ARTICLE
Received 1 Oct 2014 | Accepted 31 Mar 2015 | Published 5 June 2015
Miriam Schmidts1,2,3,4,*, Yuqing Hou5,*, Claudio R. Corts6, Dorus A. Mans2,3, Celine Huber7, Karsten Boldt8,
Mitali Patel1, Jeroen van Reeuwijk2,3, Jean-Marc Plaza9, Sylvia E.C. van Beersum2,3, Zhi Min Yap1,Stef J.F. Letteboer2,3, S Paige Taylor10, Warren Herridge11, Colin A. Johnson11, Peter J. Scambler12,Marius Uefng8, Hulya Kayserili13,14, Deborah Krakow10, Stephen M. King15, UK10Kw, Philip L. Beales1,16, Lihadh Al-Gazali17, Carol Wicking6, Valerie Cormier-Daire7, Ronald Roepman2,3, Hannah M. Mitchison1,**, George B. Witman5,**
The analysis of individuals with ciliary chondrodysplasias can shed light on sensitive mechanisms controlling ciliogenesis and cell signalling that are essential to embryonic development and survival. Here we identify TCTEX1D2 mutations causing Jeune asphyxiating thoracic dystrophy with partially penetrant inheritance. Loss of TCTEX1D2 impairs retrograde intraagellar transport (IFT) in humans and the protist Chlamydomonas, accompanied by destabilization of the retrograde IFT dynein motor. We thus dene TCTEX1D2 as an integral component of the evolutionarily conserved retrograde IFT machinery. In complex with several IFT dynein light chains, it is required for correct vertebrate skeletal formation but may be functionally redundant under certain conditions.
1 Genetics and Genomic Medicine Programme, University College London (UCL), Institute of Child Health, 30 Guilford Street, London WC1N 1EH, UK.
2 Department of Human Genetics, Radboud University Medical Center, 6525 GA Nijmegen, The Netherlands. 3 Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, 6525 GA Nijmegen, The Netherlands. 4 Center for Pediatrics and Adolescent Medicine, University Hospital Freiburg, Mathildenstrasse 1, 79112 Freiburg, Germany. 5 Department of Cell and Developmental Biology, University of Massachusetts Medical School, Worcester, Massachusetts 01655, USA. 6 Institute for Molecular Bioscience, The University of Queensland, St Lucia, Queensland 4072, Australia. 7 INSERM UMR_1163, Dpartement de gntique, Institut Imagine, Universit Paris Descartes Sorbonne Paris Cit, Hpital Necker-Enfants Malades, Assistance Publique-Hpitaux de Paris, Paris 75015, France. 8 Division of Experimental Ophthalmology and Medical Proteome Center, Center of Ophthalmology, University of Tbingen, Tbingen 72074, Germany. 9 Plateforme de Bioinformatique, Institut Imagine, Universit Paris Descartes, Paris 75015, France. 10 Departments of Orthopaedic Surgery and Human Genetics, David Geffen School of Medicine at UCLA, Los Angeles 90095, California, USA. 11 Section of Ophthalmology and Neuroscience, Leeds Institutes of Molecular Medicine, University of Leeds, Leeds LS9 7TF, UK. 12 Developmental Biology and Cancer Programme, University College London (UCL), Institute of Child Health, 30 Guilford Street, London WC1N 1EH, UK. 13 Medical Genetics Department, Istanbul Medical Faculty, Istanbul University, 34093 Istanbul, Turkey. 14 Medical Genetics Department, Koc University School of Medicine, 34010 Istanbul, Turkey. 15 Department of Molecular Biology and Biophysics and Institute for Systems Genomics, University of Connecticut Health Center, Farmington, Connecticut 06030, USA.
16 Centre for Translational Genomics-GOSgene, UCL Institute of Child Health, London WC1N 1EH, UK. 17 Department of Pediatrics, College of Medicine and Health Sciences, United Arab Emirates University, PO Box 17666, Al Ain, United Arab Emirates. * These authors contributed equally to this work. ** These authors jointly supervised the work. w A full list of consortium members appears at the end of the paper. Correspondence and requests for materials should be addressed to H.M.M. (email: mailto:[email protected]
Web End [email protected] )
NATURE COMMUNICATIONS | 6:7074 | DOI: 10.1038/ncomms8074 | http://www.nature.com/naturecommunications
Web End =www.nature.com/naturecommunications 1
& 2015 Macmillan Publishers Limited. All rights reserved.
DOI: 10.1038/ncomms8074 OPEN
TCTEX1D2 mutations underlie Jeune asphyxiating thoracic dystrophy with impaired retrograde intraagellar transport
ARTICLE NATURE COMMUNICATIONS | DOI: 10.1038/ncomms8074
The malfunction of primary cilia, conserved signalling organelles present on the surface of most cells, has emerged as the cause of a growing number of severe
congenital developmental defects1,2. Intraagellar transport (IFT), a highly conserved process required for ciliary growth and signalling, is powered by motors attached to IFT complexes A and B. The kinesin-2 motor transports cargo along ciliary microtubules towards the ciliary tip (anterograde IFT). IFT complexes and other cargo are returned to the base of the cilium (retrograde IFT) by a specialized cytoplasmic dynein3, termed dynein 2 in vertebrates and dynein 1b in Chlamydomonas; (hereafter referred to as IFT dynein). Chlamydomonas has been a key organism for elucidating the molecular and mechanistic basis of IFT. The known subunits of Chlamydomonas IFT dynein are the homodimer-forming heavy chain DHC1b, intermediate chains D1bIC1 and D1bIC2 (also known as FAP163 and FAP133, respectively4), light-intermediate chain D1bLIC and the light-chain LC8. Human IFT dynein contains the homologues of these proteins, DYNC2H1, WDR60, WDR34, DYNC2LI1 and DYNLL1/DYNLL2 (both of which are LC8 homologues), and probably additional subunits410. However, the regulation, cargo interactions and exact composition of IFT dynein have remained relatively elusive.
Primary cilia and IFT are required for vertebrate hedgehog signalling, an important regulator of skeletogenesis11. Mutations in several IFT dynein components cause short-rib polydactyly syndromes (SRPS) and Jeune asphyxiating thoracic dystrophy (JATD; Jeune syndrome), a group of autosomal recessively inherited, genetically heterogeneous ciliary chondrodysplasias with overlapping phenotypic features1216. The spectrum of disease phenotypes varies in affected individuals but hallmarks include short ribs and narrow thorax, short limbs, with sporadic polydactyly and extraskeletal disease including kidney, liver, eye, heart and brain defects. The underlying genetic basis of these skeletal ciliopathies also overlaps, with JATD at the milder end of the JATD/SRPS disease spectrum17. To date, mutations in 10 genes have been shown to cause Jeune syndrome, eight leading to classic JATD with ciliogenesis defects due to IFT dysfunction; these encode heavy and intermediate IFT dynein subunits DYNC2H1, WDR34 and WDR60 (refs 1216), IFT complex B components IFT80 and IFT172 (refs 18,20), and IFT complex A components WDR19/IFT144, TTC21B/IFT139 and IFT140 (refs 2123). Two encode the centriole-associated proteins CEP120 and CSPP1 (Jeune variants), which are important for ciliary assembly or function19,24. Disease-causing mutations are considered hypomorphic since no individuals with SRPS or JATD were previously shown to carry biallelic loss-of-function mutations13,17, and homozygosity for null alleles is embryonic lethal in mouse models around midgestation25,26.
Here we report biallelic loss-of-function mutations causing JATD in the gene encoding TCTEX1D2, an IFT dynein light chain distinct from DYNLL1/DYNLL2 (LC8). Unusually for the SRPS/JATD spectrum, affected individuals all carry biallelic null alleles where complete loss of protein function is predicted. Furthermore, the disease phenotype appears incompletely penetrant. Afnity proteomics indicates that TCTEX1D2/Tctex2b is an integral component of IFT dynein. We demonstrate a retrograde IFT defect in TCTEX1D2-decient human broblasts and Tctex2b-decient Chlamydomonas cells, and nd that IFT dynein is partially destabilized by loss of Tctex2b in Chlamydomonas. Compared with mutations in other IFT dynein components, TCTEX1D2/Tctex2b loss in human, zebrash and Chlamydomonas has a modest effect on retrograde IFT, likely explaining the partially penetrant nature of human TCTEX1D2 mutations.
ResultsVariants in TCTEX1D2 are associated with JATD. Whole-exome sequencing of 69 individuals from 60 families clinically diagnosed with JATD identied a homozygous consensus splice variant (c.113 2C4G) in TCTEX1D2 (NM_152773.4, encoding a
dynein light chain) in individual UCL82 II.1 from a consanguineous Turkish family (Supplementary Fig. 1a). Reverse transcription PCR (RT-PCR) on RNA derived from broblasts of UCL82 II.1 detected no TCTEX1D2 transcript, suggesting nonsense-mediated decay (NMD) of the mutant transcript (Supplementary Fig. 2a). All primers used for human genetic analysis are listed in Supplementary Table 1. Exome copy number variant analysis revealed a 410-kb homozygous deletion in two affected siblings (UCL4 II.6 and II.8) from a consanguineous Arabic family that removes exon 12 of TCTEX1D2 including the start codon, indicating a complete loss-of-function allele(c.(?_ 142)_247 ?del) (Supplementary Fig. 3). No other likely
disease-causing variants in known JATD/SRPS-causing genes or other known ciliary components were detected in these two siblings. The deletion also removes exon 25 of neighbouring TM4SF19 encoding a protein reportedly involved in pancreatic development but unlikely to be involved in JATD (Supplementary Fig. 3). Reverse transcription PCR on RNA derived from blood lymphocytes of individuals UCL4 II.6 and II.8 detected no TCTEX1D2 transcript, indicating likely NMD of the mutant transcript (Supplementary Fig. 2b), and no transcripts initiated at the TM4SF19 start codon continuing into TCTEX1D2 downstream of the deletion either. Analysis of a further 154 JATD/SRPS exomes and Sanger sequencing of TCTEX1D2 in 69 additional JATD/SRPS cases, previously excluded for mutations in known JATD and SRPS genes, detected compound-heterozygous variants in TCTEX1D2 in individual INS II.1 from a non-consanguineous French family comprising a nonsense (c.262C4T; p.Arg88*) and a deletion-insertion frameshift alteration (c.100delinsCT; p.Val34Leufs*12; Supplementary Fig. 1b).
The TCTEX1D2 c.113 2C4G, c.262C4T and c.100delinsCT
variants are absent from the dbSNP, 1,000 Genomes and EVC databases. The exon 12 deletion is absent from 500 exomes from the UK10K project and 100 Bedouin control chromosomes assessed by Sanger sequencing. The c.113 2C4G, c.262C4T
and c.100delinsCT variants segregated with the disease phenotype in affected families as expected (Supplementary Fig. 1). However, in family UCL4, the exon 12 deletion was detected not only in a third affected individual who died at 2 months due to respiratory insufciency (UCL4 II.9) but also in two siblings (UCL4 II.1 andII.5) for whom no clinical signs of JATD had been documented (Fig. 1a and Supplementary Fig. 1c,d). When reassessed clinically with a full X-ray exam, both UCL4 II.1 and II.5 showed mild brachydactyly and slightly shortened lower limb distal segments; they are both shorter in stature than their siblings that do not carry the deletion, and one also had pectus carinatum as a child. However, in contrast to the other ve individuals documented here as affected with typical clinical features of JATD (short horizontal ribs, narrow thorax, trident acetabulum with spurs and polydactyly; Fig. 2 and Table 1), no specic radiological signs of JATD/SRPS were found in UCL4 II.1 and II.5 (Supplementary Fig. 4). Thus, while their mild brachydactyly and short stature could represent a very mild JATD phenotype, or radiological diagnostic criteria for JATD might have been fullled in childhood, we have no clinical or radiological proof for JATD. Brachydactyly and short stature can occur with JATD but are not specic for the condition. We therefore suggest that the phenotype is not fully penetrant in this family, with the caveat that we cannot denitively exclude an extremely mild expression of the JATD phenotype in the two seemingly unaffected individuals harbouring TCTEX1D2 mutations.
2 NATURE COMMUNICATIONS | 6:7074 | DOI: 10.1038/ncomms8074 | http://www.nature.com/naturecommunications
Web End =www.nature.com/naturecommunications
& 2015 Macmillan Publishers Limited. All rights reserved.
NATURE COMMUNICATIONS | DOI: 10.1038/ncomms8074 ARTICLE
I.1 I.2
UCL4
II.1 II.2 II.3 II.4 II.5 II.6 II.7 II.8II.8
II.9II.9
M I.2 II.1 II.2 II.3 II.4 II.5 II.6 II.7
I.1
400 bp
800 bp 600 bp
TCTEX1D2 ex2
TCTEX1D2 ex4
c.(?_142)_247+?del
Exon 1 Exon 3
TCTEX1 domain
142
c.100delinsCT
c.113+2C>G
c.262C>T
Exon 2
Exon 4 Exon 5
1
p. Val34Leufs*12
c.113+2C>G
p.Arg88*
Exon 12 del
Figure 1 | TCTEX1D2 deletion in UCL4 and location of identied variants in TCTEX1D2 protein structure. (a) The absence of TCTEX1D2 exon 1 and 2 in family UCL4 is visualized by PCR of genomic DNA samples from members of the UCL4 pedigree. TCTEX1D2 exon 4 primers verify the presence of the gene in samples, but TCTEX1D2 exon 2 primers do not amplify in some individuals. Children carrying the homozygous exon 12 TCTEX1D2 deletion are marked in black (diagnosed with JATD) or grey (two siblings who were not diagnosed with JATD). The strikethrough indicates death at 2 months of age; double line indicates consanguineous marriage. See also Supplementary Fig. 1. (b) Human TCTEX1D2 (shown above, white boxes indicate untranslated regions (UTR)) consists of ve exons encoding a 142 amino-acid protein (shown below) with a C-terminal TCTEX1 domain (blue box). The location of the four identied TCTEX1D2 mutations is shown in the gene (above). Their corresponding location in the protein (below) shows the TCTEX1 domain will be at least partially lost for all variants identied in individuals with JATD.
We examined this further by single-nucleotide polymorphism-based genome-wide linkage analysis in all members of UCL4. A high penetrance recessive model coding UCL4 II.1 and II.5 as unknown identied ve homozygous linked regions all with a logarithm of odds (LOD) score of 2.9, with TCTEX1D2 in the second largest interval (Supplementary Table 2). Cross-reference to the exome-sequencing data detected the TCTEX1D2 deletion and just one other homozygous variant shared between UCL4 II.6 and II.8 in these intervals. The latter was in an untranslated pseudogene TBC1D3P2 with a minor allele frequency of 1 in 500 in our in-house control database, suggesting it is not causative for JATD. Considering UCL4 II.1 and II.5 as affected under a reduced penetrance model (penetrance set at 0.6) generated three linked regions including one with a LOD score 3.9 across TCTEX1D2, with no homozygous variants shared between UCL4II.6 and II.8 in the other intervals (Supplementary Table 3). Considering UCL4 II.1 and II.5 as unaffected found no linkage to homozygous variants shared between UCL4 II.6 and II.8 except the TBC1D3P2 pseudogene variant (LOD score 2.9; Supplementary Table 4). Finally, considering UCL4 II.1 and II.5 as affected under the high penetrance model identied just one linked region, a 9.4-Mb locus containing TCTEX1D2 with a LOD score of 4.1 (Supplementary Table 5). Thus, along with the expected segregation in two independent families, linkage modelling supports TCTEX1D2 mutations as disease-causing in family UCL4.
Functional predictions and evidence of NMD suggest that all four TCTEX1D2 variants identied are loss-of-function mutations affecting the conserved TCTEX1 domain of the protein (Fig. 1b). The questionable disease status in individuals UCL4 II.1 and II.5 suggests incomplete penetrance in this family, an inheritance pattern not previously described for JATD.
Knockdown of tctex1d2 causes a typical ciliopathy phenotype. TCTEX1D2 function was not previously investigated in vertebrates and we tested for potential redundancy of the orthologous gene in zebrash using oligonucleotide antisense morpholinos to abrogate transcription. Morpholino oligomers have been used widely to provide models for Jeune syndrome by transiently knocking down the expression of the orthologous zebrash genes1820. This approach has been criticized for its inability to reliably discriminate between specic and non-specic effects such as developmental delay and cardiac oedema27,28. To increase reliability, we established that human (NP_689986.2) and zebrash (XP_685487.3) TCTEX1D2 proteins are highly homologous, being 60% identical at 80% BLAST coverage. We separately tested two different splice-blocking morpholinos and focussed on phenotypic features described in JATD zebrash mutants as well as morphants2933.
Both morpholinos yielded identical results, producing a dose-dependent typical ciliopathy phenotype18,20, with ventrally curved body axis, hydrocephalus, abnormal otoliths and small eyes (Fig. 3ah). Loss of transcript was conrmed by RTPCR (Supplementary Fig. 5 and Supplementary Table 1). The few embryos surviving to 4 days post fertilization displayed severe generalized oedema and pronephric cysts (Fig. 3c,g). Embryos also showed defects of the craniofacial cartilage as visualized by alcian blue staining (Fig. 3i,j,m,n), which were comparable to those observed in other ciliary chondrodysplasia zebrash models such as ift80, ift172 and cspp1 (refs 1820). Immunouorescence analysis revealed shorter cilia in the pronephric duct at 24 h post fertilization (h.p.f.) in tctex1d2 morphant embryos compared with controls (Fig. 3k,o,l,p); however, this difference was no longer observed at 48 h.p.f. (not shown). This could reect delayed ciliogenesis in the morphants or could result from general mild developmental delay. Cilia in the neural tube appeared normal (Supplementary Fig. 6).
Loss of TCTEX1D2 causes a retrograde IFT defect. We proceeded to investigate skin broblasts from individual UCL82 II.1 for defects in cilia architecture and IFT disturbance. While the percentage of ciliated cells was modest but signicantly lower for TCTEX1D2 mutant broblasts compared with controls (Fig. 4e), no signicant difference in ciliary length was observed after 24-h serum starvation, in contrast to the cilia shortening reported in individuals with mutations in other JATD/SRPS genes12,19 (Fig. 4f). However, there was marked accumulation of the IFT-particle protein IFT88 in B35% of the ciliary tips of
TCTEX1D2-decient broblasts, compared with o10% of control broblasts (Fig. 4ad). Cells from an individual previously reported to carry DYNC2H1 mutations34 showed a very similar pattern, which indicates a retrograde IFT defect (Fig. 4d).
Chlamydomonas Tctex2b is a light chain of IFT dynein. We further investigated the role in IFT of Tctex2b, the Chlamydomonas homologue of TCTEX1D2 (49% identical at 80% BLAST coverage; human to Chlamydomonas reciprocal best match BLAST 9e 36). To determine whether Tctex2b is a subunit of
IFT dynein in Chlamydomonas, we used a new strain expressing D1bIC2 (homologue of human IFT dynein intermediate chain WDR34) fused to the haemagglutin-epitope tag (HA). This was
NATURE COMMUNICATIONS | 6:7074 | DOI: 10.1038/ncomms8074 | http://www.nature.com/naturecommunications
Web End =www.nature.com/naturecommunications 3
& 2015 Macmillan Publishers Limited. All rights reserved.
ARTICLE NATURE COMMUNICATIONS | DOI: 10.1038/ncomms8074
UCL4 II.6INS II.1UCL82 II.1
* * * * * *
Figure 2 | Clinical features of subjects with TCTEX1D2 mutations. Affected individuals presented with narrow thorax due to shortened ribs(ac, UCL82 II.1; g, INS family II.1; j, UCL4 II.6), typical pelvis conguration showing trident acetabulum with spurs (arrows) (d, UCL82 II.1; h, INS family II.1; k, UCL4 II.6), polydactyly (e,f, UCL82 II.1; l, UCL4 II.6), shortened extremities (a,b, UCL82 II.1) and brachydactyly (i, INS family II.1, asterisks indicate toes). UCL82 is shown at 38 days in a, c, d and f) and 5.5 yrs in b and e).
generated by transforming an insertional mutant defective for D1bIC2 (dic5-1; Hou et al., manuscript in preparation) with C-terminal HA-tagged D1bIC2. dic5-1 cells have a severe agellar assembly defect that is fully rescued by HA-tagged D1bIC2 (Supplementary Fig. 7), indicating its functional incorporation into IFT dynein. D1bIC2-HA has a normal distribution in the cell, is expressed in agella at approximately normal levels and sediments normally as part of a complex when the agellar membrane-plus-matrix is fractionated on sucrose gradients (Supplementary Fig. 7). Therefore, D1bIC2-HA behaves like wild-type D1bIC2 at all levels examined.
We used anti-HA antibody-conjugated beads to immuno-precipitate D1bIC2-HA protein from the membrane-plus-matrix fraction of isolated steady-state (that is, non-assembling) agella of the D1bIC2-HA strain. SDSpolyacrylamide gel electrophoresis (SDSPAGE) detected bands at BMr 90,000 and 15,000 in the D1bIC2-HA immunoprecipitate that were absent from the wild-type control (Fig. 5a). Mass spectrometry of excised bands identied several proteins specic for the D1bIC2-HA sample, including D1bIC1 (homologue of H.s. WDR60), Tctex2b, Tctex1 (homologous to both H.s. DYNLT1 and DYNLT3) and LC8 (homologous to both H.s. DYNLL1 and DYNLL2). LC7a (homologue of H.s. DYNLRB1 and DYNLRB2) was identied by nine peptides in the D1bIC2-HA sample and two peptides in the wild-type control. Western blotting conrmed that D1bIC1, Tctex2b, Tctex1 and LC8 were specic for the D1bIC2-HA sample (Fig. 5be), and also identied LC7b (also a homologue of
H.s. DYNLRB1 and DYNLRB2) whose peptides were not detected in the immunoprecipitate because it migrated below the excised slice (Fig. 5c). Moreover, all detectable D1bIC1, Tctex2b and Tctex1 were co-immunoprecipitated from the fraction (Fig. 5bd). About one-half of the LC7b, about one-quarter of the DHC1b, D1bLIC and LC8, and trace amounts of FLA10 (a kinesin-2 heavy chain) and several IFT-particle proteins also were co-immunoprecipitated with D1bIC2 (Fig. 5be).
Tctex2b and Tctex1 have not previously been shown to be associated with IFT dynein in Chlamydomonas, but they are known subunits of the axonemal inner arm dynein I1/f, which is involved in agellar motility35. Dynein I1/f is present at very low levels in the membrane-plus-matrix fraction of steady-state agella36. Nevertheless, to be sure that the presence of Tctex2b and Tctex1 in the D1bIC2-HA immunoprecipitate was not due to contamination by dynein I1/f, we probed blots of the unbound and bound membrane-plus-matrix with an antibody to IC140, an I1/f intermediate chain37, and found no IC140 in either the unbound or bound fractions of either the wild-type or mutant extracts. We also probed for p28, a subunit common to several other inner arm dyneins and previously found in the membrane-plus-matrix fraction of steady-state agella36. We readily detected p28 in the unbound fractions, but not in the bound fractions, indicating that the bound fractions were not contaminated by these other inner arm dyneins (Fig. 5e). We conclude that Tctex2b and Tctex1 are novel components of Chlamydomonas IFT dynein. These results also show that the light chains Tctex2b
4 NATURE COMMUNICATIONS | 6:7074 | DOI: 10.1038/ncomms8074 | http://www.nature.com/naturecommunications
Web End =www.nature.com/naturecommunications
& 2015 Macmillan Publishers Limited. All rights reserved.
NATURE COMMUNICATIONS | DOI: 10.1038/ncomms8074 ARTICLE
Table 1 | Summary of clinical ndings in individuals with TCTEX1D2 mutations.
Family Ethnicity Consanguineous
Patient Mutation Clinical diagnosis
Thorax Polydactyly Other skeletal features
Renal, liver or eye phenotype
Other remarks
Yes II.6 Homozygous deletion of exon 1 2
JATD Short horizontal ribs, narrow thorax
Foot Trident acetabulum with spurs
No Obesity with hypertension, alive at 16 years, height 10.p.
UCL4 UAE
(Yemeni)
II.8 Homozygous deletion of exon 1 2
JATD Short horizontal ribs, narrow thorax
No Prolonged tracheostomy, PEG tube, alive at 10 years, height o3.p, on growth hormone up to 5.p.
Hand and foot
Trident acetabulum with spurs
II.9 Homozygous deletion of exon 1 2
JATD Short horizontal ribs, narrow thorax
Hand and foot
Trident acetabulum with spurs
No Died aged 2 months due to respiratory failure
II.1 Homozygous deletion of exon 1 2
NA Normal as a young adult
No Not evident as a young adult (only mild brachydactyly and limb shortening)
No Height o3.p.
No X-ray documentation during childhood performed
II.5 Homozygous deletion of exon 1 2
NA Normal as a young adult.Pectus carinatum reported as a child
No Not evident as a young adult (only mild brachydactyly and limb shortening)
No Height o3.p.
No X-ray documentation during childhood performed
Hand and foot
Trident acetabulum with spurs, brachydactyly
No Height o3.p.
Alive aged 5.5 years
UCL82 Turkish Yes II.1 Homozygous splice site mutationc.113 2C4G
JATD Short horizontal ribs, narrow thorax
INS French No II.1 Comp. heterozygousc.262C4T,p.Arg88*;c.100delinsCT,p.Val34Leufs*12
JATD Short horizontal ribs
No Trident acetabulum with spurs, brachydactyly
No Height 25.-50. p.
Alive at 3 years
JATD, Jeune asphyxiating thoracic dystrophy; NA, not applicable; p, percentile.
and Tctex1, and intermediate chain D1bIC1 are more closely associated with D1bIC2 than with the heavy and light-intermediate chains DHC1b and D1bLIC. It is therefore likely that D1bIC2, D1bIC1, Tctex1, Tctex2b and LC7b together with LC8 form a discrete intermediate chain/light-chain complex within IFT dynein.
Tctex2b is involved in retrograde IFT. With this evidence that Tctex2b is a subunit of IFT dynein, we investigated whether loss of Tctex2b affects retrograde IFT. We re-examined a Chlamydomonas Tctex2b null strain (here termed tctex2b)35 previously shown to swim slower than wild-type cells owing to a defect in inner arm dynein I1/f, but not examined for defects in retrograde IFT. The tctex2b mutant has normal length agella at steady state, but its rate of agellar assembly is about one-third less than that of wild type (Fig. 6a). Differential interference contrast (DIC) microscopy revealed that anterograde IFT velocity is normal, and anterograde IFT frequency only slightly reduced (Fig. 6b,c). However, retrograde IFT velocity is B50% reduced, and retrograde IFT frequency is o25% that of wild type (Fig. 6b,c).
Therefore, loss of Tctex2b has a severe effect specically on retrograde IFT, indicating that Tctex2b is required for normal retrograde IFT.
IFT dynein is destabilized in the absence of Tctex2b. To better understand why retrograde IFT frequency and velocity are reduced in the Chlamydomonas tctex2b null mutant, we compared IFT dynein levels in wild-type and tctex2b whole-cell lysates by western blotting (Fig. 6d). DHC1b, D1bIC2 and D1bLIC were each reduced by B4060% in the mutant cells relative to wild type. These results strongly suggest that IFT dynein is destabilized in the absence of Tctex2b, with resulting degradation of the protein complex. In isolated tctex2b agella, levels of DHC1b, D1bIC2 and D1bLIC were reduced even further (Fig. 6e) to only B1525% that of wild type. This likely accounts for the reduced frequency of retrograde IFT in the mutant.
Western blotting also revealed that the mutant whole cells had normal levels of IFT-particle proteins (Fig. 6d), but the mutant agella had increased amounts of all IFT complex A and B
NATURE COMMUNICATIONS | 6:7074 | DOI: 10.1038/ncomms8074 | http://www.nature.com/naturecommunications
Web End =www.nature.com/naturecommunications 5
& 2015 Macmillan Publishers Limited. All rights reserved.
ARTICLE NATURE COMMUNICATIONS | DOI: 10.1038/ncomms8074
Control mo
tctex1d2 mo
100 m 100 m
100 m
100 m 100 m 100 m 100 m 100 m
Pronephros Pronephros
Acetylated tubulin + DAPI Acetylated tubulin + DAPI
Acetylated tubulin + DAPI
Alcian blue Alcian blue
Alcian blue
100 m
100 m
50 m
100 m
100 m
50 m
50 m
Alcian blue
Acetylated tubulin + DAPI
50 m
Figure 3 | Knockdown of tctex1d2 in zebrash leads to a typical ciliopathy phenotype. Whole-mount light microscopy showing control morpholino (mo)-injected embryos (ad) and tctex1d2 morphants at 4 days post fertilization (eh). Compared with controls, knockdown of tctex1d2 results in ventrally curved body axis (a,e), small eyes (b,f), pronephric cysts (c,g) and otolith defects (d,h). Alcian blue staining of cartilage identies craniofacial cartilage defects in tctexd2 morphants (m,n) compared with controls (i,j). Immunouorescence analysis after staining of cilia at 24 h.p.f. with anti-acetylated tubulin antibody reveals shorter cilia in the pronephric duct of tctex1d2 morphants (o, magnied in p) compared with control embryos (k, magnied in l); however, this difference was no longer evident at 48 h.p.f. (data not shown). Scale bars, 100 mm (aj,m,n) or 50 mm (k,l,o,p).
proteins examined (Fig. 6e). This is in agreement with the IFT88 accumulation in broblast cilia from affected individual UCL82II.1 (Fig. 4); both indicate a retrograde IFT defect. If nearly normal amounts of IFT complexes A and B (apparently assembled into larger IFT trains; Fig. 6b) are being moved by fewer dynein motors, it could also explain why IFT velocity is reduced in the mutant agella. In addition, since IFT dynein is reduced much more in the mutant agella than in the cell body, the IFT dynein lacking Tctex2b is likely to be imported less efciently into agella than is the complete IFT dynein.
TCTEX1D2 is a component of human IFT dynein. To conrm that TCTEX1D2 is part of human IFT dynein, we performed afnity proteomics after expressing SF-TAP (Streptavidin/FLAG Tandem Afnity Purication)-tagged TCTEX1D2 in HEK293T cells; cells expressing SF-TAP-RAF1 served as a control. We were able to identify interactions of TCTEX1D2 with IFT dynein intermediate chains WDR34 and WDR60, and light chains DYNLT1, DYNLT3 and DYNLRB1 (Supplementary Table 7). No peptides from cytoplasmic dynein 1 intermediate chains were detected, ruling out possible contamination by that dynein. An interaction between TCTEX1D2 and WDR60 was veried after overexpression of Flag-tagged TCTEX1D2 and GFP-tagged WDR60 in HEK293T cells (Supplementary Fig. 8). Identication of TCTEX1D2, DYNLT1, DYNLT3 and DYNLRB1 as part of the human IFT dynein is consistent with our nding that the homologous Tctex2b, Tctex1 and LC7b are part of IFT dynein in
Chlamydomonas, and provides independent evidence that IFT dynein contains several different light chains. A model of IFT dynein based on these ndings is shown in Fig. 7.
DiscussionHere we report that TCTEX1D2/Tctex2b is a light chain of IFT dynein in humans and Chlamydomonas, and show for the rst time that it is essential for IFT dynein stability and normal retrograde (tip-to-base) IFT. In zebrash, tctex1d2 gene silencing suggests a role conserved across vertebrates. We show that biallelic loss-of-function alleles in this gene in humans cause JATD. In all affected individuals, we identied biallelic TCTEX1D2 null alleles, which is in striking contrast to JATD and SRPS individuals carrying mutations in other IFT dynein genes such as DYNC2H1 (refs 15,16,34,38,39), WDR34 (refs 12,13), WDR60 (ref. 14) or in genes encoding IFT-A21 and IFTB18,20 components. These individuals usually carry at least one missense allele with inheritance restricted to two such hypomorphic alleles or one hypomorphic allele in combination with a null mutation13,17,39. Biallelic null alleles also cause early embryonic lethality in mouse IFT dynein knockouts25,26,4042, and complete loss of IFT dynein subunits was thus to date considered incompatible with embryonic development beyond midgestation. Our data suggest that TCTEX1D2 is an exception to this dogma, since all but one child carrying TCTEX1D2 null mutations survived beyond infancy.
The high survival rate could reect a milder thorax phenotype than in individuals with mutations in other IFT dynein genes
6 NATURE COMMUNICATIONS | 6:7074 | DOI: 10.1038/ncomms8074 | http://www.nature.com/naturecommunications
Web End =www.nature.com/naturecommunications
& 2015 Macmillan Publishers Limited. All rights reserved.
NATURE COMMUNICATIONS | DOI: 10.1038/ncomms8074 ARTICLE
Control UCL82 II.1 DYNC2H1 subject
-Tubulin + IFT88 + pericentrin -Tubulin + IFT88 + pericentrin -Tubulin + IFT88 + pericentrin
IFT88 IFT88 IFT88
0
0.90
0
Fraction of ciliated cells
0.75
0.85
0.80
0.70
0.65
0.60
1
Control UCL82 II.1
0.9
0.8
10 P=0.065
0.7
Cilia length (micrometer)
0.6
8
0.5
6
0.4
0.3
4
0.2
0.1
2
Control UCL82 II.1 DYNC2H1 individual
IFT88 accumulation at the ciliary tip Normal IFT88 distribution
* P=0.026
NS
Control UCL82 II.1
Figure 4 | Loss of human TCTEX1D2 results in retrograde IFT defects. Immunouorescence analysis using confocal microscopy revealed an accumulation of IFT88 at the ciliary tips in skin broblasts from individual UCL82 II.1 compared with a control (a,b). The accumulation is comparable to that previously reported in broblast cilia from an individual with JATD caused by biallelic variants in DYNC2H1 (ref. 34) (c). IFT88 staining is shown in green, anti-acetylated tubulin antibody (red) was used for visualization of the ciliary axoneme, anti-pericentrin antibody (white) marks the ciliary base; the IFT88 labeling also is shown separately in the lower panels. Scale bars, 5 mm. (d) Fraction of cells with IFT88 accumulation at the ciliary tip, 100 cells analysed for each condition. (e) The percentage of ciliated cells in the broblast sample from individual UCL82 II.1 compared with control broblasts as assessed by counting the number of cilia stained with anti-acetylated tubulin antibody in relation to nuclei stained with DAPI (4,6-diamidino-2-phenylindole, bluein ac) in 10 random visual elds in ve independent experiments each, revealing a very mild reduction in ciliation for the TCTEX1D2-decient cells. One hundred cells were counted per experiment, represented by a single point per experiment. (f) No difference in cilia length between UCL82 II.1 and control broblasts was visualized using anti-acetylated tubulin antibody, 100 cells analysed for each condition. Statistical signicance in e and f was measured using the Students t-test, asterisk indicates P value o0.05.
such as DYNC2H1, WDR34 and WDR60 (refs 1215,34). Similarly to individuals with mutations in the IFT dynein heavy-chain gene DYNC2H1 (refs 15,34), individuals with TCTEX1D2 mutations did not show signs of extraskeletal disease; however, these phenotypes could still emerge with age. Two siblings (UCL4 II.1 and II.5) carrying homozygous TCTEX1D2 mutations did not show overt signs of JATD, suggesting disease non-penetrance. Both are now young adults, and it is possible that early JATD radiological features (handlebar clavicles, trident acetabulum with spurs and cone-shaped epiphyses) were missed because a full X-ray exam was only performed in adulthood after we detected the unusual penetrance pattern. However, in contrast to their three affected siblings, they also do not exhibit polydactyly or shortened ribs. Although both are short statured with mildly short digits and lower limbs, more specic radiological signs of JATD/SRPS were not found.
Together, these clinical ndings suggest a less essential role of TCTEX1D2 in ciliary transport mechanisms compared with other proteins mutated in JATD and SRPS. Cilia of human broblasts with TCTEX1D2 mutations and tctex1d2 zebrash morphants lack gross structural defects, indicating a potentially non-essential role in vertebrate ciliogenesis. Similarly, agella of the Chlamydomonas tctex2b null mutant are normal length at steady state, whereas null mutants of IFT dynein subunits DHC1b or D1bLIC have a severe short agellar phenotype4345.
Our studies in Chlamydomonas provide insight into the molecular mechanism underlying the milder phenotype. Loss of Chlamydomonas Tctex2b causes IFT dynein instability and a reduced amount of IFT dynein in agella; however, surprisingly, the residual IFT dynein lacking Tctex2b retains enough functionality to maintain full-length agella. Thus, it appears
that a key role of Tctex2b is to stabilize IFT dynein. The lower level of IFT dynein is accompanied by reduced retrograde IFT velocity and frequency, causing an accumulation of IFT-particle proteins both in Chlamydomonas agella and skin broblast cilia from affected individual UCL82 II.1. That Chlamydomonas cells tolerate some reduction of retrograde IFT is in agreement with observations that the dhc1b temperature-sensitive mutants dhc1b-3 and a24 have reduced amounts of IFT dynein and reduced retrograde IFT at permissive temperature, yet still assemble normal length agella4648. In the case of the tctex2b mutant, agella are formed more slowly than in wild-type cells, probably reecting slower recycling of IFT components.
The slower agellar assembly in the Chlamydomonas tctex2b mutant, if recapitulated in vertebrates, might explain our observation that tctex1d2 morphant zebrash embryos appeared to have short cilia at 24 h.p.f. but normal length cilia at 48 h.p.f. Interestingly, this nding, and the normal length cilia and agella found in patient UCL82 II.1 broblasts and the Chlamydomonas tctex2b mutant, contrasts with a recent report that short interfering RNA-mediated depletion of TCTEX1D2 in human telomerase-immortalized retinal pigment epithelial (hTERTRPE1) cells resulted in longer cilia49. However, cilia length changes, ranging from shorter, normal length or longer than normal12,18,34, have been reported in cells from JATD patients with a number of gene defects. Furthermore, WDR34 mutant patient broblasts have shortened cilia12, whereas WDR34 short interfering RNA knockdown in hTERT-RPE1 results in longer cilia49. Vertebrate cilia length changes arising from retrograde IFT perturbations may thus be cell type specic or subject to experimental variability. These changes are not necessarily predictable, and therefore do not seem to provide a reliable
NATURE COMMUNICATIONS | 6:7074 | DOI: 10.1038/ncomms8074 | http://www.nature.com/naturecommunications
Web End =www.nature.com/naturecommunications 7
& 2015 Macmillan Publishers Limited. All rights reserved.
ARTICLE NATURE COMMUNICATIONS | DOI: 10.1038/ncomms8074
WT D1blC2-HA
Unbound
Bound
Bound
D1blC2-HA
D1blC2-HA
250
WT
150
kDa
WT
*
100
250
150
100
75 D1blC2
D1blC1
D1bLIC
IFT172
IFT139
FLA10 IFT81
DHC1b
75
75
50
Unbound
Bound
D1blC2-HA
Unbound
Bound
Unbound
D1blC2-HA
kDa
WT
WT
D1blC2-HA
D1blC2-HA
D1blC2-HA
D1blC2-HA
75
kDa
WT
WT
kDa
WT
WT
D1blC2
D1blC1
D1bLIC
Tctex1
75
D1blC2
Tctex2b
LC7b
15
75 D1blC2
p28
LC8
100
50
10 10
25
10
15
100
50 37
25 20 15 10
75
Figure 5 | Tctex2b and Tctex1 are in an IFT dynein intermediate chain/light-chain subcomplex with D1bIC2 and D1bIC1. (a) Flagellar membrane-plus-matrix fractions from wild-type (WT) cells or cells expressing D1bIC2-HA were incubated with anti-HA antibody-conjugated beads. The proteins pulled down by the beads were separated by SDSPAGE and silver stained. One band (marked *) between 75 and100 kDa and several bands around 15 kDa (marked ]) are specic for the D1bIC2-HA sample. (bd) Western blots conrming that Tctex2b, Tctex1 and D1bIC1 are specically co-precipitated with D1bIC2-HA. The unbound and bound samples were probed with the indicated antibodies. In each experiment, all of the D1bIC2-HA was immunoprecipitated from the D1bIC2-HA sample; all of the D1bIC1 (b,d), Tctex1 (b) and Tctex2b (c) was co-precipitated from the D1bIC2-HA sample. None of these proteins were pulled down from the WT control. Some but not all of the DHC1b (d), D1bLIC (b,d), LC7b (c) and LC8 (e) was co-precipitated from the D1bIC2-HA samples; (d) also shows that only very small amounts of the IFT-particle proteins or FLA10 were co-precipitated with D1bIC2-HA.(e) Similar western blot showing that p28 was not co-precipitated with D1bIC2-HA. In b, d and e, the ratio of unbound: bound protein loaded was 1:4; in c, the ratio was 1:2. All antibodies used for Chlamydomonas protein analysis are listed in Supplementary Table 6.
measure, compared with the direct IFT measurements reported here that include immunouorescence imaging of altered localization of IFT cargos and DIC imaging of IFT velocity. Although no difference in cilia length was observed in skin broblasts of JATD compared with control individuals in our study, similar to the Chlamydomonas Tctex2b mutant, any delay in cilia assembly could have severe consequences during development if full-length cilia are required for proper functioning of the hedgehog or other vertebrate ciliary signalling pathways soon after onset of cilia formation. Since retrograde IFT is critical for hedgehog signalling11,50, impaired retrograde IFT even in full-length cilia could be expected to affect skeletal development, as seen in the TCTEX1D2-mutated JATD individuals.
The Chlamydomonas Tctex2b mutant swims B30% slower than wild-type cells owing to defects in axonemal dyneins35. However, a ciliary motility defect is not the cause of JATD, because primary ciliary dyskinesia (PCD) patients with ciliary
motility defects do not have JATD symptoms, and multiciliated cells bearing motile cilia are not present in the tissues affected in JATD nor in their progenitor cells1. JATD is caused by defects in primary cilia; primary cilia lack axonemal dyneins.
Proteomic analysis in Chlamydomonas and human cells conrmed Tctex2b/TCTEX1D2 as a component of IFT dynein along with intermediate chains D1bIC2/WDR34 and D1bIC1/ WDR60, and a number of light chains. Immunoprecipitation of Chlamydomonas IFT dynein not only identied Tctex2b but also Tctex1 and LC7b as novel subunits of IFT dynein, indicating that these light chains together with LC8, D1bIC2 and D1bIC1 likely form a light-chain/intermediate chain complex within IFT dynein, which also may contain subunits not detected by our methods. Proteomics analysis of HEK293T proteins co-immuno-precipitated with SF-TAP-tagged TCTEX1D2 likewise identied DYNLT1 and DYNLT3 (human Tctex1), and additional light chains in the LC8 and roadblock families (DYNLL2 LC8-type 2
and DYNLRB1 DNLC2A). The light-chain pairs DYNLL1/
DYNLL2 and DYNLRB1/DYNLRB2 have a high sequence identity of 93% and 77% respectively. This approach did not identify any peptides specic for DYNLL1 or DYNLRB2, but one peptide each was identied for DYNLL1/DYNLL2 and DYNLRB1/DYNLRB2, which could have originated from either of the two homologous proteins. Furthermore DYNLL1 was previously conrmed as a light chain of human IFT dynein13. Therefore, we propose that these subunits are components of an ancient, highly conserved intermediate chain/light-chain complex within IFT dynein (Fig. 7). As such, these light chains are strong candidates for harbouring novel mutations causing JATD.
Dynein complexes fall into two general types based on whether they contain single or multiple heavy-chain motor units51. Those with multiple motors all associate with a core group of accessory proteins including WD-repeat intermediate chains and light chains in the Dynll/LC8, Dynlt/Tctex1 and Dynlrb/LC7 classes (axonemal outer dynein arms and inner arm I1/f also include proteins related to TCTEX1D2/Tctex2, which form a distinct subfamily within the Dynlt/Tctex1 group51). Our demonstration that the IFT dynein includes a member of this subfamily suggests that the presence of TCTEX1D2 is also a conserved dening feature of these multimotor dyneins, and raises the possibility that canonical cytoplasmic dynein 1 might also associate with members of this light chain type under certain circumstances52.
The possible disease non-penetrance in family UCL4 is of clinical interest. Variable phenotypic severity is well documented for JATD/SRPS even within families. However, never before to the extent that individuals can appear clinically unaffected but carry biallelic loss-of-function variants that cause documented JATD/SRPS in other family members. Functional redundancy of TCTEX1D2 with other proteins could explain this, if, for example, other dynein light chains might in certain conditions be able to compensate for its loss. Mice carrying Dynll1 mutations showed possible functional redundancy of IFT dynein light chains, whereby DYNLL2 may be able to function in place of DYNLL1 (ref. 53). We looked at single-nucleotide polymorphism haplotypes across dynein light-chain genes DYNLL1, DYNLL2 and TCTEX1 in individuals UCL4 II.1 and II.5 who carry the TCTEX1D2 deletion but appear clinically unaffected. Both have different haplotypes across TCTEX1 compared with their siblingsII.8 and II.9 who display a classical JATD phenotype, but whether protective alleles could exist in compensatory genes is not clear (Supplementary Fig. 9).
Alternatively, TCTEX1D2 loss could be attenuated by individual-to-individual variation in the proteins with which TCTEX1D2 normally interacts, or in other proteins involved in IFT dynein stabilization or degradation. Modier alleles have been debated as the underlying cause of the phenotypic variability
8 NATURE COMMUNICATIONS | 6:7074 | DOI: 10.1038/ncomms8074 | http://www.nature.com/naturecommunications
Web End =www.nature.com/naturecommunications
& 2015 Macmillan Publishers Limited. All rights reserved.
NATURE COMMUNICATIONS | DOI: 10.1038/ncomms8074 ARTICLE
14
4.5
4
Flagella length (m)
12
10
3.5 3
8
2.5
6
2
1.5
4
1
2
0.5
0
0 0 120 150 Anterograde velocity (m s1)
A54-e18 (n=4)
Whole cell
A54-e18 kDa
250 DHC1b
D1blC2
D1bLIC
IFT172 IFT139
IFT81
Tubulin
75
75
Retrograde velocity (m s1)
Anterograde tracks per s
Retrograde tracks per s
30
60
Time (min)
Before deflagellation
A54-e18 tctex2b tctex2b (n=5)
90
Tip
Base
tctex2b
Time (10 s) Tip
Flagella
A54-e18 kDa
250 DHC1b
D1blC2
D1bLIC
IFT172 IFT139
IFT81
Tubulin
75
75
tctex2b
A54-e18
50
50
50
200
50
tctex2b
150
150
Base
Time (10 s)
Figure 6 | Loss of Tctex2b causes IFT dynein instability and a retrograde IFT defect in Chlamydomonas. (a) The tctex2b mutant has defects in agella regeneration. The tctex2b null mutant and A54-e18 (the wild-type strain from which tctex2b was derived) were deagellated and then allowed to regrow their agella. Flagella lengths were measured before deagellation and at time points after deagellation. The two strains had identical agellar lengths(11.4 mm) before deagellation. For each time point, one agellum from each of 50 cells was measured; error bars are s.d. (b) The tctex2b mutant is defective in retrograde IFT. IFT was recorded in wild-type (A54-e18) and tctex2b agella by DIC microscopy, and kymograms generated from the video recordings. Tracks with positive slopes represent IFT particles moving anterogradely, and tracks with negative slopes represent particles moving retrogradely. Compared with wild type, few retrograde tracks are visible in the tctex2b kymogram, and these had a much reduced slope. Retrograde particles had a larger apparent size in mutants; similar ndings were reported for a temperature-sensitive dhc1b mutant47. (c) Quantitative analysis of IFT in wild type (A54-e18) and tctex2b. In tctex2b, anterograde IFT velocity is about the same as in wild type, while anterograde frequency is only slightly reduced, but both retrograde IFT velocity and frequency are greatly reduced. n, number of agella analysed. Error bars show s.d. (d) Western blot showing reduced IFT dynein subunits in tctex2b whole-cell lysates. Wild-type (A54-e18) and tctex2b whole-cell lysates were probed with antibodies to IFT dynein subunits and IFT-particle proteins. DHC1b, D1bIC2 and D1bLIC are reduced in tctex2b whole-cell lysate. No signicant changes were detected for IFT proteins. The same samples were probed for tubulin as loading control. (e) IFT dynein is greatly reduced in tctex2b agella. Wild-type (A54-e18) and tctex2b agella were probed with antibodies to IFT dynein subunits and IFT-particle proteins. IFT dynein subunits DHC1b, D1bIC2 and D1bLIC are greatly reduced in tctex2b agella. IFT-A protein IFT139 and IFT-B proteins IFT172 and IFT81 are increased in tctex2b agella, consistent with a retrograde IFT defect. The same samples were probed for tubulin as loading control.
Dynein 2 (Hs) / Dynein 1b (Cr)
in ciliary chondrodysplasias13,23,34 and other ciliopathies such as BardetBiedl, Joubert and Meckel syndromes; however, this has remained a challenge to prove in human subjects54,55. Variable penetrance in autosomal recessive conditions is still quite unusual, one example being a common CFTR variant that causes either disease or a normal phenotype depending on its genetic context56.
It may be relevant that partial suppression of mutations in genes encoding IFT-particle proteins has been observed in Chlamydomonas and Tetrahymena5759; this occurs in association with stress, suggesting possible involvement of a chaperone in stabilizing the incomplete IFT particle. Given that a key role of Tctex2b is to stabilize IFT dynein, the presence or absence of another protein such as a chaperone that helps stabilize IFT dynein could determine whether or not a developmental defect is observed in the absence of Tctex2b. Interestingly, peptides derived from chaperones were abundant in our proteomic analysis of TAP-tagged TCTEX1D2 from HEK293T cells.
In summary, we have identied mutations associated with a complete loss of TCTEX1D2 causing Jeune syndrome. TCTEX1D2 mutations are a rare cause of disease affecting o5% of cases in this study (three affected families identied after screening of 4300 individuals with JATD/SRPS). There are
DYNLL1, DYNLL2 (Hs) LC8 (Cr)
DYNLT1, DYNLT3 (Hs) TCTEX1 (Cr)
DYNLRB1, ?DYNLRB2 (Hs) LC7b, ?LC7a (Cr)
TCTEX1D2 (Hs) TCTEX2b (Cr)
WDR34 (Hs) D1blC2 (Cr)
WDR60 (Hs) D1blC1 (Cr)
Figure 7 | Proposed model of IFT dynein composition in Homo sapiens (Hs) and C. reinhardtii (Cr). Left, IFT dynein (dynein 2/1b) is composed of dynein heavy chains (DYNC2H1(Hs)/DHC1b(Cr); shown in dark grey), dynein light-intermediate chains (DYNC2LI1(Hs)/D1bLIC(Cr); shown in light grey) and different dynein intermediate and light chains (coloured, shown in detail on the right). Right, dynein intermediate chains (WDR34(Hs)/D1bIC2(Cr) and WDR60(Hs)/D1bIC1(Cr)) interact with different dynein light-chain subtypes, including TCTEX1D2(Hs)/ TCTEX2b(Cr). Question marks indicate LC7a and DYNLRB2 as unconrmed components suggested from our Chlamydomonas and human results.
NATURE COMMUNICATIONS | 6:7074 | DOI: 10.1038/ncomms8074 | http://www.nature.com/naturecommunications
Web End =www.nature.com/naturecommunications 9
& 2015 Macmillan Publishers Limited. All rights reserved.
ARTICLE NATURE COMMUNICATIONS | DOI: 10.1038/ncomms8074
important implications for genetic counselling since individuals carrying TCTEX1D2 mutations appear to have a higher survival rate than individuals affected by mutations in other IFT dynein components, and lack extraskeletal symptoms. In humans, sh and Chlamydomonas, TCTEX1D2 mutations do not confer changes in gross ciliary structure, but do impair the highly conserved retrograde IFT machinery. Proteomic analysis provides evidence that TCTEX1D2/Tctex2b is a component of an intermediate chain/light-chain subcomplex within IFT dynein. In Chlamydomonas, the retrograde IFT defect is associated with instability of IFT dynein; motor stability may be the major role of Tctex2b since the residual IFT dynein missing Tctex2b retains some functionality. Our ndings that IFT is impaired to a lesser extent in TCTEX1D2/Tctex2b null cells than in cells carrying null mutations in other IFT dynein components could explain, in part, the apparently incomplete penetrance underlying TCTEX1D2 disease. This potentially has future clinical therapeutic implications, for example, if modulation of other components affecting the IFT dynein system could be harnessed to attenuate the effects of a lack of TCTEX1D2 function.
Methods
Patients. Inclusion criteria were the clinical diagnosis of JATD based on clinical and radiological ndings including short ribs with small/narrow thorax and small ilia with acetabular spurs, handlebar clavicles and brachydactyly. All samples were obtained with approval of the UCL-ICH/Great Ormond Street Hospital Research Ethics Committee (08/H0713/82), South Yorkshire Research Ethics Committee (11/H1310/1) and collaborating institutions with informed consent.
Human Sanger sequencing and PCR. Familial segregation in accordance with a recessive inheritance pattern was conrmed by Sanger sequencing of PCR products amplied from genomic DNA samples of all available family members (SourceBiosciences, Cambridge, UK). For family UCL4, fresh repeat samples were collected for segregation analysis, indicating no chance of sampling error. For RTPCR, the Omniscript kit (Qiagen) was used to make cDNA from total RNA isolated from lymphocytes or broblasts of affected individuals and controls using Trizolchloroform extraction. PCR and RTPCR primers used are listed in Supplementary Table 1.
Western blotting. Uncropped images of all western blots are included in Supplementary Fig. 10.
Constructs used in mammalian cells. Mouse WDR60 and human TCTEX1D2 cDNA were obtained from the University of Queenslands SRC Microarray facility. A sequence-veried SF-TAP-TCTEX1D2 construct (BC021177.2) containing a double Streptavidin II and a single FLAG tag was kindly provided by Nicholas Katsanis (Duke University, Durham, USA). WDR60 was cloned into the GFP-N1 vector by PCR amplication, EcoRI/KpnI digestion and ligation. TCTEX1D2 cloned into a modied pCR3 vector for expression of FLAG-tagged proteins was a kind gift from Dr Jrg Heierhorst (St Vincents Institute of Medical Research, Melbourne). The human tGFP-tagged WDR34 mammalian expression plasmid was obtained from OriGene Technologies, Inc. (TrueORF clone RG204288).
Human cell culture. Human broblasts obtained by skin biopsy from study subjects and HEK293T cells (European Collection of Cell Cultures) were cultured under standard conditions at 37 C and 5% CO2 in DMEM-F12 Glutamax medium (Life Technologies) with 10% fetal bovine serum (Life Technologies).
Immunouorescence in mammalian cells and zebrash. Human broblasts were split onto glass coverslips and grown until conuent. For ciliogenesis experiments, cells were serum starved using cell medium without fetal bovine serum for 20 h. For immunouorescence, cells were then xed in 4% paraformaldehyde (PFA) for 10 min, washed ve times with PBS, treated with 0.05% Triton X-100/PBS for 2 min, washed ve times with PBS, blocked 1 h using 4% BSA in PBS and then incubated with the primary antibody. These were mouse monoclonal anti-acetylated tubulin 1:1,000 (IgG2b, clone 6-11-b1, Sigma), mouse monoclonal anti-pericentrin IgG1 1:200 (mAbcam 28144, Abcam) or rabbit polyclonal anti-IFT88 1:100 (13967-1-AP, Proteintech) overnight at 4 C. Cells were then washed again ve times with PBS and incubated with the appropriate secondary antibodies: goat anti mouseIgG1 Alexa Fluor 647, goat anti-mouse IgG2b Alexa Fluor 568 or goat anti-rabbit Alexa Fluor 488. Glass slides were then washed ve times in PBS, incubated with 4,6-diamidino-2-phenylindole (Molecular Probes, Invitrogen) for 5 min to obtain nuclear stain, washed ve times in PBS and
then mounted in Vectashield (Vector Laboratories). For zebrash immunouorescence studies, embryos were xed at 24 h.p.f. in 4% PFA overnight, washed ve times in PBS containing 0.1% Triton X-100/PBS, incubated in 10% methanol for 30 min at 20 C, washed ve times in PBS containing 0.1% Triton X-100 and
blocked with 5% BSA in PBS containing 0.1% Triton X-100 for 1 h. Embryos were then incubated with the primary antibody mouse monoclonal anti-acetylated tubulin 1:1,000 (IgG2b, clone 6-11-b1, Sigma) or mouse monoclonal anti-gamma tubulin 1:250 (IgG1, Sigma) overnight at 4 C, washed ve times in PBS containing0.1% Triton X-100/PBS and then incubated with the appropriate secondary antibodies: goat anti-mouse IgG1 Alexa Fluor 647 1:1,000 or goat anti-mouse IgG2b Alexa Fluor 488 1:1,000 and 4,6-diamidino-2-phenylindole 1:25,000. All imaging was performed using a Zeiss LSM710 confocal microscope.
Statistical analysis. Statistical analysis of cilia number and cilia length was performed using Students t-test with statistical analysis performed using GraphPad Prism.
Chlamydomonas cells and culture conditions. Chlamydomonas reinhardtii strain 137c (nit1, nit2, mt ) from the Chlamydomonas Resource Center (University of
Minnesota, St Paul, MN) was used as wild type. A54-e18 (nit1-1, ac17, sr1, mt ),
which is the parent of pf16-D2 (a double mutant of pf16 and tctex2b), and pf16-D2 Resc. w/PF16, which is pf16-D2 rescued for the PF16 gene35,60, were from Elizabeth Smith (Dartmouth College, NH). The dic5-1 insertional mutant was generated by transforming g1 cells (nit1, NIT2, mt )61 with the 1.7-kb chimeric
aph700 gene cut by HindIII from the Hyg3 plasmid62, followed by backcrossing of a transformant to wild-type cells. The D1bIC2-HA strain was generated by transformation of dic5-1 with a gene encoding D1bIC2 with a C-terminal 3 HA
tag. Cells were grown in TAP medium63 or M medium I64 altered to have 0.0022 M KH2PO4 and 0.00171 M K2HPO4; cultures were either aerated with 5% CO2 and 95% air or grown on 24-well plates.
Creation of DNA constructs for Chlamydomonas transformation. The wild-type DIC5 gene was amplied by PCR from genomic DNA using primer pairs FAP133-2 (50-TGTCCCGCTGCAGAGCAATG-30) and FAP133-3 (50-ACCCCG
CCTCCTTGTCCTTG-30). The blunt ends of the PCR product were modied by A-tailing, that is, an extra A was added at the 30 ends of the product strands by Taq polymerase. The modied PCR product was cloned into pGEM-T (Promega, Madison, MI). To insert an HA tag into the DIC5 gene just before the stop codon, primer pair FAP133-15 (50-GTAGAGTGGCAGTGCCGGC-30) and FAP133-16 (50-GCGTGAAGTTGCCGCGCA-30) was used to amplify the DIC5 gene and vector. The PCR product was then ligated to the 3 HA fragment excised from
plasmid p3 HA65 by SmaI.
Chlamydomonas immunoprecipitation and mass spectrometry. Flagella membrane-plus-matrix fractions from wild-type and D1bIC2-HA cells were incubated with the anti-HA afnity matrix (Roche Diagnostics GmbH) overnight. The beads were washed three times with HMEK buffer (30 mM HEPES, pH 7.4, 5 mM MgSO4, 0.5 mM EGTA and 25 mM KCl) plus 0.01% NP-40. Protease Inhibitor
Cocktail for plant cell and tissue extracts (Sigma-Aldrich) was added to prevent protein degradation. SDSPAGE sample loading buffer was used to elute the proteins from the matrix. Proteins in the bound fractions from wild-type and D1bIC2-HA samples were then separated by SDSPAGE and stained with Silver Stain Plus (Bio-Rad). Gel regions of interest were excised and analysed by mass spectrometry at the Proteomics and Mass Spectrometry Facility, University of Massachusetts Medical School, and the Vermont Genetics Network Proteomics Facility, University of Vermont66,67.
Flagellar regeneration and analysis of IFT. For DIC imaging of steady-state agella and the subsequent analysis of IFT, live cells were immobilized in 1% agarose and observed using an inverted microscope (Ti U; Nikon) equipped with DIC optics59. To determine the kinetics of agellar regeneration, cells were deagellated by the pH-shock method68 and allowed to regrow agella under the same conditions as before deagellation. Aliquots of the cell suspensions were removed at various times and xed with 1% glutaraldehyde. Images of the xed cells were acquired with an AxioCam camera, AxioVision 3.1 software, and an Axioskop 2 plus microscope (Zeiss). Flagellar lengths were measured using ImageJ (http://rsb.info.nih.gov/ij/index.html
Web End =http://rsb.info.nih.gov/ij/index.html). Images were processed using Adobe Photoshop (Adobe Systems Incorporated, San Jose, CA).
References
1. Fliegauf, M., Benzing, T. & Omran, H. When cilia go bad: cilia defects and ciliopathies. Nat. Rev. Mol. Cell. Biol. 8, 880893 (2007).
2. Hildebrandt, F., Benzing, T. & Katsanis, N. Ciliopathies. N. Engl. J. Med. 364, 15331543 (2011).
3. Pedersen, L.B. & Rosenbaum, J.L. Intraagellar transport (IFT) role in ciliary assembly, resorption and signalling. Curr. Topics Dev. Biol. 85, 2361 (2008).
10 NATURE COMMUNICATIONS | 6:7074 | DOI: 10.1038/ncomms8074 | http://www.nature.com/naturecommunications
Web End =www.nature.com/naturecommunications
& 2015 Macmillan Publishers Limited. All rights reserved.
NATURE COMMUNICATIONS | DOI: 10.1038/ncomms8074 ARTICLE
4. Patel-King, R.S., Gilberti, R.M., Hom, E.F. & King, S.M. WD60/FAP163 is a dynein intermediate chain required for retrograde intraagellar transport in cilia. Mol. Biol. Cell 24, 26682677 (2013).
5. Hom, E.F.Y. et al. A unied taxonomy for ciliary dyneins. Cytoskeleton (Hoboken NJ) 68, 555565 (2011).
6. Pazour, G.J., Wilkerson, C.G. & Witman, G.B. A dynein light chain is essential for the retrograde particle movement of intraagellar transport (IFT). J. Cell Biol. 141, 979992 (1998).
7. Rompolas, P., Pedersen, L.B., Patel-King, R.S. & King, S.M. Chlamydomonas FAP133 is a dynein intermediate chain associated with the retrograde intraagellar transport motor. J. Cell Sci. 120, 36533665 (2007).
8. Mikami, A. et al. Molecular structure of cytoplasmic dynein 2 and its distribution in neuronal and ciliated cells. J. Cell Sci. 115, 48014808 (2002).
9. Grissom, P.M., Vaisberg, E.A. & McIntosh, J.R. Identication of a novel light intermediate chain (D2LIC) for mammalian cytoplasmic dynein 2. Mol. Biol. Cell 13, 817829 (2002).
10. Pster, K.K. et al. Genetic analysis of the cytoplasmic dynein subunit families. PLoS Genet. 2, e1 (2006).
11. Goetz, S.C. & Anderson, K.V. The primary cilium: a signalling centre during vertebrate development. Nat. Rev. Genet. 11, 331344 (2010).
12. Huber, C. et al. WDR34 mutations that cause short-rib polydactyly syndrome type III/severe asphyxiating thoracic dysplasia reveal a role for the NF-kappaB pathway in cilia. Am. J. Hum. Genet. 93, 926931 (2013).
13. Schmidts, M. et al. Mutations in the gene encoding IFT dynein complex component WDR34 cause Jeune asphyxiating thoracic dystrophy. Am. J. Hum. Genet. 93, 932944 (2013).
14. McInerney-Leo, A.M. et al. Short-rib polydactyly and Jeune syndromes are caused by mutations in WDR60. Am. J. Hum. Genet. 93, 515523 (2013).
15. Dagoneau, N. et al. DYNC2H1 mutations cause asphyxiating thoracic dystrophy and short rib-polydactyly syndrome, type III. Am. J. Hum. Genet. 84, 706711 (2009).
16. Merrill, A.E. et al. Ciliary abnormalities due to defects in the retrograde transport protein DYNC2H1 in short-rib polydactyly syndrome. Am. J. Hum. Genet. 84, 542549 (2009).
17. Huber, C. & Cormier-Daire, V. Ciliary disorder of the skeleton. Am. J. Med. Genet. C Semin Med. Genet. 160C, 165174 (2012).
18. Halbritter, J. et al. Defects in the IFT-B component IFT172 cause Jeuneand Mainzer-Saldino syndromes in humans. Am. J. Hum. Genet. 93, 915925 (2013).
19. Tuz, K. et al. Mutations in CSPP1 cause primary cilia abnormalities and Joubert syndrome with or without Jeune asphyxiating thoracic dystrophy. Am. J. Hum. Genet. 94, 6272 (2014).
20. Beales, P.L. et al. IFT80, which encodes a conserved intraagellar transport protein, is mutated in Jeune asphyxiating thoracic dystrophy. Nat. Genet. 39, 727729 (2007).
21. Perrault, I. et al. Mainzer-Saldino syndrome is a ciliopathy caused by IFT140 mutations. Am. J. Hum. Genet. 90, 864870 (2012).
22. Bredrup, C. et al. Ciliopathies with skeletal anomalies and renal insufciency due to mutations in the IFT-A gene WDR19. Am. J. Hum. Genet. 89, 634643 (2011).
23. Davis, E.E. et al. TTC21B contributes both causal and modifying alleles across the ciliopathy spectrum. Nat. Genet. 43, 189196 (2011).
24. Shaheen, R. et al. A founder CEP120 mutation in Jeune asphyxiating thoracic dystrophy expands the role of centriolar proteins in skeletal ciliopathies. Hum. Mol. Genet. 24, 14101419 (2014).
25. Miller, K.A. et al. Cauli: a mouse strain with an Ift140 mutation that results in a skeletal ciliopathy modelling Jeune syndrome. PLoS Genet. 9, e1003746 (2013).
26. Ocbina, P.J., Eggenschwiler, J.T., Moskowitz, I. & Anderson, K.V. Complex interactions between genes controlling trafcking in primary cilia. Nat. Genet. 43, 547553 (2011).
27. Schulte-Merker, S. & Stainier, D.Y.R. Out with the old, in with the new: reassessing morpholino knockdowns in light of genome editing technology. Development 141, 31033104 (2014).
28. Kok, F.O. et al. Reverse genetic screening reveals poor correlation between morpholino-induced and mutant phenotypes in zebrash. Dev. Cell 32, 97108 (2015).
29. Tsujikawa, M. & Malicki, J. Intraagellar transport genes are essential for differentiation and survival of vertebrate sensory neurons. Neuron 42, 703716 (2004).
30. Kramer-Zucker, A.G. et al. Cilia-driven uid ow in the zebrash pronephros, brain and Kupffers vesicle is required for normal organogenesis. Development 132, 19071921 (2005).
31. Lunt, S.C., Haynes, T. & Perkins, B.D. Zebrash ift57, ift88, and ift172 intraagellar transport mutants disrupt cilia but do not affect hedgehog signaling. Dev. Dyn. 238, 17441759 (2009).
32. Ryan, S. et al. Rapid identication of kidney cyst mutations by whole exome sequencing in zebrash. Development 140, 44454451 (2013).
33. Sun, Z. et al. A genetic screen in zebrash identies cilia genes as a principal cause of cystic kidney. Development 131, 40854093 (2004).
34. Schmidts, M. et al. Exome sequencing identies DYNC2H1 mutations as a common cause of asphyxiating thoracic dystrophy (Jeune syndrome) without major polydactyly, renal or retinal involvement. J. Med. Genet. 50, 309323 (2013).
35. DiBella, L.M., Smith, E.F., Patel-King, R.S., Wakabayashi, K.-i. & King, S.M A novel Tctex2-related light chain is required for stability of inner dynein arm I1 and motor function in the Chlamydomonas agellum. J. Biol. Chem. 279, 2166621676 (2004).
36. Pazour, G.J., Agrin, N., Leszyk, J. & Witman, G.B. Proteomic analysis of a eukaryotic cilium. J. Cell Biol. 170, 103113 (2005).
37. Yang, P. & Sale, W.S. The Mr 140,000 intermediate chain of Chlamydomonas agellar inner arm dynein is a WD-repeat protein implicated in dynein arm anchoring. Mol. Biol. Cell 9, 33353349 (1998).
38. El Hokayem, J. et al. NEK1 and DYNC2H1 are both involved in short rib polydactyly Majewski type but not in Beemer Langer cases. J. Med. Genet. 49, 227233 (2012).
39. Baujat, G. et al. Asphyxiating thoracic dysplasia: clinical and molecular review of 39 families. J. Med. Genet. 50, 9198 (2013).
40. Pazour, G.J. et al. Chlamydomonas IFT88 and its mouse homologue, polycystic kidney disease gene tg737, are required for assembly of cilia and agella. J. Cell Biol. 151, 709718 (2000).
41. Marszalek, J.R., Ruiz-Lozano, P., Roberts, E., Chien, K.R. & Goldstein, L.S. Situs inversus and embryonic ciliary morphogenesis defects in mouse mutants lacking the KIF3A subunit of kinesin-II. Proc. Natl Acad. Sci. USA 96, 50435048 (1999).
42. Ashe, A. et al. Mutations in mouse Ift144 model the craniofacial, limb and rib defects in skeletal ciliopathies. Hum. Mol. Genet. 21, 18081823 (2012).43. Pazour, G.J., Dickert, B.L. & Witman, G.B. The DHC1b (DHC2) isoform of cytoplasmic dynein is required for agellar assembly. J. Cell Biol. 144, 473481 (1999).
44. Porter, M.E., Bower, R., Knott, J.A., Byrd, P. & Dentler, W. Cytoplasmic dynein heavy chain 1b is required for agellar assembly in Chlamydomonas. Mol. Biol. Cell 10, 693712 (1999).
45. Hou, Y., Pazour, G.J. & Witman, G.B. A dynein light intermediate chain, D1bLIC, is required for retrograde intraagellar transport. Mol. Biol. Cell 15, 43824394 (2004).
46. Iomini, C., Babaev-Khaimov, V., Sassaroli, M. & Piperno, G. Protein particles in Chlamydomonas agella undergo a transport cycle consisting of four phases.J. Cell Biol. 153, 1324 (2001).47. Engel, B.D. et al. The role of retrograde intraagellar transport in agellar assembly, maintenance, and function. J. Cell Biol. 199, 151167 (2012).
48. Lin, H., Nauman, N.P., Albee, A.J., Hsu, S. & Dutcher, S.K. New mutations in agellar motors identied by whole genome sequencing in Chlamydomonas. Cilia 2, 14 (2013).
49. Asante, D., Stevenson, N.L. & Stephens, D.J Subunit composition of the human cytoplasmic dynein-2 complex. J. Cell Sci. 127, 47744787 (2014).
50. Witman, G.B. in Dyneins: Structure, Biology and Disease. (ed. King, S.M.) 395421 (Elsevier Inc., 2012).
51. King, S.M. in Dyneins: Structure, Biology and Disease. (ed. King, S.M.) 209243 (Elsevier, Inc., 2012).
52. DiBella, L.M. et al. The Tctex1/Tctex2 class of dynein light chains. Dimerization, differential expression, and interaction with the LC8 protein family. J. Biol. Chem. 276, 1436614373 (2001).
53. Goggolidou, P. et al. ATMIN is a transcriptional regulator of both lung morphogenesis and ciliogenesis. Development 141, 39663977 (2014).
54. Lee, J.E. & Gleeson, J.G. A systems-biology approach to understanding the ciliopathy disorders. Genome Med. 3, 59 (2011).
55. Khanna, H. et al. A common allele in RPGRIP1L is a modier of retinal degeneration in ciliopathies. Nat. Genet. 41, 739745 (2009).
56. Kiesewetter, S. et al. A mutation in CFTR produces different phenotypes depending on chromosomal background. Nat. Genet. 5, 274278 (1993).
57. Brown, J.M., Fine, N.A., Pandiyan, G., Thazhath, R. & Gaertig, J. Hypoxia regulates assembly of cilia in suppressors of Tetrahymena lacking an intraagellar transport subunit gene. Mol. Biol. Cell 14, 31923207 (2003).
58. Hou, Y. et al. Functional analysis of an individual IFT protein: IFT46 is required for transport of outer dynein arms into agella. J. Cell Biol. 176, 653665 (2007).
59. Craige, B. et al. CEP290 tethers agellar transition zone microtubules to the membrane and regulates agellar protein content. J. Cell Biol. 190, 927940 (2010).
60. Smith, E.F. & Lefebvre, P.A. PF16 encodes a protein with armadillo repeats and localizes to a single microtubule of the central apparatus in Chlamydomonas agella. J. Cell Biol. 132, 359370 (1996).
61. Wilkerson, C.G., King, S.M., Koutoulis, A., Pazour, G.J. & Witman, G.B.
The 78,000 M(r) intermediate chain of Chlamydomonas outer arm dynein isa
NATURE COMMUNICATIONS | 6:7074 | DOI: 10.1038/ncomms8074 | http://www.nature.com/naturecommunications
Web End =www.nature.com/naturecommunications 11
& 2015 Macmillan Publishers Limited. All rights reserved.
ARTICLE NATURE COMMUNICATIONS | DOI: 10.1038/ncomms8074
WD-repeat protein required for arm assembly. J. Cell Biol. 129, 169178 (1995).62. Berthold, P., Schmitt, R. & Mages, W. An engineered Streptomyces hygroscopicus aph 7" gene mediates dominant resistance against hygromycin B in Chlamydomonas reinhardtii. Protist 153, 401412 (2002).
63. Gorman, D.S. & Levine, R.P. Cytochrome f and plastocyanin: their sequence in the photosynthetic electron transport chain of Chlamydomonas reinhardi. Proc. Natl Acad. Sci. USA 54, 16651669 (1965).
64. Sager, R. & Granick, S. Nutritional studies with Chlamydomonas reinhardi. Ann N Y Acad. Sci. 56, 831838 (1953).
65. Silow, C.D. et al. The V1 Protein in Chlamydomonas localizes in a rotationally asymmetric pattern at the distal ends of the basal bodies. J. Cell Biol. 153, 6374 (2001).
66. Lechtreck, K.F., Luro, S., Awata, J. & Witman, G.B. HA-tagging of putative agellar proteins in Chlamydomonas reinhardtii identies a novel protein of intraagellar transport complex B. Cell Motil. Cytoskeleton 66, 469482 (2009).
67. Awata, J., Takada, S., Standley, C., Lechtreck, K.F., Bellv, K.F., Pazour, G.J., Fogarty, K.E. & Witman, G.B. Nephrocystin-4 controls ciliary trafcking of membrane and large soluble proteins at the transition zone. J. Cell Sci. 127, 47144727 (2014).
68. Witman, G.B., Carlson, K., Berliner, J. & Rosenbaum, J.L. Chlamydomonas agella. I. Isolation and electrophoretic analysis of microtubules, matrix, membranes, and mastigonemes. J. Cell Biol. 54, 507539 (1972).
Acknowledgements
We thank Sarah Bond (Genetics and Genomics Medicine Unit, ICH, UCL) for assistance with Sanger Sequencing, Dr Elizabeth Smith (Dartmouth College) for providing Chlamydomonas strains, and Dr John Leszyk (UMMS Proteomics and Mass Spectrometry Facility) and Dr Bin Deng (Vermont Genetics Network Proteomics Facility, University of Vermont) for help with mass spectrometry. Andrew Phillips of HGMD (Cardiff) assisted in nomenclature for the TCTEX1D2 exon 12 mutation. The TCTEX1D2 SF-TAP construct was kindly provided by N. Katsanis and J. Willer. We are grateful to the UK10K consortium, in particular, the Rare Diseases Group for making this study possible; a full list of the UK10K investigators is available at http://www.uk10k.org/publications_and_posters.html
Web End =http://www.uk10k.org/ http://www.uk10k.org/publications_and_posters.html
Web End =publications_and_posters.html . Funding for UK10K was provided by the Wellcome Trust under award WT091310. This research was supported by National Institutes of Health (NIH) grants RO1 AR062651 and R01 AR066124 (to D.K.), R01 GM051293 (to S.M.K.) and R37 GM030626 (to G.B.W.), by the Robert W. Booth Endowment at UMMS (to G.B.W.) and by an NIH National Institute of General Medical Sciences Institutional Development Award (IDeA) P20 GM103449 (to the Vermont Genetics Network). C.A.J. acknowledges funding from a Sir Jules Thorn Award for Biomedical Research (JTA/09). W.H. was supported by a stipend from the Rosetrees Trust (grant A465). P.J.S. is funded by the British Heart Foundation. H.K. acknowledges funding from the Scientic and Technological Research Council of Turkey (TUBITAK, grant number 112S398) supported by the overall consortium, CRANIRARE-2, the European Research Area Network (ERA-Net) for research programmes on rare diseases (2011-2015). C.W. is supported by an Australian National Health and Medical Research Council grant (APP1045464) and a
University of Queensland (UQ) Vice Chancellors Senior Research Fellowship, and C.R.C. by a UQ International PhD Scholarship. M.U. and K.B. acknowledge funding from FP7 grant agreement no. 278568, PRIMES. R.R. is supported by the Netherlands Organization for Scientic Research (NWO Vici-865.12.005). M.U., P.L.B., R.R. and C.A.J. acknowledge funding from the European Communitys Seventh Framework Programme FP7/2009 under grant agreement no: 241955, SYSCILIA. M.S., P.L.B. and R.R. acknowledge funding from the Dutch Kidney Foundation (CP11.18). P.L.B. and H.M.M. are supported by the Great Ormond Street Hospital Childrens Charity. M.S. is supported by an Action Medical Research UK Clinical Training Fellowship (RTF-1411), a Radboud Excellence Initiative and Radboud Hypatia Fellowship, and acknowledges funding from the German Research Foundation (DFG; Collaborative Research Center 1140, KIDGEM).
Author contributions
M.S., Y.H., H.M.M. and G.B.W. conceived the study and M.S., Y.H., S.M.K., C.W., R.R., H.M.M. and G.B.W. wrote the manuscript. M.S., Y.H., C.R.C., D.A.M., C.H., K.B., J.v.R., M.U., C.W., V.C.-D., R.R., H.M.M. and G.B.W. supervised experiments and were involved in data interpretations. M.S. and S.P.T. performed whole-exome sequencing analysis and M.S. performed zebrash experiments and immunouorescence analysis of human broblasts. Y.H. performed Chlamydomonas experiments. S.M.K. prepared antibodies against Chlamydomonas dynein light chains. C.R.C. performed immunoprecipitations of human TCTEX1D2. C.H. and J.-M.P. performed linkage analysis in human subjects. M.P., Z.M.Y. and W.H. performed Sanger Sequencing of human samples. D.A.M., J.v.R., S.E.C.v.B., S.J.F.L. and R.R. performed mammalian proteomics experiments and proteomic bioinformatics analysis. K.B. and M.U. performed mammalian proteomics mass spectrometry analysis. M.S., C.A.J., P.J.S., H.K., D.K., P.L.B. L.A.-G.,V.C.-D. and H.M.M. were involved in patient enrolment. UK10K Consortium provided whole-exome sequencing data.
Additional information
Supplementary Information accompanies this paper at http://www.nature.com/naturecommunications
Web End =http://www.nature.com/ http://www.nature.com/naturecommunications
Web End =naturecommunications
Competing nancial interests: The authors declare no competing nancial interests.
Reprints and permission information is available online at http://npg.nature.com/reprintsandpermissions/
Web End =http://npg.nature.com/ http://npg.nature.com/reprintsandpermissions/
Web End =reprintsandpermissions/
How to cite this article: Schmidts, M. et al. TCTEX1D2 mutations underlie Jeune asphyxiating thoracic dystrophy with impaired retrograde intraagellar transport. Nat. Commun. 6:7074 doi: 10.1038/ncomms8074 (2015).
This work is licensed under a Creative Commons Attribution 4.0 International License. The images or other third party material in this article are included in the articles Creative Commons license, unless indicated otherwise in the credit line; if the material is not included under the Creative Commons license, users will need to obtain permission from the license holder to reproduce the material. To view a copy of this license, visit http://creativecommons.org/licenses/by/4.0/
Web End =http://creativecommons.org/licenses/by/4.0/
Saeed Al-Turki18,19, Carl Anderson18, Richard Anney20, Dinu Antony1, Jennifer Asimit18, Mohammad Ayub21, Jeff Barrett18, Ins Barroso18, Phil Beales1,15, Jamie Bentham22, Shoumo Bhattacharya22, Douglas Blackwood23, Martin Bobrow24, Elena Bochukova25, Patrick Bolton26, Chris Boustred27, Gerome Breen26,28, Marie-Jo Brion27, Andrew Brown18, Mattia Calissano29, Keren Carss18, Krishna Chatterjee25, Lu Chen18,30, Sebhattin Cirak29, Peter Clapham18, Gail Clement31, Guy Coates18, David Collier32,33, Catherine Cosgrove22, Tony Cox18,Nick Craddock34,35, Lucy Crooks18, Sarah Curran26, Allan Daly18, Petr Danecek18, Smith George Davey27, Aaron Day-Williams18,36, Ian Day27, Richard Durbin18, Sarah Edkins18, Peter Ellis18, David Evans27,I Sadaf Farooqi25, Ghazaleh Fatemifar27, David Fitzpatrick37, Paul Flicek38, Jamie Floyd18, A Reghan Foley29, Chris Franklin18, Marta Futema39, Louise Gallagher19, Tom Gaunt27, Daniel Geschwind40, Celia Greenwood41,42, Detelina Grozeva24, Xiaosen Guo43, Hugh Gurling44, Deborah Hart31, Audrey Hendricks18, Peter Holmans34,35, Jie Huang18, Steve E. Humphries39, Matt Hurles18, Pirro Hysi31, David Jackson18, Yalda Jamshidi45,David Jewell27, Joyce Chris18, Jane Kaye46, Thomas Keane18, John Kemp27, Karen Kennedy18, Alastair Kent47, Anja Kolb-Kokocinski18, Genevieve Lachance31, Cordelia Langford18, Irene Lee48, Rui Li41,49, Yingrui Li43,
12 NATURE COMMUNICATIONS | 6:7074 | DOI: 10.1038/ncomms8074 | http://www.nature.com/naturecommunications
Web End =www.nature.com/naturecommunications
& 2015 Macmillan Publishers Limited. All rights reserved.
NATURE COMMUNICATIONS | DOI: 10.1038/ncomms8074 ARTICLE
Liu Ryan50, Jouko Lnnqvist51, Margarida Lopes18,52, Daniel G. MacArthur18,53,54, Mangino Massimo31, Jonathan Marchini55, John Maslen18, Shane McCarthy18, Peter McGufn26, Andrew McIntosh6,Andrew McKechanie6, Andrew McQuillin44, Yasin Memari18, Sarah Metrustry31, Josine Min27,Alireza Moayyeri31, James Morris18, Dawn Muddyman18, Francesco Muntoni29, Kate Northstone27,Michael ODonovan34,35, Stephen ORahilly25, Alexandros Onoufriadis1, Karim Oualkacha56, Michael Owen34,35, Aarno Palotie18,57, Kalliope Panoutsopoulou18, Victoria Parker25, Jeremy Parr58, Lavinia Paternoster27,Tiina Paunio51, Felicity Payne18, John Perry31,52,59,60, Olli Pietilainen18,51,57, Vincent Plagnol61, Michael A. Quail18, Lydia Quaye31, Lucy Raymond24, Karola Rehnstrm18, J. Brent Richards31,41,49, Sue Ring27,Graham R. S. Ritchie18,38, David B. Savage25, Nadia Schoenmakers25, Robert K. Semple25, Eva Serra18, Hashem Shihab27, So-Youn Shin18, David Skuse48, Kerrin Small31, Carol Smee18, Artigas Mara Soler62,Nicole Soranzo18, Lorraine Southam18, Tim Spector31, Beate St Pourcain27, David St. Clair63, Jim Stalker18, Gabriela Surdulescu31, Jaana Suvisaari51, Ioanna Tachmazidou18, Jing Tian43, Nic Timpson27, Martin Tobin62, Ana Valdes31, Margriet van Kogelenberg18, Parthiban Vijayarangakannan18, Louise Wain62, Klaudia Walter18, Jun Wang43, Kirsten Ward31, Ellie Wheeler18, Ros Whittall39, Hywel Williams34,35, Kathy Williamson37,Scott G. Wilson31,64,65, Kim Wong18, Tamieka Whyte29, Xu ChangJiang41, Eleftheria Zeggini18,
Feng Zhang31 & Hou-Feng Zheng41,49
18 The Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton CB10 1HH, Cambridge, UK. 19 Department of Pathology, King Abdulaziz Medical City, Riyadh, Saudi Arabia. 20 Department of Psychiatry, Trinity Centre for Health Sciences, St. James Hospital, Jamess Street, Dublin 8, Ireland.
21 Durham University School of Medicine, Pharmacy and Health, Wolfson Research Institute, Queens Campus, Stockton-on-Tees TS17 6BH, UK.
22 Department of Cardiovascular Medicine and Wellcome Trust Centre for Human Genetics, Roosevelt Drive, Oxford OX3 7BN, UK. 23 Division of Psychiatry, The University of Edinburgh, Royal Edinburgh Hospital, Edinburgh EH10 5HF, UK. 24 Department of Medical Genetics, Cambridge Institute for Medical Research, University of Cambridge, Cambridge CB2 2XY, UK. 25 University of Cambridge Metabolic Research Laboratories, and NIHR Cambridge Biomedical Research Centre, Institute of Metabolic Science, Addenbrookes Hospital, Cambridge CB2 0QQ, UK. 26 Institute of Psychiatry, Kings College London, 16 De Crespigny Park, London SE5 8AF, UK. 27 MRC CAiTE Centre, School of Social and Community Medicine, University of Bristol, Oakeld House, Oakeld Grove, Clifton, Bristol BS8 2BN, UK. 28 NIHR BRC for Mental Health, Institute of Psychiatry and SLaM NHS Trust, Kings College London, 16 De Crespigny Park, London SE5 8AF, UK. 29 Dubowitz Neuromuscular Centre, UCL Institute of child health & Great Ormond Street Hospital, London WC1N 1EH, UK.
30 Department of Haematology, University of Cambridge, Long Road, Cambridge CB2 0PT, UK. 31 The Department of Twin Research & Genetic Epidemiology, Kings College London, St Thomas Campus, Lambeth Palace Road, London SE1 7EH, UK. 32 Social, Genetic and Developmental Psychiatry Centre, Institute of Psychiatry, Kings College London, Denmark Hill, London SE5 8AF, UK. 33 Lilly Research Laboratories, Eli Lilly & Co. Ltd., Erl Wood Manor, Sunninghill Road, Windlesham, Surrey, UK. 34 MRC Centre for Neuropsychiatric Genetics & Genomics, Institute of Psychological Medicine & Clinical Neurosciences, School of Medicine, Cardiff University, Cardiff CF14 4XN. 35 Barts and the London School of Medicine and Dentistry, East Lindon NHS Foundation Trust, London, UK.
36 Department of Translational Sciences, Biogen Idec, 14 Cambridge Center, Cambridge, MA 02142, USA. 37 MRC Human Genetics Unit, MRC Institute of Genetic and Molecular Medicine, at the University of Edinburgh, Western General Hospital, Edinburgh, EH4 2XU, UK. 38 European Bioinformatics Institute, Wellcome Trust Genome Campus, Hinxton, Cambridge, CB10 1SD, UK. 39 Cardiovascular Genetics, BHF Laboratories, Rayne Building, Institute Cardiovascular Sciences, University College London, London WC1E 6JJ, UK. 40 UCLA David Geffen School of Medicine, Los Angeles, California, USA. 41 Departments of Epidemiology, Biostatistics and Occupational Health, Lady Davis Institute, Jewish General Hospital, McGill University, Montreal, Quebec, Canada.
42 Department of Oncology, McGill University, Montreal, Quebec, Canada. 43 BGI-Shenzhen, Shenzhen 518083, China. 44 Molecular Psychiatry Laboratory, Mental Health Sciences Unit, University College London, 21 University St. Rockefeller Building, London WC1E 6BT, UK. 45 Human Genetics Research Centre, St Georges University of London, UK. 46 HeLEXCentre for Health, Law and Emerging Technologies, Department of Public Health, University of Oxford, Old Road Campus, Oxford, OX3 7LF, UK. 47 Genetic Alliance UK, 4D Leroy House, 436 Essex Road, London N1 3QP, UK. 48 Behavioural and Brain Sciences Unit, UCL Institute of Child Health, London, WC1N 1EH, UK. 49 Departments of Medicine & Human Genetics, Lady Davis Institute, Jewish General Hospital, McGill University, Montreal, Quebec, Canada.. 50 BGI-Europe, London. 51 National Institute for Health and Welfare (THL), Helsinki. 52 Wellcome Trust Centre for Human Genetics, Roosevelt Drive, Oxford, OX3 7BN, UK. 53 Analytic and Translational Genetics Unit, Massachusetts General Hospital, Boston MA 02113, USA. 54 Program in Medical and Population Genetics, Broad Institute of Harvard and MIT, Cambridge MA 02132, USA. 55 Department of Statistics, University of Oxford, 1 South Parks Road, Oxford OX1 3TG, UK. 56 Department of Mathematics, Universit de Qubec Montral, Montral, Qubec, Canada.
57 Institute for Molecular Medicine Finland (FIMM), University of Helsinki, Helsinki, Finland. 58 Institute of Neuroscience, Henry Wellcome Building for Neuroecology, Newcastle University, Framlington Place, Newcastle upon Tyne, NE2 4HH, UK. 59 Genetics of Complex Traits, Peninsula Medical School, University of Exeter, Exeter, UK. 60 Center for Statistical Genetics, University of Michigan, Ann Arbor, Michigan, USA. 61 University College London (UCL) Genetics Institute (UGI) Gower Street, London, WC1E 6BT, UK. 62 Departments of Health Sciences and Genetics, University of Leicester, Leicester, UK.
63 Institute of Medical Sciences, University of Aberdeen, AB25 2ZD, UK. 64 School of Medicine and Pharmacology, University of Western Australia, Perth, WA, Australia. 65 Department of Endocrinology and Diabetes, Sir Charles Gairdner Hospital, Nedlands, WA, Australia
NATURE COMMUNICATIONS | 6:7074 | DOI: 10.1038/ncomms8074 | http://www.nature.com/naturecommunications
Web End =www.nature.com/naturecommunications 13
& 2015 Macmillan Publishers Limited. All rights reserved.
You have requested "on-the-fly" machine translation of selected content from our databases. This functionality is provided solely for your convenience and is in no way intended to replace human translation. Show full disclaimer
Neither ProQuest nor its licensors make any representations or warranties with respect to the translations. The translations are automatically generated "AS IS" and "AS AVAILABLE" and are not retained in our systems. PROQUEST AND ITS LICENSORS SPECIFICALLY DISCLAIM ANY AND ALL EXPRESS OR IMPLIED WARRANTIES, INCLUDING WITHOUT LIMITATION, ANY WARRANTIES FOR AVAILABILITY, ACCURACY, TIMELINESS, COMPLETENESS, NON-INFRINGMENT, MERCHANTABILITY OR FITNESS FOR A PARTICULAR PURPOSE. Your use of the translations is subject to all use restrictions contained in your Electronic Products License Agreement and by using the translation functionality you agree to forgo any and all claims against ProQuest or its licensors for your use of the translation functionality and any output derived there from. Hide full disclaimer
Copyright Nature Publishing Group Jun 2015
Abstract
The analysis of individuals with ciliary chondrodysplasias can shed light on sensitive mechanisms controlling ciliogenesis and cell signalling that are essential to embryonic development and survival. Here we identify TCTEX1D2 mutations causing Jeune asphyxiating thoracic dystrophy with partially penetrant inheritance. Loss of TCTEX1D2 impairs retrograde intraflagellar transport (IFT) in humans and the protist Chlamydomonas, accompanied by destabilization of the retrograde IFT dynein motor. We thus define TCTEX1D2 as an integral component of the evolutionarily conserved retrograde IFT machinery. In complex with several IFT dynein light chains, it is required for correct vertebrate skeletal formation but may be functionally redundant under certain conditions.
You have requested "on-the-fly" machine translation of selected content from our databases. This functionality is provided solely for your convenience and is in no way intended to replace human translation. Show full disclaimer
Neither ProQuest nor its licensors make any representations or warranties with respect to the translations. The translations are automatically generated "AS IS" and "AS AVAILABLE" and are not retained in our systems. PROQUEST AND ITS LICENSORS SPECIFICALLY DISCLAIM ANY AND ALL EXPRESS OR IMPLIED WARRANTIES, INCLUDING WITHOUT LIMITATION, ANY WARRANTIES FOR AVAILABILITY, ACCURACY, TIMELINESS, COMPLETENESS, NON-INFRINGMENT, MERCHANTABILITY OR FITNESS FOR A PARTICULAR PURPOSE. Your use of the translations is subject to all use restrictions contained in your Electronic Products License Agreement and by using the translation functionality you agree to forgo any and all claims against ProQuest or its licensors for your use of the translation functionality and any output derived there from. Hide full disclaimer