1
|
Adedayo LD, Aitokhuehi NG, Bamidele O, Adekeye AO, Ajayi AM, Onasanwo SA. Buchholzia coriacea seed attenuates scopolamine-induced memory impairment in Mice by down-regulating oxidative stress biomarkers. JOURNAL OF ETHNOPHARMACOLOGY 2025; 349:119882. [PMID: 40345272 DOI: 10.1016/j.jep.2025.119882] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/04/2025] [Revised: 04/23/2025] [Accepted: 04/24/2025] [Indexed: 05/11/2025]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Amnesia is a common feature of neurodegenerative disorders. The seeds of Buchholzia coriacea have been traditionally employed to treat various diseases, including inflammatory conditions, pain, infections, and neurological disorders. AIM This study aimed to investigate the anti-amnesic and antioxidant potentials of methanol extract of Buchholzia coriacea seed (MEBC) in mice. METHODS Memory impairment was induced with scopolamine at 3 mg/kg, which was administered i.p. Thirty (30) male Swiss mice (23-27 g) were divided into 6 treatment groups (n = 5): vehicle (10 mL/kg), scopolamine (3 mg/kg, i.p.), MEBC (50, 100, 200 mg/kg, p.o.), and donepezil (reference drug 1 mg/kg, p.o.), respectively. Memory function was assessed using the Morris Water Maze Test (MWMT) and the Y-maze Test (YMT). On the 7th day of administration, AchE, MDA, GSH, catalase, and nitrite were estimated to evaluate the neuroprotective action of Buchholzia coriacea via AchE inhibition and antioxidant activity. The hippocampus and prefrontal cortex were grossed for histological analysis. RESULTS The scopolamine treatment resulted in a decrease in percentage alternation, whereas mice pre-treated with MEBC prior to scopolamine had increased percentage alternation in YMT. The mean target quadrant time was significantly increased in the mice pretreated with MEBC. During the probe trial, mice administered with scopolamine had increased search error and chance level of time spent in the target quadrant region. However, MEBC pre-treated mice prior to scopolamine administration had increased latency time in the platform region in MWMT. Scopolamine-mediated changes in MDA and catalase activity were significantly attenuated by MEBC. There is an increased neuronal cell count of the hippocampus and prefrontal cortex region in groups treated with MEBC. CONCLUSION The findings suggest that MEBC seems to be a potent neuropharmacological agent against amnesia, and its mechanism(s) might be modulation of lipid peroxidation and oxidative stress activity associated with the Alzheimer type of amnesia.
Collapse
Affiliation(s)
- Lawrence D Adedayo
- Neurophysiology Research Unit, Department of Human Physiology, Federal University, Wukari, Nigeria; Neurosciences Unit, Physiology Department, University of Ibadan, Nigeria.
| | | | - Olubayode Bamidele
- Physiology Programme, College of Health Sciences, Bowen University Iwo, Nigeria
| | - Adeshina O Adekeye
- Department of Anatomy, College of Health Sciences, Afe-Babalola University, Nigeria
| | - Abayomi M Ajayi
- Pharmacology and Therapeutics Department, University of Ibadan, Nigeria
| | - Samuel A Onasanwo
- Neurosciences Unit, Physiology Department, University of Ibadan, Nigeria
| |
Collapse
|
2
|
Singh A, Singh L, Dalal D. Neuroprotective potential of hispidulin and diosmin: a review of molecular mechanisms. Metab Brain Dis 2025; 40:188. [PMID: 40257619 DOI: 10.1007/s11011-025-01615-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/01/2024] [Accepted: 04/11/2025] [Indexed: 04/22/2025]
Abstract
Flavonoids are an important class of natural products, particularly, belong to a class of plant secondary metabolites having a polyphenolic structure. They are widely found in fruits, vegetables, and certain beverages. Hispidulin and diosmin are naturally occurring flavonoids recognized for their potential health benefits, such as antioxidant, anti-inflammatory, and neuroprotective properties. Hispidulin is present in several plants, including Arnica montana, Salvia officinalis (sage), and Eupatorium arnottianum. Diosmin is mainly extracted from citrus fruits like lemons and oranges and can also be synthesized from hesperidin, another flavonoid found in citrus fruits. Neurodegenerative diseases are characterized by complex signaling pathways that contribute to neuronal deterioration. The JAK/STAT pathway is involved in inflammatory responses, while the NF-κB/NLRP3 pathway is associated with metabolic stress and inflammation, both facilitating neurodegeneration. Conversely, the AMPK/pGSK3β pathway is crucial for neuroprotection, regulating cellular responses to oxidative stress and promoting neuronal survival. Additionally, the BACE/Aβ pathway exacerbates neuronal damage by triggering inflammatory and oxidative stress responses, highlighting critical targets for therapeutic strategies. Hispidulin and diosmin have emerged as promising agents in the modulation of mediators involved in neuroinflammation and neurodegenerative diseases. Oxidative stress and inflammatory pathways, including those driven by Aβ/BACE1 and JAK/STAT signaling, are central to neuronal damage and disease progression. Recent studies highlight that hispidulin and diosmin exhibit notable neuroprotective effects by targeting these mediators. Hispidulin has been shown to impact key inflammatory cytokines and adhesion molecules, while diosmin influences proinflammatory cytokine production and inflammasome activation. Both compounds offer potential therapeutic benefits by modulating crucial mediators linked to neuroinflammation and neurodegeneration. This review article is designed to explore the intricate mechanistic interplay underlying the neuroprotective effects of hispidulin and diosmin.
Collapse
Affiliation(s)
- Anish Singh
- University Institute of Pharma Sciences, Chandigarh University, Mohali, Punjab, India
| | - Lovedeep Singh
- University Institute of Pharma Sciences, Chandigarh University, Mohali, Punjab, India.
| | - Diksha Dalal
- University Institute of Pharma Sciences, Chandigarh University, Mohali, Punjab, India
| |
Collapse
|
3
|
Atta AM, Rihan N, Abdelwaly AM, Nafie MS, Elgawish MS, Moustafa SM, Helal MA, Darwish KM. Development, biological evaluation, and molecular modelling of novel isocytosine and guanidine derivatives as BACE1 inhibitors using a fragment growing strategy. RSC Med Chem 2025:d4md00698d. [PMID: 40093519 PMCID: PMC11904611 DOI: 10.1039/d4md00698d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2024] [Accepted: 01/28/2025] [Indexed: 03/19/2025] Open
Abstract
Alzheimer's disease (AD) is a neurodegenerative condition characterized by significant synaptic loss and neuronal death in brain regions critical for cognitive functions. The disease is characterized by the formation of amyloid plaques, which are extracellular constructs consisting mainly of aggregated Aβ42. The latter is a peptide formed by the proteolytic cleavage of β-amyloid precursor protein (APP) by two enzymes, β- and γ-secretase. Therefore, inhibition of the aspartic protease β-secretase (BACE1) is considered a promising therapeutic approach for the treatment and prevention of Alzheimer's disease. Unfortunately, a limited number of β-secretase inhibitors have reached human trials and eventually failed due to inconclusive therapeutic and/or safety profiles. In this study, we developed drug-like molecules with a β-secretase inhibitory activity using a fragment growing strategy on isocytosine and acyl guanidine warheads. Our approach is based on optimizing the hydrophobic part of the molecules to obtain a conformationally restrained scaffold complementary to the hydrophobic pockets within the enzyme active site. We developed 32 compounds with promising in vitro inhibitory activity against BACE1 down to sub-micromolar IC50. Docking simulation studies were performed to understand the mode of binding of the prepared compounds. We demonstrated that compounds with superior activities, such as 16b and 16g, are able to provide the best balance between the steric shape and position of the polar substituent for achieving preferential anchoring into the S1, S3, S1', and S2' sub-pockets. Further, in vivo characterization of selected drug-like candidates of the benzimidazole series AMK-IV, namely 16a and 16k, demonstrated their ability to reduce oxidation stress and their safety within brain and liver tissues.
Collapse
Affiliation(s)
- Asmaa M Atta
- Pharmaceutical Chemistry Department, Faculty of Pharmacy, Badr University in Cairo (BUC) Badr City Cairo (P.O. 11829) Egypt
| | - Nouran Rihan
- Biomedical Sciences Program, University of Science and Technology, Zewail City of Science and Technology Giza (P.O. 12587) Egypt
| | - Ahmad M Abdelwaly
- Biomedical Sciences Program, University of Science and Technology, Zewail City of Science and Technology Giza (P.O. 12587) Egypt
- Institute for Computational Molecular Science, and, Department of Chemistry, Temple University Philadelphia Pennsylvania (P.O. 19122) USA
| | - Mohamed S Nafie
- Department of Chemistry, College of Sciences, University of Sharjah Sharjah (P.O. 27272) United Arab Emirates
- Chemistry Department, Faculty of Science, Suez Canal University Ismailia (P.O. 41522) Egypt
| | - Mohamed S Elgawish
- Medicinal Chemistry Department, Faculty of Pharmacy, Suez Canal University Ismailia (P.O. 41522) Egypt
- Center for Molecular Spectroscopy and Dynamics, Institute of Basic Science, Korea University Seoul (P.O. 02841) Republic of South Korea
| | - Samia M Moustafa
- Medicinal Chemistry Department, Faculty of Pharmacy, Suez Canal University Ismailia (P.O. 41522) Egypt
| | - Mohamed A Helal
- Biomedical Sciences Program, University of Science and Technology, Zewail City of Science and Technology Giza (P.O. 12587) Egypt
- Medicinal Chemistry Department, Faculty of Pharmacy, Suez Canal University Ismailia (P.O. 41522) Egypt
| | - Khaled M Darwish
- Medicinal Chemistry Department, Faculty of Pharmacy, Suez Canal University Ismailia (P.O. 41522) Egypt
- Department of Medicinal Chemistry, Faculty of Pharmacy, Galala University New Galala (P.O. 43713) Egypt
| |
Collapse
|
4
|
Prabha S, Choudhury A, Islam A, Thakur SC, Hassan MI. Understanding of Alzheimer's disease pathophysiology for therapeutic implications of natural products as neuroprotective agents. Ageing Res Rev 2025; 105:102680. [PMID: 39922232 DOI: 10.1016/j.arr.2025.102680] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2024] [Revised: 01/20/2025] [Accepted: 02/02/2025] [Indexed: 02/10/2025]
Abstract
Alzheimer's disease (AD) is a leading cause of dementia, affecting more than 24.3 million people worldwide in 2024. Sporadic AD (SAD) is more common and occurs in the geriatric population, while familial AD (FAD) is rare and appears before the age of 65 years. Due to progressive cholinergic neuronal loss and modulation in the PKC/MAPK pathway, β-secretase gets upregulated, leading to Aβ aggregation, which further activates tau kinases that form neurofibrillary tangles (NFT). Simultaneously, antioxidant enzymes are also upregulated, increasing oxidative stress (OS) and reactive species by impairing mitochondrial function, leading to DNA damage and cell death. This review discusses the classifications and components of several natural products (NPs) that target these signaling pathways for AD treatment. NPs, including alkaloids, polyphenols, flavonoids, polysaccharides, steroids, fatty acids, tannins, and polypeptides derived from plants, microbes, marine animals, venoms, insects, and mushrooms, are explored in detail. A synergistic combination of plant metabolites, together with prebiotics and probiotics has been shown to decrease Aβ aggregates by increasing the production of bioactive compounds. Toxins derived from venomous organisms have demonstrated effectiveness in modulating signaling pathways and reducing OS. Marine metabolites have also shown neuroprotective and anti-inflammatory properties. The cholera toxin B subunit and an Aβ15 fragment have been combined to create a possible oral AD vaccine, that showed enhancement of cognitive function in mice. Insect tea is also a reliable source of antioxidants. A functional edible mushroom snack bar showed an increment in cognitive markers. Future directions and therapeutic approaches for the treatment of AD can be improved by focusing more on NPs derived from these sources.
Collapse
Affiliation(s)
- Sneh Prabha
- Centre for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, Jamia Nagar, New Delhi 110025, India
| | - Arunabh Choudhury
- Centre for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, Jamia Nagar, New Delhi 110025, India
| | - Asimul Islam
- Centre for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, Jamia Nagar, New Delhi 110025, India
| | - Sonu Chand Thakur
- Centre for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, Jamia Nagar, New Delhi 110025, India.
| | - Md Imtaiyaz Hassan
- Centre for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, Jamia Nagar, New Delhi 110025, India.
| |
Collapse
|
5
|
Tie J, Wu H, Liu W, Li Y, Li L, Zhao S, Yuan Z, Mahmood K, Chen S, Wu H. Overexpression of SFPQ Improves Cognition and Memory in AD Mice. Neural Plast 2025; 2025:3934591. [PMID: 39949834 PMCID: PMC11824863 DOI: 10.1155/np/3934591] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2024] [Accepted: 01/07/2025] [Indexed: 02/16/2025] Open
Abstract
Alzheimer's disease (AD) is a complex neurodegenerative disorder with multifaceted pathogenesis, which has been extensively investigated, yet effective treatments remain lacking. Splicing factor proline and glutamine rich (SFPQ) is known to play a crucial role in neurodegenerative diseases, including antioxidant-related functions and regulating gene expression within brain neurons. However, the specific role of SFPQ in AD pathology is not well understood. In this study, an AD mouse model was established through lateral ventricular injection of amyloid-beta1-42 (Aβ 1-42). Subsequently, adeno-associated virus was administered to overexpress SFPQ in the hippocampus of AD mice. The results demonstrate that SFPQ overexpression improves recognition and memory in AD mice, while reducing AD-related marker proteins such as amyloid precursor protein (APP) and Tau. Additionally, synaptic and memory-associated proteins, as well as antioxidant proteins like glutathione S-transferase (GST) and heme oxygenase-1 (HO-1), were upregulated. The ratio of antiapoptotic protein Bcl-2 to proapoptotic protein Bax also increased. Furthermore, phosphorylated phosphoinositide 3-kinase (p-PI3K)/PI3K and phosphorylated protein kinase B (p-AKT)/AKT ratios were elevated, indicating activation of the PI3K/AKT signaling pathway. These findings suggest that SFPQ may serve as a promising molecular target for the prevention and treatment of AD.
Collapse
Affiliation(s)
- Jinshan Tie
- School of Rehabilitation, Kunming Medical University, Kunming 650500, Yunnan Province, China
- Rehabilitation Medicine Department, The Third People's Hospital of Yunnan Province, Kunming 650011, Yunnan Province, China
| | - Hongxiang Wu
- School of Rehabilitation, Kunming Medical University, Kunming 650500, Yunnan Province, China
| | - Wei Liu
- School of Rehabilitation, Kunming Medical University, Kunming 650500, Yunnan Province, China
| | - Yuying Li
- School of Rehabilitation, Kunming Medical University, Kunming 650500, Yunnan Province, China
| | - Lu Li
- School of Rehabilitation, Kunming Medical University, Kunming 650500, Yunnan Province, China
| | - Suju Zhao
- School of Rehabilitation, Kunming Medical University, Kunming 650500, Yunnan Province, China
| | - Zhijiao Yuan
- School of Rehabilitation, Kunming Medical University, Kunming 650500, Yunnan Province, China
| | - Khan Mahmood
- School of Rehabilitation, Kunming Medical University, Kunming 650500, Yunnan Province, China
| | - Shaochun Chen
- School of Rehabilitation, Kunming Medical University, Kunming 650500, Yunnan Province, China
| | - Huidong Wu
- School of Rehabilitation, Kunming Medical University, Kunming 650500, Yunnan Province, China
| |
Collapse
|
6
|
Kozin MS, Kulakova OG, Kiselev IS, Semina EV, Kakotkin VV, Agapov MA, Favorova OO. Mitochondrial Genome Variants and Alzheimer's Disease. BIOCHEMISTRY. BIOKHIMIIA 2025; 90:S146-S163. [PMID: 40164157 DOI: 10.1134/s0006297924603174] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/22/2024] [Revised: 03/04/2024] [Accepted: 05/14/2024] [Indexed: 04/02/2025]
Abstract
Alzheimer's disease (AD), a severe neurodegenerative disease of the central nervous system, is the most common cause of cognitive impairment in people over the age of 60. The etiology and pathogenesis of Alzheimer's disease are still unclear despite decades of active research. Numerous studies have shown that neurodegenerative processes in AD are associated with the mitochondrial dysfunction. In this review, we briefly discuss the results of these studies and present the reported evidence that mitochondrial dysfunction in AD is associated with mitochondrial DNA (mtDNA) variations. The results of association analysis of mtDNA haplogroups and individual polymorphic variants, including those whose combinations define haplogroups, with AD are described in detail. These data clearly indicate the role of variations in the mitochondrial genome in the susceptibility to AD, although the problem of significance of individual mtDNA variants is far from being resolved.
Collapse
Affiliation(s)
- Maxim S Kozin
- Immanuel Kant Baltic Federal University, Kaliningrad, 236016, Russia.
- Chazov National Medical Research Center of Cardiology, Ministry of Health of the Russian Federation, Moscow, 121552, Russia
- Pirogov Russian National Research Medical University, Ministry of Health of the Russian Federation, Moscow, 117513, Russia
| | - Olga G Kulakova
- Immanuel Kant Baltic Federal University, Kaliningrad, 236016, Russia
- Chazov National Medical Research Center of Cardiology, Ministry of Health of the Russian Federation, Moscow, 121552, Russia
- Pirogov Russian National Research Medical University, Ministry of Health of the Russian Federation, Moscow, 117513, Russia
| | - Ivan S Kiselev
- Immanuel Kant Baltic Federal University, Kaliningrad, 236016, Russia
- Chazov National Medical Research Center of Cardiology, Ministry of Health of the Russian Federation, Moscow, 121552, Russia
- Pirogov Russian National Research Medical University, Ministry of Health of the Russian Federation, Moscow, 117513, Russia
| | | | - Viktor V Kakotkin
- Immanuel Kant Baltic Federal University, Kaliningrad, 236016, Russia
| | - Mikhail A Agapov
- Immanuel Kant Baltic Federal University, Kaliningrad, 236016, Russia
| | - Olga O Favorova
- Immanuel Kant Baltic Federal University, Kaliningrad, 236016, Russia
- Chazov National Medical Research Center of Cardiology, Ministry of Health of the Russian Federation, Moscow, 121552, Russia
- Pirogov Russian National Research Medical University, Ministry of Health of the Russian Federation, Moscow, 117513, Russia
| |
Collapse
|
7
|
Dastan M, Rajaei Z, Sharifi M, Salehi H. Gallic acid ameliorates LPS-induced memory decline by modulating NF-κB, TNF-α, and Caspase 3 gene expression and attenuating oxidative stress and neuronal loss in the rat hippocampus. Metab Brain Dis 2024; 40:12. [PMID: 39556267 DOI: 10.1007/s11011-024-01441-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/13/2024] [Accepted: 10/19/2024] [Indexed: 11/19/2024]
Abstract
Neuroinflammation and apoptosis play critical roles in the pathogenesis of Alzheimer's disease (AD), which is responsible for most cases of dementia in the elderly people. Gallic acid is a phenolic compound with radical scavenging, anti-inflammatory and anti-apoptotic activities. This study aimed to explore the protective effects of gallic acid on LPS-induced spatial memory impairment and find the underlying mechanisms. Gallic acid was orally administered (100 mg/kg) to male Wistar rats for 12 days. LPS was injected intraperitoneally at a dose of 1 mg/kg on days 8-12. Morris water maze paradigm was used to evaluate spatial learning and memory. The mRNA level of nuclear factor kappa B (NF-κB), tumor necrosis factor-α (TNF-α) and Caspase 3, lipid peroxidation and total thiol level was assessed in the rat hippocampus. Neuronal loss and histological changes were also evaluated in the brain. LPS treatment resulted in spatial learning and memory impairment, upregulation of NF-κB, TNF-α, and Caspase 3 mRNA expression, increased lipid peroxidation, decreased total thiol level, and neuronal loss in the hippocampus. Moreover, treatment with gallic acid at a dosage of 100 mg/kg ameliorated memory decline, reduced the mRNA level of NF-κB, TNF-α, and Caspase 3, decreased lipid peroxidation and increased total thiol level in the hippocampus. Gallic acid also prevented LPS-induced neuronal loss and histological changes in the brain. Conclusively, our study demonstrated that gallic acid exerts neuroprotective effect against LPS-induced memory decline in rats. This outcome could be due to anti-inflammatory, antioxidant, and anti-apoptotic activities of gallic acid.
Collapse
Affiliation(s)
- Maryam Dastan
- Department of Physiology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Ziba Rajaei
- Department of Physiology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran.
| | - Mohammadreza Sharifi
- Department of Genetics and Molecular Biology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Hossein Salehi
- Department of Anatomical Sciences, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| |
Collapse
|
8
|
Xu W, Yan J, Shao A, Lenahan C, Gao L, Wu H, Zheng J, Zhang J, Zhang JH. Peroxisome and pexophagy in neurological diseases. FUNDAMENTAL RESEARCH 2024; 4:1389-1397. [PMID: 39734532 PMCID: PMC11670711 DOI: 10.1016/j.fmre.2023.04.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2022] [Revised: 02/15/2023] [Accepted: 04/19/2023] [Indexed: 12/31/2024] Open
Abstract
Peroxisomes and pexophagy have gained increasing attention in their role within the central nervous system (CNS) in recent years. In this review, we comprehensively discussed the physiological and pathological mechanisms of peroxisomes and pexophagy in neurological diseases. Peroxisomes communicate with mitochondria, endoplasmic reticulum, and lipid bodies. Their types, sizes, and shapes vary in different regions of the brain. Moreover, peroxisomes play an important role in oxidative homeostasis, lipid synthesis, and degradation in the CNS, whereas its dysfunction causes various neurological diseases. Therefore, selective removal of dysfunctional or superfluous peroxisomes (pexophagy) provides neuroprotective effects, which indicate a promising therapeutic target. However, pexophagy largely remains unexplored in neurological disorders. More studies are needed to explore the pexophagy's crosstalk mechanisms in neurological pathology.
Collapse
Affiliation(s)
- Weilin Xu
- Department of Neurosurgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310009, China
- Key Laboratory of Precise Treatment and Clinical Translational Research of Neurological Diseases, Hangzhou 310009, China
| | - Jun Yan
- Department of Neurosurgery, Affiliated Tumor Hospital of Guangxi Medical University, Guangxi 537406, China
| | - Anwen Shao
- Department of Neurosurgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310009, China
- Key Laboratory of Precise Treatment and Clinical Translational Research of Neurological Diseases, Hangzhou 310009, China
| | - Cameron Lenahan
- Burrell College of Osteopathic Medicine, New Mexico State University, Las Cruces, NM 88001, USA
| | - Liansheng Gao
- Department of Neurosurgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310009, China
- Key Laboratory of Precise Treatment and Clinical Translational Research of Neurological Diseases, Hangzhou 310009, China
| | - Haijian Wu
- Department of Neurosurgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310009, China
- Key Laboratory of Precise Treatment and Clinical Translational Research of Neurological Diseases, Hangzhou 310009, China
| | - Jingwei Zheng
- Department of Neurosurgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310009, China
- Key Laboratory of Precise Treatment and Clinical Translational Research of Neurological Diseases, Hangzhou 310009, China
| | - Jianmin Zhang
- Department of Neurosurgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310009, China
- Key Laboratory of Precise Treatment and Clinical Translational Research of Neurological Diseases, Hangzhou 310009, China
| | - John H. Zhang
- Department of Physiology & Pharmacology Loma Linda University, Loma Linda, CA 92350, USA
- Department of Neurosurgery Loma Linda University, Loma Linda, CA 92350, USA
| |
Collapse
|
9
|
Blades B, Hung YH, Belaidi AA, Volitakis I, Schultz AG, Cater MA, Cheung NS, Bush AI, Ayton S, La Fontaine S. Impaired cellular copper regulation in the presence of ApoE4. J Neurochem 2024; 168:3284-3307. [PMID: 39135362 DOI: 10.1111/jnc.16198] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2024] [Revised: 07/04/2024] [Accepted: 07/17/2024] [Indexed: 10/04/2024]
Abstract
The strongest genetic risk factor for late-onset Alzheimer's disease (AD) is allelic variation of the APOE gene, with the following risk structure: ε4 > ε3 > ε2. The biochemical basis for this risk profile is unclear. Here, we reveal a new role for the APOE gene product, apolipoprotein E (ApoE) in regulating cellular copper homeostasis, which is perturbed in the AD brain. Exposure of ApoE target replacement (TR) astrocytes (immortalised astrocytes from APOE knock-in mice) to elevated copper concentrations resulted in exacerbated copper accumulation in ApoE4- compared to ApoE2- and ApoE3-TR astrocytes. This effect was also observed in SH-SY5Y neuroblastoma cells treated with conditioned medium from ApoE4-TR astrocytes. Increased intracellular copper levels in the presence of ApoE4 may be explained by reduced levels and delayed trafficking of the copper transport protein, copper-transporting ATPase 1 (ATP7A/Atp7a), potentially leading to impaired cellular copper export. This new role for ApoE in copper regulation lends further biochemical insight into how APOE genotype confers risk for AD and reveals a potential contribution of ApoE4 to the copper dysregulation that is a characteristic pathological feature of the AD brain.
Collapse
Affiliation(s)
- Bryce Blades
- School of Life and Environmental Sciences, Deakin University, Burwood, Victoria, Australia
| | - Ya Hui Hung
- The Florey Neuroscience Institute, University of Melbourne, Parkville, Victoria, Australia
| | - Abdel A Belaidi
- The Florey Neuroscience Institute, University of Melbourne, Parkville, Victoria, Australia
| | - Irene Volitakis
- The Florey Neuroscience Institute, University of Melbourne, Parkville, Victoria, Australia
| | - Aaron G Schultz
- School of Life and Environmental Sciences, Deakin University, Burwood, Victoria, Australia
| | - Michael A Cater
- School of Life and Environmental Sciences, Deakin University, Burwood, Victoria, Australia
| | - Nam Sang Cheung
- School of Life and Environmental Sciences, Deakin University, Burwood, Victoria, Australia
| | - Ashley I Bush
- The Florey Neuroscience Institute, University of Melbourne, Parkville, Victoria, Australia
| | - Scott Ayton
- The Florey Neuroscience Institute, University of Melbourne, Parkville, Victoria, Australia
| | - Sharon La Fontaine
- School of Life and Environmental Sciences, Deakin University, Burwood, Victoria, Australia
- The Florey Neuroscience Institute, University of Melbourne, Parkville, Victoria, Australia
| |
Collapse
|
10
|
Pereira ME, Lima LS, Souza JV, de Souza da Costa N, da Silva JF, Guiloski IC, Irioda AC, Oliveira CS. Evaluation of the Neuroprotective Effect of Organic Selenium Compounds: An in Vitro Model of Alzheimer's Disease. Biol Trace Elem Res 2024; 202:2954-2965. [PMID: 37803188 DOI: 10.1007/s12011-023-03893-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/19/2023] [Accepted: 09/26/2023] [Indexed: 10/08/2023]
Abstract
Selenium (Se) is an essential trace element for human health and plays an important role in the development and maintenance of central nervous system functions. Se deficiency has been associated with cognitive decline and increased oxidative stress. The increase in oxidative stress is one of the hypotheses for the emergence and worsening of neurodegenerative diseases, such as Alzheimer's disease (AD). To investigate the neuroprotective effects of organic Se compounds in human neuroblastoma cells (SH-SY5Y) differentiated into cholinergic neurons-like. The SH-SY5Y cells were differentiated into cholinergic neuron-like with retinoic acid (RA) and brain-derived neurotrophic factor (BDNF). AD was mimicked exposing the cells to okadaic acid (OA) and beta-amyloid protein (Aβ). The neuroprotective effect of organic Se compounds, selenomethionine (SeMet) and Ebselen, was evaluated through cell viability tests, acetylcholinesterase and antioxidant enzyme activities, and detection of reactive oxygen species (ROS). None of the SeMet concentrations tested protected against the toxic effect of OA + Aβ. On the other hand, previous exposure to 0.1 and 1 µM Ebselen protected cells from the toxic effect of OA + Aβ. Cell differentiation induced by RA and BDNF exposure was effective, showing characteristics of neuronal cells, and pointing to a promising model of AD. Ebselen showed a protective effect, but more studies are needed to identify the mechanism of action.
Collapse
Affiliation(s)
- Meire Ellen Pereira
- Instituto de Pesquisa Pelé Pequeno Príncipe, Curitiba, PR, Brazil
- Faculdades Pequeno Príncipe, Curitiba, PR, Brazil
| | - Luiza Siqueira Lima
- Instituto de Pesquisa Pelé Pequeno Príncipe, Curitiba, PR, Brazil
- Faculdades Pequeno Príncipe, Curitiba, PR, Brazil
| | - Júlia Vicentin Souza
- Instituto de Pesquisa Pelé Pequeno Príncipe, Curitiba, PR, Brazil
- Faculdades Pequeno Príncipe, Curitiba, PR, Brazil
| | - Nayara de Souza da Costa
- Instituto de Pesquisa Pelé Pequeno Príncipe, Curitiba, PR, Brazil
- Faculdades Pequeno Príncipe, Curitiba, PR, Brazil
| | - Juliana Ferreira da Silva
- Instituto de Pesquisa Pelé Pequeno Príncipe, Curitiba, PR, Brazil
- Faculdades Pequeno Príncipe, Curitiba, PR, Brazil
| | - Izonete Cristina Guiloski
- Instituto de Pesquisa Pelé Pequeno Príncipe, Curitiba, PR, Brazil
- Faculdades Pequeno Príncipe, Curitiba, PR, Brazil
| | | | - Cláudia Sirlene Oliveira
- Instituto de Pesquisa Pelé Pequeno Príncipe, Curitiba, PR, Brazil.
- Faculdades Pequeno Príncipe, Curitiba, PR, Brazil.
| |
Collapse
|
11
|
Ghaffari MK, Rafati A, Karbalaei N, Haghani M, Nemati M, Sefati N, Namavar MR. The effect of intra-nasal co-treatment with insulin and growth factor-rich serum on behavioral defects, hippocampal oxidative-nitrosative stress, and histological changes induced by icv-STZ in a rat model. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2024; 397:4833-4849. [PMID: 38157024 DOI: 10.1007/s00210-023-02899-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/20/2023] [Accepted: 12/09/2023] [Indexed: 01/03/2024]
Abstract
Impaired insulin and growth factor functions are thought to drive many alterations in neurodegenerative diseases like dementia and seem to contribute to oxidative stress and inflammatory responses. Recent studies revealed that nasal growth factor therapy could induce neuronal and oligodendroglia protection in rodent brain damage induction models. Impairment of several growth factors signaling was reported in neurodegenerative diseases. So, in the present study, we examined the effects of intranasal co-treatment of insulin and a pool of growth factor-rich serum (GFRS) which separated from activated platelets on memory, and behavioral defects induced by intracerebroventricular streptozotocin (icv-STZ) rat model also investigated changes in the hippocampal oxidative-nitrosative state and histology. We found that icv-STZ injection (3 mg/kg bilaterally) impairs spatial learning and memory in Morris Water Maze, leads to anxiogenic-like behavior in the open field arena, and induces oxidative-nitrosative stress, neuroinflammation, and neuronal/oligodendroglia death in the hippocampus. GFRS (1µl/kg, each other day, 9 doses) and regular insulin (4 U/40 µl, daily, 18 doses) treatments improved learning, memory, and anxiogenic behaviors. The present study showed that co-treatment (GFRS + insulin with respective dose) has more robust protection against hippocampal oxidative-nitrosative stress, neuroinflammation, and neuronal/oligodendroglia survival in comparison with the single therapy. Memory and behavioral improvements in the co-treatment of insulin and GFRS could be attributed to their effects on neuronal/oligodendroglia survival and reduction of neuroinflammation in the hippocampus.
Collapse
Affiliation(s)
- Mahdi Khorsand Ghaffari
- Department of Physiology, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Ali Rafati
- Department of Physiology, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
- Histomorphometry and Stereology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Narges Karbalaei
- Department of Physiology, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
- Histomorphometry and Stereology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Masoud Haghani
- Department of Physiology, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
- Histomorphometry and Stereology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Marzieh Nemati
- Endocrinology and Metabolism Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Niloofar Sefati
- Department of Anatomical Sciences, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Mohammad Reza Namavar
- Histomorphometry and Stereology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran.
- Department of Anatomical Sciences, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran.
- Clinical Neurology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran.
| |
Collapse
|
12
|
Babygirija R, Sonsalla MM, Mill J, James I, Han JH, Green CL, Calubag MF, Wade G, Tobon A, Michael J, Trautman MM, Matoska R, Yeh CY, Grunow I, Pak HH, Rigby MJ, Baldwin DA, Niemi NM, Denu JM, Puglielli L, Simcox J, Lamming DW. Protein restriction slows the development and progression of pathology in a mouse model of Alzheimer's disease. Nat Commun 2024; 15:5217. [PMID: 38890307 PMCID: PMC11189507 DOI: 10.1038/s41467-024-49589-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Accepted: 06/12/2024] [Indexed: 06/20/2024] Open
Abstract
Dietary protein is a critical regulator of metabolic health and aging. Low protein diets are associated with healthy aging in humans, and dietary protein restriction extends the lifespan and healthspan of mice. In this study, we examined the effect of protein restriction (PR) on metabolic health and the development and progression of Alzheimer's disease (AD) in the 3xTg mouse model of AD. Here, we show that PR promotes leanness and glycemic control in 3xTg mice, specifically rescuing the glucose intolerance of 3xTg females. PR induces sex-specific alterations in circulating and brain metabolites, downregulating sphingolipid subclasses in 3xTg females. PR also reduces AD pathology and mTORC1 activity, increases autophagy, and improves the cognition of 3xTg mice. Finally, PR improves the survival of 3xTg mice. Our results suggest that PR or pharmaceutical interventions that mimic the effects of this diet may hold promise as a treatment for AD.
Collapse
Affiliation(s)
- Reji Babygirija
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA
- William S. Middleton Memorial Veterans Hospital, Madison, WI, USA
- Cellular and Molecular Biology Graduate Program, University of Wisconsin-Madison, Madison, WI, USA
| | - Michelle M Sonsalla
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA
- William S. Middleton Memorial Veterans Hospital, Madison, WI, USA
- Comparative Biomedical Sciences, University of Wisconsin-Madison, Madison, WI, USA
| | - Jericha Mill
- Department of Biochemistry, University of Wisconsin-Madison, Madison, WI, 53706, USA
| | - Isabella James
- Department of Biochemistry, University of Wisconsin-Madison, Madison, WI, 53706, USA
- Integrated Program in Biochemistry, University of Wisconsin-Madison, Madison, WI, 53706, USA
| | - Jessica H Han
- Cellular and Molecular Biology Graduate Program, University of Wisconsin-Madison, Madison, WI, USA
- Department of Biomolecular Chemistry, University of Wisconsin-Madison, Madison, WI, 53706, USA
| | - Cara L Green
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA
- William S. Middleton Memorial Veterans Hospital, Madison, WI, USA
| | - Mariah F Calubag
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA
- William S. Middleton Memorial Veterans Hospital, Madison, WI, USA
- Cellular and Molecular Biology Graduate Program, University of Wisconsin-Madison, Madison, WI, USA
| | - Gina Wade
- Department of Biochemistry, University of Wisconsin-Madison, Madison, WI, 53706, USA
- Integrated Program in Biochemistry, University of Wisconsin-Madison, Madison, WI, 53706, USA
| | - Anna Tobon
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA
- William S. Middleton Memorial Veterans Hospital, Madison, WI, USA
| | - John Michael
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA
- William S. Middleton Memorial Veterans Hospital, Madison, WI, USA
| | - Michaela M Trautman
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA
- William S. Middleton Memorial Veterans Hospital, Madison, WI, USA
- Nutrition and Metabolism Graduate Program, University of Wisconsin-Madison, Madison, WI, USA
| | - Ryan Matoska
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA
- William S. Middleton Memorial Veterans Hospital, Madison, WI, USA
| | - Chung-Yang Yeh
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA
- William S. Middleton Memorial Veterans Hospital, Madison, WI, USA
| | - Isaac Grunow
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA
- William S. Middleton Memorial Veterans Hospital, Madison, WI, USA
| | - Heidi H Pak
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA
- William S. Middleton Memorial Veterans Hospital, Madison, WI, USA
- Nutrition and Metabolism Graduate Program, University of Wisconsin-Madison, Madison, WI, USA
| | - Michael J Rigby
- Waisman Center, University of Wisconsin-Madison, Madison, WI, 53705, USA
- Neuroscience Training Program, University of Wisconsin-Madison, Madison, WI, 53705, USA
| | - Dominique A Baldwin
- Department of Biochemistry, University of Wisconsin-Madison, Madison, WI, 53706, USA
- Howard Hughes Medical Institute, University of Wisconsin-Madison, Madison, WI, 53706, USA
| | - Natalie M Niemi
- Department of Biochemistry & Molecular Biophysics, Washington University School of Medicine in St. Louis, St. Louis, MO, USA
| | - John M Denu
- Department of Biomolecular Chemistry, University of Wisconsin-Madison, Madison, WI, 53706, USA
- Nutrition and Metabolism Graduate Program, University of Wisconsin-Madison, Madison, WI, USA
| | - Luigi Puglielli
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA
- William S. Middleton Memorial Veterans Hospital, Madison, WI, USA
| | - Judith Simcox
- Cellular and Molecular Biology Graduate Program, University of Wisconsin-Madison, Madison, WI, USA
- Department of Biochemistry, University of Wisconsin-Madison, Madison, WI, 53706, USA
- Integrated Program in Biochemistry, University of Wisconsin-Madison, Madison, WI, 53706, USA
- Nutrition and Metabolism Graduate Program, University of Wisconsin-Madison, Madison, WI, USA
- Howard Hughes Medical Institute, University of Wisconsin-Madison, Madison, WI, 53706, USA
| | - Dudley W Lamming
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA.
- William S. Middleton Memorial Veterans Hospital, Madison, WI, USA.
- Cellular and Molecular Biology Graduate Program, University of Wisconsin-Madison, Madison, WI, USA.
- Comparative Biomedical Sciences, University of Wisconsin-Madison, Madison, WI, USA.
- Nutrition and Metabolism Graduate Program, University of Wisconsin-Madison, Madison, WI, USA.
| |
Collapse
|
13
|
Homolak J, Varvaras K, Sciacca V, Babic Perhoc A, Virag D, Knezovic A, Osmanovic Barilar J, Salkovic-Petrisic M. Insights into Gastrointestinal Redox Dysregulation in a Rat Model of Alzheimer's Disease and the Assessment of the Protective Potential of D-Galactose. ACS OMEGA 2024; 9:11288-11304. [PMID: 38496956 PMCID: PMC10938400 DOI: 10.1021/acsomega.3c07152] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/18/2023] [Revised: 12/14/2023] [Accepted: 01/04/2024] [Indexed: 03/19/2024]
Abstract
Recent evidence suggests that the gut plays a vital role in the development and progression of Alzheimer's disease (AD) by triggering systemic inflammation and oxidative stress. The well-established rat model of AD, induced by intracerebroventricular administration of streptozotocin (STZ-icv), provides valuable insights into the GI implications of neurodegeneration. Notably, this model leads to pathophysiological changes in the gut, including redox dyshomeostasis, resulting from central neuropathology. Our study aimed to investigate the mechanisms underlying gut redox dyshomeostasis and assess the effects of D-galactose, which is known to benefit gut redox homeostasis and alleviate cognitive deficits in this model. Duodenal rings isolated from STZ-icv animals and control groups were subjected to a prooxidative environment using 2,2'-azobis(2-amidinopropane) dihydrochloride (AAPH) or H2O2 with or without D-galactose in oxygenated Krebs buffer ex vivo. Redox homeostasis was analyzed through protein microarrays and functional biochemical assays alongside cell survival assessment. Structural equation modeling and univariate and multivariate models were employed to evaluate the differential response of STZ-icv and control samples to the controlled prooxidative challenge. STZ-icv samples showed suppressed expression of catalase and glutathione peroxidase 4 (GPX4) and increased baseline activity of enzymes involved in H2O2 and superoxide homeostasis. The altered redox homeostasis status was associated with an inability to respond to oxidative challenges and D-galactose. Conversely, the presence of D-galactose increased the antioxidant capacity, enhanced catalase and peroxidase activity, and upregulated superoxide dismutases in the control samples. STZ-icv-induced gut dysfunction is characterized by a diminished ability of the redox regulatory system to maintain long-term protection through the transcription of antioxidant response genes as well as compromised activation of enzymes responsible for immediate antioxidant defense. D-galactose can exert beneficial effects on gut redox homeostasis under physiological conditions.
Collapse
Affiliation(s)
- Jan Homolak
- Department
of Pharmacology & Croatian Institute for Brain Research, University of Zagreb School of Medicine, 10000 Zagreb, Croatia
- Interfaculty
Institute of Microbiology and Infection Medicine Tübingen, University of Tübingen, 72074 Tübingen, Germany
| | - Konstantinos Varvaras
- Department
of Medicine, School of Health Sciences, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece
| | - Vittorio Sciacca
- Faculty
of Medicine, University of Catania, 95131 Catania, Italy
| | - Ana Babic Perhoc
- Department
of Pharmacology & Croatian Institute for Brain Research, University of Zagreb School of Medicine, 10000 Zagreb, Croatia
| | - Davor Virag
- Department
of Pharmacology & Croatian Institute for Brain Research, University of Zagreb School of Medicine, 10000 Zagreb, Croatia
| | - Ana Knezovic
- Department
of Pharmacology & Croatian Institute for Brain Research, University of Zagreb School of Medicine, 10000 Zagreb, Croatia
| | - Jelena Osmanovic Barilar
- Department
of Pharmacology & Croatian Institute for Brain Research, University of Zagreb School of Medicine, 10000 Zagreb, Croatia
| | - Melita Salkovic-Petrisic
- Department
of Pharmacology & Croatian Institute for Brain Research, University of Zagreb School of Medicine, 10000 Zagreb, Croatia
| |
Collapse
|
14
|
Manzoor S, Gabr MT, Nafie MS, Raza MK, Khan A, Nayeem SM, Arafa RK, Hoda N. Discovery of Quinolinone Hybrids as Dual Inhibitors of Acetylcholinesterase and Aβ Aggregation for Alzheimer's Disease Therapy. ACS Chem Neurosci 2024; 15:539-559. [PMID: 38149821 DOI: 10.1021/acschemneuro.3c00588] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2023] Open
Abstract
The development of multitargeted therapeutics has evolved as a promising strategy to identify efficient therapeutics for neurological disorders. We report herein new quinolinone hybrids as dual inhibitors of acetylcholinesterase (AChE) and Aβ aggregation that function as multitargeted ligands for Alzheimer's disease. The quinoline hybrids (AM1-AM16) were screened for their ability to inhibit AChE, BACE1, amyloid fibrillation, α-syn aggregation, and tau aggregation. Among the tested compounds, AM5 and AM10 inhibited AChE activity by more than 80% at single-dose screening and possessed a remarkable ability to inhibit the fibrillation of Aβ42 oligomers at 10 μM. In addition, dose-dependent screening of AM5 and AM10 was performed, giving half-maximal AChE inhibitory concentration (IC50) values of 1.29 ± 0.13 and 1.72 ± 0.18 μM, respectively. In addition, AM5 and AM10 demonstrated concentration-dependent inhibitory profiles for the aggregation of Aβ42 oligomers with estimated IC50 values of 4.93 ± 0.8 and 1.42 ± 0.3 μM, respectively. Moreover, the neuroprotective properties of the lead compounds AM5 and AM10 were determined in SH-SY5Y cells incubated with Aβ oligomers. This work would enable future research efforts aiming at the structural optimization of AM5 and AM10 to develop potent dual inhibitors of AChE and amyloid aggregation. Furthermore, the in vivo assay confirmed the antioxidant activity of compounds AM5 and AM10 through increasing GSH, CAT, and SOD activities that are responsible for scavenging the ROS and restoring its normal level. Blood investigation illustrated the protective activity of the two compounds against lead-induced neurotoxicity through retaining hematological and liver enzymes near normal levels. Finally, immunohistochemistry investigation revealed the inhibitory activity of β-amyloid (Aβ) aggregation.
Collapse
Affiliation(s)
- Shoaib Manzoor
- Drug Design and Synthesis Laboratory, Department of Chemistry, Jamia Millia Islamia, New Delhi 110025, India
- Department of Pharmacy and Pharmacology, University of Bath, Bath BA2 7AY, U.K
| | - Moustafa T Gabr
- Molecular Imaging Innovations Institute, Department of Radiology, Weill Cornell Medicine, New York, New York10021, United States
| | - Mohamed S Nafie
- Department of Chemistry, College of Sciences, University of Sharjah, Sharjah (P.O. Box 27272), United Arab Emirates
- Chemistry Department, Faculty of Science, Suez Canal University, Ismailia 41522, Egypt
| | - Md Kausar Raza
- Department of Inorganic and Physical Chemistry, Indian Institute of Science, Bangalore 560012, India
| | - Ashma Khan
- Department of Chemistry, Aligarh Muslim University, Aligarh, UP 202002, India
| | - Shahid M Nayeem
- Department of Chemistry, Aligarh Muslim University, Aligarh, UP 202002, India
| | - Reem K Arafa
- Drug Design and Discovery Lab, Helmy Institute for Medical Sciences, Zewail City of Science, Technology and Innovation, Giza 12578, Egypt
- Biomedical Sciences Program, University of Science and Technology, Zewail City of Science, Technology and Innovation, Giza12578,Egypt
| | - Nasimul Hoda
- Drug Design and Synthesis Laboratory, Department of Chemistry, Jamia Millia Islamia, New Delhi 110025, India
| |
Collapse
|
15
|
Beggiato S, Ferrara F, Romani A, Cassano T, Trentini A, Valacchi G, Cervellati C, Ferraro L. Signature of paraoxonases in the altered redox homeostasis in Alzheimer's disease. Chem Biol Interact 2024; 388:110839. [PMID: 38142921 DOI: 10.1016/j.cbi.2023.110839] [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: 10/09/2023] [Revised: 11/29/2023] [Accepted: 12/17/2023] [Indexed: 12/26/2023]
Abstract
Paraoxonase (PON) enzymes (PON1, PON2 and PON3) exert antioxidant properties through arylesterase, lactonase and paraoxonase activities. Increasing findings suggested their potential involvement, particularly PON1 and PON2, in Alzheimer's disease (AD), a neurodegenerative pathology characterized by early oxidative stress. Specifically, decreased serum PON1-arylesterase and lactonase activities seem to be associated with an increased brain oxidative damage in early AD, leading to hypothesize that PON activity alterations might be an early event in AD. To address this hypothesis, the levels of 4-hydroxynonenal (4-HNE; i.e. a marker of oxidative stress damage) along with the protein expression and enzymatic activity of PON1 and PON2 have been investigated in the brain and serum of young [Postnatal day (PD)8-10, 20-25 and 60-65] asymptomatic 3xTg-AD female mice, one of the most used transgenic models of AD. At PD 8-10, there were no differences in hippocampus and prefrontal cortex (PFC) 4-HNE expression levels between 3xTg-AD mice compared to controls (Non-Tg mice). On the other hand, significant increased levels of 4-HNE were detected in PD 20-30 3xTg-AD mice hippocampus, while a significant reduction was observed in 3xTg-AD group at PD 60-65. In the PFC, 4-HNE levels were significantly reduced in 3xTg-AD mice brain at PD 20-30, while no differences in 4-HNE levels were detected at PD 60-65. No significant differences in arylesterase and lactonase activities were observed in the plasma of 3xTg-AD and Non-Tg mice at the different considered ages. Compared to Non-Tg mice, a reduction of brain arylesterase activity was found in 3xTg-AD female at PD 20-30 and PD 60-65, but it was significant only in the younger group. Finally, a similar trend was observed also for PON1 and PON2 protein levels, with both significantly, and solely, decreased in 3xTg-AD mice brain at PD 20-30. Overall, these findings suggest that the altered oxidative stress homeostasis in the 3xTg-AD female mice may be related to an early reduction in activity and expression of PONs enzymes most likely via a reduced brain arylesterases activity.
Collapse
Affiliation(s)
- Sarah Beggiato
- Department of Life Sciences and Biotechnology and LTTA Centre, University of Ferrara, Italy; Psychiatric Department, School of Medicine, University of Maryland, Baltimore MD, USA
| | - Francesca Ferrara
- Department of Chemical, Pharmaceutical and Agricultural Sciences, University of Ferrara, Italy
| | - Arianna Romani
- Department of Translational Medicine and for Romagna, University of Ferrara, Ferrara, Italy
| | - Tommaso Cassano
- Department of Medical and Surgical Sciences, University of Foggia, Foggia, Italy
| | - Alessandro Trentini
- Department of Translational Medicine and for Romagna, University of Ferrara, Ferrara, Italy
| | - Giuseppe Valacchi
- Department of Environmental and Prevention Sciences, University of Ferrara, Italy; Dept. of Animal Sciences, Plants for Human Health Institute, NC Research Campus, NC State University, Kannapolis, NC, USA; Department of Food and Nutrition, Kyung Hee University, Seoul, South Korea
| | - Carlo Cervellati
- Department of Translational Medicine and for Romagna, University of Ferrara, Ferrara, Italy
| | - Luca Ferraro
- Department of Life Sciences and Biotechnology and LTTA Centre, University of Ferrara, Italy; Psychiatric Department, School of Medicine, University of Maryland, Baltimore MD, USA.
| |
Collapse
|
16
|
Jeyakumar M, Jaya Balan D, Kiruthiga C, Jafni S, Pandima Devi K. α-bisabolol β-d-fucopyranoside (ABFP) ameliorates scopolamine-induced memory deficits through cholinesterase inhibition and attenuation of oxidative stress in zebrafish (Danio rerio). J Biochem Mol Toxicol 2024; 38:e23580. [PMID: 37961937 DOI: 10.1002/jbt.23580] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Revised: 10/06/2023] [Accepted: 10/18/2023] [Indexed: 11/15/2023]
Abstract
Alzheimer's disease (AD) is one of the major devastating neurodegenerative disorders associated with the gradual decline of an individual's memory, cognition, and ability to carry out day-to-day activities. In the present study, the neuroprotective ability of α-bisabolol β-d-fucopyranoside (ABFP) was assessed via measurement of antioxidant parameters like lipid peroxidation, glutathione peroxidation, glutathione, protein carbonyl content assays, and caspase-3 activity estimation. Moreover, the acute toxicity of ABFP was estimated in the zebrafish larval model. The results showed that ABFP exhibits little to no toxicity at lower concentrations in the acute toxicity test. ABFP-pretreated and scopolamine-exposed fish exhibited more exploratory behavior in the behavior assay than scopolamine-only induced groups. Additionally, the results of antioxidant enzyme assays revealed reduced oxidative stress and damage in ABFP-treated fish, while enzyme activity experiments carried out with brain homogenate from ABFP-treated fish showed decreased acetylcholinesterase enzyme activity. Overall, it can be concluded that ABFP has the potential to be a promising agent for the treatment of AD in the future.
Collapse
Affiliation(s)
| | | | | | - Sakthivel Jafni
- Department of Biotechnology, Alagappa University, Karaikudi, Tamil Nadu, India
| | - Kasi Pandima Devi
- Department of Biotechnology, Alagappa University, Karaikudi, Tamil Nadu, India
| |
Collapse
|
17
|
Andrade VHBD, M Rodrigues EY, Dias NAF, Ferreira GFC, Carvalho DBD, das Neves AR, Coronel PMV, Yonekawa MKA, Parisotto EB, Santos EAD, Souza AS, Paredes-Gamero EJ, de Sousa KS, Souza LLD, Resstel LBM, Baroni ACM, Lagatta DC. Neuroprotective Profile of Triazole Grandisin Analogue against Amyloid-Beta Oligomer-Induced Cognitive Impairment. ACS Chem Neurosci 2023; 14:4298-4310. [PMID: 38048522 DOI: 10.1021/acschemneuro.3c00443] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/06/2023] Open
Abstract
Alzheimer's disease (AD) is a neurodegenerative disorder caused by accumulation of amyloid-β oligomers (AβO) in the brain, neuroinflammation, oxidative stress, and cognitive decline. Grandisin, a tetrahydrofuran neolignan, exhibits relevant anti-inflammatory and antioxidant properties. Interestingly, grandisin-based compounds were shown to prevent AβO-induced neuronal death in vitro. However, no study has assessed the effect of these compounds on the AD animal model. This study focuses on a triazole grandisin analogue (TGA) synthesized using simplification and bioisosteric drug design, which resulted in improved potency and solubility compared with the parent compound. This study aimed to investigate the possible in vivo effects of TGA against AβO-induced AD. Male C57/Bl6 mice underwent stereotaxic intracerebroventricular AβO (90 μM) or vehicle injections. 24 h after surgery, animals received intraperitoneal treatment with TGA (1 mg/kg) or vehicle, administered on a 14 day schedule. One day after treatment completion, a novel object recognition task (NORT) was performed. Memantine (10 mg/kg) was administered as a positive control. NORT retention sessions were performed on days 8 and 16 after AβO injection. Immediately after retention sessions, animals were euthanized for cortex and hippocampus collection. Specimens were subjected to oxidative stress and cytokine analyses. TGA reduced the level of cortex/hippocampus lipoperoxidation and prevented cognitive impairment in AβO-injected mice. Additionally, TGA reduced tumor necrosis factor (TNF) and interferon-γ (IFN-γ) levels in the hippocampus. By contrast, memantine failed to prevent cortex/hippocampus lipid peroxidation, recognition memory decline, and AβO-induced increases in TNF and IFN-γ levels in the hippocampus. Thus, memantine was unable to avoid the AβO-induced persistent cognitive impairment. The results showed that TGA may prevent memory impairment by exerting antioxidant and anti-inflammatory effects in AβO-injected mice. Moreover, TGA exhibited a persistent neuroprotective effect compared to memantine, reflecting an innovative profile of this promising agent against neurodegenerative diseases, such as AD.
Collapse
Affiliation(s)
- Victor H B de Andrade
- Pharmaceutical Sciences, Food and Nutrition School, Federal University of Mato Grosso do Sul (UFMS), Campo Grande 79051-470, Brazil
| | - Erick Y M Rodrigues
- Pharmaceutical Sciences, Food and Nutrition School, Federal University of Mato Grosso do Sul (UFMS), Campo Grande 79051-470, Brazil
| | - Nayara A F Dias
- School of Medicine, Federal University of Mato Grosso do Sul (UFMS), Campo Grande 79051-470, Brazil
| | - Gabriela F C Ferreira
- School of Medicine, Federal University of Mato Grosso do Sul (UFMS), Campo Grande 79051-470, Brazil
| | - Diego B de Carvalho
- Pharmaceutical Sciences, Food and Nutrition School, Federal University of Mato Grosso do Sul (UFMS), Campo Grande 79051-470, Brazil
| | - Amarith R das Neves
- Pharmaceutical Sciences, Food and Nutrition School, Federal University of Mato Grosso do Sul (UFMS), Campo Grande 79051-470, Brazil
| | - Paola M V Coronel
- Pharmaceutical Sciences, Food and Nutrition School, Federal University of Mato Grosso do Sul (UFMS), Campo Grande 79051-470, Brazil
| | - Murilo K A Yonekawa
- Institute of Biosciences, Federal University of Mato Grosso do Sul (INBIO-UFMS), Campo Grande 79051-470, Brazil
| | - Eduardo B Parisotto
- Pharmaceutical Sciences, Food and Nutrition School, Federal University of Mato Grosso do Sul (UFMS), Campo Grande 79051-470, Brazil
| | - Edson A Dos Santos
- Institute of Biosciences, Federal University of Mato Grosso do Sul (INBIO-UFMS), Campo Grande 79051-470, Brazil
| | - Albert S Souza
- Institute of Biosciences, Federal University of Mato Grosso do Sul (INBIO-UFMS), Campo Grande 79051-470, Brazil
| | - Edgar J Paredes-Gamero
- Pharmaceutical Sciences, Food and Nutrition School, Federal University of Mato Grosso do Sul (UFMS), Campo Grande 79051-470, Brazil
| | - Kamylla S de Sousa
- Pharmaceutical Sciences, Food and Nutrition School, Federal University of Mato Grosso do Sul (UFMS), Campo Grande 79051-470, Brazil
| | - Letícia L de Souza
- Pharmaceutical Sciences, Food and Nutrition School, Federal University of Mato Grosso do Sul (UFMS), Campo Grande 79051-470, Brazil
| | - Leonardo B M Resstel
- School of Medicine of Ribeirão Preto, University of São Paulo, Ribeirão Preto 14049-900, Brazil
| | - Adriano C M Baroni
- Pharmaceutical Sciences, Food and Nutrition School, Federal University of Mato Grosso do Sul (UFMS), Campo Grande 79051-470, Brazil
| | - Davi C Lagatta
- Pharmaceutical Sciences, Food and Nutrition School, Federal University of Mato Grosso do Sul (UFMS), Campo Grande 79051-470, Brazil
| |
Collapse
|
18
|
Zhao L, Zheng L. A Review on Bioactive Anthraquinone and Derivatives as the Regulators for ROS. Molecules 2023; 28:8139. [PMID: 38138627 PMCID: PMC10745977 DOI: 10.3390/molecules28248139] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2023] [Revised: 12/09/2023] [Accepted: 12/11/2023] [Indexed: 12/24/2023] Open
Abstract
Anthraquinones are bioactive natural products, which are often found in medicinal herbs. These compounds exert antioxidant-related pharmacological actions including neuroprotective effects, anti-inflammation, anticancer, hepatoprotective effects and anti-aging, etc. Considering the benefits from their pharmacological use, recently, there was an upsurge in the development and utilization of anthraquinones as reactive oxygen species (ROS) regulators. In this review, a deep discussion was carried out on their antioxidant activities and the structure-activity relationships. The antioxidant mechanisms and the chemistry behind the antioxidant activities of both natural and synthesized compounds were furtherly explored and demonstrated. Due to the specific chemical activity of ROS, antioxidants are essential for human health. Therefore, the development of reagents that regulate the imbalance between ROS formation and elimination should be more extensive and rational, and the exploration of antioxidant mechanisms of anthraquinones may provide new therapeutic tools and ideas for various diseases mediated by ROS.
Collapse
Affiliation(s)
- Lihua Zhao
- Tianjin Renai College, Tianjin 301636, China;
| | - Lin Zheng
- College of Pharmaceutical Engineering of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| |
Collapse
|
19
|
Jiang Y, MacNeil LT. Simple model systems reveal conserved mechanisms of Alzheimer's disease and related tauopathies. Mol Neurodegener 2023; 18:82. [PMID: 37950311 PMCID: PMC10638731 DOI: 10.1186/s13024-023-00664-x] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2023] [Accepted: 10/04/2023] [Indexed: 11/12/2023] Open
Abstract
The lack of effective therapies that slow the progression of Alzheimer's disease (AD) and related tauopathies highlights the need for a more comprehensive understanding of the fundamental cellular mechanisms underlying these diseases. Model organisms, including yeast, worms, and flies, provide simple systems with which to investigate the mechanisms of disease. The evolutionary conservation of cellular pathways regulating proteostasis and stress response in these organisms facilitates the study of genetic factors that contribute to, or protect against, neurodegeneration. Here, we review genetic modifiers of neurodegeneration and related cellular pathways identified in the budding yeast Saccharomyces cerevisiae, the nematode Caenorhabditis elegans, and the fruit fly Drosophila melanogaster, focusing on models of AD and related tauopathies. We further address the potential of simple model systems to better understand the fundamental mechanisms that lead to AD and other neurodegenerative disorders.
Collapse
Affiliation(s)
- Yuwei Jiang
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Canada
| | - Lesley T MacNeil
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Canada.
- Farncombe Family Digestive Health Research Institute, McMaster University, Hamilton, Canada.
- Michael G. DeGroote Institute for Infectious Disease Research, McMaster University, 1280 Main St W, Hamilton, ON, L8S 4K1, Canada.
| |
Collapse
|
20
|
Akram M, Neha, Pinky, Saqib M, Salman M, Parvez S. Neuroprotective effect of ropinirole against Aβ 1-42 -induced neurochemical perturbations and cognitive impairments in a rodent model. Kaohsiung J Med Sci 2023; 39:1119-1128. [PMID: 37902529 DOI: 10.1002/kjm2.12770] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2023] [Revised: 07/04/2023] [Accepted: 07/31/2023] [Indexed: 10/31/2023] Open
Abstract
The primary objective of this study was to investigate the protective effects of ropinirole (ROP) medication given for an extended period following the induction of cognitive decline, oxidative stress, and deterioration of mitochondria in a Wistar rat model by Aβ1-42 . This study aimed to examine the neuroprotective efficacy of ROP in a stereotaxis model of AD. The Wistar rats were randomly assigned into four groups. Group I was considered as a sham, group II served as Aβ-infusion alone, Group III was Aβ1-42 + ROP (5 mg/kg/i.p.), and Group IV was Aβ1-42 + ROP (10 mg/kg/i.p.). Our research revealed that ROP (10 mg/kg, b.wt.) attenuates the cognitive deficits caused by Aβ1-42 -infused, which also correlates with the barnes maze, where (10 mg/kg, b.w.t.) shows significant improvement in spatial learning and memory. At the same time, ROP was rescued from oxidative damage, decreased lipid peroxidation rates, and inhibited acetylcholinesterase activity caused, demonstrating antioxidant benefits. In addition, a higher dose of ROP restored mitochondrial membrane potential in Aβ1-42 rats. Furthermore, histopathological examination showed that ROP treatment reduced neuronal loss, especially in the hippocampus. We conclude that ROP's protective effects in reducing oxidative stress and modulating mitochondrial function might have a propensity in AD pathogenesis.
Collapse
Affiliation(s)
- Mohd Akram
- Department of Molecular Medicine, School of Interdisciplinary Sciences and Technology, Jamia Hamdard, New Delhi, India
- Department of Tahaffuzi Wa Samaji Tibb, School of Unani Medicine, Jamia Hamdard, New Delhi, India
| | - Neha
- Department of Toxicology, School of Chemical and Life Sciences, Jamia Hamdard, New Delhi, India
| | - Pinky
- Department of Toxicology, School of Chemical and Life Sciences, Jamia Hamdard, New Delhi, India
| | - Mohd Saqib
- Department of Toxicology, School of Chemical and Life Sciences, Jamia Hamdard, New Delhi, India
| | - Mohd Salman
- Department of Toxicology, School of Chemical and Life Sciences, Jamia Hamdard, New Delhi, India
| | - Suhel Parvez
- Department of Toxicology, School of Chemical and Life Sciences, Jamia Hamdard, New Delhi, India
| |
Collapse
|
21
|
Holubiec MI, Alloatti M, Bianchelli J, Greloni F, Arnaiz C, Gonzalez Prinz M, Fernandez Bessone I, Pozo Devoto V, Falzone TL. Mitochondrial vulnerability to oxidation in human brain organoids modelling Alzheimer's disease. Free Radic Biol Med 2023; 208:394-401. [PMID: 37657763 DOI: 10.1016/j.freeradbiomed.2023.08.028] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/03/2023] [Revised: 08/24/2023] [Accepted: 08/25/2023] [Indexed: 09/03/2023]
Abstract
Reactive Oxygen Species (ROS) and mitochondrial dysfunction are implicated in the pathogenesis of Alzheimer's disease (AD), a common neurodegenerative disorder characterized by abnormal metabolism of the amyloid precursor protein (APP) in brain tissue. However, the exact mechanism by which abnormal APP leads to oxidative distress remains unclear. Damage to mitochondrial membrane and inhibition of mitochondrial respiration are thought to contribute to the progression of the disease. However, the lack of suitable human models that replicate pathological features, together with impaired cellular pathways, constitutes a major challenge in AD studies. In this work, we induced pluripotency in patient-derived skin fibroblasts carrying the Swedish mutation in App (APPswe), to generate human brain organoids that model AD, and studied redox regulation and mitochondrial homeostasis. We found time-dependent increases in AD-related pathological hallmarks in APPswe brain organoids, including elevated Aβ levels, increased extracellular amyloid deposits, and enhanced tau phosphorylation. Interestingly, using live-imaging spinning-disk confocal microscopy, we found an increase in mitochondrial fragmentation and a significant loss of mitochondrial membrane potential in APPswe brain organoids when subjected to oxidative conditions. Moreover, ratiometric dyes in a live imaging setting revealed a selective increase in mitochondrial superoxide anion and hydrogen peroxide levels in APPswe brain organoids that were coupled to impairments in cytosolic and mitochondrial redoxin protein expression. Our results suggest a selective increase in mitochondrial vulnerability to oxidative conditions in APPswe organoids, indicating that the abnormal metabolism of APP leads to specific changes in mitochondrial homeostasis that enhance the vulnerability to oxidation in AD.
Collapse
Affiliation(s)
- Mariana I Holubiec
- Instituto de Investigación en Biomedicina de Buenos Aires IBioBA (MPSP-CONICET), Partner Institute of the Max Planck Society, Godoy Cruz 2390, CABA, 1425, Buenos Aires, Argentina; Instituto de Biología Celular y Neurociencia IBCN (UBA-CONICET), Facultad de Medicina, Universidad de Buenos Aires, Paraguay 2155, CABA, 1121, Buenos Aires, Argentina
| | - Matias Alloatti
- Instituto de Biología Celular y Neurociencia IBCN (UBA-CONICET), Facultad de Medicina, Universidad de Buenos Aires, Paraguay 2155, CABA, 1121, Buenos Aires, Argentina
| | - Julieta Bianchelli
- Instituto de Investigación en Biomedicina de Buenos Aires IBioBA (MPSP-CONICET), Partner Institute of the Max Planck Society, Godoy Cruz 2390, CABA, 1425, Buenos Aires, Argentina
| | - Francisco Greloni
- Instituto de Biología Celular y Neurociencia IBCN (UBA-CONICET), Facultad de Medicina, Universidad de Buenos Aires, Paraguay 2155, CABA, 1121, Buenos Aires, Argentina
| | - Cayetana Arnaiz
- Instituto de Investigación en Biomedicina de Buenos Aires IBioBA (MPSP-CONICET), Partner Institute of the Max Planck Society, Godoy Cruz 2390, CABA, 1425, Buenos Aires, Argentina
| | - Melina Gonzalez Prinz
- Instituto de Investigación en Biomedicina de Buenos Aires IBioBA (MPSP-CONICET), Partner Institute of the Max Planck Society, Godoy Cruz 2390, CABA, 1425, Buenos Aires, Argentina
| | - Ivan Fernandez Bessone
- Instituto de Biología Celular y Neurociencia IBCN (UBA-CONICET), Facultad de Medicina, Universidad de Buenos Aires, Paraguay 2155, CABA, 1121, Buenos Aires, Argentina
| | - Victorio Pozo Devoto
- Instituto de Biología Celular y Neurociencia IBCN (UBA-CONICET), Facultad de Medicina, Universidad de Buenos Aires, Paraguay 2155, CABA, 1121, Buenos Aires, Argentina
| | - Tomas L Falzone
- Instituto de Investigación en Biomedicina de Buenos Aires IBioBA (MPSP-CONICET), Partner Institute of the Max Planck Society, Godoy Cruz 2390, CABA, 1425, Buenos Aires, Argentina; Instituto de Biología Celular y Neurociencia IBCN (UBA-CONICET), Facultad de Medicina, Universidad de Buenos Aires, Paraguay 2155, CABA, 1121, Buenos Aires, Argentina.
| |
Collapse
|
22
|
Kazemi S, Marefati N, Beheshti F, Salmani H, Bigham M, Hosseini M. The effect of olibanum on the rats with memory deficit induced by scopolamine. Cent Nerv Syst Agents Med Chem 2023; 23:CNSAMC-EPUB-134227. [PMID: 37680155 DOI: 10.2174/1871524923666230901142436] [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: 05/07/2023] [Revised: 06/10/2023] [Accepted: 07/18/2023] [Indexed: 09/09/2023]
Abstract
BACKGROUND Oxidative stress is an important contributor to Alzheimer's disease. Olibanum has therapeutic effects on various diseases. The effect of Olibanum on memory deficit induced by scopolamine (Sco) was challenged. METHODS Four groups were considered as (1) control (2) Sco, (3-4) Sco - Olib 100 and 200 mg/kg. Treatment by Olib or vehicle was done for two weeks. The third week was accompanied by the Morris water maze (MWM) and passive avoidance (PA) with Sco injection. On the last day, the brain and hippocampus were used for evaluation of the malondialdehyde (MDA), catalase (CAT), superoxide dismutase (SOD), and a total thiol group. RESULTS Sco increased the traveled time and distance to reach the hidden platform during five days of learning (p<0.01 - p<0.001) whereas it decreased the traveled time and distance (p<0.05- p<0.01) in the target area during the probe test of MWM. Sco also decreased delay time in the PA test (P<0.05 - P<0.001). Sco also decreased CAT, SOD, and thiol, whereas it, increased MDA in both the cortex and hippocampus (p<0.01 - p<0.001). Olib attenuated the impaired performance of the rats induced by Sco in MWM and PA tests. Olib reversed the increasing effects of Sco on MDA in both cortex and hippocampus and also reversed the attenuating effects of Sco on CAT, SOD, and thiol. CONCLUSION Olib had an inhibitory effect on memory deficit induced by Sco probably through its anti-oxidant property.
Collapse
Affiliation(s)
- Sara Kazemi
- Pharmacological Research Center of Medicinal Plants, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Narges Marefati
- Department of Physiology and Medical Physics, Faculty of Medicine, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Farimah Beheshti
- Neuroscience Research Center, Torbat Heydariyeh University of Medical Sciences, Torbat Heydariyeh, Iran
- Department of Physiology, School of Paramedical Sciences, Torbat Heydariyeh University of Medical Sciences, Torbat Heydariyeh, Iran
| | - Hossein Salmani
- Student Research Committee, Jiroft University of Medical Sciences, Jiroft, Iran
| | - Maryam Bigham
- Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mahmoud Hosseini
- Psychiatry and Behavioral Sciences Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
23
|
Sousa T, Moreira PI, Cardoso S. Current Advances in Mitochondrial Targeted Interventions in Alzheimer's Disease. Biomedicines 2023; 11:2331. [PMID: 37760774 PMCID: PMC10525414 DOI: 10.3390/biomedicines11092331] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2023] [Revised: 08/16/2023] [Accepted: 08/18/2023] [Indexed: 09/29/2023] Open
Abstract
Alzheimer's disease is the most prevalent neurodegenerative disorder and affects the lives not only of those who are diagnosed but also of their caregivers. Despite the enormous social, economic and political burden, AD remains a disease without an effective treatment and with several failed attempts to modify the disease course. The fact that AD clinical diagnosis is most often performed at a stage at which the underlying pathological events are in an advanced and conceivably irremediable state strongly hampers treatment attempts. This raises the awareness of the need to identify and characterize the early brain changes in AD, in order to identify possible novel therapeutic targets to circumvent AD's cascade of events. One of the most auspicious targets is mitochondria, powerful organelles found in nearly all cells of the body. A vast body of literature has shown that mitochondria from AD patients and model organisms of the disease differ from their non-AD counterparts. In view of this evidence, preserving and/or restoring mitochondria's health and function can represent the primary means to achieve advances to tackle AD. In this review, we will briefly assess and summarize the previous and latest evidence of mitochondria dysfunction in AD. A particular focus will be given to the recent updates and advances in the strategy options aimed to target faulty mitochondria in AD.
Collapse
Affiliation(s)
- Tiago Sousa
- Faculty of Medicine, University of Coimbra, 3000-370 Coimbra, Portugal;
| | - Paula I. Moreira
- CNC—Center for Neuroscience and Cell Biology, University of Coimbra, 3004-504 Coimbra, Portugal;
- CIBB—Center for Innovative Biomedicine and Biotechnology, University of Coimbra, 3004-504 Coimbra, Portugal
- Institute of Physiology, Faculty of Medicine, University of Coimbra, 3000-370 Coimbra, Portugal
| | - Susana Cardoso
- CNC—Center for Neuroscience and Cell Biology, University of Coimbra, 3004-504 Coimbra, Portugal;
- CIBB—Center for Innovative Biomedicine and Biotechnology, University of Coimbra, 3004-504 Coimbra, Portugal
- IIIUC—Institute for Interdisciplinary Research, University of Coimbra, 3030-789 Coimbra, Portugal
| |
Collapse
|
24
|
Singh N, Sherin GR, Mugesh G. Antioxidant and Prooxidant Nanozymes: From Cellular Redox Regulation to Next-Generation Therapeutics. Angew Chem Int Ed Engl 2023; 62:e202301232. [PMID: 37083312 DOI: 10.1002/anie.202301232] [Citation(s) in RCA: 41] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2023] [Revised: 04/20/2023] [Accepted: 04/21/2023] [Indexed: 04/22/2023]
Abstract
Nanozymes, nanomaterials with enzyme-mimicking activity, have attracted tremendous interest in recent years owing to their ability to replace natural enzymes in various biomedical applications, such as biosensing, therapeutics, drug delivery, and bioimaging. In particular, the nanozymes capable of regulating the cellular redox status by mimicking the antioxidant enzymes in mammalian cells are of great therapeutic significance in oxidative-stress-mediated disorders. As the distinction of physiological oxidative stress (oxidative eustress) and pathological oxidative stress (oxidative distress) occurs at a fine borderline, it is a great challenge to design nanozymes that can differentially sense the two extremes in cells, tissues and organs and mediate appropriate redox chemical reactions. In this Review, we summarize the advances in the development of redox-active nanozymes and their biomedical applications. We primarily highlight the therapeutic significance of the antioxidant and prooxidant nanozymes in various disease model systems, such as cancer, neurodegeneration, and cardiovascular diseases. The future perspectives of this emerging area of research and the challenges associated with the biomedical applications of nanozymes are described.
Collapse
Affiliation(s)
- Namrata Singh
- Department of Inorganic and Physical Chemistry, Indian Institute of Science, Bangalore, 560012, India
- Current address: Department of Medical Biochemistry and Biophysics, Karolinska Institute, Biomedicum, Solnavägen 9, 171 65, Solna, Sweden
| | - G R Sherin
- Department of Inorganic and Physical Chemistry, Indian Institute of Science, Bangalore, 560012, India
| | - Govindasamy Mugesh
- Department of Inorganic and Physical Chemistry, Indian Institute of Science, Bangalore, 560012, India
| |
Collapse
|
25
|
McNeilly AD, Gallagher JR, Evans ML, de Galan BE, Pedersen-Bjergaard U, Thorens B, Dinkova-Kostova AT, Huang JT, Ashford MLJ, McCrimmon RJ. Chronic hyperglycaemia increases the vulnerability of the hippocampus to oxidative damage induced during post-hypoglycaemic hyperglycaemia in a mouse model of chemically induced type 1 diabetes. Diabetologia 2023; 66:1340-1352. [PMID: 37015997 PMCID: PMC10244284 DOI: 10.1007/s00125-023-05907-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/26/2022] [Accepted: 01/26/2023] [Indexed: 04/06/2023]
Abstract
AIMS/HYPOTHESIS Chronic hyperglycaemia and recurrent hypoglycaemia are independently associated with accelerated cognitive decline in type 1 diabetes. Recurrent hypoglycaemia in rodent models of chemically induced (streptozotocin [STZ]) diabetes leads to cognitive impairment in memory-related tasks associated with hippocampal oxidative damage. This study examined the hypothesis that post-hypoglycaemic hyperglycaemia in STZ-diabetes exacerbates hippocampal oxidative stress and explored potential contributory mechanisms. METHODS The hyperinsulinaemic glucose clamp technique was used to induce equivalent hypoglycaemia and to control post-hypoglycaemic glucose levels in mice with and without STZ-diabetes and Nrf2-/- mice (lacking Nrf2 [also known as Nfe2l2]). Subsequently, quantitative proteomics based on stable isotope labelling by amino acids in cell culture and biochemical approaches were used to assess oxidative damage and explore contributory pathways. RESULTS Evidence of hippocampal oxidative damage was most marked in mice with STZ-diabetes exposed to post-hypoglycaemic hyperglycaemia; these mice also showed induction of Nrf2 and the Nrf2 transcriptional targets Sod2 and Hmox-1. In this group, hypoglycaemia induced a significant upregulation of proteins involved in alternative fuel provision, reductive biosynthesis and degradation of damaged proteins, and a significant downregulation of proteins mediating the stress response. Key differences emerged between mice with and without STZ-diabetes following recovery from hypoglycaemia in proteins mediating the stress response and reductive biosynthesis. CONCLUSIONS/INTERPRETATION There is a disruption of the cellular response to a hypoglycaemic challenge in mice with STZ-induced diabetes that is not seen in wild-type non-diabetic animals. The chronic hyperglycaemia of diabetes and post-hypoglycaemic hyperglycaemia act synergistically to induce oxidative stress and damage in the hippocampus, possibly leading to irreversible damage/modification to proteins or synapses between cells. In conclusion, recurrent hypoglycaemia in sub-optimally controlled diabetes may contribute, at least in part, to accelerated cognitive decline through amplifying oxidative damage in key brain regions, such as the hippocampus. DATA AVAILABILITY The datasets generated during and/or analysed during the current study are available in ProteomeXchange, accession no. 1-20220824-173727 ( www.proteomexchange.org ). Additional datasets generated during and/or analysed during the present study are available from the corresponding author upon reasonable request.
Collapse
Affiliation(s)
- Alison D McNeilly
- Division of Systems Medicine, School of Medicine, Ninewells Hospital and Medical School, Dundee, UK
| | - Jennifer R Gallagher
- Division of Systems Medicine, School of Medicine, Ninewells Hospital and Medical School, Dundee, UK
| | - Mark L Evans
- Wellcome-MRC Institute of Metabolic Science, University of Cambridge, Cambridge, UK
| | - Bastiaan E de Galan
- Radboud University Medical Center, Nijmegen, the Netherlands
- Department of Internal Medicine, Maastricht University Medical Center, Maastricht, the Netherlands
- CARIM School for Cardiovascular Diseases, Maastricht University, Maastricht, the Netherlands
| | | | - Bernard Thorens
- Faculty of Biology and Medicine, University of Lausanne, Lausanne, Switzerland
| | - Albena T Dinkova-Kostova
- Division of Cancer Research, School of Medicine, Ninewells Hospital and Medical School, Dundee, UK
| | - Jeffrey-T Huang
- Division of Systems Medicine, School of Medicine, Ninewells Hospital and Medical School, Dundee, UK
- Biomarker and Drug Analysis Core Facility, School of Medicine, Ninewells Hospital and Medical School, Dundee, UK
| | - Michael L J Ashford
- Division of Systems Medicine, School of Medicine, Ninewells Hospital and Medical School, Dundee, UK
| | - Rory J McCrimmon
- Division of Systems Medicine, School of Medicine, Ninewells Hospital and Medical School, Dundee, UK.
| |
Collapse
|
26
|
Lane HY, Wang SH, Lin CH. Differential relationships of NMDAR hypofunction and oxidative stress with cognitive decline. Psychiatry Res 2023; 326:115288. [PMID: 37343463 DOI: 10.1016/j.psychres.2023.115288] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Revised: 05/23/2023] [Accepted: 06/06/2023] [Indexed: 06/23/2023]
Abstract
NMDAR hypofunction and oxidative stress are implicated in the pathogenesis of Alzheimer's disease. D-amino acid oxidase (DAO) regulates NMDAR function. Glutathione, superoxide dismutase, and catalase are three first-line endogenous antioxidants. This study explored the associations of these potential biomarkers with mild cognitive impairment. Cognitive function and blood levels of DAO, glutathione, superoxide dismutase, and catalase were measured in 63 mild cognitive impairment patients and 24 healthy individuals every 6 months for 2 years. Among the patients, DAO and glutathione levels at baseline contributed to the cognitive decline 2 years later. Among the healthy individuals, only glutathione levels were associated with cognitive change. The four biomarkers differed in change directions (upward vs. downward) in the patients and in the healthy individuals. Among patients, glutathione levels were negatively correlated with superoxide dismutase and positively correlated with catalase, and DAO levels were negatively correlated with superoxide dismutase. To our knowledge, this is the first study to demonstrate the differential associations of NMDAR hypofunction and oxidative stress with cognitive change between the mild cognitive impairment patients and healthy people. Glutathione may be regarded as an aging marker for both mild cognitive impairment and normal aging; and DAO, a biomarker exclusively for mild cognitive impairment.
Collapse
Affiliation(s)
- Hsien-Yuan Lane
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung, Taiwan; Department of Psychiatry & Brain Disease Research Center, China Medical University Hospital, Taichung, Taiwan; Department of Psychology, College of Medical and Health Sciences, Asia University, Taichung, Taiwan
| | - Shi-Heng Wang
- National Center for Geriatrics and Welfare Research, National Health Research Institutes, Yunlin, Taiwan; College of Public Health, China Medical University, Taichung, Taiwan
| | - Chieh-Hsin Lin
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung, Taiwan; Department of Psychiatry, Kaohsiung Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Kaohsiung, Taiwan; School of Medicine, Chang Gung University, Taoyuan, Taiwan.
| |
Collapse
|
27
|
Kinno A, Kasamatsu S, Akaike T, Ihara H. Reactive Sulfur Species Omics Analysis in the Brain Tissue of the 5xFAD Mouse Model of Alzheimer's Disease. Antioxidants (Basel) 2023; 12:antiox12051105. [PMID: 37237971 DOI: 10.3390/antiox12051105] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2023] [Revised: 05/12/2023] [Accepted: 05/13/2023] [Indexed: 05/28/2023] Open
Abstract
Alzheimer's disease (AD) is a progressive neurodegenerative disorder whereby oxidative stress augmentation results in mitochondrial dysfunction and cell death by apoptosis. Emerging evidence indicates that reactive sulfur species (RSS), such as glutathione hydropersulfide (GSSH), is endogenously produced, functions as potent antioxidants, and regulate redox signaling through the formation of protein polysulfides. However, the relationship between RSS and AD pathogenesis is not fully understood. In this study, we analyzed endogenous RSS production in the brain tissue of a familial AD model (5xFAD) mouse using multiple RSS-omics approaches. Memory impairment, increased amyloid plaques, and neuroinflammation have been confirmed in 5xFAD mice. Quantitative RSS omics analysis revealed that the total polysulfide content was significantly decreased in the brains of 5xFAD mice, whereas there was no significant difference in the levels of glutathione, GSSH, or hydrogen sulfide between wild-type and 5xFAD mice. In contrast, a significant decline in the protein polysulfide status was observed in the brains of 5xFAD mice, suggesting that RSS production and subsequent redox signaling might be altered during the onset and progression of AD. Our findings have important implications for understanding the significance of RSS in the development of preventive and therapeutic strategies for AD.
Collapse
Grants
- 19K06537 Ministry of Education, Culture, Sports, Science and Technology
- 22K06148 Ministry of Education, Culture, Sports, Science and Technology
- JPMJCR2024 Japan Science and Technology Agency
- 2017G036 Smoking Research Foundation
- 2022-HI Fuji Foundation for Protein Research
- 21H05263 Ministry of Education, Culture, Sports, Science and Technology
- 22K19397 Ministry of Education, Culture, Sports, Science and Technology
- 16H04674 Ministry of Education, Culture, Sports, Science and Technology
- 20K21256 Ministry of Education, Culture, Sports, Science and Technology
- 21H02082 Ministry of Education, Culture, Sports, Science and Technology
- 21H05263 Ministry of Education, Culture, Sports, Science and Technology
Collapse
Affiliation(s)
- Ayaka Kinno
- Department of Biological Chemistry, Graduate School of Science, Osaka Metropolitan University, Osaka 599-8531, Japan
| | - Shingo Kasamatsu
- Department of Biological Chemistry, Graduate School of Science, Osaka Metropolitan University, Osaka 599-8531, Japan
| | - Takaaki Akaike
- Department of Environmental Medicine and Molecular Toxicology, Graduate School of Medicine, Tohoku University, Sendai 980-8575, Japan
| | - Hideshi Ihara
- Department of Biological Chemistry, Graduate School of Science, Osaka Metropolitan University, Osaka 599-8531, Japan
| |
Collapse
|
28
|
Varte V, Munkelwitz JW, Rincon-Limas DE. Insights from Drosophila on Aβ- and tau-induced mitochondrial dysfunction: mechanisms and tools. Front Neurosci 2023; 17:1184080. [PMID: 37139514 PMCID: PMC10150963 DOI: 10.3389/fnins.2023.1184080] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2023] [Accepted: 03/31/2023] [Indexed: 05/05/2023] Open
Abstract
Alzheimer's disease (AD) is the most prevalent neurodegenerative dementia in older adults worldwide. Sadly, there are no disease-modifying therapies available for treatment due to the multifactorial complexity of the disease. AD is pathologically characterized by extracellular deposition of amyloid beta (Aβ) and intracellular neurofibrillary tangles composed of hyperphosphorylated tau. Increasing evidence suggest that Aβ also accumulates intracellularly, which may contribute to the pathological mitochondrial dysfunction observed in AD. According with the mitochondrial cascade hypothesis, mitochondrial dysfunction precedes clinical decline and thus targeting mitochondria may result in new therapeutic strategies. Unfortunately, the precise mechanisms connecting mitochondrial dysfunction with AD are largely unknown. In this review, we will discuss how the fruit fly Drosophila melanogaster is contributing to answer mechanistic questions in the field, from mitochondrial oxidative stress and calcium dysregulation to mitophagy and mitochondrial fusion and fission. In particular, we will highlight specific mitochondrial insults caused by Aβ and tau in transgenic flies and will also discuss a variety of genetic tools and sensors available to study mitochondrial biology in this flexible organism. Areas of opportunity and future directions will be also considered.
Collapse
Affiliation(s)
- Vanlalrinchhani Varte
- Department of Neurology, McKnight Brain Institute, Norman Fixel Institute for Neurological Diseases, University of Florida, Gainesville, FL, United States
| | - Jeremy W. Munkelwitz
- Department of Neurology, McKnight Brain Institute, Norman Fixel Institute for Neurological Diseases, University of Florida, Gainesville, FL, United States
| | - Diego E. Rincon-Limas
- Department of Neurology, McKnight Brain Institute, Norman Fixel Institute for Neurological Diseases, University of Florida, Gainesville, FL, United States
- Department of Neuroscience, University of Florida, Gainesville, FL, United States
- Genetics Institute, University of Florida, Gainesville, FL, United States
| |
Collapse
|
29
|
Liuzzi GM, Petraglia T, Latronico T, Crescenzi A, Rossano R. Antioxidant Compounds from Edible Mushrooms as Potential Candidates for Treating Age-Related Neurodegenerative Diseases. Nutrients 2023; 15:nu15081913. [PMID: 37111131 PMCID: PMC10145943 DOI: 10.3390/nu15081913] [Citation(s) in RCA: 28] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2023] [Revised: 04/06/2023] [Accepted: 04/14/2023] [Indexed: 04/29/2023] Open
Abstract
The last century has seen an increase in our life expectancy. As a result, various age-related diseases, such as neurodegenerative diseases (NDs), have emerged, representing new challenges to society. Oxidative stress (OS), a condition of redox imbalance resulting from excessive production of reactive oxygen species, represents a common feature that characterizes the brains of elderly people, thus contributing to NDs. Consequently, antioxidant supplementation or dietary intake of antioxidant-containing foods could represent an effective preventive and therapeutic intervention to maintain the integrity and survival of neurons and to counteract the neurodegenerative pathologies associated with aging. Food contains numerous bioactive molecules with beneficial actions for human health. To this purpose, a wide range of edible mushrooms have been reported to produce different antioxidant compounds such as phenolics, flavonoids, polysaccharides, vitamins, carotenoids, ergothioneine, and others, which might be used for dietary supplementation to enhance antioxidant defenses and, consequently, the prevention of age-related neurological diseases. In this review, we summarized the role of oxidative stress in age-related NDs, focusing on the current knowledge of the antioxidant compounds present in edible mushrooms, and highlighting their potential to preserve healthy aging by counteracting age-associated NDs.
Collapse
Affiliation(s)
- Grazia Maria Liuzzi
- Department of Biosciences, Biotechnologies and Environment, University of Bari "Aldo Moro", 70126 Bari, Italy
| | - Tania Petraglia
- Department of Sciences, University of Basilicata, 85100 Potenza, Italy
| | - Tiziana Latronico
- Department of Biosciences, Biotechnologies and Environment, University of Bari "Aldo Moro", 70126 Bari, Italy
| | - Aniello Crescenzi
- School of Agricultural, Forestry, Food and Environmental Sciences, University of Basilicata, 85100 Potenza, Italy
| | - Rocco Rossano
- Department of Sciences, University of Basilicata, 85100 Potenza, Italy
| |
Collapse
|
30
|
Thawkar BS, Kaur G. Betanin mitigates scopolamine-induced cognitive impairment by restoring cholinergic function, boosting brain antioxidative status, and increasing BDNF level in the zebrafish model. FISH PHYSIOLOGY AND BIOCHEMISTRY 2023; 49:335-349. [PMID: 36991213 DOI: 10.1007/s10695-023-01185-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/23/2021] [Accepted: 03/15/2023] [Indexed: 05/04/2023]
Abstract
Betalains obtained from Beta vulgaris (family Caryophyllales) are regularly consumed as part of the regular diet with medicinal benefits due to antioxidant and anti-inflammatory properties. The objective of this article was to evaluate betanin's neuroprotective properties in a scopolamine-induced zebrafish paradigm. Betanin (BET) (50, 100, and 200 mg/L), and donepezil (10 mg/L) were delivered to zebrafish in a treatment tank once a day for 8 days, while memory impairment was produced by scopolamine (100 µM), which was given 60 min before behavioral assessments. The treatment dosages were determined based on acute toxicity studies. The existence of betacyanin and betaxanthins of BET was tested using liquid chromatography-mass spectrometry (LC-MS). The Y-maze task was used to examine the novelty and spatial memory, while the novel tank diving test was used to assess anxiety-like behavior (NTT). The activities of acetylcholinesterase (AChE) and the oxidative stress sensitivity in zebrafish brains were examined. Also, brain-derived neurotrophic factor (BDNF) level is quantified by an ELISA kit. Scopolamine-induced rises in AChE activity, memory loss, anxiety, and brain oxidant capacity were all reduced by BET. These results suggest that BET (50 and 100 mg/L) has a therapeutic ability to treat brain oxidative stress and cognitive deficits in amnesic zebrafish.
Collapse
Affiliation(s)
- Baban S Thawkar
- Department of Pharmacology, SPP School of Pharmacy & Technology Management, SVKM's NMIMS, V.L. Mehta Road, Vile Parle (W), Mumbai, 400056, India
| | - Ginpreet Kaur
- Department of Pharmacology, SPP School of Pharmacy & Technology Management, SVKM's NMIMS, V.L. Mehta Road, Vile Parle (W), Mumbai, 400056, India.
| |
Collapse
|
31
|
Moreira DA, Santos SD, Leiro V, Pêgo AP. Dendrimers and Derivatives as Multifunctional Nanotherapeutics for Alzheimer's Disease. Pharmaceutics 2023; 15:pharmaceutics15041054. [PMID: 37111540 PMCID: PMC10140951 DOI: 10.3390/pharmaceutics15041054] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 03/15/2023] [Accepted: 03/18/2023] [Indexed: 04/29/2023] Open
Abstract
Alzheimer's disease (AD) is the most prevalent form of dementia. It affects more than 30 million people worldwide and costs over US$ 1.3 trillion annually. AD is characterized by the brain accumulation of amyloid β peptide in fibrillar structures and the accumulation of hyperphosphorylated tau aggregates in neurons, both leading to toxicity and neuronal death. At present, there are only seven drugs approved for the treatment of AD, of which only two can slow down cognitive decline. Moreover, their use is only recommended for the early stages of AD, meaning that the major portion of AD patients still have no disease-modifying treatment options. Therefore, there is an urgent need to develop efficient therapies for AD. In this context, nanobiomaterials, and dendrimers in particular, offer the possibility of developing multifunctional and multitargeted therapies. Due to their intrinsic characteristics, dendrimers are first-in-class macromolecules for drug delivery. They have a globular, well-defined, and hyperbranched structure, controllable nanosize and multivalency, which allows them to act as efficient and versatile nanocarriers of different therapeutic molecules. In addition, different types of dendrimers display antioxidant, anti-inflammatory, anti-bacterial, anti-viral, anti-prion, and most importantly for the AD field, anti-amyloidogenic properties. Therefore, dendrimers can not only be excellent nanocarriers, but also be used as drugs per se. Here, the outstanding properties of dendrimers and derivatives that make them excellent AD nanotherapeutics are reviewed and critically discussed. The biological properties of several dendritic structures (dendrimers, derivatives, and dendrimer-like polymers) that enable them to be used as drugs for AD treatment will be pointed out and the chemical and structural characteristics behind those properties will be analysed. The reported use of these nanomaterials as nanocarriers in AD preclinical research is also presented. Finally, future perspectives and challenges that need to be overcome to make their use in the clinic a reality are discussed.
Collapse
Affiliation(s)
- Débora A Moreira
- i3S-Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal
- INEB-Instituto de Engenharia Biomédica, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal
- FEUP-Faculdade de Engenharia, Universidade do Porto, Rua Dr. Roberto Frias, 4200-465 Porto, Portugal
| | - Sofia D Santos
- i3S-Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal
- INEB-Instituto de Engenharia Biomédica, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal
| | - Victoria Leiro
- i3S-Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal
- INEB-Instituto de Engenharia Biomédica, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal
| | - Ana P Pêgo
- i3S-Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal
- INEB-Instituto de Engenharia Biomédica, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal
- ICBAS-Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, Rua Jorge de Viterbo Ferreira 228, 4050-313 Porto, Portugal
| |
Collapse
|
32
|
Decandia D, Gelfo F, Landolfo E, Balsamo F, Petrosini L, Cutuli D. Dietary Protection against Cognitive Impairment, Neuroinflammation and Oxidative Stress in Alzheimer's Disease Animal Models of Lipopolysaccharide-Induced Inflammation. Int J Mol Sci 2023; 24:ijms24065921. [PMID: 36982996 PMCID: PMC10051444 DOI: 10.3390/ijms24065921] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2023] [Revised: 03/14/2023] [Accepted: 03/16/2023] [Indexed: 03/30/2023] Open
Abstract
Alzheimer's disease (AD) is a rapidly growing epidemic with a heavy social and economic burden. Evidence suggests that systemic inflammation, dysregulation of the immune response and the resulting neuroinflammation and neurodegeneration play a significant role in AD pathogenesis. Currently, given that there is no fully convincing cure for AD, the interest in lifestyle factors (such as diet), which potentially delay onset and reduce the severity of symptoms, is increasing. This review is aimed at summarizing the effects of dietary supplementation on cognitive decline, neuroinflammation and oxidative stress in AD-like animal models with a focus on neuroinflammation induced by lipopolysaccharide (LPS) injection, which mimics systemic inflammation in animals. The compounds reviewed include curcumin, krill oil, chicoric acid, plasmalogens, lycopene, tryptophan-related dipeptides, hesperetin and selenium peptides. Despite the heterogeneity of these compounds, there is a strong consensus on their counteracting action on LPS-induced cognitive deficits and neuroinflammatory responses in rodents by modulating cell-signaling processes, such as the NF-κB pathway. Overall, dietary interventions could represent an important resource to oppose AD due to their influence in neuroprotection and immune regulation.
Collapse
Affiliation(s)
- Davide Decandia
- IRCCS Fondazione Santa Lucia, Via Ardeatina 306, 00179 Rome, Italy
- Department of Psychology, Sapienza University of Rome, Via dei Marsi 78, 00185 Rome, Italy
| | - Francesca Gelfo
- IRCCS Fondazione Santa Lucia, Via Ardeatina 306, 00179 Rome, Italy
- Department of Human Sciences, Guglielmo Marconi University, Via Plinio 44, 00193 Rome, Italy
| | - Eugenia Landolfo
- IRCCS Fondazione Santa Lucia, Via Ardeatina 306, 00179 Rome, Italy
| | - Francesca Balsamo
- IRCCS Fondazione Santa Lucia, Via Ardeatina 306, 00179 Rome, Italy
- Department of Human Sciences, Guglielmo Marconi University, Via Plinio 44, 00193 Rome, Italy
| | - Laura Petrosini
- IRCCS Fondazione Santa Lucia, Via Ardeatina 306, 00179 Rome, Italy
| | - Debora Cutuli
- IRCCS Fondazione Santa Lucia, Via Ardeatina 306, 00179 Rome, Italy
- Department of Psychology, Sapienza University of Rome, Via dei Marsi 78, 00185 Rome, Italy
| |
Collapse
|
33
|
Antioxidant Compounds in the Treatment of Alzheimer's Disease: Natural, Hybrid, and Synthetic Products. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2023; 2023:8056462. [PMID: 36865743 PMCID: PMC9974281 DOI: 10.1155/2023/8056462] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Revised: 02/03/2023] [Accepted: 02/10/2023] [Indexed: 02/23/2023]
Abstract
Alzheimer's disease (AD) which is associated with cognitive dysfunction and memory lapse has become a health concern. Various targets and pathways have been involved in AD's progress, such as deficit of acetylcholine (ACh), oxidative stress, inflammation, β-amyloid (Aβ) deposits, and biometal dyshomeostasis. Multiple pieces of evidence indicate that stress oxidative participation in an early stage of AD and the generated ROS could enable neurodegenerative disease leading to neuronal cell death. Hence, antioxidant therapies are applied in treating AD as a beneficial strategy. This review refers to the development and use of antioxidant compounds based on natural products, hybrid designs, and synthetic compounds. The results of using these antioxidant compounds were discussed with the given examples, and future directions for the development of antioxidants were evaluated.
Collapse
|
34
|
Isaev NK, Genrikhs EE, Stelmashook EV. Antioxidant Thymoquinone and Its Potential in the Treatment of Neurological Diseases. Antioxidants (Basel) 2023; 12:antiox12020433. [PMID: 36829993 PMCID: PMC9952318 DOI: 10.3390/antiox12020433] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2023] [Revised: 02/04/2023] [Accepted: 02/07/2023] [Indexed: 02/12/2023] Open
Abstract
Oxidative stress is one of the main pathogenic factors of neuron damage in neurodegenerative processes; this makes it an important therapeutic target to which the action of neuroprotectors should be directed. One of these drugs is thymoquinone. According to modern data, this substance has a wide range of pharmacological activity, including neuroprotective, which was demonstrated in experimental modeling of various neurodegenerative diseases and pathological conditions of the brain. The neuroprotective effect of thymoquinone is largely due to its antioxidant ability. Currently available data show that thymoquinone is an effective means to reduce the negative consequences of acute and chronic forms of cerebral pathology, leading to the normalization of the content of antioxidant enzymes and preventing an increase in the level of lipid peroxidation products. Antioxidant properties make this substance a promising basis for the development of prototypes of therapeutic agents aimed at the treatment of a number of degenerative diseases of the central nervous system.
Collapse
Affiliation(s)
- Nickolay K. Isaev
- Research Center of Neurology, 125367 Moscow, Russia
- Biological Faculty, M.V. Lomonosov Moscow State University, 119991 Moscow, Russia
| | | | - Elena V. Stelmashook
- Research Center of Neurology, 125367 Moscow, Russia
- Correspondence: ; Tel.: +7-(495)-9171908
| |
Collapse
|
35
|
Lane HY, Wang SH, Lin CH. Endogenous antioxidants predicted outcome and increased after treatment: A benzoate dose-finding, randomized, double-blind, placebo-controlled trial for Alzheimer's disease. Psychiatry Clin Neurosci 2023; 77:102-109. [PMID: 36335573 PMCID: PMC10099492 DOI: 10.1111/pcn.13504] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/23/2022] [Revised: 10/25/2022] [Accepted: 10/28/2022] [Indexed: 11/08/2022]
Abstract
AIM Previous pilot studies suggest that sodium benzoate may be a potential cognitive enhancer for patients with Alzheimer's disease (AD), schizophrenia, or late-life depression. Especially for AD treatment, a confirmatory trial with predictive biomarkers is urgently needed. This study aimed to confirm benzoate as a novel treatment for AD and to discover its optimal dose and biomarkers. METHODS A 24-week, dose-finding, randomized, double-blind, placebo-controlled trial, with clinical measurements at weeks 0, 8, 16, and 24, was conducted in three major medical centers in Taiwan. Among 154 patients screened for AD, 149 were eligible and randomized to one of the four treatments: (i) benzoate 500 group (fixed 500 mg/day); (ii) benzoate 750 (500 mg/day for the first 4 weeks, 750 mg/day from the 5th week); (iii) benzoate 1000 (500 mg/day for the first 4 weeks, 1000 mg/day from the 5th week); and (iv) placebo. The primary outcome measure was AD assessment scale-cognitive subscale (ADAS-cog). RESULTS The benzoate 1000 group performed best in improving ADAS-cog (P = 0.026 at week 24), with female advantage. Higher plasma catalase at baseline predicted better outcome. Benzoate receivers tended to have higher catalase and glutathione than placebo recipients after treatment. The four intervention groups showed similar safety profiles. CONCLUSIONS By enhancing two vital endogenous antioxidants, catalase and glutathione, sodium benzoate therapy improved cognition of patients with AD, with higher baseline catalase predicting better response. Supporting the oxidative stress theory, the results show promise for benzoate as a novel treatment for AD.
Collapse
Affiliation(s)
- Hsien-Yuan Lane
- Department of Psychiatry & Brain Disease Research Center, China Medical University Hospital, Taichung, Taiwan.,Graduate Institute of Biomedical Sciences, China Medical University, Taichung, Taiwan.,Department of Psychology, College of Medical and Health Sciences, Asia University, Taichung, Taiwan
| | - Shi-Heng Wang
- Department of Occupational Safety and Health, China Medical University, Taichung, Taiwan
| | - Chieh-Hsin Lin
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung, Taiwan.,Department of Psychiatry, Kaohsiung Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Kaohsiung, Taiwan.,School of Medicine, Chang Gung University, Taoyuan, Taiwan
| |
Collapse
|
36
|
Khezri MR, Yousefi K, Esmaeili A, Ghasemnejad-Berenji M. The Role of ERK1/2 Pathway in the Pathophysiology of Alzheimer's Disease: An Overview and Update on New Developments. Cell Mol Neurobiol 2023; 43:177-191. [PMID: 35038057 PMCID: PMC11415193 DOI: 10.1007/s10571-022-01191-x] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Accepted: 01/06/2022] [Indexed: 01/07/2023]
Abstract
Alzheimer's disease (AD) is the most common neurodegenerative disorder worldwide. Several findings suggest that correcting the dysregulated signaling pathways may offer a potential therapeutic approach in this disease. Extracellular signal-regulated kinase 1/2 (ERK1/2), a member of the mitogen-activated protein kinase family, plays a major role in regulation of cell proliferation, autophagy process, and protein synthesis. The available literature suggests dysregulated ERK1/2 in AD patients with potential implications in the multifaceted underlying pathologies of AD, including amyloid-β plaque formation, tau phosphorylation, and neuroinflammation. In this regard, in the current review, we aim to summarize the reports on the potential roles of ERK1/2 in AD pathophysiology.
Collapse
Affiliation(s)
- Mohammad Rafi Khezri
- Department of Pharmacology and Toxicology, School of Pharmacy, Urmia University of Medical Sciences, Urmia, Iran
| | - Keyvan Yousefi
- Department of Molecular and Cellular Pharmacology, University of Miami-Miller School of Medicine, Miami, FL, USA.
| | - Ayda Esmaeili
- Clinical Pharmacy Department, School of Pharmacy, Urmia University of Medical Sciences, Urmia, Iran
| | - Morteza Ghasemnejad-Berenji
- Department of Pharmacology and Toxicology, School of Pharmacy, Urmia University of Medical Sciences, Urmia, Iran.
- Experimental and Applied Pharmaceutical Research Center, Urmia University of Medical Sciences, Urmia, Iran.
- Department of Pharmacology and Toxicology, School of Pharmacy, Urmia University of Medical Sciences, PO Box: 5715799313, Urmia, Iran.
| |
Collapse
|
37
|
Investigation of the protective effects of lutein on memory and learning using behavioral methods in a male rat model of Alzheimer's disease. J Funct Foods 2022. [DOI: 10.1016/j.jff.2022.105319] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
|
38
|
Holubiec MI, Gellert M, Hanschmann EM. Redox signaling and metabolism in Alzheimer's disease. Front Aging Neurosci 2022; 14:1003721. [PMID: 36408110 PMCID: PMC9670316 DOI: 10.3389/fnagi.2022.1003721] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Accepted: 10/14/2022] [Indexed: 08/11/2023] Open
Abstract
Reduction and oxidation reactions are essential for biochemical processes. They are part of metabolic pathways and signal transduction. Reactive oxygen species (ROS) as second messengers and oxidative modifications of cysteinyl (Cys) residues are key to transduce and translate intracellular and intercellular signals. Dysregulation of cellular redox signaling is known as oxidative distress, which has been linked to various pathologies, including neurodegeneration. Alzheimer's disease (AD) is a neurodegenerative pathology linked to both, abnormal amyloid precursor protein (APP) processing, generating Aβ peptide, and Tau hyperphosphorylation and aggregation. Signs of oxidative distress in AD include: increase of ROS (H2O2, O2 •-), decrease of the levels or activities of antioxidant enzymes, abnormal oxidation of macromolecules related to elevated Aβ production, and changes in mitochondrial homeostasis linked to Tau phosphorylation. Interestingly, Cys residues present in APP form disulfide bonds that are important for intermolecular interactions and might be involved in the aggregation of Aβ. Moreover, two Cys residues in some Tau isoforms have been shown to be essential for Tau stabilization and its interaction with microtubules. Future research will show the complexities of Tau, its interactome, and the role that Cys residues play in the progression of AD. The specific modification of cysteinyl residues in redox signaling is also tightly connected to the regulation of various metabolic pathways. Many of these pathways have been found to be altered in AD, even at very early stages. In order to analyze the complex changes and underlying mechanisms, several AD models have been developed, including animal models, 2D and 3D cell culture, and ex-vivo studies of patient samples. The use of these models along with innovative, new redox analysis techniques are key to further understand the importance of the redox component in Alzheimer's disease and the identification of new therapeutic targets in the future.
Collapse
Affiliation(s)
- M. I. Holubiec
- IBioBA-MPSP Instituto de Investigación en Biomedicina de Buenos Aires, Partner Institute of the Max Planck Society, Buenos Aires, Argentina
| | - M. Gellert
- Institute for Medical Biochemistry and Molecular Biology, University Medicine Greifwald, University Greifswald, Greifswald, Germany
| | | |
Collapse
|
39
|
Costantino AR, Charbe N, Duarte Y, Gutiérrez M, Giordano A, Prasher P, Dua K, Mandolesi S, Zacconi FC. Toward the cholinesterase inhibition potential of TADDOL derivatives: Seminal biological and computational studies. Arch Pharm (Weinheim) 2022; 355:e2200142. [PMID: 35892245 DOI: 10.1002/ardp.202200142] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2022] [Revised: 06/03/2022] [Accepted: 07/11/2022] [Indexed: 11/07/2022]
Abstract
Alzheimer's disease (AD) is a degenerative neurological disease characterized by gradual loss of cognitive skills and memory. The exact pathogenesis involved still remains unrevealed, but several studies indicate the involvement of an array of different enzymes, underlining the multifactorial character of the disease. Inhibition of these enzymes is therefore a powerful approach in the development of AD treatments, with promising candidates, including acetylcholinesterase (AChE), butyrylcholinesterase (BuChE), and monoamine oxidase. Interestingly, AChE is the target of a major pesticide family (organophosphates), with several reports indicating an intersection between the pesticide's activity and AD. In this study, various TADDOL derivatives were synthesized and their in vitro activities as AChE/BuChE inhibitors as well as their antioxidant activities were studied. Molecular modeling studies revealed the capability of TADDOL derivatives to bind to AChE and induce inhibition, especially compounds 2b and 3c furnishing IC50 values of 36.78 ± 8.97 and 59.23 ± 5.31 µM, respectively. Experimental biological activities and molecular modeling studies clearly demonstrate that TADDOL derivatives with specific stereochemistry have an interesting potential for the design of potent AChE inhibitors. The encouraging results for compounds 2b and 3c indicate them as promising scaffolds for selective and potent AChE inhibitors.
Collapse
Affiliation(s)
- Andrea R Costantino
- INQUISUR, Departamento de Química, Universidad Nacional del Sur, Bahía Blanca, Argentina
| | - Nitin Charbe
- Facultad de Química y de Farmacia, Pontificia Universidad Católica de Chile, Santiago, Chile
- Department of Pharmaceutical Sciences, Rangel College of Pharmacy, Texas A&M University, Kingsville, Texas, USA
| | - Yorley Duarte
- Center for Bioinformatics and Integrative Biology, Facultad de Ciencias de la Vida, Universidad Andrés Bello, Santiago, Chile
| | - Margarita Gutiérrez
- Organic Synthesis Laboratory and Biological Activity (LSO-Act-Bio), Institute of Chemistry and Natural Resources, Universidad de Talca, Talca, Chile
| | - Ady Giordano
- Facultad de Química y de Farmacia, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Parteek Prasher
- Department of Chemistry, University of Petroleum & Energy Studies, Dehradun, Uttarakhand, India
| | - Kamal Dua
- Discipline of Pharmacy, Graduate School of Health, University of Technology Sydney, Ultimo, New South Wales, Australia
- Faculty of Health, Australian Research Centre in Complementary and Integrative Medicine, University of Technology Sydney, Ultimo, New South Wales, Australia
- Uttaranchal Institute of Pharmaceutical Sciences, Uttaranchal University, Dehradun, India
| | - Sandra Mandolesi
- INQUISUR, Departamento de Química, Universidad Nacional del Sur, Bahía Blanca, Argentina
| | - Flavia C Zacconi
- Facultad de Química y de Farmacia, Pontificia Universidad Católica de Chile, Santiago, Chile
- Institute for Biological and Medical Engineering, Schools of Engineering, Medicine and Biological Sciences, Pontificia Universidad Católica de Chile, Santiago, Chile
- The Research Center for Nanotechnology and Advanced Materials, CIEN-UC, Pontificia Universidad Católica de Chile, Santiago, Chile
| |
Collapse
|
40
|
Karapetyan G, Fereshetyan K, Harutyunyan H, Yenkoyan K. The synergy of β amyloid 1-42 and oxidative stress in the development of Alzheimer's disease-like neurodegeneration of hippocampal cells. Sci Rep 2022; 12:17883. [PMID: 36284177 PMCID: PMC9596457 DOI: 10.1038/s41598-022-22761-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2022] [Accepted: 10/19/2022] [Indexed: 01/20/2023] Open
Abstract
Alzheimer's disease (AD) is a type of dementia that affects memory, thinking and behavior. Symptoms eventually become severe enough to interfere with daily tasks. Understanding the etiology and pathogenesis of AD is necessary for the development of strategies for AD prevention and/or treatment, and modeling of this pathology is an important step in achieving this goal. β-amyloid peptide (Aβ) injection is a widely used approach for modeling AD. Nevertheless, it has been reported that the model constructed by injection of Aβ in combination with a prooxidant cocktail (ferrous sulfate, Aβ, and buthionine sulfoximine (BSO) (FAB)) best reflects the natural development of this disease. The relationship between oxidative stress and Aβ deposition and their respective roles in Aβ-induced pathology in different animal models of AD have been thoroughly investigated. In the current paper, we compared the effects of Aβ 1-42 alone with that of Aβ-associated oxidative stress induced by the FAB cocktail on the neurodegeneration of hippocampal cells in vitro. We constructed a FAB-induced AD model using rat primary hippocampal cells and analyzed the contribution of each compound. The study mainly focused on the prooxidant aspects of AD pathogenesis. Moreover, cellular bioenergetics was assessed and routine metabolic tests were performed to determine the usefulness of this model. The data clearly show that aggregated Aβ1-42 alone is significantly less toxic to hippocampal cells. Aggregated Aβ damages neurons, and glial cells proliferate to remove Aβ from the hippocampus. External prooxidant agents (Fe2+) or inhibition of internal antioxidant defense by BSO has more toxic effects on hippocampal cells than aggregated Aβ alone. Moreover, hippocampal cells fight against Aβ-induced damage more effectively than against oxidative damage. However, the combination of Aβ with external oxidative damage and inhibition of internal antioxidant defense is even more toxic, impairs cellular defense systems, and may mimic the late phase of AD-associated cell damage. Our findings strongly indicate a critical role for the combination of Aβ and oxidative stress in the development of neurodegeneration in vitro.
Collapse
Affiliation(s)
- Gohar Karapetyan
- grid.427559.80000 0004 0418 5743Neuroscience Laboratory, Cobrain Center, Yerevan State Medical University named after M. Heratsi, 2 Koryun Str., 0025 Yerevan, Armenia
| | - Katarine Fereshetyan
- grid.427559.80000 0004 0418 5743Neuroscience Laboratory, Cobrain Center, Yerevan State Medical University named after M. Heratsi, 2 Koryun Str., 0025 Yerevan, Armenia ,grid.427559.80000 0004 0418 5743Department of Biochemistry, Yerevan State Medical University named after M. Heratsi, Yerevan, Armenia
| | - Hayk Harutyunyan
- grid.427559.80000 0004 0418 5743Neuroscience Laboratory, Cobrain Center, Yerevan State Medical University named after M. Heratsi, 2 Koryun Str., 0025 Yerevan, Armenia
| | - Konstantin Yenkoyan
- grid.427559.80000 0004 0418 5743Neuroscience Laboratory, Cobrain Center, Yerevan State Medical University named after M. Heratsi, 2 Koryun Str., 0025 Yerevan, Armenia ,grid.427559.80000 0004 0418 5743Department of Biochemistry, Yerevan State Medical University named after M. Heratsi, Yerevan, Armenia
| |
Collapse
|
41
|
Regmi S, Liu DD, Shen M, Kevadiya BD, Ganguly A, Primavera R, Chetty S, Yarani R, Thakor AS. Mesenchymal stromal cells for the treatment of Alzheimer’s disease: Strategies and limitations. Front Mol Neurosci 2022; 15:1011225. [PMID: 36277497 PMCID: PMC9584646 DOI: 10.3389/fnmol.2022.1011225] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2022] [Accepted: 09/20/2022] [Indexed: 11/27/2022] Open
Abstract
Alzheimer’s disease (AD) is a major cause of age-related dementia and is characterized by progressive brain damage that gradually destroys memory and the ability to learn, which ultimately leads to the decline of a patient’s ability to perform daily activities. Although some of the pharmacological treatments of AD are available for symptomatic relief, they are not able to limit the progression of AD and have several side effects. Mesenchymal stem/stromal cells (MSCs) could be a potential therapeutic option for treating AD due to their immunomodulatory, anti-inflammatory, regenerative, antioxidant, anti-apoptotic, and neuroprotective effects. MSCs not only secret neuroprotective and anti-inflammatory factors to promote the survival of neurons, but they also transfer functional mitochondria and miRNAs to boost their bioenergetic profile as well as improve microglial clearance of accumulated protein aggregates. This review focuses on different clinical and preclinical studies using MSC as a therapy for treating AD, their outcomes, limitations and the strategies to potentiate their clinical translation.
Collapse
|
42
|
Khan S, Khan HU, Khan FA, Shah A, Wadood A, Ahmad S, Almehmadi M, Alsaiari AA, Shah FU, Kamran N. Anti-Alzheimer and Antioxidant Effects of Nelumbo nucifera L. Alkaloids, Nuciferine and Norcoclaurine in Alloxan-Induced Diabetic Albino Rats. Pharmaceuticals (Basel) 2022; 15:ph15101205. [PMID: 36297317 PMCID: PMC9608663 DOI: 10.3390/ph15101205] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2022] [Revised: 09/23/2022] [Accepted: 09/24/2022] [Indexed: 12/04/2022] Open
Abstract
The present study is aimed to determine the efficacy and dose response of the nuciferine (1), norcoclaurine (2) and crude extract of Nelumbo nucifera in managements of diabetes, Alzheimer disease and related allergies. Experimentally, alloxan (100 mg/kg body weight (b.w.))-induced diabetic rats (200−250 g) were divided into seven groups (n = 6). Group I: normal control, Group II: diabetic control, Group III: standard treated with glibenclamide and Group lV-VII: treated with methanolic crude extracts (100, 200 mg/kg), nuciferine and norcoclaurine (10 mg/kg b.w.) for 15 days. Different tests were performed, including blood glucose, body weights and antioxidant enzyme assays, i.e., superoxide dismutase (SOD), catalase test (CAT), lipid peroxidation assay (TBARS), glutathione assay (GSH) and acetylcholinesterase (AChE) assay. Nuciferine and norcoclaurine significantly reduced blood glucose (p < 0.05) and restored body weight in diabetic rats. Moreover, nuciferine and norcoclaurine (10 mg/kg) significantly recovered the antioxidant enzymes (SOD, CAT, GPx and GSH) which decreased during induced diabetes. Significant increase in TBARS was also observed in the diabetic group and nuciferine as well as norcoclaurine (10 mg/kg) inhibited the increase in TBARS in diabetic animals (p < 0.05), as compared to glibenclamide. AChE activity was significantly recovered by nuciferine and norcoclaurine (10 mg/kg) both in the blood and brain of the diabetic group (p < 0.05). Nuciferine and norcoclaurine showed potent inhibitory effects against α-glucosidase and α-amylase with IC50, 19.06 ± 0.03, 15.03 ± 0.09 μM and 24.07 ± 0.05, 18.04 ± 0.021 μM, as confirmed by molecular docking studies. This study concludes that nuciferine and norcoclaurine significantly improve memory and could be considered as an effective phytomedicine for diabetes, Alzheimer’s disease (AD) and oxidative stress.
Collapse
Affiliation(s)
- Shahnaz Khan
- Department of Chemistry, University of Science and Technology, Bannu 28100, Khyber Pakhtunkhwa, Pakistan
- Correspondence: (S.K.); (F.A.K.); Tel.: +92-3339724044 (F.A.K.)
| | - Hidayat Ullah Khan
- Department of Chemistry, University of Science and Technology, Bannu 28100, Khyber Pakhtunkhwa, Pakistan
| | - Farman Ali Khan
- Department of Chemistry, Shaheed Benazir Bhutto University, Sheringal, Dir Upper 18000, Khyber Pakhtunkhwa, Pakistan
- Correspondence: (S.K.); (F.A.K.); Tel.: +92-3339724044 (F.A.K.)
| | - Afzal Shah
- Department of Chemistry, University of Science and Technology, Bannu 28100, Khyber Pakhtunkhwa, Pakistan
| | - Abdul Wadood
- Department of Biochemistry, Abdul Wali khan University, Mardan 23200, Khyber Pakhtunkhwa, Pakistan
| | - Shujaat Ahmad
- Department of Pharmacy, Shaheed Benazir Bhutto University, Sheringal, Dir Upper 18000, Khyber Pakhtunkhwa, Pakistan
| | - Mazen Almehmadi
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Taif University, P.O. Box 11099, Taif 21944, Saudi Arabia
| | - Ahad Amer Alsaiari
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Taif University, P.O. Box 11099, Taif 21944, Saudi Arabia
| | - Farid Ullah Shah
- Department of Biochemistry, Rehman Medical Collage, Peshawar 25000, Khyber Pakhtunkhwa, Pakistan
| | - Naveed Kamran
- Lady Reading Hospital, Peshawar 25000, Khyber Pakhtunkhwa, Pakistan
| |
Collapse
|
43
|
Neuroprotective and Antioxidant Role of Oxotremorine-M, a Non-selective Muscarinic Acetylcholine Receptors Agonist, in a Cellular Model of Alzheimer Disease. Cell Mol Neurobiol 2022:10.1007/s10571-022-01274-9. [PMID: 36056992 DOI: 10.1007/s10571-022-01274-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2022] [Accepted: 08/16/2022] [Indexed: 12/13/2022]
Abstract
Alzheimer disease (AD) is a multifactorial and age-dependent neurodegenerative disorder, whose pathogenesis, classically associated with the formation of senile plaques and neurofibrillary tangles, is also dependent on oxidative stress and neuroinflammation chronicization. Currently, the standard symptomatic therapy, based on acetylcholinesterase inhibitors, showed a limited therapeutic potential, whereas disease-modifying treatment strategies are still under extensive research. Previous studies have demonstrated that Oxotremorine-M (Oxo), a non-selective muscarinic acetylcholine receptors agonist, exerts neurotrophic functions in primary neurons, and modulates oxidative stress and neuroinflammation phenomena in rat brain. In the light of these findings, in this study, we aimed to investigate the neuroprotective effects of Oxo treatment in an in vitro model of AD, represented by differentiated SH-SY5Y neuroblastoma cells exposed to Aβ1-42 peptide. The results demonstrated that Oxo treatment enhances cell survival, increases neurite length, and counteracts DNA fragmentation induced by Aβ1-42 peptide. The same treatment was also able to block oxidative stress and mitochondria morphological/functional impairment associated with Aβ1-42 cell exposure. Overall, these results suggest that Oxo, by modulating cholinergic neurotransmission, survival, oxidative stress response, and mitochondria functionality, may represent a novel multi-target drug able to achieve a therapeutic synergy in AD. Illustration of the main pathological hallmarks and mechanisms underlying AD pathogenesis, including neurodegeneration and oxidative stress, efficiently counteracted by treatment with Oxo, which may represent a promising therapeutic molecule. Created with BioRender.com under academic license.
Collapse
|
44
|
Theiss EL, Griebsch LV, Lauer AA, Janitschke D, Erhardt VKJ, Haas EC, Kuppler KN, Radermacher J, Walzer O, Portius D, Grimm HS, Hartmann T, Grimm MOW. Vitamin B12 Attenuates Changes in Phospholipid Levels Related to Oxidative Stress in SH-SY5Y Cells. Cells 2022; 11:cells11162574. [PMID: 36010649 PMCID: PMC9406929 DOI: 10.3390/cells11162574] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Revised: 02/18/2022] [Accepted: 08/15/2022] [Indexed: 01/04/2023] Open
Abstract
Oxidative stress is closely linked to Alzheimer’s disease (AD), and is detected peripherally as well as in AD-vulnerable brain regions. Oxidative stress results from an imbalance between the generation and degradation of reactive oxidative species (ROS), leading to the oxidation of proteins, nucleic acids, and lipids. Extensive lipid changes have been found in post mortem AD brain tissue; these changes include the levels of total phospholipids, sphingomyelin, and ceramide, as well as plasmalogens, which are highly susceptible to oxidation because of their vinyl ether bond at the sn-1 position of the glycerol-backbone. Several lines of evidence indicate that a deficiency in the neurotropic vitamin B12 is linked with AD. In the present study, treatment of the neuroblastoma cell line SH-SY5Y with vitamin B12 resulted in elevated levels of phosphatidylcholine, phosphatidylethanolamine, sphingomyelin, and plasmalogens. Vitamin B12 also protected plasmalogens from hydrogen peroxide (H2O2)-induced oxidative stress due to an elevated expression of the ROS-degrading enzymes superoxide-dismutase (SOD) and catalase (CAT). Furthermore, vitamin B12 elevates plasmalogen synthesis by increasing the expression of alkylglycerone phosphate synthase (AGPS) and choline phosphotransferase 1 (CHPT1) in SH-SY5Y cells exposed to H2O2-induced oxidative stress.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Oliver Walzer
- Experimental Neurology, Saarland University, 66424 Homburg, Germany
| | - Dorothea Portius
- Nutrition Therapy and Counseling, Campus Gera, SRH University of Applied Health Science, 07548 Gera, Germany
| | | | - Tobias Hartmann
- Experimental Neurology, Saarland University, 66424 Homburg, Germany
- Deutsches Institut für DemenzPrävention, Saarland University, 66424 Homburg, Germany
| | - Marcus Otto Walter Grimm
- Experimental Neurology, Saarland University, 66424 Homburg, Germany
- Deutsches Institut für DemenzPrävention, Saarland University, 66424 Homburg, Germany
- Nutrition Therapy and Counseling, Campus Rheinland, SRH University of Applied Health Sciences, 51377 Leverkusen, Germany
- Correspondence: or
| |
Collapse
|
45
|
Mandal PK, Roy RG, Samkaria A. Oxidative Stress: Glutathione and Its Potential to Protect Methionine-35 of Aβ Peptide from Oxidation. ACS OMEGA 2022; 7:27052-27061. [PMID: 35967059 PMCID: PMC9366984 DOI: 10.1021/acsomega.2c02760] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/04/2022] [Accepted: 07/07/2022] [Indexed: 05/15/2023]
Abstract
Alzheimer's disease (AD) is the most common neurodegenerative disorder with heterogeneous etiology. Intracellular neurofibrillary tangles caused by tau (τ) protein phosphorylation and extracellular senile plaques caused by aggregation of amyloid-beta (Aβ) peptide are characteristic histopathological hallmarks of AD. Oxidative stress (OS) is also suggested to play a role in the pathophysiology of AD. The antioxidant glutathione (GSH) is able to mitigate OS through the detoxification of free radicals. The clearance of these free radicals is reported to be affected when there is a decline in GSH levels in AD. These radicals further react with the methionine-35 (M-35) residue of Aβ and facilitate its subsequent oligomerization. This review presents a plausible model indicating the role of master antioxidant GSH to protect M35 of Aβ1-40/Aβ1-42 from oxidation in pathological conditions as compared to healthy controls.
Collapse
Affiliation(s)
- Pravat K. Mandal
- Neuroimaging
and Neurospectroscopy (NINS) Laboratory, National Brain Research Centre, Gurgaon Haryana 122051, India
- Florey
Institute of Neuroscience and Mental Health, Melbourne School of Medicine Campus, Parkville, VIC 3052, Australia
| | - Rimil Guha Roy
- Neuroimaging
and Neurospectroscopy (NINS) Laboratory, National Brain Research Centre, Gurgaon Haryana 122051, India
| | - Avantika Samkaria
- Neuroimaging
and Neurospectroscopy (NINS) Laboratory, National Brain Research Centre, Gurgaon Haryana 122051, India
| |
Collapse
|
46
|
Fagerli E, Escobar I, Ferrier FJ, Jackson CW, Perez-Lao EJ, Perez-Pinzon MA. Sirtuins and cognition: implications for learning and memory in neurological disorders. Front Physiol 2022; 13:908689. [PMID: 35936890 PMCID: PMC9355297 DOI: 10.3389/fphys.2022.908689] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Accepted: 06/30/2022] [Indexed: 11/18/2022] Open
Abstract
Sirtuins are an evolutionarily conserved family of regulatory proteins that function in an NAD+ -dependent manner. The mammalian family of sirtuins is composed of seven histone deacetylase and ADP-ribosyltransferase proteins (SIRT1-SIRT7) that are found throughout the different cellular compartments of the cell. Sirtuins in the brain have received considerable attention in cognition due to their role in a plethora of metabolic and age-related diseases and their ability to induce neuroprotection. More recently, sirtuins have been shown to play a role in normal physiological cognitive function, and aberrant sirtuin function is seen in pathological cellular states. Sirtuins are believed to play a role in cognition through enhancing synaptic plasticity, influencing epigenetic regulation, and playing key roles in molecular pathways involved with oxidative stress affecting mitochondrial function. This review aims to discuss recent advances in the understanding of the role of mammalian sirtuins in cognitive function and the therapeutic potential of targeting sirtuins to ameliorate cognitive deficits in neurological disorders.
Collapse
Affiliation(s)
| | | | | | | | | | - Miguel A. Perez-Pinzon
- Department of Neurology, Miller School of Medicine, University of Miami, Miami, FL, United States
| |
Collapse
|
47
|
Bartolome F, Carro E, Alquezar C. Oxidative Stress in Tauopathies: From Cause to Therapy. Antioxidants (Basel) 2022; 11:antiox11081421. [PMID: 35892623 PMCID: PMC9332496 DOI: 10.3390/antiox11081421] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2022] [Accepted: 07/20/2022] [Indexed: 02/01/2023] Open
Abstract
Oxidative stress (OS) is the result of an imbalance between the production of reactive oxygen species (ROS) and the antioxidant capacity of cells. Due to its high oxygen demand, the human brain is highly susceptible to OS and, thus, it is not a surprise that OS has emerged as an essential component of the pathophysiology of several neurodegenerative diseases, including tauopathies. Tauopathies are a heterogeneous group of age-related neurodegenerative disorders characterized by the deposition of abnormal tau protein in the affected neurons. With the worldwide population aging, the prevalence of tauopathies is increasing, but effective therapies have not yet been developed. Since OS seems to play a key role in tauopathies, it has been proposed that the use of antioxidants might be beneficial for tau-related neurodegenerative diseases. Although antioxidant therapies looked promising in preclinical studies performed in cellular and animal models, the antioxidant clinical trials performed in tauopathy patients have been disappointing. To develop effective antioxidant therapies, the molecular mechanisms underlying OS in tauopathies should be completely understood. Here, we review the link between OS and tauopathies, emphasizing the causes of OS in these diseases and the role of OS in tau pathogenesis. We also summarize the antioxidant therapies proposed as a potential treatment for tauopathies and discuss why they have not been completely translated to clinical trials. This review aims to provide an integrated perspective of the role of OS and antioxidant therapies in tauopathies. In doing so, we hope to enable a more comprehensive understanding of OS in tauopathies that will positively impact future studies.
Collapse
Affiliation(s)
- Fernando Bartolome
- Group of Neurodegenerative Diseases, Hospital Universitario 12 de Octubre Research Institute (imas12), 28041 Madrid, Spain;
- Network Center for Biomedical Research in Neurodegenerative Diseases (CIBERNED), Spain;
| | - Eva Carro
- Network Center for Biomedical Research in Neurodegenerative Diseases (CIBERNED), Spain;
- Neurobiology of Alzheimer’s Disease Unit, Chronic Disease Program, Instituto de Salud Carlos III, 28222 Madrid, Spain
| | - Carolina Alquezar
- Group of Neurodegenerative Diseases, Hospital Universitario 12 de Octubre Research Institute (imas12), 28041 Madrid, Spain;
- Correspondence:
| |
Collapse
|
48
|
Anupama KP, Antony A, Shilpa O, Raghu SV, Gurushankara HP. Jatamansinol from Nardostachys jatamansi Ameliorates Tau-Induced Neurotoxicity in Drosophila Alzheimer's Disease Model. Mol Neurobiol 2022; 59:6091-6106. [PMID: 35864434 DOI: 10.1007/s12035-022-02964-7] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Accepted: 07/14/2022] [Indexed: 11/27/2022]
Abstract
Nardostachys jatamansi has long been used to prepare Medhya Rasayana in traditional Indian Ayurveda medicine to treat neurological disorders and enhance memory. Jatamansinol from the N. jatamansi against Alzheimer's disease (AD) showed that it could be a multitargeted drug against AD. Drosophila is an ideal model organism for studying a progressive age-related neurodegenerative disease such as AD since its neuronal organizations and functioning are highly similar to that of humans. The current study investigates the neuroprotective properties of jatamansinol against Tau-induced neurotoxicity in the AD Drosophila model. Results indicate jatamansinol is not an antifeedant for larva and adult Drosophila. Lifespan, locomotor activity, learning and memory, Tau protein expression level, eye degeneration, oxidative stress level, and cholinesterase activities were analyzed in 10, 20, and 30-day-old control (wild type), and tauopathy flies reared on jatamansinol supplemented food or regular food without jatamansinol supplementation. Jatamansinol treatment significantly extends the lifespan, improves locomotor activity, enhances learning and memory, and reduces Tau protein levels in tauopathy flies. It boosts the antioxidant enzyme activities, prevents Tau-induced oxidative stress, ameliorates eye degeneration, and inhibits cholinesterase activities in Tau-induced AD model. This study provides the first evidence that jatamansinol protects against Tau's neurotoxic effect in the AD Drosophila model, and it can be a potential therapeutic drug candidate for AD.
Collapse
Affiliation(s)
- Kizhakke Purayil Anupama
- Department of Zoology, School of Biological Sciences, Central University of Kerala, Thejaswini Hills, Periya, Kasaragod, 671 320, Kerala, India
| | - Anet Antony
- Department of Zoology, School of Biological Sciences, Central University of Kerala, Thejaswini Hills, Periya, Kasaragod, 671 320, Kerala, India
| | - Olakkaran Shilpa
- Department of Zoology, School of Biological Sciences, Central University of Kerala, Thejaswini Hills, Periya, Kasaragod, 671 320, Kerala, India
| | - Shamprasad Varija Raghu
- Neurogenetics Lab, Department of Applied Zoology, Mangalore University, Mangalagangotri, 574 199, Karnataka, India
| | | |
Collapse
|
49
|
Clark JS, Kayed R, Abate G, Uberti D, Kinnon P, Piccirella S. Post-translational Modifications of the p53 Protein and the Impact in Alzheimer's Disease: A Review of the Literature. Front Aging Neurosci 2022; 14:835288. [PMID: 35572126 PMCID: PMC9096077 DOI: 10.3389/fnagi.2022.835288] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Accepted: 03/04/2022] [Indexed: 11/24/2022] Open
Abstract
Our understanding of Alzheimer's disease (AD) pathogenesis has developed with several hypotheses over the last 40 years, including the Amyloid and Tau hypotheses. More recently, the p53 protein, well-known as a genome guardian, has gained attention for its potential role in the early evolution of AD. This is due to the central involvement of p53's in the control of oxidative stress and potential involvement in the Amyloid and Tau pathways. p53 is commonly regulated by post-translational modifications (PTMs), which affect its conformation, increasing its capacity to adopt multiple structural and functional states, including those that can affect brain processes, thus contributing to AD development. The following review will explore the impact of p53 PTMs on its function and consequential involvement in AD pathogenesis. The greater understanding of the role of p53 in the pathogenesis of AD could result in more targeted therapies benefiting the many patients of this debilitating disease.
Collapse
Affiliation(s)
| | - Rakez Kayed
- Mitchell Center for Neurodegenerative Diseases, The University of Texas Medical Branch, Galveston, TX, United States
- Department of Neurology, Neuroscience and Cell Biology, The University of Texas Medical Branch, Galveston, TX, United States
| | - Giulia Abate
- Division of Pharmacology, Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Daniela Uberti
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | | | | |
Collapse
|
50
|
Jatamansinol from Nardostachys jatamansi (D.Don) DC. Protects Aβ 42-induced Neurotoxicity in Alzheimer's Disease Drosophila Model. Neurotoxicology 2022; 90:62-78. [PMID: 35247505 DOI: 10.1016/j.neuro.2022.02.011] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2021] [Revised: 02/14/2022] [Accepted: 02/25/2022] [Indexed: 01/02/2023]
Abstract
Nardostachys jatamansi (D. Don) DC. is an essential plant used in Indian Ayurveda to treat neurological disorders, and it enhances memory. Its active phytochemical(s) responsible for neuroprotection is not yet studied. One of the neurological disorders, namely Alzheimer's disease (AD) causes dementia, is not having pharmacological strategies to effectively prevent the onset of AD, cure or reverse AD progression, and treat cognitive symptoms. Here is an attempt to analyze the neuroprotective effect of jatamansinol isolated from N. jatamansi against Aβ42 protein-induced neurotoxicity using the Aβ42 protein expressed Drosophila Alzheimer's disease (AD) model. Oregon-K (OK) and AD flies were reared on regular or jatamansinol supplemented food and analyzed their lifespan, locomotor activity, learning and memory, eye degeneration, oxidative stress levels, antioxidant activities, cholinesterase activities, Aβ42 protein, and Aβ42 gene expression. Jatamansinol extends the lifespan, improves locomotor activity, enhances learning and memory, and reduces Aβ42 protein levels in AD flies. Jatamansinol boosts the antioxidant enzyme activities, prevents Aβ42 protein-induced oxidative stress, ameliorates eye degeneration, and inhibits cholinesterase activities in the AD model. This study evidences the protective effect of jatamansinol against the Aβ42 protein-induced neurotoxicity in the AD Drosophila model, suggesting its possible therapeutic potential against AD.
Collapse
|