1
|
Iijima S, Takeda K, Nagahiro T, Watanabe K, Ikegaya Y, Matsumoto N. Acute curcumin administration enhances delta oscillations in the hippocampus underlying object memory improvement. J Pharmacol Sci 2025; 158:95-102. [PMID: 40288828 DOI: 10.1016/j.jphs.2025.03.007] [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: 12/04/2024] [Revised: 03/10/2025] [Accepted: 03/14/2025] [Indexed: 04/29/2025] Open
Abstract
Curcumin mitigates memory deficits or improves memory when it is chronically administered to animals. Due to limited bioavailability of curcumin, it remains almost unknown whether acutely treated curcumin influences cognitive function and underlying neural activity. To address this question, we monitored behavior and neural activity in the hippocampus and medial prefrontal cortex of mice treated with vehicle or curcumin while they were engaged in a novel object recognition task. Object recognition memory performance in the novel object recognition task was increased in curcumin-treated mice. Moreover, delta oscillations in the hippocampus were enhanced in the curcumin-administered mice in the test trial. Altogether, acute curcumin treatment boosts delta oscillations for memory recognition possibly by neuromodulation.
Collapse
Affiliation(s)
- Sena Iijima
- Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, 113-0033, Japan
| | - Kinjiro Takeda
- Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, 113-0033, Japan
| | - Takeshi Nagahiro
- Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, 113-0033, Japan
| | - Kisa Watanabe
- Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, 113-0033, Japan
| | - Yuji Ikegaya
- Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, 113-0033, Japan; Institute for AI and Beyond, The University of Tokyo, Tokyo, 113-0033, Japan; Center for Information and Neural Networks, National Institute of Information and Communications Technology, Suita City, Osaka, 565-0871, Japan
| | - Nobuyoshi Matsumoto
- Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, 113-0033, Japan; Institute for AI and Beyond, The University of Tokyo, Tokyo, 113-0033, Japan.
| |
Collapse
|
2
|
Roy KK, Mehta DK, Das R. Reevaluating Alzheimer's disease treatment: Can phytochemicals bridge the therapeutic Gap? Neuroscience 2025; 575:1-18. [PMID: 40216186 DOI: 10.1016/j.neuroscience.2025.04.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2025] [Revised: 04/07/2025] [Accepted: 04/08/2025] [Indexed: 04/16/2025]
Abstract
Alzheimer's disease (AD) is a growing neurological disorder giving impact cognition and memory, posing a global health challenge with over 55 million individuals affected. It is the 7th foremost cause of dying worldwide, and its pervasiveness is expected to twofold in each five years, reaching 115 million by 2050. AD is characterized by neurofibrillary tangles, senile plaques, and oxidative stress, leading to synaptic failure and cognitive decline. Currently, there is no cure, and available FDA-approved drugs provide only symptomatic relief. The disease progresses through five phases- mild cognitive impairment (MCI), very severe, severe, moderate and mild AD. Research on AD focuses on various neurodegenerative pathways, including inflammation, oxidative stress, genetic factors, environmental variables, and amyloid-beta accumulation. Existing FDA-accepted drugs, like rivastigmine, memantine, galantamine, and donepezil, primarily address early symptoms but have limitations, including side effects and high costs. In this context, phytochemicals from plants, such as resveratrol, huperzine, quercetin, galantamine, and rosmarinic acid, show promise as potential treatments for AD and overcome the challenges and limitation of conventional treatment. These natural substances are being investigated for their ability to lower the risk of AD safely. However, there is a lack of comprehensive knowledge about their application, necessitating further research and clinical trials to explore their potential benefits and limitations. This review serves as an essential reference for advancing future studies on Alzheimer's disease. By thoroughly analyzing neurodegenerative pathways, addressing drug limitations, and highlighting the potential of phytochemicals, we establish a strong foundation for developing innovative therapeutic strategies. Closing the knowledge gap related to the use of phytochemicals in Alzheimer's management is not just important; it is critical for creating novel and more effective treatments for this challenging neurological condition.
Collapse
Affiliation(s)
- Kishor Kumar Roy
- M.M. College of Pharmacy, Maharishi Markandeshwar (Deemed to be) University, Mullana, Ambala, HR, India
| | - Dinesh Kumar Mehta
- M.M. College of Pharmacy, Maharishi Markandeshwar (Deemed to be) University, Mullana, Ambala, HR, India
| | - Rina Das
- M.M. College of Pharmacy, Maharishi Markandeshwar (Deemed to be) University, Mullana, Ambala, HR, India.
| |
Collapse
|
3
|
Xu H, Wang G, Jiang Z, Han Y, Zhao W, Zhang H, Liu H, Liu H, Li Z, Ji F. Ultrasmall Nanoparticles Mitigate Tau Hyperphosphorylation to Restore Synaptic Integrity and Boost Cognitive Function in Alzheimer's Disease. Adv Healthc Mater 2025:e2500941. [PMID: 40376857 DOI: 10.1002/adhm.202500941] [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: 04/20/2025] [Indexed: 05/18/2025]
Abstract
Tau hyperphosphorylation represents a critical pathological hallmark of Alzheimer's disease (AD), a prevalent neurodegenerative disorder characterized by progressive cognitive decline. The ubiquitin-specific proteases 14 (USP14) impairs proteasomal function and accelerates hyperphosphorylated Tau accumulation, making it an attractive therapeutic target for modulating the ubiquitin-proteasome pathway in AD treatment. In this study, it is reported that wogonoside-functionalized ultrasmall Cu2-xSe nanoparticles (CSPW NPs) significantly reduce hyperphosphorylated Tau accumulation and alleviate AD symptoms. The therapeutic mechanism involves activation of the ubiquitin-proteasome pathway through USP14 inhibition by CSPW NPs, thereby preventing hyperphosphorylated Tau accumulation. Furthermore, after cell membrane coating (CSPW@CM NPs), these nanoparticles efficiently cross the blood-brain barrier with focused ultrasound assistance and accumulate in the brain to target neurons. Within neurons, they inhibit USP14, reduce phosphorylated Tau deposition, enhance microtubule stability, mitigate synaptic loss, restore synaptic integrity, and ultimately alleviate cognitive dysfunction in AD mice. The findings highlight the substantial potential of USP14 modulation for mitigating Tau hyperphosphorylation in the treatment of AD and related tauopathies.
Collapse
Affiliation(s)
- Hanbing Xu
- Department of Anesthesiology, the First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, 215006, China
- Center for Molecular Imaging and Nuclear Medicine, State Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X), Suzhou Medical College, Soochow University, Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Suzhou, 215123, China
- Institute of Anesthesiology, Soochow University, Suzhou, 215006, China
| | - Gang Wang
- Department of Anesthesiology, the First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, 215006, China
- Institute of Anesthesiology, Soochow University, Suzhou, 215006, China
| | - Zhilin Jiang
- Center for Molecular Imaging and Nuclear Medicine, State Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X), Suzhou Medical College, Soochow University, Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Suzhou, 215123, China
| | - Yaobao Han
- Center for Molecular Imaging and Nuclear Medicine, State Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X), Suzhou Medical College, Soochow University, Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Suzhou, 215123, China
| | - Weiming Zhao
- Department of Anesthesiology, the First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, 215006, China
- Institute of Anesthesiology, Soochow University, Suzhou, 215006, China
| | - Hao Zhang
- Center for Molecular Imaging and Nuclear Medicine, State Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X), Suzhou Medical College, Soochow University, Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Suzhou, 215123, China
| | - Hong Liu
- Department of Anesthesiology and Pain Medicine, University of California Davis Health, Sacramento, CA, 95817, USA
| | - Huayue Liu
- Department of Anesthesiology, the First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, 215006, China
- Institute of Anesthesiology, Soochow University, Suzhou, 215006, China
- Ambulatory Surgery Center, The First Affiliated Hospital of Soochow University, Suzhou, 215006, China
| | - Zhen Li
- Center for Molecular Imaging and Nuclear Medicine, State Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X), Suzhou Medical College, Soochow University, Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Suzhou, 215123, China
| | - Fuhai Ji
- Department of Anesthesiology, the First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, 215006, China
- Institute of Anesthesiology, Soochow University, Suzhou, 215006, China
| |
Collapse
|
4
|
Kamat PK, Kalani A, Debnath N, Mushtaq Z, Tyagi SC, Tyagi N. Neuroprotective mechanism of hydrogen sulfide in okadaic acid-induced alzheimer-like pathology. GeroScience 2025:10.1007/s11357-025-01662-1. [PMID: 40341601 DOI: 10.1007/s11357-025-01662-1] [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: 10/18/2024] [Accepted: 04/09/2025] [Indexed: 05/10/2025] Open
Abstract
Okadaic acid (OKA) is a marine biotoxin that accumulates in shellfish and is responsible for causing diarrheic shellfish poisoning. OKA is a powerful and selective inhibitor of serine/threonine phosphatases 1 and 2A, which induces hyperphosphorylation of tau in vitro and in vivo leading to Alzheimer's disease (AD)-like pathology and memory impairment. Hydrogen sulfide (H2S), a gaseous signaling molecule produced endogenously in the brain, has been demonstrated to possess neuroprotective properties in various models of neurodegeneration. The aim of this study was to investigate the potential of H₂S in reducing OKA-induced Alzheimer's disease (AD)-like pathology, focusing on its effects on the GSK3β/Tau and CaMKII/CREB signaling pathways in mice. To test this hypothesis, we used age 8-10 weeks-old male C57BL/6J wild-type mice, divided into the following experimental groups: 1. Control group: Received a single intracerebroventricular (ICV) injection of artificial cerebrospinal fluid (aCSF). 2. WT + OKA group: Received a single ICV injection of OKA (100 ng/5 µl) bilaterally to induce AD-like pathology. OKA was dissolved in artificial cerebrospinal fluid. 3. WT + OKA + GYY4137 group: Received a single ICV injection of OKA (100 ng/5 µl) bilaterally, followed by GYY4137 (30 µM/kg) via drinking water for 21 days. 4. WT + GYY4137 group: Received only GYY4137 per se (30 µM/kg) via drinking water for 21 days. After the treatment period, synaptic proteins and neurodegeneration were evaluated using Western blotting, RT-PCR, and immunohistochemistry techniques. Our results demonstrate that OKA administration results in memory impairment with decreased cerebral blood flow (CBF). OKA also caused a significant decrease in synapse proteins (PSD95, MAP-2, BDNF, CaMKIIα, and Tubulin-3β) levels, along with increased expression of Tau, PHF-1, and GSK-3β and memory-associated signaling molecules and pCREB. Interestingly, IP administration of GYY4137 (30 µM/Kg; an H2S donor) for 21 days significantly improved the level of synapse proteins and memory function in OKA-treated mice. The findings of this study determine the neuroprotective mechanism of H2S in OKA-induced AD-like pathology through the modulation of Tau, GSK3β, and pCREB signaling. Therefore, H2S ameliorates OKA-induced memory impairment by improving synapse function and forgetfulness. As a result, H2S could be used as a promising therapeutic molecule against Alzheimer's disease-like pathology.
Collapse
Affiliation(s)
- Pradip K Kamat
- Departments of Neurology, Medical College of Georgia, Augusta University, Augusta, GA, 30912, USA
| | - Anuradha Kalani
- Disease Biology Lab and Molecular Oncology Lab, Department of Life Science and Biotechnology, Chhatrapati Shahu Ji Maharaj University, Kanpur, 208024, India
| | - Nabendu Debnath
- Centre for Molecular Biology, Central University of Jammu, Bagla (Raya-Suchani), District Samba, 181143, Jammu & Kashmir, India
| | - Zayd Mushtaq
- Department of Physiology, School of Medicine, Health Sciences Center, University of Louisville, A- 1201, Louisville, KY, 40202, USA
| | - Suresh C Tyagi
- Department of Physiology, School of Medicine, Health Sciences Center, University of Louisville, A- 1201, Louisville, KY, 40202, USA
| | - Neetu Tyagi
- Department of Physiology, School of Medicine, Health Sciences Center, University of Louisville, A- 1201, Louisville, KY, 40202, USA.
| |
Collapse
|
5
|
Kelliny S, Zhou X, Bobrovskaya L. Alzheimer's Disease and Frontotemporal Dementia: A Review of Pathophysiology and Therapeutic Approaches. J Neurosci Res 2025; 103:e70046. [PMID: 40387258 PMCID: PMC12087441 DOI: 10.1002/jnr.70046] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2024] [Revised: 04/01/2025] [Accepted: 05/02/2025] [Indexed: 05/20/2025]
Abstract
Alzheimer's disease (AD) is a devastating form of dementia, with the number of affected individuals rising sharply. The main hallmarks of the disease include amyloid-beta plaque deposits and neurofibrillary tangles consisting of hyperphosphorylated tau protein, besides other pathological features that contribute to the disease's complexity. The causes of sporadic AD are multifactorial and mostly age-related and involve risk factors such as diabetes and cardiovascular or cerebrovascular disorders. Frontotemporal dementia (FTD) is another type of dementia characterized by a spectrum of behaviors, memory, and motor abnormalities and associated with abnormal depositions of protein aggregation, including tau protein. Currently approved medications are symptomatic, and no disease-modifying therapy is available to halt the disease progression. Therefore, the development of multi-targeted therapeutic approaches could hold promise for the treatment of AD and other neurodegenerative disorders, including tauopathies. In this article, we will discuss the pathophysiology of AD and FTD, the proposed hypotheses, and current therapeutic approaches, highlighting the development of novel drug candidates and the progress of clinical trials in this field of research.
Collapse
Affiliation(s)
- Sally Kelliny
- Health and Biomedical Innovation, Clinical and Health SciencesUniversity of South AustraliaAdelaideSouth AustraliaAustralia
- Faculty of PharmacyAssiut UniversityAssiutEgypt
| | - Xin‐Fu Zhou
- Health and Biomedical Innovation, Clinical and Health SciencesUniversity of South AustraliaAdelaideSouth AustraliaAustralia
| | - Larisa Bobrovskaya
- Health and Biomedical Innovation, Clinical and Health SciencesUniversity of South AustraliaAdelaideSouth AustraliaAustralia
| |
Collapse
|
6
|
Zhang Q, Li C, Yin B, Yan J, Gu Y, Huang Y, Chen J, Lao X, Hao J, Yi C, Zhou Y, Cheung JCW, Wong SHD, Yang M. A biomimetic upconversion nanoreactors for near-infrared driven H 2 release to inhibit tauopathy in Alzheimer's disease therapy. Bioact Mater 2024; 42:165-177. [PMID: 39280581 PMCID: PMC11402069 DOI: 10.1016/j.bioactmat.2024.08.029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Revised: 08/13/2024] [Accepted: 08/26/2024] [Indexed: 09/18/2024] Open
Abstract
Abnormal hyperphosphorylation of tau protein is a principal pathological hallmark in the onset of neurodegenerative disorders, such as Alzheimer's disease (AD), which can be induced by an excess of reactive oxygen species (ROS). As an antioxidant, hydrogen gas (H2) has the potential to mitigate AD by scavenging highly harmful ROS such as •OH. However, conventional administration methods of H2 face significant challenges in controlling H2 release on demand and fail to achieve effective accumulation at lesion sites. Herein, we report artificial nanoreactors that mimic natural photosynthesis to realize near-infrared (NIR) light-driven photocatalytic H2 evolution in situ. The nanoreactors are constructed by biocompatible crosslinked vesicles (CVs) encapsulating ascorbic acid and two photosensitizers, chlorophyll a (Chla) and indoline dye (Ind). In addition, platinum nanoparticles (Pt NPs) serve as photocatalysts and upconversion nanoparticles (UCNP) act as light-harvesting antennas in the nanoreacting system, and both attach to the surface of CVs. Under NIR irradiation, the nanoreactors release H2 in situ to scavenge local excess ROS and attenuate tau hyperphosphorylation in the AD mice model. Such NIR-triggered nanoreactors provide a proof-of-concept design for the great potential of hydrogen therapy against AD.
Collapse
Affiliation(s)
- Qin Zhang
- Department of Biomedical Engineering, The Hong Kong Polytechnic University, 999077, Hong Kong, China
- The Hong Kong Polytechnic University Shenzhen Research Institute, Shenzhen, 518000, China
| | - Chuanqi Li
- Department of Biomedical Engineering, The Hong Kong Polytechnic University, 999077, Hong Kong, China
- The Hong Kong Polytechnic University Shenzhen Research Institute, Shenzhen, 518000, China
| | - Bohan Yin
- School of Medicine and Pharmacy, Ocean University of China, Qingdao, 266003, China
- Laboratory for Marine Drugs and Bioproducts, Qingdao Marine Science and Technology Center, Qingdao, 266237, China
- Department of Biomedical Engineering, The Hong Kong Polytechnic University, 999077, Hong Kong, China
| | - Jiaxiang Yan
- Department of Biomedical Engineering, The Hong Kong Polytechnic University, 999077, Hong Kong, China
| | - Yutian Gu
- Department of Biomedical Engineering, The Hong Kong Polytechnic University, 999077, Hong Kong, China
| | - Yingying Huang
- Department of Biomedical Engineering, The Hong Kong Polytechnic University, 999077, Hong Kong, China
| | - Jiareng Chen
- Department of Biomedical Engineering, The Hong Kong Polytechnic University, 999077, Hong Kong, China
| | - Xinyue Lao
- Department of Applied Physics, The Hong Kong Polytechnic University, 999077, Hong Kong, China
| | - Jianhua Hao
- Department of Applied Physics, The Hong Kong Polytechnic University, 999077, Hong Kong, China
| | - Changqing Yi
- Key Laboratory of Sensing Technology and Biomedical Instruments (Guangdong Province), School of Biomedical Engineering, Sun Yat-Sen University, Guangzhou, 510000, China
| | - Yi Zhou
- Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080, China
| | - James Chung Wai Cheung
- Department of Biomedical Engineering, The Hong Kong Polytechnic University, 999077, Hong Kong, China
| | - Siu Hong Dexter Wong
- School of Medicine and Pharmacy, Ocean University of China, Qingdao, 266003, China
- Laboratory for Marine Drugs and Bioproducts, Qingdao Marine Science and Technology Center, Qingdao, 266237, China
- Department of Biomedical Engineering, The Hong Kong Polytechnic University, 999077, Hong Kong, China
| | - Mo Yang
- Department of Biomedical Engineering, The Hong Kong Polytechnic University, 999077, Hong Kong, China
- The Hong Kong Polytechnic University Shenzhen Research Institute, Shenzhen, 518000, China
- Research Center for Nanoscience and Nanotechnology, The Hong Kong Polytechnic University, Kowloon, 999077, Hong Kong, China
- Joint Research Center of Biosensing and Precision Theranostics, The Hong Kong Polytechnic University, Kowloon, 999077, Hong Kong, China
| |
Collapse
|
7
|
Singh K, Murti Y, Sharma AK, Dharamveer, Ved A, Yadav S, Kulshreshtha M. Pharmacological investigation of Achras sapota against scopolamine induce amnesia and cognitive impairment in laboratory animals. Toxicol Rep 2024; 13:101813. [PMID: 39669746 PMCID: PMC11635711 DOI: 10.1016/j.toxrep.2024.101813] [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: 08/01/2024] [Revised: 11/11/2024] [Accepted: 11/12/2024] [Indexed: 12/14/2024] Open
Abstract
The present study was undertaken to investigate the effect of Achras sapota (A. sapota) fruits in scopolamine induced amnesia & cognitive impairment in mice. A. sapota commonly known as Chiku belong to Sapotaceae family. Memory impairment was induced in Swiss albino mice by a single injection of scopolamine (1 mg/kg, i.p). Animals (Swiss albino mice) were divided into five separate groups of six animals each. Positive control group received CMC (carboxy methyl cellulose) as vehicle, negative control group received scopolamine along with vehicle, standard group received Donepezil (5 mg/kg, p.o) with scopolamine. Ethanolic extract of A. sapota (EEAS, 200 mg & 400 mg/kg, p.o) was administered to group Test 1 and Test 2 respectively along with scopolamine. Elevated plus maze (EPM), modified passive avoidance test, Morris water maze (MWM) models and locomotor activity were employed as exteroceptive behaviour models to assess learning and memory activity. Thereafter lipid peroxidation, reduced glutathione and catalase level were estimated in homogenized brain of mice. The extract showed the presence of different chemical constituents like flavonoids, tannins, glycosides and alkaloids. The pre-treatment of mice with EEAS (200 mg/kg & 400 mg/kg) significantly reduced the scopolamine induced increase in EL time in MWM, whereas in EPM administration of extract produces significant decrease in TL. In Modified passive avoidance test significant increase in SDL, was shown by the animals. In locomotor activity, treatment of EEAS did not alter normal locomotor activity whereas lipid peroxidation was significantly decreased, catalase & reduced glutathione levels were significantly increased in animals of test 1 & test 2 when compared to negative control group. Hence it would be worthwhile to explore the potential of this plant in management of cognitive impairment and other memory disorders.
Collapse
Affiliation(s)
| | - Yogesh Murti
- Institute of Pharmaceutical Research, G.L.A. University, Mathura, India
| | - Anuj Kumar Sharma
- Faculty of Pharmacy, Dr. A.P.J. Abdul Kalam University, Lucknow, Uttar Pradesh, India
| | - Dharamveer
- Ambekeshwar Institute of Pharmaceutical Sciences, Lucknow, India
| | - Akash Ved
- Faculty of Pharmacy, Dr. A.P.J. Abdul Kalam University, Lucknow, Uttar Pradesh, India
| | | | | |
Collapse
|
8
|
Baluchnejadmojarad T, Roghani M. Exploring the molecular mechanisms underlying neuroprotective effect of ellagic acid in okadaic acid-induced Alzheimer's phenotype. Metab Brain Dis 2024; 39:1417-1432. [PMID: 39133454 DOI: 10.1007/s11011-024-01405-9] [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: 01/25/2024] [Accepted: 08/05/2024] [Indexed: 08/13/2024]
Abstract
Pomegranate polyphenol ellagic acid has medicinal potential in neurodegenerative disorders. The advantageous effect of this polyphenol in improving cognition in okadaic acid (OA)-instigated murine model with unraveling some modes of its action was assessed. Rats received ICV okadaic acid (OA) and post-treated with oral ellagic acid for 3 weeks (25 and 100 mg/kg/day). Cognition was analyzed in behavioral tasks besides assessment of oxidative, apoptotic, and inflammatory factors in addition to hippocampal histochemical analysis. Ellagic acid at a dose of 100 mg/kg properly attenuated cognitive abnormalities in novel object recognition (NOR), Y maze, and Barnes maze tests. Additionally, ellagic acid diminished hippocampal changes of malondialdehyde (MDA), protein carbonyl, reactive oxygen species (ROS), glutathione (GSH), glutathione peroxidase, superoxide dismutase (SOD), apoptotic factors caspases 1 and 3, tumor necrosis factor α (TNFα), and acetylcholinesterase (AChE) and beta secretase 1 (BACE 1) besides reversal of AMP-activated protein kinase (AMPK) and hyperphosphorylated tau (p-tau). Moreover, lower glial fibrillary acidic protein (GFAP) and less injury of hippocampal CA1 pyramidal neurons were observed upon ellagic acid. To conclude, neuroprotective potential of ellagic acid was shown which is somewhat attributable to its reversal of oxidative, apoptotic, and neuroinflammatory events in addition to proper regulation of AMPK and p-tau.
Collapse
Affiliation(s)
| | - Mehrdad Roghani
- Neurophysiology Research Center, Shahed University, Tehran, Iran
| |
Collapse
|
9
|
Fan L, Zhang Z. Therapeutic potential of curcumin on the cognitive decline in animal models of Alzheimer's disease: a systematic review and meta-analysis. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2024; 397:4499-4509. [PMID: 38265680 DOI: 10.1007/s00210-024-02946-7] [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: 08/11/2023] [Accepted: 01/09/2024] [Indexed: 01/25/2024]
Abstract
Curcumin, a polyphenol derived from the herb turmeric, has emerged as a prospective potential therapy in the treatment of Alzheimer's disease (AD). However, the efficacy of curcumin treatment in improving cognitive decline caused controversy recently. We aimed to systematically review the effect of curcumin on cognitive impairment in an animal model of AD. We conducted an exhaustive database search of related studies. Two investigators identified studies and independently extracted data. Stratified meta-analyses and meta-regression analyses were carried out to explore the sources of heterogeneity. Publication bias was assessed using funnel plots and Egger's test. Our systematic review included 33 articles. A meta-analysis of 29 publications showed that curcumin exerts significant positive effects on cognitive performance. For acquisition, the global estimated effect of curcumin was - 2.027 (95% CI - 2.435 to - 1.619, p < 0.001); for retention, the global estimated effect of curcumin was 1.606 (95% CI 1.101 to 2.111, p < 0.001). The stratified meta-analysis demonstrated that an increased effect size depended on diverse study characteristics. Additionally, publication bias was detected. We conclude that curcumin may reduce cognitive deficits in experimental AD. Furthermore, we emphasize that additional well-designed and well-reported animal studies are needed to inform further clinical studies.
Collapse
Affiliation(s)
- Longmin Fan
- Department of Integrated Traditional Chinese & Western Medicine, The Second Xiangya Hospital, Central South University, No. 139 Middle Renmin Road, Changsha, 410011, Hunan, China
| | - Zheyu Zhang
- Department of Integrated Traditional Chinese & Western Medicine, The Second Xiangya Hospital, Central South University, No. 139 Middle Renmin Road, Changsha, 410011, Hunan, China.
| |
Collapse
|
10
|
Dubey R, Sathiyanarayanan L, Sankaran S, Arulmozhi S. Nootropic effect of Indian Royal Jelly against okadaic acid induced rat model of Alzheimer's disease: Inhibition of neuroinflammation and acetylcholineesterase. J Tradit Complement Med 2024; 14:300-311. [PMID: 38707922 PMCID: PMC11068996 DOI: 10.1016/j.jtcme.2023.11.005] [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: 12/06/2022] [Revised: 10/21/2023] [Accepted: 11/11/2023] [Indexed: 05/07/2024] Open
Abstract
Background Royal jelly is an anti-inflammatory, antioxidant, and neuroprotective bee product. There are several sources for royal jelly and one of them is Indian Royal Jelly (IRJ). However, the neuroprotective actions of IRJ and the underlying molecular mechanisms involved are not well known. Objective To evaluate the neuroprotective effect of IRJ in the okadaic acid (OKA)-induced Alzheimer's disease (AD) model in rats. Methods In male Wistar rats, OKA was intracerebroventricularly (ICV) administered, and from day 7, they were treated orally with IRJ or memantine for 21 days. Spatial and recognition learning and memory were evaluated from days 27-34; employing the Morris water maze (MWM) and the novel object recognition tests (NORT), respectively. In vitro biochemical measurements were taken of the cholinergic system and oxidative stress markers. In silico docking was used to find the role of tau protein kinase and phosphatase in the pharmacological action. Results In OKA-induced rats, IRJ decreased the escape latency and path length in MWM and increased the exploration time for novel objects and the discrimination index in NORT. ICV-OKA rats had higher free radicals and cytokines that caused inflammation and their level of free radical scavengers was back to normal with IRJ treatment. IRJ increased the level of acetylcholine and inhibited acetylcholinesterase. Moreover, the in silico docking study revealed the strong binding affinity of 10-hydroxy-2-decenoic acid (10-HDA), a bioactive constituent of IR, to the tau protein kinases and phosphatases. Conclusion IRJ may serve as a nootropic agent in the treatment of dementia, and owing to its capacity to prevent oxidative stress and neuroinflammation, and increase cholinergic tone; it has the potential to be explored as a novel strategy for the treatment of dementia and AD. More studies may be needed to develop 10-HDA as a novel drug entity for AD.
Collapse
Affiliation(s)
- Rahul Dubey
- Department of Pharmaceutical Chemistry, Poona College of Pharmacy, Bharati Vidyapeeth (Deemed to be University), Pune, Maharashtra, India
- Department of Pharmacy, Government Polytechnic, Ratnagiri, Maharashtra, India
| | - L. Sathiyanarayanan
- Department of Pharmaceutical Chemistry, Poona College of Pharmacy, Bharati Vidyapeeth (Deemed to be University), Pune, Maharashtra, India
| | - Sandeep Sankaran
- Department of Quality Assurance Techniques, Poona College of Pharmacy, Bharati Vidyapeeth (Deemed to be University), Pune, Maharashtra, India
| | - S. Arulmozhi
- Department of Pharmacology, Poona College of Pharmacy, Bharati Vidyapeeth (Deemed to be University), Pune, Maharashtra, India
| |
Collapse
|
11
|
Nazari-Serenjeh M, Baluchnejadmojarad T, Hatami-Morassa M, Fahanik-Babaei J, Mehrabi S, Tashakori-Miyanroudi M, Ramazi S, Mohamadi-Zarch SM, Nourabadi D, Roghani M. Kolaviron neuroprotective effect against okadaic acid-provoked cognitive impairment. Heliyon 2024; 10:e25564. [PMID: 38356522 PMCID: PMC10864987 DOI: 10.1016/j.heliyon.2024.e25564] [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: 06/05/2023] [Revised: 12/25/2023] [Accepted: 01/29/2024] [Indexed: 02/16/2024] Open
Abstract
Alzheimer's disease (AD) is acknowledged as the main causative factor of dementia that affects millions of people around the world and is increasing at increasing pace. Okadaic acid (OA) is a toxic compound with ability to inhibit protein phosphatases and to induce tau protein hyperphosphorylation and Alzheimer's-like phenotype. Kolaviron (KV) is a bioflavonoid derived from Garcinia kola seeds with anti-antioxidative and anti-inflammation properties. The main goal of this study was to assess whether kolaviron can exert neuroprotective effect against okadaic acid-induced cognitive deficit. Rats had an intracerebroventricular (ICV) injection of OA and pretreated with KV at 50 or 100 mg/kg and examined for cognition besides histological and biochemical factors. OA group treated with KV at 100 mg/kg had less memory deficit in passive avoidance and novel object discrimination (NOD) tasks besides lower hippocampal levels of caspases 1 and 3, tumor necrosis factor α (TNFα) and interleukin 6 (IL-6) as inflammatory factors, reactive oxygen species (ROS), protein carbonyl, malondialdehyde (MDA), and phosphorylated tau (p-tau) and higher level of acetylcholinesterase (AChE) activity, mitochondrial integrity index, superoxide dismutase (SOD), and glutathione (GSH). Moreover, KV pretreatment at 100 mg/kg attenuated hippocampal CA1 neuronal loss and glial fibrillary acidic protein (GFAP) reactivity as a factor of astrogliosis. In summary, KV was able to attenuate cognitive fall subsequent to ICV OA which is partly mediated through its neuroprotective potential linked to mitigation of tau hyperphosphorylation, apoptosis, pyroptosis, neuroinflammation, and oxidative stress and also improvement of mitochondrial health.
Collapse
Affiliation(s)
- Morteza Nazari-Serenjeh
- Student Research Committee, Iran University of Medical Sciences, Tehran, Iran
- Department of Physiology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | | | - Masoud Hatami-Morassa
- Department of Physiology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Javad Fahanik-Babaei
- Electrophysiology Research Center, Neuroscience Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Soraya Mehrabi
- Department of Neuroscience, Faculty of Advanced Technologies in Medicine, Iran University of Medical Science, Tehran, Iran
- Cellular and Molecular Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Mahsa Tashakori-Miyanroudi
- Psychiatry and Behavioral Sciences Research Center, Addiction Institute, Mazandaran University of Medical Sciences, Sari, Iran
| | - Samira Ramazi
- Student Research Committee, Iran University of Medical Sciences, Tehran, Iran
| | - Seyed-Mahdi Mohamadi-Zarch
- Department of Physiology, Faculty of Medicine, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Davood Nourabadi
- Department of Physiology, Faculty of Medicine, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Mehrdad Roghani
- Neurophysiology Research Center, Shahed University, Tehran, Iran
| |
Collapse
|
12
|
Rey F, Berardo C, Maghraby E, Mauri A, Messa L, Esposito L, Casili G, Ottolenghi S, Bonaventura E, Cuzzocrea S, Zuccotti G, Tonduti D, Esposito E, Paterniti I, Cereda C, Carelli S. Redox Imbalance in Neurological Disorders in Adults and Children. Antioxidants (Basel) 2023; 12:antiox12040965. [PMID: 37107340 PMCID: PMC10135575 DOI: 10.3390/antiox12040965] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2023] [Revised: 04/03/2023] [Accepted: 04/14/2023] [Indexed: 04/29/2023] Open
Abstract
Oxygen is a central molecule for numerous metabolic and cytophysiological processes, and, indeed, its imbalance can lead to numerous pathological consequences. In the human body, the brain is an aerobic organ and for this reason, it is very sensitive to oxygen equilibrium. The consequences of oxygen imbalance are especially devastating when occurring in this organ. Indeed, oxygen imbalance can lead to hypoxia, hyperoxia, protein misfolding, mitochondria dysfunction, alterations in heme metabolism and neuroinflammation. Consequently, these dysfunctions can cause numerous neurological alterations, both in the pediatric life and in the adult ages. These disorders share numerous common pathways, most of which are consequent to redox imbalance. In this review, we will focus on the dysfunctions present in neurodegenerative disorders (specifically Alzheimer's disease, Parkinson's disease and amyotrophic lateral sclerosis) and pediatric neurological disorders (X-adrenoleukodystrophies, spinal muscular atrophy, mucopolysaccharidoses and Pelizaeus-Merzbacher Disease), highlighting their underlining dysfunction in redox and identifying potential therapeutic strategies.
Collapse
Affiliation(s)
- Federica Rey
- Pediatric Clinical Research Center "Romeo ed Enrica Invernizzi", Department of Biomedical and Clinical Sciences, University of Milano, 20157 Milano, Italy
- Center of Functional Genomics and Rare diseases, Department of Pediatrics, Buzzi Children's Hospital, 20154 Milano, Italy
| | - Clarissa Berardo
- Pediatric Clinical Research Center "Romeo ed Enrica Invernizzi", Department of Biomedical and Clinical Sciences, University of Milano, 20157 Milano, Italy
- Center of Functional Genomics and Rare diseases, Department of Pediatrics, Buzzi Children's Hospital, 20154 Milano, Italy
| | - Erika Maghraby
- Pediatric Clinical Research Center "Romeo ed Enrica Invernizzi", Department of Biomedical and Clinical Sciences, University of Milano, 20157 Milano, Italy
- Department of Biology and Biotechnology "L. Spallanzani", University of Pavia, 27100 Pavia, Italy
| | - Alessia Mauri
- Pediatric Clinical Research Center "Romeo ed Enrica Invernizzi", Department of Biomedical and Clinical Sciences, University of Milano, 20157 Milano, Italy
- Center of Functional Genomics and Rare diseases, Department of Pediatrics, Buzzi Children's Hospital, 20154 Milano, Italy
| | - Letizia Messa
- Center of Functional Genomics and Rare diseases, Department of Pediatrics, Buzzi Children's Hospital, 20154 Milano, Italy
- Department of Electronics, Information and Bioengineering (DEIB), Politecnico di Milano, 20133 Milano, Italy
| | - Letizia Esposito
- Pediatric Clinical Research Center "Romeo ed Enrica Invernizzi", Department of Biomedical and Clinical Sciences, University of Milano, 20157 Milano, Italy
- Center of Functional Genomics and Rare diseases, Department of Pediatrics, Buzzi Children's Hospital, 20154 Milano, Italy
| | - Giovanna Casili
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, 98166 Messina, Italy
| | - Sara Ottolenghi
- Department of Medicine and Surgery, University of Milano Bicocca, 20126 Milano, Italy
| | - Eleonora Bonaventura
- Child Neurology Unit, Buzzi Children's Hospital, 20154 Milano, Italy
- Center for Diagnosis and Treatment of Leukodystrophies and Genetic Leukoencephalopathies (COALA), Buzzi Children's Hospital, 20154 Milano, Italy
| | - Salvatore Cuzzocrea
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, 98166 Messina, Italy
| | - Gianvincenzo Zuccotti
- Pediatric Clinical Research Center "Romeo ed Enrica Invernizzi", Department of Biomedical and Clinical Sciences, University of Milano, 20157 Milano, Italy
- Department of Pediatrics, Buzzi Children's Hospital, 20154 Milano, Italy
| | - Davide Tonduti
- Pediatric Clinical Research Center "Romeo ed Enrica Invernizzi", Department of Biomedical and Clinical Sciences, University of Milano, 20157 Milano, Italy
- Child Neurology Unit, Buzzi Children's Hospital, 20154 Milano, Italy
- Center for Diagnosis and Treatment of Leukodystrophies and Genetic Leukoencephalopathies (COALA), Buzzi Children's Hospital, 20154 Milano, Italy
| | - Emanuela Esposito
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, 98166 Messina, Italy
| | - Irene Paterniti
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, 98166 Messina, Italy
| | - Cristina Cereda
- Center of Functional Genomics and Rare diseases, Department of Pediatrics, Buzzi Children's Hospital, 20154 Milano, Italy
| | - Stephana Carelli
- Pediatric Clinical Research Center "Romeo ed Enrica Invernizzi", Department of Biomedical and Clinical Sciences, University of Milano, 20157 Milano, Italy
- Center of Functional Genomics and Rare diseases, Department of Pediatrics, Buzzi Children's Hospital, 20154 Milano, Italy
| |
Collapse
|
13
|
Liu X, Lai LY, Chen JX, Li X, Wang N, Zhou LJ, Jiang XW, Hu XL, Liu WW, Jiao XM, Qi ZT, Liu WJ, Wu LM, Huang YG, Xu ZH, Zhao QC. An inhibitor with GSK3β and DYRK1A dual inhibitory properties reduces Tau hyperphosphorylation and ameliorates disease in models of Alzheimer's disease. Neuropharmacology 2023; 232:109525. [PMID: 37004752 DOI: 10.1016/j.neuropharm.2023.109525] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Revised: 02/28/2023] [Accepted: 03/23/2023] [Indexed: 04/03/2023]
Abstract
Since Alzheimer's disease (AD) is a complex and multifactorial neuropathology, the discovery of multi-targeted inhibitors has gradually demonstrated greater therapeutic potential. Neurofibrillary tangles (NFTs), the main neuropathologic hallmarks of AD, are mainly associated with hyperphosphorylation of the microtubule-associated protein Tau. The overexpression of GSK3β and DYRK1A has been recognized as an important contributor to hyperphosphorylation of Tau, leading to the strategy of using dual-targets inhibitors for the treatment of this disorder. ZDWX-12 and ZDWX-25, as harmine derivatives, were found good inhibition on dual targets in our previous study. Here, we firstly evaluated the inhibition effect of Tau hyperphosphorylation using two compounds by HEK293-Tau P301L cell-based model and okadaic acid (OKA)-induced mouse model. We found that ZDWX-25 was more effective than ZDWX-12. Then, based on comprehensively investigations on ZDWX-25 in vitro and in vivo, 1) the capability of ZDWX-25 to show a reduction in phosphorylation of multiple Tau epitopes in OKA-induced neurodegeneration cell models, and 2) the effect of reduction on NFTs by 3xTg-AD mouse model under administration of ZDWX-25, an orally bioavailable, brain-penetrant dual-targets inhibitor with low toxicity. Our data highlight that ZDWX-25 is a promising drug for treating AD.
Collapse
|
14
|
Zhang Q, Yin B, Huang Y, Gu Y, Yan J, Chen J, Li C, Zhang Y, Wong SHD, Yang M. A dual “turn-on” biosensor based on AIE effect and FRET for in situ detection of miR-125b biomarker in early Alzheimer's disease. Biosens Bioelectron 2023; 230:115270. [PMID: 37023551 DOI: 10.1016/j.bios.2023.115270] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Revised: 03/22/2023] [Accepted: 03/26/2023] [Indexed: 04/03/2023]
Abstract
MicroRNA-125b (miR-125b) is highly associated with synaptic dysfunction and tau hyperphosphorylation in the early pathogenesis of Alzheimer's disease (AD), making it a promising biomarker for early AD diagnosis. Hence, there is an urgent need for a reliable sensing platform to assist in situ miR-125b detection. In this work, we report a dual "turn-on" fluorescence biosensor based on the nanocomposite of aggregation-induced emission fluorogen (AIEgen)-labeled oligonucleotide (TPET-DNA) probes immobilized on the surface of cationic dextran modified molybdenum disulfide (TPET-DNA@Dex-MoS2). In the presence of the target, TEPT-DNA can hybridize with miR-125b to form a DNA/RNA duplex, causing TPET-DNA to detach from the surface of Dex-MoS2 that simultaneously activates the dual fluorescence enhancement processes: (1) recovery of TPET-DNA signal and (2) strong fluorescent emission from AIEgen triggered by restriction of the intramolecular rotation. The sensing performance of TPET-DNA@Dex-MoS2 was demonstrated by detecting miR-125b in vitro with good sensitivity at the picomolar level and rapid response (≤1 h) without amplification procedures. Furthermore, our nanoprobes exhibited excellent imaging capabilities to aid real-time monitoring of the endogenous miR-125b in PC12 cells and brain tissues of mice AD model induced by local administration of okadaic acid (OA). The fluorescence signals of the nanoprobes indicated miR-125b was spatially associated with phosphorylated tau protein (p-tau) in vitro and in vivo. Therefore, TPET-DNA@Dex-MoS2 could be a promising tool for in situ and real-time monitoring of the AD-related microRNAs and also provide mechanistic insight into the early prognosis of AD.
Collapse
|
15
|
Liu A, Hu J, Yeh TS, Wang C, Tang J, Huang X, Chen B, Huangfu L, Yu W, Zhang L. Neuroprotective Strategies for Stroke by Natural Products: Advances and Perspectives. Curr Neuropharmacol 2023; 21:2283-2309. [PMID: 37458258 PMCID: PMC10556387 DOI: 10.2174/1570159x21666230717144752] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2022] [Revised: 12/21/2022] [Accepted: 12/23/2022] [Indexed: 09/09/2023] Open
Abstract
Cerebral ischemic stroke is a disease with high prevalence and incidence. Its management focuses on rapid reperfusion with intravenous thrombolysis and endovascular thrombectomy. Both therapeutic strategies reduce disability, but the therapy time window is short, and the risk of bleeding is high. Natural products (NPs) have played a key role in drug discovery, especially for cancer and infectious diseases. However, they have made little progress in clinical translation and pose challenges to the treatment of stroke. Recently, with the investigation of precise mechanisms in cerebral ischemic stroke and the technological development of NP-based drug discovery, NPs are addressing these challenges and opening up new opportunities in cerebral stroke. Thus, in this review, we first summarize the structure and function of diverse NPs, including flavonoids, phenols, terpenes, lactones, quinones, alkaloids, and glycosides. Then we propose the comprehensive neuroprotective mechanism of NPs in cerebral ischemic stroke, which involves complex cascade processes of oxidative stress, mitochondrial damage, apoptosis or ferroptosis-related cell death, inflammatory response, and disruption of the blood-brain barrier (BBB). Overall, we stress the neuroprotective effect of NPs and their mechanism on cerebral ischemic stroke for a better understanding of the advances and perspective in NPs application that may provide a rationale for the development of innovative therapeutic regimens in ischemic stroke.
Collapse
Affiliation(s)
- Aifen Liu
- Institute of Interdisciplinary Integrative Medicine Research, School of Medicine, Nantong University, Nantong 226001, China
| | - Jingyan Hu
- Institute of Interdisciplinary Integrative Medicine Research, School of Medicine, Nantong University, Nantong 226001, China
| | - Tzu-Shao Yeh
- Institute of Interdisciplinary Integrative Medicine Research, School of Medicine, Nantong University, Nantong 226001, China
- Department of Nutrition and Food Hygiene, School of Public Health, Nantong University, Nantong 226019, China
| | - Chengniu Wang
- Institute of Interdisciplinary Integrative Medicine Research, School of Medicine, Nantong University, Nantong 226001, China
| | - Jilong Tang
- Institute of Interdisciplinary Integrative Medicine Research, School of Medicine, Nantong University, Nantong 226001, China
| | - Xiaohong Huang
- Institute of Interdisciplinary Integrative Medicine Research, School of Medicine, Nantong University, Nantong 226001, China
| | - Bin Chen
- Institute of Interdisciplinary Integrative Medicine Research, School of Medicine, Nantong University, Nantong 226001, China
| | - Liexiang Huangfu
- Institute of Interdisciplinary Integrative Medicine Research, School of Medicine, Nantong University, Nantong 226001, China
| | - Weili Yu
- Institute of Interdisciplinary Integrative Medicine Research, School of Medicine, Nantong University, Nantong 226001, China
| | - Lei Zhang
- Institute of Interdisciplinary Integrative Medicine Research, School of Medicine, Nantong University, Nantong 226001, China
- Department of Pharmaceutical Botany, School of Pharmacy, Naval Medical University, Shanghai 200433, China
| |
Collapse
|
16
|
Curcumin-Piperlongumine Hybrids with a Multitarget Profile Elicit Neuroprotection in In Vitro Models of Oxidative Stress and Hyperphosphorylation. Antioxidants (Basel) 2021; 11:antiox11010028. [PMID: 35052532 PMCID: PMC8773050 DOI: 10.3390/antiox11010028] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2021] [Revised: 12/17/2021] [Accepted: 12/21/2021] [Indexed: 11/17/2022] Open
Abstract
Curcumin shows a broad spectrum of activities of relevance in the treatment of Alzheimer’s disease (AD); however, it is poorly absorbed and is also chemically and metabolically unstable, leading to a very low oral bioavailability. A small library of hybrid compounds designed as curcumin analogues and incorporating the key structural fragment of piperlongumine, a natural neuroinflammation inhibitor, were synthesized by a two-step route that combines a three-component reaction between primary amines, β-ketoesters and α-haloesters and a base-promoted acylation with cinnamoyl chlorides. These compounds were predicted to have good oral absorption and CNS permeation, had good scavenging properties in the in vitro DPPH experiment and in a cellular assay based on the oxidation of dichlorofluorescin to a fluorescent species. The compounds showed low toxicity in two cellular models, were potent inductors of the Nrf2-ARE phase II antioxidant response, inhibited PHF6 peptide aggregation, closely related to Tau protein aggregation and were active against the LPS-induced inflammatory response. They also afforded neuroprotection against an oxidative insult induced by inhibition of the mitochondrial respiratory chain with the rotenone-oligomycin A combination and against Tau hyperphosphorylation induced by the phosphatase inhibitor okadaic acid. This multitarget pharmacological profile is highly promising in the development of treatments for AD and provides a good hit structure for future optimization efforts.
Collapse
|
17
|
Preclinical validation of a novel oral Edaravone formulation for treatment of frontotemporal dementia. Neurotox Res 2021; 39:1689-1707. [PMID: 34599751 DOI: 10.1007/s12640-021-00405-2] [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: 05/10/2021] [Revised: 08/25/2021] [Accepted: 08/27/2021] [Indexed: 10/20/2022]
Abstract
Oxidative stress is a key factor in the pathogenesis of several neurodegenerative disorders and is involved in the accumulation of amyloid beta plaques and Tau inclusions. Edaravone (EDR) is a free radical scavenger that is approved for motor neuron disease and acute ischemic stroke. EDR alleviates pathologies and cognitive impairment of AD via targeting multiple key pathways in transgenic mice. Herein, we aimed to study the effect of EDR on Tau pathology in P301L mice; an animal model of frontotemporal dementia (FTD), at two age time points representing the early and late stages of the disease. A novel EDR formulation was utilized in the study and the drug was delivered orally in drinking water for 3 months. Then, behavioral tests were conducted followed by animal sacrifice and brain dissection. Treatment with EDR improved the reference memory and accuracy in the probe trial as evaluated in Morris water maze, as well as novel object recognition and significantly alleviated motor deficits in these mice. EDR also reduced the levels of 4-hydroxy-2-nonenal and 3-nitrotyrosine adducts. In addition, immunohistochemistry showed that EDR reduced tau phosphorylation and neuroinflammation and partially rescued neurons against oxidative neurotoxicity. Moreover, EDR attenuated downstream pathologies involved in Tau hyperphosphorylation. These results suggest that EDR may be a potential therapeutic agent for the treatment of FTD.
Collapse
|
18
|
Therapeutic Potential of Curcumin in Reversing the Depression and Associated Pseudodementia via Modulating Stress Hormone, Hippocampal Neurotransmitters, and BDNF Levels in Rats. Neurochem Res 2021; 46:3273-3285. [PMID: 34409523 DOI: 10.1007/s11064-021-03430-x] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2021] [Revised: 08/06/2021] [Accepted: 08/13/2021] [Indexed: 01/07/2023]
Abstract
Depressive state adversely affects the memory functions, especially in the geriatric population. The initial stage of memory deficits associated with depression is particularly called as pseudodementia. It is the starting point of memory disturbance before dementia. The purpose of this research was to study depression and its consequent pseudodementia. For this purpose 24 male albino Wistar rats were divided into four groups. Depression was induced by 14 days of chronic restraint stress (CRS) daily for 4 h. After developing a depression model, pattern separation test was conducted to monitor pseudodementia in rats. Morris water maze test (MWM) was also performed to observe spatial memory. It was observed that model animals displayed impaired pattern separation and spatial memory. Treatment was started after the development of pseudodementia in rats. Curcumin at a dose of 200 mg/kg was given to model rats for one week along with the stress procedure. Following the treatment with curcumin, rats were again subjected to the aforementioned behavioral tests before decapitation. Corticosterone levels, brain derived neurotrophic factor (BDNF) and neurochemical analysis were conducted. Model rats showed depressogenic behavior and impaired memory performance. In addition to this, high corticosterone levels and decreased hippocampal BDNF, 5-HT, dopamine (DA), and acetylcholine (ACh) levels were also observed in depressed animals. These behavioral biochemical and neurochemical changes were effectively restored following treatment with curcumin. Hence, it is suggested from this study that pseudodementia can be reversed unlike true dementia by controlling the factors such as depression which induce memory impairment.
Collapse
|
19
|
Liang Z, Currais A, Soriano-Castell D, Schubert D, Maher P. Natural products targeting mitochondria: emerging therapeutics for age-associated neurological disorders. Pharmacol Ther 2021; 221:107749. [PMID: 33227325 PMCID: PMC8084865 DOI: 10.1016/j.pharmthera.2020.107749] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2020] [Revised: 11/16/2020] [Accepted: 11/17/2020] [Indexed: 12/17/2022]
Abstract
Mitochondria are the primary source of energy production in the brain thereby supporting most of its activity. However, mitochondria become inefficient and dysfunctional with age and to a greater extent in neurological disorders. Thus, mitochondria represent an emerging drug target for many age-associated neurological disorders. This review summarizes recent advances (covering from 2010 to May 2020) in the use of natural products from plant, animal, and microbial sources as potential neuroprotective agents to restore mitochondrial function. Natural products from diverse classes of chemical structures are discussed and organized according to their mechanism of action on mitochondria in terms of modulation of biogenesis, dynamics, bioenergetics, calcium homeostasis, and membrane potential, as well as inhibition of the oxytosis/ferroptosis pathway. This analysis emphasizes the significant value of natural products for mitochondrial pharmacology as well as the opportunities and challenges for the discovery and development of future neurotherapeutics.
Collapse
Affiliation(s)
- Zhibin Liang
- Cellular Neurobiology Laboratory, The Salk Institute for Biological Studies, 10010 North Torrey Pines Road, La Jolla, California 92037, United States; The Paul F. Glenn Center for Biology of Aging Research, The Salk Institute for Biological Studies, 10010 North Torrey Pines Road, La Jolla, California 92037, United States.
| | - Antonio Currais
- Cellular Neurobiology Laboratory, The Salk Institute for Biological Studies, 10010 North Torrey Pines Road, La Jolla, California 92037, United States
| | - David Soriano-Castell
- Cellular Neurobiology Laboratory, The Salk Institute for Biological Studies, 10010 North Torrey Pines Road, La Jolla, California 92037, United States
| | - David Schubert
- Cellular Neurobiology Laboratory, The Salk Institute for Biological Studies, 10010 North Torrey Pines Road, La Jolla, California 92037, United States; The Paul F. Glenn Center for Biology of Aging Research, The Salk Institute for Biological Studies, 10010 North Torrey Pines Road, La Jolla, California 92037, United States
| | - Pamela Maher
- Cellular Neurobiology Laboratory, The Salk Institute for Biological Studies, 10010 North Torrey Pines Road, La Jolla, California 92037, United States.
| |
Collapse
|
20
|
Interferon Gamma-Mediated Oxidative Stress Induces Apoptosis, Neuroinflammation, Zinc Ion Influx, and TRPM2 Channel Activation in Neuronal Cell Line: Modulator Role of Curcumin. Inflammation 2021; 44:1878-1894. [PMID: 33866462 DOI: 10.1007/s10753-021-01465-4] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Revised: 03/30/2021] [Accepted: 04/03/2021] [Indexed: 10/21/2022]
Abstract
Host defenses in the brain are modulated by the activation of several factors such as oxygen free radical species (ROS), Ca2+ influx, and TRPM2 activation, and they are well-known adverse factors in neurotoxicity and neurodegenerative diseases. Importantly, recent data indicated a protective action of curcumin (CRC) via inhibition of TRPM2 on the inflammation factors, ROS, and apoptosis in hypoxia-induced SH-SY5Y neuronal cells. However, the relationship between interferon gamma (IFNg) exposure and TRPM2 activation in the SH-SY5Y cells are not fully identified. The SH-SY5Y cells as a neuronal cell line model were used in several neuroinflammation studies. Hence, we used the SH-SY5Y cells in the current study, and they were divided into four main groups as control, CRC, IFNg, and IFNg+CRC. The data presented here indicate that IFNg induced excessive Ca2+ influx via activation of TRPM2. The IFNg treatment further increased cell death, cell debris amount, apoptosis, and cytokine generations (IL-1β, IL-6, and TNF-α) which were due to increased cytosolic and mitochondrial ROS generations as well as increased activations of caspase-3 and caspase-9. The expression levels of TRPM2, PARP-1, Bax, caspase-3, and caspase-9 were increased in the cells by the IFNg treatment. However, CRC treatment reduced the increase of expression levels, cytokine generations, caspase activations, ROS release, Ca2+ influx, cell death, and apoptosis levels via inhibition of TRPM2 in the SH-SY5Y cells that were treated with IFNg. Moreover, the treatment of TRPM2 blockers (ACA and 2-APB) potentiated the modulator effects of CRC. In conclusion, these results suggest that neuroinflammation via IFNg lead to the TRPM2 activation in the SH-SY5Y cells, whereas CRC prevents IFNg-mediated TRPM2 activation, cell death, and cytokine generations.
Collapse
|
21
|
Mehta K, Kaur B, Pandey KK, Kaler S, Dhar P. Curcumin supplementation shows modulatory influence on functional and morphological features of hippocampus in mice subjected to arsenic trioxide exposure. Anat Cell Biol 2020; 53:355-365. [PMID: 32929054 PMCID: PMC7527119 DOI: 10.5115/acb.18.169] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2018] [Revised: 06/13/2019] [Accepted: 09/16/2019] [Indexed: 12/20/2022] Open
Abstract
Since, oxidative stress has been suggested as one of the mechanisms underlying arsenic-induced toxicity, the present study focused on the role of antioxidant (curcumin) supplementation on behavioral, biochemical, and morphological alterations with context to mice hippocampus (CA1) following arsenic trioxide (As2O3) administration. Healthy male Swiss albino mice were divided into control and experimental groups. As2O3 (2 mg/kg bw) alone or along with curcumin (100 mg/kg bw) was administered to experimental groups by oral route for 45 days whereas the control groups received either no treatment or vehicle for curcumin. Animals were subjected to behavioral study towards the end of the experimental period (day 33-45). On day 46, the brain samples were obtained and subjected either to immersion fixation (for morphometric observations) or used afresh for biochemical test. Behavioral tests (open field, elevated plus maze, and Morris water maze) revealed enhanced anxiety levels and impairment of cognitive functions in As2O3 alone treated groups whereas a trend of recovery was evident in mice simultaneously treated with As2O3 and curcumin. Morphological observations showed noticeable reduction in stratum pyramidale thickness (CA1), along with decrease in density and size of pyramidal neurons in As2O3 alone exposed group as compared to As2O3+Cu co-treated group. Hippocampal glutathione levels were found to be downregulated in animals receiving As2O3 as against the levels of controls and curcumin supplemented animals, thereby, suggestive of beneficial role of curcumin on As2O3 induced adverse effects.
Collapse
Affiliation(s)
- Kamakshi Mehta
- Department of Anatomy, All India Institute of Medical Sciences, New Delhi, India
| | - Balpreet Kaur
- Department of Anatomy, All India Institute of Medical Sciences, New Delhi, India
| | - Kamlesh Kumar Pandey
- Department of Anatomy, All India Institute of Medical Sciences, New Delhi, India
| | - Saroj Kaler
- Department of Anatomy, All India Institute of Medical Sciences, New Delhi, India
| | - Pushpa Dhar
- Department of Anatomy, All India Institute of Medical Sciences, New Delhi, India
| |
Collapse
|
22
|
Eghbaliferiz S, Farhadi F, Barreto GE, Majeed M, Sahebkar A. Effects of curcumin on neurological diseases: focus on astrocytes. Pharmacol Rep 2020; 72:769-782. [PMID: 32458309 DOI: 10.1007/s43440-020-00112-3] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2020] [Revised: 05/16/2020] [Accepted: 05/19/2020] [Indexed: 02/06/2023]
Abstract
Astrocytes are the most abundant glial cells in the central nervous system, and are important players in both brain injury and neurodegenerative disease. Curcumin (1,7-bis[4-hydroxy-3-methoxyphenyl]-1,6-heptadiene-3,5-dione), the major active component of turmeric, belongs to the curcuminoid family that was originally isolated from the plant Curcuma longa. Several studies suggest that curcumin may have a beneficial impact on the brain pathology and aging. These effects are due to curcumin's antioxidant, free-radical scavenging, and anti-inflammatory activity. In light of this, our current review aims to discuss the role of astrocytes as essential players in neurodegenerative diseases and suggest that curcumin is capable of direct inhibition of astrocyte activity with a particular focus on its effects in Alexander disease, Alzheimer's disease, ischemia stroke, spinal cord injury, Multiple sclerosis, and Parkinson's disease.
Collapse
Affiliation(s)
- Samira Eghbaliferiz
- Department of Pharmacognosy, School of Pharmacy, Birjand University of Medical Sciences, Birjand, Iran
| | - Faegheh Farhadi
- Department of Pharmacognosy, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - George E Barreto
- Department of Biological Sciences, University of Limerick, Limerick, Ireland
- Instituto de Ciencias Biomédicas, Universidad Autónoma de Chile, Santiago, Chile
| | | | - Amirhossein Sahebkar
- Halal Research Center of IRI, FDA, Tehran, Iran.
- Neurogenic Inflammation Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran.
- School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
23
|
Atabaki R, Roohbakhsh A, Moghimi A, Mehri S. Protective effects of maternal administration of curcumin and hesperidin in the rat offspring following repeated febrile seizure: Role of inflammation and TLR4. Int Immunopharmacol 2020; 86:106720. [PMID: 32585605 DOI: 10.1016/j.intimp.2020.106720] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2020] [Revised: 06/15/2020] [Accepted: 06/15/2020] [Indexed: 12/12/2022]
Abstract
Neuroinflammation has a key role in seizure generation and perpetuation in the neonatal period, and toll-like receptor 4 (TLR4) pathway has a prominent role in neuroinflammatory diseases. Administration of antioxidants and targeting TLR4 in the embryonic period may protect rat offspring against the next incidence of febrile seizure and its harmful effects. Curcumin and hesperidin are natural compounds with anti-inflammatory and antioxidant properties and have an inhibitory action on TLR4 receptors. We evaluated the effect of maternal administration of curcumin and hesperidin on infantile febrile seizure and subsequent memory dysfunction in adulthood. Hyperthermia febrile seizure was induced on postnatal days 9-11 on male rat pups with 24 h intervals, in a Plexiglas box that was heated to ~45 °C by a heat lamp. We used enzyme-linked immunosorbent assay, Western blotting, malondialdehyde (MDA), and glutathione (GSH) assessment for evaluation of inflammatory cytokine levels, TLR4 protein expression, and oxidative responses in the hippocampal tissues. For assessing working memory and long-term potentiation, the double Y-maze test and Schaffer collateral-CA1 in vivo electrophysiological recording were performed, respectively Our results showed that curcumin and hesperidin decreased TNF-α, IL-10, and TLR4 protein expression and reversed memory dysfunction. However, they did not provoke a significant effect on GSH content or amplitude and slope of recorded fEPSPs in the hippocampus. In addition, curcumin, but not hesperidin, decreased interleukin-1β (IL-1β) and MDA levels. These findings imply that curcumin and hesperidin induced significant protective effects on febrile seizures, possibly via their anti-inflammatory and antioxidant properties and downregulation of TLR4.
Collapse
Affiliation(s)
- Rabi Atabaki
- Rayan Center for Neuroscience & Behavior, Department of Biology, Faculty of Science, Ferdowsi University of Mashhad, Iran
| | - Ali Roohbakhsh
- Pharmaceutical Research Center, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran; Department of Pharmacodynamics and Toxicology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Ali Moghimi
- Rayan Center for Neuroscience & Behavior, Department of Biology, Faculty of Science, Ferdowsi University of Mashhad, Iran.
| | - Soghra Mehri
- Department of Pharmacodynamics and Toxicology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
24
|
Zhang H, Wang X, Xu P, Ji X, Chi T, Liu P, Zou L. Tolfenamic acid inhibits GSK-3β and PP2A mediated tau hyperphosphorylation in Alzheimer's disease models. J Physiol Sci 2020; 70:29. [PMID: 32517647 PMCID: PMC10717460 DOI: 10.1186/s12576-020-00757-y] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2020] [Accepted: 05/31/2020] [Indexed: 02/06/2023]
Abstract
Tolfenamic acid, a nonsteroidal anti-inflammatory drug, alleviated learning and memory deficits and decreased the expression of specificity protein 1 (SP1)-mediated cyclin-dependent kinase-5 (CDK5), a major protein kinase that regulates hyperphosphorylated tau, in Alzheimer's disease (AD) transgenic mice. However, whether tolfenamic acid can regulate the major tau protein kinase, glycogen synthase kinase-3β (GSK-3β), or tau protein phosphatase, protein phosphatase 2A (PP2A), further inhibiting hyperphosphorylation of tau, remains unknown. To this end, tolfenamic acid was administered i.p. in a GSK-3β overactivation postnatal rat model and orally in mice after intracerebroventricular (ICV) injection of okadaic acid (OA) to develop a PP2A inhibition model. We used four behavioural experiments to evaluate memory function in ICV-OA mice. In this study, tolfenamic acid attenuated memory dysfunction. Tolfenamic acid decreased the expression of hyperphosphorylated tau in the brain by inhibiting GSK-3β activity, decreasing phosphorylated PP2A (Tyr307), and enhancing PP2A activity. Tolfenamic acid also increased wortmannin (WT) and GF-109203X (GFX) induced phosphorylation of GSK-3β (Ser9) and prevented OA-induced downregulation of PP2A activity in PC12 cells. Altogether, these results show that tolfenamic acid not only decreased SP1/CDK5-mediated tau phosphorylation, but also inhibited GSK-3β and PP2A-mediated tau hyperphosphorylation in AD models.
Collapse
Affiliation(s)
- Huiming Zhang
- Department of Pharmacology, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenhe District, Shenyang, 110016, Liaoning, People's Republic of China
| | - Xiaojuan Wang
- Department of Pharmacology, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenhe District, Shenyang, 110016, Liaoning, People's Republic of China
| | - Pu Xu
- Department of Pharmacology, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenhe District, Shenyang, 110016, Liaoning, People's Republic of China
| | - Xuefei Ji
- Department of Pharmacology, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenhe District, Shenyang, 110016, Liaoning, People's Republic of China
| | - Tianyan Chi
- Department of Pharmacology, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenhe District, Shenyang, 110016, Liaoning, People's Republic of China
| | - Peng Liu
- Department of Pharmacology, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenhe District, Shenyang, 110016, Liaoning, People's Republic of China.
| | - Libo Zou
- Department of Pharmacology, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenhe District, Shenyang, 110016, Liaoning, People's Republic of China.
| |
Collapse
|
25
|
Oral glutathione administration inhibits the oxidative stress and the inflammatory responses in App NL-G-F/NL-G-F knock-in mice. Neuropharmacology 2020; 168:108026. [PMID: 32130977 DOI: 10.1016/j.neuropharm.2020.108026] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2019] [Revised: 02/22/2020] [Accepted: 02/27/2020] [Indexed: 01/27/2023]
Abstract
Alzheimer's disease (AD) is the most common neurodegenerative disease characterized by the presence of extracellular amyloid-β (Aβ) plaques and intracellular neurofibrillary tangles. Reduced antioxidants and increased oxidative stress and inflammation are responsible for the pathological features characteristic of an AD brain. We observed decreased levels of the reduced form of glutathione (GSH), the most abundant brain antioxidant, and decreased GSH/glutathione disulfide (GSSG) ratios in AppNL-G-F/NL-G-F knock-in (NL-G-F) mouse brains. Repeated oral GSH administration for 3 weeks dose-dependently increased GSH levels and restored the GSH/GSSH ratio. Consistent with the restoration of GSH levels, the levels of 4-hydroxy-2-nonenal (4-HNE), a marker of oxidative stress, were significantly decreased in the hippocampus of NL-G-F mice. Additionally, inflammatory responses, such as microgliosis and increased mRNA expression of inflammatory cytokines, were also inhibited. Moreover, behavioral deficits including cognitive decline, depressive-like behaviors, and anxiety-related behaviors observed in NL-G-F mice were significantly improved by oral and chronic GSH administration. Taken together, our data suggest that oral GSH administration is an attractive therapeutic strategy to reduce the excessive oxidative stress and inflammatory responses in the AD brain.
Collapse
|
26
|
Singh S, Dharamveer, Kulshreshtha M. Pharmacological Approach of Pistacia Vera Fruit to Assess Learning and Memory Potential in Chemically-Induced Memory Impairment in Mice. Cent Nerv Syst Agents Med Chem 2020; 19:125-132. [PMID: 30836928 DOI: 10.2174/1871524919666190304122927] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2018] [Revised: 02/20/2019] [Accepted: 02/21/2019] [Indexed: 01/16/2023]
Abstract
OBJECTIVE The present study was designed to investigate the potential of Pistacia vera (P. vera) fruits in experimental memory impairments in mice. MATERIAL & METHODS Memory impairment was induced in Swiss Albino mice by scopolamine (0.4mg mg/kg. i.p). Animals were divided into five separate groups of six animals each, positive control group received carboxy methyl cellulose (CMC) as vehicle, negative control group received scopolamine with vehicle, and standard group received donepezil (5mg/kg i.p) with Scopolamine. Ethanolic extract of P. vera (EEPV) at doses of 200mg/kg & 400mg/kg p.o were administered to group test1 & test 2 respectively along with scopolamine. Elevated plus maze (EPM), passive avoidance paradigms and morris water maze (MWM) were used as exteroceptive behavioral models to access learning and memory activity. Transfer latency, step down latency and escape latency parameters were evaluated plus maze, passive avoidance paradigm, morris water maze. Thereafter lipid peroxidation test, glutathione level and catalase activities were estimated in homogenized brain of mice. RESULTS Pretreatment of mice with EEPV (200mg/kg & 400mg/kg) significantly reduced scopolamine induced amnesia. The obtained data clearly revealed that there was increase in escape latency in MWM and also increase in step down latency in passive avoidance paradigm. Transfer latencey was found to be decrease in EPM and biochemical. Parameters were clearly satisfied the data as compared to negative control group which was indicative of cognitive improvement. CONCLUSION P. vera fruit extract demonstrated to improve cognitive process by enhancing memory in different experimental paradigm such as EPM, passive avoidance and MWM when administered orally. Hence it would be worthwhile to explore the potential of this plant in the management of memory disorders.
Collapse
Affiliation(s)
- Satyam Singh
- School of Pharmacy, Babu Banarasi Das University, Babu Banarasi Das City, Faizabad Road, Chinhat, Lucknow- 227105, Uttar Pradesh, India
| | - Dharamveer
- School of Pharmacy, Babu Banarasi Das University, Babu Banarasi Das City, Faizabad Road, Chinhat, Lucknow- 227105, Uttar Pradesh, India
| | - Mayank Kulshreshtha
- School of Pharmacy, Babu Banarasi Das University, Babu Banarasi Das City, Faizabad Road, Chinhat, Lucknow- 227105, Uttar Pradesh, India
| |
Collapse
|
27
|
Cassidy L, Fernandez F, Johnson JB, Naiker M, Owoola AG, Broszczak DA. Oxidative stress in alzheimer's disease: A review on emergent natural polyphenolic therapeutics. Complement Ther Med 2019; 49:102294. [PMID: 32147039 DOI: 10.1016/j.ctim.2019.102294] [Citation(s) in RCA: 165] [Impact Index Per Article: 27.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2019] [Revised: 11/26/2019] [Accepted: 12/30/2019] [Indexed: 12/21/2022] Open
Abstract
OBJECTIVES The aim of this research was to review the literature on Alzheimer's disease (AD) with a focus on polyphenolics as antioxidant therapeutics. DESIGN This review included a search of the literature up to and including September 2019 in PubMed and MEDLINE databases using search terms that included: Alzheimer's Disease, Aβ peptide, tau, oxidative stress, redox, oxidation, therapeutic, antioxidant, natural therapy, polyphenol. Any review articles, case studies, research reports and articles in English were identified and subsequently interrogated. Citations within relevant articles were also examined for consideration in this review. RESULTS Alzheimer's disease is a neurodegenerative disorder that is clinically characterised by the progressive deterioration of cognitive functions and drastic changes in behaviour and personality. Due to the significant presence of oxidative damage associated with abnormal Aβ accumulation and neurofibrillary tangle deposition in AD patients' brains, antioxidant drug therapy has been investigated as potential AD treatment. In particular, naturally occurring compounds, such as plant polyphenols, have been suggested to have potential neuroprotective effects against AD due to their diverse array of physiological actions, which includes potent antioxidant effects. CONCLUSIONS The impact of oxidative stress and various mechanisms of pathogenesis in AD pathophysiology was demonstrated along with the therapeutic potential of emergent antioxidant drugs to address such mechanism of oxidation.
Collapse
Affiliation(s)
- Luke Cassidy
- School of Behavioural & Health Sciences, Faculty of Heath Sciences, Australian Catholic University, 1100 Nudgee Rd, Banyo, QLD, 4014, Australia
| | - Francesca Fernandez
- School of Behavioural & Health Sciences, Faculty of Heath Sciences, Australian Catholic University, 1100 Nudgee Rd, Banyo, QLD, 4014, Australia.
| | - Joel B Johnson
- School of Health, Medical and Applied Sciences, Central Queensland University, 630 Ibis Ave, North Rockhampton, QLD, 4701, Australia.
| | - Mani Naiker
- School of Health, Medical and Applied Sciences, Central Queensland University, 630 Ibis Ave, North Rockhampton, QLD, 4701, Australia.
| | - Akeem G Owoola
- School of Biomedical Sciences, Faculty of Health, Queensland University of Technology, 2 George St, Brisbane, 4000, QLD, Australia; Tissue Repair & Translational Physiology Program, Institute of Health & Biomedical Innovation, Queensland University of Technology, 60 Musk Ave, Kelvin Grove, Queensland, 4059, Australia.
| | - Daniel A Broszczak
- School of Biomedical Sciences, Faculty of Health, Queensland University of Technology, 2 George St, Brisbane, 4000, QLD, Australia; Tissue Repair & Translational Physiology Program, Institute of Health & Biomedical Innovation, Queensland University of Technology, 60 Musk Ave, Kelvin Grove, Queensland, 4059, Australia.
| |
Collapse
|
28
|
Gold Nanoparticles Treatment Reverses Brain Damage in Alzheimer’s Disease Model. Mol Neurobiol 2019; 57:926-936. [DOI: 10.1007/s12035-019-01780-w] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2019] [Accepted: 09/09/2019] [Indexed: 12/20/2022]
|
29
|
Abstract
Alzheimer's disease (AD) is a devastating neurodegenerative disorder without a cure. Most AD cases are sporadic where age represents the greatest risk factor. Lack of understanding of the disease mechanism hinders the development of efficacious therapeutic approaches. The loss of synapses in the affected brain regions correlates best with cognitive impairment in AD patients and has been considered as the early mechanism that precedes neuronal loss. Oxidative stress has been recognized as a contributing factor in aging and in the progression of multiple neurodegenerative diseases including AD. Increased production of reactive oxygen species (ROS) associated with age- and disease-dependent loss of mitochondrial function, altered metal homeostasis, and reduced antioxidant defense directly affect synaptic activity and neurotransmission in neurons leading to cognitive dysfunction. In addition, molecular targets affected by ROS include nuclear and mitochondrial DNA, lipids, proteins, calcium homeostasis, mitochondrial dynamics and function, cellular architecture, receptor trafficking and endocytosis, and energy homeostasis. Abnormal cellular metabolism in turn could affect the production and accumulation of amyloid-β (Aβ) and hyperphosphorylated Tau protein, which independently could exacerbate mitochondrial dysfunction and ROS production, thereby contributing to a vicious cycle. While mounting evidence implicates ROS in the AD etiology, clinical trials with antioxidant therapies have not produced consistent results. In this review, we will discuss the role of oxidative stress in synaptic dysfunction in AD, innovative therapeutic strategies evolved based on a better understanding of the complexity of molecular mechanisms of AD, and the dual role ROS play in health and disease.
Collapse
Affiliation(s)
- Eric Tönnies
- Department of Neurology, Mayo Clinic, Rochester, MN, USA
| | - Eugenia Trushina
- Department of Neurology, Mayo Clinic, Rochester, MN, USA.,Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN, USA
| |
Collapse
|
30
|
Rajasekar N, Nath C, Hanif K, Shukla R. Intranasal insulin improves cerebral blood flow, Nrf-2 expression and BDNF in STZ (ICV)-induced memory impaired rats. Life Sci 2017; 173:1-10. [DOI: 10.1016/j.lfs.2016.09.020] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2016] [Revised: 08/19/2016] [Accepted: 09/26/2016] [Indexed: 01/26/2023]
|
31
|
Rajasekar N, Nath C, Hanif K, Shukla R. Intranasal Insulin Administration Ameliorates Streptozotocin (ICV)-Induced Insulin Receptor Dysfunction, Neuroinflammation, Amyloidogenesis, and Memory Impairment in Rats. Mol Neurobiol 2016; 54:6507-6522. [PMID: 27730514 DOI: 10.1007/s12035-016-0169-8] [Citation(s) in RCA: 64] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2016] [Accepted: 09/27/2016] [Indexed: 12/14/2022]
Abstract
Alzheimer's disease (AD) is associated with reduced insulin level and impairment of insulin receptor (IR) signaling in the brain, which correlates to amyloid pathology, neuroinflammation, and synaptic neurotoxicity. Clinical studies show that intranasal insulin improves memory in AD patients without peripheral hypoglycemia. However, neuroprotective molecular mechanism of the beneficial effect of intranasal insulin in AD pathology is unexplored. Therefore, we investigated the role of intranasal insulin on intracerebroventricular (ICV) streptozotocin (STZ)-induced memory impairment in rats as evaluated in the Morris water maze test. STZ (ICV) treated rats had shown memory impairment along with a significant decrease in IR signaling molecules (IR, pIRS-1, pAkt, and pGSK-3α/β expression) and IDE expression in both hippocampus and cerebral cortex. Intranasal insulin delivery prevented these changes. Moreover, intranasal insulin was found to inhibit significantly glial cell activation (GFAP and Iba-1 expression), neuroinflammation (COX-2 expression, NFκB translocation, TNF-α, and IL-10 level) and amyloidogenic protein expression (BACE-1 and Aβ1-42 expression) in STZ (ICV)-injected rats. STZ (ICV)-induced caspase activation and postsynaptic neurotoxicity were also prevented by treatment with intranasal insulin. Our findings reveal that insulin has the neuroprotective effect and clearly signifies the potential use of intranasal insulin delivery for the treatment of AD. Graphical Abstract Neuroprotective effects of intranasal insulin administration on streptozotocin (ICV)-induced memory impairment in rats.
Collapse
Affiliation(s)
- N Rajasekar
- Divisions of Pharmacology and Toxicology, CSIR-Central Drug Research Institute, Lucknow, 226031, India
- Academy of Scientific and Innovative Research (AcSIR), Chennai, India
| | - Chandishwar Nath
- Divisions of Pharmacology and Toxicology, CSIR-Central Drug Research Institute, Lucknow, 226031, India
- Academy of Scientific and Innovative Research (AcSIR), Chennai, India
| | - Kashif Hanif
- Divisions of Pharmacology and Toxicology, CSIR-Central Drug Research Institute, Lucknow, 226031, India
- Academy of Scientific and Innovative Research (AcSIR), Chennai, India
| | - Rakesh Shukla
- Divisions of Pharmacology and Toxicology, CSIR-Central Drug Research Institute, Lucknow, 226031, India.
- Academy of Scientific and Innovative Research (AcSIR), Chennai, India.
| |
Collapse
|
32
|
Elcioğlu HK, Aslan E, Ahmad S, Alan S, Salva E, Elcioglu ÖH, Kabasakal L. Tocilizumab's effect on cognitive deficits induced by intracerebroventricular administration of streptozotocin in Alzheimer's model. Mol Cell Biochem 2016; 420:21-8. [PMID: 27443846 DOI: 10.1007/s11010-016-2762-6] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2016] [Accepted: 07/09/2016] [Indexed: 10/21/2022]
Abstract
Neuroinflammation plays pivotal roles in the pathogenesis of Alzheimer's disease (AD). IL-6 is pleiotropic cytokine which plays significant pathological role in inflammatory diseases and causes prolonged inflammation. Additionally, IL-6 activates microglia cells and enhances the accumulation of amyloid-β peptides. Moreover, IL-6 signal transduction is mediated by membrane-bound and soluble IL-6 receptors. Tocilizumab which is a humanized anti-human IL-6 receptor (IL-6R) monoclonal antibody binds to both of these receptors and inhibits IL-6 signaling by this route. The objective was to investigate tocilizumab's potential effects in the treatment of AD. Male Sprague-Dawley rats were divided into three groups: sham (control), streptozotocin (STZ), and tocilizumab-STZ. We used a single dose of intracerebroventricular (ICV) tocilizumab, beginning 1 h prior to injection of STZ for 3 weeks. The rats in STZ and tocilizumab-STZ groups were given ICV-STZ (3 mg/kg). Behavioral parameters were evaluated on days 17-20 and the rats were sacrificed on day-21 to examine histopathological changes. STZ injection caused significant decrease in the mean escape latency in passive avoidance and also declined the performance improvement in Morris water maze tests. Tocilizumab-STZ group significantly improved learning and spatial memory functions by increasing RLT in the passive avoidance and by shortening escape latency in reaching the platform in the Morris water maze. Histopathological changes were examined using hematoxylin and eosin and immunohistochemical (IHC) stainings. IHC analysis revealed that while protein expressions of amyloid-ß (3.5 ± 0.2) and IL-6 (2.9 ± 0.4) showed intense immune-positivity in STZ group, amyloid-ß (1.3 ± 0.1) and IL-6 (1.5 ± 0.2) immunoreactivities were substantially decreased in tocilizumab treatment group. We conclude that tocilizumab treatment attenuated significantly STZ-induced cognitive impairment and histopathological changes. Further studies would be desirable to investigate clinically relevant protective effects of tocilizumab in AD.
Collapse
Affiliation(s)
- H Kübra Elcioğlu
- Department of Pharmacology, Faculty of Pharmacy, Marmara University, Tıbbiye St. No: 49, Haydarpaşa, Istanbul, 34688, Turkey.
| | - Ersin Aslan
- Department of Pharmacology, Faculty of Pharmacy, Marmara University, Tıbbiye St. No: 49, Haydarpaşa, Istanbul, 34688, Turkey
| | - Sarfraz Ahmad
- Florida Hospital Medical Center, 2501 N. Orange Ave., Suite 786, Orlando, FL, 32804, USA.
| | - Saadet Alan
- Department of Pathology, Malatya State Hospital, Malatya, Turkey
| | - Emine Salva
- Department of Pharmaceutical Biotechnology, Faculty of Pharmacy, Inönü University, Malatya, Turkey
| | | | - Levent Kabasakal
- Department of Pharmacology, Faculty of Pharmacy, Marmara University, Tıbbiye St. No: 49, Haydarpaşa, Istanbul, 34688, Turkey
| |
Collapse
|
33
|
Zhao L, Xiao Y, Wang XL, Pei J, Guan ZZ. Original Research: Influence of okadaic acid on hyperphosphorylation of tau and nicotinic acetylcholine receptors in primary neurons. Exp Biol Med (Maywood) 2016; 241:1825-33. [PMID: 27190248 DOI: 10.1177/1535370216650759] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2015] [Accepted: 04/25/2016] [Indexed: 11/15/2022] Open
Abstract
The aim of the study was to investigate the influence of hyperphosphorylation of tau induced by okadaic acid on the expression of nicotinic acetylcholine receptors and the neurotoxicity of β-amyloid peptide. Primary cultures of neurons isolated from the hippocampus of the brains of neonatal rats were exposed to okadaic acid or/and Aβ1-42 Tau phosphorylated at Ser404 and Ser202, and the protein expressions of α7, α4 and α3 nAChR subunits were quantified by Western blotting, and their corresponding mRNAs by real-time PCR. Superoxide dismutase activity was assayed biochemically and malondialdehyde by thiobarbituric acid-reactive substance. As compared to controls, phosphorylations of tau at Ser404 and Ser202 in the neurons were elevated by exposure to 20 nM okadaic acid for 48 h but not by 1 or 2 µM Aβ1-42 Treatment with 20 nM okadaic acid or 1 µM Aβ1-42 for 48 h resulted in the reduced α7, α4 and α3 proteins, and α4 and α3 mRNAs, as well as the decreased activity of superoxide dismutase and the increased malondialdehyde. Okadaic acid and Aβ1-42 together caused more pronounced changes in the expressions of α7 and α4, superoxide dismutase activity and lipid peroxidation than either alone. When pre-treatment with vitamin E or lovastatin, the neurotoxicity induced by okadaic acid was significantly attenuated. These findings indicate that hyperphosphorylation of tau induced by okadaic acid inhibits the expression of nicotinic acetylcholine receptors at both the protein and mRNA levels, as well as enhances the neurotoxicity of β-amyloid peptide.
Collapse
Affiliation(s)
- Liang Zhao
- Department of Pathology at the Affiliated Hospital, Guizhou Medical University, Guiyang 550004, Guizhou, PR China
| | - Yan Xiao
- The Key Laboratory of Medical Molecular Biology, Guizhou Medical University, Guiyang 550004, Guizhou, PR China
| | - Xiao-Liang Wang
- The Key Laboratory of Medical Molecular Biology, Guizhou Medical University, Guiyang 550004, Guizhou, PR China
| | - Jinjing Pei
- KI-Alzheimer Disease Research Center, Karolinska Institutet, Novum, SE14186 Huddinge, Sweden
| | - Zhi-Zhong Guan
- Department of Pathology at the Affiliated Hospital, Guizhou Medical University, Guiyang 550004, Guizhou, PR China The Key Laboratory of Medical Molecular Biology, Guizhou Medical University, Guiyang 550004, Guizhou, PR China
| |
Collapse
|
34
|
Baker S, Götz J. A local insult of okadaic acid in wild-type mice induces tau phosphorylation and protein aggregation in anatomically distinct brain regions. Acta Neuropathol Commun 2016; 4:32. [PMID: 27037086 PMCID: PMC4818468 DOI: 10.1186/s40478-016-0300-0] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2016] [Accepted: 03/15/2016] [Indexed: 12/21/2022] Open
Abstract
In Alzheimer’s disease (AD), the distribution and density of neurofibrillary tangles, a histological hallmark comprised predominately of phosphorylated tau protein, follows a distinct pattern through anatomically connected brain regions. Studies in transgenic mice engineered to regionally confine tau expression have suggested spreading of tau within neural networks. Furthermore, injection of protein lysates isolated from brains of transgenic mice or patients with tauopathies, including AD, were shown to behave like seeds, accelerating tau pathology and tangle formation in predisposed mice. However, it remains unclear how the initiation of primary aggregation events occurs and what triggers further dissemination throughout the neural system. To consolidate these findings, we pursued an alternative approach to assess the spreading of endogenous phosphorylated tau. To generate endogenous seeds, 130 nl of 100 μM protein phosphatase 2A inhibitor okadaic acid (OA) was injected unilaterally into the amygdala of 8-month-old C57Bl/6 wild-type mice. OA was detected in brain tissue by ELISA, and found to be restricted to the injected hemispheric quadrant, where it remained detectable a week post-injection. OA injection induced tau phosphorylation that was observed not only at the injection site but also in anatomically distinct areas across both hemispheres, including the cortex and hippocampus 24 h post-injection. An increase in insoluble tau was also observed in both hemispheres of injected brains by 7 days. Furthermore, thioflavin-S detected protein aggregation at the injection site and in the cortex of both injected and contralateral hemispheres. OA injection induced no thioflavin-positivity in tau knock-out mice. The data demonstrates that a local OA insult can rapidly initiate changes in protein phosphorylation, solubility and aggregation at anatomically distant sites. This model suggests that tau phosphorylation can be both a primary response to an insult, and a secondary response communicated to non-exposed brains regions. The study highlights the use of OA to assist in understanding the initiation of tau spreading in vivo.
Collapse
|
35
|
Gokce EC, Kahveci R, Gokce A, Sargon MF, Kisa U, Aksoy N, Cemil B, Erdogan B. Curcumin Attenuates Inflammation, Oxidative Stress, and Ultrastructural Damage Induced by Spinal Cord Ischemia-Reperfusion Injury in Rats. J Stroke Cerebrovasc Dis 2016; 25:1196-1207. [PMID: 26935117 DOI: 10.1016/j.jstrokecerebrovasdis.2016.01.008] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2015] [Revised: 10/25/2015] [Accepted: 01/02/2016] [Indexed: 10/22/2022] Open
Abstract
OBJECTIVES Curcumin is a molecule found in turmeric root that possesses anti-inflammatory and antioxidant properties and has been widely used to treat neurodegenerative diseases. We investigated whether curcumin stimulates the neurorepair process and improves locomotor function in a rat model of spinal cord ischemia-reperfusion injury. METHODS Thirty-two Wistar albino rats (190-220 g) were randomly allocated into 4 groups of 8 rats each: 1 sham-operated group and 3 ischemia-reperfusion injury groups that received intraperitoneal injections of saline vehicle, methylprednisolone (MP, 30 mg/kg following induction of ischemia-reperfusion [IR] injury), or curcumin (200 mg/kg for 7 days before induction of IR injury). Spinal cord IR injury was induced by occlusion of the abdominal aorta for 30 minutes. After 24 hours of reperfusion, locomotor function was assessed using the Basso, Beattie, and Bresnahan scale. All animals were sacrificed. Spinal cord tissues were harvested to evaluate histopathological and ultrastructural alterations and to analyze levels of malondialdehyde, tumor necrosis factor-alpha, interleukin-1 beta, nitric oxide, and caspase-3, as well as enzyme activities of superoxide dismutase and glutathione peroxidase. RESULTS Intraperitoneal administration of curcumin significantly reduced inflammatory cytokine expression, attenuated oxidative stress and lipid peroxidation, prevented apoptosis, and increased antioxidant defense mechanism activity in comparison to treatment with MP or saline. Histopathological and ultrastructural abnormalities were significantly reduced in curcumin-treated rats compared to the MP- and saline-treated groups. Furthermore, curcumin significantly improved locomotor function. CONCLUSIONS Curcumin treatment preserves neuronal viability against inflammation, oxidative stress, and apoptosis associated with ischemia-reperfusion injury.
Collapse
Affiliation(s)
- Emre Cemal Gokce
- Department of Neurosurgery, Turgut Ozal University Faculty of Medicine, Ankara, Turkey.
| | - Ramazan Kahveci
- Department of Neurosurgery, Ministry of Health Kirikkale Yuksek Ihtisas State Hospital, Kirikkale, Turkey
| | - Aysun Gokce
- Department of Pathology, Ministry of Health, Diskapi Yildirim Beyazit Education and Research Hospital, Ankara, Turkey
| | | | - Ucler Kisa
- Department of Biochemistry, Kirikkale University Faculty of Medicine, Kirikkale, Turkey
| | - Nurkan Aksoy
- Department of Biochemistry, Kirikkale University Faculty of Medicine, Kirikkale, Turkey
| | - Berker Cemil
- Department of Neurosurgery, Turgut Ozal University Faculty of Medicine, Ankara, Turkey
| | - Bulent Erdogan
- Department of Neurosurgery, Turgut Ozal University Faculty of Medicine, Ankara, Turkey
| |
Collapse
|
36
|
Ameliorative Effects of Antioxidants on the Hippocampal Accumulation of Pathologic Tau in a Rat Model of Blast-Induced Traumatic Brain Injury. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2015; 2016:4159357. [PMID: 27034735 PMCID: PMC4806690 DOI: 10.1155/2016/4159357] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/22/2015] [Accepted: 09/15/2015] [Indexed: 01/22/2023]
Abstract
Traumatic brain injury (TBI) can lead to early onset dementia and other related neurodegenerative diseases. We previously demonstrated that damage to the central auditory pathway resulting from blast-induced TBI (bTBI) could be significantly attenuated by a combinatorial antioxidant treatment regimen. In the current study, we examined the localization patterns of normal Tau and the potential blast-induced accumulation of neurotoxic variants of this microtubule-associated protein that are believed to potentiate the neurodegenerative effects associated with synaptic dysfunction in the hippocampus following three successive blast overpressure exposures in nontransgenic rats. We observed a marked increase in the number of both hyperphosphorylated and oligomeric Tau-positive hilar mossy cells and somatic accumulation of endogenous Tau in oligodendrocytes in the hippocampus. Remarkably, a combinatorial regimen of 2,4-disulfonyl α-phenyl tertiary butyl nitrone (HPN-07) and N-acetylcysteine (NAC) resulted in striking reductions in the numbers of both mossy cells and oligodendrocytes positively labeled for these pathological Tau immunoreactivity patterns in response to bTBI. This treatment strategy represents a promising therapeutic approach for simultaneously reducing or eliminating both primary auditory injury and nonauditory changes associated with bTBI-induced hippocampal neurodegeneration.
Collapse
|
37
|
Examining the potential clinical value of curcumin in the prevention and diagnosis of Alzheimer’s disease. Br J Nutr 2015; 115:449-65. [DOI: 10.1017/s0007114515004687] [Citation(s) in RCA: 157] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
AbstractCurcumin derived from turmeric is well documented for its anti-carcinogenic, antioxidant and anti-inflammatory properties. Recent studies show that curcumin also possesses neuroprotective and cognitive-enhancing properties that may help delay or prevent neurodegenerative diseases, including Alzheimer’s disease (AD). Currently, clinical diagnosis of AD is onerous, and it is primarily based on the exclusion of other causes of dementia. In addition, phase III clinical trials of potential treatments have mostly failed, leaving disease-modifying interventions elusive. AD can be characterised neuropathologically by the deposition of extracellular β amyloid (Aβ) plaques and intracellular accumulation of tau-containing neurofibrillary tangles. Disruptions in Aβ metabolism/clearance contribute to AD pathogenesis. In vitro studies have shown that Aβ metabolism is altered by curcumin, and animal studies report that curcumin may influence brain function and the development of dementia, because of its antioxidant and anti-inflammatory properties, as well as its ability to influence Aβ metabolism. However, clinical studies of curcumin have revealed limited effects to date, most likely because of curcumin’s relatively low solubility and bioavailability, and because of selection of cohorts with diagnosed AD, in whom there is already major neuropathology. However, the fresh approach of targeting early AD pathology (by treating healthy, pre-clinical and mild cognitive impairment-stage cohorts) combined with new curcumin formulations that increase bioavailability is renewing optimism concerning curcumin-based therapy. The aim of this paper is to review the current evidence supporting an association between curcumin and modulation of AD pathology, including in vitro and in vivo studies. We also review the use of curcumin in emerging retinal imaging technology, as a fluorochrome for AD diagnostics.
Collapse
|
38
|
Rajasekar N, Nath C, Hanif K, Shukla R. Inhibitory Effect of Memantine on Streptozotocin-Induced Insulin Receptor Dysfunction, Neuroinflammation, Amyloidogenesis, and Neurotrophic Factor Decline in Astrocytes. Mol Neurobiol 2015; 53:6730-6744. [PMID: 26660109 DOI: 10.1007/s12035-015-9576-5] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2015] [Accepted: 11/29/2015] [Indexed: 12/21/2022]
Abstract
Our earlier studies showed that insulin receptor (IR) dysfunction along with neuroinflammation and amyloidogenesis played a major role in streptozotocin (STZ)-induced toxicity in astrocytes. N-methyl-D-aspartate (NMDA) receptor antagonist-memantine shows beneficial effects in Alzheimer's disease (AD) pathology. However, the protective molecular and cellular mechanism of memantine in astrocytes is not properly understood. Therefore, the present study was undertaken to investigate the effect of memantine on insulin receptors, neurotrophic factors, neuroinflammation, and amyloidogenesis in STZ-treated astrocytes. STZ (100 μM) treatment for 24 h in astrocytes resulted significant decrease in brain-derived neurotrophic factor (BDNF), glial cell line-derived neurotrophic factor (GDNF), and insulin-degrading enzyme (IDE) expression in astrocytes. Treatment with memantine (1-10 μM) improved STZ-induced neurotrophic factor decline (BDNF, GDNF) along with IR dysfunction as evidenced by a significant increase in IR protein expression, phosphorylation of IRS-1, Akt, and GSK-3 α/β in astrocytes. Further, memantine attenuated STZ-induced amyloid precursor protein (APP), β-site APP-cleaving enzyme-1 and amyloid-β1-42 expression and restored IDE expression in astrocytes. In addition, memantine also displays protective effects against STZ-induced astrocyte activation showed by reduction of inflammatory markers, nuclear factor kappa-B translocation, glial fibrillary acidic protein, cyclooxygenase-2, tumor necrosis factor-α level, and oxidative-nitrostative stress. The results suggest that besides the NMDA receptor antagonisic activity, effect on astroglial IR and neurotrophic factor may also be an important factor in the beneficial effect of memantine in AD pathology. Graphical Abstract Novel neuroprotective mechanisms of memenatine in streptozotocin-induced toxicity in astrocytes.
Collapse
Affiliation(s)
- N Rajasekar
- Divisions of Pharmacology and Toxicology, CSIR-Central Drug Research Institute, Lucknow, 226031, India
- Academy of Scientific and Innovative Research (AcSIR), Chennai, India
| | - Chandishwar Nath
- Divisions of Pharmacology and Toxicology, CSIR-Central Drug Research Institute, Lucknow, 226031, India
- Academy of Scientific and Innovative Research (AcSIR), Chennai, India
| | - Kashif Hanif
- Divisions of Pharmacology and Toxicology, CSIR-Central Drug Research Institute, Lucknow, 226031, India
- Academy of Scientific and Innovative Research (AcSIR), Chennai, India
| | - Rakesh Shukla
- Divisions of Pharmacology and Toxicology, CSIR-Central Drug Research Institute, Lucknow, 226031, India.
- Academy of Scientific and Innovative Research (AcSIR), Chennai, India.
| |
Collapse
|
39
|
Sachdeva AK, Chopra K. Naringin mitigate okadaic acid-induced cognitive impairment in an experimental paradigm of Alzheimer's disease. J Funct Foods 2015. [DOI: 10.1016/j.jff.2015.08.024] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
|
40
|
Tau Hyperphosphorylation and Oxidative Stress, a Critical Vicious Circle in Neurodegenerative Tauopathies? OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2015; 2015:151979. [PMID: 26576216 PMCID: PMC4630413 DOI: 10.1155/2015/151979] [Citation(s) in RCA: 192] [Impact Index Per Article: 19.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/01/2014] [Accepted: 03/08/2015] [Indexed: 12/14/2022]
Abstract
Hyperphosphorylation and aggregation of the microtubule-associated protein tau in brain, are pathological hallmarks of a large family of neurodegenerative disorders, named tauopathies, which include Alzheimer's disease. It has been shown that increased phosphorylation of tau destabilizes tau-microtubule interactions, leading to microtubule instability, transport defects along microtubules, and ultimately neuronal death. However, although mutations of the MAPT gene have been detected in familial early-onset tauopathies, causative events in the more frequent sporadic late-onset forms and relationships between tau hyperphosphorylation and neurodegeneration remain largely elusive. Oxidative stress is a further pathological hallmark of tauopathies, but its precise role in the disease process is poorly understood. Another open question is the source of reactive oxygen species, which induce oxidative stress in brain neurons. Mitochondria have been classically viewed as a major source for oxidative stress, but microglial cells were recently identified as reactive oxygen species producers in tauopathies. Here we review the complex relationships between tau pathology and oxidative stress, placing emphasis on (i) tau protein function, (ii) origin and consequences of reactive oxygen species production, and (iii) links between tau phosphorylation and oxidative stress. Further, we go on to discuss the hypothesis that tau hyperphosphorylation and oxidative stress are two key components of a vicious circle, crucial in neurodegenerative tauopathies.
Collapse
|
41
|
Dwivedi S, Rajasekar N, Hanif K, Nath C, Shukla R. Sulforaphane Ameliorates Okadaic Acid-Induced Memory Impairment in Rats by Activating the Nrf2/HO-1 Antioxidant Pathway. Mol Neurobiol 2015; 53:5310-23. [PMID: 26433376 DOI: 10.1007/s12035-015-9451-4] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2015] [Accepted: 09/16/2015] [Indexed: 11/25/2022]
Abstract
Okadaic acid (OKA) causes memory impairment and attenuates nuclear factor erythroid 2-related factor 2 (Nrf2) along with oxidative stress and neuroinflammation in rats. Sulforaphane (dietary isothiocyanate compound), an activator of Nrf2 signaling, exhibits neuroprotective effects. However, the protective effect of sulforaphane in OKA-induced neurotoxicity remains uninvestigated. Therefore, in the present study, the role of sulforaphane in OKA-induced memory impairment in rats was explored. A significant increased Nrf2 expression in the hippocampus and cerebral cortex was observed in trained (Morris water maze) rats, and a significant decreased Nrf2 expression in memory-impaired (OKA, 200 ng icv) rats indicated its involvement in memory function. Sulforaphane administration (5 and 10 mg/kg, ip, days 1 and 2) ameliorates OKA-induced memory impairment in rats. The treatment also restored Nrf2 and its downstream antioxidant protein expression (GCLC, HO-1) and attenuated oxidative stress (ROS, nitrite, GSH), neuroinflammation (NF-κB, TNF-α, IL-10), and neuronal apoptosis in the cerebral cortex and hippocampus of OKA-treated rats. Further, to determine whether modulation of Nrf2 signaling is responsible for the protective effect of sulforaphane, in vitro, Nrf2 siRNA and its downstream HO-1 inhibition studies were carried out in a rat astrocytoma cell line (C6). The protective effects of sulforaphane were abolished with Nrf2 siRNA and HO-1 inhibition in astrocytes. The results suggest that Nrf2-dependent activation of cellular antioxidant machinery results in sulforaphane-mediated protection against OKA-induced memory impairment in rats. Graphical Abstract ᅟ.
Collapse
Affiliation(s)
- Subhash Dwivedi
- Division of Pharmacology, CSIR-Central Drug Research Institute, Lucknow, 226031, India
| | - N Rajasekar
- Division of Pharmacology, CSIR-Central Drug Research Institute, Lucknow, 226031, India
- Academy of Scientific and Innovative Research (AcSIR), Chennai, India
| | - Kashif Hanif
- Division of Pharmacology, CSIR-Central Drug Research Institute, Lucknow, 226031, India
- Academy of Scientific and Innovative Research (AcSIR), Chennai, India
| | - Chandishwar Nath
- Division of Toxicology, CSIR-Central Drug Research Institute, Lucknow, 226031, India
- Academy of Scientific and Innovative Research (AcSIR), Chennai, India
| | - Rakesh Shukla
- Division of Pharmacology, CSIR-Central Drug Research Institute, Lucknow, 226031, India.
- Academy of Scientific and Innovative Research (AcSIR), Chennai, India.
| |
Collapse
|
42
|
Parada E, Buendia I, Navarro E, Avendaño C, Egea J, López MG. Microglial HO-1 induction by curcumin provides antioxidant, antineuroinflammatory, and glioprotective effects. Mol Nutr Food Res 2015; 59:1690-700. [PMID: 26047311 DOI: 10.1002/mnfr.201500279] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2015] [Revised: 05/21/2015] [Accepted: 05/21/2015] [Indexed: 12/17/2022]
Abstract
SCOPE We have studied if curcumin can protect glial cells under an oxidative stress and inflammatory environment, which is known to be deleterious in neurodegeneration. METHODS AND RESULTS Primary rat glial cultures exposed to the combination of an oxidative (rotenone/oligomycin A) and a proinflammatory LPS stimuli reduced by 50% glial viability. Under these experimental conditions, curcumin afforded significant glial protection and reduction of reactive oxygen species; these effects were blocked by the HO-1 inhibitor tin protoporphyrin-IX (SnPP). These findings correlate with the observation that curcumin induced the antioxidative protein HO-1. Most interesting was the observation that the glial protective effects related to HO-1 induction were microglial specific as shown in glial cultures from LysM(Cre) Hmox(∆/∆) mice where curcumin lost its protective effect. Under LPS conditions, curcumin reduced the microglial proinflammatory markers iNOS and tumor necrosis factor, but increased the anti-inflammatory cytokine IL4. Analysis of the microglial phenotype showed that curcumin favored a ramified morphology toward a microglial alternative activated state against LPS insult also by a HO-1-dependent mechanism. CONCLUSION The curry constituent curcumin protects glial cells and promotes a microglial anti-inflammatory phenotype by a mechanism that implicates HO-1 induction; these effects may have impact on brain protection under oxidative and inflammatory conditions.
Collapse
Affiliation(s)
- Esther Parada
- Instituto Teófilo Hernando, Facultad de Medicina, Universidad Autónoma de Madrid, Madrid, Spain.,Departamento de Farmacología y Terapéutica, Facultad de Medicina, Universidad Autónoma de Madrid, Madrid, Spain.,Instituto de Investigación Sanitaria Princesa (IIS-IP), Hospital Universitario de la Princesa, Madrid, Spain
| | - Izaskun Buendia
- Instituto Teófilo Hernando, Facultad de Medicina, Universidad Autónoma de Madrid, Madrid, Spain.,Departamento de Farmacología y Terapéutica, Facultad de Medicina, Universidad Autónoma de Madrid, Madrid, Spain
| | - Elisa Navarro
- Instituto Teófilo Hernando, Facultad de Medicina, Universidad Autónoma de Madrid, Madrid, Spain.,Departamento de Farmacología y Terapéutica, Facultad de Medicina, Universidad Autónoma de Madrid, Madrid, Spain
| | - Carlos Avendaño
- Departamento de Anatomía, Histología y Neurociencia, Facultad de Medicina, Universidad Autónoma de Madrid, Madrid, Spain
| | - Javier Egea
- Instituto Teófilo Hernando, Facultad de Medicina, Universidad Autónoma de Madrid, Madrid, Spain.,Departamento de Farmacología y Terapéutica, Facultad de Medicina, Universidad Autónoma de Madrid, Madrid, Spain.,Instituto de Investigación Sanitaria Princesa (IIS-IP), Hospital Universitario de la Princesa, Madrid, Spain
| | - Manuela G López
- Instituto Teófilo Hernando, Facultad de Medicina, Universidad Autónoma de Madrid, Madrid, Spain.,Departamento de Farmacología y Terapéutica, Facultad de Medicina, Universidad Autónoma de Madrid, Madrid, Spain.,Instituto de Investigación Sanitaria Princesa (IIS-IP), Hospital Universitario de la Princesa, Madrid, Spain
| |
Collapse
|
43
|
The Ambiguous Relationship of Oxidative Stress, Tau Hyperphosphorylation, and Autophagy Dysfunction in Alzheimer's Disease. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2015; 2015:352723. [PMID: 26171115 PMCID: PMC4485995 DOI: 10.1155/2015/352723] [Citation(s) in RCA: 127] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/07/2014] [Revised: 03/02/2015] [Accepted: 03/03/2015] [Indexed: 12/30/2022]
Abstract
Alzheimer's disease (AD) is the most common form of dementia. The pathological hallmarks of AD are amyloid plaques [aggregates of amyloid-beta (Aβ)] and neurofibrillary tangles (aggregates of tau). Growing evidence suggests that tau accumulation is pathologically more relevant to the development of neurodegeneration and cognitive decline in AD patients than Aβ plaques. Oxidative stress is a prominent early event in the pathogenesis of AD and is therefore believed to contribute to tau hyperphosphorylation. Several studies have shown that the autophagic pathway in neurons is important under physiological and pathological conditions. Therefore, this pathway plays a crucial role for the degradation of endogenous soluble tau. However, the relationship between oxidative stress, tau protein hyperphosphorylation, autophagy dysregulation, and neuronal cell death in AD remains unclear. Here, we review the latest progress in AD, with a special emphasis on oxidative stress, tau hyperphosphorylation, and autophagy. We also discuss the relationship of these three factors in AD.
Collapse
|
44
|
Oz M, Nurullahoglu Atalik KE, Yerlikaya FH, Demir EA. Curcumin alleviates cisplatin-induced learning and memory impairments. Neurobiol Learn Mem 2015; 123:43-9. [PMID: 25982942 DOI: 10.1016/j.nlm.2015.05.001] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2015] [Revised: 04/02/2015] [Accepted: 05/05/2015] [Indexed: 11/26/2022]
Abstract
The present study has been designed to investigate the role of curcumin on cisplatin-inducedcognitive impairment and to reveal mechanisms of cisplatin's detrimental actions on cognition in rats. Animals were treated with cisplatin (5mg/kg/week) and/or curcumin (300mg/kg/day) for 5weeks. Morris water maze test was used to assess spatial learning and memory. Enzymatic activities of acetylcholinesterase (AChE) and superoxide dismutase (SOD) were evaluated from hippocampus and plasma samples, and malondialdehyde (MDA), which is the end-product of lipid peroxidation, was determined by a colorimetric method. Our results showed that cisplatin (5mg/kg/week, 5weeks) caused learning and memory deficits, elevated MDA content, decreased SOD activity in the hippocampus and plasma, and AChE activity in the hippocampus. Curcumin improved learning and memory in rats with administration of cisplatin. In addition, curcumin significantly reduced the level of MDA and increased the activities of SOD and AChE. Taken together, our findings indicate that curcumin ameliorates cisplatin-induced spatial learning and memory impairment, possibly through restored cholinergic function and enhanced oxidative status.
Collapse
Affiliation(s)
- Mehmet Oz
- School of Health Services, Mevlana (Rumi) University, Konya, Turkey.
| | | | - F Humeyra Yerlikaya
- Department of Biochemistry, Faculty of Meram Medicine, Necmettin Erbakan University, Konya, Turkey
| | - Enver Ahmet Demir
- Department of Physiology, Faculty of Medicine, Mustafa Kemal University, Hatay, Turkey
| |
Collapse
|
45
|
Tiwari SK, Agarwal S, Tripathi A, Chaturvedi RK. Bisphenol-A Mediated Inhibition of Hippocampal Neurogenesis Attenuated by Curcumin via Canonical Wnt Pathway. Mol Neurobiol 2015; 53:3010-3029. [PMID: 25963729 DOI: 10.1007/s12035-015-9197-z] [Citation(s) in RCA: 98] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2015] [Accepted: 04/22/2015] [Indexed: 12/20/2022]
Abstract
Bisphenol A (BPA) is an environmental xenoestrogenic endocrine disruptor, utilized for production of consumer products, and exerts adverse effects on the developing nervous system. Recently, we found that BPA impairs the finely tuned dynamic processes of neurogenesis (generation of new neurons) in the hippocampus of the developing rat brain. Curcumin is a natural polyphenolic compound, which provides neuroprotection against various environmental neurotoxicants and in the cellular and animal models of neurodegenerative disorders. Here, we have assessed the neuroprotective efficacy of curcumin against BPA-mediated reduced neurogenesis and the underlying cellular and molecular mechanism(s). Both in vitro and in vivo studies showed that curcumin protects against BPA-induced hippocampal neurotoxicity. Curcumin protects against BPA-mediated reduced neural stem cells (NSC) proliferation and neuronal differentiation and enhanced neurodegeneration. Curcumin also enhances the expression/levels of neurogenic and the Wnt pathway genes/proteins, which were reduced due to BPA exposure in the hippocampus. Curcumin-mediated neuroprotection against BPA-induced neurotoxicity involved activation of the Wnt/β-catenin signaling pathway, which was confirmed by the use of Wnt specific activators (LiCl and GSK-3β siRNA) and inhibitor (Dkk-1). BPA-mediated increased β-catenin phosphorylation, decreased GSK-3β levels, and β-catenin nuclear translocation were significantly reversed by curcumin, leading to enhanced neurogenesis. Curcumin-induced protective effects on neurogenesis were blocked by Dkk-1 in NSC culture treated with BPA. Curcumin-mediated enhanced neurogenesis was correlated well with improved learning and memory in BPA-treated rats. Overall, our results conclude that curcumin provides neuroprotection against BPA-mediated impaired neurogenesis via activation of the Wnt/β-catenin signaling pathway.
Collapse
Affiliation(s)
- Shashi Kant Tiwari
- Developmental Toxicology Division, Systems Toxicology Group, CSIR-Indian Institute of Toxicology Research (CSIR-IITR), 80 MG Marg, Lucknow, 226001, India
- Academy of Scientific and Innovative Research (AcSIR), New Delhi, India
| | - Swati Agarwal
- Developmental Toxicology Division, Systems Toxicology Group, CSIR-Indian Institute of Toxicology Research (CSIR-IITR), 80 MG Marg, Lucknow, 226001, India
- Academy of Scientific and Innovative Research (AcSIR), New Delhi, India
| | - Anurag Tripathi
- Food, Drugs and Chemical Toxicology Group, CSIR-IITR, 80 MG Marg, Lucknow, 226001, India
| | - Rajnish Kumar Chaturvedi
- Developmental Toxicology Division, Systems Toxicology Group, CSIR-Indian Institute of Toxicology Research (CSIR-IITR), 80 MG Marg, Lucknow, 226001, India.
- Academy of Scientific and Innovative Research (AcSIR), New Delhi, India.
| |
Collapse
|
46
|
Nazem A, Sankowski R, Bacher M, Al-Abed Y. Rodent models of neuroinflammation for Alzheimer's disease. J Neuroinflammation 2015; 12:74. [PMID: 25890375 PMCID: PMC4404276 DOI: 10.1186/s12974-015-0291-y] [Citation(s) in RCA: 170] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2015] [Accepted: 03/27/2015] [Indexed: 12/24/2022] Open
Abstract
Alzheimer's disease remains incurable, and the failures of current disease-modifying strategies for Alzheimer's disease could be attributed to a lack of in vivo models that recapitulate the underlying etiology of late-onset Alzheimer's disease. The etiology of late-onset Alzheimer's disease is not based on mutations related to amyloid-β (Aβ) or tau production which are currently the basis of in vivo models of Alzheimer's disease. It has recently been suggested that mechanisms like chronic neuroinflammation may occur prior to amyloid-β and tau pathologies in late-onset Alzheimer's disease. The aim of this study is to analyze the characteristics of rodent models of neuroinflammation in late-onset Alzheimer's disease. Our search criteria were based on characteristics of an idealistic disease model that should recapitulate causes, symptoms, and lesions in a chronological order similar to the actual disease. Therefore, a model based on the inflammation hypothesis of late-onset Alzheimer's disease should include the following features: (i) primary chronic neuroinflammation, (ii) manifestations of memory and cognitive impairment, and (iii) late development of tau and Aβ pathologies. The following models fit the pre-defined criteria: lipopolysaccharide- and PolyI:C-induced models of immune challenge; streptozotocin-, okadaic acid-, and colchicine neurotoxin-induced neuroinflammation models, as well as interleukin-1β, anti-nerve growth factor and p25 transgenic models. Among these models, streptozotocin, PolyI:C-induced, and p25 neuroinflammation models are compatible with the inflammation hypothesis of Alzheimer's disease.
Collapse
Affiliation(s)
- Amir Nazem
- Elmezzi Graduate School of Molecular Medicine, The Feinstein Institute for Medical Research, 350 Community drive, Manhasset, NY, 11030, USA.
| | - Roman Sankowski
- Elmezzi Graduate School of Molecular Medicine, The Feinstein Institute for Medical Research, 350 Community drive, Manhasset, NY, 11030, USA.
| | - Michael Bacher
- Institute of Immunology, Philipps University Marburg, Hans-Meerwein-Str., 35043, Marburg, Germany.
| | - Yousef Al-Abed
- Center for Molecular Innovation, The Feinstein Institute for Medical Research, 350 Community drive, Manhasset, NY, 11030, USA.
| |
Collapse
|
47
|
Kamat PK, Nath C. Okadaic acid: a tool to study regulatory mechanisms for neurodegeneration and regeneration in Alzheimer's disease. Neural Regen Res 2015; 10:365-7. [PMID: 25878578 PMCID: PMC4396092 DOI: 10.4103/1673-5374.153679] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/15/2015] [Indexed: 01/25/2023] Open
Affiliation(s)
- Pradip Kumar Kamat
- Division of Physiology and Biophysics, University of Louisville, School of Medicine, (KY) 40202, USA
| | - Chandishwar Nath
- Division of Pharmacology, Central Drug Research Institute (CDRI), P.O. Box 173, Luck now (U.P.) 226001, India
| |
Collapse
|
48
|
Wu A, Noble EE, Tyagi E, Ying Z, Zhuang Y, Gomez-Pinilla F. Curcumin boosts DHA in the brain: Implications for the prevention of anxiety disorders. Biochim Biophys Acta Mol Basis Dis 2014; 1852:951-61. [PMID: 25550171 DOI: 10.1016/j.bbadis.2014.12.005] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2014] [Revised: 11/17/2014] [Accepted: 12/02/2014] [Indexed: 12/29/2022]
Abstract
Dietary deficiency of docosahexaenoic acid (C22:6 n-3; DHA) is linked to the neuropathology of several cognitive disorders, including anxiety. DHA, which is essential for brain development and protection, is primarily obtained through the diet or synthesized from dietary precursors, however the conversion efficiency is low. Curcumin (diferuloylmethane), which is a principal component of the spice turmeric, complements the action of DHA in the brain, and this study was performed to determine molecular mechanisms involved. We report that curcumin enhances the synthesis of DHA from its precursor, α-linolenic acid (C18:3 n-3; ALA) and elevates levels of enzymes involved in the synthesis of DHA such as FADS2 and elongase 2 in both liver and brain tissues. Furthermore, in vivo treatment with curcumin and ALA reduced anxiety-like behavior in rodents. Taken together, these data suggest that curcumin enhances DHA synthesis, resulting in elevated brain DHA content. These findings have important implications for human health and the prevention of cognitive disease, particularly for populations eating a plant-based diet or who do not consume fish, a primary source of DHA, since DHA is essential for brain function and its deficiency is implicated in many types of neurological disorders.
Collapse
Affiliation(s)
- Aiguo Wu
- Department of Integrative Biology and Physiology, University of California at Los Angeles, 621 Charles E. Young Drive Los Angeles, CA 90095, USA
| | - Emily E Noble
- Department of Integrative Biology and Physiology, University of California at Los Angeles, 621 Charles E. Young Drive Los Angeles, CA 90095, USA
| | - Ethika Tyagi
- Department of Integrative Biology and Physiology, University of California at Los Angeles, 621 Charles E. Young Drive Los Angeles, CA 90095, USA
| | - Zhe Ying
- Department of Integrative Biology and Physiology, University of California at Los Angeles, 621 Charles E. Young Drive Los Angeles, CA 90095, USA
| | - Yumei Zhuang
- Department of Integrative Biology and Physiology, University of California at Los Angeles, 621 Charles E. Young Drive Los Angeles, CA 90095, USA
| | - Fernando Gomez-Pinilla
- Department of Integrative Biology and Physiology, University of California at Los Angeles, 621 Charles E. Young Drive Los Angeles, CA 90095, USA; Department of Neurosurgery, UCLA Brain Injury Research Center, David Geffen School of medicine at UCLA, Los Angeles, CA 90095, USA.
| |
Collapse
|
49
|
Mechanism of Oxidative Stress and Synapse Dysfunction in the Pathogenesis of Alzheimer's Disease: Understanding the Therapeutics Strategies. Mol Neurobiol 2014; 53:648-661. [PMID: 25511446 DOI: 10.1007/s12035-014-9053-6] [Citation(s) in RCA: 348] [Impact Index Per Article: 31.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2014] [Accepted: 09/28/2014] [Indexed: 10/24/2022]
Abstract
Synapses are formed by interneuronal connections that permit a neuronal cell to pass an electrical or chemical signal to another cell. This passage usually gets damaged or lost in most of the neurodegenerative diseases. It is widely believed that the synaptic dysfunction and synapse loss contribute to the cognitive deficits in patients with Alzheimer's disease (AD). Although pathological hallmarks of AD are senile plaques, neurofibrillary tangles, and neuronal degeneration which are associated with increased oxidative stress, synaptic loss is an early event in the pathogenesis of AD. The involvement of major kinases such as mitogen-activated protein kinase (MAPK), extracellular receptor kinase (ERK), calmodulin-dependent protein kinase (CaMKII), glycogen synthase-3β (GSK-3β), cAMP response element-binding protein (CREB), and calcineurin is dynamically associated with oxidative stress-mediated abnormal hyperphosphorylation of tau and suggests that alteration of these kinases could exclusively be involved in the pathogenesis of AD. N-methyl-D-aspartate (NMDA) receptor (NMDAR) activation and beta amyloid (Aβ) toxicity alter the synapse function, which is also associated with protein phosphatase (PP) inhibition and tau hyperphosphorylation (two main events of AD). However, the involvement of oxidative stress in synapse dysfunction is poorly understood. Oxidative stress and free radical generation in the brain along with excitotoxicity leads to neuronal cell death. It is inferred from several studies that excitotoxicity, free radical generation, and altered synaptic function encouraged by oxidative stress are associated with AD pathology. NMDARs maintain neuronal excitability, Ca(2+) influx, and memory formation through mechanisms of synaptic plasticity. Recently, we have reported the mechanism of the synapse redox stress associated with NMDARs altered expression. We suggest that oxidative stress mediated through NMDAR and their interaction with other molecules might be a driving force for tau hyperphosphorylation and synapse dysfunction. Thus, understanding the oxidative stress mechanism and degenerating synapses is crucial for the development of therapeutic strategies designed to prevent AD pathogenesis.
Collapse
|
50
|
Relevance of the anti-inflammatory properties of curcumin in neurodegenerative diseases and depression. Molecules 2014; 19:20864-79. [PMID: 25514226 PMCID: PMC6270664 DOI: 10.3390/molecules191220864] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2014] [Revised: 12/05/2014] [Accepted: 12/08/2014] [Indexed: 02/06/2023] Open
Abstract
This review is an attempt to summarize our current understanding of curcumin's potential as a neuroprotectant and an antidepressant. This dual property confers a unique advantage to this herbal medication, believed to be devoid of any major side effects, to combat commonly observed co-morbid conditions of a neurodegenerative and a neuropsychiatric disorder. Moreover, in line with the theme of this series, the role of inflammation and stress in these diseases and possible anti-inflammatory effects of curcumin, as well as its interaction with signal transduction proteins as a common denominator in its varied mechanisms of action, are also discussed. Thus, following a brief introduction of curcumin's pharmacology, we present research suggesting how its anti-inflammatory properties have therapeutic potential in treating a devastating neurological disorder (Parkinson's disease = PD) and a debilitating neuropsychiatric disorder (major depressive disorder = MDD). It is concluded that curcumin, or better yet, an analog with better and longer bioavailability could be of important therapeutic potential in PD and/or major depression.
Collapse
|