CatSper Channels

You might also like

Download as pdf or txt
Download as pdf or txt
You are on page 1of 13

See discussions, stats, and author profiles for this publication at: https://www.researchgate.

net/publication/343186718

CatSper: The complex main gate of calcium entry in mammalian spermatozoa

Article in Molecular and Cellular Endocrinology · July 2020


DOI: 10.1016/j.mce.2020.110951

CITATIONS READS

5 273

2 authors, including:

Rita Rahban
University of Geneva
12 PUBLICATIONS 84 CITATIONS

SEE PROFILE

Some of the authors of this publication are also working on these related projects:

prenatal exposure to DEHP View project

All content following this page was uploaded by Rita Rahban on 29 September 2020.

The user has requested enhancement of the downloaded file.


Molecular and Cellular Endocrinology xxx (xxxx) xxx

Contents lists available at ScienceDirect

Molecular and Cellular Endocrinology


journal homepage: www.elsevier.com/locate/mce

Review

CatSper: The complex main gate of calcium entry in


mammalian spermatozoa
Rita Rahban a, b, *, Serge Nef a, b, *
a
Swiss Centre for Applied Human Toxicology (SCAHT), Switzerland
b
Department of Genetic Medicine and Development, Faculty of Medicine, University of Geneva, Rue Michel-Servet 1, 1206, Geneva, Switzerland

A R T I C L E I N F O A B S T R A C T

Keywords: Calcium ions (Ca2+) are involved in nearly every aspect of cellular life. They are one of the most abundant el­
CatSper ements in mammals and play a vital role in physiological and biochemical processes acting mainly as intracel­
Calcium signaling lular messengers. In spermatozoa, several key functions are regulated by cytoplasmic Ca2+ concentration such as
Mammalian spermatozoa
sperm capacitation, chemotaxis, hyperactive motility, and acrosome reaction. The sperm-specific ion channel
Sperm ion channels
CatSper is the principal calcium channel in sperm mediating the calcium influx into the sperm flagellum and
Fertilization
acting as an essential modulator of downstream mechanisms involved in fertilization. This review aims to pro­
vide insights into the structure, localization, and function of the mammalian CatSper channel, primarily human
and mice. The activation of CatSper by progesterone and prostaglandins, as well as the ligand-independent
regulation of the channel by a change in the membrane voltage and intracellular pH are going to be
addressed. Finally, major questions, challenges, and perspectives are discussed.

1. Introduction through diverse ion channels (Gupta, 2017). Despite being motile upon
ejaculation, sperms are unable to fertilize the oocyte unless they un­
The survival of the animal species relies almost entirely on the ability dergo a “priming” step in the female reproductive tract referred to as
of a spermatozoon to fertilize an egg. The male germ cell is a unique capacitation (Austin, 1952; Chang, 1951). During this process which is
highly specialized cell that is designed to exert its main function outside partly dependent on an increase in intracellular calcium concentration
of its production site. After being released in the lumen of the seminif­ ([Ca2+]i), extensive metabolic and physiological changes occur in all
erous tubules, sperm cells have to accomplish several challenging tasks sperm compartments. These changes involve further membrane
before reaching and fertilizing the egg. The journey begins after sper­ remodeling which includes the redistribution, modification, addition, or
matogenesis has been completed culminating in the formation of highly removal of proteins acquired during epididymal transit. The female
differentiated spermatozoa that are structurally complete but are still reproductive tract is indeed rich in albumin that binds to cholesterol and
functionally immature. It is through their transit in the epididymis that induces an important cholesterol efflux leading to an increase in the
sperms cells complete important maturation steps crucial for fulfilling membrane fluidity. In addition, exposure of sperms to high levels of
their functions. These include sperm chromatin condensation, surface bicarbonate present in the female tract induces activation of soluble
protein modifications such as the incorporation of cholesterol, adenylate cyclase leading to the production of cyclic adenosine mono­
morphological changes, and most importantly sperm motility initiation phosphate (cAMP) and activation of the protein kinase A (PKA) (Gervasi
and acquisition of coordinated sperm motion (reviewed in Sullivan and and Visconti, 2017). The phosphorylation of tyrosine residues by PKA, in
Mieusset (2016)). Membrane remodeling and active glycosylation of turn, induces membrane hyperpolarization which later activates diverse
surface components are believed to stabilize the cell in order to remain calcium channels, key among them are the Cationic channel of Sperm
in a quiescent state (Cooper, 2007; Sullivan et al., 2019; Sullivan and CatSper and voltage-gated calcium channels Cav leading to an increase
Mieusset, 2016; Yeung et al., 1993). These changes are already at this in [Ca2+]i (Darszon et al., 2011; De Jonge, 2017, 2005; Puga Molina
stage, highly dependent on the exchange of ions and electrolytes et al., 2018; Visconti et al., 2011). This increase allows the sperm to

* Corresponding authors. Department of Genetic Medicine and Development, Faculty of Medicine, University of Geneva, Rue Michel-Servet 1, 1206, Geneva,
Switzerland.
E-mail addresses: Rita.Rahban@unige.ch (R. Rahban), serge.nef@unige.ch (S. Nef).

https://doi.org/10.1016/j.mce.2020.110951
Received 12 April 2020; Received in revised form 17 July 2020; Accepted 20 July 2020
Available online 24 July 2020
0303-7207/© 2020 The Authors. Published by Elsevier B.V. This is an open access article under the CC BY license (http://creativecommons.org/licenses/by/4.0/).

Please cite this article as: Rita Rahban, Serge Nef, Molecular and Cellular Endocrinology, https://doi.org/10.1016/j.mce.2020.110951
R. Rahban and S. Nef Molecular and Cellular Endocrinology xxx (xxxx) xxx

acquire hyperactive motility, a landmark of sperm capacitation, and an revealed that CatSper is the principal Ca2+ channel in mouse sperm
absolute prerequisite for fertilization. Hyperactivated sperms are char­ (Kirichok et al., 2006; Lobley et al., 2003; Qi et al., 2007; Quill et al.,
acterized by having a high amplitude whip-like asymmetrical tail 2003; Ren et al., 2001). CatSper is specifically located in the membrane
bending, are non-progressive in aqueous low-viscosity media, and are of the flagellar principal piece and is known to be pH-sensitive and
thought to be progressive in the physiological viscosity of the upper weakly voltage-dependent (Lishko et al., 2012). In the absence of
female reproductive tract (de Lamirande et al., 1997; Kirkman-Brown extracellular calcium, CatSper also allows the entry of monovalent cat­
and Smith, 2011; Mortimer, 1997). Another major consequence of ions such as Na+ and Cs+, as well as divalent cations Ba2+ with a much
capacitation is the sperm ability to undergo the acrosome reaction (AR) lower affinity compared to Ca2+ (Sun et al., 2017). The heteromeric
as a result of the acquired increase of membrane fluidity. The AR is CatSper channel is composed of at least ten subunits with four
characterized by the release of proteolytic enzymes allowing the sperm pore-forming α subunits (CatSper 1–4) and six auxiliary subunits (β, γ, δ,
to penetrate the Zona Pellucida (ZP) surrounding the oocyte and finally ζ, ε and EFCAB9) making it one of the most complex channels ever
fertilize it. described (Fig. 1, A) (Carlson et al., 2003; Chung et al., 2017, 2011;
Throughout this long journey, the sperm cell must continuously Darszon et al., 2011; Hwang et al., 2019; Quill et al., 2001; Ren et al.,
adapt to new environments to deliver the genetic information needed to 2001). The high complexity of CatSper is thought to be required for the
create a new individual. From capacitation and navigation to hyper­ diverse types of stimuli that CatSper can respond to (Visconti et al.,
activation and acrosome reaction, calcium ions play a central role by 2011).
acting at diverse spatial sites to orchestrate these various events (Pub­
licover et al., 2007, 2008). As the principal calcium channel of sperm, 2.1.1. Pore-forming α subunits: CatSper 1-4
CatSper is the main gate through which extracellular calcium enters the In 2001, the first member of CatSper (renamed CatSper 1 after the
sperm flagellum and is therefore vital for sperm fertility (Lishko and discovery of other subunits) was identified during sequence homology
Mannowetz, 2018). Even though the fertilization process is at the center searches to the voltage-gated Ca2+ channels Cav (Ren et al., 2001). Very
of creating a new life, it is far from being completely understood. shortly afterward, CatSper 2 was identified as another pore-forming α
Improving our understanding of the complexity of the CatSper channel subunit using a signal peptide trapping method (Quill et al., 2001).
is not only relevant to better decipher the fertilization process but also to CatSper 3 and CatSper 4 were later identified using in silico database
tackle the increasing male infertility problems on a global level. In mining (Lobley et al., 2003). Structurally, each of the four distinct
addition, disposing of male contraception alternatives is becoming ur­ subunits has six transmembrane segments (TM), unlike the Cav that has
gent in a world continuously seeking sexual equity. The scope of this 24 TM segments in a single polypeptide (Lobley et al., 2003). Impor­
review is, therefore, to focus on the CatSper channel and its function in tantly, targeted disruption of each of the four pore-forming α subunits
sperm fertilizing capacities. CatSper 1–4 leads to male infertility caused by loss of channel function
and absence of hyperactivated motility (Carlson et al., 2005, 2003; Qi
2. CatSper: the complex, promiscuous and essential calcium et al., 2007; Quill et al., 2003, 2001). It has also been shown that
channel of the sperm flagellum CatSper2, CatSper3 and CatSper4 co-precipitate with CatSper1 either
from mouse testis lysate or in a heterologous system indicating that
2.1. Structure CatSper pore is formed by all four subunits (Qi et al., 2007). Similarly,
humans bearing pathogenic mutations in the CatSper1 and CatSper2
Being exclusively expressed in spermatozoa and evolutionarily genes suffer from infertility. Like in all other voltage-gated cation
conserved among species from mammals to invertebrates, CatSper acts channels, CatSper 1 and CatSper 2 have several positively charged res­
as a polymodal, chemosensory calcium channel that is unique in con­ idues in the 4th TM segment. However, CatSper 3 and CatSper 4 have
trolling a variety of events leading to fertilization. A combination of only two positively charged residues which might explain the mild
mouse genetics, as well as the application of patch-clamp techniques, voltage dependence of CatSper (Ren et al., 2001). CatSper 1 is different

Fig. 1. Structure of the mammalian CatSper channel and its localization in the sperm flagellum.
A: Located in the principal piece of the sperm flagellum, the heteromeric voltage-dependent, pH-sensitive CatSper channel is composed of ten subunits. The channel
consists of four pore-forming α subunits (CatSper 1–4) as well as five auxiliary subunits: CatSper β (beta), CatSper γ (gamma), CatSper δ (delta), CatSper ζ (zeta) and
CatSper ε (epsilon). A new member of the CatSper complex has been recently identified as EF-hand calcium-binding domain-containing protein 9 or EFCAB9, a
calmodulin-like protein that binds Ca 2+ and acts as a dual calcium and pH sensor for CatSper. B: CatSper channels form functional domains within the principal piece
to enable rapid propagation of the calcium signals along the flagellum. The channel complex is organized in quadrilateral longitudinal nanodomains that form a
unique pattern of four linear threads running down the principal piece of the flagellum.

