1
|
Henderson J, O'Callaghan J, Campbell M. Gene therapy for glaucoma: Targeting key mechanisms. Vision Res 2024; 225:108502. [PMID: 39423611 PMCID: PMC11579448 DOI: 10.1016/j.visres.2024.108502] [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: 07/31/2024] [Revised: 10/04/2024] [Accepted: 10/04/2024] [Indexed: 10/21/2024]
Abstract
Glaucoma is a group of optic neuropathies characterised by progressive retinal ganglion cell (RGC) degeneration and is the leading cause of irreversible blindness worldwide. Current treatments for glaucoma focus on reducing intraocular pressure (IOP) with topical medications. However, many patients do not achieve sufficient IOP reductions with such treatments. Patient compliance to dosing schedules also poses a significant challenge, further limiting their effectiveness. While surgical options exist for resistant cases, these are invasive and carry risks of complications. Thus, there is a critical need for better strategies to prevent irreversible vision loss in glaucoma. Gene therapy holds significant promise in this regard, offering potential long-term solutions by targeting the disease's underlying causes at a molecular level. Gene therapy strategies for glaucoma primarily target the two key hallmarks of the disease: elevated IOP and RGC death. This review explores key mechanisms underlying these hallmarks and discusses the current state of gene therapies targeting them. In terms of IOP reduction, this review covers strategies aimed at enhancing extracellular matrix turnover in the conventional outflow pathway, targeting fibrosis, regulating aqueous humor production, and targeting myocilin for gene-specific therapy. Neuroprotective strategies explored include targeting neurotrophic factors and their receptors, reducing oxidative stress and mitochondrial dysfunction, and preventing Wallerian degeneration. This review also briefly highlights key research priorities for advancing gene therapies for glaucoma through the clinical pipeline, such as refining delivery vectors and improving transgene regulation. Addressing these priorities will be essential for translating advancements from preclinical models into effective clinical therapies for glaucoma.
Collapse
Affiliation(s)
- Jeff Henderson
- Smurfit Institute of Genetics, Trinity College Dublin, Dublin 2, Ireland
| | | | - Matthew Campbell
- Smurfit Institute of Genetics, Trinity College Dublin, Dublin 2, Ireland.
| |
Collapse
|
2
|
Fazzari M, Lunghi G, Carsana EV, Valsecchi M, Spiombi E, Breccia M, Casati SR, Pedretti S, Mitro N, Mauri L, Ciampa MG, Sonnino S, Landsberger N, Frasca A, Chiricozzi E. GM1 Oligosaccharide Ameliorates Rett Syndrome Phenotypes In Vitro and In Vivo via Trk Receptor Activation. Int J Mol Sci 2024; 25:11555. [PMID: 39519108 PMCID: PMC11547101 DOI: 10.3390/ijms252111555] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2024] [Revised: 10/24/2024] [Accepted: 10/26/2024] [Indexed: 11/16/2024] Open
Abstract
Rett syndrome (RTT) is a severe neurodevelopmental disorder primarily caused by mutations in the methyl-CpG binding protein 2 (MECP2) gene. Despite advancements in research, no cure exists due to an incomplete understanding of the molecular effects of MeCP2 deficiency. Previous studies have identified impaired tropomyosin receptor kinase (Trk) neurotrophin (NTP) signaling and mitochondrial redox imbalances as key drivers of the pathology. Moreover, altered glycosphingolipid metabolism has been reported in RTT. GM1 ganglioside is a known regulator of the nervous system, and growing evidence indicates its importance in maintaining neuronal homeostasis via its oligosaccharide chain, coded as GM1-OS. GM1-OS directly interacts with the Trk receptors on the cell surface, triggering neurotrophic and neuroprotective pathways in neurons. In this study, we demonstrate that GM1-OS ameliorates RTT deficits in the Mecp2-null model. GM1-OS restored synaptogenesis and reduced mitochondrial oxidative stress of Mecp2-knock-out (ko) cortical neurons. When administered in vivo, GM1-OS mitigated RTT-like symptoms. Our findings indicate that GM1-OS effects were mediated by Trk receptor activation on the neuron's plasma membrane. Overall, our results highlight GM1-OS as a promising candidate for RTT treatment.
Collapse
Affiliation(s)
- Maria Fazzari
- Department of Medical Biotechnology and Translational Medicine, Università degli Studi di Milano, 20054 Segrate, Italy; (G.L.); (E.V.C.); (M.V.); (E.S.); (M.B.); (S.R.C.); (L.M.); (M.G.C.); (S.S.); (N.L.); (A.F.)
| | - Giulia Lunghi
- Department of Medical Biotechnology and Translational Medicine, Università degli Studi di Milano, 20054 Segrate, Italy; (G.L.); (E.V.C.); (M.V.); (E.S.); (M.B.); (S.R.C.); (L.M.); (M.G.C.); (S.S.); (N.L.); (A.F.)
| | - Emma Veronica Carsana
- Department of Medical Biotechnology and Translational Medicine, Università degli Studi di Milano, 20054 Segrate, Italy; (G.L.); (E.V.C.); (M.V.); (E.S.); (M.B.); (S.R.C.); (L.M.); (M.G.C.); (S.S.); (N.L.); (A.F.)
| | - Manuela Valsecchi
- Department of Medical Biotechnology and Translational Medicine, Università degli Studi di Milano, 20054 Segrate, Italy; (G.L.); (E.V.C.); (M.V.); (E.S.); (M.B.); (S.R.C.); (L.M.); (M.G.C.); (S.S.); (N.L.); (A.F.)
| | - Eleonora Spiombi
- Department of Medical Biotechnology and Translational Medicine, Università degli Studi di Milano, 20054 Segrate, Italy; (G.L.); (E.V.C.); (M.V.); (E.S.); (M.B.); (S.R.C.); (L.M.); (M.G.C.); (S.S.); (N.L.); (A.F.)
| | - Martina Breccia
- Department of Medical Biotechnology and Translational Medicine, Università degli Studi di Milano, 20054 Segrate, Italy; (G.L.); (E.V.C.); (M.V.); (E.S.); (M.B.); (S.R.C.); (L.M.); (M.G.C.); (S.S.); (N.L.); (A.F.)
| | - Silvia Rosanna Casati
- Department of Medical Biotechnology and Translational Medicine, Università degli Studi di Milano, 20054 Segrate, Italy; (G.L.); (E.V.C.); (M.V.); (E.S.); (M.B.); (S.R.C.); (L.M.); (M.G.C.); (S.S.); (N.L.); (A.F.)
| | - Silvia Pedretti
- Department of Pharmacological and Biomolecular Sciences “Rodolfo Paoletti”, Università degli Studi di Milano, 20133 Milan, Italy; (S.P.); (N.M.)
| | - Nico Mitro
- Department of Pharmacological and Biomolecular Sciences “Rodolfo Paoletti”, Università degli Studi di Milano, 20133 Milan, Italy; (S.P.); (N.M.)
- Department of Experimental Oncology, IEO, European Institute of Oncology IRCCS, 20139 Milan, Italy
| | - Laura Mauri
- Department of Medical Biotechnology and Translational Medicine, Università degli Studi di Milano, 20054 Segrate, Italy; (G.L.); (E.V.C.); (M.V.); (E.S.); (M.B.); (S.R.C.); (L.M.); (M.G.C.); (S.S.); (N.L.); (A.F.)
| | - Maria Grazia Ciampa
- Department of Medical Biotechnology and Translational Medicine, Università degli Studi di Milano, 20054 Segrate, Italy; (G.L.); (E.V.C.); (M.V.); (E.S.); (M.B.); (S.R.C.); (L.M.); (M.G.C.); (S.S.); (N.L.); (A.F.)
| | - Sandro Sonnino
- Department of Medical Biotechnology and Translational Medicine, Università degli Studi di Milano, 20054 Segrate, Italy; (G.L.); (E.V.C.); (M.V.); (E.S.); (M.B.); (S.R.C.); (L.M.); (M.G.C.); (S.S.); (N.L.); (A.F.)
| | - Nicoletta Landsberger
- Department of Medical Biotechnology and Translational Medicine, Università degli Studi di Milano, 20054 Segrate, Italy; (G.L.); (E.V.C.); (M.V.); (E.S.); (M.B.); (S.R.C.); (L.M.); (M.G.C.); (S.S.); (N.L.); (A.F.)
| | - Angelisa Frasca
- Department of Medical Biotechnology and Translational Medicine, Università degli Studi di Milano, 20054 Segrate, Italy; (G.L.); (E.V.C.); (M.V.); (E.S.); (M.B.); (S.R.C.); (L.M.); (M.G.C.); (S.S.); (N.L.); (A.F.)
| | - Elena Chiricozzi
- Department of Medical Biotechnology and Translational Medicine, Università degli Studi di Milano, 20054 Segrate, Italy; (G.L.); (E.V.C.); (M.V.); (E.S.); (M.B.); (S.R.C.); (L.M.); (M.G.C.); (S.S.); (N.L.); (A.F.)
| |
Collapse
|
3
|
Ateaque S, Merkouris S, Barde YA. Neurotrophin signalling in the human nervous system. Front Mol Neurosci 2023; 16:1225373. [PMID: 37470055 PMCID: PMC10352796 DOI: 10.3389/fnmol.2023.1225373] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2023] [Accepted: 06/20/2023] [Indexed: 07/21/2023] Open
Abstract
This review focuses on neurotrophins and their tyrosine kinase receptors, with an emphasis on their relevance to the function and dysfunction in the human nervous system. It also deals with measurements of BDNF levels and highlights recent findings from our laboratory on TrkB and TrkC signalling in human neurons. These include ligand selectivity and Trk activation by neurotrophins and non-neurotrophin ligands. The ligand-induced down-regulation and re-activation of Trk receptors is also discussed.
Collapse
Affiliation(s)
- Sarah Ateaque
- School of Biosciences, Cardiff University, Cardiff, United Kingdom
| | - Spyros Merkouris
- School of Biosciences, Cardiff University, Cardiff, United Kingdom
| | - Yves-Alain Barde
- School of Biosciences, Cardiff University, Cardiff, United Kingdom
| |
Collapse
|
4
|
Sharma V, Singh TG, Kaur A, Mannan A, Dhiman S. Brain-Derived Neurotrophic Factor: A Novel Dynamically Regulated Therapeutic Modulator in Neurological Disorders. Neurochem Res 2023; 48:317-339. [PMID: 36308619 DOI: 10.1007/s11064-022-03755-1] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2022] [Revised: 09/02/2022] [Accepted: 09/03/2022] [Indexed: 02/04/2023]
Abstract
The growth factor brain-derived neurotrophic factor (BDNF), and its receptor tropomyosin-related kinase receptor type B (TrkB) play an active role in numerous areas of the adult brain, where they regulate the neuronal activity, function, and survival. Upregulation and downregulation of BDNF expression are critical for the physiology of neuronal circuits and functioning in the brain. Loss of BDNF function has been reported in the brains of patients with neurodegenerative or psychiatric disorders. This article reviews the BDNF gene structure, transport, secretion, expression and functions in the brain. This article also implicates BDNF in several brain-related disorders, including Alzheimer's disease, Parkinson's disease, Huntington's disease, amyotrophic lateral sclerosis, major depressive disorder, schizophrenia, epilepsy and bipolar disorder.
Collapse
Affiliation(s)
- Veerta Sharma
- Chitkara College of Pharmacy, Chitkara University, 140401, Rajpura, Punjab, India
| | - Thakur Gurjeet Singh
- Chitkara College of Pharmacy, Chitkara University, 140401, Rajpura, Punjab, India.
| | - Amarjot Kaur
- Chitkara College of Pharmacy, Chitkara University, 140401, Rajpura, Punjab, India
| | - Ashi Mannan
- Chitkara College of Pharmacy, Chitkara University, 140401, Rajpura, Punjab, India
| | - Sonia Dhiman
- Chitkara College of Pharmacy, Chitkara University, 140401, Rajpura, Punjab, India
| |
Collapse
|
5
|
mTOR and HDAC2 are simultaneously activated during electrically induced kindling of seizures. Epilepsy Res 2022; 185:106991. [DOI: 10.1016/j.eplepsyres.2022.106991] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Revised: 07/27/2022] [Accepted: 07/28/2022] [Indexed: 11/23/2022]
|
6
|
Clarke E, Stocki P, Sinclair EH, Gauhar A, Fletcher EJR, Krawczun-Rygmaczewska A, Duty S, Walsh FS, Doherty P, Rutkowski JL. A Single Domain Shark Antibody Targeting the Transferrin Receptor 1 Delivers a TrkB Agonist Antibody to the Brain and Provides Full Neuroprotection in a Mouse Model of Parkinson’s Disease. Pharmaceutics 2022; 14:pharmaceutics14071335. [PMID: 35890231 PMCID: PMC9318160 DOI: 10.3390/pharmaceutics14071335] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Revised: 06/20/2022] [Accepted: 06/22/2022] [Indexed: 02/04/2023] Open
Abstract
Single domain shark antibodies that bind to the transferrin receptor 1 (TfR1) on brain endothelial cells have been used to shuttle antibodies and other cargos across the blood brain barrier (BBB) to the brain. For these studies the TXB4 brain shuttle was fused to a TrkB neurotrophin receptor agonist antibody. The TXB4-TrkB fusion retained potent agonist activity at its cognate receptor and after systemic administration showed a 12-fold increase in brain levels over the unmodified antibody. Only the TXB4-TrkB antibody fusion was detected within the brain and localized to TrkB positive cells in the cortex and tyrosine hydroxylase (TH) positive dopaminergic neurons in the substantia nigra pars compacta (SNc), where it was associated with activated ERK1/2 signaling. When tested in the 6-hydroxydopamine (6-OHDA) mouse model of Parkinson’s disease (PD), TXB4-TrkB, but not the unmodified antibody, completely prevented the 6-OHDA induced death of TH positive neurons in the SNc. In conclusion, the fusion of the TXB4 brain shuttle allows a TrkB agonist antibody to reach neuroprotective concentrations in the brain parenchyma following systemic administration.
Collapse
Affiliation(s)
- Emily Clarke
- King’s College London, Institute of Psychiatry, Psychology and Neuroscience, Wolfson Centre for Age-Related Disease, Guy’s Campus, London SE1 1UL, UK; (E.C.); (E.J.R.F.); (A.K.-R.); (S.D.); (P.D.)
| | - Pawel Stocki
- Ossianix, Inc., Gunnels Wood Rd., Stevenage SG1 2FX, UK; (P.S.); (E.H.S.); (A.G.); (F.S.W.)
| | - Elizabeth H. Sinclair
- Ossianix, Inc., Gunnels Wood Rd., Stevenage SG1 2FX, UK; (P.S.); (E.H.S.); (A.G.); (F.S.W.)
| | - Aziz Gauhar
- Ossianix, Inc., Gunnels Wood Rd., Stevenage SG1 2FX, UK; (P.S.); (E.H.S.); (A.G.); (F.S.W.)
| | - Edward J. R. Fletcher
- King’s College London, Institute of Psychiatry, Psychology and Neuroscience, Wolfson Centre for Age-Related Disease, Guy’s Campus, London SE1 1UL, UK; (E.C.); (E.J.R.F.); (A.K.-R.); (S.D.); (P.D.)
| | - Alicja Krawczun-Rygmaczewska
- King’s College London, Institute of Psychiatry, Psychology and Neuroscience, Wolfson Centre for Age-Related Disease, Guy’s Campus, London SE1 1UL, UK; (E.C.); (E.J.R.F.); (A.K.-R.); (S.D.); (P.D.)
| | - Susan Duty
- King’s College London, Institute of Psychiatry, Psychology and Neuroscience, Wolfson Centre for Age-Related Disease, Guy’s Campus, London SE1 1UL, UK; (E.C.); (E.J.R.F.); (A.K.-R.); (S.D.); (P.D.)
| | - Frank S. Walsh
- Ossianix, Inc., Gunnels Wood Rd., Stevenage SG1 2FX, UK; (P.S.); (E.H.S.); (A.G.); (F.S.W.)
| | - Patrick Doherty
- King’s College London, Institute of Psychiatry, Psychology and Neuroscience, Wolfson Centre for Age-Related Disease, Guy’s Campus, London SE1 1UL, UK; (E.C.); (E.J.R.F.); (A.K.-R.); (S.D.); (P.D.)
| | - Julia Lynn Rutkowski
- Ossianix, Inc., Gunnels Wood Rd., Stevenage SG1 2FX, UK; (P.S.); (E.H.S.); (A.G.); (F.S.W.)
- Correspondence: ; Tel.: +1-(610)-291-1724
| |
Collapse
|
7
|
Ateaque S, Merkouris S, Wyatt S, Allen ND, Xie J, DiStefano PS, Lindsay RM, Barde YA. Selective activation and down-regulation of Trk receptors by neurotrophins in human neurons co-expressing TrkB and TrkC. J Neurochem 2022; 161:463-477. [PMID: 35536742 PMCID: PMC9321069 DOI: 10.1111/jnc.15617] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Revised: 03/22/2022] [Accepted: 04/18/2022] [Indexed: 11/30/2022]
Abstract
In the central nervous system, most neurons co-express TrkB and TrkC, the tyrosine kinase receptors for brain-derived neurotrophic factor (BDNF) and neurotrophin-3 (NT3). As NT3 can also activate TrkB, it has been difficult to understand how NT3 and TrkC can exert unique roles in the assembly of neuronal circuits. Using neurons differentiated from human embryonic stem cells expressing both TrkB and TrkC, we compared Trk activation by BDNF and NT3. To avoid the complications resulting from TrkB activation by NT3, we also generated neurons from stem cells engineered to lack TrkB. We found that NT3 activates TrkC at concentrations lower than those of BDNF needed to activate TrkB. Downstream of Trk activation, the changes in gene expression caused by TrkC activation were found to be similar to those resulting from TrkB activation by BDNF, including a number of genes involved in synaptic plasticity. At high NT3 concentrations, receptor selectivity was lost as a result of TrkB activation. In addition, TrkC was down-regulated, as was also the case with TrkB at high BDNF concentrations. By contrast, receptor selectivity as well as reactivation were preserved when neurons were exposed to low neurotrophin concentrations. These results indicate that the selectivity of NT3/TrkC signalling can be explained by the ability of NT3 to activate TrkC at concentrations lower than those needed to activate TrkB. They also suggest that in a therapeutic perspective, the dosage of Trk receptor agonists will need to be taken into account if prolonged receptor activation is to be achieved.
Collapse
Affiliation(s)
- Sarah Ateaque
- School of Biosciences, Cardiff University, Cardiff, Wales, UK
| | | | - Sean Wyatt
- School of Biosciences, Cardiff University, Cardiff, Wales, UK
| | | | - Jia Xie
- The Scripps Research Institute, La Jolla, California, USA
| | | | | | | |
Collapse
|
8
|
Tessarollo L, Yanpallewar S. TrkB Truncated Isoform Receptors as Transducers and Determinants of BDNF Functions. Front Neurosci 2022; 16:847572. [PMID: 35321093 PMCID: PMC8934854 DOI: 10.3389/fnins.2022.847572] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2022] [Accepted: 02/10/2022] [Indexed: 11/24/2022] Open
Abstract
Brain-derived neurotrophic factor (BDNF) belongs to the neurotrophin family of secreted growth factors and binds with high affinity to the TrkB tyrosine kinase receptors. BDNF is a critical player in the development of the central (CNS) and peripheral (PNS) nervous system of vertebrates and its strong pro-survival function on neurons has attracted great interest as a potential therapeutic target for the management of neurodegenerative disorders such as Amyotrophic Lateral Sclerosis (ALS), Huntington, Parkinson's and Alzheimer's disease. The TrkB gene, in addition to the full-length receptor, encodes a number of isoforms, including some lacking the catalytic tyrosine kinase domain. Importantly, one of these truncated isoforms, namely TrkB.T1, is the most widely expressed TrkB receptor in the adult suggesting an important role in the regulation of BDNF signaling. Although some progress has been made, the mechanism of TrkB.T1 function is still largely unknown. Here we critically review the current knowledge on TrkB.T1 distribution and functions that may be helpful to our understanding of how it regulates and participates in BDNF signaling in normal physiological and pathological conditions.
Collapse
Affiliation(s)
- Lino Tessarollo
- Neural Development Section, Mouse Cancer Genetics Program, Center for Cancer Research, National Cancer Institute, Frederick, MD, United States
| | | |
Collapse
|
9
|
Rousset F, Schmidbauer D, Fink S, Adel Y, Obexer B, Müller M, Glueckert R, Löwenheim H, Senn P. Phoenix auditory neurons as 3R cell model for high throughput screening of neurogenic compounds. Hear Res 2021; 414:108391. [PMID: 34844170 DOI: 10.1016/j.heares.2021.108391] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Revised: 10/25/2021] [Accepted: 10/27/2021] [Indexed: 11/25/2022]
Abstract
Auditory neurons connect the sensory hair cells from the inner ear to the brainstem. These bipolar neurons are relevant targets for pharmacological intervention aiming at protecting or improving the hearing function in various forms of sensorineural hearing loss. In the research laboratory, neurotrophic compounds are commonly used to improve survival and to promote regeneration of auditory neurons. One important roadblock delaying eventual clinical applications of these strategies in humans is the lack of powerful in vitro models allowing high throughput screening of otoprotective and regenerative compounds. The recently discovered auditory neuroprogenitors (ANPGs) derived from the A/J mouse with an unprecedented capacity to self-renew and to provide mature auditory neurons offer the possibility to overcome this bottleneck. In the present study, we further characterized the new phoenix ANPGs model and compared it to the current gold-standard spiral ganglion organotypic explant (SGE) model to assay neurite outgrowth, neurite length and glutamate-induced Ca2+ response in response to neurotrophin-3 (NT-3) and brain derived neurotrophic factor (BDNF) treatment. Whereas both, SGEs and phoenix ANPGs exhibited a robust and sensitive response to neurotrophins, the phoenix ANPGs offer a considerable range of advantages including high throughput suitability, lower experimental variability, single cell resolution and an important reduction of animal numbers. The phoenix ANPGs in vitro model therefore provides a robust high-throughput platform to screen for otoprotective and regenerative neurotrophic compounds in line with 3R principles and is of interest for the field of auditory neuroscience.
Collapse
Affiliation(s)
- Francis Rousset
- The Inner Ear & Olfaction Lab, Department of Pathology and Immunology, Faculty of Medicine, University of Geneva, Switzerland.
| | - Dominik Schmidbauer
- Inner Ear Laboratory, Department of Otolaryngology, Medical University of Innsbruck, Austria
| | - Stefan Fink
- Translational Hearing Research, Tübingen Hearing Research Center, Department of Otolaryngology, Head & Neck Surgery, University of Tübingen, Germany
| | - Youssef Adel
- Translational Hearing Research, Tübingen Hearing Research Center, Department of Otolaryngology, Head & Neck Surgery, University of Tübingen, Germany
| | - Benjamin Obexer
- Inner Ear Laboratory, Department of Otolaryngology, Medical University of Innsbruck, Austria
| | - Marcus Müller
- Translational Hearing Research, Tübingen Hearing Research Center, Department of Otolaryngology, Head & Neck Surgery, University of Tübingen, Germany
| | - Rudolf Glueckert
- Inner Ear Laboratory, Department of Otolaryngology, Medical University of Innsbruck, Austria.
| | - Hubert Löwenheim
- Translational Hearing Research, Tübingen Hearing Research Center, Department of Otolaryngology, Head & Neck Surgery, University of Tübingen, Germany
| | - Pascal Senn
- The Inner Ear & Olfaction Lab, Department of Pathology and Immunology, Faculty of Medicine, University of Geneva, Switzerland; Department of Clinical Neurosciences, Service of ORL & Head and Neck Surgery, University Hospital of Geneva, Switzerland
| |
Collapse
|
10
|
NGF-Dependent and BDNF-Dependent DRG Sensory Neurons Deploy Distinct Degenerative Signaling Mechanisms. eNeuro 2021; 8:ENEURO.0277-20.2020. [PMID: 33372032 PMCID: PMC7877462 DOI: 10.1523/eneuro.0277-20.2020] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2020] [Revised: 12/03/2020] [Accepted: 12/07/2020] [Indexed: 02/07/2023] Open
Abstract
The nerve growth factor (NGF) and brain-derived neurotrophic factor (BDNF) are trophic factors required by distinct population of sensory neurons during development of the nervous system. Neurons that fail to receive appropriate trophic support are lost during this period of naturally occurring cell death. In the last decade, our understanding of the signaling pathways regulating neuronal death following NGF deprivation has advanced substantially. However, the signaling mechanisms promoting BDNF deprivation-induced sensory neuron degeneration are largely unknown. Using a well-established in vitro culture model of dorsal root ganglion (DRG), we have examined degeneration mechanisms triggered on BDNF withdrawal in sensory neurons. Our results indicate differences and similarities between the molecular signaling pathways behind NGF and BDNF deprivation-induced death. For instance, we observed that the inhibition of Trk receptors (K252a), PKC (Gö6976), protein translation (cycloheximide; CHX), or caspases (zVAD-fmk) provides protection from NGF deprivation-induced death but not from degeneration evoked by BDNF-withdrawal. Interestingly, degeneration of BDNF-dependent sensory neurons requires BAX and appears to rely on reactive oxygen species (ROS) generation rather than caspases to induce degeneration. These results highlight the complexity and divergence of mechanisms regulating developmental sensory neuron death.
Collapse
|
11
|
Yousof SM, Awad YM, Mostafa EMA, Hosny MM, Anwar MM, Eldesouki RE, Badawy AE. The potential neuroprotective role of Amphora coffeaeformis algae against monosodium glutamate-induced neurotoxicity in adult albino rats. Food Funct 2021; 12:706-716. [PMID: 33337454 DOI: 10.1039/d0fo01957g] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Monosodium glutamate (MSG) is a neurotoxin found in most processed and infant formulas. Amphora coffeaeformis (AC) has neuroprotective properties. We investigated, for the first time, the potential neuroprotective role of AC on MSG-induced neurotoxicity in brain using a unique procedural approach. The AC extract was characterized via high performance liquid chromatography (HPLC). Animals were assigned into six groups; a control group, low dose MSG (LD-MSG), high dose MSG (HD-MSG), combined groups (LD-MSG + AC) (HD-MSG + AC) and AC only group for eight weeks. Assessment of the cognitive and mood domains was done via Barnes maze and an open field. Gene expression of Bdnf, TrkB, NMDA-B2 and mGlur5 in the hippocampus was obtained via real-time PCR. The hippocampi of the animals were assessed for structural changes. Oxidative stress was assessed in the cerebrum. The results revealed that omega-6 and β-coumaric acid represented the highest percentage among the constituents in the AC extract. The NO level was decreased in the LD-MSG + AC compared to LD-MSG. SOD was diminished in both treated groups compared to the untreated group. HD-MSG + AC exhibited an increase in the number of wrongly visited quadrants compared to the HD-MSG group. HD-MSG + AC showed decreased anxiety-like behavior compared to HD-MSG. LD-MSG + AC and AC groups revealed enhanced anxiety-like behavior. HD-MSG + AC showed under expressed NMDA-B2 and over expressed Bdnf and TrkB genes, compared to HD-MSG. LD-MSG + AC revealed under expression of Bdnf gene compared to LD-MSG. The AC group revealed under expressed TrkB gene compared to the control group. Overall, the results refer to the potential neuroprotective properties of AC alga against MSG neurotoxicity.
