Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
Add more filters










Publication year range
1.
Environ Toxicol ; 2024 Apr 26.
Article in English | MEDLINE | ID: mdl-38666519

ABSTRACT

This study investigated the fabrication of gallic acid-loaded chitosan nanoparticles (Gal-Chi-NPs) that enhanced the DNA damage and apoptotic features by inhibiting FEN-1 expressions in MDA-MB 231 cells. Gal-Chi-NPs were fabricated by the ionic gelation method, and it was characterized by several studies such as dynamic light spectroscopy, Fourier-transforms infrared spectroscopy, x-ray diffraction, scanning electron microscopy, energy-dispersive x-ray, atomic force microscopy, and thermogravimetric analysis. We have obtained that Gal-Chi-NPs displayed 182.2 nm with crystal, smooth surface, and heat stability in nature. Gal-Chi-NPs induce significant toxicity in MDA-MB-231 cells that compared with normal NIH-3T3 cells. A significant reactive oxygen species (ROS) overproduction was observed in Gal-Chi-NPs treated MDA-MB-231. Flap endonuclease-1 (FEN-1) is a crucial protein involved in long patch base excision repair that is involved in repairing the chemotherapeutic mediated DNA-damaged base. Therefore, inhibition of FEN-1 protein expression is a crucial target for enhancing chemotherapeutical efficacy. In this study, we have obtained that Gal-Chi-NPs treatment enhanced the DNA damage by observing increased p-H2AX, PARP1; and suppressed the expression of FEN-1 in MDA-MB-231 cells. Moreover, Gal-Chi-NPs inhibited the expression of tumor proliferating markers p-PI3K, AKT, cyclin-D1, PCNA, and BCL-2; induced proapoptotic proteins (Bax and caspase-3) in MDA-MB 231 cells. Thus, Gal-Chi-NPs induce DNA damage and apoptotic features and inhibit tumor proliferation by suppressing FEN-1 expression in triple-negative breast cancer cells.

2.
J Biochem Mol Toxicol ; 36(8): e23098, 2022 Aug.
Article in English | MEDLINE | ID: mdl-35608392

ABSTRACT

This study has explored the fact that geraniol prevents isoproterenol (ISO)-induced oxidative stress and inflammation-mediated myocardial infarction (MI) through enhanced expression of peroxiredoxin-1 (Prdx-1) in experimental animal models. The experimental strategies of MI were stimulated through the subcutaneous direction of ISO (85 mg/kg body weight) for 14 days. ISO-directed models showed elevated heart rate levels and cardiac markers (serum creatine kinase [CK], serum CK-myocardial band, serum C-reactive proteins, and plasma homocysteine); increased cardiac-troponins-T, and troponin-I levels in both serum and myocardium. Moreover, we perceived that a higher level of lipid peroxidation molecules (thiobarbituric acid reactive substances and lipid hydroperoxides) reduced the antioxidant enzyme levels in plasma and heart tissue of ISO-directed rats. However, geraniol treatment prevents ISO-directed enhancement of the heart rate, cardiac and lipid peroxidative genes; reverted the blood pressure, and antioxidant status in ISO-directed rats. Furthermore, gene expression results revealed that geraniol treatment inhibited the mitogen-activated protein kinase (MAPK) proteins, inflammatory responder (tumor necrosis factor-α, interleukin 6, nuclear factor-κB), and cardiac fibrotic proteins (matrix metalloproteinase-2[MMP-2], MMP-9) in ISO directed rats. Prdx-1 is an antioxidant response element, and it can regulate all the antioxidant proteins and it scavenges harmful radicals. Therefore, enhanced Prdx-1 expression is considered to have a pivotal role in preventing cardiac infarction. In this study, an elevated expression of Prdx1 was noticed in geraniol treated with ISO-directed rats. Hence, we concluded that geraniol is considered a potential phytodrug, and it prevents ISO-directed MAPKs, inflammation, and cardiac markers by enhancing the expression of Prdx1.


Subject(s)
Matrix Metalloproteinase 2 , Myocardial Infarction , Acyclic Monoterpenes , Animals , Antioxidants/pharmacology , Inflammation/chemically induced , Inflammation/drug therapy , Isoproterenol/toxicity , Lipid Peroxidation , Lipid Peroxides/metabolism , Matrix Metalloproteinase 2/metabolism , Models, Animal , Myocardial Infarction/chemically induced , Myocardial Infarction/drug therapy , Myocardium/metabolism , Oxidative Stress , Peroxiredoxins/metabolism , Rats , Rats, Wistar
3.
Oxid Med Cell Longev ; 2020: 5275178, 2020.
Article in English | MEDLINE | ID: mdl-33312336

ABSTRACT

In the present study, we investigated the potential of opuntiol, isolated from Opuntia ficus-indica, against UVA radiation-mediated inflammation and skin photoaging in experimental animals. The skin-shaved experimental mouse was subjected to UVA exposure at the dosage of 10 J/cm2 per day for ten consecutive days (cumulative UVA dose: 100 J/cm2). Opuntiol (50 mg/kg b.wt.) was topically applied one hour before each UVA exposure. UVA (100 J/cm2) exposure induces epidermal hyperplasia and collagen disarrangement which leads to the photoaging-associated molecular changes in the mouse skin. Opuntiol pretreatment prevented UVA-linked clinical macroscopic skin lesions and histological changes in the mouse skin. Further, opuntiol prevents UVA-linked dermal collagen fiber loss in the mouse skin. Short-term UVA radiation (100 J/cm2) activates MAPKs through AP-1 and NF-κB p65 transcriptional pathways and subsequently induces the expression of inflammatory proteins and matrix-degrading proteinases in the mouse skin. Interestingly, opuntiol pretreatment inhibited UVA-induced activation of iNOS, VEGF, TNF-α, and COX-2 proteins and consequent activation of MMP-2, MMP-9, and MMP-12 in the mouse skin. Moreover, opuntiol was found to prevent collagen I and III breakdown in UVA radiation-exposed mouse skin. Thus, opuntiol protects mouse skin from UVA radiation-associated photoaging responses through inhibiting inflammatory responses, MAPK activation, and degradation of matrix collagen molecules.


Subject(s)
Collagen/metabolism , Coumaric Acids/pharmacology , MAP Kinase Signaling System , Proteolysis , Skin Aging/drug effects , Ultraviolet Rays/adverse effects , Animals , Inflammation/metabolism , Inflammation/pathology , MAP Kinase Signaling System/drug effects , MAP Kinase Signaling System/radiation effects , Male , Mice , Proteolysis/drug effects , Proteolysis/radiation effects , Skin Aging/pathology
4.
J Environ Pathol Toxicol Oncol ; 39(2): 191-199, 2020.
Article in English | MEDLINE | ID: mdl-32749127

ABSTRACT

We have proven that naringin, a phytonutrient, diminishes oxidative damage and inflammatory responses by modulating PPAR-γ expressions in ultraviolet-B radiation (UVB)-induced NIH-3T3 cells. However, the role of naringin against DNA damage, photoaging, and apoptosis in NIH-3T3 cells has yet to be studied, necessitating investigation. We show that Naringin pretreatment significantly reduces UVB-induced alkaline DNA damage and potentially modulates NER gene (XPC, TFIIH, XPE, ERCC1, and GAPDH) expression, thereby augmenting DNA repair. We determined experimentally that naringin pretreatment prevents UVB-induced nuclear fragmentation in NIH-3T3 cells, as well as altering UVB-induced apoptotic marker (Bax, BCl-2, Caspase-9, and Caspase-3) expression in them. In addition, naringin pretreatment inhibits UVB-stimulated matrix metalloproteinase (MMP-2, MMP-9 and MMP-13) expression in these 3T3 cells. Therefore, we report that naringin can effectively avert UVB-mediated DNA damage, photoaging, and apoptosis in NIH-3T3 cells.


