Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
1.
Oncogene ; 40(19): 3460-3469, 2021 05.
Article in English | MEDLINE | ID: mdl-33767435

ABSTRACT

In solid cancers, high expression of the cellular prion protein (PrPC) is associated with stemness, invasiveness, and resistance to chemotherapy, but the role of PrPC in tumor response to radiotherapy is unknown. Here, we show that, in neuroblastoma, breast, and colorectal cancer cell lines, PrPC expression is increased after ionizing radiation (IR) and that PrPC deficiency increases radiation sensitivity and decreases radiation-induced radioresistance in tumor cells. In neuroblastoma cells, IR activates ATM that triggers TAK1-dependent phosphorylation of JNK and subsequent activation of the AP-1 transcription factor that ultimately increases PRNP promoter transcriptional activity through an AP-1 binding site in the PRNP promoter. Importantly, we show that this ATM-TAK1-PrPC pathway mediated radioresistance is activated in all tumor cell lines studied and that pharmacological inhibition of TAK1 activity recapitulates the effects of PrPC deficiency. Altogether, these results unveil how tumor cells activate PRNP to acquire resistance to radiotherapy and might have implications for therapeutic targeting of solid tumors radioresistance.


Subject(s)
Ataxia Telangiectasia Mutated Proteins/metabolism , MAP Kinase Kinase Kinases/metabolism , Neoplasms/metabolism , Neoplasms/radiotherapy , PrPC Proteins/biosynthesis , Cell Line, Tumor , Humans , Neoplasms/genetics , PrPC Proteins/metabolism , Radiation Tolerance
2.
Sci Adv ; 6(12): eaay3704, 2020 03.
Article in English | MEDLINE | ID: mdl-32219160

ABSTRACT

Human and murine skin wounding commonly results in fibrotic scarring, but the murine wounding model wound-induced hair neogenesis (WIHN) can frequently result in a regenerative repair response. Here, we show in single-cell RNA sequencing comparisons of semi-regenerative and fibrotic WIHN wounds, increased expression of phagocytic/lysosomal genes in macrophages associated with predominance of fibrotic myofibroblasts in fibrotic wounds. Investigation revealed that macrophages in the late wound drive fibrosis by phagocytizing dermal Wnt inhibitor SFRP4 to establish persistent Wnt activity. In accordance, phagocytosis abrogation resulted in transient Wnt activity and a more regenerative healing. Phagocytosis of SFRP4 was integrin-mediated and dependent on the interaction of SFRP4 with the EDA splice variant of fibronectin. In the human skin condition hidradenitis suppurativa, phagocytosis of SFRP4 by macrophages correlated with fibrotic wound repair. These results reveal that macrophages can modulate a key signaling pathway via phagocytosis to alter the skin wound healing fate.


Subject(s)
Macrophages/immunology , Macrophages/metabolism , Phagocytosis/immunology , Proto-Oncogene Proteins/metabolism , Wnt Proteins/metabolism , Wnt Signaling Pathway , Wound Healing , Fibroblasts/metabolism , Fibrosis , Humans , Proteolysis , Skin/immunology , Skin/injuries , Skin/metabolism , Wound Healing/immunology
3.
Epigenetics Chromatin ; 12(1): 46, 2019 07 22.
Article in English | MEDLINE | ID: mdl-31331374

ABSTRACT

BACKGROUND: Cell type-specific use of cis-acting regulatory elements is mediated by the combinatorial activity of transcription factors involved in lineage determination and maintenance of cell identity. In macrophages, specific transcriptional programs are dictated by the transcription factor PU.1 that primes distal regulatory elements for macrophage identities and makes chromatin competent for activity of stimuli-dependent transcription factors. Although the advances in genome-wide approaches have elucidated the functions of these macrophage-specific distal regulatory elements in transcriptional responses, chromatin structures associated with PU.1 priming and the underlying mechanisms of action of these cis-acting sequences are not characterized. RESULTS: Here, we show that, in macrophages, FACT subunit SPT16 can bind to positioned nucleosomes directly flanking PU.1-bound sites at previously uncharacterized distal regulatory elements located near genes essential for macrophage development and functions. SPT16 can interact with the transcriptional co-regulator TRIM33 and binds to half of these sites in a TRIM33-dependent manner. Using the Atp1b3 locus as a model, we show that FACT binds to two positioned nucleosomes surrounding a TRIM33/PU.1-bound site in a region, located 35 kb upstream the Atp1b3 TSS, that interact with the Atp1b3 promoter. At this - 35 kb region, TRIM33 deficiency leads to FACT release, loss of the two positioned nucleosomes, RNA Pol II recruitment and bidirectional transcription. These modifications are associated with higher levels of FACT binding at the Atp1b3 promoter, an increase of RNA Pol II recruitment and an increased expression of Atp1b3 in Trim33-/- macrophages. CONCLUSIONS: Thus, sequestering of SPT16/FACT by TRIM33 at PU.1-bound distal regions might represent a new regulatory mechanism for RNA Pol II recruitment and transcription output in macrophages.


Subject(s)
Chromatin Assembly and Disassembly , Transcription Factors/metabolism , Transcriptional Elongation Factors/metabolism , Animals , Binding Sites , Histones/metabolism , Macrophages/cytology , Macrophages/metabolism , Mice , Mice, Inbred C57BL , Nucleosomes/metabolism , Promoter Regions, Genetic , Protein Binding , RNA Polymerase II/metabolism , Regulatory Sequences, Nucleic Acid , Sodium-Potassium-Exchanging ATPase/genetics , Transcription Factors/deficiency , Transcription Factors/genetics , Transcription, Genetic
4.
EBioMedicine ; 44: 60-70, 2019 Jun.
Article in English | MEDLINE | ID: mdl-31130476

ABSTRACT

BACKGROUND: Mature myeloid cells play a crucial role in Crohn's disease (CD) but the molecular players that regulate their functions in CD are not fully characterized. We and others have shown that TRIM33 is involved in the innate immune response and in the inflammatory response but TRIM33 role in intestinal inflammation is not known. In this study, we investigated the role of TRIM33 in myeloid cells during dextran sulfate sodium (DSS)-induced colitis. METHODS: We study the role of TRIM33 during DSS-induced colitis which mimics intestinal inflammation using mice deleted for Trim33 only in mature myeloid cells (Trim33-/- mice) FINDINGS: We first show that Trim33 mRNA level is decreased in CD patient's blood monocytes suggesting a role of TRIM33 in CD. Using Trim33-/- mice, we show that these mice display an impaired resolution of colonic inflammation with an increased number of blood and colon monocytes and a decreased number of colonic macrophages. Trim33-/- monocytes are less competent for recruitment and macrophage differentiation. Finally, during resolution of inflammation, Trim33-/- colonic macrophages display an impaired M1/M2 switch and express a low level of membrane-bound TNF that is associated with an increased number of colonic neutrophils. INTERPRETATION: Our study shows an important role of TRIM33 in monocytes/macrophages during DSS-induced colitis and suggests that the decreased expression of TRIM33 in CD patient's blood monocytes might not be a consequence but might be involved in CD progression. FUND: La Ligue contre le Cancer (équipe labelisée), INSERM, CEA, Université Paris-Diderot, Université Paris-Sud.


Subject(s)
Colitis/etiology , Macrophages/metabolism , Monocytes/metabolism , Transcription Factors/deficiency , Animals , Biomarkers , Colitis/metabolism , Colitis/pathology , Crohn Disease/etiology , Crohn Disease/metabolism , Crohn Disease/pathology , Cytokines/genetics , Cytokines/metabolism , Disease Models, Animal , Gene Expression , Humans , Intestinal Mucosa/immunology , Intestinal Mucosa/metabolism , Intestinal Mucosa/pathology , Macrophage Activation/genetics , Macrophage Activation/immunology , Macrophages/immunology , Mice , Mice, Knockout , Monocytes/immunology , Myeloid Cells/immunology , Myeloid Cells/metabolism , RNA, Messenger
5.
Cell Rep ; 20(13): 3199-3211, 2017 Sep 26.
Article in English | MEDLINE | ID: mdl-28954235

ABSTRACT

Despite numerous observations linking protracted exposure to low-dose (LD) radiation and leukemia occurrence, the effects of LD irradiation on hematopoietic stem cells (HSCs) remain poorly documented. Here, we show that adult HSCs are hypersensitive to LD irradiation. This hyper-radiosensitivity is dependent on an immediate increase in the levels of reactive oxygen species (ROS) that also promotes autophagy and activation of the Keap1/Nrf2 antioxidant pathway. Nrf2 activation initially protects HSCs from the detrimental effects of ROS, but protection is transient, and increased ROS levels return, promoting a long-term decrease in HSC self-renewal. In vivo, LD total body irradiation (TBI) does not decrease HSC numbers unless the HSC microenvironment is altered by an inflammatory insult. Paradoxically, such an insult, in the form of granulocyte colony-stimulating factor (G-CSF) preconditioning, followed by LD-TBI facilitates efficient bone marrow transplantation without myeloablation. Thus, LD irradiation has long-term detrimental effects on HSCs that may result in hematological malignancies, but LD-TBI may open avenues to facilitate autologous bone marrow transplantation.


Subject(s)
Hematopoietic Stem Cells/metabolism , Oxidative Stress/genetics , Whole-Body Irradiation/methods , Animals , Humans , Mice
6.
Oncotarget ; 8(3): 5111-5122, 2017 Jan 17.
Article in English | MEDLINE | ID: mdl-27974684

ABSTRACT

The tripartite motif (TRIM) family of proteins plays important roles in innate immunity and antimicrobial infection. None of these proteins has been shown to directly regulate transcription of genes in monocyte/macrophage except TRIM33 that we have recently shown to be a macrophage specific transcriptional inhibitor of Ifnb1. Using ChIP-seq analyses, we now report that TRIM33 is bound to two fold more genes in immature than in mature myeloid cell lines. When located near the same genes, TRIM33 is bound to different sequences in the two cell lines suggesting a role of TRIM33 in both immature and mature myeloid cells. Accordingly, expression of TRIM33 in immature myeloid cells is necessary for efficient production of small peritoneal macrophages, monocytes and bone marrow derived macrophage (BMDM) and TRIM33 targets a subset of genes involved in the inflammatory response only in mature myeloid cells. Functionally, this targeting is associated with impaired repression of pathways regulating the late phases of lipopolysaccharide (LPS) activation of BMDM and a high sensitivity to LPS in vivo when the trim33 gene is inactivated in mature myeloid cells. These findings pinpoint TRIM33 as an important transcriptional actor of monocyte/macrophage mediated inflammation.


Subject(s)
Chromatin/metabolism , Macrophage Activation , Macrophages/cytology , Transcription Factors/metabolism , Animals , Cells, Cultured , Chromatin/genetics , Chromatin Immunoprecipitation , DNA/metabolism , Gene Regulatory Networks , Lipopolysaccharides/pharmacology , Macrophages/drug effects , Macrophages/immunology , Mice , Myeloid Cells/cytology , Myeloid Cells/immunology , RAW 264.7 Cells , Sequence Analysis, DNA , Transcription Factors/genetics
7.
Nat Commun ; 6: 8900, 2015 Nov 23.
Article in English | MEDLINE | ID: mdl-26592194

ABSTRACT

Despite its importance during viral or bacterial infections, transcriptional regulation of the interferon-ß gene (Ifnb1) in activated macrophages is only partially understood. Here we report that TRIM33 deficiency results in high, sustained expression of Ifnb1 at late stages of toll-like receptor-mediated activation in macrophages but not in fibroblasts. In macrophages, TRIM33 is recruited by PU.1 to a conserved region, the Ifnb1 Control Element (ICE), located 15 kb upstream of the Ifnb1 transcription start site. ICE constitutively interacts with Ifnb1 through a TRIM33-independent chromatin loop. At late phases of lipopolysaccharide activation of macrophages, TRIM33 is bound to ICE, regulates Ifnb1 enhanceosome loading, controls Ifnb1 chromatin structure and represses Ifnb1 gene transcription by preventing recruitment of CBP/p300. These results characterize a previously unknown mechanism of macrophage-specific regulation of Ifnb1 transcription whereby TRIM33 is critical for Ifnb1 gene transcription shutdown.


Subject(s)
Gene Expression Regulation , Interferon-beta/genetics , Macrophages/cytology , Macrophages/metabolism , Transcription Factors/metabolism , Animals , Female , Interferon-beta/metabolism , Macrophage Activation , Male , Mice , Mice, Inbred C57BL , Transcription Factors/genetics , Transcription, Genetic
8.
Cell Stem Cell ; 8(4): 412-25, 2011 Apr 08.
Article in English | MEDLINE | ID: mdl-21474105

ABSTRACT

Crosstalk between transcription factors and cytokines precisely regulates tissue homeostasis. Transcriptional intermediary factor 1γ (TIF1γ) regulates vertebrate hematopoietic development, can control transcription elongation, and is a component of the TGF-ß signaling pathway. Here we show that deletion of TIF1γ in adult hematopoiesis is compatible with life and long-term maintenance of essential blood cell lineages. However, loss of TIF1γ results in deficient long-term hematopoietic stem cell (LT-HSC) transplantation activity, deficient short-term HSC (ST-HSC) bone marrow retention, and priming ST-HSCs to myelomonocytic lineage. These defects are hematopoietic cell-autonomous, and priming of TIF1γ-deficient ST-HSCs can be partially rescued by wild-type hematopoietic cells. TIF1γ can form complexes with TAL1 or PU.1-two essential DNA-binding proteins in hematopoiesis-occupy specific subsets of their DNA binding sites in vivo, and repress their transcriptional activity. These results suggest a regulation of adult hematopoiesis through TIF1γ-mediated transcriptional repression of TAL1 and PU.1 target genes.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/genetics , Hematopoiesis/genetics , Proto-Oncogene Proteins/genetics , Trans-Activators/genetics , Transcription Factors/physiology , Down-Regulation/genetics , Repressor Proteins/genetics , T-Cell Acute Lymphocytic Leukemia Protein 1 , Transcription, Genetic
9.
J Exp Med ; 207(10): 2141-56, 2010 Sep 27.
Article in English | MEDLINE | ID: mdl-20855495

ABSTRACT

TAL1 (also known as SCL) is expressed in >40% of human T cell acute lymphoblastic leukemias (T-ALLs). TAL1 encodes a basic helix-loop-helix transcription factor that can interfere with the transcriptional activity of E2A and HEB during T cell leukemogenesis; however, the oncogenic pathways directly activated by TAL1 are not characterized. In this study, we show that, in human TAL1-expressing T-ALL cell lines, TAL1 directly activates NKX3.1, a tumor suppressor gene required for prostate stem cell maintenance. In human T-ALL cell lines, NKX3.1 gene activation is mediated by a TAL1-LMO-Ldb1 complex that is recruited by GATA-3 bound to an NKX3.1 gene promoter regulatory sequence. TAL1-induced NKX3.1 activation is associated with suppression of HP1-α (heterochromatin protein 1 α) binding and opening of chromatin on the NKX3.1 gene promoter. NKX3.1 is necessary for T-ALL proliferation, can partially restore proliferation in TAL1 knockdown cells, and directly regulates miR-17-92. In primary human TAL1-expressing leukemic cells, the NKX3.1 gene is expressed independently of the Notch pathway, and its inactivation impairs proliferation. Finally, TAL1 or NKX3.1 knockdown abrogates the ability of human T-ALL cells to efficiently induce leukemia development in mice. These results suggest that tumor suppressor or oncogenic activity of NKX3.1 depends on tissue expression.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/genetics , Homeodomain Proteins/genetics , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/genetics , Proto-Oncogene Proteins/genetics , Transcription Factors/genetics , Adaptor Proteins, Signal Transducing , Animals , Basic Helix-Loop-Helix Transcription Factors/biosynthesis , Cell Line, Tumor , Cell Proliferation , DNA-Binding Proteins/metabolism , GATA3 Transcription Factor/metabolism , Gene Knockdown Techniques , Genes, Tumor Suppressor , Homeodomain Proteins/metabolism , Humans , LIM Domain Proteins , Male , Metalloproteins/metabolism , Mice , Neoplasm Transplantation , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/metabolism , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/pathology , Prostate/metabolism , Prostate/pathology , Protein Binding , Proto-Oncogene Proteins/biosynthesis , Stem Cells/physiology , T-Cell Acute Lymphocytic Leukemia Protein 1 , Transcription Factors/metabolism
10.
Nucleic Acids Res ; 37(15): 5071-80, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19542185

ABSTRACT

Non-coding RNAs are emerging as key players in many fundamental biological processes, including specification of higher-order chromatin structure. We examined the implication of RNA transcribed from mouse centromeric minor satellite repeats in the formation and function of centromere-associated complexes. Here we show that the levels of minor satellite RNA vary during cell-cycle progression, peaking in G2/M phase, concomitant with accumulation of proteins of the chromosomal passenger complex near the centromere. Consistent with this, we describe that murine minor satellite RNA are components of CENP-A-associated centromeric fractions and associate with proteins of the chromosomal passenger complex Aurora B and Survivin at the onset of mitosis. Interactions of endogenous Aurora B with CENP-A and Survivin are sensitive to RNaseA. Likewise, the kinase activity of Aurora B requires an RNA component. More importantly, Aurora B kinase activity can be potentiated by minor satellite RNA. In addition, decreased Aurora B activity after RNA depletion can be specifically rescued by restitution of these transcripts. Together, our data provide new functional evidence for minor satellite transcripts as key partners and regulators of the mitotic kinase Aurora B.


Subject(s)
Centromere/chemistry , Chromatin/chemistry , Protein Serine-Threonine Kinases/metabolism , RNA, Untranslated/metabolism , Animals , Aurora Kinase B , Aurora Kinases , Autoantigens/analysis , Cell Cycle/genetics , Cell Line , Centromere Protein A , Chromosomal Proteins, Non-Histone/analysis , DNA, Satellite , Inhibitor of Apoptosis Proteins , Mice , Microtubule-Associated Proteins/metabolism , Mitosis/genetics , RNA, Untranslated/analysis , Repressor Proteins , Survivin
11.
J Neurosci Res ; 86(4): 776-84, 2008 Mar.
Article in English | MEDLINE | ID: mdl-17941053

ABSTRACT

We have previously demonstrated that, in C6 glioma cells, eicosapentaenoic acid (EPA) stimulates the expression of proteolipid protein (PLP) via cAMP-mediated pathways. In this study, we investigated whether n-3 polyunsaturated fatty acids can affect myelinogenesis in vivo. A single dose of either EPA or docosahexaenoic acid (DHA) was injected intracerebroventricularly into 2-day-old rats, which were then killed after 3 days post-injection (p.i.). Total RNA was isolated from the medulla, cerebellum, and cortex, and the expression of myelin-specific mRNAs was analyzed by real-time PCR. The levels of PLP, myelin basic protein, and myelin oligodendrocyte protein mRNAs increased in nearly all brain regions of DHA- and EPA-treated animals, but the effect was more pronounced in EPA-treated rats. The enhancement in PLP transcript levels was followed by an increase in PLP translation in EPA-treated rats. A further indicator of accelerated myelination was the increase in 2'-3'-cyclic nucleotide 3'-phosphodiesterase (CNPase) protein levels. In EPA-treated rats, the increased expression of myelin genes coincided with a decrease of cAMP-response element-binding protein (CREB)-DNA binding in the cerebellum and cortex (1 hr p.i.). After 16 hr, this effect was still present in the same cerebral regions even though the decrease in EPA-treated rats was less pronounced than in controls. The down-regulation of CREB activity was due to a decrease in the levels of CREB phosphorylation. In conclusion, our data suggest that EPA stimulates the expression of specific myelin proteins through decreased CREB phosphorylation. These results corroborate the clinical studies of the n-3 PUFA beneficial effects on several demyelinating diseases.


Subject(s)
Brain/drug effects , Eicosapentaenoic Acid/administration & dosage , Gene Expression/drug effects , Myelin Proteins/drug effects , Animals , Blotting, Northern , Blotting, Western , Brain/metabolism , Cyclic AMP Response Element-Binding Protein/drug effects , Docosahexaenoic Acids/administration & dosage , Electrophoretic Mobility Shift Assay , Injections, Intraventricular , Myelin Proteins/metabolism , Polymerase Chain Reaction , RNA, Messenger/analysis , Rats , Rats, Sprague-Dawley , Reverse Transcriptase Polymerase Chain Reaction
SELECTION OF CITATIONS
SEARCH DETAIL
...