Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 25
Filter
1.
Int J Biol Macromol ; 253(Pt 8): 127254, 2023 Dec 31.
Article in English | MEDLINE | ID: mdl-37813219

ABSTRACT

Ferroptosis is a non-apoptotic cell death pathway characterized by the accumulation of lipid-peroxy radicals within the affected cells. Here, we investigate the synergistic capacity of sorafenib (SOR) and simvastatin (SIM) to trigger ferroptosis for cancer therapy. For precise in-vivo delivery, SOR + SIM was ratiometrically loaded in bovine serum albumin nanoparticles (BSA-NPs) modified with 4-carboxy phenylboronic acid (CPBA). The developed CPBA-BSA(SOR + SIM)-NPs revealed size of 175.2 ± 12.8 nm, with PDI of 0.22 ± 0.03 and Z-potential of -29.6 ± 4.8 mV. Significantly, CPBA-BSA(SOR + SIM)-NPs exhibited > 2 and > 5-fold reduction in IC50 values compared to individual SOR and SIM treatments respectively, in all tested cell lines. Moreover, CPBA-BSA(SOR + SIM)-NPs treated cells exhibited decrease in glutathione levels, increase in malonaldehyde levels and depolarization of mitochondrial membrane potential (JC-1 assay). Pharmacokinetic analysis revealed enhanced AUC0-∞ and MRT levels for SOR and SIM when administered as CPBA-BSA(SOR + SIM)-NPs compared to free drugs. Crucially, in in-vivo experiments, CPBA-BSA(SOR + SIM)-NPs led to a significant reduction in tumor volume compared to various control groups. Histological and biomarker analyses underscore their biocompatibility for clinical applications. In conclusion, this study highlights the potential of CPBA-BSA(SOR + SIM)-NPs as a promising strategy for inducing ferroptosis in cancer cells, concurrently improving drug delivery and therapeutic efficacy. This approach opens new avenues in cancer treatment.


Subject(s)
Ferroptosis , Nanoparticles , Sorafenib/pharmacology , Serum Albumin, Bovine , Simvastatin/pharmacology , Drug Carriers/pharmacokinetics , Particle Size
2.
AAPS PharmSciTech ; 24(7): 180, 2023 Sep 11.
Article in English | MEDLINE | ID: mdl-37697085

ABSTRACT

Ferroptosis, a pathway dependent on oxygen and iron catalysts, holds promise as a therapeutic approach for cancer treatment due to its manageable regulation, direct control, and immunogenic properties. The sensitivity of cancer cells to ferroptosis induction varies based on their metabolic, genetic, and signalling pathways, prompting the use of combination therapy. In this study, we conducted a screening of drug combinations, including sorafenib (SOR) with simvastatin (SIM), phenethyl isothiocyanate, and trigonelline, in MDA-MB-231, A549, and HeLa cells to assess their cytotoxicity. The SOR-SIM combination exhibited a synergistic effect in MDA-MB-231, A549, and HeLa cells, with calculated CI values of ~ 0.66, 0.53, and 0.59, respectively. Furthermore, co-treatment with ferrostatin-1 resulted in a concentration-dependent increase in the IC50 values. Additionally, SOR + SIM demonstrated a significant reduction in GSH levels, an increase in MDA levels, and mitochondrial membrane depolarization across all three cell lines, indicating their ferroptosis inducing potential. In-vivo studies showed a significant reduction in tumor volume by 3.53-, 2.55-, and 1.47-fold compared to control, SIM, and SOR, respectively. Toxicity assessments revealed insignificant changes in biomarker levels and no observable deformations in isolated organs, except for erythrocyte shrinkage and membrane scrambling effects caused by the SOR + SIM combination. Overall, our findings highlight the potential of the SOR + SIM combination as an effective strategy for cancer treatment, emphasizing the importance of further research in targeted drug delivery systems to ensure its safety.


Subject(s)
Ferroptosis , Neoplasms , Humans , Early Detection of Cancer , HeLa Cells , Sorafenib/pharmacology , Drug Delivery Systems , Neoplasms/drug therapy
3.
Indian J Community Med ; 48(3): 471-477, 2023.
Article in English | MEDLINE | ID: mdl-37469914

ABSTRACT

Context: In 2018, Government of India initiated Home-Based Care for Young Child (HBYC) program having five quarterly structured home visits for children age 3 to 15 months to promote early childhood development. Assess knowledge and practices of Accredited Social Health Activist (ASHAs), other health functionaries, and mothers related to HBYC. Cross-sectional evaluation design with ASHAs, AWWs, ANMs, ASHA, and mothers of 3 to 15 month's children as participants. Material and Methods: Knowledge and practices of 801 ASHAs, 200 other health functionaries, and 787 mothers were assessed on exclusive breastfeeding, complementary feeding, hand washing, iron folic acid (IFA) and oral rehydration solution (ORS) supplementation, danger referral signs in eight aspirational districts of Madhya Pradesh. Results: 88% ASHAs had correct knowledge on ORS, 85% on complementary feeding, 85% on adequacy of IFA, and 47% on danger signs which required child referral. Similarly, 85% of mothers had knowledge on exclusive breastfeeding, 40% mothers knew about complementary feeding, and only 18% knew correct ORS preparation. Statistically significant association was observed between ASHAs home visits and availability of ORS with mothers and their knowledge on correct Initiation of IFA (p < 0.001). Conclusion: Findings of study confirmed majority of health functionaries were aware about roles, responsibilities, and key tasks under HBYC. However, there observed a gap in knowledge transfer by health functionaries and thus inadequate translation of knowledge into practices among mothers on HBYC. This necessitates the need of appropriate actions from health system strengthening to capacity building to accelerate uptake of HBYC program.

4.
Int J Biol Macromol ; 211: 380-389, 2022 Jun 30.
Article in English | MEDLINE | ID: mdl-35569681

ABSTRACT

Nanoparticles of green materials have gained enormous interest due to their broad range of applications in several disciplines since they have significantly improved multifunctional activities. This article attempts a sustainable green approach to synthesize sodium lignosulfonate nanoparticles (SLS NPs) using another biomolecule, i.e., chitosan. The synthesized SLS NPs (with an average diameter of ~125 nm to 129 nm) have demonstrated synergetic efficacy by exhibiting outstanding multifunctional properties due to the presence of two types of biomolecules (i.e., lignosulfonate as well as chitosan) in their structure. The synthesized SLS NPs have bestowed excellent antibacterial activity against both the Gram-positive (S. aureus) and Gram-negative (E. coli) bacteria. Moreover, SLS NPs have displayed ~92% antioxidant property. Having polyphenolic entities in the structure of SLS NPs, they have shown UV-visible absorption peak at 224 nm, which directly indicates that they can act as an outstanding UV protective agent which has also been proven experimentally.


Subject(s)
Chitosan , Metal Nanoparticles , Nanoparticles , Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/pharmacology , Chitosan/chemistry , Escherichia coli , Green Chemistry Technology , Lignin/analogs & derivatives , Metal Nanoparticles/chemistry , Microbial Sensitivity Tests , Plant Extracts/chemistry , Sodium , Staphylococcus aureus
5.
Lab Chip ; 20(22): 4273-4284, 2020 11 10.
Article in English | MEDLINE | ID: mdl-33090162

ABSTRACT

In this study, an ion depleted zone created by an ion-selective membrane was used to impose a high and uniform constant extracellular potential over an entire ∼1000 cell rat cardiomyocyte (rCM) colony on-a-chip to trigger synchronized voltage-gated ion channel activities while preserving cell viability, thus extending single-cell voltage-clamp ion channel studies to an entire normalized colony. Image analysis indicated that rCM beating was strengthened and accelerated (by a factor of ∼2) within minutes of ion depletion and the duration of contraction and relaxation phases was significantly reduced. After the initial synchronization, the entire colony responds collectively to external potential changes such that beating over the entire colony can be activated or deactivated within 0.1 s. These newly observed collective dynamic responses, due to simultaneous ion channel activation/deactivation by a uniform constant-potential extracellular environment, suggest that perm-selective membrane modules on cell culture chips can facilitate studies of extracellular cardiac cell electrical communication and how ion-channel related pathologies affect cardiac cell synchronization. The future applications of this new technology can lead to better drug screening platforms for cardiotoxicity as well as platforms that can facilitate synchronized maturation of pluripotent stem cells into colonies with high electrical connectivity.


Subject(s)
Ion Channels , Myocytes, Cardiac , Animals , Drug Evaluation, Preclinical , Rats
6.
Nat Prod Res ; 33(22): 3298-3302, 2019 Nov.
Article in English | MEDLINE | ID: mdl-29726721

ABSTRACT

Total amount of phenols, flavonoids and tannins in methanolic extract of heartwood Pterocarpus marsupium were found to be 124.4 ± 4.6, 485.5 ± 2.7, and 71.6 ± 4.2 mg/gm of plant extract respectively. HPLC analyses of extract showed retention time at 6.2 min and FTIR analysis confirmed presence of Liquiritigenin in methanolic extract. In DPPH scavenging activity and ABTS∙+ scavenging activity, IC50 values were found to be 138.3, 12.4, 13.5 and 47.8, 3.9, 4.2 µg/mL for aqueous, methanolic extract and standard ascorbic acid respectively. In α-amylase inhibition assay and α-glucosidase inhibition assay, IC50 values for standard Acarbose, aqueous and methanolic extract of heartwood were 44.09, 166.72, 48.20 and 45.17, 172.32, 48.12 respectively. Molecular docking study showed hydrogen bonding between Liquiritigenin and catalytic triad (Asp197, Glu233, and Asp300) of α-amylase and His407 of α -Glucosidase with -5.60 and -7.10 binding energies respectively.


Subject(s)
Flavonoids/isolation & purification , Hypoglycemic Agents/pharmacology , Plant Extracts/chemistry , Pterocarpus/chemistry , Tannins/isolation & purification , Antioxidants/chemistry , Computer Simulation , Flavanones/metabolism , Flavonoids/chemistry , Glycoside Hydrolase Inhibitors/chemistry , Hydrogen Bonding , Hypoglycemic Agents/isolation & purification , Molecular Docking Simulation , Phenols/analysis , Tannins/chemistry , alpha-Amylases/antagonists & inhibitors , alpha-Amylases/metabolism
7.
Microb Pathog ; 107: 136-143, 2017 Jun.
Article in English | MEDLINE | ID: mdl-28351711

ABSTRACT

Inter and intracellular communication in bacteria, which is known as quorum sensing (QS), is mediated by small diffusible signaling molecules known as autoinducers. QS regulates various virulence factors responsible for pathogenesis. Increasing resistance of microorganisms against traditional antibiotics has turned the focus towards the QS as it exerts less selective pressure preventing development of resistance among microorganisms. LasR, a transcription factor that controls QS in Pseudomonas aeruginosa, is an attractive therapeutic target for inhibitors. This study aimed to screen natural compounds as potential inhibitors of LasR. About 2603 compounds from ZINC database were virtually screened against the structure of LasR. Then after qualifying compounds were filtered on the parameters of Lipinski's rule and ADME. Six novel potential QS inhibiting compounds were selected on the basis of binding energy. Structures of LasR-ligand complexes were analysed to have insight of binding between inhibitors and target. It is pertinent to mention here that all the molecules are structurally different from 3-oxo-C12HSL,a native autoinducer of LasR, that play key role in formation of LasR dimer which is an active form of the protein to facilitate QS.


Subject(s)
4-Butyrolactone/analogs & derivatives , Bacterial Proteins/chemistry , Bacterial Proteins/drug effects , Homoserine/analogs & derivatives , Pseudomonas aeruginosa/drug effects , Quorum Sensing/drug effects , Trans-Activators/chemistry , Trans-Activators/drug effects , 4-Butyrolactone/antagonists & inhibitors , 4-Butyrolactone/chemistry , Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/pharmacology , Bacterial Proteins/metabolism , Chemical Phenomena , Drug Discovery , Drug Evaluation, Preclinical , Homoserine/antagonists & inhibitors , Homoserine/chemistry , Hydrogen Bonding , Molecular Conformation , Molecular Docking Simulation , Pseudomonas aeruginosa/metabolism , Transcription Factors/metabolism , User-Computer Interface , Virulence Factors/chemistry , Virulence Factors/metabolism
8.
Contraception ; 93(4): 347-355, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26738620

ABSTRACT

OBJECTIVE: As part of a strategy to revitalize postpartum family planning services, Government of India revised its policy in 2013 to permit trained nurses and midwives to insert postpartum intrauterine contraceptive devices (PPIUCDs). This study compares two key outcomes of PPIUCD insertions--expulsion and infection--for physicians and nurses/midwives to generate evidence for task sharing. STUDY DESIGN: We analyzed secondary data from the PPIUCD program in seven states using a case-control study design. We included facilities where both doctors and nurses/midwives performed PPIUCD insertions and where five or more cases of expulsion and/or infection were reported during the study period (January-December 2013). For each case of expulsion and infection, we identified a time-matched control who received a PPIUCD at the same facility and had no complaints. We performed a multiple logistic regression analysis focusing on provider cadre while controlling for potential confounding factors. RESULTS: In 137 facilities, 792 expulsion and 382 infection cases were matched with 1041 controls. Provider type was not significantly associated with either expulsion [odds ratio (OR) 1.84; 95% confidence interval (CI): 0.82-4.12] or infection (OR 0.73; 95% CI: 0.39-1.37). Compared with centralized training, odds of expulsion were higher for onsite (OR 2.32, 95% CI: 1.86-2.89) and on-the-job training (OR 1.23, 95% CI: 1.11-1.36), but odds of infection were lower for onsite (OR 0.45, 95% CI: 0.27-0.75) and on-the-job training (OR 0.31, 95% CI: 0.25-0.37). CONCLUSION: Trained nurses and midwives who conduct deliveries at public health facilities can perform PPIUCD insertions as safely as physicians. IMPLICATIONS: Institutional deliveries are increasing in India, but most normal vaginal deliveries at public health facilities are attended by nurses and midwives due to a shortage of physicians. Task sharing with nurses and midwives can increase women's access to and the acceptability of quality PPIUCD services.


Subject(s)
Intrauterine Devices , Postpartum Period , Adult , Case-Control Studies , Family Planning Services , Female , Humans , India , Infections/epidemiology , Intrauterine Device Expulsion , Intrauterine Devices/adverse effects , Logistic Models , Midwifery , Nurses , Physicians , Treatment Outcome , United States , United States Public Health Service
9.
PLoS One ; 10(8): e0135495, 2015.
Article in English | MEDLINE | ID: mdl-26263486

ABSTRACT

Quorum sensing (QS) is a system of stimuli and responses in bacterial cells governed by their population density, through which they regulate genes that control virulence factors and biofilm formation. Despite considerable research on QS and the discovery of new antibiotics, QS-controlled biofilm formation by microorganisms in clinical settings has remained a problem because of nascent drug resistance, which requires screening of diverse compounds for anti-QS activities. Cinnamon is a dietary phytochemical that is traditionally used to remedy digestive problems and assorted contagions, which suggests that cinnamon might contain chemicals that can hinder the QS process. To test this hypothesis, the anti-QS activity of cinnamon oil against P. aeruginosa was tested, measured by the inhibition of biofilm formation and other QS-associated phenomena, including virulence factors such as pyocyanin, rhamnolipid, protease, alginate production, and swarming activity. To this end, multiple microscopy analyses, including light, scanning electron and confocal microscopy, revealed the ability of cinnamon oil to inhibit P. aeruginosa PAO1 biofilms and their accompanying extracellular polymeric substances. This work is the first to demonstrate that cinnamon oil can influence various QS-based phenomena in P. aeruginosa PAO1, including biofilm formation.


Subject(s)
Biofilms/drug effects , Cinnamomum zeylanicum/chemistry , Plant Oils/pharmacology , Pseudomonas aeruginosa/drug effects , Pseudomonas aeruginosa/physiology , Quorum Sensing/drug effects , Anti-Bacterial Agents/pharmacology , Indoles/metabolism , Microbial Sensitivity Tests , Phenotype , Plant Extracts/pharmacology , Virulence Factors
10.
Clin Cancer Res ; 18(18): 4942-53, 2012 Sep 15.
Article in English | MEDLINE | ID: mdl-22832932

ABSTRACT

PURPOSE: Development of chemoresistance, poor prognosis, and metastasis often renders the current treatments for colorectal cancer (CRC) ineffective. Whether ursolic acid, a component of numerous medicinal plants, either alone or in combination with capecitabine, can inhibit the growth and metastasis of human CRC was investigated. EXPERIMENTAL DESIGN: The effect of ursolic acid on proliferation of CRC cell lines was examined by mitochondrial dye uptake assay, apoptosis by esterase staining, NF-κB activation by DNA-binding assay, and protein expression by Western blot. The effect of ursolic acid on the growth and chemosensitization was also examined in orthotopically implanted CRC in nude mice. RESULTS: We found that ursolic acid inhibited the proliferation of different colon cancer cell lines. This is correlated with inhibition of constitutive NF-κB activation and downregulation of cell survival (Bcl-xL, Bcl-2, cFLIP, and survivin), proliferative (cyclin D1), and metastatic (MMP-9, VEGF, and ICAM-1) proteins. When examined in an orthotopic nude mouse model, ursolic acid significantly inhibited tumor volume, ascites formation, and distant organ metastasis, and this effect was enhanced with capecitabine. Immunohistochemistry of tumor tissue indicated that ursolic acid downregulated biomarkers of proliferation (Ki-67) and microvessel density (CD31). This effect was accompanied by suppression of NF-κB, STAT3, and ß-catenin. In addition, ursolic acid suppressed EGF receptor (EGFR) and induced p53 and p21 expression. We also observed bioavailability of ursolic acid in the serum and tissue of animals. CONCLUSION: Overall, our results show that ursolic acid can inhibit the growth and metastasis of CRC and further enhance the therapeutic effects of capecitabine through the suppression of multiple biomarkers linked to inflammation, proliferation, invasion, angiogenesis, and metastasis.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Colorectal Neoplasms/drug therapy , Signal Transduction/drug effects , Triterpenes/therapeutic use , Animals , Antimetabolites, Antineoplastic/administration & dosage , Antimetabolites, Antineoplastic/pharmacology , Antimetabolites, Antineoplastic/toxicity , Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Apoptosis/drug effects , Capecitabine , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/pathology , Deoxycytidine/administration & dosage , Deoxycytidine/analogs & derivatives , Deoxycytidine/pharmacology , Deoxycytidine/toxicity , Disease Models, Animal , Drug Resistance, Neoplasm/drug effects , Drug Synergism , ErbB Receptors/metabolism , Fluorouracil/administration & dosage , Fluorouracil/analogs & derivatives , Fluorouracil/pharmacology , Fluorouracil/toxicity , Humans , Ki-67 Antigen/metabolism , Male , Mice , Mice, Nude , NF-kappa B/antagonists & inhibitors , NF-kappa B/metabolism , Neoplasm Metastasis/drug therapy , Platelet Endothelial Cell Adhesion Molecule-1/metabolism , STAT3 Transcription Factor/metabolism , Triterpenes/administration & dosage , Tumor Suppressor Proteins/metabolism , Xenograft Model Antitumor Assays , beta Catenin/metabolism , Ursolic Acid
11.
Int J Cancer ; 130(9): 2176-84, 2012 May 01.
Article in English | MEDLINE | ID: mdl-21702037

ABSTRACT

Numerous cancer therapeutics were originally identified from natural products used in traditional medicine. One such agent is acetyl-11-keto-beta-boswellic acid (AKBA), derived from the gum resin of the Boswellia serrata known as Salai guggal or Indian frankincense. Traditionally, it has been used in Ayurvedic medicine to treat proinflammatory conditions. In this report, we hypothesized that AKBA can affect the growth and metastasis of colorectal cancer (CRC) in orthotopically implanted tumors in nude mice. We found that the oral administration of AKBA (50-200 mg/kg) dose-dependently inhibited the growth of CRC tumors in mice, resulting in decrease in tumor volumes than those seen in vehicle-treated mice without significant decreases in body weight. In addition, we observed that AKBA was highly effective in suppressing ascites and distant metastasis to the liver, lungs and spleen in orthotopically implanted tumors in nude mice. When examined for the mechanism, we found that markers of tumor proliferation index Ki-67 and the microvessel density cluster of differentiation (CD31) were significantly downregulated by AKBA treatment. We also found that AKBA significantly suppressed nuclear factor-κB (NF-κB) activation in the tumor tissue and expression of proinflammatory (cyclooxygenase-2), tumor survival (bcl-2, bcl-xL, inhibitor of apoptosis (IAP-1) and survivin), proliferative (cyclin D1), invasive (intercellular adhesion molecule 1 and matrix metalloproteinase-9) and angiogenic C-X-C (CXC) receptor 4 and vascular endothelial growth factor) biomarkers. When examined for serum and tissue levels of AKBA, a dose-dependent increase in the levels of the drug was detected, indicating its bioavailability. Thus, our findings suggest that this boswellic acid analog can inhibit the growth and metastasis of human CRC in vivo through downregulation of cancer-associated biomarkers.


Subject(s)
Biomarkers, Tumor/metabolism , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/pathology , Enzyme Inhibitors/pharmacology , Neovascularization, Pathologic/metabolism , Triterpenes/pharmacology , Animals , Biomarkers, Tumor/genetics , Boswellia/chemistry , Cell Proliferation , Colorectal Neoplasms/blood supply , Disease Models, Animal , Dose-Response Relationship, Drug , Enzyme Inhibitors/blood , Gene Expression Regulation, Neoplastic/drug effects , HCT116 Cells , Humans , Inflammation/genetics , Inflammation/metabolism , Male , Mice , Mice, Nude , Neoplasm Invasiveness , Neoplasm Metastasis , Neovascularization, Pathologic/genetics , Triterpenes/blood , Xenograft Model Antitumor Assays
12.
Mol Nutr Food Res ; 56(3): 454-65, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22147524

ABSTRACT

SCOPE: The incidence of cancer is significantly lower in regions where turmeric is heavily consumed. Whether lower cancer incidence is due to turmeric was investigated by examining its effects on tumor cell proliferation, on pro-inflammatory transcription factors NF-κB and STAT3, and on associated gene products. METHODS AND RESULTS: Cell proliferation and cell cytotoxicity were measured by the MTT method, NF-κB activity by EMSA, protein expression by Western blot analysis, ROS generation by FACS analysis, and osteoclastogenesis by TRAP assay. Turmeric inhibited NF-κB activation and down-regulated NF-κB-regulated gene products linked to survival (Bcl-2, cFLIP, XIAP, and cIAP1), proliferation (cyclin D1 and c-Myc), and metastasis (CXCR4) of cancer cells. The spice suppressed the activation of STAT3, and induced the death receptors (DR)4 and DR5. Turmeric enhanced the production of ROS, and suppressed the growth of tumor cell lines. Furthermore, turmeric sensitized the tumor cells to chemotherapeutic agents capecitabine and taxol. Turmeric was found to be more potent than pure curcumin for cell growth inhibition. Turmeric also inhibited NF-κB activation induced by RANKL that correlated with the suppression of osteoclastogenesis. CONCLUSION: Our results indicate that turmeric can effectively block the proliferation of tumor cells through the suppression of NF-κB and STAT3 pathways.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Antineoplastic Agents/pharmacology , Curcuma/chemistry , NF-kappa B/antagonists & inhibitors , Osteoclasts/drug effects , Plant Extracts/pharmacology , Animals , Apoptosis/drug effects , Blotting, Western , Capecitabine , Cell Differentiation/drug effects , Cell Proliferation/drug effects , Cyclin D1/genetics , Cyclin D1/metabolism , Deoxycytidine/analogs & derivatives , Deoxycytidine/pharmacology , Down-Regulation , Fluorouracil/analogs & derivatives , Fluorouracil/pharmacology , HCT116 Cells , HT29 Cells , Humans , Mice , NF-kappa B/genetics , NF-kappa B/metabolism , Osteoclasts/cytology , Paclitaxel/pharmacology , RANK Ligand/genetics , RANK Ligand/metabolism , Reactive Oxygen Species/metabolism , Receptors, CXCR4/genetics , Receptors, CXCR4/metabolism , Receptors, Death Domain/genetics , Receptors, Death Domain/metabolism , STAT3 Transcription Factor/genetics , STAT3 Transcription Factor/metabolism , X-Linked Inhibitor of Apoptosis Protein/genetics , X-Linked Inhibitor of Apoptosis Protein/metabolism , bcl-2-Associated X Protein/genetics , bcl-2-Associated X Protein/metabolism
13.
Nutr Cancer ; 64(2): 173-97, 2012.
Article in English | MEDLINE | ID: mdl-22149093

ABSTRACT

Extensive research within the last half a century has revealed that cancer is caused by dysregulation of as many as 500 different gene products. Most natural products target multiple gene products and thus are ideally suited for prevention and treatment of various chronic diseases, including cancer. Dietary agents such as spices have been used extensively in the Eastern world for a variety of ailments for millennia, and five centuries ago they took a golden journey to the Western world. Various spice-derived nutraceuticals, including 1'-acetoxychavicol acetate, anethole, capsaicin, cardamonin, curcumin, dibenzoylmethane, diosgenin, eugenol, gambogic acid, gingerol, thymoquinone, ursolic acid, xanthohumol, and zerumbone derived from galangal, anise, red chili, black cardamom, turmeric, licorice, fenugreek, clove, kokum, ginger, black cumin, rosemary, hop, and pinecone ginger, respectively, are the focus of this review. The modulation of various transcription factors, growth factors, protein kinases, and inflammatory mediators by these spice-derived nutraceuticals are described. The anticancer potential through the modulation of various targets is also the subject of this review. Although they have always been used to improve taste and color and as a preservative, they are now also used for prevention and treatment of a wide variety of chronic inflammatory diseases, including cancer.


Subject(s)
Dietary Supplements , Neoplasms/drug therapy , Phytotherapy , Signal Transduction/drug effects , Spices , Animals , Antineoplastic Agents, Phytogenic/therapeutic use , Cell Line, Tumor , Humans , Inflammation Mediators , Intercellular Signaling Peptides and Proteins , NF-kappa B/antagonists & inhibitors , Neoplasms/metabolism , Neoplasms/prevention & control , Protein Kinase Inhibitors/therapeutic use , Protein Kinases/drug effects , Receptors, Growth Factor/antagonists & inhibitors , Transcription Factors/antagonists & inhibitors , Transcription Factors/drug effects
14.
PLoS One ; 6(10): e26943, 2011.
Article in English | MEDLINE | ID: mdl-22066019

ABSTRACT

Pancreatic cancer (PaCa) is one of the most lethal cancers, with an estimated 5-year survival of <5% even when patients are given the best treatment available. In addition, these treatments are often toxic and expensive, thus new agents which are safe, affordable and effective are urgently needed. We describe here the results of our study with acetyl-11-keto-ß-boswellic acid (AKBA), an agent obtained from an Ayurvedic medicine, gum resin of Boswellia serrata. Whether AKBA has an activity against human PaCa, was examined in in vitro models and in an orthotopic nude mouse model of PaCa. We found that AKBA inhibited the proliferation of four different PaCa cell lines (AsPC-1, PANC-28, and MIA PaCa-2 with K-Ras and p53 mutations, and BxPC-3 with wild-type K-Ras and p53 mutation). These effects correlated with an inhibition of constitutively active NF-κB and suppression of NF-κB regulating gene expression. AKBA also induced apoptosis, and sensitized the cells to apoptotic effects of gemcitabine. In the orthotopic nude mouse model of PaCa, p.o. administration of AKBA alone (100 mg/kg) significantly inhibited the tumor growth; this activity was enhanced by gemcitabine. In addition, AKBA inhibited the metastasis of the PaCa to spleen, liver, and lungs. This correlated with decreases in Ki-67, a biomarker of proliferation, and CD31, a biomarker of microvessel density, in the tumor tissue. AKBA produced significant decreases in the expression of NF-κB regulating genes in the tissues. Immunohistochemical analysis also showed AKBA downregulated the expression of COX-2, MMP-9, CXCR4, and VEGF in the tissues. Overall these results demonstrate that AKBA can suppress the growth and metastasis of human pancreatic tumors in an orthotopic nude mouse model that correlates with modulation of multiple targets.


Subject(s)
Molecular Targeted Therapy , Pancreatic Neoplasms/pathology , Triterpenes/pharmacology , Xenograft Model Antitumor Assays , Animals , Apoptosis/drug effects , Biomarkers, Tumor/metabolism , Cell Proliferation/drug effects , Chromatography, High Pressure Liquid , Deoxycytidine/analogs & derivatives , Deoxycytidine/pharmacology , Gene Expression Regulation, Neoplastic/drug effects , Humans , Ki-67 Antigen/metabolism , Male , Mice , Mice, Nude , NF-kappa B/metabolism , Neoplasm Metastasis , Neovascularization, Pathologic/drug therapy , Neovascularization, Pathologic/pathology , Pancreatic Neoplasms/blood , Pancreatic Neoplasms/blood supply , Pancreatic Neoplasms/genetics , Platelet Endothelial Cell Adhesion Molecule-1/metabolism , Triterpenes/blood , Triterpenes/therapeutic use , Gemcitabine
15.
Free Radic Res ; 45(8): 864-79, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21651450

ABSTRACT

Extensive research in the past decade has revealed cancer to be a multigenic disease caused by perturbation of multiple cell signalling pathways and dysregulation of numerous gene products, all of which have been linked to inflammation. It is also becoming evident that various lifestyle factors, such as tobacco and alcohol use, diet, environmental pollution, radiation and infections, can cause chronic inflammation and lead to tumourigenesis. Chronic diseases caused by ongoing inflammation therefore require chronic, not acute, treatment. Nutraceuticals, compounds derived from fruits, vegetables, spices and cereals, can be used chronically. This study discusses the molecular targets of some nutraceuticals that happen to be markers of chronic inflammation and how they can prevent or treat cancer. These naturally-occurring agents in the diet have great potential as anti-cancer drugs, thus proving Hippocrates, who proclaimed 25 centuries ago, 'Let food be thy medicine and medicine be thy food'.


Subject(s)
Diet , Neoplasms/diet therapy , Neoplasms/metabolism , Animals , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Antioxidants/pharmacology , Antioxidants/therapeutic use , Dietary Supplements , Free Radicals/pharmacology , Free Radicals/therapeutic use , Humans , Inflammation/diet therapy , Inflammation/prevention & control , Neoplasms/pathology , Neoplasms/prevention & control
16.
Cancer Prev Res (Phila) ; 4(7): 1084-94, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21490133

ABSTRACT

The transcription factor, STAT3, is associated with proliferation, survival, and metastasis of cancer cells. We investigated whether gambogic acid (GA), a xanthone derived from the resin of traditional Chinese medicine, Garcinia hanburyi (mangosteen), can regulate the STAT3 pathway, leading to suppression of growth and sensitization of cancer cells. We found that GA induced apoptosis in human multiple myeloma cells that correlated with the inhibition of both constitutive and inducible STAT3 activation. STAT3 phosphorylation at both tyrosine residue 705 and serine residue 727 was inhibited by GA. STAT3 suppression was mediated through the inhibition of activation of the protein tyrosine kinases Janus-activated kinase 1 (JAK1) and JAK2. Treatment with the protein tyrosine phosphatase (PTP) inhibitor pervanadate reversed the GA-induced downregulation of STAT3, suggesting the involvement of a PTP. We also found that GA induced the expression of the PTP SHP-1. Deletion of the SHP-1 gene by siRNA suppressed the ability of GA to inhibit STAT3 activation and to induce apoptosis, suggesting the critical role of SHP-1 in its action. Moreover, GA downregulated the expression of STAT3-regulated antiapoptotic (Bcl-2, Bcl-xL, and Mcl-1), proliferative (cyclin D1), and angiogenic (VEGF) proteins, and this correlated with suppression of proliferation and induction of apoptosis. Overall, these results suggest that GA blocks STAT3 activation, leading to suppression of tumor cell proliferation and induction of apoptosis.


Subject(s)
Apoptosis/drug effects , Cell Proliferation/drug effects , Protein Tyrosine Phosphatase, Non-Receptor Type 6/metabolism , STAT3 Transcription Factor/metabolism , Xanthones/pharmacology , Blotting, Western , Breast Neoplasms/drug therapy , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Carcinoma, Squamous Cell/drug therapy , Carcinoma, Squamous Cell/metabolism , Carcinoma, Squamous Cell/pathology , DNA, Neoplasm/metabolism , Electrophoretic Mobility Shift Assay , Female , Head and Neck Neoplasms/drug therapy , Head and Neck Neoplasms/metabolism , Head and Neck Neoplasms/pathology , Humans , Immunoenzyme Techniques , Janus Kinase 1/metabolism , Janus Kinase 2/metabolism , Male , Multiple Myeloma/drug therapy , Multiple Myeloma/metabolism , Multiple Myeloma/pathology , Phosphorylation/drug effects , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Protein Transport , Protein Tyrosine Phosphatase, Non-Receptor Type 6/antagonists & inhibitors , Protein Tyrosine Phosphatase, Non-Receptor Type 6/genetics , RNA, Small Interfering/genetics , Tumor Cells, Cultured
17.
J Biol Chem ; 286(7): 5546-57, 2011 Feb 18.
Article in English | MEDLINE | ID: mdl-21156789

ABSTRACT

Discovery of the molecular targets of traditional medicine and its chemical footprints can validate the use of such medicine. In the present report, we investigated the effect of ursolic acid (UA), a pentacyclic triterpenoid found in rosemary and holy basil, on apoptosis induced by TRAIL. We found that UA potentiated TRAIL-induced apoptosis in cancer cells. In addition, UA also sensitized TRAIL-resistant cancer cells to the cytokine. When we investigated the mechanism, we found that UA down-regulated cell survival proteins and induced the cell surface expression of both TRAIL receptors, death receptors 4 and 5 (DR4 and -5). Induction of receptors by UA occurred independently of cell type. Gene silencing of either receptor by small interfering RNA reduced the apoptosis induced by UA and the effect of TRAIL. In addition, UA also decreased the expression of decoy receptor 2 (DcR2) but not DcR1. Induction of DRs was independent of p53 because UA induced DR4 and DR5 in HCT116 p53(-/-) cells. Induction of DRs, however, was dependent on JNK because UA induced JNK, and its pharmacologic inhibition abolished the induction of the receptors. The down-regulation of survival proteins and up-regulation of the DRs required reactive oxygen species (ROS) because UA induced ROS, and its quenching abolished the effect of the terpene. Also, potentiation of TRAIL-induced apoptosis by UA was significantly reduced by both ROS quenchers and JNK inhibitor. In addition, UA was also found to induce the expression of DRs, down-regulate cell survival proteins, and activate JNK in orthotopically implanted human colorectal cancer in a nude mouse model. Overall, our results showed that UA potentiates TRAIL-induced apoptosis through activation of ROS and JNK-mediated up-regulation of DRs and down-regulation of DcR2 and cell survival proteins.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Apoptosis/drug effects , Colorectal Neoplasms/metabolism , MAP Kinase Kinase 4/metabolism , Reactive Oxygen Species/metabolism , TNF-Related Apoptosis-Inducing Ligand/pharmacology , Triterpenes/pharmacology , Tumor Suppressor Protein p53/metabolism , Up-Regulation/drug effects , Animals , Antineoplastic Agents, Phytogenic/agonists , Apoptosis/genetics , Caco-2 Cells , Cell Survival/drug effects , Cell Survival/genetics , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/genetics , Down-Regulation/drug effects , Down-Regulation/genetics , Drug Synergism , Humans , MAP Kinase Kinase 4/genetics , Mice , Mice, Nude , Neoplasm Transplantation , Receptors, TNF-Related Apoptosis-Inducing Ligand/agonists , Receptors, TNF-Related Apoptosis-Inducing Ligand/genetics , Receptors, TNF-Related Apoptosis-Inducing Ligand/metabolism , TNF-Related Apoptosis-Inducing Ligand/agonists , Triterpenes/agonists , Tumor Necrosis Factor Decoy Receptors/agonists , Tumor Necrosis Factor Decoy Receptors/genetics , Tumor Necrosis Factor Decoy Receptors/metabolism , Tumor Suppressor Protein p53/genetics , Up-Regulation/genetics , Xenograft Model Antitumor Assays/methods , Ursolic Acid
18.
Mol Pharmacol ; 79(2): 279-89, 2011 Feb.
Article in English | MEDLINE | ID: mdl-20930110

ABSTRACT

Most anticancer drugs have their origin in traditional medicinal plants. We describe here a flavone, 5,3'-dihydroxy-3,6,7,8,4'-pentamethoxyflavone (PMF), from the leaves of the Thai plant Gardenia obtusifolia, that has anti-inflammatory and anticancer potential. Because the nuclear factor-κB (NF-κB) pathway is linked to inflammation and tumorigenesis, we investigated the effect of PMF on this pathway. We found that PMF suppressed NF-κB activation induced by inflammatory agents, tumor promoters, and carcinogens. This suppression was not specific to the cell type. Although PMF did not directly modify the ability of NF-κB proteins to bind to DNA, it inhibited IκBα (inhibitory subunit of NF-κB) kinase, leading to suppression of phosphorylation and degradation of IκBα, and suppressed consequent p65 nuclear translocation, thus abrogating NF-κB-dependent reporter gene expression. Suppression of the NF-κB cell signaling pathway by the flavone led to the inhibition of expression of NF-κB-regulated gene products that mediate inflammation (cyclooxygenase-2), survival (XIAP, survivin, Bcl-xL, and cFLIP), proliferation (cyclin D1), invasion (matrix metalloproteinase-9), and angiogenesis (vascular endothelial growth factor). Suppression of antiapoptotic gene products by PMF correlated with the enhancement of apoptosis induced by tumor necrosis factor-α and the chemotherapeutic agents cisplatin, paclitaxel, and 5-flurouracil. Overall, our results indicate that PMF suppresses the activation of NF-κB and NF-κB-regulated gene expression, leading to the enhancement of apoptosis. This is the first report to demonstrate that this novel flavone has anti-inflammatory and anticancer effects by targeting the IKK complex.


Subject(s)
Angiogenesis Inducing Agents/metabolism , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Down-Regulation/drug effects , Flavonoids/pharmacology , I-kappa B Kinase/antagonists & inhibitors , Tumor Necrosis Factor-alpha/pharmacology , Base Sequence , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Enzyme Activation , Genes, Reporter , Humans , I-kappa B Kinase/metabolism , Immunohistochemistry , Oligodeoxyribonucleotides
19.
Mol Cancer Res ; 8(10): 1425-36, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20826545

ABSTRACT

Most patients with cancer die not because of the tumor in the primary site, but because it has spread to other sites. Common tumors, such as breast, multiple myeloma, and prostate tumors, frequently metastasize to the bone. It is now well recognized that osteoclasts are responsible for the osteolysis observed in bone metastases of the tumor. Receptor activator of NF-κB ligand (RANKL), a member of the tumor necrosis factor superfamily and an activator of the NF-κB signaling pathway, has emerged as a major mediator of bone loss, commonly associated with cancer and other chronic inflammatory diseases. Embelin (2,5-dihydroxy-3-undecyl-1,4-benzoquinone), derived from the Ayurvedic medicinal plant Embelia ribes, has been shown to bind and inhibit X-linked inhibitor of apoptosis protein and inhibit inflammatory pathways. We investigated whether embelin could inhibit osteoclastogenesis-associated bone loss induced by RANKL and by tumor cells in vitro. We found that embelin suppressed the RANKL-induced differentiation of monocytes into osteoclasts. This benzoquinone also suppressed the osteoclastogenesis induced by multiple myeloma and by breast cancer cells. This effect of embelin correlated with the suppression of NF-κB activation and inhibition of IκBα phosphorylation and IκBα degradation. Inhibition of IκBα phosphorylation was due to the inhibition of IκBα kinase (IKK) activation. Furthermore, by using an inhibitor of the IKKγ or NF-κB essential modulator (NEMO), the regulatory component of the IKK complex, we showed that the NF-κB signaling pathway is mandatory for RAW 264.7 cell differentiation into osteoclasts. Thus, embelin, an inhibitor of RANKL-induced NF-κB activation has great potential as a therapeutic agent for osteoporosis and cancer-linked bone loss.


Subject(s)
Benzoquinones/pharmacology , NF-kappa B/antagonists & inhibitors , Osteoclasts/drug effects , Osteoclasts/pathology , RANK Ligand/physiology , Signal Transduction/drug effects , Animals , Bone Density Conservation Agents/pharmacology , Bone Neoplasms/drug therapy , Bone Neoplasms/pathology , Bone Neoplasms/secondary , Breast Neoplasms/drug therapy , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Line , Cell Line, Tumor , Humans , Mice , Multiple Myeloma/drug therapy , Multiple Myeloma/metabolism , Multiple Myeloma/pathology , NF-kappa B/physiology , Osteoclasts/metabolism , Osteoporosis/drug therapy , Osteoporosis/pathology , Osteoporosis/prevention & control , Signal Transduction/physiology
20.
Biochem Pharmacol ; 80(12): 1833-43, 2010 Dec 15.
Article in English | MEDLINE | ID: mdl-20654584

ABSTRACT

Extensive research within the last two decades has revealed that most chronic illnesses, including cancer, diabetes, and cardiovascular and pulmonary diseases, are mediated through chronic inflammation. Thus, suppressing chronic inflammation has the potential to delay, prevent, and even treat various chronic diseases, including cancer. Various nutraceuticals from fruits, vegetables, vitamins, spices, legumes, and traditional Chinese and Ayurvedic medicine have been shown to safely suppress proinflammatory pathways; however, their low bioavailability in vivo limits their use in preventing and treating cancer. We describe here the potential of nanotechnology to fill this gap. Several nutraceuticals, including curcumin, green tea polyphenols, coenzyme Q, quercetin, thymoquinone and others, have been packaged as nanoparticles and proven to be useful in "nanochemoprevention" and "nano-chemotherapy".


Subject(s)
Anti-Inflammatory Agents/therapeutic use , Anticarcinogenic Agents/therapeutic use , Dietary Supplements , Nanoparticles , Neoplasms/therapy , Animals , Anti-Inflammatory Agents/administration & dosage , Anti-Inflammatory Agents/pharmacology , Anticarcinogenic Agents/administration & dosage , Anticarcinogenic Agents/pharmacology , Chemistry, Pharmaceutical , Humans , Inflammation/metabolism , Inflammation/prevention & control , Inflammation/therapy , Neoplasms/metabolism , Neoplasms/prevention & control
SELECTION OF CITATIONS
SEARCH DETAIL