Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters










Publication year range
1.
NAR Cancer ; 6(2): zcae024, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38751936

ABSTRACT

In this review, we explore the transformative impact of next generation sequencing technologies in the realm of translatomics (the study of how translational machinery acts on a genome-wide scale). Despite the expectation of a direct correlation between mRNA and protein content, the complex regulatory mechanisms that affect this relationship remark the limitations of standard RNA-seq approaches. Then, the review characterizes crucial techniques such as polysome profiling, ribo-seq, trap-seq, proximity-specific ribosome profiling, rnc-seq, tcp-seq, qti-seq and scRibo-seq. All these methods are summarized within the context of cancer research, shedding light on their applications in deciphering aberrant translation in cancer cells. In addition, we encompass databases and bioinformatic tools essential for researchers that want to address translatome analysis in the context of cancer biology.

2.
Life Sci Alliance ; 7(3)2024 Mar.
Article in English | MEDLINE | ID: mdl-38176727

ABSTRACT

Activating transcription factor 4 (Atf4), which is modulated by the protein kinase RNA-like ER kinase (PERK), is a stress-induced transcription factor responsible for controlling the expression of a wide range of adaptive genes, enabling cells to withstand stressful conditions. However, the impact of the Atf4 signaling pathway on airway regeneration remains poorly understood. In this study, we used mouse airway epithelial cell culture models to investigate the role of PERK/Atf4 in respiratory tract differentiation. Through pharmacological inhibition and silencing of ATF4, we uncovered the crucial involvement of PERK/Atf4 in the differentiation of basal stem cells, leading to a reduction in the number of secretory cells. ChIP-seq analysis revealed direct binding of ATF4 to regulatory elements of genes associated with osteoblast differentiation and secretory cell function. Our findings provide valuable insights into the role of ATF4 in airway epithelial differentiation and its potential involvement in innate immune responses and cellular adaptation to stress.


Subject(s)
Endoplasmic Reticulum Stress , eIF-2 Kinase , Animals , Mice , eIF-2 Kinase/genetics , Endoplasmic Reticulum Stress/genetics , Activating Transcription Factor 4/genetics , Activating Transcription Factor 4/metabolism , Cell Differentiation/genetics , Respiratory System/metabolism
3.
Genome Biol ; 19(1): 55, 2018 04 25.
Article in English | MEDLINE | ID: mdl-29695303

ABSTRACT

BACKGROUND: Animals can show very different behaviors even in isogenic populations, but the underlying mechanisms to generate this variability remain elusive. We use the zebrafish (Danio rerio) as a model to test the influence of histone modifications on behavior. RESULTS: We find that laboratory and isogenic zebrafish larvae show consistent individual behaviors when swimming freely in identical wells or in reaction to stimuli. This behavioral inter-individual variability is reduced when we impair the histone deacetylation pathway. Individuals with high levels of histone H4 acetylation, and specifically H4K12, behave similarly to the average of the population, but those with low levels deviate from it. More precisely, we find a set of genomic regions whose histone H4 acetylation is reduced with the distance between the individual and the average population behavior. We find evidence that this modulation depends on a complex of Yin-yang 1 (YY1) and histone deacetylase 1 (HDAC1) that binds to and deacetylates these regions. These changes are not only maintained at the transcriptional level but also amplified, as most target regions are located near genes encoding transcription factors. CONCLUSIONS: We suggest that stochasticity in the histone deacetylation pathway participates in the generation of genetic-independent behavioral inter-individual variability.


