1
|
Jeong Y, Kim MW, Lee SG, Park S, Jeong KS, Lee YH, Lee S, Chung HM, Kim J, Kim CY. Therapeutic effects of CGS21680, a selective A 2A receptor agonist, via BDNF-related pathways in R106W mutation Rett syndrome model. Biomed Pharmacother 2025; 183:117821. [PMID: 39813786 DOI: 10.1016/j.biopha.2025.117821] [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: 08/29/2024] [Revised: 12/24/2024] [Accepted: 01/09/2025] [Indexed: 01/18/2025] Open
Abstract
Rett syndrome (RTT) is a neurological disorder caused by a mutation in the X-linked methyl-CpG binding protein 2 (MECP2), leading to cognitive and motor skill regression. Therapeutic strategies aimed at increasing brain-derived neurotrophic factor (BDNF) levels have been reported; however, BDNF treatment has limitations, including the inability to penetrate the blood-brain barrier, a short half-life, and potential for adverse effects when administered via intrathecal injection, necessitating novel therapeutic approaches. In this study, we focused on the adenosine A2A receptor (A2AR), which modulates BDNF and its downstream pathways, and investigated the therapeutic potential of CGS21680, an A2AR agonist, through in vitro and in vivo studies using R106W RTT model. CGS21680 restored neurite outgrowth, the number of SYN1+/MAP2+ puncta pairs, genes related to the BDNF-TrkB signaling pathway (Bdnf, TrkB, and Mtor) and neural development (Tuj1 and Syn1), and electrophysiological functions in in vitro RTT primary neurons. Additionally, CGS21680 alleviated neurobehavioral impairments and modulated gene expression in an RTT in vivo model. Our findings suggest that activation of A2AR via CGS21680 enhances BDNF-TrkB signaling, which in turn activates downstream pathways, ultimately increasing neurite outgrowth and synaptic plasticity, and restoring neurobehavioral clinical symptoms. This is the first study to report the therapeutic effect of CGS21680 in R106W point mutation RTT models, both in vitro and in vivo. These research results suggest that CGS21680 could be a promising therapeutic candidate for the treatment of RTT.
Collapse
Affiliation(s)
- Youngin Jeong
- College of Veterinary Medicine, Konkuk University, 120, Neungdong-ro, Gwangjin-gu, Seoul 05029, Republic of Korea
| | - Min Woo Kim
- College of Veterinary Medicine, Konkuk University, 120, Neungdong-ro, Gwangjin-gu, Seoul 05029, Republic of Korea
| | - Seul-Gi Lee
- College of Veterinary Medicine, Konkuk University, 120, Neungdong-ro, Gwangjin-gu, Seoul 05029, Republic of Korea
| | - Shinhye Park
- College of Veterinary Medicine, Konkuk University, 120, Neungdong-ro, Gwangjin-gu, Seoul 05029, Republic of Korea
| | - Kyu Sik Jeong
- College of Veterinary Medicine, Konkuk University, 120, Neungdong-ro, Gwangjin-gu, Seoul 05029, Republic of Korea
| | - Yun Hyeong Lee
- College of Veterinary Medicine, Konkuk University, 120, Neungdong-ro, Gwangjin-gu, Seoul 05029, Republic of Korea
| | - Suemin Lee
- College of Veterinary Medicine, Konkuk University, 120, Neungdong-ro, Gwangjin-gu, Seoul 05029, Republic of Korea
| | - Hyung Min Chung
- Department of Stem Cell Biology, College of Medicine, Konkuk University, Seoul 05029, Republic of Korea
| | - Jin Kim
- Department of Physiology, College of Medicine, Soonchunhyang University, Cheonan 31151, Republic of Korea.
| | - C-Yoon Kim
- College of Veterinary Medicine, Konkuk University, 120, Neungdong-ro, Gwangjin-gu, Seoul 05029, Republic of Korea.
| |
Collapse
|
2
|
Di Palma M, Lattanzi D, Ambrogini P, Sartini S. Adenosine A2A receptor activation is necessary to gate the TrkB-dependent intramuscular nerve sprouting during muscle reinnervation after a nerve crush. Heliyon 2025; 11:e41441. [PMID: 39839513 PMCID: PMC11748686 DOI: 10.1016/j.heliyon.2024.e41441] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2024] [Revised: 12/17/2024] [Accepted: 12/22/2024] [Indexed: 01/23/2025] Open
Abstract
Compelling evidence has demonstrated that rehabilitation through physical exercise, a non-invasive and non-surgical intervention, enhances muscle reinnervation and motor recovery after peripheral nerve injury (PNI) by increasing muscle-derived brain-derived neurotrophic factor (BDNF) expression and triggering TrkB-dependent axonal plasticity. Adenosine has been widely acknowledged to trigger TrkB via A2A receptor (A2AR). Since motor nerve terminals co-express TrkBs and A2ARs and depolarizing conditions increase muscle release of BDNF and adenosine, we examined whether A2ARs activation could recapitulate the functional recovery benefits of intermittent exercise after a nerve crush. Immunohistochemical and in situ proximity ligation assay (isPLA) analyses were used to localize A2ARs and A2A-TrkB heteroreceptor complexes at the neuromuscular level in undenervated animals. The reinnervation process of the soleus muscle was examined in both sedentary and trained animals ten days following a nerve crush injury. The effects of A2A and TrkB interplay on muscle fiber multiple-innervation was assessed using a functional approach. We confirmed that A2A immunoreactivity is mainly localized at the axonal level and provided evidence that A2ARs may form heteroreceptor complexes with TrKb at muscle plasmalemma. The pharmacological activation of either TrkBs or A2ARs mirrored the effect of motor activity on target muscle reinnervation after a nerve crush. Furthermore, the block of A2ARs abolished the effect of TrkBs agonism on nerve endings sprouting. Our results demonstrated that activation of adenosine A2ARs is required to gate the activity-related TrkB-dependent enhancement of axon sprouting during the reinnervation process after a nerve crush. Moreover, our isPLA data suggest that A2ARs can physically interact with TrkBs at the muscle plasmalemma.
Collapse
Affiliation(s)
- Michael Di Palma
- Department of Experimental and Clinical Medicine, Section of Neuroscience and Cell Biology, Università Politecnica Delle Marche, 60026, Ancona, Italy
| | - Davide Lattanzi
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, Urbino, Italy
| | - Patrizia Ambrogini
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, Urbino, Italy
| | - Stefano Sartini
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, Urbino, Italy
| |
Collapse
|
3
|
Chaptal MC, Maraninchi M, Musto G, Mancini J, Chtioui H, Dupont-Roussel J, Marlinge M, Fromonot J, Lalevee N, Mourre F, Beliard S, Guieu R, Valero R, Mottola G. Low Density Lipoprotein Cholesterol Decreases the Expression of Adenosine A 2A Receptor and Lipid Rafts-Protein Flotillin-1: Insights on Cardiovascular Risk of Hypercholesterolemia. Cells 2024; 13:488. [PMID: 38534331 PMCID: PMC10969546 DOI: 10.3390/cells13060488] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Revised: 02/26/2024] [Accepted: 03/07/2024] [Indexed: 03/28/2024] Open
Abstract
High blood levels of low-density lipoprotein (LDL)-cholesterol (LDL-C) are associated with atherosclerosis, mainly by promoting foam cell accumulation in vessels. As cholesterol is an essential component of cell plasma membranes and a regulator of several signaling pathways, LDL-C excess may have wider cardiovascular toxicity. We examined, in untreated hypercholesterolemia (HC) patients, selected regardless of the cause of LDL-C accumulation, and in healthy participants (HP), the expression of the adenosine A2A receptor (A2AR), an anti-inflammatory and vasodilatory protein with cholesterol-dependent modulation, and Flotillin-1, protein marker of cholesterol-enriched plasma membrane domains. Blood cardiovascular risk and inflammatory biomarkers were measured. A2AR and Flotillin-1 expression in peripheral blood mononuclear cells (PBMC) was lower in patients compared to HP and negatively correlated to LDL-C blood levels. No other differences were observed between the two groups apart from transferrin and ferritin concentrations. A2AR and Flotillin-1 proteins levels were positively correlated in the whole study population. Incubation of HP PBMCs with LDL-C caused a similar reduction in A2AR and Flotillin-1 expression. We suggest that LDL-C affects A2AR expression by impacting cholesterol-enriched membrane microdomains. Our results provide new insights into the molecular mechanisms underlying cholesterol toxicity, and may have important clinical implication for assessment and treatment of cardiovascular risk in HC.
Collapse
Affiliation(s)
- Marie-Charlotte Chaptal
- Centre de Recherche en Cardiovasculaire et Nutrition (C2VN), Aix-Marseille Université, INSERM 1263, INRAE 1260, 13005 Marseille, France; (M.-C.C.); (M.M.); (G.M.); (J.M.); (J.F.); (N.L.); (F.M.); (S.B.); (R.G.); (R.V.)
| | - Marie Maraninchi
- Centre de Recherche en Cardiovasculaire et Nutrition (C2VN), Aix-Marseille Université, INSERM 1263, INRAE 1260, 13005 Marseille, France; (M.-C.C.); (M.M.); (G.M.); (J.M.); (J.F.); (N.L.); (F.M.); (S.B.); (R.G.); (R.V.)
| | - Giorgia Musto
- Centre de Recherche en Cardiovasculaire et Nutrition (C2VN), Aix-Marseille Université, INSERM 1263, INRAE 1260, 13005 Marseille, France; (M.-C.C.); (M.M.); (G.M.); (J.M.); (J.F.); (N.L.); (F.M.); (S.B.); (R.G.); (R.V.)
- Department of Pharmacy, University of Naples Federico II, 80131 Naples, Italy
| | - Julien Mancini
- Centre de Recherche en Cardiovasculaire et Nutrition (C2VN), Aix-Marseille Université, INSERM 1263, INRAE 1260, 13005 Marseille, France; (M.-C.C.); (M.M.); (G.M.); (J.M.); (J.F.); (N.L.); (F.M.); (S.B.); (R.G.); (R.V.)
| | - Hedi Chtioui
- Department of Nutrition, Metabolic Diseases and Endocrinology, Hospital La Conception, APHM, 13005 Marseille, France; (H.C.); (J.D.-R.)
| | - Janine Dupont-Roussel
- Department of Nutrition, Metabolic Diseases and Endocrinology, Hospital La Conception, APHM, 13005 Marseille, France; (H.C.); (J.D.-R.)
| | - Marion Marlinge
- Centre de Recherche en Cardiovasculaire et Nutrition (C2VN), Aix-Marseille Université, INSERM 1263, INRAE 1260, 13005 Marseille, France; (M.-C.C.); (M.M.); (G.M.); (J.M.); (J.F.); (N.L.); (F.M.); (S.B.); (R.G.); (R.V.)
- Secteur de Biochimie, Biogenopôle, Hôpital de la Timone, APHM, 13005 Marseille, France
| | - Julien Fromonot
- Centre de Recherche en Cardiovasculaire et Nutrition (C2VN), Aix-Marseille Université, INSERM 1263, INRAE 1260, 13005 Marseille, France; (M.-C.C.); (M.M.); (G.M.); (J.M.); (J.F.); (N.L.); (F.M.); (S.B.); (R.G.); (R.V.)
- Secteur de Biochimie, Biogenopôle, Hôpital de la Timone, APHM, 13005 Marseille, France
| | - Nathalie Lalevee
- Centre de Recherche en Cardiovasculaire et Nutrition (C2VN), Aix-Marseille Université, INSERM 1263, INRAE 1260, 13005 Marseille, France; (M.-C.C.); (M.M.); (G.M.); (J.M.); (J.F.); (N.L.); (F.M.); (S.B.); (R.G.); (R.V.)
| | - Florian Mourre
- Centre de Recherche en Cardiovasculaire et Nutrition (C2VN), Aix-Marseille Université, INSERM 1263, INRAE 1260, 13005 Marseille, France; (M.-C.C.); (M.M.); (G.M.); (J.M.); (J.F.); (N.L.); (F.M.); (S.B.); (R.G.); (R.V.)
- Department of Nutrition, Metabolic Diseases and Endocrinology, Hospital La Conception, APHM, 13005 Marseille, France; (H.C.); (J.D.-R.)
| | - Sophie Beliard
- Centre de Recherche en Cardiovasculaire et Nutrition (C2VN), Aix-Marseille Université, INSERM 1263, INRAE 1260, 13005 Marseille, France; (M.-C.C.); (M.M.); (G.M.); (J.M.); (J.F.); (N.L.); (F.M.); (S.B.); (R.G.); (R.V.)
- Department of Nutrition, Metabolic Diseases and Endocrinology, Hospital La Conception, APHM, 13005 Marseille, France; (H.C.); (J.D.-R.)
| | - Régis Guieu
- Centre de Recherche en Cardiovasculaire et Nutrition (C2VN), Aix-Marseille Université, INSERM 1263, INRAE 1260, 13005 Marseille, France; (M.-C.C.); (M.M.); (G.M.); (J.M.); (J.F.); (N.L.); (F.M.); (S.B.); (R.G.); (R.V.)
- Secteur de Biochimie, Biogenopôle, Hôpital de la Timone, APHM, 13005 Marseille, France
| | - René Valero
- Centre de Recherche en Cardiovasculaire et Nutrition (C2VN), Aix-Marseille Université, INSERM 1263, INRAE 1260, 13005 Marseille, France; (M.-C.C.); (M.M.); (G.M.); (J.M.); (J.F.); (N.L.); (F.M.); (S.B.); (R.G.); (R.V.)
- Department of Nutrition, Metabolic Diseases and Endocrinology, Hospital La Conception, APHM, 13005 Marseille, France; (H.C.); (J.D.-R.)
| | - Giovanna Mottola
- Centre de Recherche en Cardiovasculaire et Nutrition (C2VN), Aix-Marseille Université, INSERM 1263, INRAE 1260, 13005 Marseille, France; (M.-C.C.); (M.M.); (G.M.); (J.M.); (J.F.); (N.L.); (F.M.); (S.B.); (R.G.); (R.V.)
- Secteur de Biochimie, Biogenopôle, Hôpital de la Timone, APHM, 13005 Marseille, France
| |
Collapse
|
4
|
Deng C, Chen H. Brain-derived neurotrophic factor/tropomyosin receptor kinase B signaling in spinal muscular atrophy and amyotrophic lateral sclerosis. Neurobiol Dis 2024; 190:106377. [PMID: 38092270 DOI: 10.1016/j.nbd.2023.106377] [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: 09/17/2023] [Revised: 11/15/2023] [Accepted: 12/10/2023] [Indexed: 12/23/2023] Open
Abstract
Tropomyosin receptor kinase B (TrkB) and its primary ligand brain-derived neurotrophic factor (BDNF) are expressed in the neuromuscular system, where they affect neuronal survival, differentiation, and functions. Changes in BDNF levels and full-length TrkB (TrkB-FL) signaling have been revealed in spinal muscular atrophy (SMA) and amyotrophic lateral sclerosis (ALS), two common forms of motor neuron diseases that are characterized by defective neuromuscular junctions in early disease stages and subsequently progressive muscle weakness. This review summarizes the current understanding of BDNF/TrkB-FL-related research in SMA and ALS, with an emphasis on their alterations in the neuromuscular system and possible BDNF/TrkB-FL-targeting therapeutic strategies. The limitations of current studies and future directions are also discussed, giving the hope of discovering novel and effective treatments.
Collapse
Affiliation(s)
- Chunchu Deng
- Department of Rehabilitation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Hong Chen
- Department of Rehabilitation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| |
Collapse
|
5
|
O'Neill KM, Anderson ED, Mukherjee S, Gandu S, McEwan SA, Omelchenko A, Rodriguez AR, Losert W, Meaney DF, Babadi B, Firestein BL. Time-dependent homeostatic mechanisms underlie brain-derived neurotrophic factor action on neural circuitry. Commun Biol 2023; 6:1278. [PMID: 38110605 PMCID: PMC10728104 DOI: 10.1038/s42003-023-05638-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2023] [Accepted: 11/27/2023] [Indexed: 12/20/2023] Open
Abstract
Plasticity and homeostatic mechanisms allow neural networks to maintain proper function while responding to physiological challenges. Despite previous work investigating morphological and synaptic effects of brain-derived neurotrophic factor (BDNF), the most prevalent growth factor in the central nervous system, how exposure to BDNF manifests at the network level remains unknown. Here we report that BDNF treatment affects rodent hippocampal network dynamics during development and recovery from glutamate-induced excitotoxicity in culture. Importantly, these effects are not obvious when traditional activity metrics are used, so we delve more deeply into network organization, functional analyses, and in silico simulations. We demonstrate that BDNF partially restores homeostasis by promoting recovery of weak and medium connections after injury. Imaging and computational analyses suggest these effects are caused by changes to inhibitory neurons and connections. From our in silico simulations, we find that BDNF remodels the network by indirectly strengthening weak excitatory synapses after injury. Ultimately, our findings may explain the difficulties encountered in preclinical and clinical trials with BDNF and also offer information for future trials to consider.
Collapse
Affiliation(s)
- Kate M O'Neill
- Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, NJ, USA
- Biomedical Engineering Graduate Program, Rutgers University, Piscataway, NJ, USA
- Institute for Physical Science & Technology, University of Maryland, College Park, MD, USA
| | - Erin D Anderson
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, USA
| | - Shoutik Mukherjee
- Department of Electrical and Computer Engineering, University of Maryland, College Park, MD, USA
| | - Srinivasa Gandu
- Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, NJ, USA
- Cell and Developmental Biology Graduate Program, Rutgers University, Piscataway, NJ, USA
| | - Sara A McEwan
- Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, NJ, USA
- Neuroscience Graduate Program, Rutgers University, Piscataway, NJ, USA
| | - Anton Omelchenko
- Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, NJ, USA
- Neuroscience Graduate Program, Rutgers University, Piscataway, NJ, USA
| | - Ana R Rodriguez
- Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, NJ, USA
- Biomedical Engineering Graduate Program, Rutgers University, Piscataway, NJ, USA
| | - Wolfgang Losert
- Department of Physics, University of Maryland, College Park, MD, USA
- Institute for Physical Science & Technology, University of Maryland, College Park, MD, USA
| | - David F Meaney
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, USA
- Department of Neurosurgery, University of Pennsylvania, Philadelphia, PA, USA
| | - Behtash Babadi
- Department of Electrical and Computer Engineering, University of Maryland, College Park, MD, USA
| | - Bonnie L Firestein
- Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, NJ, USA.
| |
Collapse
|
6
|
Vázquez-Cuevas FG, Reyna-Jeldes M, Velázquez-Miranda E, Coddou C. Transactivation of receptor tyrosine kinases by purinergic P2Y and adenosine receptors. Purinergic Signal 2023; 19:613-621. [PMID: 36529846 PMCID: PMC10754767 DOI: 10.1007/s11302-022-09913-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2022] [Accepted: 12/05/2022] [Indexed: 12/23/2022] Open
Abstract
Transactivation of receptor tyrosine kinases (RTK) is a crosstalk mechanism exhibited by G-protein-coupled receptors (GPCR) to activate signaling pathways classically associated with growth factors. The discovery of RTK transactivation was a breakthrough in signal transduction that contributed to developing current concepts in intracellular signaling. RTK transactivation links GPCR signaling to important cellular processes, such as cell proliferation and differentiation, and explains the functional diversity of these receptors. Purinergic (P2Y and adenosine) receptors belong to class A of GPCR; in the present work, we systematically review the experimental evidence showing that purinergic receptors have the ability to transactivate RTK in multiple tissues and physiopathological conditions resulting in the modulation of cellular physiology. Of particular relevance, the crosstalk between purinergic receptors and epidermal growth factor receptor is a redundant pathway that participates in multiple pathophysiological processes. Specific and detailed knowledge of purinergic receptor-regulated pathways advances our understanding of the complexity of GPCR signal transduction and opens the way for pharmacologic intervention in the pathological context.
Collapse
Affiliation(s)
- F G Vázquez-Cuevas
- Departamento de Neurobiología Celular Y Molecular, Instituto de Neurobiología, Universidad Nacional Autónoma de México, Boulevard Juriquilla # 3001, Juriquilla, Querétaro, 76230, México.
| | - M Reyna-Jeldes
- Departamento de Ciencias Biomédicas, Facultad de Medicina, Universidad Católica del Norte, Larrondo 1281, Coquimbo, 1781421, Chile
- Millennium Nucleus for the Study of Pain (MiNuSPain), Santiago, 8331150, Chile
- Núcleo Para El Estudio del Cáncer a Nivel Básico, Aplicado Y Clínico, Universidad Católica del Norte, Larrondo 1281, Coquimbo , 1781421, Chile
| | - E Velázquez-Miranda
- Departamento de Neurobiología Celular Y Molecular, Instituto de Neurobiología, Universidad Nacional Autónoma de México, Boulevard Juriquilla # 3001, Juriquilla, Querétaro, 76230, México
| | - C Coddou
- Departamento de Ciencias Biomédicas, Facultad de Medicina, Universidad Católica del Norte, Larrondo 1281, Coquimbo, 1781421, Chile.
- Millennium Nucleus for the Study of Pain (MiNuSPain), Santiago, 8331150, Chile.
- Núcleo Para El Estudio del Cáncer a Nivel Básico, Aplicado Y Clínico, Universidad Católica del Norte, Larrondo 1281, Coquimbo , 1781421, Chile.
| |
Collapse
|
7
|
Samhan-Arias AK, Poejo J, Marques-da-Silva D, Martínez-Costa OH, Gutierrez-Merino C. Hexa-Histidine, a Peptide with Versatile Applications in the Study of Amyloid-β(1-42) Molecular Mechanisms of Action. Molecules 2023; 28:7138. [PMID: 38067638 PMCID: PMC10708093 DOI: 10.3390/molecules28237909] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2023] [Revised: 10/10/2023] [Accepted: 10/13/2023] [Indexed: 12/18/2023] Open
Abstract
Amyloid β (Aβ) oligomers are the most neurotoxic forms of Aβ, and Aβ(1-42) is the prevalent Aβ peptide found in the amyloid plaques of Alzheimer's disease patients. Aβ(25-35) is the shortest peptide that retains the toxicity of Aβ(1-42). Aβ oligomers bind to calmodulin (CaM) and calbindin-D28k with dissociation constants in the nanomolar Aβ(1-42) concentration range. Aβ and histidine-rich proteins have a high affinity for transition metal ions Cu2+, Fe3+ and Zn2+. In this work, we show that the fluorescence of Aβ(1-42) HiLyteTM-Fluor555 can be used to monitor hexa-histidine peptide (His6) interaction with Aβ(1-42). The formation of His6/Aβ(1-42) complexes is also supported by docking results yielded by the MDockPeP Server. Also, we found that micromolar concentrations of His6 block the increase in the fluorescence of Aβ(1-42) HiLyteTM-Fluor555 produced by its interaction with the proteins CaM and calbindin-D28k. In addition, we found that the His6-tag provides a high-affinity site for the binding of Aβ(1-42) and Aβ(25-35) peptides to the human recombinant cytochrome b5 reductase, and sensitizes this enzyme to inhibition by these peptides. In conclusion, our results suggest that a His6-tag could provide a valuable new tool to experimentally direct the action of neurotoxic Aβ peptides toward selected cellular targets.
Collapse
Affiliation(s)
- Alejandro K. Samhan-Arias
- Departamento de Bioquímica, Universidad Autónoma de Madrid (UAM), C/Arturo Duperier 4, 28029 Madrid, Spain;
- Instituto de Investigaciones Biomédicas ‘Sols-Morreale’ (CSIC-UAM), C/Arturo Duperier 4, 28029 Madrid, Spain
| | - Joana Poejo
- Instituto de Biomarcadores de Patologías Moleculares, Universidad de Extremadura, 06006 Badajoz, Spain;
| | - Dorinda Marques-da-Silva
- LSRE—Laboratory of Separation and Reaction Engineering and LCM—Laboratory of Catalysis and Materials, School of Management and Technology, Polytechnic Institute of Leiria, Morro do Lena-Alto do Vieiro, 2411-901 Leiria, Portugal;
- ALiCE—Associate Laboratory in Chemical Engineering, Faculty of Engineering, University of Porto, Rua Dr. Roberto Frias, 4200-465 Porto, Portugal
- School of Technology and Management, Polytechnic Institute of Leiria, Morro do Lena-Alto do Vieiro, 2411-901 Leiria, Portugal
| | - Oscar H. Martínez-Costa
- Departamento de Bioquímica, Universidad Autónoma de Madrid (UAM), C/Arturo Duperier 4, 28029 Madrid, Spain;
- Instituto de Investigaciones Biomédicas ‘Sols-Morreale’ (CSIC-UAM), C/Arturo Duperier 4, 28029 Madrid, Spain
| | - Carlos Gutierrez-Merino
- Instituto de Biomarcadores de Patologías Moleculares, Universidad de Extremadura, 06006 Badajoz, Spain;
| |
Collapse
|
8
|
Stansberry WM, Pierchala BA. Neurotrophic factors in the physiology of motor neurons and their role in the pathobiology and therapeutic approach to amyotrophic lateral sclerosis. Front Mol Neurosci 2023; 16:1238453. [PMID: 37692101 PMCID: PMC10483118 DOI: 10.3389/fnmol.2023.1238453] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2023] [Accepted: 08/11/2023] [Indexed: 09/12/2023] Open
Abstract
The discovery of the neurotrophins and their potent survival and trophic effects led to great enthusiasm about their therapeutic potential to rescue dying neurons in neurodegenerative diseases. The further discovery that brain-derived neurotrophic factor (BDNF), ciliary neurotrophic factor (CNTF) and glial cell line-derived neurotrophic factor (GDNF) had potent survival-promoting activity on motor neurons led to the proposal for their use in motor neuron diseases such as amyotrophic lateral sclerosis (ALS). In this review we synthesize the literature pertaining to the role of NGF, BDNF, CNTF and GDNF on the development and physiology of spinal motor neurons, as well as the preclinical studies that evaluated their potential for the treatment of ALS. Results from the clinical trials of these molecules will also be described and, with the aid of decades of hindsight, we will discuss what can reasonably be concluded and how this information can inform future clinical development of neurotrophic factors for ALS.
Collapse
Affiliation(s)
- Wesley M. Stansberry
- The Department of Anatomy, Cell Biology and Physiology, Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN, United States
- Medical Neuroscience Graduate Program, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Brian A. Pierchala
- The Department of Anatomy, Cell Biology and Physiology, Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN, United States
- Medical Neuroscience Graduate Program, Indiana University School of Medicine, Indianapolis, IN, United States
| |
Collapse
|
9
|
Vanecek AS, Mojsilovic-Petrovic J, Kalb RG, Tepe JJ. Enhanced Degradation of Mutant C9ORF72-Derived Toxic Dipeptide Repeat Proteins by 20S Proteasome Activation Results in Restoration of Proteostasis and Neuroprotection. ACS Chem Neurosci 2023. [PMID: 37015082 DOI: 10.1021/acschemneuro.2c00732] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/06/2023] Open
Abstract
A hexanucleotide repeat expansion (HRE) in an intron of gene C9ORF72 is the most common cause of familial amyotrophic lateral sclerosis and frontotemporal dementia. The HRE undergoes noncanonical translation (repeat-associated non-ATG translation) resulting in the production of five distinct dipeptide repeat (DPR) proteins. Arginine-rich DPR proteins have shown to be toxic to motor neurons, and recent evidence suggests this toxicity is associated with disruption of the ubiquitin-proteasome system. Here we report the ability of known 20S proteasome activator, TCH-165, to enhance the degradation of DPR proteins and overcome proteasome impairment evoked by DPR proteins. Furthermore, the 20S activator protects rodent motor neurons from DPR protein toxicity and restores proteostasis in cortical neuron cultures. This study suggests that 20S proteasome enhancers may have therapeutic efficacy in neurodegenerative diseases that display proteostasis defects.
Collapse
Affiliation(s)
- Allison S Vanecek
- Department of Chemistry, Michigan State University, East Lansing, Michigan 48824, United States
| | - Jelena Mojsilovic-Petrovic
- Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, Illinois 60611, United States
| | - Robert G Kalb
- Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, Illinois 60611, United States
| | - Jetze J Tepe
- Department of Chemistry, Michigan State University, East Lansing, Michigan 48824, United States
| |
Collapse
|
10
|
Mansour HM, El-Khatib AS. Repositioning of receptor tyrosine kinase inhibitors. RECEPTOR TYROSINE KINASES IN NEURODEGENERATIVE AND PSYCHIATRIC DISORDERS 2023:353-401. [DOI: 10.1016/b978-0-443-18677-6.00010-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
11
|
Bassani D, Pavan M, Federico S, Spalluto G, Sturlese M, Moro S. The Multifaceted Role of GPCRs in Amyotrophic Lateral Sclerosis: A New Therapeutic Perspective? Int J Mol Sci 2022; 23:4504. [PMID: 35562894 PMCID: PMC9106011 DOI: 10.3390/ijms23094504] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Revised: 04/15/2022] [Accepted: 04/15/2022] [Indexed: 02/05/2023] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a degenerating disease involving the motor neurons, which causes a progressive loss of movement ability, usually leading to death within 2 to 5 years from the diagnosis. Much effort has been put into research for an effective therapy for its eradication, but still, no cure is available. The only two drugs approved for this pathology, Riluzole and Edaravone, are onlyable to slow down the inevitable disease progression. As assessed in the literature, drug targets such as protein kinases have already been extensively examined as potential drug targets for ALS, with some molecules already in clinical trials. Here, we focus on the involvement of another very important and studied class of biological entities, G protein-coupled receptors (GPCRs), in the onset and progression of ALS. This workaimsto give an overview of what has been already discovered on the topic, providing useful information and insights that can be used by scientists all around the world who are putting efforts into the fight against this very important neurodegenerating disease.
Collapse
Affiliation(s)
- Davide Bassani
- Molecular Modeling Section (MMS), Department of Pharmaceutical and Pharmacological Sciences, University of Padova, 35131 Padova, Italy; (D.B.); (M.P.); (M.S.)
| | - Matteo Pavan
- Molecular Modeling Section (MMS), Department of Pharmaceutical and Pharmacological Sciences, University of Padova, 35131 Padova, Italy; (D.B.); (M.P.); (M.S.)
| | - Stephanie Federico
- Department of Chemical and Pharmaceutical Sciences, University of Trieste, 34127 Trieste, Italy; (S.F.); (G.S.)
| | - Giampiero Spalluto
- Department of Chemical and Pharmaceutical Sciences, University of Trieste, 34127 Trieste, Italy; (S.F.); (G.S.)
| | - Mattia Sturlese
- Molecular Modeling Section (MMS), Department of Pharmaceutical and Pharmacological Sciences, University of Padova, 35131 Padova, Italy; (D.B.); (M.P.); (M.S.)
| | - Stefano Moro
- Molecular Modeling Section (MMS), Department of Pharmaceutical and Pharmacological Sciences, University of Padova, 35131 Padova, Italy; (D.B.); (M.P.); (M.S.)
| |
Collapse
|
12
|
Neurons undergo pathogenic metabolic reprogramming in models of familial ALS. Mol Metab 2022; 60:101468. [PMID: 35248787 PMCID: PMC8958550 DOI: 10.1016/j.molmet.2022.101468] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Revised: 02/25/2022] [Accepted: 02/25/2022] [Indexed: 11/21/2022] Open
Abstract
Objectives Methods Results Conclusions Our work is the first to perform a comprehensive and quantitative analysis of intermediary metabolism in neurons in the setting of fALS causing gene products. Because the cardinal feature of ALS is death of motor neurons, these new studies are directly relevant to the pathogenesis of ALS. Our functional interrogations begin to unpack how metabolic re-wiring is induced by fALS genes and it will be very interesting, in the future, to gain insight in amino acid fueling of the TCA cycle. We suspect pleiotropic effects of amino acid fueling, and this may lead to very targeted therapeutic interventions.
Collapse
|
13
|
Banerjee P, Elliott E, Rifai OM, O'Shaughnessy J, McDade K, Abrahams S, Chandran S, Smith C, Gregory JM. NLRP3 inflammasome as a key molecular target underlying cognitive resilience in amyotrophic lateral sclerosis. J Pathol 2022; 256:262-268. [PMID: 34883532 DOI: 10.1002/path.5846] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2021] [Revised: 11/05/2021] [Accepted: 12/06/2021] [Indexed: 11/09/2022]
Abstract
Up to 50% of amyotrophic lateral sclerosis patients present with cognitive deficits in addition to motor dysfunction, but the molecular mechanisms underlying diverse clinical and pathological presentations remain poorly understood. There is therefore an unmet need to identify molecular drivers of cognitive dysfunction to enable better therapeutic targeting and prognostication. To address this, we employed a non-biased approach to identify molecular targets using a deeply phenotyped, clinically stratified cohort of cognitively affected and unaffected brain regions from three brain regions of 13 amyotrophic lateral sclerosis patients with the same cognitive screening test performed during life. Using NanoString molecular barcoding as a sensitive mRNA sequencing technique on post-mortem tissue, we profiled a data-driven panel of 770 genes using the Neuropathology Panel, followed by region and cell type-specific validation using BaseScope in situ hybridisation and immunohistochemistry. We identified 50 significantly dysregulated genes that are distinct between cognitively affected and unaffected brain regions. Using BaseScope in situ hybridisation, we also demonstrate that macromolecular complex regulation, notably NLRP3 inflammasome modulation, is a potential, therapeutically targetable, pathological correlate of cognitive resilience in ALS. © 2021 The Authors. The Journal of Pathology published by John Wiley & Sons, Ltd on behalf of The Pathological Society of Great Britain and Ireland.
