Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 21
1.
Biochem Pharmacol ; 223: 116126, 2024 May.
Article En | MEDLINE | ID: mdl-38490521

Anastasis cascade including induction of Epithelial to Mesenchymal Transition (EMT), DNA repair, and stimulation of pro-survival mediators collectively exaggerate therapy resistance in cancer prognosis. The extensive implications of DNA-damaging agents are clinically proven futile for the rapid development of disease recurrence during treatment regime. Herein we report a glycosidic derivative of Δ9-tetrahydrocannabinol (THC-9-OG) abrogates sub-toxic doses of 5-Fluorouracil (5FU) induced EMT in colon cancer cells nullifying DNA repairing mechanism. Our in vitro and in vivo data strongly proclaims that THC-9-OG could not only abrogate 5FU mediated background EMT activation through stalling matrix degradation as well as murine 4T1 lung metastasis but also vigorously diminished Rad-51 repairing mediator along with stimulation of γ-H2AX foci formation. The combinatorial treatment (5FU + THC-9-OG) in Apc knockout colorectal carcinoma model conferred remission of the crypt progenitor phenotype which was prominently identified in 5FU treatment. Mechanistically, we demonstrated that 5FU plus THC-9-OG significantly attenuated major EMT inducer Vimentin via extensive ROS generation along with autophagy induction via LC3B I-II conversion and p62 degradation in a p-ATM dependent manner. Additionally, Cannabinoid receptor CB1 was responsible for abrogation of Vimentin since we found increase in the expression of γH2AX and decrease in vimentin expression in CB1 agonist (ACEA) plus 5FU treated cells. Nutshell, our results unveil a new direction of Cannabinoid based combinatorial approach to control background EMT along with robust enhancing of DNA damage potential of sub-toxic concentration of 5FU resulting immense inhibition of distant metastasis coupled with triggering cell death in vitro and in vivo.


Cannabinoids , Humans , Animals , Mice , Fluorouracil/pharmacology , Epithelial-Mesenchymal Transition , Vimentin/genetics , Vimentin/metabolism , Cell Line, Tumor , Neoplasm Recurrence, Local , Autophagy , DNA
2.
Front Oncol ; 12: 1013500, 2022.
Article En | MEDLINE | ID: mdl-36465376

ATF-4 is a master regulator of transcription of genes essential for cellular-adaptive function. In response to the quantum and duration of stress, ATF-4 diligently responds to both pro-apoptotic and pro-survival signals converging into either autophagy or apoptosis/senescence. Despite emerging cues implying a relationship between autophagy and senescence, how these two processes are controlled remains unknown. Herein, we demonstrate ß-(4-fluorobenzyl) Arteannuin B (here after Arteannuin 09), a novel semisynthetic derivative of Arteannuin B, as a potent ER stress inducer leading to the consistent activation of ATF-4. Persistent ATF-4 expression at early time-points facilitates the autophagy program and consequently by upregulating p21 at later time-points, the signaling is shifted towards G2/M cell cycle arrest. As bZIP transcription factors including ATF-4 are obligate dimers, and because ATF-4 homodimers are not highly stable, we hypothesized that ATF-4 may induce p21 expression by physically interacting with another bZIP family member i.e., C/EBPß. Our co-immunoprecipitation and co-localization studies demonstrated that ATF-4 is principally responsible for the autophagic potential of Arteannuin 09, while as, induction of both ATF-4 and C/EBPß is indispensable for the p21 regulated-cell cycle arrest. Interestingly, inhibition of autophagy signaling switches the fate of Arteannuin 09 treated cells from senescence to apoptosis. Lastly, our data accomplished that Arteannuin 09 is a potent inhibitor of tumor growth and inducer of premature senescence in vivo.

3.
ACS Appl Bio Mater ; 4(1): 470-482, 2021 01 18.
Article En | MEDLINE | ID: mdl-35014298

Over the past few years, nanotechnology-based approaches have emerged to override drug resistance owing to their superiority over other formulations because of their diverse therapeutic advantages such as target-specific drug delivery, enhanced bioavailability, biodegradability, and minimal off-target effects. Hybrid nanomaterials as a formulation of anticancer drugs with gold nanoparticles (AuNPs) have adequately proven efficacious in controlled release as well as disintegration into ultrasmall nanoparticles dragging the drug to penetrate deep into tumor tissues and consequently getting cleared from the body. In this study, to achieve better antitumor responses, we engineered self-assembled organic nanoparticles of potent anticancer compound BZ6 (BZ6-ONPs), BZ6-gold nanoparticle conjugates (BZ6-AuNPs), and organic-inorganic nanohybrids involving amalgamation of AuNPs with BZ6-ONPs (AuNPs@BZ6-ONPs) and comparatively analyzed their physicochemical as well as biological activities. The epithelial-mesenchymal transition (EMT) is a critical biological event that facilitates metastatic spread of cancer cells and contributes to chemoresistance. AuNPs@BZ6-ONPs consistently suppressed EMT characteristics including invasion, cell scattering, and migration abilities of aggressive breast cancer (MDA-MB-231) and pancreatic adenocarcinoma (PANC-1) cells much more efficiently than BZ6-ONPs and BZ6-AuNPs. Western blotting and immunocytochemistry analysis unveiled that the nanohybrids downregulated expression of the key mesenchymal markers NF-κß p65, Twist-1, vimentin, and MMP-2, meanwhile augmenting epithelial marker E-cadherin and tumor suppressor Par-4. The in vivo syngenic mouse tumor model demonstrated remarkable reduction of tumor growth (84.3%) and metastatic lung nodules (66.1%) following 14 days of treatment without any adverse effects. Finally, the facile and ecofriendly method of synthesis of AuNPs@BZ6-ONPs demonstrating promising antitumor/antimetastatic efficacies suggests its therapeutic implication for the treatment of advanced cancers.