2
R. Rahban and S. Nef Molecular and Cellular Endocrinology xxx (xxxx) xxx

than the three other α subunits in the fact that it possesses a microtubule-based axoneme which comprises two central pairs of mi­
histidine-rich large cytoplasmic terminal domain that is speculated to crotubules doublets and nine outer microtubule doublets forming the “9
function as a pH sensor (83 histidines in the 446 amino acid composing + 2” pattern. Nine outer dense fibers (ODF) and the FS closely lying
the N terminus) (Ren et al., 2001). The sequence similarity between the under the plasma membrane surrounding the axoneme as well as two
four α subunits is indeed low and ranges between 16 and 22% (Navarro peripheral longitudinal columns (LC) connected by circumferential ribs
et al., 2008). Similarly, the interspecies homology of the different (Fig. 1, B).
CatSper subunits between mouse and human, is relatively low, ranging Even though it is situated in the principal piece of the sperm fla­
from 50% (CATSPER1) to 69% (CATSPER4) (Brown et al., 2019). This gellum, CatSper mediates calcium influx that propagates within seconds
could explain the significant differences in the function and regulation of through the midpiece and the head (Xia et al., 2007). This calcium
CatSper in these species. propagation is sustained in the sperm head and is thought to be impli­
cated in the acrosome reaction (discussed in paragraph 2.3.3) (Xia and
2.1.2. Auxiliary subunits: CatSper β, γ, δ, ζ, ε and EFCAB9 Ren, 2009a). A mathematical model developed to investigate this
CatSper β was the first reported auxiliary subunit of CatSper that was propagation proposes that CatSper mediated calcium influx induces
identified using database search and qRT-PCR (Liu et al., 2007). It additional calcium release from internal stores located in the sperm
contains two TM segments with two short cytoplasmic domains and a midpiece (Olson et al., 2010). A similar pattern has been observed when
large extracellular domain (Fig. 1, A). In contrast, CatSper γ contains human sperm are stimulated with odorant such as bourgeonal (Spehr
only one TM segment with a large extracellular domain and a short et al., 2003). The physiological relevance of this calcium propagation, in
cytoplasmic tail. The large extracellular domains of CatSper β and addition to being associated with acrosome reaction, is thought to
CatSper γ suggest possible channel modulation by external signals such induce hyperactive motility and regulate chemotaxis (discussed in
as ligands and cell-cell interactions during sperm maturation in the paragraph 2.3). Using super-resolution fluorescent microscopy, Chung
epididymis and/or in the female reproductive tract (Darszon et al., and colleagues determined the distribution of CatSper proteins at a
2011). CatSper δ has a single TM segment with a large extracellular nanometer-scale resolution (Chung et al., 2017, 2014). CatSper channel
domain and a short cytoplasmic tail, very similar to CatSper γ (Chung complex is organized in quadrilateral longitudinal nanodomains that
et al., 2011). It has been shown that the sperm plasma membrane of form a unique pattern of four linear ‘stripes’ running down the principal
CatSper1 null mice also lacks the other three components of the subunit α piece of the flagellum (Fig. 1, B). Each of these domains runs along each
(CatSper 2–4), as well as the auxiliary subunits CatSper β, CatSper γ and side of the two longitudinal columns of the sperm flagella. In addition,
CatSper δ (Babcock, 2007; Chung et al., 2011; Qi et al., 2007). CatSper ζ each of these four lines was found to be formed by two rows of CatSper
and ε co-localize with the rest of the CatSper complex. However, unlike complexes. It has been hypothesized that within a nanodomain, the
other CatSper knockout mouse models, lack of CatSperζ disrupts the CatSperζ and EFCAB9 subunits connect and interact with the neigh­
quadrilateral longitudinal nanodomain arrangement of CatSper complex boring CatSper complex either in a zigzag manner to the adjacent col­
but the channel remains functional and only leads to subfertility in the umn or linearly in the same column to structurally and functionally link
mutant mice (Chung et al., 2017, 2011). These studies indicate that the CatSper complexes (Bystroff, 2018; Vyklicka and Lishko, 2020).
CatSper ζ is involved in the compartmentalization of calcium signaling These hypothetical arrangements could be responsible for the synchro­
and can modulate the calcium mechanism of CatSper. While the quad­ nization of the CatSper opening as well as a more efficient propagation
rilateral Ca2+ signaling threads are disrupted in CATSPER 1 knockout of Ca2+ waves over the length of flagella (Fig. 1, B).
mouse sperm (Chung et al., 2014), they seem not to be in CATSPER 2
deficient human sperm (Schiffer et al., 2020). A new member of the 2.3. Function
CatSper complex was recently identified as EF-hand calcium-binding
domain-containing protein 9 or EFCAB9, a calmodulin-like protein that The undeniable importance of CatSper during fertilization lies in the
binds Ca 2+ and acts as a dual calcium and pH sensor (Hwang et al., fact that null mice lacking any of CatSper isoforms are infertile (Jin
2019). EFCAB9 is an evolutionarily conserved component of the CatSper et al., 2007; Quill et al., 2003, 2001; Ren et al., 2001) and men with
channel and is a direct binding partner of CatSperζ. The mutations or deletions in CATSPER1, CATSPER2 and CATSPERE suffer
EFCAB9-CatSperζ complex is associated with the channel pore and from infertility (Avenarius et al., 2009; Avidan et al., 2003; Brown et al.,
confers pH-dependent activation and Ca2+ sensitivity thus regulating 2018; Hildebrand et al., 2010; Luo et al., 2019; Smith et al., 2013;
the opening and closing of CatSper channel. Null-mice deficient for Williams et al., 2015; Zhang et al., 2007). To understand what makes
CatSperz, Efcab9, or lacking both subunits, were shown to be severely CatSper-deficient males infertile, it is important to summarize our cur­
subfertile with sperms that failed to acquire hyperactivated motility rent understanding of the role of CatSper in sperm fertilizing capacities.
(Chung et al., 2017; Hwang et al., 2019). The absence of EFCAB9 and/or
CatSperζ renders the channel much less responsive to alkalinization in 2.3.1. Rheotaxis, thermotaxis, and chemotaxis
the presence of physiologically relevant Ca2+, ultimately making the During their journey from ejaculation to fertilization, spermatozoa
channel a less efficient Ca2+ conductor. While CatSper channel orga­ must travel a long distance and survive in a variety of environments
nizes in four doublet bands in the flagellar membrane, loss of Efcab9 characterized by diverse physical and chemical conditions (Fig. 2).
alters these doublet tracks as well as sperm surface nanoarchitecture Because of the long distance and the complexity of the convoluted ductal
(Chung et al., 2017, 2014; Hwang et al., 2019). Interestingly, the loss of oviduct, several sperm guidance mechanisms exist employing short- and
CatSperz in mice delayed the targeting of the CatSper complex to the long-range cues enabling capacitated sperm to reach the oocyte. Most of
flagellum, but also led to a periodic thinning and disruption of the linear these mechanisms were well described in sea urchin sperm from Arabica
signaling domains of CatSper (Chung et al., 2017). This suggests that Punctulata but our current knowledge on mammalian guidance mecha­
CatSperζ could coordinate the insertion of CatSper channel complexes in nisms is still very sparse (Kaupp et al., 2008). Spermatozoa employ at
the flagellar membrane and the underlying fibrous sheath (FS) proteins least three different guidance mechanisms: (i) rheotaxis that refers to the
along the axoneme (Fig. 1, B). directed movement against the fluid flow (Miki and Clapham, 2013), (ii)
thermotaxis that refer to directed movement in a temperature gradient
2.2. Localization (Bahat et al., 2003) and (iii) chemotaxis that refers to the directed
movement of a cell in a gradient of chemoattractant (Fabro et al., 2002;
CatSper channels form functional domains within the principal piece Oliveira et al., 1999; Ralt et al., 1994). While chemotaxis is a short-range
to enable efficient and fast calcium signal propagation along the fla­ mechanism acting at the order of the millimeter, rheotaxis and ther­
gellum. The principle piece is characterized by the presence of a motaxis are long-range mechanisms guiding spermatozoa along the

3
R. Rahban and S. Nef Molecular and Cellular Endocrinology xxx (xxxx) xxx

Fig. 2. The journey of a sperm in the female reproductive tract.

oviduct to the site of fertilization. Interestingly, all three mechanisms of there is currently, no data available indicating that CatSper is directly
guidance have been reported to be effective only on capacitated sper­ involved in mammalian thermotaxis. However, the sperm’s sodium
matozoa, involve hyperactivation and depend, at least in part, on channel TRPV4 which is an important regulator of the activity of
intracellular Ca2+ suggesting commonality of guidance mechanisms in CatSper was found to be activated by warm temperature (37 ◦ C at the
mammalian spermatozoa (Boryshpolets et al., 2015; Serafín site of fertilization) (Mundt et al., 2018). TRPV4-mediated sodium influx
Pérez-Cerezales, 2015). was shown to induce membrane depolarization, which in turn activates
CatSper is ideally positioned to control sperm chemotaxis because both Hv1 and CatSper. According to the proposed model, Hv1 then ex­
chemotactic movements that guide the spermatozoa to the egg are trudes protons out of the sperm leading to intracellular alkalization and
dependent on asymmetrical movement (Eisenbach and Giojalas, 2006). further activation of CatSper (further discussed in paragraph 2.3 and
The discovery that both human and rabbit spermatozoa are sensitive to Fig. 3). It is, therefore, possible that thermotaxis indirectly regulates
picomolar concentrations of progesterone secreted by the cumulus cells CatSper. Besides, there are indications that the thermosensors for sperm
surrounding the oocyte lead to the establishment that progesterone thermotaxis in mammals may involve opsins (Pérez-Cerezales et al.,
plays the role of a sperm chemoattractant in humans (Fig. 2, B) (Gui­ 2015).
dobaldi et al., 2008; Teves et al., 2006). This remains however debatable
(Chang and Suarez, 2010) even after the discovery that progesterone act 2.3.2. Hyperactivation
mainly through CatSper in humans but not in murine sperm (Lishko Calcium influx through CatSper and mobilization of intracellular
et al., 2011; Strünker et al., 2011). Exposure of human sperm to gradi­ calcium stores are thought to be one of the key events triggering sperm
ents of progesterone leads to a gradual and transient increase in [Ca2+]i hyperactivation (Lishko et al., 2011; Marquez et al., 2007a; Mortimer,
in the sperm head and calcium oscillations at the base of the flagellum 1997; Publicover et al., 2008; Strünker et al., 2011) (Fig. 2, B). After
(Harper et al., 2004). Calcium oscillations often start at low progester­ ovulation, the egg is stored for a short period in the ampulla of the
one levels and their frequency does not change with increasing con­ oviduct, the location where fertilization occurs. The acquisition of hy­
centration of progesterone. These oscillations regulate flagellar beat but peractive motility is essential for enabling the sperms to detach from the
do not induce the acrosome reaction (Bedu-Addo et al., 2007). ciliary oviductal epithelium of the isthmus where they tend to form a
Although progesterone is the best-known chemoattractant, reservoir, arrive at the ampulla at the right time and overcome the
numerous other compounds secreted either from the cumulus cell sur­ protective vestment of the oocyte (De Jonge, 2005; Demott and Suarez,
rounding the oocyte or the oocyte itself have been reported to act as 1992; Suarez, 2008). The current model explaining this high-amplitude
chemoattractants for mammalian spermatozoa. It includes an uniden­ asymmetrical flagellar bending in mammalian sperm is thought to be
tified hydrophobic non-peptide molecule (Armon et al., 2014), chemo­ initiated by three main events: the alkalinization of the intracellular
kine CCL20 (Caballero-Campo et al., 2014), the natriuretic peptide type environment, the depolarization of the membrane, and an increase in
C (NPPC) (Kong et al., 2017) and alluring (Burnett et al., 2011), amongst the intracellular calcium concentration (Lishko and Mannowetz, 2018).
other. Whether these compounds are physiologically relevant and Comparison of the flagellar beating of CatSper1-null spermatozoa with
require CatSper and calcium signaling is still unclear. wild type revealed that flagellar beating remains symmetric with small
Concerning rheotaxis and sperm rolling (i.e. the rotation of sperm amplitude and low curvature even in capacitating medium (Carlson
around their longitudinal axis), it was initially suggested to require Ca2+ et al., 2003). CatSper-deficient sperm cells indeed are unable to acquire
influx via CatSper (Kaupp and Strünker, 2017). Indeed, mouse sperms hyperactivation and therefore fail to penetrate the egg’s vestment (Qi
lacking CatSper1 fail to undergo rolling and rheotaxis (Miki and Clap­ et al., 2007). CatSper-null mouse spermatozoa have also been shown to
ham, 2013). However, the hypothesis that rolling and rheotaxis are be unable to fertilize ZP-intact oocyte but do fertilize ZP-free oocyte,
mediated by Ca2+ influx is at odds with some data in mice and humans indicating that CatSper channels are required to allow the sperm to
showing that sperm rolling does not require extracellular Ca2+ (Muschol penetrate the ZP (Ren et al., 2001). The central role of CatSper-mediated
et al., 2018) and that exposure of human sperm to flow velocity gradi­ [Ca2+]i increase in inducing hyperactivation was however challenged
ents does not cause measurable changes in [Ca2+]i (Zhang et al., 2016). and other calcium channels from internal calcium stores by Ca2+
It is only until very recently that a new piece of the puzzle revealed that -induced Ca2+ release (CICR) were found to have an important role in
rolling and rheotaxis of human and mouse sperm do not require func­ hyperactivation (Alasmari et al., 2013). These calcium stores involve
tional CatSper and transmembrane calcium signaling (Schiffer et al., activation of Inositol 1,4,5-triphosphate receptor (IP3R) and Ryanodine
2020). These results support the concept that passive biomechanical and receptors (RyR) (Alasmari et al., 2013; Darszon et al., 2011; Publicover
hydrodynamic processes enable sperm rolling and rheotaxis rather than et al., 2008). These findings by Alasmari et al. confirm the important
calcium signaling. role of CatSper in regulating motility but show that, at least in human
With regards to sperm guidance by thermotaxis, to our knowledge, sperm, its role is not directly to support hyperactivation. While

