Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 90
Filter
1.
J Tradit Complement Med ; 12(3): 287-301, 2022 May.
Article in English | MEDLINE | ID: mdl-35493312

ABSTRACT

Background and aim: Metabolic syndrome (MetS) is a complex disease of physiological imbalances interrelated to abnormal metabolic conditions, such as abdominal obesity, type II diabetes, dyslipidemia and hypertension. In the present pilot study, we investigated the nutraceutical bitter melon (Momordica charantia L) -intake induced transcriptome and metabolome changes and the converging metabolic signaling networks underpinning its inhibitory effects against MetS-associated risk factors. Experimental procedure: Metabolic effects of lyophilized bitter melon juice (BMJ) extract (oral gavage 200 mg/kg/body weight-daily for 40 days) intake were evaluated in diet-induced obese C57BL/6J male mice [fed-high fat diet (HFD), 60 kcal% fat]. Changes in a) serum levels of biochemical parameters, b) gene expression in the hepatic transcriptome (microarray analysis using Affymetrix Mouse Exon 1.0 ST arrays), and c) metabolite abundance levels in lipid-phase plasma [liquid chromatography mass spectrometry (LC-MS)-based metabolomics] after BMJ intervention were assessed. Results and conclusion: BMJ-mediated changes showed a positive trend towards enhanced glucose homeostasis, vitamin D metabolism and suppression of glycerophospholipid metabolism. In the liver, nuclear peroxisome proliferator-activated receptor (PPAR) and circadian rhythm signaling, as well as bile acid biosynthesis and glycogen metabolism targets were modulated by BMJ (p < 0.05). Thus, our in-depth transcriptomics and metabolomics analysis suggests that BMJ-intake lowers susceptibility to the onset of high-fat diet associated MetS risk factors partly through modulation of PPAR signaling and its downstream targets in circadian rhythm processes to prevent excessive lipogenesis, maintain glucose homeostasis and modify immune responses signaling.

2.
Mol Carcinog ; 59(10): 1227-1240, 2020 10.
Article in English | MEDLINE | ID: mdl-32816368

ABSTRACT

Chemoresistance to gemcitabine (GEM)-a frontline chemotherapeutic, resulting from its dysfunctional uptake and metabolism in cancer cells, is a major contributing factor for failed therapy in pancreatic cancer (PanC) patients. Therefore, there is an urgent need for agents that could reverse GEM resistance and allow continued chemosensitivity to the drug. We employed natural nontoxic agent (with anti-PanC potential) bitter melon juice (BMJ) and GEM to examine their combinatorial benefits against tumorigenesis of PanC patient-derived xenograft (PDX)-pancreatic ductal adenocarcinomas explants PDX272 (wild-type KRAS), PDX271 (mutant KRAS and SMAD4), and PDX266 (mutant KRAS). Anti-PanC efficacy of single agents vs combination in the three tumor explants, both at the end of active dosing regimen and following a drug-washout phase were compared. In animal studies, GEM alone treatment significantly inhibited PDX tumor growth, but effects were not sustained, as GEM-treated tumors exhibited regrowth posttreatment termination. However, combination-regimen displayed enhanced and sustained efficacy. Mechanistic assessments revealed that overcoming GEM resistance by coadministration with BMJ was possibly due to modulation of GEM transport/metabolism pathway molecules (ribonucleotide reductase regulatory subunit M1, human equilibrative nucleoside transporter 1, and deoxycytidine kinase). Study outcomes, highlighting significantly higher and sustained efficacy of GEM in combination with BMJ, make a compelling case for a clinical trial in PanC patients, wherein BMJ could be combined with GEM to target and overcome GEM resistance. In addition, given their specific effectiveness against KRAS-mutant tumors, this combination could be potentially beneficial to a broader PanC patient population.


Subject(s)
Carcinoma, Pancreatic Ductal/drug therapy , Deoxycytidine/analogs & derivatives , Disease Models, Animal , Drug Resistance, Neoplasm/drug effects , Momordica charantia/chemistry , Pancreatic Neoplasms/drug therapy , Plant Extracts/pharmacology , Animals , Antimetabolites, Antineoplastic/pharmacology , Apoptosis , Carcinoma, Pancreatic Ductal/pathology , Cell Proliferation , Deoxycytidine/pharmacology , Female , Humans , Mice , Mice, Nude , Neoplasm Invasiveness , Pancreatic Neoplasms/pathology , Tumor Cells, Cultured , Xenograft Model Antitumor Assays , Gemcitabine
3.
J Tradit Complement Med ; 10(3): 236-244, 2020 May.
Article in English | MEDLINE | ID: mdl-32670818

