Home TTBK2 T3290C mutation in spinocerebellar ataxia 11 interferes with ciliogenesis
Article Open Access

TTBK2 T3290C mutation in spinocerebellar ataxia 11 interferes with ciliogenesis

  • Ruiqing Luo , Xiaoxia Zeng , Ping Li , Shuai Hu and Xueliang Qi EMAIL logo
Published/Copyright: October 3, 2024
Become an author with De Gruyter Brill

Abstract

This study aimed to elucidate the impact of the TTBK2 T3290C mutation (MUT) associated with Spinocerebellar Ataxia 11 (SCA11) on TTBK2 expression, function, and ciliogenesis. Lymphocytes were isolated from peripheral blood samples of SCA11 family members with the MUT and healthy controls (wild-type, WT). HEK-293 cells transfected with either WT or MUT TTBK2 plasmids were used to assess the MUT’s impact on TTBK2 protein expression, enzymatic activity, and its binding to Cep164 protein. Mouse embryonic fibroblast cells transfected with WT or MUT TTBK2 plasmids examined the MUT’s effect on cilia formation. Clinically, there was no significant difference in the expression of TTBK2 between the SCA11 patients and healthy individuals. The TTBK2 T3290C MUT did not affect protein expression or enzymatic activity but did reduce ciliary formation in embryonic cells and decreased binding affinity to Cep164. Therefore, our data suggested that the TTBK2 T3290C MUT in SCA11 may impair ciliogenesis by weakening the interaction with Cep164.

1 Introduction

Spinocerebellar ataxia type 11 (SCA11) is an infrequent subtype of hereditary cerebellar ataxia exhibiting an autosomal dominant inheritance pattern. SCA11 is typified by a gradually progressive cerebellar ataxia, coupled with limb and gait ataxia, dysarthria, and oculomotor dysfunctions [1]. Globally, SCA11 is extremely rare, with only seven families reported to date [2]. The diagnosis of SCA11 predominantly relies on phenotypic presentation and the identification of pathogenic variants in the TTBK2 gene, which remains the sole genetic locus linked to SCA11 [3,4].

The TTBK2 gene, positioned on chromosome 15q15.2, encompasses 14 coding exons (with exon 1 being noncoding). It encodes a serine-threonine protein kinase, a member of the casein kinase 1 family [3]. The TTBK2 protein is ubiquitously expressed in adult tissues, predominantly in the brain and testes [5], where it demonstrates elevated expression and kinase activity. Notably, within the brain, TTBK2 is abundantly expressed across all regions, including the cerebellum, hippocampus, midbrain, and substantia nigra, albeit with lower levels in the cerebral cortex [3,6], underscoring its significance in neurological disorders.

Research indicates that mutations (MUTs) associated with SCA11 function as dominant negative alleles, producing a truncated protein (TTBK2SCA11) that disrupts the functionality of the full-length TTBK2 in ciliogenesis mediation [7]. Studies have elucidated the pivotal role of TTBK2 in initiating ciliogenesis, primarily through phosphorylation and/or protein interactions that govern the initiation, maintenance, stability, and transport of cilia [8,9,10]. Beyond its basal body localization, TTBK2 is also present at the ciliary transition zone, where it potentially regulates ciliogenesis homeostasis, thereby influencing ciliary length, stability, and intracellular transport. The findings of Bowie et al. [7] revealed that TTBK2 hypomorphic mutant cells exhibit reduced ciliary length and formation frequency, alongside perturbed trafficking of the Sonic Hedgehog pathway effector Smoothened and the structural regulator KIF7. Moreover, TTBK2 localizes to the maternal centriole, where it facilitates the removal of the negative regulatory factor (centriole curl protein 110) and the recruitment of positive regulatory factors and cargo carriers in cilia (intrabundle transport proteins 88, 140, and 81) [7,11]. This maternal centriole localization, mediated by Cep164, is crucial for ciliation initiation [12,13,14].

Our preliminary research identified a new heterozygous TTBK2 MUT, c.3290T>C (exon 15), which led to an amino acid exchange (p.Val1097Ala) [15]. Therefore, this study was based on previous research to explore the effects of TTBK2 MUT (c.3290T>C) on TTBK2 protein expression level, kinase activity, and interaction with Cep164 protein. By verifying the effect of TTBK2 MUT on ciliary formation, the molecular mechanism of TTBK2 heterozygous MUT-induced SCA11 is revealed, bringing hope for the treatment of SCA11. Furthermore, enhancing awareness of SCA11 onset is advantageous for early diagnosis and the selection of appropriate treatment strategies, ultimately improving patient’s quality of life.