4
R. Rahban and S. Nef Molecular and Cellular Endocrinology xxx (xxxx) xxx

Fig. 3. Regulation of CatSper channel in human sperm.

mobilization of stored Ca2+ was a potent inducer of hyperactivated the cumulus cells is also known to trigger a wave of increase in [Ca 2+]i
motility, the main functional effect of activating CatSper was to enhance and promote priming and AR (Meizel et al., 1997; Uñates et al., 2014).
penetration into viscous medium which was abolished in presence of the We can distinguish two types of calcium signaling during this unique,
CatSper inhibitor NNC55-0396 (Alasmari et al., 2013). However, in a tightly regulated and irreversible exocytotic process; initial calcium rise
more recent finding of a potent and specific CatSper inhibitor (RU1968), upon binding of sperm to ZP or in response to progesterone activation
Rennhack et al. found that application of the inhibitor did not suppress followed by another sustained calcium rise due to calcium release from
hyperactivation in human sperm suggesting that CatSper is involved in intracellular stores (Darszon et al., 2011). It has been proposed that
their ability to acquire the whip-lash movement (Rennhack et al., 2018). CatSper is responsible for the initial transient Ca2+ rise (Singh and
Murine sperms incubated with a calcium-free medium are able to Rajender, 2015). CatSper mediated calcium influx into the flagellum
undergo hyperactivation by mobilization of stored calcium through the then leads to a calcium elevation in the sperm head probably by causing
IP3R that were shown to localize in the sperm neck region (Marquez calcium-dependent release from the intracellular store located in the
et al., 2007b). A plasma membrane calcium- ATPase 4 (PMCA 4), sperm neck (Singh and Rajender, 2015; Xia et al., 2007). The role of the
localized in the principal piece of the flagellum, is responsible for CatSper channel in inducing mammalian AR is still however highly
eliminating calcium that is also crucial for sperm motility and acquiring debatable. In a study showing a tail-to head Ca2+ propagation in mouse
hyperactivation. Indeed, mitochondrial abnormalities found in PMCA sperm, the CatSper-mediated [Ca2+]i increase seemed not to be required
4-deficient spermatozoa lead to a calcium overload resulting in male for AR (Xia et al., 2007). In another more recent study aiming at un­
infertility due to defective calcium extrusion (Okunade et al., 2004). derstanding the relation between acrosome reaction and
Calcium is therefore not only required for the initiation of hyper­ CatSper-mediated calcium influx in human sperm, the effect of proges­
activation but also to its maintenance by directly regulating components terone on AR was shown to be blunted in the presence of T-type channel
of the axonemal machinery (Darszon et al., 2007). blockers NNC at the same concentrations that inhibit 80% of the
progesterone-stimulated calcium influx (Tamburrino et al., 2014). This
2.3.3. Acrosome reaction suggests that CatSper-mediated calcium influx could be regulating the
The AR allows the sperm, by the release of proteolytic enzymes, to acrosome reaction in humans but more experiments are needed to
penetrate the cumulus oophorus and the ZP surrounding the oocyte establish the role of CatSper in this process (Beltrán et al., 2016).
(Fig. 2, B) (Jin et al., 2011). This reaction is a prerequisite for sperma­ A: The female reproductive tract is composed of different regions and
tozoa to successfully fertilize the egg and it is known to be strictly the sperm cells must constantly adapt to different environments
dependent on an increase in [Ca 2+]i (Yanagimachi, 1994). Different throughout their journey from the vagina to the ampulla. The vagina is
theories exist on the nature of the inducers of the human acrosome re­ the site of deposition of semen and where sperms begin to migrate out of
action physiologically. It is generally believed that AR is triggered by the seminal plasma and into the cervical mucus. Out of the millions of
sperm contact with the zona pellucida, although it has been shown that spermatozoa deposited in the vagina, only an estimated number of one
most fertilizing spermatozoa in mouse undergo the AR already within hundred capacitated sperms will meet the egg. Capacitation is mainly
the cumulus oophorus that surrounds the newly ovulated mouse egg induced by exposure of sperms to high concentrations of bicarbonate
(Bedford, 2011; Inoue et al., 2011; Jin et al., 2011; Sun et al., 2011). (HCO−3 ) and serum albumin. After this first selection process, most of the
However, it seems that in humans only acrosome intact sperm cells can sperm cohort continues their way up to the uterus where sperm trans­
bind to the ZP (Liu et al., 2006). While the classical view proposes the port is facilitated via peristaltic contractions and rheotaxis. As they
zona pellucida proteins, mainly ZP3, to be the main inductor of approach the egg at the ampulla, sperms are exposed to warmer tem­
mammalian AR, picomolar concentrations of progesterone secreted by perature at the site of fertilization (37 ◦ C at the ampulla), a guiding

5
R. Rahban and S. Nef Molecular and Cellular Endocrinology xxx (xxxx) xxx

mechanism referred to as thermotaxis. B: High concentrations of pro­ via a membrane endocannabinoid signaling pathway through the α/β
gesterone secreted by the cumulus cells forms a gradient that guides the hydrolase domain-containing protein 2 (ABHD2). At rest, CatSper is
sperm when in close proximity to the egg through chemotaxis. Proges­ thought to be inhibited by the endocannabinoid 2-arachidonoylglycerol
terone, as well as prostaglandins, activate CatSper leading to a rapid (2-AG) in the flagellar membrane. Upon binding of progesterone,
transient increase in intracellular calcium concentration [Ca2+]i. CatS­ ABHD2 degrades 2-AG and thereby relieves CatSper from inhibition
per activation is implicated in hyperactivated motility characterized by (Miller et al., 2016). It is noteworthy to mention that progesterone ac­
a star shaped, non-progressive trajectory in aqueous media. This will tivates CatSper only in human and macaque sperm and does not induce
facilitate sperm penetration through the extracellular matrix of cumulus any increase in [Ca2+]i in mouse sperm (Lishko et al., 2011; Strünker
cells by mechanical shear forces. Sperms will then release their acro­ et al., 2011; Sumigama et al., 2015).
somal content to digest the Zona Pellucida during the acrosome reaction Prostaglandins (PGs): these compounds derived from arachidonic
and finally fertilize the egg. fatty acids are abundant in the seminal plasma and are secreted by the
oviduct and cumulus cells surrounding the oocyte. In addition to pro­
2.4. Regulation and cross-talks gesterone, prostaglandin E1 (PGE1) has also been shown to act through
CatSper and increase intracellular calcium concentration in a very
The application of patch-clamp techniques to measure CatSper cur­ similar biphasic manner with similar amplitude and potentiates the
rents that were adapted to human sperm less than a decade ago paved ICatSper (Lishko et al., 2011; Strünker et al., 2011). The large current
the way to the elucidation of the regulation of this highly complex recorded in the presence of PGE1 was fully inhibited by NNC similarly to
channel (Kirichok et al., 2006; Kirichok and Lishko, 2011; Lishko et al., the calcium influx measured with fluorimetry. The addition of saturating
2011; Strünker et al., 2011). We can divide the mechanisms leading to concentration of PGE1 (2 μM) to sperm cells already potentiated by
CatSper activation in two fashions, the first is ligand-dependent and the progesterone resulted in a further increase in the amplitude of ICatSper
second is ligand-independent. Both mechanisms discussed below are and vice versa. This suggests that although progesterone and PGE1
activated simultaneously and are highly interconnected. activate the same channel, they have two distinct binding sites (Kaupp
and Strünker, 2017; Lishko et al., 2011; Lishko and Mannowetz, 2018;
2.4.1. Ligand-dependent pathway Miller et al., 2015; Strünker et al., 2011). Consistently, it has been
Progesterone: the nanomolar concentrations of progesterone pro­ recently reported that PGs and progesterone act in a highly synergistic
duced and released by the cumulus cells surrounding the oocyte has long manner (Brenker et al., 2018a). Other PGs also induced an increase in
been known to play a central role in human sperm fertilizing capacities intracellular calcium concentration but with different signal amplitudes
by acting primarily on acrosome reaction and sperm motility (Black­ such as PGE1α and PGE2. The relative effects of ligands activating human
more et al., 1990; Forti et al., 1999). However, the unknown mecha­ CatSper can be classified as follows: Progesterone > PGE1α ≈ PGE1 >
nisms by which progesterone act on sperm have fascinated and, at the PGA1 > PGE2 ≫ PGD2 (Lishko et al., 2011; Miller et al., 2015). Similarly
same time, frustrated reproductive scientists. Progesterone classically to progesterone, PGE1 does not induce a calcium influx in mouse sperm
acts in a genomic fashion by binding to a nuclear receptor and initiating underlining once more the important differences between human and
gene transcription, but spermatozoa have a highly condensed genome mouse CatSper regulation (Lishko and Mannowetz, 2018). For a long
and are transcriptionally inactive (Baldi et al., 2009). Various mecha­ time, PGs were thought to bind to a prostanoid receptor on human sperm
nisms have been proposed to explain this “non-genomic” action of (Schaefer et al., 1998) but this is incompatible with the biphasic signal of
progesterone on sperm. The mechanisms mainly involved G-protein PG-induced intracellular calcium increase and the pharmacology of the
coupled progestin receptors (mPR), a putative membrane-associated PGE1 response as measured by fluorimetry (Strünker et al., 2011). The
progesterone steroid receptor PGMRC1 (Progesterone Receptor Mem­ mechanism of action by which PGs act on CatSper thus remains elusive
brane Component 1), as well as second messengers such as cAMP, cGMP, and does not involve ABHD2 (Kaupp and Strünker, 2017).
PKA, PKG - induced calcium release from intracellular stores, SOC and Cyclic nucleotide cAMP and CNG: in intact mouse spermatozoa,
cGMP channels. However, the molecular mechanisms by which pro­ membrane-permeable analogs of cAMP and cGMP (such as 8-Br-cAMP
gesterone acts on sperm have remained elusive despite considerable and 8-Br-cGMP) have been shown to activate CatSper-dependent cal­
efforts by the scientific community (Baldi et al., 2009). The discovery of cium entry and neither cAMP nor cGMP was shown to elevate [Ca2+]i in
CatSper as the principal calcium channel in sperm (Ren et al., 2001) CatSper-null mice (Ren et al., 2001). It was therefore originally proposed
along with the application of patch-clamp techniques in human sperm that the CatSper channel can be directly or indirectly activated by cyclic
have finally led to the discovery of the progesterone’s gateway into nucleotides (Ren et al., 2001). The following discoveries could not
sperm (Publicover and Barratt, 2011). Progesterone, along with an demonstrate a direct [Ca2+]i increase in response to cyclic nucleotides,
alkaline pH, stimulates a rapid calcium entry with almost no latency that to photolysis of caged cAMP or in response to bicarbonate-induced
is incompatible with a signaling pathway involving metabotropic re­ elevation of cAMP (Brenker et al., 2012; Strünker et al., 2011; Wenne­
ceptors and second messengers. Using the patch-clamp technique as well muth et al., 2003). CatSper-dependent increase in [Ca2+]i induced by
as optical fluorimetry in sperms loaded with a calcium indicator, pro­ alkalinization and high [K+] was however strongly facilitated by the
gesterone was shown to induce a biphasic signal of a rapid and transient presence of bicarbonate that is known to increase cAMP levels through
calcium increase followed by a slower sustained calcium elevation sAC (Carlson et al., 2003; Xie et al., 2006). In mouse, patch-clamp re­
(Strünker et al., 2011). The T-type channel blockers, mibefradil, and cordings of CatSper currents were not affected by cyclic nucleotides
NNC, significantly altered the progesterone-induced calcium response. (Kirichok et al., 2006) suggesting that the facilitation of CatSper by
The constants of half-maximal activation (K1/2) of progesterone-induced cyclic nucleotides is indirect and involves an intermediary signaling
increase in calcium were shifted to lower concentration in capacitated cascade that is disrupted during patch-clamp recordings (Kirichok and
sperm suggesting that capacitation renders sperm more sensitive to Lishko, 2011). It has been therefore proposed that cAMP facilitates
progesterone (Strünker et al., 2011). Using the patch-clamp, Lishko and CatSper-dependent calcium increase via PKA-dependent phosphoryla­
colleagues demonstrated that the addition of progesterone dramatically tion (Orta et al., 2018).
increased the amplitude of human monovalent CatSper current (ICatSper) Albumin and Zona Pellucida glycoproteins: cholesterol efflux by serum
(Lishko et al., 2011). After the action of progesterone and PGE1 on albumin is a key component in sperm cell capacitation and the influx of
human sperm was proved to be “non-genomic” because of the rapid calcium during this process has been linked directly to the presence of
activation rate, it became clear that progesterone either binds directly to CatSper within sperm cells (Xia and Ren, 2009b). Indeed, Bovine Serum
CatSper or to a receptor that is directly associated with the channel Albumin (BSA) - induced [Ca2+]i increase was shown by patch-clamp
(Strünker et al., 2011). Progesterone was later proved to act on CatSper recordings to be absent in CatSper-deficient sperm. The mechanisms

