Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 47
Filter
Add more filters










Publication year range
1.
Int J Mol Sci ; 25(6)2024 Mar 12.
Article in English | MEDLINE | ID: mdl-38542202

ABSTRACT

Fas-associated death domain (FADD) is an adaptor protein that predominantly transduces the apoptosis signal from the death receptor (DR) to activate caspases, leading to the initiation of apoptotic signaling and the coordinated removal of damaged, infected, or unwanted cells. In addition to its apoptotic functions, FADD is involved in signaling pathways related to autophagy, cell proliferation, necroptosis, and cellular senescence, indicating its versatile role in cell survival and proliferation. The subcellular localization and intracellular expression of FADD play a crucial role in determining its functional outcomes, thereby highlighting the importance of spatiotemporal mechanisms and regulation. Furthermore, FADD has emerged as a key regulator of inflammatory signaling, contributing to immune responses and cellular homeostasis. This review provides a comprehensive summary and analysis of the cellular dynamics of FADD in regulating programmed cell death and inflammation through distinct molecular mechanisms associated with various signaling pathways.


Subject(s)
Apoptosis , Neoplasms , Humans , Death Domain , Fas-Associated Death Domain Protein/metabolism , Apoptosis/physiology , fas Receptor/metabolism , Inflammation , Caspase 8/metabolism
2.
Int J Mol Sci ; 24(3)2023 Feb 03.
Article in English | MEDLINE | ID: mdl-36769293

ABSTRACT

Endocytosis in mammalian cells is a fundamental cellular machinery that regulates vital physiological processes, such as the absorption of metabolites, release of neurotransmitters, uptake of hormone cellular defense, and delivery of biomolecules across the plasma membrane. A remarkable characteristic of the endocytic machinery is the sequential assembly of the complex proteins at the plasma membrane, followed by internalization and fusion of various biomolecules to different cellular compartments. In all eukaryotic cells, functional characterization of endocytic pathways is based on dynamics of the protein complex and signal transduction modules. To coordinate the assembly and functions of the numerous parts of the endocytic machinery, the endocytic proteins interact significantly within and between the modules. Clathrin-dependent and -independent endocytosis, caveolar pathway, and receptor mediated endocytosis have been attributed to a greater variety of physiological and pathophysiological roles such as, autophagy, metabolism, cell division, apoptosis, cellular defense, and intestinal permeabilization. Notably, any defect or alteration in the endocytic machinery results in the development of pathological consequences associated with human diseases such as cancer, cardiovascular diseases, neurological diseases, and inflammatory diseases. In this review, an in-depth endeavor has been made to illustrate the process of endocytosis, and associated mechanisms describing pathological manifestation associated with dysregulated endocytosis machinery.


Subject(s)
Caveolae , Endocytosis , Animals , Humans , Endocytosis/physiology , Caveolae/metabolism , Cell Membrane/metabolism , Signal Transduction , Biological Transport , Mammals
3.
Clin Transl Discov ; 2(2): e60, 2022 Jun.
Article in English | MEDLINE | ID: mdl-35938058

ABSTRACT

Endeavors to identify potentially protective variables for COVID-19 impact on certain populations have remained a priority. Multiple attempts have been made to attribute the reduced COVID-19 impact on populations to their Bacillus-Calmette-Guérin (BCG) vaccination coverage ignoring the fact that the effect of childhood BCG vaccination wanes within 5 years while most of the COVID-19 cases and deaths have occurred in aged with comorbidities. Since the supposed protection being investigated could come from heterologous 'trained immunity' (TI) conferred by exposure to Mycobacterium spp. (i.e., environmental and BCG), it is argued that the estimates of the prevalence of TI in populations currently available as latent tuberculosis infection (LTBI) prevalence would be a better variable to evaluate such assertions. Indeed, when we analyze the European populations (24), and erstwhile East and West Germany populations completely disregarding their BCG vaccination coverage, the populations with higher TI prevalence consistently display reduced COVID-19 impact as compared to their lower TI prevalence neighbors. The TI estimates of the populations not the BCG coverage per se, negatively correlated with pandemic phase-matched COVID-19 incidences (r(24): -0.79 to -0.57; p-value < .004), mortality (r(24): -0.63 to -0.45; p-value < .03), and interim case fatality rates (i-CFR) data. To decisively arrive at dependable conclusions about the potential protective benefit gained from BCG vaccination in COVID-19, the ongoing or planned randomized controlled trials should consciously consider including measures of TI as: (a) all individuals immunized do not respond equally, (b) small study groups from higher background TI could fail to indicate any protective effect.

4.
Apoptosis ; 27(7-8): 606-618, 2022 08.
Article in English | MEDLINE | ID: mdl-35725975

ABSTRACT

Cyclic nucleotide phosphodiesterase 5 (PDE5) has been recently identified to play a crucial role in the progression of many cancers. PDE5 promotes tumorigenesis by dysregulating various cellular processes such as proliferation, apoptosis, angiogenesis, and invasion and migration. Interestingly, multiple studies have reported the promising chemosensitizing potential of PDE5 inhibitor sildenafil in breast, colon, prostate, glioma, and lung cancers. However, to date, the chemosensitizing action of sildenafil is not evaluated in T cell lymphoma, a rare and challenging neoplastic disorder. Hence, the present investigation was undertaken to examine the chemosensitizing potential of sildenafil against T cell lymphoma along with elucidation of possible involvement of altered apoptosis and glucose metabolism. The experimental findings of this study showed that sildenafil enhances the cytotoxic ability of cisplatin by apoptosis induction through altering the levels of apoptosis regulatory molecules: Bcl-2, Bax, cytochrome c (Cyt c), cleaved caspase-3, and poly (ADP-ribose) polymerase (PARP). These molecular alterations were possibly driven by sildenafil through reactive oxygen species (ROS). Sildenafil deregulates glucose metabolism by markedly lowering the expression of glycolysis regulatory molecules, namely glucose transporter 1 (GLUT1), lactate dehydrogenase A (LDHA), hexokinase II (HKII), pyruvate kinase M2 (PKM2), and pyruvate dehydrogenase kinase 1 (PDK1) via suppressing hypoxia-inducible factor 1-alpha (HIF-1α) expression. Hence, sildenafil potentiates the tumor cell killing ability of cisplatin by augmenting ROS production through switching the glucose metabolism from glycolysis to oxidative phosphorylation (OXPHOS). Overall, our study demonstrates that sildenafil might be a promising adjunct therapeutic candidate in designing novel combinatorial chemotherapeutic regimens against T cell lymphoma.


Subject(s)
Cisplatin , Lymphoma, T-Cell , Apoptosis , Cell Line, Tumor , Cisplatin/pharmacology , Glucose/metabolism , Glycolysis , Humans , Lymphoma, T-Cell/metabolism , Male , Phosphodiesterase 5 Inhibitors/pharmacology , Reactive Oxygen Species/metabolism , Sildenafil Citrate/pharmacology
5.
Cancer Rep (Hoboken) ; 5(12): e1291, 2022 12.
Article in English | MEDLINE | ID: mdl-33052041

ABSTRACT

BACKGROUND: The acquisition of resistance to chemotherapy is a major hurdle in the successful application of cancer therapy. Several anticancer approaches, including chemotherapies, radiotherapy, surgery and targeted therapies are being employed for the treatment of cancer. However, cancer cells reprogram themselves in multiple ways to evade the effect of these therapies, and over a period of time, the drug becomes inactive due to the development of multi-drug resistance (MDR). MDR is a complex phenomenon where malignant cells become insensitive to anticancer drugs and attain the ability to survive even after several exposures of anticancer drugs. In this review, we have discussed the molecular and cellular paradigms of multidrug resistance in cancer. RECENT FINDINGS: An Extensive research in cancer biology revealed that drug resistance in cancer is the result of perpetuated intracellular and extracellular mechanisms such as drug efflux, drug inactivation, drug target alteration, oncogenic mutations, altered DNA damage repair mechanism, inhibition of programmed cell death signaling, metabolic reprogramming, epithelial mesenchymal transition (EMT), inherent cell heterogeneity, epigenetic changes, redox imbalance, or any combination of these mechanisms. An inevitable cross-link between inflammation and drug resistance has been discussed. This review provided insight molecular mechanism to understand the vulnerabilities of cancer cells to develop drug resistance. CONCLUSION: MDR is an outcome of interplays between multiple intricate pathways responsible for the inactivation of drug and development of resistance. MDR is a major obstacle in regimens of successful application of anti-cancer therapy. An improved understanding of the molecular mechanism of multi drug resistance and cellular reprogramming can provide a promising opportunity to combat drug resistance in cancer and intensify anti-cancer therapy for the upcoming future.


Subject(s)
Antineoplastic Agents , Neoplasms , Humans , Drug Resistance, Neoplasm/genetics , Drug Resistance, Multiple/genetics , Neoplasms/drug therapy , Neoplasms/genetics , Neoplasms/metabolism , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Drug Delivery Systems
6.
Mol Divers ; 26(2): 869-878, 2022 Apr.
Article in English | MEDLINE | ID: mdl-33646502

ABSTRACT

Pefloxacin-based mixed ligand Cu(II) complexes with substituted isatin of type [Cu(Isatin)(Pefloxacin)Cl] were synthesized, and characterized by EPR, mass, FT-IR, electronic spectrometry, metal content, magnetic moment, and conductance measurement. The g factors g [Formula: see text] > g [Formula: see text] > 2.0023 observed in EPR suggest a square-pyramidal environment of ligands around the copper metal. The compounds were screened for diverse biological activities. The compounds inhibit efficiently the cell proliferation of HCT 116 cancer cells. To take the insight of anticancer activity mechanism, we investigated compound-1 for further cellular assay-based biological activities like trypan blue assay, cell morphological alteration assay, colony formation assay, cell apoptosis, and cell necrosis assay. The compound-1 induced distinct morphological alteration in cells, inhibits cell viability, decreases % plating efficiency, and decreases the clonogenic ability of the HCT 116 cells. The cell death mechanism was confirmed by annexin V-FITC / PI assay and LDH release assay. The positive annexin V/PI stained cells in presence of compound-1 and the absence of a significant amount of lactate dehydrogenase suggest cell apoptosis mechanism for anticancer activity of compounds. We also screened compounds for in vitro antibacterial and cytotoxic activities. Synthesis, characterization, antibacterial, anticancer, and cytotoxicity activities of pefloxacin based Cu(II) complexes were studied. The compound -1 is more potent than standard anticancer drugs and it induced apoptosis to the HCT 116 cells.


Subject(s)
Antineoplastic Agents , Isatin , Anti-Bacterial Agents/chemistry , Antineoplastic Agents/chemistry , Copper/chemistry , Copper/pharmacology , Fluoroquinolones/pharmacology , Humans , Isatin/chemistry , Ligands , Pefloxacin , Spectroscopy, Fourier Transform Infrared
7.
Appl Biochem Biotechnol ; 194(1): 445-463, 2022 Jan.
Article in English | MEDLINE | ID: mdl-34611857

ABSTRACT

Quercetin (QCN) is a plant polyphenol with a variety of medicinal effects. Poor water solubility, on the other hand, restricts its therapeutic effectiveness. The purpose of this study was to develop mixed micellar systems using two biocompatible amphiphilic PEO-PPO-PEO triblock copolymers, Pluronic P123 (EO20-PO70-EO20) and Pluronic F88 (EO104-PO39-EO104), in order to enhance the aqueous solubility and oral bioavailability of QCN drug. The critical micelle concentrations (CMCs) of mixed P123/F88 micellar solutions were investigated using UV-visible spectroscopy with pyrene as a probe. Mixed P123/F88 micelles have low CMCs, indicating that they have a stable micelle structure even when diluted. The solubility of QCN in aqueous mixed P123/F88 micellar solutions at different temperatures was investigated to better understand drug entrapment. The QCN solubility increased with increasing temperature in the mixed P123/F88 micellar system. The QCN-incorporated mixed P123/F88 micelles were prepared using the thin-film hydration method and were well characterized in terms of size and morphology, compatibility, in vitro release and antioxidant profile. In addition, the cell proliferation activity of the mixed micelles was evaluated in the MCF-7 cell line. The QCN-incorporated mixed P123/F88 micelles had a small particle size (< 25 nm) and a negative zeta potential with a spherical shape. The in vitro release behaviour of QCN from a mixed P123/F88 micellar system was slower and more sustained at physiological conditions. The oxidation resistance of QCN-incorporating mixed P123/F88 micelles was shown to be considerably higher than that of pure QCN. An in vitro cell proliferation study revealed that QCN-incorporated mixed micells were effective in inhibiting tumour cell growth. In conclusion, the QCN-incorporated mixed P123/F88 micelle may be a promising approach to increase QCN oral bioavailability, antioxidant activity, and cell viability.


Subject(s)
Cell Proliferation/drug effects , Drug Carriers , Micelles , Polyethylene Glycols/chemistry , Propylene Glycols/chemistry , Quercetin , Drug Carriers/chemistry , Drug Carriers/pharmacology , Humans , MCF-7 Cells , Quercetin/chemistry , Quercetin/pharmacology
8.
Mol Divers ; 26(4): 2085-2101, 2022 Aug.
Article in English | MEDLINE | ID: mdl-34561737

ABSTRACT

Pyrazine-bipyrazole-based µ-oxo bridged dinuclear Au(III) complexes were synthesized and characterized by various spectrometric (1H-NMR, 13C (APT) NMR, FT-IR, Mass spectrometry) and analytical techniques (elemental analysis and conductance measurement). The evaluation of DNA binding activity by UV-Vis absorption spectra and viscosity measurement demonstrated that all the compounds intercalate in between the stacks of DNA base pair and the binding constant values were observed in the range of 5.4 × 104-2.17 × 105 M-1. The molecular docking study also supports the intercalation mode of binding. The anti-proliferation activity of complexes on A549 (Lung adenocarcinoma) cells by MTT assay demonstrated IC50 values in the range of 47.46 -298.12 µg/mL. The genotoxicity of compounds was checked by smearing observed in the DNA of S. pombe cell under the influence of complexes. The in vivo cytotoxicity of compounds against brine shrimp demonstrated the LC50 values in the range of 4.59-27.22 µg/mL. The promising results of the Au(III) complexes received significant attention and make them suitable for the new metallodrugs after the detailed mechanistic biological study.


Subject(s)
Antineoplastic Agents , Coordination Complexes , Antineoplastic Agents/chemistry , Coordination Complexes/chemistry , Coordination Complexes/toxicity , DNA/chemistry , Molecular Docking Simulation , Pyrazines/toxicity , Spectroscopy, Fourier Transform Infrared
9.
J Cancer Res Ther ; 17(6): 1483-1490, 2021.
Article in English | MEDLINE | ID: mdl-34916382

ABSTRACT

OBJECTIVE: The aim of this study was to screen plant extracts for antimitotic activity using Vigna radiata germination inhibition assay, followed by Allium cepa root tip assay and evaluation of their cytotoxic potential on colon carcinoma (HCT-116) cell lines. SUBJECTS AND METHODS: Aqueous extracts of Aconitum heterophyllum, Terminalia bellirica, Bauhinia variegata, Vanda roxburghii, and Cassia angustifolia were prepared by maceration method, and preliminary screening studies to check their antimitotic activity were done by V. radiata germination inhibition assay, followed by A. cepa root tip assay. Furthermore, cytotoxic actions were evaluated by cell proliferation assay. Effect of T. bellirica aqueous extract was analyzed to induce morphological changes, cell death, lactate dehydrogenase release, and cell survival of HCT-116 cells. STATISTICAL ANALYSIS USED: The data represented were analyzed by Student's t-test using SigmaStat 2.0 statistical analysis software. The normality of data was tested by the Shapiro-Wilk test before the Student's t-test. P values *P ≤ 0.05, **P ≤ 0.01, and ***P ≤ 0.001 were considered as statistically significant. RESULTS: All the plant extracts showed promising antimitotic activity. Out of all, T. bellirica was highly effective on HCT-116 cells and promising effect on cell proliferation assay and Annexin-propidium iodide staining revealed that T. bellirica efficiently induces apoptosis. CONCLUSIONS: T. bellirica inhibits cancer cell growth and induces apoptotic cell death. Collectively, it may hold potential for cancer therapeutics.


Subject(s)
Antimitotic Agents/pharmacology , Antineoplastic Agents, Phytogenic/pharmacology , Colonic Neoplasms/drug therapy , Plant Extracts/pharmacology , Aconitum/chemistry , Antimitotic Agents/isolation & purification , Antimitotic Agents/therapeutic use , Antineoplastic Agents, Phytogenic/isolation & purification , Antineoplastic Agents, Phytogenic/therapeutic use , Apoptosis/drug effects , Bauhinia/chemistry , Cell Proliferation/drug effects , Cell Survival/drug effects , Colonic Neoplasms/pathology , Drug Screening Assays, Antitumor , HCT116 Cells , Humans , Orchidaceae/chemistry , Plant Extracts/isolation & purification , Plant Extracts/therapeutic use , Senna Plant/chemistry , Terminalia/chemistry
10.
Article in English | MEDLINE | ID: mdl-34047249

ABSTRACT

Osmium (IV) complexes with pyrazole nucleus containing ligands were synthesized. Os(IV) compounds were characterized using ESI-MS, ICP-OES, IR spectroscopy, electronic spectroscopy, conductance, and magnetic measurements. Whereas, ligands were characterized by heteronuclear spectroscopy, (1H and 13C), IR spectroscopy, and elemental analysis. All the compounds were tested for their potential to interact with HS-DNA by absorption titration, fluorescence spectroscopy, viscosity measurement, and docking study. The quenching constant and Stern Volmer constant values were calculated using fluorescence study. The synthesized compounds were studied for in-vitro bacteriostatic and cytotoxic activities. The cancer cell line studies of all the synthesized complexes were carried out on human lung cancer cells (A549).Supplemental data for this article is available online at https://doi.org/10.1080/15257770.2021.1921795 .


Subject(s)
Coordination Complexes , Osmium , Cell Line, Tumor , DNA/chemistry , Humans , Ligands , Molecular Docking Simulation , Pyrazoles
11.
Biochemistry (Mosc) ; 86(3): 262-274, 2021 Mar.
Article in English | MEDLINE | ID: mdl-33838628

ABSTRACT

Altered expression of cellular redox genes and proteins contributes to invasion, metastasis, and drug resistance in cancer. NADPH oxidase (NOX) isoforms are the pro-oxidant enzymes that generate ROS as a primary product. Dysregulation of NOX activity and expression alters ROS generation, which either directly or indirectly modulates cell death and survival signaling during the progression of cancer. Nuclear factor erythroid 2-related factor 2 (Nrf-2) is an inducible transcription factor, which transcribes an array of antioxidant genes and protects cancer cells from the oxidative stress. Both NOXs and Nrf-2 participate in the regulation of cellular redox homeostasis; but their dysregulation promotes oxidative stress, which contributes to the progression of different types of cancer. Indeed, the role of NOX isoforms and Nrf-2 in developing the drug resistance in cancer is largely unknown. In the present study, we have explored the association of NOX isoforms and Nrf-2 signaling with the MDR1 gene expression in colon carcinoma cells (HCT-116/R). The MDR1 gene was overexpressed to develop resistant HCT-116/R cells and the NOX activation and ROS generation were monitored. We also assessed the role of NOX isoforms and Nrf-2 in the 5-fluorouracil (5-FU) mediated apoptotic cell death of HCT-116/R cells. The HCT-116/R cells demonstrated higher expression of HIF-1α, Nrf-2, and HO-1 and were highly resistant to 5-FU; they also displayed upregulated expression and activity of NOX-2, as well as elevated ROS levels. Interestingly, the treatment with HDC, a specific NOX-2 inhibitor, reduced the ROS levels in HCT-116/R cells. The treatment with HDC and ML-385 (specific inhibitor of Nrf-2) augmented the 5-FU-mediated apoptotic cell death of HCT-116/R cells, which suggests that NOX-2 and Nrf-2 are involved in the development of the chemoresistant phenotype of these cells. Taken together, NOX-2 and Nrf-2 are associated with developing drug resistance of colorectal cancer cells and might be potential targets to overcome drug resistance during cancer therapy.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 1/genetics , Colonic Neoplasms/metabolism , Drug Resistance, Neoplasm , Fluorouracil/pharmacology , NADPH Oxidase 2/metabolism , NF-E2-Related Factor 2/metabolism , Antimetabolites, Antineoplastic/pharmacology , Antimetabolites, Antineoplastic/therapeutic use , Apoptosis , Colonic Neoplasms/drug therapy , Colonic Neoplasms/genetics , Colonic Neoplasms/physiopathology , Fluorouracil/therapeutic use , Gene Expression Regulation, Neoplastic , HCT116 Cells , Humans , Oxidative Stress , Protein Isoforms , Signal Transduction
12.
J Fluoresc ; 31(2): 349-362, 2021 Mar.
Article in English | MEDLINE | ID: mdl-33389418

ABSTRACT

Osmium(IV) pyrazole compounds and ligands were synthesized and well characterised. Ligands were characterized by heteronuclear NMR spectroscopy (1H & 13C), elemental analysis, IR spectroscopy and liquid crystal mass spectroscopy. Os(IV) complexes were characterized by ESI-MS, ICP-OES, IR spectroscopy, conductance measurements, magnetic measurements and electronic spectroscopy. Binding of compounds with HS-DNA were evaluated using viscosity measurements, absorption titration, fluorescence quenching, and molecular docking, which show effective intercalation mode exhibited by compounds. Binding constant of Os(IV) complexes are found to be 8.1 to 9.2 × 104 M-1. Bacteriostatic and cytotoxic activities were carried out to evaluate MIC, LC50, and IC50. The compounds have been undergone bacteriostatic screening using three sets of Gram+ve and two sets of Gram-ve bacteria. MIC of complexes are found to be 72.5-100 µM, whereas that of ligands fall at about 122.5-150 µM.. LC50 count of ligands fall in the range of 16.22-17.28 µg/mL whereas that of complexes of Os(IV) fall in the range of 4.87-5.87 µg/mL. IC50 of osmium compounds were evaluated using HCT-116 cell line. All the Os(IV) compounds show moderate IC50.


Subject(s)
Antineoplastic Agents/pharmacology , Coordination Complexes/pharmacology , DNA/chemistry , Fluorescence , Osmium/pharmacology , Pyrazoles/pharmacology , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Cell Proliferation/drug effects , Cell Survival/drug effects , Coordination Complexes/chemical synthesis , Coordination Complexes/chemistry , Drug Screening Assays, Antitumor , HCT116 Cells , Humans , Ligands , Molecular Docking Simulation , Osmium/chemistry , Pyrazoles/chemistry
13.
J Biomol Struct Dyn ; 39(12): 4555-4562, 2021 Aug.
Article in English | MEDLINE | ID: mdl-32476567

ABSTRACT

The current cancer research focuses on the design and synthesis of chemical compounds that can modulate cell apoptosis or programmed cell death. So we synthesized and characterized ciprofloxacin based copper(II) complexes and studied their anticancer activity against HCT 116 cancer cells by MTT assay. We further investigated the influence of compound-2 (better IC50 value than cisplatin) on cancer cells to know the exact mechanism of anticancer activity. The distinct morphological change of cells due to compound-2 was observed in bright field microscopy. The trypan blue assay clearly demonstrated inhibition of cell viability. The clonogenic ability inhibition assay showed a low percentage of the plating efficiency of HCT 116 cells. The mechanism of cell death, either apoptotic or necrotic was distinguished by annexin V-FITC/PI (propidium iodide) staining assay and LDH (lactate dehydrogenase) release assay. The positive annexinV/PI cells in presence of compound-2 and absence of LDH in the LDH release assay confirmed the cell apoptotic mechanism of cell death. We also checked in vitro antibacterial activity of compounds against Gram(-ve) and Gram(+ve) bacteria in terms of MIC (minimum inhibitory concentration) and the data were in good agreement with the standard drug data. SOD mimic activity of synthesized Cu(II) complexes was also studied in terms of IC50 value. The brine shrimp lethality bioassay was also performed to evaluate the cytotoxic properties of the Cu(II) complexes.Communicated by Ramaswamy H. Sarma.