Antineoplastic Agents/chemistry , Benzimidazoles/chemistry , Epithelial-Mesenchymal Transition , Gold/chemistry , Metal Nanoparticles/chemistry , Animals , Antineoplastic Agents/metabolism , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Benzimidazoles/metabolism , Benzimidazoles/pharmacology , Benzimidazoles/therapeutic use , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Down-Regulation/drug effects , Epithelial-Mesenchymal Transition/drug effects , Female , Humans , Metal Nanoparticles/toxicity , Mice , Mice, Inbred BALB C , Neoplasms/drug therapy , Transcription Factor RelA/genetics , Transcription Factor RelA/metabolism , Vimentin/genetics , Vimentin/metabolism
4.
Eur J Cell Biol ; 99(4): 151076, 2020 May.
Article En | MEDLINE | ID: mdl-32439219

Deregulation of TGF-ß signaling is intricately engrossed in the pathophysiology of pancreatic adenocarcinomas (PDACs). The role of TGF-ß all through pancreatic cancer initiation and progression is multifarious and somewhat paradoxical. TGF-ß plays a tumor suppressive role in early-stage pancreatic cancer by promoting apoptosis and inhibiting epithelial cell cycle progression, but incites tumor promotion in late-stage by modulating genomic instability, neo-angiogenesis, immune evasion, cell motility, and metastasis. Here, we provide evidences that Par-4 acts as one of the vital mediators to regulate TGF-ß/Smad4 pathway, wherein, Par-4 induction/over-expression induced EMT which was later culminated in to apoptosis in presence of TGF-ß via positive regulation of Smad4. Intriguingly, Par-4-/- cells were devoid of significant Smad4 induction compared to Par-4+/+ cells in presence of TGF-ß and ectopic Par-4 steadily augmented Smad4 expression by restoring TGF-ß/Smad4 axis in Panc-1 cells. Further, our FACS and western blotting results unveiled that Par-4 dragged the PDAC cells to G1 arrest in presence of TGF-ß byelevating p21 and p27 levels while attenuating Cyclin E and A levels and augmenting caspase 3 cleavage triggering lethal EMT. Through restoration of Smad4, we further establish that in BxPC3 cell line (Smad4-/-), Smad4 is essential for Par-4 to indulge TGF-ß dependent lethal EMT program. The mechanistic relevance of Par-4 mediated Smad4 activation was additionally validated by co-immunoprecipitation wherein disruption of NM23H1-STRAP interaction by Par-4 rescues TGF-ß/Smad4 pathway in PDAC and mediates the tumor suppressive role of TGF-ß, therefore serving as a vital cog to restore the apoptotic functions of TGF-ß pathway.


Carcinoma, Pancreatic Ductal/metabolism , Pancreatic Neoplasms/metabolism , Receptors, Thrombin/metabolism , Smad4 Protein/metabolism , Transforming Growth Factor beta/metabolism , Carcinoma, Pancreatic Ductal/pathology , Cell Line, Tumor , Epithelial-Mesenchymal Transition , G1 Phase Cell Cycle Checkpoints , Humans , NM23 Nucleoside Diphosphate Kinases/genetics , NM23 Nucleoside Diphosphate Kinases/metabolism , Pancreatic Neoplasms/pathology , Plasmids/genetics , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Proto-Oncogene Proteins c-akt/metabolism , RNA, Small Interfering/administration & dosage , RNA, Small Interfering/genetics , RNA-Binding Proteins/metabolism , Receptors, Thrombin/genetics , Signal Transduction , Smad4 Protein/biosynthesis , Smad4 Protein/genetics , Up-Regulation
5.
Cancer Metastasis Rev ; 39(2): 553-566, 2020 06.
Article En | MEDLINE | ID: mdl-32020420

Apoptosis is a tightly controlled, coordinated cellular event responsible for inducing programmed cell death to rid the body of defective or unfit cells. Inhibition of apoptosis is, therefore, an essential process for cancer cells to harness. Genomic variants in apoptotic-controlling genes are highly prevalent in cancer and have been identified to induce pro-proliferation and pro-survival pathways, rendering cancer cells resistant to apoptosis. Traditional understanding of apoptosis defines it as an irreversible process; however, growing evidence suggests that apoptosis is a reversible process from which cells can escape, even after the activation of its most committed stages. The mechanism invoked to reverse apoptosis has been termed anastasis and poses challenges for the development and utilization of chemotherapeutic agents. Anastasis has also been identified as a mechanism by which cells can recover from apoptotic lesions and revert back to its previous functioning state. In this review, we intend to focus the attention of the reader on the comprehensive role of survival, metastasis, and epithelial mesenchymal transition (EMT), as well as DNA damage repair mechanisms in promoting anastasis. Additionally, we will emphasize the mechanistic consequences of anastasis on drug resistance and recent rational therapeutic approaches designed to combat this resistance.