6
R. Rahban and S. Nef Molecular and Cellular Endocrinology xxx (xxxx) xxx

by which BSA is coupled to the CatSper channel are still largely un­ domain of the other CatSper subunits in contrast to CatSper 1 (Navarro
known (Ren and Xia, 2010; Sun et al., 2017). Application of solubilized et al., 2008; Qi et al., 2007). Human ICatSper recordings revealed
ZP glycoproteins induces an intracellular calcium increase that is known fundamental differences with their rodent’s counterpart. The voltage
to be involved in inducing a change in motility and allowing the release dependence of human CatSper is slightly steeper (k = 20) and half
of the sperm acrosomal content, which is crucial for penetrating the egg. activation voltage in human sperm is strikingly higher with V1/2 human
A suggested role for CatSper in the early phase of ZP-induced increase in = +85 mV compared to V1/2 mouse = +11 mV at the same intracellular
[Ca2+]i was proposed. In CatSper 1- deficient mice sperm, the early pH (7.5) (Kirichok et al., 2006; Lishko et al., 2011). This again suggests
calcium response induced by solubilized ZP glycoproteins is absent and that the regulation of mouse and human CatSper is fundamentally
can be restored with exogenous expression of Green Fluorescent Proteins different. Although the CatSper channel has only weak voltage depen­
(GFP)-tagged CatSper 1 protein (Xia and Ren, 2009a). The mechanisms dence, it is still very important to the optimal functioning of the channel
involved in transducing ZP-induction to the opening of CatSper channels since it is directly connected to the channel’s pH sensitivity and the
are currently unknown (Ren and Xia, 2010). activation of pH-regulating channels such as Hv1. At low intracellular
pH, the CatSper voltage dependence regulates the channel status by
2.4.2. Ligand-independent pathway keeping it closed at sperm physiological membrane potentials (from − 70
Intracellular alkaline pH: intracellular pH is a critical regulator of mV to 0 mV). An increase in intracellular pH significantly shifts murine
sperm activity, from the moment they are released in the lumen of the CatSper voltage dependence towards the negative membrane potentials
seminiferous tubules until they reach and fertilize the egg. While stored allowing the channel to open by lower potentials in the physiological
in the epididymis, the sperms bath in an acidic milieu (pH = 5.5–6.8) range (Kirichok et al., 2006). The principal regulator of membrane po­
making their intracellular pH (pHi) also acidic (≈6) that helps them tential is attributed to the potassium permeability that is regulated by
remain quiescent before ejaculation (Acott and Carr, 1984; Jones and two members of the Slo family of potassium channels: Slo1 and Slo3
Bavister, 2000). Upon entering the female reproductive tract, sperma­ (Mannowetz et al., 2013; Navarro et al., 2008; Schreiber et al., 1998).
tozoa are exposed to a higher extracellular pH (pHO) of around 7 due to The principal potassium channel in mouse - Slo3 - is pH-sensitive, cal­
the presence of a high concentration of Na+ that elevates the sperm pHi cium-independent, and voltage-sensitive (Brenker et al., 2014; Santi
to about 6.5 as they first become motile (Carr and Acott, 1989; Kirichok et al., 2010; Schreiber et al., 1998). Slo3 deficient mice display severely
and Lishko, 2011). During their subsequent transit, pHi further increases reduced male fertility (Santi et al., 2010; Zhang et al., 2006). In contrast
as a result of sperm capacitation but remains below pHO. This increase in to mouse, human sperm potassium current (KSper) is pH-independent,
pH has been shown to activate the CatSper channel in mice and humans sensitive to [Ca2+]i and can be inhibited by progesterone (Mannowetz
(Kirichok et al., 2006; Lishko et al., 2011; Strünker et al., 2011). et al., 2013). In humans, calcium rather than pHi was shown to control
Application of the patch-clamp technique revealed that intracellular KSper suggesting that the prototypical Ca2+ - regulated K+ channel
alkaline pH strongly potentiates the murine CatSper current (ICatSper) by regulating human KSper is Slo1. However, human Slo3 is also activated
inducing a seven-fold increase leading to an increase in flagellar [Ca2+]i by calcium rather than alkalinization and is abundantly present in the
and sperm hyperactivation (Kirichok et al., 2006). In humans, intra­ human sperm flagellum. This suggests that it is rather Slo3 that repre­
cellular alkalinization by the simple addition of NH4Cl is sufficient to sents the principal K+ channel in human sperm that carries the
induce calcium influx and potentiate ICatSper independently of proges­ Ca2+-activated IKSper current (Brenker et al., 2014). The efflux of po­
terone that does not alter pHi (Strünker et al., 2011). Even if murine and tassium through Slo3 was therefore proposed to regulate CatSper by
human CatSper1 proteins are highly enriched in histidine which is an leading to the membrane hyperpolarization that in turn inhibits the
indication of a channel’s pH sensitivity, intracellular alkalinization progesterone-evoked Ca2+ influx through CatSper (Brenker et al., 2014).
alone seems to be sufficient to open murine CatSper but not human
CatSper which also depends on activation by ligands as previously dis­ 2.4.3. CatSper is promiscuous
cussed. One of the major players regulating the intracellular pH in Under physiological conditions, progesterone, and PGs, together
human but not in mouse sperm is the voltage-gated proton channel Hv1 with membrane voltage and intracellular pH, act collectively to regulate
that is confined to the principal piece of the sperm flagellum and is the calcium entry into sperm via the human CatSper. However, CatSper is
dominant proton conductance channel (Lishko et al., 2010). It allows also modulated by various endogenous or exogenous compounds and is
only outward transport of protons and is therefore dedicated to inducing known for its promiscuity. In particular, CatSper appears to be regulated
intracellular alkalinization. Human sperm was also shown to hold a by several endogenous steroids. Pregnenolone sulfate exerts effects
shorter isoform of Hv1 termed Hv1Sper (Berger et al., 2017). It is similar to those of progesterone, while physiological concentrations of
generated from Hv1 by the removal of 68 amino acids from the N-ter­ testosterone, hydrocortisone and estradiol were first shown to act as
minus by post-translational proteolytic cleavage. In both channels, the antagonists to CatSper and to reduce or prevent CatSper activation by
conductance-voltage relationship is determined by the pH difference progesterone (Mannowetz et al., 2017). However, these results could not
across the membrane (ΔpH). However, a constant ΔpH activates only be reproduced by Brenker et al. whose data lead to entirely different
Hv1Sper but not Hv1. This suggests that cleavage of the N-terminus conclusions. Testosterone, hydrocortisone, and estradiol were shown to
regulates pH sensing in Hv1 (Berger et al., 2017). Hv1 is activated by enhance CatSper currents, with varying potency and efficacy. In addi­
diverse mechanisms: 1) membrane depolarization, 2) an alkaline tion, the three steroids were shown not to antagonize the
extracellular environment similar to the one in the female reproductive progesterone-induced CatSper currents (Brenker et al., 2018b). Simi­
tract (that can reach up to pH = 8), 3) endocannabinoid anandamide, larly, it has been shown that in human follicular fluid unidentified
and 4) removal of extracellular zinc that is a potent Hv1 blocker (Lishko compounds other than progesterone contribute to the increase of [Ca2+]i
et al., 2010; Lishko and Kirichok, 2010). In mouse, the sperm-specific and modulate CatSper activation (Brown et al., 2019). Concerning
sodium/proton exchanger (sNHE) is activated by membrane hyperpo­ exogenous ligands, molecules as diverse as a variety of odorants can
larization and seems to be responsible for intracellular alkalinization directly act on CatSper without involving G protein-coupled receptors
and consequently CatSper activation (Wang et al., 2007, 2003). (GPCRs) or cAMP (Brenker et al., 2012). It has also been shown that
Change in the membrane voltage: the same initial patch-clamp re­ endocrine-disrupting chemicals (EDCs) such as chemical UV filters act
cordings of the CatSper currents also revealed that murine CatSper is directly on CatSper and induce an increase in [Ca2+]i (Brenker et al.,
weakly voltage-dependent with a slope factor (k) of 30 compared to a 2018b; Rehfeld et al., 2017, 2016; Schiffer et al., 2014; Shannon et al.,
much steeper k of about 4 in solely voltage-gated ion channels (Kirichok 2016; Tavares et al., 2013; Yuan et al., 2020). Premature activation of
et al., 2006). The low voltage sensitivity of CatSper can be explained by the CatSper channel by EDCs can potentially desensitize the sperm to
the low number of positively charged residues in the S4 transmembrane physiological ligands such as progesterone and prostaglandins and may,

7
R. Rahban and S. Nef Molecular and Cellular Endocrinology xxx (xxxx) xxx

therefore, alter the precisely coordinated sequence of fertilization. With 3. Major challenges, remaining questions, and perspectives
this in mind, taking advantage of the promiscuous binding to a wide
range of compounds renders CatSper an excellent target for new 3.1. Challenge: functional expression of CatSper
non-hormonal contraceptives. Two potential examples were thought to
be good candidates such as the plant triterpenoids Pristimerine and The regulation of mammalian CatSper by the various stimuli as well
Lupeol, which are similar in structure to steroid hormones. Both com­ as the mechanisms by which channel activity is regulated is far from
pounds were shown to inhibit CatSper activation by progesterone, sperm being completely understood for a variety of reasons. First because the
hyperactivation, and slightly reduce basal motility of capacitated human functional expression of CatSper in any heterologous systems has not yet
sperms (Mannowetz et al., 2017; Brenker et al., 2018b). However, these been achieved since the channel is resistant to functional expression
observations could not be reproduced by Brenker et al. who showed that (Darszon et al., 2011). This inability to express the channel in other cell
both compounds did not affect the progesterone-induced Ca2+ influx types may be related to the complexity of the multi-subunit constituents
(Brenker et al., 2018b). This indicates that further studies are needed to of the channel. Alternatively, it is also possible that this channel requires
address the promiscuous steroid-binding side controlling CatSper. a yet unknown essential subunit for the assembly of a functional CatSper
Regulation of CatSper is critical for proper sperm function and com­ complex. Occurrence and localization of CatSper were indeed shown to
pounds that directly affect CatSper or influence calcium signaling pose a be coordinated with the assembly of the FS proteins along the axoneme
genuine threat to sperm fertilization potential (Miller et al., 2015). (Chung et al., 2017; Smith et al., 2013). It has been also shown that it is
only after the different CatSper subunits are properly positioned on the
2.4.4. Integrated model flagellum and are docked to underlying axonemal structures that the
As sperms enter the female reproductive tract, they are first exposed channel becomes functional (Lishko et al., 2012). The last two CatSper
to a high concentration of bicarbonate and albumin, two main players in subunits - EFCAB9 and CatSper ζ - were relatively recently identified
initiating capacitation by stimulating activation of the sAC/PKA phos­ (Chung et al., 2017; Hwang et al., 2019). Second, the human and murine
phorylation pathway and cholesterol efflux, respectively. At the time of CatSper shows significant differences in both their regulation and ligand
ejaculation, sperms are combined with the seminal fluid, which contains bindings, which certainly reflects an adaptation of sperm to the local
high enough concentration of zinc to block the activity of the Hv1 environment of the female reproductive tract. While both murine and
channel, by binding to two histidine residues that stabilize the channel human CatSper are activated at alkaline pH and are weakly
in the closed state (Cherny and DeCoursey, 1999; Lishko and Kirichok, voltage-sensitive (Lishko et al., 2011; Strünker et al., 2011), only the
2010). As sperms ascend from the vagina through the cervix and into the human and macaque CatSper are sensitive to progesterone (EC50 = 10
upper female reproductive tract, divalent zinc ions are chelated by nM) and prostaglandins (EC50 = 8 nM) ((Lishko et al., 2012; Sumigama
proteins present in the oviductal fluid resulting in gradual activation of et al., 2015). Third, the application of the patch-clamp technique was
Hv1 (Lishko et al., 2010; Lu et al., 2008). PKA also phosphorylates Hv1 first made possible in 2006 on mouse sperm and was only introduced to
further activating the channel and potentiating the efflux of protons human sperm less than a decade ago which is relatively a short period of
(Puga Molina et al., 2018). In order to function properly, CatSper of time (Kirichok et al., 2006; Lishko et al., 2011; Strünker et al., 2011).
human sperm requires three concomitant activation mechanisms: 1) Finally, the lack of suitable pharmacological tools has always hindered
membrane depolarization, 2) intracellular alkalization and 3) the pres­ the elucidation of CatSper function. It is only until recently that a spe­
ence of progesterone. As sperms approach the site of fertilization, they cific inhibitor of CatSper (RU1968) was introduced to help better un­
are guided by rheotaxis and thermotaxis into warmer temperature at the derstand the pharmacological regulation of this complex channel
ampulla and by chemotaxis through the exposure of a gradient of pro­ (Rennhack et al., 2018).
gesterone secreted by cumulus cells surrounding the oocyte. The
required conditions for proper CatSper functioning are simultaneously 3.2. CatSper as a pharmacological target for male-directed contraception
present; the elevated temperature activates the sperm’s sodium channel
TRPV4 (DSper) inducing a membrane depolarization. This activates Hv1 Due to its unique composition, specific expression in sperm, and
(as well as CatSper) leading to extrusion of protons by Hv1, intracellular central role in male fertility, the CatSper channel is a prime target for
alkalization, and further activation of CatSper. The presence of sperms infertility treatment or male contraception. Early studies have already
close to a high concentration of progesterone further activates CatSper explored the contraceptive potential of Anti-CatSper1 IgG (Li et al.,
promoting the calcium influx and the potassium efflux through KSper. 2009) or the Ca2+ channel blocker HC-056456 that has been shown to
The latter leads to membrane hyperpolarization and regulation of block CatSper channel activity and reversibly prevented hyperactivated
CatSper in a negative feedback loop mechanism (Fig. 3). motility of capacitated sperm (Carlson et al., 2009).. We can expect that
The complex CatSper channel is composed of ten subunits and is in the coming years several studies testing the ability of pharmacological
activated in a ligand dependent fashion – by steroids and prostaglandins- or natural compounds to act as CatSper agonists or inhibitors will be
as well as in a ligand independent fashion via the diversified crosstalk identified and made available to the scientific community. Using a
with other channels and ion exchangers. In order to function properly, high-throughput screening technique, thousands of ion channels ligands
CatSper requires three concomitant activation mechanisms: 1) mem­ were assessed for their ability to induce calcium signaling and improve
brane depolarization, 2) intracellular alkalization and 3) presence of sperm motility (Martins Da Silva et al., 2017). This allowed the identi­
progesterone. As sperms approach to the site of fertilization, sperms are fication of a phosphodiesterase inhibitor (PDE3), the trequinsin hydro­
exposed to elevated temperature (37 ◦ C) which induce the activation of chloride, that was shown to act as an efficacious agonist of CatSper, and
sodium channel TRPV4 (also called DSper) leading to membrane de­ increase sperm hyperactivation and penetration into viscous medium
polarization following sodium efflux. This activates Hv1 and CatSper (McBrinn et al., 2019). These pharmacological tools will not only pro­
leading to extrusion of protons by Hv1 and further activation of Catsper vide additional insights into the properties, regulation, and role of the
due to intracellular alkalization. Concomitantly, progesterone binds to CatSper channel but will also allow us to evaluate their potential as
ABHD2 and releases CatSper inhibition by 2-AG promoting the calcium CatSper-targeting drugs for the treatment of infertility or as
influx through CatSper and the potassium efflux through the potassium non-hormonal male contraceptives.
channel Slo3 (also called KSper). Influx of calcium induces hyperactive
motility and efflux of potassium, leads to membrane hyperpolarization 3.3. Remaining questions
and regulation of CatSper in a negative feedback loop mechanism.
Although the role of [Ca2+]i signaling in sperm physiology is central,
several questions remain unanswered regarding CatSper and its role in

8
R. Rahban and S. Nef Molecular and Cellular Endocrinology xxx (xxxx) xxx

mediating sperm function. Future studies should address the precise Bedu-Addo, K., Barratt, C.L.R., Kirkman-Brown, J.C., Publicover, S.J., 2007. Patterns of
[Ca2+]i mobilization and cell response in human spermatozoa exposed to
organization of the CatSper channels along the four longitudinal nano­
progesterone. Dev. Biol. 302, 324–332. https://doi.org/10.1016/j.
domains in the principal piece of the flagellum and how this arrange­ ydbio.2006.09.040.
ment allows synchronization of CatSper opening, making efficient Beltrán, C., Mata-Martínez, E., Chávez, J.C., Sánchez-Cárdenas, C., 2016. Role of ion
propagation of Ca2+ waves possible. The translation of these waves into channels in the sperm acrosome reaction. https://doi.org/10.1007/978-3-319-
30567-7_3.
phosphorylation cascades and structural changes underlying hyper­ Berger, T.K., Fußhöller, D.M., Goodwin, N., Bönigk, W., Müller, A., Dokani
activated asymmetrical motility are also still to be clarified. It is also Khesroshahi, N., Brenker, C., Wachten, D., Krause, E., Kaupp, U.B., Strünker, T.,
important to better understand the species differences between human 2017. Post-translational cleavage of Hv1 in human sperm tunes pH- and voltage-
dependent gating. J. Physiol. 595, 1533–1546. https://doi.org/10.1113/JP273189.
and rodent CatSper and to identify the other endogenous CatSper li­ Blackmore, P.F., Beebe, S.J., Danforth, D.R., Alexander, N., 1990. Progesterone and 17α-
gands acting as a chemoattractant for sperm chemotaxis. In terms of hydroxyprogesterone. Novel stimulators of calcium influx in human sperm. J. Biol.
channel regulation, both progesterone and PGs were shown to act on Chem. 265, 1376–1380.
Boryshpolets, S., Pérez-Cerezales, S., Eisenbach, M., 2015. Behavioral mechanism of
CatSper synergistically and are equally important in inducing calcium human sperm in thermotaxis: a role for hyperactivation. Hum. Reprod. 30, 884–892.
downstream events leading to fertilization (Brenker et al., 2018a). The https://doi.org/10.1093/humrep/dev002.
mechanism of action by which progesterone acts on CatSper to elevate Brenker, C., Goodwin, N., Weyand, I., Kashikar, N.D., Naruse, M., Krä Hling, M., Mü
Ller, A., Kaupp, B., Strü Nker, T., 2012. The CatSper channel: a polymodal
[Ca2+]i has been previously described and is thought to be mediated chemosensor in human sperm. EMBO J. 31, 1654–1665. https://doi.org/10.1038/
through ABHD2 (Miller et al., 2016). However, the mechanism of emboj.2012.30.
CatSper activation by prostaglandins remains elusive and the relation Brenker, C., Zhou, Y., Müller, A., Echeverry, F.A., Trötschel, C., Poetsch, A., Xia, X.-M.,
Bönigk, W., Lingle, C.J., Kaupp, B., Strünker, T., 2014. The Ca 2+-activated K +
with the pH-control of the channel is not yet understood. Indeed, the
current of human sperm is mediated by Slo3. https://doi.org/10.7554/eLife.01438,
specific role of both progesterone and PGE1 during fertilization has not 3, 1438.
been yet fully established (Baldi et al., 2009). This is mainly due to the Brenker, Rehfeld, A., Schiffer, C., Kierzek, M., Kaupp, U.B., Skakkebaek, N.E.,
experimental challenges faced in reproducing the chemical composition Strünker, T., Skakkebæk, N.E., Strünker, T., 2018a. Synergistic activation of CatSper
Ca2+ channels in human sperm by oviductal ligands and endocrine disrupting
of the female reproductive tract on one hand and the lack of appropriate chemicals. Hum. Reprod. 33, 1915–1923. https://doi.org/10.1093/humrep/dey275.
pharmacological tools to decipher the complex crosstalk of ion channels Brenker, Schiffer, C., Wagner, I.V., Tüttelmann, F., Röpke, A., Rennhack, A., Kaupp, U.B.,
involved in regulating sperm physiology on the other hand (Suarez and Strünker, T., 2018b. Action of steroids and plant triterpenoids on CatSper Ca2+
channels in human sperm. Proc. Natl. Acad. Sci. U.S.A. https://doi.org/10.1073/
Pacey, 2006; Suarez, 2008; Barratt and Publicover, 2012). In addition, pnas.1717929115.
the precise role of membrane potential in regulating CatSper in human Brown, S.G., Miller, M.R., Lishko, P.V., Lester, D.H., Publicover, S.J., Barratt, C.L.R.,
sperm is still not yet elucidated and needs further exploration (Brenker Martins, S., Silva, D., 2018. Homozygous in-frame deletion in CATSPERE in a man
producing spermatozoa with loss of CatSper function and compromised fertilizing
et al., 2014; Mannowetz et al., 2013). Mechanisms of CatSper silencing capacity. Hum. Reprod. 33, 1812–1816. https://doi.org/10.1093/humrep/dey278.
in the male reproductive tract and activation in the female genital tract Brown, S.G., Publicover, S.J., Barratt, C.L.R., Martins da Silva, S.J., 2019. Human sperm
also need further investigation. ion channel (dys)function: implications for fertilization. Hum. Reprod. Update 25,
758–776. https://doi.org/10.1093/humupd/dmz032.
Burnett, L.A., Anderson, D.M., Rawls, A., Bieber, A.L., Chandler, D.E., 2011. Mouse
Acknowledgments sperm exhibit chemotaxis to allurin, a truncated member of the cysteine-rich
secretory protein family. Dev. Biol. 360, 318–328. https://doi.org/10.1016/j.
ydbio.2011.09.028.
The authors are grateful to the graphic designer Valentin Durand for Bystroff, C., 2018. Intramembranal disulfide cross-linking elucidates the super-
assistance with the artwork. This work was supported by the Swiss quaternary structure of mammalian CatSpers. Reprod. Biol. 18, 76–82. https://doi.
Centre for Applied Human Toxicology (SCAHT) and by the Département org/10.1016/j.repbio.2018.01.005.
Caballero-Campo, P., Buffone, M.G., Benencia, F., Conejo-García, J.R., Rinaudo, P.F.,
de l’Instruction Publique of the State of Geneva.
Gerton, G.L., 2014. A role for the chemokine receptor CCR6 in mammalian sperm
motility and chemotaxis. J. Cell. Physiol. 229, 68–78. https://doi.org/10.1002/
References jcp.24418.
Carlson, A.E., Burnett, L.A., del Camino, D., Quill, T.A., Hille, B., Chong, J.A., Moran, M.
M., Babcock, D.F., 2009. Pharmacological targeting of native CatSper channels
Acott, T.S., Carr, D.W., 1984. Inhibition of bovine spermatozoa by caudal epididymal
reveals a required role in maintenance of sperm hyperactivation. PloS One 4.
fluid: II. Interaction of pH and a quiescence factor 1. Biol. Reprod. 30, 926–935.
https://doi.org/10.1371/journal.pone.0006844.
https://doi.org/10.1095/biolreprod30.4.926.
Carlson, A.E., Quill, T.A., Westenbroek, R.E., Schuh, S.M., Hille, B., Babcock, D.F., 2005.
Alasmari, W., Costello, S., Correia, J., Oxenham, S.K., Morris, J., Fernandes, L., Ramalho-
Identical phenotypes of CatSper1 and CatSper2 null sperm. J. Biol. Chem. 280,
Santos, J., Kirkman-Brown, J., Michelangeli, F., Publicover, S., Barratt, C.L.R., 2013.
32238–32244. https://doi.org/10.1074/jbc.M501430200.
Ca2+ signals generated by CatSper and Ca2+ stores regulate different behaviors in
Carlson, A.E., Westenbroek, R.E., Quill, T., Ren, D., Clapham, D.E., Hille, B., Garbers, D.
human sperm. J. Biol. Chem. 288, 6248–6258. https://doi.org/10.1074/jbc.
L., Babcock, D.F., 2003. CatSper1 Required for Evoked Ca 2 Entry and Control of
M112.439356.
Flagellar Function in Sperm.
Armon, L., Ben-Ami, I., Ron-El, R., Eisenbach, M., 2014. Human oocyte-derived sperm
Carr, D.W., Acott, T.S., 1989. Intracellular pH regulates bovine sperm motility and
chemoattractant is a hydrophobic molecule associated with a carrier protein. Fertil.
protein Phosphorylation1. Biol. Reprod. 41, 907–920. https://doi.org/10.1095/
Steril. 102, 885–890. https://doi.org/10.1016/j.fertnstert.2014.06.011.
biolreprod41.5.907.
Austin, 1952. The capacitation of the mammalian sperm. Nature 170, 326. https://doi.
Chang, 1951. Fertilizing capacity of spermatozoa deposited into the fallopian tubes.
org/10.1038/170326a0.
Nature 168, 697–698. https://doi.org/10.1038/168697b0.
Avenarius, M.R., Hildebrand, M.S., Zhang, Y., Meyer, N.C., Smith, L.L.H., Kahrizi, K.,
Chang, H., Suarez, S.S., 2010. Rethinking the relationship between hyperactivation and
Najmabadi, H., Smith, R.J.H., 2009. Human male infertility caused by mutations in
chemotaxis in mammalian Sperm1. Biol. Reprod. 83, 507–513. https://doi.org/
the CATSPER1 channel protein. Am. J. Hum. Genet. 84, 505–510. https://doi.org/
10.1095/biolreprod.109.083113.
10.1016/J.AJHG.2009.03.004.
Cherny, V.V., DeCoursey, T.E., 1999. pH-Dependent inhibition of voltage-gated H+
Avidan, N., Tamary, H., Dgany, O., Cattan, D., Pariente, A., Thulliez, M., Borot, N.,
currents in rat alveolar epithelial cells by Zn2+ and other divalent cations. J. Gen.
Moati, L., Barthelme, A., Shalmon, L., Krasnov, T., Ben-Asher, E., Olender, T.,
Physiol. 114, 819–838. https://doi.org/10.1085/jgp.114.6.819.
Khen, M., Yaniv, I., Zaizov, R., Shalev, H., Delaunay, J., Fellous, M., Lancet, D.,
Chung, J.-J., Miki, K., Kim, D., Shim, S.-H., Shi, H.F., Hwang, J.Y., Cai, X., Iseri, Y.,
Beckmann, J.S., 2003. CATSPER2, a human autosomal nonsyndromic male infertility
Zhuang, X., Clapham, D.E., 2017. CatSperζ regulates the structural continuity of
gene. Eur. J. Hum. Genet. 11, 497–502. https://doi.org/10.1038/sj.ejhg.5200991.
sperm Ca2+ signaling domains and is required for normal fertility. Elife 6. https://
Babcock, D.F., 2007. Wrath of the wraiths of CatSper3 and CatSper4. Proc. Natl. Acad.
doi.org/10.7554/eLife.23082.
Sci. U.S.A. https://doi.org/10.1073/pnas.0610909104.
Chung, J.-J., Navarro, B., Krapivinsky, G., Krapivinsky, L., Clapham, D.E., 2011. A novel
Bahat, A., Tur-Kaspa, I., Gakamsky, A., Giojalas, L.C., Breitbart, H., Eisenbach, M., 2003.
gene required for male fertility and functional CATSPER channel formation in
Thermotaxis of mammalian sperm cells: a potential navigation mechanism in the
spermatozoa. Nat. Commun. 2, 153. https://doi.org/10.1038/ncomms1153.
female genital tract [1]. Nat. Med. https://doi.org/10.1038/nm0203-149.
Chung, J.J., Shim, S.H., Everley, R.A., Gygi, S.P., Zhuang, X., Clapham, D.E., 2014.
Baldi, E., Luconi, M., Muratori, M., Marchiani, S., Tamburrino, L., Forti, G., 2009.
Structurally distinct Ca2+ signaling domains of sperm flagella orchestrate tyrosine
Nongenomic activation of spermatozoa by steroid hormones: facts and fictions. Mol.
phosphorylation and motility. Cell 157, 808–822. https://doi.org/10.1016/j.
Cell. Endocrinol. 308, 39–46. https://doi.org/10.1016/J.MCE.2009.02.006.
cell.2014.02.056.
Barratt, C.L.R., Publicover, S.J., 2012. Sperm are promiscuous and CatSper is to blame.
Cooper, T.G., 2007. Sperm maturation in the epididymis: a new look at an old problem.
EMBO J. 31, 1624–1626. https://doi.org/10.1038/emboj.2012.62.
Asian J. Androl. 9, 533–539. https://doi.org/10.1111/j.1745-7262.2007.00285.x.
Bedford, J.M., 2011. Site of the mammalian sperm physiological acrosome reaction.
Proc. Natl. Acad. Sci. U.S.A. https://doi.org/10.1073/pnas.1102296108.

