RESUMO
Despite the capability of extracellular vesicles (EVs) derived from Gram-negative and Gram-positive bacteria to induce potent anti-tumour responses, large-scale production of bacterial EVs remains as a hurdle for their development as novel cancer immunotherapeutic agents. Here, we developed manufacturing processes for mass production of Escherichia coli EVs, namely, outer membrane vesicles (OMVs). By combining metal precipitation and size-exclusion chromatography, we isolated 357 mg in total protein amount of E. coli OMVs, which was equivalent to 3.93 × 1015 particles (1.10 × 1010 particles/µg in total protein amounts of OMVs) from 160 L of the conditioned medium. We show that these mass-produced E. coli OMVs led to complete remission of two mouse syngeneic tumour models. Further analysis of tumour microenvironment in neoantigen-expressing tumour models revealed that E. coli OMV treatment causes increased infiltration and activation of CD8+ T cells, especially those of cancer antigen-specific CD8+ T cells with high expression of TCF-1 and PD-1. Furthermore, E. coli OMVs showed synergistic anti-tumour activity with anti-PD-1 antibody immunotherapy, inducing substantial tumour growth inhibition and infiltration of activated cancer antigen-specific stem-like CD8+ T cells into the tumour microenvironment. These data highlight the potent anti-tumour activities of mass-produced E. coli OMVs as a novel candidate for developing next-generation cancer immunotherapeutic agents.
Assuntos
Vesículas Extracelulares , Neoplasias , Animais , Camundongos , Escherichia coli/metabolismo , Vesículas Extracelulares/química , Membrana Externa Bacteriana/metabolismo , Linfócitos T CD8-Positivos , Imunoterapia , Neoplasias/terapia , Neoplasias/metabolismoRESUMO
Dehydroabietic acid (DAA) is a naturally occurring diterpene resin acid derived from coniferous plants such as Pinus and Picea. Various bioactive effects of DAA have been studied including antibacterial, antifungal, and anticancer activities. However, the anti-inflammatory mechanism of DAA remains unclear. We evaluated the anti-inflammatory effect of DAA in macrophage cell lines. Dehydroabietic acid clearly reduced nitric oxide (NO) production and inflammatory gene expression decreased according to RT-PCR results. Dehydroabietic acid displayed anti-inflammatory activity at the transcriptional level in results from NF-κB- or AP-1-mediated luciferase assays. To identify the DAA target protein, we investigated NF-κB and AP-1 pathways by Western blotting analysis. Dehydroabietic acid suppressed the activity of proto-oncogene tyrosine protein kinase (Src) and spleen tyrosine kinase (Syk) in the NF-κB cascade and transforming growth factor beta-activated kinase 1 (TAK1) in the AP-1 cascade. Using overexpression strategies, we confirmed that DAA targeted these kinases. Our findings demonstrate the anti-inflammatory effects and molecular mechanism of DAA. This suggests that DAA has potential as a drug or supplement to ameliorate inflammation.
Assuntos
Abietanos/farmacologia , Inflamação/patologia , MAP Quinase Quinase Quinases/metabolismo , Transdução de Sinais , Quinase Syk/metabolismo , Quinases da Família src/metabolismo , Abietanos/química , Animais , Anti-Inflamatórios/farmacologia , Morte Celular/efeitos dos fármacos , Camundongos , Modelos Biológicos , NF-kappa B/metabolismo , Óxido Nítrico/biossíntese , Células RAW 264.7 , Transdução de Sinais/efeitos dos fármacos , Fator de Transcrição AP-1/metabolismo , Transcrição Gênica/efeitos dos fármacosRESUMO
BACKGROUND: Ultraviolet B (UVB) radiation is a major cause of skin photodamage, including the damage associated with photodermatoses, aging, and cancer. Although many studies have shown that red light has photoprotective effects on skin, the mechanisms underlying these effects are still poorly understood. OBJECTIVE: The aim of this study was to identify the photoprotective effects of visible red light against UVB-induced skin damage in normal human dermal fibroblast cells using a transcriptomic approach. METHODS: Next-generation sequencing-based transcriptomic analyses were used to profile transcriptomic alterations and identify genes that are differentially expressed by visible red light and by UVB exposure. To understand the biological networks among identified genes, a literature-based biological pathway analysis was performed. Quantitative real-time polymerase chain reaction assays were used for mRNA-level validation of selected key genes. RESULTS: We observed that visible red light contributes to skin cell protection against UVB by modulating gene expression that enhances the adaptive response to redox and inflammatory balancing and by upregulating genes involved in DNA excision repair processes. We also identified that several key genes in the red light-induced biological network were differentially regulated. CONCLUSIONS: Visible red light enhanced the UVB-protective effects in normal human skin cells via the transcriptomic modulation of genes involved in cell-protective processes. Our findings from this next-generation sequencing analysis may lead to a better understanding of the cytoprotective effects of visible red light and provide direction for further molecular or mechanistic studies.
