Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 31
Filter
Add more filters










Publication year range
1.
Mol Biol Rep ; 51(1): 788, 2024 Jul 06.
Article in English | MEDLINE | ID: mdl-38970704

ABSTRACT

Despite many efforts, a comprehensive understanding and clarification of the intricate connections within cancer cell metabolism remain elusive. This might pertain to intracellular dynamics and the complex interplay between cancer cells, and cells with the tumor stroma. Almost a century ago, Otto Warburg found that cancer cells exhibit a glycolytic phenotype, which continues to be a subject of thorough investigation. Past and ongoing investigations have demonstrated intricate mechanisms by which tumors modulate their functionality by utilizing extracellular glucose as a substrate, thereby sustaining the essential proliferation of cancer cells. This concept of "aerobic glycolysis," where cancer cells (even in the presence of enough oxygen) metabolize glucose to produce lactate plays a critical role in cancer progression and is regulated by various signaling pathways. Recent research has revealed that the canonical wingless-related integrated site (WNT) pathway promotes aerobic glycolysis, directly and indirectly, thereby influencing cancer development and progression. The present review seeks to gather knowledge about how the WNT/ß-catenin pathway influences aerobic glycolysis, referring to relevant studies in different types of cancer. Furthermore, we propose the concept of impeding the glycolytic phenotype of tumors by employing specific inhibitors that target WNT/ß-catenin signaling.


Subject(s)
Glycolysis , Neoplasms , Wnt Signaling Pathway , Humans , Neoplasms/metabolism , Neoplasms/pathology , Neoplasms/genetics , beta Catenin/metabolism , Warburg Effect, Oncologic , Animals , Glucose/metabolism
2.
Chem Commun (Camb) ; 59(85): 12759-12762, 2023 Oct 24.
Article in English | MEDLINE | ID: mdl-37811603

ABSTRACT

Integrin-mediated cellular delivery was attempted to optimize practical applications of hydrophobic ionophores. The potent ionophore preferentially transports H+/Cl- across the lipid bilayers following a symport mechanism. The RGD-peptide-appended tag was stimulated by glutathione to generate the active ionophore, prompting the transport of Cl- under the cellular environment.


Subject(s)
Lipid Bilayers , Oligopeptides , Ionophores/chemistry , Ion Transport , Lipid Bilayers/chemistry , Biological Transport , Oligopeptides/chemistry
3.
Biotechnol Bioeng ; 120(8): 2049-2055, 2023 08.
Article in English | MEDLINE | ID: mdl-37439093

ABSTRACT

Cancer cell lines play a crucial role as invaluable models in cancer research, facilitating the examination of cancer progression as well as the advancement of diagnostics and treatments. While they may not perfectly replicate the original tumor, they generally exhibit similar characteristics. Low-passage cancer cell lines are generally preferred due to their closer resemblance to the original tumor, as long-term culturing can alter the genetic and molecular profiles of a cell line thereby highlighting the importance of monitoring the passage number (PN). Variations in proliferation, migration, gene expression, and drug sensitivity can be linked to PN differences. PN can also influence DNA methylation levels, metabolic profiles, and the expression of genes/or proteins in cancer cell lines. When conducting research on cancer cell lines, it is crucial for researchers to carefully select the appropriate PN to maintain consistency and reliability of results. Moreover, to ensure dependability and replicability, scientists ought to actively track the growth, migration, and gene/or protein profiles of cancer cell lines at specific PNs. This approach enables the identification of the most suitable range of PNs for experiments, guaranteeing consistent and precise results. Additionally, such efforts serve to minimize disparities and uphold the integrity of research. In this review, we have laid out recommendations for laboratories to overcome these PN discrepancies when working with cancer cell lines.


Subject(s)
Neoplasms , Humans , Reproducibility of Results , Neoplasms/genetics , Cell Line , DNA Methylation/genetics , Cell Line, Tumor
4.
Biomater Adv ; 153: 213527, 2023 Oct.
Article in English | MEDLINE | ID: mdl-37418935

ABSTRACT

Light-based three-dimensional (3D) printing has been under use extensively to fabricate complex geometrical constructs which find a vast application in the fields of drug delivery and tissue engineering fields due to its ability to recapitulate the intricate biological architecture and thus provides avenues to achieve previously unachievable biomedical devices. The inherent problem associated with light-based 3D printing (from a biomedical perspective) is that of light scattering causing inaccurate and defective prints which results in erroneous drug loading in 3D printed dosage forms and can also render the environment of the polymers toxic for the biological cells and tissues. In this regard, an innovative additive comprising of a nature-derived drug-cum-photoabsorber (curcumin) entrapped in naturally derived protein (bovine serum albumin) is envisaged to act as a photoabsorbing system that can improve the printing quality of 3D printed drug delivery formulations (macroporous pills) as well as provide stimuli-responsive release of the same upon oral ingestion. The delivery system was designed to endure the chemically and mechanically hostile gastric environment and deliver the drug in the small intestine to improve absorption. A 3 × 3 grid macroporous pill was designed (specifically to withstand the mechanically hostile gastric environment) and 3D printed using Stereolithography comprising of a resin system including acrylic Acid, PEGDA and PEG 400 along with curcumin loaded BSA nanoparticles (Cu-BSA NPs) as a multifunctional additive and TPO as the photoinitiator. The 3D printed macroporous pills were found to show excellent fidelity to CAD design as evident from the resolution studies. The mechanical performance of the macroporous pills was found to be extremely superior to monolithic pills. The pills found to release curcumin in pH responsive manner with slower release at acidic pH but faster release at intestinal pH due to its similar swelling behavior. Finally, the pills were found to be cytocompatible to mammalian kidney and colon cell lines.


Subject(s)
Curcumin , Nanoparticles , Curcumin/pharmacology , Curcumin/therapeutic use , Printing, Three-Dimensional , Stereolithography , Polymers
5.
Cancers (Basel) ; 14(24)2022 Dec 10.
Article in English | MEDLINE | ID: mdl-36551563

ABSTRACT

Treatment of melanoma with a BRAF inhibitor (BRAFi) frequently initiates development of BRAFi resistance, leading to increased tumor progression and metastasis. Previously, we showed that combined inhibition of elevated WNT5A and IL-6 signaling reduced the invasion and migration of BRAFi-resistant (BRAFi-R) melanoma cells. However, the use of a combined approach per se and the need for high inhibitor concentrations to achieve this effect indicate a need for an alternative and single target. One such target could be myristoylated alanine-rich C-kinase substrate (MARCKS), a downstream target of WNT5A in BRAFi-sensitive melanoma cells. Our results revealed that MARCKS protein expression and activity are significantly elevated in PLX4032 and PLX4720 BRAFi-R A375 and HTB63 melanoma cells. Surprisingly, neither WNT5A nor IL-6 contributed to the increases in MARCKS expression and activity in BRAFi-R melanoma cells, unlike in BRAFi-sensitive melanoma cells. However, despite the above findings, our functional validation experiments revealed that MARCKS is essential for the increased metastatic behavior of BRAFi-R melanoma cells. Knockdown of MARCKS in BRAFi-R melanoma cells caused reductions in the F-actin content and the number of filopodia-like protrusions, explaining the impaired migration, invasion and metastasis of these cells observed in vitro and in an in vivo zebrafish model. In our search for an alternative explanation for the increased activity of MARCKS in BRAFi-R melanoma cells, we found elevated basal activities of PKCα, PKCε, PKCι, and RhoA. Interestingly, combined inhibition of basal PKC and RhoA effectively impaired MARCKS activity in BRAFi-R melanoma cells. Our results reveal that MARCKS is an attractive single antimetastatic target in BRAFi-R melanoma cells.

6.
Free Radic Biol Med ; 186: 17-30, 2022 06.
Article in English | MEDLINE | ID: mdl-35513128

ABSTRACT

Oxidative damage and accumulation of extracellular matrix (ECM) components play a crucial role in the adverse outcome of cardiac hypertrophy. Evidence suggests that nuclear factor erythroid-derived factor 2 related factor 2 (Nrf2) can modulate oxidative damage and adverse myocardial remodeling. Daphnetin (Daph) is a coumarin obtained from the plant genus Daphne species that exerts anti-oxidative and anti-inflammatory properties. Herein, we investigated the roles of Daph in transverse aortic constriction (TAC)-induced cardiac hypertrophy and fibrosis in mice. TAC-induced alterations in cardiac hypertrophy markers, histopathological changes, and cardiac function were markedly ameliorated by oral administration of Daph in mice. We found that Daph significantly reduced the reactive oxygen species (ROS) generation, increased the nuclear translocation of Nrf2, and consequently, reinstated the protein levels of NAD(P)H quinone dehydrogenase1 (NQO1), heme oxygenase-1 (HO-1), and other antioxidants in the heart. Besides, Daph significantly inhibited the TAC-induced accumulation of ECM components, including α-smooth muscle actin (α-SMA), collagen I, collagen III, and fibronectin, and interfered with the TGF-ß1/Smad2/3 signaling axis. Further studies revealed that TAC-induced terminal deoxynucleotidyl transferase dUTP nick end labeling (TUNEL) positive nuclei and the protein levels of Bax/Bcl2 ratio and cleaved caspase 3 were substantially decreased by Daph treatment. We further characterized the effect of Daph on angiotensin II (Ang-II)-stimulated H9c2 cardiomyoblast cells and observed that Daph markedly decreased the Ang-II induced increase in cell size, production of ROS, and proteins associated with apoptosis and fibrosis. Mechanistically, Daph alone treatment enhanced the protein levels of Nrf2, NQO1, and HO-1 in H9c2 cells. The inhibition of this axis by Si-Nrf2 transfection abolished the protective effect of Daph in H9c2 cells. Taken together, Daph effectively counteracted the TAC-induced cardiac hypertrophy and fibrosis by improving the Nrf2/HO-1 axis and inhibiting the TGF-ß1/Smad2/3 signaling axis.


Subject(s)
Heme Oxygenase-1 , Membrane Proteins , NF-E2-Related Factor 2 , Smad2 Protein , Smad3 Protein , Transforming Growth Factor beta1 , Umbelliferones , Angiotensin II/metabolism , Animals , Cardiomegaly/drug therapy , Cardiomegaly/metabolism , Collagen/metabolism , Heme Oxygenase-1/metabolism , Membrane Proteins/metabolism , Mice , NF-E2-Related Factor 2/metabolism , Reactive Oxygen Species/metabolism , Signal Transduction/drug effects , Smad2 Protein/antagonists & inhibitors , Smad2 Protein/metabolism , Smad3 Protein/antagonists & inhibitors , Smad3 Protein/metabolism , Transforming Growth Factor beta1/antagonists & inhibitors , Transforming Growth Factor beta1/metabolism , Umbelliferones/pharmacology , Up-Regulation , Ventricular Remodeling/drug effects
7.
Mol Biol Rep ; 48(5): 4733-4745, 2021 May.
Article in English | MEDLINE | ID: mdl-34047880

ABSTRACT

Among breast cancer subtypes, the triple negative breast cancer (TNBC) has the worst prognosis. In absence of any permitted targeted therapy, standard chemotherapy is the mainstay for TNBC treatment. Hence, there is a crucial need to identify potential druggable targets in TNBCs for its effective treatment. In recent times, metabolic reprogramming has emerged as cancer cells hallmark, wherein cancer cells display discrete metabolic phenotypes to fuel cell progression and metastasis. Altered glycolysis is one such phenotype, in which even in oxygen abundance majority of cancer cells harvest considerable amount of energy through elevated glycolytic-flux. In the present review, we attempt to summarize the role of key glycolytic enzymes i.e. HK, Hexokinase; PFK, Phosphofructokinase; PKM2, Pyruvate kinase isozyme type 2; and LDH, Lactate dehydrogenase in TNBCs, and possible therapeutic options presently available.


Subject(s)
Triple Negative Breast Neoplasms/enzymology , Triple Negative Breast Neoplasms/genetics , Warburg Effect, Oncologic , Carrier Proteins/genetics , Carrier Proteins/metabolism , Female , Gene Expression Regulation, Neoplastic/genetics , Hexokinase/genetics , Hexokinase/metabolism , Humans , L-Lactate Dehydrogenase/genetics , L-Lactate Dehydrogenase/metabolism , Membrane Proteins/genetics , Membrane Proteins/metabolism , Phosphofructokinases/genetics , Phosphofructokinases/metabolism , Pyruvate Kinase/genetics , Pyruvate Kinase/metabolism , Thyroid Hormones/genetics , Thyroid Hormones/metabolism , Triple Negative Breast Neoplasms/metabolism , Triple Negative Breast Neoplasms/pathology , Thyroid Hormone-Binding Proteins
8.
Cancers (Basel) ; 12(2)2020 Feb 04.
Article in English | MEDLINE | ID: mdl-32033033

ABSTRACT

WNT5A is a well-known mediator of melanoma cell invasion and metastasis via its ability to activate protein kinase C (PKC), which is monitored by phosphorylation of the endogenous PKC substrate myristoylated alanine-rich c-kinase substrate (MARCKS). However, a possible direct contribution of MARCKS in WNT5A-mediated melanoma cell invasion has not been investigated. Analyses of melanoma patient databases suggested that similar to WNT5A expression, MARCKS expression appears to be associated with increased metastasis. A relationship between the two is suggested by the findings that recombinant WNT5A (rWNT5A) induces both increased expression and phosphorylation of MARCKS, whereas WNT5A silencing does the opposite. Moreover, WNT5A-induced invasion of melanoma cells was blocked by siRNA targeting MARCKS, indicating a crucial role of MARCKS expression and/or its phosphorylation. Next, we employed a peptide inhibitor of MARCKS phosphorylation that did not affect MARCKS expression and found that it abolished WNT5A-induced melanoma cell invasion. Similarly, rWNT5A induced the accumulation of phosphorylated MARCKS in membrane protrusions at the leading edge of melanoma cells. Our results demonstrate that WNT5A-induced phosphorylation of MARCKS is not only an indicator of PKC activity but also a crucial regulator of the metastatic behavior of melanoma and therefore an attractive future antimetastatic target in melanoma patients.

9.
Mol Oncol ; 13(2): 480-494, 2019 02.
Article in English | MEDLINE | ID: mdl-30582770

ABSTRACT

The identification of novel antimetastatic therapeutic targets is necessary for improved treatment of patients with acquired BRAF inhibitor-resistant (BRAFi-R) melanoma, in whom metastasis is a major concern. Our present study focused on the identification of such targets to explore novel antimetastatic therapeutic options for BRAFi-R melanoma patients. We confirmed the development of BRAFi resistance in our BRAFi-treated melanoma cell lines by demonstrating reduced sensitivity to BRAF inhibitors, increased ERK1/2 activity and increased WNT5A expression. Here, we demonstrated for the first time that high secretion of interleukin-6 (IL-6) was associated with increased invasive migration of BRAFi-R melanoma cells. This finding could be readily explained by the increased expression of WNT5A in BRAFi-R melanoma cells and the presence of an IL-6/WNT5A positive feedback loop in parental melanoma cells. Surprisingly, however, we found that the IL-6/WNT5A positive feedback loop present in parental melanoma cells was lost during the development of acquired BRAFi resistance, meaning that IL-6 and WNT5A signalling were independent events in BRAFi-R melanoma cells. Despite the absence of an IL-6/WNT5A loop, we found that both an IL-6 blocking antibody and the WNT5A antagonist Box5 alone impaired the elevated invasive migration of BRAFi-R melanoma cells, but combined use of the two was more effective. This impaired invasive migration of BRAFi-R melanoma cells correlated well with the reduction in Cdc42-GTPase activity and alterations of the actin cytoskeleton in these cells. In summary, our novel identification of IL-6 as a key independent promoter of the invasive migration of BRAFi-R melanoma cells stresses that a combination of a blocking IL-6 antibody and administration of the WNT5A antagonist Box5 might be an attractive antimetastatic approach for future treatment of BRAFi-R melanoma patients.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Cell Movement , Drug Resistance, Neoplasm , Interleukin-6/metabolism , Melanoma/drug therapy , Melanoma/pathology , Protein Kinase Inhibitors/therapeutic use , Proto-Oncogene Proteins B-raf/antagonists & inhibitors , Actins/metabolism , Antibodies, Blocking/pharmacology , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Cell Line, Tumor , Cell Movement/drug effects , Drug Resistance, Neoplasm/drug effects , Humans , Mutation/genetics , Neoplasm Invasiveness , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins B-raf/metabolism , Signal Transduction/drug effects , Wnt-5a Protein , cdc42 GTP-Binding Protein/metabolism
10.
Cancer Metastasis Rev ; 37(4): 767-778, 2018 12.
Article in English | MEDLINE | ID: mdl-30171384

ABSTRACT

Despite the clinical development of novel adjuvant and neoadjuvant chemotherapeutic drugs, metastatic breast cancer is one of the leading causes of cancer-related death among women. The present review focuses on the relevance, mechanisms, and therapeutic potential of targeting WNT5A as a future anti-metastatic treatment strategy for breast cancer patients by restoring WNT5A signaling as an innovative therapeutic option. WNT5A is an auto- and paracrine ß-catenin-independent ligand that has been shown to induce tumor suppression as well as oncogenic signaling, depending upon cancer type. In breast cancer patients, WNT5A protein expression has been observed to be significantly reduced in between 45 and 75% of the cases and associated with early relapse and reduced disease-free survival. WNT5A triggers various downstream signaling pathways in breast cancer that primarily affect tumor cell migration and invasion. The accumulated in vitro results reveal that treatment of WNT5A-negative breast cancer cells with recombinant WNT5A caused different tumor-suppressive responses and in particular it impaired migration and invasion. The anti-migratory/invasive and anti-metastatic effects of reconstituting WNT5A signaling by the small WNT5A mimicking peptide Foxy5 form the basis for two successful clinical phase 1-studies aiming at determining safety and pharmacokinetics as well as defining dose-level for a subsequent phase 2-study. We conclude that re-installation of WNT5A signaling is an attractive and promising anti-metastatic therapeutic approach for future treatment of WNT5A-negative breast cancer patients.


Subject(s)
Breast Neoplasms/drug therapy , Breast Neoplasms/metabolism , Wnt-5a Protein/metabolism , Animals , Biomimetic Materials/pharmacology , Breast Neoplasms/pathology , Humans , Molecular Targeted Therapy , Neoplasm Invasiveness , Oligopeptides/pharmacology , Recombinant Proteins/pharmacology , Signal Transduction , Wnt-5a Protein/pharmacology
11.
Oncotarget ; 7(25): 37790-37802, 2016 Jun 21.
Article in English | MEDLINE | ID: mdl-27191257

ABSTRACT

Increased expression and signalling of WNT5A and interleukin-6 (IL-6) have both been shown to promote melanoma progression. Here, we investigated the proposed existence of a WNT5A-IL-6 positive feedback loop that drives melanoma migration and invasion. First, the HOPP algorithm revealed that the invasive phenotype of cultured melanoma cells was significantly correlated with increased expression of WNT5A or IL-6. In three invasive melanoma cell lines, endogenous WNT5A protein expression was related to IL-6 protein secretion. Knockdown with anti-IL-6 siRNAs or treating WM852 melanoma cells with a neutralising anti-IL-6 antibody reduced WNT5A protein expression. Conversely, the silencing of WNT5A expression by WNT5A siRNAs or treating WM852 melanoma cells with Box5 (a WNT5A antagonist) significantly reduced IL-6 secretion. Interestingly, these effects occurred at the protein level but not at the transcriptional levels. Functionally, we demonstrated that combined siRNA knockdown of WNT5A and IL-6 expression or the simultaneous inhibition of WNT5A and IL-6 signalling inhibited melanoma cell invasion more effectively than suppressing each factor individually. Together, our results demonstrate that WNT5A and IL-6 are connected through a positive feedback loop in melanoma cells and that the combined targeting of both molecules could serve as an effective therapeutic means to reduce melanoma metastasis.


Subject(s)
Gene Expression Regulation, Neoplastic , Interleukin-6/metabolism , Melanoma/metabolism , Skin Neoplasms/metabolism , Wnt-5a Protein/metabolism , Algorithms , Cell Line, Tumor , Cell Movement , Feedback, Physiological , Gene Expression Profiling , Gene Silencing , Humans , Neoplasm Invasiveness , Oligonucleotide Array Sequence Analysis , Phenotype , RNA, Small Interfering/metabolism
12.
Cancers (Basel) ; 7(3): 1900-24, 2015 Sep 17.
Article in English | MEDLINE | ID: mdl-26393652

ABSTRACT

In recent years, scientists have advocated the use of targeted therapies in the form of drugs that modulate genes and proteins that are directly associated with cancer progression and metastasis. Malignant melanoma is a dreadful cancer type that has been associated with the rapid dissemination of primary tumors to multiple sites, including bone, brain, liver and lungs. The discovery that approximately 40%-50% of malignant melanomas contain a mutation in BRAF at codon 600 gave scientists a new approach to tackle this disease. However, clinical studies on patients have shown that although BRAFi (BRAF inhibitors) trigger early anti-tumor responses, the majority of patients later develop resistance to the therapy. Recent studies have shown that WNT5A plays a key role in enhancing the resistance of melanoma cells to BRAFi. The focus of the current review will be on melanoma development, signaling pathways important to acquired resistance to BRAFi, and why WNT5A inhibitors are attractive candidates to be included in combinatorial therapies for melanoma.

13.
Int J Biochem Cell Biol ; 66: 75-84, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26212257

ABSTRACT

Combination therapy using two or more small molecule inhibitors of aberrant signaling cascade in aggressive breast cancers is a promising therapeutic strategy over traditional monotherapeutic approaches. Here, we have studied the synergistic mechanism of resveratrol and curcumin induced apoptosis using in vitro (cigarette smoke condensate mediated transformed breast epithelial cell, MCF-10A-Tr) and in vivo (tumor xenograft mice) model system. Resveratrol exposure increased the intracellular uptake of curcumin in a dose dependent manner and caused apoptosis in MCF-10A-Tr cells. Approximately, ten fold lower IC50 value was noted in cells treated with the combination of resveratrol (3µM) and curcumin (3µM) in comparison to 30µM of resveratrol or curcumin alone. Resveratrol+curcumin combination caused apoptosis by increasing Bax/Bcl-xL ratio, Cytochrome C release, cleaved product of PARP and caspase 3 in cells. Interestingly, this combination unaltered the protein expressions of WNT-TCF and Notch signaling components, ß-catenin and cleaved notch-1 val1744, respectively. Furthermore, the combination also significantly decreased the intermediates of Hedgehog-Gli cascade including SMO, SHH, Gli-1, c-MYC, Cyclin-D1, etc. and increased the level of p21(Waf/Cip1) in vitro and in vivo. A significant reduction of Gli- promoter activity was noted in combinational drug treated cells in comparison to individual drug treatment. Un-alteration of the expressions of the above proteins and Gli1 promoter activity in p21(Waf/Cip1) knockout cells suggests this combination caused apoptosis through p21(Waf/Cip1). Thus, our findings revealed resveratrol and curcumin synergistically caused apoptosis in cigarette smoke induced breast cancer cells through p2(Waf/Cip1) mediated inhibition of Hedgehog-Gli cascade.


Subject(s)
Apoptosis/drug effects , Curcumin/pharmacology , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Epithelial Cells/drug effects , Hedgehog Proteins/metabolism , Stilbenes/pharmacology , Transcription Factors/metabolism , Animals , Antineoplastic Agents/pharmacology , Breast Neoplasms/drug therapy , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Cell Line , Cell Transformation, Neoplastic/drug effects , Cyclin-Dependent Kinase Inhibitor p21/genetics , Drug Synergism , Epithelial Cells/metabolism , Epithelial Cells/pathology , Female , Humans , Immunoblotting , Mice, Inbred BALB C , Microscopy, Fluorescence , RNA Interference , Resveratrol , Signal Transduction/drug effects , Smoke , Nicotiana/chemistry , Nicotiana/toxicity , Transcription Factors/genetics , Xenograft Model Antitumor Assays , Zinc Finger Protein GLI1 , bcl-2-Associated X Protein/metabolism , bcl-X Protein/metabolism
14.
PLoS One ; 10(5): e0125709, 2015.
Article in English | MEDLINE | ID: mdl-25938234

ABSTRACT

Pentacyclic Triterpenoids (PTs) and their analogues as well as derivatives are emerging as important drug leads for various diseases. They act through a variety of mechanisms and a majority of them inhibit the nuclear factor kappa-beta (NF-κB) signaling pathway. In this study, we examined the effects of the naturally occurring PTs on IκB kinase-ß (IKKß), which has great scientific relevance in the NF-κB signaling pathway. On virtual screening, 109 PTs were screened through the PASS (prediction of activity spectra of substances) software for prediction of NF-κB inhibitory activity followed by docking on the NEMO/IKKß association complex (PDB: 3BRV) and testing for compliance with the softened Lipinski's Rule of Five using Schrodinger (LLC, New York, USA). Out of the projected 45 druggable PTs, Corosolic Acid (CA), Asiatic Acid (AA) and Ursolic Acid (UA) were assayed for IKKß kinase activity in the cell free medium. The UA exhibited a potent IKKß inhibitory effect on the hotspot kinase assay with IC50 of 69 µM. Whereas, CA at 50 µM concentration markedly reduced the NF-κB luciferase activity and phospho-IKKß protein expressions. The PTs tested, attenuated the expression of the NF-κB cascade proteins in the LPS-stimulated RAW 264.7 cells, prevented the phosphorylation of the IKKα/ß and blocked the activation of the Interferon-gamma (IFN-γ). The results suggest that the IKKß inhibition is the major mechanism of the PTs-induced NF-κB inhibition. PASS predictions along with in-silico docking against the NEMO/IKKß can be successfully applied in the selection of the prospective NF-κB inhibitory downregulators of IKKß phosphorylation.


Subject(s)
I-kappa B Kinase/metabolism , NF-kappa B/metabolism , Pentacyclic Triterpenes/pharmacology , Signal Transduction/drug effects , Animals , Cell Line , Cell Survival/drug effects , Computer Simulation , Dose-Response Relationship, Drug , Drug Discovery/methods , Enzyme Activation/drug effects , Gene Expression , Genes, Reporter , Humans , Hydrogen Bonding , I-kappa B Kinase/antagonists & inhibitors , I-kappa B Kinase/chemistry , Interferon-gamma/biosynthesis , Lipopolysaccharides/immunology , Macrophages/drug effects , Macrophages/immunology , Macrophages/metabolism , Mice , Molecular Conformation , Molecular Docking Simulation , Molecular Dynamics Simulation , NF-kappa B/antagonists & inhibitors , NF-kappa B/chemistry , Pentacyclic Triterpenes/chemistry , Protein Binding , Small Molecule Libraries
15.
Pathol Oncol Res ; 21(2): 405-11, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25359126

ABSTRACT

Here, we report the p53 dependent mitochondria-mediated apoptotic mechanism of plant derived silver-nanoparticle (PD-AgNPs) in colorectal cancer cells (CRCs). PD-AgNPs was synthesized by reduction of AgNO3 with leaf extract of a medicinal plant periwinkle and characterized. Uptake of PD-AgNPs (ξ - 2.52 ± 4.31 mV) in HCT116 cells was 3 fold higher in comparison to synthetic AgNPs (ξ +2.293 ± 5.1 mV). A dose dependent increase in ROS production, activated JNK and decreased mitochondrial membrane potential (MMP) were noted in HCT116 but not in HCT116 p53(-/-) cells after PD-AgNP exposure. PD-AgNP-mediated apoptosis in CRCs is a p53 dependent process involving ROS and JNK cascade.


Subject(s)
Apoptosis/drug effects , Colonic Neoplasms/pathology , MAP Kinase Signaling System/drug effects , Nanoparticles , Plant Extracts/pharmacology , Reactive Oxygen Species/metabolism , Silver/pharmacology , Tumor Suppressor Protein p53/metabolism , Antineoplastic Agents/pharmacology , Apoptosis/physiology , Cell Line, Tumor , Cell Survival/drug effects , Colonic Neoplasms/metabolism , Cytochromes c/metabolism , Dose-Response Relationship, Drug , HCT116 Cells , Humans , MAP Kinase Signaling System/physiology , Membrane Potential, Mitochondrial/drug effects , Membrane Potential, Mitochondrial/physiology , Mitogen-Activated Protein Kinase Kinases/metabolism , Treatment Outcome
16.
DNA Repair (Amst) ; 24: 15-25, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25460919

ABSTRACT

Colorectal cancer (CRC) patients with APC mutations do not benefit from 5-FU therapy. It was reported that APC physically interacts with POLß and FEN1, thus blocking LP-BER via APC's DNA repair inhibitory (DRI) domain in vitro. The aim of this study was to elucidate how APC status affects BER and the response of CRC to 5-FU. HCT-116, HT-29, and LOVO cells varying in APC status were treated with 5-FU to evaluate expression, repair, and survival responses. HCT-116 expresses wild-type APC; HT-29 expresses an APC mutant that contains DRI domain; LOVO expresses an APC mutant lacking DRI domain. 5-FU increased the expression of APC and decreased the expression of FEN1 in HCT-116 and HT-29 cells, which were sensitized to 5-FU when compared to LOVO cells. Knockdown of APC in HCT-116 rendered cells resistant to 5-FU, and FEN1 levels remained unchanged. Re-expression of full-length APC in LOVO cells caused sensitivity to 5-FU, and decreased expression of FEN1. These knockdown and addback studies confirmed that the DRI domain is necessary for the APC-mediated reduction in LP-BER and 5-FU. Modelling studies showed that 5-FU can interact with the DRI domain of APC via hydrogen bonding and hydrophobic interactions. 5-FU resistance in CRC occurs with mutations in APC that disrupt or eliminate the DRI domain's interaction with LP-BER. Understanding the type of APC mutation should better predict 5-FU resistance in CRC than simply characterizing APC status as wild-type or mutant.


Subject(s)
Adenomatous Polyposis Coli Protein/metabolism , Colonic Neoplasms/drug therapy , DNA Repair/drug effects , Fluorouracil/pharmacology , Adenomatous Polyposis Coli Protein/chemistry , Adenomatous Polyposis Coli Protein/genetics , Amino Acid Sequence , Antimetabolites, Antineoplastic/pharmacology , Cell Line, Tumor/drug effects , Colonic Neoplasms/genetics , Flap Endonucleases/metabolism , Gene Knockdown Techniques , HCT116 Cells/drug effects , Humans , Hydrogen Bonding , Molecular Docking Simulation , Molecular Sequence Data , Proteasome Endopeptidase Complex/metabolism , Protein Structure, Tertiary
17.
Eur J Med Chem ; 85: 95-106, 2014 Oct 06.
Article in English | MEDLINE | ID: mdl-25078313

ABSTRACT

A new family of andrographolide analogues were synthesized and screened in vitro against kidney (HEK-293) and breast (MCF-7) cancer cells. The anti-cancer effects of the active analogues (2b, 2c and 4c) were determined by multiple cell based assays such as MTT, immunostaining, FACS, western blotting and transcriptional inhibition of NF-κB activity. Importantly, these compounds were found to possess higher anti-cancer potency than andrographolide and low toxicity to normal (VERO and MCF-10A) cells. Increased level of Bax/Bcl-xL ratio, caspase 3, and sub G1 population, higher expression level of tumor suppressor protein p53 and lower expression level of NF-κB suggested potent apoptotic property of the active analogues. Data revealed that the andrographolide derivative-mediated cell death in cancer cells was p53 dependent.


Subject(s)
Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/pharmacology , Diterpenes/chemical synthesis , Diterpenes/pharmacology , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/metabolism , Caspase 3/metabolism , Cell Line, Tumor , Chemistry Techniques, Synthetic , Diterpenes/chemistry , Diterpenes/metabolism , Glutathione/metabolism , Half-Life , Humans , Hydrolysis , NF-kappa B/metabolism , Transcription, Genetic/drug effects
18.
Appl Biochem Biotechnol ; 174(5): 1850-8, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25149458

ABSTRACT

Two different microbial biosurfactants S9BS and CHBS were isolated from Lysinibacillus fusiformis S9 and Bacillus tequilensis CH. Cytotoxicity effect of these biosurfactants on human embryonic kidney cancerous cell (HEK-293) were studied with the help of 3-(4,5-dimethylthiazol-2yl-)-2, 5-diphenyl tetrazolium bromide (MTT) assay and morphological changes were observed under inverted microscope. The biosurfactants exhibited positive cytotoxic effect on HEK-293 cell line. It was found that LC50 of S9BS and CHBS were 75 and 100 µg ml(-1), respectively. Further cell cycle and apoptosis analysis of biosurfactant-treated HEK-293 cell line were done by FACS. In this study, cytotoxic effect of glycolipid biosurfactant against HEK-293 cell lines is reported for the first time. Mechanism towards increased membrane permeability of biosurfactant-treated cancer cell may be the incorporation of its lipid moiety into the plasma membrane leading to formation of pores and membrane disruption. Hence, these microbial biosurfactants can prove to be significant biomolecule for cancer treatment.


Subject(s)
Apoptosis/drug effects , Bacillus/metabolism , Kidney Neoplasms/drug therapy , Kidney Neoplasms/pathology , Surface-Active Agents/administration & dosage , Bacillus/classification , Dose-Response Relationship, Drug , HEK293 Cells , Humans , Lethal Dose 50
19.
Toxicol Appl Pharmacol ; 275(3): 221-31, 2014 Mar 15.
Article in English | MEDLINE | ID: mdl-24467951

