1
|
Carracedo S, Launay A, Dechelle-Marquet PA, Faivre E, Blum D, Delarasse C, Boué-Grabot E. Purinergic-associated immune responses in neurodegenerative diseases. Prog Neurobiol 2024; 243:102693. [PMID: 39579963 DOI: 10.1016/j.pneurobio.2024.102693] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2024] [Revised: 10/28/2024] [Accepted: 11/19/2024] [Indexed: 11/25/2024]
Abstract
The chronic activation of immune cells can participate in the development of pathological conditions such as neurodegenerative diseases including Alzheimer's disease (AD), Multiple Sclerosis (MS), Parkinson's disease (PD), Huntington's disease (HD) and Amyotrophic Lateral Sclerosis (ALS). In recent years, compelling evidence indicates that purinergic signaling plays a key role in neuro-immune cell functions. The extracellular release of adenosine 5'-triphosphate (ATP), and its breakdown products (ADP and adenosine) provide the versatile basis for complex purinergic signaling through the activation of several families of receptors. G-protein coupled adenosine A2A receptors, ionotropic P2X and G-protein coupled P2Y receptors for ATP and other nucleotides are abundant and widely distributed in neurons, microglia, and astrocytes of the central nervous system as well as in peripheral immune cells. These receptors are strongly linked to inflammation, with a functional interplay that may influence the intricate purinergic signaling involved in inflammatory responses. In the present review, we examine the roles of the purinergic receptors in neuro-immune cell functions with particular emphasis on A2AR, P2X4 and P2X7 and their possible relevance to specific neurodegenerative disorders. Understanding the molecular mechanisms governing purinergic receptor interaction will be crucial for advancing the development of effective immunotherapies targeting neurodegenerative diseases.
Collapse
Affiliation(s)
- Sara Carracedo
- Univ. Bordeaux, CNRS, IMN, UMR 5293, Bordeaux F-33000, France
| | - Agathe Launay
- Université de Lille, Inserm, CHU Lille, U1172, LilNCog, "Alzheimer & Tauopathies", LabEx DISTALZ, Lille F-59000, France
| | | | - Emilie Faivre
- Université de Lille, Inserm, CHU Lille, U1172, LilNCog, "Alzheimer & Tauopathies", LabEx DISTALZ, Lille F-59000, France
| | - David Blum
- Université de Lille, Inserm, CHU Lille, U1172, LilNCog, "Alzheimer & Tauopathies", LabEx DISTALZ, Lille F-59000, France
| | - Cécile Delarasse
- Sorbonne Université, Inserm, CNRS, Institut de la Vision, 17, rue Moreau, Paris F-75012, France
| | | |
Collapse
|
2
|
Vohra A, Keefe P, Puthanveetil P. Altered Metabolic Signaling and Potential Therapies in Polyglutamine Diseases. Metabolites 2024; 14:320. [PMID: 38921455 PMCID: PMC11205831 DOI: 10.3390/metabo14060320] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Revised: 05/17/2024] [Accepted: 05/27/2024] [Indexed: 06/27/2024] Open
Abstract
Polyglutamine diseases comprise a cluster of genetic disorders involving neurodegeneration and movement disabilities. In polyglutamine diseases, the target proteins become aberrated due to polyglutamine repeat formation. These aberrant proteins form the root cause of associated complications. The metabolic regulation during polyglutamine diseases is not well studied and needs more attention. We have brought to light the significance of regulating glutamine metabolism during polyglutamine diseases, which could help in decreasing the neuronal damage associated with excess glutamate and nucleotide generation. Most polyglutamine diseases are accompanied by symptoms that occur due to excess glutamate and nucleotide accumulation. Along with a dysregulated glutamine metabolism, the Nicotinamide adenine dinucleotide (NAD+) levels drop down, and, under these conditions, NAD+ supplementation is the only achievable strategy. NAD+ is a major co-factor in the glutamine metabolic pathway, and it helps in maintaining neuronal homeostasis. Thus, strategies to decrease excess glutamate and nucleotide generation, as well as channelizing glutamine toward the generation of ATP and the maintenance of NAD+ homeostasis, could aid in neuronal health. Along with understanding the metabolic dysregulation that occurs during polyglutamine diseases, we have also focused on potential therapeutic strategies that could provide direct benefits or could restore metabolic homeostasis. Our review will shed light into unique metabolic causes and into ideal therapeutic strategies for treating complications associated with polyglutamine diseases.
Collapse
Affiliation(s)
- Alisha Vohra
- Chicago College of Osteopathic Medicine, Midwestern University, Downers Grove, IL 60515, USA; (A.V.); (P.K.)
| | - Patrick Keefe
- Chicago College of Osteopathic Medicine, Midwestern University, Downers Grove, IL 60515, USA; (A.V.); (P.K.)
| | - Prasanth Puthanveetil
- College of Graduate Studies, Department of Pharmacology, Midwestern University, Downers Grove, IL 60515, USA
| |
Collapse
|
3
|
Ye H, Zhao J, Xu X, Zhang D, Shen H, Wang S. Role of adenosine A2a receptor in cancers and autoimmune diseases. Immun Inflamm Dis 2023; 11:e826. [PMID: 37102661 PMCID: PMC10091380 DOI: 10.1002/iid3.826] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2022] [Revised: 02/28/2023] [Accepted: 03/13/2023] [Indexed: 04/28/2023] Open
Abstract
Adenosine receptors are P1 class of purinergic receptors that belong to G protein-coupled receptors. There are 4 subtypes of adenosine receptors, namely A1, A2A, A2B, and A3. A2AR has a high affinity for the ligand adenosine. Under pathological conditions or external stimuli, ATP is sequentially hydrolyzed to adenosine by CD39 and CD73. The combination of adenosine and A2AR can increase the concentration of cAMP and activate a series of downstream signaling pathways, and further playing the role of immunosuppression and promotion of tumor invasion. A2AR is expressed to some extent on various immune cells, where it is abnormally expressed on immune cells in cancers and autoimmune diseases. A2AR expression also correlates with disease progression. Inhibitors and agonists of A2AR may be potential new strategies for treatment of cancers and autoimmune diseases. We herein briefly reviewed the expression and distribution of A2AR, adenosine/A2AR signaling pathway, expression, and potential as a therapeutic target.
Collapse
Affiliation(s)
- Hongling Ye
- Department of Clinical Laboratory MedicineNanjing Drum Tower Hospital, Medical School of Nanjing UniversityNanjingJiangsuP.R. China
| | - Junqi Zhao
- Department of Clinical Laboratory MedicineNanjing Drum Tower Hospital, Medical School of Nanjing UniversityNanjingJiangsuP.R. China
| | - Xuejing Xu
- Department of Clinical Laboratory MedicineNanjing Drum Tower Hospital, Medical School of Nanjing UniversityNanjingJiangsuP.R. China
| | - Dagan Zhang
- Department of Clinical Laboratory MedicineNanjing Drum Tower Hospital, Medical School of Nanjing UniversityNanjingJiangsuP.R. China
| | - Han Shen
- Department of Clinical Laboratory MedicineNanjing Drum Tower Hospital, Medical School of Nanjing UniversityNanjingJiangsuP.R. China
| | - Sen Wang
- Department of Clinical Laboratory MedicineNanjing Drum Tower Hospital, Medical School of Nanjing UniversityNanjingJiangsuP.R. China
| |
Collapse
|
4
|
Bhat SA, Ahamad S, Dar NJ, Siddique YH, Nazir A. The Emerging Landscape of Natural Small-molecule Therapeutics for Huntington's Disease. Curr Neuropharmacol 2023; 21:867-889. [PMID: 36797612 PMCID: PMC10227909 DOI: 10.2174/1570159x21666230216104621] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Revised: 11/12/2022] [Accepted: 11/18/2022] [Indexed: 02/18/2023] Open
Abstract
Huntington's disease (HD) is a rare and fatal neurodegenerative disorder with no diseasemodifying therapeutics. HD is characterized by extensive neuronal loss and is caused by the inherited expansion of the huntingtin (HTT) gene that encodes a toxic mutant HTT (mHTT) protein having expanded polyglutamine (polyQ) residues. Current HD therapeutics only offer symptomatic relief. In fact, Food and Drug Administration (FDA) approved two synthetic small-molecule VMAT2 inhibitors, tetrabenazine (1) and deutetrabenazine (2), for managing HD chorea and various other diseases in clinical trials. Therefore, the landscape of drug discovery programs for HD is evolving to discover disease- modifying HD therapeutics. Likewise, numerous natural products are being evaluated at different stages of clinical development and have shown the potential to ameliorate HD pathology. The inherent anti-inflammatory and antioxidant properties of natural products mitigate the mHTT-induced oxidative stress and neuroinflammation, improve mitochondrial functions, and augment the anti-apoptotic and pro-autophagic mechanisms for increased survival of neurons in HD. In this review, we have discussed HD pathogenesis and summarized the anti-HD clinical and pre-clinical natural products, focusing on their therapeutic effects and neuroprotective mechanism/s.
Collapse
Affiliation(s)
| | - Shakir Ahamad
- Department of Chemistry, Aligarh Muslim University, Aligarh, U.P., India
| | - Nawab John Dar
- School of Medicine, UT Health San Antonio, Texas, TX, USA
| | | | - Aamir Nazir
- Division of Neuroscience and Ageing Biology, CSIR-Central Drug Research Institute, Lucknow, U.P., India
- Academy of Scientific and Innovative Research, New Delhi, India
| |
Collapse
|
5
|
Ahamad S, Bhat SA. The Emerging Landscape of Small-Molecule Therapeutics for the Treatment of Huntington's Disease. J Med Chem 2022; 65:15993-16032. [PMID: 36490325 DOI: 10.1021/acs.jmedchem.2c00799] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Huntington's disease (HD) is a progressive neurodegenerative disorder caused by a CAG repeat expansion in the huntingtin gene (HTT). The new insights into HD's cellular and molecular pathways have led to the identification of numerous potent small-molecule therapeutics for HD therapy. The field of HD-targeting small-molecule therapeutics is accelerating, and the approval of these therapeutics to combat HD may be expected in the near future. For instance, preclinical candidates such as naphthyridine-azaquinolone, AN1, AN2, CHDI-00484077, PRE084, EVP4593, and LOC14 have shown promise for further optimization to enter into HD clinical trials. This perspective aims to summarize the advent of small-molecule therapeutics at various stages of clinical development for HD therapy, emphasizing their structure and design, therapeutic effects, and specific mechanisms of action. Further, we have highlighted the key drivers involved in HD pathogenesis to provide insights into the basic principle for designing promising anti-HD therapeutic leads.
Collapse
Affiliation(s)
- Shakir Ahamad
- Department of Chemistry, Aligarh Muslim University, Aligarh, Uttar Pradesh202002, India
| | - Shahnawaz A Bhat
- Department of Zoology, Aligarh Muslim University, Aligarh, Uttar Pradesh202002, India
| |
Collapse
|
6
|
Pasquini S, Contri C, Cappello M, Borea PA, Varani K, Vincenzi F. Update on the recent development of allosteric modulators for adenosine receptors and their therapeutic applications. Front Pharmacol 2022; 13:1030895. [PMID: 36278183 PMCID: PMC9581118 DOI: 10.3389/fphar.2022.1030895] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Accepted: 09/20/2022] [Indexed: 11/13/2022] Open
Abstract
Adenosine receptors (ARs) have been identified as promising therapeutic targets for countless pathological conditions, spanning from inflammatory diseases to central nervous system disorders, from cancer to metabolic diseases, from cardiovascular pathologies to respiratory diseases, and beyond. This extraordinary therapeutic potential is mainly due to the plurality of pathophysiological actions of adenosine and the ubiquitous expression of its receptors. This is, however, a double-edged sword that makes the clinical development of effective ligands with tolerable side effects difficult. Evidence of this is the low number of AR agonists or antagonists that have reached the market. An alternative approach is to target allosteric sites via allosteric modulators, compounds endowed with several advantages over orthosteric ligands. In addition to the typical advantages of allosteric modulators, those acting on ARs could benefit from the fact that adenosine levels are elevated in pathological tissues, thus potentially having negligible effects on normal tissues where adenosine levels are maintained low. Several A1 and various A3AR allosteric modulators have been identified so far, and some of them have been validated in different preclinical settings, achieving promising results. Less fruitful, instead, has been the discovery of A2A and A2BAR allosteric modulators, although the results obtained up to now are encouraging. Collectively, data in the literature suggests that allosteric modulators of ARs could represent valuable pharmacological tools, potentially able to overcome the limitations of orthosteric ligands.
Collapse
Affiliation(s)
- Silvia Pasquini
- Department of Chemical, Pharmaceutical and Agricultural Sciences, University of Ferrara, Ferrara, Italy
| | - Chiara Contri
- Department of Translational Medicine, University of Ferrara, Ferrara, Italy
| | - Martina Cappello
- Department of Translational Medicine, University of Ferrara, Ferrara, Italy
| | | | - Katia Varani
- Department of Translational Medicine, University of Ferrara, Ferrara, Italy
- *Correspondence: Katia Varani,
| | - Fabrizio Vincenzi
- Department of Translational Medicine, University of Ferrara, Ferrara, Italy
| |
Collapse
|
7
|
Tang ML, Wen ZH, Wang JH, Wang ML, Zhang H, Liu XH, Jin L, Chang J. Discovery of Pyridone-Substituted Triazolopyrimidine Dual A 2A/A 1 AR Antagonists for the Treatment of Ischemic Stroke. ACS Med Chem Lett 2022; 13:436-442. [PMID: 35295085 PMCID: PMC8919384 DOI: 10.1021/acsmedchemlett.1c00599] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Accepted: 02/16/2022] [Indexed: 11/28/2022] Open
Abstract
![]()
Ischemic stroke is
a complex systemic disease characterized by
high morbidity, disability, and mortality. The activation of the presynaptic
adenosine A2A and A1 receptors modifies a variety
of brain insults from excitotoxicity to stroke. Therefore, the discovery
of dual A2A/A1 adenosine receptor (AR)-targeting
therapeutic compounds could be a strategy for the treatment of ischemic
stroke. Inspired by two clinical phase III drugs, ASP-5854 (dual A2A/A1 AR antagonist) and preladenant (selective
A2A AR antagonist), and using the hybrid medicinal strategy,
we characterized novel pyridone-substituted triazolopyrimidine scaffolds
as dual A2A/A1 AR antagonists. Among them, compound 1a exerted excellent A2A/A1 AR binding
affinity (Ki = 5.58/24.2
nM), an antagonistic effect (IC50 = 5.72/25.9 nM), and
good metabolic stability in human liver microsomes, rat liver microsomes,
and dog liver microsomes. Importantly, compound 1a demonstrated
a dose–effect relationship in the oxygen-glucose deprivation/reperfusion
(OGD/R)-treated HT22 cell model. These findings support the development
of dual A2A/A1 AR antagonists as a potential
treatment for ischemic stroke.
Collapse
Affiliation(s)
- Mei-Lin Tang
- School of Pharmacy, Human Phenome Institute, Fudan University, 826 Zhangheng Road, Shanghai 201203, China
| | - Zi-Hao Wen
- School of Pharmacy, Human Phenome Institute, Fudan University, 826 Zhangheng Road, Shanghai 201203, China
| | - Jing-Huan Wang
- School of Pharmacy, Human Phenome Institute, Fudan University, 826 Zhangheng Road, Shanghai 201203, China
| | - Mei-Ling Wang
- School of Pharmacy, Human Phenome Institute, Fudan University, 826 Zhangheng Road, Shanghai 201203, China
| | - Heyanhao Zhang
- School of Pharmacy, Human Phenome Institute, Fudan University, 826 Zhangheng Road, Shanghai 201203, China
| | - Xin-Hua Liu
- School of Pharmacy, Human Phenome Institute, Fudan University, 826 Zhangheng Road, Shanghai 201203, China
| | - Lin Jin
- Department of Anesthesia, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai 200032, China
| | - Jun Chang
- School of Pharmacy, Human Phenome Institute, Fudan University, 826 Zhangheng Road, Shanghai 201203, China
| |
Collapse
|
8
|
N 6-substituated adenosine analog J4 attenuates anxiety-like behaviors in mice. Psychopharmacology (Berl) 2022; 239:887-895. [PMID: 35102423 PMCID: PMC9063204 DOI: 10.1007/s00213-022-06079-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/14/2021] [Accepted: 01/27/2022] [Indexed: 10/19/2022]
Abstract
RATIONALE Withdrawal from chronic alcohol exposure produces various physical and mental withdrawal symptoms. Activation of adenosine receptors is known to inhibit withdrawal-induced excitation. However, limited studies investigate how adenosine analogs may prove helpful tools to alleviate alcohol withdrawal-related affective behaviors. OBJECTIVES This study aimed to investigate the effects of J4 compared with saline using the mice vapor or voluntary ethanol drinking model on behavioral endpoints representing ethanol-withdrawal negative emotionality commonly observed during abstinence from chronic alcohol use. METHODS We subjected C57BL/6 J mice to chronic intermittent ethanol (CIE) exposure schedule to investigate how 72-h withdrawal from alcohol alters affective-like behavior. Next, we determined how treatment with J4, a second-generation adenosine analog, influenced affective behaviors produced by alcohol withdrawal. Finally, we determined how J4 treatment alters voluntary ethanol drinking using the two-bottle-choice drinking paradigm. RESULTS Our results show that 72-h withdrawal from chronic intermittent ethanol exposure produces limited affective-like disturbances in male C57BL/6 J mice exposed to 4 cycles ethanol vapor. Most importantly, J4 treatment irrespective of ethanol exposure decreases innate anxiety-like behavior in mice. CONCLUSIONS Withdrawal from chronic intermittent ethanol exposure and subsequent behavioral testing 72 h later produces minimal affective-like behavior. J4 treatment did however reduce marble-burying behavior and increased time spent in open arms of the elevated plus maze, suggesting J4 may be useful as a general anxiolytic.
Collapse
|
9
|
Emerging roles of dysregulated adenosine homeostasis in brain disorders with a specific focus on neurodegenerative diseases. J Biomed Sci 2021; 28:70. [PMID: 34635103 PMCID: PMC8507231 DOI: 10.1186/s12929-021-00766-y] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2021] [Accepted: 10/04/2021] [Indexed: 02/07/2023] Open
Abstract
In modern societies, with an increase in the older population, age-related neurodegenerative diseases have progressively become greater socioeconomic burdens. To date, despite the tremendous effort devoted to understanding neurodegenerative diseases in recent decades, treatment to delay disease progression is largely ineffective and is in urgent demand. The development of new strategies targeting these pathological features is a timely topic. It is important to note that most degenerative diseases are associated with the accumulation of specific misfolded proteins, which is facilitated by several common features of neurodegenerative diseases (including poor energy homeostasis and mitochondrial dysfunction). Adenosine is a purine nucleoside and neuromodulator in the brain. It is also an essential component of energy production pathways, cellular metabolism, and gene regulation in brain cells. The levels of intracellular and extracellular adenosine are thus tightly controlled by a handful of proteins (including adenosine metabolic enzymes and transporters) to maintain proper adenosine homeostasis. Notably, disruption of adenosine homeostasis in the brain under various pathophysiological conditions has been documented. In the past two decades, adenosine receptors (particularly A1 and A2A adenosine receptors) have been actively investigated as important drug targets in major degenerative diseases. Unfortunately, except for an A2A antagonist (istradefylline) administered as an adjuvant treatment with levodopa for Parkinson's disease, no effective drug based on adenosine receptors has been developed for neurodegenerative diseases. In this review, we summarize the emerging findings on proteins involved in the control of adenosine homeostasis in the brain and discuss the challenges and future prospects for the development of new therapeutic treatments for neurodegenerative diseases and their associated disorders based on the understanding of adenosine homeostasis.
Collapse
|
10
|
Hsu WH, Huang NK, Shiao YJ, Lu CK, Chao YM, Huang YJ, Yeh CH, Lin YL. Gastrodiae rhizoma attenuates brain aging via promoting neuritogenesis and neurodifferentiation. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2021; 87:153576. [PMID: 33985879 DOI: 10.1016/j.phymed.2021.153576] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/04/2020] [Revised: 03/23/2021] [Accepted: 04/15/2021] [Indexed: 06/12/2023]
Abstract
BACKGROUND Gastrodiae Rhizoma (Tianma), the dried tuber of Gastrodia elata Bl. (Orchidaceae), is listed as a top-grade herbal medicine in Shen-nong Ben-ts'ao Jing and has been used for treating headaches, dizziness, vertigo and convulsion. It has a neuroprotective effect and extends the lifespan in mouse models of Huntington's disease and Niemann-Pick type C disease. However, its effect on senescence remains unknown. PURPOSE This study aimed to investigate the anti-aging effects and the underlying mechanism of Gastrodiae Rhizoma. METHODS D-galactose (D-gal)- and BeSO4-induced cellular senescence and senescence-associated β-galactosidase (SA-β-gal) activity were evaluated in SH-SY5Y and PC12 cells. D-gal-induced aging mice were used as an in vivo model. Animal behaviors including nesting and burrowing and Morris water maze were conducted. Neurogenesis in the hippocampus was assessed by immunohistochemistry and confocal microscopy, and the aging-related proteins were assessed by Western blot analysis. The potential neuritogenesis activity of the partially purified fraction of Gastrodiae Rhizoma (TM-2) and its major ingredients were investigated in PC12 cells. RESULTS TM-2 could improve D-gal-induced learning and memory impairement by inhibiting oxidative stress, increasing hippocampal neurogenesis and regulating the SH2B1-Akt pathway. Moreover, N6-(4-hydroxybenzyl)adenine riboside (T1-11) and parishins A and B, three constituents of TM-2, had anti-aging activity, as did T1-11 and parishin A induced neuritogenesis. CONCLUSION Our data suggested that TM-2 slowed down D-gal-induced cellular and mouse brain aging. These results indicate that Gastrodiae Rhizoma has a beneficial effect on senescence. It may be used for neuroprotection and promoting neurogenesis.
Collapse
Affiliation(s)
- Wei-Hsiang Hsu
- Department of Chinese Pharmaceutical Sciences and Chinese Medicine Resources, China Medical University, Taichung 40402, Taiwan
| | - Nai-Kuei Huang
- National Research Institute of Chinese Medicine, Ministry of Health and Welfare, Taipei 11221, Taiwan
| | - Young-Ji Shiao
- National Research Institute of Chinese Medicine, Ministry of Health and Welfare, Taipei 11221, Taiwan; Institute of Biopharmaceutical Science, National Yang Ming Chiao Tung University, Taipei 11221, Taiwan
| | - Chung-Kuang Lu
- National Research Institute of Chinese Medicine, Ministry of Health and Welfare, Taipei 11221, Taiwan
| | - Yen-Ming Chao
- Department of Chinese Pharmaceutical Sciences and Chinese Medicine Resources, China Medical University, Taichung 40402, Taiwan
| | - Yi-Jeng Huang
- Department of Chinese Pharmaceutical Sciences and Chinese Medicine Resources, China Medical University, Taichung 40402, Taiwan
| | - Chih-Hsin Yeh
- Taoyuan District Agricultural Research and Extension Station, Council of Agriculture, Executive Yuan, Taoyuan 32754, Taiwan
| | - Yun-Lian Lin
- Department of Chinese Pharmaceutical Sciences and Chinese Medicine Resources, China Medical University, Taichung 40402, Taiwan; Department of Pharmacy, National Taiwan University, Taipei 10050, Taiwan.
| |
Collapse
|
11
|
Abstract
Nucleosides play central roles in all facets of life, from metabolism to cellular signaling. Because of their physiochemical properties, nucleosides are lipid bilayer impermeable and thus rely on dedicated transport systems to cross biological membranes. In humans, two unrelated protein families mediate nucleoside membrane transport: the concentrative and equilibrative nucleoside transporter families. The objective of this review is to provide a broad outlook on the current status of nucleoside transport research. We will discuss the role played by nucleoside transporters in human health and disease, with emphasis placed on recent structural advancements that have revealed detailed molecular principles of these important cellular transport systems and exploitable pharmacological features.
Collapse
Affiliation(s)
- Nicholas J. Wright
- Department of Biochemistry, Duke University Medical Center, 303 Research Drive, Durham, North Carolina, 27710, USA
| | - Seok-Yong Lee
- Department of Biochemistry, Duke University Medical Center, 303 Research Drive, Durham, North Carolina, 27710, USA
- Correspondence and requests for materials should be addressed to: S.-Y. Lee., , tel: 919-684-1005, fax: 919-684-8885
| |
Collapse
|
12
|
Salman MM, Al-Obaidi Z, Kitchen P, Loreto A, Bill RM, Wade-Martins R. Advances in Applying Computer-Aided Drug Design for Neurodegenerative Diseases. Int J Mol Sci 2021; 22:4688. [PMID: 33925236 PMCID: PMC8124449 DOI: 10.3390/ijms22094688] [Citation(s) in RCA: 61] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Revised: 04/26/2021] [Accepted: 04/26/2021] [Indexed: 12/11/2022] Open
Abstract
Neurodegenerative diseases (NDs) including Alzheimer's disease, Parkinson's disease, amyotrophic lateral sclerosis, and Huntington's disease are incurable and affect millions of people worldwide. The development of treatments for this unmet clinical need is a major global research challenge. Computer-aided drug design (CADD) methods minimize the huge number of ligands that could be screened in biological assays, reducing the cost, time, and effort required to develop new drugs. In this review, we provide an introduction to CADD and examine the progress in applying CADD and other molecular docking studies to NDs. We provide an updated overview of potential therapeutic targets for various NDs and discuss some of the advantages and disadvantages of these tools.
Collapse
Affiliation(s)
- Mootaz M. Salman
- Department of Physiology, Anatomy and Genetics, University of Oxford, Parks Road, Oxford OX1 3QX, UK;
- Oxford Parkinson’s Disease Centre, University of Oxford, South Parks Road, Oxford OX1 3QX, UK
| | - Zaid Al-Obaidi
- Department of Pharmaceutical Chemistry, College of Pharmacy, University of Alkafeel, Najaf 54001, Iraq;
- Department of Chemistry and Biochemistry, College of Medicine, University of Kerbala, Karbala 56001, Iraq
| | - Philip Kitchen
- School of Biosciences, College of Health and Life Sciences, Aston University, Aston Triangle, Birmingham B4 7ET, UK; (P.K.); (R.M.B.)
| | - Andrea Loreto
- Department of Physiology, Anatomy and Genetics, University of Oxford, Parks Road, Oxford OX1 3QX, UK;
- John Van Geest Centre for Brain Repair, University of Cambridge, Cambridge CB2 0PY, UK
| | - Roslyn M. Bill
- School of Biosciences, College of Health and Life Sciences, Aston University, Aston Triangle, Birmingham B4 7ET, UK; (P.K.); (R.M.B.)
| | - Richard Wade-Martins
- Department of Physiology, Anatomy and Genetics, University of Oxford, Parks Road, Oxford OX1 3QX, UK;
- Oxford Parkinson’s Disease Centre, University of Oxford, South Parks Road, Oxford OX1 3QX, UK
| |
Collapse
|
13
|
Lum PT, Sekar M, Gan SH, Bonam SR, Shaikh MF. Protective Effect of Natural Products against Huntington's Disease: An Overview of Scientific Evidence and Understanding Their Mechanism of Action. ACS Chem Neurosci 2021; 12:391-418. [PMID: 33475334 DOI: 10.1021/acschemneuro.0c00824] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Huntington's disease (HD), a neurodegenerative disease, normally starts in the prime of adult life, followed by a gradual occurrence of characteristic psychiatric disturbances and cognitive and motor dysfunction. To the best of our knowledge, there is no treatment available to completely mitigate the progression of HD. Among various therapeutic approaches, exhaustive literature reports have confirmed the medicinal benefits of natural products in HD experimental models. Building on this information, this review presents a brief overview of the neuroprotective mechanism(s) of natural products against in vitro/in vivo models of HD. Relevant studies were identified from several scientific databases, including PubMed, ScienceDirect, Scopus, and Google Scholar. After screening through literature from 2005 to the present, a total of 14 medicinal plant species and 30 naturally isolated compounds investigated against HD based on either in vitro or in vivo models were included in the present review. Behavioral outcomes in the HD in vivo model showed that natural compounds significantly attenuated 3-nitropropionic acid (3-NP) induced memory loss and motor incoordination. The biochemical alteration has been markedly alleviated with reduced lipid peroxidation, increased endogenous enzymatic antioxidants, reduced acetylcholinesterase activity, and increased mitochondrial energy production. Interestingly, following treatment with certain natural products, 3-NP-induced damage in the striatum was ameliorated, as seen histologically. Overall, natural products afforded varying degrees of neuroprotection in preclinical studies of HD via antioxidant and anti-inflammatory properties, preservation of mitochondrial function, inhibition of apoptosis, and induction of autophagy.
Collapse
Affiliation(s)
- Pei Teng Lum
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy and Health Sciences, Universiti Kuala Lumpur Royal College of Medicine Perak, Ipoh, 30450 Perak, Malaysia
| | - Mahendran Sekar
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy and Health Sciences, Universiti Kuala Lumpur Royal College of Medicine Perak, Ipoh, 30450 Perak, Malaysia
| | - Siew Hua Gan
- School of Pharmacy, Monash University Malaysia, Bandar Sunway, 47500 Selangor Darul Ehsan, Malaysia
| | - Srinivasa Reddy Bonam
- Institut National de la Santé et de la Recherche Médicale, Centre de Recherche des Cordeliers, Equipe-Immunopathologie et Immunointervention Thérapeutique, Sorbonne Université, Université de Paris, Paris 75006, France
| | - Mohd. Farooq Shaikh
- Neuropharmacology Research Strength, Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Bandar Sunway, 47500 Selangor, Malaysia
| |
Collapse
|
14
|
Jou SB, Tsai CJ, Fang CY, Yi PL, Chang FC. Effects of N 6 -(4-hydroxybenzyl) adenine riboside in stress-induced insomnia in rodents. J Sleep Res 2020; 30:e13156. [PMID: 32748529 DOI: 10.1111/jsr.13156] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2020] [Revised: 05/20/2020] [Accepted: 06/11/2020] [Indexed: 11/30/2022]
Abstract
Adenosine exhibits a somnogenic effect; however, there is no adenosinergic hypnotic because of cardiovascular effects. This study investigated whether N6-(4-hydroxybenzyl) adenine riboside (T1-11), extracted from Gastrodia elata, produces somnogenic effects in rodents. We determined the involvement of adenosine 2A receptors (A2ARs) in GABAergic neurons of the ventrolateral preoptic area (VLPO) and the cardiovascular effects. Change of cage bedding is employed as a stressor to induce insomnia in rodents, and electroencephalograms and electromyograms were used to acquire and analyse sleep-wake activity. We found that intracerebroventricular administration of T1-11 before a dark period increased non-rapid eye movement (NREM) and rapid eye movement (REM) sleep during a dark period, and T1-11-induced sleep increases were blocked by the A2AR antagonist, SCH58261, in naïve rats. Oral administration of T1-11 increased NREM sleep during both dark and light periods. Microinjection of the A2AR antagonist, SCH58261, into the VLPO blocked sleep effects of T1-11. In addition to the somnogenic effect in naïve mice, T1-11 suppressed the stress-induced insomnia and this suppressive effect was blocked by SCH58261. C-fos expression in GABAergic neurons of VLPO was increased after administration of T1-11 in Gad2-Cre::Ai14 mice, suggesting the activation of GABAergic neurons in the VLPO. T1-11 exhibited no effects on heart rate and the low frequency/high frequency ratio of heart rate variability. We concluded that T1-11 elicited somnogenic effects and effectively ameliorated acute stress-induced insomnia. The somnogenic effect is mediated by A2ARs to activate GABAergic neurons in the VLPO. This adenosine analogue could be a potential hypnotic because of no sympathetic and parasympathetic effects on the cardiovascular system.
Collapse
Affiliation(s)
- Shuo-Bin Jou
- Department of Neurology, Mackay Medical College, Mackay Memorial Hospital, New Taipei City, Taiwan
| | - Chung-Jen Tsai
- Department of Veterinary Medicine, School of Veterinary Medicine, National Taiwan University, Taipei, Taiwan
| | - Chun-Ying Fang
- Department of Veterinary Medicine, School of Veterinary Medicine, National Taiwan University, Taipei, Taiwan
| | - Pei-Lu Yi
- Department of Sport Management, College of Tourism, Leisure and Sports, Aletheia University, New Taipei City, Taiwan
| | - Fang-Chia Chang
- Department of Veterinary Medicine, School of Veterinary Medicine, National Taiwan University, Taipei, Taiwan.,Graduate Institute of Brain and Mind Sciences, College of Medicine, National Taiwan University, Taipei, Taiwan.,Graduate Institute of Acupuncture Science, College of Chinese Medicine, China Medical University, Taichung, Taiwan
| |
Collapse
|
15
|
T1-11, an adenosine derivative, ameliorates aging-related behavioral physiology and senescence markers in aging mice. Aging (Albany NY) 2020; 12:10556-10577. [PMID: 32501291 PMCID: PMC7346012 DOI: 10.18632/aging.103279] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2019] [Accepted: 04/20/2020] [Indexed: 12/25/2022]
Abstract
Aging is a natural human process. It is uniquely individual, taking into account experiences, lifestyle habits and environmental factors. However, many disorders and syndromes, such as osteoporosis, neurodegenerative disorders, cognitive decline etc., often come with aging. The present study was designed to investigate the possible anti-aging effect of N6-(4-hydroxybenzyl)adenine riboside (T1-11), an adenosine analog isolated from Gastrodia elata, in a mouse model of aging created by D-galactose (D-gal) and the underlying mechanism, as well as explore the role of adenosine signaling in aging. T1-11 activated A2AR and suppressed D-gal- and BeSO4-induced cellular senescence in vitro. In vivo results in mice revealed that T1-11 abated D-gal-induced reactive oxygen species generation and ameliorated cognitive decline by inducing neurogenesis and lowering D-gal-caused neuron death. T1-11 could be a potent agent for postponing senility and preventing aging-related neuroinflammation and neurodegeneration.
Collapse
|
16
|
Choudhury H, Chellappan DK, Sengupta P, Pandey M, Gorain B. Adenosine Receptors in Modulation of Central Nervous System Disorders. Curr Pharm Des 2020; 25:2808-2827. [PMID: 31309883 DOI: 10.2174/1381612825666190712181955] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2019] [Accepted: 07/01/2019] [Indexed: 12/20/2022]
Abstract
The ubiquitous signaling nucleoside molecule, adenosine is found in different cells of the human body to provide its numerous pharmacological role. The associated actions of endogenous adenosine are largely dependent on conformational change of the widely expressed heterodimeric G-protein-coupled A1, A2A, A2B, and A3 adenosine receptors (ARs). These receptors are well conserved on the surface of specific cells, where potent neuromodulatory properties of this bioactive molecule reflected by its easy passage through the rigid blood-brainbarrier, to simultaneously act on the central nervous system (CNS). The minimal concentration of adenosine in body fluids (30-300 nM) is adequate to exert its neuromodulatory action in the CNS, whereas the modulatory effect of adenosine on ARs is the consequence of several neurodegenerative diseases. Modulatory action concerning the activation of such receptors in the CNS could be facilitated towards neuroprotective action against such CNS disorders. Our aim herein is to discuss briefly pathophysiological roles of adenosine on ARs in the modulation of different CNS disorders, which could be focused towards the identification of potential drug targets in recovering accompanying CNS disorders. Researches with active components with AR modulatory action have been extended and already reached to the bedside of the patients through clinical research in the improvement of CNS disorders. Therefore, this review consist of recent findings in literatures concerning the impact of ARs on diverse CNS disease pathways with the possible relevance to neurodegeneration.
Collapse
Affiliation(s)
- Hira Choudhury
- School of Pharmacy, International Medical University, Bukit Jalil, Kuala Lumpur, Malaysia
| | - Dinesh K Chellappan
- School of Pharmacy, International Medical University, Bukit Jalil, Kuala Lumpur, Malaysia
| | - Pallav Sengupta
- Department of Physiology, Faculty of Medicine, MA`HSA University, Kuala Lumpur, Malaysia
| | - Manisha Pandey
- School of Pharmacy, International Medical University, Bukit Jalil, Kuala Lumpur, Malaysia
| | - Bapi Gorain
- School of Pharmacy, Faculty of Health and Medical Science, Taylor's University, Subang Jaya, Selangor, Malaysia
| |
Collapse
|
17
|
Kadlecová A, Maková B, Artal-Sanz M, Strnad M, Voller J. The plant hormone kinetin in disease therapy and healthy aging. Ageing Res Rev 2019; 55:100958. [PMID: 31479763 DOI: 10.1016/j.arr.2019.100958] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2019] [Revised: 08/02/2019] [Accepted: 08/30/2019] [Indexed: 12/20/2022]
Abstract
It has been more than 60 years since the discovery of kinetin, the first known member of a group of plant hormones called cytokinins. In this review we summarize the health-promoting activity of kinetin in animal systems, ranging from cells cultured in vitro through invertebrates to mammals. Kinetin has been shown to modulate aging, to delay age-related physiological decline and to protect against some neurodegenerative diseases. We also review studies on its mechanism of action, as well as point out gaps in our current knowledge.
Collapse
Affiliation(s)
- Alena Kadlecová
- Laboratory of Growth Regulators, The Czech Academy of Sciences, Institute of Experimental Botany & Palacký University, Šlechtitelů 27, CZ-78371 Olomouc, Czech Republic
| | - Barbara Maková
- Laboratory of Growth Regulators, The Czech Academy of Sciences, Institute of Experimental Botany & Palacký University, Šlechtitelů 27, CZ-78371 Olomouc, Czech Republic
| | - Marta Artal-Sanz
- Andalusian Centre for Developmental Biology, CISIC-JA-University Pablo de Olavide, Department of Molecular Biology and Biochemical Engineering, Carretera de Utrera km 1, 41013 Sevilla, Spain
| | - Miroslav Strnad
- Laboratory of Growth Regulators, The Czech Academy of Sciences, Institute of Experimental Botany & Palacký University, Šlechtitelů 27, CZ-78371 Olomouc, Czech Republic
| | - Jiří Voller
- Laboratory of Growth Regulators, The Czech Academy of Sciences, Institute of Experimental Botany & Palacký University, Šlechtitelů 27, CZ-78371 Olomouc, Czech Republic; Department of Clinical and Molecular Pathology, Institute of Molecular and Translational Medicine, Faculty of Medicine and Dentistry, Palacký University, Hněvotínská 3, 775 15 Olomouc, Czech Republic.
| |
Collapse
|
18
|
Adenosine A2A receptor as potential therapeutic target in neuropsychiatric disorders. Pharmacol Res 2019; 147:104338. [DOI: 10.1016/j.phrs.2019.104338] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/01/2019] [Revised: 06/26/2019] [Accepted: 07/01/2019] [Indexed: 01/20/2023]
|
19
|
Adenosine and Adenosine A 2AReceptors as Targets for the Treatment of Niemann Pick Type C Disease. J Caffeine Adenosine Res 2019. [DOI: 10.1089/caff.2019.0014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
|
20
|
Zhang J, He Y, Jiang X, Jiang H, Shen J. Nature brings new avenues to the therapy of central nervous system diseases—An overview of possible treatments derived from natural products. SCIENCE CHINA-LIFE SCIENCES 2019; 62:1332-1367. [DOI: 10.1007/s11427-019-9587-y] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/24/2019] [Accepted: 07/01/2019] [Indexed: 12/11/2022]
|
21
|
Hong SI, Peyton L, Chern Y, Choi DS. Novel Adenosine Analog, N6-(4-Hydroxybenzyl)-Adenosine, Dampens Alcohol Drinking and Seeking Behaviors. J Pharmacol Exp Ther 2019; 371:260-267. [PMID: 31409667 DOI: 10.1124/jpet.119.261529] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2019] [Accepted: 08/09/2019] [Indexed: 12/11/2022] Open
Abstract
Adenosine signaling is associated with ethanol-related behaviors. We previously found that adenosine A2A receptor (A2AR) activation dampens ethanol drinking behaviors in equilibrative nucleoside transporter 1 (ENT1) knockout mice, and A2AR inhibition augments reward-seeking behavior in wild-type mice. The novel adenosine analog N6-(4-hydroxybenzyl)-adenosine (NHBA), which is isolated from the rhizomes of Gastrodia elata, activates A2AR and inhibits ENT1. Here, we examined the effects of NHBA on ethanol drinking in the two-bottle choice test and operant ethanol seeking behaviors. We selected mice exhibiting high ethanol drinking behavior in the two-bottle choice test. NHBA (0.1 mg/kg, i.p.) reduced ethanol drinking behavior in a limited-access 3-hour drinking session in high-consumption ethanol drinking mice, and NHBA (0.1 mg/kg, i.p.) did not alter locomotor activity in the open-field test. Operant conditioning with 10% ethanol and 10% sucrose (10E10S) reward increased zone entries and time spent in the ethanol zone, while NHBA (0.1 mg/kg, i.p.) dampened ethanol zone preference in the Y-maze. Furthermore, NHBA (0.1 mg/kg, i.p.) devalued 10E10S and 10% ethanol (10E) reward after operant conditioning with 10E10S and 10E. Taken together, NHBA through A2AR activation and ENT1 modulation may dampen ethanol drinking and seeking behaviors, suggesting that NHBA is a potential therapeutic agent for treating alcohol use disorder. SIGNIFICANCE STATEMENT: Our work highlights that A2AR activation and ENT1 inhibition by a novel adenosine analog isolated from Gastrodia elata, N6-(4-hydroxybenzyl)-adenosine, decreases ethanol drinking and seeking behaviors. We suggest that NHBA is a potential therapeutic agent to treat alcohol use disorder.
Collapse
Affiliation(s)
- Sa-Ik Hong
- Department of Molecular Pharmacology and Experimental Therapeutics (S.-I.H., L.P., D.-S.C.), Neuroscience Program (D.-S.C.), and Department of Psychiatry and Psychology (D.-S.C.), Mayo Clinic College of Medicine, Rochester, Minnesota; and Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan (Y.C.)
| | - Lee Peyton
- Department of Molecular Pharmacology and Experimental Therapeutics (S.-I.H., L.P., D.-S.C.), Neuroscience Program (D.-S.C.), and Department of Psychiatry and Psychology (D.-S.C.), Mayo Clinic College of Medicine, Rochester, Minnesota; and Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan (Y.C.)
| | - Yijuang Chern
- Department of Molecular Pharmacology and Experimental Therapeutics (S.-I.H., L.P., D.-S.C.), Neuroscience Program (D.-S.C.), and Department of Psychiatry and Psychology (D.-S.C.), Mayo Clinic College of Medicine, Rochester, Minnesota; and Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan (Y.C.)
| | - Doo-Sup Choi
- Department of Molecular Pharmacology and Experimental Therapeutics (S.-I.H., L.P., D.-S.C.), Neuroscience Program (D.-S.C.), and Department of Psychiatry and Psychology (D.-S.C.), Mayo Clinic College of Medicine, Rochester, Minnesota; and Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan (Y.C.)
| |
Collapse
|
22
|
Huang NK, Lin CC, Lin YL, Huang CL, Chiou CT, Lee YC, Lee SY, Huang HT, Yang YC. Morphological control of mitochondria as the novel mechanism of Gastrodia elata in attenuating mutant huntingtin-induced protein aggregations. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2019; 59:152756. [PMID: 31004885 DOI: 10.1016/j.phymed.2018.11.016] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/04/2018] [Accepted: 11/13/2018] [Indexed: 06/09/2023]
Abstract
BACKGROUND According to Compendium of Materia Medica, Gastrodia elata (GE) Blume as a top grade and frequently prescribed herbal medicine has been used in treating dizziness, headaches, and epilepsy, indicating a neuroprotective effect. Because GE is capable of suppressing a hyperactive liver and thus calming endogenous wind, and because Huntington's disease (HD) can be classified as a phenomenon of disturbed liver wind, it is suggested that GE might be beneficial in treating HD. However, although current studies support GE for the prevention of diverse neurodegenerations such as HD, its detailed mechanisms remain elusive. PURPOSE To investigate the molecular mechanism of GE in preventing HD by focusing on mitochondrial morphology, which is highly associated with HD etiology and thus proposed as a therapeutic target of neurodegenerations. STUDY DESIGN/METHODS The overexpression of the mutant huntingtin (mHTT) gene in rat pheochromocytoma (PC12) cells was used as an in vitro cell model of HD. A filter retardation assay was applied to measure protein aggregations during HTT expression. Cotransfection with mitochondrial fusion and fission genes was used to test their relationships with HTT aggregates by monitoring with a confocal laser scanning microscope and filter retardation assay. Western blot analysis was used to estimate protein expression under different drug treatments or cotransfections with other related genes. RESULTS The overexpression of mutant but not normal HTT genes significantly resulted in protein aggregations in PC12 cells. GE dose-dependently attenuated mHTT-induced protein aggregations and free radical formations. GE significantly reversed mHTT-induced mitochondrial fragmentation and dysregulation of mitochondrial fusion and fission molecules. The overexpression of mitochondrial fusion genes attenuated mHTT-induced protein aggregations. Further, Mdivi-1, a DRP1 fission molecule inhibitor, significantly reversed mHTT-induced protein aggregations and mitochondrial fragmentation. CONCLUSION GE attenuated mHTT aggregations through the control of mitochondrial fusion and the fission pathway.
