Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
Add more filters










Publication year range
2.
Bioelectromagnetics ; 45(3): 110-129, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38115173

ABSTRACT

Whether human cells are impacted by environmental electromagnetic fields (EMF) is still a matter of debate. With the deployment of the fifth generation (5G) of mobile communication technologies, the carrier frequency is increasing and the human skin becomes the main biological target. Here, we evaluated the impact of 5G-modulated 3.5 GHz radiofrequency (RF) EMF on mitochondrial stress in human fibroblasts and keratinocytes that were exposed for 24 h at specific absorption rate of 0.25, 1, and 4 W/kg. We assessed cell viability, mitochondrial reactive oxygen species (ROS) production, and membrane polarization. Knowing that human skin is the main target of environmental ultraviolet (UV), using the same read-out, we investigated whether subsequent exposure to 5G signal could alter the capacity of UV-B to damage skin cells. We found a statistically significant reduction in mitochondrial ROS concentration in fibroblasts exposed to 5G signal at 1 W/kg. On the contrary, the RF exposure slightly but statistically significantly enhanced the effects of UV-B radiation specifically in keratinocytes at 0.25 and 1 W/kg. No effect was found on mitochondrial membrane potential or apoptosis in any cell types or exposure conditions suggesting that the type and amplitude of the observed effects are very punctual.


Subject(s)
Skin , Ultraviolet Rays , Humans , Reactive Oxygen Species/metabolism , Ultraviolet Rays/adverse effects , Skin/metabolism , Radio Waves/adverse effects , Keratinocytes/metabolism , Electromagnetic Fields
3.
Cell Rep ; 42(6): 112579, 2023 06 27.
Article in English | MEDLINE | ID: mdl-37267103

ABSTRACT

In mammals, about 99% of mitochondrial proteins are synthesized in the cytosol as precursors that are subsequently imported into the organelle. The mitochondrial health and functions rely on an accurate quality control of these imported proteins. Here, we show that the E3 ubiquitin ligase F box/leucine-rich-repeat protein 6 (FBXL6) regulates the quality of cytosolically translated mitochondrial proteins. Indeed, we found that FBXL6 substrates are newly synthesized mitochondrial ribosomal proteins. This E3 binds to chaperones involved in the folding and trafficking of newly synthesized peptide and to ribosomal-associated quality control proteins. Deletion of these interacting partners is sufficient to hamper interactions between FBXL6 and its substrate. Furthermore, we show that cells lacking FBXL6 fail to degrade specifically mistranslated mitochondrial ribosomal proteins. Finally, showing the role of FBXL6-dependent mechanism, FBXL6-knockout (KO) cells display mitochondrial ribosomal protein aggregations, altered mitochondrial metabolism, and inhibited cell cycle in oxidative conditions.


Subject(s)
Ribosomal Proteins , Ubiquitin-Protein Ligases , Mammals/metabolism , Mitochondria/metabolism , Mitochondrial Proteins/metabolism , Protein Domains , Ribosomal Proteins/metabolism , Ubiquitin-Protein Ligases/metabolism , Humans
4.
Cancers (Basel) ; 15(1)2022 Dec 20.
Article in English | MEDLINE | ID: mdl-36612001

ABSTRACT

Alterations in lipid handling are an important hallmark in cancer. Our aim here is to target key metabolic enzymes to reshape the oncogenic lipid metabolism triggering irreversible cell breakdown. We targeted the key metabolic player proprotein convertase subtilisin/kexin type 9 (PCSK9) using a pharmacological inhibitor (R-IMPP) alone or in combination with 3-hydroxy 3-methylglutaryl-Coenzyme A reductase (HMGCR) inhibitor, simvastatin. We assessed the effect of these treatments using 3 hepatoma cell lines, Huh6, Huh7 and HepG2 and a tumor xenograft in chicken choriorallantoic membrane (CAM) model. PCSK9 deficiency led to dose-dependent inhibition of cell proliferation in all cell lines and a decrease in cell migration. Co-treatment with simvastatin presented synergetic anti-proliferative effects. At the metabolic level, mitochondrial respiration assays as well as the assessment of glucose and glutamine consumption showed higher metabolic adaptability and surge in the absence of PCSK9. Enhanced lipid uptake and biogenesis led to excessive accumulation of intracellular lipid droplets as revealed by electron microscopy and metabolic tracing. Using xenograft experiments in CAM model, we further demonstrated the effect of anti-PCSK9 treatment in reducing tumor aggressiveness. Targeting PCSK9 alone or in combination with statins deserves to be considered as a new therapeutic option in liver cancer clinical applications.

