Skip to main content
Journal of Tissue Engineering logoLink to Journal of Tissue Engineering
. 2025 Mar 12;16:20417314241310508. doi: 10.1177/20417314241310508

From in vivo models to in vitro bioengineered neuromuscular junctions for the study of Charcot-Marie-Tooth disease

Camille Scherrer 1,, Camille Loret 1, Nicolas Védrenne 2, Colman Buckley 3, Anne-Sophie Lia 1,4,5, Vincent Kermene 3, Franck Sturtz 1,4, Frédéric Favreau 1,4, Amandine Rovini 1, Pierre-Antoine Faye 1,4
PMCID: PMC11898049  PMID: 40078221

Abstract

Peripheral neuropathies are disorders affecting the peripheral nervous system. Among them, Charcot-Marie-Tooth disease is an inherited sensorimotor neuropathy for which no effective treatment exists yet. Research on Charcot-Marie-Tooth disease has been hampered by difficulties in accessing relevant cells, such as sensory and motor neurons, Schwann cells, and myocytes, which interact at the neuromuscular junction, the specialized synapses formed between nerves and skeletal muscles. This review first outlines the various in vivo models and methods used to study neuromuscular junction deficiencies in Charcot-Marie-Tooth disease. We then explore novel in vitro techniques and models, including complex hiPSC-derived cultures, which offer promising isogenic and reproducible neuromuscular junction models. The adaptability of in vitro culture methods, including cell origin, cell-type combinations, and choice of culture format, adds complexity and excitement to this rapidly evolving field. This review aims to recapitulate available tools for studying Charcot-Marie-Tooth disease to understand its pathophysiological mechanisms and test potential therapies.

Keywords: Charcot-Marie-Tooth disease, neuromuscular Junction, hiPSCs, microphysiological systems, disease modeling


Graphical Abstract.

Graphical Abstract

Introduction to Charcot-Marie-Tooth disease

Peripheral neuropathies are among the most common inherited neurological diseases. They constitute a vast and heterogeneous group of rare disorders affecting peripheral nerves. Inherited peripheral neuropathies (IPNs) have a genetic cause and range from purely motor (Hereditary Motor Neuropathy) to purely sensory (Hereditary Sensory Neuropathy), but many patients are affected by an intermediate sensory-motor form known as Charcot-Marie-Tooth disease (CMT). CMT overall prevalence was estimated in the 1970s at 1:2500. 1 However, broader population studies have lowered the prevalence between 1:5,000 and 1:10,000 in European populations and 1:3,300 worldwide. 2

First described in 1886, CMT is characterized by distal motor and/or sensory deficits in all four limbs, following a distal-to-proximal pattern. CMT is a heterogeneous neurological disease characterized by a wide range of clinical manifestations that can vary significantly in onset, severity, and progression. Symptoms usually begin during childhood or early adulthood, although it can occur anytime from infancy to later in life. Over time, the disease leads to a gradual deterioration in muscle function, typically spanning over several decades. 1 The variability in severity and type of symptoms underscores the heterogeneous nature of CMT. Most common motor symptoms include muscle weakness and wasting in the feet and lower legs, seen in up to 99% of patients. 3 Patients frequently develop foot deformities, such as high arches (pes cavus) and curled toes (hammertoes). Therefore, patients experience frequent tripping, reduced walking speed, foot drop, and shorter stride lengths. In addition to motor symptoms, sensory issues commonly arise, typically in the lower legs and feet, with numbness, tingling, and reduced sensitivity to touch. These symptoms may progress to the hands. Pain is another key feature of CMT, often manifesting as a mix of neuropathic and nociceptive pain, which mainly affects the lower back and joints. 4 Other complications may include scoliosis, hand tremors, fatigue, hearing loss, and in rare cases, vocal cord paralysis. Chronic pain, especially in the extremities, is reported by many CMT patients, typically moderate in severity and of neuromuscular origin.46 Although CMT does not typically reduce life expectancy, it has a profound impact on quality of life, particularly by affecting balance, mobility, and manual dexterity.

Given that CMT is a complex neurological disease, its classification is essential for a better understanding of its pathophysiology and advancing research. It can be classified into subgroups based on inheritance pattern (either autosomal dominant (AD), autosomal recessive (AR), X-linked dominant (XD) or X-linked recessive (XR)) and nerve conduction velocity (NCV) testing, an electrodiagnostic that helps distinguish between demyelinating and axonal forms of the disease. Indeed, one of the major breakthroughs in CMT classification was made in 1968, by Dyck and Lambert and followed a decade later by Harding and Thomas through their electrophysiological studies.6,7 They observed differences in NCVs among CMT patients, distinguishing two major subtypes among them, either exhibiting demyelinating or axonal peripheral neuropathies based on these measurements. The first group is characterized by an NCV < 38 m/s and is referred to as CMT1 or “demyelinating CMT,” while the second group has an NCV ⩾ 38 m/s, similar to that of unaffected individuals, is known as CMT2 or “axonal CMT.” These differences arise from the specific types of cells that are damaged. CMT1, the most common form affecting approximately 70% of patients, is characterized by Schwann cell (SCs) alterations with progressive demyelination of axons. Myelin plays a crucial role in protecting and speeding up the transmission of electrical signals which explains the reduced NCV.810 In contrast, CMT2 primarily affects the neurons themselves, which explains why NCV values remain unchanged, 11 but with a reduced action potential (AP) amplitude. Additionally, several studies have identified a third group, called “intermediate CMT” (CMTI), which includes clinical cases that fall between the axonal and demyelinating forms, sharing characteristics of both. The NCV for CMTI ranges from 25 to 45 m/s.

With advancements in genetic testing, different subtypes of CMT have been identified, each associated with specific inheritance patterns and affected genes. For instance, CMT can result from dominant or recessive mutations. CMT4 corresponds to an autosomal recessive form of demyelinating CMT, with a prevalence of 1% to 9% depending on geographical regions in genetically identified CMT.12,13 When the CMT-causing gene is located on the X chromosome, the disease is referred to as CMTX. CMT has extremely heterogeneous genetic causes. The most common genes responsible for CMT are PMP22 (CMT1A), GJB1 (CMT1X), MPZ (CMT1B), and MFN2 (CMT2A) representing 90% of mutations found in patients diagnosed subjected to genetic testing. 12 Moreover, different alterations in the same gene can lead to distinct forms of CMT (demyelinating, axonal, or intermediate) and distinct modes of transmission, as is the case for GDAP1. Altogether, over 100 genes have been identified as CMT-causing genes. 14 This wide spectrum of genes intervene in ubiquitous and fundamental functions of the cell (i.e. myelination, transcription factors, adhesion, mitochondrial fission/fusion, vesicular trafficking, cell signaling and others).15,16

A striking observation is that despite the ubiquitous and crucial nature of mutated genes, deleterious conditions occur only in the peripheral nervous system (PNS), particularly affecting motor neurons (MNs) and their supporting SCs. These cells, along with skeletal muscle (SkM) fibers, are implicated in neuromuscular junctions (NMJs), which are one of the main impacted components of all forms of CMT.1719

NMJs are specialized synapses formed between lower MNs nerve terminals and motor end plates of SkMs. The MN cell body is located in the dorsal root ganglion (DRG) and innervates striated muscles. The NMJ corresponds to the physical junction, where APs are translated into muscle contractions. When APs reach the presynaptic terminal, a transient and local increase in calcium concentration triggers the exocytosis of acetylcholine (ACh)-loaded vesicles into the synaptic cleft. Upon binding to acetylcholine receptors, sodium influx triggers the release of calcium from the sarcoplasmic reticulum which translates macroscopically into contraction of SkM fibers. ACh is then degraded in the synaptic cleft by ACh-esterase.

The NMJ also includes non-myelinating SCs known as terminal SCs (tSCs, also referred to as perisynaptic SCs or teloglia). Myelinating SCs, on the other hand, are essential supporting cells wrapping around the axon and allowing the propagation of APs via saltatory conduction. Another cell type, known as kranocytes, has been identified as fibroblast-like cells that cap the NMJ and play a role in synaptic regeneration. 20

Although the NMJ is not the primary site of pathology, the axonopathy or demyelinating damage caused by CMT directly impacts this structure and leads to secondary myopathic consequences. Indeed, both CMT1 and CMT2 patients exhibit myopathic changes, 21 which are more pronounced in CMT2. Interestingly, NMJ dysfunction has been reported in patients with CMT2D and CMT2M using repetitive nerve stimulation and single fiber EMG, 22 even if NMJ defects have been scarcely directly observed. 23 Moreover, extensive studies on animal models have established this phenomenon, showing structural and morphological changes, synaptic dysfunction, and destabilization of NMJs.1719,24,25 An observational study on neuromuscular function in CMT1 and CMT2 patients as well as in healthy subjects, named ESTABLISH, has been conducted (NCT04980807). Although the results have not yet been published, this study confirms the growing interest of the scientific community in this crucial aspect of the pathology. Indeed, because the NMJ is the functional unit necessary to trigger the movement via the recruitment of muscles, muscle weakness and atrophy are an indirect proof of its contribution in some serious and invalidating effects in CMT. Studying it in vivo and in vitro definitely helps to understand the pathophysiological characteristics and evaluate the effects of therapeutic agents that could act directly upon it.

Despite significant advances in the field over the past few years, therapeutic options for patients with CMT remain limited. 26 The genetic heterogeneity of the disease and the diversity of pathophysiological mechanisms involved make it challenging to address a single therapeutic strategy. To better understand CMT disease and to test potential therapeutic approaches, it is necessary to generate a wide variety of models as close as possible to human pathology.

This review endeavors to comprehensively explore the various methodologies employed in both in vivo and in vitro models to deepen our understanding of CMT mechanisms. Moreover, we highlight significant advancements, such as the utilization of human induced pluripotent stem cells (hiPSCs) and cutting-edge microfluidic technologies, which have revolutionized the creation of more physiologically relevant in vitro models. By exploring these advancements, we endeavor to unveil a spectrum of possibilities that could pave the way for the development of a highly representative CMT model.

1. Conventional in vivo models

In the past, animal models have proven crucial in investigating neuropathy onset, molecular determinants, pathogenesis, and identifying potential druggable targets. We will address these in vivo models through the scope of the different techniques and methods used to study the NMJ pathophysiology in CMT.

Non-mammal models

Saccharomyces cerevisiae (S. cerevisiae; yeasts) is a simple organism used to study CMT at the genetic and molecular levels. S. cerevisiae are ideal for gene complementation assays2729 and are often used to study CMT-mutation consequences,3032 and to perform large-scale drug screening. 33 However, yeasts are single-cell organisms for which the study of NMJ is unattainable. Caenorhabditis elegans (C. elegans), Danio rerio (D. rerio; zebrafish), and Drosophila melanogaster (D. melanogaster) are commonly used models of NMJ diseases. They are well-characterized, genetically and molecularly traceable animals that show short generation times and are inexpensive to breed on a large scale, thereby increasing experimental throughput.3436

Behavioral assays

One of the simplest yet indirect ways to assess NMJs’ integrity is to perform behavioral assays. These can vary considerably among species. The locomotor capacities of C. elegans worm models of different CMT forms were measured using crawling or swimming/thrashing assays at the larval and adult stages, respectively.3739 Similarly, D.melanogaster can be subjected to either crawling or climbing assays depending on their developmental stage. Ali et al. tested both crawling and climbing (negative geotaxis assay) locomotor capacities of their pan-neuron-specific dSLC25A46a knock-down flies modeling CMT2, exhibiting locomotive defects. These results, supplemented by NMJ and mitochondrial visualization, lifespan analysis, learning assays, and ATP/Reactive Oxygen Species (ROS) production analysis, highlighted the role of dSLC25A46ain mitochondrial dynamics, revealing the contribution of its loss-of-function in the neurodegeneration characteristic of the disease. 40 Muscle contraction assays in larvae can also be performed by counting forward body wall contraction exhibited for a given period of time. 41 Using transgenic D. rerio lines overexpressing the human HARS1 (CMT2W) mutant in which sensory and motor divisions are tagged with GFP and RFP, respectively, Mullen et al. 42 linked abnormal PNS morphology with functional consequences through impaired touch-evoked escape response and swimming behavior of the fishes. These tests are widely used in D. rerio studies. 43 Other methods, such as the vibrational startle response state, visual motor response test, swimming endurance, and swimming posture are available to assess the locomotive behavior of D. rerio larvae.44,45 These locomotive assays are indirect but enable to draw conclusions regarding the presence of nervous system defects. In top-bottom approaches, such findings often prompt researchers to investigate the cause at cellular and molecular levels. In addition, in the context of large drug screening, phenotypic changes in motility can help discriminate between potentially efficient compounds. 39

NMJ visualization methods

At the cellular level, a convenient way of appreciating NMJ morphology and physiology is to immunolabel of pre- and/or post-synaptic proteins. Qualitative and quantitative immunolabeling techniques inform on the number and size of synaptic boutons, as well as the density of the active zone and length of synaptic branches. MNs reaching their target, and therefore establishing NMJs, can be quantified using transgenic nematodes expressing GFP in MNs subpopulations.3739 As the organization of the PNS is well established and conserved in C. elegans, abnormalities are easy to detect (Figure 1). However, this method, although simple due to the transparency of worms, does not hint at morphological and functional defects other than misguidance and/or incapability to form synapses. To specifically visualize the NMJ, the gold standard involves using α-bungarotoxin (BTX), which selectively binds to AChR on muscle cells. For D. rerio, this approach can be enhanced by staining muscle cells with fluorescent phalloidin conjugates and using D. rerio lines expressing GFP under the control of an Hb9 promoter, as demonstrated by Vettori et al. In this CMT2A model, caudal primary MNs in transparent D. rerio embryos appeared disorganized, with less arborization; phalloidin staining permitted to enlighten a reduced width of MFN2 mutant’s muscle fiber compared to control D. rerio. 46 An alternative strategy, when combined with BTX, involves marking the presynaptic SV2 protein.45,47 In addition, standardized, high-resolution quantitative imaging techniques have been developed, providing useful parameters such as axonal length, AChR clustering, and AChR-axon overlap, rendering high-throughput drug screening amenable in D. rerio models.43,48 Regarding D. melanogaster, their nerves can be specifically stained by anti-horseradish peroxidase antibodies. 49 The presynaptic active zone and the postsynaptic muscle folds can be stained with anti-Bruchpilot (Brp) and anti-Disk Large Protein (DLG1) antibodies. This combination of antibodies is the basis of many studies on NMJ in D. melanogaster.40,41,5052 Although not described in studies of NMJs in the context of CMT, it is worth noting that the use of ultrastructural microscopy can provide insight into the morphological changes of the NMJ (i.e. size, density of active zone) and organelles (i.e. mitochondria, microtubules). Nonetheless, electron microscopy is a valuable tool for studying myelin abnormalities in demyelinating neuropathy models. 53

Figure 1.

Figure 1.

Non-exhaustive listing of techniques and assays performed to study NMJ integrity in traditional models of Charcot-Marie-Tooth disease. Studies on CMT have primarily focused on C. elegans, D. rerio, D. melanogaster, and rodents. Non-mammalian models are advantageous for large-scale breeding, but rodents remain a preferred model for peripheral neuropathies. NMJ integrity is crucial for CMT research. Behavioral tests, though indirect, allow quick screening of locomotor defects. NMJ visualization can be achieved by immunolabelling, electron microscopy, or transgenic lines with fluorescent proteins, with D. rerio and C. elegans being particularly suited due to their transparent skin. Less commonly used techniques include electrophysiology, with rodents’ muscle activity assessed via electromyography. Pharmacological assays and omics approaches have also yielded interesting findings.

AChR: acetylcholine receptor; DLG1: disk large protein 1; EJP: excitatory junction potentials; FP: fluorescent protein; HRP: horseradish peroxidase.

Electrophysiological NMJ assessment

Although not reported in the context of CMT, electrophysiological recordings are technically possible and allow appreciation of the functionality of NMJs in D. rerio. 54 Notably, NMJ electrophysiological activity was recorded in nine nematode lines harboring different mutations in CMT-causing genes. The frequency of spontaneous post-synaptic currents (PSCs) in mutant nematodes was lower than that in wild type (WT) animals. 39 Spontaneous PSC frequency depends on post-synaptic membrane integrity and cholinergic receptor activity, leading to the conclusion that these structures might be affected, in accordance with morphological and behavioral data. 39 In D. melanogaster, the Giant Fiber system is a well-characterized neuronal circuit mediating the escape response in flies. Briefly, giant fibers originate from the brain and form a synapse with both an interneuron and a MN, which in turn projects to the tergotrochanteral muscle. Electrophysiological recordings permit to gather features such as response latency and ability to follow stimuli one-to-one at high frequency, representing synaptic strength and reliability, respectively. 55 D. melanogaster NMJ physiology is also assessable via excitatory junction potential electrophysiological recordings. 51

Pharmacological assays

Pharmacological assays of neuromuscular function in nematodes were performed using aldicarb, (an AChE inhibitor) and levamisole (a selective agonist of AChRs). Inhibition of AChE by aldicarb leads to spastic paralysis, the onset speed of which depends on the rate of ACh release from the presynapse. Thus, an increased sensitivity to aldicarb reflects presynaptic defects. In contrast, levamisole-induced paralysis translates to post-synaptic (muscle) defects. For example, hPDK3R158H C. elegans (CMT6X) were found to be hypersensitive to aldicarb and insensitive to levamisole, indicating that locomotor abnormalities observed are due to neuronal defects, confirming the axonal nature of neuropathy. 56 Levamisole was also used to determine whether the functionality of the body wall muscles of the nine nematode lines generated by Soh et al. 39 was compromised, which was indeed the case.

It is important to note that studies often reach their conclusions by combining the different approaches described above.

Mammal models

Rodents, including Mus musculus (mouse) and Ratus norvegicus (rat), remain the gold standard for disease modeling, 57 and CMT is not ruled out. 58 Fundamentally, the techniques used to study NMJ are the same as those used for non-mammalian models. However, the study of rodent NMJ is extremely widespread and enables detailed studies of NMJ integrity in an extensively characterized organism.

NMJ integrity, behavioral and electrophysiological tests: The example of GDAP1 deficiency

To investigate the functional consequences of loss of GDAP1 function in vivo, Gdap1 KO mice (Gdap1-/-) were subjected to the rotarod test, gait, and footprint analyses, all of which pointed out the defects in locomotor capacities of this model. 24 Subsequently, electrophysiological recordings along the sciatic nerve confirmed the occurrence of axonal neuropathy concomitant with an absence of demyelination, as assessed by ultrastructural analysis. Analysis of MN numbers and morphology confirmed MN degeneration while immunolabelling of β-tubulin and AChR (α-BTX) revealed a reduced occupancy of the NMJs in 12-month-old Gdap1-/- mice and abnormal tangle-like structures in terminal nerves. Additional investigations of GDAP1 loss were carried out on cultured DRG sensory neurons or MNs from Gdap1-/- mice. Electron microscopic ultrastructural analysis of cultured MNs extracted from KO mice revealed the presence of abnormal mitochondria in the proximal and distal regions of the mice sciatic nerves. Finally, mitochondrial protein expression levels were measured in various tissues of 5-month-old Gdap1-/- mice. Limited energy provision linked to dysfunctional mitochondria was found in the cerebellum and peripheral nerves of Gdap1-/- mice. This study reports a model that has been used in numerous other studies since.5961 This model was also used for the final in vivo assessment of drug efficiency in a wide screening of already-approved FDA drugs. In this study, motor performance and muscle strength were used as indicators of drug ability to reverse the disease course. 62 It is worth noting that a wide variety of motor behavior assessments exists in rodents (Figure 1). Interestingly, GDAP1 has also been associated with a gain-of-function phenotype. Evidence regarding whether alterations result in gain or loss of function are sometimes intricate. Therefore, these findings have been listed for each gene discussed in this review according to the corresponding subtype (Supplemental Table 1).

NMJ visualization methods and myelination insights

The pathophysiological involvement of NMJ can be a consequence of both axonal and demyelinating forms and its understanding has significantly evolved over time. Specifically, understanding how NMJ abnormalities and demyelination interact can clarify the underlying disease mechanisms, highlight the temporal sequence of pathological events, and help identify early therapeutic targets.

