Neurofilament protein-L (NF-L) is the core component of neurofilaments. Recent studies indicate that the NF-L mutations reported in human Charcot–Marie–Tooth (CMT) disease lead to the formation of NF-L aggregates and result in axon degeneration of motor and sensory neurons, which are thought to be the cause of CMT disease type 2E. In the present study, we investigated the dynamic regulation of NF-L assembly and the mechanism of aggregate formation of CMT NF-L mutants. We report that 14-3-3 proteins interact with NF-L in a phosphorylation-dependent manner. Investigation of mutations of phospho-serine sites at the head domain of NF-L revealed that several phosphorylation sites, particularly Ser43 and Ser55, were important for 14-3-3 binding. 14-3-3 overexpression resulted in a significant increase in the dynamic exchange rate of NF-L subunits and induced striking disassembly of neurofilaments. CMT NF-L mutants, particularly those with mutations in the Pro8 and Pro22 sites of the NF-L head domain, led to substantially diminished interaction between 14-3-3 and NF-L, which resulted in the formation of NF-L aggregates and the disruption of the neurofilament co-assembly of NF-L and NF-M. However, aggregate formation in CMT NF-L mutants was downregulated by 14-3-3 overexpression. Taken together, these results suggest the important role of 14-3-3 in the dynamic regulation of NF-L assembly, and in the capacity to prevent the formation of NF-L aggregates. Thus, the 14-3-3 proteins are a possible molecular target for CMT disease therapy.

Neurofilaments are type IV intermediate filaments (IFs), and are the main IFs of neurons and an important component of axons. Neurofilaments are obligate heteropolymers of neurofilament proteins: NF-L, NF-M and NF-H (Lee et al., 1993). In common with other IF proteins, neurofilament triplet proteins are composed of an α-helical rod domain, a globular N-terminal head domain and a non-α-helical C-terminal tail domain. The central core of neurofilaments is made up of the rod domains of NF-L, NF-M and NF-H, and the tail domains of NF-M and NF-H are structural components of cross-bridges between adjacent neurofilaments and are involved in the parallel bundling of neurofilaments (Hirokawa et al., 1984; Nakagawa et al., 1995; Chen et al., 2000). The expression of NF-L alone in Sf9 cells lacking endogenous cytoplasmic IFs results in the formation of 10-nm short filaments, while NF-M or NF-H alone cannot self-assemble into 10-nm filaments in vivo (Hirokawa et al., 1984; Nakagawa et al., 1995; Chen et al., 2000). Co-assembly experiments with truncated NF-L showed that the head domain of NF-L is more important for assembly than the tail domain (Chin et al., 1991; Ching and Liem, 1993; Kim et al., 2007). And phosphorylation of the NF-L head domain is important for the regulation of neurofilament assembly (Hisanaga et al., 1990; Sihag and Nixon, 1991; Hisanaga et al., 1994; Gibb et al., 1998; Hashimoto et al., 1998). A series of NF-L head domain phosphorylation sites has been reported: Ser2, 12, 26, 27, 33, 37, 41, 43, 49, 51, 55, 57, 62, 66 (Gonda et al., 1990; Sihag and Nixon, 1991; Cleverley et al., 1998; Hashimoto et al., 1998; Nakamura et al., 2000; Trimpin et al., 2004). However, only Ser2, 55, 57 and 66 are reported to be endogenous phosphorylation sites (Giasson et al., 1996; Hashimoto et al., 2000; Trimpin et al., 2004). Phosphorylation of the NF-L head domain is thought to regulate neurofilament assembly (Sihag and Nixon, 1991; Hisanaga et al., 1994; Giasson et al., 1996; Cleverley et al., 1998; Hashimoto et al., 1998).

Charcot–Marie–Tooth disease (CMT) is the most common inherited degenerative disease of the peripheral nervous system. Its clinical manifestations are limb weakness, atrophy and sensory loss. The first CMT NF-L mutation, Gln333Pro, was linked with CMT disease type 2 in 2000 (Mersiyanova et al., 2000), then, more and more CMT NF-L mutations were revealed, many of which are located in the NF-L head domain: NF-L E7K, P8Q, P8R, P8L, P22S, P22T, and E89K (Jordanova et al., 2003; Sasaki et al., 2006). These mutations affect neurofilament assembly and NF-L head domain phosphorylation (Brownlees et al., 2002; Perez-Olle et al., 2002; Perez-Olle et al., 2004; Sasaki et al., 2006). However, the mechanism of how CMT NF-L mutations lead to abnormal neurofilament assembly and finally cause the disease is unclear.

The 14-3-3 proteins, abundant in brain (Moore et al., 1968), are important signal transduction adaptors. In mammals, there are seven isoforms of these proteins: β, γ, ε, ζ, η, θ, and σ. The interactions of 14-3-3 proteins are dependent on specific phospho-serine-containing motifs of the target proteins (Muslin et al., 1996). There are two kinds of 14-3-3 binding motifs: RSXpSXP and RXXXpSXP (Muslin et al., 1996; Yaffe et al., 1997). 14-3-3 proteins interact with many IFs and these interactions depend on head domain Ser/Thr phosphorylation of the IF proteins. 14-3-3 proteins associate with keratin 18 via Ser33 phosphorylation (Liao and Omary, 1996; Ku et al., 1998); they bind to vimentin through its head domain phosphorylation (Tzivion et al., 2000); and they interact with GFAP via Ser8 phosphorylation (Li et al., 2006). It is reported that 14-3-3 proteins play important roles in the regulation of IF dynamics (Liao and Omary, 1996; Li et al., 2006; Sivaramakrishnan et al., 2009). Since NF-L assembly is regulated by head domain phosphorylation and the mechanism is unclear, we focused on elucidating whether 14-3-3 proteins interact with NF-L and regulate NF-L assembly, and whether these proteins play a role in the abnormal neurofilament assembly caused by CMT NF-L mutations. Our results showed that 14-3-3 proteins interacted with NF-L and affected NF-L dynamic assembly; furthermore, 14-3-3 proteins also alleviated the aggregate formation of CMT NF-L mutants, which reveals 14-3-3 proteins as possible molecular targets for human CMT disease therapy.

Association of 14-3-3 proteins with NF-L

To investigate whether 14-3-3 proteins associate with neurofilament proteins in mouse brain, pull-down assays of the seven isoforms of GST–14-3-3 fusion proteins were carried out. The results showed that all seven isoforms of 14-3-3 exclusively associated with NF-L, but no interaction between 14-3-3 and either NF-M or NF-H was detected (Fig. 1A). The results also showed stronger interactions of 14-3-3β and 14-3-3γ with NF-L than the other 14-3-3 isoforms (Fig. 1A). Mouse brain lysate also contained other IF proteins such as GFAP and vimentin, which were capable of co-assembly with NF-L to form 10-nm IFs (Chin et al., 1991; Ching and Liem, 1993; Li et al., 2006). To determine the interaction between 14-3-3 and NF-L is independent of other IF proteins, SW13 Cl2 Vim cells that lack endogenous cytoplasmic IF proteins were used to perform co-immunoprecipitation assays. Analysis of co-immunoprecipitation of green fluorescent protein (GFP)-tagged 14-3-3γ with anti-GFP antibody revealed that 14-3-3γ still associated with NF-L in the absence of endogenous IF proteins (Fig. 1B). Further analysis revealed that 14-3-3 not only associated with soluble NF-L subunits, but also interacted with pellets of filaments (supplementary material Fig. S4). Furthermore, co-immunoprecipitation assay with anti-14-3-3γ antibody showed that endogenous 14-3-3γ of SW13 Cl2 Vim cells also interacted with NF-L (Fig. 1C). To determine the capacity of the seven isoforms to associate with NF-L in the absence of endogenous IFs in vivo, NF-L and each GFP-tagged 14-3-3 isoform were co-transfected into SW13 Cl2 Vim cells, and the co-immunoprecipitation results showed that all seven isoforms associated with NF-L, with 14-3-3γ displaying the strongest association (Fig. 1D), which confirmed the results of the 14-3-3 pull-down assays (Fig. 1A).

Fig. 1.

Seven isoforms of 14-3-3 proteins all interact with NF-L. (A) Seven prokaryotic-expressed GST–14-3-3 proteins were incubated with supernatants of adult mouse brain homogenates. Pull-down products were analyzed by western blots with rabbit anti-NF-L and -NF-H and mouse anti-NF-M and anti-GST antibodies, respectively. (B) SW13 Cl2 Vim cells co-transfected with 14-3-3γ–GFP and NF-L–FLAG were co-immunoprecipitated (IP) with rabbit anti-GFP antibody; products were detected with mouse anti-FLAG and anti-GFP antibodies, respectively. (C) SW13 Cl2 Vim cells transfected with NF-L–FLAG were co-immunoprecipitated with rabbit anti-14-3-3γ antibody; products were detected with mouse anti-14-3-3γ and anti-FLAG antibodies, respectively. (D) SW13 Cl2 Vim cells were transfected with seven isoforms of 14-3-3–GFP and NF-L–FLAG, respectively. Products of co-immunoprecipitation with rabbit anti-GFP antibody were detected with mouse anti-FLAG and anti-GFP antibodies, respectively.

