Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 32
Filter
Add more filters










Publication year range
1.
Curr Drug Targets ; 24(11): 889-918, 2023.
Article in English | MEDLINE | ID: mdl-37519203

ABSTRACT

Microtubules are a well-known target in cancer chemotherapy because of their critical role in cell division. Chromosome segregation during mitosis depends on the establishment of the mitotic spindle apparatus through microtubule dynamics. The disruption of microtubule dynamics through the stabilization or destabilization of microtubules results in the mitotic arrest of the cells. Microtubule-targeted drugs, which interfere with microtubule dynamics, inhibit the growth of cells at the mitotic phase and induce apoptotic cell death. The principle of microtubule-targeted drugs is to arrest the cells at mitosis and reduce their growth because cancer is a disease of unchecked cell proliferation. Many anti-microtubule agents produce significant inhibition of cancer cell growth and are widely used as chemotherapeutic drugs for the treatment of cancer. The drugs that interact with microtubules generally bind at one of the three sites vinblastine site, taxol site, or colchicine site. Colchicine binds to the interface of tubulin heterodimer and induces the depolymerization of microtubules. The colchicine binding site on microtubules is a much sought-after target in the history of anti-microtubule drug discovery. Many colchicine-binding site inhibitors have been discovered, but their use in the treatment of cancer is limited due to their dose-limiting toxicity and resistance in humans. Combination therapy can be a new treatment strategy to overcome these drawbacks of currently available microtubule-targeted anticancer drugs. This review discusses the significance of microtubules as a potential pharmacological target for cancer and stresses the necessity of finding new microtubule inhibitors to fight the disease.


Subject(s)
Antineoplastic Agents , Neoplasms , Humans , HeLa Cells , Microtubules/metabolism , Mitosis , Tubulin/chemistry , Tubulin/metabolism , Colchicine/metabolism , Colchicine/pharmacology , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Antineoplastic Agents/metabolism , Neoplasms/drug therapy , Neoplasms/metabolism
2.
Chem Biodivers ; 20(5): e202201043, 2023 May.
Article in English | MEDLINE | ID: mdl-37040122

ABSTRACT

Biological activities of a series of palladium(II) complexes (M1-M9) bearing N∩ N, N∩ S, and N∩ O chelating ligands are reported. The palladium complexes were tested for their cytotoxic properties against human cervical cancer (HeLa) cells and antibacterial activity against Gm+ve and Gm-ve bacteria. Among the palladium complexes studied (M1-M9), the complex M5, M8, and M9 were found to be more effective in inhibiting the proliferation of HeLa cells. Hence, these complexes were further investigated for their potential role in cellular damage and apoptosis. DCFDA staining, Rhodamine 123 staining and DNA cleavage assay revealed that complex M5, M8 and M9 induced apoptotic cell death in HeLa cells through ROS generation, DNA damage and mitochondrial depolarization. Computational and titration studies also indicated strong electrostatic interaction with DNA groove. Most of the complexes exhibited good antibacterial activity against both Gm+ve and Gm-ve bacteria. The antibacterial activity of the compounds could not be correlated with their anticancer activity indicating a differential mechanism at their effective concentrations. The detailed study on the antibacterial mechanism of the most potent complex M7 revealed that it exerted its antibacterial activity by inhibiting the function of FtsZ and perturbing the localization of the Z-ring at the mid cell.


Subject(s)
Antineoplastic Agents , Coordination Complexes , Humans , HeLa Cells , Palladium/pharmacology , Anti-Bacterial Agents/pharmacology , Bacteria , Coordination Complexes/pharmacology , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Apoptosis
3.
Mol Divers ; 27(3): 1203-1221, 2023 Jun.
Article in English | MEDLINE | ID: mdl-35789974

