Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 902
Filtrar
1.
Sci Transl Med ; 16(765): eado7189, 2024 Sep 18.
Artículo en Inglés | MEDLINE | ID: mdl-39292800

RESUMEN

Muscle-specific kinase (MuSK) is essential for the formation, function, and preservation of neuromuscular synapses. Activation of MuSK by a MuSK agonist antibody may stabilize or improve the function of the neuromuscular junction (NMJ) in patients with disorders of the NMJ, such as congenital myasthenia (CM). Here, we generated and characterized ARGX-119, a first-in-class humanized agonist monoclonal antibody specific for MuSK, that is being developed for treatment of patients with neuromuscular diseases. We performed in vitro ligand-binding assays to show that ARGX-119 binds with high affinity to the Frizzled-like domain of human, nonhuman primate, rat, and mouse MuSK, without off-target binding, making it suitable for clinical development. Within the Fc region, ARGX-119 harbors L234A and L235A mutations to diminish potential immune-activating effector functions. Its mode of action is to activate MuSK, without interfering with its natural ligand neural Agrin, and cluster acetylcholine receptors in a dose-dependent manner, thereby stabilizing neuromuscular function. In a mouse model of DOK7 CM, ARGX-119 prevented early postnatal lethality and reversed disease relapse in adult Dok7 CM mice by restoring neuromuscular function and reducing muscle weakness and fatigability in a dose-dependent manner. Pharmacokinetic studies in nonhuman primates, rats, and mice revealed a nonlinear PK behavior of ARGX-119, indicative of target-mediated drug disposition and in vivo target engagement. On the basis of this proof-of-concept study, ARGX-119 has the potential to alleviate neuromuscular diseases hallmarked by impaired neuromuscular synaptic function, warranting further clinical development.


Asunto(s)
Modelos Animales de Enfermedad , Síndromes Miasténicos Congénitos , Proteínas Tirosina Quinasas Receptoras , Receptores Colinérgicos , Animales , Proteínas Tirosina Quinasas Receptoras/metabolismo , Humanos , Síndromes Miasténicos Congénitos/tratamiento farmacológico , Receptores Colinérgicos/metabolismo , Ratones , Unión Neuromuscular/efectos de los fármacos , Unión Neuromuscular/patología , Recurrencia , Ratas , Anticuerpos Monoclonales Humanizados/uso terapéutico , Anticuerpos Monoclonales Humanizados/farmacología
2.
Sci Rep ; 14(1): 19540, 2024 08 22.
Artículo en Inglés | MEDLINE | ID: mdl-39174694

RESUMEN

Amyotrophic lateral sclerosis (ALS) is a devastating paralytic disorder caused by the death of motoneurons. Several mutations in the KIF5A gene have been identified in patients with ALS. Some mutations affect the splicing sites of exon 27 leading to its deletion (Δ27 mutation). KIF5A Δ27 is aggregation-prone and pathogenic for motoneurons due to a toxic gain of function. Another mutation found to be enriched in ALS patients is a proline/leucine substitution at position 986 (P986L mutation). Bioinformatic analyses strongly suggest that this variant is benign. Our study aims to conduct functional studies in Drosophila to classify the KIF5A P986L variant. When expressed in motoneurons, KIF5A P986L does not modify the morphology of larval NMJ or the synaptic transmission. In addition, KIF5A P986L is uniformly distributed in axons and does not disturb mitochondria distribution. Locomotion at larval and adult stages is not affected by KIF5A P986L. Finally, both KIF5A WT and P986L expression in adult motoneurons extend median lifespan compared to control flies. Altogether, our data show that the KIF5A P986L variant is not pathogenic for motoneurons and may represent a hypomorphic allele, although it is not causative for ALS.


Asunto(s)
Esclerosis Amiotrófica Lateral , Cinesinas , Neuronas Motoras , Animales , Cinesinas/genética , Cinesinas/metabolismo , Neuronas Motoras/metabolismo , Neuronas Motoras/patología , Esclerosis Amiotrófica Lateral/genética , Esclerosis Amiotrófica Lateral/patología , Esclerosis Amiotrófica Lateral/metabolismo , Mutación , Humanos , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Drosophila , Unión Neuromuscular/metabolismo , Unión Neuromuscular/patología , Drosophila melanogaster/genética , Transmisión Sináptica/genética , Modelos Animales de Enfermedad , Axones/metabolismo , Axones/patología , Larva/genética , Larva/metabolismo
3.
Int J Mol Sci ; 25(16)2024 Aug 06.
Artículo en Inglés | MEDLINE | ID: mdl-39201265

RESUMEN

We investigated the shuttling of Homer protein isoforms identified in soluble (cytosolic) vs. insoluble (membrane-cytoskeletal) fraction and Homer protein-protein interaction/activation in the deep postural calf soleus (SOL) and non-postural gastrocnemius (GAS) muscles of het-/- mice, i.e., mice with an autosomal recessive variant responsible for a vestibular disorder, in order to further elucidate a) the underlying mechanisms of disrupted vestibular system-derived modulation on skeletal muscle, and b) molecular signaling at respective neuromuscular synapses. Heterozygote mice muscles served as the control (CTR). An increase in Homer cross-linking capacity was present in the SOL muscle of het-/- mice as a compensatory mechanism for the altered vestibule system function. Indeed, in both fractions, different Homer immunoreactive bands were detectable, as were Homer monomers (~43-48 kDa), Homer dimers (~100 kDa), and several other Homer multimer bands (>150 kDA). The het-/- GAS particulate fraction showed no Homer dimers vs. SOL. The het-/- SOL soluble fraction showed a twofold increase (+117%, p ≤ 0.0004) in Homer dimers and multimers. Homer monomers were completely absent from the SOL independent of the animals studied, suggesting muscle-specific changes in Homer monomer vs. dimer expression in the postural SOL vs. the non-postural GAS muscles. A morphological assessment showed an increase (+14%, p ≤ 0.0001) in slow/type-I myofiber cross-sectional area in the SOL of het-/- vs. CTR mice. Homer subcellular immuno-localization at the neuromuscular junction (NMJ) showed an altered expression in the SOL of het-/-mice, whereas only not-significant changes were found for all Homer isoforms, as judged by RT-qPCR analysis. Thus, muscle-specific changes, myofiber properties, and neuromuscular signaling mechanisms share causal relationships, as highlighted by the variable subcellular Homer isoform expression at the instable NMJs of vestibular lesioned het-/- mice.


Asunto(s)
Proteínas de Andamiaje Homer , Músculo Esquelético , Unión Neuromuscular , Animales , Proteínas de Andamiaje Homer/metabolismo , Proteínas de Andamiaje Homer/genética , Ratones , Músculo Esquelético/metabolismo , Músculo Esquelético/patología , Unión Neuromuscular/metabolismo , Unión Neuromuscular/patología , Ratones Noqueados , Masculino , Enfermedades Vestibulares/metabolismo , Enfermedades Vestibulares/patología , Enfermedades Vestibulares/genética , Isoformas de Proteínas/metabolismo , Isoformas de Proteínas/genética
4.
Sci Rep ; 14(1): 18575, 2024 08 10.
Artículo en Inglés | MEDLINE | ID: mdl-39127839

RESUMEN

Triosephosphate isomerase deficiency (TPI Df) is a rare multisystem disorder with severe neuromuscular symptoms which arises exclusively from mutations within the TPI1 gene. Studies of TPI Df have been limited due to the absence of mammalian disease models and difficulties obtaining patient samples. Recently, we developed a novel murine model of TPI Df which models the most common disease-causing mutation in humans, TPI1E105D. Using our model in the present study, the underlying pathogenesis of neuromuscular symptoms has been elucidated. This is the first report detailing studies of neuromuscular pathology within a murine model of TPI Df. We identified several contributors to neuromuscular symptoms, including neurodegeneration in the brain, alterations in neurotransmission at the neuromuscular junction, and reduced muscle fiber size. TPI Df mice also exhibited signs of cardiac pathology and displayed a deficit in vascular smooth muscle functionality. Together, these findings provide insight into pathogenesis of the neuromuscular symptoms in TPI Df and can guide the future development of therapeutics.


Asunto(s)
Modelos Animales de Enfermedad , Unión Neuromuscular , Triosa-Fosfato Isomerasa , Animales , Triosa-Fosfato Isomerasa/deficiencia , Triosa-Fosfato Isomerasa/genética , Triosa-Fosfato Isomerasa/metabolismo , Ratones , Unión Neuromuscular/patología , Unión Neuromuscular/metabolismo , Anemia Hemolítica Congénita no Esferocítica/genética , Anemia Hemolítica Congénita no Esferocítica/patología , Enfermedades Neuromusculares/genética , Enfermedades Neuromusculares/patología , Enfermedades Neuromusculares/etiología , Errores Innatos del Metabolismo de los Carbohidratos/genética , Mutación , Humanos
5.
Skelet Muscle ; 14(1): 17, 2024 Jul 24.
Artículo en Inglés | MEDLINE | ID: mdl-39044305

RESUMEN

BACKGROUND: Amyotrophic lateral sclerosis (ALS) is characterized by progressive motor neuron (MN) degeneration, leading to neuromuscular junction (NMJ) dismantling and severe muscle atrophy. The nuclear receptor interaction protein (NRIP) functions as a multifunctional protein. It directly interacts with calmodulin or α-actinin 2, serving as a calcium sensor for muscle contraction and maintaining sarcomere integrity. Additionally, NRIP binds with the acetylcholine receptor (AChR) for NMJ stabilization. Loss of NRIP in muscles results in progressive motor neuron degeneration with abnormal NMJ architecture, resembling ALS phenotypes. Therefore, we hypothesize that NRIP could be a therapeutic factor for ALS. METHODS: We used SOD1 G93A mice, expressing human SOD1 with the ALS-linked G93A mutation, as an ALS model. An adeno-associated virus vector encoding the human NRIP gene (AAV-NRIP) was generated and injected into the muscles of SOD1 G93A mice at 60 days of age, before disease onset. Pathological and behavioral changes were measured to evaluate the therapeutic effects of AAV-NRIP on the disease progression of SOD1 G93A mice. RESULTS: SOD1 G93A mice exhibited lower NRIP expression than wild-type mice in both the spinal cord and skeletal muscle tissues. Forced NRIP expression through AAV-NRIP intramuscular injection was observed in skeletal muscles and retrogradely transduced into the spinal cord. AAV-NRIP gene therapy enhanced movement distance and rearing frequencies in SOD1 G93A mice. Moreover, AAV-NRIP increased myofiber size and slow myosin expression, ameliorated NMJ degeneration and axon terminal denervation at NMJ, and increased the number of α-motor neurons (α-MNs) and compound muscle action potential (CMAP) in SOD1 G93A mice. CONCLUSIONS: AAV-NRIP gene therapy ameliorates muscle atrophy, motor neuron degeneration, and axon terminal denervation at NMJ, leading to increased NMJ transmission and improved motor functions in SOD1 G93A mice. Collectively, AAV-NRIP could be a potential therapeutic drug for ALS.


