Mini review Topical Sections

Integrative medicinal approaches towards Alzheimer's disease model animals–A new hope

  • Alzheimer's disease (AD), a neurodegenerative disease (ND), has been plaguing healthcare and health policy agencies due to its ever-soaring impact globally. It is timely to develop appropriate measures to combat global AD burden. Due to structural and functional similarity, rodent models offer the best alternative to study AD pathogenesis and therapeutics. In this special issue, the present article reviews the state-of-the-art findings on AD model animals from the alternative and integrative medicinal point of view. Our integrative therapeutic agents include fungi (Ganoderma lucidum), turmeric (Curcuma longa), Indian black berry (black plum, jamun) (Syzygium cumini), and madecassoside (derived from Centella asiatica). Their bio-components, especially flavonoids, polyphenols, and tri-terpenoids, resist oxidative stress, amyloid beta fibrillation, and pro-inflammatory cytokine levels; improve anti-oxidative prowess; and improve learning-related memory and anxiety-like behavior in AD model animals. Most of the findings described here are outcomes of our own studies. However, our claims have been substantiated with evidence from some other studies in the relevant field. As a whole, integrative medicinal approaches towards AD pathogenesis using these natural products seem promising in AD protection.

    Citation: Shahdat Hossain, Mohammad Azizur Rahman, Shoriful Islam Joy, Anuradha Baraik, Shadman Nazib. Integrative medicinal approaches towards Alzheimer's disease model animals–A new hope[J]. AIMS Molecular Science, 2024, 11(3): 221-230. doi: 10.3934/molsci.2024013

    Related Papers:

    [1] Jean Emmanuel Mbosso Teinkela, Philippe Belle Ebanda Kedi, Jean Baptiste Hzounda Fokou, Michelle Isaacs, Lisette Pulchérie Yoyo Ngando, Gaelle Wea Tchepnou, Hassan Oumarou, Xavier Siwe Noundou . In vitro anti-trypanosomal activity of crude extract and fractions of Trichoscypha acuminata stem bark, Spathodea campanulata flowers, and Ficus elastica lianas on Trypanosoma brucei brucei. AIMS Molecular Science, 2024, 11(1): 63-71. doi: 10.3934/molsci.2024005
    [2] Irene Mwongeli Waita, Atunga Nyachieo, Daniel Chai, Samson Muuo, Naomi Maina, Daniel Kariuki, Cleophas M. Kyama . Differential expression and functional analysis of micro RNAs in Papio anubis induced with endometriosis for early detection of the disease. AIMS Molecular Science, 2020, 7(4): 305-327. doi: 10.3934/molsci.2020015
    [3] Sarmishta Mukhopadhyay, Sayak Ganguli, Santanu Chakrabarti . Shigella pathogenesis: molecular and computational insights. AIMS Molecular Science, 2020, 7(2): 99-121. doi: 10.3934/molsci.2020007
    [4] Seyedeh Fahimeh Razavi, Leila Zarandi Miandoab, Elaheh Zadeh Hosseingholi, Nader Chaparzadeh . Radical scavenging capacity of RuBisCO bioactive peptides derived from Dunaliella salina and Spirulina platensis: An in silico and in vitro study. AIMS Molecular Science, 2025, 12(1): 49-66. doi: 10.3934/molsci.2025004
    [5] Praveen Kumar Neela, Rajeshwari B.V, Mahamad Irfanulla Khan, Shahistha Parveen Dasnadi, Gosla Srinivas Reddy, Akhter Husain, Vasavi Mohan . Genetic influences on non-syndromic cleft lip palate: The impact of BMP4, RUNX2, PAX7, and TGFB3 allelic variations. AIMS Molecular Science, 2024, 11(4): 322-329. doi: 10.3934/molsci.2024019
    [6] Ioannis Tsakmakidis, Anastasia Parisi, Dimitrios K. Papadopoulos, Maria V. Alvanou, Konstantinos V. Papageorgiou, Evanthia Petridou, Ioannis A. Giantsis . Stray dogs as carriers of E. coli resistant strains for the retracted and re-emerged antibiotic colistin, based on the mcr-1 gene presence. AIMS Molecular Science, 2024, 11(4): 367-378. doi: 10.3934/molsci.2024022
    [7] Jin Hwan Do . Genome-wide transcriptional comparison of MPP+ treated human neuroblastoma cells with the state space model. AIMS Molecular Science, 2015, 2(4): 440-460. doi: 10.3934/molsci.2015.4.440
    [8] Yutaka Takihara, Ryuji Otani, Takuro Ishii, Shunsuke Takaoka, Yuki Nakano, Kaori Inoue, Steven Larsen, Yoko Ogino, Masashi Asai, Sei-ichi Tanuma, Fumiaki Uchiumi . Characterization of the human IDH1 gene promoter. AIMS Molecular Science, 2023, 10(3): 186-204. doi: 10.3934/molsci.2023013
    [9] Anna Kapambwe Bwalya, Robinson Mugasiali Irekwa, Amos Mbugua, Matthew Mutinda Munyao, Peter Kipkemboi Rotich, Tonny Teya Nyandwaro, Caroline Wangui Njoroge, Anne Wanjiru Mwangi, Joanne Jepkemei Yego, Shahiid Kiyaga, Samson Muuo Nzou . Investigation of single nucleotide polymorphisms in MRPA and AQP-1 genes of Leishmania donovani as resistance markers in visceral leishmaniasis in Kenya. AIMS Molecular Science, 2021, 8(2): 149-160. doi: 10.3934/molsci.2021011
    [10] Patrizia A d'Alessio, Marie C Béné, Chantal Menut . d-Limonene challenging anti-inflammatory strategies. AIMS Molecular Science, 2022, 9(2): 46-65. doi: 10.3934/molsci.2022003
  • Alzheimer's disease (AD), a neurodegenerative disease (ND), has been plaguing healthcare and health policy agencies due to its ever-soaring impact globally. It is timely to develop appropriate measures to combat global AD burden. Due to structural and functional similarity, rodent models offer the best alternative to study AD pathogenesis and therapeutics. In this special issue, the present article reviews the state-of-the-art findings on AD model animals from the alternative and integrative medicinal point of view. Our integrative therapeutic agents include fungi (Ganoderma lucidum), turmeric (Curcuma longa), Indian black berry (black plum, jamun) (Syzygium cumini), and madecassoside (derived from Centella asiatica). Their bio-components, especially flavonoids, polyphenols, and tri-terpenoids, resist oxidative stress, amyloid beta fibrillation, and pro-inflammatory cytokine levels; improve anti-oxidative prowess; and improve learning-related memory and anxiety-like behavior in AD model animals. Most of the findings described here are outcomes of our own studies. However, our claims have been substantiated with evidence from some other studies in the relevant field. As a whole, integrative medicinal approaches towards AD pathogenesis using these natural products seem promising in AD protection.



    Neurodegenerative diseases (NDs) are as complex as the entire nervous system in terms of their pathogenesis and manifestation. Complications of NDs include abnormalities in speech, movement, cognition, learning, memory, and personality, among others. Interestingly, some of the NDs share common or almost common pathomechanisms, such as late age onset, prolonged incubation period, mutation, neurodegeneration, neuroinflammation, oxidative damage, impaired repair processes, and protein misfolding and aggregation. NDs affect both the structure and function of the neurons and nervous system [1]. Therefore, the preparation of disease model organisms is important to understand the pathogenesis and progression and to resist the disease [2]. In this regard, the generation of cellular and animal models of various NDs has driven numerous research and development activities worldwide [3]. However, it has not been possible to generate a model suitable for all NDs, and even the old ND models appear to be inadequate to answer the emerging problems that challenge the search for a novel model development incorporating cutting-edge scientific knowledge. Animal models are typically created through structural, genetic, and functional manipulation and evaluated based on the after-effects of NDs [1][3]. Structural similarities between the brains of humans and rodents have facilitated these processes. Both transgenic and non-transgenic animal models have shown promising results in mimicking and alleviating ND symptoms [1][3]. In this context, rat and mouse models are predominantly used worldwide.

    We have explored neurodegenerative diseases, considering Alzheimer's disease (AD) as our prime concern, and utilized AD rat models following the method of Nabeshima and Nitta (1994) [4]. Our goal was to search for an AD ameliorating agent that is natural and food-based so that it can be utilizable to all and used as an alternative or integrative medicine as well as a dietary supplement. In this series, we have studied the effects of extracts from mushrooms (Ganoderma lucidum, Reishi mushroom), herbs (Centella asiatica itself and its bio-component madecassoside), shrubs (Curcuma longa), and fruit seeds (Syzygium cumini) on AD model rats for a few years. Our investigations have entailed biochemical and memory and learning related behavioral studies of the AD model organisms along with histopathological scrutinization to substantiate our claims that would aid the ever-increasing number of AD patients, their caregivers, researchers, health professionals, and policymakers worldwide.

    With regard to the pathogenesis of Alzheimer's disease, this mini-review briefly discusses how natural components of traditional plant/mushroom extracts can be targeted to alleviate the symptoms and pathological features, as well as their inhibitory effects on various cellular pathways that cause them to provide protection against various symptoms, including learning-related memory impairment. One of the goals of this review is to shed light on research in this regard.

    Highlights

    • Ganoderma lucidum as a tool against Alzheimer's disease (AD);

    • Effects of Curcuma longa in AD;

    • The potential of Syzygium cumini seed powder sold by street vendors in third world countries against AD;

    • Madecassoside, purified from Centella asiatica, has anti-AD effects.

    Alzheimer's disease (AD) is a progressive neurodegenerative disease characterized by cognitive decline and memory impairment [5],[6]. The symptoms usually manifest gradually. Early signs often include forgetfulness, difficulties in solving problems, and challenges in performing familiar tasks [5],[6]. As the disease progresses, those affected may experience speech problems, disorientation in time and space, and changes in mood and behavior [5],[6]. AD is the most common cause of dementia and affects millions of people worldwide [7]. Although its prevalence increases with age, it is not a normal part of aging. The exact cause of Alzheimer's disease remains unclear, but it is due to a complex interplay of genetic, environmental, and lifestyle factors. Pathologically, AD is associated with the accumulation of beta-amyloid plaques and tau tangles in the brain through fibrillation of amyloid beta peptide, leading to neuronal damage and impaired communication between brain cells [6] (Figure 1). The progressive nature of the disease poses significant challenges for affected individuals and their families and highlights the need for further research to better understand its mechanisms and develop effective interventions [7].

    Figure 1.  A The processes of amyloid fibrillation, in which a soluble α-helix/random-coil amyloid beta (Abeta/Aβ/Ab) is converted into an insoluble beta-sheet monomer. These beta sheets subsequently give rise to dimers, trimers, and tetramers, resulting in oligomers that eventually form mature fibers upon further aggregation. The oligomers are the most toxic species in the pathogenesis of Alzheimer's disease (AD). B Treatment of amyloid beta (Abeta/Aβ/Ab) with neuronal culture results in degenerative changes in neuronal morphology due to the oxidative and pro-inflammatory effects induced by amyloid oligomers. Similar effects are also observed in the brains of rodents after infusion into the cerebral ventricles of AD model rats/mice. Autopsy samples of the brains of AD patients also show these characteristic features of AD pathology. It is noteworthy that G. lucidum, C. longa, S. cumini, and C. asiatica extract alleviate the symptoms of AD.

    Ganoderma lucidum, commonly known as Reishi mushroom, has become a topic of interest in relation to Alzheimer's research. Scientific research into the possible effects of Ganoderma lucidum on Alzheimer's disease has suggested that this medicinal mushroom may have neuroprotective and AD ameliorating properties [8][14]. G. lucidum polysaccharide-1 (GLP-1) has been reported to modulate cognitive impairment by increasing the expression of interleukin-10 (IL-10) and transforming growth factor-β1 (TGF-β1) in chronic cerebral hypoperfusion (CCH) model mice [15]. GLP promoted neurogenesis and self-renewal of neural progenitor cells (NPC) and therefore may be involved in the recovery of cognitive decline associated with AD [16]. A modulatory effect of GLP on amyloid beta- and lipopolysaccharide-induced neuro-inflammation has been documented for microglia [17]. GLP stimulates the expression of anti-inflammatory cytokines (TGF β) and down-regulates the expression of pro-inflammatory AD-related cytokines (IL-1β, IL-6, inducible nitric oxide synthase) [17]. Furthermore, GLP could alter the morphology, migration, and phagocytic behavior of microglia in amyloid-beta- and lipopolysaccharide-induced neuroinflammation [17]. Dysregulation of the immune pathway is a recently recognized problem in AD pathogenesis [18]. The progression of AD has been linked to the differential expression of T-helper 17 (Th17) and T-regulatory (Treg) cells in the brain. Ganoderic acid A (GAA), a tri-terpene derived from G. lucidum, was found to restore the disrupted Th17/Treg axis in the brain [18]. GAA-driven anti-inflammatory and neuroprotective effects were achieved using the JAK/STAT signaling cascade. Thus, G. lucidum could restore the mitochondrial dysfunction associated with AD and revamp mitochondrial oxidative phosphorylation [18]. Methylation of CpG DNA islands is an important aspect in old age-related NDs, particularly in AD [19]. Alcoholic extract of G. lucidum could reduce the rate of DNA methylation, decrease the expression of neuronal amyloid beta-42 protein and apoptosis, and modulate brain atrophy [19]. The active ingredients identified were ganoderic acid (Mk, C6) and lucidone [19]. Ganoderic acid A and B were identified as the ligands of microtubule affinity regulation kinase 4 (MARK4), which mediates microtubule assembly [20]. By maintaining microtubule structure and cell cycle, ganoderic acids could alter the neuronal cytoskeleton of AD patients [20].

    Age-related physiological changes are among the most discernible hallmarks of AD [21]. Ganoderic acid A could alleviate brain and physiological decline [21]. Ganoderic acid A is also associated with delaying the aging process by modulating sphingolipid metabolism and increasing autophagy and telomere length [21]. Aqueous extract of G. lucidum together with other mushrooms could reduce the generation of reactive oxygen species (ROS), stimulate an anti-oxidative state, reduce apoptosis, and boost the survival rate of neuronal cell lines [22]. By reducing Aβ-induced neurotoxicity, this mushroom could improve memory and learning related behavioral changes in AD model mice [22]. In the Morris water maze study, improved memory was observed in the G. lucidum- treated mice [23]. G. lucidum had been praised for its inhibitory effect on acetylcholine esterase, restoring brain acetylcholine levels, which decrease in AD brains [23]. In addition to maintaining memory and redox homeostasis, G. lucidum exhibited strong neuroplasticity properties [24]. Mushrooms could increase the production of neurotransmitters, resist AD pathogenesis, and improve memory and learning-related behavioral performances [25]. Apart from AD-related memory loss, amelioration of morphine-induced memory loss and relief from morphine addiction could be achieved by taking G. lucidum aqueous extract [26]. The bioactive compounds contained in G. lucidum, through their anti-oxidative and neuroprotective effects and cognitive enhancement, provide a potential avenue for AD therapeutic interventions. Although the results are promising, further comprehensive studies are required to determine the specific mechanisms involved and to determine the optimal dosage and treatment protocols for incorporating G. lucidum into the AD treatment strategy.

    Curcuma longa, commonly known as turmeric, is the subject of growing interest in Alzheimer's disease research [27]. At the center of this attention is curcumin, a bioactive compound abundant in turmeric. Numerous studies have examined the possible therapeutic effects of curcumin in Alzheimer's disease and demonstrated its diverse properties [28]. With its well-documented anti-inflammatory and antioxidant properties, curcumin shows promise in alleviating the neuro-inflammation and oxidative stress associated with Alzheimer's disease [29]. The ability of curcumin to penetrate the blood-brain barrier further highlights its potential as a neuroprotective agent [30]. In addition, research suggests that curcumin may play a role in modulating the aggregation of beta-amyloid plaques, an important pathological feature of Alzheimer's disease pathology [31]. Beta site amyloid precursor protein cleaving enzyme 1 (BACE-1) is a regulatory enzyme and the first enzyme in the Aβ synthetic pathway [31]. BACE-1 is also a target for AD drugs. The BACE-1 inhibitory effect of curcuminoids appears to be promising against the progression of AD [31]. Moreover, lifestyle modification of the AD model Drosophila melanogaster opens new perspectives for the therapeutic use of curcuminoids and related compounds in AD [31]. The anti-oxidative, anti-neuroinflammatory, and anti-hypercholesterolemic effects of curcumin conferred by its polyphenol content might also be implicated in its AD-ameliorating effects [32]. Recently, the inhibitory effects of curcumin on hippocampal Aβ and neurofibrillary tau deposits, acetylcholine esterase, caspase-3, IL-1β, and TNFα, along with reduced ROS generation and an increase in the anti-oxidative enzyme glutathione synthase (GSH), have been reported in AD model rats [33]. In parallel, AD model rats treated with curcumin had improved memory and learning related behavioral performance [33]. So far, different bio-components of C. longa providing AD amelioration are, inter alia, curcumin, calebin, demethoxycurcumin, and curcuminoids [34].

    Although the results are encouraging, it is important to recognize the complexity of Alzheimer's disease and the need for larger clinical trials to determine the effectiveness of C. longa in real-world applications. Research into the effects of turmeric on Alzheimer's represents an exciting avenue to advance our understanding and potentially develop new therapeutic strategies for this debilitating neurodegenerative disease.

    Syzygium cumini, commonly known as jamun or Indian blackberry, has recently drawn interest from researchers studying its possible effects on Alzheimer's disease. While scientific research on the effect of Syzygium cumini on Alzheimer's disease is still in its early stages, initial studies suggest that the fruit possesses bioactive compounds with antioxidant and anti-inflammatory properties [35]. Our study suggested that S. cumini seed extract could reduce the oligomerization of Aβ and decrease the levels of Aβ and TNFα in the hippocampus [36]. At the same time, it improved hippocampal levels of brain derived neurotrophic factor (BDNF), postsynaptic-density protein 95 (PSD-95), synaptosomal-associated protein (SNAP-25), and tyrosine-kinase B (TrkB). These modulatory effects resulted in improved cognitive and memory related behavioral performance of AD model rats in an 8-armed radial maze study [36]. In a relevant study, Malik et al. [37] showed that S. cumini seed extract led to improved memory and learning related behavior as well as biochemical attainments in AD model mice through reduced acetylcholine esterase activity, reduced oxidative stress, and heightened antioxidant activity [37]. The active bio-components conferring these AD amendment attributes were identified as flavonoids and phenol derivatives such as β-farnesene, caryophyllene, humulene, and phytol [37]. Additionally, the neuroprotective effects of S. cumini include an acetylcholine esterase inhibitory effect along with attenuation of neuro-inflammation [38][40]. The β-secretase inhibitory effect of S. cumini has also been associated with its AD ameliorative potential [41].

    However, it is important to note that existing evidence is limited, and further rigorous studies are essential to substantiate these findings and elucidate the specific mechanisms involved. The study of S. cumini in the context of Alzheimer's disease represents an interesting avenue for future research and holds the potential for new insights into natural compounds that may contribute to the development of therapeutic strategies for this complex and challenging neurological disease.

    Madecassoside, derived from Centella asiatica, could attenuate AD pathogenesis by decreasing hippocampal deposition of Aβ1-42, oxidative stress, proinflammatory cytokine TNFα, and cathepsin D [42]. Madecassoside could reduce oligomerization of Aβ fibrils, an important step in AD pathogenesis [42]. Madecassoside could improve the diminished spatial memory of AD model rats. Our findings were further supported by molecular docking studies where madecassoside showed strong binding affinity with Aβ1-42 [43]. In addition to our findings, the AD-alleviating effect of madecassoside has been attributed to its harmonizing effect on circadian rhythms and sleep patterns, elimination of anxiety, and improvement of cognitive performance, particularly memory and learning-related behavioral modulation [44].

    Due to its complex and multifaceted nature, Alzheimer's disease remains incurable. The underlying causes of the disease involve complex interactions of genetic, environmental, and lifestyle factors, making it difficult to determine a single target for a cure. By the time AD symptoms appear, significant neurological damage has already occurred, complicating further possible treatment strategies. Protecting against AD requires a comprehensive approach that focuses on preventive measures and lifestyle changes. Maintaining a healthy and active lifestyle, including regular exercise, a balanced diet rich in antioxidants, and adequate sleep, is associated with a lower risk of developing AD. Cognitive stimulation through activities such as reading, puzzles, and social engagement can also contribute to brain health. When exploring alternatives to Alzheimer's treatment, research is increasingly focusing on personalized medicine and precision therapies. Tailored interventions based on a person's genetic makeup and specific biomarkers can enable more targeted and effective treatments. Innovative approaches such as immunotherapies and gene therapies are also being investigated. Given the lack of a definitive cure, emphasis on early detection and intervention is crucial. Regular cognitive assessments, advanced imaging techniques, and biomarker identification can help detect Alzheimer's disease in its early stages, when interventions may be more effective.

    In summary, a holistic approach that combines lifestyle changes, early detection strategies, and innovative therapeutic options is needed to address the challenges of Alzheimer's disease. While a complete cure may remain elusive, ongoing research and a multidimensional approach provide hope for improved outcomes and quality of life for individuals affected by Alzheimer's disease and their families.



    Author contributions



    Conceptualization, SH; Writing original draft-manuscript, SH; Writing, review, editing, MAR; Figure preparation, SN; Reading and editing, SIJ, AB, and SN.
    All authors have read and agreed to the published version of the manuscript.

    Conflict of interest



    The authors declare no conflict of interest.

    [1] Gitler AD, Dhillon P, Shorter J (2017) Neurodegenerative disease: Models, mechanisms, and a new hope. Dis Model Mech 10: 499-502. https://doi.org/10.1242/dmm.030205
    [2] Dhapola R, Kumari S, Sharma P, et al. (2023) Insight into the emerging and common experimental in-vivo models of Alzheimer's disease. Lab Anim Res 39: 33. https://doi.org/10.1186/s42826-023-00184-1
    [3] Dawson TM, Golde TE, Tourenne C (2018) Animal models of neurodegenerative diseases. Nat Neurosci 21: 1370-1379. https://doi.org/10.1038/s41593-018-0236-8
    [4] Nabeshima T, Nitta A (1994) Memory impairment and neuronal dysfunction induced by beta-amyloid protein in rats. Tohoku J Exp Med 174: 241-249. https://doi.org/10.1620/tjem.174.241
    [5] Lyketsos CG, Carrillo MC, Ryan JM, et al. (2011) Neuropsychiatric symptoms in Alzheimer's disease. Alzheimers Dement 7: 532-539. https://doi.org/10.1016/j.jalz.2011.05.2410
    [6] Sadigh-Eteghad S, Sabermarouf B, Majdi A, et al. (2015) Amyloid-beta: A crucial factor in Alzheimer's disease. Med Princ Pract 24: 1-10. https://doi.org/10.1159/000369101
    [7] Alzheimer's association.Alzheimer's association report: 2019 Alzheimer's disease facts and figures. Alzheimer's Dement (2019) 15: 321-387. https://doi.org/10.1016/j.jalz.2019.01.010
    [8] Rahman MA, Alpona AK, Biswas D, et al. (2023) Neuronal malady in epileptic brain–Amendment with mushrooms. Arch Neurol Neurosci 14: 2023. http://doi.org/10.33552/ANN.2023.14.000847
    [9] Rahman MA, Hossain S, Abdullah N, et al. (2020) Ganoderma lucidum modulates neuronal distorted cytoskeletal proteomics and protein-protein interaction in Alzheimer's disease model animals. Arch Neurol Neurosci 9: 2020. http://doi.org/10.33552/ANN.2020.09.000713
    [10] Sharma P., Tulsawani R. (2020) Ganoderma lucidum aqueous extract prevents hypobaric hypoxia induced memory deficit by modulating neurotransmission, neuroplasticity and maintaining redox homeostasis. Sci Rep 10: 8944. https://doi.org/10.1038/s41598-020-65812-5
    [11] Rahman MA, Hossain S, Abdullah N, et al. (2020) Validation of Ganoderma lucidum against hypercholesterolemia and Alzheimer's disease. Eur J Biol Res 10: 314-325. http://doi.org/10.5281/zenodo.4009588
    [12] Rahman MA, Hossain S, Abdullah N, et al. (2020) Ganoderma lucidum ameliorates spatial memory and memory-related protein markers in hypercholesterolemic and Alzheimer's disease model rats. Arch Neurol Neurol Disord 3: 117-129.
    [13] Rahman MA, Hossain S, Abdullah N, et al. (2020) Lingzhi or Reishi medicinal mushroom, Ganoderma lucidum (Agaricomycetes) ameliorates non-spatial learning and memory deficits in rats with hypercholesterolemia and Alzheimer's disease. Int J Med Mushroom 22: 93-103. http://doi.org/10.1615/IntJMedMushrooms.2020033383
    [14] Rahman MA, Hossain S, Abdullah N, et al. (2019) Brain proteomics links oxidative stress with metabolic and cellular stress response proteins in behavioural alteration of Alzheimer's disease model rats. AIMS Neurosci 6: 299-315. http://doi.org/10.3934/Neuroscience.2019.4.299
    [15] Zhang Y, Song S, Li H, et al. (2022) Polysaccharide from Ganoderma lucidum alleviates cognitive impairment in a mouse model of chronic cerebral hypoperfusion by regulating CD4+ CD25+ Foxp3+ regulatory T cells. Food Funct 13: 1941-1952. http://doi.org/10.1039/d1fo03698j
    [16] Huang S, Mao J, Ding K, et al. (2017) Polysaccharides from Ganoderma lucidum promote cognitive function and neural progenitor proliferation in mouse model of Alzheimer's disease. Stem Cell Reports 8: 84-94. http://doi.org/10.1016/j.stemcr.2016.12.007
    [17] Cai Q, Li Y, Pei G (2017) Polysaccharides from Ganoderma lucidum attenuate microglia-mediated neuroinflammation and modulate microglial phagocytosis and behavioural response. J Neuroinflammation 14: 63. http://doi.org/10.1186/s12974-017-0839-0
    [18] Zhang Y, Wang X, Yang X, et al. (2021) Ganoderic acid A to alleviate neuroinflammation of Alzheimer's disease in mice by regulating the imbalance of the Th17/Tregs axis. J Agric Food Chem 69: 14204-14214. http://doi.org/10.1021/acs.jafc.1c06304
    [19] Lai G, Guo Y, Chen D, et al. (2019) Alcohol extracts from Ganoderma lucidum delay the progress of Alzheimer's Disease by regulating DNA methylation in rodents. Front Pharmacol 10: 272. http://doi.org/10.3389/fphar.2019.00272
    [20] Ahmad F, Singh G, Soni H, et al. (2022) Identification of potential neuroprotective compound from Ganoderma lucidum extract targeting microtubule affinity regulation kinase 4 involved in Alzheimer's disease through molecular dynamics simulation and MMGBSA. Aging Med (Milton) 6: 144-154. http://doi.org/10.1002/agm2.12232
    [21] Zeng M, Qi L, Guo Y, et al. (2021) Long-term administration of triterpenoids from Ganoderma lucidum mitigates age-associated brain physiological decline via regulating sphingolipid metabolism and enhancing autophagy in mice. Front Aging Neurosci 13: 628860. https://doi.org/10.3389/fnagi.2021.628860
    [22] Jeong JH, Hong GL, Jeong YG, et al. (2023) Mixed medicinal mushroom mycelia attenuates Alzheimer's disease pathologies in vitro and in vivo. Curr Issues Mol Biol 45: 6775-6789. https://doi.org/10.3390/cimb45080428
    [23] Khatian N, Aslam M (2019) Effect of Ganoderma lucidum on memory and learning in mice. Clin Phytosci 5: 4. https://doi.org/10.1186/s40816-019-0101-7
    [24] Sharma P, Tulsawani R (2020) Ganoderma lucidum aqueous extract prevents hypobaric hypoxia induced memory deficit by modulating neurotransmission, neuroplasticity and maintaining redox homeostasis. Sci Rep 10: 8944. https://doi.org/10.1038/s41598-020-65812-5
    [25] Silva AM, Preto M, Grosso C, et al. (2023) Tracing the path between mushrooms and Alzheimer's disease—A literature review. Molecules 28: 5614. https://doi.org/10.3390/molecules28145614
    [26] Tran YH, Nguyen TTT, Nguyen PT, et al. (2021) Effects of Ganoderma Lucidum extract on morphine-induced addiction and memory impairment in mice. Biointerface Res Appl Chem 12: 1076-1084. https://doi.org/10.33263/BRIAC121.10761084
    [27] Rezvanirad A, Mardani M, Shirzad H, et al. (2016) Curcuma longa: A review of therapeutic effects in traditional and modern medical references. J Chem Pharmac Sci 9: 3438-4348.
    [28] Chen M, Du ZY, Zheng X, et al. (2018) Use of curcumin in diagnosis, prevention, and treatment of Alzheimer's disease. Neural Regen Res 13: 742-752. https://doi.org/10.4103/1673-5374.230303
    [29] da Costa IM, de Moura Freire MA, de Paiva Cavalcanti JRL, et al. (2019) Supplementation with curcuma longa reverses neurotoxic and behavioral damage in models of Alzheimer's disease: A systematic review. Curr Neuropharmacol 17: 406-421. https://doi.org/10.2174/0929867325666180117112610
    [30] Chen MH, Chiang BH (2020) Modification of curcumin-loaded liposome with edible compounds to enhance ability of crossing blood brain barrier. Colloid Surfaces A 599: 124862. https://doi.org/10.1016/j.colsurfa.2020.124862
    [31] Liu ZJ, Li ZH, Liu L, et al. (2016) Curcumin attenuates beta-amyloid-induced neuroinflammation via activation of peroxisome proliferator-activated receptor-gamma function in a rat model of Alzheimer's disease. Front Pharmacol 7: 261. https://doi.org/10.3389/fphar.2016.00261
    [32] Ringman JM, Frautschy SA, Teng E, et al. (2012) Oral curcumin for Alzheimer's disease: Tolerability and efficacy in a 24-week randomized, double blind, placebo-controlled study. Alz Res Therapy 43: 4. https://doi.org/10.1186/alzrt146
    [33] Assi AA, Farrag MMY, Badary DM, et al. (2023) Protective effects of curcumin and Ginkgo biloba extract combination on a new model of Alzheimer's disease. Inflammopharmacology 31: 1449-1464. https://doi.org/10.1007/s10787-023-01164-6
    [34] Park SY, Kim DSHL (2002) Discovery of natural products from Curcuma longa that protect cells from beta-amyloid insult: A drug discovery effort against Alzheimer's disease. J Nat Prod 65: 1227-1231. https://doi.org/10.1021/np010039x
    [35] do Nascimento-Silva NR, Bastos RP, da Silva FA (2022) Jambolan (Syzygium cumini (L.) Skeels): A review on its nutrients, bioactive compounds and health benefits. J Food Compos Anal 109: 104491. https://doi.org/10.1016/j.jfca.2022.104491
    [36] Hossain S, Islam J, Bhowmick S, et al. (2017) Effects of Syzygium cumini seed extract on the memory loss of Alzheimer's disease model rats. Adv Alzh Dis 6: 53-73. https://doi.org/10.4236/aad.2017.63005
    [37] Malik N, Javaid S, Ashraf W, et al. (2023) Long-term supplementation of Syzygium cumini (L.) skeels concentrate alleviates age-related cognitive deficit and oxidative damage: A comparative study of young vs. old mice. Nutrients 15: 666. https://doi.org/10.3390/nu15030666
    [38] Amir Rawa MS, Mazlan MKN, Ahmad R, et al. (2022) Roles of syzygium in anti-cholinesterase, anti-diabetic, anti-inflammatory, and antioxidant: From Alzheimer's perspective. Plants 11: 1476. https://doi.org/10.3390/plants11111476
    [39] Basiru OA, Ojo OA, Akuboh OS, et al. (2017) The protective effect of polyphenol-rich extract of Syzygium cumini leaves on cholinesterase and brain antioxidant status in alloxan-induced diabetic rats. Jordan J Biol Sci 11: 163-169.
    [40] Hawash ZAS, Yassien EM, Alotaibi BS, et al. (2023) Assessment of anti-Alzheimer pursuit of jambolan fruit extract and/or choline against AlCl3 toxicity in rats. Toxics 11: 509. https://doi.org/10.3390/toxics11060509
    [41] Mujawah A, Rauf A, Bawazeer S, et al. (2023) Isolation, structural elucidation, in vitro anti-α-glucosidase, anti-β-secretase, and in silico studies of bioactive compound isolated from Syzygium cumini L. Processes 11: 880. https://doi.org/10.3390/pr11030880
    [42] Al Mamun A, Hashimoto M, Katakura M, et al. (2014) Neuroprotective effect of madecassoside evaluated using amyloid β1-42-mediated in vitro and in vivo Alzheimer's disease models. Int J. Indigen Med Plants 47: 1669-1682.
    [43] Mamun AA, Hashimoto M, Hossain S, et al. (2015) Confirmation of the experimentally-proven therapeutic utility of madecassoside in an Aβ1-42 infusion rat model of Alzheimer's disease by in silico analyses. Adv Alz Dis 4: 37-44. https://doi.org/10.4236/aad.2015.42005
    [44] Ling Z, Zhou S, Zhou Y, et al. (2024) Protective role of madecassoside from Centella asiatica against protein L-isoaspartyl methyltransferase deficiency-induced neurodegeneration. Neuropharmacology 246: 109834. https://doi.org/10.1016/j.neuropharm.2023.109834
  • Reader Comments
  • © 2024 the Author(s), licensee AIMS Press. This is an open access article distributed under the terms of the Creative Commons Attribution License (http://creativecommons.org/licenses/by/4.0)
通讯作者: 陈斌, bchen63@163.com
  • 1. 

    沈阳化工大学材料科学与工程学院 沈阳 110142

  1. 本站搜索
  2. 百度学术搜索
  3. 万方数据库搜索
  4. CNKI搜索

Metrics

Article views(1190) PDF downloads(67) Cited by(0)

Figures and Tables

Figures(1)

/

DownLoad:  Full-Size Img  PowerPoint
Return
Return

Catalog