Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2025 Mar 22.
Published in final edited form as: Dev Dyn. 2021 Mar 29;251(1):10–25. doi: 10.1002/dvdy.333

BMP/TGF-β signaling as a modulator of neurodegeneration in ALS

Kathryn Russo 1,2, Kristi A Wharton 2,3
PMCID: PMC11929146  NIHMSID: NIHMS2060721  PMID: 33745185

Abstract

This commentary focuses on the emerging intersection between BMP/TGF-β signaling roles in nervous system function and the amyotrophic lateral sclerosis (ALS) disease state. Future research is critical to elucidate the molecular underpinnings of this intersection of the cellular processes disrupted in ALS and those influenced by BMP/TGF-β signaling, including synapse structure, neurotransmission, plasticity, and neuroinflammation. Such knowledge promises to inform us of ideal entry points for the targeted modulation of dysfunctional cellular processes in an effort to abrogate ALS pathologies. It is likely that different interventions are required, either at discrete points in disease progression, or across multiple dysfunctional processes which together lead to motor neuron degeneration and death. We discuss the challenging, but intriguing idea that modulation of the pleiotropic nature of BMP/TGF-β signaling could be advantageous, as a way to simultaneously treat defects in more than one cell process across different forms of ALS.

Keywords: actin dynamics, ALS, amyotrophic lateral sclerosis, BMP signaling, importin-β, LIMK, mad, neurodegeneration, nucleocytoplasmic transport, p38/MAPK, Smad, TGF-β signaling


Bone morphogenetic protein (BMP) and transforming growth factor-β (TGF-β)/Activin signaling pathways are known to impact synaptogenesis, axonal and dendritic growth, synaptic transmission, and neuronal survival. Given their involvement in nervous system development and function, there is considerable potential that disruptions in signaling activity and efficacy result in neurological disease. Moreover, it is conceivable that targeted manipulations of these pathways may be effective in abrogating cellular dysfunctions typical of nervous system disorders. The BMP and TGF-β/Activin intercellular signaling pathways represent two evolutionarily related branches, composed of a family of ligands, transmembrane receptors, and signal transducers, that display significant sequence and functional conservation, all together often referred to as TGF-β superfamily, or BMP/TGF-β, signaling.14 BMP/TGF-β signaling plays critical roles in a multitude of developmental and homeostatic processes in invertebrates and vertebrates alike.1,5 BMP ligands were first identified in demineralized bone with remarkable capabilities as inducers of bone deposition,6 and TGF-β as a growth factor that could stimulate anchorage-independent growth of fibroblasts in soft agar (reviewed in [Moses, 2016 #2016]). In addition to their role in the development and homeostasis of bone,7 BMPs are well known to provide instructional cues in many aspects of development, as well as in tissue homeostasis later in life. Similarly, TGF-β signaling regulates cell proliferation, differentiation, and apoptosis during development and also acts as a critical regulator of immune function.811 While these two related branches share some pathway elements, they often antagonize one another, inducing quite different outcomes in the same cellular or tissue environment.12,13

The roles of BMP/TGF-β signaling in nervous system function represent a substantial gap in our knowledge of how these remarkably pleiotropic pathways tightly regulate cell communication between both neuronal and non-neuronal components of dynamic neural circuits. Early roles for both pathway branches in neural patterning of both invertebrates and vertebrate embryos are well known,1418 but an appreciation of the nuances of BMP/TGF-β signaling in later events, such as axon guidance, synaptic growth, neural plasticity, and axonal regeneration has now gained traction with more research in these areas of study.1921 Likewise, more reports are appearing in the literature that document disruptions in BMP/TGF-β signaling in a variety of neurological diseases, implicating both branches in, Alzheimer’s disease, Parkinson’s disease, Angelman Syndrome, multiple sclerosis (MS), and amyotrophic lateral sclerosis (ALS).2225 Here, we will focus on our current understanding of the intersection of BMP/TGF-β signaling with ALS, a lethal neurodegenerative disease, for which diagnosis is challenging for the clinician, and devastating for the patient, as no effective treatments or therapeutics are yet available. We will not attempt a comprehensive review of ALS or BMP/TGF-β signaling, but will instead summarize what is currently known about how BMP/TGF-β signaling is altered in ALS, and how it may be possible for modulation of these pathways to alter the outcome of ALS. We raise questions that remain to be addressed, in hopes that they will help drive future research towards a more thorough understanding of the underlying mechanisms of the progressive degeneration associated with ALS. Importantly, we suggest avenues of research that may aid in the identification of both biomarkers and therapeutics, so desperately in need for overcoming the loss of neuron function, motor activity and ultimately life, all hallmarks of ALS.26,27

1 |. THE ALS PHENOTYPE

ALS is characterized by a progressive loss of motor control accompanied by the death of motor neurons whose cell bodies reside in the brain (upper motor neurons) and/or the spinal cord (lower motor neurons).28,29 Neuronal death leads to muscle wasting and atrophy, manifesting as weight loss, muscle weakness, fasciculations, and changes in gait or speech. As the disease progresses, everyday motor tasks become more difficult, and the degeneration of diaphragm innervation ultimately leads to respiratory failure and death.3032 In addition to motor loss, approximately 15% of ALS patients experience frontotemporal dementia (FTD), implicating ALS as a multisystem neurodegenerative disease, known as ALS/FTD.33 (Here, we refer to the ALS/FTD spectrum, as simply ALS.) The onset of ALS typically occurs in mid- to late in life, at approximately 50–70 years of age, at an incidence of ~2 cases per 100 000 individuals. Survival time post diagnosis typically ranges from 2 to 3 years.30,34 Currently, there are two Federal Drug Administration (FDA) approved treatments for ALS: Edaravone and Riluzole. These treatments slow the progression of the disease by no more than 6 months.35,36 With no known cure, or therapeutic that provides a considerable extension in lifespan, the diagnosis of ALS is devastating to the patient, their family members and caregivers, as a rapid irreversible decline is inevitable. There is a pressing need to identify treatments that reduce, stop degeneration, or even restore motor function.

ALS is classified as familial (fALS) or sporadic (sALS), depending on whether a patient does, or does not, have a family history of the disease, respectively. Clinically, both types of ALS patients present with nearly identical pathologies, and mutations in fALS genes are often identified in sALS patients.26,27,37 Familial ALS (10% of total ALS cases) has been associated with primarily autosomal dominant mutations in at least 24 different genes which encode a variety of proteins whose functions range from roles in RNA biology to fundamental cellular metabolism, some of which have previously been associated with FTD, reinforcing an ALS-FTD spectrum.31,3840 The remaining 90% of ALS cases, classified as sporadic, may arise from de novo mutations, previously unknown genetic mutations, and/or, non-genetic associations. Environmental factors that are disproportionately correlated with sALS cases include, but are not limited to, military service, low body fat, and a variety of external and chemical insults.27,41 ALS diagnosis is often reached when no other definitive explanation for the progressive loss of motor control is found.32 As previously noted, motor neuron death is a common feature in all ALS cases. Thus, a question plaguing the field for some time is: how do varied insults, either environmental or genetic, result in the same phenotype: the demise of motor neurons and loss of motor function?

Prior to motor neuron death, neuromuscular junction (NMJ) structure and function is compromised, axonal projections retract and degenerate, inclusions appear in the cytoplasm of aggregated proteins and RNAs, and an increased number of neuroinflammatory cells infiltrate the brain and spinal cord.4246 Both patients, and animal models of ALS, exhibit NMJ abnormalities at the morphological and functional level. ALS patients exhibit a dying back of motor axons in the spinal cord coupled with NMJ degeneration.47 A mouse model of ALS has shown impaired neurotransmission accompanied by disrupted end plates and innervation at the NMJ prior to evidence of motor deficits or motor neuron loss.48 From an electrophysiological perspective, a zebrafish model provided evidence of decreased quantal neurotransmission.49 Further, a Drosophila knock-in model of ALS showed a reduction in organismal locomotion accompanied by electrophysiological changes in the sensory feedback component of the motor circuit prior to significant NMJ abnormalities.50

2 |. ALS MODELS

Directed studies of the fALS genes most affected worldwide have enabled the research community to gain considerable insight into the molecular/cellular basis of cellular pathologies associated with ALS. These studies have made use of patient tissues, induced pluripotent stem cells (iPSCs) derived from patients, as well as animal models of ALS.5154 Genetic models in organisms, such as rodents (mice, Mus musculus and rats, Rattus norvegicus), nematode (Caenorhabditis elegans), fruit fly (Drosophila melanogaster), and zebrafish (Danio rerio), have been able to reproduce ALS neurodegenerative phenotypes, and in some cases, impaired locomotor function.52,5560 The majority of models to date induce ALS-like phenotypes by overexpressing a mutant form, or even wild type form, of a human, or an orthologous fALS gene, in select cells, that is, motor neurons or astrocytes.61,62 More recently, as technological advances have been made in genome editing, ALS mutations can be “knocked-in” to endogenous orthologous loci, enabling an animal model to replicate gene dosage and proper spatial and temporal gene expression patterns, while also recapitulating progressive motor deficits and the neurodegenerative pathogenesis of ALS.6366 Advances, that eliminate the use of heterologous systems, have allowed patient derived iPSCs to be differentiated into a cell type of choice, producing a “purified” culture of human cells that harbor the relevant genetic background, that is, an “ALS genome”.6769 The primary disadvantage, of course, is that the uniform population of differentiated iPSCs have lost their in vivo context. To rectify this shortcoming, 3D co-cultures and microfluidic systems aim to replicate the in vivo environment, addressing the importance of spatial context and cell-cell communication in ALS.7073

An important element in the function of neural circuits is the synapse which constitutes the juncture between the presynaptic neuron and its postsynaptic target. The structure of the synapse facilitates neurotransmission at the NMJ, for example, to enable the efficient release of neurotransmitter from vesicle fusions at the presynaptic membrane and their reception by neurotransmitter receptors localized on the opposing muscle cell membrane, leading to subsequent activation of contractile machinery. Motor neuron degeneration in ALS is accompanied by a disruption in NMJ structure and function. While differences exist in the organization and specific neurotransmitters used at NMJs of different species, that is, Drosophila vs mice vs human,74,75 many molecular details are similar, with findings of the Drosophila glutamatergic NMJ advancing our knowledge of glutamatergic synapse in the vertebrate CNS.76 Nevertheless, when considering the behavioral consequences of ALS-related disruptions at a mammalian NMJ vs that at the Drosophila NMJ, we need to keep in mind that more than one motor neuron can innervate a single muscle in Drosophila, while vertebrates exhibit a one neuron to one muscle fiber relationship. Further, different types of synaptic boutons are found in Drosophila, with no evidence of bouton diversity in vertebrates.77,78 Despite these differences Drosophila ALS models have successfully identified cellular pathways shown to be compromised in ALS patients or mammalian models, as well as revealing genetic suppressors that have translated to human models.55,7981

Regardless of classification (fALS or sALS) or heterogeneity in disease presentation, research using genetic animal models and patient derived cells has advanced the field in its conceptual understanding of the molecular underpinnings of ALS-associated degeneration. The extreme view that each mutated gene or environmental factor uniquely triggers a different process resulting in motor neuron death is unlikely. In reality, it is more likely that individual triggers (gene mutations and environmental insults) effect one of only a handful of biological processes, such as protein homeostasis and quality control, RNA biology, or cytoskeletal dynamics,82 that initially function somewhat independently of one another (Figure 1). As each potentially minor defect triggers their own disruption in a molecular pathway, multiple failings begin to impinge upon one another, together causing a more global imbalance and dysfunction of cellular physiology, resulting in the “perfect storm” of cellular pathologies characteristic of ALS (Figure 1). A better understanding of the molecular relationships between affected cellular pathways will reveal if common targets exist and whether a single therapeutic for all ALS patients is possible. If such interventions are not effective or cannot be made early enough, a combination of treatments or therapeutics, tailored to each of the initial dysfunctional processes will be necessary, ideally prior to significant decline, requiring a “personalized” approach to treatment.

FIGURE 1.

FIGURE 1

Conceptual depiction of cellular pathways impacted in ALS leading to motor neuron degeneration and death. (Top) Healthy motor neurons (green) in an environment of microglia (yellow) and astrocytes (blue). Gene mutations associated with ALS and/or environmental insults usually lead to disruptions in three general categories of cellular processes, that is, protein homeostasis, RNA biology, and cytoskeletal dynamics. The paths initiating cellular decline appear to trigger additional shared dysfunctions (mitochondrial or cellular metabolic dysfunction, impaired neurotransmission and the formation of protein and/or RNA aggregates and inclusions) that converge and accumulate, eventually leading to degeneration of the neuron, neuroinflammation (indicated by swollen microglia, yellow), astrocytosis (purple astrocytes), and finally, cell death

3 |. DO COMMON CELLULAR PATHWAYS UNDERLIE ALL FORMS OF ALS?

Four genes with the highest worldwide prevalence for fALS mutations are C9orf72, superoxide dismutase 1 (SOD1), Transactive Response DNA-binding protein 43 (TARDBP encoding TDP-43), and fused in sarcoma (FUS).39 Expansions of a GGGGCC (G4C2) hexanucleotide repeat sequence, which normally exist as ~10 copies within the intron of the chromosome 9 open reading frame 72 (C9orf72) gene, expand to 400 to 2000 repeats in most (39.6%) fALS cases, with concomitant haploinsufficiency of C9orf72.83,84 Of the people who carry the G4C2 hexanucleotide repeat expansion, 25.1% also exhibit cognitive decline and memory loss typical of FTD.85 Toxicity associated with the expansion of G4C2 repeats has also been attributed to multiple aspects of the hexanucleotide expansion. The increase in G4C2 repeats in RNA transcribed from the C9orf72 locus is thought to result in increased sequestration of protein binding partners from their normal cellular functions. Translation of the expansion of repeats via RAN (repeat associated non-AUG) translation results in long stretches of dipeptide repeats which exhibit a high propensity to for accumulations and aggregations within the cell.86,87 The second most common gene associated with ALS is superoxide dismutase 1 (SOD1). Mutations in SOD1 not only affect the protein’s ability to prevent the accumulation of superoxide radicals that wreak havoc on a number of cellular processes (DNA and RNA metabolism, mitochondrial function and protein folding),88,89 but also result in the aggregation of mutant SOD1 protein with wild type SOD1 protein, further diminishing the enzyme’s role in reactive oxygen species (ROS) abatement.90 SOD1 inclusions have been observed in both neurons and glia,91 with a more rapid disease progression associated with misfolded SOD1 in microglia.89 Mutations in TDP-43 and in FUS account for approximately 4.5% and 5% of all fALS cases, respectively. Both TDP-43 and FUS are RNA binding proteins and mutant forms of these proteins sequester transcriptional regulators, disrupting multiple aspects of RNA biology, including transcription, splicing, and RNA trafficking, all thought to contribute to ALS pathologies.37 Like SOD1, cytoplasmic inclusions are often associated with the co-localization of mutant FUS and TDP43 with stress granules.9296 A prominent disease signature also shared with other neurodegenerative diseases is the increase in stress granule formation and protein inclusions, indicative of general disruptions in both protein and RNA folding and turn-over.37 In roughly 97% of all ALS patients, TDP43 inclusions are found in cells of the spinal cord and motor cortex, irrespective of a fALS or sALS diagnosis. In general, TDP43 aggregates are thought to be toxic to neurons, but their potential protective role is important to consider.97 Whether cytoplasmic inclusions initiate decline, or they arise as a secondary consequence of upstream pathway dysfunctions, the question remains, does preventing their formation have a significant impact on disease progression and patient outcome? Relevant to this area of research is the implication that such molecular instabilities and aggregations could be responsible for the prion-like propagation of ALS pathologies, a molecular mechanism that could account for efficient spread from one cell to another and the rapid rate of disease progression.98100

Efforts to uncover common pathways at the root of ALS decline are starting to emerge from comparative studies. In three different iPSC-derived motor neuron models harboring fALS mutations in SOD1, FUS, and C9ORF72, an increase in hyperexcitability suggests that toxicity associated with superfluous glutamatergic activity could be a common mechanism leading to abnormal increases in calcium that ultimately contribute to neuron death.51 In a similar vein, attempts to identify genetic variants that suppress neurodegenerative phenotypes across multiple ALS models is a powerful approach for revealing shared pathways that underlie the demise of neural function.101103 There are also efforts being made to identify the molecular signatures associated with dysfunctions in ALS using RNA sequencing, proteomics, and metabolomic approaches.104106 The field would benefit from comparative omic analyses across different mutations.107 One important goal will be to compare presymptomatic and post-symptomatic samples to determine how changes in gene expression, protein modifications, and metabolites correlate with ALS pathologies, such as, hyperexcitability, synaptic and axonal instability, or cytoplasmic inclusions. Ideally, such studies will examine molecular changes in specific cell types in an in vivo context through time. Such studies of the shifts in molecular signatures early in the manifestation of ALS hold greater promise for (1) identifying potential biomarkers as predictors of disease risk, (2) determining whether commonalities exist early in disease onset, and (3) uncovering targets for the discovery of therapeutics that can prevent disease progression.

It is likely that ALS treatment and/or prevention will require targeting multiple molecular pathways, either simultaneously or in succession. However, it is conceivable that targeting a single pathway could be effective, if that pathway is pleiotropic, affecting multiple cell types and cellular processes, in which case modifying its activity or efficacy could attenuate the multi-pronged reach of ALS. BMP/TGF-β signaling is one such pathway, notorious for its involvement in a dizzying array of cellular functions.10,21,108 If BMP/TGF-β signaling is affected in multiple ALS pathologies, it may be possible to take advantage of its known regulatory mechanisms, to dial up, or dial back, signaling activity, modifying multiple dysfunctional processes in order to prevent neurodegeneration and restore motor function.

4 |. BMP/TGF-B SIGNALING OVERVIEW

TGF-β superfamily ligands are expressed broadly in the brain and spinal cord of the adult central nervous system, as well as in the key musculoskeletal components of the motor circuit.25,109 Extracellular ligands that comprise the large family of dimeric BMP/TGF-β signaling molecules, bind the extracellular domain of a heterotetrameric receptor complex, composed of type I and type II transmembrane serine/threonine kinases, to transduce a signal (Figure 2). Constitutively active type II receptor kinases phosphorylate and activate the type I receptor kinase, enabling its phosphorylation of cytoplasmic proteins that act to alter the receiving cell’s physiology. The pathway is highly regulated at all levels, from processing, maturation and availability of homo- or heterodimeric ligands,110115 to different constellations of receptors in the signaling complex,1 to a variety of intracellular effectors116 (Figure 2). In general, the BMP and TGF-β pathways are based on the evolutionary relatedness of ligands, type I and type II serine/threonine receptors, receptor mediated cytoplasmic Smads (R-Smad) proteins, in association with the co-Smad (Smad4) to regulate transcription in nuclei of responding cells. Phosphorylated R-Smads, the effector in Smad-dependent signaling, are generally distinct between the two branches with pSmad1/5/8 mediating signaling induced by BMP and growth differentiation factor (GDF) ligands, and pSmad2/3 mediating signaling initiated by TGF-β, Activin, and Nodal ligands,1 with inhibitory I-Smads able to negatively regulate Smad-dependent signaling.117,118 In some contexts, pSmads act independently of co-Smad4, such as in their role in chromatin remodeling and microRNA processing.119121 R-Smads are not the only effectors of BMP/TGF-β signaling. In Smad-independent signaling, a number of different kinases including p38 MAPK, PI3K, JNK, and LIMK, are activated in response to the ligand-bound receptor complex, extending the range of targets and cellular functions effected by BMP/TGF-β ligands.2,122

FIGURE 2.

FIGURE 2

Bmp/TGF-β signaling consists of two evolutionarily related branches with sequences and structural similarities between ligands, type I and type II receptors, and Smad effectors. The pathways are regulated at multiple levels including ligand maturation and availability, receptor complex composition and effector activity. Signal transduction depends on both Smad-dependent and Smad-independent (non-Smad) mediation. p38 and LIMK are two examples of non-Smad signaling effectors. The response of specific signaling components in ALS patients and/or models are shown as, elevated (up arrow) or reduced (down arrow) levels. The consequence of a direct experimental manipulation of BMP or TGF-β signaling in the context of an ALS model is highlighted in chartreuse (see text). Various properties of signaling that are altered or may be affected in ALS are indicated, that is, the different response of signaling (pMad) in different cellular compartments of the motor neuron, differential inclusion of pSmad2 vs pSmad3 dependent on importin-β, and the potential for BMP-mediated LIMK regulation of Actin dynamics in ALS

5 |. ALS-ASSOCIATED DISRUPTIONS IN BMP/TGF-B SIGNALING

Despite the identified roles for BMP/TGF-β signaling in nervous system function and the suggestions that dysregulated signaling may underlie neurodegenerative disorders, relatively few studies have directly addressed BMP/TGF-β signaling in ALS.1,22,23,25,123 A common pattern across the pathway has yet to emerge with regard to whether signaling is generally upregulated, or down-regulated, in specific tissues or cells. A preferential involvement of Smad-dependent vs Smad-independent signaling in ALS is also not yet clear. However, to date, the findings that report a change in the TGF-β branch in the ALS disease state, all report an upregulation. A preferential elevation in TGF-β1 ligand level, compared to those of TGF-β2 and 3, is seen in the spinal cord, muscles, and astrocytes of individuals carrying a SOD1-ALS mutation, as well as, in sALS patients.109,124126 An increase in pSmad2/3 has been detected in both neurons and glial cells of sALS and fALS patients, thus the TGF-β/Activin pathway is indeed activated in both of these cell types.125 Furthermore, pSmad2/3 was found to localize to TDP-43 aggregates in lower motor neurons of sALS patients.127 Significant increases in Smad2 RNA in muscle tissue of a transgenic SOD1G93A ALS mouse model126 and the orthologous dSmad2/Smox RNA in the central nervous system of a TDP-43 Drosophila knock-out model, while alone is not indicative of an increase in signaling per se, is suggestive of altered TGF-β/Activin signaling.128 Consistent with the idea that an upregulation of TGF-β/Activin signaling could underlie ALS pathologies, administration of a TGF-β inhibitor (SB-431542) was found to extend the survival of SOD1G93A ALS mice.124

With regard to BMP signaling, a simple up- or down-regulation, does not consistently correlate with ALS. In a rat SOD1H46R ALS model, an elevated level of BMP4 was detected in reactive astrocytes in the lumbar ventral spinal horns of late symptomatic animals,129 consistent with reports that particular BMP ligands, including BMP4, increase following neuronal injury, in animal models of spinal cord injury (SCI), traumatic brain injury (TBI), or demyelinating disorders, accompanied by a neuroinflammatory response (reviewed in [Hart, 2020 #1712]). In addition to an activation of Smad-dependent signaling (ie, increased pSmad1/5), astrocytes in the rat SOD1H46R ALS model showed an increase in p38MAPK, indicating an accompanying upregulation of Smad-independent signaling.129 As a means to block BMP signaling, intrathecal injection of Noggin, an extracellular antagonist of the BMP ligands, was shown to reduce markers of neuroinflammation in the rat SOD1H46R ALS model, and to help maintain myelin integrity in independent neuronal injury models.25

Proper synapse growth, neurotransmission, and synaptic plasticity in the Drosophila larval nervous system requires Smad-dependent BMP signaling, mediated by the BMP5/6/7 ortholog, gbb, the type II BMP receptor 2 (BMPR2), wit, and the Smad1/5/8, ortholog, Mad.123,130,131 The loss of gbb results in small NMJs, and overexpression of gbb and hyperactivation of BMP signaling produces larger NMJs, with an increase in synaptic transmission.123,132,133 Reduced locomotor activity and severe NMJ degeneration exhibited by a dSod1G85R knock-in ALS model, could both be abrogated by BMP signaling activation in response to the overexpression of gbb, or SaxA, a constitutively active form of the BMP type I receptor (ACVR1).50 In this case, ALS-associated defects were attenuated by activation of BMP signaling, in contrast to the results from rodent models where down-regulation of BMP signaling was able to reduce neuroinflammation, and an inhibition of TGF-β signaling extended survival in a SOD1 model.124,129 In both cases the Drosophila and rodent models are SOD1-ALS mutants, however the nature of the genetic manipulation of SOD1 differed. Does the consequence of BMP/TGF-β signaling manipulation depend on whether the ALS model is a knock-in vs overexpression model? Or does the ability of signaling to suppress various pathologies depend on the cell or tissue in which signaling is activated?

To address the latter question, cell-type specific and cell-autonomous activation of BMP signaling was tested for rescue of multiple phenotypes in the dSod1G85R Drosophila knock-in model.50 Specific activation of signaling in motor neurons, sensory neurons, or a subpopulation of sensory neurons, the proprioceptors, showed significant rescue, but activation in muscle or glial cells did not, indicating that BMP signaling rescue of motor dysfunction is cell-type specific. While underappreciated, the involvement of sensory neurons in ALS is known from the clinical standpoint, as sensory neuropathies are evident in patients.134136 Similarly, sensory neurons are found to be stress-sensitive in ALS animal models.137 The failure of muscle or glial-specific signaling to suppress degeneration could not be simply explained by detrimental effects of high signaling in those cells, as none were detected. The potential for ectopic activation of signaling, in and of itself, to produce negative consequences is an important consideration, especially in the experimental manipulations of potent pathways such as BMP and TGF-β/Activin signaling. For example, overexpression of BMP ligands in the Drosophila eye, a popular tissue to assay for modification of human disease genes, produces disorganized and roughened eyes, a phenotype similar to that seen when a variety of human disease genes are expressed in this tissue, precluding any assessment of modification.138,139 It is important to keep in mind the limitations of various assays and models, while also recognizing that in some cases, while one may not be able to directly assess molecular functions specific to a disease, the epistatic relationships between genes and/or pathways can be explored, and then tested further in other disease models.

Another important consideration with regard to BMP/TGF-β signaling is their potential for crosstalk in part due to the evolutionary relatedness of the signaling components comprising the two BMP and TGF-β/Activin branches. The ability of receptors and Smad effectors from one pathway to form non-functional complexes with those typically promoting signaling could underlie competition or antagonism between the two pathways, observed in a variety of developmental and disease contexts (reviewed in (Hudnall, 2016 #2009), and most recently in MS and neuroinflammation.13 Certainly, a single response, such as, up- or downregulation of signaling is not associated with either the ALS disease state, or the ability to ameliorate ALS-associated degeneration. More studies elucidating the action of each BMP and TGF-β/Activin signaling branch in ALS, and comparisons in the same cellular context, are necessary for future research.

6 |. IMPORTANCE OF SPATIAL AND COMPONENT-SPECIFIC ACTIVATION OF BMP SIGNALING

An interesting and potentially important aspect of BMP signaling that has emerged from detailed studies using the Drosophila larval motor circuit, is the presence of what appears to be two distinct pools of the phosphorylated Smad1/5/8 transducer, pMad, an indicator of active signaling. In filleted preparations of the intact wild type motor circuit, pMad is evident within motor neuron nuclei, in the ventral nerve cord (VNC), the spinal cord equivalent, as expected given its function as transcriptional regulator. pMad also accumulates in the presynaptic space of NMJs.140,141 The functional difference between “nuclear pMad” and “synaptic pMad” has not been fully elucidated; however, several studies suggest that roles for BMP signaling in synaptic growth and synaptic function are separable.131,133,141,142 Interestingly, in the dSod1G85R knock-in model, “synaptic pMad” is elevated indicating that BMP signaling is upregulated at the NMJ. However, no change in pMad levels is apparent in motor neuron nuclei.50 Yet in the context of dSod1G85R locomotor rescue by gbb (BMP5/6/7) expression, a significant increase in pMad within the VNC is observed but no change to the already elevated levels of “synaptic pMad”. A differential response of pMad in these two subcellular compartments is also apparent in two other Drosophila ALS models (TDP43 and VAPB), where “nuclear pMad” is also unaffected and “synaptic pMad” is changed, but in contrast to dSod1G85R, in these models synaptic pMad is reduced.143,144 In the TDP43 model, a degenerative phenotype elicited by the overexpression of wild type hTDP43 is associated with a defect in endosomal trafficking of the BMP type I Tkv receptor at the synapse thought to be responsible for the observed reduction in “synaptic pMad”.144 Currently, few details about differential regulation of BMP signal transduction in distinct subcellular domains are known, especially in a cell as asymmetric as a neuron. The precise function of ‘synaptic pMad’ is not known albeit its presence is associated with postsynaptic clustering of GluR receptors.141 Are there distinct subcellular pools of pSmad1/5/8 or pSmad2/3 in vertebrate motor neurons? A major gap in our knowledge is how BMP/TGF-β signaling is differentially regulated in discrete subcellular domains and how those differences manifest themselves with regard to neuronal function.

Two other aspects of BMP/TGF-β pathway regulation in the nervous system that require further investigation are the mechanisms underlying and consequences of ligand-specific signaling, and transduction through Smad-independent pathways. In the lumbar spinal cords of SOD1H46R rats, researchers have found that BMP4 levels are preferentially elevated, but levels of BMP2, 6, and 7 were unchanged.129 In the Drosophila nervous system, pMad-dependent BMP signaling is activated in multiple cell types: muscles, motor neurons, and glia. The BMP5/6/7 ortholog, gbb, and the BMP2/4 ortholog, dpp, are each expressed by distinct sets of cells, and contribute differentially to the activation of signaling in target cells.145,146 This kind of spatial difference in ligand production will be first regulated at the level of gene transcription, then by dimer formation, proprotein processing, post-translational modifications during trafficking, and secretion.110,147 As such, in addition to BMP ligand maturation, their structural properties, and availability for receptor binding are also known to alter signaling output,148,149 as demonstrated by the requirement for a specific Gbb proprotein proconvertase processing event that impacts CCAP (crustacean cardioactive peptide) neuropeptide production in CNS neurons.150 Differential reception of each ligand type is further impacted by the presence/absence of different receptor types and their contribution to receptor complexes expressed at the cell. In addition to these forms of regulation, crosstalk between pathways is evident, as in the case of glial-expressed Activin, encoded by maverick (mav), where its contribution to the complexity of BMP signaling regulation and its influence on NMJ growth appears to occur through its modulation of gbb gene expression in muscles.151 Thus, the constellation of ligands, their bioavailability, and composition of receptor complexes, contribute to the tight regulation of signaling in neuronal function, providing a mechanism for a fine level of control.

7 |. INVOLVEMENT OF SMAD-INDEPENDENT SIGNALING

In addition to our growing knowledge of varied roles for different ligand forms, evidence for their ability to initiate signaling via Smad-independent pathways is also growing. While our current understanding is limited, links between BMP/TGF-β signaling and p38/MAPK in the nervous system are emerging. BMPs are important in inducing neuronal differentiation of PC12 cells, a process also shown to require p38 activation.152 Reduced neurodegeneration associated with an upregulation in BMP7 has been linked to an activation of pro survival p38/MAPK signaling in neurons.153 Hyper-phosphorylation of neurofilaments is a hallmark of ALS and the finding that p38 phosphorylates neurofilaments in culture is intriguing.154 Furthermore, p38 activation is observed in both patients and SOD1-ALS mutant mouse models,155,156 and inhibitors of p38 have been shown to correct a defect in retrograde axonal transport present in SOD1G93A mice.157 To date, no direct connections have been made between upstream BMP/TGF-β signaling components, such as ligands and receptors, with p38 activity in ALS. p38 is a known regulator of Rab5, a key factor in early endosome biology,158,159 and given the disrupted endocytic trafficking of the BMP Tkv receptor proposed in a Drosophila TDP43 model,144 it would be interesting to measure p38 levels in that context.

Smad-independent BMP signaling has also been implicated in the regulation of actin dynamics at the tips of dendrites and axons in part through the direct association of LIMK with the cytoplasmic domain of the BMPRII receptor.160 Stimulation of LIMK occurs in response to BMP ligand-receptor interactions in the absence of Smad involvement. As discussed below, activation of LIMK in response to BMP signaling has the potential to alter actin polymerization dynamics in both axons and dendrites.

8 |. CELLULAR PROCESSES AFFECTED BY BOTH ALS AND BMP/TGF-B SIGNALING

Overall, a better understanding of how components and mediators of BMP/TGF-β signaling are affected across a number of ALS mutations and models will be important for constructing the array of molecular consequences that link the two. When considering the possibility of amelioration of ALS-associated neurodegeneration by BMP/TGF-β pathway activation, it is also important to consider the overlap of cellular processes in which TGF-β/BMP signaling is known to act and those that are dysfunctional at early stages of ALS. In order to address the effectiveness of BMP/TGF-β signaling as a modulator of ALS, we must consider two important questions: Does amelioration of a single cellular process by BMP/TGF-β signaling modulation, provide phenotypic rescue across a number of different ALS mutations and models? Can the functions of multiple defective processes, that together contribute to neuronal degeneration in ALS, be restored by increasing or decreasing BMP/TGF-β signaling?

There are several areas for intersection of BMP/TGF-β signaling in processes commonly affected in ALS that are certainly worthy of further investigation. The role for BMP signaling in regulating actin dynamics through interactions between LIMK with the cytoplasmic domain of the type II receptor BMPRII receptor160 is of interest. BMP7 stimulates LIMK activity and dendrite outgrowth, through a Smad-independent pathway that is activated within a specific subcellular domain. Phosphorylation of cofilin by LIMK inactivates its ability to sever and depolymerize F-actin to enrich the G-actin pool. The yang to cofilin’s yin, is profilin, which facilitates the polymerization of actin filaments by acting on the G-actin pool, balancing the actions of cofilin to control actin filament dynamics. Mutations in profilin have been identified in ALS patients161 implicating a disruption in actin polymerization as a mechanistic failure that could result in defective axon and dendrite growth and transport.162,163 Consistent with such a model, phosphorylation of cofilin is enhanced in both ALS patients and C9orf72-ALS animal models. Further, C9orf72 gene function can modulate LIMK activity.164 The conservation of process is evident in Drosophila where the interaction between the orthologous BMPRII receptor, Wit, and LIMK, is critical for NMJ stabilization,165 and LIMK-dependent BMP signaling is required for axonal growth in the mushroom body.166

Another cellular process impacted by both BMP/TGF-β signaling and ALS that requires more investigation is nucleocytoplasmic transport. Disruptions in nucleocytoplasmic transport in ALS are multifold and involve a failure in the proper nuclear localization of RNA-binding proteins, resulting in cytoplasmic aggregation of factors such as, TDP43, and peptides composed of dipeptide repeats (DPRs) produced by RAN translation of the G4C2 expansions in C9orf72-ALS. Abnormal nuclear membrane structure was first observed in spinal motor neurons of fALS and sALS patients with SOD1 mutations.167 An association of the expanded G4C2 hexanucleotide repeat RNA with RanGAP, a mediator of nuclear pore dynamics, has been documented in a Drosophila ALS model, iPSC-derived neurons, as well as in C9orf72 ALS patient tissue and shown to rescue ALS-associated defects when over expressed.55,168,169 Importin-β is a nuclear import receptor that regulates transport through RanGAP-regulated nuclear pores from the cytoplasm to the nucleus of a number of RNA-binding proteins, such as TDP-43 and FUS which exhibit aggregation-prone properties in ALS.170,171 Mutations in importin-β result in defective NMJ structure and synaptic transmission in Drosophila.140 Synaptic pMad is reduced in importin-β mutant animals and a genetic interaction identified between importin-β mutations and mutations in BMP signaling components, highlight the connection between importin-β function and BMP signaling in the motor neuron.140 How importin-β, as a regulator of nucleocytoplasmic transport, specifically interferes with synaptic pMad abundance and not nuclear pMad levels in not clear. One key towards understanding the impact of BMP signaling on its effectors in different cellular domains may be an elucidation of how the cytoplasmic and nuclear transport of different pMad pools is regulated, coupled with what controls its localization at the NMJ, in wild type and diseased motor neurons. An intriguing parallel is the observation in both sALS and SOD1 fALS that the differential localization of pSmad2 vs pSmad3 to motor neuron inclusions is dependent on importin-β function.125

9 |. EXCITING NEW RESEARCH TO BE DONE

In conclusion, the intersection between BMP-TGF-β signaling roles in nervous system function and the cellular disturbances arising from ALS is becoming more robust. Research directed at understanding the molecular underpinnings responsible for fine tuning signaling output will reveal how disruptions or breaks in this system arise in a diseased state. Importantly, this knowledge will identify entry points for signaling manipulation through targeted modulation, as well as markers of disease risk or onset that have the potential to aid in diagnosis. Important questions to be addressed include: Can modulation of BMP/TGF-β signaling alleviate dysfunction of multiple cellular processes affected by ALS? And, is modulation of the pleiotropic nature of BMP/TGF-β signaling effective in its suppression of neurodegeneration across all forms of ALS? We identify several particularly intriguing connections between processes regulated by BMP/TGF-β signaling, such as the dynamics of actin polymerization, and nucleocytoplasmic transport, with their known dysfunction in ALS. Research elucidating the mechanistic basis of these intersections of ALS compromised cellular processes which roles for BMP/TGF-β signaling, is a top priority.

ACKNOWLEDGMENTS

K.A.W. graciously acknowledges funding from The Judith and Jean Pape Adams Foundation, ALS Finding a Cure, Biogen, Muscular Dystrophy Association, NIH GM068118, and The Rothberg Family Fund in Cognitive Sciences whose support was instrumental in our efforts to explore new areas of research. K.R. was supported by funds from NIH T32MH020068 and The Muscular Dystrophy Association. We apologize for the omission of relevant references due to space limitations.

Funding information

ALS Finding a Cure; Judith and Jean Pape Adams Charitable Foundation; Muscular Dystrophy Association; National Institute of General Medical Sciences, Grant/Award Number: GM068118; Rothberg Family Fund for Cognitive Sciences

REFERENCES

  • 1.Wang RN, Green J, Wang Z, et al. Bone morphogenetic protein (BMP) signaling in development and human diseases. Genes Dis. 2014;1(1):87–105. 10.1016/j.gendis.2014.07.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.Derynck R, Zhang YE. Smad-dependent and Smad-independent pathways in TGF. Nature. 2003;425(6958): 577–584. [DOI] [PubMed] [Google Scholar]
  • 3.Tzavlaki K, Moustakas A. TGF-β signaling. Biomolecules. 2020;10(3):487–525. 10.3390/biom10030487. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Bragdon B, Moseychuk O, Saldanha S, King D, Julian J, Nohe A. Bone morphogenetic proteins: a critical review. Cell Signal. 2011;23(4):609–620. 10.1016/j.cellsig.2010.10.003. [DOI] [PubMed] [Google Scholar]
  • 5.Chen G, Xu H, Yao Y, et al. BMP signaling in the development and regeneration of cranium bones and maintenance of calvarial stem cells. Front Cell Dev Biol. 2020;8(March):1–9. 10.3389/fcell.2020.00135. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Urist MR, Mikulski A, Lietze A. Solubilized and insolubilized bone morphogenetic protein. Proc Natl Acad Sci U S A. 1979; 76(4):1828–1832. 10.1073/pnas.76.4.1828. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Halloran D, Durbano HW, Nohe A. Developmental review bone morphogenetic protein-2 in development and bone homeostasis. J Dev Biol. 2020;8(3):28–30. 10.3390/JDB8030019. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Beyer TA, Narimatsu M, Weiss A, David L, Wrana JL. The TGFβ superfamily in stem cell biology and early mammalian embryonic development. Biochim Biophys Acta Gen Subjects. 2013;1830(2):2268–2279. 10.1016/j.bbagen.2012.08.025. [DOI] [PubMed] [Google Scholar]
  • 9.David CJ, Massagué J. Contextual determinants of TGFβ action in development, immunity and cancer. Nat Rev Mol Cell Biol. 2018;19(7):419–435. 10.1038/s41580-018-0007-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Wu MY, Hill CS. TGF-β superfamily signaling in embryonic development and homeostasis. Dev Cell. 2009;16(3):329–343. 10.1016/j.devcel.2009.02.012. [DOI] [PubMed] [Google Scholar]
  • 11.Morikawa M, Derynck R, Miyazono K. TGF-β and the TGF-β family: context-dependent roles in cell and tissue physiology. Cold Spring Harbor Perspect Biol. 2016;8(5):a021873. 10.1101/cshperspect.a021873. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Hudnall AM, Arthur JW, Lowery JW. Clinical relevance and mechanisms of antagonism between the BMP and Activin/TGF-beta signaling pathways. J Am Osteopath Assoc. 2016;116(7):452–461. 10.7556/jaoa.2016.089. [DOI] [PubMed] [Google Scholar]
  • 13.Sotiropoulos MG, Chitnis T. Opposing and potentially antagonistic effects of BMP and TGF-β in multiple sclerosis: the “yin and Yang” of neuro-immune signaling. J Neuroimmunol. 2020;347(July):577358. 10.1016/j.jneuroim.2020.577358. [DOI] [PubMed] [Google Scholar]
  • 14.Zinski J, Tajer B, Mullins MC. TGF-β family signaling in early vertebrate development. Cold Spring Harb Perspect Biol. 2018; 10(6):1–76. 10.1101/cshperspect.a033274. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Carron C, Shi DL. Specification of anteroposterior axis by combinatorial signaling during Xenopus development. Wiley Interdiscip Rev Dev Biol. 2016;5(2):150–168. 10.1002/wdev.217. [DOI] [PubMed] [Google Scholar]
  • 16.Leung B, Shimeld SM. Evolution of vertebrate spinal cord patterning. Dev Dyn. 2019;248(11):1028–1043. 10.1002/dvdy.77. [DOI] [PubMed] [Google Scholar]
  • 17.Bond AM, Bhalala OG, Kessler JA. The dynamic role of bone morphogenetic proteins in neural stem cell fate and maturation. Dev Neurobiol. 2012;72(7):1068–1084. 10.1002/dneu.22022. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Andrews MG, Kong J, Novitch BG, Butler SJ. New perspectives on the mechanisms establishing the dorsal-ventral axis of the spinal cord. Curr Top Dev Biol. 2019;132:417–450. 10.1016/bs.ctdb.2018.12.010. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Keshishian H, Kim YS. Orchestrating development and function: retrograde BMP signaling in the drosophila nervous system. Trends Neurosci. 2004;27(3):143–147. 10.1016/j.tins.2004.01.004. [DOI] [PubMed] [Google Scholar]
  • 20.Zhong J, Zou H. BMP signaling in axon regeneration. Curr Opin Neurobiol. 2014;27:127–134. 10.1016/j.conb.2014.03.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Hegarty SV, O’Keeffe GW, Sullivan AM. BMP-Smad 1/5/8 signalling in the development of the nervous system. Prog Neurobiol. 2013;109:28–41. 10.1016/j.pneurobio.2013.07.002. [DOI] [PubMed] [Google Scholar]
  • 22.Bayat V, Jaiswal M, Bellen HJ. The BMP signaling pathway at the drosophila neuromuscular junction and its links to neurodegenerative diseases. Curr Opin Neurobiol. 2011;21(1):182–188. 10.1016/j.conb.2010.08.014. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Kashima R, Hata A. The role of TGF-β superfamily signaling in neurological disorders [review]. Acta Biochim Biophys Sin. 2018;50(1):106–120. 10.1093/abbs/gmx124. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Costa C, Eixarch H, Martínez-Sáez E, et al. Expression of bone morphogenetic proteins in multiple sclerosis lesions. Am J Pathol. 2019;189(3):665–676. 10.1016/j.ajpath.2018.11.007. [DOI] [PubMed] [Google Scholar]
  • 25.Hart CG, Karimi-Abdolrezaee S. Bone morphogenetic proteins: new insights into their roles and mechanisms in CNS development, pathology and repair. Exp Neurol. 2020;334: 113455. 10.1016/j.expneurol.2020.113455. [DOI] [PubMed] [Google Scholar]
  • 26.Taylor JP, Brown RH Jr, Cleveland DW. Decoding ALS: from genes to mechanism. Nature. 2016;539(7628):197–206. 10.1038/nature20413. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Zarei S, Carr K, Reiley L, et al. A comprehensive review of amyotrophic lateral sclerosis. Surg Neurol Int. 2015;6:171. 10.4103/2152-7806.169561. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Ban J, Sámano C, Mladinić M, Munitić I. Glia in amyotrophic lateral sclerosis and spinal cord injury: common therapeutic targets. Croat Med J. 2019;60(2):109–120. 10.3325/cmj.2019.60.109. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Kang SH, Li Y, Fukaya M, et al. Degeneration and impaired regeneration of gray matter oligodendrocytes in amyotrophic lateral sclerosis. Nat Neurosci. 2013;16(5):571–579. 10.1038/nn.3357. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Brown RH, Al-Chalabi A. Amyotrophic lateral sclerosis. N Engl J Med. 2017;377(2):162–172. 10.1056/NEJMra1603471. [DOI] [PubMed] [Google Scholar]
  • 31.Masrori P, Van Damme P. Amyotrophic lateral sclerosis: a clinical review. Eur J Neurol. 2020;27(10):1918–1929. 10.1111/ene.14393. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Salameh JS, Brown RH Jr, Berry JD. Amyotrophic lateral sclerosis: review. Semin Neurol. 2015;35(4):469–476. 10.1055/s-0035-1558984. [DOI] [PubMed] [Google Scholar]
  • 33.Shuo-Chien Ling MPDWC. Converging mechanisms in ALS and FTD. Physiol Behav. 2016;176(1):139–148. 10.1016/j.neuron.2013.07.033.Converging. [DOI] [Google Scholar]
  • 34.Taylor LM, McMillan PJ, Liachko NF, et al. Pathological phosphorylation of tau and TDP-43 by TTBK1 and TTBK2 drives neurodegeneration. Mol Neurodegener. 2018;13(1):7. 10.1186/s13024-018-0237-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Khairoalsindi OA, Abuzinadah AR. Maximizing the survival of amyotrophic lateral sclerosis patients: current perspectives. Neurol Res Int. 2018;2018:1–12. 10.1155/2018/6534150. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Miller RG, Mitchell JD, Moore DH. Riluzole for amyotrophic lateral sclerosis (ALS)/motor neuron disease (MND). Cochrane Database Syst Rev. 2012;(3):1–28. 10.1002/14651858.CD001447.pub3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Prasad A, Bharathi V, Sivalingam V, Girdhar A, Patel BK. Molecular mechanisms of TDP-43 Misfolding and pathology in amyotrophic lateral sclerosis. Front Mol Neurosci. 2019;12: 25–25. 10.3389/fnmol.2019.00025. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Ling SC, Polymenidou M, Cleveland DW. Converging mechanisms in als and FTD: disrupted RNA and protein homeostasis. Neuron. 2013;79(3):416–438. 10.1016/j.neuron.2013.07.033. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Kirby J, Al Sultan A, Waller R, Heath P. The genetics of amyotrophic lateral sclerosis: current insights. Degener Neurol Neuromusc Dis. 2016;6:49–49. 10.2147/DNND.S84956. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Ghasemi M, Brown RH Jr. Genetics of amyotrophic lateral sclerosis. Cold Spring Harb Perspect Med. 2018;8(5):a024125. 10.1101/cshperspect.a024125. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Armon C Excess incidence of ALS in young gulf war veterans. Neurology. 2004;63(10):1986–1987; author reply 1986–7. 10.1212/wnl.63.10.1986. [DOI] [PubMed] [Google Scholar]
  • 42.Wong PC, Pardo CA, Borchelt DR, et al. An adverse property of a familial ALS-linked SOD1 mutation causes motor neuron disease characterized by vacuolar degeneration of mitochondria. Neuron. 1995;14(6):1105–1116. 10.1016/0896-6273(95)90259-7. [DOI] [PubMed] [Google Scholar]
  • 43.Bruijn LI, Miller TM, Cleveland DW. Unraveling the mechanisms involved in motor neuron degeneration in ALS. Annu Rev Neurosci. 2004;27:723–749. 10.1146/annurev.neuro.27.070203.144244. [DOI] [PubMed] [Google Scholar]
  • 44.Fischer LR, Culver DG, Tennant P, et al. Amyotrophic lateral sclerosis is a distal axonopathy: evidence in mice and man [comparative study]. Exp Neurol. 2004;185(2):232–240. [DOI] [PubMed] [Google Scholar]
  • 45.Mackenzie IRA, Rademakers R, Neumann M. TDP-43 and FUS in amyotrophic lateral sclerosis and frontotemporal dementia. Lancet Neurol. 2010;9(10):995–1007. 10.1016/S1474-4422(10)70195-2. [DOI] [PubMed] [Google Scholar]
  • 46.Philips T, Robberecht W. Neuroinflammation in amyotrophic lateral sclerosis: role of glial activation in motor neuron disease. Lancet Neurol. 2011;10(3):253–263. 10.1016/S1474-4422(11)70015-1. [DOI] [PubMed] [Google Scholar]
  • 47.Moloney EB, De Winter F, Verhaagen J, et al. ALS as a distal axonopathy: molecular mechanisms affecting neuromuscular junction stability in the presymptomatic stages of the disease. Front Neurosci. 2014;252:1–18. 10.3389/fnins.2014.00252. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Chand KK, Lee KM, Lee JD, et al. Defects in synaptic transmission at the neuromuscular junction precede motor deficits in a TDP-43Q331K transgenic mouse model of amyotrophic lateral sclerosis. FASEB J. 2018;32(5):2676–2689. 10.1096/fj.201700835R. [DOI] [PubMed] [Google Scholar]
  • 49.Armstrong GA, Drapeau P. Calcium channel agonists protect against neuromuscular dysfunction in a genetic model of TDP-43 mutation in ALS. J Neurosci. 2013;33(4):1741–1752. 10.1523/JNEUROSCI.4003-12.2013. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Held A, Major P, Sahin A, Reenan RA, Lipscombe D, Wharton KA. Circuit dysfunction in SOD1-ALS model first detected in sensory feedback prior to motor neuron degeneration is alleviated by BMP signaling. J Neurosci. 2019;39(12): 2347–2364. 10.1523/JNEUROSCI.1771-18.2019. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Wainger BJ, Kiskinis E, Mellin C, et al. Intrinsic membrane hyperexcitability of amyotrophic lateral sclerosis patient-derived motor neurons. Cell Rep. 2014;7(1):1–11. 10.1016/j.celrep.2014.03.019. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Watson MR, Lagow RD, Xu K, Zhang B, Bonini NM. A drosophila model for amyotrophic lateral sclerosis reveals motor neuron damage by human SOD1. J Biol Chem. 2008;283(36): 24972–24981. 10.1074/jbc.M804817200. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Han-Xiang D, Hujun J, Ronggen F, et al. Molecular dissection of ALS-associated toxicity of SOD1 in transgenic mice using an exon-fusion approach. Hum Mol Genet. 2008;17(15): 2310–2319. 10.1093/hmg/ddn131. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Philips T, Rothstein JD. Rodent models of amyotrophic lateral sclerosis. Curr Protoc Pharmacol. 2015;69:5 67 1–5 67 21. 10.1002/0471141755.ph0567s69. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.Zhang K, Donnelly CJ, Haeusler AR, et al. The C9orf72 repeat expansion disrupts nucleocytoplasmic transport. Nature. 2015; 525(7567):56–61. 10.1038/nature14973. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.Ash PE, Zhang YJ, Roberts CM, et al. Neurotoxic effects of TDP-43 overexpression in C. elegans. Hum Mol Genet. 2010;19 (16):3206–3218. 10.1093/hmg/ddq230. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Bories C, Amendola J, Lamotte D’Incamps B, Durand J. Early electrophysiological abnormalities in lumbar motoneurons in a transgenic mouse model of amyotrophic lateral sclerosis. Eur J Neurosci. 2007;25(2):451–459. 10.1111/j.1460-9568.2007.05306.x. [DOI] [PubMed] [Google Scholar]
  • 58.Yuva-Aydemir Y, Almeida S, Gao FB. Insights into C9ORF72-related ALS/FTD from drosophila and iPSC models. Trends Neurosci. 2018;41(7):457–469. 10.1016/j.tins.2018.04.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Van Damme P, Robberecht W, Van Den Bosch L. Modelling amyotrophic lateral sclerosis: progress and possibilities. Dis Models Mech. 2017;10(5):537–549. 10.1242/dmm.029058. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Picher-Martel V, Valdmanis PN, Gould PV, Julien JP, Dupré N. From animal models to human disease: a genetic approach for personalized medicine in ALS. Acta Neuropathol Commun. 2016;4(1):1–29. 10.1186/s40478-016-0340-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.Farg MA, Sundaramoorthy V, Sultana JM, et al. C9ORF72, implicated in amytrophic lateral sclerosis and frontotemporal dementia, regulates endosomal trafficking. Hum Mol Genet. 2014;23(13):3579–3595. 10.1093/hmg/ddu068. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Wang H, Yi J, Li X, Xiao Y, Dhakal K, Zhou J. ALS-associated mutation SOD1G93A leads to abnormal mitochondrial dynamics in osteocytes. Bone. 2018;106:126–138. 10.1016/j.bone.2017.10.010. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.Şahin A, Held A, Bredvik K, et al. Human SOD1 ALS mutations in a drosophila Knock-in model cause severe phenotypes and reveal dosage-sensitive gain- and loss-of-function components. Genetics. 2017;205(2):707–723. 10.1534/genetics.116.190850. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64.Baskoylu SN, Yersak J, O’Hern P, et al. Single copy/knock-in models of ALS SOD1 in C. elegans suggest loss and gain of function have different contributions to cholinergic and glutamatergic neurodegeneration. PLoS Genet. 2018;14(10): e1007682. 10.1371/journal.pgen.1007682. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65.Watanabe S, Oiwa K, Murata Y, et al. ALS-linked TDP-43M337V knock-in mice exhibit splicing deregulation without neurodegeneration. Mol Brain. 2020;13(1):13–16. 10.1186/s13041-020-0550-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66.Dong W, Zhang L, Sun C, et al. Knock in of a hexanucleotide repeat expansion in the C9orf72 gene induces ALS in rats. Anim Models Exp Med. 2020;3(3):237–244. 10.1002/ame2.12129. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67.Lee S, Huang EJ. Modeling ALS and FTD with iPSC-derived neurons. Brain Res. 1656;1(2017):88–97. 10.1016/j.brainres.2015.10.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.Fujimori K, Ishikawa M, Otomo A, et al. Modeling sporadic ALS in iPSC-derived motor neurons identifies a potential therapeutic agent. Nat Med. 2018;24(10):1579–1589. 10.1038/s41591-018-0140-5. [DOI] [PubMed] [Google Scholar]
  • 69.Hawrot J, Imhof S, Wainger BJ. Modeling cell-autonomous motor neuron phenotypes in ALS using iPSCs. Neurobiol Dis. 2020;134(November 2019):104680–104680. 10.1016/j.nbd.2019.104680. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.Zahavi EE, Ionescu A, Gluska S, Gradus T, Ben-Yaakov K, Perlson E. A compartmentalized microfluidic neuromuscular co-culture system reveals spatial aspects of GDNF functions. J Cell Sci. 2015;128(6):1241–1252. 10.1242/jcs.167544. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71.Osaki T, Sivathanu V, Kamm RD. Engineered 3D vascular and neuronal networks in a microfluidic platform. Sci Rep. 2018;8:5168–5168. 10.1038/s41598-018-23512-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.Kunze A, Lengacher S, Dirren E, Aebischer P, Magistretti PJ, Renaud P. Astrocyte-neuron co-culture on microchips based on the model of SOD mutation to mimic ALS. Integr Biol (United Kingdom). 2013;5(7):964–975. 10.1039/c3ib40022k. [DOI] [PubMed] [Google Scholar]
  • 73.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;4:5847. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74.Slater CR. The functional organization of motor nerve terminals. Prog Neurobiol. 2015;134:55–103. 10.1016/j.pneurobio.2015.09.004. [DOI] [PubMed] [Google Scholar]
  • 75.Jones RA, Harrison C, Eaton SL, et al. Cellular and molecular anatomy of the human neuromuscular junction. Cell Rep. 2017;21(9):2348–2356. 10.1016/j.celrep.2017.11.008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76.Chou VT, Johnson SA, Van Vactor D. Synapse development and maturation at the drosophila neuromuscular junction. Neural Dev. 2020;15(1):11. 10.1186/s13064-020-00147-5 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77.Wu H, Xiong WC, Mei L. To build a synapse: signaling pathways in neuromuscular junction assembly. Development. 2010;137(7):1017–1033. 10.1242/dev.038711. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78.Menon KP, Carrillo RA, Zinn K. Development and plasticity of the drosophila larval neuromuscular junction. Wiley Interdiscip Rev Dev Biol. 2013;2(5):647–670. 10.1002/wdev.108. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79.Markmiller S, Soltanieh S, Server KL, et al. Context-dependent and disease-specific diversity in protein interactions within stress granules. Cell. 2018;172(3):590–604 e13. 10.1016/j.cell.2017.12.032. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80.Yuva-Aydemir Y, Almeida S, Krishnan G, Gendron TF, Gao FB. Transcription elongation factor AFF2/FMR2 regulates expression of expanded GGGGCC repeat-containing C9ORF72 allele in ALS/FTD. Nat Commun. 2019;10(1):5466. 10.1038/s41467-019-13477-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81.Manzo E, Lorenzini I, Barrameda D, et al. Glycolysis upregulation is neuroprotective as a compensatory mechanism in ALS. Elife. 2019;8. 1–20. 10.7554/eLife.45114. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82.Castellanos-Montiel M, Chaineau M, Durcan TM. The neglected genes of ALS: cytoskeletal dynamics impact synaptic degeneration in ALS. Front Cell Neurosci. 2020;14:594975. 10.3389/fncel.2020.594975. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83.Leko MB, Župunski V, Kirincich J, et al. Molecular mechanisms of neurodegeneration related to C9orf72 hexanucleotide repeat expansion. Behav Neurol. 2019;2019:1–18. 10.1155/2019/2909168. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84.Casci I, Pandey UB. A fruitful endeavor: modeling ALS in the fruit fly. Brain Res. 2015; 1607:47–74. 10.1016/j.brainres.2014.09.064. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85.Sha SJ, Takada LT, Rankin KP, et al. Frontotemporal dementia due to C9ORF72 mutations clinical and imaging features. Neurology. 2012;79(10):1002–1011. 10.1212/WNL.0b013e318268452e. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86.Cooper-Knock J, Walsh MJ, Higginbottom A, et al. Sequestration of multiple RNA recognition motif-containing proteins by C9orf72 repeat expansions. Brain. 2014;137(Pt 7):2040–2051. 10.1093/brain/awu120. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87.Liu Y, Pattamatta A, Zu T, et al. C9orf72 BAC mouse model with motor deficits and neurodegenerative features of ALS/FTD. Neuron. 2016;90(3):521–534. 10.1016/j.neuron.2016.04.005. [DOI] [PubMed] [Google Scholar]
  • 88.Kaur SJ, McKeown SR, Rashid S. Mutant SOD1 mediated pathogenesis of amyotrophic lateral sclerosis. Gene. 2016;577 (2):109–118. 10.1016/j.gene.2015.11.049. [DOI] [PubMed] [Google Scholar]
  • 89.Ilieva H, Polymenidou M, Cleveland DW. Non-cell autonomous toxicity in neurodegenerative disorders: ALS and beyond. J Cell Biol. 2009;187(6):761–772. 10.1083/jcb.200908164. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90.de Araújo BA, de Carvalho MDC, Gerhardt E, et al. Characterization of the activity, aggregation, and toxicity of heterodimers of WT and ALS-associated mutant Sod1. Proc Natl Acad Sci U S A. 2019;116(51):25991–26000. 10.1073/pnas.1902483116. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 91.Tobisawa S, Hozumi Y, Arawaka S, et al. Mutant SOD1 linked to familial amyotrophic lateral sclerosis, but not wild-type SOD1, induces ER stress in COS7 cells and transgenic mice. Biochem Biophys Res Commun. 2003;303(2):496–503. 10.1016/S0006-291X(03)00353-X. [DOI] [PubMed] [Google Scholar]
  • 92.Gal J, Kuang L, Barnett KR, et al. ALS mutant SOD1 interacts with G3BP1 and affects stress granule dynamics. Acta Neuropathol. 2016;132(4):563–576. 10.1007/s00401-016-1601-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 93.Weisberg SJ, Lyakhovetsky R, Werdiger AC, Gitler AD, Soen Y, Kaganovich D. Compartmentalization of superoxide dismutase 1 (SOD1G93A) aggregates determines their toxicity. Proc Natl Acad Sci U S A. 2012;109(39):15811–15816. 10.1073/pnas.1205829109. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94.Baron DM, Kaushansky LJ, Ward CL, et al. Amyotrophic lateral sclerosis-linked FUS/TLS alters stress granule assembly and dynamics. Mol Neurodegener. 2013;8(1):1–18. 10.1186/1750-1326-8-30. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 95.Liu-Yesucevitz L, Bilgutay A, Zhang YJ, et al. Tar DNA binding protein-43 (TDP-43) associates with stress granules: analysis of cultured cells and pathological brain tissue. PLoS One. 2010;5(10):e13250. 10.1371/journal.pone.0013250. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 96.Mateju D, Franzmann TM, Patel A, et al. An aberrant phase transition of stress granules triggered by misfolded protein and prevented by chaperone function. EMBO J. 2017;36(12): 1669–1687. 10.15252/embj.201695957. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 97.Loganathan S, Lehmkuhl EM, Eck RJ, Zarnescu DC. To be or not to be…toxic—is RNA association with TDP-43 complexes deleterious or protective in neurodegeneration? Front Mol Bio-sci. 2020;6(January):1–11. 10.3389/fmolb.2019.00154. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 98.Pradat PF, Kabashi E, Desnuelle C. Deciphering spreading mechanisms in amyotrophic lateral sclerosis: clinical evidence and potential molecular processes. Curr Opin Neurol. 2015;28 (5):455–461. 10.1097/WCO.0000000000000239. [DOI] [PubMed] [Google Scholar]
  • 99.Sibilla C, Bertolotti A. Prion properties of SOD1 in amyotrophic lateral sclerosis and potential therapy. Cold Spring Harb Perspect Biol. 2017;9(10):a024141. 10.1101/cshperspect.a024141. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 100.Vassileff N, Cheng L, Hill AF. Extracellular vesicles – propagators of neuropathology and sources of potential biomarkers and therapeutics for neurodegenerative diseases. J Cell Sci. 2020;133(23):jcs243139. 10.1242/jcs.243139. [DOI] [PubMed] [Google Scholar]
  • 101.Kankel MW, Sen A, Lu L, et al. Amyotrophic lateral sclerosis modifiers in drosophila reveal the phospholipase d pathway as a potential therapeutic target. Gene. 2020;215(3):747–766. 10.1534/genetics.119.302985. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 102.Yanagi KS, Wu Z, Amaya J, et al. Meta-analysis of genetic modifiers reveals candidate Dysregulated pathways in amyotrophic lateral sclerosis. Neuroscience. 2019;396:A3–A20. 10.1016/j.neuroscience.2018.10.033. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 103.Dervishi I, Gozutok O, Murnan K, et al. Protein-protein interactions reveal key canonical pathways, upstream regulators, interactome domains, and novel targets in ALS. Sci Rep. 2018; 8(1):14732. 10.1038/s41598-018-32902-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 104.Suzuki N, Akiyama T, Warita H, Aoki M. Omics approach to axonal dysfunction of motor neurons in amyotrophic lateral sclerosis (ALS). Front Neurosci. 2020;14:194. 10.3389/fnins.2020.00194. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 105.Kori M, Aydin B, Unal S, Arga KY, Kazan D. Metabolic biomarkers and Neurodegeneration: a pathway enrichment analysis of Alzheimer’s disease, Parkinson’s disease, and amyotrophic lateral sclerosis. OMICS. 2016;20(11):645–661. 10.1089/omi.2016.0106. [DOI] [PubMed] [Google Scholar]
  • 106.Lanznaster D, Veyrat-Durebex C, Vourc’h P, Andres CR, Blasco H, Corcia P. Metabolomics: a tool to understand the impact of genetic mutations in amyotrophic lateral sclerosis. Genes. 2020;11(5):537. 10.3390/genes11050537. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 107.Blokhuis AM, Koppers M, Groen EJN, et al. Comparative interactomics analysis of different ALS-associated proteins identifies converging molecular pathways. Acta Neuropathol. 2016;132(2):175–196. 10.1007/s00401-016-1575-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 108.Liu A, Niswander LA. Bone morphogenetic protein signalling and vertebrate nervous system development. Nat Rev Neurosci. 2005;6:945–954. [DOI] [PubMed] [Google Scholar]
  • 109.Taetzsch T, Tenga MJ, Valdez G. Muscle fibers secrete FGFBP1 to slow degeneration of neuromuscular synapses during aging and progression of ALS. J Neurosci. 2017;37(1):70–82. 10.1523/JNEUROSCI.2992-16.2016. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 110.Akiyama T, Marqués G, Wharton KA. A large bioactive BMP ligand with distinct signaling properties is produced by alternative proconvertase processing. Sci Signal. 2012;5(218):ra28. 10.1126/scisignal.2002549. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 111.Shi Q, Zhong YS, Ren Z, et al. Analysis of the role of the BMP7-Smad4-Id2 signaling pathway in SW480 colorectal carcinoma cells. Mol Med Rep. 2011;4(4):627–631. 10.3892/mmr.2011.481. [DOI] [PubMed] [Google Scholar]
  • 112.Patin F, Baranek T, Vourc’h P, et al. Combined metabolomics and transcriptomics approaches to assess the IL-6 blockade as a therapeutic of ALS: deleterious alteration of lipid metabolism. Neurotherapeutics. 2016;13(4):905–917. 10.1007/s13311-016-0461-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 113.Zee M. v. d., Stockhammer O, Levetzow C. v., Fonseca R. N. d., Roth S. Sog/Chordin is required for ventral-to-dorsal Dpp/BMP transport and head formation in a short germ insect. Proc National Acad Sci.. 2006;103(44):16307–16312. 10.1073/pnas.0605154103. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 114.Babitt JL, Huang FW, Wrighting DM, et al. Bone morphogenetic protein signaling by hemojuvelin regulates hepcidin expression. Nat Genet. 2006;38(5):531–539. 10.1038/ng1777. [DOI] [PubMed] [Google Scholar]
  • 115.Xia Y, Yu PB, Sidis Y, et al. Repulsive guidance molecule RGMa alters utilization of bone morphogenetic protein (BMP) type II receptors by BMP2 and BMP4. J Biol Chem. 2007;282 (25):18129–18140. 10.1074/jbc.M701679200. [DOI] [PubMed] [Google Scholar]
  • 116.Luo JY, Zhang Y, Wang L, Huang Y. Regulators and effectors of bone morphogenetic protein signalling in the cardiovascular system. J Physiol. 2015;593(14):2995–3011. 10.1113/JP270207. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 117.Miyazawa K, Miyazono K. Regulation of TGF-β family signaling by inhibitory smads. Cold Spring Harb Perspect Biol. 2017; 9(3):1–26. 10.1101/cshperspect.a022095. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 118.Massagué J TGFbeta signalling in context. Nat Rev Mol Cell Biol. 2014;13(10):616–630. 10.1038/nrm3434.TGF. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 119.Coda DM, Gaarenstroom T, East P, et al. Distinct modes of SMAD2 chromatin binding and remodeling shape the transcriptional response to NODAL/Activin signaling. Elife. 2017; 6:1–31. 10.7554/eLife.22474. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 120.Yamada Y, Mashima H, Sakai T, Matsuhashi T, Jin M, Ohnishi H. Functional roles of TGF-β1 in intestinal epithelial cells through smad-dependent and non-smad pathways. Dig Dis Sci. 2013;58(5):1207–1217. 10.1007/s10620-012-2515-7. [DOI] [PubMed] [Google Scholar]
  • 121.Blahna MT, Hata A. Smad-mediated regulation of microRNA biosynthesis. FEBS Lett. 2012;586(14):1906–1912. 10.1016/j.febslet.2012.01.041. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 122.Zhang Y, O’Keefe RJ, Jonason JH. BMP-TAK1 (MAP3K7) induces adipocyte differentiation through PPARgamma signaling. J Cell Biochem. 2017;118(1):204–210. 10.1002/jcb.25626. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 123.BD MC, Marqués G, Haghighi AP, et al. The BMP homolog Gbb provides a retrograde signal that regulates synaptic growth at the drosophila neuromuscular junction. [comparative study]. Neuron. 2003;39(2):241–254. [DOI] [PubMed] [Google Scholar]
  • 124.Endo F, Komine O, Fujimori-Tonou N, et al. Astrocyte-derived TGF-β1 accelerates disease progression in ALS mice by interfering with the Neuroprotective functions of microglia and T cells. Cell Rep. 2015;11(4):592–604. 10.1016/j.celrep.2015.03.053. [DOI] [PubMed] [Google Scholar]
  • 125.Nakamura M, Ito H, Wate R, Nakano S, Hirano A, Kusaka H. Phosphorylated Smad2/3 immunoreactivity in sporadic and familial amyotrophic lateral sclerosis and its mouse model. Acta Neuropathol. 2008;115(3):327–334. 10.1007/s00401-007-0337-z. [DOI] [PubMed] [Google Scholar]
  • 126.Si Y, Kim S, Cui X, et al. Transforming growth factor Beta (TGF-beta) is a muscle biomarker of disease progression in ALS and correlates with Smad expression. PLoS One. 2015;10 (9):e0138425. 10.1371/journal.pone.0138425. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 127.Nakamura M, Kaneko S, Ito H, et al. Activation of transforming growth factor-beta/Smad signaling reduces aggregate formation of mislocalized TAR DNA-binding protein-43. Neurodegener Dis. 2013;11(4):182–193. 10.1159/000338151. [DOI] [PubMed] [Google Scholar]
  • 128.Hazelett DJ, Chang J-C, Lakeland DL, Morton DB. Comparison of parallel high-throughput RNA sequencing between knockout of TDP-43 and its overexpression reveals primarily nonreciprocal and nonoverlapping gene expression changes in the central nervous system of Drosophila. G3 (Bethesda, Md). 2012;2(7):789–802. 10.1534/g3.112.002998. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 129.Shijo T, Warita H, Suzuki N, et al. Antagonizing bone morphogenetic protein 4 attenuates disease progression in a rat model of amyotrophic lateral sclerosis. Exp Neurol. 2018;307: 164–179. 10.1016/j.expneurol.2018.06.009. [DOI] [PubMed] [Google Scholar]
  • 130.Marques G, Haerry TE, Crotty ML, Xue M, Zhang B, O’Connor MB. Retrograde Gbb signaling through the bmp type 2 receptor wishful thinking regulates systemic FMRFa expression in drosophila. Development. 2003;130(22):5457–5470. 10.1242/dev.00772. [DOI] [PubMed] [Google Scholar]
  • 131.Goold CP, Davis GW. The BMP ligand Gbb gates the expression of synaptic homeostasis independent of synaptic growth control. Neuron. 2007;56(1):109–123. 10.1016/j.neuron.2007.08.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 132.Hoover KM, Gratz SJ, Qi N, et al. The calcium channel subunit alpha2delta-3 organizes synapses via an activity-dependent and autocrine BMP signaling pathway. Nat Commun. 2019;10(1): 5575. 10.1038/s41467-019-13165-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 133.Berke B, Wittnam J, McNeill E, Van Vactor DL, Keshishian H. Retrograde BMP signaling at the synapse: a permissive signal for synapse maturation and activity-dependent plasticity. J Neurosci. 2013;33(45):17937–17950. 10.1523/JNEUROSCI.6075-11.2013. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 134.Theys PA, Peeters E, Robberecht W. Evolution of motor and sensory deficits in amyotrophic lateral sclerosis estimated by neurophysiological techniques. J Neurol. 1999;246(6):438–442. 10.1007/s004150050379. [DOI] [PubMed] [Google Scholar]
  • 135.Pugdahl K, Fuglsang-Frederiksen A, de Carvalho M, et al. Generalised sensory system abnormalities in amyotrophic lateral sclerosis: a European multicentre study. J Neurol Neurosurg Psychiatry. 2007;78(7):746–749. 10.1136/jnnp.2006.098533. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 136.Isaacs JD, Dean AF, Shaw CE, Al-Chalabi A, Mills KR, Leigh PN. Amyotrophic lateral sclerosis with sensory neuropathy: part of a multisystem disorder? J Neurol Neurosurg Psychiatry. 2007;78(7):750–753. 10.1136/jnnp.2006.098798. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 137.Vaughan SK, Sutherland NM, Zhang S, Hatzipetros T, Vieira F, Valdez G. The ALS-inducing factors, TDP43(A315T) and SOD1(G93A), directly affect and sensitize sensory neurons to stress. Sci Rep. 2018;8(1):16582. 10.1038/s41598-018-34510-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 138.Deal SL, Yamamoto S. Unraveling novel mechanisms of Neurodegeneration through a large-scale forward genetic screen in drosophila. Front Genet. 2018;9:700. 10.3389/fgene.2018.00700. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 139.Link N, Bellen HJ. Using drosophila to drive the diagnosis and understand the mechanisms of rare human diseases. Development. 2020;147(21):dev191411. 10.1242/dev.191411. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 140.Higashi-Kovtun ME, Mosca TJ, Dickman DK, Meinertzhagen IA, Schwarz TL. Importin-beta11 regulates synaptic phosphorylated mothers against decapentaplegic, and thereby influences synaptic development and function at the drosophila neuromuscular junction. J Neurosci. 2010;30 (15):5253–5268. 10.1523/JNEUROSCI.3739-09.2010. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 141.Sulkowski M, Kim Y-J, Serpe M. Postsynaptic glutamate receptors regulate local BMP signaling at the drosophila neuromuscular junction. Development (Cambridge, England). 2014;141(2):436–447. 10.1242/dev.097758. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 142.Berke B, Le L, Keshishian H. Target-dependent retrograde signaling mediates synaptic plasticity at the drosophila neuromuscular junction. Dev Neurobiol. 2019;79(11–12):895–912. 10.1002/dneu.22731. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 143.Ratnaparkhi A, Lawless GM, Schweizer FE, Golshani P, Jackson GR. A drosophila model of ALS: human ALS-associated mutation in VAP33A suggests a dominant negative mechanism. PLoS One. 2008;3(6):e2334. 10.1371/journal.pone.0002334. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 144.Deshpande M, Feiger Z, Shilton AK, Luo CC, Silverman E, Rodal AA. Role of BMP receptor traffic in synaptic growth defects in an ALS model. Mol Biol Cell. 2016;27(19):2898–2910. 10.1091/mbc.E16-07-0519. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 145.Berke B, Wittnam J, McNeill E, Van Vactor DL, Keshishian H. Retrograde BMP signaling at the synapse: a permissive signal for synapse maturation and activity-dependent plasticity. J Neurosci. 2013;33(45):17937–17950. 10.1523/jneurosci.6075-11.2013. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 146.Bartoletti M, Knight T, Held A, Rand LM, Wharton KA. Glianeuron signaling mediated by two different BMP ligands impacts synaptic growth. bioRxiV. 2021; submitted. 10.1101/2021.02.23.432606. [DOI] [Google Scholar]
  • 147.Kim HS, Neugebauer J, McKnite A, Tilak A, Christian JL. BMP7 functions predominantly as a heterodimer with BMP2 or BMP4 during mammalian embryogenesis. eLife. 2019;8. 10.7554/elife.48872. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 148.Gipson GR, Goebel EJ, Hart KN, et al. Structural perspective of BMP ligands and signaling. Bone. 2020;140:115549. 10.1016/j.bone.2020.115549. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 149.Hinck AP, Mueller TD, Springer TA. Structural biology and evolution of the TGF-β family. [review]. Cold Spring Harbor Perspect Biol. 2016;8(12):a022103. 10.1101/cshperspect.a022103. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 150.Anderson EN, Wharton KA. Alternative cleavage of the bone morphogenetic protein (BMP), Gbb, produces ligands with distinct developmental functions and receptor preferences. J Biol Chem. 2017;292(47):19160–19178. 10.1074/jbc.M117.793513. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 151.Fuentes-Medel Y, Ashley J, Barria R, Maloney R, Freeman M, Budnik V. Integration of a retrograde signal during synapse formation by glia-secreted TGF-beta ligand. Curr Biol. 2012;22 (19):1831–1838. 10.1016/j.cub.2012.07.063. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 152.Morooka T, Nishida E. Requirement of p38 mitogen-activated protein kinase for neuronal differentiation in PC12 cells. J Biol Chem. 1998;273(38):24285–24288. 10.1074/jbc.273.38.24285. [DOI] [PubMed] [Google Scholar]
  • 153.Liu X, Zhao Y, Peng S, et al. BMP7 retards peripheral myelination by activating p38 MAPK in Schwann cells. Sci Rep. 2016;6(1):31049. 10.1038/srep31049. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 154.Ackerley S, Grierson AJ, Banner S, et al. P38A stress-activated protein kinase phosphorylates neurofilaments and is associated with neurofilament pathology in amyotrophic lateral sclerosis. Mol Cell Neurosci. 2004;26(2):354–364. 10.1016/j.mcn.2004.02.009. [DOI] [PubMed] [Google Scholar]
  • 155.Bendotti C, Carrì MT. Lessons from models of SOD1-linked familial ALS. Trends Mol Med. 2004;10(8):393–400. 10.1016/j.molmed.2004.06.009. [DOI] [PubMed] [Google Scholar]
  • 156.Tortarolo M, Veglianese P, Calvaresi N, et al. Persistent activation of p38 mitogen-activated protein kinase in a mouse model of familial amyotrophic lateral sclerosis correlates with disease progression. Mol Cell Neurosci. 2003;23(2):180–192. 10.1016/S1044-7431(03)00022-8. [DOI] [PubMed] [Google Scholar]
  • 157.Gibbs KL, Kalmar B, Rhymes ER, et al. Inhibiting p38 MAPK alpha rescues axonal retrograde transport defects in a mouse model of ALS article. Cell Death Dis. 2018;9(6):1–16. 10.1038/s41419-018-0624-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 158.Cavalli V, Vilbois F, Corti M, et al. The stress-induced MAP kinase p38 regulates endocytic trafficking via the GDI:Rab5 complex. Mol Cell. 2001;7(2):421–432. 10.1016/S1097-2765(01)00189-7. [DOI] [PubMed] [Google Scholar]
  • 159.Takeda K, Ichiijo H. Neuronal p38 MAPK signalling: an emerging regulator of cell fate and function in the nervous system. Genes Cells. 2002;7(11):1099–1111. 10.1046/j.1365-2443.2002.00591.x. [DOI] [PubMed] [Google Scholar]
  • 160.Lee-Hoeflich ST, Causing CG, Podkowa M, Zhao X, Wrana JL, Attisano L. Activation of LIMK1 by binding to the BMP receptor, BMPRII, regulates BMP-dependent dendritogenesis. EMBO J. 2004;23(24):4792–4801. 10.1038/sj.emboj.7600418. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 161.Wu CH, Fallini C, Ticozzi N, et al. Mutations in the profiling 1 gene cause familial amyotrophic lateral sclerosis. Nature. 2012;488(7412):499–503. 10.1038/nature11280. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 162.Yang C, Danielson EW, Qiao T, et al. Mutant PFN1 causes ALS phenotypes and progressive motor neuron degeneration in mice by a gain of toxicity. Proc Natl Acad Sci U S A. 2016;113(41): E6209–E6218. 10.1073/pnas.1605964113. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 163.Kiaei M, Balasubramaniam M, Govind Kumar V, Shmookler Reis RJ, Moradi M, Varughese KI. ALS-causing mutations in profilin-1 alter its conformational dynamics: a computational approach to explain propensity for aggregation. Sci Rep. 2018; 8(1):1–10. 10.1038/s41598-018-31199-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 164.Sivadasan R, Hornburg D, Drepper C, et al. C9ORF72 Interaction with cofilin modulates Actin dynamics in motor neurons. Nat Neurosci. 2016;19(12):1610–1618. 10.1038/nn.4407. [DOI] [PubMed] [Google Scholar]
  • 165.Eaton BA, Davis GW. LIM Kinase1 controls synaptic stability downstream of the type II BMP receptor. Neuron. 2005;47(5): 695–708. 10.1016/j.neuron.2005.08.010. [DOI] [PubMed] [Google Scholar]
  • 166.Ng J TGFβ signals regulate axonal development through distinct Smad-independent mechanisms. Development. 2008;135 (24):4025–4035. 10.1242/dev.028209. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 167.Gertz B, Wong M, Martin LJ. Nuclear localization of human SOD1 and mutant SOD1-specific disruption of survival motor neuron protein complex in transgenic amyotrophic lateral sclerosis mice. J Neuropathol Exp Neurol. 2012;71(2):162–177. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 168.Tang X, Toro A, Sahana TG, et al. Divergence, convergence, and therapeutic implications: a cell biology perspective of C9ORF72-ALS/FTD. Mol Neurodegener. 2020;15(1):1–15. 10.1186/s13024-020-00383-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 169.Fallini C, Khalil B, Smith CL, Rossoll W. Traffic jam at the nuclear pore: all roads lead to nucleocytoplasmic transport defects in ALS/FTD. Neurobiol Dis. 2020;140(January):104835. 10.1016/j.nbd.2020.104835. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 170.Hutten S, Usluer S, Bourgeois B, et al. Nuclear import receptors directly bind to arginine-rich dipeptide repeat proteins and suppress their pathological interactions. Cell Rep. 2020;33 (12):108538. 10.1016/j.celrep.2020.108538. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 171.Hutten S, Flotho A, Melchior F, Kehlenbach RH. The Nup358-RanGAP complex is required for efficient importin alpha/beta-dependent nuclear import. Mol Biol Cell. 2008;19 (5):2300–2310. 10.1091/mbc.E07-12-1279. [DOI] [PMC free article] [PubMed] [Google Scholar]

RESOURCES