Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
2.
Drug Deliv Transl Res ; 9(6): 1143-1158, 2019 12.
Artigo em Inglês | MEDLINE | ID: mdl-31317345

RESUMO

Scarless healing of injury remains a clinical challenge because of its complicated and overlapping phases of inflammation, clearing, and regeneration. Curcumin has been already established as a potential wound healing agent for normal and diabetic-impaired wounds. Herein, the question has been addressed whether a well-known antioxidant cerium oxide nanoparticle (CNP) can potentiate the activity of curcumin to promote a cellular program for scarless healing. In this study, we have developed a biocompatible poly (acrylamide) hydrogel (PAGE)-based dressing material comprising of CNP and curcumin (ACC) and tested its wound healing activity in an animal model of acute wound. Characterization of the CNP- and curcumin-entrapped hydrogel dressing (ACC) demonstrated high loading efficiency and sustained release of curcumin. In a full-thickness acute wound healing model of rat, a single application of ACC dressing demonstrated higher wound healing efficacy (78%) and negligible scarring compared to dressings containing only curcumin or CNP in 7 days. Enhanced cell proliferation, higher collagen content, advanced wound maturity, re-epithelialization, and granulation tissue formation were observed using the combination of curcumin and CNP (ACC). Study of cellular mechanisms identified MCP-1 and TGF-ß as the key drivers of differential and accelerated healing observed in the ACC group. These, coupled with the upregulation of growth-related signaling pathways (HER2/ErbB2, TGF-ß-Smad2/3, MAPK/ERK, AKT, and VEGF), promoted almost scarless healing in animals treated with ACC. The optimized combination of curcumin and CNP used in our study shows distinct advantage and can be a better agent for complete wound healing.


Assuntos
Cério/administração & dosagem , Curcumina/administração & dosagem , Portadores de Fármacos/administração & dosagem , Hidrogéis/administração & dosagem , Nanopartículas/administração & dosagem , Cicatrização/efeitos dos fármacos , Animais , Linhagem Celular , Cério/química , Curcumina/química , Citocinas/sangue , Portadores de Fármacos/química , Liberação Controlada de Fármacos , Humanos , Hidrogéis/química , Masculino , Nanopartículas/química , Proteínas Quinases/metabolismo , Ratos Wistar , Pele/efeitos dos fármacos , Pele/metabolismo , Pele/patologia
3.
Photodermatol Photoimmunol Photomed ; 34(1): 82-90, 2018 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-28857273

RESUMO

BACKGROUND: Ultraviolet B (UVB) radiation is the major contributor to skin inflammation which leads to the development of skin cancer. Hence, in this study, we studied the effect of Nexrutine (NX) on UVB-induced cutaneous inflammation and its mediators. METHODS: Ultraviolet absorption spectra of NX were measured by spectrophotometer. To conduct the photoprotective studies, SKH-1 hairless mice were topically treated with NX, 30 minutes before to the UVB (180 mJ/cm2 ) exposure. Twenty hours of post-UVB irradiation, mouse skin was used for edema measurements, H & E staining, myeloperoxidase (MPO) activity, and estimation of plasma cytokines. In addition, expression levels of inflammatory cytokines, cyclooxygenase-2 (COX-2), and inducible nitric oxide synthase (iNOS) were also determined by Western blot analysis. RESULTS: Nexrutine displayed absorbance over the UVB spectrum. NX significantly decreased the UVB-induced epidermal edema, skin thickness, leukocyte infiltration, number of the sunburn, and TUNEL-positive cells. NX treatment also decreased the number of mast cells, MPO activity, expression of pro-inflammatory cytokines, and inflammation mediator protein in mouse skin. CONCLUSION: These results provide evidences that NX inhibits the UVB-induced cutaneous inflammatory responses in SKH-1 mouse skin.


Assuntos
Edema/prevenção & controle , Extratos Vegetais/uso terapêutico , Radiodermite/etiologia , Radiodermite/prevenção & controle , Queimadura Solar/prevenção & controle , Raios Ultravioleta/efeitos adversos , Absorção de Radiação , Administração Cutânea , Animais , Ciclo-Oxigenase 2/metabolismo , Citocinas/metabolismo , Fragmentação do DNA/efeitos dos fármacos , Fragmentação do DNA/efeitos da radiação , Edema/etiologia , Feminino , Camundongos , Camundongos Pelados , Óxido Nítrico Sintase Tipo II/metabolismo , Peroxidase/metabolismo , Extratos Vegetais/administração & dosagem , Radiodermite/enzimologia , Radiodermite/patologia , Espectrofotometria , Queimadura Solar/etiologia
4.
Artigo em Inglês | MEDLINE | ID: mdl-27542711

RESUMO

UV-induced reactive oxygen species (ROS) have been implicated in photocarcinogenesis and skin aging. This is because UV-induced ROS can induce DNA damage that, if unrepaired, can lead to carcinogenesis. Sunscreens contain UV attenuators, such as organic chemical and/or physical UV filters, which can prevent all forms of damage from UV irradiation. In recent years, the effective broad-spectrum UV attenuation properties of ZnO-nanoparticles (ZnO-NPs) have made them attractive as active components in sunscreens and other personal care products. As the use of ZnO-NPs in sunscreens is on the rise, so is public concern about their safety, particularly with exposure to sunlight. Therefore, in the present study, using various experimental approaches, we investigated the possible toxic effects resulting from exposure to UVB and ZnO-NPs in primary mouse keratinocytes (PMKs) as well as in the skin of SKH-1 hairless mice. The findings of the present study demonstrated that co-exposure to UVB and ZnO-NPs: (1) translocated the ZnO-NPs into the nucleus of PMKs; (2) caused enhanced generation of ROS; (3) induced more severe DNA damage as evident by alkaline comet assay and immunocytochemistry for γ-H2AX and 8-hydroxy-2'-deoxyguanosine (8-OHdG); and (4) subsequently caused much more pronounced cell death in PMKs. Further, to elucidate the physiological relevance of these in vitro findings, SKH-1 hairless mice were topically treated with ZnO-NPs and after 30min irradiated with UVB (50mJ/cm(2)). Interestingly, we found that co-exposure of ZnO-NPs and UVB caused increased oxidative DNA damage and cell death, indicated by immunostaining for 8-OHdG and TUNEL assay in sections of exposed mouse skin. Thus, collectively, our findings suggest that UVB exposure increases ZnO-NPs-mediated oxidative stress and oxidative damage, thereby enhancing ZnO-NPs-induced cell death.


Assuntos
Apoptose , Dano ao DNA , Nanopartículas Metálicas/toxicidade , Pele/efeitos dos fármacos , Raios Ultravioleta/efeitos adversos , Óxido de Zinco/toxicidade , Animais , Células Cultivadas , Feminino , Queratinócitos/efeitos dos fármacos , Queratinócitos/efeitos da radiação , Nanopartículas Metálicas/efeitos adversos , Nanopartículas Metálicas/química , Camundongos , Estresse Oxidativo , Pele/efeitos da radiação , Protetores Solares/efeitos adversos , Protetores Solares/química
5.
Food Chem Toxicol ; 96: 183-90, 2016 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-27507225

RESUMO

This study was conducted to explore the role of UVB on benzanthrone (BA)-induced skin inflammation and its mechanism/s. SKH-1 hairless mice were topically exposed with BA (25 and 50 mg/kg b.wt) either alone or along with UVB (50 mJ/cm(2)) for 24 h and estimation of ROS, histopathological analysis, myeloperoxidase (MPO) activity, mast cell staining, immunohistochemistry for COX-2 and iNOS as well as western blotting for MAPKs, p-NF-κB, c-jun, c-fos COX-2 and iNOS were carried out. Enhanced ROS generation, increased epidermal thickness, mast cell number, MPO activity, enhanced expression of COX-2 and iNOS, MAPKs, c-jun, c-fos, NF-κB were found in BA either alone or when followed by UVB treatment, compared to the control groups. Expression of COX-2, iNOS and phosphorylation of ERK1/2 were found to be more enhanced in BA and UVB- exposed group compared to BA and UVB only group, while phosphorylation of JNK1/2, p38, NF-κB and expression of c-jun and c-fos were comparable with BA and UVB only groups. In summary, we suggest that UVB exposure enhanced BA-induced SKH-1 skin inflammation possibly via oxidative stress-mediated activation of MAPKs-NF-κB/AP-1 signalling, which subsequently increased the expression of COX-2 and iNOS and led to inflammation in SKH-1 mouse skin.


Assuntos
Benzo(a)Antracenos/toxicidade , Ciclo-Oxigenase 2/metabolismo , Inflamação/etiologia , Proteínas Quinases Ativadas por Mitógeno/metabolismo , NF-kappa B/metabolismo , Óxido Nítrico Sintase Tipo II/metabolismo , Fator de Transcrição AP-1/metabolismo , Raios Ultravioleta/efeitos adversos , Animais , Feminino , Immunoblotting , Técnicas Imunoenzimáticas , Inflamação/metabolismo , Inflamação/patologia , Peroxidação de Lipídeos/efeitos dos fármacos , Camundongos , Camundongos Pelados , Estresse Oxidativo/efeitos dos fármacos , Fosforilação/efeitos dos fármacos , Espécies Reativas de Oxigênio/metabolismo , Transdução de Sinais/efeitos dos fármacos
6.
Mol Carcinog ; 55(8): 1262-74, 2016 08.
Artigo em Inglês | MEDLINE | ID: mdl-26259065

RESUMO

Colon cancer is the third most common cause of death in the United States. Therefore, new preventive strategies are warranted for preventing colon cancer. Nexrutine (NX), an herbal extract from Phellodendron amurense, has been shown to have anti-inflammatory, anti-microbial and anti-cancer activity for various tissue specific cancers, but its chemopreventive efficacy has not been evaluated against colon cancer. Here, we explored the mechanism of chemopreventive/chemotherapeutic efficacy of NX against colon cancer. We found that dietary exposure of NX significantly reduced the number of azoxymethane (AOM)-induced aberrant crypt foci (ACF) in rats. In addition, significant inhibition in AOM-induced cell proliferation and reduced expression of the inflammatory markers COX-2, iNOS as well as the proliferative markers PCNA and cyclin D1 were also seen. Moreover, NX exposure significantly enhanced apoptosis in the colon of AOM treated rats. Furthermore, in in vitro studies, NX (2.5, 5, 10 µg/ml, 48 h) decreased cell survival and colony formation while inducing G0/G1 cell cycle arrest and apoptosis in colon adenocarcinoma cells COLO205 and HCT-15. However, NX had minimal cytotoxic effect on IEC-6 normal rat intestinal cells, suggesting its high therapeutic index. NX treatment also modulates the level of Bax and Bcl-2 proteins along with cytochrome c release, cleavage and enhanced expression of poly (adenosine diphosphate-ribose) polymerase as well as the catalytic activity of caspase 3 and caspase 9 in both COLO205 and HCT-15 cells. Based on these in vivo and in vitro findings, we suggest that NX could be useful candidate agent for colon cancer chemoprevention and treatment. © 2015 Wiley Periodicals, Inc.


Assuntos
Focos de Criptas Aberrantes/dietoterapia , Azoximetano/toxicidade , Biomarcadores Tumorais/metabolismo , Neoplasias do Colo/dietoterapia , Extratos Vegetais/administração & dosagem , Focos de Criptas Aberrantes/induzido quimicamente , Focos de Criptas Aberrantes/metabolismo , Animais , Apoptose , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Neoplasias do Colo/induzido quimicamente , Neoplasias do Colo/metabolismo , Ciclina D1/metabolismo , Ciclo-Oxigenase 2/metabolismo , Regulação Neoplásica da Expressão Gênica , Humanos , Masculino , Óxido Nítrico Sintase Tipo II/metabolismo , Extratos Vegetais/farmacologia , Ratos , Ensaios Antitumorais Modelo de Xenoenxerto
7.
Toxicol Res (Camb) ; 5(4): 1066-1077, 2016 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-30090412

RESUMO

Besides titanium dioxide (TiO2), zinc oxide (ZnO) nanoparticles (NPs) are commonly used in sunscreen formulations as protective agents against exposure to ultraviolet radiation. Although the majority of prior studies have concluded that NPs do not penetrate healthy skin, compromised skin slightly enhanced metal oxide NP penetration. However, a question arises regarding the possible toxic consequences if consumers who had applied sunscreens containing ZnO-NPs were exposed to environmentally relevant doses of UVB. Considering this, we planned a study where SKH-1 hairless mice were topically exposed to a 5% and/or 10% dose of ZnO-NPs (<50 nm and <100 nm) either alone or along with UVB (50 mJ cm-2). In two additional groups, mice were treated with either bulk ZnO-NPs (<5 µm) or with ZnO-NPs (<5 µm) and subsequently UVB (50 mJ cm-2). Animals of all groups were sacrificed after 6, 24 and 48 h and the Zn ion content in the skin was measured. In addition, estimation of ROS generation, histopathology, myeloperoxidase (MPO) activity, immunohistochemistry for COX-2 and western blot analysis for MAPKs, p-IκBα, p-NF-κB, and COX-2 were also carried out. Significant increases in the Zn ion in exposed skin were seen. Enhanced ROS generation and MPO activity were also found in ZnO-NPs followed by UVB exposed groups at all three time points. Similarly, hyperplasia and over-expression of COX-2 were also greater in ZnO-NPs and UVB exposed groups than in the ZnO-NPs and UVB only groups. The expression of MAPKs, and transcription factors NF-κB along with COX-2 were also enhanced significantly in ZnO-NPs and the UVB treated group. Collectively, our findings suggest that UVB exposure enhanced ZnO-NP penetration in mouse skin and possibly dissolution of these ZnO-NPs takes place during this process, causing significant Zn ion generation leading to oxidative stress by ROS generation which subsequently activates MAPK-NF-κB signaling and increases COX-2 and inflammation.

8.
Toxicol Rep ; 2: 1-11, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-28962332

RESUMO

Epidemiological studies suggested that plant-based dietary supplements can reduce the risk of liver cancer. Nexrutine (NX), an herbal extract from Phellodendronamurense, has been shown to have anti-inflammatory, anti-microbial and anti-tumor activities. In the present study, we have shown the anti-tumor potential of NX against Solt-Farber model with elimination of PH, rat liver tumor induced by diethylnitrosoamine (DEN) as carcinogen and 2-acetylaminofluorene (2-AAF) as co-carcinogen. The elucidation of mechanistic pathways was explored in human liver cancer cells. Dietary intake of NX significantly decreased the cell proliferation and inflammation, as well as increased apoptosis in the liver sections of DEN/2-AAF-treated rats. Moreover, NX (2.5-10 µg/ml) exposure significantly decreased the viability of liver cancer cells and modulated the levels of Bax and Bcl-2 proteins levels. NX treatment resulted in increased cytochrome-c release and cleavage of caspases 3 and 9. In addition, NX decreased the expression of CDK2, CDK4 and associated cyclins E1 and D1, while up-regulated the expression of p21, p27 and p53 expression. NX also enhanced phosphorylation of the mitogen-activated protein kinases (MAPKs) ERK1/2, p38 and JNK1/2. Collectively, these findings suggested that NX-mediated protection against DEN/2-AAF-induced liver tumorigenesis involves decrease in cell proliferation and enhancement in apoptotic cell death of liver cancer cells.

9.
Mol Carcinog ; 53(12): 988-98, 2014 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-23813870

RESUMO

Patulin (PAT), a present day major contaminant of commercial apple and apple products is reported to be carcinogenic, embryotoxic, and immunotoxic. While oral and inhalation are considered to be the most prevalent routes of exposure to this toxin, exposure through skin is now being extensively investigated. Our previous study showed that short-term dermal exposure to PAT resulted in toxicological injury to the skin, while long-term exposure induced skin tumorigenesis. In this study, we explore the mechanism involve in proliferation of mouse keratinocytes by PAT. Our study revealed that PAT rapidly induces phosphorylation of EGFR, activation of the Ras/MAPKs, and Akt pathways. This in-turn leads to the activation of NF-κB/AP-1 transcription factors which then binds to the promoter region of the cell growth regulatory genes Cyclin D1 and COX-2 inducing their expression leading ultimately to PMKs proliferation. Inhibition of EGFR or the Ras/MAPKs, PI3/Akt pathways with different pharmacological inhibitors or knockdown of NF-κB, c-jun, c-fos, Cyclin D1, and COX-2 with siRNA inhibited PAT-induced PMKs proliferation.


Assuntos
Proliferação de Células/genética , Ciclina D1/genética , Ciclo-Oxigenase 2/genética , Receptores ErbB/genética , Queratinócitos/metabolismo , Proteínas Quinases Ativadas por Mitógeno/genética , Proteínas Proto-Oncogênicas c-akt/genética , Animais , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , Queratinócitos/efeitos dos fármacos , Camundongos , NF-kappa B/genética , Patulina/efeitos adversos , Fosfatidilinositol 3-Quinases/genética , Fosforilação/efeitos dos fármacos , Fosforilação/genética , Regiões Promotoras Genéticas/efeitos dos fármacos , Regiões Promotoras Genéticas/genética , Proteínas Proto-Oncogênicas c-fos/genética , Proteínas Proto-Oncogênicas c-jun/genética , Transdução de Sinais/genética , Fator de Transcrição AP-1/genética , Proteínas ras/genética
10.
Nutr Cancer ; 65 Suppl 1: 78-87, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23682786

RESUMO

Since bioantioxidants in plasma of Epidemic Dropsy patients [a condition caused by consumption of adulterated mustard oil with argemone oil (AO)] were found to be significantly decreased, the beneficial effect of N-acetyl cysteine (NAC) and α-tocopherol (TOCO) against AO- or sanguinarine (SANG)-induced tumorigenicity was undertaken in mice. Topical application of TOCO and NAC either alone or in combination showed significant protection against AO/TPA- and SANG/TPA-induced skin tumorigenicity. Histopathological findings suggest that papillomatous growth in AO/TPA- and SANG/TPA-treated animals were substantially protected following topical application of TOCO or NAC. Further, treatment of TOCO and NAC either alone or in combination to AO/TPA- or SANG/TPA-induced mice significantly decreased lipid peroxidation, along with significant revival in glutathione (GSH) content and activities of tyrosinase, histidase, catalase, SOD, GSH peroxidase, and GSH reductase in skin. In vitro studies showed that TOCO and/or NAC significantly decreased the AO and SANG induced cell proliferation and activation of ERK, p38, JNK MAPKs and NF-κB signaling in HaCaT cells. In summary, TOCO and NAC may be useful in preventing the tumorigenic response of AO and SANG probably by acting as scavenger of free radicals and inhibiting MAPKs and NF-κB signaling.


Assuntos
Acetilcisteína/farmacologia , Carcinogênese/efeitos dos fármacos , Mostardeira/efeitos adversos , Óleos de Plantas/efeitos adversos , alfa-Tocoferol/farmacologia , Animais , Antioxidantes/farmacologia , Benzofenantridinas/efeitos adversos , Carcinogênese/induzido quimicamente , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , Feminino , Glutationa/análise , Glutationa/metabolismo , Humanos , Isoquinolinas/efeitos adversos , Peroxidação de Lipídeos/efeitos dos fármacos , Sistema de Sinalização das MAP Quinases , Camundongos , NF-kappa B/genética , NF-kappa B/metabolismo , Pele/efeitos dos fármacos , Pele/patologia , Proteínas Quinases p38 Ativadas por Mitógeno/genética , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA