Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 30
1.
Int J Mol Sci ; 24(24)2023 Dec 12.
Article En | MEDLINE | ID: mdl-38139209

The endocannabinoid system regulates physiological processes, and the modulation of endogenous endocannabinoid (eCB) levels is an attractive tool to contrast the development of pathological skin conditions including cancers. Inhibiting FAAH (fatty acid amide hydrolase), the degradation enzyme of the endocannabinoid anandamide (AEA) leads to the increase in AEA levels, thus enhancing its biological effects. Here, we evaluated the anticancer property of the FAAH inhibitor URB597, investigating its potential to counteract epithelial-to-mesenchymal transition (EMT), a process crucially involved in tumor progression. The effects of the compound were determined in primary human keratinocytes, ex vivo skin explants, and the squamous carcinoma cell line A431. Our results demonstrate that URB597 is able to hinder the EMT process by downregulating mesenchymal markers and reducing migratory potential. These effects are associated with the dampening of the AKT/STAT3 signal pathways and reduced release of pro-inflammatory cytokines and tumorigenic lipid species. The ability of URB597 to contrast the EMT process provides insight into effective approaches that may also include the use of FAAH inhibitors for the treatment of skin cancers.


Endocannabinoids , Neoplasms , Humans , Endocannabinoids/pharmacology , Endocannabinoids/metabolism , Polyunsaturated Alkamides/pharmacology , Polyunsaturated Alkamides/metabolism , Amidohydrolases/metabolism , Keratinocytes/metabolism
2.
Cells ; 12(7)2023 03 24.
Article En | MEDLINE | ID: mdl-37048080

Cutaneous squamous cell carcinoma (cSCC) is the most common UV-induced keratinocyte-derived cancer, and its progression is characterized by the epithelial-mesenchymal transition (EMT) process. We previously demonstrated that PPARγ activation by 2,4,6-octatrienoic acid (Octa) prevents cutaneous UV damage. We investigated the possible role of the PPARγ activators Octa and the new compound (2Z,4E,6E)-2-methoxyocta-2,4,6-trienoic acid (A02) in targeting keratinocyte-derived skin cancer. Like Octa, A02 exerted a protective effect against UVB-induced oxidative stress and DNA damage in NHKs. In the squamous cell carcinoma A431 cells, A02 inhibited cell proliferation and increased differentiation markers' expression. Moreover, Octa and even more A02 counteracted the TGF-ß1-dependent increase in mesenchymal markers, intracellular ROS, the activation of EMT-related signal transduction pathways, and cells' migratory capacity. Both compounds, especially A02, counterbalanced the TGF-ß1-induced cell membrane lipid remodeling and the release of bioactive lipids involved in EMT. In vivo experiments on a murine model useful to study cell proliferation in adult animals showed the reduction of areas characterized by active cell proliferation in response to A02 topical treatment. In conclusion, targeting PPARγ may be useful for the prevention and treatment of keratinocyte-derived skin cancer.


Carcinoma, Squamous Cell , Skin Neoplasms , Mice , Animals , Carcinoma, Squamous Cell/pathology , Epithelial-Mesenchymal Transition , Transforming Growth Factor beta1/pharmacology , PPAR gamma/metabolism , Skin Neoplasms/drug therapy , Skin Neoplasms/metabolism , Carcinogenesis
3.
Int J Mol Sci ; 25(1)2023 Dec 23.
Article En | MEDLINE | ID: mdl-38203435

Glucocorticoids (GCs) are commonly used in the treatment of inflammatory skin diseases, although the balance between therapeutic benefits and side effects is still crucial in clinical practice. One of the major and well-known adverse effects of topical GCs is cutaneous atrophy, which seems to be related to the activation of the glucorticoid receptor (GR) genomic pathway. Dissociating anti-inflammatory activity from atrophogenicity represents an important goal to achieve, in order to avoid side effects on keratinocytes and fibroblasts, known target cells of GC action. To this end, we evaluated the biological activity and safety profile of two novel chemical compounds, DE.303 and KL.202, developed as non-transcriptionally acting GR ligands. In primary keratinocytes, both compounds demonstrated anti-inflammatory properties inhibiting NF-κB activity, downregulating inflammatory cytokine release and interfering with pivotal signaling pathways involved in the inflammatory process. Of note, these beneficial actions were not associated with GC-related atrophic effects: treatments of primary keratinocytes and fibroblasts with DE.303 and KL.202 did not induce, contrarily to dexamethasone-a known potent GC-alterations in extracellular matrix components and lipid synthesis, thus confirming their safety profile. These data provide the basis for evaluating these compounds as effective alternatives to the currently used GCs in managing inflammatory skin diseases.


Dermatitis , Receptors, Glucocorticoid , Humans , Skin , Anti-Inflammatory Agents/adverse effects , Keratinocytes , Glucocorticoids/adverse effects , Dermatitis/drug therapy , Dermatitis/etiology , Atrophy
4.
Sci Adv ; 8(35): eabn9299, 2022 Sep 02.
Article En | MEDLINE | ID: mdl-36054352

Vitiligo is an acquired skin depigmentation disease involving multiple pathogenetic mechanisms, which ultimately direct cytotoxic CD8+ cells to destroy melanocytes. Abnormalities have been described in several cells even in pigmented skin as an expression of a functional inherited defect. Keratinocytes regulate skin homeostasis by the assembly of a proper skin barrier and releasing and responding to cytokines and growth factors. Alterations in epidermal proliferation, differentiation, and lipid composition as triggers for immune response activation in vitiligo have not yet been investigated. By applying cellular and lipidomic approaches, we revealed a deregulated keratinocyte differentiation with altered lipid composition, associated with impaired energy metabolism and increased glycolytic enzyme expression. Vitiligo keratinocytes secreted inflammatory mediators, which further increased following mild mechanical stress, thus evidencing immune activation. These findings identify intrinsic alterations of the nonlesional epidermis, which can be the prime instigator of the local inflammatory milieu that stimulates immune responses targeting melanocytes.

5.
iScience ; 25(3): 103871, 2022 Mar 18.
Article En | MEDLINE | ID: mdl-35252805

Melasma is a hyperpigmentary disorder with photoaging features, whose manifestations appear on specific face areas, rich in sebaceous glands (SGs). To explore the SGs possible contribution to the onset, the expression of pro-melanogenic and inflammatory factors from the SZ95 SG cell line exposed to single or repetitive ultraviolet (UVA) radiation was evaluated. UVA up-modulated the long-lasting production of α-MSH, EDN1, b-FGF, SCF, inflammatory cytokines and mediators. Irradiated SZ95 sebocyte conditioned media increased pigmentation in melanocytes and the expression of senescence markers, pro-inflammatory cytokines, and growth factors regulating melanogenesis in fibroblasts cultures. Cocultures experiments with skin explants confirmed the role of sebocytes on melanogenesis promotion. The analysis on sebum collected from melasma patients demonstrated that in vivo sebocytes from lesional areas express the UVA-activated pathways markers observed in vitro. Our results indicate sebocytes as one of the actors in melasma pathogenesis, inducing prolonged skin cell stimulation, contributing to localized dermal aging and hyperpigmentation.

6.
Int J Mol Sci ; 23(3)2022 Jan 21.
Article En | MEDLINE | ID: mdl-35163077

Apoptotic cells stimulate compensatory proliferation through the caspase-3-cPLA-2-COX-2-PGE-2-STAT3 Phoenix Rising pathway as a healing process in normal tissues. Phoenix Rising is however usurped in cancer, potentially nullifying pro-apoptotic therapies. Cytotoxic therapies also promote cancer cell plasticity through epigenetic reprogramming, leading to epithelial-to-mesenchymal-transition (EMT), chemo-resistance and tumor progression. We explored the relationship between such scenarios, setting-up an innovative, straightforward one-pot in vitro model of therapy-induced prostate cancer repopulation. Cancer (castration-resistant PC3 and androgen-sensitive LNCaP), or normal (RWPE-1) prostate cells, are treated with etoposide and left recovering for 18 days. After a robust apoptotic phase, PC3 setup a coordinate tissue-like response, repopulating and acquiring EMT and chemo-resistance; repopulation occurs via Phoenix Rising, being dependent on high PGE-2 levels achieved through caspase-3-promoted signaling; epigenetic inhibitors interrupt Phoenix Rising after PGE-2, preventing repopulation. Instead, RWPE-1 repopulate via Phoenix Rising without reprogramming, EMT or chemo-resistance, indicating that only cancer cells require reprogramming to complete Phoenix Rising. Intriguingly, LNCaP stop Phoenix-Rising after PGE-2, failing repopulating, suggesting that the propensity to engage/complete Phoenix Rising may influence the outcome of pro-apoptotic therapies. Concluding, we established a reliable system where to study prostate cancer repopulation, showing that epigenetic reprogramming assists Phoenix Rising to promote post-therapy cancer repopulation and acquired cell-resistance (CRAC).


