Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 40
Filter
Add more filters










Publication year range
1.
Fitoterapia ; 143: 104552, 2020 Jun.
Article in English | MEDLINE | ID: mdl-32173422

ABSTRACT

We have shown previously that alpha-santalol, a major component of sandalwood oil inhibits growth of cultured prostate cancer cells in vitro by causing apoptosis, but the mechanism of cell death is not fully elucidated. The present study was undertaken to investigate the role of PI3K/Akt/survivin pathway in alpha-santalol-induced apoptosis employing cultured LNCaP and PC-3 human prostate cancer cells. Treatment of prostate cancer cells with alpha-santalol (20, 40 µM) resulted in the down regulation of survivin and p-AKT (s-473) expression and statistically significant reduction in total survivin levels as evidenced by survivin ELISA assay. Furthermore, inhibition of PI3K-Akt pathway by pharmacological inhibitor, LY294002 enhanced the apoptotic cell death induced by alpha-santalol as determined by cell viability, cellular morphology, active caspase-3 activity and expression of cleaved PARP, cleaved caspase-3 levels. In conclusion, the present study provides novel insight into the molecular circuitry of alpha-santalol-induced cell death and reveals that alpha-santalol targets Akt/Survivin pathway to induce cell death and that the cell death is increased in the presence of a known inhibitor of the pathway.


Subject(s)
Apoptosis/drug effects , Polycyclic Sesquiterpenes/pharmacology , Prostatic Neoplasms/pathology , Proto-Oncogene Proteins c-akt/metabolism , Survivin/metabolism , Caspase 3/metabolism , Cell Line, Tumor , Humans , Male , PC-3 Cells , Phosphatidylinositol 3-Kinases/metabolism , Plant Oils/chemistry , Sesquiterpenes/chemistry , Signal Transduction
2.
Nat Prod Res ; 33(4): 527-543, 2019 Feb.
Article in English | MEDLINE | ID: mdl-29130352

ABSTRACT

Alpha-santalol is a naturally occurring sesquiterpene that is derived from sandalwood oil. Its wide range of health benefits have been attributed to the modulation of various signalling pathways involved in the development of a particular disease. For example, the antitumour and cancer preventive properties of alpha-santalol have been shown to involve cell death induction through apoptosis and cell cycle arrest in various cancer models. A marked decrease in inflammatory markers have also been shown with alpha-santalol administration in skin tissue models. The current review is aimed at bringing the most recent advances of alpha-santalol against various disease-specific models and highlighting its associated mechanistic details.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Antineoplastic Agents, Phytogenic/pharmacology , Sesquiterpenes/pharmacology , Animals , Apoptosis/drug effects , Cell Cycle Checkpoints/drug effects , Female , Humans , Inflammation/drug therapy , Inflammation/metabolism , Male , Neoplasms/drug therapy , Neoplasms/metabolism , Neoplasms/pathology , Plant Oils/chemistry , Polycyclic Sesquiterpenes , Sesquiterpenes/chemistry , Signal Transduction/drug effects
3.
Anticancer Res ; 38(8): 4475-4480, 2018 08.
Article in English | MEDLINE | ID: mdl-30061212

ABSTRACT

BACKGROUND/AIM: Alpha-santalol, a terpenoid found in sandalwood oil has been shown to inhibit breast cancer cell growth in vitro by inducing apoptosis, but the mechanisms underlying the growth inhibitory effects of alpha-santalol are not fully understood. In this study, we demonstrate that α-santalol treatment targets Wnt/ß-catenin pathway to inhibit migration of cultured breast cancer cells. MATERIALS AND METHODS: Migration assays, immunoblotting and immunofluorescence were used to examine the mechanism of action of a-santalol in breast cancer cells. RESULTS: Exposure of MDA-MB 231 and MCF-7 cells to α-santalol resulted in a significant reduction in their migratory potential and wound healing ability. In addition, α-santalol affected the localization of ß-catenin from cytosol to nucleus in MDA-MB 231 cells. CONCLUSION: Alpha-santalol inhibited migration of breast cancer cells may be mediated, in part, by targeting Wnt//ß-catenin pathway. ß-catenin represents an important target of α-santalol's response for future pre-clinical studies.


Subject(s)
Breast Neoplasms/drug therapy , Cell Movement/drug effects , Plant Oils/pharmacology , Sesquiterpenes/pharmacology , Signal Transduction/drug effects , beta Catenin/metabolism , Apoptosis/drug effects , Breast Neoplasms/metabolism , Cell Line, Tumor , Female , Humans , MCF-7 Cells , Polycyclic Sesquiterpenes , Wnt Signaling Pathway/drug effects , Wound Healing/drug effects
4.
Pharm Res ; 34(9): 1897-1907, 2017 Sep.
Article in English | MEDLINE | ID: mdl-28589445

ABSTRACT

PURPOSE: Almost all breast cancers originate from epithelial cells lining the milk ducts in the breast. To this end, the study investigated the feasibility of localized transdermal delivery of α-santalol, a natural chemopreventive agent to the breast. METHODS: Different α-santalol formulations (cream, solution and microemulsion) were developed and the in vitro permeability was studied using excised animal (porcine and rat) and human breast skin/mammary papilla (nipple). The in vivo biodistribution and efficacy studies were conducted in female rats. A chemical carcinogenesis model of breast cancer was used for the efficacy studies. RESULTS: Phospholipid based α-santalol microemulsion showed the highest penetration through the nipple and breast skin. Delivery of α-santalol through the entire breast (breast skin and nipple) in vivo in rats resulted in significantly higher concentration in the mammary gland compared to transdermal delivery through the breast skin or nipple. There was no measurable α-santalol concentration in the blood. Transdermal delivery of α-santalol reduced the tumor incidence and tumor multiplicity. Furthermore, the tumor size was significantly reduced with α-santalol treatment. CONCLUSIONS: The findings from this study demonstrate the feasibility of localized transdermal delivery of α-santalol for chemoprevention of breast cancer.


Subject(s)
Anticarcinogenic Agents/administration & dosage , Anticarcinogenic Agents/therapeutic use , Breast Neoplasms/prevention & control , Breast/drug effects , Sesquiterpenes/administration & dosage , Sesquiterpenes/therapeutic use , Skin Absorption , Administration, Cutaneous , Animals , Anticarcinogenic Agents/pharmacokinetics , Breast/metabolism , Breast/pathology , Breast Neoplasms/pathology , Chemoprevention , Female , Humans , Nipples/drug effects , Nipples/metabolism , Nipples/pathology , Polycyclic Sesquiterpenes , Rats , Rats, Sprague-Dawley , Sesquiterpenes/pharmacokinetics , Swine
5.
Biomed Res Int ; 2017: 5496398, 2017.
Article in English | MEDLINE | ID: mdl-28194418

ABSTRACT

Honokiol, a plant lignan has been shown to have antineoplastic effects against nonmelanoma skin cancer developments in mice. In this study, antineoplastic effects of honokiol were investigated in malignant melanoma models. In vitro effects of honokiol treatment on SKMEL-2 and UACC-62 melanoma cells were evaluated by measuring the cell viability, proliferation, apoptosis, cell cycle analysis, and expressions of various proteins associated with cell cycle progression and apoptosis. For the in vivo study, male nude mice inoculated with SKMEL-2 or UACC-62 cells received injections of sesame oil or honokiol for two to seven weeks. In vitro honokiol treatment caused significant decrease in cell viability, proliferation, cell cycle arrest, increased apoptosis, and modulation of apoptotic and cell cycle regulatory proteins. Honokiol caused an accumulation of cells in the G2/M phase of the cell cycle in SKMEL-2 and G0/G1 phase in UACC-62 cells. An elevated level of caspases and PARP were observed in both cell lines treated with honokiol. A decrease in the expression of various cell cycle regulatory proteins was also observed in honokiol treated cells. Honokiol caused a significant reduction of tumor growth in SKMEL-2 and UACC-62 melanoma xenografts. These findings suggest that honokiol is a good candidate for further studies as a possible treatment for malignant melanoma.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Apoptosis/drug effects , Biphenyl Compounds/pharmacology , Cell Cycle/drug effects , Lignans/pharmacology , Melanoma/drug therapy , Animals , Cell Line, Tumor , Cell Survival/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Humans , Male , Melanoma/metabolism , Melanoma/pathology , Mice , Mice, Nude , Neoplasm Proteins/biosynthesis , Xenograft Model Antitumor Assays
6.
Anticancer Res ; 35(10): 5353-7, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26408696

ABSTRACT

BACKGROUND: α-Santalol, a terpenoid found in sandalwood oil, has been shown to inhibit cancer cell growth in vitro by inducing apoptosis. This study was performed to investigate the anticancer properties of α-santalol associated with the induction of apoptosis in cultured MCF-7 [estrogen receptor (ER)-positive, and wild-type p53)] and MDA-MB-231 (ER-negative and mutant p53) breast cancer cells. MATERIALS AND METHODS: Expression of major proteins examined in the study were determined using a standard western blot protocol and analyzed by LICOR-Odyssey infra-red scanner. Total protein levels of survivin were confirmed by survivin enzyme-linked immunosorbent assay (ELISA) kit. Cell viability was assessed by the trypan blue dye exclusion assay, and caspase-3 activity was determined by caspase-3 (active) ELISA kit. RESULTS: Treatment of breast cancer cells for 6 and 9 h with α-santalol (20, and 40 µM) resulted in statistically significant concentration-dependent down-regulation of survivin. Phosphorylated protein kinase B (pAKT) levels were found to be slightly up-regulated despite the down-regulation of survivin. Pharmacological inhibition of the phosphoinositide 3-kinase - protein kinase B (PI3K-AKT) pathway did not result in a synergistic/additive increase in cell death or caspase-3 activity caused by α-santalol. CONCLUSION: The study reveals that survivin down-regulation by α-santalol in breast cancer cells is not mediated through the PI3K-AKT pathway.


