1
|
Henríquez JP, Bermedo-García F, Zelada D, Mella J. Integrating postsynaptic morphology and dynamics to evaluate neuromuscular synapse status: Insights from α-bungarotoxin. Toxicon 2025; 262:108404. [PMID: 40354828 DOI: 10.1016/j.toxicon.2025.108404] [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: 01/18/2025] [Revised: 04/30/2025] [Accepted: 05/09/2025] [Indexed: 05/14/2025]
Abstract
The neuromuscular junction (NMJ) is a crucial peripheral synapse that controls muscle contraction. It consists of a presynaptic motor terminal, a postsynaptic muscle domain, and associated cells, such as terminal Schwann cells and kranocytes. Its larger size compared to central synapses has allowed detailed analyses of NMJ morphology that have been widely used as a reliable parameter of synaptic formation, maturation, function, and decline. Due to its high affinity for postsynaptic acetylcholine receptors (AChRs), the snake venom-derived α-bungarotoxin (BTX) has been pivotal in advancing our understanding of NMJ organization, enabling a detailed mapping of postsynaptic morphologies associated to distinct functional outcomes. Although certain morphological features are often associated with NMJ worsening, some of these cellular changes also occur in biological contexts where synaptic function remains intact. In this review, we draw on previous studies and our recent findings using BTX-based pulse-chase assays to suggest that combining morphological analyses with assessments of postsynaptic stability offers a more comprehensive understanding of NMJ function and regenerative potential. We propose that integrating diverse BTX-based tools into studies of NMJ morphology and stability will provide particularly valuable insights in contexts such as aging, injury, and neuromuscular diseases, where these combined parameters may serve as robust predictors of functional outcomes.
Collapse
Affiliation(s)
- Juan Pablo Henríquez
- Neuromuscular Studies Lab (NeSt Lab), Instituto de Anatomía, Histología y Patología, Facultad de Medicina, Universidad Austral de Chile, 5110566, Valdivia, Chile; Departamento de Biología Celular, Facultad de Ciencias Biológicas, Universidad de Concepción, 4070386, Concepción, Chile.
| | - Francisca Bermedo-García
- Neuromuscular Studies Lab (NeSt Lab), Instituto de Anatomía, Histología y Patología, Facultad de Medicina, Universidad Austral de Chile, 5110566, Valdivia, Chile
| | - Diego Zelada
- Departamento de Biología Celular, Facultad de Ciencias Biológicas, Universidad de Concepción, 4070386, Concepción, Chile
| | - Jessica Mella
- Neuromuscular Studies Lab (NeSt Lab), Instituto de Anatomía, Histología y Patología, Facultad de Medicina, Universidad Austral de Chile, 5110566, Valdivia, Chile
| |
Collapse
|
2
|
Odria R, Cardús A, Gomis-Coloma C, Balanyà-Segura M, Mercado-Amarilla A, Maestre-Mora P, Poveda-Sabuco A, Domingo JC, Nogales-Gadea G, Gomez-Sanchez JA, Hurtado E, Suelves M. HDAC11 deficiency regulates age-related muscle decline and sarcopenia. GeroScience 2025:10.1007/s11357-025-01611-y. [PMID: 40220154 DOI: 10.1007/s11357-025-01611-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2025] [Accepted: 03/09/2025] [Indexed: 04/14/2025] Open
Abstract
Sarcopenia, defined as the progressive loss of skeletal muscle mass and function associated with ageing, has devastating effects in terms of reducing the quality of life of older people. Muscle ageing is characterised by muscle atrophy and decreased capacity for muscle repair, including a reduction in the muscle stem cell pool that impedes recovery after injury. Histone deacetylase 11 (HDAC11) is the newest member of the HDAC family and it is highly expressed in skeletal muscle. Our group recently showed that genetic deficiency in HDAC11 increases skeletal muscle regeneration, mitochondrial function and globally improves muscle performance in young mice. Here, we explore for the first time the functional consequences of HDAC11 deficiency in old mice, in homeostasis and during muscle regeneration. Aged mice lacking HDAC11 show attenuated muscle atrophy and postsynaptic fragmentation of the neuromuscular junction, but no significant differences in the number or diameter of myelinated axons of peripheral nerves. Maintenance of the muscle stem cell reservoir and advanced skeletal muscle regeneration after injury are also observed. HDAC11 depletion enhances mitochondrial fatty acid oxidation and attenuates age-associated alterations in skeletal muscle fatty acid composition, reducing drastically the omega-6/omega-3 fatty acid ratio and improving significantly the omega-3 index, providing an explanation for improved muscle strength and fatigue resistance and decreased mortality. Taken together, our results point to HDAC11 as a new target for the treatment of sarcopenia. Importantly, selective HDAC11 inhibitors have recently been developed that could offer a new therapeutic approach to slow the ageing process.
Collapse
Affiliation(s)
- Renato Odria
- Grup de Recerca en Malaties Neuromusculars de Badalona (GRENBA), Institut d'Investigació en Ciències de la Salut Germans Trias i Pujol (IGTP), 08916, Badalona, Spain
- Programa de Doctorat en Biomedicina, Universitat de Barcelona, 08007, Barcelona, Spain
| | - Aina Cardús
- Grup de Recerca en Malaties Neuromusculars de Badalona (GRENBA), Institut d'Investigació en Ciències de la Salut Germans Trias i Pujol (IGTP), 08916, Badalona, Spain
| | - Clara Gomis-Coloma
- Universidad Católica de Valencia San Vicente Mártir, 46001, Valencia, Spain
| | - Marta Balanyà-Segura
- Universitat Rovira i Virgili, Unitat d'Histologia i Neurobiologia (UHNeurob), Facultat de Medicina i Ciències de la Salut, 43201, Reus, Spain
| | - Alexandra Mercado-Amarilla
- Grup de Recerca en Malaties Neuromusculars de Badalona (GRENBA), Institut d'Investigació en Ciències de la Salut Germans Trias i Pujol (IGTP), 08916, Badalona, Spain
| | - Pau Maestre-Mora
- Grup de Recerca en Malaties Neuromusculars de Badalona (GRENBA), Institut d'Investigació en Ciències de la Salut Germans Trias i Pujol (IGTP), 08916, Badalona, Spain
| | - Andrea Poveda-Sabuco
- Instituto de Investigación Sanitaria y Biomédica de Alicante (ISABIAL), 03010, Alicante, Spain
| | - Joan Carles Domingo
- Department de Bioquímica i Biomedicina Molecular, Facultat de Biologia, Universitat de Barcelona, 08028, Barcelona, Spain
| | - Gisela Nogales-Gadea
- Grup de Recerca en Malaties Neuromusculars de Badalona (GRENBA), Institut d'Investigació en Ciències de la Salut Germans Trias i Pujol (IGTP), 08916, Badalona, Spain
| | - Jose A Gomez-Sanchez
- Instituto de Investigación Sanitaria y Biomédica de Alicante (ISABIAL), 03010, Alicante, Spain
- Instituto de Neurociencias de Alicante UMH-CSIC, 03550, San Juan de Alicante, Spain
| | - Erica Hurtado
- Grup de Recerca en Malaties Neuromusculars de Badalona (GRENBA), Institut d'Investigació en Ciències de la Salut Germans Trias i Pujol (IGTP), 08916, Badalona, Spain.
- Universitat Rovira i Virgili, Unitat d'Histologia i Neurobiologia (UHNeurob), Facultat de Medicina i Ciències de la Salut, 43201, Reus, Spain.
| | - Mònica Suelves
- Grup de Recerca en Malaties Neuromusculars de Badalona (GRENBA), Institut d'Investigació en Ciències de la Salut Germans Trias i Pujol (IGTP), 08916, Badalona, Spain.
| |
Collapse
|
3
|
Fish LA, Ewing MD, Rich KA, Xi C, Chen I, Jaime D, Madigan LA, Wang X, Shahtout JL, Feder RE, Funai K, Christian JL, Wharton KA, Rich MM, Arnold WD, Fallon JR. MuSK Regulates Neuromuscular Junction Nav1.4 Localization and Excitability. J Neurosci 2025; 45:e1279232025. [PMID: 39880682 PMCID: PMC11984086 DOI: 10.1523/jneurosci.1279-23.2025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2023] [Revised: 12/16/2024] [Accepted: 01/18/2025] [Indexed: 01/31/2025] Open
Abstract
The neuromuscular junction (NMJ) is the linchpin of nerve-evoked muscle contraction. Broadly, the NMJ transduces nerve action potentials into muscle fiber action potentials (MFAPs). Efficient neuromuscular transmission requires cholinergic signaling, which generates endplate potentials (EPPs), and excitation, the amplification of an EPP by postsynaptic voltage-gated sodium channels (Nav1.4) to generate the MFAP. Compared to the cholinergic component, the signaling pathways that organize Nav1.4 are poorly characterized. Muscle-specific kinase (MuSK), in addition to its Ig1 domain-dependent role as the main organizer of acetylcholine receptors (AChRs), also binds BMPs via its Ig3 domain and shapes BMP-induced signaling. Using mice lacking the MuSK Ig3 domain ("ΔIg3-MuSK"), we probed the role of this domain at the NMJ. NMJs formed in ΔIg3-MuSK animals with pre- and postsynaptic specializations aligned at all ages examined. However, the ΔIg3-MuSK postsynaptic apparatus was fragmented from an early age. Synaptic electrophysiology showed that spontaneous and nerve-evoked acetylcholine release, AChR density, and endplate currents were comparable at WT and ΔIg3-MuSK NMJs. However, single fiber electromyography revealed that nerve-evoked MFAPs in ΔIg3-MuSK muscle were abnormal, exhibiting jitter and blocking. Nerve-evoked compound muscle action potentials and muscle force were also diminished. Finally, Nav1.4 levels were reduced at ΔIg3-MuSK NMJs, but not extrasynaptically, indicating that the observed excitability defects result from impaired synaptic localization of this ion channel. We propose distinct, domain-specific roles for MuSK at the NMJ: the Ig1 domain mediates agrin-LRP4-mediated AChR localization, while the Ig3 domain maintains postsynaptic Nav1.4 density, conferring the muscle excitability required to amplify cholinergic signals and trigger action potentials.
Collapse
Affiliation(s)
- Lauren A Fish
- Neuroscience Graduate Program, Brown University, Providence, Rhode Island 02912
- Robert J. and Nancy D. Carney Institute for Brain Science, Brown University, Providence, Rhode Island 02912
| | - Madison D Ewing
- Department of Neuroscience, Brown University, Providence, Rhode Island 02912
| | - Kelly A Rich
- Neuroscience Graduate Program, Ohio State University, Columbus, Ohio 43210
| | - Chengjie Xi
- Biotechnology Graduate Program, Brown University, Brown University, Providence, Rhode Island 02912
| | - Isabella Chen
- Department of Neuroscience, Brown University, Providence, Rhode Island 02912
| | - Diego Jaime
- Department of Molecular Biology, Cell Biology, and Biochemistry, Brown University, Providence, Rhode Island 02912
| | - Laura A Madigan
- Department of Molecular Biology, Cell Biology, and Biochemistry, Brown University, Providence, Rhode Island 02912
| | - Xueyong Wang
- Department of Neuroscience, Cell Biology, and Physiology, Wright State University, Dayton, Ohio 45435
| | - Justin L Shahtout
- Diabetes & Metabolism Research Center, University of Utah, Salt Lake City, Utah 84112
| | - Rita E Feder
- Department of Neuroscience, Brown University, Providence, Rhode Island 02912
| | - Katsuhiko Funai
- Diabetes & Metabolism Research Center, University of Utah, Salt Lake City, Utah 84112
| | - Jan L Christian
- Department of Neurobiology, University of Utah, Salt Lake City, Utah 84112
| | - Kristi A Wharton
- Department of Molecular Biology, Cell Biology, and Biochemistry, Brown University, Providence, Rhode Island 02912
| | - Mark M Rich
- Department of Neuroscience, Cell Biology, and Physiology, Wright State University, Dayton, Ohio 45435
| | - William D Arnold
- NextGen Precision Health Institute, University of Missouri, Columbia, Missouri 65211
- Department of Physical Medicine and Rehabilitation, University of Missouri, Columbia, Missouri 65212
- Department of Neurology, Neuromuscular Division, Wexner Medical Center, The Ohio State University, Columbus, Ohio 43210
| | - Justin R Fallon
- Robert J. and Nancy D. Carney Institute for Brain Science, Brown University, Providence, Rhode Island 02912
- Department of Neuroscience, Brown University, Providence, Rhode Island 02912
| |
Collapse
|
4
|
Belančić A, Janković T, Gkrinia EMM, Kristić I, Rajič Bumber J, Rački V, Pilipović K, Vitezić D, Mršić-Pelčić J. Glial Cells in Spinal Muscular Atrophy: Speculations on Non-Cell-Autonomous Mechanisms and Therapeutic Implications. Neurol Int 2025; 17:41. [PMID: 40137462 PMCID: PMC11944370 DOI: 10.3390/neurolint17030041] [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: 02/09/2025] [Revised: 03/07/2025] [Accepted: 03/11/2025] [Indexed: 03/29/2025] Open
Abstract
Spinal muscular atrophy (SMA) is a neuromuscular disorder caused by homozygous deletions or mutations in the SMN1 gene, leading to progressive motor neuron degeneration. While SMA has been classically viewed as a motor neuron-autonomous disease, increasing evidence indicates a significant role of glial cells-astrocytes, microglia, oligodendrocytes, and Schwann cells-in the disease pathophysiology. Astrocytic dysfunction contributes to motor neuron vulnerability through impaired calcium homeostasis, disrupted synaptic integrity, and neurotrophic factor deficits. Microglia, through reactive gliosis and complement-mediated synaptic stripping, exacerbate neurodegeneration and neuroinflammation. Oligodendrocytes exhibit impaired differentiation and metabolic support, while Schwann cells display abnormalities in myelination, extracellular matrix composition, and neuromuscular junction maintenance, further compromising motor function. Dysregulation of pathways such as NF-κB, Notch, and JAK/STAT, alongside the upregulation of complement proteins and microRNAs, reinforces the non-cell-autonomous nature of SMA. Despite the advances in SMN-restorative therapies, they do not fully mitigate glial dysfunction. Targeting glial pathology, including modulation of reactive astrogliosis, microglial polarization, and myelination deficits, represents a critical avenue for therapeutic intervention. This review comprehensively examines the multifaceted roles of glial cells in SMA and highlights emerging glia-targeted strategies to enhance treatment efficacy and improve patient outcomes.
Collapse
Affiliation(s)
- Andrej Belančić
- Department of Basic and Clinical Pharmacology and Toxicology, Faculty of Medicine, University of Rijeka, Braće Branchetta 20, 51000 Rijeka, Croatia; (T.J.); (I.K.); (J.R.B.); (K.P.); (D.V.); (J.M.-P.)
| | - Tamara Janković
- Department of Basic and Clinical Pharmacology and Toxicology, Faculty of Medicine, University of Rijeka, Braće Branchetta 20, 51000 Rijeka, Croatia; (T.J.); (I.K.); (J.R.B.); (K.P.); (D.V.); (J.M.-P.)
| | | | - Iva Kristić
- Department of Basic and Clinical Pharmacology and Toxicology, Faculty of Medicine, University of Rijeka, Braće Branchetta 20, 51000 Rijeka, Croatia; (T.J.); (I.K.); (J.R.B.); (K.P.); (D.V.); (J.M.-P.)
| | - Jelena Rajič Bumber
- Department of Basic and Clinical Pharmacology and Toxicology, Faculty of Medicine, University of Rijeka, Braće Branchetta 20, 51000 Rijeka, Croatia; (T.J.); (I.K.); (J.R.B.); (K.P.); (D.V.); (J.M.-P.)
| | - Valentino Rački
- Department of Neurology, Clinical Hospital Centre Rijeka, Krešimirova 42, 51000 Rijeka, Croatia;
| | - Kristina Pilipović
- Department of Basic and Clinical Pharmacology and Toxicology, Faculty of Medicine, University of Rijeka, Braće Branchetta 20, 51000 Rijeka, Croatia; (T.J.); (I.K.); (J.R.B.); (K.P.); (D.V.); (J.M.-P.)
| | - Dinko Vitezić
- Department of Basic and Clinical Pharmacology and Toxicology, Faculty of Medicine, University of Rijeka, Braće Branchetta 20, 51000 Rijeka, Croatia; (T.J.); (I.K.); (J.R.B.); (K.P.); (D.V.); (J.M.-P.)
| | - Jasenka Mršić-Pelčić
- Department of Basic and Clinical Pharmacology and Toxicology, Faculty of Medicine, University of Rijeka, Braće Branchetta 20, 51000 Rijeka, Croatia; (T.J.); (I.K.); (J.R.B.); (K.P.); (D.V.); (J.M.-P.)
| |
Collapse
|
5
|
Gould TW, Ko CP, Willison H, Robitaille R. Perisynaptic Schwann Cells: Guardians of Neuromuscular Junction Integrity and Function in Health and Disease. Cold Spring Harb Perspect Biol 2025; 17:a041362. [PMID: 38858074 PMCID: PMC11694759 DOI: 10.1101/cshperspect.a041362] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/12/2024]
Abstract
The neuromuscular junction (NMJ) is a highly reliable synapse to carry the control of the motor commands of the nervous system over the muscles. Its development, organization, and synaptic properties are highly structured and regulated to support such reliability and efficacy. Yet, the NMJ is also highly plastic, able to react to injury, and able to adapt to changes. This balance between structural stability and synaptic efficacy on one hand and structural plasticity and repair on another hand is made possible by perisynaptic Schwann cells (PSCs), glial cells at this synapse. They regulate synaptic efficacy and structural plasticity of the NMJ in a dynamic, bidirectional manner owing to their ability to decode synaptic transmission and by their interactions with trophic-related factors. Alteration of these fundamental roles of PSCs is also important in the maladapted response of NMJs in various diseases and in aging.
Collapse
Affiliation(s)
- Thomas W Gould
- Department of Physiology and Cell Biology, University of Nevada, Reno School of Medicine, Reno, Nevada 89557, USA
| | - Chien-Ping Ko
- Section of Neurobiology, Department of Biological Sciences, University of Southern California, Los Angeles, California 90089-2520, USA
| | - Hugh Willison
- School of Infection and Immunity, University of Glasgow, Glasgow G12 8TA, Scotland
| | - Richard Robitaille
- Département de neurosciences, Université de Montréal, Montréal, Québec H3C 3J7, Canada
- Centre Interdisciplinaire de Recherche sur le Cerveau et l'apprentissage, Université de Montréal, Montréal, Québec H3C 3J7, Canada
| |
Collapse
|
6
|
Petrov K, Lenina O, Leroy J, Bernard V, Germain T, Truong C, Nurullin L, Sibgatullina G, Ohno K, Samigullin D, Krejci E. An α7 nicotinic and GABA B receptor-mediated pathway controls acetylcholine release in the tripartite neuromuscular junction. J Physiol 2025; 603:507-527. [PMID: 39740234 PMCID: PMC11737540 DOI: 10.1113/jp287243] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2024] [Accepted: 11/13/2024] [Indexed: 01/02/2025] Open
Abstract
Terminal Schwann cells (TSCs) are capable of regulating acetylcholine (ACh) release at the neuromuscular junction (NMJ). We have identified GABA as a gliotransmitter at mouse NMJs. When ACh activates α7 nicotinic ACh receptor (nAChRs) on TSCs, GABA is released and activates GABAB receptors on the nerve terminal that subsequently reduce ACh release. Indeed, specific deletion of the α7 nAChR in TSCs or inhibition of the metabotropic GABAB receptor prevents the reduction in the quantal content of the end-plate potential induced by cholinesterase inhibitors. The α7/GABAB receptor-mediated pathway is activated when ACh that escapes from collagen Q (ColQ) anchored AChE in the synaptic cleft and from PRiMA-anchored butyrylcholinesterase on the TSC activates α7 nAChRs on the TSC. Consequently, prolonged tetanic stimulation of isolated muscle activates the α7/GABAB receptor pathway, which reduces post-tetanic ACh release. When AChE levels are low in neonatal mice, the α7/GABAB receptor-mediated pathway decreases ACh release and reduces ex vivo muscle fatigue. For ColQ-deficient mice where AChE is not clustered, the decrease in AСh release following activation of this pathway contributes to mouse fatigue in vivo. KEY POINTS: Acetylcholine (ACh) released from the nerve terminal at the neuromuscular junction (NMJ) can activate α7 nicotinic ACh receptor (nAChR) on terminal Schwann cells, releasing gamma-aminobutyric acid (GABA) that activates metabotropic GABAB receptors on the nerve terminal which then reduces further ACh release from the nerve. At the mature NMJ, before reaching α7 nAChRs on terminal Schwann cells ACh is normally hydrolyzed by AChE clustered in the synaptic cleft and by BChE anchored to the TSC. ACh can activate the α7/GABAB receptor-mediated pathway and depress subsequent ACh release when AChE at the NMJ is low, either during development or in congenital myasthenic syndrome. In the latter case, this pathway contributes to muscle fatigue.
Collapse
Affiliation(s)
- Konstantin Petrov
- Arbuzov Institute of Organic and Physical ChemistryFRC Kazan Scientific Center of RASKazanRussia
- Kazan Institute of Biochemistry and BiophysicsFRC Kazan Scientific Center of RASKazanRussia
| | - Oksana Lenina
- Arbuzov Institute of Organic and Physical ChemistryFRC Kazan Scientific Center of RASKazanRussia
| | - Jacqueline Leroy
- Université Paris Cité, CNRS, ENS Paris SaclayCentre Borelli UMR 9010ParisFrance
| | | | - Thibaut Germain
- Université Paris Saclay, CNRS, ENS Paris Saclay, Centre Borelli UMR 9010Gif sur YvetteFrance
| | - Charles Truong
- Université Paris Saclay, CNRS, ENS Paris Saclay, Centre Borelli UMR 9010Gif sur YvetteFrance
| | - Leniz Nurullin
- Kazan Institute of Biochemistry and BiophysicsFRC Kazan Scientific Center of RASKazanRussia
- Kazan State Medical UniversityKazanRussia
| | - Guzel Sibgatullina
- Kazan Institute of Biochemistry and BiophysicsFRC Kazan Scientific Center of RASKazanRussia
| | - Kinji Ohno
- Graduate School of Nutritional SciencesNagoya University of Arts and SciencesNisshinJapan
| | - Dmitry Samigullin
- Kazan Institute of Biochemistry and BiophysicsFRC Kazan Scientific Center of RASKazanRussia
- Department of Radiophotonics and Microwave TechnologiesKazan National Research Technical University Named after A.N. Tupolev‐KAIKazanRussia
| | - Eric Krejci
- Université Paris Cité, CNRS, ENS Paris SaclayCentre Borelli UMR 9010ParisFrance
| |
Collapse
|
7
|
Hirono T, Takeda R, Nishikawa T, Ueda S, Akamatsu Y, Uchida K, Mita Y, Watanabe K. Age-related decline of motor unit firing rate in community-dwelling healthy older adults and daily ingestion of fish protein of Alaska pollack: a randomised trial. Int J Food Sci Nutr 2024; 75:846-854. [PMID: 39370733 DOI: 10.1080/09637486.2024.2412035] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2023] [Revised: 09/03/2024] [Accepted: 09/29/2024] [Indexed: 10/08/2024]
Abstract
Neuromuscular properties decline with ageing and low-level physical activities. Alaska pollack protein (APP) is reportedly effective for improving skeletal muscular functions, even if the amount is small and exercise is not conducted. However, it is unclear whether APP intake without an exercise program affects neuromuscular dysfunction in community-dwelling adults conducting normal, everyday activities. This study aimed to investigate changes in motor unit firing properties by adding APP to daily meals in community-dwelling older adults. They were divided into an APP group or a placebo control (CON) group for randomised, double-blind treatment. Participants in APP (n = 15) and CON (n = 17) groups ingested 4.5 g/d APP and whey protein, respectively, added to daily meals for 3 months without a specific exercise intervention. High-density surface electromyography of the vastus lateralis to assess the individual motor unit firing rate during 70% of maximum voluntary ramp-up contraction (MVC), maximum knee extensor strength, muscle thickness, echo intensity, and a physical function test were evaluated at the baseline, as well as 1.5 and 3 months after the intervention. While muscle strength, muscle thickness, and motor function were not changed, the motor unit firing rate at 50%-70% of MVC showed a significant decline at 1.5 and 3 months in the CON group, but this decline was not observed in the APP group. These findings suggest that neuromuscular properties show a detectable decline on performing daily activities without a specific exercise intervention, but such a decline was not observed in the APP intake group.
Collapse
Affiliation(s)
- Tetsuya Hirono
- Laboratory of Neuromuscular Biomechanics, School of Health and Sport Science, Chukyo University, Toyota, Japan
- Research Fellow of Japan Society for the Promotion of Science, Tokyo, Japan
- Human Health Sciences, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Ryosuke Takeda
- Laboratory of Neuromuscular Biomechanics, School of Health and Sport Science, Chukyo University, Toyota, Japan
| | - Taichi Nishikawa
- Laboratory of Neuromuscular Biomechanics, School of Health and Sport Science, Chukyo University, Toyota, Japan
| | - Saeko Ueda
- Laboratory of Neuromuscular Biomechanics, School of Health and Sport Science, Chukyo University, Toyota, Japan
- Department of Human Nutrition, School of Life Studies, Sugiyama Jogakuen University, Nagoya, Japan
| | | | | | - Yukiko Mita
- Department of Human Nutrition, School of Life Studies, Sugiyama Jogakuen University, Nagoya, Japan
| | - Kohei Watanabe
- Laboratory of Neuromuscular Biomechanics, School of Health and Sport Science, Chukyo University, Toyota, Japan
| |
Collapse
|
8
|
Traoré M, Noviello C, Vergnol A, Gentil C, Halliez M, Saillard L, Gelin M, Forand A, Lemaitre M, Guesmia Z, Cadot B, Caldas de Almeida Araujo E, Marty B, Mougenot N, Messéant J, Strochlic L, Sadoine J, Slimani L, Jolly A, De la Grange P, Hogrel JY, Pietri-Rouxel F, Falcone S. GDF5 as a rejuvenating treatment for age-related neuromuscular failure. Brain 2024; 147:3834-3848. [PMID: 38584513 DOI: 10.1093/brain/awae107] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2023] [Revised: 03/08/2024] [Accepted: 03/23/2024] [Indexed: 04/09/2024] Open
Abstract
Sarcopenia involves a progressive loss of skeletal muscle force, quality and mass during ageing, which results in increased inability and death; however, no cure has been established thus far. Growth differentiation factor 5 (GDF5) has been described to modulate muscle mass maintenance in various contexts. For our proof of concept, we overexpressed GDF5 by AAV vector injection in tibialis anterior muscle of adult aged (20 months) mice and performed molecular and functional analysis of skeletal muscle. We analysed human vastus lateralis muscle biopsies from adult young (21-42 years) and aged (77-80 years) donors, quantifying the molecular markers modified by GDF5 overexpression in mouse muscle. We validated the major effects of GDF5 overexpression using human immortalized myotubes and Schwann cells. We established a preclinical study by treating chronically (for 4 months) aged mice using recombinant GDF5 protein (rGDF5) in systemic administration and evaluated the long-term effect of this treatment on muscle mass and function. Here, we demonstrated that GDF5 overexpression in the old tibialis anterior muscle promoted an increase of 16.5% of muscle weight (P = 0.0471) associated with a higher percentage of 5000-6000 µm2 large fibres (P = 0.0211), without the induction of muscle regeneration. Muscle mass gain was associated with an amelioration of 26.8% of rate of force generation (P = 0.0330) and better neuromuscular connectivity (P = 0.0098). Moreover, GDF5 overexpression preserved neuromuscular junction morphology (38.5% of nerve terminal area increase, P < 0.0001) and stimulated the expression of reinnervation-related genes, in particular markers of Schwann cells (fold-change 3.19 for S100b gene expression, P = 0.0101). To characterize the molecular events induced by GDF5 overexpression during ageing, we performed a genome-wide transcriptomic analysis of treated muscles and showed that this factor leads to a 'rejuvenating' transcriptomic signature in aged mice, as 42% of the transcripts dysregulated by ageing reverted to youthful expression levels upon GDF5 overexpression (P < 0.05). Towards a preclinical approach, we performed a long-term systemic treatment using rGDF5 and showed its effectiveness in counteracting age-related muscle wasting, improving muscle function (17.8% of absolute maximal force increase, P = 0.0079), ensuring neuromuscular connectivity and preventing neuromuscular junction degeneration (7.96% of AchR area increase, P = 0.0125). In addition, in human muscle biopsies, we found the same age-related alterations than those observed in mice and improved by GDF5 and reproduced its major effects on human cells, suggesting this treatment as efficient in humans. Overall, these data provide a foundation to examine the curative potential of GDF5 drug in clinical trials for sarcopenia and, eventually, other neuromuscular diseases.
Collapse
Affiliation(s)
- Massiré Traoré
- Sorbonne Université, INSERM, Institut de Myologie, Centre de Recherche en Myologie, F-75013 Paris, France
| | - Chiara Noviello
- Sorbonne Université, INSERM, Institut de Myologie, Centre de Recherche en Myologie, F-75013 Paris, France
| | - Amélie Vergnol
- Sorbonne Université, INSERM, Institut de Myologie, Centre de Recherche en Myologie, F-75013 Paris, France
| | - Christel Gentil
- Sorbonne Université, INSERM, Institut de Myologie, Centre de Recherche en Myologie, F-75013 Paris, France
| | - Marius Halliez
- Sorbonne Université, INSERM, Institut de Myologie, Centre de Recherche en Myologie, F-75013 Paris, France
| | - Lucile Saillard
- Sorbonne Université, INSERM, Institut de Myologie, Centre de Recherche en Myologie, F-75013 Paris, France
| | - Maxime Gelin
- Sorbonne Université, INSERM, Institut de Myologie, Centre de Recherche en Myologie, F-75013 Paris, France
| | - Anne Forand
- Sorbonne Université, INSERM, Institut de Myologie, Centre de Recherche en Myologie, F-75013 Paris, France
- Inovarion, F-75005 Paris, France
| | - Mégane Lemaitre
- Sorbonne Université, INSERM UMS28, Phénotypage du Petit Animal, 75013 Paris, France
| | - Zoheir Guesmia
- Sorbonne Université, INSERM, Institut de Myologie, Centre de Recherche en Myologie, F-75013 Paris, France
| | - Bruno Cadot
- Sorbonne Université, INSERM, Institut de Myologie, Centre de Recherche en Myologie, F-75013 Paris, France
| | | | - Benjamin Marty
- Institut de Myologie, CEA, Laboratoire d'imagerie et de spectroscopie par RMN, F-75013 Paris, France
| | - Nathalie Mougenot
- Sorbonne Université, INSERM UMS28, Phénotypage du Petit Animal, 75013 Paris, France
| | - Julien Messéant
- Sorbonne Université, INSERM, Institut de Myologie, Centre de Recherche en Myologie, F-75013 Paris, France
| | - Laure Strochlic
- Sorbonne Université, INSERM, Institut de Myologie, Centre de Recherche en Myologie, F-75013 Paris, France
| | - Jeremy Sadoine
- Université de Paris, Plateforme d'Imagerie du Vivant (PIV), F-92120 Montrouge, France
| | - Lofti Slimani
- Université de Paris, Plateforme d'Imagerie du Vivant (PIV), F-92120 Montrouge, France
| | - Ariane Jolly
- GenoSplice, Paris Biotech Santé, F-75014 Paris, France
| | | | - Jean-Yves Hogrel
- Institut de Myologie, Laboratoire de physiologie et d'évaluation neuromusculaire, F-75013 Paris, France
| | - France Pietri-Rouxel
- Sorbonne Université, INSERM, Institut de Myologie, Centre de Recherche en Myologie, F-75013 Paris, France
| | - Sestina Falcone
- Sorbonne Université, INSERM, Institut de Myologie, Centre de Recherche en Myologie, F-75013 Paris, France
| |
Collapse
|
9
|
Hastings RL, Valdez G. Origin, identity, and function of terminal Schwann cells. Trends Neurosci 2024; 47:432-446. [PMID: 38664109 PMCID: PMC11168889 DOI: 10.1016/j.tins.2024.03.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2023] [Revised: 02/20/2024] [Accepted: 03/26/2024] [Indexed: 06/14/2024]
Abstract
The highly specialized nonmyelinating glial cells present at somatic peripheral nerve endings, known collectively as terminal Schwann cells (TSCs), play critical roles in the development, function and repair of their motor and sensory axon terminals and innervating tissue. Over the past decades, research efforts across various vertebrate species have revealed that while TSCs are a diverse group of cells, they share a number of features among them. In this review, we summarize the state-of-knowledge about each TSC type and explore the opportunities that TSCs provide to treat conditions that afflict peripheral axon terminals.
Collapse
Affiliation(s)
- Robert Louis Hastings
- Department of Molecular Biology, Cell Biology and Biochemistry, Brown University, Providence, RI, USA
| | - Gregorio Valdez
- Department of Molecular Biology, Cell Biology and Biochemistry, Brown University, Providence, RI, USA; Center for Translational Neuroscience, Robert J. and Nancy D. Carney Institute for Brain Science, and Center on the Biology of Aging, Brown University, Providence, RI, USA.
| |
Collapse
|
10
|
Tao X, Zhu Z, Wang L, Li C, Sun L, Wang W, Gong W. Biomarkers of Aging and Relevant Evaluation Techniques: A Comprehensive Review. Aging Dis 2024; 15:977-1005. [PMID: 37611906 PMCID: PMC11081160 DOI: 10.14336/ad.2023.00808-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2023] [Accepted: 08/08/2023] [Indexed: 08/25/2023] Open
Abstract
The risk of developing chronic illnesses and disabilities is increasing with age. To predict and prevent aging, biomarkers relevant to the aging process must be identified. This paper reviews the known molecular, cellular, and physiological biomarkers of aging. Moreover, we discuss the currently available technologies for identifying these biomarkers, and their applications and potential in aging research. We hope that this review will stimulate further research and innovation in this emerging and fast-growing field.
Collapse
Affiliation(s)
- Xue Tao
- Department of Research, Beijing Rehabilitation Hospital, Capital Medical University, Beijing, China.
| | - Ziman Zhu
- Beijing Rehabilitation Medicine Academy, Capital Medical University, Beijing, China.
| | - Liguo Wang
- Key Laboratory of Protein Sciences, School of Pharmaceutical Sciences, Tsinghua University, Beijing, China.
| | - Chunlin Li
- School of Biomedical Engineering, Capital Medical University, Beijing, China.
- Beijing Key Laboratory of Fundamental Research on Biomechanics in Clinical Application, Capital Medical University, Beijing, China.
| | - Liwei Sun
- School of Biomedical Engineering, Capital Medical University, Beijing, China.
- Beijing Key Laboratory of Fundamental Research on Biomechanics in Clinical Application, Capital Medical University, Beijing, China.
| | - Wei Wang
- Department of Rehabilitation Radiology, Beijing Rehabilitation Hospital, Capital Medical University, Beijing, China.
| | - Weijun Gong
- Department of Neurological Rehabilitation, Beijing Rehabilitation Hospital, Capital Medical University, Beijing, China.
| |
Collapse
|
11
|
Kedlian VR, Wang Y, Liu T, Chen X, Bolt L, Tudor C, Shen Z, Fasouli ES, Prigmore E, Kleshchevnikov V, Pett JP, Li T, Lawrence JEG, Perera S, Prete M, Huang N, Guo Q, Zeng X, Yang L, Polański K, Chipampe NJ, Dabrowska M, Li X, Bayraktar OA, Patel M, Kumasaka N, Mahbubani KT, Xiang AP, Meyer KB, Saeb-Parsy K, Teichmann SA, Zhang H. Human skeletal muscle aging atlas. NATURE AGING 2024; 4:727-744. [PMID: 38622407 PMCID: PMC11108788 DOI: 10.1038/s43587-024-00613-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/20/2023] [Accepted: 03/19/2024] [Indexed: 04/17/2024]
Abstract
Skeletal muscle aging is a key contributor to age-related frailty and sarcopenia with substantial implications for global health. Here we profiled 90,902 single cells and 92,259 single nuclei from 17 donors to map the aging process in the adult human intercostal muscle, identifying cellular changes in each muscle compartment. We found that distinct subsets of muscle stem cells exhibit decreased ribosome biogenesis genes and increased CCL2 expression, causing different aging phenotypes. Our atlas also highlights an expansion of nuclei associated with the neuromuscular junction, which may reflect re-innervation, and outlines how the loss of fast-twitch myofibers is mitigated through regeneration and upregulation of fast-type markers in slow-twitch myofibers with age. Furthermore, we document the function of aging muscle microenvironment in immune cell attraction. Overall, we present a comprehensive human skeletal muscle aging resource ( https://www.muscleageingcellatlas.org/ ) together with an in-house mouse muscle atlas to study common features of muscle aging across species.
Collapse
Affiliation(s)
- Veronika R Kedlian
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, UK
| | - Yaning Wang
- Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
- Advanced Medical Technology Center, The First Affiliated Hospital, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Tianliang Liu
- Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
- Advanced Medical Technology Center, The First Affiliated Hospital, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Xiaoping Chen
- Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Liam Bolt
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, UK
| | - Catherine Tudor
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, UK
| | - Zhuojian Shen
- Department of Thoracic Surgery, Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital of Sun Yat-sen University, Guangzhou, China
| | - Eirini S Fasouli
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, UK
| | - Elena Prigmore
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, UK
| | | | - Jan Patrick Pett
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, UK
| | - Tong Li
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, UK
| | - John E G Lawrence
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, UK
| | - Shani Perera
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, UK
| | - Martin Prete
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, UK
| | - Ni Huang
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, UK
| | - Qin Guo
- Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Xinrui Zeng
- Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
- Advanced Medical Technology Center, The First Affiliated Hospital, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Lu Yang
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, UK
| | - Krzysztof Polański
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, UK
| | - Nana-Jane Chipampe
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, UK
| | - Monika Dabrowska
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, UK
| | - Xiaobo Li
- Core Facilities for Medical Science, Sun Yat-sen University, Guangzhou, China
| | - Omer Ali Bayraktar
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, UK
| | - Minal Patel
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, UK
| | - Natsuhiko Kumasaka
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, UK
| | - Krishnaa T Mahbubani
- Department of Surgery, University of Cambridge, Cambridge, UK
- Collaborative Biorepository for Translational Medicine (CBTM), NIHR Cambridge Biomedical Research Centre, Cambridge, UK
| | - Andy Peng Xiang
- Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Kerstin B Meyer
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, UK
| | - Kourosh Saeb-Parsy
- Department of Surgery, University of Cambridge, Cambridge, UK.
- Collaborative Biorepository for Translational Medicine (CBTM), NIHR Cambridge Biomedical Research Centre, Cambridge, UK.
| | - Sarah A Teichmann
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, UK.
- Cavendish Laboratory, University of Cambridge, Cambridge, UK.
| | - Hongbo Zhang
- Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China.
- Advanced Medical Technology Center, The First Affiliated Hospital, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China.
| |
Collapse
|
12
|
Willows JW, Alshahal Z, Story NM, Alves MJ, Vidal P, Harris H, Rodrigo R, Stanford KI, Peng J, Reifsnyder PC, Harrison DE, David Arnold W, Townsend KL. Contributions of mouse genetic strain background to age-related phenotypes in physically active HET3 mice. Neurobiol Aging 2024; 136:58-69. [PMID: 38325031 DOI: 10.1016/j.neurobiolaging.2024.01.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2023] [Revised: 01/06/2024] [Accepted: 01/22/2024] [Indexed: 02/09/2024]
Abstract
We assessed aging hallmarks in skin, muscle, and adipose in the genetically diverse HET3 mouse, and generated a broad dataset comparing these to individual animal diagnostic SNPs from the 4 founding inbred strains of the HET3 line. For middle- and old-aged HET3 mice, we provided running wheel exercise to ensure our observations were not purely representative of sedentary animals, but age-related phenotypes were not improved with running wheel activity. Adipose tissue fibrosis, peripheral neuropathy, and loss of neuromuscular junction integrity were consistent phenotypes in older-aged HET3 mice regardless of physical activity, but aspects of these phenotypes were moderated by the SNP% contributions of the founding strains for the HET3 line. Taken together, the genetic contribution of founder strain SNPs moderated age-related phenotypes in skin and muscle innervation and were dependent on biological sex and chronological age. However, there was not a single founder strain (BALB/cJ, C57BL/6J, C3H/HeJ, DBA/2J) that appeared to drive more protection or disease-risk across aging in this mouse line, but genetic diversity in general was more protective.
Collapse
Affiliation(s)
- Jake W Willows
- Department of Neurological Surgery, The Ohio State University, Columbus, OH, USA
| | - Zahra Alshahal
- Department of Neurological Surgery, The Ohio State University, Columbus, OH, USA
| | - Naeemah M Story
- Department of Neurological Surgery, The Ohio State University, Columbus, OH, USA
| | - Michele J Alves
- Department of Neurological Surgery, The Ohio State University, Columbus, OH, USA
| | - Pablo Vidal
- Department of Physiology and Cell Biology, The Ohio State University, Columbus, OH, USA
| | - Hallie Harris
- Department of Neurology, The Ohio State University, Columbus, OH, USA
| | - Rochelle Rodrigo
- Department of Neurology, The Ohio State University, Columbus, OH, USA
| | - Kristin I Stanford
- Department of Physiology and Cell Biology, The Ohio State University, Columbus, OH, USA
| | - Juan Peng
- Department of Biomedical Informatics, The Ohio State University, Columbus, OH, USA
| | | | | | - W David Arnold
- Department of Neurology, The Ohio State University, Columbus, OH, USA
| | - Kristy L Townsend
- Department of Neurological Surgery, The Ohio State University, Columbus, OH, USA.
| |
Collapse
|
13
|
Jagaraj CJ, Shadfar S, Kashani SA, Saravanabavan S, Farzana F, Atkin JD. Molecular hallmarks of ageing in amyotrophic lateral sclerosis. Cell Mol Life Sci 2024; 81:111. [PMID: 38430277 PMCID: PMC10908642 DOI: 10.1007/s00018-024-05164-9] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Revised: 01/21/2024] [Accepted: 02/06/2024] [Indexed: 03/03/2024]
Abstract
Amyotrophic lateral sclerosis (ALS) is a fatal, severely debilitating and rapidly progressing disorder affecting motor neurons in the brain, brainstem, and spinal cord. Unfortunately, there are few effective treatments, thus there remains a critical need to find novel interventions that can mitigate against its effects. Whilst the aetiology of ALS remains unclear, ageing is the major risk factor. Ageing is a slowly progressive process marked by functional decline of an organism over its lifespan. However, it remains unclear how ageing promotes the risk of ALS. At the molecular and cellular level there are specific hallmarks characteristic of normal ageing. These hallmarks are highly inter-related and overlap significantly with each other. Moreover, whilst ageing is a normal process, there are striking similarities at the molecular level between these factors and neurodegeneration in ALS. Nine ageing hallmarks were originally proposed: genomic instability, loss of telomeres, senescence, epigenetic modifications, dysregulated nutrient sensing, loss of proteostasis, mitochondrial dysfunction, stem cell exhaustion, and altered inter-cellular communication. However, these were recently (2023) expanded to include dysregulation of autophagy, inflammation and dysbiosis. Hence, given the latest updates to these hallmarks, and their close association to disease processes in ALS, a new examination of their relationship to pathophysiology is warranted. In this review, we describe possible mechanisms by which normal ageing impacts on neurodegenerative mechanisms implicated in ALS, and new therapeutic interventions that may arise from this.
Collapse
Affiliation(s)
- Cyril Jones Jagaraj
- MND Research Centre, Macquarie Medical School, Faculty of Medicine, Health and Human Sciences, Macquarie University, 75 Talavera Road, Sydney, NSW, 2109, Australia
| | - Sina Shadfar
- MND Research Centre, Macquarie Medical School, Faculty of Medicine, Health and Human Sciences, Macquarie University, 75 Talavera Road, Sydney, NSW, 2109, Australia
| | - Sara Assar Kashani
- MND Research Centre, Macquarie Medical School, Faculty of Medicine, Health and Human Sciences, Macquarie University, 75 Talavera Road, Sydney, NSW, 2109, Australia
| | - Sayanthooran Saravanabavan
- MND Research Centre, Macquarie Medical School, Faculty of Medicine, Health and Human Sciences, Macquarie University, 75 Talavera Road, Sydney, NSW, 2109, Australia
| | - Fabiha Farzana
- MND Research Centre, Macquarie Medical School, Faculty of Medicine, Health and Human Sciences, Macquarie University, 75 Talavera Road, Sydney, NSW, 2109, Australia
| | - Julie D Atkin
- MND Research Centre, Macquarie Medical School, Faculty of Medicine, Health and Human Sciences, Macquarie University, 75 Talavera Road, Sydney, NSW, 2109, Australia.
- La Trobe Institute for Molecular Science, La Trobe University, Bundoora, Melbourne, VIC, 3086, Australia.
| |
Collapse
|
14
|
Bahat G, Ozkok S. The Current Landscape of Pharmacotherapies for Sarcopenia. Drugs Aging 2024; 41:83-112. [PMID: 38315328 DOI: 10.1007/s40266-023-01093-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/28/2023] [Indexed: 02/07/2024]
Abstract
Sarcopenia is a skeletal muscle disorder characterized by progressive and generalized decline in muscle mass and function. Although it is mostly known as an age-related disorder, it can also occur secondary to systemic diseases such as malignancy or organ failure. It has demonstrated a significant relationship with adverse outcomes, e.g., falls, disabilities, and even mortality. Several breakthroughs have been made to find a pharmaceutical therapy for sarcopenia over the years, and some have come up with promising findings. Yet still no drug has been approved for its treatment. The key factor that makes finding an effective pharmacotherapy so challenging is the general paradigm of standalone/single diseases, traditionally adopted in medicine. Today, it is well known that sarcopenia is a complex disorder caused by multiple factors, e.g., imbalance in protein turnover, satellite cell and mitochondrial dysfunction, hormonal changes, low-grade inflammation, senescence, anorexia of aging, and behavioral factors such as low physical activity. Therefore, pharmaceuticals, either alone or combined, that exhibit multiple actions on these factors simultaneously will likely be the drug of choice to manage sarcopenia. Among various drug options explored throughout the years, testosterone still has the most cumulated evidence regarding its effects on muscle health and its safety. A mas receptor agonist, BIO101, stands out as a recent promising pharmaceutical. In addition to the conventional strategies (i.e., nutritional support and physical exercise), therapeutics with multiple targets of action or combination of multiple therapeutics with different targets/modes of action appear to promise greater benefit for the prevention and treatment of sarcopenia.
Collapse
Affiliation(s)
- Gulistan Bahat
- Division of Geriatrics, Department of Internal Medicine, Istanbul Medical School, Istanbul University, Capa, 34390, Istanbul, Turkey.
| | - Serdar Ozkok
- Division of Geriatrics, Department of Internal Medicine, Hatay Training and Research Hospital, Hatay, 31040, Turkey
| |
Collapse
|
15
|
Ding Z, Jiang M, Qian J, Gu D, Bai H, Cai M, Yao D. Role of transforming growth factor-β in peripheral nerve regeneration. Neural Regen Res 2024; 19:380-386. [PMID: 37488894 PMCID: PMC10503632 DOI: 10.4103/1673-5374.377588] [Citation(s) in RCA: 15] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2023] [Revised: 03/29/2023] [Accepted: 04/27/2023] [Indexed: 07/26/2023] Open
Abstract
Injuries caused by trauma and neurodegenerative diseases can damage the peripheral nervous system and cause functional deficits. Unlike in the central nervous system, damaged axons in peripheral nerves can be induced to regenerate in response to intrinsic cues after reprogramming or in a growth-promoting microenvironment created by Schwann cells. However, axon regeneration and repair do not automatically result in the restoration of function, which is the ultimate therapeutic goal but also a major clinical challenge. Transforming growth factor (TGF) is a multifunctional cytokine that regulates various biological processes including tissue repair, embryo development, and cell growth and differentiation. There is accumulating evidence that TGF-β family proteins participate in peripheral nerve repair through various factors and signaling pathways by regulating the growth and transformation of Schwann cells; recruiting specific immune cells; controlling the permeability of the blood-nerve barrier, thereby stimulating axon growth; and inhibiting remyelination of regenerated axons. TGF-β has been applied to the treatment of peripheral nerve injury in animal models. In this context, we review the functions of TGF-β in peripheral nerve regeneration and potential clinical applications.
Collapse
Affiliation(s)
- Zihan Ding
- School of Life Sciences, Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, Nantong University, Nantong, Jiangsu Province, China
| | - Maorong Jiang
- School of Life Sciences, Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, Nantong University, Nantong, Jiangsu Province, China
| | - Jiaxi Qian
- School of Life Sciences, Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, Nantong University, Nantong, Jiangsu Province, China
| | - Dandan Gu
- School of Life Sciences, Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, Nantong University, Nantong, Jiangsu Province, China
| | - Huiyuan Bai
- School of Life Sciences, Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, Nantong University, Nantong, Jiangsu Province, China
| | - Min Cai
- Medical School of Nantong University, Nantong, Jiangsu Province, China
| | - Dengbing Yao
- School of Life Sciences, Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, Nantong University, Nantong, Jiangsu Province, China
| |
Collapse
|
16
|
Kuang R, Zhang Y, Wu G, Zhu Z, Xu S, Liu X, Xu Y, Luo Y. Long Non-coding RNAs Influence Aging Process of Sciatic Nerves in SD Rats. Comb Chem High Throughput Screen 2024; 27:2140-2150. [PMID: 37691192 PMCID: PMC11348477 DOI: 10.2174/1386207326666230907115800] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2023] [Revised: 08/19/2023] [Accepted: 08/23/2023] [Indexed: 09/12/2023]
Abstract
OBJECTIVES To investigate the long non-coding RNAs (lncRNAs) changes in the sciatic nerve (SN) in Sprague Dawley (SD) rats during aging. METHODS Eighteen healthy SD rats were selected at the age of 1 month (1M) and 24 months (24M) and SNs were collected. High-throughput transcriptome sequencing and bioinformatics analysis were performed. Protein-protein interaction (PPI) networks and competing endogenous RNA (ceRNA) networks were established according to differentially expressed genes (DEGs). RESULT As the length of lncRNAs increased, its proportion to the total number of lncRNAs decreased. A total of 4079 DElncRNAs were identified in Con vs. 24M. GO analysis was primarily clustered in nerve and lipid metabolism, extracellular matrix, and vascularization-related fields. There were 17 nodes in the PPI network of the target genes of up-regulating genes including Itgb2, Lox, Col11a1, Wnt5a, Kras, etc. Using quantitative RT-PCR, microarray sequencing accuracy was validated. There were 169 nodes constructing the PPI network of down-regulated target genes, mainly including Col1a1, Hmgcs1, Hmgcr. CeRNA interaction networks were constructed. CONCLUSION Lipid metabolism, angiogenesis, and ECM fields might play an important role in the senescence process in SNs. Col3a1, Serpinh1, Hmgcr, and Fdps could be candidates for nerve aging research.
Collapse
Affiliation(s)
- Rui Kuang
- Department of Plastic Surgery, The First Affiliated Hospital of Sun Yat-Sen University, No. 58 Zhongshan Road 2, Guangzhou 510080, China
| | - Yi Zhang
- Department of Plastic Surgery, The First Affiliated Hospital of Sun Yat-Sen University, No. 58 Zhongshan Road 2, Guangzhou 510080, China
| | - Guanggeng Wu
- Department of Plastic Surgery, The First Affiliated Hospital of Sun Yat-Sen University, No. 58 Zhongshan Road 2, Guangzhou 510080, China
| | - Zhaowei Zhu
- Department of Plastic Surgery, The First Affiliated Hospital of Sun Yat-Sen University, No. 58 Zhongshan Road 2, Guangzhou 510080, China
| | - Shuqia Xu
- Department of Plastic Surgery, The First Affiliated Hospital of Sun Yat-Sen University, No. 58 Zhongshan Road 2, Guangzhou 510080, China
| | - Xiangxia Liu
- Department of Plastic Surgery, University of Tennessee Health Science Center, Memphis, TN, United States
| | - Yangbin Xu
- Department of Plastic Surgery, The First Affiliated Hospital of Sun Yat-Sen University, No. 58 Zhongshan Road 2, Guangzhou 510080, China
| | - Yunxiang Luo
- Department of Plastic Surgery, The First Affiliated Hospital of Sun Yat-Sen University, No. 58 Zhongshan Road 2, Guangzhou 510080, China
| |
Collapse
|
17
|
Hastings RL, Avila MF, Suneby E, Juros D, O'Young A, Peres da Silva J, Valdez G. Cellular and molecular evidence that synaptic Schwann cells contribute to aging of mouse neuromuscular junctions. Aging Cell 2023; 22:e13981. [PMID: 37771191 PMCID: PMC10652323 DOI: 10.1111/acel.13981] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Revised: 08/17/2023] [Accepted: 08/18/2023] [Indexed: 09/30/2023] Open
Abstract
Age-induced degeneration of the neuromuscular junction (NMJ) is associated with motor dysfunction and muscle atrophy. While the impact of aging on the NMJ presynapse and postsynapse is well-documented, little is known about the changes perisynaptic Schwann cells (PSCs), the synaptic glia of the NMJ, undergo during aging. Here, we examined PSCs in young, middle-aged, and old mice in three muscles with different susceptibility to aging. Using light and electron microscopy, we found that PSCs acquire age-associated cellular features either prior to or at the same time as the onset of NMJ degeneration. Notably, we found that aged PSCs fail to completely cap the NMJ even though they are more abundant in old compared with young mice. We also found that aging PSCs form processes that either intrude into the synaptic cleft or guide axonal sprouts to innervate other NMJs. We next profiled the transcriptome of PSCs and other Schwann cells (SCs) to identify mechanisms altered in aged PSCs. This analysis revealed that aged PSCs acquire a transcriptional pattern previously shown to promote phagocytosis that is absent in other SCs. It also showed that aged PSCs upregulate unique pro-inflammatory molecules compared to other aged SCs. Interestingly, neither synaptogenesis genes nor genes that are typically upregulated by repair SCs were induced in aged PSCs or other SCs. These findings provide insights into cellular and molecular mechanisms that could be targeted in PSCs to stave off the deleterious effects of aging on NMJs.
Collapse
Affiliation(s)
- Robert Louis Hastings
- Department of Molecular Biology, Cell Biology and Biochemistry, Brown University, Providence, Rhode Island, USA
| | | | - Emma Suneby
- Molecular Biology, Cell Biology, & Biochemistry Graduate Program, Brown University, Providence, Rhode Island, USA
| | - Devin Juros
- Department of Molecular Biology, Cell Biology and Biochemistry, Brown University, Providence, Rhode Island, USA
| | - Anson O'Young
- Department of Molecular Biology, Cell Biology and Biochemistry, Brown University, Providence, Rhode Island, USA
| | - Jason Peres da Silva
- Department of Molecular Biology, Cell Biology and Biochemistry, Brown University, Providence, Rhode Island, USA
| | - Gregorio Valdez
- Department of Molecular Biology, Cell Biology and Biochemistry, Brown University, Providence, Rhode Island, USA
- Center for Translational Neuroscience, Robert J. and Nancy D. Carney Institute for Brain Science, and Center on the Biology of Aging, Brown University, Providence, Rhode Island, USA
| |
Collapse
|
18
|
Fish LA, Ewing MD, Jaime D, Rich KA, Xi C, Wang X, Feder RE, Wharton KA, Rich MM, Arnold WD, Fallon JR. The MuSK-BMP pathway regulates synaptic Nav1.4 localization and muscle excitability. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.10.24.563837. [PMID: 37961580 PMCID: PMC10634800 DOI: 10.1101/2023.10.24.563837] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/15/2023]
Abstract
The neuromuscular junction (NMJ) is the linchpin of nerve-evoked muscle contraction. Broadly considered, the function of the NMJ is to transduce a nerve action potential into a muscle fiber action potential (MFAP). Efficient information transfer requires both cholinergic signaling, responsible for the generation of endplate potentials (EPPs), and excitation, the activation of postsynaptic voltage-gated sodium channels (Nav1.4) to trigger MFAPs. In contrast to the cholinergic apparatus, the signaling pathways that organize Nav1.4 and muscle fiber excitability are poorly characterized. Muscle-specific kinase (MuSK), in addition to its Ig1 domain-dependent role as an agrin-LRP4 receptor, is also a BMP co-receptor that binds BMPs via its Ig3 domain and shapes BMP-induced signaling and transcriptional output. Here we probed the function of the MuSK-BMP pathway at the NMJ using mice lacking the MuSK Ig3 domain ('ΔIg3-MuSK'). Synapses formed normally in ΔIg3-MuSK animals, but the postsynaptic apparatus was fragmented from the first weeks of life. Anatomical denervation was not observed at any age examined. Moreover, spontaneous and nerve-evoked acetylcholine release, AChR density, and endplate currents were comparable to WT. However, trains of nerve-evoked MFAPs in ΔIg3-MuSK muscle were abnormal as revealed by increased jitter and blocking in single fiber electromyography. Further, nerve-evoked compound muscle action potentials (CMAPs), as well as twitch and tetanic muscle torque force production, were also diminished. Finally, Nav1.4 levels were reduced at ΔIg3-MuSK synapses but not at the extrajunctional sarcolemma, indicating that the observed excitability defects are the result of impaired localization of this voltage-gated ion channel at the NMJ. We propose that MuSK plays two distinct roles at the NMJ: as an agrin-LRP4 receptor necessary for establishing and maintaining cholinergic signaling, and as a BMP co-receptor required for maintaining proper Nav1.4 density, nerve-evoked muscle excitability and force production. The MuSK-BMP pathway thus emerges as a target for modulating excitability and functional innervation, which are defective in conditions such as congenital myasthenic syndromes and aging.
Collapse
Affiliation(s)
- L. A. Fish
- Neuroscience Graduate Program, Brown University, Providence, RI 02912
- Carney Institute for Brain Science, Brown University, Providence, RI 02912
| | - M. D. Ewing
- Department of Neuroscience, Brown University, Providence, RI 02912
| | - D. Jaime
- Department of Molecular Biology, Cell Biology, and Biochemistry, Brown University, Providence, RI 02912
| | - K. A. Rich
- Neuroscience Graduate Program, Ohio State University, Columbus, OH 43210
| | - C. Xi
- Biotechnology Graduate Program, Brown University, Brown University, Providence, RI 02912
| | - X. Wang
- Department of Neuroscience Cell Biology and Physiology, Wright State University, Dayton, OH 45435
| | - R. E. Feder
- Department of Neuroscience, Brown University, Providence, RI 02912
| | - K. A. Wharton
- Department of Molecular Biology, Cell Biology, and Biochemistry, Brown University, Providence, RI 02912
| | - M. M. Rich
- Department of Neuroscience Cell Biology and Physiology, Wright State University, Dayton, OH 45435
| | - W. D. Arnold
- NextGen Precision Health Institute, University of Missouri, Columbia, MO 62511
| | - J. R. Fallon
- Carney Institute for Brain Science, Brown University, Providence, RI 02912
- Department of Neuroscience, Brown University, Providence, RI 02912
| |
Collapse
|
19
|
Duranti E, Villa C. Muscle Involvement in Amyotrophic Lateral Sclerosis: Understanding the Pathogenesis and Advancing Therapeutics. Biomolecules 2023; 13:1582. [PMID: 38002264 PMCID: PMC10669302 DOI: 10.3390/biom13111582] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2023] [Revised: 10/20/2023] [Accepted: 10/25/2023] [Indexed: 11/26/2023] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a fatal condition characterized by the selective loss of motor neurons in the motor cortex, brainstem, and spinal cord. Muscle involvement, muscle atrophy, and subsequent paralysis are among the main features of this disease, which is defined as a neuromuscular disorder. ALS is a persistently progressive disease, and as motor neurons continue to degenerate, individuals with ALS experience a gradual decline in their ability to perform daily activities. Ultimately, muscle function loss may result in paralysis, presenting significant challenges in mobility, communication, and self-care. While the majority of ALS research has traditionally focused on pathogenic pathways in the central nervous system, there has been a great interest in muscle research. These studies were carried out on patients and animal models in order to better understand the molecular mechanisms involved and to develop therapies aimed at improving muscle function. This review summarizes the features of ALS and discusses the role of muscle, as well as examines recent studies in the development of treatments.
Collapse
Affiliation(s)
| | - Chiara Villa
- School of Medicine and Surgery, University of Milano-Bicocca, 20900 Monza, Italy;
| |
Collapse
|
20
|
Lin YL, Nhieu J, Lerdall T, Milbauer L, Wei CW, Lee DJ, Oh SH, Thayer S, Wei LN. A novel 3D bilayer hydrogel tri-culture system for studying functional motor units. Cell Biosci 2023; 13:168. [PMID: 37700376 PMCID: PMC10496371 DOI: 10.1186/s13578-023-01115-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2023] [Accepted: 08/27/2023] [Indexed: 09/14/2023] Open
Abstract
BACKGROUND A motor unit (MU) is formed by a single alpha motor neuron (MN) and the muscle fibers it innervates. The MU is essential for all voluntary movements. Functional deficits in the MU result in neuromuscular disorders (NMDs). The pathological mechanisms underlying most NMDs remain poorly understood, in part due to the lack of in vitro models that can comprehensively recapitulate multistage intercellular interactions and physiological function of the MU. RESULTS We have designed a novel three-dimensional (3D) bilayer hydrogel tri-culture system where architecturally organized MUs can form in vitro. A sequential co-culture procedure using the three cell types of a MU, MN, myoblast, and Schwann cell was designed to construct a co-differentiating tri-culture on a bilayer hydrogel matrix. We utilized a µ-molded hydrogel with an additional Matrigel layer to form the bilayer hydrogel device. The µ-molded hydrogel layer provides the topological cues for myoblast differentiation. The Matrigel layer, with embedded Schwann cells, not only separates the MNs from myoblasts but also provides a proper micro-environment for MU development. The completed model shows key MU features including an organized MU structure, myelinated nerves, aligned myotubes innervated on clustered neuromuscular junctions (NMJs), MN-driven myotube contractions, and increases in cytosolic Ca2+ upon stimulation. CONCLUSIONS This organized and functional in vitro MU model provides an opportunity to study pathological events involved in NMDs and peripheral neuropathies, and can serve as a platform for physiological and pharmacological studies such as modeling and drug screening. Technically, the rational of this 3D bilayer hydrogel co-culture system exploits multiple distinct properties of hydrogels, facilitating effective and efficient co-culturing of diverse cell types for tissue engineering.
Collapse
Affiliation(s)
- Yu-Lung Lin
- Department of Pharmacology, University of Minnesota Medical School, Minneapolis, MN, 55455, USA
- The Ph.D. Program for Translational Medicine, College of Medical Sciences and Technology, Taipei Medical University, Taipei, Taiwan
- International Ph.D. Program for Translational Science, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan
| | - Jennifer Nhieu
- Department of Pharmacology, University of Minnesota Medical School, Minneapolis, MN, 55455, USA
| | - Thomas Lerdall
- Department of Pharmacology, University of Minnesota Medical School, Minneapolis, MN, 55455, USA
| | - Liming Milbauer
- Department of Pharmacology, University of Minnesota Medical School, Minneapolis, MN, 55455, USA
| | - Chin-Wen Wei
- Department of Pharmacology, University of Minnesota Medical School, Minneapolis, MN, 55455, USA
| | - Dong Jun Lee
- Department of Electrical and Computer Engineering, University of Minnesota, Minneapolis, MN, 55455, USA
| | - Sang-Hyun Oh
- Department of Electrical and Computer Engineering, University of Minnesota, Minneapolis, MN, 55455, USA
| | - Stanley Thayer
- Department of Pharmacology, University of Minnesota Medical School, Minneapolis, MN, 55455, USA
| | - Li-Na Wei
- Department of Pharmacology, University of Minnesota Medical School, Minneapolis, MN, 55455, USA.
| |
Collapse
|
21
|
Lloyd EM, Pinniger GJ, Murphy RM, Grounds MD. Slow or fast: Implications of myofibre type and associated differences for manifestation of neuromuscular disorders. Acta Physiol (Oxf) 2023; 238:e14012. [PMID: 37306196 DOI: 10.1111/apha.14012] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Revised: 05/30/2023] [Accepted: 06/06/2023] [Indexed: 06/13/2023]
Abstract
Many neuromuscular disorders can have a differential impact on a specific myofibre type, forming the central premise of this review. The many different skeletal muscles in mammals contain a spectrum of slow- to fast-twitch myofibres with varying levels of protein isoforms that determine their distinctive contractile, metabolic, and other properties. The variations in functional properties across the range of classic 'slow' to 'fast' myofibres are outlined, combined with exemplars of the predominantly slow-twitch soleus and fast-twitch extensor digitorum longus muscles, species comparisons, and techniques used to study these properties. Other intrinsic and extrinsic differences are discussed in the context of slow and fast myofibres. These include inherent susceptibility to damage, myonecrosis, and regeneration, plus extrinsic nerves, extracellular matrix, and vasculature, examined in the context of growth, ageing, metabolic syndrome, and sexual dimorphism. These many differences emphasise the importance of carefully considering the influence of myofibre-type composition on manifestation of various neuromuscular disorders across the lifespan for both sexes. Equally, understanding the different responses of slow and fast myofibres due to intrinsic and extrinsic factors can provide deep insight into the precise molecular mechanisms that initiate and exacerbate various neuromuscular disorders. This focus on the influence of different myofibre types is of fundamental importance to enhance translation for clinical management and therapies for many skeletal muscle disorders.
Collapse
Affiliation(s)
- Erin M Lloyd
- Department of Anatomy, Physiology and Human Biology, School of Human Sciences, The University of Western Australia, Perth, Western Australia, Australia
- Curtin Health Innovation Research Institute, Curtin Medical School, Curtin University, Bentley, Western Australia, Australia
| | - Gavin J Pinniger
- Department of Anatomy, Physiology and Human Biology, School of Human Sciences, The University of Western Australia, Perth, Western Australia, Australia
| | - Robyn M Murphy
- Department of Biochemistry and Chemistry, School of Agriculture, Biomedicine and Environment, La Trobe University, Melbourne, Victoria, Australia
| | - Miranda D Grounds
- Department of Anatomy, Physiology and Human Biology, School of Human Sciences, The University of Western Australia, Perth, Western Australia, Australia
| |
Collapse
|
22
|
Zhang H, Zhang Z, Lin H. Research progress on the reduced neural repair ability of aging Schwann cells. Front Cell Neurosci 2023; 17:1228282. [PMID: 37545880 PMCID: PMC10398339 DOI: 10.3389/fncel.2023.1228282] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2023] [Accepted: 07/06/2023] [Indexed: 08/08/2023] Open
Abstract
Peripheral nerve injury (PNI) is associated with delayed repair of the injured nerves in elderly patients, resulting in loss of nerve function, chronic pain, muscle atrophy, and permanent disability. Therefore, the mechanism underlying the delayed repair of peripheral nerves in aging patients should be investigated. Schwann cells (SCs) play a crucial role in repairing PNI and regulating various nerve-repair genes after injury. SCs also promote peripheral nerve repair through various modalities, including mediating nerve demyelination, secreting neurotrophic factors, establishing Büngner bands, clearing axon and myelin debris, and promoting axon remyelination. However, aged SCs undergo structural and functional changes, leading to demyelination and dedifferentiation disorders, decreased secretion of neurotrophic factors, impaired clearance of axonal and myelin debris, and reduced capacity for axon remyelination. As a result, aged SCs may result in delayed repair of nerves after injury. This review article aimed to examine the mechanism underlying the diminished neural repair ability of aging SCs.
Collapse
|
23
|
Liu N, Butcher JT, Nakano A, del Campo A. Changes in macrophage immunometabolism as a marker of skeletal muscle dysfunction across the lifespan. Aging (Albany NY) 2023; 15:4035-4050. [PMID: 37244285 PMCID: PMC10258037 DOI: 10.18632/aging.204750] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Accepted: 05/16/2023] [Indexed: 05/29/2023]
Abstract
One of the most pronounced changes in the elderly is loss of strength and mobility due to the decline of skeletal muscle function, resulting in a multifactorial condition termed sarcopenia. Although significant clinical changes begin to manifest at advanced ages, recent studies have shown that changes at the cellular and molecular level precede the symptomatology of sarcopenia. By utilizing a single-cell transcriptomic atlas of mouse skeletal muscle across the lifespan, we identified a clear sign of immune senescence that presents during middle age. More importantly, the change in macrophage phenotype in middle age may explain the changes in extracellular matrix composition, especially collagen synthesis, that contributes to fibrosis and overall muscle weakness with advanced age. Our results show a novel paradigm whereby skeletal muscle dysfunction is driven by alterations in tissue-resident macrophages before the appearance of clinical symptoms in middle-aged mice, providing a new therapeutic approach via regulation of immunometabolism.
Collapse
Affiliation(s)
- Norika Liu
- Department of Cell Physiology, The Jikei University School of Medicine, Tokyo, Japan
- Department of Molecular Cell and Developmental Biology, University of California Los Angeles, Los Angeles, CA 90095, USA
| | - Joshua T. Butcher
- Department of Physiological Sciences, College of Veterinary Medicine, Oklahoma State University, Stillwater, OK 74078, USA
| | - Atsushi Nakano
- Department of Cell Physiology, The Jikei University School of Medicine, Tokyo, Japan
- Department of Molecular Cell and Developmental Biology, University of California Los Angeles, Los Angeles, CA 90095, USA
- David Geffen Department of Medicine, Division of Cardiology, University of California Los Angeles, Los Angeles, CA 90095, USA
| | - Andrea del Campo
- Laboratorio de Fisiología y Bioenergetica Celular, Facultad de Química y de Farmacia, Pontificia Universidad Católica de Chile, Santiago 7810000, Chile
| |
Collapse
|
24
|
Willows JW, Gunsch G, Paradie E, Blaszkiewicz M, Tonniges JR, Pino MF, Smith SR, Sparks LM, Townsend KL. Schwann cells contribute to demyelinating diabetic neuropathy and nerve terminal structures in white adipose tissue. iScience 2023; 26:106189. [PMID: 36895649 PMCID: PMC9989657 DOI: 10.1016/j.isci.2023.106189] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Revised: 12/09/2022] [Accepted: 02/08/2023] [Indexed: 02/15/2023] Open
Abstract
Peripheral neuropathy, which can include axonal degeneration and/or demyelination, impacts adipose tissues with obesity, diabetes, and aging. However, the presence of demyelinating neuropathy had not yet been explored in adipose. Both demyelinating neuropathies and axonopathies implicate Schwann cells (SCs), a glial support cell that myelinates axons and contributes to nerve regeneration after injury. We performed a comprehensive assessment of SCs and myelination patterns of subcutaneous white adipose tissue (scWAT) nerves, and changes across altered energy balance states. We found that mouse scWAT contains both myelinated and unmyelinated nerves and is populated by SCs, including SCs that were associated with synaptic vesicle-containing nerve terminals. BTBR ob/ob mice, a model of diabetic peripheral neuropathy, exhibited small fiber demyelinating neuropathy and alterations in SC marker gene expression in adipose that were similar to obese human adipose. These data indicate that adipose SCs regulate the plasticity of tissue nerves and become dysregulated in diabetes.
Collapse
Affiliation(s)
- Jake W Willows
- Department of Neurological Surgery, The Ohio State University, Columbus, OH, USA
| | - Gilian Gunsch
- Department of Neurological Surgery, The Ohio State University, Columbus, OH, USA
| | - Emma Paradie
- Department of Neurological Surgery, The Ohio State University, Columbus, OH, USA
| | | | - Jeffrey R Tonniges
- Campus Microscopy and Imaging Facility, The Ohio State University, Columbus, OH, USA
| | - Maria F Pino
- Translational Research Institute, AdventHealth, Orlando, FL, USA
| | - Steven R Smith
- Translational Research Institute, AdventHealth, Orlando, FL, USA
| | - Lauren M Sparks
- Translational Research Institute, AdventHealth, Orlando, FL, USA
| | - Kristy L Townsend
- Department of Neurological Surgery, The Ohio State University, Columbus, OH, USA
| |
Collapse
|
25
|
Willows JW, Robinson M, Alshahal Z, Morrison SK, Mishra G, Cyr H, Blaszkiewicz M, Gunsch G, DiPietro S, Paradie E, Tero B, Harrington A, Ryzhova L, Liaw L, Reifsnyder PC, Harrison DE, Townsend KL. Age-related changes to adipose tissue and peripheral neuropathy in genetically diverse HET3 mice differ by sex and are not mitigated by rapamycin longevity treatment. Aging Cell 2023; 22:e13784. [PMID: 36798047 PMCID: PMC10086534 DOI: 10.1111/acel.13784] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Revised: 12/16/2022] [Accepted: 01/05/2023] [Indexed: 02/18/2023] Open
Abstract
Neural communication between the brain and adipose tissues regulates energy expenditure and metabolism through modulation of adipose tissue functions. We have recently demonstrated that under pathophysiological conditions (obesity, diabetes, and aging), total subcutaneous white adipose tissue (scWAT) innervation is decreased ('adipose neuropathy'). With advanced age in the C57BL/6J mouse, small fiber peripheral nerve endings in adipose tissue die back, resulting in reduced contact with adipose-resident blood vessels and other cells. This vascular neuropathy and parenchymal neuropathy together likely pose a physiological challenge for tissue function. In the current work, we used the genetically diverse HET3 mouse model to investigate the incidence of peripheral neuropathy and adipose tissue dysregulation across several ages in both male and female mice. We also investigated the anti-aging treatment rapamycin, an mTOR inhibitor, as a means to prevent or reduce adipose neuropathy. We found that HET3 mice displayed a reduced neuropathy phenotype compared to inbred C56BL/6 J mice, indicating genetic contributions to this aging phenotype. Compared to female HET3 mice, male HET3 mice had worse neuropathic phenotypes by 62 weeks of age. Female HET3 mice appeared to have increased protection from neuropathy until advanced age (126 weeks), after reproductive senescence. We found that rapamycin overall had little impact on neuropathy measures, and actually worsened adipose tissue inflammation and fibrosis. Despite its success as a longevity treatment in mice, higher doses and longer delivery paradigms for rapamycin may lead to a disconnect between life span and beneficial health outcomes.
Collapse
Affiliation(s)
- Jake W Willows
- Department of Neurological Surgery, The Ohio State University, Wexner Medical Center, Columbus, Ohio, USA
| | | | - Zahra Alshahal
- Department of Neurological Surgery, The Ohio State University, Wexner Medical Center, Columbus, Ohio, USA
| | - Samantha K Morrison
- Department of Neurological Surgery, The Ohio State University, Wexner Medical Center, Columbus, Ohio, USA
| | - Gargi Mishra
- Department of Neurological Surgery, The Ohio State University, Wexner Medical Center, Columbus, Ohio, USA
| | | | - Magdalena Blaszkiewicz
- Department of Neurological Surgery, The Ohio State University, Wexner Medical Center, Columbus, Ohio, USA
| | - Gilian Gunsch
- Department of Neurological Surgery, The Ohio State University, Wexner Medical Center, Columbus, Ohio, USA
| | - Sabrina DiPietro
- Department of Neurological Surgery, The Ohio State University, Wexner Medical Center, Columbus, Ohio, USA
| | - Emma Paradie
- Department of Neurological Surgery, The Ohio State University, Wexner Medical Center, Columbus, Ohio, USA
| | - Benjamin Tero
- Maine Medical Center Research Institute, Scarborough, Maine, USA
| | - Anne Harrington
- Maine Medical Center Research Institute, Scarborough, Maine, USA
| | - Larisa Ryzhova
- Maine Medical Center Research Institute, Scarborough, Maine, USA
| | - Lucy Liaw
- Maine Medical Center Research Institute, Scarborough, Maine, USA
| | | | | | - Kristy L Townsend
- Department of Neurological Surgery, The Ohio State University, Wexner Medical Center, Columbus, Ohio, USA.,University of Maine, Orono, Maine, USA
| |
Collapse
|
26
|
Gala DS, Titlow JS, Teodoro RO, Davis I. Far from home: the role of glial mRNA localization in synaptic plasticity. RNA (NEW YORK, N.Y.) 2023; 29:153-169. [PMID: 36442969 PMCID: PMC9891262 DOI: 10.1261/rna.079422.122] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Neurons and glia are highly polarized cells, whose distal cytoplasmic functional subdomains require specific proteins. Neurons have axonal and dendritic cytoplasmic extensions containing synapses whose plasticity is regulated efficiently by mRNA transport and localized translation. The principles behind these mechanisms are equally attractive for explaining rapid local regulation of distal glial cytoplasmic projections, independent of their cell nucleus. However, in contrast to neurons, mRNA localization has received little experimental attention in glia. Nevertheless, there are many functionally diverse glial subtypes containing extensive networks of long cytoplasmic projections with likely localized regulation that influence neurons and their synapses. Moreover, glia have many other neuron-like properties, including electrical activity, secretion of gliotransmitters and calcium signaling, influencing, for example, synaptic transmission, plasticity and axon pruning. Here, we review previous studies concerning glial transcripts with important roles in influencing synaptic plasticity, focusing on a few cases involving localized translation. We discuss a variety of important questions about mRNA transport and localized translation in glia that remain to be addressed, using cutting-edge tools already available for neurons.
Collapse
Affiliation(s)
- Dalia S Gala
- Department of Biochemistry, The University of Oxford, Oxford OX1 3QU, United Kingdom
| | - Joshua S Titlow
- Department of Biochemistry, The University of Oxford, Oxford OX1 3QU, United Kingdom
| | - Rita O Teodoro
- iNOVA4Health, NOVA Medical School-Faculdade de Ciências Médicas, Universidade Nova de Lisboa, Lisboa 1169-056, Portugal
| | - Ilan Davis
- Department of Biochemistry, The University of Oxford, Oxford OX1 3QU, United Kingdom
| |
Collapse
|
27
|
Anderson G. Amyotrophic Lateral Sclerosis Pathoetiology and Pathophysiology: Roles of Astrocytes, Gut Microbiome, and Muscle Interactions via the Mitochondrial Melatonergic Pathway, with Disruption by Glyphosate-Based Herbicides. Int J Mol Sci 2022; 24:ijms24010587. [PMID: 36614029 PMCID: PMC9820185 DOI: 10.3390/ijms24010587] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2022] [Revised: 12/23/2022] [Accepted: 12/28/2022] [Indexed: 12/31/2022] Open
Abstract
The pathoetiology and pathophysiology of motor neuron loss in amyotrophic lateral sclerosis (ALS) are still to be determined, with only a small percentage of ALS patients having a known genetic risk factor. The article looks to integrate wider bodies of data on the biological underpinnings of ALS, highlighting the integrative role of alterations in the mitochondrial melatonergic pathways and systemic factors regulating this pathway across a number of crucial hubs in ALS pathophysiology, namely glia, gut, and the muscle/neuromuscular junction. It is proposed that suppression of the mitochondrial melatonergic pathway underpins changes in muscle brain-derived neurotrophic factor, and its melatonergic pathway mimic, N-acetylserotonin, leading to a lack of metabolic trophic support at the neuromuscular junction. The attenuation of the melatonergic pathway in astrocytes prevents activation of toll-like receptor agonists-induced pro-inflammatory transcription factors, NF-kB, and yin yang 1, from having a built-in limitation on inflammatory induction that arises from their synchronized induction of melatonin release. Such maintained astrocyte activation, coupled with heightened microglia reactivity, is an important driver of motor neuron susceptibility in ALS. Two important systemic factors, gut dysbiosis/permeability and pineal melatonin mediate many of their beneficial effects via their capacity to upregulate the mitochondrial melatonergic pathway in central and systemic cells. The mitochondrial melatonergic pathway may be seen as a core aspect of cellular function, with its suppression increasing reactive oxygen species (ROS), leading to ROS-induced microRNAs, thereby altering the patterning of genes induced. It is proposed that the increased occupational risk of ALS in farmers, gardeners, and sportsmen and women is intimately linked to exposure, whilst being physically active, to the widely used glyphosate-based herbicides. This has numerous research and treatment implications.
Collapse
Affiliation(s)
- George Anderson
- CRC Scotland & London, Eccleston Square, London SW1V 1PG, UK
| |
Collapse
|
28
|
Negro S, Pirazzini M, Rigoni M. Models and methods to study Schwann cells. J Anat 2022; 241:1235-1258. [PMID: 34988978 PMCID: PMC9558160 DOI: 10.1111/joa.13606] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2021] [Revised: 11/26/2021] [Accepted: 11/29/2021] [Indexed: 12/22/2022] Open
Abstract
Schwann cells (SCs) are fundamental components of the peripheral nervous system (PNS) of all vertebrates and play essential roles in development, maintenance, function, and regeneration of peripheral nerves. There are distinct populations of SCs including: (1) myelinating SCs that ensheath axons by a specialized plasma membrane, called myelin, which enhances the conduction of electric impulses; (2) non-myelinating SCs, including Remak SCs, which wrap bundles of multiple axons of small caliber, and perysinaptic SCs (PSCs), associated with motor axon terminals at the neuromuscular junction (NMJ). All types of SCs contribute to PNS regeneration through striking morphological and functional changes in response to nerve injury, are affected in peripheral neuropathies and show abnormalities and a diminished plasticity during aging. Therefore, methodological approaches to study and manipulate SCs in physiological and pathophysiological conditions are crucial to expand the present knowledge on SC biology and to devise new therapeutic strategies to counteract neurodegenerative conditions and age-derived denervation. We present here an updated overview of traditional and emerging methodologies for the study of SCs for scientists approaching this research field.
Collapse
Affiliation(s)
- Samuele Negro
- Department of Biomedical SciencesUniversity of PaduaPaduaItaly
| | - Marco Pirazzini
- Department of Biomedical SciencesUniversity of PaduaPaduaItaly
- CIR‐MyoCentro Interdipartimentale di Ricerca di MiologiaUniversity of PaduaPadovaItaly
| | - Michela Rigoni
- Department of Biomedical SciencesUniversity of PaduaPaduaItaly
- CIR‐MyoCentro Interdipartimentale di Ricerca di MiologiaUniversity of PaduaPadovaItaly
| |
Collapse
|
29
|
Deschenes MR, Flannery R, Hawbaker A, Patek L, Mifsud M. Adaptive Remodeling of the Neuromuscular Junction with Aging. Cells 2022; 11:cells11071150. [PMID: 35406714 PMCID: PMC8997609 DOI: 10.3390/cells11071150] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2022] [Revised: 03/24/2022] [Accepted: 03/24/2022] [Indexed: 02/06/2023] Open
Abstract
Aging is associated with gradual degeneration, in mass and function, of the neuromuscular system. This process, referred to as “sarcopenia”, is considered a disease by itself, and it has been linked to a number of other serious maladies such as type II diabetes, osteoporosis, arthritis, cardiovascular disease, and even dementia. While the molecular causes of sarcopenia remain to be fully elucidated, recent findings have implicated the neuromuscular junction (NMJ) as being an important locus in the development and progression of that malady. This synapse, which connects motor neurons to the muscle fibers that they innervate, has been found to degenerate with age, contributing both to senescent-related declines in muscle mass and function. The NMJ also shows plasticity in response to a number of neuromuscular diseases such as amyotrophic lateral sclerosis (ALS) and Lambert-Eaton myasthenic syndrome (LEMS). Here, the structural and functional degradation of the NMJ associated with aging and disease is described, along with the measures that might be taken to effectively mitigate, if not fully prevent, that degeneration.
Collapse
|
30
|
Deschenes MR, Patek LG, Trebelhorn AM, High MC, Flannery RE. Juvenile Neuromuscular Systems Show Amplified Disturbance to Muscle Unloading. Front Physiol 2021; 12:754052. [PMID: 34759841 PMCID: PMC8573242 DOI: 10.3389/fphys.2021.754052] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2021] [Accepted: 09/27/2021] [Indexed: 11/21/2022] Open
Abstract
Muscle unloading results in severe disturbance in neuromuscular function. During juvenile stages of natural development, the neuromuscular system experiences a high degree of plasticity in function and structure. This study aimed to determine whether muscle unloading imposed during juvenile development would elicit more severe disruption in neuromuscular function than when imposed on fully developed, mature neuromuscular systems. Twenty juvenile (3 months old) and 20 mature (8 months old) rats were equally divided into unloaded and control groups yielding a total of four groups (N = 10/each). Following the 2 week intervention period, soleus muscles were surgically extracted and using an ex vivo muscle stimulation and recording system, were examined for neuromuscular function. The unloading protocol was found to have elicited significant (P ≤ 0.05) declines in whole muscle wet weight in both juvenile and mature muscles, but of a similar degree (P = 0.286). Results also showed that juvenile muscles displayed significantly greater decay in peak force due to unloading than mature muscles, such a finding was also made for specific tension or force/muscle mass. When examining neuromuscular efficiency, i.e., function of the neuromuscular junction, it again was noted that juvenile systems were more negatively affected by muscle unloading than mature systems. These results indicate that juvenile neuromuscular systems are more sensitive to the effects of unloading than mature ones, and that the primary locus of this developmental related difference is likely the neuromuscular junction as indicated by age-related differences in neuromuscular transmission efficiency.
Collapse
Affiliation(s)
- Michael R Deschenes
- Department of Kinesiology and Health Sciences, College of William & Mary, Williamsburg, VA, United States.,Program in Neuroscience, College of William & Mary, Williamsburg, VA, United States
| | - Leah G Patek
- Department of Kinesiology and Health Sciences, College of William & Mary, Williamsburg, VA, United States
| | - Audrey M Trebelhorn
- Department of Kinesiology and Health Sciences, College of William & Mary, Williamsburg, VA, United States
| | - Madeline C High
- Program in Neuroscience, College of William & Mary, Williamsburg, VA, United States
| | - Rachel E Flannery
- Department of Kinesiology and Health Sciences, College of William & Mary, Williamsburg, VA, United States
| |
Collapse
|
31
|
Pikatza-Menoio O, Elicegui A, Bengoetxea X, Naldaiz-Gastesi N, López de Munain A, Gerenu G, Gil-Bea FJ, Alonso-Martín S. The Skeletal Muscle Emerges as a New Disease Target in Amyotrophic Lateral Sclerosis. J Pers Med 2021; 11:671. [PMID: 34357138 PMCID: PMC8307751 DOI: 10.3390/jpm11070671] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2021] [Revised: 07/09/2021] [Accepted: 07/14/2021] [Indexed: 01/02/2023] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a fatal neurodegenerative disorder that leads to progressive degeneration of motor neurons (MNs) and severe muscle atrophy without effective treatment. Most research on ALS has been focused on the study of MNs and supporting cells of the central nervous system. Strikingly, the recent observations of pathological changes in muscle occurring before disease onset and independent from MN degeneration have bolstered the interest for the study of muscle tissue as a potential target for delivery of therapies for ALS. Skeletal muscle has just been described as a tissue with an important secretory function that is toxic to MNs in the context of ALS. Moreover, a fine-tuning balance between biosynthetic and atrophic pathways is necessary to induce myogenesis for muscle tissue repair. Compromising this response due to primary metabolic abnormalities in the muscle could trigger defective muscle regeneration and neuromuscular junction restoration, with deleterious consequences for MNs and thereby hastening the development of ALS. However, it remains puzzling how backward signaling from the muscle could impinge on MN death. This review provides a comprehensive analysis on the current state-of-the-art of the role of the skeletal muscle in ALS, highlighting its contribution to the neurodegeneration in ALS through backward-signaling processes as a newly uncovered mechanism for a peripheral etiopathogenesis of the disease.
Collapse
Affiliation(s)
- Oihane Pikatza-Menoio
- Neuromuscular Diseases Group, Neurosciences Area, Biodonostia Health Research Institute, 20014 Donostia/San Sebastián, Spain; (O.P.-M.); (A.E.); (X.B.); (N.N.-G.); (A.L.d.M.); (G.G.); (F.J.G.-B.)
- CIBERNED, Carlos III Institute, Spanish Ministry of Economy & Competitiveness, 28031 Madrid, Spain
| | - Amaia Elicegui
- Neuromuscular Diseases Group, Neurosciences Area, Biodonostia Health Research Institute, 20014 Donostia/San Sebastián, Spain; (O.P.-M.); (A.E.); (X.B.); (N.N.-G.); (A.L.d.M.); (G.G.); (F.J.G.-B.)
- CIBERNED, Carlos III Institute, Spanish Ministry of Economy & Competitiveness, 28031 Madrid, Spain
| | - Xabier Bengoetxea
- Neuromuscular Diseases Group, Neurosciences Area, Biodonostia Health Research Institute, 20014 Donostia/San Sebastián, Spain; (O.P.-M.); (A.E.); (X.B.); (N.N.-G.); (A.L.d.M.); (G.G.); (F.J.G.-B.)
| | - Neia Naldaiz-Gastesi
- Neuromuscular Diseases Group, Neurosciences Area, Biodonostia Health Research Institute, 20014 Donostia/San Sebastián, Spain; (O.P.-M.); (A.E.); (X.B.); (N.N.-G.); (A.L.d.M.); (G.G.); (F.J.G.-B.)
- CIBERNED, Carlos III Institute, Spanish Ministry of Economy & Competitiveness, 28031 Madrid, Spain
| | - Adolfo López de Munain
- Neuromuscular Diseases Group, Neurosciences Area, Biodonostia Health Research Institute, 20014 Donostia/San Sebastián, Spain; (O.P.-M.); (A.E.); (X.B.); (N.N.-G.); (A.L.d.M.); (G.G.); (F.J.G.-B.)
- CIBERNED, Carlos III Institute, Spanish Ministry of Economy & Competitiveness, 28031 Madrid, Spain
- Department of Neurology, Donostialdea Integrated Health Organization, Osakidetza Basque Health Service, 20014 Donostia/San Sebastián, Spain
- Department of Neurosciences, Faculty of Medicine and Nursery, University of the Basque Country UPV-EHU, 20014 Donostia/San Sebastián, Spain
| | - Gorka Gerenu
- Neuromuscular Diseases Group, Neurosciences Area, Biodonostia Health Research Institute, 20014 Donostia/San Sebastián, Spain; (O.P.-M.); (A.E.); (X.B.); (N.N.-G.); (A.L.d.M.); (G.G.); (F.J.G.-B.)
- CIBERNED, Carlos III Institute, Spanish Ministry of Economy & Competitiveness, 28031 Madrid, Spain
- Department of Physiology, University of the Basque Country UPV-EHU, 48940 Leioa, Spain
| | - Francisco Javier Gil-Bea
- Neuromuscular Diseases Group, Neurosciences Area, Biodonostia Health Research Institute, 20014 Donostia/San Sebastián, Spain; (O.P.-M.); (A.E.); (X.B.); (N.N.-G.); (A.L.d.M.); (G.G.); (F.J.G.-B.)
- CIBERNED, Carlos III Institute, Spanish Ministry of Economy & Competitiveness, 28031 Madrid, Spain
| | - Sonia Alonso-Martín
- Neuromuscular Diseases Group, Neurosciences Area, Biodonostia Health Research Institute, 20014 Donostia/San Sebastián, Spain; (O.P.-M.); (A.E.); (X.B.); (N.N.-G.); (A.L.d.M.); (G.G.); (F.J.G.-B.)
- CIBERNED, Carlos III Institute, Spanish Ministry of Economy & Competitiveness, 28031 Madrid, Spain
| |
Collapse
|