9
R. Rahban and S. Nef Molecular and Cellular Endocrinology xxx (xxxx) xxx

Darszon, A., Nishigaki, T., Beltran, C., Treviño, C.L., 2011. Calcium channels in the Lishko, P.V., Kirichok, Y., 2010. The role of Hv1 and CatSper channels in sperm
development, maturation, and function of spermatozoa. Physiol. Rev. 91, activation. J. Physiol. 588, 4667–4672. https://doi.org/10.1113/
1305–1355. https://doi.org/10.1152/physrev.00028.2010. jphysiol.2010.194142.
Darszon, A., Treviño, C.L., Wood, C., Galindo, B., Rodríguez-Miranda, E., Acevedo, J.J., Lishko, P.V., Kirichok, Y., Ren, D., Navarro, B., Chung, J.-J., Clapham, D.E., 2012. The
Hernandez-González, E.O., Beltrán, C., Martínez-López, P., Nishigaki, T., 2007. Ion control of male fertility by spermatozoan ion channels. Annu. Rev. Physiol. 74,
channels in sperm motility and capacitation. Soc. Reprod. Fertil. Suppl. 65, 229–244. 453–475. https://doi.org/10.1146/annurev-physiol-020911-153258.
De Jonge, C., 2017. Biological basis for human capacitation—revisited. Hum. Reprod. Lishko, P.V., Mannowetz, N., 2018. CatSper: a unique calcium channel of the sperm
Update 23, 289–299. https://doi.org/10.1093/humupd/dmw048. flagellum. Curr. Opin. Physiol. 2, 109–113. https://doi.org/10.1016/J.
De Jonge, C., 2005. Biological basis for human capacitation. Hum. Reprod. Update 11, COPHYS.2018.02.004.
205–214. https://doi.org/10.1093/humupd/dmi010. Liu, D.Y., Garrett, C., Baker, H.W.G., 2006. Acrosome-reacted human sperm in
de Lamirande, E., Leclerc, P., Gagnon, C., 1997. Capacitation as a regulatory event that insemination medium do not bind to the zona pellucida of human oocytes. Int. J.
primes spermatozoa for the acrosome reaction and fertilization. Mol. Hum. Reprod. Androl. 29, 475–481. https://doi.org/10.1111/j.1365-2605.2006.00681.x.
3, 175–194. https://doi.org/10.1093/molehr/3.3.175. Liu, J., Xia, J., Cho, K.-H., Clapham, D.E., Ren, D., 2007. CatSperbeta, a novel
Demott, R.P., Suarez, S.S., 1992. Hyperactivated sperm progress in the mouse Oviduct1. transmembrane protein in the CatSper channel complex. J. Biol. Chem. 282,
Biol. Reprod. 46, 779–785. https://doi.org/10.1095/biolreprod46.5.779. 18945–18952. https://doi.org/10.1074/jbc.M701083200.
Eisenbach, M., Giojalas, L.C., 2006. Sperm guidance in mammals — an unpaved road to Lobley, A., Pierron, V., Reynolds, L., Allen, L., Michalovich, D., 2003. Identification of
the egg. Nat. Rev. Mol. Cell Biol. 7, 276–285. https://doi.org/10.1038/nrm1893. human and mouse CatSper3 and CatSper4 genes: characterisation of a common
Fabro, G., Rovasio, R.A., Civalero, S., Frenkel, A., Caplan, S.R., Eisenbach, M., Giojalas, L. interaction domain and evidence for expression in testis. Reprod. Biol. Endocrinol. 1,
C., 2002. Chemotaxis of capacitated rabbit spermatozoa to follicular fluid revealed 53. https://doi.org/10.1186/1477-7827-1-53.
by a novel directionality-based Assay1. Biol. Reprod. 67, 1565–1571. https://doi. Lu, J., Stewart, A.J., Sadler, P.J., Pinheiro, T.J.T., Blindauer, C.A., 2008. Albumin as a
org/10.1095/biolreprod.102.006395. zinc carrier: properties of its high-affinity zinc-binding site. Biochem. Soc. Trans. 36,
Forti, G., Baldi, E., Krausz, C., Luconi, M., Bonaccorsi, L., Maggi, M., Bassi, F., 1317–1321. https://doi.org/10.1042/BST0361317.
Scarselli, G., 1999. Effects of progesterone on human spermatozoa : clinical Luo, T., Chen, H.-Y., Zou, Q.-X., Wang, T., Cheng, Y.-M., Wang, H.-F., Wang, F., Jin, Z.-L.,
implications Les effets de la progestérone sur le spermatozoïde humain : implications Chen, Y., Weng, S.-Q., Zeng, X.-H., 2019. A novel copy number variation in
cliniques. Annales d’Endocrinologie. CATSPER2 causes idiopathic male infertility with normal semen parameters. Hum.
Gervasi, M.G., Visconti, P.E., 2017. Molecular changes and signaling events occurring in Reprod. 1–10 https://doi.org/10.1093/humrep/dey377.
spermatozoa during epididymal maturation. Andrology. https://doi.org/10.1111/ Mannowetz, N., Miller, M.R., Lishko, P.V., 2017. Regulation of the sperm calcium
andr.12320. channel CatSper by endogenous steroids and plant triterpenoids. Proc. Natl. Acad.
Guidobaldi, H.A., Teves, M.E., Uñates, D.R., Anastasía, A., Giojalas, L.C., 2008. Sci. U.S.A. 114, 5743–5748. https://doi.org/10.1073/pnas.1700367114.
Progesterone from the cumulus cells is the sperm chemoattractant secreted by the Mannowetz, N., Naidoo, N.M., Choo, S.A.S., Smith, J.F., Lishko, P.V., 2013. Slo1 is the
rabbit oocyte cumulus complex. PloS One 3, e3040. https://doi.org/10.1371/ principal potassium channel of human spermatozoa. Elife. https://doi.org/10.7554/
journal.pone.0003040. eLife.01009.001, 2013.
Gupta, G., 2017. Sperm maturation in epididymis. In: Male Infertility: Understanding, Marquez, B., Ignotz, G., Suarez, S.S., 2007a. Contributions of extracellular and
Causes and Treatment. Springer Singapore, Singapore, pp. 37–45. https://doi.org/ intracellular Ca2+ to regulation of sperm motility: release of intracellular stores can
10.1007/978-981-10-4017-7_4. hyperactivate CatSper1 and CatSper2 null sperm. Dev. Biol. 303, 214–221. https://
Harper, C.V., Barratt, C.L.R., Publicover, S.J., 2004. Stimulation of human spermatozoa doi.org/10.1016/j.ydbio.2006.11.007.
with progesterone gradients to simulate approach to the oocyte. Induction of [Ca2+] Marquez, B., Ignotz, G., Suarez, S.S., 2007b. Contributions of extracellular and
i oscillations and cyclical transitions in flagellar beating. J. Biol. Chem. 279, intracellular Ca2+ to regulation of sperm motility: release of intracellular stores can
46315–46325. https://doi.org/10.1074/jbc.M401194200. hyperactivate CatSper1 and CatSper2 null sperm. Dev. Biol. 303, 214–221. https://
Hildebrand, M.S., Avenarius, M.R., Fellous, M., Zhang, Y., Meyer, N.C., Auer, J., doi.org/10.1016/j.ydbio.2006.11.007.
Serres, C., Kahrizi, K., Najmabadi, H., Beckmann, J.S., Smith, R.J.H., 2010. Genetic Martins Da Silva, S.J., Brown, S.G., Sutton, K., King, L.V., Ruso, H., Gray, D.W., Wyatt, P.
male infertility and mutation of CATSPER ion channels. Eur. J. Hum. Genet. 18, G., Kelly, M.C., Barratt, C.L.R., Hope, A.G., 2017. Drug discovery for male
1178–1184. https://doi.org/10.1038/ejhg.2010.108. subfertility using high-throughput screening: a new approach to an unsolved
Hwang, J.Y., Mannowetz, N., Zhang, Y., Everley, R.A., Gygi, S.P., Bewersdorf, J., problem. Hum. Reprod. 32, 974–984. https://doi.org/10.1093/humrep/dex055.
Lishko, P.V., Chung, J.J., 2019. Dual sensing of physiologic pH and calcium by McBrinn, R.C., Fraser, J., Hope, A.G., Gray, D.W., Barratt, C.L.R., Martins da Silva, S.J.,
EFCAB9 regulates sperm motility. Cell 177, 1480–1494. https://doi.org/10.1016/j. Brown, S.G., 2019. Novel pharmacological actions of trequinsin hydrochloride
cell.2019.03.047 e19. improve human sperm cell motility and function. Br. J. Pharmacol. 176, 4521–4536.
Inoue, N., Satouh, Y., Ikawa, M., Okabe, M., Yanagimachi, R., 2011. Acrosome-reacted https://doi.org/10.1111/bph.14814.
mouse spermatozoa recovered from the perivitelline space can fertilize other eggs. Meizel, S., Turner, K.O., Nuccitelli, R., 1997. Progesterone triggers a wave of increased
Proc. Natl. Acad. Sci. U.S.A. 108, 20008–20011. https://doi.org/10.1073/ free calcium during the human sperm acrosome reaction. Dev. Biol. 182, 67–75.
pnas.1116965108. https://doi.org/10.1006/dbio.1997.8477.
Jin, J., Jin, N., Zheng, H., Ro, S., Tafolla, D., Sanders, K.M., Yan, W., 2007. Catsper3 and Miki, K., Clapham, D.E., 2013. Rheotaxis guides mammalian sperm. Curr. Biol. 23,
Catsper4 are essential for sperm hyperactivated motility and male fertility in the 443–452. https://doi.org/10.1016/J.CUB.2013.02.007.
Mouse1. Biol. Reprod. 77, 37–44. https://doi.org/10.1095/biolreprod.107.060186. Miller, M.R., Mannowetz, N., Iavarone, A.T., Safavi, R., Gracheva, E.O., Smith, J.F.,
Jin, M., Fujiwara, E., Kakiuchi, Y., Okabe, M., Satouh, Y., Baba, S.A., Chiba, K., Hill, R.Z., Bautista, D.M., Kirichok, Y., Lishko, P.V., 2016. Unconventional
Hirohashi, N., 2011. Most fertilizing mouse spermatozoa begin their acrosome endocannabinoid signaling governs sperm activation via the sex hormone
reaction before contact with the zona pellucida during in vitro fertilization. Proc. progesterone. Science 352, 555–559. https://doi.org/10.1126/science.aad6887.
Natl. Acad. Sci. U.S.A. 108, 4892–4896. https://doi.org/10.1073/pnas.1018202108. Miller, M.R., Mansell, S.A., Meyers, S.A., Lishko, P.V., 2015. Flagellar ion channels of
Jones, J.M., Bavister, B.D., 2000. Acidification of intracellular pH in bovine spermatozoa sperm: similarities and differences between species. Cell Calcium 58, 105–113.
suppresses motility and extends viable life. J. Androl. 21, 616–624. https://doi.org/ https://doi.org/10.1016/J.CECA.2014.10.009.
10.1002/J.1939-4640.2000.TB02128.X. Mortimer, S.T., 1997. A critical review of the physiological importance and analysis of
Kaupp, U. Benjamin, et al., 2008. Mechanisms of sperm chemotaxis. Annu. Rev. Physiol. sperm movement in mammals. Hum. Reprod. Update 3, 403–439. https://doi.org/
https://doi.org/10.1146/annurev.physiol.70.113006.100654. 10.1093/humupd/3.5.403.
Kaupp, U.B., Strünker, T., 2017. Signaling in sperm: more different than similar. Trends Mundt, N., Spehr, M., Lishko, P.V., 2018. TRPV4 is the temperature-sensitive ion channel
Cell Biol. 27, 101–109. https://doi.org/10.1016/J.TCB.2016.10.002. of human sperm. Elife 7, 1–20. https://doi.org/10.7554/eLife.35853.
Kirichok, Y., Lishko, P.V., 2011. Rediscovering sperm ion channels with the patch-clamp Muschol, M., Wenders, C., Wennemuth, G., 2018. Four-dimensional analysis by high-
technique. Mol. Hum. Reprod. https://doi.org/10.1093/molehr/gar044. speed holographic imaging reveals a chiral memory of sperm flagella. PloS One 13,
Kirichok, Y., Navarro, B., Clapham, D.E., 2006. Whole-cell patch-clamp measurements of e0199678. https://doi.org/10.1371/journal.pone.0199678.
spermatozoa reveal an alkaline-activated Ca2+ channel. Nature 439, 737–740. Navarro, B., Kirichok, Y., Chung, J.-J., Clapham, D.E., 2008. Ion channels that control
https://doi.org/10.1038/nature04417. fertility in mammalian spermatozoa. Int. J. Dev. Biol. 52, 607–613. https://doi.org/
Kirkman-Brown, J.C., Smith, D.J., 2011. Sperm motility: is viscosity fundamental to 10.1387/ijdb.072554bn.
progress? Mol. Hum. Reprod. 17, 539–544. https://doi.org/10.1093/molehr/ Okunade, G.W., Miller, M.L., Pyne, G.J., Sutliff, R.L., O’Connor, K.T., Neumann, J.C.,
gar043. Andringa, A., Miller, D.A., Prasad, V., Doetschman, T., Paul, R.J., Shull, G.E., 2004.
Kong, N., Xu, X., Zhang, Y., Wang, Y., Hao, X., Zhao, Y., Qiao, J., Xia, G., Zhang, M., Targeted ablation of plasma membrane Ca2+-ATPase (PMCA) 1 and 4 indicates a
2017. Natriuretic peptide type C induces sperm attraction for fertilization in mouse. major housekeeping function for PMCA1 and a critical role in hyperactivated sperm
Sci. Rep. 7, 39711. https://doi.org/10.1038/srep39711. motility and male fertility for PMCA4. J. Biol. Chem. 279, 33742–33750. https://
Li, H., Ding, X., Guan, H., Xiong, C., 2009. Inhibition of human sperm function and doi.org/10.1074/jbc.M404628200.
mouse fertilization in vitro by an antibody against cation channel of sperm 1: the Oliveira, R.G., Tomasi, L., Rovasio, R.A., Giojalas, L.C., 1999. Increased velocity and
contraceptive potential of its transmembrane domains and pore region. Fertil. Steril. induction of chemotactic response in mouse spermatozoa by follicular and oviductal
92, 1141–1146. https://doi.org/10.1016/j.fertnstert.2008.07.1751. fluids. J. Reprod. Fertil. 115, 23–27. https://doi.org/10.1530/jrf.0.1150023.
Lishko, P.V., Botchkina, I.L., Fedorenko, A., Kirichok, Y., 2010. Acid extrusion from Olson, S.D., Suarez, S.S., Fauci, L.J., 2010. A model of CatSper channel mediated calcium
human spermatozoa is mediated by flagellar voltage-gated proton channel. Cell 140, dynamics in mammalian spermatozoa. Bull. Math. Biol. 72, 1925–1946. https://doi.
327–337. https://doi.org/10.1016/J.CELL.2009.12.053. org/10.1007/s11538-010-9516-5.
Lishko, P.V., Botchkina, I.L., Kirichok, Y., 2011. Progesterone activates the principal Ca2 Orta, G., de la Vega-Beltran, J.L., Martín-Hidalgo, D., Santi, C.M., Visconti, P.E.,
+ channel of human sperm. Nature 471, 387–391. https://doi.org/10.1038/ Darszon, A., 2018. CatSper channels are regulated by protein kinase A. J. Biol. Chem.
nature09767. 293, 16830–16841. https://doi.org/10.1074/jbc.RA117.001566.

10
R. Rahban and S. Nef Molecular and Cellular Endocrinology xxx (xxxx) xxx

Pérez-Cerezales, S., Boryshpolets, S., Afanzar, O., Brandis, A., Nevo, R., Kiss, V., Strünker, T., Goodwin, N., Brenker, C., Kashikar, N.D., Weyand, I., Seifert, R., Kaupp, U.
Eisenbach, M., 2015. Involvement of opsins in mammalian sperm thermotaxis. Sci. B., 2011. The CatSper channel mediates progesterone-induced Ca 2+ influx in
Rep. 5 https://doi.org/10.1038/srep16146. human sperm. Nature 471, 382–387. https://doi.org/10.1038/nature09769.
Publicover, S., Barratt, C., 2011. Progesterone’s gateway into sperm. Nature 471, Suarez, S.S., 2008. Regulation of sperm storage and movement in the mammalian
313–314. https://doi.org/10.1038/471313a. oviduct. Int. J. Dev. Biol. 52, 455–462. https://doi.org/10.1387/ijdb.072527ss.
Publicover, S., Harper, C.V., Barratt, C., 2007. [Ca2+]i signalling in sperm — making the Suarez, S.S., Pacey, A.A., 2006. Sperm transport in the female reproductive tract. Hum.
most of what you’ve got. Nat. Cell Biol. 9, 235–242. https://doi.org/10.1038/ Reprod. Update 12, 23–37. https://doi.org/10.1093/humupd/dmi047.
ncb0307-235. Sullivan, R., Légaré, C., Lamontagne-Proulx, J., Breton, S., Soulet, D., 2019. Revisiting
Publicover, S.J., Giojalas, L.C., Teves, M.E., de Oliveira, G.S.M.M., Garcia, A.A.M., structure/functions of the human epididymis. Andrology. https://doi.org/10.1111/
Barratt, C.L.R., Harper, C.V., 2008. Ca2+ signalling in the control of motility and andr.12633 andr.12633.
guidance in mammalian sperm. Front. Biosci. 13, 5623–5637. Sullivan, R., Mieusset, R., 2016. The human epididymis: its function in sperm
Puga Molina, L.C., Luque, G.M., Balestrini, P.A., Marín-Briggiler, C.I., Romarowski, A., maturation. Hum. Reprod. Update 22, 574–587. https://doi.org/10.1093/humupd/
Buffone, M.G., 2018. Molecular basis of human sperm capacitation. Front. Cell Dev. dmw015.
Biol. 6, 72. https://doi.org/10.3389/fcell.2018.00072. Sumigama, S., Mansell, S., Miller, M., Lishko, P.V., Cherr, G.N., Meyers, S.A., Tollner, T.,
Qi, H., Moran, M.M., Navarro, B., Chong, J.A., Krapivinsky, G., Krapivinsky, L., 2015. Progesterone accelerates the completion of sperm capacitation and activates
Kirichok, Y., Ramsey, I.S., Quill, T.A., Clapham, D.E., 2007. All Four CatSper Ion CatSper channel in spermatozoa from the rhesus Macaque1. Biol. Reprod. 93 https://
Channel Proteins Are Required for Male Fertility and Sperm Cell Hyperactivated doi.org/10.1095/biolreprod.115.129783.
Motility. Sun, T.T., Chung, C.M., Chan, H.C., 2011. Acrosome reaction in the cumulus oophorus
Quill, T.A., Ren, D., Clapham, D.E., Garbers, D.L., 2001. A voltage-gated ion channel revisited: involvement of a novel sperm-released factor NYD-SP8. Protein Cell.
expressed specifically in spermatozoa. Proc. Natl. Acad. Sci. U.S.A. 98, https://doi.org/10.1007/s13238-011-1022-5.
12527–12531. https://doi.org/10.1073/pnas.221454998. Sun, X. hong, Zhu, Y. ying, Wang, L., Liu, H. ling, Ling, Y., Li, Z. li, Sun, L. bo, 2017. The
Quill, T.A., Sugden, S.A., Rossi, K.L., Doolittle, L.K., Hammer, R.E., Garbers, D.L., 2003. Catsper channel and its roles in male fertility: a systematic review. Reprod. Biol.
Hyperactivated sperm motility driven by CatSper2 is required for fertilization. Proc. Endocrinol. https://doi.org/10.1186/s12958-017-0281-2.
Natl. Acad. Sci. U.S.A. 100, 14869–14874. https://doi.org/10.1073/ Tamburrino, L., Marchiani, S., Minetti, F., Forti, G., Muratori, M., Baldi, E., 2014. The
pnas.2136654100. CatSper calcium channel in human sperm: relation with motility and involvement in
Ralt, D., Manor, M., Cohen-Dayag, A., Tur-Kaspa, I., Ben-Shlomo, I., Makler, A., Yuli, I., progesterone-induced acrosome reaction. Hum. Reprod. 29, 418–428. https://doi.
Dor, J., Blumberg, S., Mashiach, S., Eisenbach, M., 1994. Chemotaxis and org/10.1093/humrep/det454.
chemokinesis of human spermatozoa to follicular Factors1. Biol. Reprod. 50, Tavares, R.S., Mansell, S., Barratt, C.L.R., Wilson, S.M., Publicover, S.J., Ramalho-
774–785. https://doi.org/10.1095/biolreprod50.4.774. Santos, J., 2013. p,p’-DDE activates CatSper and compromises human sperm
Rehfeld, A., Dissing, S., Skakkebæk, N.E., 2016. Chemical UV filters mimic the effect of function at environmentally relevant concentrations. Hum. Reprod. 28, 3167–3177.
progesterone on Ca 2+ signaling in human sperm cells. Endocrinology 157, https://doi.org/10.1093/humrep/det372.
4297–4308. https://doi.org/10.1210/en.2016-1473. Teves, M.E., Barbano, F., Guidobaldi, H.A., Sanchez, R., Miska, W., Giojalas, L.C., 2006.
Rehfeld, A., Egeberg, D.L., Almstrup, K., Petersen, J.H., Dissing, S., Skakkebæk, N.E., Progesterone at the picomolar range is a chemoattractant for mammalian
2017. EDC IMPACT: chemical UV filters can affect human sperm function in a spermatozoa. Fertil. Steril. 86, 745–749. https://doi.org/10.1016/j.
progesterone-like manner. Endocr. Connect. 7, 16–25. https://doi.org/10.1530/ec- fertnstert.2006.02.080.
17-0156. Uñates, D.R., Guidobaldi, H.A., Gatica, L.V., Cubilla, M.A., Teves, M.E., Moreno, A.,
Ren, D., Navarro, B., Perez, G., Jackson, A.C., Hsu, S., Shi, Q., Tilly, J.L., Clapham, D.E., Giojalas, L.C., 2014. Versatile action of picomolar gradients of progesterone on
Perez, G., Jackson, A.C., Hsu, S., Shi, Q., Tilly, J.L., Clapham, D.E., 2001. A sperm different sperm subpopulations. PloS One 9, e91181. https://doi.org/10.1371/
ion channel required for sperm motility and male fertility. https://doi.org/10.1038 journal.pone.0091181.
/35098027. Visconti, P.E., Krapf, D., de la Vega-Beltrán, J.L., Acevedo, J.J., Darszon, A., 2011. Ion
Ren, D., Xia, J., 2010. Calcium signaling through CatSper channels in mammalian channels, phosphorylation and mammalian sperm capacitation. Asian J. Androl. 13,
fertilization. Physiology 25, 165–175. https://doi.org/10.1152/physiol.00049.2009. 395–405. https://doi.org/10.1038/aja.2010.69.
Rennhack, A., Schiffer, C., Brenker, C., Fridman, D., Nitao, E.T., Cheng, Y.-M., Vyklicka, L., Lishko, P.V., 2020. Dissecting the signaling pathways involved in the
Tamburrino, L., Balbach, M., Stölting, G., Berger, T.K., Kierzek, M., Alvarez, L., function of sperm flagellum. Curr. Opin. Cell Biol. https://doi.org/10.1016/j.
Wachten, D., Zeng, X.-H., Baldi, E., Publicover, S.J., Kaupp, U.B., Strünker, T., 2018. ceb.2020.01.015.
A novel cross-species inhibitor to study the function of CatSper Ca 2+ channels in Wang, D., Hu, J., Alexandru Bobulescu, I., Quill, T.A., McLeroy, P., Moe, O.W.,
sperm. Br. J. Pharmacol. 175, 3144–3161. https://doi.org/10.1111/bph.14355. Garbers, D.L., 2007. A Sperm-specific Na/H Exchanger (sNHE) Is Critical for
Santi, C.M., Martínez-López, P., de la Vega-Beltrán, J.L., Butler, A., Alisio, A., Expression and in Vivo Bicarbonate Regulation of the Soluble Adenylyl Cyclase
Darszon, A., Salkoff, L., 2010. The SLO3 sperm-specific potassium channel plays a (sAC).
vital role in male fertility. FEBS Lett. 584, 1041–1046. https://doi.org/10.1016/j. Wang, D., King, S.M., Quill, T.A., Doolittle, L.K., Garbers, D.L., 2003. A new sperm-
febslet.2010.02.005. specific Na+/H+ exchanger required for sperm motility and fertility. Nat. Cell Biol.
Schaefer, M., Hofmann, T., Schultz, G., Gudermann, T., 1998. A new prostaglandin E 5, 1117–1122. https://doi.org/10.1038/ncb1072.
receptor mediates calcium influx and acrosome reaction in human spermatozoa. Wennemuth, G., Babcock, D.F., Hille, B., 2003. Calcium clearance mechanisms of mouse
Proc. Natl. Acad. Sci. U.S.A. 95, 3008–3013. https://doi.org/10.1073/ sperm. J. Gen. Physiol. 122, 115–128. https://doi.org/10.1085/jgp.200308839.
pnas.95.6.3008. Williams, H.L., Mansell, S., Alasmari, W., Brown, S.G., Wilson, S.M., Sutton, K.A.,
Schiffer, C., Müller, A., Egeberg, D.L., Alvarez, L., Brenker, C., Rehfeld, A., Miller, M.R., Lishko, P.V., Barratt, C.L.R., Publicover, S.J., Martins Da Silva, S., 2015.
Frederiksen, H., Wäschle, B., Kaupp, U.B., Balbach, M., Wachten, D., Skakkebaek, N. Specific loss of CatSper function is sufficient to compromise fertilizing capacity of
E., Almstrup, K., Strünker, T., 2014. Direct action of endocrine disrupting chemicals human spermatozoa. Hum. Reprod. 30, 2737–2746. https://doi.org/10.1093/
on human sperm. EMBO Rep. 15, 758–765. https://doi.org/10.15252/ humrep/dev243.
embr.201438869. Xia, J., Reigada, D., Mitchell, C.H., Ren, D., 2007. CATSPER channel-mediated Ca2+
Schiffer, C., Rieger, S., Brenker, C., Young, S., Hamzeh, H., Wachten, D., Tüttelmann, F., entry into mouse sperm triggers a tail-to-head Propagation1. Biol. Reprod. 77,
Röpke, A., Kaupp, U.B., Wang, T., Wagner, A., Krallmann, C., Kliesch, S., Fallnich, C., 551–559. https://doi.org/10.1095/biolreprod.107.061358.
Strünker, T., 2020. Rotational motion and rheotaxis of human sperm do not require Xia, J., Ren, D., 2009a. Egg coat proteins activate calcium entry into mouse sperm via
functional CatSper channels and transmembrane Ca 2+ signaling. EMBO J. 1–15. CATSPER Channels1. Biol. Reprod. 80, 1092–1098. https://doi.org/10.1095/
https://doi.org/10.15252/embj.2019102363. biolreprod.108.074039.
Schreiber, M., Wei, A., Yuan, A., Gaut, J., Saito, M., Salkoff, L., 1998. Slo3, a novel pH- Xia, J., Ren, D., 2009b. The BSA-induced Ca(2+) influx during sperm capacitation is
sensitive K + channel from mammalian spermatocytes. J. Biol. Chem. 273, CATSPER channel-dependent. Reprod. Biol. Endocrinol. 7, 119. https://doi.org/
3509–3516. https://doi.org/10.1074/jbc.273.6.3509. 10.1186/1477-7827-7-119.
Serafín Pérez-Cerezales, S.B.M.E., 2015. Behavioral mechanisms of mammalian sperm Xie, F., Garcia, M.A., Carlson, A.E., Schuh, S.M., Babcock, D.F., Jaiswal, B.S., Gossen, J.
guidance. Asian J. Androl. 628–632. https://doi.org/10.4103/1008-682X.154308. A., Esposito, G., van Duin, M., Conti, M., 2006. Soluble adenylyl cyclase (sAC) is
Shannon, M., Rehfeld, A., Frizzell, C., Livingstone, C., McGonagle, C., Skakkebaek, N.E., indispensable for sperm function and fertilization. Dev. Biol. 296, 353–362. https://
Wielogórska, E., Connolly, L., 2016. In vitro bioassay investigations of the endocrine doi.org/10.1016/J.YDBIO.2006.05.038.
disrupting potential of steviol glycosides and their metabolite steviol, components of Yanagimachi, 1994. Mammalian fertilization. In: The Physiology of Reproduction.
the natural sweetener Stevia. Mol. Cell. Endocrinol. 427, 65–72. https://doi.org/ Yeung, C.H., Cooper, T.G., Oberpenning, F., Schulze, H., Nieschlag, E., 1993. Changes in
10.1016/j.mce.2016.03.005. movement characteristics of human spermatozoa along the length of the
Singh, A.P., Rajender, S., 2015. CatSper channel, sperm function and male fertility. Epididymis1. Biol. Reprod. 49, 274–280. https://doi.org/10.1095/
Reprod. Biomed. Online 30, 28–38. https://doi.org/10.1016/J.RBMO.2014.09.014. biolreprod49.2.274.
Smith, J.F., Syritsyna, O., Fellous, M., Serres, C., Mannowetz, N., Kirichok, Y., Lishko, P. Yuan, Y., Ding, X., Cheng, Y., Kang, H., Luo, T., Zhang, X., Kuang, H., Chen, Y., Zeng, X.,
V., 2013. Disruption of the principal, progesterone-activated sperm Ca2+ channel in Zhang, D., 2020. PFOA evokes extracellular Ca2+ influx and compromises
a CatSper2-deficient infertile patient. Proc. Natl. Acad. Sci. U.S.A. 110, 6823–6828. progesterone-induced response in human sperm. Chemosphere 241. https://doi.org/
https://doi.org/10.1073/pnas.1216588110. 10.1016/j.chemosphere.2019.125074.
Spehr, M., Gisselmann, G., Poplawski, A., Riffell, J.A., Wetzel, C.H., Zimmer, R.K.,
Hatt, H., 2003. Identification of a testicular odorant receptor mediating human
sperm chemotaxis. Science 299, 2054–2058. https://doi.org/10.1126/
science.1080376.

11
R. Rahban and S. Nef Molecular and Cellular Endocrinology xxx (xxxx) xxx

Zhang, X., Zeng, X., Lingle, C.J., 2006. Slo3 K + channels: voltage and pH dependence of Sensorineural deafness and male infertility: a contiguous gene deletion syndrome.
macroscopic currents. J. Gen. Physiol. 128, 317–336. https://doi.org/10.1085/ J. Med. Genet. 44, 233–240. https://doi.org/10.1136/jmg.2006.045765.
jgp.200609552. Zhang, Z., Liu, J., Meriano, J., Ru, C., Xie, S., Luo, J., Sun, Y., 2016. Human sperm
Zhang, Y., Malekpour, M., Al-Madani, N., Kahrizi, K., Zanganeh, M., Lohr, N.J., rheotaxis: a passive physical process. Sci. Rep. 6, 23553. https://doi.org/10.1038/
Mohseni, M., Mojahedi, F., Daneshi, A., Najmabadi, H., Smith, R.J.H., 2007. srep23553.

12

View publication stats

You might also like