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










Publication year range
1.
Apoptosis ; 29(1-2): 191-209, 2024 Feb.
Article in English | MEDLINE | ID: mdl-37945815

ABSTRACT

During cancer cell invasion, integrin undergoes constant endo/exocytic trafficking. It has been found that the recycling ability of integrin ß1 through Rab11-controlled long loop pathways is directly associated with cancer invasion. Previous studies showed that gain-of-function mutant p53 regulates the Rab-coupling protein [RCP]-mediated integrin ß1 recycling by inactivating tumor suppressor TAp63. So, we were interested to investigate the involvement of miR-205 in this process. In the current study first, we evaluated that the lower expression of miR-205 in MDA-MB-231 cell line is associated with high motility and invasiveness. Further investigation corroborated that miR-205 directly targets RCP resulting in attenuated RCP-mediated integrin ß1 recycling. Overexpression of TAp63 validates our in vitro findings. To appraise the anti-metastatic role of miR-205, we developed two in vivo experimental models- xenograft-chick embryo and xenograft-immunosuppressed BALB/c mice. Our in vivo results support the negative effect of miR-205 on metastasis. Therefore, these findings advocate the tumor suppressor activity of miR-205 in breast cancer cells and suggest that in the future development of miR-205-targeting RNAi therapeutics could be a smart alternative approach to prevent the metastatic fate of the disease.


Subject(s)
Breast Neoplasms , MicroRNAs , Animals , Chick Embryo , Female , Humans , Mice , Apoptosis , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Cell Line, Tumor , Cell Movement/genetics , Integrin beta1/genetics , Integrin beta1/metabolism , MicroRNAs/genetics , Neoplasm Invasiveness , Neoplasm Metastasis
2.
Fitoterapia ; 169: 105601, 2023 Sep.
Article in English | MEDLINE | ID: mdl-37406886

ABSTRACT

Cancer continues to threat mortal alongside scientific community with burgeoning grasp. Most efforts directed to tame Cancer such as radiotherapy or chemotherapy, all came at a cost of severe side effects. The plant derived bioactive compounds on the other hand carries an inevitable advantage of being safer, bioavailable & less toxic compared to contemporary chemotherapeutics. Our strategic approach employed solvent extraction of Black Seed Oil (BSO) to highlight the orchestrated use of its oil soluble phytochemicals - Thymoquinone, Carvacrol & Trans-Anethole when used in cohort. These anti-cancer agents in unbelievably modest amounts present in BSO shows better potential to delineate migratory properties in breast cancer cells as compared to when treated individually. BSO was also observed to have apoptotic calibre when investigated in MDA-MB-231 and MCF-7 cell lines. We performed chemical characterization of the individual phytochemical as well as the oil in-whole to demonstrate the bioactive oil-soluble entities present in whole extract. BSO was observed to have significant anti-cancerous properties in cumulative proportion that is reportedly higher than the individual three components. Besides, this study also reports micro-RNA regulation on BSO administration, thereby playing a pivotal role in breast cancer alleviation. Thus, synergistic action of the integrants serves better combat force against breast cancer in the form of whole extract, hence aiming at a more lucrative paradigm while significantly regulating microRNAs associated with breast cancer migration and apoptosis.


Subject(s)
Breast Neoplasms , MicroRNAs , Nigella sativa , Humans , Female , Breast Neoplasms/drug therapy , Nigella sativa/chemistry , Molecular Structure , Plant Extracts/pharmacology , Plant Extracts/therapeutic use
3.
Apoptosis ; 28(3-4): 263-276, 2023 04.
Article in English | MEDLINE | ID: mdl-36536258

ABSTRACT

Tumor cells always have the need to produce an increased amount of proteins in the cells. This elevated amount of proteins increases the pressure on the organelles of the cell such as the endoplasmic reticulum and compels it to increase its protein folding efficiency. However, it is by a matter of fact, that the amount of proteins synthesized outweighs the protein folding capacity of the ER which in turn switches on the UPR pathway by activating the three major molecular sensors and other signaling cascades, which helps in cell survival instead of instant death. However, if this pathway is active for a prolonged period of time the tumor cells heads toward apoptosis. Again, interestingly this is not the same as in case of non- tumorogenic cells. This exhibit a straight natural pathway for tumor cells-specific destruction which has a great implication in today's world where hormone therapies and chemo-therapies are non-effective for various types of breast cancer, a major type being Triple Negative Breast Cancer. Thus a detailed elucidation of the molecular involvement of the UPR pathway in breast cancer may open new avenues for management and attract novel chemotherapeutic targets providing better hopes to patients worldwide.


Subject(s)
Breast Neoplasms , Humans , Female , Breast Neoplasms/drug therapy , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Endoplasmic Reticulum Stress , Apoptosis/genetics , Unfolded Protein Response , Signal Transduction
4.
Biomater Sci ; 9(24): 8285-8312, 2021 Dec 07.
Article in English | MEDLINE | ID: mdl-34766965