Apoptosis/physiology , Neoplasms/pathology , Animals , Cell Survival/physiology , DNA Damage , DNA Repair , Drug Resistance, Neoplasm , Epithelial-Mesenchymal Transition , Humans , Neoplasms/drug therapy , Neoplasms/genetics , Neoplasms/metabolism
6.
Front Genet ; 11: 609758, 2020.
Article En | MEDLINE | ID: mdl-33584808

Breast cancer has replaced cervical cancer as being the most common and having the highest mortality among women in India. ANKLE gene is conserved among organisms during evolutionary succession and is a member of LEM family proteins in lower metazoans and is involved in critical functions in the nuclear architecture, gene expression and cell signaling. ANKLE1 is the human orthologous of LEM-3 and is involved in DNA damage response and DNA repair. Whole Exome Sequencing (WES) of paired breast cancer samples was performed and ANKLE1 was found to be a new possible hotspot for predisposition of breast cancer. The mass array genotyping for breast cancer variant rs2363956 further confirmed the ANKLE1 association with the studied population of breast cancer. To elucidate the role of ANKLE1 in DNA damage, it was knocked down in MCF-7 breast cancer cell line and the expression of γH2AX was assessed. ANKLE1 knockdown cells displayed elevated levels of γ-H2AX foci in response to the cisplatin induced replication stress. The localization pattern of ANKLE1 further emphasized the role of ANKLE1 in DNA repair process. We observed that ANKLE1 is required for maintaining genomic stability and plays a role in DNA damage and repair process. These findings provided a molecular basis for the suspected role of ANKLE1 in human breast cancer and suggested an important role of this gene in controlling breast cancer development among women in India.

7.
Cell Death Dis ; 10(6): 467, 2019 06 13.
Article En | MEDLINE | ID: mdl-31197132

Epithelial to mesenchymal transitions (EMT) is a preparatory process for cancer cells to attain motility and further metastasis to distant sites. Majority of DNA damaging drugs have shown to develop EMT as one of the major mechanisms to attain drug resistance. Here we sought to understand the resistance/survival instincts of cancer cells during initial phase of drug treatment. We provide a tangible evidence of stimulation of EMT factors in Apc knockout colorectal carcinoma model. Our results implied that CPT-treated Apc knockout cohorts depicted increased pro-invasive and pro-survival factors (Vimentin/pser38Vimentin & NFκB). Moreover, by cell sorting experiment, we have observed the expression of Vimentin in early apoptotic cells (AnnexinV positive) from 36 to 48 h of CPT treatment. We also observed the expression of chimeric Sec-AnnexinV-mvenus protein in migrated cells on transwell membrane recapitulating signatures of early apoptosis. Notably, induction of Vimentin-mediated signaling (by CPT) delayed apoptosis progression in cells conferring survival responses by modulating the promoter activity of NFκB. Furthermore, our results unveiled a novel link between Vimentin and ATM signaling, orchestrated via binding interaction between Vimentin and ATM kinase. Finally, we observed a significant alteration of crypt-villus morphology upon combination of DIM (EMT inhibitor) with CPT nullified the background EMT signals thus improving the efficacy of the DNA damaging agent. Thus, our findings revealed a resistance strategy of cancer cells within a very initial period of drug treatment by activating EMT program, which hinders the cancer cells to achieve later phases of apoptosis thus increasing the chances of early migration.


Apoptosis , Colorectal Neoplasms/metabolism , DNA Damage , Epithelial-Mesenchymal Transition , Vimentin/metabolism , Adenomatous Polyposis Coli Protein/genetics , Animals , Apoptosis/drug effects , Apoptosis/genetics , Ataxia Telangiectasia Mutated Proteins/genetics , Ataxia Telangiectasia Mutated Proteins/metabolism , Camptothecin/administration & dosage , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Proliferation/genetics , Cell Survival/drug effects , Cell Survival/genetics , Colorectal Neoplasms/enzymology , Colorectal Neoplasms/genetics , Colorectal Neoplasms/pathology , Epithelial-Mesenchymal Transition/drug effects , Epithelial-Mesenchymal Transition/genetics , Gene Expression Regulation, Neoplastic/genetics , Humans , Mice , Mice, Knockout , NF-kappa B/metabolism , Signal Transduction/drug effects , Vimentin/genetics
8.
Breast Cancer Res Treat ; 177(2): 307-323, 2019 Sep.
Article En | MEDLINE | ID: mdl-31175498

PURPOSE: More than 90% of the breast cancer deaths occur due to the metastasis of the cancer cells to secondary organ sites. Increased Glucose-regulated protein 78 (GRP78) expression is critical for epithelial-mesenchymal transition (EMT) and invasion in breast cancer resulting in poor patient survival outcomes. Therefore, there is an urgent need of potential inhibitors of GRP78 for the abrogation of invasion and metastasis in breast cancer. METHODS: We investigated the effect of IKM5 (2-(1-(1H-indol-3-yl)octyl)-3-hydroxy-6-(hydroxymethyl)-4H-pyran-4-one) (a novel Indolylkojyl methane analogue) on invasion abilities of human breast cancer cells employing invadopodia formation, Matrigel invasion assays, and mouse models for metastasis. The mechanism underlying the anti-invasive effect of IKM5 was examined through molecular docking, immunoblotting, immunocytochemistry, co-immunoprecipitation analysis, siRNA silencing, and sub-cellular fractionation studies. RESULTS: Treatment with IKM5 at its sub-toxic concentration (200 nM) suppressed invasion and invadopodia formation, and growth factor-induced cell scattering of aggressive human breast cancer MDA-MB-231, MDA-MB-468, and MCF7 cells. IKM5 spontaneously binds to GRP78 (Ki = 1.35 µM) and downregulates its expression along with the EMT markers MMP-2, Twist1, and Vimentin. Furthermore, IKM5 amplified the expression and nuclear translocation of tumor suppressor Par-4 to control NF-kB-mediated pro-EMT activities. Interestingly, IKM5 disrupts the interaction between GRP78 and TIMP-1 by inhibiting GRP78 in a Par-4-dependent manner. Moreover, IKM5 inhibited tumor growth and lung metastasis at a safe dose of 30 mg/kg/body weight. CONCLUSION: Our study warrants IKM5, a potential anticancer agent that can abrogate invasion and metastasis, suggesting its clinical development for the treatment of patients with advanced breast cancer.


Antineoplastic Agents/pharmacology , Heat-Shock Proteins/genetics , Methane/pharmacology , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacokinetics , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Cell Survival/drug effects , Disease Models, Animal , Doxorubicin/pharmacology , Endoplasmic Reticulum Chaperone BiP , Epithelial-Mesenchymal Transition/drug effects , Epithelial-Mesenchymal Transition/genetics , Female , Heat-Shock Proteins/chemistry , Heat-Shock Proteins/metabolism , Humans , Matrix Metalloproteinases , Methane/analogs & derivatives , Methane/chemistry , Methane/pharmacokinetics , Mice , Models, Biological , Models, Molecular , Molecular Conformation , Molecular Structure , Protein Transport , Structure-Activity Relationship , Xenograft Model Antitumor Assays
9.
Mater Sci Eng C Mater Biol Appl ; 97: 467-478, 2019 Apr.
Article En | MEDLINE | ID: mdl-30678934

Organic nanoparticles (ONPs) possess great research interests for their promising effects in the enhancement of bioactivity including anticancer activity with less toxicity. The present study describes the preparation, characterization and biological evaluation of aqueous phase ONPs of potent 1,2-disubstituted benzimidazole derivative (BZ6) for anticancer activity. BZ6-ONPs were characterized through UV-absorption and fluorescence spectroscopic analysis for their photo-physical properties. DLS, TEM and SEM studies were carried out for morphological and structural analysis. Cytotoxicity determination on a panel of four different cancer cell lines (MCF-7, MiaPaca-2, HT-29 and HCT-116) revealed that the BZ6-ONPs show highest activity in human breast cancer MCF-7 cells. Surprisingly, the BZ6-ONPs were found to be non-toxic towards normal breast epithelial fR2 cells. Additionally, the FITC-ONPs showed enhanced uptake in 3D tumor spheroids of MCF-7 cells compared to the free FITC. BZ6-ONPs strongly halted cell proliferation and induced apoptosis, possibly through oxidative stress-mediated reactive oxygen species (ROS) generation and loss of mitochondrial membrane potential (MMP) in MCF-7 cells. Moreover, molecular mechanism-based studies revealed that BZ6-ONPs downregulated AKT/NF-κB/vimentin/survivin-mediated oncogenic signaling pathway promoting cell proliferation and malignancy. In a nutshell, BZ6-ONPs are therapeutically efficacious, which needs further development as a treatment option in human mammary gland carcinomas.


Benzimidazoles/chemistry , Nanoparticles/chemistry , Oxidative Stress , Apoptosis/drug effects , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Line, Tumor , Cell Proliferation/drug effects , Female , Humans , Membrane Potential, Mitochondrial/drug effects , NF-kappa B/metabolism , Nanoparticles/toxicity , Oxidative Stress/drug effects , Proto-Oncogene Proteins c-akt/metabolism , Reactive Oxygen Species/metabolism , Signal Transduction/drug effects , Spheroids, Cellular/cytology , Spheroids, Cellular/drug effects , Spheroids, Cellular/metabolism
10.
Mol Carcinog ; 57(9): 1102-1115, 2018 09.
Article En | MEDLINE | ID: mdl-29672923

Epithelial-mesenchymal transition (EMT) is a critical event that occurs during the invasion and metastatic spread of cancer cells. Here, we conceive a dual mechanism of Par-4-mediated inhibition of EMT and induction of MET in metastatic pancreatic cancer cells. First, we demonstrate that 1,1'-ß-D-glucopyranosyl-3,3'-bis(5-bromoindolyl)-octyl methane (NGD16), an N-glycosylated derivative of medicinally important phytochemical 3,3'-diindolylmethane (DIM) abrogates EMT by inducing pro-apoptotic protein Par-4. Induction of Par-4 (by NGD16 or ectopic overexpression) strongly impedes invasion with inhibition of major mesenchymal markers viz. Vimentin and Twist-1 epithelial marker- E-cadherin. Further, NGD16 triggers MET phenotypes in pancreatic cancer cells by augmenting ALK2/Smad4 signaling in a Par-4-dependent manner. Conversely, siRNA-mediated silencing of endogenous Par-4 unveil reversal of MET with diminished E-cadherin expression and invasive phenotypes. Additionally, we demonstrate that intact Smad4 is essential for Par-4-mediated maintenance of E-cadherin level in MET induced cells. Notably, we imply that Par-4 induction regulates E-cadherin levels in the pancreatic cancer cells via modulating Twist-1 promoter activity. Finally, in vivo studies with syngenic mouse metastatic pancreatic cancer model reveal that NGD16 strongly suppresses metastatic burden, ascites formation, and prolongs the overall survival of animals effectively.


Apoptosis Regulatory Proteins/genetics , Epithelial-Mesenchymal Transition , Gene Expression Regulation, Neoplastic , Neoplasm Invasiveness/genetics , Pancreatic Neoplasms/genetics , Animals , Cadherins/genetics , Cell Line, Tumor , Epithelial-Mesenchymal Transition/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Humans , Indoles/pharmacology , Indoles/therapeutic use , Male , Mice, Inbred C57BL , Neoplasm Invasiveness/pathology , Neoplasm Invasiveness/prevention & control , Pancreatic Neoplasms/drug therapy , Pancreatic Neoplasms/pathology , RNA Interference , RNA, Small Interfering/genetics
11.
Cell Death Differ ; 24(7): 1275-1287, 2017 07.
Article En | MEDLINE | ID: mdl-28498365

Twist1, a basic helix-loop-helix transcription factor is implicated as a key mediator of epithelial-mesenchymal transition (EMT) and metastatic dissemination in p53-deficient cancer cells. On the other hand, checkpoint kinase 2 (Chk2), a major cell cycle regulatory protein provides a barrier to tumorigenesis due to DNA damage response by preserving genomic stability of the cells. Here we demonstrate that Chk2 induction proficiently abrogates invasion, cell scattering and invadopodia formation ability of p53-mutated invasive cells by suppressing Twist1, indicating Chk2 confers vital role in metastasis prevention. In addition, ectopic Chk2, as well as its (Chk2) induction by natural podophyllotoxin analog, 4'-demethyl-deoxypodophyllotoxin glucoside (4DPG), strongly restrain Twist1 activity along with other mesenchymal markers, for example, ZEB-1, vimentin and Snail1, whereas the epithelial markers such as E-cadherin and TIMP-1 expression augmented robustly. However, downregulation of endogenous Chk2 by siRNA as well as Chk2 selective inhibitor PV1019 implies that 4DPG-mediated inhibition of Twist1 is Chk2-dependent. Further, mechanistic studies unveil that Chk2 negatively regulates Twist1 promoter activity and it (Chk2) interacts steadily with Snail1 protein to curb EMT. Strikingly, Chk2 overexpression triggers premature senescence in these cells with distinctive increase in senescence-associated ß-galactosidase (SA-ß-gal) activity, G2/M cell cycle arrest and induction of senescence-specific marker p21waf1/Cip1. Importantly, stable knockdown of Twist1 by shRNA markedly augments p21 expression, its nuclear accumulation, senescence-associated heterochromatin foci (SAHF) and amplifies the number of SA-ß-gal-positive cells. Moreover, our in vivo studies also validate that 4DPG treatment significantly abrogates tumor growth as well as metastatic lung nodules formation by elevating the level of phospho-Chk2, Chk2 and suppressing Twist1 activity in mouse mammary carcinoma model. In a nutshell, this report conceives a novel strategy of Twist1 suppression through Chk2 induction, which prevents metastatic dissemination and promotes premature senescence in p53-defective invasive cancer cells.