ABSTRACT

Skin is the largest human organ that shields the inner body from contact with xenobiotic and genotoxic agents, and in this process, the skin's cellular genome faces continuous stress due to direct exposure to these noxious factors. Accumulation of genetic stress results in genomic alterations leading to undesirable gene or protein alteration/expression in skin cells, which eventually causes the formation of non-melanoma skin cancers (NMSCs). Ultraviolet B (UVB) radiation from sun is the most prominent factor contributing to ∼5 million skin cancer cases (which are mostly NMSCs) in the United States (US) and western countries. UVB exposure causes aberrations in a range of biochemical and molecular pathways such as: thymine dimer formation, DNA damage, oxidative stress, inflammatory responses, altered cellular signaling, which ultimately contribute to the development of NMSCs. The focus of this review is to summarize the protective and preventive potential of silymarin and/or silibinin against UVB-induced NMSC in pre-clinical skin cancer studies. Over two decades of research has shown the strong potential of silibinin, a biologically active flavonolignan (crude form Silymarin) derived from milk thistle plant, against a wide range of cancers, including NMSCs. Silibinin protects against UVB-induced thymine dimer formation and in turn promotes DNA repair and/or initiates apoptosis in damaged cells via an increase in p53 levels. Additionally, silibinin has shown strong efficacy against NMSCs via its potential to target aberrant signaling pathways, and induction of anti-inflammatory responses. Overall, completed comprehensive studies suggest the potential use of silibinin to prevent and/or manage NMSCs in humans.

4.
Mol Carcinog ; 58(7): 1105-1117, 2019 07.
Article in English | MEDLINE | ID: mdl-30828884

ABSTRACT

The present study aimed to determine whether grape seed extract (GSE) procyanidin mix, and its active constituent procyanidin B2 3,3″-di-O-gallate (B2G2) have the potential to target cancer stem cells (CSCs) in prostate cancer (PCa). The CSC populations were isolated and purified based on CD44+ -α2ß1high surface markers in PCa cell lines LNCaP, C4-2B, 22Rv1, PC3, and DU145, and then subjected to prostasphere formation assays in the absence or presence of GSE or B2G2. Results indicated that at lower doses (<15 µg) , the GSE procyanidin mix produced activity in unsorted prostate cancer antigen (PCA) cells, but not in sorted; however, multiple treatments with low dose GSE over a course of time inhibited sphere formation by sorted PCA CSCs. Importantly, B2G2 demonstrated significant potential to target both unsorted and sorted CSCs at lower doses. As formation of spheroids, under specific in vitro conditions, is a measure of stemness, these results indicated the potential of both GSE and B2G2 to target the self-renewal of CSC in PCa cell lines, though B2G2 was more potent in its efficacy. Subsequent mechanistic studies revealed that both GSE procyanidins and B2G2 strongly decreased the constitutive as well as Jagged1 (Notch1 ligand)-induced activated Notch1 pathway. In totality, these in vitro studies warrant extensive dose-profiling-based assessments in vivo settings to conclusively determine the impact on CSC pool kinetics on the efficacy of both GSE and B2G2 to target PCa growth as well as tumor relapse.


Subject(s)
Anthocyanins/pharmacology , Antineoplastic Agents, Phytogenic/pharmacology , Biflavonoids/pharmacology , Catechin/pharmacology , Grape Seed Extract/pharmacology , Neoplastic Stem Cells/drug effects , Proanthocyanidins/pharmacology , Prostatic Neoplasms/drug therapy , Apoptosis/drug effects , Cell Movement/drug effects , Cell Proliferation/drug effects , Cell Survival/drug effects , Humans , Jagged-1 Protein/metabolism , Male , Neoplastic Stem Cells/pathology , PC-3 Cells , Prostate/pathology , Prostatic Neoplasms/pathology , Receptor, Notch1/metabolism , Spheroids, Cellular/drug effects , Tumor Cells, Cultured
5.
Semin Cancer Biol ; 40-41: 116-129, 2016 10.
Article in English | MEDLINE | ID: mdl-27452666

ABSTRACT

Recently, there is a paradigm shift that the whole food-derived components are not 'idle bystanders' but actively participate in modulating aberrant metabolic and signaling pathways in both healthy and diseased individuals. One such whole food from Cucurbitaceae family is 'bitter melon' (Momordica charantia, also called bitter gourd, balsam apple, etc.), which has gained an enormous attention in recent years as an alternative medicine in developed countries. The increased focus on bitter melon consumption could in part be due to several recent pre-clinical efficacy studies demonstrating bitter melon potential to target obesity/type II diabetes-associated metabolic aberrations as well as its pre-clinical anti-cancer efficacy against various malignancies. The bioassay-guided fractionations have also classified the bitter melon chemical constituents based on their anti-diabetic or cytotoxic effects. Thus, by definition, these bitter melon constituents are at cross roads on the bioactivity parameters; they either have selective efficacy for correcting metabolic aberrations or targeting cancer cells, or have beneficial effects in both conditions. However, given the vast, though dispersed, literature reports on the bioactivity and beneficial attributes of bitter melon constituents, a comprehensive review on the bitter melon components and the overlapping beneficial attributes is lacking; our review attempts to fulfill these unmet needs. Importantly, the recent realization that there are common risk factors associated with obesity/type II diabetes-associated metabolic aberrations and cancer, this timely review focuses on the dual efficacy of bitter melon against the risk factors associated with both diseases that could potentially impact the course of malignancy to advanced stages. Furthermore, this review also addresses a significant gap in our knowledge regarding the bitter melon drug-drug interactions which can be predicted from the available reports on bitter melon effects on metabolism enzymes and drug transporters. This has important implications, given that a large proportion of individuals, taking bitter melon based supplements/phytochemical extracts/food based home-remedies, are also likely to be taking conventional therapeutic drugs at the same time. Accordingly, the comprehensively reviewed information here could be prudently translated to the clinical implications associated with any potential concerns regarding bitter melon consumption by cancer patients.


