Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
2.
Oncotarget ; 6(28): 25452-65, 2015 Sep 22.
Article in English | MEDLINE | ID: mdl-26327537

ABSTRACT

Targeted treatment of advanced melanoma could benefit from the precise molecular characterization of melanoma samples. Using a melanoma-specific selection of 217 genes, we performed targeted deep sequencing of a series of biopsies, from advanced melanoma cases, with a Breslow index of ≥ 4 mm, and/or with a loco-regional infiltration in lymph nodes or presenting distant metastasis, as well of a collection of human cell lines. This approach detected 3-4 mutations per case, constituting unique mutational signatures associated with specific inhibitor sensitivity. Functionally, case-specific combinations of inhibitors that simultaneously targeted MAPK-dependent and MAPK-independent mechanisms were most effective at inhibiting melanoma growth, against each specific mutational background. These observations were challenged by characterizing a freshly resected biopsy from a metastatic lesion located in the skin and soft tissue and by testing its associated therapy ex vivo and in vivo using melanocytes and patient-derived xenografted mice, respectively. The results show that upon mutational characterization of advanced melanoma patients, specific mutational profiles can be used for selecting drugs that simultaneously target several deregulated genes/pathways involved in tumor generation or progression.


Subject(s)
Antineoplastic Agents/therapeutic use , Biomarkers, Tumor/genetics , DNA Mutational Analysis/methods , Gene Expression Profiling/methods , Melanoma/drug therapy , Melanoma/genetics , Mutation , Precision Medicine , Skin Neoplasms/drug therapy , Skin Neoplasms/genetics , Animals , Biopsy , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Genetic Predisposition to Disease , Humans , Lymphatic Metastasis , Melanocytes/drug effects , Melanocytes/metabolism , Melanocytes/pathology , Melanoma/secondary , Mice, Inbred BALB C , Mice, Inbred NOD , Mice, Nude , Molecular Targeted Therapy , Patient Selection , Phenotype , Predictive Value of Tests , Protein Kinase Inhibitors/therapeutic use , Signal Transduction/drug effects , Skin Neoplasms/pathology , Time Factors , Xenograft Model Antitumor Assays
5.
PLoS One ; 10(3): e0119946, 2015.
Article in English | MEDLINE | ID: mdl-25775023

ABSTRACT

We have performed a comparative ultrasequencing study of multiple colorectal lesions obtained simultaneously from four patients. Our data show that benign lesions (adenomatous or hyperplastic polyps) contain a high mutational load. Additionally multiple synchronous colorectal lesions show non overlapping mutational signatures highlighting the degree of heterogeneity between multiple specimens in the same patient. Observations in these cases imply that considering not only the number of mutations but an effective oncogenic combination of mutations can determine the malignant progression of colorectal lesions.


Subject(s)
Adenocarcinoma/genetics , Adenoma/genetics , Clonal Evolution , Colorectal Neoplasms/genetics , Mutation , Adenocarcinoma/pathology , Adenoma/pathology , Aged , Aged, 80 and over , Colorectal Neoplasms/pathology , Female , Humans , Male
6.
Cancer Cell ; 25(6): 831-45, 2014 Jun 16.
Article in English | MEDLINE | ID: mdl-24882515

ABSTRACT

Mutually exclusive activating mutations in the GNAQ and GNA11 oncogenes, encoding heterotrimeric Gαq family members, have been identified in ∼ 83% and ∼ 6% of uveal and skin melanomas, respectively. However, the molecular events underlying these GNAQ-driven malignancies are not yet defined, thus limiting the ability to develop cancer-targeted therapies. Here, we focused on the transcriptional coactivator YAP, a critical component of the Hippo signaling pathway that controls organ size. We found that Gαq stimulates YAP through a Trio-Rho/Rac signaling circuitry promoting actin polymerization, independently of phospholipase Cß and the canonical Hippo pathway. Furthermore, we show that Gαq promotes the YAP-dependent growth of uveal melanoma cells, thereby identifying YAP as a suitable therapeutic target in uveal melanoma, a GNAQ/GNA11-initiated human malignancy.