Since D. melanogaster and C. elegans differ in terms of myelination mechanisms and organization, those models are not proper to study the context of CMT disease. However, in other animal models, myelin abnormalities can, as in humans, manifest as reduced myelin thickness 63 and characteristic onion bulb formation resulting from continuous demyelination and remyelination processes (as seen in CMT1B models). 64 Myelination alterations can be effectively visualized using transmission electron microscopy (TEM), while instability in its protein and lipid structure are easily observable via immunocytochemistry or immunohistochemistry (as observed in PMP22-/- mice). 65

Regarding NMJ visualization methods, NMJ imaging and morphological analysis in CMT2D mice can unveil important parameters such as synapse elimination, endplate morphological maturation, NMJ growth, and denervation. 17

Although demyelinating forms ultimately lead to NMJ problems, 21 studies directly examining both myelination and NMJ integrity are scarce. Indirect links have been identified, particularly in CMT4C models, where distinct studies have reported NMJ abnormalities on one hand, and demyelination along with characteristic onion bulb formations on the other.

In axonal forms of CMT, where the primary defect lies within the axon itself, the effects on the NMJ are more direct. Studies on CMT2 have highlighted connections between NMJ alterations and denervation, changes in axon diameter, and myelin abnormalities, as seen in models such as CMT2D (GARS1) and DGAT2 zebrafish.47,66

In conclusion, although the direct approach to NMJ analysis in CMT research was complex, it has yielded valuable insights. The combined analysis of NMJ function and myelination now provides a comprehensive perspective on the interplay between axonal integrity, myelination, and NMJ function across CMT models, enhancing both our understanding and the therapeutic potential for CMT.

Pharmacological assays

Regarding pharmacological studies, the neurotoxic agent acrylamide can be used to explore subclinical phenotypes, as has been performed in mice genetically modified with loss-of-function mutations in Lrsam1, which showed no evident impairments in neuromuscular performance or NMJ morphological, functional, or anatomical PNS changes. 67 However, challenging LRSAM1-mice with acrylamide revealed increased neurological dysfunction, tremors, and dyscoordination, indicating that their axons are more prone to degeneration. 67 Moreover, homozygous mice presented with significantly reduced NCV arising directly from acrylamide treatment. Heterozygous mice showed significantly reduced motor axon numbers, whereas homozygous mice had a significantly decreased motor axon area compared with treated WT controls. 67 Although part of a study on the demyelinating form of CMT4C caused by mutations in SH3TC2, the applied proteomics analysis carried out by Cipriani et al. is of major importance in the field of study of NMJ pathologies, which requires mention in this section. By selecting key proteins for maintenance, function, and axonal preservation, this study revealed changes of 50% in the abundance of the selected proteins, probably explained by compensatory mechanisms and prevention of NMJ breakdown. 68

Omics approaches

Omics approaches, more particularly transcriptomics and proteomics, offer a comprehensive view of the molecular changes associated with diseases. Transcriptomics examines the full set of mRNAs, revealing genes with altered expression, while proteomics analyzes the complete set of proteins, providing insights into affected signaling pathways and cellular processes. Cipriani et al. 68 utilized transcriptomics and proteomics to study Sh3tc2cEx1/∆Ex1 mice, a model of CMT4C. Their transcriptomic analysis revealed an increased expression of AChRγ, a marker of denervation, and NGF, which may be involved in the enhanced nerve branching observed in these mice. The proteomic analysis of the sciatic nerves showed an upregulation of extracellular matrix proteins, suggesting an attempt to stabilize the NMJ in response to pathology. 68 Proteomics has also underlined the impact of florfenicol and MitoQ pharmacological derivatives on the sciatic nerves of GDAP1-null mice. 62 Both treatments prevented the decline in proteins involved in energy metabolism and antioxidant responses observed in untreated mice. Additionally, they reduced oxidative stress in nerve proteins These omics approaches are virtually transposable to any disease model.

Taken together, C. elegans, D. rerio, D. melanogaster, and rodents provide a wide collection of living organisms in which NMJs studies are feasible, each with its own advantages and limitations (Box 1). These models are particularly valuable for investigating the mutations encountered in CMT (Figure 1). Large animal models have also been studied69,70; however, despite positive arguments, their use is questionable from an ethical point of view. In any case, ethical considerations must be underlined, and compliance with 3R rules (Replace, Reduce, Refine) needs to be applied rigorously when animals are used as models.

Box 1.

Advantages and limits of animal models.

• S. cerevisiae allows for quick testing of CMT-related genetic variants although it does not provide integrative assessments.
• D. melanogaster and C. elegans offer fast, low-cost genetic study options and allow for large-scale screenings, though their lack of myelin and simplicity limit their modeling of complex diseases.
• D. rerio, a vertebrate model, facilitates high-throughput drug screening due to its transparency and neuromuscular similarity to humans, though not all findings translate to mammals as human pre-terminal axons are smaller and more fragmented, with more numerous postsynaptic folds than in other vertebrates.
• Rodents, while replicating key clinical features, are limited for studying inherited peripheral neuropathies due to cost, ethical concerns, and incomplete representation of the disease. Unlike in healthy humans, nerves are shorter and NMJs in mice undergo gradual degeneration and remodeling. Molecular profiling has shown that 97% of the shared pathway between human and mouse NMJs exhibit significant differences in protein expression levels, including key pathways such as axonal guidance signaling.
The choice of model depends on the research goal, with each organism providing unique insights while having limitations in modeling the full complexity of human diseases.

2. In vitro culture models: from monocultures to tissue bioengineering approaches

CMT has an impact on the cells of the PNs all the way to the NMJ, extending from MNs and their supporting cells, SCs and to the SkM fibers. Creating a functional in vitro NMJ is currently a focal topic in tissue bioengineering. Several emerging models hold the promise of enabling the simultaneous study of all cell-specific defects resulting from CMT gene mutations. In this context, we summarize the recent efforts made to develop CMT models that closely mimic in vivo settings.

Level 1: Progression of motor neurons, skeletal muscles, and Schwann cells monocultures

NMJs in vivo display a tripartite architecture, composed of MNs, SkM fibers, and SCs. The culture of these components individually is well-established in laboratories, whether they are obtained from cell lines, primary cultures, or induced pluripotent stem cells.

Motor neurons, Schwann cells and skeletal muscle fibers: Cell lines and primary culture

Extracting MNs from patients or healthy controls poses technical and ethical challenges, restricting resources to postmortem tissues, which only reflect severe end-stage disease. This limitation hinders the study of disease progression and therapeutic testing for CMT. To address this issue, in vitro CMT studies initially utilized primary animal cells or human cell lines. MNs were obtained by dissecting DRGs from rodents, a procedure applicable to specific disease models, 71 or from the ventral spinal cord of embryonic rats to obtain enriched primary MN cultures, further down transfected to study CMT mutations. 72 Civera-Tregón et al. 61 utilized the Gdap1-/- model described earlier to obtain easy to handle embryonic MNs primary cultures for mitochondrial defect studies related to GDAP1 deficiency. Human cell lines (such as SH-SY5Y or COS7) have also long been used to investigate gene mutation consequences in PNS disorders, including CMT.7376

Human primary SCs cultures were first described in 1980, obtained from peripheral nerve biopsies on 40 patients with various peripheral neuropathies. 77 However, large-scale human primary SCs cultivation is challenging, and these cells have a finite capacity for proliferation and passaging. 78 Thus, SC isolation and culture protocols are mostly available for rodents, 79 with primary SC cultures from adult and newborn CMT1A rats’ sciatic nerves. 80 For example, Trembler-J mice ( Pmp22TrJ ) or CMT1A rats’ sciatic nerves dissections yield primary SCs carrying Pmp22 duplication or three additional murine Pmp22copies, respectively. These cells were used to investigate the effects of curcumin or curcumin-cyclodextrin/cellulose nanocrystals (Nanocur) on the expression of key autophagy pathway markers in the accumulation of PMP22, oxidative stress and ER stress.81,82

Axonal loss and demyelination likely contribute to denervation and NMJ fragmentation, also known as the dying-back process, and linked to muscle atrophy in a CMT2A mouse model. 83 Although muscles are considered as secondary actors in CMT pathophysiology and therefore no CMT-specific muscle tissue monocultures have been developed, the potential role of satellite cells in some CMT forms cannot be dismissed. 84 Muscle tissues can easily be obtained either by culturing cell lines such as mouse myoblasts (C2C12) or by using human muscle precursor cells and primary myoblasts.85,86 However, muscle stem cells obtained from biopsies are rarely pure and rapidly lose their stem cell properties. 87

Induced pluripotent stem cells-derived models

About two decades ago, pluripotent stem cell research began to significantly advance cell type generation. CMT studies utilized human embryonic stem cells to produce MNs 88 and SCs. 89 Human induced pluripotent stem cells (hiPSCs) that share biological properties with embryonic stem cells without raising ethical issue have recently been contemplated with growing interest. Initially derived from mice, hiPSCs methods quickly adapted to human cells.9092 The first study that successfully generated MN lines from CMT2E (NEFL) and CMT2A patients’ hiPSCs in 35 days correlated with the mouse model’s pathophysiology, showing intermediate neurofilament accumulation in CMT2E MNs, reduced AP threshold, and abnormal channel current properties. 93 Other researchers developed scalable methods to obtain MNs from hiPSCs via embryonic body formation, highlighting how slight changes in concentrations and exposure times impact differentiation. 94 Guo et al. 95 later refined the protocol, achieving a highly pure culture with 70%–95% of cells expressing MN markers. Indeed, A major challenge of hiPSCs-derived MNs is achieving homogeneous and pure cultures. Sorting methods like sedimentation field flow fractionation (SdFFF) 96 or magnetic activated cell sorting (MACS) 97 can enhance differentiation outcomes. Time required to obtain mature CMT MNs was also reduced, reaching as little as 20 days.98,99 Additionally, adding retinoic acid and SAG (smoothened agonist) at specific times can refine the rostro-caudal identity of MN populations during differentiation. 100

Differentiating hiPSCs from CMT patients into SCs is attainable, though efficiency, reproducibility, and maturity remain issues.101104 Observing myelination of iPSC-derived MN axons by iPSC-derived SCs in culture would be ideal in the context of studying CMT disease, but this remains challenging in 2D cultures. Several studies have reported hiPSC-derived cell lines for various CMT-causing alterations, 105 as well as successful differentiation into MNs or SCs (Table 1).

Table 1.

Updated list of iPSC models of Charcot-Marie-Tooth disease derived from patient cells.

Gene CMT subtype Cells origin Genetic alterations Mode of generation Targeted differentiated cell type Main observations Therapeutics Reference Date of publication
AARS1 CMT2N Fibroblasts c.986G > A p.(Arg329His) mRNA reprograming kit (Stemgent) N/I N/I N/I Cai et al. 106 2021
ATP1A1 CMT2 Fibroblasts c.1798C > G p.(Pro600Ala) Episomal plasmid (pCXLE-hOCT3/4 shp53-F, pCXLE-hSK, pCXLE-hUL) Neural precursors and post-mitotic neurons Inability to differentiate patient hiPSC into mature neurons: decreased number of mature neurons in ATP1A1-CMT disease N/I Manganelli et al. 107 2019
EGR2 CMT1D PBMC c.1057C > G p.(Arg353Gly) Episomal vectors (Oct3/4, Sox2, Klf4, L-Myc, LIN28, and p53 carboxy-terminal dominant-negative fragment) NCSCs Glutathione S-transferase theta 2 upregulation and increased ROS in CMT NCSCs N/I Kitani-Morii et al. 101 2017
FIG4 CMT4J Fibroblasts c.122T > C p.(Ile41Thr) + c.718C > T p.(Arg183*) Retroviral vectors expressing SOX2, OCT3/4, KFL4 and C-MYC N/I N/I N/I Saporta et al. 93 2015
GARS1 CMT2D Fibroblasts N/I p.(Pro234LysTyr) c.2171C > A p.(Pro724His) CytoTune-iPS 2.0 Sendai Reprograming Kit Spinal MNs Impaired electrophysiological activity, reductions in mitochondrial transport velocities and acetylated α-tubulin levels Tubastatin A and CKD504 (HDAC6 inhibitors) Smith et al. 97 2022
CMT2D Fibroblasts c.880G > C p.(Gly294Arg) pCXLE-hSK, pCXLE-hUL and pCXLE-hOCT3/4-shp5-F N/I N/I N/I Lu et al. 108 2023
GDAP1 CMT2H Fibroblasts c.581C > G p.(Ser194*) c.487C > T p.(Gln163*) Episomal vectors (pCXLE-hOCT3/4 shp53-F, pCXLE-hSK, pCXLE-hUL) Spinal MNs Hyperexcitability N/I Faye et al. 98 ; subsequently used in Miressi et al. 99 and Benslimane et al. 109 2020
CMT2H Fibroblasts c.577A > T p.(Lys193*) + c.1019ins p.(Arg341fs) Retroviral vectors expressing SOX2, OCT3/4, KFL4 and C-MYC N/I N/I N/I Saporta et al. 93 2015
CMT2K Fibroblasts c.487C > T p.(Gln163*) + N/I p.(Thr288Asnfs*3) CytoTune-iPS 2.0 Sendai (OCT3/4, KLF4, SOX2 and C-MYC) Reprograming Kit N/I N/I N/I Martí et al. 110 2017
GJB1 CMT1X Fibroblasts c.193T > C (p.Tyr65His) Retroviral vectors expressing SOX2, OCT3/4, KFL4 and C-MYC N/I N/I N/I Saporta et al. 93 2015
CMTX Fibroblasts c.415G > A p.(Val139Met) Episomal vectors (OCT4, SOX2, KLF4, L-MYC, LIN28, shRNA-p53) iPSC differentiation abilities (ectodermal, mesodermal, endodermal differentiation) N/I N/I Son et al. 111 2017
HSPB1 CMT2F Fibroblasts c.545C > T p.(Pro182Leu) non-integrating Sendai virus vectors (Klf4, Oct3/4, Sox2, and cMyc) Spinal MNs Impaired chaperone activity and increased oligomers size, IxI/V motif plays an important, regulatory role in modulating protein–protein interactions. N/I Alderson et al. 112 2021
CMT2F Fibroblasts c.545C > T p.(Pro182Leu) Performed by VIB Stem Cell Center, Belgium Spinal MNs Decreased autophagic flux N/I Haidar et al. 113 2019
CMT2F Fibroblasts c.250G > A p.(Gly84Arg) Performed by VIB Stem Cell Center, Belgium Spinal MNs N/I Juneja et al. 114 ; subsequently used inVan Lent et al.115,116 2018
CMT2F Fibroblasts c.404C > T p.(Ser135Phe) c.545C > T p.(Pro182Leu) Sendai virus- vectors, expressing SOX2, OCT4,KLF4, and c-MYC MNs Mitochondrial abnormalities (velocity/movement) - Reduced α-tubulin acetylation Chemical X4, Chemical X9 (HDAC6 inhibitors) Kim et al. 117 2016
HSPB8 CMT2L Fibroblasts c.423G > T p.(Lys141Asn) Performed by VIB Stem Cell Center, Belgium Spinal MNs N/I Juneja et al. 114 ; subsequently used in Van Lent et al.115,116 2018
KIF5A CMT2/SPG10– Fibroblasts c.50G > A p.(Arg17Gln) CytoTune iPS 2.0 SendaiReprogramming Kit N/I N/I N/I Santangelo et al. 118 2023
MFN2 CMT2A2B PBMC / T-lymphocytes c.382C > T p.(His128Tyr) c.281G > A (p.Arg94Gln) Episomal vectors for OCT3/4, Sox2, Klf4, L-Myc, Lin28, and dominant negative p53 or OCT3/4, Sox2, Klf4, L-Myc, Lin28, and p53-shRNA MNs Abnormal mitochondiral morpholohy and motility predict drug-induced neurotoxicity Ohara et al. 119 2017
CMT2A2A Fibroblasts c.281G > A (p.Arg94Gln) Performed by VIB Stem Cell Center, Belgium Spinal MNs Downregulation of PFN2 in patients’ cells N/I Juneja et al. 114 ; subsequently used in Van Lent et al.115,116 2018
CMT2A2A Renal epithelial cells c.752C > T p.(Pro251Leu) Retroviral integration (OCT4, SOX2, KLF4 and c-MYC) N/I N/I N/I Xu et al. 120 2019
CMT2A2B Fibroblasts c.1188C > T p.(Ala383Val) oriP/EBNA1 vectors in which all six reprograming factors (OCT4, SOX2, NANOG, LIN28, c-Myc, and KLF4) Spinal MNs Mitochondrial abnormalities, more resistant to apoptosis, increased autophagic flux Nutlin-3 (p53 stabilization and accumulation) Rizzo et al. 121 2016
CMT2A2A Fibroblasts c.2119C > T p.(Arg707Trp) CytoTune-iPS 2.0 Sendai Reprograming Kit N/I N/I N/I Ababneh et al. 122 2022
CMT2A2A Fibroblasts c.1090C > T p.(Arg364Trp) Retroviral vectors expressing SOX2, OCT3/4, KFL4 and C-MYC Spinal MNs MFN2: mitochondrial abnormalities, hyperexcitability, altered Na+/Ca2+ channel dynamics; NEFL: mitochondrial abnormalities, NEFL aggregations, hyperexcitability, altered Na+/Ca2+ channel dynamics N/I Saporta et al. 93 ; subsequently used in Maciel et al. 123 and Feliciano et al. 124 2015
MPZ CMT1B PBMC c.292C > T p.(Arg98Cys), Episomal vectors (Oct3/4, Sox2, Klf4, L-Myc, LIN28, and p53 carboxy-terminal dominant-negative fragment) NCSCs Glutathione S-transferase theta 2 upregulation and increased ROS in CMT NCSCs N/I Kitani-Morii et al. 101 2017
CMT1B LCL from B-lymphocytes c.418T > A p.(Ser140Thr) Episomal plasmids (OCT4, SOX2, KLF4, L-MYC, LIN28, shRNA-p53) iPSC differentiation abilities (ectodermal, mesodermal, endodermal differentiation) N/I N/I Son et al. 125 2017
CMT1B Urinary renal epithelial cells c.292C > T p.(Arg98Cys) Retroviral integration (OCT4, SOX2, KLF4 and c-MYC) iPSC differentiation abilities (ectodermal, mesodermal, endodermal differentiation) N/I N/I Xu et al. 126 2019
CMT1B Fibroblasts c.116A > C p.(His39Pro) Retroviral vectors expressing SOX2, OCT3/4, KFL4 and C-MYC N/I N/I N/I Saporta et al. 93 2015
NEFL CMT1F Fibroblasts c.1099C > T p.(Arg367*) Episomal overexpression of OCT4, SOX2, KLF4 and MYC MNs Decreased NEFL mRNA linked to NMD activation Cycloheximide (NMD inhibitor) Sainio et al. 127 ; subsequently used in Sainio et al. 128 2018
CMT2E Fibroblasts N/I p.(Pro8Arg) Performed by VIB Stem Cell Center, Belgium Spinal MNs Downregulation of PFN2 in patients’ cells N/I Juneja et al. 114 ; subsequently used in Van Lent et al.115,116 2018
CMT2E Fibroblasts c.293A > G p.(Asn98Ser) Retroviral vectors expressing SOX2, OCT3/4, KFL4 and C-MYC Spinal MNs MFN2: mitochondrial abnormalities, hyperexcitability, altered Na+/Ca2+ channel dynamics; NEFL: mitochondrial abnormalities, NEFL aggregations, hyperexcitability, altered Na+/Ca2+ channel dynamics N/I Saporta et al. 93 ; subsequently used in Maciel et al. 123 and Feliciano et al. 124 2015
PDK3 CMT6X Fibroblasts c.473G > A p.(Arg158His) non-integrating episomal vectors (oriP/EBNA-1 vectors with OCT4, SOX2, NANOG, LIN28, c-Myc, KLF4) Spinal MNs Pyruvate dehydrogenase kinase 3 hyperphosphorylation, hyperphosphorylation of the E1 subunit of PDC, mitochondrial and bioenergetic abnormalities Dichloroacetate (PDK inhibitor) Perez-Siles et al. 129 2020
PMP22 CMT1A Fibroblasts Duplication Cytotune Sendai Reprograming Kit, genome editing and direct lineage-converted cells SCs Inflammatory cytokines upregulation and mysregulated pathways N/I Mukherjee-Clavin et al. 103 ; subsequently used in Van Lent et al. 115 2019
CMT1A Fibroblasts Duplication Retroviral vectors (OCT4, SOX2, c-MYC, KLF4) NCSCs, mesenchymal cells, SCs, peripheral neurons SCs developmental defects; no myelination on co-cultures; dysregulation of cholesterol/lipid biosynthesis and transportation N/I Shi et al. 102 2018
CMT1A PBMC Duplication Episomal vectors (Oct3/4, Sox2, Klf4, L-Myc, LIN28, and p53 carboxy-terminal dominant-negative fragment) NCSCs Glutathione S-transferase theta 2 upregulation and increased ROS in CMT NCSCs N/I Kitani-Morii et al. 101 2017
CMT1A Fibroblasts Duplication Retroviral vectors expressing SOX2, OCT3/4, KFL4 and C-MYC N/I N/I N/I Saporta et al. 93 2015
RAB7A CMT2B Fibroblasts c.484G > A p.(Val162Met) CytoTune2.0-iPS Sendai Reprograming Kit DRG sensory neurons Altered late endocytic pathway, increased number of lysosomes and lysosomal activity, affected EGFR signaling and cell migration N/I Romano et al. 130 2021
SH3TC2 CMT4C Fibroblats-derived hiPSCs c.211C > T, p.Gln71* c.2860C > T, p.Arg954* CRISPR base editing N/I N/I N/I Loret et al. 131 2024
VRK1 dCMT/dHMN Fibroblasts c.656G > T p.(Arg219Ile) c.761G > T p.(Thr254Leu) retroviral transduction of Oct4, Sox2, Klf4, and c-myc Spinal MNs Reduced protein levels, VRK1 mislocalization, decrease of coilin levels, abnormal Cajal Bodies, decreased neurite length and branching MG132 (proteasome inhibitor) increased VRK1 protein levels El-Bazzal et al. 132 2019

dHMN: distal hereditary motor neuropathies; NCSCs: neural crest stem cells; NPC: neural progenitor cells; N/I: no information or not assessed; PBMC: peripheral blood mononuclear cells; Px: patient x; yo: year old.

Article were selected based on a screening of PubMed database using the following key words: « iPSCs », « induced pluripotent stem cells », « Charcot-Marie-Tooth disease », « CMT ».

Compound heterozygous variants are signalled with a “+” in between the variations. Genetic alterations are stated as provided in the referenced paper, with the date of publication corresponding to the first article that introduced the cellular model.

As stated before, although muscle cells are not primarily affected in CMT, defects in MNs and SCs lead to muscle weakness and atrophy. To our knowledge, no SkM cell model from CMT patient hiPSCs exists, though healthy hiPSC lines have been differentiated into SkMs.133136

Level 2: Co-cultures

Motor neurons and skeletal muscle fibers to establish a neuromuscular junction stricto sensus

In vivo settings involve numerous cell types that interact to form functional and complex NMJs. Although monocultures of MNs and SCs alone provide useful insights into axonal and demyelinating forms, efforts should focus on establishing a NMJs closely resembling those of patients. Moreover, MNs input is essential for proper muscle maturation and functional NMJ generation.137139 Strickland et al. 140 reviewed NMJ co-culture models according to cell origin and structural complexity. Additionally, Barbeau et al. 141 have outlined an historical overview of in vitro NMJ models.

Homologous healthy rodents NMJ have been created using cell lines 142 and primary cells.143,144 Rodent ESC or iPSC-derived MNs and/or SkMs models have also been developed, including chimeric cultures with at least one cell component derived from human stem cells. 145 Co-culturing immortalized human SkMs with rat embryonic spinal cord explants formed functional NMJs with SCs for up to 14 days.146,147 Guo et al. 148 were the first to report that synapses between human spinal cord stem cell lines-derived MNs and rat SkMs could be formed under serum-free conditions in vitro. Shortly after, they presented an all-human NMJ composed of MNs and SkMs both derived from human stem cells, in a defined serum-free system. 149

A model combining MNs and myotubes from the same hiPSC line emerged later. 150 hiPSC-derived MNs were able to induce myotubes to elicite APs and contraction upon ACh or electrical stimulations. Nerve-muscle contacts, marked by AChR-α clusters, developed fully after 21 days. Co-localization of myotubes, MNs and synaptic markers was also observed. Thus, Demestre et al. 151 confirmed that hiPSC-derived MNs and SkMs could form functional NMJs, resembling those observed in heterologous NMJs. Other studies reported functional NMJs derived from single hiPSCs lines in MN-SkM mixed co-culture of independently differentiated MNs and SkMs, where MN electrical stimulations evidenced NMJ formation. 151 These human NMJ models are crucial for studying normal development, disease mechanisms, and new drug targets involving NMJ dysfunction and degeneration. However, to our knowledge, such MNs-SkMs co-culture have not been reported in the field of CMT disease. Establishing such models could be particularly relevant in a CMT context.

In pathological models, MNs derived from myotonic dystrophy type 1-hiPSC cell line were co-cultured with myotubes generated from a non-diseased human muscle cell line, Mu2b3. 152 While NMJs modeling myasthenia gravis (MG) are frequently cited, Amyotrophic Lateral Sclerosis (ALS) models are also documented.153,154 In 2021, an isogenic, MG patient specific NMJ was created allowing disease diagnosis and therapeutic evaluation using minimal blood serum, thanks to previously established functional quantification methods.155,156 In this study, optogenetically modified hiPSC-derived MNs were cultured with non-optogenetically hiPSC-derived SkMs attached to pillars in a compartmentalized system. Using an optical stimulation platform, NMJs responses were recorded and NMJs function could be evaluated in a robust and reproducible manner. 156 While the previous model aimed at diagnosing MG, other models have been designed and tested with the aim of becoming high-throughput therapeutic testing platforms. For instance, a MG co-culture model of primary SkM and WT hiPSC-derived MNs was used to investigate the effects of anti-nAChR antibodies. 157

These studies demonstrated that MNs and myotubes could be differentiated from the same individual and co-cultured to form functional NMJs. However, despite the advances and technologies developed for other pathologies, no co-culture modeling of CMT NMJs has been attempted. Additionally, determining the perfect media that permit the survival, growth and functionality of both cell types remains challenging, as maintaining the contractile capability of SkMs and the electrical activity of MNs is crucial.

Co-culturing motor neurons and Schwann cells

Incorporating SCs into NMJ models is not trivial as they are essential for accurate neuron physiology (e.g. saltatory nerve conduction, axon caliber, etc.), and their absence results in inaccurate NMJ models. 158 Different strategies exist to study MN-SC interactions. The first in vitro MN-SC model, proposed in 2009, demonstrated myelin sheath formation and nodes of Ranvier development in embryonic rat MN and neonatal SC co-cultures. 159 Using IFRS1 and NSC-34 mouse cell lines, Takaku et al. 160 observed axon-SC interactions, reminiscent of myelinated axons. Comparisons between 2D co-cultures of dissociated neurons from mouse embryo DRG and mouse sciatic nerve SC, and 3D adult mouse-derived DRG explants, highlighted the 3D DRG ex vivo model’s advantages in terms of reduced complexity, culture duration, and animal usage. 161 However, this strategy cannot be used for studies on human SC/MN interactions.

The first hiPSC-derived sensory neurons and rat SCs co-culture proved that rat SCs were able to myelinate human sensory neurons, and more importantly to enhance neuronal health and alignment of SCs to axons. 162 However, establishing such models with human SCs is more delicate, as myelination capacity of human SCs is decreased compared to rodent SCs. 163 A serum-free co-culture of hiPSC-derived MN and primary human SC displayed a 65% myelination efficiency, evaluated through the ratio of myelin segments to aligned SCs (assessed by MBP/neurofilament and S100b/neurofilament immunostainings, respectively), along with nodes of Ranvier formation. 164 Nonetheless, achieving the same efficiency with hiPSC-derived SC is more complex. 78 Enhancing myelination capabilities of hiPSC-derived SCs could be achieved by adding supportive molecules. Addition of fetal bovine serum, 165 c-AMP (via Krox20 activation)166,167, ascorbate, 164 or CoenzymeQ10 168 have been shown to positively influence myelination. Another approach, as discussed below, is to offer a 3D matrix to MN-SCs co-culture.

Level 3: Complex neuromuscular junctions

Although NMJ integrates MNs and SkMs, other cells intervene in the synapse physiology. 169 SCs are crucial for NMJ development and synaptogenesis,170172 maturation, 173 functioning, 174 maintenance and repair.175,176 Their presence is associated with the stability and longevity of NMJs in vitro. 177 Barik et al. 178 highlighted their role in AChR cluster maturation, NMJ formation and maturation by selectively removing SCs at key time points in mice. Moreover, the role of myelinating and non-myelinating SCs has been repeatedly demonstrated in vitro,147,179182 as they cap motor nerve terminals and myelinate axons in vivo. Including SCs in NMJ models would allow deeper study of their interactions with other cells and decipher their role in pathophysiology of the demyelinating forms of CMT, aiming to develop therapies targeting SCs dysfunctions. 183 Adding muscle tissues to the equation to study CMT will provide a means of assessing the functionality of the CMT MNs and SCs. Adding muscle tissues to CMT studies would assess the functionality of CMT MNs and SCs. Creating a tri-culture model of MNs, SkMs, and SCs to represent a human NMJ is technically challenging, with only a few successful functional tripartite NMJ studies.

A tri-culture mimicking NMJ in a microfluidic system was established using a rat-derived B35 neuronal cells, rat S42 SCs line, and mouse C2C12 myoblasts, where SC inclusion improved maturation and underscored the synergistic relationship among cells. 184 Numerous studies have documented myelinating co-cultures of neurons from various sources and rat SCs under 2D conditions,162,168,185 yet establishing such co-cultures with hiPSC-derived SCs remains challenging. Limitations include few protocols for SC differentiation from hiPSCs and the necessity to develop a medium accommodating all cell types. Few models have integrated healthy iPSCs-derived MNs, SCs and SkMs to create a tripartite NMJ (Tables 2 and 3). Hörner et al. created a functional NMJ by culturing hiPSCs-derived MNs and SCs with C2C12 mouse myoblasts within 9 days. SCs and MNs, which were previously characterized, were seeded on 2-day-old C2C12 myotubes. NMJ formation was assessed by immunostaining of AChRs. Cluster colocalizations of AChR-MNs were reported, and SCs aligned with neurites. Additionally, the presence of SCs and MNs synergistically increased myogenesis. The main challenge consisted in developing a tri-culture medium to meet the diverse needs of the different cell types. SCs displayed a bipolar, elongated spindle shaped morphology, decreased proliferation, and increased P0 and S100-positive cells, indicating better differentiation with the tailored tri-culture medium. 179 Hörner et al. went also established an in vitro NMJ derived entirely from hiPSCs. Interestingly, Hörner’s protocol for NMJ triculture permitted to control the purity and identity of cell populations prior to co-culture settings. Lin et al. employed an alternative approach to induce human NMJ formation within a single well using an hiPSC-line transiently expressing Dox-inducible MYOD1 coupled to mCherry. Doxycycline (dox), when added to hiPSC cultures, initiated differentiation into myotubes after 10 days. The culture medium was switched to MNs and SCs differentiation medium. NMJ presence was confirmed by AChR clustering on myotube surfaces, juxtaposed on the axon terminal, clustering of synaptic nuclei and formation of end plate structures, in addition to immunostaining and gene expression assays. TEM captured synaptic boutons capped by SCs and myelinated axons integrated with myotubes.180,186 Further tests showed NMJ functionality through muscle contraction in response to Ca²⁺ concentration and optogenetic activation of MNs. This healthy human NMJ model was adapted to a pathogenic condition by knocking down the survival motor neuron protein, hence recapitulating spinal muscular atrophy (SMA).180,186 Descriptive abnormalities and functional deficits observed recapitulated in vivo phenotypes of SMA and confirmed the interest of hiSPCs derived tri-cultures of MNs, SCs, and SkMs to evaluate the function of NMJ. While 2D NMJs described by Hörner et al. 179 detached due to massive contractions after a few days, 3D NMJs could be maintained for up to 100 days. 186

Table 2.

Tri-cultures containing motor neurons, skeletal muscle cells and Schwann cells to study the neuromuscular junction.

Reference Cell origin Disease model Time required for NMJ formation Format Cells characterization NMJ assessments Phenotype observed in tri-culture Therapeutics Maintenance of NMJ
Rat/mouse
Singh and Vazquez 184 B35 neuronal cell line, S42 Schwann cell line, C2C12 myoblast cell line N/I 12 days 2D, compartmentalized, microfluidic platform Myotubes fusion index; α-actinin and AChR (α-BTX) staining; morphology; viability N/I Facilitated SkM differentiation and increased α-BTX staining in presence of MNs and SCs N/I 14 days in presence of MNs and SCs (versus 10 days when SKM-derived cells were present alone)
Human/mouse
Hörner et al. 179 hiPSCs-derived MNs, hiPSCs-derived SCs, C2C12 cell line N/I 9 days a 2D SCs markers: S100b, vimentin; SkMs markers: desmin, myosin heavy chain, AChR (α-BTX); MNs markers: TUBB3, synaptophysin, MAP2, peripherin, ISL1, Hb9 and vAChT; elongation index N/I Promoted myogenesis, influence AChR clustering N/I Fixed after 3 days to avoid massive contraction-induced culture detachment
Human
Lin et al.180,186 Dox-inducible MYOD1-mCherry hiPSCs (line 201B7) differentiated into SkMs by dox supplementation followed by media-driven differentiation of MNs and SCs SMA SkMs formed at day 10, AChR clustering starting from day 21 2D MNs markers: ChAT, ISL1, Hb9, TUBB3, Neurofilaments, SV2; SCs markers: S100b and observed by TEM; SkMs markers: MHC, AChR (α-BTX); gene expression analysis in SkMs and MNs; SEM Calcium imaging; optogenetically driven SkM contraction SMA: Smaller area of NMJ; mitochondrial abnormalities; Myofiber integrity decreased; Altered muscular contraction Agrin Up to 100 days
Mazaleyrat et al. 187 Co-differentiation of hiPSCs derived MNs and hiPSC-derived SkMs from a healthy individual and patients with neuromuscular disorders DMD, DM1, FSHD2 and LGMD2A 19–21 days to achieve mature MNs and myogenic fibers able to contract 2D SkM markers: Pax3/7, Desmin, AChR (α-BTX), Titin, Dystrophin, MHC; MNs markers: Neurofilaments); SCs markers: S100b, MBP; transcriptomic profiling; TEM (lamina invaginations, synaptic cleft) Spontaneous myofibers contraction blocked by TTX and α-BTX addition; calcium signaling In disease models: reduced fiber section size, defects in sarcolemma anchoring, calcium amplitude disturbances and other disease specific alterations On healthy NMJ: L-Glutamic acid potassium salt monohydrate, Salbutamol, Carisoprodol, Mexiletine 30 days (maintained for 7 months)
Faustino Martins et al. 181 H9 hPSCs H1hESC and XM001 hiPSCs MG 50 days post-differentiation to reach maturity 3D organoids Transcriptomic profiling, single-cell analysis, organoid morphology, MNs markers: ChAT, TUJ1, SMI32; SCs markers: S100b, MBP; SkMs markers: Fast MyHC, AChR (α-BTX), MYOD/DESMIN; eletron microscopy Spontaneous muscle contractions blocked by curare addition; chemically triggered rhythmic activity blocked by TTX measured by MEA; calcium imaging Healthy: CPG-like circuits; MG: Severe reduction of NMJ; reduction in the contraction rate and amplitude N/I Up to 150 days (maintained for 1 year without obvious signs of deterioration)
Hörner et al. 179 hiPSCs derived MNs, hiPSCs derived SCs, hiPSCs derived SkM b N/I Data not shown 2D SCs stained with S100b; MNs stained with cAChT, Synaptophysin; SkM cells stained with Myosin heavy chain N/I N/I N/I Fixed after 10 days in culture
Urzi et al. 188 hiPSCs differentiated into self-organizing brachial spinal neurons, SkMs, and terminal SCs (among others) SMA 20 days to obtain cells; 50 days to reach maturity/functionality 2D MNs stained with HB9, ChAT, HOXC6, ISL1, FOXP1, LHX3, TUBB3, SMI32; Glial cells stained with GFAP, S100b; SkM markers: AChR (α-BTX), Desmin, MyHC.; Gene expression analysis; TEM Optogenetically driven SkM contraction and pharmacological (Acetylcholine, Curare) interventions; Calcium imaging; Patch clamp SMA: NMJs reduced in size and number; muscle contraction severely affected N/I More than 100 days
Van Lent et al. 115 hiPSCs differentiated into organoids including neurons, muscle cells and SCs (among others) CMT1A, CMT2A Confirmed by αBTX staining at day 30 3D organoids MNs markers: ISL1; MC markers: demsin, AChR (α-BTX); SkM marked with PAX7; SCs markers: P0, MBP, S100b; electron microscopy; qPCR MEA CMT1A: hypermyelination of the smallest axons, increased myelin periodic line distance, onion-bulb-like formation PXT3003 (Baclofen, Naltrexone hydrochloride and D-sorbitol); shRNAs Up to 50 days (as analyzes performed suggest)

AChR: Acetylcholine Receptor; αBTX: α-Botulinum Toxin; ChAT: Choline O-Acetyltransferase; CPG: central pattern generator; DM1: Myotonig dystrophy; DMD: Duchenne muscular dystrophy; Fast MyHC: fast myosin skeletal heavy chain; FOXP1: Forkhead Box P1; FSHD2: facio scapulo humeral dystrophy; hiPSCs: human induced pluripotent stem cells; hPSCs: human pluripotent stem cells; LGMD2A: limb girdle muscular dystrophy 2A; MEA: multi electrode array; MG: myasthenia gravis; MN: motor neuron; N/I: no information or not assessed; SC: Schwann cells; SEM: scanning electron microscopy; shRNA: short hairpin RNA; SkM: skeletal muscle cells; SMA: spinal muscular atrophy; SMI32: non-phosphorylated neurofilament; TEM: transmission electron microscopy; TTX: tetrodotoxin; vAChT: vesicular ACetylcholine transporter.

a

If differentiated cells had been prepared previously and thawed when tri-culture is needed.

b

hiPSCs-derived SkMs were not from the same line as other cell types included in the triculture. They were obtained by following Chal et al.’s 133 differentiation protocol.

Table 3.

Main microfluidic systems commercially available that could allow to recreate neuromuscular junctions to study CMT.

Company Format Product name References
Ananda 2D NeuroHTS N/I
AxoSim 3D NerveSim Sharma et al. 165
CuriBio 2D/3D Mantarray Plate kit Smith et al. 189
eNUVIO 2D/3D OMEGAACE/OMEGANMJ N/I
Hesperos N/I Service only Santhanam et al. 190
Mimetas 3D OrganoPlate 3-lane platform Spijkers et al. 191
Netri 2D DuaLink/TriaLink N/I
Xona microfluidics 2 or 3 compartments with microchannels XonaChips Costamagna et al., 192 Dinamarca et al. 193

N/I: no information or not assessed.

In studies focusing on the simultaneous generation of hiPSCs-derived MNs and SkMs, the presence of SCs was detected through immunostaining (S100β and MBP positive cells) and TEM in both 2D 187 and 3D formats.181,186 These “same dish NMJ” formed innervated and functional SkM tissue, confirmed via multi-electrode array (MEA), calcium imaging and biochemical stimulations181,187 (Table 2). These techniques, along with optogenetics, electrical stimulations, and patch clamp methods, are essential for assessing NMJ functionality. 194 Additionally, contraction strength can be quantified by calculating the muscle contraction-induced displacement of pillars to which the muscle bundle is attached. 153 Only one study examined the interaction of self-organizing neurons, muscle cells and other cell types, including myelinating SCs, in a CMT1A context. 115 A 3D organoid culture approach, favoring axon myelination by SCs, was chosen. Myelination was confirmed by electron microscopy, appearing impaired in the CMT1A model but not in the isogenic-corrected control or the CMT2A model. The defects could be corrected by short-hairpin RNA or a combinatorial drug. No other complex in vitro CMT models derived from hiPSCs have been developed. As highlighted by Van Lent’s study, such models could yield more predictive and physiologically relevant results for the various disease variants and facilitate therapeutic testing.

3. Structural complexity of Charcot-Marie-Tooth disease neuromuscular junction models: Insights into peripheral nervous system modelization with microphysiological systems

Challenges of classical 2D culture systems for NMJ studies

Classical 2D culture is a fundamental and straightforward method for in vitro analysis. NMJs were initially modeled by myotubes and MNs plated above. 148 Numerous studies involving MNs and SkMs (from mice or hiPSCs) employed these monolayer cultures to explore synaptogenesis, NMJ functionality and formation.150,195 Refinement of 2D culture includes using patterned surfaces or stimulating NMJ electrically or optogenetically. 196 However, maintaining MN/SkM co-cultures in a 2D conformation over time is challenging179,197 and the flat, rigid substrate restricts cellular migration, potentially hindering cell interactions and altering cell morphology and gene expression. 196 Traditional high-density 2D models do not allow clear visualization and orientation of axons. Functional analysis of individual MN-myotube is challenging, although plating at a lower density can mitigate this issue. 198

Emergence of microphysiological systems (MPS) in neuromuscular research

Microphysiological systems (MPS) encompass 3D culture, organoid/spheroid, and microfluidics composed of human or animal cells, tissue explants, or stem-cell derived “organoid” formations. Defined by international experts, MPS are systems “capable of emulating human (or any other animal species’) biology in vitro at the smallest biologically acceptable scale.” 199 In neuromuscular models for CMT research, 3D models have been described in two studies. Beyond the NMJ organoid from Van Lent et al. , Maciel et al. developed CMT2E spheroids derived from patients carrying mutations in either NEFL or MFN2.116,123 These spheroids/organoids facilitate cell-cell interaction and heterogeneous populations, providing a more accurate representation of human physiology. Regarding MPS, a compartmentalized microfluidic system elucidated pathomechanisms associated with CMT2B and its causing mutation gene Rab7. DRG neurons from adult rats and rat embryos, modified to express EGFP-Rab7, were plated into a compartmentalized microfluidic system, segregating axon terminals from cell bodies. Axonal transport of endosomes containing EGFP-Rab7 was observed with a high temporal resolution imaging technique (pseudo-TIRF). 200

To enrich the argumentation, the subsequent section explores strategies and techniques in the MPS field to recreate healthy human NMJs or model other peripheral neuropathies.

Designing effective NMJ-Chips

To create functional, operable NMJ models, the cell type layout on the chip must be defined. Spheroids and organoids are able to recapitulate NMJs, with Osaki’s work providing solid evidence. For instance, Faustino Martins et al. reported hiPSCs-derived self-organizing trunk neuromuscular organoids, reaching maturity 50 days post-differentiation. These organoids featured myelinating and terminal SCs and were maintained for up to 150 days. 181 If cell types are generated independently, MNs (in 2D or 3D) are typically placed in a compartment connected by channels to the muscle bundle compartment. SCs can be added in various ways.165,179

Stimulation techniques and functional readouts in NMJ-Chip designs

Designing a NMJ-chip involves careful consideration of MN stimulation methods, as it affects the overall design. Optogenetic activation of MNs via light induces muscle contraction, confirming cell interaction without requiring electric isolation between MN and SkM compartments.153,156,201 Alternatively, if genetic manipulation of the cell line is not desired, microchannels between compartments are required, as demonstrated by Santhanam et al., 190 where MNs were stimulated using an electrode in the MN compartment. Readout of NMJ functionality is essential. Osaki et al., 153 measured muscle bundle contraction force in response to MN opto-stimulation through the displacement of PDMS pillars. The study, comparing healthy and ALS-NMJ, showed reduced muscle contractions and other NMJ defects in the ALS model, validating the “ALS-on-a-chip” model for ALS pathogenesis studies and therapeutic testing. 153 Indeed, cantilevers are commonly used for real-time myotubes contraction measurements, assessing NMJ functionality. This “ALS-on-a-chip” is a derivative of organs-on-chips (OOCs), that have been primarily developed to furnish accessible models of ALS. 202 Another study reported a microfluidic device comprising a custom MEA. This upgrade permitted, for the first time, the recording of muscle AP triggered by evoked neuronal electrical stimulation of the human NMJs in culture. 203 Several adaptations of chips to MEA systems are already commercialized by several biotech companies. 204

Incorporation of Schwann cells and myelination in NMJ models

Adding SCs enhances NMJs functionality and is all the more important for the study of demyelinating pathologies such as CMT. It seems necessary for cells to be included in a matrix, enabling them to interact and rearrange. For example, co-cultures of iPSC-derived SC and MN in a 3D collagen sponge model has enabled to obtain neurites with a 0.2 μm thick myelin sheath, 205 though nodes of Ranvier were absent, likely due to an incomplete maturation. 205 Compared to 2D systems, SCs cultured in matrices such as gelatin methacryloyl, fibrin hydrogels and bioprinted alginate-gelatin hydrogel exhibited improved cell viability, proliferation, migration, differentiation, and neurotrophic factor production.206209

hiPSC-derived NMJ organoids also exhibited myelinated neurites, offering functional assessment in CMT1A organoids compared to controls. 116 In a sophisticated 3D biomimetic in vitro peripheral nerve model, 4.7% of axons were myelinated. 165 Altogether, these studies indicate that providing a scaffold/matrix is essential to obtain myelination with cells derived from hiPSCs. Regarding myelin observation in spheroids, TEM can easily be employed with cryosections of spheroids/organoids. However, if neurons are placed in a microfluidic chip, experimenters will be subjected to a dilemma between visualization of axons with TEM techniques (which prohibits the use of glass coverslips) and functional recording. Different strategies have been employed to circumvent this problem. Santhanam et al. 190 developed two distinct chips to analyze NMJs in a cytological and functional manner. AxoSim’ nerve-on-chip is particularly innovative, allowing MN/SC spheroids to grow in matrigel within hydrogel, enabling both TEM and optic microscopy observations 165 (Table 3). Finally, an alternative to TEM for myelin observation is the use of 3D confocal Raman microscopy, a viable alternative for transmission electron microscopy analysis, which gives access to the biochemical nature of the sample.

Enhancing myotube maturation through 3D culture and stimulation

Alignment of myotubes is important for coordinated, force generating muscle contraction but is difficult to achieve under standard 2D conditions.148,198,210 SkMs grown in 3D exhibit advanced structural and functional characteristics compared to those cultivated in 2D conditions. 201 Indeed, 3D engineered muscle tissues provide an optimal culture environment for SkM tissues, the latter showing enhanced contractile output over time. 97 Interestingly, parameters influencing hiPSC-derived SkM organoids maturity and functionality include cell population heterogeneity (including neuronal and mesenchymal populations), three-dimensionality (tissue cultivated on metal holders) and maturation medium supplementation with triiodo-L-thyronine. 211 A recent study on self-organizing NMJ in 2D also supported the benefits of cellular heterogeneity, maintaining cultures for over 100 days. 188 Nanoscale topographical stimulations can further increase myogenic cell alignment and improve functional maturation compared to unpatterned surfaces.212215 Another way of optimizing co-cultures is to electrically stimulate MNs and myotubes cultures, thus promoting maturation of both cell types and NMJ development, 216 stability, 217 and functionality. 218 Overall, it has been demonstrated that myotubes formation and maturation improve with chemical, mechanical, and electrical stimulations.

Alternative approaches to spheroids and microfluidic chips for NMJ models

Other solutions apart from spheroids/organoids and compartmentalized microfluidic chips are available to combine MN spheroids and muscle bundles. For example, Rimington et al. 219 cultivated primary human derived muscle cells in molds for 14 days before adding hiPSC-derived MN spheroids in a second layer of extracellular matrix, similar to perimysium, forming functional NMJ within 14 days in co-culture. In conclusion, the crucial point in the design of an MPS device is the outcome and type of data generated. Research on MPS has been one of the most prolific areas within the field of tissue engineering over the past decade, 199 and there are now turnkey commercial proposals for the cultivation of muscles, nerves and NMJ (Table 3).

As a final word, the advantages and limits of each culture setting (2D, 3D, and microfluidic chip) must be acknowledged and considered (Box 2).

Box 2.

Benefits and challenges of 2D, 3D, and microfluidic chip.

Numerous strategies exist to create in vitro NMJs.
• Among them, 2D co-cultures are the easiest to set up, offering relatively straightforward analysis. However, they lack the ability to recapitulate the architectural complexity seen in vivo and are not stable over time.
• To address these limitations, 3D co-cultures provide a more accurate representation of muscle contraction physiology. Additionally, simultaneous differentiation of iPSCs into multiple cell lineages has led to the development of neuromuscular organoids—self-organized 3D structures containing functional NMJs. These 3D structures hold great promise, though they are complex to establish and analyze, and variability among organoids can complicate interpretation.
• Finally, the use of microfluidic chips allows for precise environmental control, NMJ clustering in a specific zone and localized stimulation, although these devices require specialized fabrication skills and can be costly.
In vitro cellular models offer controlled environments and accessibility, making them ideal for isolating specific factors and performing high-throughput drug screenings. They provide various model types which can approximate NMJ physiology to different extents. However, they struggle to replicate the full physiological complexity and interconnectedness of tissues seen in vivo. Nonetheless, animal models are often costly, ethically complex, and difficult to control at the same granularity as in vitro setups.

4. CMT pathological processes at the NMJ: a contribution of all models

Animal models have been pivotal in expanding our understanding of NMJ pathology in CMT. These models have provided valuable insights into NMJ indirect anatomical and developmental changes following denervation.Compared to in vitro models, denervation is more easily observed in animal models, such as D. melanogaster and mice.52,220 For instance, in Pmp22-Tg mice, denervation was accompanied by MNs axonal sprouting, which may indicate a compensatory mechanism for muscle reinnervation. 220 More direct NMJ-related phenomena were also observed in vivo in Sh3tc2∆Ex1/∆Ex1 mice, which model CMT4C. Specifically, both pre- and post-synaptic discontinuities were detected. These anatomical defects were associated with the overexpression of the AChR-γ subunit, considered as a denervation marker. However, the observed increase in branching suggests the appearance of adaptive changes. 68 Furthermore, multiple NMJ-related pathological processes were observed specifically in vivo such as delayed maturation 25 and synaptic transmission issues.25,56 Additionally, D. melanogaster carrying alterations in dSLC25A46 and GARS exhibited reduced NMJ size and number.40,50,52

On the other hand, other pathological phenomena observed in various CMT subtypes in vivo were found to be correlated with in vitro models. For example, protein accumulation at the pre-synaptic terminal can disrupt NMJ functionality. This was observed in both the D. melanogaster CMT2D model 41 and in CMT2E mice. 221 Similar defects were, later on, detected in hiPSC-derived MNs cultures carrying the same NEFL mutations,93,116 as well as in NEFL spheroids. 123 Mitochondrial dysfunction is another critical pathological process at the NMJ associated with CMT. Mitochondrial defects have been documented in various animal models, including dSLC25A46a and dSLC25A46b knockdown flies and Gdap1 KO mice.24,40,50,61 These defects are characterized by altered mitochondrial morphology and impaired transport, which can destabilize the NMJ. For example, the CMT2A D. rerio model showed reduced mitochondrial retrograde transport velocity, a finding that was later confirmed in in vitro models using hiPSC-derived motor neurons with disrupted mitochondrial morphology and distribution.45,93,119,121 In Gdap1-/- rodents, defects included reduced NMJ occupancy, axonal tangles, motor neuron loss, and mitochondrial dysfunction with oxidative stress, findings also mirrored in hiPSC-derived MNs.24,99 Finally, a more indirect but significant pathological process observed in CMT is myelin alteration. Dysmyelination leads to reduced stimulus transmission to MNs and, ultimately, NMJ dysfunction. This phenomenon has been observed in multiple in vivo rodent models and patient nerve biopsies as well as in in vitro complex models. For instance, in CMT1A in vitro models, organoids recapitulated myelin abnormalities, such as an increased G-ratio and irregular myelin periodic line spacing, findings that align with rodent observations and patient biopsies.115,222

In vivo animal models are crucial for studying the NMJ pathophysiology in CMT, as they enable direct observation of complex phenomena such as denervation, axonal sprouting, pre- and post-synaptic disruptions, AchR-γ subunit overexpression, delayed maturation, synaptic transmission defects, myelination defects and reduced NMJ size and density. These pathological processes affecting the NMJ are illustrated in Figure 2. While in vitro models are more ethical and offer distinct advantages, they must be rigorously validated to ensure they accurately replicate disease-related phenomena and reflect human pathophysiology. Therefore, integrating both in vivo and in vitro models is essential to comprehensively understand the full spectrum of CMT-associated pathologies.115,116

Figure 2.

Figure 2.

Integrative view of the observations and processes identified in the pathophysiology of NMJ dysfunction induced by CMT disease. Blue boxes represent findings obtained from in vivo models, whereas purple boxes indicate those observed in both in vivo and in vitro models. The mechanisms illustrated in this figure ultimately lead to muscle atrophy and the characteristic muscle weakness associated with the motor forms of CMT.

5. Insights into the therapeutic perspectives for CMT: Focus on the NMJ

Currently, there is no curative treatment for CMT, and treatments mainly focus on the management of CMT symptoms. However, a large number of in vivo preclinical studies have been conducted and documented, particularly in the fields of gene therapy (aimed at upregulating or downregulating the production of a protein of interest) and small molecule trials (such as the promising PXT3003 combinatorial therapy 223 ), as highlighted in recent reviews. 224 Animal models, particularly rodents, are an essential preclinical tool for evaluating the efficacy and safety of clinical approaches. The impact on the NMJ can be assessed through both direct and indirect methods, discussed in Section 1. As shown in Table 4, these models have been used to test various therapeutic approaches for treating CMT. Regarding treatments targetting the NMJ and its transmission failures directly, new approaches are being investigated. Salbutamol, a beta-2 adrenergic receptor agonist known to improve the structural and functional integrity of the NMJ in congenital myasthenic syndromes, did not lead to significant muscle strength improvements in a cohort of 15 patients presenting dHMN including 3 CMT2D and 2 CMT2M patients, despite a subjective improvement of fatigue. 22

Table 4.

Preclinical approaches tested on in vivo models and the methods used to investigate, either directly or indirectly, the NMJ.

Approaches Aim/Gene concerned Outcome measures related to the NMJ Preclinical studies model Reference
Genetic approaches (DNA targeted)
 Gene replacement with lentiviral vector Restore Sh3tc2 levels E, GT, R Sh3tc2−/− mice (CMT4C) Schiza et al. 225
Restore Gjb1 levels N/I Gjb1-null mice (CMT1X) Sargiannidou et al. 226
 Gene replacement with AAV vector Restore Sh3tc2 levels E, GT, R Sh3tc2−/− mice (CMT4C) Georgiou et al. 227
Restore Gjb1 levels GT, E Gjb1-null mice (CMT1B) Kagiava et al. 228
Restore Ighmbp2 levels MSQ, NMJ IF (AChR), R Ighmbp2nmd-2 J/J mice (CMT2S) Nizzardo et al. 229
Restore Fig4 levels E, GT, R, SWR Fig4plt/plt mice (CMT4J) Presa et al. 230
NT-3 levels upregulation MSQ Trembler-J (TrJ/+) mice (CMT1A) Yalvac et al. 231
 CRISPR-Cas9 Pmp22 downregulation E C22 mice (CMT1A) Lee et al. 232
Genetic approaches (RNA targeted)
 siRNA Pmp22 downregulation E, R Trembler-J (TrJ/+) mice CMT1A rat (CMT1A) Boutary et al., 222 Lee et al. 233
 shRNA Pmp22 downregulation E, GT, R, RS CMT1A rat (CMT1A) Gautier et al. 234
 miRNA Pmp22 downregulation E, R C22 mice (CMT1A) Lee et al. 235
GARS downregulation E, GT, NMJ IF (AChR), GarsΔETAQ huEx8 (CMT2D) Morelli et al. 236
 ASO Pmp22 downregulation E, GT, R C22 mice and CMT1A rat (CMT1A) Zhao et al. 237
Non-genetic approaches
 PXT3003 Pmp22 downregulation BT, E, HP, IP, WB CMT1A rat (CMT1A) Chumakov et al. 238
 Curcumin and derivatives Antioxidant, Neuroprotection GT, NMJ IF (AChR), R (R98C/+) and (R98C/R98C) mice (CMT1B) Patzkó et al. 239
BB, E, GA, GT, R, TN, VF CMT1A rat; C61 (Tg+/-) and C61 (Tg+/+) mice (CMT1A) Caillaud et al., 81 El Massry et al. 240
 Neuregulin-1 type III Promoting myelination E, GT, NMJ IF (AChR) CMT1A rat (CMT1A) Fledrich et al. 241
E P0S63del-CMT1B mice (CMT1B) Scapin et al. 242
Cell therapy
 SCs like derived from human tonsil-derived stem cells transplantation Promoting remyelination E, R Trembler-J (TrJ/+) mice (CMT1A) Park et al. 243

AChR: acetylcholine receptor; BB: beam balance test; BT: bar test; E: electrophysiological assessments; GA: gait analysis; GT: grip test; HP: hot plate; IF: immunofluorescence; IP: inclined plane; MSQ: myofiber size quantification; N/I: no information or not assessed; R: Rotarod; RS: Randall Selitto test; SWR: spontaneous wheel running; TN: thermal nociception test; VF: Von Frey’s test; WB: weight bearing.

This table presents a non-exhaustive list of preclinical trials conducted on rodent CMT models. Models are named according to the princeps paper.

Other diseases, such as myasthenia gravis, benefit from molecules acting at the NMJ. For example, NMD670, a muscle specific chloride ion channel (CIC-1) inhibitor, has been shown to enhance neuromuscular transmission and, ultimately, skeletal muscle function as demonstrated in a phase I clinical trial in myasthenia gravis. 244 This compound will soon be tested in a phase II trial to assess its efficacy, safety, and tolerability in ambulatory adults with CMT (SYNPASE-CMT 245 ;). Other molecules, such as Pyridostigmine and Nifedipine, both used to enhance neuromuscular excitation in SMA, could benefit from repositioning to help alleviate symptoms related to the neuromuscular abnormalities observed in CMT.

Discussion and perspectives

Animal models, particularly genetically engineered ones, have traditionally been used to study IPNs due to their ability to overcome limitations in research materials. These models provide valuable data on physiological and behavioral mechanisms and are still models of choice to study PNs. Rodents, notably, are extremely widespread models.

The in vivo part of this review focused on studies assessing NMJ integrity mainly in axonal CMT mice models. However, demyelinating CMT forms also warrant comprehensive investigation of NMJ characteristics. Indeed, Court et al. 20 hypothesized that myelin impairments in the pre-terminal region of axons could lead to NMJ damage with functional consequences. To address this, they examined muscles of periaxin KO mice, a common CMT4A model, and found that preterminal demyelination led to abnormal innervation patterns with presence of supernumerary branches, thinning of axon branches and focal swellings. Moreover, KO mice NMJs showed compromised electrophysiological features, with impaired neuromuscular transmission at >30 Hz frequencies. Similar conclusions regarding NMJ affections in demyelinating forms of CMT have been reported in SH3TC2 deficient mice or Trembler-J/PMP22-Tg mice, modeling CMT4C and CMT1A, respectively.68,220,246

While animal models offer valuable insights, they come with ethical and biological limitations. First, from an ethical point of view, the use of animals in science is increasingly regulated, hence the 3Rs rule: reduce, refine, replace. Mice do not fully capture the length dependent symptoms of axonal CMT due to their shorter stature compared to humans. Moreover, animal models may not accurately replicate the pathogenesis seen in humans due to genetic, metabolic, 247 and developmental difference, which can impact drug testing outcomes, thus limiting the transferability of preclinical research results to clinical settings. 248 For example, human NMJs are more stable than mice’s, 249 with smaller, more fragmented pre-terminal axons and more post-synaptic folds.250,251 Molecular profiling of human versus mice NMJs revealed that despite 36 molecular pathways being similar, 97% of these pathways show statistically significant differences in protein expression levels known to be important for synapse formation and maintenance. 250 As another example of the difficulty to compare human and animal physiology, Drosophila’s major NMJ neurotransmitter is glutamate rather than acetylcholine. 252 Intriguingly, NMJ neurotransmitter release displayed sexual dysmorphism in adult Caenorhabditis elegans, 253 which is for now contrasting with human physiology. Altogether, physiological differences between in vivo models and humans represent a significant limitation and must be considered.153,254

On the scale of a disease such as CMT, establishing a model for each gene and mutation involved requires a considerably large number of models, and inevitably as many animals, 255 which raises ethical concerns as well. Moreover, axonal CMT forms are more difficult to reproduce in rodent models, as they do not fully recapitulate the clinical and pathological findings of human patients. 58 Currently, the enormous heterogeneity of CMT disease renders drug screening incredibly fastidious, theoretically requiring the creation of new models for each genetic alteration. Developing complex in vitro hiPSC-derived NMJ would not only accelerate the process, but allow the creation of clean, high-throughput platforms for testing new and promising therapeutics. This advancement would result in significant reduction in time, cost and the number of animals used.218,256

In vitro systems, particularly those utilizing hiPSCs, offer promising alternatives to in vivo models. These systems allow the generation of NMJs, containing MNs, SkMs and SCs all derived from the same individual. Studying all cell types found to interact in vivo is crucial for a rigorous representation of the cellular mechanisms and pathological aspects observed in CMT. hiPSCs, derived from easily obtainable sources, provide ethical, isogenic, and scalable options for generating NMJ components. Nevertheless, avoiding animal models does not absolve researchers from ethical consideration, which must remain an integral part of research on patient-derived cells. 257

The most widely used method to obtain cellular models is to use somatic cells of patients and reprogram them (Table 1). Yet, this method tends to be time and resource consuming and iPSCs differentiation protocols can influence the transcriptional profile of hiPSC-derived cells, such as myogenic progenitors. 258 Epigenetic memory can affect iPSC ability to differentiate into specific cell type. 259 Another concern with the use of hiPSCs is their lack of maturity. To address this, methods of artificial aging such as progerin expression 260 or trans-differentiation would further strengthen the model. Trans-differentiation refers to the direct differentiation of patient-derived somatic cells into the desired cell type without passing through a state of pluripotency, thereby retaining aging-associated transcriptomic signatures. 261 For example, fibroblasts can be trans-differentiated into MNs, 262 SCs263,264 or SkMs. In fact, MNs resulting from a trans-differentiation were shown to retain characteristics of aged cells, such as heterochromatin and nuclear organization, extensive DNA damage and increased senescence-associated SA-ß-Gal activity. 265

CRISPR technology offers a promising and innovative avenue for generating CMT models by introducing specific CMT mutations into a “healthy” hiPSCs line or by correcting mutations in hiPSCs derived from CMT patients.124,131 Developing disease-specific models, along with their isogenic controls offers genetic consistency and allows for rigorous comparison and analysis, helping distinguish the direct effects of the specific CMT mutations from other genetic variations. The choice between using isogenic models and traditional controls is critical: isogenic controls, which are genetically identical to the CMT models except for the specific mutation, provide a unique advantage by eliminating background genetic variability that might otherwise obscure or amplify disease phenotypes. In contrast, traditional controls may introduce confounding genetic differences that can influence results. Although introducing mutations into a control lineage provides crucial insights into the pathophysiology of the specific mutation, it may overlook interactions that could play an important role in the disease. For a long time, the CRISPR approach proved costly in terms of time and money, resulting in low editing rates and a high risk of indels, linked in particular to DNA double helix breakage. The refinement of genome editing techniques, in particular the base editing strategy, 266 now enables efficient and precise editing that is less deleterious to cells. This approach holds tremendous potential for advancing our understanding of CMT pathogenesis, especially linked to point alterations 115 and developing targeted therapies. However, caution is required to address off-target effects and epigenetic states.267270

Maintaining in vitro NMJ over time is necessary, as it allows the study of a mature and functional system, since pathogenesis in CMT disease often manifests progressively over time due to genetic and environmental factors, leading to an increasing pathogenicity. 2D systems often do not permit to maintain complex structures such as NMJs for extended periods and do not fully recapitulate physiological settings. So far, only one CMT1A organoid NMJ and one CMT2E spheroid NMJ have been reported,115,123 but 3D culture, as any other culture format, has its drawbacks. The lack of control over the architectural organization of cells significantly impacts reproducibility. 271 Drawbacks for 3D models are also the difficulty in visualizing and imaging NMJ, performing precise electrophysiological recordings, 272 and the unequal oxygen and nutrient distribution, potentially leading to cell death. This challenge could be addressed by implementing continuous nutrient provision and waste removal strategies. 273 Sometimes, the 3D culture format is not compatible with approaching large-scale therapeutic tests, requiring the use of 96-well plates. For example, Faustino Martins et al.’ 181 protocol permits to generate organoids measuring about 5 mm in diameter after 50 days in culture.Urzi et al. 188 adapted this protocol to produce 2D self-organizing neuromuscular organoids. The transition from 3D to 2D, which may seem obvious from a theoretical point of view, nevertheless required the addition of 2 SMAD inhibitors. 188 The optimized protocol enabled the carry out of therapeutic tests and, most importantly, was not subjected to the detachment regularly observed in 2D muscle or NMJ cultures.179,197 Nonetheless, spheroids and organoids are valuable resources for studying cell-to-cell interaction in physiological or pathological conditions.

The methods used to obtain NMJs may also differ. While some protocols, such as Hörners’, assemble the cell types in a second step, after obtaining them separately, others opt for the self-organizing method. Whether in 2D184,187,188 or 3D,115,181 it would appear that the cells can influence themselves to differentiate via autocrine and paracrine signals into the different cell types without the need to add exogenous factors. 188

In addition to MNs and SkM fibers being the main actors of the NMJ, the involvement of SCs is now established, and the trend is moving toward a systematic integration of this cell type in NMJ models.115,179,181,186,187 However, different subtypes of SCs exits, the three major ones being myelinating SCs, TSCs and Remak SCs (RSCs). Remak SCs ensheath numerous small axons forming unmyelinated fibers, also called Remak bundles. 274 At present, very little information is available on the differentiation process of precursors into non-myelinating subtypes, that is, RSCs and TSCs. 104 All myelinating and non-myelinating SCs appear to be positive for S100b staining. However, TSCs need to be detected through the expression of both S100b and NG2. 275 This renders the characterization of SCs differentiation products difficult in terms of assessing which SC subtypes are predominant. Nonetheless, excluding them from studies on CMT NMJs would be a mistake, as RSCs seem to play a role in CMT1A pathogenesis. 276 Electron microscopic examination of CMT1A patient’s transverse sural nerve cross-sections revealed a proliferation of non-myelinating SCs, although no degeneration of non-myelinated fibers was found. 276

Complex microfluidic devices allow a precise control of many aspects of the co-culture, offering the possibility to work with high density or single cell cultures, temporal and spatial control, channel and valves integration, fluid flow and integration of systems such as MEA for electrophysiological studies. Mechanical stretching regimens, topographical patterning, and anchoring points also significantly improves myotubes formation and functionality. 153 Microfluidic can be adapted to welcome any cell type and overcomes the nutrient circulation and waste evacuation problems observed in 3D cultures. Developing microfluidic devices supplying all needs of cells integrated in the model will significantly improve the quality and functionality of in vitro CMT-NMJ models, as already observed in other diseases and healthy models.153,184 Indeed, there is currently no report of such model for CMT disease. Although not yet available, custom-made chips could even recapture the length-dependent damage characteristic of CMT. These tools will be necessary for the advent of high-throughput platforms for therapeutic molecules screening and investigating normal NMJ development and functioning. In any case, the movement is underway for a shift to more complex models.179,277,278

In addition to the in vivo and in vitro approaches described above, integrating omics data and computational analyses could offer an invaluable, multifaceted approach in CMT research. Multi-omics approaches, encompassing genomics, transcriptomics, proteomics, and metabolomics, allow for the identification, characterization, and quantification of the entire spectrum of biological molecules in cells or tissues. Applying such comprehensive results to computational analysis would provide deeper insights into gene interactions, enhance our understanding of disease mechanisms, and potentially identify new therapeutic targets, a promise already demonstrated in other diseases like multiple sclerosis. 279 In the field of CMT, Agrahari and George Priya Doss’s 280 study on the PMP22 gene used computational tools to identify highly deleterious single nucleotide polymorphisms and their effects on protein structure and function. Drug-gene interactions and docking simulations suggested estradiol as a promising therapeutic candidate, surpassing ascorbic acid, which had no significant effect on neuropathy compared with placebo after 2 years of treatment in clinical trials.280,281 While estradiol’s effects on CMT1A remain unexplored, earlier studies on Schwann cells tested various hormones and observed activity only with progesterone, although these studies were under differing experimental conditions. 282 In the context of biomarker identification, Jennings et al. conducted a proteomic quantification of serum collected from patients and mice with various types of CMT. The levels of NCAM1 and GDF15 proteins were found to be elevated in the serum of both patients and mice. Notably, serum levels of NCAM1 were correlated with the severity of neuropathy in patients. These results were further validated by staining for NCAM1 in the SkM of two patients with different genetic forms of CMT (FIG4, GDAP1). 23 Thus, integration of omics and computational approaches in CMT can help identify novel biomarkers for diagnosis, prognosis, and treatment response efficacy.283,284

Very few in vitro NMJ models have been composed entirely of human cell types and even fewer have used hiPSC-derived MNs, SCs and SkMs. This scarcity is likely due to the challenges of differentiating mature, hiPSC-derived SkMs and integrating all cell types in a model supporting all their needs. When the ultimate goal is to progress toward NMJ models capable of surpassing the need for animal testing, the integration of additional cell types, notably immune cells, endothelial cells and fibro-adipogenic progenitors, becomes necessary. 285 Such a comprehensive model can be achieved in organoids. 115 However, if the research aim is to study diseases in controlled, highly pure cell content, with identified cell types, adopting a multi-culture approach supported by compartmentalized systems emerges as a necessity to achieve the desired precision and reproducibility in experimental setups. This intricate balance between complexity and control is pivotal in advancing the development of alternative models that can emulate physiological responses more comprehensively. Rather than a linear progress toward more complex models, NMJs research must be considered as a network of experimental setups, each providing valuable knowledge. Ultimately, experiments and models must be viewed as a “fit for purpose” principle.

Conclusion

Despite significant progress in the field, there is a continued need for more comprehensive in vitro NMJ models composed exclusively of human-derived cell types. Standardization remains crucial to ensure the development of high-quality NMJs, as it attests reproducibility and comparability across studies. The exciting synergy between cellular technologies and innovative culture formats holds immense promise and is poised to guide future research endeavors in this domain.

As we navigate the abundance of new technologies and diverse options, it becomes imperative to tailor our study approaches with greater specificity. The judicious selection of the most suitable strategy—consideration of cell origin or incorporation of multiple cell type—will be pivotal in achieving our research goals. The ongoing integration of cutting-edge technologies and the refinement of methodologies underscore the optimistic trajectory of NMJ research, promising significant breakthrough through the understanding and treating of neuromuscular disorders.

Supplemental Material

sj-docx-1-tej-10.1177_20417314241310508 – Supplemental material for From in vivo models to in vitro bioengineered neuromuscular junctions for the study of Charcot-Marie-Tooth disease

Supplemental material, sj-docx-1-tej-10.1177_20417314241310508 for From in vivo models to in vitro bioengineered neuromuscular junctions for the study of Charcot-Marie-Tooth disease by Camille Scherrer, Camille Loret, Nicolas Védrenne, Colman Buckley, Anne-Sophie Lia, Vincent Kermene, Franck Sturtz, Frédéric Favreau, Amandine Rovini and Pierre-Antoine Faye in Journal of Tissue Engineering

Acknowledgments

Selected artwork (cells, neuromuscular junction, muscle, petri dish, capsule) shown in the graphical abstract and the modified NMJ and myelin segment in Figure 2 were used or adapted from pictures provided by Servier Medical Art (Servier; https://smart.servier.com/), licensed under a Creative Commons Attribution 4.0 Unported License. Other illustrations were created with Procreate.

Footnotes

Authorship: Amandine Rovini: Conceptualization, Methodology, Supervision, Writing - original draft, Writing - review & editing; Anne-Sophie Lia: Writing - review & editing; Camille Loret: Conceptualization, Methodology, Visualization, Writing - original draft, Writing - review & editing; Camille Scherrer: Conceptualization, Investigation, Methodology, Visualization, Writing - original draft, Writing - review & editing; Colman Buckley: Writing - review & editing; Franck Sturtz: Writing - review & editing; Frédéric Favreau: Conceptualization, Supervision, Writing - review & editing; Nicolas Védrenne: Supervision, Writing - original draft, Writing - review & editing; Pierre-Antoine Faye: Conceptualization, Methodology, Supervision, Writing - review & editing; Vincent Kermene: Writing - review & editing.

Data availability statement: Not applicable.

The author(s) declared no potential conflicts of interest with respect to the research, authorship, and/or publication of this article.

Funding: The author(s) disclosed receipt of the following financial support for the research, authorship, and/or publication of this article: This work is supported by institutional grants from the National Research Agency under the Investments for the Future program (references: ANR-10-LABX-0074-01 Sigma-LIM and ANR-20-CE17-0026), the Nouvelle-Aquitaine Region grant (AAPR2021-2020-11765410), the 2024 Exploratory AAP from the University of Limoges (MITIC), and the French Ministry of Research (MRT). We express our gratitude to the “Association pour l’Encouragement à la Recherche en Neurologie,” the “Club 41-098,” the “Lions Club St Yrieix La Perche,” the “Rotary Club,” the “Mairie de St Yrieix La Perche,” and the University of Limoges Foundation for their financial support.

Ethical considerations: There are no human participants in this article and informed consent is not required.

Consent to participate: Not applicable.

Consent for publication: Not applicable.

Supplemental material: Supplemental material for this article is available online.

References

  • 1. Bird T. Charcot-Marie-Tooth hereditary neuropathy overview. In: Adam M, Feldman J, Mirzaa G, et al. (eds) Genereviews®. Seattle, WA: University of Washington, Seattle, 1993. [PubMed] [Google Scholar]
  • 2. Barreto LCLS, Oliveira FS, Nunes PS, et al. Epidemiologic study of Charcot-Marie-Tooth disease: a systematic review. Neuroepidemiology 2016; 46(3): 157–165. [DOI] [PubMed] [Google Scholar]
  • 3. Johnson NE, Heatwole CR, Dilek N, et al. Quality-of-life in Charcot-Marie-Tooth disease: the patient’s perspective. Neuromuscul Disord 2014; 24(11): 1018–1023. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4. Gemignani F, Melli G, Alfieri S, et al. Sensory manifestations in Charcot-Marie-Tooth disease. J Peripher Nerv Syst 2004; 9(1): 7–14. [DOI] [PubMed] [Google Scholar]
  • 5. Thomas FP, Saporta MA, Attarian S, et al. Patient-reported symptom burden of Charcot-Marie-Tooth disease type 1A: findings from an observational digital lifestyle study. J Clin Neuromuscul Dis 2022; 24(1): 7–17. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6. Harding AE, Thomas PK. Genetic aspects of hereditary motor and sensory neuropathy (types I and II). J Med Genet 1980; 17(5): 329–336. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7. Dyck PJ, Lambert EH. Lower motor and primary sensory neuron diseases with peroneal muscular atrophy. I. Neurologic, genetic, and electrophysiologic findings in hereditary polyneuropathies. Arch Neurol 1968; 18(6): 603–618. [DOI] [PubMed] [Google Scholar]
  • 8. Manganelli F, Pisciotta C, Reilly MM, et al. Nerve conduction velocity in CMT1A: what else can we tell? Eur J Neurol 2016; 23(10): 1566–1571. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9. Vallat JM, Tazir M, Magdelaine C, et al. Autosomal-recessive Charcot-Marie-Tooth diseases. J Neuropathol Exp Neurol 2005; 64(5): 363–370. [DOI] [PubMed] [Google Scholar]
  • 10. Huang LW, Lin KP, Chang MH, et al. Electrophysiological characterization of Charcot-Marie-Tooth disease type 1A in Taiwan. J Chin Med Assoc 2012; 75(5): 197–202. [DOI] [PubMed] [Google Scholar]
  • 11. Tazir M, Nouioua S. Distal hereditary motor neuropathies. Rev Neurol 2024; 180(10): 1031–1036. [DOI] [PubMed] [Google Scholar]
  • 12. Murphy SM, Laura M, Fawcett K, et al. Charcot-Marie-Tooth disease: frequency of genetic subtypes and guidelines for genetic testing. J Neurol Neurosurg Psychiatry 2012; 83(7): 706–710. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13. Karakaya T, Turkyilmaz A, Sager G, et al. Molecular characterization of Turkish patients with demyelinating Charcot-Marie-Tooth disease. Neurogenetics 2022; 23(3): 213–221. [DOI] [PubMed] [Google Scholar]
  • 14. Inherited Neuropathy Variant Browser. [Internet]. https://neuropathybrowser.zuchnerlab.net/#/ (accessed 8 June 2024).
  • 15. Suter U, Scherer SS. Disease mechanisms in inherited neuropathies. Nat Rev Neurosci 2003; 4(9): 714–726. [DOI] [PubMed] [Google Scholar]
  • 16. Nave KA, Sereda MW, Ehrenreich H. Mechanisms of disease: inherited demyelinating neuropathies—from basic to clinical research. Nat Clin Pract Neurol 2007; 3(8): 453–464. [DOI] [PubMed] [Google Scholar]
  • 17. Sleigh JN, Grice SJ, Burgess RW, et al. Neuromuscular junction maturation defects precede impaired lower motor neuron connectivity in Charcot-Marie-tooth type 2D mice. Hum Mol Genet 2014; 23(10): 2639–2650. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18. Chandhok G, Soh MS. Presynaptic deficits at neuromuscular junctions: a specific cause and potential target of axonal neuropathy in type 2 Charcot-Marie-Tooth disease. ARC J Neurosci 2016; 36(31): 8067–8069. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19. Massana Muñoz X, Buono S, Koebel P, et al. Different in vivo impacts of dynamin 2 mutations implicated in Charcot-Marie-tooth neuropathy or centronuclear myopathy. Hum Mol Genet 2019; 28(24): 4067–4077. [DOI] [PubMed] [Google Scholar]
  • 20. Court FA, Gillingwater TH, Melrose S, et al. Identity, developmental restriction and reactivity of extralaminar cells capping mammalian neuromuscular junctions. J Cell Sci 2008; 121(Pt 23): 3901–3911. [DOI] [PubMed] [Google Scholar]
  • 21. Ericson U, Ansved T, Borg K. Charcot-Marie-Tooth disease–muscle biopsy findings in relation to neurophysiology. Neuromuscul Disord 1998; 8(3–4): 175–181. [DOI] [PubMed] [Google Scholar]
  • 22. McMacken G, Whittaker RG, Wake R, et al. Neuromuscular junction involvement in inherited motor neuropathies: genetic heterogeneity and effect of oral salbutamol treatment. J Neurol 2023; 270(6): 3112–3119. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23. Jennings MJ, Kagiava A, Vendredy L, et al. NCAM1 and GDF15 are biomarkers of Charcot-Marie-Tooth disease in patients and mice. Brain 2022; 145(11): 3999–4015. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24. Barneo-Muñoz M, Juárez P, Civera-Tregón A, et al. Lack of GDAP1 induces neuronal calcium and mitochondrial defects in a knockout mouse model of charcot-marie-tooth neuropathy. PLoS Genet 2015; 11(4): e1005115. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25. Spaulding EL, Sleigh JN, Morelli KH, et al. Synaptic deficits at neuromuscular junctions in two mouse models of Charcot–Marie–Tooth type 2d. J Neurosci 2016; 36(11): 3254–3267. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26. Stavrou M, Sargiannidou I, Georgiou E, et al. Emerging therapies for Charcot-Marie-Tooth inherited neuropathies. Int J Mol Sci 2021; 22(11): 6048. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27. Jordanova A, Irobi J, Thomas FP, et al. Disrupted function and axonal distribution of mutant tyrosyl-tRNA synthetase in dominant intermediate Charcot-Marie-Tooth neuropathy. Nat Genet 2006; 38(2): 197–202. [DOI] [PubMed] [Google Scholar]
  • 28. Rips J, Meyer-Schuman R, Breuer O, et al. MARS variant associated with both recessive interstitial lung and liver disease and dominant Charcot-Marie-Tooth disease. Eur J Med Genet 2018; 61(10): 616–620. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29. Meyer AP, Forrest ME, Nicolau S, et al. Pathogenic missense variants altering codon 336 of GARS1 lead to divergent dominant phenotypes. Hum Mutat 2022; 43(7): 869–876. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30. Rzepnikowska W, Kaminska J, Kabzińska D, et al. Pathogenic effect of GDAP1 gene mutations in a yeast model. Genes 2020; 11(3): 310. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31. Estela A, Pla-Martín D, Sánchez-Piris M, et al. Charcot-Marie-Tooth-related gene GDAP1 complements cell cycle delay at G2/M phase in Saccharomyces cerevisiae fis1 gene-defective cells. J Biol Chem 2011; 286(42): 36777–36786. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32. Rzepnikowska W, Kaminska J, Kochański A. Validation of the pathogenic effect of IGHMBP2 gene mutations based on yeast S. cerevisiae model. Int J Mol Sci 2022; 23(17): 9913. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33. Binięda K, Rzepnikowska W, Kolakowski D, et al. Mutations in GDAP1 influence structure and function of the trans-Golgi network. Int J Mol Sci 2021; 22(2): 914. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34. Singh J, Patten SA. Modeling neuromuscular diseases in zebrafish. Front Mol Neurosci 2022; 15: 1054573. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35. Kitani-Morii F, Noto YI. Recent advances in Drosophila models of Charcot-Marie-Tooth disease. Int J Mol Sci 2020; 21(19): 7419. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36. Markaki M, Tavernarakis N. Caenorhabditis elegans as a model system for human diseases. Curr Opin Biotechnol 2020; 63: 118–125. [DOI] [PubMed] [Google Scholar]
  • 37. Vester A, Velez-Ruiz G, McLaughlin HM, et al. A loss-of-function variant in the human histidyl-tRNA synthetase (HARS) gene is neurotoxic in vivo. Hum Mutat 2013; 34(1): 191–199. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38. Safka Brozkova D, Deconinck T, Beth Griffin L, et al. Loss of function mutations in HARS cause a spectrum of inherited peripheral neuropathies. Brain 2015; 138(8): 2161–2172. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39. Soh MS, Cheng X, Vijayaraghavan T, et al. Disruption of genes associated with Charcot-Marie-tooth type 2 lead to common behavioural, cellular and molecular defects in Caenorhabditis elegans. PLoS One 2020; 15(4): e0231600. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40. Ali MS, Suda K, Kowada R, et al. Neuron-specific knockdown of solute carrier protein SLC25A46a induces locomotive defects, an abnormal neuron terminal morphology, learning disability, and shortened lifespan. IBRO Rep 2020; 8: 65–75. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41. Grice SJ, Sleigh JN, Motley WW, et al. Dominant, toxic gain-of-function mutations in gars lead to non-cell autonomous neuropathology. Hum Mol Genet 2015; 24(15): 4397–4406. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42. Mullen P, Abbott JA, Wellman T, et al. Neuropathy-associated histidyl-tRNA synthetase variants attenuate protein synthesis in vitro and disrupt axon outgrowth in developing zebrafish. FEBS J 2021; 288(1): 142–159. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43. Lescouzères L, Bordignon B, Bomont P. Development of a high-throughput tailored imaging method in zebrafish to understand and treat neuromuscular diseases. Front Mol Neurosci 2022; 15: 956582. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44. Vaz R, Hofmeister W, Lindstrand A. Zebrafish models of neurodevelopmental disorders: limitations and benefits of current tools and techniques. Int J Mol Sci 2019; 20(6): 1296. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45. Chapman AL, Bennett EJ, Ramesh TM, et al. Axonal transport defects in a mitofusin 2 loss of function model of Charcot-Marie-Tooth disease in zebrafish. PLoS One 2013; 8(6): e67276. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46. Vettori A, Bergamin G, Moro E, et al. Developmental defects and neuromuscular alterations due to mitofusin 2 gene (MFN2) silencing in zebrafish: a new model for Charcot-Marie-Tooth type 2A neuropathy. Neuromuscul Disord 2011; 21(1): 58–67. [DOI] [PubMed] [Google Scholar]
  • 47. Hong YB, Kang J, Kim JH, et al. DGAT2Mutation in a family with autosomal-dominant early-onset axonal Charcot-Marie-Tooth disease. Hum Mutat 2016; 37(5): 473–480. [DOI] [PubMed] [Google Scholar]
  • 48. Lescouzères L, Hassen-Khodja C, Baudot A, et al. A multilevel screening pipeline in zebrafish identifies therapeutic drugs for GAN. EMBO Mol Med 2023; 15(7): e16267. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49. Jan LY, Jan YN. Antibodies to horseradish peroxidase as specific neuronal markers in Drosophila and in grasshopper embryos. Proc Natl Acad Sci USA 1982; 79(8): 2700–2704. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50. Suda K, Ueoka I, Azuma Y, et al. Novel Drosophila model for mitochondrial diseases by targeting of a solute carrier protein SLC25A46. Brain Res 2018; 1689: 30–44. [DOI] [PubMed] [Google Scholar]
  • 51. Sandoval H, Yao CK, Chen K, et al. Mitochondrial fusion but not fission regulates larval growth and synaptic development through steroid hormone production. eLife 2014; 3: e03558. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52. Niehues S, Bussmann J, Steffes G, et al. Impaired protein translation in Drosophila models for Charcot-Marie-tooth neuropathy caused by mutant tRNA synthetases. Nat Commun 2015; 6: 7520. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53. Pogoda HM, Sternheim N, Lyons DA, et al. A genetic screen identifies genes essential for development of myelinated axons in zebrafish. Dev Biol 2006; 298(1): 118–131. [DOI] [PubMed] [Google Scholar]
  • 54. Wen H, Brehm P. Paired patch clamp recordings from motor-neuron and target skeletal muscle in zebrafish. J Vis Exp 2010; (45): 2351. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55. Storkebaum E, Leitão-Gonçalves R, Godenschwege T, et al. Dominant mutations in the tyrosyl-tRNA synthetase gene recapitulate in drosophila features of human charcot–marie–tooth neuropathy. Proc Natl Acad Sci USA 2009; 106(28): 11782–11787. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56. Narayanan RK, Brewer MH, Perez-Siles G, et al. Charcot–Marie–tooth disease causing mutation (p.R158H) in pyruvate dehydrogenase kinase 3 (PDK3) affects synaptic transmission, ATP production and causes neurodegeneration in a cmtx6c. Elegansmodel. Hum Mol Genet 2021; 31(1): 133–145. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57. Bryda EC. The mighty mouse: the impact of rodents on advances in biomedical research. Mo Med 2013; 110(3): 207–211. [PMC free article] [PubMed] [Google Scholar]
  • 58. Bosco L, Falzone YM, Previtali SC. Animal models as a tool to design therapeutical strategies for CMT-like Hereditary Neuropathies. Brain Sci 2021; 11(9): 1237. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59. Cantarero L, Juárez-Escoto E, Civera-Tregón A, et al. Mitochondria-lysosome membrane contacts are defective in GDAP1-related Charcot-Marie-Tooth disease. Hum Mol Genet 2020; 29(22): 3589–3605. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60. Fernandez-Lizarbe S, Civera-Tregón A, Cantarero L, et al. Neuroinflammation in the pathogenesis of axonal Charcot-Marie-Tooth disease caused by lack of GDAP1. Exp Neurol 2019; 320: 113004. [DOI] [PubMed] [Google Scholar]
  • 61. Civera-Tregón A, Domínguez L, Martínez-Valero P, et al. Mitochondria and calcium defects correlate with axonal dysfunction in GDAP1-related Charcot-Marie-Tooth mouse model. Neurobiol Dis 2021; 152: 105300. [DOI] [PubMed] [Google Scholar]
  • 62. Nuevo-Tapioles C, Santacatterina F, Sánchez-Garrido B, et al. Effective therapeutic strategies in a preclinical mouse model of Charcot–Marie–Tooth disease. Hum Mol Genet 2021; 30(24): 2441–2455. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63. Khajavi M, Shiga K, Wiszniewski W, et al. Oral curcumin mitigates the clinical and neuropathologic phenotype of the Trembler-J mouse: a potential therapy for inherited neuropathy. Am J Hum Genet 2007; 81(3): 438–453. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64. Rünker AE, Kobsar I, Fink T, et al. Pathology of a mouse mutation in peripheral myelin protein P0 is characteristic of a severe and early onset form of human Charcot-Marie-tooth type 1B disorder. J Cell Biol 2004; 165(4): 565–573. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65. Lee S, Amici S, Tavori H, et al. PMP22 is critical for actin-mediated cellular functions and for establishing lipid rafts. J Neurosci 2014; 34(48): 16140–16152. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66. Mo Z, Zhao X, Liu H, et al. Aberrant GlyRS-HDAC6 interaction linked to axonal transport deficits in Charcot-Marie-Tooth neuropathy. Nat Commun 2018; 9(1): 1007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67. Bogdanik LP, Sleigh JN, Tian C, et al. Loss of the E3 ubiquitin ligase LRSAM1 sensitizes peripheral axons to degeneration in a mouse model of Charcot-Marie-Tooth disease. Dis Model Mech 2013; 6(3): 780–792. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68. Cipriani S, Phan V, Médard JJ, et al. Neuromuscular junction changes in a mouse model of Charcot-Marie-Tooth disease type 4C. Int J Mol Sci 2018; 19(12): 4072. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69. Granger N, Luján Feliu-Pascual A, Spicer C, et al. Charcot-Marie-tooth type 4B2 demyelinating neuropathy in miniature Schnauzer dogs caused by a novel splicing SBF2 (MTMR13) genetic variant: a new spontaneous clinical model. PeerJ 2019; 7: e7983. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70. Cahalan SD, Boehm I, Jones RA, et al. Recognising the potential of large animals for modelling neuromuscular junction physiology and disease. J Anat 2022; 241(5): 1120–1132. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71. Zhao J, Brown K, Liem RKH. Abnormal neurofilament inclusions and segregations in dorsal root ganglia of a charcot-Marie-Tooth type 2E mouse model. PLoS One 2017; 12(6): e0180038. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72. Jacquier A, Risson V, Schaeffer L. Modeling Charcot-Marie-Tooth disease in vitro by transfecting mouse primary motoneurons. J Vis Exp 2019; (143). [DOI] [PubMed] [Google Scholar]
  • 73. Pedrola L, Espert A, Wu X, et al. GDAP1, the protein causing Charcot-Marie-Tooth disease type 4A, is expressed in neurons and is associated with mitochondria. Hum Mol Genet 2005; 14(8): 1087–1094. [DOI] [PubMed] [Google Scholar]
  • 74. Pla-Martín D, Rueda CB, Estela A, et al. Silencing of the Charcot-Marie-Tooth disease-associated gene GDAP1 induces abnormal mitochondrial distribution and affects Ca2+ homeostasis by reducing store-operated Ca2+ entry. Neurobiol Dis 2013; 55: 140–151. [DOI] [PubMed] [Google Scholar]
  • 75. González-Sánchez P, Pla-Martín D, Martínez-Valero P, et al. CMT-linked loss-of-function mutations in GDAP1 impair store-operated Ca2+ entry-stimulated respiration. Sci Rep 2017; 7: 42993. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76. Kabzińska D, Chabros K, Kamińska J, et al. The GDAP1 p.Glu222Lys variant-weak pathogenic effect, cumulative effect of weak sequence variants, or synergy of both factors? Genes 2022; 13(9): 1546. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77. Askanas V, Engel WK, Dalakas MC, et al. Human schwann cells in tissue culture: histochemical and ultrastructural studies. Arch Neurol 1980; 37(6): 329–337. [DOI] [PubMed] [Google Scholar]
  • 78. Monje PV. Schwann cell cultures: biology, technology and therapeutics. Cells 2020; 9(8): 1848. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79. Palomo Irigoyen M, Tamayo Caro M, Pérez Andrés E, et al. Isolation and purification of primary rodent Schwann cells. Methods Mol Biol Clifton NJ 2018; 1791(1): 81–93. [DOI] [PubMed] [Google Scholar]
  • 80. Nobbio L, Sturla L, Fiorese F, et al. P2X7-mediated increased intracellular calcium causes functional derangement in Schwann cells from rats with CMT1A neuropathy. J Biol Chem 2009; 284(34): 23146–23158. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81. Caillaud M, Msheik Z, Ndong-Ntoutoume GM, et al. Curcumin-cyclodextrin/cellulose nanocrystals improve the phenotype of Charcot-Marie-tooth-1a transgenic rats through the reduction of oxidative stress. Free Radic Biol Med 2020; 161: 246–262. [DOI] [PubMed] [Google Scholar]
  • 82. Vázquez Alberdi L, Rosso G, Velóz L, et al. Curcumin and ethanol effects in Trembler-J Schwann cell culture. Biomolecules 2022; 12(4): 515. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83. Franco A, Dang X, Walton EK, et al. Burst mitofusin activation reverses neuromuscular dysfunction in murine CMT2A. eLife 2020; 9: e61119. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84. Ganassi M, Zammit PS. Involvement of muscle satellite cell dysfunction in neuromuscular disorders: expanding the portfolio of satellite cell-opathies. Eur J Transl Myol 2022; 32(1): 10064. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85. Bala M, Gupta P, Gupta S, et al. Efficient and modified 2-NBDG assay to measure glucose uptake in cultured myotubes. J Pharmacol Toxicol Methods 2021; 109: 107069. [DOI] [PubMed] [Google Scholar]
  • 86. Eberli D, Soker S, Atala A, et al. Optimization of human skeletal muscle precursor cell culture and myofiber formation in vitro. Methods 2009; 47(2): 98–103. [DOI] [PubMed] [Google Scholar]
  • 87. Mamchaoui K, Trollet C, Bigot A, et al. Immortalized pathological human myoblasts: towards a universal tool for the study of neuromuscular disorders. Skelet Muscle 2011; 1(1): 34. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88. Mou Y, Dein J, Chen Z, et al. MFN2 deficiency impairs mitochondrial transport and downregulates motor protein expression in human spinal motor neurons. Front Mol Neurosci 2021; 14: 727552. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89. Ziegler L, Grigoryan S, Yang IH, et al. Efficient generation of schwann cells from human embryonic stem cell-derived neurospheres. Stem Cell Rev Rep 2011; 7(2): 394–403. [DOI] [PubMed] [Google Scholar]
  • 90. Takahashi K, Yamanaka S. Induction of pluripotent stem cells from mouse embryonic and adult fibroblast cultures by defined factors. Cell 2006; 126(4): 663–676. [DOI] [PubMed] [Google Scholar]
  • 91. Takahashi K, Tanabe K, Ohnuki M, et al. Induction of pluripotent stem cells from adult human fibroblasts by defined factors. Cell 2007; 131(5): 861–872. [DOI] [PubMed] [Google Scholar]
  • 92. León M, Prieto J, Molina-Navarro MM, et al. Rapid degeneration of iPSC-derived motor neurons lacking gdap1 engages a mitochondrial-sustained innate immune response. Cell Death Discov 2023; 9(1): 217–311. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 93. Saporta MA, Dang V, Volfson D, et al. Axonal Charcot–Marie–Tooth disease patient-derived motor neurons demonstrate disease-specific phenotypes including abnormal electrophysiological properties. Exp Neurol 2015; 263: 190–199. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94. Maury Y, Côme J, Piskorowski RA, et al. Combinatorial analysis of developmental cues efficiently converts human pluripotent stem cells into multiple neuronal subtypes. Nat Biotechnol 2015; 33(1): 89–96. [DOI] [PubMed] [Google Scholar]
  • 95. Guo W, Naujock M, Fumagalli L, et al. HDAC6 inhibition reverses axonal transport defects in motor neurons derived from FUS-ALS patients. Nat Commun 2017; 8(1): 861. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 96. Faye PA, Vedrenne N, De la Cruz-Morcillo MA, et al. New Method for sorting endothelial and neural progenitors from human induced pluripotent stem cells by sedimentation field flow fractionation. Anal Chem 2016; 88(13): 6696–6702. [DOI] [PubMed] [Google Scholar]
  • 97. Smith AST, Kim JH, Chun C, et al. HDAC6 inhibition corrects electrophysiological and axonal transport deficits in a human stem cell-based model of Charcot-Marie-Tooth disease (Type 2D). Adv Biol 2022; 6(2): e2101308. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 98. Faye PA, Vedrenne N, Miressi F, et al. Optimized protocol to generate spinal motor neuron cells from induced pluripotent stem cells from Charcot-Marie-Tooth patients. Brain Sci 2020; 10(7): 407. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 99. Miressi F, Benslimane N, Favreau F, et al. GDAP1 involvement in mitochondrial function and oxidative stress, investigated in a Charcot-Marie-Tooth model of hiPSCs-Derived motor neurons. Biomedicines 2021; 9(8): 945. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 100. Mouilleau V, Vaslin C, Robert R, et al. Dynamic extrinsic pacing of the HOX clock in human axial progenitors controls motor neuron subtype specification. Development 2021; 148(6): dev194514. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 101. Kitani-Morii F, Imamura K, Kondo T, et al. Analysis of neural crest cells from Charcot-Marie-Tooth disease patients demonstrates disease-relevant molecular signature. Neuroreport 2017; 28(13): 814–821. [DOI] [PubMed] [Google Scholar]
  • 102. Shi L, Huang L, He R, et al. Modeling the pathogenesis of Charcot-Marie-Tooth disease type 1A using patient-specific iPSCs. Stem Cell Reports 2018; 10(1): 120–133. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 103. Mukherjee-Clavin B, Mi R, Kern B, et al. Comparison of three congruent patient-specific cell types for the modelling of a human genetic Schwann-cell disorder. Nat Biomed Eng 2019; 3(7): 571–582. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 104. Hörner SJ, Couturier N, Gueiber DC, et al. Development and in vitro differentiation of Schwann cells. Cells 2022; 11(23): 3753. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 105. Juneja M, Burns J, Saporta MA, et al. Challenges in modelling the Charcot-Marie-tooth neuropathies for therapy development. J Neurol Neurosurg Psychiatry 2019; 90(1): 58–67. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 106. Cai J, Kropf E, Hou YM, et al. A stress-free strategy to correct point mutations in patient iPS cells. Stem Cell Res 2021; 53: 102332. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 107. Manganelli F, Parisi S, Nolano M, et al. Insights into the pathogenesis of ATP1A1-related CMT disease using patient-specific iPSCs. J Peripher Nerv Syst 2019; 24(4): 330–339. [DOI] [PubMed] [Google Scholar]
  • 108. Lu PJ, Zhang P, Liu YC, et al. Gene repair of iPSC line with GARS (G294R) mutation of CMT2D Disease by CRISPR/Cas9. Curr Med Sci 2023; 43(2): 261–267. [DOI] [PubMed] [Google Scholar]
  • 109. Benslimane N, Miressi F, Loret C, et al. Amlexanox: Readthrough induction and nonsense-mediated mRNA decay inhibition in a Charcot–Marie–Tooth model of hiPSCs-Derived neuronal cells harboring a nonsense mutation in GDAP1 gene. Pharmaceuticals 2023; 16(7): 1034. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 110. Martí S, León M, Orellana C, et al. Generation of a disease-specific iPS cell line derived from a patient with Charcot-Marie-tooth type 2K lacking functional GDAP1 gene. Stem Cell Res 2017; 18: 1–4. [DOI] [PubMed] [Google Scholar]
  • 111. Son D, Kang PJ, Yun W, et al. Generation of induced pluripotent stem cell (iPSC) line from a 36-year-old Charcot-Marie-Tooth disease patient with GJB1 mutation (CMTX). Stem Cell Res 2017; 21: 9–12. [DOI] [PubMed] [Google Scholar]
  • 112. Alderson TR, Adriaenssens E, Asselbergh B, et al. A weakened interface in the P182L variant of HSP27 associated with severe Charcot-Marie-tooth neuropathy causes aberrant binding to interacting proteins. EMBO J 2021; 40(8): e103811. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 113. Haidar M, Asselbergh B, Adriaenssens E, et al. Neuropathy-causing mutations in HSPB1 impair autophagy by disturbing the formation of SQSTM1/p62 bodies. Autophagy 2019; 15(6): 1051–1068. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 114. Juneja M, Azmi A, Baets J, et al. PFN2 and GAMT as common molecular determinants of axonal Charcot-Marie-Tooth disease. J Neurol Neurosurg Psychiatry 2018; 89(8): 870–878. [DOI] [PubMed] [Google Scholar]
  • 115. Van Lent J, Vendredy L, Adriaenssens E, et al. Downregulation of PMP22 ameliorates myelin defects in iPSC-derived human organoid cultures of CMT1A. Brain 2023; 146(7): 2885–2896. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 116. Van Lent J, Verstraelen P, Asselbergh B, et al. Induced pluripotent stem cell-derived motor neurons of CMT type 2 patients reveal progressive mitochondrial dysfunction. Brain 2021; 144(8): 2471–2485. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 117. Kim JY, Woo SY, Hong YB, et al. HDAC6 inhibitors rescued the defective axonal mitochondrial movement in motor neurons derived from the induced pluripotent stem cells of peripheral neuropathy patients with HSPB1 mutation. Stem Cells Int 2016; 2016: 9475981. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 118. Santangelo S, Bossolasco P, Magri S, et al. Generation of an iPSC line from a patient with spastic paraplegia type 10 carrying a novel mutation in KIF5A gene. Stem Cell Res 2023; 66: 103008. [DOI] [PubMed] [Google Scholar]
  • 119. Ohara R, Imamura K, Morii F, et al. Modeling Drug-Induced neuropathy using human iPSCs for predictive toxicology. Clin Pharmacol Ther 2017; 101(6): 754–762. [DOI] [PubMed] [Google Scholar]
  • 120. Xu J, Fu Y, Xia W, et al. Generation of induced pluripotent stem cell line, ZJUCHi002-A, from Charcot-Marie-Tooth disease type 2A (CMT2A) patient with a mutation of c.752C>T in MFN2. Stem Cell Res 2019; 36: 101411. avr 2019;36:101411. [DOI] [PubMed] [Google Scholar]
  • 121. Rizzo F, Ronchi D, Salani S, et al. Selective mitochondrial depletion, apoptosis resistance, and increased mitophagy in human Charcot-Marie-Tooth 2A motor neurons. Hum Mol Genet 2016; 25(19): 4266–4281. [DOI] [PubMed] [Google Scholar]
  • 122. Ababneh NA, Barham R, Al-Kurdi B, et al. Generation of a human induced pluripotent stem cell (iPSC) line (JUCTCi019-A) from a patient with Charcot-Marie-Tooth disease type 2A2 (CMT2A2) due to a heterozygous missense substitution c.2119C > T (p.Arg707Trp) in MFN2 gene. Stem Cell Res 2022; 62: 102786. [DOI] [PubMed] [Google Scholar]
  • 123. Maciel R, Correa R, Bosso Taniguchi J, et al. Human tridimensional neuronal cultures for phenotypic drug screening in inherited peripheral neuropathies. Clin Pharmacol Ther 2020; 107(5): 1231–1239. [DOI] [PubMed] [Google Scholar]
  • 124. Feliciano CM, Wu K, Watry HL, et al. Allele-specific gene editing rescues pathology in a human model of Charcot-Marie-Tooth disease type 2E. Front Cell Dev Biol 2021; 9: 723023. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 125. Son D, Kang PJ, Yun W, et al. Generation of induced pluripotent stem cell (iPSC) line from Charcot-Marie-Tooth disease patient with MPZ mutation (CMT1B). Stem Cell Res 2017; 24: 5–7. [DOI] [PubMed] [Google Scholar]
  • 126. Xu J, Wang Y, He J, et al. Generation of a human Charcot-Marie-Tooth disease type 1B (CMT1B) iPSC line, ZJUCHi001-A, with a mutation of c.292C>T in MPZ. Stem Cell Res 2019; 35: 101407. [DOI] [PubMed] [Google Scholar]
  • 127. Sainio MT, Ylikallio E, Mäenpää L, et al. Absence of NEFL in patient-specific neurons in early-onset Charcot-Marie-Tooth neuropathy. Neurol Genet 2018; 4(3): e244. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 128. Sainio MT, Rasila T, Molchanova SM, et al. Neurofilament light regulates axon caliber, synaptic activity, and organelle trafficking in cultured human motor neurons. Front Cell Dev Biol 2021; 9: 820105. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 129. Perez-Siles G, Cutrupi A, Ellis M, et al. Energy metabolism and mitochondrial defects in X-linked Charcot-Marie-Tooth (CMTX6) iPSC-derived motor neurons with the p.R158H PDK3 mutation. Sci Rep 2020; 10(1): 9262. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 130. Romano R, Rivellini C, De Luca M, et al. Alteration of the late endocytic pathway in Charcot–Marie–Tooth type 2B disease. Cell Mol Life Sci 2021; 78(1): 351–372. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 131. Loret C, Pauset A, Faye PA, et al. CRISPR base editing to create potential Charcot-Marie-Tooth disease models with high editing efficiency: human induced pluripotent stem cell harboring SH3TC2 variants. Biomedicines 2024; 12(7): 1550. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 132. El-Bazzal L, Rihan K, Bernard-Marissal N, et al. Loss of Cajal bodies in motor neurons from patients with novel mutations in VRK1. Hum Mol Genet 2019; 28(14): 2378–2394. [DOI] [PubMed] [Google Scholar]
  • 133. Chal J, Al Tanoury Z, Hestin M, et al. Generation of human muscle fibers and satellite-like cells from human pluripotent stem cells in vitro. Nat Protoc 2016; 11(10): 1833–1850. [DOI] [PubMed] [Google Scholar]
  • 134. Borchin B, Chen J, Barberi T. Derivation and FACS-mediated purification of PAX3+/PAX7+ skeletal muscle precursors from human pluripotent stem cells. Stem Cell Reports 2013; 1(6): 620–631. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 135. Hosoyama T, McGivern JV, Van Dyke JM, et al. Derivation of myogenic progenitors directly from human pluripotent stem cells using a sphere-based culture. Stem Cells Transl Med 2014; 3(5): 564–574. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 136. Reilly M, Robertson S, Suzuki M. Sphere-based expansion of myogenic progenitors from human pluripotent stem cells. Methods Mol Biol 2023; 2640(5): 159–174. [DOI] [PubMed] [Google Scholar]
  • 137. Misgeld T, Burgess RW, Lewis RM, et al. Roles of neurotransmitter in synapse formation: development of neuromuscular junctions lacking choline acetyltransferase. Neuron 2002; 36(4): 635–648. [DOI] [PubMed] [Google Scholar]
  • 138. Machado CB, Pluchon P, Harley P, et al. in vitro modeling of nerve–muscle connectivity in a compartmentalized tissue culture device. Adv Biosyst 2019; 3(7): 1800307. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 139. Steinbeck JA, Jaiswal MK, Calder EL, et al. Functional connectivity under optogenetic control allows modeling of human neuromuscular disease. Cell Stem Cell 2016; 18(1): 134–143. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 140. Strickland JB, Davis-Anderson K, Micheva-Viteva S, et al. Optimization of application-driven development of in vitro neuromuscular junction models. Tissue Eng Part B: Rev 2022; 28(6): 1180–1191. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 141. Barbeau S, Tahraoui-Bories J, Legay C, et al. Building neuromuscular junctions in vitro. Development 2020; 147(22): dev193920. [DOI] [PubMed] [Google Scholar]
  • 142. Solomon EA, Rooney AM, Rodriguez AM, et al. Neuromuscular Junction model optimized for electrical platforms. Tissue Eng Part C: Methods 2021; 27(4): 242–252. [DOI] [PubMed] [Google Scholar]
  • 143. Martin NR, Passey SL, Player DJ, et al. Neuromuscular junction formation in tissue-engineered skeletal muscle augments contractile function and improves cytoskeletal organization. Tissue Eng Part A 2015; 21(19–20): 2595–2604. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 144. Smith AS, Passey SL, Martin NR, et al. Creating interactions between tissue-engineered skeletal muscle and the peripheral nervous system. Cells Tissues Organs 2016; 202(3–4): 143–158. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 145. Lynch E, Peek E, Reilly M, et al. Current progress in the creation, characterization, and application of human stem cell-derived in vitro neuromuscular junction models. Stem Cell Rev Rep 2022; 18(2): 768–780. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 146. Saini J, Faroni A, Abd Al, Samid M, et al. Simplified in vitro engineering of neuromuscular junctions between rat embryonic motoneurons and immortalized human skeletal muscle cells. Stem Cells Cloning Adv Appl 2019; 12(Suppl 1): 1–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 147. Saini J, Faroni A, Reid AJ, et al. A novel bioengineered functional motor unit platform to study neuromuscular interaction. J Clin Med 2020; 9(10): 3238. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 148. Guo X, Das M, Rumsey J, et al. Neuromuscular junction formation between human stem-cell-derived motoneurons and rat skeletal muscle in a defined system. Tissue Eng Part C: Methods 2010; 16(6): 1347–1355. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 149. Guo X, Gonzalez M, Stancescu M, et al. Neuromuscular junction formation between human stem cell-derived motoneurons and human skeletal muscle in a defined system. Biomaterials 2011; 32(36): 9602–9611. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 150. Demestre M, Orth M, Föhr KJ, et al. Formation and characterisation of neuromuscular junctions between hiPSC derived motoneurons and myotubes. Stem Cell Res 2015; 15(2): 328–336. [DOI] [PubMed] [Google Scholar]
  • 151. Guo X, Badu-Mensah A, Thomas MC, et al. Characterization of functional human skeletal myotubes and neuromuscular junction derived—from the same induced pluripotent stem cell source. Bioengineering 2020; 7(4): 133. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 152. Marteyn A, Maury Y, Gauthier MM, et al. Mutant human embryonic stem cells reveal neurite and synapse formation defects in type 1 myotonic dystrophy. Cell Stem Cell 2011; 8(4): 434–444. [DOI] [PubMed] [Google Scholar]
  • 153. Osaki T, Uzel SGM, Kamm RD. Microphysiological 3D model of amyotrophic lateral sclerosis (ALS) from human iPS-derived muscle cells and optogenetic motor neurons. Sci Adv 2018; 4(10): eaat5847. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 154. Stoklund Dittlau K, Krasnow EN, Fumagalli L, et al. Human motor units in microfluidic devices are impaired by FUS mutations and improved by HDAC6 inhibition. Stem Cell Reports 2021; 16(9): 2213–2227. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 155. Vila OF, Uzel SGM, Ma SP, et al. Quantification of human neuromuscular function through optogenetics. Theranostics 2019; 9(5): 1232–1246. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 156. Vila OF, Chavez M, Ma SP, et al. Bioengineered optogenetic model of human neuromuscular junction. Biomaterials 2021; 276: 121033. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 157. Smith VM, Nguyen H, Rumsey JW, et al. A functional human-on-a-chip autoimmune disease model of myasthenia gravis for development of therapeutics. Front Cell Dev Biol 2021; 9: 745897. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 158. Saporta MA, Shy ME. A human cellular model to study peripheral myelination and demyelinating neuropathies. Brain 2017; 140(4): 856–859. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 159. Rumsey JW, Das M, Stancescu M, et al. Node of Ranvier formation on motoneurons in vitro. Biomaterials 2009; 30(21): 3567–3572. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 160. Takaku S, Yako H, Niimi N, et al. Establishment of a myelinating co-culture system with a motor neuron-like cell line NSC-34 and an adult rat Schwann cell line IFRS1. Histochem Cell Biol 2018; 149(5): 537–543. [DOI] [PubMed] [Google Scholar]
  • 161. Mo YJ, Kim YS, Kim MS, et al. Advantages of adult mouse dorsal root ganglia explant culture in investigating myelination in an inherited neuropathic mice model. Methods Protoc 2022; 5(4): 66. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 162. Clark AJ, Kaller MS, Galino J, et al. Co-cultures with stem cell-derived human sensory neurons reveal regulators of peripheral myelination. Brain 2017; 140(4): 898–913. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 163. Monje PV, Sant D, Wang G. Phenotypic and functional characteristics of human Schwann cells as revealed by cell-based assays and RNA-SEQ. Mol Neurobiol 2018; 55(8): 6637–6660. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 164. Patel A, Rumsey JW, Lorance C, et al. Myelination and node of Ranvier formation in a human motoneuron–Schwann cell serum-free coculture. ACS Chem Neurosci 2020; 11(17): 2615–2623. [DOI] [PubMed] [Google Scholar]
  • 165. Sharma AD, McCoy L, Jacobs E, et al. Engineering a 3D functional human peripheral nerve in vitro using the nerve-on-a-chip platform. Sci Rep 2019; 9(1): 8921. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 166. Vijay S, Chiu M, Dacks JB, et al. Exclusive expression of the Rab11 effector SH3TC2 in Schwann cells links integrin-α6 and myelin maintenance to Charcot-Marie-Tooth disease type 4C. Biochim Biophys Acta 2016; 1862(7): 1279–1290. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 167. Huang Z, Powell R, Kankowski S, et al. Culture conditions for human induced pluripotent stem cell-derived Schwann cells: a two-centre study. Int J Mol Sci 2023; 24(6): 5366. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 168. Hyung S, Yoon Lee B, Park JC, et al. Coculture of primary motor neurons and Schwann cells as a model for in vitro myelination. Sci Rep 2015; 5(1): 15122. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 169. Hörner SJ, Couturier N, Hafner M, et al. Schwann cells in neuromuscular in vitro models. Biol Chem 2024; 405(1): 25–30. [DOI] [PubMed] [Google Scholar]
  • 170. Feng Z, Ko CP. The role of glial cells in the formation and maintenance of the neuromuscular junction. Ann N Y Acad Sci 2008; 1132: 19–28. [DOI] [PubMed] [Google Scholar]
  • 171. Herrera AA, Qiang H, Ko CP. The role of perisynaptic Schwann cells in development of neuromuscular junctions in the frog (Xenopus laevis). J Neurobiol 2000; 45(4): 237–254. [DOI] [PubMed] [Google Scholar]
  • 172. Ullian EM, Harris BT, Wu A, et al. Schwann cells and astrocytes induce synapse formation by spinal motor neurons in culture. Mol Cell Neurosci 2004; 25(2): 241–251. [DOI] [PubMed] [Google Scholar]
  • 173. Darabid H, St-Pierre-See A, Robitaille R. Purinergic-dependent glial regulation of synaptic plasticity of competing terminals and synapse elimination at the neuromuscular junction. Cell Rep 2018; 25(8): 2070–2082.e6. [DOI] [PubMed] [Google Scholar]
  • 174. Sugiura Y, Lin W. Neuron-glia interactions: the roles of Schwann cells in neuromuscular synapse formation and function. Biosci Rep 2011; 31(5): 295–302. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 175. Riethmacher D, Sonnenberg-Riethmacher E, Brinkmann V, et al. Severe neuropathies in mice with targeted mutations in the ErbB3 receptor. Nature 1997; 389(6652): 725–730. [DOI] [PubMed] [Google Scholar]
  • 176. Balice-Gordon RJ. Dynamic roles at the neuromuscular junction. Schwann cells. Curr Biol 1996; 6(9): 1054–1056. [DOI] [PubMed] [Google Scholar]
  • 177. Mars T, Yu KJ, Tang XM, et al. Differentiation of glial cells and motor neurons during the formation of neuromuscular junctions in cocultures of rat spinal cord explant and human muscle. J Comp Neurol 2001; 438(2): 239–251. [DOI] [PubMed] [Google Scholar]
  • 178. Barik A, Li L, Sathyamurthy A, et al. Schwann cells in neuromuscular junction formation and maintenance. J Neurosci 2016; 36(38): 9770–9781. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 179. Hörner SJ, Couturier N, Bruch R, et al. hiPSC-derived Schwann cells influence myogenic differentiation in neuromuscular cocultures. Cells 2021; 10(12): 3292. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 180. Lin CY, Yoshida M, Li LT, et al. iPSC-derived functional human neuromuscular junctions model the pathophysiology of neuromuscular diseases. JCI Insight 2019; 4(18): e124299–124299. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 181. Faustino Martins JM, Fischer C, Urzi A, et al. Self-organizing 3D human trunk neuromuscular organoids. Cell Stem Cell 2020; 27(3): 498–186.e6. [DOI] [PubMed] [Google Scholar]
  • 182. Ko CP, Robitaille R. Perisynaptic Schwann cells at the neuromuscular synapse: adaptable, multitasking glial cells. Cold Spring Harb Perspect Biol 2015; 7(10): a020503. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 183. Murakami T, Sunada Y. Schwann cell and the pathogenesis of Charcot-Marie-Tooth disease. Adv Exp Med Biol 2019; 1190: 301–321. [DOI] [PubMed] [Google Scholar]
  • 184. Singh T, Vazquez M. Time-dependent addition of neuronal and Schwann cells increase myotube viability and length in an in vitro tri-culture model of the neuromuscular junction. Regen Eng Transl Med 2019; 5(4): 402–413. [Google Scholar]
  • 185. Ishii T, Kawakami E, Endo K, et al. Myelinating cocultures of rodent stem cell line-derived neurons and immortalized Schwann cells. Neuropathology 2017; 37(5): 475–481. [DOI] [PubMed] [Google Scholar]
  • 186. Lin CY, Yoshida M, Li LT, et al. In vitro neuromuscular junction induced from human induced pluripotent stem cells. J Vis Exp 2020; (166). [DOI] [PubMed] [Google Scholar]
  • 187. Mazaleyrat K, Badja C, Broucqsault N, et al. Multilineage differentiation for formation of innervated skeletal muscle fibers from healthy and diseased human pluripotent stem cells. Cells 2020; 9(6): 1531. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 188. Urzi A, Lahmann I, Nguyen LVN, et al. Efficient generation of a self-organizing neuromuscular junction model from human pluripotent stem cells. Nat Commun 2023; 14(1): 8043. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 189. Smith AS, Luttrell SM, Dupont JB, et al. High-throughput, real-time monitoring of engineered skeletal muscle function using magnetic sensing. J Tissue Eng 2022; 13: 20417314221122127. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 190. Santhanam N, Kumanchik L, Guo X, et al. Stem cell derived phenotypic human neuromuscular junction model for dose response evaluation of therapeutics. Biomaterials 2018; 166: 64–78. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 191. Spijkers XM, Pasteuning-Vuhman S, Dorleijn JC, et al. A directional 3D neurite outgrowth model for studying motor axon biology and disease. Sci Rep 2021; 11(1): 2080. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 192. Costamagna D, Bastianini V, Corvelyn M, et al. Botulinum toxin treatment of adult muscle stem cells from children with cerebral palsy and hiPSC-derived neuromuscular junctions. Cells 2023; 12(16): 2072. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 193. Dinamarca MC, Colombo L, Brykczynska U, et al. Transmission-selective muscle pathology induced by the active propagation of mutant huntingtin across the human neuromuscular synapse. Front Mol Neurosci 2023; 16: 1287510. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 194. Leng Y, Li X, Zheng F, et al. Advances in in vitro models of neuromuscular junction: focusing on organ-on-a-chip, organoids, and biohybrid robotics. Adv Mater 2023; 35(41): e2211059. [DOI] [PubMed] [Google Scholar]
  • 195. Puttonen KA, Ruponen M, Naumenko N, et al. Generation of functional neuromuscular junctions from human pluripotent stem cell lines. Front Cell Neurosci 2015; 9: 473. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 196. Luttrell SM, Smith AST, Mack DL. Creating stem cell-derived neuromuscular junctions in vitro. Muscle Nerve 2021; 64(4): 388–403. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 197. Mao Q, Acharya A, Rodríguez-delaRosa A, et al. Tension-driven multi-scale self-organisation in human iPSC-derived muscle fibers. eLife 2022; 11: e76649. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 198. Umbach JA, Adams KL, Gundersen CB, et al. Functional neuromuscular junctions formed by embryonic stem cell-derived motor neurons. PLoS One 2012; 7(5): e36049. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 199. Marx U, Akabane T, Andersson TB, et al. Biology-inspired microphysiological systems to advance medicines for patient benefit and animal welfare. ALTEX 2020; 37(3): 365–394. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 200. Zhang K, Chowdary P, Cui B. Visualizing directional Rab7 and TrkA cotrafficking in axons by pTIRF microscopy. Methods Mol Biol 2015; 1298(3): 319–329. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 201. Afshar Bakooshli M, Lippmann ES, Mulcahy B, et al. A 3D culture model of innervated human skeletal muscle enables studies of the adult neuromuscular junction. eLife 2019; 8: e44530. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 202. Arjmand B, Kokabi Hamidpour S, Rabbani Z, et al. Organ on a chip: a novel in vitro biomimetic strategy in amyotrophic lateral sclerosis (ALS) modeling. Front Neurol 2021; 12: 788462. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 203. Duc P, Vignes M, Hugon G, et al. Human neuromuscular junction on micro-structured microfluidic devices implemented with a custom micro electrode array (MEA). Lab Chip 2021; 21(21): 4223–4236. [DOI] [PubMed] [Google Scholar]
  • 204. Kundu A, McCoy L, Azim N, et al. Fabrication and characterization of 3D printed, 3D microelectrode arrays for interfacing with a peripheral nerve-on-a-chip. ACS Biomater Sci Eng 2021; 7(7): 3018–3029. [DOI] [PubMed] [Google Scholar]
  • 205. Louit A, Beaudet MJ, Pépin R, et al. Differentiation of human induced pluripotent stem cells into mature and myelinating Schwann cells. Tissue Eng Part C: Methods 2023; 29(4): 134–143. [DOI] [PubMed] [Google Scholar]
  • 206. Chen Y, Sun T, Bi Z, et al. Premature termination codon readthrough in Drosophila varies in a developmental and tissue-specific manner. Sci Rep 2020; 10(1): 8485. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 207. Bayat N, Ebrahimi-Barough S, Ardakan MM, et al. Erratum to: differentiation of human endometrial stem cells into Schwann cells in fibrin hydrogel as 3D culture. Mol Neurobiol 2016; 53(10): 7177–7186. [DOI] [PubMed] [Google Scholar]
  • 208. Li X, Wang X, Wang X, et al. 3D bioprinted rat Schwann cell-laden structures with shape flexibility and enhanced nerve growth factor expression. 3 Biotech 2018; 8(8): 342. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 209. Chen Q, Zhang Z, Liu J, et al. A fibrin matrix promotes the differentiation of EMSCs isolated from nasal respiratory mucosa to myelinating phenotypical Schwann-like cells. Mol Cells 2015; 38(3): 221–228. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 210. Das M, Rumsey JW, Bhargava N, et al. A defined long-term in vitro tissue engineered model of neuromuscular junctions. Biomaterials 2010; 31(18): 4880–4888. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 211. Shahriyari M, Islam MR, Sakib SM, et al. Engineered skeletal muscle recapitulates human muscle development, regeneration and dystrophy. J Cachexia Sarcopenia Muscle 2022; 13(6): 3106–3121. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 212. Kim J, Kim W, Kim G. Scaffold with micro/nanoscale topographical cues fabricated using E-field-assisted 3D printing combined with plasma-etching for enhancing myoblast alignment and differentiation. Appl Surf Sci 2020; 509: 145404. [Google Scholar]
  • 213. Xu B, Magli A, Anugrah Y, et al. Nanotopography-responsive myotube alignment and orientation as a sensitive phenotypic biomarker for Duchenne muscular dystrophy. Biomaterials 2018; 183: 54–66. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 214. Happe CL, Tenerelli KP, Gromova AK, et al. Mechanically patterned neuromuscular junctions-in-a-dish have improved functional maturation. Mol Biol Cell 2017; 28(14): 1950–1958. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 215. Huang NF, Patel S, Thakar RG, et al. Myotube assembly on nanofibrous and micropatterned polymers. Nano Lett 2006; 6(3): 537–542. [DOI] [PubMed] [Google Scholar]
  • 216. van der Schaft DW, van Spreeuwel AC, Boonen KJ, et al. Engineering skeletal muscle tissues from murine myoblast progenitor cells and application of electrical stimulation. J Vis Exp 2013; (73): e4267. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 217. Tomori K, Ohta Y, Nishizawa T, et al. Low-intensity electrical stimulation ameliorates disruption of transverse tubules and neuromuscular junctional architecture in denervated rat skeletal muscle fibers. J Muscle Res Cell Motil 2010; 31(3): 195–205. [DOI] [PubMed] [Google Scholar]
  • 218. Charoensook SN, Williams DJ, Chakraborty S, et al. Bioreactor model of neuromuscular junction with electrical stimulation for pharmacological potency testing. Integr Biol 2017; 9(12): 956–967. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 219. Rimington RP, Fleming JW, Capel AJ, et al. Bioengineered model of the human motor unit with physiologically functional neuromuscular junctions. Sci Rep 2021; 11(1): 11695. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 220. Ang ET, Schäfer R, Baltensperger R, et al. Motor axonal sprouting and neuromuscular junction loss in an animal model of Charcot-Marie-Tooth disease. J Neuropathol Exp Neurol 2010; 69(3): 281–293. [DOI] [PubMed] [Google Scholar]
  • 221. Adebola AA, Di Castri T, He CZ, et al. Neurofilament light polypeptide gene N98S mutation in mice leads to neurofilament network abnormalities and a Charcot-Marie-Tooth Type 2E phenotype. Hum Mol Genet 2015; 24(8): 2163–2174. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 222. Boutary S, Caillaud M, El Madani M, et al. Squalenoyl siRNA PMP22 nanoparticles are effective in treating mouse models of Charcot-Marie-Tooth disease type 1 A. Commun Biol 2021; 4(1): 317–414. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 223. Attarian S, Young P, Brannagan TH, et al. A double-blind, placebo-controlled, randomized trial of PXT3003 for the treatment of Charcot-Marie-tooth type 1aa. Orphanet J Rare Dis 2021; 16(1): 433. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 224. Pisciotta C, Pareyson D. Gene therapy and other novel treatment approaches for Charcot-Marie-Tooth disease. Neuromuscul Disord 2023; 33(8): 627–635. [DOI] [PubMed] [Google Scholar]
  • 225. Schiza N, Georgiou E, Kagiava A, et al. Gene replacement therapy in a model of Charcot-Marie-Tooth 4C neuropathy. Brain 2019; 142(5): 1227–1241. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 226. Sargiannidou I, Kagiava A, Bashiardes S, et al. Intraneural GJB1 gene delivery improves nerve pathology in a model of X-linked Charcot–Marie–Tooth disease. Ann Neurol 2015; 78(2): 303–316. [DOI] [PubMed] [Google Scholar]
  • 227. Georgiou E, Kagiava A, Sargiannidou I, et al. AAV9-mediated SH3TC2 gene replacement therapy targeted to Schwann cells for the treatment of CMT4C. Mol Ther 2023; 31(11): 3290–3307. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 228. Kagiava A, Karaiskos C, Richter J, et al. AAV9-mediated Schwann cell-targeted gene therapy rescues a model of demyelinating neuropathy. Gene Ther 2021; 28(10–11): 659–675. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 229. Nizzardo M, Simone C, Rizzo F, et al. Gene therapy rescues disease phenotype in a spinal muscular atrophy with respiratory distress type 1 (SMARD1) mouse model. Adv Mar Sci 2015; 1(2): e1500078. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 230. Presa M, Bailey RM, Davis C, et al. AAV9-mediated FIG4 delivery prolongs life span in Charcot-Marie-Tooth disease type 4J mouse model. J Clin Investig 2021; 131(11): e137159. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 231. Yalvac ME, Amornvit J, Chen L, et al. AAV1.NT-3 gene therapy increases muscle fiber diameter through activation of mTOR pathway and metabolic remodeling in a CMT mouse model. Gene Ther 2018; 25(2): 129–138. [DOI] [PubMed] [Google Scholar]
  • 232. Lee JS, Lee JY, Song DW, et al. Targeted PMP22 TATA-box editing by CRISPR/Cas9 reduces demyelinating neuropathy of Charcot-Marie-Tooth disease type 1A in mice. Nucleic Acids Res 2020; 48(1): 130–140. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 233. Lee JS, Chang EH, Koo OJ, et al. Pmp22 mutant allele-specific siRNA alleviates demyelinating neuropathic phenotype in vivo. Neurobiol Dis 2017; 100: 99–107. [DOI] [PubMed] [Google Scholar]
  • 234. Gautier B, Hajjar H, Soares S, et al. AAV2/9-mediated silencing of PMP22 prevents the development of pathological features in a rat model of Charcot-Marie-Tooth disease 1 A. Nat Commun 2021; 12(1): 2356. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 235. Lee JS, Kwak G, Kim HJ, et al. miR-381 attenuates peripheral neuropathic phenotype caused by overexpression of PMP22. Exp Neurobiol 2019; 28(2): 279–288. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 236. Morelli KH, Griffin LB, Pyne NK, et al. Allele-specific RNA interference prevents neuropathy in Charcot-Marie-Tooth disease type 2D mouse models. J Clin Investig 2019; 129(12): 5568–5583. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 237. Zhao HT, Damle S, Ikeda-Lee K, et al. PMP22 antisense oligonucleotides reverse Charcot-Marie-Tooth disease type 1A features in rodent models. J Clin Investig 2018; 128(1): 359–368. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 238. Chumakov I, Milet A, Cholet N, et al. Polytherapy with a combination of three repurposed drugs (PXT3003) down-regulates Pmp22 over-expression and improves myelination, axonal and functional parameters in models of CMT1A neuropathy. Orphanet J Rare Dis 2014; 9(1): 201. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 239. Patzkó A, Bai Y, Saporta MA, et al. Curcumin derivatives promote Schwann cell differentiation and improve neuropathy in R98C CMT1B mice. Brain 2012; 135(Pt 12): 3551–3566. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 240. El Massry M, Msheik Z, El Masri T, et al. Improvement of Charcot-Marie-Tooth Phenotype with a nanocomplex treatment in two transgenic models of CMT1A. Biomater Res 2024; 28: 0009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 241. Fledrich R, Stassart RM, Klink A, et al. Soluble neuregulin-1 modulates disease pathogenesis in rodent models of Charcot-Marie-Tooth disease 1A. Nat Med 2014; 20(9): 1055–1061. [DOI] [PubMed] [Google Scholar]
  • 242. Scapin C, Ferri C, Pettinato E, et al. Enhanced axonal neuregulin-1 type-III signaling ameliorates neurophysiology and hypomyelination in a Charcot–Marie–Tooth type 1B mouse model. Hum Mol Genet 2019; 28(6): 992–1006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 243. Park S, Choi Y, Kwak G, et al. Application of differentiated human tonsil–derived stem cells to trembler-J mice. Muscle Nerve 2018; 57(3): 478–486. [DOI] [PubMed] [Google Scholar]
  • 244. Skov M, Ruijs TQ, Grønnebæk TS, et al. The ClC-1 chloride channel inhibitor NMD670 improves skeletal muscle function in rat models and patients with myasthenia gravis. Sci Transl Med 2024; 16(739): eadk9109. [DOI] [PubMed] [Google Scholar]
  • 245. Gidaro T, Grønnebæk TS, Cornwall C, et al. 103P NMD670, a first-in-class skeletal muscle ClC-1 inhibitor in Charcot-Marie-Tooth disease: the SYNAPSE-CMT phase 2 study. Neuromuscul Disord 2024; 43: 104441.573. [Google Scholar]
  • 246. Scurry AN, Heredia DJ, Feng CY, et al. Structural and functional abnormalities of the neuromuscular junction in the Trembler-J homozygote mouse model of congenital hypomyelinating neuropathy. J Neuropathol Exp Neurol 2016; 75(4): 334–346. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 247. de Mello CPP, Rumsey J, Slaughter V, et al. A human-on-a-chip approach to tackling rare diseases. Drug Discov Today 2019; 24(11): 2139–2151. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 248. Perel P, Roberts I, Sena E, et al. Comparison of treatment effects between animal experiments and clinical trials: systematic review. BMJ 2007; 334(7586): 197. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 249. Willadt S, Nash M, Slater CR. Age-related fragmentation of the motor endplate is not associated with impaired neuromuscular transmission in the mouse diaphragm. Sci Rep 2016; 6(1): 24849. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 250. Jones RA, Harrison C, Eaton SL, et al. Cellular and molecular anatomy of the human neuromuscular junction. Cell Rep 2017; 21(9): 2348–2356. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 251. Slater CR. The structure of human neuromuscular junctions: some unanswered molecular questions. Int J Mol Sci 2017; 18(10): 2183. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 252. Jan LY, Jan YN. L-glutamate as an excitatory transmitter at the Drosophila larval neuromuscular junction. J Physiol 1976; 262(1): 215–236. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 253. Yan Z, Cheng X, Li Y, et al. Sexually dimorphic neurotransmitter release at the neuromuscular junction in Adult Caenorhabditis elegans. Front Mol Neurosci 2022; 14: 780396. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 254. Haring AP, Sontheimer H, Johnson BN. Microphysiological human brain and neural systems-on-a-chip: potential alternatives to small animal models and emerging platforms for drug discovery and personalized medicine. Stem Cell Rev Rep 2017; 13(3): 381–406. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 255. Bouhy D, Timmerman V. Animal models and therapeutic prospects for Charcot–Marie–Tooth disease. Ann Neurol 2013; 74(3): 391–396. [DOI] [PubMed] [Google Scholar]
  • 256. Vila OF, Qu Y, Vunjak-Novakovic G. In vitro models of neuromuscular junctions and their potential for novel drug discovery and development. Expert Opin Drug Discov 2020; 15(3): 307–317. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 257. Moradi S, Mahdizadeh H, Šarić T, et al. Research and therapy with induced pluripotent stem cells (iPSCs): social, legal, and ethical considerations. Stem Cell Res Ther 2019; 10(1): 341. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 258. Xi H, Langerman J, Sabri S, et al. A human skeletal muscle atlas identifies the trajectories of stem and progenitor cells across development and from human pluripotent stem cells. Cell Stem Cell 2020; 27(1): 181–185. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 259. Ohi Y, Qin H, Hong C, et al. Incomplete DNA methylation underlies a transcriptional memory of somatic cells in human iPS cells. Nat Cell Biol 2011; 13(5): 541–549. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 260. Miller JD, Ganat YM, Kishinevsky S, et al. Human iPSC-based modeling of late-onset disease via progerin-induced aging. Cell Stem Cell 2013; 13(6): 691–705. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 261. Mertens J, Paquola ACM, Ku M, et al. Directly reprogrammed human neurons retain aging-associated transcriptomic signatures and reveal age-related nucleocytoplasmic defects. Cell Stem Cell 2015; 17(6): 705–718. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 262. Lee H, Lee HY, Lee BE, et al. Sequentially induced motor neurons from human fibroblasts facilitate locomotor recovery in a rodent spinal cord injury model. eLife 2020; 9: e52069. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 263. Thoma E, Merkl C, Heckel T, et al. Chemical conversion of human fibroblasts into functional Schwann cells. Stem Cell Reports 2014; 3(4): 539–547. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 264. Kim HS, Kim JY, Song CL, et al. Directly induced human Schwann cell precursors as a valuable source of Schwann cells. Stem Cell Res Ther 2020; 11(1): 257. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 265. Tang Y, Liu ML, Zang T, et al. Direct reprogramming rather than iPSC-based reprogramming maintains aging hallmarks in human motor neurons. Front Mol Neurosci 2017; 10: 359. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 266. Komor AC, Kim YB, Packer MS, et al. Programmable editing of a target base in genomic DNA without double-stranded DNA cleavage. Nature 2016; 533(7603): 420–424. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 267. Asmamaw Mengstie M, Teshome Azezew M, Asmamaw Dejenie T, et al. Recent advancements in reducing the Off-Target effect of CRISPR-Cas9 genome editing. Biologics 2024; 18: 21–28. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 268. Liang G, Zhang Y. Genetic and epigenetic variations in iPSCs: potential causes and implications for application. Cell Stem Cell 2013; 13(2): 149–159. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 269. Kim K, Zhao R, Doi A, et al. Donor cell type can influence the epigenome and differentiation potential of human induced pluripotent stem cells. Nat Biotechnol 2011; 29(12): 1117–1119. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 270. Daer RM, Cutts JP, Brafman DA, et al. The impact of chromatin dynamics on cas9-mediated genome editing in human cells. ACS Synth Biol 2017; 6(3): 428–438. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 271. Miny L, Maisonneuve BGC, Quadrio I, et al. Modeling neurodegenerative diseases using in vitro compartmentalized microfluidic devices. Front Bioeng Biotechnol 2022; 10: 919646. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 272. Hopkins AM, DeSimone E, Chwalek K, et al. 3D in vitro modeling of the central nervous system. Prog Neurobiol 2015; 125: 1–25. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 273. J Siney E, Kurbatskaya K, Chatterjee S, et al. Modelling neurodegenerative diseases in vitro: recent advances in 3D iPSC technologies. AIMS Cell Tissue Eng 2018; 2(1): 1–23. [Google Scholar]
  • 274. Remak R. Observationes Anatomicae Et Microscopicae De Systematis Nervosi Structura, Sumtibus Et Formis Reimerianis. 1838.
  • 275. Castro R, Taetzsch T, Vaughan SK, et al. Specific labeling of synaptic schwann cells reveals unique cellular and molecular features. eLife 2020; 9: e56935. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 276. Koike H, Iijima M, Mori K, et al. Nonmyelinating Schwann cell involvement with well-preserved unmyelinated axons in Charcot-Marie-Tooth disease type 1A. J Neuropathol Exp Neurol 2007; 66(11): 1027–1036. [DOI] [PubMed] [Google Scholar]
  • 277. Jensen C, Teng Y. Is it time to start transitioning from 2D to 3D cell culture? Front Mol Biosci 2020; 7: 33. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 278. Corrò C, Novellasdemunt L, Li VSW. A brief history of organoids. Am J Physiol Cell Physiol 2020; 319(1): C151–C165. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 279. Lorefice L, Pitzalis M, Murgia F, et al. Omics approaches to understanding the efficacy and safety of disease-modifying treatments in multiple sclerosis. Front Genet 2023; 14. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 280. Agrahari AK, George Priya Doss C. A computational approach to identify a potential alternative drug with its positive impact toward PMP22. J Cell Biochem 2017; 118(11): 3730–3743. [DOI] [PubMed] [Google Scholar]
  • 281. Pareyson D, Reilly MM, Schenone A, et al. Ascorbic acid in Charcot–Marie–Tooth disease type 1A (CMT-TRIAAL and CMT-TRAUK): a double-blind randomised trial. Lancet Neurol 2011; 10(4): 320–328. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 282. Désarnaud F, Do Thi AN, Brown AM, et al. Progesterone stimulates the activity of the promoters of peripheral myelin protein-22 and protein zero genes in Schwann cells. J Neurochem 1998; 71(4): 1765–1768. [DOI] [PubMed] [Google Scholar]
  • 283. Record CJ, Pipis M, Skorupinska M, et al. Whole genome sequencing increases the diagnostic rate in Charcot-Marie-Tooth disease. Brain 2024; 147(9): 3144–3156. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 284. Hasin Y, Seldin M, Lusis A. Multi-omics approaches to disease. Genome Biol 2017; 18(1): 83. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 285. Fralish Z, Lotz EM, Chavez T, et al. Neuromuscular development and disease: learning from in vitro and in vivo models. Front Cell Dev Biol 2021; 9: 764732. [DOI] [PMC free article] [PubMed] [Google Scholar]

Associated Data

This section collects any data citations, data availability statements, or supplementary materials included in this article.

Supplementary Materials

sj-docx-1-tej-10.1177_20417314241310508 – Supplemental material for From in vivo models to in vitro bioengineered neuromuscular junctions for the study of Charcot-Marie-Tooth disease

Supplemental material, sj-docx-1-tej-10.1177_20417314241310508 for From in vivo models to in vitro bioengineered neuromuscular junctions for the study of Charcot-Marie-Tooth disease by Camille Scherrer, Camille Loret, Nicolas Védrenne, Colman Buckley, Anne-Sophie Lia, Vincent Kermene, Franck Sturtz, Frédéric Favreau, Amandine Rovini and Pierre-Antoine Faye in Journal of Tissue Engineering


Articles from Journal of Tissue Engineering are provided here courtesy of SAGE Publications

RESOURCES