Subject(s)
Biological Variation, Population , Histone Code , Acetylation , Animals , Biological Variation, Population/genetics , Gene Expression , Histone Deacetylase 1/metabolism , Histones/metabolism , Larva/genetics , Larva/metabolism , Larva/physiology , Swimming , YY1 Transcription Factor/metabolism , Zebrafish/genetics , Zebrafish/growth & development , Zebrafish/metabolism , Zebrafish/physiology , Zebrafish Proteins/metabolism
4.
Pharmacol Ther ; 185: 50-63, 2018 05.
Article in English | MEDLINE | ID: mdl-29258844

ABSTRACT

The aryl hydrocarbon receptor (AhR) is well-known for its major contributions to the cellular responses against environmental toxins and carcinogens. Notably, AhR has also emerged as a key transcription factor controlling many physiological processes including cell proliferation and apoptosis, differentiation, adhesion and migration, pluripotency and stemness. These novel functions have broadened our understanding of the signalling pathways and molecular intermediates interacting with AhR under both homeostatic and pathological conditions. Recent discoveries link AhR with the function of essential organs such as liver, skin and gonads, and with complex organismal structures including the immune and cardiovascular systems. The identification of potential endogenous ligands able to regulate AhR activity, opens the possibility of designing ad hoc molecules with pharmacological and/or therapeutic value to treat human diseases in which AhR may have a causal role. Integration of experimental data from in vitro and in vivo studies with "omic" analyses of human patients affected with cancer, immune diseases, inflammation or neurological disorders will likely contribute to validate the clinical relevance of AhR and the possible benefits of modulating its activity by pharmacologically-driven strategies. In this review, we will highlight signalling pathways involved in human diseases that could be targetable by AhR modulators and discuss the feasibility of using such molecules in therapy. The pros and cons of AhR-aimed approaches will be also mentioned.


Subject(s)
Receptors, Aryl Hydrocarbon/metabolism , Animals , Epigenesis, Genetic , Genetic Variation , Humans , Neoplasms/genetics , Receptors, Aryl Hydrocarbon/genetics , Signal Transduction
5.
Cell Commun Signal ; 12: 57, 2014 Sep 21.
Article in English | MEDLINE | ID: mdl-25238970

ABSTRACT

BACKGROUND: Adhesion and migration are relevant physiological functions that must be regulated by the cell under both normal and pathological conditions. The dioxin receptor (AhR) has emerged as a transcription factor regulating both processes in mesenchymal, epithelial and endothelial cells. Indirect results suggest that AhR could cooperate not only with additional transcription factors but also with membrane-associated proteins to drive such processes. RESULTS: In this study, we have used immortalized and primary dermal fibroblasts from wild type (AhR+/+) and AhR-null (AhR-/-) mice to show that AhR modulates membrane distribution and mobilization of caveolin-1 (Cav-1) during directional cell migration. AhR co-immunoprecipitated with Cav-1 and a fraction of both proteins co-localized to detergent-resistant membrane microdomains (DRM). Consistent with a role of AhR in the process, AhR-/- cells had a significant reduction in Cav-1 in DRMs. Moreover, high cell density reduced AhR nuclear levels and moved Cav-1 from DRMs to the soluble membrane in AhR+/+ but not in AhR-/- cells. Tyrosine-14 phosphorylation had a complex role in the mechanism since its upregulation reduced Cav-1 in DRMs in both AhR+/+ and AhR-/-cells, despite the lower basal levels of Y14-Cav-1 in the null cells. Fluorescence recovery after photobleaching revealed that AhR knock-down blocked Cav-1 transport to the plasma membrane, a deficit possibly influencing its depleted levels in DRMs. Membrane distribution of Cav-1 in AhR-null fibroblasts correlated with higher levels of cholesterol and with disrupted membrane microdomains, whereas addition of exogenous cholesterol changed the Cav-1 distribution of AhR+/+ cells to the null phenotype. Consistently, higher cholesterol levels enhanced caveolae-dependent endocytosis in AhR-null cells. CONCLUSIONS: These results suggest that AhR modulates Cav-1 distribution in migrating cells through the control of cholesterol-enriched membrane microdomains. Our study also supports the likely possibility of membrane-related, transcription factor independent, functions of AhR.


Subject(s)
Caveolin 1/metabolism , Cell Movement/physiology , Cholesterol/metabolism , Fibroblasts/metabolism , Receptors, Aryl Hydrocarbon/metabolism , Animals , Cells, Cultured , Endocytosis , Fibroblasts/physiology , Mice , Mice, Knockout , Receptors, Aryl Hydrocarbon/genetics
6.
EMBO J ; 32(15): 2125-39, 2013 Jul 31.
Article in English | MEDLINE | ID: mdl-23749212

ABSTRACT

Polarized epithelial cells take up nutrients from the blood through receptors that are endocytosed and recycle back to the basolateral plasma membrane (PM) utilizing the epithelial-specific clathrin adaptor AP-1B. Some native epithelia lack AP-1B and therefore recycle cognate basolateral receptors to the apical PM, where they carry out important functions for the host organ. Here, we report a novel transcytotic pathway employed by AP-1B-deficient epithelia to relocate AP-1B cargo, such as transferrin receptor (TfR), to the apical PM. Lack of AP-1B inhibited basolateral recycling of TfR from common recycling endosomes (CRE), the site of function of AP-1B, and promoted its transfer to apical recycling endosomes (ARE) mediated by the plus-end kinesin KIF16B and non-centrosomal microtubules, and its delivery to the apical membrane mediated by the small GTPase rab11a. Hence, our experiments suggest that the apical recycling pathway of epithelial cells is functionally equivalent to the rab11a-dependent TfR recycling pathway of non-polarized cells. They define a transcytotic pathway important for the physiology of native AP-1B-deficient epithelia and report the first microtubule motor involved in transcytosis.


Subject(s)
Adaptor Protein Complex 1 , Endosomes/metabolism , Epithelial Cells/metabolism , Kinesins/metabolism , Microtubules/metabolism , Receptors, Transferrin/metabolism , Transcytosis , Animals , CHO Cells , Cricetinae , Cricetulus , Dogs , Endosomes/genetics , Epithelial Cells/cytology , Humans , Kinesins/genetics , Madin Darby Canine Kidney Cells , Microtubules/genetics , Receptors, Transferrin/genetics , rab GTP-Binding Proteins/genetics , rab GTP-Binding Proteins/metabolism
7.
Cell Signal ; 25(4): 848-59, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23333462

ABSTRACT

Recent studies have suggested a regulatory role for the dioxin receptor (AhR) in cell adhesion and migration. Following our previous work, we report here that the C-terminal Src kinase-binding protein (Cbp) signaling pathway controls ß1 integrin activation and that this mechanism is AhR dependent. T-FGM AhR-/- fibroblasts displayed higher integrin ß1 activation, revealed by the increased binding of the activation reporter 9EG7 anti-ß1 mAb and of a soluble fibronectin fragment, as well as by enhanced talin-ß1 association. AhR-/- fibroblasts also showed increased fibronectin secretion and impaired directional migration. Notably, interfering Cbp expression in AhR-/- fibroblasts reduced ß1 integrin activation, improved cell migration and rescued wild-type cell morphology. Cbp over-expression in T-FGM AhR-/- cells enhanced the formation of inhibitory Csk-Cbp complexes which in turn reduced c-Src p-Tyr(416) activation and focal adhesion kinase (FAK) phosphorylation at the c-Src-responsive residues p-Tyr(576) and p-Tyr(577). The c-Src target and migration-related protein Cav1 was also hypophosphorylated at p-Tyr(14) in AhR-/- cells, and such effect was rescued by down-modulating Cbp levels. Thus, AhR regulates fibroblast migration by modulating ß1 integrin activation via Cbp-dependent, Src-mediated signaling.