Collapse
Affiliation(s)
- Poulomi Banerjee
- Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, UK
- UK Dementia Research Institute, University of Edinburgh, Edinburgh, UK
- The Euan MacDonald Centre, University of Edinburgh, Edinburgh, UK
| | - Elizabeth Elliott
- Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, UK
- UK Dementia Research Institute, University of Edinburgh, Edinburgh, UK
- The Euan MacDonald Centre, University of Edinburgh, Edinburgh, UK
| | - Olivia M Rifai
- Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, UK
- UK Dementia Research Institute, University of Edinburgh, Edinburgh, UK
- The Euan MacDonald Centre, University of Edinburgh, Edinburgh, UK
| | - Judi O'Shaughnessy
- Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, UK
| | - Karina McDade
- Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, UK
| | - Sharon Abrahams
- The Euan MacDonald Centre, University of Edinburgh, Edinburgh, UK
- School of Philosophy, Psychology and Language Science, University of Edinburgh, Edinburgh, UK
- The Anne Rowling Regenerative Neurology Clinic, University of Edinburgh, Edinburgh, UK
| | - Siddharthan Chandran
- Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, UK
- UK Dementia Research Institute, University of Edinburgh, Edinburgh, UK
- The Euan MacDonald Centre, University of Edinburgh, Edinburgh, UK
- The Anne Rowling Regenerative Neurology Clinic, University of Edinburgh, Edinburgh, UK
| | - Colin Smith
- Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, UK
- The Euan MacDonald Centre, University of Edinburgh, Edinburgh, UK
| | - Jenna M Gregory
- Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, UK
- UK Dementia Research Institute, University of Edinburgh, Edinburgh, UK
- The Euan MacDonald Centre, University of Edinburgh, Edinburgh, UK
| |
Collapse
|
14
|
Huang SK, Almurad O, Pejana RJ, Morrison ZA, Pandey A, Picard LP, Nitz M, Sljoka A, Prosser RS. Allosteric modulation of the adenosine A 2A receptor by cholesterol. eLife 2022; 11:e73901. [PMID: 34986091 PMCID: PMC8730723 DOI: 10.7554/elife.73901] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2021] [Accepted: 12/23/2021] [Indexed: 12/18/2022] Open
Abstract
Cholesterol is a major component of the cell membrane and commonly regulates membrane protein function. Here, we investigate how cholesterol modulates the conformational equilibria and signaling of the adenosine A2A receptor (A2AR) in reconstituted phospholipid nanodiscs. This model system conveniently excludes possible effects arising from cholesterol-induced phase separation or receptor oligomerization and focuses on the question of allostery. GTP hydrolysis assays show that cholesterol weakly enhances the basal signaling of A2AR while decreasing the agonist EC50. Fluorine nuclear magnetic resonance (19F NMR) spectroscopy shows that this enhancement arises from an increase in the receptor's active state population and a G-protein-bound precoupled state. 19F NMR of fluorinated cholesterol analogs reveals transient interactions with A2AR, indicating a lack of high-affinity binding or direct allosteric modulation. The combined results suggest that the observed allosteric effects are largely indirect and originate from cholesterol-mediated changes in membrane properties, as shown by membrane fluidity measurements and high-pressure NMR.
Collapse
Affiliation(s)
- Shuya Kate Huang
- Department of Chemistry, University of TorontoTorontoCanada
- Department of Chemical and Physical Sciences, University of Toronto MississaugaMississaugaCanada
| | - Omar Almurad
- Department of Chemistry, University of TorontoTorontoCanada
- Department of Chemical and Physical Sciences, University of Toronto MississaugaMississaugaCanada
| | - Reizel J Pejana
- Department of Chemistry, University of TorontoTorontoCanada
- Department of Chemical and Physical Sciences, University of Toronto MississaugaMississaugaCanada
| | | | - Aditya Pandey
- Department of Chemistry, University of TorontoTorontoCanada
- Department of Chemical and Physical Sciences, University of Toronto MississaugaMississaugaCanada
| | - Louis-Philippe Picard
- Department of Chemistry, University of TorontoTorontoCanada
- Department of Chemical and Physical Sciences, University of Toronto MississaugaMississaugaCanada
| | - Mark Nitz
- Department of Chemistry, University of TorontoTorontoCanada
| | - Adnan Sljoka
- RIKEN Center for Advanced Intelligence ProjectTokyoJapan
- York University, Department of ChemistryTorontoCanada
| | - R Scott Prosser
- Department of Chemistry, University of TorontoTorontoCanada
- Department of Chemical and Physical Sciences, University of Toronto MississaugaMississaugaCanada
- Department of Biochemistry, University of TorontoTorontoCanada
| |
Collapse
|
15
|
Rochat C, Bernard-Marissal N, Källstig E, Pradervand S, Perrin FE, Aebischer P, Raoul C, Schneider BL. Astrocyte-targeting RNA interference against mutated superoxide dismutase 1 induces motoneuron plasticity and protects fast-fatigable motor units in a mouse model of amyotrophic lateral sclerosis. Glia 2022; 70:842-857. [PMID: 34978340 PMCID: PMC9303637 DOI: 10.1002/glia.24140] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2021] [Revised: 12/19/2021] [Accepted: 12/22/2021] [Indexed: 11/22/2022]
Abstract
In amyotrophic lateral sclerosis (ALS) caused by SOD1 gene mutations, both cell‐autonomous and noncell‐autonomous mechanisms lead to the selective degeneration of motoneurons (MN). Here, we evaluate the therapeutic potential of gene therapy targeting mutated SOD1 in mature astrocytes using mice expressing the mutated SOD1G93A protein. An AAV‐gfaABC1D vector encoding an artificial microRNA is used to deliver RNA interference against mutated SOD1 selectively in astrocytes. The treatment leads to the progressive rescue of neuromuscular junction occupancy, to the recovery of the compound muscle action potential in the gastrocnemius muscle, and significantly improves neuromuscular function. In the spinal cord, gene therapy targeting astrocytes protects a small pool of the most vulnerable fast‐fatigable MN until disease end stage. In the gastrocnemius muscle of the treated SOD1G93A mice, the fast‐twitch type IIB muscle fibers are preserved from atrophy. Axon collateral sprouting is observed together with muscle fiber type grouping indicative of denervation/reinnervation events. The transcriptome profiling of spinal cord MN shows changes in the expression levels of factors regulating the dynamics of microtubules. Gene therapy delivering RNA interference against mutated SOD1 in astrocytes protects fast‐fatigable motor units and thereby improves neuromuscular function in ALS mice.
Collapse
Affiliation(s)
- Cylia Rochat
- Ecole Polytechnique Fédérale de Lausanne (EPFL), Brain Mind Institute, Lausanne
| | - Nathalie Bernard-Marissal
- Ecole Polytechnique Fédérale de Lausanne (EPFL), Brain Mind Institute, Lausanne.,INSERM, MMG, Aix-Marseille University, Marseille, France
| | - Emma Källstig
- Ecole Polytechnique Fédérale de Lausanne (EPFL), Brain Mind Institute, Lausanne.,Bertarelli Platform for Gene Therapy, Ecole Polytechnique Fédérale de Lausanne (EPFL), Geneva
| | - Sylvain Pradervand
- Genomic Technologies Facility, University of Lausanne, Lausanne, Switzerland
| | | | - Patrick Aebischer
- Ecole Polytechnique Fédérale de Lausanne (EPFL), Brain Mind Institute, Lausanne
| | - Cédric Raoul
- INM, Université Montpellier, INSERM, Montpellier, France
| | - Bernard L Schneider
- Ecole Polytechnique Fédérale de Lausanne (EPFL), Brain Mind Institute, Lausanne.,Bertarelli Platform for Gene Therapy, Ecole Polytechnique Fédérale de Lausanne (EPFL), Geneva
| |
Collapse
|
16
|
Sharma N, Tan MA, An SSA. Phytosterols: Potential Metabolic Modulators in Neurodegenerative Diseases. Int J Mol Sci 2021; 22:ijms222212255. [PMID: 34830148 PMCID: PMC8618769 DOI: 10.3390/ijms222212255] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2021] [Revised: 11/03/2021] [Accepted: 11/09/2021] [Indexed: 12/12/2022] Open
Abstract
Phytosterols constitute a class of natural products that are an important component of diet and have vast applications in foods, cosmetics, and herbal medicines. With many and diverse isolated structures in nature, they exhibit a broad range of biological and pharmacological activities. Among over 200 types of phytosterols, stigmasterol and β-sitosterol were ubiquitous in many plant species, exhibiting important aspects of activities related to neurodegenerative diseases. Hence, this mini-review presented an overview of the reported studies on selected phytosterols related to neurodegenerative diseases. It covered the major phytosterols based on biosynthetic considerations, including other phytosterols with significant in vitro and in vivo biological activities.
Collapse
Affiliation(s)
- Niti Sharma
- Bionano Research Institute, Gachon University, 1342 Seongnam-daero, Sujeong-gu, Seongnam-si 461-701, Gyeonggi-do, Korea;
| | - Mario A. Tan
- Research Center for the Natural and Applied Sciences, College of Science, University of Santo Tomas, Manila 1015, Philippines;
| | - Seong Soo A. An
- Bionano Research Institute, Gachon University, 1342 Seongnam-daero, Sujeong-gu, Seongnam-si 461-701, Gyeonggi-do, Korea;
- Correspondence: ; Tel.: +82-31-750-8755
| |
Collapse
|
17
|
The nuclear ubiquitin ligase adaptor SPOP is a conserved regulator of C9orf72 dipeptide toxicity. Proc Natl Acad Sci U S A 2021; 118:2104664118. [PMID: 34593637 DOI: 10.1073/pnas.2104664118] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/06/2021] [Indexed: 12/29/2022] Open
Abstract
A hexanucleotide repeat expansion in the C9orf72 gene is the most common cause of inherited amyotrophic lateral sclerosis (ALS) and frontotemporal dementia (FTD). Unconventional translation of the C9orf72 repeat produces dipeptide repeat proteins (DPRs). Previously, we showed that the DPRs PR50 and GR50 are highly toxic when expressed in Caenorhabditis elegans, and this toxicity depends on nuclear localization of the DPR. In an unbiased genome-wide RNA interference (RNAi) screen for suppressors of PR50 toxicity, we identified 12 genes that consistently suppressed either the developmental arrest and/or paralysis phenotype evoked by PR50 expression. All of these genes have vertebrate homologs, and 7 of 12 contain predicted nuclear localization signals. One of these genes was spop-1, the C. elegans homolog of SPOP, a nuclear localized E3 ubiquitin ligase adaptor only found in metazoans. SPOP is also required for GR50 toxicity and functions in a genetic pathway that includes cul-3, which is the canonical E3 ligase partner for SPOP Genetic or pharmacological inhibition of SPOP in mammalian primary spinal cord motor neurons suppressed DPR toxicity without affecting DPR expression levels. Finally, we find that knockdown of bromodomain proteins in both C. elegans and mammalian neurons, which are known SPOP ubiquitination targets, suppresses the protective effect of SPOP inhibition. Together, these data suggest a model in which SPOP promotes the DPR-dependent ubiquitination and degradation of BRD proteins. We speculate the pharmacological manipulation of this pathway, which is currently underway for multiple cancer subtypes, could also represent an entry point for therapeutic intervention to treat C9orf72 FTD/ALS.
Collapse
|
18
|
Agarwal P, Agarwal R. Tackling retinal ganglion cell apoptosis in glaucoma: role of adenosine receptors. Expert Opin Ther Targets 2021; 25:585-596. [PMID: 34402357 DOI: 10.1080/14728222.2021.1969362] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
INTRODUCTION The role of adenosine receptors as therapeutic targets for neuroprotection is now widely recognized. Their role, however, in protection against retinal ganglion cell (RGC) apoptosis in glaucoma needs further investigation. Hence, in this review, we look into the possibility of adenosine receptors as potential therapeutic targets by exploring their role in modulating various pathophysiological mechanisms underlying glaucomatous RGC loss. AREAS COVERED This review presents a summary of the adenosine receptor distribution in retina and the cellular functions mediated by them. The major pathophysiological mechanisms such as excitotoxicity, vascular dysregulation, loss of neurotrophic signaling, and inflammatory responses involved in glaucomatous RGC loss are discussed. The literature showing the role of adenosine receptors in modulating these pathophysiological mechanisms is discussed. The literature search was conducted using Pubmed search engine using key words such as 'RGC apoptosis,' 'adenosine,' adenosine receptors' 'retina' 'excitotoxicity,' 'neurotrophins,' 'ischemia', and 'cytokines' individually and in various combinations. EXPERT OPINION Use of adenosine receptor agonists and antagonists, for preservation of the RGCs in glaucomatous eyes independent of the level of intraocular pressure seems a very useful strategy. Future application of this strategy would require appropriate designing of drug formulation for tissue and disease-specific receptor targeting. Furthermore, the modulation of physiological functions and potential adverse effects need further investigations.
Collapse
Affiliation(s)
- Puneet Agarwal
- School of Medicine, International Medical University, Kuala Lumpur, Malaysia
| | - Renu Agarwal
- School of Medicine, International Medical University, Kuala Lumpur, Malaysia
| |
Collapse
|
19
|
How Are Adenosine and Adenosine A 2A Receptors Involved in the Pathophysiology of Amyotrophic Lateral Sclerosis? Biomedicines 2021; 9:biomedicines9081027. [PMID: 34440231 PMCID: PMC8392384 DOI: 10.3390/biomedicines9081027] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Revised: 08/03/2021] [Accepted: 08/12/2021] [Indexed: 12/19/2022] Open
Abstract
Adenosine is extensively distributed in the central and peripheral nervous systems, where it plays a key role as a neuromodulator. It has long been implicated in the pathogenesis of progressive neurogenerative disorders such as Parkinson’s disease, and there is now growing interest in its role in amyotrophic lateral sclerosis (ALS). The motor neurons affected in ALS are responsive to adenosine receptor function, and there is accumulating evidence for beneficial effects of adenosine A2A receptor antagonism. In this article, we focus on recent evidence from ALS clinical pathology and animal models that support dynamism of the adenosinergic system (including changes in adenosine levels and receptor changes) in ALS. We review the possible mechanisms of chronic neurodegeneration via the adenosinergic system, potential biomarkers and the acute symptomatic pharmacology, including respiratory motor neuron control, of A2A receptor antagonism to explore the potential of the A2A receptor as target for ALS therapy.
Collapse
|
20
|
Pregnolato F, Cova L, Doretti A, Bardelli D, Silani V, Bossolasco P. Exosome microRNAs in Amyotrophic Lateral Sclerosis: A Pilot Study. Biomolecules 2021; 11:biom11081220. [PMID: 34439885 PMCID: PMC8394507 DOI: 10.3390/biom11081220] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Revised: 08/11/2021] [Accepted: 08/13/2021] [Indexed: 12/11/2022] Open
Abstract
The pathogenesis of amyotrophic lateral sclerosis (ALS), a lethal neurodegenerative disease, remains undisclosed. Mutations in ALS related genes have been identified, albeit the majority of cases are unmutated. Clinical pathology of ALS suggests a prion-like cell-to-cell diffusion of the disease possibly mediated by exosomes, small endocytic vesicles involved in the propagation of RNA molecules and proteins. In this pilot study, we focused on exosomal microRNAs (miRNAs), key regulators of many signaling pathways. We analyzed serum-derived exosomes from ALS patients in comparison with healthy donors. Exosomes were obtained by a commercial kit. Purification of miRNAs was performed using spin column chromatography and RNA was reverse transcribed into cDNA. All samples were run on the miRCURY LNATM Universal RT miRNA PCR Serum/Plasma Focus panel. An average of 29 miRNAs were detectable per sample. The supervised analysis did not identify any statistically significant difference among the groups indicating that none of the miRNA of our panel has a strong pathological role in ALS. However, selecting samples with the highest miRNA content, six biological processes shared across miRNAs through the intersection of the GO categories were identified. Our results, combined to those reported in the literature, indicated that further investigation is needed to elucidate the role of exosome-derived miRNA in ALS.
Collapse
Affiliation(s)
- Francesca Pregnolato
- Experimental Laboratory of Immunological and Rheumatologic Researches, Istituto Auxologico Italiano, IRCCS, Cusano Milanino, 20095 Milan, Italy;
| | - Lidia Cova
- Department of Neurology-Stroke Unit and Laboratory of Neuroscience, Istituto Auxologico Italiano, IRCCS, 20149 Milan, Italy; (L.C.); (A.D.); (D.B.); (V.S.)
| | - Alberto Doretti
- Department of Neurology-Stroke Unit and Laboratory of Neuroscience, Istituto Auxologico Italiano, IRCCS, 20149 Milan, Italy; (L.C.); (A.D.); (D.B.); (V.S.)
| | - Donatella Bardelli
- Department of Neurology-Stroke Unit and Laboratory of Neuroscience, Istituto Auxologico Italiano, IRCCS, 20149 Milan, Italy; (L.C.); (A.D.); (D.B.); (V.S.)
| | - Vincenzo Silani
- Department of Neurology-Stroke Unit and Laboratory of Neuroscience, Istituto Auxologico Italiano, IRCCS, 20149 Milan, Italy; (L.C.); (A.D.); (D.B.); (V.S.)
- “Dino Ferrari” Center, Department of Pathophysiology and Transplantation, Università degli Studi di Milano, 20122 Milan, Italy
- “Aldo Ravelli” Center for Neurotechnology and Experimental Brain Therapeutics, Università degli Studi di Milano, 20122 Milan, Italy
| | - Patrizia Bossolasco
- Department of Neurology-Stroke Unit and Laboratory of Neuroscience, Istituto Auxologico Italiano, IRCCS, 20149 Milan, Italy; (L.C.); (A.D.); (D.B.); (V.S.)
- Correspondence:
| |
Collapse
|
21
|
Atif M, Alsrhani A, Naz F, Imran M, Imran M, Ullah MI, Alameen AAM, Gondal TA, Raza Q. Targeting Adenosine Receptors in Neurological Diseases. Cell Reprogram 2021; 23:57-72. [PMID: 33861641 DOI: 10.1089/cell.2020.0087] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Adenosine plays a significant role in neurotransmission process by controlling the blood pressure, while adenosine triphosphate (ATP) acts as a neuromodulator and neurotransmitter and by activation of P2 receptors, regulates the contractility of the heart. Adenosine signaling is essential in the process of regeneration by regulating proliferation, differentiation, and apoptosis of stem cells. In this review, we have selected neurological disorders (Alzheimer's disease, Parkinson's disease, amyotrophic lateral sclerosis, multiple sclerosis, and epilepsy) with clinical trials using antagonists and epigenetic tools targeting adenosine receptor as a therapeutic approach in the treatment of these disorders. Promising results have been reported from many clinical trials. It has been found that higher expression levels of A2A and P2X7 receptors in neurological disorders further complicate the disease condition. Therefore, modulations of these receptors by using antagonists of these receptors or SAM (S-adenosylmethionine) therapy as an epigenetic tool could be useful in reversing the complications of these disorders. Finally, we suggest that modulation of adenosine receptors in neurological disorders can increase the regenerative phase by increasing the rate of proliferation and differentiation in the damaged tissues.
Collapse
Affiliation(s)
- Muhmmad Atif
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Jouf University, Sakaka, Saudi Arabia
| | - Abdullah Alsrhani
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Jouf University, Sakaka, Saudi Arabia
| | - Farrah Naz
- Department of Microbiology, Government College University, Faisalabad, Pakistan
| | - Muhammad Imran
- University Institute of Diet and Nutritional Sciences, Faculty of Allied Health Sciences, The University of Lahore, Lahore, Pakistan
| | - Muhammad Imran
- Department of Microbiology, University of Health Sciences, Lahore, Pakistan
| | - Muhammad Ikram Ullah
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Jouf University, Sakaka, Saudi Arabia
| | - Ayman A M Alameen
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Jouf University, Sakaka, Saudi Arabia.,Department of Chemical Pathology, Faculty of Medical Laboratory Sciences, University of Khartoum, Khartoum, Sudan
| | - Tanweer Aslam Gondal
- School of Exercise and Nutrition, Faculty of Health, Deakin University, Victoria, Australia
| | - Qaisar Raza
- Department of Clinical Nutrition, NUR International University, Lahore, Pakistan
| |
Collapse
|
22
|
Nam Y, Moon GJ, Kim SR. Therapeutic Potential of AAV1-Rheb(S16H) Transduction against Neurodegenerative Diseases. Int J Mol Sci 2021; 22:ijms22063064. [PMID: 33802760 PMCID: PMC8002454 DOI: 10.3390/ijms22063064] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Revised: 03/08/2021] [Accepted: 03/16/2021] [Indexed: 01/19/2023] Open
Abstract
Neurotrophic factors (NTFs) are essential for cell growth, survival, synaptic plasticity, and maintenance of specific neuronal population in the central nervous system. Multiple studies have demonstrated that alterations in the levels and activities of NTFs are related to the pathology and symptoms of neurodegenerative disorders, such as Parkinson’s disease (PD), Alzheimer’s disease (AD), and Huntington’s disease. Hence, the key molecule that can regulate the expression of NTFs is an important target for gene therapy coupling adeno-associated virus vector (AAV) gene. We have previously reported that the Ras homolog protein enriched in brain (Rheb)–mammalian target of rapamycin complex 1 (mTORC1) axis plays a vital role in preventing neuronal death in the brain of AD and PD patients. AAV transduction using a constitutively active form of Rheb exerts a neuroprotective effect through the upregulation of NTFs, thereby promoting the neurotrophic interaction between astrocytes and neurons in AD conditions. These findings suggest the role of Rheb as an important regulator of the regulatory system of NTFs to treat neurodegenerative diseases. In this review, we present an overview of the role of Rheb in neurodegenerative diseases and summarize the therapeutic potential of AAV serotype 1 (AAV1)-Rheb(S16H) transduction in the treatment of neurodegenerative disorders, focusing on diseases, such as AD and PD.
Collapse
Affiliation(s)
- Youngpyo Nam
- Brain Science and Engineering Institute, Kyungpook National University, Daegu 41944, Korea;
| | - Gyeong Joon Moon
- Center for Cell Therapy, Asan Institute for Life Science, Asan Medical Center, Seoul 05505, Korea;
- Department of Convergence Medicine, University of Ulsan College of Medicine, Seoul 05505, Korea
| | - Sang Ryong Kim
- Brain Science and Engineering Institute, Kyungpook National University, Daegu 41944, Korea;
- School of Life Sciences, Kyungpook National University, Daegu 41566, Korea
- BK21 FOUR KNU Creative BioResearch Group, Kyungpook National University, Daegu 41566, Korea
- Correspondence: ; Tel.: +82-53-950-7362; Fax: +82-53-943-2762
| |
Collapse
|
23
|
Zakyrjanova GF, Giniatullin AR, Mukhutdinova KA, Kuznetsova EA, Petrov AM. Early differences in membrane properties at the neuromuscular junctions of ALS model mice: Effects of 25-hydroxycholesterol. Life Sci 2021; 273:119300. [PMID: 33662433 DOI: 10.1016/j.lfs.2021.119300] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2020] [Revised: 02/13/2021] [Accepted: 02/25/2021] [Indexed: 12/11/2022]
Abstract
AIMS Plasma hyperlipidemia is a protective factor in amyotrophic lateral sclerosis (ALS) while cholesterol-lowering drugs aggravate the pathology. We hypothesize that this phenomenon can be linked with membrane lipid alterations in the neuromuscular junctions (NMJs) occurring before motor neuron loss. METHODS Neurotransmitter release in parallel with lipid membrane properties in diaphragm NMJs of SOD1G93A (mSOD) mice at nine weeks of age (pre-onset stage) were assessed. KEY FINDINGS Despite on slight changes in spontaneous and evoked quantum release of acetylcholine, extracellular levels of choline at resting conditions, an indicator of non-quantum release, were significantly increased in mSOD mice. The use of lipid-sensitive fluorescent probes points to lipid raft disruption in the NMJs of mSOD mice. However, content of cholesterol, a key raft component was unchanged implying another pathway responsible for the loss of raft integrity. In the mSOD mice we found marked increase in levels of raft-destabilizing lipid ceramide. This was accompanied by enhanced ability to uptake of exogenous ceramide in NMJs. Acute and chronic administration of 25-hydroxycholesterol, whose levels increase due to hypercholesterolemia, recovered early alterations in membrane properties. Furthermore, chronic treatment with 25-hydroxycholesterol prevented increase in ceramide and extracellular choline levels as well as suppressed lipid peroxidation of NMJ membranes and fragmentation of end plates. SIGNIFICANCE Thus, lipid raft disruption likely due to ceramide accumulation could be early event in ALS which may trigger neuromuscular abnormalities. Cholesterol derivative 25-hydroxycholesterol may serve as a molecule restoring the membrane and functional properties of NMJs at the early stage.
Collapse
Affiliation(s)
- Guzel F Zakyrjanova
- Laboratory of Biophysics of Synaptic Processes, Kazan Institute of Biochemistry and Biophysics, Federal Research Center "Kazan Scientific Center of RAS", 2/31 Lobachevsky Street, box 30, Kazan 420111, Russia; Institute of Neuroscience, Kazan State Medial University, 49 Butlerova Street, Kazan, 420012, Russia
| | - Arthur R Giniatullin
- Department of Normal Physiology, Kazan State Medial University, 49 Butlerova Street, Kazan 420012, Russia
| | - Kamilla A Mukhutdinova
- Institute of Neuroscience, Kazan State Medial University, 49 Butlerova Street, Kazan, 420012, Russia
| | - Eva A Kuznetsova
- Laboratory of Biophysics of Synaptic Processes, Kazan Institute of Biochemistry and Biophysics, Federal Research Center "Kazan Scientific Center of RAS", 2/31 Lobachevsky Street, box 30, Kazan 420111, Russia
| | - Alexey M Petrov
- Laboratory of Biophysics of Synaptic Processes, Kazan Institute of Biochemistry and Biophysics, Federal Research Center "Kazan Scientific Center of RAS", 2/31 Lobachevsky Street, box 30, Kazan 420111, Russia; Institute of Neuroscience, Kazan State Medial University, 49 Butlerova Street, Kazan, 420012, Russia.
| |
Collapse
|
24
|
Scaricamazza S, Salvatori I, Ferri A, Valle C. Skeletal Muscle in ALS: An Unappreciated Therapeutic Opportunity? Cells 2021; 10:525. [PMID: 33801336 PMCID: PMC8000428 DOI: 10.3390/cells10030525] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2021] [Revised: 02/23/2021] [Accepted: 02/24/2021] [Indexed: 12/11/2022] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a neurodegenerative disorder characterized by the selective degeneration of upper and lower motor neurons and by the progressive weakness and paralysis of voluntary muscles. Despite intense research efforts and numerous clinical trials, it is still an incurable disease. ALS had long been considered a pure motor neuron disease; however, recent studies have shown that motor neuron protection is not sufficient to prevent the course of the disease since the dismantlement of neuromuscular junctions occurs before motor neuron degeneration. Skeletal muscle alterations have been described in the early stages of the disease, and they seem to be mainly involved in the "dying back" phenomenon of motor neurons and metabolic dysfunctions. In recent years, skeletal muscles have been considered crucial not only for the etiology of ALS but also for its treatment. Here, we review clinical and preclinical studies that targeted skeletal muscles and discuss the different approaches, including pharmacological interventions, supplements or diets, genetic modifications, and training programs.
Collapse
Affiliation(s)
- Silvia Scaricamazza
- Fondazione Santa Lucia IRCCS, c/o CERC, 00143 Rome, Italy; (S.S.); (I.S.)
- Department of Biology, University of Rome Tor Vergata, 00133 Rome, Italy
| | - Illari Salvatori
- Fondazione Santa Lucia IRCCS, c/o CERC, 00143 Rome, Italy; (S.S.); (I.S.)
- Department of Experimental Medicine, University of Rome “La Sapienza”, 00161 Rome, Italy
| | - Alberto Ferri
- Fondazione Santa Lucia IRCCS, c/o CERC, 00143 Rome, Italy; (S.S.); (I.S.)
- Institute of Translational Pharmacology, National Research Council, 00133 Rome, Italy
| | - Cristiana Valle
- Fondazione Santa Lucia IRCCS, c/o CERC, 00143 Rome, Italy; (S.S.); (I.S.)
- Institute of Translational Pharmacology, National Research Council, 00133 Rome, Italy
| |
Collapse
|
25
|
Miranda-Lourenço C, Ribeiro-Rodrigues L, Fonseca-Gomes J, Tanqueiro SR, Belo RF, Ferreira CB, Rei N, Ferreira-Manso M, de Almeida-Borlido C, Costa-Coelho T, Freitas CF, Zavalko S, Mouro FM, Sebastião AM, Xapelli S, Rodrigues TM, Diógenes MJ. Challenges of BDNF-based therapies: From common to rare diseases. Pharmacol Res 2020; 162:105281. [PMID: 33161136 DOI: 10.1016/j.phrs.2020.105281] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Revised: 10/20/2020] [Accepted: 10/22/2020] [Indexed: 12/11/2022]
Abstract
Neurotrophins are a well-known family of neurotrophic factors that play an important role both in the central and peripheral nervous systems, where they modulate neuronal survival, development, function and plasticity. Brain-derived neurotrophic factor (BDNF) possesses diverse biological functions which are mediated by the activation of two main classes of receptors, the tropomyosin-related kinase (Trk) B and the p75 neurotrophin receptor (p75NTR). The therapeutic potential of BDNF has drawn attention since dysregulation of its signalling cascades has been suggested to underlie the pathogenesis of both common and rare diseases. Multiple strategies targeting this neurotrophin have been tested; most have found obstacles that ultimately hampered their effectiveness. This review focuses on the involvement of BDNF and its receptors in the pathophysiology of Alzheimer's disease (AD), Amyotrophic Lateral Sclerosis (ALS) and Rett Syndrome (RTT). We describe the known mechanisms leading to the impairment of BDNF/TrkB signalling in these disorders. Such mechanistic insight highlights how BDNF signalling compromise can take various shapes, nearly disease-specific. Therefore, BDNF-based therapeutic strategies must be specifically tailored and are more likely to succeed if a combination of resources is employed.
Collapse
Affiliation(s)
- Catarina Miranda-Lourenço
- Instituto de Farmacologia e Neurociências, Faculdade de Medicina, Universidade de Lisboa, 1649-028 Lisboa, Portugal; Instituto de Medicina Molecular João Lobo Antunes, Universidade de Lisboa, 1649-028 Lisboa, Portugal
| | - Leonor Ribeiro-Rodrigues
- Instituto de Farmacologia e Neurociências, Faculdade de Medicina, Universidade de Lisboa, 1649-028 Lisboa, Portugal; Instituto de Medicina Molecular João Lobo Antunes, Universidade de Lisboa, 1649-028 Lisboa, Portugal
| | - João Fonseca-Gomes
- Instituto de Farmacologia e Neurociências, Faculdade de Medicina, Universidade de Lisboa, 1649-028 Lisboa, Portugal; Instituto de Medicina Molecular João Lobo Antunes, Universidade de Lisboa, 1649-028 Lisboa, Portugal
| | - Sara R Tanqueiro
- Instituto de Farmacologia e Neurociências, Faculdade de Medicina, Universidade de Lisboa, 1649-028 Lisboa, Portugal; Instituto de Medicina Molecular João Lobo Antunes, Universidade de Lisboa, 1649-028 Lisboa, Portugal
| | - Rita F Belo
- Instituto de Farmacologia e Neurociências, Faculdade de Medicina, Universidade de Lisboa, 1649-028 Lisboa, Portugal; Instituto de Medicina Molecular João Lobo Antunes, Universidade de Lisboa, 1649-028 Lisboa, Portugal
| | - Catarina B Ferreira
- Instituto de Farmacologia e Neurociências, Faculdade de Medicina, Universidade de Lisboa, 1649-028 Lisboa, Portugal; Instituto de Medicina Molecular João Lobo Antunes, Universidade de Lisboa, 1649-028 Lisboa, Portugal
| | - Nádia Rei
- Instituto de Farmacologia e Neurociências, Faculdade de Medicina, Universidade de Lisboa, 1649-028 Lisboa, Portugal; Instituto de Medicina Molecular João Lobo Antunes, Universidade de Lisboa, 1649-028 Lisboa, Portugal
| | - Mafalda Ferreira-Manso
- Instituto de Farmacologia e Neurociências, Faculdade de Medicina, Universidade de Lisboa, 1649-028 Lisboa, Portugal; Instituto de Medicina Molecular João Lobo Antunes, Universidade de Lisboa, 1649-028 Lisboa, Portugal
| | - Carolina de Almeida-Borlido
- Instituto de Farmacologia e Neurociências, Faculdade de Medicina, Universidade de Lisboa, 1649-028 Lisboa, Portugal; Instituto de Medicina Molecular João Lobo Antunes, Universidade de Lisboa, 1649-028 Lisboa, Portugal
| | - Tiago Costa-Coelho
- Instituto de Farmacologia e Neurociências, Faculdade de Medicina, Universidade de Lisboa, 1649-028 Lisboa, Portugal; Instituto de Medicina Molecular João Lobo Antunes, Universidade de Lisboa, 1649-028 Lisboa, Portugal
| | - Céline Felicidade Freitas
- Instituto de Farmacologia e Neurociências, Faculdade de Medicina, Universidade de Lisboa, 1649-028 Lisboa, Portugal; Instituto de Medicina Molecular João Lobo Antunes, Universidade de Lisboa, 1649-028 Lisboa, Portugal
| | - Svitlana Zavalko
- Instituto de Farmacologia e Neurociências, Faculdade de Medicina, Universidade de Lisboa, 1649-028 Lisboa, Portugal; Instituto de Medicina Molecular João Lobo Antunes, Universidade de Lisboa, 1649-028 Lisboa, Portugal
| | - Francisco M Mouro
- Instituto de Farmacologia e Neurociências, Faculdade de Medicina, Universidade de Lisboa, 1649-028 Lisboa, Portugal; Instituto de Medicina Molecular João Lobo Antunes, Universidade de Lisboa, 1649-028 Lisboa, Portugal
| | - Ana M Sebastião
- Instituto de Farmacologia e Neurociências, Faculdade de Medicina, Universidade de Lisboa, 1649-028 Lisboa, Portugal; Instituto de Medicina Molecular João Lobo Antunes, Universidade de Lisboa, 1649-028 Lisboa, Portugal
| | - Sara Xapelli
- Instituto de Farmacologia e Neurociências, Faculdade de Medicina, Universidade de Lisboa, 1649-028 Lisboa, Portugal; Instituto de Medicina Molecular João Lobo Antunes, Universidade de Lisboa, 1649-028 Lisboa, Portugal
| | - Tiago M Rodrigues
- Instituto de Farmacologia e Neurociências, Faculdade de Medicina, Universidade de Lisboa, 1649-028 Lisboa, Portugal; Instituto de Medicina Molecular João Lobo Antunes, Universidade de Lisboa, 1649-028 Lisboa, Portugal; Institute of Molecular and Clinical Ophthalmology Basel (IOB), Mittlere Strasse 91, 4031 Basel, Switzerland
| | - Maria J Diógenes
- Instituto de Farmacologia e Neurociências, Faculdade de Medicina, Universidade de Lisboa, 1649-028 Lisboa, Portugal; Instituto de Medicina Molecular João Lobo Antunes, Universidade de Lisboa, 1649-028 Lisboa, Portugal.
| |
Collapse
|
26
|
Di Palma M, Sartini S, Lattanzi D, Cuppini R, Pita-Rodriguez M, Diaz-Carmenate Y, Narvaez M, Fuxe K, Borroto-Escuela DO, Ambrogini P. Evidence for the existence of A2AR-TrkB heteroreceptor complexes in the dorsal hippocampus of the rat brain: Potential implications of A2AR and TrkB interplay upon ageing. Mech Ageing Dev 2020; 190:111289. [PMID: 32565059 DOI: 10.1016/j.mad.2020.111289] [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: 02/10/2020] [Revised: 06/14/2020] [Accepted: 06/16/2020] [Indexed: 10/24/2022]
Abstract
Adenosine A2A receptors (A2AR) are crucial in facilitating the BDNF action on synaptic transmission in the rat hippocampus primarily upon ageing. Furthermore, it has been suggested that A2AR-Tropomyosin related kinase B receptor (TrkB) crosstalk has a pivotal role in adenosine A2AR-mediated modulation of the BDNF action on hippocampal plasticity. Considering the impact of the above receptors interplay on what concerns BDNF-induced enhancement of synaptic transmission, gaining a better insight into the mechanisms behind this powerful crosstalk becomes of primary interest. Using in situ proximity ligation assay (PLA), the existence of a direct physical interaction between adenosine A2AR and TrkB is demonstrated. The A2AR-TrkB heteroreceptor complexes show a heterogeneous distribution within the rat dorsal hippocampus. High densities of the heteroreceptor complexes were observed in the pyramidal cell layers of CA1-CA3 regions and in the polymorphic layer of the dentate gyrus (DG). The stratum radiatum of the CA1-3 regions showed positive PLA signal in contrast to the oriens region. The molecular and granular layers of the DG also lacked significant densities of PLA positive heteroreceptor complexes, but subgranular zone showed some PLA positive cells. Their allosteric receptor-receptor interactions may significantly modulate BDNF signaling impacting on hippocampal plasticity which is impaired upon ageing.
Collapse
Affiliation(s)
- Michael Di Palma
- Department of Experimental and Clinical Medicine, Faculty of Medicine and Surgery, Università Politecnica Delle Marche, Ancona, Italy.
| | - Stefano Sartini
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, Urbino, Italy
| | - Davide Lattanzi
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, Urbino, Italy
| | - Riccardo Cuppini
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, Urbino, Italy
| | - Mariana Pita-Rodriguez
- Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden; Universidad de Málaga, Instituto de Investigación Biomédica de Málaga, Málaga, Spain; Departamento de Diagnóstico Molecular, Centro de Neurociencias de Cuba, La Habana, Cuba
| | | | - Manuel Narvaez
- Universidad de Málaga, Instituto de Investigación Biomédica de Málaga, Málaga, Spain
| | - Kjell Fuxe
- Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Dasiel O Borroto-Escuela
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, Urbino, Italy; Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Patrizia Ambrogini
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, Urbino, Italy
| |
Collapse
|
27
|
Andrejew R, Glaser T, Oliveira-Giacomelli Á, Ribeiro D, Godoy M, Granato A, Ulrich H. Targeting Purinergic Signaling and Cell Therapy in Cardiovascular and Neurodegenerative Diseases. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1201:275-353. [PMID: 31898792 DOI: 10.1007/978-3-030-31206-0_14] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Extracellular purines exert several functions in physiological and pathophysiological mechanisms. ATP acts through P2 receptors as a neurotransmitter and neuromodulator and modulates heart contractility, while adenosine participates in neurotransmission, blood pressure, and many other mechanisms. Because of their capability to differentiate into mature cell types, they provide a unique therapeutic strategy for regenerating damaged tissue, such as in cardiovascular and neurodegenerative diseases. Purinergic signaling is pivotal for controlling stem cell differentiation and phenotype determination. Proliferation, differentiation, and apoptosis of stem cells of various origins are regulated by purinergic receptors. In this chapter, we selected neurodegenerative and cardiovascular diseases with clinical trials using cell therapy and purinergic receptor targeting. We discuss these approaches as therapeutic alternatives to neurodegenerative and cardiovascular diseases. For instance, promising results were demonstrated in the utilization of mesenchymal stem cells and bone marrow mononuclear cells in vascular regeneration. Regarding neurodegenerative diseases, in general, P2X7 and A2A receptors mostly worsen the degenerative state. Stem cell-based therapy, mainly through mesenchymal and hematopoietic stem cells, showed promising results in improving symptoms caused by neurodegeneration. We propose that purinergic receptor activity regulation combined with stem cells could enhance proliferative and differentiation rates as well as cell engraftment.
Collapse
Affiliation(s)
- Roberta Andrejew
- Neuroscience Laboratory, Institute of Chemistry, Department of Biochemistry, University of São Paulo, São Paulo, Brazil
| | - Talita Glaser
- Neuroscience Laboratory, Institute of Chemistry, Department of Biochemistry, University of São Paulo, São Paulo, Brazil
| | - Ágatha Oliveira-Giacomelli
- Neuroscience Laboratory, Institute of Chemistry, Department of Biochemistry, University of São Paulo, São Paulo, Brazil
| | - Deidiane Ribeiro
- Neuroscience Laboratory, Institute of Chemistry, Department of Biochemistry, University of São Paulo, São Paulo, Brazil
| | - Mariana Godoy
- Neuroscience Laboratory, Institute of Chemistry, Department of Biochemistry, University of São Paulo, São Paulo, Brazil.,Laboratory of Neurodegenerative Diseases, Institute of Biophysics Carlos Chagas Filho, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Alessandro Granato
- Neuroscience Laboratory, Institute of Chemistry, Department of Biochemistry, University of São Paulo, São Paulo, Brazil
| | - Henning Ulrich
- Neuroscience Laboratory, Institute of Chemistry, Department of Biochemistry, University of São Paulo, São Paulo, Brazil.
| |
Collapse
|
28
|
Chiricozzi E, Lunghi G, Di Biase E, Fazzari M, Sonnino S, Mauri L. GM1 Ganglioside Is A Key Factor in Maintaining the Mammalian Neuronal Functions Avoiding Neurodegeneration. Int J Mol Sci 2020; 21:E868. [PMID: 32013258 PMCID: PMC7037093 DOI: 10.3390/ijms21030868] [Citation(s) in RCA: 94] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2019] [Revised: 01/23/2020] [Accepted: 01/27/2020] [Indexed: 12/11/2022] Open
Abstract
Many species of ganglioside GM1, differing for the sialic acid and ceramide content, have been characterized and their physico-chemical properties have been studied in detail since 1963. Scientists were immediately attracted to the GM1 molecule and have carried on an ever-increasing number of studies to understand its binding properties and its neurotrophic and neuroprotective role. GM1 displays a well balanced amphiphilic behavior that allows to establish strong both hydrophobic and hydrophilic interactions. The peculiar structure of GM1 reduces the fluidity of the plasma membrane which implies a retention and enrichment of the ganglioside in specific membrane domains called lipid rafts. The dynamism of the GM1 oligosaccharide head allows it to assume different conformations and, in this way, to interact through hydrogen or ionic bonds with a wide range of membrane receptors as well as with extracellular ligands. After more than 60 years of studies, it is a milestone that GM1 is one of the main actors in determining the neuronal functions that allows humans to have an intellectual life. The progressive reduction of its biosynthesis along the lifespan is being considered as one of the causes underlying neuronal loss in aged people and severe neuronal decline in neurodegenerative diseases. In this review, we report on the main knowledge on ganglioside GM1, with an emphasis on the recent discoveries about its bioactive component.
Collapse
Affiliation(s)
| | | | | | | | - Sandro Sonnino
- Department of Medical Biotechnology and Translational Medicine, University of Milano, 20090 Segrate, Milano, Italy; (E.C.)
| | | |
Collapse
|
29
|
Allen SP, Hall B, Castelli LM, Francis L, Woof R, Siskos AP, Kouloura E, Gray E, Thompson AG, Talbot K, Higginbottom A, Myszczynska M, Allen CF, Stopford MJ, Hemingway J, Bauer CS, Webster CP, De Vos KJ, Turner MR, Keun HC, Hautbergue GM, Ferraiuolo L, Shaw PJ. Astrocyte adenosine deaminase loss increases motor neuron toxicity in amyotrophic lateral sclerosis. Brain 2020; 142:586-605. [PMID: 30698736 PMCID: PMC6391613 DOI: 10.1093/brain/awy353] [Citation(s) in RCA: 79] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2018] [Revised: 10/25/2018] [Accepted: 11/22/2018] [Indexed: 12/12/2022] Open
Abstract
As clinical evidence supports a negative impact of dysfunctional energy metabolism on the disease progression in amyotrophic lateral sclerosis, it is vital to understand how the energy metabolic pathways are altered and whether they can be restored to slow disease progression. Possible approaches include increasing or rerouting catabolism of alternative fuel sources to supplement the glycolytic and mitochondrial pathways such as glycogen, ketone bodies and nucleosides. To analyse the basis of the catabolic defect in amyotrophic lateral sclerosis we used a novel phenotypic metabolic array. We profiled fibroblasts and induced neuronal progenitor-derived human induced astrocytes from C9orf72 amyotrophic lateral sclerosis patients compared to normal controls, measuring the rates of production of reduced nicotinamide adenine dinucleotides from 91 potential energy substrates. This approach shows for the first time that C9orf72 human induced astrocytes and fibroblasts have an adenosine to inosine deamination defect caused by reduction of adenosine deaminase, which is also observed in induced astrocytes from sporadic patients. Patient-derived induced astrocyte lines were more susceptible to adenosine-induced toxicity, which could be mimicked by inhibiting adenosine deaminase in control lines. Furthermore, adenosine deaminase inhibition in control induced astrocytes led to increased motor neuron toxicity in co-cultures, similar to the levels observed with patient derived induced astrocytes. Bypassing metabolically the adenosine deaminase defect by inosine supplementation was beneficial bioenergetically in vitro, increasing glycolytic energy output and leading to an increase in motor neuron survival in co-cultures with induced astrocytes. Inosine supplementation, in combination with modulation of the level of adenosine deaminase may represent a beneficial therapeutic approach to evaluate in patients with amyotrophic lateral sclerosis.
Collapse
Affiliation(s)
- Scott P Allen
- Sheffield Institute for Translational Neuroscience (SITraN), University of Sheffield, 385 Glossop Road, Sheffield, UK
- Correspondence to: Dr Scott Allen Sheffield Institute for Translational Neuroscience (SITraN), University of Sheffield, 385 Glossop Road, Sheffield S10 2HQ, UK E-mail:
| | - Benjamin Hall
- Sheffield Institute for Translational Neuroscience (SITraN), University of Sheffield, 385 Glossop Road, Sheffield, UK
| | - Lydia M Castelli
- Sheffield Institute for Translational Neuroscience (SITraN), University of Sheffield, 385 Glossop Road, Sheffield, UK
| | - Laura Francis
- The Living Systems Institute, University of Exeter, Stocker Road, Exeter, UK
| | - Ryan Woof
- Sheffield Institute for Translational Neuroscience (SITraN), University of Sheffield, 385 Glossop Road, Sheffield, UK
| | - Alexandros P Siskos
- Department of Surgery and Cancer, Imperial College London, Hammersmith Hospital Campus, London, UK
| | - Eirini Kouloura
- Department of Surgery and Cancer, Imperial College London, Hammersmith Hospital Campus, London, UK
| | - Elizabeth Gray
- Nuffield Department of Clinical Neurosciences, Oxford University, John Radcliffe Hospital, West Wing Level 6, Oxford, UK
| | - Alexander G Thompson
- Nuffield Department of Clinical Neurosciences, Oxford University, John Radcliffe Hospital, West Wing Level 6, Oxford, UK
| | - Kevin Talbot
- Nuffield Department of Clinical Neurosciences, Oxford University, John Radcliffe Hospital, West Wing Level 6, Oxford, UK
| | - Adrian Higginbottom
- Sheffield Institute for Translational Neuroscience (SITraN), University of Sheffield, 385 Glossop Road, Sheffield, UK
| | - Monika Myszczynska
- Sheffield Institute for Translational Neuroscience (SITraN), University of Sheffield, 385 Glossop Road, Sheffield, UK
| | - Chloe F Allen
- Sheffield Institute for Translational Neuroscience (SITraN), University of Sheffield, 385 Glossop Road, Sheffield, UK
| | - Matthew J Stopford
- Sheffield Institute for Translational Neuroscience (SITraN), University of Sheffield, 385 Glossop Road, Sheffield, UK
| | - Jordan Hemingway
- Sheffield Institute for Translational Neuroscience (SITraN), University of Sheffield, 385 Glossop Road, Sheffield, UK
| | - Claudia S Bauer
- Sheffield Institute for Translational Neuroscience (SITraN), University of Sheffield, 385 Glossop Road, Sheffield, UK
| | - Christopher P Webster
- Sheffield Institute for Translational Neuroscience (SITraN), University of Sheffield, 385 Glossop Road, Sheffield, UK
| | - Kurt J De Vos
- Sheffield Institute for Translational Neuroscience (SITraN), University of Sheffield, 385 Glossop Road, Sheffield, UK
| | - Martin R Turner
- Nuffield Department of Clinical Neurosciences, Oxford University, John Radcliffe Hospital, West Wing Level 6, Oxford, UK
| | - Hector C Keun
- Department of Surgery and Cancer, Imperial College London, Hammersmith Hospital Campus, London, UK
| | - Guillaume M Hautbergue
- Sheffield Institute for Translational Neuroscience (SITraN), University of Sheffield, 385 Glossop Road, Sheffield, UK
| | - Laura Ferraiuolo
- Sheffield Institute for Translational Neuroscience (SITraN), University of Sheffield, 385 Glossop Road, Sheffield, UK
| | - Pamela J Shaw
- Sheffield Institute for Translational Neuroscience (SITraN), University of Sheffield, 385 Glossop Road, Sheffield, UK
| |
Collapse
|
30
|
Seven YB, Simon AK, Sajjadi E, Zwick A, Satriotomo I, Mitchell GS. Adenosine 2A receptor inhibition protects phrenic motor neurons from cell death induced by protein synthesis inhibition. Exp Neurol 2019; 323:113067. [PMID: 31629857 DOI: 10.1016/j.expneurol.2019.113067] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2019] [Revised: 09/12/2019] [Accepted: 09/18/2019] [Indexed: 01/31/2023]
Abstract
Respiratory motor neuron survival is critical for maintenance of adequate ventilation and airway clearance, preventing dependence to mechanical ventilation and respiratory tract infections. Phrenic motor neurons are highly vulnerable in rodent models of motor neuron disease versus accessory inspiratory motor pools (e.g. intercostals, scalenus). Thus, strategies that promote phrenic motor neuron survival when faced with disease and/or toxic insults are needed to help preserve breathing ability, airway defense and ventilator independence. Adenosine 2A receptors (A2A) are emerging as a potential target to promote neuroprotection, although their activation can have both beneficial and pathogenic effects. Since the role of A2A receptors in the phrenic motor neuron survival/death is not known, we tested the hypothesis that A2A receptor antagonism promotes phrenic motor neuron survival and preserves diaphragm function when faced with toxic, neurodegenerative insults that lead to phrenic motor neuron death. We utilized a novel neurotoxic model of respiratory motor neuron death recently developed in our laboratory: intrapleural injections of cholera toxin B subunit (CtB) conjugated to the ribosomal toxin, saporin (CtB-Saporin). We demonstrate that intrapleural CtB-Saporin causes: 1) profound phrenic motor neuron death (~5% survival); 2) ~7-fold increase in phrenic motor neuron A2A receptor expression prior to cell death; and 3) diaphragm muscle paralysis (inactive in most rats; ~7% residual diaphragm EMG amplitude during room air breathing). The A2A receptor antagonist istradefylline given after CtB-Saporin: 1) reduced phrenic motor neuron death (~20% survival) and 2) preserved diaphragm EMG activity (~46%). Thus, A2A receptors contribute to neurotoxic phrenic motor neuron death, an effect mitigated by A2A receptor antagonism.
Collapse
Affiliation(s)
- Yasin B Seven
- Center for Respiratory Research and Rehabilitation, Department of Physical Therapy and McKnight Brain Institute, University of Florida, Gainesville, FL 32610, USA
| | - Alec K Simon
- Center for Respiratory Research and Rehabilitation, Department of Physical Therapy and McKnight Brain Institute, University of Florida, Gainesville, FL 32610, USA
| | - Elaheh Sajjadi
- Center for Respiratory Research and Rehabilitation, Department of Physical Therapy and McKnight Brain Institute, University of Florida, Gainesville, FL 32610, USA
| | - Amanda Zwick
- Center for Respiratory Research and Rehabilitation, Department of Physical Therapy and McKnight Brain Institute, University of Florida, Gainesville, FL 32610, USA
| | - Irawan Satriotomo
- Center for Respiratory Research and Rehabilitation, Department of Physical Therapy and McKnight Brain Institute, University of Florida, Gainesville, FL 32610, USA
| | - Gordon S Mitchell
- Center for Respiratory Research and Rehabilitation, Department of Physical Therapy and McKnight Brain Institute, University of Florida, Gainesville, FL 32610, USA.
| |
Collapse
|
31
|
Seydyousefi M, Moghanlou AE, Metz GAS, Gursoy R, Faghfoori MH, Mirghani SJ, Faghfoori Z. Exogenous adenosine facilitates neuroprotection and functional recovery following cerebral ischemia in rats. Brain Res Bull 2019; 153:250-256. [PMID: 31545998 DOI: 10.1016/j.brainresbull.2019.09.010] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2019] [Revised: 09/16/2019] [Accepted: 09/18/2019] [Indexed: 12/31/2022]
Abstract
INTRODUCTION & OBJECTIVE Cerebral ischemia causes physiological and biochemical cellular changes that ultimately result in structural and functional damage to hippocampal neurons. Ischemia also raises endogenous adenosine release that in turn has neuroprotective effects. The purpose of this study was to evaluate the effect of exogenous adenosine on mitigating neuronal lesions to the CA1 region of hippocampus and A2A protein expression following cerebral I/R in rats. METHODS Male Wistar rats were randomly assigned to three experimental groups (sham, ischemia + control, and ischemia + adenosine). A daily dose of adenosine (0.1 mg/ml/kg, i.p.) was administered starting 24 h post-ischemia for 7 days. Ischemia was induced by occlusion of both common carotid arteries for 45 min. Cresyl violet and Hematoxylin Eosin staining were used to assess lesion extent and location. To investigate the expression and protein levels, immunohistochemistry and enzyme-linked immunosorbent assay method was used. RESULTS The cerebral ischemia caused neuronal loss in the CA1 region and reduced sensorimotor functions in lesion animals. Injection of adenosine significantly diminished cell death and improved sensorimotor functional recovery. Moreover, the expression and concentration of A2A protein was significantly greater in the adenosine group compared to the ischemia group. CONCLUSION This study showed that the administration of exogenous adenosine promotes protection against cell death and supports functional recovery following ischemic injury.
Collapse
Affiliation(s)
- Mehdi Seydyousefi
- Department of Physical Education and Sport Sciences, Bojnourd Branch, Islamic Azad University, Bojnourd, Iran.
| | | | - Gerlinde A S Metz
- Canadian Centre for Behavioural Neuroscience, Department of Neuroscience, University of Lethbridge, Lethbridge, Alberta, T1K 3M4, Canada.
| | - Recep Gursoy
- Faculty of Sports Sciences, Mugla Sitki Kocman University, Mugla, Turkey.
| | - Mohammad Hasan Faghfoori
- Department of Medical Biotechnology, School of Medicine, Zanjan University of Medical Sciences, Zanjan, Iran.
| | | | - Zeinab Faghfoori
- Food Safety Research Center (salt), Semnan University of Medical Sciences, Semnan, Iran; Department of Nutrition, School of Nutrition and Food Sciences, Semnan University of Medical Sciences, Semnan, Iran.
| |
Collapse
|
32
|
Lin CY, Lai HL, Chen HM, Siew JJ, Hsiao CT, Chang HC, Liao KS, Tsai SC, Wu CY, Kitajima K, Sato C, Khoo KH, Chern Y. Functional roles of ST8SIA3-mediated sialylation of striatal dopamine D 2 and adenosine A 2A receptors. Transl Psychiatry 2019; 9:209. [PMID: 31455764 PMCID: PMC6712005 DOI: 10.1038/s41398-019-0529-z] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/05/2018] [Revised: 05/22/2019] [Accepted: 06/20/2019] [Indexed: 01/20/2023] Open
Abstract
Sialic acids are typically added to the end of glycoconjugates by sialyltransferases. Among the six ST8 α-N-acetyl-neuraminide α-2,8-sialyltransferases (ST8SIA) existing in adult brains, ST8SIA2 is a schizophrenia-associated gene. However, the in vivo substrates and physiological functions of most sialyltransferases are currently unknown. The ST8SIA3 is enriched in the striatum. Here, we showed that ablation of St8sia3 in mice (St8sia3-KO) led to fewer disialylated and trisialylated terminal glycotopes in the striatum of St8sia3-KO mice. Moreover, the apparent sizes of several striatum-enriched G-protein-coupled receptors (GPCRs) (including the adenosine A2A receptor (A2AR) and dopamine D1/D2 receptors (D1R and D2R)) were smaller in St8sia3-KO mice than in WT mice. A sialidase treatment removed the differences in the sizes of these molecules between St8sia3-KO and WT mice, confirming the involvement of sialylation. Expression of ST8SIA3 in the striatum of St8sia3-KO mice using adeno-associated viruses normalized the sizes of these proteins, demonstrating a direct role of ST8SIA3. The lack of ST8SIA3-mediated sialylation altered the distribution of these proteins in lipid rafts and the interaction between D2R and A2AR. Locomotor activity assays revealed altered pharmacological responses of St8sia3-KO mice to drugs targeting these receptors and verified that a greater population of D2R formed heteromers with A2AR in the striatum of St8sia3-KO mice. Since the A2AR-D2R heteromer is an important drug target for several basal ganglia diseases (such as schizophrenia and Parkinson's disease), the present study not only reveals a crucial role for ST8SIA3 in striatal functions but also provides a new drug target for basal ganglia-related diseases.
Collapse
Affiliation(s)
- Chien-Yu Lin
- 0000 0001 2287 1366grid.28665.3fInstitute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
| | - Hsing-Lin Lai
- 0000 0001 2287 1366grid.28665.3fInstitute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
| | - Hui-Mei Chen
- 0000 0001 2287 1366grid.28665.3fInstitute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
| | - Jian-Jing Siew
- 0000 0001 2287 1366grid.28665.3fInstitute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan ,0000 0001 0425 5914grid.260770.4Taiwan International Graduate Program in Molecular Medicine, National Yang-Ming University and Academia Sinica, Taipei, Taiwan
| | - Cheng-Te Hsiao
- 0000 0001 2287 1366grid.28665.3fInstitute of Biological Chemistry, Academia Sinica, Taipei, Taiwan
| | - Hua-Chien Chang
- 0000 0001 2287 1366grid.28665.3fInstitute of Biological Chemistry, Academia Sinica, Taipei, Taiwan
| | - Kuo-Shiang Liao
- 0000 0001 2287 1366grid.28665.3fGenomics Research Center, Academia Sinica, Taipei, Taiwan
| | - Shih-Chieh Tsai
- grid.36020.37Department of Research and Development, National Laboratory Animal Center, National Applied Research Laboratories, Taipei and Tainan, Taipei, Taiwan
| | - Chung-Yi Wu
- 0000 0001 2287 1366grid.28665.3fGenomics Research Center, Academia Sinica, Taipei, Taiwan
| | - Ken Kitajima
- 0000 0001 0943 978Xgrid.27476.30Bioscience and Biotechnology Center, Nagoya University, Nagoya, 464-860 Japan
| | - Chihiro Sato
- 0000 0001 0943 978Xgrid.27476.30Bioscience and Biotechnology Center, Nagoya University, Nagoya, 464-860 Japan
| | - Kay-Hooi Khoo
- 0000 0001 2287 1366grid.28665.3fInstitute of Biological Chemistry, Academia Sinica, Taipei, Taiwan
| | - Yijuang Chern
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan.
| |
Collapse
|
33
|
Pradhan J, Noakes PG, Bellingham MC. The Role of Altered BDNF/TrkB Signaling in Amyotrophic Lateral Sclerosis. Front Cell Neurosci 2019; 13:368. [PMID: 31456666 PMCID: PMC6700252 DOI: 10.3389/fncel.2019.00368] [Citation(s) in RCA: 73] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2019] [Accepted: 07/29/2019] [Indexed: 12/11/2022] Open
Abstract
Brain derived neurotrophic factor (BDNF) is well recognized for its neuroprotective functions, via activation of its high affinity receptor, tropomysin related kinase B (TrkB). In addition, BDNF/TrkB neuroprotective functions can also be elicited indirectly via activation of adenosine 2A receptors (A2aRs), which in turn transactivates TrkB. Evidence suggests that alterations in BDNF/TrkB, including TrkB transactivation by A2aRs, can occur in several neurodegenerative diseases, including amyotrophic lateral sclerosis (ALS). Although enhancing BDNF has been a major goal for protection of dying motor neurons (MNs), this has not been successful. Indeed, there is emerging in vitro and in vivo evidence suggesting that an upregulation of BDNF/TrkB can cause detrimental effects on MNs, making them more vulnerable to pathophysiological insults. For example, in ALS, early synaptic hyper-excitability of MNs is thought to enhance BDNF-mediated signaling, thereby causing glutamate excitotoxicity, and ultimately MN death. Moreover, direct inhibition of TrkB and A2aRs has been shown to protect MNs from these pathophysiological insults, suggesting that modulation of BDNF/TrkB and/or A2aRs receptors may be important in early disease pathogenesis in ALS. This review highlights the relevance of pathophysiological actions of BDNF/TrkB under certain circumstances, so that manipulation of BDNF/TrkB and A2aRs may give rise to alternate neuroprotective therapeutic strategies in the treatment of neural diseases such as ALS.
Collapse
Affiliation(s)
- Jonu Pradhan
- Faculty of Medicine, School of Biomedical Sciences, The University of Queensland, Brisbane, QLD, Australia
| | - Peter G Noakes
- Faculty of Medicine, School of Biomedical Sciences, The University of Queensland, Brisbane, QLD, Australia.,Queensland Brain Institute, The University of Queensland, Brisbane, QLD, Australia
| | - Mark C Bellingham
- Faculty of Medicine, School of Biomedical Sciences, The University of Queensland, Brisbane, QLD, Australia
| |
Collapse
|
34
|
L3MBTL1 regulates ALS/FTD-associated proteotoxicity and quality control. Nat Neurosci 2019; 22:875-886. [PMID: 31061493 PMCID: PMC6588399 DOI: 10.1038/s41593-019-0384-5] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2018] [Accepted: 03/13/2019] [Indexed: 12/13/2022]
Abstract
Misfolded protein toxicity and failure of protein quality control
underlie neurodegenerative diseases including amyotrophic lateral sclerosis
(ALS) and frontotemporal dementia (FTD). Here, we identified Lethal(3)malignant
brain tumor-like protein 1 (L3MBTL1) as a previously unknown regulator of
protein quality control, the loss of which protected against the proteotoxicity
of mutant SOD1 or C9orf72 dipeptide repeat proteins. L3MBTL1 acts by regulating
p53-dependent quality control systems that degrade misfolded proteins. SET
domain-containing protein 8 (SETD8), a L3MBTL1-associatd p53-binding protein,
also regulated clearance of misfolded proteins and was increased by
proteotoxicity-associated stresses in mammalian cells. Both L3MBTL1 and SETD8
were up-regulated in the central nervous systems of mouse models of ALS and
human ALS/FTD patients. The role of L3MBTL1 in protein quality control is
conserved from C. elegans to mammalian neurons. These results
indicate a previously unrecognized pathway in both normal stress response and
proteotoxicity-associated neurodegenerative diseases.
Collapse
|
35
|
Cieślak M, Roszek K, Wujak M. Purinergic implication in amyotrophic lateral sclerosis-from pathological mechanisms to therapeutic perspectives. Purinergic Signal 2019; 15:1-15. [PMID: 30430356 PMCID: PMC6439052 DOI: 10.1007/s11302-018-9633-4] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2018] [Accepted: 11/01/2018] [Indexed: 12/22/2022] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a clinically heterogeneous disorder characterized by degeneration of upper motor neurons in the brainstem and lower motor neurons in the spinal cord. Multiple mechanisms of motor neuron injury have been implicated, including more than 20 different genetic factors. The pathogenesis of ALS consists of two stages: an early neuroprotective stage and a later neurotoxic. During early phases of disease progression, the immune system through glial and T cell activities provides anti-inflammatory factors that sustain motor neuron viability. As the disease progresses and motor neuron injury accelerates, a rapidly succeeding neurotoxic phase develops. A well-orchestrated purine-mediated dialog among motor neurons, surrounding glia and immune cells control the beneficial and detrimental activities occurring in the nervous system. In general, low adenosine triphosphate (ATP) concentrations protect cells against excitotoxic stimuli through purinergic P2X4 receptor, whereas high concentrations of ATP trigger toxic P2X7 receptor activation. Finally, adenosine is also involved in ALS progression since A2A receptor antagonists prevent motor neuron death. Given the complex cellular cross-talk occurring in ALS and the recognized function of extracellular nucleotides and adenosine in neuroglia communication, the comprehensive understanding of purinome dynamics might provide new research perspectives to decipher ALS and help to design more efficient and targeted drugs. This review will focus on the purinergic players involved in ALS etiology and disease progression and current therapeutic strategies to enhance neuroprotection and suppress neurotoxicity.
Collapse
Affiliation(s)
- M Cieślak
- Neurology Clinic, Marek Cieślak, Toruń, Poland
| | - K Roszek
- Department of Biochemistry, Faculty of Biology and Environmental Protection, Nicolaus Copernicus University in Toruń, 1 Lwowska St, 87-100, Toruń, Poland
| | - M Wujak
- Department of Biochemistry, Faculty of Biology and Environmental Protection, Nicolaus Copernicus University in Toruń, 1 Lwowska St, 87-100, Toruń, Poland.
| |
Collapse
|
36
|
Petimar J, O'Reilly É, Adami HO, van den Brandt PA, Buring J, English DR, Freedman DM, Giles GG, Håkansson N, Kurth T, Larsson SC, Robien K, Schouten LJ, Weiderpass E, Wolk A, Smith-Warner SA. Coffee, tea, and caffeine intake and amyotrophic lateral sclerosis mortality in a pooled analysis of eight prospective cohort studies. Eur J Neurol 2018; 26:468-475. [PMID: 30326172 DOI: 10.1111/ene.13840] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2018] [Accepted: 09/10/2018] [Indexed: 12/13/2022]
Abstract
BACKGROUND AND PURPOSE Caffeine is associated with a lower risk of some neurological diseases, but few prospective studies have investigated caffeine intake and risk of amyotrophic lateral sclerosis (ALS) mortality. We therefore determined associations between coffee, tea and caffeine intake, and risk of ALS mortality. METHODS We conducted pooled analyses of eight international, prospective cohort studies, including 351 565 individuals (120 688 men and 230 877 women). We assessed coffee, tea and caffeine intake using validated food-frequency questionnaires administered at baseline. We used Cox regression to estimate study- and sex-specific risk ratios and 95% confidence intervals (CI) for ALS mortality, which were then pooled using a random-effects model. We conducted analyses using cohort-specific tertiles, absolute common cut-points and continuous measures of all exposures. RESULTS During follow-up, 545 ALS deaths were documented. We did not observe statistically significant associations between coffee, tea or caffeine intake and risk of ALS mortality. The pooled multivariable risk ratio (MVRR) for ≥3 cups per day vs. >0 to <1 cup per day was 1.04 (95% CI, 0.74-1.47) for coffee and 1.17 (95% CI, 0.77-1.79) for tea. The pooled MVRR comparing the highest with the lowest tertile of caffeine intake (mg/day) was 0.99 (95% CI, 0.80-1.23). No statistically significant results were observed when exposures were modeled as tertiles or continuously. CONCLUSIONS Our results do not support associations between coffee, tea or total caffeine intake and risk of ALS mortality.
Collapse
Affiliation(s)
- J Petimar
- Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - É O'Reilly
- Harvard T.H. Chan School of Public Health, Boston, MA, USA.,School of Public Health, College of Medicine, University College Cork, Cork, Ireland
| | - H-O Adami
- Karolinska Institutet, Stockholm, Sweden
| | | | - J Buring
- Division of Preventive Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - D R English
- Cancer Council Victoria, Melbourne, VIC, Australia.,Melbourne School of Population and Global Health, The University of Melbourne, Melbourne, VIC, Australia
| | - D M Freedman
- National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - G G Giles
- Cancer Council Victoria, Melbourne, VIC, Australia.,Melbourne School of Population and Global Health, The University of Melbourne, Melbourne, VIC, Australia
| | | | - T Kurth
- Institute of Public Health, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | | | - K Robien
- Milken Institute School of Public Health, The George Washington University, Washington, DC, USA
| | - L J Schouten
- Caphri School, Maastricht University, Maastricht, The Netherlands
| | - E Weiderpass
- Karolinska Institutet, Stockholm, Sweden.,Institute of Population-Based Cancer Research, Oslo, Norway
| | - A Wolk
- Karolinska Institutet, Stockholm, Sweden
| | | |
Collapse
|
37
|
McGurk L, Gomes E, Guo L, Mojsilovic-Petrovic J, Tran V, Kalb RG, Shorter J, Bonini NM. Poly(ADP-Ribose) Prevents Pathological Phase Separation of TDP-43 by Promoting Liquid Demixing and Stress Granule Localization. Mol Cell 2018; 71:703-717.e9. [PMID: 30100264 PMCID: PMC6128762 DOI: 10.1016/j.molcel.2018.07.002] [Citation(s) in RCA: 321] [Impact Index Per Article: 45.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2018] [Revised: 05/18/2018] [Accepted: 06/29/2018] [Indexed: 12/27/2022]
Abstract
In amyotrophic lateral sclerosis (ALS) and frontotemporal degeneration (FTD), cytoplasmic aggregates of hyperphosphorylated TDP-43 accumulate and colocalize with some stress granule components, but how pathological TDP-43 aggregation is nucleated remains unknown. In Drosophila, we establish that downregulation of tankyrase, a poly(ADP-ribose) (PAR) polymerase, reduces TDP-43 accumulation in the cytoplasm and potently mitigates neurodegeneration. We establish that TDP-43 non-covalently binds to PAR via PAR-binding motifs embedded within its nuclear localization sequence. PAR binding promotes liquid-liquid phase separation of TDP-43 in vitro and is required for TDP-43 accumulation in stress granules in mammalian cells and neurons. Stress granule localization initially protects TDP-43 from disease-associated phosphorylation, but upon long-term stress, stress granules resolve, leaving behind aggregates of phosphorylated TDP-43. Finally, small-molecule inhibition of Tankyrase-1/2 in mammalian cells inhibits formation of cytoplasmic TDP-43 foci without affecting stress granule assembly. Thus, Tankyrase inhibition antagonizes TDP-43-associated pathology and neurodegeneration and could have therapeutic utility for ALS and FTD.
Collapse
Affiliation(s)
- Leeanne McGurk
- Department of Biology, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Edward Gomes
- Department of Biochemistry and Biophysics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Lin Guo
- Department of Biochemistry and Biophysics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Jelena Mojsilovic-Petrovic
- Department of Neurology, Children's Hospital of Philadelphia, Joseph Stokes Jr. Research Institute, Philadelphia, PA 19104, USA
| | - Van Tran
- Department of Biology, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Robert G Kalb
- Department of Neurology, Children's Hospital of Philadelphia, Joseph Stokes Jr. Research Institute, Philadelphia, PA 19104, USA
| | - James Shorter
- Department of Biochemistry and Biophysics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA.
| | - Nancy M Bonini
- Department of Biology, University of Pennsylvania, Philadelphia, PA 19104, USA.
| |
Collapse
|
38
|
McGurk L, Mojsilovic-Petrovic J, Van Deerlin VM, Shorter J, Kalb RG, Lee VM, Trojanowski JQ, Lee EB, Bonini NM. Nuclear poly(ADP-ribose) activity is a therapeutic target in amyotrophic lateral sclerosis. Acta Neuropathol Commun 2018; 6:84. [PMID: 30157956 PMCID: PMC6114235 DOI: 10.1186/s40478-018-0586-1] [Citation(s) in RCA: 85] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2018] [Accepted: 08/19/2018] [Indexed: 12/11/2022] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a devastating and fatal motor neuron disease. Diagnosis typically occurs in the fifth decade of life and the disease progresses rapidly leading to death within ~ 2–5 years of symptomatic onset. There is no cure, and the few available treatments offer only a modest extension in patient survival. A protein central to ALS is the nuclear RNA/DNA-binding protein, TDP-43. In > 95% of ALS patients, TDP-43 is cleared from the nucleus and forms phosphorylated protein aggregates in the cytoplasm of affected neurons and glia. We recently defined that poly(ADP-ribose) (PAR) activity regulates TDP-43-associated toxicity. PAR is a posttranslational modification that is attached to target proteins by PAR polymerases (PARPs). PARP-1 and PARP-2 are the major enzymes that are active in the nucleus. Here, we uncovered that the motor neurons of the ALS spinal cord were associated with elevated nuclear PAR, suggesting elevated PARP activity. Veliparib, a small-molecule inhibitor of nuclear PARP-1/2, mitigated the formation of cytoplasmic TDP-43 aggregates in mammalian cells. In primary spinal-cord cultures from rat, Veliparib also inhibited TDP-43-associated neuronal death. These studies uncover that PAR activity is misregulated in the ALS spinal cord, and a small-molecular inhibitor of PARP-1/2 activity may have therapeutic potential in the treatment of ALS and related disorders associated with abnormal TDP-43 homeostasis.
Collapse
|
39
|
Gupta P, Uner OE, Nayak S, Grant GR, Kalb RG. SAP97 regulates behavior and expression of schizophrenia risk enriched gene sets in mouse hippocampus. PLoS One 2018; 13:e0200477. [PMID: 29995933 PMCID: PMC6040763 DOI: 10.1371/journal.pone.0200477] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2018] [Accepted: 06/27/2018] [Indexed: 01/10/2023] Open
Abstract
Synapse associated protein of 97KDa (SAP97) belongs to a family of scaffolding proteins, the membrane-associated guanylate kinases (MAGUKs), that are highly enriched in the postsynaptic density of synapses and play an important role in organizing protein complexes necessary for synaptic development and plasticity. The Dlg-MAGUK family of proteins are structurally very similar, and an effort has been made to parse apart the unique function of each Dlg-MAGUK protein by characterization of knockout mice. Knockout mice have been generated and characterized for PSD-95, PSD-93, and SAP102, however SAP97 knockout mice have been impossible to study because the SAP97 null mice die soon after birth due to a craniofacial defect. We studied the transcriptomic and behavioral consequences of a brain-specific conditional knockout of SAP97 (SAP97-cKO). RNA sequencing from hippocampi from control and SAP97-cKO male animals identified 67 SAP97 regulated transcripts. As large-scale genetic studies have implicated MAGUKs in neuropsychiatric disorders such as intellectual disability, autism spectrum disorders, and schizophrenia (SCZ), we analyzed our differentially expressed gene (DEG) set for enrichment of disease risk-associated genes, and found our DEG set to be specifically enriched for SCZ-related genes. Subjecting SAP97-cKO mice to a battery of behavioral tests revealed a subtle male-specific cognitive deficit and female-specific motor deficit, while other behaviors were largely unaffected. These data suggest that loss of SAP97 may have a modest contribution to organismal behavior. The SAP97-cKO mouse serves as a stepping stone for understanding the unique role of SAP97 in biology.
Collapse
Affiliation(s)
- Preetika Gupta
- Neuroscience Graduate Group, Department of Neuroscience, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Ogul E. Uner
- School of Arts and Sciences, Department of Biology, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Soumyashant Nayak
- Institute for Translational Medicine and Therapeutics, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Gregory R. Grant
- Institute for Translational Medicine and Therapeutics, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
- Department of Genetics, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Robert G. Kalb
- Feinberg School of Medicine, Department of Neurology, Northwestern University, Chicago, Illinois, United States of America
| |
Collapse
|
40
|
Sebastião AM, Rei N, Ribeiro JA. Amyotrophic Lateral Sclerosis (ALS) and Adenosine Receptors. Front Pharmacol 2018; 9:267. [PMID: 29713276 PMCID: PMC5911503 DOI: 10.3389/fphar.2018.00267] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2017] [Accepted: 03/09/2018] [Indexed: 12/11/2022] Open
Abstract
In the present review we discuss the potential involvement of adenosinergic signaling, in particular the role of adenosine receptors, in amyotrophic lateral sclerosis (ALS). Though the literature on this topic is not abundant, the information so far available on adenosine receptors in animal models of ALS highlights the interest to continue to explore the role of these receptors in this neurodegenerative disease. Indeed, all motor neurons affected in ALS are responsive to adenosine receptor ligands but interestingly, there are alterations in pre-symptomatic or early symptomatic stages that mirror those in advanced disease stages. Information starts to emerge pointing toward a beneficial role of A2A receptors (A2AR), most probably at early disease states, and a detrimental role of caffeine, in clear contrast with what occurs in other neurodegenerative diseases. However, some evidence also exists on a beneficial action of A2AR antagonists. It may happen that there are time windows where A2AR prove beneficial and others where their blockade is required. Furthermore, the same changes may not occur simultaneously at the different synapses. In line with this, it is not fully understood if ALS is a dying back disease or if it propagates in a centrifugal way. It thus seems crucial to understand how motor neuron dysfunction occurs, how adenosine receptors are involved in those dysfunctions and whether the early changes in purinergic signaling are compensatory or triggers for the disease. Getting this information is crucial before starting the design of purinergic based strategies to halt or delay disease progression.
Collapse
Affiliation(s)
- Ana M Sebastião
- Instituto de Farmacologia e Neurociências, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal.,Instituto de Medicina Molecular, Universidade de Lisboa, Lisbon, Portugal
| | - Nádia Rei
- Instituto de Farmacologia e Neurociências, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal.,Instituto de Medicina Molecular, Universidade de Lisboa, Lisbon, Portugal
| | - Joaquim A Ribeiro
- Instituto de Farmacologia e Neurociências, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal.,Instituto de Medicina Molecular, Universidade de Lisboa, Lisbon, Portugal
| |
Collapse
|
41
|
Oliveira-Giacomelli Á, Naaldijk Y, Sardá-Arroyo L, Gonçalves MCB, Corrêa-Velloso J, Pillat MM, de Souza HDN, Ulrich H. Purinergic Receptors in Neurological Diseases With Motor Symptoms: Targets for Therapy. Front Pharmacol 2018; 9:325. [PMID: 29692728 PMCID: PMC5902708 DOI: 10.3389/fphar.2018.00325] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2017] [Accepted: 03/21/2018] [Indexed: 12/13/2022] Open
Abstract
Since proving adenosine triphosphate (ATP) functions as a neurotransmitter in neuron/glia interactions, the purinergic system has been more intensely studied within the scope of the central nervous system. In neurological disorders with associated motor symptoms, including Parkinson's disease (PD), motor neuron diseases (MND), multiple sclerosis (MS), amyotrophic lateral sclerosis (ALS), Huntington's Disease (HD), restless leg syndrome (RLS), and ataxias, alterations in purinergic receptor expression and activity have been noted, indicating a potential role for this system in disease etiology and progression. In neurodegenerative conditions, neural cell death provokes extensive ATP release and alters calcium signaling through purinergic receptor modulation. Consequently, neuroinflammatory responses, excitotoxicity and apoptosis are directly or indirectly induced. This review analyzes currently available data, which suggests involvement of the purinergic system in neuro-associated motor dysfunctions and underlying mechanisms. Possible targets for pharmacological interventions are also discussed.
Collapse
Affiliation(s)
| | - Yahaira Naaldijk
- Department of Biochemistry, Institute of Chemistry, University of São Paulo, São Paulo, Brazil
| | - Laura Sardá-Arroyo
- Department of Biochemistry, Institute of Chemistry, University of São Paulo, São Paulo, Brazil
| | - Maria C. B. Gonçalves
- Department of Neurology and Neuroscience, Medical School, Federal University of São Paulo, São Paulo, Brazil
| | - Juliana Corrêa-Velloso
- Department of Biochemistry, Institute of Chemistry, University of São Paulo, São Paulo, Brazil
| | - Micheli M. Pillat
- Department of Biochemistry, Institute of Chemistry, University of São Paulo, São Paulo, Brazil
| | - Héllio D. N. de Souza
- Department of Biochemistry, Institute of Chemistry, University of São Paulo, São Paulo, Brazil
| | - Henning Ulrich
- Department of Biochemistry, Institute of Chemistry, University of São Paulo, São Paulo, Brazil
| |
Collapse
|
42
|
Sonnino S, Chiricozzi E, Grassi S, Mauri L, Prioni S, Prinetti A. Gangliosides in Membrane Organization. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2018; 156:83-120. [PMID: 29747825 DOI: 10.1016/bs.pmbts.2017.12.007] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Since the structure of GM1 was elucidated 55years ago, researchers have been attracted by the sialylated glycans of gangliosides. Gangliosides head groups, protruding toward the extracellular space, significantly contribute to the cell glycocalyx; and in certain cells, such as neurons, are major determinants of the features of the cell surface. Expression of glycosyltransferases involved in the de novo biosynthesis of gangliosides is tightly regulated along cell differentiation and activation, and is regarded as the main metabolic mechanism responsible for the acquisition of cell-specific ganglioside patterns. The resulting sialooligosaccharides are characterized by a high degree of geometrical complexity and by highly dynamic properties, which seem to be functional for complex interactions with other molecules sitting on the same cellular membrane (cis-interactions) or soluble molecules present in the extracellular environment, or molecules associated with the surface of other cells (trans-interactions). There is no doubt that the multifaceted biological functions of gangliosides are largely dependent on oligosaccharide-mediated molecular interactions. However, gangliosides are amphipathic membrane lipids, and their chemicophysical, aggregational, and, consequently, biological properties are dictated by the properties of the monomers as a whole, which are not merely dependent on the structures of their polar head groups. In this chapter, we would like to focus on the peculiar chemicophysical features of gangliosides (in particular, those of the nervous system), that represent an important driving force determining the organization and properties of cellular membranes, and to emphasize the causal connections between altered ganglioside-dependent membrane organization and relevant pathological conditions.
Collapse
|
43
|
Vejux A, Namsi A, Nury T, Moreau T, Lizard G. Biomarkers of Amyotrophic Lateral Sclerosis: Current Status and Interest of Oxysterols and Phytosterols. Front Mol Neurosci 2018; 11:12. [PMID: 29445325 PMCID: PMC5797798 DOI: 10.3389/fnmol.2018.00012] [Citation(s) in RCA: 50] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2017] [Accepted: 01/09/2018] [Indexed: 12/11/2022] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a non-demyelinating neurodegenerative disease in adults with motor disorders. Two forms exist: a sporadic form (90% of cases) and a family form due to mutations in more than 20 genes including the Superoxide dismutase 1, TAR DNA Binding Protein, Fused in Sarcoma, chromosome 9 open reading frame 72 and VAPB genes. The mechanisms associated with this pathology are beginning to be known: oxidative stress, glutamate excitotoxicity, protein aggregation, reticulum endoplasmic stress, neuroinflammation, alteration of RNA metabolism. In various neurodegenerative diseases, such as Alzheimer's disease or multiple sclerosis, the involvement of lipids is increasingly suggested based on lipid metabolism modifications. With regard to ALS, research has also focused on the possible involvement of lipids. Lipid involvement was suggested for clinical arguments where changes in cholesterol and LDL/HDL levels were reported with, however, differences in positivity between studies. Since lipids are involved in the membrane structure and certain signaling pathways, it may be considered to look for oxysterols, mainly 25-hydroxycholesterol and its metabolites involved in immune response, or phytosterols to find suitable biomarkers for this pathology.
Collapse
Affiliation(s)
- Anne Vejux
- Team Biochemistry of the Peroxisome, Inflammation and Lipid Metabolism EA 7270, INSERM, University of Bourgogne Franche-Comté, Dijon, France
| | - Amira Namsi
- Team Biochemistry of the Peroxisome, Inflammation and Lipid Metabolism EA 7270, INSERM, University of Bourgogne Franche-Comté, Dijon, France.,Laboratoire de Neurophysiologie Fonctionnelle et Pathologies, UR11ES/09, Faculté des Sciences Mathématiques, Physiques et Naturelles de Tunis, Université de Tunis El Manar - Bienvenue, Tunis, Tunisia
| | - Thomas Nury
- Team Biochemistry of the Peroxisome, Inflammation and Lipid Metabolism EA 7270, INSERM, University of Bourgogne Franche-Comté, Dijon, France
| | - Thibault Moreau
- Team Biochemistry of the Peroxisome, Inflammation and Lipid Metabolism EA 7270, INSERM, University of Bourgogne Franche-Comté, Dijon, France.,Department of Neurology, University Hospital/University Bourgogne Franche-Comté, Dijon, France
| | - Gérard Lizard
- Team Biochemistry of the Peroxisome, Inflammation and Lipid Metabolism EA 7270, INSERM, University of Bourgogne Franche-Comté, Dijon, France
| |
Collapse
|
44
|
Bahreyni A, Samani SS, Khazaei M, Ryzhikov M, Avan A, Hassanian SM. Therapeutic potentials of adenosine receptors agonists and antagonists in colitis; Current status and perspectives. J Cell Physiol 2017; 233:2733-2740. [DOI: 10.1002/jcp.26073] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2017] [Accepted: 06/28/2017] [Indexed: 12/15/2022]
Affiliation(s)
- Amirhossein Bahreyni
- Faculty of Medicine; Department of Clinical Biochemistry and Immunogenetic Research Center; Mazandaran University of Medical Sciences; Sari Mazandaran Iran
| | - Seyed S. Samani
- Department of Biology; Mashhad Branch; Islamic Azad University; Mashhad Iran
| | - Majid Khazaei
- Faculty of Medicine; Department of Medical Physiology; Mashhad University of Medical Sciences; Mashhad Iran
| | - Mikhail Ryzhikov
- Department of Molecular Microbiology and Immunology; St. Louis University; School of Medicine; Saint Louis Missouri
| | - Amir Avan
- Metabolic Syndrome Research Center; Mashhad University of Medical Sciences; Mashhad Iran
- Department of Modern Sciences and Technologies; School of Medicine; Mashhad University of Medical Sciences; Mashhad Iran
| | - Seyed M. Hassanian
- Metabolic Syndrome Research Center; Mashhad University of Medical Sciences; Mashhad Iran
- Faculty of Medicine; Department of Medical Biochemistry; Mashhad University of Medical Sciences; Mashhad Iran
- Microanatomy Research Center; Mashhad University of Medical Sciences; Mashhad Iran
| |
Collapse
|
45
|
Leiva A, Guzmán-Gutiérrez E, Contreras-Duarte S, Fuenzalida B, Cantin C, Carvajal L, Salsoso R, Gutiérrez J, Pardo F, Sobrevia L. Adenosine receptors: Modulators of lipid availability that are controlled by lipid levels. Mol Aspects Med 2017; 55:26-44. [DOI: 10.1016/j.mam.2017.01.007] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2016] [Revised: 01/25/2017] [Accepted: 01/25/2017] [Indexed: 12/20/2022]
|
46
|
Imamura K, Izumi Y, Watanabe A, Tsukita K, Woltjen K, Yamamoto T, Hotta A, Kondo T, Kitaoka S, Ohta A, Tanaka A, Watanabe D, Morita M, Takuma H, Tamaoka A, Kunath T, Wray S, Furuya H, Era T, Makioka K, Okamoto K, Fujisawa T, Nishitoh H, Homma K, Ichijo H, Julien JP, Obata N, Hosokawa M, Akiyama H, Kaneko S, Ayaki T, Ito H, Kaji R, Takahashi R, Yamanaka S, Inoue H. The Src/c-Abl pathway is a potential therapeutic target in amyotrophic lateral sclerosis. Sci Transl Med 2017; 9:eaaf3962. [PMID: 28539470 DOI: 10.1126/scitranslmed.aaf3962] [Citation(s) in RCA: 165] [Impact Index Per Article: 20.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2015] [Revised: 02/04/2016] [Accepted: 12/13/2016] [Indexed: 03/07/2024]
Abstract
Amyotrophic lateral sclerosis (ALS), a fatal disease causing progressive loss of motor neurons, still has no effective treatment. We developed a phenotypic screen to repurpose existing drugs using ALS motor neuron survival as readout. Motor neurons were generated from induced pluripotent stem cells (iPSCs) derived from an ALS patient with a mutation in superoxide dismutase 1 (SOD1). Results of the screen showed that more than half of the hits targeted the Src/c-Abl signaling pathway. Src/c-Abl inhibitors increased survival of ALS iPSC-derived motor neurons in vitro. Knockdown of Src or c-Abl with small interfering RNAs (siRNAs) also rescued ALS motor neuron degeneration. One of the hits, bosutinib, boosted autophagy, reduced the amount of misfolded mutant SOD1 protein, and attenuated altered expression of mitochondrial genes. Bosutinib also increased survival in vitro of ALS iPSC-derived motor neurons from patients with sporadic ALS or other forms of familial ALS caused by mutations in TAR DNA binding protein (TDP-43) or repeat expansions in C9orf72 Furthermore, bosutinib treatment modestly extended survival of a mouse model of ALS with an SOD1 mutation, suggesting that Src/c-Abl may be a potentially useful target for developing new drugs to treat ALS.
Collapse
Affiliation(s)
- Keiko Imamura
- Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto 606-8507, Japan
| | - Yuishin Izumi
- Department of Clinical Neuroscience, Institute of Biomedical Sciences, Tokushima University Graduate School, Tokushima 770-8503, Japan
| | - Akira Watanabe
- Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto 606-8507, Japan
| | - Kayoko Tsukita
- Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto 606-8507, Japan
| | - Knut Woltjen
- Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto 606-8507, Japan
- Hakubi Center for Advanced Research, Kyoto University, Kyoto 606-8501, Japan
| | - Takuya Yamamoto
- Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto 606-8507, Japan
- Institute for Integrated Cell-Material Sciences, Kyoto University, Kyoto 606-8507, Japan
| | - Akitsu Hotta
- Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto 606-8507, Japan
- Institute for Integrated Cell-Material Sciences, Kyoto University, Kyoto 606-8507, Japan
- Precursory Research for Embryonic Science and Technology (PRESTO), Japan Science and Technology Agency, Saitama 332-0012, Japan
| | - Takayuki Kondo
- Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto 606-8507, Japan
| | - Shiho Kitaoka
- Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto 606-8507, Japan
| | - Akira Ohta
- Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto 606-8507, Japan
| | - Akito Tanaka
- Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto 606-8507, Japan
| | - Dai Watanabe
- Department of Biological Sciences, Graduate School of Medicine, Kyoto University, Kyoto 606-8501, Japan
| | - Mitsuya Morita
- Division of Neurology, Department of Internal Medicine, Jichi Medical University, Tochigi 329-0498, Japan
| | - Hiroshi Takuma
- Department of Neurology, Faculty of Medicine, University of Tsukuba, Tsukuba 305-8575, Japan
| | - Akira Tamaoka
- Department of Neurology, Faculty of Medicine, University of Tsukuba, Tsukuba 305-8575, Japan
| | - Tilo Kunath
- MRC Centre for Regenerative Medicine, School of Biological Sciences, University of Edinburgh, 5 Little France Drive, Edinburgh EH16 4UU, UK
| | - Selina Wray
- Department of Molecular Neuroscience, University College London Institute of Neurology, Queen Square, London WC1N 3BG, UK
| | - Hirokazu Furuya
- Department of Neurology, Kochi Medical School, Kochi University, Kochi 783-8505, Japan
| | - Takumi Era
- Department of Cell Modulation, Institute of Molecular Embryology and Genetics, Kumamoto University, Kumamoto 860-0811, Japan
| | - Kouki Makioka
- Department of Neurology, Gunma University Graduate School of Medicine, Maebashi 371-8511, Japan
| | - Koichi Okamoto
- Geriatrics Research Institute and Hospital, Maebashi 371-0847, Japan
| | - Takao Fujisawa
- Cell Signaling, Graduate School of Pharmaceutical Sciences, University of Tokyo, Bunkyo-ku, Tokyo 113-0033, Japan
| | - Hideki Nishitoh
- Department of Medical Sciences, University of Miyazaki, Miyazaki 889-1601, Japan
| | - Kengo Homma
- Cell Signaling, Graduate School of Pharmaceutical Sciences, University of Tokyo, Bunkyo-ku, Tokyo 113-0033, Japan
| | - Hidenori Ichijo
- Cell Signaling, Graduate School of Pharmaceutical Sciences, University of Tokyo, Bunkyo-ku, Tokyo 113-0033, Japan
| | - Jean-Pierre Julien
- Department of Psychiatry and Neurosciences, Research Centre of Mental Health Institute of Quebec (IUSMQ), Laval University, Québec, Canada
| | - Nanako Obata
- Tokyo Metropolitan Institute of Medical Science, 2-1-6 Kamikitazawa, Setagaya-ku, Tokyo 156-8506, Japan
| | - Masato Hosokawa
- Tokyo Metropolitan Institute of Medical Science, 2-1-6 Kamikitazawa, Setagaya-ku, Tokyo 156-8506, Japan
| | - Haruhiko Akiyama
- Tokyo Metropolitan Institute of Medical Science, 2-1-6 Kamikitazawa, Setagaya-ku, Tokyo 156-8506, Japan
| | - Satoshi Kaneko
- Department of Neurology, Kansai Medical University, Hirakata 573-1191, Japan
| | - Takashi Ayaki
- Department of Neurology, Graduate School of Medicine, Kyoto University, Kyoto 606-8507, Japan
| | - Hidefumi Ito
- Department of Neurology, Wakayama Medical University, Kimiidera, Wakayama 641-8509, Japan
| | - Ryuji Kaji
- Institute for Integrated Cell-Material Sciences, Kyoto University, Kyoto 606-8507, Japan
| | - Ryosuke Takahashi
- Department of Neurology, Graduate School of Medicine, Kyoto University, Kyoto 606-8507, Japan
| | - Shinya Yamanaka
- Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto 606-8507, Japan
- Gladstone Institute of Cardiovascular Disease, San Francisco, CA 94158, USA
| | - Haruhisa Inoue
- Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto 606-8507, Japan.
| |
Collapse
|
47
|
Schmidt J, Ferk P. Safety issues of compounds acting on adenosinergic signalling. ACTA ACUST UNITED AC 2017; 69:790-806. [PMID: 28397249 DOI: 10.1111/jphp.12720] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2016] [Accepted: 03/04/2017] [Indexed: 12/20/2022]
Abstract
OBJECTIVES Much research has been performed on the field of identifying the roles of adenosine and adenosinergic signalling, but a relatively low number of marketing authorizations have been granted for adenosine receptor (AdR) ligands. In part, this could be related to their safety issues; therefore, our aim was to examine the toxicological and adverse effects data of different compounds acting on adenosinergic signalling, including different AdR ligands and compounds resembling the structure of adenosine. We also wanted to present recent pharmaceutical developments of experimental compounds that showed promising results in clinical trial setting. KEY FINDINGS Safety issues of compounds modulating adenosinergic signalling were investigated, and different mechanisms were presented. Structurally different classes of compounds act on AdRs, the most important being adenosine, adenosine derivatives and other non-nucleoside compounds. Many of them are either not selective enough or are targeting other targets of adenosinergic signalling such as metabolizing enzymes that regulate adenosine levels. Many other targets are also involved that are not part of adenosinergic signalling system such as GABA receptors, different channels, enzymes and others. Some synthetic AdR ligands even showed to be genotoxic. SUMMARY Current review presents safety data of adenosine, adenosine derivatives and other non-nucleoside compounds that modulate adenosinergic signalling. We have presented different mechanisms that participate to an adverse effect or toxic outcome. A separate section also deals with possible organ-specific toxic effects on different in-vitro and in-vivo models.
Collapse
Affiliation(s)
- Jan Schmidt
- Faculty of Chemistry and Chemical Engineering, University of Maribor, Maribor, Slovenia.,Faculty of Medicine, University of Maribor, Maribor, Slovenia
| | - Polonca Ferk
- Faculty of Medicine, University of Maribor, Maribor, Slovenia
| |
Collapse
|
48
|
The Proline/Arginine Dipeptide from Hexanucleotide Repeat Expanded C9ORF72 Inhibits the Proteasome. eNeuro 2017; 4:eN-NWR-0249-16. [PMID: 28197542 PMCID: PMC5282547 DOI: 10.1523/eneuro.0249-16.2017] [Citation(s) in RCA: 52] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2016] [Revised: 01/05/2017] [Accepted: 01/12/2017] [Indexed: 12/11/2022] Open
Abstract
An intronic hexanucleotide repeat expansion (HRE) mutation in the C9ORF72 gene is the most common cause of familial ALS and frontotemporal dementia (FTD) and is found in ∼7% of individuals with apparently sporadic disease. Several different diamino acid peptides can be generated from the HRE by noncanonical translation (repeat-associated non-ATG translation, or RAN translation), and some of these peptides can be toxic. Here, we studied the effects of two arginine containing RAN translation products [proline/arginine repeated 20 times (PR20) and glycine/arginine repeated 20 times (GR20)] in primary rat spinal cord neuron cultures grown on an astrocyte feeder layer. We find that PR20 kills motor neurons with an LD50 of 2 µM, but in contrast to the effects of other ALS-causing mutant proteins (i.e., SOD or TDP43), PR20 does not evoke the biochemical signature of mitochondrial dysfunction, ER stress, or mTORC down-regulation. PR20 does result in a time-dependent build-up of ubiquitylated substrates, and this is associated with a reduction of flux through both autophagic and proteasomal degradation pathways. GR20, however, does not have these effects. The effects of PR20 on the proteasome are likely to be direct because (1) PR20 physically associates with proteasomes in biochemical assays, and (2) PR20 inhibits the degradation of a ubiquitylated test substrate when presented to purified proteasomes. Application of a proteasomal activator (IU1) blocks the toxic effects of PR20 on motor neuron survival. This work suggests that proteasomal activators have therapeutic potential in individuals with C9ORF72 HRE.
Collapse
|
49
|
Cunha RA. How does adenosine control neuronal dysfunction and neurodegeneration? J Neurochem 2016; 139:1019-1055. [PMID: 27365148 DOI: 10.1111/jnc.13724] [Citation(s) in RCA: 350] [Impact Index Per Article: 38.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2016] [Revised: 05/23/2016] [Accepted: 06/23/2016] [Indexed: 12/11/2022]
Abstract
The adenosine modulation system mostly operates through inhibitory A1 (A1 R) and facilitatory A2A receptors (A2A R) in the brain. The activity-dependent release of adenosine acts as a brake of excitatory transmission through A1 R, which are enriched in glutamatergic terminals. Adenosine sharpens salience of information encoding in neuronal circuits: high-frequency stimulation triggers ATP release in the 'activated' synapse, which is locally converted by ecto-nucleotidases into adenosine to selectively activate A2A R; A2A R switch off A1 R and CB1 receptors, bolster glutamate release and NMDA receptors to assist increasing synaptic plasticity in the 'activated' synapse; the parallel engagement of the astrocytic syncytium releases adenosine further inhibiting neighboring synapses, thus sharpening the encoded plastic change. Brain insults trigger a large outflow of adenosine and ATP, as a danger signal. A1 R are a hurdle for damage initiation, but they desensitize upon prolonged activation. However, if the insult is near-threshold and/or of short-duration, A1 R trigger preconditioning, which may limit the spread of damage. Brain insults also up-regulate A2A R, probably to bolster adaptive changes, but this heightens brain damage since A2A R blockade affords neuroprotection in models of epilepsy, depression, Alzheimer's, or Parkinson's disease. This initially involves a control of synaptotoxicity by neuronal A2A R, whereas astrocytic and microglia A2A R might control the spread of damage. The A2A R signaling mechanisms are largely unknown since A2A R are pleiotropic, coupling to different G proteins and non-canonical pathways to control the viability of glutamatergic synapses, neuroinflammation, mitochondria function, and cytoskeleton dynamics. Thus, simultaneously bolstering A1 R preconditioning and preventing excessive A2A R function might afford maximal neuroprotection. The main physiological role of the adenosine modulation system is to sharp the salience of information encoding through a combined action of adenosine A2A receptors (A2A R) in the synapse undergoing an alteration of synaptic efficiency with an increased inhibitory action of A1 R in all surrounding synapses. Brain insults trigger an up-regulation of A2A R in an attempt to bolster adaptive plasticity together with adenosine release and A1 R desensitization; this favors synaptotocity (increased A2A R) and decreases the hurdle to undergo degeneration (decreased A1 R). Maximal neuroprotection is expected to result from a combined A2A R blockade and increased A1 R activation. This article is part of a mini review series: "Synaptic Function and Dysfunction in Brain Diseases".
Collapse
Affiliation(s)
- Rodrigo A Cunha
- CNC-Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal.,FMUC-Faculty of Medicine, University of Coimbra, Coimbra, Portugal
| |
Collapse
|
50
|
Loss of RAD-23 Protects Against Models of Motor Neuron Disease by Enhancing Mutant Protein Clearance. J Neurosci 2016; 35:14286-306. [PMID: 26490867 DOI: 10.1523/jneurosci.0642-15.2015] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
UNLABELLED Misfolded proteins accumulate and aggregate in neurodegenerative disease. The existence of these deposits reflects a derangement in the protein homeostasis machinery. Using a candidate gene screen, we report that loss of RAD-23 protects against the toxicity of proteins known to aggregate in amyotrophic lateral sclerosis. Loss of RAD-23 suppresses the locomotor deficit of Caenorhabditis elegans engineered to express mutTDP-43 or mutSOD1 and also protects against aging and proteotoxic insults. Knockdown of RAD-23 is further neuroprotective against the toxicity of SOD1 and TDP-43 expression in mammalian neurons. Biochemical investigation indicates that RAD-23 modifies mutTDP-43 and mutSOD1 abundance, solubility, and turnover in association with altering the ubiquitination status of these substrates. In human amyotrophic lateral sclerosis spinal cord, we find that RAD-23 abundance is increased and RAD-23 is mislocalized within motor neurons. We propose a novel pathophysiological function for RAD-23 in the stabilization of mutated proteins that cause neurodegeneration. SIGNIFICANCE STATEMENT In this work, we identify RAD-23, a component of the protein homeostasis network and nucleotide excision repair pathway, as a modifier of the toxicity of two disease-causing, misfolding-prone proteins, SOD1 and TDP-43. Reducing the abundance of RAD-23 accelerates the degradation of mutant SOD1 and TDP-43 and reduces the cellular content of the toxic species. The existence of endogenous proteins that act as "anti-chaperones" uncovers new and general targets for therapeutic intervention.
Collapse
|