The mitotic spindle protein SPAG5/Astrin connects to the Usher protein network postmitotically

Ferry Fj Kersten, Erwin van Wijk, Lisette Hetterschijt, Katharina Bauβ, Theo A Peters, Mariam G Aslanyan, Bert van der Zwaag, Uwe Wolfrum, Jan Ee Keunen, Ronald Roepman, Hannie Kremer, Ferry Fj Kersten, Erwin van Wijk, Lisette Hetterschijt, Katharina Bauβ, Theo A Peters, Mariam G Aslanyan, Bert van der Zwaag, Uwe Wolfrum, Jan Ee Keunen, Ronald Roepman, Hannie Kremer

Abstract

Background: Mutations in the gene for Usher syndrome 2A (USH2A) are causative for non-syndromic retinitis pigmentosa and Usher syndrome, a condition that is the most common cause of combined deaf-blindness. To gain insight into the molecular pathology underlying USH2A-associated retinal degeneration, we aimed to identify interacting proteins of USH2A isoform B (USH2AisoB) in the retina.

Results: We identified the centrosomal and microtubule-associated protein sperm-associated antigen (SPAG)5 in the retina. SPAG5 was also found to interact with another previously described USH2AisoB interaction partner: the centrosomal ninein-like protein NINLisoB. Using In situ hybridization, we found that Spag5 was widely expressed during murine embryonic development, with prominent signals in the eye, cochlea, brain, kidney and liver. SPAG5 expression in adult human tissues was detected by quantitative PCR, which identified expression in the retina, brain, intestine, kidney and testis. In the retina, Spag5, Ush2aisoB and NinlisoB were present at several subcellular structures of photoreceptor cells, and colocalized at the basal bodies.

Conclusions: Based on these results and on the suggested roles for USH proteins in vesicle transport and providing structural support to both the inner ear and the retina, we hypothesize that SPAG5, USH2AisoB and NINLisoB may function together in microtubule-based cytoplasmic trafficking of proteins that are essential for cilium formation, maintenance and/or function.

Figures

Figure 1
Figure 1
Interactions between sperm-associated antigen (SPAG)5 and Usher syndrome 2A isoform B (USH2AisoB). (A) Schematic protein structures of SPAG5 and the peptides encoded by deletion constructs. Numbers represent amino acids (aa; NP_006452). Protein fragments encoded by deletion constructs of SPAG5 and the intracellular domain (ICD) of USH2AisoB were used in a yeast two-hybrid assay, which identified a specific interaction between USH2AisoB (aa 5064 to 5196) and the C-terminal two coiled-coil domains (aa 973to 1193) of SPAG5. (B) Glutathione S-transferase (GST) pull-down assays showing that Flag-tagged SPAG5 is efficiently pulled down by GST-USH2AisoBICD, but not by GST alone, as detected by an anti-Flag antibody. The first lane shows 5% of the input of COS-1 cell lysate. (C) Hemagluttinin (HA)-SPAG5 aa 973-1193 from COS-1 lysates co-immunoprecipitated with GFP-fused USH2AisoBICD, but not with an unrelated HA-tagged protein (EPS8). As a positive control, HA-whirlin co-immunoprecipitated with green fluorescent protein (GFP)-fused USH2AisoBICD. The upper protein band in the middle blot represents the heavy chain of the anti-GFP antibody.
Figure 2
Figure 2
Validation of the interaction between sperm-associated antigen (SPAG)5 and ninein-like protein isoform B (NINLisoB) interaction. (A) The schematic protein structure of NINLisoB and the predicted intermediate filament domain (IF; aa 656-925), encoded by a deletion construct. A liquid β-galactosidase assay identified a specific interaction between SPAG5 (aa 973-1193) and the intermediate filament (IF) domain of NINLisoB. (B) Glutathione S-transferase (GST) pull-down assay showing that Flag-tagged NINLisoB was efficiently pulled down by GST-SPAG5 (aa 973-1193), but not by GST alone, as detected by an anti-Flag antibody. The first lane shows 5% of the protein input. (C) Flag-NINLisoB co-immunoprecipitated with full-length HA-SPAG5 and the positive control HA-USH2AisoB ICD, but not with the HA-tagged unrelated protein MDA5.
Figure 3
Figure 3
Expression of the gene sperm-associated antigen (SPAG)5 in human adult tissues and Spag5 expression during murine development. (A) Using quantitative PCR, SPAG5 expression was detected in the human retina, brain, duodenum, kidney and testis. (B-M). RNA in situ hybridization of Spag5 mRNA in mouse embryos at embryonic day (E)12.5-E18.5 and mouse eyes at postnatal day (P)7. (B-D) Spag5 was widely expressed during murine development (E12.5 to E16.5), with the most intense signals in the following structures (numbers and arrowheads) ventricular zone of forebrain (1), midbrain (2), liver (3), lung (4), kidney (5), adrenal gland (6), thymus gland (7), submandibular salivary gland (8), limbs (9) and eye (10). (E) Expression was also seen in the cochlea, primordial vibrissal follicles, upper and lower jaw, intestine, heart, olfactory epithelium and testis (data not shown). (D) From E16.5 onwards, expression was mainly detected in the eye, brain, kidney, lung, thymus, primordial vibrissal follicles and in the liver (data not shown). (F-K) A strong signal for Spag5 was seen in the eye during embryonic development E12.5-E18.5 in the neuroblastic layer of the retina. At (I) E16.5 and (K) E18.5, Spag5 expression was present in the developing inner nuclear layer (INL; 11) and, (L-M) although much weaker, in the ganglion cell layer (GCL; 12), and this was maintained at postnatal day 7.
Figure 4
Figure 4
Subcellular localization of sperm-associated antigen (Spag)5 in retina cryosections of adult (postnatal day (P)20) rats. (A-C) Co-immunostaining using(A) anti-SPAG5 (green) and (B) anti-pan-centrin (red, a marker for the connecting cilium, basal body and centrioles) antibodies. (C) Colocalization (C; yellow) was detected at the (Ci) basal body (BB) and (Cii) in one of the two centrioles of the inner nuclear layer (INL). (A,C) Centriole staining is also visible in the ganglion cell layer (GCL). In addition, weaker Spag5 labeling was detected at the outer limiting membrane (OLM; filled arrowhead), the outer plexiform layer (OPL), at the distal part of the connecting cilium (CC) and at the accessory centriole. (A-Ci-ii).High-magnification fluorescence microscopy of the BB/CC region and the INL.
Figure 5
Figure 5
Localization of sperm-associated antigen (Spag)5 by immunoelectron microscopy. Electron micrographs of pre-embedded anti-SPAG5 labeling in (A,B) longitudinal and (C) cross-sections of mouse rod photoreceptor cells. Spag5 was detected (A-B) around the basal body (BB), (A-C) in the collar-like extension (CE) of the apical inner segment; and (A) in some photoreceptors at the region of the rootlet (R). CC = connecting cilium; OS = outer segment; IS = inner segment. Scale bars: 0.5 μm.
Figure 6
Figure 6
Colocalization of sperm-associated antigen (Spag)5 and isoform B of Ush2a (Ush2aisoB) and ninein-like (NINLisoB) proteins in the retina of young adult (P20) rat. Co-immunostaining of Spag5 and Ush2aisoB in retinal cryosections using (A) anti-SPAG5 antibodies (red) and (B) anti-USH2AisoB antibodies (green) show (C) partial colocalization (yellow) at the basal body. Co-immunostaining using (D) anti-SPAG5 antibodies (green) and (E) anti-NINLisoB antibodies (red). (F) A partial colocalization (yellow) was detected at the basal body. In addition, both antibodies labeled (Di,Ei) the ciliary rootlets, which was confirmed by (I) co-staining (yellow) using (G) anti-NINLisoB (green) and (H) anti-rootletin (red) antibodies.
Figure 7
Figure 7
Specificity of anti-sperm-associated antigen (SPAG)5 antibodies. (A) Co-immunostaining of SPAG5 (green) and Usher syndrome 2A (USH2A; red) in retinal cryosections of adult (P20) rat using anti-USH2A and anti-SPAG5 antibodies. Both antibodies were blocked together with the SPAG5 antigen and after pre-absorption no specific SPAG5 signals were detected in contrast to the normal USH2A staining. (A') Western blot analysis of HA-tagged SPAG5 using (pre-absorbed) anti-hemagglutinin (HA) and anti-SPAG5 antibodies. After pre-absorption of the antibodies with the SPAG5 antigen, HA-SPAG5 could only be detected by anti-HA antibodies and not by the blocked anti-SPAG5 antibodies, indicating the specificity of the antibodies.

References

    1. Kaiserman N, Obolensky A, Banin E, Sharon D. Novel USH2A mutations in Israeli patients with retinitis pigmentosa and Usher syndrome type 2. Arch Ophthalmol. 2007;125:219–224. doi: 10.1001/archopht.125.2.219.
    1. Liu X, Tang Z, Li C, Yang K, Gan G, Zhang Z, Liu J, Jiang F, Wang Q, Liu M. Novel USH2A compound heterozygous mutations cause RP/USH2 in a Chinese family. Mol Vis. 2010;16:454–461.
    1. McGee TL, Seyedahmadi BJ, Sweeney MO, Dryja TP, Berson EL. Novel mutations in the long isoform of the USH2A gene in patients with Usher syndrome type II or non-syndromic retinitis pigmentosa. J Med Genet. 2010;47:499–506. doi: 10.1136/jmg.2009.075143.
    1. Rivolta C, Sweklo EA, Berson EL, Dryja TP. Missense mutation in the USH2A gene: association with recessive retinitis pigmentosa without hearing loss. Am J Hum Genet. 2000;66:1975–1978. doi: 10.1086/302926.
    1. Smith RJ, Berlin CI, Hejtmancik JF, Keats BJ, Kimberling WJ, Lewis RA, Moller CG, Pelias MZ, Tranebjaerg L. Clinical diagnosis of the Usher syndromes. Usher Syndrome Consortium. Am J Med Genet. 1994;50:32–38. doi: 10.1002/ajmg.1320500107.
    1. Hartong DT, Berson EL, Dryja TP. Retinitis pigmentosa. Lancet. 2006;368:1795–1809. doi: 10.1016/S0140-6736(06)69740-7.
    1. Cohen M, Bitner-Glindzicz M, Luxon L. The changing face of Usher syndrome: clinical implications. Int J Audiol. 2007;46:82–93. doi: 10.1080/14992020600975279.
    1. Adato A, Michel V, Kikkawa Y, Reiners J, Alagramam KN, Weil D, Yonekawa H, Wolfrum U, El Amraoui A, Petit C. Interactions in the network of Usher syndrome type 1 proteins. Hum Mol Genet. 2005;14:347–356.
    1. Kremer H, van Wijk E, Marker T, Wolfrum U, Roepman R. Usher syndrome: molecular links of pathogenesis, proteins and pathways. Hum Mol Genet. 2006;15(Spec No 2):R262–R270.
    1. Maerker T, van Wijk E, Overlack N, Kersten FFJ, McGee J, Goldmann T, Sehn E, Roepman R, Walsh EJ, Kremer H, Wolfrum U. A novel Usher protein network at the periciliary reloading point between molecular transport machineries in vertebrate photoreceptor cells. Hum Mol Genet. 2008;17:71–86.
    1. Reiners J, van Wijk E, Maerker T, Zimmermann U, Jurgens K, te Brinke H, Overlack N, Roepman R, Knipper M, Kremer H, Wolfrum U. Scaffold protein harmonin (USH1C) provides molecular links between Usher syndrome type 1 and type 2. Hum Mol Genet. 2005;14:3933–3943. doi: 10.1093/hmg/ddi417.
    1. van Wijk E, van der Zwaag B, Peters TA, Zimmermann U, te Brinke H, Kersten FFJ, Maerker T, Aller E, Hoefsloot LH, Cremers CWRJ, Cremers FPM, Wolfrum U, Knipper M, Roepman R, Kremer H. The DFNB31 gene product whirlin connects to the Usher protein network in the cochlea and retina by direct association with USH2A and VLGR1. Hum Mol Genet. 2006;15:751–765. doi: 10.1093/hmg/ddi490.
    1. Van Wijk E, Kersten FFJ, Kartono A, Mans DA, Brandwijk K, Letteboer SJF, Peters TA, Maerker T, Yan X, Cremers CWRJ, Cremers FPM, Wolfrum U, Roepman R, Kremer H. Usher syndrome and Leber congenital amaurosis are molecularly linked via a novel isoform of the centrosomal ninein-like protein. Hum Mol Genet. 2009;18:51–64.
    1. Sedmak T, Wolfrum U. Intraflagellar transport molecules in ciliary and nonciliary cells of the retina. J Cell Biol. 2010;189:171–186. doi: 10.1083/jcb.200911095.
    1. Mack GJ, Compton DA. Analysis of mitotic microtubule-associated proteins using mass spectrometry identifies astrin, a spindle-associated protein. Proc Natl Acad Sci USA. 2001;98:14434–14439. doi: 10.1073/pnas.261371298.
    1. Thein KH, Kleylein-Sohn J, Nigg EA, Gruneberg U. Astrin is required for the maintenance of sister chromatid cohesion and centrosome integrity. J Cell Biol. 2007;178:345–354. doi: 10.1083/jcb.200701163.
    1. Cheng TS, Hsiao YL, Lin CC, Hsu CM, Chang MS, Lee CI, Yu RC, Huang CY, Howng SL, Hong YR. hNinein is required for targeting spindle-associated protein Astrin to the centrosome during the S and G2 phases. Exp Cell Res. 2007;313:1710–1721. doi: 10.1016/j.yexcr.2007.02.023.
    1. Wolfrum U, Salisbury JL. Expression of centrin isoforms in the mammalian retina. Exp Cell Res. 1998;242:10–17. doi: 10.1006/excr.1998.4038.
    1. Chang MS, Huang CJ, Chen ML, Chen ST, Fan CC, Chu JM, Lin WC, Yang YC. Cloning and characterization of hMAP126, a new member of mitotic spindle-associated proteins. Biochem Biophys Res Commun. 2001;287:116–121. doi: 10.1006/bbrc.2001.5554.
    1. Gruber J, Harborth J, Schnabel J, Weber K, Hatzfeld M. The mitotic-spindle-associated protein astrin is essential for progression through mitosis. J Cell Sci. 2002;115:4053–4059. doi: 10.1242/jcs.00088.
    1. Du J, Jablonski S, Yen TJ, Hannon GJ. Astrin regulates Aurora-A localization. Biochem Biophys Res Commun. 2008;370:213–219. doi: 10.1016/j.bbrc.2008.03.072.
    1. Casenghi M, Meraldi P, Weinhart U, Duncan PI, Korner R, Nigg EA. Polo-like kinase 1 regulates Nlp, a centrosome protein involved in microtubule nucleation. Dev Cell. 2003;5:113–125. doi: 10.1016/S1534-5807(03)00193-X.
    1. Casenghi M, Barr FA, Nigg EA. Phosphorylation of Nlp by Plk1 negatively regulates its dynein-dynactin-dependent targeting to the centrosome. J Cell Sci. 2005;118:5101–5108. doi: 10.1242/jcs.02622.
    1. Rapley J, Baxter JE, Blot J, Wattam SL, Casenghi M, Meraldi P, Nigg EA, Fry AM. Coordinate regulation of the mother centriole component nlp by nek2 and plk1 protein kinases. Mol Cell Biol. 2005;25:1309–1324. doi: 10.1128/MCB.25.4.1309-1324.2005.
    1. Yan J, Jin S, Li J, Zhan Q. Aurora B Interaction of Centrosomal Nlp Regulates Cytokinesis. J Biol Chem. 2010;285:40230–40239. doi: 10.1074/jbc.M110.140541.
    1. Yang YC, Hsu YT, Wu CC, Chen HT, Chang MS. Silencing of astrin induces the p53-dependent apoptosis by suppression of HPV18 E6 expression and sensitizes cells to paclitaxel treatment in HeLa cells. Biochem Biophys Res Commun. 2006;343:428–434. doi: 10.1016/j.bbrc.2006.02.166.
    1. Shao S, Liu R, Wang Y, Song Y, Zuo L, Xue L, Lu N, Hou N, Wang M, Yang X, Zhan Q. Centrosomal Nlp is an oncogenic protein that is gene-amplified in human tumors and causes spontaneous tumorigenesis in transgenic mice. J Clin Invest. 2010;120:498–507. doi: 10.1172/JCI39447.
    1. Wang Y, Zhan Q. Cell cycle-dependent expression of centrosomal ninein-like protein in human cells is regulated by the anaphase-promoting complex. J Biol Chem. 2007;282:17712–17719. doi: 10.1074/jbc.M701350200.
    1. Christensen ST, Pedersen LB, Schneider L, Satir P. Sensory cilia and integration of signal transduction in human health and disease. Traffic. 2007;8:97–109. doi: 10.1111/j.1600-0854.2006.00516.x.
    1. Singla V, Reiter JF. The primary cilium as the cell's antenna: signaling at a sensory organelle. Science. 2006;313:629–633. doi: 10.1126/science.1124534.
    1. Quarmby LM, Parker JD. Cilia and the cell cycle? J Cell Biol. 2005;169:707–710. doi: 10.1083/jcb.200503053.
    1. Smith KR, Kieserman EK, Wang PI, Basten SG, Giles RH, Marcotte EM, Wallingford JB. A role for central spindle proteins in cilia structure and function. Cytoskeleton (Hoboken) 2011;68:112–124.
    1. Liu Q, Tan G, Levenkova N, Li T, Pugh EN Jr, Rux JJ, Speicher DW, Pierce EA. The proteome of the mouse photoreceptor sensory cilium complex. Mol Cell Proteomics. 2007;6:1299–1317. doi: 10.1074/mcp.M700054-MCP200.
    1. Roepman R, Wolfrum U. Protein networks and complexes in photoreceptor cilia. Subcell Biochem. 2007;43:209–235. doi: 10.1007/978-1-4020-5943-8_10.
    1. Wolfrum U, Schmitt A. Rhodopsin transport in the membrane of the connecting cilium of mammalian photoreceptor cells. Cell Motil Cytoskeleton. 2000;46:95–107. doi: 10.1002/1097-0169(200006)46:2<95::AID-CM2>;2-Q.
    1. Liu X, Bulgakov OV, Darrow KN, Pawlyk B, Adamian M, Liberman MC, Li T. Usherin is required for maintenance of retinal photoreceptors and normal development of cochlear hair cells. Proc Natl Acad Sci USA. 2007;104:4413–4418. doi: 10.1073/pnas.0610950104.
    1. Papermaster DS. The birth and death of photoreceptors: the Friedenwald Lecture. Invest Ophthalmol Vis Sci. 2002;43:1300–1309.
    1. Famulski JK, Vos LJ, Rattner JB, Chan GK. Dynein/dynactin-mediated transport of kinetochore components off kinetochores and onto spindle poles induced by nordihydroguaiaretic acid. PLoS One. 2011;6:e16494. doi: 10.1371/journal.pone.0016494.
    1. Zallocchi M, Sisson JH, Cosgrove D. Biochemical characterization of native Usher protein complexes from a vesicular subfraction of tracheal epithelial cells. Biochemistry. 2010;49:1236–1247. doi: 10.1021/bi9020617.
    1. Xue J, Tarnasky HA, Rancourt DE, van der Hoorn FA. Targeted disruption of the testicular SPAG5/deepest protein does not affect spermatogenesis or fertility. Mol Cell Biol. 2002;22:1993–1997. doi: 10.1128/MCB.22.7.1993-1997.2002.
    1. Suzuki H, Yagi M, Suzuki K. Duplicated insertion mutation in the microtubule-associated protein Spag5 (astrin/MAP126) and defective proliferation of immature Sertoli cells in rat hypogonadic (hgn/hgn) testes. Reproduction. 2006;132:79–93. doi: 10.1530/rep.1.01104.
    1. Suzuki H, Yagi M, Saito K, Suzuki K. Embryonic pathogenesis of hypogonadism and renal hypoplasia in hgn/hgn rats characterized by male sterility, reduced female fertility and progressive renal insufficiency. Congenit Anom (Kyoto) 2007;47:34–44. doi: 10.1111/j.1741-4520.2006.00138.x.
    1. Suzuki H, Tokuriki T, Saito K, Hishida A, Suzuki K. Glomerular hyperfiltration and hypertrophy in the rat hypoplastic kidney as a model of oligomeganephronic disease. Nephrol Dial Transplant. 2005;20:1362–1369. doi: 10.1093/ndt/gfh782.
    1. Suzuki H, Tokuriki T, Kamita H, Oota C, Takasu M, Saito K, Suzuki K. Age-related pathophysiological changes in rat oligomeganephronic hypoplastic kidney. Pediatr Nephrol. 2006;21:637–642. doi: 10.1007/s00467-006-0089-3.
    1. Janin-Mercier A, Palcoux JB, Gubler MC, de Latour M, Dalens H, Fonck Y. Oligomeganephronic renal hypoplasia with tapetoretinal degeneration. Report of one case with ultrastructural study of the renal biopsy. Virchows Arch A Pathol Anat Histopathol. 1985;407:477–483. doi: 10.1007/BF00709994.
    1. Reiners J, Nagel-Wolfrum K, Jurgens K, Maerker T, Wolfrum U. Molecular basis of human Usher syndrome: deciphering the meshes of the Usher protein network provides insights into the pathomechanisms of the Usher disease. Exp Eye Res. 2006;83:97–119. doi: 10.1016/j.exer.2005.11.010.
    1. Roepman R, Schick D, Ferreira PA. Isolation of retinal proteins that interact with retinitis pigmentosa GTPase regulator by interaction trap screen in yeast. Methods Enzymol. 2000;316:688–704.

Source: PubMed

3
Abonnieren