Subject(s)
Antineoplastic Agents/pharmacology , Breast Neoplasms/metabolism , Inhibitor of Apoptosis Proteins/metabolism , Sesquiterpenes/pharmacology , Apoptosis , Breast Neoplasms/drug therapy , Cell Line, Tumor , Cell Proliferation/drug effects , Chromones/pharmacology , Down-Regulation , Female , Gene Expression Regulation, Neoplastic/drug effects , Humans , MAP Kinase Signaling System/drug effects , MCF-7 Cells , Morpholines/pharmacology , Polycyclic Sesquiterpenes , Survivin
7.
Anticancer Res ; 35(6): 3137-45, 2015 Jun.
Article in English | MEDLINE | ID: mdl-26026073

ABSTRACT

Effective management of tumorigenesis requires development of better anticancer agents with greater efficacy and fewer side-effects. Natural products are important sources for the development of chemotherapeutic agents and almost 60% of anticancer drugs are of natural origin. α-Santlol, a sesquiterpene isolated from Sandalwood, is known for a variety of therapeutic properties including anti-inflammatory, anti-oxidant, anti-viral and anti-bacterial activities. Cell line and animal studies reported chemopreventive effects of sandalwood oil and α-santalol without causing toxic side-effects. Our laboratory identified its anticancer effects in chemically-induced skin carcinogenesis in CD-1 and SENCAR mice, ultraviolet-B-induced skin carcinogenesis in SKH-1 mice and in vitro models of melanoma, non-melanoma, breast and prostate cancer. Its ability to induce cell-cycle arrest and apoptosis in cancer cells is its most reported anticancer mechanism of action. The present review discusses studies that support the anticancer effect and the mode of action of sandalwood oil and α-santalol in carcinogenesis.


Subject(s)
Carcinogenesis/drug effects , Plant Oils/therapeutic use , Santalum/chemistry , Skin Neoplasms/drug therapy , Animals , Apoptosis/drug effects , Cell Cycle Checkpoints/drug effects , Humans , Mice , Plant Extracts/chemistry , Plant Extracts/therapeutic use , Skin Neoplasms/chemically induced
8.
Nanoscale ; 7(9): 3903-14, 2015 Mar 07.
Article in English | MEDLINE | ID: mdl-25436837

ABSTRACT

The study was aimed at investigating the feasibility of using a poly (amidoamine) (PAMAM) dendrimer as a carrier for topical iontophoretic delivery of an antisense oligonucleotide (ASO). Bcl-2, an anti-apoptotic protein implicated in skin cancer, was used as the model target protein to demonstrate the topical gene silencing approach. Confocal laser scanning microscopy studies demonstrated that the iontophoretically delivered ASO-dendrimer complex can reach the viable epidermis in porcine skin. In contrast, passively delivered free or dendrimer complexed ASO was mainly localized to the stratum corneum. The cell uptake of ASO was significantly enhanced by the dendrimer complex and the complex suppressed Bcl-2 levels in the cell. In the skin cancer mouse model, the iontophoretically delivered ASO-dendrimer complex reduced the tumor volume by 45% and was consistent with the reduction in Bcl-2 protein levels. The iontophoretically delivered ASO-dendrimer complex caused significant apoptosis in skin tumor. Overall, the findings from this study demonstrate that dendrimers are promising nanocarriers for developing topical gene silencing approaches for skin diseases.


Subject(s)
Dendrimers/chemistry , Gene Silencing , Nanostructures/chemistry , Oligonucleotides, Antisense/therapeutic use , Skin Neoplasms/drug therapy , Administration, Topical , Animals , Apoptosis , Cell Line, Tumor , Disease Models, Animal , Humans , Mice , Microscopy, Confocal , Oligonucleotides, Antisense/chemistry , Oligonucleotides, Antisense/metabolism , Proto-Oncogene Proteins c-bcl-2/antagonists & inhibitors , Proto-Oncogene Proteins c-bcl-2/genetics , Proto-Oncogene Proteins c-bcl-2/metabolism
9.
Photochem Photobiol ; 89(4): 919-26, 2013.
Article in English | MEDLINE | ID: mdl-23480292

ABSTRACT

This study is designed to investigate the chemopreventive effect and molecular mechanisms of α-santalol on UVB-induced skin tumor development in SKH-1 hairless mouse, a widely used model for human photocarcinogenesis. A dose of UVB radiation (30 mJ cm(-2) day(-1)) that is in the range of human sunlight exposure was used for the initiation and promotion of tumor. Topical treatment of mice with α-santalol (10%, wt/vol in acetone) caused reduction in tumor incidence, multiplicity and volume. In our study, the anticarcinogenic action of α-santalol against UVB-induced photocarcinogenesis was found to be associated with inhibition of inflammation and epidermal cell proliferation, cell cycle arrest and induction of apoptosis. α-Santalol pretreatment strongly inhibited UVB-induced epidermal hyperplasia and thickness of the epidermis, expression of proliferation and inflammation markers proliferating cell nuclear antigen (PCNA), Ki-67 and cyclooxygenase 2 (Cox-2). Significant decrease in the expression of cyclins A, B1, D1 and D2 and cyclin-dependent kinases (Cdk)s Cdk1 (Cdc2), Cdk2, Cdk4 and Cdk6 and an upregulated expression of cyclin-dependent kinase (CDK) inhibitor Cip1/p21 were found in α-santalol pretreated group. Furthermore, an elevated level of cleaved caspase 3 and cleaved poly (ADP-ribose) polymerase (PARP) were observed in α-santalol-treated group. Our data suggested that α-santalol is a safer and promising skin cancer chemopreventive agent with potential to target various pathways involved in photocarcinogenesis.


Subject(s)
Sesquiterpenes/pharmacology , Skin Neoplasms/prevention & control , Ultraviolet Rays/adverse effects , Animals , Caspase 3/genetics , Caspase 3/metabolism , Cell Cycle/physiology , Cell Proliferation , Cyclooxygenase 2/genetics , Cyclooxygenase 2/metabolism , Female , Gene Expression Regulation/drug effects , Mice , Mice, Hairless , Poly (ADP-Ribose) Polymerase-1 , Poly(ADP-ribose) Polymerases/genetics , Poly(ADP-ribose) Polymerases/metabolism , Polycyclic Sesquiterpenes
10.
PLoS One ; 8(2): e56982, 2013.
Article in English | MEDLINE | ID: mdl-23451128

ABSTRACT

Anticancer efficacy and the mechanism of action of α-santalol, a terpenoid isolated from sandalwood oil, were investigated in human breast cancer cells by using p53 wild-type MCF-7 cells as a model for estrogen receptor (ER)-positive and p53 mutated MDA-MB-231 cells as a model for ER-negative breast cancer. α-Santalol inhibited cell viability and proliferation in a concentration and time-dependent manner in both cells regardless of their ER and/or p53 status. However, α-santalol produced relatively less toxic effect on normal breast epithelial cell line, MCF-10A. It induced G2/M cell cycle arrest and apoptosis in both MCF-7 and MDA-MB-231 cells. Cell cycle arrest induced by α-santalol was associated with changes in the protein levels of BRCA1, Chk1, G2/M regulatory cyclins, Cyclin dependent kinases (CDKs), Cell division cycle 25B (Cdc25B), Cdc25C and Ser-216 phosphorylation of Cdc25C. An up-regulated expression of CDK inhibitor p21 along with suppressed expression of mutated p53 was observed in MDA-MB-231 cells treated with α-santalol. On the contrary, α-santalol did not increase the expression of wild-type p53 and p21 in MCF-7 cells. In addition, α-santalol induced extrinsic and intrinsic pathways of apoptosis in both cells with activation of caspase-8 and caspase-9. It led to the activation of the executioner caspase-6 and caspase-7 in α-santalol-treated MCF-7 cells and caspase-3 and caspase-6 in MDA-MB-231 cells along with strong cleavage of poly(ADP-ribose) polymerase (PARP) in both cells. Taken together, this study for the first time identified strong anti-neoplastic effects of α-santalol against both ER-positive and ER-negative breast cancer cells.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Breast Neoplasms/pathology , Cell Division/drug effects , G2 Phase/drug effects , Receptors, Estrogen/metabolism , Sesquiterpenes/pharmacology , Breast Neoplasms/metabolism , Cell Line, Tumor , Cell Proliferation , Female , Flow Cytometry , Humans , In Situ Nick-End Labeling , Polycyclic Sesquiterpenes
11.
Mar Drugs ; 10(9): 2111-2125, 2012 Sep.
Article in English | MEDLINE | ID: mdl-23118725

ABSTRACT

Sarcophine-diol (SD) is a lactone ring-opened analogue of sarcophine. It has shown chemopreventive effects on chemically-induced skin tumor development in female CD-1 mice, as well as in a UVB-induced skin tumor development model in hairless SKH-1 mice at a dose of 30 µg SD applied topically and 180 mJ/cm(2) UVB. The objective of this study was to determine the dose-response on the chemopreventive effects of SD on SKH-1 hairless mice when exposed to a UVB radiation dose of 30 mJ/cm(2). This UVB dose better represents chronic human skin exposure to sunlight leading to skin cancer than previous studies applying much higher UVB doses. Carcinogenesis was initiated and promoted by UVB radiation. Female hairless SKH-1 mice were divided into five groups. The control group was topically treated with 200 µL of acetone (vehicle), and the SD treatment groups were topically treated with SD (30 µg, 45 µg, and 60 µg dissolved in 200 µL of acetone) 1 h before UVB radiation (30 mJ/cm(2)). The last group of animals received 60 µg SD/200 µL acetone without UVB exposure. These treatments were continued for 27 weeks. Tumor multiplicity and tumor volumes were recorded on a weekly basis for 27 weeks. Weight gain and any signs of toxicity were also closely monitored. Histological characteristics and the proliferating cell nuclear antigen (PCNA) were evaluated in the mice skin collected at the end of the experiment. The dose-response study proved a modest increase in chemopreventive effects with the increase in SD dose. SD reduced the number of cells positively stained with PCNA proliferation marker in mice skin. The study also showed that SD application without UVB exposure has no effect on the structure of skin. The results from this study suggest that broader range doses of SD are necessary to improve the chemopreventive effects.


Subject(s)
Anticarcinogenic Agents/pharmacology , Carcinoma, Squamous Cell/prevention & control , Diterpenes/pharmacology , Neoplasms, Radiation-Induced/prevention & control , Skin Neoplasms/prevention & control , Sunlight/adverse effects , Animals , Body Weight/radiation effects , Carcinoma, Squamous Cell/etiology , Carcinoma, Squamous Cell/metabolism , Female , Mice , Mice, Hairless , Neoplasms, Radiation-Induced/etiology , Neoplasms, Radiation-Induced/metabolism , Proliferating Cell Nuclear Antigen/metabolism , Skin/drug effects , Skin/radiation effects , Skin Neoplasms/etiology , Skin Neoplasms/metabolism , Ultraviolet Rays , Weight Gain/radiation effects
12.
Drug Discov Ther ; 6(3): 140-6, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22890204

ABSTRACT

Honokiol has shown chemopreventive effects in chemically-induced and UVB-induced skin cancer in mice. In this investigation, we assessed the time-effects of a topical low dose of honokiol (30 µg), and then the effects of different honokiol doses (30, 45, and 60 µg) on a UVB-induced skin cancer model to find an optimal dose and time for desirable chemopreventive effects. UVB radiation (30 mJ/cm(2), 5 days/week for 25 or 27 weeks) was used to induce skin carcinogenesis in SKH-1 mice. For the time-response experiment 30 µg honokiol in acetone was applied topically to the animals before the UVB exposure (30 min, 1 h, and 2 h) and after the UVB exposure (immediately, 30 min, and 1 h). Control groups were treated with acetone. For the dose-response study, animals were treated topically with acetone or honokiol (30, 45, and 60 µg) one hour before the UVB exposure. In the time-response experiment, honokiol inhibited skin tumor multiplicity by 49-58% while reducing tumor volumes by 70-89%. In the dose-response study, honokiol (30, 45, and 60 µg) significantly decreased skin tumor multiplicity by 36-78% in a dose-dependent manner, while tumor area was reduced by 76-94%. Honokiol (60 µg) significantly reduced tumor incidence by 40% as compared to control group. Honokiol applied in very low doses (30 µg) either before or after UVB radiation shows chemopreventive effects. Honokiol (30, 45, and 60 µg) prevents UVB-induced skin cancer in a dose-dependent manner. Honokiol can be an effective chemopreventive agent against skin cancer.


Subject(s)
Antineoplastic Agents, Phytogenic/therapeutic use , Biphenyl Compounds/therapeutic use , Drugs, Chinese Herbal/therapeutic use , Lignans/therapeutic use , Neoplasms, Radiation-Induced/drug therapy , Skin Neoplasms/drug therapy , Animals , Dose-Response Relationship, Drug , Female , Mice , Mice, Hairless , Neoplasms, Radiation-Induced/pathology , Skin Neoplasms/pathology , Tumor Burden/drug effects , Ultraviolet Rays , Weight Gain/drug effects
13.
Phytomedicine ; 19(8-9): 804-11, 2012 Jun 15.
Article in English | MEDLINE | ID: mdl-22571975

ABSTRACT

The anticancer effects of α-santalol, a major component of sandalwood oil, have been reported against the development of certain cancers such as skin cancer both in vitro and in vivo. The primary objectives of the current study were to investigate the cancer preventive properties of α-santalol on human prostate cancer cells PC-3 (androgen independent and P-53 null) and LNCaP (androgen dependent and P-53 wild-type), and determine the possible mechanisms of its action. The effect of α-santalol on cell viability was determined by trypan blue dye exclusion assay. Apoptosis induction was confirmed by analysis of cytoplasmic histone-associated DNA fragmentation using both an apoptotic ELISA kit and a DAPI fluorescence assay. Caspase-3 activity was determined using caspase-3 (active) ELISA kit. PARP cleavage was analyzed using immunoblotting. α-Santalol at 25-75 µM decreased cell viability in both cell lines in a concentration and time dependent manner. Treatment of prostate cancer cells with α-santalol resulted in induction of apoptosis as evidenced by DNA fragmentation and nuclear staining of apoptotic cells by DAPI. α-Santalol treatment also resulted in activation of caspase-3 activity and PARP cleavage. The α-santalol-induced apoptotic cell death and activation of caspase-3 was significantly attenuated in the presence of pharmacological inhibitors of caspase-8 and caspase-9. In conclusion, the present study reveals the apoptotic effects of α-santalol in inhibiting the growth of human prostate cancer cells.


Subject(s)
Apoptosis/drug effects , Caspase 3/metabolism , Plant Oils/chemistry , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/pathology , Sesquiterpenes/chemistry , Sesquiterpenes/pharmacology , Androgens/metabolism , Antineoplastic Agents, Phytogenic/pharmacology , Caspase 8/metabolism , Caspase 9/metabolism , Cell Line, Tumor , Cell Survival/drug effects , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , Humans , Male , Poly(ADP-ribose) Polymerases/metabolism , Polycyclic Sesquiterpenes , Prostatic Neoplasms/metabolism
14.
BMC Cancer ; 11: 456, 2011 Oct 20.
Article in English | MEDLINE | ID: mdl-22014088

