Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 5 de 5
Filter
Add more filters










Database
Language
Publication year range
1.
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
2.
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
3.
J Invest Dermatol ; 131(9): 1793-805, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21633368

ABSTRACT

Besides lung, postnatal human epidermis is the only epithelium in direct contact with atmospheric oxygen. Skin epidermal oxygenation occurs mostly through atmospheric oxygen rather than tissue vasculature, resulting in a mildly hypoxic microenvironment that favors increased expression of hypoxia-inducible factor-1α (HIF-1α). Considering the wide spectrum of biological processes, such as angiogenesis, inflammation, bioenergetics, proliferation, motility, and apoptosis, that are regulated by this transcription factor, its high expression level in the epidermis might be important to HIF-1α in skin physiology and pathophysiology. Here, we review the role of HIF-1α in cutaneous angiogenesis, skin tumorigenesis, and several skin disorders.


Subject(s)
Carcinoma, Squamous Cell/physiopathology , Epidermis/physiology , Hypoxia-Inducible Factor 1, alpha Subunit/physiology , Neovascularization, Physiologic/physiology , Skin Neoplasms/physiopathology , Carcinoma, Squamous Cell/metabolism , Humans , Oxygen/metabolism , Skin Diseases/metabolism , Skin Diseases/physiopathology , Skin Neoplasms/metabolism
4.
Stem Cells ; 28(9): 1639-48, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20681019

ABSTRACT

Tissue stem cells must be endowed with superior maintenance and repair systems to ensure genomic stability over multiple generations, which would be less necessary in more differentiated cells. We previously reported that human keratinocyte stem cells were more resistant to ionizing radiation toxicity than their direct progeny, the keratinocyte progenitor cells. In the present study we addressed the mechanisms underlying this difference. Investigations of DNA repair showed that both single and double DNA strand breaks were repaired more rapidly and more efficiently in stem cells than in progenitors. As cell signaling is a key regulatory step in the management of DNA damage, a gene profiling study was performed. Data revealed that several genes of the fibroblast growth factor type 2 (FGF2) signaling pathway were induced by DNA damage in stem cells and not in progenitors. Furthermore, an increased content of the FGF2 protein was found in irradiated stem cells, both for the secreted and the cellular forms of the protein. To examine the role of endogenous FGF2 in DNA repair, stem cells were exposed to FGF2 pathway inhibitors. Blocking the FGF2 receptor (FGF receptor 1) or the kinase (Ras-mitogen-activated protein kinase 1) resulted in a inhibition of single and double DNA strand-break repair in the keratinocyte stem cells. Moreover, supplementing the progenitor cells with exogenous FGF2 activated their DNA repair. We propose that, apart from its well-known role as a strong mitogen and prosurvival factor, FGF2 helps to maintain genomic integrity in stem cells by activating stress-induced DNA repair.


Subject(s)
DNA Breaks, Double-Stranded , DNA Breaks, Single-Stranded , DNA Repair , Fibroblast Growth Factor 2/metabolism , Keratinocytes/metabolism , Signal Transduction , Stem Cells/metabolism , Antibodies, Monoclonal/pharmacology , Butadienes/pharmacology , Cell Cycle , Cells, Cultured , Chromatin Assembly and Disassembly , Comet Assay , Fibroblast Growth Factor 2/antagonists & inhibitors , Fibroblast Growth Factor 2/genetics , Gene Expression Profiling , Gene Regulatory Networks , Genomic Instability , Histones/metabolism , Humans , Keratinocytes/drug effects , Keratinocytes/radiation effects , Nitriles/pharmacology , Phosphorylation , Protein Kinase Inhibitors/pharmacology , Recombinant Proteins/metabolism , Serine , Signal Transduction/drug effects , Signal Transduction/genetics , Signal Transduction/radiation effects , Stem Cells/drug effects , Stem Cells/radiation effects , Time Factors
5.
Mutat Res ; 704(1-3): 167-74, 2010.
Article in English | MEDLINE | ID: mdl-20117235

ABSTRACT

Stem cells have been described in most adult tissues, where they play a key role in maintaining tissue homeostasis. As they self-renew throughout life, accumulating genetic anomalies can compromise their genomic integrity and potentially give rise to cancer. Stem cells (SCs) may thus be a major target of radiation carcinogenesis. In addition, unrepaired genotoxic damage may cause cell death and stem cell pool depletion, impairing lineage functionality and accelerating aging. Developments in SC biology enabled the characterization of the responses of stem cells to genotoxic stress and their role in tissue damage. We here examine how these cells react to ionizing radiation (IR), and more specifically their radiosensitivity, stress signaling and DNA repair. We first review embryonic SCs, as a paradigm of primitive pluripotent cells, then three adult tissues, bone marrow, skin and intestine, capable of long-term regeneration and at high risk for acute radiation syndromes and long-term carcinogenesis. We discuss IR disruption of the fine balance between maintenance of tissue homeostasis and genomic stability. We show that stem cell radiosensitivity does not follow a unique model, but differs notably according to the turnover rates of the tissues.


Subject(s)
Genomic Instability/radiation effects , Homeostasis , Radiation, Ionizing , Stem Cells/radiation effects , Bone Marrow Cells/radiation effects , DNA Damage , DNA Repair , Embryonic Stem Cells/radiation effects , Hematopoietic Stem Cells , Humans , Intestines/cytology , Radiation Tolerance , Signal Transduction , Skin/cytology , Time
SELECTION OF CITATIONS
SEARCH DETAIL
...