1
|
Bhondwe P, Sengar N, Bodiwala HS, Singh IP, Panda D. An adamantyl-caffeoyl-anilide exhibits broad-spectrum antibacterial activity by inhibiting FtsZ assembly and Z-ring formation. Int J Biol Macromol 2024; 259:129255. [PMID: 38199552 DOI: 10.1016/j.ijbiomac.2024.129255] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2023] [Revised: 01/01/2024] [Accepted: 01/03/2024] [Indexed: 01/12/2024]
Abstract
Several harmful bacteria have evolved resistance to conventional antibiotics due to their extensive usage. FtsZ, a principal bacterial cell division protein, is considered as an important drug target to combat resistance. We identified a caffeoyl anilide derivative, (E)-N-(4-(3-(3,4-dihydroxyphenyl)acryloyl)phenyl)-1-adamantylamide (compound 11) as a new antimicrobial agent targeting FtsZ. Compound 11 caused cell elongation in Mycobacterium smegmatis, Bacillus subtilis, and Escherichia coli cells, indicating that it inhibits cell partitioning. Compound 11 inhibited the assembly of Mycobacterium smegmatis FtsZ (MsFtsZ), forming short and thin filaments in vitro. Interestingly, the compound increased the rate of GTP hydrolysis of MsFtsZ. Compound 11 also impeded the assembly of Mycobacterium tuberculosis FtsZ. Fluorescence and absorption spectroscopic analysis suggested that compound 11 binds to MsFtsZ and produces conformational changes in FtsZ. The docking analysis indicated that the compound binds at the interdomain cleft of MsFtsZ. Further, it caused delocalization of the Z-ring in Mycobacterium smegmatis and Bacillus subtilis without affecting DNA segregation. Notably, compound 11 did not inhibit tubulin polymerization, the eukaryotic homolog of FtsZ, suggesting its specificity on bacteria. The evidence indicated that compound 11 exerts its antibacterial effect by impeding FtsZ assembly and has the potential to be developed as a broad-spectrum antimicrobial agent.
Collapse
Affiliation(s)
- Prajakta Bhondwe
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Mumbai 400076, India
| | - Neha Sengar
- Department of Natural Products, National Institute of Pharmaceutical Education and Research (NIPER), Sector 67, S.A.S. Nagar, Mohali 160062, Punjab, India
| | - Hardik S Bodiwala
- Department of Natural Products, National Institute of Pharmaceutical Education and Research (NIPER), Sector 67, S.A.S. Nagar, Mohali 160062, Punjab, India
| | - Inder Pal Singh
- Department of Natural Products, National Institute of Pharmaceutical Education and Research (NIPER), Sector 67, S.A.S. Nagar, Mohali 160062, Punjab, India
| | - Dulal Panda
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Mumbai 400076, India; Department of Natural Products, National Institute of Pharmaceutical Education and Research (NIPER), Sector 67, S.A.S. Nagar, Mohali 160062, Punjab, India.
| |
Collapse
|
2
|
Andres AE, Mariano A, Rane D, Peterson BR. Quantification of Engagement of Microtubules by Small Molecules in Living Cells by Flow Cytometry. ACS BIO & MED CHEM AU 2022; 2:529-537. [PMID: 36281300 PMCID: PMC9585582 DOI: 10.1021/acsbiomedchemau.2c00031] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Revised: 07/25/2022] [Accepted: 07/27/2022] [Indexed: 11/29/2022]
Abstract
![]()
Drugs such as paclitaxel (Taxol) that bind microtubules
are widely
used for the treatment of cancer. Measurements of the affinity and
selectivity of these compounds for their targets are largely based
on studies of purified proteins, and only a few quantitative methods
for the analysis of interactions of small molecules with microtubules
in living cells have been reported. We describe here a novel method
for rapidly quantifying the affinities of compounds that bind polymerized
tubulin in living HeLa cells. This method uses the fluorescent molecular
probe Pacific Blue-GABA-Taxol in conjunction with verapamil to block
cellular efflux. Under physiologically relevant conditions of 37 °C,
this combination allowed quantification of equilibrium saturation
binding of this probe to cellular microtubules (Kd = 1.7 μM) using flow cytometry. Competitive binding
of the microtubule stabilizers paclitaxel (cellular Ki = 22 nM), docetaxel (cellular Ki = 16 nM), cabazitaxel (cellular Ki = 6 nM), and ixabepilone (cellular Ki = 10 nM) revealed intracellular affinities for microtubules that
closely matched previously reported biochemical affinities. By including
a cooperativity factor (α) for curve fitting of allosteric modulators,
this probe also allowed quantification of binding (Kb) of the microtubule destabilizers colchicine (Kb = 80 nM, α = 0.08), vinblastine (Kb = 7 nM, α = 0.18), and maytansine (Kb = 3 nM, α = 0.21). Screening of this
assay against 1008 NCI diversity compounds identified NSC 93427 as
a novel microtubule destabilizer (Kb =
485 nM, α = 0.02), illustrating the potential of this approach
for drug discovery.
Collapse
Affiliation(s)
- Angelo E. Andres
- Division of Medicinal Chemistry and Pharmacognosy, The Ohio State University, Columbus, Ohio 43210, United States
| | - Andres Mariano
- Division of Medicinal Chemistry and Pharmacognosy, The Ohio State University, Columbus, Ohio 43210, United States
| | - Digamber Rane
- Division of Medicinal Chemistry and Pharmacognosy, The Ohio State University, Columbus, Ohio 43210, United States
| | - Blake R. Peterson
- Division of Medicinal Chemistry and Pharmacognosy, The Ohio State University, Columbus, Ohio 43210, United States
| |
Collapse
|
3
|
Venkatramani A, Mukherjee S, Kumari A, Panda D. Shikonin impedes phase separation and aggregation of tau and protects SH-SY5Y cells from the toxic effects of tau oligomers. Int J Biol Macromol 2022; 204:19-33. [PMID: 35120943 DOI: 10.1016/j.ijbiomac.2022.01.172] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2021] [Revised: 01/20/2022] [Accepted: 01/28/2022] [Indexed: 12/22/2022]
Abstract
Tauopathies such as Alzheimer's and Parkinson's diseases involve the abnormal deposition of tau aggregates in the brain and neuronal tissues. We report that a natural naphthoquinone, shikonin, impeded the oligomerization and fibrillization of tau. The compound strongly inhibited heparin, arachidonic acid, and RNA-induced tau aggregation. Atomic force microscopy, dynamic light scattering, SDS-PAGE, and dot blot assays revealed that shikonin diminished tau oligomerization and decreased the mean size of tau oligomers. Transmission electron microscopy and atomic force microscopy analysis further showed that shikonin could suppress tau fibrillization and shorten the tau filaments. Shikonin inhibited tau droplet formation. The compound significantly reduced the aggregation rate of a tryptophan mutant (Y310W-tau) of tau. In addition, shikonin disaggregated preformed tau filaments with a half-maximal disaggregation concentration (DC50) of 6.3 ± 0.4 μM. Pre-treatment of neuroblastoma cells (SH-SY5Y) with shikonin protected the cells from the toxicity induced by tau oligomers and increased their viability. The findings imply that shikonin inhibited several steps in the tau aggregation pathways, especially the early stages, such as liquid-liquid phase separation. Therefore, shikonin is an attractive candidate for developing a therapy against tauopathy.
Collapse
Affiliation(s)
- Anuradha Venkatramani
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Mumbai 400076, India
| | - Sandipan Mukherjee
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Mumbai 400076, India
| | - Anuradha Kumari
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Mumbai 400076, India
| | - Dulal Panda
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Mumbai 400076, India; National Institute of Pharmaceutical Education and Research (NIPER), Sector 67, SAS. Nagar, Mohali 160062, Punjab, India.
| |
Collapse
|
4
|
Abstract
Taxoids such as paclitaxel (Taxol) are an important class of anticancer drugs that bind β-tubulin and stabilize cellular microtubules. To provide new chemical tools for studies of microtubules, we synthesized derivatives of paclitaxel modified at the 7-position with the small coumarin-derived fluorophore Pacific Blue (PB). Three of these Pacific Blue-Taxoids termed PB-Gly-Taxol, PB-β-Ala-Taxol, and PB-GABA-Taxol bind purified crosslinked microtubules with affinities of 34-265 nM, where the affinity can be tuned based on the length of an amino acid linker. When added to living cells in the presence of verapamil or probenecid as inhibitors of efflux, these compounds allow visualization of the microtubule network by confocal microscopy. We describe methods for the synthesis of these probes, determination of their affinities for crosslinked tubulin, and imaging of microtubules in living HeLa cells. We further describe their uptake by Caco-2 cells and two transporter-deficient Caco-2 knockout cell lines in the absence and presence of efflux inhibitors by flow cytometry. These studies revealed that p-glycoprotein (MDR1) and multidrug-resistance protein 2 (MRP2) are major mediators of efflux of these molecular probes. These compounds provide useful tools for studies of microtubules and cellular efflux transporters in living cells.
Collapse
Affiliation(s)
- Angelo E Andres
- Division of Medicinal Chemistry and Pharmacognosy, College of Pharmacy, The Ohio State University, Columbus, OH, USA
| | - Digamber Rane
- Division of Medicinal Chemistry and Pharmacognosy, College of Pharmacy, The Ohio State University, Columbus, OH, USA
| | - Blake R Peterson
- Division of Medicinal Chemistry and Pharmacognosy, College of Pharmacy, The Ohio State University, Columbus, OH, USA.
| |
Collapse
|
5
|
Ashraf SM, Mahanty S, Rathinasamy K. Securinine induces mitotic block in cancer cells by binding to tubulin and inhibiting microtubule assembly: A possible mechanistic basis for its anticancer activity. Life Sci 2021; 287:120105. [PMID: 34756929 DOI: 10.1016/j.lfs.2021.120105] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2021] [Revised: 10/20/2021] [Accepted: 10/25/2021] [Indexed: 10/20/2022]
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.
Collapse
Affiliation(s)
- Shabeeba M Ashraf
- School of Biotechnology, National Institute of Technology Calicut, Calicut, Kerala, India
| | - Susobhan Mahanty
- School of Biotechnology, National Institute of Technology Calicut, Calicut, Kerala, India
| | - Krishnan Rathinasamy
- School of Biotechnology, National Institute of Technology Calicut, Calicut, Kerala, India.
| |
Collapse
|
6
|
Mukherjee S, Panda D. Contrasting Effects of Ferric and Ferrous Ions on Oligomerization and Droplet Formation of Tau: Implications in Tauopathies and Neurodegeneration. ACS Chem Neurosci 2021; 12:4393-4405. [PMID: 34783530 DOI: 10.1021/acschemneuro.1c00377] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
The dysregulation of metal homeostasis is reported to enhance the aggregation of tau, a key neuronal microtubule-associated protein. Herein, we found that ferric (Fe3+) ions enhanced tau aggregation. Fe3+ and Al3+ induced tau aggregation while several trivalent metal ions such as Cr3+, La3+, and V3+ had no discernable effect on tau aggregation. Fe3+ reduced the critical concentration of tau required for the liquid-liquid phase separation (LLPS); however, Cr3+, La3+, and V3+ did not affect tau droplet formation. Dynamic light scattering, atomic force microscopic, and transmission electron microscopic analysis suggested that Fe3+ significantly increased the formation of tau oligomers and fibrils. In contrast, Fe2+ neither enhanced tau droplet formation nor increased the heparin-induced aggregation of tau. Using a tryptophan mutant (Y310W-tau) of tau, Fe3+ was found to bind to tau with four times higher affinity than Fe2+. Acrylamide quenching of the tryptophan fluorescence of Y310W-tau, 1-anilino-8-naphthalene sulfonate (ANS) fluorescence experiment, and far-UV circular dichroism analysis indicated that Fe3+ decreased the solvent exposure of the tryptophan residue, perturbed the hydrophobic surface arrangement, and disrupted the secondary structure of tau, respectively. The increase in the β-sheet content and a subsequent decrease in the disordered content of tau due to the binding of Fe3+ may favor tau aggregation. Fe3+ may enhance and stabilize the non-covalent interactions between disordered domains of tau molecules leading to tau aggregation. The data highlighted the relationship between the dysregulation of ferric ions and neurodegenerative disorders.
Collapse
Affiliation(s)
- Sandipan Mukherjee
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Mumbai 400076, India
| | - Dulal Panda
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Mumbai 400076, India
- National Institute of Pharmaceutical Education and Research, S.A.S. Nagar, Punjab 160062, India
| |
Collapse
|
7
|
Andrade S, Ramalho MJ, Loureiro JA, Pereira MC. Liposomes as biomembrane models: Biophysical techniques for drug-membrane interaction studies. J Mol Liq 2021. [DOI: 10.1016/j.molliq.2021.116141] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
|
8
|
Mdivi-1 induces spindle abnormalities and augments taxol cytotoxicity in MDA-MB-231 cells. Cell Death Discov 2021; 7:118. [PMID: 34016960 PMCID: PMC8137698 DOI: 10.1038/s41420-021-00495-z] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Revised: 04/03/2021] [Accepted: 04/23/2021] [Indexed: 01/19/2023] Open
Abstract
Taxol is a first-line chemotherapeutic for numerous cancers, including the highly refractory triple-negative breast cancer (TNBC). However, it is often associated with toxic side effects and chemoresistance in breast cancer patients, which greatly limits the clinical utility of the drug. Hence, compounds that act in concert with taxol to promote cytotoxicity may be useful to improve the efficacy of taxol-based chemotherapy. In this study, we demonstrated that mdivi-1, a putative inhibitor of mitochondrial fission protein Drp1, enhances the anticancer effects of taxol and overcomes taxol resistance in a TNBC cell line (MDA-MB-231). Not only did mdivi-1 induce mitotic spindle abnormalities and mitotic arrest when used alone, but it also enhanced taxol-induced antimitotic effects when applied in combination. In addition, mdivi-1 induced pronounced spindle abnormalities and cytotoxicity in a taxol-resistant cell line, indicating that it can overcome taxol resistance. Notably, the antimitotic effects of mdivi-1 were not accompanied by prominent morphological or functional alterations in mitochondria and were Drp1-independent. Instead, mdivi-1 exhibited affinity to tubulin at μM level, inhibited tubulin polymerization, and immediately disrupted spindle assembly when cells entered mitosis. Together, our results show that mdivi-1 associates with tubulin and impedes tubulin polymerization, actions which may underlie its antimitotic activity and its ability to enhance taxol cytotoxicity and overcome taxol resistance in MDA-MB-231 cells. Furthermore, our data imply a possibility that mdivi-1 could be useful to improve the therapeutic efficacy of taxol in breast cancer.
Collapse
|
9
|
Nag D, Dastidar DG, Chakrabarti G. Natural flavonoid morin showed anti-bacterial activity against Vibrio cholera after binding with cell division protein FtsA near ATP binding site. Biochim Biophys Acta Gen Subj 2021; 1865:129931. [PMID: 34023444 DOI: 10.1016/j.bbagen.2021.129931] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2021] [Revised: 05/17/2021] [Accepted: 05/17/2021] [Indexed: 11/27/2022]
Abstract
BACKGROUND Increasing antibiotic-resistance in bacterial strains has boosted the need to find new targets for drug delivery. FtsA, a major bacterial divisome protein can be a potent novel drug-target. METHODS AND RESULTS This study finds, morin (3,5,7,2',4'-pentahydroxyflavone), a bio-available flavonoid, had anti-bacterial activities against Vibrio cholerae, IC50 (50 μM) and MIC (150 μM). Morin (2 mM) kills ~20% of human lung fibroblast (WI38) and human intestinal epithelial (HIEC-6) cells in 24 h in-vitro. Fluorescence studies showed morin binds to VcFtsA (FtsA of V. cholerae) with a Kd of 4.68 ± 0.4 μM, inhibiting the protein's polymerization by 72 ± 7% at 25 μM concentration. Morin also affected VcFtsA's ATPase activity, recording ~80% reduction at 20 μM concentration. The in-silico binding study indicated binding sites of morin and ATP on VcFtsA had overlapping amino acids. Mant-ATP, a fluorescent ATP-derivative, showed increased fluorescence on binding to VcFtsA in absence of morin, but in its presence, Mant-ATP fluorescence decreased. VcFtsA-S40A mutant protein did not bind to morin. CONCLUSIONS VcFtsA-morin interaction inhibits the polymerization of the protein by affecting its ATPase activity. The destabilized VcFtsA assembly in-turn affected the cell division in V. cholerae, yielding an elongated morphology. GENERAL SIGNIFICANCE Collectively, these findings explore the anti-bacterial effect of morin on V. cholerae cells targeting VcFtsA, encouraging it to become a potent anti-bacterial agent. Low cytotoxicity of morin against human cells (host) is therapeutically advantageous. This study will also help in synthesizing novel derivatives that can target VcFtsA more efficiently.
Collapse
Affiliation(s)
- Debasish Nag
- Department of Biotechnology And Dr. B. C. Guha Centre for Genetic Engineering And Biotechnology, University of Calcutta, 35 Ballygunge Circular Road, Kolkata, WB 700 019, India
| | - Debabrata Ghosh Dastidar
- Department of Biotechnology And Dr. B. C. Guha Centre for Genetic Engineering And Biotechnology, University of Calcutta, 35 Ballygunge Circular Road, Kolkata, WB 700 019, India; Guru Nanak Institute of Pharmaceutical Science & Technology, 157/F Nilgunj Road, Panihati, Kolkata 700114, West Bengal, India
| | - Gopal Chakrabarti
- Department of Biotechnology And Dr. B. C. Guha Centre for Genetic Engineering And Biotechnology, University of Calcutta, 35 Ballygunge Circular Road, Kolkata, WB 700 019, India.
| |
Collapse
|
10
|
NMK-BH2, a novel microtubule-depolymerising bis (indolyl)-hydrazide-hydrazone, induces apoptotic and autophagic cell death in cervical cancer cells by binding to tubulin at colchicine - site. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2020; 1867:118762. [PMID: 32502617 DOI: 10.1016/j.bbamcr.2020.118762] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/28/2020] [Revised: 05/10/2020] [Accepted: 05/27/2020] [Indexed: 01/06/2023]
Abstract
BACKGROUND Microtubules, the key components of the eukaryotic cytoskeleton and mitotic spindle, are one of the most sought-after targets for cancer chemotherapy, especially due to their indispensible role in mitosis. Cervical cancer is a prevalent malignancy among women of developing countries including India. In spite of the remarkable therapeutic advancement, the non-specificity of chemotherapeutic drugs adversely affect the patients' survival and well-being, thus, necessitating the quest for novel indole-based anti-microtubule agent against cervical cancer, with high degree of potency and selectivity. METHODS For in vitro studies, we used MTT assay, confocal microscopy, fluorescence microscopy, flow cytometry and Western blot analysis. Study in cell free system was accomplished by spectrophotometry, fluorescence spectroscopy and TEM and computational analysis was done by AutodockTools 1.5.6. RESULTS NMK-BH2 exhibited significant and selective anti-proliferative activity against cervical cancer HeLa cells (IC50 = 1.5 μM) over normal cells. It perturbed the cytoskeletal and spindle microtubules of HeLa cells leading to mitotic block and cell death by apoptosis and autophagy. Furthermore, NMK-BH2 targeted the tubulin-microtubule system through fast and strong binding to the αβ-tubulin heterodimers at colchicine-site. CONCLUSION This study identifies and characterises NMK-BH2 as a novel anti-microtubule agent and provides insights into its key anti-cancer mechanism through two different cell death pathways: apoptosis and autophagy, which are mutually independent. GENERAL SIGNIFICANCE It navigates the potential of the novel bis (indolyl)-hydrazide-hydrazone, NMK-BH2, to serve as lead for development of new generation microtubule-disrupting chemotherapeutic with improved efficacy and remarkable selectivity towards better cure of cervical cancer.
Collapse
|
11
|
Liao VWY, Kumari A, Narlawar R, Vignarajan S, Hibbs DE, Panda D, Groundwater PW. Tubulin-Binding 3,5-Bis(styryl)pyrazoles as Lead Compounds for the Treatment of Castration-Resistant Prostate Cancer. Mol Pharmacol 2020; 97:409-422. [PMID: 32241960 DOI: 10.1124/mol.119.118539] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2019] [Accepted: 03/24/2020] [Indexed: 11/22/2022] Open
Abstract
The microtubule-binding taxanes, docetaxel and cabazitaxel, are administered intravenously for the treatment of castration-resistant prostate cancer (CRPC) as the oral administration of these drugs is largely hampered by their low and highly variable bioavailabilities. Using a simple, rapid, and environmentally friendly microwave-assisted protocol, we have synthesized a number of 3,5-bis(styryl)pyrazoles 2a-l, thus allowing for their screening for antiproliferative activity in the androgen-independent PC3 prostate cancer cell line. Surprisingly, two of these structurally simple 3,5-bis(styryl)pyrazoles (2a and 2l) had concentrations which gave 50% of the maximal inhibition of cell proliferation (GI50) in the low micromolar range in the PC3 cell line and were thus selected for extensive further biologic evaluation (apoptosis and cell cycle analysis, and effects on tubulin and microtubules). Our findings from these studies show that 3,5-bis[(1E)-2(2,6-dichlorophenyl)ethenyl]-1H-pyrazole 2l 1) caused significant effects on the cell cycle in PC3 cells, with the vast majority of treated cells in the G2/M phase (89%); 2) induces cell death in PC3 cells even after the removal of the compound; 3) binds to tubulin [dissociation constant (Kd) 0.4 ± 0.1 μM] and inhibits tubulin polymerization in vitro; 4) had no effect upon the polymerization of the bacterial cell division protein FtsZ (a homolog of tubulin); 5) is competitive with paclitaxel for binding to tubulin but not with vinblastine, crocin, or colchicine; and 6) leads to microtubule depolymerization in PC3 cells. Taken together, these results suggest that 3,5-bis(styryl)pyrazoles warrant further investigation as lead compounds for the treatment of CRPC. SIGNIFICANCE STATEMENT: The taxanes are important components of prostate cancer chemotherapy regimens, but their oral administration is hampered by very low and highly variable oral bioavailabilities resulting from their poor absorption, poor solubility, high first-pass metabolism, and efficient efflux by P-glycoprotein. New chemical entities for the treatment of prostate cancer are thus required, and we report here the synthesis and investigation of the mechanism of action of some bis(styryl)pyrazoles, demonstrating their potential as lead compounds for the treatment of prostate cancer.
Collapse
Affiliation(s)
- Vivian W Y Liao
- Sydney Pharmacy School, Faculty of Medicine and Health (V.W.Y.L., R.N., D.E.H., P.W.G.) and Charles Perkins Centre (S.V.), The University of Sydney, Sydney, New South Wales, Australia; and Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Mumbai, India (A.K., D.P.)
| | - Anuradha Kumari
- Sydney Pharmacy School, Faculty of Medicine and Health (V.W.Y.L., R.N., D.E.H., P.W.G.) and Charles Perkins Centre (S.V.), The University of Sydney, Sydney, New South Wales, Australia; and Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Mumbai, India (A.K., D.P.)
| | - Rajeshwar Narlawar
- Sydney Pharmacy School, Faculty of Medicine and Health (V.W.Y.L., R.N., D.E.H., P.W.G.) and Charles Perkins Centre (S.V.), The University of Sydney, Sydney, New South Wales, Australia; and Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Mumbai, India (A.K., D.P.)
| | - Soma Vignarajan
- Sydney Pharmacy School, Faculty of Medicine and Health (V.W.Y.L., R.N., D.E.H., P.W.G.) and Charles Perkins Centre (S.V.), The University of Sydney, Sydney, New South Wales, Australia; and Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Mumbai, India (A.K., D.P.)
| | - David E Hibbs
- Sydney Pharmacy School, Faculty of Medicine and Health (V.W.Y.L., R.N., D.E.H., P.W.G.) and Charles Perkins Centre (S.V.), The University of Sydney, Sydney, New South Wales, Australia; and Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Mumbai, India (A.K., D.P.)
| | - Dulal Panda
- Sydney Pharmacy School, Faculty of Medicine and Health (V.W.Y.L., R.N., D.E.H., P.W.G.) and Charles Perkins Centre (S.V.), The University of Sydney, Sydney, New South Wales, Australia; and Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Mumbai, India (A.K., D.P.)
| | - Paul W Groundwater
- Sydney Pharmacy School, Faculty of Medicine and Health (V.W.Y.L., R.N., D.E.H., P.W.G.) and Charles Perkins Centre (S.V.), The University of Sydney, Sydney, New South Wales, Australia; and Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Mumbai, India (A.K., D.P.)
| |
Collapse
|
12
|
Rane JS, Kumari A, Panda D. The Acetyl Mimicking Mutation, K274Q in Tau, Enhances the Metal Binding Affinity of Tau and Reduces the Ability of Tau to Protect DNA. ACS Chem Neurosci 2020; 11:291-303. [PMID: 31886644 DOI: 10.1021/acschemneuro.9b00455] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
The aggregation of tau, a microtubule-associated protein, is known to play an important role in several neurological disorders including Alzheimer's disease. Alzheimer's disease is considered to be associated with the dyshomeostasis of metal ions such as aluminum, zinc, copper, and ferric ions. Tau is predominately acetylated at the K274 residue in Alzheimer's disease, and the acetylation of the K274 residue is thought to be linked with dementia. Using acetyl mimicking K274Q mutation in tau, we have examined the effects of the acetylation at K274 residue of tau on the interactions of tau with metal ions and also on the ability of tau to protect DNA from the heat and other stressors. We found that Zn2+ and Al3+ increased the liquid-liquid phase separation of tau, an initial stage of tau aggregation. Further, Zn2+ and Al3+ considerably reduced the critical concentration for the phase separation of K274Q tau. Using far-UV circular dichroism and fluorescence spectroscopy, we provide evidence suggesting that the binding of Zn2+ and Al3+ induces conformational changes in tau. The K274Q mutation enhanced the binding affinity of tau for Zn2+, Al3+, Cu2+, and Fe3+ ions. In addition, Zn2+, Al3+, Cu2+, and Fe3+ significantly enhanced the aggregation propensity of K274Q tau in comparison to tau. Interestingly, tau binds to DNA with a higher affinity than K274Q tau. Tau protects DNA from the DNase treatment in vitro as well as from the heat stress in neuroblastoma cells more efficiently than K274Q tau. The results indicated that the acetylation of K274 residue of tau may increase metal ion-induced toxicity and diminish the ability of tau to protect DNA.
Collapse
Affiliation(s)
- Jitendra Subhash Rane
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Powai, Mumbai-400076, India
| | - Anuradha Kumari
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Powai, Mumbai-400076, India
| | - Dulal Panda
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Powai, Mumbai-400076, India
| |
Collapse
|
13
|
Sasaki JC, Allemang A, Bryce SM, Custer L, Dearfield KL, Dietz Y, Elhajouji A, Escobar PA, Fornace AJ, Froetschl R, Galloway S, Hemmann U, Hendriks G, Li HH, Luijten M, Ouedraogo G, Peel L, Pfuhler S, Roberts DJ, Thybaud V, van Benthem J, Yauk CL, Schuler M. Application of the adverse outcome pathway framework to genotoxic modes of action. ENVIRONMENTAL AND MOLECULAR MUTAGENESIS 2020; 61:114-134. [PMID: 31603995 DOI: 10.1002/em.22339] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/04/2019] [Revised: 09/17/2019] [Accepted: 09/23/2019] [Indexed: 05/22/2023]
Abstract
In May 2017, the Health and Environmental Sciences Institute's Genetic Toxicology Technical Committee hosted a workshop to discuss whether mode of action (MOA) investigation is enhanced through the application of the adverse outcome pathway (AOP) framework. As AOPs are a relatively new approach in genetic toxicology, this report describes how AOPs could be harnessed to advance MOA analysis of genotoxicity pathways using five example case studies. Each of these genetic toxicology AOPs proposed for further development includes the relevant molecular initiating events, key events, and adverse outcomes (AOs), identification and/or further development of the appropriate assays to link an agent to these events, and discussion regarding the biological plausibility of the proposed AOP. A key difference between these proposed genetic toxicology AOPs versus traditional AOPs is that the AO is a genetic toxicology endpoint of potential significance in risk characterization, in contrast to an adverse state of an organism or a population. The first two detailed case studies describe provisional AOPs for aurora kinase inhibition and tubulin binding, leading to the common AO of aneuploidy. The remaining three case studies highlight provisional AOPs that lead to chromosome breakage or mutation via indirect DNA interaction (inhibition of topoisomerase II, production of cellular reactive oxygen species, and inhibition of DNA synthesis). These case studies serve as starting points for genotoxicity AOPs that could ultimately be published and utilized by the broader toxicology community and illustrate the practical considerations and evidence required to formalize such AOPs so that they may be applied to genetic toxicity evaluation schemes. Environ. Mol. Mutagen. 61:114-134, 2020. © 2019 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
| | | | | | - Laura Custer
- Bristol-Myers Squibb Company, Drug Safety Evaluation, New Brunswick, New Jersey
| | | | - Yasmin Dietz
- Sanofi-Aventis Deutschland GmbH, Frankfurt, Germany
| | | | | | | | | | | | | | | | - Heng-Hong Li
- Georgetown University, Washington, District of Columbia
| | - Mirjam Luijten
- National Institute for Public Health and the Environment, Bilthoven, The Netherlands
| | | | - Lauren Peel
- Health and Environmental Sciences Institute, Washington, District of Columbia
| | | | | | - Véronique Thybaud
- Sanofi, Research and Development, Preclinical Safety, Vitry-sur-Seine, France
| | - Jan van Benthem
- National Institute for Public Health and the Environment, Bilthoven, The Netherlands
| | - Carole L Yauk
- Environmental Health Science and Research Bureau, Health Canada, Ottawa, Ontario, Canada
| | - Maik Schuler
- Pfizer Inc, World Wide Research and Development, Groton, Connecticut
| |
Collapse
|
14
|
FtsA-FtsZ interaction in Vibrio cholerae causes conformational change of FtsA resulting in inhibition of ATP hydrolysis and polymerization. Int J Biol Macromol 2019; 142:18-32. [PMID: 31790740 DOI: 10.1016/j.ijbiomac.2019.11.217] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2019] [Revised: 11/26/2019] [Accepted: 11/26/2019] [Indexed: 11/23/2022]
Abstract
Proper interaction between the divisome proteins FtsA and FtsZ is important for the bacterial cell division which is not well characterized till date. In this study, the objective was to understand the mechanism of FtsA-FtsZ interaction using full-length recombinant proteins. We cloned, over-expressed, purified and subsequently characterized FtsA of Vibrio cholerae (VcFtsA). We found that VcFtsA polymerization assembly was dependent on Ca2+ ions, which is unique among FtsA proteins reported until now. VcFtsA also showed ATPase activity and its assembly was ATP dependent. Binding parameters of the interaction between the two full-length proteins, VcFtsA, and VcFtsZ determined by fluorescence spectrophotometry yielded a Kd value of around 38 μM. The Kd value of the interaction was 3 μM when VcFtsA was in ATP bound state. We found that VcFtsZ after interacting with VcFtsA causes a change of secondary structure in the later one leading to loss of its ability to hydrolyze ATP, subsequently halting the VcFtsA polymerization. On the other hand, a double-mutant of VcFtsA (VcFtsA-D242E,R300E), that does not bind to VcFtsZ, polymerized in the presence of VcFtsZ. Though FtsA proteins among different organisms show 70-80% homology in their sequences, assembly of VcFtsA showed a difference in its regulatory processes.
Collapse
|
15
|
Chakrabarty S, Nag D, Ganguli A, Das A, Ghosh Dastidar D, Chakrabarti G. Theaflavin and epigallocatechin-3-gallate synergistically induce apoptosis through inhibition of PI3K/Akt signaling upon depolymerizing microtubules in HeLa cells. J Cell Biochem 2018; 120:5987-6003. [PMID: 30390323 DOI: 10.1002/jcb.27886] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2018] [Accepted: 09/21/2018] [Indexed: 12/20/2022]
Abstract
Theaflavin (TF) and epigallocatechin-3-gallate (EGCG) both have been reported previously as microtubule depolymerizing agents that also have anticancer effects on various cancer cell lines and in animal models. Here, we have applied TF and EGCG in combination on HeLa cells to investigate if they can potentiate each other to improve their anticancer effect in lower doses and the underlying mechanism. We found that TF and EGCG acted synergistically, in lower doses, to inhibit the growth of HeLa cells. We found the combination of 50 µg/mL TF and 20 µg/mL EGCG to be the most effective combination with a combination index of 0.28. The same combination caused larger accumulation of cells in the G 2 /M phase of the cell cycle, potent mitochondrial membrane potential loss, and synergistic augmentation of apoptosis. We have shown that synergistic activity might be due to stronger microtubule depolymerization by simultaneous binding of TF and EGCG at different sites on tubulin: TF binds at vinblastine binding site on tubulin, and EGCG binds near colchicines binding site on tubulin. A detailed mechanistic analysis revealed that stronger microtubule depolymerization caused effective downregulation of PI3K/Akt signaling and potently induced mitochondrial apoptotic signals, which ultimately resulted in the apoptotic death of HeLa cells in a synergistic manner.
Collapse
Affiliation(s)
- Subhendu Chakrabarty
- Department of Biotechnology, and Dr. B. C. Guha Centre for Genetic Engineering and Biotechnology, University of Calcutta, Kolkata, West Bengal, India.,Department of Microbiology, M.U.C. Women's College, Burdwan, West Bengal, India
| | - Debasish Nag
- Department of Biotechnology, and Dr. B. C. Guha Centre for Genetic Engineering and Biotechnology, University of Calcutta, Kolkata, West Bengal, India
| | - Arnab Ganguli
- Department of Biotechnology, and Dr. B. C. Guha Centre for Genetic Engineering and Biotechnology, University of Calcutta, Kolkata, West Bengal, India
| | - Amlan Das
- Department of Biotechnology, NIT Sikkim, Sikkim, India
| | - Debabrata Ghosh Dastidar
- Department of Biotechnology, and Dr. B. C. Guha Centre for Genetic Engineering and Biotechnology, University of Calcutta, Kolkata, West Bengal, India.,Division of Pharmaceutics, Guru Nanak Institute of Pharmaceutical Science and Technology, Kolkata, West Bengal, India
| | - Gopal Chakrabarti
- Department of Biotechnology, and Dr. B. C. Guha Centre for Genetic Engineering and Biotechnology, University of Calcutta, Kolkata, West Bengal, India
| |
Collapse
|
16
|
Wu X, Li Z, Shen Y. The small molecule CS1 inhibits mitosis and sister chromatid resolution in HeLa cells. Biochim Biophys Acta Gen Subj 2018; 1862:1134-1147. [PMID: 29410075 DOI: 10.1016/j.bbagen.2018.01.012] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2017] [Revised: 12/01/2017] [Accepted: 01/18/2018] [Indexed: 01/03/2023]
Abstract
BACKGROUND Mitosis, the most dramatic event in the cell cycle, involves the reorganization of virtually all cellular components. Antimitotic agents are useful for dissecting the mechanism of this reorganization. Previously, we found that the small molecule CS1 accumulates cells in G2/M phase [1], but the mechanism of its action remains unknown. METHODS Cell cycle analysis, live cell imaging and nuclear staining were used. Chromosomal morphology was detected by chromosome spreading. The effects of CS1 on microtubules were confirmed by tubulin polymerization, colchicine tubulin-binding, cellular tubulin polymerization and immunofluorescence assays and by analysis of microtubule dynamics and molecular modeling. Histone phosphoproteomics was performed using mass spectrometry. Cell signaling cascades were analyzed using immunofluorescence, immunoprecipitation, immunoblotting, siRNA knockdown and chemical inhibition of specific proteins. RESULTS The small molecule CS1 was shown to be an antimitotic agent. CS1 potently inhibited microtubule polymerization via interaction with the colchicine-binding pocket of tubulin in vitro and inhibited the formation of the spindle apparatus by reducing the bulk of growing microtubules in HeLa cells, which led to activation of the spindle assembly checkpoint (SAC) and mitotic arrest of HeLa cells. Compared with colchicine, CS1 impaired the progression of sister chromatid resolution independent of cohesin dissociation, and this was reversed by the removal of CS1. Additionally, CS1 induced unique histone phosphorylation patterns distinct from those induced by colchicine. CONCLUSIONS AND SIGNIFICANCE CS1 is a unique antimitotic small molecule and a powerful tool with unprecedented value over colchicine that makes it possible to specifically and conditionally perturb mitotic progression.
Collapse
Affiliation(s)
- Xingkang Wu
- Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Shandong University, No. 44 West Wenhua Road, Jinan, Shandong 250012, PR China
| | - Zhenyu Li
- Department of Pharmacy, Shandong Provincial Hospital Affiliated to Shandong University, Jinan 250021, PR China
| | - Yuemao Shen
- Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Shandong University, No. 44 West Wenhua Road, Jinan, Shandong 250012, PR China; State Key Laboratory of Microbial Technology, Shandong University, No. 27 South Shanda Road, Jinan, Shandong 250100, PR China.
| |
Collapse
|
17
|
Das Mukherjee D, Kumar NM, Tantak MP, Das A, Ganguli A, Datta S, Kumar D, Chakrabarti G. Development of Novel Bis(indolyl)-hydrazide–Hydrazone Derivatives as Potent Microtubule-Targeting Cytotoxic Agents against A549 Lung Cancer Cells. Biochemistry 2016; 55:3020-35. [DOI: 10.1021/acs.biochem.5b01127] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Affiliation(s)
- Dipanwita Das Mukherjee
- Department
of Biotechnology and Dr. B. C. Guha Centre for Genetic Engineering
and Biotechnology, University of Calcutta, 35 Ballygunge Circular Road, Kolkata, WB 700
019, India
| | - N. Maruthi Kumar
- Department
of Chemistry, Birla Institute of Technology and Science, Pilani, Rajasthan 333 031, India
| | - Mukund P. Tantak
- Department
of Chemistry, Birla Institute of Technology and Science, Pilani, Rajasthan 333 031, India
| | - Amlan Das
- Department
of Biotechnology and Dr. B. C. Guha Centre for Genetic Engineering
and Biotechnology, University of Calcutta, 35 Ballygunge Circular Road, Kolkata, WB 700
019, India
| | - Arnab Ganguli
- Department
of Biotechnology and Dr. B. C. Guha Centre for Genetic Engineering
and Biotechnology, University of Calcutta, 35 Ballygunge Circular Road, Kolkata, WB 700
019, India
| | - Satabdi Datta
- Department
of Biotechnology and Dr. B. C. Guha Centre for Genetic Engineering
and Biotechnology, University of Calcutta, 35 Ballygunge Circular Road, Kolkata, WB 700
019, India
| | - Dalip Kumar
- Department
of Chemistry, Birla Institute of Technology and Science, Pilani, Rajasthan 333 031, India
| | - Gopal Chakrabarti
- Department
of Biotechnology and Dr. B. C. Guha Centre for Genetic Engineering
and Biotechnology, University of Calcutta, 35 Ballygunge Circular Road, Kolkata, WB 700
019, India
| |
Collapse
|
18
|
Patra D, Mishra P, Vijayan M, Surolia A. Negative Cooperativity and High Affinity in Chitooligosaccharide Binding by a Mycobacterium smegmatis Protein Containing LysM and Lectin Domains. Biochemistry 2015; 55:49-61. [DOI: 10.1021/acs.biochem.5b00841] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Affiliation(s)
- Dhabaleswar Patra
- Molecular Biophysics Unit, Indian Institute of Science, Bangalore 560 012, India
| | - Padmanabh Mishra
- Molecular Biophysics Unit, Indian Institute of Science, Bangalore 560 012, India
| | - Mamannamana Vijayan
- Molecular Biophysics Unit, Indian Institute of Science, Bangalore 560 012, India
| | - Avadhesha Surolia
- Molecular Biophysics Unit, Indian Institute of Science, Bangalore 560 012, India
| |
Collapse
|
19
|
Rashid A, Kuppa A, Kunwar A, Panda D. Thalidomide (5HPP-33) suppresses microtubule dynamics and depolymerizes the microtubule network by binding at the vinblastine binding site on tubulin. Biochemistry 2015; 54:2149-59. [PMID: 25747795 DOI: 10.1021/bi501429j] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Thalidomides were initially thought to be broad-range drugs specifically for curing insomnia and relieving morning sickness in pregnant women. However, its use was discontinued because of a major drawback of causing teratogenicity. In this study, we found that a thalidomide derivative, 5-hydroxy-2-(2,6-diisopropylphenyl)-1H-isoindole-1,3-dione (5HPP-33), inhibited the proliferation of MCF-7 with a half-maximal inhibitory concentration of 4.5 ± 0.4 μM. 5HPP-33 depolymerized microtubules and inhibited the reassembly of cold-depolymerized microtubules in MCF-7 cells. Using time-lapse imaging, the effect of 5HPP-33 on the dynamics of individual microtubules in live MCF-7 cells was analyzed. 5HPP-33 (5 μM) decreased the rates of growth and shortening excursions by 34 and 33%, respectively, and increased the time microtubules spent in the pause state by 92% as compared to that of the vehicle-treated MCF-7 cells. 5HPP-33 (5 μM) reduced the dynamicity of microtubules by 62% compared to the control. 5HPP-33 treatment reduced the distance between the two poles of a bipolar spindle, induced multipolarity in some of the treated cells, and blocked cells at mitosis. In vitro, 5HPP-33 bound to tubulin with a weak affinity. Vinblastine inhibited the binding of 5HPP-33 to tubulin, and 5HPP-33 inhibited the binding of BODIPY FL-vinblastine to tubulin. Further, a molecular docking analysis suggested that 5HPP-33 shares its binding site on tubulin with vinblastine. The results provided significant insight into the antimitotic mechanism of action of 5HPP-33 and also suggest a possible mechanism for the teratogenicity of thalidomides.
Collapse
Affiliation(s)
- Aijaz Rashid
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Mumbai 400076, India
| | - Annapurna Kuppa
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Mumbai 400076, India
| | - Ambarish Kunwar
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Mumbai 400076, India
| | - Dulal Panda
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Mumbai 400076, India
| |
Collapse
|
20
|
Dhar G, Chakravarty D, Hazra J, Dhar J, Poddar A, Pal M, Chakrabarti P, Surolia A, Bhattacharyya B. Actin–Curcumin Interaction: Insights into the Mechanism of Actin Polymerization Inhibition. Biochemistry 2015; 54:1132-43. [DOI: 10.1021/bi5014408] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Affiliation(s)
- Gopa Dhar
- Department
of Biochemistry, Bose Institute, Kolkata 700054, India
| | | | - Joyita Hazra
- Division
of Molecular Medicine, Bose Institute, Kolkata 700054, India
| | - Jesmita Dhar
- Bioinformatics
Centre, Bose Institute, Kolkata 700054, India
| | - Asim Poddar
- Department
of Biochemistry, Bose Institute, Kolkata 700054, India
| | - Mahadeb Pal
- Division
of Molecular Medicine, Bose Institute, Kolkata 700054, India
| | | | - Avadhesha Surolia
- Molecular
Biophysics Unit, Indian Institute of Science, Bangalore 560012, India
| | | |
Collapse
|
21
|
Boscá F, Sastre G, Andreu JM, Jornet D, Tormos R, Miranda MA. Drug–tubulin interactions interrogated by transient absorption spectroscopy. RSC Adv 2015. [DOI: 10.1039/c5ra05636e] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023] Open
Abstract
The triplet excited state of complexed COL and MTC gives well defined transient spectra undetectable in the absence of TU.
Collapse
Affiliation(s)
- F. Boscá
- Departamento de Química/Instituto Universitario Mixto de Tecnología Química UPV-CSIC
- E-46022 Valencia
- Spain
| | - G. Sastre
- Departamento de Química/Instituto Universitario Mixto de Tecnología Química UPV-CSIC
- E-46022 Valencia
- Spain
| | - J. M. Andreu
- Centro de Investigaciones Biológicas
- CSIC
- E-28040 Madrid
- Spain
| | - D. Jornet
- Departamento de Química/Instituto Universitario Mixto de Tecnología Química UPV-CSIC
- E-46022 Valencia
- Spain
| | - R. Tormos
- Departamento de Química/Instituto Universitario Mixto de Tecnología Química UPV-CSIC
- E-46022 Valencia
- Spain
| | - M. A. Miranda
- Departamento de Química/Instituto Universitario Mixto de Tecnología Química UPV-CSIC
- E-46022 Valencia
- Spain
| |
Collapse
|
22
|
Nacoulma AP, Megalizzi V, Pottier LR, De Lorenzi M, Thoret S, Dubois J, Vandeputte OM, Duez P, Vereecke D, Jaziri ME. Potent antiproliferative cembrenoids accumulate in tobacco upon infection with Rhodococcus fascians and trigger unusual microtubule dynamics in human glioblastoma cells. PLoS One 2013; 8:e77529. [PMID: 24167576 PMCID: PMC3805576 DOI: 10.1371/journal.pone.0077529] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2013] [Accepted: 09/10/2013] [Indexed: 12/26/2022] Open
Abstract
AIMS Though plant metabolic changes are known to occur during interactions with bacteria, these were rarely challenged for pharmacologically active compounds suitable for further drug development. Here, the occurrence of specific chemicals with antiproliferative activity against human cancer cell lines was evidenced in hyperplasia (leafy galls) induced when plants interact with particular phytopathogens, such as the Actinomycete Rhodococcus fascians. METHODS We examined leafy galls fraction F3.1.1 on cell proliferation, cell division and cytoskeletal disorganization of human cancer cell lines using time-lapse videomicroscopy imaging, combined with flow cytometry and immunofluorescence analysis. We determined the F3.1.1-fraction composition by gas chromatography coupled to mass spectrometry. RESULTS The leafy galls induced on tobacco by R. fascians yielded fraction F3.1.1 which inhibited proliferation of glioblastoma U373 cells with an IC50 of 4.5 µg/mL, F.3.1.1 was shown to increase cell division duration, cause nuclear morphological deformations and cell enlargement, and, at higher concentrations, karyokinesis defects leading to polyploidization and apoptosis. F3.1.1 consisted of a mixture of isomers belonging to the cembrenoids. The cellular defects induced by F3.1.1 were caused by a peculiar cytoskeletal disorganization, with the occurrence of fragmented tubulin and strongly organized microtubule aggregates within the same cell. Colchicine, paclitaxel, and cembrene also affected U373 cell proliferation and karyokinesis, but the induced microtubule rearrangement was very different from that provoked by F3.1.1. Altogether our data indicate that the cembrenoid isomers in F3.1.1 have a unique mode of action and are able to simultaneously modulate microtubule polymerization and stability.
Collapse
Affiliation(s)
- Aminata P. Nacoulma
- Laboratoire de Toxicologie, Faculté de Pharmacie, Université Libre de Bruxelles, Brussels, Belgium
| | - Veronique Megalizzi
- Laboratoire de Toxicologie, Faculté de Pharmacie, Université Libre de Bruxelles, Brussels, Belgium
| | - Laurent R. Pottier
- Laboratoire de Pharmacognosie, de Bromatologie et de Nutrition Humaine, Faculté de Pharmacie, Université Libre de Bruxelles, Brussels, Belgium
| | - Manuela De Lorenzi
- Laboratoire de Toxicologie, Faculté de Pharmacie, Université Libre de Bruxelles, Brussels, Belgium
| | - Sylviane Thoret
- Institut de Chimie des Substances Naturelles, Centre national de la recherche scientifique Unité PR 2301, Gif-sur-Yvette, France
| | - Joëlle Dubois
- Institut de Chimie des Substances Naturelles, Centre national de la recherche scientifique Unité PR 2301, Gif-sur-Yvette, France
| | - Olivier M. Vandeputte
- Laboratoire de Biotechnologie Végétale, Faculté des Sciences, Université Libre de Bruxelles, Gosselies, Belgium
| | - Pierre Duez
- Laboratoire de Pharmacognosie, de Bromatologie et de Nutrition Humaine, Faculté de Pharmacie, Université Libre de Bruxelles, Brussels, Belgium
| | - Danny Vereecke
- Department of Plant Production, University College Ghent, Ghent University, Gent, Belgium
| | - Mondher El Jaziri
- Laboratoire de Biotechnologie Végétale, Faculté des Sciences, Université Libre de Bruxelles, Gosselies, Belgium
| |
Collapse
|
23
|
Bhattacharya S, Kumar NM, Ganguli A, Tantak MP, Kumar D, Chakrabarti G. NMK-TD-100, a novel microtubule modulating agent, blocks mitosis and induces apoptosis in HeLa cells by binding to tubulin. PLoS One 2013; 8:e76286. [PMID: 24116100 PMCID: PMC3792137 DOI: 10.1371/journal.pone.0076286] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2013] [Accepted: 08/22/2013] [Indexed: 11/19/2022] Open
Abstract
Thiadiazoles are one of the most widely utilized agents in medicinal chemistry, having a wide range of pharmacologic activity. Microtubules (MTs) have always remained a sought-after target in rapidly proliferating cancer cells. We screened for the growth inhibitory effect of synthetic 5-(3-indolyl)-2-substituted-1,3,4-thiadiazoles on cancer cells and identified NMK-TD-100, as the most potent agent. Cell viability experiments using human cervical carcinoma cell line (HeLa cells) indicated that the IC50 value was 1.42±0.11 µM for NMK-TD-100 for 48 h treatment. In further study, we examined the mode of interaction of NMK-TD-100 with tubulin and unraveled the cellular mechanism responsible for its anti-tumor activity. NMK-TD-100 induced arrest in mitotic phase of cell cycle, caused decline in mitochondrial membrane potential and induced apoptosis in HeLa cells. Immunofluorescence studies using an anti-α-tubulin antibody showed a significant depolymerization of the interphase microtubule network and spindle microtubule in HeLa cells in a concentration-dependent manner. However, the cytotoxicity of NMK-TD-100 towards human peripheral blood mononuclear cells (PBMC) was lower compared to that in cancer cells. Polymerization of tissue purified tubulin into microtubules was inhibited by NMK-TD-100 with an IC50 value of 17.5±0.35 µM. The binding of NMK-TD-100 with tubulin was studied using NMK-TD-100 fluorescence enhancement and intrinsic tryptophan fluorescence of tubulin. The stoichiometry of NMK-TD-100 binding to tubulin is 1:1 (molar ratio) with a dissociation constant of ~1 µM. Fluorescence spectroscopic and molecular modeling data showed that NMK-TD-100 binds to tubulin at a site which is very near to the colchicine binding site. The binding of NMK-TD-100 to tubulin was estimated to be ~10 times faster than that of colchicine. The results indicated that NMK-TD-100 exerted anti-proliferative activity by disrupting microtubule functions through tubulin binding and provided insights into its potential of being a chemotherapeutic agent.
Collapse
MESH Headings
- Amino Acids/chemistry
- Amino Acids/metabolism
- Apoptosis/drug effects
- Blotting, Western
- Caspase 3/metabolism
- Cell Proliferation/drug effects
- Cell Survival/drug effects
- Cells, Cultured
- Dose-Response Relationship, Drug
- HeLa Cells
- Humans
- Indoles/chemistry
- Indoles/metabolism
- Indoles/pharmacology
- Leukocytes, Mononuclear/cytology
- Leukocytes, Mononuclear/drug effects
- Leukocytes, Mononuclear/metabolism
- M Phase Cell Cycle Checkpoints/drug effects
- Membrane Potential, Mitochondrial/drug effects
- Microscopy, Electron
- Microtubules/drug effects
- Microtubules/metabolism
- Mitosis/drug effects
- Models, Molecular
- Molecular Structure
- Protein Binding
- Protein Structure, Tertiary
- Proto-Oncogene Proteins c-bcl-2/metabolism
- Thiadiazoles/chemistry
- Thiadiazoles/metabolism
- Thiadiazoles/pharmacology
- Tubulin/chemistry
- Tubulin/metabolism
- Tubulin/ultrastructure
- Tubulin Modulators/chemistry
- Tubulin Modulators/metabolism
- Tubulin Modulators/pharmacology
- Tumor Suppressor Protein p53/metabolism
Collapse
Affiliation(s)
- Surela Bhattacharya
- Department of Biotechnology and Dr. B.C. Guha Centre for Genetic Engineering and Biotechnology, University of Calcutta, Kolkata, India
| | - N. Maruthi Kumar
- Department of Chemistry, Birla Institute of Technology and Science, Pilani, Rajasthan, India
| | - Arnab Ganguli
- Department of Biotechnology and Dr. B.C. Guha Centre for Genetic Engineering and Biotechnology, University of Calcutta, Kolkata, India
| | - Mukund P. Tantak
- Department of Chemistry, Birla Institute of Technology and Science, Pilani, Rajasthan, India
| | - Dalip Kumar
- Department of Chemistry, Birla Institute of Technology and Science, Pilani, Rajasthan, India
- * E-mail: (GC); (DK)
| | - Gopal Chakrabarti
- Department of Biotechnology and Dr. B.C. Guha Centre for Genetic Engineering and Biotechnology, University of Calcutta, Kolkata, India
- * E-mail: (GC); (DK)
| |
Collapse
|
24
|
Bhattacharya A, Jindal B, Singh P, Datta A, Panda D. Plumbagin inhibits cytokinesis inBacillus subtilisby inhibiting FtsZ assembly - a mechanistic study of its antibacterial activity. FEBS J 2013; 280:4585-99. [DOI: 10.1111/febs.12429] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2013] [Revised: 06/24/2013] [Accepted: 07/04/2013] [Indexed: 11/28/2022]
Affiliation(s)
- Anusri Bhattacharya
- Department of Chemistry; Indian Institute of Technology Bombay; Mumbai India
- Department of Biosciences and Bioengineering; Indian Institute of Technology Bombay; Mumbai India
| | - Bhavya Jindal
- Department of Biosciences and Bioengineering; Indian Institute of Technology Bombay; Mumbai India
| | - Parminder Singh
- Department of Biosciences and Bioengineering; Indian Institute of Technology Bombay; Mumbai India
| | - Anindya Datta
- Department of Chemistry; Indian Institute of Technology Bombay; Mumbai India
| | - Dulal Panda
- Department of Biosciences and Bioengineering; Indian Institute of Technology Bombay; Mumbai India
| |
Collapse
|
25
|
CXI-benzo-84 reversibly binds to tubulin at colchicine site and induces apoptosis in cancer cells. Biochem Pharmacol 2013; 86:378-91. [PMID: 23747346 DOI: 10.1016/j.bcp.2013.05.024] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2013] [Revised: 05/21/2013] [Accepted: 05/22/2013] [Indexed: 01/02/2023]
|
26
|
Rai A, Surolia A, Panda D. An antitubulin agent BCFMT inhibits proliferation of cancer cells and induces cell death by inhibiting microtubule dynamics. PLoS One 2012; 7:e44311. [PMID: 22952952 PMCID: PMC3432122 DOI: 10.1371/journal.pone.0044311] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2012] [Accepted: 08/01/2012] [Indexed: 12/30/2022] Open
Abstract
Using cell based screening assay, we identified a novel anti-tubulin agent (Z)-5-((5-(4-bromo-3-chlorophenyl)furan-2-yl)methylene)-2-thioxothiazolidin-4-one (BCFMT) that inhibited proliferation of human cervical carcinoma (HeLa) (IC50, 7.2±1.8 µM), human breast adenocarcinoma (MCF-7) (IC50, 10.0±0.5 µM), highly metastatic breast adenocarcinoma (MDA-MB-231) (IC50, 6.0±1 µM), cisplatin-resistant human ovarian carcinoma (A2780-cis) (IC50, 5.8±0.3 µM) and multi-drug resistant mouse mammary tumor (EMT6/AR1) (IC50, 6.5±1µM) cells. Using several complimentary strategies, BCFMT was found to inhibit cancer cell proliferation at G2/M phase of the cell cycle apparently by targeting microtubules. In addition, BCFMT strongly suppressed the dynamics of individual microtubules in live MCF-7 cells. At its half maximal proliferation inhibitory concentration (10 µM), BCFMT reduced the rates of growing and shortening phases of microtubules in MCF-7 cells by 37 and 40%, respectively. Further, it increased the time microtubules spent in the pause (neither growing nor shortening detectably) state by 135% and reduced the dynamicity (dimer exchange per unit time) of microtubules by 70%. In vitro, BCFMT bound to tubulin with a dissociation constant of 8.3±1.8 µM, inhibited tubulin assembly and suppressed GTPase activity of microtubules. BCFMT competitively inhibited the binding of BODIPY FL-vinblastine to tubulin with an inhibitory concentration (Ki) of 5.2±1.5 µM suggesting that it binds to tubulin at the vinblastine site. In cultured cells, BCFMT-treatment depolymerized interphase microtubules, perturbed the spindle organization and accumulated checkpoint proteins (BubR1 and Mad2) at the kinetochores. BCFMT-treated MCF-7 cells showed enhanced nuclear accumulation of p53 and its downstream p21, which consequently activated apoptosis in these cells. The results suggested that BCFMT inhibits proliferation of several types of cancer cells including drug resistance cells by suppressing microtubule dynamics and indicated that the compound may have chemotherapeutic potential.
Collapse
Affiliation(s)
- Ankit Rai
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Mumbai, Maharashtra, India
| | - Avadhesha Surolia
- Molecular Biophysics Unit, Indian Institute of Science, Bangalore, Karnataka, India
- * E-mail: (DP); (AS)
| | - Dulal Panda
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Mumbai, Maharashtra, India
- * E-mail: (DP); (AS)
| |
Collapse
|
27
|
Gireesh KK, Rashid A, Chakraborti S, Panda D, Manna T. CIL-102 binds to tubulin at colchicine binding site and triggers apoptosis in MCF-7 cells by inducing monopolar and multinucleated cells. Biochem Pharmacol 2012; 84:633-45. [PMID: 22705644 DOI: 10.1016/j.bcp.2012.06.008] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2012] [Revised: 06/05/2012] [Accepted: 06/07/2012] [Indexed: 11/18/2022]
Abstract
A plant dictamine analog, 1-[4-(furo[2,3-b]quinolin-4-ylamino)phenyl]ethanone (CIL-102) has been shown to exert potent anti-tumor activity. In this study, we examined the mode of interaction of CIL-102 with tubulin and unraveled the cellular mechanism responsible for its anti-tumor activity. CIL-102 bound to tubulin at a single site with a dissociation constant ~0.4 μM. Isothermal titration calorimetry revealed that CIL-102-tubulin interaction is highly enthalpy driven and that the binding affords a large negative heat capacity change (ΔC(p) = -790 cal mol(-1) K(-1)) with an enthalpy-entropy compensation. An analysis of the modified Dixon plot suggested that CIL-102 competitively inhibited the binding of podophyllotoxin, a colchicine-binding site agent, to tubulin. Computational modeling indicated that CIL-102 binds exclusively at the β-subunit of tubulin and that CIL-102 and colchicine partially share their binding sites on tubulin. It bound to tubulin reversibly and the binding was estimated to be ~1000 times faster than that of colchicine. CIL-102 potently inhibited the proliferation of MCF-7 cells, induced monopolar spindle formation and multi-nucleation. At half-maximal inhibitory concentration, the spindle microtubules were visibly depolymerized and disorganized. CIL-102 reduced the inter-polar distances of bipolar mitotic cells indicating that it impaired microtubule-kinetochore attachments. CIL-102-treatment induced apoptosis in MCF-7 cells in association with increased nuclear accumulation of p53 and p21 suggesting that apoptosis is triggered through a p53-p21 dependent pathway. The results indicated that CIL-102 exerted anti-proliferative activity by disrupting microtubule functions through tubulin binding and provided important insights into the differential mode of tubulin binding by CIL-102 and colchicine.
Collapse
Affiliation(s)
- K K Gireesh
- School of Biology, Indian Institute of Science Education and Research, Thiruvananthapuram, CET Campus, Thiruvananthapuram 695016, Kerala, India
| | | | | | | | | |
Collapse
|