Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 30
Filter
1.
J Clin Invest ; 129(11): 4962-4978, 2019 11 01.
Article in English | MEDLINE | ID: mdl-31609245

ABSTRACT

Macrophages are important in mounting an innate immune response to injury as well as in repair of injury. Gene expression of Rho proteins is known to be increased in fibrotic models; however, the role of these proteins in idiopathic pulmonary fibrosis (IPF) is not known. Here, we show that BAL cells from patients with IPF have a profibrotic phenotype secondary to increased activation of the small GTPase Rac1. Rac1 activation requires a posttranslational modification, geranylgeranylation, of the C-terminal cysteine residue. We found that by supplying more substrate for geranylgeranylation, Rac1 activation was substantially increased, resulting in profibrotic polarization by increasing flux through the mevalonate pathway. The increased flux was secondary to greater levels of acetyl-CoA from metabolic reprogramming to ß oxidation. The polarization mediated fibrotic repair in the absence of injury by enhancing macrophage/fibroblast signaling. These observations suggest that targeting the mevalonate pathway may abrogate the role of macrophages in dysregulated fibrotic repair.


Subject(s)
Idiopathic Pulmonary Fibrosis/metabolism , Macrophages/metabolism , Mevalonic Acid/metabolism , Acetyl Coenzyme A/genetics , Acetyl Coenzyme A/metabolism , Adolescent , Adult , Aged , Animals , Female , Humans , Idiopathic Pulmonary Fibrosis/genetics , Idiopathic Pulmonary Fibrosis/pathology , Macrophages/pathology , Male , Mice , Mice, Knockout , Middle Aged , Neuropeptides/genetics , Neuropeptides/metabolism , Oxidation-Reduction , rac1 GTP-Binding Protein/genetics , rac1 GTP-Binding Protein/metabolism
2.
Oncotarget ; 8(30): 49625-49636, 2017 Jul 25.
Article in English | MEDLINE | ID: mdl-28548949

ABSTRACT

Ultraviolet B (UVB) radiation induces regulatory T cells (Treg cells) and depletion of these Treg cells alleviates immunosuppression and inhibits photocarcinogenesis in mice. Here, we determined the effects of dietary grape seed proanthocyanidins (GSPs) on the development and activity of UVB-induced Treg cells. C3H/HeN mice fed a GSPs (0.5%, w/w)-supplemented or control diet were exposed to UVB (150 mJ/cm2) radiation, sensitized to 2,4-dinitrofluorobenzene (DNFB) and sacrificed 5 days later. FACS analysis indicated that dietary GSPs decrease the numbers of UVB-induced Treg cells. ELISA analysis of cultured sorted Treg cells indicated that secretion of immunosuppressive cytokines (interleukin-10, TGF-ß) was significantly lower in Treg cells from GSPs-fed mice. Dietary GSPs also enhanced the ability of Treg cells from wild-type mice to stimulate production of IFNγ by T cells. These effects of dietary GSPs on Treg cell function were not found in XPA-deficient mice, which are incapable of repairing UVB-induced DNA damage. Adoptive transfer experiments revealed that naïve recipients that received Treg cells from GSPs-fed UVB-irradiated wild-type donors that had been sensitized to DNFB exhibited a significantly higher contact hypersensitivity (CHS) response to DNFB than mice that received Treg cells from UVB-exposed mice fed the control diet. There was no significant difference in the CHS response between mice that received Treg cells from UVB-irradiated XPA-deficient donors fed GSPs or the control diet. Furthermore, dietary GSPs significantly inhibited UVB-induced skin tumor development in wild-type mice but not in XPA-deficient mice. These results suggest that GSPs inactivate Treg cells by promoting DNA repair in dendritic cells in UVB-exposed skin.


Subject(s)
DNA End-Joining Repair/drug effects , Dendritic Cells/drug effects , Dendritic Cells/physiology , Grape Seed Extract/pharmacology , Proanthocyanidins/pharmacology , Skin Physiological Phenomena/radiation effects , Skin/radiation effects , T-Lymphocytes, Regulatory/drug effects , T-Lymphocytes, Regulatory/physiology , Adoptive Transfer , Animals , Biomarkers , Cytokines/metabolism , Immunomodulation , Immunophenotyping , Interferon-gamma/metabolism , Mice , Mice, Knockout , Skin Neoplasms/etiology , Skin Neoplasms/metabolism , Skin Neoplasms/pathology , Tumor Microenvironment , Ultraviolet Rays
3.
Int J Oncol ; 48(2): 624-34, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26676402

ABSTRACT

Melanoma is a highly aggressive form of skin cancer with poor survival rate. Aberrant activation of Wnt/ß-catenin has been observed in nearly one-third of human melanoma cases thereby indicating that targeting Wnt/ß-catenin signaling could be a promising strategy against melanoma development. In the present study, we determined chemotherapeutic effect of grape seed proanthocyanidins (GSPs) on the growth of melanoma cells and validated their protective effects in vivo using a xenograft mouse model, and assessed if ß-catenin is the target of GSP chemotherapeutic effect. Our in vitro data show that treatment of A375 and Hs294t human melanoma cells with GSPs inhibit the growth of melanoma cells, which was associated with the reduction in the levels of ß-catenin. Administration of dietary GSPs (0.2 and 0.5%, w/w) in supplementation with AIN76A control diet significantly inhibited the growth of melanoma tumor xenografts in nude mice. Furthermore, dietary GSPs inhibited the xenograft growth of Mel928 (ß-catenin-activated), while did not inhibit the xenograft growth of Mel1011 (ß-catenin-inactivated) cells. These observations were further verified by siRNA knockdown of ß-catenin and forced overexpression of ß-catenin in melanoma cells using a cell culture model.


