Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 28
Filter
Add more filters










Publication year range
1.
J Med Virol ; 96(5): e29684, 2024 May.
Article in English | MEDLINE | ID: mdl-38773828

ABSTRACT

Kaposi's sarcoma (KS) may derive from Kaposi's sarcoma herpesvirus (KSHV)-infected human mesenchymal stem cells (hMSCs) that migrate to sites characterized by inflammation and angiogenesis, promoting the initiation of KS. By analyzing the RNA sequences of KSHV-infected primary hMSCs, we have identified specific cell subpopulations, mechanisms, and conditions involved in the initial stages of KSHV-induced transformation and reprogramming of hMSCs into KS progenitor cells. Under proangiogenic environmental conditions, KSHV can reprogram hMSCs to exhibit gene expression profiles more similar to KS tumors, activating cell cycle progression, cytokine signaling pathways, endothelial differentiation, and upregulating KSHV oncogenes indicating the involvement of KSHV infection in inducing the mesenchymal-to-endothelial (MEndT) transition of hMSCs. This finding underscores the significance of this condition in facilitating KSHV-induced proliferation and reprogramming of hMSCs towards MEndT and closer to KS gene expression profiles, providing further evidence of these cell subpopulations as precursors of KS cells that thrive in a proangiogenic environment.


Subject(s)
Herpesvirus 8, Human , Mesenchymal Stem Cells , Sarcoma, Kaposi , Humans , Herpesvirus 8, Human/physiology , Herpesvirus 8, Human/genetics , Sarcoma, Kaposi/virology , Mesenchymal Stem Cells/virology , Cell Differentiation , Cells, Cultured , Gene Expression Profiling , Cell Proliferation
2.
Front Cell Dev Biol ; 11: 1265475, 2023.
Article in English | MEDLINE | ID: mdl-38274271

ABSTRACT

Tristetraprolin (TTP) is an RNA binding protein that destabilizes mRNAs of factors involved in proliferation, invasiveness, and inflammation. Disruption of the gene that codes for TTP (Zfp36) led to severe arthritis, autoimmunity, cachexia and dermatitis in mice. It has been shown that these phenotypes were mostly due to excessive TNFα levels in the affected tissues. We have previously reported that TTP expression is required for lactation maintenance. Our results indicated that conditional MG TTP-KO female mice displayed early involution due to the untimely induction of pro-inflammatory pathways led mostly by TNFα overexpression. Here we show that reducing TTP levels not only affects the fully differentiated mammary gland, but also harms morphogenesis of this tissue by impairing the progenitor cell population. We found that Zfp36 expression is linked to mammary stemness in human and mice. In addition, diminishing TTP expression and activity induced apoptosis of stem-like mouse mammary cells, reduced its ability to form mammospheres in culture and to develop into complete glands when implanted into cleared mammary fat pads in vivo. Our results show that survival of the stem-like cells is compromised by increased levels of inflammatory cytokines and stimulation of signaling cascades involving NFκB, STAT3 and MAPK-p38 activation. Moreover, TNFα overexpression and the consequent p38 phosphorylation would be the leading cause of progenitor cell death upon TTP expression restriction. Taken together, our results reveal the relevance of TTP for the maintenance of the mammary progenitor cell compartment by maintaining local TNFα levels at bay.

3.
PLoS Pathog ; 16(10): e1009006, 2020 10.
Article in English | MEDLINE | ID: mdl-33057440

ABSTRACT

Kaposi's sarcoma-associated herpesvirus (KSHV) vGPCR is a constitutively active G protein-coupled receptor that subverts proliferative and inflammatory signaling pathways to induce cell transformation in Kaposi's sarcoma. Cyclooxygenase-2 (COX-2) is an inflammatory mediator that plays a key regulatory role in the activation of tumor angiogenesis. Using two different transformed mouse models and tumorigenic full KSHV genome-bearing cells, including KSHV-Bac16 based mutant system with a vGPCR deletion, we demostrate that vGPCR upregulates COX-2 expression and activity, signaling through selective MAPK cascades. We show that vGPCR expression triggers signaling pathways that upregulate COX-2 levels due to a dual effect upon both its gene promoter region and, in mature mRNA, the 3'UTR region that control mRNA stability. Both events are mediated by signaling through ERK1/2 MAPK pathway. Inhibition of COX-2 in vGPCR-transformed cells impairs vGPCR-driven angiogenesis and treatment with the COX-2-selective inhibitory drug Celecoxib produces a significant decrease in tumor growth, pointing to COX-2 activity as critical for vGPCR oncogenicity in vivo and indicating that COX-2-mediated angiogenesis could play a role in KS tumorigenesis. These results, along with the overexpression of COX-2 in KS lesions, define COX-2 as a potential target for the prevention and treatment of KSHV-oncogenesis.


Subject(s)
Herpesvirus 8, Human/metabolism , Matrix Metalloproteinase 2/biosynthesis , Receptors, G-Protein-Coupled/metabolism , Sarcoma, Kaposi/blood supply , Animals , Carcinogenesis , Cell Transformation, Neoplastic/genetics , Endothelial Cells/metabolism , GTP-Binding Proteins/genetics , Herpesvirus 8, Human/genetics , MAP Kinase Signaling System , Matrix Metalloproteinase 2/genetics , Matrix Metalloproteinase 2/metabolism , Mice , Mice, Nude , NIH 3T3 Cells , Neovascularization, Pathologic/metabolism , Neovascularization, Pathologic/pathology , Neovascularization, Pathologic/virology , Oncogenes , Receptors, G-Protein-Coupled/genetics , Sarcoma, Kaposi/metabolism , Sarcoma, Kaposi/pathology , Sarcoma, Kaposi/virology , Signal Transduction , Transcriptional Activation
4.
Cancer Res ; 78(16): 4497-4511, 2018 08 15.
Article in English | MEDLINE | ID: mdl-29748375

ABSTRACT

R-spondin3 (RSPO3) is a member of a family of secreted proteins that enhance Wnt signaling pathways in diverse processes, including cancer. However, the role of RSPO3 in mammary gland and breast cancer development remains unclear. In this study, we show that RSPO3 is expressed in the basal stem cell-enriched compartment of normal mouse mammary glands but is absent from committed mature luminal cells in which exogenous RSPO3 impairs lactogenic differentiation. RSPO3 knockdown in basal-like mouse mammary tumor cells reduced canonical Wnt signaling, epithelial-to-mesenchymal transition-like features, migration capacity, and tumor formation in vivo Conversely, RSPO3 overexpression, which was associated with some LGR and RUNX factors, highly correlated with the basal-like subtype among patients with breast cancer. Thus, we identified RSPO3 as a novel key modulator of breast cancer development and a potential target for treatment of basal-like breast cancers.Significance: These findings identify RSPO3 as a potential therapetuic target in basal-like breast cancers.Graphical Abstract: http://cancerres.aacrjournals.org/content/canres/78/16/4497/F1.large.jpg Cancer Res; 78(16); 4497-511. ©2018 AACR.


Subject(s)
Breast Neoplasms/genetics , Breast/metabolism , Mammary Neoplasms, Animal/genetics , Thrombospondins/genetics , Animals , Breast/pathology , Breast Neoplasms/pathology , Cell Differentiation/genetics , Cell Line, Tumor , Cell Proliferation/genetics , Core Binding Factor alpha Subunits/genetics , Epithelial Cells/pathology , Epithelial-Mesenchymal Transition/genetics , Female , Gene Expression Regulation, Neoplastic , Humans , Mammary Neoplasms, Animal/pathology , Mice , Receptors, G-Protein-Coupled/genetics , Wnt Signaling Pathway/genetics
5.
Mol Cell Endocrinol ; 419: 259-67, 2016 Jan 05.
Article in English | MEDLINE | ID: mdl-26522133

ABSTRACT

Epidermal growth factor (EGF) has been suggested to play a key role in the maintenance of epithelial cell survival during lactation. Previously, we demonstrated that EGF dependent activation of PI3K pathway prevents apoptosis in confluent murine HC11 cells cultured under low nutrient conditions. The EGF protective effect is associated with increased levels of the antiapoptotic protein Bcl-XL. Here, we identify the EGF-dependent mechanism involved in cell survival that converges in the regulation of bcl-X expression by activated CREB. EGF induces Bcl-XL expression through activation of a unique bcl-X promoter, the P1; being not only the PI3K/AKT signaling pathway but also the increase in cAMP levels and the concomitant PKA/CREB activation necessary for both bcl-XL upregulation and apoptosis avoidance. Results presented in this work suggest the existence of a novel connection between the EGF receptor and the adenylate cyclase that would have an impact in preventing apoptosis under low nutrient conditions.


Subject(s)
Culture Media/chemistry , Cyclic AMP/metabolism , Epidermal Growth Factor/pharmacology , Mammary Glands, Animal/cytology , Animals , Cell Culture Techniques , Cell Line , Cell Survival/drug effects , Culture Media/pharmacology , Cyclic AMP Response Element-Binding Protein/genetics , Cyclic AMP Response Element-Binding Protein/metabolism , Female , Mammary Glands, Animal/drug effects , Mice , Promoter Regions, Genetic , Signal Transduction/drug effects , bcl-X Protein/genetics , bcl-X Protein/metabolism
6.
Biochem J ; 467(1): 77-90, 2015 Apr 01.
Article in English | MEDLINE | ID: mdl-25588078

ABSTRACT

Mitogen-activated protein kinase (MAPK) pathways constitute key regulatory elements linking extracellular stimuli to nuclear gene expression. Immediate-early responsive genes (IEGs) of the activator protein 1 (AP-1) family, such as fos, achieve peak expression levels shortly after cells are stimulated with growth factors and sharply decrease thereafter. Several AU-rich binding proteins (AUBPs), including HuR (Hu-antigen R, Elav-like protein 1, ELAVL1) and KSRP (far upstream element-binding protein 2, KHSRP) bind to a fos AU-rich element (ARE) present in the 3'-UTR (untranslated region) of fos mRNA regulating its stability by a still poorly defined mechanism. We show in the present study that, whereas HuR binds and stabilizes transcribed reporter mRNAs bearing the fos 3'-UTR, KSRP counteracts this effect. Furthermore, we found that fos mRNA stability and HuR phosphorylation status are dependent on the activity of p38 MAPK in both epithelial cells and fibroblasts upon proliferative stimulation. Analysing PPI (protein-protein interaction) networks, we performed a thorough query of interacting proteins for p38 MAPKs, HuR and other AUBPs upon growth factor stimulation. This revealed novel HuR interactors including inhibitors of protein phosphatase 2 (PP2A) activity. Over-expression of two of these interactors, pp32 and APRIL (acidic leucine-rich nuclear phosphoprotein 32 family member B, ANP32B) and pharmacological inhibition of PP2A stabilized a fos reporter mRNA. Our results indicate that p38 MAPK regulates fos mRNA decay by affecting the state of phosphorylation of HuR while controlling yet to be fully elucidated PP regulatory networks.


Subject(s)
ELAV Proteins/metabolism , MAP Kinase Signaling System/drug effects , Mitogens/pharmacology , Proto-Oncogene Proteins c-fos/metabolism , RNA Stability/drug effects , RNA, Messenger/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism , 3' Untranslated Regions/drug effects , Animals , Cell Proliferation/drug effects , ELAV Proteins/genetics , ELAV-Like Protein 1 , HEK293 Cells , HeLa Cells , Humans , Mice , Mutation , NIH 3T3 Cells , Phosphorylation/drug effects , Protein Kinase Inhibitors/pharmacology , Protein Processing, Post-Translational/drug effects , Proto-Oncogene Proteins c-fos/chemistry , Proto-Oncogene Proteins c-fos/genetics , RNA, Messenger/chemistry , RNA-Binding Proteins/chemistry , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism , Recombinant Fusion Proteins/metabolism , Trans-Activators/chemistry , Trans-Activators/genetics , Trans-Activators/metabolism
7.
Biochem J ; 454(2): 345-57, 2013 Sep 01.
Article in English | MEDLINE | ID: mdl-23789592

ABSTRACT

Integrins are heterodimeric cell-surface adhesion receptors that play a critical role in tissue development. Characterization of the full-length mRNA encoding the ß1 subunit (Itgb1) revealed an alternative functional cleavage and polyadenylation site that yields a new Itgb1 mRNA isoform 578 bp shorter than that previously reported. Using a variety of experimental and bioinformatic approaches, we found that the two Itgb1 isoforms are expressed at different levels in a variety of mouse tissues, including the mammary gland, where they are differentially regulated at successive developmental stages. The longer mRNA species is prevelant during lactation, whereas the shorter is induced after weaning. In 3D cultures, where expression of integrin ß1 protein is required for normal formation of acini, experimental blockade of the longer isoform induced enhanced expression of the shorter species which allowed normal morphological mammary differentiation. The short isoform lacks AU-rich motifs and miRNA target sequences that are potentially implicated in the regulation of mRNA stability and translation efficiency. We further determined that the AU-binding protein HuR appears to selectively stabilize the longer isoform in the mammary gland. In summary, the results of the present study identify a new regulatory instance involved in the fine-tuning of Itgb1 expression during mammary gland development and function.


Subject(s)
Integrin beta1/metabolism , Mammary Glands, Animal/growth & development , Mammary Glands, Animal/metabolism , RNA Isoforms/metabolism , RNA Processing, Post-Transcriptional , RNA Stability , RNA, Messenger/metabolism , Animals , Cell Culture Techniques , Cell Differentiation , Cell Line , Data Mining , Female , Gene Expression Regulation, Developmental , Integrin beta1/chemistry , Integrin beta1/genetics , Lactation/metabolism , Mammary Glands, Animal/cytology , Mice , Mice, Inbred BALB C , Polyadenylation , Pregnancy , RNA Isoforms/antagonists & inhibitors , RNA, Messenger/antagonists & inhibitors , RNA, Small Interfering , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Specific Pathogen-Free Organisms , Weaning
8.
Lung Cancer ; 77(1): 168-75, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22418244

ABSTRACT

While changes in heme oxygenase (HO-1) in lung cancer have already been reported, conflicting results were obtained for enzyme expression in human lung cancer specimens. Therefore, the aim of this work was to study HO-1 expression in a large collection of human lung cancer samples. For this purpose, we analyzed the expression of HO-1 in an organized tissue microarray (TMA) and investigated its correlation with clinicopathological data. Ninety-six percent of tumor samples were positive for HO-1, and the expression of HO-1 was significantly higher in cancerous than in non-cancerous tissues. Importantly, HO-1 expression correlated with advanced stages and lymph node involvement. Additionally, quantitative RT-PCR in 18 pairs of human lung carcinomas and their adjacent non-malignant tissues was performed. Our results demonstrate that HO-1 protein is upregulated in epithelial malignant cells in NSCLC and its expression is associated with higher stages of the disease. Additionally, different subcellular localization is observed between tumor and adjacent non-malignant tissues.


Subject(s)
Carcinoma, Non-Small-Cell Lung/enzymology , Gene Expression , Heme Oxygenase-1/metabolism , Lung Neoplasms/enzymology , Animals , Carcinoma, Non-Small-Cell Lung/mortality , Carcinoma, Non-Small-Cell Lung/secondary , Cell Line, Tumor , Epithelial Cells/enzymology , Epithelial Cells/pathology , Female , Heme Oxygenase-1/genetics , Humans , Kaplan-Meier Estimate , Lung Neoplasms/mortality , Lung Neoplasms/pathology , Lymphatic Metastasis , Male , Mice , NIH 3T3 Cells , Real-Time Polymerase Chain Reaction , Tissue Array Analysis , Up-Regulation
9.
J Cell Biochem ; 110(4): 857-65, 2010 Jul 01.
Article in English | MEDLINE | ID: mdl-20564184

ABSTRACT

It has been reported that expression of tumor necrosis factor superfamily members occur at the onset of the mammary gland post-lactational involution. One of these proteins, tumor necrosis factor alpha (TNFalpha), is a major mediator of inflammation that is able to induce expression of several cytokines. Leukemia inhibitory factor (LIF) is an inflammatory cytokine that is induced and plays a fundamental role during post-lactational involution of the mammary gland. Therefore, our goal was to determine whether TNFalpha activity in the mammary epithelium might include regulation of LIF expression. This biological role would increase the significance of TNFalpha expression at the end of lactation. Our results show that TNFalpha was able to induce LIF transcription through ERK1/2 activation in a non-tumorigenic mouse mammary epithelial cell line, SCp2. We found that activation of TNFalpha receptor-2 (TNFR2) was specifically involved in triggering this signaling pathway. In addition, our data suggest the participation of AP-1 transcription factor family members in this pathway. We determined that TNFalpha treatment induced c-fos transcription, and blocking AP-1 activity resulted in a significant inhibition of TNFalpha-induced LIF expression. Finally, we found that TNFalpha was also able to trigger LIF expression and ERK1/2 activation in the mouse mammary gland in vivo. Therefore, our data suggest that TNFalpha may contribute to mammary gland involution by, among other activities, eliciting LIF expression through ERK1/2 and AP1 activation.


Subject(s)
Leukemia Inhibitory Factor/metabolism , Mammary Glands, Human/enzymology , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Tumor Necrosis Factor-alpha/physiology , Animals , Blotting, Western , Cell Line , Electrophoretic Mobility Shift Assay , Enzyme Activation , Humans , Immunohistochemistry , Mammary Glands, Human/cytology , Mice , Mice, Inbred BALB C , Transcription Factor AP-1/metabolism
10.
Biochim Biophys Acta ; 1793(3): 496-505, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19121340

ABSTRACT

Apoptosis is the predominant process controlling cell deletion during post-lactational mammary gland remodeling. The members of the Bcl-2 protein family, whose expression levels are under the control of lactogenic hormones, internally control this mechanism. Epidermal growth factor (EGF) belongs to a family of proteins that act as survival factors for mammary epithelial cells upon binding to specific membrane tyrosine kinase receptors. Expression of EGF peaks during lactation and dramatically decreases in the involuting mammary gland. Though it was suggested that the protective effect of EGF is mediated through the phosphatidylinositol-3-kinase (PI3K) or MEK/ERK kinases activities, little is known about the downstream mechanisms involved on the anti-apoptotic effect of EGF on mammary epithelial cells; particularly the identity of target genes controlling apoptosis. Here, we focused on the effect of EGF on the survival of mammary epithelial cells. We particularly aimed at the characterization of the signaling pathways that were triggered by this growth factor, impinge upon expression of Bcl-2 family members and therefore have an impact on the regulation of cell survival. We demonstrate that EGF provokes the induction of the anti-apoptotic isoform Bcl-XL and the phosphorylation and down-regulation of the pro-apoptotic protein Bad. The activation of JNK and PI3K/AKT signaling pathways promotes the induction of Bcl-XL while AKT activation also leads to Bad phosphorylation and down-regulation. This protective effect of EGF correlates mainly with the up-regulation of Bcl-XL than with the down-regulation of Bad. In fact, HC11 cells unable to express bcl-X, die even in the presence of EGF. In this context, Bcl-XL emerges as a key anti-apoptotic molecule critical for mediating EGF cell survival.


Subject(s)
Epidermal Growth Factor/metabolism , Epithelial Cells/metabolism , Mammary Glands, Animal/metabolism , bcl-X Protein/metabolism , Animals , Apoptosis , Cell Survival , Cytochromes c/metabolism , Down-Regulation , Female , Humans , JNK Mitogen-Activated Protein Kinases/metabolism , Mice , Mitochondria/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Phosphorylation , Proto-Oncogene Proteins c-akt/metabolism , bcl-Associated Death Protein/genetics , bcl-Associated Death Protein/metabolism
11.
J Biol Chem ; 282(47): 34510-24, 2007 Nov 23.
Article in English | MEDLINE | ID: mdl-17881360

ABSTRACT

Heme oxygenase-1 (HO-1), an inducible enzyme that metabolizes the heme group, is highly expressed in human Kaposi sarcoma lesions. Its expression is up-regulated by the G protein-coupled receptor from the Kaposi sarcoma-associated herpes virus (vGPCR). Although recent evidence shows that HO-1 contributes to vGPCR-induced tumorigenesis and vascular endothelial growth factor (VEGF) expression, the molecular steps that link vGPCR to HO-1 remain unknown. Here we show that vGPCR induces HO-1 expression and transformation through the Galpha(12/13) family of heterotrimeric G proteins and the small GTPase RhoA. Targeted small hairpin RNA knockdown expression of Galpha(12), Galpha(13), or RhoA and inhibition of RhoA activity impair vGPCR-induced transformation and ho-1 promoter activity. Knockdown expression of RhoA also reduces vGPCR-induced VEFG-A secretion and blocks tumor growth in a murine allograft tumor model. NIH-3T3 cells expressing constitutively activated Galpha(13) or RhoA implanted in nude mice develop tumors displaying spindle-shaped cells that express HO-1 and VEGF-A, similarly to vGPCR-derived tumors. RhoAQL-induced tumor growth is reduced 80% by small hairpin RNA-mediated knockdown expression of HO-1 in the implanted cells. Likewise, inhibition of HO-1 activity by chronic administration of the HO-1 inhibitor tin protoporphyrin IX to mice reduces RhoAQL-induced tumor growth by 70%. Our study shows that vGPCR induces HO-1 expression through the Galpha(12/13)/RhoA axes and shows for the first time a potential role for HO-1 as a therapeutic target in tumors where RhoA has oncogenic activity.


Subject(s)
Cell Transformation, Viral , GTP-Binding Protein alpha Subunits, Gq-G11/metabolism , Heme Oxygenase-1/metabolism , Herpesvirus 8, Human/metabolism , Receptors, Chemokine/metabolism , rhoA GTP-Binding Protein/metabolism , Animals , Cell Transformation, Viral/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Heme Oxygenase-1/antagonists & inhibitors , Humans , Mice , Mice, Nude , NIH 3T3 Cells , Photosensitizing Agents/pharmacology , Promoter Regions, Genetic/genetics , Protoporphyrins/pharmacology , Receptors, Chemokine/genetics , Vascular Endothelial Growth Factor A/metabolism , Xenograft Model Antitumor Assays
12.
J Immunol ; 177(6): 4037-46, 2006 Sep 15.
Article in English | MEDLINE | ID: mdl-16951367

ABSTRACT

We have previously shown that bacterial DNA activates human neutrophils in a CpG-independent manner. In this study, we have characterized the signaling pathways involved in the activation mechanism. We found that p38 MAPK, ERK1/2, and JNK pathways, as well as the PI3K/Akt pathway, are activated by bacterial DNA. We also determined that bacterial DNA induces NF-kappaB and AP-1 activation. When analyzing the role of these pathways on neutrophil functions, we observed that up-regulation of CD11b triggered by bacterial DNA was decreased by pharmacological inhibitors of the p38 MAPK, ERK1/2, and JNK, whereas stimulation of IL-8 release was dependent on p38, ERK1/2, and NF-kappaB. Moreover, we found that IL-8 production was markedly enhanced by inhibition of JNK, suggesting that this pathway negatively modulates NF-kappaB-dependent transcription. We also observed that bacterial DNA stimulated IL-1R-associated kinase-1 kinase activity and its partial degradation. Finally, we determined that bacterial DNA stimulated CD11b up-regulation in TLR9(-/-) but not in MyD88(-/-) mouse neutrophils, supporting that bacterial DNA induces neutrophil activation through a TLR9-independent and MyD88-dependent pathway.


Subject(s)
DNA, Bacterial/physiology , MAP Kinase Signaling System/immunology , Neutrophils/enzymology , Neutrophils/microbiology , Animals , Cells, Cultured , Escherichia coli/genetics , Escherichia coli/immunology , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mitogen-Activated Protein Kinases/physiology , Neutrophils/metabolism
13.
FEBS Lett ; 580(10): 2512-6, 2006 May 01.
Article in English | MEDLINE | ID: mdl-16638579

ABSTRACT

Prostaglandin F2alpha (PGF2alpha) induces cyclin D1 expression and DNA synthesis in Swiss 3T3 cells. In order to assess which signaling mechanisms are implicated in these processes, we have used both a pharmacological approach and interfering mutants. We demonstrate that PGF2alpha induces extracellular-signal-regulated kinase (ERK1-2) and p38MAPK activation, and inhibition of any of these signaling pathways completely blocks PGF2alpha-stimulated DNA synthesis. We also show that ERK1-2, but not p38MAPK activation is required to induce cyclin D1 expression, strongly suggesting that the concerted action of cyclin D1 gene expression and other events are required to induce complete phosphorylation of retinoblastoma protein and S-phase entry in response to PGF2alpha.


Subject(s)
Cell Division/drug effects , Dinoprost/pharmacology , Mitogen-Activated Protein Kinases/metabolism , Animals , Blotting, Western , Cyclin D1/genetics , DNA Replication , Electrophoresis, Polyacrylamide Gel , Enzyme Activation , Mice , Swiss 3T3 Cells
14.
J Biol Chem ; 281(16): 11332-46, 2006 Apr 21.
Article in English | MEDLINE | ID: mdl-16476737

ABSTRACT

Heme oxygenase-1 (HO-1), the inducible enzyme responsible for the rate-limiting step in the heme catabolism, is expressed in AIDS-Kaposi sarcoma (KS) lesions. Its expression is up-regulated by the Kaposi sarcoma-associated herpesvirus (KSHV) in endothelial cells, but the mechanisms underlying KSHV-induced HO-1 expression are still unknown. In this study we investigated whether the oncogenic G protein-coupled receptor (KSHV-GPCR or vGPCR), one of the key KSHV genes involved in KS development, activated HO-1 expression. Here we show that vGPCR induces HO-1 mRNA and protein levels in fibroblasts and endothelial cells. Moreover, targeted knock-down gene expression of HO-1 by small hairpin RNA and chemical inhibition of HO-1 enzymatic activity by tin protoporphyrin IX (SnPP), impaired vGPCR-induced survival, proliferation, transformation, and vascular endothelial growth factor (VEGF)-A expression. vGPCR-expressing cells implanted in the dorsal flank of nude mice developed tumors with elevated HO-1 expression and activity. Chronic administration of SnPP to the implanted mice, under conditions that effectively blocked HO-1 activity and VEGF-A expression in the transplanted cells, strikingly reduced tumor growth, without apparent side effects. On the contrary, administration of the HO-1 inducer cobalt protoporphyrin (CoPP) further enhanced vGPCR-induced tumor growth. These data postulate HO-1 as an important mediator of vGPCR-induced tumor growth and suggest that inhibition of intratumoral HO-1 activity by SnPP may be a potential therapeutic strategy.


Subject(s)
Enzyme Inhibitors/pharmacology , Heme Oxygenase-1/antagonists & inhibitors , Herpesvirus 8, Human/metabolism , Receptors, G-Protein-Coupled/metabolism , Animals , Annexin A5/pharmacology , Apoptosis , Blotting, Western , Cell Proliferation , Cell Survival , Culture Media, Serum-Free/metabolism , DNA/metabolism , Dose-Response Relationship, Drug , Endothelial Cells/metabolism , Fibroblasts/metabolism , Fluorescent Antibody Technique, Indirect , Genes, Reporter , Heme/chemistry , Heme Oxygenase (Decyclizing)/metabolism , Heme Oxygenase-1/metabolism , Immunohistochemistry , Luciferases/metabolism , Metalloporphyrins/metabolism , Mice , Mice, Nude , Models, Biological , NIH 3T3 Cells , Neoplasm Transplantation , Neoplasms/metabolism , Promoter Regions, Genetic , Protoporphyrins/chemistry , Protoporphyrins/metabolism , RNA/metabolism , RNA, Messenger/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Time Factors , Transfection , Up-Regulation , Vascular Endothelial Growth Factor A/metabolism
15.
J Immunol ; 176(2): 1163-71, 2006 Jan 15.
Article in English | MEDLINE | ID: mdl-16394005

ABSTRACT

Inflammation in peripheral tissues is usually associated with the development of local acidosis; however, there are few studies aimed at analyzing the influence of acidosis on immune cells. We have shown previously that extracellular acidosis triggers human neutrophil activation, inducing a transient increase in intracellular Ca2+ concentration, a shape change response, the up-regulation of CD18 expression, and a delay of apoptosis. In this study, we analyzed the signaling pathways responsible for neutrophil activation. We found that acidosis triggers the phosphorylation of Akt (the main downstream target of PI3K) and ERK MAPK, but not that of p38 and JNK MAPK. No degradation of IkappaB was observed, supporting the hypothesis that NF-kappaB is not activated under acidosis. Inhibition of PI3K by wortmannin or LY294002 markedly decreased the shape change response and the induction of Ca2+ transients triggered by acidosis, whereas the inhibition of MEK by PD98059 or U0126 significantly inhibited the shape change response without affecting the induction of Ca2+ transients. We also found that acidosis not only induces a shape change response and the induction of Ca2+ transients in human neutrophils but also stimulates the endocytosis of FITC-OVA and FITC-dextran. Stimulation of endocytosis was partially prevented by inhibitors of PI3K and MEK. Together, our results support the notion that the stimulation of human neutrophils by extracellular acidosis is dependent on the activation of PI3K/Akt and ERK pathways. Of note, using mouse peritoneal neutrophils we observed that the enhancement of endocytosis induced by acidosis was associated with an improved ability to present extracellular Ags through a MHC class I-restricted pathway.


Subject(s)
Acidosis/immunology , MAP Kinase Signaling System , Neutrophil Activation/physiology , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Androstadienes/pharmacology , Animals , Antigen Presentation , Calcium Signaling , Cell Shape , Endocytosis , Enzyme Activation , Enzyme Inhibitors/pharmacology , Extracellular Fluid/metabolism , Female , Flavonoids/pharmacology , Histocompatibility Antigens Class I/metabolism , Humans , In Vitro Techniques , JNK Mitogen-Activated Protein Kinases/metabolism , MAP Kinase Signaling System/drug effects , Mice , Mice, Inbred C57BL , NF-kappa B/metabolism , Neutrophil Activation/drug effects , Neutrophils/cytology , Neutrophils/drug effects , Neutrophils/immunology , Neutrophils/metabolism , Phosphoinositide-3 Kinase Inhibitors , Signal Transduction , Wortmannin , p38 Mitogen-Activated Protein Kinases/metabolism
16.
J Biol Chem ; 281(10): 6136-43, 2006 Mar 10.
Article in English | MEDLINE | ID: mdl-16291739

ABSTRACT

Leukemia inhibitory factor (LIF) and oncostatin M (OSM) induce DNA synthesis in Swiss 3T3 cells through common signaling mechanism(s), whereas other related cytokines such as interleukin-6 and ciliary neurotrophic factor do not cause this response. Induction of DNA replication by LIF or prostaglandin F2alpha (PGF2alpha) occurs, in part, through different signaling events. LIF and OSM specifically trigger STAT1 cytoplasmic to nuclear translocation, whereas PGF2alpha fails to do so. However, LIF and PGF2alpha can trigger increases in ERK1/2 activity, which are required for their mitogenic responses because U0126, a MEK1/2 inhibitor, prevents both ERK1/2 activation and induction of DNA synthesis by LIF or PGF2alpha treatment. PGF2alpha induces cyclin D expression and full phosphorylation of retinoblastoma protein. In contrast, LIF fails to promote increases in cyclin D mRNA/protein levels; consequently, LIF induces DNA synthesis without promoting full phosphorylation of retinoblastoma protein (Rb). However, both LIF and PGF2alpha increase cyclin E expression. Furthermore, LIF mitogenic action does not involve protein kinase C (PKC) activation, because a PKC inhibitor does not block this effect. In contrast, PKC activity is required for PGF2alpha mitogenic action. More importantly, the synergistic effect between LIF and PGF2alpha to promote S phase entry is independent of PKC activation. These results show fundamental differences between LIF- and PGF2alpha-dependent mechanism(s) that induce cellular entry into S phase. These findings are critical in understanding how LIF and other related cytokine-regulated events participate in normal cell cycle control and may also provide clues to unravel crucial processes underlying cancerous cell division.


Subject(s)
Cyclin D1/biosynthesis , DNA Replication/physiology , Extracellular Signal-Regulated MAP Kinases/metabolism , Interleukin-6/physiology , Mitogen-Activated Protein Kinase Kinases/metabolism , Animals , Cyclin D1/genetics , Cyclin G , Cyclin G1 , Cyclins/biosynthesis , Cyclins/genetics , Cytokines/physiology , Dinoprost/physiology , Enzyme Activation/genetics , Extracellular Signal-Regulated MAP Kinases/physiology , Kinetics , Leukemia Inhibitory Factor , Mice , Mitogen-Activated Protein Kinase Kinases/physiology , Oncostatin M , Phosphorylation , Protein Kinase C/metabolism , Protein-Tyrosine Kinases/physiology , Retinoblastoma Protein/metabolism , S Phase/physiology , STAT Transcription Factors/physiology , Signal Transduction/physiology , Swiss 3T3 Cells
17.
Nat Struct Mol Biol ; 12(12): 1037-44, 2005 Dec.
Article in English | MEDLINE | ID: mdl-16299516

ABSTRACT

Serine/arginine-rich (SR) proteins are important regulators of mRNA splicing. Several postsplicing activities have been described for a subset of shuttling SR proteins, including regulation of mRNA export and translation. Using the fibronectin gene to study the links between signal-transduction pathways and SR protein activity, we show that growth factors not only modify the alternative splicing pattern of the fibronectin gene but also alter translation of reporter messenger RNAs in an SR protein-dependent fashion, providing two coregulated levels of isoform-specific amplification. These effects are inhibited by specific small interfering RNAs against SR proteins and are mediated by the AKT kinase, which elicits opposite effects to those evoked by overexpressing SR protein kinases Clk and SRPK. These results show how SR protein activity is modified in response to extracellular stimulation, leading to a concerted regulation of splicing and translation.


Subject(s)
Nuclear Proteins/metabolism , Phosphoproteins/metabolism , Protein Biosynthesis/genetics , Proto-Oncogene Proteins c-akt/metabolism , RNA Splicing , Amino Acid Sequence , Animals , Cell Nucleus/chemistry , Cell Nucleus/metabolism , Cytoplasm/chemistry , Cytoplasm/metabolism , Fibronectins/genetics , Growth Substances/metabolism , Humans , Molecular Sequence Data , Nuclear Proteins/analysis , Nuclear Proteins/genetics , Phosphatidylinositol 3-Kinases/metabolism , Phosphoproteins/analysis , Phosphoproteins/genetics , Protein Biosynthesis/drug effects , RNA, Messenger/metabolism , RNA, Small Interfering/genetics , RNA, Small Interfering/pharmacology , RNA-Binding Proteins , Serine-Arginine Splicing Factors , Signal Transduction
18.
J Biol Chem ; 280(19): 18842-52, 2005 May 13.
Article in English | MEDLINE | ID: mdl-15708845

ABSTRACT

Exposure to sources of UV radiation, such as sunlight, induces a number of cellular alterations that are highly dependent on its ability to affect gene expression. Among them, the rapid activation of genes coding for two subfamilies of proto-oncoproteins, Fos and Jun, which constitute the AP-1 transcription factor, plays a key role in the subsequent regulation of expression of genes involved in DNA repair, cell proliferation, cell cycle arrest, death by apoptosis, and tissue and extracellular matrix remodeling proteases. Besides being regulated at the transcriptional level, Jun and Fos transcriptional activities are also regulated by phosphorylation as a result of the activation of intracellular signaling cascades. In this regard, the phosphorylation of c-Jun by UV-induced JNK has been readily documented, whereas a role for Fos proteins in UV-mediated responses and the identification of Fos-activating kinases has remained elusive. Here we identify p38 MAPKs as proteins that can associate with c-Fos and phosphorylate its transactivation domain both in vitro and in vivo. This phosphorylation is transduced into changes in its transcriptional ability as p38-activated c-Fos enhances AP1-driven gene expression. Our findings indicate that as a consequence of the activation of stress pathways induced by UV light, endogenous c-Fos becomes a substrate of p38 MAPKs and, for the first time, provide evidence that support a critical role for p38 MAPKs in mediating stress-induced c-Fos phosphorylation and gene transcription activation. Using a specific pharmacological inhibitor for p38alpha and -beta, we found that most likely these two isoforms mediate UV-induced c-Fos phosphorylation in vivo. Thus, these newly described pathways act concomitantly with the activation of c-Jun by JNK/MAPKs, thereby contributing to the complexity of AP1-driven gene transcription regulation.


Subject(s)
Proto-Oncogene Proteins c-fos/chemistry , Transcription Factor AP-1/chemistry , p38 Mitogen-Activated Protein Kinases/chemistry , Active Transport, Cell Nucleus , Animals , Apoptosis , Binding Sites , Blotting, Western , Cell Cycle , Cell Line , Cell Nucleus/metabolism , DNA/chemistry , DNA Damage , Electrophoresis, Polyacrylamide Gel , Enzyme Inhibitors/pharmacology , Extracellular Matrix/metabolism , Fluorescent Antibody Technique, Indirect , Genes, Reporter , Glutathione Transferase/metabolism , HeLa Cells , Humans , Luciferases/metabolism , Mice , Microscopy, Fluorescence , Models, Biological , NIH 3T3 Cells , Phosphorylation , Protein Isoforms , Recombinant Fusion Proteins/chemistry , Subcellular Fractions , Time Factors , Transcription, Genetic , Transcriptional Activation , Transfection , Two-Hybrid System Techniques , Ultraviolet Rays , p38 Mitogen-Activated Protein Kinases/metabolism
19.
J Biol Chem ; 279(20): 21029-37, 2004 May 14.
Article in English | MEDLINE | ID: mdl-15028734

ABSTRACT

The way alternative splicing is regulated within tissues is not understood. A relevant model of this process is provided by fibronectin, an important extracellular matrix protein that plays a key role in cell adhesion and migration and contains three alternatively spliced regions known as EDI, EDII, and IIICS. We used a cell culture system to simulate mammary epithelial-stromal communication, a process that is crucial for patterning and function of the mammary gland, and studied the effects of extracellular signals on the regulation of fibronectin pre-mRNA alternative splicing. We found that soluble factors from a mammary mesenchymal cell-conditioned medium, as well as the growth factors HGF/SF (hepatocyte growth factor/scatter factor), KGF (keratinocyte growth factor), and aFGF (acidic fibroblast growth factor), stimulate EDI and IIICS but not EDII inclusion into fibronectin mRNA in the mammary epithelial cell line SCp2, favoring fibronectin isoforms associated with proliferation, migration, and tissue remodeling. We explored the signaling pathways involved in this regulation and found that the mammary mesenchymal cell-conditioned medium and HGF/SF act through a phosphatidylinositol 3-kinase-dependent cascade to alter fibronectin alternative splicing. This splicing regulation is independent from promoter structure and de novo protein synthesis but does require two exonic elements within EDI. These results shed light on how extracellular stimuli are converted into changes in splicing patterns.


Subject(s)
Alternative Splicing/genetics , Epithelial Cells/physiology , Fibronectins/genetics , Mammary Glands, Animal/cytology , Mesoderm/physiology , Phosphatidylinositol 3-Kinases/metabolism , Alternative Splicing/drug effects , Animals , Coculture Techniques , Epithelial Cells/cytology , Exons , Female , Growth Substances/pharmacology , Mesoderm/cytology , Mice , Recombinant Proteins/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Transfection
20.
Eur J Immunol ; 34(1): 200-9, 2004 Jan.
Article in English | MEDLINE | ID: mdl-14971046

ABSTRACT

Given that arginase activation may effectively influence nitric oxide (NO) production in macrophages, we have investigated the intracellular signals that regulate L-arginine metabolism and its influence on Trypanosoma cruzi growth. We demonstrate that cruzipain (Cz), a parasite antigen, induces arginase I expression in J774 cells, and the pretreatment of Cz-treated cells with N-omega-hydroxy-L-arginine (arginase inhibitor) leads to a dramatic decrease in amastigote growth. The study of intracellular signals shows that genistein [tyrosine kinase (TK) inhibitor], KT5720 [protein kinase (PK) A inhibitor] and SB203580 [p38 mitogen-activated protein kinase (MAPK) inhibitor] significantly decrease Cz-induced arginase activation. However, calphostin C (PKC inhibitor) and PD98059 [p44/p42 MAPK kinase (MEK) inhibitor] did not cause a significant change. To determine if signaling pathways triggered by Cz were involved in the T. cruzi growth, we studied the effect of those inhibitors. In Cz-treated cells--pre-incubated with TK, PKA or p38 MAPK inhibitors--the balance of NO/urea was biased towards NO, and the amastigote growth was diminished. Besides, genistein and mainly KT5720 induced down-regulation of arginase I expression in Cz-treated cells. Thus, activation of TK, PKA and p38 MAPK by Cz induces an increase of arginase activity in macrophages and the subsequent T. cruzi growth.


Subject(s)
Arginase/biosynthesis , Arginine/analogs & derivatives , Cell Division/drug effects , Enzyme Induction/physiology , Trypanosoma cruzi/enzymology , Animals , Arginase/antagonists & inhibitors , Arginine/pharmacology , Carbazoles/pharmacology , Cyclic AMP-Dependent Protein Kinases/antagonists & inhibitors , Cyclic AMP-Dependent Protein Kinases/metabolism , Cysteine Endopeptidases/pharmacology , Genistein/pharmacology , Growth Inhibitors/pharmacology , Imidazoles/pharmacology , Indoles/pharmacology , Mitogen-Activated Protein Kinases/metabolism , Protein-Tyrosine Kinases/antagonists & inhibitors , Protein-Tyrosine Kinases/metabolism , Protozoan Proteins , Pyridines/pharmacology , Pyrroles/pharmacology , Signal Transduction/drug effects , Trypanosoma cruzi/drug effects , Trypanosoma cruzi/growth & development , p38 Mitogen-Activated Protein Kinases
SELECTION OF CITATIONS
SEARCH DETAIL
...