Apoptosis , Cellular Reprogramming , Drug Resistance, Neoplasm , Epigenesis, Genetic , Etoposide/pharmacology , Gene Expression Regulation, Neoplastic/drug effects , Prostatic Neoplasms/pathology , Antineoplastic Agents, Phytogenic/pharmacology , Epithelial-Mesenchymal Transition , Humans , Male , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/genetics , Prostatic Neoplasms/metabolism , Receptors, Androgen/genetics , Receptors, Androgen/metabolism , Signal Transduction , Tumor Cells, Cultured
7.
Metabolites ; 11(12)2021 Nov 29.
Article En | MEDLINE | ID: mdl-34940576

Lipidomics is strategic in the discovery of biomarkers of neurodegenerative diseases (NDDs). The skin surface lipidome bears the potential to provide biomarker candidates in the detection of pathological processes occurring in distal organs. We investigated the sebum composition to search diagnostic and, possibly, prognostic, biomarkers of Alzheimer's disease (AD) and Parkinson's disease (PD). The observational study included 64 subjects: 20 characterized as "probable AD with documented decline", 20 as "clinically established PD", and 24 healthy subjects (HS) of comparable age. The analysis of sebum by GCMS and TLC retrieved the amounts (µg) of 41 free fatty acids (FFAs), 7 fatty alcohols (FOHs), vitamin E, cholesterol, squalene, and total triglycerides (TGs) and wax esters (WEs). Distributions of sebum lipids in NDDs and healthy conditions were investigated with multivariate ANOVA-simultaneous component analysis (ASCA). The deranged sebum composition associated with the PD group showed incretion of most composing lipids compared to HS, whereas only two lipid species (vitamin E and FOH14:0) were discriminant of AD samples and presented lower levels than HS sebum. Thus, sebum lipid biosynthetic pathways are differently affected in PD and AD. The characteristic sebum bio-signatures detected support the value of sebum lipidomics in the biomarkers search in NDDs.

8.
Pigment Cell Melanoma Res ; 34(1): 72-88, 2021 01.
Article En | MEDLINE | ID: mdl-32608114

The melanocortin-1 receptor (MC1R) belongs to the family of the G protein-coupled receptor (GPCR). Activated GPCRs can promote the phosphoinositide 3-kinase (PI3K) pathway. Few studies deal with the role of the PI3K pathway activation in response to αMSH. On B16-F10 cell line, we investigated the αMSH-dependent modulation of pAKT/AKT, as a key element of the PI3K pathway after rapid and prolonged stimulation. We demonstrated that αMSH triggers a rapid modulation of AKT which culminates in an increase in its phosphorylation. We highlighted a comparable upregulation of pAKT after exposure to αMSH on primary cultures of normal human melanocytes (NHMs) expressing a wild-type MC1R. On B16-F10 cells, NHMs, and an ex vivo model of human skin biopsies, we explored the influence of PI3K/AKT signaling triggered by αMSH, focusing on the control of melanogenesis and pigment release. We showed that the αMSH-dependent PI3K/AKT pathway exerts a negative feedback on melanogenesis and promotes the extracellular release of pigment. We strengthened the role of the PI3K/AKT pathway triggered by αMSH in preserving redox equilibrium and genome integrity, highlighting its role in affecting cell survival.


Feedback, Physiological , Melanins/metabolism , Melanocytes/cytology , Melanoma, Experimental/pathology , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , alpha-MSH/pharmacology , Animals , Humans , Melanocytes/drug effects , Melanocytes/metabolism , Melanoma, Experimental/metabolism , Mice , Phosphatidylinositol 3-Kinases/genetics , Phosphorylation , Pigmentation , Proto-Oncogene Proteins c-akt/genetics
9.
Exp Dermatol ; 29(9): 833-839, 2020 09.
Article En | MEDLINE | ID: mdl-32779245

Acne is the most common skin disease in adolescent Westernized populations. Several data support the involvement of the mammalian target of rapamycin complex 1 (mTORC1) signalling in the interplay between androgens, insulin, insulin-like growth factor (IGF1) and high-glycaemic index diet in acne. The peroxisome proliferator-activated receptor γ (PPARγ) is involved in both differentiation and anti-inflammatory response. Low differentiated sebocytes showed decreased expression of PPARγ and increased level of insulin and IGF-1 receptors, resulting in the production of acne-like sebum and inflammatory mediators. In this viewpoint, we discuss how in acne the dysregulation of proliferation and differentiation processes in sebocytes and keratinocytes may be associated with altered response to androgens and other hormones, such as insulin or IGF-1. Moreover, we propose PPARγ modulation as an innovative therapeutic approach to normalize sebocyte differentiation process, interfering with the different mechanisms involved in altered pilosebaceous unit.


Acne Vulgaris/etiology , Cell Differentiation , Hormones/metabolism , Acne Vulgaris/metabolism , Humans , Insulin-Like Growth Factor I/metabolism , Keratinocytes/metabolism , PPAR gamma/metabolism
10.
Front Pharmacol ; 9: 1183, 2018.
Article En | MEDLINE | ID: mdl-30459604

Cerium oxide nanoparticles (CNPs) are potent radical scavengers protecting cells from oxidative insults, including ionizing radiation. Here we show that CNPs prevent X-ray-induced oxidative imbalance reducing DNA breaks on HaCat keratinocytes, nearly abating mutagenesis. At the same time, and in spite of the reduced damage, CNPs strengthen radiation-induced cell cycle arrest and apoptosis outcome, dropping colony formation; notably, CNPs do not possess any intrinsic toxicity toward non-irradiated HaCat, indicating that they act on damaged cells. Thus CNPs, while exerting their antioxidant action, also reinforce the stringency of damage-induced cell integrity checkpoints, promoting elimination of the "tolerant" cells, being in fact radio-sensitizers. These two contrasting pathways are mediated by different activities of CNPs: indeed Sm-doped CNPs, which lack the Ce3+/Ce4+ redox switch and the correlated antioxidant action, fail to decrease radiation-induced superoxide formation, as expected, but surprisingly maintain the radio-sensitizing ability and the dramatic decrease of mutagenesis. The latter is thus attributable to elimination of damaged cells rather than decreased oxidative damage. This highlights a novel redox-independent activity of CNPs, allowing selectively eliminating heavily damaged cells through non-toxic mechanisms, rather reactivating endogenous anticancer pathways in transformed cells.

11.
Free Radic Biol Med ; 115: 266-277, 2018 02 01.
Article En | MEDLINE | ID: mdl-29229551

Pleural mesothelioma is a deadly form of cancer. The prognosis is extremely poor due to the limited treatment modalities. Uptake of asbestos fibres, the leading cause of mesothelioma, lead to the accumulation of reactive-oxygen-species (ROS). Interestingly, increasing ROS production by using ROS-generating drugs may offer a strategy to selectively trigger cell death. Exemestane, an aromatase inhibitor, has previously shown anti-tumor properties in mesothelioma preclinical models suggesting a role of G protein-coupled receptor 30 (GPR30) in the drug response. As exemestane, in addition to blocking estrogen biosynthesis, generates ROS that are able to arrest the growth of breast cancer, we explored the role of ROS, antioxidant defense system, and ROS-induced signalling pathways in mesothelioma cells during exemestane response. Here we report that exemestane treatment reduced cell proliferation with an increase in ROS production and reduction of cyclic adenosine monophosphate (cAMP) levels in MSTO-H211, Ist-Mes1, Ist-Mes2 and MPP89 exemestane-sensitive mesothelioma cell lines, but not in NCI-H2452 exemestane-insensitive mesothelioma cells. Exemestane induced a significant antioxidant response in NCI-H2452 cells, as highlighted by an increase in γ-glutamylcysteine levels, catalase (Cat), superoxide-dismutase and (SOD) and glutathione-peroxidase (GSH-Px) activity and nuclear factor E2-related factor 2 (Nrf2) activation, responsible for drug insensitivity. Conversely, exemestane elevated ROS levels along with increased ERK phosphorylation and a reduction of p-STA3 in exemestane-sensitive mesothelioma cells. ROS generation was the crucial event of exemestane action because ROS inhibitor N-acetyl-L-cysteine (NAC) abrogated p-ERK and p-STAT3 modulation and cellular death. Exemestane also modulates ERK and STAT3 signalling via GPR30. Results indicate an essential role of ROS in the antiproliferative action of exemestane in mesothelioma cells. It is likely that the additional oxidative insults induced by exemestane results in the lethal effects of mesothelioma cells by increasing ROS production. As such, manipulating ROS levels with exemestane seems to be a feasible strategy to selectively kill mesothelioma cells with less toxicity to normal cells by regulating ERK and STAT3 activity.


Androstadienes/pharmacology , Antineoplastic Agents/pharmacology , Aromatase Inhibitors/pharmacology , Mesothelioma/drug therapy , Pleural Neoplasms/drug therapy , Acetylcysteine/pharmacology , Asbestos/adverse effects , Cell Death , Cell Line, Tumor , Cell Proliferation/drug effects , Cyclic AMP/metabolism , Environmental Exposure/adverse effects , Extracellular Signal-Regulated MAP Kinases/metabolism , Humans , Oxidative Stress , Reactive Oxygen Species/metabolism , Receptors, Estrogen , Receptors, G-Protein-Coupled , STAT3 Transcription Factor/metabolism , Signal Transduction , Treatment Outcome
12.
Sci Rep ; 7(1): 9241, 2017 08 23.
Article En | MEDLINE | ID: mdl-28835664

Increasing attention is addressed to identify products able to enhance skin photoprotection and to prevent skin carcinogenesis. Several studies have demonstrated that the α-melanocyte stimulating hormone (αMSH), acting on a functional MC1R, provides a photoprotective effect by inducing pigmentation, antioxidants and DNA repair. We discovered a link between αMSH and the nuclear receptor Peroxisome Proliferator-Activated Receptor-γ (PPARγ), suggesting that some of the αMSH protective effects may be dependent on PPARγ transcriptional activity. Moreover, we demonstrated that the activation of PPARγ by the parrodiene 2,4,6-octatrienoic acid (Octa) induces melanogenesis and antioxidant defence in human melanocytes and counteracts senescence-like phenotype in human fibroblasts. In this study, we demonstrate that the activation of PPARγ by Octa exerts a protective effect against UVA- and UVB-induced damage on normal human keratinocytes (NHKs), the major target cells of UV radiation. Octa promotes the antioxidant defence, augments DNA repair and reduces the induction of proteins involved in UV-induced DNA damage response. Our results contribute to deepen the analysis of the αMSH/PPARγ connection and suggest perspectives for the development of new molecules and formulations able to prevent cutaneous UV damage by acting on the different skin cell populations through PPARγ activation.


Fatty Acids, Unsaturated/pharmacology , Keratinocytes/drug effects , Keratinocytes/enzymology , Keratinocytes/radiation effects , PPAR gamma/agonists , Radiation-Protective Agents/pharmacology , Ultraviolet Rays/adverse effects , Antioxidants/metabolism , Cells, Cultured , DNA Damage/drug effects , DNA Damage/radiation effects , Enzyme Activation/drug effects , Gene Expression Regulation/drug effects , Humans , Keratinocytes/pathology , PPAR gamma/genetics , PPAR gamma/metabolism , Receptor, Melanocortin, Type 1/genetics , Receptor, Melanocortin, Type 1/metabolism , alpha-MSH/metabolism
14.
PLoS One ; 9(8): e104045, 2014.
Article En | MEDLINE | ID: mdl-25101957

Peroxisome proliferator-activated receptor gamma (PPARγ) may be involved in a key mechanism of the skin aging process, influencing several aspects related to the age-related degeneration of skin cells, including antioxidant unbalance. Therefore, we investigated whether the up-modulation of this nuclear receptor exerts a protective effect in a stress-induced premature senescence (SIPS) model based on a single exposure of human dermal fibroblasts to 8-methoxypsoralen plus + ultraviolet-A-irradiation (PUVA). Among possible PPARγ modulators, we selected 2,4,6-octatrienoic acid (Octa), a member of the parrodiene family, previously reported to promote melanogenesis and antioxidant defense in normal human melanocytes through a mechanism involving PPARγ activation. Exposure to PUVA induced an early and significant decrease in PPARγ expression and activity. PPARγ up-modulation counteracted the antioxidant imbalance induced by PUVA and reduced the expression of stress response genes with a synergistic increase of different components of the cell antioxidant network, such as catalase and reduced glutathione. PUVA-treated fibroblasts grown in the presence of Octa are partially but significantly rescued from the features of the cellular senescence-like phenotype, such as cytoplasmic enlargement, the expression of senescence-associated-ß-galactosidase, matrix-metalloproteinase-1, and cell cycle proteins. Moreover, the alterations in the cell membrane lipids, such as the decrease in the polyunsaturated fatty acid content of phospholipids and the increase in cholesterol levels, which are typical features of cell aging, were prevented. Our data suggest that PPARγ is one of the targets of PUVA-SIPS and that its pharmacological up-modulation may represent a novel therapeutic approach for the photooxidative skin damage.


Cellular Senescence , Fibroblasts/drug effects , PPAR gamma/physiology , Stress, Physiological , Cell Membrane/chemistry , Cell Membrane/metabolism , Cholesterol/metabolism , Fatty Acids, Unsaturated/metabolism , Fatty Acids, Unsaturated/pharmacology , Fibroblasts/radiation effects , Humans , Lipid Peroxidation , Methoxsalen/pharmacology , PPAR gamma/genetics , PPAR gamma/metabolism , Phospholipids/metabolism , Protective Agents/pharmacology , Reactive Oxygen Species/metabolism , Ultraviolet Rays , Up-Regulation
15.
Pigment Cell Melanoma Res ; 26(5): 616-33, 2013 Sep.
Article En | MEDLINE | ID: mdl-23710556

Despite considerable advances in the past decade, melanin research still suffers from the lack of universally accepted and shared nomenclature, methodologies, and structural models. This paper stems from the joint efforts of chemists, biochemists, physicists, biologists, and physicians with recognized and consolidated expertise in the field of melanins and melanogenesis, who critically reviewed and experimentally revisited methods, standards, and protocols to provide for the first time a consensus set of recommended procedures to be adopted and shared by researchers involved in pigment cell research. The aim of the paper was to define an unprecedented frame of reference built on cutting-edge knowledge and state-of-the-art methodology, to enable reliable comparison of results among laboratories and new progress in the field based on standardized methods and shared information.


Chemistry Techniques, Analytical/methods , Chemistry Techniques, Analytical/standards , Melanins/biosynthesis , Animals , Biophysical Phenomena , Humans , Melanins/chemistry , Melanins/classification , Spectrum Analysis
16.
Exp Dermatol ; 22(1): 41-7, 2013 Jan.
Article En | MEDLINE | ID: mdl-23278893

Azelaic acid (AzA) has been used for the treatment for inflammatory skin diseases, such as acne and rosacea. Interestingly, an improvement in skin texture has been observed after long-time treatment with AzA. We previously unrevealed that anti-inflammatory activity of AzA involves a specific activation of PPARγ, a nuclear receptor that plays a relevant role in inflammation and even in ageing processes. As rosacea has been considered as a photo-aggravated disease, we investigated the ability of AzA to counteract stress-induced premature cell senescence (SIPS). We employed a SIPS model based on single exposure of human dermal fibroblasts (HDFs) to UVA and 8-methoxypsoralen (PUVA), previously reported to activate a senescence-like phenotype, including long-term growth arrest, flattened morphology and increased synthesis of matrix metalloproteinases (MMPs) and senescence-associated ß-galactosidase (SA-ß-gal). We found that PUVA-treated HDFs grown in the presence of AzA maintained their morphology and reduced MMP-1 release and SA-ß-galactosidase-positive cells. Moreover, AzA induced a reduction in ROS generation, an up-modulation of antioxidant enzymes and a decrease in cell membrane lipid damages in PUVA-treated HDFs. Further evidences of AzA anti-senescence effect were repression of p53 and p21, increase in type I pro-collagen and abrogation of the enhanced expression of growth factors, such as HGF and SCF. Interestingly, PUVA-SIPS showed a decreased activation of PPARγ and AzA counteracted this effect, suggesting that AzA effect involves PPARγ modulation. All together these data showed that AzA interferes with PUVA-induced senescence-like phenotype and its ability to activate PPAR-γ provides relevant insights into the anti-senescence mechanism.


Cellular Senescence/drug effects , Dermatologic Agents/pharmacology , Dicarboxylic Acids/pharmacology , Fibroblasts/drug effects , Fibroblasts/metabolism , PPAR gamma/metabolism , Cell Membrane/drug effects , Cell Membrane/metabolism , Cells, Cultured , Cellular Senescence/radiation effects , Collagen Type I/metabolism , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Fibroblasts/cytology , Hepatocyte Growth Factor/metabolism , Humans , Matrix Metalloproteinase 1/metabolism , Methoxsalen/pharmacology , PUVA Therapy , Phenotype , Phospholipids/metabolism , Photosensitizing Agents/pharmacology , Procollagen/metabolism , Reactive Oxygen Species/metabolism , Stem Cell Factor/metabolism , Tumor Suppressor Protein p53/metabolism , Ultraviolet Rays , beta-Galactosidase/metabolism
17.
FASEB J ; 26(9): 3779-89, 2012 Sep.
Article En | MEDLINE | ID: mdl-22649030

Cystinosis is a rare autosomal recessive disease characterized by cystine crystal accumulation leading to multiorgan dysfunctions and caused by mutation in CTNS. CTNS encodes cystinosin, a cystine/H(+) symporter that exports cystine out of the lysosomes. Patients with cystinosis frequently exhibit blond hair and fair complexion, suggesting an alteration in melanogenesis. However, the pigmentation singularities of these patients have not been studied, and the role of cystinosin in melanogenesis has remained unknown. In our study, a clinical evaluation of 27 patients with cystinosis showed that 44% had a cutaneous pigmentation dilution compared to their relatives. Analysis of the hair melanin content in these patients by HPLC demonstrated a 50% decrease in eumelanin (4360 vs. 9360 ng/mg), and a 2-fold increase in pheomelanin (53 vs. 20 ng/mg), the yellow/red pigments. Cystinosin-deficient mice also showed a 4-fold increase in hair pheomelanin content. In vitro studies showed that cystinosin was located at melanosomes. CTNS silencing led to a 75% reduction of melanin synthesis that was caused by a degradation of tyrosinase by lysosomal proteases. Our results objectify the pigmentation defect in patients with cystinosis. We also identify the role of CTNS in melanogenesis and add a new gene to the list of the genes involved in the control of skin and hair pigmentation.


Amino Acid Transport Systems, Neutral/physiology , Melanins/biosynthesis , Melanosomes/metabolism , Adolescent , Adult , Amino Acid Transport Systems, Neutral/genetics , Amino Acid Transport Systems, Neutral/metabolism , Animals , Cell Line, Tumor , Child , Child, Preschool , Cystinosis/metabolism , Female , Humans , Male , Mice , Mice, Knockout , Mutation , Reverse Transcriptase Polymerase Chain Reaction , Skin Pigmentation/genetics , Young Adult
18.
Pigment Cell Melanoma Res ; 24(4): 618-30, 2011 Aug.
Article En | MEDLINE | ID: mdl-21762468

Given the importance of the tanning response in protecting human skin from the harmful effects of UV radiation, one important research priority is to identify novel molecules that are capable of promoting pigmentation and/or antioxidant defence. Parrodienes share some structural features with carotenoids and retinoids, stimulate cell antioxidant defence and counteract senescence-like phenotype in fibroblasts. We selected the parrodiene-derivative 2,4,6-octatrienoic acid (Octa) to study its impact on key parameters of melanogenesis and antioxidant defence in organ-cultured human skin and in normal human melanocytes. Octa promoted melanogenesis by up-regulating tyrosinase and microphthalmia-associated transcription factor expression. This correlated with an increase of melanin content in both human epidermis in situ and cultured human epidermal melanocytes. Moreover, Octa increased the biological antioxidant potential content and the expression and activity of catalase. Activation of peroxisome proliferator-activated receptor (PPAR)-γ was necessary to evoke these effects. These data strongly encourage the systematic study of Octa as a novel candidate promoter of human skin photoprotection.