Subject(s)
Apoptosis/drug effects , Cell Proliferation/drug effects , Grape Seed Extract/pharmacology , Melanoma/drug therapy , Melanoma/metabolism , Proanthocyanidins/pharmacology , beta Catenin/metabolism , Animals , Cell Line, Tumor , Female , Humans , Mice , Mice, Nude , Xenograft Model Antitumor Assays/methods
4.
Biomark Cancer ; 7(Suppl 1): 29-37, 2015.
Article in English | MEDLINE | ID: mdl-26568685

ABSTRACT

Colon cancer development and malignant progression are driven by genetic and epigenetic alterations in tumor cells and by factors from the tumor microenvironment. Cancer cells become reliant on the activity of specific oncogenes and on prosurvival and proliferative signals they receive from the abnormal environment they create and reside in. Accordingly, the response to anticancer therapy is determined by genetic and epigenetic changes that are intrinsic to tumor cells and by the factors present in the tumor microenvironment. Recent advances in the understanding of the involvement of the tumor microenvironment in tumor progression and therapeutic response are optimizing the application of prognostic and predictive factors in colon cancer. Moreover, new targets in the tumor microenvironment that are amenable to therapeutic intervention have been identified. Because stromal cells are with rare exceptions genetically stable, the tumor microenvironment has emerged as a preferred target for therapeutic drugs. In this review, we discuss the role of stromal fibroblasts and macrophages in colon cancer progression and in the response of colon cancer patients to therapy.

5.
Am J Cancer Res ; 5(11): 3325-38, 2015.
Article in English | MEDLINE | ID: mdl-26807314

ABSTRACT

Melanoma is a highly aggressive form of skin cancer and a leading cause of death from skin diseases mainly due to its propensity to metastasis. Due to metastatic tendency, melanoma is often associated with activation of Wnt/ß-catenin signaling mechanism. Blocking ß-catenin activation may be a good strategy to block melanoma-associated mortality. We have shown earlier that grape seed proanthocyanidins (GSPs) inhibit melanoma cell migration via targeting cyclooxygenase-2 (COX-2) overexpression. Here we explored further whether inhibition of inflammatory mediators-mediated activation of ß-catenin by GSPs is associated with the inhibition of melanoma cell migration. Our study revealed that PGE2 receptors (EP2 and EP4) agonists promote melanoma cell migration while PGE2 receptor antagonist suppressed the migration capacity of melanoma cells. GSPs treatment inhibit butaprost (EP2 agonist) or Cay10580 (EP4 agonist) induced migration of melanoma cells. Western blot analysis revealed that GSPs reduced cellular accumulation of ß-catenin, and decreased the expressions of matrix metalloproteinase (MMP)-2, MMP-9 and MITF, downstream targets of ß-catenin in melanoma cells. GSPs also reduced the protein expressions of PI3K and p-Akt in the same set of experiment. To verify that ß-catenin is a specific molecular target of GSPs, we compared the effect of GSPs on cell migration of ß-catenin-activated (Mel1241) and ß-catenin-inactivated (Mel1011) melanoma cells. GSPs inhibit cell migration of Mel1241 cells but not of Mel1011 cells. Additionally, in vivo bioluminescence imaging data indicate that dietary administration of GSPs (0.5%, w/w) in supplementation with AIN76A control diet inhibited the migration/extravasation of intravenously injected melanoma cells in lungs of immune-compromised nude mice, and that this effect of GSPs was associated with an inhibitory effect on the activation of ß-catenin and its downstream targets, such as MMPs, in lungs as a target organ.

6.
Am J Physiol Lung Cell Mol Physiol ; 308(1): L58-75, 2015 Jan 01.
Article in English | MEDLINE | ID: mdl-25326576

ABSTRACT

Surfactant protein A (SP-A), a molecule with roles in lung innate immunity and surfactant-related functions, is encoded by two genes in humans: SFTPA1 (SP-A1) and SFTPA2 (SP-A2). The mRNAs from these genes differ in their 5'-untranslated regions (5'-UTR) due to differential splicing. The 5'-UTR variant ACD' is exclusively found in transcripts of SP-A1, but not in those of SP-A2. Its unique exon C contains two upstream AUG codons (uAUGs) that may affect SP-A1 translation efficiency. The first uAUG (u1) is in frame with the primary start codon (p), but the second one (u2) is not. The purpose of this study was to assess the impact of uAUGs on SP-A1 expression. We employed RT-qPCR to determine the presence of exon C-containing SP-A1 transcripts in human RNA samples. We also used in vitro techniques including mutagenesis, reporter assays, and toeprinting analysis, as well as in silico analyses to determine the role of uAUGs. Exon C-containing mRNA is present in most human lung tissue samples and its expression can, under certain conditions, be regulated by factors such as dexamethasone or endotoxin. Mutating uAUGs resulted in increased luciferase activity. The mature protein size was not affected by the uAUGs, as shown by a combination of toeprint and in silico analysis for Kozak sequence, secondary structure, and signal peptide and in vitro translation in the presence of microsomes. In conclusion, alternative splicing may introduce uAUGs in SP-A1 transcripts, which in turn negatively affect SP-A1 translation, possibly affecting SP-A1/SP-A2 ratio, with potential for clinical implication.


Subject(s)
Alternative Splicing/physiology , Codon, Initiator/physiology , Lung/metabolism , Pulmonary Surfactant-Associated Protein A/biosynthesis , RNA, Messenger/biosynthesis , Alternative Splicing/drug effects , Anti-Inflammatory Agents/pharmacology , Cell Line, Tumor , Dexamethasone/pharmacology , Endotoxins/pharmacology , Exons/physiology , Female , Humans , Male , Microsomes/metabolism , Mutation , Protein Sorting Signals/physiology , Pulmonary Surfactant-Associated Protein A/genetics , RNA, Messenger/genetics
7.
Mol Carcinog ; 54 Suppl 1: E61-71, 2015 Jun.
Article in English | MEDLINE | ID: mdl-24798688

ABSTRACT

Cigarette smoking is the major cause of lung cancer. It is therefore important to develop effective strategies that target molecular abnormalities induced by cigarette smoke condensate (CSC). Cigarette smoking increases oxidative stress particularly via activation of NADPH oxidase (NOX), a key source of superoxide anion production. Here, we report that grape seed proanthocyanidins (GSPs) exert an inhibitory effect on the CSC-induced migration of non-small cell lung cancer (NSCLC) cells (A549, H460, and H1299). Using an in vitro invasion assay, we found that treatment of NSCLC cells with CSC increased NSCLC cell migration by enhancing NOX mediated-oxidative stress. Treatment of NSCLC cells with GSPs inhibited the CSC-induced cell migration through reduction in oxidative stress levels and a reduction in the epithelial-to-mesenchymal transition. To identify the molecular targets of GSPs, we examined the effects of GSPs on CSC-induced alterations in the levels of key NOX components, namely p22(phox) and p47(phox) proteins, using A549 cells. We also determined the effect of GSPs on CSC-induced interaction/binding between these proteins, which is a key event in NOX activation. We found that treatment of A549 cells with GSPs not only inhibited the CSC-induced increase in the expression levels of p22(phox) and p47(phox) , but also reduced the binding of p22(phox) to p47(phox) proteins. This new insight into the anti-lung cancer cell migration activity of GSPs could serve as a basis for development of improved chemopreventive or therapeutic strategies for lung cancer.


Subject(s)
Carcinoma, Non-Small-Cell Lung/pathology , Grape Seed Extract/pharmacology , Lung Neoplasms/pathology , NADPH Oxidases/antagonists & inhibitors , Neoplasm Metastasis/prevention & control , Proanthocyanidins/pharmacology , Smoke/adverse effects , Carcinoma, Non-Small-Cell Lung/enzymology , Carcinoma, Non-Small-Cell Lung/metabolism , Cell Line, Tumor , Cell Movement/drug effects , Enzyme Activation , Humans , Lung Neoplasms/enzymology , Lung Neoplasms/metabolism , NADPH Oxidases/metabolism , Oxidative Stress , Protein Binding , Nicotiana
8.
Mol Carcinog ; 54(11): 1328-39, 2015 Nov.
Article in English | MEDLINE | ID: mdl-25174976

ABSTRACT

Cutaneous malignant melanoma is the leading cause of death from skin diseases and is often associated with activating mutations of the proto-oncogene BRAF. To develop more effective strategies for the prevention or treatment of melanoma, we have examined the inhibitory effects of silymarin, a flavanoid from Silybum marianum, on melanoma cells. Using A375 (BRAF-mutated) and Hs294t (non BRAF-mutated but highly metastatic) human melanoma cell lines, we found that in vitro treatment with silymarin resulted in a dose-dependent: (i) reduction in cell viability; (ii) enhancement of either Go/G1 (A375) or G2-M (Hs294t) phase cell cycle arrest with corresponding alterations in cyclins and cyclin-dependent kinases; and (iii) induction of apoptosis. The silymarin-induced apoptosis of human melanoma cells was associated with a reduction in the levels of anti-apoptotic proteins (Bcl-2 and Bcl-xl), an increase in the levels of pro-apoptotic protein (Bax), and activation of caspases. Further, oral administration of silymarin (500 mg/kg body weight/2× a week) significantly inhibited (60%, P < 0.01) the growth of BRAF-mutated A375 melanoma tumor xenografts, and this was associated with: (i) inhibition of cell proliferation; (ii) induction of apoptosis of tumor cells; (iii) alterations in cell cycle regulatory proteins; and (iv) reduced expression of tumor angiogenic biomarkers in tumor xenograft tissues. These results indicate that silymarin may have a chemotherapeutic effect on human melanoma cell growth and warrant its further evaluation.


Subject(s)
Apoptosis/drug effects , Biomarkers, Tumor/genetics , Cell Cycle Checkpoints/drug effects , Cell Proliferation/drug effects , Melanoma/drug therapy , Neovascularization, Pathologic/genetics , Silymarin/pharmacology , Animals , Caspases/genetics , Cell Cycle Proteins/genetics , Cell Line, Tumor , Female , Humans , Melanoma/genetics , Mice , Mice, Nude , Proto-Oncogene Mas , bcl-2-Associated X Protein/genetics , bcl-X Protein/genetics
9.
Toxicol Appl Pharmacol ; 274(1): 147-55, 2014 Jan 01.
Article in English | MEDLINE | ID: mdl-24211275