Subject(s)
Anticarcinogenic Agents/pharmacology , Antineoplastic Agents, Phytogenic/pharmacology , Momordica charantia/chemistry , Neoplasms/prevention & control , Plant Extracts/pharmacology , Animals , Anticarcinogenic Agents/therapeutic use , Antineoplastic Agents, Phytogenic/therapeutic use , Drug Screening Assays, Antitumor , Humans , Neoplasms/drug therapy , Plant Extracts/therapeutic use
6.
Oncotarget ; 7(22): 32566-78, 2016 May 31.
Article in English | MEDLINE | ID: mdl-27105525

ABSTRACT

The transcription factor NFAT1 and the oncogene MDM2 have crucial roles in breast cancer development, progression, and metastasis. We have recently discovered that NFAT1 activates MDM2 expression. Here, we identified a small molecule (named Inulanolide A) that dually inhibited both NFAT1 and MDM2 in breast cancer cells in vitro and in vivo. Unlike conventional MDM2 inhibitors, Inulanolide A (InuA) exerted its selective anticancer activity in both p53-dependent and -independent manners. InuA decreased cell proliferation and induced G2/M phase arrest and apoptosis in breast cancer cells; it also led to a decrease in MDM2, NFAT1 and proteins associated with cell proliferation, and an increase in apoptotic signal related proteins. In a mouse orthotopic model, JapA suppressed tumor growth and lung metastasis without host toxicity. Thus, InuA is a novel NFAT1 and MDM2 dual targeting agent and may be a clinical candidate for breast cancer therapy. This study also validates the effectiveness of dually targeting NFAT1 and MDM2 in breast cancer.


Subject(s)
Breast Neoplasms/drug therapy , NFATC Transcription Factors/antagonists & inhibitors , Proto-Oncogene Proteins c-mdm2/antagonists & inhibitors , Sesquiterpenes/pharmacology , A549 Cells , Animals , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Growth Processes/drug effects , Female , Humans , MCF-7 Cells , Mice , Mice, Nude , NFATC Transcription Factors/metabolism , Proto-Oncogene Proteins c-mdm2/metabolism , Signal Transduction/drug effects , Tumor Suppressor Protein p53/metabolism , Xenograft Model Antitumor Assays
7.
Phytochemistry ; 126: 4-10, 2016 Jun.
Article in English | MEDLINE | ID: mdl-27048854

ABSTRACT

Four meroterpenoids [talarolutins A-D] and one known compound [purpurquinone A] were characterized from an endophytic fungal isolate of Talaromyces minioluteus (G413), which was obtained from the leaves of the medicinal plant milk thistle [Silybum marianum (L.) Gaertn. (Asteraceae)]. The structures of talarolutins A-D were determined by the analysis of various NMR and MS techniques. The relative and absolute configuration of talarolutin A was determined by X-ray diffraction analysis. A combination of NOESY data and comparisons of ECD spectra were employed to assign the relative and absolute configuration of the other analogs. Talarolutins B-D were tested for cytotoxicity against human prostate carcinoma (PC-3) cell line, antimicrobial activity, and induction of quinone reductase; no notable bioactivity was observed in any assay.


Subject(s)
Talaromyces/chemistry , Terpenes/isolation & purification , Crystallography, X-Ray , Humans , Male , Microbial Sensitivity Tests , Silybum marianum/microbiology , Molecular Conformation , Molecular Structure , NAD(P)H Dehydrogenase (Quinone)/drug effects , Nuclear Magnetic Resonance, Biomolecular , Plant Leaves/microbiology , Prostatic Neoplasms/drug therapy , Terpenes/chemistry , Terpenes/pharmacology
8.
Sci Rep ; 6: 23135, 2016 Mar 16.
Article in English | MEDLINE | ID: mdl-26979487

ABSTRACT

Prostate cancer (PCa) is the leading malignancy among men. Importantly, this disease is mostly diagnosed at early stages offering a unique chemoprevention opportunity. Therefore, there is an urgent need to identify and target signaling molecules with higher expression/activity in prostate tumors and play critical role in PCa growth and progression. Here we report that NADPH oxidase (NOX) expression is directly associated with PCa progression in TRAMP mice, suggesting NOX as a potential chemoprevention target in controlling PCa. Accordingly, we assessed whether NOX activity in PCa cells could be inhibited by Graviola pulp extract (GPE) that contains unique acetogenins with strong anti-cancer effects. GPE (1-5 µg/ml) treatment strongly inhibited the hypoxia-induced NOX activity in PCa cells (LNCaP, 22Rv1 and PC3) associated with a decrease in the expression of NOX catalytic and regulatory sub-units (NOX1, NOX2 and p47(phox)). Furthermore, GPE-mediated NOX inhibition was associated with a strong decrease in nuclear HIF-1α levels as well as reduction in the proliferative and clonogenic potential of PCa cells. More importantly, GPE treatment neither inhibited NOX activity nor showed any cytotoxicity against non-neoplastic prostate epithelial PWR-1E cells. Overall, these results suggest that GPE could be useful in the prevention of PCa progression via inhibiting NOX activity.


Subject(s)
Adenocarcinoma/drug therapy , Antineoplastic Agents, Phytogenic/pharmacology , NADH, NADPH Oxidoreductases/metabolism , Plant Extracts/pharmacology , Prostatic Neoplasms/drug therapy , Adenocarcinoma/enzymology , Animals , Annonaceae/chemistry , Cell Hypoxia , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Humans , Male , Membrane Glycoproteins/metabolism , Mice, Transgenic , NADPH Oxidase 1 , NADPH Oxidase 2 , NADPH Oxidases/metabolism , Prostatic Neoplasms/enzymology
9.
J Nutr Biochem ; 26(12): 1641-9, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26383537

ABSTRACT

Cancer stem cells (CSCs) are shown to be responsible for initiation and progression of tumors in a variety of cancers. We previously showed that anthocyanin-containing baked purple-fleshed potato (PP) extracts (PA) suppressed early and advanced human colon cancer cell proliferation and induced apoptosis, but their effect on colon CSCs is not known. Considering the evidence of bioactive compounds, such as anthocyanins, against cancers, there is a critical need to study anticancer activity of PP, a global food crop, against colon CSCs. Thus, isolated colon CSCs (positive for CD44, CD133 and ALDH1b1 markers) with functioning p53 and shRNA-attenuated p53 were treated with PA at 5.0 µg/ml. Effects of baked PP (20% wt/wt) against colon CSCs were also tested in vivo in mice with azoxymethane-induced colon tumorigenesis. Effects of PA/PP were compared to positive control sulindac. In vitro, PA suppressed proliferation and elevated apoptosis in a p53-independent manner in colon CSCs. PA, but not sulindac, suppressed levels of Wnt pathway effector ß-catenin (a critical regulator of CSC proliferation) and its downstream proteins (c-Myc and cyclin D1) and elevated Bax and cytochrome c, proteins-mediating mitochondrial apoptosis. In vivo, PP reduced the number of crypts containing cells with nuclear ß-catenin (an indicator of colon CSCs) via induction of apoptosis and suppressed tumor incidence similar to that of sulindac. Combined, our data suggest that PP may contribute to reduced colon CSCs number and tumor incidence in vivo via suppression of Wnt/ß-catenin signaling and elevation of mitochondria-mediated apoptosis.


Subject(s)
Anthocyanins/chemistry , Colonic Neoplasms/metabolism , Neoplastic Stem Cells/metabolism , Solanum tuberosum/chemistry , Animals , Antineoplastic Agents/chemistry , Apoptosis , Azoxymethane/chemistry , Carcinogenesis , Cell Line, Tumor , Cell Proliferation , Cell Survival , Cell Transformation, Neoplastic/metabolism , Colonic Neoplasms/diet therapy , Colonic Neoplasms/prevention & control , Cytochromes c/metabolism , Food , Humans , In Situ Nick-End Labeling , Lentivirus , Male , Mice , Mitochondria/metabolism , Neoplastic Stem Cells/cytology , RNA, Small Interfering/metabolism , Sulindac/chemistry , Tumor Suppressor Protein p53/metabolism , Wnt Proteins/metabolism , bcl-2-Associated X Protein/metabolism , beta Catenin/metabolism
10.
Mol Carcinog ; 54(9): 688-97, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26053157

ABSTRACT

Basal cell carcinoma (BCC) is a major health problem with approximately 2.8 million new cases diagnosed each year in the United States. BCC incidences have continued to rise due to lack of effective chemopreventive options. One of the key molecular characteristics of BCC is the sustained activation of hedgehog signaling through inactivating mutations in the tumor suppressor gene patch (Ptch) or activating mutations in Smoothened. In the past, several studies have addressed targeting the activated hedgehog pathway for the treatment and prevention of BCC, although with toxic effects. Other studies have attempted BCC chemoprevention through targeting the promotional phase of the disease especially the inflammatory component. The compounds that have been utilized in pre-clinical and/or clinical studies include green and black tea, difluoromethylornithine, thymidine dinucleotide, retinoids, non-steroidal anti-inflammatory drugs, vitamin D3, and silibinin. In this review, we have discussed genetic and epigenetic modifications that occur during BCC development as well as the current state of BCC pre-clinical and clinical chemoprevention studies.


Subject(s)
Biological Products/therapeutic use , Carcinoma, Basal Cell/prevention & control , Skin Neoplasms/prevention & control , Animals , Antioxidants/therapeutic use , Carcinoma, Basal Cell/genetics , Carcinoma, Basal Cell/metabolism , Carcinoma, Basal Cell/pathology , Hedgehog Proteins/metabolism , Humans , Silybin , Silymarin/therapeutic use , Skin/drug effects , Skin/metabolism , Skin/pathology , Skin Neoplasms/genetics , Skin Neoplasms/metabolism , Skin Neoplasms/pathology
11.
Mycology ; 6(1): 8-27, 2015 Jan 02.
Article in English | MEDLINE | ID: mdl-26000195

ABSTRACT

Use of the herb milk thistle (Silybum marianum) is widespread, and its chemistry has been studied for over 50 years. However, milk thistle endophytes have not been studied previously for their fungal and chemical diversity. We examined the fungal endophytes inhabiting this medicinal herb to determine: (1) species composition and phylogenetic diversity of fungal endophytes; (2) chemical diversity of secondary metabolites produced by these organisms; and (3) cytotoxicity of the pure compounds against the human prostate carcinoma (PC-3) cell line. Forty-one fungal isolates were identified from milk thistle comprising 25 operational taxonomic units based on BLAST search via GenBank using published authentic sequences from nuclear ribosomal internal transcribed spacer sequence data. Maximum likelihood analyses of partial 28S rRNA gene showed that these endophytes had phylogenetic affinities to four major classes of Ascomycota, the Dothideomycetes, Sordariomycetes, Eurotiomycetes, and Leotiomycetes. Chemical studies of solid-substrate fermentation cultures led to the isolation of four new natural products. In addition, 58 known secondary metabolites, representing diverse biosynthetic classes, were isolated and characterized using a suite of nuclear magnetic resonance and mass spectrometry techniques. Selected pure compounds were tested against the PC-3 cell line, where six compounds displayed cytotoxicity.

12.
Toxicol Appl Pharmacol ; 285(1): 71-8, 2015 May 15.
Article in English | MEDLINE | ID: mdl-25791923

ABSTRACT

Currently, there is no effective antidote to prevent skin injuries by sulfur mustard (SM) and nitrogen mustard (NM), which are vesicating agents with potential relevance to chemical warfare, terrorist attacks, or industrial/laboratory accidents. Our earlier report has demonstrated the therapeutic efficacy of silibinin, a natural flavanone, in reversing monofunctional alkylating SM analog 2-chloroethyl ethyl sulfide-induced toxic effects in mouse skin. To translate this effect to a bifunctional alkylating vesicant, herein, efficacy studies were carried out with NM. Topical application of silibinin (1 or 2mg) 30 min after NM exposure on the dorsal skin of male SKH-1 hairless mice significantly decreased NM-induced toxic lesions at 24, 72 or 120 h post-exposure. Specifically, silibinin treatment resulted in dose-dependent reduction of NM-induced increase in epidermal thickness, dead and denuded epidermis, parakeratosis and microvesication. Higher silibinin dose also caused a 79% and 51%reversal in NM-induced increases in myeloperoxidase activity and COX-2 levels, respectively. Furthermore, silibinin completely prevented NM-induced H2A.X phosphorylation, indicating reversal of DNA damage which could be an oxidative DNA damage as evidenced by high levels of 8-oxodG in NM-exposed mouse skin that was significantly reversed by silibinin. Together, these findings suggest that attenuation of NM-induced skin injury by silibinin is due to its effects on the pathways associated with DNA damage, inflammation, vesication and oxidative stress. In conclusion, results presented here support the optimization of silibinin as an effective treatment of skin injury by vesicants.


Subject(s)
Antidotes/pharmacology , Irritants/toxicity , Mechlorethamine/toxicity , Silymarin/pharmacology , Skin/drug effects , 8-Hydroxy-2'-Deoxyguanosine , Administration, Cutaneous , Animals , Antidotes/administration & dosage , Apoptosis/drug effects , Cyclooxygenase 2/metabolism , Cytoprotection , DNA Damage , Deoxyguanosine/analogs & derivatives , Deoxyguanosine/metabolism , Dose-Response Relationship, Drug , Histones/metabolism , Male , Mice, Hairless , Necrosis , Oxidative Stress/drug effects , Peroxidase/metabolism , Phosphorylation , Signal Transduction/drug effects , Silybin , Silymarin/administration & dosage , Skin/metabolism , Skin/pathology , Time Factors
13.
Int J Oncol ; 46(4): 1849-57, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25672620

ABSTRACT

Pancreatic cancer (PanC) is one of the most lethal malignancies, and resistance towards gemcitabine, the front-line chemotherapy, is the main cause for dismal rate of survival in PanC patients; overcoming this resistance remains a major challenge to treat this deadly malignancy. Whereas several molecular mechanisms are known for gemcitabine resistance in PanC cells, altered metabolism and bioenergetics are not yet studied. Here, we compared metabolic and bioenergetic functions between gemcitabine-resistant (GR) and gemcitabine-sensitive (GS) PanC cells and underlying molecular mechanisms, together with efficacy of a natural agent bitter melon juice (BMJ). GR PanC cells showed distinct morphological features including spindle-shaped morphology and a decrease in E-cadherin expression. GR cells also showed higher ATP production with an increase in oxygen consumption rate (OCR) and extracellular acidification rate (ECAR). Molecular studies showed higher expression of glucose transporters (GLUT1 and 4) suggesting an increase in glucose uptake by GR cells. Importantly, GR cells showed a significant increase in Akt and ERK1/2 phosphorylation and their inhibition decreased cell viability, suggesting their role in survival and drug resistance of these cells. Recently, we reported strong efficacy of BMJ against a panel of GS cells in culture and nude mice, which we expanded here and found that BMJ was also effective in decreasing both Akt and ERK1/2 phosphorylation and viability of GR PanC cells. Overall, we have identified novel mechanisms of gemcitabine resistance in PanC cells which are targeted by BMJ. Considering the short survival in PanC patients, our findings could have high translational potential in controlling this deadly malignancy.


Subject(s)
Antineoplastic Agents/pharmacology , Drug Resistance, Neoplasm/drug effects , MAP Kinase Signaling System/drug effects , Momordica charantia/chemistry , Pancreatic Neoplasms/drug therapy , Plant Extracts/pharmacology , Cadherins/metabolism , Cell Line, Tumor , Cell Survival/drug effects , Deoxycytidine/analogs & derivatives , Deoxycytidine/pharmacology , Gene Expression Regulation, Neoplastic/drug effects , Humans , Oxygen Consumption/drug effects , Pancreatic Neoplasms/metabolism , Gemcitabine
14.
Mol Carcinog ; 54(12): 1734-47, 2015 Dec.
Article in English | MEDLINE | ID: mdl-25557495

ABSTRACT

Head and neck squamous cell carcinoma (HNSCC) is a major killer worldwide and innovative measures are urgently warranted to lower the morbidity and mortality caused by this malignancy. Aberrant redox and metabolic status in HNSCC cells offer a unique opportunity to specifically target cancer cells. Therefore, we investigated the efficacy of grape seed extract (GSE) to target the redox and bioenergetic alterations in HNSCC cells. GSE treatment decreased the mitochondrial electron transport chain complex III activity, increased the mitochondrial superoxide levels and depleted the levels of cellular antioxidant (glutathione), thus resulting in the loss of mitochondrial membrane potential in human HNSCC Detroit 562 and FaDu cells. Polyethylene glycol-SOD addition reversed the GSE-mediated apoptosis without restoring complex III activity. Along with redox changes, GSE inhibited the extracellular acidification rate (representing glycolysis) and oxygen consumption rate (indicating oxidative phosphorylation) leading to metabolic stress in HNSCC cells. Molecular studies revealed that GSE activated AMP-activated protein kinase (AMPK), and suppressed Akt/mTOR/4E-BP1/S6K signaling in both Detroit 562 and FaDu cells. Interestingly, GSE increased the autophagic load specifically in FaDu cells, and autophagy inhibition significantly augmented the apoptosis in these cells. Consistent with in vitro results, in vivo analyses also showed that GSE feeding in nude mice activated AMPK and induced-autophagy in FaDu xenograft tumor tissues. Overall, these findings are innovative as we for the first time showed that GSE targets ETC complex III and induces oxidative and metabolic stress, thereby, causing autophagy and apoptotic death in HNSCC cells.


Subject(s)
Autophagy/drug effects , Electron Transport Complex III/metabolism , Grape Seed Extract/pharmacology , Head and Neck Neoplasms/drug therapy , Mitochondria/drug effects , Oxidative Stress/drug effects , Stress, Physiological/drug effects , AMP-Activated Protein Kinases/metabolism , Animals , Apoptosis/drug effects , Carcinoma, Squamous Cell/drug therapy , Carcinoma, Squamous Cell/metabolism , Cytoprotection/drug effects , Head and Neck Neoplasms/metabolism , Humans , Mice , Mice, Nude , Mitochondria/metabolism , Mitochondrial Membranes/drug effects , Mitochondrial Membranes/metabolism , Oxidation-Reduction/drug effects , Phosphorylation/drug effects , Signal Transduction/drug effects , Squamous Cell Carcinoma of Head and Neck , Tumor Cells, Cultured
15.
Mol Carcinog ; 54(4): 291-300, 2015 Apr.
Article in English | MEDLINE | ID: mdl-24243690

ABSTRACT

Preventive measures against oral carcinogenesis are urgently warranted to lower the high morbidity and mortality associated with this malignancy worldwide. Here, we investigated the chemopreventive efficacy of grape seed extract (GSE) and resveratrol (Res) in 4-nitroquinoline-1-oxide (4NQO)-induced tongue tumorigenesis in C57BL/6 mice. Following 8 weeks of 4NQO exposure (100 µg/ml in drinking water), mice were fed with either control AIN-76A diet or diet containing 0.2% GSE (w/w) or 0.25% Res (w/w) for 8 subsequent weeks, while continued on 4NQO. Upon termination of the study at 16 weeks, tongue tissues were histologically evaluated for hyperplasia, dysplasia, and papillary lesions, and then analyzed for molecular targets by immunohistochemistry. GSE and Res feeding for 8 weeks, moderately decreased the incidence, but significantly prevented the multiplicity and severity of 4NQO-induced preneoplastic and neoplastic lesions, without any apparent toxicity. In tongue tissues, both 4NQO + GSE and 4NQO + Res treatment correlated with a decreased proliferation (BrdU labeling index) but increased apoptotic death (TUNEL-positive cells) as compared to the 4NQO group. Furthermore, tongue tissues from both the 4NQO + GSE and 4NQO + Res groups showed an increase in activated metabolic regulator phospho-AMPK (Thr172) and decreased autophagy flux marker p62. Together, these findings suggest that GSE and Res could effectively prevent 4NQO-induced oral tumorigenesis through modulating AMPK activation, and thereby, inhibiting proliferation and inducing apoptosis and autophagy, as mechanisms of their efficacy.


Subject(s)
4-Nitroquinoline-1-oxide/toxicity , AMP-Activated Protein Kinases/metabolism , Anticarcinogenic Agents/therapeutic use , Carcinogens/toxicity , Carcinoma, Squamous Cell/prevention & control , Grape Seed Extract/therapeutic use , Stilbenes/therapeutic use , Tongue Neoplasms/prevention & control , AMP-Activated Protein Kinases/analysis , Animals , Apoptosis/drug effects , Carcinogenesis/chemically induced , Carcinogenesis/drug effects , Carcinogenesis/pathology , Carcinoma, Squamous Cell/chemically induced , Carcinoma, Squamous Cell/pathology , Female , In Situ Nick-End Labeling , Mice , Mice, Inbred C57BL , Resveratrol , Tongue/drug effects , Tongue/pathology , Tongue Neoplasms/chemically induced , Tongue Neoplasms/pathology
16.
Oncotarget ; 5(20): 10151-69, 2014 Oct 30.
Article in English | MEDLINE | ID: mdl-25294814

ABSTRACT

With global rise in obesity, it is imperative that we identify obesity-driven factors that increase growth and progression of colorectal cancer (CRC), and also discover and develop agents with anti-CRC efficacy under obese conditions. Here in, we investigated grape seed extract (GSE), a well-defined agent with both preventive and anti-CRC efficacy, for its potential to impair pro-tumorigenic signaling of adipocytes on CRC/colon cancer stem cells (CSCs) and associated molecular mechanisms, to control CRC under obese conditions. GSE treatment significantly decreased the growth and invasion promoting effects of both mouse and human adipocytes on CRC cells. Moreover, GSE exerted a direct inhibitory effect, as well as it strongly reduced the growth promoting signals of adipocytes, on colon CSCs. These GSE effects were associated with a decrease in both mRNA and protein levels of various CSC-associated molecules. Notably, GSE effects on adipocytes were not due to changes in lipid content, but by inducing the 'browning' of adipocytes as evidenced by an increase in UCP-1 mRNA level and mitochondriogenesis. Together, these findings, for the first time, suggest the ability of GSE to induce 'brown remodeling' of white adipocytes, which causes functional modification of adipocytes thus impairing their pro-tumorigenic signals on colon CSCs/CRC cells.


Subject(s)
Adipocytes/drug effects , Antineoplastic Agents, Phytogenic/pharmacology , Colorectal Neoplasms/drug therapy , Grape Seed Extract/pharmacology , Neoplastic Stem Cells/drug effects , Adipocytes/metabolism , Adipocytes/pathology , Animals , Apoptosis/drug effects , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/pathology , HCT116 Cells , HT29 Cells , Humans , Mice , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Signal Transduction/drug effects
17.
Curr Cancer Drug Targets ; 14(4): 323-36, 2014.
Article in English | MEDLINE | ID: mdl-24724981

ABSTRACT

Various natural agents, including grape seed extract (GSE), have shown considerable chemopreventive and anti-cancer efficacy against different cancers in pre-clinical studies; however, their specific protein targets are largely unknown and thus, their clinical usefulness is marred by limited scientific evidences about their direct cellular targets. Accordingly, herein, employing, for the first time, the recently developed drug affinity responsive target stability (DARTS) technique, we aimed to profile the potential protein targets of GSE in human colorectal cancer (CRC) cells. Unlike other methods, which can cause chemical alteration of the drug components to allow for detection, this approach relies on the fact that a drug bound protein may become less susceptible to proteolysis and hence the enriched proteins can be detected by Mass Spectroscopy methods. Our results, utilizing the DARTS technique followed by examination of the spectral output by LC/MS and the MASCOT data, revealed that GSE targets endoplasmic reticulum (ER) stress response proteins resulting in overall down regulation of proteins involved in translation and that GSE also causes oxidative protein modifications, specifically on methionine amino acids residues on its protein targets. Corroborating these findings, mechanistic studies revealed that GSE indeed caused ER stress and strongly inhibited PI3k-Akt-mTOR pathway for its biological effects in CRC cells. Furthermore, bioenergetics studies indicated that GSE also interferes with glycolysis and mitochondrial metabolism in CRC cells. Together, the present study identifying GSE molecular targets in CRC cells, combined with its efficacy in vast pre-clinical CRC models, further supports its usefulness for CRC prevention and treatment.


Subject(s)
Antineoplastic Agents, Phytogenic/metabolism , Colorectal Neoplasms/metabolism , Dietary Supplements , Endoplasmic Reticulum Stress , Grape Seed Extract/metabolism , Models, Biological , Neoplasm Proteins/antagonists & inhibitors , Antineoplastic Agents, Phytogenic/therapeutic use , Cell Line, Tumor , Colorectal Neoplasms/prevention & control , Down-Regulation , Energy Metabolism , Grape Seed Extract/therapeutic use , Humans , Kinetics , Methionine/chemistry , Neoplasm Proteins/chemistry , Neoplasm Proteins/metabolism , Oxidation-Reduction , Peptide Mapping , Phosphatidylinositol 3-Kinase/chemistry , Phosphatidylinositol 3-Kinase/metabolism , Phosphoinositide-3 Kinase Inhibitors , Protein Processing, Post-Translational , Protein Stability , Proteomics/methods , Signal Transduction
18.
Exp Toxicol Pathol ; 66(5-6): 235-42, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24670932

