Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 64
Filter
1.
Crit Rev Microbiol ; 43(4): 466-480, 2017 Aug.
Article in English | MEDLINE | ID: mdl-28502224

ABSTRACT

Tuberculosis (TB) is a major public health problem, invading all age groups world-wide. It is an opportunistic infection affecting the individuals alone or with co-infections. Childhood TB is a neglected aspect and a significant health problem in epidemic areas. It constitutes more than 20% of TB incidence. Pediatric TB exists in the shadow of adult TB. The clinicians concentrate on pulmonary manifestation of TB, whereas it is a major problem in both pulmonary and extra-pulmonary infections. The rate of infection with this disease is mostly associated with poverty, social disruption and human immunodeficiency virus (HIV) infection. The diagnosis of extra-pulmonary TB (EPTB) is more difficult than pulmonary TB (PTB). Delayed diagnosis and executive treatment contribute to increase in the mortality rate in endemic areas. This article provides the evidence-based simple and safe screening method, indicating rapid, highly sensitive and specific diagnostic tests for pulmonary and EPTB in children. The most important aspect of treatment is the correct course of anti-tubercular drugs. This review serves the purpose of quick reference for microbiologists, epidemiologists, academicians, students and researchers. It provides guidance regarding early diagnosis and treatment accuracy of pediatric TB.


Subject(s)
Otitis Media/diagnosis , Tuberculoma/diagnosis , Tuberculosis, Lymph Node/diagnosis , Tuberculosis, Meningeal/diagnosis , Tuberculosis, Pulmonary/diagnosis , Tuberculosis, Pulmonary/epidemiology , Tuberculosis, Spinal/diagnosis , Tuberculosis, Urogenital/diagnosis , Adult , Child , Child, Preschool , Diagnostic Tests, Routine , Humans , Mycobacterium tuberculosis/drug effects , Mycobacterium tuberculosis/pathogenicity , Otitis Media/microbiology , Tuberculoma/microbiology , Tuberculosis, Lymph Node/microbiology , Tuberculosis, Meningeal/microbiology , Tuberculosis, Pulmonary/drug therapy , Tuberculosis, Pulmonary/microbiology , Tuberculosis, Spinal/microbiology , Tuberculosis, Urogenital/microbiology
2.
J Exp Ther Oncol ; 11(3): 181-188, 2016 Jul.
Article in English | MEDLINE | ID: mdl-28471123

ABSTRACT

Pristimerin (PM) is a quinonemethide triterpenoid with cytotoxic activity against a wide range of cancer cell lines. However, the effect of PM on IL-2 induced activation of T lymphocytes, which play a major role in antitumor immunity has not been studied. The objective of the present study was to evaluate the effect of PM on IL-2 induced proliferation of T cells, generation of lymphokine activated killer cells (LAK cells) and the signaling pathways involved in activation of T cells by IL-2. PM inhibited the IL-2 induced proliferation of mouse splenic T cells and the generation LAK cells at very low concentrations. The suppression of T cell proliferation by PM was associated with the inhibition of IL-2 induced Janus kinase/signal transducers and activators of transcription (Jak/STAT) and extracellular signal-regulated kinase 1 and 2 (Erk1/2) signaling pathways. PM also inhibited the proliferation and differentiation-related immediate early gene products such as p-c-fos, p-c-jun, c-myc and cyclin D1. In addition, antiapoptotic (prosurvival) NF-ƐĀŗB, p-Akt and p-mTOR were also inhibited by PM. These data demonstrated that PM inhibits IL-2 induced T cell activation and generation of LAK cells by disrupting multiple cell signaling pathways induced by IL-2.


Subject(s)
Antineoplastic Agents/pharmacology , Interleukin-2/pharmacology , Lymphocyte Activation/drug effects , Spleen/drug effects , T-Lymphocytes/drug effects , Triterpenes/pharmacology , Animals , Apoptosis Regulatory Proteins/metabolism , Cell Cycle Proteins/metabolism , Cell Differentiation/drug effects , Cell Proliferation/drug effects , Cells, Cultured , Dose-Response Relationship, Drug , Extracellular Signal-Regulated MAP Kinases/metabolism , Janus Kinases/metabolism , Killer Cells, Lymphokine-Activated/drug effects , Killer Cells, Lymphokine-Activated/immunology , Killer Cells, Lymphokine-Activated/metabolism , Mice , Pentacyclic Triterpenes , Phosphorylation , STAT Transcription Factors/metabolism , Signal Transduction/drug effects , Spleen/immunology , Spleen/metabolism , T-Lymphocytes/immunology , T-Lymphocytes/metabolism
3.
J Exp Ther Oncol ; 11(4): 251-260, 2016 Nov.
Article in English | MEDLINE | ID: mdl-27849335