Subject(s)
DNA Damage/drug effects , DNA Repair/drug effects , Flavanones/pharmacology , Ultraviolet Rays/adverse effects , Animals , Apoptosis/drug effects , Apoptosis/physiology , Apoptosis/radiation effects , DNA Repair/radiation effects , Matrix Metalloproteinases/genetics , Matrix Metalloproteinases/metabolism , Mice , NIH 3T3 Cells , Radiation-Protective Agents/pharmacology , Skin Aging/drug effects
5.
Antioxidants (Basel) ; 9(1)2019 Dec 21.
Article in English | MEDLINE | ID: mdl-31877697

ABSTRACT

We synthesized the gold nanoparticles (AuNPs) using wedelolactone (WDL) and characterized them using UV-visible spectroscopy, fourier transform infrared spectroscopy (FTIR), X-ray diffraction (XRD), scanning electron microscopic (SEM), transmission electron microscopic (TEM), energy dispersive X-ray diffraction, and atomic force microscopic (AFM) studies. The electronic spectrum exhibited an absorption peak at 535 nm. The FT-IR results proved that WDL was stabilized on the surface of AuNPs by acting as a capping or reducing agent. The crystalline structure was affirmed by XRD pattern and the spherical shape of WDL-AuNPs was evidenced by SEM, TEM, and AFM. The synthesized WDL-AuNPS were evaluated for anti-diabetic activity in pancreatic RIN-5F cell lines. In vitro results showed that WDL-AuNPs did not only improve the insulin secretion affected by di-(2-ethylhexyl) phthalate (DEHP), but also the cell viability in RIN5F cells. WDL-AuNPs treatment modulates the pro-apoptotic proteins and anti-apoptotic proteins expression to prevent the cells undergoing apoptosis in DEHP-exposed RIN-5F cells. The exposure of DEHP causes an increase in ROS production and lipid peroxidation levels. The free radical scavenging and antioxidant properties of WDL-AuNPs increase the deleterious effect caused by DEHP. On the other side, WDL-AuNPs increase mRNA expressions of insulin-signaling proteins in RIN-5F cells. This study concludes that WDL-AuNPs can be successfully used to regulate the expression of Bcl-2 family proteins, reduce lipid peroxidation, and to improve the secretion of antioxidants and insulin through the GLUT2 pathway in RIN-5F cell lines.

6.
Photochem Photobiol Sci ; 18(11): 2816-2817, 2019 11 01.
Article in English | MEDLINE | ID: mdl-31647084

ABSTRACT

Correction for 'The preventive effect of linalool on acute and chronic UVB-mediated skin carcinogenesis in Swiss albino mice' by Srithar Gunaseelan, et al., Photochem. Photobiol. Sci., 2016, 15, 851-860.

7.
J Biochem Mol Toxicol ; 33(3): e22263, 2019 Mar.
Article in English | MEDLINE | ID: mdl-30512238

ABSTRACT

The present study, we investigate the preventive role of naringin, a dietary flavonoid, against ultraviolet-B (UVB) radiation (280-320 nm) induced oxidative damage and inflammatory responses in mouse embryonic fibroblast cell lines (NIH-3T3). In this study, 20 mJ/cm 2 of UVB radiation induces cell cytotoxicity, reactive oxygen species (ROS) generation, DNA damage, and antioxidants depletion in NIH-3T3 cells. Treatment with naringin (60 µM) prior UVB exposure prevented the cell cytotoxicity, ROS generation, DNA damage, and antioxidants depletion in NIH-3T3 cells. Furthermore, naringin prevents UVB-induced mitogen-activated protein kinase families and nuclear factor-κB (NF-κB)-mediated activation of inflammatory factors, that is TNF-α, IL-6, IL-10, and COX-2 in NIH-3T3 cells. Peroxisome proliferator-activated receptor γ (PPARγ) is an anti-inflammatory agent and it suppressed the UVB-mediated oxidative and inflammatory responses. In this study, naringin activates PPARγ and prevents inflammatory biomarkers in NIH-3T3 cells. Thus, naringin prevents UVB-mediated inflammation and oxidative damage in NIH-3T3 cells probably over controlling NF-κB expression and activation of PPARγ.


Subject(s)
Flavanones/pharmacology , Inflammation/drug therapy , Oxidative Stress/drug effects , PPAR gamma/metabolism , Ultraviolet Rays , Animals , Anti-Inflammatory Agents/pharmacology , DNA Damage , Mice , NF-kappa B/metabolism , NIH 3T3 Cells , Oxidative Stress/radiation effects , Signal Transduction
8.
Article in Chinese | WPRIM (Western Pacific) | ID: wpr-950363

ABSTRACT

To examine the effects of p-coumaric acid on ethanol-induced kidney injury in Swiss Wistar rats. Methods: Ethanol (25% v/v) was used to induce nephrotoxicity in rats. p-Coumaric acid was orally administered at 50, 100, or 200 mg/kg body weight. The levels of oxidative parameters were determined; pro-inflammatory biomarkers were analyzed by Western blotting and apoptotic protein was analyzed by immunohistochemistry. Results: Ethanol treated rats showed decreased levels of antioxidants and aberrant production of pro-inflammatory cytokines (IL-6, IL1P, TNF-), NF- and kappa;B activation and imbalance of pro- and anti-apoptotic proteins (Bcl-2, Bax, caspase 3). Meanwhile, jp-coumaric acid restored antioxidant levels and decreased the levels of inflammatory cytokines, NF- and kappa;B, and pro- apoptotic proteins and increased Bcl-2 expression. Conclusions: jp-Coumaric acid ameliorates ethanol-induced kidney injury by restoring antioxidant production and suppressing cellular apoptosis and inhibiting NF- and kappa;B expression. jp-Coumaric acid should be further investigated as a promising candidate for ethanol-induced kidney toxicity.

9.
Toxicol Appl Pharmacol ; 352: 87-96, 2018 08 01.
Article in English | MEDLINE | ID: mdl-29802912

ABSTRACT

Previously, we proved that caffeic acid (CA), a major dietary phenolic acid, prevents skin carcinogenesis by modulating inflammatory signaling in mouse skin. However, the actual mechanisms of CA against UVB (280-320 nm) induced photocarcinogenesis remains unclear. The present results confirms that CA significantly inhibits single UVB-induced CPDs formation, oxidative DNA damage, ROS generation and frequency of apoptotic cell death in human dermal fibroblasts (HDFa). Furthermore, CA prevents UVB-induced expression of PI3K and AKT kinases through activation of PTEN which subsequently promotes XPC dependant NER proteins such as XPC, XPE, TFIIH (p44) and ERCC1 in HDFa cells and mouse skin tissue. Further, CA directly activates PTEN through hydrogen bond and hydrophobic interactions. Taken together, these findings suggest that CA prevents UVB-induced photodamage through the activation of PTEN expression in human dermal fibroblasts and mouse skin.


Subject(s)
Anticarcinogenic Agents/pharmacology , Caffeic Acids/pharmacology , Cell Transformation, Neoplastic/drug effects , Fibroblasts/drug effects , Neoplasms, Experimental/prevention & control , Neoplasms, Radiation-Induced/prevention & control , PTEN Phosphohydrolase/metabolism , Signal Transduction/drug effects , Skin Neoplasms/prevention & control , Skin/drug effects , Ultraviolet Rays/adverse effects , Animals , Apoptosis/drug effects , Cell Transformation, Neoplastic/metabolism , Cell Transformation, Neoplastic/pathology , Cell Transformation, Neoplastic/radiation effects , DNA Damage/drug effects , DNA Repair/drug effects , DNA Repair Enzymes/metabolism , Fibroblasts/enzymology , Fibroblasts/pathology , Fibroblasts/radiation effects , Humans , Mice , Neoplasms, Experimental/enzymology , Neoplasms, Experimental/pathology , Neoplasms, Radiation-Induced/enzymology , Neoplasms, Radiation-Induced/pathology , Oxidative Stress/drug effects , Oxidative Stress/radiation effects , Signal Transduction/radiation effects , Skin/enzymology , Skin/pathology , Skin/radiation effects , Skin Neoplasms/enzymology , Skin Neoplasms/pathology
10.
PLoS One ; 12(5): e0176699, 2017.
Article in English | MEDLINE | ID: mdl-28467450

ABSTRACT

Ultraviolet-B radiation (285-320 nm) elicits a number of cellular signaling elements. We investigated the preventive effect of linalool, a natural monoterpene, against UVB-induced oxidative imbalance, activation of mitogen-activated protein kinase (MAPK) and nuclear factor kappa-B (NF-κB) signaling in HDFa cells. We observed that linalool treatment (30 µM) prevented acute UVB-irradiation (20 mJ/cm2) mediated loss of activities of antioxidant enzymes in HDFa cells. The comet assay results illustrate that linalool significantly prevents UVB-mediated 8-deoxy guanosine formation (oxidative DNA damage) rather than UVB-induced cyclobutane pyrimidine (CPD) formation. This might be due to its ability to prevent UVB-induced ROS formation and to restore the oxidative imbalance of cells. This has been reflected in UVB-induced overexpression of MAPK and NF-κB signaling. We observed that linalool inhibited UVB-induced phosphorylation of ERK1, JNK and p38 proteins of MAPK family. Linalool inhibited UVB-induced activation of NF-κB/p65 by activating IκBa. We further observed that UVB-induced expression of TNF-α, IL6, IL-10, MMP-2 and MMP-9 was modulated by linalool treatment in HDFa cells. Thus, linalool protects the human skin cells from the oxidative damages of UVB radiation and modulates MAPK and NF-κB signaling in HDFa cells. The present findings substantiate that linalool may act as a photoprotective agent against UVB-induced skin damages.


Subject(s)
Mitogen-Activated Protein Kinases/drug effects , Monoterpenes/pharmacology , Oxidative Stress/radiation effects , Radiation-Protective Agents/pharmacology , Skin/radiation effects , Ultraviolet Rays/adverse effects , Acyclic Monoterpenes , Antioxidants/pharmacology , Apoptosis/drug effects , Apoptosis/radiation effects , DNA Damage/drug effects , DNA Damage/radiation effects , Humans , Mitogen-Activated Protein Kinases/radiation effects , NF-kappa B/drug effects , Oxidative Stress/drug effects , Signal Transduction/drug effects , Signal Transduction/radiation effects , Skin/drug effects , Skin/metabolism
11.
Photochem Photobiol Sci ; 15(7): 851-60, 2016 07 06.
Article in English | MEDLINE | ID: mdl-27251985

ABSTRACT

In this study, we evaluated the role of linalool in acute ultraviolet-B (UVB; 280-320 nm) radiation-induced inflammation and chronic UVB-mediated photocarcinogenesis in mouse skin. Acute UVB-irradiation (180 mJ cm(-2)) causes hyperplasia, edema formation, lipid peroxidation, antioxidant depletion, and overexpression of cyclooxygenase-2 (COX-2) and ornithine decarboxylase (ODC) in mouse skin. Topical or intraperitoneal (i.p.) treatment of linalool prevented acute UVB-induced hyperplasia, edema formation, lipid peroxidation, and antioxidant depletion in mouse skin. Further, linalool treatment prevented UVB-induced overexpression of COX-2 and ODC in mouse skin. In the chronic study, mice were subjected to UVB-exposure thrice weekly for 30 weeks. Chronic UVB-exposure induced tumor incidence and expression of proliferative markers such as NF-κB, TNF-α, IL-6, COX-2, VEGF, TGF-ß1, Bcl-2 and mutated p53 in mouse skin. Treatment with linalool before each UVB-exposure significantly prevented the expression of these proliferative markers and subsequently decreased the tumor incidence in mice skin. Histopathological studies confirmed the development of dysplasia and squamous cell carcinoma (SCC) in the chronic UVB-exposed mouse skin; and this was prevented by both topical and i.p. linalool treatment. Therefore, linalool may be considered as a photochemopreventive agent against UVB radiation induced skin carcinogenesis.


