Skip to main content

From neurodevelopment to neurodegeneration: the interaction of neurofibromin and valosin-containing protein/p97 in regulation of dendritic spine formation

Abstract

Both Neurofibromatosis type I (NF1) and inclusion body myopathy with Paget's disease of bone and frontotemporal dementia (IBMPFD) are autosomal dominant genetic disorders. These two diseases are fully penetrant but with high heterogeneity in phenotypes, suggesting the involvement of genetic modifiers in modulating patients' phenotypes. Although NF1 is recognized as a developmental disorder and IBMPFD is associated with degeneration of multiple tissues, a recent study discovered the direct protein interaction between neurofibromin, the protein product of the NF1 gene, and VCP/p97, encoded by the causative gene of IBMPFD. Both NF1 and VCP/p97 are critical for dendritic spine formation, which provides the cellular mechanism explaining the cognitive deficits and dementia found in patients. Moreover, disruption of the interaction between neurofibromin and VCP impairs dendritic spinogenesis. Neurofibromin likely influences multiple downstream pathways to control dendritic spinogenesis. One is to activate the protein kinase A pathway to initiate dendritic spine formation; another is to regulate the synaptic distribution of VCP and control the activity of VCP in dendritic spinogenesis. Since neurofibromin and VCP/p97 also regulate cell growth and bone metabolism, the understanding of neurofibromin and VCP/p97 in neurons may be applied to study of cancer and bone. Statin treatment rescues the spine defects caused by VCP deficiency, suggesting the potential role of statin in clinical treatment for these two diseases.

Review

Neurodevelopmental disorders and neurodegeneration

Neurons are highly differentiated cells composed of several specialized subcellular structures, including soma, dendrites, axon, and numerous synapses. All of these structures play specific roles in signal transduction between or within neurons. Besides dendrites and axons, dendritic spines are particularly interesting structures, which are the tiny protrusions (~0.5-1 μm in width and ~1-2 μm in length) extending from dendrites. The majority of excitatory synapses in the mammalian nervous system are localized at the tips of dendritic spines [1]. Morphology and density of dendritic spines are controlled by genetic program, neuronal activity, and environmental insults. Indeed, defects of neural development leading to alterations of neuronal morphology are associated with many neurological and neuropsychiatric disorders, including mental retardation, learning disability, autism, attention-deficient hyperactivity disorder, and schizophrenia [2, 3]. On the other hand, impairment in maintenance of neuronal morphology is also frequently found in neurodegenerative disorders. For instance, synaptic loss has been suggested as a cause of dementia [4–6]. This review will focus on an example showing that neurodegeneration may be relevant with neurodevelopment in terms of regulation of neuronal morphology. It is regarding a recent finding of the interaction between neurofibromin and valosin-containing protein (VCP).

Neurofibromin, the protein product of the neurofibromatosis type I (NF1) gene, regulates the formation of dendritic spines [7], which explains at least partially why patients with NF1 suffer from cognitive defects. Recently, we further identified that VCP/p97 interacts with neurofibromin and demonstrated that VCP/p97 also controls dendritic spinogenesis of neurons [8]. Mutations in the VCP gene result in inclusion body myopathy with Paget's disease of bone and frontotemporal dementia (IBMPFD) and amyotrophic lateral sclerosis (ALS). The function of VCP in spinogenesis provides a potential explanation how VCP mutations result in dementia in patients. Although both neurofibromin and VCP/p97 are ubiquitously expressed in many tissues and are also involved in tumorigenesis [9–11] and bone metabolism [12, 13], the current review focuses on the molecular functions of neurofibromin and VCP/p97 in neurons.

Neurofibromatosis type I (NF1)

Neurofibromatosis type I (NF1, MIM:162200) is one of the most common autosomal dominant disorders affecting about one in 3,500 individuals. NF1 is characterized by skin pigmentations (café-au-lait spots and freckling), Lisch nodules in the iris, and formations of benign peripheral nerve sheath tumors (neurofibromas and schwannomas). In addition, many other features are frequently found in patients with NF1, including neurobehavioral developmental disorders as well as skeletal lesions and malformations. Children with NF1 are frequently associated with learning difficulty [14–16] and are more susceptible to autism and attention-deficient hyperactivity disorder [15–18]. Additionally, scoliosis, kyphosis, short stature, and tibial bowing are common skeleton manifestations occurring in NF1 patients [19]. Based on the symptoms identified in patients with NF1, it is clear that the NF1 gene plays important roles in cell growth (tumor formation), pigmentation, neuronal activity and function, and bone metabolism. Although NF1 is fully penetrant, the phenotypes of NF1 patients are extremely variable, even among patients in the same family. It is unclear why clinical features of NF1 patients vary so markedly. Genetic modifiers have been suggested to influence the impact of NF1 deficits [20, 21].

The function of neurofibromin in signaling

The NF1 gene encodes a large protein of 2818 amino acid residues, named neurofibromin (Figure 1), with a well-known RasGAP-related domain (GRD, ~350 amino acid residues) at the central region of the protein [22]. The RasGAP activity is believed to account for the tumor suppression activity of neurofibromin [23–27], which is the most well known function of neurofibromin. In addition to downregulating the Ras pathway, neurofibromin increases adenylyl cyclase activity and thus upregulates the cAMP pathway through both Gαs-dependent and Gαs-independent pathways [28, 29]. More details about the role of neurofibromin in signaling are available in the previous review [30].

Figure 1
figure 1

The interacting proteins and corresponding functions of neurofibromin. GRD, RasGAP-related domain; LRD, Leucine-rich domain; CTD, C-terminal domain. The amino acid (aa) residue number is based on the sequence of neurofibromin with type I GRD. The LRD can be further separated into the N-terminal Sec14-like domain (residue 1560-1699) for lipid binding and the C-terminal pleckstrin homology-like domain (residues 1713-1818). The braces indicate the interacting regions for each binding proteins. 1Note that the GRD and entire C-terminal region of neurofibromin are involved in activation of the adenylate cyclase. However, the mechanism is unknown. It is not clear whether the direct protein-protein interaction between neurofibromin and adenylate cyclase is involved.

The obstacles of NF1 study

The NF1 gene was identified more than 20 years ago. However, several obstacles impede the progress of NF1 study. First of all, perhaps due to cytotoxic effects, it is not easy to express full length neurofibromin in the cell. Therefore it is difficult to perform genetic manipulation to study the molecular function of neurofibromin. Secondly, mutational hot spots are not present in the NF1 gene. The mutations identified from patients are dispersed throughout the entire NF1 gene. It is therefore difficult to figure out the pathogenic mechanism of NF1. Finally, with the exception of the GRD, neurofibromin lacks a recognizable enzymatic protein domain. Therefore, numerous efforts have been directed to identify neurofibromin-interacting proteins (Figure 1) [7, 8, 28, 31–41]. Identification and characterization of proteins that interact with neurofibromin provide a better understanding of the molecular functions of neurofibromin.

Neurofibromin regulates neuronal morphogenesis

The fact that patients with NF1 are frequently associated with multiple neurobehavioral developmental disorders suggests an important role of neurofibromin in brain development and function. Indeed, several studies have revealed the role of neurofibromin in neural development. Through its C-terminal domain, neurofibromin interacts with collapsing response mediator proteins, CRMP-2 and CRMP-4 [35, 39]. This interaction regulates neurite outgrowth of neurons [39]. In addition to neurite outgrowth, our previous study showed that neurofibromin acts downstream of syndecan-2, a synaptic heparan sulfate proteoglycan, in the regulation of dendritic spine formation [7]. Neurofibromin interacts with syndecan-2 [34] and then activates the PKA-Ena/VASP pathway through the cAMP pathway to promote actin polymerization and bundle formation [7], thus promoting the formation of dendritic filopodia, the precursors of dendritic spines. The role of neurofibromin in dendritic spinogenesis explains at least partially why patients with NF1 are often associated with cognitive abnormalities. Moreover, although the PKA pathway is essential for dendritic filopodia and spine formation, activation of PKA alone is not sufficient for the process [7], possibly due to the involvement of multiple downstream pathways of neurofibromin in spinogenesis. Indeed, from a series of proteomic and biochemical studies, we identified that neurofibromin uses its leucine-rich domain (LRD) to directly interact with the valosin-containing protein (VCP) [8]. Knockdown of endogenous VCP reduces the density of dendritic spines, but does not noticeably influence their width or length. Interruption of the interaction between neurofibromin and VCP also reduces the spine density [8]. The studies suggest that neurofibromin regulates dendritic spine formation through at least two pathways: one cAMP-dependent and the other VCP-dependent.

Valosin-containing protein (VCP) and degeneration disorders

IBMPFD MIM:167320, [42] is a dominant inherited disorder with three major symptoms: myopathy, osteolytic bone lesion, and frontotemporal dementia (FTD). Similar to NF1, IBMPFD is also fully penetrant, but expressivity is variable, suggesting the involvement of genetic modifiers in the influence of phenotype [43, 44]. Around 80-90% and 50% of patients develop inclusion body myopathy and osteolytic lesions, respectively, with a mean age of 42 years. Only about 30-35% of patients had early-onset FTD typically presenting at age 53 years [42–44]. In addition to IBMPFD, recent human genetic analysis indicated that VCP mutations account for 1-2% of autosomal dominantly inherited amyotrophic lateral sclerosis (ALS) [45]. VCP interacts with the polyglutamine-containing aggregates that are found in patients with Huntington's and Machado-Joseph diseases [46]. VCP also associates with ubiquitinated protein aggregates and is involved in TDP-43-related frontotemporal dementia [47–49].

Molecular functions of VCP

VCP, a multifunctional AAA (A TPases A ssociated with a variety of cellular A ctivities) protein, forms a homohexameric barrel and hydrolyses ATP to generate the mechanical force for its function as a molecular chaperon [50–52]. It possesses two ATPase domains (D1 and D2). The D2 domain carries the major ATPase activity, while the D1 domain is also the hexamerization domain of VCP. The N-terminal region (N-domain) of VCP is involved in its interaction with various adaptors. The identified IBMPFD mutations are highly clustered in the N- and D1 domains of VCP [42, 53–55]. Therefore, IBMPFD mutations can change the conformation of the catalytic domains, alter the ATPase activity, and compromise the function of VCP [56]. Through the N-terminus interacting adaptors, VCP regulates a variety of cellular events, including cell-cycle control, membrane fusion, ubiquitin-dependent protein degradation, endoplasmic reticulum-associated protein degradation (ERAD), and autophagy [51, 57–64].

So far, it is not clear how mutations in the VCP gene lead to IBMPFD or other degeneration diseases. An IBMPFD-associated VCP mutant was shown to induce aggregation of polyubiquitin-conjugated proteins in myoblastoma cells [65]. TDP-43-positive aggregates are also found in the muscles of patients with IBMPFD [48]. VCP mutations have also been shown to cause autophagy dysfunction, which may additionally contribute to the pathogenesis of IBMPFD [62, 63, 66]. Therefore, dysregulation of protein degradation has been proposed to play a role in IBMPFD and VCP-related disorders.

Although the formation of VCP-positive protein aggregates induced by IBMPFD mutation have been demonstrated in muscle, the significance of protein aggregation in neurons is not so clear. Expression of IBMPFD mutant VCP in cultured neurons reduces the density of dendritic spines, but does not induce VCP aggregation [8]. It suggests that VCP aggregation is not the cause of synaptic defects induced by IBMPFD mutations. However, it does not suggest the irrelevance of dysregulation of protein degradation in IBMPFD. Perhaps after expression for six days in cultured neurons, protein degradation (through either proteasomal degradation or autophagy) is impaired and thus spine density is reduced, although the undegraded proteins have not yet formed aggregates. The involvement of ERAD, proteasomal degradation, and/or autophagy in VCP-dependent dendritic spine formation should be investigated directly in the future.

The previous studies had demonstrated that VCP forms complexes with cofactor p47 and regulates multiple membrane fusion events in cells [67, 68], including nuclear envelope assembly [69], ER/Golgi membrane assembly [70, 71], ER morphogenesis [72, 73], and membrane fusion of autophagy [74]. Because p47 also coexists with VCP in the immunoprecipitated complex containing neurofibromin [8], it is likely that membrane fusion mediated by VCP also contributes to dendritic spine formation. More investigations have to been performed to address the possible role of VCP-mediated membrane fusion in dendritic spine formation.

VCP and neuronal morphogenesis

Although the molecular mechanism underlying IBMPFD pathogenesis is still unclear, the accumulated evidence indicates that VCP plays critical roles in neuronal morphogenesis, which may account for the dementia phenotype associated with IBMPFD patients. Firstly, dystrophic neurites are frequently found in patients with FTD [55, 75, 76]. Moreover, VCP also regulates remodeling of dendritic arbor in Drosophila [77]. We also found that VCP is critical for the density of dendritic spines in cultured neurons. Expression of IBMPFD mutant VCP reduces the density of dendritic spines in cultured rat hippocampal neurons [8]. It is very possible that VCP actively contributes to neuronal morphogenesis and thus dysfunction of VCP may result in neurodegeneration.

The interaction between neurofibromin and VCP

Using a variety of proteomic and biochemical methods, we identified a direct interaction between neurofibromin and VCP. The LRD of neurofibromin and the D1D2 region of VCP are required for the interaction [8]. We further showed that VCP acts downstream of neurofibromin in modulating dendritic spine formation. Compared with wild-type (WT) neurons, Nf1+/- neurons have a lower density of dendritic spines [8]. Overexpression of WT VCP, but not the IBMPFD mutant, rescues the defects of spine density caused by Nf1 haploinsufficiency [8]. Moreover, the synaptic distribution of VCP is reduced in Nf1+/- brains [8]. These results suggest that neurofibromin targets VCP to synapses and directs the function of VCP in spinogenesis.

To further elucidate the role of the neurofibromin-VCP interaction in NF1 and IBMPFD, the influence of mutations in the genes identified in patients has been investigated. The neurofibromin mutant (Y1587delta) lacking the Y1587 residue in the LRD has a much weaker interaction with VCP [8]. Patient carrying the Y1587delta mutation presents clinically with short stature and dementia [8], which echo the functions of VCP in dementia and bone metabolism. Moreover, unlike full length WT neurofibromin, expression of full length Y1587delta mutant does not rescue spine deficiency caused by Nf1 haploinsufficiency [8]. On the other hand, the VCP R155H and R95G mutations, the most common mutations in VCP, also reduce the interaction between VCP and neurofibromin [8]. In contrast to WT VCP, expression of these VCP mutants do not rescue the dendritic spine defects in Nf1+/- neurons [8]. Taken together, these data show that reduction of the interaction between neurofibromin and VCP occurs in both NF1 and IBMPFD patients.

Genetic modifications of NF1 and IBMPFD

Since the phenotypes of both NF1 and IBMPFD vary markedly, genetic modifications have been suggested to influence the function of neurofibromin and VCP. The interacting proteins of neurofibromin and VCP are possible genetic modifier candidates. Indeed, VCP interacts with different cofactors and regulates various cellular processes [43, 59, 64]. The variant expression levels of cofactors or other interacting proteins of VCP are likely to influence the functions of VCP. In addition to Ras and VCP, neurofibromin has been shown to also interact with syndecans [30, 34], 14-3-3 [38], CRMP2/4 [35, 39], and APP [37] (Figure 1). Our recent study indicated that overexpression of VCP rescues the defects of dendritic spines in cultured Nf1+/- cortical neurons [8]. This suggests that variation of VCP protein levels affects the phenotypes caused by Nf1 haploinsufficiency. It is important to further explore this possibility in NF1 patients. Examination of the VCP expression levels and their correlation with the phenotypes of NF1 patients will provide clues whether the VCP expression levels influence the phenotypes of patients with NF1.

The potential clinical effect of statin

Statin, an inhibitor of 3-hydroxy-3-methylglutaryl-coenzyme A (HMG-CoA) reductase, has been shown to rescue the learning and attention deficits of Nf1 mutant mice [78]. HMG-CoA reductase is the rate-limiting enzyme of the mevalonate pathway, which produces cholesterol and other isoprenoids. The role of statin in NF1 was examined because of the essential role of lipid modification in Ras activity, which is upregulated in NF1 mutant cells [79, 80]. Additionally, cholesterol is involved in regulation of dendritic spinogenesis [81]. Thus, control of lipid metabolism may be critical for cognitive function. Recent studies indicated that VCP is required for downregulation of the protein levels of HMG-CoA reductase [82–84]. HMG-CoA reductase expression is tightly controlled by the concentration of sterol in cells, which influences both gene expression and protein stability of HMG-CoA reductase. VCP plays an essential role in sterol-induced dislocation of HMG-CoA reductase from the ER, which leads to ubiquitination and degradation of HMG-CoA reductase [82–84]. Thus, statin may correct the defects of Nf1 haploinsufficiency in both Ras- and VCP-dependent pathways. It also implies that statin may be applied to NF1 patients who have defects in the interaction between neurofibromin and VCP.

Conclusions

The heterogeneity of patients' phenotypes presents an obstacle to establishing effective and specific clinical treatments for NF1 and IBMPFD. Understanding the molecular mechanisms of the pathogenesis of NF1 and IBMPFD, which still need more investigation to elucidate the finer details, is essential for overcoming these obstacles. Identification of the interacting molecules and elucidation of the crosstalk between or among the interacting proteins are certainly important to understanding the pathogenic mechanisms. The connection of the interaction between neurofibromin and VCP and the regulation of dendritic spinogenesis has provided the first molecular evidence that VCP functions as a genetic modifier for NF1. Studies in mouse genetic models and patients now have to be performed to confirm the role of VCP as a downstream effector of neurofibromin. It will also be interesting to explore whether mutations in the NF1 gene modulate the phenotypes of IBMPFD.

Abbreviations

AAA ATPase:

ATPases Associated with a variety of cellular Activities APTase

ALS:

amyotrophic lateral sclerosis

CRMP:

collapsing response mediator protein

CTD:

C-terminal domain

Ena/VASP:

Enabled/vasodilator-stimulated phosphoprotein

ERAD:

endoplasmic reticulum-associated protein degradation

FTD:

frontotemporal dementia

GRD:

RasGAP-related domain

IBMPFD:

Inclusion body myopathy with Paget disease of the bone and frontotemporal dementia

HMG-CoA:

3-hydroxy-3-methylglutaryl-coenzyme A

LRD:

Leucine-rich domain

NF1:

Neurofibromatosis Type I

TDP-43:

TAR DNA binding protein 43

VCP:

Valosin-containing protein.

References

  1. Harris KM, Stevens JK: Dendritic spines of CA 1 pyramidal cells in the rat hippocampus: serial electron microscopy with reference to their biophysical characteristics. J Neurosci. 1989, 9: 2982-2997.

    CAS  PubMed  Google Scholar 

  2. Ehninger D, Li W, Fox K, Stryker MP, Silva AJ: Reversing neurodevelopmental disorders in adults. Neuron. 2008, 60: 950-960. 10.1016/j.neuron.2008.12.007.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  3. Ramocki MB, Zoghbi HY: Failure of neuronal homeostasis results in common neuropsychiatric phenotypes. Nature. 2008, 455: 912-918. 10.1038/nature07457.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  4. Giusti-Rodriguez P, Gao J, Graff J, Rei D, Soda T, Tsai LH: Synaptic deficits are rescued in the p25/Cdk5 model of neurodegeneration by the reduction of beta-secretase (BACE1). J Neurosci. 2011, 31: 15751-15756. 10.1523/JNEUROSCI.3588-11.2011.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  5. Ferreira ST, Klein WL: The Abeta oligomer hypothesis for synapse failure and memory loss in Alzheimer's disease. Neurobiol Learn Mem. 2011, 96: 529-543. 10.1016/j.nlm.2011.08.003.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  6. Spires-Jones T, Knafo S: Spines, plasticity, and cognition in Alzheimer's model mice. Neural Plast. 2012, 2012: 319836-

    PubMed Central  PubMed  Google Scholar 

  7. Lin Y-L, Lei Y-T, Hong C-J, Hsueh YP: Syndecan-2 induces filopodia formation via the neurofibromin-PKA-Ena/VASP pathway. J Cell Biol. 2007, 177: 829-841. 10.1083/jcb.200608121.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  8. Wang HF, Shih YT, Chen CY, Chao HW, Lee MJ, Hsueh YP: Valosin-containing protein and neurofibromin interact to regulate dendritic spine density. J Clin Invest. 2011, 121: 4820-4837. 10.1172/JCI45677.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  9. Upadhyaya M: Genetic basis of tumorigenesis in NF1 malignant peripheral nerve sheath tumors. Front Biosci. 2011, 16: 937-951. 10.2741/3727.

    Article  CAS  Google Scholar 

  10. Brossier NM, Carroll SL: Genetically engineered mouse models shed new light on the pathogenesis of neurofibromatosis type I-related neoplasms of the peripheral nervous system. Brain Res Bull. 2011, 10.1016/j.brainresbull.2011.08.005 [doi]

    Google Scholar 

  11. Haines DS: p97-containing complexes in proliferation control and cancer: emerging culprits or guilt by association?. Genes Cancer. 2010, 1: 753-763. 10.1177/1947601910381381.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  12. Ralston SH: Pathogenesis of Paget's disease of bone. Bone. 2008, 43: 819-825. 10.1016/j.bone.2008.06.015.

    Article  CAS  PubMed  Google Scholar 

  13. Schindeler A, Little DG: Recent insights into bone development, homeostasis, and repair in type 1 neurofibromatosis (NF1). Bone. 2008, 42: 616-622. 10.1016/j.bone.2007.11.006.

    Article  CAS  PubMed  Google Scholar 

  14. Costa RM, Federov NB, Kogan JH, Murphy GG, Stern J, Ohno M, Kucherlapati R, Jacks T, Silva AJ: Mechanism for the learning deficits in a mouse model of neurofibromatosis type 1. Nature. 2002, 415: 526-530. 10.1038/nature711.

    Article  CAS  PubMed  Google Scholar 

  15. Hyman SL, Shores A, North KN: The nature and frequency of cognitive deficits in children with neurofibromatosis type 1. Neurology. 2005, 65: 1037-1044. 10.1212/01.wnl.0000179303.72345.ce.

    Article  PubMed  Google Scholar 

  16. Hyman SL, Arthur Shores 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: 973-977. 10.1017/S0012162206002131.

    Article  PubMed  Google Scholar 

  17. Mbarek O, Marouillat S, Martineau J, Barthelemy C, Muh JP, Andres C: Association study of the NF1 gene and autistic disorder. Am J Med Genet. 1999, 88: 729-732. 10.1002/(SICI)1096-8628(19991215)88:6<729::AID-AJMG26>3.0.CO;2-Q.

    Article  CAS  PubMed  Google Scholar 

  18. Marui T, Hashimoto O, Nanba E, Kato C, Tochigi M, Umekage T, Ishijima M, Kohda K, Kato N, Sasaki T: Association between the neurofibromatosis-1 (NF1) locus and autism in the Japanese population. Am J Med Genet B Neuropsychiatr Genet. 2004, 131B: 43-47. 10.1002/ajmg.b.20119.

    Article  PubMed  Google Scholar 

  19. Kuorilehto T, Poyhonen M, Bloigu R, Heikkinen J, Vaananen K, Peltonen J: Decreased bone mineral density and content in neurofibromatosis type 1: lowest local values are located in the load-carrying parts of the body. Osteoporos Int. 2005, 16: 928-936. 10.1007/s00198-004-1801-4.

    Article  CAS  PubMed  Google Scholar 

  20. 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: 305-313.

    PubMed Central  CAS  PubMed  Google Scholar 

  21. Szudek J, Birch P, Riccardi VM, Evans DG, Friedman JM: Associations of clinical features in neurofibromatosis 1 (NF1). Genet Epidemiol. 2000, 19: 429-439. 10.1002/1098-2272(200012)19:4<429::AID-GEPI13>3.0.CO;2-N.

    Article  CAS  PubMed  Google Scholar 

  22. Xu GF, O'Connell P, Viskochil D, Cawthon R, Robertson M, Culver M, Dunn D, Stevens J, Gesteland R, White R: The neurofibromatosis type 1 gene encodes a protein related to GAP. Cell. 1990, 62: 599-608. 10.1016/0092-8674(90)90024-9.

    Article  CAS  PubMed  Google Scholar 

  23. Brannan CI, Perkins AS, Vogel KS, Ratner N, Nordlund ML, Reid SW, Buchberg AM, Jenkins NA, Parada LF, Copeland NG: Targeted disruption of the neurofibromatosis type-1 gene leads to developmental abnormalities in heart and various neural crest-derived tissues. Genes Dev. 1994, 8: 1019-1029. 10.1101/gad.8.9.1019.

    Article  CAS  PubMed  Google Scholar 

  24. Jacks T, Shih TS, Schmitt EM, Bronson RT, Bernards A, Weinberg RA: Tumour predisposition in mice heterozygous for a targeted mutation in Nf1. Nat Genet. 1994, 7: 353-361. 10.1038/ng0794-353.

    Article  CAS  PubMed  Google Scholar 

  25. Xu GF, Lin B, Tanaka K, Dunn D, Wood D, Gesteland R, White R, Weiss R, Tamanoi F: The catalytic domain of the neurofibromatosis type 1 gene product stimulates ras GTPase and complements ira mutants of S. cerevisiae. Cell. 1990, 63: 835-841. 10.1016/0092-8674(90)90149-9.

    Article  CAS  PubMed  Google Scholar 

  26. Ballester R, Marchuk D, Boguski M, Saulino A, Letcher R, Wigler M, Collins F: The NF1 locus encodes a protein functionally related to mammalian GAP and yeast IRA proteins. Cell. 1990, 63: 851-859. 10.1016/0092-8674(90)90151-4.

    Article  CAS  PubMed  Google Scholar 

  27. Martin GA, Viskochil D, Bollag G, McCabe PC, Crosier WJ, Haubruck H, Conroy L, Clark R, O'Connell P, Cawthon RM: The GAP-related domain of the neurofibromatosis type 1 gene product interacts with ras p21. Cell. 1990, 63: 843-849. 10.1016/0092-8674(90)90150-D.

    Article  CAS  PubMed  Google Scholar 

  28. Guo HF, The I, Hannan F, Bernards A, Zhong Y: Requirement of Drosophila NF1 for activation of adenylyl cyclase by PACAP38-like neuropeptides. Science. 1997, 276: 795-798. 10.1126/science.276.5313.795.

    Article  CAS  PubMed  Google Scholar 

  29. 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: 1087-1098. 10.1093/hmg/ddl023.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  30. Hsueh YP: Neurofibromin signaling and synapses. J Biomed Sci. 2007, 14: 461-466. 10.1007/s11373-007-9158-2.

    Article  CAS  PubMed  Google Scholar 

  31. Guo HF, Tong J, Hannan F, Luo L, Zhong Y: A neurofibromatosis-1-regulated pathway is required for learning in Drosophila. Nature. 2000, 403: 895-898. 10.1038/35002593.

    Article  CAS  PubMed  Google Scholar 

  32. Tong J, Hannan F, Zhu Y, Bernards A, Zhong Y: Neurofibromin regulates G protein-stimulated adenylyl cyclase activity. Nat Neurosci. 2002, 5: 95-96. 10.1038/nn792.

    Article  CAS  PubMed  Google Scholar 

  33. Kweh F, Zheng M, Kurenova E, Wallace M, Golubovskaya V, Cance WG: Neurofibromin physically interacts with the N-terminal domain of focal adhesion kinase. Mol Carcinog. 2009, 48: 1005-1017. 10.1002/mc.20552.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  34. Hsueh YP, Roberts AM, Volta M, Sheng M, Roberts RG: Bipartite interaction between neurofibromatosis type I protein (neurofibromin) and syndecan transmembrane heparan sulfate proteoglycans. J Neurosci. 2001, 21: 3764-3770.

    CAS  PubMed  Google Scholar 

  35. Lin YL, Hsueh YP: Neurofibromin interacts with CRMP-2 and CRMP-4 in rat brain. Biochem Biophys Res Commun. 2008, 369: 747-752. 10.1016/j.bbrc.2008.02.095.

    Article  CAS  PubMed  Google Scholar 

  36. Volta M, Calza S, Roberts AM, Roberts RG: Characterisation of the interaction between syndecan-2, neurofibromin and CASK: dependence of interaction on syndecan dimerization. Biochem Biophys Res Commun. 2010, 391: 1216-1221. 10.1016/j.bbrc.2009.12.043.