ABSTRACT

Cigarette smoking is a key factor for the development and progression of different cancers including mammary tumor in women. Resveratrol (Res) is a promising natural chemotherapeutic agent that regulates many cellular targets including p21, a cip/kip family of cyclin kinase inhibitors involved in DNA damage-induced cell cycle arrest and blocking of DNA replication and repair. We have recently shown that cigarette smoke condensate (CSC) prepared from commercially available Indian cigarette can cause neoplastic transformation of normal breast epithelial MCF-10A cell. Here we studied the mechanism of Res mediated apoptosis in CSC transformed (MCF-10A-Tr) cells in vitro and in vivo. Res mediated apoptosis in MCF-10A-Tr cells was a p21 dependent event. It increased the p21 protein expression in MCF-10A-Tr cells and MCF-10A-Tr cells-mediated tumors in xenograft mice. Res treatment reduced the tumor size(s) and expression of anti-apoptotic proteins (e.g. PI3K, AKT, NFκB) in solid tumor. The expressions of cell cycle regulatory (Cyclins, CDC-2, CDC-6, etc.), BER associated (Pol-ß, Pol-δ, Pol-ε, Pol-η, RPA, Fen-1, DNA-Ligase-I, etc.) proteins and LP-BER activity decreased in MCF-10A-Tr cells but remain significantly unaltered in isogenic p21 null MCF-10A-Tr cells after Res treatment. Interestingly, no significant changes were noted in SP-BER activity in both the cell lines after Res exposure. Finally, it was observed that increased p21 blocks the LP-BER in MCF-10A-Tr cells by increasing its interaction with PCNA via competing with Fen-1 after Res treatment. Thus, Res caused apoptosis in CSC-induced cancer cells by reduction of LP-BER activity and this phenomenon largely depends on p21.


Subject(s)
Antineoplastic Agents/pharmacology , Breast Neoplasms/drug therapy , Cell Transformation, Neoplastic/drug effects , Cyclin-Dependent Kinase Inhibitor p21/metabolism , DNA Repair/drug effects , Epithelial Cells/drug effects , Mammary Glands, Human/drug effects , Smoke/adverse effects , Smoking/adverse effects , Stilbenes/pharmacology , Animals , Apoptosis/drug effects , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Cycle/drug effects , Cell Death/drug effects , Cell Line, Transformed , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/metabolism , Cell Transformation, Neoplastic/pathology , Cyclin-Dependent Kinase Inhibitor p21/genetics , DNA Damage/drug effects , Dose-Response Relationship, Drug , Epithelial Cells/metabolism , Epithelial Cells/pathology , Female , Flap Endonucleases/metabolism , Humans , Mammary Glands, Human/metabolism , Mammary Glands, Human/pathology , Mice , Mice, Inbred BALB C , Proliferating Cell Nuclear Antigen/metabolism , RNA Interference , Resveratrol , Signal Transduction/drug effects , Time Factors , Transfection , Up-Regulation , Xenograft Model Antitumor Assays
20.
Toxicol Appl Pharmacol ; 274(1): 168-79, 2014 Jan 01.
Article in English | MEDLINE | ID: mdl-24099783

ABSTRACT

Cigarette smoking is a crucial factor in the development and progression of multiple cancers including breast. Here, we report that repeated exposure to a fixed, low dose of cigarette smoke condensate (CSC) prepared from Indian cigarettes is capable of transforming normal breast epithelial cells, MCF-10A, and delineate the biochemical basis for cellular transformation. CSC transformed cells (MCF-10A-Tr) were capable of anchorage-independent growth, and their anchorage dependent growth and colony forming ability were higher compared to the non-transformed MCF-10A cells. Increased expression of biomarkers representative of oncogenic transformation (NRP-1, Nectin-4), and anti-apoptotic markers (PI3K, AKT, NFκB) were also noted in the MCF-10A-Tr cells. Short tandem repeat (STR) profiling of MCF-10A and MCF-10A-Tr cells revealed that transformed cells acquired allelic variation during transformation, and had become genetically distinct. MCF-10A-Tr cells formed solid tumors when implanted into the mammary fat pads of Balb/c mice. Data revealed that CSC contained approximately 1.011µg Cd per cigarette equivalent, and Cd (0.0003µg Cd/1×10(7) cells) was also detected in the lysates from MCF-10A cells treated with 25µg/mL CSC. In similar manner to CSC, CdCl2 treatment in MCF-10A cells caused anchorage independent colony growth, higher expression of oncogenic proteins and increased PI3K-AKT-NFκB protein expression. An increase in the expression of PI3K-AKT-NFκB was also noted in the mice xenografts. Interestingly, it was noted that CSC and CdCl2 treatment in MCF-10A cells increased ROS. Collectively, results suggest that heavy metals present in cigarettes of Indian origin may substantially contribute to tumorigenesis by inducing intercellular ROS accumulation and increased expression of PI3K, AKT and NFκB proteins.


Subject(s)
Cell Transformation, Neoplastic/chemically induced , Epithelial Cells/drug effects , Metals, Heavy/toxicity , NF-kappa B/biosynthesis , Phosphatidylinositol 3-Kinase/biosynthesis , Proto-Oncogene Proteins c-akt/biosynthesis , Smoke/adverse effects , Animals , Breast/drug effects , Breast/metabolism , Breast/pathology , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Line, Transformed , Cell Transformation, Neoplastic/metabolism , Cell Transformation, Neoplastic/pathology , Dose-Response Relationship, Drug , Epithelial Cells/metabolism , Epithelial Cells/pathology , Female , Humans , Mice , Mice, Inbred BALB C , Smoking/adverse effects , Xenograft Model Antitumor Assays/methods
SELECTION OF CITATIONS
SEARCH DETAIL
...