1
|
Nadeem M, Majid H, Ansari MD, Ahmad FJ, Parvez S, Akhtar M, Ahmad S, Najmi AK. Development and Optimization of Piracetam and Shatavarin IV-Loaded Nanoemulsion for Alzheimer's Disease Therapy: In Silico and Experimental Analysis. ACS OMEGA 2025; 10:9132-9153. [PMID: 40092812 PMCID: PMC11904661 DOI: 10.1021/acsomega.4c09072] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/04/2024] [Revised: 02/13/2025] [Accepted: 02/14/2025] [Indexed: 03/19/2025]
Abstract
Alzheimer's disease (AD) presents a significant challenge due to cognitive decline resulting from nerve cell degeneration. Shatavarin IV, a prominent bioactive compound fromAsparagus racemosus and Piracetam, has been investigated for its neuroprotective potential. This study examines the molecular docking, formulation, and characterization of a nanoemulsion containing Piracetam and Shatavarin IV for treating AD. The in silico study demonstrated that Shatavarin IV exhibited strong binding affinities with multiple AD-related targets, including TNF-α (-7.29 kcal/mol), the GSK-3 axin complex (-9.6785 kcal/mol), amyloid-β (-6.8326 kcal/mol), and GSK-3 β (-8.8243 kcal/mol). The extraction of Shatavarin IV from Asparagus racemosus roots yielded 401.1 ± 2.3 mg with a purity of 66%, as confirmed by HPTLC. A combination index study revealed a synergistic effect with a CI value of 0.10843 at a 1:1 ratio of Piracetam and Shatavarin IV. The nanoemulsion was optimized using a Box-Behnken design, with oil concentration, surfactant mixture (S mix), and sonication time as key factors. The optimized formulation exhibited a particle size of 183.6 nm and a PDI of 0.194. Characterization techniques, including TEM and DSC, confirmed the uniformity, stability, and incorporation of the drugs in the nanoemulsion. The in vitro drug release study revealed a significantly higher release profile (84.30 ± 1.03% in 24 h) for the nanoemulsion than the drug suspension. Ex vivo studies demonstrated a superior permeability rate for the nanoemulsion (56.35 ± 1.19%) compared to the conventional suspension. Additionally, the nanoemulsion showed enhanced antioxidant activity compared with the pure extract. Stability studies indicated that the formulation remained stable with only minor changes in particle size, PDI, and zeta potential over time. This nanoemulsion presents a promising therapeutic strategy for AD.
Collapse
Affiliation(s)
- Mohd Nadeem
- Department
of Pharmacology. School of Pharmaceutical Education and Research. Jamia Hamdard, New Delhi 110062, India
| | - Haya Majid
- Department
of Translational and Clinical Research, School of Chemical and Life
Sciences, Jamia Hamdard, New Delhi 110062, India
| | - Mohd Danish Ansari
- Department
of Pharmaceutical Technology, Faculty of Pharmacy, Universiti Malaya, Kuala
Lumpur 50603, Malaysia
| | - Farhan Jalees Ahmad
- Department
of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi 110062, India
| | - Suhel Parvez
- Department
of Medical Elementology and Toxicology, School of Chemical and Life
Sciences, Jamia Hamdard, New Delhi 110062, India
| | - Mohd Akhtar
- Department
of Pharmacology. School of Pharmaceutical Education and Research. Jamia Hamdard, New Delhi 110062, India
| | - Sayeed Ahmad
- Department
of Pharmacognosy and Phytochemistry, School of Pharmaceutical Education
and Research, Jamia Hamdard, New Delhi 110062, India
| | - Abul Kalam Najmi
- Department
of Pharmacology. School of Pharmaceutical Education and Research. Jamia Hamdard, New Delhi 110062, India
| |
Collapse
|
2
|
Sheldon P, Mendez-Montejano J, Michalak AJ, Navarro JJ, Bradley C, Mongalo M, Zavala-Cerna MG. Pharmacological Cognitive Enhancement Among International Medical Students: Assessing Study Habits as a Novel Factor. MEDICAL SCIENCE EDUCATOR 2024; 34:1353-1360. [PMID: 39758485 PMCID: PMC11699064 DOI: 10.1007/s40670-024-02113-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 07/01/2024] [Indexed: 01/07/2025]
Abstract
Introduction Pharmacological cognitive enhancement (PCE) among healthy individuals with off-label use of medications raises ethical and safety concerns in academia. Among the few studies that have examined this phenomenon in medical students, the desire to improve performance and cope with high levels of stress are the most common associated factors implicated. Our main objective was to update current literature by exploring a novel factor by investigating the association between PCE use and student learning approaches among a cohort of international medical students. Methods We conducted a cross-sectional study with an anonymous, online survey, distributed to year 1 US international medical students at Universidad Autónoma de Guadalajara. Participants reported PCE medication use, underlying motivators, and answered items characterizing their study habits via the Approaches to Learning and Studying Inventory (ALSI). Results A total of 103 student responses were included, with 18 students (17.3%) reporting PCE use and a majority (61.1%) of whom were female. PCE users were less likely to adopt deep or strategic study approaches and were mainly motivated to improve academic performance. Discussion We encourage universities to have open discussions about medication risks and forms to improve an active learning without its use, which might be beneficial in curbing the perceived need for pharmacologic enhancement. Further explorations are required to confirm if certain study habits are truly associated with PCE use. Supplementary Information The online version contains supplementary material available at 10.1007/s40670-024-02113-w.
Collapse
Affiliation(s)
- Parker Sheldon
- Milken Institute of Public Health, George Washington University, Washington, D.C. USA
| | - Javier Mendez-Montejano
- Unidad Académica Ciencias de la Salud, Universidad Autónoma de Guadalajara, Zapopan, Mexico 45129
| | | | | | - Cersten Bradley
- University of Texas Health Science Center at San Antonio, San Antonio, TX USA
| | | | - Maria G. Zavala-Cerna
- Unidad Académica Ciencias de la Salud, Universidad Autónoma de Guadalajara, Zapopan, Mexico 45129
| |
Collapse
|
3
|
Wang B, Zou L, Li M, Zhou L. Astrocyte: A Foe or a Friend in Intellectual Disability-Related Diseases. Front Synaptic Neurosci 2022; 14:877928. [PMID: 35812794 PMCID: PMC9259964 DOI: 10.3389/fnsyn.2022.877928] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2022] [Accepted: 05/27/2022] [Indexed: 11/24/2022] Open
Abstract
Intellectual disabilities are a type of neurodevelopmental disease caused by neurological dysfunction. Their incidence is largely associated with neural development. Astrocytes are the most widely distributed cells in the mammalian brain. Previous studies have reported that astrocytes only supported and separated the neurons in the brain. However, recent studies have found that they also play an important role in neural development. Understanding the astrocyte mechanism in intellectual development disorder-related diseases will help provide new therapeutic targets for the treatment of intellectual disability-related diseases. This mini-review introduced the association between astrocyte and intellectual disabilities. Furthermore, recent advances in genetic and environmental factors causing intellectual disability and different pharmaceutical effects of intellectual disability-related drugs on astrocytes have been summarised. Finally, we discussed future perspectives of astrocyte-based therapy for intellectual disability.