Subject(s)
GTP Phosphohydrolases/metabolism , GTP-Binding Protein alpha Subunits/genetics , Melanoma/genetics , Nuclear Proteins/genetics , Protein Serine-Threonine Kinases/metabolism , Transcription Factors/genetics , Uveal Neoplasms/genetics , Animals , Cell Cycle Proteins , Cell Growth Processes/physiology , Cell Line, Tumor , Female , GTP Phosphohydrolases/genetics , GTP-Binding Protein alpha Subunits/metabolism , GTP-Binding Protein alpha Subunits, Gq-G11 , Gene Knockdown Techniques , HEK293 Cells , Heterografts , Hippo Signaling Pathway , Humans , Melanoma/enzymology , Melanoma/metabolism , Mice , Mice, Inbred NOD , Mutation , Nuclear Proteins/metabolism , Protein Serine-Threonine Kinases/genetics , Signal Transduction , Skin Neoplasms , Transcription Factors/metabolism , Transfection , Uveal Neoplasms/enzymology , Uveal Neoplasms/metabolism , rho GTP-Binding Proteins/genetics , rho GTP-Binding Proteins/metabolism , Melanoma, Cutaneous Malignant
7.
Blood ; 123(13): 2034-43, 2014 Mar 27.
Article in English | MEDLINE | ID: mdl-24497536

ABSTRACT

Cutaneous T-cell lymphoma (CTCL) is a heterogeneous group of primary cutaneous T-cell lymphoproliferative processes, mainly composed of mycosis fungoides and Sézary syndrome, the aggressive forms of which lack an effective treatment. The molecular pathogenesis of CTCL is largely unknown, although neoplastic cells show increased signaling from T-cell receptors (TCRs). DNAs from 11 patients with CTCL, both normal and tumoral, were target-enriched and sequenced by massive parallel sequencing for a selection of 524 TCR-signaling-related genes. Identified variants were validated by capillary sequencing. Multiple mutations were found that affected several signaling pathways, such as TCRs, nuclear factor κB, or Janus kinase/signal transducer and activator of transcription, but PLCG1 was found to be mutated in 3 samples, 2 of which featured a redundant mutation (c.1034T>C, S345F) in exon 11 that affects the PLCx protein catalytic domain. This mutation was further analyzed by quantitative polymerase chain reaction genotyping in a new cohort of 42 patients with CTCL, where it was found in 19% of samples. Immunohistochemical analysis for nuclear factor of activated T cells (NFAT) showed that PLCG1-mutated cases exhibited strong NFAT nuclear immunostaining. Functional studies demonstrated that PLCG1 mutants elicited increased downstream signaling toward NFAT activation, and inhibition of this pathway resulted in reduced CTCL cell proliferation and cell viability. Thus, increased proliferative and survival mechanisms in CTCL may partially depend on the acquisition of somatic mutations in PLCG1 and other genes that are essential for normal T-cell differentiation.


Subject(s)
Lymphoma, T-Cell/genetics , Mutation , Phospholipase C gamma/genetics , Skin Neoplasms/genetics , Animals , Cell Line, Tumor , Cell Survival/genetics , Cohort Studies , DNA Mutational Analysis , Female , High-Throughput Nucleotide Sequencing , Humans , Lymphoma, T-Cell/pathology , Male , Mice , NIH 3T3 Cells , Skin Neoplasms/pathology
8.
Haematologica ; 99(2): 222-31, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24497559

ABSTRACT

B-cell lymphomas comprise an increasing number of clinicopathological entities whose characterization has historically been based mainly on histopathological features. In recent decades, the analysis of chromosomal aberrations as well as gene and miRNA expression profile studies have helped distinguish particular tumor types and also enabled the detection of a number of targets with therapeutic implications, such as those activated downstream of the B-cell receptor. Our ability to identify the mechanisms involved in B-cell lymphoma pathogenesis has been boosted recently through the use of Next Generation Sequencing techniques in the analysis of human cancer. This work summarizes the recent findings in the molecular pathogenesis of B-cell neoplasms with special focus on those clinically relevant somatic mutations with the potential to be explored as candidates for the development of new targeted therapies. Our work includes a comparison between the mutational indexes and ranges observed in B-cell lymphomas and also with other solid tumors and describes the most striking mutational data for the major B-cell neoplasms. This review describes a highly dynamic field that currently offers many opportunities for personalized therapy, although there is still much to be gained from the further molecular characterization of these clinicopathological entities.


Subject(s)
Chromosome Aberrations , Gene Expression Regulation, Neoplastic , Lymphoma, B-Cell , MicroRNAs , RNA, Neoplasm , Humans , Lymphoma, B-Cell/diagnosis , Lymphoma, B-Cell/genetics , Lymphoma, B-Cell/metabolism , Lymphoma, B-Cell/therapy , MicroRNAs/biosynthesis , MicroRNAs/genetics , RNA, Neoplasm/biosynthesis , RNA, Neoplasm/genetics
9.
Mol Cell ; 49(1): 94-108, 2013 Jan 10.
Article in English | MEDLINE | ID: mdl-23177739

ABSTRACT

Activating mutations in GNAQ and GNA11, encoding members of the Gα(q) family of G protein α subunits, are the driver oncogenes in uveal melanoma, and mutations in Gq-linked G protein-coupled receptors have been identified recently in numerous human malignancies. How Gα(q) and its coupled receptors transduce mitogenic signals is still unclear because of the complexity of signaling events perturbed upon Gq activation. Using a synthetic-biology approach and a genome-wide RNAi screen, we found that a highly conserved guanine nucleotide exchange factor, Trio, is essential for activating Rho- and Rac-regulated signaling pathways acting on JNK and p38, and thereby transducing proliferative signals from Gα(q) to the nucleus independently of phospholipase C-ß. Indeed, whereas many biological responses elicited by Gq depend on the transient activation of second-messenger systems, Gq utilizes a hard-wired protein-protein-interaction-based signaling circuitry to achieve the sustained stimulation of proliferative pathways, thereby controlling normal and aberrant cell growth.


Subject(s)
Guanine Nucleotide Exchange Factors/physiology , Mitosis , Protein Serine-Threonine Kinases/physiology , Receptors, G-Protein-Coupled/metabolism , Signal Transduction , Transcription Factor AP-1/metabolism , rho GTP-Binding Proteins/metabolism , Animals , Cell Line, Tumor , Cell Proliferation , Clozapine/analogs & derivatives , Clozapine/pharmacology , Drosophila/genetics , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Enzyme Activation , Female , GTP-Binding Protein alpha Subunits/metabolism , GTP-Binding Protein alpha Subunits, Gq-G11 , Gene Knockdown Techniques , Guanine Nucleotide Exchange Factors/metabolism , Humans , Mice , Mice, Nude , Mitogen-Activated Protein Kinases/metabolism , Mitogens/pharmacology , NIH 3T3 Cells , Neoplasm Transplantation , Neoplasms/pathology , Protein Serine-Threonine Kinases/metabolism , RNA Interference , Receptors, G-Protein-Coupled/genetics
10.
Medicina (B Aires) ; 72(4): 315-20, 2012.
Article in Spanish | MEDLINE | ID: mdl-22892083

ABSTRACT

In C4-HD murine mammary carcinomas and in human breast cancer T47D cells, we showed that medroxyprogesterone acetate (MPA) induces a nuclear physical association between estrogen receptor alpha (ERa) and progesterone receptors (PR). The blockade of ERa inhibits cell proliferation mediated by progestins. We hypothesized that this nuclear association between ERa/PR is necessary to trigger progestin-induced cell proliferation and tumor growth. We demonstrated that fulvestrant (FUL, ICI182.780) induced complete regression of C4-HD tumors growing with progestins. MPA treatment induced an early increase in both CCND1 and MYC expression in T47D cells. The blockade of ERa prevented the MPA-dependent transcription of both genes. Specific binding of PR/ERa was observed at the same MPA-sensitive regions at the CCND1 and MYC gene promoters after chromatin immunoprecipitation (ChIP) analysis. ICI inhibited binding of ERa to both gene regulatory sequences while PR binding was unaffected. The nuclear colocalization between both receptors in T47D cells was confirmed by: confocal microscopy, Duolink assays and co-immunoprecipitation assays. In breast cancer samples we also observed a nuclear interaction between both steroid receptors. Our results indicate that the presence of ERa interacting with activated PR at the CCND1 and MYC promoters is required to trigger progestin-induced gene transcription and cell proliferation in breast cancer cells.


Subject(s)
Carcinoma/pathology , Estradiol/analogs & derivatives , Estrogen Receptor alpha/physiology , Mammary Neoplasms, Experimental/pathology , Receptors, Progesterone/physiology , Animals , Antineoplastic Agents, Hormonal/pharmacology , Breast Neoplasms/genetics , Carcinoma/chemically induced , Carcinoma/drug therapy , Cell Proliferation , Chromatin Immunoprecipitation , Cyclin D1/metabolism , Estradiol/administration & dosage , Estrogen Receptor alpha/drug effects , Female , Fulvestrant , Genes, myc , Humans , Mammary Neoplasms, Experimental/chemically induced , Mammary Neoplasms, Experimental/drug therapy , Medroxyprogesterone Acetate/pharmacology , Murinae , Progestins/metabolism , Receptors, Progesterone/drug effects , Transcription, Genetic
11.
Cancer Res ; 72(9): 2416-27, 2012 May 01.
Article in English | MEDLINE | ID: mdl-22396492

ABSTRACT

Synthetic progesterone used in contraception drugs (progestins) can promote breast cancer growth, but the mechanisms involved are unknown. Moreover, it remains unclear whether cytoplasmic interactions between the progesterone receptor (PR) and estrogen receptor alpha (ERα) are required for PR activation. In this study, we used a murine progestin-dependent tumor to investigate the role of ERα in progestin-induced tumor cell proliferation. We found that treatment with the progestin medroxyprogesterone acetate (MPA) induced the expression and activation of ERα, as well as rapid nuclear colocalization of activated ERα with PR. Treatment with the pure antiestrogen fulvestrant to block ERα disrupted the interaction of ERα and PR in vitro and induced the regression of MPA-dependent tumor growth in vivo. ERα blockade also prevented an MPA-induced increase in CYCLIN D1 (CCND1) and MYC expression. Chromatin immunoprecipitation studies showed that MPA triggered binding of ERα and PR to the CCND1 and MYC promoters. Interestingly, blockade or RNAi-mediated silencing of ERα inhibited ERα, but not PR binding to both regulatory sequences, indicating that an interaction between ERα and PR at these sites is necessary for MPA-induced gene expression and cell proliferation. We confirmed that nuclear colocalization of both receptors also occurred in human breast cancer samples. Together, our findings argued that ERα-PR association on target gene promoters is essential for progestin-induced cell proliferation.


Subject(s)
Breast Neoplasms/pathology , Cyclin D1/genetics , Estrogen Receptor alpha/metabolism , Genes, myc , Mammary Neoplasms, Experimental/pathology , Receptors, Progesterone/metabolism , Animals , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Cell Growth Processes/genetics , Cell Nucleus/genetics , Cell Nucleus/metabolism , Estradiol/analogs & derivatives , Estradiol/pharmacology , Estrogen Receptor alpha/antagonists & inhibitors , Estrogen Receptor alpha/genetics , Female , Fulvestrant , Gene Expression Regulation, Neoplastic/drug effects , Genes, Reporter , Humans , Mammary Neoplasms, Experimental/genetics , Mammary Neoplasms, Experimental/metabolism , Medroxyprogesterone Acetate/pharmacology , Mice , Mice, Inbred BALB C , Neoplasms, Hormone-Dependent/genetics , Neoplasms, Hormone-Dependent/metabolism , Neoplasms, Hormone-Dependent/pathology , Promoter Regions, Genetic , Receptors, Progesterone/genetics
12.
J Periodontol ; 83(7): 948-54, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22181687

ABSTRACT

BACKGROUND: Interleukin-21 (IL-21) controls the differentiation of T-helper Th17 cells and induces the production of IL-17 in this T-cell subtype. The aim of this study is to determine the relative expression of IL-21 in gingival tissues of chronic periodontitis patients and correlate/associate this expression with proinflammatory cytokines and clinical parameters of disease. METHODS: Samples of gingival biopsies were collected from chronic periodontitis patients (n = 10) and controls (n = 8). The mRNA expressions of IL-21, IL-1ß, IL-6, IL-17, IL-23, IL-10, and transforming growth factor-ß1 (TGF-ß1) were quantified using real-time reverse transcription-polymerase chain reaction. IL-21 levels were compared between chronic periodontitis and healthy gingival tissues and correlated with cytokine and clinical parameters of tissue destruction. RESULTS: A significant overexpression of IL-21, IL-1ß, IL-6, IL-17, and IL-23p19 was detected in periodontal disease-affected tissues compared to healthy gingival tissues. IL-10 and TGF-ß1 were, however, downregulated in periodontal lesions. IL-21 yielded significant positive correlations with probing depth, clinical attachment level, IL-1ß, and IL-6. In addition, IL-21 was negatively correlated with IL-10 and TGF-ß1. CONCLUSIONS: IL-21 was overexpressed in chronic periodontitis gingival tissues and correlated with clinical parameters of periodontal destruction and with proinflammatory cytokines. Therefore, IL-21 might play a role in the tissue destruction that characterizes chronic periodontal disease.


Subject(s)
Chronic Periodontitis/immunology , Cytokines/analysis , Inflammation Mediators/analysis , Interleukins/analysis , Adult , Dental Plaque Index , Female , Gingiva/immunology , Humans , Interleukin-10/analysis , Interleukin-17/analysis , Interleukin-1beta/analysis , Interleukin-23/analysis , Interleukin-23 Subunit p19/analysis , Interleukin-6/analysis , Male , Middle Aged , Periodontal Attachment Loss/immunology , Periodontal Index , Periodontal Pocket/immunology , T-Lymphocytes, Regulatory/immunology , Th17 Cells/immunology , Transforming Growth Factor beta1/analysis
13.
Cancer Res ; 71(10): 3720-31, 2011 May 15.
Article in English | MEDLINE | ID: mdl-21464042

ABSTRACT

Fibroblast growth factor (FGF) receptor 2 (FGFR-2) polymorphisms have been associated with an increase in estrogen receptor and progesterone receptor (PR)-positive breast cancer risk; however, a clear mechanistic association between FGFR-2 and steroid hormone receptors remains elusive. In previous works, we have shown a cross talk between FGF2 and progestins in mouse mammary carcinomas. To investigate the mechanisms underlying these interactions and to validate our findings in a human setting, we have used T47D human breast cancer cells and human cancer tissue samples. We showed that medroxyprogesterone acetate (MPA) and FGF2 induced cell proliferation and activation of ERK, AKT, and STAT5 in T47D and in murine C4-HI cells. Nuclear interaction between PR, FGFR-2, and STAT5 after MPA and FGF2 treatment was also showed by confocal microscopy and immunoprecipitation. This effect was associated with increased transcription of PRE and/or GAS reporter genes, and of PR/STAT5-regulated genes and proteins. Two antiprogestins and the FGFR inhibitor PD173074, specifically blocked the effects induced by FGF2 or MPA respectively. The presence of PR/FGFR-2/STAT5 complexes bound to the PRE probe was corroborated by using NoShift transcription and chromatin immunoprecipitation of the MYC promoter. Additionally, we showed that T47D cells stably transfected with constitutively active FGFR-2 gave rise to invasive carcinomas when transplanted into NOD/SCID mice. Nuclear colocalization between PR and FGFR-2/STAT5 was also observed in human breast cancer tissues. This study represents the first demonstration of a nuclear interaction between FGFR-2 and STAT5, as PR coactivators at the DNA progesterone responsive elements, suggesting that FGFRs are valid therapeutic targets for human breast cancer treatment.


Subject(s)
Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Gene Expression Regulation, Neoplastic , Receptor, Fibroblast Growth Factor, Type 2/metabolism , Receptors, Progesterone/metabolism , STAT5 Transcription Factor/metabolism , Animals , Antineoplastic Agents, Hormonal/pharmacology , Cell Line, Tumor , Extracellular Signal-Regulated MAP Kinases/metabolism , Female , Humans , Medroxyprogesterone Acetate/pharmacology , Mice , Mice, Inbred BALB C , Mice, Inbred NOD , Mice, SCID , Neoplasm Transplantation
14.
Mol Cancer Res ; 9(5): 564-76, 2011 May.
Article in English | MEDLINE | ID: mdl-21460180

ABSTRACT

Untreated chronic myeloid leukemia (CML) progresses from chronic phase to blastic crisis (BC). Increased genomic instability, deregulated proliferation, and loss of differentiation appear associated to BC, but the molecular alterations underlying the progression of CML are poorly characterized. MYC oncogene is frequently deregulated in human cancer, often associated with tumor progression. Genomic instability and induction of aberrant DNA replication are described as effects of MYC. In this report, we studied MYC activities in CML cell lines with conditional MYC expression with and without exposure to imatinib, the front-line drug in CML therapy. In cells with conditional MYC expression, MYC did not rescue the proliferation arrest mediated by imatinib but provoked aberrant DNA synthesis and accumulation of cells with 4C content. We studied MYC mRNA expression in 66 CML patients at different phases of the disease, and we found that MYC expression was higher in CML patients at diagnosis than control bone marrows or in patients responding to imatinib. Further, high MYC levels at diagnosis correlated with a poor response to imatinib. MYC expression did not directly correlate with BCR-ABL levels in patients treated with imatinib. Overall our study suggests that, as in other tumor models, MYC-induced aberrant DNA synthesis in CML cells is consistent with MYC overexpression in untreated CML patients and nonresponding patients and supports a role for MYC in CML progression, possibly through promotion of genomic instability.