ABSTRACT

Trichothecenes are powerful mycotoxins that inhibit protein synthesis and induce ribotoxic stress response in mammalian cells. Verrucarin A (VC-A) is a Type D macrocyclic mycotoxin which inhibits cell proliferation and induces apoptosis in cancer cells. However, the antitumor activity of VC-A for prostate cancer cells has not been investigated. The objective of the present study was to determine the anticancer activity and its mechanism of action in hormone-responsive (LNCaP) and hormone-refractory (PC-3) carcinoma of the prostate (CaP) cell lines. VC-A strongly inhibited the proliferation and induced cell cycle arrest in G2/M phase associated with the inhibition of cell cycle regulatory proteins cyclin D, cyclin E, cyclin-dependent kinases (cdks) cdk2, cdk4, cdk6 and cdk inhibitors WAF1/21 and KIP1/27. VC-A also induced apoptosis in CaP cells as characterized by the cleavage of poly (ADP-ribose) polymerase (PARP-1), procaspases-3, -8 and -9 and the inhibition of Bcl-2 family proteins that regulate apoptosis (Bcl-2, Bcl-xL, Bax, Bak and Bad). In addition, VC-A also down-regulated the expression of prosurvival phospho-AKT (p-AKT), nuclear factor kappa B (NF-kB) (p65) and phospho-mammalian target of rapamycin (p-mTOR) signaling proteins. Taken together, these results demonstrated strong antiproliferative and apoptosis-inducing activity of verrucarin A against CaP cells through cell cycle arrest and inhibition of the prosurvival (antiapoptotic) AKT/NF-kB/mTOR signaling pathway.


Subject(s)
Apoptosis/drug effects , Cell Proliferation/drug effects , NF-kappa B/metabolism , Prostatic Neoplasms/drug therapy , Proto-Oncogene Proteins c-akt/metabolism , TOR Serine-Threonine Kinases/metabolism , Trichothecenes/pharmacology , Apoptosis Regulatory Proteins/metabolism , Cell Cycle Checkpoints/drug effects , Cell Line, Tumor , Down-Regulation/drug effects , G2 Phase Cell Cycle Checkpoints/drug effects , Humans , Male , Mycotoxins/pharmacology , Prostate/drug effects , Prostate/metabolism , Prostatic Neoplasms/metabolism , Signal Transduction/drug effects
4.
J Exp Ther Oncol ; 11(1): 41-9, 2015.
Article in English | MEDLINE | ID: mdl-26259389

ABSTRACT

Pristimerin (PM) is a quinonemethide triterpenoid present in various plant species with strong antiprolifertive and proapoptotic activities in cancer cells. The effect of PM on telomerase which is reactivated in most cancers including carcinoma of the prostate (CaP) has not been studied. We investigated the effect of PM on the expression of human telomerase reverse transcriptase (hTERT) gene that codes for the catalytic subunit of the telomerase holoenzyme complex in prostate cancer cell lines LNCaP and PC-3 cells. The inhibition of cell proliferation and induction of apoptosis by PM in both cell lines was associated with the inhibition of hTERT mRNA expression, suppression of native and phosphorylated hTERT protein and hTERT telomerase activity. The ablation of hTERT expression increased the sensitivity of cancer cells to PM. In addition, results also revealed that the inhibition of hTERT expression is attributed to the inhibition of transcription factors SP1, c-Myc and STAT3 and protein kinase B/Akt which regulate hTERT transcriptionally and post-translationally, respectively. These data provide evidence that telomerase is a potential target of PM in prostate cancer.


Subject(s)
Antineoplastic Agents/pharmacology , Enzyme Inhibitors/pharmacology , Prostatic Neoplasms/enzymology , Telomerase/antagonists & inhibitors , Triterpenes/pharmacology , Apoptosis/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Dose-Response Relationship, Drug , Gene Expression Regulation, Neoplastic/drug effects , Humans , Male , Pentacyclic Triterpenes , Phosphorylation , Prostatic Neoplasms/genetics , Prostatic Neoplasms/pathology , RNA Interference , RNA, Messenger/metabolism , Signal Transduction/drug effects , Telomerase/genetics , Telomerase/metabolism , Transcription, Genetic/drug effects , Transfection
5.
J Exp Ther Oncol ; 10(4): 275-83, 2014.
Article in English | MEDLINE | ID: mdl-25509983

ABSTRACT

Pristimerin isaquinonemethidetriterpenoidthathasshown anticancer activity against some cancer types. However, the antitumor effects of pristimerin (PM) in ovarian cancer cells have not been adequately studied. The objective of the present study was to determine the anticancer activity and its mechanism of action in human ovarian carcinoma cell lines. PM strongly inhibited the proliferation of ovarian cancer cells by inducing apoptosis characterized by increased annexin V-binding, cleavage of poly (ADP-ribose) polymerase (PARP-1) and procaspases-3, -8 and -9. Furthermore, PM caused mitochondrial depolarization. Western blot analysis showed inhibition of prosurvival phospho-AKT (p-AKT), nuclear factor kappa B (NF-κB) (p65) and phospho-mammalian target of rapamycin (p-mTOR) signaling proteins in cells treated with PM. Treatment with PM also inhibited the expression of NF-κB-regulated antiapoptotic Bcl-2, Bcl-xL, c-IAP1 and survivin. Thus, our data showing potent antiproliferative and apoptosis-inducing activity of PM in ovarian carcinoma cells through the inhibition of AKT/ NF-κB/ mTOR signaling pathway warrant further investigation of PM for the management of ovarian cancer.


Subject(s)
Antineoplastic Agents/pharmacology , NF-kappa B/antagonists & inhibitors , Ovarian Neoplasms/drug therapy , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Signal Transduction/drug effects , TOR Serine-Threonine Kinases/antagonists & inhibitors , Triterpenes/pharmacology , Apoptosis/drug effects , Carcinoma/drug therapy , Carcinoma/metabolism , Carcinoma/pathology , Cell Line, Tumor , Cell Proliferation/drug effects , Female , Humans , Mitochondria/drug effects , NF-kappa B/metabolism , Ovarian Neoplasms/metabolism , Ovarian Neoplasms/pathology , Pentacyclic Triterpenes , Proto-Oncogene Proteins c-akt/metabolism , TOR Serine-Threonine Kinases/metabolism
6.
Molecules ; 18(3): 3250-65, 2013 Mar 13.
Article in English | MEDLINE | ID: mdl-23486104

ABSTRACT

Methyl-2-cyano-3,12-dioxooleana-1,9(11)-dien-28-oate (CDDO-Me) is a synthetic derivative of oleanolic acid, a triterpene, with apoptosis-inducing activity in a wide range of cancer cells. Induction of apoptosis by CDDO-Me is associated with the generation of reactive oxygen species (ROS) and inhibition of telomerase activity. In the present study, we investigated the role of ROS in inhibition of telomerase by CDDO-me. Treatment of MiaPaCa-2 and Panc-1 pancreatic cancer cell lines with CDDO-Me induced the production of hydrogen peroxide and superoxide anions and inhibited the telomerase activity. Pretreatment of cells with N-acetylcycsteine, a general purpose antioxidant or overexpression of glutathione peroxidase (GPx) or superoxide dismutase-1 (SOD-1) blocked the telomerase inhibitory activity of CDDO-Me. Furthermore, blocking ROS generation also prevented the inhibition of hTERT gene expression, hTERT protein production and expression of a number of hTERT-regulatory proteins by CDDO-Me (e.g., c-Myc, Sp1, NF-κB and p-Akt). Data also showed that Akt plays an important role in the activation of telomerase activity. Together, these data suggest that inhibition of telomerase activity by CDDO-Me is mediated through a ROS-dependent mechanism; however, more work is needed to fully understand the role of ROS in down-regulation of hTERT gene and hTERT-regulatory proteins by CDDO-Me.


Subject(s)
Oleanolic Acid/analogs & derivatives , Pancreatic Neoplasms/metabolism , Reactive Oxygen Species/metabolism , Telomerase/antagonists & inhibitors , Antioxidants/pharmacology , Apoptosis/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Humans , Oleanolic Acid/pharmacology , Pancreatic Neoplasms/genetics , Proto-Oncogene Proteins c-akt/metabolism , Telomerase/genetics , Telomerase/metabolism
7.
Biochem Biophys Res Commun ; 417(1): 570-5, 2012 Jan 06.
Article in English | MEDLINE | ID: mdl-22177954

ABSTRACT

Our previous studies have shown that methyl-2-cyano-3,12-dioxooleana-1,9(11)-dien-28-oate (CDDO-Me), a oleanane synthetic triterpenoid induces apoptosis in prostate cancer cells by inhibiting the Akt/NF-κB/mTOR signaling cascade; however, the mechanism by which CDDO-Me inhibits Akt/NF-κB/mTOR signaling has remained undetermined. Present studies show that Akt plays a critical role in the response of prostate cancer cells to CDDO-Me. Silencing of Akt sensitized PC-3 cells to CDDO-Me, whereas its overexpression rendered them resistant to CDDO-Me. Evaluation of the effect of CDDO-Me on Akt which lies upstream of NF-κB and mTOR showed that CDDO-Me directly inhibits the Akt kinase activity in cell-free kinase activity assay and in vivo without modulating the activity of PDK1, the upstream kinase that phosphorylates and activates Akt. The inhibition of Akt activity resulted in inhibition of phosphorylation/inactivation of proapoptotic procaspase-9, Bad and Foxo3a. Further, inhibition of p-Akt by CDDO-Me was not attributable to an increase in the activity of protein phosphatase 2A (PP2A) or PH domain/leucine-rich repeat protein phosphatase1 (PHLPP1) both of which dephosphorylate p-Akt. These findings show that Akt is a direct target of CDDO-Me in the Akt/NF-κB/mTOR prosurvival signaling axis.


Subject(s)
Antineoplastic Agents/pharmacology , Oleanolic Acid/analogs & derivatives , Prostatic Neoplasms/enzymology , Protein Phosphatase 2/metabolism , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Cell Line, Tumor , Humans , Male , NF-kappa B/metabolism , Oleanolic Acid/pharmacology , Pyruvate Dehydrogenase Acetyl-Transferring Kinase , TOR Serine-Threonine Kinases/metabolism
8.
Biochem Biophys Res Commun ; 422(4): 561-7, 2012 Jun 15.
Article in English | MEDLINE | ID: mdl-22609405

ABSTRACT

Methyl-2-cyano-3,12-dioxooleana-1,9(11)-dien-28-oate (CDDO-Me) is a multifunctional oleanane synthetic triterpenoid with potent anti-inflammatory and antitumorigenic properties. The mechanisms of the antisurvival and apoptosis-inducing activities of CDDO-Me and related derivatives of oleanolic acid have been defined; however, to date, no study has been carried out on the effect of CDDOs on human telomerase reverse transcriptase (hTERT) gene or telomerase activity. Here we report for the first time that inhibition of cell proliferation and induction of apoptosis by CDDO-Me in pancreatic cancer cell lines is associated with the inhibition of hTERT gene expression, hTERT telomerase activity and a number of proteins that regulate hTERT expression and activity. Furthermore, abrogation or overexpression of hTERT protein altered the susceptibility of tumor cells to CDDO-Me. These findings suggest that telomerase (hTERT) is a relevant target of CDDO-Me in pancreatic cancer cells.


Subject(s)
Antineoplastic Agents/pharmacology , Cell Proliferation/drug effects , Gene Expression Regulation, Enzymologic/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Oleanolic Acid/analogs & derivatives , Pancreatic Neoplasms/enzymology , Suppression, Genetic , Telomerase/antagonists & inhibitors , Cell Line, Tumor , Humans , Oleanolic Acid/pharmacology , Pancreatic Neoplasms/genetics , Telomerase/genetics
9.
J Exp Ther Oncol ; 10(1): 51-64, 2012.
Article in English | MEDLINE | ID: mdl-22946344

ABSTRACT

Oleanolic acid-derived synthetic triterpenoids are broad spectrum antiproliferative and antitumorigenic agents. In this study, we investigated the role of reactive oxygen species (ROS) in induction of apoptosis and inhibition of prosurvival Akt, NF-kappaB and mTOR signaling pro-teins by methyl-2-cyano-3,12-dioxooleana-1,9(11)-dien-28-oate (CDDO-Me) in pancreatic cancer cells. Micromolar concentrations of CDDO-Me inhibited proliferation and induced apoptosis in MiaPaCa-2 and Panc-1 pancreatic cancer cells. Treatment with CDDO-Me caused the generation of hydrogen peroxide and superoxide anion and pretreatment of cells with NADPH oxidase inhibitor diphylene iodonium (DPI) or respiratory chain complex 1 inhibitor rotenone prevented ROS generation. Pretreatment with N-acetylcysteine (NAC) or overexpression of glutathione peroxidase (GPx) or superoxide dismutase-1 (SOD-1) blocked the antiproliferative effects of CDDO-Me. Likewise, NAC prevented the induction of apoptosis (annexin V-FITC binding and cleavage of PARP-1 and procaspases-3,-8 and -9) and reversed the loss of mitochondrial membrane potential and release of cytochrome c from mitochondria by CDDO-Me. CDDO-Me down-regulated p-Akt, p-mTOR and NF-kappaB (p65) but increased the activation of Erk1/2 and NAC blocked the modulation of these cell signaling proteins by CDDO-Me. Thus, the results of this study indicate that the antiproliferative and apoptosis inducing effects of CDDO-Me are mediated through a ROS-dependent mechanism and the role of ROS in modulation of signaling proteins by CDDO-Me warrants further investigation.


Subject(s)
Acetylcysteine/pharmacology , Oleanolic Acid/analogs & derivatives , Pancreatic Neoplasms/metabolism , Reactive Oxygen Species , Apoptosis/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Glutathione Peroxidase/metabolism , Humans , Hydrogen Peroxide/metabolism , Membrane Potential, Mitochondrial , Oleanolic Acid/administration & dosage , Reactive Oxygen Species/antagonists & inhibitors , Reactive Oxygen Species/metabolism , Signal Transduction/drug effects , Superoxide Dismutase/metabolism , Superoxide Dismutase-1 , Superoxides/metabolism
10.
J Exp Ther Oncol ; 10(1): 1-8, 2012.
Article in English | MEDLINE | ID: mdl-22946339

ABSTRACT

Xanthohumol (XN), a prenylated chalcone present in hops exhibits anti-inflammatory, antioxidant and anticancer activity. In the present study we show that XN inhibits the proliferation of mouse lymphoma cells and IL-2 induced proliferation and cell cycle progression in mouse splenic T cells. The suppression of T cell proliferation by XN was due to the inhibition of IL-2 induced Janus kinase/signal transducers and activators of transcription (Jak/STAT) and extracellular signal-regulated kinase 1 and 2 (Erk1/2) signaling pathways. XN also inhibited proliferation-related cellular proteins such as c-Myc, c-Fos and NF-kappaB and cyclin D1. Thus, understanding of IL-2 induced cell signaling pathways in normal T cells, which are constitutively turned on in T cell lymphomas may facilitate development of XN for the treatment of hematologic cancers.


