1
|
Maglione AV, do Nascimento BPP, Ribeiro MO, de Souza TJL, da Silva REC, Sato MA, Penatti CAA, Britto LRG, de Souza JS, Maciel RMB, da Conceição RR, Laureano-Melo R, Giannocco G. Triiodothyronine Treatment reverses Depression-Like Behavior in a triple-transgenic animal model of Alzheimer's Disease. Metab Brain Dis 2022; 37:2735-2750. [PMID: 35951206 DOI: 10.1007/s11011-022-01055-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Accepted: 07/19/2022] [Indexed: 11/29/2022]
Abstract
Alzheimer disease's (AD) is a neurodegenerative disorder characterized by cognitive and behavioral impairment. The central nervous system is an important target of thyroid hormones (TH). An inverse association between serum triiodothyronine (T3) levels and the risk of AD symptoms and progression has been reported. We investigated the effects of T3 treatment on the depression-like behavior in male transgenic 3xTg-AD mice. Animals were divided into 2 groups treated with daily intraperitoneal injections of 20 ng/g of body weight (b.w.) L-T3 (T3 group) or saline (vehicle, control group). The experimental protocol lasted 21 days, and behavioral tests were conducted on days 18-20. At the end of the experiment, the TH profile and hippocampal gene expression were evaluated. The T3-treated group significantly increased serum T3 and decreased thyroxine (T4) levels. When compared to control hippocampal samples, the T3 group exhibited attenuated glycogen synthase kinase-3 (GSK3), metalloproteinase 10 (ADAM10), amyloid-beta precursor-protein (APP), serotonin transporter (SERT), 5HT1A receptor, monocarboxylate transporter 8 (MCT8) and bone morphogenetic protein 7 (BMP-7) gene expression, whereas augmented superoxide dismutase 2 (SOD2) and Hairless gene expression. T3-treated animals also displayed reduced immobility time in both the tail suspension and forced swim tests, and in the latter presented a higher latency time compared to the control group. Therefore, our findings suggest that in an AD mouse model, T3 supplementation promotes improvements in depression-like behavior, through the modulation of the serotonergic related genes involved in the transmission mediated by 5HT1A receptors and serotonin reuptake, and attenuated disease progression.
Collapse
Affiliation(s)
- Andréa V Maglione
- Dept. Medicine, Laboratory of Endocrinology and Translational Medicine, Universidade Federal de São Paulo, UNIFESP/EPM, São Paulo, Brazil
| | - Bruna P P do Nascimento
- Laboratory of Translational Medicine, Universidade Federal de São Paulo, UNIFESP/EPM, São Paulo, Brazil
- Developmental Disorders Program, Center of Biological Science and Health, Mackenzie Presbyterian University, São Paulo, Brazil
| | - Miriam O Ribeiro
- Developmental Disorders Program, Center of Biological Science and Health, Mackenzie Presbyterian University, São Paulo, Brazil
| | - Talytha J L de Souza
- Dept. Medicine, Laboratory of Endocrinology and Translational Medicine, Universidade Federal de São Paulo, UNIFESP/EPM, São Paulo, Brazil
| | - Renata E C da Silva
- Dept. Medicine, Laboratory of Endocrinology and Translational Medicine, Universidade Federal de São Paulo, UNIFESP/EPM, São Paulo, Brazil
| | - Monica A Sato
- Dept. Morphology and Physiology, Faculdade de Medicina do ABC, Centro Universitário FMABC, Santo André- Brazil, São Paulo, Santo André, Brazil
| | - Carlos A A Penatti
- Laboratory of Human Physiology, Universidade Nove de Julho, São Paulo, Brazil
| | - Luiz R G Britto
- Institute of Biomedical Sciences, Universidade de São Paulo, São Paulo, Brazil
| | - Janaina S de Souza
- Dept. Medicine, Laboratory of Endocrinology and Translational Medicine, Universidade Federal de São Paulo, UNIFESP/EPM, São Paulo, Brazil
| | - Rui M B Maciel
- Dept. Medicine, Laboratory of Endocrinology and Translational Medicine, Universidade Federal de São Paulo, UNIFESP/EPM, São Paulo, Brazil
| | - Rodrigo Rodrigues da Conceição
- Dept. Medicine, Laboratory of Endocrinology and Translational Medicine, Universidade Federal de São Paulo, UNIFESP/EPM, São Paulo, Brazil.
| | - Roberto Laureano-Melo
- Laboratory of Physiopharmacoly and Behavior, Universidade de Barra Mansa, Rio de Janeiro, Brazil
| | - Gisele Giannocco
- Dept. Medicine, Laboratory of Endocrinology and Translational Medicine, Universidade Federal de São Paulo, UNIFESP/EPM, São Paulo, Brazil.
| |
Collapse
|
2
|
Go MJ, Kim JM, Kang JY, Park SK, Lee CJ, Kim MJ, Lee HR, Kim TY, Joo SG, Kim DO, Heo HJ. Korean Red Pine ( Pinus densiflora) Bark Extract Attenuates Aβ-Induced Cognitive Impairment by Regulating Cholinergic Dysfunction and Neuroinflammation. J Microbiol Biotechnol 2022; 32:1154-1167. [PMID: 36039041 PMCID: PMC9628973 DOI: 10.4014/jmb.2207.07015] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2022] [Revised: 07/28/2022] [Accepted: 07/29/2022] [Indexed: 12/15/2022]
Abstract
In this study, we investigated the anti-amnesic effect of Korean red pine (Pinus densiflora) bark extract (KRPBE) against amyloid beta1-42 (Aβ1-42)-induced neurotoxicity. We found that treatment with KRPBE improved the behavioral function in Aβ-induced mice, and also boosted the antioxidant system in mice by decreasing malondialdehyde (MDA) content, increasing superoxide dismutase (SOD) activities, and reducing glutathione (GSH) levels. In addition, KRPBE improved the cholinergic system by suppressing reduced acetylcholine (ACh) content while also activating acetylcholinesterase (AChE), regulating the expression of choline acetyltransferase (ChAT), postsynaptic density protein-95 (PSD-95), and synaptophysin. KRPBE also showed an ameliorating effect on cerebral mitochondrial deficit by regulating reactive oxygen species (ROS), mitochondrial membrane potential (MMP) and ATP levels. Moreover, KRPBE modulated the expression levels of neurotoxicity indicators Aβ and phosphorylated tau (p-tau) and inflammatory cytokines TNF-α, p-IκB-α, and IL-1β. Furthermore, we found that KRPBE improved the expression levels of neuronal apoptosis-related markers BAX and BCl-2 and increased the expression levels of BDNF and p-CREB. Therefore, this study suggests that KRPBE treatment has an anti-amnestic effect by modulating cholinergic system dysfunction and neuroinflammation in Aβ1-42-induced cognitive impairment in mice.
Collapse
Affiliation(s)
- Min Ji Go
- Division of Applied Life Science (BK21), Institute of Agriculture and Life Science, Gyeongsang National University, Jinju 52828, Republic of Korea
| | - Jong Min Kim
- Division of Applied Life Science (BK21), Institute of Agriculture and Life Science, Gyeongsang National University, Jinju 52828, Republic of Korea
| | - Jin Yong Kang
- Division of Applied Life Science (BK21), Institute of Agriculture and Life Science, Gyeongsang National University, Jinju 52828, Republic of Korea,Advanced Process Technology and Fermentation Research Group, World Institute of Kimchi, Gwangju 61755, Republic of Korea
| | - Seon Kyeong Park
- Division of Applied Life Science (BK21), Institute of Agriculture and Life Science, Gyeongsang National University, Jinju 52828, Republic of Korea,Korea Food Research Institute, Wanju-gun 55365, Republic of Korea
| | - Chang Jun Lee
- Division of Applied Life Science (BK21), Institute of Agriculture and Life Science, Gyeongsang National University, Jinju 52828, Republic of Korea,Korea Food Research Institute, Wanju-gun 55365, Republic of Korea
| | - Min Ji Kim
- Division of Applied Life Science (BK21), Institute of Agriculture and Life Science, Gyeongsang National University, Jinju 52828, Republic of Korea
| | - Hyo Rim Lee
- Division of Applied Life Science (BK21), Institute of Agriculture and Life Science, Gyeongsang National University, Jinju 52828, Republic of Korea
| | - Tae Yoon Kim
- Division of Applied Life Science (BK21), Institute of Agriculture and Life Science, Gyeongsang National University, Jinju 52828, Republic of Korea
| | - Seung Gyum Joo
- Division of Applied Life Science (BK21), Institute of Agriculture and Life Science, Gyeongsang National University, Jinju 52828, Republic of Korea
| | - Dae-Ok Kim
- Department of Food Science and Biotechnology, Kyung Hee University, Yongin 17104, Republic of Korea
| | - Ho Jin Heo
- Division of Applied Life Science (BK21), Institute of Agriculture and Life Science, Gyeongsang National University, Jinju 52828, Republic of Korea,Corresponding author Phone: +82-55-772-1907 Fax: +82-55-772-1909 E-mail:
| |
Collapse
|
3
|
Haghighijoo Z, Zamani L, Moosavi F, Emami S. Therapeutic potential of quinazoline derivatives for Alzheimer's disease: A comprehensive review. Eur J Med Chem 2022; 227:113949. [PMID: 34742016 DOI: 10.1016/j.ejmech.2021.113949] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2021] [Revised: 10/02/2021] [Accepted: 10/22/2021] [Indexed: 12/24/2022]
Abstract
Quinazolines are considered as a promising class of bioactive heterocyclic compounds with broad properties. Particularly, the quinazoline scaffold has an impressive role in the design and synthesis of new CNS-active drugs. The drug-like properties and pharmacological characteristics of quinazoline could lead to different drugs with various targets. Among CNS disorders, Alzheimer's disease (AD) is a progressive neurodegenerative disorder with memory loss, cognitive decline and language dysfunction. AD is a complex and multifactorial disease therefore, the need for finding multi-target drugs against this devastative disease is urgent. A literature survey revealed that quinazoline derivatives have diverse therapeutic potential for AD as modulators/inhibitors of β-amyloid, tau protein, cholinesterases, monoamine oxidases, and phosphodiesterases as well as other protective effects. Thus, we describe here the most relevant and recent studies about anti-AD agents with quinazoline structure which can further aid the development and discovery of new anti-AD agents.
Collapse
Affiliation(s)
- Zahra Haghighijoo
- Department of Chemistry, University of Louisiana at Lafayette, Lafayette, LA, 70504, USA
| | - Leila Zamani
- Worcester Polytechnic Institute, 100 Institute Road, Worcester, MA, 01609, USA
| | - Fatemeh Moosavi
- Medicinal and Natural Products Chemistry Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Saeed Emami
- Department of Medicinal Chemistry and Pharmaceutical Sciences Research Center, Faculty of Pharmacy, Mazandaran University of Medical Sciences, Sari, Iran.
| |
Collapse
|
4
|
Gulcan HO, Kosar M. The hybrid compounds as multi-target ligands for the treatment of Alzheimer's Disease: Considerations on Donepezil. Curr Top Med Chem 2021; 22:395-407. [PMID: 34766890 DOI: 10.2174/1568026621666211111153626] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Revised: 08/31/2021] [Accepted: 11/11/2021] [Indexed: 11/22/2022]
Abstract
The strategies to combat Alzheimer's Disease (AD) have been changing with respect to the failures of many drug candidates assessed in clinical studies, the complex pathophysiology of AD, and the limitations of the current drugs employed. So far, none of the targets, either validated or nonvalidated, have been shown to be purely causative in the generation and development of AD. Considering the progressive and the neurodegenerative characteristics of the disease, the main strategy has been based on the design of molecules capable of showing activity on more than one receptor, and it is defined as multi-target ligand design strategy. The hybrid molecule concept is an outcome of this approach. Donepezil, as one of the currently employed drugs for AD therapy, has also been utilized in hybrid drug design studies. This review has aimed to present the promising donepezil-like hybrid molecules introduced in the recent period. Particularly, multi-target ligands with additional activities concomitant to cholinesterase inhibition are preferred.
Collapse
Affiliation(s)
- Hayrettin Ozan Gulcan
- Faculty of Pharmacy, Eastern Mediterranean University, Famagusta, T.R. North Cyprus, via Mersin 10, Turkey
| | - Muberra Kosar
- Faculty of Pharmacy, Eastern Mediterranean University, Famagusta, T.R. North Cyprus, via Mersin 10, Turkey
| |
Collapse
|
5
|
Morton H, Kshirsagar S, Orlov E, Bunquin LE, Sawant N, Boleng L, George M, Basu T, Ramasubramanian B, Pradeepkiran JA, Kumar S, Vijayan M, Reddy AP, Reddy PH. Defective mitophagy and synaptic degeneration in Alzheimer's disease: Focus on aging, mitochondria and synapse. Free Radic Biol Med 2021; 172:652-667. [PMID: 34246776 DOI: 10.1016/j.freeradbiomed.2021.07.013] [Citation(s) in RCA: 113] [Impact Index Per Article: 28.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/18/2021] [Revised: 06/06/2021] [Accepted: 07/06/2021] [Indexed: 12/12/2022]
Abstract
Alzheimer's disease (AD) is a progressive neurodegenerative disease characterized by memory loss and multiple cognitive impairments. AD is marked by multiple cellular changes, including deregulation of microRNAs, activation of glia and astrocytes, hormonal imbalance, defective mitophagy, synaptic degeneration, in addition to extracellular neuritic amyloid-beta (Aβ) plaques, phosphorylated tau (P-tau), and intracellular neurofibrillary tangles (NFTs). Recent research in AD revealed that defective synaptic mitophagy leads to synaptic degeneration and cognitive dysfunction in AD neurons. Our critical analyses of mitochondria and Aβ and P-tau revealed that increased levels of Aβ and P-Tau, and abnormal interactions between Aβ and Drp1, P-Tau and Drp1 induced increased mitochondrial fragmentation and proliferation of dysfunctional mitochondria in AD neurons and depleted Parkin and PINK1 levels. These events ultimately lead to impaired clearance of dead and/or dying mitochondria in AD neurons. The purpose of our article is to highlight the recent research on mitochondria and synapses in relation to Aβ and P-tau, focusing on recent developments.
Collapse
Affiliation(s)
- Hallie Morton
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, TX, USA
| | - Sudhir Kshirsagar
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, TX, USA
| | - Erika Orlov
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, TX, USA
| | - Lloyd E Bunquin
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, TX, USA
| | - Neha Sawant
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, TX, USA
| | - Lauren Boleng
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, TX, USA; Nutritional Sciences Nutritional Science, College of Human Sciences, Texas Tech University, 1301Akron Ave, Lubbock, TX, 79409, USA
| | - Mathew George
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, TX, USA
| | - Tanisha Basu
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, TX, USA
| | | | | | - Subodh Kumar
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, TX, USA
| | - Murali Vijayan
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, TX, USA
| | - Arubala P Reddy
- Nutritional Sciences Nutritional Science, College of Human Sciences, Texas Tech University, 1301Akron Ave, Lubbock, TX, 79409, USA
| | - P Hemachandra Reddy
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, TX, USA; Neuroscience & Pharmacology, Texas Tech University Health Sciences Center, Lubbock, TX, USA; Neurology, Departments of School of Medicine, Texas Tech University Health Sciences Center, Lubbock, TX, USA; Public Health Department of Graduate School of Biomedical Sciences, Texas Tech University Health Sciences Center, Lubbock, TX, USA; Department of Speech, Language and Hearing Sciences, School Health Professions, Texas Tech University Health Sciences Center, Lubbock, TX, USA.
| |
Collapse
|
6
|
The structural simplification of lysergic acid as a natural lead for synthesizing novel anti-Alzheimer agents. Bioorg Med Chem Lett 2021; 47:128205. [PMID: 34139326 DOI: 10.1016/j.bmcl.2021.128205] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Revised: 06/06/2021] [Accepted: 06/11/2021] [Indexed: 11/23/2022]
Abstract
Alzheimer's disease (AD) is a neurodegenerative disorder, projected to be the second leading cause of mortality by 2040. AD is characterized by a progressive impairment of memory leading to dementia and loss of ability to carry out daily functions. In addition to the deficiency of acetylcholine release in synapse, there are other mechanisms explaining the etiology of the disease. The most disputing ones are associated with the accumulation of damaged proteins β-amyloid (Aβ) and hyperphosphorylated tau outside and inside neurons, respectively. Lysergic acid derivatives have been shown to possess promising anti-Alzheimer effect. Moreover, lysergic acid structure encompasses the general structural requirements for acetylcholinesterase inhibition. In this study, sixteen analogues, derived from lysergic acid structure, were synthesized. Heck and Mannich reactions were carried out to 4-bromo indole nucleus to generate potentially active analogues. Some of them were subsequently cyclized by nitromethane and zinc reduction procedures. Some of these compounds showed neuroprotective and anti-inflammatory effects stronger than the currently used anti-Alzheimer drug; donepezil. Some of the synthesized com-pounds showed a noticeable acetylcholinesterase inhibition. Twelve molecular targets attributed with AD etiology were tested versus the synthesized compounds by in silico modeling. Docking scores of modeling were plotted against in vitro activity of the compounds. The one afforded the strongest positive correlation was ULK-1 which has a significant role in autophagy.
Collapse
|
7
|
Qiao O, Ji H, Zhang Y, Zhang X, Zhang X, Liu N, Huang L, Liu C, Gao W. New insights in drug development for Alzheimer's disease based on microglia function. Biomed Pharmacother 2021; 140:111703. [PMID: 34083109 DOI: 10.1016/j.biopha.2021.111703] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2021] [Revised: 04/29/2021] [Accepted: 05/05/2021] [Indexed: 12/26/2022] Open
Abstract
One of the biggest challenges in drug development for Alzheimer's disease (AD) is how to effectively remove deposits of amyloid-beta (Aβ). Recently, the relationship between microglia and Aβ has become a research hotspot. Emerging evidence suggests that Aβ-induced microglia-mediated neuroinflammation further aggravates the decline of cognitive function, while microglia are also involved in the process of Aβ clearance. Hence, microglia have become a potential therapeutic target for the treatment or prevention of AD. An in-depth understanding of the role played by microglia in the development of AD will help us to broaden therapeutic strategies for AD. In this review, we provide an overview of the dual roles of microglia in AD progression: the positive effect of phagocytosis of Aβ and its negative effect on neuroinflammation after over-activation. With the advantages of novel structure, high efficiency, and low toxicity, small-molecule compounds as modulators of microglial function have attracted considerable attention in the therapeutic areas of AD. In this review, we also summarize the therapeutic potential of small molecule compounds (SMCs) and their structure-activity relationship for AD treatment through modulating microglial phagocytosis and inhibiting neuroinflammation. For example, the position and number of phenolic hydroxyl groups on the B ring are the key to the activity of flavonoids, and the substitution of hydroxyl groups on the benzene ring enhances the anti-inflammatory activity of phenolic acids. This review is expected to be useful for developing effective modulators of microglial function from SMCs for the amelioration and treatment of AD.
Collapse
Affiliation(s)
- Ou Qiao
- Tianjin Key Laboratory for Modern Drug Delivery and High-Efficiency, School of Pharmaceutical Science and Technology, Tianjin University, Weijin Road, Tianjin 300072, China
| | - Haixia Ji
- Tianjin Key Laboratory for Modern Drug Delivery and High-Efficiency, School of Pharmaceutical Science and Technology, Tianjin University, Weijin Road, Tianjin 300072, China
| | - Yi Zhang
- Tianjin Key Laboratory for Modern Drug Delivery and High-Efficiency, School of Pharmaceutical Science and Technology, Tianjin University, Weijin Road, Tianjin 300072, China
| | - Xinyu Zhang
- Tianjin Key Laboratory for Modern Drug Delivery and High-Efficiency, School of Pharmaceutical Science and Technology, Tianjin University, Weijin Road, Tianjin 300072, China
| | - Xueqian Zhang
- Tianjin Key Laboratory for Modern Drug Delivery and High-Efficiency, School of Pharmaceutical Science and Technology, Tianjin University, Weijin Road, Tianjin 300072, China
| | - Na Liu
- Tianjin Key Laboratory for Modern Drug Delivery and High-Efficiency, School of Pharmaceutical Science and Technology, Tianjin University, Weijin Road, Tianjin 300072, China
| | - Luqi Huang
- Chinese Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Changxiao Liu
- The State Key Laboratories of Pharmacodynamics and Pharmacokinetics, Tianjin 300193, China
| | - Wenyuan Gao
- Tianjin Key Laboratory for Modern Drug Delivery and High-Efficiency, School of Pharmaceutical Science and Technology, Tianjin University, Weijin Road, Tianjin 300072, China.
| |
Collapse
|
8
|
Hejazi SA, Rohampour K, Sharifipour E, Sharifimoghadam S, Paybast S, Ghoreishi A, Hassanzadeh N, Vahedian M. The correlation of serum adiponectin and insulin resistance with the presence and severity of dementia in non-obese Alzheimer's patients. Clin Nutr ESPEN 2020; 40:376-382. [PMID: 33183566 DOI: 10.1016/j.clnesp.2020.07.019] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2020] [Revised: 07/24/2020] [Accepted: 07/30/2020] [Indexed: 01/21/2023]
Abstract
BACKGROUND AND AIMS Alteration in the insulin signaling could contribute to the development of Alzheimer's disease (AD) through metabolic or inflammatory processes, adipokines could affect insulin dysregulation. This study aimed to investigate whether there is a correlation between serum adiponectin level alteration and insulin resistance with the presence and severity of AD, compared to normal controls. METHODS This analytical observational study was conducted on 60 non-overweight and non-diabetic participants who were assigned to AD patients (n = 34) and healthy volunteers (n = 26). The diagnosis and severity of dementia were evaluated by the same protocol, and the Mini-Mental Score Exam (MMSE) questionnaire was utilized to collect the data. Moreover, adiponectin concentration, fasting blood sugar, and plasma insulin levels were measured using enzyme-linked immunosorbent assay. Furthermore, the homeostasis model assessment for insulin resistance (HOMA-IR) was utilized in this study. RESULTS The mean ages of the AD patients and control participants were 71.35 and 70.46, respectively. In addition, the mean values of the serum adiponectin level of the participants were 9660 and 12,730 ng/mL in control and AD groups, respectively (P ≤ 0.05). Additionally, the insulin resistance (IR) was 2.90 and 5.10 in the control and AD groups, respectively (P ≤ 0.05). According to the results, there was a significant positive correlation between serum adiponectin level and HOMA-IR in the AD group; however, no significant correlation was observed between serum adiponectin level and MMSE score in this group. The MMSE score of AD patients significantly decreased by 1.2 times with an increase in each score of the IR (P ≤ 0.05). CONCLUSION A significant direct positive correlation was observed between the serum adiponectin level and IR among the AD patients. However, a significant decrease in cognition levels was detected following an increase in IR scores of the AD patients.
Collapse
Affiliation(s)
- Seyed Amir Hejazi
- Neuroscience Research Center, Qom University of Medical Sciences, Qom, Iran
| | - Kambiz Rohampour
- Neuroscience Research Center, Guilan University of Medical Sciences, Rasht, Iran
| | - Ehsan Sharifipour
- Neuroscience Research Center, Qom University of Medical Sciences, Qom, Iran.
| | | | | | - Abdoreza Ghoreishi
- Department of Neurology, Vali-e-Asr Hospital, School of Medicine, University of Medical Sciences, Zanjan, Iran
| | - Navid Hassanzadeh
- Student Research Committee, Qom University of Medical Sciences, Qom, Iran
| | - Mostafa Vahedian
- Neuroscience Research Center, Qom University of Medical Sciences, Qom, Iran
| |
Collapse
|
9
|
Paroni G, Bisceglia P, Seripa D. Understanding the Amyloid Hypothesis in Alzheimer's Disease. J Alzheimers Dis 2020; 68:493-510. [PMID: 30883346 DOI: 10.3233/jad-180802] [Citation(s) in RCA: 98] [Impact Index Per Article: 19.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
The amyloid hypothesis (AH) is still the most accepted model to explain the pathogenesis of inherited Alzheimer's disease (IAD). However, despite the neuropathological overlapping with the non-inherited form (NIAD), AH waver in explaining NIAD. Thus, 30 years after its first statement several questions are still open, mainly regarding the role of amyloid plaques (AP) and apolipoprotein E (APOE). Accordingly, a pathogenetic model including the role of AP and APOE unifying IAD and NIAD pathogenesis is still missing. In the present understanding of the AH, we suggested that amyloid-β (Aβ) peptides production and AP formation is a physiological aging process resulting from a systemic age-related decrease in the efficiency of the proteins catabolism/clearance machinery. In this pathogenetic model Aβ peptides act as neurotoxic molecules, but only above a critical concentration [Aβ]c. A threshold mechanism triggers IAD/NIAD onset only when [Aβ]≥[Aβ]c. In this process, APOE modifies [Aβ]c threshold in an isoform-specific way. Consequently, all factors influencing Aβ anabolism, such as amyloid beta precursor protein (APP), presenilin 1 (PSEN1), and presenilin 2 (PSEN2) gene mutations, and/or Aβ catabolism/clearance could contribute to exceed the threshold [Aβ]c, being characteristic of each individual. In this model, AP formation does not depend on [Aβ]c. The present interpretation of the AH, unifying the pathogenetic theories for IAD and NIAD, will explain why AP and APOE4 may be observed in healthy aging and why they are not the cause of AD. It is clear that further studies are needed to confirm our pathogenetic model. Nevertheless, our suggestion may be useful to better understand the pathogenesis of AD.
Collapse
Affiliation(s)
- Giulia Paroni
- Research Laboratory, Complex Structure of Geriatrics, Fondazione IRCCS Casa Sollievo della Sofferenza, San Giovanni Rotondo, Foggia, Italy
| | - Paola Bisceglia
- Research Laboratory, Complex Structure of Geriatrics, Fondazione IRCCS Casa Sollievo della Sofferenza, San Giovanni Rotondo, Foggia, Italy
| | - Davide Seripa
- Research Laboratory, Complex Structure of Geriatrics, Fondazione IRCCS Casa Sollievo della Sofferenza, San Giovanni Rotondo, Foggia, Italy
| |
Collapse
|
10
|
Liao Q, Li Q, Zhao Y, Jiang P, Yan Y, Sun H, Liu W, Feng F, Qu W. Design, synthesis and biological evaluation of novel carboline-cinnamic acid hybrids as multifunctional agents for treatment of Alzheimer’s disease. Bioorg Chem 2020; 99:103844. [DOI: 10.1016/j.bioorg.2020.103844] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2020] [Accepted: 04/08/2020] [Indexed: 01/28/2023]
|
11
|
Iraji A, Khoshneviszadeh M, Firuzi O, Khoshneviszadeh M, Edraki N. Novel small molecule therapeutic agents for Alzheimer disease: Focusing on BACE1 and multi-target directed ligands. Bioorg Chem 2020; 97:103649. [PMID: 32101780 DOI: 10.1016/j.bioorg.2020.103649] [Citation(s) in RCA: 65] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2019] [Revised: 01/05/2020] [Accepted: 02/03/2020] [Indexed: 12/17/2022]
Abstract
Alzheimer's Disease (AD) is a progressive neurodegenerative disorder that effects 50 million people worldwide. In this review, AD pathology and the development of novel therapeutic agents targeting AD were fully discussed. In particular, common approaches to prevent Aβ production and/or accumulation in the brain including α-secretase activators, specific γ-secretase modulators and small molecules BACE1 inhibitors were reviewed. Additionally, natural-origin bioactive compounds that provide AD therapeutic advances have been introduced. Considering AD is a multifactorial disease, the therapeutic potential of diverse multi target-directed ligands (MTDLs) that combine the efficacy of cholinesterase (ChE) inhibitors, MAO (monoamine oxidase) inhibitors, BACE1 inhibitors, phosphodiesterase 4D (PDE4D) inhibitors, for the treatment of AD are also reviewed. This article also highlights descriptions on the regulator of serotonin receptor (5-HT), metal chelators, anti-aggregants, antioxidants and neuroprotective agents targeting AD. Finally, current computational methods for evaluating the structure-activity relationships (SAR) and virtual screening (VS) of AD drugs are discussed and evaluated.
Collapse
Affiliation(s)
- Aida Iraji
- Medicinal and Natural Products Chemistry Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Mahsima Khoshneviszadeh
- Medicinal and Natural Products Chemistry Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Omidreza Firuzi
- Medicinal and Natural Products Chemistry Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Mehdi Khoshneviszadeh
- Medicinal and Natural Products Chemistry Research Center, Shiraz University of Medical Sciences, Shiraz, Iran; Department of Medicinal Chemistry, Faculty of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Najmeh Edraki
- Medicinal and Natural Products Chemistry Research Center, Shiraz University of Medical Sciences, Shiraz, Iran.
| |
Collapse
|
12
|
Xu A, He F, Yu C, Qu Y, Zhang Q, Lv J, Zhang X, Ran Y, Wei C, Wu J. The Development of Small Molecule Inhibitors of Glutaminyl Cyclase and Isoglutaminyl Cyclase for Alzheimer's Disease. ChemistrySelect 2019. [DOI: 10.1002/slct.201902852] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Affiliation(s)
- Ana Xu
- College of PharmacyShanDong University, 4 4 West WenHua Road JiNan 250012 China
| | - Feng He
- College of PharmacyShanDong University, 4 4 West WenHua Road JiNan 250012 China
| | - Chenggong Yu
- College of PharmacyShanDong University, 4 4 West WenHua Road JiNan 250012 China
| | - Ying Qu
- College of PharmacyShanDong University, 4 4 West WenHua Road JiNan 250012 China
| | - Qiuqiong Zhang
- College of PharmacyShanDong University, 4 4 West WenHua Road JiNan 250012 China
| | - Jiahui Lv
- College of PharmacyShanDong University, 4 4 West WenHua Road JiNan 250012 China
| | - Xiangna Zhang
- College of PharmacyShanDong University, 4 4 West WenHua Road JiNan 250012 China
| | - Yingying Ran
- College of PharmacyShanDong University, 4 4 West WenHua Road JiNan 250012 China
| | - Chao Wei
- College of PharmacyShanDong University, 4 4 West WenHua Road JiNan 250012 China
| | - Jingde Wu
- College of PharmacyShanDong University, 4 4 West WenHua Road JiNan 250012 China
| |
Collapse
|
13
|
Jaafaru MS, Nordin N, Rosli R, Shaari K, Bako HY, Saad N, Noor NM, Abdull Razis AF. Neuroprotective effects of glucomoringin-isothiocyanate against H 2O 2-Induced cytotoxicity in neuroblastoma (SH-SY5Y) cells. Neurotoxicology 2019; 75:89-104. [PMID: 31521693 DOI: 10.1016/j.neuro.2019.09.008] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2019] [Revised: 09/07/2019] [Accepted: 09/09/2019] [Indexed: 12/26/2022]
Abstract
Neurodegenerative diseases (NDDs) are pathological conditions characterised by progressive damage of neuronal cells leading to eventual loss of structure and function of the cells. Due to implication of multi-systemic complexities of signalling pathways in NDDs, the causes and preventive mechanisms are not clearly delineated. The study was designed to investigate the potential signalling pathways involved in neuroprotective activities of purely isolated glucomoringin isothiocyanate (GMG-ITC) against H2O2-induced cytotoxicity in neuroblastoma (SH-SY5Y) cells. GMG-ITC was isolated from Moringa oleifera seeds, and confirmed with NMR and LC-MS based methods. Gene expression analysis of phase II detoxifying markers revealed significant increase in the expression of all the genes involved, due to GMG-ITC pre-treatment. GMG-ITC also caused significant decreased in the expression of NF-kB, BACE1, APP and increased the expressions of IkB and MAPT tau genes in the differentiated cells as confirmed by multiplex genetic system analysis. The effect was reflected on the expressed proteins in the differentiated cells, where GMG-ITC caused increased in expression level of Nrf2, SOD-1, NQO1, p52 and c-Rel of nuclear factor erythroid factor 2 (Nrf2) and nuclear factor kappa-B (NF-kB) pathways respectively. The findings revealed the potential of GMG-ITC to abrogate oxidative stress-induced neurodegeneration through Nrf2 and NF-kB signalling pathways.
Collapse
Affiliation(s)
- Mohammed Sani Jaafaru
- Laboratory of Molecular Biomedicine, Institute of Bioscience, Universiti Putra Malaysia, 43400, UPM Serdang, Selangor, Malaysia; Department of Biochemistry, Kaduna State University, Main Campus, PMB 2339, Kaduna, Nigeria.
| | - Norshariza Nordin
- Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, 43400, UPM Serdang, Selangor, Malaysia.
| | - Rozita Rosli
- Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, 43400, UPM Serdang, Selangor, Malaysia; UPM-MAKNA Cancer Research Laboratory, Institute of Bioscience, Universiti Putra Malaysia, 43400, UPM Serdang, Selangor, Malaysia.
| | - Khozirah Shaari
- Laboratory of Natural Products, Institute of Bioscience, Universiti Putra Malaysia, 43400, UPM Serdang, Selangor, Malaysia; Department of Chemistry, Faculty of Science, Universiti Putra Malaysia, 43400, UPM Serdang, Selangor, Malaysia.
| | - Hauwa'u Yakubu Bako
- Department of Biochemistry, Kaduna State University, Main Campus, PMB 2339, Kaduna, Nigeria.
| | - Norazalina Saad
- UPM-MAKNA Cancer Research Laboratory, Institute of Bioscience, Universiti Putra Malaysia, 43400, UPM Serdang, Selangor, Malaysia.
| | - Noramaliza Mohd Noor
- Department of Imaging, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, 43400, UPM Serdang, Selangor, Malaysia.
| | - Ahmad Faizal Abdull Razis
- Laboratory of Molecular Biomedicine, Institute of Bioscience, Universiti Putra Malaysia, 43400, UPM Serdang, Selangor, Malaysia; Department of Food Science, Faculty of Food Science and Technology, Universiti Putra Malaysia, 43400, UPM Serdang, Selangor, Malaysia; Laboratory of Food Safety and Food Integrity, Institute of Tropical Agriculture and Food Security, Universiti Putra Malaysia, 43400, UPM Serdang, Selangor, Malaysia.
| |
Collapse
|
14
|
Suppression of gut dysbiosis by Bifidobacterium longum alleviates cognitive decline in 5XFAD transgenic and aged mice. Sci Rep 2019; 9:11814. [PMID: 31413350 PMCID: PMC6694197 DOI: 10.1038/s41598-019-48342-7] [Citation(s) in RCA: 105] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2019] [Accepted: 08/02/2019] [Indexed: 02/07/2023] Open
Abstract
To understand the role of commensal gut bacteria on the progression of cognitive decline in Alzheimer’s disease via the microbiota-gut-brain axis, we isolated anti-inflammatory Bifidobacterium longum (NK46) from human gut microbiota, which potently inhibited gut microbiota endotoxin production and suppressed NF-κB activation in lipopolysaccharide (LPS)-stimulated BV-2 cells, and examined whether NK46 could simultaneously alleviate gut dysbiosis and cognitive decline in male 5xFAD-transgenic (5XFAD-Tg, 6 months-old) and aged (18 months-old) mice. Oral administration of NK46 (1 × 109 CFU/mouse/day for 1 and 2 months in aged and Tg mice, respectively) shifted gut microbiota composition, particularly Proteobacteria, reduced fecal and blood LPS levels, suppressed NF-κB activation and TNF-α expression, and increased tight junction protein expression in the colon of 5XFAD-Tg and aged mice. NK46 treatment also alleviated cognitive decline in 5XFAD-Tg and aged mice. Furthermore, NK46 treatment suppressed amyloid-β, β/γ-secretases, and caspase-3 expression and amyloid-β accumulation in the hippocampus of 5XFAD-Tg mice. NK46 treatment also reduced Iba1+, LPS+/CD11b+, and caspase-3+/NeuN+ cell populations and suppressed NF-κB activation in the hippocampus of 5XFAD-Tg and aged mice, while BDNF expression was increased. These findings suggest that the suppression of gut dysbiosis and LPS production by NK46 can mitigate cognitive decline through the regulation of microbiota LPS-mediated NF-κB activation.
Collapse
|
15
|
Tiwari S, Atluri V, Kaushik A, Yndart A, Nair M. Alzheimer's disease: pathogenesis, diagnostics, and therapeutics. Int J Nanomedicine 2019; 14:5541-5554. [PMID: 31410002 PMCID: PMC6650620 DOI: 10.2147/ijn.s200490] [Citation(s) in RCA: 745] [Impact Index Per Article: 124.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2019] [Accepted: 03/02/2019] [Indexed: 12/12/2022] Open
Abstract
Currently, 47 million people live with dementia globally, and it is estimated to increase more than threefold (~131 million) by 2050. Alzheimer's disease (AD) is one of the major causative factors to induce progressive dementia. AD is a neurodegenerative disease, and its pathogenesis has been attributed to extracellular aggregates of amyloid β (Aβ) plaques and intracellular neurofibrillary tangles made of hyperphosphorylated τ-protein in cortical and limbic areas of the human brain. It is characterized by memory loss and progressive neurocognitive dysfunction. The anomalous processing of APP by β-secretases and γ-secretases leads to production of Aβ40 and Aβ42 monomers, which further oligomerize and aggregate into senile plaques. The disease also intensifies through infectious agents like HIV. Additionally, during disease pathogenesis, the presence of high concentrations of Aβ peptides in central nervous system initiates microglial infiltration. Upon coming into vicinity of Aβ, microglia get activated, endocytose Aβ, and contribute toward their clearance via TREM2 surface receptors, simultaneously triggering innate immunoresponse against the aggregation. In addition to a detailed report on causative factors leading to AD, the present review also discusses the current state of the art in AD therapeutics and diagnostics, including labeling and imaging techniques employed as contrast agents for better visualization and sensing of the plaques. The review also points to an urgent need for nanotechnology as an efficient therapeutic strategy to increase the bioavailability of drugs in the central nervous system.
Collapse
Affiliation(s)
- Sneham Tiwari
- Department of Immunology and Nano-Medicine, Institute of NeuroImmune Pharmacology, Herbert Wertheim College of Medicine, Florida International University, Miami, FL33199, USA
| | - Venkata Atluri
- Department of Immunology and Nano-Medicine, Institute of NeuroImmune Pharmacology, Herbert Wertheim College of Medicine, Florida International University, Miami, FL33199, USA
| | - Ajeet Kaushik
- Department of Immunology and Nano-Medicine, Institute of NeuroImmune Pharmacology, Herbert Wertheim College of Medicine, Florida International University, Miami, FL33199, USA
| | - Adriana Yndart
- Department of Immunology and Nano-Medicine, Institute of NeuroImmune Pharmacology, Herbert Wertheim College of Medicine, Florida International University, Miami, FL33199, USA
| | - Madhavan Nair
- Department of Immunology and Nano-Medicine, Institute of NeuroImmune Pharmacology, Herbert Wertheim College of Medicine, Florida International University, Miami, FL33199, USA
| |
Collapse
|
16
|
Castro MA, Hadziselimovic A, Sanders CR. The vexing complexity of the amyloidogenic pathway. Protein Sci 2019; 28:1177-1193. [PMID: 30897251 PMCID: PMC6566549 DOI: 10.1002/pro.3606] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2019] [Revised: 03/18/2019] [Accepted: 03/18/2019] [Indexed: 02/06/2023]
Abstract
The role of the amyloidogenic pathway in the etiology of Alzheimer's disease (AD), particularly the common sporadic late onset forms of the disease, is controversial. To some degree, this is a consequence of the failure of drug and therapeutic antibody trials based either on targeting the proteases in this pathway or its amyloid end products. Here, we explore the formidable complexity of the biochemistry and cell biology associated with this pathway. For example, we review evidence that the immediate precursor of amyloid-β, the C99 domain of the amyloid precursor protein (APP), may itself be toxic. We also review important new results that appear to finally establish a direct genetic link between mutations in APP and the sporadic forms of AD. Based on the complexity of amyloidogenesis, it seems possible that a major contributor to the failure of related drug trials is that we have an incomplete understanding of this pathway and how it is linked to Alzheimer's pathogenesis. If so, this highlights a need for further characterization of this pathway, not its abandonment.
Collapse
Affiliation(s)
- Manuel A. Castro
- Departments of Biochemistry and MedicineVanderbilt University School of MedicineNashvilleTennessee 37240
| | - Arina Hadziselimovic
- Departments of Biochemistry and MedicineVanderbilt University School of MedicineNashvilleTennessee 37240
| | - Charles R. Sanders
- Departments of Biochemistry and MedicineVanderbilt University School of MedicineNashvilleTennessee 37240
| |
Collapse
|
17
|
Chaubey MG, Patel SN, Rastogi RP, Srivastava PL, Singh AK, Madamwar D, Singh NK. Therapeutic potential of cyanobacterial pigment protein phycoerythrin: in silico and in vitro study of BACE1 interaction and in vivo Aβ reduction. Int J Biol Macromol 2019; 134:368-378. [PMID: 31059742 DOI: 10.1016/j.ijbiomac.2019.05.006] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2018] [Revised: 03/29/2019] [Accepted: 05/02/2019] [Indexed: 12/18/2022]
Abstract
Cyanobacteria are an immense source of innovative classes of pharmacologically active compounds exhibiting various biological activities ranging from antioxidants, antibiotics, anticancer, anti-inflammatory to anti-Alzheimer's disease. In the present study, we primarily targeted the inhibition of Beta-site amyloid precursor protein cleaving enzyme-1 (BACE1) by a naturally occurring cyanobacterial protein phycoerythrin (C-PE). BACE1 cleaves amyloid-β precursor protein (APP) and leads to accumulation of neurotoxic amyloid beta (Aβ) plaques in the brain, as an attribute of Alzheimer's disease (AD). Inhibition of BACE1 was measured in terms of their association and dissociation rate constants, thermodynamics of binding using surface plasmon resonance (SPR) and isothermal titration calorimetry (ITC). The kinetic parameters for enzyme activity were also measured using synthetic decapeptide as a substrate. We further validated the potential of PE by in-vivo histopathological staining of Aβ aggregate mutant Caenorhabditis elegans CL4176 by Thioflavin-T. The present studies pave the way for the application of naturally occurring C-PE as a putative therapeutic drug for the AD.
Collapse
Affiliation(s)
- Mukesh Ghanshyam Chaubey
- Department of Biotechnology, Shree A. N. Patel PG Institute of Science and Research, Sardar Patel University, Anand 388001, Gujarat, India
| | - Stuti Nareshkumar Patel
- Post-Graduate Department of Biosciences, UGC-Centre of Advanced Study, Satellite Campus, Vadtal Road, Sardar Patel University, Bakrol, 388315 Anand, Gujarat, India
| | - Rajesh Prasad Rastogi
- Post-Graduate Department of Biosciences, UGC-Centre of Advanced Study, Satellite Campus, Vadtal Road, Sardar Patel University, Bakrol, 388315 Anand, Gujarat, India
| | - Prabhakar Lal Srivastava
- Symbiosis School of Biological Science, Symbiosis International (Deemed University), Lavale, 412115 Pune, Maharashtra, India
| | - Arun Kumar Singh
- Zydus Research Centre, Sarkhej-Bavla N.H. No. 8A, Moraiya, Sanand, Ahmedabad 382210, India
| | - Datta Madamwar
- Post-Graduate Department of Biosciences, UGC-Centre of Advanced Study, Satellite Campus, Vadtal Road, Sardar Patel University, Bakrol, 388315 Anand, Gujarat, India
| | - Niraj Kumar Singh
- Department of Biotechnology, Shree A. N. Patel PG Institute of Science and Research, Sardar Patel University, Anand 388001, Gujarat, India.
| |
Collapse
|
18
|
McConnell HL, Li Z, Woltjer RL, Mishra A. Astrocyte dysfunction and neurovascular impairment in neurological disorders: Correlation or causation? Neurochem Int 2019; 128:70-84. [PMID: 30986503 DOI: 10.1016/j.neuint.2019.04.005] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2019] [Revised: 04/08/2019] [Accepted: 04/09/2019] [Indexed: 12/14/2022]
Abstract
The neurovascular unit, consisting of neurons, astrocytes, and vascular cells, has become the focus of much discussion in the last two decades and emerging literature now suggests an association between neurovascular dysfunction and neurological disorders. In this review, we synthesize the known and suspected contributions of astrocytes to neurovascular dysfunction in disease. Throughout the brain, astrocytes are centrally positioned to dynamically mediate interactions between neurons and the cerebral vasculature, and play key roles in blood-brain barrier maintenance and neurovascular coupling. It is increasingly apparent that the changes in astrocytes in response to a variety of insults to brain tissue -collectively referred to as "reactive astrogliosis" - are not just an epiphenomenon restricted to morphological alterations, but comprise functional changes in astrocytes that contribute to the phenotype of neurological diseases with both beneficial and detrimental effects. In the context of the neurovascular unit, astrocyte dysfunction accompanies, and may contribute to, blood-brain barrier impairment and neurovascular dysregulation, highlighting the need to determine the exact nature of the relationship between astrocyte dysfunction and neurovascular impairments. Targeting astrocytes may represent a new strategy in combinatorial therapeutics for preventing the mismatch of energy supply and demand that often accompanies neurological disorders.
Collapse
Affiliation(s)
- Heather L McConnell
- Knight Cardiovascular Institute, Oregon Health & Science University, Portland, OR, United States
| | - Zhenzhou Li
- Knight Cardiovascular Institute, Oregon Health & Science University, Portland, OR, United States; Department of Anesthesiology, General Hospital of Ningxia Medical University, Yinchuan City, China
| | - Randall L Woltjer
- Department of Neuropathology, Oregon Health & Science University, Portland, OR, United States
| | - Anusha Mishra
- Knight Cardiovascular Institute, Oregon Health & Science University, Portland, OR, United States.
| |
Collapse
|
19
|
de Oliveira JS, Abdalla FH, Dornelles GL, Palma TV, Signor C, da Silva Bernardi J, Baldissarelli J, Lenz LS, de Oliveira VA, Chitolina Schetinger MR, Melchiors Morsch VM, Rubin MA, de Andrade CM. Neuroprotective effects of berberine on recognition memory impairment, oxidative stress, and damage to the purinergic system in rats submitted to intracerebroventricular injection of streptozotocin. Psychopharmacology (Berl) 2019; 236:641-655. [PMID: 30377748 DOI: 10.1007/s00213-018-5090-6] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/05/2018] [Accepted: 10/21/2018] [Indexed: 12/15/2022]
Abstract
Alzheimer's disease (AD) is a progressive and irreversible neurodegenerative disease. The present study investigated the effects of 50 and 100 mg/kg berberine (BRB) on recognition memory, oxidative stress, and purinergic neurotransmission, in a model of sporadic dementia of the Alzheimer's type induced by intracerebroventricular (ICV) injection of streptozotocin (STZ) in rats. Rats were submitted to ICV-STZ 3 mg/kg or saline, and 3 days later, were started on a treatment of BRB or saline for 21 days. The results demonstrated that BRB was effective in protecting against memory impairment, increased reactive oxygen species, and the subsequent increase in protein and lipid oxidation in the cerebral cortex and hippocampus, as well as δ-aminolevulinate dehydratase inhibition in the cerebral cortex. Moreover, the decrease in total thiols, and the reduced glutathione and glutathione S-transferase activity in the cerebral cortex and hippocampus of ICV-STZ rats, was prevented by BRB treatment. Besides an antioxidant effect, BRB treatment was capable of preventing decreases in ecto-nucleoside triphosphate diphosphohydrolase (NTPDase), 5'-nucleotidase (EC-5'-Nt), and adenosine deaminase (ADA) activities in synaptosomes of the cerebral cortex and hippocampus. Thus, our data suggest that BRB exerts a neuroprotective effect on recognition memory, as well as on oxidative stress and oxidative stress-related damage, such as dysfunction of the purinergic system. This suggests that BRB may act as a promising multipotent agent for the treatment of AD.
Collapse
Affiliation(s)
- Juliana Sorraila de Oliveira
- Programa de Pós Graduação em Ciências Biológicas: Bioquímica Toxicológica, Setor de Bioquímica e Estresse Oxidativo do Laboratório de Terapia Celular, Centro de Ciências Rurais, Universidade Federal de Santa Maria, Santa Maria, RS, Brazil. .,Post-Graduation Program in Toxicological Biochemistry, Department of Chemistry of the Center of Natural and Exact Sciences of the Federal University of Santa Maria, Santa Maria, RS, Brazil.
| | - Fátima Husein Abdalla
- Programa de Pós Graduação em Ciências Biológicas: Bioquímica Toxicológica, Departamento de Bioquímica e Biologia Molecular, Centro de Ciências Naturais e Exatas, Universidade Federal de Santa Maria, Santa Maria, RS, Brazil
| | - Guilherme Lopes Dornelles
- Programa de Pós graduação em Medicina Veterinária, Centro de Ciência Rurais/Departamento de Clínica de Pequenos Animais, Laboratório de Patologia Clínica Veternária/Hospital Veterinário, Universidade Federal de Santa Maria, Santa Maria, RS, Brazil
| | - Taís Vidal Palma
- Programa de Pós Graduação em Ciências Biológicas: Bioquímica Toxicológica, Setor de Bioquímica e Estresse Oxidativo do Laboratório de Terapia Celular, Centro de Ciências Rurais, Universidade Federal de Santa Maria, Santa Maria, RS, Brazil.,Post-Graduation Program in Toxicological Biochemistry, Department of Chemistry of the Center of Natural and Exact Sciences of the Federal University of Santa Maria, Santa Maria, RS, Brazil
| | - Cristiane Signor
- Programa de Pós Graduação em Ciências Biológicas: Bioquímica Toxicológica, Departamento de Bioquímica e Biologia Molecular, Centro de Ciências Naturais e Exatas, Laboratório de Neuropsicofarmacologia Universidade Federal de Santa Maria, Santa Maria, Brazil
| | - Jamile da Silva Bernardi
- Programa de Pós Graduação em Ciências Biológicas: Bioquímica Toxicológica, Departamento de Bioquímica e Biologia Molecular, Centro de Ciências Naturais e Exatas, Universidade Federal de Santa Maria, Santa Maria, RS, Brazil
| | - Jucimara Baldissarelli
- Programa de Pós Graduação em Ciências Biológicas: Bioquímica Toxicológica, Departamento de Bioquímica e Biologia Molecular, Centro de Ciências Naturais e Exatas, Universidade Federal de Santa Maria, Santa Maria, RS, Brazil
| | - Luana Suéling Lenz
- Programa de Pós Graduação em Ciências Biológicas: Bioquímica Toxicológica, Setor de Bioquímica e Estresse Oxidativo do Laboratório de Terapia Celular, Centro de Ciências Rurais, Universidade Federal de Santa Maria, Santa Maria, RS, Brazil.,Post-Graduation Program in Toxicological Biochemistry, Department of Chemistry of the Center of Natural and Exact Sciences of the Federal University of Santa Maria, Santa Maria, RS, Brazil
| | - Vitor Antunes de Oliveira
- Programa de Pós Graduação em Ciências Biológicas: Bioquímica Toxicológica, Departamento de Bioquímica e Biologia Molecular, Centro de Ciências Naturais e Exatas, Universidade Federal de Santa Maria, Santa Maria, RS, Brazil
| | - Maria Rosa Chitolina Schetinger
- Programa de Pós Graduação em Ciências Biológicas: Bioquímica Toxicológica, Departamento de Bioquímica e Biologia Molecular, Centro de Ciências Naturais e Exatas, Universidade Federal de Santa Maria, Santa Maria, RS, Brazil
| | - Vera Maria Melchiors Morsch
- Programa de Pós Graduação em Ciências Biológicas: Bioquímica Toxicológica, Departamento de Bioquímica e Biologia Molecular, Centro de Ciências Naturais e Exatas, Universidade Federal de Santa Maria, Santa Maria, RS, Brazil
| | - Maribel Antonello Rubin
- Programa de Pós Graduação em Ciências Biológicas: Bioquímica Toxicológica, Departamento de Bioquímica e Biologia Molecular, Centro de Ciências Naturais e Exatas, Laboratório de Neuropsicofarmacologia Universidade Federal de Santa Maria, Santa Maria, Brazil
| | - Cinthia Melazzo de Andrade
- Programa de Pós graduação em Medicina Veterinária, Centro de Ciência Rurais/Departamento de Clínica de Pequenos Animais, Laboratório de Patologia Clínica Veternária/Hospital Veterinário, Universidade Federal de Santa Maria, Santa Maria, RS, Brazil.,Department of Small Animal Clinic, Center of Rural Sciences Federal University of Santa Maria, Santa Maria, RS, Brazil
| |
Collapse
|
20
|
An Aβ42 variant that inhibits intra- and extracellular amyloid aggregation and enhances cell viability. Biochem J 2018; 475:3087-3103. [DOI: 10.1042/bcj20180247] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2018] [Revised: 09/02/2018] [Accepted: 09/07/2018] [Indexed: 01/06/2023]
Abstract
Aggregation and accumulation of the 42-residue amyloid β peptide (Aβ42) in the extracellular matrix and within neuronal cells is considered a major cause of neuronal cell cytotoxicity and death in Alzheimer's disease (AD) patients. Therefore, molecules that bind to Aβ42 and prevent its aggregation are therapeutically promising as AD treatment. Here, we show that a non-self-aggregating Aβ42 variant carrying two surface mutations, F19S and L34P (Aβ42DM), inhibits wild-type Aβ42 aggregation and significantly reduces Aβ42-mediated cell cytotoxicity. In addition, Aβ42DM inhibits the uptake and internalization of extracellularly added pre-formed Aβ42 aggregates into cells. This was the case in both neuronal and non-neuronal cells co-expressing Aβ42 and Aβ42DM or following pre-treatment of cells with extracellular soluble forms of the two peptides, even at high Aβ42 to Aβ42DM molar ratios. In cells, Aβ42DM associates with Aβ42, while in vitro, the two soluble recombinant peptides exhibit nano-molar binding affinity. Importantly, Aβ42DM potently suppresses Aβ42 amyloid aggregation in vitro, as demonstrated by thioflavin T fluorescence and transmission electron microscopy for detecting amyloid fibrils. Overall, we present a new approach for inhibiting Aβ42 fibril formation both within and outside cells. Accordingly, Aβ42DM should be evaluated in vivo for potential use as a therapeutic lead for treating AD.
Collapse
|
21
|
Lee HJ, Hwang YH, Kim DH. Lactobacillus plantarum C29-Fermented Soybean (DW2009) Alleviates Memory Impairment in 5XFAD Transgenic Mice by Regulating Microglia Activation and Gut Microbiota Composition. Mol Nutr Food Res 2018; 62:e1800359. [PMID: 30152045 DOI: 10.1002/mnfr.201800359] [Citation(s) in RCA: 55] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2018] [Revised: 07/23/2018] [Indexed: 12/29/2022]
Abstract
SCOPE The study aims to determine whether Lactobacillus plantarum C29-fermented defatted soybean (FDS, DW2009) can attenuate memory impairment in 5XFAD transgenic (Tg) mice. METHODS AND RESULTS Oral administration of FDS or C29 increases cognitive function in Tg mice in passive avoidance, Y-maze, novel object recognition, and Morris water maze tasks. FDS or C29 treatment significantly suppresses amyloid-β, β/γ-secretases, caspase-3 expression, and NF-κB activation, and activates microglia and apoptotic neuron cell populations, and increases BDNF expression in the brain. FDS or C29 treatment suppresses blood and fecal lipopolysaccharide levels and Enterobacteriaceae population and increases lactobacilli/bifidobacteria populations. CONCLUSION FDS and C29 alleviates the decrease in cognitive function and inhibited amyloid-β expression in Tg mice by regulating microglia activation and gut microbiota composition.
Collapse
Affiliation(s)
- Hae-Ji Lee
- Department of Life and Nanopharmaceutical Sciences, College of Pharmacy, Kyung Hee University, 26, Kyungheedae-ro, Dongdaemun-gu, Seoul, 02447, Korea
| | - Yun-Ha Hwang
- DongWha Pharm Research Institute, 35-71, Topsil-ro, Giheung-gu, Yongin-Shi, Gyeonggi, 17084, Korea
| | - Dong-Hyun Kim
- Department of Life and Nanopharmaceutical Sciences, College of Pharmacy, Kyung Hee University, 26, Kyungheedae-ro, Dongdaemun-gu, Seoul, 02447, Korea
| |
Collapse
|
22
|
Kumar D, Ganeshpurkar A, Kumar D, Modi G, Gupta SK, Singh SK. Secretase inhibitors for the treatment of Alzheimer's disease: Long road ahead. Eur J Med Chem 2018; 148:436-452. [DOI: 10.1016/j.ejmech.2018.02.035] [Citation(s) in RCA: 115] [Impact Index Per Article: 16.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2017] [Revised: 01/30/2018] [Accepted: 02/10/2018] [Indexed: 10/18/2022]
|
23
|
Li Z, Liu XC, Li R, Chang J. Reduction of Aβ Generation by Schisandrin B through Restraining Beta-Secretase 1 Transcription and Translation. Med Sci Monit 2018; 24:1219-1224. [PMID: 29487280 PMCID: PMC5841190 DOI: 10.12659/msm.905127] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2017] [Accepted: 08/17/2017] [Indexed: 11/29/2022] Open
Abstract
BACKGROUND Beta-secretase 1 (BACE1) is a rate-limiting enzyme in the generation of amyloid beta peptides, which are associated with Alzheimer's disease (AD). It has been reported that Schisandrin B could improve cognitive functions in animal models of AD, but the underlying mechanisms are not completely understood. MATERIAL AND METHODS In this research, in order to investigate the effects of Schisandrin B on amyloid-β (Aβ) metabolism and its mechanisms, amyloid precursor protein (APP) and its proteolytic products were determined by enzyme-linked immunosorbent assay (ELISA), western blotting, and RT-PCR after incubation of N2a/Swe cells with Schisandrin B. RESULTS The results indicated that Schisandrin B can significantly reduce the level of secretion of Aβ40 and Aβ42 secreted in N2a/Swe cells. Additionally, there was nonsignificant change in APP level after Schisandrin B treatment. Treatment of Schisandrin B dramatically reduced the mRNA and protein expression levels of BACE1. Moreover, Schisandrin B treatment resulted in a reduction of protein level of sAPPβ, an APP fragment cleavage by BACE1. CONCLUSIONS These results suggest that Schisandrin B inhibits the transcription and translation of BACE1, suppresses the activity of BACE1, and ultimately attenuates Aβ generation, which provides a novel mechanism for the regulation of Aβ metabolism by Schisandrin B.
Collapse
Affiliation(s)
- Zeng Li
- School of Pharmacy, Anhui Key Laboratory of Bioactivity of Natural Products, Anhui Medical University, Hefei, Anhui, P.R. China
| | - Xiao-chang Liu
- Department of Pharmacy, Zhuhai Hospital of Guangdong General Hospital, Zhuhai, Guangdong, P.R. China
| | - Rui Li
- Department of Pharmacy, Anhui Academy of Medical Sciences, Hefei, Anhui, P.R. China
| | - Jun Chang
- Department of Orthopedics, The Fourth Affiliated Hospital of Anhui Medical University, Hefei, Anhui, P.R. China
| |
Collapse
|
24
|
Zdarova Karasova J, Mzik M, Hroch M, Korabecny J, Nepovimova E, Vorisek V, Palicka V, Kuca K. The New Acetylcholinesterase Inhibitors PC-37 and PC-48 (7-Methoxytacrine-Donepezil-Like Compounds): Characterization of Their Metabolites in Human Liver Microsomes, Pharmacokinetics and In Vivo Formation of the Major Metabolites in Rats. Basic Clin Pharmacol Toxicol 2017; 122:373-382. [PMID: 29067789 DOI: 10.1111/bcpt.12922] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2017] [Accepted: 10/16/2017] [Indexed: 12/22/2022]
Abstract
The objective of this study was to elucidate the pharmacokinetics and metabolite formation of newly developed non-selective AChE/BChE 7-MEOTA-donepezil-like inhibitors for potential therapeutic use in Alzheimer's disease (AD) patients. The chemical structures of metabolites were defined during incubation with human liver microsomes, and subsequently, the metabolization was verified in in vivo study. In vitro metabolic profiling revealed the formation of nine major metabolites in the case of PC-37 and eight metabolites of PC-48. Hydroxylation and the enzymatic hydrolysis of bonds close to the piperazine ring appeared to be the principal metabolic pathways in vitro. Of these metabolites, M1-M7 of PC-37 and M1-M6 of PC-48 were confirmed under in vivo conditions. Pilot pharmacokinetic experiments in rats were focused on the absorption, distribution and elimination of these compounds. Absorption after i.m. application was relatively fast; the bioavailability expressed as AUCtotal was 28179 ± 4691 min.ng/mL for PC-37 and 23374 ± 4045 min.ng/mL for PC-48. Both compounds showed ability to target the central nervous system, with brain concentrations exceeding those in plasma. The maximal brain concentrations are approximately two times higher than the plasma concentrations. The relatively high brain concentrations persisted throughout the experiment until 24 hr after application. Elimination via the kidneys (urine) significantly exceeded elimination via the liver (bile). All these characteristics are crucial for new candidates intended for AD treatment. The principle metabolic pathways that were verified in the in vivo study do not show any evidence for formation of extremely toxic metabolites, but this needs to be confirmed by further studies.
Collapse
Affiliation(s)
- Jana Zdarova Karasova
- Department of Toxicology and Military Pharmacy, Faculty of Military Health Sciences, University of Defense, Hradec Kralove, Brno, Czech Republic.,Biomedical Research Center, University Hospital, Hradec Kralove, Czech Republic
| | - Martin Mzik
- Institute of Clinical Biochemistry and Diagnostic, University Hospital, Hradec Kralove, Czech Republic
| | - Milos Hroch
- Department of Medical Biochemistry, Faculty of Medicine in Hradec Kralove, Charles University, Hradec Kralove, Czech Republic
| | - Jan Korabecny
- Department of Toxicology and Military Pharmacy, Faculty of Military Health Sciences, University of Defense, Hradec Kralove, Brno, Czech Republic.,Biomedical Research Center, University Hospital, Hradec Kralove, Czech Republic
| | - Eugenie Nepovimova
- Department of Toxicology and Military Pharmacy, Faculty of Military Health Sciences, University of Defense, Hradec Kralove, Brno, Czech Republic
| | - Viktor Vorisek
- Institute of Clinical Biochemistry and Diagnostic, University Hospital, Hradec Kralove, Czech Republic
| | - Vladimir Palicka
- Institute of Clinical Biochemistry and Diagnostic, University Hospital, Hradec Kralove, Czech Republic
| | - Kamil Kuca
- Biomedical Research Center, University Hospital, Hradec Kralove, Czech Republic.,Department of Cellular Biology and Pharmacology, Florida International University, Miami, FL, USA
| |
Collapse
|
25
|
Suprun EV, Radko SP, Farafonova TE, Mitkevich VA, Makarov AA, Archakov AI, Shumyantseva VV. Application of an Electrochemical Method to Evaluation of Amyloid-β Aggregation Inhibitors: Testing the RGKLVFFGR-NH2Peptide Antiaggregant. ELECTROANAL 2017. [DOI: 10.1002/elan.201700499] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Affiliation(s)
- Elena V. Suprun
- Institute of Biomedical Chemistry; Pogodinskaya Street 10/8 Moscow 119121 Russia
| | - Sergey P. Radko
- Institute of Biomedical Chemistry; Pogodinskaya Street 10/8 Moscow 119121 Russia
| | | | - Vladimir A. Mitkevich
- Engelhardt Institute of Molecular Biology; Russian Academy of Sciences; Vavilov Street 32 Moscow 119991 Russia
| | - Alexander A. Makarov
- Engelhardt Institute of Molecular Biology; Russian Academy of Sciences; Vavilov Street 32 Moscow 119991 Russia
| | | | | |
Collapse
|
26
|
Morimoto BH. Therapeutic peptides for CNS indications: Progress and challenges. Bioorg Med Chem 2017; 26:2859-2862. [PMID: 28951091 DOI: 10.1016/j.bmc.2017.09.011] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2017] [Revised: 09/05/2017] [Accepted: 09/08/2017] [Indexed: 10/18/2022]
Abstract
Attacking neurodegeneration and promoting neuroprotection have been the holy grail in neurology for almost 20years and represent an area of high unmet medical need. However, indications like Alzheimer's disease and stroke are areas in drug development fraught with failure. This review will highlight three CNS peptide programs which are tackling targets and indications in which traditional small molecule approaches have been difficult and challenging. The targets for these potential peptide therapeutics include the NMDA receptor, γ-secretase, and cyclin-dependent kinase in which direct inhibition has resulted in on-target (not compound related) problems. For example, direct inhibition of γ-secretase has resulted in gastrointestinal abnormalities and inhibition of the NMDA receptor can result in hallucinations, dizziness, out-of-body sensations, and nightmares. When confronted with show-stopping side effects, the CNS peptide programs profiled in this review strike the problem with intervention and disruption of selective protein-protein interactions. The goal of these peptide programs is to produce selective therapeutics with a better safety profile.
Collapse
Affiliation(s)
- Bruce H Morimoto
- Scientific Affairs, Celerion, Inc., 621 Rose Street, Lincoln, NE 68502, USA.
| |
Collapse
|
27
|
Kuruva CS, Manczak M, Yin X, Ogunmokun G, Reddy AP, Reddy PH. Aqua-soluble DDQ reduces the levels of Drp1 and Aβ and inhibits abnormal interactions between Aβ and Drp1 and protects Alzheimer's disease neurons from Aβ- and Drp1-induced mitochondrial and synaptic toxicities. Hum Mol Genet 2017; 26:3375-3395. [PMID: 28854701 PMCID: PMC5886305 DOI: 10.1093/hmg/ddx226] [Citation(s) in RCA: 61] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2017] [Revised: 05/28/2017] [Accepted: 06/06/2017] [Indexed: 11/14/2022] Open
Abstract
The purpose of our study was to develop a therapeutic target that can reduce Aβ and Drp1 levels, and also can inhibit abnormal interactions between Aβ and Drp1 in AD neurons. To achieve this objective, we designed various compounds and their 3-dimensional molecular structures were introduced into Aβ and Drp1 complex and identified their inhibitory properties against Aβ-Drp1 interaction. Among all, DDQ was selected for further investigation because of 1) its best docking score and 2) its binding capability at interacting sites of Drp1 and Aβ complex. We synthesized DDQ using retro-synthesis and analyzed its structure spectrally. Using biochemical, molecular biology, immunostaining and transmission electron microscopy (TEM) methods, we studied DDQ's beneficial effects in AD neurons. We measured the levels of Aβ and Drp1, Aβ and Drp1 interaction, mRNA and protein levels of mitochondrial dynamics, biogenesis and synaptic genes, mitochondrial function and cell viability and mitochondrial number in DDQ-treated and untreated AD neurons. Our qRT-PCR and immunoblotting analysis revealed that reduced levels of mitochondrial fission and increased fusion, biogenesis and synaptic genes in DDQ-treated AD neurons. Our immunoblotting and immunostaining analyses revealed that Aβ and Drp1 levels were reduced in DDQ-treated AD neurons. Interaction between Aβ and Drp1 is reduced in DDQ-treated AD neurons. Aβ42 levels were significantly reduced in DDQ-treated mutant APPSwe/Ind cells. Mitochondrial number is significantly reduced and mitochondrial length is significantly increased. Mitochondrial function and cell viability were maintained in AD neurons treated with DDQ. These observations indicate that DDQ reduces excessive mitochondrial fragmentation, enhances fusion, biogenesis and synaptic activity and reduces Aβ42 levels and protects AD neurons against Aβ-induced mitochondrial and synaptic toxicities.
Collapse
Affiliation(s)
| | | | | | | | | | - P. Hemachandra Reddy
- Garrison Institute on Aging
- Cell Biology & Biochemistry
- Pharmacology/Neuroscience
- Neurology
- Health
- Speech, Language and Hearing Sciences Departments, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA
- Garrison Institute on Aging, South West Campus, Texas Tech University Health Sciences Center, Lubbock, TX 79413, USA
| |
Collapse
|
28
|
Simoni E, Serafini MM, Caporaso R, Marchetti C, Racchi M, Minarini A, Bartolini M, Lanni C, Rosini M. Targeting the Nrf2/Amyloid-Beta Liaison in Alzheimer's Disease: A Rational Approach. ACS Chem Neurosci 2017; 8:1618-1627. [PMID: 28421738 DOI: 10.1021/acschemneuro.7b00100] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Amyloid is a prominent feature of Alzheimer's disease (AD). Yet, a linear linkage between amyloid-β peptide (Aβ) and the disease onset and progression has recently been questioned. In this context, the crucial partnership between Aβ and Nrf2 pathways is acquiring paramount importance, offering prospects for deciphering the Aβ-centered disease network. Here, we report on a new class of antiaggregating agents rationally designed to simultaneously activate transcription-based antioxidant responses, whose lead 1 showed interesting properties in a preliminary investigation. Relying on the requirements of Aβ recognition, we identified the catechol derivative 12. In SH-SY5Y neuroblastoma cells, 12 combined remarkable free radical scavenger properties to the ability to trigger the Nrf2 pathway and induce the Nrf2-dependent defensive gene NQO1 by means of electrophilic activation of the transcriptional response. Moreover, 12 prevented the formation of cytotoxic stable oligomeric intermediates, being significantly more effective, and per se less toxic, than prototype 1. More importantly, as different chemical features were exploited to regulate Nrf2 and Aβ activities, the two pathways could be tuned independently. These findings point to compound 12 and its derivatives as promising tools for investigating the therapeutic potential of the Nrf2/Aβ cellular network, laying foundation for generating new drug leads to confront AD.
Collapse
Affiliation(s)
- Elena Simoni
- Department of Pharmacy
and Biotechnology, Alma Mater Studiorum-University of Bologna, Via Belmeloro
6, 40126 Bologna, Italy
| | - Melania M. Serafini
- Department of Drug Sciences (Pharmacology
Section), University of Pavia, V.le Taramelli 14, 27100 Pavia, Italy
- Scuola Universitaria Superiore IUSS Pavia, P.zza Vittoria, 15, 27100 Pavia, Italy
| | - Roberta Caporaso
- Department of Pharmacy
and Biotechnology, Alma Mater Studiorum-University of Bologna, Via Belmeloro
6, 40126 Bologna, Italy
| | - Chiara Marchetti
- Department of Pharmacy
and Biotechnology, Alma Mater Studiorum-University of Bologna, Via Belmeloro
6, 40126 Bologna, Italy
| | - Marco Racchi
- Department of Drug Sciences (Pharmacology
Section), University of Pavia, V.le Taramelli 14, 27100 Pavia, Italy
| | - Anna Minarini
- Department of Pharmacy
and Biotechnology, Alma Mater Studiorum-University of Bologna, Via Belmeloro
6, 40126 Bologna, Italy
| | - Manuela Bartolini
- Department of Pharmacy
and Biotechnology, Alma Mater Studiorum-University of Bologna, Via Belmeloro
6, 40126 Bologna, Italy
| | - Cristina Lanni
- Department of Drug Sciences (Pharmacology
Section), University of Pavia, V.le Taramelli 14, 27100 Pavia, Italy
| | - Michela Rosini
- Department of Pharmacy
and Biotechnology, Alma Mater Studiorum-University of Bologna, Via Belmeloro
6, 40126 Bologna, Italy
| |
Collapse
|
29
|
Dual Inhibition of AChE and BChE with the C-5 Substituted Derivative of Meldrum’s Acid: Synthesis, Structure Elucidation, and Molecular Docking Studies. CRYSTALS 2017. [DOI: 10.3390/cryst7070211] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
|
30
|
Zhang L, Tu R, Wang Y, Hu Y, Li X, Cheng X, Yin Y, Li W, Huang H. Early-Life Exposure to Lead Induces Cognitive Impairment in Elder Mice Targeting SIRT1 Phosphorylation and Oxidative Alterations. Front Physiol 2017; 8:446. [PMID: 28706491 PMCID: PMC5489681 DOI: 10.3389/fphys.2017.00446] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2017] [Accepted: 06/13/2017] [Indexed: 11/21/2022] Open
Abstract
Pb is a potential risk factor for cognition, mainly mediated by enhanced oxidative stress. Resveratrol, a natural polyphenol with crucial anti-oxidative property, is recently implicated in preventing cognitive deficits in normal aging and neurodegenerative disorders. Its beneficial effects have been linked to sirtuin 1(SIRT1) activation. The aim of this work is to investigate the possible linkage between alterations in Pb-induced oxidative damage and cognitive impairment by prolonged treatment of resveratrol. Male C57BL/6 mice were given Pb(Ac)2 treatment or deionized H2O for 12 weeks, and subjected to resveratrol gavage at the dose of 50 mg/kgBw•d or vehicle after Pb exposure. Results from biochemical analysis and immunohistofluorescence showed that Pb induced oxidative DNA damage and decreased cortical antioxidant biomarker. As expected, these abnormalities were improved by resveratrol treatment. Morris water maze test, Western blotting, immunohistofluorescence staining and RT-qPCR indicated that resveratrol ameliorated spatial learning and memory deficits with alterations in hippocampal BDNF-TrkB signaling, promoted nuclear localization and phosphorylation of hippocampal SIRT1, partly increased protein levels of AMPK and PGC-1α involving in modulation of antioxidant response in Pb-exposed mice. Our results support the hypothesis that resveratrol could attenuate Pb-induced cognitive impairment which was associated with activating SIRT1 via modulation of oxidative stress. Additionally, resveratrol also repressed the Pb-induce amyloidogenic processing with resultant decline in cortical Aβ1−−40. Noteworthy, such effects were not mediated by resveratrol treatment alone. These findings emphasize the potential of SIRT1 activator as an efficacious dietary intervention to downgrade the Pb-induced neurotoxic lesion.
Collapse
Affiliation(s)
- Lijie Zhang
- College of Public Health, Zhengzhou UniversityZhengzhou, China
| | - Runqi Tu
- College of Public Health, Zhengzhou UniversityZhengzhou, China
| | - Yawei Wang
- College of Public Health, Zhengzhou UniversityZhengzhou, China
| | - Yazhen Hu
- College of Public Health, Zhengzhou UniversityZhengzhou, China
| | - Xing Li
- College of Public Health, Zhengzhou UniversityZhengzhou, China
| | - Xuemin Cheng
- College of Public Health, Zhengzhou UniversityZhengzhou, China
| | - Yanyan Yin
- College of Public Health, Zhengzhou UniversityZhengzhou, China
| | - Wenjie Li
- College of Public Health, Zhengzhou UniversityZhengzhou, China
| | - Hui Huang
- College of Public Health, Zhengzhou UniversityZhengzhou, China
| |
Collapse
|
31
|
First dual AK/GSK-3β inhibitors endowed with antioxidant properties as multifunctional, potential neuroprotective agents. Eur J Med Chem 2017; 138:438-457. [PMID: 28689095 DOI: 10.1016/j.ejmech.2017.06.017] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2017] [Revised: 06/07/2017] [Accepted: 06/08/2017] [Indexed: 02/02/2023]
Abstract
The manuscript deals with the design, synthesis and biological evaluation of novel benzoxazinone-based and indole-based compounds as multifunctional neuroprotective agents. These compounds inhibit human adenosine kinase (hAK) and human glycogen synthase kinase 3 beta (hGSK-3β) enzymes. Computational analysis based on a molecular docking approach underlined the potential structural requirements for simultaneously targeting both proteins' allosteric sites. In silico hints drove the synthesis of appropriately decorated benzoxazinones and indoles (5a-s, and 6a-c) and biochemical analysis revealed their behavior as allosteric inhibitors of hGSK-3β. For both our hit 4 and the best compounds of the series (5c,l and 6b) the potential antioxidant profile was assessed in human neuroblastoma cell lines (IMR 32, undifferentiated and neuronal differentiated), by evaluating the protective effect of selected compounds against H2O2 cytotoxicity and reactive oxygen species (ROS) production. Results showed a strong efficacy of the tested compounds, even at the lower doses, in counteracting the induced oxidative stress (50 μM of H2O2) and in preventing ROS formation. In addition, the tested compounds did not show any cytotoxic effect determined by the LDH release, at the concentration range analyzed (from 0.1 to 50 μM). This study allowed the identification of compound 5l, as the first dual hAK/hGSK-3β inhibitor reported to date. Compound 5l, which behaves as an effective antioxidant, holds promise for the development of new series of potential therapeutic agents for the treatment of neurodegenerative diseases characterized by an innovative pharmacological profile.
Collapse
|