ABSTRACT

Previously, we showed that administration of a high-fat diet (HF-diet) to C57BL/6 mice exacerbates their response to short-term UVB radiation-induced inflammation in the skin. To explore the effects of an HF-diet on UVB-induced tumorigenesis, we have used the SKH-1 hairless mouse model in which the mice are exposed to UVB radiation (180mJ/cm(2)) three times a week for 24weeks. The development of UVB-induced skin tumors was rapid and the tumor multiplicity and tumor size were significantly higher (P<0.01-0.005) in the mice fed an HF-diet than the mice fed a control-diet (C-diet). Moreover, the malignant progression of UVB-induced papillomas to carcinomas was higher in HF-diet-fed mice. On analysis of tumors and tumor-uninvolved skin samples from the tumor-bearing mice, we found that administration of an HF-diet significantly enhanced the levels of UVB-induced expression of cyclooxygenase-2 (COX-2), prostaglandin E2 (P<0.01), and PGE2 receptors, and activation of NF-κB in the UVB-exposed skin as well as in tumors. In addition the HF-diet enhanced the expression of proinflammatory cytokines, including tumor necrosis factor-α (P<0.01), interleukin (IL)-1ß (P<0.01) and IL-6 (P<0.05) in the UVB-exposed skin as well as in tumors. Western blot analysis revealed that HF-diet enhanced the levels of epidermal cell proliferation, phosphatidylinositol 3-kinase and phosphorylation of Akt at Ser(473) in UVB-exposed skin and skin tumors. Collectively, these data demonstrate that the regular consumption of an HF-diet increases the risk of photocarcinogenesis in mice and that this is associated with enhanced expression of inflammatory mediators in the UVB-exposed skin and tumors.


Subject(s)
Diet, High-Fat/adverse effects , Disease Progression , Neoplasms, Radiation-Induced/pathology , Papilloma/pathology , Skin Neoplasms/pathology , Animals , Female , Mice , Mice, Hairless , Neoplasms, Radiation-Induced/metabolism , Papilloma/metabolism , Risk Factors , Skin Neoplasms/metabolism , Ultraviolet Rays/adverse effects
10.
Biochem Pharmacol ; 85(8): 1066-76, 2013 Apr 15.
Article in English | MEDLINE | ID: mdl-23395695

ABSTRACT

Silymarin inhibits UVB-induced immunosuppression in mouse skin. To identify the molecular mechanisms underlying this effect, we used an adoptive transfer approach in which dendritic cells (DCs) from the draining lymph nodes of donor mice that had been UVB-exposed and sensitized to 2,4,-dinitrofluorobenzene (DNFB) were transferred into naïve recipient mice. The contact hypersensitivity (CHS) response of the recipient mice to DNFB was then measured. When DCs were obtained from UVB-exposed donor mice that were not treated with silymarin, the CHS response was suppressed confirming the role of DCs in the UVB-induced immunosuppression. Silymarin treatment of UVB-exposed donor mice relieved this suppression of the CHS response in the recipients. Silymarin treatment was associated with rapid repair of UVB-induced cyclobutane pyrimidine dimers (CPDs) in DCs and silymarin treatment did not prevent UV-induced immunosuppression in XPA-deficient mice which are unable to repair UV-induced DNA damage. The CHS response in mice receiving DCs from silymarin-treated UV-exposed donor mice also was associated with enhanced secretion of Th1-type cytokines and stimulation of T cells. Adoptive transfer of T cells revealed that transfer of either CD8(+) or CD4(+) cells from silymarin-treated, UVB-exposed donors resulted in enhancement of the CHS response. Cell culture study showed enhanced secretion of IL-2 and IFNγ by CD8(+) T cells, and reduced secretion of Th2 cytokines by CD4(+) T cells, obtained from silymarin-treated UVB-exposed mice. These data suggest that DNA repair-dependent functional activation of DCs, a reduction in CD4(+) regulatory T-cell activity, and stimulation of CD8(+) effector T cells contribute to silymarin-mediated inhibition of UVB-induced immunosuppression.


Subject(s)
DNA Repair , Dendritic Cells/drug effects , Immune Tolerance/drug effects , Lymphocyte Activation/drug effects , Silymarin/pharmacology , T-Lymphocytes/drug effects , Ultraviolet Rays/adverse effects , Adoptive Transfer , Animals , Dendritic Cells/immunology , Dermatitis, Contact/prevention & control , Dinitrofluorobenzene , Female , Mice , Mice, Inbred C3H , T-Lymphocytes/immunology
11.
Cancer Prev Res (Phila) ; 6(3): 242-52, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23321928

ABSTRACT

Ultraviolet (UV) radiation-induced immunosuppression has been implicated in skin carcinogenesis. Grape seed proanthocyanidins (GSPs) have anti-skin carcinogenic effects in mice and GSPs-fed mice exhibit a reduction in UV-induced suppression of allergic contact hypersensitivity (CHS), a prototypic T-cell-mediated response. Here, we report that dietary GSPs did not inhibit UVB-induced suppression of CHS in xeroderma pigmentosum complementation group A (XPA)-deficient mice, which lack nucleotide excision repair mechanisms. GSPs enhanced repair of UVB-induced DNA damage (cyclobutane pyrimidine dimers) in wild-type, but not XPA-deficient, dendritic cells (DC). Co-culture of CD4(+) T cells with DCs from UVB-irradiated wild-type mice resulted in suppression of T-cell proliferation and secretion of T-helper (TH) 1-type cytokines that was ameliorated when the DCs were obtained from GSP-fed mice, whereas DCs obtained from GSP-fed XPA-KO mice failed to restore T-cell proliferation. In adoptive transfer experiments, donor DCs were positively selected from the draining lymph nodes of UVB-exposed donor mice that were sensitized to 2,4,-dinitrofluorobenzene were transferred into naïve recipient mice and the CHS response assessed. Naïve recipients that received DCs from UVB-exposed wild-type donors that had been fed GSPs exhibited a full CHS response, whereas no significant CHS was observed in mice that received DCs from XPA-KO mice fed GSPs. These results suggest that GSPs prevent UVB-induced immunosuppression through DNA repair-dependent functional activation of dendritic cells in mice. Cancer Prev Res; 6(3); 242-52. ©2013 AACR.


Subject(s)
Dendritic Cells/drug effects , Grape Seed Extract/pharmacology , Proanthocyanidins/pharmacology , Skin/radiation effects , Ultraviolet Rays/adverse effects , Animals , Blotting, Southern , Blotting, Western , DNA Damage/drug effects , Dendritic Cells/immunology , Dermatitis, Contact/immunology , Female , Immune System/drug effects , Immune System/radiation effects , Immune Tolerance/drug effects , Immune Tolerance/immunology , Mice , Mice, Knockout , Skin/drug effects , Skin/immunology , Vitis/chemistry
12.
PLoS One ; 7(8): e43064, 2012.
Article in English | MEDLINE | ID: mdl-22905202

ABSTRACT

Pancreatic cancer is an aggressive malignancy that is frequently diagnosed at an advanced stage with poor prognosis. Here, we report the chemotherapeutic effects of bioactive proanthocyanidins from grape seeds (GSPs) as assessed using In Vitro and In Vivo models. Treatment of human pancreatic cancer cells (Miapaca-2, PANC-1 and AsPC-1) with GSPs In Vitro reduced cell viability and increased G2/M phase arrest of the cell cycle leading to induction of apoptosis in a dose- and time-dependent manner. The GSPs-induced apoptosis of pancreatic cancer cells was associated with a decrease in the levels of Bcl-2 and Bcl-xl and an increase in the levels of Bax and activated caspase-3. Treatment of Miapaca-2 and PANC-1 cells with GSPs also decreased the levels of phosphatidylinositol-3-kinase (PI3K) and phosphorylation of Akt at ser(473). siRNA knockdown of PI3K from pancreatic cancer cells also reduced the phosphorylation of Akt. Further, dietary administration of GSPs (0.5%, w/w) as a supplemented AIN76A control diet significantly inhibited the growth of Miapaca-2 pancreatic tumor xenografts grown subcutaneously in athymic nude mice, which was associated with: (i) inhibition of cell proliferation, (ii) induction of apoptosis of tumor cells, (iii) increased expression of Bax, reduced expression of anti-apoptotic proteins and activation of caspase-3-positive cells, and (iv) decreased expression of PI3K and p-Akt in tumor xenograft tissues. Together, these results suggest that GSPs may have a potential chemotherapeutic effect on pancreatic cancer cell growth.


Subject(s)
Apoptosis , Pancreatic Neoplasms/drug therapy , Phosphatidylinositol 3-Kinases/metabolism , Plant Extracts/pharmacology , Proanthocyanidins/pharmacology , Proto-Oncogene Proteins c-akt/metabolism , Seeds/metabolism , Animals , Caspase 3/metabolism , Cell Cycle , Cell Line, Tumor , Dose-Response Relationship, Drug , Female , Humans , In Vitro Techniques , Mice , Mice, Nude , Phosphorylation , Proto-Oncogene Proteins c-bcl-2/metabolism , Time Factors , Vitis , bcl-2-Associated X Protein/metabolism , bcl-X Protein/metabolism
13.
Toxicol Appl Pharmacol ; 263(1): 122-30, 2012 Aug 15.
Article in English | MEDLINE | ID: mdl-22749965

ABSTRACT

Grape seed proanthocyanidins (GSPs) have been shown to have anti-skin carcinogenic effects in in vitro and in vivo models. However, the precise epigenetic molecular mechanisms remain unexplored. This study was designed to investigate whether GSPs reactivate silenced tumor suppressor genes following epigenetic modifications in skin cancer cells. For this purpose, A431 and SCC13 human squamous cell carcinoma cell lines were used as in vitro models. The effects of GSPs on DNA methylation, histone modifications and tumor suppressor gene expressions were studied in these cell lines using enzyme activity assays, western blotting, dot-blot analysis and real-time polymerase chain reaction (RT-PCR). We found that treatment of A431 and SCC13 cells with GSPs decreased the levels of: (i) global DNA methylation, (ii) 5-methylcytosine, (iii) DNA methyltransferase (DNMT) activity and (iv) messenger RNA (mRNA) and protein levels of DNMT1, DNMT3a and DNMT3b in these cells. Similar effects were noted when these cancer cells were treated identically with 5-aza-2'-deoxycytidine, an inhibitor of DNA methylation. GSPs decreased histone deacetylase activity, increased levels of acetylated lysines 9 and 14 on histone H3 (H3-Lys 9 and 14) and acetylated lysines 5, 12 and 16 on histone H4, and reduced the levels of methylated H3-Lys 9. Further, GSP treatment resulted in re-expression of the mRNA and proteins of silenced tumor suppressor genes, RASSF1A, p16(INK4a) and Cip1/p21. Together, this study provides a new insight into the epigenetic mechanisms of GSPs and may have significant implications for epigenetic therapy in the treatment/prevention of skin cancers in humans.


Subject(s)
Antineoplastic Agents/pharmacology , Carcinoma, Squamous Cell/genetics , Gene Silencing/drug effects , Genes, Tumor Suppressor/drug effects , Grape Seed Extract/pharmacology , Proanthocyanidins/pharmacology , Skin Neoplasms/genetics , Azacitidine/analogs & derivatives , Azacitidine/pharmacology , Blotting, Western , Carcinoma, Squamous Cell/drug therapy , Cell Line, Tumor , DNA Methylation/drug effects , Decitabine , Histone Acetyltransferases/metabolism , Humans , Immunoblotting , Real-Time Polymerase Chain Reaction , Skin Neoplasms/drug therapy
14.
Photochem Photobiol ; 88(5): 1066-74, 2012.
Article in English | MEDLINE | ID: mdl-22017262