Subject(s)
Cell Proliferation , Flavonoids/pharmacology , Interleukin-2/metabolism , Propiophenones/pharmacology , T-Lymphocytes , Animals , Cell Cycle Proteins/drug effects , Cell Cycle Proteins/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Janus Kinases/metabolism , MAP Kinase Signaling System/drug effects , Mice , Mice, Inbred C57BL , Spleen/cytology , Spleen/metabolism , T-Lymphocytes/drug effects , T-Lymphocytes/metabolism
11.
Molecules ; 17(12): 14795-809, 2012 Dec 11.
Article in English | MEDLINE | ID: mdl-23519253

ABSTRACT

Methyl-2-cyano-3,12-dioxooleana-1,9(11)-dien-28-oate (CDDO-Me) is an synthetic oleanane triterpenoid with strong antiprolifertive and proapoptotic activities in cancer cells. However, the effect of CDDO-Me on human telomerase reverse transcriptase (hTERT) and its telomerase activity in prostate cancer cells has not been studied. We investigated the role of hTERT in mediating the anticancer activity of CDDO-Me in prostate cancer cells in vitro and in vivo. The inhibition of cell proliferation and induction of apoptosis by CDDO-Me in LNCaP and PC-3 prostate cancer cell lines was associated with the inhibition of hTERT gene expression, hTERT telomerase activity and a number of proteins that regulate hTERT transcriptionally and post-translationally. Furthermore, ablation of hTERT protein increased the sensitivity of cancer cells to CDDO-Me, whereas its overexpression rendered them resistant to CDDO-Me. In addition, inhibition of progression of preneoplastic lesions (i.e., low and high-grade prostate intraepithelial neoplasms, PINs) to adenocarcinoma of the prostate by CDDO-Me in TRAMP mice was associated with significant decrease in TERT and its regulatory proteins in the prostate gland. These data provide evidence that telomerase is a potential target of CDDO-Me for the prevention and treatment of prostate cancer.


Subject(s)
Oleanolic Acid/analogs & derivatives , Telomerase/antagonists & inhibitors , Telomerase/metabolism , Animals , Apoptosis/drug effects , Blotting, Western , Cell Line, Tumor , Cell Proliferation/drug effects , Gene Expression Regulation , Humans , Male , Mice , NF-kappa B/antagonists & inhibitors , NF-kappa B/metabolism , Oleanolic Acid/pharmacology , Prostatic Neoplasms/drug therapy , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Proto-Oncogene Proteins c-akt/metabolism , STAT3 Transcription Factor/antagonists & inhibitors , STAT3 Transcription Factor/metabolism
12.
Carcinogenesis ; 32(5): 757-64, 2011 May.
Article in English | MEDLINE | ID: mdl-21325633

ABSTRACT

In an extension of our previous studies showing potent antitumorigenic activity of synthetic triterpenoids of oleanolic acid against prostate cancer cell lines, we examined the efficacy of 2-cyano-3,12-dioxooleana-1,9(11)-dien-28-oic acid (CDDO) in preventing the development and/or progression of prostate cancer in the transgenic adenocarcinoma of the mouse prostate (TRAMP) model. Data show that oral gavage with CDDO (10 Āµmol/kg) for 20 weeks resulted in inhibition of the progression of preneoplastic lesions in the dorsolateral prostate and ventral prostate to adenocarcinoma without toxicity. CDDO also inhibited metastasis of tumor to the distant organs. Treatment with CDDO significantly inhibited cell proliferation, reduced the density of blood vessels and promoted apoptosis in the prostatic tissue. Further, Akt, NF-κB and NF-κB regulated Bcl-2, Bcl-xL, survivin and cIAP1 appear to be the molecular targets of CDDO for inhibiting the progression of prostate cancer in TRAMP mice. Thus, these studies show for the first time the potential of CDDO for chemoprevention of human prostate cancer.


Subject(s)
Apoptosis/drug effects , Kidney Neoplasms/drug therapy , Liver Neoplasms/drug therapy , Lung Neoplasms/drug therapy , Oleanolic Acid/analogs & derivatives , Prostatic Neoplasms/drug therapy , Signal Transduction/drug effects , Adenocarcinoma/drug therapy , Adenocarcinoma/pathology , Animals , Animals, Genetically Modified , Blotting, Western , Cell Proliferation/drug effects , Disease Progression , Female , Humans , Immunoenzyme Techniques , Kidney Neoplasms/secondary , Liver Neoplasms/secondary , Lung Neoplasms/secondary , Lymphatic Metastasis , Male , Mice , Mice, Inbred C57BL , Nitric Oxide/antagonists & inhibitors , Oleanolic Acid/therapeutic use , Prostatic Intraepithelial Neoplasia/drug therapy , Prostatic Intraepithelial Neoplasia/pathology , Prostatic Neoplasms/pathology , RNA, Messenger/genetics , Reverse Transcriptase Polymerase Chain Reaction
13.
J Exp Ther Oncol ; 9(2): 119-27, 2011.
Article in English | MEDLINE | ID: mdl-21699019

ABSTRACT

CDDO-Me, an oleanane synthetic triterpenoid has shown strong antitumorigeic activity towards diverse cancer cell types including colorectal cancer cells. In the present study, we investigated the role of free radicals in the growth inhibitory and apoptosis-inducing activity of CDDO-Me in colorectal cancer cells lines. Results demonstrated that CDDO-Me potently inhibited the growth of colorectal cancer cells and pretreatment of cancer cells with small-molecule antioxidant N-acetylcysteine (NAC) completely blocked the growth inhibitory activity of CDDO-Me. CDDO-Me caused the generation of reactive oxygen species, which was inhibited by NAC and mitochondrial chain 1 complex inhibitors DPI and rotenone. CDDO-Me induced apoptosis as demonstrated by the cleavage of PARP-1, activation of procaspases -3, -8, and -9 and mitochondrial depolarization and NAC blocked the activation of these apoptosis related processes. Furthermore, induction of apoptosis by CDDO-Me was associated with the inhibition of antiapoptotic/ prosurvival Akt, mTOR and NF-kappaB signaling proteins and the inhibition of these signaling molecules was blocked by NAG. Together these studies provided evidence that CDDO-Me is a potent anticancer agent, which imparts growth inhibition and apoptosis in colorectal cancer cells through the generation of free radicals.


