Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 2.443
Filter
1.
Int J Nanomedicine ; 19: 5381-5395, 2024.
Article in English | MEDLINE | ID: mdl-38859950

ABSTRACT

Background: Current immunotherapies with unexpected severe side effects and treatment resistance have not resulted in the desired outcomes for patients with melanoma, and there is a need to discover more effective medications. Cytotoxin (CTX) from Cobra Venom has been established to have favorable cytolytic activity and antitumor efficacy and is regarded as a promising novel anticancer agent. However, amphiphilic CTX with excellent anionic phosphatidylserine lipid-binding ability may also damage normal cells. Methods: We developed pH-responsive liposomes with a high CTX load (CTX@PSL) for targeted acidic-stimuli release of drugs in the tumor microenvironment. The morphology, size, zeta potential, drug-release kinetics, and preservation stability were characterized. Cell uptake, apoptosis-promoting effects, and cytotoxicity were assessed using MTT assay and flow cytometry. Finally, the tissue distribution and antitumor effects of CTX@PSL were systematically assessed using an in vivo imaging system. Results: CTX@PSL exhibited high drug entrapment efficiency, drug loading, stability, and a rapid release profile under acidic conditions. These nanoparticles, irregularly spherical in shape and small in size, can effectively accumulate at tumor sites (six times higher than free CTX) and are rapidly internalized into cancer cells (2.5-fold higher cell uptake efficiency). CTX@PSL displayed significantly stronger cytotoxicity (IC50 0.25 µg/mL) and increased apoptosis in than the other formulations (apoptosis rate 71.78±1.70%). CTX@PSL showed considerably better tumor inhibition efficacy than free CTX or conventional liposomes (tumor inhibition rate 79.78±5.93%). Conclusion: Our results suggest that CTX@PSL improves tumor-site accumulation and intracellular uptake for sustained and targeted CTX release. By combining the advantages of CTX and stimuli-responsive nanotechnology, the novel CTX@PSL nanoformulation is a promising therapeutic candidate for cancer treatment.


Subject(s)
Antineoplastic Agents , Elapid Venoms , Liposomes , Liposomes/chemistry , Hydrogen-Ion Concentration , Animals , Elapid Venoms/chemistry , Elapid Venoms/pharmacology , Humans , Cell Line, Tumor , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Antineoplastic Agents/pharmacokinetics , Mice , Apoptosis/drug effects , Drug Liberation , Cytotoxins/chemistry , Cytotoxins/pharmacology , Cytotoxins/pharmacokinetics , Drug Delivery Systems/methods , Tissue Distribution , Tumor Microenvironment/drug effects , Nanoparticles/chemistry
2.
Sci Rep ; 14(1): 10561, 2024 05 08.
Article in English | MEDLINE | ID: mdl-38719884

ABSTRACT

This study focuses on understanding the structural and molecular changes in lipid membranes under the influence of six halogenated flavonoid derivatives differing in the number and position of substitution of chlorine and bromine atoms (D1-D6). Utilizing various analytical techniques, including fluorometric methods, dynamic light scattering (DLS), attenuated Fourier transform infrared spectroscopy (ATR- FTIR), and FT-Raman spectroscopy, the research aims to elucidate the mechanisms underlying the interaction of flavonoids with cell membranes. Additionally, the study includes in silico analyses to explore the physicochemical properties of these compounds and their potential pharmaceutical applications, along with toxicity studies to assess their effects on cancer, normal, and red blood cells. Our study showed the ability of halogenated derivatives to interact mostly with the outer part of the membrane, especially in the lipid heads region however, some of them were able to penetrate deeper into the membrane and affect the fluidity of hydrocarbon chains. The potential to reduce cancer cell viability, the lack of toxicity towards erythrocytes, and the favourable physicochemical and pharmacokinetic properties suggest these halogenated flavonoids potential candidates for exploring their potential for medical use.


Subject(s)
Flavonoids , Membrane Lipids , Flavonoids/chemistry , Flavonoids/pharmacology , Flavonoids/metabolism , Humans , Membrane Lipids/metabolism , Membrane Lipids/chemistry , Cell Membrane/metabolism , Halogenation , Cytotoxins/chemistry , Cytotoxins/pharmacology , Cytotoxins/metabolism , Erythrocytes/drug effects , Erythrocytes/metabolism , Cell Survival/drug effects , Spectrum Analysis, Raman , Spectroscopy, Fourier Transform Infrared , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemistry , Antineoplastic Agents/metabolism , Cell Line, Tumor
3.
Int J Nanomedicine ; 19: 4163-4180, 2024.
Article in English | MEDLINE | ID: mdl-38751660

ABSTRACT

Purpose: The study aimed to address the non-specific toxicity of cytotoxins (CTX) in liver cancer treatment and explore their combined application with the photosensitizer Ce6, co-loaded into carbonized Zn/Co bimetallic organic frameworks. The goal was to achieve controlled CTX release and synergistic photodynamic therapy, with a focus on evaluating anti-tumor activity against human liver cancer cell lines (Hep G2). Methods: Purified cobra cytotoxin (CTX) and photosensitizer Ce6 were co-loaded into carbonized Zn/Co bimetallic organic frameworks, resulting in RGD-PDA@C-ZIF@(CTX+Ce6). The formulation was designed with surface-functionalization using polydopamine and tumor-penetrating peptide RGD. This approach aimed to facilitate controlled CTX release and enhance the synergistic effect of photodynamic therapy. The accumulation of RGD-PDA@C-ZIF@(CTX+Ce6) at tumor sites was achieved through RGD's active targeting and the enhanced permeability and retention (EPR) effect. In the acidic tumor microenvironment, the porous structure of the metal-organic framework disintegrated, releasing CTX and Ce6 into tumor cells. Results: Experiments demonstrated that RGD-PDA@C-ZIF@(CTX+Ce6) nanoparticles, combined with near-infrared laser irradiation, exhibited optimal anti-tumor effects against human liver cancer cells. The formulation showcased heightened anti-tumor activity without discernible systemic toxicity. Conclusion: The study underscores the potential of utilizing metal-organic frameworks as an efficient nanoplatform for co-loading cytotoxins and photodynamic therapy in liver cancer treatment. The developed formulation, RGD-PDA@C-ZIF@(CTX+Ce6), offers a promising avenue for advancing the clinical application of cytotoxins in oncology, providing a solid theoretical foundation for future research and development.


Subject(s)
Indoles , Liver Neoplasms , Metal-Organic Frameworks , Photochemotherapy , Photosensitizing Agents , Zinc , Humans , Photochemotherapy/methods , Metal-Organic Frameworks/chemistry , Metal-Organic Frameworks/pharmacology , Liver Neoplasms/drug therapy , Zinc/chemistry , Zinc/pharmacology , Indoles/chemistry , Indoles/pharmacology , Indoles/administration & dosage , Photosensitizing Agents/pharmacology , Photosensitizing Agents/chemistry , Photosensitizing Agents/administration & dosage , Animals , Hep G2 Cells , Cobalt/chemistry , Cobalt/pharmacology , Oligopeptides/chemistry , Oligopeptides/pharmacology , Oligopeptides/pharmacokinetics , Polymers/chemistry , Mice , Cytotoxins/chemistry , Cytotoxins/pharmacology , Cytotoxins/pharmacokinetics , Mice, Nude , Mice, Inbred BALB C , Cell Survival/drug effects
4.
Appl Biochem Biotechnol ; 196(1): 160-181, 2024 Jan.
Article in English | MEDLINE | ID: mdl-37103736

ABSTRACT

Snake venoms are a potential source of bioactive peptides, which have multiple therapeutic properties in treating diseases such as diabetes, cancer, and neurological disorders. Among bioactive peptides, cytotoxins (CTXs) and neurotoxins are low molecular weight proteins belonging to the three-finger-fold toxins (3FTxs) family composed of two ß sheets that are stabilized by four to five conserved disulfide bonds containing 58-72 amino acid residues. These are highly abundant in snake venom and are predicted to have insulinotropic activities. In this study, the CTXs were purified from Indian cobra snake venom using preparative HPLC and characterized using high-resolution mass spectrometry (HRMS) TOF-MS/MS. Further SDS-PAGE analysis confirmed the presence of low molecular weight cytotoxic proteins. The CTXs in fractions A and B exhibited dose-dependent insulinotropic activity from 0.001 to 10 µM using rat pancreatic beta-cell lines (RIN-5F) in the ELISA. Nateglinide and repaglinide are synthetic small-molecule drugs that control sugar levels in the blood in type 2 diabetes, which were used as a positive control in ELISA. Concluded that purified CTXs have insulinotropic activity, and there is a scope to use these proteins as small molecules to stimulate insulinotropic activities. At this stage, the focus is on the efficiency of the cytotoxins to induce insulin. Additional work is ongoing on animal models to see the extent of the beneficial effects and efficiency to cure diabetes using streptozotocin-induced models.


Subject(s)
Diabetes Mellitus, Type 2 , Elapid Venoms , Rats , Animals , Elapid Venoms/chemistry , Elapid Venoms/toxicity , Naja naja , Cytotoxins/pharmacology , Tandem Mass Spectrometry , Peptides
5.
Biophys J ; 122(20): 4068-4081, 2023 10 17.
Article in English | MEDLINE | ID: mdl-37740492

ABSTRACT

Plasma membrane-induced protein folding and conformational transitions play a central role in cellular homeostasis. Several transmembrane proteins are folded in the complex lipid milieu to acquire a specific structure and function. Bacterial pore forming toxins (PFTs) are proteins expressed by a large class of pathogenic bacteria that exploit the plasma membrane environment to efficiently undergo secondary structure changes, oligomerize, and form transmembrane pores. Unregulated pore formation causes ion imbalance, leading to cell death and infection. Determining the free energy landscape of these membrane-driven-driven transitions remains a challenging problem. Although cholesterol recognition is required for lytic activity of several proteins in the PFT family of toxins, the regulatory role of cholesterol for the α-PFT, cytolysin A expressed by Escherichia coli remains unexplained. In a recent free energy computation, we showed that the ß tongue, a critical membrane-inserted motif of the ClyA toxin, has an on-pathway partially unfolded intermediate that refolds into the helix-turn-helix motif of the pore state. To understand the molecular role played by cholesterol, we carry out string-method-based computations in membranes devoid of cholesterol, which reveals an increase of ∼30 times in the free energy barrier for the loss of ß sheet secondary structure when compared with membranes containing cholesterol. Specifically, the tyrosine-cholesterol interaction was found to be critical to creating the unfolded intermediate. Cholesterol also increases the packing and hydrophobicity of the bilayer, resulting in enhanced interactions of the bound protein before complete membrane insertion. Our study illustrates that cholesterol is critical to catalyzing and stabilizing the membrane-inserted unfolded state of the ß tongue motif of ClyA, opening up fresh insights into cholesterol-assisted unfolding of membrane proteins.


Subject(s)
Bacterial Toxins , Escherichia coli , Cell Membrane/metabolism , Escherichia coli/metabolism , Porins/metabolism , Protein Structure, Secondary , Cytotoxins/analysis , Cytotoxins/metabolism , Cytotoxins/pharmacology , Cholesterol/metabolism
6.
Eur J Med Chem ; 259: 115712, 2023 Nov 05.
Article in English | MEDLINE | ID: mdl-37567059

ABSTRACT

Several studies have indicated the potential therapeutic outcomes of combining selective COX-2 inhibitors with tubulin-targeting anticancer agents. In the current study, a novel series of thiazolidin-4-one-based derivatives (7a-q) was designed by merging the pharmacophoric features of some COXs inhibitors and tubulin polymerization inhibitors. Compounds 7a-q were synthesized and evaluated for their cytotoxic activity against MCF7, HT29, and A2780 cancer cell lines (IC50 = 0.02-17.02 µM). The cytotoxicity of 7a-q was also assessed against normal MRC5 cells (IC50 = 0.47-13.46 µM). Compounds 7c, 7i, and 7j, the most active in the MTT assay, significantly reduced the number of HT29 colonies compared to the control. Compounds 7c, 7i, and 7j also induced significant decreases in the tumor volumes and masses in Ehrlich solid carcinoma-bearing mice compared to the control. The three compounds also exhibited significant anti-HT29 migration activity in the wound-healing assay. They have also induced cell cycle arrest in HT29 cells at the S and G2/M phases. In addition, they induced significant increases in both early and late apoptotic events in HT29 cells compared to the control, where 7j showed the highest effect. On the other hand, compound 7j (1 µM) displayed weak inhibitory activity against tubulin polymerization compared to colchicine (3 µM). On the other hand, compounds 7a-q inhibited the activity of COX-2 (IC50 = 0.42-29.11 µM) compared to celecoxib (IC50 = 0.86 µM). In addition, 7c, 7i, and 7j showed moderate inhibition of inflammation in rats compared to indomethacin, with better GIT safety profiles. Molecular docking analysis revealed that 7c, 7i, and 7j have higher binding free energies towards COX-2 than COX-1. These above results suggested that 7j could serve as a potential anticancer drug candidate.


Subject(s)
Antineoplastic Agents , Ovarian Neoplasms , Rats , Mice , Humans , Animals , Female , Cell Line, Tumor , Cytotoxins/pharmacology , Tubulin/metabolism , Molecular Docking Simulation , Cyclooxygenase 2/metabolism , Anti-Inflammatory Agents/pharmacology , Antineoplastic Agents/chemistry , Cyclooxygenase 2 Inhibitors/pharmacology , Cell Proliferation , Drug Screening Assays, Antitumor , Structure-Activity Relationship
7.
Mar Drugs ; 21(7)2023 Jul 13.
Article in English | MEDLINE | ID: mdl-37504931

ABSTRACT

The semisynthesis of renieramycin-type derivatives was achieved under mild and facile conditions by attaching a 1,3-dioxole-bridged phenolic moiety onto ring A of the renieramycin structure and adding a 4'-pyridinecarbonyl ester substituent at its C-5 or C-22 position. These were accomplished through a light-induced intramolecular photoredox reaction using blue light (4 W) and Steglich esterification, respectively. Renieramycin M (4), a bis-tetrahydroisoquinolinequinone compound isolated from the Thai blue sponge (Xestospongia sp.), served as the starting material. The cytotoxicity of the 10 natural and semisynthesized renieramycins against non-small-cell lung cancer (NSCLC) cell lines was evaluated. The 5-O-(4'-pyridinecarbonyl) renieramycin T (11) compound exhibited high cytotoxicity with half-maximal inhibitory concentration (IC50) values of 35.27 ± 1.09 and 34.77 ± 2.19 nM against H290 and H460 cells, respectively. Notably, the potency of compound 11 was 2-fold more than that of renieramycin T (7) and equal to those of 4 and doxorubicin. Interestingly, the renieramycin-type derivatives with a hydroxyl group at C-5 and C-22 exhibited weak cytotoxicity. In silico molecular docking and dynamics studies confirmed that the mitogen-activated proteins, kinase 1 and 3 (MAPK1 and MAPK3), are suitable targets for 11. Thus, the structure-cytotoxicity study of renieramycins was extended to facilitate the development of potential anticancer agents for NSCLC cells.


Subject(s)
Antineoplastic Agents , Carcinoma, Non-Small-Cell Lung , Lung Neoplasms , Humans , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/metabolism , Cytotoxins/pharmacology , Lung Neoplasms/drug therapy , Lung Neoplasms/metabolism , Molecular Docking Simulation , Antineoplastic Agents/chemistry , Cell Line, Tumor , Molecular Structure , Cell Proliferation , Structure-Activity Relationship , Drug Screening Assays, Antitumor
8.
Int J Mol Sci ; 24(9)2023 Apr 28.
Article in English | MEDLINE | ID: mdl-37175727

ABSTRACT

Oncological diseases are difficult to treat even with strong drugs due to development the multidrug resistance (MDR) of cancer cells. A strategy is proposed to increase the efficiency and selectivity of cytotoxic agents against cancer cells to engage the differences in the morphology and microenvironment of tumor and healthy cells, including the pH, membrane permeability, and ion channels. Using this approach, we managed to develop enhanced formulations of cytotoxic agents with adjuvants (which are known as efflux inhibitors and as ion channel inhibitors in tumors)-with increased permeability in A549 and a protective effect on healthy HEK293T cells. The composition of the formulation is as follows: cytotoxic agents (doxorubicin (Dox), paclitaxel (Pac), cisplatin) + adjuvants (allylbenzenes and terpenoids) in the form of inclusion complexes with ß-cyclodextrin. Modified cyclodextrins make it possible to obtain soluble forms of pure substances of the allylbenzene and terpenoid series and increase the solubility of cytotoxic agents. A comprehensive approach based on three methods for studying the interaction of drugs with cells is proposed: MTT test-quantitative identification of surviving cells; FTIR spectroscopy-providing information on the molecular mechanisms inaccessible to study by any other methods (including binding to DNA, surface proteins, or lipid membrane); confocal microscopy for the visualization of observed effects of Dox accumulation in cancer or healthy cells depending on the drug formulation as a direct control of the correctness of interpretation of the results obtained by the two other methods. We found that eugenol (EG) and apiol increase the intracellular concentration of cytostatic in A549 cells by 2-4 times and maintain it for a long time. However, an important aspect is the selectivity of the enhancing effect of adjuvants on tumor cells in relation to healthy ones. Therefore, the authors focused on adjuvant's effect on the control healthy cells (HEK293T): EG and apiol demonstrate "protective" properties from cytostatic penetration by reducing intracellular concentrations by about 2-3 times. Thus, a combined formulation of cytostatic drugs has been found, showing promise in the aspects of improving the efficiency and selectivity of antitumor drugs; thereby, one of the perspective directions for overcoming MDR is suggested.


Subject(s)
Antineoplastic Agents , Cytostatic Agents , Neoplasms , Humans , Terpenes/pharmacology , Cytostatic Agents/pharmacology , Cytotoxins/pharmacology , HEK293 Cells , Drug Resistance, Neoplasm , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemistry , Drug Resistance, Multiple , Doxorubicin/pharmacology , Doxorubicin/chemistry , Plant Extracts/pharmacology , Adjuvants, Immunologic/pharmacology
9.
Eur J Histochem ; 67(2)2023 May 17.
Article in English | MEDLINE | ID: mdl-37195011

ABSTRACT

Metformin can enhance cancer cell chemosensitivity to anticancer drugs. IGF-1R is involved in cancer chemoresistance. The current study aimed to elucidate the role of metformin in osteosarcoma (OS) cell chemosensitivity modulation and identify its underlying mechanism in IGF-1R/miR-610/FEN1 signalling. IGF-1R, miR-610, and FEN1 were aberrantly expressed in OS and participated in apoptosis modulation; this effect was abated by metformin treatment. Luciferase reporter assays confirmed that FEN1 is a direct target of miR-610. Moreover, metformin treatment decreased IGF-1R and FEN1 but elevated miR-610 expression. Metformin sensitised OS cells to cytotoxic agents, while FEN1 overexpression partly compromised metformin's sensitising effects. Furthermore, metformin was observed to enhance adriamycin's effects in a murine xenograft model. Metformin enhanced OS cell sensitivity to cytotoxic agents via the IGF-1R/miR-610/FEN1 signalling axis, highlighting its potential as an adjuvant during chemotherapy.


Subject(s)
Bone Neoplasms , Metformin , MicroRNAs , Osteosarcoma , Humans , Mice , Animals , MicroRNAs/metabolism , Metformin/pharmacology , Metformin/therapeutic use , Osteosarcoma/drug therapy , Bone Neoplasms/drug therapy , Cytotoxins/pharmacology , Cell Proliferation , Cell Line, Tumor , Flap Endonucleases
10.
Int J Mol Sci ; 24(8)2023 Apr 14.
Article in English | MEDLINE | ID: mdl-37108435

ABSTRACT

This study describes, to some extent, the VCC contribution as an early stimulation of the macrophage lineage. Regarding the onset of the innate immune response caused by infection, the ß form of IL-1 is the most important interleukin involved in the onset of the inflammatory innate response. Activated macrophages treated in vitro with VCC induced the activation of the MAPK signaling pathway in a one-hour period, with the activation of transcriptional regulators for a surviving and pro-inflammatory response, suggesting an explanation inspired and supported by the inflammasome physiology. The mechanism of IL-1ß production induced by VCC has been gracefully outlined in murine models, using bacterial knockdown mutants and purified molecules; nevertheless, the knowledge of this mechanism in the human immune system is still under study. This work shows the soluble form of 65 kDa of the Vibrio cholerae cytotoxin (also known as hemolysin), as it is secreted by the bacteria, inducing the production of IL-1ß in the human macrophage cell line THP-1. The mechanism involves triggering the early activation of the signaling pathway MAPKs pERK and p38, with the subsequent activation of (p50) NF-κB and AP-1 (cJun and cFos), determined by real-time quantitation. The evidence shown here supports that the monomeric soluble form of the VCC in the macrophage acts as a modulator of the innate immune response, which is consistent with the assembly of the NLRP3 inflammasome actively releasing IL-1ß.


Subject(s)
NF-kappa B , Vibrio cholerae , Humans , Animals , Mice , NF-kappa B/metabolism , Transcription Factor AP-1/metabolism , Inflammasomes/metabolism , Vibrio cholerae/metabolism , Transcriptional Activation , Cytotoxins/pharmacology , Signal Transduction , Macrophages/metabolism , THP-1 Cells , Interleukin-1beta/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism
11.
Appl Biochem Biotechnol ; 195(12): 7338-7378, 2023 Dec.
Article in English | MEDLINE | ID: mdl-37000353

ABSTRACT

The pathophysiology of lung cancer is dependent on the dysregulation in the apoptotic and autophagic pathways. The intricate link between apoptosis and autophagy through shared signaling pathways complicates our understanding of how lung cancer pathophysiology is regulated. As drug resistance is the primary reason behind treatment failure, it is crucial to understand how cancer cells may respond to different therapies and integrate crosstalk between apoptosis and autophagy in response to them, leading to cell death or survival. Thus, in this study, we have tried to evaluate the crosstalk between autophagy and apoptosis in A549 lung cancer cell line that could be modulated by employing a combination therapy of metformin (6 mM), an anti-diabetic drug, with gedunin (12 µM), an Hsp90 inhibitor, to provide insights into the development of new cancer therapeutics. Our results demonstrated that metformin and gedunin were cytotoxic to A549 lung cancer cells. Combination of metformin and gedunin generated ROS and promoted MMP loss and DNA damage. The combination further increased the expression of AMPKα1 and promoted the nuclear localization of AMPKα1/α2. The expression of Hsp90 was downregulated, further decreasing the expression of its clients, EGFR, PIK3CA, AKT1, and AKT3. Inhibition of the EGFR/PI3K/AKT pathway upregulated TP53 and inhibited autophagy. The combination was promoting nuclear localization of p53; however, some cytoplasmic signals were also detected. Further increase in the expression of caspase 9 and caspase 3 was observed. Thus, we concluded that the combination of metformin and gedunin upregulates apoptosis by inhibiting the EGFR/PI3K/AKT pathway and autophagy in A549 lung cancer cells.


Subject(s)
Antineoplastic Agents , Apoptosis , Autophagy , Limonins , Lung Neoplasms , Metformin , Lung Neoplasms/drug therapy , Lung Neoplasms/metabolism , Humans , A549 Cells , Apoptosis/drug effects , Metformin/pharmacology , Limonins/pharmacology , Antineoplastic Agents/pharmacology , HSP90 Heat-Shock Proteins/antagonists & inhibitors , Lung/drug effects , Cell Line , Cytotoxins/pharmacology , Drug Synergism , Reactive Oxygen Species/metabolism , Drug Combinations , DNA Damage/drug effects , AMP-Activated Protein Kinases/metabolism , Cell Nucleus/metabolism , Signal Transduction/drug effects , Caspase 3/metabolism , Caspase 9/metabolism
12.
J Microbiol Methods ; 207: 106696, 2023 04.
Article in English | MEDLINE | ID: mdl-36898586

ABSTRACT

Cholesterol-dependent cytolysins (CDCs) are proteinaceous toxins widely distributed in gram-positive pathogenic bacteria. CDCs can be classified into three groups (I-III) based on the mode of receptor recognition. Group I CDCs recognize cholesterol as their receptor. Group II CDC specifically recognizes human CD59 as the primary receptor on the cell membrane. Only intermedilysin from Streptococcus intermedius has been reported as a group II CDC. Group III CDCs recognize both human CD59 and cholesterol as receptors. CD59 contains five disulfide bridges in its tertiary structure. Therefore, we treated human erythrocytes with dithiothreitol (DTT) to inactivate CD59 on membranes. Our data showed that DTT treatment caused a complete loss of recognition of intermedilysin and an anti-human CD59 monoclonal antibody. In contrast, this treatment did not affect the recognition of group I CDCs, judging from the fact that DTT-treated erythrocytes were lysed with the same efficiency as mock-treated human erythrocytes. The recognition of group III CDCs toward DTT-treated erythrocytes was partially reduced, and these results are likely due to the loss of human CD59 recognition. Therefore, the degree of human CD59 and cholesterol requirements of uncharacterized group III CDCs frequently found in Mitis group streptococci can be easily estimated by comparing the amounts of hemolysis between DTT-treated and mock-treated erythrocytes.


Subject(s)
Bacterial Toxins , Bacterial Toxins/metabolism , Cytotoxins/pharmacology , Cell Membrane/metabolism , Erythrocytes/metabolism , Cholesterol/chemistry , Cholesterol/metabolism , Cholesterol/pharmacology
13.
Brain Res ; 1807: 148315, 2023 05 15.
Article in English | MEDLINE | ID: mdl-36878343

ABSTRACT

Microglia are the resident immune cells of the brain which regulate both the innate and adaptive neuroimmune responses in health and disease. In response to specific endogenous and exogenous stimuli, microglia transition to one of their reactive states characterized by altered morphology and function, including their secretory profile. A component of the microglial secretome is cytotoxic molecules capable of causing damage and death to nearby host cells, thus contributing to the pathogenesis of neurodegenerative disorders. Indirect evidence from secretome studies and measurements of mRNA expression using diverse microglial cell types suggest different stimuli may induce microglia to secrete distinct subsets of cytotoxins. We demonstrate the accuracy of this hypothesis directly by challenging murine BV-2 microglia-like cells with eight different immune stimuli and assessing secretion of four potentially cytotoxic molecules, including nitric oxide (NO), tumor necrosis factor α (TNF), C-X-C motif chemokine ligand 10 (CXCL10), and glutamate. Lipopolysaccharide (LPS) and a combination of interferon (IFN)-γ plus LPS induced secretion of all toxins studied. IFN-ß, IFN-γ, polyinosinic:polycytidylic acid (poly I:C), and zymosan A upregulated secretion of subsets of these four cytotoxins. LPS and IFN-γ, alone or in combination, as well as IFN-ß induced toxicity of BV-2 cells towards murine NSC-34 neuronal cells, while ATP, N-formylmethionine-leucyl-phenylalanine (fMLP), and phorbol 12-myristate 13-acetate (PMA) did not affect any parameters studied. Our observations contribute to a growing body of knowledge on the regulation of the microglial secretome, which may inform future development of novel therapeutics for neurodegenerative diseases, where dysregulated microglia are key contributors to pathogenesis.


Subject(s)
Microglia , Neurotoxins , Mice , Animals , Microglia/metabolism , Neurotoxins/metabolism , Lipopolysaccharides/pharmacology , Lipopolysaccharides/metabolism , Interferon-gamma/pharmacology , Interferon-gamma/metabolism , Tumor Necrosis Factor-alpha/metabolism , Cytotoxins/metabolism , Cytotoxins/pharmacology
14.
J Biochem Mol Toxicol ; 37(6): e23334, 2023 Jun.
Article in English | MEDLINE | ID: mdl-36843476

ABSTRACT

Levofloxacin, the optical S-(-) isomer of ofloxacin, is a broad-spectrum antibacterial agent widely used to control various infections caused by Gram-positive and Gram-negative bacteria. While the COOH group is necessary for antibacterial activity, its modification can offer anticancer activity to the fluoroquinolone framework. Therefore, several levofloxacin carboxamides 11a-j and 12 containing 5-substituted-1,3,4-thiadiazole residue were synthesized and screened in vitro for their anticancer activity. The in vitro MTT viability assay revealed that the most compounds had significant activity against cancer cells MCF-7, A549, and SKOV3. In particular, the 3-chloro- and 4-fluoro- benzyl derivatives (11b and 11h), with IC50 values of 1.69-4.76 µM were as potent as or better than doxorubicin. It should be noted that the mother quinolone levofloxacin showed no activity on the tested cancer cell lines. The SAR analysis demonstrated that the 3-chloro or 4-fluoro substituent on the S-benzyl moiety had positive effect on the activity. Further in vitro evaluations of the most promising compounds 11b and 11h by flow cytometric analysis and comet test revealed the ability of compounds in the induction of apoptosis and blockage of the cell proliferation at the G1-phase by nuclear fragmentation and DNA degradation in cancer cells. The obtained results demonstrated that the alteration of 6-COOH functional group in the levofloxacin structure and conjugation with a proper heterocyclic pharmacophore is a good strategy to obtain new anticancer agents.


Subject(s)
Antineoplastic Agents , Quinolones , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/chemistry , Levofloxacin/pharmacology , Quinolones/pharmacology , Cytotoxins/pharmacology , Structure-Activity Relationship , Gram-Negative Bacteria , Gram-Positive Bacteria , Cell Proliferation , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemistry , Drug Screening Assays, Antitumor , Molecular Structure
15.
Bioorg Chem ; 133: 106411, 2023 04.
Article in English | MEDLINE | ID: mdl-36801792

ABSTRACT

A new series of 2,3-diaryl-1,3thiazolidin-4-one derivatives was designed, synthesized, and evaluated for their cytotoxicity and COXs inhibitory activities. Among these derivatives, compounds 4 k and 4j exhibited the highest inhibitory activities against COX-2 at IC50 values of 0.05 and 0.06 µM, respectively. Compounds 4a, 4b, 4e, 4 g, 4j, 4 k, 5b, and 6b, which exhibited the highest inhibition% against COX-2, were evaluated for their anti-inflammatory activity in rats. Results showed 41.08-82.00 % inhibition of paw edema thickness by the test compounds compared to celecoxib (inhibition% = 89.51 %). In addition, compounds 4b, 4j, 4 k, and 6b exhibited better GIT safety profiles compared to celecoxib and indomethacin. The four compounds were also evaluated for their antioxidant activity. The results revealed the highest antioxidant activity for 4j (IC50 = 45.27 µM) comparable to torolox (IC50 = 62.03 µM). The antiproliferative activity of the new compounds was evaluated against HePG-2, HCT-116, MCF-7, and PC-3 cancer cell lines. The results showed the highest cytotoxicity for compounds 4b, 4j, 4 k, and 6b (IC50 = 2.31-27.19 µM), with 4j being the most potent. Mechanistic studies revealed the ability of 4j and 4 k by inducing marked apoptosis and cell cycle arrest at the G1 phase in HePG-2 cancer cells. These biological results may also suggest a role for COX-2 inhibition in the antiproliferative activity of these compounds. The results of the molecular docking study for 4 k and 4j into the active site of COX-2 revealed good fitting and correlation with the results of the in vitro COX­2 inhibition assay.


Subject(s)
Antineoplastic Agents , Cytotoxins , Rats , Animals , Celecoxib , Molecular Docking Simulation , Cyclooxygenase 2/metabolism , Thiazolidines/pharmacology , Cytotoxins/pharmacology , Antioxidants/pharmacology , Anti-Inflammatory Agents/pharmacology , Structure-Activity Relationship , Molecular Structure , Antineoplastic Agents/chemistry , Drug Design
16.
Nat Prod Res ; 37(7): 1161-1167, 2023 Apr.
Article in English | MEDLINE | ID: mdl-34720015

ABSTRACT

Highly efficient methodology was developed for the construction of functionalized Kojic acid involving Click linker via 1,3-dipolar cycloaddition and their cytotoxicity against MCF-7, MIAPaCa-2 and DU145 mammalian cell lines were evaluated. Preliminary studies on structure-activity-relationship (SAR) revealed that substitution at C-2 of kojic acid as well as C-5 of 1,2,3-triazole motif played a major role in the activity profile. Kojic acid 1,2,3-triazole analogue 3 b containing an alkyl chain (n = 6) exhibited two fold potent activity than the parent compound, kojic acid against MCF-7 and MIA PaCa-2 cell lines. It induced apoptosis in these cell lines via ID1/PARP1 mediated pathway. The structures of the new analogues of kojic acid 1,2,3-triazole were confirmed by the detailed spectroscopic data analysis.


Subject(s)
Antineoplastic Agents , Cytotoxins , Animals , Molecular Structure , Cytotoxins/pharmacology , Structure-Activity Relationship , Triazoles/chemistry , Antineoplastic Agents/chemistry , Cell Proliferation , Drug Screening Assays, Antitumor , Mammals
17.
Steroids ; 191: 109171, 2023 03.
Article in English | MEDLINE | ID: mdl-36581085

ABSTRACT

Steroidal heterocyclic compounds constitute interesting and promising scaffolds for drug discovery as they have displayed diverse chemical reactivity and several types of biological activities. This study is a concise report on the most recent advancements in the chemistry of the steroid skeleton, including reactions at the A, B, and D ring systems. The modern synthetic methods for the steroidal nitrogen-containing six-membered heterocyclic derivatives from 3-keto-, 6-keto-, 17-keto-, and 20-keto-steroids, as well as 2-Aldo-, 4-Aldo-, 6-Aldo-, and 16-Aldo-steroids, are discussed. However, some other methods for the synthesis of steroidal N-containing 6-membered heterocyclic derivatives are also included. These compounds have shown therapeutic potential as cytotoxic agents against various cell lines and have also shown antiproliferative, anti-inflammatory, and antioxidant activities. Therefore, they could be used as prospective candidates for the development of various medications. This paper not only describes synthetic details involved in creating N-containing 6-membered heterocyclic steroid derivatives, but also provides a brief overview of the medicinal applications of these compounds. This information will be highly useful for the medicinal chemists conducting research in this field.


Subject(s)
Cytotoxins , Nitrogen , Steroids, Heterocyclic , Cytotoxins/chemical synthesis , Cytotoxins/pharmacology , Drug Discovery , Humans , Animals , Cell Line , Steroids, Heterocyclic/chemical synthesis , Steroids, Heterocyclic/pharmacology
18.
Molecules ; 27(19)2022 Oct 09.
Article in English | MEDLINE | ID: mdl-36235247

ABSTRACT

In this study, a series of coumarin derivatives, either alone or as hybrids with cinnamic acid, were synthesized and evaluated for their cytotoxicity against a panel of cancer cells using the MTT assay. Then, the most active compounds were inspected for their mechanism of cytotoxicity by cell-cycle analysis, RT-PCR, DNA fragmentation, and Western blotting techniques. Cytotoxic results showed that compound (4) had a significant cytotoxic effect against HL60 cells (IC50 = 8.09 µM), while compound (8b) had a noticeable activity against HepG2 cells (IC50 = 13.14 µM). Compounds (4) and (8b) mediated their cytotoxicity via PI3K/AKT pathway inhibition. These results were assured by molecular docking studies. These results support further exploratory research focusing on the therapeutic activity of coumarin derivatives as cytotoxic agents.


Subject(s)
Antineoplastic Agents , Neoplasms , Antineoplastic Agents/pharmacology , Apoptosis , Coumarins/pharmacology , Cytotoxins/pharmacology , Drug Screening Assays, Antitumor , HL-60 Cells , Hep G2 Cells , Humans , Molecular Docking Simulation , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction
19.
J Cell Biol ; 221(12)2022 12 05.
Article in English | MEDLINE | ID: mdl-36194176

ABSTRACT

Pore-forming toxins (PFTs) are important virulence factors produced by many pathogenic bacteria. Here, we show that the Vibrio cholerae toxin MakA is a novel cholesterol-binding PFT that induces non-canonical autophagy in a pH-dependent manner. MakA specifically binds to cholesterol on the membrane at pH < 7. Cholesterol-binding leads to oligomerization of MakA on the membrane and pore formation at pH 5.5. Unlike other cholesterol-dependent cytolysins (CDCs) which bind cholesterol through a conserved cholesterol-binding motif (Thr-Leu pair), MakA contains an Ile-Ile pair that is essential for MakA-cholesterol interaction. Following internalization, endosomal acidification triggers MakA pore-assembly followed by ESCRT-mediated membrane repair and V-ATPase-dependent unconventional LC3 lipidation on the damaged endolysosomal membranes. These findings characterize a new cholesterol-binding toxin that forms pores in a pH-dependent manner and reveals the molecular mechanism of host autophagy manipulation.


Subject(s)
Autophagy , Bacterial Proteins , Cholesterol , Cytotoxins , Vibrio cholerae , Virulence Factors , Adenosine Triphosphatases/metabolism , Amino Acid Motifs , Autophagy/drug effects , Bacterial Proteins/chemistry , Bacterial Proteins/metabolism , Cholesterol/metabolism , Cytotoxins/metabolism , Cytotoxins/pharmacology , Endosomal Sorting Complexes Required for Transport/metabolism , Endosomes/chemistry , Endosomes/metabolism , Hydrogen-Ion Concentration , Lysosomes/chemistry , Lysosomes/metabolism , Protein Binding , Vibrio cholerae/chemistry , Vibrio cholerae/metabolism , Virulence Factors/chemistry , Virulence Factors/metabolism
20.
Mar Drugs ; 20(10)2022 Oct 20.
Article in English | MEDLINE | ID: mdl-36286475

ABSTRACT

Seaweeds are a great source of compounds with cytotoxic properties with the potential to be used as anticancer agents. This study evaluated the cytotoxic and proteasome inhibitory activities of 12R-hydroxy-bromosphaerol, 12S-hydroxy-bromosphaerol, and bromosphaerol isolated from Sphaerococcus coronopifolius. The cytotoxicity was evaluated on malignant cell lines (A549, CACO-2, HCT-15, MCF-7, NCI-H226, PC-3, SH-SY5Y, and SK-MEL-28) using the MTT and LDH assays. The ability of compounds to stimulate the production of hydrogen peroxide (H2O2) and to induce mitochondrial dysfunction, the externalization of phosphatidylserine, Caspase-9 activity, and changes in nuclear morphology was also studied on MCF-7 cells. The ability to induce DNA damage was also studied on L929 fibroblasts. The proteasome inhibitory activity was estimated through molecular docking studies. The compounds exhibited IC50 values between 15.35 and 53.34 µM. 12R-hydroxy-bromosphaerol and 12S-hydroxy-bromosphaerol increased the H2O2 levels on MCF-7 cells, and bromosphaerol induced DNA damage on fibroblasts. All compounds promoted a depolarization of mitochondrial membrane potential, Caspase-9 activity, and nuclear condensation and fragmentation. The compounds have been shown to interact with the chymotrypsin-like catalytic site through molecular docking studies; however, only 12S-hydroxy-bromosphaerol evidenced interaction with ALA20 and SER169, key residues of the proteasome catalytic mechanism. Further studies should be outlined to deeply characterize and understand the potential of those bromoditerpenes for anticancer therapeutics.


Subject(s)
Antineoplastic Agents , Neuroblastoma , Rhodophyta , Seaweed , Humans , Proteasome Inhibitors/pharmacology , Hydrogen Peroxide/pharmacology , Cytotoxins/pharmacology , Cell Line, Tumor , Molecular Docking Simulation , Phosphatidylserines/pharmacology , Proteasome Endopeptidase Complex , Caco-2 Cells , Caspase 9 , Chymotrypsin/pharmacology , Rhodophyta/chemistry , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemistry , Apoptosis
SELECTION OF CITATIONS
SEARCH DETAIL
...