Asunto(s)
Esclerosis Amiotrófica Lateral , Dependovirus , Modelos Animales de Enfermedad , Terapia Genética , Ratones Transgénicos , Neuronas Motoras , Atrofia Muscular , Animales , Esclerosis Amiotrófica Lateral/genética , Esclerosis Amiotrófica Lateral/terapia , Esclerosis Amiotrófica Lateral/metabolismo , Esclerosis Amiotrófica Lateral/patología , Terapia Genética/métodos , Atrofia Muscular/genética , Atrofia Muscular/terapia , Atrofia Muscular/metabolismo , Atrofia Muscular/patología , Neuronas Motoras/metabolismo , Neuronas Motoras/patología , Dependovirus/genética , Ratones , Humanos , Músculo Esquelético/metabolismo , Músculo Esquelético/patología , Unión Neuromuscular/metabolismo , Unión Neuromuscular/patología , Vectores Genéticos/administración & dosificación , Degeneración Nerviosa/genética , Degeneración Nerviosa/terapia , Masculino , Superóxido Dismutasa-1/genética , Superóxido Dismutasa-1/metabolismo
6.
Biochem Biophys Res Commun ; 727: 150311, 2024 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-38950494

RESUMEN

In human Alzheimer's disease (AD), the aggregation of tau protein is considered a significant hallmark, along with amyloid-beta. The formation of neurofibrillary tangles due to aberrant phosphorylation of tau disrupts microtubule stability, leading to neuronal toxicity, dysfunction, and subsequent cell death. Nesfatin-1 is a neuropeptide primarily known for regulating appetite and energy homeostasis. However, the function of Nesfatin-1 in a neuroprotective role has not been investigated. In this study, we aimed to elucidate the effect of Nesfatin-1 on tau pathology using the Drosophila model system. Our findings demonstrate that Nesfatin-1 effectively mitigates the pathological phenotypes observed in Drosophila human Tau overexpression models. Nesfatin-1 overexpression rescued the neurodegenerative phenotypes in the adult fly's eye and bristle. Additionally, Nesfatin-1 improved locomotive behavior, neuromuscular junction formation, and lifespan in the hTau AD model. Moreover, Nesfatin-1 controls tauopathy by reducing the protein level of hTau. Overall, this research highlights the potential therapeutic applications of Nesfatin-1 in ameliorating the pathological features associated with Alzheimer's disease.


Asunto(s)
Enfermedad de Alzheimer , Modelos Animales de Enfermedad , Nucleobindinas , Proteínas tau , Animales , Enfermedad de Alzheimer/patología , Enfermedad de Alzheimer/metabolismo , Nucleobindinas/metabolismo , Nucleobindinas/genética , Proteínas tau/metabolismo , Proteínas tau/genética , Humanos , Proteínas de Drosophila/metabolismo , Proteínas de Drosophila/genética , Drosophila melanogaster/metabolismo , Proteínas de Unión al ADN/metabolismo , Proteínas de Unión al ADN/genética , Proteínas de Unión al Calcio/metabolismo , Proteínas de Unión al Calcio/genética , Unión Neuromuscular/metabolismo , Unión Neuromuscular/patología , Animales Modificados Genéticamente , Drosophila , Locomoción , Longevidad
7.
Brain Res Bull ; 216: 111036, 2024 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-39084570

RESUMEN

Amyotrophic lateral sclerosis (ALS) is a fatal neurological disease characterized by the progressive destruction of the neuromuscular junction (NMJ) and the degeneration of motor neurons, eventually leading to atrophy and paralysis of voluntary muscles responsible for motion and breathing. NMJs, synaptic connections between motor neurons and skeletal muscle fibers, are extremely fragile in ALS. To determine the effects of early electroacupuncture (EA) intervention on nerve reinnervation and regeneration following injury, a model of sciatic nerve injury (SNI) was first established using SOD1G93A mice, and early electroacupuncture (EA) intervention was conducted at Baihui (DU20), and bilateral Zusanli (ST36). The results revealed that EA increased the Sciatic nerve Functional Index, the structural integrity of the gastrocnemius muscles, and the cross-sectional area of muscle fibers, as well as up-regulated the expression of acetylcholinesterase and facilitated the co-location of α7 nicotinic acetate choline receptors and α-actinin. Overall, these results suggested that EA can promote the repair and regeneration of injured nerves and delay NMJ degeneration in SOD1G93A-SNI mice. Moreover, analysis of the cerebral cortex demonstrated that EA alleviated cortical motor neuron damage in SOD1G93A mice, potentially attributed to the inhibition of the cyclic GMP-AMP synthase-stimulator of interferon genes pathway and the release of interferon-ß suppressing the activation of natural killer cells and the secretion of interferon-γ, thereby further inhibiting microglial activation and the expression of inflammatory factors. In summary, EA delayed the degeneration of NMJ and mitigated the loss of cortical motor neurons, thus delaying disease onset, accompanied by alleviation of muscle atrophy and improvements in motor function in SOD1G93A mice.


Asunto(s)
Esclerosis Amiotrófica Lateral , Electroacupuntura , Ratones Transgénicos , Neuronas Motoras , Unión Neuromuscular , Animales , Electroacupuntura/métodos , Unión Neuromuscular/patología , Unión Neuromuscular/metabolismo , Neuronas Motoras/patología , Neuronas Motoras/fisiología , Ratones , Esclerosis Amiotrófica Lateral/terapia , Esclerosis Amiotrófica Lateral/patología , Esclerosis Amiotrófica Lateral/genética , Modelos Animales de Enfermedad , Masculino , Degeneración Nerviosa/terapia , Degeneración Nerviosa/patología , Músculo Esquelético/patología , Superóxido Dismutasa-1/genética , Superóxido Dismutasa-1/metabolismo , Nervio Ciático/lesiones , Nervio Ciático/patología , Ratones Endogámicos C57BL
8.
Int J Mol Sci ; 25(13)2024 Jul 03.
Artículo en Inglés | MEDLINE | ID: mdl-39000416

RESUMEN

5q-Spinal muscular atrophy (5q-SMA) is one of the most common neuromuscular diseases due to homozygous mutations in the SMN1 gene. This leads to a loss of function of the SMN1 gene, which in the end determines lower motor neuron degeneration. Since the generation of the first mouse models of SMA neuropathology, a complex degenerative involvement of the neuromuscular junction and peripheral axons of motor nerves, alongside lower motor neurons, has been described. The involvement of the neuromuscular junction in determining disease symptoms offers a possible parallel therapeutic target. This narrative review aims at providing an overview of the current knowledge about the pathogenesis and significance of neuromuscular junction dysfunction in SMA, circulating biomarkers, outcome measures and available or developing therapeutic approaches.


Asunto(s)
Neuronas Motoras , Atrofia Muscular Espinal , Unión Neuromuscular , Proteína 1 para la Supervivencia de la Neurona Motora , Unión Neuromuscular/metabolismo , Unión Neuromuscular/patología , Atrofia Muscular Espinal/genética , Atrofia Muscular Espinal/patología , Atrofia Muscular Espinal/metabolismo , Humanos , Animales , Neuronas Motoras/metabolismo , Neuronas Motoras/patología , Proteína 1 para la Supervivencia de la Neurona Motora/genética , Proteína 1 para la Supervivencia de la Neurona Motora/metabolismo , Biomarcadores , Modelos Animales de Enfermedad , Mutación , Ratones
9.
Biomolecules ; 14(7)2024 Jul 22.
Artículo en Inglés | MEDLINE | ID: mdl-39062592

RESUMEN

Amyotrophic lateral sclerosis (ALS) is a complex neuromuscular disease characterized by progressive motor neuron degeneration, neuromuscular junction dismantling, and muscle wasting. The pathological and therapeutic studies of ALS have long been neurocentric. However, recent insights have highlighted the significance of peripheral tissue, particularly skeletal muscle, in disease pathology and treatment. This is evidenced by restricted ALS-like muscle atrophy, which can retrogradely induce neuromuscular junction and motor neuron degeneration. Moreover, therapeutics targeting skeletal muscles can effectively decelerate disease progression by modulating muscle satellite cells for muscle repair, suppressing inflammation, and promoting the recovery or regeneration of the neuromuscular junction. This review summarizes and discusses therapeutic strategies targeting skeletal muscles for ALS treatment. It aims to provide a comprehensive reference for the development of novel therapeutics targeting skeletal muscles, potentially ameliorating the progression of ALS.


Asunto(s)
Esclerosis Amiotrófica Lateral , Músculo Esquelético , Esclerosis Amiotrófica Lateral/metabolismo , Esclerosis Amiotrófica Lateral/patología , Esclerosis Amiotrófica Lateral/terapia , Humanos , Músculo Esquelético/metabolismo , Músculo Esquelético/patología , Animales , Unión Neuromuscular/metabolismo , Unión Neuromuscular/patología , Neuronas Motoras/metabolismo , Neuronas Motoras/patología
10.
Mol Brain ; 17(1): 43, 2024 Jul 13.
Artículo en Inglés | MEDLINE | ID: mdl-39003465

RESUMEN

Dorsal switch protein 1(DSP1), a mammalian homolog of HMGB1, is firstly identified as a dorsal co-repressor in 1994. DSP1 contains HMG-box domain and functions as a transcriptional regulator in Drosophila melanogaster. It plays a crucial role in embryonic development, particularly in dorsal-ventral patterning during early embryogenesis, through the regulation of gene expression. Moreover, DSP1 is implicated in various cellular processes, including cell fate determination and tissue differentiation, which are essential for embryonic development. While the function of DSP1 in embryonic development has been relatively well-studied, its role in the adult Drosophila brain remains less understood. In this study, we investigated the role of DSP1 in the brain by using neuronal-specific DSP1 overexpression flies. We observed that climbing ability and life span are decreased in DSP1-overexpressed flies. Furthermore, these flies demonstrated neuromuscular junction (NMJ) defect, reduced eye size and a decrease in tyrosine hydroxylase (TH)-positive neurons, indicating neuronal toxicity induced by DSP1 overexpression. Our data suggest that DSP1 overexpression leads to neuronal dysfunction and toxicity, positioning DSP1 as a potential therapeutic target for neurodegenerative diseases.


Asunto(s)
Proteínas de Drosophila , Drosophila melanogaster , Degeneración Nerviosa , Unión Neuromuscular , Neuronas , Fenotipo , Animales , Drosophila melanogaster/genética , Drosophila melanogaster/metabolismo , Proteínas de Drosophila/metabolismo , Proteínas de Drosophila/genética , Ojo/patología , Longevidad/genética , Degeneración Nerviosa/patología , Degeneración Nerviosa/genética , Unión Neuromuscular/metabolismo , Unión Neuromuscular/patología , Neuronas/metabolismo , Neuronas/patología , Factores de Transcripción/metabolismo , Factores de Transcripción/genética , Tirosina 3-Monooxigenasa/metabolismo , Tirosina 3-Monooxigenasa/genética
11.
Am J Pathol ; 194(9): 1752-1763, 2024 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-38885925

RESUMEN

Local tetanus develops when limited amounts of tetanus neurotoxin (TeNT) are released by Clostridium tetani generated from spores inside a necrotic wound. Within days, a spastic paralysis restricted to the muscles of the affected anatomical area develops. This paralysis follows the retrograde transport of TeNT inside the axons of motoneurons and its uptake by inhibitory interneurons with cleavage of a vesicle-associated membrane protein required for neurotransmitter release. Consequently, incontrollable excitation of motoneurons causes contractures of innervated muscles and leads to local spastic paralysis. Here, the initial events occurring close to the site of TeNT release were investigated in a mouse model of local tetanus. A peripheral flaccid paralysis was found to occur, before or concurrent to the spastic paralysis. At variance from the confined TeNT proteolytic activity taking place within motor neuron terminals, central protein cleavage was detected within inhibitory interneurons controlling motor neuron efferents innervating muscle groups distant from the site of TeNT release. These results indicate peripheral activity of TeNT in tetanus and explains why the spastic paralysis observed in local tetanus, although confined to single limbs, generally affects multiple muscles. The initial TeNT neuroparalytic activity can be detected by measuring the compound muscle action potential, providing a very early diagnosis and therapy, thus preventing the ensuing life-threatening generalized tetanus.