ABSTRACT

The importance of epigenetic alterations in the development of various diseases including the cancers has been realized. As epigenetic changes are reversible heritable changes, these can be utilized as an effective strategy for the prevention of cancers. DNA methylation is the most characterized epigenetic mechanism that can be inherited without changing the DNA sequence. Although limited available data suggest that silencing of tumor suppressor genes in ultraviolet (UV) radiation-exposed epidermis leads to photocarcinogenesis and is associated with a network of epigenetic modifications including alterations in DNA methylation, DNA methyltransferases and histone acetylations. Various bioactive dietary components have been shown to protect skin from UV radiation-induced skin tumors in animal models. The role of bioactive dietary components, such as, (-)-epicatechins from green tea and proanthocyanidins from grape seeds has been assessed in chemoprevention of UV-induced skin carcinogenesis and underlying epigenetic mechanism in vitro and in vivo animal models. These bioactive components have the ability to block UV-induced DNA hypermethylation and histone modifications in the skin required for the silencing of tumor suppressor genes (e.g. Cip1/p21, p16(INK4a) ). This information is of importance for understanding the role of epigenetic modulation in UV-induced skin tumor and the chemopreventive mechanism of bioactive dietary components.


Subject(s)
Biological Products/pharmacology , Cell Transformation, Neoplastic/radiation effects , Epigenesis, Genetic/radiation effects , Melanoma/genetics , Skin Neoplasms/genetics , Skin/metabolism , Animals , Biological Products/metabolism , Catechin/metabolism , Catechin/pharmacology , Cell Transformation, Neoplastic/drug effects , DNA Methylation , Diet , Epigenesis, Genetic/drug effects , Gene Expression Regulation/drug effects , Gene Expression Regulation/radiation effects , Humans , Melanoma/etiology , Melanoma/metabolism , Melanoma/prevention & control , Proanthocyanidins/pharmacology , Skin/drug effects , Skin/radiation effects , Skin Neoplasms/etiology , Skin Neoplasms/metabolism , Skin Neoplasms/prevention & control , Tumor Suppressor Proteins/antagonists & inhibitors , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/metabolism , Ultraviolet Rays/adverse effects
15.
PLoS One ; 6(7): e23000, 2011.
Article in English | MEDLINE | ID: mdl-21829575

ABSTRACT

Metastatic melanoma is a leading cause of death from skin diseases, and is often associated with activation of Wnt/ß-catenin signaling pathway. We have examined the inhibitory effect of silymarin, a plant flavanoid from Silybum marianum, on cell migration of metastasis-specific human melanoma cell lines (A375 and Hs294t) and assessed whether Wnt/ß-catenin signaling is the target of silymarin. Using an in vitro invasion assay, we found that treatment of human melanoma cell lines with silymarin resulted in concentration-dependent inhibition of cell migration, which was associated with accumulation of cytosolic ß-catenin, while reducing the nuclear accumulation of ß-catenin (i.e., ß-catenin inactivation) and reducing the levels of matrix metalloproteinase (MMP) -2 and MMP-9 which are the down-stream targets of ß-catenin. Silymarin enhanced: (i) the levels of casein kinase 1α, glycogen synthase kinase-3ß and phosphorylated-ß-catenin on critical residues Ser(45), Ser(33/37) and Thr(41), and (ii) the binding of ß-transducin repeat-containing proteins (ß-TrCP) with phospho forms of ß-catenin in melanoma cells. These events play important roles in degradation or inactivation of ß-catenin. To verify whether ß-catenin is a potent molecular target of silymarin, the effect of silymarin was determined on ß-catenin-activated (Mel 1241) and ß-catenin-inactivated (Mel 1011) melanoma cells. Treatment of Mel 1241 cells with silymarin or FH535, an inhibitor of Wnt/ß-catenin pathway, significantly inhibited cell migration of Mel 1241 cells, which was associated with the elevated levels of casein kinase 1α and glycogen synthase kinase-3ß, and decreased accumulation of nuclear ß-catenin and inhibition of MMP-2 and MMP-9 levels. However, this effect of silymarin and FH535 was not found in Mel 1011 melanoma cells. These results indicate for the first time that silymarin inhibits melanoma cell migration by targeting ß-catenin signaling pathway.


Subject(s)
Cell Movement/drug effects , Melanoma/metabolism , Melanoma/pathology , Signal Transduction/drug effects , Silymarin/pharmacology , beta Catenin/metabolism , Antioxidants/pharmacology , Apoptosis/drug effects , Blotting, Western , Casein Kinase Ialpha/metabolism , Cell Adhesion/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Fluorescent Antibody Technique , Gene Expression Regulation, Neoplastic , Glycogen Synthase Kinases/metabolism , Humans , Matrix Metalloproteinase 2/metabolism , Matrix Metalloproteinase 9/metabolism , Melanoma/drug therapy , Neoplasm Invasiveness , Phosphorylation , Wnt Proteins/metabolism , Wound Healing
16.
PLoS One ; 6(6): e21539, 2011.
Article in English | MEDLINE | ID: mdl-21738696

