Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters










Database
Language
Publication year range
1.
J Biol Chem ; 285(19): 14842, 2010 May 07.
Article in English | MEDLINE | ID: mdl-29874310

ABSTRACT

[This retracts the article on p. 28897 in vol. 283, PMID: 18715874.].

2.
Int J Mol Med ; 23(6): 771-7, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19424603

ABSTRACT

Aquaporins (AQPs) are a family of 13 small ( approximately 30 kDa/monomer), hydrophobic, integral membrane proteins. AQPs are expressed in various epithelial and endothelial cells involved in fluid transport. Here, we demonstrated for the first time that AQP1 is expressed in cultured human retinal pigment epithelial (RPE) cells (ARPE-19 cell line). Ultraviolet radiation (UVB) and H2O2, two major factors causing RPE cell damage, induced AQP1 downregulation which was mediated by MEK/ERK activation. UV and H2O2 as well as AQP1-specific siRNA knockdown impaired water permeability of ARPE-19 cells. Notably, pretreatment with all-trans retinoic acid attenuated UV- and H2O2-induced AQP1 downregulation and water permeability impairment. Considering that water permeability is involved in multiple functions of RPE cells such as cellular junction formation, fluid or protein exchange and barrier formation, our data elucidated a novel mechanism through which UV radiation and oxidative stress induce eye cell damage. Our results further support the notion that all-trans retinoic acid might be useful for protection against UV or oxidative stress-induced eye cell damage.


Subject(s)
Aquaporin 1/metabolism , Cell Membrane Permeability/radiation effects , Down-Regulation/radiation effects , Mitogen-Activated Protein Kinase Kinases/metabolism , Pigment Epithelium of Eye/cytology , Ultraviolet Rays , Water/metabolism , Aquaporin 1/genetics , Blotting, Western , Cell Line , Cell Membrane Permeability/drug effects , Down-Regulation/drug effects , Down-Regulation/genetics , Humans , Hydrogen Peroxide/pharmacology , RNA, Small Interfering/genetics , RNA, Small Interfering/physiology , Reactive Oxygen Species/metabolism , Tretinoin/pharmacology
3.
J Cell Mol Med ; 13(9B): 3632-43, 2009 Sep.
Article in English | MEDLINE | ID: mdl-18681908

ABSTRACT

SIRT1 is a member of a highly conserved gene family (sirtuins) encoding nicotinamide adenine dinucleotide (NAD)(+)-dependent deacetylases, originally found to deacetylate histones leading to increased DNA stability and prolonged survival in yeast and higher organisms, including mammals. SIRT1 has been found to function as a deacetylase for numerous protein targets involved in various cellular pathways, including stress responses, apoptosis and axonal degeneration. However, the role of SIRT1 in ultraviolet (UV) signalling pathways remains unknown. Using cell culture and Western blot analysis in this study we found that SIRT1 is expressed in cultured human skin keratinocytes. Both UV radiation and H(2)O(2), two major inducers of skin cell damage, down-regulate SIRT1 in a time- and dose-dependent manner. We observed that reactive oxygen species-mediated JNK activation is involved in this SIRT1 down-regulation. SIRT1 activator, resveratrol, which has been considered as an important antioxidant, protects against UV- and H(2)O(2)-induced cell death, whereas SIRT inhibitors such as sirtinol and nicotinamide enhance cell death. Activation of SIRT1 negatively regulates UV- and H(2)O(2)-induced p53 acetylation, because nicotinamide and sirtinol as well as SIRT1 siRNA enhance UV- and H(2)O(2)-induced p53 acetylation, whereas SIRT1 activator resveratrol inhibits it. We also found that SIRT1 is involved in UV-induced AMP-activated protein kinase (AMPK) and downstream acetyl-CoA carboxylase (ACC), phosphofructose kinase-2 (PFK-2) phosphorylation. Collectively, our data provide new insights into understanding of the molecular mechanisms of UV-induced skin aging, suggesting that SIRT1 activators such as resveratrol could serve as new anti-skin aging agents.


Subject(s)
Cells, Cultured/metabolism , Gene Expression Regulation , Hydrogen Peroxide/pharmacology , MAP Kinase Kinase 4/metabolism , Sirtuin 1/metabolism , Skin/cytology , Tumor Suppressor Protein p53/metabolism , Animals , Cell Death , Cell Proliferation , Dose-Response Relationship, Drug , Humans , Mice , Reactive Oxygen Species
4.
Oncol Rep ; 20(6): 1553-9, 2008 Dec.
Article in English | MEDLINE | ID: mdl-19020741

ABSTRACT

AMP-activated protein kinase (AMPK), an evolutionarily conserved serine/threonine protein kinase, serves as an energy sensor in all eukaryotic cells. Recent findings suggest that AMPK activation strongly suppresses cell proliferation and induces cell apoptosis in a variety of cancer cells. Our study demonstrated that chemopreventive agent curcumin strongly activates AMPK in a p38-dependent manner in CaOV3 ovarian cancer cells. Pretreatment of cells with compound C (AMPK inhibitor) and SB203580 (p38 inhibitor) attenuates curcumin-induced cell death. We also observed that curcumin induces p53 phosphorylation (Ser 15) and both compound C and SB203580 pretreatment inhibit p53 phosphorylation. Collectively, our data suggest that AMPK is a new molecular target of curcumin and AMPK activation partially contributes to the cytotoxic effect of curcumin in ovarian cancer cells.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Curcumin/pharmacology , Gene Expression Regulation, Enzymologic , Gene Expression Regulation, Neoplastic , Ovarian Neoplasms/drug therapy , Ovarian Neoplasms/enzymology , Antineoplastic Agents/pharmacology , Cell Death , Cell Line, Tumor , Cell Survival , Enzyme Inhibitors/pharmacology , Female , Humans , Phosphorylation , Tetrazolium Salts/pharmacology , Thiazoles/pharmacology , p38 Mitogen-Activated Protein Kinases/metabolism
5.
J Biol Chem ; 283(43): 28897-908, 2008 Oct 24.
Article in English | MEDLINE | ID: mdl-18715874

ABSTRACT

AMP-activated protein kinase or AMPK is an evolutionarily conserved sensor of cellular energy status, activated by a variety of cellular stresses that deplete ATP. However, the possible involvement of AMPK in UV- and H(2)O(2)-induced oxidative stresses that lead to skin aging or skin cancer has not been fully studied. We demonstrated for the first time that UV and H(2)O(2) induce AMPK activation (Thr(172) phosphorylation) in cultured human skin keratinocytes. UV and H(2)O(2) also phosphorylate LKB1, an upstream signal of AMPK, in an epidermal growth factor receptor-dependent manner. Using compound C, a specific inhibitor of AMPK and AMPK-specific small interfering RNA knockdown as well as AMPK activator, we found that AMPK serves as a positive regulator for p38 and p53 (Ser(15)) phosphorylation induced by UV radiation and H(2)O(2) treatment. We also observed that AMPK serves as a negative feedback signal against UV-induced mTOR (mammalian target of rapamycin) activation in a TSC2-dependent manner. Inhibiting mTOR and positively regulating p53 and p38 might contribute to the pro-apoptotic effect of AMPK on UV- or H(2)O(2)-treated cells. Furthermore, activation of AMPK also phosphorylates acetyl-CoA carboxylase or ACC, the pivotal enzyme of fatty acid synthesis, and PFK2, the key protein of glycolysis in UV-radiated cells. Collectively, we conclude that AMPK contributes to UV- and H(2)O(2)-induced apoptosis via multiple mechanisms in human skin keratinocytes and AMPK plays important roles in UV-induced signal transduction ultimately leading to skin photoaging and even skin cancer.


Subject(s)
AMP-Activated Protein Kinases/chemistry , Apoptosis , Hydrogen Peroxide/chemistry , Keratinocytes/cytology , Skin/metabolism , Acetyl-CoA Carboxylase/metabolism , Adenylate Kinase/metabolism , Cell Line , Epidermal Growth Factor/metabolism , Humans , Keratinocytes/metabolism , Models, Biological , Skin Neoplasms/metabolism , Tumor Suppressor Protein p53/metabolism , Ultraviolet Rays , p38 Mitogen-Activated Protein Kinases/metabolism
6.
Int J Mol Med ; 22(2): 229-36, 2008 Aug.
Article in English | MEDLINE | ID: mdl-18636178

ABSTRACT

The most common adverse effects that are related to all-trans retinoic acid (atRA) treatment are irritation and dryness of the skin. atRA therapy is reported to impair barrier function as achieved by trans-epidermal water loss (TEWL). Treatment with nicotinamide prior to initiation of atRA therapy provides additional barrier protection and thus reduces susceptibility of retinoic acid. Our previous studies showed that atRA upregulates aquaporin 3 (AQP3) in cultured human skin keratinocytes and fibroblasts. Others have demonstrated that in atopic dermatitis, overexpression of AQP3 is linked to elevated TEWL and that nicotinamide treatment reduces skin TEWL. In this study, we observed that while atRA upregulates AQP3 expression in cultured human skin keratinocytes (HaCaT cells), nicotinamide attenuates the effect of atRA in a concentration-dependent manner. atRA treatment induces EGFR and ERK activation. PD153035, an EGFR inhibitor, and U0126, an ERK inhibitor, inhibit atRA-induced upregulation of AQP3. Nicotinamide also inhibits atRA-induced activation of EGFR/ERK signal transduction and decreases water permeability by downregulating AQP3 expression. Collectively, our results indicate that the effect of atRA on AQP3 expression is at least partly mediated by EGFR/ERK signaling in cultured human skin keratinocytes. Nicotinamide attenuates atRA-induced AQP3 expression through inhibition of EGFR/ERK signal transduction and eventually decreases water permeability and water loss. Our study provides insights into the molecular mechanism through which nicotinamide reverses the side effects of dryness in human skin after treatment with atRA.


Subject(s)
Aquaporin 3/metabolism , ErbB Receptors/metabolism , Extracellular Signal-Regulated MAP Kinases/metabolism , Keratinocytes/metabolism , Niacinamide/pharmacology , Tretinoin/pharmacology , Animals , Aquaporin 3/genetics , Cells, Cultured , Epidermal Growth Factor/metabolism , ErbB Receptors/antagonists & inhibitors , Extracellular Signal-Regulated MAP Kinases/antagonists & inhibitors , Humans , Keratinocytes/cytology , Keratolytic Agents/metabolism , MAP Kinase Signaling System/physiology , Permeability , RNA Interference , Skin/cytology , Vitamin B Complex/metabolism
7.
Cell Signal ; 20(10): 1830-8, 2008 Oct.
Article in English | MEDLINE | ID: mdl-18644433

ABSTRACT

Ultraviolet radiation (UV) induces apoptosis and functional maturation in skin dendritic cells (DCs). However, the molecular mechanisms through which UV activates DCs have not been thoroughly investigated. In this study, we examined the mechanisms of activation and apoptosis of DCs after UV irradiation by focusing on epidermal growth factor receptor (EGFR). Our previous studies have demonstrated that in addition to cognate ligands, EGFR is also activated by UVB irradiation in cultured human skin keratinocytes in vitro and in human skin in vivo. We found for the first time in this study that UV also induces EGFR activation in cultured mouse skin DCs (XS 106 cell line) as well as mouse monocyte-derived dendritic cells (MoDCs). Pharmacological inhibition of EGFR tyrosine kinase significantly inhibits UV-induced ERK, p38, and JNK MAP kinases, and their effectors, transcription factors c-Fos and c-Jun. Inhibition of EGFR also suppresses UV-induced activation of PI3K/AKT/mTOR/S6K and NF-kappaB signal transduction pathways. Our data demonstrated that UV induces LKB1/AMPK pathway, also dependent on EGFR trans-activation. We further observed that MAPK, LKB1/AMPK, PI3K/AKT/mTOR/S6K as well as NF-kappaB activation are impaired in EGFR-/- cells compared to wide type MEF cells after UV radiation. Taken together, we conclude that UV induces multiple signaling pathways mediated by EGFR trans-activation leading to possible maturation, apoptosis and survival, and EGFR activation protects against UV-induced apoptosis in cultured mouse dendritic cells.


Subject(s)
Apoptosis/radiation effects , Dendritic Cells/cytology , Dendritic Cells/enzymology , ErbB Receptors/metabolism , Skin/cytology , Ultraviolet Rays , AMP-Activated Protein Kinases , Adaptor Proteins, Signal Transducing , Animals , Carrier Proteins/metabolism , Cell Cycle Proteins , Cells, Cultured , Dendritic Cells/radiation effects , Enzyme Activation/radiation effects , ErbB Receptors/genetics , Eukaryotic Initiation Factors , Humans , Mice , Mitogen-Activated Protein Kinases/metabolism , Models, Biological , Multienzyme Complexes/metabolism , NF-kappa B/metabolism , Phosphoproteins/metabolism , Protein Kinases/metabolism , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Ribosomal Protein S6 Kinases/metabolism , Signal Transduction/radiation effects , Skin/enzymology , TOR Serine-Threonine Kinases , Transcriptional Activation/radiation effects
8.
Int J Mol Med ; 22(1): 9-16, 2008 Jul.
Article in English | MEDLINE | ID: mdl-18575770

ABSTRACT

Vitiligo is an acquired and progressive disorder manifested by the selective destruction of melanocytes in the skin. An extremely high level of hydrogen peroxide (H2O2) in plasma as well as in lesional skin has been reported in vitiligo patients. High H2O2 level has been suggested to be responsible for the disappearance of melanocytes in vitiligo. JNK and p38 MAPK are strongly induced by oxidative stress and related to neuron loss in neurodegenerative disorders. Minocycline, an antibiotic possessing antioxidant activity, is capable of attenuating oxidative stress-induced neurotoxicity. To investigate whether minocycline rescues melanocytes from H2O2-induced apoptosis, cultured mouse melanocytes (B10BR) were treated with H2O2 in the presence or absence of minocycline. Our data showed that H2O2 decreases cell viability in a concentration-dependent manner which is attenuated by minocycline. Also, H2O2 treatment activates JNK and p38 MAPK, and executive caspase 3 in B10BR cells. Minocycline significantly inhibits H2O2-induced activation of JNK, p38 MAPK and caspase 3. Collectively, we concluded that minocycline protects melanocytes against H2O2-induced apoptosis in vitro. Its protective effect is associated with the inhibition of JNK and p38 MAPK. Our findings suggest that minocycline, a clinically well-tolerated, safe antibiotic, may be used to prevent melanocyte loss in the early stage of vitiligo.


Subject(s)
Hydrogen Peroxide/pharmacology , JNK Mitogen-Activated Protein Kinases/metabolism , MAP Kinase Kinase Kinases/metabolism , MAP Kinase Signaling System/drug effects , Melanocytes/cytology , Melanocytes/drug effects , Minocycline/pharmacology , Proto-Oncogene Proteins/metabolism , Animals , Caspase 3/metabolism , Cell Death/drug effects , Cell Line , Enzyme Activation/drug effects , Melanins/biosynthesis , Melanocytes/enzymology , Mice , Phosphorylation/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...