Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 5.739
Filter
1.
Int J Mol Sci ; 25(14)2024 Jul 18.
Article in English | MEDLINE | ID: mdl-39063088

ABSTRACT

This study investigated the effects of cilostazol on motor dysfunction, spinal motor neuron abnormalities, and schwannopathy in rats with diabetes. Diabetes mellitus (DM) was induced in rats via femoral intravenous streptozotocin (STZ) injection (60 mg/kg). After successful DM induction, cilostazol was administered on day 15 via oral gavage (100 mg/kg/day) for 6 weeks until sacrifice. Behavioral assays, including motor function, were performed weekly. The sciatic nerve, L5 spinal cord, and spinal ventral root were collected to evaluate the expression of the glial fibrillary acidic protein (GFAP), myelin protein zero (P0), and choline acetyltransferase (ChAT) by immunofluorescence and Western blotting. DM rats displayed decreased running speeds, running distances, and toe spread but increased foot pressure. In addition, loss of non-myelinating Schwann cells and myelin sheaths was observed in the sciatic nerve and L5 spinal ventral root. Reduced numbers of motor neurons were also found in the L5 spinal ventral horn. Cilostazol administration significantly potentiated running speed and distance; increased hind paw toe spread; and decreased foot pressure. In the sciatic nerve and L5 spinal ventral root, cilostazol treatment significantly improved non-myelinated Schwann cells and increased myelin mass. ChAT expression in motor neurons in the spinal ventral horn was improved, but not significantly. Cilostazol administration may protect sensorimotor function in diabetic rats.


Subject(s)
Cilostazol , Diabetes Mellitus, Experimental , Schwann Cells , Sciatic Nerve , Animals , Cilostazol/pharmacology , Cilostazol/therapeutic use , Schwann Cells/drug effects , Schwann Cells/metabolism , Diabetes Mellitus, Experimental/drug therapy , Diabetes Mellitus, Experimental/metabolism , Rats , Male , Sciatic Nerve/drug effects , Sciatic Nerve/metabolism , Choline O-Acetyltransferase/metabolism , Rats, Sprague-Dawley , Spinal Cord/drug effects , Spinal Cord/metabolism , Motor Neurons/drug effects , Motor Neurons/metabolism , Glial Fibrillary Acidic Protein/metabolism , Myelin P0 Protein/metabolism , Streptozocin
2.
Int J Mol Sci ; 25(13)2024 Jun 27.
Article in English | MEDLINE | ID: mdl-39000168

ABSTRACT

Amyotrophic lateral sclerosis (ALS) is an extremely complex neurodegenerative disease involving different cell types, but motoneuronal loss represents its main pathological feature. Moreover, compensatory plastic changes taking place in parallel to neurodegeneration are likely to affect the timing of ALS onset and progression and, interestingly, they might represent a promising target for disease-modifying treatments. Therefore, a simplified animal model mimicking motoneuronal loss without the other pathological aspects of ALS has been established by means of intramuscular injection of cholera toxin-B saporin (CTB-Sap), which is a targeted neurotoxin able to kill motoneurons by retrograde suicide transport. Previous studies employing the mouse CTB-Sap model have proven that spontaneous motor recovery is possible after a subtotal removal of a spinal motoneuronal pool. Although these kinds of plastic changes are not enough to counteract the functional effects of the progressive motoneuron degeneration, it would nevertheless represent a promising target for treatments aiming to postpone ALS onset and/or delay disease progression. Herein, the mouse CTB-Sap model has been used to test the efficacy of mitochondrial division inhibitor 1 (Mdivi-1) as a tool to counteract the CTB-Sap toxicity and/or to promote neuroplasticity. The homeostasis of mitochondrial fission/fusion dynamics is indeed important for cell integrity, and it could be affected during neurodegeneration. Lesioned mice were treated with Mdivi-1 and then examined by a series of behavioral test and histological analyses. The results have shown that the drug may be capable of reducing functional deficits after the lesion and promoting synaptic plasticity and neuroprotection, thus representing a putative translational approach for motoneuron disorders.


Subject(s)
Amyotrophic Lateral Sclerosis , Disease Models, Animal , Mitochondrial Dynamics , Motor Neurons , Animals , Motor Neurons/drug effects , Motor Neurons/metabolism , Motor Neurons/pathology , Mitochondrial Dynamics/drug effects , Mice , Amyotrophic Lateral Sclerosis/metabolism , Amyotrophic Lateral Sclerosis/drug therapy , Amyotrophic Lateral Sclerosis/pathology , Cholera Toxin/metabolism , Saporins , Quinazolinones/pharmacology , Neuronal Plasticity/drug effects , Male , Mitochondria/drug effects , Mitochondria/metabolism
3.
Cell Mol Life Sci ; 81(1): 315, 2024 Jul 27.
Article in English | MEDLINE | ID: mdl-39066803

ABSTRACT

Chemotherapy-induced peripheral neuropathy (CIPN) is a disabling side effect of cancer chemotherapy that can often limit treatment options for cancer patients or have life-long neurodegenerative consequences that reduce the patient's quality of life. CIPN is caused by the detrimental actions of various chemotherapeutic agents on peripheral axons. Currently, there are no approved preventative measures or treatment options for CIPN, highlighting the need for the discovery of novel therapeutics and improving our understanding of disease mechanisms. In this study, we utilized human-induced pluripotent stem cell (hiPSC)-derived motor neurons as a platform to mimic axonal damage after treatment with vincristine, a chemotherapeutic used for the treatment of breast cancers, osteosarcomas, and leukemia. We screened a total of 1902 small molecules for neuroprotective properties in rescuing vincristine-induced axon growth deficits. From our primary screen, we identified 38 hit compounds that were subjected to secondary dose response screens. Six compounds showed favorable pharmacological profiles - AZD7762, A-674563, Blebbistatin, Glesatinib, KW-2449, and Pelitinib, all novel neuroprotectants against vincristine toxicity to neurons. In addition, four of these six compounds also showed efficacy against vincristine-induced growth arrest in human iPSC-derived sensory neurons. In this study, we utilized high-throughput screening of a large library of compounds in a therapeutically relevant assay. We identified several novel compounds that are efficacious in protecting different neuronal subtypes from the toxicity induced by a common chemotherapeutic agent, vincristine which could have therapeutic potential in the clinic.


Subject(s)
Induced Pluripotent Stem Cells , Neuroprotective Agents , Vincristine , Vincristine/pharmacology , Humans , Induced Pluripotent Stem Cells/drug effects , Induced Pluripotent Stem Cells/metabolism , Induced Pluripotent Stem Cells/cytology , Neuroprotective Agents/pharmacology , Motor Neurons/drug effects , Motor Neurons/pathology , Motor Neurons/metabolism , Axons/drug effects , Axons/metabolism , Axons/pathology , Neurons/drug effects , Neurons/metabolism , Neurons/pathology , Cells, Cultured , Peripheral Nervous System Diseases/chemically induced , Peripheral Nervous System Diseases/pathology , Peripheral Nervous System Diseases/drug therapy
4.
PLoS One ; 19(6): e0305173, 2024.
Article in English | MEDLINE | ID: mdl-38875300

ABSTRACT

Chlorpyrifos is an organophosphate pesticide associated with numerous health effects including motor performance decrements. While many studies have focused on the health effects following acute chlorpyrifos poisonings, almost no studies have examined the effects on motoneurons following occupational-like exposures. The main objective of this study was to examine the broad effects of repeated occupational-like chlorpyrifos exposures on spinal motoneuron soma size relative to motor activity. To execute our objective, adult rats were exposed to chlorpyrifos via oral gavage once a day, five days a week for two weeks. Chlorpyrifos exposure effects were assessed either three days or two months following the last exposure. Three days following the last repeated chlorpyrifos exposure, there were transient effects in open-field motor activity and plasma cholinesterase activity levels. Two months following the chlorpyrifos exposures, there were delayed effects in sensorimotor gating, pro-inflammatory cytokines and spinal lumbar motoneuron soma morphology. Overall, these results offer support that subacute repeated occupational-like chlorpyrifos exposures have both short-term and longer-term effects in motor activity, inflammation, and central nervous system mechanisms.


Subject(s)
Chlorpyrifos , Motor Activity , Motor Neurons , Animals , Chlorpyrifos/toxicity , Motor Neurons/drug effects , Motor Neurons/pathology , Rats , Male , Motor Activity/drug effects , Insecticides/toxicity , Spinal Cord/drug effects , Spinal Cord/pathology , Rats, Sprague-Dawley , Lumbosacral Region , Cholinesterases/metabolism , Cholinesterases/blood , Cholinesterase Inhibitors/toxicity
5.
CNS Neurosci Ther ; 30(6): e14692, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38872258

ABSTRACT

AIM: Amyotrophic lateral sclerosis (ALS) is a severe neurodegenerative disease characterized by progressive death of upper and lower motor neurons, leading to generalized muscle atrophy, paralysis, and even death. Mitochondrial damage and neuroinflammation play key roles in the pathogenesis of ALS. In the present study, the efficacy of A-1, a derivative of arctigenin with AMP-activated protein kinase (AMPK) and silent information regulator 1 (SIRT1) activation for ALS, was investigated. METHODS: A-1 at 33.3 mg/kg was administrated in SOD1G93A transgenic mice orally from the 13th week for a 6-week treatment period. Motor ability was assessed before terminal anesthesia. Muscle atrophy and fibrosis, motor neurons, astrocytes, and microglia in the spinal cord were evaluated by H&E, Masson, Sirius Red, Nissl, and immunohistochemistry staining. Protein expression was detected with proteomics analysis, Western blotting, and ELISA. Mitochondrial adenosine triphosphate (ATP) and malondialdehyde (MDA) levels were measured using an assay kit. RESULTS: A-1 administration in SOD1G93A mice enhanced mobility, decreased skeletal muscle atrophy and fibrosis, mitigated loss of spinal motor neurons, and reduced glial activation. Additionally, A-1 treatment improved mitochondrial function, evidenced by elevated ATP levels and increased expression of key mitochondrial-related proteins. The A-1 treatment group showed decreased levels of IL-1ß, pIκBα/IκBα, and pNF-κB/NF-κB. CONCLUSIONS: A-1 treatment reduced motor neuron loss, improved gastrocnemius atrophy, and delayed ALS progression through the AMPK/SIRT1/PGC-1α pathway, which promotes mitochondrial biogenesis. Furthermore, the AMPK/SIRT1/IL-1ß/NF-κB pathway exerted neuroprotective effects by reducing neuroinflammation. These findings suggest A-1 as a promising therapeutic approach for ALS.


