1
|
Ghosh A, Banik S, Takahama K, Nishida Y, Honda M. Formulation and physicochemical characterization of astaxanthin-loaded liposomes: A comparative study of free and ester forms with different E/Z-isomer ratios of astaxanthin. Food Res Int 2025; 212:116448. [PMID: 40382042 DOI: 10.1016/j.foodres.2025.116448] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2024] [Revised: 03/18/2025] [Accepted: 04/15/2025] [Indexed: 05/20/2025]
Abstract
The present study aimed to developed astaxanthin (AST)-loaded liposomes in different forms (free and ester forms) and with varying E/Z-isomer ratios and compare their physicochemical properties. Four liposomes, i.e., liposomes of free-form all-E- and Z-isomer-rich AST (E- and Z-AST-Free-L, respectively) and ester-form all-E- and Z-isomer-rich AST (E- and Z-AST-Ester-L, respectively) were prepared using soybean lecithin and cholesterol via thin-film dispersion technique, and encapsulation efficiency of AST, E/Z-isomer ratio of encapsulated AST, morphology, release characteristics, and storage stability were investigated. All liposomes exhibited multilamellar structures with high encapsulation efficiency (> 98 %). Under simulated intestinal conditions, E- and Z-AST-Ester-L, especially Z-AST-Ester-L, demonstrated higher AST release compared to E- and Z-AST-Free-L. Storage tests at 5 and 30 °C revealed that E- and Z-AST-Ester-L and E-AST-Free-L showed high storage stability, whereas Z-AST-Free-L exhibited comparatively low stability. These observations provide valuable insights into the production of AST-loaded liposomes with enhanced health benefits and practical applications.
Collapse
Affiliation(s)
- Antara Ghosh
- Department of Chemistry, Faculty of Science & Technology, Meijo University, 1-501 Shiogamaguchi, Tempaku-ku, Nagoya, Aichi 468-8502, Japan
| | - Sujan Banik
- Department of Pharmacy, Noakhali Science and Technology University, Sonapur, Noakhali 3814, Bangladesh
| | - Kentaro Takahama
- Technical Center, Nagoya University, Furo-cho, Nagoya, Aichi 464-8601, Japan
| | - Yasuhiro Nishida
- Fuji Chemical Industries, Co., Ltd., Kamiich-machi, Nakaniikawa-gun, Toyama, Japan
| | - Masaki Honda
- Department of Chemistry, Faculty of Science & Technology, Meijo University, 1-501 Shiogamaguchi, Tempaku-ku, Nagoya, Aichi 468-8502, Japan; Graduate School of Environmental and Human Sciences, Meijo University, 1-501 Shiogamaguchi, Tempaku-ku, Nagoya, Aichi 468-8502, Japan.
| |
Collapse
|
2
|
Jiang Y, Chen H, Xu J, Le J, Rong W, Zhu Z, Chen Y, Hu C, Cai J, Hong Y, Huang S, Zheng M, Zhang X, Zhou C, Zhang J, He S, Yan X, Cui W. Long-term fucoxanthin treatment prevents cognitive impairments and neuroinflammation via the inhibition of Nogo-A in APP/PS1 transgenic mice. Food Funct 2025; 16:3891-3903. [PMID: 40272460 DOI: 10.1039/d4fo05034g] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/25/2025]
Abstract
Alzheimer's disease (AD) is a progressive neurodegenerative disorder characterized by neuroinflammation and cognitive impairments. Although short-term treatment with fucoxanthin, a marine carotenoid with anti-neuroinflammatory activity, has been reported to prevent cognitive impairments in scopolamine- and β-amyloid (Aβ)-treated mice, it remains uncertain whether long-term fucoxanthin treatment could produce similar effects in transgenic AD models. Moreover, the anti-neuroinflammatory mechanism of fucoxanthin is still unclear. In this study, long-term treatment with fucoxanthin (15-150 mg kg-1, twice a week for 20 weeks) significantly prevented cognitive deficits and Aβ-related neuroinflammation in APP/PS1 transgenic mice. In addition, fucoxanthin largely prevented Aβ oligomer-induced secretion of pro-inflammatory cytokines and the activation of BV2 microglial cells. Furthermore, fucoxanthin reduced the increased expression of Nogo-A, a central player in AD pathophysiology, as well as the activation of downstream Rho-associated protein kinase 2 (ROCK2) and nuclear factor kappa-B (NF-κB) pathways in AD models. Most importantly, the inhibition of neuroinflammation by fucoxanthin was not reduced by shRNA-mediated knockdown of Nogo-A, suggesting that fucoxanthin significantly prevented cognitive impairments and neuroinflammation via the inhibition of Nogo-A. These results not only elucidate an anti-neuroinflammatory mechanism of fucoxanthin, but also provide strong support for the development of fucoxanthin as a novel food ingredient or drug for the treatment of AD.
Collapse
Affiliation(s)
- Yujie Jiang
- Translational Medicine Center of Pain, Emotion and Cognition, Health Science Center, Ningbo University, Ningbo, 315211, China.
| | - Huiyue Chen
- Translational Medicine Center of Pain, Emotion and Cognition, Health Science Center, Ningbo University, Ningbo, 315211, China.
| | - Jiayi Xu
- Translational Medicine Center of Pain, Emotion and Cognition, Health Science Center, Ningbo University, Ningbo, 315211, China.
| | - Jingyang Le
- Translational Medicine Center of Pain, Emotion and Cognition, Health Science Center, Ningbo University, Ningbo, 315211, China.
| | - Wenni Rong
- Translational Medicine Center of Pain, Emotion and Cognition, Health Science Center, Ningbo University, Ningbo, 315211, China.
| | - Zengyu Zhu
- Translational Medicine Center of Pain, Emotion and Cognition, Health Science Center, Ningbo University, Ningbo, 315211, China.
| | - Yuan Chen
- Translational Medicine Center of Pain, Emotion and Cognition, Health Science Center, Ningbo University, Ningbo, 315211, China.
| | - Chenwei Hu
- Translational Medicine Center of Pain, Emotion and Cognition, Health Science Center, Ningbo University, Ningbo, 315211, China.
| | - Jinhan Cai
- Translational Medicine Center of Pain, Emotion and Cognition, Health Science Center, Ningbo University, Ningbo, 315211, China.
| | - Yirui Hong
- Translational Medicine Center of Pain, Emotion and Cognition, Health Science Center, Ningbo University, Ningbo, 315211, China.
| | - Shangwei Huang
- Translational Medicine Center of Pain, Emotion and Cognition, Health Science Center, Ningbo University, Ningbo, 315211, China.
| | - Meilin Zheng
- Translational Medicine Center of Pain, Emotion and Cognition, Health Science Center, Ningbo University, Ningbo, 315211, China.
| | - Xinyu Zhang
- Translational Medicine Center of Pain, Emotion and Cognition, Health Science Center, Ningbo University, Ningbo, 315211, China.
| | - Chenhui Zhou
- The First Affiliated Hospital of Ningbo University, Ningbo, 315211, China
| | - Jinrong Zhang
- School of Food and Pharmaceutical Sciences, Ningbo University, Ningbo, 315211, China
| | - Shan He
- Translational Medicine Center of Pain, Emotion and Cognition, Health Science Center, Ningbo University, Ningbo, 315211, China.
| | - Xiaojun Yan
- Marine Science and Technical College, Zhejiang Ocean University, Zhoushan, 316022, China.
| | - Wei Cui
- Translational Medicine Center of Pain, Emotion and Cognition, Health Science Center, Ningbo University, Ningbo, 315211, China.
- The First Affiliated Hospital of Ningbo University, Ningbo, 315211, China
- Ningbo Kangning Hospital, Ningbo, 315211, China
| |
Collapse
|
3
|
Iraji A, Hariri R, Hashempur MH, Ghasemi M, Pourtaher H, Saeedi M, Akbarzadeh T. Design and synthesis of new 1,2,3-triazole-methoxyphenyl-1,3,4-oxadiazole derivatives: selective butyrylcholinesterase inhibitors against Alzheimer's disease. BMC Chem 2025; 19:97. [PMID: 40234998 PMCID: PMC11998406 DOI: 10.1186/s13065-025-01475-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2024] [Accepted: 04/04/2025] [Indexed: 04/17/2025] Open
Abstract
Alzheimer's disease (AD) remains a significant public health challenge due to its progressive cognitive impairment and the absence of proven treatments. In this study, several novel 1,2,3-triazole-methoxyphenyl-1,3,4-oxadiazole derivatives were synthesized and evaluated for their ability to inhibit key enzymes associated with AD: acetylcholinesterase (AChE) and butyrylcholinesterase (BChE). Structure-activity relationship (SAR) analysis revealed that derivatives featuring electron-withdrawing groups, particularly nitro and fluorine substituents, exhibited remarkable inhibitory activity against BChE while showing minimal effectiveness against AChE. Among these, compound 13s (R = 4-CH3, R' = 4-NO2) demonstrated the highest potency, selectively targeting BChE with an IC50 value of 11.01 µM. Molecular docking and molecular dynamics (MD) simulations provided deeper insights into the favorable interactions between these compounds and BChE. Additionally, cytotoxicity studies confirmed the active compound's limited toxicity toward normal cells, indicating a promising therapeutic profile. These findings suggest that the synthesized selective anti-BChE compounds hold potential for consideration in the later stages of AD treatment.
Collapse
Affiliation(s)
- Aida Iraji
- Stem Cells Technology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
- Department of Persian Medicine, School of Medicine, Research Center for Traditional Medicine and History of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Roshanak Hariri
- Department of Medicinal Chemistry, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Mohammad Hashem Hashempur
- Department of Persian Medicine, School of Medicine, Research Center for Traditional Medicine and History of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Mahshad Ghasemi
- Stem Cells Technology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Hormoz Pourtaher
- Department of Chemistry, Faculty of Sciences, Persian Gulf University, Bushehr, 75169, Iran
| | - Mina Saeedi
- Medicinal Plants Research Center, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran.
- Persian Medicine and Pharmacy Research Center, Tehran University of Medical Sciences, Tehran, Iran.
| | - Tahmineh Akbarzadeh
- Department of Medicinal Chemistry, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran.
- Persian Medicine and Pharmacy Research Center, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
4
|
Oliyaei N, Zekri S, Iraji A, Oliyaei A, Tanideh R, Mussin NM, Tamadon A, Tanideh N. Health benefits of algae and marine-derived bioactive metabolites for modulating ulcerative colitis symptoms. J Funct Foods 2025; 125:106690. [DOI: 10.1016/j.jff.2025.106690] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/30/2025] Open
|
5
|
Le NHT, Park SA, Kim YM, Ahn DK, Jung W, Han SK. Fucoxanthin Inhibits the NMDA and AMPA Receptors Through Regulating the Calcium Response on Substantia Gelatinosa Neurons of the Trigeminal Subnucleus Caudalis in Juvenile Mice. Neural Plast 2025; 2025:2553040. [PMID: 39949835 PMCID: PMC11824308 DOI: 10.1155/np/2553040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2024] [Accepted: 01/08/2025] [Indexed: 02/16/2025] Open
Abstract
Glutamate excitotoxicity is considered as the etiology of stroke and neurodegenerative diseases, namely, Parkinson's disease (PD), Alzheimer's disease (AD), and others. Meanwhile, substantia gelatinosa (SG) neurons of the trigeminal subnucleus caudalis (Vc), a pivotal site in regulating orofacial nociceptive transmission via Aδ and C primary afferent fibers, majorly utilize glutamate as the principal excitatory neurotransmitter. Fucoxanthin (FCX), a carotenoid pigment extracted from brown seaweed, possesses various pharmaceutical properties including neuroprotective effect in multiple neuronal populations. To date, the direct activity of FCX on the SG of the Vc has not been extensively clarified. Consequently, we investigated the effect of FCX on excitatory signaling mediated by ionotropic glutamate receptors (iGluRs), using the patch-clamp technique recorded from SG neurons of the Vc. Here, FCX directly acted on glutamate receptors independent of voltage-gated sodium channel and γ-aminobutyric acid (GABA)A/glycine receptors in the voltage-clamp mode. Specifically, the N-methyl-D-aspartic acid (NMDA)- and α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA)-induced responses but not the kainic acid receptor (KAR)-mediated response were suppressed by FCX in standard extracellular solution. Additionally, the inhibitory effect of FCX on NMDA currents was repeatable and concentration-dependent. The FCX blockade of NMDA-mediated excitotoxicity was associated with the modulation of Ca2+ response without affecting Na+ ions. The Ca2+-dependent fluorescence intensity of brain slice was reduced in the presence of FCX. Notably, FCX significantly attenuated the spontaneous firing activity of SG neurons. Altogether, these results reveal that FCX may protect SG neurons against glutamate excitotoxicity via primarily regulating Ca2+ response, thereby inhibiting the excitatory signaling induced by NMDA and AMPA receptors (AMPARs).
Collapse
Affiliation(s)
- Nhung Ha Thuy Le
- Department of Oral Physiology, School of Dentistry and Institute of Oral Bioscience, Jeonbuk National University, Jeonju, Republic of Korea
- Faculty of Odonto-Stomatology, Hue University of Medicine and Pharmacy, Hue University, Hue, Vietnam
| | - Seon Ah Park
- Department of Oral Physiology, School of Dentistry and Institute of Oral Bioscience, Jeonbuk National University, Jeonju, Republic of Korea
| | - Yu Mi Kim
- Department of Oral Physiology, School of Dentistry, Kyungpook National University, Daegu, Republic of Korea
| | - Dong Kuk Ahn
- Department of Oral Physiology, School of Dentistry, Kyungpook National University, Daegu, Republic of Korea
| | - Won Jung
- Department of Oral Medicine, School of Dentistry and Institute of Oral Bioscience, Jeonbuk National University; Research Institute of Clinical Medicine of Jeonbuk National University—Biomedical Research Institute of Jeonbuk National University Hospital, Jeonju 54896, Republic of Korea
| | - Seong Kyu Han
- Department of Oral Physiology, School of Dentistry and Institute of Oral Bioscience, Jeonbuk National University, Jeonju, Republic of Korea
| |
Collapse
|
6
|
Shokouhi Asl AS, Sayahi MH, Hashempur MH, Irajie C, Alaeddini AH, Ghafouri SN, Noori M, Dastyafteh N, Mottaghipisheh J, Asadi M, Larijani B, Mahdavi M, Iraji A. Cinnamic acid conjugated with triazole acetamides as anti-Alzheimer and anti-melanogenesis candidates: an in vitro and in silico study. Sci Rep 2025; 15:655. [PMID: 39754023 PMCID: PMC11698978 DOI: 10.1038/s41598-024-83020-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2024] [Accepted: 12/10/2024] [Indexed: 01/06/2025] Open
Abstract
In this study, new cinnamic acid linked to triazole acetamide derivatives was synthesized and evaluated for anti-Alzheimer and anti-melanogenesis activities. The structural elucidation of all analogs was performed using different analytical techniques, including 1H-NMR, 13C-NMR, mass spectrometry, and IR spectroscopy. The synthesized compounds were assessed in vitro for their inhibitory activities against acetylcholinesterase (AChE), butyrylcholinesterase (BChE), and tyrosinase enzymes. Among synthesize derivative compound 3-(4-((1-(2-((2,4-dichlorophenyl)amino)-2-oxoethyl)-1H-1,2,3-triazol-4-yl)methoxy)-3-methoxyphenyl)acrylic acid (10j) exhibited the highest activity against BChE with an IC50 value of 11.99 ± 0.53 µM. Derivative 3-(3-methoxy-4-((1-(2-oxo-2-(p-tolylamino)ethyl)-1H-1,2,3-triazol-4-yl)methoxy)phenyl)acrylic acid (10d), bearing a 4-CH3 group, was identified as the most potent AChE inhibitor. In terms of tyrosinase inhibition, 3-(3-methoxy-4-((1-(2-((2-methyl-4-nitrophenyl)amino)-2-oxoethyl)-1H-1,2,3-triazol-4-yl)methoxy)phenyl)acrylic acid (compound 10n), demonstrated 44.87% inhibition at a concentration of 40 µM. Additionally, a kinetic study of compound 10j which 2,4-dichlorophenyl substituents against BChE revealed a mixed-type inhibition pattern. Furthermore, molecular docking and molecular dynamic studies of compound 10j were conducted to thoroughly evaluate its mode of action within the BChE active site.
Collapse
Affiliation(s)
- Amir Shervin Shokouhi Asl
- Department of Medicinal Chemistry, School of Pharmacy, Iran University of Medical Sciences, Tehran, Iran
| | | | - Mohammad Hashem Hashempur
- Research Center for Traditional Medicine and History of Medicine, Department of Persian Medicine, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Cambyz Irajie
- Department of Medical Biotechnology, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran
| | | | | | - Milad Noori
- Endocrinology and Metabolism Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Navid Dastyafteh
- Endocrinology and Metabolism Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Javad Mottaghipisheh
- Department of Aquatic Sciences and Assessment, Swedish University of Agricultural Sciences, 7050, SE-750 07, Uppsala, Sweden
| | - Mehdi Asadi
- Department of Medicinal Chemistry, School of Pharmacy, Iran University of Medical Sciences, Tehran, Iran
| | - Bagher Larijani
- Department of Medicinal Chemistry, School of Pharmacy, Iran University of Medical Sciences, Tehran, Iran
| | - Mohammad Mahdavi
- Endocrinology and Metabolism Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran.
| | - Aida Iraji
- Stem Cells Technology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran.
| |
Collapse
|
7
|
Noori M, Khalili Ghomi M, Dastyafteh N, Oliyaei N, Hamedifar H, Javanshir S, Tanideh N, Sattarinezhad E, Sattari F, Haghani M, Rahmani H, Larijani B, Mahdavi M, Hajimiri MH, Iraji A. Isoindolinedione-Benzamide Pyridinium Derivatives for Targeting Alzheimer's Disease. ACS OMEGA 2024; 9:48032-48043. [PMID: 39676969 PMCID: PMC11635510 DOI: 10.1021/acsomega.4c04027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/04/2024] [Revised: 10/26/2024] [Accepted: 11/05/2024] [Indexed: 12/17/2024]
Abstract
An Isoindolinedione-benzamide pyridinium derivatives were designed through a structure-based strategy and synthesized as novel multifunctional anti-Alzheimer agents. The inhibitory activities of all 17 derivatives against acetylcholinesterase and butyrylcholinesterase were evaluated. Results exhibited that compound 7j displayed promising AChE inhibitory activity with an IC50 value of 0.26 ± 0.07 μM, and compound 7c exhibited an IC50 value of 0.08 ± 0.01 μM against BChE with 132-fold better inhibitory activity in comparison with positive control. Next, the enzyme kinetics studies and detailed binding mode via molecular docking were performed for the most potent compounds. Additionally, molecular dynamics simulations were accomplished to further investigate the potent compound's interaction, orientation, and conformation over the related enzymes. The neurotoxicity of the most potent derivative was executed against SH-SY5Y, and the mRNA levels of GSK-3α and GSK-3β after treatment with 7c on SH-SY5Y were evaluated. Results exhibited the mRNA levels of GSK-3β were decreased compared to the control group. All these results indicate that 7c is a good starting point for developing a multifunctional anti-Alzheimer compound.
Collapse
Affiliation(s)
- Milad Noori
- Pharmaceutical
and Heterocyclic Chemistry Research Laboratory, Department of Chemistry, Iran University of Science and Technology, Tehran 16846-13114, Iran
| | - Minoo Khalili Ghomi
- Endocrinology
and Metabolism Research Center, Endocrinology
and Metabolism Clinical Sciences Institute, Tehran University of Medical
Sciences, Tehran 1416634793, Iran
| | - Navid Dastyafteh
- Pharmaceutical
and Heterocyclic Chemistry Research Laboratory, Department of Chemistry, Iran University of Science and Technology, Tehran 16846-13114, Iran
| | - Najmeh Oliyaei
- Stem
Cells Technology Research Center, Shiraz
University of Medical Sciences, Shiraz 71348-14336, Iran
| | - Haleh Hamedifar
- CinnaGen
Medical Biotechnology Research Center, Alborz
University of Medical Sciences, Karaj 1461965381, Iran
- CinnaGen
Research and Production Co., Alborz 3164819712, Iran
| | - Shahrzad Javanshir
- Pharmaceutical
and Heterocyclic Chemistry Research Laboratory, Department of Chemistry, Iran University of Science and Technology, Tehran 16846-13114, Iran
| | - Nader Tanideh
- Stem
Cells Technology Research Center, Shiraz
University of Medical Sciences, Shiraz 71348-14336, Iran
| | - Elahe Sattarinezhad
- Department
of Pharmacology, School of Medicine, Shiraz
University of Medical Sciences, Shiraz 71348-14336, Iran
| | - Fateme Sattari
- Student
Research Committee, Shiraz University of
Medical Sciences, Shiraz 71348-14336, Iran
| | - Masoud Haghani
- Department
of Physiology, The Medical School, Shiraz
University of Medical Sciences, Shiraz 71348-14336, Iran
| | - Hojjat Rahmani
- Department
of Health Management, Policy and Economics, School of Public Health, Tehran University of Medical Sciences, Tehran 1416634793, Iran
| | - Bagher Larijani
- Endocrinology
and Metabolism Research Center, Endocrinology
and Metabolism Clinical Sciences Institute, Tehran University of Medical
Sciences, Tehran 1416634793, Iran
| | - Mohammad Mahdavi
- Endocrinology
and Metabolism Research Center, Endocrinology
and Metabolism Clinical Sciences Institute, Tehran University of Medical
Sciences, Tehran 1416634793, Iran
| | - Mir H. Hajimiri
- CinnaGen
Research and Production Co., Alborz 3164819712, Iran
- CinnaGen
Medical Biotechnology Research Center, Alborz
University of Medical Sciences, Karaj 1461965381, Iran
| | - Aida Iraji
- Research
Center for Traditional Medicine and History of Medicine, Department
of Persian Medicine, School of Medicine, Shiraz University of Medical Sciences, Shiraz 7134845794, Iran
| |
Collapse
|
8
|
Palanisamy M, Ramalingam S. Microbial Bacterioruberin: A Comprehensive Review. Indian J Microbiol 2024; 64:1477-1501. [PMID: 39678945 PMCID: PMC11645389 DOI: 10.1007/s12088-024-01312-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2024] [Accepted: 05/11/2024] [Indexed: 12/17/2024] Open
Abstract
Bacterioruberin (BR) is a fat-soluble, dipolar, reddish pigment predominantly found in halophilic archaea. BR is a rare C50 carotenoid from the xanthophyll family, and it has been extensively studied for its potent antioxidant properties, such as its ability to protect cells from oxidative stress. In addition, several studies have shown that BR-rich extracts and its derivatives exhibit significant antiviral, antidiabetic, antibacterial, and anti-inflammatory effects, making them ideal candidates for the development of novel therapeutic interventions against various diseases. Although it possesses remarkable biological properties, studies related to the regulatory aspects of biosynthesis, in vitro and in vivo studies of purified BR have been rare. However, investigations are needed to explore the potential application of BR in various industries. Additionally, optimization of the culture conditions of BR-producing haloarchaea could pave the way for their sustainable production and utilization. The current review provides comprehensive information on BR, which includes the sources of this compound and its bioproduction, extraction, stability, toxicity, and biological activities in relation to its commercial applications. This review also discusses the potential challenges and limitations associated with BR bioproduction and its utilization in various industries. In addition, this treatise highlights the need for further research to optimize production and extraction methods and explore avenues for novel applications of BR in various sectors, such as pharmaceuticals, food, and cosmetics. Graphical Abstract
Collapse
Affiliation(s)
- Mouliraj Palanisamy
- Plant Genetic Engineering Laboratory, Department of Biotechnology, Bharathiar University, Coimbatore, India
| | - Sathishkumar Ramalingam
- Plant Genetic Engineering Laboratory, Department of Biotechnology, Bharathiar University, Coimbatore, India
| |
Collapse
|
9
|
Iraji A, Nikfar P, Nazari Montazer M, Karimi M, Edraki N, Saeedi M, Mirfazli SS. Synthesis, biological evaluation and molecular modeling studies of methyl indole-isoxazole carbohydrazide derivatives as multi-target anti-Alzheimer's agents. Sci Rep 2024; 14:21115. [PMID: 39256495 PMCID: PMC11387822 DOI: 10.1038/s41598-024-71729-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2023] [Accepted: 08/30/2024] [Indexed: 09/12/2024] Open
Abstract
Alzheimer's disease (AD) is a progressive neurodegenerative disorder that affects the elderly population globally and there is an urgent demand for developing novel anti-AD agents. In this study, a new series of indole-isoxazole carbohydrazides were designed and synthesized. The structure of all compounds was elucidated using spectroscopic methods including FTIR, 1H NMR, and 13C NMR as well as mass spectrometry and elemental analysis. All derivatives were screened for their acetylcholinesterase (AChE) and butyrylcholinesterase (BuChE) inhibitory activity. Out of all synthesized compounds, compound 5d exhibited the highest potency as AChE inhibitor with an IC50 value of 29.46 ± 0.31 µM. It showed significant selectivity towards AChE, with no notable inhibition against BuChE. A kinetic study on AChE for compound 5d indicated a competitive inhibition pattern. Also, 5d exhibited promising BACE1 inhibitory potential with an IC50 value of 2.85 ± 0.09 µM and in vitro metal chelating ability against Fe3+. The molecular dynamic studies of 5d against both AChE and BACE1 were executed to evaluate the behavior of this derivative in the binding site. The results showed that the new compounds deserve further chemical optimization to be considered potential anti-AD agents.
Collapse
Affiliation(s)
- Aida Iraji
- Department of Persian Medicine, School of Medicine, Research Center for Traditional Medicine and History of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
- Stem Cells Technology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Parisa Nikfar
- Department of Medicinal Chemistry, School of Pharmacy, Iran University of Medical Sciences, Tehran, Iran
| | - Mohammad Nazari Montazer
- Endocrinology and Metabolism Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Mona Karimi
- Department of Chemistry, North Tehran Branch, Islamic Azad University, Tehran, Iran
| | - Najmeh Edraki
- Medicinal and Natural Products Chemistry Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Mina Saeedi
- Medicinal Plants Research Center, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran.
- Persian Medicine and Pharmacy Research Center, Tehran University of Medical Sciences, Tehran, Iran.
| | - Seyedeh Sara Mirfazli
- Department of Medicinal Chemistry, School of Pharmacy, Iran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
10
|
Flieger J, Forma A, Flieger W, Flieger M, Gawlik PJ, Dzierżyński E, Maciejewski R, Teresiński G, Baj J. Carotenoid Supplementation for Alleviating the Symptoms of Alzheimer's Disease. Int J Mol Sci 2024; 25:8982. [PMID: 39201668 PMCID: PMC11354426 DOI: 10.3390/ijms25168982] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2024] [Revised: 08/12/2024] [Accepted: 08/16/2024] [Indexed: 09/03/2024] Open
Abstract
Alzheimer's disease (AD) is characterized by, among other things, dementia and a decline in cognitive performance. In AD, dementia has neurodegenerative features and starts with mild cognitive impairment (MCI). Research indicates that apoptosis and neuronal loss occur in AD, in which oxidative stress plays an important role. Therefore, reducing oxidative stress with antioxidants is a natural strategy to prevent and slow down the progression of AD. Carotenoids are natural pigments commonly found in fruits and vegetables. They include lipophilic carotenes, such as lycopene, α- and β-carotenes, and more polar xanthophylls, for example, lutein, zeaxanthin, canthaxanthin, and β-cryptoxanthin. Carotenoids can cross the blood-brain barrier (BBB) and scavenge free radicals, especially singlet oxygen, which helps prevent the peroxidation of lipids abundant in the brain. As a result, carotenoids have neuroprotective potential. Numerous in vivo and in vitro studies, as well as randomized controlled trials, have mostly confirmed that carotenoids can help prevent neurodegeneration and alleviate cognitive impairment in AD. While carotenoids have not been officially approved as an AD therapy, they are indicated in the diet recommended for AD, including the consumption of products rich in carotenoids. This review summarizes the latest research findings supporting the potential use of carotenoids in preventing and alleviating AD symptoms. A literature review suggests that a diet rich in carotenoids should be promoted to avoid cognitive decline in AD. One of the goals of the food industry should be to encourage the enrichment of food products with functional substances, such as carotenoids, which may reduce the risk of neurodegenerative diseases.
Collapse
Affiliation(s)
- Jolanta Flieger
- Department of Analytical Chemistry, Medical University of Lublin, Chodźki 4a, 20-093 Lublin, Poland
| | - Alicja Forma
- Department of Forensic Medicine, Medical University of Lublin, ul. Jaczewskiego 8b, 20-090 Lublin, Poland; (A.F.); (M.F.); (G.T.)
| | - Wojciech Flieger
- Department of Plastic Surgery, St. John’s Cancer Center, ul. Jaczewskiego 7, 20-090 Lublin, Poland; (W.F.)
| | - Michał Flieger
- Department of Forensic Medicine, Medical University of Lublin, ul. Jaczewskiego 8b, 20-090 Lublin, Poland; (A.F.); (M.F.); (G.T.)
| | - Piotr J. Gawlik
- Department of Plastic Surgery, St. John’s Cancer Center, ul. Jaczewskiego 7, 20-090 Lublin, Poland; (W.F.)
| | - Eliasz Dzierżyński
- Department of Plastic Surgery, St. John’s Cancer Center, ul. Jaczewskiego 7, 20-090 Lublin, Poland; (W.F.)
| | - Ryszard Maciejewski
- Institute of Health Sciences, John Paul II Catholic University of Lublin, Konstantynów 1 H, 20-708 Lublin, Poland;
| | - Grzegorz Teresiński
- Department of Forensic Medicine, Medical University of Lublin, ul. Jaczewskiego 8b, 20-090 Lublin, Poland; (A.F.); (M.F.); (G.T.)
| | - Jacek Baj
- Department of Correct, Clinical and Imaging Anatomy, Medical University of Lublin, ul. Jaczewskiego 4, 20-090 Lublin, Poland;
| |
Collapse
|
11
|
Shareghi-Boroujeni D, Iraji A, Dara M, Hashempur MH, Zare S, Hariri R, Akbarzadeh T, Saeedi M. Synthesis of novel hybrids of 1,2,3-triazoles-hydrazone: targeting cholinesterases and Alzheimer's related genes. Future Med Chem 2024; 16:1519-1535. [PMID: 38864182 PMCID: PMC11370907 DOI: 10.1080/17568919.2024.2359894] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2024] [Accepted: 05/13/2024] [Indexed: 06/13/2024] Open
Abstract
Aim: A new series of 1,2,3-triazole-hydrazone derivatives were developed to evaluate their anti-Alzheimer's activity. Materials & methods: All compounds were screened toward cholinesterases via the modified Ellman's method. The toxicity assay on SH-SY5Y cells was performed using the MTT assay, and the expression levels of GSK-3α, GSK-3β, DYRK1 and CDK5 were assessed in the presence of compounds 6m and 6p.Results:6m and 6p; acting as mixed-type inhibitors, exhibited promising acetylcholinesterase and butyrylcholinesterase inhibitory activity, respectively. 6m demonstrated no toxicity under tested concentrations on the SH-SY5Y cells and positively impacted neurodegenerative pathways. Notably, 6m displayed a significant downregulation in mRNA levels of GSK-3α, GSK-3β and CDK5.Conclusion: The target compounds could be considered in developing anti-Alzheimer's disease agents.
Collapse
Affiliation(s)
- Diba Shareghi-Boroujeni
- Medicinal Plants Research Center, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Aida Iraji
- Research Center for Traditional Medicine & History of Medicine, Department of Persian Medicine, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
- Stem Cells Technology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Mahintaj Dara
- Stem Cells Technology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Mohammad Hashem Hashempur
- Research Center for Traditional Medicine & History of Medicine, Department of Persian Medicine, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Shahrokh Zare
- Stem Cells Technology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Roshanak Hariri
- Department of Medicinal Chemistry, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Tahmineh Akbarzadeh
- Department of Medicinal Chemistry, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
- Persian Medicine & Pharmacy Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Mina Saeedi
- Medicinal Plants Research Center, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
- Persian Medicine & Pharmacy Research Center, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
12
|
Guardado Yordi E, Pérez Martínez A, Radice M, Scalvenzi L, Abreu-Naranjo R, Uriarte E, Santana L, Matos MJ. Seaweeds as Source of Bioactive Pigments with Neuroprotective and/or Anti-Neurodegenerative Activities: Astaxanthin and Fucoxanthin. Mar Drugs 2024; 22:327. [PMID: 39057436 PMCID: PMC11277739 DOI: 10.3390/md22070327] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2024] [Revised: 07/14/2024] [Accepted: 07/19/2024] [Indexed: 07/28/2024] Open
Abstract
The marine kingdom is an important source of a huge variety of scaffolds inspiring the design of new drugs. The complex molecules found in the oceans present a great challenge to organic and medicinal chemists. However, the wide variety of biological activities they can display is worth the effort. In this article, we present an overview of different seaweeds as potential sources of bioactive pigments with activity against neurodegenerative diseases, especially due to their neuroprotective effects. Along with a broad introduction to seaweed as a source of bioactive pigments, this review is especially focused on astaxanthin and fucoxanthin as potential neuroprotective and/or anti-neurodegenerative agents. PubMed and SciFinder were used as the main sources to search and select the most relevant scientific articles within the field.
Collapse
Affiliation(s)
- Estela Guardado Yordi
- Universidad Estatal Amazónica, 160101 Puyo, Ecuador; (E.G.Y.); (A.P.M.); (M.R.); (L.S.); (R.A.-N.)
| | - Amaury Pérez Martínez
- Universidad Estatal Amazónica, 160101 Puyo, Ecuador; (E.G.Y.); (A.P.M.); (M.R.); (L.S.); (R.A.-N.)
| | - Matteo Radice
- Universidad Estatal Amazónica, 160101 Puyo, Ecuador; (E.G.Y.); (A.P.M.); (M.R.); (L.S.); (R.A.-N.)
| | - Laura Scalvenzi
- Universidad Estatal Amazónica, 160101 Puyo, Ecuador; (E.G.Y.); (A.P.M.); (M.R.); (L.S.); (R.A.-N.)
| | - Reinier Abreu-Naranjo
- Universidad Estatal Amazónica, 160101 Puyo, Ecuador; (E.G.Y.); (A.P.M.); (M.R.); (L.S.); (R.A.-N.)
| | - Eugenio Uriarte
- Departamento de Química Orgánica, Facultad de Farmacia, Universidade de Santiago de Compostela, 15782 Santiago de Compostela, Spain; (E.U.); (L.S.)
| | - Lourdes Santana
- Departamento de Química Orgánica, Facultad de Farmacia, Universidade de Santiago de Compostela, 15782 Santiago de Compostela, Spain; (E.U.); (L.S.)
| | - Maria Joao Matos
- Departamento de Química Orgánica, Facultad de Farmacia, Universidade de Santiago de Compostela, 15782 Santiago de Compostela, Spain; (E.U.); (L.S.)
| |
Collapse
|
13
|
Loori S, Pourtaher H, Mehranpour A, Hasaninejad A, Eftekharian M, Iraji A. Synthesis of novel aryl-substituted 2-aminopyridine derivatives by the cascade reaction of 1,1-enediamines with vinamidinium salts to develop novel anti-Alzheimer agents. Sci Rep 2024; 14:13780. [PMID: 38877034 PMCID: PMC11178820 DOI: 10.1038/s41598-024-64179-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2024] [Accepted: 06/05/2024] [Indexed: 06/16/2024] Open
Abstract
Alzheimer's disease (AD), a severe neurodegenerative disorder, imposes socioeconomic burdens and necessitates innovative therapeutic strategies. Current therapeutic interventions are limited and underscore the need for novel inhibitors of acetylcholinesterase (AChE) and butyrylcholinesterase (BChE), enzymes implicated in the pathogenesis of AD. In this study, we report a novel synthetic strategy for the generation of 2-aminopyridine derivatives via a two-component reaction converging aryl vinamidinium salts with 1,1-enediamines (EDAMs) in a dimethyl sulfoxide (DMSO) solvent system, catalyzed by triethylamine (Et3N). The protocol introduces a rapid, efficient, and scalable synthetic pathway, achieving good to excellent yields while maintaining simplistic workup procedures. Seventeen derivatives were synthesized and subsequently screened for their inhibitory activity against AChE and BChE. The most potent derivative, 3m, exhibited an IC50 value of 34.81 ± 3.71 µM against AChE and 20.66 ± 1.01 µM against BChE compared to positive control donepezil with an IC50 value of 0.079 ± 0.05 µM against AChE and 10.6 ± 2.1 µM against BChE. Also, detailed kinetic studies were undertaken to elucidate their modes of enzymatic inhibition of the most potent compounds against both AChE and BChE. The promising compound was then subjected to molecular docking and dynamics simulations, revealing significant binding affinities and favorable interaction profiles against AChE and BChE. The in silico ADMET assessments further determined the drug-like properties of 3m, suggesting it as a promising candidate for further pre-clinical development.
Collapse
Affiliation(s)
- Sama Loori
- Department of Chemistry, Faculty of Sciences, Persian Gulf University, Bushehr, 75169, Iran
| | - Hormoz Pourtaher
- Department of Chemistry, Faculty of Sciences, Persian Gulf University, Bushehr, 75169, Iran
| | | | - Alireza Hasaninejad
- Department of Chemistry, Faculty of Sciences, Persian Gulf University, Bushehr, 75169, Iran
| | | | - Aida Iraji
- Department of Persian Medicine, School of Medicine, Research Center for Traditional Medicine and History of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran.
- Stem Cells Technology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran.
| |
Collapse
|
14
|
Guo B, Zhang W, Zhou Y, Zhang J, Zeng C, Sun P, Liu B. Fucoxanthin restructures the gut microbiota and metabolic functions of non-obese individuals in an in vitro fermentation model. Food Funct 2024; 15:4805-4817. [PMID: 38563411 DOI: 10.1039/d3fo05671f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/04/2024]
Abstract
Fucoxanthin, a carotenoid exclusively derived from algae, exerts its bioactivities with the modulation of the gut microbiota in mice. However, mechanisms through which fucoxanthin regulates the gut microbiota and its derived metabolites/metabolism in humans remain unclear. In this study, we investigated the effects of fucoxanthin on the gut microbiota and metabolism of non-obese individuals using an in vitro simulated digestion-fermentation cascade model. The results showed that about half of the fucoxanthin was not absorbed in the intestine, thus reaching the colon. The gut microbiota from fecal samples underwent significant changes after 48 or 72 hours in vitro fermentation. Specifically, fucoxanthin significantly enhanced the relative abundance of Bacteroidota and Parabacteroides, leading to improved functions of the gut microbiota in its development, glycan biosynthesis and metabolism as well as in improving the digestive system, endocrine system and immune system. The recovery of fucoxanthin during fermentation showed a decreasing trend with the slight bio-conversion of fucoxanthinol. Notably, fucoxanthin supplementation significantly altered metabolites, especially bile acids and indoles in the simulated human gut ecosystem. Correlation analysis indicated the involvement of the gut microbiota in the manipulation of these metabolites by fucoxanthin. Moreover, all these altered metabolites revealed the improvement in the capacity of fucoxanthin in manipulating gut metabolism, especially lipid metabolism. Overall, fucoxanthin determinedly reshaped the gut microbiota and metabolism, implying its potential health benefits in non-obese individuals.
Collapse
Affiliation(s)
- Bingbing Guo
- College of Chemistry and Life Science, Beijing University of Technology, Beijing, 100124, China
| | - Weihao Zhang
- College of Chemistry and Life Science, Beijing University of Technology, Beijing, 100124, China
| | - Yonghui Zhou
- College of Chemistry and Life Science, Beijing University of Technology, Beijing, 100124, China
| | - Jingyi Zhang
- College of Chemistry and Life Science, Beijing University of Technology, Beijing, 100124, China
| | - Chengchu Zeng
- College of Chemistry and Life Science, Beijing University of Technology, Beijing, 100124, China
| | - Peipei Sun
- Institute of Food Science and Technology, Chinese Academy of Agricultural Sciences, Beijing 100193, China
| | - Bin Liu
- Shenzhen Key Laboratory of Food Nutrition and Health, Institute for Innovative Development of Food Industry, Department of Food Science and Engineering, College of Chemistry and Environmental Engineering, Shenzhen University, Shenzhen, China.
| |
Collapse
|
15
|
Behzadnia A, Moosavi-Nasab M, Oliyaei N. Anti-biofilm activity of marine algae-derived bioactive compounds. Front Microbiol 2024; 15:1270174. [PMID: 38680918 PMCID: PMC11055458 DOI: 10.3389/fmicb.2024.1270174] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Accepted: 03/27/2024] [Indexed: 05/01/2024] Open
Abstract
A large number of microbial species tend to communicate and produce biofilm which causes numerous microbial infections, antibiotic resistance, and economic problems across different industries. Therefore, advanced anti-biofilms are required with novel attributes and targets, such as quorum sensing communication system. Meanwhile, quorum sensing inhibitors as promising anti-biofilm molecules result in the inhibition of particular phenotype expression blocking of cell-to-cell communication, which would be more acceptable than conventional strategies. Many natural products are identified as anti-biofilm agents from different plants, microorganisms, and marine extracts. Marine algae are promising sources of broadly novel compounds with anti-biofilm activity. Algae extracts and their metabolites such as sulfated polysaccharides (fucoidan), carotenoids (zeaxanthin and lutein), lipid and fatty acids (γ-linolenic acid and linoleic acid), and phlorotannins can inhibit the cell attachment, reduce the cell growth, interfere in quorum sensing pathway by blocking related enzymes, and disrupt extracellular polymeric substances. In this review, the mechanisms of biofilm formation, quorum sensing pathway, and recently identified marine algae natural products as anti-biofilm agents will be discussed.
Collapse
Affiliation(s)
- Asma Behzadnia
- Seafood Processing Research Center, School of Agriculture, Shiraz University, Shiraz, Iran
- Department of Food Science and Technology, School of Agriculture, Shiraz University, Shiraz, Iran
| | - Marzieh Moosavi-Nasab
- Seafood Processing Research Center, School of Agriculture, Shiraz University, Shiraz, Iran
- Department of Food Science and Technology, School of Agriculture, Shiraz University, Shiraz, Iran
| | - Najmeh Oliyaei
- Seafood Processing Research Center, School of Agriculture, Shiraz University, Shiraz, Iran
- Department of Food Science and Technology, School of Agriculture, Shiraz University, Shiraz, Iran
| |
Collapse
|
16
|
Asadipour A, Pourshojaei Y, Mansouri M, Mahdavizadeh E, Irajie C, Mottaghipisheh J, Faghih-Mirzaei E, Mahdavi M, Iraji A. Amino-7,8-dihydro-4H-chromenone derivatives as potential inhibitors of acetylcholinesterase and butyrylcholinesterase for Alzheimer's disease management; in vitro and in silico study. BMC Chem 2024; 18:70. [PMID: 38600537 PMCID: PMC11007943 DOI: 10.1186/s13065-024-01170-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2023] [Accepted: 03/22/2024] [Indexed: 04/12/2024] Open
Abstract
In this article, we present the design and synthesis of amino-7,8-dihydro-4H-chromenone derivatives as possible inhibitors of acetylcholinesterase (AChE) and butyrylcholinesterase (BChE) for the management of Alzheimer's disease (AD). The target compounds were evaluated against AChE and BChE in vitro, and 4k exhibited good potency against BChE (IC50 = 0.65 ± 0.13 µM) compared with donepezil used as a positive control. Kinetic studies revealed that compound 4k exhibited a competitive-type inhibition with a Ki value of 0.55 µM. Molecular docking and molecular dynamics simulations further supported the rationality of our design strategy, as 4k showed promising binding interactions with the active sites of BChE. Overall, our findings highlight the potential of amino-7,8-dihydro-4H-chromenone derivatives as promising candidates for developing novel therapeutics targeting cholinesterase in managing AD.
Collapse
Affiliation(s)
- Ali Asadipour
- Department of Medicinal Chemistry, Faculty of Pharmacy, Kerman University of Medical Sciences, Kerman, Iran
| | - Yaghoub Pourshojaei
- Department of Medicinal Chemistry, Faculty of Pharmacy, Kerman University of Medical Sciences, Kerman, Iran
- Extremophile and Productive Microorganisms Research Center, Kerman University of Medical Sciences, Kerman, Iran
| | - Moein Mansouri
- Department of Medicinal Chemistry, Faculty of Pharmacy, Kerman University of Medical Sciences, Kerman, Iran
| | - Elham Mahdavizadeh
- Department of Medicinal Chemistry, Faculty of Pharmacy, Kerman University of Medical Sciences, Kerman, Iran
| | - Cambyz Irajie
- Department of Medical Biotechnology, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Javad Mottaghipisheh
- Department of Aquatic Sciences and Assessment, Swedish University of Agricultural Sciences, 75007, Uppsala, Sweden
- Research Center for Traditional Medicine and History of Medicine, Department of Persian Medicine, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Ehsan Faghih-Mirzaei
- Department of Medicinal Chemistry, Faculty of Pharmacy, Kerman University of Medical Sciences, Kerman, Iran
| | - Mohammad Mahdavi
- Endocrinology and Metabolism Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Aida Iraji
- Research Center for Traditional Medicine and History of Medicine, Department of Persian Medicine, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran.
- Stem Cells Technology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran.
| |
Collapse
|
17
|
Nazarian A, Abedinifar F, Hamedifar H, Hashempur MH, Mahdavi M, Sepehri N, Iraji A. Anticholinesterase activities of novel isoindolin-1,3-dione-based acetohydrazide derivatives: design, synthesis, biological evaluation, molecular dynamic study. BMC Chem 2024; 18:64. [PMID: 38561813 PMCID: PMC10985906 DOI: 10.1186/s13065-024-01169-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2024] [Accepted: 03/21/2024] [Indexed: 04/04/2024] Open
Abstract
In pursuit of developing novel cholinesterase (ChE) inhibitors through molecular hybridization theory, a novel series of isoindolin-1,3-dione-based acetohydrazides (compounds 8a-h) was designed, synthesized, and evaluated as possible acetylcholinesterase (AChE) and butyrylcholinesterase (BChE) inhibitors. In vitro results revealed IC50 values ranging from 0.11 ± 0.05 to 0.86 ± 0.02 µM against AChE and 5.7 ± 0.2 to 30.2 ± 2.8 µM against BChE. A kinetic study was conducted on the most potent compound, 8a, to ascertain its mode of inhibition, revealing its competitive mode against AChE. Furthermore, the binding interaction modes of the most active compound within the AChE active site was elucidated. Molecular dynamics simulations of compound 8a were performed to assess the stability of the 8a-AChE complex. In silico pharmacokinetic predictions for the most potent compounds indicated their potential as promising lead structure for the development of new anti-Alzheimer's disease (anti-AD) agents.
Collapse
Affiliation(s)
- Ahmad Nazarian
- Endocrinology and Metabolism Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Fahime Abedinifar
- Endocrinology and Metabolism Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Haleh Hamedifar
- CinnaGen Medical Biotechnology Research Center, Alborz University of Medical Sciences, Karaj, Iran
- CinnaGen Research and Production Co., Alborz, Iran
| | - Mohammad Hashem Hashempur
- Research Center for Traditional Medicine and History of Medicine, Department of Persian Medicine, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Mohammad Mahdavi
- Endocrinology and Metabolism Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Nima Sepehri
- CinnaGen Medical Biotechnology Research Center, Alborz University of Medical Sciences, Karaj, Iran.
- CinnaGen Research and Production Co., Alborz, Iran.
| | - Aida Iraji
- Research Center for Traditional Medicine and History of Medicine, Department of Persian Medicine, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran.
- Stem Cells Technology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran.
| |
Collapse
|
18
|
Reiss AB, Gulkarov S, Jacob B, Srivastava A, Pinkhasov A, Gomolin IH, Stecker MM, Wisniewski T, De Leon J. Mitochondria in Alzheimer's Disease Pathogenesis. Life (Basel) 2024; 14:196. [PMID: 38398707 PMCID: PMC10890468 DOI: 10.3390/life14020196] [Citation(s) in RCA: 16] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2024] [Revised: 01/23/2024] [Accepted: 01/25/2024] [Indexed: 02/25/2024] Open
Abstract
Alzheimer's disease (AD) is a progressive and incurable neurodegenerative disorder that primarily affects persons aged 65 years and above. It causes dementia with memory loss and deterioration in thinking and language skills. AD is characterized by specific pathology resulting from the accumulation in the brain of extracellular plaques of amyloid-β and intracellular tangles of phosphorylated tau. The importance of mitochondrial dysfunction in AD pathogenesis, while previously underrecognized, is now more and more appreciated. Mitochondria are an essential organelle involved in cellular bioenergetics and signaling pathways. Mitochondrial processes crucial for synaptic activity such as mitophagy, mitochondrial trafficking, mitochondrial fission, and mitochondrial fusion are dysregulated in the AD brain. Excess fission and fragmentation yield mitochondria with low energy production. Reduced glucose metabolism is also observed in the AD brain with a hypometabolic state, particularly in the temporo-parietal brain regions. This review addresses the multiple ways in which abnormal mitochondrial structure and function contribute to AD. Disruption of the electron transport chain and ATP production are particularly neurotoxic because brain cells have disproportionately high energy demands. In addition, oxidative stress, which is extremely damaging to nerve cells, rises dramatically with mitochondrial dyshomeostasis. Restoring mitochondrial health may be a viable approach to AD treatment.
Collapse
Affiliation(s)
- Allison B. Reiss
- Department of Medicine and Biomedical Research Institute, NYU Grossman Long Island School of Medicine, Mineola, NY 11501, USA; (S.G.); (B.J.); (A.S.); (A.P.); (I.H.G.); (J.D.L.)
| | - Shelly Gulkarov
- Department of Medicine and Biomedical Research Institute, NYU Grossman Long Island School of Medicine, Mineola, NY 11501, USA; (S.G.); (B.J.); (A.S.); (A.P.); (I.H.G.); (J.D.L.)
| | - Benna Jacob
- Department of Medicine and Biomedical Research Institute, NYU Grossman Long Island School of Medicine, Mineola, NY 11501, USA; (S.G.); (B.J.); (A.S.); (A.P.); (I.H.G.); (J.D.L.)
| | - Ankita Srivastava
- Department of Medicine and Biomedical Research Institute, NYU Grossman Long Island School of Medicine, Mineola, NY 11501, USA; (S.G.); (B.J.); (A.S.); (A.P.); (I.H.G.); (J.D.L.)
| | - Aaron Pinkhasov
- Department of Medicine and Biomedical Research Institute, NYU Grossman Long Island School of Medicine, Mineola, NY 11501, USA; (S.G.); (B.J.); (A.S.); (A.P.); (I.H.G.); (J.D.L.)
| | - Irving H. Gomolin
- Department of Medicine and Biomedical Research Institute, NYU Grossman Long Island School of Medicine, Mineola, NY 11501, USA; (S.G.); (B.J.); (A.S.); (A.P.); (I.H.G.); (J.D.L.)
| | - Mark M. Stecker
- The Fresno Institute of Neuroscience, Fresno, CA 93730, USA;
| | - Thomas Wisniewski
- Center for Cognitive Neurology, Departments of Neurology, Pathology and Psychiatry, New York University Grossman School of Medicine, New York, NY 10016, USA;
| | - Joshua De Leon
- Department of Medicine and Biomedical Research Institute, NYU Grossman Long Island School of Medicine, Mineola, NY 11501, USA; (S.G.); (B.J.); (A.S.); (A.P.); (I.H.G.); (J.D.L.)
| |
Collapse
|
19
|
Pourtaher H, Mohammadi Y, Hasaninejad A, Iraji A. Highly efficient, catalyst-free, one-pot sequential four-component synthesis of novel spiroindolinone-pyrazole scaffolds as anti-Alzheimer agents: in silico study and biological screening. RSC Med Chem 2024; 15:207-222. [PMID: 38283217 PMCID: PMC10809384 DOI: 10.1039/d3md00255a] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2023] [Accepted: 10/20/2023] [Indexed: 01/30/2024] Open
Abstract
Alzheimer's disease is a neurodegenerative disorder that impacts memory, thinking, and behavior, and currently, there is no effective cure available for its treatment. This study explored a one-pot strategy for synthesizing spiroindolinone-pyrazole derivatives through a sequential four-component condensation reaction. These derivatives were further investigated for their potential as anti-Alzheimer's disease agents. The developed synthetic procedure provides remarkable advantages, including a clean reaction profile, abundant starting materials, operational simplicity, and easy purification without traditional methods with good to excellent yields (84-96%). Next, the biological potencies of the newly synthesized spiroindolinone-pyrazole derivatives against AChE and BChE as Alzheimer's disease-related targets were determined. Also, the kinetic study and cytotoxicity of the most potent derivative were investigated. Furthermore, molecular docking and molecular dynamics evaluations were performed employing in silico tools to investigate the interaction, orientation, and conformation of the potent analog over the active site of the enzyme.
Collapse
Affiliation(s)
- Hormoz Pourtaher
- Department of Chemistry, Faculty of Sciences, Persian Gulf University Bushehr Iran
| | - Yasaman Mohammadi
- Research Center for Traditional Medicine and History of Medicine, Department of Persian Medicine, School of Medicine, Shiraz University of Medical Sciences Shiraz 7134845794 Iran
- Stem Cells Technology Research Center, Shiraz University of Medical Sciences Shiraz Iran
| | - Alireza Hasaninejad
- Department of Chemistry, Faculty of Sciences, Persian Gulf University Bushehr Iran
| | - Aida Iraji
- Research Center for Traditional Medicine and History of Medicine, Department of Persian Medicine, School of Medicine, Shiraz University of Medical Sciences Shiraz 7134845794 Iran
- Stem Cells Technology Research Center, Shiraz University of Medical Sciences Shiraz Iran
| |
Collapse
|
20
|
Abstract
Alzheimer's, Parkinson's, and dementia are the leading neurodegenerative diseases that threaten the world with the aging population. Although the pathophysiology of each disease is unique, the steps to be taken to prevent diseases are similar. One of the changes that a person can make alone is to gain the habit of an antioxidant-rich diet. Phytochemicals known for their antioxidant properties have been reported to prevent neurodegenerative diseases in various studies. Phytochemicals with similar chemical structures are grouped. Accordingly, there are two main groups of phytochemicals, flavonoid and non-flavonoid. Various in vitro and in vivo studies on phytochemicals have proven neuroprotective effects by increasing cognitive function with their anti-inflammatory and antioxidant mechanisms. The purpose of this review is to summarize the in vitro and in vivo studies on phytochemicals with neuroprotective effects and to provide insight.
Collapse
Affiliation(s)
- Basak Can
- Nutrition and Dietetics, School of Health Sciences, Istanbul Gelisim University, Istanbul, Turkey
| | - Nevin Sanlier
- School of Health Sciences, Nutrition and Dietetics, Ankara Medipol University, Ankara, Turkey
| |
Collapse
|
21
|
Hong DD, Thom LT, Ha NC, Thu NTH, Hien HTM, Tam LT, Dat NM, Duc TM, Tru NV, Hang NTM, Ambati RR. Isolation of Fucoxanthin from Sargassum oligocystum Montagne, 1845 Seaweed in Vietnam and Its Neuroprotective Activity. Biomedicines 2023; 11:2310. [PMID: 37626806 PMCID: PMC10452663 DOI: 10.3390/biomedicines11082310] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2023] [Revised: 08/08/2023] [Accepted: 08/17/2023] [Indexed: 08/27/2023] Open
Abstract
Fucoxanthin extracted and purified from Vietnamese Sargassum oligocystum Montagne, 1845 exhibits various biological activities. In this study, the ability of fucoxanthin to inhibit acetylcholinesterase (AChE), the antioxidant activities, and the expression of antioxidant enzymes were investigated. Fucoxanthin isolated from Vietnamese S. oligocystum showed no cytotoxic effects; moreover, it exhibited AChE inhibitory activity (with an IC50 value of 130.12 ± 6.65 μg mL-1) and antioxidant activity (with an IC50 value of 3.42 ± 0.15 mg mL-1). At concentrations of 50 and 100 µg mL-1, fucoxanthin provided protection against amyloid β-protein fragment 25-35-induced neurotoxicity in a C6 neuronal cell line, and the survival of C6 cells was higher than 81.01% and 80.98%, respectively, compared to the control group (59%). Moreover, antioxidant enzyme activity and quantitative PCR analysis suggested that the neuroprotective effect of fucoxanthin resulted from regulation of the gene expression of antioxidant enzymes (CAT and GPx) and ER pathways (caspase-3 and Bax), as well as the promotion of expression of genes involved in PI3K/Akt signaling (GSK-3β), autophagy (p62 and ATG5), and the biosynthesis of ACh (VAChT and ChAT). Therefore, fucoxanthin extracted from the seaweed S. oligocystum in Vietnam is a potential feedstock source for the production of health foods that exert neuroprotective effects.
Collapse
Affiliation(s)
- Dang Diem Hong
- Institute of Biotechnology, Vietnam Academy of Science and Technology, 18 Hoang Quoc Viet Str., Cau Giay, Hanoi 100000, Vietnam; (L.T.T.); (N.C.H.); (N.T.H.T.); (H.T.M.H.); (L.T.T.); (N.M.D.); (N.V.T.)
- Department of Microbiology, Graduate University of Science and Technology, Vietnam Academy of Science and Technology, 18 Hoang Quoc Viet Str., Cau Giay, Hanoi 100000, Vietnam
| | - Le Thi Thom
- Institute of Biotechnology, Vietnam Academy of Science and Technology, 18 Hoang Quoc Viet Str., Cau Giay, Hanoi 100000, Vietnam; (L.T.T.); (N.C.H.); (N.T.H.T.); (H.T.M.H.); (L.T.T.); (N.M.D.); (N.V.T.)
| | - Nguyen Cam Ha
- Institute of Biotechnology, Vietnam Academy of Science and Technology, 18 Hoang Quoc Viet Str., Cau Giay, Hanoi 100000, Vietnam; (L.T.T.); (N.C.H.); (N.T.H.T.); (H.T.M.H.); (L.T.T.); (N.M.D.); (N.V.T.)
| | - Ngo Thi Hoai Thu
- Institute of Biotechnology, Vietnam Academy of Science and Technology, 18 Hoang Quoc Viet Str., Cau Giay, Hanoi 100000, Vietnam; (L.T.T.); (N.C.H.); (N.T.H.T.); (H.T.M.H.); (L.T.T.); (N.M.D.); (N.V.T.)
| | - Hoang Thi Minh Hien
- Institute of Biotechnology, Vietnam Academy of Science and Technology, 18 Hoang Quoc Viet Str., Cau Giay, Hanoi 100000, Vietnam; (L.T.T.); (N.C.H.); (N.T.H.T.); (H.T.M.H.); (L.T.T.); (N.M.D.); (N.V.T.)
| | - Luu Thi Tam
- Institute of Biotechnology, Vietnam Academy of Science and Technology, 18 Hoang Quoc Viet Str., Cau Giay, Hanoi 100000, Vietnam; (L.T.T.); (N.C.H.); (N.T.H.T.); (H.T.M.H.); (L.T.T.); (N.M.D.); (N.V.T.)
| | - Nguyen Manh Dat
- Institute of Biotechnology, Vietnam Academy of Science and Technology, 18 Hoang Quoc Viet Str., Cau Giay, Hanoi 100000, Vietnam; (L.T.T.); (N.C.H.); (N.T.H.T.); (H.T.M.H.); (L.T.T.); (N.M.D.); (N.V.T.)
| | - Tran Mai Duc
- Nha Trang Institute of Technology Research and Application, Vietnam Academy of Science and Technology, Nha Trang 57100, Vietnam;
| | - Nguyen Van Tru
- Institute of Biotechnology, Vietnam Academy of Science and Technology, 18 Hoang Quoc Viet Str., Cau Giay, Hanoi 100000, Vietnam; (L.T.T.); (N.C.H.); (N.T.H.T.); (H.T.M.H.); (L.T.T.); (N.M.D.); (N.V.T.)
| | - Nguyen Thi Minh Hang
- Institute of Marine Biochemistry, Vietnam Academy of Science and Technology, 18 Hoang Quoc Viet Str., Cau Giay, Hanoi 100000, Vietnam;
| | - Ranga Rao Ambati
- Department of Biotechnology, School of Biotechnology and Pharmaceutical Sciences, Vignan’s Foundation for Science, Technology and Research (Deemed to Be University), Vadlamudi, Guntur 522213, India;
| |
Collapse
|
22
|
Shehata MK, Ismail AA, Kamel MA. Combined Donepezil with Astaxanthin via Nanostructured Lipid Carriers Effective Delivery to Brain for Alzheimer's Disease in Rat Model. Int J Nanomedicine 2023; 18:4193-4227. [PMID: 37534058 PMCID: PMC10391537 DOI: 10.2147/ijn.s417928] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2023] [Accepted: 07/19/2023] [Indexed: 08/04/2023] Open
Abstract
Introduction Donepezil (DPL), a specific acetylcholinesterase inhibitor, is used as a first-line treatment to improve cognitive deficits in Alzheimer's disease (AD) and it might have a disease modifying effect. Astaxanthin (AST) is a natural potent antioxidant with neuroprotective, anti-amyloidogenic, anti-apoptotic, and anti-inflammatory effects. This study aimed to prepare nanostructured lipid carriers (NLCs) co-loaded with donepezil and astaxanthin (DPL/AST-NLCs) and evaluate their in vivo efficacy in an AD-like rat model 30 days after daily intranasal administration. Methods DPL/AST-NLCs were prepared using a hot high-shear homogenization technique, in vitro examined for their physicochemical parameters and in vivo evaluated. AD induction in rats was performed by aluminum chloride. The cortex and hippocampus were isolated from the brain of rats for biochemical testing and histopathological examination. Results DPL/AST-NLCs showed z-average diameter 149.9 ± 3.21 nm, polydispersity index 0.224 ± 0.017, zeta potential -33.7 ± 4.71 mV, entrapment efficiency 81.25 ±1.98% (donepezil) and 93.85 ±1.75% (astaxanthin), in vitro sustained release of both donepezil and astaxanthin for 24 h, spherical morphology by transmission electron microscopy, and they were stable at 4-8 ± 2°C for six months. Differential scanning calorimetry revealed that donepezil and astaxanthin were molecularly dispersed in the NLC matrix in an amorphous state. The DPL/AST-NLC-treated rats showed significantly lower levels of nuclear factor-kappa B, malondialdehyde, β-site amyloid precursor protein cleaving enzyme-1, caspase-3, amyloid beta (Aβ1‑42), and acetylcholinesterase, and significantly higher levels of glutathione and acetylcholine in the cortex and hippocampus than the AD-like untreated rats and that treated with donepezil-NLCs. DPL/AST-NLCs showed significantly higher anti-amyloidogenic, antioxidant, anti-acetylcholinesterase, anti-inflammatory, and anti-apoptotic effects, resulting in significant improvement in the cortical and hippocampal histopathology. Conclusion Nose-to-brain delivery of DPL/AST-NLCs is a promising strategy for the management of AD.
Collapse
Affiliation(s)
- Mustafa K Shehata
- Department of Pharmaceutics, Faculty of Pharmacy, Alexandria University, Alexandria, Egypt
| | - Assem A Ismail
- Department of Pharmaceutics, Faculty of Pharmacy, Alexandria University, Alexandria, Egypt
| | - Maher A Kamel
- Department of Biochemistry, Medical Research Institute, Alexandria University, Alexandria, Egypt
| |
Collapse
|
23
|
Pourtaher H, Hasaninejad A, Zare S, Tanideh N, Iraji A. The anti-Alzheimer potential of novel spiroindolin-1,2-diazepine derivatives as targeted cholinesterase inhibitors with modified substituents. Sci Rep 2023; 13:11952. [PMID: 37488177 PMCID: PMC10366214 DOI: 10.1038/s41598-023-38236-0] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2023] [Accepted: 07/05/2023] [Indexed: 07/26/2023] Open
Abstract
In this study, a new series of spiro indolin-1,2-diazepine were designed, synthesized, and screened for their cholinesterase inhibitory activities. A novel, green, high-yielding approach was constructed to synthesize spiro indolin-1,2-diazepine derivatives through a cascade reaction of different isatins, malononitrile and 1,1-enediamines (EDAMs) via sequential four-component reactions to produce the target compounds with good to excellent yields. Next the inhibitory potencies of all derivatives were determined spectroscopically at 415 nm using the modified Ellman method. The results of the in vitro screening indicated that 5l with spiroindolin-1,2-diazepine core bearing 5-NO2 at R1 and 4-OH at R2 was the most potent and selective AChE inhibitor with an IC50 value of 3.98 ± 1.07 µM with no significant inhibition against BChE while 5j was the most active analog against both AChE and BChE enzymes. The structure-activity relationships suggested the variation in the inhibitory activities of derivatives was affected by different substitutions on the indolinone ring as well as the phenyl moiety. The enzyme kinetic studies of the most potent compound 5l at five different concentrations and acetylthiocholine substrate (0.1-1 mM) by Ellman's method revealed that it inhibited AChE in a mixed mode with a Ki of 0.044 μM. A molecular docking study was performed via induced fit docking protocol to predict the putative binding interaction. It was shown that the moieties used in the initial structure design play a fundamental role in interacting with the enzyme's binding site. Further, molecular dynamics simulations with the Schrödinger package were performed for 5l in a complex with AChE and revealed that compound 5l formed the stable complex with the enzyme. The MTT toxicity assessments against the neuroblastoma cell line were executed, and no toxicity was seen for 5l under the tested concentrations.
Collapse
Affiliation(s)
- Hormoz Pourtaher
- Department of Chemistry, Faculty of Sciences, Persian Gulf University, Bushehr, 75169, Iran
| | - Alireza Hasaninejad
- Department of Chemistry, Faculty of Sciences, Persian Gulf University, Bushehr, 75169, Iran.
| | - Shahrokh Zare
- Stem Cells Technology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Nader Tanideh
- Stem Cells Technology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Aida Iraji
- Research Center for Traditional Medicine and History of Medicine, Department of Persian Medicine, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran.
| |
Collapse
|
24
|
Cunha SA, Borges S, Baptista-Silva S, Ribeiro T, Oliveira-Silva P, Pintado M, Batista P. Astaxanthin impact on brain: health potential and market perspective. Crit Rev Food Sci Nutr 2023; 64:11067-11090. [PMID: 37417323 DOI: 10.1080/10408398.2023.2232866] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/08/2023]
Abstract
Nowadays, there is an emergent interest in new trend-driven biomolecules to improve health and wellbeing, which has become an interesting and promising field, considering their high value and biological potential. Astaxanthin is one of these promising biomolecules, with impressive high market growth, especially in the pharmaceutical and food industries. This biomolecule, obtained from natural sources (i.e., microalgae), has been reported in the literature to have several beneficial health effects due to its biological properties. These benefits seem to be mainly associated with Astaxanthin's high antioxidant and anti-inflammatory properties, which may act on several brain issues, thus attenuating symptoms. In this sense, several studies have demonstrated the impact of astaxanthin on a wide range of diseases, namely on brain disorders (such as Alzheimer's disease, Parkinson, depression, brain stroke and autism). Therefore, this review highlights its application in mental health and illness. Furthermore, a S.W.O.T. analysis was performed to display an approach from the market/commercial perspective. However, to bring the molecule to the market, there is still a need for more studies to increase deep knowledge regarding the real impact and mechanisms in the human brain.HIGHLIGHTSAstaxanthin has been mainly extracted from the algae Haematococcus pluvialisAstaxanthin, bioactive molecule with high antioxidant and anti-inflammatory propertiesAstaxanthin has an important protective effect on brain disordersAstaxanthin is highly marketable, mainly for food and pharmaceutical industries.
Collapse
Affiliation(s)
- Sara A Cunha
- CBQF - Centro de Biotecnologia e Química Fina - Laboratório Associado, Escola Superior de Biotecnologia, Universidade Católica Portuguesa, Porto, Portugal
| | - Sandra Borges
- CBQF - Centro de Biotecnologia e Química Fina - Laboratório Associado, Escola Superior de Biotecnologia, Universidade Católica Portuguesa, Porto, Portugal
| | - Sara Baptista-Silva
- CBQF - Centro de Biotecnologia e Química Fina - Laboratório Associado, Escola Superior de Biotecnologia, Universidade Católica Portuguesa, Porto, Portugal
| | - Tânia Ribeiro
- CBQF - Centro de Biotecnologia e Química Fina - Laboratório Associado, Escola Superior de Biotecnologia, Universidade Católica Portuguesa, Porto, Portugal
| | - Patrícia Oliveira-Silva
- Universidade Católica Portuguesa, Research Centre for Human Development, Human Neurobehavioral Laboratory, Porto, Portugal
| | - Manuela Pintado
- CBQF - Centro de Biotecnologia e Química Fina - Laboratório Associado, Escola Superior de Biotecnologia, Universidade Católica Portuguesa, Porto, Portugal
| | - Patrícia Batista
- CBQF - Centro de Biotecnologia e Química Fina - Laboratório Associado, Escola Superior de Biotecnologia, Universidade Católica Portuguesa, Porto, Portugal
- Universidade Católica Portuguesa, Research Centre for Human Development, Human Neurobehavioral Laboratory, Porto, Portugal
| |
Collapse
|
25
|
Su W, Xu W, Liu E, Su W, Polyakov NE. Improving the Treatment Effect of Carotenoids on Alzheimer's Disease through Various Nano-Delivery Systems. Int J Mol Sci 2023; 24:ijms24087652. [PMID: 37108814 PMCID: PMC10142927 DOI: 10.3390/ijms24087652] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Revised: 04/10/2023] [Accepted: 04/20/2023] [Indexed: 04/29/2023] Open
Abstract
Natural bioactive compounds have recently emerged as a current strategy for Alzheimer's disease treatment. Carotenoids, including astaxanthin, lycopene, lutein, fucoxanthin, crocin and others are natural pigments and antioxidants, and can be used to treat a variety of diseases, including Alzheimer's disease. However, carotenoids, as oil-soluble substances with additional unsaturated groups, suffer from low solubility, poor stability and poor bioavailability. Therefore, the preparation of various nano-drug delivery systems from carotenoids is a current measure to achieve efficient application of carotenoids. Different carotenoid delivery systems can improve the solubility, stability, permeability and bioavailability of carotenoids to a certain extent to achieve Alzheimer's disease efficacy. This review summarizes recent data on different carotenoid nano-drug delivery systems for the treatment of Alzheimer's disease, including polymer, lipid, inorganic and hybrid nano-drug delivery systems. These drug delivery systems have been shown to have a beneficial therapeutic effect on Alzheimer's disease to a certain extent.
Collapse
Affiliation(s)
- Wenjing Su
- Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, Zhejiang University of Technology, Hangzhou 310014, China
| | - Wenhao Xu
- Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, Zhejiang University of Technology, Hangzhou 310014, China
| | - Enshuo Liu
- Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, Zhejiang University of Technology, Hangzhou 310014, China
| | - Weike Su
- Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, Zhejiang University of Technology, Hangzhou 310014, China
| | - Nikolay E Polyakov
- Institute of Solid State Chemistry and Mechanochemistry, 630128 Novosibirsk, Russia
- Institute of Chemical Kinetics and Combustion, 630090 Novosibirsk, Russia
| |
Collapse
|
26
|
Nieri P, Carpi S, Esposito R, Costantini M, Zupo V. Bioactive Molecules from Marine Diatoms and Their Value for the Nutraceutical Industry. Nutrients 2023; 15:464. [PMID: 36678334 PMCID: PMC9861441 DOI: 10.3390/nu15020464] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Revised: 01/12/2023] [Accepted: 01/13/2023] [Indexed: 01/17/2023] Open
Abstract
The search for novel sources of nutrients is among the basic goals for achievement of sustainable progress. In this context, microalgae are relevant organisms, being rich in high-value compounds and able to grow in open ponds or photobioreactors, thus enabling profitable exploitation of aquatic resources. Microalgae, a huge taxon containing photosynthetic microorganisms living in freshwater, as well as in brackish and marine waters, typically unicellular and eukaryotic, include green algae (Chlorophyceae), red algae (Rhodophyceae), brown algae (Phaeophyceae) and diatoms (Bacillariophyceae). In recent decades, diatoms have been considered the most sustainable sources of nutrients for humans with respect to other microalgae. This review focuses on studies exploring their bio-pharmacological activities when relevant for human disease prevention and/or treatment. In addition, we considered diatoms and their extracts (or purified compounds) when relevant for specific nutraceutical applications.
Collapse
Affiliation(s)
- Paola Nieri
- Department of Pharmacy, University of Pisa, Via Bonanno 6, 56126 Pisa, Italy
- Interdepartmental Center of Marine Pharmacology, University of Pisa, Via Bonanno 6, 56126 Pisa, Italy
| | - Sara Carpi
- National Enterprise for NanoScience and Nanotechnology (NEST), Piazza San Silvestro, 56127 Pisa, Italy
| | - Roberta Esposito
- Stazione Zoologica Antorn Dohrn, Department of Ecosustainable Marine Biotechnology, Via Ammiraglio Ferdinando Acton, 80133 Naples, Italy
- Department of Biology, University of Naples Federico II, Complesso Universitario di Monte Sant’Angelo, Via Cinthia 21, 80126 Naples, Italy
| | - Maria Costantini
- Stazione Zoologica Antorn Dohrn, Department of Ecosustainable Marine Biotechnology, Via Ammiraglio Ferdinando Acton, 80133 Naples, Italy
| | - Valerio Zupo
- Stazione Zoologica Antorn Dohrn, Department of Ecosustainable Marine Biotechnology, Ischia Marine Centre, 80077 Ischia, Italy
| |
Collapse
|