Collapse
Affiliation(s)
- Nai-Kuei Huang
- National Research Institute of Chinese Medicine, Ministry of Health and Welfare, Taipei, Taiwan, ROC; Program for Neural Regenerative Medicine, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan, ROC
| | - Chung-Chih Lin
- Brain Research Center, National Yang-Ming University, Taipei, Taiwan, ROC; Department of Life Sciences and Institute of Genome Sciences, National Yang-Ming University, Taipei, Taiwan, ROC; Biophotonics Interdisciplinary Research Center, National Yang-Ming University, Taipei, Taiwan, ROC
| | - Yun-Lian Lin
- School of Chinese Pharmaceutical Sciences and Chinese Medicine Resources, College of Biopharmaceutical and Food Sciences, China Medical University, Taichung, Taiwan, ROC
| | - Chuen-Lin Huang
- Medical Research Center, Cardinal Tien Hospital, Hsintien, New Taipei City, Taiwan, ROC; Graduate Institute of Physiology & Department of Physiology and Biophysics, National Defense Medical Center, Taipei, Taiwan, ROC
| | - Chun-Tang Chiou
- National Research Institute of Chinese Medicine, Ministry of Health and Welfare, Taipei, Taiwan, ROC
| | - Yi-Chao Lee
- Program for Neural Regenerative Medicine, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan, ROC
| | - Shu-Yi Lee
- Department of Life Sciences and Institute of Genome Sciences, National Yang-Ming University, Taipei, Taiwan, ROC
| | - Hung-Tse Huang
- National Research Institute of Chinese Medicine, Ministry of Health and Welfare, Taipei, Taiwan, ROC
| | - Ying-Chen Yang
- Department of Biotechnology and Animal Science, National Ilan University, Ilan, Taiwan, ROC.
| |
Collapse
|
23
|
Chen IC, Chang CN, Chen WL, Lin TH, Chao CY, Lin CH, Lin HY, Cheng ML, Chiang MC, Lin JY, Wu YR, Lee-Chen GJ, Chen CM. Targeting Ubiquitin Proteasome Pathway with Traditional Chinese Medicine for Treatment of Spinocerebellar Ataxia Type 3. THE AMERICAN JOURNAL OF CHINESE MEDICINE 2019; 47:63-95. [PMID: 30612452 DOI: 10.1142/s0192415x19500046] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Nine autosomal dominant spinocerebellar ataxias (SCAs) are caused by an abnormal expansion of CAG trinucleotide repeats that encodes a polyglutamine (polyQ) tract within different genes. Accumulation of aggregated mutant proteins is a common feature of polyQ diseases, leading to progressive neuronal dysfunction and degeneration. SCA type 3 (SCA3), the most common form of SCA worldwide, is characterized by a CAG triplet expansion in chromosome 14q32.1 ATXN3 gene. As accumulation of the mutated polyQ protein is a possible initial event in the pathogenic cascade, clearance of aggregated protein by ubiquitin proteasome system (UPS) has been proposed to inhibit downstream detrimental events and suppress neuronal cell death. In this study, Chinese herbal medicine (CHM) extracts were studied for their proteasome-activating, polyQ aggregation-inhibitory and neuroprotective effects in GFPu and ATXN3/Q 75 -GFP 293/SH-SY5Y cells. Among the 14 tested extracts, 8 displayed increased proteasome activity, which was confirmed by 20S proteasome activity assay and analysis of ubiquitinated and fused GFP proteins in GFPu cells. All the eight extracts displayed good aggregation-inhibitory potential when tested in ATXN3/Q 75 -GFP 293 cells. Among them, neuroprotective effects of five selected extracts were shown by analyses of polyQ aggregation, neurite outgrowth, caspase 3 and proteasome activities, and ATXN3-GFP, ubiquitin, BCL2 and BAX protein levels in neuronal differentiated ATXN3/Q 75 -GFP SH-SY5Y cells. Finally, enhanced proteasome function, anti-oxidative activity and neuroprotection of catalpol, puerarin and daidzein (active constituents of Rehmannia glutinosa and Pueraria lobata) were demonstrated in GFPu and/or ATXN3/Q 75 -GFP 293/SH-SY5Y cells. This study may have therapeutic implication in polyQ-mediated disorders.
Collapse
Affiliation(s)
- I-Cheng Chen
- * Department of Neurology, Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Taoyuan 33302, Taiwan
| | - Chia-Ning Chang
- † Department of Life Science, National Taiwan Normal University, Taipei 11677, Taiwan
| | - Wan-Ling Chen
- * Department of Neurology, Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Taoyuan 33302, Taiwan
| | - Te-Hsien Lin
- † Department of Life Science, National Taiwan Normal University, Taipei 11677, Taiwan
| | - Chih-Ying Chao
- * Department of Neurology, Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Taoyuan 33302, Taiwan
| | - Chih-Hsin Lin
- * Department of Neurology, Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Taoyuan 33302, Taiwan
| | - Hsuan-Yuan Lin
- † Department of Life Science, National Taiwan Normal University, Taipei 11677, Taiwan
| | - Mei-Ling Cheng
- ‡ Department of Biomedical Sciences, College of Medicine, Chang Gung University, TaoYuan 33302, Taiwan
| | | | - Jung-Yaw Lin
- † Department of Life Science, National Taiwan Normal University, Taipei 11677, Taiwan
| | - Yih-Ru Wu
- * Department of Neurology, Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Taoyuan 33302, Taiwan
| | - Guey-Jen Lee-Chen
- † Department of Life Science, National Taiwan Normal University, Taipei 11677, Taiwan
| | - Chiung-Mei Chen
- * Department of Neurology, Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Taoyuan 33302, Taiwan
| |
Collapse
|
24
|
Duarte AI, Sjögren M, Santos MS, Oliveira CR, Moreira PI, Björkqvist M. Dual Therapy with Liraglutide and Ghrelin Promotes Brain and Peripheral Energy Metabolism in the R6/2 Mouse Model of Huntington's Disease. Sci Rep 2018; 8:8961. [PMID: 29895889 PMCID: PMC5997749 DOI: 10.1038/s41598-018-27121-w] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2018] [Accepted: 05/29/2018] [Indexed: 01/14/2023] Open
Abstract
Neuronal loss alongside altered energy metabolism, are key features of Huntington’s disease (HD) pathology. The orexigenic gut-peptide hormone ghrelin is known to stimulate appetite and affect whole body energy metabolism. Liraglutide is an efficient anti-type 2 diabetes incretin drug, with neuroprotective effects alongside anorectic properties. Combining liraglutide with the orexigenic peptide ghrelin may potentially promote brain/cognitive function in HD. The R6/2 mouse model of HD exhibits progressive central pathology, weight loss, deranged glucose metabolism, skeletal muscle atrophy and altered body composition. In this study, we targeted energy metabolism in R6/2 mice using a co-administration of liraglutide and ghrelin. We investigated their effect on brain cortical hormone-mediated intracellular signalling pathways, metabolic and apoptotic markers, and the impact on motor function in HD. We here demonstrate that liraglutide, alone or together with ghrelin (subcutaneous daily injections of 150 µg/kg (ghrelin) and 0.2 mg/kg (liraglutide), for 2 weeks), normalized glucose homeostatic features in the R6/2 mouse, without substantially affecting body weight or body composition. Liraglutide alone decreased brain cortical active GLP-1 and IGF-1 levels in R6/2 mice, alongside higher ADP levels. Liraglutide plus ghrelin decreased brain insulin, lactate, AMP and cholesterol levels in R6/2 mice. Taken together, our findings encourage further studies targeting energy metabolism in HD.
Collapse
Affiliation(s)
- Ana I Duarte
- CNC - Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal. .,Institute for Interdisciplinary Research (IIIUC), University of Coimbra, Coimbra, Portugal. .,Brain Disease Biomarker Unit, Department of Experimental Medical Sciences, Wallenberg Neuroscience Center, Lund University, Lund, Sweden.
| | - Marie Sjögren
- Brain Disease Biomarker Unit, Department of Experimental Medical Sciences, Wallenberg Neuroscience Center, Lund University, Lund, Sweden
| | - Maria S Santos
- CNC - Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal.,Life Sciences Department, Faculty of Sciences and Technology, University of Coimbra, Coimbra, Portugal
| | - Catarina R Oliveira
- CNC - Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal.,Laboratory of Biochemistry, Faculty of Medicine, University of Coimbra, Coimbra, Portugal
| | - Paula I Moreira
- CNC - Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal.,Laboratory of Physiology, Faculty of Medicine, University of Coimbra, Coimbra, Portugal
| | - Maria Björkqvist
- Brain Disease Biomarker Unit, Department of Experimental Medical Sciences, Wallenberg Neuroscience Center, Lund University, Lund, Sweden
| |
Collapse
|
25
|
Blum D, Chern Y, Domenici MR, Buée L, Lin CY, Rea W, Ferré S, Popoli P. The Role of Adenosine Tone and Adenosine Receptors in Huntington's Disease. J Caffeine Adenosine Res 2018; 8:43-58. [PMID: 30023989 PMCID: PMC6049521 DOI: 10.1089/caff.2018.0006] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Huntington's disease (HD) is a hereditary neurodegenerative disorder caused by a mutation in the IT15 gene that encodes for the huntingtin protein. Mutated hungtingtin, although widely expressed in the brain, predominantly affects striato-pallidal neurons, particularly enriched with adenosine A2A receptors (A2AR), suggesting a possible involvement of adenosine and A2AR is the pathogenesis of HD. In fact, polymorphic variation in the ADORA2A gene influences the age at onset in HD, and A2AR dynamics is altered by mutated huntingtin. Basal levels of adenosine and adenosine receptors are involved in many processes critical for neuronal function and homeostasis, including modulation of synaptic activity and excitotoxicity, the control of neurotrophin levels and functions, and the regulation of protein degradation mechanisms. In the present review, we critically analyze the current literature involving the effect of altered adenosine tone and adenosine receptors in HD and discuss why therapeutics that modulate the adenosine system may represent a novel approach for the treatment of HD.
Collapse
Affiliation(s)
- David Blum
- University of Lille, Inserm, CHU Lille, UMR-S 1172 - JPArc, LabEx DISTALZ, Lille, France
| | - Yijuang Chern
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
| | - Maria Rosaria Domenici
- National Center for Drug Research and Evaluation, Istituto Superiore di Sanità, Rome, Italy
| | - Luc Buée
- University of Lille, Inserm, CHU Lille, UMR-S 1172 - JPArc, LabEx DISTALZ, Lille, France
| | - Chien-Yu Lin
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
| | - William Rea
- Integrative Neurobiology Section, National Institute on Drug Abuse, National Institutes of Health, Baltimore, Maryland
| | - Sergi Ferré
- Integrative Neurobiology Section, National Institute on Drug Abuse, National Institutes of Health, Baltimore, Maryland
| | - Patrizia Popoli
- National Center for Drug Research and Evaluation, Istituto Superiore di Sanità, Rome, Italy
| |
Collapse
|
26
|
Neuronal adenosine A 2A receptor overexpression is neuroprotective towards 3-nitropropionic acid-induced striatal toxicity: a rat model of Huntington's disease. Purinergic Signal 2018; 14:235-243. [PMID: 29770921 DOI: 10.1007/s11302-018-9609-4] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2017] [Accepted: 05/01/2018] [Indexed: 10/16/2022] Open
Abstract
The A2A adenosine receptor (A2AR) is widely distributed on different cellular types in the brain, where it exerts a broad spectrum of pathophysiological functions, and for which a role in different neurodegenerative diseases has been hypothesized or demonstrated. To investigate the role of neuronal A2ARs in neurodegeneration, we evaluated in vitro and in vivo the effect of the neurotoxin 3-nitropropionic acid (3-NP) in a transgenic rat strain overexpressing A2ARs under the control of the neural-specific enolase promoter (NSEA2A rats). We recorded extracellular field potentials (FP) in corticostriatal slice and found that the synaptotoxic effect of 3-NP was significantly reduced in NSEA2A rats compared with wild-type animals (WT). In addition, after exposing corticostriatal slices to 3-NP 10 mM for 2 h, we found that striatal cell viability was significantly higher in NSEA2A rats compared to control rats. These in vitro results were confirmed by in vivo experiments: daily treatment of female rats with 3-NP 10 mg/kg for 8 days induced a selective bilateral lesion in the striatum, which was significantly reduced in NSEA2A compared to WT rats. These results demonstrate that the overexpression of the A2AR selectively at the neuronal level reduced 3-NP-induced neurodegeneration, and suggest an important function of the neuronal A2AR in the modulation of neurodegeneration.
Collapse
|
27
|
Lee CC, Chang CP, Lin CJ, Lai HL, Kao YH, Cheng SJ, Chen HM, Liao YP, Faivre E, Buée L, Blum D, Fang JM, Chern Y. Adenosine Augmentation Evoked by an ENT1 Inhibitor Improves Memory Impairment and Neuronal Plasticity in the APP/PS1 Mouse Model of Alzheimer's Disease. Mol Neurobiol 2018; 55:8936-8952. [PMID: 29616397 DOI: 10.1007/s12035-018-1030-z] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2018] [Accepted: 03/20/2018] [Indexed: 02/07/2023]
Abstract
Alzheimer's disease (AD) is a neurodegenerative disorder characterized by cognitive impairment and synaptic dysfunction. Adenosine is an important homeostatic modulator that controls the bioenergetic network in the brain through regulating receptor-evoked signaling pathways, bioenergetic machineries, and epigenetic-mediated gene regulation. Equilibrative nucleoside transporter 1 (ENT1) is a major adenosine transporter that recycles adenosine from the extracellular space. In the present study, we report that a small adenosine analogue (designated J4) that inhibited ENT1 prevented the decline in spatial memory in an AD mouse model (APP/PS1). Electrophysiological and biochemical analyses further demonstrated that chronic treatment with J4 normalized the impaired basal synaptic transmission and long-term potentiation (LTP) at Schaffer collateral synapses as well as the aberrant expression of synaptic proteins (e.g., NR2A and NR2B), abnormal neuronal plasticity-related signaling pathways (e.g., PKA and GSK3β), and detrimental elevation in astrocytic A2AR expression in the hippocampus and cortex of APP/PS1 mice. In conclusion, our findings suggest that modulation of adenosine homeostasis by J4 is beneficial in a mouse model of AD. Our study provides a potential therapeutic strategy to delay the progression of AD.
Collapse
Affiliation(s)
- Chia-Chia Lee
- Institute of Biomedical Sciences, Academia Sinica, Nankang, Taipei, 115, Taiwan
| | - Ching-Pang Chang
- Institute of Biomedical Sciences, Academia Sinica, Nankang, Taipei, 115, Taiwan
| | - Chun-Jung Lin
- School of Pharmacy, National Taiwan University, Taipei, Taiwan
| | - Hsing-Lin Lai
- Institute of Biomedical Sciences, Academia Sinica, Nankang, Taipei, 115, Taiwan
| | - Yu-Han Kao
- School of Pharmacy, National Taiwan University, Taipei, Taiwan
| | - Sin-Jhong Cheng
- Institute of Biomedical Sciences, Academia Sinica, Nankang, Taipei, 115, Taiwan.,Neuroscience Program of Academia Sinica, Academia Sinica, Taipei, Taiwan
| | - Hui-Mei Chen
- Institute of Biomedical Sciences, Academia Sinica, Nankang, Taipei, 115, Taiwan
| | - Yu-Ping Liao
- Institute of Biomedical Sciences, Academia Sinica, Nankang, Taipei, 115, Taiwan
| | - Emilie Faivre
- Université de Lille, Inserm, CHU-Lille, LabEx DISTALZ, Jean-Pierre Aubert research centre UMR-S1172, Lille, France
| | - Luc Buée
- Université de Lille, Inserm, CHU-Lille, LabEx DISTALZ, Jean-Pierre Aubert research centre UMR-S1172, Lille, France
| | - David Blum
- Université de Lille, Inserm, CHU-Lille, LabEx DISTALZ, Jean-Pierre Aubert research centre UMR-S1172, Lille, France
| | - Jim-Min Fang
- Department of Chemistry, National Taiwan University, Taipei, Taiwan
| | - Yijuang Chern
- Institute of Biomedical Sciences, Academia Sinica, Nankang, Taipei, 115, Taiwan.
| |
Collapse
|
28
|
Moreno E, Chiarlone A, Medrano M, Puigdellívol M, Bibic L, Howell LA, Resel E, Puente N, Casarejos MJ, Perucho J, Botta J, Suelves N, Ciruela F, Ginés S, Galve-Roperh I, Casadó V, Grandes P, Lutz B, Monory K, Canela EI, Lluís C, McCormick PJ, Guzmán M. Singular Location and Signaling Profile of Adenosine A 2A-Cannabinoid CB 1 Receptor Heteromers in the Dorsal Striatum. Neuropsychopharmacology 2018; 43:964-977. [PMID: 28102227 PMCID: PMC5854787 DOI: 10.1038/npp.2017.12] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/04/2016] [Revised: 01/13/2017] [Accepted: 01/14/2017] [Indexed: 12/16/2022]
Abstract
The dorsal striatum is a key node for many neurobiological processes such as motor activity, cognitive functions, and affective processes. The proper functioning of striatal neurons relies critically on metabotropic receptors. Specifically, the main adenosine and endocannabinoid receptors present in the striatum, ie, adenosine A2A receptor (A2AR) and cannabinoid CB1 receptor (CB1R), are of pivotal importance in the control of neuronal excitability. Facilitatory and inhibitory functional interactions between striatal A2AR and CB1R have been reported, and evidence supports that this cross-talk may rely, at least in part, on the formation of A2AR-CB1R heteromeric complexes. However, the specific location and properties of these heteromers have remained largely unknown. Here, by using techniques that allowed a precise visualization of the heteromers in situ in combination with sophisticated genetically modified animal models, together with biochemical and pharmacological approaches, we provide a high-resolution expression map and a detailed functional characterization of A2AR-CB1R heteromers in the dorsal striatum. Specifically, our data unveil that the A2AR-CB1R heteromer (i) is essentially absent from corticostriatal projections and striatonigral neurons, and, instead, is largely present in striatopallidal neurons, (ii) displays a striking G protein-coupled signaling profile, where co-stimulation of both receptors leads to strongly reduced downstream signaling, and (iii) undergoes an unprecedented dysfunction in Huntington's disease, an archetypal disease that affects striatal neurons. Altogether, our findings may open a new conceptual framework to understand the role of coordinated adenosine-endocannabinoid signaling in the indirect striatal pathway, which may be relevant in motor function and neurodegenerative diseases.
Collapse
Affiliation(s)
- Estefanía Moreno
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas, Instituto de Salud Carlos III, Madrid, Spain,Department of Biochemistry and Molecular Biology, University of Barcelona, Barcelona, Spain
| | - Anna Chiarlone
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas, Instituto de Salud Carlos III, Madrid, Spain,Instituto Universitario de Investigación Neuroquímica and Department of Biochemistry and Molecular Biology I, Complutense University, Madrid, Spain,Instituto Ramón y Cajal de Investigación Sanitaria, Madrid, Spain
| | - Mireia Medrano
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas, Instituto de Salud Carlos III, Madrid, Spain,Department of Biochemistry and Molecular Biology, University of Barcelona, Barcelona, Spain
| | - Mar Puigdellívol
- School of Pharmacy, University of East Anglia, Norwich Research Park, Norwich, UK
| | - Lucka Bibic
- School of Pharmacy, University of East Anglia, Norwich Research Park, Norwich, UK
| | - Lesley A Howell
- School of Pharmacy, University of East Anglia, Norwich Research Park, Norwich, UK,School of Biological and Chemical Sciences, Queen Mary, University of London, London, UK
| | - Eva Resel
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas, Instituto de Salud Carlos III, Madrid, Spain,Instituto Universitario de Investigación Neuroquímica and Department of Biochemistry and Molecular Biology I, Complutense University, Madrid, Spain,Instituto Ramón y Cajal de Investigación Sanitaria, Madrid, Spain
| | - Nagore Puente
- Department of Neurosciences, University of the Basque Country UPV/EHU, Leioa, Spain,Achucarro Basque Center for Neuroscience, Bizkaia Science and Technology Park, Zamudio, Spain
| | | | - Juan Perucho
- Instituto Ramón y Cajal de Investigación Sanitaria, Madrid, Spain
| | - Joaquín Botta
- School of Pharmacy, University of East Anglia, Norwich Research Park, Norwich, UK
| | - Nuria Suelves
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas, Instituto de Salud Carlos III, Madrid, Spain,Biomedical Science Department, School of Medicine; Institut d’Investigacions Biomèdiques August Pi i Sunyer, and Neuroscience Institute, Barcelona University, Barcelona, Spain
| | - Francisco Ciruela
- Pharmacology Unit, Department of Pathology and Experimental Therapeutics, IDIBELL, and Neuroscience Institute, Barcelona University, Barcelona, Spain
| | - Silvia Ginés
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas, Instituto de Salud Carlos III, Madrid, Spain,Biomedical Science Department, School of Medicine; Institut d’Investigacions Biomèdiques August Pi i Sunyer, and Neuroscience Institute, Barcelona University, Barcelona, Spain
| | - Ismael Galve-Roperh
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas, Instituto de Salud Carlos III, Madrid, Spain,Instituto Universitario de Investigación Neuroquímica and Department of Biochemistry and Molecular Biology I, Complutense University, Madrid, Spain,Instituto Ramón y Cajal de Investigación Sanitaria, Madrid, Spain
| | - Vicent Casadó
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas, Instituto de Salud Carlos III, Madrid, Spain,Department of Biochemistry and Molecular Biology, University of Barcelona, Barcelona, Spain
| | - Pedro Grandes
- Department of Neurosciences, University of the Basque Country UPV/EHU, Leioa, Spain,Achucarro Basque Center for Neuroscience, Bizkaia Science and Technology Park, Zamudio, Spain
| | - Beat Lutz
- Institute of Physiological Chemistry, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - Krisztina Monory
- Institute of Physiological Chemistry, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - Enric I Canela
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas, Instituto de Salud Carlos III, Madrid, Spain,Department of Biochemistry and Molecular Biology, University of Barcelona, Barcelona, Spain
| | - Carmen Lluís
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas, Instituto de Salud Carlos III, Madrid, Spain,Department of Biochemistry and Molecular Biology, University of Barcelona, Barcelona, Spain,Department of Biochemistry and Molecular Biology, University of Barcelona, Barcelona 08028, Spain, Tel: +34 93 4021208, Fax: +34 93 4021559, E-mail:
| | - Peter J McCormick
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas, Instituto de Salud Carlos III, Madrid, Spain,Department of Biochemistry and Molecular Biology, University of Barcelona, Barcelona, Spain,School of Pharmacy, University of East Anglia, Norwich Research Park, Norwich, UK,Faculty of Health and Medical Sciences, University of Surrey, Guildford, Surrey, UK,School of Veterinary Medicine Faculty of Health & Medical Sciences, University of Surrey, Daphne Jackson Road, Guildford, Surrey, GU2 7AL, UK, Tel: +44 (0)1483 684399, Fax: +44 (0)1483 684399, E-mail:
| | - Manuel Guzmán
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas, Instituto de Salud Carlos III, Madrid, Spain,Instituto Universitario de Investigación Neuroquímica and Department of Biochemistry and Molecular Biology I, Complutense University, Madrid, Spain,Instituto Ramón y Cajal de Investigación Sanitaria, Madrid, Spain,Instituto Universitario de Investigación Neuroquímica (IUIN) and Department of Biochemistry and Molecular Biology I, School of Biology, Complutense University, Madrid 28040, Spain, Tel: +34 91 3944668, Fax: +34 91 3944672, E-mail:
| |
Collapse
|
29
|
Lai CY, Liu YJ, Lai HL, Chen HM, Kuo HC, Liao YP, Chern Y. The D2 Dopamine Receptor Interferes With the Protective Effect of the A 2A Adenosine Receptor on TDP-43 Mislocalization in Experimental Models of Motor Neuron Degeneration. Front Neurosci 2018; 12:187. [PMID: 29615863 PMCID: PMC5869924 DOI: 10.3389/fnins.2018.00187] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2017] [Accepted: 03/07/2018] [Indexed: 12/13/2022] Open
Abstract
The A2A adenosine receptor (A2AR) and D2 dopamine receptor (D2R) are two G-protein-coupled receptors that can form dimers and negatively regulate their partners. TAR DNA-binding protein (TDP-43) is a nuclear protein that has been implicated in amyotrophic lateral sclerosis (ALS). Mislocalization of TDP-43 from the nucleus to the cytoplasm is an early step of TDP-43 proteinopathy. Our previous studies indicated that A2AR is a potential drug target for ALS because treatment with an A2AR agonist (JMF1907; a T1-11 analog) prevents reactive oxygen species (ROS)-induced TDP-43 mislocalization in a motor neuron cell line (NSC34) and delays motor impairment in a TDP-43 transgenic ALS mouse model. Here, we set out to assess whether activation of D2R interferes with the beneficial effects of an A2AR agonist on motor neurons. We first demonstrated that A2AR and D2R are both located in motor neurons of mouse and human spinal cords and human iPSC-derived motor neurons. Expression of A2AR and D2R in NSC34 cells led to dimer formation without affecting the binding affinity of A2AR toward T1-11. Importantly, activation of D2R reduced T1-11-mediated activation of cAMP/PKA signaling and subsequent inhibition of TDP-43 mislocalization in NSC34 cells. Treatment with quinpirole (a D2 agonist) blunted the rescuing effect of T1-11 on TDP-43 mislocalization and impaired grip strength in a mouse model of ALS. Our findings suggest that D2R activation may limit the beneficial responses of an A2AR agonist in motor neurons and may have an important role in ALS pathogenesis.
Collapse
Affiliation(s)
- Chia-You Lai
- Graduate Institute of Life Sciences, National Defense Medical Center, Taipei, Taiwan
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
| | - Yu-Ju Liu
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
| | - Hsing-Lin Lai
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
| | - Hui-Mei Chen
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
| | - Hung-Chi Kuo
- Institute of Cellular and Organismic Biology, Academia Sinica, Taipei, Taiwan
| | - Yu-Ping Liao
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
| | - Yijuang Chern
- Graduate Institute of Life Sciences, National Defense Medical Center, Taipei, Taiwan
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
| |
Collapse
|
30
|
Sustained reversal of central neuropathic pain induced by a single intrathecal injection of adenosine A 2A receptor agonists. Brain Behav Immun 2018; 69:470-479. [PMID: 29366930 DOI: 10.1016/j.bbi.2018.01.005] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/28/2016] [Revised: 01/09/2018] [Accepted: 01/11/2018] [Indexed: 01/19/2023] Open
Abstract
Central neuropathic pain is a debilitating outcome of spinal cord injury (SCI) and current treatments to alleviate this pain condition are ineffective. A growing body of literature suggests that activating adenosine A2A receptors (A2ARs) decreases the production of proinflammatory cytokines and increases the production of anti-inflammatory cytokines. Here, the effect of administering intrathecal A2AR agonists on central neuropathic pain was measured using hindpaw mechanical allodynia in a rat model of SCI termed spinal neuropathic avulsion pain (SNAP). Other models of SCI cause extensive damage to the spinal cord, resulting in paralysis and health problems. SNAP rats with unilateral low thoracic (T13)/high lumbar (L1) dorsal root avulsion develop below-level bilateral allodynia, without concomitant motor or health problems. A single intrathecal injection of the A2AR agonist 2-p-(2-carboxyethyl)phenethylamino-5'-N-ethylcarboxamido adenosine HCl (CGS21680) reversed SCI-induced allodynia for at least 6 weeks. The reversal is likely in part mediated by interleukin (IL)-10, as intrathecally administering neutralizing IL-10 antibodies 1 week after CGS21680 abolished the anti-allodynic effect of CGS21680. Dorsal spinal cord tissue from the ipsilateral site of SCI (T13/L1) was assayed 1 and 6 weeks after CGS21680 for IL-10, CD11b, and tumor necrosis factor (TNF) gene expression. CGS21680 treatment did not change IL-10 gene expression but did significantly decrease CD11b and TNF gene expression at both timepoints. A second A2AR agonist, 4-(3-(6-amino-9-(5-cyclopropylcarbamoyl-3,4-dihydroxytetrahydrofuran-2-yl)-9H-purin-2-yl)prop-2-ynyl)piperidine-1-carboxylic acid methyl ester (ATL313), was also able to significantly prevent and reverse SCI-induced allodynia for several weeks after a single intrathecal injection, providing converging lines of evidence of A2AR involvement. The enduring pain reversal after a single intrathecal injection of A2AR agonists suggests that A2AR agonists could be exciting new candidates for treating SCI-induced central neuropathic pain.
Collapse
|
31
|
Huang CL, Wang KC, Yang YC, Chiou CT, Tan CH, Lin YL, Huang NK. Gastrodia elata alleviates mutant huntingtin aggregation through mitochondrial function and biogenesis mediation. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2018; 39:75-84. [PMID: 29433686 DOI: 10.1016/j.phymed.2017.12.017] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/27/2017] [Revised: 11/01/2017] [Accepted: 12/17/2017] [Indexed: 06/08/2023]
Abstract
BACKGROUND According to the Compendium of Materia Medica, Gastrodia elata (GE) Blume is a top-grade herbal medicine frequently used to treat dizziness, headaches, tetanus, and epilepsy, suggesting that it affects neurological functions. Although studies have supported its effects in preventing diverse neurodegenerations such as Huntington's disease (HD), its mechanisms require further investigation. PURPOSE To investigate the ability of the molecular mechanism of GE to prevent mutant huntingtin (mHTT) protein aggregation by focusing on mitochondrial function and biogenesis, which have been proposed as the therapeutic targets of HD. STUDY DESIGN/METHODS mHtt overexpression in pheochromocytoma (PC12) cells was used as an in vitro cell model of HD. A retardation assay was applied to measure protein aggregation during Htt expression. Cotransfection with transcriptional genes was used to test their relationships with HTT aggregates by monitoring with a confocal laser scanning microscope. Western blot analysis was used to estimate protein expression under different drug treatments or when cotransfected with other related genes. RESULTS Mutant, abnormal Htt overexpression resulted in significant protein aggregation in PC12 cells. GE dose-dependently attenuated mHTT aggregates and increased cyclic-AMP response element-binding protein (CREB) phosphorylation. Adenosine A2A-R receptor (A2A-R) antagonist counteracted these phenomena. CREB overexpression significantly attenuated mHTT aggregation. GE increased the promoter activity and expression of peroxisome proliferator-activated receptor gamma coactivator 1-alpha (PGC-1α). Furthermore, wild-type PGC-1α but not mutant PGC-1α overexpression attenuated mHTT aggregates. CONCLUSION GE attenuated mHtt aggregation by mediating mitochondrial function and biogenesis through the A2A-R/PKA/CREB/PGC-1α-dependent pathway.
Collapse
Affiliation(s)
- Chuen-Lin Huang
- Medical Research Center, Cardinal Tien Hospital, Hsintien, New Taipei City, Taiwan, ROC; Graduate Institute of Physiology & Department of Physiology and Biophysics, National Defense Medical Center, Taipei, Taiwan, ROC
| | - Kaw-Chen Wang
- Department of Neurology, Cardinal-Tien Hospital, New Taipei City, Taiwan, ROC
| | - Ying-Chen Yang
- Department of Biotechnology and Animal Science, National Ilan University, Ilan, Taiwan, ROC
| | - Chun-Tang Chiou
- National Research Institute of Chinese Medicine, Ministry of Health and Welfare, Taipei, Taiwan, ROC
| | - Chia-Hui Tan
- National Research Institute of Chinese Medicine, Ministry of Health and Welfare, Taipei, Taiwan, ROC
| | - Yun-Lian Lin
- School of Chinese Pharmaceutical Sciences and Chinese Medicine Resources, College of Biopharmaceutical and Food Sciences, China Medical University, Taichung, Taiwan, ROC
| | - Nai-Kuei Huang
- National Research Institute of Chinese Medicine, Ministry of Health and Welfare, Taipei, Taiwan, ROC; Ph.D. Program for Neural Regenerative Medicine, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan, ROC.
| |
Collapse
|
32
|
Chien T, Weng YT, Chang SY, Lai HL, Chiu FL, Kuo HC, Chuang DM, Chern Y. GSK3β negatively regulates TRAX, a scaffold protein implicated in mental disorders, for NHEJ-mediated DNA repair in neurons. Mol Psychiatry 2018; 23:2375-2390. [PMID: 29298990 PMCID: PMC6294740 DOI: 10.1038/s41380-017-0007-z] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/11/2017] [Revised: 10/28/2017] [Accepted: 10/30/2017] [Indexed: 12/27/2022]
Abstract
Translin-associated protein X (TRAX) is a scaffold protein with various functions and has been associated with mental illnesses, including schizophrenia. We have previously demonstrated that TRAX interacts with a Gsα protein-coupled receptor, the A2A adenosine receptor (A2AR), and mediates the function of this receptor in neuritogenesis. In addition, stimulation of the A2AR markedly ameliorates DNA damage evoked by elevated oxidative stress in neurons derived from induced pluripotent stem cells (iPSCs). Here, we report that glycogen synthase kinase 3 beta (GSK3β) and disrupted-in-schizophrenia 1 (DISC1) are two novel interacting proteins of TRAX. We present evidence to suggest that the stimulation of A2AR markedly facilitated DNA repair through the TRAX/DISC1/GSK3β complex in a rat neuronal cell line (PC12), primary mouse neurons, and human medium spiny neurons derived from iPSCs. A2AR stimulation led to the inhibition of GSK3β, thus dissociating the TRAX/DISC1/GSK3β complex and facilitating the non-homologous end-joining pathway (NHEJ) by enhancing the activation of a DNA-dependent protein kinase via phosphorylation at Thr2609. Similarly, pharmacological inhibition of GSK3β by SB216763 also facilitated the TRAX-mediated repair of oxidative DNA damage. Collectively, GSK3β binds with TRAX and negatively affects its ability to facilitate NHEJ repair. The suppression of GSK3β by A2AR activation or a GSK3β inhibitor releases TRAX for the repair of oxidative DNA damage. Our findings shed new light on the molecular mechanisms underlying diseases associated with DNA damage and provides a novel target (i.e., the TRAX/DISC1/GSK3β complex) for future therapeutic development for mental disorders.
Collapse
Affiliation(s)
- Ting Chien
- 0000 0004 0634 0356grid.260565.2Graduate Institute of Life Sciences, National Defense Medical Center, Taipei, Taiwan ,0000 0004 0633 7958grid.482251.8Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
| | - Yu-Ting Weng
- 0000 0004 0633 7958grid.482251.8Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan ,0000 0001 2287 1366grid.28665.3fProgram in Molecular Medicine, National Yang-Ming University and Academia Sinica, Taipei, Taiwan
| | - Shu-Yung Chang
- 0000 0004 0633 7958grid.482251.8Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan ,0000 0001 0425 5914grid.260770.4Institute of Neuroscience, National Yang-Ming University, Taipei, Taiwan
| | - Hsing-Lin Lai
- 0000 0004 0633 7958grid.482251.8Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
| | - Feng-Lan Chiu
- 0000 0001 2287 1366grid.28665.3fInstitute of Cellular and Organismic Biology, Academia Sinica, Taipei, Taiwan
| | - Hung-Chih Kuo
- 0000 0001 2287 1366grid.28665.3fInstitute of Cellular and Organismic Biology, Academia Sinica, Taipei, Taiwan
| | - De-Maw Chuang
- 0000 0004 0464 0574grid.416868.5Intramural Research Program, National Institute of Mental Health, National Institutes of Health, Bethesda, MD USA
| | - Yijuang Chern
- Graduate Institute of Life Sciences, National Defense Medical Center, Taipei, Taiwan. .,Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan. .,Institute of Neuroscience, National Yang-Ming University, Taipei, Taiwan. .,Program in Molecular Medicine, National Yang-Ming University and Academia Sinica, Taipei, Taiwan.
| |
Collapse
|
33
|
Sehgal SA, Hammad MA, Tahir RA, Akram HN, Ahmad F. Current Therapeutic Molecules and Targets in Neurodegenerative Diseases Based on in silico Drug Design. Curr Neuropharmacol 2018; 16:649-663. [PMID: 29542412 PMCID: PMC6080102 DOI: 10.2174/1570159x16666180315142137] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2017] [Revised: 01/01/2018] [Accepted: 03/02/2018] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND As the number of elderly persons increases, neurodegenerative diseases are becoming ubiquitous. There is currently a great need for knowledge concerning management of oldage neurodegenerative diseases; the most important of which are: Alzheimer's disease, Parkinson's disease, Amyotrophic Lateral Sclerosis, and Huntington's disease. OBJECTIVE To summarize the potential of computationally predicted molecules and targets against neurodegenerative diseases. METHOD Review of literature published since 1997 against neurodegenerative diseases, utilizing as keywords: in silico, Alzheimer's disease, Parkinson's disease, Amyotrophic Lateral Sclerosis ALS, and Huntington's disease was conducted. RESULTS AND CONCLUSION Due to the costs associated with experimentation and current ethical law, performing experiments directly on living organisms has become much more difficult. In this scenario, in silico techniques have been successful and have become powerful tools in the search to cure disease. Researchers use the Computer Aided Drug Design pipeline which: 1) generates 3- dimensional structures of target proteins through homology modeling 2) achieves stabilization through molecular dynamics simulation, and 3) exploits molecular docking through large compound libraries. Next generation sequencing is continually producing enormous amounts of raw sequence data while neuroimaging is producing a multitude of raw image data. To solve such pressing problems, these new tools and algorithms are required. This review elaborates precise in silico tools and techniques for drug targets, active molecules, and molecular docking studies, together with future prospects and challenges concerning possible breakthroughs in Alzheimer's, Parkinson's, Amyotrophic Lateral Sclerosis, and Huntington's disease.
Collapse
Affiliation(s)
- Sheikh Arslan Sehgal
- Address correspondence to this author at the State Key Laboratory of Biomembrane and Membrane Biotechnology, Institute of Zoology, Chinese Academy of Sciences; Beijing, China; E-mail:
| | | | | | | | | |
Collapse
|
34
|
Bortolatto CF, Reis AS, Pinz MP, Voss GT, Oliveira RL, Vogt AG, Roman S, Jesse CR, Luchese C, Wilhelm EA. Selective A 2A receptor antagonist SCH 58261 modulates striatal oxidative stress and alleviates toxicity induced by 3-Nitropropionic acid in male Wistar rats. Metab Brain Dis 2017; 32:1919-1927. [PMID: 28795281 DOI: 10.1007/s11011-017-0086-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/31/2017] [Accepted: 08/01/2017] [Indexed: 10/19/2022]
Abstract
The aim of the present study was to investigate the effects of SCH58261, a selective adenosine A2A receptor antagonist, on striatal toxicity induced by 3-nitropropionic acid (3-NP) in rats. The experimental protocol consisted of 10 administrations (once a day) of SCH58261 (0.01 or 0.05 mg/kg/day, intraperitoneal, i.p.). From 7th to 10th day, 3-NP (20 mg/kg/day, i.p.) was injected 1 h after SCH58261 administration. Twenty-four hours after the last 3-NP injection, the body weight gain, locomotor activity (open-field test), motor coordination (rotarod test), striatal succinate dehydrogenase (SDH) activity and parameters linked to striatal oxidative status were evaluated in rats. The marked body weight loss resulting from 3-NP injections in rats was partially protected by SCH 58261 at both doses. SCH 58261 at the highest dose was effective against impairments on motor coordination and locomotor activity induced by 3-NP. SCH 58261 was unable to restore the inhibition of SDH activity caused by 3-NP. In addition, the increase in striatal reactive species (RS) levels, depletion of reduced glutathione (GSH) content and stimulation of glutathione reductase (GR) activity provoked by 3-NP injections were alleviated by both doses of SCH 58261. The highest dose of SCH 58261 was also effective in attenuating the increase of protein carbonyl levels as well as the inhibition of glutathione peroxidase (GPx) activity in rats exposed to 3-NP. Our results revealed that reduction of oxidative stress in rat striatum by adenosine A2A receptor antagonism contributes for alleviating 3-NP-induced toxicity.
Collapse
Affiliation(s)
- Cristiani F Bortolatto
- Programa de Pós-graduação em Bioquímica e Bioprospecção (PPGBio), Centro de Ciências Químicas, Farmacêuticas e de Alimentos, Universidade Federal de Pelotas (UFPel), Campus Capão do Leão, Pelotas, RS, CEP 96010-900, Brazil.
| | - Angélica S Reis
- Programa de Pós-graduação em Bioquímica e Bioprospecção (PPGBio), Centro de Ciências Químicas, Farmacêuticas e de Alimentos, Universidade Federal de Pelotas (UFPel), Campus Capão do Leão, Pelotas, RS, CEP 96010-900, Brazil
| | - Mikaela P Pinz
- Programa de Pós-graduação em Bioquímica e Bioprospecção (PPGBio), Centro de Ciências Químicas, Farmacêuticas e de Alimentos, Universidade Federal de Pelotas (UFPel), Campus Capão do Leão, Pelotas, RS, CEP 96010-900, Brazil
| | - Guilherme T Voss
- Programa de Pós-graduação em Bioquímica e Bioprospecção (PPGBio), Centro de Ciências Químicas, Farmacêuticas e de Alimentos, Universidade Federal de Pelotas (UFPel), Campus Capão do Leão, Pelotas, RS, CEP 96010-900, Brazil
| | - Renata L Oliveira
- Programa de Pós-graduação em Bioquímica e Bioprospecção (PPGBio), Centro de Ciências Químicas, Farmacêuticas e de Alimentos, Universidade Federal de Pelotas (UFPel), Campus Capão do Leão, Pelotas, RS, CEP 96010-900, Brazil
| | - Ane G Vogt
- Programa de Pós-graduação em Bioquímica e Bioprospecção (PPGBio), Centro de Ciências Químicas, Farmacêuticas e de Alimentos, Universidade Federal de Pelotas (UFPel), Campus Capão do Leão, Pelotas, RS, CEP 96010-900, Brazil
| | - Silvane Roman
- Universidade Regional Integrada, Campus Erechim, Erechim, RS, CEP 99700-000, Brazil
| | - Cristiano R Jesse
- Laboratório de Avaliações Farmacológicas e Toxicológicas Aplicadas às Moléculas Bioativas - LaftamBio Pampa, Universidade Federal do Pampa, Itaqui, RS, CEP 97650-000, Brazil
| | - Cristiane Luchese
- Programa de Pós-graduação em Bioquímica e Bioprospecção (PPGBio), Centro de Ciências Químicas, Farmacêuticas e de Alimentos, Universidade Federal de Pelotas (UFPel), Campus Capão do Leão, Pelotas, RS, CEP 96010-900, Brazil
| | - Ethel A Wilhelm
- Programa de Pós-graduação em Bioquímica e Bioprospecção (PPGBio), Centro de Ciências Químicas, Farmacêuticas e de Alimentos, Universidade Federal de Pelotas (UFPel), Campus Capão do Leão, Pelotas, RS, CEP 96010-900, Brazil.
| |
Collapse
|
35
|
Ferrante A, Pezzola A, Matteucci A, Di Biase A, Attorri L, Armida M, Martire A, Chern Y, Popoli P. The adenosine A 2A receptor agonist T1-11 ameliorates neurovisceral symptoms and extends the lifespan of a mouse model of Niemann-Pick type C disease. Neurobiol Dis 2017; 110:1-11. [PMID: 29079454 DOI: 10.1016/j.nbd.2017.10.013] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2017] [Revised: 09/29/2017] [Accepted: 10/23/2017] [Indexed: 12/12/2022] Open
Abstract
Niemann-Pick C is a fatal neurovisceral disorder caused, in 95% of cases, by mutation of NPC1 gene. Therapeutic options are extremely limited and new "druggable" targets are highly warranted. We previously demonstrated that the stimulation of the adenosine A2A receptor (A2AR) normalized the pathological phenotype of cellular models of NPC1. Since the validation of A2ARs as a therapeutic target for NPC1 can be obtained only conducting studies in in vivo models of the disease, in the present paper, the effects of two agonists of A2ARs were evaluated in the mouse model Balb/c Npc1nih, hereafter indicated as NPC1-/-. The agonists CGS21680 (2.5 and 5mg/kg/day by intraperitoneal injection) and T1-11 (50mg/kg/day in drinking water) were administered at a presymptomatic stage of the disease of NPC1-/- mice (PN28 and PN30, respectively); the experimental groups were the following: vehicle-treated WT mice (N=16 for both CGS and T1-11 treatments); vehicle-treated NPC1-/- mice (N=14 for CGS and 12 for T1-11 treatment); CGS-treated NPC1-/- mice (N=7) and T1-11-treated NPC1-/- mice (N=11). The efficacy of the treatments was evaluated by comparing vehicle-treated and CGS or T1-11-treated NPC1-/- mice for their motor deficits (analyzed by both rotarod and footprint tests), hippocampal cognitive impairment (by Novel Object Recognition (NOR) test), cerebellar neurodegeneration (Purkinje neurons counting), and cholesterol and sphingomyelin accumulation in spleen and liver. Finally, the effect of both agonists on survival was evaluated by applying a humane late endpoint (weight loss >30% of peak weight, punched posture and reduced activity in the cage). The results demonstrated that, while CGS21680 only slightly attenuated cognitive deficits, T1-11 ameliorated motor coordination, significantly improved cognitive impairments, increased the survival of Purkinje neurons and reduced sphingomyelin accumulation in the liver. More importantly, it significantly prolonged the lifespan of NPC1-/- mice. In vitro experiments conducted in a neuronal model of NPC1 demonstrated that the ability of T1-11 to normalize cell phenotype was mediated by the selective activation of A2ARs and modulation of intracellular calcium levels. In conclusion, our results fully confirm the validity of A2ARs as a new target for NPC1 treatment. As soon as new ligands with improved pharmacokinetic characteristics (i.e. orally active, with brain bioavailability and metabolic stability) will be obtained, A2AR agonists could represent a breakthrough in the treatment of NPC.
