Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 26
Filter
1.
Nucleic Acids Res ; 48(10): e55, 2020 06 04.
Article in English | MEDLINE | ID: mdl-32196115

ABSTRACT

Single-cell transcriptomics offers unprecedented opportunities to infer the ligand-receptor (LR) interactions underlying cellular networks. We introduce a new, curated LR database and a novel regularized score to perform such inferences. For the first time, we try to assess the confidence in predicted LR interactions and show that our regularized score outperforms other scoring schemes while controlling false positives. SingleCellSignalR is implemented as an open-access R package accessible to entry-level users and available from https://github.com/SCA-IRCM. Analysis results come in a variety of tabular and graphical formats. For instance, we provide a unique network view integrating all the intercellular interactions, and a function relating receptors to expressed intracellular pathways. A detailed comparison of related tools is conducted. Among various examples, we demonstrate SingleCellSignalR on mouse epidermis data and discover an oriented communication structure from external to basal layers.


Subject(s)
Gene Expression Profiling/methods , Signal Transduction , Single-Cell Analysis/methods , Software , Animals , Epidermis/metabolism , Ligands , Mice , Workflow
2.
Int J Mol Sci ; 23(16)2022 Aug 16.
Article in English | MEDLINE | ID: mdl-36012478

ABSTRACT

E4F1 is essential for early embryonic mouse development and for controlling the balance between proliferation and survival of actively dividing cells. We previously reported that E4F1 is essential for the survival of murine p53-deficient cancer cells by controlling the expression of genes involved in mitochondria functions and metabolism, and in cell-cycle checkpoints, including CHEK1, a major component of the DNA damage and replication stress responses. Here, combining ChIP-Seq and RNA-Seq approaches, we identified the transcriptional program directly controlled by E4F1 in Human Triple-Negative Breast Cancer cells (TNBC). E4F1 binds and regulates a limited list of direct target genes (57 genes) in these cells, including the human CHEK1 gene and, surprisingly, also two other genes encoding post-transcriptional regulators of the ATM/ATR-CHK1 axis, namely, the TTT complex component TTI2 and the phosphatase PPP5C, that are essential for the folding and stability, and the signaling of ATM/ATR kinases, respectively. Importantly, E4F1 also binds the promoter of these genes in vivo in Primary Derived Xenograft (PDX) of human TNBC. Consequently, the protein levels and signaling of CHK1 but also of ATM/ATR kinases are strongly downregulated in E4F1-depleted TNBC cells resulting in a deficiency of the DNA damage and replicative stress response in these cells. The E4F1-depleted cells fail to arrest into S-phase upon treatment with the replication-stalling agent Gemcitabine, and are highly sensitized to this drug, as well as to other DNA-damaging agents, such as Cisplatin. Altogether, our data indicate that in breast cancer cells the ATM/ATR-CHK1 signaling pathway and DNA damage-stress response are tightly controlled at the transcriptional and post-transcriptional level by E4F1.


Subject(s)
Repressor Proteins , Transcription Factors , Triple Negative Breast Neoplasms , Ubiquitin-Protein Ligases , Animals , Ataxia Telangiectasia Mutated Proteins/metabolism , Cell Cycle Proteins/metabolism , Checkpoint Kinase 1/metabolism , DNA Damage , DNA-Binding Proteins/metabolism , Humans , Mice , Phosphorylation , Protein Kinases/metabolism , Repressor Proteins/metabolism , Transcription Factors/metabolism , Triple Negative Breast Neoplasms/metabolism , Ubiquitin-Protein Ligases/metabolism
3.
Proc Natl Acad Sci U S A ; 113(39): 11004-9, 2016 09 27.
Article in English | MEDLINE | ID: mdl-27621431

ABSTRACT

The multifunctional protein E4 transcription factor 1 (E4F1) is an essential regulator of epidermal stem cell (ESC) maintenance. Here, we found that E4F1 transcriptionally regulates a metabolic program involved in pyruvate metabolism that is required to maintain skin homeostasis. E4F1 deficiency in basal keratinocytes resulted in deregulated expression of dihydrolipoamide acetyltransferase (Dlat), a gene encoding the E2 subunit of the mitochondrial pyruvate dehydrogenase (PDH) complex. Accordingly, E4f1 knock-out (KO) keratinocytes exhibited impaired PDH activity and a redirection of the glycolytic flux toward lactate production. The metabolic reprogramming of E4f1 KO keratinocytes associated with remodeling of their microenvironment and alterations of the basement membrane, led to ESC mislocalization and exhaustion of the ESC pool. ShRNA-mediated depletion of Dlat in primary keratinocytes recapitulated defects observed upon E4f1 inactivation, including increased lactate secretion, enhanced activity of extracellular matrix remodeling enzymes, and impaired clonogenic potential. Altogether, our data reveal a central role for Dlat in the metabolic program regulated by E4F1 in basal keratinocytes and illustrate the importance of PDH activity in skin homeostasis.