Subject(s)
Monoterpenes/pharmacology , Neoplasms, Radiation-Induced/prevention & control , Protective Agents/pharmacology , Skin/drug effects , Ultraviolet Rays , Acyclic Monoterpenes , Administration, Topical , Animals , Antioxidants/metabolism , Carcinogenesis/drug effects , Catalase/metabolism , Cyclooxygenase 2/metabolism , Immunohistochemistry , Injections, Intraperitoneal , Interleukin-6/metabolism , Lipid Peroxidation/drug effects , Lipid Peroxidation/radiation effects , Mice , Mice, Hairless , Monoterpenes/therapeutic use , Ornithine Decarboxylase/metabolism , Protective Agents/therapeutic use , Skin/pathology , Skin/radiation effects , Superoxide Dismutase/metabolism
12.
Eur J Pharmacol ; 786: 194-203, 2016 Sep 05.
Article in English | MEDLINE | ID: mdl-27262378

ABSTRACT

Multidrug resistance (MDR) remains a major obstacle in cancer chemotherapy. The use of the dietary phytochemicals as chemosensitizing agents to enhance the efficacy of conventional cytostatic drugs has recently gained the attention as a plausible approach for overcoming the drug resistance. The aim of this study was to investigate whether a naturally occurring diet-based phenolic acid, ferulic acid, could sensitize paclitaxel efficacy in ABCB1 overexpressing (P-glycoprotein) colchicine selected KB Ch(R)8-5 cell line. In vitro drug efflux assays demonstrated that ferulic acid inhibits P-glycoprotein transport function in drug resistant KB Ch(R)8-5 cell lines. However, ferulic acid significantly downregulates ABCB1 expression in a concentration dependent manner. Cytotoxicity assay reveals that ferulic acid decreased paclitaxel resistance in KBCh(R)8-5 and HEK293/ABCB1 cells, which indicates its chemosensitizing potential. Clonogenic cell survival assay and apoptotic morphological staining further confirm the chemosensitizing potential of ferulic acid in drug resistant KB Ch(R)8-5 cell lines. Ferulic acid treatment enhances paclitaxel mediated cell cycle arrest and upregulates paclitaxel-induced apoptotic signaling in KB resistant cells. Hence, it has been concluded that downregulation of ABCB1 and subsequent induction of paclitaxel-mediated cell cycle arrest and apoptotic signaling may be the cause for the chemosensitizing potential of ferulic acid in P-gp overexpressing cell lines.


Subject(s)
Coumaric Acids/pharmacology , Drug Resistance, Multiple/drug effects , Paclitaxel/pharmacology , ATP Binding Cassette Transporter, Subfamily B/chemistry , ATP Binding Cassette Transporter, Subfamily B/genetics , ATP Binding Cassette Transporter, Subfamily B/metabolism , Apoptosis/drug effects , Cell Movement/drug effects , Cell Survival/drug effects , Coumaric Acids/metabolism , Fluoresceins/metabolism , G2 Phase Cell Cycle Checkpoints/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Humans , KB Cells , M Phase Cell Cycle Checkpoints/drug effects , Molecular Docking Simulation , Protein Conformation
13.
Photochem Photobiol ; 91(6): 1458-68, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26303058

ABSTRACT

In this study, the effect of caffeic acid (CA) on both acute and chronic UVB-irradiation-induced inflammation and photocarcinogenesis was investigated in Swiss albino mice. Animals were exposed to 180 mJ cm(-2) of UVB once daily for 10 consecutive days and thrice weekly for 30 weeks for acute and chronic study respectively. UVB exposure for 10 consecutive days showed edema formation, increased lipid peroxidation and decreased antioxidant status with activation of inflammatory molecules such as TNF-α, IL-6, COX-2 and NF-κB. However, CA (15 mg per kg.b.wt.) administration before each UVB exposure decreased lipid peroxidation, inflammatory markers expression and enhanced antioxidant status probably through the activation of peroxisome proliferator-activated receptors (PPARγ) in the mice skin. PPARγ is considered a potential target for photochemoprevention because it inhibits UVB-mediated inflammatory responses. In this study, UVB exposure for 30 weeks caused squamous cell carcinoma and upregulation of iNOS, VEGF and TGF-ß and downregulation of p53 and tumor incidence in the mice skin. Both topical (CAT) and intraperitoneal (CAIP) treatment before each UVB exposure downregulates iNOS, VEGF, TGF-ß, upregulates p53 and reduces tumors multiplicity in the mice skin. Thus, CA offers protection against UVB-induced photocarcinogenesis probably through activation of anti-inflammatory transcription factor PPARγ in the mice.


Subject(s)
Caffeic Acids/pharmacology , Carcinogenesis/drug effects , PPAR gamma/metabolism , Skin/drug effects , Transcriptional Activation/drug effects , Ultraviolet Rays , Animals , Anti-Inflammatory Agents/pharmacology , Antioxidants/pharmacology , Female , Inflammation , Mice , Mice, Hairless
14.
Food Chem Toxicol ; 82: 72-8, 2015 Aug.
Article in English | MEDLINE | ID: mdl-25983265

ABSTRACT

The aim of this study was to evaluate the photochemopreventive effects of ferulic acid (FA) against chronic ultraviolet-B (290-320 nm) induced oxidative stress, inflammation and angiogenesis in the skin of Swiss albino mice. Chronic UVB exposure (180 mJ/cm(2) for 30 weeks; thrice in a week) induced tumor formation in the mice skin that showed increased expression of carcinogenic and inflammatory markers when compared with the control animals. The intraperitoneal (FAIP) and topical (FAT) administration of FA significantly reduced the incidence of UVB-induced tumor volume and tumor weight in the mice skin. Histopathological studies revealed that both FAIP and FAT administration prevented the UVB-induced hyperplasia, squamous cell carcinoma (SCC) and dysplastic feature in the mice skin. Further, it has been observed that FA treatment reverted chronic UVB-induced oxidative damage (thiobarbituric acid reactive substances, superoxide dismutase, catalase, glutathione peroxidase) accompanied with modulation of vascular endothelial growth factor (VEGF), inducible nitric oxide synthase (iNOS), TNF-α and IL-6 in the mice skin tumor. FA treatment also modulates mutated p53, Bcl-2 and Bax expressions in the UVB-induced mice skin tumor. Thus, the results of the present study indicate ferulic acid has potential against UVB-induced carcinogenesis in the Swiss albino mice.


Subject(s)
Apoptosis/drug effects , Carcinoma, Squamous Cell/prevention & control , Coumaric Acids/pharmacology , Skin Neoplasms/prevention & control , Animals , Anticarcinogenic Agents/pharmacology , Apoptosis/radiation effects , Carcinoma, Squamous Cell/metabolism , Inflammation/etiology , Inflammation/metabolism , Interleukin-6/metabolism , Male , Mice , Nitric Oxide Synthase Type II , Radiation-Protective Agents/pharmacology , Signal Transduction/drug effects , Signal Transduction/radiation effects , Skin Neoplasms/pathology , Tumor Necrosis Factor-alpha/metabolism , Ultraviolet Rays/adverse effects
SELECTION OF CITATIONS
SEARCH DETAIL
...