Subject(s)
Integrin beta1/metabolism , Membrane Proteins/metabolism , Phosphoproteins/metabolism , Receptors, Aryl Hydrocarbon/metabolism , Sialoglycoproteins/metabolism , src-Family Kinases/metabolism , Actins/metabolism , Animals , CSK Tyrosine-Protein Kinase , Caveolin 1/metabolism , Cell Adhesion , Cell Movement , Fibroblasts/metabolism , Fibronectins/metabolism , Focal Adhesion Protein-Tyrosine Kinases/metabolism , Mice , Phosphorylation , RNA Interference , RNA, Small Interfering/metabolism , Receptors, Aryl Hydrocarbon/antagonists & inhibitors , Receptors, Aryl Hydrocarbon/genetics , Signal Transduction
8.
J Biol Chem ; 286(4): 2896-909, 2011 Jan 28.
Article in English | MEDLINE | ID: mdl-21115475

ABSTRACT

Aryl hydrocarbon receptor (Ahr) is a transcriptional factor involved in detoxification responses to pollutants and in intrinsic biological processes of multicellular organisms. We recently described that Vav3, an activator of Rho/Rac GTPases, is an Ahr transcriptional target in embryonic fibroblasts. These results prompted us to compare the Ahr(-/-) and Vav3(-/-) mouse phenotypes to investigate the implications of this functional interaction in vivo. Here, we show that Ahr is important for Vav3 expression in kidney, lung, heart, liver, and brainstem regions. This process is not affected by the administration of potent Ahr ligands such as benzo[a]pyrene. We also report that Ahr- and Vav3-deficient mice display hypertension, tachypnea, and sympathoexcitation. The Ahr gene deficiency also induces the GABAergic transmission defects present in the Vav3(-/-) ventrolateral medulla, a main cardiorespiratory brainstem center. However, Ahr(-/-) mice, unlike Vav3-deficient animals, display additional defects in fertility, perinatal growth, liver size and function, closure, spleen size, and peripheral lymphocytes. These results demonstrate that Vav3 is a bona fide Ahr target that is in charge of a limited subset of the developmental and physiological functions controlled by this transcriptional factor. Our data also reveal the presence of sympathoexcitation and new cardiorespiratory defects in Ahr(-/-) mice.


Subject(s)
Cardiovascular System/metabolism , Gene Expression Regulation/physiology , Proto-Oncogene Proteins c-vav/metabolism , Receptors, Aryl Hydrocarbon/metabolism , Respiratory System/metabolism , Animals , Benzo(a)pyrene/pharmacology , Brain Stem/metabolism , Cells, Cultured , Embryo, Mammalian/cytology , Embryo, Mammalian/metabolism , Fibroblasts/cytology , Fibroblasts/metabolism , Gene Expression Regulation/drug effects , Hypertension/genetics , Hypertension/metabolism , Mice , Mice, Knockout , Organ Specificity/drug effects , Organ Specificity/physiology , Proto-Oncogene Proteins c-vav/genetics , Receptors, Aryl Hydrocarbon/genetics , Sleep Wake Disorders/genetics , Sleep Wake Disorders/metabolism , rac GTP-Binding Proteins/genetics , rac GTP-Binding Proteins/metabolism
9.
Kidney Int ; 78(4): 382-8, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20531453

ABSTRACT

It is well established that many cognate basolateral plasma membrane proteins are expressed apically in proximal tubule cells thus optimizing the reabsorption capacity of the kidney. The protein clathrin and its adapter proteins normally regulate basolateral polarity. Here we tested whether the unique proximal tubule polarity is dependent on an epithelial-specific basolateral clathrin adapter, AP1B, present in most other epithelia. Quantitative PCR of isolated mouse renal tubules showed that AP1B was absent in proximal tubules but present in medullary and cortical thick ascending limbs of Henle, and cortical collecting ducts. Western blot confirmed the absence of AP1B in three established proximal tubule cell lines. Knockdown of AP1B by shRNA in prototypical distal tubule MDCK cells resulted in redistribution of the basolateral parathyroid hormone receptor, the insulin-like growth factor II receptor/calcium-independent mannose-6-phosphate receptor, and the junctional adhesion molecule, JAM-C, to a proximal tubule-like nonpolar localization. Yeast two-hybrid assays detected direct interactions between the cytoplasmic tails of these plasma membrane proteins and the cargo-binding region of the AP1B complex. Hence, our results show that differential expression of AP1B contributes to normal kidney function and illustrates possible roles of this adapter protein in kidney development, physiology, and pathology.


Subject(s)
Adaptor Protein Complex beta Subunits/analysis , Adaptor Proteins, Vesicular Transport/analysis , Cell Polarity/physiology , Kidney Tubules, Proximal/physiology , Absorption , Adaptor Protein Complex beta Subunits/genetics , Adaptor Protein Complex beta Subunits/metabolism , Adaptor Proteins, Vesicular Transport/genetics , Adaptor Proteins, Vesicular Transport/metabolism , Animals , Cell Line , Dogs , Membrane Proteins/metabolism , Protein Binding
10.
J Biol Chem ; 284(37): 25135-48, 2009 Sep 11.
Article in English | MEDLINE | ID: mdl-19617630

ABSTRACT

Angiogenesis has key roles in development and in the progression of human diseases such as cancer. Consequently, identifying the novel markers and regulators of angiogenesis is a critical task. The dioxin receptor (AhR) contributes to vascular homeostasis and to the endothelial response to toxins, although the mechanisms involved are largely uncharacterized. Here, we show that AhR-null mice (AhR(-/-)) have impaired angiogenesis in vivo that compromises tumor xenograft growth. Aortic rings emigration experiments and RNA interference indicated that AhR(-/-) endothelial cells failed to branch and to form tube-like structures. Such a phenotype was found to be vascular endothelial growth factor (VEGF)-dependent, as AhR(-/-) aortic endothelial cells (MAECs) secreted lower amounts of active VEGF-A and their treatment with VEGF-A rescued angiogenesis in culture and in vivo. Further, the addition of anti-VEGF antibody to AhR(+/+) MAECs reduced angiogenesis. Treatment under hypoxic conditions with 2-methoxyestradiol suggested that HIF-1alpha modulates endothelial VEGF expression in an AhR-dependent manner. Importantly, AhR-null stromal myofibroblasts produced increased transforming growth factor-beta (TGFbeta) activity, which inhibited angiogenesis in human endothelial cells (HMECs) and AhR(-/-) mice, whereas the co-culture of HMECs with AhR(-/-) myofibroblasts or with their conditioned medium inhibited branching, which was restored by an anti-TGFbeta antibody. Moreover, VEGF and TGFbeta activities cooperated in modulating angiogenesis, as the addition of TGFbeta to AhR(-/-) MAECs further reduced their low basal VEGF-A activity. Thus, AhR modulates angiogenesis through a mechanism requiring VEGF activation in the endothelium and TGFbeta inactivation in the stroma. These data highlight the role of AhR in cardiovascular homeostasis and suggest that this receptor can be a novel regulator of angiogenesis during tumor development.


Subject(s)
Endothelium/metabolism , Neovascularization, Pathologic , Receptors, Aryl Hydrocarbon/deficiency , Receptors, Aryl Hydrocarbon/physiology , Transforming Growth Factor beta/metabolism , Vascular Endothelial Growth Factor A/metabolism , Animals , Aorta/pathology , Cobalt/pharmacology , Embryonic Stem Cells/cytology , Fibroblasts/metabolism , Hypoxia , Melanoma, Experimental , Mice , Neoplasm Transplantation , Recombination, Genetic
11.
J Cell Sci ; 122(Pt 11): 1823-33, 2009 Jun 01.
Article in English | MEDLINE | ID: mdl-19435800

ABSTRACT

Delayed wound healing caused by inefficient re-epithelialization underlines chronic skin lesions such as those found in diabetes. The dioxin receptor (AhR) modulates cell plasticity and migration and its activation by occupational polycyclic aromatic hydrocarbons (PAHs) results in severe skin lesions such as contact hypersensitivity, dermatitis and chloracne. Using wild-type (Ahr+/+) and AhR-null (Ahr-/-) mouse primary keratinocyte cultures and tissue explants, we show that lack of AhR increases keratinocyte migration and accelerates skin re-epithelialization without affecting cell proliferation or recruitment of inflammatory cells. Wounds in Ahr-/- animals had elevated numbers of fibroblasts and increased collagen content in their granulation tissue. Importantly, Ahr-/- dermal fibroblasts secreted higher levels of active TGFbeta that increased keratinocyte migration in culture and that could account for over-activation of the TGFbeta pathway and for faster wound healing in the AhR-null neo-epithelium. Consistently, a TGFbeta neutralizing antibody decreased keratinocyte migration in culture and halted re-epithelialization in Ahr-/- mice. Moreover, in vivo treatment with an antisense oligonucleotide for AhR increased TGFbeta signaling and improved re-epithelialization in wounds of wild-type mice. These data indicate that AhR is relevant for wound repair and suggest that AhR downmodulation might be a potential new tool for the treatment of chronic, surgical or accidental wounds.


Subject(s)
Receptors, Aryl Hydrocarbon/metabolism , Transforming Growth Factor beta/metabolism , Wound Healing/physiology , Animals , Cells, Cultured , Collagen/metabolism , Fibroblasts/cytology , Fibroblasts/metabolism , Humans , Keratinocytes/cytology , Keratinocytes/metabolism , Male , Mice , Mice, Knockout , Oligonucleotides, Antisense/genetics , Oligonucleotides, Antisense/metabolism , Receptors, Aryl Hydrocarbon/genetics , Skin/cytology , Skin/metabolism , Skin/pathology , Transforming Growth Factor beta/genetics
12.
Mol Biol Cell ; 20(6): 1715-27, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19158396

ABSTRACT

The dioxin receptor (AhR) modulates cell plasticity and migration, although the signaling involved remains unknown. Here, we report a mechanism that integrates AhR into these cytoskeleton-related functions. Immortalized and mouse embryonic fibroblasts lacking AhR (AhR-/-) had increased cell area due to spread cytoplasms that reverted to wild-type morphology upon AhR re-expression. The AhR-null phenotype included increased F-actin stress fibers, depolarized focal adhesions, and enhanced spreading and adhesion. The cytoskeleton alterations of AhR-/- cells were due to down-regulation of constitutive Vav3 expression, a guanosine diphosphate/guanosine triphosphate exchange factor for Rho/Rac GTPases and a novel transcriptional target of AhR. AhR was recruited to the vav3 promoter and maintained constitutive mRNA expression in a ligand-independent manner. Consistently, AhR-/- fibroblasts had reduced Rac1 activity and increased activation of the RhoA/Rho kinase (Rock) pathway. Pharmacological inhibition of Rac1 shifted AhR+/+ fibroblasts to the null phenotype, whereas Rock inhibition changed AhR-null cells to the AhR+/+ morphology. Knockdown of vav3 transcripts by small interfering RNA induced cytoskeleton defects and changes in adhesion and spreading mimicking those of AhR-null cells. Moreover, vav3-/- MEFs, as AhR-/- mouse embryonic fibroblasts, had increased cell area and enhanced stress fibers. By modulating Vav3-dependent signaling, AhR could regulate cell shape, adhesion, and migration under physiological conditions and, perhaps, in certain pathological states.


Subject(s)
Cell Shape , Proto-Oncogene Proteins c-vav/metabolism , Receptors, Aryl Hydrocarbon/metabolism , Actins/metabolism , Animals , Cell Adhesion , Cells, Cultured , Cytoskeleton/metabolism , Mice , Mice, Knockout , Phenotype , Proto-Oncogene Proteins c-vav/deficiency , Proto-Oncogene Proteins c-vav/genetics , RNA, Messenger/genetics , Receptors, Aryl Hydrocarbon/deficiency , Receptors, Aryl Hydrocarbon/genetics , Transcription, Genetic/genetics , rac1 GTP-Binding Protein/metabolism , rhoA GTP-Binding Protein/metabolism
13.
Biochem Pharmacol ; 77(4): 700-12, 2009 Feb 15.
Article in English | MEDLINE | ID: mdl-18812170

ABSTRACT

As our knowledge on the mechanisms that control cell function increases, more complex signaling pathways and quite intricate cross-talks among regulatory proteins are discovered. Establishing accurate interactions between cellular networks is essential for a healthy cell and different alterations in signaling are known to underline human disease. Transforming growth factor beta (TGFbeta) is an extracellular cytokine that regulates such critical cellular responses as proliferation, apoptosis, differentiation, angiogenesis and migration, and it is assumed that the latency-associated protein LTBP-1 plays a relevant role in TGFbeta targeting and activation in the extracellular matrix (ECM). The dioxin receptor (AhR) is a unique intracellular protein long studied because of its critical role in xenobiotic-induced toxicity and carcinogenesis. Yet, a large set of studies performed in cellular systems and in vivo animal models have suggested important xenobiotic-independent functions for AhR in cell proliferation, differentiation and migration and in tissue homeostasis. Remarkably, AhR activity converges with TGFbeta-dependent signaling through LTBP-1 since cells lacking AhR expression have phenotypic alterations that can be explained, at least in part, by the coordinated regulation of both proteins. Here, we will discuss the existence of functional interactions between AhR and TGFbeta signaling. We will focus on regulatory and functional aspects by analyzing how AhR status determines TGFbeta activity and by proposing a mechanism through which LTBP-1, a novel AhR target gene, mediates such effects. We will integrate ECM proteases in the AhR-LTBP-1-TGFbeta axis and suggest a model that could help explain some in vivo phenotypes associated to AhR deficiency.


Subject(s)
Cell Proliferation , Homeostasis , Receptor Cross-Talk/physiology , Receptors, Aryl Hydrocarbon/physiology , Signal Transduction/physiology , Transforming Growth Factor beta/physiology , Animals , Cell Proliferation/drug effects , Homeostasis/drug effects , Humans , Receptor Cross-Talk/drug effects , Receptors, Aryl Hydrocarbon/genetics , Receptors, Aryl Hydrocarbon/metabolism , Signal Transduction/drug effects , Transforming Growth Factor beta/genetics , Transforming Growth Factor beta/metabolism , Xenobiotics/toxicity
14.
J Mol Biol ; 380(1): 1-16, 2008 Jun 27.
Article in English | MEDLINE | ID: mdl-18508077

ABSTRACT

Latent TGFbeta-binding protein 1 (LTBP-1) is a key regulator of TGFbeta targeting and activation in the extracellular matrix. LTBP-1 is recognized as a major docking molecule to localize, and possibly to activate, TGFbeta in the extracellular matrix. Despite this relevant function, the molecular mechanisms regulating Ltbp-1 transcription remain largely unknown. Previous results from our laboratory revealed that mouse embryonic fibroblasts (MEF) lacking dioxin receptor (AhR) had increased Ltbp-1 mRNA expression and elevated TGFbeta activity, suggesting that AhR repressed Ltbp-1 transcription. Here, we have cloned the mouse Ltbp-1 gene promoter and analysed its mechanism of transcriptional repression by AhR. Reporter gene assays, AhR over-expression and site-directed mutagenesis showed that basal Ltbp-1 transcription is AhR-dependent. Chromatin immunoprecipitation (ChIP) and RNA interference (RNAi) revealed that AhR regulates Ltbp-1 transcription by a mechanism involving recruitment of co-activators such as CREB1 and co-repressors such as HDAC2 to the Ltbp-1 promoter. In AhR-expressing (AhR+/+) MEF cells, the recruitment of HDAC1, 2 and 4 correlated with decreased K8H4 acetylation and impaired binding of pCREB(Ser133) to the Ltbp-1 promoter, likely maintaining a constitutive repressed state. AhR-/- MEF cells had the opposite pattern of HDACs and pCREB1(Ser133) binding to Ltbp-1 promoter, and therefore, over-expressed Ltbp-1 mRNA. In agreement, siRNA for HDAC2 increased Ltbp-1 expression and K8H4 acetylation in AhR+/+ but not in AhR-/- MEF cells. We suggest that HDAC2 binding keeps Ltbp-1 promoter repressed in AhR+/+ MEF cells, whereas in AhR-null MEF cells the absence of HDAC2 and the binding of pCREB(Ser133) allow Ltbp-1 transcription. Thus, epigenetics can contribute to constitutive Ltbp-1 repression by a mechanism requiring AhR activity.


Subject(s)
Cyclic AMP Response Element-Binding Protein/metabolism , Gene Expression Regulation , Histone Deacetylases/metabolism , Latent TGF-beta Binding Proteins/genetics , Promoter Regions, Genetic/genetics , Receptors, Aryl Hydrocarbon/metabolism , Repressor Proteins/metabolism , Acetylation , Animals , Base Sequence , Cloning, Molecular , DNA Methylation , Genotype , Histone Deacetylase 2 , Histone Deacetylases/genetics , Histones/metabolism , Latent TGF-beta Binding Proteins/metabolism , Mice , Molecular Sequence Data , Mutagenesis, Site-Directed , Protein Binding , RNA Interference , Repressor Proteins/genetics , Response Elements/genetics
15.
Proc Natl Acad Sci U S A ; 105(5): 1632-7, 2008 Feb 05.
Article in English | MEDLINE | ID: mdl-18223155

ABSTRACT

Alterations in tissue-specific gene expression greatly affect cell function. Transcription factors (TFs) interact with cis-acting binding sites in noncoding enhancer promoter regions. Transposable elements (TEs) are abundant and similarly represented among mammalian genomes. TEs are important in gene regulation, but their function is not well understood. We have characterized a TE containing functional TF-binding sites for the carcinogen-activated dioxin receptor xenobiotic responsive element (XRE) and the epithelial-mesenchymal transition regulator Slug (Slug site). A Mus promoter database was scanned for XREs to predict coregulation with other TFs. We identified an overrepresented (1,398 genes) B1 retrotransposon containing XRE and Slug sites within 35 bp of each other (designated as B1-X35S). This B1-X35S retrotransposon differed from classic B1s by the presence of the Slug site and by its differential nucleotide conservation outside the X35S region. Phylogenetically, B1-X35S appeared recently in evolution, close to the B1-B subfamily. Comparative gene expression in 61 mouse tissues revealed that B1-X35S-containing genes had lower median expression levels than those with canonical B1 TEs, suggesting a repressive role for X35S. Indeed, X35S was functional and able to bind aryl hydrocarbon (dioxin) receptor (AhR) and Slug and, importantly, to repress cis-reporter genes. Moreover, AhR and Slug were recruited to X35S in vivo and repressed the endogenous expression of X35S-containing genes. Our results demonstrate the existence of a widely present B1 subfamily in the mouse. Because AhR and Slug are relevant in tumor development and differentiation, X35S may represent a genome-wide regulatory mechanism and a tool to modulate gene expression.


Subject(s)
Gene Expression Regulation , Receptors, Aryl Hydrocarbon/metabolism , Retroelements , Transcription Factors/metabolism , Animals , Base Sequence , Cell Line, Tumor , Gene Expression , Genome/genetics , Mice , Molecular Sequence Data , Promoter Regions, Genetic , Snail Family Transcription Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...