1
|
Turner-Ivey B, Jenkins DP, Carroll SL. Multiple Roles for Neuregulins and Their ERBB Receptors in Neurodegenerative Disease Pathogenesis and Therapy. THE AMERICAN JOURNAL OF PATHOLOGY 2025:S0002-9440(25)00119-1. [PMID: 40254133 DOI: 10.1016/j.ajpath.2025.03.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/07/2025] [Revised: 03/06/2025] [Accepted: 03/17/2025] [Indexed: 04/22/2025]
Abstract
The role that neurotrophins, such as nerve growth factor, play in the pathogenesis of neurodegenerative diseases has long been appreciated. However, the neuregulin (NRG) family of growth factors and/or their v-erb-B2 avian erythroblastic leukemia viral oncogene homolog (ERBB) receptors have also been implicated in the pathogenesis of conditions, such as Alzheimer disease (AD), frontotemporal lobar degeneration (FTLD), and amyotrophic lateral sclerosis (ALS). In this review, we consider i) the structural variability of NRG isoforms generated by alternative RNA splicing, the use of multiple promoters and proteolysis, and the impact that this structural variability has on neuronal and glial physiology during development and adulthood. We discuss ii) the NRG receptors ERBB2, ERBB3, and ERBB4, how activation of each of these receptors further diversifies NRG actions in the central nervous system, and how dementia-related proteins, such as γ-secretase modulate the action of NRGs and their ERBB receptors. We then iii) turn to the abnormalities in NRG and ERBB expression and function evident in human AD and mouse AD models, how these abnormalities affect brain function, and attempts to use NRGs to treat AD. Finally, iv) we discuss NRG effects on the survival and function of neurons relevant to FTLD and ALS, alterations in NRG/ERBB signaling identified in these conditions, and the recent discovery of multiple human pedigrees in which autosomal dominant FTLD/ALS potentially results from point mutations in ERBB4.
Collapse
Affiliation(s)
- Brittany Turner-Ivey
- Department of Pathology and Laboratory Medicine, Medical University of South Carolina, Charleston, South Carolina
| | - Dorea P Jenkins
- Department of Pathology and Laboratory Medicine, Medical University of South Carolina, Charleston, South Carolina
| | - Steven L Carroll
- Department of Pathology and Laboratory Medicine, Medical University of South Carolina, Charleston, South Carolina.
| |
Collapse
|
2
|
Goffin L, Lemoine D, Clotman F. Potential contribution of spinal interneurons to the etiopathogenesis of amyotrophic lateral sclerosis. Front Neurosci 2024; 18:1434404. [PMID: 39091344 PMCID: PMC11293063 DOI: 10.3389/fnins.2024.1434404] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2024] [Accepted: 06/21/2024] [Indexed: 08/04/2024] Open
Abstract
Amyotrophic lateral sclerosis (ALS) consists of a group of adult-onset fatal and incurable neurodegenerative disorders characterized by the progressive death of motor neurons (MNs) throughout the central nervous system (CNS). At first, ALS was considered to be an MN disease, caused by cell-autonomous mechanisms acting specifically in MNs. Accordingly, data from ALS patients and ALS animal models revealed alterations in excitability in multiple neuronal populations, including MNs, which were associated with a variety of cellular perturbations such as protein aggregation, ribonucleic acid (RNA) metabolism defects, calcium dyshomeostasis, modified electrophysiological properties, and autophagy malfunctions. However, experimental evidence rapidly demonstrated the involvement of other types of cells, including glial cells, in the etiopathogenesis of ALS through non-cell autonomous mechanisms. Surprisingly, the contribution of pre-motor interneurons (INs), which regulate MN activity and could therefore critically modulate their excitability at the onset or during the progression of the disease, has to date been severely underestimated. In this article, we review in detail how spinal pre-motor INs are affected in ALS and their possible involvement in the etiopathogenesis of the disease.
Collapse
Affiliation(s)
| | | | - Frédéric Clotman
- Université catholique de Louvain, Louvain Institute of Biomolecular Science and Technology, Animal Molecular and Cellular Biology, Louvain-la-Neuve, Belgium
| |
Collapse
|
3
|
Odierna GL, Vucic S, Dyer M, Dickson T, Woodhouse A, Blizzard C. How do we get from hyperexcitability to excitotoxicity in amyotrophic lateral sclerosis? Brain 2024; 147:1610-1621. [PMID: 38408864 PMCID: PMC11068114 DOI: 10.1093/brain/awae039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Revised: 11/15/2023] [Accepted: 12/10/2023] [Indexed: 02/28/2024] Open
Abstract
Amyotrophic lateral sclerosis is a devastating neurodegenerative disease that, at present, has no effective cure. Evidence of increased circulating glutamate and hyperexcitability of the motor cortex in patients with amyotrophic lateral sclerosis have provided an empirical support base for the 'dying forward' excitotoxicity hypothesis. The hypothesis postulates that increased activation of upper motor neurons spreads pathology to lower motor neurons in the spinal cord in the form of excessive glutamate release, which triggers excitotoxic processes. Many clinical trials have focused on therapies that target excitotoxicity via dampening neuronal activation, but not all are effective. As such, there is a growing tension between the rising tide of evidence for the 'dying forward' excitotoxicity hypothesis and the failure of therapies that target neuronal activation. One possible solution to these contradictory outcomes is that our interpretation of the current evidence requires revision in the context of appreciating the complexity of the nervous system and the limitations of the neurobiological assays we use to study it. In this review we provide an evaluation of evidence relevant to the 'dying forward' excitotoxicity hypothesis and by doing so, identify key gaps in our knowledge that need to be addressed. We hope to provide a road map from hyperexcitability to excitotoxicity so that we can better develop therapies for patients suffering from amyotrophic lateral sclerosis. We conclude that studies of upper motor neuron activity and their synaptic output will play a decisive role in the future of amyotrophic lateral sclerosis therapy.
Collapse
Affiliation(s)
- G Lorenzo Odierna
- Tasmanian School of Medicine, University of Tasmania, Hobart, TAS 7000, Australia
| | - Steve Vucic
- Brain and Nerve Research Center, The University of Sydney, Sydney 2050, Australia
| | - Marcus Dyer
- Menzies Institute for Medical Research, University of Tasmania, Hobart, TAS 7000, Australia
- Department of Pharmaceutical and Pharmacological Sciences, Center for Neurosciences, Vrije Universiteit Brussel (VUB), 1090 Brussels, Belgium
| | - Tracey Dickson
- Menzies Institute for Medical Research, University of Tasmania, Hobart, TAS 7000, Australia
| | - Adele Woodhouse
- The Wicking Dementia Centre, University of Tasmania, Hobart, TAS 7000, Australia
| | - Catherine Blizzard
- Tasmanian School of Medicine, University of Tasmania, Hobart, TAS 7000, Australia
- Menzies Institute for Medical Research, University of Tasmania, Hobart, TAS 7000, Australia
| |
Collapse
|
4
|
Kang Q, Jiang S, Min J, Hu F, Xu R. Parvalbumin interneurons dysfunction is potentially associated with FαMNs decrease and NRG1-ErbB4 signaling inhibition in spinal cord in amyotrophic lateral sclerosis. Aging (Albany NY) 2023; 15:15324-15339. [PMID: 38157256 PMCID: PMC10781496 DOI: 10.18632/aging.205351] [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: 05/11/2023] [Accepted: 11/06/2023] [Indexed: 01/03/2024]
Abstract
OBJECTIVE To investigate the alteration of PV interneurons in ALS mainly focusing its dynamic changes and its relationship with motor neurons and ErbB4 signaling. METHODS SOD1G93A mice were used as ALS model. ALS animals were divided into different groups according to birth age: symptomatic prophase (50~60 days), symptomatic phase (90~100 days), and symptomatic progression (130~140 days). Immunofluorescence was performed for measurement of PV-positive interneurons, MMP-9, ChAT, NeuN and ErbB4. RT-qPCR and western blot were used to determine the expression of PV and MMP-9. RESULTS PV expression was remarkably higher in the anterior horn of gray matter compared with posterior horn and area in the middle of gray matter in control mice. In ALS mice, PV, MMP-9 and ErbB4 levels were gradually decreased along with onset. PV, MMP-9 and ErbB4 levels in ALS mice were significantly down-regulated than control mice after onset, indicating the alteration of PV interneurons, FαMNs and ErbB4. SαMNs levels only decreased remarkably at symptomatic progression in ALS mice compared with control mice, while γMNs levels showed no significant change during whole period in all mice. MMP-9 and ErbB4 were positively correlated with PV. NRG1 treatment significantly enhanced the expression of ErBb4, PV and MMP-9 in ALS mice. CONCLUSION PV interneurons decrease is along with FαMNs and ErbB4 decrease in ALS mice.
Collapse
Affiliation(s)
- Qin Kang
- Department of Neurology, Medical College of Nanchang University, Nanchang 330006, Jiangxi, P.R. China
- Department of Neurology, Jiangxi Provincial People’s Hospital, First Affiliated Hospital of Nanchang Medical College, Clinical College of Nanchang Medical College, Nanchang 330006, Jiangxi, P.R. China
| | - Shishi Jiang
- Department of Neurology, Medical College of Nanchang University, Nanchang 330006, Jiangxi, P.R. China
| | - Jun Min
- Department of Neurology, The Second Affiliated Hospital of Nanchang University, Nanchang 330006, Jiangxi, P.R. China
| | - Fan Hu
- Department of Neurology, Jiangxi Provincial People’s Hospital, First Affiliated Hospital of Nanchang Medical College, Clinical College of Nanchang Medical College, Nanchang 330006, Jiangxi, P.R. China
| | - Renshi Xu
- Department of Neurology, Medical College of Nanchang University, Nanchang 330006, Jiangxi, P.R. China
- Department of Neurology, Jiangxi Provincial People’s Hospital, First Affiliated Hospital of Nanchang Medical College, Clinical College of Nanchang Medical College, Nanchang 330006, Jiangxi, P.R. China
| |
Collapse
|
5
|
Hernández S, Salvany S, Casanovas A, Piedrafita L, Soto-Bernardini MC, Tarabal O, Blasco A, Gras S, Gatius A, Schwab MH, Calderó J, Esquerda JE. Persistent NRG1 Type III Overexpression in Spinal Motor Neurons Has No Therapeutic Effect on ALS-Related Pathology in SOD1 G93A Mice. Neurotherapeutics 2023; 20:1820-1834. [PMID: 37733208 PMCID: PMC10684470 DOI: 10.1007/s13311-023-01424-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/31/2023] [Indexed: 09/22/2023] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a devastating neurodegenerative disease affecting upper and lower motor neurons (MNs). Neuregulin-1 (NRG1) is a pleiotropic growth factor that has been shown to be potentially valuable for ALS when supplemented by means of viral-mediated gene therapy. However, these results are inconsistent with other reports. An alternative approach for investigating the therapeutic impact of NRG1 on ALS is the use of transgenic mouse lines with genetically defined NRG1 overexpression. Here, we took advantage of a mouse line with NRG1 type III overexpression in spinal cord α motor neurons (MN) to determine the impact of steadily enhanced NRG1 signalling on mutant superoxide dismutase 1 (SOD1)-induced disease. The phenotype of SOD1G93A-NRG1 double transgenic mice was analysed in detail, including neuropathology and extensive behavioural testing. At least 3 animals per condition and sex were histopathologically assessed, and a minimum of 10 mice per condition and sex were clinically evaluated. The accumulation of misfolded SOD1 (mfSOD1), MN degeneration, and a glia-mediated neuroinflammatory response are pathological hallmarks of ALS progression in SOD1G93A mice. None of these aspects was significantly improved when examined in double transgenic NRG1-SOD1G93A mice. In addition, behavioural testing revealed that NRG1 type III overexpression did not affect the survival of SOD1G93A mice but accelerated disease onset and worsened the motor phenotype.
Collapse
Affiliation(s)
- Sara Hernández
- Unitat de Neurobiologia Cel·lular, Departament de Medicina Experimental, Facultat de Medicina, Universitat de Lleida and Institut de Recerca Biomèdica de Lleida (IRBLleida), Lleida, Catalonia, Spain
| | - Sara Salvany
- Unitat de Neurobiologia Cel·lular, Departament de Medicina Experimental, Facultat de Medicina, Universitat de Lleida and Institut de Recerca Biomèdica de Lleida (IRBLleida), Lleida, Catalonia, Spain
| | - Anna Casanovas
- Unitat de Neurobiologia Cel·lular, Departament de Medicina Experimental, Facultat de Medicina, Universitat de Lleida and Institut de Recerca Biomèdica de Lleida (IRBLleida), Lleida, Catalonia, Spain
| | - Lídia Piedrafita
- Unitat de Neurobiologia Cel·lular, Departament de Medicina Experimental, Facultat de Medicina, Universitat de Lleida and Institut de Recerca Biomèdica de Lleida (IRBLleida), Lleida, Catalonia, Spain
| | - M Clara Soto-Bernardini
- Department of Neurology, University Hospital Leipzig, Leipzig, Germany
- Center for Research in Biotechnology (CIB), Costa Rica Institute of Technology (TEC), Cartago, Costa Rica
| | - Olga Tarabal
- Unitat de Neurobiologia Cel·lular, Departament de Medicina Experimental, Facultat de Medicina, Universitat de Lleida and Institut de Recerca Biomèdica de Lleida (IRBLleida), Lleida, Catalonia, Spain
| | - Alba Blasco
- Unitat de Neurobiologia Cel·lular, Departament de Medicina Experimental, Facultat de Medicina, Universitat de Lleida and Institut de Recerca Biomèdica de Lleida (IRBLleida), Lleida, Catalonia, Spain
| | - Sílvia Gras
- Unitat de Neurobiologia Cel·lular, Departament de Medicina Experimental, Facultat de Medicina, Universitat de Lleida and Institut de Recerca Biomèdica de Lleida (IRBLleida), Lleida, Catalonia, Spain
| | - Alaó Gatius
- Unitat de Neurobiologia Cel·lular, Departament de Medicina Experimental, Facultat de Medicina, Universitat de Lleida and Institut de Recerca Biomèdica de Lleida (IRBLleida), Lleida, Catalonia, Spain
| | - Markus H Schwab
- Paul Flechsig Institute of Neuropathology, University Hospital Leipzig, Leipzig, Germany
| | - Jordi Calderó
- Unitat de Neurobiologia Cel·lular, Departament de Medicina Experimental, Facultat de Medicina, Universitat de Lleida and Institut de Recerca Biomèdica de Lleida (IRBLleida), Lleida, Catalonia, Spain
| | - Josep E Esquerda
- Unitat de Neurobiologia Cel·lular, Departament de Medicina Experimental, Facultat de Medicina, Universitat de Lleida and Institut de Recerca Biomèdica de Lleida (IRBLleida), Lleida, Catalonia, Spain.
| |
Collapse
|
6
|
Eleftheriadis PE, Pothakos K, Sharples SA, Apostolou PE, Mina M, Tetringa E, Tsape E, Miles GB, Zagoraiou L. Peptidergic modulation of motor neuron output via CART signaling at C bouton synapses. Proc Natl Acad Sci U S A 2023; 120:e2300348120. [PMID: 37733738 PMCID: PMC10523464 DOI: 10.1073/pnas.2300348120] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2023] [Accepted: 07/17/2023] [Indexed: 09/23/2023] Open
Abstract
The intensity of muscle contraction, and therefore movement vigor, needs to be adaptable to enable complex motor behaviors. This can be achieved by adjusting the properties of motor neurons, which form the final common pathway for all motor output from the central nervous system. Here, we identify roles for a neuropeptide, cocaine- and amphetamine-regulated transcript (CART), in the control of movement vigor. We reveal distinct but parallel mechanisms by which CART and acetylcholine, both released at C bouton synapses on motor neurons, selectively amplify the output of subtypes of motor neurons that are recruited during intense movement. We find that mice with broad genetic deletion of CART or selective elimination of acetylcholine from C boutons exhibit deficits in behavioral tasks that require higher levels of motor output. Overall, these data uncover spinal modulatory mechanisms that control movement vigor to support movements that require a high degree of muscle force.
Collapse
Affiliation(s)
| | - Konstantinos Pothakos
- Center of Basic Research, Biomedical Research Foundation Academy of Athens, Athens11527, Greece
| | - Simon A. Sharples
- School of Psychology and Neuroscience, University of St. Andrews, St. AndrewsKY16 9JP, United Kingdom
| | - Panagiota E. Apostolou
- Center of Basic Research, Biomedical Research Foundation Academy of Athens, Athens11527, Greece
| | - Maria Mina
- Center of Basic Research, Biomedical Research Foundation Academy of Athens, Athens11527, Greece
| | - Efstathia Tetringa
- Center of Basic Research, Biomedical Research Foundation Academy of Athens, Athens11527, Greece
| | - Eirini Tsape
- Center of Basic Research, Biomedical Research Foundation Academy of Athens, Athens11527, Greece
| | - Gareth B. Miles
- School of Psychology and Neuroscience, University of St. Andrews, St. AndrewsKY16 9JP, United Kingdom
| | - Laskaro Zagoraiou
- Center of Basic Research, Biomedical Research Foundation Academy of Athens, Athens11527, Greece
| |
Collapse
|
7
|
Kong L, Hassinan CW, Gerstner F, Buettner JM, Petigrow JB, Valdivia DO, Chan-Cortés MH, Mistri A, Cao A, McGaugh SA, Denton M, Brown S, Ross J, Schwab MH, Simon CM, Sumner CJ. Boosting neuregulin 1 type-III expression hastens SMA motor axon maturation. Acta Neuropathol Commun 2023; 11:53. [PMID: 36997967 PMCID: PMC10061791 DOI: 10.1186/s40478-023-01551-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2023] [Accepted: 03/12/2023] [Indexed: 04/01/2023] Open
Abstract
Intercellular communication between axons and Schwann cells is critical for attaining the complex morphological steps necessary for axon maturation. In the early onset motor neuron disease spinal muscular atrophy (SMA), many motor axons are not ensheathed by Schwann cells nor grow sufficiently in radial diameter to become myelinated. These developmentally arrested motor axons are dysfunctional and vulnerable to rapid degeneration, limiting efficacy of current SMA therapeutics. We hypothesized that accelerating SMA motor axon maturation would improve their function and reduce disease features. A principle regulator of peripheral axon development is neuregulin 1 type III (NRG1-III). Expressed on axon surfaces, it interacts with Schwann cell receptors to mediate axon ensheathment and myelination. We examined NRG1 mRNA and protein expression levels in human and mouse SMA tissues and observed reduced expression in SMA spinal cord and in ventral, but not dorsal root axons. To determine the impact of neuronal NRG1-III overexpression on SMA motor axon development, we bred NRG1-III overexpressing mice to SMA∆7 mice. Neonatally, elevated NRG1-III expression increased SMA ventral root size as well as axon segregation, diameter, and myelination resulting in improved motor axon conduction velocities. NRG1-III was not able to prevent distal axonal degeneration nor improve axon electrophysiology, motor behavior, or survival of older mice. Together these findings demonstrate that early SMA motor axon developmental impairments can be ameliorated by a molecular strategy independent of SMN replacement providing hope for future SMA combinatorial therapeutic approaches.
Collapse
Affiliation(s)
- Lingling Kong
- Departments of Neurology, Johns Hopkins University School of Medicine, 855 North Wolfe Street, Rangos Building Room 234, Baltimore, MD, 21205, USA
| | - Cera W Hassinan
- Departments of Neurology, Johns Hopkins University School of Medicine, 855 North Wolfe Street, Rangos Building Room 234, Baltimore, MD, 21205, USA
| | - Florian Gerstner
- Carl-Ludwig-Institute for Physiology, Leipzig University, Leipzig, Germany
| | - Jannik M Buettner
- Carl-Ludwig-Institute for Physiology, Leipzig University, Leipzig, Germany
| | - Jeffrey B Petigrow
- Departments of Neurology, Johns Hopkins University School of Medicine, 855 North Wolfe Street, Rangos Building Room 234, Baltimore, MD, 21205, USA
| | - David O Valdivia
- Departments of Neurology, Johns Hopkins University School of Medicine, 855 North Wolfe Street, Rangos Building Room 234, Baltimore, MD, 21205, USA
| | - Michelle H Chan-Cortés
- Departments of Neurology, Johns Hopkins University School of Medicine, 855 North Wolfe Street, Rangos Building Room 234, Baltimore, MD, 21205, USA
| | - Amy Mistri
- Departments of Neurology, Johns Hopkins University School of Medicine, 855 North Wolfe Street, Rangos Building Room 234, Baltimore, MD, 21205, USA
| | - Annie Cao
- Departments of Neurology, Johns Hopkins University School of Medicine, 855 North Wolfe Street, Rangos Building Room 234, Baltimore, MD, 21205, USA
| | - Scott Alan McGaugh
- Departments of Neurology, Johns Hopkins University School of Medicine, 855 North Wolfe Street, Rangos Building Room 234, Baltimore, MD, 21205, USA
| | - Madeline Denton
- Departments of Neurology, Johns Hopkins University School of Medicine, 855 North Wolfe Street, Rangos Building Room 234, Baltimore, MD, 21205, USA
| | - Stephen Brown
- Departments of Neurology, Johns Hopkins University School of Medicine, 855 North Wolfe Street, Rangos Building Room 234, Baltimore, MD, 21205, USA
| | - Joshua Ross
- Departments of Neurology, Johns Hopkins University School of Medicine, 855 North Wolfe Street, Rangos Building Room 234, Baltimore, MD, 21205, USA
| | - Markus H Schwab
- Department of Neuropathology, University Hospital Leipzig, Leipzig, Germany
| | - Christian M Simon
- Carl-Ludwig-Institute for Physiology, Leipzig University, Leipzig, Germany
| | - Charlotte J Sumner
- Departments of Neurology, Johns Hopkins University School of Medicine, 855 North Wolfe Street, Rangos Building Room 234, Baltimore, MD, 21205, USA.
- Departments of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA.
| |
Collapse
|
8
|
Bak AN, Djukic S, Kadlecova M, Braunstein TH, Jensen DB, Meehan CF. Cytoplasmic TDP-43 accumulation drives changes in C-bouton number and size in a mouse model of sporadic Amyotrophic Lateral Sclerosis. Mol Cell Neurosci 2023; 125:103840. [PMID: 36921783 DOI: 10.1016/j.mcn.2023.103840] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2022] [Revised: 02/11/2023] [Accepted: 03/04/2023] [Indexed: 03/14/2023] Open
Abstract
An altered neuronal excitability of spinal motoneurones has consistently been implicated in Amyotrophic Lateral Sclerosis (ALS) leading to several investigations of synaptic input to these motoneurones. One such input that has repeatedly been shown to be affected is a population of large cholinergic synapses terminating mainly on the soma of the motoneurones referred to as C-boutons. Most research on these synapses during disease progression has used transgenic Superoxide Dismutase 1 (SOD1) mouse models of the disease which have not only produced conflicting findings, but also fail to recapitulate the key pathological feature seen in ALS; cytoplasmic accumulations of TAR DNA-binding protein 43 (TDP-43). Additionally, they fail to distinguish between slow and fast motoneurones, the latter of which have more C-boutons, but are lost earlier in the disease. To circumvent these issues, we quantified the frequency and volume of C-boutons on traced soleus and gastrocnemius motoneurones, representing predominantly slow and fast motor pools respectively. Experiments were performed using the TDP-43ΔNLS mouse model that carries a transgenic construct of TDP-43 devoid of its nuclear localization signal, preventing its nuclear import. This results in the emergence of pathological TDP-43 inclusions in the cytoplasm, modelling the main pathology seen in this disorder, accompanied by a severe and lethal ALS phenotype. Our results confirmed changes in both the number and volume of C-boutons with a decrease in number on the more vulnerable, predominantly fast gastrocnemius motoneurones and an increase in number on the less vulnerable, predominantly slow soleus motoneurones. Importantly, these changes were only found in male mice. However, both sexes and motor pools showed a decrease in C-bouton volume. Our experiments confirm that cytoplasmic TDP-43 accumulation is sufficient to drive C-bouton changes.
Collapse
Affiliation(s)
| | - Svetlana Djukic
- Department of Neuroscience, University of Copenhagen, Denmark
| | | | | | | | | |
Collapse
|
9
|
Vullhorst D, Bloom MS, Akella N, Buonanno A. ER-PM Junctions on GABAergic Interneurons Are Organized by Neuregulin 2/VAP Interactions and Regulated by NMDA Receptors. Int J Mol Sci 2023; 24:2908. [PMID: 36769244 PMCID: PMC9917868 DOI: 10.3390/ijms24032908] [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: 01/03/2023] [Revised: 01/24/2023] [Accepted: 01/27/2023] [Indexed: 02/05/2023] Open
Abstract
Neuregulins (NRGs) signal via ErbB receptors to regulate neural development, excitability, synaptic and network activity, and behaviors relevant to psychiatric disorders. Bidirectional signaling between NRG2/ErbB4 and NMDA receptors is thought to homeostatically regulate GABAergic interneurons in response to increased excitatory neurotransmission or elevated extracellular glutamate levels. Unprocessed proNRG2 forms discrete clusters on cell bodies and proximal dendrites that colocalize with the potassium channel Kv2.1 at specialized endoplasmic reticulum-plasma membrane (ER-PM) junctions, and NMDA receptor activation triggers rapid dissociation from ER-PM junctions and ectodomain shedding by ADAM10. Here, we elucidate the mechanistic basis of proNRG2 clustering at ER-PM junctions and its regulation by NMDA receptors. Importantly, we demonstrate that proNRG2 promotes the formation of ER-PM junctions by directly binding the ER-resident membrane tether VAP, like Kv2.1. The proNRG2 intracellular domain harbors two non-canonical, low-affinity sites that cooperatively mediate VAP binding. One of these is a cryptic and phosphorylation-dependent VAP binding motif that is dephosphorylated following NMDA receptor activation, thus revealing how excitatory neurotransmission promotes the dissociation of proNRG2 from ER-PM junctions. Therefore, proNRG2 and Kv2.1 can independently function as VAP-dependent organizers of neuronal ER-PM junctions. Based on these and prior studies, we propose that proNRG2 and Kv2.1 serve as co-regulated downstream effectors of NMDA receptors to homeostatically regulate GABAergic interneurons.
Collapse
Affiliation(s)
- Detlef Vullhorst
- Section on Molecular Neurobiology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, Bethesda, MD 20892, USA
| | | | | | | |
Collapse
|
10
|
Salvany S, Casanovas A, Piedrafita L, Gras S, Calderó J, Esquerda JE. Accumulation of misfolded SOD1 outlines distinct patterns of motor neuron pathology and death during disease progression in a SOD1 G93A mouse model of amyotrophic lateral sclerosis. Brain Pathol 2022; 32:e13078. [PMID: 35584812 PMCID: PMC9616096 DOI: 10.1111/bpa.13078] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Accepted: 04/11/2022] [Indexed: 12/12/2022] Open
Abstract
Early misfolded superoxide dismutase 1 (mfSOD1) accumulation, motor neuron (MN) degeneration, and microgliosis are hallmark pathological features in SOD1G93A amyotrophic lateral sclerosis (ALS) mice. Because of the different vulnerabilities of distinct MN subtypes, degenerating and surviving MNs coexist in different proportions during disease progression. By examining the expression of misfolded conformers of SOD1 using specific antibodies, we defined distinct MN phenotypes that were evaluated during disease progression and the local neuroinflammatory reaction. The most severe phenotype corresponded to somata of fast‐twitch subtype MNs, which exhibited highly positive mfSOD1 immunostaining and an extreme degree of vacuolar degeneration. Vacuoles, which are of mitochondrial origin, contain mfSOD1 in conjunction with nonmitochondrial proteins, such as chromogranin, CD81, and flotillin. The fusion of ER‐derived vesicles enriched in mfSOD1 with outer mitochondrial membranes is thought to be the primary mechanism for vacuole formation. In addition, the ulterior coalescence of enlarged mitochondria may lead to the formation of giant vacuoles. Vacuolar degeneration is a transient degenerative process occurring early during the presymptomatic stages of the disease in ALS mice. Some vacuolated MNs are also positive for pMLKL, the effector protein of necroptosis. This indicates a newly described mechanism in which extracellular vesicles derived from damaged MNs, via cellular secretion or necroptotic disruption, may be the triggers for initiating neuroinflammation, glial‐mediated neurotoxicity, and disease spreading. Furthermore, as MN degeneration in mutant SOD1 mice is noncell autonomous, the effects of experimentally increasing or decreasing the microglial response on the expression of MN phenotypes were also evaluated, demonstrating bidirectional cross talk signaling between the degree of expression of mfSOD1 and local neuroinflammation. More detailed knowledge regarding these processes occurring long before the end stages of the disease is necessary to identify novel molecular targets for future preclinical testing.
Collapse
Affiliation(s)
- Sara Salvany
- Patologia Neuromuscular Experimental, Departament de Medicina Experimental, Facultat de Medicina, Universitat de Lleida and Institut de Recerca Biomèdica de Lleida (IRBLleida), Lleida, Catalonia, Spain
| | - Anna Casanovas
- Patologia Neuromuscular Experimental, Departament de Medicina Experimental, Facultat de Medicina, Universitat de Lleida and Institut de Recerca Biomèdica de Lleida (IRBLleida), Lleida, Catalonia, Spain
| | - Lídia Piedrafita
- Patologia Neuromuscular Experimental, Departament de Medicina Experimental, Facultat de Medicina, Universitat de Lleida and Institut de Recerca Biomèdica de Lleida (IRBLleida), Lleida, Catalonia, Spain
| | - Sílvia Gras
- Patologia Neuromuscular Experimental, Departament de Medicina Experimental, Facultat de Medicina, Universitat de Lleida and Institut de Recerca Biomèdica de Lleida (IRBLleida), Lleida, Catalonia, Spain
| | - Jordi Calderó
- Patologia Neuromuscular Experimental, Departament de Medicina Experimental, Facultat de Medicina, Universitat de Lleida and Institut de Recerca Biomèdica de Lleida (IRBLleida), Lleida, Catalonia, Spain
| | - Josep E Esquerda
- Patologia Neuromuscular Experimental, Departament de Medicina Experimental, Facultat de Medicina, Universitat de Lleida and Institut de Recerca Biomèdica de Lleida (IRBLleida), Lleida, Catalonia, Spain
| |
Collapse
|
11
|
Recabal-Beyer AJ, Senecal JMM, Senecal JEM, Lynn BD, Nagy JI. On the Organization of Connexin36 Expression in Electrically Coupled Cholinergic V0c Neurons (Partition Cells) in the Spinal Cord and Their C-terminal Innervation of Motoneurons. Neuroscience 2022; 485:91-115. [PMID: 35090881 DOI: 10.1016/j.neuroscience.2022.01.015] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2021] [Revised: 01/07/2022] [Accepted: 01/19/2022] [Indexed: 12/13/2022]
Abstract
Large cholinergic neurons (V0c neurons; aka, partition cells) in the spinal cord project profusely to motoneurons on which they form C-terminal contacts distinguished by their specialized postsynaptic subsurface cisterns (SSCs). The V0c neurons are known to be rhythmically active during locomotion and release of acetylcholine (ACh) from their terminals is known to modulate the excitability of motoneurons in what appears to be a task-dependent manner. Here, we present evidence that a subpopulation of V0c neurons express the gap junction forming protein connexin36 (Cx36), indicating that they are coupled by electrical synapses. Based on immunofluorescence imaging and the use of Cx36BAC-enhanced green fluorescent protein (eGFP) mice in which C-terminals immunolabelled for their marker vesicular acetylcholine transporter (vAChT) are also labelled for eGFP, we found a heterogeneous distribution of eGFP+ C-terminals on motoneurons at cervical, thoracic and lumber spinal levels. The density of C-terminals on motoneurons varied as did the proportion of those that were eGFP+ vs. eGFP-. We present evidence that fast vs. slow motoneurons have a greater abundance of these terminals and fast motoneurons also have the highest density that were eGFP+. Thus, our results indicate that a subpopulation of V0c neurons projects preferentially to fast motoneurons, suggesting that the capacity for synchronous activity conferred by electrical synapses among networks of coupled V0c neurons enhances their dynamic capabilities for synchronous regulation of motoneuron excitability during high muscle force generation. The eGFP+ vs. eGFP- V0c neurons were more richly innervated by serotonergic terminals, suggesting their greater propensity for regulation by descending serotonergic systems.
Collapse
Affiliation(s)
- A J Recabal-Beyer
- Department of Physiology and Pathophysiology, Rady Faculty of Health Sciences, Max Rady College of Medicine, University of Manitoba, Winnipeg, Canada
| | - J M M Senecal
- Department of Physiology and Pathophysiology, Rady Faculty of Health Sciences, Max Rady College of Medicine, University of Manitoba, Winnipeg, Canada
| | - J E M Senecal
- Department of Physiology and Pathophysiology, Rady Faculty of Health Sciences, Max Rady College of Medicine, University of Manitoba, Winnipeg, Canada
| | - B D Lynn
- Department of Physiology and Pathophysiology, Rady Faculty of Health Sciences, Max Rady College of Medicine, University of Manitoba, Winnipeg, Canada
| | - J I Nagy
- Department of Physiology and Pathophysiology, Rady Faculty of Health Sciences, Max Rady College of Medicine, University of Manitoba, Winnipeg, Canada.
| |
Collapse
|
12
|
Lorca C, Mulet M, Arévalo-Caro C, Sanchez MÁ, Perez A, Perrino M, Bach-Faig A, Aguilar-Martínez A, Vilella E, Gallart-Palau X, Serra A. Plant-derived nootropics and human cognition: A systematic review. Crit Rev Food Sci Nutr 2022; 63:5521-5545. [PMID: 34978226 DOI: 10.1080/10408398.2021.2021137] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
Substances with modulatory capabilities on certain aspects of human cognition have been revered as nootropics from the dawn of time. The plant kingdom provides most of the currently available nootropics of natural origin. Here, in this systematic review, we aim to provide state-of-the-art information regarding proven and unproven effects of plant-derived nootropics (PDNs) on human cognition in conditions of health and disease. Six independent searches, one for each neurocognitive domain (NCD), were performed in parallel using three independent scientific library databases: PubMed, Cochrane and Scopus. Only scientific studies and systematic reviews with humans published between January 2000 and November 2021 were reviewed, and 256 papers were included. Ginkgo biloba was the most relevant nootropic regarding perceptual and motor functions. Bacopa monnieri improves language, learning and memory. Withania somnifera (Ashwagandha) modulates anxiety and social-related cognitions. Caffeine enhances attention and executive functions. Together, the results from the compiled studies highlight the nootropic effects and the inconsistencies regarding PDNs that require further research.Supplemental data for this article is available online at https://doi.org/10.1080/10408398.2021.2021137.
Collapse
Affiliation(s)
- Cristina Lorca
- IMDEA-Food Research Institute, +Pec Proteomics, Campus of International Excellence UAM + CSIC, Old Cantoblanco Hospital, Madrid, Spain
- Biomedical Research Institute of Lleida Dr. Pifarré Foundation (IRB Lleida) - Neuroscience Area - University Hospital Arnau de Vilanova (HUAV) - School of Medicine, University of Lleida (UdL), Lleida, Spain
- Proteored - Instituto de Salud Carlos III (ISCIII), Madrid, Spain
| | - María Mulet
- Biomedical Research Institute of Lleida Dr. Pifarré Foundation (IRB Lleida) - Neuroscience Area - University Hospital Arnau de Vilanova (HUAV) - School of Medicine, University of Lleida (UdL), Lleida, Spain
| | - Catalina Arévalo-Caro
- Biomedical Research Institute of Lleida Dr. Pifarré Foundation (IRB Lleida) - Neuroscience Area - University Hospital Arnau de Vilanova (HUAV) - School of Medicine, University of Lleida (UdL), Lleida, Spain
| | - M Ángeles Sanchez
- IMDEA-Food Research Institute, +Pec Proteomics, Campus of International Excellence UAM + CSIC, Old Cantoblanco Hospital, Madrid, Spain
| | - Ainhoa Perez
- IMDEA-Food Research Institute, +Pec Proteomics, Campus of International Excellence UAM + CSIC, Old Cantoblanco Hospital, Madrid, Spain
| | - María Perrino
- IMDEA-Food Research Institute, +Pec Proteomics, Campus of International Excellence UAM + CSIC, Old Cantoblanco Hospital, Madrid, Spain
| | - Anna Bach-Faig
- FoodLab Research Group (2017SGR 83), Faculty of Health Sciences, Open University of Catalonia (UOC), Barcelona, Spain
- Food and Nutrition Area, Barcelona Official College of Pharmacists, Barcelona, Spain
| | - Alicia Aguilar-Martínez
- FoodLab Research Group (2017SGR 83), Faculty of Health Sciences, Open University of Catalonia (UOC), Barcelona, Spain
| | - Elisabet Vilella
- Hospital Universitari Institut Pere Mata (HUIPM), Institut Investigació Sanitària Pere Virgili (IISPV), Reus, Spain
- Universitat Rovira i Virgili (URV), Reus, Spain
- Centro de investigación Biomédica en Salud Mental CIBERSAM, Instituto de Salud Carlos III, Madrid, Spain
| | - Xavier Gallart-Palau
- Biomedical Research Institute of Lleida Dr. Pifarré Foundation (IRB Lleida) - Neuroscience Area - University Hospital Arnau de Vilanova (HUAV) - School of Medicine, University of Lleida (UdL), Lleida, Spain
- Proteored - Instituto de Salud Carlos III (ISCIII), Madrid, Spain
- Hospital Universitari Institut Pere Mata (HUIPM), Institut Investigació Sanitària Pere Virgili (IISPV), Reus, Spain
- Centro de investigación Biomédica en Salud Mental CIBERSAM, Instituto de Salud Carlos III, Madrid, Spain
| | - Aida Serra
- IMDEA-Food Research Institute, +Pec Proteomics, Campus of International Excellence UAM + CSIC, Old Cantoblanco Hospital, Madrid, Spain
- Biomedical Research Institute of Lleida Dr. Pifarré Foundation (IRB Lleida) - Neuroscience Area - University Hospital Arnau de Vilanova (HUAV) - School of Medicine, University of Lleida (UdL), Lleida, Spain
- Proteored - Instituto de Salud Carlos III (ISCIII), Madrid, Spain
| |
Collapse
|
13
|
Abstract
Motoneurons are the 'final common path' between the central nervous system (that intends, selects, commands, and organises movement) and muscles (that produce the behaviour). Motoneurons are not passive relays, but rather integrate synaptic activity to appropriately tune output (spike trains) and therefore the production of muscle force. In this chapter, we focus on studies of mammalian motoneurons, describing their heterogeneity whilst providing a brief historical account of motoneuron recording techniques. Next, we describe adult motoneurons in terms of their passive, transition, and active (repetitive firing) properties. We then discuss modulation of these properties by somatic (C-boutons) and dendritic (persistent inward currents) mechanisms. Finally, we briefly describe select studies of human motor unit physiology and relate them to findings from animal preparations discussed earlier in the chapter. This interphyletic approach to the study of motoneuron physiology is crucial to progress understanding of how these diverse neurons translate intention into behaviour.
Collapse
|
14
|
Mòdol-Caballero G, Herrando-Grabulosa M, Verdés S, García-Lareu B, Hernández N, Francos-Quijorna I, López-Vales R, Bosch A, Navarro X. Gene Therapy Overexpressing Neuregulin 1 Type I in Combination With Neuregulin 1 Type III Promotes Functional Improvement in the SOD1 G93A ALS Mice. Front Neurol 2021; 12:693309. [PMID: 34630277 PMCID: PMC8492910 DOI: 10.3389/fneur.2021.693309] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2021] [Accepted: 08/26/2021] [Indexed: 11/13/2022] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a fatal neurodegenerative disease affecting the neuromuscular system for which currently there is no effective therapy. Motoneuron (MN) degeneration involves several complex mechanisms, including surrounding glial cells and skeletal muscle contributions. Neuregulin 1 (NRG1) is a trophic factor present particularly in MNs and neuromuscular junctions. Our previous studies revealed that gene therapy overexpressing the isoform I (NRG1-I) in skeletal muscles as well as overexpressing the isoform III (NRG1-III) directly in the central nervous system are both effective in preserving MNs in the spinal cord of ALS mice, opening novel therapeutic approaches. In this study, we combined administration of both viral vectors overexpressing NRG1-I in skeletal muscles and NRG1-III in spinal cord of the SOD1G93A mice in order to obtain a synergistic effect. The results showed that the combinatorial gene therapy increased preservation of MNs and of innervated neuromuscular junctions and reduced glial reactivity in the spinal cord of the treated SOD1G93A mice. Moreover, NRG1 isoforms overexpression improved motor function of hindlimb muscles and delayed the onset of clinical disease. However, this combinatory gene therapy did not produce a synergic effect compared with single therapies, suggesting an overlap between NRG1-I and NRG1-III activated pathways and their beneficial effects.
Collapse
Affiliation(s)
- Guillem Mòdol-Caballero
- Department of Cell Biology, Physiology and Immunology, Institute of Neurosciences, Universitat Autonoma de Barcelona, Barcelona, Spain
- Centro de Investigación Biomédica en Red Sobre Enfermedades Neurodegenerativas (CIBERNED), Barcelona, Spain
| | - Mireia Herrando-Grabulosa
- Department of Cell Biology, Physiology and Immunology, Institute of Neurosciences, Universitat Autonoma de Barcelona, Barcelona, Spain
- Centro de Investigación Biomédica en Red Sobre Enfermedades Neurodegenerativas (CIBERNED), Barcelona, Spain
| | - Sergi Verdés
- Department of Biochemistry and Molecular Biology, Institute of Neurosciences, Universitat Autònoma De Barcelona, Barcelona, Spain
- Unitat Mixta UAB-VHIR, Vall d'Hebron Institut de Recerca (VHIR), Barcelona, Spain
| | - Belén García-Lareu
- Centro de Investigación Biomédica en Red Sobre Enfermedades Neurodegenerativas (CIBERNED), Barcelona, Spain
- Department of Biochemistry and Molecular Biology, Institute of Neurosciences, Universitat Autònoma De Barcelona, Barcelona, Spain
| | - Neus Hernández
- Department of Cell Biology, Physiology and Immunology, Institute of Neurosciences, Universitat Autonoma de Barcelona, Barcelona, Spain
- Centro de Investigación Biomédica en Red Sobre Enfermedades Neurodegenerativas (CIBERNED), Barcelona, Spain
| | - Isaac Francos-Quijorna
- Department of Cell Biology, Physiology and Immunology, Institute of Neurosciences, Universitat Autonoma de Barcelona, Barcelona, Spain
- Centro de Investigación Biomédica en Red Sobre Enfermedades Neurodegenerativas (CIBERNED), Barcelona, Spain
| | - Rubén López-Vales
- Department of Cell Biology, Physiology and Immunology, Institute of Neurosciences, Universitat Autonoma de Barcelona, Barcelona, Spain
- Centro de Investigación Biomédica en Red Sobre Enfermedades Neurodegenerativas (CIBERNED), Barcelona, Spain
| | - Assumpció Bosch
- Centro de Investigación Biomédica en Red Sobre Enfermedades Neurodegenerativas (CIBERNED), Barcelona, Spain
- Department of Biochemistry and Molecular Biology, Institute of Neurosciences, Universitat Autònoma De Barcelona, Barcelona, Spain
- Unitat Mixta UAB-VHIR, Vall d'Hebron Institut de Recerca (VHIR), Barcelona, Spain
| | - Xavier Navarro
- Department of Cell Biology, Physiology and Immunology, Institute of Neurosciences, Universitat Autonoma de Barcelona, Barcelona, Spain
- Centro de Investigación Biomédica en Red Sobre Enfermedades Neurodegenerativas (CIBERNED), Barcelona, Spain
| |
Collapse
|
15
|
Sun L, Cheng B, Zhou Y, Fan Y, Li W, Qiu Q, Fang Y, Xiao S, Zheng H, Li X. ErbB4 Mutation that Decreased NRG1-ErbB4 Signaling Involved in the Pathogenesis of Amyotrophic Lateral Sclerosis/Frontotemporal Dementia. J Alzheimers Dis 2021; 74:535-544. [PMID: 32065797 DOI: 10.3233/jad-191230] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
BACKGROUND Amyotrophic lateral sclerosis/frontotemporal dementia (ALS/FTD) includes a large spectrum of neurodegenerative disorders. OBJECTIVE To identify the relationship of ErbB4 mutation and ALS/FTD. METHODS Here, we report an atypical case of frontal variant behavioral abnormalities at the initial stage, a stable plateau stage of 5 years, and paralysis involving both upper and lower motor neurons followed by progressive cognitive dysfunction at the advanced stage. The clinical findings suggested a diagnosis of ALS/FTD, and genetic testing revealed erb-b2 receptor tyrosine kinase 4 (ErbB4) heterozygous mutation (c.2136 T>G, p.I712M), identified in an ALS pedigree previously. We modeled mutant ErbB4 protein through the SWISS-MODEL Server, and speculated on the structural change caused by the mutation. We also identified that ErbB4 (I712M) mutation led to reduced auto-phosphorylation of ErbB4 upon neuregulin-1 (NRG1) stimulation. RESULTS A functional analysis of ErbB4 mutation demonstrated an obviously decreased auto-phosphorylation of ErbB4 involving in the pathogenesis of ALS/FTD. CONCLUSION We firstly found ErbB4 mutation to be identified in ALS/FTD.
Collapse
Affiliation(s)
- Lin Sun
- Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Baoying Cheng
- Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, Institute of Neuroscience, College of Medicine, Xiamen University, Xiamen, China.,Department of Neuroscience, Shenzhen Research Institute of Xiamen University, Shenzhen, China
| | - Yuxun Zhou
- The College of Chemistry, Chemical Engineering & Biotechnology, Donghua University, Shanghai, China
| | - Yating Fan
- The College of Chemistry, Chemical Engineering & Biotechnology, Donghua University, Shanghai, China
| | - Wei Li
- Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Qi Qiu
- Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yuan Fang
- Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Shifu Xiao
- Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Honghua Zheng
- Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, Institute of Neuroscience, College of Medicine, Xiamen University, Xiamen, China.,Department of Neuroscience, Shenzhen Research Institute of Xiamen University, Shenzhen, China
| | - Xia Li
- Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
16
|
Gras S, Blasco A, Mòdol-Caballero G, Tarabal O, Casanovas A, Piedrafita L, Barranco A, Das T, Rueda R, Pereira SL, Navarro X, Esquerda JE, Calderó J. Beneficial effects of dietary supplementation with green tea catechins and cocoa flavanols on aging-related regressive changes in the mouse neuromuscular system. Aging (Albany NY) 2021; 13:18051-18093. [PMID: 34319911 PMCID: PMC8351677 DOI: 10.18632/aging.203336] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Accepted: 06/19/2021] [Indexed: 12/17/2022]
Abstract
Besides skeletal muscle wasting, sarcopenia entails morphological and molecular changes in distinct components of the neuromuscular system, including spinal cord motoneurons (MNs) and neuromuscular junctions (NMJs); moreover, noticeable microgliosis has also been observed around aged MNs. Here we examined the impact of two flavonoid-enriched diets containing either green tea extract (GTE) catechins or cocoa flavanols on age-associated regressive changes in the neuromuscular system of C57BL/6J mice. Compared to control mice, GTE- and cocoa-supplementation significantly improved the survival rate of mice, reduced the proportion of fibers with lipofuscin aggregates and central nuclei, and increased the density of satellite cells in skeletal muscles. Additionally, both supplements significantly augmented the number of innervated NMJs and their degree of maturity compared to controls. GTE, but not cocoa, prominently increased the density of VAChT and VGluT2 afferent synapses on MNs, which were lost in control aged spinal cords; conversely, cocoa, but not GTE, significantly augmented the proportion of VGluT1 afferent synapses on aged MNs. Moreover, GTE, but not cocoa, reduced aging-associated microgliosis and increased the proportion of neuroprotective microglial phenotypes. Our data indicate that certain plant flavonoids may be beneficial in the nutritional management of age-related deterioration of the neuromuscular system.
Collapse
Affiliation(s)
- Sílvia Gras
- Unitat de Neurobiologia Cel·lular, Departament de Medicina Experimental, Facultat de Medicina, Universitat de Lleida and Institut de Recerca Biomèdica de Lleida (IRBLleida), Lleida, Spain
| | - Alba Blasco
- Unitat de Neurobiologia Cel·lular, Departament de Medicina Experimental, Facultat de Medicina, Universitat de Lleida and Institut de Recerca Biomèdica de Lleida (IRBLleida), Lleida, Spain
| | - Guillem Mòdol-Caballero
- Grup de Neuroplasticitat i Regeneració, Institut de Neurociències, Departament de Biologia Cellular, Fisiologia i Immunologia, Universitat Autònoma de Barcelona and CIBERNED, Bellaterra, Spain
| | - Olga Tarabal
- Unitat de Neurobiologia Cel·lular, Departament de Medicina Experimental, Facultat de Medicina, Universitat de Lleida and Institut de Recerca Biomèdica de Lleida (IRBLleida), Lleida, Spain
| | - Anna Casanovas
- Unitat de Neurobiologia Cel·lular, Departament de Medicina Experimental, Facultat de Medicina, Universitat de Lleida and Institut de Recerca Biomèdica de Lleida (IRBLleida), Lleida, Spain
| | - Lídia Piedrafita
- Unitat de Neurobiologia Cel·lular, Departament de Medicina Experimental, Facultat de Medicina, Universitat de Lleida and Institut de Recerca Biomèdica de Lleida (IRBLleida), Lleida, Spain
| | - Alejandro Barranco
- Department of Biochemistry and Molecular Biology II, School of Pharmacy, University of Granada, Granada, Spain
| | - Tapas Das
- Abbott Nutrition, Research and Development, Columbus, OH 43215, USA
| | - Ricardo Rueda
- Abbott Nutrition, Research and Development, Granada, Spain
| | | | - Xavier Navarro
- Grup de Neuroplasticitat i Regeneració, Institut de Neurociències, Departament de Biologia Cellular, Fisiologia i Immunologia, Universitat Autònoma de Barcelona and CIBERNED, Bellaterra, Spain
| | - Josep E. Esquerda
- Unitat de Neurobiologia Cel·lular, Departament de Medicina Experimental, Facultat de Medicina, Universitat de Lleida and Institut de Recerca Biomèdica de Lleida (IRBLleida), Lleida, Spain
| | - Jordi Calderó
- Unitat de Neurobiologia Cel·lular, Departament de Medicina Experimental, Facultat de Medicina, Universitat de Lleida and Institut de Recerca Biomèdica de Lleida (IRBLleida), Lleida, Spain
| |
Collapse
|
17
|
Morphological Heterogeneity of the Endoplasmic Reticulum within Neurons and Its Implications in Neurodegeneration. Cells 2021; 10:cells10050970. [PMID: 33919188 PMCID: PMC8143122 DOI: 10.3390/cells10050970] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2021] [Revised: 04/13/2021] [Accepted: 04/19/2021] [Indexed: 12/19/2022] Open
Abstract
The endoplasmic reticulum (ER) is a multipurpose organelle comprising dynamic structural subdomains, such as ER sheets and tubules, serving to maintain protein, calcium, and lipid homeostasis. In neurons, the single ER is compartmentalized with a careful segregation of the structural subdomains in somatic and neurite (axodendritic) regions. The distribution and arrangement of these ER subdomains varies between different neuronal types. Mutations in ER membrane shaping proteins and morphological changes in the ER are associated with various neurodegenerative diseases implying significance of ER morphology in maintaining neuronal integrity. Specific neurons, such as the highly arborized dopaminergic neurons, are prone to stress and neurodegeneration. Differences in morphology and functionality of ER between the neurons may account for their varied sensitivity to stress and neurodegenerative changes. In this review, we explore the neuronal ER and discuss its distinct morphological attributes and specific functions. We hypothesize that morphological heterogeneity of the ER in neurons is an important factor that accounts for their selective susceptibility to neurodegeneration.
Collapse
|
18
|
Mille T, Quilgars C, Cazalets J, Bertrand SS. Acetylcholine and spinal locomotor networks: The insider. Physiol Rep 2021; 9:e14736. [PMID: 33527727 PMCID: PMC7851432 DOI: 10.14814/phy2.14736] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2020] [Revised: 01/05/2021] [Accepted: 01/07/2021] [Indexed: 01/07/2023] Open
Abstract
This article aims to review studies that have investigated the role of neurons that use the transmitter acetylcholine (ACh) in controlling the operation of locomotor neural networks within the spinal cord. This cholinergic system has the particularity of being completely intraspinal. We describe the different effects exerted by spinal cholinergic neurons on locomotor circuitry by the pharmacological activation or blockade of this propriospinal system, as well as describing its different cellular and subcellular targets. Through the activation of one ionotropic receptor, the nicotinic receptor, and five metabotropic receptors, the M1 to M5 muscarinic receptors, the cholinergic system exerts a powerful control both on synaptic transmission and locomotor network neuron excitability. Although tremendous advances have been made in our understanding of the spinal cholinergic system's involvement in the physiology and pathophysiology of locomotor networks, gaps still remain, including the precise role of the different subtypes of cholinergic neurons as well as their pre- and postsynaptic partners. Improving our knowledge of the propriospinal cholinergic system is of major relevance to finding new cellular targets and therapeutics in countering the debilitating effects of neurodegenerative diseases and restoring motor functions after spinal cord injury.
Collapse
Affiliation(s)
- Théo Mille
- Université de BordeauxCNRS UMR 5287INCIABordeauxFrance
| | | | | | | |
Collapse
|
19
|
Deardorff AS, Romer SH, Fyffe RE. Location, location, location: the organization and roles of potassium channels in mammalian motoneurons. J Physiol 2021; 599:1391-1420. [DOI: 10.1113/jp278675] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2020] [Accepted: 01/08/2021] [Indexed: 11/08/2022] Open
Affiliation(s)
- Adam S. Deardorff
- Department of Neuroscience, Cell Biology and Physiology, Wright State University Boonshoft School of Medicine Dayton OH 45435 USA
- Department of Neurology and Internal Medicine, Wright State University Boonshoft School of Medicine Dayton OH 45435 USA
| | - Shannon H. Romer
- Odyssey Systems Environmental Health Effects Laboratory, Navy Medical Research Unit‐Dayton Wright‐Patterson Air Force Base OH 45433 USA
| | - Robert E.W. Fyffe
- Department of Neuroscience, Cell Biology and Physiology, Wright State University Boonshoft School of Medicine Dayton OH 45435 USA
| |
Collapse
|
20
|
Konsolaki E, Koropouli E, Tsape E, Pothakos K, Zagoraiou L. Genetic Inactivation of Cholinergic C Bouton Output Improves Motor Performance but not Survival in a Mouse Model of Amyotrophic Lateral Sclerosis. Neuroscience 2020; 450:71-80. [DOI: 10.1016/j.neuroscience.2020.07.047] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2020] [Revised: 07/06/2020] [Accepted: 07/26/2020] [Indexed: 12/17/2022]
|
21
|
Blasco A, Gras S, Mòdol-Caballero G, Tarabal O, Casanovas A, Piedrafita L, Barranco A, Das T, Pereira SL, Navarro X, Rueda R, Esquerda JE, Calderó J. Motoneuron deafferentation and gliosis occur in association with neuromuscular regressive changes during ageing in mice. J Cachexia Sarcopenia Muscle 2020; 11:1628-1660. [PMID: 32691534 PMCID: PMC7749545 DOI: 10.1002/jcsm.12599] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/03/2020] [Revised: 06/05/2020] [Accepted: 06/15/2020] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND The cellular mechanisms underlying the age-associated loss of muscle mass and function (sarcopenia) are poorly understood, hampering the development of effective treatment strategies. Here, we performed a detailed characterization of age-related pathophysiological changes in the mouse neuromuscular system. METHODS Young, adult, middle-aged, and old (1, 4, 14, and 24-30 months old, respectively) C57BL/6J mice were used. Motor behavioural and electrophysiological tests and histological and immunocytochemical procedures were carried out to simultaneously analyse structural, molecular, and functional age-related changes in distinct cellular components of the neuromuscular system. RESULTS Ageing was not accompanied by a significant loss of spinal motoneurons (MNs), although a proportion (~15%) of them in old mice exhibited an abnormally dark appearance. Dark MNs were also observed in adult (~9%) and young (~4%) animals, suggesting that during ageing, some MNs undergo early deleterious changes, which may not lead to MN death. Old MNs were depleted of cholinergic and glutamatergic inputs (~40% and ~45%, respectively, P < 0.01), suggestive of age-associated alterations in MN excitability. Prominent microgliosis and astrogliosis [~93% (P < 0.001) and ~100% (P < 0.0001) increase vs. adults, respectively] were found in old spinal cords, with increased density of pro-inflammatory M1 microglia and A1 astroglia (25-fold and 4-fold increase, respectively, P < 0.0001). Ageing resulted in significant reductions in the nerve conduction velocity and the compound muscle action potential amplitude (~30%, P < 0.05, vs. adults) in old distal plantar muscles. Compared with adult muscles, old muscles exhibited significantly higher numbers of both denervated and polyinnervated neuromuscular junctions, changes in fibre type composition, higher proportion of fibres showing central nuclei and lipofuscin aggregates, depletion of satellite cells, and augmented expression of different molecules related to development, plasticity, and maintenance of neuromuscular junctions, including calcitonin gene-related peptide, growth associated protein 43, agrin, fibroblast growth factor binding protein 1, and transforming growth factor-β1. Overall, these alterations occurred at varying degrees in all the muscles analysed, with no correlation between the age-related changes observed and myofiber type composition or muscle topography. CONCLUSIONS Our data provide a global view of age-associated neuromuscular changes in a mouse model of ageing and help to advance understanding of contributing pathways leading to development of sarcopenia.
Collapse
Affiliation(s)
- Alba Blasco
- Unitat de Neurobiologia Cel·lular, Departament de Medicina Experimental, Facultat de Medicina, Universitat de Lleida, Institut de Recerca Biomèdica de Lleida (IRBLleida), Lleida, Spain
| | - Sílvia Gras
- Unitat de Neurobiologia Cel·lular, Departament de Medicina Experimental, Facultat de Medicina, Universitat de Lleida, Institut de Recerca Biomèdica de Lleida (IRBLleida), Lleida, Spain
| | - Guillem Mòdol-Caballero
- Grup de Neuroplasticitat i Regeneració, Institut de Neurociències, Departament de Biologia Cel·lular, Fisiologia i Immunologia, Universitat Autònoma de Barcelona, CIBERNED, Bellaterra, Spain
| | - Olga Tarabal
- Unitat de Neurobiologia Cel·lular, Departament de Medicina Experimental, Facultat de Medicina, Universitat de Lleida, Institut de Recerca Biomèdica de Lleida (IRBLleida), Lleida, Spain
| | - Anna Casanovas
- Unitat de Neurobiologia Cel·lular, Departament de Medicina Experimental, Facultat de Medicina, Universitat de Lleida, Institut de Recerca Biomèdica de Lleida (IRBLleida), Lleida, Spain
| | - Lídia Piedrafita
- Unitat de Neurobiologia Cel·lular, Departament de Medicina Experimental, Facultat de Medicina, Universitat de Lleida, Institut de Recerca Biomèdica de Lleida (IRBLleida), Lleida, Spain
| | | | - Tapas Das
- Abbott Nutrition Research and Development, Columbus, OH, USA
| | | | - Xavier Navarro
- Grup de Neuroplasticitat i Regeneració, Institut de Neurociències, Departament de Biologia Cel·lular, Fisiologia i Immunologia, Universitat Autònoma de Barcelona, CIBERNED, Bellaterra, Spain
| | - Ricardo Rueda
- Abbott Nutrition Research and Development, Granada, Spain
| | - Josep E Esquerda
- Unitat de Neurobiologia Cel·lular, Departament de Medicina Experimental, Facultat de Medicina, Universitat de Lleida, Institut de Recerca Biomèdica de Lleida (IRBLleida), Lleida, Spain
| | - Jordi Calderó
- Unitat de Neurobiologia Cel·lular, Departament de Medicina Experimental, Facultat de Medicina, Universitat de Lleida, Institut de Recerca Biomèdica de Lleida (IRBLleida), Lleida, Spain
| |
Collapse
|
22
|
Crabé R, Aimond F, Gosset P, Scamps F, Raoul C. How Degeneration of Cells Surrounding Motoneurons Contributes to Amyotrophic Lateral Sclerosis. Cells 2020; 9:cells9122550. [PMID: 33260927 PMCID: PMC7760029 DOI: 10.3390/cells9122550] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2020] [Revised: 11/19/2020] [Accepted: 11/24/2020] [Indexed: 12/13/2022] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a fatal neurological disorder characterized by the progressive degeneration of upper and lower motoneurons. Despite motoneuron death being recognized as the cardinal event of the disease, the loss of glial cells and interneurons in the brain and spinal cord accompanies and even precedes motoneuron elimination. In this review, we provide striking evidence that the degeneration of astrocytes and oligodendrocytes, in addition to inhibitory and modulatory interneurons, disrupt the functionally coherent environment of motoneurons. We discuss the extent to which the degeneration of glial cells and interneurons also contributes to the decline of the motor system. This pathogenic cellular network therefore represents a novel strategic field of therapeutic investigation.
Collapse
Affiliation(s)
- Roxane Crabé
- The Neuroscience Institute of Montpellier, INSERM, UMR1051, University of Montpellier, 34091 Montpellier, France; (R.C.); (F.A.); (P.G.); (F.S.)
| | - Franck Aimond
- The Neuroscience Institute of Montpellier, INSERM, UMR1051, University of Montpellier, 34091 Montpellier, France; (R.C.); (F.A.); (P.G.); (F.S.)
| | - Philippe Gosset
- The Neuroscience Institute of Montpellier, INSERM, UMR1051, University of Montpellier, 34091 Montpellier, France; (R.C.); (F.A.); (P.G.); (F.S.)
| | - Frédérique Scamps
- The Neuroscience Institute of Montpellier, INSERM, UMR1051, University of Montpellier, 34091 Montpellier, France; (R.C.); (F.A.); (P.G.); (F.S.)
| | - Cédric Raoul
- The Neuroscience Institute of Montpellier, INSERM, UMR1051, University of Montpellier, 34091 Montpellier, France; (R.C.); (F.A.); (P.G.); (F.S.)
- Laboratory of Neurobiology, Kazan Federal University, 420008 Kazan, Russia
- Correspondence:
| |
Collapse
|
23
|
Herrando-Grabulosa M, Gaja-Capdevila N, Vela JM, Navarro X. Sigma 1 receptor as a therapeutic target for amyotrophic lateral sclerosis. Br J Pharmacol 2020; 178:1336-1352. [PMID: 32761823 DOI: 10.1111/bph.15224] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Revised: 07/13/2020] [Accepted: 07/25/2020] [Indexed: 12/13/2022] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is an adult disease causing a progressive loss of upper and lower motoneurons, muscle paralysis and early death. ALS has a poor prognosis of 3-5 years after diagnosis with no effective cure. The aetiopathogenic mechanisms involved include glutamate excitotoxicity, oxidative stress, protein misfolding, mitochondrial alterations, disrupted axonal transport and inflammation. Sigma non-opioid intracellular receptor 1 (sigma 1 receptor) is a protein expressed in motoneurons, mainly found in the endoplasmic reticulum (ER) on the mitochondria-associated ER membrane (MAM) or in close contact with cholinergic postsynaptic sites. MAMs are sites that allow the assembly of several complexes implicated in essential survival cell functions. The sigma 1 receptor modulates essential mechanisms for motoneuron survival including excitotoxicity, calcium homeostasis, ER stress and mitochondrial dysfunction. This review updates sigma 1 receptor mechanisms and its alterations in ALS, focusing on MAM modulation, which may constitute a novel target for therapeutic strategies. LINKED ARTICLES: This article is part of a themed issue on Neurochemistry in Japan. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v178.6/issuetoc.
Collapse
Affiliation(s)
- Mireia Herrando-Grabulosa
- Institute of Neurosciences, Department Cell Biology, Physiology and Immunology, Universitat Autònoma de Barcelona, Bellaterra, Spain.,Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | - Núria Gaja-Capdevila
- Institute of Neurosciences, Department Cell Biology, Physiology and Immunology, Universitat Autònoma de Barcelona, Bellaterra, Spain.,Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | - José M Vela
- Esteve Pharmaceuticals S.A., Drug Discovery and Preclinical Development, Barcelona, Spain
| | - Xavier Navarro
- Institute of Neurosciences, Department Cell Biology, Physiology and Immunology, Universitat Autònoma de Barcelona, Bellaterra, Spain.,Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain.,Institut Guttmann de Neurorehabilitació, Badalona, Spain
| |
Collapse
|
24
|
Mòdol-Caballero G, García-Lareu B, Verdés S, Ariza L, Sánchez-Brualla I, Brocard F, Bosch A, Navarro X, Herrando-Grabulosa M. Therapeutic Role of Neuregulin 1 Type III in SOD1-Linked Amyotrophic Lateral Sclerosis. Neurotherapeutics 2020; 17:1048-1060. [PMID: 31965551 PMCID: PMC7609630 DOI: 10.1007/s13311-019-00811-7] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a devastating motoneuron (Mn) disease without effective cure currently available. Death of MNs in ALS is preceded by failure of neuromuscular junctions and axonal retraction. Neuregulin 1 (NRG1) is a neurotrophic factor highly expressed in MNs and neuromuscular junctions that support axonal and neuromuscular development and maintenance. NRG1 and its ErbB receptors are involved in ALS. Reduced NRG1 expression has been found in ALS patients and in the ALS SOD1G93A mouse model; however, the expression of the isoforms of NRG1 and its receptors is still controversial. Due to the reduced levels of NRG1 type III (NRG1-III) in the spinal cord of ALS patients, we used gene therapy based on intrathecal administration of adeno-associated virus to overexpress NRG1-III in SOD1G93A mice. The mice were evaluated from 9 to 16 weeks of age by electrophysiology and rotarod tests. At 16 weeks, samples were harvested for histological and molecular analyses. Our results indicate that overexpression of NRG1-III is able to preserve neuromuscular function of the hindlimbs, improve locomotor performance, increase the number of surviving MNs, and reduce glial reactivity in the treated female SOD1G93A mice. Furthermore, the NRG1-III/ErbB4 axis appears to regulate MN excitability by modulating the chloride transporter KCC2 and reduces the expression of the MN vulnerability marker MMP-9. However, NRG1-III did not have a significant effect on male mice, indicating relevant sex differences. These findings indicate that increasing NRG1-III at the spinal cord is a promising approach for promoting MN protection and functional improvement in ALS.
Collapse
Affiliation(s)
- Guillem Mòdol-Caballero
- Department of Cell Biology, Physiology and Immunology, Institute of Neurosciences, Universitat Autònoma de Barcelona, 08193, Bellaterra, Spain
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), 08193, Bellaterra, Spain
| | - Belén García-Lareu
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), 08193, Bellaterra, Spain
- Department of Biochemistry and Molecular Biology, Institute of Neurosciences, Universitat Autònoma de Barcelona, 08193, Bellaterra, Spain
| | - Sergi Verdés
- Department of Biochemistry and Molecular Biology, Institute of Neurosciences, Universitat Autònoma de Barcelona, 08193, Bellaterra, Spain
| | - Lorena Ariza
- Department of Biochemistry and Molecular Biology, Institute of Neurosciences, Universitat Autònoma de Barcelona, 08193, Bellaterra, Spain
| | - Irene Sánchez-Brualla
- Department of Cell Biology, Physiology and Immunology, Institute of Neurosciences, Universitat Autònoma de Barcelona, 08193, Bellaterra, Spain
- Team P3M, Institut de Neurosciences de la Timone, UMR7289, Aix-Marseille Université and Centre National de la Recherche Scientifique (CNRS), 13005, Marseille, France
| | - Frédéric Brocard
- Team P3M, Institut de Neurosciences de la Timone, UMR7289, Aix-Marseille Université and Centre National de la Recherche Scientifique (CNRS), 13005, Marseille, France
| | - Assumpció Bosch
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), 08193, Bellaterra, Spain
- Department of Biochemistry and Molecular Biology, Institute of Neurosciences, Universitat Autònoma de Barcelona, 08193, Bellaterra, Spain
| | - Xavier Navarro
- Department of Cell Biology, Physiology and Immunology, Institute of Neurosciences, Universitat Autònoma de Barcelona, 08193, Bellaterra, Spain.
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), 08193, Bellaterra, Spain.
| | - Mireia Herrando-Grabulosa
- Department of Cell Biology, Physiology and Immunology, Institute of Neurosciences, Universitat Autònoma de Barcelona, 08193, Bellaterra, Spain.
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), 08193, Bellaterra, Spain.
| |
Collapse
|
25
|
Andrés-Benito P, Povedano M, Torres P, Portero-Otín M, Ferrer I. Altered Dynein Axonemal Assembly Factor 1 Expression in C-Boutons in Bulbar and Spinal Cord Motor-Neurons in Sporadic Amyotrophic Lateral Sclerosis. J Neuropathol Exp Neurol 2020; 78:416-425. [PMID: 30939186 DOI: 10.1093/jnen/nlz019] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Dyneins are major components of microtubules. Dynein assembly is modulated by a heterogeneous group of dynein axonemal assembly factors (DNAAFs). The present study analyzes dynein axonemal assembly factor 1 (DNAAF1) and leucine-rich repeat-containing protein 50 (LRRC50), the corresponding encoded protein, in lower motor neurons in spinal cord of sALS postmortem samples and hSOD1-G93A transgenic mice compared with controls. DNAAF1 mRNA is significantly reduced in the anterior horn in sALS, and LRRC50 immunoreactivity is significantly reduced in C-boutons of the remaining motor neurons of the anterior horn, dorsal nucleus of the vagus nerve, and hypoglossal nuclei at terminal stages of ALS. LRRC50 immunoreactivity has a perinuclear distribution in motor neurons in sALS thus suggesting a disorder of transport. The number of LRRC50-/S1R-immunoreactive structures is also significantly decreased in hSOD1-G93A transgenic mice at the age of 90 days (preclinical stages), and the number of motor neurons with LRRC50-immunoreactive structures is significantly reduced in animals aged 150 days (clinical stages). These observations suggest cholinergic denervation of motor neurons as a pathogenic factor in motor neuron disease. LRRC50 protein levels were not detected in human CSF.
Collapse
Affiliation(s)
- Pol Andrés-Benito
- Department of Pathology and Experimental Therapeutics, University of Barcelona, Spain.,Institute Carlos III, Biomedical Network Research Center on Neurodegenerative Diseases (CIBERNED), Hospitalet de Llobregat, Spain.,Bellvitge Biomedical Research Institute (IDIBELL), Hospitalet de Llobregat, Barcelona, Spain
| | - Mònica Povedano
- Functional Unit of Amyotrophic Lateral Sclerosis (UFELA), Service of Neurology, Bellvitge University Hospital, Hospitalet de Llobregat, Spain
| | - Pascual Torres
- Departament Medicina Experimental, Facultat de Medicina, Universitat de Lleida, IRBLLEIDA, Lleida, Spain
| | - Manuel Portero-Otín
- Departament Medicina Experimental, Facultat de Medicina, Universitat de Lleida, IRBLLEIDA, Lleida, Spain
| | - Isidro Ferrer
- Department of Pathology and Experimental Therapeutics, University of Barcelona, Spain.,Institute Carlos III, Biomedical Network Research Center on Neurodegenerative Diseases (CIBERNED), Hospitalet de Llobregat, Spain.,Bellvitge Biomedical Research Institute (IDIBELL), Hospitalet de Llobregat, Barcelona, Spain.,Neuropathology, Pathologic Anatomy Service, Bellvitge University Hospital, IDIBELL, L'Hospitalet de Llobregat, Spain.,Institute of Neurosciences, University of Barcelona, Barcelona, Spain
| |
Collapse
|
26
|
Schram S, Loeb JA, Song F. Disease propagation in amyotrophic lateral sclerosis (ALS): an interplay between genetics and environment. J Neuroinflammation 2020; 17:175. [PMID: 32505190 PMCID: PMC7276078 DOI: 10.1186/s12974-020-01849-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2020] [Accepted: 05/19/2020] [Indexed: 12/11/2022] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a progressive, fatal disease affecting the neuromuscular system. While there have been a number of important genetic discoveries, there are no therapeutics capable of stopping its insidious progression. Lessons from clinical histories reveal that ALS can start focally at a single limb, but then segmentally spread up and down the spinal cord as well as in the motor cortex and cortex of frontal and temporal lobes until respiratory muscles fail. With or without a clear genetic etiology, often there is no explanation as to why it starts in one region of the body versus another. Similarly, once the disease starts the mechanisms by which the neurodegenerative process spreads are not known. Here, we summarize recent work in animal models that support the hypothesis that critical environmental contributions, such as a nerve injury, can initiate the disease process. We also propose that pathological axoglial signaling by the glial growth factor neuregulin-1 leads to the slow propagation of neuroinflammation resulting in neurodegeneration up and down the spinal cord and that locally applied drugs that block neuregulin-1 signaling could slow or halt the spread of disease.
Collapse
Affiliation(s)
- Sarah Schram
- Department of Neurology and Rehabilitation, University of Illinois at Chicago, NPI North Bldg., Room 657, M/C 796, 912 S. Wood Street, Chicago, IL, 60612, USA
| | - Jeffrey A Loeb
- Department of Neurology and Rehabilitation, University of Illinois at Chicago, NPI North Bldg., Room 657, M/C 796, 912 S. Wood Street, Chicago, IL, 60612, USA.
| | - Fei Song
- Department of Neurology and Rehabilitation, University of Illinois at Chicago, NPI North Bldg., Room 657, M/C 796, 912 S. Wood Street, Chicago, IL, 60612, USA.
| |
Collapse
|
27
|
Dragunas G, Woest ME, Nijboer S, Bos ST, van Asselt J, de Groot AP, Vohlídalová E, Vermeulen CJ, Ditz B, Vonk JM, Koppelman GH, van den Berge M, Ten Hacken NHT, Timens W, Munhoz CD, Prakash YS, Gosens R, Kistemaker LEM. Cholinergic neuroplasticity in asthma driven by TrkB signaling. FASEB J 2020; 34:7703-7717. [PMID: 32277855 PMCID: PMC7302963 DOI: 10.1096/fj.202000170r] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2020] [Revised: 03/23/2020] [Accepted: 03/24/2020] [Indexed: 12/11/2022]
Abstract
Parasympathetic neurons in the airways control bronchomotor tone. Increased activity of cholinergic neurons are mediators of airway hyperresponsiveness (AHR) in asthma, however, mechanisms are not elucidated. We describe remodeling of the cholinergic neuronal network in asthmatic airways driven by brain‐derived neurotrophic factor (BDNF) and Tropomyosin receptor kinase B (TrkB). Human bronchial biopsies were stained for cholinergic marker vesicular acetylcholine transporter (VAChT). Human lung gene expression and single nucleotide polymorphisms (SNP) in neuroplasticity‐related genes were compared between asthma and healthy patients. Wild‐type (WT) and mutated TrkB knock‐in mice (Ntrk2tm1Ddg/J) with impaired BDNF signaling were chronically exposed to ovalbumin (OVA). Neuronal VAChT staining and airway narrowing in response to electrical field stimulation in precision cut lung slices (PCLS) were assessed. Increased cholinergic fibers in asthmatic airway biopsies was found, paralleled by increased TrkB gene expression in human lung tissue, and SNPs in the NTRK2 [TrkB] and BDNF genes linked to asthma. Chronic allergen exposure in mice resulted in increased density of cholinergic nerves, which was prevented by inhibiting TrkB. Increased nerve density resulted in AHR in vivo and in increased nerve‐dependent airway reactivity in lung slices mediated via TrkB. These findings show cholinergic neuroplasticity in asthma driven by TrkB signaling and suggest that the BDNF‐TrkB pathway may be a potential target.
Collapse
Affiliation(s)
- Guilherme Dragunas
- Department of Molecular Pharmacology, University of Groningen, Groningen, the Netherlands.,GRIAC Research Institute, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands.,Department of Pharmacology, University of São Paulo, São Paulo, Brazil
| | - Manon E Woest
- Department of Molecular Pharmacology, University of Groningen, Groningen, the Netherlands.,GRIAC Research Institute, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Susan Nijboer
- Department of Molecular Pharmacology, University of Groningen, Groningen, the Netherlands.,GRIAC Research Institute, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Sophie T Bos
- Department of Molecular Pharmacology, University of Groningen, Groningen, the Netherlands.,GRIAC Research Institute, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Janet van Asselt
- Department of Molecular Pharmacology, University of Groningen, Groningen, the Netherlands.,GRIAC Research Institute, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Anne P de Groot
- Department of Molecular Pharmacology, University of Groningen, Groningen, the Netherlands.,GRIAC Research Institute, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Eva Vohlídalová
- Department of Molecular Pharmacology, University of Groningen, Groningen, the Netherlands.,GRIAC Research Institute, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Corneel J Vermeulen
- GRIAC Research Institute, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands.,Department of Pulmonary Diseases, UMCG, Groningen, the Netherlands
| | - Benedikt Ditz
- GRIAC Research Institute, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands.,Department of Pulmonary Diseases, UMCG, Groningen, the Netherlands
| | - Judith M Vonk
- GRIAC Research Institute, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands.,Department of Epidemiology, UMCG, Groningen, the Netherlands
| | - Gerard H Koppelman
- GRIAC Research Institute, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands.,Department of Pediatric Pulmonology and Pediatric Allergology, University Medical Center Groningen, University of Groningen, Beatrix Children's Hospital, Groningen, the Netherlands
| | - Maarten van den Berge
- GRIAC Research Institute, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands.,Department of Pulmonary Diseases, UMCG, Groningen, the Netherlands
| | - Nick H T Ten Hacken
- GRIAC Research Institute, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands.,Department of Pulmonary Diseases, UMCG, Groningen, the Netherlands
| | - Wim Timens
- GRIAC Research Institute, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands.,Department of Pathology, UMCG, Groningen, the Netherlands
| | - Carolina D Munhoz
- Department of Pharmacology, University of São Paulo, São Paulo, Brazil
| | - Y S Prakash
- Department of Anesthesiology and Perioperative Medicine, Mayo Clinic, Rochester, MN, USA
| | - Reinoud Gosens
- Department of Molecular Pharmacology, University of Groningen, Groningen, the Netherlands.,GRIAC Research Institute, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Loes E M Kistemaker
- Department of Molecular Pharmacology, University of Groningen, Groningen, the Netherlands.,GRIAC Research Institute, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| |
Collapse
|
28
|
Gatius A, Tarabal O, Cayuela P, Casanovas A, Piedrafita L, Salvany S, Hernández S, Soler RM, Esquerda JE, Calderó J. The Y172 Monoclonal Antibody Against p-c-Jun (Ser63) Is a Marker of the Postsynaptic Compartment of C-Type Cholinergic Afferent Synapses on Motoneurons. Front Cell Neurosci 2020; 13:582. [PMID: 32038174 PMCID: PMC6992659 DOI: 10.3389/fncel.2019.00582] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2019] [Accepted: 12/20/2019] [Indexed: 12/02/2022] Open
Abstract
C-bouton-type cholinergic afferents exert an important function in controlling motoneuron (MN) excitability. During the immunocytochemical analysis of the role of c-Jun in MNs with a monoclonal (clone Y172) antibody against phospho (p)-c-Jun (serine [Ser]63), unexpected labeling was identified in the cell body cytoplasm. As predicted for c-Jun in adult spinal cord, very few, if any MNs exhibited nuclear immunoreactivity with the Y172 antibody; conversely, virtually all MNs displayed strong Y172 immunostaining in cytoplasmic structures scattered throughout the soma and proximal dendrites. The majority of these cytoplasmic Y172-positive profiles was closely associated with VAChT-positive C-boutons, but not with other types of nerve afferents contacting MNs. Ultrastructural analysis revealed that cytoplasmic Y172 immunostaining was selectively located at the subsurface cistern (SSC) of C-boutons and also in the inner areas of the endoplasmic reticulum (ER). We also described changes in cytoplasmic Y172 immunoreactivity in injured and degenerating MNs. Moreover, we noticed that MNs from NRG1 type III-overexpressing transgenic mice, which show abnormally expanded SSCs, exhibited an increase in the density and size of peripherally located Y172-positive profiles. A similar immunocytochemical pattern to that of the Y172 antibody in MNs was found with a polyclonal antibody against p-c-Jun (Ser63) but not with another polyclonal antibody that recognizes c-Jun phosphorylated at a different site. No differential band patterns were found by western blotting with any of the antibodies against c-Jun or p-c-Jun used in our study. In cultured MNs, Y172-positive oval profiles were distributed in the cell body and proximal dendrites. The in vitro lentiviral-based knockdown of c-Jun resulted in a dramatic decrease in nuclear Y172 immunostaining in MNs without any reduction in the density of cytoplasmic Y172-positive profiles, suggesting that the synaptic antigen recognized by the antibody corresponds to a C-bouton-specific protein other than p-c-Jun. Our results lay the foundation for further studies aimed at identifying this protein and determining its role in this particular type of synapse.
Collapse
Affiliation(s)
- Alaó Gatius
- Unitat de Neurobiologia Cel·lular, Departament de Medicina Experimental, Facultat de Medicina, Universitat de Lleida and Institut de Recerca Biomèdica de Lleida (IRBLleida), Lleida, Spain
| | - Olga Tarabal
- Unitat de Neurobiologia Cel·lular, Departament de Medicina Experimental, Facultat de Medicina, Universitat de Lleida and Institut de Recerca Biomèdica de Lleida (IRBLleida), Lleida, Spain
| | - Paula Cayuela
- Unitat de Neurobiologia Cel·lular, Departament de Medicina Experimental, Facultat de Medicina, Universitat de Lleida and Institut de Recerca Biomèdica de Lleida (IRBLleida), Lleida, Spain
| | - Anna Casanovas
- Unitat de Neurobiologia Cel·lular, Departament de Medicina Experimental, Facultat de Medicina, Universitat de Lleida and Institut de Recerca Biomèdica de Lleida (IRBLleida), Lleida, Spain
| | - Lídia Piedrafita
- Unitat de Neurobiologia Cel·lular, Departament de Medicina Experimental, Facultat de Medicina, Universitat de Lleida and Institut de Recerca Biomèdica de Lleida (IRBLleida), Lleida, Spain
| | - Sara Salvany
- Unitat de Neurobiologia Cel·lular, Departament de Medicina Experimental, Facultat de Medicina, Universitat de Lleida and Institut de Recerca Biomèdica de Lleida (IRBLleida), Lleida, Spain
| | - Sara Hernández
- Unitat de Neurobiologia Cel·lular, Departament de Medicina Experimental, Facultat de Medicina, Universitat de Lleida and Institut de Recerca Biomèdica de Lleida (IRBLleida), Lleida, Spain
| | - Rosa M Soler
- Unitat de Senyalització Neuronal, Departament de Medicina Experimental, Facultat de Medicina, Universitat de Lleida and Institut de Recerca Biomèdica de Lleida (IRBLleida), Lleida, Spain
| | - Josep E Esquerda
- Unitat de Neurobiologia Cel·lular, Departament de Medicina Experimental, Facultat de Medicina, Universitat de Lleida and Institut de Recerca Biomèdica de Lleida (IRBLleida), Lleida, Spain
| | - Jordi Calderó
- Unitat de Neurobiologia Cel·lular, Departament de Medicina Experimental, Facultat de Medicina, Universitat de Lleida and Institut de Recerca Biomèdica de Lleida (IRBLleida), Lleida, Spain
| |
Collapse
|
29
|
Gene therapy for overexpressing Neuregulin 1 type I in skeletal muscles promotes functional improvement in the SOD1 G93A ALS mice. Neurobiol Dis 2020; 137:104793. [PMID: 32032731 DOI: 10.1016/j.nbd.2020.104793] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2019] [Revised: 01/10/2020] [Accepted: 02/03/2020] [Indexed: 12/11/2022] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a neurodegenerative disorder affecting motoneurons (MNs), with no effective treatment currently available. The molecular mechanisms that are involved in MN death are complex and not fully understood, with partial contributions of surrounding glial cells and skeletal muscle to the disease. Neuregulin 1 (NRG1) is a trophic factor highly expressed in MNs and neuromuscular junctions. Recent studies have suggested a crucial role of the isoform I (NRG1-I) in the collateral reinnervation process in skeletal muscle, and NRG1-III in the preservation of MNs in the spinal cord, opening a window for developing novel therapies for neuromuscular diseases like ALS. In this study, we overexpressed NRG1-I widely in the skeletal muscles of the SOD1G93A transgenic mouse. The results show that NRG1 gene therapy activated the survival pathways in muscle and spinal cord, increasing the number of surviving MNs and neuromuscular junctions and reducing the astroglial reactivity in the spinal cord of the treated SOD1G93A mice. Furthermore, NRG1-I overexpression preserved motor function and delayed the onset of clinical disease. In summary, our data indicates that NRG1 plays an important role on MN survival and muscle innervation in ALS, and that viral-mediated overexpression of NRG1 isoforms may be considered as a promising approach for ALS treatment.
Collapse
|
30
|
Serra A, Gallart-Palau X, Park JE, Lim GGY, Lim KL, Ho HH, Tam JP, Sze SK. Vascular Bed Molecular Profiling by Differential Systemic Decellularization In Vivo. Arterioscler Thromb Vasc Biol 2019; 38:2396-2409. [PMID: 30354219 DOI: 10.1161/atvbaha.118.311552] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Objective- Vascular endothelial dysfunction is a key component of several major human diseases, but the molecular basis of this complex disorder has been difficult to determine in vivo. Previous attempts to identify key mediators of vascular endothelial dysfunction in experimental models have been limited by the lack of suitable methods for system-wide analyses of vascular bed biology. Here, we aimed to develop a novel method for investigating vascular endothelial dysfunction pathogenesis that enables system-wide analyses of molecular interactions between endothelial glycocalyx, endothelial cells, and smooth muscle cells in murine. Approach and Results- We developed a new technique using whole-body differential perfusion with increasing concentrations of detergent buffer to selectively solubilize distinct layers of vascular bed tissue in rodents. When combined with proteomics techniques, our novel approach of differential systemic decellularization in vivo enabled quantitative profiling of vascular beds throughout the body. Initial perfusion with phosphate buffer was used to obtain the endothelial glycocalyx, followed by subsequent extraction of endothelial cell components, and finally by smooth muscle cell constituents with increasing concentrations of detergent. Differential systemic decellularization in vivo has also been successfully applied to characterize molecular events in the vascular bed pathology of lipopolysaccharide-challenged mice. Conclusions- Together, these data indicate that differential systemic decellularization in vivo permits system-wide molecular characterization of vascular bed proteomes in rodent models and can be used to advance our current understanding of vascular endothelial dysfunction pathogenesis and progression in a wide range of disease settings.
Collapse
Affiliation(s)
- Aida Serra
- From the School of Biological Sciences, Nanyang Technological University, Singapore (A.S., X.G.-P., J.E.P., J.P.T., S.K.S.)
| | - Xavier Gallart-Palau
- From the School of Biological Sciences, Nanyang Technological University, Singapore (A.S., X.G.-P., J.E.P., J.P.T., S.K.S.)
| | - Jung Eun Park
- From the School of Biological Sciences, Nanyang Technological University, Singapore (A.S., X.G.-P., J.E.P., J.P.T., S.K.S.)
| | - Grace Gui Yin Lim
- Neurodegeneration Research Laboratory, National Neuroscience Institute, Singapore (G.G.Y.L., K.L.L.)
| | - Kah Leong Lim
- Neurodegeneration Research Laboratory, National Neuroscience Institute, Singapore (G.G.Y.L., K.L.L.)
- Department of Physiology, National University of Singapore (K.L.L.)
| | - Hee Hwa Ho
- Department of Cardiology, Tan Tock Seng Hospital, Singapore (H.H.H.)
| | - James P Tam
- From the School of Biological Sciences, Nanyang Technological University, Singapore (A.S., X.G.-P., J.E.P., J.P.T., S.K.S.)
| | - Siu Kwan Sze
- From the School of Biological Sciences, Nanyang Technological University, Singapore (A.S., X.G.-P., J.E.P., J.P.T., S.K.S.)
| |
Collapse
|
31
|
Nishino K, Watanabe S, Shijie J, Murata Y, Oiwa K, Komine O, Endo F, Tsuiji H, Abe M, Sakimura K, Mishra A, Yamanaka K. Mice deficient in the C-terminal domain of TAR DNA-binding protein 43 develop age-dependent motor dysfunction associated with impaired Notch1-Akt signaling pathway. Acta Neuropathol Commun 2019; 7:118. [PMID: 31345270 PMCID: PMC6657153 DOI: 10.1186/s40478-019-0776-5] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2019] [Accepted: 07/18/2019] [Indexed: 02/06/2023] Open
Abstract
Intracellular mislocalization of TAR DNA-binding protein 43 (TDP-43), a nuclear DNA/RNA-binding protein involved in RNA metabolism, is a pathological hallmark of amyotrophic lateral sclerosis (ALS). Although the aggregation-prone, TDP-43 C-terminal domain is widely considered as a key component of TDP-43 pathology in ALS, recent studies including ours suggest that TDP-43 N-terminal fragments (TDP-∆C) may also contribute to the motor dysfunction in ALS. However, the specific pathological functions of TDP-43 N-terminal fragments in mice have not been elucidated. Here, we established TDP-∆C knock-in mice missing a part of exon 6 of murine Tardbp gene, which encodes the C-terminal region of TDP-43. Homozygous TDP-∆C mice showed embryonic lethality, indicating that the N-terminal domain of TDP-43 alone is not sufficient for normal development. In contrast, heterozygous TDP-∆C mice developed normally but exhibited age-dependent mild motor dysfunction with a loss of C-boutons, large cholinergic synaptic terminals on spinal α-motor neurons. TDP-∆C protein broadly perturbed gene expression in the spinal cords of aged heterozygous TDP-∆C mice, including downregulation of Notch1 mRNA. Moreover, the level of Notch1 mRNA was suppressed both by TDP-43 depletion and TDP-∆C expression in Neuro2a cells. Decreased Notch1 mRNA expression in aged TDP-∆C mice was associated with the age-dependent motor dysfunction and loss of Akt surviving signal. Our findings indicate that the N-terminal region of TDP-43 derived from TDP-∆C induces the age-dependent motor dysfunction associated with impaired Notch1-Akt axis in mice.
Collapse
Affiliation(s)
- Kohei Nishino
- Department of Neuroscience and Pathobiology, Research Institute of Environmental Medicine, Nagoya University, Chikusa-ku, Nagoya, Aichi 464-8601 Japan
- Department of Neuroscience and Pathobiology, Graduate School of Medicine, Nagoya University, Nagoya, Aichi 466-8550 Japan
| | - Seiji Watanabe
- Department of Neuroscience and Pathobiology, Research Institute of Environmental Medicine, Nagoya University, Chikusa-ku, Nagoya, Aichi 464-8601 Japan
- Department of Neuroscience and Pathobiology, Graduate School of Medicine, Nagoya University, Nagoya, Aichi 466-8550 Japan
| | - Jin Shijie
- Department of Neuroscience and Pathobiology, Research Institute of Environmental Medicine, Nagoya University, Chikusa-ku, Nagoya, Aichi 464-8601 Japan
- Department of Neuroscience and Pathobiology, Graduate School of Medicine, Nagoya University, Nagoya, Aichi 466-8550 Japan
| | - Yuri Murata
- Department of Neuroscience and Pathobiology, Research Institute of Environmental Medicine, Nagoya University, Chikusa-ku, Nagoya, Aichi 464-8601 Japan
- Department of Neuroscience and Pathobiology, Graduate School of Medicine, Nagoya University, Nagoya, Aichi 466-8550 Japan
| | - Kotaro Oiwa
- Department of Neuroscience and Pathobiology, Research Institute of Environmental Medicine, Nagoya University, Chikusa-ku, Nagoya, Aichi 464-8601 Japan
- Department of Neuroscience and Pathobiology, Graduate School of Medicine, Nagoya University, Nagoya, Aichi 466-8550 Japan
| | - Okiru Komine
- Department of Neuroscience and Pathobiology, Research Institute of Environmental Medicine, Nagoya University, Chikusa-ku, Nagoya, Aichi 464-8601 Japan
- Department of Neuroscience and Pathobiology, Graduate School of Medicine, Nagoya University, Nagoya, Aichi 466-8550 Japan
| | - Fumito Endo
- Department of Neuroscience and Pathobiology, Research Institute of Environmental Medicine, Nagoya University, Chikusa-ku, Nagoya, Aichi 464-8601 Japan
- Department of Neuroscience and Pathobiology, Graduate School of Medicine, Nagoya University, Nagoya, Aichi 466-8550 Japan
| | - Hitomi Tsuiji
- Department of Biomedical Science, Graduate School of Pharmaceutical Sciences, Nagoya City University, Nagoya, Aichi 467-8603 Japan
| | - Manabu Abe
- Department of Animal Model Development, Brain Research Institute, Niigata University, Niigata, 951-8585 Japan
| | - Kenji Sakimura
- Department of Animal Model Development, Brain Research Institute, Niigata University, Niigata, 951-8585 Japan
| | - Amit Mishra
- Cellular and Molecular Neurobiology Unit, Indian Institute of Technology Jodhpur, Jodhpur, Rajasthan 342011 India
| | - Koji Yamanaka
- Department of Neuroscience and Pathobiology, Research Institute of Environmental Medicine, Nagoya University, Chikusa-ku, Nagoya, Aichi 464-8601 Japan
- Department of Neuroscience and Pathobiology, Graduate School of Medicine, Nagoya University, Nagoya, Aichi 466-8550 Japan
| |
Collapse
|
32
|
Vullhorst D, Buonanno A. NMDA Receptors Regulate Neuregulin 2 Binding to ER-PM Junctions and Ectodomain Release by ADAM10 [corrected]. Mol Neurobiol 2019; 56:8345-8363. [PMID: 31240601 DOI: 10.1007/s12035-019-01659-w] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2019] [Accepted: 05/20/2019] [Indexed: 12/13/2022]
Abstract
Unprocessed pro-neuregulin 2 (pro-NRG2) accumulates on neuronal cell bodies at junctions between the endoplasmic reticulum and plasma membrane (ER-PM junctions). NMDA receptors (NMDARs) trigger NRG2 ectodomain shedding from these sites followed by activation of ErbB4 receptor tyrosine kinases, and ErbB4 signaling cell-autonomously downregulates intrinsic excitability of GABAergic interneurons by reducing voltage-gated sodium channel currents. NMDARs also promote dispersal of Kv2.1 clusters from ER-PM junctions and cause a hyperpolarizing shift in its voltage-dependent channel activation, suggesting that NRG2/ErbB4 and Kv2.1 work together to regulate intrinsic interneuron excitability in an activity-dependent manner. Here we explored the cellular processes underlying NMDAR-dependent NRG2 shedding in cultured rat hippocampal neurons. We report that NMDARs control shedding by two separate but converging mechanisms. First, NMDA treatment disrupts binding of pro-NRG2 to ER-PM junctions by post-translationally modifying conserved Ser/Thr residues in its intracellular domain. Second, using a mutant NRG2 protein that cannot be modified at these residues and that fails to accumulate at ER-PM junctions, we demonstrate that NMDARs also directly promote NRG2 shedding by ADAM-type metalloproteinases. Using pharmacological and shRNA-mediated knockdown, and metalloproteinase overexpression, we unexpectedly find that ADAM10, but not ADAM17/TACE, is the major NRG2 sheddase acting downstream of NMDAR activation. Together, these findings reveal how NMDARs exert tight control over the NRG2/ErbB4 signaling pathway, and suggest that NRG2 and Kv2.1 are co-regulated components of a shared pathway that responds to elevated extracellular glutamate levels.
Collapse
Affiliation(s)
- Detlef Vullhorst
- Section on Molecular Neurobiology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, 35 Lincoln Drive, Room 2C-1000, Bethesda, MD, 20892, USA
| | - Andres Buonanno
- Section on Molecular Neurobiology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, 35 Lincoln Drive, Room 2C-1000, Bethesda, MD, 20892, USA.
| |
Collapse
|
33
|
Takahashi Y, Uchino A, Shioya A, Sano T, Matsumoto C, Numata-Uematsu Y, Nagano S, Araki T, Murayama S, Saito Y. Altered immunoreactivity of ErbB4, a causative gene product for ALS19, in the spinal cord of patients with sporadic ALS. Neuropathology 2019; 39:268-278. [PMID: 31124187 PMCID: PMC6852233 DOI: 10.1111/neup.12558] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2018] [Revised: 03/30/2019] [Accepted: 04/03/2019] [Indexed: 12/13/2022]
Abstract
ErbB4 is the protein implicated in familial amyotrophic lateral sclerosis (ALS), designated as ALS19. ErbB4 is a receptor tyrosine kinase activated by its ligands, neuregulins (NRG), and plays an essential role in the function and viability of motor neurons. Mutations in the ALS19 gene lead to the reduced autophosphorylation capacity of the ErbB4 protein upon stimulation with NRG‐1, suggesting that the disruption of the NRG–ErbB4 pathway causes motor neuron degeneration. We used immunohistochemistry to study ErbB4 in the spinal cord of patients with sporadic ALS (SALS) to test the hypothesis that ErbB4 may be involved in the pathogenesis of SALS. ErbB4 was specifically immunoreactive in the cytoplasm of motor neurons in the anterior horns of the spinal cord. In patients with SALS, some of the motor neurons lost immunoreactivity with ErbB4, with the proportion of motor neurons with a loss of immunoreactivity correlated with the severity of motor neuron loss. The subcellular localization was altered, demonstrating nucleolar or nuclear localization, threads/dots and spheroids. The ectopic glial immunoreactivity was observed, mainly in the oligodendrocytes of the lateral columns and anterior horns. The reduction in the ErbB4 immunoreactivity was significantly correlated with the cytoplasmic mislocalization of transactivation response DNA‐binding protein 43 kDa (TDP‐43) in the motor neurons. No alteration in immunoreactivity was observed in the motor neurons of mice carrying atransgene for mutant form of the superoxide dismutase 1 gene (SOD1). This study provided compelling evidence that ErbB4 is also involved in the pathophysiology of SALS, and that the disruption of the NRG–ErbB4 pathway may underlie the TDP‐43‐dependent motor neuron degeneration in ALS.
Collapse
Affiliation(s)
- Yuji Takahashi
- Department of Neurology, National Center Hospital, National Center of Neurology and Psychiatry, Kodaira, Japan
| | - Akiko Uchino
- Department of Neurology and Neuropathology and Brain Bank for Aging Research, Tokyo Metropolitan Geriatric Hospital and Institute of Gerontology, Tokyo, Japan
| | - Ayako Shioya
- Department of Laboratory Medicine, National Center Hospital, National Center of Neurology and Psychiatry, Kodaira, Japan
| | - Terunori Sano
- Department of Laboratory Medicine, National Center Hospital, National Center of Neurology and Psychiatry, Kodaira, Japan.,Tokyo Medical and Dental University Graduate School of Medical and Dental Sciences, Tokyo, Japan
| | - Chihiro Matsumoto
- Tokyo Medical and Dental University Graduate School of Medical and Dental Sciences, Tokyo, Japan.,Department of Peripheral Nervous System Research, National Institute of Neuroscience, National Center of Neurology and Psychiatry, Kodaira, Japan
| | - Yurika Numata-Uematsu
- Department of Peripheral Nervous System Research, National Institute of Neuroscience, National Center of Neurology and Psychiatry, Kodaira, Japan.,Department of Pediatrics, Tohoku University School of Medicine, Sendai, Japan
| | - Seiichi Nagano
- Department of Peripheral Nervous System Research, National Institute of Neuroscience, National Center of Neurology and Psychiatry, Kodaira, Japan.,Department of Neurology, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Toshiyuki Araki
- Department of Peripheral Nervous System Research, National Institute of Neuroscience, National Center of Neurology and Psychiatry, Kodaira, Japan
| | - Shigeo Murayama
- Department of Neurology and Neuropathology and Brain Bank for Aging Research, Tokyo Metropolitan Geriatric Hospital and Institute of Gerontology, Tokyo, Japan
| | - Yuko Saito
- Department of Laboratory Medicine, National Center Hospital, National Center of Neurology and Psychiatry, Kodaira, Japan
| |
Collapse
|
34
|
Salvany S, Casanovas A, Tarabal O, Piedrafita L, Hernández S, Santafé M, Soto-Bernardini MC, Calderó J, Schwab MH, Esquerda JE. Localization and dynamic changes of neuregulin-1 at C-type synaptic boutons in association with motor neuron injury and repair. FASEB J 2019; 33:7833-7851. [PMID: 30912977 DOI: 10.1096/fj.201802329r] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
C-type synaptic boutons (C-boutons) provide cholinergic afferent input to spinal cord motor neurons (MNs), which display an endoplasmic reticulum (ER)-related subsurface cistern (SSC) adjacent to their postsynaptic membrane. A constellation of postsynaptic proteins is clustered at C-boutons, including M2 muscarinic receptors, potassium channels, and σ-1 receptors. In addition, we previously found that neuregulin (NRG)1 is associated with C-boutons at postsynaptic SSCs, whereas its ErbB receptors are located in the presynaptic compartment. C-bouton-mediated regulation of MN excitability has been implicated in MN disease, but NRG1-mediated functions and the impact of various pathologic conditions on C-bouton integrity have not been studied in detail. Here, we investigated changes in C-boutons after electrical stimulation, pharmacological treatment, and peripheral nerve axotomy. SSC-linked NRG1 clusters were severely disrupted in acutely stressed MNs and after tunicamycin-induced ER stress. In axotomized MNs, C-bouton loss occurred in concomitance with microglial recruitment and was prevented by the ER stress inhibitor salubrinal. Activated microglia displayed a positive chemotaxis to C-boutons. Analysis of transgenic mice overexpressing NRG1 type I and type III isoforms in MNs indicated that NRG1 type III acts as an organizer of SSC-like structures, whereas NRG1 type I promotes synaptogenesis of presynaptic cholinergic terminals. Moreover, MN-derived NRG1 signals may regulate the activity of perineuronal microglial cells. Together, these data provide new insights into the molecular and cellular pathology of C-boutons in MN injury and suggest that distinct NRG1 isoform-mediated signaling functions regulate the complex matching between pre- and postsynaptic C-bouton elements.-Salvany, S., Casanovas, A., Tarabal, O., Piedrafita, L., Hernández, S., Santafé, M., Soto-Bernardini, M. C., Calderó, J., Schwab, M. H., Esquerda, J. E. Localization and dynamic changes of neuregulin-1 at C-type synaptic boutons in association with motor neuron injury and repair.
Collapse
Affiliation(s)
- Sara Salvany
- Unitat de Neurobiologia Cellular, Departament de Medicina Experimental, Facultat de Medicina, Universitat de Lleida-Institut de Recerca Biomèdica de Lleida (IRBLLEIDA), Lleida, Catalonia, Spain
| | - Anna Casanovas
- Unitat de Neurobiologia Cellular, Departament de Medicina Experimental, Facultat de Medicina, Universitat de Lleida-Institut de Recerca Biomèdica de Lleida (IRBLLEIDA), Lleida, Catalonia, Spain
| | - Olga Tarabal
- Unitat de Neurobiologia Cellular, Departament de Medicina Experimental, Facultat de Medicina, Universitat de Lleida-Institut de Recerca Biomèdica de Lleida (IRBLLEIDA), Lleida, Catalonia, Spain
| | - Lídia Piedrafita
- Unitat de Neurobiologia Cellular, Departament de Medicina Experimental, Facultat de Medicina, Universitat de Lleida-Institut de Recerca Biomèdica de Lleida (IRBLLEIDA), Lleida, Catalonia, Spain
| | - Sara Hernández
- Unitat de Neurobiologia Cellular, Departament de Medicina Experimental, Facultat de Medicina, Universitat de Lleida-Institut de Recerca Biomèdica de Lleida (IRBLLEIDA), Lleida, Catalonia, Spain
| | - Manuel Santafé
- Unitat d'Histologia i Neurobiologia (UHN), Facultat de Medicina i Ciències de la Salut, Universitat Rovira i Virgili, Reus, Catalonia, Spain
| | - María Clara Soto-Bernardini
- Instituto Tecnológico de Costa Rica (TEC), Centro de Investigación en Biotecnología (CIB), Escuela de Biología, Cartago, Costa Rica
| | - Jordi Calderó
- Unitat de Neurobiologia Cellular, Departament de Medicina Experimental, Facultat de Medicina, Universitat de Lleida-Institut de Recerca Biomèdica de Lleida (IRBLLEIDA), Lleida, Catalonia, Spain
| | - Markus H Schwab
- Institute of Cellular Neurophysiology, Hannover Medical School, Hannover, Germany.,Center for Systems Neuroscience (ZSN), University of Veterinary Medicine Hannover, Hannover, Germany
| | - Josep E Esquerda
- Unitat de Neurobiologia Cellular, Departament de Medicina Experimental, Facultat de Medicina, Universitat de Lleida-Institut de Recerca Biomèdica de Lleida (IRBLLEIDA), Lleida, Catalonia, Spain
| |
Collapse
|
35
|
Rozani I, Tsapara G, Witts EC, Deaville SJ, Miles GB, Zagoraiou L. Pitx2 cholinergic interneurons are the source of C bouton synapses on brainstem motor neurons. Sci Rep 2019; 9:4936. [PMID: 30894556 PMCID: PMC6426951 DOI: 10.1038/s41598-019-39996-4] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2018] [Accepted: 01/15/2019] [Indexed: 11/29/2022] Open
Abstract
Cholinergic neuromodulation has been described throughout the brain and has been implicated in various functions including attention, food intake and response to stress. Cholinergic modulation is also thought to be important for regulating motor systems, as revealed by studies of large cholinergic synapses on spinal motor neurons, called C boutons, which seem to control motor neuron excitability in a task-dependent manner. C boutons on spinal motor neurons stem from spinal interneurons that express the transcription factor Pitx2. C boutons have also been identified on the motor neurons of specific cranial nuclei. However, the source and roles of cranial C boutons are less clear. Previous studies suggest that they originate from Pitx2+ and Pitx2- neurons, in contrast to spinal cord C boutons that originate solely from Pitx2 neurons. Here, we address this controversy using mouse genetics, and demonstrate that brainstem C boutons are Pitx2+ derived. We also identify new Pitx2 populations and map the cholinergic Pitx2 neurons of the mouse brain. Taken together, our data present important new information about the anatomical organization of cholinergic systems which impact motor systems of the brainstem. These findings will enable further analyses of the specific roles of cholinergic modulation in motor control.
Collapse
Affiliation(s)
- Ismini Rozani
- Center of Basic Research, Biomedical Research Foundation of the Academy of Athens, 4 Soranou Ephessiou Str., 11527, Athens, Greece
- Division of Animal and Human Physiology, Department of Biology, National & Kapodistrian University of Athens, Panepistimiopolis, Ilisia, Greece
| | - Georgia Tsapara
- Center of Basic Research, Biomedical Research Foundation of the Academy of Athens, 4 Soranou Ephessiou Str., 11527, Athens, Greece
- Institute of Molecular Life Sciences, University of Zurich, Winterthurerstrasse 190, CH-8057, Zurich, Switzerland
| | - Emily C Witts
- Sainsbury Wellcome Centre, 25 Howland Street, London, W1T 4JG, UK
- School of Psychology & Neuroscience, University of St Andrews, Fife, KY169JP, UK
| | - S James Deaville
- School of Psychology & Neuroscience, University of St Andrews, Fife, KY169JP, UK
| | - Gareth B Miles
- School of Psychology & Neuroscience, University of St Andrews, Fife, KY169JP, UK
| | - Laskaro Zagoraiou
- Center of Basic Research, Biomedical Research Foundation of the Academy of Athens, 4 Soranou Ephessiou Str., 11527, Athens, Greece.
| |
Collapse
|
36
|
Breit M, Kessler M, Stepniewski M, Vlachos A, Queisser G. Spine-to-Dendrite Calcium Modeling Discloses Relevance for Precise Positioning of Ryanodine Receptor-Containing Spine Endoplasmic Reticulum. Sci Rep 2018; 8:15624. [PMID: 30353066 PMCID: PMC6199256 DOI: 10.1038/s41598-018-33343-9] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2017] [Accepted: 09/18/2018] [Indexed: 12/15/2022] Open
Abstract
The endoplasmic reticulum (ER) forms a complex endomembrane network that reaches into the cellular compartments of a neuron, including dendritic spines. Recent work discloses that the spine ER is a dynamic structure that enters and leaves spines. While evidence exists that ER Ca2+ release is involved in synaptic plasticity, the role of spine ER morphology remains unknown. Combining a new 3D spine generator with 3D Ca2+ modeling, we addressed the relevance of ER positioning on spine-to-dendrite Ca2+ signaling. Our simulations, which account for Ca2+ exchange on the plasma membrane and ER, show that spine ER needs to be present in distinct morphological conformations in order to overcome a barrier between the spine and dendritic shaft. We demonstrate that RyR-carrying spine ER promotes spine-to-dendrite Ca2+ signals in a position-dependent manner. Our simulations indicate that RyR-carrying ER can initiate time-delayed Ca2+ reverberation, depending on the precise position of the spine ER. Upon spine growth, structural reorganization of the ER restores spine-to-dendrite Ca2+ communication, while maintaining aspects of Ca2+ homeostasis in the spine head. Our work emphasizes the relevance of precise positioning of RyR-containing spine ER in regulating the strength and timing of spine Ca2+ signaling, which could play an important role in tuning spine-to-dendrite Ca2+ communication and homeostasis.
Collapse
Affiliation(s)
- Markus Breit
- Goethe Center for Scientific Computing, Computational Neuroscience, Goethe University Frankfurt, Frankfurt, Germany
| | - Marcus Kessler
- Goethe Center for Scientific Computing, Computational Neuroscience, Goethe University Frankfurt, Frankfurt, Germany
| | - Martin Stepniewski
- Goethe Center for Scientific Computing, Computational Neuroscience, Goethe University Frankfurt, Frankfurt, Germany
| | - Andreas Vlachos
- Department of Neuroanatomy, Institute of Anatomy and Cell Biology, Faculty of Medicine, University of Freiburg, Freiburg, 79104, Germany. .,Bernstein Center Freiburg, University of Freiburg, Freiburg, 79104, Germany.
| | - Gillian Queisser
- Department of Mathematics, Temple University, Philadelphia, USA.
| |
Collapse
|
37
|
Wang ZQ, Xiu DH, Liu GF, Jiang JL. Overexpression of Neuregulin-1 (NRG-1) Gene Contributes to Surgical Repair of Brachial Plexus Injury After Contralateral C7 Nerve Root Transfer in Rats. Med Sci Monit 2018; 24:5779-5787. [PMID: 30121695 PMCID: PMC6111774 DOI: 10.12659/msm.908144] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Background Surgeons usually transfer the contralateral C7 to the median nerve on the injured side via a nerve graft to recover sensation and movement in a paralyzed hand. The purpose of our study was to determine whether NRG-1 affects the recovery of nerve function in brachial plexus injury after contralateral C7 nerve root transfer in a rat model. Material/Methods An injury model of left brachial plexus and contralateral C7 nerve root transfer was established. Four weeks after the operation, NRG-1 expression was examined by reverse transcription quantitative polymerase chain reaction and Western blot analysis. The diameter rate differences of the healthy limb and affected limb were estimated. The postoperative mass of the left latissimus dorsi, triceps, extensor carpi radialis brevis, and musculus extensor digitorum were examined. The number of nerve fibers and typical area of the affected side were assessed. Postoperative left motor nerve conduction velocity (MNCV) and motor nerve action potential (MNAP) were tested by use of a biological information recording and collecting system. Results Eukaryotic expression plasmid of pcDNA4/myc/A-NRG-1 was successfully constructed, and NRG-1 was overexpressed. Compared with the model group, the NRG-1 group had a lower rate of differences of the limbs; higher mass of left latissimus dorsi, triceps, extensor carpi radialis brevis, and musculus extensor digitorum; more nerve fibers and larger typical area in the affected side, left MNCV, and MNAP; and wider CSA of the left triceps. Conclusions These results demonstrated that NRG-1 can promote recovery of nerve function in brachial plexus injury after contralateral C7 nerve root transfer in rats.
Collapse
Affiliation(s)
- Zong-Qiang Wang
- Medical Department, China-Japan Union Hospital of Jilin University, Changchun, Jilin, China (mainland)
| | - Dian-Hui Xiu
- Department of Radiology, China-Japan Union Hospital of Jilin University, Changchun, Jilin, China (mainland)
| | - Gui-Feng Liu
- Department of Orthopedics, China-Japan Union Hospital of Jilin University, Changchun, Jilin, China (mainland)
| | - Jin-Lan Jiang
- Department of Orthopedics, China-Japan Union Hospital of Jilin University, Changchun, Jilin, China (mainland).,Scientific Research Center, China-Japan Union Hospital of Jilin University, Changchun, Jilin, China (mainland)
| |
Collapse
|
38
|
Cerveró C, Blasco A, Tarabal O, Casanovas A, Piedrafita L, Navarro X, Esquerda JE, Calderó J. Glial Activation and Central Synapse Loss, but Not Motoneuron Degeneration, Are Prevented by the Sigma-1 Receptor Agonist PRE-084 in the Smn2B/- Mouse Model of Spinal Muscular Atrophy. J Neuropathol Exp Neurol 2018; 77:577-597. [PMID: 29767748 DOI: 10.1093/jnen/nly033] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023] Open
Abstract
Spinal muscular atrophy (SMA) is characterized by the loss of α-motoneurons (MNs) with concomitant muscle denervation. MN excitability and vulnerability to disease are particularly regulated by cholinergic synaptic afferents (C-boutons), in which Sigma-1 receptor (Sig1R) is concentrated. Alterations in Sig1R have been associated with MN degeneration. Here, we investigated whether a chronic treatment with the Sig1R agonist PRE-084 was able to exert beneficial effects on SMA. We used a model of intermediate SMA, the Smn2B/- mouse, in which we performed a detailed characterization of the histopathological changes that occur throughout the disease. We report that Smn2B/- mice exhibited qualitative differences in major alterations found in mouse models of severe SMA: Smn2B/- animals showed more prominent MN degeneration, early motor axon alterations, marked changes in sensory neurons, and later MN deafferentation that correlated with conspicuous reactive gliosis and altered neuroinflammatory M1/M2 microglial balance. PRE-084 attenuated reactive gliosis, mitigated M1/M2 imbalance, and prevented MN deafferentation in Smn2B/- mice. These effects were also observed in a severe SMA model, the SMNΔ7 mouse. However, the prevention of gliosis and MN deafferentation promoted by PRE-084 were not accompanied by any improvements in clinical outcome or other major pathological changes found in SMA mice.
Collapse
Affiliation(s)
- Clàudia Cerveró
- Unitat de Neurobiologia Cel·lular, Departament de Medicina Experimental, Facultat de Medicina, Universitat de Lleida and Institut de Recerca Biomèdica de Lleida (IRBLLEIDA), Lleida, Catalonia, Spain
| | - Alba Blasco
- Unitat de Neurobiologia Cel·lular, Departament de Medicina Experimental, Facultat de Medicina, Universitat de Lleida and Institut de Recerca Biomèdica de Lleida (IRBLLEIDA), Lleida, Catalonia, Spain
| | - Olga Tarabal
- Unitat de Neurobiologia Cel·lular, Departament de Medicina Experimental, Facultat de Medicina, Universitat de Lleida and Institut de Recerca Biomèdica de Lleida (IRBLLEIDA), Lleida, Catalonia, Spain
| | - Anna Casanovas
- Unitat de Neurobiologia Cel·lular, Departament de Medicina Experimental, Facultat de Medicina, Universitat de Lleida and Institut de Recerca Biomèdica de Lleida (IRBLLEIDA), Lleida, Catalonia, Spain
| | - Lídia Piedrafita
- Unitat de Neurobiologia Cel·lular, Departament de Medicina Experimental, Facultat de Medicina, Universitat de Lleida and Institut de Recerca Biomèdica de Lleida (IRBLLEIDA), Lleida, Catalonia, Spain
| | - Xavier Navarro
- Group of Neuroplasticity and Regeneration, Institute of Neurosciences and Department of Cell Biology, Physiology and Immunology, Universitat Autònoma de Barcelona and CIBERNED, Bellaterra, Catalonia, Spain
| | - Josep E Esquerda
- Unitat de Neurobiologia Cel·lular, Departament de Medicina Experimental, Facultat de Medicina, Universitat de Lleida and Institut de Recerca Biomèdica de Lleida (IRBLLEIDA), Lleida, Catalonia, Spain
| | - Jordi Calderó
- Unitat de Neurobiologia Cel·lular, Departament de Medicina Experimental, Facultat de Medicina, Universitat de Lleida and Institut de Recerca Biomèdica de Lleida (IRBLLEIDA), Lleida, Catalonia, Spain
| |
Collapse
|
39
|
Mòdol-Caballero G, Santos D, Navarro X, Herrando-Grabulosa M. Neuregulin 1 Reduces Motoneuron Cell Death and Promotes Neurite Growth in an in Vitro Model of Motoneuron Degeneration. Front Cell Neurosci 2018; 11:431. [PMID: 29375317 PMCID: PMC5767462 DOI: 10.3389/fncel.2017.00431] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2017] [Accepted: 12/21/2017] [Indexed: 12/11/2022] Open
Abstract
Amyotrophic Lateral Sclerosis (ALS) is a devastating neurodegenerative disorder with no effective treatment currently available. Although the mechanisms of motoneuron (MN) death are still unclear, glutamate excitotoxicity and neuroinflammatory reaction are two main features in the neurodegenerative process of ALS. Neuregulin 1 (NRG1) is a trophic factor highly expressed in MNs and neuromuscular junctions. Several recent evidences suggest that NRG1 and their ErbB receptors are involved in ALS. However, further knowledge is still needed to clarify the role of the NRG1-ErbB pathway on MN survival. In this study we used an in vitro model of spinal cord organotypic cultures (SCOCs) subject to chronic excitotoxicity caused by DL-threo-β-hydroxyaspartic acid (THA) to characterize the effect of NRG1 on MN survival. Our results show that addition of recombinant human NRG1 (rhNRG1) to the medium significantly increased MN survival through the activation of ErbB receptors which was ablated with lapatinib (LP), an ErbB inhibitor, and reduced microglial reactivity overcoming the excitotoxicity effects. rhNRG1 activated the pro-survival PI3K/AKT pathway and restored the autophagic flux in the spinal cord culture. Moreover, addition of rhNRG1 to the medium promoted motor and sensory neurite outgrowth. These findings indicate that increasing NRG1 at the spinal cord is an interesting approach for promoting MN protection and regeneration.
Collapse
Affiliation(s)
- Guillem Mòdol-Caballero
- Department of Cell Biology, Physiology and Immunology, Institute of Neurosciences, Universitat Autònoma de Barcelona, Bellaterra, Spain.,Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Bellaterra, Spain
| | - Daniel Santos
- Department of Cell Biology, Physiology and Immunology, Institute of Neurosciences, Universitat Autònoma de Barcelona, Bellaterra, Spain.,Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Bellaterra, Spain
| | - Xavier Navarro
- Department of Cell Biology, Physiology and Immunology, Institute of Neurosciences, Universitat Autònoma de Barcelona, Bellaterra, Spain.,Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Bellaterra, Spain
| | - Mireia Herrando-Grabulosa
- Department of Cell Biology, Physiology and Immunology, Institute of Neurosciences, Universitat Autònoma de Barcelona, Bellaterra, Spain.,Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Bellaterra, Spain
| |
Collapse
|
40
|
Structural Similarities between Neuregulin 1-3 Isoforms Determine Their Subcellular Distribution and Signaling Mode in Central Neurons. J Neurosci 2017; 37:5232-5249. [PMID: 28432142 DOI: 10.1523/jneurosci.2630-16.2017] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2016] [Revised: 03/14/2017] [Accepted: 04/09/2017] [Indexed: 11/21/2022] Open
Abstract
The Neuregulin (NRG) family of ErbB ligands is comprised of numerous variants originating from the use of different genes, alternative promoters, and splice variants. NRGs have generally been thought to be transported to axons and presynaptic terminals where they signal via ErbB3/4 receptors in paracrine or juxtacrine mode. However, we recently demonstrated that unprocessed pro-NRG2 accumulates on cell bodies and proximal dendrites, and that NMDAR activity is required for shedding of its ectodomain by metalloproteinases. Here we systematically investigated the subcellular distribution and processing of major NRG isoforms in rat hippocampal neurons. We show that NRG1 isotypes I and II, which like NRG2 are single-pass transmembrane proteins with an Ig-like domain, share the same subcellular distribution and ectodomain shedding properties. We furthermore show that NRG3, like CRD-NRG1, is a dual-pass transmembrane protein that harbors a second transmembrane domain near its amino terminus. Both NRG3 and CRD-NRG1 cluster on axons through juxtacrine interactions with ErbB4 present on GABAergic interneurons. Interestingly, although single-pass NRGs accumulate as unprocessed proforms, axonal puncta of CRD-NRG1 and NRG3 are comprised of processed protein. Mutations of CRD-NRG1 and NRG3 that render them resistant to BACE cleavage, as well as BACE inhibition, result in the loss of axonal puncta and in the accumulation of unprocessed proforms in neuronal soma. Together, these results define two groups of NRGs with distinct membrane topologies and fundamentally different targeting and processing properties in central neurons. The implications of this functional diversity for the regulation of neuronal processes by the NRG/ErbB pathway are discussed.SIGNIFICANCE STATEMENT Numerous Neuregulins (NRGs) are generated through the use of different genes, promoters, and alternative splicing, but the functional significance of this evolutionary conserved diversity remains poorly understood. Here we show that NRGs can be categorized by their membrane topologies. Single-pass NRGs, such as NRG1 Types I/II and NRG2, accumulate as unprocessed proforms on cell bodies, and their ectodomains are shed by metalloproteinases in response to NMDA receptor activation. By contrast, dual-pass CRD-NRG1 and NRG3 are constitutively processed by BACE and accumulate on axons where they interact with ErbB4 in juxtacrine mode. These findings reveal a previously unknown functional relationship between membrane topology, protein processing, and subcellular distribution, and suggest that single- and dual-pass NRGs regulate neuronal functions in fundamentally different ways.
Collapse
|
41
|
Experimental Design and Data Analysis Issues Contribute to Inconsistent Results of C-Bouton Changes in Amyotrophic Lateral Sclerosis. eNeuro 2017; 4:eN-FTR-0281-16. [PMID: 28101533 PMCID: PMC5241941 DOI: 10.1523/eneuro.0281-16.2016] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2016] [Revised: 11/23/2016] [Accepted: 12/26/2016] [Indexed: 12/12/2022] Open
Abstract
The possible presence of pathological changes in cholinergic synaptic inputs [cholinergic boutons (C-boutons)] is a contentious topic within the ALS field. Conflicting data reported on this issue makes it difficult to assess the roles of these synaptic inputs in ALS. Our objective was to determine whether the reported changes are truly statistically and biologically significant and why replication is problematic. This is an urgent question, as C-boutons are an important regulator of spinal motoneuron excitability, and pathological changes in motoneuron excitability are present throughout disease progression. Using male mice of the SOD1-G93A high-expresser transgenic (G93A) mouse model of ALS, we examined C-boutons on spinal motoneurons. We performed histological analysis at high statistical power, which showed no difference in C-bouton size in G93A versus wild-type motoneurons throughout disease progression. In an attempt to examine the underlying reasons for our failure to replicate reported changes, we performed further histological analyses using several variations on experimental design and data analysis that were reported in the ALS literature. This analysis showed that factors related to experimental design, such as grouping unit, sampling strategy, and blinding status, potentially contribute to the discrepancy in published data on C-bouton size changes. Next, we systematically analyzed the impact of study design variability and potential bias on reported results from experimental and preclinical studies of ALS. Strikingly, we found that practices such as blinding and power analysis are not systematically reported in the ALS field. Protocols to standardize experimental design and minimize bias are thus critical to advancing the ALS field.
Collapse
|
42
|
Casanovas A, Salvany S, Lahoz V, Tarabal O, Piedrafita L, Sabater R, Hernández S, Calderó J, Esquerda JE. Neuregulin 1-ErbB module in C-bouton synapses on somatic motor neurons: molecular compartmentation and response to peripheral nerve injury. Sci Rep 2017; 7:40155. [PMID: 28065942 PMCID: PMC5220293 DOI: 10.1038/srep40155] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2016] [Accepted: 12/02/2016] [Indexed: 12/30/2022] Open
Abstract
The electric activity of lower motor neurons (MNs) appears to play a role in determining cell-vulnerability in MN diseases. MN excitability is modulated by cholinergic inputs through C-type synaptic boutons, which display an endoplasmic reticulum-related subsurface cistern (SSC) adjacent to the postsynaptic membrane. Besides cholinergic molecules, a constellation of proteins involved in different signal-transduction pathways are clustered at C-type synaptic sites (M2 muscarinic receptors, Kv2.1 potassium channels, Ca2+ activated K+ [SK] channels, and sigma-1 receptors [S1R]), but their collective functional significance so far remains unknown. We have previously suggested that neuregulin-1 (NRG1)/ErbBs-based retrograde signalling occurs at this synapse. To better understand signalling through C-boutons, we performed an analysis of the distribution of C-bouton-associated signalling proteins. We show that within SSC, S1R, Kv2.1 and NRG1 are clustered in highly specific, non-overlapping, microdomains, whereas ErbB2 and ErbB4 are present in the adjacent presynaptic compartment. This organization may define highly ordered and spatially restricted sites for different signal-transduction pathways. SSC associated proteins are disrupted in axotomised MNs together with the activation of microglia, which display a positive chemotactism to C-bouton sites. This indicates that C-bouton associated molecules are also involved in neuroinflammatory signalling in diseased MNs, emerging as new potential therapeutic targets.
Collapse
Affiliation(s)
- Anna Casanovas
- Departament de Medicina Experimental, Patologia Neuromuscular Experimental, Facultat de Medicina, Universitat de Lleida/IRBLLEIDA, Av. Rovira Roure 80, 25198 Lleida, Catalonia, Spain
| | - Sara Salvany
- Departament de Medicina Experimental, Patologia Neuromuscular Experimental, Facultat de Medicina, Universitat de Lleida/IRBLLEIDA, Av. Rovira Roure 80, 25198 Lleida, Catalonia, Spain
| | - Víctor Lahoz
- Departament de Medicina Experimental, Patologia Neuromuscular Experimental, Facultat de Medicina, Universitat de Lleida/IRBLLEIDA, Av. Rovira Roure 80, 25198 Lleida, Catalonia, Spain
| | - Olga Tarabal
- Departament de Medicina Experimental, Patologia Neuromuscular Experimental, Facultat de Medicina, Universitat de Lleida/IRBLLEIDA, Av. Rovira Roure 80, 25198 Lleida, Catalonia, Spain
| | - Lídia Piedrafita
- Departament de Medicina Experimental, Patologia Neuromuscular Experimental, Facultat de Medicina, Universitat de Lleida/IRBLLEIDA, Av. Rovira Roure 80, 25198 Lleida, Catalonia, Spain
| | - Raimundo Sabater
- Departament de Medicina Experimental, Patologia Neuromuscular Experimental, Facultat de Medicina, Universitat de Lleida/IRBLLEIDA, Av. Rovira Roure 80, 25198 Lleida, Catalonia, Spain
| | - Sara Hernández
- Departament de Medicina Experimental, Patologia Neuromuscular Experimental, Facultat de Medicina, Universitat de Lleida/IRBLLEIDA, Av. Rovira Roure 80, 25198 Lleida, Catalonia, Spain
| | - Jordi Calderó
- Departament de Medicina Experimental, Patologia Neuromuscular Experimental, Facultat de Medicina, Universitat de Lleida/IRBLLEIDA, Av. Rovira Roure 80, 25198 Lleida, Catalonia, Spain
| | - Josep E. Esquerda
- Departament de Medicina Experimental, Patologia Neuromuscular Experimental, Facultat de Medicina, Universitat de Lleida/IRBLLEIDA, Av. Rovira Roure 80, 25198 Lleida, Catalonia, Spain
| |
Collapse
|
43
|
Mavlyutov TA, Baker EM, Losenegger TM, Kim JR, Torres B, Epstein ML, Ruoho AE. The Sigma-1 Receptor-A Therapeutic Target for the Treatment of ALS? ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 964:255-265. [PMID: 28315276 DOI: 10.1007/978-3-319-50174-1_17] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
The membrane bound 223 amino acid Sigma-1 Receptor (S1R) serves as a molecular chaperone and functional regulator of many signaling proteins. Spinal cord motor neuron activation occurs, in part, via large ventral horn cholinergic synapses called C-boutons/C-terminals. Chronic excitation of motor neurons and alterations in C-terminals has been associated with Amyotrophic Lateral Sclerosis (ALS ). The S1R has an important role in regulating motor neuron function. High levels of the S1R are localized in postsynaptic endoplasmic reticulum (ER) subsurface cisternae within 10-20 nm of the plasma membrane that contain muscarinic type 2 acetylcholine receptors (M2AChR), calcium activated potassium channels (Kv2.1) and slow potassium (SK) channels. An increase in action potentials in the S1R KO mouse motor neurons indicates a critical role for the S1R as a "brake" on motor neuron function possibly via calcium dependent hyperpolarization mechanisms involving the aforementioned potassium channels. The longevity of SOD-1/S1R KO ALS mice is significantly reduced compared to SOD-1/WT ALS controls. The S1R colocalizes in C-terminals with Indole(ethyl)amine-N-methyl transferase (INMT ), the enzyme that produces the S1R agonist , N,N'- dimethyltryptamine (DMT). INMT methylation can additionally neutralize endogenous toxic sulfur and selenium derivatives thus providing functional synergism with DMT to reduce oxidative stress in motor neurons . Small molecule activation of the S1R and INMT thus provides a possible therapeutic strategy to treat ALS .
Collapse
Affiliation(s)
- Timur A Mavlyutov
- Department of Neuroscience, School of Medicine and Public Health, University of Wisconsin, Madison, WI, USA
| | - Erin M Baker
- Department of Neuroscience, School of Medicine and Public Health, University of Wisconsin, Madison, WI, USA
| | - Tasher M Losenegger
- Department of Neuroscience, School of Medicine and Public Health, University of Wisconsin, Madison, WI, USA
| | - Jaimie R Kim
- Department of Neuroscience, School of Medicine and Public Health, University of Wisconsin, Madison, WI, USA
| | - Brian Torres
- Department of Neuroscience, School of Medicine and Public Health, University of Wisconsin, Madison, WI, USA
| | - Miles L Epstein
- Department of Neuroscience, School of Medicine and Public Health, University of Wisconsin, Madison, WI, USA
| | - Arnold E Ruoho
- Department of Neuroscience, School of Medicine and Public Health, University of Wisconsin, Madison, WI, USA.
| |
Collapse
|
44
|
Mancuso R, Navarro X. Sigma-1 Receptor in Motoneuron Disease. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 964:235-254. [PMID: 28315275 DOI: 10.1007/978-3-319-50174-1_16] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Amyotrophic Lateral Sclerosis (ALS ) is a neurodegenerative disease affecting spinal cord and brain motoneurons , leading to paralysis and early death. Multiple etiopathogenic mechanisms appear to contribute in the development of ALS , including glutamate excitotoxicity, oxidative stress , protein misfolding, mitochondrial defects, impaired axonal transport, inflammation and glial cell alterations. The Sigma-1 receptor is highly expressed in motoneurons of the spinal cord, particularly enriched in the endoplasmic reticulum (ER) at postsynaptic cisternae of cholinergic C-terminals. Several evidences point to participation of Sigma-1R alterations in motoneuron degeneration. Thus, mutations of the transmembrane domain of the Sigma-1R have been described in familial ALS cases. Interestingly, Sigma-1R KO mice display muscle weakness and motoneuron loss. On the other hand, Sigma-1R agonists promote neuroprotection and neurite elongation through activation of protein kinase C on motoneurons in vitro and in vivo after ventral root avulsion. Remarkably, treatment of SOD1 mice, the most usual animal model of ALS , with Sigma-1R agonists resulted in significantly enhanced motoneuron function and preservation, and increased animal survival. Sigma-1R activation also reduced microglial reactivity and increased the glial expression of neurotrophic factors. Two main interconnected mechanisms seem to underlie the effects of Sigma-1R manipulation on motoneurons: modulation of neuronal excitability and regulation of calcium homeostasis. In addition, Sigma-1R also contributes to regulating protein degradation, and reducing oxidative stress. Therefore, the multi-functional nature of the Sigma-1R represents an attractive target for treating aspects of ALS and other motoneuron diseases .
Collapse
Affiliation(s)
- Renzo Mancuso
- Center for Biological Sciences, University of Southampton, Southampton General Hospital, SO16 6YD, Southampton, UK
| | - Xavier Navarro
- Institute of Neurosciences and Department of Cell Biology, Physiology and Immunology, Universitat Autònoma de Barcelona, and Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Bellaterra, Spain.
| |
Collapse
|
45
|
Zufiría M, Gil-Bea FJ, Fernández-Torrón R, Poza JJ, Muñoz-Blanco JL, Rojas-García R, Riancho J, López de Munain A. ALS: A bucket of genes, environment, metabolism and unknown ingredients. Prog Neurobiol 2016; 142:104-129. [DOI: 10.1016/j.pneurobio.2016.05.004] [Citation(s) in RCA: 113] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2015] [Revised: 03/22/2016] [Accepted: 05/09/2016] [Indexed: 12/11/2022]
|
46
|
Gallart-Palau X, Serra A, Sze SK. Enrichment of extracellular vesicles from tissues of the central nervous system by PROSPR. Mol Neurodegener 2016; 11:41. [PMID: 27216497 PMCID: PMC4877958 DOI: 10.1186/s13024-016-0108-1] [Citation(s) in RCA: 70] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2015] [Accepted: 05/16/2016] [Indexed: 01/26/2023] Open
Abstract
BACKGROUND Extracellular vesicles (EVs) act as key mediators of intercellular communication and are secreted and taken up by all cell types in the central nervous system (CNS). While detailed study of EV-based signaling is likely to significantly advance our understanding of human neurobiology, the technical challenges of isolating EVs from CNS tissues have limited their characterization using 'omics' technologies. We therefore developed a new Protein Organic Solvent Precipitation (PROSPR) method that can efficiently isolate the EV repertoire from human biological samples. RESULTS In the current report, we present a novel experimental workflow that outlines the process of sample extraction and enrichment of CNS-derived EVs using PROSPR. Subsequent LC-MS/MS-based proteomic profiling of EVs enriched from brain homogenates successfully identified 86 of the top 100 exosomal markers. Proteomic profiling of PROSPR-enriched CNS EVs indicated that > 75 % of the proteins identified matched previously reported exosomal and microvesicle cargoes, while also expanded the known human EV-associated proteome with 685 novel identifications. Similarly, lipidomic characterization of enriched CNS vesicles not only identified previously reported EV-specific lipid families (PS, SM, lysoPC, lysoPE) but also uncovered novel lipid isoforms not previously detected in human EVs. Finally, dedicated flow cytometry of PROSPR-CNS-EVs revealed that ~80 % of total microparticles observed were exosomes ranging in diameter from ≤100 nm to 300 nm. CONCLUSIONS These data demonstrate that the optimized use of PROSPR represents an easy-to-perform and inexpensive method of enriching EVs from human CNS tissues for detailed characterization by 'omics' technologies. We predict that widespread use of the methodology described herein will greatly accelerate the study of EVs biology in neuroscience.
Collapse
Affiliation(s)
- Xavier Gallart-Palau
- School of Biological Sciences, Division of Chemical Biology & BioTechnology, Nanyang Technological University, 60 Nanyang Drive, Singapore, 637551, Singapore
| | - Aida Serra
- School of Biological Sciences, Division of Chemical Biology & BioTechnology, Nanyang Technological University, 60 Nanyang Drive, Singapore, 637551, Singapore
| | - Siu Kwan Sze
- School of Biological Sciences, Division of Chemical Biology & BioTechnology, Nanyang Technological University, 60 Nanyang Drive, Singapore, 637551, Singapore.
| |
Collapse
|
47
|
Alsultan AA, Waller R, Heath PR, Kirby J. The genetics of amyotrophic lateral sclerosis: current insights. Degener Neurol Neuromuscul Dis 2016; 6:49-64. [PMID: 30050368 PMCID: PMC6053097 DOI: 10.2147/dnnd.s84956] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a progressive neurodegenerative disorder that results in loss of the upper and lower motor neurons from motor cortex, brainstem, and spinal cord. While the majority of cases are sporadic, approximately 10% show familial inheritance. ALS is usually inherited in an autosomal dominant manner, although autosomal recessive and X-linked inheritance do occur. To date, 24 of the genes at 26 loci have been identified; these include loci linked to ALS and to frontotemporal dementia-ALS, where family pedigrees contain individuals with frontotemporal dementia with/without ALS. The most commonly established genetic causes of familial ALS (FALS) to date are the presence of a hexanucleotide repeat expansion in the C9ORF72 gene (39.3% FALS) and mutation of SOD1, TARDBP, and FUS, with frequencies of 12%-23.5%, 5%, and 4.1%, respectively. However, with the increasing use of next-generation sequencing of small family pedigrees, this has led to an increasing number of genes being associated with ALS. This review provides a comprehensive review on the genetics of ALS and an update of the pathogenic mechanisms associated with these genes. Commonly implicated pathways have been established, including RNA processing, the protein degradation pathways of autophagy and ubiquitin-proteasome system, as well as protein trafficking and cytoskeletal function. Elucidating the role genetics plays in both FALS and sporadic ALS is essential for understanding the subsequent cellular dysregulation that leads to motor neuron loss, in order to develop future effective therapeutic strategies.
Collapse
Affiliation(s)
- Afnan A Alsultan
- Sheffield Institute for Translational Neuroscience (SITraN), Department of Neuroscience, University of Sheffield, Sheffield, UK,
| | - Rachel Waller
- Sheffield Institute for Translational Neuroscience (SITraN), Department of Neuroscience, University of Sheffield, Sheffield, UK,
| | - Paul R Heath
- Sheffield Institute for Translational Neuroscience (SITraN), Department of Neuroscience, University of Sheffield, Sheffield, UK,
| | - Janine Kirby
- Sheffield Institute for Translational Neuroscience (SITraN), Department of Neuroscience, University of Sheffield, Sheffield, UK,
| |
Collapse
|
48
|
Gallart-Palau X, Ng CH, Ribera J, Sze SK, Lim KL. Drosophila expressing human SOD1 successfully recapitulates mitochondrial phenotypic features of familial amyotrophic lateral sclerosis. Neurosci Lett 2016; 624:47-52. [PMID: 27163198 DOI: 10.1016/j.neulet.2016.05.006] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2015] [Revised: 02/12/2016] [Accepted: 05/05/2016] [Indexed: 11/30/2022]
Abstract
Mitochondrial pathology is a seminal pathogenic hallmark of familial amyotrophic lateral sclerosis (FALS) which is extensively manifested by human patients and mutant SOD1(G93A) mammalian models. Rodents expressing human FALS-associated mutations successfully mimic several human disease features; although they are not as amenable to genetic and therapeutic compound screenings as non-mammalian models. In this study, we report a newly generated and characterized Drosophila model that expresses human SOD1(G93A) in muscle fibers. Presence of SOD1(G93A) in thoracic muscles causes mitochondrial pathology and impairs normal motor behavior in these flies. Use of this new FALS-24B-SOD1(G93A) fly model holds promise for better understanding of the mitochondrial affectation process in FALS and for the discovery of novel therapeutic compounds able to reverse mitochondrial dysfunction in this fatal disease.
Collapse
Affiliation(s)
- Xavier Gallart-Palau
- Experimental Medicine Department, Medical School, University of Lleida, 25198 Lleida, Catalonia, Spain; Institut de Recerca Biomèdica de Lleida (IRBLLEIDA), 25198 Lleida, Catalonia, Spain; School of Biological Sciences, Nanyang Technological University, Singapore 637551, Singapore.
| | - Chee-Hoe Ng
- Department of Research, National Neuroscience Institute, Singapore 308433, Singapore; Duke-NUS Graduate Medical School, Singapore 169857, Singapore
| | - Joan Ribera
- Experimental Medicine Department, Medical School, University of Lleida, 25198 Lleida, Catalonia, Spain; Institut de Recerca Biomèdica de Lleida (IRBLLEIDA), 25198 Lleida, Catalonia, Spain
| | - Siu Kwan Sze
- School of Biological Sciences, Nanyang Technological University, Singapore 637551, Singapore
| | - Kah-Leong Lim
- Department of Physiology, National University of Singapore, Singapore 117543, Singapore; Department of Research, National Neuroscience Institute, Singapore 308433, Singapore; Duke-NUS Graduate Medical School, Singapore 169857, Singapore
| |
Collapse
|
49
|
Lasiene J, Komine O, Fujimori-Tonou N, Powers B, Endo F, Watanabe S, Shijie J, Ravits J, Horner P, Misawa H, Yamanaka K. Neuregulin 1 confers neuroprotection in SOD1-linked amyotrophic lateral sclerosis mice via restoration of C-boutons of spinal motor neurons. Acta Neuropathol Commun 2016; 4:15. [PMID: 26891847 PMCID: PMC4758105 DOI: 10.1186/s40478-016-0286-7] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2016] [Accepted: 02/10/2016] [Indexed: 12/25/2022] Open
Abstract
Introduction Increasing evidence implicates the role of the cell types surrounding motor neurons, such as interneurons and glial cells, in non-cell autonomous neurodegeneration of amyotrophic lateral sclerosis (ALS). C-boutons, the large cholinergic synapses that innervate spinal α-motor neurons to control their excitability, are progressively lost from motor neurons in both human ALS and mutant Cu/Zn superoxide dismutase 1 (SOD1)-ALS mice. Neuregulin-1 (NRG1), a trophic factor implicated in neural development, transmission, and synaptic plasticity, has been reported to localize in the synapse of C-boutons. However, the roles of NRG1 in maintenance of motor neuron health and activity, as well as the functional consequences of its alteration in motor neuron disease, are not fully understood. Results NRG1 was localized to the post-synaptic face of C-boutons and its expression was significantly lost in SOD1-ALS mice and human ALS patients. Losses of NRG1 expression and C-boutons occured almost contemporaneously in SOD1-ALS mice. In addition, expressions of ErbB3 and ErbB4, receptors for NRG1, were reduced in the motor neurons of SOD1-ALS mice. Furthermore, viral-mediated delivery of type III-NRG1 to the spinal cord restored the number of C-boutons and extended the survival time of SOD1-ALS mice. Conclusions These results suggest that maintenance of NRG1-ErbB4/3 axis by supplementation of NRG1 confers neuroprotection in motor neuron disease, partly through the maintenance of C-boutons of spinal motor neurons. Electronic supplementary material The online version of this article (doi:10.1186/s40478-016-0286-7) contains supplementary material, which is available to authorized users.
Collapse
|
50
|
Arii Y, Yamaguchi H, Yamasaki M, Fukuoka SI. Detection of an interaction between prion protein and neuregulin I-β1 by fluorescence resonance energy transfer analysis. Biosci Biotechnol Biochem 2016; 80:761-8. [PMID: 26796243 DOI: 10.1080/09168451.2015.1116934] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
Abstract
Cellular prion protein (PrP) copurifies with neuregulin type I-β1 (NRG I-β1), but no interaction has been detected by a general immunoprecipitation study. We speculate that PrP interacts with NRG I-β1. Here, the interaction of PrP with NRG I-β1 was detected by measuring fluorescence resonance energy transfer (FRET) between enhanced blue (EBFP) and enhanced green (EGFP) fluorescent protein-fusion proteins. Full-length PrP interacted with EGFP in addition to NRG I-β1. From this result, we deduced that PrP interacts with EGFP through its unstructured N-terminal domain. We therefore detected FRET between PrP deleting the N-terminal domain and NRG I-β1. In contrast, the C-terminal domain of PrP interacted with NRG I-β1 and the proteins dissociated completely in the presence of sodium chloride. This interaction occurs at the nanomolar level, which is important for the reaction to be functional in organisms. We concluded that PrP interacted with NRG I-β1 through its C-terminal domain.
Collapse
Affiliation(s)
- Yasuhiro Arii
- a Department of Food Science and Nutrition, School of Human Environmental Sciences , Mukogawa Women's University , Nishinomiya , Japan
| | - Hidenori Yamaguchi
- b Biological Science Course, Graduate School of Science and Engineering , Aoyama Gakuin University , Sagamihara , Japan
| | | | - Shin-Ichi Fukuoka
- b Biological Science Course, Graduate School of Science and Engineering , Aoyama Gakuin University , Sagamihara , Japan
| |
Collapse
|