1
|
Zhang M, Xiang C, Niu R, He X, Luo W, Liu W, Gu R. Liposomes as versatile agents for the management of traumatic and nontraumatic central nervous system disorders: drug stability, targeting efficiency, and safety. Neural Regen Res 2025; 20:1883-1899. [PMID: 39254548 DOI: 10.4103/nrr.nrr-d-24-00048] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2024] [Accepted: 05/28/2024] [Indexed: 09/11/2024] Open
Abstract
Various nanoparticle-based drug delivery systems for the treatment of neurological disorders have been widely studied. However, their inability to cross the blood-brain barrier hampers the clinical translation of these therapeutic strategies. Liposomes are nanoparticles composed of lipid bilayers, which can effectively encapsulate drugs and improve drug delivery across the blood-brain barrier and into brain tissue through their targeting and permeability. Therefore, they can potentially treat traumatic and nontraumatic central nervous system diseases. In this review, we outlined the common properties and preparation methods of liposomes, including thin-film hydration, reverse-phase evaporation, solvent injection techniques, detergent removal methods, and microfluidics techniques. Afterwards, we comprehensively discussed the current applications of liposomes in central nervous system diseases, such as Alzheimer's disease, Parkinson's disease, Huntington's disease, amyotrophic lateral sclerosis, traumatic brain injury, spinal cord injury, and brain tumors. Most studies related to liposomes are still in the laboratory stage and have not yet entered clinical trials. Additionally, their application as drug delivery systems in clinical practice faces challenges such as drug stability, targeting efficiency, and safety. Therefore, we proposed development strategies related to liposomes to further promote their development in neurological disease research.
Collapse
Affiliation(s)
- Mingyu Zhang
- Department of Orthopedic Surgery, China-Japan Union Hospital of Jilin University, Changchun, Jilin Province, China
| | | | | | | | | | | | | |
Collapse
|
2
|
Suhandi C, Wilar G, Narsa AC, Mohammed AFA, El-Rayyes A, Muchtaridi M, Shamsuddin S, Safuan S, Wathoni N. Updating the Pharmacological Effects of α-Mangostin Compound and Unraveling Its Mechanism of Action: A Computational Study Review. Drug Des Devel Ther 2024; 18:4723-4748. [PMID: 39469723 PMCID: PMC11514645 DOI: 10.2147/dddt.s478388] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2024] [Accepted: 10/07/2024] [Indexed: 10/30/2024] Open
Abstract
α-Mangostin, initially identified in 1855, is a xanthone derivative compound predominantly located in the pericarp of the mangosteen fruit (Garcinia mangostana L). This compound is known for its beneficial properties as an antioxidant and anti-inflammatory agent, still holding promise for potential benefits in other related pathologies. In the investigative process, computational studies have proven highly valuable in providing evidence and initial screening before progressing to preclinical and clinical studies. This review aims to present the pharmacological findings and mechanisms of action of α-mangostin based on computational studies. The compilation of this review is founded on the analysis of relevant articles obtained from PubMed, Scopus, and ScienceDirect databases. The study commences with an elucidation of the physicochemical characteristics, drug-likeness, pharmacokinetics, and toxicity profile of α-mangostin, which demonstrates that α-mangostin complies with the Lipinski's Rule of Five, exhibits favorable profiles of absorption, distribution, metabolism, and excretion, and presents low toxicity. Subsequent investigations have revealed that computational studies employing various software tools including ArgusLab, AutoDock, AutoDock Vina, Glide, HEX, and MOE, have been pivotal to comprehend the pharmacology of α-mangostin. Beyond its well established roles as an antioxidant and anti-inflammatory agent, α-mangostin is now recognized for its pharmacological effects in Alzheimer's disease, diabetes, cancer, chronic kidney disease, chronic periodontitis, infectious diseases, and rheumatoid arthritis. Moreover, α-mangostin is projected to have applications in pain management and as a potent mosquito larvicide. All of these findings are based on the attainment of adequate binding affinity to specific target receptors associated with each respective pathological condition. Consequently, it is anticipated that these findings will serve as a foundation for future scientific endeavours, encompassing both in vitro and in vivo studies, as well as clinical investigations, to better understand the pharmacological effects of α-mangostin.
Collapse
Affiliation(s)
- Cecep Suhandi
- Department of Pharmaceutics and Pharmaceutical Technology, Faculty of Pharmacy, Universitas Padjadjaran, Jatinangor, 45363, Indonesia
| | - Gofarana Wilar
- Department of Pharmacology and Clinical Pharmacy, Faculty of Pharmacy, Universitas Padjadjaran, Jatinangor, 45363, Indonesia
| | - Angga Cipta Narsa
- Department of Pharmaceutics and Pharmaceutical Technology, Faculty of Pharmacy, Mulawarman University, Samarinda, 71157, Indonesia
| | | | - Ali El-Rayyes
- Department of Chemistry, College of Science, Northern Border University, Arar, Saudi Arabia
| | - Muchtaridi Muchtaridi
- Department of Analytical Pharmacy and Medicinal Chemistry, Faculty of Pharmacy, Universitas Padjadjaran, Jatinangor, 45363, Indonesia
| | - Shaharum Shamsuddin
- School of Health Sciences, Universiti Sains Malaysia, Kubang Kerian, Kelantan, 16150, Malaysia
| | - Sabreena Safuan
- Department of Biomedicine, School of Health Sciences, Universiti Sains Malaysia, Kubang Kerian, Kelantan, 16150, Malaysia
| | - Nasrul Wathoni
- Department of Pharmaceutics and Pharmaceutical Technology, Faculty of Pharmacy, Universitas Padjadjaran, Jatinangor, 45363, Indonesia
| |
Collapse
|
3
|
Thew HY, Boon Keat K, Tan YC, Ong YS, Parat MO, Murugaiyah V, Goh BH, Khaw KY. Probing the anti-Aβ42 aggregation and protective effects of prenylated xanthone against Aβ42-induced toxicity in transgenic Caenorhabditis elegans model. Chem Biol Interact 2024; 394:110978. [PMID: 38552766 DOI: 10.1016/j.cbi.2024.110978] [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: 02/02/2024] [Revised: 03/12/2024] [Accepted: 03/26/2024] [Indexed: 04/10/2024]
Abstract
Alzheimer's disease (AD) is a progressive neurodegenerative disorder characterized by the accumulation of amyloid-β (Aβ) protein aggregates, leading to synaptic dysfunction and neuronal cell death. In this study, we used a comprehensive approach encompassing in vitro assays, computational analyses, and an in vivo Caenorhabditis elegans model to evaluate the inhibitory effects of various xanthones, focusing on Garcinone D (GD), on Aβ42 oligomer formation. Dot blot analysis revealed concentration-dependent responses among xanthones, with GD consistently inhibiting Aβ42 oligomer formation at low concentrations (0.1 and 0.5 μM, inhibitions of 84.66 ± 2.25% and 85.06 ± 6.57%, respectively). Molecular docking and dynamics simulations provided insights into the molecular interactions between xanthones and Aβ42, highlighting the disruption of key residues involved in Aβ42 aggregation. The neuroprotective potential of GD was established using transgenic C. elegans GMC101, with substantial delays in paralysis reported at higher concentrations. Our findings show that GD is a potent suppressor of Aβ42 oligomer formation, suggesting its potential as a therapeutic candidate for AD. The concentration-dependent effects observed in both in vitro and in vivo models underscore the need for nuanced dose-response assessments. These findings contribute novel insights into the therapeutic landscape of xanthones against AD, emphasizing the multifaceted potential of GD for further translational endeavors in neurodegenerative disorder research.
Collapse
Affiliation(s)
- Hin Yee Thew
- School of Pharmacy, Monash University Malaysia, Bandar Sunway, 47500 Subang Jaya, Selangor Darul Ehsan, Malaysia
| | - Khor Boon Keat
- Discipline of Pharmacology, School of Pharmaceutical Sciences, Universiti Sains Malaysia, 11800, Penang, Malaysia
| | - Yong Chiang Tan
- International Medical University, 57000 Kuala Lumpur, Federal Territory of Kuala Lumpur, Malaysia
| | - Yong Sze Ong
- School of Pharmacy, Monash University Malaysia, Bandar Sunway, 47500 Subang Jaya, Selangor Darul Ehsan, Malaysia
| | - Marie-Odile Parat
- School of Pharmacy, Pharmacy Australia Centre of Excellence, University of Queensland, Brisbane, QLD 4102, Australia
| | - Vikneswaran Murugaiyah
- Centre for Drug Research, Universiti Sains Malaysia, 11800, Penang, Malaysia; Discipline of Pharmacology, School of Pharmaceutical Sciences, Universiti Sains Malaysia, 11800, Penang, Malaysia
| | - Bey Hing Goh
- Sunway Biofunctional Molecules Discovery Centre (SBMDC), School of Medical and Life Sciences, Sunway University, Sunway City, Selangor, Malaysia; Faculty of Health, Australian Research Centre in Complementary and Integrative Medicine, University of Technology Sydney, Ultimo, NSW 2007, Australia; Biofunctional Molecule Exploratory (BMEX) Research Group, School of Pharmacy, Monash University Malaysia, Subang Jaya, Malaysia; College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou, Zhejiang, 310058, China
| | - Kooi Yeong Khaw
- School of Pharmacy, Monash University Malaysia, Bandar Sunway, 47500 Subang Jaya, Selangor Darul Ehsan, Malaysia.
| |
Collapse
|
4
|
Panghal A, Flora SJS. Nanotechnology in the diagnostic and therapy for Alzheimer's disease. Biochim Biophys Acta Gen Subj 2024; 1868:130559. [PMID: 38191034 DOI: 10.1016/j.bbagen.2024.130559] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2023] [Revised: 12/21/2023] [Accepted: 01/04/2024] [Indexed: 01/10/2024]
Abstract
Alzheimer's disease (AD) is a neurodegenerative disorder primarily characterized by β-amyloid plaque, intraneuronal tangles, significant neuronal loss and cognitive deficit. Treatment in the early stages of the disease is crucial for preventing or perhaps reversing the neurodegeneration in the AD cases. However, none of the current diagnostic procedures are capable of early diagnosis of AD. Further, the available treatments merely provide symptomatic alleviation in AD and do not address the underlying illness. Therefore, there is no permanent cure for AD currently. Better therapeutic outcomes need the optimum drug concentration in the central nervous system (CNS) by traversing blood-brain-barrier (BBB). Nanotechnology offers enormous promise to transform the treatment and diagnostics of neurodegenerative diseases. Nanotechnology based diagnostic tools, drug delivery systems and theragnostic are capable of highly sensitive molecular detection, effective drug targeting and their combination. Significant work has been done in this area over the last decade and prospective results have been obtained in AD therapy. This review explores the various applications of nanotechnology in addressing the varied facets of AD, ranging from early detection to therapeutic interventions. This review also looks at how nanotechnology can help with the development of disease-modifying medicines, such as the delivery of anti-amyloid, anti-tau, cholinesterase inhibitors, antioxidants and hormonal drugs. In conclusion, this paper discusses the role of nanotechnology in the early detection of AD, effective drug targeting to the CNS and theragnostic applications in the management of AD.
Collapse
Affiliation(s)
- Archna Panghal
- National Institute of Pharmaceutical Education and Research, SAS Nagar, Mohali, Panjab 160012, India
| | - S J S Flora
- National Institute of Pharmaceutical Education and Research, SAS Nagar, Mohali, Panjab 160012, India; Institute of Pharmaceutical Sciences, Era Medical University, Safarajganj, Lucknow 226003, U.P., India.
| |
Collapse
|
5
|
Pang LW, Hamzah S, Tan SLJ, Mah SH, Yow HY. The Effects and Mechanisms of Xanthones in Alzheimer's Disease: A Systematic Review. Neurochem Res 2023; 48:3485-3511. [PMID: 37578655 DOI: 10.1007/s11064-023-04005-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2023] [Revised: 07/11/2023] [Accepted: 07/27/2023] [Indexed: 08/15/2023]
Abstract
Xanthones are natural secondary metabolites that possess great potential as neuroprotective agents due to their prominent biological effects on Alzheimer's disease (AD). However, their underlying mechanisms in AD remain unclear. This study aimed to systematically review the effects and mechanisms of xanthones in cell culture and animal studies, gaining a better understanding of their roles in AD. A comprehensive literature search was conducted in the Medline and Scopus databases using specific keywords to identify relevant articles published up to June 2023. After removing duplicates, all articles were imported into the Rayyan software. The article titles were screened based on predefined inclusion and exclusion criteria. Relevant full-text articles were assessed for biases using the OHAT tool. The results were presented in tables. Xanthones have shown various pharmacological effects towards AD from the 21 preclinical studies included. Cell culture studies demonstrated the anti-cholinesterase activity of xanthones, which protects against the loss of acetylcholine. Xanthones exhibited neuroprotective effects by promoting cell viability, reducing the accumulation of β-amyloid and tau aggregation. The administration of xanthones in animal models resulted in a reduction in neuronal inflammation by decreasing microglial and astrocyte burden. In terms of molecular mechanisms, xanthones prevented neuroinflammation through the modulation of signaling pathways, including TLR4/TAK1/NF-κB and MAPK pathways. Mechanisms such as activation of caspase-3 and -9 and suppression of endoplasmic reticulum stress were also reported. Despite the various neuroprotective effects associated with xanthones, there are limited studies reported on their underlying mechanisms in AD. Further studies are warranted to fully understand their potential roles in AD.
Collapse
Affiliation(s)
- Li Wen Pang
- School of Pharmacy, Faculty of Health and Medical Sciences, Taylor's University, 47500, Subang Jaya, Selangor, Malaysia
| | - Sharina Hamzah
- School of Pharmacy, Faculty of Health and Medical Sciences, Taylor's University, 47500, Subang Jaya, Selangor, Malaysia
- Medical Advancement for Better Quality of Life Impact Lab, Taylor's University, 47500, Subang Jaya, Selangor, Malaysia
| | - Sui Ling Janet Tan
- Department of Pharmaceutical Technology, Faculty of Pharmacy, Universiti Malaya, 50603, Kuala Lumpur, Malaysia
| | - Siau Hui Mah
- School of Biosciences, Faculty of Health and Medical Sciences, Taylor's University, 47500, Subang Jaya, Selangor, Malaysia
| | - Hui Yin Yow
- School of Pharmacy, Faculty of Health and Medical Sciences, Taylor's University, 47500, Subang Jaya, Selangor, Malaysia.
- Department of Pharmaceutical Life Sciences, Faculty of Pharmacy, Universiti Malaya, 50603, Kuala Lumpur, Malaysia.
| |
Collapse
|
6
|
Grabska-Kobyłecka I, Szpakowski P, Król A, Książek-Winiarek D, Kobyłecki A, Głąbiński A, Nowak D. Polyphenols and Their Impact on the Prevention of Neurodegenerative Diseases and Development. Nutrients 2023; 15:3454. [PMID: 37571391 PMCID: PMC10420887 DOI: 10.3390/nu15153454] [Citation(s) in RCA: 21] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2023] [Revised: 07/13/2023] [Accepted: 07/18/2023] [Indexed: 08/13/2023] Open
Abstract
It is well known that neurodegenerative diseases' development and progression are accelerated due to oxidative stress and inflammation, which result in impairment of mitochondrial function, cellular damage, and dysfunction of DNA repair systems. The increased consumption of antioxidants can postpone the development of these disorders and improve the quality of patients' lives who have already been diagnosed with neurodegenerative diseases. Prolonging life span in developed countries contributes to an increase in the incidence ratio of chronic age-related neurodegenerative disorders, such as PD (Parkinson's disease), AD (Alzheimer's disease), or numerous forms of age-related dementias. Dietary supplementation with neuroprotective plant-derived polyphenols might be considered an important element of healthy aging. Some polyphenols improve cognition, mood, visual functions, language, and verbal memory functions. Polyphenols bioavailability differs greatly from one compound to another and is determined by solubility, degree of polymerization, conjugation, or glycosylation resulting from chemical structure. It is still unclear which polyphenols are beneficial because their potential depends on efficient transport across the BBB (blood-brain barrier), bioavailability, and stability in the CNS (central nervous system). Polyphenols improve brain functions by having a direct impact on cells and processes in the CNS. For a direct effect, polyphenolic compounds must be able to overcome the BBB and accumulate in brain tissue. In this review, the latest achievements in studies (animal models and clinical trials) on the effect of polyphenols on brain activity and function are described. The beneficial impact of plant polyphenols on the brain may be summarized by their role in increasing brain plasticity and related cognition improvement. As reversible MAO (monoamine oxidase) inhibitors, polyphenols are mood modulators and improve neuronal self-being through an increase in dopamine, serotonin, and noradrenaline amounts in the brain tissue. After analyzing the prohealth effects of various eating patterns, it was postulated that their beneficial effects result from synergistic interactions between individual dietary components. Polyphenols act on the brain endothelial cells and improve the BBB's integrity and reduce inflammation, thus protecting the brain from additional injury during stroke or autoimmune diseases. Polyphenolic compounds are capable of lowering blood pressure and improving cerebral blood flow. Many studies have revealed that a nutritional model based on increased consumption of antioxidants has the potential to ameliorate the cognitive impairment associated with neurodegenerative disorders. Randomized clinical trials have also shown that the improvement of cognitive functions resulting from the consumption of foods rich in flavonoids is independent of age and health conditions. For therapeutic use, sufficient quantities of polyphenols must cross the BBB and reach the brain tissue in active form. An important issue in the direct action of polyphenols on the CNS is not only their penetration through the BBB, but also their brain metabolism and localization. The bioavailability of polyphenols is low. The most usual oral administration also conflicts with bioavailability. The main factors that limit this process and have an effect on therapeutic efficacy are: selective permeability across BBB, gastrointestinal transformations, poor absorption, rapid hepatic and colonic metabolism, and systemic elimination. Thus, phenolic compounds have inadequate bioavailability for human applications to have any beneficial effects. In recent years, new strategies have been attempted in order to exert cognitive benefits and neuroprotective effects. Converting polyphenols into nanostructures is one of the theories proposed to enhance their bioavailability. The following nanoscale delivery systems can be used to encapsulate polyphenols: nanocapsules, nanospheres, micelles, cyclodextrins, solid lipid nanoparticles, and liposomes. It results in great expectations for the wide-scale and effective use of polyphenols in the prevention of neurodegenerative diseases. Thus far, only natural polyphenols have been studied as neuroprotectors. Perhaps some modification of the chemical structure of a given polyphenol may increase its neuroprotective activity and transportation through the BBB. However, numerous questions should be answered before developing neuroprotective medications based on plant polyphenols.
Collapse
Affiliation(s)
- Izabela Grabska-Kobyłecka
- Department of Clinical Physiology, Medical University of Lodz, Mazowiecka 6/8 Street, 92-215 Łódź, Poland
| | - Piotr Szpakowski
- Department of Neurology and Stroke, Medical University of Lodz, Zeromskiego 113 Street, 90-549 Łódź, Poland; (P.S.); (D.K.-W.); (A.G.)
| | - Aleksandra Król
- Department of Experimental Physiology, Medical University of Lodz, Mazowiecka 6/8 Street, 92-215 Łódź, Poland;
| | - Dominika Książek-Winiarek
- Department of Neurology and Stroke, Medical University of Lodz, Zeromskiego 113 Street, 90-549 Łódź, Poland; (P.S.); (D.K.-W.); (A.G.)
| | - Andrzej Kobyłecki
- Interventional Cardiology Lab, Copernicus Hospital, Pabianicka Str. 62, 93-513 Łódź, Poland;
| | - Andrzej Głąbiński
- Department of Neurology and Stroke, Medical University of Lodz, Zeromskiego 113 Street, 90-549 Łódź, Poland; (P.S.); (D.K.-W.); (A.G.)
| | - Dariusz Nowak
- Department of Clinical Physiology, Medical University of Lodz, Mazowiecka 6/8 Street, 92-215 Łódź, Poland
| |
Collapse
|
7
|
Gao B, Qu YC, Cai MY, Zhang YY, Lu HT, Li HX, Tang YX, Shen H. Phytochemical interventions for post-traumatic stress disorder: A cluster co-occurrence network analysis using CiteSpace. JOURNAL OF INTEGRATIVE MEDICINE 2023:S2095-4964(23)00048-1. [PMID: 37380564 DOI: 10.1016/j.joim.2023.06.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/05/2022] [Accepted: 03/16/2023] [Indexed: 06/30/2023]
Abstract
OBJECTIVE This study investigated trends in the study of phytochemical treatment of post-traumatic stress disorder (PTSD). METHODS The Web of Science database (2007-2022) was searched using the search terms "phytochemicals" and "PTSD," and relevant literature was compiled. Network clustering co-occurrence analysis and qualitative narrative review were conducted. RESULTS Three hundred and one articles were included in the analysis of published research, which has surged since 2015 with nearly half of all relevant articles coming from North America. The category is dominated by neuroscience and neurology, with two journals, Addictive Behaviors and Drug and Alcohol Dependence, publishing the greatest number of papers on these topics. Most studies focused on psychedelic intervention for PTSD. Three timelines show an "ebb and flow" phenomenon between "substance use/marijuana abuse" and "psychedelic medicine/medicinal cannabis." Other phytochemicals account for a small proportion of the research and focus on topics like neurosteroid turnover, serotonin levels, and brain-derived neurotrophic factor expression. CONCLUSION Research on phytochemicals and PTSD is unevenly distributed across countries/regions, disciplines, and journals. Since 2015, the research paradigm shifted to constitute the mainstream of psychedelic research thus far, leading to the exploration of botanical active ingredients and molecular mechanisms. Other studies focus on anti-oxidative stress and anti-inflammation. Please cite this article as: Gao B, Qu YC, Cai MY, Zhang YY, Lu HT, Li HX, Tang YX, Shen H. Phytochemical interventions for post-traumatic stress disorder: A cluster co-occurrence network analysis using CiteSpace. J Integr Med. 2023; Epub ahead of print.
Collapse
Affiliation(s)
- Biao Gao
- Department of Naval Nutrition and Food Hygiene, Naval Medical University, Shanghai 200433, China; Teaching and Research Support Center, Naval Medical University, Shanghai 200433, China
| | - Yi-Cui Qu
- Department of Naval Nutrition and Food Hygiene, Naval Medical University, Shanghai 200433, China
| | - Meng-Yu Cai
- Department of Naval Nutrition and Food Hygiene, Naval Medical University, Shanghai 200433, China
| | - Yin-Yin Zhang
- Department of Naval Nutrition and Food Hygiene, Naval Medical University, Shanghai 200433, China
| | - Hong-Tao Lu
- Department of Naval Nutrition and Food Hygiene, Naval Medical University, Shanghai 200433, China
| | - Hong-Xia Li
- Department of Naval Nutrition and Food Hygiene, Naval Medical University, Shanghai 200433, China
| | - Yu-Xiao Tang
- Department of Naval Nutrition and Food Hygiene, Naval Medical University, Shanghai 200433, China
| | - Hui Shen
- Department of Naval Nutrition and Food Hygiene, Naval Medical University, Shanghai 200433, China.
| |
Collapse
|
8
|
Alam M, Rashid S, Fatima K, Adnan M, Shafie A, Akhtar MS, Ganie AH, Eldin SM, Islam A, Khan I, Hassan MI. Biochemical features and therapeutic potential of α-Mangostin: Mechanism of action, medicinal values, and health benefits. Biomed Pharmacother 2023; 163:114710. [PMID: 37141737 DOI: 10.1016/j.biopha.2023.114710] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2023] [Revised: 04/12/2023] [Accepted: 04/12/2023] [Indexed: 05/06/2023] Open
Abstract
α-Mangostin (α-MG) is a natural xanthone obtained from the pericarps of mangosteen. It exhibits excellent potential, including anti-cancer, neuroprotective, antimicrobial, antioxidant, and anti-inflammatory properties, and induces apoptosis. α-MG controls cell proliferation by modulating signaling molecules, thus implicated in cancer therapy. It possesses incredible pharmacological features and modulates crucial cellular and molecular factors. Due to its lesser water solubility and pitiable target selectivity, α-MG has limited clinical application. As a known antioxidant, α-MG has gained significant attention from the scientific community, increasing interest in extensive technical and biomedical applications. Nanoparticle-based drug delivery systems were designed to improve the pharmacological features and efficiency of α-MG. This review is focused on recent developments on the therapeutic potential of α-MG in managing cancer and neurological diseases, with a special focus on its mechanism of action. In addition, we highlighted biochemical and pharmacological features, metabolism, functions, anti-inflammatory, antioxidant effects and pre-clinical applications of α-MG.
Collapse
Affiliation(s)
- Manzar Alam
- Centre for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, Jamia Nagar, New Delhi 110025, India
| | - Summya Rashid
- Department of Pharmacology & Toxicology, College of Pharmacy, Prince Sattam Bin Abdulaziz University, PO Box 173, Al-kharj 11942, Saudi Arabia
| | - Kisa Fatima
- Department of Biotechnology, Jamia Millia Islamia, Jamia Nagar, New Delhi 110025, India
| | - Mohd Adnan
- Department of Biology, College of Science, University of Hail, PO Box 2440, Hail 2440, Saudi Arabia
| | - Alaa Shafie
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Taif University, Taif, Saudi Arabia
| | - Mohammad Salman Akhtar
- Department of Basic Medical Sciences, Faculty of Applied Medical Sciences, Albaha University, Albaha, Saudi Arabia
| | - A H Ganie
- Basic Sciences Department, College of Science and Theoretical Studies, Saudi Electronic University, Abha Male 61421, Saudi Arabia
| | - Sayed M Eldin
- Center of Research, Faculty of Engineering, Future University in Egypt, New Cairo 11835, Egypt
| | - Asimul Islam
- Centre for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, Jamia Nagar, New Delhi 110025, India
| | - Ilyas Khan
- Department of Mathematics, College of Science Al-Zulfi, Majmaah University, Al-Majmaah 11952, Saudi Arabia.
| | - Md Imtaiyaz Hassan
- Centre for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, Jamia Nagar, New Delhi 110025, India.
| |
Collapse
|
9
|
Tyler SEB, Tyler LDK. Therapeutic roles of plants for 15 hypothesised causal bases of Alzheimer's disease. NATURAL PRODUCTS AND BIOPROSPECTING 2022; 12:34. [PMID: 35996065 PMCID: PMC9395556 DOI: 10.1007/s13659-022-00354-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/07/2022] [Accepted: 06/15/2022] [Indexed: 05/26/2023]
Abstract
Alzheimer's disease (AD) is progressive and ultimately fatal, with current drugs failing to reverse and cure it. This study aimed to find plant species which may provide therapeutic bioactivities targeted to causal agents proposed to be driving AD. A novel toolkit methodology was employed, whereby clinical symptoms were translated into categories recognized in ethnomedicine. These categories were applied to find plant species with therapeutic effects, mined from ethnomedical surveys. Survey locations were mapped to assess how this data is at risk. Bioactivities were found of therapeutic relevance to 15 hypothesised causal bases for AD. 107 species with an ethnological report of memory improvement demonstrated therapeutic activity for all these 15 causal bases. The majority of the surveys were found to reside within biodiversity hotspots (centres of high biodiversity under threat), with loss of traditional knowledge the most common threat. Our findings suggest that the documented plants provide a large resource of AD therapeutic potential. In demonstrating bioactivities targeted to these causal bases, such plants may have the capacity to reduce or reverse AD, with promise as drug leads to target multiple AD hallmarks. However, there is a need to preserve ethnomedical knowledge, and the habitats on which this knowledge depends.
Collapse
Affiliation(s)
| | - Luke D K Tyler
- School of Natural Sciences, Bangor University, Gwynedd, UK
| |
Collapse
|
10
|
Hu X, Liu C, Wang K, Zhao L, Qiu Y, Chen H, Hu J, Xu J. Multifunctional Anti-Alzheimer’s Disease Effects of Natural Xanthone Derivatives: A Primary Structure-Activity Evaluation. Front Chem 2022; 10:842208. [PMID: 35646819 PMCID: PMC9130743 DOI: 10.3389/fchem.2022.842208] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Accepted: 04/26/2022] [Indexed: 12/15/2022] Open
Abstract
Background: A series of α-Mangostin (α-M) derivatives were designed and synthesized. α-M and four analogues were evaluated for their multifunctional anti-Alzheimer’s disease (anti-AD) effects on fibrillogenesis, microglial uptake, microglial degradation, and anti-neurotoxicity of Aβ, as well as LPS-induced neuroinflammation. The differences in bioactivities were analyzed to understand the structure-activity relationship for further modifications. Purpose: This study aims to investigate the anti-AD effects of α-M and elucidate its structure-activity relationship by comparing difference between α-M and several analogues. Methods: Aβ fibrillogenesis was detected by Thioflavin T fluorometric assay. The levels of Aβ1-42 and inflammatory cytokines were evaluated by enzyme-linked immunosorbent assay. Neuron viability was examined by the CCK-8 assay. The morphology of ZO-1 of bEnd.3 cultured in BV-2-conditioned medium was evaluated by immunofluorescence staining. Results: Aβ fibrillogenesis was significantly inhibited by co-incubation with α-M, Zcbd-2 or Zcbd-3. α-M, Zcbd-2, Zcbd-3, and Zcbd-4 decreased the levels of Aβ1-42 and inflammatory cytokines, and promoted Aβ uptake, degradation and anti-inflammation effects inflammation in microglia. α-M and Zcbd-3 protected neuron viability from Aβ-induced neurotoxicity, and preserved tight junction integrity of bEnd.3 against LPS-induced neuroinflammation. Conclusion: Zcbd-3 acted as α-M almost in all effects. The structure-activity analysis indicated that the 3-methyl-2-butenyl group at C-8 is essential for the bioactivity of α-M, while modifying the double hydroxylation at the C-2 position may improve the multifunctional anti-AD effects.
Collapse
Affiliation(s)
- Xiaoyu Hu
- Academy of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Department of Pharmacology and Chemical Biology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Chan Liu
- Department of Pharmacology and Chemical Biology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Joint International Research Laboratory of Ethnomedicine of Ministry of Education and Key Laboratory of Basic Pharmacology of Ministry of Education, Zunyi Medical University, Zunyi, China
| | - Kaichun Wang
- Academy of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Lanxue Zhao
- Department of Pharmacology and Chemical Biology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yu Qiu
- Department of Pharmacology and Chemical Biology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Hongzhuan Chen
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Jiangmiao Hu
- State Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming, China
- *Correspondence: Jiangmiao Hu, ; Jianrong Xu,
| | - Jianrong Xu
- Academy of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Department of Pharmacology and Chemical Biology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- *Correspondence: Jiangmiao Hu, ; Jianrong Xu,
| |
Collapse
|
11
|
Khamphukdee C, Turkmani I, Chotritthirong Y, Chulikhit Y, Boonyarat C, Sekeroglu N, Silva AMS, Monthakantirat O, Kijjoa A. Effects of the Bark Resin Extract of Garcinia nigrolineata on Chronic Stress-Induced Memory Deficit in Mice Model and the In Vitro Monoamine Oxidases and β-Amyloid Aggregation Inhibitory Activities of Its Prenylated Xanthone Constituents. Molecules 2022; 27:molecules27093014. [PMID: 35566362 PMCID: PMC9103351 DOI: 10.3390/molecules27093014] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Revised: 05/05/2022] [Accepted: 05/06/2022] [Indexed: 12/04/2022] Open
Abstract
The present study describes investigation of the effects of the bark resin extract of Garcinia nigrolineata (Clusiaceae) on the cognitive function and the induction of oxidative stress in both frontal cortex and hippocampus by unpredictable chronic mild stress (UCMS). By using behavioral mouse models, i.e., the Y-maze test, the Novel Object Recognition Test (NORT), and the Morris Water Maze Test (MWMT), it was found that the negative impact of repeated mild stress-induced learning and memory deficit through brain oxidative stress in the UCMS mice was reversed by treatment with the bark resin extract G. nigrolineata. Moreover, the prenylated xanthones viz. cowagarcinone C, cowaxanthone, α-mangostin, cowaxanthone B, cowanin, fuscaxanthone A, fuscaxanthone B, xanthochymusxanthones A, 7-O-methylgarcinone E, and cowagarcinone A, isolated from the bark resin of G. nigrolineata, were assayed for their inhibitory activities against β-amyloid (Aβ) aggregation and monoamine oxidase enzymes (MAOs).
Collapse
Affiliation(s)
- Charinya Khamphukdee
- Division of Pharmacognosy and Toxicology, Faculty of Pharmaceutical Sciences, Khon Kaen University, Khon Kaen 40002, Thailand;
| | - Ibrahim Turkmani
- ICBAS-Instituo de Ciências Biomédicas Abel Salazar and CIIMAR, Universidade do Porto, Rua de Jorge Viterbo Ferreira 228, 4050-313 Porto, Portugal;
| | - Yutthana Chotritthirong
- Graduate School of Pharmaceutical Sciences, Khon Kaen University, Khon Kaen 40002, Thailand;
| | - Yaowared Chulikhit
- Division of Pharmaceutical Chemistry, Faculty of Pharmaceutical Sciences, Khon Kaen University, Khon Kaen 40002, Thailand; (Y.C.); (C.B.)
| | - Chantana Boonyarat
- Division of Pharmaceutical Chemistry, Faculty of Pharmaceutical Sciences, Khon Kaen University, Khon Kaen 40002, Thailand; (Y.C.); (C.B.)
| | - Nazim Sekeroglu
- Phytotherapy, Medicinal and Aromatic Plants Application & Research Center and Biology Department, Faculty of Arts and Science, Gaziantep University, 27310 Gaziantep, Turkey;
| | - Artur M. S. Silva
- Departamento de Química & QOPNA, Universidade de Aveiro, 3810-193 Aveiro, Portugal;
| | - Orawan Monthakantirat
- Division of Pharmaceutical Chemistry, Faculty of Pharmaceutical Sciences, Khon Kaen University, Khon Kaen 40002, Thailand; (Y.C.); (C.B.)
- Correspondence: (O.M.); (A.K.); Tel.: +66-81-3404677 (O.M.); +351-220428331 (A.K.)
| | - Anake Kijjoa
- ICBAS-Instituo de Ciências Biomédicas Abel Salazar and CIIMAR, Universidade do Porto, Rua de Jorge Viterbo Ferreira 228, 4050-313 Porto, Portugal;
- Correspondence: (O.M.); (A.K.); Tel.: +66-81-3404677 (O.M.); +351-220428331 (A.K.)
| |
Collapse
|
12
|
de Sousa NF, Scotti L, de Moura ÉP, dos Santos Maia M, Soares Rodrigues GC, de Medeiros HIR, Lopes SM, Scotti MT. Computer Aided Drug Design Methodologies with Natural Products in the Drug Research Against Alzheimer's Disease. Curr Neuropharmacol 2022; 20:857-885. [PMID: 34636299 PMCID: PMC9881095 DOI: 10.2174/1570159x19666211005145952] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2021] [Revised: 08/19/2021] [Accepted: 08/26/2021] [Indexed: 11/22/2022] Open
Abstract
Natural products are compounds isolated from plants that provide a variety of lead structures for the development of new drugs by the pharmaceutical industry. The interest in these substances increases because of their beneficial effects on human health. Alzheimer's disease (AD) affects occur in about 80% of individuals aged 65 years. AD, the most common cause of dementia in elderly people, is characterized by progressive neurodegenerative alterations, as decrease of cholinergic impulse, increased toxic effects caused by reactive oxygen species and the inflammatory process that the amyloid plaque participates. In silico studies is relevant in the process of drug discovery; through technological advances in the areas of structural characterization of molecules, computational science and molecular biology have contributed to the planning of new drugs used against neurodegenerative diseases. Considering the social impairment caused by an increased incidence of disease and that there is no chemotherapy treatment effective against AD; several compounds are studied. In the researches for effective neuroprotectants as potential treatments for Alzheimer's disease, natural products have been extensively studied in various AD models. This study aims to carry out a literature review with articles that address the in silico studies of natural products aimed at potential drugs against Alzheimer's disease (AD) in the period from 2015 to 2021.
Collapse
Affiliation(s)
- Natália Ferreira de Sousa
- Postgraduate Program of Natural and Synthetic Bioactive Products (PgPNSB), Health Sciences Center, Federal University of Paraíba, João Pessoa-PB, Brazil
| | - Luciana Scotti
- Postgraduate Program of Natural and Synthetic Bioactive Products (PgPNSB), Health Sciences Center, Federal University of Paraíba, João Pessoa-PB, Brazil;,Lauro Wanderley University Hospital (HULW), Health Sciences Center, Federal University of Paraíba, João Pessoa-PB, Brazil,Address correspondence to this author at the Health Sciences Center, Chemioinformatic Laboratory, Federal University of Paraíba, Paraíba, Brazil; E-mail:
| | - Érika Paiva de Moura
- Postgraduate Program of Natural and Synthetic Bioactive Products (PgPNSB), Health Sciences Center, Federal University of Paraíba, João Pessoa-PB, Brazil
| | - Mayara dos Santos Maia
- Postgraduate Program of Natural and Synthetic Bioactive Products (PgPNSB), Health Sciences Center, Federal University of Paraíba, João Pessoa-PB, Brazil
| | - Gabriela Cristina Soares Rodrigues
- Postgraduate Program of Natural and Synthetic Bioactive Products (PgPNSB), Health Sciences Center, Federal University of Paraíba, João Pessoa-PB, Brazil
| | - Herbert Igor Rodrigues de Medeiros
- Postgraduate Program of Natural and Synthetic Bioactive Products (PgPNSB), Health Sciences Center, Federal University of Paraíba, João Pessoa-PB, Brazil
| | - Simone Mendes Lopes
- Postgraduate Program of Natural and Synthetic Bioactive Products (PgPNSB), Health Sciences Center, Federal University of Paraíba, João Pessoa-PB, Brazil
| | - Marcus Tullius Scotti
- Lauro Wanderley University Hospital (HULW), Health Sciences Center, Federal University of Paraíba, João Pessoa-PB, Brazil
| |
Collapse
|
13
|
Meylina L, Muchtaridi M, Joni IM, Mohammed AFA, Wathoni N. Nanoformulations of α-Mangostin for Cancer Drug Delivery System. Pharmaceutics 2021; 13:1993. [PMID: 34959275 PMCID: PMC8708633 DOI: 10.3390/pharmaceutics13121993] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Revised: 11/07/2021] [Accepted: 11/08/2021] [Indexed: 12/24/2022] Open
Abstract
Natural compounds are emerging as effective agents for the treatment of malignant diseases. The active constituent of α-mangostin from the pericarp of Garcinia mangostana L. has earned significant interest as a plant base compound with anticancer properties. Despite α-mangostin's superior properties as an anticancer agent, its applications are limited due to its poor solubility and physicochemical stability, rapid systemic clearance, and low cellular uptake. Our review aimed to summarize and discuss the nanoparticle formulations of α-mangostin for cancer drug delivery systems from published papers recorded in Scopus, PubMed, and Google Scholar. We investigated various types of α-mangostin nanoformulations to improve its anticancer efficacy by improving bioavailability, cellular uptake, and localization to specific areas These nanoformulations include nanofibers, lipid carrier nanostructures, solid lipid nanoparticles, polymeric nanoparticles, nanomicelles, liposomes, and gold nanoparticles. Notably, polymeric nanoparticles and nanomicelles can increase the accumulation of α-mangostin into tumors and inhibit tumor growth in vivo. In addition, polymeric nanoparticles with the addition of target ligands can increase the cellular uptake of α-mangostin. In conclusion, nanoformulations of α-mangostin are a promising tool to enhance the cellular uptake, accumulation in cancer cells, and the efficacy of α-mangostin as a candidate for anticancer drugs.
Collapse
Affiliation(s)
- Lisna Meylina
- Department of Pharmaceutics and Pharmaceutical Technology, Faculty of Pharmacy, Universitas Padjadjaran, Sumedang 45363, Indonesia;
- Department of Pharmaceutics and Pharmaceutical Technology, Faculty of Pharmacy, Universitas Mulawarman, Samarinda 75119, Indonesia
| | - Muchtaridi Muchtaridi
- Department of Pharmaceutical Analysis and Medicinal Chemistry, Faculty of Pharmacy, Universitas Padjadjaran, Sumedang 45363, Indonesia;
| | - I Made Joni
- Department of Physics, Faculty of Mathematics and Natural Sciences, Universitas Padjadjaran, Sumedang 45363, Indonesia;
- Functional Nano Powder University Center of Excellence, Universitas Padjadjaran, Sumedang 45363, Indonesia
| | | | - Nasrul Wathoni
- Department of Pharmaceutics and Pharmaceutical Technology, Faculty of Pharmacy, Universitas Padjadjaran, Sumedang 45363, Indonesia;
| |
Collapse
|
14
|
Riccardi C, Napolitano F, Montesarchio D, Sampaolo S, Melone MAB. Nanoparticle-Guided Brain Drug Delivery: Expanding the Therapeutic Approach to Neurodegenerative Diseases. Pharmaceutics 2021; 13:1897. [PMID: 34834311 PMCID: PMC8623286 DOI: 10.3390/pharmaceutics13111897] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2021] [Revised: 10/31/2021] [Accepted: 11/04/2021] [Indexed: 02/07/2023] Open
Abstract
Neurodegenerative diseases (NDs) represent a heterogeneous group of aging-related disorders featured by progressive impairment of motor and/or cognitive functions, often accompanied by psychiatric disorders. NDs are denoted as 'protein misfolding' diseases or proteinopathies, and are classified according to their known genetic mechanisms and/or the main protein involved in disease onset and progression. Alzheimer's disease (AD), Parkinson's disease (PD) and Huntington's disease (HD) are included under this nosographic umbrella, sharing histopathologically salient features, including deposition of insoluble proteins, activation of glial cells, loss of neuronal cells and synaptic connectivity. To date, there are no effective cures or disease-modifying therapies for these NDs. Several compounds have not shown efficacy in clinical trials, since they generally fail to cross the blood-brain barrier (BBB), a tightly packed layer of endothelial cells that greatly limits the brain internalization of endogenous substances. By engineering materials of a size usually within 1-100 nm, nanotechnology offers an alternative approach for promising and innovative therapeutic solutions in NDs. Nanoparticles can cross the BBB and release active molecules at target sites in the brain, minimizing side effects. This review focuses on the state-of-the-art of nanoengineered delivery systems for brain targeting in the treatment of AD, PD and HD.
Collapse
Affiliation(s)
- Claudia Riccardi
- Department of Chemical Sciences, University of Naples Federico II, Via Cintia 21, I-80126 Naples, Italy; (C.R.); (D.M.)
| | - Filomena Napolitano
- Department of Advanced Medical and Surgical Sciences, 2nd Division of Neurology, Center for Rare Diseases and InterUniversity Center for Research in Neurosciences, University of Campania Luigi Vanvitelli, Via Sergio Pansini, 5, I-80131 Naples, Italy; (F.N.); (S.S.)
| | - Daniela Montesarchio
- Department of Chemical Sciences, University of Naples Federico II, Via Cintia 21, I-80126 Naples, Italy; (C.R.); (D.M.)
| | - Simone Sampaolo
- Department of Advanced Medical and Surgical Sciences, 2nd Division of Neurology, Center for Rare Diseases and InterUniversity Center for Research in Neurosciences, University of Campania Luigi Vanvitelli, Via Sergio Pansini, 5, I-80131 Naples, Italy; (F.N.); (S.S.)
| | - Mariarosa Anna Beatrice Melone
- Department of Advanced Medical and Surgical Sciences, 2nd Division of Neurology, Center for Rare Diseases and InterUniversity Center for Research in Neurosciences, University of Campania Luigi Vanvitelli, Via Sergio Pansini, 5, I-80131 Naples, Italy; (F.N.); (S.S.)
- Sbarro Institute for Cancer Research and Molecular Medicine, Center for Biotechnology, Temple University, Philadelphia, PA 19122-6078, USA
| |
Collapse
|
15
|
Wang Q, Dong X, Zhang R, Zhao C. Flavonoids with Potential Anti-Amyloidogenic Effects as Therapeutic Drugs for Treating Alzheimer's Disease. J Alzheimers Dis 2021; 84:505-533. [PMID: 34569961 DOI: 10.3233/jad-210735] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Alzheimer's disease (AD) is a central neurodegenerative disease generally among the elderly; it accounts for approximately 50-75%of total cases of dementia patients and poses a serious threat to physical and mental health. Currently available treatments for AD mainly relieves its symptoms, and effective therapy is urgently needed. Deposition of amyloid-β protein in the brain is an early and invariant neuropathological feature of AD. Currently the main efforts in developing anti-AD drugs focus on anti-amyloidogenic therapeutics that prevent amyloid-β production or aggregation and decrease the occurrence of neurotoxic events. The results of an increasing number of studies suggest that natural extracts and phytochemicals have a positive impact on brain aging. Flavonoids belong to the broad group of polyphenols and recent data indicate a favorable effect of flavonoids on brain aging. In this review, we collect relevant discoveries from 1999 to 2021, discuss 75 flavonoids that effectively influence AD pathogenesis, and summarize their functional mechanisms in detail. The data we have reviewed show that, these flavonoids belong to various subclasses, including flavone, flavanone, biflavone, etc. Our results provide a reference for further study of the effects of flavonoids on AD and the progress of anti-AD therapy.
Collapse
Affiliation(s)
- Qixin Wang
- Gene Engineering and Biotechnology Beijing Key Laboratory, College of Life Science, Beijing Normal University, Beijing, China
| | - Xiaofang Dong
- Gene Engineering and Biotechnology Beijing Key Laboratory, College of Life Science, Beijing Normal University, Beijing, China
| | - Ran Zhang
- Gene Engineering and Biotechnology Beijing Key Laboratory, College of Life Science, Beijing Normal University, Beijing, China
| | - Changqi Zhao
- Gene Engineering and Biotechnology Beijing Key Laboratory, College of Life Science, Beijing Normal University, Beijing, China
| |
Collapse
|
16
|
Ndongo TM, Passos MLC, Durães F, Resende DISP, Pinto M, Sousa E, Saraiva MLMFS. Microsequential injection analysis/lab-on-valve system for the automatic evaluation of acetylcholinesterase inhibitors. Arch Pharm (Weinheim) 2021; 354:e2100150. [PMID: 34105191 DOI: 10.1002/ardp.202100150] [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: 04/19/2021] [Revised: 05/18/2021] [Accepted: 05/22/2021] [Indexed: 11/08/2022]
Abstract
A miniaturized microsequential injection/lab-on-valve (µSIA-LOV) system was developed and shown to be a useful alternative to perform inhibitory studies on acetylcholinesterase. These studies are essential for the evaluation of the potential therapeutic effect of drugs commonly used in the treatment of Alzheimer's disease. Donepezil, galantamine, and rivastigmine were tested, in addition to compounds based on the xanthone scaffold. Four of these xanthone derivatives were identified as having EC50 values between 676 and 4466 µmol/l, showing a potential inhibitory effect higher than the clinical agent rivastigmine. The developed automatic system added advantages of reduction of reagents and sample consumption (around 55 µl per analysis), lower cost per analysis, and the generation of less waste (around 1.2 ml per analysis). The µSIA-LOV system is also a robust, rapid, reliable, and simple system to use. Docking studies suggested a possible mode of interaction with the target acetylcholinesterase protein.
Collapse
Affiliation(s)
- Thierno Moussa Ndongo
- LAQV, REQUIMTE, Laboratory of Applied Chemistry, Department of Chemical Sciences, Faculty of Pharmacy, Porto University, Porto, Portugal.,Faculté des sciences et techniques, Le Mans Université, Le Mans, France
| | - Marieta L C Passos
- LAQV, REQUIMTE, Laboratory of Applied Chemistry, Department of Chemical Sciences, Faculty of Pharmacy, Porto University, Porto, Portugal
| | - Fernando Durães
- Laboratório de Química Orgânica e Farmacêutica, Departamento de Ciências Químicas, Faculdade de Farmácia, Universidade do Porto, Porto, Portugal.,Centro Interdisciplinar de Investigação Marinha e Ambiental (CIIMAR/CIMAR), Universidade do Porto, Matosinhos, Portugal
| | - Diana I S P Resende
- Laboratório de Química Orgânica e Farmacêutica, Departamento de Ciências Químicas, Faculdade de Farmácia, Universidade do Porto, Porto, Portugal.,Centro Interdisciplinar de Investigação Marinha e Ambiental (CIIMAR/CIMAR), Universidade do Porto, Matosinhos, Portugal
| | - Madalena Pinto
- Laboratório de Química Orgânica e Farmacêutica, Departamento de Ciências Químicas, Faculdade de Farmácia, Universidade do Porto, Porto, Portugal.,Centro Interdisciplinar de Investigação Marinha e Ambiental (CIIMAR/CIMAR), Universidade do Porto, Matosinhos, Portugal
| | - Emília Sousa
- Laboratório de Química Orgânica e Farmacêutica, Departamento de Ciências Químicas, Faculdade de Farmácia, Universidade do Porto, Porto, Portugal.,Centro Interdisciplinar de Investigação Marinha e Ambiental (CIIMAR/CIMAR), Universidade do Porto, Matosinhos, Portugal
| | - Maria Lúcia M F S Saraiva
- LAQV, REQUIMTE, Laboratory of Applied Chemistry, Department of Chemical Sciences, Faculty of Pharmacy, Porto University, Porto, Portugal
| |
Collapse
|
17
|
Li H, Xu J, Li X, Hu Y, Liao Y, Zhou W, Song Z. Anti-inflammatory activity of psoralen in human periodontal ligament cells via estrogen receptor signaling pathway. Sci Rep 2021; 11:8754. [PMID: 33888745 PMCID: PMC8062431 DOI: 10.1038/s41598-021-85145-1] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2020] [Accepted: 02/25/2021] [Indexed: 12/11/2022] Open
Abstract
Psoralen is one of the most effective ingredients extracted from the Chinese herb, Psoralea corylifolia L. Studies have found that psoralen has anti-inflammatory and estrogen-like effects; however, little research has been conducted to elucidate the mechanisms underlying these effects. Through the molecule docking assay, psoralen was found to have a better combination with ERα than ERβ. In human periodontal ligament cells, psoralen was found to upregulate the estrogen target genes (e.g., CTSD, PGR, TFF1) and down-regulate the expression of inflammatory cytokines (TNF-α, IL-1β, IL-6 and IL-8) stimulated by P. gingivalis LPS, as well as TLR4-IRAK4-NF-κb signaling pathway proteins. These effects were reversed by the ER antagonist ICI 182780. These results indicated that psoralen may exert anti-inflammatory effects as an agonist to ER, which could provide a theoretical basis for the use of psoralen for adjuvant therapy and prevention of periodontitis.
Collapse
Affiliation(s)
- Huxiao Li
- Department of Periodontology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China.,College of Stomatology, Shanghai Jiao Tong University; National Center for Stomatology; National Clinical Research Center for Oral Diseases; Shanghai Key Laboratory of Stomatology; Research Unit of Oral and Maxillofacial Regenerative Medicine, Chinese Academy of Medical Sciences, Shanghai, 200011, China
| | - Jianrong Xu
- Academy of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China.,Department of Pharmacology and Chemical Biology, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Xiaotian Li
- Department of Periodontology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China.,College of Stomatology, Shanghai Jiao Tong University; National Center for Stomatology; National Clinical Research Center for Oral Diseases; Shanghai Key Laboratory of Stomatology; Research Unit of Oral and Maxillofacial Regenerative Medicine, Chinese Academy of Medical Sciences, Shanghai, 200011, China
| | - Yi Hu
- Department of Periodontology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China.,College of Stomatology, Shanghai Jiao Tong University; National Center for Stomatology; National Clinical Research Center for Oral Diseases; Shanghai Key Laboratory of Stomatology; Research Unit of Oral and Maxillofacial Regenerative Medicine, Chinese Academy of Medical Sciences, Shanghai, 200011, China
| | - Yue Liao
- Department of Periodontology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China.,College of Stomatology, Shanghai Jiao Tong University; National Center for Stomatology; National Clinical Research Center for Oral Diseases; Shanghai Key Laboratory of Stomatology; Research Unit of Oral and Maxillofacial Regenerative Medicine, Chinese Academy of Medical Sciences, Shanghai, 200011, China
| | - Wei Zhou
- Laboratory of Oral Microbiota and Systemic Diseases, Shanghai Research Institute of Stomatology,Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China. .,College of Stomatology, Shanghai Jiao Tong University; National Center for Stomatology; National Clinical Research Center for Oral Diseases; Shanghai Key Laboratory of Stomatology; Research Unit of Oral and Maxillofacial Regenerative Medicine, Chinese Academy of Medical Sciences, Shanghai, 200011, China.
| | - Zhongchen Song
- Department of Periodontology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China. .,College of Stomatology, Shanghai Jiao Tong University; National Center for Stomatology; National Clinical Research Center for Oral Diseases; Shanghai Key Laboratory of Stomatology; Research Unit of Oral and Maxillofacial Regenerative Medicine, Chinese Academy of Medical Sciences, Shanghai, 200011, China.
| |
Collapse
|
18
|
Nale SD, Maiti D, Lee YR. Construction of Highly Functionalized Xanthones via Rh-Catalyzed Cascade C-H Activation/ O-Annulation. Org Lett 2021; 23:2465-2470. [PMID: 33719464 DOI: 10.1021/acs.orglett.1c00391] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
A facile and efficient strategy for obtaining functionalized and multihydroxylated xanthones via Rh catalysis under redox-neutral conditions is developed. Diverse salicylaldehydes bearing heterocycles, aromatics, and fused aromatics can be rapidly coupled with 1,4-benzoquinones or 1,4-hydroquinones to afford valuable xanthones via cascade C-H/O-H functionalization and annulation. This protocol provides a rapid synthetic approach to obtain biologically active materials through late-stage functionalization and prepares natural products such as subelliptenone, pruniflorone N, and ravenelin.
Collapse
Affiliation(s)
- Sagar D Nale
- School of Chemical Engineering, Yeungnam University, Gyeongsan 38541, Republic of Korea
| | - Debabrata Maiti
- Department of Chemistry, IIT Bombay, Powai, Mumbai 400076, India
| | - Yong Rok Lee
- School of Chemical Engineering, Yeungnam University, Gyeongsan 38541, Republic of Korea
| |
Collapse
|
19
|
Tiwari A, Khera R, Rahi S, Mehan S, Makeen HA, Khormi YH, Rehman MU, Khan A. Neuroprotective Effect of α-Mangostin in the Ameliorating Propionic Acid-Induced Experimental Model of Autism in Wistar Rats. Brain Sci 2021; 11:288. [PMID: 33669120 PMCID: PMC7996534 DOI: 10.3390/brainsci11030288] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2021] [Revised: 02/09/2021] [Accepted: 02/23/2021] [Indexed: 02/07/2023] Open
Abstract
Several studies have documented the role of hyper-activation of extracellular signal-regulated kinases (ERK) in Autism pathogenesis. Alpha-mangostin (AMG) is a phytoconstituents with anti-oxidants, anti-inflammatory, and ERK inhibition properties in many diseases. Our research aims to investigate the neuroprotective effect of AMG in the rat model of intracerebroventricular-propionic acid (ICV-PPA) induced autism with a confirmation of its effect on the ERK signaling. Autism was induced in Wistar rats (total 36 rats; 18 male/18 female) by multiple doses of PPA through ICV injection for 11 days. Actophotometer and beam walking tasks were used to evaluate animals' motor abilities, and the Morris water maze task was utilized to confirm the cognition and memory in animals. Long term administration of AMG 100 mg/kg and AMG 200mg/kg continued from day 12 to day 44 of the experiment. Before that, animals were sacrificed, brains isolated, morphological, gross pathological studies were performed, and neurochemical analysis was performed in the brain homogenates. Cellular and molecular markers, including ERK, myelin basic protein, apoptotic markers including caspase-3, Bax, Bcl-2, neuroinflammatory markers, neurotransmitters, and oxidative stress markers, have been tested throughout the brain. Thus, AMG reduces the overactivation of the ERK signaling and also restored autism-like behavioral and neurochemical alterations.
Collapse
Affiliation(s)
- Aarti Tiwari
- Department of Pharmacology, Neuropharmacology Division, ISF College of Pharmacy, Moga, Punjab 142001, India; (A.T.); (R.K.); (S.R.)
| | - Rishabh Khera
- Department of Pharmacology, Neuropharmacology Division, ISF College of Pharmacy, Moga, Punjab 142001, India; (A.T.); (R.K.); (S.R.)
| | - Saloni Rahi
- Department of Pharmacology, Neuropharmacology Division, ISF College of Pharmacy, Moga, Punjab 142001, India; (A.T.); (R.K.); (S.R.)
| | - Sidharth Mehan
- Department of Pharmacology, Neuropharmacology Division, ISF College of Pharmacy, Moga, Punjab 142001, India; (A.T.); (R.K.); (S.R.)
| | - Hafiz Antar Makeen
- Department of Clinical Pharmacy, College of Pharmacy, Jazan University, Jazan 45142, Saudi Arabia;
| | - Yahya H. Khormi
- Division of Neurosurgery, Department of Surgery, Faculty of Medicine, Jazan University, Jazan 45142, Saudi Arabia;
| | - Muneeb U Rehman
- Department of Clinical Pharmacy, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia;
| | - Andleeb Khan
- Department of Pharmacology & Toxicology, College of Pharmacy, Jazan University, Jazan 45142, Saudi Arabia
| |
Collapse
|
20
|
A review on α-mangostin as a potential multi-target-directed ligand for Alzheimer's disease. Eur J Pharmacol 2021; 897:173950. [PMID: 33607107 DOI: 10.1016/j.ejphar.2021.173950] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2020] [Revised: 02/03/2021] [Accepted: 02/12/2021] [Indexed: 12/20/2022]
Abstract
Alzheimer's disease (AD) is an age-related neurodegenerative disease characterized by progressive memory loss, declining language skills and other cognitive disorders. AD has brought great mental and economic burden to patients, families and society. However due to the complexity of AD's pathology, drugs developed for the treatment of AD often fail in clinical or experimental trials. The main problems of current anti-AD drugs are low efficacy due to mono-target method or side effects, especially high hepatotoxicity. To tackle these two main problems, multi-target-directed ligand (MTDL) based on "one molecule, multiple targets" has been studied. MTDLs can regulate multiple biological targets at the same time, so it has shown higher efficacy, better safety. As a natural active small molecule, α-mangostin (α-M) has shown potential multi-factor anti-AD activities in a series of studies, furthermore it also has a certain hepatoprotective effect. The good availability of α-M also provides support for its application in clinical research. In this work, multiple activities of α-M related to AD therapy were reviewed, which included anti-cholinesterase, anti-amyloid-cascade, anti-inflammation, anti-oxidative stress, low toxicity, hepatoprotective effects and drug formulation. It shows that α-M is a promising candidate for the treatment of AD.
Collapse
|
21
|
Wu M, Lu G, Lao YZ, Zhang H, Zheng D, Zheng ZQ, Yi J, Xiang Q, Wang LM, Tan HS, Zhou H, Shen HM, Xu HX. Garciesculenxanthone B induces PINK1-Parkin-mediated mitophagy and prevents ischemia-reperfusion brain injury in mice. Acta Pharmacol Sin 2021; 42:199-208. [PMID: 32759963 PMCID: PMC8026581 DOI: 10.1038/s41401-020-0480-9] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2020] [Accepted: 07/07/2020] [Indexed: 12/14/2022] Open
Abstract
Mitophagy is a selective form of autophagy involving the removal of damaged mitochondria via the autophagy-lysosome pathway. PINK1-Parkin-mediated mitophagy is one of the most important mechanisms in cardiovascular disease, cerebral ischemia-reperfusion (I/R) injury, and neurodegenerative diseases. In this study we conducted an image-based screening in YFP-Parkin HeLa cells to discover new mitophagy regulators from natural xanthone compounds. We found that garciesculenxanthone B (GeB), a new xanthone compound from Garcinia esculenta, induced the formation of YFP-Parkin puncta, a well known mitophagy marker. Furthermore, treatment with GeB dose-dependently promoted the degradation of mitochondrial proteins Tom20, Tim23, and MFN1 in YFP-Parkin HeLa cells and SH-SY5Y cells. We revealed that GeB stabilized PINK1 and triggered Parkin translocation to the impaired mitochondria to induce mitophagy, and these effects were abolished by knockdown of PINK1. Finally, in vivo experiments demonstrated that GeB partially rescued ischemia-reperfusion-induced brain injury in mice. Taken together, our findings demonstrate that the natural compound GeB can promote the PINK1-Parkin-mediated mitophagy pathway, which may be implicated in protection against I/R brain injury.
Collapse
Affiliation(s)
- Man Wu
- Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Guang Lu
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Yuan-Zhi Lao
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Hong Zhang
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Dan Zheng
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Zhao-Qing Zheng
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Juan Yi
- School of Basic Medical Science, Lanzhou University, Lanzhou, 730000, China
| | - Qian Xiang
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Li-Ming Wang
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Hong-Sheng Tan
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Hua Zhou
- Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Han-Ming Shen
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.
- Faculty of Health Sciences, University of Macau, Macau, China.
| | - Hong-Xi Xu
- Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China.
| |
Collapse
|
22
|
Cascione M, De Matteis V, Leporatti S, Rinaldi R. The New Frontiers in Neurodegenerative Diseases Treatment: Liposomal-Based Strategies. Front Bioeng Biotechnol 2020; 8:566767. [PMID: 33195128 PMCID: PMC7649361 DOI: 10.3389/fbioe.2020.566767] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2020] [Accepted: 09/14/2020] [Indexed: 12/12/2022] Open
Abstract
In the last decade, the onset of neurodegenerative (ND) diseases is strongly widespread due to the age increase of the world population. Despite the intensive investigations boosted by the scientific community, an efficacious therapy has not been outlined yet. The drugs commonly used are only able to relieve symptom severity; following their oral or intravenous administration routes, their effectiveness is strictly limited due to their low ability to reach the Central Nervous System (CNS) overcoming the Blood Brain Barrier (BBB). Starting from these assumptions, the engineered-nanocarriers, such as lipid-nanocarriers, are suitable agents to enhance the delivery of drugs into the CNS due to their high solubility, bioavailability, and stability. Liposomal delivery systems are considered to be the ideal carriers, not only for conventional drugs but also for neuroprotective small molecules and green-extracted compounds. In the current work, the LP-based drug delivery improvements in in vivo applications against ND disorders were carefully assessed.
Collapse
Affiliation(s)
- Mariafrancesca Cascione
- Department of Mathematics and Physics "Ennio De Giorgi," University of Salento, Lecce, Italy
| | - Valeria De Matteis
- Department of Mathematics and Physics "Ennio De Giorgi," University of Salento, Lecce, Italy
| | - Stefano Leporatti
- National Research Council Nanotec Institute of Nanotechnology, Lecce, Italy
| | - Rosaria Rinaldi
- Department of Mathematics and Physics "Ennio De Giorgi," University of Salento, Lecce, Italy
| |
Collapse
|
23
|
Do HTT, Cho J. Mangosteen Pericarp and Its Bioactive Xanthones: Potential Therapeutic Value in Alzheimer's Disease, Parkinson's Disease, and Depression with Pharmacokinetic and Safety Profiles. Int J Mol Sci 2020; 21:E6211. [PMID: 32867357 PMCID: PMC7504283 DOI: 10.3390/ijms21176211] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2020] [Revised: 08/22/2020] [Accepted: 08/25/2020] [Indexed: 12/11/2022] Open
Abstract
Alzheimer's disease (AD), Parkinson's disease (PD), and depression are growing burdens for society globally, partly due to a lack of effective treatments. Mangosteen (Garcinia mangostana L.,) pericarp (MP) and its xanthones may provide therapeutic advantages for these disorders. In this review, we discuss potential therapeutic value of MP-derived agents in AD, PD, and depression with their pharmacokinetic and safety profiles. MP-derived agents have shown multifunctional effects including neuroprotective, antioxidant, and anti-neuroinflammatory actions. In addition, they target specific disease pathologies, such as amyloid beta production and deposition as well as cholinergic dysfunction in AD; α-synuclein aggregation in PD; and modulation of monoamine disturbance in depression. Particularly, the xanthone derivatives, including α-mangostin and γ-mangostin, exhibit potent pharmacological actions. However, low oral bioavailability and poor brain penetration may limit their therapeutic applications. These challenges can be overcome in part by administering as a form of MP extract (MPE) or using specific carrier systems. MPE and α-mangostin are generally safe and well-tolerated in animals. Furthermore, mangosteen-based products are safe for humans. Therefore, MPE and its bioactive xanthones are promising candidates for the treatment of AD, PD, and depression. Further studies including clinical trials are essential to decipher their efficacy, and pharmacokinetic and safety profiles in these disorders.
Collapse
Affiliation(s)
| | - Jungsook Cho
- College of Pharmacy, Dongguk University-Seoul, Dongguk-ro 32, Ilsandong-gu, Goyang, Gyeonggi 10326, Korea;
| |
Collapse
|
24
|
Hao S, Li X, Han A, Yang Y, Luo X, Fang G, Wang H, Liu J, Wang S. Hydroxycinnamic Acid from Corncob and Its Structural Analogues Inhibit Aβ40 Fibrillation and Attenuate Aβ40-Induced Cytotoxicity. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2020; 68:8788-8796. [PMID: 32700906 DOI: 10.1021/acs.jafc.0c01841] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/15/2023]
Abstract
The aggregation of amyloid-β protein (Aβ) is deemed a vital pathological feature of Alzheimer's disease (AD). Hence, inhibiting Aβ aggregation is noticed as a major tactic for the prevention and therapy of AD. Hydroxycinnamic acid, as a natural phenolic compound, is widely present in plant foods and has several biological activities including anti-inflammation, antioxidation, and neuroprotective effects. Here, it was found that hydroxycinnamic acid and its structural analogues (3-hydroxycinnamic acid, 2-hydroxycinnamic acid, cinnamic acid, 3,4-dihydroxycinnamic acid, 2,4-dihydroxycinnamic acid, and 3,4,5-trihydroxycinnamic acid) could inhibit Aβ40 fibrillogenesis and reduce Aβ40-induced cytotoxicity in a dose-dependent manner. Among these small molecules investigated, 3,4,5-trihydroxycinnamic acid is considered to be the most effective inhibitor, which reduces the ThT fluorescence intensity to 30.79% and increases cell viability from 49.47 to 84.78% at 200 μM. Also, the results with Caenorhabditis elegans verified that these small molecules can ameliorate AD-like symptoms of worm paralysis. Moreover, molecular docking studies showed that these small molecules interact with the Aβ40 mainly via hydrogen bonding. These results suggest that hydroxycinnamic acid and its structural analogues could inhibit Aβ40 fibrillogenesis and the inhibition activity is enhanced with the increase of phenolic hydroxyl groups of inhibitors. These small molecules have huge potential to be developed into novel aggregation inhibitors in neurodegenerative disorders.
Collapse
Affiliation(s)
- Sijia Hao
- State Key Laboratory of Food Nutrition and Safety, College of Food Science and Engineering, Tianjin University of Science and Technology, Tianjin 300457, PR China
| | - Xia Li
- State Key Laboratory of Food Nutrition and Safety, College of Food Science and Engineering, Tianjin University of Science and Technology, Tianjin 300457, PR China
| | - Ailing Han
- State Key Laboratory of Food Nutrition and Safety, College of Food Science and Engineering, Tianjin University of Science and Technology, Tianjin 300457, PR China
| | - Yayu Yang
- State Key Laboratory of Food Nutrition and Safety, College of Food Science and Engineering, Tianjin University of Science and Technology, Tianjin 300457, PR China
| | - Xiaoyu Luo
- State Key Laboratory of Food Nutrition and Safety, College of Food Science and Engineering, Tianjin University of Science and Technology, Tianjin 300457, PR China
| | - Guozhen Fang
- State Key Laboratory of Food Nutrition and Safety, College of Food Science and Engineering, Tianjin University of Science and Technology, Tianjin 300457, PR China
| | - Hao Wang
- State Key Laboratory of Food Nutrition and Safety, College of Food Science and Engineering, Tianjin University of Science and Technology, Tianjin 300457, PR China
| | - Jifeng Liu
- State Key Laboratory of Food Nutrition and Safety, College of Food Science and Engineering, Tianjin University of Science and Technology, Tianjin 300457, PR China
| | - Shuo Wang
- State Key Laboratory of Food Nutrition and Safety, College of Food Science and Engineering, Tianjin University of Science and Technology, Tianjin 300457, PR China
- Research Center of Food Science and Human Health, School of Medicine, Nankai University, Tianjin 300071, PR China
| |
Collapse
|
25
|
Khaw KY, Kumar P, Yusof SR, Ramanathan S, Murugaiyah V. Probing simple structural modification of α-mangostin on its cholinesterase inhibition and cytotoxicity. Arch Pharm (Weinheim) 2020; 353:e2000156. [PMID: 32716578 DOI: 10.1002/ardp.202000156] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2020] [Revised: 06/22/2020] [Accepted: 07/11/2020] [Indexed: 11/08/2022]
Abstract
α-Mangostin has been reported to possess a broad range of pharmacological effects including potent cholinesterase inhibition, but the development of α-mangostin as a potential lead compound is impeded by its toxicity. The present study investigated the impact of simple structural modification of α-mangostin on its cholinesterase inhibitory activities and toxicity toward neuroblastoma and liver cancer cells. The dialkylated derivatives retained good acetylcholinesterase (AChE) inhibitory activities with IC50 values between 4.15 and 6.73 µM, but not butyrylcholinesterase (BChE) inhibitory activities, compared with α-mangostin, a dual inhibitor (IC50 : AChE, 2.48 µM; BChE, 5.87 µM). Dialkylation of α-mangostin produced AChE selective inhibitors that formed hydrophobic interactions at the active site of AChE. Interestingly, all four dialkylated derivatives of α-mangostin showed much lower cytotoxicity, being 6.4- to 9.0-fold and 3.8- to 5.5-fold less toxic than their parent compound on neuroblastoma and liver cancer cells, respectively. Likewise, their selectivity index was higher by 1.9- to 4.4-fold; in particular, A2 and A4 showed improved selectivity index compared with α-mangostin. Taken together, modification of the hydroxyl groups of α-mangostin at positions C-3 and C-6 greatly influenced its BChE inhibitory and cytotoxic but not its AChE inhibitory activities. These dialkylated derivatives are viable candidates for further structural modification and refinement, worthy in the search of new AChE inhibitors with higher safety margins.
Collapse
Affiliation(s)
- Kooi-Yeong Khaw
- Discipline of Pharmacology, School of Pharmaceutical Sciences, Penang, Malaysia.,School of Pharmacy, Monash University Malaysia, Bandar Sunway, Selangor Darul Ehsan, Malaysia
| | | | | | | | | |
Collapse
|
26
|
Huang Y, Sun L, Zhu S, Xu L, Liu S, Yuan C, Guo Y, Wang X. Neuroprotection Against Parkinson's Disease Through the Activation of Akt/GSK3β Signaling Pathway by Tovophyllin A. Front Neurosci 2020; 14:723. [PMID: 32742256 PMCID: PMC7364155 DOI: 10.3389/fnins.2020.00723] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2020] [Accepted: 06/17/2020] [Indexed: 12/18/2022] Open
Abstract
Parkinson’s disease (PD) is one of the most prevalent and life-threatening neurodegenerative disease and mainly characterized by lack of sufficient dopaminergic neurons in the substantia nigra pars compacta (SNc). Although current treatments help to alleviate clinical symptoms, effective therapies preventing neuronal loss remain scarce. Tovophyllin A (TA), one of the xanthones extracted from Garcinia mangostana L. (GM), has recently been reported to play a beneficial role in the therapy of neurodegenerative diseases. In our research, we explored whether TA has protective effects on dopaminergic neurons in PD models. We found that TA significantly reduced apoptotic cell death in primary cortical neurons treated with 1-methyl-4-phenyl pyridinium (MPP+) or paraquat (PQ) in the in vitro PD model. In an in vivo acute PD model induced by 1-methyl4-phenyl-1,2,3,5-tetrahydropyridine (MPTP) treatment, TA also attenuated the resulting behavioral dysfunctions and dopaminergic neuron loss. In the collected brain tissues, TA increased the phosphorylation of Akt and GSK-3β, which may be related to TA-mediated dopaminergic neuronal protective effects. In summary, our results illustrated that TA is a powerful cytoprotective agent for dopaminergic neurons in the MPTP-induced PD model, suggesting TA as a possible therapeutic candidate for PD.
Collapse
Affiliation(s)
- Yanjun Huang
- Key Laboratory of Mental Health of the Ministry of Education, Guangdong-Hong Kong-Macao Greater Bay Area Center for Brain Science and Brain-Inspired Intelligence, Guangdong Province Key Laboratory of Psychiatric Disorders, Department of Neurobiology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Lirong Sun
- Key Laboratory of Mental Health of the Ministry of Education, Guangdong-Hong Kong-Macao Greater Bay Area Center for Brain Science and Brain-Inspired Intelligence, Guangdong Province Key Laboratory of Psychiatric Disorders, Department of Neurobiology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Shuzhen Zhu
- Department of Neurology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Liu Xu
- Key Laboratory of Mental Health of the Ministry of Education, Guangdong-Hong Kong-Macao Greater Bay Area Center for Brain Science and Brain-Inspired Intelligence, Guangdong Province Key Laboratory of Psychiatric Disorders, Department of Neurobiology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Shuhu Liu
- Key Laboratory of Mental Health of the Ministry of Education, Guangdong-Hong Kong-Macao Greater Bay Area Center for Brain Science and Brain-Inspired Intelligence, Guangdong Province Key Laboratory of Psychiatric Disorders, Department of Neurobiology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Chunhua Yuan
- Key Laboratory of Mental Health of the Ministry of Education, Guangdong-Hong Kong-Macao Greater Bay Area Center for Brain Science and Brain-Inspired Intelligence, Guangdong Province Key Laboratory of Psychiatric Disorders, Department of Neurobiology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Yanwu Guo
- Department of Neurosurgery, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Xuemin Wang
- Key Laboratory of Mental Health of the Ministry of Education, Guangdong-Hong Kong-Macao Greater Bay Area Center for Brain Science and Brain-Inspired Intelligence, Guangdong Province Key Laboratory of Psychiatric Disorders, Department of Neurobiology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| |
Collapse
|
27
|
Guan H, Li J, Tan X, Luo S, Liu Y, Meng Y, Wu B, Zhou Y, Yang Y, Chen H, Hou L, Qiu Y, Li J. Natural Xanthone α-Mangostin Inhibits LPS-Induced Microglial Inflammatory Responses and Memory Impairment by Blocking the TAK1/NF-κB Signaling Pathway. Mol Nutr Food Res 2020; 64:e2000096. [PMID: 32506806 DOI: 10.1002/mnfr.202000096] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2020] [Revised: 05/14/2020] [Indexed: 12/18/2022]
Abstract
SCOPE The effect of α-mangostin (α-M), a polyphenolic xanthone isolated from mangostin, on lipopolysaccharide (LPS)-induced microglial activation and memory impairment is explored. The possible underlying mechanisms are also investigated. METHODS AND RESULTS Cytokine production and activation of transforming growth factor activated kinase-1 (TAK1) and nuclear factor-κB (NF-κB) are detected by enzyme-linked immunosorbent assay (ELISA) or Western blot. Microglial migration and phagocytosis are evaluated with scratch wound-healing assay and phagocytosis of fluorescent latex beads, respectively. Learning and memory abilities of mice are evaluated with the Morris water maze test. The nanomolar (100-500 nm) α-M suppresses LPS-induced pro-inflammatory cytokine production and inducible nitric oxide synthase (iNOS) expression in microglia. It also inhibits LPS-induced microglial migration and phagocytosis. α-M rescues LPS-caused, microglia-mediated neuronal dendritic damage. Moreover, α-M represses LPS-induced toll-like receptor 4 (TLR4) expression and activation of TAK1 and NF-κB. In a mouse neuroinflammation model, α-M (50 mg kg-1 day-1 ) shows obvious anti-neuroinflammatory, neuroprotective, and memory-improving effects in vivo. CONCLUSION α-M inhibits microglia-mediated neuroinflammation and prevents neurotoxicity and memory impairment from inflammatory damage. These results indicate that α-M has great potential to be used as a nutritional preventive strategy for neuroinflammation-related neurodegenerative disorders such as Alzheimer's disease.
Collapse
Affiliation(s)
- Huifeng Guan
- Department of Pharmacology and Chemical Biology, Shanghai Jiao Tong University School of Medicine, 280 South Chongqing Road, Shanghai, 200025, P. R. China
| | - Jiabing Li
- Department of Pharmacology and Chemical Biology, Shanghai Jiao Tong University School of Medicine, 280 South Chongqing Road, Shanghai, 200025, P. R. China
| | - Xiaofang Tan
- Department of Pharmacology and Chemical Biology, Shanghai Jiao Tong University School of Medicine, 280 South Chongqing Road, Shanghai, 200025, P. R. China
| | - Shenying Luo
- Department of Pharmacology and Chemical Biology, Shanghai Jiao Tong University School of Medicine, 280 South Chongqing Road, Shanghai, 200025, P. R. China
| | - Yangdan Liu
- Department of Pharmacology and Chemical Biology, Shanghai Jiao Tong University School of Medicine, 280 South Chongqing Road, Shanghai, 200025, P. R. China
| | - Yiwen Meng
- Department of Pharmacology and Chemical Biology, Shanghai Jiao Tong University School of Medicine, 280 South Chongqing Road, Shanghai, 200025, P. R. China
| | - Baichuan Wu
- Department of Pharmacology and Chemical Biology, Shanghai Jiao Tong University School of Medicine, 280 South Chongqing Road, Shanghai, 200025, P. R. China
| | - Yan Zhou
- Department of Pharmacology and Chemical Biology, Shanghai Jiao Tong University School of Medicine, 280 South Chongqing Road, Shanghai, 200025, P. R. China
| | - Yang Yang
- Department of Pharmacology and Chemical Biology, Shanghai Jiao Tong University School of Medicine, 280 South Chongqing Road, Shanghai, 200025, P. R. China
| | - Hongzhuan Chen
- Institute of Interdisciplinary Integrative Biomedical Research, Shanghai University of Traditional Chinese Medicine, 1200 Cailun Road, Shanghai, 201210, P. R. China
| | - Lina Hou
- Department of Pharmacology and Chemical Biology, Shanghai Jiao Tong University School of Medicine, 280 South Chongqing Road, Shanghai, 200025, P. R. China
| | - Yu Qiu
- Department of Pharmacology and Chemical Biology, Shanghai Jiao Tong University School of Medicine, 280 South Chongqing Road, Shanghai, 200025, P. R. China
| | - Juan Li
- Department of Pharmacology and Chemical Biology, Shanghai Jiao Tong University School of Medicine, 280 South Chongqing Road, Shanghai, 200025, P. R. China
| |
Collapse
|
28
|
Abouelela ME, Orabi MA, Abdelhamid RA, Abdelkader MS, Darwish FM, Hotsumi M, Konno H. Anti-Alzheimer's flavanolignans from Ceiba pentandra aerial parts. Fitoterapia 2020; 143:104541. [DOI: 10.1016/j.fitote.2020.104541] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2019] [Revised: 03/01/2020] [Accepted: 03/02/2020] [Indexed: 11/30/2022]
|
29
|
Chen Y, Bian Y, Wang JW, Gong TT, Ying YM, Ma LF, Shan WG, Xie XQ, Zhan ZJ. Effects of α-Mangostin Derivatives on the Alzheimer's Disease Model of Rats and Their Mechanism: A Combination of Experimental Study and Computational Systems Pharmacology Analysis. ACS OMEGA 2020; 5:9846-9863. [PMID: 32391472 PMCID: PMC7203693 DOI: 10.1021/acsomega.0c00057] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/05/2020] [Accepted: 04/16/2020] [Indexed: 06/09/2023]
Abstract
α-Mangostin (α-M) is a natural xanthone from the pericarp of fruit Garcinia mangostana and possesses versatile biological activities. α-M has a therapeutic potential to treat Alzheimer's disease (AD) because of its anti-inflammatory, antioxidative, and neuroprotective activities. However, the use of α-M for AD treatment is limited due to its cytotoxic activities and relatively low potency. Modifications of its chemical structure were needed to reduce its cytotoxicity and improve its therapeutic potential against AD. For this purpose, 16 α-M carbamate derivatives were synthesized. An animal model of AD was established, and the effects of AMG-1 on the spatial learning ability and memory ability were evaluated using behavioral tests. The effect on neuropathology was tested by histopathological evaluation, Nissl staining, and silver staining. Computational systems pharmacology analysis using the chemogenomics knowledgebase was applied for network studies. Compound-target, target-pathway, and target-disease networks were constructed, integrating both in silico analysis and reported experimental data. The results show that AMG-1 can demonstrate its therapeutic effects in a one-molecule, multiple-targets manner to remarkably ameliorate neurological changes and reverse behavioral deficits in AD model rats. The improved cognitive function and alleviated neuronal injury can be observed. The ability of AMG-1 to scavenge β-amyloid in the hippocampus was validated in AD model rats.
Collapse
Affiliation(s)
- Yan Chen
- College
of Pharmacology Sciences Zhejiang University of Technology, Hangzhou 310014, P. R. China
- Department of Pharmaceutical
Sciences and Computational Chemical
Genomics Screening Center, School of Pharmacy; NIH National Center of Excellence
for Computational Drug Abuse Research; Drug Discovery Institute; Departments of Computational
Biology and Structural Biology, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania 15261, United States
| | - Yuemin Bian
- Department of Pharmaceutical
Sciences and Computational Chemical
Genomics Screening Center, School of Pharmacy; NIH National Center of Excellence
for Computational Drug Abuse Research; Drug Discovery Institute; Departments of Computational
Biology and Structural Biology, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania 15261, United States
| | - Jian-Wei Wang
- College
of Pharmacology Sciences Zhejiang University of Technology, Hangzhou 310014, P. R. China
| | - Ting-Ting Gong
- College
of Pharmacology Sciences Zhejiang University of Technology, Hangzhou 310014, P. R. China
| | - You-Min Ying
- College
of Pharmacology Sciences Zhejiang University of Technology, Hangzhou 310014, P. R. China
| | - Lie-Feng Ma
- College
of Pharmacology Sciences Zhejiang University of Technology, Hangzhou 310014, P. R. China
| | - Wei-Guang Shan
- College
of Pharmacology Sciences Zhejiang University of Technology, Hangzhou 310014, P. R. China
| | - Xiang-Qun Xie
- Department of Pharmaceutical
Sciences and Computational Chemical
Genomics Screening Center, School of Pharmacy; NIH National Center of Excellence
for Computational Drug Abuse Research; Drug Discovery Institute; Departments of Computational
Biology and Structural Biology, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania 15261, United States
| | - Zha-Jun Zhan
- College
of Pharmacology Sciences Zhejiang University of Technology, Hangzhou 310014, P. R. China
| |
Collapse
|
30
|
Ghosh S, Lalani R, Patel V, Bhowmick S, Misra A. Surface engineered liposomal delivery of therapeutics across the blood brain barrier: recent advances, challenges and opportunities. Expert Opin Drug Deliv 2019; 16:1287-1311. [DOI: 10.1080/17425247.2019.1676721] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Affiliation(s)
- Saikat Ghosh
- Department of Pharmaceutics, Faculty of Pharmacy, Kalabhavan Campus, The Maharaja Sayajirao University of Baroda, Vadodara, India
- Formulation Development Department-Novel Drug Delivery Systems, Sun Pharmaceutical Industries Ltd, Vadodara, India
| | - Rohan Lalani
- Department of Pharmaceutics, Faculty of Pharmacy, Kalabhavan Campus, The Maharaja Sayajirao University of Baroda, Vadodara, India
- Formulation Development Department-Novel Drug Delivery Systems, Sun Pharmaceutical Industries Ltd, Vadodara, India
| | - Vivek Patel
- Department of Pharmaceutics, Faculty of Pharmacy, Kalabhavan Campus, The Maharaja Sayajirao University of Baroda, Vadodara, India
| | - Subhas Bhowmick
- Department of Pharmaceutics, Faculty of Pharmacy, Kalabhavan Campus, The Maharaja Sayajirao University of Baroda, Vadodara, India
- Formulation Development Department-Novel Drug Delivery Systems, Sun Pharmaceutical Industries Ltd, Vadodara, India
| | - Ambikanandan Misra
- Department of Pharmaceutics, Faculty of Pharmacy, Kalabhavan Campus, The Maharaja Sayajirao University of Baroda, Vadodara, India
| |
Collapse
|
31
|
Kuo YC, Rajesh R. Challenges in the treatment of Alzheimer’s disease: recent progress and treatment strategies of pharmaceuticals targeting notable pathological factors. Expert Rev Neurother 2019; 19:623-652. [DOI: 10.1080/14737175.2019.1621750] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Affiliation(s)
- Yung-Chih Kuo
- Department of Chemical Engineering, National Chung Cheng University, Chia-Yi, Taiwan, Republic of China
| | - Rajendiran Rajesh
- Department of Chemical Engineering, National Chung Cheng University, Chia-Yi, Taiwan, Republic of China
| |
Collapse
|
32
|
Ge Y, Xu X, Liang Q, Xu Y, Huang M. α-Mangostin suppresses NLRP3 inflammasome activation via promoting autophagy in LPS-stimulated murine macrophages and protects against CLP-induced sepsis in mice. Inflamm Res 2019; 68:471-479. [PMID: 30927050 DOI: 10.1007/s00011-019-01232-0] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2018] [Revised: 03/05/2019] [Accepted: 03/26/2019] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND The major mechanism of sepsis is immunosuppression caused by host response dysfunction. It has been found that α-Mangostin (α-M) is a potential candidate as a treatment for multiple inflammatory and immune disorders. To date, the role of α-M in host response during sepsis remains unexplored. METHODS AND RESULTS Herein, we examined the effect of α-M on macrophages-mediated host response in the presence of lipopolysaccharide (LPS), and the vital organ function, inflammatory response, and survival rate in septic mice. In murine peritoneal macrophages, α-M induced autophagy and then inhibited LPS-stimulated NLRP3 inflammasome activation, as well as interleukin-1β (IL-1β) production. Moreover, α-M improved phagocytosis and killing of macrophages, and increased M2 macrophages numbers after LPS stimulation. Furthermore, in vivo experiment suggested that α-M reduced serum levels of tumor necrosis factor-α (TNF-α), interferon-γ (IFN-γ), IL-1β, alanine transaminase (ALT), aspartate transaminase (AST), and creatinine (Cr), whilst increased that of interleukin-10 (IL-10) in caecal ligation and puncture (CLP) mice. CONCLUSION Taken together, these findings showed that α-M-mediated macrophages autophagy contributed to NLRP3 inflammasome inactivation and α-M exerted organ protection in septic mice.
Collapse
Affiliation(s)
- Yun Ge
- Department of General Intensive Care Unit, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, 310052, China
| | - Xin Xu
- Department of General Intensive Care Unit, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, 310052, China
| | - Qiqiang Liang
- Department of General Intensive Care Unit, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, 310052, China
| | - Yongshan Xu
- Department of General Intensive Care Unit, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, 310052, China
| | - Man Huang
- Department of General Intensive Care Unit, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, 310052, China.
| |
Collapse
|
33
|
Inhibition of Oxidative Neurotoxicity and Scopolamine-Induced Memory Impairment by γ-Mangostin: In Vitro and In Vivo Evidence. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2019; 2019:3640753. [PMID: 31019651 PMCID: PMC6451816 DOI: 10.1155/2019/3640753] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/04/2018] [Revised: 12/14/2018] [Accepted: 01/09/2019] [Indexed: 01/08/2023]
Abstract
Among a series of xanthones identified from mangosteen, the fruit of Garcinia mangostana L. (Guttifereae), α- and γ-mangostins are known to be major constituents exhibiting diverse biological activities. However, the effects of γ-mangostin on oxidative neurotoxicity and impaired memory are yet to be elucidated. In the present study, the protective effect of γ-mangostin on oxidative stress-induced neuronal cell death and its underlying action mechanism(s) were investigated and compared to that of α-mangostin using primary cultured rat cortical cells. In addition, the effect of orally administered γ-mangostin on scopolamine-induced memory impairment was evaluated in mice. We found that γ-mangostin exhibited prominent protection against H2O2- or xanthine/xanthine oxidase-induced oxidative neuronal death and inhibited reactive oxygen species (ROS) generation triggered by these oxidative insults. In contrast, α-mangostin had no effects on the oxidative neuronal damage or associated ROS production. We also found that γ-mangostin, not α-mangostin, significantly inhibited H2O2-induced DNA fragmentation and activation of caspases 3 and 9, demonstrating its antiapoptotic action. In addition, only γ-mangostin was found to effectively inhibit lipid peroxidation and DPPH radical formation, while both mangostins inhibited β-secretase activity. Furthermore, we observed that the oral administration of γ-mangostin at dosages of 10 and 30 mg/kg markedly improved scopolamine-induced memory impairment in mice. Collectively, these results provide both in vitro and in vivo evidences for the neuroprotective and memory enhancing effects of γ-mangostin. Multiple mechanisms underlying this neuroprotective action were suggested in this study. Based on our findings, γ-mangostin could serve as a potentially preferable candidate over α-mangostin in combatting oxidative stress-associated neurodegenerative diseases including Alzheimer's disease.
Collapse
|
34
|
Sun Q, Liu F, Sang J, Lin M, Ma J, Xiao X, Yan S, Naman CB, Wang N, He S, Yan X, Cui W, Liang H. 9-Methylfascaplysin Is a More Potent Aβ Aggregation Inhibitor than the Marine-Derived Alkaloid, Fascaplysin, and Produces Nanomolar Neuroprotective Effects in SH-SY5Y Cells. Mar Drugs 2019; 17:md17020121. [PMID: 30781608 PMCID: PMC6409607 DOI: 10.3390/md17020121] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2019] [Revised: 02/02/2019] [Accepted: 02/07/2019] [Indexed: 02/07/2023] Open
Abstract
β-Amyloid (Aβ) is regarded as an important pathogenic target for Alzheimer’s disease (AD), the most prevalent neurodegenerative disease. Aβ can assemble into oligomers and fibrils, and produce neurotoxicity. Therefore, Aβ aggregation inhibitors may have anti-AD therapeutic efficacies. It was found, here, that the marine-derived alkaloid, fascaplysin, inhibits Aβ fibrillization in vitro. Moreover, the new analogue, 9-methylfascaplysin, was designed and synthesized from 5-methyltryptamine. Interestingly, 9-methylfascaplysin is a more potent inhibitor of Aβ fibril formation than fascaplysin. Incubation of 9-methylfascaplysin with Aβ directly reduced Aβ oligomer formation. Molecular dynamics simulations revealed that 9-methylfascaplysin might interact with negatively charged residues of Aβ42 with polar binding energy. Hydrogen bonds and π–π interactions between the key amino acid residues of Aβ42 and 9-methylfascaplysin were also suggested. Most importantly, compared with the typical Aβ oligomer, Aβ modified by nanomolar 9-methylfascaplysin produced less neuronal toxicity in SH-SY5Y cells. 9-Methylfascaplysin appears to be one of the most potent marine-derived compounds that produces anti-Aβ neuroprotective effects. Given previous reports that fascaplysin inhibits acetylcholinesterase and induces P-glycoprotein, the current study results suggest that fascaplysin derivatives can be developed as novel anti-AD drugs that possibly act via inhibition of Aβ aggregation along with other target mechanisms.
Collapse
Affiliation(s)
- Qingmei Sun
- Ningbo Key Laboratory of Behavioral Neuroscience, Zhejiang Provincial Key Laboratory of Pathophysiology, School of Medicine, Ningbo University, Ningbo 315211, China.
- Li Dak Sum Yip Yio Chin Kenneth Li Marine Biopharmaceutical Research Center, College of Food and Pharmaceutical Sciences, Ningbo University, Ningbo 315211, China.
| | - Fufeng Liu
- Key Laboratory of Industrial Fermentation Microbiology of Education, State Key Laboratory of Food Nutrition and Safety, College of Biotechnology, Tianjin University of Science & Technology, Tianjin 300457, China.
| | - Jingcheng Sang
- Key Laboratory of Industrial Fermentation Microbiology of Education, State Key Laboratory of Food Nutrition and Safety, College of Biotechnology, Tianjin University of Science & Technology, Tianjin 300457, China.
| | - Miaoman Lin
- School of Materials Science and Chemical Engineering, Ningbo University, Ningbo 315211, China.
| | - Jiale Ma
- School of Materials Science and Chemical Engineering, Ningbo University, Ningbo 315211, China.
| | - Xiao Xiao
- Ningbo Key Laboratory of Behavioral Neuroscience, Zhejiang Provincial Key Laboratory of Pathophysiology, School of Medicine, Ningbo University, Ningbo 315211, China.
| | - Sicheng Yan
- Ningbo Key Laboratory of Behavioral Neuroscience, Zhejiang Provincial Key Laboratory of Pathophysiology, School of Medicine, Ningbo University, Ningbo 315211, China.
| | - C Benjamin Naman
- Li Dak Sum Yip Yio Chin Kenneth Li Marine Biopharmaceutical Research Center, College of Food and Pharmaceutical Sciences, Ningbo University, Ningbo 315211, China.
| | - Ning Wang
- Li Dak Sum Yip Yio Chin Kenneth Li Marine Biopharmaceutical Research Center, College of Food and Pharmaceutical Sciences, Ningbo University, Ningbo 315211, China.
| | - Shan He
- Li Dak Sum Yip Yio Chin Kenneth Li Marine Biopharmaceutical Research Center, College of Food and Pharmaceutical Sciences, Ningbo University, Ningbo 315211, China.
| | - Xiaojun Yan
- Li Dak Sum Yip Yio Chin Kenneth Li Marine Biopharmaceutical Research Center, College of Food and Pharmaceutical Sciences, Ningbo University, Ningbo 315211, China.
| | - Wei Cui
- Ningbo Key Laboratory of Behavioral Neuroscience, Zhejiang Provincial Key Laboratory of Pathophysiology, School of Medicine, Ningbo University, Ningbo 315211, China.
- Li Dak Sum Yip Yio Chin Kenneth Li Marine Biopharmaceutical Research Center, College of Food and Pharmaceutical Sciences, Ningbo University, Ningbo 315211, China.
- Key Laboratory of Industrial Fermentation Microbiology of Education, State Key Laboratory of Food Nutrition and Safety, College of Biotechnology, Tianjin University of Science & Technology, Tianjin 300457, China.
| | - Hongze Liang
- School of Materials Science and Chemical Engineering, Ningbo University, Ningbo 315211, China.
| |
Collapse
|
35
|
Sivaranjani M, Leskinen K, Aravindraja C, Saavalainen P, Pandian SK, Skurnik M, Ravi AV. Deciphering the Antibacterial Mode of Action of Alpha-Mangostin on Staphylococcus epidermidis RP62A Through an Integrated Transcriptomic and Proteomic Approach. Front Microbiol 2019; 10:150. [PMID: 30787919 PMCID: PMC6372523 DOI: 10.3389/fmicb.2019.00150] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2018] [Accepted: 01/21/2019] [Indexed: 11/25/2022] Open
Abstract
Background: Alpha-mangostin (α-MG) is a natural xanthone reported to exhibit rapid bactericidal activity against Gram-positive bacteria, and may therefore have potential clinical application in healthcare sectors. This study sought to identify the impact of α-MG on Staphylococcus epidermidis RP62A through integrated advanced omic technologies. Methods: S. epidermidis was challenged with sub-MIC (0.875 μg/ml) of α-MG at various time points and the differential expression pattern of genes/proteins were analyzed in the absence and presence of α-MG using RNA sequencing and LC-MS/MS experiments. Bioinformatic tools were used to categorize the biological processes, molecular functions and KEGG pathways of differentially expressed genes/proteins. qRT-PCR was employed to validate the results obtained from these analyses. Results: Transcriptomic and proteomic profiling of α-MG treated cells indicated that genes/proteins affected by α-MG treatment were associated with diverse cellular functions. The greatest reduction in expression was observed in transcription of genes conferring cytoplasmic membrane integrity (yidC2, secA and mscL), cell division (ftsY and divlB), teichoic acid biosynthesis (tagG and dltA), fatty-acid biosynthesis (accB, accC, fabD, fabH, fabI, and fabZ), biofilm formation (icaA) and DNA replication and repair machinery (polA, polC, dnaE, and uvrA). Those with increased expression were involved in oxidative (katA and sodA) and cellular stress response (clpB, clpC, groEL, and asp23). The qRT-PCR analysis substantiated the results obtained from transcriptomic and proteomic profiling studies. Conclusion: Combining transcriptomic and proteomic methods provided comprehensive information about the antibacterial mode of action of α-MG. The obtained results suggest that α-MG targets S. epidermidis through multifarious mechanisms, and especially prompts that loss of cytoplasmic membrane integrity leads to rapid onset of bactericidal activity.
Collapse
Affiliation(s)
| | - Katarzyna Leskinen
- Department of Bacteriology and Immunology, Medicum, Research Programs Unit, Immunobiology Research Program, University of Helsinki, Helsinki, Finland
| | | | - Päivi Saavalainen
- Department of Bacteriology and Immunology, Medicum, Research Programs Unit, Immunobiology Research Program, University of Helsinki, Helsinki, Finland
| | | | - Mikael Skurnik
- Department of Bacteriology and Immunology, Medicum, Research Programs Unit, Immunobiology Research Program, University of Helsinki, Helsinki, Finland
- Division of Clinical Microbiology, Helsinki University Hospital, HUSLAB, Helsinki, Finland
| | | |
Collapse
|
36
|
Chi XQ, Hou B, Yang L, Zi CT, Lv YF, Li JY, Ren FC, Yuan MY, Hu JM, Zhou J. Design, synthesis and cholinesterase inhibitory activity of α-mangostin derivatives. Nat Prod Res 2018; 34:1380-1388. [PMID: 30456989 DOI: 10.1080/14786419.2018.1510925] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Affiliation(s)
- Xiao-Qian Chi
- State Key Laboratory of Phytochemistry and Plant Resources in West China, and Yunnan Key Laboratory of Natural Medicinal Chemistry, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming, People’ s Republic of China
- University of Chinese Academy of Sciences, Beijing, People’s Republic of China
| | - Bo Hou
- State Key Laboratory of Phytochemistry and Plant Resources in West China, and Yunnan Key Laboratory of Natural Medicinal Chemistry, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming, People’ s Republic of China
- University of Chinese Academy of Sciences, Beijing, People’s Republic of China
| | - Liu Yang
- State Key Laboratory of Phytochemistry and Plant Resources in West China, and Yunnan Key Laboratory of Natural Medicinal Chemistry, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming, People’ s Republic of China
| | - Cheng-Ting Zi
- College of Science, Yunnan Agricultural University, Kunming, People’s Republic of China
| | - Yong-Feng Lv
- State Key Laboratory of Phytochemistry and Plant Resources in West China, and Yunnan Key Laboratory of Natural Medicinal Chemistry, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming, People’ s Republic of China
- University of Chinese Academy of Sciences, Beijing, People’s Republic of China
| | - Jin-Yu Li
- State Key Laboratory of Phytochemistry and Plant Resources in West China, and Yunnan Key Laboratory of Natural Medicinal Chemistry, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming, People’ s Republic of China
- University of Chinese Academy of Sciences, Beijing, People’s Republic of China
| | - Fu-Cai Ren
- State Key Laboratory of Phytochemistry and Plant Resources in West China, and Yunnan Key Laboratory of Natural Medicinal Chemistry, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming, People’ s Republic of China
- University of Chinese Academy of Sciences, Beijing, People’s Republic of China
| | - Ming-Yan Yuan
- State Key Laboratory of Phytochemistry and Plant Resources in West China, and Yunnan Key Laboratory of Natural Medicinal Chemistry, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming, People’ s Republic of China
| | - Jiang-Miao Hu
- State Key Laboratory of Phytochemistry and Plant Resources in West China, and Yunnan Key Laboratory of Natural Medicinal Chemistry, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming, People’ s Republic of China
| | - Jun Zhou
- State Key Laboratory of Phytochemistry and Plant Resources in West China, and Yunnan Key Laboratory of Natural Medicinal Chemistry, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming, People’ s Republic of China
| |
Collapse
|
37
|
Shrestha R, Lee YR. Base-Promoted Denitrogenative/Deoxygenative/Deformylative Benzannulation of N-Tosylhydrazones with 3-Formylchromones for Diverse and Polyfunctionalized Xanthones. Org Lett 2018; 20:7167-7171. [PMID: 30370769 DOI: 10.1021/acs.orglett.8b03106] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
A simple and efficient base-promoted denitrogenative/deoxygenative/deformylative benzannulation is developed for the construction of biologically interesting polyfunctionalized xanthones starting from N-tosylhydrazones and two molecules of 3-formylchromones. This unprecedented protocol proceeds via a cascade diazo formation/Michael addition/denitrogenation/[4 + 2] cycloaddition/elimination/ring opening. The synthesized xanthones possess potent UV-filter, fluorescent sensor, and antioxidant properties.
Collapse
Affiliation(s)
- Rajeev Shrestha
- School of Chemical Engineering , Yeungnam University , Gyeongsan 38541 , Republic of Korea
| | - Yong Rok Lee
- School of Chemical Engineering , Yeungnam University , Gyeongsan 38541 , Republic of Korea
| |
Collapse
|
38
|
Basavan D, Chalichem NSS, Kumar MKS. Phytoconstituents and their Possible Mechanistic Profile for Alzheimer's Disease - A Literature Review. Curr Drug Targets 2018; 20:263-291. [PMID: 30101703 DOI: 10.2174/1389450119666180813095637] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2018] [Revised: 07/24/2018] [Accepted: 08/08/2018] [Indexed: 11/22/2022]
Abstract
Memory is an associated part of life without which livelihood of a human being becomes miserable. As the global aged population is increasing tremendously, time has come to concentrate on tail end life stage diseases. Alzheimer's disease (AD) is one of such diseases whose origin is enigmatic, having an impact on later stage of life drastically due to irreparable damage of cognition, characterised by the presence of neurotoxic amyloid-beta (Aβ) plaques and hyper phosphorylated Tau protein as fibrillary tangles. Existing therapeutic regimen mainly focuses on symptomatic relief by targeting neurotransmitters that are secondary to AD pathology. Plant derived licensed drugs, Galantamine and Huperzine-A were studied extensively due to their AChE inhibitory action for mild to moderate cases of AD. Although many studies have proved the efficacy of AChEIs as a preferable symptom reliever, they cannot offer long term protection. The future generation drugs of AD is expected to alter various factors that underlie the disease course with a symptomatic benefit promise. As AD involves complex pathology, it is essential to consider several molecular divergent factors apart from the events that result in the production of toxic plaques and neurofibrillary tangles. Even though several herbals have shown neuroprotective actions, we have mentioned about the phytoconstituents that have been tested experimentally against different Alzheimer's pathology models. These phytoconstituents need to be considered by the researchers for further drug development process to make them viable clinically, which is currently a lacuna.
Collapse
Affiliation(s)
- Duraiswamy Basavan
- Department of Pharmacognosy and Phytopharmacy, JSS College of pharmacy (Constituent College of JSS Academy of Higher Education and Research, Mysuru), Ooty-643001, India
| | - Nehru S S Chalichem
- Department of Pharmacognosy and Phytopharmacy, JSS College of pharmacy (Constituent College of JSS Academy of Higher Education and Research, Mysuru), Ooty-643001, India
| | - Mohan K S Kumar
- TIFAC CORE Herbal drugs, Department of Pharmacognosy and Phytopharmacy, JSS College of Pharmacy (Constituent College of JSS Academy of Higher Education and Research, Mysuru), ooty-643001, India
| |
Collapse
|
39
|
Mostafavi N, Ebrahimi A. The role of chlorine substituents in lichexanthones properties: the ionic and halogen bond interactions. Theor Chem Acc 2018. [DOI: 10.1007/s00214-018-2294-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/28/2022]
|
40
|
Wang P, Zheng X, Guo Q, Yang P, Pang X, Qian K, Lu W, Zhang Q, Jiang X. Systemic delivery of BACE1 siRNA through neuron-targeted nanocomplexes for treatment of Alzheimer's disease. J Control Release 2018; 279:220-233. [DOI: 10.1016/j.jconrel.2018.04.034] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2017] [Revised: 03/22/2018] [Accepted: 04/17/2018] [Indexed: 10/17/2022]
|
41
|
Shal B, Ding W, Ali H, Kim YS, Khan S. Anti-neuroinflammatory Potential of Natural Products in Attenuation of Alzheimer's Disease. Front Pharmacol 2018; 9:548. [PMID: 29896105 PMCID: PMC5986949 DOI: 10.3389/fphar.2018.00548] [Citation(s) in RCA: 123] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2018] [Accepted: 05/08/2018] [Indexed: 12/22/2022] Open
Abstract
Alzheimer's disease (AD) is a chronic progressive neurodegenerative disorder associated with dementia and cognitive impairment most common in elderly population. Various pathophysiological mechanisms have been proposed by numerous researcher, although, exact mechanism is not yet elucidated. Several studies have been indicated that neuroinflammation associated with deposition of amyloid- beta (Aβ) in brain is a major hallmark toward the pathology of neurodegenerative diseases. So, there is a need to unravel the link of inflammatory process in neurodegeneration. Increased microglial activation, expression of cytokines, reactive oxygen species (ROS), and nuclear factor kappa B (NF-κB) participate in inflammatory process of AD. This review mainly concentrates on involvement of neuroinflammation and the molecular mechanisms adapted by various natural compounds, phytochemicals and herbal formulations in various signaling pathways involved in neuroprotection. Currently, pharmacologically active natural products, having anti-neuroinflammatory potential are being focused which makes them potential candidate to cure AD. A number of preclinical and clinical trials have been done on nutritional and botanical agents. Analysis of anti-inflammatory and neuroprotective phytochemicals such as terpenoids, phenolic derivatives, alkaloids, glycosides, and steroidal saponins displays therapeutic potential toward amelioration and prevention of devastating neurodegeneration observed in AD.
Collapse
Affiliation(s)
- Bushra Shal
- Department of Pharmacy, Faculty of Biological Sciences, Quaid-i-Azam University, Islamabad, Pakistan
| | - Wei Ding
- Department of Neurosurgery, Rizhao Hospital of Traditional Chinese Medicine, Rizhao, China
| | - Hussain Ali
- Department of Pharmacy, Faculty of Biological Sciences, Quaid-i-Azam University, Islamabad, Pakistan
| | - Yeong S Kim
- College of Pharmacy, Seoul National University, Seoul, South Korea
| | - Salman Khan
- Department of Pharmacy, Faculty of Biological Sciences, Quaid-i-Azam University, Islamabad, Pakistan
| |
Collapse
|
42
|
In Silico Studies Applied to Natural Products with Potential Activity Against Alzheimer’s Disease. NEUROMETHODS 2018. [DOI: 10.1007/978-1-4939-7404-7_18] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/11/2023]
|
43
|
Avila-Vazquez MF, Altamirano-Bustamante NF, Altamirano-Bustamante MM. Amyloid Biomarkers in Conformational Diseases at Face Value: A Systematic Review. Molecules 2017; 23:molecules23010079. [PMID: 29286329 PMCID: PMC6017595 DOI: 10.3390/molecules23010079] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2017] [Revised: 10/03/2017] [Accepted: 10/11/2017] [Indexed: 12/20/2022] Open
Abstract
Conformational diseases represent a new aspect of proteomic medicine where diagnostic and therapeutic paradigms are evolving. In this context, the early biomarkers for target cell failure (neurons, β-cells, etc.) represent a challenge to translational medicine and play a multidimensional role as biomarkers and potential therapeutic targets. This systematic review, which follows the PICO and Prisma methods, analyses this new-fangled multidimensionality, its strengths and limitations, and presents the future possibilities it opens up. The nuclear diagnosis methods are immunoassays: ELISA, immunodot, western blot, etc., while the therapeutic approach is focused on pharmaco- and molecular chaperones.
Collapse
Affiliation(s)
- Maria Fernanda Avila-Vazquez
- Unidad de Investigación en Enfermedades Metabólicas, Centro Médico Nacional Siglo XXI, IMSS, Mexico City 06720, Mexico.
- Health Department, Universidad Iberoamericana, Mexico City 01219, Mexico.
| | | | | |
Collapse
|
44
|
Ovalle-Magallanes B, Eugenio-Pérez D, Pedraza-Chaverri J. Medicinal properties of mangosteen (Garcinia mangostana L.): A comprehensive update. Food Chem Toxicol 2017; 109:102-122. [PMID: 28842267 DOI: 10.1016/j.fct.2017.08.021] [Citation(s) in RCA: 113] [Impact Index Per Article: 16.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2017] [Revised: 08/16/2017] [Accepted: 08/18/2017] [Indexed: 12/22/2022]
Abstract
Garcinia mangostana L. (Clusiaceae) is a tropical tree native to Southeast Asia known as mangosteen which fruits possess a distinctive and pleasant taste that has granted them the epithet of "queen of the fruits". The seeds and pericarps of the fruit have a long history of use in the traditional medicinal practices of the region, and beverages containing mangosteen pulp and pericarps are sold worldwide as nutritional supplements. The main phytochemicals present in the species are isoprenylated xanthones, a class of secondary metabolites with multiple reports of biological effects, such as antioxidant, pro-apoptotic, anti-proliferative, antinociceptive, anti-inflammatory, neuroprotective, hypoglycemic and anti-obesity. The diversity of actions displayed by mangosteen xanthones shows that these compounds target multiple signaling pathways involved in different pathologies, and place them as valuable sources for developing new drugs to treat chronic and degenerative diseases. This review article presents a comprehensive update of the toxicological findings on animal models, and the preclinical anticancer, analgesic, neuroprotective, antidiabetic and hypolipidemic effects of G. mangostana L. extracts and its main isolates. Pharmacokinetics, drug delivery systems and reports on dose-finding human trials are also examined.
Collapse
Affiliation(s)
- Berenice Ovalle-Magallanes
- Department of Biology, Faculty of Chemistry, National Autonomous University of Mexico (UNAM), Mexico City 04510, Mexico
| | - Dianelena Eugenio-Pérez
- Department of Biology, Faculty of Chemistry, National Autonomous University of Mexico (UNAM), Mexico City 04510, Mexico
| | - José Pedraza-Chaverri
- Department of Biology, Faculty of Chemistry, National Autonomous University of Mexico (UNAM), Mexico City 04510, Mexico.
| |
Collapse
|
45
|
Dual/multitargeted xanthone derivatives for Alzheimer's disease: where do we stand? Future Med Chem 2017; 9:1611-1630. [PMID: 28832188 DOI: 10.4155/fmc-2017-0086] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
To date, the current therapy for Alzheimer's disease (AD) based on acetylcholinesterase inhibitors is only symptomatic, being its efficacy limited. Hence, the recent research has been focused in the development of different pharmacological approaches. Here we discuss the potential of xanthone derivatives as new anti-Alzheimer agents. The interference of xanthone derivatives with acetylcholinesterase and other molecular targets and cellular mechanisms associated with AD have been recently systematically reported. Therefore, we report xanthones with anticholinesterase, monoamine oxidase and amyloid β aggregation inhibitory activities as well as antioxidant properties, emphasizing xanthone derivatives with dual/multitarget activity as potential agents to treat AD. We also propose the structural features for these activities that may guide the design of new, more effective xanthone derivatives. [Formula: see text].
Collapse
|
46
|
Effects of hydroxy-xanthones on dipalmitoylphosphatidylcholine lipid bilayers: A theoretical and experimental study. Chem Phys Lipids 2017; 206:1-8. [DOI: 10.1016/j.chemphyslip.2017.05.004] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2016] [Revised: 05/15/2017] [Accepted: 05/16/2017] [Indexed: 11/22/2022]
|
47
|
Hao XM, Li LD, Duan CL, Li YJ. Neuroprotective effect of α-mangostin on mitochondrial dysfunction and α-synuclein aggregation in rotenone-induced model of Parkinson's disease in differentiated SH-SY5Y cells. JOURNAL OF ASIAN NATURAL PRODUCTS RESEARCH 2017; 19:833-845. [PMID: 28696167 DOI: 10.1080/10286020.2017.1339349] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/10/2017] [Accepted: 06/01/2017] [Indexed: 06/07/2023]
Abstract
The study was designed to evaluate the protective effect of α-mangostin and explore its mechanism in an in vitro model of Parkinson's disease (PD) induced by rotenone. SH-SY5Y cells were treated with rotenone and α-mangostin for 24 h. α-Mangostin significantly and concentration-dependently inhibited rotenone-induced cytotoxicity. The rotenone-induced aggregation of α-synuclein and loss of TH were alleviated by α-mangostin. α-Mangostin treatment also reversed the rotenone-induced overproduction of reactive oxygen species, activation of caspases (-8 and -3) and mitochondrial dysfunction, reflected by decrease in mitochondrial membrane potential and cellular ATP levels. These findings suggest that α-mangostin has neuroprotective effects against PD-related neuronal injury.
Collapse
Affiliation(s)
- Xin-Mei Hao
- a Graduate School, Beijing University of Chinese Medicine , Beijing 100029 , China
- b Laboratory of Academician, Experimental Research Center , China Academy of Chinese Medical Sciences , Beijing 100700 , China
| | - Lian-Da Li
- b Laboratory of Academician, Experimental Research Center , China Academy of Chinese Medical Sciences , Beijing 100700 , China
| | - Chang-Ling Duan
- c Shanghai University of Traditional Chinese Medicine , Shanghai 201203 , China
| | - Yu-Juan Li
- b Laboratory of Academician, Experimental Research Center , China Academy of Chinese Medical Sciences , Beijing 100700 , China
| |
Collapse
|
48
|
Recent advancements in liposomes targeting strategies to cross blood-brain barrier (BBB) for the treatment of Alzheimer's disease. J Control Release 2017; 260:61-77. [PMID: 28549949 DOI: 10.1016/j.jconrel.2017.05.019] [Citation(s) in RCA: 211] [Impact Index Per Article: 30.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2017] [Revised: 05/12/2017] [Accepted: 05/13/2017] [Indexed: 12/20/2022]
Abstract
In this modern era, with the help of various advanced technologies, medical science has overcome most of the health-related issues successfully. Though, some diseases still remain unresolved due to various physiological barriers. One such condition is Alzheimer; a neurodegenerative disorder characterized by progressive memory impairment, behavioral abnormalities, mood swing and disturbed routine activities of the person suffering from. It is well known to all that the brain is entirely covered by a protective layer commonly known as blood brain barrier (BBB) which is responsible to maintain the homeostasis of brain by restricting the entry of toxic substances, drug molecules, various proteins and peptides, small hydrophilic molecules, large lipophilic substances and so many other peripheral components to protect the brain from any harmful stimuli. This functionally essential structure creates a major hurdle for delivery of any drug into the brain. Still, there are some provisions on BBB which facilitate the entry of useful substances in the brain via specific mechanisms like passive diffusion, receptor-mediated transcytosis, carrier-mediated transcytosis etc. Another important factor for drug transport is the selection of a suitable drug delivery systems like, liposome, which is a novel drug carrier system offering a potential approach to resolving this problem. Its unique phospholipid bilayer structure (similar to physiological membrane) had made it more compatible with the lipoidal layer of BBB and helps the drug to enter the brain. The present review work focused on various surface modifications with functional ligand (like lactoferrin, transferrin etc.) and carrier molecules (such as glutathione, glucose etc.) on the liposomal structure to enhance its brain targeting ability towards the successful treatment of Alzheimer disease.
Collapse
|
49
|
Zhao LX, Wang Y, Liu T, Wang YX, Chen HZ, Xu JR, Qiu Y. α-Mangostin decreases β-amyloid peptides production via modulation of amyloidogenic pathway. CNS Neurosci Ther 2017; 23:526-534. [PMID: 28429536 DOI: 10.1111/cns.12699] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2016] [Revised: 03/17/2017] [Accepted: 03/19/2017] [Indexed: 12/13/2022] Open
Abstract
AIMS β-amyloid (Aβ) aggregation and deposition play a central role in the pathogenic process of Alzheimer's disease (AD). α-Mangostin (α-M), a polyphenolic xanthone, have been shown to dissociate Aβ oligomers. In this study, we further investigated the effect of α-M on Aβ production and its molecular mechanism. METHODS The Aβ and soluble amyloid precursor protein α (sAPPα) in culture medium of cortical neurons were measured by ELISA. The activities of α-, β-, and γ-secretases were assayed, and the interaction between α-M and β- or γ-secretases was simulated by molecular docking. RESULTS α-M significantly decreased Aβ40 and Aβ42 production. α-M did not affect the expression of enzymes involved in nonamyloidogenic and amyloidogenic pathways, but significantly decreased the activities of β-secretase and likely γ-secretase with IC50 13.22 nmol·L-1 and 16.98 nmol·L-1 , respectively. Molecular docking demonstrated that α-M interacted with β-site amyloid precursor protein cleaving enzyme 1 and presenilin 1 to interfere with their active sites. CONCLUSIONS Our data demonstrate that α-M decreases Aβ production through inhibiting activities of β-secretase and likely γ-secretase in the amyloidogenic pathway. The current data together with previous study indicated that α-M could be a novel neuroprotective agent through intervention of multiple pathological processes of AD.
Collapse
Affiliation(s)
- Lan-Xue Zhao
- Department of Pharmacology, Institute of Medical Sciences, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yan Wang
- Department of Pharmacology, Institute of Medical Sciences, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Center for Cellular Immunotherapy, Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA, USA
| | - Ting Liu
- Department of Pharmacology, Institute of Medical Sciences, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Key Laboratory of Gynecologic Oncology, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yan-Xia Wang
- Department of Pharmacology, Institute of Medical Sciences, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Hong-Zhuan Chen
- Department of Pharmacology, Institute of Medical Sciences, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jian-Rong Xu
- Department of Pharmacology, Institute of Medical Sciences, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yu Qiu
- Department of Pharmacology, Institute of Medical Sciences, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
50
|
Abstract
Mangosteen (Garcinia mangostana Linn.) is a well-known tropical tree indigenous to Southeast Asia. Its fruit's pericarp abounds with a class of isoprenylated xanthones which are referred as mangostins. Numerous in vitro and in vivo studies have shown that mangostins and their derivatives possess diverse pharmacological activities, such as antibacterial, antifungal, antimalarial, anticarcinogenic, antiatherogenic activities as well as neuroprotective properties in Alzheimer's disease (AD). This review article provides a comprehensive review of the pharmacological activities of mangostins and their derivatives to reveal their promising utilities in the treatment of certain important diseases, mainly focusing on the discussions of the underlying molecular targets/pathways, modes of action, and relevant structure-activity relationships (SARs). Meanwhile, the pharmacokinetics (PK) profile and recent toxicological studies of mangostins are also described for further druggability exploration in the future.
Collapse
|