MMAF

The genetic architecture of morphological abnormalities of the sperm tail
Aminata Touré1,2,3 · Guillaume Martinez4,5 · Zine‑Eddine Kherraf4,6 · Caroline Cazin4 · Julie Beurois4 · Christophe Arnoult4 · Pierre F. Ray4,6 · Charles Coutton4,5

Received: 10 October 2019 / Accepted: 6 January 2020
© Springer-Verlag GmbH Germany, part of Springer Nature 2020

Abstract
Spermatozoa contain highly specialized structural features reflecting unique functions required for fertilization. Among them, the flagellum is a sperm-specific organelle required to generate the motility, which is essential to reach the egg. The flagellum integrity is, therefore, critical for normal sperm function and flagellum defects consistently lead to male infertility due to reduced or absent sperm motility defined as asthenozoospermia. Multiple morphological abnormalities of the flagella (MMAF), also called short tails, is among the most severe forms of sperm flagellum defects responsible for male infertility and is characterized by the presence in the ejaculate of spermatozoa being short, coiled, absent and of irregular caliber. Recent studies have demonstrated that MMAF is genetically heterogeneous which is consistent with the large number of proteins (over one thousand) localized in the human sperm flagella. In the past 5 years, genomic investigation of the MMAF phenotype allowed the identification of 18 genes whose mutations induce MMAF and infertility. Here we will review information about those genes including their expression pattern, the features of the encoded proteins together with their localization within the different flagellar protein complexes (axonemal or peri-axonemal) and their potential functions. We will categorize the identified MMAF genes following the protein complexes, functions or biological processes they may be associated with, based on the current knowledge in the field.

Electronic supplementary material The online version of this article (https://doi.org/10.1007/s00439-020-02113-x) contains supplementary material, which is available to authorized users.
 Charles Coutton [email protected]
1 Faculté de Médecine, Université Paris Descartes, Sorbonne Paris Cité, 75014 Paris, France
2 INSERM U1016, Institut Cochin, 75014 Paris, France
3 Centre National de La Recherche Scientifique UMR8104, 75014 Paris, France
4 INSERM U1209, CNRS UMR 5309, Institute for Advanced Biosciences, Team Genetics Epigenetics and Therapies
of Infertility, Univ. Grenoble Alpes, 38000 Grenoble, France
5 CHU Grenoble Alpes, UM de Génétique Chromosomique, 38000 Grenoble, France
6 CHU Grenoble Alpes, UM GI-DPI, 38000 Grenoble, France

Introduction
The sperm tail or flagellum is an evolutionary conserved organelle, which sustains sperm motility and is, therefore, indispensable for sperm progression within the female geni- tal tract and fertilization. It is composed of a microtubule- based structure, called the axoneme, which comprises nine microtubules doublets (MTD), each with a type-A and -B microtubule, organized around a central pair of microtu- bules (CP) (9 + 2 pattern) (Inaba 2007, 2011). The peripheral doublets are connected to each another by the nexin–dynein regulatory complex (NDRC) and to the central pair by mul- tiprotein T-shaped structures, called the radial spokes (RSs). This latter protein complex not only confers stability to the axonemal structure but also functions as a scaffold for sig- nalling molecules such as Calmodulin and Protein kinase A that regulate flagellar beating (Heuser et al. 2012b; Yang et al. 2006). Importantly, multiprotein ATPase complexes, called the Inner and Outer Dynein Arms (IDAs, ODAs), constitute the motor protein complexes and are key elements for flagellar beating. IDAs and ODAs are attached to the type-A microtubule of each MTD and provide sliding of the

axoneme and flagellar movement by anchoring to the type-B microtubule of the contiguous MTD (Fig. 1). In mammals, the sperm flagellum also harbours peri-axonemal structures that surround the axoneme and are required for structural cohesion, energy regulation and cell signalling (Eddy 2007; Eddy et al. 2003). The sperm tail is divided into three main regions based on the nature of these peri-axonemal struc- tures. The midpiece (MP) contains the mitochondrial sheath (MS) and Outer Dense Fibers (ODFs), which ensure elastic- ity and structural integrity (Lehti and Sironen 2017). The principal piece (PP) comprises the Fibrous Sheath (FS) with two large longitudinal columns attached to doublets 3 and 8 that partially displace ODFs and are connected by semi- circular ribs. The FS is enriched in proteins stabilized by di-sulphide bonds and contributes to sperm structure and flexibility (Eddy et al. 2003); it also harbours glycolytic enzymes and signalling molecules, which are essential for regulating sperm motility and functionality (Eddy 2007; Lehti and Sironen 2017). Last, the terminal piece is only composed of the axoneme and devoid of any peri-axonemal structure. Along the flagellum, two specific structures are also important: the connecting piece, which anchors the tail to the sperm head (Inaba 2007, Inaba 2011) and the annulus (also called Jensen’s ring), which is a Septin-ring structure located at the junction of the midpiece and the principal piece, behaving as a diffusion barrier (Toure et al. 2011) (Fig. 1).
Morphological and/or functional defects of the sperm
flagellum induce asthenozoospermia, which, according to the normal values established by the World Health

Organization reference values, is defined by less than 32% of progressive motile sperm in the ejaculate (Cooper et al. 2010). Due to the conserved axonemal structure between motile cilia and sperm flagella, asthenozoospermia may be part of a syndromic pathological conditions known as Primary Ciliary Dyskinesia (PCD, #MIM 244,400), which is mainly characterized by chronic airway infections with bronchitis and rhinosinusitis due to motile cilia dysfunc- tion (Horani et al. 2016; Knowles et al. 2016). However, asthenozoospermia is also evidenced in many infertile men with no other symptomatology (i.e. isolated asthenozoo- spermia) and overall it constitutes the most frequent sperm defect observed in infertile men as it is found with variable degrees of severity in more than 80% of infertile men (Curi et al. 2003). Accordingly, asthenozoospermia is associated with a wide range of sperm tail morphological defects such as abnormal mitochondrial sheath, abnormal head–tail or midpiece–principal piece junction, abnormal tail bending or coiling, irregular tail caliber or abnormal residual cyto- plasm (Auger et al. 2016; Escalier and Toure 2012). The most severe form of these morphological defects consists in the total or near absence of the tail, a phenotype called “short tails” or “stump tails” (Barthelemy et al. 1990; Chemes et al. 1987; Escalier 2006; Ohmori et al. 1993) and more recently termed as ‘Multiple Morphological Abnormalities of the sperm Flagellum’ (MMAF), which is associated with a condition of extreme asthenozoospermia with near zero progressive sperm (Ben Khelifa et al. 2014). MMAF is characterized by a mosaic of sperm cells with absent, short, irregular and coiled flagellum that can be

Fig. 1 Schematic representation of mammalian spermatozoa and fla- gellum structure. Left panel: overall view of the spermatozoa show- ing the main head and flagellum structures and compartments. Right panel: cross section from the principal piece of the flagellum show- ing the organization of the axoneme: microtubule doublets (MTD),

central pair (CP), radial spokes (RS), nexin dynein regulatory com- plex (N-DRC), inner and outer dynein arms (IDA, ODA), together with some of the peri-axonemal structures: fibrous sheath (FS), outer dense fiber (ODF) and longitudinal columns (LC)

easily evidenced during semen routine analysis by means of optic microscopy (Fig. 2). At the ultra-structural level, evaluated by transmission electron microscopy, MMAF is associated with a severe disorganization of both axonemal and peri-axonemal structures (Chemes et al. 1987; Escalier 2006; Nsota Mbango et al. 2019) (Fig. 3). In particular, the sperm heads are often attached to large cytoplasmic bags with unassembled microtubule and peri-axonemal

elements in due place of the flagella. The flagellum when present shows a disorganized axonemal structure lacking the central pair and/or peripheral microtubule doublets, and loss of dynein arms can also be observed (Fig. 3). In addition, the longitudinal columns and the fibrous sheath are frequently abnormal, the latter being evidenced as a thickening of the fibrous sheath, which led to an additional appellation for this phenotype, namely ‘Dysplasia of the

Fig. 2 Light microscopy imag- ing of control and MMAF sperm. a Control individual.
b MMAF individual. Semen analysis of a MMAF infertile man showing a mosaic of morphological defects such as sperm with absent (#) and short flagella (*); sperm head are also observed attached to large cytoplasmic bag in due place of the flagella (**). Adapted from Nsota Mbango et al. (2019)

Fig. 3 Transmission Electron Microscopy analysis of control and MMAF sperm. a Human spermatozoa with the head on the left, and the flagellum on the right side. The flagellum is divided into two main compartments: the midpiece, which comprises the mitochondrial sheath, and the principal piece, characterized by the presence of a fibrous sheath surrounding the axoneme. b Sperm from MMAF indi- vidual displays incomplete flagellum with short midpiece and abnor- mal fibrous sheath disposition; c Some sperms lack flagellum and display a large cytoplasmic bag with unassembled axonemal and peri-

axonemal components. d Transversal section of the axoneme showing the regular microtubule organization with nine microtubule doublets surrounding the central pair (9 + 2), in normal sperm. e, f In MMAF individual, the axoneme often display a lack of the central pair or total disorganization. Ac acrosome, Ax axoneme, CP central pair, ODF outer dense fibers, FS fibrous sheath, LC longitudinal column, MTD microtubule doublets, M mitochondria, N nucleus. Adapted from Nsota Mbango et al. (2019)

fibrous sheath’ (DFS) (Chemes et al. 1987; Escalier 2006; Linck et al. 2016; Ross et al. 1973).
Although morphological and ultra-structural defects asso- ciated with MMAF condition were thoroughly described since several decades (Alexandre et al. 1978; Chemes et al. 1987; Escalier 2006; Escalier and Albert 2006; Ohmori et al. 1993; Terquem and Dadoune 1980), genetic investigation of their causes were only recently investigated, such was initi- ated by work performed in 2014 by Ben Khelifa et al., who discovered mutations in DNAH1, encoding for an axonemal dynein of the IDAs preferentially expressed in the testis, as the first genetic cause of MMAF (Ben Khelifa et al. 2014). Since then, the development of NGS and the increasing number of phenotypically well-characterized cohorts from different ethnicities contributed to a burst in the identifica- tion of MMAF-associated genes. Remarkably, the number of MMAF identified genes has reached 18 in less than 5 years and accounts for 30 to 60% of the MMAF cases in the dif- ferent cohorts (Coutton et al. 2019; Liu et al. 2019a). Those identified MMAF-associated genes are distinct from genes known to induce a PCD phenotype in humans and encode for proteins locating to both axonemal and peri-axonemal struc- tures of the sperm flagellum (Nsota Mbango et al. 2019). If some of these proteins, such as DNAH1, correspond to well- defined components of the core axoneme, in most cases the exact functions and molecular mechanisms of the ‘MMAF proteins’ are so far unknown, which in fine constitutes a pre- cious source to decipher some of the mechanisms required for flagellum assembly and function. In this review, we will describe the current knowledge about the MMAF-associ- ated genes, which were uncovered by genetic screening of MMAF patients combined with information provided by the studies of their orthologs in various animal models including the mouse and some flagellated protists such as T. brucei, Tetrahymena and the algae Chlamydomonas reinhardtii. Only genes which were demonstrated to induce isolated male infertility due to MMAF phenotype, in patients with no clinical features of PCD or ciliopathies, will be reported in this review.

Repertory of the gene mutations inducing MMAF
In the past 5 years, genetic investigation of the MMAF phenotype allowed the identification of 18 genes whose mutations induce MMAF and infertility without any clini- cal manifestation of PCD. Here we will review information about those genes including their expression pattern, the fea- tures of the encoded proteins together with their localization within the different flagellar protein complexes (axonemal or peri-axonemal) and their potential functions. We will cat- egorize the identified MMAF genes following the protein

complexes, functions or biological processes they may be associated with, based on the current knowledge in the field (Table 1; Fig. 4). We also fully reported all the pathogenic mutations identified in these MMAF-related genes includ- ing loss-of-function mutations (i.e. frameshift, splice site and stop variants which are predicted to have a profound impact on the mRNA transcript and translated protein) and predicted deleterious missense mutations (Table S1).
Inner and outer dynein arm complexes: IDA, ODA

Inner and Outer Dynein Arms (IDAs, ODAs) are multipro- tein ATPase complexes, which constitute genuine motor pro- teins and are indispensable for flagellar beating. Their com- position and properties have been exhaustively studied and described in protozoa (Kobayashi and Takeda 2012; Vin- censini et al. 2011). In Chlamydomonas, ODAs are required for ciliary beat frequency, whereas IDAs are responsible for bend formation and beating form (Viswanadha et al. 2017). In mammals and in particular in humans, much less informa- tion is known and when available, it mostly refers to motile cilia from respiratory cells (Linck et al. 2016; Viswanadha et al. 2017). In addition, some components are not evolu- tionary conserved (Viswanadha et al. 2017) as illustrated by dynein heavy chains, which exist in three types in Chla- mydomonas (α, β and γ), whereas vertebrate ODAs have only two (β and γ) (King 2012). All this makes it very dif- ficult to extend the knowledge established in protozoa to human sperm flagellum. In this regard, the recent advances in the genetics of MMAF helped in pinpointing some evolu- tionary conserved ODA and IDA proteins, which in humans are specifically required for human sperm flagellum due to highly specialized and restricted functions.
DNAH1

The first gene formally identified in humans with muta- tions causative for a MMAF phenotype and male sterility is DNAH1 (MIM #603332) (Ben Khelifa et al. 2014), a gene preferentially expressed in human testis (Ben Khelifa et al. 2014). This work was performed by Ben Khelifa et al. on a cohort of 20 MMAF consanguineous patients from North Africa and led to the identification of homozygous deleteri- ous mutations (Table S1), associated with severe axonemal disorganisation such as mislocalisation of the peripheral microtubule doublets, absence of the central pair together with the lack of IDAs (Ben Khelifa et al. 2014). Importantly, DNAH1 was subsequently reported to be mutated by other teams in MMAF patients from additional ethnical origins (i.e. Chinese, Iranian, Italian) (Amiri-Yekta et al. 2016; Sha et al. 2017a; Wang et al. 2017, Tu et al. 2019) confirming that mutations in DNAH1 constitute a recurrent cause of isolated male sterility. Comprehensive analysis performed

Table 1 Repertory of the genes identified with mutations in MMAF infertile men together with the corresponding phenotypes in different mutant models and ICSI outcomes for patients
Gene Protein features Protein localisa- Phenotype of mutant models Clinical outcome of patients References

chain, ATPase

(IDA)

mutant): reduced speed of swimming

and Li et al. (2019c)

DNAH6 Dynein Heavy chain, ATPase

Inner Dynein Arm (IDA)

– – – – 1 patient Abortion (1/1) Tu et al. 2019

DNAH17 Dynein Heavy chain, ATPase

Outer Dynein Arm (ODA)

KO model: MMAF phenotype
KI model: asthe- nozoospermia (no MMAF)

– – – 4 patients Pregnancy (0/4) Whitfield et al.
(2019) and Zhang et al. (2019a)

CFAP43 WD repeat domains, coiled- coil domain

Tetrahymena, Chla- mydomonas: Fap43p location toT/TH complex
Trypanosoma: TbCFAP43
location between MTD 5, 6 and parafla- gellar rod

MMAF phenotype Fap43 mutant:
reduced sliding and swimming velocity

Fap43 gene dele- tion: altered waveform,
beat stroke and reduced swim- ming speed

TbCfap43
(Tb.927.4.5380)
RNAi mutant: cell growth defects, Abnormal flagellum beat- ing (axonemal disorganization), normal flagellum length

1 patient Pregnancy (1/1) Tang et al. (2017),
Fu et al. (2018), Kubo et al. (2018), Urbanska
et al. (2018), Coutton et al. (2018) and Sha et al. (2019b, c)

CFAP44 WD repeat

Tetrahymena,

Asthenozoo-

Fap44 mutant:

Fap44 gene dele-

TbCfap44

4 patients Pregnancy (2/4) Tang et al. 2017;

Trypanosoma: TbCFAP44
location between MTD 5, 6 and parafla-

2017)

ing (axonemal disorganization), normal flagellum length

et al. (2019b, c)

gellar rod

Table 1 (continued)
Gene Protein features Protein localisa-

Phenotype of mutant models Clinical outcome of patients References

ependyma Cfap70 RNAi: reduced cilia beating fre- quency

defects in ODA activity and reduced flagellar motility

(2018) and Beurois et al. (2019)

WDR66 (CFAP251)

WD repeats, cal- cium regulating EF-hand domain

Mammalian sperm: flagella
Trypanosoma: flagella
Tetrahymena, Chla- mydomonas: radial spoke

– – Fap251 mutant: reduced cell swimming, Impaired ciliary beating coordi- nation, normal length

Tbwdr66
(Tb927.3.1670)
deletion: abnormal motil- ity (axonemal disorganization), growth defects, normal flagellum length

No – Heuser et al. (2012a, b), Urban- ska et al. (2015) and Kherraf et al. (2018)

FSIP2 AKAP4 Interact- ing domain

Mammalian sperm: fibrous sheath of the flagella

– – – – No – Brown et al. (2003)

CEP135 – Human sperm: centriole

– – – – 1 patient Pregnancy (0/1) Sha et al. (2017b)

TTC21A TPR repeats – MMAF phenotype – – – No – Liu et al. (2019c)

TTC29 TPR repeats Mammalian sperm: flagella
Trypanosome: TbTTC29 loca- tion to axoneme
Chlamydomonas: p44 location to IDA

Asthenozoo- spermia (no MMAF)

– – TbTTC29 (Tb927.3.1990)
RNAi mutant: normal falgel- lar length and
structure, reduced swimming, increased sedi- mentation

3 patients Pregnancy (2/3) Yamamoto et al.
(2008), Lorès
et al. (2019) and Liu et al. (2019a)

SPEF2 IFT20 binding domain

Human: sperm flagellum mid- piece

MMAF phenotype – – – 1 patient Pregnancy (0/1) Lehti et al. (2017)
and Liu et al. (2019b; d)

by means of exome screening of different cohorts, reaching up to 178 MMAF individuals from various ethnicities (Cau- casians, North and Sub-Saharan Africans, Iranians) found that DNAH1 mutations account for 6% of the MMAF cases in this cohort (Coutton et al. 2018, 2019).
DNAH1 encodes for an inner arm Dynein Heavy Chain (DHC), and in Chlamydomonas, its paralogs, Dhc1, is required for IDA assembly and flagellar motility (Myster et al. 1997, 1999) (Fig. 4). Invalidation of the mouse par- alog, Mdhc7, also induces asthenozoospermia but no mor- phological defects of the sperm tail were observed and surprisingly, a reduced ciliary beating was also described (Neesen et al. 2001) while in humans, no PCD clinical man- ifestations were reported in MMAF individuals carrying DNAH1mutations (Ben Khelifa et al. 2014; Amiri-Yekta et al. 2016; Sha et al. 2017a; Wang et al. 2017). Moreover, to date, no mutations in DNAH1 were formally identified in PCD patients (Horani et al. 2016; Kurkowiak et al. 2015). Only one study reported a homozygous DNAH1 missense mutation (p.Lys1154Gln) in two sisters with PCD (Imtiaz et al. 2015). However, the causality of this missense muta- tion in the PCD phenotype observed in both sisters was not demonstrated. The report of the identification of a relatively mild mutation without any functional validation in single familial case remains somewhat unconvincing. The discrep- ancy observed between the mouse and human phenotypes may result from the persistence of a truncated protein in the mouse model (Neesen et al. 2001) in contrast to the MMAF individuals; such truncated protein could indeed act as a dominant negative in motile cilia while it could be absent in the sperm cells due to residual cytoplasm elimination occurring during spermiogenesis. Nonetheless, this clearly suggests possible functional differences between species. In support of this, recent work from Hu et al. reported the phe- notypical characterization of a ENU-induced mutant allele of Dnah1, ferf1, which carries the homozygous p.Y3897H missense mutation in DNAH1. Intriguingly, those mutant mice produce apparently normal sperm cells but with clumping and motility abnormalities leading to fertiliza- tion failure (Hu et al. 2019).
DNAH2

Very recently Li et al. performed whole-exome sequencing (WES) of 38 Han Chinese infertile patients with a MMAF phenotype and identified three probands with bi-allelic mutations in DNAH2 (MIM *603,333) (Table S1), which based on public expression databases, also encodes for an axonemal inner arm Dynein Heavy Chain (DHC) prefer- entially expressed in the testes (Li et al. 2019c) (Fig. 4). Although four out of the five identified mutations were missense, they observed their low frequency or absence in variant databases and reduced amount of DNAH2 protein in

Fig. 4 Localization in mature spermatozoa of proteins encoded by the MMAF-related genes identified in human. Schematic representation of the flagellum structure and cross sections through a representative segment of the intermediate piece, principal piece and terminal piece are shown. Cross-sections of the flagellum showing the axoneme are enlarged and the offset shows: the nine outer microtubule doublets of the axoneme with associated inner dynein arms, outer dynein arms, radial spokes, nexin–dynein regulator complex (N-DRC), nexin links

and the central pair of microtubule doublets. The longitudinal view of the axoneme shows in particular the localization of the calmodulin and spoke-associated complex (CSC), the T/TH complex, the inter- mediate chain–light chain complex of inner dynein arms (IC/LC) and the modifier of inner arms (MIA) complex 1/2. Proteins encoded by the MMAF-related genes identified in human are reported in red. Pro- teins with unknown localization (AK7 and ARMC2) are indicated separately (black box)

sperm from the mutated MMAF individuals. Similar to indi- viduals with DNAH1 mutations, they observed the absence of the DNAL11 protein suggesting that IDAs are severely impaired in sperm from DNAH2 patients (Li et al. 2019c). In keeping with DNAH2 preferential expression in the testis, to date no patients suffering from PCD were identified with mutations in DNAH2 (Horani et al. 2016; Kurkowiak et al. 2015). However, recent work revealed that in vitro, Dnah2 siRNAi-mediated depletion in mouse multiciliated ependymal cells results in cilia immotility (Zhang et al. 2019c), suggesting that in the mouse, this dynein heavy
chain might also be essential for ciliary function.
DNAH6

Recently, mutations in DNAH6 (MIM *603336), encoding for an inner dynein heavy chain, were identified in three infertile patients with MMAF phenotype from two unre- lated Han Chinese families (Tu et al. 2019) (Table S1). Two

infertile brothers harbored two compound heterozygous mis- sense variants of DNAH6 (p. D2194E and p. G3753D) and the third unrelated patient had also two compound heterozy- gous variants including a frameshift mutation (p.S941fs) and a missense mutation (p.R3342H). Three-dimensional analysis of DNAH6 protein structure was used to confirm the impact of the missense mutations (p. D2194E and p. G3753D) and evidenced that the mutations may gener- ate a misfolded protein with potentially reduced ATPase activity (Tu et al. 2019). Immunofluorescence experiments showed that DNHA6 protein locates to the sperm flagel- lum of control individuals and that its amount and distribu- tion pattern were not changed in the sperm from patients with DNA6 mutations. In contrast, the protein amounts of DNAH1, SPAG6 and AKAP4 were remarkably reduced in spermatozoa from patients carrying DNAH6 mutations rela- tive to the controls, indicating that DNAH6 mutations are associated with severe axonemal and peri-axonemal disor- ganization (Tu et al. 2019). Intriguingly, DNAH6 mutations

were previously associated with heterotaxia, azoospermia, premature ovarian insufficiency and changes of lung func- tion in cystic fibrosis in humans (Norling et al. 2014; Li et al. 2016; Gershoni et al. 2017; Blue et al. 2018). In addi- tion, other compound heterozygous mutations were found in patients with sperm head abnormalities (30% headless spermatozoa and 69% globozoospermia) (Li et al. 2018). Moreover, Li et al. showed that DNHA6 protein is located in the neck region of normal spermatozoa (Li et al. 2018). This wide range of different phenotypes associated with DNAH6 mutations as well as the nature of the reported mutations in MMAF individuals (missense mutations) raise some doubts about the pathogenicity of the identified mutations and also questions about the precise function of DNAH6. Overall this recent identification of DNAH6 mutations in MMAF indi- viduals should be supported by strong functional validation.
DNAH17

Whitfield et al. identified mutations in DNAH17 (MIM
*610063), encoding for an outer dynein heavy chain, in five asthenozoospermic patients with a MMAF phenotype (Whitfield et al. 2019) (Fig. 4) (Table S1). They formally demonstrated by TEM that these mutations were associated with a loss of the ODAs in the sperm cells of the patients but not in their respiratory ciliated cells. In humans, the com- position of ODAs was so far only described in respiratory cells, where they are known to form a globular head domain, which comprises β- and γ-type heavy chains (β-HCs, γ-HC), associated with an intermediate domain and various light chains (Hom et al. 2011; Nicastro et al. 2006). By analysing data from tissue expression databases and literature (Amaral et al. 2013; Darde et al. 2015, 2019; Wang et al. 2013) and performing immunodetections and western blot assays of all outer arm dynein heavy chains, Whitfield et al. demonstrated that ODAs from the sperm cells harbour a set of dynein heavy chains (γ-HC DNAH8 and β-HC DNAH17), which is distinct from that of human airway epithelial cells (γ-HC DNAH5 and β-HCs DNAH9, DNAH11) (Whitfield et al. 2019). This plainly explains the absence of ciliary dysfunc- tion and the sperm-specific phenotype observed in MMAF individuals carrying mutations DNAH17. Importantly, they also demonstrated that in contrast to the ciliary β heavy chains, which display a regional localisation, with DNAH11 located at the proximal end and DNAH9 locating at the dis- tal end, the sperm-specific β-HC DNAH17 is homogene- ously distributed along the flagellum (Whitfield et al. 2019). Zhang et al. have just reported the identification of the homozygous DNAH17 missense mutation (p.C1803Y) in three unrelated men presenting with asthenozoospermia but no MMAF phenotype and no ciliary defects (Zhang et al. 2019a) (Table S1). Importantly, the three affected men showed morphologically normal spermatozoa and a less

severe asthenozoospermia than MMAF individuals: 11–25% of total motile sperm (normal value > 40%) (Zhang et al. 2019a) compared to 0–5% in MMAF individuals (Nsota Mbango et al. 2019). Zhang et al. performed an exhaustive description of the ultra-structural defects associated with this mutation based on patient semen analyses and a mutant mouse model harbouring the same mutation. They show that the (p.C1803Y) missense mutation results in the absence of microtubule peripheral doublets 4–7 from the principal piece of the axoneme and in reduced sperm motility and progression, both in humans and mice. They also show that in mice, the phenotype of microtubule destabilization is increased during sperm storage in the cauda epididymides (Zhang et al. 2019a). Importantly, Zhang et al. also have generated a Crispr-Cas9 KO mouse model and demonstrated that DNAH17 loss of function induces a MMAF phenotype in mice (Zhang et al. 2019a), further confirming the patho- genicity of the DNAH17 truncating mutations previously identified by Whitfield et al. in MMAF patients (Whitfield et al. 2019). Overall, the data provided by Zhang et al. indi- cate that the DNAH17 (p.C1803Y) missense has less impact in sperm axonemal structure and stability than DNAH17 loss of function and is compatible with minimal sperm motility (Zhang et al. 2019a). This again underlines possible geno- type–phenotype correlation in the MMAF phenotype and asthenozoospermia, which should be further investigated.

IDA associated complex: T/TH complex
A peculiar complex, called the tether/tether head (T/TH) complex, was recently identified by cryo-electron tomogra- phy in Chlamydomonas (Heuser et al. 2012a). This complex, although tightly connected to the IDAf (I1), is not required for IDA assembly but important for regulating their motor activity (Urbanska et al. 2018). The T/TH complex was shown to interact with the I1 dynein motor domain and with the radial spoke RS3, also involved in beating regulation (Fu et al. 2018; Kubo et al. 2018). Genetic analysis of MMAF patients have identified mutations in CFAP43, CFAP 44 and CFAP65, which correspond to orthologous genes encoding proteins of the (T/TH) complex, in Chlamydomonas and Tet- rahymena. Importantly, in humans the absence of CFAP43, 44 and 65 is deleterious for sperm axoneme assembly in contrast to what was observed in Chlamydomonas, where Fap43 and Fap44 deletion only impacted the regulation of flagellar beating while the integrity of the axoneme structure and of IDA was preserved.
CFAP43, CFAP44

Tang et al. first identified bi-allelic loss of function muta- tions in CFAP43/WDR96 (MIM #617592) and CFAP44/

WDR52 (MIM #617593) in Han Chinese patients (Table S1) and generated two knockout mouse models deficient for the orthologous proteins, which they show to induce male steril- ity and to recapitulate the human MMAF phenotype (Tang et al. 2017). Subsequently Coutton et al. also identified CFAP43 and CFAP44 mutations in their cohort of MMAF individuals, accounting for 12.8% and 7.7%, of the cases (Coutton et al. 2018) (Table S1). Coutton et al. assessed the impact of Cfap43 and Cfap44 absence on mouse sper- matogenesis by generating knockout (KO) animals using the CRISPR-Cas9 technology. All KO strains for Cfap43 presented the same reproductive phenotype with 100% of sperm displaying a typical MMAF phenotype. In contrast, sperm from Cfap44−/− males had sperm cells showing com- plete immotility but with a normal flagellum length asso- ciated with subtle morphological abnormalities (Coutton et al. 2018). They also demonstrated that RNAi-depletion of Cfap43 and CfapP44 in the flagellated parasite T. bru- cei induced growth defects, abnormal flagellum beating, axonemal disorganization but normal flagellum length (Coutton et al. 2018). Several other mutations in CFAP43 and CFAP44 were identified later in different replicative studies confirming that the CFAP43 and CFAP44 mutations are important and recurrent causes of MMAF (Sha et al. 2019c; Wu et al. 2019) (Table S1). CFAP43 and CFAP44 encode cilia- and flagella-associated proteins (CFAPs) with WD repeat domains (WDR), involved in protein interactions (Smith 2008). To date, due to the lack of efficient antibodies, their exact function and localization in human or mammalian sperm are unknown. However, very recent work performed in Tetrahymena reported the existence of a small complex, which is composed of both orthologous Fap43p and Fap44p proteins. In addition, Fap43p and Fap44p were shown to interact with Fap57Ap, the ortholog of human CFAP65/ WDR65 (Urbanska et al. 2018). Importantly, lack of either Fap43p or Fap44p affected ciliary beating, and location of both proteins in Tetrahymena cilia was interdependent. In addition, from biochemical and mutant analyses, the Fap43p/ Fap44p protein complex was shown to locate in close proximity to IDA I1 although it is not required for IDA I1 assembly (Urbanska et al. 2018). This protein complex cor- responds to the so-called tether/tether head (T/TH) complex identified by cryo-electron tomography in Chlamydomonas, which connects the motor domain of IDA I1 to the outer microtubule doublet and regulate motor activity (Heuser et al. 2012a) (Fig. 4). Consistent with this, Fap43 and Fap44 proteins were also identified in Chlamydomonas as com- ponents of the T/TH complex and shown to be required for flagella beating and movement speed (Fu et al. 2018; Kubo et al. 2018; Urbanska et al. 2018). Intriguingly, Coutton et al. reported a distinct localization of the orthologous TbC- FAP43 and TbCFAP44 proteins in T. brucei. They observed that both proteins were not uniformly distributed along the

axoneme but primarily localized between the peripheral doublets (5 and 6) and the paraflagellar rod. Conversely to Tetrahymena or Chlamydomonas organelles in which such peri-axonemal structure is not present, mammalian sperm flagellum harbours accessory structures. Therefore, CFAP43 and CFAP44 proteins may have a specific localiza- tion in mammalian sperm and may be involved in connecting axonemal and peri-axonemal structures. Nevertheless, the localization of TbCFAP43 and TbCFAP44 in the (T/TH) complex of T. brucei and mammalian sperm would need to be properly investigated.
A recent study reported a nonsense variant in CFAP43 that segregated with normal-pressure hydrocephalus (NPH) in one Japanese family (Morimoto et al. 2019) suggesting that CFAP43 mutation may affect not only the testis but also other ciliated tissues. Intriguingly, the mutation identified in the family was heterozygous. This nonsense variant is located in the exon 35 (p.Trp1502Ter) corresponding to the C-terminal domain of the protein which seems essen- tial for the localization of CFAP43 in cilia (Urbanska et al. 2018). To confirm the effect of CFAP43 truncation on the pathogenesis of hydrocephalus, Morimoto et al. introduced a 14-bp deletion in exon 35 of the mouse ortholog Cfap43 using CRISPR/Cas9 technology. The generated Cfap43 knockout mouse also exhibited a hydrocephalus phenotype with morphologic abnormality of motile cilia mimicking human NPH (Morimoto et al. 2019). Such clinical features were, however, not evidenced in the two other Cfap43 KO mice previously reported (Tang et al. 2017; Coutton et al. 2018). Moreover, homozygous nonsense mutations in the C-terminal domain of CFAP43 were previously reported in MMAF patients but none presented with NPH or other clini- cal features of ciliopathies (Tang et al. 2017; Coutton et al. 2018; Sha et al. 2019c; Wu et al. 2019). Interestingly, Mori- moto et al. detected the mRNA Sharbouring the premature termination codon in the brain and trachea of mutant mice (Morimoto et al. 2019) suggesting that a truncated protein may still be produced. We may thus suppose that such trun- cated protein could act as a dominant negative in motile cilia and may thus explain the NPH phenotype observed in patients with a heterozygous nonsense variant in the C-ter- minal part of the CFAP43 gene.
Phenotypic differences were observed between some KO
mouse models generated to study the function of CFAP43 and CFAP44 during the spermatogenesis. For CFAP43, the absence of other clinical features in the model presented by Coutton et al. and Tang et al. may be due to the fact that the guide RNAs were designed against the central exons 21 and 22, respectively, while Morimoto et al. targeted the terminal exon 35 (Tang et al. 2017; Coutton et al. 2018; Morimoto et al. 2019). As well for CFAP44, the phenotype discrepan- cies between the KO model generated by Coutton et al. (pure asthenozoospermia) and Tang et al. (MMAF-like phenotype)

may be explained by the choice of the exon targeted (3 and 15, respectively) (Tang et al. 2017; Coutton et al. 2018). The mild phenotype reported by Coutton et al. might be due to the persistence of small isoforms not inactivated by the guide RNA against the proximal exon 3 or the presence of truncated functional protein as well as a different genetic background of the mouse used but it remains to be clearly elucidated.

CFAP65

Tang et al. reported a first homozygous nonsense mutation in CFAP65 (MIM #617592) in a single MMAF case (Tang et al. 2017) (Table S1). Further analyses of Chinese and non-Chinese cohorts identified MMAF individuals with null mutations in CFAP65. Five novel bi-allelic deleterious mutations in CFAP65 (one homozygous nonsense muta- tion (p.E1781X), two compound heterozygous mutations (p.R762X and p.P584fs) and two compound heterozygous mutations (p.L1905fs and p.N1007K)) were then identified in three individuals by Wang et al., but the mutated MMAF patients also displayed an acrosomal hypoplasia, a pheno- type which was not previously highlighted for other MMAF individuals (Wang et al. 2019) (Table S1). In line with the acrosome defects, Wang et al. reported that the CFAP65 protein locates both at the acrosome and the midpiece of human sperm flagellum. More importantly, the outcome of Intracytoplasmic Sperm Injection (ICSI) for those CFAP65 mutated MMAF individuals was compromised, suggesting that CFAP65 deficiency may also affect the sperm head, chromatin and/or the integrity of the sperm centrosome (Wang et al. 2019). By performing exome sequencing of 88 Han Chinese MMAF infertile individuals, Li et al. also identified CFAP65 homozygous and compound heterozy- gous mutations (including four frameshift and three stop- gain mutations) in six unrelated probands (Table S1). They also analysed exome data from a previously reported cohort of 167 additional MMAF non-Chinese individuals and iden- tified two additional homozygous CFAP65 variants likely affecting splicing (Li et al. 2019b). Li et al. also generated a mouse mutant model with a homozygous Cfap65 frame shift mutation, which induced a MMAF-like phenotype and male sterility, thus confirming that CFAP65 is required for sperm flagellum assembly and structure (Li et al. 2019b) (Fig. 4). Last, a novel homozygous nonsense mutation in CFAP65 was described in another Han Chinese MMAF patient leading to the absence of CFAP65 protein in sperm flagellum (Zhang et al. 2019b) (Table S1). In addition, the authors investigated the expression level of Cfap65 in dif- ferent organs of adult mice and in mouse testes at different postnatal days and demonstrated that Cfap65 is restricted in mouse testes and is mainly expressed in the round and

elongated spermatids during the spermiogenesis (Zhang et al. 2019b).
In protists the Fap43p and Fap44p are part of a complex including Fap57Ap, the ortholog of CFAP65, but in mam- mals, no information regarding CFAP65 interacting proteins is currently known and the link with head sperm function- ality remains obscure. However, the mammalian CFAP65 protein is speculated to be a transmembrane protein as it contains a transmembrane helix region near the N-termi- nus. In addition, it also contains an ASH domain present in proteins associated with cilia, flagella, centrosome and the Golgi complex together with modules involved in pro- tein–protein interaction (MSP domain and coiled-coil near the C-terminus) (Wang et al. 2019). Further studies may help in better defining the relation between CFAP65 and sperm head functionality.

ODA associated complex: CFAP70 complex
In 2018, the existence of a novel regulatory component of ODAs, in both cilia and flagella, was reported by Shamoto et al. In this work, the authors characterized the CFAP70 protein, which contains a cluster of tetratricopeptide repeat (TPR) domains located at the N-terminus known to be pre- sent in many proteins involved in the intraflagellar transport (IFT) complex (Zeytuni and Zarivach 2012). They demon- strated that the TPR region is required for CFAP70 axone- mal localization (Shamoto et al. 2018). They also showed that the knock down of CFAP70 in cultured mouse epend- yma resulted in reduction of ciliary beating frequency while ODA assembly and ultra-structure were unaffected (Shamoto et al. 2018). Interestingly, in Chlamydomonas, cryo-electron tomography indicated that the N-terminus of the orthologous protein FAP70 resided stably at the base of the ODAs (Sha- moto et al. 2018) and in zebrafish, the CFAP70 orthologous gene was identified as a novel cilia-related TTC gene, called ttc18 (Xu et al. 2015), involved in ciliary length and function (van Dam et al. 2019). This strongly suggested the existence of a molecular protein complex including CFAP70, which is involved in ODA regulation in both cilia and flagella. This strongly suggested the existence of a molecular protein com- plex including CFAP70, which is involved in ODA regula- tion in both cilia and flagella. In line with these findings, recent genetic analyses identified mutations in CFAP70, con- firming the importance of this protein in mammalian flagella (Beurois et al. 2019).
CFAP70

Beurois et al. identified two unrelated MMAF individuals carrying mutations in CFAP70: one patient had a homozy- gous splice variant altering a consensus splice acceptor site

of CFAP70 exon 16, and one had a likely deleterious mis- sense variant in exon 3 (p.Phe60Ile) (Table S1). In the sperm cells from the patient carrying the splice variant, immuno- fluoresence analysis demonstrated the absence of CFAP70 protein in the sperm flagellum. Interestingly, authors also showed the absence of SPAG6 and DNAI2 protein in sperm from the MMAF patient carrying a CFAP70 splic- ing mutation, indicating that the CPC and ODA complexes are impacted (Beurois et al. 2019). In addition, whereas CFAP70 staining was present in sperm cells from patients with mutations in the three other MMAF-related genes (ARMC2, FSIP2 and CFAP43), Beurois et al. observed an absence of staining in sperm cells from patients mutated in the WDR66 gene, suggesting a possible interaction between two different axonemal components (Beurois et al. 2019). Regrettably, no information regarding the axonemal ultra- structure of the sperm cells from the MMAF patient carrying a CFAP70 mutation was reported, thus, precluding to state on the integrity of the ODAs (Beurois et al. 2019). Further characterization of MMAF patients with CFAP70 mutations should help to state on this point and to clearly define the function of CFAP70 as a sole regulator of ODA activity (as hypothesized by Shamaoto et al. in the mouse (Shamoto et al. 2018) or as a protein also required for ODA assembly/ structure. Nonetheless, this work demonstrates that muta- tions in ODA components/regulators, similarly to IDA, are deleterious for the whole axonemal structure of the sperm flagellum in contrast to motile cilia.

Radial spoke complex: RS
The radial spoke (RS) is a multiprotein complex, which comprises at least 23 proteins identified in Chlamydomonas, and behaves as a mechanochemical transducer between the central pair and the peripheral microtubule doublets (Pigino and Ishikawa 2012). In Chlamydomonas, three types of RS have been described, RS1, RS2 and RS3, and mutants lack- ing RSs show no flagellar beating. By regulating the activity of the dyneins, RSs are essential for beating of both motile cilia and flagella (Pigino and Ishikawa 2012).
CFAP251 (WDR66)

Kheraff et al. identified by exome sequencing a homozy- gous intragenic deletion encompassing exons 20 and 21 of CFAP251/WDR66, (MIM #618152), in 7 patients out of 78 MMAF individuals (Table S1). This mutation event, con- firmed by comparative genomic hybridization, was only observed in North African (Tunisia) individuals from this cohort and absent from all public variant databases, sug- gesting a founder effect (Kherraf et al. 2018). CFAP251 encodes for a 941 amino acid protein with 9 WD40-repeat

domains, known to be important for protein–protein interaction (Smith 2008), and a calcium-binding EF hand domain located at the carboxy-terminal extremity. The genomic deletion affected the carboxy-terminal region of WDR66, and removed the calcium regulating EF-hand domain, suggesting its importance for flagellar structure and beating (Kherraf et al. 2018). Auguste et al. concomi- tantly demonstrated CFAP251 implication in the MMAF phenotype and identified bi-allelic frameshift mutation (p.Asp42Metfs*4) in CFAP251 in two MMAF siblings from Lebanon and two compound heterozygous mutations (p.Glu111Ter and p.Leu530Valfs*4) in a third French unre- lated MMAF individual (Auguste et al. 2018) (Table S1). The authors showed that CFAP251 is absent from sper- matozoa in two independent MMAF individuals carry- ing these distinct WDR66 mutations. In addition, using immunofluorescent and transmission electron microscopy, Auguste et al. provided evidence that loss of CFAP251 affects the formation of the mitochondrial sheath. Last, Li et al. identified three patients with bi-allelic CFAP251 loss of function mutations out of 65 Han Chinese MMAF indi- viduals: one patient with a homozygous nonsense mutation (p.Arg267*), one with a homozygous splice-site mutation (c.1286+2T>C) and the third patient with two compound heterozygous mutations including a nonsense mutation (p.Gln139*) and a frameshift mutation (p.Phe574Leu*3) (Li et al. 2019a). Overall this indicated that CFAP251 mutations constitute a recurrent cause of MMAF and male sterility in various geographical populations.
Although in humans CFAP251 precise location and function within the axoneme remain to be determined, it was shown to locate to the sperm flagellum (Kherraf et al. 2018; Auguste et al. 2018). In the unicellular flag- ellated alga Chlamydomonas, CFAP251 was identified in the calmodulin and spoke-associated complex (CSC), a complex which binds to the MTD and is essential for anchoring RS2 and RS3 (Dymek et al. 2011; Dymek and Smith 2007) (Fig. 4). In addition, in the ciliated protozoa Tetrahymena, CFAP251 is required for efficient waveform and coordinated ciliary beating (Urbanska et al. 2015). In
T. brucei, CFAP251 was also shown to locate to the flagel- lum and its depletion through RNAi-induced severe axone- mal and peri-axonemal disorganization and impairment of the parasite movement. Kherraf et al. showed that the terminal WDR66 exonic deletion led to a truncated pro- tein which is still detected using immunofluorescence in the sperm flagella from the mutated patients. The authors subsequently generated Trypanosoma cell lines express- ing a truncated CFAP251 protein without its C-terminal domain mimicking the human deletion and showed that this truncated protein could properly locate to the parasite flagellum but could not rescue the phenotype induced by

the RNAi, confirming that the EF-hand domain is required for flagellar structure and beating (Kherraf et al. 2018).

Peri‑axonemal structures
The fibrous sheath (FS) constitutes a major accessory struc- ture of the sperm flagellum, which contributes to sperm structure and flexibility (Eddy et al. 2003), together with motility regulation (Eddy 2007; Lehti and Sironen 2017). As the organisation of the FS is severely impacted in sperm from MMAF individuals, one would expect that muta- tion in FS components or proteins interacting with the FS could result in a MMAF phenotype and male sterility. Indeed in 2002, invalidation of the Akap4 gene, encoding for one the most abundant protein of the FS (Carrera et al. 1994), was shown to induce a MMAF-like phenotype in the mouse (Miki et al. 2002). Following this work, Bac- etti et al. reported partial genomic deletions of both AKAP3 (chromosome 12) and AKAP4 (chromosome X), which they identified by PCR in a single MMAF subject (Baccetti et al. 2005), but no further molecular analyses of the transcripts or protein were performed to confirm the mutation, and this result, therefore, remains uncertain. To date no other MMAF individuals were reported with a mutation in AKAP pro- teins; however, mutations in FSIP2, another component of the FS, were recently identified (Fig. 4).
FSIP2

Martinez et al. first reported mutations in FSIP2 (MIM #618153), encoding for the Fibrous Sheath (FS) Integra- tion Protein, which they show to account for 5.1% of their cohort (Martinez et al. 2018) (Table S1). A more recent study also identified homozygous loss-of-function muta- tions (one nonsense and one frameshift) of FSIP2 in two (5%) out of 40 Chinese MMAF subjects analysed (Liu et al. 2019e) (Table S1). Interestingly, the absence of AKAP4 pro- tein, known to interact with FSIP2 (Brown et al. 2003), was observed in sperm from FSIP2 mutated patients (Martinez et al. 2018; Liu et al. 2019e), which is not observed with other MMAF gene mutations and, therefore, constitutes a specific feature of FSIP2 mutations (Martinez et al. 2018). At the morphological and ultra-structural level, FSIP2 muta- tions were associated with a complete disorganization of the FS and severe axonemal defects including a lack of the CP and of dynein arms, as described for other MMAF muta- tions. The severity of the axonemal defects is quite intrigu- ing but might be secondary to the FS defects. Hence, a statis- tically significant lower proportion of sperm with an absent flagellum was observed in comparison to other MMAF individuals (Martinez et al. 2018), suggesting that flagel- lum growth defects might occur later during spermiogenesis.

Overall, this underlines the existence of a complex network of interacting proteins located within the sperm axoneme and the peri-axonemal structures.

Centrosome
Centrosomes are present in all eukaryotic cells and are com- posed of a pair of centrioles, which correspond to cylindri- cal structures with nine triplet microtubules in a “pinwheel” arrangement. The two centrioles are positioned perpendicu- lar to each other and are surrounded by the pericentriolar material (PCM) that constitutes the microtubule organizing center (MOCT) of the cell. Centrosomes are involved in many cell functions such as the movement of cell organelles along microtubules, the organization of the cytoskeleton, the mitotic spindle, and zygote sperm aster. Hence microtubules can originate from the PCM, which constitutes a scaffold for numerous proteins including microtubule binding proteins such as kinesins and dyneins, centrin, pericentrin and spin- dle checkpoint proteins (Schatten and Sun 2009).
The centrosomes have alternative functions in dividing and non-dividing cells. During mitosis, the centrosomes are essential to organize the mitotic spindle for chromosome alignment, duplication and correct segregation between the daughter cells, while in the process of cilia and flagella assembly, centrosomes migrate to the cell periphery and are critical to initiate the assembly of the axoneme with the distal centriole serving as a basal body. In line with these critical functions, many mutations in genes encoding for centrosome and basal body proteins have been identified in human disorders, including cancer and ciliopathies (Betten- court-Dias et al. 2011). However, despite the observations that some sperm defects observed in humans might originate from centrosome defects (i.e. headless sperm flagella or ace- phalic sperm, abnormal tail assembly or abnormal head–tail alignment) (Chemes 2012), to date only one MMAF gene encoding for a centrosome protein was identified and this only concerned one individual (Fig. 4).
CEP135

Sha et al. have identified a single case of Han Chinese MMAF individual carrying the homozygous (p.D455V) missense mutation in CEP135 (MIM *611423) (Sha et al. 2017b) (Table S1). The variant was absent from all public databases and analysis of sperm form the patient indicated abnormal protein distribution, which was found diffused along the midpiece instead of the punctiform pattern char- acteristic of centriolar proteins. In humans, loss-of-func- tion mutations in CEP135 were previously associated with autosomal-recessive primary microcephaly and no informa- tion regarding the fertility was described in those patients

(Hussain et al. 2012). Conversely, no other phenotype was reported for the MMAF sterile individual carrying the p.D455V mutation in CEP135 (Sha et al. 2017b); it is, there- fore, possible that this missense mutation differently affects the centrosome and results in a sperm-specific dysfunction.
The CEP135 gene encodes a centrosomal protein with a coiled-coil domain which binds to microtubules (Kraatz et al. 2016). In vitro studies in human and mammalian cells indicates that CEP135 is required for centriole biogenesis (Kleylein-Sohn et al. 2007; Ohta et al. 2002). In Drosophila, the CEP135 othologue, Bld10, gene localizes to the sper- matid basal body (Mottier-Pavie and Megraw 2009), and Bld10 mutation induces short centrioles and sperm with immotile flagella lacking the central pair (Mottier-Pavie and Megraw 2009). In Chlamydomonas reinhardtii, the Bld10 protein localizes to the cartwheel (basal bodies) and Bld10 mutant also showed abnormal centriole (Hiraki et al. 2007). Overall, CEP135 appears essential for centriole biogenesis and in particular, central pair assembly within the axoneme. Unfortunately, no TEM analysis was performed to specify the axonemal anomalies associated with CEP135 muta- tion in the MMAF patient (Sha et al. 2017b). However, Sha et al. reported that in contrast to what was observed for other MMAF patients so far (Wambergue et al. 2016; Yang et al. 2016), the patient carrying the CEP135 mutation did not achieve any pregnancy when his sperm was used for ICSI (Sha et al. 2017b). This could be due to the essential role of the sperm centrosome in the control of the first division after egg fertilization. The identification of further MMAF genes encoding for centrosome proteins should help in establishing precise genotype–phenotype correlations and better defining the prognosis of MMAF patients for ICSI.

Intraflagellar transport (IFT) complex
In most cells, the assembly of cilia and flagella requires a molecular motor-driven process, which ensures the selective transport of proteins from the proximal to the distal part of the growing organelle. Such process is called intraflagellar transport (IFT) and conveys the multiprotein complexes of the axoneme in a bidirectional way, namely the anterograde IFT (from base to tip; IFT complex B) and the retrograde IFT (from tip to base; IFT complex A) [for review see (Ishi- kawa and Marshall 2011)]. For a long time, no clear evi- dences for IFT in mammalian sperm flagella were obtained. The characterization of the Ift88 (Tg737Rpw) mutant mouse model was pioneer in this field and indicated that similar to cilia, the IFT88 protein (also known as TTC10) is required in spermatids to assemble the flagella, but not in mature spermatozoa (San Agustin et al. 2015). Subsequently a series of other IFT proteins (i.e. IFT20, IFT25, IFT27 and IFT140) were discovered to be essential for flagellum assembly

during mouse spermiogenesis (Liu et al. 2017; Zhang et al. 2016,2017,2018) and very recent work on the genetics of MMAF phenotype confirmed the importance of IFT for human sperm flagellum assembly (Liu et al. 2019c) (Fig. 4).
TTC21A

In 2019, Liu et al. screened two distinct cohorts of MMAF individuals and identified bi-allelic loss-of-function muta- tions in TTC21A (MIM #618429), also called IFT139 (Liu et al. 2019c). Whole-exome sequencing in a cohort of 65 Han Chinese men with MMAF revealed three unrelated patients with bi-allelic mutations of TTC21A including two with homozygous stop-gain mutations and one with com- pound heterozygous mutations (Table S1). Furthermore, a homozygous TTC21A splicing mutation was identified in two Tunisian cases from an independent MMAF cohort (Table S1) (Liu et al. 2019c). Invalidation of the orthologous gene in the mouse resulted in a MMAF phenotype, confirm- ing the pathogenicity of the identified mutations. TTC21A encoded protein is highly enriched in TPR repeats (19 repeats spanning the entire protein sequence) and does not contain any other protein motif. The precise localization pat- tern of TTC21A in sperm flagella is undefined but the pro- tein was detected in human sperm proteome (Amaral et al. 2013; Wang et al. 2013) and in purified flagellar fractions from mouse spermatozoa (our unpublished data). Impor- tantly, TTC21A was connected to several established IFT proteins including IFT20 and IFT140 based on the analysis of protein functional networks (Liu et al. 2019c). Some pre- liminary experiments were performed by Liu et al. to con- firm the interaction of TTC21A with those two IFT proteins (Liu et al. 2019c) but further investigations are definitely required to confirm the exact molecular protein complexes involving TTC21A.
TTC29

In the same line, two recent publications reported the iden- tification of pathogenic mutations in TTC29 (Lorès et al. 2019; Liu et al. 2019a) (Table S1), a gene whose ortholog in Xenopus was demonstrated to be involved in the IFT B complex (Chung et al. 2014) (Table S1). TTC29 is prefer- entially expressed in the testis and encodes a protein with several TPR motifs. Similar to TTC21A, the protein was identified in proteome analyses from human spermatozoa and mouse purified sperm flagella (Lorès et al. 2019). In humans, the identified TTC29 mutations resulted in a clear MMAF phenotype, and in Leishmania, the deletion of TTC29 also induced both shortening of the flagellum and motility defects (Beneke et al. 2019). However, invalidation of the orthologous genes in the mouse (Lorès et al. 2019; Liu et al. 2019a) and in the trypanosome (Lorès et al. 2019)

yielded a near-normal flagella structure but impaired flagel- lar beating and cell progression, suggesting potential com- pensatory mechanisms in those species.
SPEF2 (IFT20)

Mutations in SPEF2 were recently reported by three dis- tinct laboratories (Liu et al. 2019b, d; Sha et al. 2019a) (Table S1). SPEF2 was first identified as a PCD gene based on the big giant head (bgh) mutant mouse model, which displays a severe ciliary phenotype and was shown to carry a SPEF2 loss of function mutation (Sironen et al. 2011), but to date in humans, no mutation in SPEF2 was reported to cause PCD. Several transcripts with different tissue distribution were described, and Liu et al. demonstrated that the SPEF2 mutations they identified in MMAF individuals only impact those that are expressed in the testis (Liu et al. 2019b).
Sironen et al. previously demonstrated that SPEF2 colo- calizes and interacts with IFT20 in the mouse testis (Sironen et al. 2010). More recently, Lehti et al. generated a male germ cell-specific Spef2 knock out mouse model and dem- onstrated that SPEF2 is required for sperm tail development; the mutant mice displaying a MMAF-like phenotype (Lehti et al. 2017). Interestingly, they also reported a duplication of the basal body, a structure required for axonemal assem- bly, and a failure in manchette migration, which results in abnormal head shape. Biochemical studies indicated that in addition to IFT20, SPEF2 also interacts with the cytoplas- mic Dynein1, strongly supporting its involvement in IFT and manchette-dependent protein transport during sper- miogenesis (Lehti et al. 2017). Taken together, the above observations are in line with the function of the manchette, which serves as platform for protein delivery during sperm tail assembly, through a process, called intra-manchette transport (IMT) (Kierszenbaum 2002). Among the MMAF genes associated with IFT, SPEF2 clearly appears as the most documented gene in this process as its function is sup- ported by comprehensive biochemical and cellular analyses. Similar studies would have to be performed for TTC21A and TTC29 to firmly demonstrate their function in the processes of sperm flagellar protein transport.
CFAP69

A few mutations were identified in CFAP69 (MIM #617959), encoding a protein of unknown function, which contains Armadillo-like helical repeats (Dong et al. 2018; He et al. 2019) (Table S1). In the mouse, Cfap69 gene invalidation was also found to recapitulate the MMAF phenotype (Dong et al. 2018; He et al. 2019), confirm- ing its implication in the processes of sperm flagellum structure and/or assembly. The precise localisation of CFAP69 within the established sperm protein complexes is

unknown but in Chlamydomonas, the ortholog was found enriched in the flagellar fractions (Pazour et al. 2005; Yang et al. 2006) and the protein was found to locate to the midpiece of human mature sperm by immunofluorescence (Dong et al. 2018).
Interestingly, proteomic analysis in mouse testis indi- cated that CFAP69 (identified with the reference Q8BH53) is part of SPEF2 immunoprecipitated protein complex, thus,suggesting a function of CFAP69 in IMT/IFT pro- cesses (Lehti et al. 2017). Consistent with this hypothesis, defects of the sperm head shape were observed in MMAF patients and in mice carrying CFAP69 pathogenic muta- tions. Study of CFAP69 localization in sperm cells’ ongo- ing spermiogenesis and in the manchette will have to be performed to confirm its involvement in the processes of protein transport.
Intriguingly, in mouse CFAP69 was also identified in cilia from olfactory sensory neurons where it was shown to regulate the odour–response kinetics with no apparent function in structure or organization of the cilia (Talaga et al. 2017). Such information suggests that in humans, ciliary dysfunction of the olfactory neurons could be associated with CFAP69 mutations; unfortunately, to date, no clinical information from the patients were obtained to state on this possible function.

Protein degradation complex
Protein degradation in eukaryotic cells is principally medi- ated by the ubiquitin–proteasome pathway (UPP), a machin- ery which comprises several hundreds of proteins working in a highly regulated manner. UPP involves the modification of the substrate protein by covalent attachment of multiple ubiquitin molecules (conjugation step) and the subsequent degradation of the tagged protein by the 26S proteasome (degradation step). UPP plays very important roles in the regulation of protein amount, activity, interaction and signal- ling in the cells and is, therefore, essential for various cel- lular functions including stress response, growth and devel- opment. Spermatogenesis is a complex process which is tightly regulated in order to produce qualitatively functional spermatozoa. In particular, aberrant gametes are eliminated throughout all duration of the process and many proteins and organelles are also degraded during spermiogenesis. Hence, in mammals, UPP was demonstrated to be required during spermiogenesis, in particular by regulating acrosome formation and flagellum assembly (Meccariello et al. 2014; Dong et al. 2016). We summarize below the recent work reporting the identification of mutations in QRICH2, which were shown to impact ubiquitination and degradation of a set of proteins required for sperm tail structure and assembly.

QRICH2

Mutations in QRICH2 were identified in a couple of MMAF infertile men (Kherraf et al. 2019; Shen et al. 2019) (MIM #618341) (Table S1) and functional analyses of the QRICH2 protein revealed an unexpected function in stabilizing the expression of a set of proteins required for sperm tail devel- opment (Shen et al. 2019). QRICH2 is specifically expressed in the testis and the protein is abundant in germ cells ongo- ing spermiogenesis. In mature sperm, the QRICH2 protein was found to co-localize with the Tubulin along the flagel- lum. Shen et al. created a Crispr-invalidated Qrich2 mouse model, which recapitulated the human MMAF phenotype and induces male infertility (Shen et al. 2019).
Proteomic analysis of the Qrich2 mutant testes revealed a total of 108 proteins differentially expressed between the mutant and control mice. Interestingly, the majority of these proteins were downregulated in the mutant testes, and among them, 33 are well-established proteins required for sperm tail structure and/or development, such as AKAP3, ODF2, TTSK4 CABYR, whose deletion in the mouse can induce sperm tail defects (Tarnasky et al. 2010; Wang et al. 2015; Young et al. 2016; Zhao et al. 2018). Importantly, QRICH2 was found to stabilize the protein amounts of AKAP3 and TTSK4 by suppressing the ubiquitination- dependent degradation of these proteins (Shen et al. 2019). In addition, Shen et al. demonstrated that QRICH2 can act as a trans-regulating factor by binding to ODF2 and CABYR promoters and enhancing their expression (Shen et al. 2019). Last, several downregulated genes identified in the Qrich2 mutant mouse model were related to energy metabolism and consistent with this, Shen et al. demonstrated the impact of heterozygous QRICH2 mutations on sperm motility parame- ters, both in mice and human with a pure asthenozoospermia without morphological abnormalities (Shen et al. 2019). All in all, QRICH2 appears as a key factor in stabilizing proteins and regulating gene transcription that are required for sperm energy, motility and flagella assembly (Fig. 4).

Undefined sperm localization or associated process
AK7

Lorès et al. reported the familial case of two MMAF siblings carrying the homozygous missense mutation (p.Leu673Pro) in AK7 (MIM #617965) which encodes an adenylate kinase expressed in ciliated cells and flagella (Lorès et al. 2018) (Table S1). Adenylate kinase (AK) catalyses the reversible transphosphorylation reaction of two molecules of ADP to one molecule each of ATP and AMP and are essential in situations of high-energy utilization and for relaying

energy to compartments that are distant from the produc- tion sites of ATP (Dzeja and Terzic 2009). While in the mouse, Ak7 invalidation was shown to induce a PCD-like phenotype with hydrocephalus, respiratory cilia defects and male infertility (Fernandez-Gonzalez et al. 2009), in human the p.Leu673Pro mutation only induced a MMAF phenotype, as revealed by normal ultra-structure and func- tion of the patient respiratory cells (Lorès et al. 2018). By performing transcript and protein analyses of biological samples from individual carrying the mutation, Lorès et al. demonstrated the absence of AK7 protein in sperm cells, but not in respiratory ciliated cells, while both cell types harboured the mutated transcript and no tissue-specific iso- forms were detected (Lorès et al. 2018). This indicated that proteins, although shared by cilia and sperm flagella. may have specific properties and/or function in each organelle. It also constitutes an additional example of functional differ- ences between human and mouse.
Several adenylate kinases were described as compo- nents of the mouse sperm flagellum; AK1 and AK2 were detected in the peri-axonemal structures (ODF and mito- chondrial sheath, respectively) (Cao et al. 2006), while AK8 was detected in the axonemal fraction (Vadnais et al. 2014). In humans, considering AK7 expression in motile cilia that are devoid of peri-axonemal structures (Milara et al. 2010), and its co-localization with tubulin in the sperm cells (Lorès et al. 2018), it is very likely that AK7 is also part of the axonemal fraction but its exact localization is so far unknown. Among mammalian AK family members, AK7 is unique in harbouring a DPY30 domain, known to be involved in the interaction with AKAP proteins (Welch et al. 2010). Considering that in Chlamydomonas reinhardtii, AK activity was reported to be tightly associated with ODAs (Milara et al. 2010), it is possible that in mammals, the DPY30 domain also provides specific targeting of AK activ- ity close to dyneins. Besides considering that AKAP4 pro- tein distribution was altered in sperm form patient carrying AK7 mutation, it is also conceivable that AK7 interacts with components of the fibrous sheath (Lorès et al. 2018). Overall in mammals and in humans, the precise localisation of AK7 in sperm flagellum remains to be determined.
ARMC2

Coutton et al. recently identified five unrelated MMAF infer- tile patients harbouring homozygous deleterious mutations in ARMC2 (MIM #618433) among a cohort of 168 patients (Coutton et al. 2019) (Table S1). They generated a Crispr mutant mouse model, which also displayed a MMAF phe- notype and male sterility, confirming the causality of the identified ARMC2 mutations in humans. The localization and precise function of ARMC2 is so far unknown. ARMC2 is a member of the ARM-repeat-containing protein family,

which all contain 42-amino-acid motifs, called ARM repeats that behave as scaffold for protein–protein interactions (Coates 2003). Arm repeats are composed of α helices (Pei- fer et al. 1994) and are present in tandem folding together to form “superhelixes” (Coates 2003).
Intriguingly, while ARMC2 transcripts are highly abun- dant in human and mouse testis and detected in human male germ cells at spermiogenesis stages (Darde et al. 2019, 2015), the protein is not reported in any dataset from sperm flagellum proteome, neither in humans (Amaral et al. 2013; Wang et al. 2013) nor in mice (Skerget et al. 2015) (and our unpublished data). This suggests that ARMC2 might fulfill transitory functions in the sperm cells and not per- sist in mature sperm. Further studies in testicular germ cells and spermatozoa may help in better defining its precise localization.

What lesson could be drawn from these genetic findings?
In the past 5 years, genetic investigations of male infertility due to MMAF resulted in the identification of 18 novel genes whose mutations account for up 30 to 60% of the cases, depending on the cohorts that were analysed. Such remark- able production was possible because of the significant advances made in the field of high-throughput sequencing technologies and mouse gene editing procedures, in particu- lar the Crispr-Cas9 technology. An important asset for this successful outcome also comes from the knowledge previ- ously established on cilio- and flagello-genesis from the study of unicellular organism models such as the flagellated parasite T. brucei and the algae Chlamydomonas. Despite regular new gene identification and this effective combined strategy, we, however, observe that about half of MMAF individuals remain with unknown genetic causes. Part of the explanation may be that some variants of unknown sig- nificance in identified MMAF genes are rarely reported and/ or accounted in published studies due to a lack of rapid and reliable techniques to validate the pathogenicity of these variations. Second, we know that WES approach cannot be expected to provide 100% positive diagnoses. This could also be explained in part by the fact that some variants are not detected by the technique used (deep intronic variants or some exonic variants due to a lack of coverage) or by the current bio-informatic pipeline used for the analysis (e.g., small duplications, complex structural variations rear- rangements). To improve this diagnostic yield and detect new causative genes, more powerful techniques such as whole genome sequencing (WGS) may now be envisaged for MMAF patients with WES negative results (Meienberg et al. 2016).

From a clinical point of view, the identification of causal mutations for the MMAF phenotype is of a clear benefit for infertile asthenozoospermic patients. In addition to improving the genetic diagnosis, this will offer the possi- bility to investigate potential genotype–phenotype correla- tions and provide ICSI prognosis for those patients. A study on infertile patients with ultra-structural abnormalities of the sperm flagellum has reported an association with poor ICSI outcome and foetal development (Fauque et al. 2009); however, this included only few MMAF cases and no infor- mation regarding the genetic aetiology of those phenotypes was available at this time. With the genetics progress made in this field, it is now possible to re-address those clinical aspects; in particular, to perform statistical analyses to pre- cisely compare the rate of fertilization and embryos gener- ated from ICSI with sperm from MMAF patients compared with that of non-MMAF patients. A limiting aspect is the number of MMAF patients carrying the same mutations or mutations in the same gene; nonetheless, such study could be performed for DNAH1 mutated patients (with a series of six and nine patients, independently analysed by two labo- ratories (Wambergue et al. 2016; Liu et al. 2019a) and indi- cated a positive ICSI outcome for those patients and most importantly, similar success rate to that obtained for other infertile patients (Wambergue et al. 2016; Liu et al. 2019a). The ICSI outcome and the associated clinical pregnancies were also analysed for a few patients carrying mutations in DNAH2, DNAH6, DNAH17, CFAP43, CFAP44, CFAP65, CFAP70, CEP135, TTC29 and SPEF 2 (Table 1). Consid-
ering that most of the mutated genes encode for axonemal
components that are not required after fertilization, one could expect a good ICSI prognosis for MMAF patients. However, a few genes have drawn our attention despite the small number of cases that were analysed. for instance, no pregnancies were reported for DNAH17 mutated patients (0/4 patients analysed) (Whitfield et al. 2019) and clini- cal pregnancies followed by abortion were reported for all CFAP65 mutated patients (3/3 patients analysed) (Li et al. 2019b). Importantly, the ICSI outcome of mutations impact- ing centrosomal proteins, which persist after fertilization and are required for embryo development, might be different. To date only one patient was identified with a mutation in a component of the centrosome (CEP135) (Sha et al. 2017b) and no pregnancy could be obtained with sperm from this patient; this would need to be further documented. Over- all, we clearly are at the beginning of a novel era, which should expand more and more, with the genetic diagnosis that can currently be provided to MMAF patients. Such will undoubtedly improve the clinical care provided to couples in the course of Assisted Reproduction Technologies and in particular MMAF patients.
From a fundamental point of view, the genetic advances
made in the field of male infertility and MMAF contribute

to improving our knowledge about the molecular mecha- nisms involved in sperm flagellum structure and beating. Amazingly, only few of the identified MMAF genes encode for proteins whose functions are already established; those are in particular the component of dynein arms required for flagellar beating. For the rest, the majority of the MMAF genes encode for proteins of unknown functions, providing a precious entry point for the discovery of novel protein networks and cellular pathways required for the building and the stabilization of the sperm flagella.
Another intriguing and fascinating aspect is the variable localizations observed for the MMAF encoded proteins, which include the axoneme, the peri-axonemal structures, the centriole and the midpiece. Obviously, such diversity emphasizes the complexity of the molecular networks and mechanisms, which sustain sperm flagellum assembly and beating. Last, although the axoneme is a common struc- ture shared between cilia and flagellum, MMAF patients with homozygous deleterious mutations in genes encoding for axonemal components (DNAH1, DNAH2, DNAH17, CFAP43, CFAP44, CFAP251/WDR66) presented only with isolated infertility without any other clinical features of PCD or other ciliopathies. These observations raise the unresolved question about the difference between flagellum and cilia and support the assumption that these axonemal MMAF-related proteins are dispensable for the structure and function of cilia in other cells. In support of this hypothesis, some of these proteins display an expression profile nearly restricted to sperm cells, thus suggesting that axonemal biogenesis/ structure of sperm flagella and cilia may require different proteins and mechanisms. Intriguingly, we noted that many of these MMAF-related genes encoding axonemal proteins (DNAH1, WDR66, CFAP43, CFAP44) are linked directly or indirectly to the radial spoke 3 (RS3). Remarkably, the morphology of the head and stem of RS3 is very different from that of the other two RS (RS1 and RS2), suggesting that there are important differences in protein composition and that each radial spoke might have specific requirements for its assembly (Pigino et al. 2011; Urbanska et al. 2015; Zhu et al. 2017). Therefore, the RS3 biogenesis and function may be one of the major differences between cilia and sperm flagella and could be the key to unravel this paradox.

Conclusion
Now is a very exciting time in the field of the genetics of infertility and in particular for male infertility due to mor- phological abnormalities of the sperm tail. The rapid pro- gress made in this field allows a better understanding of the physiopathology of MMAF, which constitutes a prerequisite for improving patient management. Besides, it clearly allows to expand our knowledge of mammalian spermatogenesis

and flagellum biogenesis, which in comparison to cilia, still remain poorly defined in a molecular point of view.
Acknowledgements We thank the Cellular Imaging Facility of Institut Cochin (INSERM U1016, CNRS UMR8104, Université Paris Descartes), in particular, Alain Schmitt, Jean-Marc Massé and Azzedine Yacia for electron microscopy procedures.

Compliance with ethical standards
Conflict of interest On behalf of all authors, the corresponding author states that there is no conflict of interest.

References
Alexandre C, Bisson JP, David G (1978) Total asthenospermia with an ultrastructural anomaly of the flagellum in two sterile brothers (author’s transl). J Gynecol Obstet Biol Reprod (Paris) 7:31–38 Amaral A, Castillo J, Estanyol JM, Ballesca JL, Ramalho-Santos J, Oliva R (2013) Human sperm tail proteome suggests new endog- enous metabolic pathways. Mol Cell Proteomics 12:330–342.

https://doi.org/10.1074/mcp.M112.020552

Amiri-Yekta A et al (2016) Whole-exome sequencing of familial cases of multiple morphological abnormalities of the sperm flagella (MMAF) reveals new DNAH1 mutations. Hum Reprod 31:2872– 2880. https://doi.org/10.1093/humrep/dew262
Auger J, Jouannet P, Eustache F (2016) Another look at human sperm morphology. Hum Reprod 31:10–23. https://doi.org/10.1093/ humrep/dev251
Auguste Y et al (2018) Loss of calmodulin- and radial-spoke-associ- ated complex protein CFAP251 leads to immotile spermatozoa lacking mitochondria and infertility in men. Am J Hum Genet 103:413–420. https://doi.org/10.1016/j.ajhg.2018.07.013
Baccetti B, Collodel G, Estenoz M, Manca D, Moretti E, Piomboni P (2005) Gene deletions in an infertile man with sperm fibrous sheath dysplasia. Hum Reprod 20:2790–2794
Barthelemy C, Tharanne MJ, Lebos C, Lecomte P, Lansac J (1990) Tail stump spermatozoa: morphogenesis of the defect. An ultra- structural study of sperm and testicular biopsy. Andrologia 22:417–425
Ben Khelifa M et al (2014) Mutations in DNAH1, which encodes an inner arm heavy chain dynein, lead to male infertility from multi- ple morphological abnormalities of the sperm flagella. Am J Hum Genet 94:95–104. https://doi.org/10.1016/j.ajhg.2013.11.017
Beneke T et al (2019) Genetic dissection of a Leishmania flagellar proteome demonstrates requirement for directional motility in sand fly infections. PLoS Pathog 15:e1007828. https://doi. org/10.1371/journal.ppat.1007828
Bettencourt-Dias M, Hildebrandt F, Pellman D, Woods G, Godinho SA (2011) Centrosomes and cilia in human disease. Trends Genet 27:307–315. https://doi.org/10.1016/j.tig.2011.05.004
Beurois J et al (2019) CFAP70 mutations lead to male infertility due to severe astheno-teratozoospermia. A case report. Hum Reprod Oxf Engl 34:2071–2079. https://doi.org/10.1093/humrep/dez166
Blue E et al (2018) Variation in cilia protein genes and progression of lung disease in cystic fibrosis. Ann Am Thorac Soc 15:440–448. https://doi.org/10.1513/AnnalsATS.201706-451OC
Brown PR, Miki K, Harper DB, Eddy EM (2003) A-kinase anchoring protein 4 binding proteins in the fibrous sheath of the sperm flagellum. Biol Reprod 68:2241–2248. https://doi.org/10.1095/ biolreprod.102.013466

Cao W, Haig-Ladewig L, Gerton GL, Moss SB (2006) Adenylate kinases 1 and 2 are part of the accessory structures in the mouse sperm flagellum. Biol Reprod 75:492–500. https://doi. org/10.1095/biolreprod.106.053512
Carrera A, Gerton GL, Moss SB (1994) The major fibrous sheath poly- peptide of mouse sperm: structural and functional similarities to the A-kinase anchoring proteins. Dev Biol 165:272–284. https
://doi.org/10.1006/dbio.1994.1252
Chemes H (2012) Sperm centrioles and their dual role in flagello- genesis and cell cycle of the zygote, pp 33–48. https://doi. org/10.1007/978-1-62703-035-9_2
Chemes HE, Brugo S, Zanchetti F, Carrere C, Lavieri JC (1987) Dys- plasia of the fibrous sheath: an ultrastructural defect of human spermatozoa associated with sperm immotility and primary ste- rility. Fertil Steril 48:664–669
Chung MI et al (2014) Coordinated genomic control of ciliogen- esis and cell movement by RFX2. eLife 3:e01439. https://doi. org/10.7554/eLife.01439
Coates JC (2003) Armadillo repeat proteins: beyond the animal king- dom. Trends Cell Biol 13:463–471
Cooper TG et al (2010) World Health Organization reference values for human semen characteristics. Hum Reprod Update 16:231–245. https://doi.org/10.1093/humupd/dmp048
Coutton C et al (2015) Teratozoospermia: spotlight on the main genetic actors in the human. Hum Reprod Update 21:455–485. https:// doi.org/10.1093/humupd/dmv020
Coutton C et al (2019) Bi-allelic mutations in ARMC2 Lead to severe astheno-teratozoospermia due to sperm flagellum malformations in humans and mice. Am J Hum Genet 104:331–340. https://doi. org/10.1016/j.ajhg.2018.12.013
Coutton C et al (2018) Mutations in CFAP43 and CFAP44 cause male infertility and flagellum defects in Trypanosoma and human. Nat Commun 9:686. https://doi.org/10.1038/s41467-017-02792-7
Curi SM et al (2003) Asthenozoospermia: analysis of a large popula- tion. Arch Androl 49:343–349
Darde TA et al (2019) The ReproGenomics Viewer: a multi-omics and cross-species resource compatible with single-cell studies for the reproductive science community. Bioinformatics. https:// doi.org/10.1093/bioinformatics/btz047
Darde TA et al (2015) The ReproGenomics Viewer: an integrative cross-species toolbox for the reproductive science community. Nucleic Acids Res 43:W109–W116. https://doi.org/10.1093/nar/ gkv345
Dong FN et al (2018) Absence of CFAP69 causes male infertility due to multiple morphological abnormalities of the flagella in human and mouse. Am J Hum Genet 102:636–648. https://doi. org/10.1016/j.ajhg.2018.03.007
Dong L et al (2016) The role of ubiquitin-proteasome pathway in spermatogenesis. Yi Chuan Hered 38:791–800. https://doi. org/10.16288/j.yczz.16-120
Dymek EE, Heuser T, Nicastro D, Smith EF (2011) The CSC is required for complete radial spoke assembly and wild-type ciliary motility. Mol Biol Cell 22:2520–2531. https://doi.org/10.1091/ mbc.E11-03-0271
Dymek EE, Smith EF (2007) A conserved CaM- and radial spoke asso- ciated complex mediates regulation of flagellar dynein activity. J Cell Biol 179:515–526. https://doi.org/10.1083/jcb.200703107
Dzeja P, Terzic A (2009) Adenylate kinase and AMP signaling net- works: metabolic monitoring, signal communication and body energy sensing. Int J Mol Sci 10:1729–1772. https://doi. org/10.3390/ijms10041729
Eddy EM (2007) The scaffold role of the fibrous sheath. Soc Reprod Fertil Suppl 65:45–62
Eddy EM, Toshimori K, O’Brien DA (2003) Fibrous sheath of mam- malian spermatozoa. Microsc Res Tech 61:103–115. https://doi. org/10.1002/jemt.10320

Escalier D (2006) Arrest of flagellum morphogenesis with fibrous sheath immaturity of human spermatozoa. Andrologia 38:54–60
Escalier D, Albert M (2006) New fibrous sheath anomaly in sperma- tozoa of men with consanguinity. Fertil Steril 86(219):e211–219 Escalier D, Toure A (2012) Morphological defects of sperm flagellum implicated in human male infertility. Med Sci 28:503–511. https
://doi.org/10.1051/medsci/2012285015
Fauque P et al (2009) From ultrastructural flagellar sperm defects to the health of babies conceived by ICSI. Reprod Biomed Online 19:326–336
Fernandez-Gonzalez A, Kourembanas S, Wyatt TA, Mitsialis SA (2009) Mutation of murine adenylate kinase 7 underlies a pri- mary ciliary dyskinesia phenotype. Am J Respir Cell Mol Biol 40:305–313. https://doi.org/10.1165/rcmb.2008-0102OC
Fu G et al (2018) The I1 dynein-associated tether and tether head com- plex is a conserved regulator of ciliary motility. Mol Biol Cell 29:1048–1059. https://doi.org/10.1091/mbc.E18-02-0142
Gershoni M et al (2017) A familial study of azoospermic men identifies three novel causative mutations in three new human azoospermia genes. Genet Med Off J Am Coll Med Genet 19:998–1006. https
://doi.org/10.1038/gim.2016.225
He X et al (2019) Novel homozygous CFAP69 mutations in humans and mice cause severe asthenoteratospermia with multiple mor- phological abnormalities of the sperm flagella. J Med Genet 56:96–103. https://doi.org/10.1136/jmedgenet-2018-105486
Heuser T, Barber CF, Lin J, Krell J, Rebesco M, Porter ME, Nicastro D (2012a) Cryoelectron tomography reveals doublet-specific struc- tures and unique interactions in the I1 dynein. Proc Natl Acad Sci USA 109:E2067–2076. https://doi.org/10.1073/pnas.1120690109
Heuser T, Dymek EE, Lin J, Smith EF, Nicastro D (2012b) The CSC connects three major axonemal complexes involved in dynein regulation. Mol Biol Cell 23:3143–3155. https://doi.org/10.1091/ mbc.E12-05-0357
Hiraki M, Nakazawa Y, Kamiya R, Hirono M (2007) Bld10p con- stitutes the cartwheel-spoke tip and stabilizes the 9-fold sym- metry of the centriole. Curr Biol 17:1778–1783. https://doi. org/10.1016/j.cub.2007.09.021
Hom EF et al (2011) A unified taxonomy for ciliary dyneins. Cytoskel- eton 68:555–565. https://doi.org/10.1002/cm.20533
Horani A, Ferkol TW, Dutcher SK, Brody SL (2016) Genetics and biol- ogy of primary ciliary dyskinesia. Paediatr Respir Rev 18:18–24. https://doi.org/10.1016/j.prrv.2015.09.001
Hu J et al (2019) ENU-induced mutant allele of Dnah1, ferf1, causes abnormal sperm behavior and fertilization failure in mice. Mol Reprod Dev 86:416–425. https://doi.org/10.1002/mrd.23120
Hussain MS et al (2012) A truncating mutation of CEP135 causes primary microcephaly and disturbed centrosomal function. Am J Hum Genet 90:871–878. https://doi.org/10.1016/j. ajhg.2012.03.016
Imtiaz F, Allam R, Ramzan K, Al-Sayed M (2015) Variation in DNAH1 may contribute to primary ciliary dyskinesia. BMC Med Genet 16:14. https://doi.org/10.1186/s12881-015-0162-5
Inaba K (2007) Molecular basis of sperm flagellar axonemes: struc- tural and evolutionary aspects. Ann N Y Acad Sci 1101:506–526. https://doi.org/10.1196/annals.1389.017
Inaba K (2011) Sperm flagella: comparative and phylogenetic perspec- tives of protein components. Mol Hum Reprod 17:524–538. https
://doi.org/10.1093/molehr/gar034
Ishikawa H, Marshall WF (2011) Ciliogenesis: building the cell’s antenna. Nat Rev Mol Cell Biol 12:222–234. https://doi. org/10.1038/nrm3085
Kherraf ZE et al (2018) A homozygous ancestral SVA-insertion-medi- ated deletion in WDR66 induces multiple morphological abnor- malities of the sperm flagellum and male infertility. Am J Hum Genet 103:400–412. https://doi.org/10.1016/j.ajhg.2018.07.014

Kherraf ZE et al (2019) Whole exome sequencing of men with mul- tiple morphological abnormalities of the sperm flagella reveals novel homozygous QRICH2 mutations. Clin Genet 5:5. https:// doi.org/10.1111/cge.13604
Kierszenbaum AL (2002) Intramanchette transport (IMT): managing the making of the spermatid head, centrosome, and tail. Mol Reprod Dev 63:1–4. https://doi.org/10.1002/mrd.10179
King SM (2012) Integrated control of axonemal dynein AAA(+) motors. J Struct Biol 179:222–228. https://doi.org/10.1016/j. jsb.2012.02.013
Kleylein-Sohn J, Westendorf J, Le Clech M, Habedanck R, Stierhof YD, Nigg EA (2007) Plk4-induced centriole biogenesis in human cells. Dev Cell 13:190–202. https://doi.org/10.1016/j.devce l.2007.07.002
Knowles MR, Zariwala M, Leigh M (2016) Primary ciliary dyski- nesia. Clin Chest Med 37:449–461. https://doi.org/10.1016/j. ccm.2016.04.008
Kobayashi D, Takeda H (2012) Ciliary motility: the components and cytoplasmic preassembly mechanisms of the axonemal dyneins. Differentiation 83:S23–29. https://doi.org/10.1016/j. diff.2011.11.009
Kraatz S et al (2016) The human centriolar protein CEP135 contains a two-stranded coiled-coil domain critical for microtubule binding. Structure 24:1358–1371. https://doi.org/10.1016/j. str.2016.06.011
Kubo T, Hou Y, Cochran DA, Witman GB, Oda T (2018) A micro- tubule-dynein tethering complex regulates the axonemal inner dynein f (I1). Mol Biol Cell 29:1060–1074. https://doi. org/10.1091/mbc.E17-11-0689
Kurkowiak M, Zietkiewicz E, Witt M (2015) Recent advances in pri- mary ciliary dyskinesia genetics. J Med Genet 52:1–9. https:// doi.org/10.1136/jmedgenet-2014-102755
Lehti MS, Sironen A (2017) Formation and function of sperm tail structures in association with sperm motility defects. Biol Reprod 97:522–536. https://doi.org/10.1093/biolre/iox096
Lehti MS, Zhang FP, Kotaja N, Sironen A (2017) SPEF2 functions in microtubule-mediated transport in elongating spermatids to ensure proper male germ cell differentiation. Development 144:2683–2693. https://doi.org/10.1242/dev.152108
Li Y et al (2016) DNAH6 and its interactions with PCD genes in heter- otaxy and primary ciliary dyskinesia. PLoS Genet 12:e1005821. https://doi.org/10.1371/journal.pgen.1005821
Li L et al (2018) DNAH6 is a novel candidate gene associated with sperm head anomaly. Andrologia. https://doi.org/10.1111/ and.12953
Li W et al (2019a) Bi-allelic mutations of CFAP251 cause sperm fla- gellar defects and human male infertility. J Hum Genet. https:// doi.org/10.1038/s10038-018-0520-1
Li W et al (2019b) Bi-allelic mutations in CFAP65 cause male infertil- ity with multiple morphological abnormalities of the sperm fla- gella in humans and mice. J Med Genet. https://doi.org/10.1136/ jmedgenet-2019-106344
Li Y et al (2019c) DNAH2 is a novel candidate gene associated with multiple morphological abnormalities of the sperm flagella. Clin Genet 95:590–600. https://doi.org/10.1111/cge.13525
Linck RW, Chemes H, Albertini DF (2016) The axoneme: the propul- sive engine of spermatozoa and cilia and associated ciliopathies leading to infertility. J Assist Reprod Genet 33:141–156. https:// doi.org/10.1007/s10815-016-0652-1
Liu C et al (2019a) Bi-allelic mutations in TTC29 cause male subfertil- ity with asthenoteratospermia in humans and mice. Am J Hum Genet. https://doi.org/10.1016/j.ajhg.2019.10.010
Liu C et al (2019b) Homozygous mutations in SPEF2 induce multi- ple morphological abnormalities of the sperm flagella and male infertility. J Med Genet. https://doi.org/10.1136/jmedgenet-2019- 106011

Liu H et al (2017) IFT25, an intraflagellar transporter protein dispen- sable for ciliogenesis in somatic cells, is essential for sperm flagella formation. Biol Reprod 96:993–1006. https://doi. org/10.1093/biolre/iox029
Liu W et al (2019a) Bi-allelic mutations in TTC21A induce asthe- noteratospermia in humans and mice. Am J Hum Genet 104:738–748. https://doi.org/10.1016/j.ajhg.2019.02.020
Liu W et al (2019b) Loss-of-function mutations in SPEF2 cause multiple morphological abnormalities of the sperm flagella (MMAF). J Med Genet 56:678–684. https://doi.org/10.1136/ jmedgenet-2018-105952
Liu W et al (2019c) Homozygous loss-of-function mutations in FSIP2 cause male infertility with asthenoteratospermia. J Genet Genomics Yi Chuan Xue Bao 46:53–56. https://doi. org/10.1016/j.jgg.2018.09.006
Lorès P et al (2018) Homozygous missense mutation L673P in ade- nylate kinase 7 (AK7) leads to primary male infertility and multiple morphological anomalies of the flagella but not to primary ciliary dyskinesia. Hum Mol Genet 27:1196–1211. https://doi.org/10.1093/hmg/ddy034
Lorès P et al (2019) Mutations in TTC29, encoding an evolutionarily conserved axonemal protein, result in asthenozoospermia and male infertility. Am J Hum Genet 1:1. https://doi.org/10.1016/j. ajhg.2019.10.007
Martinez G et al (2018) Whole-exome sequencing identifies muta- tions in FSIP2 as a recurrent cause of multiple morphological abnormalities of the sperm flagella. Hum Reprod 33:1973– 1984. https://doi.org/10.1093/humrep/dey264
Meccariello R et al (2014) Molecular chaperones, cochaperones, and ubiquitination/deubiquitination system: involvement in the production of high quality spermatozoa. BioMed Res Int 2014:561426. https://doi.org/10.1155/2014/561426
Meienberg J, Bruggmann R, Oexle K, Matyas G (2016) Clinical sequencing: is WGS the better WES? Hum Genet 135:359– 362. https://doi.org/10.1007/s00439-015-1631-9
Miki K, Willis WD, Brown PR, Goulding EH, Fulcher KD, Eddy EM (2002) Targeted disruption of the Akap4 gene causes defects in sperm flagellum and motility. Dev Biol 248:331–342
Milara J, Armengot M, Mata M, Morcillo EJ, Cortijo J (2010) Role of adenylate kinase type 7 expression on cilia motility: pos- sible link in primary ciliary dyskinesia. Am J Rhinol Allergy 24:181–185. https://doi.org/10.2500/ajra.2010.24.3468
Morimoto Y et al (2019) Nonsense mutation in CFAP43 causes nor- mal-pressure hydrocephalus with ciliary abnormalities. Neu- rology 92:e2364–e2374. https://doi.org/10.1212/WNL.00000 00000007505
Mottier-Pavie V, Megraw TL (2009) Drosophila bld10 is a centri- olar protein that regulates centriole, basal body, and motile cilium assembly. Mol Biol Cell 20:2605–2614. https://doi. org/10.1091/mbc.E08-11-1115
Myster SH, Knott JA, O’Toole E, Porter ME (1997) The Chla- mydomonas Dhc1 gene encodes a dynein heavy chain subunit required for assembly of the I1 inner arm complex. Mol Biol Cell 8:607–620. https://doi.org/10.1091/mbc.8.4.607
Myster SH, Knott JA, Wysocki KM, O’Toole E, Porter ME (1999) Domains in the 1alpha dynein heavy chain required for inner arm assembly and flagellar motility in Chlamydomonas. J Cell Biol 146:801–818
Neesen J et al (2001) Disruption of an inner arm dynein heavy chain gene results in asthenozoospermia and reduced ciliary beat frequency. Hum Mol Genet 10:1117–1128
Nicastro D, Schwartz C, Pierson J, Gaudette R, Porter ME, McIntosh JR (2006) The molecular architecture of axonemes revealed by cryoelectron tomography. Science 313:944–948. https://doi. org/10.1126/science.1128618

Norling A et al (2014) Identification of a duplication within the GDF9 gene and novel candidate genes for primary ovarian insuffi- ciency (POI) by a customized high-resolution array compara- tive genomic hybridization platform. Hum Reprod Oxf Engl 29:1818–1827. https://doi.org/10.1093/humrep/deu149
Nsota Mbango J-F, Coutton C, Arnoult C, Ray P, Toure A (2019) Genetic causes of male infertility: snapshot on morphological abnormalities of the sperm flagellum. Basic Clin Androl 29:2. https://doi.org/10.1186/s12610-019-0083-9 (eCollection 2019) Ohmori K, Matsuda T, Horii Y, Yoshida O (1993) Three cases with different types of short-tailed spermatozoa. Urol Int 50:174–178.

https://doi.org/10.1159/000282478

Ohta T, Essner R, Ryu JH, Palazzo RE, Uetake Y, Kuriyama R (2002) Characterization of Cep135, a novel coiled-coil centrosomal pro- tein involved in microtubule organization in mammalian cells. J Cell Biol 156:87–99. https://doi.org/10.1083/jcb.200108088
Pazour GJ, Agrin N, Leszyk J, Witman GB (2005) Proteomic analy- sis of a eukaryotic cilium. J Cell Biol 170:103–113. https://doi. org/10.1083/jcb.200504008
Peifer M, Berg S, Reynolds AB (1994) A repeating amino acid motif shared by proteins with diverse cellular roles. Cell 76:789–791. https://doi.org/10.1016/0092-8674(94)90353-0
Pigino G, Ishikawa T (2012) Axonemal radial spokes: 3D structure, function and assembly. Bioarchitecture 2:50–58
Pigino G et al (2011) Cryoelectron tomography of radial spokes in cilia and flagella. J Cell Biol 195:673–687. https://doi.org/10.1083/ jcb.201106125
Ross A, Christie S, Edmond P (1973) Ultrastructural tail defects in the spermatozoa from two men attending a subfertility clinic. J Reprod Fertil 32:243–251. https://doi.org/10.1530/jrf.0.0320243
San Agustin JT, Pazour GJ, Witman GB (2015) Intraflagellar trans- port is essential for mammalian spermiogenesis but is absent in mature sperm. Mol Biol Cell 26:4358–4372. https://doi. org/10.1091/mbc.E15-08-0578
Schatten H, Sun QY (2009) The role of centrosomes in mammalian fertilization and its significance for ICSI. Mol Hum Reprod 15:531–538. https://doi.org/10.1093/molehr/gap049
Sha Y et al (2019) Biallelic mutations in Sperm flagellum 2 cause human multiple morphological abnormalities of the sperm fla- gella (MMAF) phenotype. Clin Genet 96:385–393. https://doi. org/10.1111/cge.13602
Sha Y et al (2017) DNAH1 gene mutations and their potential associa- tion with dysplasia of the sperm fibrous sheath and infertility in the Han Chinese population. Fertil Steril 107:1312–1318. https
://doi.org/10.1016/j.fertnstert.2017.04.007
Sha YW et al (2019a) Patients with multiple morphological abnormali- ties of the sperm flagella harbouring CFAP44 or CFAP43 muta- tions have a good pregnancy outcome following intracytoplasmic sperm injection. Andrologia 51:e13151. https://doi.org/10.1111/ and.13151
Sha YW et al (2019b) Novel mutations in CFAP44 and CFAP43 cause multiple morphological abnormalities of the sperm flagella (MMAF). Reprod Sci Thousand Oaks Calif 26:26–34. https:// doi.org/10.1177/1933719117749756
Sha YW, Xu X, Mei LB, Li P, Su ZY, He XQ, Li L (2017) A homozy- gous CEP135 mutation is associated with multiple morphologi- cal abnormalities of the sperm flagella (MMAF). Gene 633:48– 53. https://doi.org/10.1016/j.gene.2017.08.033
Shamoto N, Narita K, Kubo T, Oda T, Takeda S (2018) CFAP70 is a novel axoneme-binding protein that localizes at the base of the outer dynein arm and regulates ciliary motility. Cells. https://doi. org/10.3390/cells7090124
Shen Y et al (2019) Loss-of-function mutations in QRICH2 cause male infertility with multiple morphological abnormalities of the sperm flagella. Nat Commun 10:433. https://doi.org/10.1038/ s41467-018-08182-x

Sironen A, Hansen J, Thomsen B, Andersson M, Vilkki J, Toppari J, Kotaja N (2010) Expression of SPEF2 during mouse spermato- genesis and identification of IFT20 as an interacting protein. Biol Reprod 82:580–590. https://doi.org/10.1095/biolreprod.108.07497
1
Sironen A et al (2011) Loss of SPEF2 function in mice results in sper- matogenesis defects and primary ciliary dyskinesia. Biol Reprod 85:690–701. https://doi.org/10.1095/biolreprod.111.091132
Skerget S, Rosenow MA, Petritis K, Karr TL (2015) Sperm proteome maturation in the mouse epididymis. PLoS ONE 10:e0140650. https://doi.org/10.1371/journal.pone.0140650
Smith TF (2008) Diversity of WD-repeat proteins. Subcell Biochem 48:20–30. https://doi.org/10.1007/978-0-387-09595-0_3
Talaga AK, Dong FN, Reisert J, Zhao H (2017) Cilia- and flagella- associated protein 69 regulates olfactory transduction kinetics in mice. J Neurosci 37:5699–5710. https://doi.org/10.1523/JNEUR OSCI.0392-17.2017
Tang S et al (2017) Bi-allelic mutations in CFAP43 and CFAP44 Cause male infertility with multiple morphological abnormalities of the sperm flagella. Am J Hum Genet 100:854–864. https://doi. org/10.1016/j.ajhg.2017.04.012
Tarnasky H, Cheng M, Ou Y, Thundathil JC, Oko R, van der Hoorn FA (2010) Gene trap mutation of murine outer dense fiber pro- tein-2 gene can result in sperm tail abnormalities in mice with high percentage chimaerism. BMC Dev Biol 10:67. https://doi. org/10.1186/1471-213X-10-67
Terquem A, Dadoune JP (1980) Structural anomalies in disorders human of sperm motility in 60 cases of asthenospermia. Bull Assoc Anat (Nancy) 64:567–576
Toure A, Rode B, Hunnicutt GR, Escalier D, Gacon G (2011) Septins at the annulus of mammalian sperm. Biol Chem 392:799–803. https
://doi.org/10.1515/BC.2011.074
Tu C, Nie H, Meng L et al (2019) Identification of DNAH6 mutations in infertile men with multiple morphological abnormalities of the sperm flagella. Sci Rep 9:15864. https://doi.org/10.1038/s4159 8-019-52436-7
Urbanska P et al (2018) Ciliary proteins Fap43 and Fap44 interact with each other and are essential for proper cilia and flagella beating. CMLS 75:4479–4493. https://doi.org/10.1007/s00018-018-2819-7
Urbanska P et al (2015) The CSC proteins FAP61 and FAP251 build the basal substructures of radial spoke 3 in cilia. Mol Biol Cell 26:1463–1475. https://doi.org/10.1091/mbc.E14-11-1545
Vadnais ML, Cao W, Aghajanian HK, Haig-Ladewig L, Lin AM, Al-Alao O, Gerton GL (2014) Adenine nucleotide metabolism and a role for AMP in modulating flagellar waveforms in mouse sperm. Biol Reprod. https://doi.org/10.1095/biolreprod.113.114447
van Dam TJP et al (2019) CiliaCarta: an integrated and validated com- pendium of ciliary genes. PLoS ONE 14:e0216705. https://doi. org/10.1371/journal.pone.0216705
Vincensini L, Blisnick T, Bastin P (2011) 1001 model organisms to study cilia and flagella. Biol Cell 103:109–130. https://doi.org/10.1042/ BC20100104
Viswanadha R, Sale WS, Porter ME (2017) Ciliary motility: regulation of axonemal dynein motors. Cold Spring Harb Perspect Biol. https
://doi.org/10.1101/cshperspect.a018325
Wambergue C et al (2016) Patients with multiple morphological abnor- malities of the sperm flagella due to DNAH1 mutations have a good prognosis following intracytoplasmic sperm injection. Hum Reprod 31:1164–1172. https://doi.org/10.1093/humrep/dew083
Wang G et al (2013) In-depth proteomic analysis of the human sperm reveals complex protein compositions. J Proteomics 79:114–122. https://doi.org/10.1016/j.jprot.2012.12.008
Wang W et al (2019) Bi-allelic mutations in CFAP65 lead to severe asthenoteratospermia due to acrosome hypoplasia and flagellum malformations. J Med Genet. https://doi.org/10.1136/jmedgenet- 2019-106031

Wang X et al (2017) Homozygous DNAH1 frameshift mutation causes multiple morphological anomalies of the sperm flagella in Chinese. Clin Genet 91:313–321. https://doi.org/10.1111/cge.12857
Wang X et al (2015) Tssk4 is essential for maintaining the structural integrity of sperm flagellum. Mol Hum Reprod 21:136–145. https
://doi.org/10.1093/molehr/gau097
Welch EJ, Jones BW, Scott JD (2010) Networking with AKAPs: context- dependent regulation of anchored enzymes. Mol Interv 10:86–97. https://doi.org/10.1124/mi.10.2.6
Whitfield M et al (2019) Mutations in DNAH17, Encoding a sperm-spe- cific axonemal outer dynein arm heavy chain, cause isolated male infertility due to asthenozoospermia. Am J Hum Genet 105:198– 212. https://doi.org/10.1016/j.ajhg.2019.04.015
Wu H et al (2019) NovelCFAP43 andCFAP44 mutations cause male infertility with multiple morphological abnormalities of the sperm flagella (MMAF). Reprod Biomed Online 38:769–778. https://doi. org/10.1016/j.rbmo.2018.12.037
Xu Y, Cao J, Huang S, Feng D, Zhang W, Zhu X, Yan X (2015) Char- acterization of tetratricopeptide repeat-containing proteins critical for cilia formation and function. PLoS ONE 10:e0124378. https:// doi.org/10.1371/journal.pone.0124378
Yang P et al (2006) Radial spoke proteins of Chlamydomonas flagella. J Cell Sci 119:1165–1174. https://doi.org/10.1242/jcs.02811
Yang SM, Yang XY, Ding Y, Li H, Wang W, Liu JY, Wen DG (2016) Intracytoplasmic sperm injection outcomes in Chinese men with multiple morphological abnormalities of sperm flagella. Asian J Androl 18:809–811. https://doi.org/10.4103/1008-682X.167722
Young SA, Miyata H, Satouh Y, Aitken RJ, Baker MA, Ikawa M (2016) CABYR is essential for fibrous sheath integrity and progressive motility in mouse spermatozoa. J Cell Sci 129:4379–4387. https:// doi.org/10.1242/jcs.193151
Zeytuni N, Zarivach R (2012) Structural and functional discussion of the tetra-trico-peptide repeat, a protein interaction module. Structure 20:397–405. https://doi.org/10.1016/j.str.2012.01.006

Zhang B et al (2019) A DNAH17 missense variant causes flagella destabilization and asthenozoospermia. J Exp Med. https://doi. org/10.1084/jem.20182365
Zhang X, Shen Y, Wang X et al (2019) A novel homozygous CFAP65 mutation in humans causes male infertility with multiple morpho- logical abnormalities of the sperm flagella. Clin Genet 96:541–548. https://doi.org/10.1111/cge.13644
Zhang Y et al (2019c) Vertebrate Dynein-f depends on Wdr78 for axone- mal localization and is essential for ciliary beat J Mol Cell Biol 11:383–394. https://doi.org/10.1093/jmcb/mjy043
Zhang Y et al (2017) Intraflagellar transporter protein (IFT27), an IFT25 binding partner, is essential for male fertility and spermiogenesis in mice Dev Biol 432:125–139. https://doi.org/10.1016/j.ydbio
.2017.09.023
Zhang Y et al (2018) Intraflagellar transporter protein 140 (IFT140), a component of IFT-A complex, is essential for male fertility and spermiogenesis in mice. Cytoskeleton 75:70–84. https://doi. org/10.1002/cm.21427
Zhang Z et al (2016) Intraflagellar transport protein IFT20 is essential for male fertility and spermiogenesis in mice. Mol Biol Cell 1:1. https
://doi.org/10.1091/mbc.E16-05-0318
Zhao W, Li Z, Ping P, Wang G, Yuan X, Sun F (2018) Outer dense fibers stabilize the axoneme to maintain sperm motility. J Cell Mol Med 22:1755–1768. https://doi.org/10.1111/jcmm.13457
Zhu X, Liu Y, Yang P (2017) Radial Spokes—a snapshot of the motil- ity regulation, assembly, and evolution of Cilia and Flagella. Cold Spring Harb Perspect Biol 9:a028126. https://doi.org/10.1101/ cshperspect.a028126
Publisher’s Note Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Leave a Reply

Your email address will not be published. Required fields are marked *

*

You may use these HTML tags and attributes: <a href="" title=""> <abbr title=""> <acronym title=""> <b> <blockquote cite=""> <cite> <code> <del datetime=""> <em> <i> <q cite=""> <strike> <strong>