ABSTRACT

Epidermal growth factor receptor (EGFR) normally over-expresses in non-small cell lung cancer (NSCLC) cells. Its mutations act as oncogenic drivers in the cellular signal transduction pathway, and induce the downstream activation of numerous key cellular events involved in cellular proliferation and survival. EGFR tyrosine kinase inhibitors (EGFR-TK inhibitors), such as gefitinib and erlotinib, have been used for a long time in the treatment of NSCLC. However, they fail to overcome the EGFR-TK mutation due to the acquisition of drug resistance. It is strongly believed that the epithelial-to-mesenchymal transition (EMT) is a key player for acquired resistance and consequent limitation of the clinical efficiency of EGFR-TKIs. Therefore, a new strategy needs to be developed to overcome the resistance in NSCLC. In this current study, we have disclosed for the first time the efficiency of transferrin-modified PLGA-thymoquinone-nanoparticles in combination with gefitinib (NP-dual-1, NP-dual-2 and NP-dual-3) towards gefitinib-resistant A549 cells. The gefitinib-resistant A549 cells (A549/GR) showed 12.3-fold more resistance to gefitinib in comparison to non-resistant A549 cells. The phenotypic alteration resembling spindle-cell shape and increased pseudopodia integuments featured the EMT phenomena in A549/GR cells. EMT in A549/GR was later coupled with the loss of Ecad and expansion of Ncad, along with upregulated vimentin expression, as compared to the control A549 cells. Moreover, the invasive nature and migration potential are more amplified in A549/GR cells. Pre-incubation of A549 cells with TGFß1 also initiated EMT, leading to drug resistance. Conversely, treatment of A549 or A549/GR cells with NP-dual-3 effectively retrieved the sensitivity to gefitinib, restricted the EMT phenomenon, and impaired the TGFß1-induced EMT. On unveiling the underlying mechanism of therapeutic action, we found that STAT3 and miR-21 were individually overexpressed in the A549/GR cells by transfection, and followed by treatment with NP-dual-3. Simultaneously, NP-dual-3 fragmented HIF1-α induced EMT in A549/GR cells and reduced the CSCs markers, viz., Oct-4, Sox-2, Nanog, and Aldh1. These data are self-sufficient to suggest that NP-dual-3 re-sensitizes the drug-resistant A549/GR cells to gefitinib, possibly by retrieving MET phenomena via modulation of STAT3/mir-21/Akt/PTEN/HIF1-α axis. Thus, TQ nanoparticles combined with TKI gefitinib may provide an effective platform to treat NSCLC.


Subject(s)
Antineoplastic Agents , Carcinoma, Non-Small-Cell Lung , Lung Neoplasms , Nanoparticles , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Benzoquinones , Carcinoma, Non-Small-Cell Lung/drug therapy , Cell Line, Tumor , Cell Proliferation , Drug Resistance, Neoplasm , Gefitinib/pharmacology , Gefitinib/therapeutic use , Humans , Lung Neoplasms/drug therapy , Transferrin
5.
Biomater Sci ; 9(16): 5665-5690, 2021 Aug 21.
Article in English | MEDLINE | ID: mdl-34259681

ABSTRACT

To date, most of the accessible therapeutic options are virtually non-responsive towards triple-negative breast cancer (TNBC) due to its highly aggressive and metastatic nature. Interestingly, chemotherapy reacts soundly in many TNBC cases compared to other types of breast cancer. However, the side effects of many chemotherapeutic agents are still under cross-examination, and thus prohibit their extensive uses. In this present study, we have developed a series of coumarin-dihydropyrimidinone conjugates (CDHPs) and subsequently their poly(lactic-co-glycolic acid) (PLGA)-PEG4000 mixed copolymer nanoparticles as excellent chemotherapeutic nanomedicine to control TNBC. Among all the synthesized CDHPs, CDHP-4 (prepared by the combination of EDCO with 3,4-difluorobenzaldehyde) showed excellent therapeutic effect on a wide variety of cancer cell lines, including TNBC. Besides, it can control the metastasis and stemness property of TNBC. Furthermore, the nano-encapsulation of CDHP-4 in a mixed polymer nanoparticle system (CDHP-4@PP-NPs) and simultaneous delivery showed much improved therapeutic efficacy at a much lower dose, and almost negligible side effects in normal healthy cells or organs. The effectiveness of the present therapeutic agent was observed both in intravenous and oral mode of administration in in vivo experiments. Moreover, on elucidating the molecular mechanism, we found that CDHP-4@PP-NPs could exhibit apoptotic, anti-migratory, as well as anti-stemness activity against TNBC cell lines through the downregulation of miR-138. We validated our findings in MDA-MB-231 xenograft chick embryos, as well as in 4T1-induced mammary tumor-bearing BALB/c mice models, and studied the bio-distribution of CDHP-4@PP-NPs on the basis of the photoluminescence property of nanoparticles. Our recent study, hence for the first time, unravels the synthesis of CDHP-4@PP-NPs and the molecular mechanism behind the anti-migration, anti-stemness and anti-tumor efficacy of the nanoparticles against the TNBC cells through the miR-138/p65/TUSC2 axis.


Subject(s)
Coumarins , Nanoparticles , Triple Negative Breast Neoplasms , Animals , Cell Line, Tumor , Chick Embryo , Humans , Mice , Mice, Inbred BALB C , MicroRNAs , Triple Negative Breast Neoplasms/drug therapy , Tumor Suppressor Proteins
6.
Biochim Biophys Acta Gen Subj ; 1864(11): 129695, 2020 11.
Article in English | MEDLINE | ID: mdl-32735937

ABSTRACT

BACKGROUND: Breast cancer intimidates the contemporary medical advances, attempting to revolutionize cancer therapeutics. While patients suffering an advanced breast cancer are dependent on mono drugs, yet the build out of resistance leading to treatment fails has become inevitable. METHODS: Cell viability Assay with MTT revealed the "IC50" concentrations of the drugs in both cancer as well as PBMC. Cell cycle arrest, flow cytometric ROS analysis & apoptosis evaluation pointed out the efficacy of the dual drug. Wound Healing, Transwell Migration & Immunocytochemistry indicated anti-migratory potential of TQ-Emo while expression patterns of Cl-Cas3, p53, Bax, Bcl2 & the stemness markers further vouched the potential of the combinatorial drug. Furthermore, validation of tumor inhibitory effect was earned by an ex-ovo xenograft model. RESULTS: Dual dosage enhanced apoptosis through ROS generation, anti- migratory effect by targeting FAK &Integrins, displaying effective stemness control by assessing regulatory proteins- Oct4, Sox2, Nanog, ALDH1/2. Ex-ovo xenograft model validated tumor regression. Our study thereby deals with devastating effects of cancer drug resistance while trying to abate enhanced migratory potential & stemness, utilizing the synergism of the combinable therapy. CONCLUSION: TQ/Emo inhibited breast cancer proliferation synergistically while enhancing cytotoxicity, inducing apoptosis on MCF-7 cells while curbing migration & stemness. GENERAL SIGNIFICANCE: Employment of the combinatorial phytochemicals, Thymoquinone & Emodin attempted to achieve deliverables like reduced cellular toxicity, drug resistance, anti-migratory potency & stemness. Besides, decreased p-FAK expression or regression in Mammosphere & tumor size in ex-ovo xenograft model is indicative of the better anti-tumorigenic potential of the dual formulation.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Benzoquinones/pharmacology , Breast Neoplasms/drug therapy , Emodin/pharmacology , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Breast Neoplasms/pathology , Cell Movement/drug effects , Cell Proliferation/drug effects , Drug Synergism , Female , Humans , MCF-7 Cells , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/pathology , Protein Kinase Inhibitors/pharmacology
7.
Biomater Sci ; 8(10): 2939-2954, 2020 May 21.
Article in English | MEDLINE | ID: mdl-32319481

ABSTRACT

The development of new therapeutic strategies to target triple-negative breast cancer (TNBC) is in much demand to overcome the roadblocks associated with the existing treatment procedures. In this regard, therapies targeting the CD44 receptor have drawn attention for more than a decade. MicroRNAs (miRNAs) modulate post-transcriptional gene regulation and thus, the correction of specific miRNA alterations using miRNA mimics or antagomiRs is an emerging strategy to normalize the genetic regulation in the tumor microenvironment. It has been acknowledged that miR-34a is downregulated and miR-10b is upregulated in TNBC, which promotes tumorigenesis and metastatic dissemination. However, there are a few barriers related to miRNA delivery. Herein, we have introduced tailored mesoporous silica nanoparticles (MSNs) for the co-delivery of miR-34a-mimic and antisense-miR-10b. MSN was functionalized with a cationic basic side chain and then loaded with the dual combination to overexpress miR-34a and downregulate miR-10b simultaneously. Finally, the loaded MSNs were coated with an hyaluronic acid-appended PEG-PLGA polymer for specific targeting. The cellular uptake, release profile, and subsequent effect in TNBC cells were evaluated. In vitro and in vivo studies demonstrated high specificity in TNBC tumor targeting, leading to efficient tumor growth inhibition as well as the retardation of metastasis, which affirmed the clinical application potential of the system.


Subject(s)
Gene Transfer Techniques , MicroRNAs/genetics , Triple Negative Breast Neoplasms/therapy , Animals , Female , Humans , Hyaluronan Receptors/genetics , Hyaluronan Receptors/metabolism , Mice , Mice, Inbred BALB C , MicroRNAs/metabolism , Nanoparticles/chemistry , Particle Size , Polylactic Acid-Polyglycolic Acid Copolymer/chemistry , Porosity , Silicon Dioxide/chemistry , Surface Properties , Triple Negative Breast Neoplasms/genetics , Triple Negative Breast Neoplasms/metabolism , Tumor Cells, Cultured
8.
J Control Release ; 322: 357-374, 2020 06 10.
Article in English | MEDLINE | ID: mdl-32243981

ABSTRACT

Triple-negative breast cancer (TNBC) is a highly aggressive and metastatic subtype of breast cancer showing non-responsiveness to most available therapeutic options. Therefore, smart therapeutic approaches to selectively transport and target TNBCs are required. Herein, we developed thymoquinone (TQ)-loaded, hyaluronic acid (HA)-conjugated Pluronic® P123 and F127 copolymer nanoparticles (HA-TQ-Nps) as a selective drug-carrying vehicle to deliver anticancer phytochemical TQ to TNBC cells. The mean size of nanoparticles was around 19.3 ± 3.2 nm. and they were stable at room temperature up to 4 months. HA-TQ-Nps were immensely cytotoxic towards TNBC cells but did not show the toxic effect on normal cells. Detailed investigations also demonstrated its pro-apoptotic, anti-metastatic and anti-angiogenic activity. In-depth mechanistic studies highlighted that HA-TQ-Nps retarded cell migration of TNBC cells through up-regulation of microRNA-361 which in turn down-regulated Rac1 and RhoA mediated cell migration and also perturbed the cancer cell migration under the influence of the autocrine effect of VEGF-A. Moreover, HA-TQ-Np-treatment also perturbed tumor-induced vascularization by reducing the secretion of VEGF-A. The anti-metastatic and anti-angiogenic activity of HA-TQ-Nps was found to be evident in both MDA-MB-231 xenograft chick embryos and 4T1-mammary solid tumor model in syngeneic mice. Thus, an innovative targeted nano-therapeutic approach is being established to reduce the tumor burden and inhibit metastasis and angiogenesis simultaneously for better management of TNBC.