Subject(s)
Antineoplastic Agents/pharmacology , Colorectal Neoplasms/drug therapy , Oleanolic Acid/analogs & derivatives , Reactive Oxygen Species/metabolism , Acetylcysteine/metabolism , Antioxidants/metabolism , Apoptosis/drug effects , Cell Line, Tumor , Colorectal Neoplasms/pathology , Free Radicals/metabolism , Humans , Oleanolic Acid/pharmacology
14.
BMC Med Imaging ; 11: 21, 2011 Dec 13.
Article in English | MEDLINE | ID: mdl-22165848

ABSTRACT

BACKGROUND: The current method to determine the efficacy of chemoprevention in TRAMP mouse model of carcinoma of prostate (CaP) is by extracting and weighing the prostate at different time points or by immunohistochemistry analysis. Non-invasive determination of volumes of prostate glands and seminal vesicles before, during and after treatment would be valuable in investigating the efficacy of newer chemopreventive agents in CaP. The purpose of this study was to determine whether in vivo magnetic resonance imaging (MRI) using a 3 tesla clinical MRI system can be used to follow the effect of chemoprevention in TRAMP model of mouse CaP. METHODS: Mice were randomized into control and treated groups. The animals in treated group received 10 Āµmol/kg of CDDO, 5 days a week for 20 weeks. Animals underwent in vivo MRI of prostate gland and seminal vesicles by a clinical 3 Tesla MRI system just before (at 5 weeks), during and at the end of treatment, at 25 weeks. T1-weighted and fat saturation (FATSAT) multiecho fast spin echo T2-weighted images (T2WI) were acquired. Volume of the prostate glands and seminal vesicles was determined from MR images. T2 signal intensity changes in the seminal vesicles were determined by subtracting higher echo time (TE) from lower TE T2WI. Following treatments all animals were sacrificed, prostate and seminal vesicles collected, and the tissues prepared for histological staining. All data were expressed as mean Ā± 1 standard deviation. Two-way or multivariate analysis of variance followed by post-hoc test was applied to determine the significant differences. A p-value of <0.05 was considered significant. RESULTS: Histological analysis indicated tumor in 100% of control mice, whereas 10% of the treated mice showed tumor in prostate gland. Both MRI and measured prostate weights showed higher volume/weight in control mouse group. MRI showed significantly higher volume of seminal vesicles in control animals and T2 signal intensity changes in seminal vesicles of control mice indicating higher number of tumor foci, which was also proven by histology. CONCLUSIONS: In vivo MRI is helpful in determining the efficacy of chemoprevention of prostate cancer in TRAMP mice.


Subject(s)
Chemoprevention , Magnetic Resonance Imaging , Oleanolic Acid/analogs & derivatives , Prostatic Neoplasms/prevention & control , Animals , Disease Progression , Male , Mice , Mice, Transgenic , Oleanolic Acid/therapeutic use , Prostate/pathology , Prostatic Neoplasms/pathology , Seminal Vesicles/pathology
15.
Neurochem Res ; 35(1): 152-61, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19685289

ABSTRACT

Resveratrol is a naturally occurring polyphenolic compound highly enriched in grapes, peanuts, red wine, and a variety of food sources. Sulforaphane belongs to the family of isothiocyanates and is highly enriched in cruciferous vegetables. Our previous study showed that resveratrol, when used at high concentrations, inhibited cell proliferation, caused the cell cycle arrest and induced apoptotic cell death in glioma cells. In the current study, we tested the effect of combination treatment with resveratrol and sulforaphane, when both were used at low concentrations, on cell proliferation, migration and death in human U251 glioma cells. Our study shows that combination treatment with resveratrol and sulforaphane inhibits cell proliferation and migration, reduces cell viability, induces lactate dehydrogenase release, decreases pro-survival Akt phosphorylation and increases caspase-3 activation. The use of combination of bioactive food components, such as resveratrol and sulforaphane, may be a viable approach for the treatment of glioma.


Subject(s)
Apoptosis/drug effects , Glioma/pathology , Stilbenes/pharmacology , Thiocyanates/pharmacology , Cell Cycle/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Humans , Isothiocyanates , Resveratrol , Sulfoxides
16.
Prostate ; 69(8): 851-60, 2009 Jun 01.
Article in English | MEDLINE | ID: mdl-19189297

ABSTRACT

BACKGROUND: Synthetic triterpenoids are potent anticancer agents, but their therapeutic efficacy or mechanism of action for prostate cancer has not been investigated. The goal of this study was to determine the antitumor activity and the mechanism of action of methyl-2-cyano-3,12-dioxooleana-1,9(11)-dien-28-oate (CDDO-Me), a oleanane-derived synthetic triterpenoid for human prostate cancer cells. METHODS: The antitumor activity of CDDO-Me for hormone-refractory PC-3 (AR(-)) and C4-2 (AR(+)) prostate cancer cell lines was determined by effects on cell growth and induction of apoptosis, identification of molecular targets, and therapeutic efficacy in vivo in PC-3 xenograft model. RESULTS: CDDO-Me inhibited the growth and induced apoptosis in PC-3 and C4-2 cells at extremely low concentrations. The antitumor activity of CDDO-Me was associated with the inhibition of p-Akt, mammalian target of rapamycin (mTOR), and nuclear factor kappa B (NF-kappaB) signaling proteins and their downstream targets such as p-Bad and p-Foxo3a (Akt); p-S6K1, p-eIF-4E and p-4E-BP1 (mTOR); and COX-2, VEGF and cyclin D1(NF-kappaB). Silencing of Akt sensitized the PC-3 cells to CDDO-Me, whereas overexpression of Akt induced resistance to CDDO-Me. Targeted silencing of Akt showed that Akt does not regulate mTOR activation in PC-3 cells, but targeted silencing of mTOR sensitized PC-3 cells to CDDO-Me mediated growth inhibition. Further, treatment with CDDO-Me inhibited the growth of PC-3 xenografts in nude mice. CONCLUSIONS: This study demonstrated potent antitumor activity of CDDO-Me against prostate cancer cells both in vitro and in vivo. Data also identified Akt and mTOR as molecular targets of CDDO-Me in prostate cancer cells.


Subject(s)
Apoptosis/drug effects , Cell Division/drug effects , Oleanolic Acid/analogs & derivatives , Prostatic Neoplasms/pathology , Protein Kinases/genetics , Proto-Oncogene Proteins c-akt/genetics , Triterpenes/pharmacology , Animals , Blotting, Western , Cell Line, Tumor , Cell Survival/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Humans , Male , Mice , Mice, Nude , NF-kappa B/drug effects , NF-kappa B/metabolism , Nuclear Proteins/drug effects , Nuclear Proteins/isolation & purification , Oleanolic Acid/pharmacology , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/genetics , Protein Kinases/drug effects , Proto-Oncogene Proteins c-akt/drug effects , TOR Serine-Threonine Kinases , Transplantation, Heterologous
17.
J Exp Ther Oncol ; 8(1): 25-33, 2009.
Article in English | MEDLINE | ID: mdl-19827268

ABSTRACT

Resveratrol (trans-3,4', 5-trihydroxystilbene) is a naturally occurring polyphenolic compound that has antiinflammatory, antioxidant, neuroprotective properties and acts as a chemopreventive agent. Resveratrol causes cell cycle arrest and induces apoptotic cell death in various types of cancer cells. In the current studies, the effect of resveratrol on phosphoinositide kinase-3 (PI3K)/protein kinase B (Akt)/mammalian target of rapamycin (mTOR) signaling pathway was examined in human U251 glioma cells. Resveratrol decreased both the expression and phosphorylation of Akt. Inhibitors of PI3K (LY294002) and Akt (SH-6) enhanced resveratrol-induced LDH release and caspase-3 activation. Resveratrol reduced phosphorylation of ribosomal protein S6 and the mTOR inhibitor rapamycin further enhanced resveratrol-induced cell death. These results suggest that the downregulation of PI3K/Akt/mTOR signaling pathways may be an important mediator in resveratrol-induced apoptosis in glioma cells.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Glioma/drug therapy , Phosphoinositide-3 Kinase Inhibitors , Protein Kinases/drug effects , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Signal Transduction/drug effects , Stilbenes/pharmacology , Caspase 3/metabolism , Cell Line, Tumor , Cyclin D1/antagonists & inhibitors , Dose-Response Relationship, Drug , Down-Regulation , Glioma/pathology , Humans , Resveratrol , TOR Serine-Threonine Kinases
18.
Immunopharmacol Immunotoxicol ; 31(3): 477-84, 2009.
Article in English | MEDLINE | ID: mdl-19555200

ABSTRACT

Xanthohumol (XN), a prenylated chalcone present in hops (Humulus lupus L.) and beer, exhibits anti-inflammatory, antioxidant and antiproliferative activity, but has not been studied for effects on T cell-mediated immune responses. Here we demonstrate that XN has profound immunosuppressive effects on T cell proliferation, development of IL-2 activated killer (LAK) cells, cytotoxic T lymphocytes (CTLs), and production of Th1 cytokines (IL-2, IFN-gamma and TNF-alpha). The suppression of these cell-mediated immune responses by XN was at, least in part, due to the inhibition of nuclear factor kappa B (NF-kappaB) transcription factor through suppression of phosphorylation of IkappaBalpha, an inhibitor of NF-kappaB.


Subject(s)
Cell Proliferation/drug effects , Flavonoids/pharmacology , Immunosuppressive Agents/pharmacology , Lymphocytes/immunology , NF-kappa B/antagonists & inhibitors , Propiophenones/pharmacology , Animals , Cytokines/biosynthesis , Cytokines/immunology , Flavonoids/chemistry , Gene Expression Regulation/drug effects , Gene Expression Regulation/immunology , Humulus/chemistry , I-kappa B Proteins/immunology , I-kappa B Proteins/metabolism , Immunity, Cellular/drug effects , Immunity, Cellular/immunology , Immunosuppressive Agents/chemistry , Lymphocytes/metabolism , Male , Mice , Mice, Inbred C3H , NF-KappaB Inhibitor alpha , NF-kappa B/immunology , NF-kappa B/metabolism , Phosphorylation/drug effects , Phosphorylation/immunology , Propiophenones/chemistry
19.
J Exp Ther Oncol ; 7(1): 31-9, 2008.
Article in English | MEDLINE | ID: mdl-18472640

ABSTRACT

Chemoprevention represents a promising strategy to reducing the incidence of prostate cancer which afflicts more than 240,000 males annually in the U.S. 2-cyano-3,12-dioxooleana-1,9(11)-dien-28-oic acid (CDDO) and its C-28 methyl ester (CCDO-Me) and C-28 imidazole (CDDO-Im) derivatives are synthetic oleanane triterpenoids that exhibit several-fold more potent antiinflammatory activity than naturally occurring oleanolic acid, but have not been investigated for prevention of the prostate. In order to evaluate the anticancer activity of CDDOs for prostate cancer, we have investigated the effect of synthetic oleanane triterpenoids on molecular targets relevant to the chemoprevention and treatment of prostate cancer in vitro in TRAMPC-1 cells derived from the primary tumor in the prostate of a transgenic adenocarcinoma of the mouse prostate (TRAMP) mouse. Data demonstrate that CDDOs strongly inhibit the proliferation of TRAMPC-1 cells with a potency order of CDDO-Me>CDDO-Im>CDDO. Because CDDO-Me showed the most growth inhibitory activity it was further analyzed for the anticancer activity. CDDO-Me induced apoptosis in TRAMPC-1 cells as shown by the increased binding of annexin V-FITC and cleavage of procaspases 3, -8, and -9. It effectively inhibited the molecular targets such as p-Akt, NF-kappaB, and p-mTOR and downstream effectors of mTOR (p-S6K1, cyclin-D1, and cdk4). Further, CDDO-Me inhibited NF-kappaB-regulated antiapoptotic Bcl-2, Bcl-xL, and XIAP and proangiogenic VEGF. Taken together, these data demonstrate that CDDO-Me is potentially a potent chemopreventive agent that inhibits several molecular targets that are known to play critical roles in the development and progression of prostate cancer.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Oleanolic Acid/analogs & derivatives , Prostatic Neoplasms/metabolism , Animals , Animals, Genetically Modified , Antineoplastic Agents/therapeutic use , Cell Line, Tumor/drug effects , Cell Proliferation/drug effects , Down-Regulation/drug effects , Enzyme Inhibitors/pharmacology , Enzyme Inhibitors/therapeutic use , Humans , Male , Mice , NF-kappa B/antagonists & inhibitors , NF-kappa B/metabolism , Nitric Oxide/antagonists & inhibitors , Nitric Oxide/metabolism , Oleanolic Acid/pharmacology , Oleanolic Acid/therapeutic use , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/pathology , Protein Kinases/metabolism , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Proto-Oncogene Proteins c-akt/metabolism , TOR Serine-Threonine Kinases
20.
Oncol Res ; 17(6): 257-67, 2008.
Article in English | MEDLINE | ID: mdl-19192720

ABSTRACT

Because of lack of effective treatment options for hormone-refractory prostate cancer at the present time, the need for developing novel therapeutic strategies and targets to treat and prevent the progression of hormone-sensitive prostate cancer to the hormone-refractory stage is paramount. Our previous in vitro studies have shown that curcumin sensitizes both hormone-sensitive and hormone-resistant prostate cancer cells to tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) and that combined curcumin/TRAIL treatment induces apoptosis in cancer cells by inhibiting antiapoptotic p-Akt and nuclear factor-kappaB (NF-kappaB). In the present study, we demonstrate that curcumin and TRAIL combination regimen is also the most effective treatment for inhibiting the growth of PC3 xenografts compared to curcumin or TRAIL monotherpy. The inhibition of PC3 tumors by combined treatment correlated with significant reduction in expression of p-Akt and NF-kappaB in tumor tissue. Furthermore, tumor growth inhibition by curcumin/TRAIL combination regimen was associated with significant decrease in cell proliferation and an increase in terminal deoxynucleotidyl transferase dUTP nick end labeling (TUNEL)-positive cells in the tumors without significant change in microvessel density. Based on the significant efficacy in this preclinical model, combined curcumin/TRAIL regimen may be an effective adjuvant therapy for hormone-refractory prostate cancer.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Prostatic Neoplasms/drug therapy , Animals , Antineoplastic Agents, Hormonal/therapeutic use , Apoptosis/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Curcumin/administration & dosage , Drug Resistance, Neoplasm , Humans , Male , Mice , Mice, Inbred BALB C , Mice, Nude , NF-kappa B/metabolism , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Proto-Oncogene Proteins c-akt/metabolism , TNF-Related Apoptosis-Inducing Ligand/administration & dosage , Treatment Outcome , Xenograft Model Antitumor Assays
SELECTION OF CITATIONS
SEARCH DETAIL