Fig. 1.

Seven isoforms of 14-3-3 proteins all interact with NF-L. (A) Seven prokaryotic-expressed GST–14-3-3 proteins were incubated with supernatants of adult mouse brain homogenates. Pull-down products were analyzed by western blots with rabbit anti-NF-L and -NF-H and mouse anti-NF-M and anti-GST antibodies, respectively. (B) SW13 Cl2 Vim cells co-transfected with 14-3-3γ–GFP and NF-L–FLAG were co-immunoprecipitated (IP) with rabbit anti-GFP antibody; products were detected with mouse anti-FLAG and anti-GFP antibodies, respectively. (C) SW13 Cl2 Vim cells transfected with NF-L–FLAG were co-immunoprecipitated with rabbit anti-14-3-3γ antibody; products were detected with mouse anti-14-3-3γ and anti-FLAG antibodies, respectively. (D) SW13 Cl2 Vim cells were transfected with seven isoforms of 14-3-3–GFP and NF-L–FLAG, respectively. Products of co-immunoprecipitation with rabbit anti-GFP antibody were detected with mouse anti-FLAG and anti-GFP antibodies, respectively.

Co-localization of 14-3-3 and NF-L in neurons and SW13 Vim cells

To investigate the co-localization of 14-3-3 and NF-L, indirect immunofluorescence assays were carried out in SW13 Cl2 Vim cells and dorsal root ganglion (DRG) neurons. In SW13 Cl2 Vim cells, transfected NF-L displayed as morphology of small granules dispersed throughout cytoplasm (Fig. 2A), and the granule morphology is common to either human NF-L (hNF-L) or mouse NF-L (mNF-L) or mouse NF-L (mNF-L) with a FLAG tag (mNF-L–FLAG) (supplementary material Fig. S1A,B). Interestingly, these granules were revealed as a mass of short 10-nm filaments under transmission electron microscopy (TEM) (supplementary material Fig. S1B). To confirm this phenomenon, Sf9 cells that are also lack of endogenous IF proteins were infected with baculoviruses expressing hNF-L, mNF-L or mNF-L–FLAG, clusters of short 10-nm filaments were observed in the cytoplasm (data not shown), and 0.1% Triton X-100 treatment of the infected Sf9 cells before fixation got a clear view of 10-nm short filaments assemble of mNF-L–FLAG (supplementary material Fig. S1C). In SW13 Cl2 Vim cells, co-localizations between endogenous 14-3-3γ dots and NF-L granules were observed (Fig. 2A). When cells were co-transfected with NF-L and NF-M, they co-assembled into neurofilaments (Fig. 2B; supplementary material Fig. S1D). Some of endogenous 14-3-3γ stained as fibrous distribution and co-localized with neurofilaments (Fig. 2B, top). When cells were co-transfected with NF-L, NF-M and 14-3-3γ–red fluorescent protein (RFP), co-localization between fibrously distributed 14-3-3γ–RFP and neurofilament bundles was also observed (Fig. 2B, bottom). And there are also some other neurofilament bundles showed no co-localization with 14-3-3γ–RFP (Fig. 2B, bottom). Immunofluorescence staining of DRG neurons revealed that, some fibrously distributed 14-3-3 co-localized with neurofilaments in axons (Fig. 2C, top) and growth cone (Fig. 2C, bottom). In order to determine the direct interaction between 14-3-3 and NF-L, we performed fluorescence resonance energy transfer (FRET) analyses between cyan fluorescent protein (CFP)-tagged 14-3-3 (donor) and yellow fluorescent protein (YFP)-tagged NF-L (acceptor) in SW13 Cl2 Vim cells. When the selected co-localization granules were bleached of acceptor fluorescence (NF-L–YFP), the intensity of donor fluorescence (CFP–14-3-3) increased over the value before bleaching (Fig. 2D,E, labels A1, D1). However, the control granules without bleaching showed no significant change in the fluorescence intensity of donor and acceptor (Fig. 2D,E, labels A2, D2). FRET analysis of CFP–14-3-3 and NF-L–YFP showed about 17% FRET efficiency (Ef) of the bleached zone compared with the control efficiency (Cf) of about 3% in the unbleached zone (Fig. 2F).

Fig. 2.

Co-localization of 14-3-3γ and NF-L in SW13 Cl2 Vim cells and DRG neurons. (A) SW13 Cl2 Vim cells transfected with NF-L were stained with anti-NF-L and anti-14-3-3 antibodies, arrows indicating the co-localizing granules. (B) SW13 Cl2 Vim cells were co-transfected with NF-L and NF-M, and stained with anti-NF-L and anti-14-3-3γ antibodies (upper panels); and co-transfected with 14-3-3γ–RFP, NF-L–FLAG and NF-M, and stained with anti-FLAG antibody (lower panels), arrows indicating the co-localization of 14-3-3 and neurofilaments, and arrowheads indicating neurofilaments alone. (C) Immunofluorescent staining of endogenous 14-3-3γ and NF-L of DRG neurons with anti-NF-L and anti-14-3-3 antibodies, arrows indicating the co-localization of 14-3-3 and neurofilaments. (D) FRET assay in SW13 Cl2 Vim cells co-transfected with CFP–14-3-3γ and NF-L–YFP. A1 and D1 represent the fluorescence intensity of CFP–14-3-3γ and NF-L–YFP, respectively, of the bleached zone; A2 and D2 represent the fluorescence intensity of CFP–14-3-3γ and NF-L–YFP, respectively, of the control unbleached zone. (E) Change in the fluorescence intensity of A1, A2, D1, and D2 before and after photobleaching. (F) FRET efficiency between CFP–14-3-3γ and NF-L–YFP in the bleached zone (Ef) and control unbleached zone (Cf). Values are means±s.e.m., n = 11; ***P<0.001 (Student's t-test). Scale bar: 10 µm.

Fig. 2.

Co-localization of 14-3-3γ and NF-L in SW13 Cl2 Vim cells and DRG neurons. (A) SW13 Cl2 Vim cells transfected with NF-L were stained with anti-NF-L and anti-14-3-3 antibodies, arrows indicating the co-localizing granules. (B) SW13 Cl2 Vim cells were co-transfected with NF-L and NF-M, and stained with anti-NF-L and anti-14-3-3γ antibodies (upper panels); and co-transfected with 14-3-3γ–RFP, NF-L–FLAG and NF-M, and stained with anti-FLAG antibody (lower panels), arrows indicating the co-localization of 14-3-3 and neurofilaments, and arrowheads indicating neurofilaments alone. (C) Immunofluorescent staining of endogenous 14-3-3γ and NF-L of DRG neurons with anti-NF-L and anti-14-3-3 antibodies, arrows indicating the co-localization of 14-3-3 and neurofilaments. (D) FRET assay in SW13 Cl2 Vim cells co-transfected with CFP–14-3-3γ and NF-L–YFP. A1 and D1 represent the fluorescence intensity of CFP–14-3-3γ and NF-L–YFP, respectively, of the bleached zone; A2 and D2 represent the fluorescence intensity of CFP–14-3-3γ and NF-L–YFP, respectively, of the control unbleached zone. (E) Change in the fluorescence intensity of A1, A2, D1, and D2 before and after photobleaching. (F) FRET efficiency between CFP–14-3-3γ and NF-L–YFP in the bleached zone (Ef) and control unbleached zone (Cf). Values are means±s.e.m., n = 11; ***P<0.001 (Student's t-test). Scale bar: 10 µm.

Requirement for head domain phosphorylation of NF-L for 14-3-3 association

To determine which domain of NF-L is required for binding of 14-3-3, mutants of NF-L with the head or tail domain truncated were constructed (Fig. 3A). After co-transfection of NF-M and either wild-type or truncated NF-L into SW13 Cl2 Vim cells, immunofluorescence analyses revealed the disrupted co-assembly of neurofilaments in head domain-truncated NF-L and NF-M, while the tail domain-truncated mutant still co-assembled with NF-M into filaments (Fig. 3B). These filament networks formed by tail domain-truncated NF-L and NF-M were irregular and distributed randomly in the cytosol, unlike the classical filament array extending from the nucleus to the periphery of the cell formed by wild-type NF-L and NF-M. These immunofluorescence data suggested the importance of the head domain for the assembly of NF-L (Fig. 3B). Co-immunoprecipitation assays demonstrated that deletion of the head domain of NF-L prevented the association of 14-3-3, whereas NF-L with the tail domain deleted retained association, further suggesting the necessity of the NF-L head domain for 14-3-3 association (Fig. 3C). Commonly, phosphorylation of binding targets is required for 14-3-3 interaction. To determine whether interaction between 14-3-3 and NF-L depends on phosphorylation, two 14-3-3 binding motifs at the head domain of NF-L were predicted at website: http://scansite.mit.edu/motifscan_seq.phtml (Fig. 3D). However, mutation of both predicted phosphorylation sites only diminished 14-3-3 binding to some extent, but not completely (Fig. 3E), indicating that some other phosphorylation sites were also required for 14-3-3 binding. It is known that Ser2, 55, 57 and 66 are phosphorylation sites of the NF-L head domain in vivo (Giasson et al., 1996; Hashimoto et al., 2000; Trimpin et al., 2004). Mutation of these sites and the predicted 14-3-3 binding sites simultaneously further diminished the 14-3-3 binding. Only the NF-L with mutation of all serines of the head domain showed completely inhibited 14-3-3 binding (Fig. 3E,F), indicating multiple phosphorylation sites involved in the interaction of 14-3-3 with NF-L. Treatment with the phosphatase-2A inhibitor, okadaic acid (OA), increases the head domain phosphorylation of NF-L (Sacher et al., 1994). After OA treatment, we found that the association between wild-type NF-L and 14-3-3 was increased, whereas the interaction with NF-L having all serines of the head domain mutated remained unchanged (Fig. 3G,H), which confirmed the necessity of phosphorylation of the NF-L head domain for binding of 14-3-3.

Fig. 3.

14-3-3γ interacts with NF-L head domain and is dependent on phosphorylation. (A) Schematic representation of wild-type NF-L, NF-L head domain deletion mutant (NF-L-ΔH), and NF-L tail domain deletion mutant (NF-L-ΔT). (B) SW13 Cl2 Vim cells co-transfected with NF-L, NF-L-ΔH or NF-L-ΔT and with NF-M were immunostained with anti-NF-L and anti-NF-M antibodies. (C) SW13 Cl2 Vim cells co-transfected with NF-L–FLAG, NF-L-ΔH–FLAG or NF-L-ΔT–FLAG and with 14-3-3γ–GFP, were co-immunoprecipitated with rabbit anti-GFP antibody and subjected to western blotting. (D) Predication of 14-3-3 binding motifs of NF-L head domain. (E) SW13 Cl2 Vim cells co-transfected with NF-L-wt, NF-L-Ala43, 57, NF-L-Ala2, 43, 55, 57 and 66 or NF-L-all mut (mutation of all serines of NF-L head domain) and 14-3-3γ–GFP were co-immunoprecipitated with rabbit anti-GFP antibody and subjected to western blotting. (F) Relative quantification of 14-3-3γ–GFP interaction by standardizing to NF-L-wt. Values are means±s.e.m., n = 3; **P<0.01, ***P<0.001 versus NF-L-wt (Student's t-test). (G) SW13 Cl2 Vim cells co-transfected with 14-3-3γ–GFP and NF-L-wt or NF-L-all mut were subjected to okadaic acid (OA) treatment or DMSO as control, and co-immunoprecipitated with rabbit anti-GFP antibody. (H) Relative quantification of 14-3-3γ–GFP interaction by standardizing to DMSO control. Values are means±s.e.m., n = 3; *P<0.05 (Student's t-test). Scale bar: 10 µm. n.s., not significant; wt, wild-type.

Fig. 3.

14-3-3γ interacts with NF-L head domain and is dependent on phosphorylation. (A) Schematic representation of wild-type NF-L, NF-L head domain deletion mutant (NF-L-ΔH), and NF-L tail domain deletion mutant (NF-L-ΔT). (B) SW13 Cl2 Vim cells co-transfected with NF-L, NF-L-ΔH or NF-L-ΔT and with NF-M were immunostained with anti-NF-L and anti-NF-M antibodies. (C) SW13 Cl2 Vim cells co-transfected with NF-L–FLAG, NF-L-ΔH–FLAG or NF-L-ΔT–FLAG and with 14-3-3γ–GFP, were co-immunoprecipitated with rabbit anti-GFP antibody and subjected to western blotting. (D) Predication of 14-3-3 binding motifs of NF-L head domain. (E) SW13 Cl2 Vim cells co-transfected with NF-L-wt, NF-L-Ala43, 57, NF-L-Ala2, 43, 55, 57 and 66 or NF-L-all mut (mutation of all serines of NF-L head domain) and 14-3-3γ–GFP were co-immunoprecipitated with rabbit anti-GFP antibody and subjected to western blotting. (F) Relative quantification of 14-3-3γ–GFP interaction by standardizing to NF-L-wt. Values are means±s.e.m., n = 3; **P<0.01, ***P<0.001 versus NF-L-wt (Student's t-test). (G) SW13 Cl2 Vim cells co-transfected with 14-3-3γ–GFP and NF-L-wt or NF-L-all mut were subjected to okadaic acid (OA) treatment or DMSO as control, and co-immunoprecipitated with rabbit anti-GFP antibody. (H) Relative quantification of 14-3-3γ–GFP interaction by standardizing to DMSO control. Values are means±s.e.m., n = 3; *P<0.05 (Student's t-test). Scale bar: 10 µm. n.s., not significant; wt, wild-type.

Phospho-serines required for 14-3-3 binding

To investigate which phosphorylation sites of the NF-L head domain mediated 14-3-3 binding, a series of single mutations of reported serine phosphorylation sites of this domain were constructed. Co-immunoprecipitation results demonstrated that single site mutations of Ser41, 43, 49 and 55 significantly reduced 14-3-3 binding, and mutations of Ser26 and 62 also decreased it, although not so markedly (Fig. 4A). Immunofluorescence assays of SW13 Cl2 Vim cells transfected with these NF-L mutants revealed that those mutants affecting 14-3-3 binding, including S26A, S49A, S55A and S62A, resulted in the formation of sheet-like aggregates. In contrast, wild-type NF-L displayed as granules dispersed in the cytosol (Fig. 4B,C), indicating the important role of 14-3-3 in regulating NF-L assembly. However, some mutants, such as S41A and S43A, which significantly reduced 14-3-3 binding, displayed no formation of aggregates, suggesting these sites affected 14-3-3 binding only (Fig. 4B,C). To investigate whether these NF-L mutants affect the co-assembly with NF-M, NF-L mutants and NF-M were co-transfected into SW13 Cl2 Vim cells. Results showed that all of these mutants could co-assembly with NF-M into neurofilament networks (supplementary material Fig. S2). Therefore, co-assembly with NF-M into neurofilament networks was not affected by mutation of single phosphorylation site of NF-L. It is reported that Ser41, 43, 49, 55 and 62 are phosphorylated by protein kinase A (PKA) (Cleverley et al., 1998; Hashimoto et al., 2000; Nakamura et al., 2000), and our study demonstrated that mutation of these phosphorylation sites inhibited the interaction between 14-3-3 and NF-L (Fig. 4A), which together suggested the role of PKA in the regulation of 14-3-3 binding to NF-L. Assays with the PKA inhibitor KT5720 and the activator forskolin showed that the inhibitor decreased the interaction between 14-3-3 and NF-L, while the activator increased the interaction (Fig. 4D). Furthermore, the interaction between the NF-L S55A mutant and 14-3-3 was still increased by treatment with the PKA activator, indicating that several PKA phosphorylation sites of NF-L were involved in 14-3-3 association besides Ser55.

Fig. 4.

Phosphorylation sites of NF-L head domain that are involved in 14-3-3 interaction. (A) SW13 Cl2 Vim cells co-transfected with NF-L-wt, or single site mutation of Ser2, 12, 26, 27, 33, 41, 43, 49, 51, 55, 57, 62 or 66, and 14-3-3γ–GFP were co-immunoprecipitated with rabbit anti-GFP antibody and subjected to western blotting. (B) Exogenous expression of single phosphorylation site mutation of Ser2, 12, 26, 27, 33, 37, 41, 43, 49, 51, 55, 57, 62 or 66 in SW13 Cl2 Vim cells, and stained with anti-NF-L antibody. (C) Statistical analysis of cells containing sheet-like structure, standardized to NF-L-wt. Values are means±s.e.m., n = 3. (D) SW13 Cl2 Vim cells co-transfected with NF-L–FLAG and 14-3-3–GFP were subjected to DMSO, KT5720 (PKA inhibitor) or forskolin (PKA activator) treatment respectively before co-immunoprecipitation with rabbit anti-GFP antibody. Also, SW13 Cl2 Vim cells co-transfected with NF-L-S55A–FLAG and 14-3-3–GFP were subjected to DMSO or forskolin treatment respectively before co-immunoprecipitation. Scale bar: 10 µm. WT, wild-type.

Fig. 4.

Phosphorylation sites of NF-L head domain that are involved in 14-3-3 interaction. (A) SW13 Cl2 Vim cells co-transfected with NF-L-wt, or single site mutation of Ser2, 12, 26, 27, 33, 41, 43, 49, 51, 55, 57, 62 or 66, and 14-3-3γ–GFP were co-immunoprecipitated with rabbit anti-GFP antibody and subjected to western blotting. (B) Exogenous expression of single phosphorylation site mutation of Ser2, 12, 26, 27, 33, 37, 41, 43, 49, 51, 55, 57, 62 or 66 in SW13 Cl2 Vim cells, and stained with anti-NF-L antibody. (C) Statistical analysis of cells containing sheet-like structure, standardized to NF-L-wt. Values are means±s.e.m., n = 3. (D) SW13 Cl2 Vim cells co-transfected with NF-L–FLAG and 14-3-3–GFP were subjected to DMSO, KT5720 (PKA inhibitor) or forskolin (PKA activator) treatment respectively before co-immunoprecipitation with rabbit anti-GFP antibody. Also, SW13 Cl2 Vim cells co-transfected with NF-L-S55A–FLAG and 14-3-3–GFP were subjected to DMSO or forskolin treatment respectively before co-immunoprecipitation. Scale bar: 10 µm. WT, wild-type.

Roles of 14-3-3 in regulation of NF-L dynamics and structural reorganization of neurofilaments

Previous studies revealed the importance of 14-3-3 in the regulation of the dynamics of keratin and GFAP (Liao and Omary, 1996; Li et al., 2006; Sivaramakrishnan et al., 2009). To determine the role of 14-3-3 in the regulation of NF-L dynamics and neurofilament assembly, fluorescence recovery after photobleaching (FRAP) analyses were carried out (Fig. 5A). The results revealed that co-expression of 14-3-3 clearly increased the dynamic exchange rate of NF-L, with an average t1/2 of 16±1 s and a rate constant of 0.049±0.004, compared to an average t1/2 of 23±3 s and rate constant of 0.035±0.003 in control cells (Fig. 5B,C). Since 14-3-3 facilitated the dynamic exchange rate of NF-L, we set out to assess the role of 14-3-3 overexpression in the regulation of the neurofilament network. Normally, NF-L and NF-M could co-assemble into extensive neurofilament networks; however, upon co-expression of NF-L and NF-M with 14-3-3, overexpression of 14-3-3 disassembled the neurofilament network and little extensive neurofilament networks were observed (Fig. 5D, lower panels). However, in cells with low amounts of 14-3-3 expression, the neurofilament assembly was unaffected (Fig. 5D, upper panels). Statistical analysis showed that the percentage of cells with neurofilament network was decreased from 93% to 49% owing to 14-3-3 overexpression (Fig. 5E).

Fig. 5.

Overexpression of 14-3-3 enhances the NF-L dynamic exchange rate and leads to neurofilament disassembly. (A) SW13 Cl2 Vim cells co-transfected with NF-L-EGFP and HA–14-3-3γ or empty vector as control were subjected to FRAP assays. Numbers 0–90 represent recovery time (seconds) after photobleaching. (B) FRAP t1/2 of control group and 14-3-3 overexpression group were calculated. Values are means±s.e.m., n = 18; *P<0.05 (Student's t test). (C) Rate constants of FRAP of control group and 14-3-3 overexpression group were calculated. Values are means±s.e.m., n = 18; **P<0.01 (Student's t test). (D) SW13 Cl2 Vim cells were co-transfected with NF-L–FLAG, NF-M and 14-3-3γ–RFP, and neurofilaments were stained with anti–FLAG antibody. (E) Statistical analysis of percentage of cells with a neurofilament network. Values are means±s.e.m., n = 3; **P<0.01 (Student's t test). Scale bar: 10 µm.

Fig. 5.

Overexpression of 14-3-3 enhances the NF-L dynamic exchange rate and leads to neurofilament disassembly. (A) SW13 Cl2 Vim cells co-transfected with NF-L-EGFP and HA–14-3-3γ or empty vector as control were subjected to FRAP assays. Numbers 0–90 represent recovery time (seconds) after photobleaching. (B) FRAP t1/2 of control group and 14-3-3 overexpression group were calculated. Values are means±s.e.m., n = 18; *P<0.05 (Student's t test). (C) Rate constants of FRAP of control group and 14-3-3 overexpression group were calculated. Values are means±s.e.m., n = 18; **P<0.01 (Student's t test). (D) SW13 Cl2 Vim cells were co-transfected with NF-L–FLAG, NF-M and 14-3-3γ–RFP, and neurofilaments were stained with anti–FLAG antibody. (E) Statistical analysis of percentage of cells with a neurofilament network. Values are means±s.e.m., n = 3; **P<0.01 (Student's t test). Scale bar: 10 µm.

Disruption of 14-3-3 binding by CMT NF-L mutations and the role of 14-3-3 in aggregate formation in these mutations

Reported CMT NF-L mutants show defects in neurofilament assembly and transport (Perez-Olle et al., 2004), while P22S and P22T mutants abolish phosphorylation of Thr21 in the NF-L head domain (Sasaki et al., 2006). P8 and P22 residues are mutational hot-spots of NF-L in CMT disease, and so far, five mutations of these two residues have been reported in CMT disease: P8Q, P8R, P8L, P22S and P22T (Jordanova et al., 2003; Perez-Olle et al., 2004; Sasaki et al., 2006; Dequen et al., 2010). To investigate the effects of these mutations on the association of 14-3-3 with NF-L, all five mutants were constructed. The results of co-immunoprecipitation showed that mutants P8Q, P8L and P22S clearly inhibited 14-3-3 binding (Fig. 6A). The fluorescence result demonstrated that these CMT mutants formed huge aggregates in the cell body, in contrast to the small granules formed by wild-type NF-L (Fig. 6B). Since these CMT mutants affected 14-3-3 association and NF-L self-assembly, we then investigated their effect on the co-assembly with NF-M into neurofilaments. Fluorescence analysis of cells co-transfected with NF-M and CMT mutants revealed that the P8Q and P8R mutants retained the ability to co-assemble with NF-M into neurofilament networks, whereas the P8L, P22S and P22T mutants significantly inhibited this co-assembly (Fig. 6C). Statistical analysis showed that 91.0% of cells co-transfected with wild-type NF-L and NF-M had networks of co-assembly neurofilaments, and this value was decreased to 50.3% in the P8Q mutant and 63.7% in the P8R mutant; and the percentage of cells with neurofilament networks was significantly decreased to 19.7% in the P8L mutant, 16.9% in P22S, and P22T mutant completely inhibited formation of neurofilament network (Fig. 6D), which is consistent with the report that among the three mutations of the P8 residue in patients, a more serious phenotype is caused by P8L mutation (Jordanova et al., 2003). Interestingly, statistical analysis demonstrated that co-expression of 14-3-3 with CMT mutants reduced the formation of huge aggregates by CMT mutants (Fig. 6E,F). However, co-expression of 14-3-3 with CMT mutants and NF-M, failed to rescue the deficiency of CMT mutants to co-assembly with NF-M into neurofilament networks (supplementary material Fig. S3).

Fig. 6.

CMT NF-L mutations inhibit 14-3-3 interaction and result in aggregate formation and disruption of neurofilament assembly. (A) SW13 Cl2 Vim cells co-transfected with 14-3-3γ–GFP together with NF-L-WT, P8Q, P8R, P8L, P22S or P22T respectively were co-immunoprecipitated with rabbit anti-GFP antibody and subjected to western blotting. (B) Exogenous expression of NF-L-WT, P8Q, P8R, P8L, P22S or P22T in SW13 Cl2 Vim cells, and stained with anti-NF-L antibody. (C) Co-expression of NF-L-WT, P8Q, P8R, P8L, P22S or P22T together with NF-M in SW13 Cl2 Vim cells, and double-stained with rabbit anti-NF-L and mouse anti-NF-M antibodies. (D) Statistical analysis of neurofilament array formation of NF-M and NF-L or CMT NF-L mutants in SW13 Cl2 Vim cells. Values are means±s.e.m., n = 3. (E) SW13 Cl2 Vim cells were co-transfected with CMT NF-L mutants together with control empty vector or HA–14-3-3, and stained with anti-NF-L and anti-HA antibodies. (F) Statistical analysis of aggregate formation of CMT NF-L mutants in control group and 14-3-3 overexpression group. Values are means±s.e.m., n = 3. Scale bar: 10 µm. WT, wild-type; ctrl, control.

Fig. 6.

CMT NF-L mutations inhibit 14-3-3 interaction and result in aggregate formation and disruption of neurofilament assembly. (A) SW13 Cl2 Vim cells co-transfected with 14-3-3γ–GFP together with NF-L-WT, P8Q, P8R, P8L, P22S or P22T respectively were co-immunoprecipitated with rabbit anti-GFP antibody and subjected to western blotting. (B) Exogenous expression of NF-L-WT, P8Q, P8R, P8L, P22S or P22T in SW13 Cl2 Vim cells, and stained with anti-NF-L antibody. (C) Co-expression of NF-L-WT, P8Q, P8R, P8L, P22S or P22T together with NF-M in SW13 Cl2 Vim cells, and double-stained with rabbit anti-NF-L and mouse anti-NF-M antibodies. (D) Statistical analysis of neurofilament array formation of NF-M and NF-L or CMT NF-L mutants in SW13 Cl2 Vim cells. Values are means±s.e.m., n = 3. (E) SW13 Cl2 Vim cells were co-transfected with CMT NF-L mutants together with control empty vector or HA–14-3-3, and stained with anti-NF-L and anti-HA antibodies. (F) Statistical analysis of aggregate formation of CMT NF-L mutants in control group and 14-3-3 overexpression group. Values are means±s.e.m., n = 3. Scale bar: 10 µm. WT, wild-type; ctrl, control.

Previous studies demonstrated that 14-3-3 interacts with several IF proteins such as keratin, vimentin and GFAP (Ku et al., 1998; Tzivion et al., 2000; Kim et al., 2006; Li et al., 2006). However, the keratins are Type I/II IFs, while vimentin and GFAP are Type III. Here, we report that 14-3-3 proteins interact with a Type IV IF, NF-L, which is the core component of neurofilaments.

Specific interaction between 14-3-3 proteins and NF-L

Seven isoforms of 14-3-3 are expressed in brain (Moore et al., 1968), named 14-3-3 β, γ, ε, ζ, η, θ and σ. GST–14-3-3 binding assays showed that all seven isoforms specifically interacted with NF-L, but not NF-M or NF-H (Fig. 1A). NF-L associated with GST–14-3-3 was in the soluble state, and it is thought that soluble IFs take the form of tetramers (Herrmann and Aebi, 1998). Although NF-M and NF-H are known to co-assemble with NF-L into neurofilaments, GST–14-3-3 binding assays suggested that these soluble NF-L tetramers associated with 14-3-3 were homopolymeric, explaining the specific binding of 14-3-3 to NF-L, but not NF-M or NF-H. There was another possibility that NF-L co-assembled into heteropolymers with vimentin or GFAP in brain, and thus the interaction with 14-3-3 was dependent on vimentin or GFAP, which were shown to associate with 14-3-3 in previous studies. To rule out this possibility, further co-immunoprecipitation assays were carried out in the SW13 Cl2 Vim cell line with no endogenous cytoplasmic IFs. Results showed that all seven isoforms of 14-3-3 still interacted with NF-L, independent of vimentin or GFAP (Fig. 1D). Although the seven isoforms are highly homologous, GST–14-3-3 binding assays and co-immunoprecipitation assays showed that different isoforms showed differential associations with NF-L. These assays demonstrated that 14-3-3γ had the strongest interaction. The differences in association may be due to competition between NF-L and other favored binding targets of each 14-3-3 isoform, since they are important signal transduction adaptor proteins with many binding targets. Previous studies also reported competitive interactions between 14-3-3 and IF proteins as well as other signal transduction proteins (Tzivion et al., 2000; Ku et al., 2002; Kim et al., 2006; Margolis et al., 2006).

14-3-3 association depends on head domain phosphorylation of NF-L

14-3-3 proteins usually interact with phosphorylated targets. Two common 14-3-3 binding motifs are reported: RSXpSXP and RXXXpSXP (Muslin et al., 1996; Yaffe et al., 1997); pS represents the phosphorylation site of target proteins. So far, reported interactions between 14-3-3 and IF proteins are all dependent on phosphorylation of target the IFs, including keratin18 (Liao and Omary, 1996), vimentin (Tzivion et al., 2000), GFAP (Li et al., 2006) and keratin17 (Kim et al., 2006). Our study also demonstrated the requirement of head domain phosphorylation for interaction between 14-3-3 and NF-L (Fig. 3). However, among these binding targets, only Thr9 and Ser44 residues of keratin17 have been shown to affect 14-3-3 association, consistent with the predicted 14-3-3 binding motifs, although they do not exactly match the two classical 14-3-3 binding motifs (Kim et al., 2006). The reported Ser33 residue of keratin18 and the Ser8 residue of GFAP and vimentin do not match or contain classical 14-3-3 binding motifs. In our study, co-immunoprecipitation assays showed that several serine residues were necessary for 14-3-3 association (Fig. 4A). Among these residues, only Ser43 was the site of a predicted 14-3-3 binding motif of the NF-L head domain, which still partially matched the classical 14-3-3 binding motifs. None of the remaining residues matched 14-3-3 binding motifs. Notably, Ser57, another predicted 14-3-3 binding site and simultaneously a phosphorylation hot-spot of the NF-L head domain, had no effect on the interaction between 14-3-3 and NF-L (Fig. 4A). Together, the lack of classical 14-3-3 binding motifs in these 14-3-3-associated IF proteins suggested that the interaction between 14-3-3 and IF proteins is weak, unlike signal transduction proteins such as Raf-1 (Fu et al., 1994; Muslin et al., 1996), BAD (Zha et al., 1996) and Cdc25C (Peng et al., 1997), which contain classical 14-3-3 binding motifs and show strong interaction with 14-3-3.

Regulation of interaction between 14-3-3 and NF-L

It is reported that the head domain phosphorylation of NF-L is regulated by phosphatase-2A (Sacher et al., 1994; Giasson et al., 1996), and okadaic acid (OA) treatment, which specifically inhibits phosphatase-2A activity, increases the head domain phosphorylation of NF-L (Sacher et al., 1994). Since interaction between 14-3-3 and NF-L was dependent on phosphorylation, OA treatment markedly increased the association between 14-3-3 and NF-L (Fig. 3G), which was probably due to the increase phosphorylation level of the NF-L head domain by the inhibitory activity of phosphatase-2A. There are seven PKA phosphorylation sites at the head domain of NF-L: Ser2, 12, 41, 43, 49, 55 and 62 (Giasson et al., 1996; Cleverley et al., 1998; Nakamura et al., 2000). Our study revealed that mutation of Ser41, 43, 49, 55 and 62 had an evident effect on reducing the association between 14-3-3 and NF-L (Fig. 4A), indicating the role of PKA in the regulation of 14-3-3 binding. Further assays of PKA inhibitor and activator treatment confirmed this hypothesis (Fig. 4D). Mutation of Ser26, a phosphorylation site of Rho kinase (Hashimoto et al., 1998), also decreased 14-3-3 association (Fig. 4A), indicating that other kinases are also involved in the regulation of 14-3-3 association. The phosphorylation level of the NF-L head domain is regulated by a series of kinases and phosphatases, which results in changeable 14-3-3 associations. To some extent, binding of 14-3-3 to specific phosphorylation sites may prevent dephosphorylation of NF-L by phosphatase. Since many serine phosphorylation sites at the head domain of NF-L were involved in 14-3-3 association, mutation of only one or even several serine phosphorylation sites resulted only in partial reduction of 14-3-3 association (Fig. 3E; Fig. 4A). In fact, only mutation of all serines of the NF-L head domain completely disrupted 14-3-3 binding (Fig. 3E,G). Therefore, in terms of each functional phosphorylation site alone, the interaction with 14-3-3 is not strong, but the many phosphorylation sites of the NF-L head domain substantially enhance the association of 14-3-3, which also complicates the regulation of interaction between 14-3-3 and NF-L.

Role of 14-3-3 in regulation of NF-L dynamic assembly

Transmission electron microscopy (TEM) shows that NF-L self-assembles into 10-nm filaments (Nakagawa et al., 1995; Chen et al., 2000), however, in this study, no filament structure was observed under fluorescence microscopy when expressed NF-L alone in SW13 Cl2 Vim cells. NF-L alone showed the morphology of small granules diffused throughout the cytoplasm in SW13 Cl2 Vim cells (Fig. 2A; supplementary material Fig. S1B). According to our TEM result, these granules are tangles of short 10-nm filaments (supplementary material Fig. S1B). However, co-transfected NF-L and NF-M can co-assembled into clear neurofilament networks (Fig. 2B; Fig. 3B; supplementary material Fig. S1D). It is suggested that these co-assembled neurofilaments seen under fluorescence microscopy are parallel bundle of 10-nm filaments, just like the phenomenon in Sf9 cells reported in previous studies (Nakagawa et al., 1995; Chen et al., 2000).

The disassembly of neurofilament network due to 14-3-3 overexpression (Fig. 5D) and the association of 14-3-3 with soluble NF-L subunits (supplementary material Fig. S4) together indicate the disassembly role of 14-3-3. Therefore, on one hand, 14-3-3 associates with the soluble NF-L subunits to inhibit assembly; on the other hand, 14-3-3 binds to phosphorylated neurofilaments and leads to neurofilament network disassembly. Since the regulation of IF disassembly is dependent on phosphorylation state, hyperphosphorylation of NF-L leads to the disassembly of neurofilaments (Giasson and Mushynski, 1998). Here, specific interaction of 14-3-3 proteins with phosphorylated NF-L subunits also indicated the role of 14-3-3 and NF-L phosphorylation in the disassembly of neurofilaments. What is more, binding of 14-3-3 to phosphorylated NF-L subunits may prevent the dephosphorylation of these subunits by phosphatases, maintaining the hyperphosphorylation state of the subunits, which facilitates the disassembly of neurofilaments. 14-3-3 specifically interacts with phosphorylated IF proteins, and many studies have revealed the role of 14-3-3 in IF disassembly. For example, during astrocyte mitosis, phosphorylation of GFAP and binding of 14-3-3 are followed by disassembly of glial filaments (Li et al., 2006); phosphorylation of the Ser33 site of keratin18 together with 14-3-3 binding during mitosis results in sequestration of keratin18 into the soluble pool, enhancing its solubility (Liao and Omary, 1996). It is reported that disruption of association between 14-3-3 and keratin18 results in the inhibition of keratin network disassembly induced by shear-stress and significantly retards FRAP recovery of keratin18 (Sivaramakrishnan et al., 2009). In our study, 14-3-3 overexpression not only resulted in neurofilament disassembly, but also increased the dynamic exchange rate of NF-L subunits (Fig. 5), indicating the important role of 14-3-3 in the regulation of NF-L assembly. And the co-localization data suggests that NF-L granules co-localized with 14-3-3 maybe the soluble portion of NF-L, whereas the independent NF-L granules without 14-3-3 association maybe the assemble 10-nm filaments (Fig. 2A). There are also fibrously distributed 14-3-3 co-localized with neurofilament bundles in SW13 Cl2 Vim cells co-transfected with NF-L and NF-M, and with neurofilaments in DRG neurons (Fig. 2B,C). These neurofilaments associated with 14-3-3 are possibly going to disassembly.

CMT NF-L mutants and 14-3-3

Mutations in NF-L cause CMT disease in humans. CMT patients with NF-L mutations are classified as CMT2E (Mersiyanova et al., 2000; De Jonghe et al., 2001). In our study, proline mutations of the NF-L head domain in CMT disease disrupted 14-3-3 association (Fig. 6A). It is possible that mutations of proline residues change the secondary and tertiary structure of the NF-L head domain, and as a result, inhibit 14-3-3 association, since this is dependent on the head domain phosphorylation of NF-L. There is another possibility, that proline mutations affect Ser/Thr phosphorylation of the NF-L head domain, therefore inhibiting phosphorylation-dependent 14-3-3 association; there is a report that Pro22 mutation of the NF-L head domain inhibits nearby Thr21 phosphorylation (Sasaki et al., 2006). Among five head domain proline mutations of NF-L (P8Q, P8R, P8L, P22S and P22T), mutations P8Q, P8L and P22S significantly inhibited 14-3-3 interaction (Fig. 6A). Interestingly, there is a correlation between the ability of CMT mutants to interact with 14-3-3 and the motor nerve conduction velocity (NCV). With regard to the mutants P8Q, P8R and P8L, a lower interaction with 14-3-3 (Fig. 6A), was correlate with a lower motor NCV in the CMT patients (Jordanova et al., 2003). This correlation suggests a role of 14-3-3 in the pathogenesis of CMT disease, due to abnormal regulation of NF-L assembly. These reported proline mutations all induced the formation of huge aggregates of NF-L (Fig. 6B). Furthermore, the P8L, P22S and P22T mutations even disrupted co-assembly with NF-M into neurofilament arrays (Fig. 6C). It is possible that disruption of 14-3-3 binding leads to the formation of aggregates. One of CMT patients' clinical symptoms is distal predominance of motor and sensory neurons. Since NF-L is an important component of axons, formation of NF-L aggregates has a negative effect on axon development. Therefore, prevention of NF-L aggregate formation may be an aspect of therapy for CMT disease.

In summary, this study reports the interactions between 14-3-3 proteins and NF-L, and the regulation of NF-L assembly and dynamic exchange by 14-3-3. The formation of NF-L aggregates in CMT patients with NF-L mutations may be due to inhibition of 14-3-3 binding to these mutants, result in abnormal neurofilament assembly. Our results suggest 14-3-3 proteins as potential molecular targets for human CMT disease therapy.

cDNA construction

All experiments on animals were performed according to the standards of Animal Facility of Peking University on Animal Care and National Institutes of Health guidelines. Seven isoforms of 14-3-3 cDNA were synthesized by RT-PCR using mRNA samples from mouse brain. The cDNA of NF-L and NF-M were kindly provided by Dr Nobutaka Hirokawa (University of Tokyo, Tokyo, Japan) (Nakagawa et al., 1995). NF-L deletion mutants (NF-L-ΔH (Δ1-93aa), NF-L-ΔT (Δ403-543aa), NF-LΔ1-40aa, NF-LΔ1-50aa, NF-LΔ1-60aa, NF-LΔ1-70aa and NF-LΔ42-98aa) and phosphorylation site mutants (S2A, S12A, S26A, S27A, S33A, S37A, S41A, S43A, S49A, S51A, S55A, S57A, S62A and S66A) were constructed by PCR amplification of NF-L cDNA. The NF-L cDNA, NF-L mutants and NF-M cDNA were subcloned into p3XFLAG-CMV-14 vector (Sigma-Aldrich, Inc.). NF-L cDNA was also subcloned into pEGFP-N3 vector (Clontech Laboratories, Inc.). The seven isoforms of 14-3-3 cDNA were inserted into pEGFP-N3 vector (Clontech Laboratories, Inc.). 14-3-3γ was also inserted into pHM6 vector (Roche Molecular Biochemicals) and pmRFP-N3 vector [enhanced GFP (EGFP) tag of pEGFP-N3 vector replaced with monomeric RFP (mRFP) tag].

Antibodies

GFP cDNA was subcloned into vector pET-28a (Novagen). His–GFP expressed in E. coli BL21 was purified and used as an antigen to immunize rabbits.

Antibodies were sourced as follows: mouse anti-NF-L (clone DA2; Abcam), rabbit anti-NF-L (a gift from Dr Nobutaka Hirokawa), mouse anti-NF-M (clone NN18; Sigma), rabbit anti-NF-H (a gift from Hirokawa), mouse anti-14-3-3γ (clone CG31; Millipore), rabbit anti-14-3-3γ (Santa Cruz Biotechnology), mouse anti-GFP (clone 1E4; Abcam), mouse anti-FLAG (clone M2; Sigma).

Cell culture and transfection

Cultures of DRG neurons were prepared as described previously (Okabe and Hirokawa, 1990). SW13 Cl2 Vim cell lines (a kind gift from Dr Robert M. Evans, University of Colorado Health Sciences Center, Denver, CO) were grown in Dulbecco's modified Eagle's medium (DMEM) (Gibco) with 10% fetal bovine serum (FBS) at 37°C in 5% CO2, and transfected with jetPEI (Polyplus transfection) for immunofluorescence experiments, or Lipofectamine 2000 reagent (Invitrogen) for immunoprecipitation experiments.

GST–14-3-3 binding assay

The seven isoforms of GST–14-3-3 fusion proteins were immobilized on glutathione–Sepharose 4B beads (Amersham) for binding assays. Adult mouse brains were homogenized in lysis buffer (50 mM Tris-HCl, pH 7.4, 150 mM NaCl, 1% Triton X-100, protease inhibitor cocktail; Roche). The homogenate was centrifuged twice at 12,000 g for 30 min and the supernatant was added to the affinity column. After washing with lysis buffer several times, the eluted samples were analyzed by western blotting.

Immunoprecipitation

Cells were washed twice with PBS and lysed in lysis buffer (50 mM Tris-HCl, pH 7.4, 150 mM NaCl, 1% Triton X-100, 2 mM EDTA, protease inhibitor cocktail). The lysate was centrifuged at 16,000 g for 30 min. The supernatant was mixed with antibody at 4°C overnight. Then 20 µl Protein A–Sepharose beads (Pharmacia) were added and the mixture was rotated at 4°C for 2 h. After washing three times with lysis buffer, the beads were eluted and boiled in 2×SDS sample buffer and subjected to western blot analysis.

Immunofluorescence

Cells grown on coverslips were washed with PBS, fixed in 4% paraformaldehyde for 15 min and permeabilized with 0.2% Triton X-100 for 10 min at room temperature. After blocking with 1% BSA for 30 min, cells were incubated with primary antibodies for 1 h at room temperature. The secondary antibodies were FITC, TRITC, Alexa-488- or Alexa-568-conjugated goat anti-mouse or -rabbit IgG. Cell samples were observed under an LSM-710 confocal laser-scanning microscope (Zeiss, Germany), a TCS SP2 confocal microscope (Leica, Germany), or a TH4-200 fluorescence microscope (Olympus, Japan).

Fluorescence resonance energy transfer (FRET)

The acceptor photobleaching method was used to measure FRET. FRET was carried out with the LSM-710 confocal microscope. The fluorescence of GFP-linked fusion protein (donor) and RFP-linked protein (acceptor) was recorded before and after bleaching of RFP. FRET efficiency was calculated by Zeiss FRET software. GFP-tagged NF-L and RFP-tagged 14-3-3γ were co-transfected into SW13 Cl2 Vim cells to detect direct interactions between 14-3-3γ and NF-L.

Fluorescence recovery after photobleaching (FRAP)

FRAP was carried out with the TCS SP2 confocal microscope. Phase-contrast images of cells were taken before and immediately after FRAP to ensure that there were no significant changes in cell shape or position. Bar-shaped regions were bleached using the line-scan function at 488 nm (100% power), and recovery of fluorescence was monitored (15% power) using the time-series function at 10 s for up to 100 s. SW13 Cl2 Vim cells were transfected with pEGFP-NF-L and pHM6-14-3-3γ simultaneously or with control pHM6 empty vector, to assess the effect of 14-3-3γ on NF-L dynamics.

OA, KT5720 and forskolin treatment

Transfected cells were treated with 100 nM OA (Calbiochem), 1 µM KT5720 (Calbiochem) or 15 µM forskolin (Sigma) for 2 h, then harvested for further experiments.

Measurements and statistical analysis

Pixel density of the bands from western blots was measured by Image J (NIH). The unpaired Student's t test was used to perform comparisons. P<0.05 was considered significant.

We are grateful to Robert M. Evans (University of Colorado Health Sciences Center, USA) for SW13 Cl2 Vim cell lines.

Author contributions

This study was conceived by J.T. and J.C.; L.M. designed the study, carried out experiments and analysed the data; J.L. and X.L. carried out experiments and provided conceptual advice; L.M. designed the figures and wrote the manuscript.

Funding

This work was supported by the National Natural Science Foundation of China (NSFC) [grant numbers 30770663, 31071177].

Brownlees
J.
,
Ackerley
S.
,
Grierson
A. J.
,
Jacobsen
N. J.
,
Shea
K.
,
Anderton
B. H.
,
Leigh
P. N.
,
Shaw
C. E.
,
Miller
C. C.
(
2002
).
Charcot-Marie-Tooth disease neurofilament mutations disrupt neurofilament assembly and axonal transport.
Hum. Mol. Genet.
11
,
2837
2844
.
Chen
J.
,
Nakata
T.
,
Zhang
Z.
,
Hirokawa
N.
(
2000
).
The C-terminal tail domain of neurofilament protein-H (NF-H) forms the crossbridges and regulates neurofilament bundle formation.
J. Cell Sci.
113
,
3861
3869
.
Chin
S. S.
,
Macioce
P.
,
Liem
R. K.
(
1991
).
Effects of truncated neurofilament proteins on the endogenous intermediate filaments in transfected fibroblasts.
J. Cell Sci.
99
,
335
350
.
Ching
G. Y.
,
Liem
R. K.
(
1993
).
Assembly of type IV neuronal intermediate filaments in nonneuronal cells in the absence of preexisting cytoplasmic intermediate filaments.
J. Cell Biol.
122
,
1323
1335
.
Cleverley
K. E.
,
Betts
J. C.
,
Blackstock
W. P.
,
Gallo
J. M.
,
Anderton
B. H.
(
1998
).
Identification of novel in vitro PKA phosphorylation sites on the low and middle molecular mass neurofilament subunits by mass spectrometry.
Biochemistry
37
,
3917
3930
.
De Jonghe
P.
,
Mersivanova
I.
,
Nelis
E.
,
Del Favero
J.
,
Martin
J. J.
,
Van Broeckhoven
C.
,
Evgrafov
O.
,
Timmerman
V.
(
2001
).
Further evidence that neurofilament light chain gene mutations can cause Charcot-Marie-Tooth disease type 2E.
Ann. Neurol.
49
,
245
249
.
Dequen
F.
,
Filali
M.
,
Larivière
R. C.
,
Perrot
R.
,
Hisanaga
S.
,
Julien
J. P.
(
2010
).
Reversal of neuropathy phenotypes in conditional mouse model of Charcot-Marie-Tooth disease type 2E.
Hum. Mol. Genet.
19
,
2616
2629
.
Fu
H.
,
Xia
K.
,
Pallas
D. C.
,
Cui
C.
,
Conroy
K.
,
Narsimhan
R. P.
,
Mamon
H.
,
Collier
R. J.
,
Roberts
T. M.
(
1994
).
Interaction of the protein kinase Raf-1 with 14-3-3 proteins.
Science
266
,
126
129
.
Giasson
B. I.
,
Mushynski
W. E.
(
1998
).
Intermediate filament disassembly in cultured dorsal root ganglion neurons is associated with amino-terminal head domain phosphorylation of specific subunits.
J. Neurochem.
70
,
1869
1875
.
Giasson
B. I.
,
Cromlish
J. A.
,
Athlan
E. S.
,
Mushynski
W. E.
(
1996
).
Activation of cyclic AMP-dependent protein kinase in okadaic acid-treated neurons potentiates neurofilament fragmentation and stimulates phosphorylation of Ser2 in the low-molecular-mass neurofilament subunit.
J. Neurochem.
66
,
1207
1213
.
Gibb
B. J.
,
Brion
J. P.
,
Brownlees
J.
,
Anderton
B. H.
,
Miller
C. C.
(
1998
).
Neuropathological abnormalities in transgenic mice harbouring a phosphorylation mutant neurofilament transgene.
J. Neurochem.
70
,
492
500
.
Gonda
Y.
,
Nishizawa
K.
,
Ando
S.
,
Kitamura
S.
,
Minoura
Y.
,
Nishi
Y.
,
Inagaki
M.
(
1990
).
Involvement of protein kinase C in the regulation of assembly-disassembly of neurofilaments in vitro.
Biochem. Biophys. Res. Commun.
167
,
1316
1325
.
Hashimoto
R.
,
Nakamura
Y.
,
Goto
H.
,
Wada
Y.
,
Sakoda
S.
,
Kaibuchi
K.
,
Inagaki
M.
,
Takeda
M.
(
1998
).
Domain- and site-specific phosphorylation of bovine NF-L by Rho-associated kinase.
Biochem. Biophys. Res. Commun.
245
,
407
411
.
Hashimoto
R.
,
Nakamura
Y.
,
Komai
S.
,
Kashiwagi
Y.
,
Tamura
K.
,
Goto
T.
,
Aimoto
S.
,
Kaibuchi
K.
,
Shiosaka
S.
,
Takeda
M.
(
2000
).
Site-specific phosphorylation of neurofilament-L is mediated by calcium/calmodulin-dependent protein kinase II in the apical dendrites during long-term potentiation.
J. Neurochem.
75
,
373
382
.
Herrmann
H.
,
Aebi
U.
(
1998
).
Intermediate filament assembly: fibrillogenesis is driven by decisive dimer-dimer interactions.
Curr. Opin. Struct. Biol.
8
,
177
185
.
Hirokawa
N.
,
Glicksman
M. A.
,
Willard
M. B.
(
1984
).
Organization of mammalian neurofilament polypeptides within the neuronal cytoskeleton.
J. Cell Biol.
98
,
1523
1536
.
Hisanaga
S.
,
Gonda
Y.
,
Inagaki
M.
,
Ikai
A.
,
Hirokawa
N.
(
1990
).
Effects of phosphorylation of the neurofilament L protein on filamentous structures.
Cell Regul.
1
,
237
248
.
Hisanaga
S.
,
Matsuoka
Y.
,
Nishizawa
K.
,
Saito
T.
,
Inagaki
M.
,
Hirokawa
N.
(
1994
).
Phosphorylation of native and reassembled neurofilaments composed of NF-L, NF-M, and NF-H by the catalytic subunit of cAMP-dependent protein kinase.
Mol. Biol. Cell
5
,
161
172
.
Jordanova
A.
,
De Jonghe
P.
,
Boerkoel
C. F.
,
Takashima
H.
,
De Vriendt
E.
,
Ceuterick
C.
,
Martin
J. J.
,
Butler
I. J.
,
Mancias
P.
,
Papasozomenos
S. C.
et al.  (
2003
).
Mutations in the neurofilament light chain gene (NEFL) cause early onset severe Charcot-Marie-Tooth disease.
Brain
126
,
590
597
.
Kim
S.
,
Wong
P.
,
Coulombe
P. A.
(
2006
).
A keratin cytoskeletal protein regulates protein synthesis and epithelial cell growth.
Nature
441
,
362
365
.
Kim
S. K.
,
Cho
S. M.
,
Lee
I. B.
,
Lee
Y. H.
,
Kang
J. H.
,
Choi
J. H.
,
Suh
P. G.
,
Chang
J. S.
(
2007
).
In vitro assay of neurofilament light chain self-assembly using truncated mutants.
J. Neurosci. Methods
161
,
199
204
.
Ku
N. O.
,
Liao
J.
,
Omary
M. B.
(
1998
).
Phosphorylation of human keratin 18 serine 33 regulates binding to 14-3-3 proteins.
EMBO J.
17
,
1892
1906
.
Ku
N. O.
,
Michie
S.
,
Resurreccion
E. Z.
,
Broome
R. L.
,
Omary
M. B.
(
2002
).
Keratin binding to 14-3-3 proteins modulates keratin filaments and hepatocyte mitotic progression.
Proc. Natl. Acad. Sci. USA
99
,
4373
4378
.
Lee
M. K.
,
Xu
Z.
,
Wong
P. C.
,
Cleveland
D. W.
(
1993
).
Neurofilaments are obligate heteropolymers in vivo.
J. Cell Biol.
122
,
1337
1350
.
Li
H.
,
Guo
Y.
,
Teng
J.
,
Ding
M.
,
Yu
A. C.
,
Chen
J.
(
2006
).
14-3-3gamma affects dynamics and integrity of glial filaments by binding to phosphorylated GFAP.
J. Cell Sci.
119
,
4452
4461
.
Liao
J.
,
Omary
M. B.
(
1996
).
14-3-3 proteins associate with phosphorylated simple epithelial keratins during cell cycle progression and act as a solubility cofactor.
J. Cell Biol.
133
,
345
357
.
Margolis
S. S.
,
Perry
J. A.
,
Forester
C. M.
,
Nutt
L. K.
,
Guo
Y.
,
Jardim
M. J.
,
Thomenius
M. J.
,
Freel
C. D.
,
Darbandi
R.
,
Ahn
J. H.
et al.  (
2006
).
Role for the PP2A/B56delta phosphatase in regulating 14-3-3 release from Cdc25 to control mitosis.
Cell
127
,
759
773
.
Mersiyanova
I. V.
,
Perepelov
A. V.
,
Polyakov
A. V.
,
Sitnikov
V. F.
,
Dadali
E. L.
,
Oparin
R. B.
,
Petrin
A. N.
,
Evgrafov
O. V.
(
2000
).
A new variant of Charcot-Marie-Tooth disease type 2 is probably the result of a mutation in the neurofilament-light gene.
Am. J. Hum. Genet.
67
,
37
46
.
Moore
B. W.
,
Perez
V. J.
,
Gehring
M.
(
1968
).
Assay and regional distribution of a soluble protein characteristic of the nervous system.
J. Neurochem.
15
,
265
272
.
Muslin
A. J.
,
Tanner
J. W.
,
Allen
P. M.
,
Shaw
A. S.
(
1996
).
Interaction of 14-3-3 with signaling proteins is mediated by the recognition of phosphoserine.
Cell
84
,
889
897
.
Nakagawa
T.
,
Chen
J.
,
Zhang
Z.
,
Kanai
Y.
,
Hirokawa
N.
(
1995
).
Two distinct functions of the carboxyl-terminal tail domain of NF-M upon neurofilament assembly: cross-bridge formation and longitudinal elongation of filaments.
J. Cell Biol.
129
,
411
429
.
Nakamura
Y.
,
Hashimoto
R.
,
Kashiwagi
Y.
,
Aimoto
S.
,
Fukusho
E.
,
Matsumoto
N.
,
Kudo
T.
,
Takeda
M.
(
2000
).
Major phosphorylation site (Ser55) of neurofilament L by cyclic AMP-dependent protein kinase in rat primary neuronal culture.
J. Neurochem.
74
,
949
959
.
Okabe
S.
,
Hirokawa
N.
(
1990
).
Turnover of fluorescently labelled tubulin and actin in the axon.
Nature
343
,
479
482
.
Peng
C. Y.
,
Graves
P. R.
,
Thoma
R. S.
,
Wu
Z.
,
Shaw
A. S.
,
Piwnica–Worms
H.
(
1997
).
Mitotic and G2 checkpoint control: regulation of 14-3-3 protein binding by phosphorylation of Cdc25C on serine-216.
Science
277
,
1501
1505
.
Perez–Olle
R.
,
Leung
C. L.
,
Liem
R. K.
(
2002
).
Effects of Charcot-Marie-Tooth-linked mutations of the neurofilament light subunit on intermediate filament formation.
J. Cell Sci.
115
,
4937
4946
.
Perez–Olle
R.
,
Jones
S. T.
,
Liem
R. K.
(
2004
).
Phenotypic analysis of neurofilament light gene mutations linked to Charcot-Marie-Tooth disease in cell culture models.
Hum. Mol. Genet.
13
,
2207
2220
.
Sacher
M. G.
,
Athlan
E. S.
,
Mushynski
W. E.
(
1994
).
Increased phosphorylation of the amino-terminal domain of the low molecular weight neurofilament subunit in okadaic acid-treated neurons.
J. Biol. Chem.
269
,
18480
18484
.
Sasaki
T.
,
Gotow
T.
,
Shiozaki
M.
,
Sakaue
F.
,
Saito
T.
,
Julien
J. P.
,
Uchiyama
Y.
,
Hisanaga
S.
(
2006
).
Aggregate formation and phosphorylation of neurofilament-L Pro22 Charcot-Marie-Tooth disease mutants.
Hum. Mol. Genet.
15
,
943
952
.
Sihag
R. K.
,
Nixon
R. A.
(
1991
).
Identification of Ser-55 as a major protein kinase A phosphorylation site on the 70-kDa subunit of neurofilaments. Early turnover during axonal transport.
J. Biol. Chem.
266
,
18861
18867
.
Sivaramakrishnan
S.
,
Schneider
J. L.
,
Sitikov
A.
,
Goldman
R. D.
,
Ridge
K. M.
(
2009
).
Shear stress induced reorganization of the keratin intermediate filament network requires phosphorylation by protein kinase C zeta.
Mol. Biol. Cell
20
,
2755
2765
.
Trimpin
S.
,
Mixon
A. E.
,
Stapels
M. D.
,
Kim
M. Y.
,
Spencer
P. S.
,
Deinzer
M. L.
(
2004
).
Identification of endogenous phosphorylation sites of bovine medium and low molecular weight neurofilament proteins by tandem mass spectrometry.
Biochemistry
43
,
2091
2105
.
Tzivion
G.
,
Luo
Z. J.
,
Avruch
J.
(
2000
).
Calyculin A-induced vimentin phosphorylation sequesters 14-3-3 and displaces other 14-3-3 partners in vivo.
J. Biol. Chem.
275
,
29772
29778
.
Yaffe
M. B.
,
Rittinger
K.
,
Volinia
S.
,
Caron
P. R.
,
Aitken
A.
,
Leffers
H.
,
Gamblin
S. J.
,
Smerdon
S. J.
,
Cantley
L. C.
(
1997
).
The structural basis for 14-3-3:phosphopeptide binding specificity.
Cell
91
,
961
971
.
Zha
J.
,
Harada
H.
,
Yang
E.
,
Jockel
J.
,
Korsmeyer
S. J.
(
1996
).
Serine phosphorylation of death agonist BAD in response to survival factor results in binding to 14-3-3 not BCL-X(L).
Cell
87
,
619
628
.

Supplementary information