Subject(s)
AMP-Activated Protein Kinases , Amyotrophic Lateral Sclerosis , Furans , Interleukin-1beta , Mice, Transgenic , NF-kappa B , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha , Sirtuin 1 , Animals , Sirtuin 1/metabolism , Mice , NF-kappa B/metabolism , AMP-Activated Protein Kinases/metabolism , Furans/pharmacology , Amyotrophic Lateral Sclerosis/drug therapy , Amyotrophic Lateral Sclerosis/pathology , Amyotrophic Lateral Sclerosis/metabolism , Interleukin-1beta/metabolism , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/metabolism , Lignans/pharmacology , Lignans/therapeutic use , Signal Transduction/drug effects , Superoxide Dismutase-1/genetics , Superoxide Dismutase-1/metabolism , Male , Motor Neurons/drug effects , Motor Neurons/pathology , Motor Neurons/metabolism , Spinal Cord/drug effects , Spinal Cord/pathology , Spinal Cord/metabolism
6.
PLoS One ; 19(6): e0306099, 2024.
Article in English | MEDLINE | ID: mdl-38917189

ABSTRACT

Orexin-mediated stimulation of orexin receptors 1/2 (OX[1/2]R) may stimulate the diaphragm and genioglossus muscle via activation of inspiratory neurons in the pre-Bötzinger complex, which are critical for the generation of inspiratory rhythm, and phrenic and hypoglossal motoneurons. Herein, we assessed the effects of OX2R-selective agonists TAK-925 (danavorexton) and OX-201 on respiratory function. In in vitro electrophysiologic analyses using rat medullary slices, danavorexton and OX-201 showed tendency and significant effect, respectively, in increasing the frequency of inspiratory synaptic currents of inspiratory neurons in the pre-Bötzinger complex. In rat medullary slices, both danavorexton and OX-201 significantly increased the frequency of inspiratory synaptic currents of hypoglossal motoneurons. Danavorexton and OX-201 also showed significant effect and tendency, respectively, in increasing the frequency of burst activity recorded from the cervical (C3-C5) ventral root, which contains axons of phrenic motoneurons, in in vitro electrophysiologic analyses from rat isolated brainstem-spinal cord preparations. Electromyogram recordings revealed that intravenous administration of OX-201 increased burst frequency of the diaphragm and burst amplitude of the genioglossus muscle in isoflurane- and urethane-anesthetized rats, respectively. In whole-body plethysmography analyses, oral administration of OX-201 increased respiratory activity in free-moving mice. Overall, these results suggest that OX2R-selective agonists enhance respiratory function via activation of the diaphragm and genioglossus muscle through stimulation of inspiratory neurons in the pre-Bötzinger complex, and phrenic and hypoglossal motoneurons. OX2R-selective agonists could be promising drugs for various conditions with respiratory dysfunction.


Subject(s)
Diaphragm , Hypoglossal Nerve , Motor Neurons , Orexin Receptors , Phrenic Nerve , Animals , Diaphragm/drug effects , Diaphragm/innervation , Diaphragm/physiology , Motor Neurons/drug effects , Motor Neurons/physiology , Orexin Receptors/agonists , Orexin Receptors/metabolism , Rats , Phrenic Nerve/drug effects , Phrenic Nerve/physiology , Mice , Male , Hypoglossal Nerve/drug effects , Hypoglossal Nerve/physiology , Rats, Sprague-Dawley , Inhalation , Medulla Oblongata/drug effects , Medulla Oblongata/physiology , Isoquinolines , Pyridines
7.
Synapse ; 78(4): e22304, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38896000

ABSTRACT

The goal of this report is to explore how K2P channels modulate axonal excitability by using the crayfish ventral superficial flexor preparation. This preparation allows for simultaneous recording of motor nerve extracellular action potentials (eAP) and intracellular excitatory junctional potential (EJP) from a muscle fiber. Previous pharmacological studies have demonstrated the presence of K2P-like channels in crayfish. Fluoxetine (50 µM) was used to block K2P channels in this study. The blocker caused a gradual decline, and eventually complete block, of motor axon action potentials. At an intermediate stage of the block, when the peak-to-peak amplitude of eAP decreased to ∼60%-80% of the control value, the amplitude of the initial positive component of eAP declined at a faster rate than that of the negative peak representing sodium influx. Furthermore, the second positive peak following this sodium influx, which corresponds to the after-hyperpolarizing phase of intracellularly recorded action potentials (iAP), became larger during the intermediate stage of eAP block. Finally, EJP recorded simultaneously with eAP showed no change in amplitude, but did show a significant increase in synaptic delay. These changes in eAP shape and EJP delay are interpreted as the consequence of depolarized resting membrane potential after K2P channel block. In addition to providing insights to possible functions of K2P channels in unmyelinated axons, results presented here also serve as an example of how changes in eAP shape contain information that can be used to infer alterations in intracellular events. This type of eAP-iAP cross-inference is valuable for gaining mechanistic insights here and may also be applicable to other model systems.


Subject(s)
Action Potentials , Astacoidea , Axons , Fluoxetine , Motor Neurons , Animals , Astacoidea/drug effects , Astacoidea/physiology , Fluoxetine/pharmacology , Action Potentials/drug effects , Action Potentials/physiology , Motor Neurons/drug effects , Motor Neurons/physiology , Axons/drug effects , Axons/physiology
8.
Neurobiol Dis ; 199: 106576, 2024 Sep.
Article in English | MEDLINE | ID: mdl-38914173

ABSTRACT

Variability in disease onset and progression is a hallmark of amyotrophic lateral sclerosis (ALS), both in sporadic and genetic forms. Recently, we found that SOD1-G93A transgenic mice expressing the same amount of mutant SOD1 but with different genetic backgrounds, C57BL/6JOlaHsd and 129S2/SvHsd, show slow and rapid muscle wasting and disease progression, respectively. Here, we investigated the different molecular mechanisms underlying muscle atrophy. Although both strains showed similar denervation-induced degradation of muscle proteins, only the rapidly progressing mice exhibited early and sustained STAT3 activation that preceded atrophy in gastrocnemius muscle. We therefore investigated the therapeutic potential of sunitinib, a tyrosine kinase inhibitor known to inhibit STAT3 and prevent cancer-induced muscle wasting. Although sunitinib treatment reduced STAT3 activation in the gastrocnemius muscle and lumbar spinal cord, it did not preserve spinal motor neurons, improve neuromuscular impairment, muscle atrophy and disease progression in the rapidly progressing SOD1-G93A mice. Thus, the effect of sunitinib is not equally positive in different diseases associated with muscle wasting. Moreover, given the complex role of STAT3 in the peripheral and central compartments of the neuromuscular system, the present study suggests that its broad inhibition may lead to opposing effects, ultimately preventing a potential positive therapeutic action in ALS.


Subject(s)
Amyotrophic Lateral Sclerosis , Disease Models, Animal , Indoles , Mice, Inbred C57BL , Mice, Transgenic , Muscle, Skeletal , Pyrroles , STAT3 Transcription Factor , Spinal Cord , Sunitinib , Animals , Amyotrophic Lateral Sclerosis/metabolism , Amyotrophic Lateral Sclerosis/drug therapy , Amyotrophic Lateral Sclerosis/pathology , Sunitinib/pharmacology , Muscle, Skeletal/drug effects , Muscle, Skeletal/metabolism , Muscle, Skeletal/pathology , STAT3 Transcription Factor/metabolism , STAT3 Transcription Factor/antagonists & inhibitors , Indoles/pharmacology , Mice , Spinal Cord/metabolism , Spinal Cord/drug effects , Spinal Cord/pathology , Pyrroles/pharmacology , Superoxide Dismutase/metabolism , Superoxide Dismutase/genetics , Muscular Atrophy/metabolism , Muscular Atrophy/pathology , Motor Neurons/drug effects , Motor Neurons/metabolism , Motor Neurons/pathology , Disease Progression
9.
Int J Pharm ; 659: 124198, 2024 Jun 25.
Article in English | MEDLINE | ID: mdl-38816263

ABSTRACT

Autophagy, an intracellular degradation system, plays a vital role in protecting cells by clearing damaged organelles, pathogens, and protein aggregates. Autophagy upregulation through pharmacological interventions has gained significant attention as a potential therapeutic avenue for proteinopathies. Here, we report the development of an autophagy-inducing peptide (BCN4) derived from the Beclin 1 protein, the master regulator of autophagy. To deliver the BCN4 into cells and the central nervous system (CNS), it was conjugated to our previously developed cell and blood-brain barrier-penetrating peptide (CPP). CPP-BCN4 significantly upregulated autophagy and reduced protein aggregates in motor neuron (MN)-like cells. Moreover, its systemic administration in a reporter mouse model of autophagy resulted in a significant increase in autophagy activity in the spinal MNs. Therefore, this novel autophagy-inducing peptide with a demonstrated ability to upregulate autophagy in the CNS has significant potential for the treatment of various neurodegenerative diseases with protein aggregates as a characteristic feature.


Subject(s)
Autophagy , Beclin-1 , Motor Neurons , Up-Regulation , Animals , Autophagy/drug effects , Beclin-1/metabolism , Motor Neurons/drug effects , Mice , Up-Regulation/drug effects , Spinal Cord/drug effects , Spinal Cord/metabolism , Peptides/pharmacology , Peptides/administration & dosage , Peptides/chemistry , Cell-Penetrating Peptides/administration & dosage , Cell-Penetrating Peptides/chemistry , Humans , Male , Protein Aggregates/drug effects
10.
Physiol Behav ; 282: 114585, 2024 Aug 01.
Article in English | MEDLINE | ID: mdl-38762195

ABSTRACT

We investigated the effects of one-week quercetin ingestion on motor unit (MU) behavior and muscle contractile properties before, during, and after a single session of resistance exercise in older adults. Twenty-four older adults were divided into two groups: those receiving quercetin glycosides (QUE) or placebo (PLA), and they performed a single session of resistance exercise. MU behavior before and during resistance exercise and electrically elicited contraction before and after resistance exercise were measured (Day 1), and the same measurements were conducted again after 7 days of placebo or quercetin glycoside ingestion (Day 8). The MU recruitment threshold (RT) was decreased (p < 0.001, 25.6 ± 10.1 to 23.6 ± 9.5 %MVC) and the exerted force normalized by the MU firing rate (FR) was increased (p = 0.003, 1.13 ± 0.24 to 1.18 ± 0.22 %MVC/pps) from Days 1 to 8, respectively, in QUE but not PLA (p = 0.263, 22.6 ± 11.9 to 21.9 ± 11.6 %MVC; p = 0.713, 1.09 ± 0.20 to 1.10 ± 0.19 %MVC/pps, respectively). On Day 1, a significant correlation between MURT and%change in MUFR from the first to last contractions during the resistance exercise was observed in both groups (QUE: p = 0.009, rs = 0.308; PLA: p < 0.001, rs = 0.403). On Day 8 %change in MUFR was negatively correlated with MURT in QUE (p = 0.044, rs = -0.251), but there was no significant correlation in PLA (p = 0.844). There was no difference in electrically elicited contraction before and after the resistance exercise between QUE and PLA (p < 0.05). These results suggest that one-week quercetin ingestion in older adults lowered MURT and led to greater fatigue in MU with higher RT than with lower RT during resistance training.


Subject(s)
Muscle, Skeletal , Quercetin , Recruitment, Neurophysiological , Resistance Training , Humans , Quercetin/pharmacology , Quercetin/administration & dosage , Male , Aged , Female , Recruitment, Neurophysiological/drug effects , Recruitment, Neurophysiological/physiology , Muscle, Skeletal/drug effects , Muscle, Skeletal/physiology , Muscle Contraction/drug effects , Muscle Contraction/physiology , Double-Blind Method , Motor Neurons/drug effects , Motor Neurons/physiology , Electromyography/drug effects , Electric Stimulation , Antioxidants/administration & dosage , Antioxidants/pharmacology , Exercise/physiology
11.
Mol Ther ; 32(7): 2176-2189, 2024 Jul 03.
Article in English | MEDLINE | ID: mdl-38734896

ABSTRACT

The disassembly of the neuromuscular junction (NMJ) is an early event in amyotrophic lateral sclerosis (ALS), ultimately leading to motor dysfunction and lethal respiratory paralysis. The hexanucleotide GGGGCC repeat expansion in the C9orf72 gene is the most common genetic mutation, and the dipeptide repeat (DPR) proteins have been shown to cause neurodegeneration. While no drugs can treat ALS patients efficiently, new treatment strategies are urgently needed. Here, we report that a MuSK agonist antibody alleviates poly-PR-induced NMJ deficits in C9orf72-ALS mice. The HB9-PRF/F mice, which express poly-PR proteins in motor neurons, exhibited impaired motor behavior and NMJ deficits. Mechanistically, poly-PR proteins interacted with Agrin to disrupt the interaction between Agrin and Lrp4, leading to attenuated activation of MuSK. Treatment with a MuSK agonist antibody rescued NMJ deficits, and extended the lifespan of C9orf72-ALS mice. Moreover, impaired NMJ transmission was observed in C9orf72-ALS patients. These findings identify the mechanism by which poly-PR proteins attenuate MuSK activation and NMJ transmission, highlighting the potential of promoting MuSK activation with an agonist antibody as a therapeutic strategy to protect NMJ function and prolong the lifespan of ALS patients.


Subject(s)
Amyotrophic Lateral Sclerosis , C9orf72 Protein , Disease Models, Animal , Neuromuscular Junction , Receptor Protein-Tyrosine Kinases , Animals , Neuromuscular Junction/metabolism , Neuromuscular Junction/drug effects , Mice , Amyotrophic Lateral Sclerosis/genetics , Amyotrophic Lateral Sclerosis/metabolism , Amyotrophic Lateral Sclerosis/drug therapy , C9orf72 Protein/genetics , C9orf72 Protein/metabolism , Humans , Receptor Protein-Tyrosine Kinases/metabolism , Receptor Protein-Tyrosine Kinases/genetics , Longevity/drug effects , Motor Neurons/metabolism , Motor Neurons/drug effects , Agrin/metabolism , Agrin/genetics , Mice, Transgenic , Antibodies/pharmacology , Receptors, Cholinergic/metabolism , Receptors, Cholinergic/genetics , LDL-Receptor Related Proteins/metabolism , LDL-Receptor Related Proteins/genetics
12.
Nat Commun ; 15(1): 4120, 2024 May 15.
Article in English | MEDLINE | ID: mdl-38750052

ABSTRACT

5q-associated spinal muscular atrophy (SMA) is a motoneuron disease caused by mutations in the survival motor neuron 1 (SMN1) gene. Adaptive immunity may contribute to SMA as described in other motoneuron diseases, yet mechanisms remain elusive. Nusinersen, an antisense treatment, enhances SMN2 expression, benefiting SMA patients. Here we have longitudinally investigated SMA and nusinersen effects on local immune responses in the cerebrospinal fluid (CSF) - a surrogate of central nervous system parenchyma. Single-cell transcriptomics (SMA: N = 9 versus Control: N = 9) reveal NK cell and CD8+ T cell expansions in untreated SMA CSF, exhibiting activation and degranulation markers. Spatial transcriptomics coupled with multiplex immunohistochemistry elucidate cytotoxicity near chromatolytic motoneurons (N = 4). Post-nusinersen treatment, CSF shows unaltered protein/transcriptional profiles. These findings underscore cytotoxicity's role in SMA pathogenesis and propose it as a therapeutic target. Our study illuminates cell-mediated cytotoxicity as shared features across motoneuron diseases, suggesting broader implications.


Subject(s)
Brain , Killer Cells, Natural , Motor Neurons , Muscular Atrophy, Spinal , Oligonucleotides , Humans , Muscular Atrophy, Spinal/drug therapy , Muscular Atrophy, Spinal/pathology , Muscular Atrophy, Spinal/genetics , Motor Neurons/drug effects , Motor Neurons/pathology , Motor Neurons/metabolism , Killer Cells, Natural/immunology , Killer Cells, Natural/drug effects , Brain/pathology , Brain/drug effects , Female , Male , Survival of Motor Neuron 2 Protein/genetics , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/drug effects , Survival of Motor Neuron 1 Protein/genetics , Survival of Motor Neuron 1 Protein/metabolism , Single-Cell Analysis , Cytotoxicity, Immunologic/drug effects , Infant , Child, Preschool , Child , Transcriptome
13.
Cell Stress Chaperones ; 29(3): 359-380, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38570009

ABSTRACT

Protein misfolding and mislocalization are common themes in neurodegenerative disorders, including motor neuron disease, and amyotrophic lateral sclerosis (ALS). Maintaining proteostasis is a crosscutting therapeutic target, including the upregulation of heat shock proteins (HSP) to increase chaperoning capacity. Motor neurons have a high threshold for upregulating stress-inducible HSPA1A, but constitutively express high levels of HSPA8. This study compared the expression of these HSPs in cultured motor neurons expressing three variants linked to familial ALS: TAR DNA binding protein 43 kDa (TDP-43)G348C, fused in sarcoma (FUS)R521G, or superoxide dismutase I (SOD1)G93A. All variants were poor inducers of Hspa1a, and reduced levels of Hspa8 mRNA and protein, indicating multiple compromises in chaperoning capacity. To promote HSP expression, cultures were treated with the putative HSP coinducer, arimoclomol, and class I histone deacetylase inhibitors, to promote active chromatin for transcription, and with the combination. Treatments had variable, often different effects on the expression of Hspa1a and Hspa8, depending on the ALS variant expressed, mRNA distribution (somata and dendrites), and biomarker of toxicity measured (histone acetylation, maintaining nuclear TDP-43 and the neuronal Brm/Brg-associated factor chromatin remodeling complex component Brg1, mitochondrial transport, FUS aggregation). Overall, histone deacetylase inhibition alone was effective on more measures than arimoclomol. As in the FUS model, arimoclomol failed to induce HSPA1A or preserve Hspa8 mRNA in the TDP-43 model, despite preserving nuclear TDP-43 and Brg1, indicating neuroprotective properties other than HSP induction. The data speak to the complexity of drug mechanisms against multiple biomarkers of ALS pathogenesis, as well as to the importance of HSPA8 for neuronal proteostasis in both somata and dendrites.


Subject(s)
Amyotrophic Lateral Sclerosis , Biomarkers , DNA-Binding Proteins , Histone Deacetylase Inhibitors , Motor Neurons , Amyotrophic Lateral Sclerosis/metabolism , Amyotrophic Lateral Sclerosis/genetics , Amyotrophic Lateral Sclerosis/drug therapy , Histone Deacetylase Inhibitors/pharmacology , Biomarkers/metabolism , DNA-Binding Proteins/metabolism , DNA-Binding Proteins/genetics , Humans , Motor Neurons/metabolism , Motor Neurons/drug effects , Motor Neurons/pathology , Animals , HSP70 Heat-Shock Proteins/metabolism , HSP70 Heat-Shock Proteins/genetics , HSC70 Heat-Shock Proteins/metabolism , HSC70 Heat-Shock Proteins/genetics , Hydroxylamines/pharmacology , Cells, Cultured , RNA-Binding Protein FUS/metabolism , RNA-Binding Protein FUS/genetics , Superoxide Dismutase-1/metabolism , Superoxide Dismutase-1/genetics
14.
Ecotoxicol Environ Saf ; 276: 116327, 2024 May.
Article in English | MEDLINE | ID: mdl-38626605

ABSTRACT

Roxithromycin (ROX), a commonly used macrolide antibiotic, is extensively employed in human medicine and livestock industries. Due to its structural stability and resistance to biological degradation, ROX persists as a resilient environmental contaminant, detectable in aquatic ecosystems and food products. However, our understanding of the potential health risks to humans from continuous ROX exposure remains limited. In this study, we used the zebrafish as a vertebrate model to explore the potential developmental toxicity of early ROX exposure, particularly focusing on its effects on locomotor functionality and CaP motoneuron development. Early exposure to ROX induces marked developmental toxicity in zebrafish embryos, significantly reducing hatching rates (n=100), body lengths (n=100), and increased malformation rates (n=100). The zebrafish embryos treated with a corresponding volume of DMSO (0.1%, v/v) served as vehicle controls (veh). Moreover, ROX exposure adversely affected the locomotive capacity of zebrafish embryos, and observations in transgenic zebrafish Tg(hb9:eGFP) revealed axonal loss in motor neurons, evident through reduced or irregular axonal lengths (n=80). Concurrently, abnormal apoptosis in ROX-exposed zebrafish embryos intensified alongside the upregulation of apoptosis-related genes (bax, bcl2, caspase-3a). Single-cell sequencing further disclosed substantial effects of ROX on genes involved in the differentiation of motor neuron progenitor cells (ngn1, olig2), axon development (cd82a, mbpa, plp1b, sema5a), and neuroimmunity (aplnrb, aplnra) in zebrafish larvae (n=30). Furthermore, the CaP motor neuron defects and behavioral deficits induced by ROX can be rescued by administering ngn1 agonist (n=80). In summary, ROX exposure leads to early-life abnormalities in zebrafish motor neurons and locomotor behavior by hindering the differentiation of motor neuron progenitor cells and inducing abnormal apoptosis.


Subject(s)
Cell Differentiation , Motor Neurons , Zebrafish , Animals , Motor Neurons/drug effects , Motor Neurons/pathology , Cell Differentiation/drug effects , Apoptosis/drug effects , Water Pollutants, Chemical/toxicity , Anti-Bacterial Agents/toxicity , Embryo, Nonmammalian/drug effects , Locomotion/drug effects , Stem Cells/drug effects , Animals, Genetically Modified , Behavior, Animal/drug effects
15.
Ecotoxicol Environ Saf ; 277: 116359, 2024 Jun 01.
Article in English | MEDLINE | ID: mdl-38663194

ABSTRACT

2,6-Dihalogenated nitrophenols (2,6-DHNPs) are emerging halogenated nitroaromatic pollutants that have been detected in various water environments. However, there is currently limited research available regarding their potential impacts on locomotion behavior and neurotoxicity. Therefore, this study utilized zebrafish embryos to investigate the potential neurotoxic effects of 2,6-DHNPs by examining their impact on the nervous system at a concentration defined as 10% of the median lethal concentration. Our findings demonstrated that exposure to 2,6-DHNPs resulted in a significant 30 % decrease in the total swimming distance of zebrafish larvae, accompanied by notable impairments in motor neuron development and central nervous system. These effects were evidenced by a substantial 25% decrease in axonal growth, as well as disruptions in synapse formation and neuronal differentiation. Additionally, neurotransmitter analysis revealed marked decreases of 40%, 35%, and 30% in dopamine, 5-hydroxytryptamine, and acetylcholine levels respectively, highlighting disturbances in their synthesis, transport, and degradation mechanisms. These results emphasize the considerable neurotoxicity of 2,6-DHNPs at concentrations previously considered safe; thus necessitating a re-evaluation of environmental risk assessments and regulatory standards for such emerging contaminants.


Subject(s)
Embryo, Nonmammalian , Water Pollutants, Chemical , Zebrafish , Animals , Water Pollutants, Chemical/toxicity , Embryo, Nonmammalian/drug effects , Neurotoxicity Syndromes/etiology , Motor Neurons/drug effects , Swimming , Neurotransmitter Agents/metabolism , Larva/drug effects
16.
J Appl Physiol (1985) ; 136(5): 1113-1121, 2024 May 01.
Article in English | MEDLINE | ID: mdl-38511211

ABSTRACT

The number of motor neurons (MNs) declines precipitously during the final trimester before birth. Thereafter, the number of MNs remains relatively stable, with their connections to skeletal muscle dependent on neurotrophins, including brain-derived neurotrophic factor (BDNF) signaling through its high-affinity full-length tropomyosin-related kinase receptor subtype B (TrkB.FL) receptor. As a genetic knockout of BDNF leads to extensive MN loss and postnatal death within 1-2 days after birth, we tested the hypothesis that postnatal inhibition of BDNF/TrkB.FL signaling is important for postnatal phrenic MN (PhMN) survival. In the present study, we used a 1NMPP1-sensitive TrkBF616A mutant mouse to evaluate the effects of inhibition of TrkB kinase activity on phrenic MN (PhMN) numbers and diaphragm muscle (DIAm) fiber cross-sectional area (CSA). Pups were exposed to 1NMPP1 or vehicle (DMSO) from birth to 21 days old (weaning) via the mother's ingestion in the drinking water. Following weaning, the right phrenic nerve was exposed in the neck and the proximal end dipped in a rhodamine solution to retrogradely label PhMNs. After 24 h, the cervical spinal cord and DIAm were excised. Labeled PhMNs were imaged using confocal microscopy, whereas DIAm strips were frozen at ∼1.5× resting length, cryosectioned, and stained with hematoxylin and eosin to assess CSA. We observed an ∼34% reduction in PhMN numbers and increased primary dendrite numbers in 1NMPP1-treated TrkBF616A mice. The distribution of PhMN size (somal surface area) DIAm fiber cross-sectional areas did not differ. We conclude that survival of PhMNs during early postnatal development is sensitive to BDNF/TrkB.FL signaling.NEW & NOTEWORTHY During early postnatal development, BDNF/TrkB signaling promotes PhMN survival. Inhibition of BDNF/TrkB signaling in early postnatal development does not impact PhMN size. Inhibition of BDNF/TrkB signaling in early postnatal development does not impact the number or CSA of DIAm fibers.


Subject(s)
Brain-Derived Neurotrophic Factor , Motor Neurons , Phrenic Nerve , Receptor, trkB , Signal Transduction , Animals , Female , Male , Mice , Animals, Newborn , Brain-Derived Neurotrophic Factor/metabolism , Cell Survival/physiology , Cell Survival/drug effects , Diaphragm/metabolism , Mice, Inbred C57BL , Motor Neurons/metabolism , Motor Neurons/physiology , Motor Neurons/drug effects , Phrenic Nerve/physiology , Phrenic Nerve/metabolism , Phrenic Nerve/drug effects , Pyrazoles , Pyrimidines , Receptor, trkB/metabolism , Signal Transduction/physiology
17.
J Physiol ; 602(9): 2061-2087, 2024 May.
Article in English | MEDLINE | ID: mdl-38554126

ABSTRACT

Motoneuron properties and their firing patterns undergo significant changes throughout development and in response to neuromodulators such as serotonin. Here, we examined the age-related development of self-sustained firing and general excitability of tibialis anterior motoneurons in a young development (7-17 years), young adult (18-28 years) and adult (32-53 years) group, as well as in a separate group of participants taking selective serotonin reuptake inhibitors (SSRIs, aged 11-28 years). Self-sustained firing, as measured by ΔF, was larger in the young development (∼5.8 Hz, n = 20) compared to the young adult (∼4.9 Hz, n = 13) and adult (∼4.8 Hz, n = 8) groups, consistent with a developmental decrease in self-sustained firing mediated by persistent inward currents (PIC). ΔF was also larger in participants taking SSRIs (∼6.5 Hz, n = 9) compared to their age-matched controls (∼5.3 Hz, n = 26), consistent with increased levels of spinal serotonin facilitating the motoneuron PIC. Participants in the young development and SSRI groups also had higher firing rates and a steeper acceleration in initial firing rates (secondary ranges), consistent with the PIC producing a steeper acceleration in membrane depolarization at the onset of motoneuron firing. In summary, both the young development and SSRI groups exhibited increased intrinsic motoneuron excitability compared to the adults, which, in the young development group, was also associated with a larger unsteadiness in the dorsiflexion torque profiles. We propose several intrinsic and extrinsic factors that affect both motoneuron PICs and cell discharge which vary during development, with a time course similar to the changes in motoneuron firing behaviour observed in the present study. KEY POINTS: Neurons in the spinal cord that activate muscles in the limbs (motoneurons) undergo increases in excitability shortly after birth to help animals stand and walk. We examined whether the excitability of human ankle flexor motoneurons also continues to change from child to adulthood by recording the activity of the muscle fibres they innervate. Motoneurons in children and adolescents aged 7-17 years (young development group) had higher signatures of excitability that included faster firing rates and more self-sustained activity compared to adults aged ≥18 years. Participants aged 11-28 years of age taking serotonin reuptake inhibitors had the highest measures of motoneuron excitability compared to their age-matched controls. The young development group also had more unstable contractions, which might partly be related to the high excitability of the motoneurons.


Subject(s)
Motor Neurons , Humans , Motor Neurons/physiology , Motor Neurons/drug effects , Adult , Adolescent , Female , Male , Child , Young Adult , Middle Aged , Action Potentials/physiology , Muscle, Skeletal/physiology , Muscle, Skeletal/growth & development , Muscle, Skeletal/innervation , Selective Serotonin Reuptake Inhibitors/pharmacology
18.
Arch Toxicol ; 98(6): 1859-1875, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38555327

ABSTRACT

Poisoning with the organophosphorus nerve agent VX can be life-threatening due to limitations of the standard therapy with atropine and oximes. To date, the underlying pathomechanism of VX affecting the neuromuscular junction has not been fully elucidated structurally. Results of recent studies investigating the effects of VX were obtained from cells of animal origin or immortalized cell lines limiting their translation to humans. To overcome this limitation, motor neurons (MN) of this study were differentiated from in-house feeder- and integration-free-derived human-induced pluripotent stem cells (hiPSC) by application of standardized and antibiotic-free differentiation media with the aim to mimic human embryogenesis as closely as possible. For testing VX sensitivity, MN were initially exposed once to 400 µM, 600 µM, 800 µM, or 1000 µM VX and cultured for 5 days followed by analysis of changes in viability and neurite outgrowth as well as at the gene and protein level using µLC-ESI MS/HR MS, XTT, IncuCyte, qRT-PCR, and Western Blot. For the first time, VX was shown to trigger neuronal cell death and decline in neurite outgrowth in hiPSC-derived MN in a time- and concentration-dependent manner involving the activation of the intrinsic as well as the extrinsic pathway of apoptosis. Consistent with this, MN morphology and neurite network were altered time and concentration-dependently. Thus, MN represent a valuable tool for further investigation of the pathomechanism after VX exposure. These findings might set the course for the development of a promising human neuromuscular test model and patient-specific therapies in the future.


Subject(s)
Cell Differentiation , Cell Survival , Induced Pluripotent Stem Cells , Motor Neurons , Nerve Agents , Organothiophosphorus Compounds , Humans , Induced Pluripotent Stem Cells/drug effects , Motor Neurons/drug effects , Organothiophosphorus Compounds/toxicity , Nerve Agents/toxicity , Cell Differentiation/drug effects , Cell Survival/drug effects , Neuronal Outgrowth/drug effects , Chemical Warfare Agents/toxicity , Dose-Response Relationship, Drug , Cells, Cultured
19.
J Neurosci ; 44(16)2024 Apr 17.
Article in English | MEDLINE | ID: mdl-38413232

ABSTRACT

Amyotrophic lateral sclerosis (ALS) is a debilitating neurodegenerative disorder marked by progressive motor neuron degeneration and muscle denervation. A recent transcriptomic study integrating a wide range of human ALS samples revealed that the upregulation of p53, a downstream target of inflammatory stress, is commonly detected in familial and sporadic ALS cases by a mechanism linked to a transactive response DNA-binding protein 43 (TDP-43) dysfunction. In this study, we show that prolonged interferon-gamma (IFNγ) treatment of human induced pluripotent stem cell-derived spinal motor neurons results in a severe cytoplasmic aggregation of TDP-43. TDP-43 dysfunction resulting from either IFNγ exposure or an ALS-associated TDP-43 mutation was associated with the activation of the p53 pathway. This was accompanied by the hyperactivation of neuronal firing, followed by the complete loss of their electrophysiological function. Through a comparative single-cell transcriptome analysis, we have identified significant alterations in ALS-associated genes in motor neurons exposed to IFNγ, implicating their direct involvement in ALS pathology. Interestingly, IFNγ was found to induce significant levels of programmed death-ligand 1 (PD-L1) expression in motor neurons without affecting the levels of any other immune checkpoint proteins. This finding suggests a potential role of excessive PD-L1 expression in ALS development, given that PD-L1 was recently reported to impair neuronal firing ability in mice. Our findings suggest that exposing motor neurons to IFNγ could directly derive ALS pathogenesis, even without the presence of the inherent genetic mutation or functional glia component. Furthermore, this study provides a comprehensive list of potential candidate genes for future immunotherapeutic targets with which to treat sporadic forms of ALS, which account for 90% of all reported cases.


Subject(s)
Amyotrophic Lateral Sclerosis , Induced Pluripotent Stem Cells , Animals , Humans , Mice , Amyotrophic Lateral Sclerosis/genetics , Amyotrophic Lateral Sclerosis/metabolism , Amyotrophic Lateral Sclerosis/pathology , B7-H1 Antigen/metabolism , Biomarkers , DNA-Binding Proteins/genetics , Induced Pluripotent Stem Cells/metabolism , Interferon-gamma/metabolism , Interferon-gamma/pharmacology , Motor Neurons/drug effects , Motor Neurons/metabolism , Motor Neurons/pathology , Tumor Suppressor Protein p53/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL