55 results on '"Patro BS"'
Search Results
2. Drug release potential of polyacrylamide grafted-Assam Bora rice polysaccharide graft copolymer (ABRS-g-PAM) as effective controlled release polymer.
- Author
-
Singh N, Das IJ, Satpathi S, Das D, Patro BS, Jena K, Chakroborty S, Dagar P, and Bal T
- Subjects
- Polysaccharides chemistry, Drug Carriers chemistry, Polymers chemistry, Oryza chemistry, Acrylic Resins chemistry, Drug Liberation, Delayed-Action Preparations chemistry
- Abstract
The research focuses on the characterization and evaluation of drug delivery efficiency of a microwave-assisted, free-radical synthesized polyacrylamide-grafted Assam Bora rice starch (ABRS) graft copolymer (ABRS-g-PAM). Percentage grafting efficiency (% GE) and intrinsic viscosity were chosen as the optimization parameters. The optimized ABRS-g-PAM Grade Formulation 4 (GF4) was found to be the best grade. GF4 was characterized through XRD, FTIR, TGA, DSC, and
1 ³C NMR, confirming efficient polyacrylamide grafting onto ABRS, besides swelling studies. SEM and FESEM showed a rough GF4 surface. Further analysis using Atomic Force Microscopy (AFM) detailed the irregular, rough internal architecture. The maximum swelling of GF4 occurred at pH 7 and 0.005 (M) NaCl, demonstrating second-order swelling kinetics. The soil biodegradability study of GF4 confirmed its biocompatibility. In vitro drug release studies showed that GF4 achieved 99 % release of Rosuvastatin over 26 h, thereby supporting the controlled release behaviour of the optimized polymer., Competing Interests: Declaration of competing interest The authors declare that they have no known competing financial interests or personal relationships that could have appeared to influence the work reported in this paper., (Copyright © 2024 Elsevier Ltd. All rights reserved.)- Published
- 2025
- Full Text
- View/download PDF
3. Experimental and theoretical validation for transmutation of palladium at electrochemical interfaces.
- Author
-
Gadly T, Phapale S, Gamre S, Jain P, Kumar H, Poudel SK, Desai S, Pathak AK, Srivastava A, Kumar A, Pal R, Chandwadkar P, Acharya C, Bakshi AK, Varma S, Patro BS, and Goswami D
- Abstract
Electrolysis of D
2 O may be used as a portable neutron source with numerous applications without the complexity of huge reactor operations. Herein, we report reproducible fast neutron generation by electrolysis of D2 O using palladium cathode and platinum anode, which was detected with diamond detector, gas filled3 He detectors after thermalisation with high density polythene, as well as novel epoxy resin and CR-39 detectors. Notably, a highly reproducible neutron generation at electrochemical surfaces of palladium electrode was observed and signature transmutation via Pd (d, n) Ag was corroborated. This was further explained using a theoretical model based on second order quantum perturbation theory., (© 2024. The Author(s).)- Published
- 2024
- Full Text
- View/download PDF
4. A BODIPY-Naphtholimine-BF 2 Dyad for Precision Photodynamic Therapy, Targeting, and Dual Imaging of Endoplasmic Reticulum and Lipid Droplets in Cancer.
- Author
-
Chauhan N, Koli M, Ghosh R, Majumdar AG, Ghosh A, Ghanty TK, Mula S, and Patro BS
- Abstract
Currently, effective therapeutic modalities for pancreatic ductal adenocarcinoma (PDAC) are quite limited, leading to gloomy prognosis and ∼6-months median patient survival. Recent advances showed the promise of photodynamic therapy (PDT) for PDAC patients. Next generation photosensitizers (PS) are based on "organelle-targeted-PDT" and provide new paradigm in the field of precision medicines to address the current challenge for treating PDAC. In this investigation, we have constructed a novel PS, named as N b B , for precise and simultaneous targeting of endoplasmic reticulum (ER) and lipid droplets (LDs) in PDAC, based on the fact that malignant PDAC cells are heavily relying on ER for hormone synthesis. Our live cell imaging and fluorescence recovery after photobleaching (FRAP) experiments revealed that N b B is quickly targeted to ER and subsequently to LDs and shows simultaneous dual fluorescence color due to polar and nonpolar milieu of ER and LDs. Interestingly, the same molecule generates triplet state and singlet oxygen efficiently and causes robust ER stress and cellular lipid peroxidation, leading to apoptosis in two different PDAC cells in the presence of light. Together, we present, for the first time, a potential next generation precision medicine for ER-LD organelle specific imaging and PDT of pancreatic cancer., Competing Interests: The authors declare no competing financial interest., (© 2024 The Authors. Published by American Chemical Society.)
- Published
- 2024
- Full Text
- View/download PDF
5. CLU (clusterin) and PPARGC1A/PGC1α coordinately control mitophagy and mitochondrial biogenesis for oral cancer cell survival.
- Author
-
Praharaj PP, Patra S, Singh A, Panigrahi DP, Lee HY, Kabir MF, Hossain MK, Patra SK, Patro BS, Patil S, Klionsky DJ, Chae HJ, and Bhutia SK
- Subjects
- Humans, Animals, Cell Line, Tumor, Cisplatin pharmacology, Organelle Biogenesis, Mice, Apoptosis drug effects, Mice, Nude, Reactive Oxygen Species metabolism, Autophagy physiology, Autophagy drug effects, Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha metabolism, Clusterin metabolism, Clusterin genetics, Mitophagy drug effects, Mitophagy physiology, Mitochondria metabolism, Mitochondria drug effects, Mouth Neoplasms pathology, Mouth Neoplasms metabolism, Mouth Neoplasms genetics, Cell Survival drug effects
- Abstract
Mitophagy involves the selective elimination of defective mitochondria during chemotherapeutic stress to maintain mitochondrial homeostasis and sustain cancer growth. Here, we showed that CLU (clusterin) is localized to mitochondria to induce mitophagy controlling mitochondrial damage in oral cancer cells. Moreover, overexpression and knockdown of CLU establish its mitophagy-specific role, where CLU acts as an adaptor protein that coordinately interacts with BAX and LC3 recruiting autophagic machinery around damaged mitochondria in response to cisplatin treatment. Interestingly, CLU triggers class III phosphatidylinositol 3-kinase (PtdIns3K) activity around damaged mitochondria, and inhibition of mitophagic flux causes the accumulation of excessive mitophagosomes resulting in reactive oxygen species (ROS)-dependent apoptosis during cisplatin treatment in oral cancer cells. In parallel, we determined that PPARGC1A/PGC1α (PPARG coactivator 1 alpha) activates mitochondrial biogenesis during CLU-induced mitophagy to maintain the mitochondrial pool. Intriguingly, PPARGC1A inhibition through small interfering RNA (si PPARGC1A ) and pharmacological inhibitor (SR-18292) treatment counteracts CLU-dependent cytoprotection leading to mitophagy-associated cell death. Furthermore, co-treatment of SR-18292 with cisplatin synergistically suppresses tumor growth in oral cancer xenograft models. In conclusion, CLU and PPARGC1A are essential for sustained cancer cell growth by activating mitophagy and mitochondrial biogenesis, respectively, and their inhibition could provide better therapeutic benefits against oral cancer.
- Published
- 2024
- Full Text
- View/download PDF
6. Recent advancement of autophagy in polyploid giant cancer cells and its interconnection with senescence and stemness for therapeutic opportunities.
- Author
-
Patra S, Naik PP, Mahapatra KK, Alotaibi MR, Patil S, Patro BS, Sethi G, Efferth T, and Bhutia SK
- Subjects
- Humans, Animals, Epithelial-Mesenchymal Transition, Autophagy, Polyploidy, Cellular Senescence drug effects, Neoplasms pathology, Neoplasms genetics, Neoplasms drug therapy, Neoplasms metabolism, Neoplastic Stem Cells pathology, Neoplastic Stem Cells metabolism, Neoplastic Stem Cells drug effects
- Abstract
Recurrent chemotherapy-induced senescence and resistance are attributed to the polyploidization of cancer cells that involve genomic instability and poor prognosis due to their unique form of cellular plasticity. Autophagy, a pre-dominant cell survival mechanism, is crucial during carcinogenesis and chemotherapeutic stress, favouring polyploidization. The selective autophagic degradation of essential proteins associated with cell cycle progression checkpoints deregulate mitosis fidelity and genomic integrity, imparting polyploidization of cancer cells. In connection with cytokinesis failure and endoreduplication, autophagy promotes the formation, maintenance, and generation of the progeny of polyploid giant cancer cells. The polyploid cancer cells embark on autophagy-guarded elevation in the expression of stem cell markers, along with triggered epithelial and mesenchymal transition and senescence. The senescent polyploid escapers represent a high autophagic index than the polyploid progeny, suggesting regaining autophagy induction and subsequent autophagic degradation, which is essential for escaping from senescence/polyploidy, leading to a higher proliferative phenotypic progeny. This review documents the various causes of polyploidy and its consequences in cancer with relevance to autophagy modulation and its targeting for therapeutic intervention as a novel therapeutic strategy for personalized and precision medicine., Competing Interests: Declaration of competing interest The authors declare that they have no known competing financial interests or personal relationships that could have appeared to influence the work reported in this paper., (Copyright © 2024 Elsevier B.V. All rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
7. Emerging roles of lysosome homeostasis (repair, lysophagy and biogenesis) in cancer progression and therapy.
- Author
-
Chauhan N and Patro BS
- Subjects
- Humans, Lysosomes metabolism, Proteins metabolism, Ubiquitination, Homeostasis, Autophagy physiology, Macroautophagy, Neoplasms drug therapy, Neoplasms metabolism
- Abstract
In the era of personalized therapy, precise targeting of subcellular organelles holds great promise for cancer modality. Taking into consideration that lysosome represents the intersection site in numerous endosomal trafficking pathways and their modulation in cancer growth, progression, and resistance against cancer therapies, the lysosome is proposed as an attractive therapeutic target for cancer treatment. Based on the recent advances, the current review provides a comprehensive understanding of molecular mechanisms of lysosome homeostasis under 3R responses: Repair, Removal (lysophagy) and Regeneration of lysosomes. These arms of 3R responses have distinct role in lysosome homeostasis although their interdependency along with switching between the pathways still remain elusive. Recent advances underpinning the crucial role of (1) ESCRT complex dependent/independent repair of lysosome, (2) various Galectins-based sensing and ubiquitination in lysophagy and (3) TFEB/TFE proteins in lysosome regeneration/biogenesis of lysosome are outlined. Later, we also emphasised how these recent advancements may aid in development of phytochemicals and pharmacological agents for targeting lysosomes for efficient cancer therapy. Some of these lysosome targeting agents, which are now at various stages of clinical trials and patents, are also highlighted in this review., Competing Interests: Declaration of competing interest The authors declare no competing interest for this paper., (Copyright © 2023 Elsevier B.V. All rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
8. In house synthesized novel distyryl-BODIPY dye and polymer assembly as deep-red emitting probe for protamine detection.
- Author
-
Gorai S, Mula S, Jonnalgadda PN, Patro BS, and Chakraborty G
- Subjects
- Humans, Boron Compounds, Limit of Detection, Fluorescent Dyes, Protamines
- Abstract
In this contribution, we designed and synthesized a deep-red emitting distyryl-BODIPY dye (dye 3) which is non-fluorescent in aqueous solution due to the formation of non-emissive aggregates. However, in presence of an amphiphilic polymer (polystyrene sulfonate, PSS), the aggregated dye molecules de-aggregate and form dye 3-PSS complex, which significantly modulates the optical features of the bound dye. Interestingly, the dye 3-PSS complex shows turn-on fluorescence response in deep-red region in presence of protamine (Pr) due to the formation of dye 3-PSS-Pr ternary complex. Such enhancement follows a linear trend in the dynamic range of 0-8.75 μM of Pr which has been utilized to determine Pr with limit of detection (LOD) of 15.04(±0.5) nM in phosphate buffer. Furthermore, excellent selectivity of the dye 3-PSS system towards Pr allows us to determine Pr even in complex biological matrix like 1% human serum. Thus, dye 3-PSS system can be applied as a very effective tool for the detection and quantification of Pr in deep-red region, overcoming several limitations encountered with the probes in the shorter wavelength region. This is the first report on BODIPY dye based supramolecular assembly for sensing and quantification of protamine., Competing Interests: Declaration of competing interest There is no competing conflict of interest to be declared., (Copyright © 2023 Elsevier B.V. All rights reserved.)
- Published
- 2023
- Full Text
- View/download PDF
9. SIRT1 inhibits mitochondrial hyperfusion associated mito-bulb formation to sensitize oral cancer cells for apoptosis in a mtROS-dependent signalling pathway.
- Author
-
Patra S, Singh A, Praharaj PP, Mohanta NK, Jena M, Patro BS, Abusharha A, Patil S, and Bhutia SK
- Subjects
- Humans, Superoxides, Sirtuin 1 genetics, Sirtuin 1 metabolism, Apoptosis, Cisplatin pharmacology, Mitomycin, Mitochondrial Dynamics, Mouth Neoplasms drug therapy, Mouth Neoplasms genetics
- Abstract
SIRT1 (NAD-dependent protein deacetylase sirtuin-1), a class III histone deacetylase acting as a tumor suppressor gene, is downregulated in oral cancer cells. Non-apoptotic doses of cisplatin (CDDP) downregulate SIRT1 expression advocating the mechanism of drug resistance. SIRT1 downregulation orchestrates inhibition of DNM1L-mediated mitochondrial fission, subsequently leading to the formation of hyperfused mitochondrial networks. The hyperfused mitochondrial networks preserve the release of cytochrome C (CYCS) by stabilizing the mitochondrial inner membrane cristae (formation of mitochondrial nucleoid clustering mimicking mito-bulb like structures) and reducing the generation of mitochondrial superoxide to inhibit apoptosis. Overexpression of SIRT1 reverses the mitochondrial hyperfusion by initiating DNM1L-regulated mitochondrial fission. In the overexpressed cells, inhibition of mitochondrial hyperfusion and nucleoid clustering (mito-bulbs) facilitates the cytoplasmic release of CYCS along with an enhanced generation of mitochondrial superoxide for the subsequent induction of apoptosis. Further, low-dose priming with gallic acid (GA), a bio-active SIRT1 activator, nullifies CDDP-mediated apoptosis inhibition by suppressing mitochondrial hyperfusion. In this setting, SIRT1 knockdown hinders apoptosis activation in GA-primed oral cancer cells. Similarly, SIRT1 overexpression in the CDDP resistance oral cancer-derived polyploid giant cancer cells (PGCCs) re-sensitizes the cells to apoptosis. Interestingly, synergistically treated with CDDP, GA induces apoptosis in the PGCCs by inhibiting mitochondrial hyperfusion., (© 2023. The Author(s).)
- Published
- 2023
- Full Text
- View/download PDF
10. Design and Synthesis of BODIPY-Hetero[5]helicenes as Heavy-Atom-Free Triplet Photosensitizers for Photodynamic Therapy of Cancer.
- Author
-
Koli M, Gupta S, Chakraborty S, Ghosh A, Ghosh R, Wadawale AP, Ghanty TK, Patro BS, and Mula S
- Abstract
Designing heavy-atom-free triplet photosensitizers (PSs) is a challenge for the efficient photodynamic therapy (PDT) of cancer. Helicenes are twisted polycyclic aromatic hydrocarbons (PAHs) with an efficient intersystem crossing (ISC) that is proportional to their twisting angle. But their difficult syntheses and weak absorption profile in the visible spectral region restrict their use as heavy-atom-free triplet PSs for PDT. On the other hand, boron-containing PAHs, BODIPYs are highly recognized for their outstanding optical properties. However, planar BODIPY dyes has low ISC and thus they are not very effective as PDT agents. We have designed and synthesized fused compounds containing both BODIPY and hetero[5]helicene structures to develop red-shifted chromophores with efficient ISC. One of the pyrrole units of the BODIPY core was also replaced by a thiazole unit to further enhance the triplet conversion. All the fused compounds have helical structure, and their twisting angles are also increased by substitutions at the boron centre. The helical structures of the BODIPY-hetero[5]helicenes were confirmed by X-ray crystallography and DFT structure optimization. The designed BODIPY-hetero[5]helicenes showed superior optical properties and high ISC with respect to [5]helicene. Interestingly their ISC efficiencies increase proportionally with their twisting angles. This is the first report on the relationship between the twisting angle and the ISC efficiency in twisted BODIPY-based compounds. Theoretical calculations showed that energy gap of the S1 and T1 states decreases in BODIPY-hetero[5]helicene as compared to planar BODIPY. This enhances the ISC rate in BODIPY-hetero[5]helicene, which is responsible for their high generation of singlet oxygen. Finally, their potential applications as PDT agents were investigated, and one BODIPY-hetero[5]helicene showed efficient cancer cell killing upon photo-exposure. This new design strategy will be very useful for the future development of heavy-atom-free PDT agents., (© 2023 Wiley-VCH GmbH.)
- Published
- 2023
- Full Text
- View/download PDF
11. Design, synthesis and development of a dual inhibitor of Topoisomerase 1 and poly (ADP-ribose) polymerase 1 for efficient killing of cancer cells.
- Author
-
Guha Majumdar A, Shree S, Das A, Kumar BK, Dey P, Subramanian M, and Patro BS
- Subjects
- Humans, Poly (ADP-Ribose) Polymerase-1, Topoisomerase I Inhibitors pharmacology, DNA, Ribose, Neoplasms drug therapy
- Abstract
Combinatorial inhibition of Topoisomerase 1 (TOP1) and Poly (ADP-ribose) polymerase 1 (PARP1) is an attractive therapeutic strategy which is under active investigation to address chemoresistance to TOP1 inhibitors. However, this combinatorial regimen suffers from severe dose limiting toxicities. Dual inhibitors often offer significant advantages over combinatorial therapies involving individual agents by minimizing toxicity and providing conducive pharmacokinetic profiles. In this study, we have designed, synthesized and evaluated a library of 11 candidate conjugated dual inhibitors for PARP1 and TOP1, named as DiPT-1 to DiPT-11. Our extensive screening showed that one of the hits i.e.DiPT-4 has promising cytotoxicity profile against multiple cancers with limited toxicities towards normal cells. DiPT-4 induces extensive DNA double stand breaks (DSBs), cell cycle arrest and apoptosis in cancer cells. Mechanistically, DiPT-4 has the propensity to bind catalytic pockets of TOP1 and PARP1, leading to significant inhibition of both TOP1 and PARP1 at in vitro and cellular level. Interestingly, DiPT-4 causes extensive stabilization of TOP1-DNA covalent complex (TOP1cc), a key lethal intermediate associated with induction of DSBs and cell death. Moreover, DiPT-4 inhibited poly (ADP-ribosylation) i.e. PARylation of TOP1cc, leading to long lived TOP1cc with a slower kinetics of degradation. This is one of the important molecular processes which helps in overcoming resistance in cancer in response to TOP1 inhibitors. Together, our investigation showed DiPT-4 as a promising dual inhibitor of TOP1 and PARP1, which may have the potential to offer significant advantages over combinatorial therapy in clinical settings., Competing Interests: Declaration of competing interest The authors declare the following financial interests/personal relationships which may be considered as potential competing interests: There is no conflict of interest., (Copyright © 2023 Elsevier Masson SAS. All rights reserved.)
- Published
- 2023
- Full Text
- View/download PDF
12. Functional inhibition of RECQL5 helicase elicits non-homologous end joining response and sensitivity of breast cancers to PARP inhibitor.
- Author
-
Philip KT, Dutta K, Chakraborty S, and Patro BS
- Subjects
- Humans, Female, DNA End-Joining Repair, Breast pathology, DNA Replication, Cell Line, Tumor, Homologous Recombination, RecQ Helicases genetics, Poly(ADP-ribose) Polymerase Inhibitors pharmacology, Poly(ADP-ribose) Polymerase Inhibitors therapeutic use, Breast Neoplasms drug therapy, Breast Neoplasms genetics, Breast Neoplasms pathology
- Abstract
Poly (ADPRibose) Polymerase inhibitor (PARPi) are clinically approved for the treatment of BRCA-mutated hereditary breast and ovarian cancers with homologous recombination (HR) deficiency, based on synthetic lethality concept. However, ∼90% of breast cancers are BRCA-wild type; they repair PARPi mediated damage through HR, leading to intrinsic de novo resistance. Hence, there is an unmet need of exploring novel targets in HR-proficient aggressive breast cancers for PARPi treatment. RECQL5 physically interacts and disrupts RAD51 from pre-synaptic filaments, aiding HR resolution, replication fork protection and preventing illegitimate recombination. In the current investigation, we show that targeted inhibition of HR by stabilization of RAD51-RECQL5 complex by a pharmacological inhibitor of RECQL5 (4a; 1,3,4-oxadiazole derivative) in the presence of PARPi [talazoparib (BMN673)] leads to abolition of functional HR with uncontrolled activation of NHEJ repair. This was assessed by GFP based NHEJ reporter assay, KU80 recruitment and in vitro NHEJ based plasmid ligation assay. Concomitant treatment with talazoparib and 4a generates copious amounts of replication stress, prolonged cell cycle arrest, extensive double strand breaks (DSBs) and mitotic catastrophe, leading to sensitization of HR-proficient breast cancers. Suppression of NHEJ activity abolishes 4a-mediated sensitization of breast cancers to PARPi treatment. Imperatively, 4a was ineffective against normal mammary epithelial cells, which expresses low RECQL5 vis-à-vis breast cancer cells. Moreover, functional inhibition of RECQL5 suppresses metastatic potential of breast cancer cells in response to PARPi. Together, we identified RECQL5 as a novel pharmacological target for expanding PARPi based treatment horizon for HR-proficient cancers., Competing Interests: Declaration of Competing Interest The authors declare no competing interests., (Copyright © 2023 Elsevier Ltd. All rights reserved.)
- Published
- 2023
- Full Text
- View/download PDF
13. The inner mitochondrial membrane fission protein MTP18 serves as a mitophagy receptor to prevent apoptosis in oral cancer.
- Author
-
Panigrahi DP, Praharaj PP, Behera BP, Patra S, Patil S, Patro BS, and Bhutia SK
- Subjects
- Humans, Apoptosis genetics, Carbonyl Cyanide m-Chlorophenyl Hydrazone metabolism, Carbonyl Cyanide m-Chlorophenyl Hydrazone pharmacology, Membrane Proteins genetics, Membrane Proteins metabolism, Mitochondrial Dynamics, Mitophagy genetics, Protein Kinases genetics, Protein Kinases metabolism, Ubiquitin-Protein Ligases genetics, Ubiquitin-Protein Ligases metabolism, Mitochondrial Membranes metabolism, Mouth Neoplasms genetics, Mouth Neoplasms metabolism
- Abstract
MTP18 (also known as MTFP1), an inner mitochondrial membrane protein, plays a vital role in maintaining mitochondrial morphology by regulating mitochondrial fission. Here, we found that MTP18 functions as a mitophagy receptor that targets dysfunctional mitochondria into autophagosomes for elimination. Interestingly, MTP18 interacts with members of the LC3 (also known as MAP1LC3) family through its LC3-interacting region (LIR) to induce mitochondrial autophagy. Mutation in the LIR motif (mLIR) inhibited that interaction, thus suppressing mitophagy. Moreover, Parkin or PINK1 deficiency abrogated mitophagy in MTP18-overexpressing human oral cancer-derived FaDu cells. Upon exposure to the mitochondrial oxidative phosphorylation uncoupler CCCP, MTP18[mLIR]-FaDu cells showed decreased TOM20 levels without affecting COX IV levels. Conversely, loss of Parkin or PINK1 resulted in inhibition of TOM20 and COX IV degradation in MTP18[mLIR]-FaDu cells exposed to CCCP, establishing Parkin-mediated proteasomal degradation of outer mitochondrial membrane as essential for effective mitophagy. We also found that MTP18 provides a survival advantage to oral cancer cells exposed to cellular stress and that inhibition of MTP18-dependent mitophagy induced cell death in oral cancer cells. These findings demonstrate that MTP18 is a novel mitophagy receptor and that MTP18-dependent mitophagy has pathophysiologic implications for oral cancer progression, indicating inhibition of MTP18-mitophagy could thus be a promising cancer therapy strategy., Competing Interests: Competing interests The authors declare no competing or financial interests., (© 2023. Published by The Company of Biologists Ltd.)
- Published
- 2023
- Full Text
- View/download PDF
14. Editorial: TGF-β signalling pathways and their enigmatic role as a friend and foe in human health and diseases.
- Author
-
Betapudi V, Patro BS, Saika S, and Rahaman SO
- Abstract
Competing Interests: The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.
- Published
- 2023
- Full Text
- View/download PDF
15. Fluorogenic gemcitabine based light up sensor for serum albumin detection in complex biological matrices.
- Author
-
Gadly T, Patro BS, and Chakraborty G
- Subjects
- Humans, Gemcitabine, Serum Albumin, Deoxycytidine
- Abstract
Herein we report fluorogenic derivative of gemcitabine (GEM-DNS), synthesized from gemcitabine hydrochloride and dansyl chloride in a single step. Owing to its large stoke shift of ∼200 nm and intriguing photophysical properties, the said dye has been utilized to estimate albumin concentration in complex bio-media such as human urine and blood serum. High sensitivity and selectivity towards albumin make the aforementioned dye a powerful diagnostic tool to detect ailments such as liver cirrhosis, diabetes, hypertension etc., Competing Interests: Declaration of Competing Interest The authors declare that they have no known competing financial interests or personal relationships that could have appeared to influence the work reported in this paper., (Copyright © 2022 Elsevier B.V. All rights reserved.)
- Published
- 2022
- Full Text
- View/download PDF
16. Dysregulation of mitophagy and mitochondrial homeostasis in cancer stem cells: Novel mechanism for anti-cancer stem cell-targeted cancer therapy.
- Author
-
Praharaj PP, Patro BS, and Bhutia SK
- Subjects
- Homeostasis, Humans, Mitochondria metabolism, Mitochondrial Dynamics, Neoplastic Stem Cells, Tumor Microenvironment, Mitophagy, Neoplasms metabolism
- Abstract
Despite the potential of cancer medicine, cancer stem cells (CSCs) associated with chemoresistance and disease recurrence are the significant challenges currently opposing the efficacy of available cancer treatment options. Mitochondrial dynamics involving the fission-fusion cycle and mitophagy are the major contributing factors to better adaptation, enabling CSCs to survive and grow better under tumour micro-environment-associated stress. Moreover, mitophagy is balanced with mitochondrial biogenesis to maintain mitochondrial homeostasis in CSCs, which are necessary for the growth and maintenance of CSCs and regulate metabolic switching from glycolysis to oxidative phosphorylation. In this review, we discuss different aspects of mitochondrial dynamics, mitophagy, and mitochondrial homeostasis and their effects on modulating CSCs behaviour during cancer development. Moreover, the efficacy of pharmacological targeting of these cellular processes using anti-CSC drugs in combination with currently available chemotherapeutic drugs improves the patient's survival of aggressive cancer types., (© 2021 The British Pharmacological Society.)
- Published
- 2022
- Full Text
- View/download PDF
17. MTP18 inhibition triggers mitochondrial hyperfusion to induce apoptosis through ROS-mediated lysosomal membrane permeabilization-dependent pathway in oral cancer.
- Author
-
Panigrahi DP, Patra S, Behera BP, Behera PK, Patil S, Patro BS, Rout L, Sarangi I, and Bhutia SK
- Subjects
- Apoptosis physiology, Humans, Lysosomes metabolism, Mitochondria metabolism, Mitochondrial Proteins metabolism, Reactive Oxygen Species metabolism, Superoxides metabolism, Mitochondrial Dynamics, Mouth Neoplasms drug therapy, Mouth Neoplasms genetics, Mouth Neoplasms metabolism
- Abstract
Although stress-induced mitochondrial hyperfusion (SIMH) exerts a protective role in aiding cell survival, in the absence of mitochondrial fission, SIMH drives oxidative stress-related induction of apoptosis. In this study, our data showed that MTP18, a mitochondrial fission-promoting protein expression, was increased in oral cancer. We have screened and identified S28, a novel inhibitor of MTP18, which was found to induce SIMH and subsequently trigger apoptosis. Interestingly, it inhibited MTP18-mediated mitochondrial fission, as shown by a decrease in p-Drp1 along with increased Mfn1 expression in oral cancer cells. Moreover, S28 induced autophagy but not mitophagy due to the trouble in engulfment of hypoperfused mitochondria. Interestingly, S28-mediated SIMH resulted in the loss of mitochondrial membrane potential, leading to the consequent generation of mitochondrial superoxide to induce intrinsic apoptosis. Mechanistically, S28-induced mitochondrial superoxide caused lysosomal membrane permeabilization (LMP), resulting in decreased lysosomal pH, which impaired autophagosome-lysosome fusion. In this setting, it showed that overexpression of MTP18 resulted in mitochondrial fission leading to mitophagy and inhibition of superoxide-mediated LMP and apoptosis. Further, S28, in combination with FDA-approved anticancer drugs, exhibited higher apoptotic activity and decreased cell viability, suggesting the MTP18 inhibition combined with the anticancer drug could have greater efficacy against cancer., Competing Interests: Declaration of competing interest No competing interests declared., (Copyright © 2022 Elsevier Inc. All rights reserved.)
- Published
- 2022
- Full Text
- View/download PDF
18. Non-enzymatic function of WRN RECQL helicase regulates removal of topoisomerase-I-DNA covalent complexes and triggers NF-κB signaling in cancer.
- Author
-
Gupta P, Majumdar AG, and Patro BS
- Subjects
- Animals, DNA metabolism, DNA, Single-Stranded, Exodeoxyribonucleases metabolism, Female, Genetic Predisposition to Disease, Humans, Mice, NF-kappa B metabolism, Proteasome Endopeptidase Complex metabolism, RecQ Helicases genetics, RecQ Helicases metabolism, Signal Transduction, Werner Syndrome Helicase genetics, Breast Neoplasms, Werner Syndrome genetics
- Abstract
Mutation in Werner (WRN) RECQL helicase is associated with premature aging syndrome (Werner syndrome, WS) and predisposition to multiple cancers. In patients with solid cancers, deficiency of the WRN RECQL helicase is paradoxically associated with enhanced overall survival in response to treatment with TOP1 inhibitors, which stabilize pathological TOP1-DNA-covalent-complexes (TOP1cc) on the genome. However, the underlying mechanism of WRN in development of chemoresistance to TOP1 inhibitors is not yet explored. Our whole-genome transcriptomic analysis for ~25,000 genes showed robust activation of NF-κB-dependent prosurvival genes in response to TOP1cc. CRISPR-Cas9 knockout, shRNA silencing, and under-expression of WRN confer high-sensitivity of multiple cancers to TOP1 inhibitor. We demonstrated that WRN orchestrates TOP1cc repair through proteasome-dependent and proteasome-independent process, unleashing robust ssDNA generation. This in turn ensues signal transduction for CHK1 mediated NF-κB-activation through IκBα-degradation and nuclear localization of p65 protein. Intriguingly, our site-directed mutagenesis and rescue experiments revealed that neither RECQL-helicase nor DNA-exonuclease enzyme activity of WRN (WRN
E84A , WRNK577M , and WRNE84A-K577M ) were required for TOP1cc removal, ssDNA generation and signaling for NF-κB activation. In correlation with patient data and above results, the TOP1 inhibitor-based targeted therapy showed that WRN-deficient melanoma tumors were highly sensitive to TOP1 inhibition in preclinical in vivo mouse model. Collectively, our findings identify hitherto unknown non-enzymatic role of WRN RECQL helicase in pathological mechanisms underlying TOP1cc processing and subsequent NF-κB-activation, offering a potential targeted therapy for WRN-deficient cancer patients., (© 2022 The Authors. Aging Cell published by the Anatomical Society and John Wiley & Sons Ltd.)- Published
- 2022
- Full Text
- View/download PDF
19. Synthesis of Dihydrobenzofuro[3,2-b]chromenes as Potential 3CLpro Inhibitors of SARS-CoV-2: A Molecular Docking and Molecular Dynamics Study.
- Author
-
Gorai S, Junghare V, Kundu K, Gharui S, Kumar M, Patro BS, Nayak SK, Hazra S, and Mula S
- Subjects
- Antiviral Agents chemistry, Antiviral Agents pharmacology, Benzopyrans pharmacology, Benzopyrans therapeutic use, Flavonoids chemistry, Flavonoids pharmacology, Humans, Molecular Docking Simulation, Molecular Dynamics Simulation, Protease Inhibitors chemistry, COVID-19, SARS-CoV-2
- Abstract
The recent emergence of pandemic of coronavirus (COVID-19) caused by SARS-CoV-2 has raised significant global health concerns. More importantly, there is no specific therapeutics currently available to combat against this deadly infection. The enzyme 3-chymotrypsin-like cysteine protease (3CLpro) is known to be essential for viral life cycle as it controls the coronavirus replication. 3CLpro could be a potential drug target as established before in the case of severe acute respiratory syndrome coronavirus (SARS-CoV) and Middle East respiratory syndrome coronavirus (MERS-CoV). In the current study, we wanted to explore the potential of fused flavonoids as 3CLpro inhibitors. Fused flavonoids (5a,10a-dihydro-11H-benzofuro[3,2-b]chromene) are unexplored for their potential bioactivities due to their low natural occurrences. Their synthetic congeners are also rare due to unavailability of general synthetic methodology. Here we designed a simple strategy to synthesize 5a,10a-dihydro-11H-benzofuro[3,2-b]chromene skeleton and it's four novel derivatives. Our structural bioinformatics study clearly shows excellent potential of the synthesized compounds in comparison to experimentally validated inhibitor N3. Moreover, in-silico ADMET study displays excellent druggability and extremely low level of toxicity of the synthesized molecules. Further, for better understanding, the molecular dynamic approach was implemented to study the change in dynamicity after the compounds bind to the protein. A detailed investigation through clustering analysis and distance calculation gave us sound comprehensive data about their molecular interaction. In summary, we anticipate that the currently synthesized molecules could not only be a potential set of inhibitors against 3CLpro but also the insights acquired from the current study would be instrumental in further developing novel natural flavonoid based anti-COVID therapeutic spectrums., (© 2022 Wiley-VCH GmbH.)
- Published
- 2022
- Full Text
- View/download PDF
20. Carbon nano-dot for cancer studies as dual nano-sensor for imaging intracellular temperature or pH variation.
- Author
-
Gadly T, Chakraborty G, Tyagi M, Patro BS, Dutta B, Potnis A, Chandwadkar P, Acharya C, Suman SK, Mukherjee A, Neogy S, Wadawale A, Sahoo S, Chauhan N, and Ghosh SK
- Subjects
- Female, Humans, Hydrogen-Ion Concentration, MCF-7 Cells, Breast Neoplasms pathology, Carbon chemistry, Fluorescent Dyes chemistry, Quantum Dots, Temperature
- Abstract
Cellular temperature and pH govern many cellular physiologies, especially of cancer cells. Besides, attaining higher cellular temperature plays key role in therapeutic efficacy of hyperthermia treatment of cancer. This requires bio-compatible, non-toxic and sensitive probe with dual sensing ability to detect temperature and pH variations. In this regard, fluorescence based nano-sensors for cancer studies play an important role. Therefore, a facile green synthesis of orange carbon nano-dots (CND) with high quantum yield of 90% was achieved and its application as dual nano-sensor for imaging intracellular temperature and pH was explored. CND was synthesized from readily available, bio-compatible citric acid and rhodamine 6G hydrazide using solvent-free and simple heating technique requiring purification by dialysis. Although the particle size of 19 nm (which is quite large for CND) was observed yet CND exhibits no surface defects leading to decrease in photoluminescence (PL). On the contrary, very high fluorescence was observed along with good photo-stability. Temperature and pH dependent fluorescence studies show linearity in fluorescence intensity which was replicated in breast cancer cells. In addition, molecular nature of PL of CND was established using pH dependent fluorescence study. Together, the current investigation showed synthesis of highly fluorescent orange CND, which acts as a sensitive bio-imaging probe: an optical nano-thermal or nano-pH sensor for cancer-related studies., (© 2021. The Author(s).)
- Published
- 2021
- Full Text
- View/download PDF
21. Pharmacological targeting of differential DNA repair, radio-sensitizes WRN-deficient cancer cells in vitro and in vivo.
- Author
-
Gupta P, Saha B, Chattopadhyay S, and Patro BS
- Subjects
- Animals, Cell Line, Tumor, Checkpoint Kinase 1 antagonists & inhibitors, Checkpoint Kinase 1 metabolism, DNA Repair physiology, Enzyme Inhibitors administration & dosage, Humans, MAP Kinase Signaling System drug effects, MAP Kinase Signaling System physiology, Male, Melanoma, Experimental, Mice, Mice, Inbred C57BL, Xenograft Model Antitumor Assays methods, DNA Repair drug effects, Drug Delivery Systems methods, Radiation-Sensitizing Agents administration & dosage, Werner Syndrome Helicase antagonists & inhibitors, Werner Syndrome Helicase deficiency
- Abstract
Werner (WRN) expression is epigenetically downregulated in various tumors. It is imperative to understand differential repair process in WRN-proficient and WRN-deficient cancers to find pharmacological targets for radio-sensitization of WRN-deficient cancer. In the current investigation, we showed that pharmacological inhibition of CHK1 mediated homologous recombination repair (HRR), but not non-homologous end joining (NHEJ) repair, can causes hyper-radiosensitization of WRN-deficient cancers. This was confirmed in cancer cell lines of different tissue origin (osteosarcoma, colon adenocarcinoma and melanoma) with WRN silencing and overexpression. We established that WRN-depleted cells are dependent on a critical but compromised CHK1-mediated HRR-pathway for repairing ionizing radiation (IR) induced DSBs for their survival. Mechanistically, we unraveled a new finding that the MRE11, CTIP and WRN proteins are largely responsible for resections of late and persistent DSBs. In response to IR-treatment, MRE11 and CTIP-positively and WRN-negatively regulate p38-MAPK reactivation in a CHK1-dependent manner. A degradation resistant WRN protein, mutated at serine 1141, abrogates p38-MAPK activation. We also showed that CHK1-p38-MAPK axis plays important role in RAD51 mediated HRR in WRN-silenced cells. Like CHK1 inhibition, pharmacological-inhibition of p38-MAPK also hyper-radiosensitizes WRN-depleted cells by targeting HR-pathway. Combination treatment of CHK1-inhibitor (currently under various clinical trials) and IR exhibited a strong synergy against WRN-deficient melanoma tumor in vivo. Taken together, our findings suggest that pharmacological targeting of CHK1-p38-MAPK mediated HRR is an attractive strategy for enhancing therapeutic response of radiation treatment of cancer., (Copyright © 2021 Elsevier Inc. All rights reserved.)
- Published
- 2021
- Full Text
- View/download PDF
22. Targeting autophagy reverses de novo resistance in homologous recombination repair proficient breast cancers to PARP inhibition.
- Author
-
Pai Bellare G, Saha B, and Patro BS
- Subjects
- Animals, Apoptosis, Breast Neoplasms genetics, Breast Neoplasms pathology, Cell Proliferation, Female, Gene Expression Regulation, Neoplastic, Humans, Mice, Mice, SCID, Tumor Cells, Cultured, Xenograft Model Antitumor Assays, Autophagy, Breast Neoplasms drug therapy, Drug Resistance, Neoplasm, Homologous Recombination, Phthalazines pharmacology, Poly(ADP-ribose) Polymerase Inhibitors pharmacology, Recombinational DNA Repair
- Abstract
Background: Poly(ADP-ribose) polymerase inhibitors (PARPi) target tumours defective in homologous recombination (HR). Most BRCA-wild-type (WT) HR-proficient breast cancers are intrinsically resistant to PARP inhibitors, e.g., talazoparib. We evaluated the role of autophagy in this de novo resistance and determined the underlying mechanism to overcome this., Methods: Autophagosome formation and autophagic flux were assessed by evaluating endogenous LC3-II levels and ectopic expression of EGFP-LC3 and mRFP-EGFP-LC3 in breast cancer cells. Autophagy-defective cells were generated by genetic depletion of BECN1, ATG5, p62/SQSTM1 and LAMP1 by using CRISPR-Cas9 double nickase system. The response of PARPi was evaluated in autophagy-proficient and -defective breast cancer cells and in xenograft SCID-mice model., Results: Pro-survival autophagy was significantly enhanced upon talazoparib treatment in BRCA-WT breast cancer cell lines. Autophagy-deficient cells were hypersensitive to talazoparib. Targeting autophagy synergistically enhanced the therapeutic efficacy of talazoparib in BRCA1-WT breast cancer cells in vitro and in vivo xenograft tumour mouse model. Mechanistically, autophagy inhibition by chloroquine promoted deleterious NHEJ mediated DSB-repair, leading to extensive genomic instability and mitotic catastrophe., Conclusions: Autophagy confers de novo resistance to PARP inhibitor, talazoparib. Autophagy inhibition improves the therapeutic outcome of PARPi treatment in preclinical mice model, bearing HR-proficient breast tumours, warranting its usage in the clinical settings.
- Published
- 2021
- Full Text
- View/download PDF
23. Synthesis of Bioactive Diarylheptanoids from Alpinia officinarum and Their Mechanism of Action for Anticancer Properties in Breast Cancer Cells.
- Author
-
Gamre S, Tyagi M, Chatterjee S, Patro BS, Chattopadhyay S, and Goswami D
- Subjects
- Antineoplastic Agents, Phytogenic chemical synthesis, Apoptosis, Cell Cycle Checkpoints drug effects, Diarylheptanoids chemical synthesis, Eugenol, Humans, Lysosomes, MCF-7 Cells, Membrane Potential, Mitochondrial, Molecular Structure, Oxidative Stress, Reactive Oxygen Species metabolism, Structure-Activity Relationship, Alpinia chemistry, Antineoplastic Agents, Phytogenic pharmacology, Diarylheptanoids pharmacology
- Abstract
An efficient synthesis of the Alpinia officinarum -derived diarylheptanoids, viz., enantiomers of a β-hydroxyketone ( 1 ) and an α,β-unsaturated ketone ( 2 ) was developed starting from commercially available eugenol. Among these, compound 2 showed a superior antiproliferative effect against human breast adenocarcinoma MCF-7 cells. Besides reducing clonogenic cell survival, compound 2 dose-dependently increased the sub G1 cell population and arrested the G2-phase of the cell cycle, as revealed by flow cytometry. Mechanistically, compound 2 acts as an intracellular pro-oxidant by generating copious amounts of reactive oxygen species. Compound 2 also induced both loss of mitochondrial membrane potential (MMP) as well as lysosomal membrane permeabilization (LMP) in the MCF-7 cells. The impaired mitochondrial and lysosomal functions due to reactive oxygen species (ROS)-generation by compound 2 may contribute to its apoptotic property.
- Published
- 2021
- Full Text
- View/download PDF
24. Targeting RECQL5 Functions, by a Small Molecule, Selectively Kills Breast Cancer in Vitr o and in Vivo .
- Author
-
Chakraborty S, Dutta K, Gupta P, Das A, Das A, Ghosh SK, and Patro BS
- Subjects
- Administration, Oral, Animals, Antineoplastic Agents toxicity, Cell Cycle drug effects, Cell Line, Tumor, Cisplatin pharmacology, Computer Simulation, Drug Resistance, Neoplasm, Drug Screening Assays, Antitumor, Female, Gene Expression Regulation, Neoplastic drug effects, Gene Knockout Techniques, Humans, Mice, Mice, Inbred C57BL, Mice, Nude, Models, Molecular, RecQ Helicases genetics, Xenograft Model Antitumor Assays, Antineoplastic Agents chemical synthesis, Antineoplastic Agents pharmacology, Breast Neoplasms drug therapy, RecQ Helicases drug effects
- Abstract
Clinical and preclinical data reveal that RECQL5 protein overexpression in breast cancer was strongly correlated with poor prognosis, survival, and therapeutic resistance. In the current investigation, we report design, synthesis, and specificity of a small molecule, 4a , which can preferentially kill RECQL5-expressing breast cancers but not RECQL5 knockout. Our stringent analysis showed that compound 4a specifically sensitizes RECQL5-expressing cancers, while it did not have any effect on other members of DNA RECQL-helicases. Integrated approaches of organic synthesis, biochemical, in silico molecular simulation, knockouts, functional mutation, and rescue experiments showed that 4a potently inhibits RECQL5-helicase activity and stabilizes RECQL5-RAD51 physical interaction, leading to impaired HRR and preferential killing of RECQL5-expressing breast cancer. Moreover, 4a treatment led to the efficient sensitization of cisplatin-resistant breast cancers but not normal mammary epithelial cells. Pharmacologically, compound 4a was orally effective in reducing the growth of RECQL5-expressing breast tumors (human xenograft) in NUDE-mice with no appreciable toxicity to the vital organs.
- Published
- 2021
- Full Text
- View/download PDF
25. Is DAPI assay of cellular nucleic acid reliable in the presence of protein aggregates?
- Author
-
Mora AK, Khan S, Patro BS, and Nath S
- Subjects
- Cell Line, Tumor, Humans, Protein Aggregates, Spectrometry, Fluorescence, Amyloid beta-Peptides chemistry, DNA, Neoplasm analysis, Fluorescent Dyes chemistry, Indoles chemistry, Peptide Fragments chemistry
- Abstract
DAPI is used extensively to identify and quantify DNA in cellular systems assuming its exclusive staining of nucleic acids. However, the present results show that DAPI has much higher affinity towards protein aggregates than DNA. Thus, the use of DAPI for the nucleic acid assay of cellular systems with protein aggregates may not be very reliable.
- Published
- 2020
- Full Text
- View/download PDF
26. Thiol antioxidants sensitize malabaricone C induced cancer cell death via reprogramming redox sensitive p53 and NF-κB proteins in vitro and in vivo.
- Author
-
Tyagi M, Bauri AK, Chattopadhyay S, and Patro BS
- Subjects
- Animals, Apoptosis, Cell Death, Humans, Mice, NF-kappa B genetics, Oxidation-Reduction, Reactive Oxygen Species metabolism, Resorcinols, Sulfhydryl Compounds, Tumor Suppressor Protein p53 genetics, Antioxidants pharmacology, Neoplasms
- Abstract
Specific focus on "redox cancer therapy" by targeting drugs to redox homeostasis of the cancer cells is growing rapidly. Recent clinical studies showed that N-acetyl cysteine (NAC) treatment significantly decreased the metabolic heterogeneity and reduced Ki67 (a proliferation marker) with simultaneous enhancement in apoptosis of tumor cells in patients. However, it is not yet precisely known how thiol antioxidants enhance killing of cancer cells in a context dependent manner. To this end, we showed that a dietary compound, malabaricone C (mal C) generated copious amounts of reactive oxygen species (ROS) and also reduced GSH level in lung cancer cells. Paradoxically, although antioxidants supplementation reduced mal C-induced ROS, thiol-antioxidants (NAC/GSH) restored intracellular GSH level but enhanced DNA DSBs and apoptotic cell death induced by mal C. Our results unraveled two tightly coupled biochemical mechanisms attributing this sensitization process by thiol antioxidants. Firstly, thiol antioxidants enable the "catechol-quinone redox cycle" of mal C and ameliorate ROS generation and bio-molecular damage (DNA and protein). Secondly, thiol antioxidants cause rapid glutathionylation of transcription factors [p53, p65 (NF-κB) etc.], oxidized by mal C, and abrogates their nuclear sequestration and transcription of the anti-apoptotic genes. Furthermore, analyses of the mitochondrial fractions of p53 expressing and silenced cells revealed that cytoplasmic accumulation of glutathionylated p53 (p53-SSG) triggers a robust mitochondrial death process. Interestingly, mutation of redox sensitive cysteine residues at 124, 141 and 182 position in p53 significantly reduces mal C plus NAC mediated sensitization of cancer cells. The preclinical results, in two different tumor models in mice, provides further support our conclusion that NAC is able to sensitize mal C induced suppression of tumor growth in vivo., Competing Interests: Declaration of competing interest The authors have no conflict of interest to report., (Copyright © 2020 Elsevier Inc. All rights reserved.)
- Published
- 2020
- Full Text
- View/download PDF
27. Salinomycin reduces growth, proliferation and metastasis of cisplatin resistant breast cancer cells via NF-kB deregulation.
- Author
-
Tyagi M and Patro BS
- Subjects
- Breast Neoplasms drug therapy, Breast Neoplasms metabolism, Cell Proliferation drug effects, Cell Survival drug effects, Humans, MCF-7 Cells, Antineoplastic Agents pharmacology, Cisplatin, Drug Resistance, Neoplasm, NF-kappa B metabolism, Pyrans pharmacology
- Abstract
Cisplatin (cis-diamminedichloro-platinum, CDDP), is a widely used platinum compound for various solid tumors including breast cancer as first line of therapy. However, its positive effects are limited due to acquired drug resistance and severe side effects in non-malignant tissue, especially due to dose-dependent nephro- and/or neuro-toxicity. Salinomycin is an antibiotic with coccidiostat effect and has shown anticancer efficacy against various cancer cells with selectivity in targeting cancer stem cells. In the present study, anticancer efficacy and mechanism of action of salinomycin in CDDP-resistant human breast cancer (MCF7
DDP ) cells has been examined. Initially, we generated CDDP-resistant cells by a new protocol followed by checking the anticancer efficacy of salinomycin through MTT, clonogenic, annexin-V/PI and sub-G1 assay. Our results demonstrated that salinomycin diminished both cell proliferation and metastatic migration of MCF7DDP cells. Salinomycin also induced mitochondrial dysfunction in CDDP-resistant breast cancer cells. The analysis of nuclear translocation of pro-survival transcription factors by western blotting showed a distinct role of p65 (NF-κB) in CDDP-mediated resistance in breast cancer. Salinomycin abrogated nuclear translocation of NF-κB proteins and also caused a concurrent reduction in NF-κB regulated expression of pro-survival proteins e.g., survivin, XIAP and BCL-2 in CDDP-resistant cells. These results suggest that a follow up treatment of salinomycin may be promising strategy against CDDP resistant breast cancer cells and metastasis and help in reducing CDDP-induced side effects., (Copyright © 2019 Elsevier Ltd. All rights reserved.)- Published
- 2019
- Full Text
- View/download PDF
28. Mechanism of coralyne-mediated DNA photo-nicking process.
- Author
-
Patro BS, Bhattacharyya R, Gupta P, Bandyopadhyay S, and Chattopadhyay S
- Subjects
- Berberine analogs & derivatives, Berberine pharmacology, Light, Berberine Alkaloids pharmacology, DNA Cleavage drug effects, DNA Cleavage radiation effects
- Abstract
Previously, we reported that coralyne and UVA combination sensitized a wide range of human carcinoma cells regardless of their p53 status. The coralyne induced photosensitization of cancer cells may be clinically attractive, as mutation in the p53 gene is prevalent in many types of tumors. Coralyne mediated photosensitization of cancer cells is attributable to its ability to cause extensive DNA single strand breaks (SSB). However, the precise mechanism of coralyne induced DNA photo-damage is not yet known. The present study was aimed to understand the hitherto unknown mechanism of the coralyne-induced DNA photo-cleavage process. To this end, we compared the DNA photo-nicking properties of berberine, jatrorrhizine and coralyne, and deciphered involvement of the photochemical processes in the photo-nuclease action of coralyne using absorption and electron spin resonance spectroscopy, high performance liquid chromatography and mass spectroscopy (MS) techniques in conjunction with relevant in vitro studies with plasmid DNA. In association with UVA, coralyne, but not berberine and jatrorrhizine induced significant nicking of plasmid DNA via an O
2 -independent photo-chemical process. The Job's plot of our spectrophotometric data suggested that one coralyne molecule remains intercalated with two DNA base pairs (i. e., 1:2) and starts forming aggregates beyond this molar ratio. The DNA photo-nicking by the combination of coralyne and UVA (designated as CUVA) was primarily caused by the coralyne aggregates without any significant contribution from the DNA-intercalated coralyne monomer., (Copyright © 2019 Elsevier B.V. All rights reserved.)- Published
- 2019
- Full Text
- View/download PDF
29. Cell Permeable Imidazole-Desferrioxamine Conjugates: Synthesis and In Vitro Evaluation.
- Author
-
Pramanik S, Chakraborty S, Sivan M, Patro BS, Chatterjee S, and Goswami D
- Subjects
- Biphenyl Compounds chemistry, Deferoxamine metabolism, Fluorescein chemistry, Fluoresceins chemistry, Imidazoles metabolism, In Vitro Techniques, Iron chemistry, Picrates chemistry, Spectrophotometry, Ultraviolet, Cell Membrane Permeability, Deferoxamine chemistry, Imidazoles chemistry
- Abstract
Desferrioxamine (DFO), a clinically approved iron chelator used for iron overload, is unable to chelate labile plasma iron (LPI) because of its limited cell permeability. Herein, alkyl chain modified imidazolium cations with varied hydrophobicities have been conjugated with DFO. The iron binding abilities and the antioxidant properties of the conjugates were found to be similar to DFO. The degree of cellular internalization was much higher in the octyl-imidazolium-DFO conjugate (IV) compared with DFO, and IV was able to chelate LPI in vitro. This opens up a new avenue in using N-alkyl imidazolium salts as a delivery vector for hydrophilic cell-impermeable drugs.
- Published
- 2019
- Full Text
- View/download PDF
30. Spice-derived phenolic, malabaricone B induces mitochondrial damage in lung cancer cells via a p53-independent pathway.
- Author
-
Tyagi M, Maity B, Saha B, Bauri AK, Subramanian M, Chattopadhyay S, and Patro BS
- Subjects
- A549 Cells, Animals, Apoptosis drug effects, Caspase 3 genetics, Caspase 3 metabolism, Caspase 9 genetics, Caspase 9 metabolism, Cell Line, Tumor, Curcumin pharmacology, Cytochromes c metabolism, DNA Fragmentation, HEK293 Cells, Humans, Inhibitory Concentration 50, Membrane Potential, Mitochondrial drug effects, Mice, Mice, SCID, Reactive Oxygen Species metabolism, Tumor Suppressor Protein p53 genetics, Xenograft Model Antitumor Assays, bcl-2-Associated X Protein genetics, bcl-2-Associated X Protein metabolism, Lung Neoplasms drug therapy, Mitochondria drug effects, Resorcinols pharmacology
- Abstract
The spice-derived phenolic, malabaricone B (mal B) showed selective toxicity to human lung cancer (A549), malignant melanoma (A375) and T cell leukemia (Jurkat) cell lines, without showing toxicity to human normal intestinal (INT407), human kidney (HEK293) and lung fibroblast (WI-38) cells. Among the chosen cancer cell lines, mal B showed maximum cytotoxicity to the A549 cells (IC50 = 8.1 ± 1.0 μM), which was significantly better than that of curcumin (IC50 = 26.7 ± 3.1 μM). Further morphological studies by phase contrast microscopy and a clonogenic assay of the A549 cells revealed that mal B treatment increased the number of shrinking cells and also abolished the clonal proliferation of the cells. Mal B induced apoptotic cell death was confirmed by DNA laddering and quantified by cytoplasmic oligonucleosome formation and annexin V/PI assays. The mal B-induced apoptosis was mediated by an increase in the intracellular reactive oxygen species (ROS), because the cell-permeable antioxidants, N-acetylcysteine (NAC) and PEG-SOD, strongly inhibited its cytotoxicity to the A549 cells. Mal B increased the BAX level while simultaneously decreasing the BCL-2 and BCL-XL levels in the A549 cells, triggering the mitochondrial apoptotic pathway as revealed from the release of cytochrome c, and the activation of caspase-9 and caspase-3. Pre-treatment of cells with caspase-9, caspase-3 and pan-caspase inhibitors made them more resistant to mal B treatment. This effect of mal B was strongly associated with the concomitant decrease in anti-apoptotic (IAP1, IAP2 and survivin), angiogenic (growth factors) and cancer invasiveness (matrix metalloproteinase-9, COX-2) modulating proteins. Mal B induced cytotoxicity was unaffected by the shRNA-mediated depletion of p53 in A549 cells. Most importantly, mal B sensitized a wide range of human carcinoma cells regardless of their p53 status. Finally, mal B (100 mg kg-1) also inhibited lung tumor (xenograft) growth in SCID mice.
- Published
- 2018
- Full Text
- View/download PDF
31. Resveratrol analogue, trans-4,4'-dihydroxystilbene (DHS), inhibits melanoma tumor growth and suppresses its metastatic colonization in lungs.
- Author
-
Saha B, Pai GB, Subramanian M, Gupta P, Tyagi M, Patro BS, and Chattopadhyay S
- Subjects
- Animals, Antineoplastic Agents, Phytogenic pharmacology, Apoptosis drug effects, Cell Line, Tumor, Cell Movement drug effects, Cell Proliferation drug effects, G1 Phase Cell Cycle Checkpoints drug effects, Gene Expression Regulation, Neoplastic drug effects, Humans, Lung Neoplasms secondary, Male, Melanoma drug therapy, Melanoma pathology, Melanoma, Experimental pathology, Mice, Mice, Inbred C57BL, Neoplasm Invasiveness prevention & control, Neoplasm Metastasis prevention & control, Skin Neoplasms pathology, Lung Neoplasms prevention & control, Melanoma, Experimental drug therapy, Skin Neoplasms drug therapy, Stilbenes pharmacology
- Abstract
The prevalence of melanoma and the lack of effective therapy for metastatic melanoma warrant extensive and systematic evaluations of small molecules in cellular and pre-clinical models. We investigated, herein, the antitumor and anti-metastatic effects of trans-4,4'-dihydroxystilbene (DHS), a natural product present in bark of Yucca periculosa, using in vitro and in vivo melanoma murine models. DHS showed potent melanoma cytotoxicity, as determined by MTT and clonogenic assay. Further, DHS induced cytotoxicity was mediated through apoptosis, which was assessed by annexin V-FITC/PI, sub-G1 and caspase activation assays. In addition, DHS inhibited cell proliferation by inducing robust cell cycle arrest in G1-phase. Imperatively, these inhibitory effects led to a significant reduction of melanoma tumor in pre-clinical murine model. DHS also inhibited cell migration and invasion of melanoma cells, which were examined using wound healing and Transwell migration/invasion assays. Mechanistically, DHS modulated the expressions of several key metastasis regulating proteins e.g., MMP-2/9, N-cadherin, E-cadherin and survivin. We also showed the anti-metastatic effect of DHS in a melanoma mediated lung metastasis model in vivo. DHS significantly reduced large melanoma nodule formation in the parenchyma of lungs. Therefore, DHS may represent a promising natural drug in the repertoire of treatment against melanoma tumor growth and metastasis., (Copyright © 2018 Elsevier Masson SAS. All rights reserved.)