ABSTRACT

We discovered tolvaptan as a new Eg5 inhibitor using molecular dynamics simulation-based virtual screening. The Eg5-monastrol, Eg5-ispinesib, and Eg5-STLC complexes with "closed" L5 conformation obtained in MD simulation were used to generate a combined pharmacophore model, and this model was used during the process of virtual screening. Further, the MD simulation for 1 µs showed that the binding of tolvaptan to Eg5 was stable due to the closure of the α2/L5/α3 pocket. Tolvaptan belongs to the class of drugs called vaptans which are non-peptide vasopressin receptor antagonists. Since our virtual search for mitotic inhibitors identified tolvaptan as a potential candidate, we were interested in unraveling its antimitotic mechanism. Tolvaptan bound to purified Eg5-437H with a dissociation constant of 27 ± 3.8 µM. Tolvaptan inhibited the growth of HeLa cells through the mitotic block, and around 70% of these mitotic cells exhibited a characteristic monopolar spindle. Tolvaptan bound to goat brain tubulin with a dissociation constant of 103 ± 13 µM. The binding location of tolvaptan on tubulin overlapped with that of colchicine, according to molecular docking analysis. The combination of tolvaptan with STLC augmented mitotic bock with monopolar cells, whereas its combination with vinblastine increased mitotic block with bipolar cells. Since tolvaptan is found to have a significant cytotoxic effect on HeLa cells, it can be developed as a prospective anticancer agent either alone or in combination with other antimitotic drugs. Tolvaptan was identified as an inhibitor of Eg5 in a MD simulation-based virtual screening using a combined pharmacophore model.


Subject(s)
Antimitotic Agents , Antineoplastic Agents , Humans , Tolvaptan/pharmacology , HeLa Cells , Molecular Docking Simulation , Tubulin , Prospective Studies , Kinesins/chemistry , Kinesins/metabolism , Antineoplastic Agents/pharmacology
4.
Cytotechnology ; 74(1): 141-162, 2022 Feb.
Article in English | MEDLINE | ID: mdl-35185291

ABSTRACT

Tioconazole is one of the drugs used to treat topical mycotic infections. It exhibited severe toxicity during systemic administration; however, the molecular mechanism behind the cytotoxic effect was not well established. We employed HeLa cells as a model to investigate the molecular mechanism of its toxicity and discovered that tioconazole inhibited HeLa cell growth through mitotic block (37%). At the half-maximal inhibitory concentration (≈ 15 µM) tioconazole apparently depolymerized microtubules and caused defects in chromosomal congression at the metaphase plate. Tioconazole induced apoptosis and significantly hindered the migration of HeLa cells. Tioconazole bound to goat brain tubulin (K d, 28.3 ± 0.5 µM) and inhibited the assembly of microtubules in the in vitro assays. We report for the first time that tioconazole binds near to the colchicine site, based on the evidence from in vitro tubulin competition experiment and computational analysis. The conformation of tubulin dimer was found to be "curved" upon binding with tioconazole in the MD simulation. Tioconazole in combination with vinblastine synergistically inhibited the growth of HeLa cells and augmented the percentage of mitotic block by synergistically inhibiting the assembly of microtubules. Our study indicates that the systemic adverse effects of tioconazole are partly due to its effects on microtubules and cell cycle arrest. Since tioconazole is well tolerated at the topical level, it could be developed as a topical anticancer agent in combination with other systemic anticancer drugs. SUPPLEMENTARY INFORMATION: The online version contains supplementary material available at 10.1007/s10616-021-00516-w.

5.
J Fluoresc ; 32(1): 247-256, 2022 Jan.
Article in English | MEDLINE | ID: mdl-34731386

ABSTRACT

Purpurin (1,2,4-trihydroxy-9,10-anthraquinone) is a natural red dye obtained from the red madder plant that is widely used in food and dyeing industries. The present study investigated the characteristics of purpurin and its application as a pH-sensitive probe to detect the pH of solutions and intracellular pH of mammalian and bacterial cells. Purpurin exhibited high pH-sensitive behavior, low analytes interference, high stability with pKa of 4.6 and visible colorimetric change. 1H NMR and FTIR studies indicated protonation of phenolic hydroxyl group under acidic condition with hypsochromic shift in the absorption and fluorescence spectra relative to that of basic condition. Cell culture studies using HeLa cells revealed that purpurin is well tolerated by the cells and the fluorescent imaging result indicated excellent cell permeability with possible use of the dye to detect the pH fluctuations in living cells under various physiological conditions such as apoptosis. Microbiological studies indicated that the dye could be used for visualization of bacteria under acidic condition.


Subject(s)
Anthraquinones , Apoptosis , Biosensing Techniques/methods , Coloring Agents , Molecular Imaging/methods , Animals , Anthraquinones/isolation & purification , Coloring Agents/isolation & purification , HeLa Cells , Humans , Hydrogen-Ion Concentration , Magnetic Resonance Spectroscopy , Microscopy, Fluorescence , Plants/chemistry
6.
Life Sci ; 287: 120105, 2021 Dec 15.
Article in English | MEDLINE | ID: mdl-34756929

ABSTRACT

AIM: Analysis of the anticancer and antimitotic activity of the plant derived alkaloid securinine along with its effect on the organization of cellular microtubules as well as its binding with purified goat brain tubulin in-vitro. MATERIALS AND METHODS: The cytotoxicity of securinine on different cell lines was conducted using SRB assay. The effect of securinine on the cellular microtubules was analyzed using immunofluorescence microscopy. The binding of securinine on purified goat brain tubulin was evaluated using fluorescent spectroscopy. KEY FINDINGS: Securinine effectively prevented the proliferation of cervical, breast and lung cancer cells with an IC50 of 6, 10 and 11 µM respectively and induced minimal toxicity in HEK cell line. Securinine at concentrations higher than IC50 induced significant depolymerization in interphase and mitotic microtubules and it suppressed the reassembly of cold depolymerized spindle microtubules in HeLa cells. In the wound healing assay, securinine effectively suppressed the migration of HeLa cells to close the wound. Securinine bound to tubulin with a Kd of 9.7 µM and inhibited the assembly of tubulin into microtubules. The treatment with securinine induced a mitochondrial dependent ROS response in HeLa cells which enhanced the cytotoxic effect of securinine. The result from gene expression studies indicates that securinine induced apoptosis in MCF-7 cells through p53 dependent pathway. SIGNIFICANCE: Considering the strong anticancer and anti-metastatic property and low toxicity in non-malignant cell lines, we suggest that securinine can be used as a chemotherapeutic drug either alone or in combination with other known anticancer molecules.


Subject(s)
Antineoplastic Agents/metabolism , Azepines/metabolism , Heterocyclic Compounds, Bridged-Ring/metabolism , Lactones/metabolism , Microtubules/drug effects , Mitosis/drug effects , Neoplasms/metabolism , Piperidines/metabolism , Tubulin/metabolism , A549 Cells , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Azepines/pharmacology , Azepines/therapeutic use , Dose-Response Relationship, Drug , HEK293 Cells , HeLa Cells , Heterocyclic Compounds, Bridged-Ring/pharmacology , Heterocyclic Compounds, Bridged-Ring/therapeutic use , Humans , Lactones/pharmacology , Lactones/therapeutic use , MCF-7 Cells , Microtubules/metabolism , Mitosis/physiology , Neoplasms/drug therapy , Piperidines/pharmacology , Piperidines/therapeutic use
7.
Bioorg Med Chem ; 50: 116463, 2021 11 15.
Article in English | MEDLINE | ID: mdl-34700238

ABSTRACT

There is an increasing demand to discover novel antibacterial drugs to counter the ever-evolving genetic machinery of bacteria. The cell division protein FtsZ plays a vital role in bacterial cytokinesis and has been recognized as an effective antibacterial drug target. In this study, we have shown that the madder dye purpurin inhibited bacterial cytokinesis through perturbation of FtsZ assembly. Purpurin inhibited the growth of bacterial cells in a concentration-dependent manner and induced bacterial cell filamentation. Microscopy studies showed that it inhibited the localization of the Z ring at the midcell, and FtsZ was dispersed throughout the cells. Further, purpurin bound firmly to FtsZ with a dissociation constant of 11 µM and inhibited its assembly in vitro. It reduced the GTP hydrolysis by binding closer to the nucleotide-binding site of FtsZ. Purpurin inhibited the proliferation of mammalian cancer cells at higher concentrations without disturbing the polymerization of tubulin. The results collectively suggest that the natural anthraquinone purpurin can potently inhibit the growth of bacteria and serve as a lead molecule for the development of antibacterial agents.


Subject(s)
Anthraquinones/pharmacology , Anti-Bacterial Agents/pharmacology , Antineoplastic Agents/pharmacology , Bacillus subtilis/drug effects , Bacterial Proteins/antagonists & inhibitors , Biological Products/pharmacology , Cytoskeletal Proteins/antagonists & inhibitors , Escherichia coli/drug effects , Anthraquinones/chemistry , Anthraquinones/isolation & purification , Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/isolation & purification , Antineoplastic Agents/chemistry , Antineoplastic Agents/isolation & purification , Bacillus subtilis/chemistry , Biological Products/chemistry , Biological Products/isolation & purification , Cell Proliferation/drug effects , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , Escherichia coli/chemistry , HeLa Cells , Humans , Models, Molecular , Molecular Structure , Structure-Activity Relationship , Tumor Cells, Cultured
8.
J Biol Inorg Chem ; 26(4): 511-531, 2021 06.
Article in English | MEDLINE | ID: mdl-34057639

ABSTRACT

Vanadocene dichloride (VDC), a vanadium containing metallocene dihalide exhibits promising anticancer activity. However, its mechanism of action remains elusive as several diverse targets and pathways have been proposed for its anticancer activity. In this study, we observed that VDC inhibited the proliferation of mammalian cancer cells and induced apoptotic cell death by altering the mitochondrial membrane potential and the expression of bcl2 and bax. Probing further into its anticancer mechanism, we found that VDC caused depolymerization of interphase microtubules and blocked the cells at mitosis with considerable proportion of cells exhibiting monopolar spindles. The reassembly of cold depolymerized microtubules was strongly inhibited in the presence of 10 µM VDC. VDC perturbed the microtubule-kinetochore interactions during mitosis as indicated by the absence of cold stable spindle microtubules in the cells treated with 20 µM VDC. Using goat brain tubulin, we found that VDC inhibited the steady-state polymer mass of microtubules and bound to tubulin at a novel site with a Kd of 9.71 ± 0.19 µM and perturbed the secondary structure of tubulin dimer. In addition, VDC was also found to bind to the mitotic kinesin Eg5 and inhibit its basal as well as microtubule stimulated ATPase activity. The results suggest that disruption of microtubule assembly dynamics and inhibition of the ATPase activity of Eg5 could be a plausible mechanism for the antiproliferative and antimitotic activity of VDC.Graphic abstract.


Subject(s)
Apoptosis/drug effects , Gene Expression Regulation/drug effects , Kinesins/metabolism , Microtubules/drug effects , Vanadium Compounds/pharmacology , Acridine Orange , Cell Proliferation/drug effects , HeLa Cells , Hep G2 Cells , Humans , Kinesins/genetics , Microtubules/metabolism
9.
Naunyn Schmiedebergs Arch Pharmacol ; 394(6): 1231-1249, 2021 06.
Article in English | MEDLINE | ID: mdl-33620548

ABSTRACT

Econazole, miconazole, and sertaconazole, the structurally related azoles with imidazole moiety, were evaluated for their cytotoxicity and their ability to bind to mammalian tubulin. Our results indicated that sertaconazole and econazole bound to goat brain tubulin with a dissociation constant of 9 and 19 µM respectively, while miconazole did not bind to goat brain tubulin. Econazole, miconazole, and sertaconazole inhibited the proliferation of HeLa cells with an IC50 of 28, 98, and 38 µM respectively with sertaconazole alone inducing a mitotic block in the treated cells. Since sertaconazole bound to goat brain tubulin with higher affinity and blocked the cells at mitosis, we hypothesized that its cytotoxic mechanism might involve inhibition of tubulin and econazole which did not block the cells at mitosis may have additional targets than tubulin. Sertaconazole inhibited the polymerization of tubulin in HeLa cells and the in vitro assembled goat brain tubulin. Competitive tubulin-binding assay using colchicine and computational simulation studies showed that sertaconazole bound closer to the colchicine site and induced the tubulin dimer to adopt a "bent" conformation which is incompetent for the polymerization. Results from RT-PCR analysis of the A549 cells treated with sertaconazole indicated activation of apoptosis. Sertaconazole significantly inhibited the migration of HeLa cells and showed synergistic antiproliferative potential with vinblastine. Collectively, the results suggest that sertaconazole which is already in clinical practice could be useful as a topical chemotherapy agent for the treatment of skin cancers in combination with other systemic anticancer agents.


Subject(s)
Antineoplastic Agents/pharmacology , Imidazoles/pharmacology , Microtubules/drug effects , Mitosis/drug effects , Thiophenes/pharmacology , A549 Cells , Animals , Apoptosis/drug effects , Cell Movement/drug effects , Cell Proliferation/drug effects , Drug Synergism , Econazole/pharmacology , Goats , HEK293 Cells , HeLa Cells , Humans , Inhibitory Concentration 50 , Miconazole/pharmacology , Tubulin/drug effects , Tubulin/metabolism , Vinblastine/administration & dosage
11.
Mol Inform ; 39(3): e1900035, 2020 03.
Article in English | MEDLINE | ID: mdl-31347789

ABSTRACT

The interaction of Kif15 with kinesin binding protein (KBP) is critical for its microtubule localization, bundling of kinetochore microtubules and proper alignment of chromosomes at the metaphase plate. The Kif15-KBP structure was prepared from the crystal structure of Kif15 and nonhomologous model of KBP through docking. Benserazide was retrieved when we did a screening of the ZINC Drug Database using the pharmacophore model generated from the potential binding site on Kif15 in an effort to identify molecules for repurposing as Kif15 inhibitors. Live cell imaging of HeLa cells revealed that benserazide delayed metaphase to anaphase-onset by 47±10 min compared to control cells. Benserazide treatment perturbed the kinetochore and microtubule interaction and inhibited the proliferation of HeLa cells with an IC50 of 101 µM with a mitotic block of 12 %. It did not bind to tubulin in the in vitro assays suggesting that the observed effects could be due to its perturbation of Kif15-KBP interaction.


Subject(s)
Benserazide/pharmacology , Chromosomes/drug effects , Computer Simulation , Kinesins/chemistry , Metaphase/drug effects , Benserazide/chemistry , Binding Sites/drug effects , HeLa Cells , Humans , Models, Molecular , Optical Imaging , Tumor Cells, Cultured
13.
Spectrochim Acta A Mol Biomol Spectrosc ; 226: 117584, 2020 Feb 05.
Article in English | MEDLINE | ID: mdl-31698317

ABSTRACT

Trifluoperazine is a potent antipsychotic drug used in the treatment of neurological disorders. The usage of trifluoperazine is often found to be associated with more adverse side effects as compared to other low-potency antipsychotic agents. Plasma proteins play an inevitable role in determining the pharmacokinetic properties of a drug. Hence, this study was conducted with an aim to characterize the interactions of trifluoperazine with bovine serum albumin and determine the influence of other small molecules on its interaction with serum albumin. Trifluoperazine bound to BSA at two independent sites with Kd values of 9.5 and 172.6 µM. Förster resonance energy transfer and computational docking analysis revealed that both the binding sites of trifluoperazine were located closer to TRP 213 in subdomain IIA of BSA. Evaluation of trifluoperazine-BSA interactions at three different temperatures indicated that there was a stable complex formation between the two molecules at the ground state and that the static quenching mechanism was predominant behind these interactions. Binding studies in the presence of pharmaceutically relevant drugs indicated that warfarin, paracetamol, and caffeine negatively influenced the binding of trifluoperazine on BSA. Lastly, Fourier transformed infrared spectroscopy and circular dichroism spectroscopy indicated that the binding of trifluoperazine induced a conformational change by reducing the α-helical content of BSA. The study implicates that the small molecules which prefer binding to the Sudlow site I of BSA might compete with trifluoperazine for its binding site thereby increasing the concentration of free trifluoperazine in the plasma which could lead to adverse side effects in patients.


Subject(s)
Molecular Docking Simulation , Serum Albumin, Bovine/chemistry , Serum Albumin, Bovine/drug effects , Serum Albumin, Bovine/metabolism , Trifluoperazine/pharmacokinetics , Animals , Antipsychotic Agents/chemistry , Antipsychotic Agents/pharmacokinetics , Binding Sites/drug effects , Cattle , Drug Interactions , Fluorescence Resonance Energy Transfer , Molecular Docking Simulation/methods , Protein Binding/drug effects , Protein Conformation , Spectrometry, Fluorescence , Spectrophotometry, Ultraviolet , Trifluoperazine/chemistry , Warfarin/chemistry , Warfarin/metabolism , Warfarin/pharmacokinetics
14.
Mater Sci Eng C Mater Biol Appl ; 103: 109753, 2019 Oct.
Article in English | MEDLINE | ID: mdl-31349477

ABSTRACT

Type 1 diabetes mellitus (DM) is a metabolic disorder associated with impaired carbohydrate metabolism. We present a promising bioinspired approach against type 1 DM using yeast microcapsule (YMC). The glucan component in the outer shell of baker's yeast undergoes receptor-mediated uptake by phagocytic cells through M cell-mediated endocytosis. Thus, a drug can be expected to be delivered to the systemic circulation via lymphatic transport if it is attached to the surface of YMC. For the first time, this possibility has been explored by surface loading of insulin onto YMC. The electrostatic interaction between oppositely charged YMC and insulin resulted in the formation of insulin-loaded yeast microcapsule (IYMC) which was confirmed by fluorescence imaging. Alginate coating provided to IYMC protects YMC from the harsh environment of the gastrointestinal tract and prevents the degradation of insulin in IYMC. Cellular uptake of FITC conjugated IYMC by RAW macrophages confirmed the proposed mechanism of insulin uptake. Moreover, an in vitro method using YMC-imprinted gel was developed for insulin release study from the bioinspired system. Molecular docking studies proved the interaction of insulin with ß-glucan and alginate. A significant hypoglycemic effect was observed after oral administration of the alginate coated insulin-loaded yeast microcapsule (AL-IYMC) in diabetic rats. The AL-IYMCs could serve as a promising approach towards the oral delivery of insulin.


Subject(s)
Capsules/administration & dosage , Drug Delivery Systems/methods , Insulin/administration & dosage , Insulin/pharmacokinetics , Saccharomyces cerevisiae , Administration, Oral , Alginates , Animals , Capsules/chemistry , Diabetes Mellitus, Experimental/drug therapy , Drug Carriers/administration & dosage , Drug Carriers/chemistry , Drug Liberation , Hypoglycemic Agents/administration & dosage , Hypoglycemic Agents/analysis , Hypoglycemic Agents/pharmacokinetics , Insulin/analysis , Intestines/drug effects , Intestines/pathology , Mice , Molecular Docking Simulation , Molecular Imaging , Particle Size , RAW 264.7 Cells , Rats, Sprague-Dawley , Saccharomyces cerevisiae/chemistry , Saccharomyces cerevisiae/cytology , Spectroscopy, Fourier Transform Infrared
15.
Pestic Biochem Physiol ; 157: 108-121, 2019 Jun.
Article in English | MEDLINE | ID: mdl-31153458

ABSTRACT

Karanjin, a furanoflavonol from Pongamia pinnata L is used in agricultural practices for its pesticidal, insecticidal and acaricidal activities. It is commercially available as a bio-pesticide targeting a wide variety of pests. The present study was intended to evaluate the biochemical interactions of karanjin with bovine serum albumin (BSA) and study its toxicological effects on mammalian and bacterial cell lines. Karanjin bound to BSA at a single site with a dissociation constant of 19.7 µM. Evaluation of BSA-karanjin interactions at three different temperatures indicated the involvement of static mode of quenching. Binding experiments in the presence of warfarin and computational docking analysis indicated that karanjin bound closer to the warfarin binding site located in the Subdomain IIA of BSA. Using Förster resonance energy transfer analysis the distance between TRP 213 of BSA and karanjin was found to be 20 Å. Collective results from synchronous fluorescence spectra analysis, differential scanning calorimetry, and circular dichroism analysis indicated that binding of karanjin induced conformational changes in the secondary structure of BSA. Karanjin exhibited low toxicity against human cervical cancer cells and normal mouse fibroblast L929 cells and modestly inhibited the growth of B. subtilis and E. coli cells. The data presented in this study provides insights for understanding the binding interactions of karanjin with BSA and its possible toxicological effects on mammalian cell lines and bacteria.


Subject(s)
Benzopyrans/metabolism , Benzopyrans/toxicity , Plant Oils/chemistry , Plant Oils/toxicity , Pongamia/chemistry , Seeds/chemistry , Animals , Binding Sites , Biological Control Agents/chemistry , Biological Control Agents/toxicity , Cattle , Cell Line , Cell Line, Tumor , Cell Survival/drug effects , Escherichia coli/drug effects , Fluorescence Resonance Energy Transfer , Humans , Mice , Protein Binding , Serum Albumin, Bovine/metabolism
16.
Int J Biol Macromol ; 127: 628-636, 2019 Apr 15.
Article in English | MEDLINE | ID: mdl-30708020

ABSTRACT

Gene therapy is a promising alternative that ensures effective treatment and cure for cancer. Here, we report graphene-reinforced chitosan (CS) construct based non-viral vector for tumor-targeted gene therapy. The therapeutic gene, pDNA-TNF-α, was loaded on to chitosan-carboxylated graphene oxide (CS-CGO) construct via electrostatic interaction. The pDNA-TNF-α-CS-CGO thus obtained was further passivated with 4,7,10-trioxa-1,13-tridecanediamine for protecting the vector from the mononuclear phagocyte system that contributes to the prolongation of circulation half-life. The surface passivated carrier (PEG-pDNA-TNF-α-CS-CGO) then festooned with the folic acid derived carbon dots (C-dots) for targeting folate receptors that are overexpressed in most of the cancer cells. The results of TEM images and zeta potential values ensured the occurrence of desired changes in each stage of C-dot-PEG-pDNA-TNF-α-CS-CGO formulation. After 14 days of incubation, the anti-angiogenesis effect was observed for final formulation in the chorioallantoic membrane. The results of in vitro gene expression study in cancer cell line show a comparatively higher transfection efficacy of the developed system (C-dot-PEG-pDNA-TNF-α-CS-CGO) than pDNA-TNF-α. The efficiency of the developed gene delivery system was further confirmed using a developed and validated artificial tumor cell apparatus.


Subject(s)
Chitosan , Gene Transfer Techniques , Genetic Therapy/methods , Graphite , Nanoparticles/chemistry , Neoplasms , Tumor Necrosis Factor-alpha , Carbon/chemistry , Carbon/pharmacology , Chitosan/chemistry , Chitosan/pharmacology , Graphite/chemistry , Graphite/pharmacology , HeLa Cells , Humans , Neoplasms/genetics , Neoplasms/metabolism , Neoplasms/pathology , Neoplasms/therapy , Tumor Necrosis Factor-alpha/biosynthesis , Tumor Necrosis Factor-alpha/genetics
17.
Cell Prolif ; 52(2): e12558, 2019 Mar.
Article in English | MEDLINE | ID: mdl-30525278

ABSTRACT

OBJECTIVES: The aim of this study was to elucidate the antimitotic mechanism of zerumbone and to investigate its effect on the HeLa cells in combination with other mitotic blockers. MATERIALS AND METHODS: HeLa cells and fluorescence microscopy were used to analyse the effect of zerumbone on cancer cell lines. Cellular internalization of zerumbone was investigated using FITC-labelled zerumbone. The interaction of zerumbone with tubulin was characterized using fluorescence spectroscopy. The Chou and Talalay equation was used to calculate the combination index. RESULTS: Zerumbone selectively inhibited the proliferation of HeLa cells with an IC50 of 14.2 ± 0.5 µmol/L through enhanced cellular uptake compared to the normal cell line L929. It induced a strong mitotic block with cells exhibiting bipolar spindles at the IC50 and monopolar spindles at 30 µmol/L. Docking analysis indicated that tubulin is the principal target of zerumbone. In vitro studies indicated that it bound to goat brain tubulin with a Kd of 4 µmol/L and disrupted the assembly of tubulin into microtubules. Zerumbone and colchicine had partially overlapping binding site on tubulin. Zerumbone synergistically enhanced the anti-proliferative activity of vinblastine and paclitaxel through augmented mitotic block. CONCLUSION: Our data suggest that disruption of microtubule assembly dynamics is one of the mechanisms of the anti-cancer activity of zerumbone and it can be used in combination therapy targeting cell division.


Subject(s)
Antimitotic Agents/pharmacology , Antineoplastic Agents, Phytogenic/pharmacology , Paclitaxel/pharmacology , Sesquiterpenes/pharmacology , Tubulin/metabolism , Vinblastine/pharmacology , Cell Proliferation/drug effects , Drug Synergism , HeLa Cells , Humans , Mitosis/drug effects , Neoplasms/drug therapy , Neoplasms/metabolism , Neoplasms/pathology
18.
Biomed Pharmacother ; 105: 506-517, 2018 Sep.
Article in English | MEDLINE | ID: mdl-29883946

ABSTRACT

Indirubin, a bis-indole alkaloid used in traditional Chinese medicine has shown remarkable anticancer activity against chronic myelocytic leukemia. The present work was aimed to decipher the underlying molecular mechanisms responsible for its anticancer attributes. Our findings suggest that indirubin inhibited the proliferation of HeLa cells with an IC50 of 40 µM and induced a mitotic block. At concentrations higher than its IC50, indirubin exerted a moderate depolymerizing effect on the interphase microtubular network and spindle microtubules in HeLa cells. Studies with goat brain tubulin indicated that indirubin bound to tubulin at a single site with a dissociation constant of 26 ±â€¯3 µM and inhibited the in vitro polymerization of tubulin into microtubules in the presence of glutamate as well as microtubule-associated proteins. Molecular docking analysis and molecular dynamics simulation studies indicate that indirubin stably binds to tubulin at the interface of the α-ß tubulin heterodimer. Further, indirubin stabilized the binding of colchicine on tubulin and promoted the cysteine residue modification by 5,5'-dithiobis-2-nitrobenzoic acid, indicating towards alteration of tubulin conformation upon binding. In addition, we found that indirubin synergistically enhanced the anti-mitotic and anti-proliferative activity of vinblastine, a known microtubule-targeted agent. Collectively our studies indicate that perturbation of microtubule polymerization dynamics could be one of the possible mechanisms behind the anti-cancer activities of indirubin.


Subject(s)
Alkaloids/metabolism , Antimitotic Agents/pharmacology , Tubulin/metabolism , Vinblastine/pharmacology , Animals , Binding Sites , Brain/metabolism , Cell Cycle/drug effects , Cell Cycle Checkpoints/drug effects , Cell Movement/drug effects , Cell Proliferation/drug effects , Colchicine/metabolism , Drug Synergism , Fibroblasts/cytology , Fibroblasts/drug effects , Goats , HeLa Cells , Humans , Indoles/metabolism , Mice , Microtubules/drug effects , Microtubules/metabolism , Mitosis/drug effects , Molecular Docking Simulation , Polymerization , Protein Binding/drug effects , Protein Multimerization , Spindle Apparatus/drug effects , Spindle Apparatus/metabolism , Tryptophan/metabolism , Tubulin/chemistry , Wound Healing/drug effects
19.
ACS Appl Bio Mater ; 1(5): 1705-1716, 2018 Nov 19.
Article in English | MEDLINE | ID: mdl-34996219

ABSTRACT

Tin(IV) porphyrin scaffolds having long-chain (C8 and C16) alkoxyphenyl group at one meso position were synthesized with axial dichloro and dihydroxo ligands. Modifications with respect to the alkoxy chain length and the axial ligands were investigated using UV-vis and fluorescence spectroscopy as well as cyclic voltammetry. Significantly higher molar absorption coefficients were noted with dichloro axial ligands whereas higher fluorescence quantum yields were observed for C8 alkoxy chain substituents. Electrochemical redox data revealed that the porphyrin ring became difficult to reduce consequent to the presence of electron donating alkoxy chain. Evaluation of the aggregation behavior in different organic solvents and DMF-water mixture has been carried out. The singlet oxygen production and photostability data are supportive of their suitability for antibacterial photodynamic therapy. The antibacterial studies carried out in Gram negative (E. coli) and Gram positive (B. subtilis) bacteria using agar well diffusion and LB broth assay revealed selective toxicity in B. Subtilis cells. Fluorescence microscopy and competitive DNA binding studies with Hoechst 33342 revealed DNA binding at the minor groove which was further substantiated with molecular docking studies.

20.
Nat Prod Res ; 32(23): 2856-2860, 2018 Dec.
Article in English | MEDLINE | ID: mdl-28934859

ABSTRACT

The cashew nut shell liquid (CNSL) from the cashew nut shell of Anacardium occidentale L. has been used to treat skin infections, cracks on soles of feet and cancerous ulcers. In this study, we have purified the technical CNSL, systematically evaluated its anticancer, antibacterial and wound healing activity. The LC-MS data revealed that the purified CNSL contains the compounds, cardanol, anacardic acid and methylcardol. It inhibited the proliferation of HeLa cells with an IC50 of 0.004%(v/v) and caused moderate mitotic block with spindle abnormality. It induced apoptosis in HeLa cells and accelerated wound closure in L929 cells. It inhibited the growth of Bacillus subtilis with an IC50 of 0.35%(v/v) and the treated cells exhibited elongated morphology indicating that suppression of cell division is one of the possible mechanisms of its action. The study suggests that the purified CNSL might have potential applications in anticancer and antibacterial drug development.


Subject(s)
Anacardium/chemistry , Anti-Bacterial Agents/pharmacology , Antineoplastic Agents/pharmacology , Wound Healing/drug effects , Anacardic Acids , Anti-Bacterial Agents/isolation & purification , Antineoplastic Agents/isolation & purification , Cell Proliferation/drug effects , HeLa Cells , Humans , Nuts/chemistry , Phenols , Plant Extracts/chemistry , Plant Extracts/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...