Antioxidants/metabolism , Fatty Acids, Unsaturated/pharmacology , Melanins/biosynthesis , Melanocytes/drug effects , Melanocytes/immunology , PPAR gamma/metabolism , Catalase/metabolism , Cell Proliferation/drug effects , Cell Survival/drug effects , Enzyme Induction/drug effects , Epidermis/drug effects , Epidermis/physiology , Gene Expression Regulation/drug effects , Gene Silencing/drug effects , Humans , Male , Melanocytes/cytology , Melanocytes/enzymology , Microphthalmia-Associated Transcription Factor/genetics , Microphthalmia-Associated Transcription Factor/metabolism , Monophenol Monooxygenase/biosynthesis , Monophenol Monooxygenase/genetics , Organ Culture Techniques , PPAR gamma/antagonists & inhibitors , PPAR gamma/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , Scalp/drug effects , Scalp/physiology , Skin Pigmentation/drug effects
19.
Melanoma Res ; 19(6): 372-8, 2009 Dec.
Article En | MEDLINE | ID: mdl-19741552

Malignant melanoma incidence is increasing rapidly in Western countries. Its prevention requires a deep knowledge of the biological basis of the neoplasm leading to the identification of new biological risk markers. In in-vitro and ex-vivo models we demonstrated that catalase was modified not only in its activity but also in its charge properties after ultraviolet A irradiation through pheomelanin. Here we focus on the electrophoretic behaviour of catalase in the human skin in vivo, in association with cutaneous phototype. Zymographic analysis of the enzyme on skin biopsies from Caucasian population (phototype I-IV), collected from the trunk in autumn-winter, to exclude possible influences of an acute photoexposure, evidenced a protein doublet, representing the coexistence of two active isoforms of catalase with different charge properties. In the skin from low-phototype subjects, the percent contribution of the more acidic component of the doublet was prevalent, inversely correlated with total melanin concentration in hair, and associated with a high number of melanocytic nevi. In summary, this study shows for the first time the existence of an acidic catalase in association with clinically defined risk characteristics in low phototype skin in vivo, contributing to the knowledge of a new biochemical marker of cutaneous photosusceptibility.


Catalase/metabolism , Hair/enzymology , Melanoma/enzymology , Melanoma/metabolism , Skin/enzymology , Adolescent , Adult , Aged , Biomarkers, Tumor , Female , Hair/metabolism , Humans , Hydrogen-Ion Concentration , Male , Melanins , Middle Aged , Nevus, Pigmented , Protein Isoforms , Skin/metabolism , Skin Neoplasms/enzymology , Skin Neoplasms/metabolism , Ultraviolet Rays , Young Adult
20.
J Dermatol Sci ; 54(2): 106-13, 2009 May.
Article En | MEDLINE | ID: mdl-19250802

BACKGROUND: Exposure to ultraviolet (UV) radiation causes a complex cellular response, mostly mediated by the production of reactive oxygen species (ROS), which can be counteracted by exogenous treatments and endogenous mechanisms with anti-oxidant and scavenger properties. Keratinocyte growth factor (KGF/FGF7), a member of the fibroblast growth factor family, promotes epithelial growth and differentiation and is involved in cell survival after oxidant injuries. OBJECTIVE: We analyzed the role of KGF in the control of intracellular ROS production and oxidative stress after UVB exposure on KGF receptor (KGFR) transfected cells and human immortalized and primary keratinocytes. METHODS: We assessed the intracellular ROS production measuring the intensity of the oxidation-sensitive fluorescent probe 2',7'-dichlorofluorescein diacetate (DCFH-DA) by confocal microscopy, as well as the catalase activity by spectrophotometric assay. Moreover, morphological and biochemical analysis of actin cytoskeleton reorganization was evaluated as a further marker of oxidative damage. RESULTS: Our data show that KGF significantly reduces intracellular ROS generation in response to UVB, preserves the decrease of catalase activity and prevents actin cytoskeleton rearrangement. CONCLUSION: Our results provide a further evidence that KGF may be crucial for an efficient skin photoprotection, demonstrating a direct role for KGF in the reduction of intracellular ROS content following UVB exposure.


Actins/metabolism , Catalase/metabolism , Fibroblast Growth Factor 7/physiology , Keratinocytes/metabolism , Reactive Oxygen Species/metabolism , Receptor, Fibroblast Growth Factor, Type 2/metabolism , Actins/drug effects , Actins/radiation effects , Animals , Catalase/drug effects , Catalase/radiation effects , Cell Line , Cytoskeleton/drug effects , Cytoskeleton/metabolism , Cytoskeleton/radiation effects , Down-Regulation/drug effects , Fibroblast Growth Factor 7/pharmacology , Humans , Keratinocytes/drug effects , Keratinocytes/radiation effects , Mice , NIH 3T3 Cells , Oxidative Stress/drug effects , Oxidative Stress/radiation effects , Receptor, Fibroblast Growth Factor, Type 2/agonists , Transfection , Ultraviolet Rays
...