Cellular Senescence , Checkpoint Kinase 2/metabolism , Nuclear Proteins/metabolism , Tumor Suppressor Protein p53/metabolism , Twist-Related Protein 1/metabolism , Animals , Biomarkers, Tumor/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Cellular Senescence/drug effects , Cellular Senescence/genetics , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Epithelial-Mesenchymal Transition/drug effects , Epithelial-Mesenchymal Transition/genetics , Female , Gene Expression Regulation, Neoplastic/drug effects , Gene Knockdown Techniques , Glucosides/chemistry , Glucosides/pharmacology , Humans , Mice , Models, Biological , Neoplasm Invasiveness , Neoplasm Metastasis , Nuclear Proteins/genetics , Podophyllotoxin/analogs & derivatives , Podophyllotoxin/chemistry , Podophyllotoxin/pharmacology , Promoter Regions, Genetic/genetics , Transcription, Genetic/drug effects , Twist-Related Protein 1/genetics
12.
Eur J Cell Biol ; 96(2): 164-171, 2017 Mar.
Article En | MEDLINE | ID: mdl-28216015

Multiple stresses are prevalent inside the tumor microenvironment rendering tumor growth, neighboring invasion and metastasis of the cancer cells to distant organs. NM23-H1 is the first metastasis suppressor gene identified and known to be implicated as an important regulator of stress-induced metastasis. Herein, we demonstrated that prototypical NM23-H1 expression diminished during hypoxia and serum starvation in Panc-1/MDA-MB-231 cells, but converse invasion patterns were obtained in these two diverse stresses. Supportingly, a compelling discrete difference in mRNA and protein levels of NM23-H1 was achieved in hypoxia as well as serum starvation. Knockdown of NM23-H1 activates EMT whereas the similar effects are subdued in serum starvation where NM23-H1 down-modulation prompted E-cadherin upregulation. Stable NM23-H1 expression augmented E-cadherin levels along with retardation in invadopodea formation and invasion. In hypoxia/serum starvation excess NM23-H1 effectively modulated the Twist1 promoter activity. Thus, differential regulation of NM23-H1 may corroborate/abrogate EMT depending on the nature of stress, tumor microenvironment and cellular context.


NM23 Nucleoside Diphosphate Kinases/metabolism , Neoplasms/enzymology , Neoplasms/pathology , Cell Differentiation/physiology , Cell Hypoxia/physiology , Cell Line, Tumor , Epithelial-Mesenchymal Transition , Gene Knockdown Techniques , Humans , MCF-7 Cells , NM23 Nucleoside Diphosphate Kinases/genetics , Neoplasm Metastasis , Neoplasms/genetics , Transfection , Tumor Microenvironment
13.
Clin Exp Metastasis ; 33(8): 757-764, 2016 12.
Article En | MEDLINE | ID: mdl-27568374

Prostate apoptotic response 4 (Par-4) is coined as a therapeutic protein since owing to its diverse physiologically relevant properties, especially in the cancer perspective. Albeit, Par-4 expression is not restricted to any specific tissue/organ, apart from cell death promotion (due to challenging threats), the other biological role of Par-4 is convincingly emerging. In the recent years, several laboratories have intended to dissect the signaling or mechanisms involved in Par-4 activation to augment apoptosis cascades but new developments in Par-4 research have widened its therapeutic potential. One of these important avenues is the prevention of metastasis by pro-apoptotic Par-4. In this review, we will focus on the therapeutic perspective of Par-4 with a special reference to its (Par-4) virgin prospect of devastating metastasis control.


Apoptosis Regulatory Proteins/genetics , Apoptosis/genetics , Neoplasm Metastasis/genetics , Apoptosis Regulatory Proteins/biosynthesis , Gene Expression Regulation, Neoplastic/genetics , Humans , Signal Transduction
14.
Age (Dordr) ; 38(3): 62, 2016 Jun.
Article En | MEDLINE | ID: mdl-27246693

Stress-induced premature senescence (SIPS) is quite similar to replicative senescence that is committed by cells exposed to various stress conditions viz. ultraviolet radiation (DNA damage), hydrogen peroxide (oxidative stress), chemotherapeutic agents (cytotoxic threat), etc. Here, we report that cristacarpin, a natural product obtained from the stem bark of Erythrina suberosa, promotes endoplasmic reticulum (ER) stress, leading to sub-lethal reactive oxygen species (ROS) generation and which eventually terminates by triggering senescence in pancreatic and breast cancer cells through blocking the cell cycle in the G1 phase. The majority of cristacarpin-treated cells responded to conventional SA-ß-gal stains; showed characteristic p21(waf1) upregulation along with enlarged and flattened morphology; and increased volume, granularity, and formation of heterochromatin foci-all of these features are the hallmarks of senescence. Inhibition of ROS generation by N-acetyl-L-cysteine (NAC) significantly reduced the expression of p21(waf1), confirming that the modulation in p21(waf1) by anti-proliferative cristacarpin was ROS dependent. Further, the elevation in p21(waf1) expression in PANC-1 and MCF-7 cells was consistent with the decrease in the expression of Cdk-2 and cyclinD1. Here, we provide evidence that cristacarpin promotes senescence in a p53-independent manner. Moreover, cristacarpin treatment induced p38MAPK, indicating the ROS-dependent activation of the MAP kinase pathway, and thus abrogates the tumor growth in mouse allograft tumor model.


Aging/genetics , Cyclin-Dependent Kinase Inhibitor p21/genetics , DNA Damage/drug effects , Endoplasmic Reticulum Stress/drug effects , Oxidative Stress , Pterocarpans/pharmacology , Aging/drug effects , Animals , Cell Cycle/drug effects , Cellular Senescence/drug effects , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Female , Flow Cytometry , Humans , Immunoblotting , Mice , Reactive Oxygen Species/metabolism , Signal Transduction/drug effects
16.
Sci Rep ; 6: 18800, 2016 Jan 05.
Article En | MEDLINE | ID: mdl-26728896

The eukaryotic translation initiation factor 4E (eIF4E) is considered as a key survival protein involved in cell cycle progression, transformation and apoptosis resistance. Herein, we demonstrate that medicinal plant derivative 3-AWA (from Withaferin A) suppressed the proliferation and metastasis of CaP cells through abrogation of eIF4E activation and expression via c-FLIP dependent mechanism. This translational attenuation prevents the de novo synthesis of major players of metastatic cascades viz. c-FLIP, c-Myc and cyclin D1. Moreover, the suppression of c-FLIP due to inhibition of translation initiation complex by 3-AWA enhanced FAS trafficking, BID and caspase 8 cleavage. Further ectopically restored c-Myc and GFP-HRas mediated activation of eIF4E was reduced by 3-AWA in transformed NIH3T3 cells. Detailed underlying mechanisms revealed that 3-AWA inhibited Ras-Mnk and PI3-AKT-mTOR, two major pathways through which eIF4E converges upon eIF4F hub. In addition to in vitro studies, we confirmed that 3-AWA efficiently suppressed tumor growth and metastasis in different mouse models. Given that 3-AWA inhibits c-FLIP through abrogation of translation initiation by co-targeting mTOR and Mnk-eIF4E, it (3-AWA) can be exploited as a lead pharmacophore for promising anti-cancer therapeutic development.


Adenosine Triphosphatases/metabolism , Cation Transport Proteins/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction , TOR Serine-Threonine Kinases/metabolism , ras Proteins/metabolism , Adaptor Proteins, Signal Transducing , Animals , CASP8 and FADD-Like Apoptosis Regulating Protein/genetics , CASP8 and FADD-Like Apoptosis Regulating Protein/metabolism , Carcinoma, Ehrlich Tumor/genetics , Carcinoma, Ehrlich Tumor/metabolism , Carcinoma, Ehrlich Tumor/pathology , Carrier Proteins/metabolism , Caspase 8/metabolism , Cell Cycle Proteins , Cell Line, Tumor , Cell Movement/genetics , Copper-Transporting ATPases , Disease Models, Animal , Eukaryotic Initiation Factor-4E/metabolism , Eukaryotic Initiation Factors , Humans , Male , Mice , Neoplasm Metastasis , Phosphoproteins/metabolism , Phosphorylation , Protein Biosynthesis/drug effects , Protein Transport , Withanolides/pharmacology , fas Receptor/metabolism
17.
Mol Carcinog ; 55(5): 864-81, 2016 May.
Article En | MEDLINE | ID: mdl-25969134

Here, we provide evidences that natural product derivative 3-azido Withaferin A (3-AWA) abrogated EMT and invasion by modulating ß-catenin localization and its transcriptional activity in the prostate as well as in breast cancer cells. This study, for the first time, reveals 3-AWA treatment consistently sequestered nuclear ß-catenin and augmented its cytoplasmic pool as evidenced by reducing ß-catenin transcriptional activity in these cells. Moreover, 3-AWA treatment triggered robust induction of pro-apoptotic intracellular Par-4, attenuated Akt activity and rescued Phospho-GSK3ß (by Akt) to promote ß-catenin destabilization. Further, our in vitro studies demonstrate that 3-AWA treatment amplified E-cadherin expression along with sharp downregulation of c-Myc and cyclin D1 proteins. Strikingly, endogenous Par-4 knock down by siRNA underscored 3-AWA mediated inhibition of nuclear ß-catenin was Par-4 dependent and suppression of Par-4 activity, either by Bcl-2 or by Ras transfection, restored the nuclear ß-catenin level suggesting Par-4 mediated ß-catenin regulation was not promiscuous. In vivo results further demonstrated that 3-AWA was effective inhibitor of tumor growth and immunohistochemical studies indicated that increased expression of total ß-catenin and decreased expression of phospho-ß-catenin and Par-4 in breast cancer tissues as compared to normal breast tissue suggesting Par-4 and ß-catenin proteins are mutually regulated and inversely co-related in normal as well as cancer condition. Thus, strategic regulation of intracellular Par-4 by 3-AWA in diverse cancers could be an effective tool to control cancer cell metastasis. Conclusively, this report puts forward a novel approach of controlling deregulated ß-catenin signaling by 3-AWA induced Par-4 protein.


Antineoplastic Agents, Phytogenic/administration & dosage , Apoptosis Regulatory Proteins/metabolism , Neoplasms/drug therapy , Withanolides/agonists , beta Catenin/metabolism , Animals , Antineoplastic Agents, Phytogenic/pharmacology , Apoptosis , Cell Line, Tumor , Epithelial-Mesenchymal Transition/drug effects , Female , Gene Expression Regulation, Neoplastic/drug effects , Humans , MCF-7 Cells , Mice , Neoplasms/metabolism , Phosphorylation/drug effects , Plants, Medicinal/chemistry , Signal Transduction/drug effects
18.
ACS Med Chem Lett ; 6(10): 1071-4, 2015 Oct 08.
Article En | MEDLINE | ID: mdl-26487914

The present work describes the anti-invasive effect of conjugate BC06, a novel conjugate of EPA, (2E,4E)-4-(benzo[d][1,3]dioxol-5-ylmethylene) hex-2-enoic acid with ß,ß-disubstituted-ß-amino acid, ß(3,3)-Pip-OH (2-(4-aminopiperidin-4-yl)acetic acid), in human pancreatic carcinoma. The conjugate BC06 inhibited invasion and migration of PANC-1 cells in wound healing, matrigel invasion, and gelatin degradation assays. Apart from suppressing PI3K/Akt/NF-kB signaling, which is involved in the up-regulation of matrix metalloproteinases, our study also demonstrated that dose-dependent treatment of BC06 results in the upregulation of TIMP-1 and E-cadherin expression. Further, BC06 was found to be inhibiting the metastatic ability of PANC-1 cells by reducing MMP-2 and MMP-9 expression. These findings suggest that EPA conjugate with ß(3,3)-Pip-OH, BC06, may be used as an anti-invasive agent against human pancreatic carcinoma.

19.
Chem Biol Interact ; 239: 1-11, 2015 Sep 05.
Article En | MEDLINE | ID: mdl-26115782

The chemical investigation of the bioactive nonpolar fractions of Tanacetum gracile afforded two flavonoid analogues namely, 5-hydroxy-3,6,7,3',4'-pentamethoxyflavone (1) and 5,4'-dihydroxy-3,6,7,3',4'-tetramethoxyflavone (2) which were identical to the previously reported artemetin and chrysosplenetin respectively. The structure of the compounds was elucidated on the basis of spectroscopic evidences and they showed significant cytotoxic activity against human breast cancer cells (MCF-7 and T47D). Mechanism based study showed that the compounds modulated microtubule depolymerization by activating mitotic spindle checkpoint. Molecular docking at the colchicine binding pocket revealed that the compounds bind at α-ß interfacial site of tubulin, correlating binding interactions with probable inhibition mechanism. The study reveals important observations to generate improved flavonoids that leads to cell apoptosis. The compounds were also evaluated for absorption, metabolism and toxicity by online webserver admetSAR. The significant microtubule disassembling property and less toxicity paves way for consideration of the compounds as chemopreventive agents.


Antineoplastic Agents, Phytogenic/chemistry , Antineoplastic Agents, Phytogenic/pharmacology , Breast Neoplasms/drug therapy , Flavonoids/pharmacology , Microtubules/drug effects , Tanacetum/chemistry , Animals , Antineoplastic Agents, Phytogenic/toxicity , Apoptosis/drug effects , Binding Sites , Blood-Brain Barrier/drug effects , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Colchicine/chemistry , Colchicine/metabolism , Colchicine/pharmacology , Drug Screening Assays, Antitumor/methods , Female , Flavonoids/chemistry , Humans , MCF-7 Cells/drug effects , Membrane Potential, Mitochondrial/drug effects , Molecular Docking Simulation , Molecular Structure , Rats , Reactive Oxygen Species/metabolism , Tubulin Modulators/chemistry , Tubulin Modulators/pharmacology
20.
J Med Chem ; 58(8): 3432-44, 2015 Apr 23.
Article En | MEDLINE | ID: mdl-25825934

Sclareol, a promising anticancer labdane diterpene, was isolated from Salvia sclarea. Keeping the basic stereochemistry-rich framework of the molecule intact, a method for the synthesis of novel sclareol analogues was designed using palladium(II)-catalyzed oxidative Heck coupling reaction in order to study their structure-activity relationship. Both sclareol and its derivatives showed an interesting cytotoxicity profile, with 15-(4-fluorophenyl)sclareol (SS-12) as the most potent analogue, having IC50 = 0.082 µM against PC-3 cells. It was found that SS-12 commonly interacts with Bcl-2 and Beclin 1 BH3 domain proteins and enhances autophagic flux by modulating autophagy-related proteins. Moreover, inhibition of autophagy by autophagy inhibitors protected against SS-12-induced apoptosis. Finally, SS-12 effectively suppressed tumor growth in vivo in Ehrlich's ascitic and solid Sarcoma-180 mouse models.


Diterpenes/chemistry , Diterpenes/therapeutic use , Sarcoma 180/drug therapy , Animals , Apoptosis/drug effects , Apoptosis Regulatory Proteins/metabolism , Autophagy/drug effects , Beclin-1 , Cell Line, Tumor , Diterpenes/pharmacology , Drug Design , Female , Halogenation , Humans , Membrane Proteins/metabolism , Mice , Proto-Oncogene Proteins c-bcl-2/metabolism , Salvia/chemistry , Sarcoma 180/metabolism , Sarcoma 180/pathology , Signal Transduction/drug effects , TOR Serine-Threonine Kinases/metabolism
...