2 Materials and methods

2.1 Sample collection

All subjects originated from the same Chinese family afflicted with SCA11, as depicted in Figure S1. The family spanned three generations and comprised a total of 26 individuals, 10 of whom had manifested the disease across these generations. Clinical data were obtained from the 10 affected family members (I2, II2, II3, II5, II7, III3, III6, III8, III9, and III15). Following the acquisition of informed consent, 4 ml of venous blood was collected from the proband (III3) as well as from other affected family members (III6, III8, III9, and III15, carrying the pathogenic variant) and unaffected individuals (III4, III5, III7, III10, III12, and III16, wild type). The blood specimens were placed in ethylenediamine tetraacetic acid-containing anticoagulant tubes for lymphocyte isolation and subsequent lymphocyte separation for further analysis.

2.2 Reverse transcription-polymerase chain reaction (RT-PCR) analysis

Total RNA was extracted from isolated lymphocytes or transfected HEK-293/mouse embryonic fibroblast (MEF) cells using TRIzol reagent. Subsequently, cDNA synthesis was performed using the 5× Prime Script RT Master Mix, followed by real-time quantitative polymerase chain reaction with the SYBR Premix Ex Taq State II kit. The primer sequence is as follows: TTBK2(F): 5′CTCCTCACAATCCAAAAACACC3′, TTBK2(R): 5′CTAGATGGTGAGGAACTAGACG3′, GAPDH(F): 5′GAAGGTGAAGGTCGGAGTC3′, and GAPDH(R): 5′GAAGATGGTGATGGGATTTC3′.

2.3 Western blotting and immunoprecipitation

The cellular protein was extracted and resolved using sodium dodecyl sulfate polyacrylamide gel electrophoresis. Subsequently, the proteins were transferred onto a polyvinylidene fluoride membrane, blocked with milk, and incubated overnight at 4°C with either anti-TTBK2 or anti-Moesin. The protein bands were visualized using an enhanced chemiluminescence reagent, and band intensities were quantified using Image Pro Plus software.

For co-immunoprecipitation (Co-IP) experiments, transfected cells were lysed, and protein concentrations were determined. Immunoprecipitation was then carried out using anti-IgG and anti-Myc antibodies. Subsequently, western blotting analysis was performed using anti-Myc or anti-Flag antibodies.

2.4 Cell transfection

The full-length complementary (c) DNA of human TTBK2 was synthesized and cloned into the expression vector pEGFP-N1 (Beijing Olinger Biotechnology Co., Ltd). The TTBK2 T3290C mutant plasmid was obtained from the same company. Transfection of full-length cDNA or mutant TTBK2 plasmids into cells was performed using Lipofectamine 3000 (Invitrogen, Carlsbad, CA, USA).

2.5 Isolation of MEF cells and induction of cilia formation

The MEFs were isolated from embryos of E10.5 or E12.5 and maintained them as previously described [16]. Subsequently, the isolated MEFs were transfected with wild-type and mutant (T3290C) TTBK2 plasmids using Lipofectamine 2000. Cilia formation was induced by shifting cells from 10 to 0.5% fetal bovine serum (FBS) and maintaining them under low serum conditions for 48 h.

2.6 TTBK2 enzyme activity

TTBK2 enzyme activity was assessed utilizing the Universal Kinase Assay Kit (Abcam, ab138879). Samples were prepared and processed following the manufacturer’s instructions. The procedure for conducting the ADP Assay is as follows: prepare a 50× ADP Sensor I stock solution by combining 50 µL of DMSO with the vial of ADP Sensor I. Subsequently, generate the ADP Sensor by mixing 50 µL of the 50× ADP Sensor I stock solution with the vial of ADP Sensor II. Then, add 20 µL of ADP Sensor Buffer and 10 µL of ADP Sensor to each well containing 20 µL of kinase reaction solution, resulting in a total ADP assay volume of 50 µL/well. Incubate the reaction mixture at room temperature for 30 min, and measure the fluorescence intensity using a fluorescence plate reader set at E x/E m = 540/590 nm (cutoff 570 nm).

2.7 Immunofluorescence

The transfected cells were cultured in a 24-well plate with low serum (0.5% FBS) for 48 h, followed by fixation in 4% paraformaldehyde for 30 min. Subsequently, the cells were permeabilized with 0.1% Triton X-100 for 15 min after washing with phosphate-buffered saline. After washing again, the cell was blocked by 5% bovine serum albumin for 2 h at 37°C. Next, the cells were then incubated overnight at 4°C with primary antibodies, ARL13b (66739-1-Ig; Proteintech, 1:200) and γ-tubulin (ab179503; Abcam, 1:500), followed by 1-h incubation at room temperature with secondary antibodies (CY3 R 1:50, FITC M 1:50). Finally, the cells were stained with DAPI in phosphate-buffered solution (PBS) for 10 min, washed with PBS, and fixed on a glass slide.

2.8 Statistical analysis

Data were analyzed using GraphPad Prism 7 software and presented as means ± standard deviation. Two groups of data were analyzed by Student’s t-test. For multiple comparisons, we used one-way analysis of variance and the Tukey honestly significant difference test. P  <  0.05 was considered significant.

  1. Ethical approval: The research related to human use has been complied with all the relevant national regulations and institutional policies and in accordance with the tenets of the Helsinki Declaration and has been approved by the authors’ institutional review board or equivalent committee. This study was approved by the Second Affiliated Hospital of Nanchang University.

  2. Informed consent: Informed consent has been obtained from all individuals included in this study.

3 Results

3.1 TTBK2T3290C MUT in SCA11 does not affect its mRNA and protein expression

To compare the expression levels of TTBK2 in individuals with pathogenic variants (mutant type, n = 5) and healthy controls (wild type, n = 6) within the SCA11 family, peripheral blood samples were collected for lymphocyte extraction. The mRNA and protein expression of TTBK2 were subsequently analyzed using RT-PCR and Western blot techniques. Statistical evaluation revealed no significant difference in TTBK2 mRNA expression between the patient group and healthy controls (Figure 1a, p = 0.8536). Similarly, protein expression analysis showed no significant difference between the two groups (Figure 1b, p = 0.8636). These findings suggested that the TTBK2 T3290C MUT in SCA11 does not impact its gene or protein expression.

Figure 1 
                  TTBK2 expression in SCA11 patients. Peripheral blood and lymphocytes were extracted from patients (pathogenic variant individuals, mutant types) and healthy individuals (healthy individuals, wild-type) in the SCA11 family. (a) The TTBK2 mRNA level was detected by RT-PCR. (b) The TTBK2 protein level was detected by western blotting (membrane tissue extension spike protein, Moesin as the control). Results are pooled from three independent experiments. Statistical comparison was performed by Student’s t-test. ns indicates no significance.
Figure 1

TTBK2 expression in SCA11 patients. Peripheral blood and lymphocytes were extracted from patients (pathogenic variant individuals, mutant types) and healthy individuals (healthy individuals, wild-type) in the SCA11 family. (a) The TTBK2 mRNA level was detected by RT-PCR. (b) The TTBK2 protein level was detected by western blotting (membrane tissue extension spike protein, Moesin as the control). Results are pooled from three independent experiments. Statistical comparison was performed by Student’s t-test. ns indicates no significance.

3.2 The SCA11-associated TTBK2T3290C MUT does not affect its protein expression and enzyme activity

To elucidate the impact of the SCA11-associated TTBK2 T3290C MUT on its expression and enzymatic function, we engineered wild-type and mutant (T3290C) TTBK2 plasmids and transfected them into HEK-293 cells. Non-transfected cells served as controls, while cells transfected with empty plasmids were treated as negative controls (NC). Subsequently, TTBK2 protein levels and kinase activity were assessed. Western blot analysis revealed that the SCA11-associated TTBK2 T3290C MUT did not alter its protein expression (Figure 2a, p = 0.6544). Data from the Universal Kinase Assay Kit indicated that the TTBK2 T3290C MUT did not influence TTBK2’s enzymatic activity (Figure 2b, p = 0.9194). These findings suggested that the SCA11-associated TTBK2 T3290C MUT does not affect its protein expression or functional activity.

Figure 2 
                  
                     TTBK2
                     T3290C mutation does not affect protein expression and enzyme activity. The empty, wild-type, and mutant (T3290C) TTBK2 plasmids were transfected into HEK-293 cells, and the untransfected cell as the control group. (a) The TTBK2 protein level was detected by western blotting. (b) The TTBK2 enzyme activity was measured by Universal Kinase Assay Kit. Results are pooled from three independent experiments. Statistical comparison was performed by Student’s t-test. ns indicates no significance.
Figure 2

TTBK2 T3290C mutation does not affect protein expression and enzyme activity. The empty, wild-type, and mutant (T3290C) TTBK2 plasmids were transfected into HEK-293 cells, and the untransfected cell as the control group. (a) The TTBK2 protein level was detected by western blotting. (b) The TTBK2 enzyme activity was measured by Universal Kinase Assay Kit. Results are pooled from three independent experiments. Statistical comparison was performed by Student’s t-test. ns indicates no significance.

3.3 TTBK2T3290C MUT reduces cilia formation in embryonic cells

Next, to investigate the effect of SCA11-associated TTBK2 T3290C MUT on MEF cell cilia formation, we isolated mouse embryonic MEF cells and transfected them with empty (NC), wild-type, and mutant TTBK2 plasmids. Immunofluorescence staining for the primary cilia marker ARL13B was utilized to assess cilia formation, with γ-Tubulin serving as a centrosomal marker (Figure 3a and b). Our findings demonstrated that MEF cells transfected with the wild-type TTBK2 plasmid exhibited a significantly greater ciliary length compared to those transfected with the NC empty plasmid (p = 0.0104). Conversely, MEF cells transfected with the TTBK2 T3290C mutant plasmid displayed shorter cilia than those transfected with the wild-type TTBK2 plasmid (p = 0.0108) (Figure 3c). These results indicated that the TTBK2 T3290C MUT impairs cilia formation in MEF cells.

Figure 3 
                  SCA11-associated TTBK2
                     T3290C mutation affects cilia formation in MEF cells. Mouse embryonic MEF cells were isolated and divided into three groups, transfected with empty (NC), wild-type, and mutant (T3290C) TTBK2 plasmid, respectively. Then, the cells were maintained under low serum conditions for 48 h to induce cilia formation. (a) and (b) Cilia were immunostained for ARL13b (green) to label cilia and γ-Tubulin (red) to label centrosomes, scar bar = 20 and 1 µm. (c) Quantification of the cilia length from MEFs. Results are pooled from three independent experiments. Statistical comparison was performed by Student’s t-test. *p < 0.05.
Figure 3

SCA11-associated TTBK2 T3290C mutation affects cilia formation in MEF cells. Mouse embryonic MEF cells were isolated and divided into three groups, transfected with empty (NC), wild-type, and mutant (T3290C) TTBK2 plasmid, respectively. Then, the cells were maintained under low serum conditions for 48 h to induce cilia formation. (a) and (b) Cilia were immunostained for ARL13b (green) to label cilia and γ-Tubulin (red) to label centrosomes, scar bar = 20 and 1 µm. (c) Quantification of the cilia length from MEFs. Results are pooled from three independent experiments. Statistical comparison was performed by Student’s t-test. *p < 0.05.

3.4 TTBK2T3290C MUT reduces its binding to Cep164 protein

The aforementioned study demonstrated that the SCA11-associated TTBK2 T3290C MUT does not impact TTBK2 protein expression or enzymatic activity, but it significantly impedes ciliogenesis. To elucidate the mechanism by which the SCA11-associated TTBK2 T3290C MUT influences ciliogenesis, we assessed the impact of the TTBK2 T3290C MUT on its interaction with the Cep164 protein using Co-IP assays. HEK-293 cells were transfected with MYC-Cep164 plasmids alongside either wild-type (Flag-TTBK2) or mutant (Flag-TTBK2 T3290C) plasmids. The findings revealed that the TTBK2 T3290C MUT diminished its binding affinity to the Cep164 protein (Figure 4).

Figure 4 
                  
                     TTBK2
                     T3290C mutation reduces its binding to Cep164 protein. HEK-293 cells were transfected with MYC-Cep164 plasmids and wild-type (Flag-TTBK2)/mutant (Flag-TTBK2
                     T3290C) plasmid. Pull-down assessment for interaction between TTBK2 and Cep164 in vitro.
Figure 4

TTBK2 T3290C mutation reduces its binding to Cep164 protein. HEK-293 cells were transfected with MYC-Cep164 plasmids and wild-type (Flag-TTBK2)/mutant (Flag-TTBK2 T3290C) plasmid. Pull-down assessment for interaction between TTBK2 and Cep164 in vitro.

4 Discussion

SCA11 is a very rare progressive degenerative disease. The diagnosis of SCA11 mainly depends on clinical features and identification of TTBK2 MUTs. Previously, we documented a Chinese SCA11 pedigree, wherein 10 affected individuals harbored a novel heterozygous TTBK2 MUT, c.3290T>C (exon 15), which results in an amino acid substitution (p.Val1097Ala) [15]. Although categorized as potentially benign, our prior studies [15] suggested that this variant aligns with the SCA11 diagnosis. Primary cilia exert a crucial regulatory influence on neurogenesis, influencing neuronal differentiation, proliferation, and migration during corticogenesis [17,18,19,20]. Hence, this study aimed to elucidate the impact of the TTBK2 T3290C MUT linked to SCA11 on TTBK2 expression, function, and ciliogenesis.

Research indicates that TTBK2 sustains primary cilia by inhibiting their breakdown, thereby fostering the proliferation of granule neural progenitor cells and modulating cerebellar development [21]. Pathological phosphorylation of TDP-43 by TTBK2 may precipitate neurodegeneration [22]. Meanwhile, numerous studies affirmed that TTBK2 is a crucial ciliogenesis regulator [23,24]. Bowie et al.’s [7] study demonstrated that SCA11-associated MUT disrupts the function of full-length TTBK2 in ciliogenesis mediation. Goetz et al. [25] proved that dominant truncating MUTs in human TTBK2 cause SCA11, and these mutant proteins do not promote ciliogenesis and inhibit ciliogenesis in wild-type cells. Consistent with the findings of Bowie et al. and Goetz et al., our study also observed that the TTBK2 T3290C MUT impeded ciliogenesis. The TTBK2 MUTs led to either nonsense-mediated decay of the mutant transcripts or the production of truncated TTBK2 proteins with varying lengths due to premature stop codons in the mRNA. The nonsense-mediated decay or the presence of truncated TTBK2 protein significantly diminished kinase activity and acted as a dominant negative allele, interfering with the function of the wild-type TTBK2 protein [3]. As reported by Bowie et al. [7], truncated proteins generated by SCA11-related TTBK2 MUT can disrupt the function of full-length TTBK2, thus affecting ciliogenesis. Our study found no difference in expression of TTBK2 in peripheral blood lymphocytes between patients (pathogenic variant individuals, mutant types) and healthy individuals (healthy individuals, wild type) in the SCA11 family. The TTBK2 T3290C MUT did not impact TTBK2 protein expression or enzyme activity, differing from Bouskila et al.’s findings, suggesting that TTBK2 T3290C MUT may interfere with cilia formation through alternative mechanisms.

Gene MUTs can affect protein–protein interactions, as evidenced by Liu et al. [26], who reported that the MPP2 N315 MUT exhibited enhanced binding affinity to ANXA2 compared to MPP2 K315. Cep164 is localized to the apical domain of the mother centriole, forming a molecular bridge between the mother centriole and the membrane biogenesis machinery essential for cilia initiation [27]. Previous studies identified the recruitment of TTBK2 by the distal appendage protein Cep164 as an early event in ciliogenesis, with Cep164 MUTs disrupting the Cep164-TTBK2 complex [12]. We have confirmed that the TTBK2 T3290C MUT diminished its binding affinity to the Cep164 protein, implying that TTBK2 T3290C MUT in SCA11 may impair cilia formation by reducing binding to Cep164. TTBK2 interacts with Cep164 via its proline-rich motif. Oda et al. [14] established that Cep164 binding is crucial for TTBK2’s role in promoting ciliogenesis. Further investigations revealed that TTBK2 can phosphorylate Cep164 and Cep97, inhibiting the interaction between Cep164 and Dishevelled-3, a key ciliogenesis regulator, in a kinase activity-dependent manner.

The MUT site (c.3290T>C, p.Val1097Ala, Figure S2) is located outside the protease active region, and our results demonstrated that the TTBK2 T3290C MUT did not impact TTBK2 protease activity. However, this MUT decreased TTBK2’s binding to the Cep164 protein. The mechanism by which TTBK2 T3290C MUT affects its interaction with Cep164 protein still needs further exploration. We hypothesize that the (c.3290T>C, p.Val1097Ala) MUT may induce secondary and tertiary structural alterations in the protein, thereby influencing TTBK2’s interaction with other proteins such as Cep164. This article evaluates the effects of MUTs on TTBK2 protein expression, enzyme activity, cilia formation, and binding to Cep164 protein through cellular assays. Future research will involve RNA sequencing analysis of wild-type and mutant TTBK2 in mouse cerebellar neurons to identify distinct functional pathways and elucidate the mechanisms underlying the onset and progression of the TTBK2 T3290C MUT.

5 Conclusion

In summary, this study corroborates the link between the TTBK2 T3290C MUT and SCA11. Our findings indicate no significant difference in TTBK2 expression levels between SCA11 patients and healthy controls. Although the TTBK2 T3290C MUT does not alter its protein expression or enzymatic activity, it impairs ciliogenesis via a mechanism involving altered interaction with the Cep164 protein. This research provides new theoretical insights into the pathogenic mechanisms of SCA11.


tel: +86-0791-86300624

  1. Funding information: This study was supported by the Natural Science Foundation of Jiangxi, China (No. 20202BABL206050) and the Scientific Research Project of Jiangxi Health Commission (No. 202130342).

  2. Author contributions: (1) Conception and design: RL; (2) administrative support: XQ; (3) experiments: all authors; (4) data analysis and interpretation: RL; (5) manuscript writing: RL and XQ; and (6) final approval of manuscript: all authors.

  3. Conflict of interest: Authors state no conflict of interest.

  4. Data availability statement: The datasets generated during and/or analyzed during the current study are available from the corresponding author on reasonable request.

References

[1] Gong Z, Lei L. Spinocerebellar ataxia type 11 (SCA11): An update. Eur J Neurosci. 2023;58:2623–40.10.1111/ejn.16054Search in Google Scholar PubMed

[2] Lu YQ, Chen JM, Huang YL, Zou ZY. A Novel TTBK2 mutation in a chinese pedigree with spinocerebellar ataxia 11. Cerebellum. 2024;23:1221–5.10.1007/s12311-023-01616-3Search in Google Scholar PubMed

[3] Houlden H, Johnson J, Gardner-Thorpe C, Lashley T, Hernandez D, Worth P, et al. Mutations in TTBK2, encoding a kinase implicated in tau phosphorylation, segregate with spinocerebellar ataxia type 11. Nat Genet. 2007;39:1434–6.10.1038/ng.2007.43Search in Google Scholar PubMed

[4] Bauer P, Stevanin G, Beetz C, Synofzik M, Schmitz-Hubsch T, Wullner U, et al. Spinocerebellar ataxia type 11 (SCA11) is an uncommon cause of dominant ataxia among French and German kindreds. J Neurol Neurosurg Psychiatry. 2010;81:1229–32.10.1136/jnnp.2009.202150Search in Google Scholar PubMed

[5] Bouskila M, Esoof N, Gay L, Fang E, Deak M, Begley M, et al. TTBK2 kinase substrate specificity and the impact of spinocerebellar-ataxia-causing mutations on expression, activity, localization and development. Biochem J. 2011;437:157–67.10.1042/BJ20110276Search in Google Scholar PubMed PubMed Central

[6] Bowie E, Goetz SC. TTBK2 and primary cilia are essential for the connectivity and survival of cerebellar Purkinje neurons. Elife. 2020;9:e51166.10.7554/eLife.51166Search in Google Scholar PubMed PubMed Central

[7] Bowie E, Norris R, Anderson KV, Goetz SC. Spinocerebellar ataxia type 11-associated alleles of Ttbk2 dominantly interfere with ciliogenesis and cilium stability. PLoS Genet. 2018;14:e1007844.10.1371/journal.pgen.1007844Search in Google Scholar PubMed PubMed Central

[8] Rosa ESI, Bino L, Johnson CM, Rutherford TJ, Neuhaus D, Andreeva A, et al. Molecular mechanisms underlying the role of the centriolar CEP164-TTBK2 complex in ciliopathies. Structure. 2022;30:114–28.e119.Search in Google Scholar

[9] Lo CH, Lin IH, Yang TT, Huang YC, Tanos BE, Chou PC, et al. Phosphorylation of CEP83 by TTBK2 is necessary for cilia initiation. J Cell Biol. 2019;218:3489–505.10.1083/jcb.201811142Search in Google Scholar PubMed PubMed Central

[10] Bernatik O, Pejskova P, Vyslouzil D, Hanakova K, Zdrahal Z, Cajanek L. Phosphorylation of multiple proteins involved in ciliogenesis by Tau Tubulin kinase 2. Mol Biol Cell. 2020;31:1032–46.10.1091/mbc.E19-06-0334Search in Google Scholar PubMed PubMed Central

[11] Sobu Y, Wawro PS, Dhekne HS, Yeshaw WM, Pfeffer SR. Pathogenic LRRK2 regulates ciliation probability upstream of tau tubulin kinase 2 via Rab10 and RILPL1 proteins. Proc Natl Acad Sci U S A. 2021;118:e2005894118.10.1073/pnas.2005894118Search in Google Scholar PubMed PubMed Central

[12] Rosa E, Silva I, Binó L, Johnson C, Rutherford T, Neuhaus D, et al. Molecular mechanisms underlying the role of the centriolar CEP164-TTBK2 complex in ciliopathies. Structure (London, England: 1993). 2022;30:114–28.e119.10.1016/j.str.2021.08.007Search in Google Scholar PubMed PubMed Central

[13] Cajanek L, Nigg EA. Cep164 triggers ciliogenesis by recruiting Tau tubulin kinase 2 to the mother centriole. Proc Natl Acad Sci U S A. 2014;111:E2841–50.10.1073/pnas.1401777111Search in Google Scholar PubMed PubMed Central

[14] Oda T, Chiba S, Nagai T, Mizuno K. Binding to Cep164, but not EB1, is essential for centriolar localization of TTBK2 and its function in ciliogenesis. Genes Cells. 2014;19:927–40.10.1111/gtc.12191Search in Google Scholar PubMed

[15] Deng Y, Fu J, Zhong Y, Zhang M, Qi X. First finding of familial spinal cerebellar Ataxia11 in China: clinical, imaging and genetic features. Neurol Sci. 2020;41:155–60.10.1007/s10072-019-04052-6Search in Google Scholar PubMed

[16] Ocbina PJ, Anderson KV. Intraflagellar transport, cilia, and mammalian Hedgehog signaling: analysis in mouse embryonic fibroblasts. Dev Dyn. 2008;237:2030–8.10.1002/dvdy.21551Search in Google Scholar PubMed PubMed Central

[17] Guo J, Otis JM, Higginbotham H, Monckton C, Cheng J, Asokan A, et al. Primary cilia signaling shapes the development of interneuronal connectivity. Dev Cell. 2017;42:286–300.e284.10.1016/j.devcel.2017.07.010Search in Google Scholar PubMed PubMed Central

[18] Chang CH, Zanini M, Shirvani H, Cheng JS, Yu H, Feng CH, et al. Atoh1 controls primary cilia formation to allow for SHH-triggered granule neuron progenitor proliferation. Dev Cell. 2019;48:184–99.e185.10.1016/j.devcel.2018.12.017Search in Google Scholar PubMed

[19] Coschiera A, Yoshihara M, Lauter G, Ezer S, Pucci M, Li H, et al. Primary cilia promote the differentiation of human neurons through the WNT signaling pathway. BMC Biol. 2024;22:48.10.1186/s12915-024-01845-wSearch in Google Scholar PubMed PubMed Central

[20] Stoufflet J, Chaulet M, Doulazmi M, Fouquet C, Dubacq C, Metin C, et al. Primary cilium-dependent cAMP/PKA signaling at the centrosome regulates neuronal migration. Sci Adv. 2020;6:eaba3992.10.1126/sciadv.aba3992Search in Google Scholar PubMed PubMed Central

[21] Lin IH, Li YR, Chang CH, Cheng YW, Wang YT, Tsai YS, et al. Regulation of primary cilia disassembly through HUWE1-mediated TTBK2 degradation plays a crucial role in cerebellar development and medulloblastoma growth. Cell Death Differ. 2024. 10.1038/s41418-024-01325-2.Search in Google Scholar PubMed PubMed Central

[22] Taylor LM, McMillan PJ, Liachko NF, Strovas TJ, Ghetti B, Bird TD, et al. Pathological phosphorylation of tau and TDP-43 by TTBK1 and TTBK2 drives neurodegeneration. Mol Neurodegener. 2018;13:7.10.1186/s13024-018-0237-9Search in Google Scholar PubMed PubMed Central

[23] Binó L, Čajánek L. Tau tubulin kinase 1 and 2 regulate ciliogenesis and human pluripotent stem cells-derived neural rosettes. Sci Rep. 2023;13:12884.10.1038/s41598-023-39887-9Search in Google Scholar PubMed PubMed Central

[24] Bashore F, Marquez A, Chaikuad A, Howell S, Dunn A, Beltran A, et al. Modulation of tau tubulin kinases (TTBK1 and TTBK2) impacts ciliogenesis. Sci Rep. 2023;13:6118.10.1038/s41598-023-32854-4Search in Google Scholar PubMed PubMed Central

[25] Goetz S, Liem K, Anderson K. The spinocerebellar ataxia-associated gene Tau tubulin kinase 2 controls the initiation of ciliogenesis. Cell. 2012;151:847–58.10.1016/j.cell.2012.10.010Search in Google Scholar PubMed PubMed Central

[26] Liu X, Meng J, Liao X, Liu Y, Zhou Q, Xu Z, et al. A de novo missense mutation in MPP2 confers an increased risk of Vogt-Koyanagi-Harada disease as shown by trio-based whole-exome sequencing. Cell Mol Immunol. 2023;20:1379–92.10.1038/s41423-023-01088-9Search in Google Scholar PubMed PubMed Central

[27] Schmidt KN, Kuhns S, Neuner A, Hub B, Zentgraf H, Pereira G. Cep164 mediates vesicular docking to the mother centriole during early steps of ciliogenesis. J Cell Biol. 2012;199:1083–101.10.1083/jcb.201202126Search in Google Scholar PubMed PubMed Central

Received: 2024-05-28
Revised: 2024-08-28
Accepted: 2024-09-09
Published Online: 2024-10-03

© 2024 the author(s), published by De Gruyter

This work is licensed under the Creative Commons Attribution 4.0 International License.

Articles in the same Issue

  1. Research Articles
  2. Brain expression profiles of two SCN1A antisense RNAs in children and adolescents with epilepsy
  3. Silibinin suppresses glioblastoma cell growth, invasion, stemness, and glutamine metabolism by YY1/SLC1A5 pathway
  4. Early exercise intervention promotes myelin repair in the brains of ischemic rats by inhibiting the MEK/ERK pathway
  5. Comparative analysis of CRASH and IMPACT in predicting the outcome of 340 patients with traumatic brain injury
  6. Association between FOXP3 polymorphisms and expression and neuromyelitis optica spectrum disorder risk in the Northern Chinese Han population
  7. Trehalose improves the movement ability of Aβarc Drosophila by restoring the damaged mitochondria
  8. The ACE2/Ang-(1-7)/MasR axis alleviates brain injury after cardiopulmonary resuscitation in rabbits by activating PI3K/Akt signaling
  9. Single cocaine exposure attenuates the intrinsic excitability of CRH neurons in the ventral BNST via Sigma-1 receptors
  10. Effect of dopamine on limbic network connectivity at rest in Parkinson’s disease patients with freezing of gait
  11. FT4-to-FT3 ratio is a novel prognostic marker in subacute combined spinal cord degeneration patients
  12. Suanzaoren decoction exerts its antidepressant effect via the CaMK signaling pathway
  13. Acute ischemic STROKE – from laboratory to the Patient’s BED (STROKELABED): A translational approach to reperfusion injury. Study Protocol
  14. Thyroid hormone T3 induces Fyn modification and modulates palmitoyltransferase gene expression through αvβ3 integrin receptor in PC12 cells during hypoxia
  15. Activating α7nAChR suppresses systemic inflammation by mitigating neuroinflammation of the medullary visceral zone in sepsis in a rat model
  16. Amelioration of behavioral and histological impairments in somatosensory cortex injury rats by limbal mesenchymal stem cell transplantation
  17. TTBK2 T3290C mutation in spinocerebellar ataxia 11 interferes with ciliogenesis
  18. In a rodent model of autism, probiotics decrease gut leakiness in relation to gene expression of GABA receptors: Emphasize how crucial the gut–brain axis
  19. A data science approach to optimize ADHD assessment with the BRIEF-2 questionnaire
  20. Cystatin C alleviates unconjugated bilirubin-induced neurotoxicity by promoting bilirubin clearance from neurocytes via exosomes, dependent on hepatocyte UGT1A1 activity
  21. Macrophage accumulation in dorsal root ganglion is associated with neuropathic pain in experimental autoimmune neuritis
  22. Identifying key biomarkers and therapeutic candidates for post-COVID-19 depression through integrated omics and bioinformatics approaches
  23. The hidden link: Investigating functional connectivity of rarely explored sub-regions of thalamus and superior temporal gyrus in Schizophrenia
  24. A pilot evaluation of the diagnostic accuracy of ChatGPT-3.5 for multiple sclerosis from case reports
  25. Review Articles
  26. Adaptation of the layer V supraspinal motor corticofugal projections from the primary (M1) and premotor (PM) cortices after CNS motor disorders in non-human primates: A survey
  27. Comorbidity in spinal cord injury in Iran: A narrative review
  28. Lipid-based nanoparticles for drug delivery in Parkinson’s disease
  29. Disgust sensitivity and psychopathic behavior: A narrative review
  30. Rapid Communications
  31. Long COVID elevated MMP-9 and release from microglia by SARS-CoV-2 Spike protein
  32. Internal consistency of the Mental Health Professional Culture Inventory: A pilot study in Romanian population
  33. Retraction
  34. Retraction of “Effect of C-phycocyanin on HDAC3 and miRNA-335 in Alzheimer’s disease”
  35. Corrigendum
  36. Corrigendum to “The ACE2/Ang-(1-7)/MasR axis alleviates brain injury after cardiopulmonary resuscitation in rabbits by activating PI3K/Akt signaling”
  37. Corrigendum to “Tongxinluo promotes axonal plasticity and functional recovery after stroke”
Downloaded on 10.9.2025 from https://www.degruyterbrill.com/document/doi/10.1515/tnsci-2022-0353/html
Scroll to top button