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
|
Miranda-Lourenço C, Rosa J, Rei N, Belo RF, Lopes AL, Silva D, Vieira C, Magalhães-Cardoso T, Viais R, Correia-de-Sá P, Sebastião AM, Diógenes MJ. Adenosinergic System and BDNF Signaling Changes as a Cross-Sectional Feature of RTT: Characterization of Mecp2 Heterozygous Mouse Females. Int J Mol Sci 2023; 24:16249. [PMID: 38003438 PMCID: PMC10671708 DOI: 10.3390/ijms242216249] [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: 09/23/2023] [Revised: 10/25/2023] [Accepted: 11/01/2023] [Indexed: 11/26/2023] Open
Abstract
Rett Syndrome is an X-linked neurodevelopmental disorder (RTT; OMIM#312750) associated to MECP2 mutations. MeCP2 dysfunction is seen as one cause for the deficiencies found in brain-derived neurotrophic factor (BDNF) signaling, since BDNF is one of the genes under MeCP2 jurisdiction. BDNF signaling is also dependent on the proper function of the adenosinergic system. Indeed, both BDNF signaling and the adenosinergic system are altered in Mecp2-null mice (Mecp2-/y), a representative model of severe manifestation of RTT. Considering that symptoms severity largely differs among RTT patients, we set out to investigate the BDNF and ADO signaling modifications in Mecp2 heterozygous female mice (Mecp2+/-) presenting a less severe phenotype. Symptomatic Mecp2+/- mice have lower BDNF levels in the cortex and hippocampus. This is accompanied by a loss of BDNF-induced facilitation of hippocampal long-term potentiation (LTP), which could be restored upon selective activation of adenosine A2A receptors (A2AR). While no differences were observed in the amount of adenosine in the cortex and hippocampus of Mecp2+/- mice compared with healthy littermates, the density of the A1R and A2AR subtype receptors was, respectively, upregulated and downregulated in the hippocampus. Data suggest that significant changes in BDNF and adenosine signaling pathways are present in an RTT model with a milder disease phenotype: Mecp2+/- female animals. These features strengthen the theory that boosting adenosinergic activity may be a valid therapeutic strategy for RTT patients, regardless of their genetic penetrance.
Collapse
Affiliation(s)
- Catarina Miranda-Lourenço
- Instituto de Farmacologia e Neurociências, Faculdade de Medicina, Universidade de Lisboa, 1649-028 Lisboa, Portugal; (C.M.-L.); (A.M.S.)
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, 1649-028 Lisboa, Portugal
| | - Jéssica Rosa
- Instituto de Farmacologia e Neurociências, Faculdade de Medicina, Universidade de Lisboa, 1649-028 Lisboa, Portugal; (C.M.-L.); (A.M.S.)
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, 1649-028 Lisboa, Portugal
| | - Nádia Rei
- Instituto de Farmacologia e Neurociências, Faculdade de Medicina, Universidade de Lisboa, 1649-028 Lisboa, Portugal; (C.M.-L.); (A.M.S.)
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, 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; (C.M.-L.); (A.M.S.)
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, 1649-028 Lisboa, Portugal
| | - Ana Luísa Lopes
- Laboratório de Farmacologia e Neurobiologia/MedInUP, Instituto de Ciências Biomédicas Abel Salazar—Universidade do Porto (ICBAS-UP), 4050-313 Porto, Portugal (D.S.); (P.C.-d.-S.)
| | - Diogo Silva
- Laboratório de Farmacologia e Neurobiologia/MedInUP, Instituto de Ciências Biomédicas Abel Salazar—Universidade do Porto (ICBAS-UP), 4050-313 Porto, Portugal (D.S.); (P.C.-d.-S.)
| | - Cátia Vieira
- Laboratório de Farmacologia e Neurobiologia/MedInUP, Instituto de Ciências Biomédicas Abel Salazar—Universidade do Porto (ICBAS-UP), 4050-313 Porto, Portugal (D.S.); (P.C.-d.-S.)
| | - Teresa Magalhães-Cardoso
- Laboratório de Farmacologia e Neurobiologia/MedInUP, Instituto de Ciências Biomédicas Abel Salazar—Universidade do Porto (ICBAS-UP), 4050-313 Porto, Portugal (D.S.); (P.C.-d.-S.)
| | - Ricardo Viais
- Instituto de Farmacologia e Neurociências, Faculdade de Medicina, Universidade de Lisboa, 1649-028 Lisboa, Portugal; (C.M.-L.); (A.M.S.)
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, 1649-028 Lisboa, Portugal
| | - Paulo Correia-de-Sá
- Laboratório de Farmacologia e Neurobiologia/MedInUP, Instituto de Ciências Biomédicas Abel Salazar—Universidade do Porto (ICBAS-UP), 4050-313 Porto, Portugal (D.S.); (P.C.-d.-S.)
| | - Ana M. Sebastião
- Instituto de Farmacologia e Neurociências, Faculdade de Medicina, Universidade de Lisboa, 1649-028 Lisboa, Portugal; (C.M.-L.); (A.M.S.)
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, 1649-028 Lisboa, Portugal
| | - Maria J. Diógenes
- Instituto de Farmacologia e Neurociências, Faculdade de Medicina, Universidade de Lisboa, 1649-028 Lisboa, Portugal; (C.M.-L.); (A.M.S.)
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, 1649-028 Lisboa, Portugal
| |
Collapse
|
3
|
Agosto-Marlin IM, Nikodemova M, Dale EA, Mitchell GS. BDNF-induced phrenic motor facilitation shifts from PKCθ to ERK dependence with mild systemic inflammation. J Neurophysiol 2023; 129:455-464. [PMID: 36695529 PMCID: PMC9942899 DOI: 10.1152/jn.00345.2022] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2022] [Revised: 01/17/2023] [Accepted: 01/19/2023] [Indexed: 01/26/2023] Open
Abstract
Moderate acute intermittent hypoxia (mAIH) elicits a form of phrenic motor plasticity known as phrenic long-term facilitation (pLTF), which requires spinal 5-HT2 receptor activation, ERK/MAP kinase signaling, and new brain-derived neurotrophic factor (BDNF) synthesis. New BDNF protein activates TrkB receptors that normally signal through PKCθ to elicit pLTF. Phrenic motor plasticity elicited by spinal drug administration (e.g., BDNF) is referred to by a more general term: phrenic motor facilitation (pMF). Although mild systemic inflammation elicited by a low lipopolysaccharide (LPS) dose (100 µg/kg; 24 h prior) undermines mAIH-induced pLTF upstream from BDNF protein synthesis, it augments pMF induced by spinal BDNF administration through unknown mechanisms. Here, we tested the hypothesis that mild inflammation shifts BDNF/TrkB signaling from PKCθ to alternative pathways that enhance pMF. We examined the role of three known signaling pathways associated with TrkB (MEK/ERK MAP kinase, PI3 kinase/Akt, and PKCθ) in BDNF-induced pMF in anesthetized, paralyzed, and ventilated Sprague Dawley rats 24 h post-LPS. Spinal PKCθ inhibitor (TIP) attenuated early BDNF-induced pMF (≤30 min), with minimal effect 60-90 min post-BDNF injection. In contrast, MEK inhibition (U0126) abolished BDNF-induced pMF at 60 and 90 min. PI3K/Akt inhibition (PI-828) had no effect on BDNF-induced pMF at any time. Thus, whereas BDNF-induced pMF is exclusively PKCθ-dependent in normal rats, MEK/ERK is recruited by neuroinflammation to sustain, and even augment downstream plasticity. Because AIH is being developed as a therapeutic modality to restore breathing in people living with multiple neurological disorders, it is important to understand how inflammation, a common comorbidity in many traumatic or degenerative central nervous system disorders, impacts phrenic motor plasticity.NEW & NOTEWORTHY We demonstrate that even mild systemic inflammation shifts signaling mechanisms giving rise to BDNF-induced phrenic motor plasticity. This finding has important experimental, biological, and translational implications, particularly since BDNF-dependent spinal plasticity is being translated to restore breathing and nonrespiratory movements in diverse clinical disorders, such as spinal cord injury (SCI) and amyotrophic lateral sclerosis (ALS).
Collapse
Affiliation(s)
- Ibis M Agosto-Marlin
- Department of Comparative Biosciences, University of Wisconsin, Madison, Wisconsin, United States
| | - Maria Nikodemova
- Breathing Research and Therapeutics Center, University of Florida, Gainesville, Florida, United States
- Department of Physical, Therapy University of Florida, Gainesville, Florida, United States
- McKnight Brain Institute, University of Florida, Gainesville, Florida, United States
| | - Erica A Dale
- Breathing Research and Therapeutics Center, University of Florida, Gainesville, Florida, United States
- Department of Physiology and Aging, University of Florida, Gainesville, Florida, United States
- McKnight Brain Institute, University of Florida, Gainesville, Florida, United States
| | - Gordon S Mitchell
- Department of Comparative Biosciences, University of Wisconsin, Madison, Wisconsin, United States
- Breathing Research and Therapeutics Center, University of Florida, Gainesville, Florida, United States
- Department of Physical, Therapy University of Florida, Gainesville, Florida, United States
- McKnight Brain Institute, University of Florida, Gainesville, Florida, United States
| |
Collapse
|
4
|
c-Abl Tyrosine Kinase Is Required for BDNF-Induced Dendritic Branching and Growth. Int J Mol Sci 2023; 24:ijms24031944. [PMID: 36768268 PMCID: PMC9916151 DOI: 10.3390/ijms24031944] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2022] [Revised: 01/09/2023] [Accepted: 01/09/2023] [Indexed: 01/20/2023] Open
Abstract
Brain-derived neurotrophic factor (BDNF) induces activation of the TrkB receptor and several downstream pathways (MAPK, PI3K, PLC-γ), leading to neuronal survival, growth, and plasticity. It has been well established that TrkB signaling regulation is required for neurite formation and dendritic arborization, but the specific mechanism is not fully understood. The non-receptor tyrosine kinase c-Abl is a possible candidate regulator of this process, as it has been implicated in tyrosine kinase receptors' signaling and trafficking, as well as regulation of neuronal morphogenesis. To assess the role of c-Abl in BDNF-induced dendritic arborization, wild-type and c-Abl-KO neurons were stimulated with BDNF, and diverse strategies were employed to probe the function of c-Abl, including the use of pharmacological inhibitors, an allosteric c-Abl activator, and shRNA to downregulates c-Abl expression. Surprisingly, BDNF promoted c-Abl activation and interaction with TrkB receptors. Furthermore, pharmacological c-Abl inhibition and genetic ablation abolished BDNF-induced dendritic arborization and increased the availability of TrkB in the cell membrane. Interestingly, inhibition or genetic ablation of c-Abl had no effect on the classic TrkB downstream pathways. Together, our results suggest that BDNF/TrkB-dependent c-Abl activation is a novel and essential mechanism in TrkB signaling.
Collapse
|
5
|
Bogacheva PO, Molchanova AI, Pravdivceva ES, Miteva AS, Balezina OP, Gaydukov AE. ProBDNF and Brain-Derived Neurotrophic Factor Prodomain Differently Modulate Acetylcholine Release in Regenerating and Mature Mouse Motor Synapses. Front Cell Neurosci 2022; 16:866802. [PMID: 35591942 PMCID: PMC9110780 DOI: 10.3389/fncel.2022.866802] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Accepted: 03/18/2022] [Indexed: 11/30/2022] Open
Abstract
The effects of brain-derived neurotrophic factor (BDNF) processing by-products (proBDNF and BDNF prodomain) on the activity of mouse neuromuscular junctions (NMJs) were studied in synapses formed during the reinnervation of extensor digitorum longus muscle (m. EDL) and mature synapses of the diaphragm. The parameters of spontaneous miniature endplate potentials (MEPPs) and evoked endplate potentials (EPPs) were analyzed in presence of each of the BDNF maturation products (both – 1 nM). In newly formed NMJs, proBDNF caused an increase in the resting membrane potential of muscle fibers and a decrease in the frequency of MEPPs, which was prevented by tertiapin-Q, a G-protein-coupled inwardly rectifying potassium channels (GIRK) blocker but not by p75 receptor signaling inhibitor TAT-Pep5. proBDNF had no effect on the parameters of EPPs. BDNF prodomain in newly formed synapses had effects different from those of proBDNF: it increased the amplitude of MEPPs, which was prevented by vesamicol, an inhibitor of vesicular acetylcholine (ACh) transporter; and reduced the quantal content of EPPs. In mature NMJs, proBDNF did not influence MEPPs parameters, but BDNF prodomain suppressed both spontaneous and evoked ACh release: decreased the frequency and amplitude of MEPPs, and the amplitude and quantal content of EPPs. This effect of the BDNF prodomain was prevented by blocking GIRK channels, by TAT-Pep5 or by Rho-associated protein kinase (ROCK) inhibitor Y-27632. At the same time, the BDNF prodomain did not show any inhibitory effects in diaphragm motor synapses of pannexin 1 knockout mice, which have impaired purinergic regulation of neuromuscular transmission. The data obtained suggest that there is a previously unknown mechanism for the acute suppression of spontaneous and evoked ACh release in mature motor synapses, which involves the activation of p75 receptors, ROCK and GIRK channels by BDNF prodomain and requires interaction with metabotropic purinoreceptors. In general, our results show that both the precursor of BDNF and the product of its maturation have predominantly inhibitory effects on spontaneous and evoked ACh release in newly formed or functionally mature neuromuscular junctions, which are mainly opposite to the effects of BDNF. The inhibitory influences of both proteins related to brain neurotrophin are mediated via GIRK channels of mouse NMJs.
Collapse
|
6
|
Hossain MS, Mawatari S, Fujino T. Plasmalogens, the Vinyl Ether-Linked Glycerophospholipids, Enhance Learning and Memory by Regulating Brain-Derived Neurotrophic Factor. Front Cell Dev Biol 2022; 10:828282. [PMID: 35223852 PMCID: PMC8864319 DOI: 10.3389/fcell.2022.828282] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2021] [Accepted: 01/17/2022] [Indexed: 01/22/2023] Open
Abstract
Plasmalogens (Pls), a kind of glycerophospholipids, have shown potent biological effects but their role in hippocampus-dependent memory remained mostly elusive. Here, we first report Pls can enhance endogenous expression of brain-derived neurotrophic factor (Bdnf) in the hippocampus and promotes neurogenesis associated with improvement of learning and memory in mice. Genomic and proteomic studies revealed that Pls enhanced recruitment of CREB transcription factor onto the murine Bdnf promoter region via upregulating ERK-Akt signaling pathways in neuronal cells. Reduction of endogenous Pls in murine hippocampus significantly reduced learning and memory associated with the reduction of memory-related protein expression, suggesting that Pls can regulate memory-related gene expression in the hippocampus.
Collapse
Affiliation(s)
| | - Shiro Mawatari
- Institute of Rheological Functions of Food, Fukuoka, Japan
| | | |
Collapse
|
7
|
A maestro role of adenosine A 2A receptors in GABAergic synapses stabilization during postnatal neuronal maturation. Purinergic Signal 2022; 18:157-159. [PMID: 35119605 DOI: 10.1007/s11302-022-09845-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2022] [Accepted: 01/13/2022] [Indexed: 10/19/2022] Open
|
8
|
Wang T, Meunier FA. Live-Cell Superresolution Imaging of Retrograde Axonal Trafficking Using Pulse-Chase Labeling in Cultured Hippocampal Neurons. Methods Mol Biol 2022; 2473:101-128. [PMID: 35819762 DOI: 10.1007/978-1-0716-2209-4_9] [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] [Indexed: 06/15/2023]
Abstract
The entanglement of long axons found in cultured dissociated hippocampal neurons restricts the analysis of the machinery underlying directed axonal trafficking. Further, hippocampal neurons exhibit "en passant" presynapses that may confound the analysis of long-range retrograde axonal transport. To solve these issues, we and others have developed microfluid-based methods to specifically follow the fates of the retrograde axonal cargoes following pulse-chase labeling by super-resolution live-cell imaging, and automatically tracking their directed transport and analyzing their kinetical properties. These methods have allowed us to visualize the trafficking of fluorescently tagged signaling endosomes and autophagosomes derived from axonal terminals and resolve their localizations and movements with high spatial and temporal accuracy. In this chapter, we describe how to use a commercially available microfluidic device to enable the labeling and tracking of retrograde axonal carriers, including (1) how to culture and transfect rat hippocampal neurons in the microfluidic device; (2) how to perform pulse-chase to label specific populations of retrograde axonal carriers; and (3) how to conduct the automatic tracking and data analysis using open-source software.
Collapse
Affiliation(s)
- Tong Wang
- Center for Brain Science, School of Life Science and Technology, Shanghaitech University, Shanghai, China.
| | - Frédéric A Meunier
- Clem Jones Centre for Ageing Dementia Research, Queensland Brain Institute, The University of Queensland, Brisbane, QLD, Australia
| |
Collapse
|
9
|
Ribeiro FF, Ferreira F, Rodrigues RS, Soares R, Pedro DM, Duarte-Samartinho M, Aroeira RI, Ferreiro E, Valero J, Solá S, Mira H, Sebastião AM, Xapelli S. Regulation of hippocampal postnatal and adult neurogenesis by adenosine A 2A receptor: Interaction with brain-derived neurotrophic factor. Stem Cells 2021; 39:1362-1381. [PMID: 34043863 DOI: 10.1002/stem.3421] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2020] [Revised: 05/06/2021] [Accepted: 05/06/2021] [Indexed: 06/12/2023]
Abstract
Adenosine A2A receptor (A2A R) activation modulates several brain processes, ranging from neuronal maturation to synaptic plasticity. Most of these actions occur through the modulation of the actions of the neurotrophin brain-derived neurotrophic factor (BDNF). In this work, we studied the role of A2A Rs in regulating postnatal and adult neurogenesis in the rat hippocampal dentate gyrus (DG). Here, we show that A2A R activation with CGS 21680 promoted neural stem cell self-renewal, protected committed neuronal cells from cell death and contributed to a higher density of immature and mature neuronal cells, particularly glutamatergic neurons. Moreover, A2A R endogenous activation was found to be essential for BDNF-mediated increase in cell proliferation and neuronal differentiation. Our findings contribute to further understand the role of adenosinergic signaling in the brain and may have an impact in the development of strategies for brain repair under pathological conditions.
Collapse
Affiliation(s)
- Filipa F Ribeiro
- Instituto de Farmacologia e Neurociências, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal
- Instituto de Medicina Molecular João Lobo Antunes (iMM, JLB), Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal
| | - Filipa Ferreira
- Instituto de Farmacologia e Neurociências, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal
- Instituto de Medicina Molecular João Lobo Antunes (iMM, JLB), Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal
| | - Rui S Rodrigues
- Instituto de Farmacologia e Neurociências, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal
- Instituto de Medicina Molecular João Lobo Antunes (iMM, JLB), Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal
| | - Rita Soares
- Instituto de Farmacologia e Neurociências, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal
- Instituto de Medicina Molecular João Lobo Antunes (iMM, JLB), Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal
- iMed.ULisboa, Faculdade de Farmácia, Universidade de Lisboa, Lisbon, Portugal
| | - Diogo M Pedro
- Instituto de Farmacologia e Neurociências, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal
- Instituto de Medicina Molecular João Lobo Antunes (iMM, JLB), Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal
| | - Marta Duarte-Samartinho
- Instituto de Farmacologia e Neurociências, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal
- Instituto de Medicina Molecular João Lobo Antunes (iMM, JLB), Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal
| | - Rita I Aroeira
- Instituto de Farmacologia e Neurociências, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal
- Instituto de Medicina Molecular João Lobo Antunes (iMM, JLB), Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal
| | - Elisabete Ferreiro
- Center for Neuroscience and Cell Biology (CNC), University of Coimbra, Coimbra, Portugal
| | - Jorge Valero
- Laboratory of Glial Cell Biology, Achucarro Basque Center for Neuroscience, Leioa, Spain
- Ikerbasque Foundation, Bilbao, Spain
- University of the Basque Country EHU/UPV, Leioa, Spain
| | - Susana Solá
- iMed.ULisboa, Faculdade de Farmácia, Universidade de Lisboa, Lisbon, Portugal
| | - Helena Mira
- Instituto de Salud Carlos III (ISCIII), Majadahonda, Madrid, Spain
- Instituto de Biomedicina de Valencia (IBV-CSIC), Valencia, Spain
| | - Ana M Sebastião
- Instituto de Farmacologia e Neurociências, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal
- Instituto de Medicina Molecular João Lobo Antunes (iMM, JLB), Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal
| | - Sara Xapelli
- Instituto de Farmacologia e Neurociências, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal
- Instituto de Medicina Molecular João Lobo Antunes (iMM, JLB), Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal
| |
Collapse
|
10
|
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
|
11
|
Miranda-Lourenço C, Duarte ST, Palminha C, Gaspar C, Rodrigues TM, Magalhães-Cardoso T, Rei N, Colino-Oliveira M, Gomes R, Ferreira S, Rosa J, Xapelli S, Armstrong J, García-Cazorla À, Correia-de-Sá P, Sebastião AM, Diógenes MJ. Impairment of adenosinergic system in Rett syndrome: Novel therapeutic target to boost BDNF signalling. Neurobiol Dis 2020; 145:105043. [PMID: 32798727 DOI: 10.1016/j.nbd.2020.105043] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2020] [Revised: 07/23/2020] [Accepted: 08/08/2020] [Indexed: 01/20/2023] Open
Abstract
Rett syndrome (RTT; OMIM#312750) is mainly caused by mutations in the X-linked MECP2 gene (methyl-CpG-binding protein 2 gene; OMIM*300005), which leads to impairments in the brain-derived neurotrophic factor (BDNF) signalling. The boost of BDNF mediated effects would be a significant breakthrough but it has been hampered by the difficulty to administer BDNF to the central nervous system. Adenosine, an endogenous neuromodulator, may accomplish that role since through A2AR it potentiates BDNF synaptic actions in healthy animals. We thus characterized several hallmarks of the adenosinergic and BDNF signalling in RTT and explored whether A2AR activation could boost BDNF actions. For this study, the RTT animal model, the Mecp2 knockout (Mecp2-/y) (B6.129P2 (C)-Mecp2tm1.1Bird/J) mouse was used. Whenever possible, parallel data was also obtained from post-mortem brain samples from one RTT patient. Ex vivo extracellular recordings of field excitatory post-synaptic potentials in CA1 hippocampal area were performed to evaluate synaptic transmission and long-term potentiation (LTP). RT-PCR was used to assess mRNA levels and Western Blot or radioligand binding assays were performed to evaluate protein levels. Changes in cortical and hippocampal adenosine content were assessed by liquid chromatography with diode array detection (LC/DAD). Hippocampal ex vivo experiments revealed that the facilitatory actions of BDNF upon LTP is absent in Mecp2-/y mice and that TrkB full-length (TrkB-FL) receptor levels are significantly decreased. Extracts of the hippocampus and cortex of Mecp2-/y mice revealed less adenosine amount as well as less A2AR protein levels when compared to WT littermates, which may partially explain the deficits in adenosinergic tonus in these animals. Remarkably, the lack of BDNF effect on hippocampal LTP in Mecp2-/y mice was overcome by selective activation of A2AR with CGS21680. Overall, in Mecp2-/y mice there is an impairment on adenosinergic system and BDNF signalling. These findings set the stage for adenosine-based pharmacological therapeutic strategies for RTT, highlighting A2AR as a therapeutic target in this devastating pathology.
Collapse
Affiliation(s)
- Catarina Miranda-Lourenço
- Instituto de Farmacologia e Neurociências, Faculdade de Medicina e Instituto de Medicina Molecular - João Lobo Antunes, Universidade de Lisboa, Lisboa, Portugal.
| | - Sofia T Duarte
- Instituto de Farmacologia e Neurociências, Faculdade de Medicina e Instituto de Medicina Molecular - João Lobo Antunes, Universidade de Lisboa, Lisboa, Portugal; Child Neurology Department, Hospital Dona Estefânia - Centro Hospitalar Universitário de Lisboa Central, Portugal.
| | - Cátia Palminha
- Instituto de Farmacologia e Neurociências, Faculdade de Medicina e Instituto de Medicina Molecular - João Lobo Antunes, Universidade de Lisboa, Lisboa, Portugal.
| | - Cláudia Gaspar
- Instituto de Medicina Molecular - João Lobo Antunes, Universidade de Lisboa, Lisboa, Portugal.
| | - Tiago M Rodrigues
- Instituto de Farmacologia e Neurociências, Faculdade de Medicina e Instituto de Medicina Molecular - João Lobo Antunes, Universidade de Lisboa, Lisboa, Portugal; Institute of Molecular and Clinical Ophtalmology, Mittlere Strasse 91, CH-4031 Basel, Switzerland.
| | - Teresa Magalhães-Cardoso
- Laboratório de Farmacologia e Neurobiologia / MedInUP, Instituto de Ciências Biomédicas Abel Salazar - Universidade do Porto (ICBAS-UP), Porto, Portugal.
| | - Nádia Rei
- Instituto de Farmacologia e Neurociências, Faculdade de Medicina e Instituto de Medicina Molecular - João Lobo Antunes, Universidade de Lisboa, Lisboa, Portugal.
| | - Mariana Colino-Oliveira
- Instituto de Farmacologia e Neurociências, Faculdade de Medicina e Instituto de Medicina Molecular - João Lobo Antunes, Universidade de Lisboa, Lisboa, Portugal.
| | - Rui Gomes
- Instituto de Medicina Molecular - João Lobo Antunes, Universidade de Lisboa, Lisboa, Portugal.
| | - Sara Ferreira
- Instituto de Medicina Molecular - João Lobo Antunes, Universidade de Lisboa, Lisboa, Portugal.
| | - Jéssica Rosa
- Instituto de Farmacologia e Neurociências, Faculdade de Medicina e Instituto de Medicina Molecular - João Lobo Antunes, Universidade de Lisboa, Lisboa, Portugal.
| | - Sara Xapelli
- Instituto de Farmacologia e Neurociências, Faculdade de Medicina e Instituto de Medicina Molecular - João Lobo Antunes, Universidade de Lisboa, Lisboa, Portugal.
| | - Judith Armstrong
- Genetics Department, Hospital Sant Joan de Deu. Institut Pediàtric de Recerca and CIBERER. (ISCIII), Barcelona, Spain.
| | - Àngels García-Cazorla
- Synaptic Metabolism Laboratory, Neurology Department; Institut Pediàtric de Recerca and CIBERER. (ISCIII), Barcelona, Spain.
| | - Paulo Correia-de-Sá
- Laboratório de Farmacologia e Neurobiologia / MedInUP, Instituto de Ciências Biomédicas Abel Salazar - Universidade do Porto (ICBAS-UP), Porto, Portugal.
| | - Ana M Sebastião
- Instituto de Farmacologia e Neurociências, Faculdade de Medicina e Instituto de Medicina Molecular - João Lobo Antunes, Universidade de Lisboa, Lisboa, Portugal.
| | - Maria José Diógenes
- Instituto de Farmacologia e Neurociências, Faculdade de Medicina e Instituto de Medicina Molecular - João Lobo Antunes, Universidade de Lisboa, Lisboa, Portugal.
| |
Collapse
|
12
|
Ma Y, Deng M, Zhao XQ, Liu M. Alternatively Polarized Macrophages Regulate the Growth and Differentiation of Ependymal Stem Cells through the SIRT2 Pathway. Exp Neurobiol 2020; 29:150-163. [PMID: 32408405 PMCID: PMC7237271 DOI: 10.5607/en19078] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2019] [Revised: 04/15/2020] [Accepted: 04/16/2020] [Indexed: 02/06/2023] Open
Abstract
Ependymal stem cells (EpSCs) are dormant stem cells in the adult spinal cord that proliferate rapidly and migrate to the site of injury after spinal cord injury (SCI). Although they can differentiate into neurons under appropriate conditions in vitro, EpSCs mainly differentiate into astrocytes in vivo. Our previous study confirmed that alternatively polarized macrophages (M2) facilitate the differentiation of EpSCs towards neurons, but the detailed mechanism remains elusive. In the present study, we found that M2 conditioned medium could upregulate the expression of Sirtuin 2 (SIRT2) in EpSCs in vitro through the BDNF/TrkB-MEK/ERK signaling pathway. As an important deacetylase, SIRT2 deacetylated stable Ac-α-tubulin (Acetyl alpha Tubulin) in microtubules and thus promoted EpSC differentiation into neurons. The present study provides a theoretical basis and a new way to improve neural recovery, such as regulating the growth and differentiation of EpSCs by increasing the proportion of M2 cells in the local microenvironment or upregulating the expression of SIRT2 in EpSCs.
Collapse
Affiliation(s)
- Yonggang Ma
- Department of Orthopaedics, Renmin Hospital, Wuhan University, Wuhan 430060, China
| | - Ming Deng
- Department of Orthopaedics, Renmin Hospital, Wuhan University, Wuhan 430060, China
| | - Xiao-Qi Zhao
- Department of Pharmacology, School of Basic Medical Sciences, Wuhan University, Wuhan 430071, China
| | - Min Liu
- Department of Immunology, School of Basic Medical Sciences, Wuhan University, Wuhan 430071, China
| |
Collapse
|
13
|
Zhao R, Qiao J, Zhang X, Zhao Y, Meng X, Sun D, Peng X. Toll-Like Receptor-Mediated Activation of CD39 Internalization in BMDCs Leads to Extracellular ATP Accumulation and Facilitates P2X7 Receptor Activation. Front Immunol 2019; 10:2524. [PMID: 31736956 PMCID: PMC6834529 DOI: 10.3389/fimmu.2019.02524] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2019] [Accepted: 10/10/2019] [Indexed: 12/19/2022] Open
Abstract
Toll-like receptors (TLRs) trigger innate immune responses through their recognition of conserved molecular ligands of either endogenous or microbial origin. Although activation, function, and signaling pathways of TLRs were already well-studied, their precise function in specific cell types, especially innate immune cells, needs to be further clarified. In this study, we showed that when significantly decreased amounts of membrane CD39, an adenosine triphosphate (ATP)-degrading enzyme, were detected in lipopolysaccharide (LPS)-treated bone marrow-derived dendritic cells (BMDCs), Cd39 mRNA expression, and whole-cell CD39 expression were at the same levels as those in untreated BMDCs. Further experiments demonstrated that the downregulation of membrane CD39 expression in LPS-treated BMDCs was mediated by endocytosis, leading to membrane-exposed CD39 downregulation, which was positively associated with decreased enzymatic activity in ATP metabolism and increased extracellular ATP accumulation. The accumulated ATP promoted intracellular calcium accumulation and IL-1β production in BMDCs through P2X7 signaling activation. Further research revealed that not only LPS but also other TLR ligands, excluding polyI:C, induced CD39 internalization in BMDCs and that the MyD88 pathway was critical in this process. The results suggested that the activation of CD39 internalization in DCs induced by a TLR ligand caused increased ATP accumulation, leading to P2X7 receptor activation that mediated a proinflammatory effect. Considering the strong modulatory effect of extracellular ATP accumulation on the immune response and inflammation, the manipulation of membrane CD39 expression on DCs may have implications on the regulation and treatment of inflammatory responses.
Collapse
Affiliation(s)
- Ronglan Zhao
- Department of Laboratory Medicine, Weifang Medical University, Weifang, China.,Institutional Key Laboratory of Clinical Laboratory Diagnostics, 12th 5-Year Project of Shandong Province, Weifang Medical University, Weifang, China
| | - Jinjuan Qiao
- Department of Laboratory Medicine, Weifang Medical University, Weifang, China.,Institutional Key Laboratory of Clinical Laboratory Diagnostics, 12th 5-Year Project of Shandong Province, Weifang Medical University, Weifang, China
| | - Xumei Zhang
- Department of Pathology, Affiliated Hospital of Weifang Medical University, Weifang, China
| | - Yansong Zhao
- Department of Ophthalmology, Affiliated Hospital of Weifang Medical University, Weifang, China
| | - Xiangying Meng
- Department of Laboratory Medicine, Weifang Medical University, Weifang, China.,Institutional Key Laboratory of Clinical Laboratory Diagnostics, 12th 5-Year Project of Shandong Province, Weifang Medical University, Weifang, China
| | - Deming Sun
- Department of Ophthalmology, David Geffen School of Medicine at UCLA, Doheny Eye Institute, Los Angeles, CA, United States
| | - Xiaoxiang Peng
- Department of Laboratory Medicine, Weifang Medical University, Weifang, China.,Institutional Key Laboratory of Clinical Laboratory Diagnostics, 12th 5-Year Project of Shandong Province, Weifang Medical University, Weifang, China
| |
Collapse
|
14
|
Zhang P, Zhang Y, Liu K, Liu B, Xu W, Gao J, Ding L, Tao L. Ivermectin induces cell cycle arrest and apoptosis of HeLa cells via mitochondrial pathway. Cell Prolif 2019; 52:e12543. [PMID: 30515909 PMCID: PMC6496724 DOI: 10.1111/cpr.12543] [Citation(s) in RCA: 55] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2018] [Revised: 09/16/2018] [Accepted: 09/17/2018] [Indexed: 11/27/2022] Open
Abstract
OBJECTIVES The aim of study was to investigate the anticancer activities of Ivermectin (IVM) and the possible mechanisms in cells level via cell proliferation inhibition, apoptosis and migration inhibition in model cancer cell HeLa. MATERIALS AND METHODS The MTT assay was used to study the inhibitory effect of IVM on the proliferation of Hela cells, and the cell cycle was analysed by flow cytometry. The neutral comet assay was used to study the DNA damage. The presence of apoptosis was confirmed by DAPI nuclear staining and flow cytometry. Changes in mitochondrial membrane potential and reactive oxygen species (ROS) levels were determined using Rhodamine 123 staining and DCFH-DA staining. Western blot analysis for apoptosis-related proteins was carried out. We use scratch test to analyse the antimigration potential of IVM. RESULTS Ivermectin can inhibit the viability of HeLa cells significantly. In addition, treatment with IVM resulted in cell cycle arrest at the G1/S phase which partly account for the suppressed proliferation. Typical apoptosis morphological changes were shown in IVM treatment cells including DNA fragmentation and chromatin condensation. At the same time, the results of flow cytometry analysis showed that the number of apoptotic cells increased significantly with the increase of IVM concentration. Moreover, we observed that the mitochondrial membrane potential collapses and the ratio of Bax/Bcl-2 in the cytoplasm increases, which induces cytochrome c release from the mitochondria to the cytoplasm, activates caspase-9/-3 and finally induces apoptosis. We also found that IVM can significantly increase intracellular ROS content. At the same time, we determined that IVM can significantly inhibit the migration of HeLa cells. CONCLUSIONS Our experimental results show that IVM might be a new potential anticancer drug for therapy of human cancer.
Collapse
Affiliation(s)
- Ping Zhang
- Shanghai Key Laboratory of Chemical Biology, School of PharmacyEast China University of Science and TechnologyShanghaiChina
| | - Yang Zhang
- Shanghai Key Laboratory of Chemical Biology, School of PharmacyEast China University of Science and TechnologyShanghaiChina
| | - Kuikui Liu
- Shandong Key Laboratory of Chemical MedicineShandong Academy of Pharmaceutical SciencesJinanChina
| | - Bin Liu
- Vegetable Technical Extension Station Qingpu District ShanghaiShanghaiChina
| | - Wenping Xu
- Shanghai Key Laboratory of Chemical Biology, School of PharmacyEast China University of Science and TechnologyShanghaiChina
| | - Jufang Gao
- College of Life and Environmental SciencesShanghai Normal UniversityShanghaiChina
| | - Lei Ding
- Shanghai Key Laboratory of Chemical Biology, School of PharmacyEast China University of Science and TechnologyShanghaiChina
| | - Liming Tao
- Shanghai Key Laboratory of Chemical Biology, School of PharmacyEast China University of Science and TechnologyShanghaiChina
| |
Collapse
|
15
|
24S-hydroxycholesterol suppresses neuromuscular transmission in SOD1(G93A) mice: A possible role of NO and lipid rafts. Mol Cell Neurosci 2018; 88:308-318. [PMID: 29550246 DOI: 10.1016/j.mcn.2018.03.006] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2017] [Revised: 02/08/2018] [Accepted: 03/12/2018] [Indexed: 12/12/2022] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a neurodegenerative disorder characterized by the initial denervation of skeletal muscle and subsequent death of motor neurons. A dying-back pattern of ALS suggests a crucial role for neuromuscular junction dysfunction. In the present study, microelectrode recording of postsynaptic currents and optical detection of synaptic vesicle traffic (FM1-43 dye) and intracellular NO levels (DAF-FM DA) were used to examine the effect of the major brain-derived cholesterol metabolite 24S-hydroxycholesterol (24S-HC, 0.4 μM) on neuromuscular transmission in the diaphragm of transgenic mice carrying a mutant superoxide dismutase 1 (SODG93A). We found that 24S-HC suppressed spontaneous neurotransmitter release and neurotransmitter exocytosis during high-frequency stimulation. The latter was accompanied by a decrease in both the rate of synaptic vesicle recycling and activity-dependent enhancement of NO production. Inhibition of NO synthase with L-NAME also attenuated synaptic vesicle exocytosis during high-frequency stimulation and completely abolished the effect of 24S-HC itself. Of note, 24S-HC enhanced the labeling of synaptic membranes with B-subunit of cholera toxin, suggesting an increase in lipid ordering. Lipid raft-disrupting agents (methyl-β-cyclodextrin, sphingomyelinase) prevented the action of 24S-HC on both lipid raft marker labeling and NO synthesis. Together, these experiments indicate that 24S-HC is able to suppress the exocytotic release of neurotransmitter in response to intense activity via a NO/lipid raft-dependent pathway in the neuromuscular junctions of SODG93A mice.
Collapse
|
16
|
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]
|
17
|
Zhao H, Alam A, San CY, Eguchi S, Chen Q, Lian Q, Ma D. Molecular mechanisms of brain-derived neurotrophic factor in neuro-protection: Recent developments. Brain Res 2017; 1665:1-21. [PMID: 28396009 DOI: 10.1016/j.brainres.2017.03.029] [Citation(s) in RCA: 70] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2016] [Revised: 03/02/2017] [Accepted: 03/28/2017] [Indexed: 12/13/2022]
Abstract
Neuronal cell injury, as a consequence of acute or chronic neurological trauma, is a significant cause of mortality around the world. On a molecular level, the condition is characterized by widespread cell death and poor regeneration, which can result in severe morbidity in survivors. Potential therapeutics are of major interest, with a promising candidate being brain-derived neurotrophic factor (BDNF), a ubiquitous agent in the brain which has been associated with neural development and may facilitate protective and regenerative effects following injury. This review summarizes the available information on the potential benefits of BDNF and the molecular mechanisms involved in several pathological conditions, including hypoxic brain injury, stroke, Alzheimer's disease and Parkinson's disease. It further explores the methods in which BDNF can be applied in clinical and therapeutic settings, and the potential challenges to overcome.
Collapse
Affiliation(s)
- Hailin Zhao
- Anaesthetics, Pain Medicine and Intensive Care, Department of Surgery and Cancer, Faculty of Medicine, Imperial College London, Chelsea & Westminster Hospital, London, UK
| | - Azeem Alam
- Anaesthetics, Pain Medicine and Intensive Care, Department of Surgery and Cancer, Faculty of Medicine, Imperial College London, Chelsea & Westminster Hospital, London, UK
| | - Chun-Yin San
- Anaesthetics, Pain Medicine and Intensive Care, Department of Surgery and Cancer, Faculty of Medicine, Imperial College London, Chelsea & Westminster Hospital, London, UK
| | - Shiori Eguchi
- Anaesthetics, Pain Medicine and Intensive Care, Department of Surgery and Cancer, Faculty of Medicine, Imperial College London, Chelsea & Westminster Hospital, London, UK
| | - Qian Chen
- Anaesthetics, Pain Medicine and Intensive Care, Department of Surgery and Cancer, Faculty of Medicine, Imperial College London, Chelsea & Westminster Hospital, London, UK; Department of Anaesthesiology, Southwest Hospital, Third Military Medical University, Chongqing, China
| | - Qingquan Lian
- Department of Anesthesiology, Second Affiliated Hospital, Wenzhou Medical University, Wenzhou, China.
| | - Daqing Ma
- Anaesthetics, Pain Medicine and Intensive Care, Department of Surgery and Cancer, Faculty of Medicine, Imperial College London, Chelsea & Westminster Hospital, London, UK.
| |
Collapse
|
18
|
Understanding the Functional Plasticity in Neural Networks of the Basal Ganglia in Cocaine Use Disorder: A Role for Allosteric Receptor-Receptor Interactions in A2A-D2 Heteroreceptor Complexes. Neural Plast 2016; 2016:4827268. [PMID: 27872762 PMCID: PMC5107220 DOI: 10.1155/2016/4827268] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2016] [Revised: 08/28/2016] [Accepted: 10/03/2016] [Indexed: 11/17/2022] Open
Abstract
Our hypothesis is that allosteric receptor-receptor interactions in homo- and heteroreceptor complexes may form the molecular basis of learning and memory. This principle is illustrated by showing how cocaine abuse can alter the adenosine A2AR-dopamine D2R heterocomplexes and their receptor-receptor interactions and hereby induce neural plasticity in the basal ganglia. Studies with A2AR ligands using cocaine self-administration procedures indicate that antagonistic allosteric A2AR-D2R heterocomplexes of the ventral striatopallidal GABA antireward pathway play a significant role in reducing cocaine induced reward, motivation, and cocaine seeking. Anticocaine actions of A2AR agonists can also be produced at A2AR homocomplexes in these antireward neurons, actions in which are independent of D2R signaling. At the A2AR-D2R heterocomplex, they are dependent on the strength of the antagonistic allosteric A2AR-D2R interaction and the number of A2AR-D2R and A2AR-D2R-sigma1R heterocomplexes present in the ventral striatopallidal GABA neurons. It involves a differential cocaine-induced increase in sigma1Rs in the ventral versus the dorsal striatum. In contrast, the allosteric brake on the D2R protomer signaling in the A2AR-D2R heterocomplex of the dorsal striatopallidal GABA neurons is lost upon cocaine self-administration. This is potentially due to differences in composition and allosteric plasticity of these complexes versus those in the ventral striatopallidal neurons.
Collapse
|
19
|
Ribeiro FF, Sebastião AM. Adenosine A2A receptors in neuronal outgrowth: a target for nerve regeneration? Neural Regen Res 2016; 11:706-8. [PMID: 27335540 PMCID: PMC4904447 DOI: 10.4103/1673-5374.182683] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/15/2016] [Indexed: 01/19/2023] Open
Affiliation(s)
- Filipa F. Ribeiro
- Instituto de Farmacologia e Neurociências, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal
| | - Ana M. Sebastião
- Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal
| |
Collapse
|
20
|
Kasimov MR, Zakyrjanova GF, Giniatullin AR, Zefirov AL, Petrov AM. Similar oxysterols may lead to opposite effects on synaptic transmission: Olesoxime versus 5α-cholestan-3-one at the frog neuromuscular junction. Biochim Biophys Acta Mol Cell Biol Lipids 2016; 1861:606-16. [PMID: 27102612 DOI: 10.1016/j.bbalip.2016.04.010] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2015] [Revised: 03/17/2016] [Accepted: 04/15/2016] [Indexed: 02/03/2023]
Abstract
Cholesterol oxidation products frequently have a high biological activity. In the present study, we have used microelectrode recording of end plate currents and FM-based optical detection of synaptic vesicle exo-endocytosis to investigate the effects of two structurally similar oxysterols, olesoxime (cholest-4-en-3-one, oxime) and 5ɑ-cholestan-3-one (5ɑCh3), on neurotransmission at the frog neuromuscular junction. Olesoxime is an exogenous, potentially neuroprotective, substance and 5ɑCh3 is an intermediate product in cholesterol metabolism, which is elevated in the case of cerebrotendinous xanthomatosis. We found that olesoxime slightly increased evoked neurotransmitter release in response to a single stimulus and significantly reduced synaptic depression during high frequency activity. The last effect was due to an increase in both the number of synaptic vesicles involved in exo-endocytosis and the rate of synaptic vesicle recycling. In contrast, 5ɑCh3 reduced evoked neurotransmitter release during the low- and high frequency synaptic activities. The depressant action of 5ɑCh3 was associated with a reduction in the number of synaptic vesicles participating in exo- and endocytosis during high frequency stimulation, without a change in rate of the synaptic vesicle recycling. Of note, olesoxime increased the staining of synaptic membranes with the B-subunit of cholera toxin and the formation of fluorescent ganglioside GM1 clusters, and decreased the fluorescence of 22-NBD-cholesterol, while 5ɑCh3 had the opposite effects, suggesting that the two oxysterols have different effects on lipid raft stability. Taken together, these data show that these two structurally similar oxysterols induce marked different changes in neuromuscular transmission which are related with the alteration in synaptic vesicle cycle.
Collapse
Affiliation(s)
- M R Kasimov
- Department of Normal Physiology, Kazan State Medical University, Kazan 420012, Russia
| | - G F Zakyrjanova
- Department of Normal Physiology, Kazan State Medical University, Kazan 420012, Russia
| | - A R Giniatullin
- Department of Normal Physiology, Kazan State Medical University, Kazan 420012, Russia
| | - A L Zefirov
- Department of Normal Physiology, Kazan State Medical University, Kazan 420012, Russia
| | - A M Petrov
- Department of Normal Physiology, Kazan State Medical University, Kazan 420012, Russia.
| |
Collapse
|
21
|
Kalirin is required for BDNF-TrkB stimulated neurite outgrowth and branching. Neuropharmacology 2016; 107:227-238. [PMID: 27036892 DOI: 10.1016/j.neuropharm.2016.03.050] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2015] [Revised: 03/03/2016] [Accepted: 03/28/2016] [Indexed: 01/19/2023]
Abstract
Exogenous brain-derived neurotrophic factor (BDNF), acting through TrkB, is known to promote neurite formation and branching. This response to BDNF was eliminated by inhibition of TrkB kinase and by specific inhibition of the GEF1 domain of Kalirin, which activates Rac1. Neurons from Kalrn knockout mice were unable to activate Rac1 in response to BDNF. BDNF-triggered neurite outgrowth was abolished when Kalrn expression was reduced using shRNA that targets all of the major Kalrn isoforms, and reduced in neurons from Kalrn knockout mice. The Kalrn isoforms expressed early in development also include a GEF2 domain that activates RhoA. However, BDNF-stimulated neurite outgrowth in Kalrn knockout neurons was rescued by expression of Kalirin-7, which includes only the GEF1 domain but lacks the GEF2 domain. Dendritic morphogenesis, which requires spatially restricted, coordinated changes in the actin cytoskeleton and in the organization of microtubules, involves essential contributions from multiple Rho GEFs. Since Tiam1, another Rho GEF, is also required for BDNF-stimulated neurite outgrowth, an inhibitory fragment of Tiam1 (PHn-CC-EX) was tested and found to interfere with both Kalirin and Tiam1 GEF activity. The prolonged TrkB activation observed in response to BDNF in Kalrn knockout neurons and the altered time course and extent of ERK, CREB and Akt activation observed in the absence of Kalrn would be expected to alter the response of these neurons to other regulatory factors.
Collapse
|
22
|
Benedetti E, Di Loreto S, D'Angelo B, Cristiano L, d'Angelo M, Antonosante A, Fidoamore A, Golini R, Cinque B, Cifone MG, Ippoliti R, Giordano A, Cimini A. The PPARβ/δ Agonist GW0742 Induces Early Neuronal Maturation of Cortical Post-Mitotic Neurons: Role of PPARβ/δ in Neuronal Maturation. J Cell Physiol 2016. [PMID: 26206209 DOI: 10.1002/jcp.25103] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Increasing evidences support that signaling lipids participate in synaptic plasticity and cell survival, and that the lipid signaling is closely associated with neuronal differentiation, learning, and memory and with pathologic events, such as epilepsy and Alzheimer's disease. The Peroxisome Proliferator-Activated Receptors (PPAR) are strongly involved in the fatty acid cell signaling, as many of the natural lypophylic compounds are PPAR ligands. We have previously shown that PPARβ/δ is the main isotype present in cortical neuron primary cultures and that during neuronal maturation, PPARβ/δ is gradually increased and activated. To get more insight into the molecular mechanism by which PPARβ/δ may be involved in neuronal maturation processes, in this work a specific PPARβ/δ agonist, GW0742 was used administered alone or in association with a specific PPARβ/δ antagonist, the GSK0660, and the parameters involved in neuronal differentiation and maturation were assayed. The data obtained demonstrated the strong involvement of PPARβ/δ in neuronal maturation, triggering the agonist an anticipation of neuronal differentiation, and the antagonist abolishing the observed effects. These effects appear to be mediated by the activation of BDNF pathway.
Collapse
Affiliation(s)
- Elisabetta Benedetti
- Department of Life Health and Environmental Sciences, University of L'Aquila, Italy
| | - Silvia Di Loreto
- Institute of Translational Pharmacology (IFT)-CNR, L'Aquila, Italy
| | - Barbara D'Angelo
- Sbarro Institute for Cancer Research and Molecular Medicine and Center for Biotechnology, Temple University, Philadelphia, Pennsylvania
| | - Loredana Cristiano
- Department of Life Health and Environmental Sciences, University of L'Aquila, Italy
| | - Michele d'Angelo
- Department of Life Health and Environmental Sciences, University of L'Aquila, Italy
| | - Andrea Antonosante
- Department of Life Health and Environmental Sciences, University of L'Aquila, Italy
| | - Alessia Fidoamore
- Department of Life Health and Environmental Sciences, University of L'Aquila, Italy
| | - Raffaella Golini
- Department of Life Health and Environmental Sciences, University of L'Aquila, Italy
| | - Benedetta Cinque
- Department of Life Health and Environmental Sciences, University of L'Aquila, Italy
| | - Maria Grazia Cifone
- Department of Life Health and Environmental Sciences, University of L'Aquila, Italy
| | - Rodolfo Ippoliti
- Department of Life Health and Environmental Sciences, University of L'Aquila, Italy
| | - Antonio Giordano
- Sbarro Institute for Cancer Research and Molecular Medicine and Center for Biotechnology, Temple University, Philadelphia, Pennsylvania.,Department of Medicine, Surgery and Neuroscience, University of Siena, Siena, Italy
| | - Annamaria Cimini
- Department of Life Health and Environmental Sciences, University of L'Aquila, Italy.,Sbarro Institute for Cancer Research and Molecular Medicine and Center for Biotechnology, Temple University, Philadelphia, Pennsylvania.,National Institute for Nuclear Physics (INFN), Gran Sasso National Laboratory (LNGS), Assergi, Italy
| |
Collapse
|
23
|
BDNF-induced presynaptic facilitation of GABAergic transmission in the hippocampus of young adults is dependent of TrkB and adenosine A2A receptors. Purinergic Signal 2016; 12:283-94. [PMID: 26897393 DOI: 10.1007/s11302-016-9502-y] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2015] [Accepted: 02/10/2016] [Indexed: 01/03/2023] Open
Abstract
Brain-derived neurotrophic factor (BDNF) and adenosine are widely recognized as neuromodulators of glutamatergic transmission in the adult brain. Most BDNF actions upon excitatory plasticity phenomena are under control of adenosine A2A receptors (A2ARs). Concerning gamma-aminobutyric acid (GABA)-mediated transmission, the available information refers to the control of GABA transporters. We now focused on the influence of BDNF and the interplay with adenosine on phasic GABAergic transmission. To assess this, we evaluated evoked and spontaneous synaptic currents recorded from CA1 pyramidal cells in acute hippocampal slices from adult rat brains (6 to 10 weeks old). BDNF (10-100 ng/mL) increased miniature inhibitory postsynaptic current (mIPSC) frequency, but not amplitude, as well as increased the amplitude of inhibitory postsynaptic currents (IPSCs) evoked by afferent stimulation. The facilitatory action of BDNF upon GABAergic transmission was lost in the presence of a Trk inhibitor (K252a, 200 nM), but not upon p75(NTR) blockade (anti-p75(NTR) IgG, 50 μg/mL). Moreover, the facilitatory action of BDNF onto GABAergic transmission was also prevented upon A2AR antagonism (SCH 58261, 50 nM). We conclude that BDNF facilitates GABAergic signaling at the adult hippocampus via a presynaptic mechanism that depends on TrkB and adenosine A2AR activation.
Collapse
|
24
|
Purine nucleosides in neuroregeneration and neuroprotection. Neuropharmacology 2015; 104:226-42. [PMID: 26577017 DOI: 10.1016/j.neuropharm.2015.11.006] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2015] [Revised: 11/05/2015] [Accepted: 11/06/2015] [Indexed: 12/20/2022]
Abstract
In the present review, we stress the importance of the purine nucleosides, adenosine and guanosine, in protecting the nervous system, both centrally and peripherally, via activation of their receptors and intracellular signalling mechanisms. A most novel part of the review focus on the mechanisms of neuronal regeneration that are targeted by nucleosides, including a recently identified action of adenosine on axonal growth and microtubule dynamics. Discussion on the role of the purine nucleosides transversally with the most established neurotrophic factors, e.g. brain derived neurotrophic factor (BDNF), glial derived neurotrophic factor (GDNF), is also focused considering the intimate relationship between some adenosine receptors, as is the case of the A2A receptors, and receptors for neurotrophins. This article is part of the Special Issue entitled 'Purines in Neurodegeneration and Neuroregeneration'.
Collapse
|
25
|
Adenosine A2A receptor activation is determinant for BDNF actions upon GABA and glutamate release from rat hippocampal synaptosomes. Purinergic Signal 2015; 11:607-12. [PMID: 26452489 DOI: 10.1007/s11302-015-9476-1] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2015] [Accepted: 09/28/2015] [Indexed: 01/07/2023] Open
Abstract
Adenosine, through A(2A) receptor (A(2A)R) activation, can act as a metamodulator, controlling the actions of other modulators, as brain-derived neurotrophic factor (BDNF). Most of the metamodulatory actions of adenosine in the hippocampus have been evaluated in excitatory synapses. However, adenosine and BDNF can also influence GABAergic transmission. We thus evaluated the role of A(2A)R on the modulatory effect of BDNF upon glutamate and GABA release from isolated hippocampal nerve terminals (synaptosomes). BDNF (30 ng/ml) enhanced K(+)-evoked [(3)H]glutamate release and inhibited the K(+)-evoked [(3)H]GABA release from synaptosomes. The effect of BDNF on both glutamate and GABA release requires tonic activation of adenosine A(2A)R since for both neurotransmitters, the BDNF action was blocked by the A(2A)R antagonist SCH 58261 (50 nM). In the presence of the A(2A)R agonist, CGS21680 (30 nM), the effect of BDNF on either glutamate or GABA release was, however, not potentiated. It is concluded that both the inhibitory actions of BDNF on GABA release as well as the facilitatory action of the neurotrophin on glutamate release are dependent on the activation of adenosine A(2A)R by endogenous adenosine. However, these actions could not be further enhanced by exogenous activation of A(2A)R.
Collapse
|
26
|
Ribeiro FF, Neves-Tomé R, Assaife-Lopes N, Santos TE, Silva RFM, Brites D, Ribeiro JA, Sousa MM, Sebastião AM. Axonal elongation and dendritic branching is enhanced by adenosine A2A receptors activation in cerebral cortical neurons. Brain Struct Funct 2015; 221:2777-99. [DOI: 10.1007/s00429-015-1072-1] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2015] [Accepted: 05/27/2015] [Indexed: 01/09/2023]
|
27
|
Jerónimo-Santos A, Fonseca-Gomes J, Guimarães DA, Tanqueiro SR, Ramalho RM, Ribeiro JA, Sebastião AM, Diógenes MJ. Brain-derived neurotrophic factor mediates neuroprotection against Aβ-induced toxicity through a mechanism independent on adenosine 2A receptor activation. Growth Factors 2015; 33:298-308. [PMID: 26365294 DOI: 10.3109/08977194.2015.1080696] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
Brain-derived neurotrophic factor (BDNF) promotes neuronal survival through TrkB-FL activation. The activation of adenosine A2A receptors (A2AR) is essential for most of BDNF-mediated synaptic actions, such as synaptic plasticity, transmission and neurotransmitter release. We now aimed at evaluating the A2AR influence upon BDNF-mediated neuroprotection against Aβ25-35 toxicity in cultured neurons. Results showed that BDNF increases cell survival and reduces the caspase-3 and calpain activation induced by amyloid-β (Aβ) peptide, in a mechanism probably dependent on PLCγ pathway. This BDNF-mediated neuroprotection is not affected by A2AR activation or inhibition. Moreover neither activation nor inhibition of A2AR, per se, significantly influenced Aβ-induced neuronal death on calpain-mediated cleavage of TrkB induced by Aβ. In conclusion, these results suggest that, in opposition to the fast synaptic actions of BDNF, the neuroprotective actions of this neurotrophin against a strong Aβ insult do not require the activation of A2AR.
Collapse
Affiliation(s)
- André Jerónimo-Santos
- a Institute of Pharmacology and Neurosciences, Faculty of Medicine, University of Lisbon , Lisbon , Portugal and
- b Instituto de Medicina Molecular, University of Lisbon , Lisbon , Portugal
| | - João Fonseca-Gomes
- a Institute of Pharmacology and Neurosciences, Faculty of Medicine, University of Lisbon , Lisbon , Portugal and
- b Instituto de Medicina Molecular, University of Lisbon , Lisbon , Portugal
| | - Diogo Andrade Guimarães
- a Institute of Pharmacology and Neurosciences, Faculty of Medicine, University of Lisbon , Lisbon , Portugal and
- b Instituto de Medicina Molecular, University of Lisbon , Lisbon , Portugal
| | - Sara Ramalho Tanqueiro
- a Institute of Pharmacology and Neurosciences, Faculty of Medicine, University of Lisbon , Lisbon , Portugal and
- b Instituto de Medicina Molecular, University of Lisbon , Lisbon , Portugal
| | - Rita Mira Ramalho
- a Institute of Pharmacology and Neurosciences, Faculty of Medicine, University of Lisbon , Lisbon , Portugal and
- b Instituto de Medicina Molecular, University of Lisbon , Lisbon , Portugal
| | - Joaquim Alexandre Ribeiro
- a Institute of Pharmacology and Neurosciences, Faculty of Medicine, University of Lisbon , Lisbon , Portugal and
- b Instituto de Medicina Molecular, University of Lisbon , Lisbon , Portugal
| | - Ana Maria Sebastião
- a Institute of Pharmacology and Neurosciences, Faculty of Medicine, University of Lisbon , Lisbon , Portugal and
- b Instituto de Medicina Molecular, University of Lisbon , Lisbon , Portugal
| | - Maria José Diógenes
- a Institute of Pharmacology and Neurosciences, Faculty of Medicine, University of Lisbon , Lisbon , Portugal and
- b Instituto de Medicina Molecular, University of Lisbon , Lisbon , Portugal
| |
Collapse
|
28
|
Diógenes MJ, Ribeiro JA, Sebastião AM. Adenosine A2A Receptors and Neurotrophic Factors: Relevance for Parkinson’s Disease. CURRENT TOPICS IN NEUROTOXICITY 2015. [DOI: 10.1007/978-3-319-20273-0_4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
|
29
|
Sebastião AM, Ribeiro JA. Neuromodulation and metamodulation by adenosine: Impact and subtleties upon synaptic plasticity regulation. Brain Res 2014; 1621:102-13. [PMID: 25446444 DOI: 10.1016/j.brainres.2014.11.008] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2014] [Revised: 10/30/2014] [Accepted: 11/05/2014] [Indexed: 01/06/2023]
Abstract
Synaptic plasticity mechanisms, i.e. the sequence of events that underlies persistent changes in synaptic strength as a consequence of transient alteration in neuronal firing, are greatly influenced by the 'chemical atmosphere' of the synapses, that is to say by the presence of molecules at the synaptic cleft able to fine-tune the activity of other molecules more directly related to plasticity. One of those fine tuners is adenosine, known for a long time as an ubiquitous neuromodulator and metamodulator and recognized early as influencing synaptic plasticity. In this review we will refer to the mechanisms that adenosine can use to affect plasticity, emphasizing aspects of the neurobiology of adenosine relevant to its ability to control synaptic functioning. This article is part of a Special Issue entitled Brain and Memory.
Collapse
Affiliation(s)
- Ana M Sebastião
- Instituto de Farmacologia e Neurociências, Faculdade de Medicina e Unidade de Neurociências, Instituto de Medicina Molecular, Universidade de Lisboa, Lisboa, Portugal.
| | - Joaquim A Ribeiro
- Instituto de Farmacologia e Neurociências, Faculdade de Medicina e Unidade de Neurociências, Instituto de Medicina Molecular, Universidade de Lisboa, Lisboa, Portugal.
| |
Collapse
|
30
|
Rodrigues TM, Jerónimo-Santos A, Outeiro TF, Sebastião AM, Diógenes MJ. Challenges and promises in the development of neurotrophic factor-based therapies for Parkinson's disease. Drugs Aging 2014; 31:239-61. [PMID: 24610720 DOI: 10.1007/s40266-014-0160-x] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Parkinson's disease (PD) is a chronic movement disorder typically coupled to progressive degeneration of dopaminergic neurons in the substantia nigra (SN). The treatments currently available are satisfactory for symptomatic management, but the efficacy tends to decrease as neuronal loss progresses. Neurotrophic factors (NTFs) are endogenous proteins known to promote neuronal survival, even in degenerating states. Therefore, the use of these factors is regarded as a possible therapeutic approach, which would aim to prevent PD or to even restore homeostasis in neurodegenerative disorders. Intriguingly, although favorable results in in vitro and in vivo models of the disease were attained, clinical trials using these molecules have failed to demonstrate a clear therapeutic benefit. Therefore, the development of animal models that more closely reproduce the mechanisms known to underlie PD-related neurodegeneration would be a major step towards improving the capacity to predict the clinical usefulness of a given NTF-based approach in the experimental setting. Moreover, some adjustments to the design of clinical trials ought to be considered, which include recruiting patients in the initial stages of the disease, improving the efficacy of the delivery methods, and combining synergetic NTFs or adding NTF-boosting drugs to the already available pharmacological approaches. Despite the drawbacks on the road to the use of NTFs as pharmacological tools for PD, very relevant achievements have been reached. In this article, we review the current status of the potential relevance of NTFs for treating PD, taking into consideration experimental evidence, human observational studies, and data from clinical trials.
Collapse
Affiliation(s)
- Tiago Martins Rodrigues
- Instituto de Farmacologia e Neurociências, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal
| | | | | | | | | |
Collapse
|
31
|
Chen JF. Adenosine receptor control of cognition in normal and disease. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2014; 119:257-307. [PMID: 25175970 DOI: 10.1016/b978-0-12-801022-8.00012-x] [Citation(s) in RCA: 107] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Adenosine and adenosine receptors (ARs) are increasingly recognized as important therapeutic targets for controlling cognition under normal and disease conditions for its dual roles of neuromodulation as well as of homeostatic function in the brain. This chapter first presents the unique ability of adenosine, by acting on the inhibitory A1 and facilitating A2A receptor, to integrate dopamine, glutamate, and BNDF signaling and to modulate synaptic plasticity (e.g., long-term potentiation and long-term depression) in brain regions relevant to learning and memory, providing the molecular and cellular bases for adenosine receptor (AR) control of cognition. This led to the demonstration of AR modulation of social recognition memory, working memory, reference memory, reversal learning, goal-directed behavior/habit formation, Pavlovian fear conditioning, and effort-related behavior. Furthermore, human and animal studies support that AR activity can also, through cognitive enhancement and neuroprotection, reverse cognitive impairments in animal models of Alzheimer's disease (AD), Parkinson's disease (PD), Huntington's disease, and schizophrenia. Lastly, epidemiological evidence indicates that regular human consumption of caffeine, the most widely used psychoactive drug and nonselective AR antagonists, is associated with the reduced cognitive decline in aging and AD patients, and with the reduced risk in developing PD. Thus, there is a convergence of the molecular studies revealing AR as molecular targets for integrating neurotransmitter signaling and controlling synaptic plasticity, with animal studies demonstrating the strong procognitive impact upon AR antagonism in normal and disease brains and with epidemiological and clinical evidences in support of caffeine and AR drugs for therapeutic modulation of cognition. Since some of adenosine A2A receptor antagonists are already in phase III clinical trials for motor benefits in PD patients with remarkable safety profiles, additional animal and human studies to better understand the mechanism underlying the AR-mediated control of cognition under normal and disease conditions will provide the required rationale to stimulate the necessary clinical investigation to rapidly translate adenosine and AR drug as a novel strategy to control memory impairment in neuropsychiatric disorders.
Collapse
Affiliation(s)
- Jiang-Fan Chen
- Department of Neurology, Boston University School of Medicine, Boston, Massachusetts, USA; The Molecular Medicine Institute, Wenzhou Medical University, Wenzhou, Zhejiang, PR China.
| |
Collapse
|
32
|
Sonnino S, Aureli M, Grassi S, Mauri L, Prioni S, Prinetti A. Lipid Rafts in Neurodegeneration and Neuroprotection. Mol Neurobiol 2013; 50:130-48. [DOI: 10.1007/s12035-013-8614-4] [Citation(s) in RCA: 63] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2013] [Accepted: 12/08/2013] [Indexed: 11/28/2022]
|