- Published
- 2018
- Full Text
- View/download PDF
32. Coralyne, a protoberberine alkaloid, causes robust photosenstization of cancer cells through ATR-p38 MAPK-BAX and JAK2-STAT1-BAX pathways.
- Author
-
Bhattacharyya R, Gupta P, Bandyopadhyay SK, Patro BS, and Chattopadhyay S
- Subjects
- Apoptosis drug effects, Ataxia Telangiectasia Mutated Proteins metabolism, Berberine Alkaloids therapeutic use, Cell Line, Tumor, Cells, Cultured, Humans, Janus Kinase 2 metabolism, Keratinocytes drug effects, Neoplasms drug therapy, Photosensitizing Agents pharmacology, STAT1 Transcription Factor metabolism, bcl-2-Associated X Protein metabolism, p38 Mitogen-Activated Protein Kinases metabolism, Berberine Alkaloids pharmacology, Photosensitivity Disorders, Signal Transduction drug effects
- Abstract
Photodynamic therapy (PDT) provides an effective cancer treatment option but it requires sufficient cellular oxygen concentration to exert its photosensitizing effects. Due to hypoxic nature of most tumors, widespread clinical application of PDT is restricted and warrants development of photosensitizers which can kill cancer cells in ROS independent manner. Previously, we reported significant enhancement of the anti-cancer property of coralyne in presence of ultraviolet-A (UVA) light exposure against several human carcinoma cell lines. This study aimed at unravelling molecular cascades of events in CUVA treatment (coralyne and UVA light)-mediated photosensitization of human skin cancer. The CUVA-treatment caused robust apoptosis of A431 cancer cells, primarily through mitochondrial and lysosomal dysfunctions. Silencing of BAX conferred a significant protection against CUVA-induced apoptosis. Both lysosomal proteases and caspase-8 activation contributed to BID cleavage. Further, our results revealed that a dual signaling axis e.g., ATR-p38 MAPK and JAK2-STAT1 pathways functioned upstream of BAX activation in apoptosis response. Moreover, transient silencing of ATR and pharmacological inhibition of p38-MAPK or JAK2 significantly abolished the effect of CUVA treatment induced BAX expression and cell death, linking the extrinsic and intrinsic pathways with the observed cell death. Our data suggest that coralyne, which is known topoisomerase-I inhibitor, may be an attractive agent for photo-chemotherapeutic treatment of human skin cancers., (Copyright © 2018 Elsevier B.V. All rights reserved.)
- Published
- 2018
- Full Text
- View/download PDF
33. Synergistic enhancement in the drug sequestration power and reduction in the cytotoxicity of surfactants.
- Author
-
Kalel R, Mora AK, Patro BS, Palit DK, and Nath S
- Subjects
- Hydrophobic and Hydrophilic Interactions, Intercalating Agents, Micelles, Pharmaceutical Preparations, Pharmacokinetics, Poloxamer, Polymers, Solubility, DNA chemistry, Sodium Dodecyl Sulfate chemistry, Surface-Active Agents chemistry
- Abstract
Surfactants have often been employed for the sequestration of drugs from DNA. However, for an effective sequestration, the concentration of the surfactant needs to be higher than its critical micellar concentration (CMC). Use of such high concentrations of the surfactant may limit its practical usage as a sequestering agent due to its cytotoxicity. In the present study we have shown that sodium dodecyl sulfate (SDS) itself at a concentration less than its CMC failed to sequester a drug from DNA. However, the sequestration power of SDS at sub-CMC concentration could be enhanced to a significant extent when incorporated into Pluronic polymer micelles in the form of supramolecular assemblies. Such a sequestration process was monitored through detailed photophysical properties of a model drug using steady-state and time-resolved fluorescence techniques. It has also been demonstrated that unlike a conventional surfactant, the sequestration of drugs by SDS-polymer supramolecular assemblies can be controlled by their compositions. Two Pluronic polymers with different compositions have been used to understand the effect of polymer composition on the sequestration process. It has been shown that with the increase in the length of the hydrophilic blocks of the polymer, the extent of sequestration decreases due to the decrease in the sequestering force exerted on the intercalated drug. Most importantly, our in vitro cell viability studies show that the toxicity of the SDS surfactant is reduced to a remarkable extent due to its incorporation into the polymer micelles.
- Published
- 2017
- Full Text
- View/download PDF
34. Differential modes of photosensitisation in cancer cells by berberine and coralyne.
- Author
-
Bhattacharyya R, Saha B, Tyagi M, Bandyopadhyay SK, Patro BS, and Chattopadhyay S
- Subjects
- A549 Cells, Berberine analogs & derivatives, Caspases metabolism, Cell Proliferation drug effects, Cell Proliferation radiation effects, DNA Breaks, Double-Stranded drug effects, DNA Replication drug effects, DNA Replication radiation effects, Humans, Lung Neoplasms pathology, Photosensitizing Agents administration & dosage, Reactive Oxygen Species radiation effects, Ultraviolet Rays, Berberine administration & dosage, Berberine Alkaloids administration & dosage, Lung Neoplasms drug therapy, Lung Neoplasms radiotherapy
- Abstract
In this study, we demonstrated that the cytotoxicity of the protoberberine alkaloids such as coralyne, berberine and jatrorrhizine to several human cancer cell lines can be improved significantly in combination with UVA exposure. However, the phototoxic property of coralyne was much higher than that of the other two alkaloids. The combination of coralyne and UVA (designated as CUVA) induced oxygen-independent cytotoxicity in the human lung cancer A549 cells by producing more lethal DNA double-strand breaks, and the effect was mediated via the replication machinery. In comparison, the berberine-induced phototoxicity to the A549 cells was mediated by reactive oxygen species generation, mitochondrial membrane permeabilisation and caspase-9/caspase-3 activation.
- Published
- 2017
- Full Text
- View/download PDF
35. trans -4,4'-Dihydroxystilbene (DHS) inhibits human neuroblastoma tumor growth and induces mitochondrial and lysosomal damages in neuroblastoma cell lines.
- Author
-
Saha B, Patro BS, Koli M, Pai G, Ray J, Bandyopadhyay SK, and Chattopadhyay S
- Abstract
In view of the inadequacy of neuroblastoma treatment, five hydroxystilbenes and resveratrol (Resv) were screened for their cytotoxic property against human neuroblastoma cell lines. The mechanism of cytotoxic action of the most potent compound, trans -4,4'-dihydroxystilbene (DHS) was investigated in vitro using human neuroblastoma cell lines. DHS was also tested in a mouse xenograft model of human neuroblastoma tumor. The MTT, sub-G1, annexin V and clonogenic assays as well as microscopy established higher cytotoxicity of DHS than Resv to the IMR32 cell line. DHS (20 μM) induced mitochondrial membrane permeabilization (MMP) in the cells, as revealed from JC-1 staining, cytochrome c and ApaF1 release and caspases-9/3 activation. DHS also induced lysosomal membrane permeabilization (LMP) to release cathepsins B, L and D, and the cathepsins inhibitors partially reduced MMP/caspase-3 activation. The ROS, produced by DHS activated the p38 and JNK MAPKs to augment the BAX activity and BID-cleavage, and induce LMP and MMP in the cells. DHS (100 mg/kg) also inhibited human neuroblastoma tumor growth in SCID mice by 51%. Hence, DHS may be a potential chemotherapeutic option against neuroblastoma. The involvement of an independent LMP as well as a partially LMP-dependent MMP by DHS is attractive as it provides options to target both mitochondria and lysosome., Competing Interests: CONFLICTS OF INTEREST The authors have no conflicts of interest to report.
- Published
- 2017
- Full Text
- View/download PDF
36. trans-4,4'-Dihydroxystilbene (DHS) protects PC12 cells from oxidative damage but induces reactive oxygen species-mediated apoptosis in SHSY-5Y cell line.
- Author
-
Saha B, Subramanian M, Gupta P, Patro BS, Ray J, Bandyopadhyay SK, and Chattopadhyay S
- Subjects
- Animals, Cell Survival, Humans, Hydrogen Peroxide, PC12 Cells, Rats, Reactive Oxygen Species, Apoptosis drug effects, Oxidative Stress drug effects, Stilbenes pharmacology
- Abstract
Polyphenols can exert both, antioxidant and pro-oxidant properties, depending on cell types as well as their concentrations. Hence, it was of interest to examine if the naturally occurring resveratrol analog, trans-4,4'-dihydroxystilbene (DHS) also exert both these activities in a biphasic or cell-specific manner. In this study, we established the cytoprotective action of DHS against hydrogen peroxide (H2O2)-induced apoptotic death of the PC12 cells. DHS reduced mitochondrial membrane permeabilization and deactivated reactive oxygen species (ROS)-mediated caspase-3 activation in the H2O2-treated PC12 cells. However, it induced apoptosis in the human neuroblastoma SHSY-5Y cell line by destabilizing mitochondrial membrane, augmenting ROS and activating caspapse-3. DHS showed better activity than resveratrol in both the chosen models.
- Published
- 2016
37. Syntheses and photodynamic activity of some glucose-conjugated BODIPY dyes.
- Author
-
Shivran N, Tyagi M, Mula S, Gupta P, Saha B, Patro BS, and Chattopadhyay S
- Subjects
- Biological Transport, Boron Compounds chemistry, Cell Line, Tumor, Chemistry Techniques, Synthetic, Coloring Agents chemistry, Humans, Photochemotherapy, Boron Compounds chemical synthesis, Boron Compounds pharmacology, Coloring Agents chemical synthesis, Coloring Agents pharmacology, Glucose chemistry
- Abstract
The syntheses of three water-soluble glucose-conjugated BODIPY dyes with different wavelength emissions and studies of their photodynamic therapeutic (PDT) action on human lung cancer A549 cell line are disclosed. Amongst the chosen compounds, the BODIPY dye 4 possessing a glycosylated styryl moiety at the C-3 position showed best PDT property against the A549 cell line. In particular, it induced reactive oxygen species-mediated caspase-8/caspase-3-dependent apoptosis as revealed from the increased sub G1 cell population and changes in cell morphology. These results along with its localization in the endoplasmic reticulum, as revealed by confocal microscopy suggested that mitochondria may not be directly involved in the photo-cytotoxicity of 4. Compound 4 did not induce any dark toxicity to the A549 cells, and was non-toxic to normal lung cells., (Copyright © 2016 Elsevier Masson SAS. All rights reserved.)
- Published
- 2016
- Full Text
- View/download PDF
38. PicoGreen: a better amyloid probe than Thioflavin-T.
- Author
-
Mora AK, Singh PK, Patro BS, and Nath S
- Subjects
- Benzothiazoles, Humans, Models, Molecular, Molecular Docking Simulation, Organic Chemicals chemistry, Protein Binding, Amyloid chemistry, Fluorescent Dyes chemistry, Insulin chemistry, Molecular Probes chemistry, Thiazoles chemistry
- Abstract
PicoGreen, a cyanine based ultrafast molecular rotor, shows high affinity towards amyloid fibrils and scores a much better sensitivity than Thioflavin-T, a gold standard probe for amyloid fibrils. Detailed spectroscopic and molecular docking studies have been performed to understand the mode of interaction between PicoGreen and amyloid fibrils.
- Published
- 2016
- Full Text
- View/download PDF
39. Mechanism of the anti-hypertensive property of the naturally occurring phenolic, malabaricone C in DOCA-salt rats.
- Author
-
Rathee JS, Patro BS, Brown L, and Chattopadhyay S
- Subjects
- Animals, Antioxidants pharmacology, Aorta drug effects, Aorta pathology, Desoxycorticosterone Acetate, Disease Models, Animal, Heart drug effects, Hypertension chemically induced, Hypertrophy drug therapy, Male, Myocardium pathology, Oxidative Stress drug effects, Rats, Vasodilation drug effects, Antihypertensive Agents therapeutic use, Hypertension drug therapy, Resorcinols therapeutic use
- Abstract
In this study, we studied whether chronic oral administration of the natural antioxidant, malabaricone C (mal C) can reduce blood pressure (BP) and attenuate cardio-vascular remodeling in deoxycorticosterone acetate (DOCA)-salt hypertensive rats. The dose of mal C for its anti-hypertensive action was optimized by measuring the systolic BP (SBP). DOCA-salt rats showed very high SBP, associated with organ hypertrophy, collagen depositions, and inflammatory infiltrations in cardiac and aortic sections, reduced plasma total antioxidant status and NO level, and increased levels of TBARS, PGI2 as well as vasoconstrictors (AVP, Big ET, and ET-1). DOCA-salt also reduced smooth muscle- and endothelium-dependent vascular relaxation in rats. Mal C reversed all these changes of the DOCA-salt rats and improved their vascular reactivity. Mal C exerts anti-hypertensive property in DOCA-salt rats by reducing oxidative stress and organ hypertrophy, and improving endothelial and vascular functions. Given that mal C has appreciable natural abundance and is non-toxic to rodents, further studies would help in establishing its medicinal potential against hypertension.
- Published
- 2016
- Full Text
- View/download PDF
40. Mechanism of the malabaricone C-induced toxicity to the MCF-7 cell line.
- Author
-
Tyagi M, Patro BS, and Chattopadhyay S
- Subjects
- Apoptosis, Cell Line, Tumor, Cell Proliferation, Female, Humans, MCF-7 Cells, Signal Transduction, Breast Neoplasms metabolism, Resorcinols metabolism
- Abstract
In this study, we studied the mechanism of the cytotoxicity of malabaricone C (mal C) against human breast cancer MCF-7 cell line. Mal C dose-dependently increased the sub G1 cell population, associated with cytoplasmic oligonucleosome formation and chromatin condensation. The mal C-induced apoptosis led to mitochondrial damage as revealed by fluorescence microscopy and flow cytometry of the JC-1-stained cells as well as from the release of mitochondrion-specific nuclease proteins AIF and endo G. Mal C also released intracellular Ca(2+) from the MCF-7 cells, but the Ca(2+)-modulators BAPTA-AM and Ru360 only partially abrogated the apoptosis. The calpain activation by mal C did not have any effect on its cytotoxicity. On the other hand, after mal C treatment significant lysosomal membrane permeabilization (LMP), along with release of cathepsin B, as well as Bid-cleavage and its translocation to mitochondria were observed much earlier than the mitochondrial damage. This suggested that cytotoxicity of mal C against human MCF-7 human breast cancer cell line may proceed through LMP as the initial event that triggered a caspase-independent, but cathepsin B and t-Bid-dependent intrinsic mitochondrial apoptotic pathway. A significant accumulation of cells in the S or G2-M phases along with upregulation of the cyclins E and A due to mal C exposure promises it to be a potential anti-cancer agent.
- Published
- 2014
- Full Text
- View/download PDF
41. DNA damage dependent activation of checkpoint kinase-1 and mitogen-activated protein kinase-p38 are required in malabaricone C-induced mitochondrial cell death.
- Author
-
Tyagi M, Bhattacharyya R, Bauri AK, Patro BS, and Chattopadhyay S
- Subjects
- Ataxia Telangiectasia Mutated Proteins physiology, Cell Line, Tumor, Checkpoint Kinase 1, Enzyme Activation, Humans, Mitochondria pathology, Proto-Oncogene Proteins c-bcl-2 analysis, bcl-2-Associated X Protein analysis, Apoptosis drug effects, DNA Damage, Mitochondria drug effects, Protein Kinases physiology, Resorcinols pharmacology, p38 Mitogen-Activated Protein Kinases physiology
- Abstract
Background: Given that lung cancer is the second leading cause of cancer-related deaths with low survival rates, the project was aimed to formulate an efficient drug with minimum side effects, and rationalize its action mechanistically., Methods: Mitochondria deficient cells, shRNA-mediated BCL2 and ATM depleted cells and pharmacological inhibition of DNA-damage response proteins were employed to explore the signaling mechanism governed between nucleus and mitochondria in response to mal C., Results: Mal C decreased cell viability in three lung carcinoma cells, associated with DNA damage, p38-MAPK activation, imbalance in BAX/BCL2 expression, mitochondrial dysfunction and cytochrome-c release. Mitochondria depletion and p38-MAPK inhibition made A549 cells extremely resistant, but BCL2 knock-down partially sensitized the cells to mal C treatment. The mal C-induced apoptosis in A549 cells was initiated by DNA single strand breaks that led to double strand breaks (DSBs). DSB generation paralleled the induction of ATM- and ATR-mediated CHK1 phosphorylation. ATM silencing and ATR inhibition partially attenuated the mal C-induced p38-MAPK activation, CHK1 phosphorylation and apoptosis, which were completely suppressed by CHK1 inhibition., Conclusions: Mal C activates the ATM-CHK1-p38 MAPK cascade to cause mitochondrial cell death in lung carcinoma cells., General Significance: Given that mal C has appreciable natural abundance and is non-toxic to mice, further in vivo evaluation would help in establishing its anti-cancer property., (Copyright © 2013 Elsevier B.V. All rights reserved.)
- Published
- 2014
- Full Text
- View/download PDF
42. Molecular mechanism of the anti-inflammatory activity of a natural diarylnonanoid, malabaricone C.
- Author
-
Maity B, Yadav SK, Patro BS, Tyagi M, Bandyopadhyay SK, and Chattopadhyay S
- Subjects
- Animals, Base Sequence, Cell Line, DNA Primers, Interleukin-1beta metabolism, Lipopolysaccharides pharmacology, Macrophages drug effects, Macrophages metabolism, Magnetic Resonance Spectroscopy, Male, Mice, NF-kappa B metabolism, Nitric Oxide metabolism, Phosphorylation, Tumor Necrosis Factor-alpha metabolism, Anti-Inflammatory Agents pharmacology, Resorcinols pharmacology
- Abstract
The spice-derived phenolic, malabaricone C (mal C), has recently been shown to accelerate healing of the indomethacin-induced gastric ulceration in mice. In this study, we explored its anti-inflammatory activity and investigated the underlying mechanism of the action. Mal C suppressed the microvascular permeability and the levels of tumor necrosis factor-α, interleukin-1β, and nitric oxide in the lipopolysaccharide (LPS)-administered mice. At a dose of 10 mg/kg, it showed anti-inflammatory activity comparable to that of omeprazole (5 mg/kg) and dexamethasone (50 mg/kg). It also reduced the expression and activities of inducible nitric oxide synthase, cyclooxygenase-2, as well as the pro- vs anti-inflammatory cytokine ratio in the LPS-treated RAW macrophages. Mal C was found to inhibit LPS-induced NF-kB activation in RAW 264.7 cells by blocking the MyD88-dependent pathway. Mal C suppressed NF-κB activation and iNOS promoter activity, which correlated with its inhibitory effect on IκB phosphorylation and degradation, and NF-κB nuclear translocation, in the LPS-stimulated macrophages. It also inhibited LPS-induced phosphorylation of p38 and JNK, which are also upstream activators of NF-κB, without affecting Akt phosphorylation. Mal C also effectively blocked the PKR-mediated activation of NF-κB. These findings indicate that mal C exerts an anti-inflammatory effect through NF-κB-responsive inflammatory gene expressions by inhibiting the p38 and JNK-dependent canonical NF-κB pathway as well as the PKR pathway, and is a potential therapeutic agent against acute inflammation., (Copyright © 2012 Elsevier Inc. All rights reserved.)
- Published
- 2012
- Full Text
- View/download PDF
43. WRN helicase regulates the ATR-CHK1-induced S-phase checkpoint pathway in response to topoisomerase-I-DNA covalent complexes.
- Author
-
Patro BS, Frøhlich R, Bohr VA, and Stevnsner T
- Subjects
- Ataxia Telangiectasia Mutated Proteins, Camptothecin pharmacology, Cell Cycle Proteins genetics, Cell Line, Tumor, Checkpoint Kinase 1, Chromatin genetics, Chromatin metabolism, Chromatin Immunoprecipitation, DNA Damage, DNA Replication, DNA, Single-Stranded genetics, DNA, Single-Stranded metabolism, Exodeoxyribonucleases genetics, Gene Expression Regulation, Enzymologic, Gene Knockdown Techniques, Genomic Instability, Humans, Hydroxyurea pharmacology, Phosphorylation, Protein Kinases genetics, Protein Serine-Threonine Kinases genetics, RNA, Small Interfering genetics, RNA, Small Interfering metabolism, RecQ Helicases genetics, Topoisomerase I Inhibitors pharmacology, Werner Syndrome Helicase, Cell Cycle Proteins metabolism, Exodeoxyribonucleases metabolism, Protein Kinases metabolism, Protein Serine-Threonine Kinases metabolism, RecQ Helicases metabolism, S Phase Cell Cycle Checkpoints
- Abstract
Checkpoints are cellular surveillance and signaling pathways that coordinate the response to DNA damage and replicative stress. Consequently, failure of cellular checkpoints increases susceptibility to DNA damage and can lead to profound genome instability. This study examines the role of a human RECQ helicase, WRN, in checkpoint activation in response to DNA damage. Mutations in WRN lead to genomic instability and the premature aging condition Werner syndrome. Here, the role of WRN in a DNA-damage-induced checkpoint was analyzed in U-2 OS (WRN wild type) and isogenic cells stably expressing WRN-targeted shRNA (WRN knockdown). The results of our studies suggest that WRN has a crucial role in inducing an S-phase checkpoint in cells exposed to the topoisomerase I inhibitor campthothecin (CPT), but not in cells exposed to hydroxyurea. Intriguingly, WRN decreases the rate of replication fork elongation, increases the accumulation of ssDNA and stimulates phosphorylation of CHK1, which releases CHK1 from chromatin in CPT-treated cells. Importantly, knockdown of WRN expression abolished or delayed all these processes in response to CPT. Together, our results strongly suggest an essential regulatory role for WRN in controlling the ATR-CHK1-mediated S-phase checkpoint in CPT-treated cells.
- Published
- 2011
- Full Text
- View/download PDF
44. Mitochondrial dysfunction mediated by quinone oxidation products of dopamine: Implications in dopamine cytotoxicity and pathogenesis of Parkinson's disease.
- Author
-
Jana S, Sinha M, Chanda D, Roy T, Banerjee K, Munshi S, Patro BS, and Chakrabarti S
- Subjects
- Animals, Apoptosis drug effects, Brain drug effects, Caspases metabolism, Dopamine metabolism, Energy Metabolism drug effects, Membrane Potential, Mitochondrial drug effects, Mitochondria physiology, Oxidation-Reduction, Oxidative Phosphorylation drug effects, PC12 Cells, Parkinson Disease metabolism, Rats, Reactive Oxygen Species metabolism, Dopamine toxicity, Mitochondria drug effects, Parkinson Disease etiology, Quinones toxicity
- Abstract
The study has demonstrated that dopamine induces membrane depolarization and a loss of phosphorylation capacity in dose-dependent manner in isolated rat brain mitochondria during extended in vitro incubation and the phenomena are not prevented by oxyradical scavengers or metal chelators. Dopamine effects on brain mitochondria are, however, markedly prevented by reduced glutathione and N-acetyl cysteine and promoted by tyrosinase present in the incubation medium. The results imply that quinone oxidation products of dopamine are involved in mitochondrial damage under this condition. When PC12 cells are exposed to dopamine in varying concentrations (100-400μM) for up to 24h, a pronounced impairment of mitochondrial bio-energetic functions at several levels is observed along with a significant (nearly 40%) loss of cell viability with features of apoptotic nuclear changes and increased activities of caspase 3 and caspase 9 and all these effects of dopamine are remarkably prevented by N-acetyl cysteine. N-acetyl cysteine also blocks nearly completely the dopamine induced increase in reactive oxygen species production and the formation of quinoprotein adducts in mitochondrial fraction within PC12 cells and also the accumulation of quinone products in the culture medium. Clorgyline, an inhibitor of MAO-A, markedly decreases the formation of reactive oxygen species in PC12 cells upon dopamine exposure but has only mild protective actions against quinoprotein adduct formation, mitochondrial dysfunctions, cell death and caspase activation induced by dopamine. The results have indicated that quinone oxidation products and not reactive oxygen species are primarily involved in cytotoxic effects of dopamine and the mitochondrial impairment plays a central role in the latter process. The data have clear implications in the pathogenesis of Parkinson's disease., (2011 Elsevier B.V. All rights reserved.)