ABSTRACT

Melanoma is the leading cause of death from skin disease due, in large part, to its propensity to metastasize. We have examined the effect of grape seed proanthocyanidins (GSPs) on melanoma cancer cell migration and the molecular mechanisms underlying these effects using highly metastasis-specific human melanoma cell lines, A375 and Hs294t. Using in vitro cell invasion assays, we observed that treatment of A375 and Hs294t cells with GSPs resulted in a concentration-dependent inhibition of invasion or cell migration of these cells, which was associated with a reduction in the levels of cyclooxygenase (COX)-2 expression and prostaglandin (PG) E(2) production. Treatment of cells with celecoxib, a COX-2 inhibitor, or transient transfection of melanoma cells with COX-2 small interfering RNA, also inhibited melanoma cell migration. Treatment of cells with 12-O-tetradecanoylphorbol-13-acetate, an inducer of COX-2, enhanced the phosphorylation of ERK1/2, a protein of mitogen-activated protein kinase family, and subsequently cell migration whereas both GSPs and celecoxib significantly inhibited 12-O-tetradecanoylphorbol-13-acetate-promoted cell migration as well as phosphorylation of ERK1/2. Treatment of cells with UO126, an inhibitor of MEK, also inhibited the migration of melanoma cells. Further, GSPs inhibited the activation of NF-κB/p65, an upstream regulator of COX-2, in melanoma cells, and treatment of cells with caffeic acid phenethyl ester, an inhibitor of NF-κB, also inhibited cell migration. Additionally, inhibition of melanoma cell migration by GSPs was associated with reversal of epithelial-mesenchymal transition process, which resulted in an increase in the levels of epithelial biomarkers (E-cadherin and cytokeratins) while loss of mesenchymal biomarkers (vimentin, fibronectin and N-cadherin) in melanoma cells. Together, these results indicate that GSPs have the ability to inhibit melanoma cell invasion/migration by targeting the endogenous expression of COX-2 and reversing the process of epithelial-to-mesenchymal transition.


Subject(s)
Dinoprostone/biosynthesis , Epithelial-Mesenchymal Transition/drug effects , Grape Seed Extract/pharmacology , Proanthocyanidins/pharmacology , Cell Line, Tumor , Cell Movement/drug effects , Cyclooxygenase 2/genetics , Cyclooxygenase 2/metabolism , Epithelial-Mesenchymal Transition/genetics , Humans , Immunoassay , Transfection
17.
Carcinogenesis ; 32(4): 537-44, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21209038

ABSTRACT

The anti-skin carcinogenic effects of green tea catechins have been studied extensively in vitro and in vivo models but the precise epigenetic molecular mechanisms are still unclear. Accumulating data suggest that dietary phytochemicals may alter cancer risk by modifications of epigenetic processes in the cells. The present study was designed to investigate whether tea catechins, particularly (-)-epigallocatechin-3-gallate (EGCG), would modify epigenetic events to regulate DNA methylation-silenced tumor suppressor genes in skin cancer cells. DNA methylation, histone modifications and tumor suppressor gene expressions were studied in detail using human epidermoid carcinoma A431 cells as an in vitro model after EGCG treatment using cytostaining, western blotting, dot blot analysis, real-time polymerase chain reaction and enzymatic activity assays. Our study shows that EGCG treatment decreased global DNA methylation levels in A431 cells in a dose-dependent manner. EGCG decreased the levels of 5-methylcytosine, DNA methyltransferase (DNMT) activity, messenger RNA (mRNA) and protein levels of DNMT1, DNMT3a and DNMT3b. EGCG decreased histone deacetylase activity and increased levels of acetylated lysine 9 and 14 on histone H3 (H3-Lys 9 and 14) and acetylated lysine 5, 12 and 16 on histone H4 but decreased levels of methylated H3-Lys 9. Additionally, EGCG treatment resulted in re-expression of the mRNA and proteins of silenced tumor suppressor genes, p16INK4a and Cip1/p21. Together, our study provides new insight into the epigenetic mechanism of action of EGCG that may contribute to the chemoprevention of skin cancer and may have important implications for epigenetic therapy.


Subject(s)
Anticarcinogenic Agents/pharmacology , Catechin/analogs & derivatives , Cyclin-Dependent Kinase Inhibitor p21/genetics , DNA Methylation , Genes, p16 , Histones/metabolism , Skin Neoplasms/prevention & control , Acetylation , Azacitidine/analogs & derivatives , Azacitidine/pharmacology , Catechin/pharmacology , Cell Line, Tumor , DNA (Cytosine-5-)-Methyltransferase 1 , DNA (Cytosine-5-)-Methyltransferases/genetics , Decitabine , Histone Deacetylases/metabolism , Humans , Hydroxamic Acids/pharmacology , Skin Neoplasms/pathology , Transcriptional Activation
18.
Carcinogenesis ; 32(1): 86-92, 2011 Jan.
Article in English | MEDLINE | ID: mdl-20974686

ABSTRACT

Melanoma is the leading cause of death from skin disease due, in large part, to its propensity to metastasize. We have examined the effect of berberine, an isoquinoline alkaloid, on human melanoma cancer cell migration and the molecular mechanisms underlying these effects using melanoma cell lines, A375 and Hs294. Using an in vitro cell migration assay, we show that over expression of cyclooxygenase (COX)-2, its metabolite prostaglandin E2 (PGE2) and PGE2 receptors promote the migration of cells. We found that treatment of A375 and Hs294 cells with berberine resulted in concentration-dependent inhibition of migration of these cells, which was associated with a reduction in the levels of COX-2, PGE2 and PGE2 receptors (EP2 and EP4). Treatment of cells with celecoxib, a COX-2 inhibitor, or transient transfection of cells with COX-2 small interfering RNA, also inhibited cell migration. Treatment of the cells with 12-O-tetradecanoylphorbol-13-acetate (TPA), an inducer of COX-2 or PGE2, enhanced cell migration, whereas berberine inhibited TPA- or PGE2-promoted cell migration. Berberine reduced the basal levels as well as PGE2-stimulated expression levels of EP2 and EP4. Treatment of the cells with the EP4 agonist stimulated cell migration and berberine blocked EP4 agonist-induced cell migration activity. Moreover, berberine inhibited the activation of nuclear factor-kappa B (NF-κB), an upstream regulator of COX-2, in A375 cells, and treatment of cells with caffeic acid phenethyl ester, an inhibitor of NF-κB, inhibited cell migration. Together, these results indicate for the first time that berberine inhibits melanoma cell migration, an essential step in invasion and metastasis, by inhibition of COX-2, PGE2 and PGE2 receptors.


Subject(s)
Antineoplastic Agents/pharmacology , Berberine/pharmacology , Cell Movement/drug effects , Melanoma/metabolism , Blotting, Western , Cell Line, Tumor , Cyclooxygenase 2/biosynthesis , Cyclooxygenase 2/drug effects , Dinoprostone/biosynthesis , Gene Expression/drug effects , Humans , Melanoma/genetics , Receptors, Prostaglandin E/biosynthesis , Receptors, Prostaglandin E/drug effects , Transfection
19.
Carcinogenesis ; 32(4): 597-604, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21186298

ABSTRACT

Overexposure of the human skin to solar ultraviolet (UV) radiation is the major etiologic factor for development of skin cancers. Here, we report the results of epigenetic modifications in UV-exposed skin and skin tumors in a systematic manner. The skin and tumor samples were collected after chronic exposure of the skin of SKH-1 hairless mice to UVB radiation using a well-established photocarcinogenesis protocol. We found a distinct DNA hypermethylation pattern in the UVB-exposed epidermal skin and UVB-induced skin tumors that was associated with the elevated expression and activity of the DNA methyltransferases (Dnmt) 1, Dnmt3a and Dnmt3b. To explore the role of hypermethylation in skin photocarcinogenesis, we focused on the p16(INK4a) and RASSF1A tumor suppressor genes, which are transcriptionally silenced on methylation. We established that the silencing of these genes in UVB-exposed epidermis and UVB-induced skin tumors is associated with a network of epigenetic modifications, including hypoacetylation of histone H3 and H4 and increased histone deacetylation, as well as recruitment of methyl-binding proteins, including MeCP2 and MBD1, to the methylated CpGs. Higher levels of DNA methylation and DNMT activity in human squamous cell carcinoma specimens than in normal human skin suggest that the data are relevant clinically. Our data indicate for the first time that UVB-induced DNA hypermethylation, enhanced Dnmt activity and histone modifications occur in UVB-exposed skin and UVB-induced skin tumors and suggest that these events are involved in the silencing of tumor suppressor genes and in skin tumor development.


Subject(s)
DNA Methylation , Gene Silencing , Genes, Suppressor , Neoplasms, Radiation-Induced/genetics , Skin Neoplasms/genetics , Ultraviolet Rays , Animals , Chromatin Assembly and Disassembly , DNA (Cytosine-5-)-Methyltransferase 1 , DNA (Cytosine-5-)-Methyltransferases/genetics , DNA (Cytosine-5-)-Methyltransferases/metabolism , Epigenesis, Genetic , Female , Genes, p16 , Mice , Mice, Hairless , Tumor Suppressor Proteins/genetics
20.
Cancer Prev Res (Phila) ; 4(2): 238-47, 2011 Feb.
Article in English | MEDLINE | ID: mdl-21075976

ABSTRACT

The inhibition of UVB-induced immunosuppression by dietary grape seed proanthocyanidins (GSP) has been associated with the induction of interleukin (IL)-12 in mice, and we now confirm that GSPs do not inhibit UVB-induced immunosuppression in IL-12p40 knockout (IL-12 KO) mice and that treatment of these mice with recombinant IL-12 restores the inhibitory effect. To characterize the cell population responsible for the GSP-mediated inhibition of UVB-induced immunosuppression and the role of IL-12 in this process, we used an adoptive transfer approach. Splenocytes and draining lymph nodes were harvested from mice that had been administered dietary GSPs (0.5%-1.0%, w/w), exposed to UVB, and sensitized by the application of 2,4-dinitrofluorobenzene (DNFB) onto the UVB-exposed skin. CD8(+) and CD4(+) T cells were positively selected and transferred into naive mice that were subsequently challenged by application of DNFB on the ear skin. Naive recipients that received CD8(+) T cells from GSP-treated, UVB-irradiated donors exhibited full contact hypersensitivity (CHS) response. Naive mice that received CD4(+) suppressor T cells from GSP-treated, UVB-exposed mice could mount a CHS response after sensitization and subsequent challenge with DNFB. On culture, the CD8(+) T cells from GSP-treated, UVB-exposed mice secreted higher levels (5- to 8-fold) of Th1 cytokines than CD8(+) T cells from UVB-irradiated mice not treated with GSPs. CD4(+) T cells from GSP-treated, UVB-exposed mice secreted significantly lower levels (80%-100%) of Th2 cytokines than CD4(+) T cells from UVB-exposed mice not treated with GSPs. These data suggest that GSPs inhibit UVB-induced immunosuppression by stimulating CD8(+) effector T cells and diminishing regulatory CD4(+) T cells.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Dermatitis, Irritant/immunology , Grape Seed Extract/pharmacology , Immune Tolerance/drug effects , Interleukin-12 Subunit p40/physiology , Proanthocyanidins/pharmacology , Ultraviolet Rays , Adoptive Transfer , Animals , Dendritic Cells/drug effects , Dendritic Cells/immunology , Dendritic Cells/radiation effects , Dermatitis, Irritant/etiology , Dietary Supplements , Dinitrofluorobenzene/pharmacology , Female , Grape Seed Extract/administration & dosage , Lymph Nodes/cytology , Lymphocyte Activation/radiation effects , Mice , Mice, Inbred CBA , Mice, Knockout , Proanthocyanidins/administration & dosage , Skin/drug effects , Skin/immunology , Skin/radiation effects , Spleen/cytology , Vitis/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL
...