Collapse
Affiliation(s)
- Antonella Ferrante
- National Center for Drug Research and Evaluation, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161 Rome, Italy.
| | - Antonella Pezzola
- National Center for Drug Research and Evaluation, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161 Rome, Italy
| | - Andrea Matteucci
- National Center for Drug Research and Evaluation, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161 Rome, Italy
| | - Antonella Di Biase
- Dept. Food Safety, Nutrition and Veterinary Public Health, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161 Rome, Italy
| | - Lucilla Attorri
- Dept. Food Safety, Nutrition and Veterinary Public Health, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161 Rome, Italy
| | - Monica Armida
- National Center for Drug Research and Evaluation, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161 Rome, Italy
| | - Alberto Martire
- National Center for Drug Research and Evaluation, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161 Rome, Italy
| | - Yijuang Chern
- Institute of Biomedical Sciences N333, Academia Sinica, Taipei 11529, Taiwan
| | - Patrizia Popoli
- National Center for Drug Research and Evaluation, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161 Rome, Italy
| |
Collapse
|
36
|
Kao YH, Lin MS, Chen CM, Wu YR, Chen HM, Lai HL, Chern Y, Lin CJ. Targeting ENT1 and adenosine tone for the treatment of Huntington's disease. Hum Mol Genet 2017; 26:467-478. [PMID: 28069792 DOI: 10.1093/hmg/ddw402] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2016] [Accepted: 11/21/2016] [Indexed: 11/14/2022] Open
Abstract
Huntington's disease (HD) is caused by an abnormal CAG expansion in the exon 1 of huntingtin gene. The treatment of HD is an unmet medical need. Given the important role of adenosine in modulating brain activity, in this study, levels of adenosine and adenine nucleotides in the cerebral spinal fluid of patients with HD and in the brain of two mouse models of HD (R6/2 and Hdh150Q) were analysed. The expression and activity of ENT1 in the striatum of mice with HD were measured. Targeting adenosine tone for treating HD was examined in R6/2 mice by genetic removal of ENT1 and by giving an ENT1 inhibitor, respectively. The results showed that the adenosine homeostasis is dysregulated in the brain of patients and mice with HD. In patients, the ratio of adenosine/ATP in the cerebral spinal fluid was negatively correlated with the disease duration, and tended to have a positive correlation with independence scale and functional capacity. In comparison to controls, mRNA level of ENT1 was higher in the striatum of R6/2 and Hdh150Q mice. Intrastriatal administration of ENT1 inhibitors increased extracellular level of adenosine in the striatum of R6/2 mice to a much higher level than controls. Chronic inhibition of ENT1 or by genetic removal of ENT1 enhanced the survival of R6/2 mice. Collectively, adenosine homeostasis and ENT1 expression are altered in HD. The inhibition of ENT1 can enhance extracellular adenosine level and be a potential therapeutic approach for treating HD.
Collapse
Affiliation(s)
- Yu-Han Kao
- School of Pharmacy, National Taiwan University, Taipei, Taiwan
| | - Meng-Syuan Lin
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
| | - Chiung-Mei Chen
- Department of Neurology, Chang Gung Memorial Hospital Linkou Medical Center and College of Medicine, Chang-Gung University, Tao-Yuan, Taiwan
| | - Yih-Ru Wu
- Department of Neurology, Chang Gung Memorial Hospital Linkou Medical Center and College of Medicine, Chang-Gung University, Tao-Yuan, Taiwan
| | - Hui-Mei Chen
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
| | - Hsing-Lin Lai
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
| | - Yijuang Chern
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
| | - Chun-Jung Lin
- School of Pharmacy, National Taiwan University, Taipei, Taiwan
| |
Collapse
|
37
|
Pharmacological targeting of adenosine receptor signaling. Mol Aspects Med 2017; 55:4-8. [PMID: 28088486 DOI: 10.1016/j.mam.2016.12.002] [Citation(s) in RCA: 53] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2016] [Revised: 12/22/2016] [Accepted: 12/23/2016] [Indexed: 12/20/2022]
Abstract
Adenosine receptor signaling plays important roles in normal physiology, but is also known to modulate the development or progression of several different diseases. The design of new, efficient, and safe pharmacological approaches to target the adenosine system may have considerable therapeutic potential, but is also associated with many challenges. This review summarizes the main challenges of adenosine receptor targeted treatment including tolerance, disease stage, cell type-specific effects, caffeine intake, adenosine level assessment and receptor distribution in vivo. Moreover, we discuss several potential ways to overcome these obstacles (i.e., the use of partial agonists, indirect receptor targeting, allosteric enhancers, prodrugs, non-receptor-mediated effects, neoreceptors, conditional knockouts). It is important to address these concerns during development of new and successful therapeutic approaches targeting the adenosine system.
Collapse
|
38
|
Voller J, Maková B, Kadlecová A, Gonzalez G, Strnad M. Plant Hormone Cytokinins for Modulating Human Aging and Age-Related Diseases. HEALTHY AGEING AND LONGEVITY 2017. [DOI: 10.1007/978-3-319-63001-4_14] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
|
39
|
The Role of Reactive Oxygen Species in the Pathogenesis of Alzheimer's Disease, Parkinson's Disease, and Huntington's Disease: A Mini Review. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2016; 2016:8590578. [PMID: 28116038 PMCID: PMC5223034 DOI: 10.1155/2016/8590578] [Citation(s) in RCA: 314] [Impact Index Per Article: 34.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/08/2016] [Revised: 10/06/2016] [Accepted: 11/13/2016] [Indexed: 11/18/2022]
Abstract
Neurodegenerative diseases affect not only the life quality of aging populations, but also their life spans. All forms of neurodegenerative diseases have a massive impact on the elderly. The major threat of these brain diseases includes progressive loss of memory, Alzheimer's disease (AD), impairments in the movement, Parkinson's disease (PD), and the inability to walk, talk, and think, Huntington's disease (HD). Oxidative stress and mitochondrial dysfunction are highlighted as a central feature of brain degenerative diseases. Oxidative stress, a condition that occurs due to imbalance in oxidant and antioxidant status, has been known to play a vital role in the pathophysiology of neurodegenerative diseases including AD, PD, and HD. A large number of studies have utilized oxidative stress biomarkers to investigate the severity of these neurodegenerative diseases and medications are available, but these only treat the symptoms. In traditional medicine, a large number of medicinal plants have been used to treat the symptoms of these neurodegenerative diseases. Extensive studies scientifically validated the beneficial effect of natural products against neurodegenerative diseases using suitable animal models. This short review focuses the role of oxidative stress in the pathogenesis of AD, PD, and HD and the protective efficacy of natural products against these diseases.
Collapse
|
40
|
Gastrolatathioneine, an unusual ergothioneine derivative from an aqueous extract of “tian ma”: A natural product co-produced by plant and symbiotic fungus. CHINESE CHEM LETT 2016. [DOI: 10.1016/j.cclet.2016.06.040] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
|
41
|
Guitart X, Bonaventura J, Rea W, Orrú M, Cellai L, Dettori I, Pedata F, Brugarolas M, Cortés A, Casadó V, Chang CP, Narayanan M, Chern Y, Ferré S. Equilibrative nucleoside transporter ENT1 as a biomarker of Huntington disease. Neurobiol Dis 2016; 96:47-53. [PMID: 27567601 DOI: 10.1016/j.nbd.2016.08.013] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2016] [Revised: 08/05/2016] [Accepted: 08/22/2016] [Indexed: 02/03/2023] Open
Abstract
The initial goal of this study was to investigate alterations in adenosine A2A receptor (A2AR) density or function in a rat model of Huntington disease (HD) with reported insensitivity to an A2AR antagonist. Unsuspected negative results led to the hypothesis of a low striatal adenosine tone and to the search for the mechanisms involved. Extracellular striatal concentrations of adenosine were measured with in vivo microdialysis in two rodent models of early neuropathological stages of HD disease, the Tg51 rat and the zQ175 knock-in mouse. In view of the crucial role of the equilibrative nucleoside transporter (ENT1) in determining extracellular content of adenosine, the binding properties of the ENT1 inhibitor [3H]-S-(4-Nitrobenzyl)-6-thioinosine were evaluated in zQ175 mice and the differential expression and differential coexpression patterns of the ENT1 gene (SLC29A1) were analyzed in a large human cohort of HD disease and controls. Extracellular striatal levels of adenosine were significantly lower in both animal models as compared with control littermates and striatal ENT1 binding sites were significantly upregulated in zQ175 mice. ENT1 transcript was significantly upregulated in HD disease patients at an early neuropathological severity stage, but not those with a higher severity stage, relative to non-demented controls. ENT1 transcript was differentially coexpressed (gained correlations) with several other genes in HD disease subjects compared to the control group. The present study demonstrates that ENT1 and adenosine constitute biomarkers of the initial stages of neurodegeneration in HD disease and also predicts that ENT1 could constitute a new therapeutic target to delay the progression of the disease.
Collapse
Affiliation(s)
- Xavier Guitart
- Integrative Neurobiology Section, National Institute on Drug Abuse, Intramural Research Program, National Institutes of Health, Baltimore, MD 21224, United States
| | - Jordi Bonaventura
- Integrative Neurobiology Section, National Institute on Drug Abuse, Intramural Research Program, National Institutes of Health, Baltimore, MD 21224, United States
| | - William Rea
- Integrative Neurobiology Section, National Institute on Drug Abuse, Intramural Research Program, National Institutes of Health, Baltimore, MD 21224, United States
| | - Marco Orrú
- Integrative Neurobiology Section, National Institute on Drug Abuse, Intramural Research Program, National Institutes of Health, Baltimore, MD 21224, United States
| | - Lucrezia Cellai
- Department NEUROFARBA, Division of Pharmacology and Toxicology, University of Florence, 50139 Florence, Italy
| | - Ilaria Dettori
- Department NEUROFARBA, Division of Pharmacology and Toxicology, University of Florence, 50139 Florence, Italy
| | - Felicita Pedata
- Department NEUROFARBA, Division of Pharmacology and Toxicology, University of Florence, 50139 Florence, Italy
| | - Marc Brugarolas
- Department of Biochemistry and Molecular Biology, Faculty of Biology, University of Barcelona and Center for Biomedical Research in Neurodegenerative Diseases Network and Institute of Biomedicine, 08028 Barcelona, Spain
| | - Antonio Cortés
- Department of Biochemistry and Molecular Biology, Faculty of Biology, University of Barcelona and Center for Biomedical Research in Neurodegenerative Diseases Network and Institute of Biomedicine, 08028 Barcelona, Spain
| | - Vicent Casadó
- Department of Biochemistry and Molecular Biology, Faculty of Biology, University of Barcelona and Center for Biomedical Research in Neurodegenerative Diseases Network and Institute of Biomedicine, 08028 Barcelona, Spain
| | - Ching-Pang Chang
- Division of Neuroscience Institute of Biomedical Sciences, Academia Sinica, 11529 Taipei, Taiwan
| | - Manikandan Narayanan
- Systems Genomics and Bioinformatics Unit, Laboratory of Systems Biology, National Institute of Allergy and Infectious Diseases, Intramural Research Program, National Institutes of Health, Bethesda, MD 20892, United States
| | - Yijuang Chern
- Division of Neuroscience Institute of Biomedical Sciences, Academia Sinica, 11529 Taipei, Taiwan
| | - Sergi Ferré
- Integrative Neurobiology Section, National Institute on Drug Abuse, Intramural Research Program, National Institutes of Health, Baltimore, MD 21224, United States.
| |
Collapse
|
42
|
Chiu FL, Lin JT, Chuang CY, Chien T, Chen CM, Chen KH, Hsiao HY, Lin YS, Chern Y, Kuo HC. Elucidating the role of the A 2Aadenosine receptor in neurodegeneration using neurons derived from Huntington's disease iPSCs. Hum Mol Genet 2015; 24:6066-6079. [DOI: 10.1093/hmg/ddv318] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/30/2023] Open
|
43
|
Chou AH, Chen YL, Chiu CC, Yuan SJ, Weng YH, Yeh TH, Lin YL, Fang JM, Wang HL. T1-11 and JMF1907 ameliorate polyglutamine-expanded ataxin-3-induced neurodegeneration, transcriptional dysregulation and ataxic symptom in the SCA3 transgenic mouse. Neuropharmacology 2015; 99:308-17. [PMID: 26254860 DOI: 10.1016/j.neuropharm.2015.08.009] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2015] [Revised: 08/04/2015] [Accepted: 08/04/2015] [Indexed: 11/24/2022]
Abstract
More studies are required to develop therapeutic agents for treating spinocerebellar ataxia type 3 (SCA3), which is caused by mutant polyglutamine-expanded ataxin-3 and is the most prevalent subtype of spinocerebellar ataxias. T1-11 [N6-(4-Hydroxybenzyl) adenosine], isolated from a Chinese medicinal herb Gastordia elata, is an adenosine A2A receptor agonist. SCA3 and Huntington's disease (HD) belong to a family of polyglutamine neurodegenerative diseases. T1-11 exerted a therapeutic effect on HD transgenic mouse by decreasing protein level of polyglutamine-expanded huntingtin in the striatum. In the present study, we test the possibility that T1-11 or JMF1907 [N6-(3-Indolylethyl) adenosine], a synthetic analog of T1-11, alleviates pontine neuronal death, cerebellar transcriptional downregulation and ataxic symptom in the SCA3 transgenic mouse expressing HA-tagged polyglutamine-expanded ataxin-3-Q79 (ataxin-3-Q79HA). Daily oral administration of T1-11 or JMF1907 prevented neuronal death of pontine nuclei in the SCA3 mouse with a dose-dependent manner. Oral application of T1-11 or JMF1907 reversed mutant ataxin-3-Q79-induced cerebellar transcriptional repression in the SCA3 transgenic mouse. T1-11 or JMF1907 ameliorated the symptom of motor incoordination displayed by SCA3 mouse. Oral administration of T1-11 or JMF1907 significantly decreased protein level of ataxin-3-Q79HA in the pontine nuclei or cerebellum of SCA3 mouse. T1-11 or JMF1907 significantly augmented the chymotrypsin-like activity of proteasome in the pontine nuclei or cerebellum of SCA3 mouse. Our results suggests that T1-11 and JMF1907 alleviate pontine neuronal death, cerebellar transcriptional downregulation and ataxic symptom of SCA3 transgenic mouse by augmenting the proteasome activity and reducing the protein level of polyglutamine-expanded ataxin-3-Q79 in the pontine nuclei and cerebellum.