Subject(s)
MicroRNAs , Nanoparticles , Triple Negative Breast Neoplasms , Animals , Benzoquinones , Cell Line, Tumor , Chick Embryo , Humans , Hyaluronic Acid , Mice , Poloxamer , Triple Negative Breast Neoplasms/drug therapy
9.
Biomater Sci ; 7(10): 4325-4344, 2019 Oct 01.
Article in English | MEDLINE | ID: mdl-31411213

ABSTRACT

Non-small cell lung carcinoma (NSCLC) is a highly lethal type of cancer with limited therapeutic avenues available to date. In the present study, we formulated PEGylated PLGA thymoquinone nanoparticles (TQ-Np) for improved TQ delivery to NSCLC cells. Transferrin (TF), a biodegradable, non-immunogenic and non-toxic protein, is well known to bind to TFR (transferrin receptor) over-expressed in non-small cell lung carcinoma A549 cells. Thus, the further decoration of the PEGylated PLGA thymoquinone nanoparticles with transferrin (TF-TQ-Np) enhanced the internalization of the nanoparticles within the A549 cells and the activity of TQ. We established TF-TQ-Np as a potent anti-tumorigenic agent through the involvement of p53 and the ROS feedback loop in regulating the microRNA (miRNA) circuitry to control apoptosis and migration of NSCLC cells. TF-TQ-Np-mediated p53 up-regulation favored the potential simultaneous activation of miR-34a and miR-16 targeting Bcl2 to induce apoptosis in the A549 cells. Additionally, TF-TQ-Np also restricted the migration through actin de-polymerization via activation of the p53/miR-34a axis. Further studies in chick CAM xenograft models confirmed the anti-cancer activity of TF-TQ-Np by controlling the p53/miR-34a/miR-16 axis. Furthermore, in vivo experiments conducted in a xenograft model in immunosuppressed Balb/c mice also proved the efficacy of the nanoparticles as an antitumor agent against NSCLC. Thus, our findings cumulatively suggest that the transferrin-adorned TQ-Np successfully coupled two distinct miRNA pathways to potentiate the apoptotic death cascade in the very lethal NSCLC cells and also restricts the migration of these cells without imparting any significant toxicity, which occurs in the widely used chemotherapeutic combinations. Thereby, our findings rekindle new hopes for the development of improved targeted therapeutic options with specified molecular objectives for combating the deadly NSCLC.


Subject(s)
Antineoplastic Agents/administration & dosage , Benzoquinones/administration & dosage , Carcinoma, Non-Small-Cell Lung/drug therapy , Lung Neoplasms/drug therapy , MicroRNAs , Nanoparticles/administration & dosage , Polyesters/administration & dosage , Polyethylene Glycols/administration & dosage , Transferrin/administration & dosage , Animals , Apoptosis/drug effects , Carcinoma, Non-Small-Cell Lung/genetics , Cell Line, Tumor , Cell Movement/drug effects , Chick Embryo , Female , Humans , Lung Neoplasms/genetics , Mice, Inbred BALB C , Reactive Oxygen Species/metabolism , Wound Healing/drug effects
10.
Apoptosis ; 21(10): 1106-24, 2016 10.
Article in English | MEDLINE | ID: mdl-27392939

ABSTRACT

The efficacy of cancer chemotherapeutics is limited by side effects resulting from narrow therapeutic windows between the anticancer activity of a drug and its cytotoxicity. Thus identification of small molecules that can selectively target cancer cells has gained major interest. Cancer cells under stress utilize the Unfolded protein response (UPR) as an effective cell adaptation mechanism. The purpose of the UPR is to balance the ER folding environment and calcium homeostasis under stress. If ER stress is prolonged, tumor cells undergo apoptosis. In the present study we demonstrated an 3,3'-(Arylmethylene)-bis-1H-indole (AMBI) derivative 3,3'-[(4-Methoxyphenyl) methylene]-bis-(5-bromo-1H-indole), named as Mephebrindole (MPB) as an effective anti-cancer agent in breast cancer cells. MPB disrupted calcium homeostasis in MCF7 cells which triggered ER stress development. Detailed evaluations revealed that mephebrindole by activating p38MAPK also regulated GRP78 and eIF2α/ATF4 downstream to promote apoptosis. Studies extended to in vivo allograft mice models revalidated its anti-carcinogenic property thus highlighting the role of MPB as an improved chemotherapeutic option.


Subject(s)
Activating Transcription Factor 4/metabolism , Apoptosis/drug effects , Breast Neoplasms/drug therapy , Carcinoma/drug therapy , Eukaryotic Initiation Factor-2/metabolism , Indoles/administration & dosage , Transcription Factor CHOP/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism , Activating Transcription Factor 4/genetics , Animals , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Breast Neoplasms/physiopathology , Carcinoma/genetics , Carcinoma/metabolism , Carcinoma/physiopathology , Cell Line, Tumor , Endoplasmic Reticulum Chaperone BiP , Eukaryotic Initiation Factor-2/genetics , Female , Humans , Indoles/chemical synthesis , Indoles/chemistry , Mice , Receptor Cross-Talk/drug effects , Signal Transduction/drug effects , Transcription Factor CHOP/genetics , p38 Mitogen-Activated Protein Kinases/genetics
11.
Front Pharmacol ; 7: 114, 2016.
Article in English | MEDLINE | ID: mdl-27199756

ABSTRACT

Triple-negative breast cancer (TNBC), is a specific subtype of epithelial breast tumors that are immuno-histochemically negative for the protein expression of the estrogen receptor (ER), the progesterone receptor (PR) and lack over expression/gene amplification of HER2. This subtype of breast cancers is highly metastatic, shows poor prognosis and hence represents an important clinical challenge to researchers worldwide. Thus alternative approaches of drug development for TNBC have gained utmost importance in the present times. Dietary indole and its derivatives have gained prominence as anti-cancer agents and new therapeutic approaches are being developed to target them against TNBC. But a major drawback with 3, 3'di Indolyl methane (DIM) is their poor bioavailability and high effective concentration against TNBC. However, the Aryl methyl ring substituted analogs of DIM display interesting anti-cancer activity in breast cancer cells. In the current study we report the synthesis of a novel synthetic aryl methyl ring substituted analog of DIM, named as Phemindole as an effective anti-tumor agent against TNBC cells. Furthermore, we enumerated that Phemindole caused reactive oxygen species mediated mitochondrial-dependent apoptosis in MDAMB-231 cells. Furthermore, Phemindole mediated Store Operated Calcium Entry (SOCE) retardation favored inactivation of STIM1 and henceforth activated ER stress to induce apoptosis in TNBC cells. Simultaneously, Phemindole was also found to restrict the in vitro cell migration through its anti mitotic property and pFAK regulation. Studies extended to ex ovo and in vivo mice models further validated the efficacy of Phemindole. Thus our results cumulatively propose Phemindole as a new chemotherapeutic regime which might be effective to target the deadly aspects of the TNBC.

12.
Biomaterials ; 76: 115-32, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26520043

ABSTRACT

Metal oxide nanoparticles are the forthcoming anti-tumor therapeutics and provide a versatile platform in the development of therapeutic approaches for drug-resistant cancers such as triple negative breast cancer (TNBC). Copper oxide nanoparticles have been characterized as anti-cancer agents but its toxicity has been a matter of concern. Herein, we have developed a targeted CuO Nanowire fabricated with Folic acid (CuO-Nw-FA) that enables enhanced cellular uptake in TNBC cells without imparting significant toxicity in normal cellular system. In the present study, we enumerated that CuO-Nw-FA caused mitochondrial-dependent apoptosis in MDAMB-231 cells. Furthermore, CuO-Nw-FA mediated cytosolic retardation of NF-κB favoured inactivation of miR-425 and henceforth activated PTEN to induce apoptosis in TNBC cells. Simultaneously, CuO-Nw-FA also restricted the in-vitro cell migration through the miR-425/PTEN axis via pFAK. Studies extended to ex-ovo and in-vivo mice models further validated the efficacy of CuO-Nw-FA. Additionally, the accumulations of nanoparticles in tumor as well as different organs in mice were examined by in-vivo biodistribution and ex-vivo optical imaging studies. Thus our results cumulatively propose that CuO-Nw-FA cross-talks two distinct signalling pathways to induce apoptosis and retard migration in TNBC cells and raises the possibility for the use of CuO-Nw-FA as a potent anti-tumor agent.


Subject(s)
Copper/chemistry , Folic Acid/chemistry , MicroRNAs/metabolism , Mitochondria/metabolism , Nanowires , PTEN Phosphohydrolase/metabolism , Reactive Oxygen Species/metabolism , Triple Negative Breast Neoplasms/drug therapy , Animals , Antineoplastic Agents/therapeutic use , Apoptosis , Cell Line, Tumor , Chick Embryo , Female , Humans , Mice , Mice, Inbred BALB C , Triple Negative Breast Neoplasms/metabolism , Triple Negative Breast Neoplasms/pathology
13.
Biomaterials ; 51: 91-107, 2015 May.
Article in English | MEDLINE | ID: mdl-25771001

ABSTRACT

Thymoquinone (TQ), a major active constituent of black seeds of Nigella sativa, has potential medical applications including spectrum of therapeutic properties against different cancers. However, little is known about their effect on breast cancer cell migration, which is the cause of over 90% of deaths worldwide. Herein, we have synthesized TQ-encapsulated nanoparticles using biodegradable, hydrophilic polymers like polyvinylpyrrolidone (PVP) and polyethyleneglycol (PEG) to overcome TQ's poor aqueous solubility, thermal and light sensitivity as well as consequently, minimal systemic bioavailability which can greatly improve the cancer treatment efficiency. Sizes of synthesized TQ-Nps were found to be below 50 nm and they were mostly spherical in shape with smooth surface texture. Estimation of the zeta potential also revealed that all the three TQ-Nps were negatively charged which also facilitated their cellular uptake. In the present investigation, we provide direct evidence that TQ-Nps showed more efficiency in killing cancer cells as well as proved to be less toxic to normal cells at a significantly lower dose than TQ. Interestingly, evaluation of the anti-migratory effect of the TQ-Nps, revealed that PEG4000-TQ-Nps showed much potent anti-migratory properties than the other types. Further studies indicated that PEG4000-TQ-Nps could significantly increase the expression of miR-34a through p53. Moreover, NPs mediated miR-34a up-regulation directly down-regulated Rac1 expression followed by actin depolymerisation thereby disrupting the actin cytoskeleton which leads to significant reduction in the lamellipodia and filopodia formation on cell surfaces thus retarding cell migration. Considering the biodegradability, non-toxicity and effectivity of PEG4000-TQ-Nps against cancer cell migration, TQ-Nps may provide new insights into specific therapeutic approach for cancer treatment.