ABSTRACT

The second leading cause of cancer-related deaths (both genders combined) in the United States is colorectal cancer (CRC). This emphasizes the need to develop both effective therapies for CRC patients and pre-clinical models mimicking human disease that carry translational potential in drug-development. Notably, at present there are no in situ models of CRC metastasis to lung. In our azoxymethane-induced colon tumorigenesis study in A/J mice assessing grape seed extract (GSE) efficacy, during necropsy we also found multiple lung nodules suggestive of colon tumor metastasis to lung that were significantly inhibited in GSE fed group. Both histopathological and molecular studies were performed to characterize and establish the origin of these lesions in lung. Histologically these nodules were determined as adenocarcinoma of mucin origin. Molecular analyses by immunohistochemistry (IHC) and RT-PCR revealed strong protein and transcript levels of colon specific markers CDX2 and CK20 in these lung nodules compared to uninvolved control lung tissue. Vis-à-vis, these nodules also showed minimally expressed lung specific biomarkers, specifically surfactant D and TTF-1, in IHC analysis. Additionally, 0.25% GSE supplementation in diet (w/w) decreased the incidence of these lung nodules by 53% and their total number by 66%. Together, the characterization of this unique in situ mouse model of CRC metastasis to lung provides translational opportunities in developing effective therapies to clinically manage and treat CRC at the advanced stage. Moreover, GSE efficacy in inhibiting CRC metastasis to lung in this model further supports its translational potential in controlling CRC growth, progression and metastasis in patients.


Subject(s)
Antineoplastic Agents, Phytogenic/therapeutic use , Colonic Neoplasms/drug therapy , Grape Seed Extract/therapeutic use , Lung Neoplasms/drug therapy , Lung Neoplasms/secondary , Lung/drug effects , Animals , Antineoplastic Agents, Phytogenic/administration & dosage , Azoxymethane , Colonic Neoplasms/chemically induced , Colonic Neoplasms/pathology , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/etiology , Colorectal Neoplasms/pathology , Drug Screening Assays, Antitumor , Grape Seed Extract/administration & dosage , Humans , Lung/pathology , Lung Neoplasms/chemically induced , Lung Neoplasms/pathology , Male , Mice, Inbred Strains
19.
Mutat Res ; 760: 1-15, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24389535

ABSTRACT

Prostate cancer (PCA) is the most commonly diagnosed cancer in men in the United States with growing worldwide incidence. Despite intensive investment in improving early detection, PCA often escapes timely detection and mortality remains high; this malignancy being the second highest cancer-associated mortality in American men. Collectively, health care costs of PCA results in an immense financial burden that is only expected to grow. Additionally, even in cases of successful treatment, PCA is associated with long-term and pervasive effects on patients. A proactive alternative to treat PCA is to prevent its occurrence and progression prior to symptomatic malignancy. This may serve to address the issue of burgeoning healthcare costs and increasing number of sufferers. One potential regimen in service of this alternative is PCA chemoprevention. Here, chemical compounds with cancer preventive efficacy are identified on the basis of their potential in a host of categories: their historical medicinal use, correlation with reduced risk in population studies, non-toxicity, their unique chemical properties, or their role in biological systems. PCA chemopreventive agents are drawn from multiple broad classes of chemicals, themselves further subdivided based on source or potential effect, with most derived from natural products. Many such compounds have shown efficacy, varying from inhibiting deregulated PCA cell signaling, proliferation, epithelial to mesenchymal transition (EMT), invasion, metastasis, tumor growth and angiogenesis and inducing apoptosis. Overall, these chemopreventive agents show great promise in PCA pre-clinical models, though additional work remains to be done in effectively translating these findings into clinical use.


Subject(s)
Anticarcinogenic Agents/therapeutic use , Prostatic Neoplasms/prevention & control , Animals , Humans , Male
20.
Nutr Cancer ; 66(4): 736-46, 2014.
Article in English | MEDLINE | ID: mdl-24191894

ABSTRACT

Recently, we identified procyanidin B2 3,3(″)-di-O-gallate (B2G2) as most active constituent of grape seed extract (GSE) for efficacy against prostate cancer (PCa). Isolating large quantities of B2G2 from total GSE is labor intensive and expensive, thereby limiting both efficacy and mechanistic studies with this novel anticancer agent. Accordingly, here we synthesized gram-scale quantities of B2G2, compared it with B2G2 isolated from GSE for possible equivalent biological activity and conducted mechanistic studies. Both B2G2 preparations inhibited cell growth, decreased clonogenicity, and induced cell cycle arrest and apoptotic death, comparable to each other, in various human PCa cell lines. Mechanistic studies focusing on transcription factors involved in apoptotic and survival pathways revealed that B2G2 significantly inhibits NF-κB and activator protein1 (AP1) transcriptional activity and nuclear translocation of signal transducer and activator of transcription3 (Stat3) in PCa cell lines, irrespective of their functional androgen receptor status. B2G2 also decreased survivin expression which is regulated by NF-κB, AP1, and Stat3 and increased cleaved PARP level. In summary, we report B2G2 chemical synthesis at gram-quantity with equivalent biological efficacy against human PCa cell lines and same molecular targeting profiles at key transcription factors level. The synthetic B2G2 will stimulate more research on prostate and possibly other malignancies in preclinical models and clinical translation.


Subject(s)
Anthocyanins/pharmacology , Apoptosis/drug effects , Grape Seed Extract/pharmacology , NF-kappa B/metabolism , Receptors, Androgen/metabolism , STAT3 Transcription Factor/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival , Humans , Male , NF-kappa B/genetics , Prostatic Neoplasms/metabolism , Receptors, Androgen/genetics , STAT3 Transcription Factor/genetics
SELECTION OF CITATIONS
SEARCH DETAIL