    Article  CAS  PubMed  Google Scholar 

  37. De Schepper S, Boucneau JM, Westbroek W, Mommaas M, Onderwater J, Messiaen L, Naeyaert JM, Lambert JL: Neurofibromatosis type 1 protein and amyloid precursor protein interact in normal human melanocytes and colocalize with melanosomes. J Invest Dermatol. 2006, 126: 653-659. 10.1038/sj.jid.5700087.

    Article  CAS  PubMed  Google Scholar 

  38. Feng L, Yunoue S, Tokuo H, Ozawa T, Zhang D, Patrakitkomjorn S, Ichimura T, Saya H, Araki N: PKA phosphorylation and 14-3-3 interaction regulate the function of neurofibromatosis type I tumor suppressor, neurofibromin. FEBS Lett. 2004, 557: 275-282. 10.1016/S0014-5793(03)01507-2.

    Article  CAS  PubMed  Google Scholar 

  39. Patrakitkomjorn S, Kobayashi D, Morikawa T, Wilson MM, Tsubota N, Irie A, Ozawa T, Aoki M, Arimura N, Kaibuchi K, Saya H, Araki N: Neurofibromatosis type 1 (NF1) tumor suppressor, neurofibromin, regulates the neuronal differentiation of PC12 cells via its associating protein, CRMP-2. J Biol Chem. 2008, 283: 9399-9413. 10.1074/jbc.M708206200.

    Article  CAS  PubMed  Google Scholar 

  40. D'Angelo I, Welti S, Bonneau F, Scheffzek K: A novel bipartite phospholipid-binding module in the neurofibromatosis type 1 protein. EMBO Rep. 2006, 7: 174-179. 10.1038/sj.embor.7400602.

    Article  PubMed Central  PubMed  Google Scholar 

  41. Welti S, Fraterman S, D'Angelo I, Wilm M, Scheffzek K: The sec14 homology module of neurofibromin binds cellular glycerophospholipids: mass spectrometry and structure of a lipid complex. J Mol Biol. 2007, 366: 551-562. 10.1016/j.jmb.2006.11.055.

    Article  CAS  PubMed  Google Scholar 

  42. Watts GD, Wymer J, Kovach MJ, Mehta SG, Mumm S, Darvish D, Pestronk A, Whyte MP, Kimonis VE: Inclusion body myopathy associated with Paget disease of bone and frontotemporal dementia is caused by mutant valosin-containing protein. Nat Genet. 2004, 36: 377-381. 10.1038/ng1332.

    Article  CAS  PubMed  Google Scholar 

  43. Weihl CC: Another VCP interactor: NF is enough. J Clin Invest. 2011, 121: 4627-4630. 10.1172/JCI61126.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  44. Weihl CC: Valosin containing protein associated fronto-temporal lobar degeneration: clinical presentation, pathologic features and pathogenesis. Curr Alzheimer Res. 2011, 8: 252-260. 10.2174/156720511795563773.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  45. Johnson JO, Mandrioli J, Benatar M, Abramzon Y, Van Deerlin VM, Trojanowski JQ, Gibbs JR, Brunetti M, Gronka S, Wuu J, Ding J, McCluskey L, Martinez-Lage M, Falcone D, Hernandez DG, Arepalli S, Chong S, Schymick JC, Rothstein J, Landi F, Wang YD, Calvo A, Mora G, Sabatelli M, Monsurro MR, Battistini S, Salvi F, Spataro R, Sola P: Borghero Get al.: Exome sequencing reveals VCP mutations as a cause of familial ALS. Neuron. 2010, 68: 857-864. 10.1016/j.neuron.2010.11.036.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  46. Hirabayashi M, Inoue K, Tanaka K, Nakadate K, Ohsawa Y, Kamei Y, Popiel AH, Sinohara A, Iwamatsu A, Kimura Y, Uchiyama Y, Hori S, Kakizuka A: VCP/p97 in abnormal protein aggregates, cytoplasmic vacuoles, and cell death, phenotypes relevant to neurodegeneration. Cell Death Differ. 2001, 8: 977-984. 10.1038/sj.cdd.4400907.

    Article  CAS  PubMed  Google Scholar 

  47. Neumann M, Mackenzie IR, Cairns NJ, Boyer PJ, Markesbery WR, Smith CD, Taylor JP, Kretzschmar HA, Kimonis VE, Forman MS: TDP-43 in the ubiquitin pathology of frontotemporal dementia with VCP gene mutations. J Neuropathol Exp Neurol. 2007, 66: 152-157. 10.1097/nen.0b013e31803020b9.

    Article  PubMed  Google Scholar 

  48. Weihl CC, Temiz P, Miller SE, Watts G, Smith C, Forman M, Hanson PI, Kimonis V, Pestronk A: TDP-43 accumulation in inclusion body myopathy muscle suggests a common pathogenic mechanism with frontotemporal dementia. J Neurol Neurosurg Psychiatry. 2008, 79: 1186-1189. 10.1136/jnnp.2007.131334.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  49. Ritson GP, Custer SK, Freibaum BD, Guinto JB, Geffel D, Moore J, Tang W, Winton MJ, Neumann M, Trojanowski JQ, Lee VM, Forman MS, Taylor JP: TDP-43 mediates degeneration in a novel Drosophila model of disease caused by mutations in VCP/p97. J Neurosci. 2010, 30: 7729-7739. 10.1523/JNEUROSCI.5894-09.2010.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  50. Brunger AT, DeLaBarre B: NSF and p97/VCP: similar at first, different at last. FEBS Lett. 2003, 555: 126-133. 10.1016/S0014-5793(03)01107-4.

    Article  CAS  PubMed  Google Scholar 

  51. Wang Q, Song C, Li CC: Molecular perspectives on p97-VCP: progress in understanding its structure and diverse biological functions. J Struct Biol. 2004, 146: 44-57. 10.1016/j.jsb.2003.11.014.

    Article  CAS  PubMed  Google Scholar 

  52. Rouiller I, DeLaBarre B, May AP, Weis WI, Brunger AT, Milligan RA, Wilson-Kubalek EM: Conformational changes of the multifunction p97 AAA ATPase during its ATPase cycle. Nat Struct Biol. 2002, 9: 950-957. 10.1038/nsb872.

    Article  CAS  PubMed  Google Scholar 

  53. Tang WK, Li D, Li CC, Esser L, Dai R, Guo L, Xia D: A novel ATP-dependent conformation in p97 N-D1 fragment revealed by crystal structures of disease-related mutants. EMBO J. 2010, 29: 2217-2229. 10.1038/emboj.2010.104.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  54. Schroder R, Watts GD, Mehta SG, Evert BO, Broich P, Fliessbach K, Pauls K, Hans VH, Kimonis V, Thal DR: Mutant valosin-containing protein causes a novel type of frontotemporal dementia. Ann Neurol. 2005, 57: 457-461. 10.1002/ana.20407.

    Article  PubMed  Google Scholar 

  55. Kimonis VE, Mehta SG, Fulchiero EC, Thomasova D, Pasquali M, Boycott K, Neilan EG, Kartashov A, Forman MS, Tucker S, Kimonis K, Mumm S, Whyte MP, Smith CD, Watts GD: Clinical studies in familial VCP myopathy associated with Paget disease of bone and frontotemporal dementia. Am J Med Genet A. 2008, 146: 745-757.

    Article  Google Scholar 

  56. Halawani D, LeBlanc AC, Rouiller I, Michnick SW, Servant MJ, Latterich M: Hereditary inclusion body myopathy-linked p97/VCP mutations in the NH2 domain and the D1 ring modulate p97/VCP ATPase activity and D2 ring conformation. Mol Cell Biol. 2009, 29: 4484-4494. 10.1128/MCB.00252-09.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  57. Vij N: AAA ATPase p97/VCP: cellular functions, disease and therapeutic potential. J Cell Mol Med. 2008, 12: 2511-2518. 10.1111/j.1582-4934.2008.00462.x.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  58. Jarosch E, Taxis C, Volkwein C, Bordallo J, Finley D, Wolf DH, Sommer T: Protein dislocation from the ER requires polyubiquitination and the AAA-ATPase Cdc48. Nat Cell Biol. 2002, 4: 134-139. 10.1038/ncb746.

    Article  CAS  PubMed  Google Scholar 

  59. Dreveny I, Kondo H, Uchiyama K, Shaw A, Zhang X, Freemont PS: Structural basis of the interaction between the AAA ATPase p97/VCP and its adaptor protein p47. EMBO J. 2004, 23: 1030-1039. 10.1038/sj.emboj.7600139.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  60. Yoshida H: ER stress and diseases. FEBS J. 2007, 274: 630-658. 10.1111/j.1742-4658.2007.05639.x.

    Article  CAS  PubMed  Google Scholar 

  61. Uchiyama K, Kondo H: p97/p47-Mediated biogenesis of Golgi and ER. J Biochem. 2005, 137: 115-119. 10.1093/jb/mvi028.

    Article  CAS  PubMed  Google Scholar 

  62. Ju JS, Weihl CC: p97/VCP at the intersection of the autophagy and the ubiquitin proteasome system. Autophagy. 2010, 6: 283-285. 10.4161/auto.6.2.11063.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  63. Tresse E, Salomons FA, Vesa J, Bott LC, Kimonis V, Yao TP, Dantuma NP, Taylor JP: VCP/p97 is essential for maturation of ubiquitin-containing autophagosomes and this function is impaired by mutations that cause IBMPFD. Autophagy. 2010, 6: 217-227. 10.4161/auto.6.2.11014.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  64. Meyer H, Bug M, Bremer S: Emerging functions of the VCP/p97 AAA-ATPase in the ubiquitin system. Nat Cell Biol. 2012, 14: 117-123. 10.1038/ncb2407.

    Article  CAS  PubMed  Google Scholar 

  65. Weihl CC, Dalal S, Pestronk A, Hanson PI: Inclusion body myopathy-associated mutations in p97/VCP impair endoplasmic reticulum-associated degradation. Hum Mol Genet. 2006, 15: 189-199.

    Article  CAS  PubMed  Google Scholar 

  66. Ju JS, Fuentealba RA, Miller SE, Jackson E, Piwnica-Worms D, Baloh RH, Weihl CC: Valosin-containing protein (VCP) is required for autophagy and is disrupted in VCP disease. J Cell Biol. 2009, 187: 875-888. 10.1083/jcb.200908115.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  67. Kondo H, Rabouille C, Newman R, Levine TP, Pappin D, Freemont P, Warren G: p47 is a cofactor for p97-mediated membrane fusion. Nature. 1997, 388: 75-78. 10.1038/40411.

    Article  CAS  PubMed  Google Scholar 

  68. Otter-Nilsson M, Hendriks R, Pecheur-Huet EI, Hoekstra D, Nilsson T: Cytosolic ATPases, p97 and NSF, are sufficient to mediate rapid membrane fusion. EMBO J. 1999, 18: 2074-2083. 10.1093/emboj/18.8.2074.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  69. Hetzer M, Meyer HH, Walther TC, Bilbao-Cortes D, Warren G, Mattaj IW: Distinct AAA-ATPase p97 complexes function in discrete steps of nuclear assembly. Nat Cell Biol. 2001, 3: 1086-1091. 10.1038/ncb1201-1086.

    Article  CAS  PubMed  Google Scholar 

  70. Uchiyama K, Jokitalo E, Kano F, Murata M, Zhang X, Canas B, Newman R, Rabouille C, Pappin D, Freemont P, Kondo H: VCIP135, a novel essential factor for p97/p47-mediated membrane fusion, is required for Golgi and ER assembly in vivo. J Cell Biol. 2002, 159: 855-866. 10.1083/jcb.200208112.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  71. Uchiyama K, Jokitalo E, Lindman M, Jackman M, Kano F, Murata M, Zhang X, Kondo H: The localization and phosphorylation of p47 are important for Golgi disassembly-assembly during the cell cycle. J Cell Biol. 2003, 161: 1067-1079. 10.1083/jcb.200303048.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  72. Kano F, Kondo H, Yamamoto A, Tanaka AR, Hosokawa N, Nagata K, Murata M: The maintenance of the endoplasmic reticulum network is regulated by p47, a cofactor of p97, through phosphorylation by cdc2 kinase. Genes Cells. 2005, 10: 333-344. 10.1111/j.1365-2443.2005.00837.x.

    Article  CAS  PubMed  Google Scholar 

  73. Vedrenne C, Hauri HP: Morphogenesis of the endoplasmic reticulum: beyond active membrane expansion. Traffic. 2006, 7: 639-646. 10.1111/j.1600-0854.2006.00419.x.

    Article  CAS  PubMed  Google Scholar 

  74. Krick R, Bremer S, Welter E, Schlotterhose P, Muehe Y, Eskelinen EL, Thumm M: Cdc48/p97 and Shp1/p47 regulate autophagosome biogenesis in concert with ubiquitin-like Atg8. J Cell Biol. 2010, 190: 965-973. 10.1083/jcb.201002075.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  75. Mizuno Y, Hori S, Kakizuka A, Okamoto K: Vacuole-creating protein in neurodegenerative diseases in humans. Neurosci Lett. 2003, 343: 77-80. 10.1016/S0304-3940(03)00280-5.

    Article  CAS  PubMed  Google Scholar 

  76. Forman MS, Mackenzie IR, Cairns NJ, Swanson E, Boyer PJ, Drachman DA, Jhaveri BS, Karlawish JH, Pestronk A, Smith TW, Tu PH, Watts GD, Markesbery WR, Smith CD, Kimonis VE: Novel ubiquitin neuropathology in frontotemporal dementia with valosin-containing protein gene mutations. J Neuropathol Exp Neurol. 2006, 65: 571-581. 10.1097/00005072-200606000-00005.

    Article  CAS  PubMed  Google Scholar 

  77. Rumpf S, Lee SB, Jan LY, Jan YN: Neuronal remodeling and apoptosis require VCP-dependent degradation of the apoptosis inhibitor DIAP1. Development. 2011, 138: 1153-1160. 10.1242/dev.062703.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  78. Li W, Cui Y, Kushner SA, Brown RA, Jentsch JD, Frankland PW, Cannon TD, Silva AJ: The HMG-CoA reductase inhibitor lovastatin reverses the learning and attention deficits in a mouse model of neurofibromatosis type 1. Curr Biol. 2005, 15: 1961-1967. 10.1016/j.cub.2005.09.043.

    Article  CAS  PubMed  Google Scholar 

  79. Mendola CE, Backer JM: Lovastatin blocks N-ras oncogene-induced neuronal differentiation. Cell Growth Differ. 1990, 1: 499-502.

    CAS  PubMed  Google Scholar 

  80. Sebti SM, Tkalcevic GT, Jani JP: Lovastatin, a cholesterol biosynthesis inhibitor, inhibits the growth of human H-ras oncogene transformed cells in nude mice. Cancer Commun. 1991, 3: 141-147.

    CAS  PubMed  Google Scholar 

  81. Hering H, Lin CC, Sheng M: Lipid rafts in the maintenance of synapses, dendritic spines, and surface AMPA receptor stability. J Neurosci. 2003, 23: 3262-3271.

    CAS  PubMed  Google Scholar 

  82. Cao J, Wang J, Qi W, Miao HH, Ge L, DeBose-Boyd RA, Tang JJ, Li BL, Song BL: Ufd1 is a cofactor of gp78 and plays a key role in cholesterol metabolism by regulating the stability of HMG-CoA reductase. Cell Metab. 2007, 6: 115-128. 10.1016/j.cmet.2007.07.002.

    Article  CAS  PubMed  Google Scholar 

  83. Leichner GS, Avner R, Harats D, Roitelman J: Dislocation of HMG-CoA reductase and Insig-1, two polytopic endoplasmic reticulum proteins, en route to proteasomal degradation. Mol Biol Cell. 2009, 20: 3330-3341. 10.1091/mbc.E08-09-0953.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  84. Hartman IZ, Liu P, Zehmer JK, Luby-Phelps K, Jo Y, Anderson RG, DeBose-Boyd RA: Sterol-induced dislocation of 3-hydroxy-3-methylglutaryl coenzyme A reductase from endoplasmic reticulum membranes into the cytosol through a subcellular compartment resembling lipid droplets. J Biol Chem. 2010, 285: 19288-19298. 10.1074/jbc.M110.134213.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

Yi-Ping Hsueh is supported by Academia Sinica (AS-100-TP-B09) and National Science Council (NSC 98-2321-B-001-002, NSC 99-2321-B-001-032, NSC 100-2321-B-001-022, and NSC 98-2311-B-001-012-MY3).

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Yi-Ping Hsueh.

Additional information

Competing interests

The author declares that they have no competing interests.

Authors' contributions

Y-PH conceived and prepared this review.

Authors’ original submitted files for images

Below are the links to the authors’ original submitted files for images.

Authors’ original file for figure 1

Rights and permissions

This article is published under license to BioMed Central Ltd. This is an Open Access article distributed under the terms of the Creative Commons Attribution License (http://creativecommons.org/licenses/by/2.0), which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited.

Reprints and permissions

About this article

Cite this article

Hsueh, YP. From neurodevelopment to neurodegeneration: the interaction of neurofibromin and valosin-containing protein/p97 in regulation of dendritic spine formation. J Biomed Sci 19, 33 (2012). https://doi.org/10.1186/1423-0127-19-33

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/1423-0127-19-33

Keywords