ABSTRACT

BACKGROUND: Magnolol, a plant lignan isolated from the bark and seed cones of Magnolia officinalis, has been shown to have chemopreventive effects on chemically-induced skin cancer development. The objectives of this investigation are to study the anticarcinogenic effects of magnolol on UVB-induced skin tumor development in SKH-1 mice, a model relevant to humans, and determine the possible role of apoptosis and cell cycle arrest involved in the skin tumor development. METHODS: UVB-induced skin carcinogenesis model in SKH-1 mice was used for determining the preventive effects of magnolol on skin cancer development. Western blottings and flow cytometric analysis were used to study the effects of magnolol on apoptosis and cell cycle. RESULTS: Magnolol pretreated groups (30, 60 µ g) before UVB treatments (30 mJ/cm2, 5 days/week) resulted in 27-55% reduction in tumor multiplicity as compared to control group in SKH-1 mice. Magnolol pretreatment increased the cleavage of caspase-8 and poly-(-ADP-ribose) polymerase (PARP), increased the expression of p21, a cell cycle inhibitor, and decreased the expression of proteins involved in the G2/M phase of cell cycle in skin samples from SKH-1 mice.Treatment of A431 cells with magnolol decreased cell viability and cell proliferation in a concentration dependent manner. Magnolol induced G2/M phase cell cycle arrest in A431 cells at 12 h with a decreased expression of cell cycle proteins such as cyclin B1, cyclin A, CDK4, Cdc2 and simultaneous increase in the expression of Cip/p21, a cyclin-dependent kinase inhibitor. Magnolol induced apoptosis in vivo and in vitro with an increased cleavage of caspase-8 and PARP. Phospho-signal transducers and activators of transcription 3 (Tyr705), B-Raf, p-MEK, and p-AKT were down-regulated, whereas phosphorylation of ERK was induced by magnolol in A431 cells. CONCLUSIONS: Magnolol pretreatments prevent UVB-induced skin cancer development by enhancing apoptosis, causing cell cycle arrest at G2/M phase, and affecting various signaling pathways. Magnolol could be a potentially safe and potent anticarcinogenic agent against skin cancer.


Subject(s)
Anticarcinogenic Agents/pharmacology , Biphenyl Compounds/pharmacology , Carcinoma, Squamous Cell/drug therapy , Lignans/pharmacology , Neoplasms, Radiation-Induced/drug therapy , Skin Neoplasms/drug therapy , Animals , Apoptosis/drug effects , Blotting, Western , Carcinoma, Squamous Cell/pathology , Carcinoma, Squamous Cell/prevention & control , Caspases/metabolism , Cell Cycle Checkpoints/drug effects , Cell Cycle Proteins/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Cyclin-Dependent Kinases/metabolism , Cyclins/metabolism , Flow Cytometry , Intracellular Signaling Peptides and Proteins/metabolism , Mice , Neoplasms, Radiation-Induced/pathology , Neoplasms, Radiation-Induced/prevention & control , Phosphorylation/drug effects , STAT3 Transcription Factor/metabolism , Skin Neoplasms/pathology , Skin Neoplasms/prevention & control , Ultraviolet Rays
15.
Exp Biol Med (Maywood) ; 236(11): 1351-9, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21908486

ABSTRACT

Honokiol is a plant lignan isolated from bark and seed cones of Magnolia officinalis. Recent studies from our laboratory indicated that honokiol pretreatment decreased ultraviolet B-induced skin cancer development in SKH-1 mice. The aim of the present investigation was to study the effects of honokiol on human epidermoid squamous carcinoma A431 cells and to elucidate possible mechanisms involved in preventing skin cancer. A431 cells were pretreated with different concentrations of honokiol for a specific time period and investigated for effects on apoptosis and cell cycle analysis. Treatment with honokiol significantly decreased cell viability and cell proliferation in a concentration- and time-dependent manner. Honokiol pretreatment at 50 µmol/L concentration induced G0/G1 cell cycle arrest significantly (P < 0.05) and decreased the percentage of cells in the S and G2/M phase. Honokiol down-regulated the expression of cyclin D1, cyclin D2, Cdk2, Cdk4 and Cdk6 proteins and up-regulated the expression of Cdk's inhibitor proteins p21 and p27. Pretreatment of A431 cells with honokiol leads to induction of apoptosis and DNA fragmentation. These findings indicate that honokiol provides its effects in squamous carcinoma cells by inducing cell cycle arrest at G0/G1 phase and apoptosis.


Subject(s)
Anticarcinogenic Agents/pharmacology , Biphenyl Compounds/pharmacology , Carcinoma, Squamous Cell/drug therapy , Cell Cycle Checkpoints/drug effects , Lignans/pharmacology , Skin Neoplasms/drug therapy , Animals , Anticarcinogenic Agents/therapeutic use , Apoptosis/drug effects , Biphenyl Compounds/therapeutic use , Carcinoma, Squamous Cell/pathology , Carcinoma, Squamous Cell/prevention & control , Cell Line, Tumor , Cell Proliferation/drug effects , Cyclin D1/genetics , Cyclin D2/genetics , Cyclin-Dependent Kinase 2/genetics , Cyclin-Dependent Kinase 2/metabolism , Cyclin-Dependent Kinase 4/genetics , Cyclin-Dependent Kinase 4/metabolism , Cyclin-Dependent Kinase 6/genetics , Cyclin-Dependent Kinase 6/metabolism , Cyclin-Dependent Kinase Inhibitor p21/genetics , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Cyclin-Dependent Kinase Inhibitor p27/genetics , Cyclin-Dependent Kinase Inhibitor p27/metabolism , DNA Fragmentation/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Humans , Lignans/therapeutic use , Mice , Skin Neoplasms/pathology , Skin Neoplasms/prevention & control
16.
Nutr Cancer ; 63(2): 242-7, 2011.
Article in English | MEDLINE | ID: mdl-21264790

ABSTRACT

Fatty acid composition of dietary fat plays a vital role in colon tumor development in animal models. Fats containing ω-6 fatty acids (e.g., corn oil) enhanced and ω-3 fatty acids (e.g., flaxseed oil) reduced chemically induced colon tumor development in rats. The objective of the present investigation was to study the effects of dietary canola oil, a source of ω-3 fatty acid on azoxymethane-induced colon cancer development in Fischer rats and compare with dietary corn oil. Dietary canola oil significantly (P<0.05) decreased colonic tumor incidence and tumor multiplicity as compared to dietary corn oil in rats. Fatty acid analysis showed that corn oil group had higher levels of ω-6 fatty acid levels, whereas the canola oil groups exhibited higher levels of ω-3 fatty acids from the colon and serum samples of rats. For the mechanistic study, COX-2 expression in the colon samples from the canola oil group was significantly lower (P<0.05) as compared to the corn oil group. Taken together, dietary canola oil may be chemopreventive for colon tumor development in Fischer rats as compared to possibly by increasing ω-3 fatty acid levels and decreasing COX-2 levels.


Subject(s)
Azoxymethane/toxicity , Chemoprevention , Colonic Neoplasms/prevention & control , Diet , Dietary Fats/administration & dosage , Fatty Acids, Monounsaturated/administration & dosage , Animals , Colon/pathology , Colonic Neoplasms/pathology , Corn Oil/administration & dosage , Cyclooxygenase 2/metabolism , Dietary Supplements , Fatty Acids, Omega-3/administration & dosage , Fatty Acids, Omega-6/administration & dosage , Linseed Oil/administration & dosage , Male , Rapeseed Oil , Rats , Rats, Inbred F344
17.
Front Biosci (Schol Ed) ; 3(2): 777-87, 2011 01 01.
Article in English | MEDLINE | ID: mdl-21196411

ABSTRACT

Alpha-santalol, a naturally occurring terpenoid, has been shown to have chemopreventive effects on both 7, 12-dimethylbenz(a)anthracene (DMBA)-initiated and 12-O- tetradecanoylphorbol-13-acetate (TPA)-promoted skin cancer development in CD-1 and SENCAR mice, and UVB-induced skin cancer developments in SKH-1 hairless mice in a concentration-dependent manner. Studies have demonstrated that α-santalol could be effective against skin carcinogenesis through both induction of apoptosis via caspase activation together with dissipation of mitochondria membrane potential and cytochrome c release in A431 cells, and inhibition of cell growth via induction of G2/M phase arrest in both A431 cells and melanoma UACC-62 cells by altering multiple cell cycle regulatory proteins and complexes. This review summarizes the chemopreventive effects and molecular mechanisms of α-santalol on skin cancer development in both animal models and skin cancer cell lines.


Subject(s)
Drugs, Chinese Herbal/therapeutic use , Phytotherapy/methods , Plant Oils/chemistry , Santalum/chemistry , Sesquiterpenes/chemistry , Sesquiterpenes/pharmacology , Sesquiterpenes/therapeutic use , Skin Neoplasms/prevention & control , 9,10-Dimethyl-1,2-benzanthracene/toxicity , Animals , Apoptosis/drug effects , Cell Cycle/drug effects , Cell Line, Tumor , Humans , Mice , Mice, Mutant Strains , Polycyclic Sesquiterpenes , Skin Neoplasms/chemically induced , Skin Neoplasms/drug therapy , Tetradecanoylphorbol Acetate/toxicity
18.
BMC Res Notes ; 3: 220, 2010 Aug 03.
Article in English | MEDLINE | ID: mdl-20682067

ABSTRACT

BACKGROUND: alpha-Santalol, an active component of sandalwood oil, has shown chemopreventive effects on skin cancer in different murine models. However, effects of alpha-santalol on cell cycle have not been studied. Thus, the objective of this study was to investigate effects of alpha-santalol on cell cycle progression in both p53 mutated human epidermoid carcinoma A431 cells and p53 wild-type human melanoma UACC-62 cells to elucidate the mechanism(s) of action. METHODS: MTT assay was used to determine cell viability in A431 cells and UACC-62; fluorescence-activated cell sorting (FACS) analysis of propidium iodide staining was used for determining cell cycle distribution in A431 cells and UACC-62 cells; immunoblotting was used for determining the expression of various proteins and protein complexes involved in the cell cycle progression; siRNA were used to knockdown of p21 or p53 in A431 and UACC-62 cells and immunofluorescence microscopy was used to investigate microtubules in UACC-62 cells. RESULTS: alpha-Santalol at 50-100 muM decreased cell viability from 24 h treatment and alpha-santalol at 50 muM-75 muM induced G2/M phase cell cycle arrest from 6 h treatment in both A431 and UACC-62 cells. alpha-Santalol altered expressions of cell cycle proteins such as cyclin A, cyclin B1, Cdc2, Cdc25c, p-Cdc25c and Cdk2. All of these proteins are critical for G2/M transition. alpha-Santalol treatment up-regulated the expression of p21 and suppressed expressions of mutated p53 in A431 cells; whereas, alpha-santalol treatment increased expressions of wild-type p53 in UACC-62 cells. Knockdown of p21 in A431 cells, knockdown of p21 and p53 in UACC-62 cells did not affect cell cycle arrest caused by alpha-santalol. Furthermore, alpha-santalol caused depolymerization of microtubules similar to vinblastine in UACC-62 cells. CONCLUSIONS: This study for the first time identifies effects of alpha-santalol in G2/M phase arrest and describes detailed mechanisms of G2/M phase arrest by this agent, which might be contributing to its overall cancer preventive efficacy in various mouse skin cancer models.

19.
Pharmacology ; 86(3): 168-73, 2010.
Article in English | MEDLINE | ID: mdl-20714207

ABSTRACT

The aim of the present study was to examine the effects of two neuronal nicotinic acetylcholine receptor ligands on acute ethanol-induced dopamine (DA) function in the C57BL/6J mouse ventral striatumusing an ex vivo assay and high-performance liquid chromatography coupled with electrochemicaldetection. Acute systemic injection of ethanol (2.5 g/kg) significantly increased the DA and dihydroxyphenylacetic acid (DOPAC) content in the ventral striatum. Pretreatment with lobeline (1 or 10 mg/kg) inhibited the ethanol-induced increase in the tissue DA and DOPAC content in the ventral striatum. Similarly, pretreatment with cytisine (0.5 or 3 mg/kg) also reduced the ethanol-induced increase in the tissue DA and DOPAC content in the ventral striatum. However, when given alone lobeline or cytisine did not produce significant effect on the DA or DOPAC content in the ventral striatum compared with controls. These findings provide evidence that lobeline and cytisine modulate ethanol-induced DA function by targeting nicotinic acetylcholine receptors in the ventral striatum, a reward-relevant brain region implicated in ethanol dependence.


Subject(s)
Alkaloids/pharmacology , Basal Ganglia/metabolism , Dopamine/metabolism , Ethanol/pharmacology , Lobeline/pharmacology , Receptors, Nicotinic/metabolism , 3,4-Dihydroxyphenylacetic Acid/metabolism , Alkaloids/metabolism , Animals , Azocines/metabolism , Azocines/pharmacology , Basal Ganglia/drug effects , Dose-Response Relationship, Drug , Ethanol/administration & dosage , Ligands , Lobeline/metabolism , Male , Mecamylamine/metabolism , Mecamylamine/pharmacology , Mice , Mice, Inbred C57BL , Nicotinic Agonists/metabolism , Nicotinic Agonists/pharmacology , Nicotinic Antagonists/metabolism , Nicotinic Antagonists/pharmacology , Quinolizines/metabolism , Quinolizines/pharmacology
20.
Anticancer Res ; 30(3): 777-83, 2010 Mar.
Article in English | MEDLINE | ID: mdl-20392996

ABSTRACT

BACKGROUND: Skin cancer is the most prevalent of all cancer types and its incidence is expected to increase substantially. Chemoprevention involves the administration of chemical agents to prevent initiation, promotion and/or progression that occurs during neoplastic development. Honokiol, a plant lignan isolated from bark and seed cones of Magnolia officinalis, has been shown to have chemopreventive effects on chemically induced skin cancer development. AIM: The objective of this investigation was to study the chemopreventive effects of honokiol on UVB-induced skin tumor development in SKH-1 mice, a model relevant to humans, and to elucidate the possible role of apoptotic proteins involved in the prevention of skin tumor development. MATERIALS AND METHODS: Female SKH-1 mice were divided into two groups. Group 1 received acetone (0.2 ml, topical) and Group 2 received honokiol (30 microg in 0.2 ml acetone, topical) one hour before UVB treatment. Tumor initiation and promotion were carried out by UVB radiation (30 mJ/cm(2)/day), 5 days a week for 30 weeks. Tumor counts and mouse weights were taken weekly. RESULTS: The honokiol-pretreated group exhibited a 45% reduction in tumor multiplicity as compared to the control group. Mechanistic studies showed the possible involvement of caspase-3, caspase-8, caspase-9, poly (ADP-ribose) polymerase (PARP) and p53 activation (p<0.05) leading to the induction of DNA fragmentation and apoptosis. CONCLUSION: Pretreatment with honokiol, at concentrations in micrograms per application compared with milligram applications of other potential chemopreventive agents, prevents UVB-induced skin cancer development, possibly by activating proapoptotic proteins through both intrinsic and extrinsic pathways.


Subject(s)
Anticarcinogenic Agents/pharmacology , Biphenyl Compounds/pharmacology , Lignans/pharmacology , Neoplasms, Radiation-Induced/prevention & control , Skin Neoplasms/prevention & control , Animals , Antineoplastic Agents, Phytogenic/pharmacology , Apoptosis/drug effects , Caspases/metabolism , Disease Models, Animal , Female , Mice , Neoplasms, Radiation-Induced/etiology , Neoplasms, Radiation-Induced/metabolism , Neoplasms, Radiation-Induced/pathology , Skin Neoplasms/etiology , Skin Neoplasms/metabolism , Skin Neoplasms/pathology , Tumor Suppressor Protein p53/metabolism , Ultraviolet Rays
SELECTION OF CITATIONS
SEARCH DETAIL