- Published
- 2011
- Full Text
- View/download PDF
45. Comparative nuclease and anti-cancer properties of the naturally occurring malabaricones.
- Author
-
Patro BS, Tyagi M, Saha J, and Chattopadhyay S
- Subjects
- Animals, Antineoplastic Agents chemistry, Apoptosis drug effects, Cattle, Cell Line, Tumor, Cell Proliferation drug effects, Chelating Agents chemistry, Chelating Agents pharmacology, Copper chemistry, DNA drug effects, DNA Cleavage, DNA Damage, Dose-Response Relationship, Drug, Drug Screening Assays, Antitumor, Humans, Molecular Structure, Reactive Oxygen Species metabolism, Resorcinols chemistry, Stereoisomerism, Structure-Activity Relationship, Antineoplastic Agents pharmacology, Copper pharmacology, Resorcinols pharmacology
- Abstract
The nuclease activities of the malabaricones have been studied so as to establish a structure-activity correlation and deduce the mechanistic pathway of the process. The inactivity of malabaricone A and malabaricone D revealed that the resorcinol moiety, present in the malabaricones did not contribute to the nuclease activity. Amongst the test compounds, malabaricone C (mal C) containing a B-ring catechol moiety showed significantly better Cu(II)-dependent nuclease activity than the partially methylated catechol derivative, mal B and curcumin. Mal C was found to bind efficiently with Cu(II) and DNA to facilitate the DNA nicking via a site-specifically generated Cu(I)-peroxo complex. Consistent with its Cu(II)-dependent nuclease property, mal C showed better cytotoxicity (IC(50)=5.26±1.20 μM) than curcumin (IC(50)=24.46±3.32 μM) against the MCF-7 human breast cancer cell line. The mal C-induced killing of the MCF-7 cells followed an apoptotic pathway involving oxidative damage to the cellular DNA., (Copyright © 2010 Elsevier Ltd. All rights reserved.)
- Published
- 2010
- Full Text
- View/download PDF
46. Topoisomerase inhibitor coralyne photosensitizes DNA, leading to elicitation of Chk2-dependent S-phase checkpoint and p53-independent apoptosis in cancer cells.
- Author
-
Patro BS, Maity B, and Chattopadhyay S
- Subjects
- Animals, Apoptosis physiology, Ataxia Telangiectasia Mutated Proteins, Berberine Alkaloids chemistry, Berberine Alkaloids pharmacology, Caspases metabolism, Cell Cycle Proteins metabolism, Cell Line, Tumor drug effects, Cell Line, Tumor radiation effects, Checkpoint Kinase 2, DNA Damage, DNA Repair, DNA-Binding Proteins metabolism, Enzyme Activation, Humans, Molecular Structure, Neoplasms pathology, Photosensitizing Agents chemistry, Photosensitizing Agents pharmacology, Protein Serine-Threonine Kinases genetics, Tumor Suppressor Protein p53 genetics, Tumor Suppressor Proteins metabolism, Ultraviolet Rays, Berberine Alkaloids metabolism, DNA metabolism, Neoplasms metabolism, Photosensitizing Agents metabolism, Protein Serine-Threonine Kinases metabolism, S Phase physiology, Topoisomerase I Inhibitors, Tumor Suppressor Protein p53 metabolism
- Abstract
The possibility of synergism between the topoisomerase inhibition by coralyne and its DNA photonicking properties being used to kill cancer cells was explored. Compared with coralyne alone, the CUVA treatment dramatically enhanced DNA damage and apoptosis in cells. Despite causing an increased p53 expression, the CUVA treatment led to p53-independent apoptosis, causing almost similar cell death in wild-type, p53 mutant, and p53-silenced tumor cells. Expression of the p53-regulated downstream proteins like p21, and DNA-damage-dependent p53 phosphorylation at serine-15 residue also was not elicited by the CUVA treatment, at a low coralyne concentration. Instead, it led to an immediate activation of the Chk2-mediated S-phase arrest, despite activating PARP protein for DNA repair. The S-phase arrest subsequently ensures apoptosis through activation of caspases-3 and -9, the latter being reflected from the results with a specific caspase-9 inhibitor. Abrogation of Chk2 activity by shRNA or by using ATM-specific inhibitor (ATMi) led to a defective S-phase checkpoint and further augmentation in apoptosis. However, at a high coralyne concentration, the CUVA-induced apoptosis followed multiple and independent pathways, involving several caspases. The CUVA treatment may represent a novel mechanism-based protocol for increasing the efficacy of coralyne in inducing apoptosis in both p53 wild-type and mutant tumor cells.
- Published
- 2010
- Full Text
- View/download PDF
47. Inhibitory property of the Piper betel phenolics against photosensitization-induced biological damages.
- Author
-
Mula S, Banerjee D, Patro BS, Bhattacharya S, Barik A, Bandyopadhyay SK, and Chattopadhyay S
- Subjects
- Animals, Antioxidants, Mice, Mitochondria, Liver metabolism, Oxygen, Plant Extracts, Rats, Lipid Peroxidation drug effects, Phenols pharmacology, Photosensitizing Agents adverse effects, Piper betle chemistry, Radiation-Protective Agents pharmacology
- Abstract
The Piper betel phenolics, allylpyrocatechol (APC) and chavibetol (CHV), were found to protect photosensitization-mediated lipid peroxidation (LPO) of rat liver mitochondria effectively, APC being significantly more potent. The better activity of APC vis-à-vis CHV could be attributed to its higher reactivity with (1)O(2), as revealed from the rate constant values of (1)O(2) quenching by the respective phenolics. APC also prevented the detrimental effects of the Type II photosensitization-induced toxicity to mouse fibroblast L929 cells. The results suggest that APC may play an important role in protecting biological systems against damage, by eliminating (1)O(2) generated from certain endogenous photosensitizers.
- Published
- 2008
- Full Text
- View/download PDF
48. Antioxidant activity of piper betel leaf extract and its constituents.
- Author
-
Rathee JS, Patro BS, Mula S, Gamre S, and Chattopadhyay S
- Subjects
- Animals, Antioxidants chemistry, Antioxidants therapeutic use, Biphenyl Compounds, DNA Damage, Free Radical Scavengers chemistry, Free Radical Scavengers pharmacology, Free Radical Scavengers therapeutic use, Gamma Rays, Hydrogen Peroxide chemistry, Lipid Peroxidation drug effects, Liposomes, Mice, Picrates chemistry, Plant Extracts chemistry, Plant Extracts therapeutic use, Plant Leaves, Radiation-Protective Agents chemistry, Radiation-Protective Agents therapeutic use, Rats, Superoxides chemistry, Antioxidants pharmacology, Phytotherapy, Piper, Plant Extracts pharmacology, Radiation-Protective Agents pharmacology
- Abstract
The 1,1-diphenyl-2-picrylhydrazyl (DPPH) assay of the ethanol extracts of three varieties (Bangla, sweet, and Mysore) of Piper betel (pan) revealed the Bangla variety to possess the best antioxidant activity that can be correlated with the total phenolic content and reducing powers of the respective extracts. Column chromatography of the extract of the Bangla variety led to the isolation of chevibetol (CHV), allylpyrocatechol (APC), and their respective glucosides. The HPTLC analyses of the extracts revealed similar chemical profiles in all three P. betel varieties, although the concentrations of CHV and APC were significantly less in the sweet and Mysore varieties. Among the isolated compounds, APC showed the best results in all the in vitro experiments. It could prevent Fe(II)-induced lipid peroxidation (LPO) of liposomes and rat brain homogenates as well as gamma-ray-induced damage of pBR322 plasmid DNA more efficiently than CHV. The superior anti-LPO and radioprotective activities of APC vis-à-vis those of CHV could not be explained by their respective Fe(II) chelation and .OH radical scavenging capacities. The better ability of APC to scavenge O2-. radicals and H2O2 might account for the results.
- Published
- 2006
- Full Text
- View/download PDF
49. Folic acid as a Fenton-modulator: possible physiological implication.
- Author
-
Patro BS, Adhikari S, Mukherjee T, and Chattopadhyay S
- Subjects
- Animals, Cell Line, Cell Survival drug effects, Deoxyribose chemistry, Edetic Acid chemistry, Fibroblasts drug effects, Fibroblasts metabolism, Hydrogen Peroxide pharmacology, Iron pharmacology, Mice, Molecular Structure, Oxidation-Reduction, Pterins chemistry, Reactive Oxygen Species metabolism, Folic Acid chemistry, Folic Acid pharmacology, Folic Acid physiology, Hydrogen Peroxide chemistry, Iron chemistry
- Abstract
Acting as a redox switch, folic acid (1) might be a promising iron modulator to protect cellular machinery against oxidative stress and iron overload. The vitamin 1 can directly control the iron concentration by oxidizing it even if present in chelated forms. In addition, during its role as a reducing agent for the biologically relevant reactive oxygen species (ROS), it furnishes 6-formyl pterin. This folate-derived intermediate possesses a stronger Fe2+-oxidizing capacity than 1. Thus, compound 1 can reduce the iron toxicity in two ways. Although, the Fe2+-oxidizing capacity is nullified in the presence of a strong biological reductant like ascorbic acid, this property may play a predominant role during pathogenesis when the cellular ascorbic acid levels deplete significantly. The iron-modulatory property of 1 was also confirmed with the L929 mouse fibroblast cell line.
- Published
- 2006
- Full Text
- View/download PDF
50. Antioxidant activity of Myristica malabarica extracts and their constituents.
- Author
-
Patro BS, Bauri AK, Mishra S, and Chattopadhyay S
- Subjects
- Animals, Antioxidants pharmacology, Biphenyl Compounds, DNA Damage drug effects, Free Radical Scavengers pharmacology, Hydroxyl Radical, Lipid Peroxidation drug effects, Mitochondria, Liver chemistry, Phenols analysis, Picrates, Plant Extracts pharmacology, Rats, Resorcinols isolation & purification, Resorcinols pharmacology, Antioxidants analysis, Myristicaceae chemistry, Plant Extracts chemistry
- Abstract
The 1,1-diphenyl-2-picrylhydrazyl (DPPH) assay of the ether, methanol, and aqueous extracts of the spice Myristica malabarica (rampatri) revealed the methanol extract to possess the best antioxidant activity. Column chromatography of the methanol extract led to the isolation of a new 2-acylresorcinol and four known diarylnonanoids of which the diarylnonanoid, malabaricone C, showed the maximum DPPH scavenging activity. Malabaricone C could prevent both Fe(II)- and 2,2'-azobis(2-amidinopropane)dihydrochloride-induced lipid peroxidation (LPO) of rat liver mitochondria more efficiently than curcumin. The anti-LPO activity of malabaricone C was attributed to its better radical scavenging and Fe(II) chelation capacities. The superior activity of malabaricone C was rationalized by a systematic structure-activity correlation of the results obtained with the structurally related diarylnonanoids and curcumin. Malabaricone C also prevented the gamma-ray-induced damage of pBR322 plasmid DNA in a concentration-dependent manner. The radioprotective activity was found to correlate with its (*)OH radical scavenging property, which matched well with that of d-mannitol.
- Published
- 2005
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.