Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 47
Filter
Add more filters










Publication year range
1.
Nanomedicine (Lond) ; 15(11): 1147-1162, 2020 05.
Article in English | MEDLINE | ID: mdl-32292109

ABSTRACT

Many dietary polyphenols have been investigated for their therapeutic potential either as single agents or in combinations. Despite the significant anticancer potential of these polyphenols in in vitro cell culture and in vivo animal models, their clinical applications have been limited because of challenges such as ineffective systemic delivery, stability and low bioavailability. Nanoencapsulation of these polyphenols could prolong circulation, improve localization, enhance efficacy and reduce the chances of multidrug resistance. This review summarized the use of various polyphenols especially epigallocatechin gallate, quercetin, curcumin and resveratrol as nanoformulations for cancer prevention and treatment. Despite some success, more research is warranted to design a nanoencapsulated combination of polyphenols, effective in in vitro, in vivo and human systems.


Subject(s)
Catechin , Curcumin , Neoplasms , Animals , Catechin/pharmacology , Curcumin/pharmacology , Humans , Neoplasms/drug therapy , Neoplasms/prevention & control , Polyphenols/pharmacology , Quercetin , Resveratrol
2.
FASEB J ; 33(3): 3198-3211, 2019 03.
Article in English | MEDLINE | ID: mdl-30379590

ABSTRACT

Growth factor receptor-binding protein 10 (GRB10) is a well-known adaptor protein and a recently identified substrate of the mammalian target of rapamycin (mTOR). Depletion of GRB10 increases insulin sensitivity and overexpression suppresses PI3K/Akt signaling. Because the major reason for the limited efficacy of PI3K/Akt-targeted therapies in prostate cancer (PCa) is loss of mTOR-regulated feedback suppression, it is therefore important to assess the functional importance and regulation of GRB10 under these conditions. On the basis of these background observations, we explored the status and functional impact of GRB10 in PCa and found maximum expression in phosphatase and tensin homolog (PTEN)-deficient PCa. In human PCa samples, GRB10 inversely correlated with PTEN and positively correlated with pAKT levels. Knockdown of GRB10 in nontumorigenic PTEN null mouse embryonic fibroblasts and tumorigenic PCa cell lines reduced Akt phosphorylation and selectively activated a panel of receptor tyrosine kinases. Similarly, overexpression of GRB10 in PTEN wild-type PCa cell lines accelerated tumorigenesis and induced Akt phosphorylation. In PTEN wild-type PCa, GRB10 overexpression promoted mediated PTEN interaction and degradation. PI3K (but not mTOR) inhibitors reduced GRB10 expression, suggesting primarily PI3K-driven regulation of GRB10. In summary, our results suggest that GRB10 acts as a major downstream effector of PI3K and has tumor-promoting effects in prostate cancer.-Khan, M. I., Al Johani, A., Hamid, A., Ateeq, B., Manzar, N., Adhami, V. M., Lall, R. K., Rath, S., Sechi, M., Siddiqui, I. A., Choudhry, H., Zamzami, M. A., Havighurst, T. C., Huang, W., Ntambi, J. M., Mukhtar, H. Proproliferatve function of adaptor protein GRB10 in prostate carcinoma.


Subject(s)
GRB10 Adaptor Protein/metabolism , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Animals , Carcinogens/antagonists & inhibitors , Carcinogens/metabolism , Cell Line, Tumor , Cell Proliferation/genetics , Cell Proliferation/physiology , Fibroblasts/cytology , Fibroblasts/metabolism , GRB10 Adaptor Protein/antagonists & inhibitors , GRB10 Adaptor Protein/genetics , Gene Knockdown Techniques , Humans , Male , Mechanistic Target of Rapamycin Complex 1/metabolism , Mice , Models, Biological , PTEN Phosphohydrolase/deficiency , PTEN Phosphohydrolase/genetics , PTEN Phosphohydrolase/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Prostatic Neoplasms/genetics , RNA, Messenger , Signal Transduction
3.
Mol Cancer Ther ; 18(2): 356-363, 2019 02.
Article in English | MEDLINE | ID: mdl-30446585

ABSTRACT

Hyperactivated AKT kinase due to loss of its negative regulator PTEN influences many aspects of cancer biology, including chromatin. AKT primarily regulates acetyl-CoA production and phosphorylates many histone-modulating enzymes, resulting in their activation or inhibition. Therefore, understanding the therapeutic impact of AKT inhibition on chromatin-related events is essential. Here, we report that AKT inhibition in prostate-specific PTEN knockout mice significantly induces di- and trimethylation of H3K4 with concomitant reduction in H3K9 acetylation. Mechanistically, we observed that AKT inhibition reduces expression of the H3K4 methylation-specific histone demethylases KDM5 family, especially KDM5B expression at transcriptional levels. Furthermore, we observed that AKT negatively regulates miR-137 levels, which transcriptionally represses KDM5B expression. Overexpression of miR-137 significantly reduced KDM5B and increased H3K4 methylation levels but failed to change AKT phosphorylation. Overall, we observed that AKT transcriptionally regulates KDM5B mainly via repression of miR-137. Our data identify a mechanism by which AKT kinase modulates the prostate cancer epigenome through regulating H3K4 methylation. Additional studies on AKT inhibition-mediated induction of H3K4 methylation will help in designing strategies to enhance the therapeutic efficacy of PI3K/AKT inhibitors.


Subject(s)
Jumonji Domain-Containing Histone Demethylases/genetics , MicroRNAs/genetics , Nuclear Proteins/genetics , PTEN Phosphohydrolase/genetics , Phosphorylcholine/analogs & derivatives , Prostatic Neoplasms/drug therapy , Repressor Proteins/genetics , Acetylation/drug effects , Animals , Cell Line, Tumor , Gene Expression Regulation, Neoplastic/drug effects , Gene Knockout Techniques , Humans , Male , Methylation/drug effects , Mice , Phosphorylcholine/administration & dosage , Phosphorylcholine/pharmacology , Prostatic Neoplasms/genetics , Prostatic Neoplasms/metabolism , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Xenograft Model Antitumor Assays
4.
Pharmacol Res ; 129: 375-387, 2018 03.
Article in English | MEDLINE | ID: mdl-29233676

ABSTRACT

Tumorigenesis and epigenetic are closely linked with each other. Epigenetic changes are potential regulators of gene expression without involving any change in the DNA itself. More interestingly, epigenetic changes are reversible heritable changes which pass through generations. Many dietary bioactive ingredients regulate epigenetic control of cells and influence biochemical processes. Correlation between epigenetic regulation and cancer onset has been well established. Recent studies provide important information on the role of bioactive dietary components in cancer prevention and therapy. Several bioactive components are responsible for modification of the epigenome by affecting DNA methylation, histone modification, micro RNAs (miRNAs) and long non-coding RNAs (lncRNAs). This review summarizes recent advancements in this field and describes the role of many bioactive components in regulating human epigenome and how these modifications can be exploited for prevention and treatment of cancer.


Subject(s)
Diet , Epigenesis, Genetic , Neoplasms/genetics , Nutrients/pharmacology , Animals , Humans , Neoplasms/therapy , Nutrients/therapeutic use
5.
Semin Cancer Biol ; 49: 75-82, 2018 04.
Article in English | MEDLINE | ID: mdl-28546110

ABSTRACT

Tumor masses are deprived of oxygen and characterized by enhanced glucose uptake followed by glycolysis. Elevated glucose levels induce non-enzymatic glycosylation or glycation of proteins which leads to accumulation of advanced glycation end products (AGE). These AGE molecules bind to their respective receptors called the receptor for advanced glycation end products (RAGE) and initiate several aberrant signaling pathways leading to onset of diseases such as diabetes, Alzheimer's, atherosclerosis, heart failure and cancer. The role of AGE in cancer progression is being extensively studied in recent years. As cancer cells are hypoxic in nature and adapted to glycolysis, which induces glycation, its effects need to be understood in greater detail. Since AGE-RAGE signaling is involved in cancer progression, inhibition of AGE-RAGE interaction could be a potential therapeutic target. The purpose of this review is to highlight the role of AGE-RAGE interaction in hypoxic cancer cells.


Subject(s)
Antineoplastic Agents/pharmacology , Cell Transformation, Neoplastic/drug effects , Cell Transformation, Neoplastic/metabolism , Glycosylation , Hypoxia/metabolism , Animals , Glycation End Products, Advanced/antagonists & inhibitors , Glycation End Products, Advanced/metabolism , Glycosylation/drug effects , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Molecular Targeted Therapy , Protein Binding/drug effects , Receptor for Advanced Glycation End Products/genetics , Receptor for Advanced Glycation End Products/metabolism , Signal Transduction/drug effects
6.
Mol Cancer Ther ; 15(12): 2863-2874, 2016 12.
Article in English | MEDLINE | ID: mdl-27765854

ABSTRACT

Although treatment of prostate cancer has improved over the past several years, taxanes, such as cabazitaxel, remain the only form of effective chemotherapy that improves survival in patients with metastatic castration-resistant prostate cancer. However, the effectiveness of this class of drugs has been associated with various side effects and drug resistance. We previously reported that fisetin, a hydroxyflavone, is a microtubule-stabilizing agent and inhibits prostate cancer cell proliferation, migration, and invasion and suggested its use as an adjuvant for treatment of prostate and other cancer types. In this study, we investigated the effect of fisetin in combination with cabazitaxel with the objective to achieve maximum therapeutic benefit, reduce dose and toxicity, and minimize or delay the induction of drug resistance and metastasis. Our data show for the first time that a combination of fisetin (20 µmol/L) enhances cabazitaxel (5 nmol/L) and synergistically reduces 22Rν1, PC-3M-luc-6, and C4-2 cell viability and metastatic properties with minimal adverse effects on normal prostate epithelial cells. In addition, the combination of fisetin with cabazitaxel was associated with inhibition of proliferation and enhancement of apoptosis. Furthermore, combination treatment resulted in the inhibition of tumor growth, invasion, and metastasis when assessed in two in vivo xenograft mouse models. These results provide evidence that fisetin may have therapeutic benefit for patients with advanced prostate cancer through enhancing the efficacy of cabazitaxel under both androgen-dependent and androgen-independent conditions. This study underscores the benefit of the combination of fisetin with cabazitaxel for the treatment of advanced and resistant prostate cancer and possibly other cancer types. Mol Cancer Ther; 15(12); 2863-74. ©2016 AACR.


Subject(s)
Antineoplastic Agents/pharmacology , Flavonoids/pharmacology , Taxoids/pharmacology , Animals , Apoptosis/drug effects , Cell Line, Tumor , Cell Survival/drug effects , Disease Models, Animal , Drug Synergism , Drug Therapy, Combination , Flavonols , Humans , Male , Mice , Mice, Nude , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/metabolism , Prostatic Neoplasms/diagnosis , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/genetics , Prostatic Neoplasms/metabolism , Treatment Outcome , Tumor Stem Cell Assay , Xenograft Model Antitumor Assays
8.
Semin Cancer Biol ; 40-41: 130-140, 2016 10.
Article in English | MEDLINE | ID: mdl-27163728

ABSTRACT

The last few decades have seen a resurgence of interest among the scientific community in exploring the efficacy of natural compounds against various human cancers. Compounds of plant origin belonging to different groups such as alkaloids, flavonoids and polyphenols evaluated for their cancer preventive effects have yielded promising data, thereby offering a potential therapeutic alternative against this deadly disease. The flavonol fisetin (3,3',4',7-tetrahydroxyflavone), present in fruits and vegetables such as strawberries, apple, cucumber, persimmon, grape and onion, was shown to possess anti-microbial, anti-inflammatory, anti-oxidant and more significantly anti-carcinogenic activity when assessed in diverse cell culture and animal model systems. The purpose of this review is to update and discuss key findings obtained till date from in vitro and in vivo studies on fisetin, with special focus on its anti-cancer role. The molecular mechanism(s) described in the observed growth inhibitory effects of fisetin in different cancer cell types is also summarized. Moreover, an attempt is made to delineate the direction of future studies that could lead to the development of fisetin as a potent chemopreventive/chemotherapeutic agent against cancer.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Flavonoids/pharmacology , Neoplasms/drug therapy , Animals , Antineoplastic Agents, Phytogenic/therapeutic use , Drug Screening Assays, Antitumor , Flavonoids/therapeutic use , Flavonols , Humans , Molecular Targeted Therapy , Plant Extracts/pharmacology , Plant Extracts/therapeutic use
9.
Mol Nutr Food Res ; 60(6): 1396-405, 2016 06.
Article in English | MEDLINE | ID: mdl-27059089

ABSTRACT

Cancer remains a major public health concern and a significant cause of death worldwide. Identification of bioactive molecules that have the potential to inhibit carcinogenesis continues to garner interest among the scientific community. In particular, flavonoids from dietary sources are the most sought after because of their safety, cost-effectiveness, and feasibility of oral administration. Emerging data have provided newer insights into understanding the molecular mechanisms that are essential to identify novel mechanism-based strategies for cancer prevention and treatment. Dietary flavonoid fisetin (3,3',4',7-tetrahydroxyflavone) found in many fruits and vegetables has been shown in preclinical studies to inhibit cancer growth through alteration of cell cycle, inducing apoptosis, angiogenesis, invasion, and metastasis without causing any toxicity to normal cells. Although data from in-vitro and in-vivo studies look convincing, well-designed clinical trials in humans are needed to conclusively determine the efficacy across various cancers. This review highlights the chemopreventive and therapeutic effects, molecular targets, and mechanisms that contribute to the observed anticancer activity of fisetin against various cancers.


Subject(s)
Anticarcinogenic Agents/pharmacology , Flavonoids/pharmacology , Neoplasms/drug therapy , Neoplasms/prevention & control , Animals , Carcinogenesis/drug effects , Cell Line, Tumor , Chemoprevention , Female , Flavonols , Fruit/chemistry , Humans , Male , Polyphenols/pharmacology , Vegetables/chemistry
10.
Cancer Lett ; 367(2): 173-83, 2015 Oct 28.
Article in English | MEDLINE | ID: mdl-26235140

ABSTRACT

Microtubule targeting based therapies have revolutionized cancer treatment; however, resistance and side effects remain a major limitation. Therefore, novel strategies that can overcome these limitations are urgently needed. We made a novel discovery that fisetin, a hydroxyflavone, is a microtubule stabilizing agent. Fisetin binds to tubulin and stabilizes microtubules with binding characteristics far superior than paclitaxel. Surface plasmon resonance and computational docking studies suggested that fisetin binds to ß-tubulin with superior affinity compared to paclitaxel. Fisetin treatment of human prostate cancer cells resulted in robust up-regulation of microtubule associated proteins (MAP)-2 and -4. In addition, fisetin treated cells were enriched in α-tubulin acetylation, an indication of stabilization of microtubules. Fisetin significantly inhibited PCa cell proliferation, migration, and invasion. Nudc, a protein associated with microtubule motor dynein/dynactin complex that regulates microtubule dynamics, was inhibited with fisetin treatment. Further, fisetin treatment of a P-glycoprotein overexpressing multidrug-resistant cancer cell line NCI/ADR-RES inhibited the viability and colony formation. Our results offer in vitro proof-of-concept for fisetin as a microtubule targeting agent. We suggest that fisetin could be developed as an adjuvant for treatment of prostate and other cancer types.


Subject(s)
Flavonoids/pharmacology , Microtubules/drug effects , Prostatic Neoplasms/metabolism , Tubulin Modulators/pharmacology , Tubulin/metabolism , Acetylation , Animals , Binding Sites , Cell Cycle Proteins/metabolism , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Cell Survival/drug effects , Dose-Response Relationship, Drug , Flavonoids/chemistry , Flavonoids/metabolism , Flavonols , Male , Microtubule-Associated Proteins/metabolism , Microtubules/metabolism , Molecular Docking Simulation , Neoplasm Invasiveness , Nuclear Proteins/metabolism , Paclitaxel/pharmacology , Prostatic Neoplasms/genetics , Prostatic Neoplasms/pathology , Protein Binding , Protein Conformation , Protein Processing, Post-Translational , Protein Stability , Signal Transduction/drug effects , Surface Plasmon Resonance , Time Factors , Tubulin/chemistry , Tubulin Modulators/metabolism
11.
Curr Pharm Des ; 21(10): 1240-8, 2015.
Article in English | MEDLINE | ID: mdl-25506896

ABSTRACT

Globally, the cancer associated deaths are generally attributed to the spread of cancerous cells or their features to the nearby or distant secondary organs by a process known as metastasis. Among other factors, the metastatic dissemination of cancer cells is attributed to the reactivation of an evolutionary conserved developmental program known as epithelial to mesenchymal transition (EMT). During EMT, fully differentiated epithelial cells undergo a series of dramatic changes in their morphology, along with loss of cell to cell contact and matrix remodeling into less differentiated and invasive mesenchymal cells. Many studies provide evidence for the existence of EMT like states in prostate cancer (PCa) and suggest its possible involvement in PCa progression and metastasis. At the same time, the lack of conclusive evidence regarding the presence of full EMT in human PCa samples has somewhat dampened the interest in the field. However, ongoing EMT research provides new perspectives and unveils the enormous potential of this field in tailoring new therapeutic regimens for PCa management. This review summarizes the role of many transcription factors and other molecules that drive EMT during prostate tumorigenesis.


Subject(s)
Carcinogenesis/metabolism , Cell Transformation, Neoplastic/metabolism , Epithelial-Mesenchymal Transition/physiology , Prostate/metabolism , Prostatic Neoplasms/metabolism , Animals , Carcinogenesis/genetics , Carcinogenesis/pathology , Cell Line, Tumor , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/pathology , Humans , Male , Prostate/pathology , Prostatic Neoplasms/genetics , Prostatic Neoplasms/pathology
13.
Oncotarget ; 5(9): 2462-74, 2014 May 15.
Article in English | MEDLINE | ID: mdl-24770864

ABSTRACT

Epithelial-to-mesenchymal transition (EMT) plays an important role in prostate cancer (PCa) metastasis. The transcription/translation regulatory Y-box binding protein-1 (YB-1) is known to be associated with cancer metastasis. We observed that YB-1 expression increased with tumor grade and showed an inverse relationship with E-cadherin in a human PCa tissue array. Forced YB-1 expression induced a mesenchymal morphology that was associated with down regulation of epithelial markers. Silencing of YB-1 reversed mesenchymal features and decreased cell proliferation, migration and invasion in PCa cells. YB-1 is activated directly via Akt mediated phosphorylation at Ser102 within the cold shock domain (CSD). We next identified fisetin as an inhibitor of YB-1 activation. Computational docking and molecular dynamics suggested that fisetin binds on the residues from ß1 - ß4 strands of CSD, hindering Akt's interaction with YB-1. Calculated free binding energy ranged from -11.9845 to -9.6273 kcal/mol. Plasmon Surface Resonance studies showed that fisetin binds to YB-1 with an affinity of approximately 35 µM, with both slow association and dissociation. Fisetin also inhibited EGF induced YB-1 phosphorylation and markers of EMT both in vitro and in vivo. Collectively our data suggest that YB-1 induces EMT in PCa and identify fisetin as an inhibitor of its activation.


Subject(s)
Cell Movement/drug effects , Epithelial-Mesenchymal Transition/drug effects , Flavonoids/pharmacology , Prostate/cytology , Prostatic Neoplasms/pathology , Y-Box-Binding Protein 1/antagonists & inhibitors , Y-Box-Binding Protein 1/metabolism , Animals , Apoptosis/drug effects , Blotting, Western , Cell Adhesion/drug effects , Cell Proliferation/drug effects , Flavonols , Fluorescent Antibody Technique , Humans , Immunoenzyme Techniques , Male , Mice , Mice, Nude , Neoplasm Grading , Phosphorylation/drug effects , Prostate/drug effects , Prostate/metabolism , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/metabolism , RNA, Messenger/genetics , RNA, Small Interfering/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Tissue Array Analysis , Tumor Cells, Cultured , Xenograft Model Antitumor Assays , Y-Box-Binding Protein 1/genetics
14.
Mol Cancer Ther ; 13(2): 275-84, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24435445

ABSTRACT

Natural compounds that target microtubules and disrupt the normal function of the mitotic spindle have proven to be one of the best classes of cancer chemotherapeutic drugs available in clinics to date. There is increasing evidence showing that even minor alteration of microtubule dynamics can engage the spindle checkpoint, arresting cell-cycle progression at mitosis and subsequently leading to cell death. Our improved understanding of tumor biology and our continued appreciation for what the microtubule targeting agents (MTAs) can do have helped pave the way for a new era in the treatment of cancer. The effectiveness of these agents for cancer therapy has been impaired, however, by various side effects and drug resistance. Several new MTAs have shown potent activity against the proliferation of various cancer cells, including resistance to the existing MTAs. Sustained investigation of the mechanisms of action of MTAs, development and discovery of new drugs, and exploring new treatment strategies that reduce side effects and circumvent drug resistance could provide more effective therapeutic options for patients with cancer. This review focuses on the successful cancer chemotherapy from natural compounds in clinical settings and the challenges that may abort their usefulness.


Subject(s)
Antineoplastic Agents/therapeutic use , Biological Products/therapeutic use , Microtubules/metabolism , Neoplasms/drug therapy , Tubulin Modulators/therapeutic use , Antineoplastic Agents/adverse effects , Biological Products/adverse effects , Cell Cycle Checkpoints/drug effects , Humans , Models, Biological , Neoplasms/metabolism , Treatment Outcome , Tubulin Modulators/adverse effects
15.
Top Curr Chem ; 329: 203-20, 2013.
Article in English | MEDLINE | ID: mdl-22790416

ABSTRACT

Cancer is considered a disease of aging since the risk for developing the disease considerably increases with age. It is estimated that 77% of all cancers are diagnosed in people who fall within the age group of 55 or older. Also, it takes several years from initiation to the development of detectable cancer. One advantage of the long latency is that it provides numerous opportunities for intervention. While intervention approaches cannot be geared towards a whole population, they can nevertheless be directed towards a defined group of people who have a greater relative risk for developing the disease. The idea of cancer prevention through the use of nontoxic agents, preferably from dietary sources, has therefore emerged as an appropriate strategy for controlling the disease. An important aspect of chemoprevention is that agents can be designed for intervention at any stage during the multistage process of carcinogenesis. This process of slowing the progression of cancer is applicable to many cancers with long latency, including prostate cancer. Over the past two decades we have put considerable effort into identifying dietary substances in the form of extracts and pure compounds that can be used for the prevention of prostate and other cancers. Although cancer chemoprevention has proven to be a successful strategy in animals and, to some extent, we can say that the mission has been accomplished, its application to humans has met with limited success. This chapter will discuss various challenges associated with chemoprevention of cancer with the focus on studies with green tea and prostate cancer.


Subject(s)
Chemoprevention , Neoplasms/prevention & control , Animals , Humans , Middle Aged
16.
Curr Drug Targets ; 13(14): 1831-41, 2012 Dec.
Article in English | MEDLINE | ID: mdl-23140293

ABSTRACT

Nontoxic naturally occurring compounds, especially those from dietary sources, are receiving increasing consideration for prevention and treatment of diseases including cancer. There is a growing need for innovative anticancer therapies and therefore search for natural compounds with novel biological activities or antineoplastic potential is currently an important area in drug discovery. Support for this interest also comes from increasing concern over the efficacy and safety of many conventional therapies, especially those that run over a long course of time. Laboratory studies in different in vitro and in vivo systems have shown that many natural compounds possess the capacity to regulate response to oxidative stress and DNA damage, suppress angiogenesis, inhibit cell proliferation and induce autophagy and apoptosis. This review discusses the induction of apoptosis and autophagy as a mechanism of cancer prevention by some of the most studied naturally occurring dietary compounds.


Subject(s)
Antineoplastic Agents, Phytogenic/therapeutic use , Apoptosis/drug effects , Autophagy/drug effects , Biological Products/therapeutic use , Diet/trends , Neoplasms/prevention & control , Animals , Antineoplastic Agents, Phytogenic/chemistry , Antineoplastic Agents, Phytogenic/pharmacology , Apoptosis/physiology , Autophagy/physiology , Biological Products/chemistry , Biological Products/pharmacology , Chemoprevention/methods , Chemoprevention/trends , Diet/methods , Humans , Neoplasms/drug therapy , Neoplasms/pathology , Plant Extracts/chemistry , Plant Extracts/pharmacology , Plant Extracts/therapeutic use
17.
Biochem Pharmacol ; 84(10): 1277-81, 2012 Nov 15.
Article in English | MEDLINE | ID: mdl-22842629

ABSTRACT

Epidemiologic and case control population based studies over the past few decades have identified diet as an important determinant of cancer risk. This evidence has kindled interest into research on bioactive food components and has till date resulted in the identification of many compounds with cancer preventive and therapeutic potential. Among such compounds has been fisetin (3,7,3',4'-tetrahydroxyflavone), a flavonol and a member of the flavonoid polyphenols that also include quercetin, myricetin and kaempferol. Fisetin is commonly found in many fruits and vegetables such as apples, persimmons, grapes, kiwis, strawberries, onions and cucumbers. We evaluated the effects of fisetin against melanoma and cancers of the prostate, pancreas and the lungs. Using prostate and lung adenocarcinoma cells, we demonstrated that fisetin acts as a dual inhibitor of the PI3K/Akt and the mTOR pathways. This is a significant finding considering the fact that mTOR is phosphorylated and its activation is more frequent in tumors with overexpression of PI3K/Akt. Dual inhibitors of PI3K/Akt and mTOR signaling have been suggested as valuable agents for treating such cancers. Here, we summarize our findings on the dietary flavonoid fisetin and its effects on cancer with particular focus on prostate cancer. Our observations and findings from other laboratories suggest that fisetin could be a useful chemotherapeutic agent that could be used either alone or as an adjuvant with conventional chemotherapeutic drugs for the management of prostate and other cancers.


Subject(s)
Flavonoids/pharmacology , Phosphoinositide-3 Kinase Inhibitors , Prostatic Neoplasms/drug therapy , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , TOR Serine-Threonine Kinases/antagonists & inhibitors , Animals , Cell Line, Tumor , Flavonoids/therapeutic use , Flavonols , Fruit , Humans , Male , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Vegetables
18.
Carcinogenesis ; 33(3): 644-51, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22198212

ABSTRACT

We earlier provided evidence that oral consumption of pomegranate fruit extract (PFE) inhibits prostate cancer (PCa) cell growth in nude mice. To ascertain convincing evidence of chemopreventive effects of PFE against PCa, its efficacy requires to be evaluated in animal models that closely emulate human disease. Here, we provide evidence of remarkable tumor growth inhibitory effects of PFE using the TRAMP model. Mice received 0.1 and 0.2% PFE, equivalent to 250 and 500 ml of pomegranate juice, in drinking water, starting at 6 weeks and examined at 12, 20 and 34 weeks of age. In water-fed group, 100% mice developed palpable tumors by 20 weeks compared with only 30 and 20% in the 0.1 and 0.2% PFE-supplemented groups, respectively. At 34 weeks, palpable tumors were observed in 70 of 0.1% and only 50 of 0.2% PFE-supplemented mice. Compared with median survival of 43 weeks in water-fed mice, 0.1 and 0.2% PFE-supplemented mice exhibited median life expectancy of 73 and 92 weeks, respectively. Compared with respective water-fed groups, none of the mice in PFE-supplemented groups exhibited metastases to any of the distant organs at 20 weeks and only 20% mice exhibited metastasis at 34 weeks of age. Many of the PFE-supplemented animals had multiple foci of well-differentiated carcinoma but no evidence of poorly differentiated carcinoma. PFE supplementation resulted in simultaneous and significant inhibition of IGF-I/Akt/mTOR pathways in the prostate tissues and tumors. We suggest that pomegranate juice be evaluated in clinical trials in patients at high risk for developing PCa.


Subject(s)
Anticarcinogenic Agents/administration & dosage , Lythraceae , Phytotherapy/methods , Plant Extracts/administration & dosage , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/prevention & control , Animals , Anticarcinogenic Agents/pharmacology , Anticarcinogenic Agents/therapeutic use , Cell Transformation, Neoplastic/drug effects , Insulin-Like Growth Factor I/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Neoplasm Metastasis , Plant Extracts/pharmacology , Plant Extracts/therapeutic use , Prostate/drug effects , Prostate/pathology , Prostatic Neoplasms/pathology , Proto-Oncogene Proteins c-akt/metabolism , Random Allocation , TOR Serine-Threonine Kinases/metabolism
19.
Endocr Relat Cancer ; 17(1): R39-52, 2010 Mar.
Article in English | MEDLINE | ID: mdl-19926708

ABSTRACT

Accumulating data clearly indicate that induction of apoptosis is an important event for chemoprevention of cancer by naturally occurring dietary agents. In mammalian cells, apoptosis has been divided into two major pathways: the extrinsic pathway, activated by pro-apoptotic receptor signals at the cellular surface; and the intrinsic pathway, which involves the disruption of mitochondrial membrane integrity. This process is strictly controlled in response to integrity of pro-death signaling and plays critical roles in development, maintenance of homeostasis, and host defense in multicellular organisms. For chemoprevention studies, prostate cancer (PCa) represents an ideal disease due to its long latency, its high incidence, tumor marker availability, and identifiable preneoplastic lesions and risk groups. In this article, we highlight the studies of various apoptosis-inducing dietary compounds for prevention of PCa in vitro in cell culture, in preclinical studies in animals, and in human clinical trials.


Subject(s)
Adenocarcinoma/diet therapy , Adenocarcinoma/prevention & control , Apoptosis , Phytotherapy , Prostatic Neoplasms/diet therapy , Prostatic Neoplasms/prevention & control , Adenocarcinoma/pathology , Aged , Animals , Carotenoids/pharmacology , Carotenoids/therapeutic use , Catechin/analogs & derivatives , Catechin/pharmacology , Catechin/therapeutic use , Clinical Trials as Topic , Curcumin/pharmacology , Curcumin/therapeutic use , Drug Screening Assays, Antitumor , Flavonoids/pharmacology , Flavonoids/therapeutic use , Flavonols , Genistein/pharmacology , Genistein/therapeutic use , Humans , Lycopene , Lythraceae , Male , Mice , Mice, Nude , Mice, Transgenic , Middle Aged , Neoplasm Proteins/antagonists & inhibitors , Neoplasm Proteins/metabolism , Pentacyclic Triterpenes/pharmacology , Pentacyclic Triterpenes/therapeutic use , Plant Extracts/administration & dosage , Plant Extracts/therapeutic use , Prostatic Neoplasms/pathology , Resveratrol , Stilbenes/pharmacology , Stilbenes/therapeutic use , Tumor Cells, Cultured/drug effects
20.
Int J Cancer ; 125(10): 2465-73, 2009 Nov 15.
Article in English | MEDLINE | ID: mdl-19670328

ABSTRACT

Death receptors of the tumor necrosis factor (TNF) receptor super family have been implicated in constitutive activation of nuclear factor-kappa B (NF-kappaB) in pancreatic cancer (PaC) cells. In this study, we demonstrate that fisetin, a natural flavonoid, induces apoptosis and inhibits invasion of chemoresistant PaC AsPC-1 cells through suppression of DR3-mediated NF-kappaB activation. Fisetin treatment resulted in dose-dependent inhibition of PaC cell growth and cell proliferation with concomitant induction of apoptosis. A cDNA array analysis revealed that fisetin modulates expression of more than 20 genes at transcription level with maximum decrease observed in DR3 expression and a parallel increase observed in the expression levels of IkappaBalpha, an NF-kappaB inhibitor. Down-regulation of DR3 in PaC cells was found to down regulate activated pNF-kappaB/p65, pIkBalpha/beta kinases (pIKK's), MMP9 and XIAP that mostly impart chemoresistance in PaC. Immunoblotting and EMSA analysis showed a marked decrease in pNF-kappaB and NF-kappaB DNA binding activity, respectively, with modest decrease in NF-kappaB promoter activity and significant decrease in MMP9 promoter activity with fisetin treatment. Importantly, consistent with these findings, we further found that transient down-regulation of DR3 by RNA interference significantly augmented fisetin induced changes in cell proliferation, cell invasion and apoptosis paralleled with decrease in pNF-kappaB, pIKKalpha/beta, MMP9, XIAP and NF-kappaB DNA binding activity. Blocking of DR3 receptor with an extra cellular domain blocking antibody demonstrated similar effects. These data provide evidence that fisetin could provide a biological rationale for treatment of pancreatic cancer or as an adjuvant with conventional therapeutic regimens.


Subject(s)
Drug Resistance, Neoplasm , Flavonoids/pharmacology , NF-kappa B/antagonists & inhibitors , Pancreatic Neoplasms/drug therapy , Pancreatic Neoplasms/metabolism , Receptors, Tumor Necrosis Factor, Member 25/metabolism , Apoptosis/drug effects , Blotting, Western , Cell Line, Tumor , Cell Proliferation/drug effects , Electrophoretic Mobility Shift Assay , Flavonols , Gene Expression Profiling , Humans , I-kappa B Proteins/metabolism , Matrix Metalloproteinase 9/metabolism , NF-KappaB Inhibitor alpha , NF-kappa B/genetics , NF-kappa B/metabolism , Neoplasm Invasiveness , Oligonucleotide Array Sequence Analysis , Pancreatic Neoplasms/pathology , Promoter Regions, Genetic , RNA, Small Interfering/pharmacology , Receptors, Tumor Necrosis Factor, Member 25/antagonists & inhibitors , Receptors, Tumor Necrosis Factor, Member 25/genetics , Signal Transduction , X-Linked Inhibitor of Apoptosis Protein/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL