Skip to main content

Unraveling neuronal and metabolic alterations in neurofibromatosis type 1

Abstract

Neurofibromatosis type 1 (OMIM 162200) affects ~ 1 in 3,000 individuals worldwide and is one of the most common monogenetic neurogenetic disorders that impacts brain function. The disorder affects various organ systems, including the central nervous system, resulting in a spectrum of clinical manifestations. Significant progress has been made in understanding the disorder’s pathophysiology, yet gaps persist in understanding how the complex signaling and systemic interactions affect the disorder. Two features of the disorder are alterations in neuronal function and metabolism, and emerging evidence suggests a potential relationship between them. This review summarizes neurofibromatosis type 1 features and recent research findings on disease mechanisms, with an emphasis on neuronal and metabolic features.

Introduction

Neurofibromatosis type 1 (NF1) is a multisystemic autosomal-dominant condition affecting ~ 1 in 3,000 live births worldwide [1,2,3,4]. Historical recognition of the disorder dates to ancient Egypt [5, 6], with Friedrich Daniel von Recklinghausen providing the first comprehensive clinical description in 1882. Initially termed “von Recklinghausen disease,” it is now known as neurofibromatosis type 1 [7, 8]. Significant progress has been made in understanding NF1’s genetic underpinnings. The disorder is caused by mutations in a single gene, neurofibromin 1 (NF1) [9], which encodes the protein neurofibromin (Nf1). Several pivotal discoveries made in the late 1990s include the establishment of diagnostic criteria for accurate NF1 diagnosis [10, 11], mapping of the NF1 gene to chromosome 17q11.2 [12,13,14], and the identification of its protein product [12, 15,16,17,18,19]. Despite these advances (and more since then), gaps persist in our understanding of the disorder and its underlying genetic, cellular, and systemic mechanisms. Features of the disorder include alterations in neuronal and brain function as well as metabolic alterations [20,21,22,23,24]. Some of the features of the disorder, including the brain/cognitive symptoms, could be influenced by the metabolic alterations. Here we review the mechanisms of NF1 pathophysiology, with a focus on the emerging understanding of neuronal and metabolic alterations.

Diagnostic criteria and clinical features

NF1 is characterized by a broad spectrum of clinical manifestations that begin in infancy and progressively worsen (Fig. 1) [25, 26]. Diagnostic criteria for NF1, first established in 1987 [10] and updated in 2021 [11] (Table 1), rely on a physical examination and family history review. Some symptoms emerge in an age-dependent manner, making proper diagnosis during early childhood challenging, particularly for those lacking a family history of the disease (half of NF1 cases stem from de novo mutations) [3, 11, 20]. These challenges led to revisions of the diagnostic criteria, incorporating mosaic neurofibromatosis and genetic testing [11]. Symptoms of NF1 include increased susceptibility to various tumors, including peripheral nerve tumors like neurofibromas, plexiform neurofibromas, and malignant peripheral nerve sheath tumors, as well as brain tumors such as optic pathway gliomas and brainstem gliomas [1, 25, 27, 28]. Although tumors are a primary clinical characteristic of NF1, it also produces non-tumor symptoms including pigmentation defects, skeletal abnormalities, stunted growth, cognitive impairments, and behavioral alterations [27, 29]. NF1 reduces life expectancy by 8–15 years [30,31,32] and significantly impacts quality of life, with up to 80% of children experiencing moderate to severe cognitive impairments [22, 29].

Fig. 1
figure 1

Disease progression and clinical features of NF1. The onset and severity of NF1 clinical features vary between individuals. In children, the most common clinical physical manifestations are skeletal abnormalities such as scoliosis, tibial dysplasia, and café-au-lait spots. Young children are at risk of developing juvenile myelomonocytic leukemia, optic gliomas, and behavioral and cognitive deficits, with attention-deficit/hyperactivity disorder (ADHD) and autism spectrum disorder (ASD) being the most common. The risk of developing plexiform neurofibromas (pNF) is high during the early stages of life, but other malignancies such as malignant peripheral nerve sheath tumors (MPNST) and breast cancer occur more often after the third decade of life [25, 26]. Created with BioRender.com

Table 1 Diagnostic criteria for NF1

The NF1 gene and neurofibromin protein

NF1 results from mutations in the NF1 gene, which encodes a large 2,818 amino acid protein called neurofibromin (Nf1) [17, 33]. The Nf1 protein contains a central Ras-GTPase activating protein (GAP)-related domain (GRD) [16, 34]. It primarily localizes to the cytoplasm, interacting with Ras at the plasma membrane, and is also found in the nucleus, endoplasmic reticulum, and mitochondria [35,36,37,38]. Nf1 is ubiquitously expressed throughout development, with the highest levels in nervous system cells, including Schwann cells, neurons, astrocytes, and oligodendrocytes [17, 19, 39]. Clinical manifestations of NF1 are variable, even among identical germline NF1 mutations, and some exhibit segmental or mosaic NF1.

Role of the NF1 gene and neurofibromin GAP-related domain

Over 2,600 unique mutations within the NF1 gene have been identified [40] (Fig. 2). Clinical heterogeneity of NF1 can be attributed to multiple factors, including allelic variation, second-hit mutations, epigenetic changes, differences across the NF1 mutations, and tissue-specific Nf1 functions [41]. Nf1 protein expression across between different mutations (i.e., Nf1 heterozygosity can result in ~ 12–89% of normal protein expression level) [42]. Neurofibromas result from loss of NF1 heterozygosity following second-hit mutations [43]. Other symptoms of the disorder emanate from haploinsufficiency due to the heterozygous mutation itself. Heterozygous germline mutation in NF1 is associated with notable impacts on cognitive functions, affecting attention and learning and increasing the prevalence of autism spectrum disorder (ASD) [44, 45]. Among the NF1 isoforms, certain variants have a tissue-specific role. The alternatively-spliced exon 11alt12 (formerly known exon 9a) is predominantly expressed in the central nervous system (CNS), particularly within forebrain neurons [46]. In contrast, the alternatively spliced exon 30alt31 (formerly known as exon 23a) contains an alternative exon that lies within the GRD and diminishes Ras GAP activity [47]; mice lacking this exon have learning and communication impairments but are not susceptible to tumor formation [48,49,50]. Alternative splicing at the 3’ end of NF1 produces the alternative exon 56alt57 (formerly 48a) which is expressed highly in fetal and adult cardiac and skeletal tissue and may contribute to reduced muscle strength and muscle weight [47, 51,52,53,54]. Exon 12alt13 (formerly known as 10a-2) is a low-level ubiquitous isoform concentrated in perinuclear granular structures [55]. Moreover, the NF1-∆E43 isoform shows elevated expression in the liver, kidneys, lungs, placenta, and skeletal muscle relative to the general expression of NF1 [56, 57]. Dimerization of the Nf1 protein and the varied impacts of different mutations on protein stability further complicate the disease’s pathophysiology [58].

Fig. 2
figure 2

Nf1 protein structure, interacting domains, and genotype–phenotype correlations. Nf1 protein contains several domains (squares) and interacting proteins (ovals). Nf1 protein domains include the following: cysteine-serine-rich domain (CSRD), tubulin binding domain (TBD), central GTPase-activating-protein-related domain (GRD), SEC14 domain, leucine-rich domain (LRD), pleckstrin homolog (PH), HEAT-like repeats (HLR), C-terminal domain (CTD), syndecan-binding domain (SBD). Phospholipids and proteins identified as Nf1-interacting proteins are shown in association with their described function, such as: trafficking (green), neuronal (yellow), membrane localization (purple), cell adhesion (gray), and cell signaling (blue). Nf1 mutations reported to correlate with certain phenotypes are shown above/below the protein and associated phenotypes. Numbers along protein indicate amino acid residues. Figure adapted and modified from Ratner and Miller (2015) [26], Mo et al. (2022) [59], and Anastasaki et al. (2022) [60]. Created with BioRender.com

Neurofibromin’s impact on cellular processes via Ras and cAMP signaling

The Nf1 protein features multiple structural domains, with the GRD being the most extensively studied [16, 59,60,61,62]. The GRD plays a pivotal role in the regulation of Ras signaling, catalyzing hydrolysis of Ras-bound GTP into GDP and thereby attenuating Ras signaling (Fig. 3). Consequently, NF1 loss-of-function mutations lead to the accumulation of active Ras-GTP and aberrant activation of downstream pathways, including Raf/MEK/ERK and PI3K/AKT/mTOR [45, 61, 63, 64]. Numerous mutations that compromise the function of the GRD have been identified in patients (Fig. 3) [61]. Analysis of the crystallographic structure of the Nf1 GRD revealed a critical arginine finger residue (R1276) that stabilizes and positions Ras association with the catalytic domain. Notably, a patient mutation (R1276P), which substitutes arginine with proline, results in a > 1000-fold reduction in Ras-GAP activity [61, 62, 65].

Fig. 3
figure 3

Nf1 regulates molecular functions in key biological signaling pathways via Ras. Nf1 affects diverse cellular functions by regulating several signaling pathways. Nf1 accelerates the conversion of active GTP-bound Ras into inactive GFP-bound Ras, thereby regulating numerous downstream effectors. Known signaling pathways downstream of Nf1 and Ras signaling include the Raf/MEK/ERK and PI3K/AKT/mTOR pathways. Figure created with BioRender.com and adapted from Anastasaki et al. (2022) [60], Masgras and Rasola (2021) [66], and Ratner and Miller (2015) [26]

Nf1 regulates multiple cellular processes, including metabolism, cell proliferation, differentiation, and survival via its regulatory effects on Ras signaling. Nf1/Ras activity is regulated by upstream signal transduction pathways involving receptor tyrosine kinases (RTK). One such RTK is the Anaplastic Lymphoma Kinase (ALK), which interacts with Nf1 and functions as an upstream activator of Nf1-regulated Ras signaling pathway [67,68,69,70]. In addition to RTKs, other upstream regulators of Nf1/Ras include G protein-coupled receptors (GPCRs), specifically the Gβγ subunits that bind to Nf1 in striatal neurons and inhibits its capacity to suppress Ras/AKT/mTOR signaling [71].

Loss-of-function mutations in Nf1 dysregulate multiple signaling pathways downstream of Ras, including the canonical mitogen-activated protein kinase (MAPK) signaling pathway (Raf/MEK/ERK), PI3K/AKT/mTOR, and others (Fig. 3) [18, 63, 64]. These pathways in turn regulate multiple cellular and metabolic processes, including cell growth, survival, nutrient uptake, proliferation, and the modulation of neuronal metabolism in response to growth factors, nutrients, and changes in the cellular energy state [72]. Hyperactivation of Raf/MEK/ERK due to loss of Nf1 is one of the major mechanisms implicated in NF1 phenotypes and is a current therapeutic target [73,74,75]. Conditional Nf1 knockout in various brain cells (astrocytes, pyramidal cells, GABAergic neurons, and inhibitory/excitatory neurons) increases ERK signaling [37]. In addition, hyperactivation of the PI3K/AKT/mTOR pathway contributes to the development of some NF1-associated phenotypes. The convergence of these two Ras effector pathways, each of which will be discussed in greater detail below, underscores the complexity of cellular signaling alterations in NF1. Overall, the NF1 gene and the Nf1 GRD function as a central regulator of Ras signaling, modulating downstream signaling targets and affecting diverse cellular functions.

In addition to its role in regulating Ras signaling, Nf1 is a positive regulator of cyclic adenosine monophosphate (cAMP) and downstream protein kinase A (PKA) activity (Fig. 3). Nf1 is required for normal cAMP generation in neurons and astrocytes, as observed in Drosophila and rodent models of NF1 [76, 77]. In turn, alterations in cAMP/PKA signaling are implicated in many of the NF1 phenotypes, including cell differentiation/growth and learning [68, 78,79,80,81]. Furthermore, NF1-related changes in cAMP/PKA levels are altered via a non-canonical mechanism involving Ras-dependent phosphorylation of protein kinase C zeta (PKCζ) [82, 83]. This pathway regulates neuronal cAMP homeostasis in both human-induced pluripotent stem cell (hiPSC)-derived neurons and primary mouse neuron cultures [82].

Mechanisms of pathophysiology in neurofibromatosis type 1

Tumors

Tumor formation is a primary concern in NF1, with cutaneous neurofibromas (CN) and plexiform neurofibromas (pNF) pervasive among patients. CNs, affecting over 99% of NF1-afflicted adults, are benign but prolific tumors that emerge during late childhood and experience rapid grow during puberty and pregnancy [4]. The number of CNs in adults can reach into the thousands, leading to significant disfigurement and considerable physical and psychological distress [84, 85]. Conversely, pNFs affect about 50% of patients, proliferating rapidly during childhood and adolescence [86]. These tumors are located in peripheral nerve sheaths, with Schwann cells representing the major neoplastic cell type [87]. Neurofibromas can cause pain, disfigurement, and impair neurovascular structures and airways. Notably, pNFs are major contributors to the elevated mortality rates in NF1, as they can transform into malignant peripheral nerve sheath tumors (MPNST), which have a low survival rate [31, 88, 89]. Although surgical intervention is the standard treatment for pNF, it is typically palliative [88]. In 2020, selumetinib, a MEK inhibitor, gained United States Food and Drug Administration (FDA) approval for treating symptomatic and inoperable pNF in children [73,74,75].

In addition to neurofibromas, optic pathway gliomas (OPGs) are the second most common tumors in NF1—approximately 15–20% of children with NF1 develop OPGs [90]. Although OPGs are often non-lethal, about 30% of affected individuals will experience visual decline or loss due to these tumors, significantly reducing their quality of life. Given the severe delayed toxicity of radiotherapy and increased risk of visual loss with surgery, chemotherapy is the first line of treatment for OPGs that cause visual decline. Notably, females are three times more likely to require treatment [90,91,92,93,94,95].

Experimental animal models of NF1 and in vitro cellular studies have provided significant insights into NF1-related tumor development. These models show significant alterations in growth, cell proliferation, and tumor progression. Such phenotypes stem from the interplay between multiple tissues, signaling pathways, neurite growth, and neuronal excitability. Introducing patient-derived NF1 mutations into hiPSCs impairs Schwann cell differentiation, promotes stemness, and fosters neurofibroma formation [96]. Furthermore, studies using hiPSCs and murine models have revealed that Nf1 mutations increase neuronal excitability, exacerbating tumor progression in both the central and peripheral nervous system [97,98,99]. Neuronal activity and midkine expression directly impact the development and progression of mouse Nf1-OPG [97, 98]. Importantly, the progression of optic glioma growth in Nf1 mutant mice can be selectively suppressed with clinically relevant dosing of lamotrigine, an anti-epileptic drug, for months after treatment cessation [100].

Aberrant regulation of the Raf/MEK/ERK signaling pathway plays a pivotal role of NF1-related tumorigenesis [26, 101], and MEK is a therapeutic target [73,74,75, 102, 103]. Both human and mouse models of MPNST exhibit aberrant activation of ERK (one molecular step downstream of MEK). Targeted pharmacological inhibition of the Raf/MEK/ERK pathway has been shown to inhibit tumor progression [101]. Proteomic analysis has also revealed Ras/PI3K-dependent activation of mTOR signaling in astrocytes from human or mutant mice optic nerve gliomas [63]. In NF1-deficient cells and human tumors, mTOR is constitutively activated [63, 64]. Notably, pharmacological inhibition of mTOR, MEK, and AKT signaling can restore normal proliferation in Nf1-deficient astrocytes [63, 104]. Additional pathways, such as cAMP/PKA, are also targets of interest for therapeutic interventions, playing roles in regulating cell differentiation and growth arrest [78]. In Drosophila, Nf1 regulates growth through non-cell-autonomous control of cAMP/PKA signaling in neuroendocrine cells [68]. In human neural progenitor cells, loss of Nf1 decreases cAMP levels, resulting in smaller growth cone areas and shorter axonal lengths [82]. These neural deficits can be restored through increased cAMP levels and by inhibiting Ras activity [82].

The zebrafish model, known for its transparent embryos, offers a unique lens through which to study the role of Nf1 during development. The zebrafish genome contains two NF1 orthologs, nf1a and nf1b, each with over 90% similarity to human NF1 at the amino acid level [105]. Experiments involving transient knockdown of these nf1 orthologs during embryogenesis result in vascular patterning defects, echoing observations seen in murine NF1 models and mirroring hallmarks of the human disease [105]. Furthermore, nf1a and nf1b zebrafish larvae exhibit hyperplasia of oligodendrocyte progenitor and Schwann cells [106, 107]. Additionally, nf1 knockout initiates gliomagenesis in adult zebrafish brain tissue [108].

NF1 mutations introduced into Yucatan miniature pigs (minipigs) mimic characteristics commonly observed in NF1 patients. Two mutations have been introduced, which model prevalent human NF1 mutations: NF1R1947X, representing a common nonsense mutation, and NF1+/ex42del, emulating a heterozygous NF1 mutation [109, 110]. Minipigs with either of these NF1 mutations exhibit major clinical hallmarks of NF1, including café-au-lait macules (CALMs), OPGs, and neurofibromas [109, 110]. Notably, the minipig is unique among model organisms in that it exhibits spontaneous loss of NF1 heterozygosity, which drives tumor formation in humans [109].

Behavioral deficits and neuronal alterations

Cognitive impairment is a prevalent complication of NF1, affecting approximately 80% of those diagnosed with NF1 [22, 111]. Individuals with NF1 are significantly more likely to encounter a spectrum of developmental delays, such as deficits in learning, memory, executive function, broad language deficits, and fine motor skills [22, 29, 112]. NF1 patients may exhibit below-average IQ scores, with a small subset (4–8%) falling into the intellectually impaired range [22, 113]. The disorder is also highly comorbid with attention-deficit/hyperactivity disorder (ADHD) and ASD. Approximately half of the children with NF1 are diagnosed with ADHD [22, 111], and 12–49% exhibit symptoms of ASD [114,115,116,117,118]. These cognitive and behavioral challenges significantly impact quality of life of NF1 patients, affecting their emotional well-being, physical health, role functioning, and social interactions [25].

Given that NF1 increases risk for cognitive/behavioral symptoms, a major question is how loss of neurofibromin affects neuronal/brain function. Studies utilizing various animal models, including flies, zebrafish, mice, and minipigs, have contributed to understanding the role of Nf1 function in the nervous system. The Drosophila Nf1 protein, sharing 60% amino acid sequence homology with its human counterpart and conserved Ras GAP functionality [80], serves as an outstanding model for investigating genetics, neuronal function, and molecular signaling pathways in vivo [119,120,121]. In Drosophila, Nf1 is ubiquitously expressed during development and is prominently localized in the adult nervous system [39]. Loss of Nf1 function in flies disrupts sleep and circadian rhythms [122,123,124]; the circadian rhythm deficit can be rescued by restoring the expression of wild-type Drosophila Nf1 in neurons or by attenuating Ras/ERK signaling pathways [122]. Additionally, Drosophila Nf1 mutants exhibit learning and memory deficits, including impaired olfactory associative learning and deficits in short-, middle-, and long-term memory [67, 79, 125, 126]. These learning and memory impairments can be rescued by restoring wild-type Nf1 protein in a neuron-specific manner [125] and ameliorated by enhancing PKA activity [79, 80]. Additionally, pharmacologically and genetically attenuating ALK, an upstream RTK, rescues associative learning deficits in nf1 mutants [67].

Mutations in the Drosophila NF1 ortholog increase locomotor activity and spontaneous grooming [127, 128], phenotypic analogs of the ADHD symptoms common in NF1 patients [29, 111]. Nf1/Ras signaling regulates grooming behavior, as the Nf1 GRD is required in neurons to maintain normal levels of grooming in Drosophila [127]. Besides motor-related behaviors, Drosophila nf1 mutants display social and behavioral alterations, including delayed flight and climbing responses and altered sleep patterns [80, 122, 129]. Loss of Nf1 alters social behavior, specifically male courtship [130]. Synaptic transmission at the neuromuscular junction is altered in nf1 mutants, suggesting that synaptic physiology changes may contribute to the phenotypes [131, 132].

Murine models of NF1 have been invaluable in unraveling Nf1's functions within the nervous system via structural plasticity and modulation of signaling pathways. In the rat hippocampus, the loss of Nf1 function disrupts pyramidal dendritic spine structural plasticity, resulting in the activity-dependent loss of dendritic spines due to sustained Ras activation [133]. In mice, Nf1 haploinsufficiency (Nf1±) replicate cognitive and behavioral deficits observed in NF1 patients, manifesting as deficits in hippocampal spatial learning and reduced long-term potentiation driven by increased GABA-mediated inhibition [44, 45, 48]. Rescue of Raf/MEK/ERK activity, either pharmacologically or genetically, ameliorates learning deficits and rescues long-term potentiation [44, 134, 135]. Moreover, Nf1± mice show heightened excitability in sensory neurons. Along with dopamine deficiency, this could contribute to learning impairment [99, 136,137,138]. Additionally, both human NF1 and mouse Nf1 are enriched in inhibitory neurons within the cortex [139]. Nf1 plays a crucial role in the nervous system beyond cognition and physiology, as human-derived Nf1 mutations increase neuronal excitability in mice, accelerating tumor progression in the central and peripheral nervous system [97, 98].

Several other vertebrate models like zebrafish and minipigs have recapitulated neurocognitive deficits similar to those observed in NF1 patients. Zebrafish with nf1 mutations display learning and memory deficits, including short- and long-term habituation; these can be restored either through pharmacological inhibition of Ras downstream targets or by increasing cAMP signaling [81, 107]. The NF1+/ex42del mutation in the minipig model produces neurocognitive deficits akin to those observed in NF1 patients, including learning and memory impairments and hyperactivity [110]. In addition, the NF1 minipig model exhibits altered pain sensitivity associated with NF1—examination of dorsal root ganglia expressing mutant NF1+/ex42del revealed dysregulation of calcium and sodium channels [110]. Overall, these findings underscore the importance of Nf1 function in regulating neuronal development, structure, activity, and function.

Metabolic alterations

Metabolism is altered in multiple ways in NF1, and these changes may contribute to the pathophysiology of the disease. Patients with NF1 exhibit systemic metabolic shifts (Fig. 4) [32, 140]. Studies on body composition reveal multiple anomalies, including a lower body mass index (BMI) [141], reduced triglyceride stores [142], decreased bone mineral density [143], and shorter stature relative to unaffected individuals [144]. NF1 patients display lower muscle function [141], reduced maximal muscular strength [24, 145], and compromised motor proficiency [146]. In a comprehensive analysis of resting energy expenditure (REE), women with NF1 display heightened REE despite lower BMI [24]. Additionally, NF1 patients have a lower respiratory quotient (RQ), which indicates a differential reliance on fat oxidation over carbohydrate metabolism [24].

Fig. 4
figure 4

Neurofibromatosis type 1 metabolic-related symptoms. Multisystemic alterations in metabolism that are commonly associated with NF1. Modified from Masgras and Rasola (2021) [66]. Created with BioRender.com

In addition to altered body composition, individuals with NF1 present a metabolic profile characterized by lower fasting blood glucose levels [147], heightened insulin sensitivity [148], and a reduced incidence of diabetes mellitus [140, 149, 150]. Hormonal dysregulation in NF1 patients, involving alterations in leptin, vistafin, and adiponectin, may contribute to these metabolic features [148]. Also noted are decreased levels of calcium, calcitonin, and vitamin D [143]. In adults with NF1, there is a notable reduction in cerebral glucose metabolism in the thalamus, as evidenced by positron emission tomography scans [23]. In addition to these differences, individuals with NF1 often experience significant cognitive and physiological fatigue [151], suggesting that metabolic dysregulation may impact brain function.

The Ras/Raf/MEK/ERK pathway regulates multiple metabolic processes, including cell proliferation, protein synthesis, lipid and cholesterol homeostasis, adipocyte differentiation, lipolysis, lipogenesis, gluconeogenesis, and gene expression (Fig. 3) [152, 153]. Consequently, hyperactivation of this pathway due to loss of Nf1 may mediate the observed cellular and systemic metabolic dysfunctions. Nf1-deficient cells exhibit increased glycolysis and reduced mitochondrial respiration mediated through the Ras/MEK/ERK pathway [154].

Metabolic features of the disorder have been recapitulated in animal models of NF1 (Fig. 5). Heterozygous Nf1 (Nf1±) mice exhibit altered body composition, represented by a reduction in fat mass and increased percentage of lean mass [155]. Similar to NF1 patients, the loss of Nf1 function enhances insulin sensitivity and glucose utilization in Nf1± mice [148, 155]. Conditional Nf1 knockout results in metabolic changes in muscles, including reduced muscle growth, increased triglyceride content, malformations (cardiac, renal, hepatic, and skeletal muscle defects), and prenatal lethality [155,156,157]. Inactivation of Nf1 in skeletal muscle (Nf1MyoD−/−) proves lethal within the first week of life; during development, animals with the mutation exhibit stunted growth and intramyocellular lipid accumulation, indicative of impaired long chain fatty acid metabolism [156, 158]. Notably, muscle samples from limb-specific Nf1 conditional knockout (Nf1Prx1−/−) mice recapitulate some of the pathological findings observed in human NF1 muscle biopsies, including intramyocellular lipid accumulation, elevated oxidative metabolic enzyme activity, heightened expression of leptin and fatty acid synthase, and reduced fatty acid transporters [156, 158].

Fig. 5
figure 5

Animal and in vitro models of NF1. Strengths of in vitro, murine, Drosophila, zebrafish, and minipig models to investigate NF1. Created with BioRender.com

Muscle weakness in NF1 may stem from changes in lipid storage resembling lipid storage myopathies. Nf1 manipulations in mice suggest a role for Nf1 in metabolic regulation within muscle tissue, suggesting avenues for potential therapeutic interventions. For example, when Nf1 is lost in mesenchymal tissues (Nf1Prx1−/− mice), dietary interventions that reduce long chain fatty acid intake and enrich medium-fatty acids with L-carnitine effectively rescue lipid accumulation and muscle weakness [158]. Additionally, pharmacological intervention using the selective MEK inhibitor, PD98059, rescues postnatal body weight loss and lipid accumulation in mice with muscle-specific Nf1 knockout when administered during pregnancy in Nf1MyoD−/− dams [159]. In pediatric NF1 patients, pharmacological inhibition of MEK with selumetinib or PD0325901 has led to clinically significant improvements in muscular strength [74], supporting a MEK/ERK-dependent mechanism underlying Nf1-associated muscle metabolism. Lastly, recent studies have highlighted the cell-autonomous role of Nf1 in postnatal muscle growth and metabolic homeostasis, with homozygous Nf1 mutations resulting in neonatal lethality [160]. Overall, these data suggest a critical role for Nf1 in muscle development and function, providing insights into potential therapeutic interventions.

Research involving several models suggests that the metabolic alterations associated with NF1 extend beyond muscle tissue. In Drosophila, loss of Nf1 drives multiple phenotypes indicative of metabolic dysfunction. Nf1 mutations decrease body size by 15–25%, mirroring the short stature observed in NF1 patients [68, 80, 141, 161]. Restoring human or Drosophila Nf1 expression in nf1 mutant neurons can rescue the mutant growth defect [77, 161, 162]. Additionally, Drosophila nf1 mutants experience a significant reduction in lifespan due to altered mitochondrial respiration and increased production of reactive oxygen species (ROS) [163]. Overexpressing Nf1 in nf1 mutants rescues lifespan, enhances mitochondrial respiration, and significantly reduces ROS production [163]. Furthermore, Nf1 regulates metabolic homeostasis, with Nf1 deficiency increasing metabolic rate (CO2 production and O2 consumption), decreasing glycogen and triglyceride stores, and increasing the rate of lipid turnover [164, 165]. Similar to human NF1 patients, Nf1 knockdown in Drosophila results in a reduced RQ [164], indicating increased fat utilization. These effects emanate, at least in part, from neuronal mechanisms [164]. Additionally, loss of Nf1 heightens starvation susceptibility and increases feeding, likely in a compensatory manner to the metabolic alterations [164]. The metabolic phenotype in Drosophila resembles the increased REE observed in NF1 patients [24], suggesting metabolic dysfunction across different species (and cell types). Notably, the Nf1 metabolic and motor (grooming) phenotypes are caused by the loss of Nf1 function in different neural subsets, as knockdown in metabolism-regulating neurons does not affect grooming [164]. Collectively, these metabolic alterations highlight Nf1's tight regulation of metabolic function and its susceptibility to disruption when Nf1 is lost.

The molecular mechanism of Nf1’s effects on metabolism involves its activity as a Ras GAP. The catalytic activity within the GRD of Nf1 is required for Nf1-dependent modulation of metabolic rate. A patient-derived mutation in the GRD (R1320P) fails to rescue metabolic phenotypes [164]. This mutation is at the equivalent residue as R1276P in humans, which reduces Ras-GAP activity by over 1000-fold without impairing any other Nf1 function [61]. Transgenic expression of full-length, wild-type Nf1 selectively in neurons of nf1 mutants restores normal metabolic function [164] (as well as grooming activity) [127]. Additionally, the activation of downstream targets of Nf1 and Ras, such as ERK, play a significant role in driving metabolic effects. Constitutive ERK activation in metabolic-regulating neurons increases metabolic rate, phenocopying the metabolic dysregulation observed in Nf1 mutations [164]. Beyond neuronal and muscle-specific effects, metabolic profiling of mouse embryonic fibroblasts (MEFs) derived from Nf1 knockout animals has revealed significant alterations in cellular bioenergetics. Specifically, Nf1 knockout MEFs displayed diminished mitochondrial activity, driven by elevated glycolysis and decreased respiration [154]. These metabolic shifts stem from heightened Ras/MEK/ERK signaling within mitochondria [154]. Collectively, these results suggest that Nf1 acts in neurons, muscles, and potentially additional cell types to regulate cellular bioenergetics in multiple model systems. Further, the mechanism involves the overactivation of Ras/Raf/MEK/ERK activity. The contributions of other downstream signaling pathways (e.g., the metabolism-regulating mTOR pathway) provide potentially promising avenues for exploration in the study of metabolic regulation in NF1.

Pigmentary lesions

Pigmentary features are crucial for early diagnosis of NF1. CALMs, which are observed in 99% of NF1 patients by age 1 [166], consist of melanocytes with biallelic NF1 inactivation [10, 167]. These features are among the earlies signs of the disease. Additionally, axillary and inguinal freckling, typically appear between 3 to 5 years of age and are present in about 90% of patients by age 7 [166]. Another significant marker are Lisch nodules, which are asymptomatic hyperpigmented iris hamartomas, typically appear by age 5–6. These nodules are present in over 70% of patients by the age of 10 and are observed in over 90% of adults with NF1 [166, 168]. Two NF1 mutations in minipig models, NF1R1947X and NF1+/ex42del, have successfully replicated these pigmentary features, including CALMs and axillary freckling [109, 110]. The loss of Nf1 expression in minipigs models of NF1 results in hyperactivation of the Ras pathway and its effector molecules, linking the signaling cascades to the pathogenesis of cutaneous NF1 features [110]. Finally, homozygous nf1a and nf1b mutant zebrafish larvae exhibit pigmentation anomalies, providing a novel vertebrate model to study pigmentation lesions associated with NF1 [107].

Skeletal abnormalities

Patients with NF1 exhibit a range of skeletal abnormalities, leading to significant morbidity. These osseous defects include both localized and generalized bone deformities, contributing to bone weakening and an increased fracture risk. One of the most significant manifestations is long-bone dysplasia, which affects approximately 5% of individuals with NF1. This condition is characterized by anterolateral bowing of the lower limbs, predominately affecting the tibia, resulting in decreased bone density, increased fracture risk, and pseudarthrosis [11, 92, 169, 170]. Another notable skeletal anomaly is sphenoid-wing dysplasia, which affects up to 11% of NF1 patients and results in distinct cranial deformities [88, 171, 172]. Scoliosis is the most prevalent skeletal defect associated with NF1, occurring in up to 30% of patients and often necessitating surgical intervention in severe cases [173]. Furthermore, NF1 patients tend to be shorter than their healthy counterparts, with 8–15% experiencing a generalized reduction in skeletal bone growth [174, 175]. Individuals with NF1 exhibit both local and general dysregulation of bone resorption and remodeling, leading to increased formation of osteoclast [176, 177]. NF1 patients often have a reduced bone mineral density, osteoporosis, and increased risk of bone fractures [178].

In animal models, Nf1 is critical in skeletal development. In mice, Nf1 is essential for joint development; conditional Nf1 loss during early limb development induces multiple joint abnormalities, including deformities in the hip, knee, and elbow [179]. Similar to human NF1 patients, tibia bowing occurs in mice due to Nf1 deficiency, leading to growth retardation and abnormal growth plate development [179]. Moreover, the expression of Nf1 in bone marrow osteoprogenitors is crucial for maintaining adult skeletal integrity [180]. Loss of Nf1 in these cells leads to skeletal anomalies resembling those seen in NF1 patients, including progressive scoliosis, kyphosis, tibial bowing, and deformities in the skull and anterior chest wall [181]. Additionally, Nf1 loss in osteochondroprogenitors results in decreased bone mass, increased cortical porosity, severe short stature, and intervertebral disc defects [181]. Similar skeletal phenotypes are observed in a minipig model of NF1. The NF1+/ex42del minipig model develops tibial bone curvature and shorter long bones such as the femur, tibia, humerus, ulna, metacarpals, indicative of reduced stature [182]. These animal models further substantiate the critical role of Nf1 in skeletal integrity and development.

Developmental alterations

The loss of Nf1 results in developmental alterations that may contribute to the clinical manifestations of NF1. Magnetic resonance imaging studies have documented alterations in neuronal development among NF1 patients, including increased total brain and white matter volumes. Notably, enlargements in subregions including corpus callosum and brainstem, as well as increased optic nerve tortuosity, are commonly observed [20, 183,184,185,186,187,188,189,190,191,192,193,194,195,196]. Along with structural differences, loss of Nf1 is linked to a range of functional changes in neuronal activity. These include changes in cortical association networks and functional connectivity within the default network, corticostriatal functional circuits, and areas critical for cognition, social functioning, executive functioning, and spatial working memory [197,198,199,200,201,202,203,204,205]. Collectively, these observations underscore the role of Nf1 in modulating brain development, connectivity, and function.

Conditional knockout of neuronal Nf1 in mice mirrors human pathology by enlarging the corpus callosum, an effect which can be rescued by inhibiting Raf/MEK/ERK signaling during neonatal development [206]. In a more severe manifestation, homozygous Nf1 knockout (Nf1−/−) mice exhibit gestational lethality due to severe cardiovascular abnormalities, highlighting a significant role for Nf1 during tissue development [157]. Heterozygous Nf1 mutant (Nf1±) mice, although viable, exhibit numerous brain abnormalities, including enlarged glia, increased neuron numbers, astrocyte proliferation, and neural tube closure defects [157, 207,208,209]. Neuron-specific Nf1 knockout in mice also reveals brain abnormalities, including abnormal cortex development, increased cell density, heightened astrocyte proliferation, and reduced cortical thickness [210]. Loss of Nf1 function in neurons, rather than glia, in mice causes growth defects, further underscoring Nf1’s critical role in neuronal growth and development [210, 211].

The structural alterations resulting from the loss of Nf1 raise significant questions about whether and how these developmental alterations influence behavior. Understanding this relationship is crucial for optimizing the timing of therapeutic interventions by, for instance, allowing for targeted treatments during appropriate developmental times. While direct behavioral correlations in humans are yet to be established, animal models have provided valuable insights. For example, in Drosophila, the developmental contribution of Nf1 to adult behavior has been parsed. Loss of Nf1 increases the frequency of spontaneous grooming behavior in adult animals [128]. Additionally, studies using conditional knockdown of Nf1 in neurons across developmental time windows revealed that loss of Nf1 during a critical developmental period impairs motor (grooming) behavior, whereas similar alterations either earlier (embryonic stage) or later (adult stage) do not have the same effect [127]. The mechanisms by which Nf1 loss impacts neuronal development in NF1 are diverse and complex. They may include altered cell growth, division, differentiation/specification, apoptosis, dendrite & axon targeting, synaptogenesis, activity-dependent synaptic refinement, hormone responsivity, and nutrient responsivity [210,211,212,213]. Future mechanistic studies are necessary to dissect how developmental disruptions due to the loss of Nf1 result in adult phenotypes.

Conclusions

Research utilizing animal models and in vitro studies has elucidated the significant effects of Nf1 in the nervous system and behavior, identifying its significance in normal development and function. Nf1 influences cellular and systemic physiology via multiple molecular and cellular mechanisms, including alterations in metabolism. Several major models, such as mice, Drosophila, minipigs, and zebrafish, have considerably advanced our understanding of Nf1's mechanistic role within the nervous system and its effects on metabolic regulation. Continued advancements in these areas hold promise for the development of novel targeted therapies and interventions aimed at improving the outcomes and quality of life for individuals with NF1.

Availability of data and materials

N/A (Review).

Abbreviations

ADHD:

Attention-deficit/hyperactivity disorder

ALK:

Anaplastic Lymphoma Kinase

ASD:

Autism spectrum disorder

BMI:

Body mass index

CALM:

Café-au-lait macule

cAMP:

Cyclic adenosine monophosphate

CN:

Cutaneous neurofibromas

CNS:

Central nervous system

FDA:

Food and Drug Administration

GAP:

GTPase activating protein

GPCR:

G protein-coupled receptor

GRD:

GAP-related domain

hiPSC:

Human induced pluripotent stem cells

MAPK:

Mitogen-activated protein kinase

MEF:

Mouse embryonic fibroblasts

MPNST:

Malignant peripheral nerve sheath tumors

NF1:

Neurofibromatosis type 1

NF1 :

Human neurofibromin 1 gene

Nf1 :

Non-human neurofibromin 1 gene

Nf1:

Neurofibromin protein

nf1 :

Invertebrate mutant neurofibromin protein

OPG:

Optic pathway glioma

PKA:

Protein kinase A

PKCζ:

Protein kinase C zeta

pNF:

Plexiform neurofibroma

REE:

Resting energy expenditure

ROS:

Reactive oxygen species

RQ:

Respiratory quotient

RTK:

Receptor tyrosine kinase

References

  1. Hirbe AC, Gutmann DH. Neurofibromatosis type 1: a multidisciplinary approach to care. Lancet Neurol. 2014;13(8):834–43.

    Article  PubMed  Google Scholar 

  2. Uusitalo E, Leppävirta J, Koffert A, Suominen S, Vahtera J, Vahlberg T, et al. Incidence and mortality of neurofibromatosis: a total population study in finland. J Investig Dermatol. 2015;135(3):904–6.

    Article  PubMed  CAS  Google Scholar 

  3. Evans DG, Howard E, Giblin C, Clancy T, Spencer H, Huson SM, et al. Birth incidence and prevalence of tumor-prone syndromes: estimates from a UK family genetic register service. Am J Med Genet A. 2010;152A(2):327–32.

    Article  PubMed  CAS  Google Scholar 

  4. Huson SM, Compston DA, Clark P, Harper PS. A genetic study of von Recklinghausen neurofibromatosis in south east Wales. I. Prevalence, fitness, mutation rate, and effect of parental transmission on severity. J Med Genet. 1989;26(11):704–11.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  5. Sanchez GM, Siuda T. Ebers Papyrus case #873: a probable case of neurofibromatosis 1. S D J Med. 2002;55(12):529–35.

    PubMed  Google Scholar 

  6. Ruggieri M, Praticò AD, Caltabiano R, Polizzi A. Early history of the different forms of neurofibromatosis from ancient Egypt to the British Empire and beyond: First descriptions, medical curiosities, misconceptions, landmarks, and the persons behind the syndromes. Am J Med Genet A. 2018;176(3):515–50.

    Article  PubMed  Google Scholar 

  7. Von Recklinghausen FD. Uber ide multiplen Fibrome der Haut und ihre beziehung zu den multiplen Neuromen. Berlin: Hirschwald; 1882. p. 3–18.

    Google Scholar 

  8. Crump T. Advances in neurology: Translation of case reports in Ueber die multiplen Fibrome der Haut und ihre Beziehung zu den multiplen Neuromen by F. v. Recklinghausen. Arch Neurol. 1981;29:259–75.

    CAS  Google Scholar 

  9. Schull WJ, Neel JV. A clinical, pathological, and genetic study of multiple neurofibromatosis: Charles C. Thomas. 1956.

    Google Scholar 

  10. Neurofibromatosis: Conference Statement. Arch Neurol. 1988;45(5):575–8. https://doi.org/10.1001/archneur.1988.00520290115023.

  11. Legius E, Messiaen L, Wolkenstein P, Pancza P, Avery RA, Berman Y, et al. Revised diagnostic criteria for neurofibromatosis type 1 and Legius syndrome: an international consensus recommendation. Genet Med. 2021;23(8):1506–13.

    Article  PubMed  PubMed Central  Google Scholar 

  12. Barker D, Wright E, Nguyen K, Cannon L, Fain P, Goldgar D, et al. Gene for von Recklinghausen neurofibromatosis is in the pericentromeric region of chromosome 17. Science. 1987;236(4805):1100–2.

    Article  PubMed  CAS  Google Scholar 

  13. Seizinger BR, Rouleau GA, Ozelius LJ, Lane AH, Faryniarz AG, Chao MV, et al. Genetic linkage of von Recklinghausen neurofibromatosis to the nerve growth factor receptor gene. Cell. 1987;49(5):589–94.

    Article  PubMed  CAS  Google Scholar 

  14. Collins FS, O’Connell P, Ponder BAJ, Seizinger BR. Progress towards identifying the neurofibromatosis (NF1) gene. Trends Genet. 1989;5:217–21.

    Article  PubMed  CAS  Google Scholar 

  15. Ballester R, Marchuk D, Boguski M, Saulino A, Letcher R, Wigler M, et al. The NF1 locus encodes a protein functionally related to mammalian GAP and yeast IRA proteins. Cell. 1990;63(4):851–9.

    Article  PubMed  CAS  Google Scholar 

  16. Martin GA, Viskochil D, Bollag G, McCabe PC, Crosier WJ, Haubruck H, et al. The GAP-related domain of the neurofibromatosis type 1 gene product interacts with ras p21. Cell. 1990;63(4):843–9.

    Article  PubMed  CAS  Google Scholar 

  17. Wallace MR, Marchuk DA, Andersen LB, Letcher R, Odeh HM, Saulino AM, et al. Type 1 neurofibromatosis gene: identification of a large transcript disrupted in three NF1 patients. Science. 1990;249(4965):181–6.

    Article  PubMed  CAS  Google Scholar 

  18. DeClue JE, Cohen BD, Lowy DR. Identification and characterization of the neurofibromatosis type 1 protein product. Proc Natl Acad Sci U S A. 1991;88(22):9914–8.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  19. Gutmann DH, Wood DL, Collins FS. Identification of the neurofibromatosis type 1 gene product. Proc Natl Acad Sci. 1991;88(21):9658–62.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  20. Pride N, Payne JM, Webster R, Shores EA, Rae C, North KN. Corpus callosum morphology and its relationship to cognitive function in neurofibromatosis type 1. J Child Neurol. 2010;25(7):834–41.

    Article  PubMed  Google Scholar 

  21. Gill DS, Hyman SL, Steinberg A, North KN. Age-related findings on MRI in neurofibromatosis type 1. Pediatr Radiol. 2006;36(10):1048–56.

    Article  PubMed  Google Scholar 

  22. Hyman SL, Shores A, North KN. The nature and frequency of cognitive deficits in children with neurofibromatosis type 1. Neurology. 2005;65(7):1037–44.

    Article  PubMed  Google Scholar 

  23. Apostolova I, Derlin T, Salamon J, Amthauer H, Granstrom S, Brenner W, et al. Cerebral glucose metabolism in adults with neurofibromatosis type 1. Brain Res. 2015;1625:97–101.

    Article  PubMed  CAS  Google Scholar 

  24. Souza MLR, Jansen AK, Rodrigues LOC, Vilela DLS, Kakehasi AM, Martins AS, et al. Increased resting metabolism in neurofibromatosis type 1. Clin Nutr ESPEN. 2019;32:44–9.

    Article  PubMed  Google Scholar 

  25. Gutmann DH, Ferner RE, Listernick RH, Korf BR, Wolters PL, Johnson KJ. Neurofibromatosis type 1. Nat Rev Dis Primers. 2017;3:17004.

    Article  PubMed  Google Scholar 

  26. Ratner N, Miller SJ. A RASopathy gene commonly mutated in cancer: the neurofibromatosis type 1 tumour suppressor. Nat Rev Cancer. 2015;15(5):290–301.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  27. Upadhyaya M, Cooper DN. Neurofibromatosis Type 1. Heidelberg: Springer; 2012.

    Book  Google Scholar 

  28. Campian J, Gutmann DH. CNS Tumors in Neurofibromatosis. J Clin Oncol. 2017;35(21):2378–85.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  29. Hyman SL, Arthur E, North KN. Learning disabilities in children with neurofibromatosis type 1: subtypes, cognitive profile, and attention-deficit- hyperactivity disorder. Dev Med Child Neurol. 2006;48(12):973–7.

    PubMed  Google Scholar 

  30. Stewart DR, Korf BR, Nathanson KL, Stevenson DA, Yohay K. Care of adults with neurofibromatosis type 1: a clinical practice resource of the American College of Medical Genetics and Genomics (ACMG). Genet Med. 2018;20(7):671–82.

    Article  PubMed  Google Scholar 

  31. Evans DGR, O’Hara C, Wilding A, Ingham SL, Howard E, Dawson J, et al. Mortality in neurofibromatosis 1: in North West England: an assessment of actuarial survival in a region of the UK since 1989. Eur J Hum Genet. 2011;19(11):1187–91.

    Article  PubMed  PubMed Central  Google Scholar 

  32. Rasmussen SA, Yang Q, Friedman JM. Mortality in neurofibromatosis 1: an analysis using U.S. death certificates. Am J Hum Genet. 2001;68(5):1110–8.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  33. Viskochil D, Buchberg AM, Xu G, Cawthon RM, Stevens J, Wolff RK, et al. Deletions and a translocation interrupt a cloned gene at the neurofibromatosis type 1 locus. Cell. 1990;62(1):187–92.

    Article  PubMed  CAS  Google Scholar 

  34. Shen MH, Harper PS, Upadhyaya M. Molecular genetics of neurofibromatosis type 1 (NF1). J Med Genet. 1996;33(1):2–17.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  35. Li C, Cheng Y, Gutmann DA, Mangoura D. Differential localization of the neurofibromatosis 1 (NF1) gene product, neurofibromin, with the F-actin or microtubule cytoskeleton during differentiation of telencephalic neurons. Dev Brain Res. 2001;130(2):231–48.

    Article  CAS  Google Scholar 

  36. Stowe IB, Mercado EL, Stowe TR, Bell EL, Oses-Prieto JA, Hernandez H, et al. A shared molecular mechanism underlies the human rasopathies Legius syndrome and Neurofibromatosis-1. Genes Dev. 2012;26(13):1421–6.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  37. Nordlund M, Gu X, Shipley M, Ratner N. Neurofibromin is enriched in the endoplasmic reticulum of CNS neurons. J Neurosci. 1993;13(4):1588–600.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  38. Roudebush M, Slabe T, Sundaram V, Hoppel CL, Golubic M, Stacey DW. Neurofibromin Colocalizes with Mitochondria in Cultured Cells. Exp Cell Res. 1997;236(1):161–72.

    Article  PubMed  CAS  Google Scholar 

  39. Daston MM, Scrable H, Nordlund M, Sturbaum AK, Nissen LM, Ratner N. The protein product of the neurofibromatosis type 1 gene is expressed at highest abundance in neurons, Schwann cells, and oligodendrocytes. Neuron. 1992;8(3):415–28.

    Article  PubMed  CAS  Google Scholar 

  40. Philpott C, Tovell H, Frayling IM, Cooper DN, Upadhyaya M. The NF1 somatic mutational landscape in sporadic human cancers. Hum Genomics. 2017;11(1):13.

    Article  PubMed  PubMed Central  Google Scholar 

  41. Easton DF, Ponder MA, Huson SM, Ponder BA. An analysis of variation in expression of neurofibromatosis (NF) type 1 (NF1): evidence for modifying genes. Am J Hum Genet. 1993;53(2):305–13.

    PubMed  PubMed Central  CAS  Google Scholar 

  42. Anastasaki C, Woo AS, Messiaen LM, Gutmann DH. Elucidating the impact of neurofibromatosis-1 germline mutations on neurofibromin function and dopamine-based learning. Hum Mol Genet. 2015;24(12):3518–28.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  43. Bajenaru ML, Donahoe J, Corral T, Reilly KM, Brophy S, Pellicer A, et al. Neurofibromatosis 1 (NF1) heterozygosity results in a cell-autonomous growth advantage for astrocytes. Glia. 2001;33(4):314–23.

    Article  PubMed  CAS  Google Scholar 

  44. Costa RM, Federov NB, Kogan JH, Murphy GG, Stern J, Ohno M, et al. Mechanism for the learning deficits in a mouse model of neurofibromatosis type 1. Nature. 2002;415(6871):526–30.

    Article  PubMed  CAS  Google Scholar 

  45. Cui Y, Costa RM, Murphy GG, Elgersma Y, Zhu Y, Gutmann DH, et al. Neurofibromin regulation of ERK signaling modulates GABA release and learning. Cell. 2008;135(3):549–60.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  46. Geist RT, Gutmann DH. Expression of a developmentally-regulated neuron-specific isoform of the neurofibromatosis 1 (NF1) gene. Neurosci Lett. 1996;211(2):85–8.

    Article  PubMed  CAS  Google Scholar 

  47. Gutmann DH, Andersen LB, Cole JL, Swaroop M, Collins FS. An alternatively-spliced mRNA in the carboxy terminus of the neurofibromatosis type 1 (NF1) gene is expressed in muscle. Hum Mol Genet. 1993;2(7):989–92.

    Article  CAS  Google Scholar 

  48. Costa RM, Yang T, Huynh DP, Pulst SM, Viskochil DH, Silva AJ, et al. Learning deficits, but normal development and tumor predisposition, in mice lacking exon 23a of Nf1. Nat Genet. 2001;27(4):399–405.

    Article  PubMed  CAS  Google Scholar 

  49. Gutmann DH, Cole JL, Collins FS. Expression of the neurofibromatosis type 1 (NF1) gene during mouse embryonic development. Prog Brain Res. 1995;105:327–35.

    Article  PubMed  CAS  Google Scholar 

  50. Xu H, Gutmann DH. Mutations in the GAP-related domain impair the ability of neurofibromin to associate with microtubules. Brain Res. 1997;759(1):149–52.

    Article  PubMed  CAS  Google Scholar 

  51. Gutmann DH, Geist RT, Rose K, Wright DE. Expression of two new protein isoforms of the neurofibromatosis type 1 gene product, neurofibromin, in muscle tissues. Dev Dyn. 1995;202(3):302–11.

    Article  PubMed  CAS  Google Scholar 

  52. Gutmann DH, Zhang Y, Hirbe A. Developmental regulation of a neuron-specific neurofibromatosis 1 isoform. Ann Neurol. 1999;46(5):777–82.

    Article  PubMed  CAS  Google Scholar 

  53. Summers MA, Quinlan KG, Payne JM, Little DG, North KN, Schindeler A. Skeletal muscle and motor deficits in Neurofibromatosis Type 1. J Musculoskelet Neuronal Interact. 2015;15(2):161–70.

    PubMed  PubMed Central  CAS  Google Scholar 

  54. Gutman DH, Cole JL, Collins FS. Modulation of neurofibromatosis type 1 gene expression during in vitro myoblast differentiation. J Neurosci Res. 1994;37(3):398–405.

    Article  Google Scholar 

  55. Kaufmann D, Müller R, Kenner O, Leistner W, Hein C, Vogel W, et al. The N-terminal splice product NF1-10a-2 of the NF1 gene codes for a transmembrane segment. Biochem Biophys Res Commun. 2002;294(2):496–503.

    Article  PubMed  CAS  Google Scholar 

  56. Gutmann DH, Geist RT, Wright DE, Snider WD. Expression of the neurofibromatosis 1 (NF1) isoforms in developing and adult rat tissues. Cell Growth Differ. 1995;6(3):315–23.

    PubMed  CAS  Google Scholar 

  57. Vandenbroucke I, Vandesompele J, De Paepe A, Messiaen L. Quantification of NF1 transcripts reveals novel highly expressed splice variants. FEBS Lett. 2002;522(1–3):71–6.

    Article  PubMed  CAS  Google Scholar 

  58. Young LC, de GoldsteinSalazar R, Han SW, Huang ZYS, Merk A, Drew M, et al. Destabilizing NF1 variants act in a dominant negative manner through neurofibromin dimerization. Proc Natl Acad Sci U S A. 2023;120(5):e2208960120.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  59. Mo J, Moye SL, McKay RM, Le LQ. Neurofibromin and suppression of tumorigenesis: beyond the GAP. Oncogene. 2022;41:1235–51.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  60. Anastasaki C, Orozco P, Gutmann DH. RAS and beyond: the many faces of the neurofibromatosis type 1 protein. Dis Models Mech. 2022;15(2):dmm049362.

    Article  CAS  Google Scholar 

  61. Klose A, Ahmadian MR, Schuelke M, Scheffzek K, Hoffmeyer S, Gewies A, et al. Selective disactivation of neurofibromin GAP activity in neurofibromatosis type 1. Hum Mol Genet. 1998;7(8):1261–8.

    Article  PubMed  CAS  Google Scholar 

  62. Scheffzek K, Ahmadian MR, Wiesmüller L, Kabsch W, Stege P, Schmitz F, et al. Structural analysis of the GAP-related domain from neurofibromin and its implications. Embo j. 1998;17(15):4313–27.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  63. Dasgupta B, Yi Y, Chen DY, Weber JD, Gutmann DH. Proteomic analysis reveals hyperactivation of the mammalian target of rapamycin pathway in neurofibromatosis 1-associated human and mouse brain tumors. Cancer Res. 2005;65(7):2755–60.

    Article  PubMed  CAS  Google Scholar 

  64. Johannessen CM, Reczek EE, James MF, Brems H, Legius E, Cichowski K. The NF1 tumor suppressor critically regulates TSC2 and mTOR. Proc Natl Acad Sci U S A. 2005;102(24):8573–8.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  65. Scheffzek K, Shivalingaiah G. Ras-specific GTPase-activating proteins-structures, mechanisms, and interactions. Cold Spring Harb Perspect Med. 2019;9(3):a031500.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  66. Masgras I, Rasola A. Metabolic Features of Neurofibromatosis Type 1-Associated Tumors. In: Juichiro N, Yuichi Y, editors. Clinical and Basic Aspects of Neurofibromatosis Type 1. Rijeka: IntechOpen; 2021. p. Ch. 9.

    Google Scholar 

  67. Gouzi JY, Moressis A, Walker JA, Apostolopoulou AA, Palmer RH, Bernards A, et al. The receptor tyrosine kinase Alk controls neurofibromin functions in Drosophila growth and learning. PLoS Genet. 2011;7(9):e1002281.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  68. Walker JA, Gouzi JY, Long JB, Huang S, Maher RC, Xia H, et al. Genetic and functional studies implicate synaptic overgrowth and ring gland cAMP/PKA signaling defects in the Drosophila melanogaster neurofibromatosis-1 growth deficiency. PLoS Genet. 2013;9(11):e1003958.

    Article  PubMed  PubMed Central  Google Scholar 

  69. Lorén CE, Scully A, Grabbe C, Edeen PT, Thomas J, McKeown M, et al. Identification and characterization of DAlk: a novel Drosophila melanogaster RTK which drives ERK activation in vivo. Genes Cells. 2001;6(6):531–44.

    Article  PubMed  PubMed Central  Google Scholar 

  70. Palmer RH, Vernersson E, Grabbe C, Hallberg B. Anaplastic lymphoma kinase: signalling in development and disease. Biochem J. 2009;420(3):345–61.

    Article  PubMed  CAS  Google Scholar 

  71. Xie K, Colgan LA, Dao MT, Muntean BS, Sutton LP, Orlandi C, et al. NF1 is a direct g protein effector essential for opioid signaling to ras in the striatum. Current biology : CB. 2016;26(22):2992–3003.

    Article  PubMed  CAS  Google Scholar 

  72. Sánchez-Alegría K, Flores-León M, Avila-Muñoz E, Rodríguez-Corona N, Arias C. PI3K signaling in neurons: a central node for the control of multiple functions. Int J Mol Sci. 2018;19(12):3725.

    Article  PubMed  PubMed Central  Google Scholar 

  73. Mukhopadhyay S, Maitra A, Choudhury S. Selumetinib: the first ever approved drug for neurofibromatosis-1 related inoperable plexiform neurofibroma. Curr Med Res Opin. 2021;37(5):789–94.

    Article  PubMed  CAS  Google Scholar 

  74. Gross AM, Wolters PL, Dombi E, Baldwin A, Whitcomb P, Fisher MJ, et al. Selumetinib in Children with Inoperable Plexiform Neurofibromas. N Engl J Med. 2020;382(15):1430–42.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  75. Bergqvist C, Wolkenstein P. MEK inhibitors in RASopathies. Curr Opin Oncol. 2021;33(2):110–9.

    Article  PubMed  CAS  Google Scholar 

  76. Dasgupta B, Dugan LL, Gutmann DH. The neurofibromatosis 1 gene product neurofibromin regulates pituitary adenylate cyclase-activating polypeptide-mediated signaling in astrocytes. J Neurosci. 2003;23(26):8949–54.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  77. Tong J, Hannan F, Zhu Y, Bernards A, Zhong Y. Neurofibromin regulates G protein-stimulated adenylyl cyclase activity. Nat Neurosci. 2002;5(2):95–6.

    Article  PubMed  CAS  Google Scholar 

  78. Kim HA, Ratner N, Roberts TM, Stiles CD. Schwann cell proliferative responses to cAMP and Nf1 are mediated by cyclin D1. J Neurosci. 2001;21(4):1110–6.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  79. Guo HF, Tong J, Hannan F, Luo L, Zhong Y. A neurofibromatosis-1-regulated pathway is required for learning in Drosophila. Nature. 2000;403(6772):895–8.

    Article  PubMed  CAS  Google Scholar 

  80. The I, Hannigan GE, Cowley GS, Reginald S, Zhong Y, Gusella JF, et al. Rescue of a Drosophila NF1 mutant phenotype by protein kinase A. Science. 1997;276(5313):791–4.

    Article  PubMed  CAS  Google Scholar 

  81. Wolman MA, de Groh ED, McBride SM, Jongens TA, Granato M, Epstein JA. Modulation of cAMP and ras signaling pathways improves distinct behavioral deficits in a zebrafish model of neurofibromatosis type 1. Cell Rep. 2014;8(5):1265–70.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  82. Anastasaki C, Gutmann DH. Neuronal NF1/RAS regulation of cyclic AMP requires atypical PKC activation. Hum Mol Genet. 2014;23(25):6712–21.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  83. Brown JA, Diggs-Andrews KA, Gianino SM, Gutmann DH. Neurofibromatosis-1 heterozygosity impairs CNS neuronal morphology in a cAMP/PKA/ROCK-dependent manner. Mol Cell Neurosci. 2012;49(1):13–22.

    Article  PubMed  CAS  Google Scholar 

  84. Jouhilahti EM, Peltonen S, Heape AM, Peltonen J. The pathoetiology of neurofibromatosis 1. Am J Pathol. 2011;178(5):1932–9.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  85. Laycock-van Spyk S, Thomas N, Cooper DN, Upadhyaya M. Neurofibromatosis type 1-associated tumours: their somatic mutational spectrum and pathogenesis. Hum Genomics. 2011;5(6):623–90.

    Article  PubMed  Google Scholar 

  86. Dombi E, Solomon J, Gillespie AJ, Fox E, Balis FM, Patronas N, et al. NF1 plexiform neurofibroma growth rate by volumetric MRI: relationship to age and body weight. Neurology. 2007;68(9):643–7.

    Article  PubMed  CAS  Google Scholar 

  87. Reed N, Gutmann DH. Tumorigenesis in neurofibromatosis: new insights and potential therapies. Trends Mol Med. 2001;7(4):157–62.

    Article  PubMed  CAS  Google Scholar 

  88. Choi J, An S, Lim SY. Current concepts of neurofibromatosis type 1: pathophysiology and treatment. Arch Craniofac Surg. 2022;23(1):6–16.

    Article  PubMed  PubMed Central  Google Scholar 

  89. Hwang IK, Hahn SM, Kim HS, Kim SK, Kim HS, Shin KH, et al. Outcomes of treatment for malignant peripheral nerve sheath tumors: different clinical features associated with neurofibromatosis Type 1. Cancer Res Treat. 2017;49(3):717–26.

    Article  PubMed  CAS  Google Scholar 

  90. Listernick R, Charrow J, Greenwald M, Mets M. Natural history of optic pathway tumors in children with neurofibromatosis type 1: a longitudinal study. J Pediatr. 1994;125(1):63–6.

    Article  PubMed  CAS  Google Scholar 

  91. Ferner RE. Neurofibromatosis 1 and neurofibromatosis 2: a twenty first century perspective. Lancet Neurol. 2007;6(4):340–51.

    Article  PubMed  Google Scholar 

  92. Cimino PJ, Gutmann DH. Chapter 51 - Neurofibromatosis type 1. In: Geschwind DH, Paulson HL, Klein C, editors. Handbook of Clinical Neurology. 148. Amsterdam: Elsevier; 2018. p. 799–811.

    Google Scholar 

  93. Diggs-Andrews KA, Brown JA, Gianino SM, Rubin JB, Wozniak DF, Gutmann DH. Sex Is a major determinant of neuronal dysfunction in neurofibromatosis type 1. Ann Neurol. 2014;75(2):309–16.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  94. Fisher MJ, Loguidice M, Gutmann DH, Listernick R, Ferner RE, Ullrich NJ, et al. Visual outcomes in children with neurofibromatosis type 1–associated optic pathway glioma following chemotherapy: a multicenter retrospective analysis. Neuro Oncol. 2012;14(6):790–7.

    Article  PubMed  PubMed Central  Google Scholar 

  95. Sani I, Albanese A. Endocrine long-term follow-up of children with neurofibromatosis type 1 and optic pathway glioma. Horm Res Paediatr. 2017;87(3):179–88.

    Article  PubMed  CAS  Google Scholar 

  96. Mo J, Anastasaki C, Chen Z, Shipman T, Papke J, Yin K, et al. Humanized neurofibroma model from induced pluripotent stem cells delineates tumor pathogenesis and developmental origins. J Clin Investig. 2021;131(1):e139807.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  97. Anastasaki C, Mo J, Chen JK, Chatterjee J, Pan Y, Scheaffer SM, et al. Neuronal hyperexcitability drives central and peripheral nervous system tumor progression in models of neurofibromatosis-1. Nat Commun. 2022;13(1):2785.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  98. Pan Y, Hysinger JD, Barron T, Schindler NF, Cobb O, Guo X, et al. NF1 mutation drives neuronal activity-dependent initiation of optic glioma. Nature. 2021;594(7862):277–82.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  99. Wang Y, Nicol GD, Clapp DW, Hingtgen CM. Sensory neurons from Nf1 haploinsufficient mice exhibit increased excitability. J Neurophysiol. 2005;94(6):3670–6.

    Article  PubMed  CAS  Google Scholar 

  100. Anastasaki C, Chatterjee J, Koleske JP, Gao Y, Bozeman SL, Kernan CM, et al. NF1 mutation-driven neuronal hyperexcitability sets a threshold for tumorigenesis and therapeutic targeting of murine optic glioma. Neuro Oncol. 2024;26:1496–508.

    Article  PubMed  Google Scholar 

  101. Jessen WJ, Miller SJ, Jousma E, Wu J, Rizvi TA, Brundage ME, et al. MEK inhibition exhibits efficacy in human and mouse neurofibromatosis tumors. J Clin Investig. 2013;123(1):340–7.

    Article  PubMed  CAS  Google Scholar 

  102. Dombi E, Baldwin A, Marcus LJ, Fisher MJ, Weiss B, Kim A, et al. Activity of Selumetinib in Neurofibromatosis Type 1-Related Plexiform Neurofibromas. N Engl J Med. 2016;375(26):2550–60.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  103. Gross AM, Dombi E, Widemann BC. Current status of MEK inhibitors in the treatment of plexiform neurofibromas. Childs Nerv Syst. 2020;36(10):2443–52.

    Article  PubMed  Google Scholar 

  104. Kaul A, Toonen JA, Cimino PJ, Gianino SM, Gutmann DH. Akt- or MEK-mediated mTOR inhibition suppresses Nf1 optic glioma growth. Neuro Oncol. 2015;17(6):843–53.

    Article  PubMed  CAS  Google Scholar 

  105. Padmanabhan A, Lee JS, Ismat FA, Lu MM, Lawson ND, Kanki JP, et al. Cardiac and vascular functions of the zebrafish orthologues of the type I neurofibromatosis gene NFI. Proc Natl Acad Sci U S A. 2009;106(52):22305–10.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  106. Lee JS, Padmanabhan A, Shin J, Zhu S, Guo F, Kanki JP, et al. Oligodendrocyte progenitor cell numbers and migration are regulated by the zebrafish orthologs of the NF1 tumor suppressor gene. Hum Mol Genet. 2010;19(23):4643–53.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  107. Shin J, Padmanabhan A, de Groh ED, Lee JS, Haidar S, Dahlberg S, et al. Zebrafish neurofibromatosis type 1 genes have redundant functions in tumorigenesis and embryonic development. Dis Model Mech. 2012;5(6):881–94.

    PubMed  PubMed Central  CAS  Google Scholar 

  108. Luo J, Liu P, Lu C, Bian W, Su D, Zhu C, et al. Stepwise crosstalk between aberrant Nf1, Tp53 and Rb signalling pathways induces gliomagenesis in zebrafish. Brain. 2021;144(2):615–35.

    Article  PubMed  Google Scholar 

  109. Isakson SH, Rizzardi AE, Coutts AW, Carlson DF, Kirstein MN, Fisher J, et al. Genetically engineered minipigs model the major clinical features of human neurofibromatosis type 1. Commun Biol. 2018;1(1):158.

    Article  PubMed  PubMed Central  Google Scholar 

  110. White KA, Swier VJ, Cain JT, Kohlmeyer JL, Meyerholz DK, Tanas MR, et al. A porcine model of neurofibromatosis type 1 that mimics the human disease. JCI Insight. 2018;3(12):e120402.

    Article  PubMed  PubMed Central  Google Scholar 

  111. Mautner VF, Kluwe L, Thakker SD, Leark RA. Treatment of ADHD in neurofibromatosis type 1. Dev Med Child Neurol. 2002;44(3):164–70.

    Article  PubMed  Google Scholar 

  112. Soucy EA, Gao F, Gutmann DH, Dunn CM. Developmental delays in children with neurofibromatosis type 1. J Child Neurol. 2012;27(5):641–4.

    Article  PubMed  Google Scholar 

  113. Ferner RE, Hughes RAC, Weinman J. Intellectual impairment in neurofibromatosis 1. J Neurol Sci. 1996;138(1):125–33.

    Article  PubMed  CAS  Google Scholar 

  114. Morris SM, Acosta MT, Garg S, Green J, Huson S, Legius E, et al. Disease Burden and Symptom Structure of Autism in Neurofibromatosis Type 1: a study of the international NF1-ASD Consortium Team (INFACT). JAMA Psychiatry. 2016;73:1276–84.

    Article  PubMed  PubMed Central  Google Scholar 

  115. Plasschaert E, Van Eylen L, Descheemaeker M-J, Noens I, Legius E, Steyaert J. Executive functioning deficits in children with neurofibromatosis type 1: The influence of intellectual and social functioning. Am J Med Genet B Neuropsychiatr Genet. 2016;171(3):348–62.

    Article  Google Scholar 

  116. Plasschaert E, Descheemaeker MJ, Van Eylen L, Noens I, Steyaert J, Legius E. Prevalence of Autism Spectrum Disorder symptoms in children with neurofibromatosis type 1. Am J Med Genet B Neuropsychiatr Genet. 2015;168b(1):72–80.

    Article  PubMed  Google Scholar 

  117. Garg S, Green J, Leadbitter K, Emsley R, Lehtonen A, Evans DG, et al. Neurofibromatosis type 1 and autism spectrum disorder. Pediatrics. 2013;132(6):e1642–8.

    Article  PubMed  Google Scholar 

  118. Walsh KS, Velez JI, Kardel PG, Imas DM, Muenke M, Packer RJ, et al. Symptomatology of autism spectrum disorder in a population with neurofibromatosis type 1. Dev Med Child Neurol. 2013;55(2):131–8.

    Article  PubMed  Google Scholar 

  119. Chatterjee N, Perrimon N. What fuels the fly: energy metabolism in Drosophila and its application to the study of obesity and diabetes. Sci Adv. 2021;7(24):eabg4336.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  120. Musselman LP, Kühnlein RP. Drosophila as a model to study obesity and metabolic disease. J Exp Biol. 2018;221(Suppl_1):jeb163881.

    Article  PubMed  Google Scholar 

  121. Gatto CL, Broadie K. Drosophila modeling of heritable neurodevelopmental disorders. Curr Opin Neurobiol. 2011;21(6):834–41.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  122. Williams JA, Su HS, Bernards A, Field J, Sehgal A. A circadian output in Drosophila mediated by neurofibromatosis-1 and Ras/MAPK. Science. 2001;293(5538):2251–6.

    Article  PubMed  CAS  Google Scholar 

  123. Machado Almeida P, Lago Solis B, Stickley L, Feidler A, Nagoshi E. Neurofibromin 1 in mushroom body neurons mediates circadian wake drive through activating cAMP–PKA signaling. Nat Commun. 2021;12(1):5758.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  124. Bai L, Sehgal A. Anaplastic Lymphoma Kinase Acts in the Drosophila Mushroom Body to Negatively Regulate Sleep. PLoS Genet. 2015;11(11):e1005611.

    Article  PubMed  PubMed Central  Google Scholar 

  125. Buchanan ME, Davis RL. A distinct set of Drosophila brain neurons required for neurofibromatosis type 1-dependent learning and memory. J Neurosci. 2010;30(30):10135–43.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  126. Ho IS, Hannan F, Guo HF, Hakker I, Zhong Y. Distinct functional domains of neurofibromatosis type 1 regulate immediate versus long-term memory formation. J Neurosci. 2007;27(25):6852–7.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  127. King LB, Boto T, Botero V, Aviles AM, Jomsky BM, Joseph C, et al. Developmental loss of neurofibromin across distributed neuronal circuits drives excessive grooming in Drosophila. PLoS Genet. 2020;16(7):e1008920.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  128. King LB, Koch M, Murphy KR, Velazquez Y, Ja WW, Tomchik SM. Neurofibromin loss of function drives excessive grooming in drosophila. G3 (Bethesda). 2016;6(4):1083–93.

    Article  PubMed  CAS  Google Scholar 

  129. Brown EB, Zhang J, Lloyd E, Lanzon E, Botero V, Tomchik S, et al. Neurofibromin 1 mediates sleep depth in Drosophila. PLoS Genet. 2023;19(12):e1011049.

    Article  PubMed  PubMed Central  Google Scholar 

  130. Moscato EH, Dubowy C, Walker JA, Kayser MS. Social behavioral deficits with loss of Neurofibromin emerge from peripheral chemosensory neuron dysfunction. Cell Rep. 2020;32(1):107856.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  131. Dyson A, Ryan M, Garg S, Evans DG, Baines RA. Loss of NF1 in drosophila larvae causes tactile hypersensitivity and impaired synaptic transmission at the neuromuscular junction. J Neurosci. 2022;42(50):9450–72.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  132. Dyson A, Ryan M, Garg S, Evans DG, Baines RA. A Targeted, Low-Throughput Compound Screen in a Drosophila Model of Neurofibromatosis Type 1 Identifies Simvastatin and BMS-204352 as Potential Therapies for Autism Spectrum Disorder (ASD). Eneuro. 2023;10(5):ENEURO.0461–ENEURO.0461.

    Article  PubMed  Google Scholar 

  133. Oliveira AF, Yasuda R. Neurofibromin is the major ras inactivator in dendritic spines. J Neurosci. 2014;34(3):776–83.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  134. Li W, Cui Y, Kushner SA, Brown RA, Jentsch JD, Frankland PW, et al. The HMG-CoA reductase inhibitor lovastatin reverses the learning and attention deficits in a mouse model of neurofibromatosis type 1. Curr Biol. 2005;15(21):1961–7.

    Article  PubMed  CAS  Google Scholar 

  135. Guilding C, McNair K, Stone TW, Morris BJ. Restored plasticity in a mouse model of neurofibromatosis type 1 via inhibition of hyperactive ERK and CREB. Eur J Neurosci. 2007;25(1):99–105.

    Article  PubMed  Google Scholar 

  136. Silva AJ, Frankland PW, Marowitz Z, Friedman E, Laszlo GS, Cioffi D, et al. A mouse model for the learning and memory deficits associated with neurofibromatosis type I. Nat Genet. 1997;15(3):281–4.

    Article  PubMed  CAS  Google Scholar 

  137. Hingtgen CM. Neurofibromatosis: the role of guanosine triphosphatase activating proteins in sensory neuron function. Sheng Li Xue Bao. 2008;60(5):581–3.

    PubMed  CAS  Google Scholar 

  138. Diggs-Andrews KA, Tokuda K, Izumi Y, Zorumski CF, Wozniak DF, Gutmann DH. Dopamine deficiency underlies learning deficits in neurofibromatosis-1 mice. Ann Neurol. 2013;73(2):309–15.

    Article  PubMed  CAS  Google Scholar 

  139. Ryu HH, Kang M, Park J, Park SH, Lee YS. Enriched expression of NF1 in inhibitory neurons in both mouse and human brain. Mol Brain. 2019;12(1):60.

    Article  PubMed  PubMed Central  Google Scholar 

  140. Friedman JM, Riccardi VM. Neurofibromatosis : phenotype, natural history, and pathogenesis. 3rd ed. Baltimore: Johns Hopkins University Press Baltimore; 1999.

    Google Scholar 

  141. Souza M, Jansen A, Martins A, Rodrigues L, Rezende N. Body composition in adults with neurofibromatosis type 1. Rev Assoc Med Bras (1992). 2016;62(9):831–6.

    Article  PubMed  Google Scholar 

  142. Koga M, Yoshida Y, Imafuku S. Nutritional, muscular and metabolic characteristics in patients with neurofibromatosis type 1. J Dermatol. 2016;43(7):799–803.

    Article  PubMed  CAS  Google Scholar 

  143. Ferrara UP, Tortora C, Rosano C, Assunto A, Rossi A, Pagano S, et al. Bone metabolism in patients with type 1 neurofibromatosis: key role of sun exposure and physical activity. Sci Rep. 2022;12(1):4368.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  144. Soucy EA, van Oppen D, Nejedly NL, Gao F, Gutmann DH, Hollander AS. Height assessments in children with neurofibromatosis type 1. J Child Neurol. 2013;28(3):303–7.

    Article  PubMed  Google Scholar 

  145. Souza JF, Passos RL, Guedes AC, Rezende NA, Rodrigues LO. Muscular force is reduced in neurofibromatosis type 1. J Musculoskelet Neuronal Interact. 2009;9(1):15–7.

    PubMed  CAS  Google Scholar 

  146. Johnson BA, MacWilliams BA, Carey JC, Viskochil DH, D’Astous JL, Stevenson DA. Motor proficiency in children with neurofibromatosis type 1. Pediatr Phys Ther. 2010;22(4):344–8.

    Article  PubMed  PubMed Central  Google Scholar 

  147. Martins AS, Jansen AK, Rodrigues LO, Matos CM, Souza ML, de Souza JF, et al. Lower fasting blood glucose in neurofibromatosis type 1. Endocr Connect. 2016;5(1):28–33.

    Article  PubMed  CAS  Google Scholar 

  148. Martins AS, Jansen AK, Rodrigues LOC, Matos CM, Souza MLR, Miranda DM, et al. Increased insulin sensitivity in individuals with neurofibromatosis type 1. Arch Endocrinol Metab. 2018;62(1):41–6.

    Article  PubMed  PubMed Central  Google Scholar 

  149. Ozhan B, Ozguven AA, Ersoy B. Neurofibromatosis type 1 and diabetes mellitus: an unusual association. Case Rep Endocrinol. 2013;2013:689107.

    PubMed  PubMed Central  Google Scholar 

  150. Kallionpää RA, Peltonen S, Leppävirta J, Pöyhönen M, Auranen K, Järveläinen H, et al. Haploinsufficiency of the NF1 gene is associated with protection against diabetes. J Med Genet. 2021;58(6):378–84.

    Article  PubMed  Google Scholar 

  151. Vassallo G, Mughal Z, Robinson L, Weisberg D, Roberts SA, Hupton E, et al. Perceived fatigue in children and young adults with neurofibromatosis type 1. J Paediatr Child Health. 2020;56(6):878–83.

    Article  PubMed  Google Scholar 

  152. Gehart H, Kumpf S, Ittner A, Ricci R. MAPK signalling in cellular metabolism: stress or wellness? EMBO Rep. 2010;11(11):834–40.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  153. Guo YJ, Pan WW, Liu SB, Shen ZF, Xu Y, Hu LL. ERK/MAPK signalling pathway and tumorigenesis (Review). Exp Ther Med. 2020;19(3):1997–2007.

    PubMed  PubMed Central  Google Scholar 

  154. Masgras I, Ciscato F, Brunati AM, Tibaldi E, Indraccolo S, Curtarello M, et al. Absence of Neurofibromin Induces an Oncogenic Metabolic Switch via Mitochondrial ERK-Mediated Phosphorylation of the Chaperone TRAP1. Cell Rep. 2017;18(3):659–72.

    Article  PubMed  CAS  Google Scholar 

  155. Tritz R, Benson T, Harris V, Hudson FZ, Mintz J, Zhang H, et al. Nf1 heterozygous mice recapitulate the anthropometric and metabolic features of human neurofibromatosis type 1. Transl Res. 2021;228:52–63.

    Article  PubMed  CAS  Google Scholar 

  156. Sullivan K, El-Hoss J, Quinlan KG, Deo N, Garton F, Seto JT, et al. NF1 is a critical regulator of muscle development and metabolism. Hum Mol Genet. 2014;23(5):1250–9.

    Article  PubMed  CAS  Google Scholar 

  157. Brannan CI, Perkins AS, Vogel KS, Ratner N, Nordlund ML, Reid SW, et al. Targeted disruption of the neurofibromatosis type-1 gene leads to developmental abnormalities in heart and various neural crest-derived tissues. Genes Dev. 1994;8(9):1019–29.

    Article  PubMed  CAS  Google Scholar 

  158. Summers MA, Rupasinghe T, Vasiljevski ER, Evesson FJ, Mikulec K, Peacock L, et al. Dietary intervention rescues myopathy associated with neurofibromatosis type 1. Hum Mol Genet. 2018;27(4):577–88.

    Article  PubMed  CAS  Google Scholar 

  159. Summers MA, Vasiljevski ER, Mikulec K, Peacock L, Little DG, Schindeler A. Developmental dosing with a MEK inhibitor (PD0325901) rescues myopathic features of the muscle-specific but not limb-specific Nf1 knockout mouse. Mol Genet Metab. 2018;123(4):518–25.

    Article  PubMed  CAS  Google Scholar 

  160. Wei X, Franke J, Ost M, Wardelmann K, Borno S, Timmermann B, et al. Cell autonomous requirement of neurofibromin (Nf1) for postnatal muscle hypertrophic growth and metabolic homeostasis. J Cachexia Sarcopenia Muscle. 2020;11(6):1758–78.

    Article  PubMed  PubMed Central  Google Scholar 

  161. Walker JA, Tchoudakova AV, McKenney PT, Brill S, Wu D, Cowley GS, et al. Reduced growth of Drosophila neurofibromatosis 1 mutants reflects a non-cell-autonomous requirement for GTPase-Activating Protein activity in larval neurons. Genes Dev. 2006;20(23):3311–23.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  162. Hannan F, Ho I, Tong JJ, Zhu Y, Nurnberg P, Zhong Y. Effect of neurofibromatosis type I mutations on a novel pathway for adenylyl cyclase activation requiring neurofibromin and Ras. Hum Mol Genet. 2006;15(7):1087–98.

    Article  PubMed  CAS  Google Scholar 

  163. Tong JJ, Schriner SE, McCleary D, Day BJ, Wallace DC. Life extension through neurofibromin mitochondrial regulation and antioxidant therapy for neurofibromatosis-1 in Drosophila melanogaster. Nat Genet. 2007;39(4):476–85.

    Article  PubMed  CAS  Google Scholar 

  164. Botero V, Stanhope BA, Brown EB, Grenci EC, Boto T, Park SJ, et al. Neurofibromin regulates metabolic rate via neuronal mechanisms in Drosophila. Nat Commun. 2021;12(1):4285.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  165. Maurer GW, Malita A, Nagy S, Koyama T, Werge TM, Halberg KA, et al. Analysis of genes within the schizophrenia-linked 22q11.2 deletion identifies interaction of night owl/LZTR1 and NF1 in GABAergic sleep control. PLoS genetics. 2020;16(4):e1008727.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  166. DeBella K, Szudek J, Friedman JM. Use of the National Institutes of Health Criteria for Diagnosis of Neurofibromatosis 1 in Children. Pediatrics. 2000;105(3):608–14.

    Article  PubMed  CAS  Google Scholar 

  167. De Schepper S, Maertens O, Callens T, Naeyaert J-M, Lambert J, Messiaen L. Somatic Mutation Analysis in NF1 Cafe au lait Spots Reveals Two NF1 Hits in the Melanocytes. J Investig Dermatol. 2008;128(4):1050–3.

    Article  PubMed  Google Scholar 

  168. Lubs MLE, Bauer MS, Formas ME, Djokic B. Lisch Nodules in Neurofibromatosis Type 1. N Engl J Med. 1991;324(18):1264–6.

    Article  PubMed  CAS  Google Scholar 

  169. Stevenson DA, Birch PH, Friedman JM, Viskochil DH, Balestrazzi P, Boni S, et al. Descriptive analysis of tibial pseudarthrosis in patients with neurofibromatosis 1. Am J Med Genet. 1999;84(5):413–9.

    Article  PubMed  CAS  Google Scholar 

  170. Kjell VR, Hilde B, Eric L, Johan L, Armand L. Prevalence of neurofibromatosis type 1 in congenital pseudarthrosis of the tibia. Eur J Pediatr. 2016;175(9):1193–8.

    Article  Google Scholar 

  171. Jackson IT, Carbonnel A, Potparic Z, Shaw K. Orbitotemporal neurofibromatosis: classification and treatment. Plast Reconstr Surg. 1993;92(1):1–11.

    Article  PubMed  CAS  Google Scholar 

  172. Brunetti-Pierri N, Doty SB, Hicks J, Phan K, Mendoza-Londono R, Blazo M, et al. Generalized metabolic bone disease in Neurofibromatosis type I. Mol Genet Metab. 2008;94(1):105–11.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  173. Crawford AH, Herrera-Soto J. Scoliosis Associated with Neurofibromatosis. Orthop Clin North Am. 2007;38(4):553–62.

    Article  PubMed  Google Scholar 

  174. Szudek J, Birch P, Friedman JM. Growth in North American white children with neurofibromatosis 1 (NF1). J Med Genet. 2000;37(12):933–8.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  175. Zessis NR, Gao F, Vadlamudi G, Gutmann DH, Hollander AS. Height growth impairment in children with neurofibromatosis type 1 is characterized by decreased pubertal growth velocity in both sexes. J Child Neurol. 2018;33(12):762–6.

    Article  PubMed  Google Scholar 

  176. Stevenson DA, Yan J, He Y, Li H, Liu Y, Zhang Q, et al. Multiple increased osteoclast functions in individuals with neurofibromatosis type 1. Am J Med Genet A. 2011;155a(5):1050–9.

    Article  PubMed  Google Scholar 

  177. Caffarelli C, Gonnelli S, Tanzilli L, Vivarelli R, Tamburello S, Balestri P, et al. Quantitative ultrasound and dual-energy x-ray absorptiometry in children and adolescents with neurofibromatosis of type 1. J Clin Densitom. 2010;13(1):77–83.

    Article  PubMed  Google Scholar 

  178. Lammert M, Kappler M, Mautner VF, Lammert K, Störkel S, Friedman JM, et al. Decreased bone mineral density in patients with neurofibromatosis 1. Osteoporos Int. 2005;16(9):1161–6.

    Article  PubMed  Google Scholar 

  179. Kolanczyk M, Kossler N, Kühnisch J, Lavitas L, Stricker S, Wilkening U, et al. Multiple roles for neurofibromin in skeletal development and growth. Hum Mol Genet. 2007;16(8):874–86.

    Article  PubMed  CAS  Google Scholar 

  180. Paria N, Khalid A, Shen B, Lemoine B, Chan J, Kidane YH, et al. Molecular dissection of somatic skeletal disease in neurofibromatosis type 1. J Bone Miner Res. 2022;38(2):288–99.

    Article  PubMed  Google Scholar 

  181. Wang W, Nyman JS, Ono K, Stevenson DA, Yang X, Elefteriou F. Mice lacking Nf1 in osteochondroprogenitor cells display skeletal dysplasia similar to patients with neurofibromatosis type I. Hum Mol Genet. 2011;20(20):3910–24.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  182. Uthoff J, Larson J, Sato TS, Hammond E, Schroeder KE, Rohret F, et al. Longitudinal phenotype development in a minipig model of neurofibromatosis type 1. Sci Rep. 2020;10(1):5046.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  183. Wang S, Mautner VF, Buchert R, Flibotte S, Suppa P, Friedman JM, et al. Alterations in brain morphology by MRI in adults with neurofibromatosis 1. Orphanet J Rare Dis. 2021;16(1):462.

    Article  PubMed  PubMed Central  Google Scholar 

  184. Cutting LE, Cooper KL, Koth CW, Mostofsky SH, Kates WR, Denckla MB, et al. Megalencephaly in NF1: predominantly white matter contribution and mitigation by ADHD. Neurology. 2002;59(9):1388–94.

    Article  PubMed  CAS  Google Scholar 

  185. Greenwood RS, Tupler LA, Whitt JK, Buu A, Dombeck CB, Harp AG, et al. Brain morphometry, T2-weighted hyperintensities, and IQ in children with neurofibromatosis type 1. Arch Neurol. 2005;62(12):1904–8.

    Article  PubMed  Google Scholar 

  186. Moore BD 3rd, Slopis JM, Jackson EF, De Winter AE, Leeds NE. Brain volume in children with neurofibromatosis type 1: relation to neuropsychological status. Neurology. 2000;54(4):914–20.

    Article  PubMed  Google Scholar 

  187. Steen RG, Taylor JS, Langston JW, Glass JO, Brewer VR, Reddick WE, et al. Prospective evaluation of the brain in asymptomatic children with neurofibromatosis type 1: relationship of macrocephaly to T1 relaxation changes and structural brain abnormalities. AJNR Am J Neuroradiol. 2001;22(5):810–7.

    PubMed  PubMed Central  CAS  Google Scholar 

  188. Karlsgodt KH, Rosser T, Lutkenhoff ES, Cannon TD, Silva A, Bearden CE. Alterations in white matter microstructure in neurofibromatosis-1. PLoS One. 2012;7(10):e47854.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  189. Said SM, Yeh TL, Greenwood RS, Whitt JK, Tupler LA, Krishnan KR. MRI morphometric analysis and neuropsychological function in patients with neurofibromatosis. NeuroReport. 1996;7(12):1941–4.

    Article  PubMed  CAS  Google Scholar 

  190. Dubovsky EC, Booth TN, Vezina G, Samango-Sprouse CA, Palmer KM, Brasseux CO. MR imaging of the corpus callosum in pediatric patients with neurofibromatosis type 1. AJNR Am J Neuroradiol. 2001;22(1):190–5.

    PubMed  PubMed Central  CAS  Google Scholar 

  191. Wignall EL, Griffiths PD, Papadakis NG, Wilkinson ID, Wallis LI, Bandmann O, et al. Corpus callosum morphology and microstructure assessed using structural MR imaging and diffusion tensor imaging: initial findings in adults with neurofibromatosis type 1. AJNR Am J Neuroradiol. 2010;31(5):856–61.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  192. DiMario FJ Jr, Ramsby G, Greenstein R, Langshur S, Dunham B. Neurofibromatosis type 1: magnetic resonance imaging findings. J Child Neurol. 1993;8(1):32–9.

    Article  PubMed  Google Scholar 

  193. Ji J, Shimony J, Gao F, McKinstry RC, Gutmann DH. Optic nerve tortuosity in children with neurofibromatosis type 1. Pediatr Radiol. 2013;43(10):1336–43.

    Article  PubMed  PubMed Central  Google Scholar 

  194. Wang S, Friedman JM, Suppa P, Buchert R, Mautner VF. White matter is increased in the brains of adults with neurofibromatosis 1. Orphanet J Rare Dis. 2022;17(1):115.

    Article  PubMed  PubMed Central  Google Scholar 

  195. Kayl AE, Moore BD 3rd, Slopis JM, Jackson EF, Leeds NE. Quantitative morphology of the corpus callosum in children with neurofibromatosis and attention-deficit hyperactivity disorder. J Child Neurol. 2000;15(2):90–6.

    Article  PubMed  CAS  Google Scholar 

  196. Aydin S, Kurtcan S, Alkan A, Guler S, Filiz M, Yilmaz TF, et al. Relationship between the corpus callosum and neurocognitive disabilities in children with NF-1: diffusion tensor imaging features. Clin Imaging. 2016;40(6):1092–5.

    Article  PubMed  Google Scholar 

  197. Shofty B, Bergmann E, Zur G, Asleh J, Bosak N, Kavushansky A, et al. Autism-associated Nf1 deficiency disrupts corticocortical and corticostriatal functional connectivity in human and mouse. Neurobiol Dis. 2019;130:104479.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  198. Tomson SN, Schreiner MJ, Narayan M, Rosser T, Enrique N, Silva AJ, et al. Resting state functional MRI reveals abnormal network connectivity in neurofibromatosis 1. Hum Brain Mapp. 2015;36(11):4566–81.

    Article  PubMed  PubMed Central  Google Scholar 

  199. Loitfelder M, Huijbregts SC, Veer IM, Swaab HS, Van Buchem MA, Schmidt R, et al. Functional Connectivity Changes and Executive and Social Problems in Neurofibromatosis Type I. Brain Connect. 2015;5(5):312–20.

    Article  PubMed  PubMed Central  Google Scholar 

  200. Baudou E, Nemmi F, Peran P, Cignetti F, Blais M, Maziero S, et al. Atypical connectivity in the cortico-striatal network in NF1 children and its relationship with procedural perceptual-motor learning and motor skills. J Neurodev Disord. 2022;14(1):15.

    Article  PubMed  PubMed Central  Google Scholar 

  201. Ibrahim AFA, Montojo CA, Haut KM, Karlsgodt KH, Hansen L, Congdon E, et al. Spatial working memory in neurofibromatosis 1: Altered neural activity and functional connectivity. Neuroimage Clin. 2017;15:801–11.

    Article  PubMed  PubMed Central  Google Scholar 

  202. Baudou E, Nemmi F, Biotteau M, Maziero S, Peran P, Chaix Y. Can the Cognitive Phenotype in Neurofibromatosis Type 1 (NF1) Be Explained by Neuroimaging? A Review Front Neurol. 2019;10:1373.

    Article  PubMed  Google Scholar 

  203. Mennigen E, Schuette P, Vajdi A, Pacheco L, Rosser T, Bearden CE. Reduced higher dimensional temporal dynamism in neurofibromatosis type 1. Neuroimage Clin. 2019;22:101692.

    Article  PubMed  PubMed Central  Google Scholar 

  204. Chabernaud C, Mennes M, Kardel PG, Gaillard WD, Kalbfleisch ML, Vanmeter JW, et al. Lovastatin regulates brain spontaneous low-frequency brain activity in neurofibromatosis type 1. Neurosci Lett. 2012;515(1):28–33.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  205. Nemmi F, Cignetti F, Assaiante C, Maziero S, Audic F, Peran P, et al. Discriminating between neurofibromatosis-1 and typically developing children by means of multimodal MRI and multivariate analyses. Hum Brain Mapp. 2019;40(12):3508–21.

    Article  PubMed  PubMed Central  Google Scholar 

  206. Wang Y, Kim E, Wang X, Novitch Bennett G, Yoshikawa K, Chang L-S, et al. ERK Inhibition Rescues Defects in Fate Specification of Nf1-Deficient Neural Progenitors and Brain Abnormalities. Cell. 2012;150(4):816–30.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  207. Lakkis MM, Golden JA, O’Shea KS, Epstein JA. Neurofibromin deficiency in mice causes Exencephaly and is a modifier for splotch neural tube defects. Dev Biol. 1999;212(1):80–92.

    Article  PubMed  CAS  Google Scholar 

  208. Gutmann DH, Loehr A, Zhang Y, Kim J, Henkemeyer M, Cashen A. Haploinsufficiency for the neurofibromatosis 1 (NF1) tumor suppressor results in increased astrocyte proliferation. Oncogene. 1999;18(31):4450–9.

    Article  PubMed  CAS  Google Scholar 

  209. Gutmann DH, Wu YL, Hedrick NM, Zhu Y, Guha A, Parada LF. Heterozygosity for the neurofibromatosis 1 (NF1) tumor suppressor results in abnormalities in cell attachment, spreading and motility in astrocytes. Hum Mol Genet. 2001;10(26):3009–16.

    Article  PubMed  CAS  Google Scholar 

  210. Zhu Y, Romero MI, Ghosh P, Ye Z, Charnay P, Rushing EJ, et al. Ablation of NF1 function in neurons induces abnormal development of cerebral cortex and reactive gliosis in the brain. Genes Dev. 2001;15(7):859–76.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  211. Zhu Y, Harada T, Liu L, Lush ME, Guignard F, Harada C, et al. Inactivation of NF1 in CNS causes increased glial progenitor proliferation and optic glioma formation. Development. 2005;132(24):5577–88.

    Article  PubMed  CAS  Google Scholar 

  212. Hegedus B, Dasgupta B, Shin JE, Emnett RJ, Hart-Mahon EK, Elghazi L, et al. Neurofibromatosis-1 Regulates Neuronal and Glial Cell Differentiation from Neuroglial Progenitors In Vivo by Both cAMP- and Ras-Dependent Mechanisms. Cell Stem Cell. 2007;1(4):443–57.

    Article  PubMed  CAS  Google Scholar 

  213. Hegedus B, Yeh TH, Lee DY, Emnett RJ, Li J, Gutmann DH. Neurofibromin regulates somatic growth through the hypothalamic-pituitary axis. Hum Mol Genet. 2008;17(19):2956–66.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

Download references

Acknowledgements

We thank Drs. Damon Page, Ronald L. Davis, Baoji Xu, and David Gutmann for advice and feedback during Dr. Botero’s dissertation research, and Lisa Ringen, Linda Buckner, Melissa Benilous, and Katherine O’Brien for administrative support.

Funding

Our research is supported by NIH/NINDS R01 NS097237, R01 NS126361, R01 NS114403, R01 NS124716, and R21 NS124198. V.B. was supported by NIH/NINDS F31 NS124245.

Author information

Authors and Affiliations

Authors

Contributions

V.B. and S.M.T. conceived, wrote, and edited the manuscript. The authors read and approved the final manuscript.

Corresponding author

Correspondence to Seth M. Tomchik.

Ethics declarations

Ethics approval and consent to participate

Not applicable.

Consent of publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

Additional information

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Botero, V., Tomchik, S.M. Unraveling neuronal and metabolic alterations in neurofibromatosis type 1. J Neurodevelop Disord 16, 49 (2024). https://doi.org/10.1186/s11689-024-09565-6

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s11689-024-09565-6

Keywords