Collapse
Affiliation(s)
- Shimaa Mohammad Yousof
- Department of Medical Physiology, Faculty of Medicine, Suez Canal University, Ismailia, Egypt.
| | | | | | | | | | | | | |
Collapse
|
12
|
Fudalej E, Justyniarska M, Kasarełło K, Dziedziak J, Szaflik JP, Cudnoch-Jędrzejewska A. Neuroprotective Factors of the Retina and Their Role in Promoting Survival of Retinal Ganglion Cells: A Review. Ophthalmic Res 2021; 64:345-355. [PMID: 33454713 DOI: 10.1159/000514441] [Citation(s) in RCA: 61] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2020] [Accepted: 01/09/2021] [Indexed: 11/19/2022]
Abstract
Retinal ganglion cells (RGCs) play a crucial role in the visual pathway. As their axons form the optic nerve, apoptosis of these cells causes neurodegenerative vision loss. RGC death could be triggered by increased intraocular pressure, advanced glycation end products, or mitochondrial dysfunction. In this review, we summarize the role of some neuroprotective factors in RGC injury: ciliary neurotrophic factor (CNTF), nerve growth factor (NGF), brain-derived neurotrophic factor, vascular endothelial growth factor, pigment epithelium-derived factor, glial cell line-derived neurotrophic factor, and Norrin. Each, in their own unique way, prevents RGC damage caused by glaucoma, ocular hypertension, ischemic neuropathy, and even oxygen-induced retinopathy. These factors are produced mainly by neurons, leukocytes, glial cells, and epithelial cells. Neuroprotective factors act via various signaling pathways, including JAK/STAT, MAPK, TrkA, and TrkB, which promotes RGC survival. Many attempts have been made to develop therapeutic strategies using these factors. There are ongoing clinical trials with CNTF and NGF, but they have not yet been accepted for clinical use.
Collapse
Affiliation(s)
- Ewa Fudalej
- Department of Experimental and Clinical Physiology, Center for Preclinical Research, Medical University of Warsaw, Warsaw, Poland
| | - Magdalena Justyniarska
- Department of Experimental and Clinical Physiology, Center for Preclinical Research, Medical University of Warsaw, Warsaw, Poland
| | - Kaja Kasarełło
- Department of Experimental and Clinical Physiology, Center for Preclinical Research, Medical University of Warsaw, Warsaw, Poland,
| | - Jacek Dziedziak
- Department of Experimental and Clinical Physiology, Center for Preclinical Research, Medical University of Warsaw, Warsaw, Poland.,Department of Ophthalmology, SPKSO Ophthalmic University Hospital, Medical University of Warsaw, Warsaw, Poland
| | - Jacek P Szaflik
- Department of Ophthalmology, SPKSO Ophthalmic University Hospital, Medical University of Warsaw, Warsaw, Poland
| | - Agnieszka Cudnoch-Jędrzejewska
- Department of Experimental and Clinical Physiology, Center for Preclinical Research, Medical University of Warsaw, Warsaw, Poland
| |
Collapse
|
13
|
Sidorova YA, Saarma M. Can Growth Factors Cure Parkinson's Disease? Trends Pharmacol Sci 2020; 41:909-922. [PMID: 33198924 DOI: 10.1016/j.tips.2020.09.010] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2020] [Revised: 09/25/2020] [Accepted: 09/30/2020] [Indexed: 01/03/2023]
Abstract
Growth factors (GFs) hold considerable promise for disease modification in neurodegenerative disorders because they can protect and restore degenerating neurons and also enhance their functional activity. However, extensive efforts applied to utilize their therapeutic potential in humans have achieved limited success so far. Multiple clinical trials with GFs were performed in Parkinson's disease (PD) patients, in whom diagnostic symptoms of the disease are caused by advanced degeneration of nigrostriatal dopamine neurons (DNs), but the results of these trials are controversial. This review discusses recent developments in the field of therapeutic use of GFs, problems and obstacles related to this use, suggests the ways to overcome these issues, and alternative approaches that can be used to utilize the potential ofGFsin PD management.
Collapse
Affiliation(s)
- Yulia A Sidorova
- Institute of Biotechnology, HiLIFE, University of Helsinki, Helsinki, Finland
| | - Mart Saarma
- Institute of Biotechnology, HiLIFE, University of Helsinki, Helsinki, Finland.
| |
Collapse
|
14
|
Wójcik-Gryciuk A, Gajewska-Woźniak O, Kordecka K, Boguszewski PM, Waleszczyk W, Skup M. Neuroprotection of Retinal Ganglion Cells with AAV2-BDNF Pretreatment Restoring Normal TrkB Receptor Protein Levels in Glaucoma. Int J Mol Sci 2020; 21:ijms21176262. [PMID: 32872441 PMCID: PMC7504711 DOI: 10.3390/ijms21176262] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2020] [Revised: 08/26/2020] [Accepted: 08/27/2020] [Indexed: 02/07/2023] Open
Abstract
Intravitreal delivery of brain-derived neurotrophic factor (BDNF) by injection of recombinant protein or by gene therapy can alleviate retinal ganglion cell (RGC) loss after optic nerve injury (ONI) or laser-induced ocular hypertension (OHT). In models of glaucoma, BDNF therapy can delay or halt RGCs loss, but this protection is time-limited. The decreased efficacy of BDNF supplementation has been in part attributed to BDNF TrkB receptor downregulation. However, whether BDNF overexpression causes TrkB downregulation, impairing long-term BDNF signaling in the retina, has not been conclusively proven. After ONI or OHT, when increased retinal BDNF was detected, a concomitant increase, no change or a decrease in TrkB was reported. We examined quantitatively the retinal concentrations of the TrkB protein in relation to BDNF, in a course of adeno-associated viral vector gene therapy (AAV2-BDNF), using a microbead trabecular occlusion model of glaucoma. We show that unilateral glaucoma, with intraocular pressure ( IOP) increased for five weeks, leads to a bilateral decrease of BDNF in the retina at six weeks, accompanied by up to four-fold TrkB upregulation, while a moderate BDNF overexpression in a glaucomatous eye triggers changes that restore normal TrkB concentrations, driving signaling towards long-term RGCs neuroprotection. We conclude that for glaucoma therapy, the careful selection of the appropriate BDNF concentration is the main factor securing the long-term responsiveness of RGCs and the maintenance of normal TrkB levels.
Collapse
Affiliation(s)
- Anna Wójcik-Gryciuk
- Laboratory of Neurobiology of Vision, Nencki Institute of Experimental Biology, 02-093 Warsaw, Poland; (A.W.-G.); (K.K.); (W.W.)
- Mediq Clinic, 05-120 Legionowo, Poland
| | - Olga Gajewska-Woźniak
- Group of Restorative Neurobiology, Nencki Institute of Experimental Biology, 02-093 Warsaw, Poland;
| | - Katarzyna Kordecka
- Laboratory of Neurobiology of Vision, Nencki Institute of Experimental Biology, 02-093 Warsaw, Poland; (A.W.-G.); (K.K.); (W.W.)
| | - Paweł M. Boguszewski
- Laboratory of Behavioral Methods, Nencki Institute of Experimental Biology, 02-093 Warsaw, Poland;
| | - Wioletta Waleszczyk
- Laboratory of Neurobiology of Vision, Nencki Institute of Experimental Biology, 02-093 Warsaw, Poland; (A.W.-G.); (K.K.); (W.W.)
| | - Małgorzata Skup
- Group of Restorative Neurobiology, Nencki Institute of Experimental Biology, 02-093 Warsaw, Poland;
- Correspondence:
| |
Collapse
|
15
|
Current Medical Therapy and Future Trends in the Management of Glaucoma Treatment. J Ophthalmol 2020; 2020:6138132. [PMID: 32774906 PMCID: PMC7391108 DOI: 10.1155/2020/6138132] [Citation(s) in RCA: 54] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2020] [Accepted: 06/29/2020] [Indexed: 01/02/2023] Open
Abstract
Glaucoma is a neurodegenerative disease characterized by progressive loss of retinal ganglion cells and their axons. Lowering of intraocular pressure (IOP) is currently the only proven treatment strategy for glaucoma. However, some patients show progressive loss of visual field and quality of life despite controlled IOP which indicates that other factors are implicated in glaucoma. Therefore, approaches that could prevent or decrease the rate of progression and do not rely on IOP lowering have gained much attention. Effective neuroprotection has been reported in animal models of glaucoma, but till now, no neuroprotective agents have been clinically approved. The present update provides an overview of currently available IOP-lowering medications. Moreover, potential new treatment targets for IOP-lowering and neuroprotective therapy are discussed. Finally, future trends in glaucoma therapy are addressed, including sustained drug delivery systems and progress toward personalized medicine.
Collapse
|
16
|
Therapeutic Potential of Mesenchymal Stem Cells and Their Secretome in the Treatment of Glaucoma. Stem Cells Int 2019; 2019:7869130. [PMID: 31949441 PMCID: PMC6948292 DOI: 10.1155/2019/7869130] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2019] [Accepted: 12/09/2019] [Indexed: 12/15/2022] Open
Abstract
Glaucoma represents a group of progressive optic neuropathies characterized by gradual loss of retinal ganglion cells (RGCs), the neurons that conduct visual information from the retina to the brain. Elevated intraocular pressure (IOP) is considered the main reason for enhanced apoptosis of RGCs in glaucoma. Currently used therapeutic agents are not able to repopulate and/or regenerate injured RGCs and, therefore, are ineffective in most patients with advanced glaucoma. Accordingly, several new therapeutic approaches, including stem cell-based therapy, have been explored for the glaucoma treatment. In this review article, we emphasized current knowledge regarding molecular and cellular mechanisms responsible for beneficial effects of mesenchymal stem cells (MSCs) and their secretome in the treatment of glaucoma. MSCs produce neurotrophins and in an exosome-dependent manner supply injured RGCs with growth factors enhancing their survival and regeneration. Additionally, MSCs are able to generate functional RGC-like cells and induce proliferation of retinal stem cells. By supporting integrity of trabecular meshwork, transplanted MSCs alleviate IOP resulting in reduced loss of RGCs. Moreover, MSCs are able to attenuate T cell-driven retinal inflammation providing protection to the injured retinal tissue. In summing up, due to their capacity for neuroprotection and immunomodulation, MSCs and their secretome could be explored in upcoming clinical studies as new therapeutic agents for glaucoma treatment.
Collapse
|
17
|
Fred SM, Laukkanen L, Brunello CA, Vesa L, Göös H, Cardon I, Moliner R, Maritzen T, Varjosalo M, Casarotto PC, Castrén E. Pharmacologically diverse antidepressants facilitate TRKB receptor activation by disrupting its interaction with the endocytic adaptor complex AP-2. J Biol Chem 2019; 294:18150-18161. [PMID: 31631060 PMCID: PMC6885648 DOI: 10.1074/jbc.ra119.008837] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2019] [Revised: 10/15/2019] [Indexed: 01/19/2023] Open
Abstract
Several antidepressant drugs activate tropomyosin-related kinase B (TRKB) receptor, but it remains unclear whether these compounds employ a common mechanism for TRKB activation. Here, using MS, we found that a single intraperitoneal injection of fluoxetine disrupts the interaction of several proteins with TRKB in the hippocampus of mice. These proteins included members of adaptor protein complex-2 (AP-2) involved in vesicular endocytosis. The interaction of TRKB with the cargo-docking μ subunit of the AP-2 complex (AP2M) was confirmed to be disrupted by both acute and repeated fluoxetine treatments. Of note, fluoxetine disrupted the coupling between full-length TRKB and AP2M, but not the interaction between AP2M and the TRKB C-terminal region, indicating that the fluoxetine-binding site in TRKB lies outside the TRKB:AP2M interface. ELISA experiments revealed that in addition to fluoxetine, other chemically diverse antidepressants, such as imipramine, rolipram, phenelzine, ketamine, and its metabolite 2R,6R-hydroxynorketamine, also decreased the interaction between TRKB and AP2M in vitro Silencing the expression of AP2M in a TRKB-expressing mouse fibroblast cell line (MG87.TRKB) increased cell-surface expression of TRKB and facilitated its activation by brain-derived neurotrophic factor (BDNF), observed as levels of phosphorylated TRKB. Moreover, animals haploinsufficient for the Ap2m1 gene displayed increased levels of active TRKB, along with enhanced cell-surface expression of the receptor in cultured hippocampal neurons. Taken together, our results suggest that disruption of the TRKB:AP2M interaction is a common mechanism underlying TRKB activation by several chemically diverse antidepressants.
Collapse
Affiliation(s)
- Senem Merve Fred
- Neuroscience Center-HiLIFE, University of Helsinki, 00014 Helsinki, Finland
| | - Liina Laukkanen
- Neuroscience Center-HiLIFE, University of Helsinki, 00014 Helsinki, Finland
| | - Cecilia A Brunello
- Neuroscience Center-HiLIFE, University of Helsinki, 00014 Helsinki, Finland
| | - Liisa Vesa
- Neuroscience Center-HiLIFE, University of Helsinki, 00014 Helsinki, Finland
| | - Helka Göös
- Institute of Biotechnology-HiLIFE, University of Helsinki, 00014 Helsinki, Finland
| | - Iseline Cardon
- Brain Master Program, Faculty of Science, Aix-Marseille Université, 13007 Marseille, France
| | - Rafael Moliner
- Neuroscience Center-HiLIFE, University of Helsinki, 00014 Helsinki, Finland
| | - Tanja Maritzen
- Leibniz-Forschungsinstitut für Molekulare Pharmakologie (FMP), 13125 Berlin, Germany
| | - Markku Varjosalo
- Institute of Biotechnology-HiLIFE, University of Helsinki, 00014 Helsinki, Finland
| | - Plinio C Casarotto
- Neuroscience Center-HiLIFE, University of Helsinki, 00014 Helsinki, Finland.
| | - Eero Castrén
- Neuroscience Center-HiLIFE, University of Helsinki, 00014 Helsinki, Finland
| |
Collapse
|
18
|
Szobota S, Mathur PD, Siegel S, Black K, Saragovi HU, Foster AC. BDNF, NT-3 and Trk receptor agonist monoclonal antibodies promote neuron survival, neurite extension, and synapse restoration in rat cochlea ex vivo models relevant for hidden hearing loss. PLoS One 2019; 14:e0224022. [PMID: 31671109 PMCID: PMC6822712 DOI: 10.1371/journal.pone.0224022] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2019] [Accepted: 10/03/2019] [Indexed: 12/21/2022] Open
Abstract
Neurotrophins and their mimetics are potential treatments for hearing disorders because of their trophic effects on spiral ganglion neurons (SGNs) whose connections to hair cells may be compromised in many forms of hearing loss. Studies in noise or ototoxin-exposed animals have shown that local delivery of NT-3 or BDNF has beneficial effects on SGNs and hearing. We evaluated several TrkB or TrkC monoclonal antibody agonists and small molecules, along with BDNF and NT-3, in rat cochlea ex vivo models. The TrkB agonists BDNF and a monoclonal antibody, M3, had the greatest effects on SGN survival, neurite outgrowth and branching. In organotypic cochlear explants, BDNF and M3 enhanced synapse formation between SGNs and inner hair cells and restored these connections after excitotoxin-induced synaptopathy. Loss of these synapses has recently been implicated in hidden hearing loss, a condition characterized by difficulty hearing speech in the presence of background noise. The unique profile of M3 revealed here warrants further investigation, and the broad activity profile of BDNF observed underpins its continued development as a hearing loss therapeutic.
Collapse
Affiliation(s)
- Stephanie Szobota
- Otonomy, Inc., San Diego, California, United States of America
- * E-mail:
| | | | - Sairey Siegel
- Otonomy, Inc., San Diego, California, United States of America
| | | | - H. Uri Saragovi
- Lady Davis Institute-Jewish General Hospital, McGill University, Montreal, Quebec, Canada
| | - Alan C. Foster
- Otonomy, Inc., San Diego, California, United States of America
| |
Collapse
|
19
|
Zanin JP, Montroull LE, Volosin M, Friedman WJ. The p75 Neurotrophin Receptor Facilitates TrkB Signaling and Function in Rat Hippocampal Neurons. Front Cell Neurosci 2019; 13:485. [PMID: 31736712 PMCID: PMC6828739 DOI: 10.3389/fncel.2019.00485] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2019] [Accepted: 10/14/2019] [Indexed: 01/02/2023] Open
Abstract
Neurotrophins activate Trk receptor signaling to support neuronal survival and many aspects of neuronal function. Early studies demonstrated that TrkA formed a complex with the p75 neurotrophin receptor (p75NTR), which increased the affinity and selectivity of NGF binding, however, whether interaction of p75NTR with other Trk receptors performs a similar function to enhance ligand binding has not been demonstrated. We investigated the interaction of TrkB with full length p75NTR in hippocampal neurons in response to BDNF and found that the association of these receptors occurs after ligand binding and requires phosphorylation of TrkB, indicating that formation of this receptor complex was not necessary for ligand binding. Moreover, the interaction of these receptors required internalization and localization to early endosomes. We found that association of TrkB with p75NTR was necessary for optimal downstream signaling of the PI3K-Akt pathway, but not the Erk pathway, in hippocampal neurons. The absence of p75NTR impaired the ability of BDNF to rescue hippocampal neurons in a trophic deprivation model, suggesting that p75NTR facilitates the ability of TrkB to activate specific pathways to promote neuronal survival.
Collapse
Affiliation(s)
- Juan P Zanin
- Department of Biological Sciences, Rutgers University, Newark, NJ, United States
| | - Laura E Montroull
- Department of Biological Sciences, Rutgers University, Newark, NJ, United States
| | - Marta Volosin
- Department of Biological Sciences, Rutgers University, Newark, NJ, United States
| | - Wilma J Friedman
- Department of Biological Sciences, Rutgers University, Newark, NJ, United States
| |
Collapse
|
20
|
Targeting BDNF/TrkB pathways for preventing or suppressing epilepsy. Neuropharmacology 2019; 167:107734. [PMID: 31377199 DOI: 10.1016/j.neuropharm.2019.107734] [Citation(s) in RCA: 63] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2019] [Revised: 07/25/2019] [Accepted: 08/01/2019] [Indexed: 12/15/2022]
Abstract
Traumatic brain injury (TBI) and status epilepticus (SE) have both been linked to development of human epilepsy. Although distinct etiologies, current research has suggested the convergence of molecular mechanisms underlying epileptogenesis following these insults. One such mechanism involves the neurotrophin brain-derived neurotrophic factor (BDNF) and its high-affinity receptor, tropomyosin related kinase B (TrkB). In this review, we focus on currently available data regarding the pathophysiologic role of BDNF/TrkB signaling in epilepsy development. We specifically examine the axonal injury and SE epilepsy models, two animal models that recapitulate many aspects of TBI- and SE-induced epilepsy in humans respectively. Thereafter, we discuss aspiring strategies for targeting BDNF/TrkB signaling so as to prevent epilepsy following an insult or suppress its expression once developed. This article is part of the special issue entitled 'New Epilepsy Therapies for the 21st Century - From Antiseizure Drugs to Prevention, Modification and Cure of Epilepsy'.
Collapse
|
21
|
Dedoni S, Marras L, Olianas MC, Ingianni A, Onali P. Downregulation of TrkB Expression and Signaling by Valproic Acid and Other Histone Deacetylase Inhibitors. J Pharmacol Exp Ther 2019; 370:490-503. [PMID: 31308194 DOI: 10.1124/jpet.119.258129] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2019] [Accepted: 06/14/2019] [Indexed: 01/27/2023] Open
Abstract
Valproic acid (VPA) has been shown to regulate the levels of brain-derived neurotrophic factor (BDNF), but it is not known whether this drug can affect the neuronal responses to BDNF. In the present study, we show that in retinoic acid-differentiated SH-SY5Y human neuroblastoma cells, prolonged exposure to VPA reduces the expression of the BDNF receptor TrkB at the protein and mRNA levels and inhibits the intracellular signaling, neurotrophic activity, and prosurvival function of BDNF. VPA downregulates TrkB and curtails BDNF-induced signaling also in differentiated Kelly and LAN-1 neuroblastoma cells and primary mouse cortical neurons. The VPA effect is mimicked by several histone deacetylase (HDAC) inhibitors, including the class I HDAC inhibitors entinostat and romidepsin. Conversely, the class II HDAC inhibitor MC1568, the HDAC6 inhibitor tubacin, the HDAC8 inhibitor PCI-34051, and the VPA derivative valpromide have no effect. In neuroblastoma cells and primary neurons both VPA and entinostat increase the cellular levels of the transcription factor RUNX3, which negatively regulates TrkB gene expression. Treatment with RUNX3 siRNA attenuates VPA-induced RUNX3 elevation and TrkB downregulation. VPA, entinostat, HDAC1 depletion by siRNA, and 3-deazaneplanocin A (DZNep), an inhibitor of the polycomb repressor complex 2 (PRC2), decrease the PRC2 core component EZH2, a RUNX3 suppressor. Like VPA, HDAC1 depletion and DZNep increase RUNX3 and decrease TrkB expression. These results indicate that VPA downregulates TrkB through epigenetic mechanisms involving the EZH2/RUNX3 axis and provide evidence that this effect implicates relevant consequences with regard to BDNF efficacy in stimulating intracellular signaling and functional responses. SIGNIFICANCE STATEMENT: The tropomyosin-related kinase receptor B (TrkB) mediates the stimulatory effects of brain-derived neurotrophic factor (BDNF) on neuronal growth, differentiation, and survival and is highly expressed in aggressive neuroblastoma and other tumors. Here we show that exposure to valproic acid (VPA) downregulates TrkB expression and functional activity in retinoic acid-differentiated human neuroblastoma cell lines and primary mouse cortical neurons. The effects of VPA are mimicked by other histone deacetylase (HDAC) inhibitors and HDAC1 knockdown and appear to be mediated by an epigenetic mechanism involving the upregulation of RUNX3, a suppressor of TrkB gene expression. TrkB downregulation may have relevance for the use of VPA as a potential therapeutic agent in neuroblastoma and other pathologies characterized by an excessive BDNF/TrkB signaling.
Collapse
Affiliation(s)
- Simona Dedoni
- Laboratory of Cellular and Molecular Pharmacology, Section of Neurosciences, Department of Biomedical Sciences (S.D., M.C.O., P.O.) and Section of Microbiology, Department of Biomedical Sciences (L.M., A.I.), University of Cagliari, Cagliari, Italy
| | - Luisa Marras
- Laboratory of Cellular and Molecular Pharmacology, Section of Neurosciences, Department of Biomedical Sciences (S.D., M.C.O., P.O.) and Section of Microbiology, Department of Biomedical Sciences (L.M., A.I.), University of Cagliari, Cagliari, Italy
| | - Maria C Olianas
- Laboratory of Cellular and Molecular Pharmacology, Section of Neurosciences, Department of Biomedical Sciences (S.D., M.C.O., P.O.) and Section of Microbiology, Department of Biomedical Sciences (L.M., A.I.), University of Cagliari, Cagliari, Italy
| | - Angela Ingianni
- Laboratory of Cellular and Molecular Pharmacology, Section of Neurosciences, Department of Biomedical Sciences (S.D., M.C.O., P.O.) and Section of Microbiology, Department of Biomedical Sciences (L.M., A.I.), University of Cagliari, Cagliari, Italy
| | - Pierluigi Onali
- Laboratory of Cellular and Molecular Pharmacology, Section of Neurosciences, Department of Biomedical Sciences (S.D., M.C.O., P.O.) and Section of Microbiology, Department of Biomedical Sciences (L.M., A.I.), University of Cagliari, Cagliari, Italy
| |
Collapse
|
22
|
Lucas-Ruiz F, Galindo-Romero C, García-Bernal D, Norte-Muñoz M, Rodríguez-Ramírez KT, Salinas-Navarro M, Millán-Rivero JE, Vidal-Sanz M, Agudo-Barriuso M. Mesenchymal stromal cell therapy for damaged retinal ganglion cells, is gold all that glitters? Neural Regen Res 2019; 14:1851-1857. [PMID: 31290434 PMCID: PMC6676874 DOI: 10.4103/1673-5374.259601] [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] [Indexed: 01/06/2023] Open
Abstract
Mesenchymal stromal cells are an excellent source of stem cells because they are isolated from adult tissues or perinatal derivatives, avoiding the ethical concerns that encumber embryonic stem cells. In preclinical models, it has been shown that mesenchymal stromal cells have neuroprotective and immunomodulatory properties, both of which are ideal for central nervous system treatment and repair. Here we will review the current literature on mesenchymal stromal cells, focusing on bone marrow mesenchymal stromal cells, adipose-derived mesenchymal stromal cells and mesenchymal stromal cells from the umbilical cord stroma, i.e., Wharton's jelly mesenchymal stromal cells. Finally, we will discuss the use of these cells to alleviate retinal ganglion cell degeneration following axonal trauma.
Collapse
Affiliation(s)
- Fernando Lucas-Ruiz
- Grupo de Oftalmología Experimental, Instituto Murciano de Investigación Biosanitaria Virgen de la Arrixaca (IMIB-Arrixaca); Department of Talmología, Universidad de Murcia, Murcia, Spain
| | - Caridad Galindo-Romero
- Grupo de Oftalmología Experimental, Instituto Murciano de Investigación Biosanitaria Virgen de la Arrixaca (IMIB-Arrixaca); Department of Talmología, Universidad de Murcia, Murcia, Spain
| | - David García-Bernal
- Unidad de Terapia Celular y Trasplante Hematopoyético, Instituto Murciano de Investigación Biosanitaria Virgen de la Arrixaca (IMIB-Arrixaca); Department of Medicina Interna, Universidad de Murcia, Murcia, Spain
| | - María Norte-Muñoz
- Grupo de Oftalmología Experimental, Instituto Murciano de Investigación Biosanitaria Virgen de la Arrixaca (IMIB-Arrixaca); Department of Talmología, Universidad de Murcia, Murcia, Spain
| | - Kristy T Rodríguez-Ramírez
- Grupo de Oftalmología Experimental, Instituto Murciano de Investigación Biosanitaria Virgen de la Arrixaca (IMIB-Arrixaca); Department of Talmología, Universidad de Murcia, Murcia, Spain
| | - Manuel Salinas-Navarro
- Grupo de Oftalmología Experimental, Instituto Murciano de Investigación Biosanitaria Virgen de la Arrixaca (IMIB-Arrixaca); Department of Talmología, Universidad de Murcia, Murcia, Spain
| | - Jose E Millán-Rivero
- Unidad de Terapia Celular y Trasplante Hematopoyético, Instituto Murciano de Investigación Biosanitaria Virgen de la Arrixaca (IMIB-Arrixaca); Department of Medicina Interna, Universidad de Murcia, Murcia, Spain
| | - Manuel Vidal-Sanz
- Grupo de Oftalmología Experimental, Instituto Murciano de Investigación Biosanitaria Virgen de la Arrixaca (IMIB-Arrixaca); Department of Talmología, Universidad de Murcia, Murcia, Spain
| | - Marta Agudo-Barriuso
- Grupo de Oftalmología Experimental, Instituto Murciano de Investigación Biosanitaria Virgen de la Arrixaca (IMIB-Arrixaca); Department of Talmología, Universidad de Murcia, Murcia, Spain
| |
Collapse
|
23
|
Osborne A, Khatib TZ, Songra L, Barber AC, Hall K, Kong GYX, Widdowson PS, Martin KR. Neuroprotection of retinal ganglion cells by a novel gene therapy construct that achieves sustained enhancement of brain-derived neurotrophic factor/tropomyosin-related kinase receptor-B signaling. Cell Death Dis 2018; 9:1007. [PMID: 30258047 PMCID: PMC6158290 DOI: 10.1038/s41419-018-1041-8] [Citation(s) in RCA: 106] [Impact Index Per Article: 15.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2018] [Revised: 09/04/2018] [Accepted: 09/07/2018] [Indexed: 01/17/2023]
Abstract
Previous studies have demonstrated that intravitreal delivery of brain-derived neurotrophic factor (BDNF) by injection of recombinant protein or by gene therapy can alleviate retinal ganglion cell (RGC) loss after optic nerve injury. BDNF gene therapy can improve RGC survival in experimental models of glaucoma, the leading cause of irreversible blindness worldwide. However, the therapeutic efficacy of BDNF supplementation alone is time limited at least in part due to BDNF receptor downregulation. Tropomyosin-related receptor kinase-B (TrkB) downregulation has been reported in many neurological diseases including glaucoma, potentially limiting the effect of sustained or repeated BDNF delivery. Here, we characterize a novel adeno-associated virus (AAV) gene therapy (AAV2 TrkB-2A-mBDNF) that not only increases BDNF production but also improves long-term neuroprotective signaling by increasing expression of the BDNF receptor (TrkB) within the inner retina. This approach leads to significant and sustained elevation of survival signaling pathways ERK and AKT within RGCs over 6 months and avoids the receptor downregulation which we observe with treatment with AAV2 BDNF alone. We validate the neuroprotective efficacy of AAV2 TrkB-2A-mBDNF in a mouse model of optic nerve injury, where it outperforms conventional AAV2 BDNF or AAV2 TrkB therapy, before showing powerful proof of concept neuroprotection of RGCs and axons in a rat model of chronic intraocular pressure (IOP) elevation. We also show that there are no adverse effects of the vector on retinal structure or function as assessed by histology and electroretinography in young or aged animals. Further studies are underway to explore the potential of this vector as a candidate for progression into clinical studies to protect RGCs in patients with glaucoma and progressive visual loss despite conventional IOP-lowering treatment.
Collapse
Affiliation(s)
- Andrew Osborne
- John van Geest Centre for Brain Repair, Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK.,Quethera Ltd, Babraham Research Campus, Cambridge, UK
| | - Tasneem Z Khatib
- John van Geest Centre for Brain Repair, Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK.,Eye Department, Addenbrooke's Hospital, Cambridge, UK
| | - Lalana Songra
- John van Geest Centre for Brain Repair, Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK
| | - Amanda C Barber
- John van Geest Centre for Brain Repair, Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK
| | - Katie Hall
- John van Geest Centre for Brain Repair, Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK
| | - George Y X Kong
- Centre for Eye Research Australia, Royal Victorian Eye and Ear Hospital, Melbourne, Australia.,University of Melbourne, Melbourne, Australia
| | | | - Keith R Martin
- John van Geest Centre for Brain Repair, Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK. .,Quethera Ltd, Babraham Research Campus, Cambridge, UK. .,Eye Department, Addenbrooke's Hospital, Cambridge, UK. .,Cambridge NIHR Biomedical Research Centre, Cambridge, UK. .,Wellcome Trust-MRC Cambridge Stem Cell Institute, University of Cambridge, Cambridge, UK.
| |
Collapse
|
24
|
Ahmed E, Tawfik MK, Essawy SS, Ahmed AS, Hermans E. Cysteamine Potentiates the Anti-Depressive Effects of Venlafaxine in Corticosterone-Induced Anxiety/Depression Mouse Model: Effect on Brain-Derived Neurotrophic Factor and Tropomyosin-Related Kinase B. EGYPTIAN JOURNAL OF BASIC AND CLINICAL PHARMACOLOGY 2018. [DOI: 10.11131/2018/101383] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023] Open
Affiliation(s)
- Eman Ahmed
- Department of Clinical Pharmacology, Faculty of Medicine, Suez Canal University, Ismailia, Egypt
- Neuropharmacology Group, Institute of Neuroscience, Université Catholique de Louvain, Brussels, Belgium
| | - Mona K. Tawfik
- Department of Clinical Pharmacology, Faculty of Medicine, Suez Canal University, Ismailia, Egypt
| | - Soha S. Essawy
- Department of Clinical Pharmacology, Faculty of Medicine, Suez Canal University, Ismailia, Egypt
| | - Amal S. Ahmed
- Department of Clinical Pathology, Faculty of Medicine, Suez Canal University, Ismailia, Egypt
| | - Emmanuel Hermans
- Neuropharmacology Group, Institute of Neuroscience, Université Catholique de Louvain, Brussels, Belgium
| |
Collapse
|
25
|
Enomoto S, Shimizu K, Nibuya M, Suzuki E, Nagata K, Kondo T. Activated brain-derived neurotrophic factor/TrkB signaling in rat dorsal and ventral hippocampi following 10-day electroconvulsive seizure treatment. Neurosci Lett 2017; 660:45-50. [DOI: 10.1016/j.neulet.2017.09.011] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2017] [Revised: 08/31/2017] [Accepted: 09/06/2017] [Indexed: 12/15/2022]
|
26
|
Cognitive impairment and gene expression alterations in a rodent model of binge eating disorder. Physiol Behav 2017; 180:78-90. [PMID: 28821448 DOI: 10.1016/j.physbeh.2017.08.004] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2017] [Revised: 08/10/2017] [Accepted: 08/11/2017] [Indexed: 12/21/2022]
Abstract
Binge eating disorder (BED) is defined as recurrent, distressing over-consumption of palatable food (PF) in a short time period. Clinical studies suggest that individuals with BED may have impairments in cognitive processes, executive functioning, impulse control, and decision-making, which may play a role in sustaining binge eating behavior. These clinical reports, however, are limited and often conflicting. In this study, we used a limited access rat model of binge-like behavior in order to further explore the effects of binge eating on cognition. In binge eating prone (BEP) rats, we found novel object recognition (NOR) as well as Barnes maze reversal learning (BM-RL) deficits. Aberrant gene expression of brain derived neurotrophic factor (Bdnf) and tropomyosin receptor kinase B (TrkB) in the hippocampus (HPC)-prefrontal cortex (PFC) network was observed in BEP rats. Additionally, the NOR deficits were correlated with reductions in the expression of TrkB and insulin receptor (Ir) in the CA3 region of the hippocampus. Furthermore, up-regulation of serotonin-2C (5-HT2C) receptors in the orbitoprefrontal cortex (OFC) was associated with BM-RL deficit. Finally, in the nucleus accumbens (NAc), we found decreased dopamine receptor 2 (Drd2) expression among BEP rats. Taken together, these data suggest that binge eating vegetable shortening may induce contextual and reversal learning deficits which may be mediated, at least in part, by the altered expression of genes in the CA3-OFC-NAc neural network.
Collapse
|
27
|
Ubiquitin C-Terminal Hydrolase L1 (UCH-L1) Promotes Hippocampus-Dependent Memory via Its Deubiquitinating Effect on TrkB. J Neurosci 2017; 37:5978-5995. [PMID: 28500221 DOI: 10.1523/jneurosci.3148-16.2017] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2016] [Revised: 04/21/2017] [Accepted: 05/04/2017] [Indexed: 11/21/2022] Open
Abstract
Multiple studies have established that brain-derived neurotrophic factor (BDNF) plays a critical role in the regulation of synaptic plasticity via its receptor, TrkB. In addition to being phosphorylated, TrkB has also been demonstrated to be ubiquitinated. However, the mechanisms of TrkB ubiquitination and its biological functions remain poorly understood. In this study, we demonstrate that ubiquitin C-terminal hydrolase L1 (UCH-L1) promotes contextual fear conditioning learning and memory via the regulation of ubiquitination of TrkB. We provide evidence that UCH-L1 can deubiquitinate TrkB directly. K460 in the juxtamembane domain of TrkB is the primary ubiquitination site and is regulated by UCH-L1. By using a peptide that competitively inhibits the association between UCH-L1 and TrkB, we show that the blockade of UCH-L1-regulated TrkB deubiquitination leads to increased BDNF-induced TrkB internalization and consequently directs the internalized TrkB to the degradation pathway, resulting in increased degradation of surface TrkB and attenuation of TrkB activation and its downstream signaling pathways. Moreover, injection of the peptide into the DG region of mice impairs hippocampus-dependent memory. Together, our results suggest that the ubiquitination of TrkB is a mechanism that controls its downstream signaling pathways via the regulation of its endocytosis and postendocytic trafficking and that UCH-L1 mediates the deubiquitination of TrkB and could be a potential target for the modulation of hippocampus-dependent memory.SIGNIFICANCE STATEMENT Ubiquitin C-terminal hydrolase L1 (UCH-L1) has been demonstrated to play important roles in the regulation of synaptic plasticity and learning and memory. TrkB, the receptor for brain-derived neurotrophic factor, has also been shown to be a potent regulator of synaptic plasticity. In this study, we demonstrate that UCH-L1 functions as a deubiquitinase for TrkB. The blockage of UCH-L1-regulated deubiquitination of TrkB eventually results in the increased degradation of surface TrkB and decreased activation of TrkB and its downstream signaling pathways. In vivo, UCH-L1-regulated TrkB deubiquitination is necessary for hippocampus-dependent memory. Overall, our study provides novel insights into the mechanisms of UCH-L1-mediated neurobiological functions and suggests that ubiquitination is an important regulatory signal for TrkB functions.
Collapse
|
28
|
Grape powder consumption affects the expression of neurodegeneration-related brain proteins in rats chronically fed a high-fructose–high-fat diet. J Nutr Biochem 2017; 43:132-140. [DOI: 10.1016/j.jnutbio.2017.02.013] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2016] [Revised: 01/11/2017] [Accepted: 02/08/2017] [Indexed: 01/19/2023]
|
29
|
Sánchez-Sánchez J, Arévalo JC. A Review on Ubiquitination of Neurotrophin Receptors: Facts and Perspectives. Int J Mol Sci 2017; 18:ijms18030630. [PMID: 28335430 PMCID: PMC5372643 DOI: 10.3390/ijms18030630] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2017] [Revised: 03/07/2017] [Accepted: 03/10/2017] [Indexed: 02/04/2023] Open
Abstract
Ubiquitination is a reversible post-translational modification involved in a plethora of different physiological functions. Among the substrates that are ubiquitinated, neurotrophin receptors (TrkA, TrkB, TrkC, and p75NTR) have been studied recently. TrkA is the most studied receptor in terms of its ubiquitination, and different E3 ubiquitin ligases and deubiquitinases have been implicated in its ubiquitination, whereas not much is known about the other neurotrophin receptors aside from their ubiquitination. Additional studies are needed that focus on the ubiquitination of TrkB, TrkC, and p75NTR in order to further understand the role of ubiquitination in their physiological and pathological functions. Here we review what is currently known regarding the ubiquitination of neurotrophin receptors and its physiological and pathological relevance.
Collapse
Affiliation(s)
- Julia Sánchez-Sánchez
- Department of Cell Biology and Pathology, Institute of Neuroscience Castile & Leon, University of Salamanca, 37007 Salamanca, Spain.
| | | |
Collapse
|
30
|
Zhao J, Boyd AW, Bartlett PF. The identification of a novel isoform of EphA4 and ITS expression in SOD1 G93A mice. Neuroscience 2017; 347:11-21. [PMID: 28153688 DOI: 10.1016/j.neuroscience.2017.01.038] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2016] [Revised: 01/18/2017] [Accepted: 01/23/2017] [Indexed: 12/12/2022]
Abstract
Amyotrophic lateral sclerosis (ALS) is characterized by the degeneration of motor neurons, leading to progressive muscle atrophy and fatal paralysis. Mutations in more than 20 genes, including full-length EphA4 (EphA4-FL), have been implicated in this pathogenesis. The present study aimed to identify novel isoforms of EphA4-FL and to investigate the expression of EphA4-FL and its isoforms in the superoxide dismutase 1 (SOD1) mutant mouse model of ALS. Two novel transcripts were verified in mouse and humans. In transfected cells, both transcripts could be translated into proteins, which respectively contained the N- and C-termini of EphA4-FL, referred as EphA4-N and EphA4-C. EphA4-N, which was expressed on the surface of transfected cells, was shown to act as a dominant negative receptor by significantly suppressing the activation of EphA4-FL in vitro. The expression of both EphA4-FL and EphA4-N was significantly higher in the nervous tissue of SOD1G93A compared to wild-type mice suggesting that both forms are modulated during the disease process.
Collapse
Affiliation(s)
- Jing Zhao
- Queensland Brain Institute, University of Queensland, Qld 4072, Australia
| | - Andrew W Boyd
- School of Medicine, University of Queensland, Qld 4072, Australia; QIMR Berghofer Medical Research Institute, Qld 4006, Australia
| | - Perry F Bartlett
- Queensland Brain Institute, University of Queensland, Qld 4072, Australia.
| |
Collapse
|
31
|
Ieraci A, Madaio AI, Mallei A, Lee FS, Popoli M. Brain-Derived Neurotrophic Factor Val66Met Human Polymorphism Impairs the Beneficial Exercise-Induced Neurobiological Changes in Mice. Neuropsychopharmacology 2016; 41:3070-3079. [PMID: 27388329 PMCID: PMC5101555 DOI: 10.1038/npp.2016.120] [Citation(s) in RCA: 64] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/03/2016] [Revised: 06/15/2016] [Accepted: 06/27/2016] [Indexed: 12/11/2022]
Abstract
Several studies have shown that exercise improves cognitive functions and emotional behaviors. Positive effects of exercise have been associated with enhanced brain plasticity, adult hippocampal neurogenesis, and increased levels of brain-derived neurotrophic factor (BDNF). However, a substantial variability of individual response to exercise has been described, which may be accounted for by individual genetic variants. Here, we have assessed whether and how the common human BDNF Val66Met polymorphism influences the neurobiological effects modulated by exercise in BDNF Val66Met knock-in male mice. Wild-type (BDNFVal/Val) and homozygous BDNF Val66Met (BDNFMet/Met) male mice were housed in cages equipped with or without running wheels for 4 weeks. Changes in behavioral phenotype, hippocampal adult neurogenesis, and gene expression were evaluated in exercised and sedentary control mice. We found that exercise reduced the latency to feed in the novelty suppressed feeding and the immobility time in the forced swimming test in BDNFVal/Val but not in BDNFMet/Met mice. Hippocampal neurogenesis was reduced in BDNFMet/Met mice compared with BDNFVal/Val mice. BDNFMet/Met mice had lower basal BDNF protein levels in the hippocampus, which was not recovered following exercise. Moreover, exercise-induced expression of total BDNF, BDNF splice variants 1, 2, 4, 6 and fibronectin type III domain-containing protein 5 (FNDC5) mRNA levels were absent or reduced in the dentate gyrus of BDNFMet/Met mice. Exercise failed to enhance PGC-1α and FNDC5 mRNA levels in the BDNFMet/Met muscle. Overall these results indicate that, in adult male mice, the BDNF Val66Met polymorphism impairs the beneficial behavioral and neuroplasticity effects induced by physical exercise.
Collapse
Affiliation(s)
- Alessandro Ieraci
- Laboratory of Neuropsychopharmacology and Functional Neurogenomics, Dipartimento di Scienze Farmacologiche e Biomolecolari and Center of Excellence on Neurodegenerative Diseases, Università di Milano, Milano, Italy
| | - Alessandro I Madaio
- Laboratory of Neuropsychopharmacology and Functional Neurogenomics, Dipartimento di Scienze Farmacologiche e Biomolecolari and Center of Excellence on Neurodegenerative Diseases, Università di Milano, Milano, Italy
| | - Alessandra Mallei
- Laboratory of Neuropsychopharmacology and Functional Neurogenomics, Dipartimento di Scienze Farmacologiche e Biomolecolari and Center of Excellence on Neurodegenerative Diseases, Università di Milano, Milano, Italy
| | - Francis S Lee
- Department of Psychiatry, Weill Medical College of Cornell University, New York, NY, USA
| | - Maurizio Popoli
- Laboratory of Neuropsychopharmacology and Functional Neurogenomics, Dipartimento di Scienze Farmacologiche e Biomolecolari and Center of Excellence on Neurodegenerative Diseases, Università di Milano, Milano, Italy
| |
Collapse
|
32
|
Chang HA, Wang YH, Tung CS, Yeh CB, Liu YP. 7,8-Dihydroxyflavone, a Tropomyosin-Kinase Related Receptor B Agonist, Produces Fast-Onset Antidepressant-Like Effects in Rats Exposed to Chronic Mild Stress. Psychiatry Investig 2016; 13:531-540. [PMID: 27757132 PMCID: PMC5067348 DOI: 10.4306/pi.2016.13.5.531] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/26/2015] [Revised: 12/13/2015] [Accepted: 12/16/2015] [Indexed: 12/14/2022] Open
Abstract
OBJECTIVE Brain-derived neurotrophic factor (BDNF) and its specific receptor, tropomyosin-related kinase (TrkB), play important roles in treating depression. In this experiment, we examined whether 7,8-dihydroxyflavone, a novel potent TrkB agonist, could reverse the behavioral and biochemical abnormalities induced by the chronic mild stress (CMS) paradigm in rats. METHODS SD rats were exposed to a battery of stressors for 56 days. 7,8-dihydroxyflavone (5 and 20 mg/kg) were administered intraperitoneally during the last 28 days of the CMS paradigm. Rats were tested in sucrose consumption test (SCT), forced-swimming test (FST) and elevated T-maze (ETM). Serum corticosterone levels and hippocampal BDNF levels of the rats were measured. RESULTS Four-week CMS on the rats induced their depression-like behavior in SCT. The CMS-reduced sucrose consumption was reversed starting from 7 days after the 7,8-dihydroxyflavone (20 mg/kg) treatment and remained across the subsequent treatment regime. 7,8-dihydroxyflavone, when given at 5 mg/kg for 3 weeks, reduced the immobility time in the FST in the CMS-subjected rats. Additionally, the 4-week treatment with 7,8-dihydroxyflavone (20 mg/kg) attenuated the CMS-induced increase in anxiety-like behavior in the ETM. For the CMS-subjected rats, 7,8-dihydroxyflavone treatment dose-dependently reduced their serum corticosterone levels but increased their hippocampal BDNF levels only at 5 mg/kg. CONCLUSION 7,8-dihydroxyflavone was beneficial for both depression and anxiety-like behaviors, and may exert fast-onset antidepressant effects. This provides a new insight into the pharmacological management of depression.
Collapse
Affiliation(s)
- Hsin-An Chang
- Department of Psychiatry, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Ying-Hsiu Wang
- Department of Physiology and Biophysics, National Defense Medical Center, Taipei, Taiwan
| | - Che-Se Tung
- Department of Physiology and Biophysics, National Defense Medical Center, Taipei, Taiwan
- Division of Medical Research and Education, Cheng Hsin General Hospital, Taipei, Taiwan
| | - Chin-Bin Yeh
- Department of Psychiatry, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Yia-Ping Liu
- Department of Psychiatry, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
- Department of Physiology and Biophysics, National Defense Medical Center, Taipei, Taiwan
| |
Collapse
|
33
|
Proenca CC, Song M, Lee FS. Differential effects of BDNF and neurotrophin 4 (NT4) on endocytic sorting of TrkB receptors. J Neurochem 2016; 138:397-406. [PMID: 27216821 DOI: 10.1111/jnc.13676] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2015] [Revised: 05/03/2016] [Accepted: 05/19/2016] [Indexed: 01/31/2023]
Abstract
Neurotrophins are a family of growth factors playing key roles in the survival, development, and function of neurons. The neurotrophins brain-derived neurotrophic factor (BDNF) and NT4 both bind to and activate TrkB receptors, however, they mediate distinct neuronal functions. The molecular mechanism of how TrkB activation by BDNF and NT4 leads to diverse outcomes is unknown. Here, we report that BDNF and NT4 lead to differential endocytic sorting of TrkB receptors resulting in diverse biological functions in cultured cortical neurons. Fluorescent microscopy and surface biotinylation experiments showed that both neurotrophins stimulate internalization of TrkB with similar kinetics. Exposure to BDNF for 2-3 h reduced the surface pool of TrkB receptors to half, whereas a longer treatment (4-5 h) with NT4 was necessary to achieve a similar level of down-regulation. Although BDNF and NT4 induced TrkB phosphorylation with similar intensities, BDNF induced more rapid ubiquitination and degradation of TrkB than NT4. Interestingly, TrkB receptor ubiquitination by these ligands have substantially different pH sensitivities, resulting in varying degrees of receptor ubiquitination at lower pH levels. Consequently, NT4 was capable of maintaining longer sustained downstream signaling activation that correlated with reduced TrkB ubiquitination at endosomal pH. Thus, by leading to altered endocytic trafficking itineraries for TrkB receptors, BDNF and NT4 elicit differential TrkB signaling in terms of duration, intensity, and specificity, which may contribute to their functional differences in vivo. The neurotrophins, brain-derived neurotrophic factor (BDNF) and neurotrophin-4 (NT4), both bind to and activate TrkB receptors, however, they mediate distinct neuronal functions. Here, we propose that BDNF and NT4 lead to differential endocytic sorting of TrkB receptors resulting in diverse biological functions. BDNF induces more rapid ubiquitination and degradation of TrkB than NT4. Consequently, NT4 is capable of maintaining more sustained signaling downstream of TrkB receptors.
Collapse
Affiliation(s)
- Catia C Proenca
- Developmental and Molecular Pathways, Novartis Institutes for Biomedical Research, Basel, Switzerland
| | - Minseok Song
- Synaptic Circuit Plasticity Laboratory, Department of Structure & Function of Neural Network, Korea Brain Research Institute, 61 Cheomdan-ro, Dong-gu, Daegu, Korea
| | - Francis S Lee
- Department of Psychiatry, Weill Medical College of Cornell University, New York City, New York, USA.,Department of Pharmacology, Weill Medical College of Cornell University, New York City, New York, USA
| |
Collapse
|
34
|
Current Neurogenic and Neuroprotective Strategies to Prevent and Treat Neurodegenerative and Neuropsychiatric Disorders. Neuromolecular Med 2015; 17:404-22. [PMID: 26374113 DOI: 10.1007/s12017-015-8369-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2015] [Accepted: 08/22/2015] [Indexed: 12/31/2022]
Abstract
The adult central nervous system is commonly known to have a very limited regenerative capacity. The presence of functional stem cells in the brain can therefore be seen as a paradox, since in other organs these are known to counterbalance cell loss derived from pathological conditions. This fact has therefore raised the possibility to stimulate neural stem cell differentiation and proliferation or survival by either stem cell replacement therapy or direct administration of neurotrophic factors or other proneurogenic molecules, which in turn has also originated regenerative medicine for the treatment of otherwise incurable neurodegenerative and neuropsychiatric disorders that take a huge toll on society. This may be facilitated by the fact that many of these disorders converge on similar pathophysiological pathways: excitotoxicity, oxidative stress, neuroinflammation, mitochondrial failure, excessive intracellular calcium and apoptosis. This review will therefore focus on the most promising achievements in promoting neuroprotection and neuroregeneration reported to date.
Collapse
|
35
|
Abstract
Peripheral axonal regeneration requires surface-expanding membrane addition. The continuous incorporation of new membranes into the axolemma allows the pushing force of elongating microtubules to drive axonal growth cones forwards. Hence, a constant supply of membranes and cytoskeletal building blocks is required, often for many weeks. In human peripheral nerves, axonal tips may be more than 1 m away from the neuronal cell body. Therefore, in the initial phase of regeneration, membranes are derived from pre-existing vesicles or synthesised locally. Only later stages of axonal regeneration are supported by membranes and proteins synthesised in neuronal cell bodies, considering that the fastest anterograde transport mechanisms deliver cargo at 20 cm/day. Whereas endocytosis and exocytosis of membrane vesicles are balanced in intact axons, membrane incorporation exceeds membrane retrieval during regeneration to compensate for the loss of membranes distal to the lesion site. Physiological membrane turnover rates will not be established before the completion of target reinnervation. In this review, the current knowledge on membrane traffic in axonal outgrowth is summarised, with a focus on endosomal vesicles as the providers of membranes and carriers of growth factor receptors required for initiating signalling pathways to promote the elongation and branching of regenerating axons in lesioned peripheral nerves.
Collapse
Affiliation(s)
- Barbara Hausott
- Division of Neuroanatomy, Department of Anatomy, Histology and Embryology, Medical University Innsbruck, 6020, Innsbruck, Austria
| | - Lars Klimaschewski
- Division of Neuroanatomy, Department of Anatomy, Histology and Embryology, Medical University Innsbruck, 6020, Innsbruck, Austria
| |
Collapse
|
36
|
Song M, Giza J, Proenca CC, Jing D, Elliott M, Dincheva I, Shmelkov SV, Kim J, Schreiner R, Huang SH, Castrén E, Prekeris R, Hempstead BL, Chao MV, Dictenberg JB, Rafii S, Chen ZY, Rodriguez-Boulan E, Lee FS. Slitrk5 Mediates BDNF-Dependent TrkB Receptor Trafficking and Signaling. Dev Cell 2015; 33:690-702. [PMID: 26004511 DOI: 10.1016/j.devcel.2015.04.009] [Citation(s) in RCA: 68] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2014] [Revised: 02/25/2015] [Accepted: 04/08/2015] [Indexed: 11/25/2022]
Abstract
Recent studies in humans and in genetic mouse models have identified Slit- and NTRK-like family (Slitrks) as candidate genes for neuropsychiatric disorders. All Slitrk isotypes are highly expressed in the CNS, where they mediate neurite outgrowth, synaptogenesis, and neuronal survival. However, the molecular mechanisms underlying these functions are not known. Here, we report that Slitrk5 modulates brain-derived neurotrophic factor (BDNF)-dependent biological responses through direct interaction with TrkB receptors. Under basal conditions, Slitrk5 interacts primarily with a transsynaptic binding partner, protein tyrosine phosphatase δ (PTPδ); however, upon BDNF stimulation, Slitrk5 shifts to cis-interactions with TrkB. In the absence of Slitrk5, TrkB has a reduced rate of ligand-dependent recycling and altered responsiveness to BDNF treatment. Structured illumination microscopy revealed that Slitrk5 mediates optimal targeting of TrkB receptors to Rab11-positive recycling endosomes through recruitment of a Rab11 effector protein, Rab11-FIP3. Thus, Slitrk5 acts as a TrkB co-receptor that mediates its BDNF-dependent trafficking and signaling.
Collapse
Affiliation(s)
- Minseok Song
- Department of Psychiatry, Weill Medical College of Cornell University, 1300 York Avenue, New York, NY 10065, USA
| | - Joanna Giza
- Department of Psychiatry, Weill Medical College of Cornell University, 1300 York Avenue, New York, NY 10065, USA
| | - Catia C Proenca
- Developmental and Molecular Pathways, Novartis Institutes for Biomedical Research, 4002 Basel, Switzerland
| | - Deqiang Jing
- Department of Psychiatry, Weill Medical College of Cornell University, 1300 York Avenue, New York, NY 10065, USA
| | - Mark Elliott
- Department of Psychiatry, University of California at San Francisco, 600 16th Street, San Francisco, CA 94158, USA
| | - Iva Dincheva
- Department of Psychiatry, Weill Medical College of Cornell University, 1300 York Avenue, New York, NY 10065, USA
| | - Sergey V Shmelkov
- Department of Biochemistry and Molecular Pharmacology, Langone Medical Center, Skirball Institute of Biomolecular Medicine, New York University School of Medicine, 540 First Avenue, New York, NY 10016, USA
| | - Jihye Kim
- Department of Psychiatry, Weill Medical College of Cornell University, 1300 York Avenue, New York, NY 10065, USA; Albert Einstein College of Medicine, 1300 Morris Park Avenue, Bronx, NY 10461, USA
| | - Ryan Schreiner
- Dyson Vision Research Institute, Department of Ophthalmology, Weill Medical College of Cornell University, 1300 York Avenue, New York, NY 10065, USA
| | - Shu-Hong Huang
- Department of Neurobiology, Shandong Provincial Key Laboratory of Mental Disorders, School of Medicine and the Collaborative Innovation Center for Brain Science, Shandong University, No. 44 Wenhua Xi Road, Jinan, Shandong 250012, China
| | - Eero Castrén
- Neuroscience Centre, University of Helsinki, 00790 Helsinki, Finland
| | - Rytis Prekeris
- Department of Cell and Developmental Biology, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Barbara L Hempstead
- Division of Hematology/Medical Oncology, Weill Medical College of Cornell University, 1300 York Avenue, New York, NY 10065, USA
| | - Moses V Chao
- Molecular Neurobiology Program, Skirball Institute of Biomolecular Medicine, New York University School of Medicine, 540 First Avenue, New York, NY 10016, USA
| | - Jason B Dictenberg
- AccelBio, DMC Advanced Biotechnology Incubator, Brooklyn, NY 11226, USA; Department of Cell Biology, SUNY Downstate Medical School, 450 Clarkson Avenue, Brooklyn, NY 11203, USA
| | - Shahin Rafii
- Department of Genetic Medicine, Weill Medical College of Cornell University, 1300 York Avenue, New York, NY 10065, USA
| | - Zhe-Yu Chen
- Department of Neurobiology, Shandong Provincial Key Laboratory of Mental Disorders, School of Medicine and the Collaborative Innovation Center for Brain Science, Shandong University, No. 44 Wenhua Xi Road, Jinan, Shandong 250012, China.
| | - Enrique Rodriguez-Boulan
- Dyson Vision Research Institute, Department of Ophthalmology, Weill Medical College of Cornell University, 1300 York Avenue, New York, NY 10065, USA.
| | - Francis S Lee
- Department of Psychiatry, Weill Medical College of Cornell University, 1300 York Avenue, New York, NY 10065, USA; Department of Pharmacology, Weill Medical College of Cornell University, 1300 York Avenue, New York, NY 10065, USA
| |
Collapse
|
37
|
Mead B, Berry M, Logan A, Scott RAH, Leadbeater W, Scheven BA. Stem cell treatment of degenerative eye disease. Stem Cell Res 2015; 14:243-57. [PMID: 25752437 PMCID: PMC4434205 DOI: 10.1016/j.scr.2015.02.003] [Citation(s) in RCA: 147] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/24/2014] [Revised: 02/12/2015] [Accepted: 02/14/2015] [Indexed: 12/16/2022] Open
Abstract
Stem cell therapies are being explored extensively as treatments for degenerative eye disease, either for replacing lost neurons, restoring neural circuits or, based on more recent evidence, as paracrine-mediated therapies in which stem cell-derived trophic factors protect compromised endogenous retinal neurons from death and induce the growth of new connections. Retinal progenitor phenotypes induced from embryonic stem cells/induced pluripotent stem cells (ESCs/iPSCs) and endogenous retinal stem cells may replace lost photoreceptors and retinal pigment epithelial (RPE) cells and restore vision in the diseased eye, whereas treatment of injured retinal ganglion cells (RGCs) has so far been reliant on mesenchymal stem cells (MSC). Here, we review the properties of non-retinal-derived adult stem cells, in particular neural stem cells (NSCs), MSC derived from bone marrow (BMSC), adipose tissues (ADSC) and dental pulp (DPSC), together with ESC/iPSC and discuss and compare their potential advantages as therapies designed to provide trophic support, repair and replacement of retinal neurons, RPE and glia in degenerative retinal diseases. We conclude that ESCs/iPSCs have the potential to replace lost retinal cells, whereas MSC may be a useful source of paracrine factors that protect RGC and stimulate regeneration of their axons in the optic nerve in degenerate eye disease. NSC may have potential as both a source of replacement cells and also as mediators of paracrine treatment.
Collapse
Affiliation(s)
- Ben Mead
- Neurotrauma Research Group, Neurobiology Section, School of Clinical and Experimental Medicine, University of Birmingham, B15 2TT, UK; School of Dentistry, University of Birmingham, B4 6NN, UK.
| | - Martin Berry
- Neurotrauma Research Group, Neurobiology Section, School of Clinical and Experimental Medicine, University of Birmingham, B15 2TT, UK
| | - Ann Logan
- Neurotrauma Research Group, Neurobiology Section, School of Clinical and Experimental Medicine, University of Birmingham, B15 2TT, UK
| | - Robert A H Scott
- Neurotrauma Research Group, Neurobiology Section, School of Clinical and Experimental Medicine, University of Birmingham, B15 2TT, UK
| | - Wendy Leadbeater
- Neurotrauma Research Group, Neurobiology Section, School of Clinical and Experimental Medicine, University of Birmingham, B15 2TT, UK
| | - Ben A Scheven
- School of Dentistry, University of Birmingham, B4 6NN, UK
| |
Collapse
|
38
|
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
|
39
|
Post-injury treatment with 7,8-dihydroxyflavone, a TrkB receptor agonist, protects against experimental traumatic brain injury via PI3K/Akt signaling. PLoS One 2014; 9:e113397. [PMID: 25415296 PMCID: PMC4240709 DOI: 10.1371/journal.pone.0113397] [Citation(s) in RCA: 80] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2014] [Accepted: 10/23/2014] [Indexed: 01/21/2023] Open
Abstract
Tropomyosin-related kinase B (TrkB) signaling is critical for promoting neuronal survival following brain damage. The present study investigated the effects and underlying mechanisms of TrkB activation by the TrkB agonist 7,8-dihydroxyflavone (7,8-DHF) on traumatic brain injury (TBI). Mice subjected to controlled cortical impact received intraperitoneal 7,8-DHF or vehicle injection 10 min post-injury and subsequently daily for 3 days. Behavioral studies, histology analysis and brain water content assessment were performed. Levels of TrkB signaling-related molecules and apoptosis-related proteins were analyzed. The protective effect of 7,8-DHF was also investigated in primary neurons subjected to stretch injury. Treatment with 20 mg/kg 7,8-DHF attenuated functional deficits and brain damage up to post-injury day 28. 7,8-DHF also reduced brain edema, neuronal death, and apoptosis at day 4. These changes were accompanied by a significant decrease in cleaved caspase-3 and increase in Bcl-2/Bax ratio. 7,8-DHF enhanced phosphorylation of TrkB, Akt (Ser473/Thr308), and Bad at day 4, but had no effect on Erk 1/2 phosphorylation. Moreover, 7,8-DHF increased brain-derived neurotrophic factor levels and promoted cAMP response element-binding protein (CREB) activation. This beneficial effect was attenuated by inhibition of TrkB or PI3K/Akt. 7,8-DHF also promoted survival and reduced apoptosis in cortical neurons subjected to stretch injury. Remarkably, delayed administration of 7,8-DHF at 3 h post-injury reduced brain tissue damage. Our study demonstrates that activation of TrkB signaling by 7,8-DHF protects against TBI via the PI3K/Akt but not Erk pathway, and this protective effect may be amplified via the PI3K/Akt-CREB cascades.
Collapse
|
40
|
de Miranda AS, Brant F, Campos AC, Vieira LB, Rocha NP, Cisalpino D, Binda NS, Rodrigues DH, Ransohoff RM, Machado FS, Rachid MA, Teixeira AL. Evidence for the contribution of adult neurogenesis and hippocampal cell death in experimental cerebral malaria cognitive outcome. Neuroscience 2014; 284:920-933. [PMID: 25451296 DOI: 10.1016/j.neuroscience.2014.10.062] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2014] [Revised: 10/09/2014] [Accepted: 10/27/2014] [Indexed: 12/22/2022]
Abstract
Cognitive dysfunction is a major sign of cerebral malaria (CM). However, the underlying mechanisms of CM cognitive outcome remain poorly understood. A body of evidence suggests that adult neurogenesis may play a role in learning and memory processes. It has also been reported that these phenomena can be regulated by the immune system. We hypothesized that memory dysfunction in CM results from hippocampal neurogenesis impairment mediated by the deregulated immune response during the acute phase of CM. C57Bl/6 mice were infected with Plasmodium berghei ANKA (PbA) strain, using a standardized inoculation of 10(6) parasitized erythrocytes. Long-term working memory was evaluated using the novel object recognition test. The mRNA expression of brain-derived neurotrophic factor (BDNF), tropomyosin-receptor-kinase (TRK-B) and nerve growth factor (NGF) in the frontal cortex and hippocampus was estimated by real-time polymerase chain reaction (PCR). The protein levels of cytokine interleukin-2 (IL-2), IL-4, IL-6, IL-10, interferon-γ (IFN-γ), tumor necrosis factor-α (TNF-α), and CCL11 and neurotrophins BDNF and NGF were determined using a cytometric bead array (CBA) kit or enzyme-linked immunosorbent assay. Cell viability in the hippocampus was analyzed by Confocal Microscopy. Neurogenesis in the dentate gyrus was determined through quantification of doublecortin (DCX) positive cells. PbA-infected mice presented working memory impairment on day 5 post-infection. At this same time point, CM mice exhibited a decrease in DCX-positive cells in the dentate gyrus in parallel with increased cell death and elevated inflammatory cytokines (IL-6, TNF-α, IFN-γ and CCL11) in the hippocampus and frontal cortex. A significant reduction of BDNF mRNA expression was also found. IL-6 and TNF-α correlated negatively with BDNF and NGF levels in the hippocampus of CM mice. In summary, we provide further evidence that neuroinflammation following PbA-infection influences neurotrophin expression, impairs adult hippocampal neurogenesis and increases hippocampal cell death in association with memory impairment following CM course. The current study identified potential mediators of memory impairment in CM.
Collapse
Affiliation(s)
- A S de Miranda
- Postgraduate Program in Health Sciences: Infectious Diseases and Tropical Medicine, School of Medicine, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil; Immunopharmacology Group, Department of Biochemistry and Immunology, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil; Neuroscience Branch, Interdisciplinary Laboratory of Medical Investigation, School of Medicine, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil; Neuroinflammation Research Center, Department of Neuroscience, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA.
| | - F Brant
- Postgraduate Program in Health Sciences: Infectious Diseases and Tropical Medicine, School of Medicine, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil; Immunopharmacology Group, Department of Biochemistry and Immunology, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | - A C Campos
- Postgraduate Program in Health Sciences: Infectious Diseases and Tropical Medicine, School of Medicine, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil; Immunopharmacology Group, Department of Biochemistry and Immunology, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | - L B Vieira
- Department of Pharmacology, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | - N P Rocha
- Immunopharmacology Group, Department of Biochemistry and Immunology, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil; Neuroscience Branch, Interdisciplinary Laboratory of Medical Investigation, School of Medicine, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil; Department of Pharmacology, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | - D Cisalpino
- Department of Microbiology, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | - N S Binda
- National Institute of Science and Technology in Molecular Medicine, School of Medicine, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | - D H Rodrigues
- Immunopharmacology Group, Department of Biochemistry and Immunology, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil; Neuroscience Branch, Interdisciplinary Laboratory of Medical Investigation, School of Medicine, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | - R M Ransohoff
- Neuroinflammation Research Center, Department of Neuroscience, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
| | - F S Machado
- Postgraduate Program in Health Sciences: Infectious Diseases and Tropical Medicine, School of Medicine, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil; Immunopharmacology Group, Department of Biochemistry and Immunology, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | - M A Rachid
- Department of Pathology, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | - A L Teixeira
- Postgraduate Program in Health Sciences: Infectious Diseases and Tropical Medicine, School of Medicine, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil; Immunopharmacology Group, Department of Biochemistry and Immunology, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil; Neuroscience Branch, Interdisciplinary Laboratory of Medical Investigation, School of Medicine, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil.
| |
Collapse
|
41
|
Harvey AR, Lovett SJ, Majda BT, Yoon JH, Wheeler LPG, Hodgetts SI. Neurotrophic factors for spinal cord repair: Which, where, how and when to apply, and for what period of time? Brain Res 2014; 1619:36-71. [PMID: 25451132 DOI: 10.1016/j.brainres.2014.10.049] [Citation(s) in RCA: 77] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2014] [Revised: 10/20/2014] [Accepted: 10/23/2014] [Indexed: 12/22/2022]
Abstract
A variety of neurotrophic factors have been used in attempts to improve morphological and behavioural outcomes after experimental spinal cord injury (SCI). Here we review many of these factors, their cellular targets, and their therapeutic impact on spinal cord repair in different, primarily rodent, models of SCI. A majority of studies report favourable outcomes but results are by no means consistent, thus a major aim of this review is to consider how best to apply neurotrophic factors after SCI to optimize their therapeutic potential. In addition to which factors are chosen, many variables need be considered when delivering trophic support, including where and when to apply a given factor or factors, how such factors are administered, at what dose, and for how long. Overall, the majority of studies have applied neurotrophic support in or close to the spinal cord lesion site, in the acute or sub-acute phase (0-14 days post-injury). Far fewer chronic SCI studies have been undertaken. In addition, comparatively fewer studies have administered neurotrophic factors directly to the cell bodies of injured neurons; yet in other instructive rodent models of CNS injury, for example optic nerve crush or transection, therapies are targeted directly at the injured neurons themselves, the retinal ganglion cells. The mode of delivery of neurotrophic factors is also an important variable, whether delivered by acute injection of recombinant proteins, sub-acute or chronic delivery using osmotic minipumps, cell-mediated delivery, delivery using polymer release vehicles or supporting bridges of some sort, or the use of gene therapy to modify neurons, glial cells or precursor/stem cells. Neurotrophic factors are often used in combination with cell or tissue grafts and/or other pharmacotherapeutic agents. Finally, the dose and time-course of delivery of trophic support should ideally be tailored to suit specific biological requirements, whether they relate to neuronal survival, axonal sparing/sprouting, or the long-distance regeneration of axons ending in a different mode of growth associated with terminal arborization and renewed synaptogenesis. This article is part of a Special Issue entitled SI: Spinal cord injury.
Collapse
Affiliation(s)
- Alan R Harvey
- School of Anatomy, Physiology and Human Biology, The University of Western Australia, 35 Stirling Highway, Crawley, WA 6009, Australia.
| | - Sarah J Lovett
- School of Anatomy, Physiology and Human Biology, The University of Western Australia, 35 Stirling Highway, Crawley, WA 6009, Australia
| | - Bernadette T Majda
- School of Anatomy, Physiology and Human Biology, The University of Western Australia, 35 Stirling Highway, Crawley, WA 6009, Australia
| | - Jun H Yoon
- School of Anatomy, Physiology and Human Biology, The University of Western Australia, 35 Stirling Highway, Crawley, WA 6009, Australia
| | - Lachlan P G Wheeler
- School of Anatomy, Physiology and Human Biology, The University of Western Australia, 35 Stirling Highway, Crawley, WA 6009, Australia
| | - Stuart I Hodgetts
- School of Anatomy, Physiology and Human Biology, The University of Western Australia, 35 Stirling Highway, Crawley, WA 6009, Australia
| |
Collapse
|
42
|
Shen J, Yu Q. Gambogic amide selectively upregulates TrkA expression and triggers its activation. Pharmacol Rep 2014; 67:217-23. [PMID: 25712642 DOI: 10.1016/j.pharep.2014.09.002] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2014] [Revised: 08/30/2014] [Accepted: 09/08/2014] [Indexed: 01/01/2023]
Abstract
BACKGROUND Gambogic amide is the first identified small molecular agonist for TrkA receptor. It mimics NGF functions by selectively activating TrkA receptor and preventing neuron death. However, its function different from that of NGF remains unknown. METHODS In the current study, we detect the effect of gambogic amide on TrkA expression using TrkA-expressing cell lines in vitro and hippocampi from mice treated with gambogic amide. RESULTS We have confirmed that gambogic amide displays robust neurotrophic activities in provoking neurite outgrowth in vitro. However, gambiogic amide displays a different kinetics from NGF in activating TrkA signals. NGF swiftly provokes TrkA activation and quickly induces TrkA degradation, while gambogic amid selectively upregulates TrkA protein and mRNA levels in a time-dependent manner. Administration of this compound in mice also activates TrkA receptor in hippocampus and promotes TrkA transcription and expression. CONCLUSION This study provides a novel mechanism of how gambogic amide regulates TrkA receptor, other than mimicking NGF in triggering TrkA activation.
Collapse
Affiliation(s)
- Jianying Shen
- Artemisinin Research Center, Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, China
| | - Qingsheng Yu
- Center for Osteonecrosis and Joint Preserving and Reconstruction, Department of Orthopedics, China-Japan Friendship Hospital, Beijing, China.
| |
Collapse
|
43
|
Mead B, Logan A, Berry M, Leadbeater W, Scheven BA. Dental pulp stem cells, a paracrine-mediated therapy for the retina. Neural Regen Res 2014; 9:577-8. [PMID: 25206857 PMCID: PMC4146241 DOI: 10.4103/1673-5374.130089] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/26/2014] [Indexed: 12/12/2022] Open
Affiliation(s)
- Ben Mead
- Neurotrauma and Neurodegeneration Section, School of Clinical and Experimental Medicine, University of Birmingham, Birmingham B15 2TT, United Kingdom ; School of Dentistry, University of Birmingham, Birmingham B4 6NN, United Kingdom
| | - Ann Logan
- Neurotrauma and Neurodegeneration Section, School of Clinical and Experimental Medicine, University of Birmingham, Birmingham B15 2TT, United Kingdom
| | - Martin Berry
- Neurotrauma and Neurodegeneration Section, School of Clinical and Experimental Medicine, University of Birmingham, Birmingham B15 2TT, United Kingdom
| | - Wendy Leadbeater
- Neurotrauma and Neurodegeneration Section, School of Clinical and Experimental Medicine, University of Birmingham, Birmingham B15 2TT, United Kingdom
| | - Ben A Scheven
- School of Dentistry, University of Birmingham, Birmingham B4 6NN, United Kingdom
| |
Collapse
|
44
|
Jerónimo-Santos A, Batalha VL, Müller CE, Baqi Y, Sebastião AM, Lopes LV, Diógenes MJ. Impact of in vivo chronic blockade of adenosine A2A receptors on the BDNF-mediated facilitation of LTP. Neuropharmacology 2014; 83:99-106. [DOI: 10.1016/j.neuropharm.2014.04.006] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2013] [Revised: 03/20/2014] [Accepted: 04/08/2014] [Indexed: 11/29/2022]
|
45
|
Terenzio M, Golding M, Russell MRG, Wicher KB, Rosewell I, Spencer-Dene B, Ish-Horowicz D, Schiavo G. Bicaudal-D1 regulates the intracellular sorting and signalling of neurotrophin receptors. EMBO J 2014; 33:1582-98. [PMID: 24920579 PMCID: PMC4198053 DOI: 10.15252/embj.201387579] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2013] [Revised: 03/14/2014] [Accepted: 04/23/2014] [Indexed: 12/31/2022] Open
Abstract
We have identified a new function for the dynein adaptor Bicaudal D homolog 1 (BICD1) by screening a siRNA library for genes affecting the dynamics of neurotrophin receptor-containing endosomes in motor neurons (MNs). Depleting BICD1 increased the intracellular accumulation of brain-derived neurotrophic factor (BDNF)-activated TrkB and p75 neurotrophin receptor (p75(NTR)) by disrupting the endosomal sorting, reducing lysosomal degradation and increasing the co-localisation of these neurotrophin receptors with retromer-associated sorting nexin 1. The resulting re-routing of active receptors increased their recycling to the plasma membrane and altered the repertoire of signalling-competent TrkB isoforms and p75(NTR) available for ligand binding on the neuronal surface. This resulted in attenuated, but more sustained, AKT activation in response to BDNF stimulation. These data, together with our observation that Bicd1 expression is restricted to the developing nervous system when neurotrophin receptor expression peaks, indicate that BICD1 regulates neurotrophin signalling by modulating the endosomal sorting of internalised ligand-activated receptors.
Collapse
Affiliation(s)
- Marco Terenzio
- Molecular NeuroPathobiology Laboratory, Cancer Research UK London Research Institute, London, UK
| | - Matthew Golding
- Molecular NeuroPathobiology Laboratory, Cancer Research UK London Research Institute, London, UK
| | - Matthew R G Russell
- Electron Microscopy Laboratory, Cancer Research UK London Research Institute, London, UK
| | - Krzysztof B Wicher
- Developmental Genetics Laboratory, Cancer Research UK London Research Institute, London, UK
| | - Ian Rosewell
- Transgenic Services laboratory, Cancer Research UK London Research Institute, London, UK
| | - Bradley Spencer-Dene
- Experimental Histopathology Laboratory, Cancer Research UK London Research Institute, London, UK
| | - David Ish-Horowicz
- Developmental Genetics Laboratory, Cancer Research UK London Research Institute, London, UK
| | - Giampietro Schiavo
- Molecular NeuroPathobiology Laboratory, Cancer Research UK London Research Institute, London, UK Sobell Department of Motor Neuroscience & Movement Disorders, UCL-Institute of Neurology, University College London, London, UK
| |
Collapse
|
46
|
Guo W, Ji Y, Wang S, Sun Y, Lu B. Neuronal activity alters BDNF-TrkB signaling kinetics and downstream functions. J Cell Sci 2014; 127:2249-60. [PMID: 24634513 DOI: 10.1242/jcs.139964] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Differential kinetics of the same signaling pathway might elicit different cellular outcomes. Here, we show that high-frequency neuronal activity converts BDNF-induced TrkB (also known as NTRK2) signaling from a transient to a sustained mode. A prior depolarization (15 mM KCl, 1 hour) resulted in a long-lasting (>24 hours) activation of the TrkB receptor and its downstream signaling, which otherwise lasts less than an hour. The long-term potentiation (LTP)-inducing theta-burst stimulation but not the long-term depression (LTD)-inducing low-frequency stimulation also induced sustained activation of TrkB. This sustained signaling facilitated dendritic branching and rescued neuronal apoptosis induced by glutamate. The change in TrkB signaling kinetics is mediated by Ca(2+) elevation and CaMKII activation, leading to an increase in TrkB expression on the neuronal surface. Physical exercise also alters the kinetics of TrkB phosphorylation induced by exogenous BDNF. Sustained TrkB signaling might serve as a key mechanism underlying the synergistic effects of neuronal activity and BDNF.
Collapse
Affiliation(s)
- Wei Guo
- Tsinghua-Peking Center for Life Sciences, Beijing, China School of Medicine, Tsinghua University, 1 Qinghuayuan Road, Beijing, 100084, China School of Life Sciences, Tsinghua University, 1 Qinghuayuan Road, Beijing, 100084, China
| | - Yuanyuan Ji
- GlaxoSmithKline, R&D China, Building 3, 898 Halei Road, Zhangjiang Hi-tech Park, Pudong, Shanghai, 201203, China
| | - Shudan Wang
- Tsinghua-Peking Center for Life Sciences, Beijing, China School of Life Sciences, Tsinghua University, 1 Qinghuayuan Road, Beijing, 100084, China
| | - Yun Sun
- Tsinghua-Peking Center for Life Sciences, Beijing, China National Institute of Biological Sciences, Beijing, 102206, China School of Life Sciences, Peking University, Beijing, 100871, China
| | - Bai Lu
- Tsinghua-Peking Center for Life Sciences, Beijing, China School of Medicine, Tsinghua University, 1 Qinghuayuan Road, Beijing, 100084, China
| |
Collapse
|
47
|
Modulation of c-Fos and BDNF protein expression in pentylenetetrazole-kindled mice following the treatment with novel antiepileptic compound HHL-6. BIOMED RESEARCH INTERNATIONAL 2014; 2014:876712. [PMID: 24605339 PMCID: PMC3925558 DOI: 10.1155/2014/876712] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/15/2013] [Revised: 10/22/2013] [Accepted: 10/30/2013] [Indexed: 01/10/2023]
Abstract
Brain-derived neurotrophic factor (BDNF) and c-Fos are shown to promote epileptogenesis and are taken as a marker of neuronal activity. The present study investigated the expression of BDNF and c-Fos in mice brain with pentylenetetrazol- (PTZ-) induced generalized seizure and evaluated the effect of novel tryptamine derivative HHL-6 on the expression of these two markers. The subconvulsive dose of PTZ (50 mg/kg) was administered on alternate days in the experimental groups until the seizure scores 4-5 developed in the PTZ-control group. At the end of each experiment, animals were sacrificed, brain samples were collected and cryosectioned, and immunohistochemical analysis of BDNF and c-Fos protein was performed. Data obtained from two sections per mouse (n = 12 animals/group) is presented as means ± S.E.M. The test compound HHL-6 demonstrated a potent anticonvulsant activity in the PTZ-induced seizure in mice. Significant reduction in the BDNF (P < 0.003) and c-Fos (P < 0.01) protein expression was observed in the HHL-6 treated group. Based on these results we suggest that one of the possible mechanisms of HHL-6 to inhibit epileptogenesis might be due to its controlling effect on the cellular and molecular expression of the factors that contribute to the development of epileptogenic plasticity in the CNS.
Collapse
|
48
|
Abstract
The tropomyosin-related tyrosine kinase (Trk) receptors were initially described as a family of growth factor receptors required for neuronal survival. They have since been shown to influence many aspects of neuronal development and function, including differentiation, outgrowth, and synaptic plasticity. This chapter will give an overview on the biology of Trk receptors within the nervous system. The structure and downstream signaling pathways of the full-length receptors will be described, as well as the biological functions of their truncated isoforms. Finally, the role of Trk receptors in the nervous system in health and disease will be discussed.
Collapse
|
49
|
Lu B, Nagappan G, Lu Y. BDNF and synaptic plasticity, cognitive function, and dysfunction. Handb Exp Pharmacol 2014; 220:223-50. [PMID: 24668475 DOI: 10.1007/978-3-642-45106-5_9] [Citation(s) in RCA: 671] [Impact Index Per Article: 61.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Among all neurotrophins, brain-derived neurotrophic factor (BDNF) stands out for its high level of expression in the brain and its potent effects on synapses. It is now widely accepted that the main function of BDNF in the adult brain is to regulate synapses, with structural and functional effects ranging from short-term to long-lasting, on excitatory or inhibitory synapses, in many brain regions. The diverse effects of BDNF on brain synapses stem from its complex downstream signaling cascades, as well as the diametrically opposing effects of the pro- and mature form through distinct receptors, TrkB and p75(NTR). Many aspects of BDNF cell biology are regulated by neuronal activity. The synergistic interactions between neuronal activity and synaptic plasticity by BDNF make it an ideal and essential regulator of cellular processes that underlie cognition and other complex behaviors. Indeed, numerous studies firmly established that BDNF plays a critical role in hippocampal long-term potentiation (LTP), a long-term enhancement of synaptic efficacy thought to underlie learning and memory. Converging evidence now strongly suggest that deficits in BDNF signaling contribute to the pathogenesis of several major diseases and disorders such as Huntington's disease, Alzheimer's disease, and depression. Thus, manipulating BDNF pathways represents a viable treatment approach to a variety of neurological and psychiatric disorders.
Collapse
Affiliation(s)
- B Lu
- GlaxoSmithKline, R&D China, Building 3, 898 Halei Road, Zhangjiang Hi-tech Park, Pudong, Shanghai, 201203, China,
| | | | | |
Collapse
|
50
|
D'Amore DE, Tracy BA, Parikh V. Exogenous BDNF facilitates strategy set-shifting by modulating glutamate dynamics in the dorsal striatum. Neuropharmacology 2013; 75:312-23. [DOI: 10.1016/j.neuropharm.2013.07.033] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2013] [Revised: 07/29/2013] [Accepted: 07/30/2013] [Indexed: 01/19/2023]
|