Collapse
Affiliation(s)
| | | | | | - Liang Zhou
- *Correspondence: Liang Zhou, , orcid.org/0000-0003-0820-1520
| |
Collapse
|
4
|
Satoh R, Kawakami K, Nakadate K. Effects of Smart Drugs on Cholinergic System and Non-Neuronal Acetylcholine in the Mouse Hippocampus: Histopathological Approach. J Clin Med 2022; 11:jcm11123310. [PMID: 35743382 PMCID: PMC9224974 DOI: 10.3390/jcm11123310] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2022] [Revised: 05/30/2022] [Accepted: 06/07/2022] [Indexed: 11/26/2022] Open
Abstract
In recent years, people in the United States and other countries have been using smart drugs, called nootropic or cognitive enhancers, to improve concentration and memory learning skills. However, these drugs were originally prescribed for attention-deficit hyperactivity disorder and dementia, and their efficacy in healthy people has not yet been established. We focused on acetylcholine in the hippocampus, which is responsible for memory learning, and elucidate the long-term effects of smart drugs on the neural circuits. Smart drugs were administered orally in normal young mice for seven weeks. The hippocampus was sectioned and compared histologically by hematoxylin and eosin (HE) staining, immunohistochemistry for acetylcholine, and immunoelectron microscopy. There were no significant changes in acetylcholinesterase staining. However, in HE, we found perivascular edema, and choline acetyltransferase staining showed increased staining throughout the hippocampus and new signal induction in the perivascular area in the CA3, especially in the aniracetam and α-glyceryl phosphoryl choline group. Additionally, new muscarinic acetylcholine receptor signals were observed in the CA1 due to smart drug intake, suggesting that vasodilation might cause neuronal activation by increasing the influx of nutrients and oxygen. Moreover, these results suggest a possible new mechanism of acetylcholine-mediated neural circuit activation by smart drug intake.
Collapse
|
5
|
Bandaru LJM, Ayyalasomayajula N, Murumulla L, Challa S. Mechanisms associated with the dysregulation of mitochondrial function due to lead exposure and possible implications on the development of Alzheimer's disease. Biometals 2022; 35:1-25. [PMID: 35048237 DOI: 10.1007/s10534-021-00360-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2021] [Accepted: 12/09/2021] [Indexed: 01/17/2023]
Abstract
Lead (Pb) is a multimedia contaminant with various pathophysiological consequences, including cognitive decline and neural abnormalities. Recent findings have reported an association of Pb toxicity with Alzheimer's disease (AD). Studies have revealed that mitochondrial dysfunction is a pathological characteristic of AD. According to toxicology reports, Pb promotes mitochondrial oxidative stress by lowering complex III activity in the electron transport chain, boosting reactive oxygen species formation, and reducing the cell's antioxidant defence system. Here, we review recent advances in the role of mitochondria in Pb-induced AD pathology, as well as the mechanisms associated with the mitochondrial dysfunction, such as the depolarisation of the mitochondrial membrane potential, mitochondrial permeability transition pore opening; mitochondrial biogenesis, bioenergetics and mitochondrial dynamics alterations; and mitophagy and apoptosis. We also discuss possible therapeutic options for mitochondrial-targeted neurodegenerative disease (AD).
Collapse
Affiliation(s)
- Lakshmi Jaya Madhuri Bandaru
- Department of Cell Biology, National Institute of Nutrition, Indian Council of Medical Research (ICMR), Hyderabad, Telangana, 500007, India
| | - Neelima Ayyalasomayajula
- Department of Cell Biology, National Institute of Nutrition, Indian Council of Medical Research (ICMR), Hyderabad, Telangana, 500007, India
| | - Lokesh Murumulla
- Department of Cell Biology, National Institute of Nutrition, Indian Council of Medical Research (ICMR), Hyderabad, Telangana, 500007, India
| | - Suresh Challa
- Department of Cell Biology, National Institute of Nutrition, Indian Council of Medical Research (ICMR), Hyderabad, Telangana, 500007, India.
| |
Collapse
|
6
|
Khan MA, Srivastava V, Kabir M, Samal M, Insaf A, Ibrahim M, Zahiruddin S, Ahmad S. Development of Synergy-Based Combination for Learning and Memory Using in vitro, in vivo and TLC-MS-Bioautographic Studies. Front Pharmacol 2021; 12:678611. [PMID: 34276370 PMCID: PMC8283279 DOI: 10.3389/fphar.2021.678611] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2021] [Accepted: 06/22/2021] [Indexed: 12/26/2022] Open
Abstract
The present study is aimed at developing a synergistic combination to enhance learning and memory in Alzheimer’s patients with the help of eight common medicinal plants used in the AYUSH system. Aqueous and hydroalcoholic extracts of eight medicinal plants from the AYUSH system of medicine were prepared. These were subjected to in vitro anticholinesterase activity, to find out the combination index of synergistic combination. The synergistic combination and their individual extracts were subjected to total phenol, flavonoid and antioxidant activity estimation. Further, in vivo neurobehavioral studies in rats were carried out followed by TLC-MS-bioautographic identification of bioactive metabolites. Out of the sixteen extracts, aqueous extracts of Withania somnifera (L.) Dunal (WSA) and Myristica fragrans (L.) Dunal (MFA) were selected for the development of synergistic combination based on their IC50 value in vitro anticholinesterase assay. The synergistic combination inhibited the anticholinesterase activity significantly as compared to the individual extracts of WSA and MFA. The synergistic combination also showed more phenolic and flavonoid contents with potential antioxidant activity. The TLC-bioautography showed four white spots in WSA, signifying sitoindosides VII, VIII, quercetin, isopelletierine and Withanolide S as AChE inhibitory compounds while showing five white spots of anti-cholinesterase active metabolites identified as eugenol, methyl eugenol, myristic acid, galbacin and β-sitosterol in MFA. The observation of neurocognitive behavior in amnesia induced subjects manifested that both the synergistic combinations showed comparable results to that of standard piracetam, though the synergistic combination containing a higher concentration of WSA showed more appreciable results in ameliorating dementia in rats. The study suggests that the synergy based combination successfully enhanced memory and learning by abating free radical and acetylcholine levels, and increased learning and memory in rats, providing a strong rationale for its use in the treatment of dementia and Alzheimer’s disease.
Collapse
Affiliation(s)
- Maaz Ahmed Khan
- Bioactive Natural Product Laboratory, Department of Pharmacognosy and Phytochemistry, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, India
| | - Varsha Srivastava
- Bioactive Natural Product Laboratory, Department of Pharmacognosy and Phytochemistry, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, India
| | - Mariya Kabir
- Bioactive Natural Product Laboratory, Department of Pharmacognosy and Phytochemistry, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, India
| | - Monalisha Samal
- Bioactive Natural Product Laboratory, Department of Pharmacognosy and Phytochemistry, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, India
| | - Areeba Insaf
- Bioactive Natural Product Laboratory, Department of Pharmacognosy and Phytochemistry, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, India
| | - Mohammad Ibrahim
- Bioactive Natural Product Laboratory, Department of Pharmacognosy and Phytochemistry, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, India
| | - Sultan Zahiruddin
- Bioactive Natural Product Laboratory, Department of Pharmacognosy and Phytochemistry, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, India
| | - Sayeed Ahmad
- Bioactive Natural Product Laboratory, Department of Pharmacognosy and Phytochemistry, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, India
| |
Collapse
|
7
|
González-Fernández R, Grigoruţă M, Chávez-Martínez S, Ruiz-May E, Elizalde-Contreras JM, Valero-Galván J, Martínez-Martínez A. Liver proteome alterations in psychologically distressed rats and a nootropic drug. PeerJ 2021; 9:e11483. [PMID: 34055494 PMCID: PMC8140599 DOI: 10.7717/peerj.11483] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Accepted: 04/27/2021] [Indexed: 11/20/2022] Open
Abstract
BACKGROUND Chronic psychological distress is considered today a pandemic due to the modern lifestyle and has been associated with various neurodegenerative, autoimmune, or systemic inflammation-related diseases. Stress is closely related to liver disease exacerbation through the high activity of the endocrine and autonomic nervous systems, and the connection between the development of these pathologies and the physiological effects induced by oxidative stress is not yet completely understood. The use of nootropics, as the cognitive enhancer and antioxidant piracetam, is attractive to repair the oxidative damage. A proteomic approach provides the possibility to obtain an in-depth comprehension of the affected cellular processes and the possible consequences for the body. Therefore, we considered to describe the effect of distress and piracetam on the liver proteome. METHODS We used a murine model of psychological stress by predatory odor as a distress paradigm. Female Sprague-Dawley rats were distributed into four experimental groups (n = 6 - 7/group) and were exposed or not to the stressor for five days and treated or not with piracetam (600 mg/kg) for six days. We evaluated the liver proteome by one-dimensional sodium dodecyl sulfate-polyacrylamide gel electrophoresis (1D-SDS-PAGE) followed by liquid chromatography-tandem mass spectrometry (GeLC-MS/MS). Besides, we analyzed the activity of liver antioxidant enzymes, the biochemical parameters in plasma and rat behavior. RESULTS Our results showed that distress altered a wide range of proteins involved in amino acids metabolism, glucose, and fatty acid mobilization and degradation on the way to produce energy, protein folding, trafficking and degradation, redox metabolism, and its implications in the development of the non-alcoholic fatty liver disease (NAFLD). Piracetam reverted the changes in metabolism caused by distress exposure, and, under physiological conditions, it increased catabolism rate directed towards energy production. These results confirm the possible relationship between chronic psychological stress and the progression of NAFLD, as well as we newly evidenced the controversial beneficial effects of piracetam. Finally, we propose new distress biomarkers in the liver as the protein DJ-1 (PARK7), glutathione peroxidase 1 (GPX), peroxiredoxin-5 (PRDX5), glutaredoxin 5 (GLRX5), and thioredoxin reductase 1 (TXNDR1), and in plasma as biochemical parameters related to kidney function such as urea and blood urea nitrogen (BUN) levels.
Collapse
Affiliation(s)
- Raquel González-Fernández
- Departamento de Ciencias Químico Biológicas, Instituto de Ciencias Biomédicas, Universidad Autónoma de Ciudad Juárez, Ciudad Juárez, Chihuahua, Mexico
| | - Mariana Grigoruţă
- Departamento de Ciencias Químico Biológicas, Instituto de Ciencias Biomédicas, Universidad Autónoma de Ciudad Juárez, Ciudad Juárez, Chihuahua, Mexico
| | - Sarahi Chávez-Martínez
- Departamento de Ciencias Químico Biológicas, Instituto de Ciencias Biomédicas, Universidad Autónoma de Ciudad Juárez, Ciudad Juárez, Chihuahua, Mexico
| | - Eliel Ruiz-May
- Red de Estudios Moleculares Avanzados, Instituto de Ecología A.C. (INECOL), Xalapa, Veracruz, México
| | | | - José Valero-Galván
- Departamento de Ciencias Químico Biológicas, Instituto de Ciencias Biomédicas, Universidad Autónoma de Ciudad Juárez, Ciudad Juárez, Chihuahua, Mexico
| | - Alejandro Martínez-Martínez
- Departamento de Ciencias Químico Biológicas, Instituto de Ciencias Biomédicas, Universidad Autónoma de Ciudad Juárez, Ciudad Juárez, Chihuahua, Mexico
| |
Collapse
|
8
|
Li S, Bianconi S, van der Veen JW, Do AD, Stolinski J, Cecil KM, Hannah-Shmouni F, Porter FD, Shen J. Oxidative phosphorylation in creatine transporter deficiency. NMR IN BIOMEDICINE 2021; 34:e4419. [PMID: 32990357 PMCID: PMC7722185 DOI: 10.1002/nbm.4419] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/15/2020] [Revised: 09/03/2020] [Accepted: 09/10/2020] [Indexed: 06/11/2023]
Abstract
X-linked creatine transporter deficiency (CTD) is one of the three types of cerebral creatine deficiency disorders. CTD arises from pathogenic variants in the X-linked gene SLC6A8. We report the first phosphorus (31 P) MRS study of patients with CTD, where both phosphocreatine and total creatine concentrations were found to be markedly reduced. Despite the diminished role of creatine and phosphocreatine in oxidative phosphorylation in CTD, we found no elevation of lactate or lowered pH, indicating that the brain energy supply still largely relied on oxidative metabolism. Our results suggest that mitochondrial function is a potential therapeutic target for CTD.
Collapse
Affiliation(s)
- Shizhe Li
- Molecular Imaging Branch, National Institute of Mental Health, Bethesda, MD, USA
| | - Simona Bianconi
- Division of Translational Medicine, Eunice Kennedy Shriver National Institute of Child Health and Human Development, Bethesda, MD, USA
| | | | - An Dang Do
- Division of Translational Medicine, Eunice Kennedy Shriver National Institute of Child Health and Human Development, Bethesda, MD, USA
| | - JoEllyn Stolinski
- NMR Facility, National Institute of Neurological Disorders and Stroke, Bethesda, MD, USA
| | - Kim M. Cecil
- Department of Radiology, Cincinnati Children’s Hospital Medical Center and the University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| | - Fady Hannah-Shmouni
- Division of Translational Medicine, Eunice Kennedy Shriver National Institute of Child Health and Human Development, Bethesda, MD, USA
| | - Forbes D. Porter
- Division of Translational Medicine, Eunice Kennedy Shriver National Institute of Child Health and Human Development, Bethesda, MD, USA
| | - Jun Shen
- Molecular Imaging Branch, National Institute of Mental Health, Bethesda, MD, USA
| |
Collapse
|
9
|
Napoletano F, Schifano F, Corkery JM, Guirguis A, Arillotta D, Zangani C, Vento A. The Psychonauts' World of Cognitive Enhancers. Front Psychiatry 2020; 11:546796. [PMID: 33024436 PMCID: PMC7516264 DOI: 10.3389/fpsyt.2020.546796] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/29/2020] [Accepted: 08/17/2020] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND There is growing availability of novel psychoactive substances (NPS), including cognitive enhancers (CEs) which can be used in the treatment of certain mental health disorders. While treating cognitive deficit symptoms in neuropsychiatric or neurodegenerative disorders using CEs might have significant benefits for patients, the increasing recreational use of these substances by healthy individuals raises many clinical, medico-legal, and ethical issues. Moreover, it has become very challenging for clinicians to keep up-to-date with CEs currently available as comprehensive official lists do not exist. METHODS Using a web crawler (NPSfinder®), the present study aimed at assessing psychonaut fora/platforms to better understand the online situation regarding CEs. We compared NPSfinder® entries with those from the European Monitoring Centre for Drugs and Drug Addiction (EMCDDA) and from the United Nations Office on Drugs and Crime (UNODC) NPS databases up to spring 2019. Any substance that was identified by NPSfinder® was considered a CE if it was either described as having nootropic abilities by psychonauts or if it was listed among the known CEs by Froestl and colleagues. RESULTS A total of 142 unique CEs were identified by NPSfinder®. They were divided into 10 categories, including plants/herbs/products (29%), prescribed drugs (17%), image and performance enhancing drugs (IPEDs) (15%), psychostimulants (15%), miscellaneous (8%), Phenethylamines (6%), GABAergic drugs (5%), cannabimimetic (4%), tryptamines derivatives (0.5%), and piperazine derivatives (0.5%). A total of 105 chemically different substances were uniquely identified by NPSfinder®. Only one CE was uniquely identified by the EMCDDA; no CE was uniquely identified by the UNODC. CONCLUSIONS These results show that NPSfinder® is helpful as part of an Early Warning System, which could update clinicians with the growing numbers and types of nootropics in the increasingly difficult-to-follow internet world. Improving clinicians' knowledge of NPS could promote more effective prevention and harm reduction measures in clinical settings.
Collapse
Affiliation(s)
- Flavia Napoletano
- Department of Mental Health, Homerton University Hospital, East London Foundation Trust, London, United Kingdom
- Psychopharmacology, Drug Misuse, and Novel Psychoactive Substances Research Unit, School of Life and Medical Sciences, University of Hertfordshire, Hatfield, United Kingdom
| | - Fabrizio Schifano
- Psychopharmacology, Drug Misuse, and Novel Psychoactive Substances Research Unit, School of Life and Medical Sciences, University of Hertfordshire, Hatfield, United Kingdom
| | - John Martin Corkery
- Psychopharmacology, Drug Misuse, and Novel Psychoactive Substances Research Unit, School of Life and Medical Sciences, University of Hertfordshire, Hatfield, United Kingdom
| | - Amira Guirguis
- Psychopharmacology, Drug Misuse, and Novel Psychoactive Substances Research Unit, School of Life and Medical Sciences, University of Hertfordshire, Hatfield, United Kingdom
- Swansea University Medical School, Institute of Life Sciences 2, Swansea University, Swansea, United Kingdom
| | - Davide Arillotta
- Psychopharmacology, Drug Misuse, and Novel Psychoactive Substances Research Unit, School of Life and Medical Sciences, University of Hertfordshire, Hatfield, United Kingdom
- Psychiatry Unit, Department of Clinical and Experimental Medicine, University of Catania, Catania, Italy
| | - Caroline Zangani
- Psychopharmacology, Drug Misuse, and Novel Psychoactive Substances Research Unit, School of Life and Medical Sciences, University of Hertfordshire, Hatfield, United Kingdom
- Department of Health Sciences, University of Milan, Milan, Italy
| | - Alessandro Vento
- Department of Mental Health, Addictions’ Observatory (ODDPSS), Rome, Italy
- Department of Mental Health, Guglielmo Marconi” University, Rome, Italy
- Department of Mental Health, ASL Roma 2, Rome, Italy
| |
Collapse
|
10
|
Grigoruţă M, Vargas-Caraveo A, Vázquez-Mayorga E, Castillo-Michel HA, Díaz-Sánchez ÁG, Reyes-Herrera J, Martínez-Martínez A. Blood mononuclear cells as speculum of emotional stress analyzed by synchrotron infrared spectroscopy and a nootropic drug. SPECTROCHIMICA ACTA. PART A, MOLECULAR AND BIOMOLECULAR SPECTROSCOPY 2018; 204:475-483. [PMID: 29966903 DOI: 10.1016/j.saa.2018.06.075] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/06/2018] [Revised: 06/20/2018] [Accepted: 06/20/2018] [Indexed: 06/08/2023]
Abstract
Chronic psychological stress is an important public health issue which generates behavioral changes, anxiety, immunosuppression and oxidative damage. Piracetam is a cognitive enhancer, at cellular level it protects from oxidative stress. The aim of this study was to evaluate the effect of psychological stress and of piracetam on circulating mononuclear cells by analyzing the biochemical spectrome using Synchrotron Radiation Fourier Transform Infrared Microspectroscopy (SR-μFTIR). Rats were exposed for five days to a stressor (cat odor) under oral administration of piracetam (600 mg/kg). SR-μFTIR analysis showed a decrease in bands associated to the lipids region (2852 cm-1, 2923 cm-1 and 2962 cm-1) and an increase absorption of the amide I band (1654 cm-1) under stress conditions. The principal component analysis showed increase oxidation of lipids (decrease of 3010 cm-1, 2923 cm-1 and 2852 cm-1 bands) as well as proteins denaturation (increase of 1610 cm-1 and 1690 cm-1 bands) under stress. Piracetam provided protection to polyunsaturated lipids (p ≤ 0.001) and lipids/proteins ratio (p ≤ 0.001). Behaviorally, this drug diminished fear and anxiety in stressed animals by the plus maze test (p ≤ 0.002). However, this drug induced oxidative stress in mononuclear cells from unstressed animals and altered their behavior.
Collapse
Affiliation(s)
- Mariana Grigoruţă
- Departamento de Ciencias Químico Biológicas, Instituto de Ciencias Biomédicas, Universidad Autónoma de Ciudad Juárez (UACJ), Anillo envolvente Pronaf y Estocolmo s/n, 32310 Cd. Juárez, Mexico
| | | | - Emmanuel Vázquez-Mayorga
- Departamento de Ciencias Químico Biológicas, Instituto de Ciencias Biomédicas, Universidad Autónoma de Ciudad Juárez (UACJ), Anillo envolvente Pronaf y Estocolmo s/n, 32310 Cd. Juárez, Mexico
| | | | - Ángel G Díaz-Sánchez
- Departamento de Ciencias Químico Biológicas, Instituto de Ciencias Biomédicas, Universidad Autónoma de Ciudad Juárez (UACJ), Anillo envolvente Pronaf y Estocolmo s/n, 32310 Cd. Juárez, Mexico
| | - Juan Reyes-Herrera
- European Synchrotron Radiation Facility (ESRF), B.P. 220, Grenoble, France
| | - Alejandro Martínez-Martínez
- Departamento de Ciencias Químico Biológicas, Instituto de Ciencias Biomédicas, Universidad Autónoma de Ciudad Juárez (UACJ), Anillo envolvente Pronaf y Estocolmo s/n, 32310 Cd. Juárez, Mexico; El Colegio de Chihuahua, Calle Partido Díaz 4723 esquina con Anillo Envolvente del PRONAF, colonia Progresista, Ciudad Juárez, Chihuahua C.P. 32310, Mexico.
| |
Collapse
|
11
|
Verma DK, Gupta S, Biswas J, Joshi N, Sivarama Raju K, Wahajuddin M, Singh S. Metabolic Enhancer Piracetam Attenuates the Translocation of Mitochondrion-Specific Proteins of Caspase-Independent Pathway, Poly [ADP-Ribose] Polymerase 1 Up-regulation and Oxidative DNA Fragmentation. Neurotox Res 2018; 34:198-219. [PMID: 29532444 DOI: 10.1007/s12640-018-9878-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2017] [Revised: 01/29/2018] [Accepted: 01/31/2018] [Indexed: 11/28/2022]
Abstract
Piracetam, a nootropic drug, has been clinically used for decades; however, its mechanism of action still remains enigmatic. The present study was undertaken to evaluate the role of mitochondrion-specific factors of caspase-independent pathway like apoptotic-inducing factor (AIF) and endonuclease-G (endo-G) in piracetam-induced neuroprotection. N2A cells treated with lipopolysaccharide (LPS) exhibited significant cytotoxicity, impaired mitochondrial activity, and reactive oxygen species generation which was significantly attenuated with piracetam co-treatment. Cells co-treated with LPS and piracetam exhibited significant uptake of piracetam in comparison to only piracetam-treated cells as estimated by liquid chromatography-mass spectrometry (LC-MSMS). LPS treatment caused significant translocation of AIF and endonuclease-G in neuronal N2A cells which were significantly attenuated with piracetam co-treatment. Significant over-expression of proinflammatory cytokines was also observed after treatment of LPS to cells which was inhibited with piracetam co-treatment demonstrating its anti-inflammatory property. LPS-treated cells exhibited significant oxidative DNA fragmentation and poly [ADP-ribose] polymerase-1 (PARP-1) up-regulation in nucleus, both of which were attenuated with piracetam treatment. Antioxidant melatonin but not z-VAD offered the inhibited LPS-induced DNA fragmentation indicating the involvement of oxidative DNA fragmentation. Further, we did not observe the altered caspase-3 level after LPS treatment initially while at a later time point, significantly augmented level of caspase-3 was observed which was not inhibited with piracetam treatment. In total, our findings indicate the interference of piracetam in mitochondrion-mediated caspase-independent pathway, as well as its anti-inflammatory and antioxidative properties. Graphical Abstract Graphical abstract indicating the novel interference of metabolic enhancer piracetam (P) in neuronal death mechanisms.
Collapse
Affiliation(s)
- Dinesh Kumar Verma
- Toxicology Division, CSIR-Central Drug Research Institute, Lucknow, Uttar Pradesh, 226031, India
| | - Sonam Gupta
- Toxicology Division, CSIR-Central Drug Research Institute, Lucknow, Uttar Pradesh, 226031, India
| | - Joyshree Biswas
- Toxicology Division, CSIR-Central Drug Research Institute, Lucknow, Uttar Pradesh, 226031, India
| | - Neeraj Joshi
- Department of Biochemistry and Biophysics, Helen Diller Comprehensive Cancer Center, University of California San Francisco, San Francisco, CA, USA
| | - K Sivarama Raju
- Pharmacokinetics & Metabolism Division, CSIR-Central Drug Research Institute, Lucknow, Uttar Pradesh, 226031, India
| | - Mu Wahajuddin
- Pharmacokinetics & Metabolism Division, CSIR-Central Drug Research Institute, Lucknow, Uttar Pradesh, 226031, India
| | - Sarika Singh
- Toxicology Division, CSIR-Central Drug Research Institute, Lucknow, Uttar Pradesh, 226031, India.
| |
Collapse
|
12
|
Ramesh S, Govindarajulu M, Lynd T, Briggs G, Adamek D, Jones E, Heiner J, Majrashi M, Moore T, Amin R, Suppiramaniam V, Dhanasekaran M. SIRT3 activator Honokiol attenuates β-Amyloid by modulating amyloidogenic pathway. PLoS One 2018; 13:e0190350. [PMID: 29324783 PMCID: PMC5764272 DOI: 10.1371/journal.pone.0190350] [Citation(s) in RCA: 57] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2017] [Accepted: 12/13/2017] [Indexed: 01/06/2023] Open
Abstract
Honokiol (poly-phenolic lignan from Magnolia grandiflora) is a Sirtuin-3 (SIRT3) activator which exhibit antioxidant activity and augment mitochondrial functions in several experimental models. Modern evidence suggests the critical role of SIRT3 in the progression of several metabolic and neurodegenerative diseases. Amyloid beta (Aβ), the precursor to extracellular senile plaques, accumulates in the brains of patients with Alzheimer's disease (AD) and is related to the development of cognitive impairment and neuronal cell death. Aβ is generated from amyloid-β precursor protein (APP) through sequential cleavages, first by β-secretase and then by γ-secretase. Drugs modulating this pathway are believed to be one of the most promising strategies for AD treatment. In the present study, we found that Honokiol significantly enhanced SIRT3 expression, reduced reactive oxygen species generation and lipid peroxidation, enhanced antioxidant activities, and mitochondrial function thereby reducing Aβ and sAPPβ levels in Chinese Hamster Ovarian (CHO) cells (carrying the amyloid precursor protein-APP and Presenilin PS1 mutation). Mechanistic studies revealed that Honokiol affects neither protein levels of APP nor α-secretase activity. In contrast, Honokiol increased the expression of AMPK, CREB, and PGC-1α, thereby inhibiting β-secretase activity leading to reduced Aβ levels. These results suggest that Honokiol is an activator of SIRT3 capable of improving antioxidant activity, mitochondrial energy regulation, while decreasing Aβ, thereby indicating it to be a lead compound for AD drug development.
Collapse
Affiliation(s)
- Sindhu Ramesh
- Department of Drug Discovery and Development, Harrison School of Pharmacy, Auburn University, Auburn, AL, United States of America
| | - Manoj Govindarajulu
- Department of Drug Discovery and Development, Harrison School of Pharmacy, Auburn University, Auburn, AL, United States of America
| | - Tyler Lynd
- Department of Drug Discovery and Development, Harrison School of Pharmacy, Auburn University, Auburn, AL, United States of America
| | - Gwyneth Briggs
- Department of Drug Discovery and Development, Harrison School of Pharmacy, Auburn University, Auburn, AL, United States of America
| | - Danielle Adamek
- Department of Drug Discovery and Development, Harrison School of Pharmacy, Auburn University, Auburn, AL, United States of America
| | - Ellery Jones
- Department of Drug Discovery and Development, Harrison School of Pharmacy, Auburn University, Auburn, AL, United States of America
| | - Jake Heiner
- Department of Drug Discovery and Development, Harrison School of Pharmacy, Auburn University, Auburn, AL, United States of America
| | - Mohammed Majrashi
- Department of Drug Discovery and Development, Harrison School of Pharmacy, Auburn University, Auburn, AL, United States of America
| | - Timothy Moore
- Department of Drug Discovery and Development, Harrison School of Pharmacy, Auburn University, Auburn, AL, United States of America
| | - Rajesh Amin
- Department of Drug Discovery and Development, Harrison School of Pharmacy, Auburn University, Auburn, AL, United States of America
| | - Vishnu Suppiramaniam
- Department of Drug Discovery and Development, Harrison School of Pharmacy, Auburn University, Auburn, AL, United States of America
| | - Muralikrishnan Dhanasekaran
- Department of Drug Discovery and Development, Harrison School of Pharmacy, Auburn University, Auburn, AL, United States of America
| |
Collapse
|
13
|
Beckervordersandforth R. Mitochondrial Metabolism-Mediated Regulation of Adult Neurogenesis. Brain Plast 2017; 3:73-87. [PMID: 29765861 PMCID: PMC5928529 DOI: 10.3233/bpl-170044] [Citation(s) in RCA: 65] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
The life-long generation of new neurons from radial glia-like neural stem cells (NSCs) is achieved through a stereotypic developmental sequence that requires precise regulatory mechanisms to prevent exhaustion or uncontrolled growth of the stem cell pool. Cellular metabolism is the new kid on the block of adult neurogenesis research and the identity of stage-specific metabolic programs and their impact on neurogenesis turns out to be an emerging research topic in the field. Mitochondrial metabolism is best known for energy production but it contains a great deal more. Mitochondria are key players in a variety of cellular processes including ATP synthesis through functional coupling of the electron transport chain and oxidative phosphorylation, recycling of hydrogen carriers, biosynthesis of cellular building blocks, and generation of reactive oxygen species that can modulate signaling pathways in a redox-dependent fashion. In this review, I will discuss recent findings describing stage-specific modulations of mitochondrial metabolism within the adult NSC lineage, emphasizing its importance for NSC self-renewal, proliferation of neural stem and progenitor cells (NSPCs), cell fate decisions, and differentiation and maturation of newborn neurons. I will furthermore summarize the important role of mitochondrial dysfunction in tissue regeneration and ageing, suggesting it as a potential therapeutic target for regenerative medicine practice.
Collapse
Affiliation(s)
- Ruth Beckervordersandforth
- Institute of Biochemistry, Emil Fischer Center, Friedrich-Alexander Universität Erlangen-Nürnberg, Germany
| |
Collapse
|
14
|
Beckervordersandforth R, Ebert B, Schäffner I, Moss J, Fiebig C, Shin J, Moore DL, Ghosh L, Trinchero MF, Stockburger C, Friedland K, Steib K, von Wittgenstein J, Keiner S, Redecker C, Hölter SM, Xiang W, Wurst W, Jagasia R, Schinder AF, Ming GL, Toni N, Jessberger S, Song H, Lie DC. Role of Mitochondrial Metabolism in the Control of Early Lineage Progression and Aging Phenotypes in Adult Hippocampal Neurogenesis. Neuron 2017; 93:560-573.e6. [PMID: 28111078 DOI: 10.1016/j.neuron.2016.12.017] [Citation(s) in RCA: 199] [Impact Index Per Article: 24.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2015] [Revised: 08/06/2016] [Accepted: 11/23/2016] [Indexed: 12/20/2022]
Abstract
Precise regulation of cellular metabolism is hypothesized to constitute a vital component of the developmental sequence underlying the life-long generation of hippocampal neurons from quiescent neural stem cells (NSCs). The identity of stage-specific metabolic programs and their impact on adult neurogenesis are largely unknown. We show that the adult hippocampal neurogenic lineage is critically dependent on the mitochondrial electron transport chain and oxidative phosphorylation machinery at the stage of the fast proliferating intermediate progenitor cell. Perturbation of mitochondrial complex function by ablation of the mitochondrial transcription factor A (Tfam) reproduces multiple hallmarks of aging in hippocampal neurogenesis, whereas pharmacological enhancement of mitochondrial function ameliorates age-associated neurogenesis defects. Together with the finding of age-associated alterations in mitochondrial function and morphology in NSCs, these data link mitochondrial complex function to efficient lineage progression of adult NSCs and identify mitochondrial function as a potential target to ameliorate neurogenesis-defects in the aging hippocampus.
Collapse
Affiliation(s)
- Ruth Beckervordersandforth
- Institute of Biochemistry, Emil Fischer Center, Friedrich-Alexander Universität Erlangen-Nürnberg, 91054 Erlangen, Germany.
| | - Birgit Ebert
- Institute of Biochemistry, Emil Fischer Center, Friedrich-Alexander Universität Erlangen-Nürnberg, 91054 Erlangen, Germany; Institute of Developmental Genetics, Helmholtz Center Munich, German Research Center for Environmental Health, 85764 Munich-Neuherberg, Germany
| | - Iris Schäffner
- Institute of Biochemistry, Emil Fischer Center, Friedrich-Alexander Universität Erlangen-Nürnberg, 91054 Erlangen, Germany
| | - Jonathan Moss
- Department of Fundamental Neuroscience, University of Lausanne, 1005 Lausanne, Switzerland
| | - Christian Fiebig
- Institute of Biochemistry, Emil Fischer Center, Friedrich-Alexander Universität Erlangen-Nürnberg, 91054 Erlangen, Germany
| | - Jaehoon Shin
- Institute for Cell Engineering, Department of Neurology, The Solomon Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Darcie L Moore
- Brain Research Institute, Faculty of Medicine and Science, University of Zurich, 8057 Zurich, Switzerland
| | - Laboni Ghosh
- Brain Research Institute, Faculty of Medicine and Science, University of Zurich, 8057 Zurich, Switzerland
| | - Mariela F Trinchero
- Laboratory of Neuronal Plasticity, Leloir Institute (IIBBA, CONICET), C1405BWE Buenos Aires, Argentina
| | - Carola Stockburger
- Molecular and Clinical Pharmacy, Friedrich-Alexander Universität Erlangen-Nürnberg, 91054 Erlangen, Germany
| | - Kristina Friedland
- Molecular and Clinical Pharmacy, Friedrich-Alexander Universität Erlangen-Nürnberg, 91054 Erlangen, Germany
| | - Kathrin Steib
- Institute of Developmental Genetics, Helmholtz Center Munich, German Research Center for Environmental Health, 85764 Munich-Neuherberg, Germany
| | - Julia von Wittgenstein
- Institute of Biochemistry, Emil Fischer Center, Friedrich-Alexander Universität Erlangen-Nürnberg, 91054 Erlangen, Germany
| | - Silke Keiner
- Hans Berger Department of Neurology, Jena University Hospital, 07747 Jena, Germany
| | - Christoph Redecker
- Hans Berger Department of Neurology, Jena University Hospital, 07747 Jena, Germany
| | - Sabine M Hölter
- Institute of Developmental Genetics, Helmholtz Center Munich, German Research Center for Environmental Health, 85764 Munich-Neuherberg, Germany
| | - Wei Xiang
- Institute of Biochemistry, Emil Fischer Center, Friedrich-Alexander Universität Erlangen-Nürnberg, 91054 Erlangen, Germany
| | - Wolfgang Wurst
- Institute of Developmental Genetics, Helmholtz Center Munich, German Research Center for Environmental Health, 85764 Munich-Neuherberg, Germany
| | - Ravi Jagasia
- Institute of Developmental Genetics, Helmholtz Center Munich, German Research Center for Environmental Health, 85764 Munich-Neuherberg, Germany; F. Hoffmann-La Roche Ltd, CNS Discovery; Pharma Research and Early Development, 4070 Basel, Switzerland
| | - Alejandro F Schinder
- Laboratory of Neuronal Plasticity, Leloir Institute (IIBBA, CONICET), C1405BWE Buenos Aires, Argentina
| | - Guo-Li Ming
- Institute for Cell Engineering, Department of Neurology, The Solomon Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Nicolas Toni
- Department of Fundamental Neuroscience, University of Lausanne, 1005 Lausanne, Switzerland
| | - Sebastian Jessberger
- Brain Research Institute, Faculty of Medicine and Science, University of Zurich, 8057 Zurich, Switzerland
| | - Hongjun Song
- Institute for Cell Engineering, Department of Neurology, The Solomon Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - D Chichung Lie
- Institute of Biochemistry, Emil Fischer Center, Friedrich-Alexander Universität Erlangen-Nürnberg, 91054 Erlangen, Germany.
| |
Collapse
|
15
|
Hou Y, Song H, Croteau DL, Akbari M, Bohr VA. Genome instability in Alzheimer disease. Mech Ageing Dev 2017; 161:83-94. [PMID: 27105872 PMCID: PMC5195918 DOI: 10.1016/j.mad.2016.04.005] [Citation(s) in RCA: 67] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2016] [Revised: 04/05/2016] [Accepted: 04/15/2016] [Indexed: 02/06/2023]
Abstract
Alzheimer's disease (AD) is a progressive neurodegenerative disorder and the most common form of dementia. Autosomal dominant, familial AD (fAD) is very rare and caused by mutations in amyloid precursor protein (APP), presenilin-1 (PSEN-1), and presenilin-2 (PSEN-2) genes. The pathogenesis of sporadic AD (sAD) is more complex and variants of several genes are associated with an increased lifetime risk of AD. Nuclear and mitochondrial DNA integrity is pivotal during neuronal development, maintenance and function. DNA damage and alterations in cellular DNA repair capacity have been implicated in the aging process and in age-associated neurodegenerative diseases, including AD. These findings are supported by research using animal models of AD and in DNA repair deficient animal models. In recent years, novel mechanisms linking DNA damage to neuronal dysfunction have been identified and have led to the development of noninvasive treatment strategies. Further investigations into the molecular mechanisms connecting DNA damage to AD pathology may help to develop novel treatment strategies for this debilitating disease. Here we provide an overview of the role of genome instability and DNA repair deficiency in AD pathology and discuss research strategies that include genome instability as a component.
Collapse
Affiliation(s)
- Yujun Hou
- Laboratory of Molecular Gerontology, National Institute on Aging, NIH, Baltimore, MD 21224, USA
| | - Hyundong Song
- Laboratory of Molecular Gerontology, National Institute on Aging, NIH, Baltimore, MD 21224, USA
| | - Deborah L Croteau
- Laboratory of Molecular Gerontology, National Institute on Aging, NIH, Baltimore, MD 21224, USA
| | - Mansour Akbari
- Center for Healthy Aging, SUND, University of Copenhagen, Denmark
| | - Vilhelm A Bohr
- Laboratory of Molecular Gerontology, National Institute on Aging, NIH, Baltimore, MD 21224, USA.
| |
Collapse
|
16
|
Enhanced Neuroplasticity by the Metabolic Enhancer Piracetam Associated with Improved Mitochondrial Dynamics and Altered Permeability Transition Pore Function. Neural Plast 2016; 2016:8075903. [PMID: 27747106 PMCID: PMC5056292 DOI: 10.1155/2016/8075903] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2016] [Accepted: 08/01/2016] [Indexed: 02/07/2023] Open
Abstract
The mitochondrial cascade hypothesis of dementia assumes mitochondrial dysfunction leading to reduced energy supply, impaired neuroplasticity, and finally cell death as one major pathomechanism underlying the continuum from brain aging over mild cognitive impairment to initial and advanced late onset Alzheimer's disease. Accordingly, improving mitochondrial function has become an important strategy to treat the early stages of this continuum. The metabolic enhancer piracetam has been proposed as possible prototype for those compounds by increasing impaired mitochondrial function and related aspects like mechanisms of neuroplasticity. We here report that piracetam at therapeutically relevant concentrations improves neuritogenesis in the human cell line SH-SY5Y over conditions mirroring the whole spectrum of age-associated cognitive decline. These effects go parallel with improvement of impaired mitochondrial dynamics shifting back fission and fusion balance to the energetically more favorable fusion site. Impaired fission and fusion balance can also be induced by a reduction of the mitochondrial permeability transition pore (mPTP) function as atractyloside which indicates the mPTP has similar effects on mitochondrial dynamics. These changes are also reduced by piracetam. These findings suggest the mPTP as an important target for the beneficial effects of piracetam on mitochondrial function.
Collapse
|
17
|
Laßek M, Weingarten J, Wegner M, Volknandt W. The Amyloid Precursor Protein-A Novel Player within the Molecular Array of Presynaptic Nanomachines. Front Synaptic Neurosci 2016; 7:21. [PMID: 26834621 PMCID: PMC4719097 DOI: 10.3389/fnsyn.2015.00021] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2015] [Accepted: 12/24/2015] [Indexed: 12/27/2022] Open
Abstract
More than 20 years ago the amyloid precursor protein (APP) was identified as the precursor protein of the Aβ peptide, the main component of senile plaques in brains affected by Alzheimer’s disease (AD). The pathophysiology of AD, characterized by a massive loss of synapses, cognitive decline, and behavioral changes was in principle attributed to the accumulation of Aβ. Within the last decades, much effort has gone into understanding the molecular basis of the progression of AD. However, little is known about the actual physiological function of APPs. Allocating APP to the proteome of the structurally and functionally dynamic presynaptic active zone (PAZ) highlights APP as a hitherto unknown player within the setting of the presynapse. The molecular array of presynaptic nanomachines comprising the life cycle of synaptic vesicles, exo- and endocytosis, cytoskeletal rearrangements, and mitochondrial activity provides a balance between structural and functional maintenance and diversity. The generation of genetically designed mouse models further deciphered APP as an essential player in synapse formation and plasticity. Deletion of APP causes an age-dependent phenotype: while younger mice revealed almost no physiological impairments, this condition was changed in the elderly mice. Interestingly, the proteomic composition of neurotransmitter release sites already revealed substantial changes at young age. These changes point to a network that incorporates APP into a cluster of nanomachines. Currently, the underlying mechanism of how APP acts within these machines is still elusive. Within the scope of this review, we shall construct a network of APP interaction partners within the PAZ. Furthermore, we intend to outline how deletion of APP affects this network during space and time leading to impairments in learning and memory. These alterations may provide a molecular link to the pathogenesis of AD and the physiological function of APP in the central nervous system.
Collapse
Affiliation(s)
- Melanie Laßek
- Department of Molecular and Cellular Neurobiology, Goethe University Frankfurt Frankfurt am Main, Germany
| | - Jens Weingarten
- Department of Molecular and Cellular Neurobiology, Goethe University Frankfurt Frankfurt am Main, Germany
| | - Martin Wegner
- Department of Molecular Bioinformatics, Goethe University Frankfurt Frankfurt am Main, Germany
| | - Walter Volknandt
- Department of Molecular and Cellular Neurobiology, Goethe University Frankfurt Frankfurt am Main, Germany
| |
Collapse
|
18
|
Han SH, Mook-Jung I. Diverse molecular targets for therapeutic strategies in Alzheimer's disease. J Korean Med Sci 2014; 29:893-902. [PMID: 25045220 PMCID: PMC4101776 DOI: 10.3346/jkms.2014.29.7.893] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/27/2014] [Accepted: 06/12/2014] [Indexed: 11/20/2022] Open
Abstract
Alzheimer's disease (AD) is the most common form of dementia caused by neurodegenerative process and is tightly related to amyloid β (Aβ) and neurofibrillary tangles. The lack of early diagnostic biomarker and therapeutic remedy hinders the prevention of increasing population of AD patients every year. In spite of accumulated scientific information, numerous clinical trials for candidate drug targets have failed to be preceded into therapeutic development, therefore, AD-related sufferers including patients and caregivers, are desperate to seek the solution. Also, effective AD intervention is desperately needed to reduce AD-related societal threats to public health. In this review, we summarize various drug targets and strategies in recent preclinical studies and clinical trials for AD therapy: Allopathic treatment, immunotherapy, Aβ production/aggregation modulator, tau-targeting therapy and metabolic targeting. Some has already failed in their clinical trials and the others are still in various stages of investigations, both of which give us valuable information for future research in AD therapeutic development.
Collapse
Affiliation(s)
- Sun-Ho Han
- Department of Biochemistry and Biomedical Sciences, Seoul National University, College of Medicine, Seoul, Korea
| | - Inhee Mook-Jung
- Department of Biochemistry and Biomedical Sciences, Seoul National University, College of Medicine, Seoul, Korea
| |
Collapse
|
19
|
Singh GK, Rai G, Chatterjee SS, Kumar V. Effects of ethanolic extract of Fumaria indica L. on rat cognitive dysfunctions. Ayu 2013; 34:421-9. [PMID: 24696581 PMCID: PMC3968708 DOI: 10.4103/0974-8520.127727] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/04/2022] Open
Abstract
Fumaria indica L. in Ayurveda is known as Parpat and traditionally used to calm the brain. Due to lack of scientific validation, 50% ethanolic extract of F. indica L. (FI) was evaluated for putative cognitive function modulating effects. Suspension of FI in 0.3% carboxymethyl cellulose (CMC) was orally administered to rats during the entire experimental period of 16 days at dose levels of 100, 200, and 400 mg/kg/day. Piracetam was used as standard nootropic. Behavioral models of learning and memory used were modified elevated plus-maze (M-EPM) and passive avoidance (PA) tests. Scopolamine (I mg/kg, s.c.), sodium nitrite (25 mg/kg, i.p.), and electroconvulsive shock (150 mA, 0.2 sec) were used to induce amnesia. Acetylcholinesterase (AChE) activity, muscarinic receptor density, oxidative status, and cytokine expressions [tumor necrosis factor alpha (TNF-α), interleukin (IL)-1β, and IL-10] were also assessed. Piracetam (500 mg/kg/day)-like memory-enhancing and anti-amnesic activity of the extract was observed. FI showed dose-dependent decrease in brain AChE activity and increase in muscarinic receptor density, and such was also the case for its observed beneficial effects on the brain antioxidative status. FI also inhibited the scopolamine-induced overexpression of the three tested cytokines observed in rat's brain. FI possesses nootropic-like beneficial effects on cognitive functions.
Collapse
Affiliation(s)
- Gireesh Kumar Singh
- Ph.D. Scholar, Department of Pharmaceutics, Neuropharmacology Research Laboratory, Indian Institute of Technology, Varanasi, India
| | - Geeta Rai
- Assistant Professor, Department of Molecular and Human Genetics, Faculty of Science, Banaras Hindu University, Varanasi, Uttar Pradesh, India
| | - Shyam Sunder Chatterjee
- Retired Head, Pharmacology Labs, Willmar Schwabe, Stettiner Str. 1, D-76138 Karlsruhe, Germany
| | - Vikas Kumar
- Associate Professor, Department of Pharmaceutics, Neuropharmacology Research Laboratory, Indian Institute of Technology, Banaras Hindu University, Varanasi, Uttar Pradesh, India
| |
Collapse
|
20
|
Marisco PC, Carvalho FB, Rosa MM, Girardi BA, Gutierres JM, Jaques JAS, Salla APS, Pimentel VC, Schetinger MRC, Leal DBR, Mello CF, Rubin MA. Piracetam prevents scopolamine-induced memory impairment and decrease of NTPDase, 5'-nucleotidase and adenosine deaminase activities. Neurochem Res 2013; 38:1704-14. [PMID: 23677777 DOI: 10.1007/s11064-013-1072-6] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2012] [Revised: 04/29/2013] [Accepted: 05/07/2013] [Indexed: 11/26/2022]
Abstract
Piracetam improves cognitive function in animals and in human beings, but its mechanism of action is still not completely known. In the present study, we investigated whether enzymes involved in extracellular adenine nucleotide metabolism, adenosine triphosphate diphosphohydrolase (NTPDase), 5'-nucleotidase and adenosine deaminase (ADA) are affected by piracetam in the hippocampus and cerebral cortex of animals subjected to scopolamine-induced memory impairment. Piracetam (0.02 μmol/5 μL, intracerebroventricular, 60 min pre-training) prevented memory impairment induced by scopolamine (1 mg/kg, intraperitoneal, immediately post-training) in the inhibitory avoidance learning and in the object recognition task. Scopolamine reduced the activity of NTPDase in hippocampus (53 % for ATP and 53 % for ADP hydrolysis) and cerebral cortex (28 % for ATP hydrolysis). Scopolamine also decreased the activity of 5'-nucleotidase (43 %) and ADA (91 %) in hippocampus. The same effect was observed in the cerebral cortex for 5'-nucleotidase (38 %) and ADA (68 %) activities. Piracetam fully prevented scopolamine-induced memory impairment and decrease of NTPDase, 5'-nucleotidase and adenosine deaminase activities in synaptosomes from cerebral cortex and hippocampus. In vitro experiments show that piracetam and scopolamine did not alter enzymatic activity in cerebral cortex synaptosomes. Moreover, piracetam prevented scopolamine-induced increase of TBARS levels in hippocampus and cerebral cortex. These results suggest that piracetam-induced improvement of memory is associated with protection against oxidative stress and maintenance of NTPDase, 5'-nucleotidase and ADA activities, and suggest the purinergic system as a putative target of piracetam.
Collapse
Affiliation(s)
- Patricia C Marisco
- Graduation Program in Pharmacology, Center of Health Sciences (CCS), Federal University of Santa Maria, Santa Maria, RS, CEP 97105-900, Brazil
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
21
|
Abdel-Salam OM, Salem NA, El-Sayed El-Shamarka M, Al-Said Ahmed N, Seid Hussein J, El-Khyat ZA. Cannabis-induced impairment of learning and memory: effect of different nootropic drugs. EXCLI JOURNAL 2013; 12:193-214. [PMID: 26417227 PMCID: PMC4552130] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/13/2013] [Accepted: 02/27/2013] [Indexed: 11/09/2022]
Abstract
Cannabis sativa preparations are the most commonly used illicit drugs worldwide. The present study aimed to investigate the effect of Cannabis sativa extract in the working memory version of the Morris water maze (MWM; Morris, 1984[43]) test and determine the effect of standard memory enhancing drugs. Cannabis sativa was given at doses of 5, 10 or 20 mg/kg (expressed as Δ(9)-tetrahydrocannabinol) alone or co-administered with donepezil (1 mg/kg), piracetam (150 mg/ kg), vinpocetine (1.5 mg/kg) or ginkgo biloba (25 mg/kg) once daily subcutaneously (s.c.) for one month. Mice were examined three times weekly for their ability to locate a submerged platform. Mice were euthanized 30 days after starting cannabis injection when biochemical assays were carried out. Malondialdehyde (MDA), reduced glutathione (GSH), nitric oxide, glucose and brain monoamines were determined. Cannabis resulted in a significant increase in the time taken to locate the platform and enhanced the memory impairment produced by scopolamine. This effect of cannabis decreased by memory enhancing drugs with piracetam resulting in the most-shorter latency compared with the cannabis. Biochemically, cannabis altered the oxidative status of the brain with decreased MDA, increased GSH, but decreased nitric oxide and glucose. In cannabis-treated rats, the level of GSH in brain was increased after vinpocetine and donepezil and was markedly elevated after Ginkgo biloba. Piracetam restored the decrease in glucose and nitric oxide by cannabis. Cannabis caused dose-dependent increases of brain serotonin, noradrenaline and dopamine. After cannabis treatment, noradrenaline is restored to its normal value by donepezil, vinpocetine or Ginkgo biloba, but increased by piracetam. The level of dopamine was significantly reduced by piracetam, vinpocetine or Ginkgo biloba. These data indicate that cannabis administration is associated with impaired memory performance which is likely to involve decreased brain glucose availability as well as alterations in brain monoamine neurotransmitter levels. Piracetam is more effective in ameliorating the cognitive impairments than other nootropics by alleviating the alterations in glucose, nitric oxide and dopamine in brain.
Collapse
Affiliation(s)
| | - Neveen A. Salem
- Department of Toxicology and Narcotics, National Research Centre, Cairo
| | | | | | | | | |
Collapse
|