1
|
Conjugates of Methylene Blue with Cycloalkaneindoles as New Multifunctional Agents for Potential Treatment of Neurodegenerative Disease. Int J Mol Sci 2022; 23:ijms232213925. [PMID: 36430413 PMCID: PMC9697446 DOI: 10.3390/ijms232213925] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2022] [Revised: 11/03/2022] [Accepted: 11/07/2022] [Indexed: 11/16/2022] Open
Abstract
The development of multi-target-directed ligands (MTDLs) would provide effective therapy of neurodegenerative diseases (ND) with complex and nonclear pathogenesis. A promising method to create such potential drugs is combining neuroactive pharmacophoric groups acting on different biotargets involved in the pathogenesis of ND. We developed a synthetic algorithm for the conjugation of indole derivatives and methylene blue (MB), which are pharmacophoric ligands that act on the key stages of pathogenesis. We synthesized hybrid structures and performed a comprehensive screening for a specific set of biotargets participating in the pathogenesis of ND (i.e., cholinesterases, NMDA receptor, mitochondria, and microtubules assembly). The results of the screening study enabled us to find two lead compounds (4h and 4i) which effectively inhibited cholinesterases and bound to the AChE PAS, possessed antioxidant activity, and stimulated the assembly of microtubules. One of them (4i) exhibited activity as a ligand for the ifenprodil-specific site of the NMDA receptor. In addition, this lead compound was able to bypass the inhibition of complex I and prevent calcium-induced mitochondrial depolarization, suggesting a neuroprotective property that was confirmed using a cellular calcium overload model of neurodegeneration. Thus, these new MB-cycloalkaneindole conjugates constitute a promising class of compounds for the development of multitarget neuroprotective drugs which simultaneously act on several targets, thereby providing cognitive stimulating, neuroprotective, and disease-modifying effects.
Collapse
|
2
|
Onder S, Biberoglu K, Yuksel M, Tacal O. Toluidine blue O attenuates tau phosphorylation in N2a-APPSwe cells. Chem Biol Interact 2022; 366:110126. [PMID: 36027949 DOI: 10.1016/j.cbi.2022.110126] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2022] [Revised: 08/15/2022] [Accepted: 08/18/2022] [Indexed: 11/27/2022]
Abstract
Alzheimer's disease (AD) is characterized by extracellular amyloid plaques composed of amyloid-β peptide (Aβ), intracellular neurofibrillary tangles containing hyperphosphorylated tau protein and neuronal loss. Most of the FDA-approved AD drugs currently on the market are cholinesterase inhibitors, which are only effective in relieving the symptoms of AD. However, recent studies in AD drug discovery focus on multi-targeted strategies, including anti-amyloid and anti-tau therapy. In the current study, we have investigated the effects of toluidine blue O (TBO), a cholinesterase inhibitor, on amyloid precursor protein (APP) processing, tau phosphorylation, and tau kinases/phosphatase in N2a mouse neuroblastoma cells stably expressing the Swedish mutation of human APP695 (N2a-APPSwe). The results demonstrated that TBO reduces Aβ40/42 levels by decreasing expression levels of β-secretase 1 (BACE1), presenilin 1 (PS1) and total APP without causing cytotoxic effects in N2a-APPSwe cells. TBO also decreased the levels of both total tau and phosphorylated tau at residues Ser202/Thr205, Thr181, Ser396 and Ser 396/Ser404. Moreover, when the possible mechanisms underlying its effects on tau pathology were explored, TBO was found to decrease tau phosphorylation at those sites by reducing the expression levels of Akt, GSK-3β, Cdk5, inactive p-PP2A and increasing the expression levels of p-Akt Ser473 and inactive p-GSK-3β Ser9. Our new data support the idea that TBO may be a promising multi-target drug candidate for the treatment of AD.
Collapse
Affiliation(s)
- Seda Onder
- Department of Biochemistry, Faculty of Pharmacy, Hacettepe University, 06100, Ankara, Turkey.
| | - Kevser Biberoglu
- Department of Biochemistry, Faculty of Pharmacy, Hacettepe University, 06100, Ankara, Turkey
| | - Melike Yuksel
- Department of Biochemistry, Faculty of Pharmacy, Hacettepe University, 06100, Ankara, Turkey; Department of Molecular Medicine, Morsani College of Medicine and Byrd Alzheimer's Center and Research Institute, University of South Florida, Tampa, FL, 33613, USA
| | - Ozden Tacal
- Department of Biochemistry, Faculty of Pharmacy, Hacettepe University, 06100, Ankara, Turkey.
| |
Collapse
|
3
|
BIBEROGLU K. Kinetics of human butyrylcholinesterase inhibition by 1,9-dimethyl-methylene blue. JOURNAL OF THE TURKISH CHEMICAL SOCIETY, SECTION A: CHEMISTRY 2021. [DOI: 10.18596/jotcsa.853598] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022] Open
|
4
|
Inhibition of cholinesterases by safranin O: Integration of inhibition kinetics with molecular docking simulations. Arch Biochem Biophys 2020; 698:108728. [PMID: 33345803 DOI: 10.1016/j.abb.2020.108728] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2020] [Revised: 12/03/2020] [Accepted: 12/08/2020] [Indexed: 12/11/2022]
Abstract
In the present study, the inhibitory mechanisms and effects of a synthetic phenazine dye, safranin O (SO) on human plasma butyrylcholinesterase (BChE), human erythrocyte acetylcholinesterase (AChE) and recombinant BChE mutants were investigated. Kinetic studies showed the following information: SO leaded to linear competitive inhibition of human plasma BChE with Ki = 0.44 ± 0.085 μM; α = ∞. It acted as a hyperbolic noncompetitive inhibitor of human erythrocyte AChE with Ki = 0.69 ± 0.13; α = 1; β = 0.08 ± 0.02. On the other hand, the inhibitory effects of SO on two BChE mutants, where A328 was modified to either F or Y, revealed differences in terms of inhibitory patterns and Ki values, compared to the obtained results with recombinant wild type BChE. SO was found to act as a linear competitive inhibitor of A328F and A328Y BChE mutants. Compared to recombinant wild type BChE, A328Y and A328F BChE mutants caused a 4- and 10-fold decrease in Ki value for SO, respectively. These findings were supported by molecular modelling studies. In conclusion, SO is a potent inhibitor of human cholinesterases and may be useful in the design and development of new drugs for the treatment of AD.
Collapse
|
5
|
Phenothiazine‐based chalcones as potential dual‐target inhibitors toward cholinesterases (AChE, BuChE) and monoamine oxidases (MAO‐A, MAO‐B). J Heterocycl Chem 2020. [DOI: 10.1002/jhet.4156] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
6
|
Onder S, Biberoglu K, Tacal O. The kinetics of inhibition of human acetylcholinesterase and butyrylcholinesterase by methylene violet 3RAX. Chem Biol Interact 2019; 314:108845. [DOI: 10.1016/j.cbi.2019.108845] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2019] [Revised: 09/26/2019] [Accepted: 10/04/2019] [Indexed: 02/03/2023]
|
7
|
ElZorkany HE, Youssef T, Mohamed MB, Amin RM. Photothermal versus photodynamic treatment for the inactivation of the bacteria Escherichia coli and Bacillus cereus: An in vitro study. Photodiagnosis Photodyn Ther 2019; 27:317-326. [PMID: 31252144 DOI: 10.1016/j.pdpdt.2019.06.020] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2019] [Revised: 06/18/2019] [Accepted: 06/24/2019] [Indexed: 02/08/2023]
Abstract
The widespread occurrence of microbial pathogens, including multidrug-resistant (MDR) bacteria, has ignited research efforts to discover alternative strategies to combat infections in patients. Recently, photodynamic therapy (PDT) and photothermal therapy (PTT) have been proposed for the inactivation of pathogens. Although PDT and PTT are very promising antipathogenic tools, further effort is needed to determine their real impact on pathogens apart from the effects of individual elements involved in the photodynamic/photothermal processes, i.e., light, photosensitizers (PSs), and nanoparticles. Accordingly, in the current study, toluidine blue O (TBO) and gold nanoparticles (GNP) were used as generators of reactive oxygen species (ROS) and hyperthermia in the presence of light, respectively. Escherichia coli (E. coli) and Bacillus cereus (B. cereus) bacteria were chosen as examples of gram-negative and gram-positive bacteria, respectively. Before the bactericidal activity of PDT was assessed, the aggregation of TBO and its effect on the growth of both strains of bacteria were studied. Additionally, E. coli and B. cereus were exposed to a range of doses of 633 nm helium-neon laser light to investigate its effect. In a separate set of experiments, the bactericidal activity of PTT was assessed after the effects of GNP and green light (530 nm) had been assessed. The results showed that PDT and PTT should be considered useful tools for bacterial eradication even when the light, PSs, and nanoparticles are each used at doses safe for bacterial growth. Moreover, different photodynamic responses were observed for E. coli and B. cereus, and light from a 633 nm laser and a 530 nm light-emitting diode (LED) showed disparate responses when applied alone to both bacteria.
Collapse
Affiliation(s)
- Heba ElSayed ElZorkany
- Nanotechnology and Advanced Materials Central Lab, Agriculture Research Center, El Gamaa St., Giza, Egypt; National Institute of Laser Enhanced Sciences, Cairo University, Giza, Egypt.
| | - Tareq Youssef
- National Institute of Laser Enhanced Sciences, Cairo University, Giza, Egypt
| | - Mona B Mohamed
- National Institute of Laser Enhanced Sciences, Cairo University, Giza, Egypt
| | - Rehab M Amin
- National Institute of Laser Enhanced Sciences, Cairo University, Giza, Egypt
| |
Collapse
|
8
|
Conjugates of methylene blue with γ-carboline derivatives as new multifunctional agents for the treatment of neurodegenerative diseases. Sci Rep 2019; 9:4873. [PMID: 30890752 PMCID: PMC6424957 DOI: 10.1038/s41598-019-41272-4] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2018] [Accepted: 03/05/2019] [Indexed: 02/08/2023] Open
Abstract
We studied the inhibitory activity of methylene blue (MB) γ-carbolines (gC) conjugates (MB-gCs) against human erythrocyte acetylcholinesterase (AChE), equine serum butyrylcholinesterase (BChE), and a structurally related enzyme, porcine liver carboxylesterase (CaE). In addition, we determined the ability of MB-gCs to bind to the peripheral anionic site (PAS) of Electrophorus electricus AChE (EeAChE) and competitively displace propidium iodide from this site. Moreover, we examined the ability of MB-gCs to scavenge free radicals as well as their influence on mitochondrial potential and iron-induced lipid peroxidation. We found that MB-gCs effectively inhibited AChE and BChE with IC50 values in the range 1.73–10.5 μM and exhibited low potencies against CaE (9.8–26% inhibition at 20 μM). Kinetic studies showed that MB-gCs were mixed-type reversible inhibitors of both cholinesterases. Molecular docking results showed that the MB-gCs could bind both to the catalytic active site and to the PAS of human AChE and BChE. Accordingly, MB-gCs effectively displaced propidium from the peripheral anionic site of EeAChE. In addition, MB-gCs were extremely active in both radical scavenging tests. Quantum mechanical DFT calculations suggested that free radical scavenging was likely mediated by the sulfur atom in the MB fragment. Furthermore, the MB-gCs, in like manner to MB, can restore mitochondrial membrane potential after depolarization with rotenone. Moreover, MB-gCs possess strong antioxidant properties, preventing iron-induced lipid peroxidation in mitochondria. Overall, the results indicate that MB-gCs are promising candidates for further optimization as multitarget therapeutic agents for neurodegenerative diseases.
Collapse
|
9
|
Optimization of hydrogel containing toluidine blue O for photodynamic therapy in treating acne. Lasers Med Sci 2019; 34:1535-1545. [PMID: 30825010 DOI: 10.1007/s10103-019-02727-2] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2018] [Accepted: 01/17/2019] [Indexed: 01/23/2023]
Abstract
Antibiotics and photodynamic therapy (PDT) are widely employed in curing acne. However, antibiotics as an effective treatment would lead to bacterial resistance and severe side effects. In this study, we aimed to develop a novel TBO hydrogel, which could prolong the retention time of photosensitizer (TBO) at the lesion site and improve therapeutic effect. In vitro antibacterial experiments (against Staphylococcus aureus and Escherichia coli), the response surface methodology was used to optimize the formulation of TBO hydrogel. The results indicated that the optimal formulation was 0.5% (v/v) carbomer, 0.01 mg/mL TBO, 0.5% (v/v) ethanol concentration, 0.5% (v/v) Tween 80, the mass ratio of NaOH to carbomer of 0.4 (w/w). The TBO hydrogel formulation showed the strong antibacterial activity for Propionibacterium acnes. The stability, pH, and antibacterial activity of TBO hydrogel did not significantly change under 4 °C, 25 °C, and 40 °C during 6-week storage. Furthermore, TBO combined with carbomer hydrogel showed the 51.28% (4 h) and 69.80% (24 h) release. In summary, the hydrogel TBO might be a vital therapeutic strategy to promote the PDT applied in the topical therapy of acne. Graphical abstract A TBO hydrogel for photodynamic therapy in the treatment of acne.
Collapse
|
10
|
Azure B affects amyloid precursor protein metabolism in PS70 cells. Chem Biol Interact 2019; 299:88-93. [PMID: 30500345 DOI: 10.1016/j.cbi.2018.11.023] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2018] [Revised: 10/22/2018] [Accepted: 11/27/2018] [Indexed: 01/01/2023]
Abstract
Alzheimer's disease (AD), the most common form of dementia, is characterized by abundant deposition of amyloid-β (Aβ) peptide that is the result of sequential cleavage of amyloid precursor protein (APP) by β-secretase and γ-secretase. Several studies have documented that inhibition of Aβ peptide synthesis or facilitating its degradation is one of the attractive therapeutic strategies in AD. Methylene blue (MethB), which has recently been investigated in Phase II clinical trials, is a prominent inhibitor in reducing Aβ oligomers. Herein, we wonder whether the mitigating effects of MethB on amyloid metabolism are related to the activity of its major metabolite, azure B. The goal of this study was to investigate the effects of azure B, which is also a cholinesterase inhibitor, on APP processing by using Chinese hamster ovary cells stably expressing human wild-type APP and presenilin 1 (PS70). Azure B significantly decreased the levels of secreted APPα (sAPPα) and Aβ40/42 in culture medium with a dose-dependent manner. A significant decrease was also observed in the levels of intracellular APP without affecting the cell viability. In parallel with the decrease of APP and APP metabolites, the activity of β-secretase 1 (BACE1) was significantly attenuated compared to control. Overall, our results show that azure B has a large contribution for the pharmacological profile of MethB in APP metabolism.
Collapse
|
11
|
Yuksel M, Biberoglu K, Onder S, Akbulut KG, Tacal O. Toluidine blue O modifies hippocampal amyloid pathology in a transgenic mouse model of Alzheimer's disease. Biochimie 2018; 146:105-112. [DOI: 10.1016/j.biochi.2017.12.004] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2017] [Accepted: 12/12/2017] [Indexed: 01/04/2023]
|
12
|
Yuksel M, Biberoglu K, Onder S, Akbulut KG, Tacal O. Effects of phenothiazine-structured compounds on APP processing in Alzheimer's disease cellular model. Biochimie 2017; 138:82-89. [DOI: 10.1016/j.biochi.2017.04.012] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2017] [Accepted: 04/25/2017] [Indexed: 01/04/2023]
|