#PAGE_PARAMS# #ADS_HEAD_SCRIPTS# #MICRODATA#

Mutations in PIH proteins MOT48, TWI1 and PF13 define common and unique steps for preassembly of each, different ciliary dynein


Authors: Ryosuke Yamamoto aff001;  Shiho Yanagi aff001;  Masahito Nagao aff001;  Yuya Yamasaki aff001;  Yui Tanaka aff001;  Winfield S. Sale aff002;  Toshiki Yagi aff003;  Takahide Kon aff001
Authors place of work: Department of Biological Sciences, Graduate School of Science, Osaka University, Osaka, Japan aff001;  Department of Cell Biology, School of Medicine, Emory University, Atlanta, Georgia, United States of America aff002;  Faculty of Life and Environmental Sciences, Prefectural University of Hiroshima, Shobara, Hiroshima, Japan aff003
Published in the journal: Mutations in PIH proteins MOT48, TWI1 and PF13 define common and unique steps for preassembly of each, different ciliary dynein. PLoS Genet 16(11): e1009126. doi:10.1371/journal.pgen.1009126
Category: Research Article
doi: https://doi.org/10.1371/journal.pgen.1009126

Summary

Ciliary dyneins are preassembled in the cytoplasm before being transported into cilia, and a family of proteins containing the PIH1 domain, PIH proteins, are involved in the assembly process. However, the functional differences and relationships between members of this family of proteins remain largely unknown. Using Chlamydomonas reinhardtii as a model, we isolated and characterized two novel Chlamydomonas PIH preassembly mutants, mot48-2 and twi1-1. A new allele of mot48 (ida10), mot48-2, shows large defects in ciliary dynein assembly in the axoneme and altered motility. A second mutant, twi1-1, shows comparatively smaller defects in motility and dynein assembly. A double mutant mot48-2; twi1-1 displays greater reduction in motility and in dynein assembly compared to each single mutant. Similarly, a double mutant twi1-1; pf13 also shows a significantly greater defect in motility and dynein assembly than either parent mutant. Thus, MOT48 (IDA10), TWI1 and PF13 may define different steps, and have partially overlapping functions, in a pathway required for ciliary dynein preassembly. Together, our data suggest the three PIH proteins function in preassembly steps that are both common and unique for different ciliary dyneins.

Keywords:

Cilia – Cytoplasm – Dyneins – Chlamydomonas reinhardtii – Immunoblotting – Motor proteins – Phenotypes – Swimming

Introduction

Motile cilia (also interchangeably referred to as flagella) are intriguing antenna-like organelles that play various important roles in eukaryotes [1, 2]. In lower eukaryotes such as Paramecium and Trypanosoma, these organelles play an indispensable role in cell motility. In higher eukaryotes including humans, cilia are essential for proper development, fertilization, and homeostasis. Defects in ciliary motility cause various symptoms including situs inversus, infertility, congenital heart disease and hydrocephalus in humans, collectively called as primary ciliary dyskinesia (PCD) [3, 4]. While diagnosis of PCD has attracted a good deal of attention, diagnosis can be difficult and a permanent treatment for PCD has not been established [5, 6].

The motility of cilia is driven by gigantic motor-protein complexes, referred to as ciliary dyneins that are composed of several subunits (HC: heavy chain, IC: intermediate chain, LC: light chain) and located on ciliary microtubules [1, 79]. Ciliary dyneins are classified into two major classes: outer dynein arm (ODA) and inner dynein arm (IDA). ODAs are particularly important for the high beat frequency of cilia, whereas IDAs are essential for creating a proper ciliary waveform [10]. Large ciliary components, including ciliary dyneins, are first assembled in the cytoplasm before being transported into and within the cilia by the intra-flagellar transport (IFT) mechanism [1113]. This process is referred to as cytoplasmic preassembly, and many factors that are essential for the preassembly of ciliary dyneins (referred to as preassembly factors) have been reported increasing our understanding of this enigmatic process [14, 15]. Moreover, defects in the preassembly of ciliary dyneins understandably cause motility defects in cilia, resulting in PCD in humans [1620]. In spite of its importance, the detailed mechanism of ciliary dynein preassembly in the cytoplasmic compartment, and the relationship between each preassembly factor, largely remain obscure.

In recent studies aimed at understanding of the dynein preassembly mechanism, a family of chaperone-cofactor-like proteins referred to as PIH proteins, which contain a Protein Interacting with HSP90 1 (PIH1) domain, have been shown to be tightly linked to this process [13, 17, 21] (Table 1). In vertebrates, at least four main PIH proteins (DNAAF2/KTU, PIH1D1, PIH1D2, and DNAAF6/PIH1D3) have been identified to date [13, 21, 22], and each protein has been shown to play a role in ciliary dynein preassembly [22] (Table 1), possibly residing in cytoplasmic complexes including the dynein axonemal particles (DynAPs) [23]. In Chlamydomonas reinhardtii, a ciliated green alga, PF13, a DNAAF2/KTU orthologue, has been shown to play an important role in the preassembly of ODAs as well as one species of IDA (IDA c)[13, 17]. Another PIH protein, MOT48 (also known as IDA10) has been shown to be necessary for the preassembly of the ODAs and a fraction of several IDA species (IDAs b, c, d, and e)[13]. In addition, a third PIH protein, TWI1 was identified in Chlamydomonas as an orthologue of DNAAF6/PIH1D3 [13, 21]. Among these three Chlamydomonas PIH proteins, knowledge of the precise function of MOT48 and TWI1 is limited, partly due to the lack of mutant alleles of the genes encoding the PIH proteins. Only the original mutant allele of mot48, mot48-1 (ida10-1), was available for study [13]. A recent report briefly described the phenotype of a twi1 mutant found in the CLiP library [24] as similar to wild-type [25], suggesting that there is no relationship between TWI1 and dynein preassembly. However, neither detailed study of ciliary dynein assembly nor an examination of the relationship of TWI1 to the other PIH proteins has been performed.

Tab. 1. PIH proteins involved in dynein preassembly.
PIH proteins involved in dynein preassembly.

In this study, we report the isolation/characterization of a twi1-1 mutant and a new allele of the mot48, mot48-2. The mot48-2 mutants swim more slowly than wild-type and have a large defect in assembly of dyneins in the ciliary axoneme. The twi1-1 mutant has only a slightly reduced motility, as previously described [25], and has only a slight defect in dynein assembly in the axoneme. In addition, a double mutant mot48-2; twi1-1 are more severely defective in motility and dynein assembly than either of the parent PIH mutant strains. Similarly, the double mutant pf13; twi1-1 also shows a more severe phenotype than the parent, single mutants. These results strongly suggest that PIH proteins MOT48, TWI1 and PF13 define different steps, and have partially overlapping functions, in a pathway required for ciliary dynein preassembly.

Results

Isolation and characterization of mot48-2, a novel allele of mot48 (ida10)

We identified a slow swimming strain (LMJ.RY0402.055540) in the Chlamydomonas mutant library (CLiP) [24], with a swimming pattern that was reminiscent of the mot48-1 mutant previously described [13]. The strain carried an additional mutation other than the original APHVIII insertion used to establish the library [24]. After back-crossed to wild type (CC-125) cells, we determined the MOT48 protein [13, 28] is indeed missing from mutant progeny from the cross (Fig 1A). We examined the MOT48 sequence in the mutant progeny, and found a mutation (G>T) in the fourth exon, which results in a pre-mature stop codon. Thus, we named the new mutant allele mot48-2 (Fig 1B) [13]. Immunoblots of whole cell samples from mot48-1 and mot48-2 show no evidence of MOT48 (Fig 1A) [13]. The mutation in mot48-2 is predicted to disrupt the PIH1 domain in MOT48, deleting the potential binding motif for chaperones including HSP90 (Fig 1C) [13, 17, 29, 30]. As has been noted for mot48-1 [13], the mot48-2 mutants typically swim much slower (~ 85 ± 14 μm/s) than wild-type (CC-125 = ~ 136 ± 21 μm/s), although the motility varies slightly from day to day and culture to culture. A transgene expressing the wild-type MOT48 with a 3HA tag at the C-terminus (mot48-2; MOT48::HA) rescued swimming velocity (~ 131 ± 31 μm/s) and expression of the 3HA-tagged MOT48 (Fig 1D). The Mot48 phenotype was also rescued by a transgene expressing MOT48 with an added C-terminal mCherry-3HA tag (mot48-2; MOT48::mCherry-HA)(swimming velocity = ~ 120 ± 21 μm/s)(Fig 1D and S1 Fig).

Characterization of a novel <i>mot48</i> allele, <i>mot48-2</i>.
Fig. 1. Characterization of a novel mot48 allele, mot48-2.
A) Immunoblotting analysis of whole cell samples from wild-type (CC-124, cw15), mot48-1, and two clones of mot48-2 (G2/G5) using an anti-MOT48 antibody. The MOT48 protein band (red arrowhead) is missing in all mot48 strains, although we cannot completely rule out the possibility that tiny amounts of MOT48 are expressed in an altered form in mot48-1. Gray arrowheads: non-specific bands. B) Sequence analysis of the mot48-2 genomic DNA identified a point mutation (G>T) in the fourth exon of MOT48, resulting in a premature stop codon. The MOT48 genomic structure is based on/from Phytozome (v5.5: https://phytozome.jgi.doe.gov/pz/portal.html#!info?alias = Org_Creinhardtii) and JGI (v4: https://genome.jgi.doe.gov/Chlre4/Chlre4.home.html) Chlamydomonas genome databases (Pink: Exon, Blue: Intron, Green: UTR). The mutation sites in mot48-1 and mot48-2 are indicated. C) The molecular structure of the MOT48 protein was predicted using a SMART analysis (http://smart.embl-heidelberg.de/). MOT48 has a PIH1 domain in the middle of its structure (gray). The pink bar represents a low-complexity region. The mot48-1 mutant has a mutation near the C-terminus of the MOT48 molecule [13], while the new allele (mot48-2) has a mutation in the middle of the molecule. D) Immunoblot analyses of whole cell samples of wild-type (cw15), mot48-2, three independent colonies of mot48-2; MOT48::HA (5D, 6B, 7D) and one colony of mot48-2; MOT48::mCherry-HA using the anti-MOT48 and anti-HA antibodies. The MOT48 protein band (black arrowhead) present in the wild-type strain is absent in mot48-2. In the mot48-2; MOT48::HA strain, exogeneous MOT48 with a 3HA tag is expressed in all three independent colonies (green arrowheads). In the mot48-2; MOT48::mCherry-HA, exogeneous MOT48 with a large mCherry-3HA tag is expressed (pink arrowhead), partially rescuing the Mot48 (Ida10) phenotype. Gray arrowheads: non-specific bands.

Since MOT48 has previously been reported to function in ciliary dynein preassembly and mot48-1 cilia lacked a subset of dynein species [13], we assessed ciliary dynein assembly in the mot48-2 mutants. To semi-quantitatively estimate the amount of each dynein species in the mot48-2 cilia compared to wild-type (CC-124), spectral counting experiments were performed on isolated axonemes (Fig 2). Among the 15 species of ciliary dynein HCs present in the Chlamydomonas genome [8, 26], ODA α, IDAs b (DHC5), c (DHC9), and e (DHC8), and three minor dyneins DHC3, DHC4, and DHC11 levels are greatly reduced compared to wild-type axonemes (< 50%). In addition, ODAs β and γ, IDAs a (DHC6), d (DHC2), and g (DHC7), and one minor dynein (DHC12/PCR4 [26, 31]) show a more modest reduction in the mot48-2 mutants (50% ~ 80% of the levels of wild type) (Relationship of dynein subunits in Chlamydomonas and humans is summarized in S1 Table). Immunofluorescent microscopic observation also confirmed the defects of IDA c (DHC9) and DHC11 (a minor species) in the mot48-2 axonemes (S2 Fig). The HCs of the two-headed IDA f/I1 showed only a slight reduction in mot48-2 mutants (Fig 2).

Several IDAs are deficient in the <i>mot48-2</i> ciliary axonemes.
Fig. 2. Several IDAs are deficient in the mot48-2 ciliary axonemes.
Spectral counting results (first set) of ciliary axonemal dyneins in wild-type (CC-124 and cw15), mot48-1, and mot48-2 strains. The spectral numbers observed in the mutants were normalized using spectral numbers observed in the CC-124 wild-type strain. Asterisks indicate the ciliary axonemal dynein species for which the spectral numbers in mot48-2 were below 50% of the levels in the CC-124 strain.

Characterization of the twi1-1 mutant, which lacks a PIH protein required for dynein preassembly

In addition to MOT48, two other PIH proteins have been identified in Chlamydomonas [13]. One is PF13, a protein required for ciliary dynein assembly based on characterization of the dynein-deficient mutant pf13 [32]. Defects of its orthologue in mammals (DNAAF2/KTU) have been reported to cause the ciliopathy [17] (Table 1). The other is the TWI1 protein (Fig 3A) (predicted molecular weight = ~ 20590), and defects in the TWI1 orthologue (DNAAF6/PIH1D3) also cause the ciliopathy [18, 19, 21] (Table 1). In addition, the expression of TWI1 is highly induced upon deciliation [33]. A twi1 strain (LMJ.RY0402.076787) was recently identified in the CliP library [24], and reported that the swimming phenotype of this LMJ.RY0402.076787 strain is similar to wild-type [25]. Thus, we back-crossed the LMJ.RY0402.076787 strain to wild-type (CC-125) and the twi1 progeny (twi1-1) phenotype was characterized. Compared to wild-type Chlamydomonas (CC-125)(~ 136 ± 21 μm/s), the twi1-1 swims at a slightly reduced rate (~ 103 ± 14 μm/s) after the 3-day liquid culture, suggesting twi1-1 had subtle defects in assembly of ciliary dyneins (S1 Fig). Like other mutants defective in genes encoding PIH proteins, the swimming phenotypes vary slightly from day to day.

TWI1 is a dynein preassembly factor present in various ciliary mutants.
Fig. 3. TWI1 is a dynein preassembly factor present in various ciliary mutants.
A) Genomic structure of Chlamydomonas TWI1 gene based on/from the Phytozome (v5.5: https://phytozome.jgi.doe.gov/pz/portal.html#!info?alias = Org_Creinhardtii) and JGI (v4: https://genome.jgi.doe.gov/Chlre4/Chlre4.home.html) genome databases (Pink: Exon, Blue: Intron, Green: UTR). The insertional mutation site in the twi1-1 mutant is based on previous reports [24, 25]. The insertion site of the transposon, MRC1, in some of the pf22/pf23 strains determined in this study is shown with a light blue arrowhead. The molecular structure of the TWI1 protein predicted by SMART analysis (http://smart.embl-heidelberg.de/) is also shown. TWI1 has a PIH1 domain in the middle of its structure (gray). B) Immunoblot of whole cell samples from various IFT-related and dynein preassembly mutants using anti-PF23, MOT48, and TWI1 antibodies. All mutants, except for twi1-1 and pf22 (CC-1382) show the presence of TWI1 in whole cells. As discussed in the text, TWI1 protein was visualized as consisting of two or three bands on immunoblots (pink arrowheads). The pf23 (5–4) strain had a slightly smaller and mutated PF23 protein, as described previously (red arrowhead) [28]. The mot48-2 mutant lacked the MOT48 protein (green arrowhead). C) Immunoblotting of whole cell samples from wild-type (CC-125) and three pf22 strains (pf22 (CC-1382), pf22 (CC-2495), and pf22A (CC-2493)) using an anti-TWI1 antibody. pf22 (CC-1382) lacked the TWI1 protein (pink arrowheads) in whole cells, because of the insertion of the Chlamydomonas transposon, MRC1, in the fourth intron (see A in this figure). D) Immunoblotting of whole cell samples from wild-type (CC-125) and three pf23 strains (pf23 (CC-1383), pf23 (CC-3660), and pf23 (5–4)) using the anti-TWI1 antibody. pf23 (CC-1383) and pf23 (CC-3660) lacked the TWI1 protein (pink arrowheads) in whole cells, because of insertion of the transposon, MRC1. pf23 (5–4) had a normal TWI1 protein. Gray arrowheads: non-specific bands. E) Immunoblottings of the de-ciliated cell body samples from wild-type (CC-125) and single or double PIH mutants using the antibodies against various IDA subunits (p28/IDA4, actin/IDA5, IC138/BOP5, and DHC9/IDA c HC).

An antibody against TWI1 was generated, and we noticed that TWI1 shows two or three bands on immunoblots (Fig 3B-3D). Since these TWI1 bands could be observed even in freshly-boiled Chlamydomonas whole cell SDS-PAGE samples, we presume that these band shifts represent modified forms of TWI1, rather than protein degradation. We assessed if the TWI1 band patterns were altered following treatment with calf intestinal phosphatase, and found that no changes occurred, suggesting these shifts did not arise as a result of TWI1 phosphorylation. Also, we could not identify alternative-splicing variants of TWI1 cDNA. One possibility is that the TWI1 protein is structurally stable, and that boiling/SDS-treatment is not sufficient to completely denature the protein, as has been observed for another preassembly factor CCDC103 [34].

To test if the IFT-related or preassembly-related mutations affect the stability of the TWI1 proteins, we performed the immunoblots using the TWI1 antibody on the whole cell samples from various IFT-related and preassembly-related mutants. Immunoblots reveal the TWI1 protein is present in most IFT-related and preassembly-related mutants (e.g. ift46-1, ift74-1, and oda5)(Fig 3B)(See S1 Table summarizing Chlamydomonas and human proteins). Surprisingly, pf22 (CC-1382) and pf23 (CC-1383 and CC-3660) strains completely lack TWI1 (Fig 3B–3D). In contrast, immunoblots of whole cell samples from pf22 (CC-2495), pf22A (CC-2493), and pf23 (5–4) strains have normal levels of TWI1 (Fig 3C and 3D). The result strongly suggested TWI1 loss in pf22 (CC-1382) and pf23 (CC-1383 and CC-3660) strains occurs because of an additional mutation. TWI1 sequence in these mutant strains revealed the transposon MRC1 (~ 1,600 bp) [35] inserted in the fourth intron of the TWI1 gene (Fig 3A). Since these strains were first isolated in Dr. David Luck’s laboratory in the 1970’s [32], we suspect that some of the parent strains used for mutagenesis in Luck laboratory had this transposon insertion in TWI1 gene, and that these strains (pf22 (CC-1382) and pf23 (CC-1383 and CC-3660)) are actually double mutants lacking their respective proteins (Chlamydomonas PF22 or PF23) and TWI1. Other pf strains from Luck laboratory might also carry this twi1 (twi1-2) background. We carried out spectral counting experiments (Fig 4) of the ciliary axonemal dyneins in the twi1-1 mutant, and found that only the levels of IDAs c (DHC9) and e (DHC8) were modestly reduced compared to wild-type (CC-125). This observation is consistent with the mild motility phenotype of twi1-1.

Dynein defects are profound in double PIH preassembly mutants.
Fig. 4. Dynein defects are profound in double PIH preassembly mutants.
Spectral counting results (second and third sets combined) of ciliary axonemal dyneins from wild-type (CC-125), twi1-1, mot48-2, mot48-2; twi1-1, pf13, pf13; twi1-1 and twi1-1; TWI1::HA. The spectral numbers observed in the mutants were normalized using the spectral numbers from the CC-125 strain. Asterisks indicate the ciliary dynein species for which the spectral numbers in the mot48-2; twi1-1 strain showed more than a 50% reduction compared to the mot48-2 strain. The black triangles indicate the ciliary dynein species for which the spectral numbers in the pf13; twi1-1 strain showed a more than 30% reduction compared to the pf13 strain. DHC11 and DHC12 showed ~ 50% increase in pf13; twi1-1 compared to pf13 (white triangles).

Chlamydomonas PIH proteins MOT48, TWI1, and PF13 have overlapping and unique roles in assembly of different ciliary dyneins

We took advantage of Chlamydomonas genetics by isolating double PIH mutants (mot48-2; pf13, mot48-2; twi1-1 and pf13; twi1-1) from crosses between single PIH mutants (pf13, mot48-2, and twi1-1). The predictions included that if deleted proteins function together in the same path and a phenotype of one mutant is similar or the same as the other, then the double mutant phenotype would nearly match the phenotype of the single mutants. Also, if deleted proteins function together in the same path but a phenotype of one mutant is more severe to the other, then the double mutant phenotype would match the phenotype of the more deleterious single mutant. Alternatively, if the deleted proteins operate in different pathways, or have some overlapping function but do not function together in the same path, then the double mutants would have a more severe phenotype than the single mutants.

The motility phenotype of the mot48-2; twi1-1 double mutant is worse than the mot48-2 single mutant (swimming velocity: mot48-2 = ~ 85 ± 14 μm/s; mot48-2; twi1-1 = ~ 49 ± 13 μm/s). Furthermore, about half of the double mutant cells have completely non-motile cilia while the other half of the cells display a slow swimming phenotype (S1 Fig). In addition, the percentage of ciliated cells of the pf13; twi1-1 double mutant (~ 16%) is much lower than the pf13 (~ 58%) single mutant. These observations strongly suggest that TWI1 protein is involved in ciliary dynein preassembly.

To further test the idea that dynein assembly is more defective in the double mutants, we performed spectral counting experiments on dyneins in isolated axonemes from the double and single PIH preassembly mutants to compare the amount of ciliary dyneins assembled (Fig 4). The mot48-2; pf13 double mutant grew extremely short cilia or was missing cilia. This severe phenotype hindered comparison of the amount of ciliary dynein assembled in this strain. This short cilia phenotype was also previously observed in the mot48-1; pf13 mutant [13]. Predictably, this short-cilia phenomenon was a consequence of pre-assembly failure of sufficient number of ciliary dyneins required for ciliary elongation.

The peptide numbers for a subset of ciliary dyneins in the mot48-2; twi1-1 double mutant are greatly reduced compared to mot48-2 (Fig 4). In particular, the IDAs b (DHC5), c (DHC9), and e (DHC8), and one minor dynein, DHC3 are greatly reduced in the double mutant compared to mot48-2 alone. This result indicates that both MOT48 and TWI1 function in the preassembly of these dynein species, but in possibly different steps. Alternatively, MOT48 and TWI1 have overlapping functions in the same step of preassembly, and loss of the two PIH proteins cause severe defects for some ciliary dyneins. Peptide numbers of ODAs α and β, and IDA b (DHC5) in the pf13; twi1-1 double mutant are modestly reduced compared to the pf13 mutant, indicating that PF13 and TWI1 both function in the preassembly of these dyneins possibly in different steps, and/or have some overlapping function in the same step (see Discussion).

It is intriguing that peptide numbers of some minor dyneins (DHC12 in the mot48-2; twi1-1 double mutant, and DHC11 and DHC12 in the pf13; twi1-1 double mutant) are much greater in the double PIH mutants than in the single PIH mutants (pf13, mot48-2, and twi1-1)(Fig 4). This result suggests that the preassembly of these minor dynein species is not affected by the double PIH mutations and that these minor dyneins partially replace the major dynein species that are affected in these double mutants. While some major IDAs are predicted to be replaced by minor dynein species at the proximal end of the cilia [36, 37], replacement of major dynein species by minor dynein species, especially DHC12 in the double PIH mutants, must be confirmed by future biochemical studies.

In addition, to check the stability of various dynein subunits in the cytoplasm, we performed immunoblots of de-ciliated cell-body samples. Immunoblots of dynein subunits (S1 Table) on the de-ciliated cell-body samples of the single or double PIH mutants revealed that one IDA subunit, p28/IDA4 was apparently reduced in the cell bodies from PIH mutants with mot48 background (Fig 3E). In contrast, another IDA subunit, actin/IDA5 appeared to accumulate in the cell bodies from PIH mutants with the pf13 and/or mot48 background (Fig 3E). The reduction in p28/IDA4 and increase in actin/IDA5 were also previously observed in mot48-1 [13]. In addition, IDA c HC (DHC9) is reduced in the PIH mutants with the mot48 background (Fig 3E).

As a further test, we performed rescue experiments to see if recovery of the TWI1 protein in the twi1-1 and mot48-2; twi1-1 would rescue the observed phenotypes. Exogenously expressed TWI1::3HA proteins in twi1-1; TWI1::HA and mot48-2; twi1-1; TWI1::HA (S2 Table) successfully rescue both the swimming defects (swimming velocity: twi1-1 = ~ 103 ± 14 μm/s; twi1-1; TWI1::HA = ~ 121 ± 21 μm/s; mot48-2; twi1-1 = ~ 49 ± 13 μm/s; mot48-2; twi1-1; TWI1::HA = ~ 71 ± 11 μm/s) and ciliary dynein assembly (Figs 4 and 5A–5C and S1 Fig). Notably, the expressed exogeneous TWI1::3HA proteins by the cDNA rescue show several forms in the immunoblots (Fig 5A), suggesting these variants most likely derive from structural differences or some modification rather than alternative splicing or protein degradation. These results indicate that the observed phenotypes in twi1-1 and mot48-2; twi1-1 were indeed derived from loss of the TWI1 protein.

Exogeneous TWI1 protein can rescue the Twi1 phenotype.
Fig. 5. Exogeneous TWI1 protein can rescue the Twi1 phenotype.
A) Immunoblot of whole cell samples from wild-type (CC-125 and cw15), twi1-1, mot48-2; twi1-1, twi1-1; TWI1::HA and mot48-2; twi1-1; TWI1::HA strains using anti-HA (left)/MOT48 and TWI1 (right) antibodies. The black arrowheads indicate the wild-type endogenous TWI1 protein. Red arrowheads indicate the exogeneous TWI1 protein with the 3HA tag. The cDNA-driven exogeneous TWI1::3HA proteins show two bands in these blots (red arrowheads). A green arrowhead indicates the MOT48 protein. B) Urea-PAGE of axonemes from wild-type (CC-125), twi1-1, twi1-1; TWI1::HA, mot48-2, mot48-2; twi1-1, and mot48-2; twi1-1; TWI1::HA strains. For presentation, gel regions of ciliary dyneins and lower parts are shown. The relative positions of ciliary dyneins were adjusted between all strains for comparison. The black arrowheads indicate the HCs of ODA. The red arrowheads indicate the HCs of IDAs. HCs of ODAγ and IDAs a, d, e and fβ form a large band in the urea gel. A green arrowhead indicates HC degradation products. In the mot48-2; twi1-1 strain, the ODAα and IDA bands were missing (asterisks), but these dyneins were recovered in the mot48-2; twi1-1; TWI1::HA strain. The correspondence between bands in the Urea-PAGE gel and DHCs was based on [6567]. C) Immunoblots of axonemal samples from wild-type (CC-125), mot48-2, mot48-2; twi1-1 and mot48-2; twi1-1; TWI1::HA strains using dynein-subunit antibodies (anti-p28/IDA4, p38, actin/IDA5, centrin/VFL2, and IC2/IC69/ODA6; S1 Table).

TWI1 may work together with other preassembly factors

In addition to the PIH mutants, we also performed spectral counting of dyneins in isolated axonemes from pf23 (5–4) and pf22A (CC-2493)(which contain a wild-type TWI1 gene) and compared to pf23 (CC-1383) and pf22 (CC-1382)(which contain a mutation in the TWI1 gene, described above). The dynein defects in pf23 (CC-1383) are more profound than pf23 (5–4)(S3A Fig). Particularly, the defects in IDA d (DHC2) and IDA g (DHC7) are larger in pf23 (CC-1383) than in pf23 (5–4) [28]. On the other hand, the dynein defects in pf22A (CC-2493) and pf22 (CC-1382) are relatively similar to each other (S3A Fig). In addition to ODAs and IDAs b (DHC5) and c (DHC9) as previously described [16, 32], these pf22 mutants have large defects (< 50% of wild-type) in IDAs a (DHC6) and e (DHC8) and minor dyneins DHC3 and DHC4. Also, the axonemal amount of one minor dynein DHC12 is increased in the pf22 mutants (S3A and S3B Fig). Given that DNAAF4/DYX1C1 (PF23 orthologue)(S1 Table) and DNAAF6/PIH1D3 (TWI1 orthologue)(Table 1) in mammals are predicted to form a complex and work together in dynein preassembly [18], the large dynein defects observed in pf23 (CC-1383; with twi1-2 background) may indicate that Chlamydomonas TWI1 is needed for efficient function of the PF23 protein in dynein preassembly. Additionally, the swimming phenotype of the pf23 (CC-1383) strain, rescued with the wild-type PF23 gene, which also harbored the twi1-2 mutation, was indistinguishable from wild-type [28], consistent with the subtle swimming defect observed in the twi1-1 mutant.

Discussion

In this report, we characterized two novel PIH preassembly mutants in Chlamydomonas reinhardtii, mot48-2 and twi1-1. Although recent studies reveal a conserved role(s) of PIH proteins in ciliary dynein preassembly [13, 1719, 21, 22], the specificity of their molecular function(s) and interaction(s) is not fully understood. Our study of assembly of specific dyneins in the axoneme, in the single and double PIH mutants, revealed partially overlapping and specific roles for three Chlamydomonas PIH proteins, MOT48, TWI1, and PF13. Accordingly, we have updated our previous model [13] of the preassembly pathway involving PIH proteins and ciliary dynein species (Fig 6). The preassembly pathway of the PIH proteins is more complicated than previously predicted [13], with assembly of each ciliary dynein requiring a specific complement of PIH proteins (Fig 6). We observed assembly defects in some dynein species are more severe in the double preassembly mutants (e.g. mot48-2; twi1-1, pf13; twi1-1 and pf23; twi1-2 (CC-1383)) than a single mutant (e.g. mot48-2, pf13 and pf23). In addition, dynein f/I1, and possibly IDA a, do not require PIH proteins for assembly (Fig 6 and see [22]). We discuss possibilities in context to the severe phenotypes in the double PIH mutants.

Updated model of the PIH-protein pathways in the dynein preassembly.
Fig. 6. Updated model of the PIH-protein pathways in the dynein preassembly.
A proposed model of the functional pathways of PIH preassembly proteins in Chlamydomonas adapted from [13]. The bold lines indicate the primary pathways, showing the dynein species that were reduced more than 30% in spectral numbers in each PIH mutant compared to the wild-type. The dotted thin lines indicate relatively secondary pathways, showing the dynein species that were reduced more than 30% in spectral numbers in the double PIH-mutants compared to the single PIH-mutants. The pathways are more complicated than previously thought [13]. Since the three-headed ODA of Chlamydomonas cannot be assembled in axonemes in the absence of ODA β or γ HC [68, 69], in this study we could not assess the direct contribution of each PIH protein in the cytoplasmic preassembly of each ODA HC since the amount of ODA α in cilia could be reduced simply in the absence of ODA β or γ HC. Thus, we categorized ODA HCs into one group in this model (asterisk). This is also the case for the two-headed IDA f/I1, in which this species cannot be assembled without each HC (f/I1α or f/I1β)[70, 71], thus we also categorized IDA f/I1 HCs into one group (asterisk). In this study, we could not find any primary pathway of the PIH proteins for the preassembly of IDAs a and f/I1. These dynein species may be preassembled by factors other than PIH proteins (e.g. PF22 or PF23, see S3 Fig), or can assemble spontaneously in cytoplasm to some extent without the help of preassembly factors.

Preassembly of ciliary dyneins likely requires a series of ordered steps each requiring a PIH preassembly protein. For example, full assembly of dynein species IDAs b and c in the axoneme require the activity of at least two PIH proteins including PF13, MOT48 and possibly TWI1 (Figs 4 and 6). Sequential steps in the dynein preassembly have been predicted in previous studies [3840]. PIH proteins and other preassembly factors may be organized in complexes/organelles such as the DynAP [23] or organized in a series of individual complexes operating in ordered steps (Table 2). Thus, whether in a single complex or in a series of complexes, each PIH protein may operate in a different step in preassembly, and loss of PIH proteins would attenuate the whole preassembly process.

Tab. 2. Interacting partners of PIH proteins predicted/identified in previous studies.
Interacting partners of PIH proteins predicted/identified in previous studies.

The apparent functional overlap between PIH proteins required for preassembly of certain dyneins, such as IDA c, suggests that the PIH proteins can work with partially overlapping function. Thus, loss of two PIH proteins would cause a more severe phenotype than a single mutation. Consistent with this idea, missing PIH proteins appear to be partially compensated by the other PIH proteins to some extent. For example, the apparent subtle motility defect in twi1-1 likely derives from a compensatory function of MOT48 and PF13 in the cytoplasm of twi1-1 cells (see IDA c in Fig 4). This hypothesis is also consistent with our observation that the motility of the mot48-1/mot48-2 mutant improves as the cells in liquid culture grow old, implying that in the mot48 cells, PF13 and TWI1 eventually compensate and help to preassemble ciliary dyneins that are usually dependent on MOT48. This idea could explain the occasional ODA assembly in pf13 axonemes observed by Huang et al., [32]. Predictably, MOT48 and/or TWI1 partially compensate, with time, for PF13 in the pf13 mutant.

As mentioned above, preassembly factors including PIH proteins, PF22 and PF23 may form in the molecular complex/organelle DynAPs [23]. Based on our data, loss of one subunit protein from this complex may have a modest effect on the activity of the whole complex, but loss of two or more specific subunits largely blocks the activity of the complex for dynein pre-assembly. Interestingly, in contrast to IDAs b and c, assembly of the minor dyneins, particularly DHC12, only seems to require MOT48. Although we focused on the assembly of dynein HCs in the axoneme, we also observed either an increase or decrease in specific LCs in the cytoplasmic compartment. For example, DHC9 (IDA c HC) and p28/IDA4, a LC of several single-headed IDAs, are reduced in the cytoplasm of PIH mutants with the mot48 background (Fig 3E, see also [13]). In addition, actin/IDA5, another LC of single-headed dyneins, accumulates in the cytoplasm of PIH mutants with the pf13 and/or mot48-2 background (Fig 3E). Thus, stability of dynein subunits in the cytoplasm may offer another approach toward understanding PIH protein function. One model is that the PIH proteins play roles in the folding and stability of dynein HCs, and also in the following LC assembly to HC [1719, 21, 40]. Further biochemical analyses, in combination with the in vitro reconstitution, are required to define the details of PIH protein function.

Although Chlamydomonas MOT48 was first identified as a protein that is conserved in organisms with motile cilia [44], an exact orthologous MOT48 gene in vertebrates remains unclear. A recent study showed that among the four PIH proteins found in vertebrates, PIH1D1, PIH1D2, DNAAF6/PIH1D3 (TWI1 orthologue), and DNAAF2/KTU (PF13 orthologue), MOT48 groups near DNAAF2/KTU and PIH1D1 in a phylogenetic tree [22]. We also generated a phylogenetic tree using the full-length sequences of PIH proteins (S4 Fig), and also found that MOT48 fell into a group with PIH1D1 proteins. A BLAST search against the NCBI database (https://blast.ncbi.nlm.nih.gov/Blast.cgi) also revealed that among the four PIH proteins in vertebrates, PIH1D1 showed the highest similarity to MOT48, although the E-values were relatively low (< 7E-15). In addition, recently MOT48 has been reported to interact with RPAP3 and RuvBL1 [25], components of the known R2TP chaperone complex [41] to form a potential R2TP-like complex in Chlamydomonas cytoplasm (Table 2). Thus, MOT48 may have a function as a co-factor in the Chlamydomonas R2TP-like complex, which is similar to the PIH1D1 function in higher eukaryotes [41].

The TWI1 orthologue, DNAAF6/PIH1D3, has also been postulated to interact with DNAAF4/DYX1C1 [18], orthologous to the PF23 protein in Chlamydomonas [28]. Thus, TWI1 may function as part of a large chaperone complex [18, 19, 23](Table 2). Using the 3HA-tagged rescued strains (mot48-2; MOT48::HA and twi1-1; TWI1::HA) for identification of interacting partners, we failed to identify chaperone related proteins. This negative result could indicate weak and/or transient interactions of the PIH proteins with chaperones and/or interacting partners of MOT48 and TWI1. Further structural and biochemical studies of PIH interacting proteins, including specific dynein HC/IC/LCs, are required for understanding assembly complexes, steps and specificity of each PIH protein required for ciliary dynein assembly.

Summary

The mechanisms for PIH proteins in assembly of ciliary dyneins are more complicated than previously thought. Chlamydomonas uses the three PIH proteins, MOT48, TWI1, and PF13, for ciliary dynein preassembly. Based on analysis in single and double PIH mutants, specific PIH proteins are required for assembly of specific dyneins, in some cases, and in other cases the PIH proteins can work in partially overlapping fashion. Further biochemical studies, and the two novel Chlamydomonas PIH preassembly mutants, mot48-2 and twi1-1, from this study, will define our understanding of dynein assembly.

Materials and methods

Chlamydomonas strains and samples

The Chlamydomonas strains used in this study are listed in S2 Table. The novel mot48 (ida10) allele, referred to as mot48-2 was isolated from a CLiP library strain (LMJ.RY0402.055540)[24] having a mutation that was unlinked to the paromomycin resistant (APHVIII) insertional cassette used for selection but caused a slow-swimming phenotype. The original LMJ.RY0402.055540 strain was backcrossed with the wild-type (CC-125) strain to separate this mutation, and two mot48-2 progeny were isolated. The identification of the mot48 background was confirmed by Sanger sequencing using the primer pair: Ida10-2 GF2 (5’-TGGCAGCACATTCATAAGCA-3’) and Ida10-2 GR2 (5’-CGCTGTACTAGAGCCCCTCA-3’). The twi1 strain was first obtained from the CLiP library (LMJ.RY0402.076787)[24] and backcrossed with the wild-type strain (CC-125), and the twi1 mutant progeny (twi1-1) were used for experiments. Cells were grown in the tris-acetic acid-phosphate (TAP) liquid/solid media as previously described [45]. Double Chlamydomonas PIH mutants were obtained using the standard tetrad procedure [45]. The deciliation was performed following the standard procedure [46]. For preparation of whole cell samples or de-ciliated cell-body samples, whole cells/cell bodies were extracted with water/methanol/chloroform (volume ratio = 3:4:1) to remove the nucleic acids, lipids and chlorophyll, and the denatured proteins were boiled in the SDS-sample buffer as previously described [47].

Rescue of mot48-2, twi1-1, and mot48-2; twi1-1

Phenotypic rescue of the mot48-2, twi1-1, and mot48-2; twi1-1 strains was performed by the electroporation method using each wild-type gene cloned in the modified pGend vector (pGend-MCS-3HA-AphVIII/pGend-MCS-mCherry-3HA-AphVIII/pGend-MCS-3HA-Hyg)[13, 48, 49]. The rescued strain, mot48-2; MOT48::HA expressed exogeneous MOT48 with a 3HA tag at the C-terminus. The rescued strain, mot48-2; MOT48::mCherry-HA expressed exogeneous MOT48 with the mCherry-3HA tag at the C-terminus. The pGend-MOT48-3HA-AphVIII vector was previously described [13] and also used in this study. The two twi1-1 rescued strains, twi1-1; TWI1::HA and mot48-2; twi1-1; TWI1::HA expressed exogeneous TWI1 with a 3HA tag at the C-terminus. The primer pair used for wild-type TWI1 cloning was as follows, TWI1-pGend-F1 (5’-CACAACAAGCCCATATGGACATTGGGAGCTTCACTGCTGA-3’) and TWI1-pGend-R1 (5’-GGTATCGATCGAATTCGAATGGCTCTTCCCGAATGATGCG-3’). The NdeI/EcoRI sites used for cloning are underlined.

Spectral counting analysis

A semi-quantitative estimation of the amount of dyneins in the isolated axonemes [28, 50, 51] was conducted using the spectral counting analyses on some LC-MS/MS spectrometers at University of Massachusetts Medical School Mass Spectrometry Facility.

A first set of experiments was performed on dyneins from the wild-type (CC-124, cw15), mot48-1, and mot48-2 strains with the aim of comparing the dynein levels between the mot48-1 and mot48-2 strains. For normalization and comparison between samples, the peptide numbers of ciliary dyneins observed in the CC-124 strain were assigned a ratio of 1.0, and the observed peptide numbers of Hydin [52], a central-pair protein were also used as an internal standard. The first-set of experiments was generously performed by Dr. John Leszyk (University of Massachusetts Medical School). Thresholds in the Scaffold 4 software (http://www.proteomesoftware.com/products/scaffold/) for the first-set of analyses were set as follows: Protein Threshold: 90%/Minimal Peptides Number: 2/ Peptide Threshold: 70%. The averages of two independent experiments are summarized in Fig 2.

A second set of experiments was performed on dyneins from wild-type (CC-125), twi1-1, mot48-2, mot48-2; twi1-1, pf13, pf13; twi1-1, and pf23 (5–4) strains with the aim of comparing the dynein levels between the single and double PIH preassembly mutants. For normalization and comparison between samples, observed peptide numbers in the CC-125 strain and also the peptide numbers of Hydin were used. The second set of experiments was generously performed by Drs. Scott Shaffer and Xuni Li (University of Massachusetts Medical School). Thresholds in the Scaffold 4 software for the second-set analyses were set as follows: Protein Threshold: 99%/Minimal Peptides Number: 2/ Peptide Threshold: 70%. The results are summarized in Fig 4 and S3 Fig.

A third set of experiments was performed on dyneins from wild-type (CC-125), twi1-1; TWI1::HA, pf22 (CC-1382 with the twi1-2 background) and pf22A (CC-2493) strains with the aim to check the dynein rescue and effect of the twi1-2 mutation in dynein assembly in pf22 strains. Observed peptide numbers were normalized using peptide numbers of CC-125 and Hydin, and the results were combined/incorporated into Fig 4 and S3 Fig. The third set of experiments was generously performed by Drs. Scott Shaffer and Roshanak Aslebagh (University of Massachusetts Medical School). Thresholds in the Scaffold 4 software for the third-set analyses were set as follows: Protein Threshold: 99%/Minimal Peptides Number: 2/ Peptide Threshold: 77%. The spectral counting data of pf23 (CC-1383 with the twi1-2 background) normalized with the wild-type (137c) peptide counts were reanalyzed/refined from our previous paper [28] with the Hydin normalization in S3 Fig.

TWI1 antibody production

The TWI1 cDNA sequence was determined using the Chlamydomonas cDNA library (Chlamydomonas Resource Center) and cloned into the NdeI/BamHI site of the pET15b vector (Novagen) by the In-Fusion HD Cloning enzyme (TAKARA). The primer pair to amplify the TWI1 cDNA sequence was as follows: TWI1-CF1 (5’-CGCGCGGCAGCCATATGGACATTGGGAGCTTCACTG-3’) and TWI1-CR1 (5’-GTTAGCAGCCGGATCCGAATGGCTCTTCCCGAATGATGCG-3’)(The NdeI/BamHI sites are underlined). The purified TWI1 protein with a 6His tag at the N-terminus was used as antigen to immunize two rabbits. The antisera from rabbits were blot and Protein-A purified before use, as previously described [53, 54]. The Chlamydomonas TWI1 cDNA sequence determined in this study was deposited in the DNA Data Bank of Japan (DDBJ) under the accession No. LC461993.

Other methods

SDS-PAGE and immunoblotting were performed following standard procedures [55, 56]. For the immunoblotting, antibodies used included: primary antibodies (anti-MOT48 [28], anti-HA (Y-11)(Santa Cruz), anti-HA (3F10)-HRP (Roche), anti-PF23 [28], anti-TWI1 (this study), anti-actin/IDA5 [57], anti-p28/IDA4 [58], anti-centrin/VFL2 (20H5)(MilliporeSigma), anti-p38 [59], anti-IC138/BOP5 [60], anti-IC2/IC69/ODA6 [61], anti-DHC9 (IDA c HC)[37]); secondary antibody (Goat-anti-Rabbit or Mouse-HRP (Roche)). Immunofluorescent microscopic observation of nucleo-flagellar apparatuses was performed as described previously [36, 37, 62], and the acquired images were adjusted for presentation using Photoshop (Adobe). The urea PAGE used to resolve ciliary dynein bands was performed as previously described [63]. The swimming velocity of Chlamydomonas was assessed on free-swimming cells in liquid culture using our in-lab video system and ImageJ software (https://www.google.com/search?client=firefox-b-d&q=imagej+software) [64]. Velocities were measured on the 3-day liquid cultured cells, and reported in the text as average ± standard deviation. The ciliated cell ratio in the pf13 and pf13; twi1-1 strains was counted and averaged on three days. Student’s t-test was performed on Excel (Microsoft).

Supporting information

S1 Fig [tif]
Swimming velocity measurement of -related strains.

S2 Fig [tif]
Immunofluorescent microscopic observation of ciliary dyneins in axonemes from PIH mutants.

S3 Fig [tif]
Dynein defects are more severe in the mutant also containing the mutation.

S4 Fig [tif]
Phylogenetic analysis of PIH proteins from and other organisms.

S1 Table [pdf]
dynein subunits, IFT proteins, and non-PIH preassembly proteins mentioned in this study and their potential human orthologues.

S2 Table [pdf]
strains used in this study.


Zdroje

1. Ishikawa T. Axoneme Structure from Motile Cilia. Cold Spring Harb Perspect Biol. 2017;9(1):a028076. doi: 10.1101/cshperspect.a028076 27601632

2. Satir P. CILIA: before and after. Cilia. 2017;6:1. doi: 10.1186/s13630-017-0046-8 28293419

3. Zariwala MA, Omran H, Ferkol TW. The emerging genetics of primary ciliary dyskinesia. Proc Am Thorac Soc. 2011;8(5):430–3. doi: 10.1513/pats.201103-023SD 21926394

4. Chodhari R, Mitchison HM, Meeks M. Cilia, primary ciliary dyskinesia and molecular genetics. Paediatr Respir Rev. 2004;5(1):69–76. doi: 10.1016/j.prrv.2003.09.005 15222957

5. Rubbo B, Lucas JS. Clinical care for primary ciliary dyskinesia: current challenges and future directions. Eur Respir Rev. 2017;26(145):170023. doi: 10.1183/16000617.0023-2017 28877972

6. Horani A, Ferkol TW. Advances in the Genetics of Primary Ciliary Dyskinesia: Clinical Implications. Chest. 2018;154(3):645–52. doi: 10.1016/j.chest.2018.05.007 29800551

7. Viswanadha R, Sale WS, Porter ME. Ciliary Motility: Regulation of Axonemal Dynein Motors. Cold Spring Harb Perspect Biol. 2017;9(8):a018325. doi: 10.1101/cshperspect.a018325 28765157

8. Kamiya R, Yagi T. Functional diversity of axonemal dyneins as assessed by in vitro and in vivo motility assays of Chlamydomonas mutants. Zoolog Sci. 2014;31(10):633–44. doi: 10.2108/zs140066 25284382

9. King SM. Axonemal Dynein Arms. Cold Spring Harb Perspect Biol. 2016;8(11):a028100. doi: 10.1101/cshperspect.a028100 27527589

10. Brokaw CJ, Kamiya R. Bending patterns of Chlamydomonas flagella: IV. Mutants with defects in inner and outer dynein arms indicate differences in dynein arm function. Cell Motil Cytoskeleton. 1987;8(1):68–75. doi: 10.1002/cm.970080110 2958145

11. Fowkes ME, Mitchell DR. The role of preassembled cytoplasmic complexes in assembly of flagellar dynein subunits. Mol Biol Cell. 1998;9(9):2337–47. doi: 10.1091/mbc.9.9.2337 9725897

12. Viswanadha R, Hunter EL, Yamamoto R, Wirschell M, Alford LM, Dutcher SK, et al. The ciliary inner dynein arm, I1 dynein, is assembled in the cytoplasm and transported by IFT before axonemal docking. Cytoskeleton (Hoboken). 2014;71(10):573–86. doi: 10.1002/cm.21192 25252184

13. Yamamoto R, Hirono M, Kamiya R. Discrete PIH proteins function in the cytoplasmic preassembly of different subsets of axonemal dyneins. J Cell Biol. 2010;190(1):65–71. doi: 10.1083/jcb.201002081 20603327

14. Kobayashi D, Takeda H. Ciliary motility: the components and cytoplasmic preassembly mechanisms of the axonemal dyneins. Differentiation. 2012;83(2):S23–9. doi: 10.1016/j.diff.2011.11.009 22154137

15. Desai PB, Dean AB, Mitchell DR. Cytoplasmic preassembly and trafficking of axonemal dyneins: Dyneins (The Biology of Dynein Motors). 2nd Edition ed: Academic Press; 2017. 684 p.

16. Mitchison HM, Schmidts M, Loges NT, Freshour J, Dritsoula A, Hirst RA, et al. Mutations in axonemal dynein assembly factor DNAAF3 cause primary ciliary dyskinesia. Nat Genet. 2012;44(4):381–9, s1-2. doi: 10.1038/ng.1106 22387996

17. Omran H, Kobayashi D, Olbrich H, Tsukahara T, Loges NT, Hagiwara H, et al. Ktu/PF13 is required for cytoplasmic pre-assembly of axonemal dyneins. Nature. 2008;456(7222):611–16. doi: 10.1038/nature07471 19052621

18. Olcese C, Patel MP, Shoemark A, Kiviluoto S, Legendre M, Williams HJ, et al. X-linked primary ciliary dyskinesia due to mutations in the cytoplasmic axonemal dynein assembly factor PIH1D3. Nat Commun. 2017;8:14279. doi: 10.1038/ncomms14279 28176794

19. Paff T, Loges NT, Aprea I, Wu K, Bakey Z, Haarman EG, et al. Mutations in PIH1D3 cause X-linked primary ciliary dyskinesia with outer and inner dynein arm defects. Am J Hum Genet. 2017;100(1):160–8. doi: 10.1016/j.ajhg.2016.11.019 28041644

20. Tarkar A, Loges NT, Slagle CE, Francis R, Dougherty GW, Tamayo JV, et al. DYX1C1 is required for axonemal dynein assembly and ciliary motility. Nat Genet. 2013;45(9):995–1003. doi: 10.1038/ng.2707 23872636

21. Dong F, Shinohara K, Botilde Y, Nabeshima R, Asai Y, Fukumoto A, et al. Pih1d3 is required for cytoplasmic preassembly of axonemal dynein in mouse sperm. J Cell Biol. 2014;204(2):203–13. doi: 10.1083/jcb.201304076 24421334

22. Yamaguchi H, Oda T, Kikkawa M, Takeda H. Systematic studies of all PIH proteins in zebrafish reveal their distinct roles in axonemal dynein assembly. Elife. 2018;7:e36979. doi: 10.7554/eLife.36979 29741156

23. Huizar RL, Lee C, Boulgakov AA, Horani A, Tu F, Marcotte EM, et al. A liquid-like organelle at the root of motile ciliopathy. Elife. 2018;7:e38497. doi: 10.7554/eLife.38497 30561330

24. Li X, Zhang R, Patena W, Gang SS, Blum SR, Ivanova N, et al. An indexed, mapped mutant library enables reverse genetics studies of biological processes in Chlamydomonas reinhardtii. Plant Cell. 2016;28(2):367–87. doi: 10.1105/tpc.15.00465 26764374

25. Liu G, Wang L, Pan J. Chlamydomonas WDR92 in association with R2TP-like complex and multiple DNAAFs to regulate ciliary dynein preassembly. J Mol Cell Biol. 2019;11(9):770–80. doi: 10.1093/jmcb/mjy067 30428028

26. Hom EF, Witman GB, Harris EH, Dutcher SK, Kamiya R, Mitchell DR, et al. A unified taxonomy for ciliary dyneins. Cytoskeleton (Hoboken). 2011;68(10):555–65. doi: 10.1002/cm.20533 21953912

27. Fabczak H, Osinka A. Role of the novel Hsp90 co-chaperones in dynein arms' preassembly. Int J Mol Sci. 2019;20(24):6174. doi: 10.3390/ijms20246174 31817850

28. Yamamoto R, Obbineni JM, Alford LM, Ide T, Owa M, Hwang J, et al. Chlamydomonas DYX1C1/PF23 is essential for axonemal assembly and proper morphology of inner dynein arms. PLoS Genet. 2017;13(9):e1006996. doi: 10.1371/journal.pgen.1006996 28892495

29. Zhao R, Kakihara Y, Gribun A, Huen J, Yang G, Khanna M, et al. Molecular chaperone Hsp90 stabilizes Pih1/Nop17 to maintain R2TP complex activity that regulates snoRNA accumulation. J Cell Biol. 2008;180(3):563–78. doi: 10.1083/jcb.200709061 18268103

30. Pal M, Morgan M, Phelps SEL, Roe SM, Parry-Morris S, Downs JA, et al. Structural basis for phosphorylation-dependent recruitment of Tel2 to Hsp90 by Pih1. Structure (London, England: 1993). 2014;22(6):805–18. doi: 10.1016/j.str.2014.04.001 24794838

31. King SM. 7—Composition and Assembly of Axonemal Dyneins. In: King SM, editor. Dyneins. Boston: Academic Press; 2012. p. 208–43.

32. Huang B, Piperno G, Luck DJ. Paralyzed flagella mutants of Chlamydomonas reinhardtii. Defective for axonemal doublet microtubule arms. J Biol Chem. 1979;254(8):3091–9. 429335

33. Stolc V, Samanta MP, Tongprasit W, Marshall WF. Genome-wide transcriptional analysis of flagellar regeneration in Chlamydomonas reinhardtii identifies orthologs of ciliary disease genes. Proc Natl Acad Sci U S A. 2005;102(10):3703–7. doi: 10.1073/pnas.0408358102 15738400

34. King SM, Patel-King RS. The oligomeric outer dynein arm assembly factor CCDC103 is tightly integrated within the ciliary axoneme and exhibits periodic binding to microtubules. J Biol Chem. 2015;290(12):7388–401. doi: 10.1074/jbc.M114.616425 25572396

35. Kim KS, Kustu S, Inwood W. Natural history of transposition in the green alga Chlamydomonas reinhardtii: use of the AMT4 locus as an experimental system. Genetics. 2006;173(4):2005–19. doi: 10.1534/genetics.106.058263 16702425

36. Bui KH, Yagi T, Yamamoto R, Kamiya R, Ishikawa T. Polarity and asymmetry in the arrangement of dynein and related structures in the Chlamydomonas axoneme. J Cell Biol. 2012;198(5):913–25. doi: 10.1083/jcb.201201120 22945936

37. Yagi T, Uematsu K, Liu Z, Kamiya R. Identification of dyneins that localize exclusively to the proximal portion of Chlamydomonas flagella. J Cell Sci. 2009;122(9):1306–14. doi: 10.1242/jcs.045096 19351714

38. Horani A, Ustione A, Huang T, Firth AL, Pan J, Gunsten SP, et al. Establishment of the early cilia preassembly protein complex during motile ciliogenesis. Proc Natl Acad Sci U S A. 2018;115(6):E1221–28. doi: 10.1073/pnas.1715915115 29358401

39. Mali GR, Yeyati PL, Mizuno S, Dodd DO, Tennant PA, Keighren MA, et al. ZMYND10 functions in a chaperone relay during axonemal dynein assembly. Elife. 2018;7:e34389. doi: 10.7554/eLife.34389 29916806

40. Zur Lage P, Stefanopoulou P, Styczynska-Soczka K, Quinn N, Mali G, von Kriegsheim A, et al. Ciliary dynein motor preassembly is regulated by Wdr92 in association with HSP90 co-chaperone, R2TP. J Cell Biol. 2018;217(7):2583–98. doi: 10.1083/jcb.201709026 29743191

41. Kakihara Y, Houry WA. The R2TP complex: discovery and functions. Biochim Biophys Acta. 2012;1823(1):101–7. doi: 10.1016/j.bbamcr.2011.08.016 21925213

42. Maurizy C, Quinternet M, Abel Y, Verheggen C, Santo PE, Bourguet M, et al. The RPAP3-Cterminal domain identifies R2TP-like quaternary chaperones. Nat Commun. 2018;9(1):2093. doi: 10.1038/s41467-018-04431-1 29844425

43. Patel-King RS, Sakato-Antoku M, Yankova M, King SM. WDR92 is required for axonemal dynein heavy chain stability in cytoplasm. Mol Biol Cell. 2019;30(15):1834–45. doi: 10.1091/mbc.E19-03-0139 31116681

44. Merchant SS, Prochnik SE, Vallon O, Harris EH, Karpowicz SJ, Witman GB, et al. The Chlamydomonas genome reveals the evolution of key animal and plant functions. Science. 2007;318(5848):245–50. doi: 10.1126/science.1143609 17932292

45. Harris EH. The Chlamydomonas sourcebook: a comprehensive guide to biology and laboratory use: Academic Press, San Diego, 780pp; 1989.

46. Craige B, Brown JM, Witman GB. Isolation of Chlamydomonas flagella. Curr Protoc Cell Biol. 2013;Chapter 3:Unit 3.41.1–9. doi: 10.1002/0471143030.cb0341s59 23728744

47. Bölling C, Fiehn O. Metabolite profiling of Chlamydomonas reinhardtii under nutrient deprivation. Plant Physiol. 2005;139(4):1995–2005. doi: 10.1104/pp.105.071589 16306140

48. Shimogawara K, Fujiwara S, Grossman A, Usuda H. High-efficiency transformation of Chlamydomonas reinhardtii by electroporation. Genetics. 1998;148(4):1821–8. 9560396

49. Fischer N, Rochaix JD. The flanking regions of PsaD drive efficient gene expression in the nucleus of the green alga Chlamydomonas reinhardtii. Mol Genet Genomics. 2001;265(5):888–94. doi: 10.1007/s004380100485 11523806

50. Wirschell M, Olbrich H, Werner C, Tritschler D, Bower R, Sale WS, et al. The nexin-dynein regulatory complex subunit DRC1 is essential for motile cilia function in algae and humans. Nat Genet. 2013;45(3):262–8. doi: 10.1038/ng.2533 23354437

51. Lin J, Le TV, Augspurger K, Tritschler D, Bower R, Fu G, et al. FAP57/WDR65 targets assembly of a subset of inner arm dyneins and connects to regulatory hubs in cilia. Mol Biol Cell. 2019;30(21):2659–80. doi: 10.1091/mbc.E19-07-0367 31483737

52. Lechtreck KF, Witman GB. Chlamydomonas reinhardtii hydin is a central pair protein required for flagellar motility. J Cell Biol. 2007;176(4):473–82. doi: 10.1083/jcb.200611115 17296796

53. Tang W-JY. Chapter 5 Blot-Affinity Purification of Antibodies. In: Asai DJ, editor. Methods Cell Biol. 37: Academic Press; 1993. p. 95–104. doi: 10.1016/s0091-679x(08)60245-9 8255253

54. Grodzki AC, Berenstein E. Antibody purification: affinity chromatography—protein A and protein G Sepharose. Methods Mol Biol. 2010;588:33–41. doi: 10.1007/978-1-59745-324-0_5 20012816

55. Laemmli UK. Cleavage of structural proteins during the assembly of the head of bacteriophage T4. Nature. 1970;227(5259):680–5. doi: 10.1038/227680a0 5432063

56. Kurien BT, Scofield RH. Western blotting. Methods. 2006;38(4):283–93. doi: 10.1016/j.ymeth.2005.11.007 16483794

57. Kato-Minoura T, Hirono M, Kamiya R. Chlamydomonas inner-arm dynein mutant, ida5, has a mutation in an actin-encoding gene. J Cell Biol. 1997;137(3):649–56. doi: 10.1083/jcb.137.3.649 9151671

58. LeDizet M, Piperno G. The light chain p28 associates with a subset of inner dynein arm heavy chains in Chlamydomonas axonemes. Mol Biol Cell. 1995;6(6):697–711. doi: 10.1091/mbc.6.6.697 7579689

59. Yamamoto R, Yanagisawa HA, Yagi T, Kamiya R. A novel subunit of axonemal dynein conserved among lower and higher eukaryotes. FEBS Lett. 2006;580(27):6357–60. doi: 10.1016/j.febslet.2006.10.047 17094970

60. Hendrickson TW, Perrone CA, Griffin P, Wuichet K, Mueller J, Yang P, et al. IC138 is a WD-repeat dynein intermediate chain required for light chain assembly and regulation of flagellar bending. Mol Biol Cell. 2004;15(12):5431–42. doi: 10.1091/mbc.e04-08-0694 15469982

61. King SM, Otter T, Witman GB. Characterization of monoclonal antibodies against Chlamydomonas flagellar dyneins by high-resolution protein blotting. Proc Natl Acad Sci U S A. 1985;82(14):4717–21. doi: 10.1073/pnas.82.14.4717 3161075

62. Sanders MA, Salisbury JL. Immunofluorescence microscopy of cilia and flagella. Methods Cell Biol. 1995;47:163–9. doi: 10.1016/s0091-679x(08)60805-5 7476482

63. Jarvik JW, Rosenbaum JL. Oversized flagellar membrane protein in paralyzed mutants of Chlamydomonas reinhardrii. J Cell Biol. 1980;85(2):258–72. doi: 10.1083/jcb.85.2.258 7372708

64. Schneider CA, Rasband WS, Eliceiri KW. NIH Image to ImageJ: 25 years of image analysis. Nat Methods. 2012;9(7):671–5. doi: 10.1038/nmeth.2089 22930834

65. Yamamoto R, Yagi T, Kamiya R. Functional binding of inner-arm dyneins with demembranated flagella of Chlamydomonas mutants. Cell Motil Cytoskeleton. 2006;63(5):258–65. doi: 10.1002/cm.20121 16518818

66. Piperno G. Regulation of dynein activity within Chlamydomonas flagella. Cell Motil Cytoskeleton. 1995;32(2):103–5. doi: 10.1002/cm.970320206 8681388

67. Kagami O, Kamiya R. Translocation and rotation of microtubules caused by multiple species of Chlamydomonas inner-arm dynein. J Cell Sci. 1992;103(3):653–64.

68. Sakakibara H, Takada S, King SM, Witman GB, Kamiya R. A Chlamydomonas outer arm dynein mutant with a truncated beta heavy chain. J Cell Biol. 1993;122(3):653–61. doi: 10.1083/jcb.122.3.653 8335691

69. Liu Z, Takazaki H, Nakazawa Y, Sakato M, Yagi T, Yasunaga T, et al. Partially functional outer-arm dynein in a novel Chlamydomonas mutant expressing a truncated gamma heavy chain. Eukaryot Cell. 2008;7(7):1136–45. doi: 10.1128/EC.00102-08 18487347

70. Myster SH, Knott JA, O'Toole E, Porter ME. The Chlamydomonas Dhc1 gene encodes a dynein heavy chain subunit required for assembly of the I1 inner arm complex. Mol Biol Cell. 1997;8(4):607–20. doi: 10.1091/mbc.8.4.607 9247642

71. Perrone CA, Myster SH, Bower R, O'Toole ET, Porter ME. Insights into the structural organization of the I1 inner arm dynein from a domain analysis of the 1beta dynein heavy chain. Mol Biol Cell. 2000;11(7):2297–313. doi: 10.1091/mbc.11.7.2297 10888669

72. Saitou N, Nei M. The neighbor-joining method: a new method for reconstructing phylogenetic trees. Mol Biol Evol. 1987;4(4):406–25. doi: 10.1093/oxfordjournals.molbev.a040454 3447015

73. Felsenstein J. Confidence limits on phylogenies: an approach using the bootstrap. Evolution. 1985;39(4):783–91. doi: 10.1111/j.1558-5646.1985.tb00420.x 28561359

74. Nei M, Kumar S. Molecular Evolution and Phylogenetics: Oxford University Press, New York; 2000.


Článek vyšel v časopise

PLOS Genetics


2020 Číslo 11
Nejčtenější tento týden
Nejčtenější v tomto čísle
Kurzy

Zvyšte si kvalifikaci online z pohodlí domova

Hypertenze a hypercholesterolémie – synergický efekt léčby
nový kurz
Autoři: prof. MUDr. Hana Rosolová, DrSc.

Multidisciplinární zkušenosti u pacientů s diabetem
Autoři: Prof. MUDr. Martin Haluzík, DrSc., prof. MUDr. Vojtěch Melenovský, CSc., prof. MUDr. Vladimír Tesař, DrSc.

Úloha kombinovaných preparátů v léčbě arteriální hypertenze
Autoři: prof. MUDr. Martin Haluzík, DrSc.

Halitóza
Autoři: MUDr. Ladislav Korábek, CSc., MBA

Terapie roztroušené sklerózy v kostce
Autoři: MUDr. Dominika Šťastná, Ph.D.

Všechny kurzy
Přihlášení
Zapomenuté heslo

Zadejte e-mailovou adresu, se kterou jste vytvářel(a) účet, budou Vám na ni zaslány informace k nastavení nového hesla.

Přihlášení

Nemáte účet?  Registrujte se

#ADS_BOTTOM_SCRIPTS#