Subject(s)
DNA-Binding Proteins/metabolism , Dihydrolipoyllysine-Residue Acetyltransferase/metabolism , Homeostasis , Mitochondrial Proteins/metabolism , Skin/metabolism , Transcription Factors/metabolism , Animals , Animals, Newborn , Basement Membrane/metabolism , Cell Adhesion , Cells, Cultured , Cellular Microenvironment , DNA-Binding Proteins/deficiency , Dihydrolipoyllysine-Residue Acetyltransferase/genetics , Epidermal Cells , Epidermis/metabolism , Gene Expression Regulation , Keratinocytes/cytology , Keratinocytes/metabolism , Mice, Knockout , Mitochondrial Proteins/genetics , Monocarboxylic Acid Transporters/metabolism , Muscle Proteins/metabolism , Pyruvates/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Repressor Proteins , Stem Cells/metabolism , Transcription Factors/deficiency , Ubiquitin-Protein Ligases
4.
Proc Natl Acad Sci U S A ; 113(39): 10998-1003, 2016 09 27.
Article in English | MEDLINE | ID: mdl-27621446

ABSTRACT

The mitochondrial pyruvate dehydrogenase (PDH) complex (PDC) acts as a central metabolic node that mediates pyruvate oxidation and fuels the tricarboxylic acid cycle to meet energy demand. Here, we reveal another level of regulation of the pyruvate oxidation pathway in mammals implicating the E4 transcription factor 1 (E4F1). E4F1 controls a set of four genes [dihydrolipoamide acetlytransferase (Dlat), dihydrolipoyl dehydrogenase (Dld), mitochondrial pyruvate carrier 1 (Mpc1), and solute carrier family 25 member 19 (Slc25a19)] involved in pyruvate oxidation and reported to be individually mutated in human metabolic syndromes. E4F1 dysfunction results in 80% decrease of PDH activity and alterations of pyruvate metabolism. Genetic inactivation of murine E4f1 in striated muscles results in viable animals that show low muscle PDH activity, severe endurance defects, and chronic lactic acidemia, recapitulating some clinical symptoms described in PDC-deficient patients. These phenotypes were attenuated by pharmacological stimulation of PDH or by a ketogenic diet, two treatments used for PDH deficiencies. Taken together, these data identify E4F1 as a master regulator of the PDC.


Subject(s)
DNA-Binding Proteins/metabolism , Pyruvate Dehydrogenase Complex/metabolism , Transcription Factors/metabolism , Transcription, Genetic , Animals , Base Sequence , DNA-Binding Proteins/deficiency , Diet, Ketogenic , Mice, Inbred C57BL , Mice, Knockout , Models, Biological , Muscle Fibers, Skeletal/metabolism , Muscle, Striated/metabolism , Phenotype , Pyruvic Acid/metabolism , Repressor Proteins , Transcription Factors/deficiency , Ubiquitin-Protein Ligases
5.
Nat Cell Biol ; 9(3): 331-8, 2007 Mar.
Article in English | MEDLINE | ID: mdl-17293853

ABSTRACT

The p300-CBP-associated factor (PCAF) is a histone acetyltransferase (HAT) involved in the reversible acetylation of various transcriptional regulators, including the tumour suppressor p53. It is implicated in many cellular processes, such as transcription, differentiation, proliferation and apoptosis. We observed that knockdown of PCAF expression in HeLa or U2OS cell lines induces stabilization of the oncoprotein Hdm2, a RING finger E3 ligase primarily known for its role in controlling p53 stability. To investigate the molecular basis of this effect, we examined whether PCAF is involved in Hdm2 ubiquitination. Here, we show that PCAF, in addition to its acetyltransferase activity, possesses an intrinsic ubiquitination activity that is critical for controlling Hdm2 expression levels, and thus p53 functions. Our data highlight a regulatory crosstalk between PCAF and Hdm2 activities, which is likely to have a central role in the subtle control of p53 activity after DNA damage.


Subject(s)
Cell Cycle Proteins/metabolism , Histone Acetyltransferases/metabolism , Proto-Oncogene Proteins c-mdm2/metabolism , Transcription Factors/metabolism , Ubiquitin-Protein Ligases/metabolism , Apoptosis/drug effects , Apoptosis/genetics , Apoptosis/radiation effects , Binding Sites/genetics , Catalytic Domain/genetics , Cell Cycle Proteins/genetics , Cell Line, Tumor , Cyclin-Dependent Kinase Inhibitor p21/genetics , DNA Damage/drug effects , DNA Damage/radiation effects , Gene Expression/drug effects , Gene Expression/radiation effects , HeLa Cells , Histone Acetyltransferases/genetics , Humans , Mutation , Proto-Oncogene Proteins c-bcl-2/genetics , Proto-Oncogene Proteins c-mdm2/genetics , RNA, Antisense/genetics , RNA, Small Interfering/genetics , Transcription Factors/genetics , Transfection , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism , Ubiquitin/metabolism , Ubiquitin-Protein Ligases/genetics , Ultraviolet Rays , Zinostatin/pharmacology , p300-CBP Transcription Factors
6.
Proc Natl Acad Sci U S A ; 107(49): 21076-81, 2010 Dec 07.
Article in English | MEDLINE | ID: mdl-21088222

ABSTRACT

A growing body of evidence suggests that the multifunctional protein E4F1 is involved in signaling pathways that play essential roles during normal development and tumorigenesis. We generated E4F1 conditional knockout mice to address E4F1 functions in vivo in newborn and adult skin. E4F1 inactivation in the entire skin or in the basal compartment of the epidermis induces skin homeostasis defects, as evidenced by transient hyperplasia in the interfollicular epithelium and alteration of keratinocyte differentiation, followed by loss of cellularity in the epidermis and severe skin ulcerations. E4F1 depletion alters clonogenic activity of epidermal stem cells (ESCs) ex vivo and ends in exhaustion of the ESC pool in vivo, indicating that the lesions observed in the E4F1 mutant skin result, at least in part, from cell-autonomous alterations in ESC maintenance. The clonogenic potential of E4F1 KO ESCs is rescued by Bmi1 overexpression or by Ink4a/Arf or p53 depletion. Skin phenotype of E4F1 KO mice is also delayed in animals with Ink4a/Arf and E4F1 compound gene deficiencies. Our data identify a regulatory axis essential for ESC-dependent skin homeostasis implicating E4F1 and the Bmi1-Arf-p53 pathway.


Subject(s)
DNA-Binding Proteins/physiology , Epidermal Cells , Homeostasis , Stem Cells/physiology , Transcription Factors/physiology , Age Factors , Animals , Cyclin-Dependent Kinase Inhibitor p16/metabolism , Mice , Mice, Knockout , Nuclear Proteins/metabolism , Phenotype , Polycomb Repressive Complex 1 , Proto-Oncogene Proteins/metabolism , Repressor Proteins/metabolism , Stem Cells/cytology , Tumor Suppressor Protein p53/metabolism , Ubiquitin-Protein Ligases
7.
Med Sci (Paris) ; 29(12): 1125-30, 2013 Dec.
Article in French | MEDLINE | ID: mdl-24356143

ABSTRACT

The p53 tumor suppressor is an essential downstream effector of various cellular stress response pathways that is functionally inactivated in most, if not all, tumors. Since its discovery more than 30 years ago, its role in the control of cell proliferation, senescence and cell survival has been widely described. However, growing evidences from several laboratories indicate that p53 has important transcriptional and non-transcriptional functions in the control of metabolism, including the regulation of glycolysis, glutaminolysis or mitochondrial respiration. Originally identified using in vitro cellular models, this previously underestimated role of p53 has been confirmed in vivo in various genetically engineered mouse models. These recent data suggest that p53 functions in various metabolic pathways significantly contribute to its role in adult tissue homeostasis, aging as well as tumor suppression.


