1
|
Suryawanshi MV, Gujarathi PP, Mulla T, Bagban I. Hypericum perforatum: a comprehensive review on pharmacognosy, preclinical studies, putative molecular mechanism, and clinical studies in neurodegenerative diseases. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2024; 397:3803-3818. [PMID: 38175276 DOI: 10.1007/s00210-023-02915-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/17/2023] [Accepted: 12/20/2023] [Indexed: 01/05/2024]
Abstract
The herb Hypericum perforatum, also referred to as St. John's wort, has drawn a lot of interest because of its potential therapeutic benefits in treating neurodegenerative illnesses. Due to the absence of effective therapies, illnesses like Alzheimer's and Parkinson's disease pose an increasing worldwide health concern. Because of its wide variety of phytochemicals, especially hyperforin, and hypericin, Hypericum perforatum is well known for its neuroprotective properties. These substances have proven to be able to affect different cellular processes linked to neurodegeneration. They can act as anti-inflammatory, antioxidant, and neurotransmitter system regulators, which may help halt neurodegenerative illnesses' progression. The use of Hypericum perforatum extracts and its contents has shown encouraging results in research on animal models of neurodegenerative disorders. These advantages include higher nerve cell survival, lowered oxidative stress, and higher cognitive performance. Underscoring its versatile potential to combat neurodegeneration, Hypericum perforatum has neuroprotective mechanisms that modulate neuroinflammation and prevent apoptotic pathways. In conclusion, Hypericum perforatum shows tremendous promise as a potential treatment for neurological illnesses due to its wide variety of phytochemicals. To completely comprehend its specific mechanisms of action and turn these discoveries into efficient clinical therapies, additional research is needed. Investigating Hypericum perforatum's function in neurodegenerative disorders may present new opportunities for the advancement of ground-breaking therapeutic strategies.
Collapse
Affiliation(s)
- Meghraj Vivekanand Suryawanshi
- School of Pharmaceutical Sciences, Jaipur National University, Jaipur, Rajasthan, 302017, India
- Department of Pharmaceutics and Pharmaceutical Technology, Krishna School of Pharmacy and Research, Drs. Kiran and Pallavi Patel Global University, Varnama, Vadodara, Gujarat, 391240, India
- AllWell Neuritech LLP, Dharngaon, Maharashtra, 425105, India
| | - Pranjal P Gujarathi
- Department of Pharmacology, Vidhyadeep Institute of Pharmacy, Vidhyadeep University, Anita, Kim, Surat, Gujarat, 394110, India.
- Centre for Advance Research, Bhagwan Mahavir College of Pharmacy, Bhagwan Mahavir University, Vesu, Surat, Gujarat, 395007, India.
| | - Taufik Mulla
- Department of Pharmaceutics and Pharmaceutical Technology, Krishna School of Pharmacy and Research, Drs. Kiran and Pallavi Patel Global University, Varnama, Vadodara, Gujarat, 391240, India
| | - Imtiyaz Bagban
- Department of Pharmacology, Krishna School of Pharmacy and Research, Drs. Kiran and Pallavi Patel Global University, Varnama, Vadodara, Gujarat, 391240, India
| |
Collapse
|
2
|
Bouron A. Cellular neurobiology of hyperforin. Phytother Res 2024; 38:636-645. [PMID: 37963759 DOI: 10.1002/ptr.8063] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2023] [Revised: 10/16/2023] [Accepted: 10/22/2023] [Indexed: 11/16/2023]
Abstract
Hyperforin is a phloroglucinol derivative isolated from the medicinal plant Hypericum perforatum (St John's wort, SJW). This lipophilic biomolecule displays antibacterial, pro-apoptotic, antiproliferative, and anti-inflammatory activities. In addition, in vitro and in vivo data showed that hyperforin is a promising molecule with potential applications in neurology and psychiatry. For instance, hyperforin possesses antidepressant properties, impairs the uptake of neurotransmitters, and stimulates the brain derived neurotrophic factor (BDNF)/TrkB neurotrophic signaling pathway, the adult hippocampal neurogenesis, and the brain homeostasis of zinc. In fact, hyperforin is a multi-target biomolecule with a complex neuropharmacological profile. However, one prominent pharmacological feature of hyperforin is its ability to influence the homeostasis of cations such as Ca2+ , Na+ , Zn2+ , and H+ . So far, the pathophysiological relevance of these actions is currently unknown. The main objective of the present work is to provide an overview of the cellular neurobiology of hyperforin, with a special focus on its effects on neuronal membranes and the movement of cations.
Collapse
Affiliation(s)
- Alexandre Bouron
- Université Grenoble Alpes, CNRS, CEA, Inserm UA13 BGE, Grenoble, France
| |
Collapse
|
3
|
Shehata NI, Abd EL-Salam DM, Hussein RM, Rizk SM. Effect of safranal or candesartan on 3-nitropropionicacid-induced biochemical, behavioral and histological alterations in a rat model of Huntington's disease. PLoS One 2023; 18:e0293660. [PMID: 37910529 PMCID: PMC10619823 DOI: 10.1371/journal.pone.0293660] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Accepted: 10/17/2023] [Indexed: 11/03/2023] Open
Abstract
3-nitropropionic acid (3-NP) is a potent mitochondrial inhibitor mycotoxin. Systemic administration of 3-NP can induce Huntington's disease (HD)-like symptoms in experimental animals. Safranal (Safr) that is found in saffron essential oil has antioxidant, anti-inflammatory and anti-apoptotic actions. Candesartan (Cands) is an angiotensin receptor blocker that has the potential to prevent cognitive deficits. The present study aims to investigate the potential neuroprotective efficacy of Safr or Cands in 3-NP-induced rat model of HD. The experiments continued for nine consecutive days. Rats were randomly assigned into seven groups. The first group (Safr-control) was daily intraperitoneally injected with paraffin oil. The second group (Cands- and 3-NP-control) daily received an oral dose of 0.5% carboxymethylcellulose followed by an intraperitoneal injection of 0.9% saline. The third and fourth groups received a single daily dose of 50 mg/kg Safr (intraperitoneal) and 1 mg/kg Cands (oral), respectively. The sixth group was daily treated with 50 mg Safr kg/day (intraperitoneal) and was intraperitoneally injected with 20 mg 3-NP/ kg, from the 3rd till the 9th day. The seventh group was daily treated with 1 mg Cands /kg/day (oral) and was intraperitoneally injected with 20 mg 3-NP/ kg, from the 3rd till the 9th day. The present results revealed that 3-NP injection induced a considerable body weight loss, impaired memory and locomotor activity, reduced striatal monoamine levels. Furthermore, 3-NP administration remarkably increased striatal malondialdehyde and nitric oxide levels, whereas markedly decreased the total antioxidant capacity. Moreover, 3-NP significantly upregulated the activities of inducible nitric oxide synthase and caspase-3 as well as the Fas ligand, in striatum. On the contrary, Safr and Cands remarkably alleviated the above-mentioned 3-NP-induced alterations. In conclusion, Safr and Cands may prevent or delay the progression of HD and its associated impairments through their antioxidant, anti-inflammatory, anti-apoptotic and neuromodulator effects.
Collapse
Affiliation(s)
| | | | | | - Sherine Maher Rizk
- Faculty of Pharmacy, Biochemistry Department, Cairo University, Cairo, Egypt
| |
Collapse
|
4
|
Li X, Li T, Zhan F, Cheng F, Lu L, Zhang B, Li J, Hu Z, Zhou S, Jia Y, Allen S, White L, Phillips J, Zhu Z, Xu J, Yao H. Design, Synthesis, and Biological Evaluation of Novel Chromanone Derivatives as Multifunctional Agents for the Treatment of Alzheimer's Disease. ACS Chem Neurosci 2022; 13:3488-3501. [PMID: 36383455 DOI: 10.1021/acschemneuro.2c00520] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Based on a multitarget strategy, a series of novel chromanone-1-benzyl-1,2,3,6-tetrahydropyridin hybrids were identified for the potential treatment of Alzheimer's disease (AD). Biological evaluation demonstrated that these hybrids exhibited significant inhibitory activities toward acetylcholinesterase (AChE) and monoamine oxidase B (MAO-B). The optimal compound C10 possessed excellent dual AChE/MAO-B inhibition both in terms of potency and equilibrium (AChE: IC50 = 0.58 ± 0.05 μM; MAO-B: IC50 = 0.41 ± 0.04 μM). Further molecular modeling and kinetic investigations revealed that compound C10 was a dual-binding inhibitor bound to both the catalytic anionic site and peripheral anionic site of AChE. In addition, compound C10 exhibited low neurotoxicity and potently inhibited AChE enzymatic activity. Furthermore, compound C10 more effectively protected against mitochondrial dysfunction and oxidation than donepezil, strongly inhibited AChE-induced amyloid aggregation, and moderately reduced glutaraldehyde-induced phosphorylation of tau protein in SH-SY5Y cells. Moreover, compound C10 displayed largely enhanced improvements in cognitive behaviors and spatial memory in a scopolamine-induced AD mice model with better efficacy than donepezil. Overall, the multifunctional profiles of compound C10 suggest that it deserves further investigation as a promising lead for the prospective treatment of AD.
Collapse
Affiliation(s)
- Xinnan Li
- State Key Laboratory of Natural Medicines and Department of Medicinal Chemistry, China Pharmaceutical University, 24 Tong Jia Xiang, Nanjing 210009, P. R. China
| | - Tiantian Li
- School of Pharmacy, The University of Nottingham, University Park Campus, Nottingham NG7 2RD, U.K
| | - Feiyan Zhan
- State Key Laboratory of Natural Medicines and Department of Medicinal Chemistry, China Pharmaceutical University, 24 Tong Jia Xiang, Nanjing 210009, P. R. China
| | - Feiyue Cheng
- School of Pharmacy, The University of Nottingham, University Park Campus, Nottingham NG7 2RD, U.K
| | - Li Lu
- School of Pharmacy, The University of Nottingham, University Park Campus, Nottingham NG7 2RD, U.K
| | - Bocheng Zhang
- School of Pharmacy, The University of Nottingham, University Park Campus, Nottingham NG7 2RD, U.K
| | - Junda Li
- State Key Laboratory of Natural Medicines and Department of Medicinal Chemistry, China Pharmaceutical University, 24 Tong Jia Xiang, Nanjing 210009, P. R. China
| | - Zhaoxin Hu
- State Key Laboratory of Natural Medicines and Department of Medicinal Chemistry, China Pharmaceutical University, 24 Tong Jia Xiang, Nanjing 210009, P. R. China
| | - Shengnan Zhou
- State Key Laboratory of Natural Medicines and Department of Medicinal Chemistry, China Pharmaceutical University, 24 Tong Jia Xiang, Nanjing 210009, P. R. China
| | - Yilin Jia
- State Key Laboratory of Natural Medicines and Department of Medicinal Chemistry, China Pharmaceutical University, 24 Tong Jia Xiang, Nanjing 210009, P. R. China
| | - Stephanie Allen
- School of Pharmacy, The University of Nottingham, University Park Campus, Nottingham NG7 2RD, U.K
| | - Lisa White
- School of Pharmacy, The University of Nottingham, University Park Campus, Nottingham NG7 2RD, U.K
| | - James Phillips
- School of Pharmacy, University of College London, London WC1N 1AX, U.K
| | - Zheying Zhu
- School of Pharmacy, The University of Nottingham, University Park Campus, Nottingham NG7 2RD, U.K
| | - Jinyi Xu
- State Key Laboratory of Natural Medicines and Department of Medicinal Chemistry, China Pharmaceutical University, 24 Tong Jia Xiang, Nanjing 210009, P. R. China
| | - Hequan Yao
- State Key Laboratory of Natural Medicines and Department of Medicinal Chemistry, China Pharmaceutical University, 24 Tong Jia Xiang, Nanjing 210009, P. R. China
| |
Collapse
|
5
|
Nayeri T, Sarvi S, Sharif M, Daryani A. Toxoplasma gondii: A possible etiologic agent for Alzheimer's disease. Heliyon 2021; 7:e07151. [PMID: 34141920 PMCID: PMC8187970 DOI: 10.1016/j.heliyon.2021.e07151] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2021] [Revised: 04/03/2021] [Accepted: 05/24/2021] [Indexed: 01/03/2023] Open
Abstract
Toxoplasma gondii (T. gondii) is one of the most pervasive neurotropic pathogens causing different lesions in a wide variety of mammals as intermediate hosts, including humans. It is estimated that one-third of the world population is infected with T. gondii; however, for a long time, there has been much interest in the examination of the possible role of this parasite in the development of mental disorders, such as Alzheimer's disease (AD). T. gondii may play a role in the progression of AD using mechanisms, such as the induction of the host's immune responses, inflammation of the central nervous system (CNS), alteration in the levels of neurotransmitters, and activation of indoleamine-2,3-dyoxigenase. This paper presents an appraisal of the literature, reports, and studies that seek to the possible role of T. gondii in the development of AD. For achieving the purpose of the current study, a search of six English databases (PubMed, ScienceDirect, Web of Science, Scopus, ProQuest, and Google Scholar) was performed. The results support the involvement of T. gondii in the induction and development of AD. Indeed, T. gondii can be considered a risk factor for the development of AD and requires the special attention of specialists and patients. Furthermore, the results of this study may contribute to prevent or delay the progress of AD worldwide. Therefore, it is required to carry out further studies in order to better perceive the parasitic mechanisms in the progression of AD.
Collapse
Affiliation(s)
- Tooran Nayeri
- Toxoplasmosis Research Center, Mazandaran University of Medical Sciences, Sari, Iran
- Department of Parasitology, School of Medicine, Mazandaran University of Medical Sciences, Sari, Iran
- Student Research Committee, Mazandaran University of Medical Sciences, Sari, Iran
| | - Shahabeddin Sarvi
- Toxoplasmosis Research Center, Mazandaran University of Medical Sciences, Sari, Iran
- Department of Parasitology, School of Medicine, Mazandaran University of Medical Sciences, Sari, Iran
| | - Mehdi Sharif
- Toxoplasmosis Research Center, Mazandaran University of Medical Sciences, Sari, Iran
- Department of Parasitology, School of Medicine, Mazandaran University of Medical Sciences, Sari, Iran
| | - Ahmad Daryani
- Toxoplasmosis Research Center, Mazandaran University of Medical Sciences, Sari, Iran
- Department of Parasitology, School of Medicine, Mazandaran University of Medical Sciences, Sari, Iran
| |
Collapse
|
6
|
Castro-Silva ES, Bello M, Rosales-Hernández MC, Correa-Basurto J, Hernández-Rodríguez M, Villalobos-Acosta D, Méndez-Méndez JV, Estrada-Pérez A, Murillo-Álvarez J, Muñoz-Ochoa M. Fucosterol from Sargassum horridum as an amyloid-beta (Aβ 1-42) aggregation inhibitor: in vitro and in silico studies. J Biomol Struct Dyn 2021; 39:1271-1283. [PMID: 32159448 DOI: 10.1080/07391102.2020.1729863] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2019] [Accepted: 02/07/2020] [Indexed: 10/25/2022]
Abstract
The number of patients diagnosed with Alzheimer's disease (AD) increases each year, and there are currently few treatment strategies to decrease the symptoms of AD; furthermore, these strategies are not sufficient to reduce memory loss in AD patients. In this work, in vitro and in silico studies were performed to evaluate the effects of fucosterol, which was extracted from an algal source and characterized by liquid chromatography-mass spectra (LC-MS), as an inhibitor of Aβ1-42 aggregation. Experimental studies, including protein gel electrophoresis, atomic force microscopy and fluorescence studies with thioflavin T (ThT), highlighted that fucosterol can decrease oligomer formation more than galantamine, which was used as a positive control. Docking and molecular dynamics simulations coupled with an MMGBSA approach showed that fucosterol is capable of recognizing the hydrophobic regions of monomeric Aβ1-42, suggesting that fucosterol could affect amyloid-beta (Aβ1-42) aggregation by preventing the formation of oligomers, preventing the development of AD.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Elena Sthephanie Castro-Silva
- Laboratorio de Química de Macroalgas, Centro Interdisciplinario de Ciencias Marinas, Instituto Politécnico Nacional. Av. Instituto Politécnico, La Paz, B.C.S. México
| | - Martiniano Bello
- Laboratorio de Diseño y Desarrollo de Nuevos Fármacos e Innovación Biotecnológica (Laboratory for the Design and Development of New Drugs and Biotechnological Innovation), Escuela Superior de Medicina, Instituto Politécnico Nacional, Ciudad de México, México
| | - Martha Cecilia Rosales-Hernández
- Laboratorio de Biofísica y Biocatálisis, Sección de Estudios de Posgrado e Investigación, Escuela Superior de Medicina, Instituto Politécnico Nacional, Ciudad de México, Mexico
| | - José Correa-Basurto
- Laboratorio de Diseño y Desarrollo de Nuevos Fármacos e Innovación Biotecnológica (Laboratory for the Design and Development of New Drugs and Biotechnological Innovation), Escuela Superior de Medicina, Instituto Politécnico Nacional, Ciudad de México, México
| | - Maricarmen Hernández-Rodríguez
- Laboratorio de Biofísica y Biocatálisis, Sección de Estudios de Posgrado e Investigación, Escuela Superior de Medicina, Instituto Politécnico Nacional, Ciudad de México, Mexico
| | - Daniel Villalobos-Acosta
- Laboratorio de Biofísica y Biocatálisis, Sección de Estudios de Posgrado e Investigación, Escuela Superior de Medicina, Instituto Politécnico Nacional, Ciudad de México, Mexico
| | - Juan Vicente Méndez-Méndez
- Instituto Politécnico Nacional, Centro de Nanociencias y Micro y Nanotecnologías, Ciudad de México, Mexico
| | - Alan Estrada-Pérez
- Laboratorio de Diseño y Desarrollo de Nuevos Fármacos e Innovación Biotecnológica (Laboratory for the Design and Development of New Drugs and Biotechnological Innovation), Escuela Superior de Medicina, Instituto Politécnico Nacional, Ciudad de México, México
| | - Jesus Murillo-Álvarez
- Laboratorio de Química de Macroalgas, Centro Interdisciplinario de Ciencias Marinas, Instituto Politécnico Nacional. Av. Instituto Politécnico, La Paz, B.C.S. México
| | - Mauricio Muñoz-Ochoa
- Laboratorio de Química de Macroalgas, Centro Interdisciplinario de Ciencias Marinas, Instituto Politécnico Nacional. Av. Instituto Politécnico, La Paz, B.C.S. México
| |
Collapse
|
7
|
Novelli M, Masiello P, Beffy P, Menegazzi M. Protective Role of St. John's Wort and Its Components Hyperforin and Hypericin against Diabetes through Inhibition of Inflammatory Signaling: Evidence from In Vitro and In Vivo Studies. Int J Mol Sci 2020; 21:E8108. [PMID: 33143088 PMCID: PMC7662691 DOI: 10.3390/ijms21218108] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Revised: 10/26/2020] [Accepted: 10/27/2020] [Indexed: 02/06/2023] Open
Abstract
Diabetes mellitus is a very common chronic disease with progressively increasing prevalence. Besides the well-known autoimmune and inflammatory pathogenesis of type 1 diabetes, in many people, metabolic changes and inappropriate lifestyle favor a subtle chronic inflammatory state that contributes to development of insulin resistance and progressive loss of β-cell function and mass, eventually resulting in metabolic syndrome or overt type 2 diabetes. In this paper, we review the anti-inflammatory effects of the extract of Hypericum perforatum L. (St. John's wort, SJW) and its main active ingredients firstly in representative pathological situations on inflammatory basis and then in pancreatic β cells and in obese or diabetic animal models. The simultaneous and long-lasting inhibition of signal transducer and activator of transcription (STAT)-1, nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB) and mitogen-activated protein kinases (MAPKs)/c-jun N-terminal kinase (JNK) signaling pathways involved in pro-inflammatory cytokine-induced β-cell dysfunction/death and insulin resistance make SJW particularly suitable for both preventive and therapeutic use in metabolic diseases. Hindrance of inflammatory cytokine signaling is likely dependent on the hyperforin content of SJW extract, but recent data reveal that hypericin can also exert relevant protective effects, mediated by activation of the cyclic adenosine monophosphate (cAMP)/protein kinase cAMP-dependent (PKA)/adenosine monophosphate activated protein kinase (AMPK) pathway, against high-fat-diet-induced metabolic abnormalities. Actually, the mechanisms of action of the two main components of SJW appear complementary, strengthening the efficacy of the plant extract. Careful quantitative analysis of SJW components and suitable dosage, with monitoring of possible drug-drug interaction in a context of remarkable tolerability, are easily achievable pre-requisites for forthcoming clinical applications.
Collapse
Affiliation(s)
- Michela Novelli
- Department of Translational Research and New Technologies in Medicine and Surgery, School of Medicine, University of Pisa, 56126 Pisa, Italy
| | - Pellegrino Masiello
- Department of Translational Research and New Technologies in Medicine and Surgery, School of Medicine, University of Pisa, 56126 Pisa, Italy
| | - Pascale Beffy
- Institute of Clinical Physiology, CNR, 56124 Pisa, Italy;
| | - Marta Menegazzi
- Department of Neuroscience, Biomedicine and Movement Sciences, Biochemistry Section, School of Medicine, University of Verona, 37134 Verona, Italy;
| |
Collapse
|
8
|
Is Toxoplasma gondii a potential risk factor for Alzheimer's disease? A systematic review and meta-analysis. Microb Pathog 2019; 137:103751. [PMID: 31536800 DOI: 10.1016/j.micpath.2019.103751] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2019] [Revised: 09/15/2019] [Accepted: 09/16/2019] [Indexed: 02/07/2023]
Abstract
Toxoplasmosis is a major public health concern due to neurotropic nature and role in the development of mental and behavioral disorders. Alzheimer's disease (AD) is an important nervous disease that results in the reduction of the amount of beta-amyloid plaque deposition and irreversible loss of neurons in the brain. Although a few studies evaluated the association between AD and toxoplasmosis, the present study as a systematic review and meta-analysis of published studies investigated the possible association between Toxoplasma gondii (T. gondii) and AD. A systematic literature search was conducted using seven electronic databases from the inception to 25th of November 2018 with no restriction of language that looked at toxoplasmosis (as an exposure) and AD (as a disease). The random effect model was used to determine the total odds ratio (OR) and total p-value. Generally, eight studies containing 3239 subjects (360 patients and 2879 controls) met the eligibility criteria. Then, eight articles were used for meta-analysis with respect to inclusion and exclusion criteria. The results of the meta-analysis (random effect model) showed a common OR of 1.53 (95% CI: 1.07-2.18). Despite the fact that there was no evidence of publication bias (P = 0.079) using formal statistical test, the visual inspection of the funnel graph suggested that the observed effect was fueled mainly by three studies with large effects (and large standard errors). Moreover, the file-drawer effect (i.e. publishing mainly studies with positive results) might play a role in the phenomenon. Results of this meta-analytic study suggested that T. gondii can be considered a risk factor for the development of AD and exacerbation of its symptoms. However, the number of published relevant studies is still relatively low, and the risk of the presence of publication bias is relatively high. Therefore, the investigation of the clinically important question of the possible association between toxoplasmosis and AD definitively deserves further attention.
Collapse
|
9
|
Andrade-Jorge E, Bribiesca-Carlos J, Martínez-Martínez FJ, Soriano-Ursúa MA, Padilla-Martínez II, Trujillo-Ferrara JG. Crystal structure, DFT calculations and evaluation of 2-(2-(3,4-dimethoxyphenyl)ethyl)isoindoline-1,3-dione as AChE inhibitor. Chem Cent J 2018; 12:74. [PMID: 29938351 PMCID: PMC6020091 DOI: 10.1186/s13065-018-0442-1] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2018] [Accepted: 06/19/2018] [Indexed: 02/07/2023] Open
Abstract
Dioxoisoindolines have been included as a pharmacophore group in diverse drug-like molecules with a wide range of biological activity. Various reports have shown that phthalimide derivatives are potent inhibitors of AChE, a key enzyme involved in the deterioration of the cholinergic system during the development of Alzheimer's disease. In the present study, 2-(2-(3,4-dimethoxyphenyl)ethyl)isoindoline-1,3-dione was synthesized, crystallized and evaluated as an AChE inhibitor. The geometric structure of the crystal and the theoretical compound (from molecular modeling) were analyzed and compared, finding a close correlation. The formation of the C6-H6···O19 interaction could be responsible for the non-negligible out of phenyl plane deviation of the C19 methoxy group, the O3 from the carbonyl group lead to C16-H16···O3i intermolecular interactions to furnish C(9) and C(14) infinite chains within the (- 4 0 9) and (- 3 1 1) families of planes. Finally, the biological experiments reveal that the isoindoline-1,3-dione exerts a good competitive inhibition on AChE (Ki = 0.33-0.93 mM; 95% confidence interval) and has very low acute toxicity (LD50 > 1600 mg/kg) compared to the AChE inhibitors currently approved for clinical use.
Collapse
Affiliation(s)
- Erik Andrade-Jorge
- Laboratorio de Investigación en Bioquímica, Sección de Estudios de Posgrado e Investigación, Escuela Superior de Medicina del Instituto Politécnico Nacional, Plan de San Luis y Díaz Mirón s/n Casco de Santo Tomás, 11340 Mexico City, Mexico
| | - José Bribiesca-Carlos
- Laboratorio de Investigación en Bioquímica, Sección de Estudios de Posgrado e Investigación, Escuela Superior de Medicina del Instituto Politécnico Nacional, Plan de San Luis y Díaz Mirón s/n Casco de Santo Tomás, 11340 Mexico City, Mexico
| | - Francisco J. Martínez-Martínez
- Facultad de Ciencias Químicas, Universidad de Colima, Km. 9 Carretera Colima-Coquimatlán, C.P. 28400 Coquimatlán, Colima Mexico
| | - Marvin A. Soriano-Ursúa
- Laboratorio de Investigación en Fisiología, Sección de Estudios de Posgrado e Investigación, Escuela Superior de Medicina del Instituto Politécnico Nacional, Plan de San Luis y Díaz Mirón s/n Casco de Santo Tomás, 11340 Mexico City, Mexico
| | - Itzia I. Padilla-Martínez
- Laboratorio de Química Supramolecular y Nanociencias, Unidad Profesional Interdisciplinaria de Biotecnología del Instituto Politécnico Nacional, Av. Acueducto s/n Barrio la Laguna Ticomán, 07340 Mexico City, Mexico
| | - José G. Trujillo-Ferrara
- Laboratorio de Investigación en Bioquímica, Sección de Estudios de Posgrado e Investigación, Escuela Superior de Medicina del Instituto Politécnico Nacional, Plan de San Luis y Díaz Mirón s/n Casco de Santo Tomás, 11340 Mexico City, Mexico
| |
Collapse
|
10
|
Hofrichter J, Krohn M, Schumacher T, Lange C, Feistel B, Walbroel B, Pahnke J. Sideritis spp. Extracts Enhance Memory and Learning in Alzheimer's β-Amyloidosis Mouse Models and Aged C57Bl/6 Mice. J Alzheimers Dis 2018; 53:967-80. [PMID: 27258424 PMCID: PMC4981905 DOI: 10.3233/jad-160301] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Nowadays, Alzheimer’s disease is the most prevalent epiphenomenon of the aging population. Although soluble amyloid-β (Aβ) species (monomers, oligomers) are recognized triggers of the disease, no therapeutic approach is able to stop it. Herbal medicines are used to treat different diseases in many regions of the world. On the Balkan Peninsula, at the eastern Mediterranean Sea, and adjacent regions, Sideritis species are used as traditional medicine to prevent age-related problems in elderly. To evaluate this traditional knowledge in controlled experiments, we tested extracts of two commonly used Sideritis species, Sideritis euboea and Sideritis scardica, with regard to their effects on cognition in APP-transgenic and aged, non-transgenic C57Bl/6 mice. Additionally, histomorphological and biochemical changes associated with Aβ deposition and treatment were assessed. We found that daily oral treatment with Sideritis spp. extracts highly enhanced cognition in aged, non-transgenic as well as in APP-transgenic mice, an effect that was even more pronounced when extracts of both species were applied in combination. The treatment strongly reduced Aβ42 load in APP-transgenic mice, accompanied by increased phagocytic activity of microglia, and increased expression of the α-secretase ADAM10. Moreover, the treatment was able to fully rescue neuronal loss of APP-transgenic mice to normal levels as seen in non-transgenic controls. Having the traditional knowledge in mind, our results imply that treatment with Sideritis spp. extracts might be a potent, well-tolerated option for treating symptoms of cognitive impairment in elderly and with regard to Alzheimer’s disease by affecting its most prominent hallmarks: Aβ pathology and cognitive decline.
Collapse
Affiliation(s)
| | - Markus Krohn
- University of Oslo (UiO) and Oslo University Hospital (OUS), Department of Neuro-/Pathology, Translational Neurodegeneration Research and Neuropathology Lab, Oslo, Norway.,University of Rostock, Department of Neurology, Rostock, Germany
| | - Toni Schumacher
- University of Rostock, Department of Neurology, Rostock, Germany
| | - Cathleen Lange
- University of Rostock, Department of Neurology, Rostock, Germany
| | | | | | - Jens Pahnke
- University of Oslo (UiO) and Oslo University Hospital (OUS), Department of Neuro-/Pathology, Translational Neurodegeneration Research and Neuropathology Lab, Oslo, Norway.,University of Rostock, Department of Neurology, Rostock, Germany.,University of Lübeck, Lübeck (LIED), Lübeck, Germany.,Leibniz Institute for Plant Biochemistry (IPB), Halle, Germany
| |
Collapse
|
11
|
McHardy SF, Wang HYL, McCowen SV, Valdez MC. Recent advances in acetylcholinesterase Inhibitors and Reactivators: an update on the patent literature (2012-2015). Expert Opin Ther Pat 2017; 27:455-476. [PMID: 27967267 DOI: 10.1080/13543776.2017.1272571] [Citation(s) in RCA: 86] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
INTRODUCTION Acetylcholinesterase (AChE) is the major enzyme that hydrolyzes acetylcholine, a key neurotransmitter for synaptic transmission, into acetic acid and choline. Mild inhibition of AChE has been shown to have therapeutic relevance in Alzheimer's disease (AD), myasthenia gravis, and glaucoma among others. In contrast, strong inhibition of AChE can lead to cholinergic poisoning. To combat this, AChE reactivators have to be developed to remove the offending AChE inhibitor, restoring acetylcholine levels to normal. Areas covered: This article covers recent advances in the development of acetylcholinesterase modulators, including both inhibitors of acetylcholinesterase for the efforts in development of new chemical entities for treatment of AD, as well as re-activators for resurrection of organophosphate bound acetylcholinesterase. Expert opinion: Over the past three years, research efforts have continued to identify novel small molecules as AChE inhibitors for both CNS and peripheral diseases. The more recent patent activity has focused on three AChE ligand design areas: derivatives of known AChE ligands, natural product based scaffolds and multifunctional ligands, all of which have produced some unique chemical matter with AChE inhibition activities in the mid picomolar to low micromolar ranges. New AChE inhibitors with polypharmacology or dual inhibitory activity have also emerged as highlighted by new AChE inhibitors with dual activity at L-type calcium channels, GSK-3, BACE1 and H3, although most only show low micromolar activity, thus further research is warranted. New small molecule reactivators of organophosphate-inhibited AChE have also been disclosed, which focused on the design of neutral ligands with improved pharmaceutical properties and blood-brain barrier (BBB) penetration. Gratifyingly, some research in this area is moving away from the traditional quaternary pyridinium oximes AChE reactivators, while still employing the necessary reactivation group (oximes). However, selectivity over inhibition of native AChE enzyme, effectiveness of reactivation, broad-spectrum reactivation against multiple organophosphates and reactivation of aged-enzyme continue to be hurdles for this area of research.
Collapse
Affiliation(s)
- Stanton F McHardy
- a Center for Innovative Drug Discovery, Department of Chemistry , University of Texas San Antonio, One UTSA Circle , San Antonio , TX , USA
| | - Hua-Yu Leo Wang
- a Center for Innovative Drug Discovery, Department of Chemistry , University of Texas San Antonio, One UTSA Circle , San Antonio , TX , USA
| | - Shelby V McCowen
- a Center for Innovative Drug Discovery, Department of Chemistry , University of Texas San Antonio, One UTSA Circle , San Antonio , TX , USA
| | - Matthew C Valdez
- a Center for Innovative Drug Discovery, Department of Chemistry , University of Texas San Antonio, One UTSA Circle , San Antonio , TX , USA
| |
Collapse
|
12
|
Seroprevalence of Toxoplasma gondii Infection in Patients with Alzheimer’s Disease. ARCHIVES OF CLINICAL INFECTIOUS DISEASES 2016. [DOI: 10.5812/archcid.37205] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
|
13
|
Oliveira AI, Pinho C, Sarmento B, Dias ACP. Neuroprotective Activity of Hypericum perforatum and Its Major Components. FRONTIERS IN PLANT SCIENCE 2016; 7:1004. [PMID: 27462333 PMCID: PMC4939296 DOI: 10.3389/fpls.2016.01004] [Citation(s) in RCA: 62] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/15/2016] [Accepted: 06/27/2016] [Indexed: 05/15/2023]
Abstract
Hypericum perforatum is a perennial plant, with worldwide distribution, commonly known as St. John's wort. It has been used for centuries in traditional medicine for the treatment of several disorders, such as minor burns, anxiety, and mild to moderate depression. In the past years, its antidepressant properties have been extensively studied. Despite that, other H. perforatum biological activities, as its neuroprotective properties have also been evaluated. The present review aims to provide a comprehensive summary of the main biologically active compounds of H. perforatum, as for its chemistry, pharmacological activities, drug interactions and adverse reactions and gather scattered information about its neuroprotective abilities. As for this, it has been demonstrated that H. perforatum extracts and several of its major molecular components have the ability to protect against toxic insults, either directly, through neuroprotective mechanisms, or indirectly, through is antioxidant properties. H. perforatum has therefore the potential to become an effective neuroprotective therapeutic agent, despite further studies that need to be carried out.
Collapse
Affiliation(s)
- Ana I. Oliveira
- Nucleo de Investigação e Informação em Farmácia, Centro de Investigação em Saúde e Ambiente, Escola Superior de Tecnologia de Saúde do Porto – Instituto Politécnico do Porto, Vila Nova de GaiaPortugal
- Agrobioplant Group (CITAB-UM), Department of Biology, University of Minho, BragaPortugal
| | - Cláudia Pinho
- Nucleo de Investigação e Informação em Farmácia, Centro de Investigação em Saúde e Ambiente, Escola Superior de Tecnologia de Saúde do Porto – Instituto Politécnico do Porto, Vila Nova de GaiaPortugal
- Agrobioplant Group (CITAB-UM), Department of Biology, University of Minho, BragaPortugal
| | - Bruno Sarmento
- Cooperativa de Ensino Superior Politécnico e Universitário, Instituto de Investigação e Formação Avançada em Ciências e Tecnologias da Saúde, Gandra PRDPortugal
- Instituto de Investigação e Inovação em Saúde, PortoPortugal
- Instituto de Engenharia Biomédica, PortoPortugal
| | - Alberto C. P. Dias
- Agrobioplant Group (CITAB-UM), Department of Biology, University of Minho, BragaPortugal
- *Correspondence: Alberto C. P. Dias,
| |
Collapse
|
14
|
Asadi F, Jamshidi AH, Khodagholi F, Yans A, Azimi L, Faizi M, Vali L, Abdollahi M, Ghahremani MH, Sharifzadeh M. Reversal effects of crocin on amyloid β-induced memory deficit: Modification of autophagy or apoptosis markers. Pharmacol Biochem Behav 2015; 139:47-58. [DOI: 10.1016/j.pbb.2015.10.011] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/23/2015] [Revised: 09/25/2015] [Accepted: 10/16/2015] [Indexed: 12/29/2022]
|
15
|
Fracasso C, Bagnati R, Passoni A, Guiso G, Cantoni L, Riva A, Morazzoni P, Gobbi M. Brain Uptake of Tetrahydrohyperforin and Potential Metabolites after Repeated Dosing in Mice. JOURNAL OF NATURAL PRODUCTS 2015; 78:2029-2035. [PMID: 26287496 DOI: 10.1021/acs.jnatprod.5b00302] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/04/2023]
Abstract
Tetrahydrohyperforin (IDN-5706) is a semisynthetic derivative of hyperforin, one of the main active components of Hypericum perforatum extracts. It showed remarkable positive effects on memory and cognitive performances in wild-type mice and in a transgenic mouse model of Alzheimer's disease, but little was known about the concentrations it can reach in the brain. The investigations reported herein show that repeated treatment of mice with tetrahydrohyperforin (20 mg/kg intraperitoneally, twice daily for 4 days and once on the fifth day) results in measurable concentrations in the brain, up to 367 ng/g brain (∼700 nM) 6 h after the last dose; these concentrations have significant effects on synaptic function in hippocampal slices. The other main finding was the identification and semiquantitative analysis of tetrahydrohyperforin metabolites. In plasma, three hydroxylated/dehydrogenated metabolites were the largest (M1-3) and were also formed in vitro on incubation of tetrahydrohyperforin with mouse liver microsomes; the fourth metabolite in abundance was a hydroxylated/deisopropylated derivative (M13), which was not predicted in vitro. These metabolites were all detected in the brain, with peak areas from 10% (M1) to ∼1.5% (M2, M3, and M13) of the parent compound. In summary, repeated treatment of mice with tetrahydrohyperforin gave brain concentrations that might well underlie its central pharmacological effects. We also provide the first metabolic profile of this compound.
Collapse
Affiliation(s)
- Claudia Fracasso
- IRCCS - Istituto di Ricerche Farmacologiche "Mario Negri" , 20156 Milan, Italy
| | - Renzo Bagnati
- IRCCS - Istituto di Ricerche Farmacologiche "Mario Negri" , 20156 Milan, Italy
| | - Alice Passoni
- IRCCS - Istituto di Ricerche Farmacologiche "Mario Negri" , 20156 Milan, Italy
| | - Giovanna Guiso
- IRCCS - Istituto di Ricerche Farmacologiche "Mario Negri" , 20156 Milan, Italy
| | - Lavinia Cantoni
- IRCCS - Istituto di Ricerche Farmacologiche "Mario Negri" , 20156 Milan, Italy
| | | | | | - Marco Gobbi
- IRCCS - Istituto di Ricerche Farmacologiche "Mario Negri" , 20156 Milan, Italy
| |
Collapse
|
16
|
Inestrosa NC, Ríos JA, Cisternas P, Tapia-Rojas C, Rivera DS, Braidy N, Zolezzi JM, Godoy JA, Carvajal FJ, Ardiles AO, Bozinovic F, Palacios AG, Sachdev PS. Age Progression of Neuropathological Markers in the Brain of the Chilean Rodent Octodon degus, a Natural Model of Alzheimer's Disease. Brain Pathol 2015; 25:679-91. [PMID: 25351914 DOI: 10.1111/bpa.12226] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2014] [Accepted: 10/21/2014] [Indexed: 02/06/2023] Open
Abstract
Alzheimer's disease (AD) is the most common neurodegenerative disorder and the leading cause of age-related dementia worldwide. Several models for AD have been developed to provide information regarding the initial changes that lead to degeneration. Transgenic mouse models recapitulate many, but not all, of the features of AD, most likely because of the high complexity of the pathology. In this context, the validation of a wild-type animal model of AD that mimics the neuropathological and behavioral abnormalities is necessary. In previous studies, we have reported that the Chilean rodent Octodon degus could represent a natural model for AD. In the present work, we further describe the age-related neurodegeneration observed in the O. degus brain. We report some histopathological markers associated with the onset progression of AD, such as glial activation, increase in oxidative stress markers, neuronal apoptosis and the expression of the peroxisome proliferative-activated receptor γ coactivator-1α (PGC-1α). With these results, we suggest that the O. degus could represent a new model for AD research and a powerful tool in the search for therapeutic strategies against AD.
Collapse
Affiliation(s)
- Nibaldo C Inestrosa
- Centro de Envejecimiento y Regeneración (CARE), Departamento de Biología Celular y Molecular, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile.,Centro UC Síndrome de Down, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile.,Centro de Excelencia en Biomedicina de Magallanes (CEBIMA), Universidad de Magallanes, Punta Arenas, Chile.,Centre for Healthy Brain Ageing, School of Psychiatry, Faculty of Medicine, University of New South Wales, Sydney, New South Wales, Australia
| | - Juvenal A Ríos
- Centro de Envejecimiento y Regeneración (CARE), Departamento de Biología Celular y Molecular, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Pedro Cisternas
- Centro de Envejecimiento y Regeneración (CARE), Departamento de Biología Celular y Molecular, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Cheril Tapia-Rojas
- Centro de Envejecimiento y Regeneración (CARE), Departamento de Biología Celular y Molecular, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Daniela S Rivera
- Departamento de Ecología and Center of Applied Ecology and Sustainability (CAPES), Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Nady Braidy
- Centre for Healthy Brain Ageing, School of Psychiatry, Faculty of Medicine, University of New South Wales, Sydney, New South Wales, Australia
| | - Juan M Zolezzi
- Departamento de Biología, Facultad de Ciencias, Universidad de Tarapacá, Arica, Chile
| | - Juan A Godoy
- Centro de Envejecimiento y Regeneración (CARE), Departamento de Biología Celular y Molecular, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Francisco J Carvajal
- Centro de Envejecimiento y Regeneración (CARE), Departamento de Biología Celular y Molecular, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Alvaro O Ardiles
- Centro Interdisciplinario de Neurociencia de Valparaíso, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso, Chile
| | - Francisco Bozinovic
- Centro UC Síndrome de Down, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile.,Departamento de Ecología and Center of Applied Ecology and Sustainability (CAPES), Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Adrián G Palacios
- Centro Interdisciplinario de Neurociencia de Valparaíso, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso, Chile
| | - Perminder S Sachdev
- Centre for Healthy Brain Ageing, School of Psychiatry, Faculty of Medicine, University of New South Wales, Sydney, New South Wales, Australia.,Neurosychiatric Institute, Prince of Wales Hospital, Randwick, New South Wales, Australia
| |
Collapse
|
17
|
Silva T, Bravo J, Summavielle T, Remião F, Pérez C, Gil C, Martínez A, Borges F. Biology-oriented development of novel lipophilic antioxidants with neuroprotective activity. RSC Adv 2015. [DOI: 10.1039/c4ra15164j] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Discovery of hydroxycinnamic acid derivatives with enhanced in lipophilicity, blood brain barrier permeability and neuroprotective potential.
Collapse
Affiliation(s)
- T. Silva
- CIQ/Department of Chemistry and Biochemistry
- Faculty of Sciences
- University of Porto
- Porto
- Portugal
| | - J. Bravo
- Addiction Biology Group
- Institute for Molecular and Cell Biology
- University of Porto
- Porto
- Portugal
| | - T. Summavielle
- Addiction Biology Group
- Institute for Molecular and Cell Biology
- University of Porto
- Porto
- Portugal
| | - F. Remião
- REQUIMTE/Laboratory of Toxicology
- Department of Biological Sciences
- Faculty of Pharmacy
- University of Porto
- Porto
| | - C. Pérez
- Instituto de Química Médica
- CSIC
- Madrid
- Spain
| | - C. Gil
- Instituto de Química Médica
- CSIC
- Madrid
- Spain
| | | | - F. Borges
- CIQ/Department of Chemistry and Biochemistry
- Faculty of Sciences
- University of Porto
- Porto
- Portugal
| |
Collapse
|
18
|
Serrano FG, Tapia-Rojas C, Carvajal FJ, Hancke J, Cerpa W, Inestrosa NC. Andrographolide reduces cognitive impairment in young and mature AβPPswe/PS-1 mice. Mol Neurodegener 2014; 9:61. [PMID: 25524173 PMCID: PMC4414355 DOI: 10.1186/1750-1326-9-61] [Citation(s) in RCA: 90] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2014] [Accepted: 12/06/2014] [Indexed: 12/29/2022] Open
Abstract
Alzheimer’s disease (AD) is a neurodegenerative disorder in which the amyloid-β (Aβ) oligomers are a key factor in synaptic impairment and in spatial memory decline associated with neuronal dysfunction. This impairment includes synaptic failure associated with the loss of synaptic proteins that contribute to AD progression. Interestingly, the use of natural compounds is an emergent conceptual strategy in the search for drugs with therapeutic potentials for treating neurodegenerative disorders. In the present study, we report that andrographolide (ANDRO), which is a labdane diterpene extracted from Andrographis paniculata, increases slope of field excitatory postsynaptic potentials (fEPSP) in the CA1 region of hippocampal slices and inhibits long-term depression (LTD), protecting the long-term potentiation (LTP) against the damage induced by Aβ oligomers in vitro, most likely by inhibiting glycogen synthase kinase-3β (GSK-3β). Additionally, ANDRO prevents changes in neuropathology in two different age groups (7- and 12-month-old mice) of an AβPPswe/PS-1 Alzheimer’s model. ANDRO reduces the Aβ levels, changing the ontogeny of amyloid plaques in hippocampi and cortices in 7-month-old mice, and reduces tau phosphorylation around the Aβ oligomeric species in both age groups. Additionally, we observed that ANDRO recovers spatial memory functions that correlate with protecting synaptic plasticity and synaptic proteins in two different age groups. Our results suggest that ANDRO could be used in a potential preventive therapy during AD progression.
Collapse
Affiliation(s)
- Felipe G Serrano
- Centro de Envejecimiento y Regeneración (CARE), Santiago, Chile.
| | | | - Francisco J Carvajal
- Departamento de Biología Celular y Molecular, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile.
| | - Juan Hancke
- Instituto de Farmacología y Morfofisiología, Universidad Austral de Chile, Valdivia, Chile.
| | - Waldo Cerpa
- Departamento de Biología Celular y Molecular, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile.
| | - Nibaldo C Inestrosa
- Centro de Envejecimiento y Regeneración (CARE), Santiago, Chile. .,Departamento de Biología Celular y Molecular, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile. .,Center of Healthy Brain Aging, School of Psychiatry, Faculty of Medicine, University of New South Wales, Sydney, Australia. .,Centro UC Síndrome de Down, Pontificia Universidad Católica de Chile, Santiago, Chile. .,Centro de Excelencia en Biomedicina de Magallanes (CEBIMA), Universidad de Magallanes, Punta Arenas, Chile. .,CARE Biomedical Center, P. Catholic University of Chile, Postal code 8331150, PO Box 114-D, Santiago, Chile.
| |
Collapse
|
19
|
Godoy JA, Rios JA, Zolezzi JM, Braidy N, Inestrosa NC. Signaling pathway cross talk in Alzheimer's disease. Cell Commun Signal 2014; 12:23. [PMID: 24679124 PMCID: PMC3977891 DOI: 10.1186/1478-811x-12-23] [Citation(s) in RCA: 112] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2014] [Accepted: 03/11/2014] [Indexed: 01/11/2023] Open
Abstract
Numerous studies suggest energy failure and accumulative intracellular waste play a causal role in the pathogenesis of several neurodegenerative disorders and Alzheimer's disease (AD) in particular. AD is characterized by extracellular amyloid deposits, intracellular neurofibrillary tangles, cholinergic deficits, synaptic loss, inflammation and extensive oxidative stress. These pathobiological changes are accompanied by significant behavioral, motor, and cognitive impairment leading to accelerated mortality. Currently, the potential role of several metabolic pathways associated with AD, including Wnt signaling, 5' adenosine monophosphate-activated protein kinase (AMPK), mammalian target of rapamycin (mTOR), Sirtuin 1 (Sirt1, silent mating-type information regulator 2 homolog 1), and peroxisome proliferator-activated receptor gamma co-activator 1-α (PGC-1α) have widened, with recent discoveries that they are able to modulate several pathological events in AD. These include reduction of amyloid-β aggregation and inflammation, regulation of mitochondrial dynamics, and increased availability of neuronal energy. This review aims to highlight the involvement of these new set of signaling pathways, which we have collectively termed "anti-ageing pathways", for their potentiality in multi-target therapies against AD where cellular metabolic processes are severely impaired.
Collapse
Affiliation(s)
- Juan A Godoy
- Centro de Envejecimiento y Regeneración (CARE); Departamento de Biología Celular y Molecular; Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Av. Alameda 340, Santiago, Chile
| | - Juvenal A Rios
- Centro de Envejecimiento y Regeneración (CARE); Departamento de Biología Celular y Molecular; Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Av. Alameda 340, Santiago, Chile
| | - Juan M Zolezzi
- Departamento de Biología, Facultad de Ciencias, Universidad de Tarapacá, Arica, Chile
| | - Nady Braidy
- Center for Healthy Brain Ageing, School of Psychiatry, Faculty of Medicine, University of New South Wales, Sydney, Australia
| | - Nibaldo C Inestrosa
- Centro de Envejecimiento y Regeneración (CARE); Departamento de Biología Celular y Molecular; Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Av. Alameda 340, Santiago, Chile
- Center for Healthy Brain Ageing, School of Psychiatry, Faculty of Medicine, University of New South Wales, Sydney, Australia
| |
Collapse
|
20
|
Richard JA. Chemistry and Biology of the Polycyclic Polyprenylated Acylphloroglucinol Hyperforin. European J Org Chem 2013. [DOI: 10.1002/ejoc.201300815] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
|
21
|
Chitra L, Kumar CR, Basha HM, Ponne S, Boopathy R. Interaction of metal chelators with different molecular forms of acetylcholinesterase and its significance in Alzheimer's disease treatment. Proteins 2013; 81:1179-91. [DOI: 10.1002/prot.24267] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2012] [Revised: 01/22/2013] [Accepted: 02/06/2013] [Indexed: 01/31/2023]
Affiliation(s)
- Loganathan Chitra
- DRDO-BU Center for Life Sciences; Bharathiar University; Coimbatore; 641 046; Tamil Nadu; India
| | - Chinnadurai Raj Kumar
- Department of Biotechnology; School of Biotechnology and Genetic Engineering; Bharathiar University; Coimbatore; 641 046; Tamil Nadu; India
| | - Haleema M. Basha
- Department of Biotechnology; School of Biotechnology and Genetic Engineering; Bharathiar University; Coimbatore; 641 046; Tamil Nadu; India
| | - Saravanaraman Ponne
- Department of Biotechnology; School of Biotechnology and Genetic Engineering; Bharathiar University; Coimbatore; 641 046; Tamil Nadu; India
| | | |
Collapse
|
22
|
Nisticò R, Pignatelli M, Piccinin S, Mercuri NB, Collingridge G. Targeting synaptic dysfunction in Alzheimer's disease therapy. Mol Neurobiol 2012; 46:572-87. [PMID: 22914888 DOI: 10.1007/s12035-012-8324-3] [Citation(s) in RCA: 71] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2012] [Accepted: 08/06/2012] [Indexed: 12/22/2022]
Abstract
In the past years, major efforts have been made to understand the genetics and molecular pathogenesis of Alzheimer's disease (AD), which has been translated into extensive experimental approaches aimed at slowing down or halting disease progression. Advances in transgenic (Tg) technologies allowed the engineering of different mouse models of AD recapitulating a range of AD-like features. These Tg models provided excellent opportunities to analyze the bases for the temporal evolution of the disease. Several lines of evidence point to synaptic dysfunction as a cause of AD and that synapse loss is a pathological correlate associated with cognitive decline. Therefore, the phenotypic characterization of these animals has included electrophysiological studies to analyze hippocampal synaptic transmission and long-term potentiation, a widely recognized cellular model for learning and memory. Transgenic mice, along with non-Tg models derived mainly from exogenous application of Aβ, have also been useful experimental tools to test the various therapeutic approaches. As a result, numerous pharmacological interventions have been reported to attenuate synaptic dysfunction and improve behavior in the different AD models. To date, however, very few of these findings have resulted in target validation or successful translation into disease-modifying compounds in humans. Here, we will briefly review the synaptic alterations across the different animal models and we will recapitulate the pharmacological strategies aimed at rescuing hippocampal plasticity phenotypes. Finally, we will highlight intrinsic limitations in the use of experimental systems and related challenges in translating preclinical studies into human clinical trials.
Collapse
Affiliation(s)
- Robert Nisticò
- Department of Pharmacobiology, University of Calabria, 87036 Rende, Italy.
| | | | | | | | | |
Collapse
|
23
|
Carvajal FJ, Inestrosa NC. Interactions of AChE with Aβ Aggregates in Alzheimer's Brain: Therapeutic Relevance of IDN 5706. Front Mol Neurosci 2011; 4:19. [PMID: 21949501 PMCID: PMC3172730 DOI: 10.3389/fnmol.2011.00019] [Citation(s) in RCA: 106] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2011] [Accepted: 08/21/2011] [Indexed: 12/20/2022] Open
Abstract
Acetylcholinesterase (AChE; EC 3.1.1.7) plays a crucial role in the rapid hydrolysis of the neurotransmitter acetylcholine, in the central and peripheral nervous system and might also participate in non-cholinergic mechanism related to neurodegenerative diseases. Alzheimer’s disease (AD) is a neurodegenerative disorder characterized by a progressive deterioration of cognitive abilities, amyloid-β (Aβ) peptide accumulation and synaptic alterations. We have previously shown that AChE is able to accelerate the Aβ peptide assembly into Alzheimer-type aggregates increasing its neurotoxicity. Furthermore, AChE activity is altered in brain and blood of Alzheimer’s patients. The enzyme associated to amyloid plaques changes its enzymatic and pharmacological properties, as well as, increases its resistant to low pH, inhibitors and excess of substrate. Here, we reviewed the effects of IDN 5706, a hyperforin derivative that has potential preventive effects on the development of AD. Our results show that treatment with IDN 5706 for 10 weeks increases brain AChE activity in 7-month-old double transgenic mice (APPSWE–PS1) and decreases the content of AChE associated with different types of amyloid plaques in this Alzheimer’s model. We concluded that early treatment with IDN 5706 decreases AChE–Aβ interaction and this effect might be of therapeutic interest in the treatment of AD.
Collapse
Affiliation(s)
- Francisco J Carvajal
- Centro de Envejecimiento y Regeneración (CARE), Departamento de Biología Celular y Molecular, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile Santiago, Chile
| | | |
Collapse
|
24
|
The emergence of designed multiple ligands for neurodegenerative disorders. Prog Neurobiol 2011; 94:347-59. [PMID: 21536094 DOI: 10.1016/j.pneurobio.2011.04.010] [Citation(s) in RCA: 69] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2010] [Revised: 04/13/2011] [Accepted: 04/15/2011] [Indexed: 12/21/2022]
Abstract
The incidence of neurodegenerative diseases has seen a constant increase in the global population, and is likely to be the result of extended life expectancy brought about by better health care. Despite this increase in the incidence of neurodegenerative diseases, there has been a dearth in the introduction of new disease-modifying therapies that are approved to prevent or delay the onset of these diseases, or reverse the degenerative processes in brain. Mounting evidence in the peer-reviewed literature shows that the etiopathology of these diseases is extremely complex and heterogeneous, resulting in significant comorbidity and therefore unlikely to be mitigated by any drug acting on a single pathway or target. A recent trend in drug design and discovery is the rational design or serendipitous discovery of novel drug entities with the ability to address multiple drug targets that form part of the complex pathophysiology of a particular disease state. In this review we discuss the rationale for developing such multifunctional drugs (also called designed multiple ligands or DMLs), and why these drug candidates seem to offer better outcomes in many cases compared to single-targeted drugs in pre-clinical studies for neurodegenerative diseases such as Alzheimer's and Parkinson's disease. Examples are drawn from the literature of drug candidates that have already reached the market, some unsuccessful attempts, and others that are still in the drug development pipeline.
Collapse
|
25
|
Balducci C, Forloni G. APP transgenic mice: their use and limitations. Neuromolecular Med 2010; 13:117-37. [PMID: 21152995 DOI: 10.1007/s12017-010-8141-7] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2010] [Accepted: 11/20/2010] [Indexed: 12/12/2022]
Abstract
Alzheimer's disease is the most widespread form of dementia. Its histopathological hallmarks include vascular and extracellular β-amyloid (Aβ) deposition and intraneuronal neurofibrillary tangles (NFTs). Gradual decline of cognitive functions linked to progressive synaptic loss makes patients unable to store new information in the earlier stages of the pathology, later becoming completely dependent because they are unable to do even elementary daily life actions. Although more than a hundred years have passed since Alois Alzheimer described the first case of AD, and despite many years of intense research, there are still many crucial points to be discovered in the neuropathological pathway. The development of transgenic mouse models engineered with overexpression of the amyloid precursor protein carrying familial AD mutations has been extremely useful. Transgenic mice present the hallmarks of the pathology, and histological and behavioural examination supports the amyloid hypothesis. As in human AD, extracellular Aβ deposits surrounded by activated astrocytes and microglia are typical features, together with synaptic and cognitive defects. Although animal models have been widely used, they are still being continuously developed in order to recapitulate some missing aspects of the disease. For instance, AD therapeutic agents tested in transgenic mice gave encouraging results which, however, were very disappointing in clinical trials. Neuronal cell death and NFTs typical of AD are much harder to replicate in these mice, which thus offer a fundamental but still imperfect tool for understanding and solving dementia pathology.
Collapse
Affiliation(s)
- Claudia Balducci
- Department of Neuroscience, Mario Negri Institute for Pharmacological Research, via G. La Masa, 19, 20156, Milan, Italy.
| | | |
Collapse
|
26
|
Shi C, Wu F, Xu J. H2O2 and PAF mediate Abeta1-42-induced Ca2+ dyshomeostasis that is blocked by EGb761. Neurochem Int 2010; 56:893-905. [PMID: 20362023 DOI: 10.1016/j.neuint.2010.03.016] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2009] [Revised: 03/05/2010] [Accepted: 03/23/2010] [Indexed: 11/27/2022]
Abstract
Calcium (Ca2+) dyshomeostasis may be of pivotal importance in mediating the neurotoxic action of amyloid beta peptide (Abeta), but the mechanism whereby Abeta disrupts Ca2+ homeostasis remains unclear. Using hippocampal neuronal cultures, the present study investigated possible mechanisms underlying Ca2+ dyshomeostasis induced by the oligomeric form of Abeta1-42 and two possible mediators of its toxicity, hydrogen peroxide (H2O2) and platelet-activating factor (PAF). It was found that, both H2O2 and PAF were able to reproduce each of the events induced by oligomeric Abeta1-42, including (a) Ca2+ influx via N-methyl-D-aspartic acid (NMDA) receptors, (b) enhancement of Ca2+ response to NMDA via activation of protein kinase C (PKC), (c) the increase of extracellular concentrations of glutamate and (d) the increase in cytosolic free Ca2+ ([Ca2+]i). Moreover, each of these events could be blocked by Ginkgo biloba extract EGb761, a free radical scavenger with PAF antagonism, and by quercetin, a constituent with well-established free radical scavenging property. In contrast, ginkgolide B, another constituent of EGb761 with well-established PAF-antagonizing activity protected the neurons against Ca2+ dyshomeostasis induced by Abeta1-42 and PAF, but not by H2O2. These results suggested the possibility that Abeta1-42-induced Ca2+ dyshomeostasis might be mediated by formation of toxic mediators such as H2O2 and PAF. Therefore, increased production of toxic mediators such as H2O2 and PAF in the brain may be critical in the pathological mechanism of neurodegenerative diseases, particularly Alzheimer's disease (AD), and may serve as major therapeutic targets for these diseases.
Collapse
Affiliation(s)
- Chun Shi
- Department of Anatomy, Zhongshan School of Medicine, Sun Yat-Sen University Guangzhou, Guangdong 510080, China.
| | | | | |
Collapse
|
27
|
Shi C, Fang L, Yew DT, Yao Z, Xu J. Ginkgo biloba extract EGb761 protects against mitochondrial dysfunction in platelets and hippocampi in ovariectomized rats. Platelets 2010; 21:53-9. [PMID: 19938886 DOI: 10.3109/09537100903395180] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Using ovariectomized middle-aged rats to mimic the post-menopausal pathophysiological changes in women, we have previously demonstrated that estrogen withdrawal and age-related decrease in the functional reserve of mitochondria might co-operate to induce persistent mitochondrial dysfunction, which may be critical in inducing degenerative processes in the brain later in post-menopausal women. The standardized Ginkgo biloba extract EGb761 has long been considered a natural antioxidant. More recently it has also proposed to have direct protective effects on the mitochondria. In this work, effects of EGb761 on mitochondrial function in platelets and hippocampi of ovariectomized and sham-operated rats were investigated. It was found that EGb761 protected against the decrease of cytochrome c oxidase (COX) activity, mitochondrial ATP (adenosine-5'-triphosphate) content and mitochondrial glutathione (GSH) content in both platelets and hippocampi of ovariectomized rats, suggesting its peripheral and central effects against estrogen withdrawal-induced degeneration. In contrast, in sham-operated rats, EGb761 increased mitochondrial GSH content in platelets but failed to show similar effect on hippocampi, suggesting that EGb761 may help to enhance the functional reserve of mitochondria, but this effect was limited to the outside of the central nervous system. EGb761 displayed similar effects on platelets and hippocampi of ovariectomized rats but showed differential effects on platelets and hippocampi of sham-operated rats, possibly because estrogen withdrawal induced an increase of blood brain barrier (BBB) permeability. Therefore, while EGb761's effect may be limited to the outside of the nervous system under normal physiological conditions, EGb761 may be a potential protective agent against central neurodegeneration in post-menopausal women.
Collapse
Affiliation(s)
- Chun Shi
- Department of Anatomy, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, Guangdong 510080, China
| | | | | | | | | |
Collapse
|
28
|
Ginkgo biloba extract in Alzheimer's disease: from action mechanisms to medical practice. Int J Mol Sci 2010; 11:107-23. [PMID: 20162004 PMCID: PMC2820992 DOI: 10.3390/ijms11010107] [Citation(s) in RCA: 98] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2009] [Revised: 12/31/2009] [Accepted: 01/01/2010] [Indexed: 12/15/2022] Open
Abstract
Standardized extract from the leaves of the Ginkgo biloba tree, labeled EGb761, is one of the most popular herbal supplements. Numerous preclinical studies have shown the neuroprotective effects of EGb761 and support the notion that it may be effective in the treatment and prevention of neurodegenerative disorders such as Alzheimer’s disease (AD). Despite the preclinical promise, the clinical efficacy of this drug remains elusive. In this review, possible mechanisms underlying neuroprotective actions of EGb761 are described in detail, together with a brief discussion of the problem of studying this herb clinically to verify its efficacy in the treatment and prevention of AD. Moreover, various parameters e.g., the dosage and the permeability of the blood brain barrier (BBB), impacting the outcome of the clinical effectiveness of the extract are also discussed. Overall, the findings summarized in this review suggest that, a better understanding of the neuroprotective mechanisms of EGb761 may contribute to better understanding of the effectiveness and complexity of this herb and may also be helpful for design of therapeutic strategies in future clinical practice. Therefore, in future clinical studies, different factors that could interfere with the effect of EGb761 should be considered.
Collapse
|
29
|
Gao W, Zhu H, Zhang JY, Zhang XJ. Calcium signaling-induced Smad3 nuclear accumulation induces acetylcholinesterase transcription in apoptotic HeLa cells. Cell Mol Life Sci 2009; 66:2181-93. [PMID: 19468687 PMCID: PMC11115644 DOI: 10.1007/s00018-009-0037-z] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2009] [Revised: 04/01/2009] [Accepted: 04/21/2009] [Indexed: 10/20/2022]
Abstract
Recently, acetylcholinesterase (AChE) has been studied as an important apoptosis regulator. We previously showed that cellular calcium mobilization upregulated AChE expression by modulating promoter activity and mRNA stability. In this study, we have identified a potential Smad3/4 binding element, TGCCAGACA, located within the -601 to -571 bp fragment of the AChE promoter, as an important calcium response motif. Smad2/3 and Smad4 were shown to bind this element. Overexpression of human Smad3 increased AChE transcription activity in a dose-dependent manner in HeLa cells, whereas dominant-negative Smad3 blocked this activation. Upon A23187 and thapsigargin treatment, nuclear Smad3 accumulation was observed, an effect that was blocked by the intracellular Ca(2+) chelator BAPTA-AM. Calcium-induced AChE transcriptional activation was significantly blocked when the nuclear localization signal of Smad3 was destroyed. Taken together, our data suggest Smad3 can regulate AChE transcriptional activation following calcium-induced nuclear accumulation.
Collapse
Affiliation(s)
- Wei Gao
- Laboratory of Molecular Cell Biology, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, 320 YueYang Road, Shanghai, 200031 China
| | - Hui Zhu
- Laboratory of Molecular Cell Biology, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, 320 YueYang Road, Shanghai, 200031 China
| | - Jing-Ya Zhang
- Laboratory of Molecular Cell Biology, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, 320 YueYang Road, Shanghai, 200031 China
| | - Xue-Jun Zhang
- Laboratory of Molecular Cell Biology, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, 320 YueYang Road, Shanghai, 200031 China
| |
Collapse
|