Subject(s)
Actin Cytoskeleton/metabolism , Benzoquinones/pharmacology , Breast Neoplasms/pathology , Cell Movement/drug effects , MicroRNAs/metabolism , Nanoparticles/chemistry , Polyethylene Glycols/chemistry , Actin Cytoskeleton/drug effects , Animals , Breast Neoplasms/genetics , Cell Death/drug effects , Down-Regulation/drug effects , Drug Liberation , Endocytosis/drug effects , Female , Humans , MCF-7 Cells , Mice , Nanoparticles/ultrastructure , Particle Size , Polymerization , Pseudopodia/drug effects , Pseudopodia/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Spectroscopy, Fourier Transform Infrared , Tumor Suppressor Protein p53/metabolism , rac1 GTP-Binding Protein/genetics , rac1 GTP-Binding Protein/metabolism
14.
J Biol Chem ; 290(7): 3936-49, 2015 Feb 13.
Article in English | MEDLINE | ID: mdl-25527500

ABSTRACT

Triple negative breast cancers (TNBC) are among the most aggressive and therapy-resistant breast tumors and currently possess almost no molecular targets for therapeutic options in this horizon. In the present study we discerned the molecular mechanisms of potential interaction between the endoplasmic reticulum (ER) stress response and the MEK/ERK pathway in inducing apoptosis in TNBC cells. Here we observed that induction of ER stress alone was not sufficient to trigger significant apoptosis but simultaneous inhibition of the MEK/ERK pathway enhanced ER stress-induced apoptosis via a caspase-dependent mechanism. Our study also demonstrated nifetepimine, a dihydropyrimidone derivative as a potent anti-cancer agent in TNBC cells. Nifetepimine down-regulated the MEK/ERK pathway in MDAMB-231 and MDAMB-468 cells and resulted in blockage of ER stress-mediated GRP78 up-regulation. Detailed mechanistic studies also revealed that nifetepimine by down-regulating pERK expression also declined the promoter binding activity of TFII-I to the GRP78 promoter and in turn regulated GRP78 transcription. Studies further extended to in vivo Swiss albino and SCID mice models also revalidated the anti-carcinogenic property of nifetepimine. Thus our findings cumulatively suggest that nifetepimine couples two distinct signaling pathways to induce the apoptotic death cascade in TNBC cells and raises the possibility for the use of nifetepimine as a potent anti-cancer agent with strong immune-restoring properties for therapeutic intervention for this group of cancer bearers.


Subject(s)
Apoptosis/drug effects , Endoplasmic Reticulum Stress/drug effects , MAP Kinase Kinase Kinases/metabolism , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Pyrimidinones/pharmacology , Triple Negative Breast Neoplasms/drug therapy , Triple Negative Breast Neoplasms/pathology , Animals , Blotting, Western , Cell Cycle/drug effects , Cell Proliferation/drug effects , Endoplasmic Reticulum Chaperone BiP , Female , Heat-Shock Proteins/antagonists & inhibitors , Heat-Shock Proteins/genetics , Heat-Shock Proteins/metabolism , Humans , Immunoenzyme Techniques , MAP Kinase Kinase Kinases/genetics , Male , Mice , Mice, Nude , Mice, SCID , Mitogen-Activated Protein Kinase 1/genetics , Mitogen-Activated Protein Kinase 3/genetics , RNA, Messenger/genetics , RNA, Small Interfering/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Triple Negative Breast Neoplasms/metabolism , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
15.
J Biol Chem ; 289(37): 25431-44, 2014 Sep 12.
Article in English | MEDLINE | ID: mdl-25086032

ABSTRACT

The evolution of the cancer cell into a metastatic entity is the major cause of death in patients with cancer. It has been acknowledged that aberrant activation of a latent embryonic program, known as the epithelial-mesenchymal transition (EMT), can endow cancer cells with the migratory and invasive capabilities associated with metastatic competence for which E-cadherin switch is a well-established hallmark. Discerning the molecular mechanisms that regulate E-cadherin expression is therefore critical for understanding tumor invasiveness and metastasis. Here we report that SMAR1 overexpression inhibits EMT and decelerates the migratory potential of breast cancer cells by up-regulating E-cadherin in a bidirectional manner. While SMAR1-dependent transcriptional repression of Slug by direct recruitment of SMAR1/HDAC1 complex to the matrix attachment region site present in the Slug promoter restores E-cadherin expression, SMAR1 also hinders E-cadherin-MDM2 interaction thereby reducing ubiquitination and degradation of E-cadherin protein. Consistently, siRNA knockdown of SMAR1 expression in these breast cancer cells results in a coordinative action of Slug-mediated repression of E-cadherin transcription, as well as degradation of E-cadherin protein through MDM2, up-regulating breast cancer cell migration. These results indicate a crucial role for SMAR1 in restraining breast cancer cell migration and suggest the candidature of this scaffold matrix-associated region-binding protein as a tumor suppressor.


Subject(s)
Breast Neoplasms/genetics , Cadherins/biosynthesis , Cell Cycle Proteins/biosynthesis , DNA-Binding Proteins/biosynthesis , Epithelial-Mesenchymal Transition/genetics , Nuclear Proteins/biosynthesis , Breast Neoplasms/pathology , Cadherins/genetics , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Cell Line, Tumor , Cell Movement/genetics , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Female , Gene Expression Regulation, Neoplastic , Histone Deacetylase 1/genetics , Histone Deacetylase 1/metabolism , Humans , Neoplasm Metastasis , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Proteolysis , Proto-Oncogene Proteins c-mdm2/genetics , Proto-Oncogene Proteins c-mdm2/metabolism , Snail Family Transcription Factors , Transcription Factors/genetics
16.
J Biol Chem ; 287(39): 32881-96, 2012 Sep 21.
Article in English | MEDLINE | ID: mdl-22851172

ABSTRACT

Multiple mechanisms have been proposed by which tumors induce T cell apoptosis to circumvent tumor immune-surveillance. Although sarco/endoplasmic reticulum Ca(2+)-ATPase (SERCA) have long been known to regulate intracellular Ca(2+) homeostasis, few studies have examined the role of SERCA in processes of T lymphocyte survival and activation. In this context it remains largely unexplored as to how tumors jeopardize SERCA function to disable T cell-mediated anti-tumor immunity. Here, we show that human CD4(+) T cells in the presence of tumor conditions manifested an up-regulation of SERCA3 expression that resulted in development of endoplasmic reticulum stress leading to CD4(+) T cell apoptosis. Prostaglandin E(2) produced by the tumor cell plays a critical role in up-regulating SERCA3 by enhancing the binding of its transcription factor Sp1. Gene manipulation and pharmacological approaches further established that an increase in SERCA expression also resulted in subsequent inhibition of PKCα and -θ and retention of NFκB in the cytosol; however, down-modulation of SERCA3 expression by a dihydropyrimidone derivative, ethyl-4-(3-nitro)-phenyl-6-methyl-2-oxo-1,2,3,4-tetrahydropyrimidine-5 carboxylate (nifetepimine), protected the CD4(+) T cells from tumor-induced apoptosis. In fact, nifetepimine-mediated restoration of PKC activity resulted in nuclear translocation of p65NFκB, thereby ensuring its survival. Studies further undertaken in a tumor-bearing mice model revalidated the immunoprotective role of nifetepimine. Our present study thus strongly suggests that imbalance in cellular calcium homeostasis is an important factor leading to CD4(+) T cell death during cancer and holds promise that nifetepimine may have the potential to be used as an immunorestoring agent in cancer bearers.


Subject(s)
Breast Neoplasms/enzymology , CD4-Positive T-Lymphocytes/metabolism , Calcium/metabolism , Immunologic Factors/pharmacology , Neoplasm Proteins/metabolism , Pyrimidinones/pharmacology , Sarcoplasmic Reticulum Calcium-Transporting ATPases/biosynthesis , Tumor Microenvironment/drug effects , Animals , Breast Neoplasms/genetics , Breast Neoplasms/immunology , CD4-Positive T-Lymphocytes/immunology , Cell Survival/drug effects , Cell Survival/genetics , Cell Survival/immunology , Dinoprostone/genetics , Dinoprostone/immunology , Dinoprostone/metabolism , Endoplasmic Reticulum Stress/drug effects , Endoplasmic Reticulum Stress/genetics , Endoplasmic Reticulum Stress/immunology , Female , Gene Expression Regulation, Enzymologic/drug effects , Gene Expression Regulation, Enzymologic/genetics , Gene Expression Regulation, Enzymologic/immunology , Gene Expression Regulation, Neoplastic/drug effects , Gene Expression Regulation, Neoplastic/genetics , Gene Expression Regulation, Neoplastic/immunology , Humans , Lymphocyte Activation/drug effects , Lymphocyte Activation/genetics , Lymphocyte Activation/immunology , Male , Mice , Neoplasm Proteins/genetics , Neoplasm Proteins/immunology , Neoplasm Transplantation , Protein Kinase C-alpha/genetics , Protein Kinase C-alpha/immunology , Protein Kinase C-alpha/metabolism , Sarcoplasmic Reticulum Calcium-Transporting ATPases/genetics , Sarcoplasmic Reticulum Calcium-Transporting ATPases/immunology , Sp1 Transcription Factor/genetics , Sp1 Transcription Factor/immunology , Sp1 Transcription Factor/metabolism , Transplantation, Heterologous , Tumor Microenvironment/genetics , Up-Regulation/drug effects , Up-Regulation/genetics , Up-Regulation/immunology
17.
Eur J Med Chem ; 54: 223-31, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22658336

ABSTRACT

A regioselective N1-alkylation of 3,4-dihydropyrimidin-2(1H)-ones using a very efficient mild base Cs(2)CO(3) and alkyl halides at room temperature has been reported. The selectivity of this methodology is excellent and the yields of the alkylated products are very good. Furthermore inhibitory action of both the 3,4-dihydropyrimidin-2(1H)-ones and the N1-alkylated derivatives were tested on Ca(2+)-ATPase, which revealed that the parent compounds can act as Ca(2+)-ATPase inhibitors whereas the N1-alkylated derivatives are inefficient for this purpose.


Subject(s)
Calcium-Transporting ATPases/antagonists & inhibitors , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/pharmacology , Pyrimidinones/chemical synthesis , Pyrimidinones/pharmacology , Alkylation , Animals , Calcium-Transporting ATPases/chemistry , Drug Evaluation, Preclinical , Enzyme Inhibitors/chemistry , Models, Molecular , Molecular Conformation , Pyrimidinones/chemistry , Stereoisomerism , Substrate Specificity
18.
PLoS One ; 7(1): e30552, 2012.
Article in English | MEDLINE | ID: mdl-22291985

ABSTRACT

Mammalian sperm capacitation is an essential prerequisite to fertilization. Although progress is being made in understanding the physiology and biochemistry of capacitation, little has been yet explored about the potential role(s) of individual sperm cell protein during this process. Therefore elucidation of the role of different sperm proteins in the process of capacitation might be of great importance to understand the process of fertilization. The present work describes the partial characterization of a 14-kDa protein (p14) detected in goat spermatozoa using an antibody directed against the purified protein. Confocal microscopic analysis reveals that the protein is present in both the intracellular and extracellular regions of the acrosomal and postacrosomal portion of caudal sperm head. Though subcellular localization shows that p14 is mainly cytosolic, however it is also seen to be present in peripheral plasma membrane and soluble part of acrosome. Immuno-localization experiment shows change in the distribution pattern of this protein upon induction of capacitation in sperm cells. Increased immunolabeling in the anterior head region of live spermatozoa is also observed when these cells are incubated under capacitating conditions, whereas most sperm cells challenged with the calcium ionophore A23187 to acrosome react, lose their labeling almost completely. Intracellular distribution of p14 also changes significantly during acrosome reaction. Interestingly, on the other hand the antibody raised against this 14-kDa sperm protein enhances the forward motility of caprine sperm cells. Rose-Bengal staining method shows that this anti-p14 antibody also decreases the number of acrosome reacted cells if incubated with capacitated sperm cells before induction of acrosome reaction. All these results taken together clearly indicate that p14 is intimately involved and plays a critical role in the acrosomal membrane fusion event.


Subject(s)
Acrosome Reaction , Goats/physiology , Seminal Plasma Proteins/physiology , Sperm Capacitation , Spermatozoa/metabolism , Spermatozoa/physiology , Acrosome Reaction/physiology , Animals , Epididymis/metabolism , Epithelial Cells/metabolism , Flow Cytometry , Goats/metabolism , Male , Microscopy, Fluorescence , Semen Analysis , Seminal Plasma Proteins/metabolism , Sperm Capacitation/physiology , Spermatogenesis/physiology , Tissue Distribution
19.
Eur J Med Chem ; 45(12): 6012-9, 2010 Dec.
Article in English | MEDLINE | ID: mdl-20952103

ABSTRACT

Synthesis of a series of novel (6-deoxy-glycopyranosid-6-yl) sulfone derivatives has been achieved using a general synthetic strategy. Yields were excellent in every case. The synthetic compounds were evaluated for their biological potential against Ca2+-ATPase, an important enzyme involves in transporting Ca2+ across the cell membranes.


Subject(s)
Calcium-Transporting ATPases/antagonists & inhibitors , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/pharmacology , Glucosides/chemical synthesis , Glucosides/pharmacology , Sulfones/chemical synthesis , Sulfones/pharmacology , Animals , Carbohydrate Conformation , Cell Membrane/drug effects , Enzyme Inhibitors/chemistry , Glucosides/chemistry , Goats , Male , Spermatozoa/drug effects , Spermatozoa/enzymology , Structure-Activity Relationship , Sulfones/chemistry
20.
Mol Cell Biochem ; 336(1-2): 39-48, 2010 Mar.
Article in English | MEDLINE | ID: mdl-19802524

ABSTRACT

Protein tyrosine phosphorylation is a key event accompanying sperm capacitation. Although this signaling cascade generates an array of tyrosine-phosphorylated polypeptides, their molecular characterization is still limited. It is necessary to differentiate the localization of the tyrosine-phosphorylated proteins in spermatozoa to understand the link between the different phosphorylated proteins and the corresponding regulated sperm function. cAMP plays a pivotal role in the regulation of tyrosine phosphorylation. The intracellular cAMP levels were raised in goat spermatozoa by the addition of the phosphodiesterase inhibitor, IBMX in conjugation with caffeine. Tyrosine phosphorylation was significantly up-regulated following treatment with these two reagents. Treatment of caudal spermatozoa with IBMX and caffeine, time dependent up-regulated phosphorylation of the protein of molecular weights 50 and 200 kDa was observed. Increased phosphorylation was observed with a combination of IBMX and caffeine treatment. Tyrosine phosphorylation in caput spermatozoa was not affected significantly under these conditions. The expression level of tyrosine kinase in sperm was examined with specific inhibitors and with anti-phosphotyrosine antibody. The indirect immunofluorescence staining was carried out on ethanol permeabilized sperm using anti-phosphotyrosine antibody. Western blot analysis was done using two separate PKA antibodies: anti-PKA catalytic and anti-PKA RIalpha. Almost no difference was found in the intracellular presence of the PKA RIalpha and RIIalpha subunits in caput and caudal epididymal spermatozoa. However, the catalytic subunit seemed to be present in higher amount in caudal spermatozoa. The results show that caprine sperm displays an enhancement of phosphorylation in the tyrosine residues of specific proteins under in vitro capacitation conditions.


Subject(s)
Phosphoproteins/metabolism , Protein-Tyrosine Kinases/metabolism , Sperm Capacitation/physiology , Spermatozoa/physiology , Tyrosine/metabolism , 1-Methyl-3-isobutylxanthine/pharmacology , Animals , Antibodies, Phospho-Specific , Caffeine/pharmacology , Cyclic AMP/metabolism , Cyclic AMP-Dependent Protein Kinases/antagonists & inhibitors , Cyclic AMP-Dependent Protein Kinases/metabolism , Epididymis/cytology , Goats , Male , Organ Specificity , Phosphodiesterase Inhibitors/pharmacology , Phosphorylation/drug effects , Protein Subunits/metabolism , Protein Transport/drug effects , Protein-Tyrosine Kinases/antagonists & inhibitors , Signal Transduction , Sperm Capacitation/drug effects , Sperm Head/drug effects , Sperm Head/metabolism , Spermatozoa/drug effects , Spermatozoa/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...