Asunto(s)
Unión Neuromuscular , Parálisis , Toxina Tetánica , Tétanos , Animales , Tétanos/metabolismo , Tétanos/complicaciones , Toxina Tetánica/metabolismo , Ratones , Unión Neuromuscular/metabolismo , Unión Neuromuscular/patología , Unión Neuromuscular/efectos de los fármacos , Parálisis/metabolismo , Neuronas Motoras/metabolismo , Neuronas Motoras/patología , Interneuronas/metabolismo , Ratones Endogámicos C57BL , Modelos Animales de Enfermedad , Femenino
12.
Dis Model Mech ; 17(8)2024 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-38903011

RESUMEN

Pathogenic variants in GFPT1, encoding a key enzyme to synthesize UDP-N-acetylglucosamine (UDP-GlcNAc), cause congenital myasthenic syndrome (CMS). We made a knock-in (KI) mouse model carrying a frameshift variant in Gfpt1 exon 9, simulating that found in a patient with CMS. As Gfpt1 exon 9 is exclusively expressed in striated muscles, Gfpt1-KI mice were deficient for Gfpt1 only in skeletal muscles. In Gfpt1-KI mice, (1) UDP-HexNAc, CMP-NeuAc and protein O-GlcNAcylation were reduced in skeletal muscles; (2) aged Gfpt1-KI mice showed poor exercise performance and abnormal neuromuscular junction structures; and (3) markers of the unfolded protein response (UPR) were elevated in skeletal muscles. Denervation-mediated enhancement of endoplasmic reticulum (ER) stress in Gfpt1-KI mice facilitated protein folding, ubiquitin-proteasome degradation and apoptosis, whereas autophagy was not induced and protein aggregates were markedly increased. Lack of autophagy was accounted for by enhanced degradation of FoxO1 by increased Xbp1-s/u proteins. Similarly, in Gfpt1-silenced C2C12 myotubes, ER stress exacerbated protein aggregates and activated apoptosis, but autophagy was attenuated. In both skeletal muscles in Gfpt1-KI mice and Gfpt1-silenced C2C12 myotubes, maladaptive UPR failed to eliminate protein aggregates and provoked apoptosis.


Asunto(s)
Autofagia , Estrés del Retículo Endoplásmico , Músculo Esquelético , Pliegue de Proteína , Respuesta de Proteína Desplegada , Animales , Ratones , Apoptosis , Proteína Forkhead Box O1/metabolismo , Técnicas de Sustitución del Gen , Fibras Musculares Esqueléticas/metabolismo , Fibras Musculares Esqueléticas/patología , Músculo Esquelético/metabolismo , Músculo Esquelético/patología , Unión Neuromuscular/metabolismo , Unión Neuromuscular/patología , Especificidad de Órganos , Complejo de la Endopetidasa Proteasomal/metabolismo , Agregado de Proteínas , Proteína 1 de Unión a la X-Box/metabolismo
13.
Mol Neurodegener ; 19(1): 50, 2024 Jun 20.
Artículo en Inglés | MEDLINE | ID: mdl-38902734

RESUMEN

BACKGROUND: The key pathological signature of ALS/ FTLD is the mis-localization of endogenous TDP-43 from the nucleus to the cytoplasm. However, TDP-43 gain of function in the cytoplasm is still poorly understood since TDP-43 animal models recapitulating mis-localization of endogenous TDP-43 from the nucleus to the cytoplasm are missing. METHODS: CRISPR/Cas9 technology was used to generate a zebrafish line (called CytoTDP), that mis-locates endogenous TDP-43 from the nucleus to the cytoplasm. Phenotypic characterization of motor neurons and the neuromuscular junction was performed by immunostaining, microglia were immunohistochemically localized by whole-mount tissue clearing and muscle ultrastructure was analyzed by scanning electron microscopy. Behavior was investigated by video tracking and quantitative analysis of swimming parameters. RNA sequencing was used to identify mis-regulated pathways with validation by molecular analysis. RESULTS: CytoTDP fish have early larval phenotypes resembling clinical features of ALS such as progressive motor defects, neurodegeneration and muscle atrophy. Taking advantage of zebrafish's embryonic development that solely relys on yolk usage until 5 days post fertilization, we demonstrated that microglia proliferation and activation in the hypothalamus is independent from food intake. By comparing CytoTDP to a previously generated TDP-43 knockout line, transcriptomic analyses revealed that mis-localization of endogenous TDP-43, rather than TDP-43 nuclear loss of function, leads to early onset metabolic dysfunction. CONCLUSIONS: The new TDP-43 model mimics the ALS/FTLD hallmark of progressive motor dysfunction. Our results suggest that functional deficits of the hypothalamus, the metabolic regulatory center, might be the primary cause of weight loss in ALS patients. Cytoplasmic gain of function of endogenous TDP-43 leads to metabolic dysfunction in vivo that are reminiscent of early ALS clinical non-motor metabolic alterations. Thus, the CytoTDP zebrafish model offers a unique opportunity to identify mis-regulated targets for therapeutic intervention early in disease progression.