Subject(s)
DNA Replication/drug effects , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics , Piperazines/therapeutic use , Proto-Oncogene Proteins c-myc/metabolism , Pyrimidines/therapeutic use , Antineoplastic Agents/therapeutic use , Benzamides , Cell Line, Tumor , Disease Progression , Fusion Proteins, bcr-abl/metabolism , Genomic Instability/genetics , Humans , Imatinib Mesylate , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology , Protein Kinase Inhibitors/therapeutic use , Protein-Tyrosine Kinases/metabolism , Proto-Oncogene Proteins c-myc/genetics
15.
Nat Immunol ; 12(1): 86-95, 2011 Jan.
Article in English | MEDLINE | ID: mdl-21131965

ABSTRACT

The molecular mechanisms that direct transcription of the gene encoding the transcription factor Foxp3 in CD4(+) T cells remain ill-defined. We show here that deletion of the DNA-binding inhibitor Id3 resulted in the defective generation of Foxp3(+) regulatory T cells (T(reg) cells). We identify two transforming growth factor-ß1 (TGF-ß1)-dependent mechanisms that were vital for activation of Foxp3 transcription and were defective in Id3(-/-) CD4(+) T cells. Enhanced binding of the transcription factor E2A to the Foxp3 promoter promoted Foxp3 transcription. Id3 was required for relief of inhibition by the transcription factor GATA-3 at the Foxp3 promoter. Furthermore, Id3(-/-) T cells showed greater differentiation into the T(H)17 subset of helper T cells in vitro and in a mouse asthma model. Therefore, a network of factors acts in a TGF-ß-dependent manner to control Foxp3 expression and inhibit the development of T(H)17 cells.


Subject(s)
Asthma/metabolism , Forkhead Transcription Factors/metabolism , Inhibitor of Differentiation Proteins/metabolism , T-Lymphocytes, Regulatory/metabolism , Th17 Cells/metabolism , Animals , Asthma/chemically induced , Asthma/genetics , Asthma/immunology , Basic Helix-Loop-Helix Transcription Factors/metabolism , Cell Differentiation/genetics , Cells, Cultured , Disease Models, Animal , Forkhead Transcription Factors/genetics , Inhibitor of Differentiation Proteins/genetics , Inhibitor of Differentiation Proteins/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Promoter Regions, Genetic/genetics , Protein Binding/genetics , Sequence Deletion/genetics , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/pathology , Th17 Cells/immunology , Th17 Cells/pathology , Transcriptional Activation/genetics , Transforming Growth Factor beta1/metabolism
16.
Methods Mol Biol ; 661: 137-50, 2010.
Article in English | MEDLINE | ID: mdl-20811981

ABSTRACT

A complex intracellular signaling network mediates the multiple biological activities of G-protein-coupled receptors (GPCRs). Among them, monomeric GTPases and a family of closely related proline-targeted serine-threonine kinases, collectively known as Mitogen-Activated Protein Kinases (MAPKs), appears to play central roles in orchestrating the proliferative responses to multiple mitogens that act on GPCRs. Upon GDP/GTP exchange, monomeric GTPases control the phosphorylation of conserved threonine and tyrosine residues in MAPKs by their immediate upstream kinases, increasing their enzymatic activity and inducing their translocation to the nucleus where they phosphorylate transcription factors, thereby regulating the expression of genes playing a key role in normal and aberrant cell growth. Recently, a number of GPCRs have been engineered to provide exclusive activation by synthetic drug-like compounds while becoming insensitive to endogenous ligands. These engineered receptors, named Receptors Activated Solely by Synthetic Ligands (RASSLs), promise better understanding of GPCRs signaling in vitro and in vivo, thus representing ideal tools to selectively modulate MAPK signaling routes controlling a wide range of biological functions, from proliferation to differentiation, migration, invasion, and cell survival or death by apoptosis.


Subject(s)
Mitogen-Activated Protein Kinases/metabolism , Receptors, G-Protein-Coupled/metabolism , ras Proteins/metabolism , rho GTP-Binding Proteins/metabolism , Amino Acid Motifs , Antibodies/immunology , Antibody Specificity , Blotting, Western , Cell Line , Conserved Sequence , Enzyme Activation , Guanosine Triphosphate/metabolism , Humans , Mitogen-Activated Protein Kinases/chemistry , Mitogen-Activated Protein Kinases/immunology , Phosphoproteins/immunology , Phosphorylation
17.
J Biol Chem ; 284(32): 21139-56, 2009 Aug 07.
Article in English | MEDLINE | ID: mdl-19509292

ABSTRACT

The TP73 gene gives rise to transactivation domain-p73 isoforms (TAp73) as well as DeltaNp73 variants with a truncated N terminus. Although TAp73alpha and -beta proteins are capable of inducing cell cycle arrest, apoptosis, and differentiation, DeltaNp73 acts in many cell types as a dominant-negative repressor of p53 and TAp73. It has been proposed that p73 is involved in myeloid differentiation, and its altered expression is involved in leukemic degeneration. However, there is little evidence as to which p73 variants (TA or DeltaN) are expressed during differentiation and whether specific p73 isoforms have the capacity to induce, or hinder, this differentiation in leukemia cells. In this study we identify GATA1 as a direct transcriptional target of TAp73alpha. Furthermore, TAp73alpha induces GATA1 activity, and it is required for erythroid differentiation. Additionally, we describe a functional cooperation between TAp73 and DeltaNp73 in the context of erythroid differentiation in human myeloid cells, K562 and UT-7. Moreover, the impaired expression of GATA1 and other erythroid genes in the liver of p73KO embryos, together with the moderated anemia observed in p73KO young mice, suggests a physiological role for TP73 in erythropoiesis.


Subject(s)
DNA-Binding Proteins/physiology , Erythrocytes/metabolism , GATA1 Transcription Factor/physiology , Gene Expression Regulation, Neoplastic , Nuclear Proteins/physiology , Tumor Suppressor Proteins/physiology , Animals , Apoptosis , Cell Differentiation , DNA-Binding Proteins/biosynthesis , Erythropoiesis , GATA1 Transcription Factor/biosynthesis , Gene Expression Regulation, Developmental , Humans , K562 Cells , Liver/embryology , Liver/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Nuclear Proteins/biosynthesis , Tumor Protein p73 , Tumor Suppressor Proteins/biosynthesis
18.
Mol Cancer Res ; 6(2): 325-39, 2008 Feb.
Article in English | MEDLINE | ID: mdl-18314492

ABSTRACT

Although mutant Ras proteins were originally described as transforming oncoproteins, they induce growth arrest, senescence, and/or differentiation in many cell types. c-Myc is an oncogenic transcription factor that cooperates with Ras in cellular transformation and oncogenesis. However, the Myc-Ras relationship in cellular differentiation is largely unknown. Here, we have analyzed the effects of c-Myc on PC12-derived cells (UR61 cell line), harboring an inducible N-Ras oncogene. In these cells, Ras activation induces neuronal-like differentiation by a process involving c-Jun activation. We found that c-Myc inhibited Ras-mediated differentiation by a mechanism that involves the blockade of c-Jun induction in response to Ras signal. Accordingly, ectopically expressed c-Jun could bypass c-Myc impediment of Ras-induced differentiation and activator protein 1 activation. Interestingly, it did not rescue the proliferative arrest elicited by Ras and did not enhance the differentiation-associated apoptosis. The blockade of Ras-mediated induction of c-Jun takes place at the level of c-Jun proximal promoter. Mutational analysis revealed that c-Myc regions involved in DNA binding and transactivation are required to block differentiation and c-Jun induction. c-Myc does not seem to require Miz-1 to inhibit differentiation and block c-Jun induction. Furthermore, Max is not required for c-Myc activity, as UR61 cells lack a functional Max gene. c-Myc-inhibitory effect on the Ras/c-Jun connection is not restricted to UR61 cells as it can occur in other cell types as K562 or HEK293. In conclusion, we describe a novel interplay between c-Myc and c-Jun that controls the ability of Ras to trigger the differentiation program of pheochromocytoma cells.


Subject(s)
Cell Differentiation , Pheochromocytoma/metabolism , Pheochromocytoma/pathology , Proto-Oncogene Proteins c-jun/genetics , Proto-Oncogene Proteins c-myc/metabolism , Up-Regulation , ras Proteins/antagonists & inhibitors , Animals , Cell Proliferation , Enzyme Activation , Enzyme Induction , Gene Expression Regulation, Neoplastic , Gene Silencing , Humans , PC12 Cells , Pheochromocytoma/enzymology , Promoter Regions, Genetic/genetics , Protein Structure, Tertiary , Proto-Oncogene Proteins c-myc/chemistry , Rats , Transcription Factor AP-1/metabolism , Transcriptional Activation/genetics , ras Proteins/biosynthesis , ras Proteins/metabolism
19.
Mol Cell Neurosci ; 37(3): 622-33, 2008 Mar.
Article in English | MEDLINE | ID: mdl-18255312

ABSTRACT

Nuclear aggregation of the expanded polyalanine tract in the poly(A)-binding protein nuclear 1 (PABPN1) is the pathological hallmark of oculopharyngeal muscular dystrophy. However, wild type PABPN1 aggregates into nuclear inclusion in oxytocin-producing neurons under physiological conditions. In this study we have analyzed the nuclear organization and dynamics of PABPN1 inclusions in oxytocin-producing neurons. We demonstrated that PABPN1 inclusions represent a distinct compartment of the interchromatin region. They establish a spatial relationship with nuclear speckles, Cajal bodies and clastosomes. PABPN1 inclusions accumulate poly(A) RNA, but do not concentrate highly expressed mRNAs in oxytocin producing neurons and the mRNA-binding proteins hnRNP C, Y14 and REF. PABPN1 inclusions are dynamic structures that appear during the postnatal period and their number decrease in response to the activation of transcription. Our results support that the RNA retained in the PABPN1 inclusions is a noncoding regulatory RNA involved in some aspects of nuclear RNA metabolism.


Subject(s)
Cell Nucleus/metabolism , Gene Expression Regulation/physiology , Intranuclear Inclusion Bodies/metabolism , Neurons/ultrastructure , Poly(A)-Binding Protein I/metabolism , Supraoptic Nucleus/cytology , Age Factors , Animals , Animals, Newborn , Histones/metabolism , Microscopy, Electron, Transmission/methods , Nonlinear Dynamics , Nuclear Proteins/metabolism , Osmotic Pressure , Oxytocin/metabolism , RNA, Messenger/metabolism , Rats , Rats, Wistar , Ribonucleoproteins, Small Nucleolar/metabolism , Supraoptic Nucleus/growth & development
20.
Chromosoma ; 116(5): 441-51, 2007 Oct.
Article in English | MEDLINE | ID: mdl-17549507

ABSTRACT

The neuron-like UR61 cell is a stable PC12 subline that contains a mouse N-ras oncogene. Dexamethasone (Dex) treatment induces a neuron-like differentiation, which is associated with neuritogenesis and nuclear expression of the glucocorticoid receptor and c-Jun. In differentiated UR61 cells, small ubiquitin-like modifiers 1 (SUMO-1) is concentrated in a new category of SUMO-1 nuclear bodies (SNBs) distinct from promyelocytic leukemia (PML) bodies by their large size and absence of PML protein. SNBs are 1 to 3 mum in diameter and exhibit a fine granular texture by electron microscopy. They are free of splicing factors and transcription foci and show spatial associations with Cajal bodies. In addition to SUMO-1 and the E2-conjugating enzyme Ubc9, which is essential for sumoylation, SNBs concentrate the transcriptional regulators CBP, CREB, and c-Jun. Moreover, transfection experiments demonstrate that SNBs accumulate the active conjugating form of SUMO-1 but not the conjugation defective variant of SUMO-1, supporting that SNBs are sites of sumoylation. Our results suggest that SNBs play a role in the control of the nucleoplasmic concentration of transcription regulators involved in neuroprotection and survival of the UR61 cells.


Subject(s)
CREB-Binding Protein/metabolism , JNK Mitogen-Activated Protein Kinases/metabolism , Neurons/metabolism , SUMO-1 Protein/isolation & purification , Animals , Cell Nucleolus/metabolism , Cell Nucleolus/ultrastructure , Cells, Cultured , Dexamethasone , Gene Expression Regulation , Neurons/chemistry , PC12 Cells , Rats , SUMO-1 Protein/chemistry , SUMO-1 Protein/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...