51
|
Tung YT, Peng KC, Chen YC, Yen YP, Chang M, Thams S, Chen JA. Mir-17∼92 Confers Motor Neuron Subtype Differential Resistance to ALS-Associated Degeneration. Cell Stem Cell 2019; 25:193-209.e7. [PMID: 31155482 DOI: 10.1016/j.stem.2019.04.016] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2018] [Revised: 01/14/2019] [Accepted: 04/22/2019] [Indexed: 12/11/2022]
Abstract
Progressive degeneration of motor neurons (MNs) is the hallmark of amyotrophic lateral sclerosis (ALS). Limb-innervating lateral motor column MNs (LMC-MNs) seem to be particularly vulnerable and are among the first MNs affected in ALS. Here, we report association of this differential susceptibility with reduced expression of the mir-17∼92 cluster in LMC-MNs prior to disease onset. Reduced mir-17∼92 is accompanied by elevated nuclear PTEN in spinal MNs of presymptomatic SOD1G93A mice. Selective dysregulation of the mir-17∼92/nuclear PTEN axis in degenerating SOD1G93A LMC-MNs was confirmed in a double-transgenic embryonic stem cell system and recapitulated in human SOD1+/L144F-induced pluripotent stem cell (iPSC)-derived MNs. We further show that overexpression of mir-17∼92 significantly rescues human SOD1+/L144F MNs, and intrathecal delivery of adeno-associated virus (AAV)9-mir-17∼92 improves motor deficits and survival in SOD1G93A mice. Thus, mir-17∼92 may have value as a prognostic marker of MN degeneration and is a candidate therapeutic target in SOD1-linked ALS. VIDEO ABSTRACT.
Collapse
Affiliation(s)
- Ying-Tsen Tung
- Institute of Molecular Biology, Academia Sinica, Taipei 11529, Taiwan.
| | - Kuan-Chih Peng
- Institute of Molecular Biology, Academia Sinica, Taipei 11529, Taiwan
| | - Yen-Chung Chen
- Institute of Molecular Biology, Academia Sinica, Taipei 11529, Taiwan
| | - Ya-Ping Yen
- Institute of Molecular Biology, Academia Sinica, Taipei 11529, Taiwan
| | - Mien Chang
- Institute of Molecular Biology, Academia Sinica, Taipei 11529, Taiwan
| | - Sebastian Thams
- Department of Pathology and Cell Biology, Center for Motor Neuron Biology and Disease, Columbia Stem Cell Initiative, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Jun-An Chen
- Institute of Molecular Biology, Academia Sinica, Taipei 11529, Taiwan.
| |
Collapse
|
52
|
Valdez G. Effects of disease-afflicted and aging neurons on the musculoskeletal system. Bone 2019; 122:31-37. [PMID: 30695738 PMCID: PMC6444351 DOI: 10.1016/j.bone.2019.01.023] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/18/2019] [Revised: 01/24/2019] [Accepted: 01/25/2019] [Indexed: 01/09/2023]
Abstract
The musculoskeletal system includes skeletal muscles, bones and innervating axons from neurons in the central and peripheral nervous systems. Together, they form the largest structure in the body. They also initiate and coordinate locomotion, provide structural stability, and contribute to metabolism and homeostasis. Because of these functions, much effort has been devoted to ascertaining the impact of acute and chronic stress, such as disease, injury and aging, on the musculoskeletal system. This review will examine the role of the nervous system in the deleterious changes that accrue in skeletal muscles and bones during the progression of neurologic diseases and with advancing age.
Collapse
Affiliation(s)
- Gregorio Valdez
- Fralin Biomedical Research Institute, Virginia Tech Carilion, Roanoke, VA, USA; Department of Biological Sciences, Virginia Tech, Blacksburg, VA, USA.
| |
Collapse
|
53
|
Just-Borràs L, Hurtado E, Cilleros-Mañé V, Biondi O, Charbonnier F, Tomàs M, Garcia N, Lanuza MA, Tomàs J. Overview of Impaired BDNF Signaling, Their Coupled Downstream Serine-Threonine Kinases and SNARE/SM Complex in the Neuromuscular Junction of the Amyotrophic Lateral Sclerosis Model SOD1-G93A Mice. Mol Neurobiol 2019; 56:6856-6872. [PMID: 30929165 DOI: 10.1007/s12035-019-1550-1] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2018] [Accepted: 03/13/2019] [Indexed: 12/12/2022]
Abstract
Amyotrophic lateral sclerosis (ALS) is a chronic neurodegenerative disease characterized by progressive motor weakness. It is accepted that it is caused by motoneuron degeneration leading to a decrease in muscle stimulation. However, ALS is being redefined as a distal axonopathy, in that neuromuscular junction dysfunction precedes and may even influence motoneuron loss. In this synapse, several metabotropic receptor-mediated signaling pathways converge on effector kinases that phosphorylate targets that are crucial for synaptic stability and neurotransmission quality. We have previously shown that, in physiological conditions, nerve-induced muscle contraction regulates the brain-derived neurotrophic factor/tropomyosin-related kinase B (BDNF/TrkB) signaling to retrogradely modulate presynaptic protein kinases PKC and PKA, which are directly involved in the modulation of acetylcholine release. In ALS patients, the alteration of this signaling may significantly contribute to a motor impairment. Here, we investigate whether BDNF/TrkB signaling, the downstream PKC (cPKCβI, cPKCα, and nPKCε isoforms), and PKA (regulatory and catalytic subunits) and some SNARE/SM exocytotic machinery proteins (Munc18-1 and SNAP-25) are altered in the skeletal muscle of pre- and symptomatic SOD1-G93A mice. We found that this pathway is strongly affected in symptomatic ALS mice muscles including an unbalance between (I) BDNF and TrkB isoforms, (II) PKC isoforms and PKA subunits, and (III) Munc18-1 and SNAP-25 phosphorylation ratios. Changes in TrkB.T1 and cPKCβI are precociously observed in presymptomatic mice. Altogether, several of these molecular alterations can be partly associated with the known fast-to-slow motor unit transition during the disease process but others can be related with the initial disease pathogenesis.
Collapse
Affiliation(s)
- Laia Just-Borràs
- Unitat d'Histologia i Neurobiologia (UHNEUROB), Facultat de Medicina i Ciències de la Salut, Universitat Rovira i Virgili, Carrer St Llorenç num 21, 43201, Reus, Spain
| | - Erica Hurtado
- Unitat d'Histologia i Neurobiologia (UHNEUROB), Facultat de Medicina i Ciències de la Salut, Universitat Rovira i Virgili, Carrer St Llorenç num 21, 43201, Reus, Spain
| | - Víctor Cilleros-Mañé
- Unitat d'Histologia i Neurobiologia (UHNEUROB), Facultat de Medicina i Ciències de la Salut, Universitat Rovira i Virgili, Carrer St Llorenç num 21, 43201, Reus, Spain
| | - Olivier Biondi
- INSERM UMRS 1124 and Université Paris Descartes, 45 rue des Saints-Pères, 75270, Paris Cedex 06, France
| | - Frédéric Charbonnier
- INSERM UMRS 1124 and Université Paris Descartes, 45 rue des Saints-Pères, 75270, Paris Cedex 06, France
| | - Marta Tomàs
- Unitat d'Histologia i Neurobiologia (UHNEUROB), Facultat de Medicina i Ciències de la Salut, Universitat Rovira i Virgili, Carrer St Llorenç num 21, 43201, Reus, Spain
| | - Neus Garcia
- Unitat d'Histologia i Neurobiologia (UHNEUROB), Facultat de Medicina i Ciències de la Salut, Universitat Rovira i Virgili, Carrer St Llorenç num 21, 43201, Reus, Spain
| | - Maria A Lanuza
- Unitat d'Histologia i Neurobiologia (UHNEUROB), Facultat de Medicina i Ciències de la Salut, Universitat Rovira i Virgili, Carrer St Llorenç num 21, 43201, Reus, Spain.
| | - Josep Tomàs
- Unitat d'Histologia i Neurobiologia (UHNEUROB), Facultat de Medicina i Ciències de la Salut, Universitat Rovira i Virgili, Carrer St Llorenç num 21, 43201, Reus, Spain.
| |
Collapse
|
54
|
Held A, Major P, Sahin A, Reenan RA, Lipscombe D, Wharton KA. Circuit Dysfunction in SOD1-ALS Model First Detected in Sensory Feedback Prior to Motor Neuron Degeneration Is Alleviated by BMP Signaling. J Neurosci 2019; 39:2347-2364. [PMID: 30659087 PMCID: PMC6433758 DOI: 10.1523/jneurosci.1771-18.2019] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2018] [Revised: 12/24/2018] [Accepted: 01/10/2019] [Indexed: 12/13/2022] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a devastating neurodegenerative disease for which the origin and underlying cellular defects are not fully understood. Although motor neuron degeneration is the signature feature of ALS, it is not clear whether motor neurons or other cells of the motor circuit are the site of disease initiation. To better understand the contribution of multiple cell types in ALS, we made use of a Drosophila Sod1G85R knock-in model, in which all cells harbor the disease allele. End-stage dSod1G85R animals of both sexes exhibit severe motor deficits with clear degeneration of motor neurons. Interestingly, earlier in dSod1G85R larvae, motor function is also compromised, but their motor neurons exhibit only subtle morphological and electrophysiological changes that are unlikely to cause the observed decrease in locomotion. We analyzed the intact motor circuit and identified a defect in sensory feedback that likely accounts for the altered motor activity of dSod1G85R We found cell-autonomous activation of bone morphogenetic protein signaling in proprioceptor sensory neurons which are critical for the relay of the contractile status of muscles back to the central nerve cord, completely rescues early-stage motor defects and partially rescue late-stage motor function to extend lifespan. Identification of a defect in sensory feedback as a potential initiating event in ALS motor dysfunction, coupled with the ability of modified proprioceptors to alleviate such motor deficits, underscores the critical role that nonmotor neurons play in disease progression and highlights their potential as a site to identify early-stage ALS biomarkers and for therapeutic intervention.SIGNIFICANCE STATEMENT At diagnosis, many cellular processes are already disrupted in the amyotrophic lateral sclerosis (ALS) patient. Identifying the initiating cellular events is critical for achieving an earlier diagnosis to slow or prevent disease progression. Our findings indicate that neurons relaying sensory information underlie early stage motor deficits in a Drosophila knock-in model of ALS that best replicates gene dosage in familial ALS (fALS). Importantly, studies on intact motor circuits revealed defects in sensory feedback before evidence of motor neuron degeneration. These findings strengthen our understanding of how neural circuit dysfunctions lead to neurodegeneration and, coupled with our demonstration that the activation of bone morphogenetic protein signaling in proprioceptors alleviates both early and late motor dysfunction, underscores the importance of considering nonmotor neurons as therapeutic targets.
Collapse
Affiliation(s)
- Aaron Held
- Department of Molecular Biology, Cell Biology and Biochemistry
- The Robert J. and Nancy D. Carney Institute for Brain Science, Brown University, Providence, Rhode Island 02912
| | - Paxton Major
- Department of Molecular Biology, Cell Biology and Biochemistry
| | - Asli Sahin
- Department of Molecular Biology, Cell Biology and Biochemistry
| | - Robert A Reenan
- Department of Molecular Biology, Cell Biology and Biochemistry
| | - Diane Lipscombe
- Department of Neuroscience, and
- The Robert J. and Nancy D. Carney Institute for Brain Science, Brown University, Providence, Rhode Island 02912
| | - Kristi A Wharton
- Department of Molecular Biology, Cell Biology and Biochemistry,
- The Robert J. and Nancy D. Carney Institute for Brain Science, Brown University, Providence, Rhode Island 02912
| |
Collapse
|
55
|
Xu Y, Halievski K, Katsuno M, Adachi H, Sobue G, Breedlove SM, Jordan CL. Pre-clinical symptoms of SBMA may not be androgen-dependent: implications from two SBMA mouse models. Hum Mol Genet 2019; 27:2425-2442. [PMID: 29897452 DOI: 10.1093/hmg/ddy142] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2018] [Accepted: 04/16/2018] [Indexed: 12/31/2022] Open
Abstract
A distinguishing aspect of spinal and bulbar muscular atrophy (SBMA) is its androgen-dependence, possibly explaining why only males are clinically affected. This disease, which impairs neuromuscular function, is linked to a polyglutamine expansion mutation in the androgen receptor (AR). In mouse models of SBMA, motor dysfunction is associated with pronounced defects in neuromuscular transmission, including defects in evoked transmitter release (quantal content, QC) and fiber membrane excitability (based on the resting membrane potential, RMP). However, whether such defects are androgen-dependent is unknown. Thus, we recorded synaptic potentials intracellularly from adult muscle fibers of transgenic (Tg) AR97Q male mice castrated pre-symptomatically. Although castration largely protects both QC and the RMP of fibers, correlating with the protective effect of castration on motor function, significant deficits in QC and RMP remained. Surprisingly, comparable defects in QC and RMP were also observed in pre-symptomatic AR97Q males, indicating that such defects emerge early and are pre-clinical. Exposing asymptomatic Tg females to androgens also induces both motor dysfunction and comparable defects in QC and RMP. Notably, asymptomatic Tg females also showed significant deficits in QC and RMP, albeit less severe, supporting their pre-clinical nature, but also raising questions about the androgen-dependence of pre-clinical symptoms. In summary, current evidence indicates that disease progression depends on androgens, but early pathogenic events may be triggered by the mutant AR allele independent of androgens. Such early, androgen-independent disease mechanisms may also be relevant to females carrying the SBMA allele.
Collapse
Affiliation(s)
- Youfen Xu
- Neuroscience Program, Michigan State University, East Lansing, MI, USA
| | | | - Masahisa Katsuno
- Department of Neurology, Nagoya University Graduate School of Medicine, Showa-ku, Nagoya, Japan
| | - Hiroaki Adachi
- Department of Neurology, University of Occupational and Environment Health School of Medicine, Yahatanishi-ku, Kitakyushu Fukuoka, Japan
| | - Gen Sobue
- Department of Neurology, Nagoya University Graduate School of Medicine, Showa-ku, Nagoya, Japan
| | - S Marc Breedlove
- Neuroscience Program, Michigan State University, East Lansing, MI, USA
| | - Cynthia L Jordan
- Neuroscience Program, Michigan State University, East Lansing, MI, USA
| |
Collapse
|
56
|
Dysfunction of Neuromuscular Synaptic Transmission and Synaptic Vesicle Recycling in Motor Nerve Terminals of mSOD1 Transgenic Mice with Model of Amyotrophic Lateral Sclerosis. BIONANOSCIENCE 2019. [DOI: 10.1007/s12668-018-0590-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
57
|
So E, Mitchell JC, Memmi C, Chennell G, Vizcay-Barrena G, Allison L, Shaw CE, Vance C. Mitochondrial abnormalities and disruption of the neuromuscular junction precede the clinical phenotype and motor neuron loss in hFUSWT transgenic mice. Hum Mol Genet 2019; 27:463-474. [PMID: 29194538 PMCID: PMC5886082 DOI: 10.1093/hmg/ddx415] [Citation(s) in RCA: 69] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2017] [Accepted: 11/26/2017] [Indexed: 11/16/2022] Open
Abstract
FUS (fused in sarcoma) mislocalization and cytoplasmic aggregation are hallmark pathologies in FUS-related amyotrophic lateral sclerosis and frontotemporal dementia. Many of the mechanistic hypotheses have focused on a loss of nuclear function in the FUS-opathies, implicating dysregulated RNA transcription and splicing in driving neurodegeneration. Recent studies describe an additional somato-dendritic localization for FUS in the cerebral cortex implying a regulatory role in mRNA transport and local translation at the synapse. Here, we report that FUS is also abundant at the pre-synaptic terminal of the neuromuscular junction (NMJ), suggesting an important function for this protein at peripheral synapses. We have previously reported dose and age-dependent motor neuron degeneration in transgenic mice overexpressing human wild-type FUS, resulting in a motor phenotype detected by ∼28 days and death by ∼100 days. Now, we report the earliest structural events using electron microscopy and quantitative immunohistochemistry. Mitochondrial abnormalities in the pre-synaptic motor nerve terminals are detected at postnatal day 6, which are more pronounced at P15 and accompanied by a loss of synaptic vesicles and synaptophysin protein coupled with NMJs of a smaller size at a time when there is no detectable motor neuron loss. These changes occur in the presence of abundant FUS and support a peripheral toxic gain of function. This appearance is typical of a ‘dying-back’ axonopathy, with the earliest manifestation being mitochondrial disruption. These findings support our hypothesis that FUS has an important function at the NMJ, and challenge the ‘loss of nuclear function’ hypothesis for disease pathogenesis in the FUS-opathies.
Collapse
Affiliation(s)
- Eva So
- Department of Basic and Clinical Neuroscience
| | | | | | - George Chennell
- Department of Basic and Clinical Neuroscience.,Wohl Cellular Imaging Centre, Institute of Psychiatry, Psychology and Neuroscience, King's College London, Denmark Hill, London SE5 8AF, UK
| | - Gema Vizcay-Barrena
- Centre for Ultrastructural Imaging, King's College London, New Hunts House, Guy's Campus, London SE1 1UL, UK
| | - Leanne Allison
- Centre for Ultrastructural Imaging, King's College London, New Hunts House, Guy's Campus, London SE1 1UL, UK
| | | | | |
Collapse
|
58
|
Fusco AF, McCall AL, Dhindsa JS, Pucci LA, Strickland LM, Kahn AF, ElMallah MK. The Respiratory Phenotype of Rodent Models of Amyotrophic Lateral Sclerosis and Spinocerebellar Ataxia. JOURNAL OF NEUROINFLAMMATION AND NEURODEGENERATIVE DISEASES 2019; 3:100011. [PMID: 31893284 PMCID: PMC6938301] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Amyotrophic lateral sclerosis (ALS) and spinocerebellar ataxia (SCA) are neurodegenerative disorders that result in progressive motor dysfunction and ultimately lead to respiratory failure. Rodent models of neurodegenerative disorders provide a means to study the respiratory motor unit pathology that results in respiratory failure. In addition, they are important for pre-clinical studies of novel therapies that improve breathing, quality of life, and survival. The goal of this review is to compare the respiratory phenotype of two neurodegenerative disorders that have different pathological origins, but similar physiological outcomes. Manuscripts reviewed were identified using specific search terms and exclusion criteria. We excluded manuscripts that investigated novel therapeutics and only included those manuscripts that describe the respiratory pathology. The ALS manuscripts describe pathology in respiratory physiology, the phrenic and hypoglossal motor units, respiratory neural control centers, and accessory respiratory muscles. The SCA rodent model manuscripts characterized pathology in overall respiratory function, phrenic motor units and hypoglossal motor neurons. Overall, a combination of pathology in the respiratory motor units and control centers contribute to devastating respiratory dysfunction.
Collapse
Affiliation(s)
- Anna F. Fusco
- Department of Pediatrics, School of Medicine, Duke University, Durham, NC
| | - Angela L. McCall
- Department of Pediatrics, School of Medicine, Duke University, Durham, NC
| | - Justin S. Dhindsa
- Department of Pediatrics, School of Medicine, Duke University, Durham, NC
| | - Logan A. Pucci
- Department of Pediatrics, School of Medicine, Duke University, Durham, NC
| | | | - Amanda F. Kahn
- Department of Pediatrics, School of Medicine, Duke University, Durham, NC
| | - Mai K. ElMallah
- Department of Pediatrics, School of Medicine, Duke University, Durham, NC,Corresponding author: Mai K. ElMallah, Division of Allergy, Immunology and Pulmonary Medicine, Department of Pediatrics, Duke University Medical Center Box 2644, Durham, NC 27710, USA, Tel: 919-684-3577;
| |
Collapse
|
59
|
Single-copy expression of an amyotrophic lateral sclerosis-linked TDP-43 mutation (M337V) in BAC transgenic mice leads to altered stress granule dynamics and progressive motor dysfunction. Neurobiol Dis 2019; 121:148-162. [DOI: 10.1016/j.nbd.2018.09.024] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2018] [Revised: 09/10/2018] [Accepted: 09/30/2018] [Indexed: 12/12/2022] Open
|
60
|
Abstract
The synapse is an incredibly specialized structure that allows for the coordinated communication of information from one neuron to another. When assembled into circuits, steady streams of excitatory and inhibitory synaptic activity shape neural outputs. At the organismal level, ensembles of neural networks underlie behavior, emotion and memory. Disorder or dysfunctions of synapses, a synaptopathy, may underlie a host of developmental and degenerative neurological conditions. There is a possibility that amyotrophic lateral sclerosis may be a result of a synaptopathy within the neuromotor system. To this end, particular attention has been trained on the excitatory glutamatergic synapses and their morphological proxy, the dendritic spine. The extensive detailing of these dysfunctions in vulnerable neuronal populations, including corticospinal neurons and motor neurons, has recently been the subject of original research in rodents and humans. If amyotrophic lateral sclerosis is indeed a synaptopathy, it is entirely consistent with other proposed pathogenic mechanisms – including glutamate excitotoxicity, accumulation of misfolded proteins and mitochondrial dysfunction at distal axon terminals (cortico-motor neuron and neuromuscular). Further, although the exact mechanism of disease spread from region to region is unknown, the synaptopathy hypothesis is consistent with emerging die-forward evidence and the prion-like propagation of misfolded protein aggregates to distant neuronal populations. Here in this mini-review, we focus on the timeline of synaptic observations in both cortical and spinal neurons from different rodent models, and provide a conceptual framework for assessing the synaptopathy hypothesis in amyotrophic lateral sclerosis.
Collapse
Affiliation(s)
- Matthew J Fogarty
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN, USA; School of Biomedical Sciences, The University of Queensland, St. Lucia, Australia
| |
Collapse
|
61
|
Morgan S, Duguez S, Duddy W. Personalized Medicine and Molecular Interaction Networks in Amyotrophic Lateral Sclerosis (ALS): Current Knowledge. J Pers Med 2018; 8:E44. [PMID: 30551677 PMCID: PMC6313785 DOI: 10.3390/jpm8040044] [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: 10/31/2018] [Revised: 11/27/2018] [Accepted: 12/05/2018] [Indexed: 12/12/2022] Open
Abstract
Multiple genes and mechanisms of pathophysiology have been implicated in amyotrophic lateral sclerosis (ALS), suggesting it is a complex systemic disease. With this in mind, applying personalized medicine (PM) approaches to tailor treatment pipelines for ALS patients may be necessary. The modelling and analysis of molecular interaction networks could represent valuable resources in defining ALS-associated pathways and discovering novel therapeutic targets. Here we review existing omics datasets and analytical approaches, in order to consider how molecular interaction networks could improve our understanding of the molecular pathophysiology of this fatal neuromuscular disorder.
Collapse
Affiliation(s)
- Stephen Morgan
- Northern Ireland Centre for Stratified Medicine, Altnagelvin Hospital Campus, Ulster University, Londonderry, BT47 6SB, Northern Ireland, UK.
| | - Stephanie Duguez
- Northern Ireland Centre for Stratified Medicine, Altnagelvin Hospital Campus, Ulster University, Londonderry, BT47 6SB, Northern Ireland, UK.
| | - William Duddy
- Northern Ireland Centre for Stratified Medicine, Altnagelvin Hospital Campus, Ulster University, Londonderry, BT47 6SB, Northern Ireland, UK.
| |
Collapse
|
62
|
Motor neuron degeneration, severe myopathy and TDP-43 increase in a transgenic pig model of SOD1-linked familiar ALS. Neurobiol Dis 2018; 124:263-275. [PMID: 30471417 DOI: 10.1016/j.nbd.2018.11.021] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2018] [Revised: 10/26/2018] [Accepted: 11/19/2018] [Indexed: 12/11/2022] Open
Abstract
Amyotrophic Lateral Sclerosis (ALS) is a neural disorder gradually leading to paralysis of the whole body. Alterations in superoxide dismutase SOD1 gene have been linked with several variants of familial ALS. Here, we investigated a transgenic (Tg) cloned swine model expressing the human pathological hSOD1G93A allele. As in patients, these Tg pigs transmitted the disease to the progeny with an autosomal dominant trait and showed ALS onset from about 27 months of age. Post mortem analysis revealed motor neuron (MN) degeneration, gliosis and hSOD1 protein aggregates in brainstem and spinal cord. Severe skeletal muscle pathology including necrosis and inflammation was observed at the end stage, as well. Remarkably, as in human patients, these Tg pigs showed a quite long presymptomatic phase in which gradually increasing amounts of TDP-43 were detected in peripheral blood mononuclear cells. Thus, this transgenic swine model opens the unique opportunity to investigate ALS biomarkers even before disease onset other than testing novel drugs and possible medical devices.
Collapse
|
63
|
Walker CL, Meadows RM, Merfeld-Clauss S, Du Y, March KL, Jones KJ. Adipose-derived stem cell conditioned medium impacts asymptomatic peripheral neuromuscular denervation in the mutant superoxide dismutase (G93A) transgenic mouse model of amyotrophic lateral sclerosis. Restor Neurol Neurosci 2018; 36:621-627. [PMID: 30010155 DOI: 10.3233/rnn-180820] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
BACKGROUND Amyotrophic lateral sclerosis (ALS) is devastating, leading to paralysis and death. Disease onset begins pre-symptomatically through spinal motor neuron (MN) axon die-back from musculature at ∼47 days of age in the mutant superoxide dismutase 1 (mSOD1G93A) transgenic ALS mouse model. This period may be optimal to assess potential therapies. We previously demonstrated that post-symptomatic adipose-derived stem cell conditioned medium (ASC-CM) treatment is neuroprotective in mSOD1G93A mice. We hypothesized that early disease onset treatment could ameliorate neuromuscular junction (NMJ) disruption. OBJECTIVE To determine whether pre-symptom administration of ASC-CM prevents early NMJ disconnection. METHODS We confirmed the NMJ denervation time course in mSOD1G93A mice using co-labeling of neurofilament and post-synaptic acetylcholine receptors (AchR) by α-bungarotoxin. We determined whether ASC-CM ameliorates early NMJ loss in mSOD1G93A mice by systemically administering 200μl ASC-CM or vehicle medium daily from post-natal days 35 to 47 and quantifying intact NMJs through co-labeling of neurofilament and synaptophysin with α-bungarotoxin in gastrocnemius muscle. RESULTS Intact NMJs were significantly decreased in 47 day old mSOD1G93A mice (p < 0.05), and daily systemic ASC-CM prevented disease-induced NMJ denervation compared to vehicle treated mice (p < 0.05). CONCLUSIONS Our results lay the foundation for testing the long-term neurological benefits of systemic ASC-CM therapy in the mSOD1G93A mouse model of ALS.
Collapse
Affiliation(s)
- Chandler L Walker
- Department of Biomedical and Applied Sciences, Indiana University School of Dentistry, Indianapolis, IN, USA.,Department of Anatomy and Cell Biology, Indiana University School of Medicine, Indianapolis, IN, USA.,Roudebush VA Medical Center, Indianapolis, IN, USA
| | - Rena M Meadows
- Department of Anatomy and Cell Biology, Indiana University School of Medicine, Indianapolis, IN, USA.,Roudebush VA Medical Center, Indianapolis, IN, USA
| | - Stephanie Merfeld-Clauss
- Roudebush VA Medical Center, Indianapolis, IN, USA.,Division of Cardiovascular Medicine, Center for Regenerative Medicine, University of Florida, Gainesville, FL, USA
| | - Yansheng Du
- Department of Neurology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Keith L March
- Department of Medicine, Indiana University School of Medicine, Indianapolis, IN, USA.,Roudebush VA Medical Center, Indianapolis, IN, USA.,Division of Cardiovascular Medicine, Center for Regenerative Medicine, University of Florida, Gainesville, FL, USA
| | - Kathryn J Jones
- Department of Anatomy and Cell Biology, Indiana University School of Medicine, Indianapolis, IN, USA.,Roudebush VA Medical Center, Indianapolis, IN, USA
| |
Collapse
|
64
|
Srinivasan E, Rajasekaran R. Quantum chemical and molecular mechanics studies on the assessment of interactions between resveratrol and mutant SOD1 (G93A) protein. J Comput Aided Mol Des 2018; 32:1347-1361. [PMID: 30368622 DOI: 10.1007/s10822-018-0175-1] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2018] [Accepted: 10/24/2018] [Indexed: 12/29/2022]
Abstract
Amyotrophic lateral sclerosis (ALS) is a neurodegenerative disease that has been associated with mutations in metalloenzyme superoxide dismutase (SOD1) causing protein structural destabilization and aggregation. However, the mechanistic action and the cure for the disease still remain obscure. Herein, we initially studied the conformational preferences of SOD1 protein structures upon substitution of Ala at Gly93 in comparison with that of wild type. Our results corroborated with the previous experimental studies on the aggregation and the destabilizing activity of mutant SOD1 protein G93A. On the therapeutic point of view, we computationally analyzed the influence of resveratrol, a natural polyphenol widely found in red wine on mutant SOD1 relative to wild type, using molecular docking studies. Further, FMO calculations were performed, using GAMESS to study the pair residual interaction on the wild type and mutant complex systems. Consequently, the resveratrol showed greater interaction with mutant than the wild type. Subsequently, we evaluated the conformational preferences of wild type and mutant complex systems, where the protein conformational structures of mutant that were earlier found to lose their conformational stability was regained, upon binding with resveratrol. Similar trend of results were found on the 2-D free energy landscapes of both the wild type and mutant systems. Hence, the combined biophysical and quantum chemical studies in our study supported the results of previous experimental studies, thereby stipulating an action of resveratrol on mutant SOD1 and paving a way for the design of highly potent effective inhibitors against fALS affecting the mankind.
Collapse
Affiliation(s)
- E Srinivasan
- Bioinformatics Lab, Department of Biotechnology, School of Bio Sciences and Technology, VIT (Deemed to be University), Vellore, Tamil Nadu, 632014, India
| | - R Rajasekaran
- Bioinformatics Lab, Department of Biotechnology, School of Bio Sciences and Technology, VIT (Deemed to be University), Vellore, Tamil Nadu, 632014, India.
| |
Collapse
|
65
|
Martineau É, Di Polo A, Vande Velde C, Robitaille R. Dynamic neuromuscular remodeling precedes motor-unit loss in a mouse model of ALS. eLife 2018; 7:41973. [PMID: 30320556 PMCID: PMC6234026 DOI: 10.7554/elife.41973] [Citation(s) in RCA: 68] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2018] [Accepted: 10/14/2018] [Indexed: 12/13/2022] Open
Abstract
Despite being an early event in ALS, it remains unclear whether the denervation of neuromuscular junctions (NMJ) is simply the first manifestation of a globally degenerating motor neuron. Using in vivo imaging of single axons and their NMJs over a three-month period, we identify that single motor-units are dismantled asynchronously in SOD1G37R mice. We reveal that weeks prior to complete axonal degeneration, the dismantling of axonal branches is accompanied by contemporaneous new axonal sprouting resulting in synapse formation onto nearby NMJs. Denervation events tend to propagate from the first lost NMJ, consistent with a contribution of neuromuscular factors extrinsic to motor neurons, with distal branches being more susceptible. These results show that NMJ denervation in ALS is a complex and dynamic process of continuous denervation and new innervation rather than a manifestation of sudden global motor neuron degeneration. Amyotrophic lateral sclerosis (ALS), also known as Lou Gehrig’s disease, is a fatal neurodegenerative disorder. It occurs when the neurons that control muscles – the motoneurons – disconnect from their target muscles and die. This causes the muscles to weaken and waste away. More and more muscles become affected over time until eventually the muscles that control breathing also become paralyzed. Most patients die within two to five years of diagnosis. Motoneurons consist of a cell body plus a cable-like structure called the axon. The cell body of each motoneuron sits within the spinal cord, and the axon extends out of the spinal cord to the motoneuron’s target muscle. Within the muscle the axon divides into branches, each of which connects with multiple muscle fibers. The breakdown of these connections, known as neuromuscular junctions, is one of the first signs of ALS. Does a motoneuron lose all of its connections with muscle fibers at once, or do the connections break down a few at a time? This distinction is important as it will help to identify the events that lead to muscle paralysis in ALS. To find out, Martineau et al. studied mice that had two genetic mutations: one that causes ALS and another that produces fluorescent molecules in some motoneurons. This allowed the branches of the motoneurons to be tracked over time with a fluorescence microscope. Martineau et al. found that individual neurons lose their connections to muscle fibers gradually. Moreover, motoneurons grow new branches and form new connections even while losing their old ones. This dual process of pruning and budding lasts for several weeks, until eventually the motoneuron dies. Developing drugs to stabilize neuromuscular junctions during the period when motoneurons gradually disconnect from muscles could be a promising avenue to explore to improve the quality of life of ALS patients. One advantage of this treatment strategy is that neuromuscular junctions in muscles are easier to access than motoneurons inside the spinal cord. To identify potential drugs, future studies will need to focus on the proteins and signals that cause the neuromuscular junctions to break down.
Collapse
Affiliation(s)
- Éric Martineau
- Département de neurosciences, Université de Montréal, Québec, Canada.,Groupe de recherche sur le système nerveux central, Université de Montréal, Québec, Canada
| | - Adriana Di Polo
- Département de neurosciences, Université de Montréal, Québec, Canada.,Centre de recherche du Centre Hospitalier de l'Université de Montréal, Québec, Canada
| | - Christine Vande Velde
- Département de neurosciences, Université de Montréal, Québec, Canada.,Centre de recherche du Centre Hospitalier de l'Université de Montréal, Québec, Canada
| | - Richard Robitaille
- Département de neurosciences, Université de Montréal, Québec, Canada.,Groupe de recherche sur le système nerveux central, Université de Montréal, Québec, Canada
| |
Collapse
|
66
|
Machamer JB, Woolums BM, Fuller GG, Lloyd TE. FUS causes synaptic hyperexcitability in Drosophila dendritic arborization neurons. Brain Res 2018; 1693:55-66. [PMID: 29625118 PMCID: PMC6347466 DOI: 10.1016/j.brainres.2018.03.037] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2018] [Revised: 03/25/2018] [Accepted: 03/31/2018] [Indexed: 12/13/2022]
Abstract
Mutations in the nuclear localization signal of the RNA binding protein FUS cause both Frontotemporal Dementia (FTD) and Amyotrophic Lateral Sclerosis (ALS). These mutations result in a loss of FUS from the nucleus and the formation of FUS-containing cytoplasmic aggregates in patients. To better understand the role of cytoplasmic FUS mislocalization in the pathogenesis of ALS, we identified a population of cholinergic neurons in Drosophila that recapitulate these pathologic hallmarks. Expression of mutant FUS or the Drosophila homolog, Cabeza (Caz), in class IV dendritic arborization neurons results in cytoplasmic mislocalization and axonal transport to presynaptic terminals. Interestingly, overexpression of FUS or Caz causes the progressive loss of neuronal projections, reduction of synaptic mitochondria, and the appearance of large calcium transients within the synapse. Additionally, we find that overexpression of mutant but not wild type FUS results in a reduction in presynaptic Synaptotagmin, an integral component of the neurotransmitter release machinery, and mutant Caz specifically disrupts axonal transport and induces hyperexcitability. These results suggest that FUS/Caz overexpression disrupts neuronal function through multiple mechanisms, and that ALS-causing mutations impair the transport of synaptic vesicle proteins and induce hyperexcitability.
Collapse
Affiliation(s)
- James B Machamer
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, United States
| | - Brian M Woolums
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, United States
| | - Gregory G Fuller
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, United States
| | - Thomas E Lloyd
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, United States; The Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205, United States.
| |
Collapse
|
67
|
Gomes C, Cunha C, Nascimento F, Ribeiro JA, Vaz AR, Brites D. Cortical Neurotoxic Astrocytes with Early ALS Pathology and miR-146a Deficit Replicate Gliosis Markers of Symptomatic SOD1G93A Mouse Model. Mol Neurobiol 2018; 56:2137-2158. [DOI: 10.1007/s12035-018-1220-8] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2018] [Accepted: 06/29/2018] [Indexed: 12/13/2022]
|
68
|
Kjældgaard AL, Pilely K, Olsen KS, Pedersen SW, Lauritsen AØ, Møller K, Garred P. Amyotrophic lateral sclerosis: The complement and inflammatory hypothesis. Mol Immunol 2018; 102:14-25. [PMID: 29933890 DOI: 10.1016/j.molimm.2018.06.007] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2018] [Revised: 05/15/2018] [Accepted: 06/06/2018] [Indexed: 12/28/2022]
Abstract
Amyotrophic lateral sclerosis (ALS) is a devastating, neurodegenerative motor neuron disease. The aetiology of ALS remains an enigma which hinders the design of an effective treatment to prevent, postpone, or reverse the pathophysiological changes occurring during the aggressive progression of this disease. During the last decade, basic research within the innate immune system, and in particular the complement system, has revealed new, important roles of the innate immune system during development, homeostasis, and ageing within as well as outside the central nervous system. Several lines of evidence indicate that aberrant activation of the complement system locally in the central nervous system as well as systemically may be involved in the pathophysiology of ALS. This exciting new knowledge could point towards the innate immune system as a potential target of medical intervention in ALS. Recently, the historic perception of ALS as a central neurodegenerative disease has been challenged due to the significant amount of evidence of a dying-back mechanism causing the selective destruction of the motor neurons, indicating that disease onset occurs outside the borders of the blood-brain-barrier. This review addresses the function of the innate immune system during ALS. We emphasize the role of the complement system and specifically suggest the involvement of ficolin-3 from the lectin pathway in the pathophysiology of ALS.
Collapse
Affiliation(s)
- Anne-Lene Kjældgaard
- Laboratory of Molecular Medicine, Department of Clinical Immunology, Diagnostic Centre, Section 7631; Department of Neuroanaesthesiology.
| | - Katrine Pilely
- Laboratory of Molecular Medicine, Department of Clinical Immunology, Diagnostic Centre, Section 7631
| | | | - Stephen Wørlich Pedersen
- Department of Neurology, Neuroscience Centre, Rigshospitalet, Institute of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Denmark
| | | | | | - Peter Garred
- Laboratory of Molecular Medicine, Department of Clinical Immunology, Diagnostic Centre, Section 7631
| |
Collapse
|
69
|
Buonvicino D, Felici R, Ranieri G, Caramelli R, Lapucci A, Cavone L, Muzzi M, Di Pietro L, Bernardini C, Zwergel C, Valente S, Mai A, Chiarugi A. Effects of Class II-Selective Histone Deacetylase Inhibitor on Neuromuscular Function and Disease Progression in SOD1-ALS Mice. Neuroscience 2018; 379:228-238. [PMID: 29588251 DOI: 10.1016/j.neuroscience.2018.03.022] [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] [Received: 12/22/2017] [Revised: 03/13/2018] [Accepted: 03/15/2018] [Indexed: 12/13/2022]
Abstract
Emerging evidence indicates that transcriptome alterations due to epigenetic deregulation concur to ALS pathogenesis. Accordingly, pan-histone deacetylase (HDAC) inhibitors delay ALS development in mice, but these compounds failed when tested in ALS patients. Possibly, lack of selectivity toward specific classes of HDACs weakens the therapeutic effects of pan-HDAC inhibitors. Here, we tested the effects of the HDAC Class II selective inhibitor MC1568 on disease evolution, motor neuron survival as well as skeletal muscle function in SOD1G93A mice. We report that HDACs did not undergo expression changes during disease evolution in isolated motor neurons of adult mice. Conversely, increase in specific Class II HDACs (-4, -5 and -6) occurs in skeletal muscle of mice with severe neuromuscular impairment. Importantly, treatment with MC1568 causes early improvement of motor performances that vanishes at later stages of disease. Notably, motor improvement is not paralleled by reduced motor neuron degeneration but by increased skeletal muscle electrical potentials, reduced activation of mir206/FGFBP1-dependent muscle reinnervation signaling, and increased muscle expression of myogenic genes.
Collapse
Affiliation(s)
- Daniela Buonvicino
- Department of Health Sciences, Section of Clinical Pharmacology and Oncology, University of Florence, Italy.
| | - Roberta Felici
- Department of Health Sciences, Section of Clinical Pharmacology and Oncology, University of Florence, Italy
| | - Giuseppe Ranieri
- Department of Health Sciences, Section of Clinical Pharmacology and Oncology, University of Florence, Italy
| | - Riccardo Caramelli
- Neurophysiology Unit, Department of Neurology and Psychiatry, Azienda Ospedaliera Careggi, Florence, Italy
| | - Andrea Lapucci
- Department of Health Sciences, Section of Clinical Pharmacology and Oncology, University of Florence, Italy
| | - Leonardo Cavone
- Department of Health Sciences, Section of Clinical Pharmacology and Oncology, University of Florence, Italy
| | - Mirko Muzzi
- Department of Health Sciences, Section of Clinical Pharmacology and Oncology, University of Florence, Italy
| | - Lorena Di Pietro
- Institute of Anatomy and Cell Biology, University Cattolica del Sacro Cuore, Rome, Italy
| | - Camilla Bernardini
- Institute of Anatomy and Cell Biology, University Cattolica del Sacro Cuore, Rome, Italy
| | - Clemens Zwergel
- Pasteur Institute, Cenci Bolognetti Foundation, Sapienza University of Rome, Italy
| | - Sergio Valente
- Pasteur Institute, Cenci Bolognetti Foundation, Sapienza University of Rome, Italy
| | - Antonello Mai
- Pasteur Institute, Cenci Bolognetti Foundation, Sapienza University of Rome, Italy
| | - Alberto Chiarugi
- Department of Health Sciences, Section of Clinical Pharmacology and Oncology, University of Florence, Italy
| |
Collapse
|
70
|
Lehmann S, Esch E, Hartmann P, Goswami A, Nikolin S, Weis J, Beyer C, Johann S. Expression profile of pattern recognition receptors in skeletal muscle of SOD1 (G93A) amyotrophic lateral sclerosis (ALS) mice and sporadic ALS patients. Neuropathol Appl Neurobiol 2018; 44:606-627. [PMID: 29575052 DOI: 10.1111/nan.12483] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2017] [Accepted: 02/20/2018] [Indexed: 12/14/2022]
Abstract
AIMS Amyotrophic lateral sclerosis (ALS) is characterized by degeneration of motoneurons and progressive muscle wasting. Inflammatory processes, mediated by non-neuronal cells, such as glial cells, are known to contribute to disease progression. Inflammasomes consist of pattern recognition receptors (PRRs), apoptosis-associated speck-like protein (ASC) and caspase 1 and are essential for interleukin (IL) processing and a rapid immune response after tissue damage. Recently, we described inflammasome activation in the spinal cord of ALS patients and in SOD1(G93A) ALS mice. Since pathological changes in the skeletal muscle are early events in ALS, we hypothesized that PRRs might be abnormally expressed in muscle fibre degeneration. METHODS Western blot analysis, real-time PCR and immunohistochemistry were performed with muscle tissue from presymptomatic and early-symptomatic male SOD1(G93A) mice and with muscle biopsies of control and sporadic ALS (sALS) patients. Analysed PRRs include nucleotide-binding oligomerization domain-like (NOD-like) receptor protein 1 (NLRP1), NLR protein 3 (NLRP3), NLR family CARD domain-containing 4 (NLRC4) and absent in melanoma 2. Additionally, expression levels of ASC, caspase 1, interleukin 1 beta (IL1β) and interleukin 18 (IL18) were evaluated. RESULTS Expression of PRRs and ASC was detected in murine and human tissue. The PRR NLRC4, caspase 1 and IL1β were significantly elevated in denervated muscle of SOD1(G93A) mice and sALS patients. Furthermore, levels of caspase 1 and IL1β were already increased in presymptomatic animals. CONCLUSION Our findings suggest that increased inflammasome activation may be involved in skeletal muscle pathology in ALS. Furthermore, elevated levels of NLRC4, caspase 1 and IL1β reflect early changes in the skeletal muscle and may contribute to the denervation process.
Collapse
Affiliation(s)
- S Lehmann
- Institute of Neuroanatomy, Medical Clinic RWTH Aachen University, Aachen, Germany.,Institute Molecular and Cellular Anatomy (MOCA), Medical Clinic RWTH Aachen University, Aachen, Germany
| | - E Esch
- Institute of Neuroanatomy, Medical Clinic RWTH Aachen University, Aachen, Germany
| | - P Hartmann
- Institute of Neuroanatomy, Medical Clinic RWTH Aachen University, Aachen, Germany
| | - A Goswami
- Institute of Neuropathology, Medical Clinic RWTH Aachen University, Aachen, Germany
| | - S Nikolin
- Institute of Neuropathology, Medical Clinic RWTH Aachen University, Aachen, Germany
| | - J Weis
- Institute of Neuropathology, Medical Clinic RWTH Aachen University, Aachen, Germany
| | - C Beyer
- Institute of Neuroanatomy, Medical Clinic RWTH Aachen University, Aachen, Germany.,JARA - Translational Brain Medicine, Aachen, Germany
| | - S Johann
- Institute of Neuroanatomy, Medical Clinic RWTH Aachen University, Aachen, Germany.,Institute of Anatomy II, Medical Faculty Heinrich-Heine-University, Düsseldorf, Germany
| |
Collapse
|
71
|
Shanmukha S, Narayanappa G, Nalini A, Alladi PA, Raju TR. Sporadic amyotrophic lateral sclerosis (SALS) - skeletal muscle response to cerebrospinal fluid from SALS patients in a rat model. Dis Model Mech 2018; 11:11/4/dmm031997. [PMID: 29666144 PMCID: PMC5963857 DOI: 10.1242/dmm.031997] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2017] [Accepted: 03/05/2018] [Indexed: 01/17/2023] Open
Abstract
Skeletal muscle atrophy is the most prominent feature of amyotrophic lateral sclerosis (ALS), an adult-onset neurodegenerative disease of motor neurons. However, the contribution of skeletal muscle to disease progression remains elusive. Our previous studies have shown that intrathecal injection of cerebrospinal fluid from sporadic ALS patients (ALS-CSF) induces several degenerative changes in motor neurons and glia of neonatal rats. Here, we describe various pathologic events in the rat extensor digitorum longus muscle following intrathecal injection of ALS-CSF. Adenosine triphosphatase staining and electron microscopic (EM) analysis revealed significant atrophy and grouping of type 2 fibres in ALS-CSF-injected rats. Profound neuromuscular junction (NMJ) damage, such as fragmentation accompanied by denervation, were revealed by α-bungarotoxin immunostaining. Altered expression of key NMJ proteins, rapsyn and calpain, was also observed by immunoblotting. In addition, EM analysis showed sarcolemmal folding, Z-line streaming, structural alterations of mitochondria and dilated sarcoplasmic reticulum. The expression of trophic factors was affected, with significant downregulation of vascular endothelial growth factor (VEGF), marginal reduction in insulin-like growth factor-1 (IGF-1), and upregulation of brain-derived neurotrophic factor (BDNF) and glial-derived neurotrophic factor (GDNF). However, motor neurons might be unable to harness the enhanced levels of BDNF and GDNF, owing to impaired NMJs. We propose that ALS-CSF triggers motor neuronal degeneration, resulting in pathological changes in the skeletal muscle. Muscle damage further aggravates the motor neuronal pathology, because of the interdependency between them. This sets in a vicious cycle, leading to rapid and progressive loss of motor neurons, which could explain the relentless course of ALS.This article has an associated First Person interview with the first author of the paper.
Collapse
Affiliation(s)
- Shruthi Shanmukha
- Department of Neurophysiology, National Institute of Mental Health and Neurosciences, Hosur Road, Bangalore 560 029, India
| | - Gayathri Narayanappa
- Department of Neuropathology, National Institute of Mental Health and Neurosciences, Hosur Road, Bangalore 560 029, India
| | - Atchayaram Nalini
- Department of Neurology, National Institute of Mental Health and Neurosciences, Hosur Road, Bangalore 560 029, India
| | - Phalguni Anand Alladi
- Department of Neurophysiology, National Institute of Mental Health and Neurosciences, Hosur Road, Bangalore 560 029, India
| | - Trichur R Raju
- Department of Neurophysiology, National Institute of Mental Health and Neurosciences, Hosur Road, Bangalore 560 029, India
| |
Collapse
|
72
|
Sebastião AM, Rei N, Ribeiro JA. Amyotrophic Lateral Sclerosis (ALS) and Adenosine Receptors. Front Pharmacol 2018; 9:267. [PMID: 29713276 PMCID: PMC5911503 DOI: 10.3389/fphar.2018.00267] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2017] [Accepted: 03/09/2018] [Indexed: 12/11/2022] Open
Abstract
In the present review we discuss the potential involvement of adenosinergic signaling, in particular the role of adenosine receptors, in amyotrophic lateral sclerosis (ALS). Though the literature on this topic is not abundant, the information so far available on adenosine receptors in animal models of ALS highlights the interest to continue to explore the role of these receptors in this neurodegenerative disease. Indeed, all motor neurons affected in ALS are responsive to adenosine receptor ligands but interestingly, there are alterations in pre-symptomatic or early symptomatic stages that mirror those in advanced disease stages. Information starts to emerge pointing toward a beneficial role of A2A receptors (A2AR), most probably at early disease states, and a detrimental role of caffeine, in clear contrast with what occurs in other neurodegenerative diseases. However, some evidence also exists on a beneficial action of A2AR antagonists. It may happen that there are time windows where A2AR prove beneficial and others where their blockade is required. Furthermore, the same changes may not occur simultaneously at the different synapses. In line with this, it is not fully understood if ALS is a dying back disease or if it propagates in a centrifugal way. It thus seems crucial to understand how motor neuron dysfunction occurs, how adenosine receptors are involved in those dysfunctions and whether the early changes in purinergic signaling are compensatory or triggers for the disease. Getting this information is crucial before starting the design of purinergic based strategies to halt or delay disease progression.
Collapse
Affiliation(s)
- Ana M Sebastião
- Instituto de Farmacologia e Neurociências, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal.,Instituto de Medicina Molecular, Universidade de Lisboa, Lisbon, Portugal
| | - Nádia Rei
- Instituto de Farmacologia e Neurociências, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal.,Instituto de Medicina Molecular, Universidade de Lisboa, Lisbon, Portugal
| | - Joaquim A Ribeiro
- Instituto de Farmacologia e Neurociências, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal.,Instituto de Medicina Molecular, Universidade de Lisboa, Lisbon, Portugal
| |
Collapse
|
73
|
24S-hydroxycholesterol suppresses neuromuscular transmission in SOD1(G93A) mice: A possible role of NO and lipid rafts. Mol Cell Neurosci 2018; 88:308-318. [PMID: 29550246 DOI: 10.1016/j.mcn.2018.03.006] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2017] [Revised: 02/08/2018] [Accepted: 03/12/2018] [Indexed: 12/12/2022] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a neurodegenerative disorder characterized by the initial denervation of skeletal muscle and subsequent death of motor neurons. A dying-back pattern of ALS suggests a crucial role for neuromuscular junction dysfunction. In the present study, microelectrode recording of postsynaptic currents and optical detection of synaptic vesicle traffic (FM1-43 dye) and intracellular NO levels (DAF-FM DA) were used to examine the effect of the major brain-derived cholesterol metabolite 24S-hydroxycholesterol (24S-HC, 0.4 μM) on neuromuscular transmission in the diaphragm of transgenic mice carrying a mutant superoxide dismutase 1 (SODG93A). We found that 24S-HC suppressed spontaneous neurotransmitter release and neurotransmitter exocytosis during high-frequency stimulation. The latter was accompanied by a decrease in both the rate of synaptic vesicle recycling and activity-dependent enhancement of NO production. Inhibition of NO synthase with L-NAME also attenuated synaptic vesicle exocytosis during high-frequency stimulation and completely abolished the effect of 24S-HC itself. Of note, 24S-HC enhanced the labeling of synaptic membranes with B-subunit of cholera toxin, suggesting an increase in lipid ordering. Lipid raft-disrupting agents (methyl-β-cyclodextrin, sphingomyelinase) prevented the action of 24S-HC on both lipid raft marker labeling and NO synthesis. Together, these experiments indicate that 24S-HC is able to suppress the exocytotic release of neurotransmitter in response to intense activity via a NO/lipid raft-dependent pathway in the neuromuscular junctions of SODG93A mice.
Collapse
|
74
|
Miyoshi S, Tezuka T, Arimura S, Tomono T, Okada T, Yamanashi Y. DOK7 gene therapy enhances motor activity and life span in ALS model mice. EMBO Mol Med 2018; 9:880-889. [PMID: 28490573 PMCID: PMC5494517 DOI: 10.15252/emmm.201607298] [Citation(s) in RCA: 57] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a progressive, multifactorial motor neurodegenerative disease with severe muscle atrophy. The glutamate release inhibitor riluzole is the only medication approved by the FDA, and prolongs patient life span by a few months, testifying to a strong need for new treatment strategies. In ALS, motor neuron degeneration first becomes evident at the motor nerve terminals in neuromuscular junctions (NMJs), the cholinergic synapse between motor neuron and skeletal muscle; degeneration then progresses proximally, implicating the NMJ as a therapeutic target. We previously demonstrated that activation of muscle‐specific kinase MuSK by the cytoplasmic protein Dok‐7 is essential for NMJ formation, and forced expression of Dok‐7 in muscle activates MuSK and enlarges NMJs. Here, we show that therapeutic administration of an adeno‐associated virus vector encoding the human DOK7 gene suppressed motor nerve terminal degeneration at NMJs together with muscle atrophy in the SOD1‐G93A ALS mouse model. Ultimately, we show that DOK7 gene therapy enhanced motor activity and life span in ALS model mice.
Collapse
Affiliation(s)
- Sadanori Miyoshi
- Division of Genetics, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Tohru Tezuka
- Division of Genetics, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Sumimasa Arimura
- Division of Genetics, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Taro Tomono
- Department of Biochemistry and Molecular Biology, Nippon Medical School, Tokyo, Japan.,Graduate School of Comprehensive Human Sciences, Majors in Medical Sciences University of Tsukuba, Ibaraki, Japan
| | - Takashi Okada
- Department of Biochemistry and Molecular Biology, Nippon Medical School, Tokyo, Japan
| | - Yuji Yamanashi
- Division of Genetics, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| |
Collapse
|
75
|
Cantor S, Zhang W, Delestrée N, Remédio L, Mentis GZ, Burden SJ. Preserving neuromuscular synapses in ALS by stimulating MuSK with a therapeutic agonist antibody. eLife 2018; 7:34375. [PMID: 29460776 PMCID: PMC5837562 DOI: 10.7554/elife.34375] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2017] [Accepted: 02/02/2018] [Indexed: 12/22/2022] Open
Abstract
In amyotrophic lateral sclerosis (ALS) and animal models of ALS, including SOD1-G93A mice, disassembly of the neuromuscular synapse precedes motor neuron loss and is sufficient to cause a decline in motor function that culminates in lethal respiratory paralysis. We treated SOD1-G93A mice with an agonist antibody to MuSK, a receptor tyrosine kinase essential for maintaining neuromuscular synapses, to determine whether increasing muscle retrograde signaling would slow nerve terminal detachment from muscle. The agonist antibody, delivered after disease onset, slowed muscle denervation, promoting motor neuron survival, improving motor system output, and extending the lifespan of SOD1-G93A mice. These findings suggest a novel therapeutic strategy for ALS, using an antibody format with clinical precedence, which targets a pathway essential for maintaining attachment of nerve terminals to muscle. Amyotrophic lateral sclerosis – often shortened to ALS – is a disease that starts with difficulties moving and progresses to paralysis of many muscles, including those used for breathing. The disease is usually lethal, with patients rarely surviving more than a few years after diagnosis. There is no cure or effective treatment for the disease. It begins with the breakdown of the connections, or synapses, between the muscles and the nerve cells that connect with them. After this, the nerve cell itself breaks down. Many therapeutic approaches have focused on attempts to prevent the nerve cells from dying, but few target the initial degeneration of the synapse. Cantor et al. asked if intervening when the synapse has already begun to break down could slow the progression of the disease in mice with ALS. Their approach involved using an antibody to bind to a receptor protein called MuSK, which plays an important role in maintaining the synapse between muscle and nerve cell. The antibody boosted the receptor’s activity, helping to preserve synapses, including those that connect nerve cells to the diaphragm muscle. The experiments showed that the antibody treatment led to fewer synapses breaking down, and kept more of the nerve cells alive. Healthier connections between the nervous system and the diaphragm improved the function of this muscle. As a result, the mice given the antibody treatment had a slightly extended lifespan, compared with those given no treatment. The findings suggest a possible new way to develop treatments for ALS, which could be used in combination with other therapies, such as those aimed at improving the health of the nerve cells. Together, this could improve quality of life for the majority of patients with ALS. Similar strategies could be used to develop treatments to preserve synapses in other neurodegenerative diseases, such as Alzheimer’s, Parkinson’s and Huntington’s disease, as well as some kinds of dementia. Preserving synapses early on, before the significant loss of nerve cells, could help to slow the progression of these diseases, improve the patients' quality of life and extend their lifespans too.
Collapse
Affiliation(s)
- Sarah Cantor
- Molecular Neurobiology Program, Helen L. and Martin S. Kimmel Center for Biology and Medicine at the Skirball Institute of Biomolecular Medicine, NYU Medical School, New York, United States
| | - Wei Zhang
- Molecular Neurobiology Program, Helen L. and Martin S. Kimmel Center for Biology and Medicine at the Skirball Institute of Biomolecular Medicine, NYU Medical School, New York, United States
| | - Nicolas Delestrée
- Center for Motor Neuron Biology and Disease and Departments of Pathology and Cell Biology and Neurology, Columbia University, New York, United States
| | - Leonor Remédio
- Molecular Neurobiology Program, Helen L. and Martin S. Kimmel Center for Biology and Medicine at the Skirball Institute of Biomolecular Medicine, NYU Medical School, New York, United States
| | - George Z Mentis
- Center for Motor Neuron Biology and Disease and Departments of Pathology and Cell Biology and Neurology, Columbia University, New York, United States
| | - Steven J Burden
- Molecular Neurobiology Program, Helen L. and Martin S. Kimmel Center for Biology and Medicine at the Skirball Institute of Biomolecular Medicine, NYU Medical School, New York, United States
| |
Collapse
|
76
|
Starr A, Sattler R. Synaptic dysfunction and altered excitability in C9ORF72 ALS/FTD. Brain Res 2018; 1693:98-108. [PMID: 29453960 DOI: 10.1016/j.brainres.2018.02.011] [Citation(s) in RCA: 52] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2017] [Revised: 02/06/2018] [Accepted: 02/10/2018] [Indexed: 02/08/2023]
Abstract
Amyotrophic lateral sclerosis (ALS) is characterized by a progressive degeneration of upper and lower motor neurons, resulting in fatal paralysis due to denervation of the muscle. Due to genetic, pathological and symptomatic overlap, ALS is now considered a spectrum disease together with frontotemporal dementia (FTD), the second most common cause of dementia in individuals under the age of 65. Interestingly, in both diseases, there is a large prevalence of RNA binding proteins (RBPs) that are mutated and considered disease-causing, or whose dysfunction contribute to disease pathogenesis. The most common shared genetic mutation in ALS/FTD is a hexanucleuotide repeat expansion within intron 1 of C9ORF72 (C9). Three potentially overlapping, putative toxic mechanisms have been proposed: loss of function due to haploinsufficient expression of the C9ORF72 mRNA, gain of function of the repeat RNA aggregates, or RNA foci, and repeat-associated non-ATG-initiated translation (RAN) of the repeat RNA into toxic dipeptide repeats (DPRs). Regardless of the causative mechanism, disease symptoms are ultimately caused by a failure of neurotransmission in three regions: the brain, the spinal cord, and the neuromuscular junction. Here, we review C9 ALS/FTD-associated synaptic dysfunction and aberrant neuronal excitability in these three key regions, focusing on changes in morphology and synapse formation, excitability, and excitotoxicity in patients, animal models, and in vitro models. We compare these deficits to those seen in other forms of ALS and FTD in search of shared pathways, and discuss the potential targeting of synaptic dysfunctions for therapeutic intervention in ALS and FTD patients.
Collapse
Affiliation(s)
- Alexander Starr
- Division of Neurobiology, Barrow Neurological Institute, St. Joseph's Hospital and Medical Center, Phoenix, AZ 85013, United States
| | - Rita Sattler
- Division of Neurobiology, Barrow Neurological Institute, St. Joseph's Hospital and Medical Center, Phoenix, AZ 85013, United States.
| |
Collapse
|
77
|
Chand KK, Lee KM, Lee JD, Qiu H, Willis EF, Lavidis NA, Hilliard MA, Noakes PG. Defects in synaptic transmission at the neuromuscular junction precede motor deficits in a TDP‐43
Q331K
transgenic mouse model of amyotrophic lateral sclerosis. FASEB J 2018; 32:2676-2689. [DOI: 10.1096/fj.201700835r] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Affiliation(s)
- Kirat K. Chand
- School of Biomedical Sciences Brisbane Queensland Australia
| | - Kah Meng Lee
- School of Biomedical Sciences Brisbane Queensland Australia
| | - John D. Lee
- School of Biomedical Sciences Brisbane Queensland Australia
| | - Hao Qiu
- School of Biomedical Sciences Brisbane Queensland Australia
| | | | | | | | | |
Collapse
|
78
|
Bowerman M, Murray LM, Scamps F, Schneider BL, Kothary R, Raoul C. Pathogenic commonalities between spinal muscular atrophy and amyotrophic lateral sclerosis: Converging roads to therapeutic development. Eur J Med Genet 2017; 61:685-698. [PMID: 29313812 DOI: 10.1016/j.ejmg.2017.12.001] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2017] [Revised: 10/04/2017] [Accepted: 12/03/2017] [Indexed: 12/12/2022]
Abstract
Spinal muscular atrophy (SMA) and amyotrophic lateral sclerosis (ALS) are the two most common motoneuron disorders, which share typical pathological hallmarks while remaining genetically distinct. Indeed, SMA is caused by deletions or mutations in the survival motor neuron 1 (SMN1) gene whilst ALS, albeit being mostly sporadic, can also be caused by mutations within genes, including superoxide dismutase 1 (SOD1), Fused in Sarcoma (FUS), TAR DNA-binding protein 43 (TDP-43) and chromosome 9 open reading frame 72 (C9ORF72). However, it has come to light that these two diseases may be more interlinked than previously thought. Indeed, it has recently been found that FUS directly interacts with an Smn-containing complex, mutant SOD1 perturbs Smn localization, Smn depletion aggravates disease progression of ALS mice, overexpression of SMN in ALS mice significantly improves their phenotype and lifespan, and duplications of SMN1 have been linked to sporadic ALS. Beyond genetic interactions, accumulating evidence further suggests that both diseases share common pathological identities such as intrinsic muscle defects, neuroinflammation, immune organ dysfunction, metabolic perturbations, defects in neuron excitability and selective motoneuron vulnerability. Identifying common molecular effectors that mediate shared pathologies in SMA and ALS would allow for the development of therapeutic strategies and targeted gene therapies that could potentially alleviate symptoms and be equally beneficial in both disorders. In the present review, we will examine our current knowledge of pathogenic commonalities between SMA and ALS, and discuss how furthering this understanding can lead to the establishment of novel therapeutic approaches with wide-reaching impact on multiple motoneuron diseases.
Collapse
Affiliation(s)
- Melissa Bowerman
- School of Medicine, Keele University, Staffordshire, United Kingdom; Institute for Science and Technology in Medicine, Stoke-on-Trent, United Kingdom; Wolfson Centre for Inherited Neuromuscular Disease, RJAH Orthopaedic Hospital, Oswestry, United Kingdom
| | - Lyndsay M Murray
- Euan McDonald Centre for Motor Neuron Disease Research and Centre for Integrative Physiology, University of Edinburgh, Edinburgh, United Kingdom
| | - Frédérique Scamps
- The Institute for Neurosciences of Montpellier, Inserm UMR1051, Univ Montpellier, Saint Eloi Hospital, Montpellier, France
| | - Bernard L Schneider
- Brain Mind Institute, Ecole Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
| | - Rashmi Kothary
- Regenerative Medicine Program, Ottawa Hospital Research Institute, Ottawa, Canada; Departments of Medicine and Cellular and Molecular Medicine, University of Ottawa, Ottawa, Canada
| | - Cédric Raoul
- The Institute for Neurosciences of Montpellier, Inserm UMR1051, Univ Montpellier, Saint Eloi Hospital, Montpellier, France.
| |
Collapse
|
79
|
Cappello V, Francolini M. Neuromuscular Junction Dismantling in Amyotrophic Lateral Sclerosis. Int J Mol Sci 2017; 18:ijms18102092. [PMID: 28972545 PMCID: PMC5666774 DOI: 10.3390/ijms18102092] [Citation(s) in RCA: 70] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2017] [Revised: 09/25/2017] [Accepted: 09/28/2017] [Indexed: 12/13/2022] Open
Abstract
Neuromuscular junction assembly and plasticity during embryonic, postnatal, and adult life are tightly regulated by the continuous cross-talk among motor nerve endings, muscle fibers, and glial cells. Altered communications among these components is thought to be responsible for the physiological age-related changes at this synapse and possibly for its destruction in pathological states. Neuromuscular junction dismantling plays a crucial role in the onset of Amyotrophic Lateral Sclerosis (ALS). ALS is characterized by the degeneration and death of motor neurons leading to skeletal muscle denervation, atrophy and, most often, death of the patient within five years from diagnosis. ALS is a non-cell autonomous disease as, besides motor neuron degeneration, glial cells, and possibly muscle fibers, play a role in its onset and progression. Here, we will review the recent literature regarding the mechanisms leading to neuromuscular junction disassembly and muscle denervation focusing on the role of the three players of this peripheral tripartite synapse.
Collapse
Affiliation(s)
- Valentina Cappello
- Center for Nanotechnology Innovation@NEST, Istituto Italiano di Tecnologia Piazza San Silvestro 12, 56127 Pisa, Italy.
| | - Maura Francolini
- Department of Medical Biotechnology and Translational Medicine, Università degli Studi di Milano-Via Vanvitelli 32, 20129 Milano, Italy.
| |
Collapse
|
80
|
Defects in Neuromuscular Transmission May Underlie Motor Dysfunction in Spinal and Bulbar Muscular Atrophy. J Neurosci 2017; 36:5094-106. [PMID: 27147661 DOI: 10.1523/jneurosci.3485-15.2016] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2015] [Accepted: 03/22/2016] [Indexed: 11/21/2022] Open
Abstract
UNLABELLED Spinal and bulbar muscular atrophy (SBMA) in men is an androgen-dependent neuromuscular disease caused by expanded CAG repeats in the androgen receptor (AR). Whether muscle or motor neuron dysfunction or both underlies motor impairment in SBMA is unknown. Muscles of SBMA mice show significant contractile dysfunction, implicating them as a likely source of motor dysfunction, but whether disease also impairs neuromuscular transmission is an open question. Thus, we examined synaptic function in three well-studied SBMA mouse models-the AR97Q, knock-in (KI), and myogenic141 models-by recording in vitro miniature and evoked end-plate potentials (MEPPs and EPPs, respectively) intracellularly from adult muscle fibers. We found striking defects in neuromuscular transmission suggesting that toxic AR in SBMA impairs both presynaptic and postsynaptic mechanisms. Notably, SBMA causes neuromuscular synapses to become weak and muscles to become hyperexcitable in all three models. Presynaptic defects included deficits in quantal content, reduced size of the readily releasable pool, and impaired short-term facilitation. Postsynaptic defects included prolonged decay times for both MEPPs and EPPs, marked resistance to μ-conotoxin (a sodium channel blocker), and enhanced membrane excitability. Quantitative PCR revealed robust upregulation of mRNAs encoding neonatal isoforms of the AChR (γ-subunit) and the voltage-gated sodium channel (NaV1.5) in diseased adult muscles of all three models, consistent with the observed slowing of synaptic potentials and resistance to μ-conotoxin. These findings suggest that muscles of SBMA patients regress to an immature state that impairs neuromuscular function. SIGNIFICANCE STATEMENT We have discovered that SBMA is accompanied by marked defects in neuromuscular synaptic transmission involving both presynaptic and postsynaptic mechanisms. For three different mouse models, we find that diseased synapses are weak, having reduced quantal content due to reductions in the size of the readily releasable pool and/or probability of release. Synaptic potentials in diseased adult fibers are slowed, explained by an aberrant upregulation of the neonatal isoform of the acetylcholine receptor. Diseased fibers also show marked resistance to μ-conotoxin, explained by an aberrant upregulation in the neonatal isoform of the sodium channel, and are hyperexcitable, reminiscent of myotonic dystrophy, showing anode-break action potentials. This work identifies several new molecular targets for recovering function in SBMA.
Collapse
|
81
|
Opposite Synaptic Alterations at the Neuromuscular Junction in an ALS Mouse Model: When Motor Units Matter. J Neurosci 2017; 37:8901-8918. [PMID: 28821658 DOI: 10.1523/jneurosci.3090-16.2017] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2016] [Revised: 06/29/2017] [Accepted: 08/02/2017] [Indexed: 12/13/2022] Open
Abstract
Denervation of the neuromuscular junction (NMJ) precedes the loss of motor neurons (MNs) in amyotrophic lateral sclerosis (ALS). ALS is characterized by a motor unit (MU)-dependent vulnerability where MNs with fast-fatigable (FF) characteristics are lost first, followed by fast fatigue-resistant (FR) and slow (S) MNs. However, changes in NMJ properties as a function of MU types remain debated. We hypothesized that NMJ synaptic functions would be altered precociously in an MU-specific manner, before structural alterations of the NMJ. Synaptic transmission and morphological changes of NMJs have been explored in two nerve-muscle preparations of male SOD1G37R mice and their wild-type (WT) littermates: the soleus (S and FR MU); and the extensor digitorum longus (FF MU). S, FR, and FF NMJs of WT mice showed distinct synaptic properties from which we build an MU synaptic profile (MUSP) that reports MU-dependent NMJ synaptic properties. At postnatal day 180 (P180), FF and S NMJs of SOD1 already showed, respectively, lower and higher quantal content compared with WT mice, before signs of MN death and before NMJ morphological alterations. Changes persisted in both muscles until preonset (P380), while denervation was frequent in the mutant mouse. MN death was evident at this stage. Additional changes occurred at clinical disease onset (P450) for S and FR MU. As a whole, our results reveal a reversed MUSP in SOD1 mutants and highlight MU-specific synaptic changes occurring in a precise temporal sequence. Importantly, changes in synaptic properties appear to be good predictors of vulnerability to neurodegeneration.SIGNIFICANCE STATEMENT The inadequate excitability of motor neurons and their output, the neuromuscular junctions (NMJs), has been considered a key factor in the detrimental outcome of the motor function in amyotrophic lateral sclerosis. However, a conundrum persists at the NMJ whereby persistent but incoherent opposite neurotransmission changes have been reported to take place. This article untangles this conundrum by systematically analyzing the changes in synaptic properties over the course of the disease progression as a function of the motor unit type. This temporal analysis reveals that early synaptic alterations evolve with disease progression but precede NMJ neurodegeneration. These data provide a novel framework of analysis and comparison of synaptic transmission alterations in neurodegenerative disorders.
Collapse
|
82
|
MacDonald R, Barbat-Artigas S, Cho C, Peng H, Shang J, Moustaine A, Carbonetto S, Robitaille R, Chalifour LE, Paudel H. A Novel Egr-1-Agrin Pathway and Potential Implications for Regulation of Synaptic Physiology and Homeostasis at the Neuromuscular Junction. Front Aging Neurosci 2017; 9:258. [PMID: 28824419 PMCID: PMC5541023 DOI: 10.3389/fnagi.2017.00258] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2017] [Accepted: 07/19/2017] [Indexed: 12/11/2022] Open
Abstract
Synaptic transmission requires intricate coordination of the components involved in processing of incoming signals, formation and stabilization of synaptic machinery, neurotransmission and in all related signaling pathways. Changes to any of these components cause synaptic imbalance and disruption of neuronal circuitry. Extensive studies at the neuromuscular junction (NMJ) have greatly aided in the current understanding of synapses and served to elucidate the underlying physiology as well as associated adaptive and homeostatic processes. The heparan sulfate proteoglycan agrin is a vital component of the NMJ, mediating synaptic formation and maintenance in both brain and muscle, but very little is known about direct control of its expression. Here, we investigated the relationship between agrin and transcription factor early growth response-1 (Egr-1), as Egr-1 regulates the expression of many genes involved in synaptic homeostasis and plasticity. Using chromatin immunoprecipitation (ChIP), cell culture with cell lines derived from brain and muscle, and animal models, we show that Egr-1 binds to the AGRN gene locus and suppresses its expression. When compared with wild type (WT), mice deficient in Egr-1 (Egr-1−/−) display a marked increase in AGRN mRNA and agrin full-length and cleavage fragment protein levels, including the 22 kDa, C-terminal fragment in brain and muscle tissue homogenate. Because agrin is a crucial component of the NMJ, we explored possible physiological implications of the Egr-1-agrin relationship. In the diaphragm, Egr-1−/− mice display increased NMJ motor endplate density, individual area and area of innervation. In addition to increased density, soleus NMJs also display an increase in fragmented and faint endplates in Egr-1−/− vs. WT mice. Moreover, the soleus NMJ electrophysiology of Egr-1−/− mice revealed increased quantal content and motor testing showed decreased movement and limb muscle strength compared with WT. This study provides evidence for the potential involvement of a novel Egr-1-agrin pathway in synaptic homeostatic and compensatory mechanisms at the NMJ. Synaptic homeostasis is greatly affected by the process of aging. These and other data suggest that changes in Egr-1 expression may directly or indirectly promote age-related pathologies.
Collapse
Affiliation(s)
- Ryen MacDonald
- Lady Davis Institute for Medical Research, Jewish General HospitalMontreal, QC, Canada.,Integrated Program in Neuroscience, McGill UniversityMontreal, QC, Canada
| | | | - Chulmin Cho
- Lady Davis Institute for Medical Research, Jewish General HospitalMontreal, QC, Canada.,Integrated Program in Neuroscience, McGill UniversityMontreal, QC, Canada
| | - Huashan Peng
- Center for Research in NeuroscienceMontreal, QC, Canada
| | - Jijun Shang
- Lady Davis Institute for Medical Research, Jewish General HospitalMontreal, QC, Canada
| | - Ayman Moustaine
- Département de neurosciences, Université de MontréalMontreal, QC, Canada
| | - Salvatore Carbonetto
- Integrated Program in Neuroscience, McGill UniversityMontreal, QC, Canada.,Center for Research in NeuroscienceMontreal, QC, Canada.,Department of Medicine, McGill UniversityMontreal, QC, Canada
| | - Richard Robitaille
- Département de neurosciences, Université de MontréalMontreal, QC, Canada
| | - Lorraine E Chalifour
- Lady Davis Institute for Medical Research, Jewish General HospitalMontreal, QC, Canada.,Department of Medicine, McGill UniversityMontreal, QC, Canada
| | - Hemant Paudel
- Lady Davis Institute for Medical Research, Jewish General HospitalMontreal, QC, Canada.,Integrated Program in Neuroscience, McGill UniversityMontreal, QC, Canada.,Department of Medicine, McGill UniversityMontreal, QC, Canada
| |
Collapse
|
83
|
Muscle Fibers Secrete FGFBP1 to Slow Degeneration of Neuromuscular Synapses during Aging and Progression of ALS. J Neurosci 2017; 37:70-82. [PMID: 28053031 DOI: 10.1523/jneurosci.2992-16.2016] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2016] [Revised: 11/02/2016] [Accepted: 11/07/2016] [Indexed: 01/13/2023] Open
Abstract
The identity of muscle secreted factors critical for the development and maintenance of neuromuscular junctions (NMJs) remains largely unknown. Here, we show that muscle fibers secrete and concentrate the fibroblast growth factor binding protein 1 (FGFBP1) at NMJs. Although FGFBP1 expression increases during development, its expression decreases before NMJ degeneration during aging and in SOD1G93A mice, a mouse model for amyotrophic lateral sclerosis (ALS). Based on these findings, we examined the impact of deleting FGFBP1 on NMJs. In the absence of FGFBP1, NMJs exhibit structural abnormalities in developing and middle age mice. Deletion of FGFBP1 from SOD1G93A mice also accelerates NMJ degeneration and death. Based on these findings, we sought to identify the mechanism responsible for decreased FGFBP1 in stressed skeletal muscles. We show that FGFBP1 expression is inhibited by increased accumulation of the transforming growth factor-β1 (TGF-β1) in skeletal muscles and at their NMJs. These findings suggest that targeting the FGFBP1 and TGF-β1 signaling axis holds promise for slowing age- and disease-related degeneration of NMJs. SIGNIFICANCE STATEMENT The neuromuscular junction (NMJ) is critical for all voluntary movement. Its malformation during development and degeneration in adulthood impairs motor function. Therefore, it is important to identify factors that function to maintain the structural integrity of NMJs. We show that muscle fibers secrete and concentrate the fibroblast growth factor binding protein 1 (FGFBP1) at NMJs. However, FGFBP1 expression decreases in skeletal muscles during aging and before NMJ degeneration in SOD1G93A mice, a mouse model for amyotrophic lateral sclerosis. We show that transforming growth factor-β1 is responsible for the decreased levels of FGFBP1. Importantly, we demonstrate critical roles for FGFBP1 at NMJs in developing, aging and SOD1G93A mice.
Collapse
|
84
|
Depressed Synaptic Transmission and Reduced Vesicle Release Sites in Huntington's Disease Neuromuscular Junctions. J Neurosci 2017; 37:8077-8091. [PMID: 28724748 DOI: 10.1523/jneurosci.0313-17.2017] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2017] [Revised: 07/06/2017] [Accepted: 07/11/2017] [Indexed: 01/25/2023] Open
Abstract
Huntington's disease (HD) is a progressive and fatal degenerative disorder that results in debilitating cognitive and motor dysfunction. Most HD studies have focused on degeneration of the CNS. We previously discovered that skeletal muscle from transgenic R6/2 HD mice is hyperexcitable due to decreased chloride and potassium conductances. The progressive and early onset of these defects suggest a primary myopathy in HD. In this study, we examined the relationship between neuromuscular transmission and skeletal muscle hyperexcitability. We used an ex vivo preparation of the levator auris longus muscle from male and female late-stage R6/2 mice and age-matched wild-type controls. Immunostaining of the synapses and molecular analyses revealed no evidence of denervation. Physiologically, we recorded spontaneous miniature endplate currents (mEPCs) and nerve-evoked EPCs (eEPCs) under voltage-clamp, which, unlike current-clamp records, were independent of the changes in muscle membrane properties. We found a reduction in the number of vesicles released per action potential (quantal content) in R6/2 muscle, which analysis of eEPC variance and morphology indicate is caused by a reduction in the number of vesicle release sites (n) rather than a change in the probability of release (prel). Furthermore, analysis of high-frequency stimulation trains suggests an impairment in vesicle mobilization. The depressed neuromuscular transmission in R6/2 muscle may help compensate for the muscle hyperexcitability and contribute to motor impersistence.SIGNIFICANCE STATEMENT Recent evidence indicates that Huntington's disease (HD) is a multisystem disorder. Our examination of neuromuscular transmission in this study reveals defects in the motor nerve terminal that may compensate for the muscle hyperexcitability in HD. The technique we used eliminates the effects of the altered muscle membrane properties on synaptic currents and thus provides hitherto the most detailed analysis of synaptic transmission in HD. Clinically, the striking depression of neurotransmission we found may help explain the motor impersistence in HD patients. Therapies that target the highly accessible peripheral nerve and muscle system provide a promising new avenue to lessen the debilitating motor symptoms of HD.
Collapse
|
85
|
|
86
|
Cunha C, Santos C, Gomes C, Fernandes A, Correia AM, Sebastião AM, Vaz AR, Brites D. Downregulated Glia Interplay and Increased miRNA-155 as Promising Markers to Track ALS at an Early Stage. Mol Neurobiol 2017; 55:4207-4224. [PMID: 28612258 DOI: 10.1007/s12035-017-0631-2] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2017] [Accepted: 05/22/2017] [Indexed: 12/13/2022]
Abstract
Amyotrophic lateral sclerosis (ALS) is a fatal neurodegenerative disease of unknown cause. Absence of specific targets and biomarkers compromise the development of new therapeutic strategies and of innovative tools to stratify patients and assess their responses to treatment. Here, we investigate changes in neuroprotective-neuroinflammatory actions in the spinal cord of SOD1 G93A mice, at presymptomatic and symptomatic stages to identify stage-specific biomarkers and potential targets. Results showed that in the presymptomatic stage, there are alterations in both astrocytes and microglia, which comprise decreased expression of GFAP and S100B and upregulation of GLT-1, as well as reduced expression of CD11b, M2-phenotype markers, and a set of inflammatory mediators. Reduced levels of Connexin-43, Pannexin-1, CCL21, and CX3CL1 further indicate the existence of a compromised intercellular communication. In contrast, in the symptomatic stage, increased markers of inflammation became evident, such as NF-κB/Nlrp3-inflammasome, Iba1, pro-inflammatory cytokines, and M1-polarizion markers, together with a decreased expression of M2-phenotypic markers. We also observed upregulation of the CX3CL1-CX3CR1 axis, Connexin-43, Pannexin-1, and of microRNAs (miR)-124, miR-125b, miR-146a and miR-21. Reduced motor neuron number and presence of reactive astrocytes with decreased GFAP, GLT-1, and GLAST further characterized this inflammatory stage. Interestingly, upregulation of miR-155 and downregulation of MFG-E8 appear as consistent biomarkers of both presymptomatic and symptomatic stages. We hypothesize that downregulated cellular interplay at the early stages may represent neuroprotective mechanisms against inflammation, SOD1 aggregation, and ALS onset. The present study identified a set of inflamma-miRNAs, NLRP3-inflammasome, HMGB1, CX3CL1-CX3CR1, Connexin-43, and Pannexin-1 as emerging candidates and promising pharmacological targets that may represent potential neuroprotective strategies in ALS therapy.
Collapse
Affiliation(s)
- Carolina Cunha
- Neuron Glia Biology in Health and Disease Group, Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Avenida Professor Gama Pinto, 1649-003, Lisbon, Portugal
| | - Catarina Santos
- Neuron Glia Biology in Health and Disease Group, Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Avenida Professor Gama Pinto, 1649-003, Lisbon, Portugal
| | - Cátia Gomes
- Neuron Glia Biology in Health and Disease Group, Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Avenida Professor Gama Pinto, 1649-003, Lisbon, Portugal
| | - Adelaide Fernandes
- Neuron Glia Biology in Health and Disease Group, Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Avenida Professor Gama Pinto, 1649-003, Lisbon, Portugal.,Department of Biochemistry and Human Biology, Faculty of Pharmacy, Universidade de Lisboa, Lisbon, Portugal
| | | | - Ana Maria Sebastião
- Instituto de Farmacologia e Neurociências, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal.,Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal
| | - Ana Rita Vaz
- Neuron Glia Biology in Health and Disease Group, Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Avenida Professor Gama Pinto, 1649-003, Lisbon, Portugal.,Department of Biochemistry and Human Biology, Faculty of Pharmacy, Universidade de Lisboa, Lisbon, Portugal
| | - Dora Brites
- Neuron Glia Biology in Health and Disease Group, Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Avenida Professor Gama Pinto, 1649-003, Lisbon, Portugal. .,Department of Biochemistry and Human Biology, Faculty of Pharmacy, Universidade de Lisboa, Lisbon, Portugal.
| |
Collapse
|
87
|
Hensel N, Claus P. The Actin Cytoskeleton in SMA and ALS: How Does It Contribute to Motoneuron Degeneration? Neuroscientist 2017; 24:54-72. [PMID: 28459188 DOI: 10.1177/1073858417705059] [Citation(s) in RCA: 55] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Amyotrophic lateral sclerosis (ALS) and spinal muscular atrophy (SMA) are neurodegenerative diseases with overlapping clinical phenotypes based on impaired motoneuron function. However, the pathomechanisms of both diseases are largely unknown, and it is still unclear whether they converge on the molecular level. SMA is a monogenic disease caused by low levels of functional Survival of Motoneuron (SMN) protein, whereas ALS involves multiple genes as well as environmental factors. Recent evidence argues for involvement of actin regulation as a causative and dysregulated process in both diseases. ALS-causing mutations in the actin-binding protein profilin-1 as well as the ability of the SMN protein to directly bind to profilins argue in favor of a common molecular mechanism involving the actin cytoskeleton. Profilins are major regulat ors of actin-dynamics being involved in multiple neuronal motility and transport processes as well as modulation of synaptic functions that are impaired in models of both motoneuron diseases. In this article, we review the current literature in SMA and ALS research with a focus on the actin cytoskeleton. We propose a common molecular mechanism that explains the degeneration of motoneurons for SMA and some cases of ALS.
Collapse
Affiliation(s)
- Niko Hensel
- 1 Institute of Neuroanatomy and Cell Biology, Hannover Medical School, Hannover, Germany.,2 Niedersachsen Network on Neuroinfectiology (N-RENNT), Hannover, Germany
| | - Peter Claus
- 1 Institute of Neuroanatomy and Cell Biology, Hannover Medical School, Hannover, Germany.,2 Niedersachsen Network on Neuroinfectiology (N-RENNT), Hannover, Germany.,3 Center for Systems Neuroscience (ZSN), Hannover, Germany
| |
Collapse
|
88
|
Marcuzzo S, Bonanno S, Figini M, Scotti A, Zucca I, Minati L, Riva N, Domi T, Fossaghi A, Quattrini A, Galbardi B, D'Alessandro S, Bruzzone MG, García-Verdugo JM, Moreno-Manzano V, Mantegazza R, Bernasconi P. A longitudinal DTI and histological study of the spinal cord reveals early pathological alterations in G93A-SOD1 mouse model of amyotrophic lateral sclerosis. Exp Neurol 2017; 293:43-52. [PMID: 28351750 DOI: 10.1016/j.expneurol.2017.03.018] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2016] [Revised: 03/02/2017] [Accepted: 03/24/2017] [Indexed: 12/13/2022]
Abstract
Amyotrophic lateral sclerosis (ALS) is a neurodegenerative disease characterized by selective motor neuron degeneration in the motor cortex, brainstem and spinal cord. It is generally accepted that ALS is caused by death of motor neurons, however the exact temporal cascade of degenerative processes is not yet completely known. To identify the early pathological changes in spinal cord of G93A-SOD1 ALS mice we performed a comprehensive longitudinal analysis employing diffusion-tensor magnetic resonance imaging alongside histology and electron microscopy, in parallel with peripheral nerve histology. We showed the gradient of degeneration appearance in spinal cord white and gray matter, starting earliest in the ventral white matter, due to a cascade of pathological events including axon dysfunction and mitochondrial changes. Notably, we found that even the main sensory regions are affected by the neurodegenerative process at symptomatic disease phase. Overall our results attest the applicability of DTI in determining disease progression in ALS mice. These findings suggest that DTI could be potentially adapted in humans to aid the assessment of ALS progression and eventually the evaluation of treatment efficacy.
Collapse
Affiliation(s)
- Stefania Marcuzzo
- Neurology IV -Neuroimmunology and Neuromuscular Diseases Unit, Fondazione Istituto Neurologico "Carlo Besta", Milan 20133, Italy
| | - Silvia Bonanno
- Neurology IV -Neuroimmunology and Neuromuscular Diseases Unit, Fondazione Istituto Neurologico "Carlo Besta", Milan 20133, Italy; PhD Program in Neuroscience, University of Milano-Bicocca, Milan 20126, Italy
| | - Matteo Figini
- Scientific Department, Fondazione Istituto Neurologico "Carlo Besta", Milan 20133, Italy
| | - Alessandro Scotti
- Scientific Department, Fondazione Istituto Neurologico "Carlo Besta", Milan 20133, Italy
| | - Ileana Zucca
- Scientific Department, Fondazione Istituto Neurologico "Carlo Besta", Milan 20133, Italy
| | - Ludovico Minati
- Scientific Department, Fondazione Istituto Neurologico "Carlo Besta", Milan 20133, Italy
| | - Nilo Riva
- Neuropathology Unit, Institute of Experimental Neurology, Division of Neuroscience, San Raffaele Scientific Institute, Milan 20132, Italy
| | - Teuta Domi
- Neuropathology Unit, Institute of Experimental Neurology, Division of Neuroscience, San Raffaele Scientific Institute, Milan 20132, Italy
| | - Andrea Fossaghi
- Neuropathology Unit, Institute of Experimental Neurology, Division of Neuroscience, San Raffaele Scientific Institute, Milan 20132, Italy
| | - Angelo Quattrini
- Neuropathology Unit, Institute of Experimental Neurology, Division of Neuroscience, San Raffaele Scientific Institute, Milan 20132, Italy
| | - Barbara Galbardi
- Scientific Department, Fondazione Istituto Neurologico "Carlo Besta", Milan 20133, Italy
| | - Sara D'Alessandro
- Neurology IV -Neuroimmunology and Neuromuscular Diseases Unit, Fondazione Istituto Neurologico "Carlo Besta", Milan 20133, Italy
| | | | | | - Victoria Moreno-Manzano
- Neuronal and Tissue Regeneration Laboratory, Centro de Investigación Príncipe Felipe, Valencia 46012, Spain
| | - Renato Mantegazza
- Neurology IV -Neuroimmunology and Neuromuscular Diseases Unit, Fondazione Istituto Neurologico "Carlo Besta", Milan 20133, Italy
| | - Pia Bernasconi
- Neurology IV -Neuroimmunology and Neuromuscular Diseases Unit, Fondazione Istituto Neurologico "Carlo Besta", Milan 20133, Italy.
| |
Collapse
|
89
|
Moloney EB, Hobo B, De Winter F, Verhaagen J. Expression of a Mutant SEMA3A Protein with Diminished Signalling Capacity Does Not Alter ALS-Related Motor Decline, or Confer Changes in NMJ Plasticity after BotoxA-Induced Paralysis of Male Gastrocnemic Muscle. PLoS One 2017; 12:e0170314. [PMID: 28103314 PMCID: PMC5245795 DOI: 10.1371/journal.pone.0170314] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2016] [Accepted: 01/02/2017] [Indexed: 12/12/2022] Open
Abstract
Terminal Schwann cells (TSCs) are specialized cells that envelop the motor nerve terminal, and play a role in the maintenance and regeneration of neuromuscular junctions (NMJs). The chemorepulsive protein semaphorin 3A (SEMA3A) is selectively up-regulated in TSCs on fast-fatigable muscle fibers following experimental denervation of the muscle (BotoxA-induced paralysis or crush injury to the sciatic nerve) or in the motor neuron disease amyotrophic lateral sclerosis (ALS). Re-expression of SEMA3A in this subset of TSCs is thought to play a role in the selective plasticity of nerve terminals as observed in ALS and following BotoxA-induced paralysis. Using a mouse model expressing a mutant SEMA3A with diminished signaling capacity, we studied the influence of SEMA3A signaling at the NMJ with two denervation paradigms; a motor neuron disease model (the G93A-hSOD1 ALS mouse line) and an injury model (BotoxA-induced paralysis). ALS mice that either expressed 1 or 2 mutant SEMA3A alleles demonstrated no difference in ALS-induced decline in motor behavior. We also investigated the effects of BotoxA-induced paralysis on the sprouting capacity of NMJs in the K108N-SEMA3A mutant mouse, and observed no change in the differential neuronal plasticity found at NMJs on fast-fatigable or slow muscle fibers due to the presence of the SEMA3A mutant protein. Our data may be explained by the residual repulsive activity of the mutant SEMA3A, or it may imply that SEMA3A alone is not a key component of the molecular signature affecting NMJ plasticity in ALS or BotoxA-induced paralysis. Interestingly, we did observe a sex difference in motor neuron sprouting behavior after BotoxA-induced paralysis in WT mice which we speculate may be an important factor in the sex dimorphic differences seen in ALS.
Collapse
Affiliation(s)
- Elizabeth B. Moloney
- Department of Regeneration of Sensorimotor Systems, Netherlands Institute for Neuroscience, an Institute of the Royal Netherlands Academy of Arts and Science, Amsterdam, The Netherlands
| | - Barbara Hobo
- Department of Regeneration of Sensorimotor Systems, Netherlands Institute for Neuroscience, an Institute of the Royal Netherlands Academy of Arts and Science, Amsterdam, The Netherlands
| | - Fred De Winter
- Department of Regeneration of Sensorimotor Systems, Netherlands Institute for Neuroscience, an Institute of the Royal Netherlands Academy of Arts and Science, Amsterdam, The Netherlands
- Department of Neurosurgery, Leiden University Medical Centre, Leiden, The Netherlands
| | - Joost Verhaagen
- Department of Regeneration of Sensorimotor Systems, Netherlands Institute for Neuroscience, an Institute of the Royal Netherlands Academy of Arts and Science, Amsterdam, The Netherlands
- Centre for Neurogenomics and Cognitive Research, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
- * E-mail:
| |
Collapse
|
90
|
Chand KK, Lee KM, Lavidis NA, Noakes PG. Loss of laminin‐a4 results in pre‐ and postsynaptic modifications at the neuromuscular junction. FASEB J 2016; 31:1323-1336. [DOI: 10.1096/fj.201600899r] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2016] [Accepted: 12/06/2016] [Indexed: 11/11/2022]
Affiliation(s)
- Kirat K. Chand
- School of Biomedical Sciences St. Lucia Queensland Australia
| | - Kah Meng Lee
- School of Biomedical Sciences St. Lucia Queensland Australia
| | | | - Peter G. Noakes
- School of Biomedical Sciences St. Lucia Queensland Australia
- Queensland Brain InstituteThe University of Queensland St. Lucia Queensland Australia
| |
Collapse
|
91
|
Heitzer M, Kaiser S, Kanagaratnam M, Zendedel A, Hartmann P, Beyer C, Johann S. Administration of 17β-Estradiol Improves Motoneuron Survival and Down-regulates Inflammasome Activation in Male SOD1(G93A) ALS Mice. Mol Neurobiol 2016; 54:8429-8443. [PMID: 27957680 DOI: 10.1007/s12035-016-0322-4] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2016] [Accepted: 11/29/2016] [Indexed: 12/12/2022]
Abstract
Amyotrophic lateral sclerosis (ALS) is a fatal neurodegenerative disease manifested by the progressive loss of upper and lower motoneurons. The pathomechanism of ALS is complex and not yet fully understood. Neuroinflammation is believed to significantly contribute to disease progression. Inflammasome activation was recently shown in the spinal cord of human sporadic ALS patients and in the SOD1(G93A) mouse model for ALS. In the present study, we investigated the neuroprotective and anti-inflammatory effects of 17β-estradiol (E2) treatment in pre-symptomatic and symptomatic male SOD1(G93A) mice. Symptomatic mice with E2 substitution exhibited improved motor performance correlating with an increased survival of motoneurons in the lumbar spinal cord. Expression of NLRP3 inflammasome proteins and levels of activated caspase 1 and mature interleukin 1 beta were significantly reduced in SOD1(G93A) mice supplemented with E2.
Collapse
Affiliation(s)
- Marius Heitzer
- Institute of Neuroanatomy, Faculty of Medicine, RWTH Aachen University, Wendlingweg 2, 52074, Aachen, Germany
| | - Sarah Kaiser
- Institute of Neuroanatomy, Faculty of Medicine, RWTH Aachen University, Wendlingweg 2, 52074, Aachen, Germany
| | - Mithila Kanagaratnam
- Institute of Neuroanatomy, Faculty of Medicine, RWTH Aachen University, Wendlingweg 2, 52074, Aachen, Germany
| | - Adib Zendedel
- Institute of Neuroanatomy, Faculty of Medicine, RWTH Aachen University, Wendlingweg 2, 52074, Aachen, Germany.,Department of Anatomical Sciences, Faculty of Medicine, Guilan University of Medical Sciences, Rasht, Iran
| | - Philipp Hartmann
- Institute of Neuroanatomy, Faculty of Medicine, RWTH Aachen University, Wendlingweg 2, 52074, Aachen, Germany
| | - Cordian Beyer
- Institute of Neuroanatomy, Faculty of Medicine, RWTH Aachen University, Wendlingweg 2, 52074, Aachen, Germany.,JARA-BRAIN, 52074, Aachen, Germany
| | - Sonja Johann
- Institute of Neuroanatomy, Faculty of Medicine, RWTH Aachen University, Wendlingweg 2, 52074, Aachen, Germany.
| |
Collapse
|
92
|
Gasco S, Rando A, Zaragoza P, García-Redondo A, Calvo AC, Osta R. Hematopoietic stem and progenitor cells as novel prognostic biomarkers of longevity in a murine model for amyotrophic lateral sclerosis. Am J Physiol Cell Physiol 2016; 311:C910-C919. [PMID: 27681176 DOI: 10.1152/ajpcell.00081.2016] [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: 03/24/2016] [Accepted: 09/21/2016] [Indexed: 11/22/2022]
Abstract
Amyotrophic lateral sclerosis (ALS) is a fatal neurodegenerative disease with a difficult diagnosis and prognosis. In this regard, new and more reliable biomarkers for the disease are needed. We propose peripheral blood, and, more specifically, the hematopoietic stem and progenitor cells (HSPCs) as potential prognostic biomarkers in the SOD1G93A murine model of ALS. We accurately and serially studied three HSPCs-hematopoietic stem cells (HSCs), common lymphoid progenitors (CLPs), and common myeloid progenitors (CMPs)-in both control and SOD1G93A mice along the disease's progression by RT-PCR and flow cytometry analysis. We found interesting differences for every HSPC type in the transgenic mice compared with the control mice at every time point selected, as well as differences along the disease course. The results showed a maintained compensatory increase of HSCs along disease progression. However, the downregulated levels of CLPs and CMPs suggested an exit of these cell populations to the peripheral tissues, probably due to their supporting role to the damaged tissues. In addition, a positive correlation of the percentage of CLPs and CMPs with the longevity was found, as well as a positive correlation of HSCs and CMPs with motor function and weight, thus reinforcing the idea that HSPCs play a relevant role in the longevity of the SOD1G93A mice. On the basis of these results, both CLPs and CMPs could be considered prognostic biomarkers of longevity in this animal model, opening the door to future studies in human patients for their potential clinical use.
Collapse
Affiliation(s)
- Samanta Gasco
- Laboratorio de Genética Bioquímica, Veterinary Faculty of Zaragoza, Instituto Agroalimentario de Aragón, Health Research Institute of Aragon, University of Zaragoza, Zaragoza, Spain; and
| | - Amaya Rando
- Laboratorio de Genética Bioquímica, Veterinary Faculty of Zaragoza, Instituto Agroalimentario de Aragón, Health Research Institute of Aragon, University of Zaragoza, Zaragoza, Spain; and
| | - Pilar Zaragoza
- Laboratorio de Genética Bioquímica, Veterinary Faculty of Zaragoza, Instituto Agroalimentario de Aragón, Health Research Institute of Aragon, University of Zaragoza, Zaragoza, Spain; and
| | - Alberto García-Redondo
- Biochemistry Department, Centre for Biomedical Network Research on Rare Diseases, Health Research Institute, October 12th Hospital, Madrid, Spain
| | - Ana Cristina Calvo
- Laboratorio de Genética Bioquímica, Veterinary Faculty of Zaragoza, Instituto Agroalimentario de Aragón, Health Research Institute of Aragon, University of Zaragoza, Zaragoza, Spain; and
| | - Rosario Osta
- Laboratorio de Genética Bioquímica, Veterinary Faculty of Zaragoza, Instituto Agroalimentario de Aragón, Health Research Institute of Aragon, University of Zaragoza, Zaragoza, Spain; and
| |
Collapse
|
93
|
Arbour D, Vande Velde C, Robitaille R. New perspectives on amyotrophic lateral sclerosis: the role of glial cells at the neuromuscular junction. J Physiol 2016; 595:647-661. [PMID: 27633977 DOI: 10.1113/jp270213] [Citation(s) in RCA: 51] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2016] [Accepted: 09/12/2016] [Indexed: 12/11/2022] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a disease leading to the death of motor neurons (MNs). It is also recognized as a non-cell autonomous disease where glial cells in the CNS are involved in its pathogenesis and progression. However, although denervation of neuromuscular junctions (NMJs) represents an early and major event in ALS, the importance of glial cells at this synapse receives little attention. An interesting possibility is that altered relationships between glial cells and MNs in the spinal cord in ALS may also take place at the NMJ. Perisynaptic Schwann cells (PSCs), which are glial cells at the NMJ, show great morphological and functional adaptability to ensure NMJ stability, maintenance and repair. More specifically, PSCs change their properties according to the state of innervation. Hence, abnormal changes or lack of changes can have detrimental effects on NMJs in ALS. This review will provide an overview of known and hypothesized interactions between MN nerve terminals and PSCs at NMJs during development, aging and ALS-induced denervation. These neuron-PSC interactions may be crucial to the understanding of how degenerative changes begin and progress at NMJs in ALS, and represent a novel therapeutic target.
Collapse
Affiliation(s)
- Danielle Arbour
- Département de neurosciences, Université de Montréal, Montréal, Québec, Canada, H3C 3J7.,Groupe de recherche sur le système nerveux central, Université de Montréal, Montréal, Québec, Canada, H3C 3J7
| | - Christine Vande Velde
- Département de neurosciences, Université de Montréal, Montréal, Québec, Canada, H3C 3J7.,Groupe de recherche sur le système nerveux central, Université de Montréal, Montréal, Québec, Canada, H3C 3J7.,Centre de recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Montréal, Québec, Canada, H2X 0A9
| | - Richard Robitaille
- Département de neurosciences, Université de Montréal, Montréal, Québec, Canada, H3C 3J7.,Groupe de recherche sur le système nerveux central, Université de Montréal, Montréal, Québec, Canada, H3C 3J7
| |
Collapse
|
94
|
Sugita S, Fleming LL, Wood C, Vaughan SK, Gomes MPSM, Camargo W, Naves LA, Prado VF, Prado MAM, Guatimosim C, Valdez G. VAChT overexpression increases acetylcholine at the synaptic cleft and accelerates aging of neuromuscular junctions. Skelet Muscle 2016; 6:31. [PMID: 27713817 PMCID: PMC5050580 DOI: 10.1186/s13395-016-0105-7] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2016] [Accepted: 08/26/2016] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Cholinergic dysfunction occurs during aging and in a variety of diseases, including amyotrophic lateral sclerosis (ALS). However, it remains unknown whether changes in cholinergic transmission contributes to age- and disease-related degeneration of the motor system. Here we investigated the effect of moderately increasing levels of synaptic acetylcholine (ACh) on the neuromuscular junction (NMJ), muscle fibers, and motor neurons during development and aging and in a mouse model for amyotrophic lateral sclerosis (ALS). METHODS Chat-ChR2-EYFP (VAChTHyp) mice containing multiple copies of the vesicular acetylcholine transporter (VAChT), mutant superoxide dismutase 1 (SOD1G93A), and Chat-IRES-Cre and tdTomato transgenic mice were used in this study. NMJs, muscle fibers, and α-motor neurons' somata and their axons were examined using a light microscope. Transcripts for select genes in muscles and spinal cords were assessed using real-time quantitative PCR. Motor function tests were carried out using an inverted wire mesh and a rotarod. Electrophysiological recordings were collected to examine miniature endplate potentials (MEPP) in muscles. RESULTS We show that VAChT is elevated in the spinal cord and at NMJs of VAChTHyp mice. We also show that the amplitude of MEPPs is significantly higher in VAChTHyp muscles, indicating that more ACh is loaded into synaptic vesicles and released into the synaptic cleft at NMJs of VAChTHyp mice compared to control mice. While the development of NMJs was not affected in VAChTHyp mice, NMJs prematurely acquired age-related structural alterations in adult VAChTHyp mice. These structural changes at NMJs were accompanied by motor deficits in VAChTHyp mice. However, cellular features of muscle fibers and levels of molecules with critical functions at the NMJ and in muscle fibers were largely unchanged in VAChTHyp mice. In the SOD1G93A mouse model for ALS, increasing synaptic ACh accelerated degeneration of NMJs caused motor deficits and resulted in premature death specifically in male mice. CONCLUSIONS The data presented in this manuscript demonstrate that increasing levels of ACh at the synaptic cleft promote degeneration of adult NMJs, contributing to age- and disease-related motor deficits. We thus propose that maintaining normal cholinergic signaling in muscles will slow degeneration of NMJs and attenuate loss of motor function caused by aging and neuromuscular diseases.
Collapse
Affiliation(s)
- Satoshi Sugita
- Virginia Tech Carilion Research Institute, Virginia Tech, Roanoke, VA USA
| | - Leland L. Fleming
- Virginia Tech Carilion Research Institute, Virginia Tech, Roanoke, VA USA
- Virginia Tech Postbaccalaureate Research and Education (VT PREP) Scholar, Virginia Tech, Blacksburg, VA USA
| | - Caleb Wood
- Virginia Tech Carilion Research Institute, Virginia Tech, Roanoke, VA USA
| | - Sydney K. Vaughan
- Virginia Tech Carilion Research Institute, Virginia Tech, Roanoke, VA USA
- Graduate Program in Translational Biology, Medicine, and Health, Virginia Tech, Blacksburg, VA USA
| | - Matheus P. S. M. Gomes
- Departamento de Morfologia, Instiuto Ciencias Biologicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais Brazil
| | - Wallace Camargo
- Departamento de Fisiologia e Biofísica, Instiuto Ciencias Biologicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais Brazil
| | - Ligia A. Naves
- Departamento de Fisiologia e Biofísica, Instiuto Ciencias Biologicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais Brazil
| | - Vania F. Prado
- Robarts Research Institute, Department of Physiology and Pharmacology, Department of Anatomy & Cell Biology, Schulich School of Medicine & Dentistry, University of Western Ontario, London, ON N6A5K8 Canada
| | - Marco A. M. Prado
- Robarts Research Institute, Department of Physiology and Pharmacology, Department of Anatomy & Cell Biology, Schulich School of Medicine & Dentistry, University of Western Ontario, London, ON N6A5K8 Canada
| | - Cristina Guatimosim
- Departamento de Morfologia, Instiuto Ciencias Biologicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais Brazil
| | - Gregorio Valdez
- Virginia Tech Carilion Research Institute, Virginia Tech, Roanoke, VA USA
- Department of Biological Sciences, Virginia Tech, Blacksburg, VA USA
| |
Collapse
|
95
|
Axonal degeneration, distal collateral branching and neuromuscular junction architecture alterations occur prior to symptom onset in the SOD1G93A mouse model of amyotrophic lateral sclerosis. J Chem Neuroanat 2016; 76:35-47. [DOI: 10.1016/j.jchemneu.2016.03.003] [Citation(s) in RCA: 56] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2015] [Revised: 01/29/2016] [Accepted: 03/19/2016] [Indexed: 12/11/2022]
|
96
|
Romer SH, Seedle K, Turner SM, Li J, Baccei ML, Crone SA. Accessory respiratory muscles enhance ventilation in ALS model mice and are activated by excitatory V2a neurons. Exp Neurol 2016; 287:192-204. [PMID: 27456268 DOI: 10.1016/j.expneurol.2016.05.033] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2016] [Revised: 05/09/2016] [Accepted: 05/26/2016] [Indexed: 02/06/2023]
Abstract
Inspiratory accessory respiratory muscles (ARMs) enhance ventilation when demands are high, such as during exercise and/or pathological conditions. Despite progressive degeneration of phrenic motor neurons innervating the diaphragm, amyotrophic lateral sclerosis (ALS) patients and rodent models are able to maintain ventilation at early stages of disease. In order to assess the contribution of ARMs to respiratory compensation in ALS, we examined the activity of ARMs and ventilation throughout disease progression in SOD1G93A ALS model mice at rest using a combination of electromyography and unrestrained whole body plethysmography. Increased ARM activity, accompanied by increased ventilation, is observed beginning at the onset of symptoms. However, ARM recruitment fails to occur at rest at late stages of disease, even though the same ARMs are used for other behaviors. Using a chemogenetic approach, we demonstrate that a glutamatergic class of neurons in the brainstem and spinal cord, the V2a class, is sufficient to drive increased ARM activity at rest in healthy mice. Additionally, we reveal pathology in the medial reticular formation of the brainstem of SOD1G93A mice using immunohistochemistry and confocal imaging. Both spinal and brainstem V2a neurons degenerate in ALS model mice, accompanied by regional activation of astrocytes and microglia. These results establish inspiratory ARM recruitment as one of the compensatory mechanisms that maintains breathing at early stages of disease and indicate that V2a neuron degeneration may contribute to ARM failure at late stages of disease.
Collapse
Affiliation(s)
- Shannon H Romer
- Division of Neurosurgery, Cincinnati Children's Hospital Medical Center, 3333 Burnet Avenue, Cincinnati, OH 45229
| | - Kari Seedle
- Division of Neurosurgery, Cincinnati Children's Hospital Medical Center, 3333 Burnet Avenue, Cincinnati, OH 45229
| | - Sarah M Turner
- Division of Neurosurgery, Cincinnati Children's Hospital Medical Center, 3333 Burnet Avenue, Cincinnati, OH 45229
| | - Jie Li
- Pain Research Center, Dept. of Anesthesiology, University of Cincinnati Medical Center, 231 Albert Sabin Way, Cincinnati, OH 45267
| | - Mark L Baccei
- Pain Research Center, Dept. of Anesthesiology, University of Cincinnati Medical Center, 231 Albert Sabin Way, Cincinnati, OH 45267
| | - Steven A Crone
- Division of Neurosurgery, Cincinnati Children's Hospital Medical Center, 3333 Burnet Avenue, Cincinnati, OH 45229; Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, 3333 Burnet Avenue, Cincinnati, OH 45229.
| |
Collapse
|
97
|
de Carvalho M, Swash M. Lower motor neuron dysfunction in ALS. Clin Neurophysiol 2016; 127:2670-81. [DOI: 10.1016/j.clinph.2016.03.024] [Citation(s) in RCA: 53] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2015] [Revised: 02/08/2016] [Accepted: 03/01/2016] [Indexed: 12/11/2022]
|
98
|
Vernay A, Therreau L, Blot B, Risson V, Dirrig-Grosch S, Waegaert R, Lequeu T, Sellal F, Schaeffer L, Sadoul R, Loeffler JP, René F. A transgenic mouse expressing CHMP2Bintron5 mutant in neurons develops histological and behavioural features of amyotrophic lateral sclerosis and frontotemporal dementia. Hum Mol Genet 2016; 25:3341-3360. [PMID: 27329763 DOI: 10.1093/hmg/ddw182] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2016] [Revised: 06/06/2016] [Accepted: 06/07/2016] [Indexed: 12/11/2022] Open
Abstract
Mutations in the charged multivesicular body protein 2B (CHMP2B) are associated with frontotemporal dementia (FTD), amyotrophic lateral sclerosis (ALS), and with a mixed ALS-FTD syndrome. To model this syndrome, we generated a transgenic mouse line expressing the human CHMP2Bintron5 mutant in a neuron-specific manner. These mice developed a dose-dependent disease phenotype. A longitudinal study revealed progressive gait abnormalities, reduced muscle strength and decreased motor coordination. CHMP2Bintron5 mice died due to generalized paralysis. When paralyzed, signs of denervation were present as attested by altered electromyographic profiles, by decreased number of fully innervated neuromuscular junctions, by reduction in size of motor endplates and by a decrease of sciatic nerve axons area. However, spinal motor neurons cell bodies were preserved until death. In addition to the motor dysfunctions, CHMP2Bintron5 mice progressively developed FTD-relevant behavioural modifications such as disinhibition, stereotypies, decrease in social interactions, compulsivity and change in dietary preferences. Furthermore, neurons in the affected spinal cord and brain regions showed accumulation of p62-positive cytoplasmic inclusions associated or not with ubiquitin and CHMP2Bintron5 As observed in FTD3 patients, these inclusions were negative for TDP-43 and FUS. Moreover, astrogliosis and microgliosis developed with age. Altogether, these data indicate that the neuronal expression of human CHMP2Bintron5 in areas involved in motor and cognitive functions induces progressive motor alterations associated with dementia symptoms and with histopathological hallmarks reminiscent of both ALS and FTD.
Collapse
Affiliation(s)
- Aurélia Vernay
- INSERM, U1118, Mécanismes Centraux et Périphériques de la Neurodégénérescence, F-67000 Strasbourg, France.,Université de Strasbourg, UMRS1118, Faculté de Médecine, Fédération de Médecine Translationelle de Strasbourg, F-67000 Strasbourg, France
| | - Ludivine Therreau
- INSERM, U1118, Mécanismes Centraux et Périphériques de la Neurodégénérescence, F-67000 Strasbourg, France.,Université de Strasbourg, UMRS1118, Faculté de Médecine, Fédération de Médecine Translationelle de Strasbourg, F-67000 Strasbourg, France
| | - Béatrice Blot
- INSERM U836, Grenoble Institut des Neurosciences, Université Joseph Fourier, F-38700 La Tronche, France
| | - Valérie Risson
- Laboratoire de Biologie Moléculaire de la Cellule, UMR5239 CNRS/ENS Lyon/UCBL/HCL Ecole normale supérieure de Lyon, F-69364 Lyon Cedex 07, France
| | - Sylvie Dirrig-Grosch
- INSERM, U1118, Mécanismes Centraux et Périphériques de la Neurodégénérescence, F-67000 Strasbourg, France.,Université de Strasbourg, UMRS1118, Faculté de Médecine, Fédération de Médecine Translationelle de Strasbourg, F-67000 Strasbourg, France
| | - Robin Waegaert
- INSERM, U1118, Mécanismes Centraux et Périphériques de la Neurodégénérescence, F-67000 Strasbourg, France.,Université de Strasbourg, UMRS1118, Faculté de Médecine, Fédération de Médecine Translationelle de Strasbourg, F-67000 Strasbourg, France
| | - Thiebault Lequeu
- INSERM, U1118, Mécanismes Centraux et Périphériques de la Neurodégénérescence, F-67000 Strasbourg, France.,Université de Strasbourg, UMRS1118, Faculté de Médecine, Fédération de Médecine Translationelle de Strasbourg, F-67000 Strasbourg, France
| | - François Sellal
- INSERM, U1118, Mécanismes Centraux et Périphériques de la Neurodégénérescence, F-67000 Strasbourg, France.,Neurology department, Hôpitaux civils and CMRR, F-68000 Colmar, France
| | - Laurent Schaeffer
- Laboratoire de Biologie Moléculaire de la Cellule, UMR5239 CNRS/ENS Lyon/UCBL/HCL Ecole normale supérieure de Lyon, F-69364 Lyon Cedex 07, France
| | - Rémy Sadoul
- INSERM U836, Grenoble Institut des Neurosciences, Université Joseph Fourier, F-38700 La Tronche, France
| | - Jean-Philippe Loeffler
- INSERM, U1118, Mécanismes Centraux et Périphériques de la Neurodégénérescence, F-67000 Strasbourg, France.,Université de Strasbourg, UMRS1118, Faculté de Médecine, Fédération de Médecine Translationelle de Strasbourg, F-67000 Strasbourg, France
| | - Frédérique René
- INSERM, U1118, Mécanismes Centraux et Périphériques de la Neurodégénérescence, F-67000 Strasbourg, France .,Université de Strasbourg, UMRS1118, Faculté de Médecine, Fédération de Médecine Translationelle de Strasbourg, F-67000 Strasbourg, France
| |
Collapse
|
99
|
Morisaki Y, Niikura M, Watanabe M, Onishi K, Tanabe S, Moriwaki Y, Okuda T, Ohara S, Murayama S, Takao M, Uchida S, Yamanaka K, Misawa H. Selective Expression of Osteopontin in ALS-resistant Motor Neurons is a Critical Determinant of Late Phase Neurodegeneration Mediated by Matrix Metalloproteinase-9. Sci Rep 2016; 6:27354. [PMID: 27264390 PMCID: PMC4893611 DOI: 10.1038/srep27354] [Citation(s) in RCA: 49] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2016] [Accepted: 05/17/2016] [Indexed: 12/13/2022] Open
Abstract
Differential vulnerability among motor neuron (MN) subtypes is a fundamental feature of amyotrophic lateral sclerosis (ALS): fast-fatigable (FF) MNs are more vulnerable than fast fatigue-resistant (FR) or slow (S) MNs. The reason for this selective vulnerability remains enigmatic. We report here that the extracellular matrix (ECM) protein osteopontin (OPN) is selectively expressed by FR and S MNs and ALS-resistant motor pools, whereas matrix metalloproteinase-9 (MMP-9) is selectively expressed by FF MNs. OPN is secreted and accumulated as extracellular granules in ECM in three ALS mouse models and a human ALS patient. In SOD1(G93A) mice, OPN/MMP-9 double positivity marks remodeled FR and S MNs destined to compensate for lost FF MNs before ultimately dying. Genetic ablation of OPN in SOD1(G93A) mice delayed disease onset but then accelerated disease progression. OPN induced MMP-9 up-regulation via αvβ3 integrin in ChAT-expressing Neuro2a cells, and also induced CD44-mediated astrocyte migration and microglial phagocytosis in a non-cell-autonomous manner. Our results demonstrate that OPN expressed by FR/S MNs is involved in the second-wave neurodegeneration by up-regulating MMP-9 through αvβ3 integrin in the mouse model of ALS. The differences in OPN/MMP-9 expression profiles in MN subsets partially explain the selective MN vulnerability in ALS.
Collapse
Affiliation(s)
- Yuta Morisaki
- Division of Pharmacology, Faculty of Pharmacy, Keio University, Tokyo 105-8512, Japan
| | - Mamiko Niikura
- Division of Pharmacology, Faculty of Pharmacy, Keio University, Tokyo 105-8512, Japan
| | - Mizuho Watanabe
- Division of Pharmacology, Faculty of Pharmacy, Keio University, Tokyo 105-8512, Japan
| | - Kosuke Onishi
- Division of Pharmacology, Faculty of Pharmacy, Keio University, Tokyo 105-8512, Japan
| | - Shogo Tanabe
- Division of Pharmacology, Faculty of Pharmacy, Keio University, Tokyo 105-8512, Japan
| | - Yasuhiro Moriwaki
- Division of Pharmacology, Faculty of Pharmacy, Keio University, Tokyo 105-8512, Japan
| | - Takashi Okuda
- Division of Pharmacology, Faculty of Pharmacy, Keio University, Tokyo 105-8512, Japan
| | - Shinji Ohara
- Department of Neurology, Matsumoto Medical Center, Chushin-Matsumoto Hospital, Matsumoto 399-0021, Japan
| | - Shigeo Murayama
- Department of Neuropathology, Tokyo Metropolitan Institute of Gerontology, Tokyo 173-0015, Japan
| | - Masaki Takao
- Department of Neuropathology, Tokyo Metropolitan Institute of Gerontology, Tokyo 173-0015, Japan
| | - Sae Uchida
- Department of Autonomic Neuroscience, Tokyo Metropolitan Institute of Gerontology, Tokyo 173-0015, Japan
| | - Koji Yamanaka
- Department of Neuroscience and Pathobiology, Research Institute of Environmental Medicine, Nagoya University, Nagoya 464-8601, Japan
| | - Hidemi Misawa
- Division of Pharmacology, Faculty of Pharmacy, Keio University, Tokyo 105-8512, Japan
| |
Collapse
|
100
|
Jiang Y, Arounleut P, Rheiner S, Bae Y, Kabanov AV, Milligan C, Manickam DS. SOD1 nanozyme with reduced toxicity and MPS accumulation. J Control Release 2016; 231:38-49. [DOI: 10.1016/j.jconrel.2016.02.038] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2015] [Revised: 02/20/2016] [Accepted: 02/24/2016] [Indexed: 01/15/2023]
|