Asunto(s)
Esclerosis Amiotrófica Lateral , Proteínas de Unión al ADN , Modelos Animales de Enfermedad , Neuronas Motoras , Proteínas de Pez Cebra , Pez Cebra , Animales , Esclerosis Amiotrófica Lateral/metabolismo , Esclerosis Amiotrófica Lateral/patología , Esclerosis Amiotrófica Lateral/genética , Proteínas de Unión al ADN/metabolismo , Proteínas de Unión al ADN/genética , Neuronas Motoras/metabolismo , Neuronas Motoras/patología , Proteínas de Pez Cebra/metabolismo , Proteínas de Pez Cebra/genética , Animales Modificados Genéticamente , Unión Neuromuscular/metabolismo , Unión Neuromuscular/patología
14.
Cells ; 13(10)2024 May 07.
Artículo en Inglés | MEDLINE | ID: mdl-38786016

RESUMEN

The primary neural circuit affected in Amyotrophic Lateral Sclerosis (ALS) patients is the corticospinal motor circuit, originating in upper motor neurons (UMNs) in the cerebral motor cortex which descend to synapse with the lower motor neurons (LMNs) in the spinal cord to ultimately innervate the skeletal muscle. Perturbation of these neural circuits and consequent loss of both UMNs and LMNs, leading to muscle wastage and impaired movement, is the key pathophysiology observed. Despite decades of research, we are still lacking in ALS disease-modifying treatments. In this review, we document the current research from patient studies, rodent models, and human stem cell models in understanding the mechanisms of corticomotor circuit dysfunction and its implication in ALS. We summarize the current knowledge about cortical UMN dysfunction and degeneration, altered excitability in LMNs, neuromuscular junction degeneration, and the non-cell autonomous role of glial cells in motor circuit dysfunction in relation to ALS. We further highlight the advances in human stem cell technology to model the complex neural circuitry and how these can aid in future studies to better understand the mechanisms of neural circuit dysfunction underpinning ALS.


Asunto(s)
Esclerosis Amiotrófica Lateral , Neuronas Motoras , Esclerosis Amiotrófica Lateral/fisiopatología , Esclerosis Amiotrófica Lateral/patología , Humanos , Neuronas Motoras/patología , Neuronas Motoras/fisiología , Animales , Red Nerviosa/fisiopatología , Red Nerviosa/patología , Unión Neuromuscular/fisiopatología , Unión Neuromuscular/patología , Modelos Animales de Enfermedad , Corteza Motora/fisiopatología , Corteza Motora/patología
15.
Skelet Muscle ; 14(1): 10, 2024 May 17.
Artículo en Inglés | MEDLINE | ID: mdl-38760872

RESUMEN

Loss-of-function mutations in MEGF10 lead to a rare and understudied neuromuscular disorder known as MEGF10-related myopathy. There are no treatments for the progressive respiratory distress, motor impairment, and structural abnormalities in muscles caused by the loss of MEGF10 function. In this study, we deployed cellular and molecular assays to obtain additional insights about MEGF10-related myopathy in juvenile, young adult, and middle-aged Megf10 knockout (KO) mice. We found fewer muscle fibers in juvenile and adult Megf10 KO mice, supporting published studies that MEGF10 regulates myogenesis by affecting satellite cell differentiation. Interestingly, muscle fibers do not exhibit morphological hallmarks of atrophy in either young adult or middle-aged Megf10 KO mice. We next examined the neuromuscular junction (NMJ), in which MEGF10 has been shown to concentrate postnatally, using light and electron microscopy. We found early and progressive degenerative features at the NMJs of Megf10 KO mice that include increased postsynaptic fragmentation and presynaptic regions not apposed by postsynaptic nicotinic acetylcholine receptors. We also found perisynaptic Schwann cells intruding into the NMJ synaptic cleft. These findings strongly suggest that the NMJ is a site of postnatal pathology in MEGF10-related myopathy. In support of these cellular observations, RNA-seq analysis revealed genes and pathways associated with myogenesis, skeletal muscle health, and NMJ stability dysregulated in Megf10 KO mice compared to wild-type mice. Altogether, these data provide new and valuable cellular and molecular insights into MEGF10-related myopathy.


Asunto(s)
Modelos Animales de Enfermedad , Ratones Noqueados , Unión Neuromuscular , Animales , Unión Neuromuscular/metabolismo , Unión Neuromuscular/patología , Ratones , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Enfermedades Musculares/genética , Enfermedades Musculares/patología , Enfermedades Musculares/metabolismo , Enfermedades Musculares/fisiopatología , Células de Schwann/metabolismo , Células de Schwann/patología , Músculo Esquelético/metabolismo , Músculo Esquelético/patología , Músculo Esquelético/fisiopatología , Ratones Endogámicos C57BL , Fibras Musculares Esqueléticas/metabolismo , Fibras Musculares Esqueléticas/patología , Masculino
16.
Respir Physiol Neurobiol ; 326: 104282, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-38782084

RESUMEN

Duchenne muscular dystrophy (DMD) is the most common X-linked disease. DMD is caused by a lack of dystrophin, a critical structural protein in striated muscle. Dystrophin deficiency leads to inflammation, fibrosis, and muscle atrophy. Boys with DMD have progressive muscle weakness within the diaphragm that results in respiratory failure in the 2nd or 3rd decade of life. The most common DMD mouse model - the mdx mouse - is not sufficient for evaluating genetic medicines that specifically target the human DMD (hDMD) gene sequence. Therefore, a novel transgenic mouse carrying the hDMD gene with an exon 52 deletion was created (hDMDΔ52;mdx). We characterized the respiratory function and pathology in this model using whole body plethysmography, histology, and immunohistochemistry. At 6-months-old, hDMDΔ52;mdx mice have reduced maximal respiration, neuromuscular junction pathology, and fibrosis throughout the diaphragm, which worsens at 12-months-old. In conclusion, the hDMDΔ52;mdx exhibits moderate respiratory pathology, and serves as a relevant animal model to study the impact of novel genetic therapies, including gene editing, on respiratory function.


Asunto(s)
Modelos Animales de Enfermedad , Ratones Transgénicos , Distrofia Muscular de Duchenne , Animales , Distrofia Muscular de Duchenne/genética , Distrofia Muscular de Duchenne/patología , Distrofia Muscular de Duchenne/fisiopatología , Ratones , Humanos , Masculino , Distrofina/genética , Distrofina/deficiencia , Ratones Endogámicos mdx , Diafragma/fisiopatología , Diafragma/patología , Insuficiencia Respiratoria/etiología , Unión Neuromuscular/patología , Unión Neuromuscular/metabolismo , Ratones Endogámicos C57BL
17.
J Cachexia Sarcopenia Muscle ; 15(4): 1358-1375, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-38646816

RESUMEN

BACKGROUND: Alzheimer's disease (AD) impairs cognitive functions and peripheral systems, including skeletal muscles. The PS19 mouse, expressing the human tau P301S mutation, shows cognitive and muscular pathologies, reflecting the central and peripheral atrophy seen in AD. METHODS: We analysed skeletal muscle morphology and neuromuscular junction (NMJ) through immunohistochemistry and advanced image quantification. A factorial Analysis of Variance assessed muscle weight, NCAM expression, NMJ, myofibre type distribution, cross-sectional areas, expression of single or multiple myosin heavy-chain isoforms, and myofibre grouping in PS19 and wild type (WT) mice over their lifespan (1-12 months). RESULTS: Significant weight differences in extensor digitorum longus (EDL) and soleus muscles between WT and PS19 mice were noted by 7-8 months. For EDL muscle in females, WT weighed 0.0113 ± 0.0005 compared with PS19's 0.0071 ± 0.0008 (P < 0.05), and in males, WT was 0.0137 ± 0.0001 versus PS19's 0.0069 ± 0.0006 (P < 0.005). Similarly, soleus muscle showed significant differences; females (WT: 0.0084 ± 0.0004; PS19: 0.0057 ± 0.0005, P < 0.005) and males (WT: 0.0088 ± 0.0003; PS19: 0.0047 ± 0.0004, P < 0.0001). Analysis of the NMJ in PS19 mice revealed a marked reduction in myofibre innervation at 5 months, with further decline by 10 months. NMJ pre-terminals in PS19 mice became shorter and simpler by 5 months, showing a steep decline by 10 months. Genotype and age strongly influenced muscle NCAM immunoreactivity, denoting denervation as early as 5-6 months in EDL muscle Type II fibres, with earlier effects in soleus muscle Type I and II fibres at 3-4 months. Muscle denervation and subsequent myofibre atrophy were linked to a reduction in Type IIB fibres in the EDL muscle and Type IIA fibres in the soleus muscle, accompanied by an increase in hybrid fibres. The EDL muscle showed Type IIB fibre atrophy with WT females at 1505 ± 110 µm2 versus PS19's 1208 ± 94 µm2, and WT males at 1731 ± 185 µm2 versus PS19's 1227 ± 116 µm2. Similarly, the soleus muscle demonstrated Type IIA fibre atrophy from 5 to 6 months, with WT females at 1194 ± 52 µm2 versus PS19's 858 ± 62 µm2, and WT males at 1257 ± 43 µm2 versus PS19's 1030 ± 55 µm2. Atrophy also affected Type IIX, I + IIA, and IIA + IIX fibres in both muscles. The timeline for both myofibre and overall muscle atrophy in PS19 mice was consistent, indicating a simultaneous decline. CONCLUSIONS: Progressive and accelerated neurogenic sarcopenia may precede and potentially predict cognitive deficits observed in AD.


Asunto(s)
Enfermedad de Alzheimer , Modelos Animales de Enfermedad , Trastornos de la Memoria , Ratones Transgénicos , Músculo Esquelético , Sarcopenia , Animales , Enfermedad de Alzheimer/patología , Enfermedad de Alzheimer/metabolismo , Ratones , Sarcopenia/metabolismo , Sarcopenia/patología , Masculino , Femenino , Trastornos de la Memoria/etiología , Trastornos de la Memoria/metabolismo , Humanos , Músculo Esquelético/patología , Músculo Esquelético/metabolismo , Tauopatías/patología , Tauopatías/metabolismo , Unión Neuromuscular/metabolismo , Unión Neuromuscular/patología , Proteínas tau/metabolismo
18.
Front Immunol ; 15: 1342213, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38605954

RESUMEN

Myasthenia gravis (MG) stands as a perplexing autoimmune disorder affecting the neuromuscular junction, driven by a multitude of antibodies targeting postsynaptic elements. However, the mystery of MG pathogenesis has yet to be completely uncovered, and its heterogeneity also challenges diagnosis and treatment. Growing evidence shows the differential expression of non-coding RNAs (ncRNAs) in MG has played an essential role in the development of MG in recent years. Remarkably, these aberrantly expressed ncRNAs exhibit distinct profiles within diverse clinical subgroups and among patients harboring various antibody types. Furthermore, they have been implicated in orchestrating the production of inflammatory cytokines, perturbing the equilibrium of T helper 1 cells (Th1), T helper 17 cells (Th17), and regulatory T cells (Tregs), and inciting B cells to generate antibodies. Studies have elucidated that certain ncRNAs mirror the clinical severity of MG, while others may hold therapeutic significance, showcasing a propensity to return to normal levels following appropriate treatments or potentially foretelling the responsiveness to immunosuppressive therapies. Notably, the intricate interplay among these ncRNAs does not follow a linear trajectory but rather assembles into a complex network, with competing endogenous RNA (ceRNA) emerging as a prominent hub in some cases. This comprehensive review consolidates the landscape of dysregulated ncRNAs in MG, briefly delineating their pivotal role in MG pathogenesis. Furthermore, it explores their promise as prospective biomarkers, aiding in the elucidation of disease subtypes, assessment of disease severity, monitoring therapeutic responses, and as novel therapeutic targets.


Asunto(s)
Miastenia Gravis , Humanos , Miastenia Gravis/terapia , Miastenia Gravis/tratamiento farmacológico , Células TH1 , Linfocitos T Reguladores , Unión Neuromuscular/patología , Células Th17/patología
19.
Aging Cell ; 23(7): e14156, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38532712

RESUMEN

Neuromuscular junction (NMJ) degeneration is one of pathological factors of sarcopenia. Low-magnitude high-frequency vibration (LMHFV) was reported effective in alleviating the sarcopenia progress. However, no previous study has investigated treatment effects of LMHFV targeting NMJ degeneration in sarcopenia. We first compared morphological differences of NMJ between sarcopenic and non-sarcopenic subjects, as well as young and old C57BL/6 mice. We then systematically characterized the age-related degeneration of NMJ in SAMP8 against its control strain, SAMR1 mice, from 3 to 12 months old. We also investigated effects of LMHFV in SAMP8 on the maintenance of NMJ during the onset of sarcopenia with respect to the Agrin-LRP4-MuSK-Dok7 pathway and investigated the mechanism related to ERK1/2 signaling. We observed sarcopenic/old NMJ presented increased acetylcholine receptors (AChRs) cluster fragmentation and discontinuity than non-sarcopenic/young NMJ. In SAMP8, NMJ degeneration (morphologically at 6 months and functionally at 8 months) was observed associated with the sarcopenia onset (10 months). SAMR1 showed improved NMJ morphology and function compared with SAMP8 at 10 months. Skeletal muscle performance was improved at Month 4 post-LMHFV treatment. Vibration group presented improved NMJ function at Months 2 and 6 posttreatment, accompanied with alleviated morphological degeneration at Month 4 posttreatment. LMHFV increased Dok7 expression at Month 4 posttreatment. In vitro, LMHFV could promote AChRs clustering in myotubes by increasing Dok7 expression through suppressing ERK1/2 phosphorylation. In conclusion, NMJ degeneration was observed associated with the sarcopenia onset in SAMP8. LMHFV may attenuate NMJ degeneration and sarcopenia progression by increasing Dok7 expression through suppressing ERK1/2 phosphorylation.


Asunto(s)
Ratones Endogámicos C57BL , Unión Neuromuscular , Sarcopenia , Vibración , Sarcopenia/patología , Sarcopenia/metabolismo , Animales , Vibración/uso terapéutico , Unión Neuromuscular/metabolismo , Unión Neuromuscular/patología , Ratones , Masculino , Humanos , Envejecimiento , Femenino
20.
Bone ; 184: 117086, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38552893

RESUMEN

PURPOSE: Mitofusin 2 (Mfn2) is one of two mitofusins involved in regulating mitochondrial size, shape and function, including mitophagy, an important cellular mechanism to limit oxidative stress. Reduced expression of Mfn2 has been associated with impaired osteoblast differentiation and function and a reduction in the number of viable osteocytes in bone. We hypothesized that the genetic absence of Mfn2 in these cells would increase their susceptibility to aging-associated metabolic stress, leading to a progressive impairment in skeletal homeostasis over time. METHODS: Mfn2 was selectively deleted in vivo at three different stages of osteoblast lineage commitment by crossing mice in which the Mfn2 gene was floxed with transgenic mice expressing Cre under the control of the promoter for Osterix (OSX), collagen1a1, or DMP1 (Dentin Matrix Acidic Phosphoprotein 1). RESULTS: Mice in which Mfn2 was deleted using DMP1-cre demonstrated a progressive and dramatic decline in bone mineral density (BMD) beginning at 10 weeks of age (n = 5 for each sex and each genotype from age 10 to 20 weeks). By 15 weeks, there was evidence for a functional decline in muscle performance as assessed using a rotarod apparatus (n = 3; 2 males/ 1 female for each genotype), accompanied by a decline in lean body mass. A marked reduction in trabecular bone mass was evident on bone histomorphometry, and biomechanical testing at 25 weeks (k/o: 2 male/1 female, control 2 male/2 female) revealed severely impaired femur strength. Extensive regional myofiber atrophy and degeneration was observed on skeletal muscle histology. Electron microscopy showed progressive disruption of cellular architecture, with disorganized sarcomeres and a bloated mitochondrial reticulum. There was also evidence of neurodegeneration within the ventral horn and roots of the lumbar spinal cord, which was accompanied by myelin loss and myofiber atrophy. Deletion of Mfn2 using OSX-cre or Col1a1-cre did not result in a musculoskeletal phenotype. Where possible, male and female animals were analyzed separately, but small numbers of animals in each group limited statistical power. For other outcomes, where sex was not considered, small sample sizes might still limit the strength of the observation. CONCLUSION: Despite known functional overlap of Mfn1 and Mfn2 in some tissues, and their co-expression in bone, muscle and spinal cord, deletion of Mfn2 using the 8 kB DMP1 promoter uncovered an important non-redundant role for Mfn2 in maintaining the neuromuscular/bone axis.


Asunto(s)
Densidad Ósea , GTP Fosfohidrolasas , Animales , Femenino , GTP Fosfohidrolasas/metabolismo , GTP Fosfohidrolasas/genética , Masculino , Ratones , Densidad Ósea/genética , Densidad Ósea/fisiología , Ratones Transgénicos , Músculo Esquelético/metabolismo , Músculo Esquelético/patología , Huesos/patología , Huesos/metabolismo , Unión Neuromuscular/metabolismo , Unión Neuromuscular/patología , Osteoblastos/metabolismo , Proteínas Mitocondriales/metabolismo , Proteínas Mitocondriales/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA