoplasms/pathology , Liver Neoplasms/virology , Mice , Mice, Transgenic , Proto-Oncogene Proteins c-jun/genetics , Transcription Factor AP-1/genetics
5.
Cell Death Differ ; 22(2): 336-50, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25301070

ABSTRACT

Epithelial-to-mesenchymal transition (EMT) is essential for embryonic morphogenesis and wound healing and critical for tumour cell invasion and dissemination. The AP-1 transcription factor Fra-1 has been implicated in tumorigenesis and in tumour-associated EMT in human breast cancer. We observed a significant inverse correlation between Fra-1 mRNA expression and distant-metastasis-free survival in a large cohort of breast cancer patients derived from multiple array data sets. This unique correlation among Fos genes prompted us to assess the evolutionary conservation between Fra-1 functions in EMT of human and mouse cells. Ectopic expression of Fra-1 in fully polarized, non-tumourigenic, mouse mammary epithelial EpH4 cells induced a mesenchymal phenotype, characterized by a loss of epithelial and gain of mesenchymal markers. Proliferation, motility and invasiveness were also increased in the resulting EpFra1 cells, and the cells were tumourigenic and efficiently colonized the lung upon transplantation. Molecular analyses revealed increased expression of Tgfß1 and the EMT-inducing transcription factors Zeb1, Zeb2 and Slug. Mechanistically, Fra-1 binds to the tgfb1 and zeb2 promoters and to an evolutionarily conserved region in the first intron of zeb1. Furthermore, increased activity of a zeb2 promoter reporter was detected in EpFra1 cells and shown to depend on AP-1-binding sites. Inhibiting TGFß signalling in EpFra1 cells moderately increased the expression of epithelial markers, whereas silencing of zeb1 or zeb2 restored the epithelial phenotype and decreased migration in vitro and tumorigenesis in vivo. Thus Fra-1 induces changes in the expression of genes encoding EMT-related transcription factors leading to the acquisition of mesenchymal, invasive and tumorigenic capacities by epithelial cells. This study defines a novel function of Fra-1/AP-1 in modulating tgfb1, zeb1 and zeb2 expression through direct binding to genomic regulatory regions, which establishes a basis for future in vivo genetic manipulations and preclinical studies using mouse models.


Subject(s)
Epithelial Cells/metabolism , Epithelial-Mesenchymal Transition , Homeodomain Proteins/metabolism , Proto-Oncogene Proteins c-fos/metabolism , Repressor Proteins/metabolism , Transcription Factor AP-1/metabolism , Transcription Factors/metabolism , Transforming Growth Factor beta/metabolism , Animals , Breast Neoplasms/pathology , Cadherins/genetics , Cell Line, Tumor , Disease Models, Animal , Female , Homeodomain Proteins/genetics , Humans , Mammary Glands, Human/cytology , Mice , Promoter Regions, Genetic , Proto-Oncogene Proteins c-fos/genetics , Repressor Proteins/genetics , Transcription Factor AP-1/genetics , Transcription Factors/genetics , Zinc Finger E-box Binding Homeobox 2 , Zinc Finger E-box-Binding Homeobox 1
6.
Cell Death Differ ; 22(4): 574-82, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25526087

ABSTRACT

Prostate cancer is a frequent cause of male death in the Western world. Relatively few genetic alterations have been identified, likely owing to disease heterogeneity. Here, we show that the transcription factor JUNB/AP-1 limits prostate cancer progression. JUNB expression is increased in low-grade prostate cancer compared with normal human prostate, but downregulated in high-grade samples and further decreased in all metastatic samples. To model the hypothesis that this downregulation is functionally significant, we genetically inactivated Junb in the prostate epithelium of mice. When combined with Pten (phosphatase and tensin homologue) loss, double-mutant mice were prone to invasive cancer development. Importantly, invasive tumours also developed when Junb and Pten were inactivated in a small cell population of the adult anterior prostate by topical Cre recombinase delivery. The resulting tumours displayed strong histological similarity with human prostate cancer. Loss of JunB expression led to increased proliferation and decreased senescence, likely owing to decreased p16(Ink4a) and p21(CIP1) in epithelial cells. Furthermore, the tumour stroma was altered with increased osteopontin and S100 calcium-binding protein A8/9 expression, which correlated with poor prognoses in patients. These data demonstrate that JUNB/AP-1 cooperates with PTEN signalling as barriers to invasive prostate cancer, whose concomitant genetic or epigenetic suppression induce malignant progression.


Subject(s)
Prostatic Neoplasms/pathology , Transcription Factors/metabolism , Aging , Animals , Calgranulin A/metabolism , Calgranulin B/metabolism , Cyclin-Dependent Kinase Inhibitor p16/metabolism , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Disease Progression , Down-Regulation , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Neoplasm Invasiveness , Osteopontin/metabolism , PTEN Phosphohydrolase/genetics , PTEN Phosphohydrolase/metabolism , Prostatic Neoplasms/genetics , Prostatic Neoplasms/metabolism , RNA, Messenger/metabolism , Signal Transduction , Transcription Factors/genetics
7.
Cell Death Differ ; 21(4): 655-64, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24464219

ABSTRACT

Adipocyte cell number is a crucial factor for controlling of body weight and metabolic function. The regulation of adipocyte numbers in the adult organism is not fully understood but is considered to depend on the homeostasis of cell differentiation and apoptosis. Herein, we show that targeted deletion of the activator protein (AP-1)-related transcription factor Fra-2 in adipocytes in vivo (Fra-2(Δadip) mice) induces a high-turnover phenotype with increased differentiation and apoptosis of adipocytes, leading to a decrease in body weight and fat pad mass. Importantly, adipocyte cell numbers were significantly reduced in Fra-2(Δadip) mice. At the molecular level, Fra-2 directly binds to the PPARγ2 promoter and represses PPARγ2 expression. Deletion of Fra-2 leads to increased PPARγ2 expression and adipocyte differentiation as well as increased adipocyte apoptosis through upregulation of hypoxia-inducible factors (HIFs). These findings suggest that Fra-2 is an important checkpoint to control adipocyte turnover. Therefore, inhibition of Fra-2 may emerge as a useful strategy to increase adipocyte turnover and to reduce adipocyte numbers and fat mass in the body.


Subject(s)
Adipocytes/metabolism , Cell Differentiation/genetics , Cell Hypoxia/genetics , Fos-Related Antigen-2/metabolism , PPAR gamma/metabolism , Adipocytes/cytology , Animals , Antineoplastic Agents, Hormonal/pharmacology , Apoptosis/drug effects , Basic Helix-Loop-Helix Transcription Factors/antagonists & inhibitors , Basic Helix-Loop-Helix Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/metabolism , Cells, Cultured , Diet, High-Fat , Fos-Related Antigen-2/genetics , Hypoxia-Inducible Factor 1/antagonists & inhibitors , Hypoxia-Inducible Factor 1/genetics , Hypoxia-Inducible Factor 1/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , PPAR gamma/genetics , Promoter Regions, Genetic , Protein Binding , Tamoxifen/pharmacology , Up-Regulation
8.
Oncogene ; 33(19): 2513-9, 2014 May 08.
Article in English | MEDLINE | ID: mdl-23752184

ABSTRACT

NANOG is a key transcription factor for pluripotency in embryonic stem cells. The analysis of NANOG in human cells is confounded by the presence of multiple and highly similar paralogs. In particular, there are three paralogs encoding full-length proteins, namely, NANOG1, NANOG2 and NANOGP8, and at least eight additional paralogs that do not encode full-length NANOG proteins. Here, we have examined NANOG family expression in human embryonic stem cells (hESCs) and in human cancer cell lines using a multi-NANOG PCR that amplifies the three functional paralogs and most of the non-functional ones. As anticipated, we found that hESCs express large amounts of NANOG1 and, interestingly, they also express NANOG2. In contrast, most human cancer cells tested express NANOGP8 and the non-coding paralogs NANOGP4 and NANOGP5. Notably, in some cancer cell lines, the NANOG protein levels produced by NANOGP8 are comparable to those produced by NANOG1 in pluripotent cells. Finally, we show that NANOGP8 is as active as NANOG1 in the reprogramming of human and murine fibroblasts into induced pluripotent stem cells. These results show that cancer-associated NANOGP8 can contribute to promote de-differentiation and/or cellular plasticity.


Subject(s)
Cell Dedifferentiation/physiology , Homeodomain Proteins/metabolism , Neoplasms/metabolism , Animals , Cell Differentiation/physiology , Cell Line, Tumor , Embryonic Stem Cells/metabolism , Gene Knockdown Techniques , Homeodomain Proteins/genetics , Humans , Mice , Mutagenesis, Site-Directed , Nanog Homeobox Protein , Neoplasms/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction
9.
Oncogene ; 31(13): 1723-32, 2012 Mar 29.
Article in English | MEDLINE | ID: mdl-21841823

ABSTRACT

Destruction of insulin-producing pancreatic ß-cells by local autoimmune inflammation is a hallmark of type 1 diabetes. Histochemical analysis of pancreases from non-obese diabetic mice indicated activation of the transcription factor JunB/AP-1 (activator protein-1) after autoimmune infiltration of the islets. In vitro studies demonstrated that the cytokines tumor necrosis factor (TNF)-α and interferon (IFN)-γ induce JunB expression as a protective mechanism against apoptosis in both human and rodent ß-cells. The gene network affected was studied by microarray analysis showing that JunB regulates nearly 20% of the cytokine-modified ß-cell genes, including the transcription factor ATF3. Direct transcriptional induction of ATF3 by JunB is a key event for ß-cell survival after TNF-α+IFN-γ treatment. Moreover, pharmacological upregulation of JunB/ATF3 via increased cAMP protected rodent primary ß-cells and human islet cells against pro-inflammatory mediators. These results were confirmed in genetically modified islets derived from Ubi-JunB transgenic mice. Our findings identify ATF3 as a novel downstream target of JunB in the survival mechanism of ß-cells under inflammatory stress.


Subject(s)
Activating Transcription Factor 3/metabolism , Diabetes Mellitus, Type 1/metabolism , Inflammation/metabolism , Insulin-Secreting Cells/metabolism , Proto-Oncogene Proteins c-jun/metabolism , Animals , Gene Knockdown Techniques , Humans , Mice , Mice, Inbred NOD , Mice, Transgenic , Proto-Oncogene Proteins c-jun/genetics , Signal Transduction , Tumor Necrosis Factor-alpha/pharmacology
10.
Oncogene ; 30(36): 3821-32, 2011 Sep 08.
Article in English | MEDLINE | ID: mdl-21460858

ABSTRACT

The role of the AP-1 transcription factor Fra-1 (encoded by Fosl1) in inflammatory responses associated with lung disease is largely unknown. Here, we show that Fra-1 overexpression in mice reduced proinflammatory cytokine production in response to injection of lipopolysaccharide (LPS), a Toll-like receptor (TLR)-ligand. Unexpectedly, Fra-1 transgenic mice died rapidly following LPS treatment, showing severe interstitial lung disease and displaying massive accumulation of macrophages and overproduction of several chemokines, including macrophage chemoattractant protein-1 (MCP-1, encoded by Ccl2). To assess the clinical relevance of Fra-1 in lung pathology, mice were treated with the anticancer drug gefitinib (Iressa), which can lead to interstitial lung disease in patients. Gefitinib-treated mice showed increased Fosl1 and Ccl2 expression and developed interstitial lung disease in response to LPS, endogenous TLR ligands and chemotherapy. Moreover, deletion of Fra-1 or blocking MCP-1 receptor signaling in mice attenuated gefitinib-enhanced lethality in response to LPS. Importantly, human alveolar macrophages showed enhanced LPS-induced FOSL1 and CCL2 expression after gefitinib treatment. These results indicate that Fra-1 is an important mediator of interstitial lung disease following gefitinib treatment.


Subject(s)
Ligands , Lung Diseases, Interstitial/chemically induced , Proto-Oncogene Proteins c-fos/metabolism , Quinazolines/adverse effects , Toll-Like Receptors/metabolism , Animals , Chemokine CCL2/metabolism , Female , Gefitinib , Humans , Lipopolysaccharides/metabolism , Macrophages, Alveolar/cytology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Inbred ICR , Mice, Transgenic
11.
Oncogene ; 30(13): 1506-17, 2011 Mar 31.
Article in English | MEDLINE | ID: mdl-21119595

ABSTRACT

Mice lacking c-fos develop osteopetrosis due to a block in osteoclast differentiation. Carboxy-terminal phosphorylation of Fos on serine 374 by ERK1/2 and serine 362 by RSK1/2 regulates Fos stability and transactivation potential in vitro. To assess the physiological relevance of Fos phosphorylation in vivo, serine 362 and/or serine 374 was replaced by alanine (Fos362A, Fos374A and FosAA) or by phospho-mimetic aspartic acid (FosDD). Homozygous mutants were healthy and skeletogenesis was largely unaffected. Fos C-terminal phosphorylation, predominantly on serine 374, was found important for osteoclast differentiation in vitro and affected lipopolysaccharide (LPS)-induced cytokine response in vitro and in vivo. Importantly, skin papilloma development was delayed in FosAA, Fos362A and Rsk2-deficient mice, accelerated in FosDD mice and unaffected in Fos374A mutants. Furthermore, the related Fos protein and putative RSK2 target Fra1 failed to substitute for Fos in papilloma development. This indicates that phosphorylation of serines 362 and 374 exerts context-dependent roles in modulating Fos activity in vivo. Inhibition of Fos C-terminal phosphorylation on serine 362 by targeting RSK2 might be of therapeutic relevance for skin tumours.


Subject(s)
Bone and Bones/metabolism , Cell Transformation, Neoplastic/metabolism , Cytokines/biosynthesis , Homeostasis/physiology , Proto-Oncogene Proteins c-fos/physiology , Skin Neoplasms/etiology , Animals , Bone Remodeling , Cell Differentiation , Cells, Cultured , Gene Expression Regulation , Mice , Mice, Inbred C57BL , Osteoclasts/cytology , Phosphorylation , Phosphoserine/metabolism
12.
Cell Death Differ ; 17(9): 1409-19, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20300111

ABSTRACT

Jun is essential for fetal development, as fetuses lacking Jun die at mid-gestation with multiple cellular defects in liver and heart. Embryos expressing JunD in place of Jun (Jun(d/d)) can develop to term with normal fetal livers, but display cardiac defects as observed in fetuses lacking Jun. Jun(d/d) mouse embryonic fibroblasts (MEFs) exhibit early senescence, which can be rescued by EGF and HB-EGF stimulation, probably through activation of Akt signaling. Thus, JunD cannot functionally replace Jun in regulating fibroblast proliferation. In Jun(-/-) fetal livers, increased hydrogen peroxide levels are detected and expression of Nrf1 and Nrf2 (nuclear erythroid 2-related transcription factors) is downregulated. Importantly, increased oxidative stress as well as expression of Nrf1 and Nrf2 is rescued by JunD in Jun(d/d) fetal livers. These data show that Jun is of critical importance for cellular protection against oxidative stress in fetal livers and fibroblasts, and Jun-dependent cellular senescence can be restored by activation of the epidermal growth factor receptor pathway.


Subject(s)
Cell Proliferation , Oxidative Stress , Proto-Oncogene Proteins c-jun/metabolism , Proto-Oncogene Proteins/metabolism , Signal Transduction/physiology , Animal Structures/abnormalities , Animal Structures/metabolism , Animal Structures/pathology , Animals , Antioxidants/pharmacology , Cell Proliferation/drug effects , Cellular Senescence/drug effects , Cellular Senescence/genetics , Cyclin D1/genetics , Cyclin D1/metabolism , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Embryo, Mammalian/abnormalities , Embryo, Mammalian/metabolism , Embryo, Mammalian/pathology , Epidermal Growth Factor/pharmacology , ErbB Receptors/genetics , Fibroblasts/metabolism , Fibroblasts/pathology , Gene Expression/drug effects , Gene Expression/genetics , Heart Defects, Congenital/genetics , Heart Defects, Congenital/pathology , Hepatocytes/cytology , Hepatocytes/metabolism , Hydrogen Peroxide/metabolism , Liver/metabolism , Liver/pathology , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , NF-E2-Related Factor 2/genetics , Nuclear Respiratory Factor 1/genetics , Oxidative Stress/genetics , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins c-akt/metabolism , Proto-Oncogene Proteins c-jun/genetics , Signal Transduction/drug effects , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism
13.
Ann Rheum Dis ; 69 Suppl 1: i86-88, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19995753

ABSTRACT

The Fos and Jun proteins are members of the AP-1 transcription factor complex, which is a central regulator for many cellular functions. This paper summarises the important functions of Fos proteins in bone development, with special emphasis on the Fos-related proteins Fra-1 and Fra-2. These factors determine the functions of osteoblasts and osteoclasts and regulate cytokine signalling during bone development. Likewise, the Jun proteins control the expression of cytokines and chemokines and are probably causally involved in inflammatory skin diseases such as psoriasis. Investigations into the molecular mechanisms responsible for skin inflammation have revealed that Jun proteins control cytokine expression, such as granulocyte colony-stimulating factor, IL-6 and tumour necrosis factor alpha by transcriptional and posttranscriptional pathways. Finally, the paper discusses the relevance of the Jun-dependent mouse model for psoriasis for preclinical studies in the field of anti-angiogenic therapies.


Subject(s)
Bone Development/physiology , Inflammation/physiopathology , Transcription Factor AP-1/physiology , Angiogenesis Inhibitors/therapeutic use , Animals , Dermatitis/physiopathology , Disease Models, Animal , Humans , Inflammation/drug therapy , Mice , Proto-Oncogene Proteins c-fos/physiology , Proto-Oncogene Proteins c-jun/physiology , Psoriasis/metabolism
14.
Oncogene ; 27(47): 6102-9, 2008 Oct 16.
Article in English | MEDLINE | ID: mdl-18679426

ABSTRACT

Chronic inflammation is an important cancer risk factor but the molecular pathways linking inflammation and cancer are incompletely understood. The transcription factor c-Jun/AP-1 (activator protein 1) is involved in inflammatory responses and tumorigenesis and has been proposed as an essential mediator of oncogenic beta-catenin signaling in the intestine. Here, we examined the functions of c-Jun in two distinct mouse models of conditional and intestine-specific activation of beta-catenin. c-Jun is strongly expressed in the small intestine of mutant mice. However, beta-catenin-dependent cell proliferation is surprisingly not affected in mice lacking c-jun in intestinal epithelium, suggesting that c-Jun is not an essential immediate target of beta-catenin signaling in the small intestine. To examine the functions of Jun and Fos proteins during inflammation and cancer in the colon, colitis-associated tumors were induced chemically in the respective knockout mice. Tumors were characterized by activated beta-catenin and strongly expressed c-Jun and JunB. However, tumorigenesis was not affected by inactivation of c-Jun in either intestinal epithelium or myeloid cells. Moreover, tumorigenesis was not altered in mice lacking junB, junD, c-fos, fra-1 or fra-2, suggesting that inhibition of c-Jun or other single AP-1 proteins is not a determining factor in colitis-associated cancer in mice.


Subject(s)
Colitis/complications , Colonic Neoplasms/etiology , Proto-Oncogene Proteins c-jun/physiology , Transcription Factor AP-1/physiology , beta Catenin/physiology , Animals , Azoxymethane/toxicity , Genes, APC , Intestinal Mucosa/chemistry , Mice , Proto-Oncogene Proteins/physiology , Proto-Oncogene Proteins c-jun/analysis , Signal Transduction
15.
Oncogene ; 27(5): 641-52, 2008 Jan 24.
Article in English | MEDLINE | ID: mdl-17667939

ABSTRACT

The activating protein-1 transcription factor, in particular the Jun proteins play critical roles in the regulation of cell proliferation and tumor progression. To study the potential clinical relevance of interfering with JunB expression, we generated retroviruses expressing short hairpin RNA. Reduction of JunB levels causes increased proliferation and tumorigenicity in wild-type murine fibroblasts, whereas in c-Jun knockout cells p53-independent cell cycle arrest and apoptosis are induced. Using melanoma-derived B16-F10 cancer cells the combination of JunB knockdown and c-Jun/JNK inactivation leads to cell cycle arrest and apoptosis-inducing factor-dependent apoptosis. Furthermore, the combined treatment extends survival of mice inoculated with the tumor cells. These results indicate that in the absence of c-Jun, JunB can act as a tumor promoter and inactivation of both, c-Jun and JunB, could provide a valuable strategy for antitumor intervention.


Subject(s)
Cell Proliferation , Proto-Oncogene Proteins c-jun/metabolism , RNA Interference , Animals , Apoptosis , Fibroblasts , Humans , Melanoma/pathology , Mice , Neoplasms/therapy , Retroviridae , Skin Neoplasms/pathology , Tumor Cells, Cultured
16.
Cell Death Differ ; 15(1): 89-93, 2008 Jan.
Article in English | MEDLINE | ID: mdl-17762881

ABSTRACT

During the development and organogenesis of all multicellular organisms, cell fate decisions determine whether cells undergo proliferation, differentiation, or aging. Two independent stress kinase signaling pathways, p38-MAPK, and JNKs, have evolved that relay developmental and environmental cues to determine cell responses. Although multiple stimuli can activate these two stress kinase pathways, the functional interactions and molecular cross-talks between these common second signaling cascades are poorly elucidated. Here we report that JNK and p38-MAPK pathways antagonistically control cellular senescence, oncogenic transformation, and proliferation in primary mouse embryonic fibroblasts (MEFs). Similarly, genetic inactivation of the JNK pathway results in impaired proliferation of fetal hepatoblasts in vitro and defective adult liver regeneration in vivo, which is rescued by inhibition of the p38-MAPK pathway. Thus, the balance between the two stress-signaling pathways, MKK7-JNK and MKK3/6-p38-MAPK, determines cell fate and links environmental and developmental stress to cell cycle arrest, senescence, oncogenic transformation, and adult tissue regeneration.


Subject(s)
Cell Proliferation , Cell Transformation, Neoplastic , Cellular Senescence , JNK Mitogen-Activated Protein Kinases/metabolism , Liver Regeneration , MAP Kinase Signaling System , p38 Mitogen-Activated Protein Kinases/metabolism , Animals , CDC2 Protein Kinase/metabolism , Cells, Cultured , Fibroblasts/metabolism , Hepatocytes/metabolism , Mice , Mice, Mutant Strains
17.
Oncogene ; 26(33): 4863-71, 2007 Jul 19.
Article in English | MEDLINE | ID: mdl-17297445

ABSTRACT

Loss of JunB has been observed in human leukemia and lymphoma, but it remains unknown, whether this loss is relevant to disease progression. Here, we investigated the consequences of JunB deficiency using Abelson-induced B-lymphoid leukemia as a model system. Mice deficient in JunB expression succumbed to Abelson-induced leukemia with increased incidence and significantly reduced latency. Similarly, bcr/abl p185-transformed JunB-deficient (junB(Delta/Delta)) cells induced leukemia in RAG2(-/-) mice displaying a more malignant phenotype. These observations indicated that cell intrinsic effects within the junB(Delta/Delta) tumor cells accounted for the accelerated leukemia development. Indeed, explantated bcr/abl p185 transformed junB(Delta/Delta) cells proliferated faster than the control cells. The proliferative advantage emerged slowly after the initial transformation process and was associated with increased expression levels of the cell cycle kinase cdk6 and with decreased levels of the cell cycle inhibitor p16(INK4a). These alterations were due to irreversible reprogramming of the cell, because - once established - accelerated disease induced by junB(Delta/Delta) cells was not reverted by re-introducing JunB. Consistent with this observation, we found that the p16 promoter was methylated. Thus, JunB functions as a gatekeeper during tumor evolution. In its absence, transformed leukemic cells acquire an enhanced proliferative capacity, which presages a more malignant disease.


Subject(s)
Leukemia, Lymphoid/pathology , Proto-Oncogene Proteins c-jun/physiology , Animals , Blotting, Western , Cell Line , Cell Line, Tumor , Cell Proliferation , Cell Survival , Cells, Cultured , Cyclin-Dependent Kinase 6/genetics , Cyclin-Dependent Kinase 6/metabolism , Cyclin-Dependent Kinase Inhibitor p16/genetics , Cyclin-Dependent Kinase Inhibitor p16/metabolism , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , DNA-Binding Proteins/physiology , Flow Cytometry , Fusion Proteins, bcr-abl/genetics , Fusion Proteins, bcr-abl/metabolism , Fusion Proteins, bcr-abl/physiology , Gene Expression , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Leukemia, Experimental/genetics , Leukemia, Experimental/metabolism , Leukemia, Experimental/pathology , Leukemia, Lymphoid/genetics , Leukemia, Lymphoid/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Nude , Proto-Oncogene Proteins c-jun/genetics , Proto-Oncogene Proteins c-jun/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Time Factors , Transfection
18.
Ann N Y Acad Sci ; 1018: 458-65, 2004 Jun.
Article in English | MEDLINE | ID: mdl-15240402

ABSTRACT

The c-fos knockout mice (c-fos KO) and corticotropin-releasing hormone knockout mice (CRH KO) can serve as interesting models for studying mechanisms involved in response of the organism to stress, focused mainly on the hypothalamic-pituitary-adrenal (HPA) axis and sympathoadrenal system (SAS). The present study focused on the investigation of changes in gene expression of catecholamine biosynthesizing enzymes tyrosine hydroxylase (TH), dopamine-beta-hydroxylase (DBH), and phenylethanolamine N-methyltransferase (PNMT) in adrenal medulla of c-fos KO and CRH KO mice stressed by immobilization. Levels of TH, DBH, and PNMT mRNA were determined by reverse transcription-polymerase chain reaction (RT-PCR). Single immobilization for 2 h significantly increased adrenomedullary TH, DBH, and PNMT mRNA levels in both c-fos KO and wild-type (WT) mice compared to unstressed controls. In CRH KO mice, PNMT gene expression was not increased to the same extent after single, but especially after repeated immobilization as in WT mice, in contrast to TH and DBH mRNA levels. Thus, our data indicate that CRH deficiency can influence the PNMT mRNA level in adrenal medulla during stress, confirming the idea that the HPA axis plays the crucial role in PNMT gene regulation in mice. On the other hand, c-Fos protein probably does not play a crucial role in TH, DBH, and PNMT gene expression in adrenal medulla under stress conditions.


Subject(s)
Catecholamines/biosynthesis , Immobilization , Stress, Physiological/enzymology , Adrenal Medulla/enzymology , Animals , Corticotropin-Releasing Hormone/genetics , Corticotropin-Releasing Hormone/physiology , Female , Male , Mice , Mice, Knockout , Proto-Oncogene Proteins c-fos/genetics , Proto-Oncogene Proteins c-fos/physiology , RNA, Messenger/genetics , RNA, Messenger/metabolism
19.
Bone ; 34(5): 776-82, 2004 May.
Article in English | MEDLINE | ID: mdl-15121008

ABSTRACT

Increased bone mass due to elevated number of active osteoblasts has been reported for transgenic mice overexpressing the transcription factor Fra-1. To explore the potential of the anabolic action of Fra-1 in treatment of osteoporosis, we examined the integrity of bone matrix generated in Fra-1 transgenic mice. Femora from Fra-1 transgenic (Fra-1 tg) and wild-type littermates were analyzed for bone mineralization density distribution (BMDD) and nanostructure using quantitative backscattered electron imaging (qBEI) and scanning small angle X-ray scattering (scanning-SAXS), respectively. For comparison, we studied mice lacking c-Fos (Fos-/-), which develop osteopetrosis because of the absence of osteoclasts. Morphometrical analysis of metaphyseal spongiosa revealed an up to 5-fold increase in bone volume for Fra-1 transgenic compared to wild type. BMDD indicated a transient lower mineralization of bone for Fra-1 transgenic at 5 and 8 weeks, which became comparable to that of wild-type mice by 8 months. The homogeneity of mineralization was not altered in the Fra-1 transgenic mice at any ages examined. However, it was strikingly reduced in Fos-/- due to an abundance of hypermineralized cartilage. The bone nanostructure did not show abnormalities in Fra-1 transgenic or Fos-/-. These results provide a rationale for the development of therapeutic applications involving Fra-1-induced bone formation.


Subject(s)
Bone and Bones/ultrastructure , Calcification, Physiologic , Osteoporosis/physiopathology , Proto-Oncogene Proteins c-fos/physiology , Animals , Bone and Bones/anatomy & histology , Mice , Mice, Transgenic , Nanotechnology , Proto-Oncogene Proteins c-fos/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...