Subject(s)
Antineoplastic Agents , Coordination Complexes , Anti-Bacterial Agents/pharmacology , Antineoplastic Agents/pharmacology , Apoptosis , Ciprofloxacin/pharmacology , Coordination Complexes/pharmacology , Humans , Microbial Sensitivity Tests , Superoxide Dismutase
14.
Mol Cell Biochem ; 476(2): 585-598, 2021 Feb.
Article in English | MEDLINE | ID: mdl-33025314

ABSTRACT

Advanced glycation end products (AGEs) are formed as a result of non-enzymatic reaction between the free reducing sugars and proteins, lipids, or nucleic acids. AGEs are predominantly synthesized during chronic hyperglycemic conditions or aging. AGEs interact with their receptor RAGE and activate various sets of genes and proteins of the signal transduction pathway. Accumulation of AGEs and upregulated expression of RAGE is associated with various pathological conditions including diabetes, cardiovascular diseases, neurodegenerative disorders, and cancer. The role of AGE-RAGE signaling has been demonstrated in the progression of various types of cancer and other pathological disorders. The expression of RAGE increases manifold during cancer progression. The activation of AGE-RAGE signaling also perturbs the cellular redox balance and modulates various cell death pathways. The programmed cell death signaling often altered during the progression of malignancies. The cellular reprogramming of AGE-RAGE signaling with cell death machinery during tumorigenesis is interesting to understand the complex signaling mechanism of cancer cells. The present review focus on multiple molecular paradigms relevant to cell death particularly Apoptosis, Autophagy, and Necroptosis that are considerably influenced by the AGE-RAGE signaling in the cancer cells. Furthermore, the review also attempts to shed light on the provenience of AGE-RAGE signaling on oxidative stress and consequences of cell survival mechanism of cancer cells.


Subject(s)
Glycation End Products, Advanced/metabolism , Neoplasms/metabolism , Neoplasms/pathology , Receptor for Advanced Glycation End Products/metabolism , Animals , Apoptosis/physiology , Humans
15.
Mol Divers ; 25(2): 687-699, 2021 May.
Article in English | MEDLINE | ID: mdl-32006296

ABSTRACT

N, S donor ligands (L1-L5){L1-L5 = 1,5-bis(4-chlorophenyl)-3-(thiophen-2-yl)-4,5-dihydro-1H-pyrazole (L1), 1-(4-bromophenyl)-5-(4-chlorophenyl)-3-(thiophen-2-yl)-4,5-dihydro-1H-pyrazole (L2), 5-(4-chlorophenyl)-3-(thiophen-2-yl)-1-(p-tolyl)-4,5-dihydro-1H-pyrazole (L3), 5-(4-chlorophenyl)-1-(4-methoxyphenyl)-3-(thiophen-2-yl)-4,5-dihydro-1H-pyrazole (L4), 5-(4-chlorophenyl)-1-(4-nitrophenyl)-3-(thiophen-2-yl)-4,5-dihydro-1H-pyrazole (L5)} were synthesized by Claisen-Schmidt condensation and characterized by spectrometric methods. The complexes (I-V) were synthesized by ligand combination followed by metal chelation. The binding of the rhenium complexes to Herrin sperm DNA was monitored by UV spectroscopy and viscosity measurements. The groove binding was suggested as the most possible mode, and the Kb values of the complexes were calculated. The mode of interaction was furthermore confirmed by molecular docking. Brine shrimp lethality and Saccharomyces cerevisiae cytotoxicity against the eukaryotic and prokaryotic cells showed the toxic nature of the synthesized compounds. All compounds were found active against S. cerevisiae, which was confirmed by increased ROS production, and DNA damage as compared to untreated yeast cell culture. The oxidative harm to cell structures was affirmed by lipid peroxidation. An antimicrobial study was carried out by estimating minimum inhibitory concentration against two Gram-positive and three Gram-negative bacteria. All complexes show good antiproliferative activity against the HCT 116 cell line. All synthesized complexes are biologically more active than the corresponding ligands.


Subject(s)
Anti-Infective Agents , Antineoplastic Agents , Coordination Complexes , Pyrazoles , Rhenium , Animals , Anti-Infective Agents/chemistry , Anti-Infective Agents/pharmacology , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Artemia/drug effects , Bacteria/drug effects , Bacteria/growth & development , Cell Line , Coordination Complexes/chemistry , Coordination Complexes/pharmacology , DNA Damage , DNA, Fungal/drug effects , Humans , Lipid Peroxidation/drug effects , Pyrazoles/chemistry , Pyrazoles/pharmacology , Reactive Oxygen Species/metabolism , Rhenium/chemistry , Rhenium/pharmacology , Saccharomyces cerevisiae/drug effects , Saccharomyces cerevisiae/genetics
16.
J Biomol Struct Dyn ; 39(8): 2894-2903, 2021 May.
Article in English | MEDLINE | ID: mdl-32299292

ABSTRACT

Biological applications of platinum group metal-based complexes have been widely explored in synthetic and inorganic chemistry. The compounds have been subjected to DNA binding, DNA cleavage, In-vivo and In-vitro photocytotoxicity (HCT-116 cell line) and bacteriostatic activities. Binding constant of complexes are 1.42-5.62 × 104 M-1, whereas that of ligands are 1.12-4.72 × 104 M-1. Ksv of complexes are about 1.32-5.21 × 103 M-1, whereas Kf is about 1.24-6.83 × 103 M-1. IC50 of compounds screened using HCT-116 cell line in dark are found to be 121-342 µg/mL. Whereas photocytotoxicity is found in the range of 48-316 µg/mL. Docking energy of molecules have been evaluated to evaluate efficacy of binding. Molecular docking energy of complexes are in the range of -286.00 to -303.11 kJ/mol. Whereas that of ligands are -254.03 to -282.96 kJ/mol. MIC of complexes are 47 ± 2.5 to 77.50 ± 7.5 µM. LC50 values of ligands fall in the range of 4.05-19.72 µg/mL and that of Os(IV) complexes fall in the range of 3.99-15.99 µg/mL. The Os(IV) complexes dominate in proving its potentiality compared to N, N-donor ligands in biological activities. [Formula: see text]Communicated by Ramaswamy H. Sarma.


Subject(s)
Antineoplastic Agents , Coordination Complexes , Quinolines , Coordination Complexes/pharmacology , DNA , DNA Cleavage , Ligands , Molecular Docking Simulation
17.
Environ Toxicol ; 36(4): 628-641, 2021 Apr.
Article in English | MEDLINE | ID: mdl-33274819

ABSTRACT

Nimbolide is a tetranortriterpenoid derived from the leaves and flowers of Azadirachta indica (Neem). It exhibits anticancer activity against a variety of cancers by modulating various crucial features, including cell proliferation, apoptosis, and invasion and metastasis. More importantly, the cytotoxic effect of nimbolide has also been observed against T cell lymphoma, but the underlying mechanisms are still unexplored. So far, no study has been conducted to observe the effect of nimbolide on cancer cell metabolism. Therefore, the present investigation was designed to explore the molecular mechanisms of the antitumor potential of nimbolide against T cell lymphoma, a neoplastic disorder of thymic origin. In addition, we also unraveled the anti-glycolytic activity of nimbolide against T lymphoma cells with possible molecular mechanisms. Our results showed the cytotoxic action of nimbolide against three different cell lines of T cell lymphoma, namely Dalton's lymphoma, HuT-78, and J6. Nimbolide-induced apoptosis in T lymphoma cells by altering the level of reactive oxygen species, p53, Bcl2, Bax, and cytochrome c, with subsequent cleavage of caspase 3. Remarkably, nimbolide inhibited the expression of hypoxia-inducible factor-1α, glucose transporter 3, hexokinase II, and pyruvate dehydrogenase kinase 1, which led to the suppression of glycolysis with concomitant activation of oxidative phosphorylation. Hence, the results of the present investigation demonstrate that nimbolide exerts tumoricidal activity against T lymphoma cells via augmentation of apoptosis and reversal of altered cell metabolism. Thus, the present study provides a new insight for the therapeutic utilization of nimbolide against T cell lymphoma.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Apoptosis/drug effects , Cell Proliferation/drug effects , Glucose/metabolism , Limonins/pharmacology , Caspase 3/metabolism , Cell Culture Techniques , Cell Line, Tumor , Cell Survival/drug effects , Humans , Lymphoma, T-Cell/metabolism , Lymphoma, T-Cell/pathology , Reactive Oxygen Species/metabolism
18.
Cell Biochem Funct ; 39(2): 218-234, 2021 Mar.
Article in English | MEDLINE | ID: mdl-32975319

ABSTRACT

Reactive oxygen species (ROS), formed by the partial reduction of oxygen, were for a long time considered to be a byproduct of cellular metabolism. Since, increase in cellular levels of ROS results in oxidative stress leading to damage of nucleic acids, proteins, and lipids resulting in numerous pathological conditions; ROS was considered a bane for aerobic species. Hence, the discovery of NADPH oxidases (NOX), an enzyme family that specifically generates ROS as its prime product came as a surprise to redox biologists. NOX family proteins participate in various cellular functions including cell proliferation and differentiation, regulation of genes and protein expression, apoptosis, and host defence immunological response. Balanced expression and activation of NOX with subsequent production of ROS are critically important to regulate various genes and proteins to maintain homeostasis of the cell. However, dysregulation of NOX activation leading to enhanced ROS levels is associated with various pathophysiologies including diabetes, cardiovascular diseases, neurodegenerative diseases, ageing, atherosclerosis, and cancer. Although our current knowledge on NOX signifies its importance in the normal functioning of various cellular pathways; yet the choice of ROS producing enzymes which can tip the scale from homeostasis toward damage, as mediators of biological functions remain an oddity. Though the role of NOX in maintaining normal cellular functions is now deemed essential, yet its dysregulation leading to catastrophic events cannot be denied. Hence, this review focuses on the involvement of NOX enzymes in various pathological conditions imploring them as possible targets for therapies. SIGNIFICANCE OF THE STUDY: The NOXs are multi-subunit enzymes that generate ROS as a prime product. NOX generated ROS are usually regulated by various molecular factors and play a vital role in different physiological processes. The dysregulation of NOX activity is associated with pathological consequences. Recently, the dynamic proximity of NOX enzymes with different molecular signatures of pathologies has been studied extensively. It is essential to identify the precise role of NOX machinery in its niche during the progression of pathology. Although inhibition of NOX could be a promising approach for therapeutic interventions, it is critical to expand the current understanding of NOX's dynamicity and shed light on their molecular partners and regulators.


Subject(s)
Cardiovascular Diseases/pathology , NADPH Oxidases/metabolism , Neoplasms/pathology , Acetophenones/therapeutic use , Animals , Cardiovascular Diseases/drug therapy , Cardiovascular Diseases/metabolism , Enzyme Inhibitors/therapeutic use , Humans , Isoenzymes/antagonists & inhibitors , Isoenzymes/classification , Isoenzymes/metabolism , NADPH Oxidases/antagonists & inhibitors , NADPH Oxidases/classification , Neoplasms/drug therapy , Neoplasms/metabolism , Oxidative Stress , Reactive Oxygen Species/metabolism
19.
J Biomol Struct Dyn ; 39(18): 6853-6869, 2021 11.
Article in English | MEDLINE | ID: mdl-32752940

ABSTRACT

To overcome the obstacle of anti-cancer therapy significant attention has been drawn for improving drug delivery system. Since recent past, different approaches were applied using synthetic or natural derivatives for improving efficacy of anti-cancer drugs in cancer therapeutics. Gallic acid (GA) is a natural polyphenol, which exhibits a broad spectrum of biological activities, but its therapeutic application was limited due to poor bioavailability and toxicity. In the present study, we had conjugated the GA with PAMAM dendrimers and proposed the insights of molecular mechanism on inhibition of cell proliferation and programmed cell death through apoptotic pathway in human colon carcinoma cells. GA was chemically conjugated with 4.0 G PAMAM dendrimer at outer surface and characterized by different biophysical methods. We further examined its bioavailability, anti-cancer activity and explored the molecular mechanism of programmed cell death signaling in HCT116 cells. The results show that PAMAM-GA conjugate inhibits cell proliferation of different origin of cancer cells, improves cellular uptake of GA, inhibits colonogenic ability, restricts cancer cell migration by down regulating the expression of MMP-9, inhibits NF-kB activation and release of pro-inflammatory cytokines to manifest apoptotic cell death in HCT 116 cells rather than necrosis. On other hand, PAMAM-GA conjugate showed negligible cytotoxic response as compared to the free Gallic acid to the normal cells. In conclusion, findings of this study revealed that PAMAM-GA conjugate improves the bioavailability of GA and specificity towards cancer cellsto manifests apoptotic cell death. This indispensable approach may be beneficial for the revolution of anti-cancer therapy.Communicated by Ramaswamy H. Sarma.


Subject(s)
Carcinoma , Colonic Neoplasms , Dendrimers , Apoptosis , Cell Movement , Cell Proliferation , Colonic Neoplasms/drug therapy , Gallic Acid/pharmacology , Humans
20.
Int J Mol Sci ; 21(18)2020 Sep 19.
Article in English | MEDLINE | ID: mdl-32961826

ABSTRACT

Dysregulated expression of Fas-associated death domain (FADD) is associated with the impediment of various cellular pathways, including apoptosis and inflammation. The adequate cytosolic expression of FADD is critical to the regulation of cancer cell proliferation. Importantly, cancer cells devise mechanisms to suppress FADD expression and, in turn, escape from apoptosis signaling. Formulating strategies, for direct delivery of FADD proteins into cancer cells in a controlled manner, may represent a promising therapeutic approach in cancer therapy. We chemically conjugated purified FADD protein with cell permeable TAT (transactivator of transcription) peptide, to deliver in cancer cells. TAT-conjugated FADD protein internalized through the caveolar pathway of endocytosis and retained in the cytosol to augment cell death. Inside cancer cells, TAT-FADD rapidly constituted DISC (death inducing signaling complex) assembly, which in turn, instigate apoptosis signaling. The apoptotic competency of TAT-FADD showed comparable outcomes with the conventional apoptosis inducers. Notably, TAT-FADD mitigates constitutive NF-κB activation and associated downstream anti-apoptotic genes Bcl2, cFLIPL, RIP1, and cIAP2, independent of pro-cancerous TNF-α priming. In cancer cells, TAT-FADD suppresses the canonical NLRP3 inflammasome priming and restricts the processing and secretion of proinflammatory IL-1ß. Our results demonstrate that TAT-mediated intracellular delivery of FADD protein can potentially recite apoptosis signaling with simultaneous regulation of anti-apoptotic and proinflammatory NF-κB signaling activation in cancer cells.


Subject(s)
Apoptosis/drug effects , Cell-Penetrating Peptides , Fas-Associated Death Domain Protein , Gene Expression Regulation/drug effects , Neoplasm Proteins/biosynthesis , Neoplasms , Animals , Cell-Penetrating Peptides/chemistry , Cell-Penetrating Peptides/pharmacology , Fas-Associated Death Domain Protein/chemistry , Fas-Associated Death Domain Protein/pharmacology , HCT116 Cells , HEK293 Cells , HeLa Cells , Hep G2 Cells , Humans , MCF-7 Cells , Mice , Neoplasms/drug therapy , Neoplasms/metabolism , Neoplasms/pathology , RAW 264.7 Cells
SELECTION OF CITATIONS
SEARCH DETAIL
...