Collapse
Affiliation(s)
- An-Hsun Chou
- Department of Anesthesiology, Chang Gung Memorial Hospital, Taoyuan, Taiwan, ROC
| | - Ying-Ling Chen
- Chang Gung University of Science and Technology, Taoyuan, Taiwan, ROC
| | - Ching-Chi Chiu
- Neuroscience Research Center, Chang Gung Memorial Hospital, Taoyuan, Taiwan, ROC
| | - Shin-Je Yuan
- Department of Physiology and Pharmacology, College of Medicine, Chang Gung University, Taoyuan, Taiwan, ROC
| | - Yi-Hsin Weng
- Neuroscience Research Center, Chang Gung Memorial Hospital, Taoyuan, Taiwan, ROC; Department of Neurology, Chang Gung Memorial Hospital, Taoyuan, Taiwan, ROC; College of Medicine, Chang Gung University, Taoyuan, Taiwan, ROC
| | - Tu-Hsueh Yeh
- Neuroscience Research Center, Chang Gung Memorial Hospital, Taoyuan, Taiwan, ROC; Department of Neurology, Chang Gung Memorial Hospital, Taoyuan, Taiwan, ROC; College of Medicine, Chang Gung University, Taoyuan, Taiwan, ROC
| | - Yun-Lian Lin
- National Research Institute of Chinese Medicine, Taipei, Taiwan, ROC
| | - Jim-Min Fang
- Department of Chemistry, National Taiwan University, Taipei, Taiwan, ROC
| | - Hung-Li Wang
- Neuroscience Research Center, Chang Gung Memorial Hospital, Taoyuan, Taiwan, ROC; Department of Physiology and Pharmacology, College of Medicine, Chang Gung University, Taoyuan, Taiwan, ROC; Healthy Aging Research Center, Chang Gung University, Taoyuan, Taiwan, ROC.
| |
Collapse
|
44
|
Guo Q, Wang Y, Lin S, Zhu C, Chen M, Jiang Z, Xu C, Zhang D, Wei H, Shi J. 4-Hydroxybenzyl-substituted amino acid derivatives from Gastrodia elata. Acta Pharm Sin B 2015; 5:350-7. [PMID: 26579466 PMCID: PMC4629276 DOI: 10.1016/j.apsb.2015.02.002] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2015] [Revised: 02/26/2015] [Accepted: 02/26/2015] [Indexed: 11/29/2022] Open
Abstract
Seven new 4-hydroxybenzyl-substituted amino acid derivatives (1−7), together with 11 known compounds, were isolated from an aqueous extract of the rhizomes of Gastrodia elata Blume. Their structures were determined by spectroscopic and chemical methods. Compounds 1−3 are pyroglutamate derivatives containing 4-hydroxybenzyl units at the N atom and 4−7 are the first examples of natural products with the 4-hydroxybenzyl unit linked via a thioether bond to 2-hydroxy-3-mercaptopropanoic acid (4−6) and 2-hydroxy-4-mercaptobutanoic acid (7), which would be biogenetically derived from cysteine and homocysteine, respectively. The structures of 1 and 2 were verified by synthesis, while the absolute configurations of 4, 5 and 7 were assigned using Mosher’s method based on the MPA determination rule of ΔδRS values. The known compound 4-(hydroxymethyl)-5-nitrobenzene-1,2-diol (8) exhibited activity against Fe2+-cysteine induced rat liver microsomal lipid peroxidation with IC50 values of 9.99×10−6 mol/L.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | - Jiangong Shi
- Corresponding author. Tel.: +86 10 83154789; fax: +86 10 63037757.
| |
Collapse
|
45
|
Mo C, Hannan AJ, Renoir T. Environmental factors as modulators of neurodegeneration: Insights from gene–environment interactions in Huntington's disease. Neurosci Biobehav Rev 2015; 52:178-92. [DOI: 10.1016/j.neubiorev.2015.03.003] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2014] [Revised: 02/13/2015] [Accepted: 03/03/2015] [Indexed: 12/11/2022]
|
46
|
Li W, Silva HB, Real J, Wang YM, Rial D, Li P, Payen MP, Zhou Y, Muller CE, Tomé AR, Cunha RA, Chen JF. Inactivation of adenosine A2A receptors reverses working memory deficits at early stages of Huntington's disease models. Neurobiol Dis 2015; 79:70-80. [PMID: 25892655 DOI: 10.1016/j.nbd.2015.03.030] [Citation(s) in RCA: 78] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2014] [Revised: 03/18/2015] [Accepted: 03/31/2015] [Indexed: 01/23/2023] Open
Abstract
Cognitive impairments in Huntington's disease (HD) are attributed to a dysfunction of the cortico-striatal pathway and significantly affect the quality of life of the patients, but this has not been a therapeutic focus in HD to date. We postulated that adenosine A(2A) receptors (A(2A)R), located at pre- and post-synaptic elements of the cortico-striatal pathways, modulate striatal neurotransmission and synaptic plasticity and cognitive behaviors. To critically evaluate the ability of A(2A)R inactivation to prevent cognitive deficits in early HD, we cross-bred A(2A)R knockout (KO) mice with two R6/2 transgenic lines of HD (CAG120 and CAG240) to generate two double transgenic R6/2-CAG120-A(2A)R KO and R6/2-CAG240-A(2A)R KO mice and their corresponding wild-type (WT) littermates. Genetic inactivation of A(2A)R prevented working memory deficits induced by R6/2-CAG120 at post-natal week 6 and by R6/2-CAG240 at post-natal month 2 and post-natal month 3, without modifying motor deficits. Similarly the A2(A)R antagonist KW6002 selectively reverted working memory deficits in R6/2-CAG240 mice at post-natal month 3. The search for possible mechanisms indicated that the genetic inactivation of A(2A)R did not affect ubiquitin-positive neuronal inclusions, astrogliosis or Thr-75 phosphorylation of DARPP-32 in the striatum. Importantly, A(2A)R blockade preferentially controlled long-term depression at cortico-striatal synapses in R6/2-CAG240 at post-natal week 6. The reported reversal of working memory deficits in R6/2 mice by the genetic and pharmacological inactivation of A(2A)R provides a proof-of-principle for A(2A)R as novel targets to reverse cognitive deficits in HD, likely by controlling LTD deregulation.
Collapse
Affiliation(s)
- Wei Li
- Department of Neurology, Boston University School of Medicine, Boston, MA, USA; Department of Neurology, Research Institute of Surgery, Daping Hospital, Third Military Medical University, Chongqing, China
| | - Henrique B Silva
- CNC-Center for Neuroscience and Cell Biology, University of Coimbra, Portugal
| | - Joana Real
- CNC-Center for Neuroscience and Cell Biology, University of Coimbra, Portugal
| | - Yu-Mei Wang
- Department of Neurology, Boston University School of Medicine, Boston, MA, USA
| | - Daniel Rial
- CNC-Center for Neuroscience and Cell Biology, University of Coimbra, Portugal
| | - Ping Li
- Department of Neurology, Boston University School of Medicine, Boston, MA, USA; Molecular Biology Center, State Key Laboratory of Trauma, Burn, and Combined Injury, Research Institute of Surgery, Daping Hospital, Third Military Medical University, Chongqing, China
| | - Marie-Pierce Payen
- Department of Neurology, Boston University School of Medicine, Boston, MA, USA
| | - Yuanguo Zhou
- Molecular Biology Center, State Key Laboratory of Trauma, Burn, and Combined Injury, Research Institute of Surgery, Daping Hospital, Third Military Medical University, Chongqing, China
| | - Christa E Muller
- Department of Neurology, Boston University School of Medicine, Boston, MA, USA
| | - Angelo R Tomé
- CNC-Center for Neuroscience and Cell Biology, University of Coimbra, Portugal
| | - Rodrigo A Cunha
- CNC-Center for Neuroscience and Cell Biology, University of Coimbra, Portugal; FMUC-Faculty of Medicine, University of Coimbra, Portugal
| | - Jiang-Fan Chen
- Department of Neurology, Boston University School of Medicine, Boston, MA, USA.
| |
Collapse
|
47
|
Liu YJ, Lee LM, Lai HL, Chern Y. Aberrant activation of AMP-activated protein kinase contributes to the abnormal distribution of HuR in amyotrophic lateral sclerosis. FEBS Lett 2015; 589:432-9. [DOI: 10.1016/j.febslet.2014.12.029] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2014] [Revised: 12/08/2014] [Accepted: 12/26/2014] [Indexed: 10/24/2022]
|
48
|
Cho JA, Zhang X, Miller GM, Lencer WI, Nery FC. 4-Phenylbutyrate attenuates the ER stress response and cyclic AMP accumulation in DYT1 dystonia cell models. PLoS One 2014; 9:e110086. [PMID: 25379658 PMCID: PMC4224384 DOI: 10.1371/journal.pone.0110086] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2014] [Accepted: 09/13/2014] [Indexed: 01/01/2023] Open
Abstract
Dystonia is a neurological disorder in which sustained muscle contractions induce twisting and repetitive movements or abnormal posturing. DYT1 early-onset primary dystonia is the most common form of hereditary dystonia and is caused by deletion of a glutamic acid residue (302/303) near the carboxyl-terminus of encoded torsinA. TorsinA is localized primarily within the contiguous lumen of the endoplasmic reticulum (ER) and nuclear envelope (NE), and is hypothesized to function as a molecular chaperone and an important regulator of the ER stress-signaling pathway, but how the mutation in torsinA causes disease remains unclear. Multiple lines of evidence suggest that the clinical symptoms of dystonia result from abnormalities in dopamine (DA) signaling, and possibly involving its down-stream effector adenylate cyclase that produces the second messenger cyclic adenosine-3', 5'-monophosphate (cAMP). Here we find that mutation in torsinA induces ER stress, and inhibits the cyclic adenosine-3', 5'-monophosphate (cAMP) response to the adenylate cyclase agonist forskolin. Both defective mechanins are corrected by the small molecule 4-phenylbutyrate (4-PBA) that alleviates ER stress. Our results link torsinA, the ER-stress-response, and cAMP-dependent signaling, and suggest 4-PBA could also be used in dystonia treatment. Other pharmacological agents known to modulate the cAMP cascade, and ER stress may also be therapeutic in dystonia patients and can be tested in the models described here, thus supplementing current efforts centered on the dopamine pathway.
Collapse
Affiliation(s)
- Jin A. Cho
- Division of Gastroenterology/Cell Biology, Boston Children's Hospital, Harvard Medical School, Boston, MA, United States of America
| | - Xuan Zhang
- Neuroscience Center, Department of Neurology, and Center for Molecular Imaging Research, Department of Radiology, Massachusetts General Hospital and Program in Neuroscience, Harvard Medical School, Boston, MA, United States of America
| | - Gregory M. Miller
- Department of Pharmaceutical Sciences and Center for Drug Discovery, Northeastern University, Boston, MA, United States of America
| | - Wayne I. Lencer
- Division of Gastroenterology/Cell Biology, Boston Children's Hospital, Harvard Medical School, Boston, MA, United States of America
- Harvard Digestive Diseases Center, Harvard Medical School, Boston, MA, United States of America
| | - Flavia C. Nery
- Neuroscience Center, Department of Neurology, and Center for Molecular Imaging Research, Department of Radiology, Massachusetts General Hospital and Program in Neuroscience, Harvard Medical School, Boston, MA, United States of America
| |
Collapse
|
49
|
Liu YJ, Ju TC, Chen HM, Jang YS, Lee LM, Lai HL, Tai HC, Fang JM, Lin YL, Tu PH, Chern Y. Activation of AMP-activated protein kinase α1 mediates mislocalization of TDP-43 in amyotrophic lateral sclerosis. Hum Mol Genet 2014; 24:787-801. [DOI: 10.1093/hmg/ddu497] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
|
50
|
Dos Santos-Rodrigues A, Grañé-Boladeras N, Bicket A, Coe IR. Nucleoside transporters in the purinome. Neurochem Int 2014; 73:229-37. [PMID: 24704797 DOI: 10.1016/j.neuint.2014.03.014] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2013] [Revised: 03/23/2014] [Accepted: 03/24/2014] [Indexed: 01/20/2023]
Abstract
The purinome is a rich complex of proteins and cofactors that are involved in fundamental aspects of cellular homeostasis and cellular responses. The purinome is evolutionarily ancient and is made up of thousands of members. Our understanding of the mechanisms linking some parts of this complex network and the physiological relevance of the various connections is well advanced. However, our understanding of other parts of the purinome is less well developed. Our research focuses on the adenosine or nucleoside transporters (NTs), which are members of the membrane purinome. Nucleoside transporters are integral membrane proteins that are responsible for the flux of nucleosides, such as adenosine, and nucleoside analog drugs, used in a variety of anti-cancer, anti-viral and anti-parasite therapies, across cell membranes. Nucleoside transporters form the SLC28 and SLC29 families of solute carriers and the protein members of these families are widely distributed in human tissues including the central nervous system (CNS). NTs modulate purinergic signaling in the CNS primarily through their effects on modulating prevailing adenosine levels inside and outside the cell. By clearing the extracellular milieu of adenosine, NTs can terminate adenosine receptor-dependent signaling and this raises the possibility of regulatory feedback loops that tie together receptor signaling with transporter function. Despite the important role of NTs as modulators of purinergic signaling in the human body, very little is known about the nature or underlying mechanisms of regulation of either the SLC28 or SLC29 families, particularly within the context of the CNS purinome. Here we provide a brief overview of our current understanding of the regulation of members of the SLC29 family and highlight some interesting avenues for future research.
Collapse
Affiliation(s)
| | - Natalia Grañé-Boladeras
- Department of Chemistry and Biology, Faculty of Science, Ryerson University, Toronto, ON, Canada
| | - Alex Bicket
- Department of Biology, Faculty of Science, York University, Toronto, ON, Canada
| | - Imogen R Coe
- Department of Biology, Faculty of Science, York University, Toronto, ON, Canada; Department of Chemistry and Biology, Faculty of Science, Ryerson University, Toronto, ON, Canada.
| |
Collapse
|