Subject(s)
Metabolism/physiology , Tumor Suppressor Protein p53/physiology , Animals , Energy Metabolism/genetics , Energy Metabolism/physiology , Glycolysis/genetics , Glycolysis/physiology , Homeostasis , Humans , Lipid Metabolism/genetics , Lipid Metabolism/physiology , Metabolism/genetics , Mitochondria/metabolism
8.
Front Oncol ; 12: 857572, 2022.
Article in English | MEDLINE | ID: mdl-35494017

ABSTRACT

Cutaneous melanoma is a highly invasive tumor and, despite the development of recent therapies, most patients with advanced metastatic melanoma have a poor clinical outcome. The most frequent mutations in melanoma affect the BRAF oncogene, a protein kinase of the MAPK signaling pathway. Therapies targeting both BRAF and MEK are effective for only 50% of patients and, almost systematically, generate drug resistance. Genetic and non-genetic mechanisms associated with the strong heterogeneity and plasticity of melanoma cells have been suggested to favor drug resistance but are still poorly understood. Recently, we have introduced a novel mathematical formalism allowing the representation of the relation between tumor heterogeneity and drug resistance and proposed several models for the development of resistance of melanoma treated with BRAF/MEK inhibitors. In this paper, we further investigate this relationship by using a new computational model that copes with multiple cell states identified by single cell mRNA sequencing data in melanoma treated with BRAF/MEK inhibitors. We use this model to predict the outcome of different therapeutic strategies. The reference therapy, referred to as "continuous" consists in applying one or several drugs without disruption. In "combination therapy", several drugs are used sequentially. In "adaptive therapy" drug application is interrupted when the tumor size is below a lower threshold and resumed when the size goes over an upper threshold. We show that, counter-intuitively, the optimal protocol in combination therapy of BRAF/MEK inhibitors with a hypothetical drug targeting cell states that develop later during the tumor response to kinase inhibitors, is to treat first with this hypothetical drug. Also, even though there is little difference in the timing of emergence of the resistance between continuous and adaptive therapies, the spatial distribution of the different melanoma subpopulations is more zonated in the case of adaptive therapy.

9.
Cancers (Basel) ; 13(1)2021 Jan 04.
Article in English | MEDLINE | ID: mdl-33406607

ABSTRACT

The p53 pathway is functionally inactivated in most, if not all, human cancers. The p53 protein is a central effector of numerous stress-related molecular cascades. p53 controls a safeguard mechanism that prevents accumulation of abnormal cells and their transformation by regulating DNA repair, cell cycle progression, cell death, or senescence. The multiple cellular processes regulated by p53 were more recently extended to the control of metabolism and many studies support the notion that perturbations of p53-associated metabolic activities are linked to cancer development, as well as to other pathophysiological conditions including aging, type II diabetes, and liver disease. Although much less documented than p53 metabolic activities, converging lines of evidence indicate that other key components of this tumor suppressor pathway are also involved in cellular metabolism through p53-dependent as well as p53-independent mechanisms. Thus, at least from a metabolic standpoint, the p53 pathway must be considered as a non-linear pathway, but the complex metabolic network controlled by these p53 regulators and the mechanisms by which their activities are coordinated with p53 metabolic functions remain poorly understood. In this review, we highlight some of the metabolic pathways controlled by several central components of the p53 pathway and their role in tissue homeostasis, metabolic diseases, and cancer.

10.
Nat Commun ; 12(1): 7037, 2021 12 02.
Article in English | MEDLINE | ID: mdl-34857760

ABSTRACT

Growing evidence supports the importance of the p53 tumor suppressor in metabolism but the mechanisms underlying p53-mediated control of metabolism remain poorly understood. Here, we identify the multifunctional E4F1 protein as a key regulator of p53 metabolic functions in adipocytes. While E4F1 expression is upregulated during obesity, E4f1 inactivation in mouse adipose tissue results in a lean phenotype associated with insulin resistance and protection against induced obesity. Adipocytes lacking E4F1 activate a p53-dependent transcriptional program involved in lipid metabolism. The direct interaction between E4F1 and p53 and their co-recruitment to the Steaoryl-CoA Desaturase-1 locus play an important role to regulate monounsaturated fatty acids synthesis in adipocytes. Consistent with the role of this E4F1-p53-Steaoryl-CoA Desaturase-1 axis in adipocytes, p53 inactivation or diet complementation with oleate partly restore adiposity and improve insulin sensitivity in E4F1-deficient mice. Altogether, our findings identify a crosstalk between E4F1 and p53 in the control of lipid metabolism in adipocytes that is relevant to obesity and insulin resistance.


Subject(s)
Adipocytes/metabolism , Adipose Tissue/metabolism , Obesity/genetics , Repressor Proteins/genetics , Stearoyl-CoA Desaturase/genetics , Tumor Suppressor Protein p53/genetics , Ubiquitin-Protein Ligases/genetics , Adipocytes/pathology , Adipose Tissue/pathology , Adult , Aged , Animals , Body Mass Index , Fatty Acids, Monounsaturated/metabolism , Female , Gene Expression Regulation , Humans , Insulin Resistance , Lipid Metabolism/genetics , Male , Mice , Mice, Knockout , Middle Aged , Obesity/metabolism , Obesity/pathology , Repressor Proteins/deficiency , Repressor Proteins/metabolism , Signal Transduction , Stearoyl-CoA Desaturase/metabolism , Tumor Suppressor Protein p53/metabolism , Ubiquitin-Protein Ligases/deficiency , Ubiquitin-Protein Ligases/metabolism
11.
Mol Metab ; 33: 2-22, 2020 03.
Article in English | MEDLINE | ID: mdl-31685430

ABSTRACT

BACKGROUND: The TP53 gene is one of the most commonly inactivated tumor suppressors in human cancers. p53 functions during cancer progression have been linked to a variety of transcriptional and non-transcriptional activities that lead to the tight control of cell proliferation, senescence, DNA repair, and cell death. However, converging evidence indicates that p53 also plays a major role in metabolism in both normal and cancer cells. SCOPE OF REVIEW: We provide an overview of the current knowledge on the metabolic activities of wild type (WT) p53 and highlight some of the mechanisms by which p53 contributes to whole body energy homeostasis. We will also pinpoint some evidences suggesting that deregulation of p53-associated metabolic activities leads to human pathologies beyond cancer, including obesity, diabetes, liver, and cardiovascular diseases. MAJOR CONCLUSIONS: p53 is activated when cells are metabolically challenged but the origin, duration, and intensity of these stresses will dictate the outcome of the p53 response. p53 plays pivotal roles both upstream and downstream of several key metabolic regulators and is involved in multiple feedback-loops that ensure proper cellular homeostasis. The physiological roles of p53 in metabolism involve complex mechanisms of regulation implicating both cell autonomous effects as well as autocrine loops. However, the mechanisms by which p53 coordinates metabolism at the organismal level remain poorly understood. Perturbations of p53-regulated metabolic activities contribute to various metabolic disorders and are pivotal during cancer progression.


Subject(s)
Energy Metabolism/genetics , Metabolic Diseases/metabolism , Neoplasms/metabolism , Tumor Suppressor Protein p53/metabolism , Homeostasis/genetics , Humans , Metabolic Diseases/genetics , Metabolic Diseases/pathology , Neoplasms/genetics , Neoplasms/pathology , Signal Transduction/genetics , Tumor Suppressor Protein p53/genetics
12.
Mol Cell Biol ; 24(14): 6467-75, 2004 Jul.
Article in English | MEDLINE | ID: mdl-15226446

ABSTRACT

The ubiquitously expressed E4F protein was originally identified as an E1A-regulated cellular transcription factor required for adenovirus replication. The function of this protein in normal cell physiology remains largely unknown. To address this issue, we generated E4F knockout mice by gene targeting. Embryos lacking E4F die at the peri-implantation stage, while in vitro-cultured E4F(-/-) blastocysts exhibit defects in mitotic progression, chromosomal missegregation, and increased apoptosis. Consistent with these observations, we found that E4F localizes to the mitotic spindle during the M phase of early embryos. Our results establish a crucial role for E4F during early embryonic cell cycles and reveal an unexpected function for E4F in mitosis.


Subject(s)
Cell Cycle/physiology , DNA-Binding Proteins/metabolism , Embryo, Mammalian/physiology , Mitosis/physiology , Transcription Factors/metabolism , Animals , Cell Cycle Proteins , Cell Death , DNA-Binding Proteins/genetics , Gene Targeting , Gestational Age , Humans , In Situ Nick-End Labeling , Mice , Mice, Knockout , Protein Kinases/metabolism , Protein Serine-Threonine Kinases , Repressor Proteins , Transcription Factors/genetics , Ubiquitin-Protein Ligases
14.
J Dermatol Sci ; 88(1): 10-19, 2017 Oct.
Article in English | MEDLINE | ID: mdl-28728908

ABSTRACT

BACKGROUND: The ubiquitin proteasome pathway is involved in the pathogenesis of psoriasis and proteasome subunits are increased in lesional psoriatic skin. Recent works have highlighted that proteasome levels can be regulated through modulation of proteasome assembly notably by the proteasome maturation protein POMP. OBJECTIVES: To investigate whether proteasome assembly and POMP expression are modified in psoriatic skin. METHODS: Proteasome assembly as well as expression of proteasome regulators were assessed in non-lesional and lesional psoriatic skin using native gel electrophoresis and western blots respectively. The protein and mRNA expression levels of POMP were compared by western blots, immunohistochemistry and quantitative polymerase chain reaction. The role of POMP in keratinocyte proliferation and differentiation was assessed by silencing POMP gene expression by RNA interference in human immortalized keratinocyte HaCaT cells. RESULTS: Both 20S and 26S proteasomes (and their respective proteolytic activities) as well as the main proteasome regulators are increased in lesional psoriatic skin. POMP binds to 20S precursor complexes and is overexpressed in lesional epidermal psoriatic skin, supporting that POMP-mediated proteasome assembly is increased in psoriatic skin. POMP silencing inhibited HaCaT cell proliferation and induced apoptosis through the inhibition of the proteasome assembly. Moreover POMP partial depletion decreased the expression of the differentiation markers keratin 10 and involucrin during the [Ca2+]-induced HaCaT cells differentiation. CONCLUSION: Altogether these results establish a potential role for POMP and proteasome assembly in psoriasis pathogenesis.


Subject(s)
Keratinocytes/pathology , Molecular Chaperones/metabolism , Proteasome Endopeptidase Complex/metabolism , Psoriasis/pathology , RNA, Messenger/metabolism , Apoptosis , Biopsy , Blotting, Western , Cell Differentiation , Cell Line , Cell Proliferation , Cytoplasm , Epidermal Cells , Epidermis/pathology , Humans , Keratinocytes/metabolism , Molecular Chaperones/genetics , Native Polyacrylamide Gel Electrophoresis , RNA Interference , RNA, Small Interfering/metabolism
15.
Oncogene ; 22(2): 256-65, 2003 Jan 16.
Article in English | MEDLINE | ID: mdl-12527894

ABSTRACT

Estrogen-receptor (ER) status is an important parameter in breast cancer management as ER-positive breast cancers have a better prognosis than ER-negative tumors. This difference comes essentially from the lower aggressiveness and invasiveness of ER-positive tumors. Here, we demonstrate, that interleukin-8 (IL-8) was clearly overexpressed in most ER-negative breast, ovary cell lines and breast tumor samples tested, whereas no significant IL-8 level could be detected in ER-positive breast or ovarian cell lines. We have also cloned human IL-8 from ER-negative MDA-MB-231 cells, and we show that IL-8 produced by breast cancer cells is identical to monocyte-derived IL-8. Interestingly, the invasion potential of ER-negative breast cancer cells is associated at least in part with expression of IL-8, but not with IL-8 receptor levels. Moreover, IL-8 increases the invasiveness of ER-positive breast cancer cells by two fold, thus confirming the invasion-promoting role of IL-8. On the other hand, exogenous expression of estrogen receptors in ER-negative cells led to a decrease of IL-8 levels. In summary, our data show that IL-8 expression is negatively linked to ER status of breast and ovarian cancer cells. We also support the idea that IL-8 expression is associated with a higher invasiveness potential of cancer cells in vitro, which suggests that IL-8 could be a novel marker of tumor aggressiveness.