5.
Front Genet ; 11: 561687, 2020.
Article in English | MEDLINE | ID: mdl-33329698

ABSTRACT

Xeroderma Pigmentosum C (XPC) is a multi-functional protein that is involved not only in the repair of bulky lesions, post-irradiation, via nucleotide excision repair (NER) per se but also in oxidative DNA damage mending. Since base excision repair (BER) is the primary regulator of oxidative DNA damage, we characterized, post-Ultraviolet B-rays (UVB)-irradiation, the detailed effect of three different XPC mutations in primary fibroblasts derived from XP-C patients on mRNA, protein expression and activity of different BER factors. We found that XP-C fibroblasts are characterized by downregulated expression of different BER factors including OGG1, MYH, APE1, LIG3, XRCC1, and Polß. Such a downregulation was also observed at OGG1, MYH, and APE1 protein levels. This was accompanied with an increase in DNA oxidative lesions, as evidenced by 8-oxoguanine levels, immediately post-UVB-irradiation. Unlike in normal control cells, these oxidative lesions persisted over time in XP-C cells having lower excision repair capacities. Taken together, our results indicated that an impaired BER pathway in XP-C fibroblasts leads to longer persistence and delayed repair of oxidative DNA damage. This might explain the diverse clinical phenotypes in XP-C patients suffering from cancer in both photo-protected and photo-exposed areas. Therapeutic strategies based on reinforcement of BER pathway might therefore represent an innovative path for limiting the drawbacks of NER-based diseases, as in XP-C case.

6.
Sci Rep ; 10(1): 12246, 2020 07 22.
Article in English | MEDLINE | ID: mdl-32699238

ABSTRACT

Developing trustworthy, cost effective, minimally or non-invasive glucose sensing strategies is of great need for diabetic patients. In this study, we used an experimental type I diabetic mouse model to examine whether the skin would provide novel means for identifying biomarkers associated with blood glucose level. We first showed that skin glucose levels are rapidly influenced by blood glucose concentrations. We then conducted a proteomic screen of murine skin using an experimental in vivo model of type I diabetes and wild-type controls. Among the proteins that increased expression in response to high blood glucose, Trisk 95 expression was significantly induced independently of insulin signalling. A luciferase reporter assay demonstrated that the induction of Trisk 95 expression occurs at a transcriptional level and is associated with a marked elevation in the Fluo-4AM signal, suggesting a role for intracellular calcium changes in the signalling cascade. Strikingly, these changes lead concurrently to fragmentation of the mitochondria. Moreover, Trisk 95 knockout abolishes both the calcium flux and the mitochondrial phenotype changes indicating dependency of glucose flux in the skin on Trisk 95 function. The data demonstrate that the skin reacts robustly to systemic blood changes, and that Trisk 95 is a promising biomarker for a glucose monitoring assembly.


Subject(s)
Carrier Proteins/metabolism , Diabetes Mellitus/metabolism , Glucose/metabolism , Skin/metabolism , Animals , Biomarkers/metabolism , Blood Glucose/metabolism , Blood Glucose Self-Monitoring/methods , Calcium Signaling/physiology , Cells, Cultured , Insulin/metabolism , Keratinocytes/metabolism , Mice , Mice, Inbred C57BL , Mitochondria/metabolism , Proteomics/methods , Transcription, Genetic/physiology
7.
Antioxid Redox Signal ; 33(13): 883-902, 2020 11 01.
Article in English | MEDLINE | ID: mdl-32475148

ABSTRACT

Aims: REDOX signaling from reactive oxygen species (ROS) generated by the mitochondria (mitochondrial reactive oxygen species [mtROS]) has been implicated in cancer growth and survival. Here, we investigated the effect of 5-(4-methoxyphenyl)-3H-1,2-dithiole-3-thione (AOL), a recently characterized member of the new class of mtROS suppressors (S1QELs), on human lung adenocarcinoma proteome reprogramming, bioenergetics, and growth. Results: AOL reduced steady-state cellular ROS levels in human lung cancer cells without altering the catalytic activity of complex I. AOL treatment induced dose-dependent inhibition of lung cancer cell proliferation and triggered a reduction in tumor growth in vivo. Molecular investigations demonstrated that AOL reprogrammed the proteome of human lung cancer cells. In particular, AOL suppressed the determinants of the Warburg effect and increased the expression of the complex I subunit NDUFV1 which was also identified as AOL binding site using molecular modeling computer simulations. Comparison of the molecular changes induced by AOL and MitoTEMPO, an mtROS scavenger that is not an S1QEL, identified a core component of 217 proteins commonly altered by the two treatments, as well as drug-specific targets. Innovation: This study provides proof-of-concept data on the anticancer effect of AOL on mouse orthotopic human lung tumors. A unique dataset on proteomic reprogramming by AOL and MitoTEMPO is also provided. Lastly, our study revealed the repression of NDUFV1 by S1QEL AOL. Conclusion: Our findings demonstrate the preclinical anticancer properties of S1QEL AOL and delineate its mode of action on REDOX and cancer signaling.


Subject(s)
Adenocarcinoma of Lung/etiology , Adenocarcinoma of Lung/metabolism , Mitochondria/metabolism , Oxidation-Reduction , Reactive Oxygen Species/metabolism , Signal Transduction , Adenocarcinoma of Lung/drug therapy , Adenocarcinoma of Lung/pathology , Cyclic N-Oxides/metabolism , Electron Transport Complex I/metabolism , Humans
8.
Oncogenesis ; 8(10): 52, 2019 Sep 24.
Article in English | MEDLINE | ID: mdl-31551419

ABSTRACT

The leading cause of cutaneous squamous cell carcinomas (cSCCs) is exposure to ultraviolet radiation (UV). Unlike most other cancers, the incidence rates of cSCCs are still on the rise and the treatment options currently available are limited. We have recently found that dihydroorotate dehydrogenase (DHODH), which is the rate-limiting enzyme in the de novo pyrimidine synthesis pathway, plays a critical role in UVB-induced energy metabolism reprogramming. Using a multistage model of UVB radiation-induced skin cancer, we show that UVB-induced DHODH upregulation is mainly regulated transcriptionally by STAT3. Our results indicate that chronic inhibition of DHODH by leflunomide (LFN) blocks UVB-induced tumor initiation. Human tumor xenograft studies showed that LFN treatment reduces growth of established tumors when used in combination with a genotoxic agent, 5-fluorouracil (5-FU). Our data suggest that DHODH is a promising target for chemoprevention and combination therapy of UVB-induced cSCCs.

9.
J Invest Dermatol ; 139(9): 2016-2028.e7, 2019 09.
Article in English | MEDLINE | ID: mdl-30878676

ABSTRACT

HIF-1α is constitutively expressed in mouse and human epidermis. It plays a crucial role in skin physiology, including the response of keratinocytes to UVR. However, little information is available about its role in photocarcinogenesis. Using a multistage model of UVB radiation-induced skin cancer, we show that the knockout of Hif-1α in the epidermis prevents tumorigenesis but at the same time triggers the formation of hyperkeratotic plaques. Our results indicate that the absence of oncogenic transformation in Hif-1α-ablated mice is related to increased DNA repair in keratinocytes, whereas the formation of hyperkeratotic plaques is caused by an increase in the levels of reactive oxygen species. Indeed, impairing the DNA repair machinery by ablating xeroderma pigmentosum C restored the UVB-induced neoplastic transformation of Hif-1α-ablated keratinocytes, whereas the development of hyperkeratotic plaques was blocked by chronic antioxidant treatment. We conclude that HIF-1α plays a procarcinogenic role in UVB-induced tumorigenesis.


Subject(s)
Carcinogenesis/pathology , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Keratosis, Actinic/pathology , Skin Neoplasms/pathology , Ultraviolet Rays/adverse effects , Animals , Apoptosis/genetics , Apoptosis/radiation effects , Carcinogenesis/radiation effects , DNA Damage/radiation effects , DNA Repair/genetics , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Epidermis/pathology , Epidermis/radiation effects , Female , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Keratinocytes/pathology , Keratinocytes/radiation effects , Keratosis, Actinic/etiology , Mice , Mice, Knockout , Neoplasms, Experimental/etiology , Neoplasms, Experimental/pathology , Oxidative Stress/genetics , Oxidative Stress/radiation effects , Skin Neoplasms/etiology
10.
Cell Rep ; 23(12): 3621-3634, 2018 06 19.
Article in English | MEDLINE | ID: mdl-29925003

ABSTRACT

Although growing evidence indicates that bioenergetic metabolism plays an important role in the progression of tumorigenesis, little information is available on the contribution of reprogramming of energy metabolism in cancer initiation. By applying a quantitative proteomic approach and targeted metabolomics, we find that specific metabolic modifications precede primary skin tumor formation. Using a multistage model of ultraviolet B (UVB) radiation-induced skin cancer, we show that glycolysis, tricarboxylic acid (TCA) cycle, and fatty acid ß-oxidation are decreased at a very early stage of photocarcinogenesis, while the distal part of the electron transport chain (ETC) is upregulated. Reductive glutamine metabolism and the activity of dihydroorotate dehydrogenase (DHODH) are both necessary for maintaining high ETC. Mice with decreased DHODH activity or impaired ETC failed to develop pre-malignant and malignant lesions. DHODH activity represents a major link between DNA repair efficiency and bioenergetic patterning during skin carcinogenesis.


Subject(s)
Carcinogenesis/metabolism , Carcinogenesis/radiation effects , Energy Metabolism/radiation effects , Skin Neoplasms/metabolism , Skin Neoplasms/pathology , Ultraviolet Rays , Animals , DNA-Binding Proteins/metabolism , Dihydroorotate Dehydrogenase , Down-Regulation/radiation effects , Electron Transport/radiation effects , Epidermis/pathology , Epidermis/radiation effects , Glutamine/metabolism , High Mobility Group Proteins/metabolism , Keratinocytes/metabolism , Keratinocytes/pathology , Keratinocytes/radiation effects , Metabolic Networks and Pathways , Mice , Mice, Hairless , Oxidoreductases Acting on CH-CH Group Donors/metabolism , Phenotype , Up-Regulation/radiation effects
12.
J Invest Dermatol ; 137(6): 1311-1321, 2017 06.
Article in English | MEDLINE | ID: mdl-28132856

ABSTRACT

The nicotinamide adenine dinucleotide phosphate oxidase (NOX) family enzymes are involved in several physiological functions. However, their roles in keratinocyte responses to UV radiation have not been clearly elucidated. This study shows that, among other NOX family members, UVB irradiation results in a biphasic activation of NOX1 that plays a critical role in defining keratinocyte fate through the modulation of the DNA damage response network. Indeed, suppression of both bursts of UVB-induced NOX1 activation by using a specific peptide inhibitor of NOX1 (InhNOX1) is associated with increased nucleotide excision repair efficiency and reduction of apoptosis, which is finally translated into decreased photocarcinogenesis. On the contrary, when only the second peak of UVB-induced NOX1 activation is blocked, both nucleotide excision repair efficiency and apoptosis are decreased. Our results show that inhibition of NOX1 activation could be a promising target for the prevention and treatment of UVB-induced skin cancer in nucleotide excision repair-proficient and -deficient patients.


Subject(s)
Carcinogenesis/radiation effects , Keratinocytes/radiation effects , NADH, NADPH Oxidoreductases/genetics , NADH, NADPH Oxidoreductases/radiation effects , NADPH Oxidases/drug effects , Ultraviolet Rays/adverse effects , Animals , Apoptosis/genetics , Apoptosis/radiation effects , Cells, Cultured , Disease Models, Animal , Female , Keratinocytes/cytology , Mice , Mice, Hairless , Mice, Transgenic , Molecular Targeted Therapy , NADPH Oxidase 1 , NADPH Oxidases/metabolism , Neoplasms, Radiation-Induced/physiopathology , Neoplasms, Radiation-Induced/prevention & control , Pyrazoles/pharmacology , Pyrazolones , Pyridines/pharmacology , Pyridones , Random Allocation , Risk Factors , Skin Neoplasms/etiology , Skin Neoplasms/physiopathology
13.
J Invest Dermatol ; 135(4): 1108-1118, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25437426

ABSTRACT

Xeroderma pigmentosum type C (XP-C) is characterized mostly by a predisposition to skin cancers and accelerated photoaging, but little is known about premature skin aging in this disease. By comparing young and old mice, we found that the level of progerin and p16(INK4a) expression, ß-galactosidase activity, and reactive oxygen species, which increase with age, were higher in young Xpc(-/-) mice than in young Xpc(+/+) ones. The expression level of mitochondrial complexes and mitochondrial functions in the skin of young Xpc(-/-) was as low as in control aged Xpc(+/+)animals. Furthermore, the metabolic profile in young Xpc(-/-) mice resembled that found in aged Xpc(+/+) mice. Furthermore, premature skin aging features in young Xpc(-/-) mice were mostly rescued by inhibition of nicotinamide adenine dinucleotide phosphate oxidase 1 (NOX1) activity by using a NOX1 peptide inhibitor, suggesting that the continuous oxidative stress due to overactivation of NOX1 has a causative role in the underlying pathophysiology.


Subject(s)
DNA-Binding Proteins/genetics , NADH, NADPH Oxidoreductases/antagonists & inhibitors , NADH, NADPH Oxidoreductases/metabolism , Nuclear Proteins/metabolism , Protein Precursors/metabolism , Skin Aging , Adaptor Proteins, Signal Transducing , Animals , Cyclin-Dependent Kinase Inhibitor p16/metabolism , Humans , Keratinocytes/cytology , Lamin Type A , Light , Mice , Mice, Knockout , Mitochondria/metabolism , NADPH Oxidase 1 , Oxidative Stress , Proteins/genetics , Reactive Oxygen Species/metabolism , Xeroderma Pigmentosum/metabolism , beta-Galactosidase/metabolism
14.
PLoS One ; 9(5): e97245, 2014.
Article in English | MEDLINE | ID: mdl-24824222

ABSTRACT

p38 mitogen-activated protein kinases (MAPKs) respond to a wide range of extracellular stimuli. While the inhibition of p38 signaling is implicated in the impaired capacity to repair ultraviolet (UV)-induced DNA damage-a primary risk factor for human skin cancers-its mechanism of action in skin carcinogenesis remains unclear, as both anti-proliferative and survival functions have been previously described. In this study, we utilized cultured keratinocytes, murine tumorigenesis models, and human cutaneous squamous cell carcinoma (SCC) specimens to assess the effect of p38 in this regard. UV irradiation of normal human keratinocytes increased the expression of all four p38 isoforms (α/ß/γ/δ); whereas irradiation of p53-deficient A431 keratinocytes derived from a human SCC selectively decreased p38α, without affecting other isoforms. p38α levels are decreased in the majority of human cutaneous SCCs assessed by tissue microarray, suggesting a tumor-suppressive effect of p38α in SCC pathogenesis. Genetic and pharmacological inhibition of p38α and in A431 cells increased cell proliferation, which was in turn associated with increases in NAPDH oxidase (NOX2) activity as well as intracellular reactive oxygen species (ROS). These changes led to enhanced invasiveness of A431 cells as assessed by the matrigel invasion assay. Chronic treatment of p53-/-/SKH-1 mice with the p38 inhibitor SB203580 accelerated UV-induced SCC carcinogenesis and increased the expression of NOX2. NOX2 knockdown suppressed the augmented growth of A431 xenografts treated with SB203580. These findings indicate that in the absence of p53, p38α deficiency drives SCC growth and progression that is associated with enhanced NOX2 expression and ROS formation.


Subject(s)
Carcinoma, Squamous Cell/metabolism , Gene Expression Regulation, Enzymologic/radiation effects , MAP Kinase Signaling System/physiology , Reactive Oxygen Species/metabolism , Skin Neoplasms/metabolism , Ultraviolet Rays , Animals , Carcinogenesis/drug effects , Cell Line, Tumor , Cell Proliferation , Cells, Cultured , Humans , Imidazoles/adverse effects , Keratinocytes/metabolism , Keratinocytes/radiation effects , MAP Kinase Signaling System/radiation effects , Membrane Glycoproteins/metabolism , Mice , Mice, Knockout , Microarray Analysis , NADPH Oxidase 2 , NADPH Oxidases/metabolism , Pyridines/adverse effects , Tumor Suppressor Protein p53/deficiency
15.
Cell Metab ; 17(5): 719-30, 2013 May 07.
Article in English | MEDLINE | ID: mdl-23602449

ABSTRACT

Mitophagy has been recently described as a mechanism of elimination of damaged organelles. Although the regulation of the amount of mitochondria is a core issue concerning cellular energy homeostasis, the relationship between mitochondrial degradation and energetic activity has not yet been considered. Here, we report that the stimulation of mitochondrial oxidative phosphorylation enhances mitochondrial renewal by increasing its degradation rate. Upon high oxidative phosphorylation activity, we found that the small GTPase Rheb is recruited to the mitochondrial outer membrane. This mitochondrial localization of Rheb promotes mitophagy through a physical interaction with the mitochondrial autophagic receptor Nix and the autophagosomal protein LC3-II. Thus, Rheb-dependent mitophagy contributes to the maintenance of optimal mitochondrial energy production. Our data suggest that mitochondrial degradation contributes to a bulk renewal of the organelle in order to prevent mitochondrial aging and to maintain the efficiency of oxidative phosphorylation.


Subject(s)
Mitochondria/physiology , Mitochondrial Proteins/metabolism , Mitophagy/physiology , Animals , Autophagy/physiology , HeLa Cells , Humans , Membrane Proteins/metabolism , Mice , Mice, Inbred C57BL , Mitochondria/metabolism , Mitochondrial Membranes/metabolism , Monomeric GTP-Binding Proteins/metabolism , Oxidative Phosphorylation
16.
J Clin Invest ; 121(1): 195-211, 2011 Jan.
Article in English | MEDLINE | ID: mdl-21123941

ABSTRACT

DNA damage is a well-known initiator of tumorigenesis. Studies have shown that most cancer cells rely on aerobic glycolysis for their bioenergetics. We sought to identify a molecular link between genomic mutations and metabolic alterations in neoplastic transformation. We took advantage of the intrinsic genomic instability arising in xeroderma pigmentosum C (XPC). The XPC protein plays a key role in recognizing DNA damage in nucleotide excision repair, and patients with XPC deficiency have increased incidence of skin cancer and other malignancies. In cultured human keratinocytes, we showed that lentivirus-mediated knockdown of XPC reduced mitochondrial oxidative phosphorylation and increased glycolysis, recapitulating cancer cell metabolism. Accumulation of unrepaired DNA following XPC silencing increased DNA-dependent protein kinase activity, which subsequently activated AKT1 and NADPH oxidase-1 (NOX1), resulting in ROS production and accumulation of specific deletions in mitochondrial DNA (mtDNA) over time. Subcutaneous injection of XPC-deficient keratinocytes into immunodeficient mice led to squamous cell carcinoma formation, demonstrating the tumorigenic potential of transduced cells. Conversely, simultaneous knockdown of either NOX1 or AKT1 blocked the neoplastic transformation induced by XPC silencing. Our results demonstrate that genomic instability resulting from XPC silencing results in activation of AKT1 and subsequently NOX1 to induce ROS generation, mtDNA deletions, and neoplastic transformation in human keratinocytes.


Subject(s)
Carcinoma, Squamous Cell/etiology , Cell Transformation, Neoplastic/genetics , DNA-Binding Proteins/antagonists & inhibitors , DNA-Binding Proteins/genetics , Keratinocytes/metabolism , Skin Neoplasms/etiology , Animals , Carcinoma, Squamous Cell/genetics , Carcinoma, Squamous Cell/metabolism , Carcinoma, Squamous Cell/pathology , Cell Proliferation , DNA Damage , DNA Repair , DNA, Mitochondrial/genetics , DNA, Mitochondrial/metabolism , Energy Metabolism , Gene Knockdown Techniques , Gene Silencing , Humans , Keratinocytes/pathology , Keratinocytes/transplantation , Mice , Mice, Inbred NOD , Mice, SCID , Mitochondria/metabolism , Models, Biological , Mutation , NADPH Oxidase 1 , NADPH Oxidases/genetics , NADPH Oxidases/metabolism , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , Reactive Oxygen Species/metabolism , Skin Neoplasms/genetics , Skin Neoplasms/metabolism , Skin Neoplasms/pathology , Transplantation, Heterologous
17.
Nucleic Acids Res ; 38(3): 797-809, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19934262

ABSTRACT

The regulation of DNA repair enzymes is crucial for cancer prevention, initiation, and therapy. We have studied the effect of ultraviolet B (UVB) radiation on the expression of the two nucleotide excision repair factors (XPC and XPD) in human keratinocytes. We show that hypoxia-inducible factor-1alpha (HIF-1alpha) is involved in the regulation of XPC and XPD. Early UVB-induced downregulation of HIF-1alpha increased XPC mRNA expression due to competition between HIF-1alpha and Sp1 for their overlapping binding sites. Late UVB-induced enhanced phosphorylation of HIF-1alpha protein upregulated XPC mRNA expression by direct binding to a separate hypoxia response element (HRE) in the XPC promoter region. HIF-1alpha also regulated XPD expression by binding to a region of seven overlapping HREs in its promoter. Quantitative chromatin immunoprecipitation assays further revealed putative HREs in the genes encoding other DNA repair proteins (XPB, XPG, CSA and CSB), suggesting that HIF-1alpha is a key regulator of the DNA repair machinery. Analysis of the repair kinetics of 6-4 photoproducts and cyclobutane pyrimidine dimers also revealed that HIF-1alpha downregulation led to an increased rate of immediate removal of both photolesions but attenuated their late removal following UVB irradiation, indicating the functional effects of HIF-1alpha in the repair of UVB-induced DNA damage.


Subject(s)
DNA Repair , DNA-Binding Proteins/genetics , Gene Expression Regulation , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Keratinocytes/metabolism , Xeroderma Pigmentosum Group D Protein/metabolism , Binding, Competitive , Cells, Cultured , DNA Damage , DNA-Binding Proteins/metabolism , Down-Regulation , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/antagonists & inhibitors , Keratinocytes/radiation effects , Promoter Regions, Genetic , Response Elements , Sp1 Transcription Factor/metabolism , Ultraviolet Rays
18.
PLoS One ; 4(5): e5493, 2009.
Article in English | MEDLINE | ID: mdl-19430526

ABSTRACT

BACKGROUND: The amount of inosine monophosphate dehydrogenase (IMPDH), a pivotal enzyme for the biosynthesis of the guanosine tri-phosphate (GTP), is frequently increased in tumor cells. The anti-viral agent ribavirin and the immunosuppressant mycophenolic acid (MPA) are potent inhibitors of IMPDH. We recently showed that IMPDH inhibition led to a necrotic signal requiring the activation of Cdc42. METHODOLOGY/PRINCIPAL FINDINGS: Herein, we strengthened the essential role played by this small GTPase in the necrotic signal by silencing Cdc42 and by the ectopic expression of a constitutive active mutant of Cdc42. Since resistance to apoptosis is an essential step for the tumorigenesis process, we next examined the effect of the MPA-mediated necrotic signal on different tumor cells demonstrating various mechanisms of resistance to apoptosis (Bcl2-, HSP70-, Lyn-, BCR-ABL-overexpressing cells). All tested cells remained sensitive to MPA-mediated necrotic signal. Furthermore, inhibition of IMPDH activity in Chronic Lymphocytic Leukemia cells was significantly more efficient at eliminating malignant cells than apoptotic inducers. CONCLUSIONS/SIGNIFICANCE: These findings indicate that necrosis and apoptosis are split signals that share few if any common hub of signaling. In addition, the necrotic signaling pathway induced by depletion of the cellular amount of GTP/GDP would be of great interest to eliminate apoptotic-resistant tumor cells.


Subject(s)
Apoptosis/drug effects , Drug Resistance, Neoplasm , Mycophenolic Acid/pharmacology , Signal Transduction/drug effects , Blotting, Western , Cell Line , Cell Line, Tumor , Dose-Response Relationship, Drug , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Guanosine Diphosphate/metabolism , Guanosine Triphosphate/metabolism , Humans , IMP Dehydrogenase/antagonists & inhibitors , IMP Dehydrogenase/metabolism , Jurkat Cells , K562 Cells , Leukemia, Lymphocytic, Chronic, B-Cell/blood , Leukemia, Lymphocytic, Chronic, B-Cell/pathology , Lymphocytes/metabolism , Lymphocytes/pathology , Lymphocytes/ultrastructure , Microscopy, Electron , Mutation , Necrosis/chemically induced , RNA, Small Interfering/genetics , Transfection , Tumor Cells, Cultured , cdc42 GTP-Binding Protein/genetics , cdc42 GTP-Binding Protein/metabolism
19.
J Immunol ; 181(11): 7630-8, 2008 Dec 01.
Article in English | MEDLINE | ID: mdl-19017951

ABSTRACT

Mycophenolate mofetil (MMF) is an immunosuppressive agent used in transplantation. Over the last decade, MMF has also emerged as an alternative therapeutic regimen for autoimmune diseases, mainly for patients refractory to other therapies. The active compound of MMF, mycophenolic acid (MPA), depletes the intracellular pool of guanosine tri-phosphate through inosine monophosphate dehydrogenase blockade. The molecular mechanism involved in the elimination of T and B lymphocytes upon inhibition of inosine monophosphate dehydrogenase remains elusive. In this study, we showed that in contrast to the immunosuppressors azathioprine, cyclosporin A, and tacrolimus, MPA killed lymphocytes through the activation of a caspase-independent necrotic signal. Furthermore, the MPA-mediated necrotic signal relied on the transmission of a novel intracellular signal involving Rho-GTPase Cdc42 activity and actin polymerization. In addition to its medical interest, this study sheds light on a novel and atypical molecular mechanism leading to necrotic cell death.


Subject(s)
Actins/immunology , B-Lymphocytes/immunology , Immunosuppressive Agents/pharmacology , Lymphocyte Activation/drug effects , Mycophenolic Acid/pharmacology , Signal Transduction/drug effects , T-Lymphocytes/immunology , Actins/metabolism , Adenosine Triphosphatases/immunology , Adenosine Triphosphatases/metabolism , B-Lymphocytes/enzymology , Enzyme Inhibitors/pharmacology , Enzyme Inhibitors/therapeutic use , Guanosine Triphosphate/immunology , Guanosine Triphosphate/metabolism , Humans , IMP Dehydrogenase/immunology , IMP Dehydrogenase/metabolism , Immunosuppressive Agents/therapeutic use , Jurkat Cells , Lymphocyte Activation/immunology , Mycophenolic Acid/therapeutic use , Necrosis/chemically induced , Necrosis/immunology , Organ Transplantation , Signal Transduction/immunology , T-Lymphocytes/enzymology , cdc42 GTP-Binding Protein/immunology , cdc42 GTP-Binding Protein/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...