Assuntos
Reparo do DNA/genética , Fibroblastos/efeitos da radiação , Luz , Pele/efeitos da radiação , Raios Ultravioleta/efeitos adversos , Linhagem Celular , Dano ao DNA/efeitos da radiação , Reparo do DNA/efeitos da radiação , Fibroblastos/metabolismo , Perfilação da Expressão Gênica , Humanos , RNA Mensageiro/metabolismo , Transdução de Sinais/genética , Transdução de Sinais/efeitos da radiação , Pele/citologia , Regulação para Cima/efeitos da radiaçãoRESUMO
Exposure to fine particulate matter (PM) with diameter <2.5 µm (PM2.5) causes epithelium injury and endothelial dysfunction. Primary cilia are sensory organelles that transmit extracellular signals into intracellular biochemical responses and have roles in physiology. To date, there have been no studies investigating whether PM2.5 affects primary cilia in skin. We addressed this in the present study using normal human epidermal keratinocytes (NHEKs) and retinal pigment epithelium (RPE) cells. We found that formation of primary cilium is increased in differentiated NHEKs. However, treatment with PM2.5 blocked increased ciliogenesis in NHEKs and RPE cells. Furthermore, PM2.5 transcriptionally upregulated small proline rich protein 3 (SPRR3) expression by activating c-Jun, and ectopic expression of SPRR3 inhibits suppressed the ciliogenesis. Accordingly, treatment with c-Jun activator (anisomycin) induced SPRR3 expression, whereas the inhibitor (SP600125) recovered the ciliated cells and cilium length in PM2.5-treated cells. Moreover, c-Jun inhibitor suppressed upregulation of SPRR3 in PM2.5-treated cells. Taken together, our finding suggested that PM2.5 inhibits ciliogenesis by increasing SPRR3 expression via c-Jun activation in RPE cells and keratinocytes.
Assuntos
Cílios/efeitos dos fármacos , Proteínas Ricas em Prolina do Estrato Córneo/metabolismo , Queratinócitos/efeitos dos fármacos , Material Particulado/farmacologia , Proteínas Proto-Oncogênicas c-jun/metabolismo , Epitélio Pigmentado da Retina/efeitos dos fármacos , Pele/efeitos dos fármacos , Diferenciação Celular/efeitos dos fármacos , Linhagem Celular , Cílios/metabolismo , Humanos , Queratinócitos/metabolismo , Estresse Oxidativo/efeitos dos fármacos , Espécies Reativas de Oxigênio/metabolismo , Epitélio Pigmentado da Retina/metabolismo , Pele/metabolismo , Regulação para Cima/efeitos dos fármacosRESUMO
Hyperpigmentation is caused by excessive production of melanin in melanocytes. Mannosylerythritol lipids (MELs) are glycolipid biosurfactants that are abundantly produced by yeasts and used commercially in cosmetics. However, the potential depigmenting efficacy of MELs has not been evaluated. In this study, the depigmentary effect of MELs was tested in primary normal human melanocytes (NHMs), α-melanocyte-stimulating hormone (MSH)-stimulated B16 cells (murine melanoma cells) and a human skin equivalent (MelanoDerm) using photography, Fontana-Masson (F&M) staining and two-photon microscopy. Mannosylerythritol lipids significantly decreased the melanin contents in NHMs and α-MSH-stimulated B16 cells. Consistent with these findings, MELs treatment had a clear whitening effect in a human skin equivalent, brightening the tissue colour and reducing the melanin content. The molecular mechanism underlying the anti-melanogenic effect of MELs treatment was examined by real-time PCR and Western blotting. Mechanistically, MELs clearly suppressed the gene expression levels of representative melanogenic enzymes, including tyrosinase, Tyrp-1 and Tyrp-2, by inhibiting the ERK/CREB/MiTF signalling pathway in NHMs. This work demonstrates for the first time that MELs exert whitening effects on human melanocytes and skin equivalent. Thus, we suggest that MELs could be developed as a potent anti-melanogenic agent for effective whitening, beyond their use as a biosurfactant in cosmetics.
Assuntos
Glicolipídeos/farmacologia , Hiperpigmentação/tratamento farmacológico , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Melanócitos/efeitos dos fármacos , Animais , Linhagem Celular , Avaliação Pré-Clínica de Medicamentos , Glicolipídeos/uso terapêutico , Humanos , Melaninas/biossíntese , Melanócitos/metabolismo , Camundongos , Cultura Primária de CélulasRESUMO
Hypoxia-inducible factor-1α (HIF-1α) has been reported to be up-regulated in psoriatic epidermis, resulting in increased proliferation and abnormal differentiation of human keratinocytes (KCs). However, the role of HIF-1α in psoriatic epidermis, which is mainly composed of KCs, is poorly understood. Here, we show that morphogenic protein 6 (BMP6) is down-regulated when HIF-1α is upregulated in patients with psoriasis skin lesions. HIF-1α overexpression in primary human KCs promoted proliferation and inhibited terminal differentiation. Furthermore, HIF1-α repressed the expression of BMP6 by binding directly to the hypoxia-response element (HRE) in the BMP6 promotor region, which shows that BMP6 is a novel target gene of HIF-1α. We also found that HIF-1α-mediated BMP6 suppression could alter the proliferation status by modulating the expression levels of cell cycle regulatory proteins and also affect the early differentiation of KCs. Therefore, we suggest that HIF-1α-dependent BMP6 suppression has a critical role in the induction of hyper-proliferation and abnormal differentiation in psoriatic KCs.
Assuntos
Proteína Morfogenética Óssea 6/metabolismo , Diferenciação Celular/genética , Proliferação de Células/genética , Subunidade alfa do Fator 1 Induzível por Hipóxia/genética , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Psoríase/genética , Antígenos de Neoplasias/metabolismo , Proteína Morfogenética Óssea 6/farmacologia , Anidrase Carbônica IX/metabolismo , Ciclo Celular/genética , Diferenciação Celular/efeitos dos fármacos , Hipóxia Celular , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , Regulação para Baixo , Transportador de Glucose Tipo 1/metabolismo , Humanos , Queratinócitos/fisiologia , Cultura Primária de Células , Regiões Promotoras Genéticas , Psoríase/metabolismo , TransfecçãoRESUMO
Mitochondrial dysfunction can drive cellular senescence, which is accompanied by changes in metabolism and increases in senescence-associated secretory phenotypes. Although pyruvate, a key metabolite for numerous aspects of metabolism, has been used as general supplement in synthetic media, the physiological function of pyruvate underlying its protective role against cellular senescence under normal conditions has remained unknown. Here, we show that extracellular pyruvate prevents senescence in normal human dermal fibroblasts through increasing the generation of oxidized nicotinamide adenine dinucleotide (NAD+) during the conversion to lactate. Acetylated peroxisome proliferator-activated receptor gamma coactivator 1α (PGC-1α), vacuolar-type H+-ATPaseV0A1 (v-ATPaseV0A1), NF-κB p65 subunit (RelA), and histone H3 accumulate under pyruvate deprivation conditions, resulting in the onset of senescence in normal human dermal fibroblasts through the accumulation of abnormal mitochondria generated by lysosomal inactivation-induced mitophagy defects, and through an increase in senescence-associated secretory phenotypes. Furthermore, pyruvate showed a protective effect against aging phenotypes in skin equivalents, which consist of a dermis and epidermis that act similarly to in vivo skin tissues. Our findings reveal a connection between pyruvate and mitochondrial dysfunction in the progression of senescence that is, to our knowledge, previously unreported. These results suggest that the pyruvate deprivation-induced senescence model can be used to study the connection between metabolism and senescence under normal conditions.
Assuntos
Senescência Celular , Derme/patologia , Epiderme/patologia , Fibroblastos/fisiologia , Lisossomos/metabolismo , Mitocôndrias/metabolismo , Ácido Pirúvico/metabolismo , Células Cultivadas , Derme/metabolismo , Epiderme/metabolismo , Histonas/metabolismo , Humanos , Ligases/metabolismo , Mitocôndrias/patologia , Mitofagia , NAD/metabolismo , PPAR gama/metabolismoRESUMO
8-Hydroxydaidzein (8-HD) is a daidzein metabolite isolated from soybeans. This compound has been studied for its anti-proliferation, depigmentation, and antioxidant activities. However, the anti-inflammatory activities of 8-HD are not well-understood. Through its antioxidant effects in ABTS and DPPH assays, 8-HD reduces the production of sodium nitroprusside (SNP)-induced radical oxygen species (ROS). By triggering various Toll-like receptors (TLRs), 8-HD suppresses the inflammatory mediator nitric oxide (NO) without cytotoxicity. We examined the regulatory mechanism of 8-HD in lipopolysaccharide (LPS)-induced conditions. We found that 8-HD diminishes inflammatory gene expression (e.g., inducible nitric oxide synthase (iNOS), cyclooxygenase (COX)-2, and tumor necrosis factor (TNF)-α) by regulating the transcriptional activities of nuclear factor (NF)-κB and activator protein 1 (AP-1). To find the potential targets of 8-HD, signaling pathways were investigated by immunoblotting analyses. These analyses revealed that 8-HD inhibits the activation of TAK1 and that phosphorylated levels of downstream molecules decrease in sequence. Together, our results demonstrate the antioxidant and anti-inflammatory actions of 8-HD and suggest its potential use in cosmetics or anti-inflammatory drugs.
Assuntos
Anti-Inflamatórios/farmacologia , Antioxidantes/farmacologia , Regulação da Expressão Gênica/efeitos dos fármacos , Isoflavonas/farmacologia , Lipopolissacarídeos/antagonistas & inibidores , Animais , Anti-Inflamatórios/isolamento & purificação , Antioxidantes/isolamento & purificação , Ciclo-Oxigenase 2/genética , Ciclo-Oxigenase 2/imunologia , Isoflavonas/isolamento & purificação , Lipopolissacarídeos/farmacologia , MAP Quinase Quinase Quinases/genética , MAP Quinase Quinase Quinases/imunologia , Ativação de Macrófagos/efeitos dos fármacos , Camundongos , NF-kappa B/antagonistas & inibidores , NF-kappa B/genética , NF-kappa B/imunologia , Óxido Nítrico Sintase Tipo II/antagonistas & inibidores , Óxido Nítrico Sintase Tipo II/genética , Óxido Nítrico Sintase Tipo II/imunologia , Células RAW 264.7 , Transdução de Sinais , Glycine max/química , Fator de Transcrição AP-1/genética , Fator de Transcrição AP-1/imunologia , Fator de Necrose Tumoral alfa/antagonistas & inibidores , Fator de Necrose Tumoral alfa/genética , Fator de Necrose Tumoral alfa/imunologiaRESUMO
Melanin synthesis in melanocytes is affected by various cytokines. Here, we reported for the first time that tumor necrosis factor superfamily member 14 (TNFSF14) inhibits melanogenesis in the primary culture of human epidermal melanocytes. TNFSF14 is known to bind to its receptors herpes virus entry mediator (HVEM) and lymphotoxin ß receptor (LTßR) for signal transduction, but TNFSF14-induced hypopigmentation was independent of HVEM and LTßR in melanocytes. To explore signaling in melanocytes treated with TNFSF14, we performed RNA-seq and found that nuclear factor κ-light-chain-enhancer of activated B cells (NF-κB) signaling is activated by TNFSF14. Further, we observed that inhibition of NF-kB effectively blocks the hypopigmentation induced by TNFSF14. We conclude that TNFSF14 inhibits melanogenesis in melanocytes via NF-κB signaling and could be applied in the treatment of cutaneous pigment disorders.
Assuntos
Melanócitos/metabolismo , NF-kappa B/metabolismo , Transdução de Sinais/fisiologia , Membro 14 da Superfamília de Ligantes de Fatores de Necrose Tumoral/metabolismo , Linhagem Celular , Humanos , Ativação Linfocitária/fisiologia , Receptor beta de Linfotoxina/metabolismo , Melaninas/metabolismo , Membro 14 de Receptores do Fator de Necrose Tumoral/metabolismoRESUMO
BACKGROUND: Ultraviolet radiation (UVR) is a well-known factor in skin aging and pigmentation, and daily exposure to subcytotoxic doses of UVR might accelerate senescence and senescence-associated phenomena in human melanocytes. OBJECTIVE: To establish an in vitro melanocyte model to mimic the conditions of repeated exposure to subcytotoxic doses of UVB irradiation and to investigate key factor(s) for melanocyte senescence and senescence-associated phenomena. METHODS: Human epidermal melanocytes were exposed twice with 20â¯mJ/cm2 UVB over a 24-h interval and subsequently cultivated for 2 weeks. Senescent phenotypes were addressed morphologically, and by measuring the senescence-associated ß-galactosidase (SA-ß-Gal) activity, cell proliferation capacity with cell cycle analysis, and melanin content. RESULTS: The established protocol successfully induced melanocyte senescence, and senescent melanocytes accompanied hyperpigmentation. Prolonged expression of p53 was responsible for melanocyte senescence and hyperpigmentation, and treatment with the p53-inhibitor pifithrin-α at 2-weeks post-UVB irradiation, but not at 48â¯h, significantly reduced melanin content along with decreases in tyrosinase levels. CONCLUSION: Melanocyte senescence model will be useful for studying the long-term effects of UVB irradiation and pigmentation relevant to physiological photoaging, and screening compounds effective for senescence-associated p53-mediated pigmentation.
Assuntos
Senescência Celular/efeitos da radiação , Envelhecimento da Pele/efeitos da radiação , Pigmentação da Pele/efeitos da radiação , Proteína Supressora de Tumor p53/metabolismo , Raios Ultravioleta/efeitos adversos , Apoptose/efeitos da radiação , Benzotiazóis/farmacologia , Proliferação de Células/efeitos da radiação , Células Cultivadas , Senescência Celular/efeitos dos fármacos , Células Epidérmicas , Epiderme/patologia , Epiderme/efeitos da radiação , Humanos , Recém-Nascido , Masculino , Melaninas/metabolismo , Melanócitos/efeitos da radiação , Envelhecimento da Pele/patologia , Pigmentação da Pele/efeitos dos fármacos , Tolueno/análogos & derivados , Tolueno/farmacologia , Proteína Supressora de Tumor p53/antagonistas & inibidoresRESUMO
Benzophenone-3 (BP-3), which is extensively used in organic sunscreen, has phototoxic potential in human skin. Phosphodiesterase 4B (PDE4B) has a well-established role in inflammatory responses in immune cells. Currently, it is unknown if PDE4B is associated with BP-3-induced phototoxicity in normal human keratinocytes (NHKs). We found that BP-3 significantly increased PDE4B expression in ultraviolet B (UVB)-irradiated NHKs. Notably, BP-8, a sunscreen agent that shares the 2-hydroxy-4-methoxyphenyl methanone moiety with BP-3, also upregulated PDE4B expression in NHKs. Upon UVB irradiation, BP-3 upregulated the expression of pro-inflammatory factors, such as prostaglandin endoperoxide synthase 2, tumor necrosis factor α, interleukin 8, and S100A7, and downregulated the level of cornified envelope associated proteins, which are important in the development of the epidermal permeability barrier. The additive effects of UVB-activated BP-3 on the expression of both pro-inflammatory mediators and cornified envelope associated proteins were antagonized by treatment with the PDE4 inhibitor rolipram. The BP-3 and UVB co-stimulation-induced PDE4B upregulation and its association with the upregulation of pro-inflammatory mediators and the downregulation of epidermal differentiation markers were confirmed in a reconstituted three dimensional human epidermis model. Therefore, PDE4B has a role in the mechanism of BP-3-induced phototoxicity.
Assuntos
Benzofenonas/toxicidade , Nucleotídeo Cíclico Fosfodiesterase do Tipo 4/fisiologia , Dermatite Fototóxica/etiologia , Queratinócitos/efeitos dos fármacos , AMP Cíclico/fisiologia , Nucleotídeo Cíclico Fosfodiesterase do Tipo 4/genética , Dinoprostona/biossíntese , Humanos , Interleucina-8/biossíntese , Fator de Necrose Tumoral alfa/biossíntese , Raios UltravioletaRESUMO
We introduce a novel microfluidic device to co-culture a blood vessel network and cell tissues in an in vivo-like niche. Our "open-top" microfluidic device is composed of microchannels with micropores in the ceiling, which provides direct fluid access from reservoir to microchannel. Fluid connections through micropores afford novel advantages, including: i) the long-term culture of large-scale microvessel network, ii) access of different fluids to inner and exterior sides of the microvessel, and iii) co-culturing of the microvessel network and small cell tissue. In this study, we have successfully assembled microvessels with 5 mm channel widths. We were also able to mimic capillary bed conditions by co-culturing microvessels with cancer spheroids. Intimate contact between the cancer spheroid and microvessel caused vessel recruitment and an increase in vessel formation, and affected vessel morphology. We expect this device to be used as a novel platform for vascularized tissue models.
Assuntos
Dispositivos Lab-On-A-Chip , Técnicas Analíticas Microfluídicas/instrumentação , Linhagem Celular , Técnicas de Cocultura , Técnicas Citológicas/instrumentação , Técnicas Citológicas/métodos , Desenho de Equipamento , Humanos , Microvasos/citologia , Microvasos/metabolismo , Microvasos/fisiologia , Esferoides Celulares , Células Tumorais CultivadasRESUMO
Ultraviolet (UV) radiation causes the harmful effects on skin by the photochemical reaction and gene expression regulation. Recent evidences have shown that long non-coding RNAs (lncRNAs) play critical roles in a diverse range of biological functions. However, research on the effects of UV irradiation on lncRNA expression in epidermal cells is limited. The aim of this study was to identify changes in the expression profile of lncRNAs after UVB irradiation. To accomplish this, we performed a microarray analysis of both mRNA and lncRNA expression levels in irradiated skin cells. Gene ontology (GO) analysis of differentially expressed mRNAs showed that the expression of immune response- and cell membrane-related genes was up-regulated, while cell-cell adhesion-associated genes were down-regulated by UVB irradiation. Moreover, we found that lncRNAs up-regulated by UVB irradiation were associated with the regulation of gene transcription, while lncRNAs down-regulated by UVB irradiation were associated with tumorigenesis. Finally, we compiled a list of the lncRNAs that showed the strongest association with the development of non-melanoma skin cancers caused by UV exposure. These findings lay a foundation for future investigations into the expression patterns of lncRNAs with roles in the response to UV irradiation and in non-melanoma skin cancers.
Assuntos
Transformação Celular Neoplásica/genética , Epiderme/metabolismo , Perfilação da Expressão Gênica , Regulação Neoplásica da Expressão Gênica/efeitos da radiação , Queratinócitos/metabolismo , RNA Longo não Codificante/genética , Raios Ultravioleta , Transformação Celular Neoplásica/patologia , Transformação Celular Neoplásica/efeitos da radiação , Células Cultivadas , Células Epidérmicas , Epiderme/efeitos da radiação , Ontologia Genética , Humanos , Queratinócitos/citologia , Queratinócitos/efeitos da radiaçãoRESUMO
Our previous work has identified miR-125b as a negative regulator of melanogenesis. However, the specific melanogenesis-related genes targeted by this miRNA had not been identified. In this study, we established a screening strategy involving three consecutive analytical approaches-analysis of target genes of miR-125b, expression correlation analysis between each target gene and representative pigmentary genes, and functional analysis of candidate genes related to melanogenesis-to discover melanogenesis-related genes targeted by miR-125b. Through these analyses, we identified SRC homology 3 domain-binding protein 4 (SH3BP4) as a novel pigmentation gene. In addition, by combining bioinformatics analysis and experimental validation, we demonstrated that SH3BP4 is a direct target of miR-125b. Finally, we found that SH3BP4 is transcriptionally regulated by microphthalmia-associated transcription factor as its direct target. These findings provide important insights into the roles of miRNAs and their targets in melanogenesis.
Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Regulação da Expressão Gênica , Melaninas/biossíntese , MicroRNAs/metabolismo , Fator de Transcrição Associado à Microftalmia/metabolismo , Pigmentação da Pele/genética , Proteínas Adaptadoras de Transdução de Sinal/genética , Linhagem Celular Tumoral , Expressão Gênica , Humanos , Melaninas/genética , Melanócitos/metabolismo , MicroRNAs/genética , Fator de Transcrição Associado à Microftalmia/genética , RNA Mensageiro , Estatística como Assunto , Domínios de Homologia de srcRESUMO
Expression profiles revealed miR-1299 downregulation concomitant with arginase-2 (ARG2) upregulation in hyperpigmented skin of melasma patients. Opposite regulation of tyrosinase and PMEL17 by miR-1299 and inverse relationship between miR-1299 and ARG2 expression denoted a role of miR-1299 in pigmentation with ARG2 as a miR-1299 target. ARG2 overexpression or knock-down in keratinocytes, the main source of ARG2 in epidermis, positively regulated tyrosinase and PMEL17 protein levels, but not mRNA levels or melanosome transfer. ARG2 overexpression in keratinocytes reduced autophagy equivalent to 3-MA, an autophagy inhibitor which also increased tyrosinase and PMEL17 protein levels, whereas ARG2 knock-down induced opposite results. Autophagy inducer rapamycin reduced ARG2-increased tyrosinase and PMEL17 protein levels. Also, autophagy was reduced in late passage-induced senescent keratinocytes showing ARG2 upregulation. ARG2, but not 3-MA, stimulated keratinocyte senescence. These results suggest that ARG2 reduces autophagy in keratinocytes by stimulating cellular senescence, resulting in skin pigmentation by reducing degradation of transferred melanosomes.
Assuntos
Arginase/metabolismo , Autofagia/genética , Senescência Celular/genética , Melanose/genética , Melanose/patologia , Melanossomas/metabolismo , MicroRNAs/metabolismo , Pigmentação da Pele/genética , Adulto , Sequência de Bases , Feminino , Regulação da Expressão Gênica , Técnicas de Silenciamento de Genes , Humanos , Melaninas/biossíntese , MicroRNAs/genética , Pessoa de Meia-IdadeRESUMO
The phototherapeutic effects of visible red light on skin have been extensively investigated, but the underlying biological mechanisms remain poorly understood. We aimed to elucidate the protective mechanism of visible red light in terms of DNA repair of UV-induced oxidative damage in normal human dermal fibroblasts. The protective effect of visible red light on UV-induced DNA damage was identified by several assays in both two-dimensional and three-dimensional cell culture systems. With regard to the protective mechanism of visible red light, our data showed alterations in base excision repair mediated by growth arrest and DNA damage inducible, alpha (GADD45A). We also observed an enhancement of the physical activity of GADD45A and apurinic/apyrimidinic endonuclease 1 (APE1) by visible red light. Moreover, UV-induced DNA damages were diminished by visible red light in an APE1-dependent manner. On the basis of the decrease in GADD45A-APE1 interaction in the activating transcription factor-2 (ATF2)-knockdown system, we suggest a role for ATF2 modulation in GADD45A-mediated DNA repair upon visible red light exposure. Thus, the enhancement of GADD45A-mediated base excision repair modulated by ATF2 might be a potential protective mechanism of visible red light.
Assuntos
Proteínas de Ciclo Celular/fisiologia , Citoproteção , Reparo do DNA , Luz , Proteínas Nucleares/fisiologia , Pele/efeitos da radiação , Fator 2 Ativador da Transcrição/fisiologia , Células Cultivadas , DNA Liase (Sítios Apurínicos ou Apirimidínicos)/fisiologia , Fibroblastos/efeitos da radiação , Humanos , Pele/metabolismo , Proteína Supressora de Tumor p53/fisiologiaRESUMO
The primary cilium is an organelle protruding from the cell body that senses external stimuli including chemical, mechanical, light, osmotic, fluid flow, and gravitational signals. Skin is always exposed to the external environment and responds to external stimuli. Therefore, it is possible that primary cilia have an important role in skin. Ciliogenesis was reported to be involved in developmental processes in skin, such as keratinocyte differentiation and hair formation. However, the relation between skin pigmentation and primary cilia is largely unknown. Here, we observed that increased melanogenesis in melanocytes treated with a melanogenic inducer was inhibited by a ciliogenesis inducer, cytochalasin D, and serum-free culture. However, these inhibitory effects disappeared in GLI2 knockdown cells. In addition, activation of sonic hedgehog (SHH)-smoothened (Smo) signaling pathway by a Smo agonist, SAG inhibited melanin synthesis in melanocytes and pigmentation in a human skin model. On the contrary, an inhibitor of primary cilium formation, ciliobrevin A1, activated melanogenesis in melanocytes. These results suggest that skin pigmentation may be regulated partly by the induction of ciliogenesis through Smo-GLI2 signaling.
Assuntos
Cílios/fisiologia , Melaninas/biossíntese , Melanócitos/fisiologia , Fenômenos Fisiológicos da Pele , Pigmentação da Pele/fisiologia , Pele/citologia , Animais , Linhagem Celular , Linhagem Celular Tumoral , Cílios/efeitos dos fármacos , Citocalasina D/farmacologia , Proteínas Hedgehog/metabolismo , Humanos , Fatores de Transcrição Kruppel-Like/metabolismo , Melanócitos/citologia , Camundongos , Camundongos Endogâmicos C57BL , Proteínas Nucleares/metabolismo , Quinazolinonas/farmacologia , Transdução de Sinais , Pele/efeitos dos fármacos , Pele/metabolismo , Fenômenos Fisiológicos da Pele/efeitos dos fármacos , Pigmentação da Pele/efeitos dos fármacos , Proteína Gli2 com Dedos de ZincoRESUMO
The regulation of melanin production is important for managing skin darkness and hyperpigmentary disorders. Numerous anti-melanogenic agents that target tyrosinase activity/stability, melanosome maturation/transfer, or melanogenesis-related signaling pathways have been developed. As a rate-limiting enzyme in melanogenesis, tyrosinase has been the most attractive target, but tyrosinase-targeted treatments still pose serious potential risks, indicating the necessity of developing lower-risk anti-melanogenic agents. Sugars are ubiquitous natural compounds found in humans and other organisms. Here, we review the recent advances in research on the roles of sugars and sugar-related agents in melanogenesis and in the development of sugar-based anti-melanogenic agents. The proposed mechanisms of action of these agents include: (a) (natural sugars) disturbing proper melanosome maturation by inducing osmotic stress and inhibiting the PI3 kinase pathway and (b) (sugar derivatives) inhibiting tyrosinase maturation by blocking N-glycosylation. Finally, we propose an alternative strategy for developing anti-melanogenic sugars that theoretically reduce melanosomal pH by inhibiting a sucrose transporter and reduce tyrosinase activity by inhibiting copper incorporation into an active site. These studies provide evidence of the utility of sugar-based anti-melanogenic agents in managing skin darkness and curing pigmentary disorders and suggest a future direction for the development of physiologically favorable anti-melanogenic agents.
Assuntos
Carboidratos/química , Carboidratos/farmacologia , Melaninas/antagonistas & inibidores , Melaninas/metabolismo , Pigmentação da Pele/efeitos dos fármacos , Animais , Antígenos de Neoplasias/metabolismo , Humanos , Proteínas de Membrana Transportadoras/metabolismoRESUMO
Psoriasin (S100A7), a member of the S100 protein family, is a well-known antimicrobial peptide and a signalling molecule which regulates cellular function and is highly expressed in hyperproliferative skin conditions such as atopic dermatitis (AD) and psoriasis with disrupted skin barrier function. However, its role in epidermal differentiation remains unknown. We examined the effect of S100A7 on epidermal differentiation in normal human keratinocytes (NHKs) and on a reconstituted human epidermis model. When NHKs were exposed to disruptive stimuli such as Staphylococcus aureus, ultraviolet irradiation and retinoic acid, the secretion of S100A7 into the culture medium increased and the expression of epidermal differentiation markers decreased. Treatment of NHKs with S100A7 significantly inhibited epidermal differentiation by reducing the expression of keratin 1, keratin 10, involucrin and loricrin and by increasing the expression of abnormal differentiation markers (keratin 6 and keratin 16). We verified that the MyD88-IκB/NF-κB signal cascade was activated via RAGE after S100A7 treatment, resulting in the upregulation of interleukin-6. Finally, we confirmed that S100A7 is a negative regulator of epidermal differentiation using a reconstituted human epidermis model. This study suggests that S100A7-related signalling molecules could be potent targets for recovering skin barrier function in AD and psoriasis where S100A7 is accumulated excessively.
Assuntos
Diferenciação Celular , Epiderme/metabolismo , Interleucina-6/metabolismo , Queratinócitos/metabolismo , Proteína A7 Ligante de Cálcio S100/metabolismo , Células Cultivadas , Células Epidérmicas , Humanos , Queratinócitos/citologia , Transdução de Sinais , Estresse FisiológicoRESUMO
NO regulates a variety of physiological processes, including cell proliferation, differentiation, and inflammation. S-nitrosylation, a NO-mediated reversible protein modification, leads to changes in the activity and function of proteins. In particular, the role of S-nitrosylation during adipogenesis is largely unknown. We hypothesized that the normal physiological levels of NO, but not the excess levels generated under severe conditions, such as inflammation, may be critically involved in the proper regulation of adipogenesis. We found that endogenous S-nitrosylation of proteins was required for adipocyte differentiation. By performing a biotin-switch assay, we identified FAS, a key lipogenic enzyme in adipocytes, as a target of S-nitrosylation during adipogenesis. Interestingly, we also observed that the dimerization of FAS increased in parallel with the amount of S-nitrosylated FAS during adipogenesis. In addition, we found that exogenous NO enhanced the dimerization and the enzymatic activity of FAS. Moreover, site-directed mutagenesis of three predicted S-nitrosylation sites indicated that S-nitrosylation of FAS at Cys(1471)and Cys(2091), but not at Cys(1127), increased its enzymatic activity. Taken together, these results suggest that the S-nitrosylation of FAS at normal physiological levels of NO increases its activity through dimerization and may contribute to the proper regulation of adipogenesis.