Subject(s)
Breast Neoplasms/metabolism , Carcinoma, Ductal, Breast/metabolism , Interleukin-18/metabolism , Receptors, Estrogen/metabolism , Amino Acid Sequence , Base Sequence , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Carcinogenicity Tests , Carcinoma, Ductal, Breast/genetics , Carcinoma, Ductal, Breast/pathology , Cell Division/drug effects , Cell Division/genetics , Female , Gene Expression Regulation, Neoplastic , Humans , Interleukin-18/genetics , Interleukin-18/pharmacology , Molecular Sequence Data , Neoplasm Invasiveness , Tumor Cells, Cultured
17.
J Invest Dermatol ; 135(11): 2732-2741, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26176762

ABSTRACT

IL-20 is involved in the development of skin psoriasis. The molecular mechanisms underlying IL-20 overexpression in psoriatic epidermis remain to be elucidated. We showed that IL-20 was primarily upregulated in psoriatic skin at the post-transcriptional level. The RNA-binding protein HuR relocalized to the cytoplasm of keratinocytes (KCs) of psoriatic patients, suggesting that it stabilizes numerous transcripts, as observed in the human KC cell lines used to assess IL-20 mRNA. We characterized epidermal HuR RNA targets in psoriatic skin using ribonucleoprotein immunoprecipitation analyzed via high-throughput sequencing. Numerous transcripts that are upregulated in psoriasis were targeted by HuR, supporting the participation of HuR in pathogenic processes such as morphological changes, innate and adaptive immune responses, and metabolic inflammatory responses. Finally, we identified the metabolic sensor AMP-activated protein kinase (AMPK) as being responsible for HuR cytoplasmic relocalization because its activity was severely impaired in human psoriatic epidermis, and in vivo drug-mediated AMPK inhibition in mouse epidermis promoted HuR cytoplasmic localization, IL-20 overproduction, acanthosis, and hyperkeratosis. These results provide insights into the molecular links between metabolism and post-transcriptional networks during chronic inflammation.


Subject(s)
AMP-Activated Protein Kinases/metabolism , ELAV-Like Protein 1/metabolism , Gene Expression Regulation , Interleukins/genetics , Psoriasis/genetics , Psoriasis/pathology , AMP-Activated Protein Kinases/genetics , Animals , Biopsy, Needle , Cells, Cultured , Disease Models, Animal , ELAV-Like Protein 1/genetics , Humans , Immunohistochemistry , Keratinocytes/cytology , Keratinocytes/metabolism , Mice , Mice, Inbred C57BL , RNA Interference , RNA, Messenger/genetics , Random Allocation , Real-Time Polymerase Chain Reaction/methods , Skin/cytology , Skin/pathology , Statistics, Nonparametric , Up-Regulation
18.
Cell Rep ; 11(2): 220-33, 2015 Apr 14.
Article in English | MEDLINE | ID: mdl-25843721

ABSTRACT

Recent data support the notion that a group of key transcriptional regulators involved in tumorigenesis, including MYC, p53, E2F1, and BMI1, share an intriguing capacity to simultaneously regulate metabolism and cell cycle. Here, we show that another factor, the multifunctional protein E4F1, directly controls genes involved in mitochondria functions and cell-cycle checkpoints, including Chek1, a major component of the DNA damage response. Coordination of these cellular functions by E4F1 appears essential for the survival of p53-deficient transformed cells. Acute inactivation of E4F1 in these cells results in CHK1-dependent checkpoint deficiency and multiple mitochondrial dysfunctions that lead to increased ROS production, energy stress, and inhibition of de novo pyrimidine synthesis. This deadly cocktail leads to the accumulation of uncompensated oxidative damage to proteins and extensive DNA damage, ending in cell death. This supports the rationale of therapeutic strategies simultaneously targeting mitochondria and CHK1 for selective killing of p53-deficient cancer cells.


Subject(s)
DNA-Binding Proteins/genetics , Mitochondria/metabolism , Neoplasms/genetics , Protein Kinases/genetics , Transcription Factors/genetics , Tumor Suppressor Protein p53/genetics , Animals , Cell Survival , Checkpoint Kinase 1 , DNA Damage/genetics , DNA-Binding Proteins/biosynthesis , Gene Expression Regulation, Neoplastic , Humans , Mice , Mitochondria/pathology , Mouse Embryonic Stem Cells/metabolism , Neoplasms/metabolism , Protein Kinases/biosynthesis , Pyrimidines/biosynthesis , Repressor Proteins , Stress, Physiological/genetics , Transcription Factors/biosynthesis , Tumor Suppressor Protein p53/biosynthesis , Ubiquitin-Protein Ligases
19.
J Invest Dermatol ; 132(3 Pt 1): 575-82, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22089833

ABSTRACT

Netherton syndrome (NS) is a severe skin disease caused by loss-of-function mutations in SPINK5 (serine protease inhibitor Kazal-type 5) encoding the serine protease inhibitor LEKTI (lympho-epithelial Kazal type-related inhibitor). Here, we disclose new SPINK5 defects in 12 patients, who presented a clinical triad suggestive of NS with variations in inter- and intra-familial disease expression. We identified a new and frequent synonymous mutation c.891C>T (p.Cys297Cys) in exon 11 of the 12 NS patients. This mutation disrupts an exonic splicing enhancer sequence and causes out-of-frame skipping of exon 11. Haplotype analysis indicates that this mutation is a founder mutation in Greece. Two other new deep intronic mutations, c.283-12T>A in intron 4 and c.1820+53G>A in intron 19, induced partial intronic sequence retention. A new nonsense c.2557C>T (p.Arg853X) mutation was also identified. All mutations led to a premature termination codon resulting in no detectable LEKTI on skin sections. Two patients with deep intronic mutations showed residual LEKTI fragments in cultured keratinocytes. These fragments retained some functional activity, and could therefore, together with other determinants, contribute to modulate the disease phenotype. This new founder mutation, the most frequent mutation described in European populations so far, and these unusual intronic mutations, widen the clinical and molecular spectrum of NS and offer new diagnostic perspectives for NS patients.


Subject(s)
Founder Effect , Introns/genetics , Mutation , Netherton Syndrome/diagnosis , Netherton Syndrome/genetics , Proteinase Inhibitory Proteins, Secretory/genetics , RNA Splicing/genetics , Adolescent , Adult , Base Sequence , Child , Child, Preschool , Codon, Nonsense/genetics , Exons/genetics , Female , Humans , Infant , Male , Molecular Sequence Data , Serine Peptidase Inhibitor Kazal-Type 5
20.
J Exp Med ; 208(7): 1403-17, 2011 Jul 04.
Article in English | MEDLINE | ID: mdl-21708927

ABSTRACT

The multifunctional E4F1 protein was originally discovered as a target of the E1A viral oncoprotein. Growing evidence indicates that E4F1 is involved in key signaling pathways commonly deregulated during cell transformation. In this study, we investigate the influence of E4F1 on tumorigenesis. Wild-type mice injected with fetal liver cells from mice lacking CDKN2A, the gene encoding Ink4a/Arf, developed histiocytic sarcomas (HSs), a tumor originating from the monocytic/macrophagic lineage. Cre-mediated deletion of E4F1 resulted in the death of HS cells and tumor regression in vivo and extended the lifespan of recipient animals. In murine and human HS cell lines, E4F1 inactivation resulted in mitochondrial defects and increased production of reactive oxygen species (ROS) that triggered massive cell death. Notably, these defects of E4F1 depletion were observed in HS cells but not healthy primary macrophages. Short hairpin RNA-mediated depletion of E4F1 induced mitochondrial defects and ROS-mediated death in several human myeloid leukemia cell lines. E4F1 protein is overexpressed in a large subset of human acute myeloid leukemia samples. Together, these data reveal a role for E4F1 in the survival of myeloid leukemic cells and support the notion that targeting E4F1 activities might have therapeutic interest.


Subject(s)
DNA-Binding Proteins/deficiency , Leukemia, Myeloid, Acute/metabolism , Leukemia, Myeloid, Acute/pathology , Repressor Proteins/deficiency , Transcription Factors/deficiency , Animals , Autophagy/physiology , Base Sequence , Cell Death/physiology , Cell Line, Tumor , Cell Transformation, Neoplastic , DNA-Binding Proteins/antagonists & inhibitors , DNA-Binding Proteins/genetics , Histiocytic Sarcoma/genetics , Histiocytic Sarcoma/metabolism , Histiocytic Sarcoma/pathology , Humans , Macrophages/metabolism , Macrophages/pathology , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Inbred DBA , Mice, Knockout , Mice, Transgenic , Oxidative Stress , RNA, Small Interfering/genetics , Reactive Oxygen Species/metabolism , Repressor Proteins/antagonists & inhibitors , Repressor Proteins/genetics , Signal Transduction , Transcription Factors/antagonists & inhibitors , Transcription Factors/genetics , Ubiquitin-Protein Ligases
SELECTION OF CITATIONS
SEARCH DETAIL