You are on page 1of 13

Pflugers Arch - Eur J Physiol (2014) 466:819831

DOI 10.1007/s00424-014-1478-2

INVITED REVIEW

T-type Ca2+ channels in spermatogenic cells and sperm


Alberto Darszon & Arturo Hernndez-Cruz

Received: 3 February 2014 / Accepted: 8 February 2014 / Published online: 6 March 2014
# Springer-Verlag Berlin Heidelberg 2014

Abstract Cell function is importantly regulated by the intra- and initiate their maturation. Thereafter, in the female repro-
cellular concentration of Ca2+ ([Ca2+]i). Sperm development ductive tract, mammalian sperm continue maturing in a pro-
and function are deeply influenced by [Ca2+]i which is mod- cess called capacitation. This later process is needed so that
ulated amongst other ion transporters by plasma membrane sperm can respond to physiological inducers of the acrosome
Ca2+ permeable channels. The presence and role of voltage- reaction (AR) [71, 139]. In parallel or as a part of this matu-
dependent Ca2+ channels (CaV) of the T-type (CaV3) in sperm rational process, the sperm flagellar beat changes from an
physiology have become a matter of debate in recent years. active, symmetric mode to a more vigorous and asymmetric
Though they are functionally present in later stages of devel- one. This motility change is known as hyperactivation [63].
opment in spermatogenic cells and testicular sperm and their Once capacitated, sperm are ready to undergo the AR, a one-
mRNAs and proteins detected from spermatogenic cells to time exocytotic event. Where and when this reaction occurs
mature mammalian spermatozoa, their currents have not been has recently become a matter of debate [69]. For many years,
recorded in mature spermatozoa. This review critically sum- it had been thought that the eggs extracellular matrix, the
marizes the evidence for the involvement of CaV3 channels in zona pellucida (ZP), was the physiological inductor of the AR
sperm development and function. but recent experiments in mice spermatozoa and earlier ones
in other species have questioned this notion [11, 69].
Keywords Sperm acrosome reaction . Motility . As in most cells, the concentration of intracellular Ca2+
Capacitation . Voltage-dependent Ca2+ channels ([Ca2+]i) plays a key role in sperm signaling [19, 27, 67, 83,
128]. This divalent cation regulates motility, capacitation and
the AR. As CaV channels convert membrane potential (Em)
Introduction changes into [Ca2+]i signals, their involvement in sperm phys-
iology has been examined thoroughly. Two major functional
Generating a new individual requires a finely orchestrated classes of CaVs are known: high-voltage-activated (HVA,
interaction between sperm and egg in organisms with sexual CaV1 and CaV2) and low-voltage-activated (LVA, CaV3)
reproduction. Within the gonads, sperm and egg are produced channels. HVA channels require strong depolarizations to
open and inactivate slowly. Weaker depolarizations are need-
This article is published as part of the Special Issue on T-type (CaV3) ed to open LVA channels. They inactivate faster and at more
calcium channels in health and disease. negative potentials than HVA channels. CaV3 channels were
A. Darszon (*) initially called T-type due to their transient nature [41]. The
Departamento de Gentica del Desarrollo y Fisiologa Molecular, reader is referred to excellent reviews containing the detailed
Instituto de Biotecnologa, Universidad Nacional Autnoma de
description of the biophysical, biochemical and molecular
Mxico (UNAM, Campus Cuernavaca), Avenida Universidad
#2001, Col. Chamilpa, Cuernavaca, Morelos CP 62210, Mexico characteristics of these channels [98, 112, 145].
e-mail: darszon@ibt.unam.mx As for HVA channels, the 1 subunit contains the pore-
forming region of CaV3 channels (CaV3.1, CaV3.2 and
A. Hernndez-Cruz
CaV3.3; 1G, 1H and 1I, respectively) [97]. This subunit
Departamento de Neurociencia Cognitiva, Instituto de Fisiologa
Celular, UNAM (Campus Ciudad Universitaria), Mexico City, consisted of four homologous domains symmetrically ar-
Mexico ranged around the pore in a single polypeptide. Six
820 Pflugers Arch - Eur J Physiol (2014) 466:819831

transmembrane alpha helices (S1S6) form each of the four whole-cell patch-clamp currents recorded from mouse sper-
domains. The pore or P loop connecting S5 and S6 has a matogenic cells have the most salient features of somatic cell
conserved alpha helix and a glutamate that form the selectivity LVA currents [98] (see Fig. 2). They display low-voltage
filter [98]. thresholds for activation and inactivation and similar Ba2+
As stated earlier, sperm are tiny specialized cells of complex versus Ca2+ selectivity. Mibefradil, amiloride, pimozide,
morphology (Fig. 1) and whose plasma membrane is overall kurtoxin and Ni2+, as well as nifedipine and related 1,4-
rigid [26]. These characteristics precluded the in situ character- dihydropyridines, inhibit these currents at micromolar con-
ization of sperm ion channels by traditional electrophysiologi- centrations [3, 5, 93, 107].
cal approaches [reviewed in 25]. Also, with the exception of a
few reports [55, 73], researchers in the field still believe that CaV3.1 (1G) Northern blots revealed the presence of CaV3.1
spermatozoa are not able to synthesize proteins and thus mo- mRNAs in mouse and human testis [76], and RT-PCR exper-
lecular strategies such as interference RNA (RNAi) cannot be iments showed their mRNA in mouse and human spermato-
used. As alternative strategies to circumvent these drawbacks, genic cells and sperm [34, 76, 103]. Thereafter, CaV3.1 pro-
sperm Ca2+ channels were studied in vivo using Ca2+-sensitive tein was immunolocalized to the mouse and human sperm
and/or Em-sensitive fluorescent dyes and channel incorporation head and principal flagellar piece and to the human midpiece
into planar lipid bilayers. Furthermore, the presence of Ca2+ [125]. Using Ni2+ as a specific blocker, patch-clamp record-
channel messengers and proteins, and specifically, CaV3s, was ings on pachytene spermatocytes suggested that CaV3.2 is
examined using RT-PCR and specific antibodies. mainly responsible for their T-type currents with CaV3.1 also
Considering the above discussion, the ion channels present making a minor contribution [125]. Indeed, as currents re-
in mature sperm are most likely synthesized during spermato- corded from spermatogenic cells of CaV3.1 null mice are
genesis by their progenitor cells, the spermatogenic cells, indistinguishable from those of wild-type spermatozoa,
particularly during the more mature stages (pachytene sper- CaV3.2 channels are likely responsible for most of the T-
matocytes and round spermatids). These later cells are ame- type currents of these cells [118]. The presence of functional
nable for electrophysiology (Fig. 2) and molecular biology CaV 3.1 or CaV 3.2 channels in epididymal mouse sperm is
[26, 39, 67]. The presence of several CaV channel messengers still not fully settled [32, 125].
and proteins has been reported and reviewed recently [27].
Here, we will focus on CaV3 channels, their presence and CaV3.2 (1H) mRNAs encoding CaV3.2 subunits were de-
function in spermatogenic cells and in spermatozoa. In the tected in mouse testis by in situ hybridization and PCR exper-
following sections, we will describe the CaV3 messengers, iments [117], and CaV3.2 protein was immunolocalized to the
proteins and electrophysiological properties that have been head region overlying the acrosome [32, 125]. As mentioned
reported in these cells. above, CaV 3.1 and CaV 3.2 null mice experiments suggest
that CaV3.2 channels are mainly responsible for T-type cur-
rents of mouse spermatogenic cells [32, 118, 125]. In human
CaV3 channels in spermatogenic cells and sperm sperm, Jagannathan [66] found both CaV3.1 and CaV3.2 tran-
scripts, while Son [116] reported only CaV3.2 mRNA in testis
Before their molecular identification, T-type Ca2+ channels and Park [96] detected mRNAs encoding the three CaV3s.
had been biophysically characterized [97, 98]. The main CaV Only CaV3.2 immunostaining was faintly detected in the

Fig. 1 Sperm morphology.


Schematics of human sperm. The
different cell regions are indicated
Pflugers Arch - Eur J Physiol (2014) 466:819831 821

Fig. 2 Recording of T-type Ca2+ currents from mouse pachytene sper- were bathed in a solution containing (in millomolars) CaCl2 (2), NaCl
matocytes. Left: Representative family of Ca2+ currents recorded from (130), KCl (3), MgCl2 (2), NaHCO3 (1), NaH2PO4 (0.5), HEPES (5) and
pachytene spermatocytes obtained by the whole-cell patch-clamp tech- glucose (10) (pH 7.3/NaOH). The internal (patch pipette) solution
nique. The ionic currents were elicited by 200-ms depolarizing steps contained (in millimolars) CsMeSO3 (110), CsF (10), CsCl (15), EGTA
ranging from 90 to 10 mV from a holding potential of 90 mV with (10), HEPES (5); ATP-Mg2 (4) and phosphocreatine (10) (pH 7.3/CsOH).
steps increasing by 10 mV. The inward currents display the typical Right: Current-voltage (I-V) relationships of Ca2+ currents (n=11). The
voltage dependence and criss-cross pattern of T-type Ca2+ currents. values in the I-V plots are the meanS.E.M
Inward current amplitude was measured at the peak and normalized. Cells

human sperm head and a stronger signal was found in the sperm maturation, motility and the AR is under debate, T-type
flagellar midpiece [125]. Similar to rodent species, T-type currents from spermatogenic cells have been utilized to test
currents were recorded in spermatogenic cells from human the effects of compounds of clinical and biological relevance
biopsies [66]. in male fertility and reproduction [i.e. 6, 8, 9, 36, 130, 131].
For instance, gossypol, a male antifertility compound iso-
CaV3.3 (1I) CaV3.3 channel kinetics of activation, inactiva- lated from cotton, irreversibly inhibits T-type Ca2+ currents in
tion and recovery from inactivation are significantly slower spermatogenic cells in a dose- and time-dependent manner.
than those of CaV3.1 and CaV3.2 Ca2+ channels [97]. These Complete blockade occurs at 5 M gossypol [7]. Similarly,
characteristics are dissimilar from those of the T-type currents the extract of the root xylem of Tripterygium wilfordii and one
recorded from mouse spermatogenic cells. However, mRNAs of its monomers, demethylzeylasteral, inhibit T-type Ca2+
encoding the CaV3.3 subunits have been detected in mouse currents with an IC50 of 6.4 and 8.8 g/ml, respectively [6].
sperm [125]. Likewise, while CaV3.3 protein was reported to ZD7288, a blocker of hyperpolarization and cyclic nucleotide-
be present at the flagellar mouse midpiece, Stamboulian et al. gated (HCN) channels, inhibits spermatogenic T-type Ca2+
[118] were unable to find electrophysiological evidence for currents with an IC50 of 100 M and it also inhibits CaV3
their presence in mouse spermatogenic cells from either wild- channels heterologously expressed in HEK cells [36].
type or CaV3.1 null mice. In human testicular sperm, Son et al. Urocortin (UCN), a peptide related to the corticotropin-
[117] did not find CaV3.3 mRNAs; however, they were de- releasing factor (CRF) and present in the seminal fluid, in-
tected in spermatogenic cells [26]. In motile human sperm, hibits T-type Ca2+ currents with an IC50 of ~0.7 M. UCN
Park et al. [96] recognized small amounts of CaV3.3 mRNAs. inhibition of Ca2+ currents does not involve binding to the
CaV3.3 protein was immunolocalized to the human flagellar CRF receptor [123]. Fenvalerate, a type II pyrethroid insecti-
midpiece, similarly to mouse sperm [125]. cide, blocks spermatogenic T-type Ca2+ currents with an
Overall, the biophysical and pharmacological characteris- IC50 =0.25 M. Maximal inhibition (~40 %) is attained at a
tics of the voltage-dependent Ca2+ currents recorded from concentration of 1 M. [137]. Fenvalerate effects are slow and
mouse spermatogenic cells, including those obtained in the irreversible. 17-Estradiol (E2) significantly inhibits T-type
CaV3.1 and CaV3.2 null mice, are consistent with the notion Ca2+ currents in a dose-dependent manner (IC50 =8.9 M). At
that T-type Ca2+ currents result from the activity of channels 100 M, E2 produced maximal inhibition (53 %) and shifted
formed by the CaV3.2 isoform. These channels have been activation and inactivation curves by 5 mV in the negative
reported to be regulated by protein kinases [4], albumin [35] direction [87].
and cell adhesion molecule [86]. Even though the The protein tyrosine kinase inhibitor genistein, a phytoes-
involvement of CaV channels and specifically the CaV3s in trogen from soy, reversibly decreased spermatogenic T-type
822 Pflugers Arch - Eur J Physiol (2014) 466:819831

Ca2+ currents in a concentration-dependent manner (IC50 glass nanopipette. The patch-clamp electrode is scanned
22.7 M). Genistein inhibition does not involve TK activity and gathers topographic information of the cell surface
but rather acts directly, stabilizing the channel in the with greater spatial resolution than the optical microscope.
inactivated state. In transfected HEK293 cells, 50 M genis- This information allows positioning of the pipette onto the
tein inhibits CaV3.1 and CaV3.2 channels (by 43 and 52 %, cell surface with nanometer precision to obtain single-
respectively), but not CaV3.3 channels [123]. Ghrelin, a newly channel recordings from small cells and submicron cellu-
isolated peptide, also reversibly inhibits these currents in a lar structures inaccessible by conventional methods [77].
dose-dependent manner (IC50 =35 nM). Maximal inhibitory The spatial resolution of the smart patch-clamp is now
effect of T-type Ca2+ currents is 38 %. Inhibition was blocked able to resolve plasma membrane protein complexes
by an antagonist of the growth hormone secretagogue receptor [115].
1a (GHS-R1a), a G-protein coupled receptor expressed in 3. The perforated-cell patch-clamp technique to record ionic
spermatogenic cells and mature spermatozoa [85]. Ghrelin- currents in the whole-cell mode from the mature human
induced T-type Ca2+ current inhibition does not involve inter- sperm head was derived from work on cardiac myocytes
action of G subunits with the channels. The chloride chan- and neurons [104]. A saponin/-escin mixture is used in
nel blocker niflumic acid (NA) blocks T-type Ca2+ currents the pipette tip to induce controlled patch permeabilization.
with an IC50 of 73.5 M. NA also blocks CaV3 channels Stable whole-cell currents can be achieved after patch
expressed in HEK2h3 cells. NA-estimated IC50 values are perforation from dialyzed spermatozoa of several species
78, 192 and 137 M for CaV3.1, CaV3.2 and CaV3.3, respec- with the advantage of not requiring the presence of a
tively [8]. Finally, celecoxib (Cx), an anti-inflammatory drug cytoplasmic droplet. Using this strategy, a Ca2+-regulated
designed to inhibit cycloxygenase 2, has been shown to inhibit Cl channel was first characterized in human spermatozoa
T-type Ca2+ currents with an IC50 of 27 M. Surprisingly, Cx [94].
also induces intracellular Ca2+ rise, depolarizes membrane
potential and triggers the acrosome reaction [9]. Patch-clamp recording on the cytoplasmic droplet of tes-
ticular and epididymal mouse spermatozoa has revealed the
activity of several ion channels [27, 82, 88, 91, 100, 108, 138].
Sperm electrophysiology Examples are mouse and human CatSper [73] and mouse and
now human Slo3 channels [80, 90, 108, 111, 143]. Until now,
Even though in principle the ion channels that will end up in these are the only two sperm-specific channels whose genetic
mature spermatozoa could be studied in spermatogenic cells removal results in infertility. The CatSper cation channel has a
[3, 56, 81], we now know that they undergo drastic changes, tetrameric organization of four subunits (CatSper 14) plus
re-distribute and even functionally disappear during the last three auxiliary subunits till now (CatSper, CatSper,
stages of spermatogenesis [27, 114, 138]. Early on, electro- CatSper). It is Ca2+-permeable and potently regulated by
physiological recording of ion channel activity in the sperm pHi [83]. The Slo3 channel belongs to the Slo K+ channel
cell membrane was extremely difficult. Only 1 % of the family. Slo3 currents in mouse testicular and epididymal
attempts were successful [33, 50]. Swelling of sea urchin sperm are activated by intracellular alkalinization, depolariza-
sperm, by making sealing slightly easier than in normal sperm, tion and cAMP (testicular); blocked by 1 mM Ba2+ and high
allowed cell-attached patch-clamp recordings, but they were [TEA] (60 mM); and poorly K+-selective (PK+/PNa+ ~8), in a
still difficult to obtain and only lasted for a few minutes [106]. similar fashion as they are when heterologously expressed in
Fortunately, new strategies have been developed to study oocytes. These currents are absent in testicular and epididymal
sperm ion channels in mature cells in situ. Three possibilities sperm from Slo3 null mice, confirming their identity [108,
are now available: 140, 143]. Whole-sperm recordings have also revealed the
presence of a voltage-sensitive H+ channel involved in pHi
1. Whole-sperm patch-clamp recordings can be obtained regulation, more prevalent in human than in mouse sperm [74]
sealing on the cytoplasmic sperm droplet, a remnant of and of ATP-gated channels of the purinergic family, P2X2, in
the precursor germ cell cytoplasm located along the mouse epididymal sperm [92]. TRPM8-like currents, absent
midpiece of maturing sperm, that is shed around the time in sperm from TRPM8 null mice, have been recorded in
of ejaculation in mouse and other species but retained in testicular sperm [44, 89]. Pharmacological and functional
human spermatozoa [73, 82, 84]. experiments are consistent with the presence of a similar
2. The smart patch-clamp allows the simultaneous record- channel in human spermatozoa [29].
ing of the topography of the sperm surface and gigaseal The T-type Ca2+ currents were recorded from testicular
formation directly on mature sperm heads [23, 24, 47, 49, mouse sperm sensitive to micromolar concentrations of
68]. This approach combines scanning ion conductance mibefradil, nifedipine and Ni2+. These results confirmed pre-
microscopy and patch-clamp recording through a single dictions from expression patterns of CaV3 channels detected
Pflugers Arch - Eur J Physiol (2014) 466:819831 823

with specific antibodies with the presence of at least some condensation and a dramatic nuclear and cellular reorganiza-
CaVs in mature mouse sperm [27, 32, 125]. In addition, they tion. Morphological considerations have led to a classification
are consistent with earlier experimental work using pharma- of mouse spermiogenesis that involves 16 developmental
cological tools, [Ca2+]i imaging and AR determinations, sug- steps. They can be summarized as follows: round (steps 5
gesting the participation of CaV channels in the sperm AR. 8], elongating (steps 911), condensing (steps 1214) and
In spite of the fact that CaV3 currents similar to those condensed (steps 1416) spermatids. Early on, round sperma-
recorded in spermatogenic cells were detected in testicular tids are transcriptionally active and this activity decreases in
mouse spermatozoa, they have not been detected in epididymal elongating spermatids to finally turn off [120, 134].
sperm [27, 83, 138]. Even examination of the currents in sperm Spermatogenic cells are unable to differentiate alone; they
from CatSper and Slo3 null mice has not revealed the functional are nursed by Sertoli cells inside the tubules. Leydig cells from
presence of any CaVs [144]. These results question previous the interstitial compartment also provide hormonal stimula-
functional, molecular and pharmacological data indicating the tion [43]. Intriguingly, not much is known about the role of
involvement of CaVs in diverse sperm functions as they indicate Ca2+ during spermatogenesis. Spermatogenic cells, as all
that the CatSper channel is the principal Ca2+-conducting chan- cells, finely regulate their [Ca2+]i. For example, in rat sper-
nel in mammalian spermatozoa [83, 100, 144]. matogenic cells, high temperatures can induce programmed
There are puzzling and contrasting findings regarding the cell death which involves [Ca2+]i regulation [60]. On the other
presence of CaV channels in sperm (see Concluding re- hand, an external supply of oxidative glycolytic substrates
marks). Sound immunolocalization and functional evidence such as glucose and lactate differentially regulates [Ca2+]i in
recording [Ca2+]i increases triggered by K+-induced depolar- round spermatids and pachytene spermatocytes [101].
ization and the AR from several laboratories indicate the Our group recently reported that mice spermatogenic cells
presence of at least CaV3.2 and/or other HVA CaV channels undergo spontaneous [Ca2+]i oscillations that depend on extra-
such as CaV2.3 in mouse and human spermatozoa [32, 70, cellular Ca2+. These oscillations are partially inhibited by CaV3
103, 133, reviewed in 27]. Notably, the motility and [Ca2+]i Ca2+ channel blockers and Ni2+ [105]. It would be very impor-
increase induced by K+ depolarization are altered in sperm tant to fully characterize these spontaneous [Ca2+]i oscillations
from CaV2.3 null mice [103, 109]. However, whole-cell to determine if and how they influence spermatogenesis.
patch-clamp recording at the cytoplasmic droplet does not The physiological importance of CaVs in spermatogenesis
reveal any CaV currents. Single-channel recordings on the and steroidogenesis is underscored by the finding that CaV3
mammalian sperm head will be necessary to determine if channels are the main voltage-gated Ca2+ channels functional-
and which type of CaVs are present. ly expressed in meiotic and post-meiotic male germ cells.
Steroidogenesis is a complex multienzyme process by which
cholesterol is converted to biologically active steroid hormones
Possible roles of T-type Ca2+ (CaV3) channels not only in the adrenal cortex but also in the testis. Moreover,
in spermatogenesis and sperm function functional CaVs are required for normal germ cell develop-
ment: blocking of either CaV3 or CaV1 channels by their
Mammalian spermatogenesis respective blockers during mice prepubertal testicular matura-
tion results in abnormal spermatogenesis and steroidogenesis.
Spermatogenesis occurs inside the seminiferous tubules of the This alteration leads to male sterility, by arresting developing
testicle and involves the transformation of diploid spermatids and diminishing the Leydig cell population [78].
spematogonial stem cells into haploid spermatozoa. Commit-
ted spermatogonia proliferate (mitosis) and develop into sper- Motility
matocytes which through meiosis produce round spermatids.
After undergoing a morphological transformation called sper- In many species, flagellar propulsion is a key feature that
miogenesis, spermatids mature into spermatozoa. Spermato- allows spermatozoa to reach the egg and achieve fertilization.
genesis is a highly conserved process throughout vertebrate The ionic composition of the sperm environment and the
species that is hormonally controlled mainly by the hypothal- regulation of its ion plasma membrane permeability are funda-
amus and the pituitary gland. However, as spermatogenesis is mental for motility regulation. The flagellar propulsion ma-
a complex stage-specific, multifactorial process, a variety of chinery, the axoneme, is the site of Ca2+ regulation and thus
endocrine, paracrine and autocrine factors converge to control [Ca2+]i influences flagellar beat symmetry [16, 17]. Sperm
it [62]. from many marine species display chemotaxis, a motility
During spermiogenesis, the terminal differentiation process response that is dependent on external Ca2+ and encompasses
of spermatozoa, haploid round spermatids go through exten- a series of turns interspersed with periods of straight swimming
sive biochemical and morphological changes that include [27, 54, 72, 135]. In swimming sea urchin sperm, [Ca2+]i
acrosome formation, flagellar development, chromatin transients regulate chemotaxis [13, 136, reviewed in 27, 72].
824 Pflugers Arch - Eur J Physiol (2014) 466:819831

Immunolocalization and western blots using mammalian CaV to stimuli that induce the AR and to penetrate the egg invest-
antibodies suggested the presence of CaV1.2 and CaV12.3 in ments prior to fertilization [38, 127]. Cholesterol removal by
the flagella and acrosomal region of sea urchin sperm, respec- albumin, bicarbonate and Ca2+ are amongst the stringent
tively. Furthermore, CaV channel blockers, nifedipine and requirements of capacitation which lead to mammalian sperm
nimodipine, that inhibit the AR in this sperm species decrease plasma membrane reorganization, extensive changes in pro-
Ca2+ uptake caused by a K+-induced depolarization in tein phosphorylation state, increases in pHi and [Ca2+]i and
valinomycin-treated cells [48] Such Ca V blockers as plasma membrane hyperpolarization. A cAMP- and PKA-
nimodipine and micromolar Ni2+ also eliminate the [Ca2+]i dependent non-linear pathway regulates the phosphorylation
transients triggered by speract, a sperm motility regulator changes associated to capacitation which encompasses hyper-
[135]. The Strongylocentrotus purpuratus genome contains activation [reviewed in 26, 65, 127, 128].
CaV3 sequences; however, the presence of these proteins in In mouse sperm, it was proposed that plasma membrane
sea urchin sperm has not been demonstrated. On the other hyperpolarization removes CaV3 channel inactivation, leaving
hand, CaV channel blockers inhibit ascidian sperm motility Ca2+ channels in a state from which they could open upon
activation by an egg-derived factor which also induces chemo- induction of the AR by ZP3 (see the The AR section) [54].
taxis that is sensitive to a store-operated Ca2+ channel (SOC) This proposal is based on the assumption that CaV3 channels,
inhibitor [141]. which are functional in spermatogenic cells [3, 81] and also in
Ion channels have been implicated in the modulation of testicular sperm [88], are functional in mature spermatozoa.
sperm motility; however, only after sperm from CatSper null However, the latter is now debated and the role of this hyper-
mice were shown to be infertile and incapable of polarization is being re-examined.
hyperactivating was this established [20, 99]. Mammalian The mouse sperm hyperpolarization involves activation of
sperm can swim in two modes, activated and hyperactivated. K+ channels and inhibition of epithelial-like Na+ channels
Upon ejaculation, sperm display a relatively low-amplitude (ENaCs) [59]. The cystic fibrosis transmembrane conductance
flagellar beat called activated motility. In contrast, after capac- regulator (CFTR) has been suggested to activate during ca-
itation, a fraction of sperm swims in figure of eight trajec- pacitation and somehow mediate ENaC-type channel inhibi-
tories in low-viscosity medium, due to large-amplitude asym- tion contributing to the hyperpolarization [37]. Recent find-
metric flagellar beats. In the upper female reproductive tract, ings show that amiloride, an ENaC inhibitor, genistein, a
which has a higher viscosity, hyperactivated sperm are more CFTR agonist, and valinomycin, a K+ ionophore, are able to
progressive. Progression through the oviductal higher- mimic the capacitation-associated hyperpolarization. This
viscosity media and possibly egg cumulus penetration may membrane potential change enhances the ability of a K+-
require hyperactivated motility which is Ca2+-dependent [22]. induced depolarization or of soluble ZP to increase [Ca2+]i
The flagellar beat frequency of hyperactivated hamster sperm and induce the AR in non-capacitated sperm, although no
is correlated with transitory [Ca2+]i increases they display protein phosphorylation is promoted. These findings indicate
[121]. The redundant nuclear envelope, a reticular structure that sperm hyperpolarization by itself is the key to allow mice
at the flagellum neck, is an internal sperm Ca2+ store appar- sperm to acrosome react [30]. Future work will define if a
ently involved in hyperactivation. Stimulation of InsP3 recep- Ca2+ channel is involved in this process.
tors found in this store or of Ca2+ release from such internal CaV channels and particularly CaV3.2 could contribute to
stores with thimerosal or 4-AP induces hyperactivation [2, the rise in [Ca2+]i that accompanies capacitation. Notably,
63]. At this time, the presence of functional CaV3 channels bovine serum albumin (BSA) stimulates CaV3 currents from
in mammalian flagella is unlikely, as CaV3.1 immunolocal- mouse spermatogenic cells and shifts both their voltage-
ization results obtained in the mouse flagella [125] were not dependent steady-state activation and inactivation [56]. BSA
corroborated using sperm from CaV3.1 null mice [32], and no also increases the window current that results from the
Ca2+ currents have been detected in mouse epididymal sper- overlap of the steady-state activation and inactivation curves
matozoa [138], our unpublished recordings] nor in ejaculated of these CaV3 currents. As a result, a small persistent Ca2+
human sperm [83]. Single-channel recordings would corrob- influx through CaV3 channels could contribute to the [Ca2+]i
orate the absence of any CaV channels in the mammalian rise that occurs during capacitation [35].
sperm flagella.
The AR
Capacitation
The head of spermatozoa from many species contains a
After ejaculation, mammalian sperm need to mature within membrane-delimited organelle found overlying the sperm
the female tract to acquire the ability to fertilize the egg. This nucleus. Certain physiological or pharmacological stimuli
complex maturational process is called capacitation [128]. trigger the fusion of this organelle and the closely apposed
This non-linear set of events allows spermatozoa to respond plasma membrane at multiple sites, releasing the acrosomal
Pflugers Arch - Eur J Physiol (2014) 466:819831 825

contents to the extracellular media [18]. This single secretory ZP are consistent with those displayed by the CaV3 currents in
vesicle exocytosis process is called the AR and is essential for mouse spermatogenic cells [reviewed in 27, 67, 71].
fertilization in all sperm species possessing an acrosome. The Notably, for a long time, ZP has been considered the
transformation of the sperm plasma membrane associated to physiological inductor of the AR [132]. However, novel stud-
this reaction, which incorporates the inner acrosomal mem- ies using transgenic mice expressing different GFPs in the
brane, is required for penetrating the outer envelope of the egg sperm acrosome and mitochondria have shown that sperma-
and for fusion with its plasma membrane. External Ca2+ is tozoa undergo the AR before contacting the ZP during in vitro
essential for the AR, and certain ion channel blockers, includ- fertilization of cumulus-egg complexes [11, 69, 129]. These
ing those selective for CaV channels, interfere with the reac- findings pose new questions regarding the identity, location
tion [reviewed in 26, 42, 71]. and signaling pathway of the physiological inducer of the AR
Upon spawning, sea urchins release billions of their sperm and the ion channels involved in it. In spite of the fact that null
and eggs into the sea. Sperm interact with the outer layer of the mice for either CaV3.1 or CaV3.2 are fertile, as stated before, it
egg, the egg jelly, which triggers the AR. The egg jelly will be necessary to generate the CaV3.1/CaV3.2 double
contains a fucose sulfate polymer (FSP) that is the inducer knockout to completely rule out the participation of CaV3
of the AR [126]. FSP binds to suREJ1, a 210-kDa membrane channels in the mouse sperm AR.
glycoprotein, and triggers K+-dependent membrane potential Although evidence has been presented for the presence of
changes which involve increases in [Ca2+]i, [Na+]i and pHi CaV3 channel transcripts and protein in human sperm [66, 96,
[reviewed in 26]. The sea urchin sperm AR is triggered by at 125], and the ZP- and neoglycoprotein-induced AR pharma-
least two different Ca2+ channels [51, 53, 110]. A Ca2+-selec- cology displays similarities with that of CaV3 channels [10,
tive channel sensitive to verapamil and dihydropyridines is the 12, 14, 70, 124], their functional participation in these cells
first to transiently open when FSP binds to suREJ1. Its phar- has not been established.
macology is compatible with CaV channels. A second chan- As indicated earlier, progesterone is secreted by the
nel, insensitive to the latter blockers, opens 5 s later, is sensi- cumulus in the female reproductive tract. In human
tive to pHi, does not inactivate and results in the AR [52]. It is sperm, low-micromolar progesterone induces a rapid
likely that the second channel is of the SOC type [46]. Sperm [Ca2+]i increase followed by a plateau phase in sperm
are devoid of endoplasmic reticulum, so it is likely that the population and can trigger the AR [and reviewed in 58,
acrosome is the internal store which is known to contain InsP3 102]. This hormone is now known to potently activate
receptors [31, 142]. Furthermore, sea urchin sperm produce CatSper [83, 119]. [Ca2+]i imaging in single human
InsP3 during the AR [reviewed in 27]. sperm has revealed that progesterone induces an initial
In Lytechinus pictus sperm, FSP induces a transient K+- transient increase beginning at the sperm flagella and
dependent hyperpolarization that is likely due to K+ channels traveling towards the head [18, 79, 113]. In a fraction
and blocked by TEA [45]. It was proposed that this hyperpo- of the sperm population, progesterone can induce
larization could be necessary to remove inactivation from CaV [Ca2+]i oscillations [75], which has been shown to be
channels [45], as it was later proposed for mammalian sperm sensitive to Ca V 3 and ryanodine receptor channel
[40]. A CaV3 transcript is present in the S. purpuratus ge- blockers [1, 57].
nome, but protein expression in sea urchin sperm has not been Unfortunately, in pharmacological terms, CatSper is a pro-
demonstrated. miscuous channel with multiple ligands modulating its func-
Ca2+ influx is the key to the mammalian sperm AR, and tion [15]. It is sensitive to CaV3 channel blockers such as
several Ca2+-permeable channels have been proposed to par- mibefradil [15, 83] which complicates dissecting the contri-
ticipate. As in sea urchin sperm and other marine species, the bution of CaV3 channels in [Ca2+]i measurements.
mammalian sperm AR involves, after external Ca2+ uptake, CaV3 channels are subject to multiple forms of regulation
Ca2+ release from internal stores (the acrosome itself and that have been reviewed in detail [98, 112]. Of interest regard-
possibly residual nuclear vesicles found in the sperm neck ing the AR and the controversial role of CaV3 channels in
and midpiece) and the activation of store-operated Ca2+ chan- sperm physiology is the observation that CaM antagonists W7
nels [28, 46, 61, 95; reviewed in 18, 40]. and trifluoperazine inhibit mouse spermatogenic CaV3 cur-
The ZP, the extracellular coat surrounding the egg, and rents in a concentration-dependent manner [86]. Both com-
progesterone, which is present in the oocyte vicinity, are two pounds shift the voltage dependence of activation to more
physiologically relevant AR inducers that activate two or positive values and slow activation and inactivation kinetics.
more Ca2+-permeable channels most likely through distinct W5, a less potent homolog of W7 used as a control, does not
pathways [reviewed in 27]. Findings derived from studies perturb these currents. The inhibitory effect of W7 is Ca2+-
using Ca2+-sensitive fluorescent dyes suggested the involve- dependent, suggesting a role for the Ca2+/CaM complex in
ment of CaV channels in the ZP-induced AR. The pharmaco- this regulation. These currents were not altered by inhibitors
logical characteristics of the initial Ca2+ transient triggered by of CaM-activated phosphatase, CaMKII and protein kinase A.
826 Pflugers Arch - Eur J Physiol (2014) 466:819831

It is worth pointing out that the ZP-induced mouse sperm AR specific membrane resistivity of 20 kohm cm2, a flagellum of
and the transient [Ca2+]i increase associated to this reaction 0.5 m in diameter has a space constant () of ~500 m.
are blocked by the CaM inhibitors [86]. In retrospective, these Therefore, a 150-m-long sperm flagellum would be reason-
findings are still consistent with the possible participation of ably space-clamped. However, because of the reduced cyto-
CaV3 channels in the mouse sperm AR and call our attention plasmic space inside the flagella and its packing with axone-
to the need for a better understanding of the involvement of mal structural components, molecular mobility under such
CaM in the AR. Along this line, recent studies showed that crowded conditions is severely restricted compared with that
Ca2+ and CaM modulate CaV3.2/calcineurin interactions. in the typical cell cytoplasm. Fluorescence recovery after
Calcineurins phosphatase activity is decreased when it binds photo-bleaching (FRAP) experiments, using small fluorescent
to CaV3.2 which diminishes the channels current density tracers (calcein or carboxyfluorescein) in the sea urchin sperm
[64]. As calcineurin is required in the human sperm AR flagella, gave diffusion coefficients of ~60 m2/s, i.e., five
[21], CaM modulation would be consistent with the participa- times less than in aqueous media (~300 m2/s) [122]. Assum-
tion of CaV3.2 in mammalian sperm AR. From the informa- ing a proportional increase of intracellular resistivity in the
tion stated above, we would have to conclude that more work flagellum (500 ohm cm), then the space constant falls to
is needed to determine if CaV channels participate in the ~200 m, implying that near the tip of the mouse sperm
signaling cascades leading to the AR in mammalian sperm. flagellum (100150 m), membrane voltage is attenuated by
4050 % from that applied at the cytoplasmic droplet. Mem-
brane currents recorded from such distant, poorly clamped
Concluding remarks sources should be similarly attenuated. This situation gets
worst for the movement of small molecules between the
The findings of Zeng et al. [144] with spermatozoa from the flagellum and the head: the fluorescence recovery rate is >60
double CatSper and Slo3 null mouse suggest that during times slower in the head than in the flagellum, suggesting the
alkalinization, these two channels are the main and possibly presence of additional diffusion barriers around the neck
the only voltage- and pHi-activated cationic currents present region of the spermatozoa (and even stronger space-clamp
in the mouse corpus epididymal sperm principal piece. The- problems). Altogether, these arguments point out that caution
se authors state that at a lower pHi, their conclusion may not must be exercised still regarding the exact set of ion channels
hold. Non-capacitated sperm have a pHi of around 6.8 that determine their behaviour and influence fertilization.
which alkalinizes to ~7.27.4 after capacitation [reviewed Summarizing, there are still pending issues that need to be
in 128]. Small currents buried in the background pipette addressed to establish if CaVs, and particularly CaV3 channels,
leak conductance at lower pHi values could nonetheless do participate in the mammalian sperm acrosome reaction. In
have a profound influence on sperm physiology. As sper- this regard, it will be important to carry out single-channel
matozoa are small and have high input impedance, opening recordings in the sperm head to determine if CaVs are func-
or closing of a single channel might significantly contribute tionally present and if they participate in this important reac-
to their resting potential [50, 144]. Because of these consid- tion. Furthermore, it is very interesting and relevant to under-
erations and the fact that in the Zeng et al. experiments, stand why and how CaV3.2 channel expression gets turned off
sperm did not undergo other physiological changes associ- as testicular spermatozoa complete their maturation if it is the
ated to capacitation, which may alter ion channel behaviour, case and if it is possible that their expression gets turned on
one cannot rule out the presence and participation of a few again after capacitation just before the AR is triggered.
CaV channels or other cationic permeable channels in the Sperm research is undergoing revolutionary changes.
sperm plasma membrane particularly if they are located in Well-established paradigms such as the role of the zona
the head (see below). pellucida in inducing the sperm acrosome reaction are
In the voltage-clamp technique, current should be re- being debated and re-examined. Novel strategies to
corded ideally, from a membrane area of uniform potential, study ion channels are challenging to work with, but
i.e. from a population of channels experiencing the same this fascinating cells are revealing some of their unex-
voltage. Whole-cell patch-clamp is ideal for small round pected properties and will help to define the complete
cells, where good space clamp can be achieved. However, set of key ion transporters necessary for sperm matura-
in long processes such as the sperm flagella, the membrane tion, regulated swimming, capacitation and the AR.
potential distal to the recording electrode is not well Advances in temporal and spatial resolution of the
clamped, and therefore, current recorded by the voltage- available imaging techniques are unveiling mechanistic
clamp electrode combines local trans-membrane current details of where, when and how intracellular Ca2+ and
with axial currents originating from locations experiencing pHi profoundly influence sperm physiology. This new
different potentials. Assuming a specific intracellular resis- information is essential to advance our understanding of
tivity value of 100 ohm cm (similar to that of a neurite) and a reproduction, and species preservation and control.
Pflugers Arch - Eur J Physiol (2014) 466:819831 827

Acknowledgements This work was supported by the Direccin Gen- 14. Brandelli A, Miranda PV, Tezon JG (1996) Voltage-dependent
eral de Asuntos del Personal Acadmico; by the Universidad Nacional calcium channels and Gi regulatory protein mediate the human
Autnoma de Mxico (DGAPA-UNAM) grants IN225406-3 and sperm acrosomal exocytosis induced by N-acetylglucosaminyl/
IN222413 to to AD and AH-C, respectively; by the Consejo Nacional mannosyl neoglycoproteins. J Androl 17:522529, PMID: 8957696
de Ciencia y Tecnologa (CONACyT) grant 39908-Q to AD and RO3 15. Brenker C, Goodwin N, Weyand I, Kashikar ND, Naruse M,
TW 006121 to Pablo Visconti; and by the Secretara de Ciencia, Krhling M, Mller A, Kaupp UB, Strnker T (2012) The
Tecnologa e Innovacin del Distrito Federal (SECITI) grant 039/2013 CatSper channel: a polymodal chemosensor in human sperm.
to AHC and AD. EMBO J 31(7):16541665. doi:10.1038/emboj.2012.30, PMID:
22354039
16. Brokaw CJ (1979) Calcium-induced asymmetrical beating of triton-
demembranated sea urchin sperm flagella. J Cell Biol 82:401411,
References PMID: 479307
17. Brokaw CJ (1991) Calcium sensors in sea urchin sperm flagella.
Cell Motil Cytoskeleton 18:123130, PMID: 2013108
1. Aitken RJ, McLaughlin EA (2007) Molecular mechanisms of sperm 18. Buffone MG, Ijiri TW, Cao W, Merdiushev T, Aghajanian HK,
capacitation: progesterone-induced secondary calcium oscillations Gerton GL (2012) Heads or tails? Structural events and molecular
reflect the attainment of a capacitated state. Soc Reprod Fertil Suppl mechanisms that promote mammalian sperm acrosomal exocytosis
63:273293, PMID: 17566279 and motility. Mol Reprod Dev 79(1):418. doi:10.1002/mrd.21393,
2. Alasmari W, Costello S, Correia J, Oxenham SK, Morris J, PMID: 22031228
Fernandes L, Ramalho-Santos J, Kirkman-Brown J, Michelangeli 19. Carafoli E (2005) Calcium a universal carrier of biological signals.
F, Publicover S, Barratt CL (2013) Ca2+ signals generated by Delivered on 3 July 2003 at the Special FEBS Meeting in Brussels.
CatSper and Ca2+ stores regulate different behaviors in human FEBS J 272:10731089, PMID: 15720383
sperm. J Biol Chem 288(9):62486258. doi:10.1074/jbc.M112. 20. Carlson AE, Westenbroek RE, Quill T, Ren D, Clapham DE, Hille
439356, PMID: 23344959 B, Garbers DL, Babcock DF (2003) CatSper1 required for evoked
3. Arnoult C, Cardullo RA, Lemos JR, Florman HM (1996) Ca2+ entry and control of flagellar function in sperm. Proc Natl
Activation of mouse sperm T-type Ca2+ channels by adhesion to Acad Sci U S A 100(25):1486414868, PMID: 14657352
the egg zona pellucida. Proc Natl Acad Sci U S A 93:1300413009, 21. Castillo Bennett J, Roggero CM, Mancifesta FE, Mayorga LS
PMID: 8917534 (2010) Calcineurin-mediated dephosphorylation of synaptotagmin
4. Arnoult C, Lemos JR, Florman HM (1997) Voltage-dependent VI is necessary for acrosomal exocytosis. Biol Chem 285(34):
modulation of T-type calcium channels by protein tyrosine phos- 2626926278. doi:10.1074/jbc.M109.095752, PMID: 20551332
phorylation. EMBO J 16(7):15931599, PMID: 9130704 22. Chang H, Suarez SS (2010) Rethinking the relationship between
5. Arnoult C, Villaz M, Florman HM (1998) Pharmacological proper- hyperactivation and chemotaxis in mammalian sperm. Biol Reprod
ties of the T-type Ca2+ current of mouse spermatogenic cells. Mol 83(4):507513. doi:10.1095/biolreprod.109.083113, PMID:
Pharmacol 53:11041111, PMID: 9614215 2046335
6. Bai JP, Shi YL (2002) Inhibition of Ca(2+) channels in mouse 23. Cisneros-Mejorado A, Snchez D (2012) cGMP and cyclic
spermatogenic cells by male antifertility compounds from nucleotide-gated channels participate in mouse sperm capacitation.
Tripterygium wilfordii Hook. f. Contraception 65(6):441445, FEBS Lett 586:149153. doi:10.1016/j.febslet.2011.12.006,
PMID: 12127645 PMID: 22192355
7. Bai J, Shi Y (2002) Inhibition of T-type Ca(2+) currents in mouse 24. Cisneros-Mejorado A, Hernndez-Soberanis L, Islas-Carbajal MC,
spermatogenic cells by gossypol, an antifertility compound. Eur J Snchez D (2014) Capacitation and Ca(2+) influx in spermatozoa:
Pharmacol 440(1):16, PMID: 11959082 role of CNG channels and protein kinase G. Andrology 2(1):145
8. Balderas E, Ateaga-Tlecuitl R, Rivera M, Gomora JC, Darszon A 154. doi:10.1111/j.2047-2927.2013.00169.x, PMID: 24293181
(2012) Niflumic acid blocks native and recombinant T-type chan- 25. Darszon A, Labarca P, Nishigaki T, Espinosa F (1999) Ion channels
nels. J Cell Physiol 227(6):25422555. doi:10.1002/jcp.22992, in sperm physiology. Physiol Rev 79(2):481510, PMID: 10221988
PMID: 21898399 26. Darszon A, Nishigaki T, Wood C, Trevino CL, Felix R, Beltran C
9. Balderas E, Snchez-Crdenas C, Chvez JC, de la Vega Beltrn JL, (2005) Calcium channels and Ca2+ fluctuations in sperm physiolo-
Gmez-Lagunas F, Trevio CL, Darszon A (2013) The anti- gy. Int Rev Cytol 243:79172, PMID: 15797459
inflammatory drug celecoxib inhibits T-type Ca2+ currents in sper- 27. Darszon A, Nishigaki T, Beltran C, Trevio CL (2011) Calcium
matogenic cells yet it elicits the acrosome reaction in mature sperm. channels in the development, maturation, and function of sperma-
FEBS Lett 587(15):24122419. doi:10.1016/j.febslet.2013.05.068, tozoa. Physiol Rev 91(4):13051355. doi:10.1152/physrev.00028.
PMID: 23770093 2010, PMID: 22013213
10. Bhandari B, Bansal P, Talwar P, Gupta SK (2010) Delineation of 28. De Blas G, Michaut M, Trevio CL, Tomes CN, Yunes R, Darszon
downstream signaling components during acrosome reaction medi- A, Mayorga LS (2002) The intraacrosomal calcium pool plays a
ated by heat solubilized human zona pellucida. Reprod Biol direct role in acrosomal exocytosis. J Biol Chem 277(51):49326
Endocrinol 8:7. doi:10.1186/1477-7827-8-7 49331, PMID: 12379648
11. Bedford JM (2011) Site of the mammalian sperm physiological 29. De Blas GA, Darszon A, Ocampo AY, Serrano CJ, Castellano
acrosome reaction. Proc Natl Acad Sci USA. 22;108(12):4703- LE, Hernndez-Gonzlez EO, Chirinos M, Larrea F, Beltrn C,
4704. doi: 10.1073/pnas.1102296108. PMID: 21398585 Trevio CL (2009) TRPM8, a versatile channel in human sperm.
12. Blackmore PF, Eisoldt S (1999) The neoglycoprotein mannose- PLoS One 4(6):e6095. doi:10.1371/journal.pone.0006095,
bovine serum albumin, but not progesterone, activates T-type calci- PMID: 19582168
um channels in human spermatozoa. Mol Hum Reprod 5:498506, 30. De La Vega-Beltrn JL, Snchez-Crdenas C, Krapf D, Hernandez-
PMID: 10340995 Gonzlez EO, Wertheimer E, Trevio CL, Visconti PE, Darszon A
13. Bhmer M, Van Q, Weyand I, Hagen V, Beyermann M, Matsumoto (2012) Mouse sperm membrane potential hyperpolarization is nec-
M, Hoshi M, Hildebrand E, Kaupp UB (2005) Ca2+ spikes in the essary and sufficient to prepare sperm for the acrosome reaction. J
flagellum control chemotactic behavior of sperm. EMBO J 24(15): Biol Chem 287(53):4438444393. doi:10.1074/jbc.M112.393488,
27412752, PMID: 16001082 PMID: 23095755
828 Pflugers Arch - Eur J Physiol (2014) 466:819831

31. Domino SE, Garbers DL (1989) Stimulation of phospholipid turn- 46. Gonzalez-Martinez MT, Galindo BE, de De La Torre L, Zapata O,
over in isolated sea urchin sperm heads by the fucose-sulfate Rodriguez E, Florman HM, Darszon A (2001) A sustained increase
glycoconjugate that induces an acrosome reaction. Biol Reprod in intracellular Ca(2+) is required for the acrosome reaction in sea
41:133141, PMID: 2804203 urchin sperm. Dev Biol 236:220229, PMID: 11456456
32. Escoffier J, Boisseau S, Serres C, Chen CC, Kim D, Stamboulian S, 47. Gorelik J, Gu Y, Spohr HA, Shevchuk AI, Lab MJ, Harding SE,
Shin HS, Campbell KP, De Waard M, Arnoult C (2007) Expression, Edwards CR, Whitaker M, Moss GW, Benton DC, Sanchez D,
localization and functions in acrosome reaction and sperm motility Darszon A, Vodyanoy I, Klenerman D, Korchev YE (2002) Ion
of Ca(V)3.1 and Ca(V)3.2 channels in sperm cells: an evaluation channels in small cells and subcellular structures can be studied with
from Ca(V)3.1 and Ca(V)3.2 deficient mice. J Cell Physiol 212(3): a smart patch-clamp system. Biophys J 83:32963303, PMID:
753763, PMID: 17450521 12496097
33. Espinosa F, de la Vega-Beltrn JL, Lpez-Gonzlez I, 48. Granados-Gonzalez G, Mendoza-Lujambio I, Rodriguez E, Galindo
Delgado R, Labarca P, Darszon A (1998) Mouse sperm BE, Beltrn C, Darszon A (2005) Identification of voltage-
patch-clamp recordings reveal single Cl- channels sensitive dependent Ca2+ channels in sea urchin sperm. FEBS Lett 579(29):
to niflumic acid, a blocker of the sperm acrosome reaction. 66676672, PMID: 16307742
FEBS Lett 426(1):4751, PMID: 9598976 49. Gu Y, Kirkman-Brown JC, Korchev Y, Barratt CL, Publicover SJ
34. Espinosa F, Lpez-Gonzlez I, Serrano CJ, Gasque G, de la (2004) Multi-state, 4-aminopyridine-sensitive ion channels in hu-
Vega-Beltrn JL, Trevio CL, Darszon A (1999) Anion man spermatozoa. Dev Biol 274:308317, PMID: 15385161
channel blockers differentially affect T-type Ca(2+) currents 50. Guerrero A, Snchez JA, Darszon A (1987) Single-channel activity
of mouse spermatogenic cells, alpha1E currents expressed in in sea urchin sperm revealed by the patch-clamp technique. FEBS
Xenopus oocytes and the sperm acrosome reaction. Dev Lett 220:295298
Genet 25(2):103114, PMID: 10440844 51. Guerrero A, Darszon A (1989) Evidence for the activation of two
35. Espinosa F, Lpez-Gonzlez I, Muoz-Garay C, Felix R, De la different Ca2+ channels during the egg jelly-induced acrosome
Vega-Beltrn JL, Kopf GS, Visconti PE, Darszon A (2000) Dual reaction of sea urchin sperm. J Biol Chem 264:1959319599,
regulation of the T-type Ca(2+) current by serum albumin and beta- PMID: 2555326
estradiol in mammalian spermatogenic cells. FEBS Lett 475(3): 52. Guerrero A, Darszon A (1989) Egg jelly triggers a calcium influx
251256, PMID: 10869566 which inactivates and is inhibited by calmodulin antagonists in the
36. Felix R, Sandoval A, Snchez D, Gmora JC, De la Vega-Beltrn sea urchin sperm. Biochim Biophys Acta 980:109116, PMID:
JL, Trevio CL, Darszon A (2003) ZD7288 inhibits low-threshold 2923893
Ca(2+) channel activity and regulates sperm function. Biochem 53. Guerrero A, Garca L, Zapata O, Rodrguez E, Darszon A (1998)
Biophys Res Commun 311(1):187192, PMID: 14575712 Acrosome reaction inactivation in sea urchin sperm. Biochim
37. Figueiras-Fierro D, Acevedo JJ, Martnez-Lpez P, Escoffier J, Biophys Acta 1401(3):329338, PMID: 9540823
Seplveda FV, Balderas E, Orta G, Visconti PE, Darszon A 54. Guerrero A, Carneiro J, Pimentel A, Wood CD, Corkidi G, Darszon
(2013) Electrophysiological evidence for the presence of cystic A (2011) Strategies for locating the female gamete: the importance
fibrosis transmembrane conductance regulator (CFTR) in mouse of measuring sperm trajectories in three spatial dimensions. Mol
sperm. J Cell Physiol 228(3):590601. doi:10.1002/jcp.24166, Hum Reprod. (8):511-523 doi: 10.1093/molehr/gar042. PMID:
PMID: 22833409 21642645
38. Flesch FM, Gadella BM (2000) Dynamics of the mammalian sperm 55. Gur Y, Breitbart H (2006) Mammalian sperm translate nuclear-
plasma membrane in the process of fertilization. Biochim Biophys encoded proteins by mitochondrial-type ribosomes. Genes Dev
Acta 1469:197235, PMID: 1106388 20:411416, PMID: 16449571
39. Florman HM, Arnoult C, Kazam IG, Li C, O'Toole CM (1998) A 56. Hagiwara S, Kawa K (1984) Calcium and potassium currents in
perspective on the control of mammalian fertilization by egg- spermatogenic cells dissociated from rat seminiferous tubules. J
activated ion channels in sperm: a tale of two channels. Biol Physiol 356:135149, PMID: 6151599
Reprod 59:1216, PMID: 9674987 57. Harper CV, Barratt CL, Publicover SJ (2004) Stimulation of human
40. Florman HM, Jungnickel MK, Sutton KA (2008) Regulating the spermatozoa with progesterone gradients to simulate approach to
acrosome reaction. Int J Dev Biol 52(56):503510. doi:10.1387/ the oocyte. Induction of [Ca(2+)](i) oscillations and cyclical transi-
ijdb.082696hf, PMID: 18649263 tions in flagellar beating. J Biol Chem 279:4631546325
41. Fox AP, Nowycky MC, Tsien RW (1987) Single-channel recordings 58. Harper CV, Publicover SJ (2005) Reassessing the role of
of three types of calcium channels in chick sensory neurones. J progesterone in fertilizationcompartmentalized calcium sig-
Physiol 394:173200, PMID: 2451017 nalling in human spermatozoa? Hum Reprod 20:26752680,
42. Fukami K, Nakao K, Inoue T, Kataoka Y, Kurokawa M, PMID: 15980011
Fissore RA, Nakamura K, Katsuki M, Mikoshiba K, 59. Hernndez-Gonzlez EO, Sosnik J, Edwards J, Acevedo JJ,
Yoshida N, Takenawa T (2001) Requirement of phospholi- Mendoza-Lujambio I, Lpez-Gonzlez I, Demarco I, Wertheimer
pase Cdelta4 for the zona pellucida-induced acrosome reac- E, Darszon A, Visconti PE (2006) Sodium and epithelial sodium
tion. Science 292:920923, PMID: 11340203 channels participate in the regulation of the capacitation-associated
43. Ge R, Chen G, Hardy MP (2008) The role of the Leydig cell in hyperpolarization in mouse sperm. J Biol Chem 281(9):56235633,
spermatogenic function. Adv Exp Med Biol 636:255269. doi:10. PMID: 16407190
1007/978-0-387-09597-4_14, PMID: 19856172 60. Herrera E, Salas K, Lagos N, Benos DJ, Reyes JG (2001)
44. Gibbs GM, Orta G, Reddy T, Koppers AJ, Martnez-Lpez P, de la Temperature dependence of intracellular Ca2+ homeostasis in rat
Vega-Beltrn JL, Lo JC, Veldhuis N, Jamsai D, McIntyre P, Darszon meiotic and postmeiotic spermatogenic cells. Reproduction 122(4):
A, O'Bryan MK (2011) Cysteine-rich secretory protein 4 is an 545551, PMID: 11570961
inhibitor of transient receptor potential M8 with a role in establish- 61. Herrick SB, Schweissinger DL, Kim SW, Bayan KR, Mann S,
ing sperm function. Proc Natl Acad Sci U S A 108(17):70347039. Cardullo RA (2005) The acrosomal vesicle of mouse sperm is a
doi:10.1073/pnas.1015935108, PMID: 21482758 calcium store. J Cell Physiol 202(3):663671, PMID: 15389568
45. Gonzalez-Martinez M, Darszon A (1987) A fast transient hyperpo- 62. Hess RA, Renato de Franca L (2008) Spermatogenesis and cycle of
larization occurs during the sea urchin sperm acrosome reaction the seminiferous epithelium. Adv Exp Med Biol 636:115. doi:10.
induced by egg jelly. FEBS Lett 218:247250, PMID: 3595869 1007/978-0-387-09597-4_1, PMID: 19856159
Pflugers Arch - Eur J Physiol (2014) 466:819831 829

63. Ho HC, Suarez SS (2003) Characterization of the intracellular 80. Leonetti MD, Yuan P, Hsiung Y, Mackinnon R (2012) Functional
calcium store at the base of the sperm flagellum that regulates and structural analysis of the human SLO3 pH- and voltage-gated
hyperactivated motility. Biol Reprod 68:15901596, PMID: K+ channel. Proc Natl Acad Sci U S A 109(47):1927419279. doi:
12606347 10.1073/pnas.1215078109, PMID: 23129643
64. Huang CH, Chen YC, Chen CC (2013) Physical interaction be- 81. Lievano A, Santi CM, Serrano CJ, Trevino CL, Bellve AR,
tween calcineurin and CaV3.2 T-type Ca2+ channel modulates their Hernandez-Cruz A, Darszon A (1996) T-type Ca2+ channels and
functions. FEBS Lett 587(12):17231730. doi:10.1016/j.febslet. alpha1E expression in spermatogenic cells, and their possible rele-
2013.04.040, PMID: 23669360 vance to the sperm acrosome reaction. FEBS Lett 388:150154,
65. Ickowicz D, Finkelstein M, Breitbart H (2012) Mechanism of sperm PMID: 8690075
capacitation and the acrosome reaction: role of protein kinases. 82. Lishko PV, Kirichok Y (2010) The role of Hv1 and CatSper chan-
Asian J Androl 14(6):816821. doi:10.1038/aja.2012.81, PMID: nels in sperm activation. J Physiol 588(Pt 23):46674672. doi:10.
23001443 1113/jphysiol.2010.194142, PMID: 20679352
66. Jagannathan S, Punt EL, Gu Y, Arnoult C, Sakkas D, Barratt CL, 83. Lishko PV, Kirichok Y, Ren D, Navarro B, Chung JJ, Clapham DE
Publicover SJ (2002) Identification and localization of T-type (2012) The control of male fertility by spermatozoan ion channels.
voltage-operated calcium channel subunits in human male germ Annu Rev Physiol 74:453475. doi:10.1146/annurev-physiol-
cells. Expression of multiple isoforms. J Biol Chem 277:8449 020911-153258, PMID: 22017176
8456, PMID: 11751928 84. Lishko P, Clapham DE, Navarro B, Kirichok Y (2013) Sperm patch-
67. Jimnez-Gonzlez C, Michelangeli F, Harper CV, Barratt CL, clamp. Methods Enzymol 525:5983. doi:10.1016/B978-0-12-
Publicover SJ (2006) Calcium signalling in human spermatozoa: a 397944-5.00004-3, PMID: 23522465
specialized 'toolkit' of channels, transporters and stores. Hum 85. Liu K, Jiang D, Zhang T, Tao J, Shen L, Sun X (2011)
Reprod Update 12(3):253267, PMID: 16338990 Activation of growth hormone secretagogue type 1a receptor
68. Jimnez-Gonzlez MC, Gu Y, Kirkman-Brown J, Barratt CL, inhibits T-type Ca2+ channel currents through pertussis toxin-
Publicover S (2007) Patch-clamp 'mapping' of ion channel activity sensitive novel protein kinase C pathway in mouse spermato-
in human sperm reveals regionalisation and co localisation into genic cells. Cell Physiol Biochem 27(5):613624. doi:10.
mixed clusters. J Cell Physiol 213(3):801808, PMID: 17516540 1159/000329983, PMID: 21691079
69. Jin M, Fujiwara E, Kakiuchi Y, Okabe M, Satouh Y, Baba SA, 86. Lpez-Gonzlez I, De La Vega-Beltrn JL, Santi CM, Florman HM,
Chiba K, Hirohashi N (2011) Most fertilizing mouse spermatozoa Felix R, Darszon A (2001) Calmodulin antagonists inhibit T-type
begin their acrosome reaction before contact with the zona pellucida Ca(2+) currents in mouse spermatogenic cells and the zona
during in vitro fertilization. Proc Natl Acad Sci U S A 108(12): pellucida-induced sperm acrosome reaction. Dev Biol 236(1):210
48924896. doi:10.1073/pnas.1018202108, PMID: 21383182 219, PMID: 11456455
70. Jos O, Hernndez-Hernndez O, Chirinos M, Gonzlez- 87. Lu L, Wang CS, Gao XH, Wang J, Cheng J, Gao R, Xiao H (2008)
Gonzlez ME, Larrea F, Almanza A, Felix R, Darszon A, [17 beta-estradiol inhibits T-type calcium channels in mouse sper-
Trevio CL (2010) Recombinant human ZP3-induced sperm matogenic cells]. Zhonghua Nan Ke Xue 14(6):483488, Chinese.
acrosome reaction: evidence for the involvement of T- and PMID: 18649742
L-type voltage-gated calcium channels. Biochem Biophys 88. Martnez-Lpez P, Santi CM, Trevio CL, Ocampo-Gutirrez AY,
Res Commun 14(395):530534 Acevedo JJ, Alisio A, Salkoff LB, Darszon A (2009) Mouse sperm
71. Jungnickel MK, Sutton KA, Florman HM (2003) In the beginning: K+ currents stimulated by pH and cAMP possibly coded by Slo3
lessons from fertilization in mice and worms. Cell 114:401404, channels. Biochem Biophys Res Commun 381(2):204209. doi:10.
PMID: 12941269 1016/j.bbrc.2009.02.008, PMID: 19338774
72. Kaupp UB, Kashikar ND, Weyand I (2008) Mechanisms of sperm 89. Martnez-Lpez P, Trevio CL, de la Vega-Beltrn JL, De Blas G,
chemotaxis. Annu Rev Physiol 70:93117, PMID: 17988206 Monroy E, Beltrn C, Orta G, Gibbs GM, O'Bryan MK, Darszon A
73. Kirichok Y, Navarro B, Clapham DE (2006) Whole-cell patch- (2011) TRPM8 in mouse sperm detects temperature changes and
clamp measurements of spermatozoa reveal an alkaline-activated may influence the acrosome reaction. J Cell Physiol 226(6):1620
Ca2+ channel. Nature 439(7077):737740, PMID: 16467839 1631. doi:10.1002/jcp.22493, PMID: 21413020
74. Kirichok Y, Lishko PV (2011) Rediscovering sperm ion channels 90. Navarro B, Kirichok Y, Clapham DE (2007) KSper, a pH-sensitive
with the patch-clamp technique. Mol Hum Reprod 8:478499. doi: K1 current that controls sperm membrane potential. Proc Natl Acad
10.1093/molehr/gar044 Sci U S A 104:76887692, PMID: 17460039
75. Kirkman-Brown JC, Barratt CL, Publicover SJ (2004) Slow calci- 91. Navarro B, Kirichok Y, Chung JJ, Clapham DE (2008) Ion
um oscillations in human spermatozoa. Biochem J 378:827832, channels that control fertility in mammalian spermatozoa. Int
PMID: 14606954 J Dev Biol 52:607613. doi:10.1387/ijdb.072554bn, PMID:
76. Klugbauer N, Marais E, Lacinova L, Hofmann F (1999) A T-type 18649274
calcium channel from mouse brain. Pflugers Arch 437:710715, 92. Navarro B, Miki K, Clapham DE (2011) ATP-activated P2X2
PMID: 10087148 current in mouse spermatozoa. Proc Natl Acad Sci U S A
77. Korchev YE, Gorelik J, Lab MJ, Sviderskaya EV, Johnston CL, 108(34):1434214347. doi:10.1073/pnas.1111695108, PMID:
Coombes CR, Vodyanoy I, Edwards CR (2000) Cell volume mea- 21831833
surement using scanning ion conductance microscopy. Biophys J 93. Olamendi-Portugal T, Garca BI, Lpez-Gonzlez I, Van Der Walt J,
78:451457, PMID: 10620308 Dyason K, Ulens C, Tytgat J, Felix R, Darszon A, Possani LD
78. Lee JH, Ahn HJ, Lee SJ, Gye MC, Min CK (2011) Effects of L- and (2002) Two new scorpion toxins that target voltage-gated Ca2+ and
T-type Ca2(+) channel blockers on spermatogenesis and steroido- Na+ channels. Biochem Biophys Res Commun 299(4):562568,
genesis in the prepubertal mouse testis. J Assist Reprod Genet 1:23 PMID: 12459175
30. doi:10.1007/s10815-010-9480-x 94. Orta G, Ferreira G, Jos O, Trevio CL, Beltrn C, Darszon A
79. Lefivre L, Nash K, Mansell S, Costello S, Punt E, Correia J, Morris (2012) Human spermatozoa possess a calcium-dependent chloride
J, Kirkman-Brown J, Wilson SM, Barratt CL, Publicover S (2012) channel that may participate in the acrosomal reaction. J Physiol
2-APB-potentiated channels amplify CatSper-induced Ca(2+) sig- 590(11):26592675. doi:10.1113/jphysiol.2011.224485
nals in human sperm. Biochem J 448(2):189200. doi:10.1042/ 95. O'Toole CM, Arnoult C, Darszon A, Steinhardt RA, Florman HM
BJ20120339, PMID: 22943284 (2000) Ca(2+) entry through store-operated channels in mouse
830 Pflugers Arch - Eur J Physiol (2014) 466:819831

sperm is initiated by egg ZP3 and drives the acrosome reaction. Mol Reproduction 144(1):101109. doi:10.1530/REP-11-0268, PMID:
Biol Cell 5:15711584 22580372
96. Park JY, Ahn HJ, Gu JG, Lee KH, Kim JS, Kang HW, Lee JH 114. Serrano CJ, Trevino CL, Felix R, Darszon A (1999) Voltage-
(2003) Molecular identification of Ca2+ channels in human sperm. dependent Ca(2+) channel subunit expression and immunolocaliza-
Exp Mol Med 35:285292, PMID: 14508069 tion in mouse spermatogenic cells and sperm. FEBS Lett 462:171
97. Perez-Reyes E (2003) Molecular physiology of low-voltage- 176, PMID: 10580114
activated t-type calcium channels. Physiol Rev 83:117161, 115. Shevchuk AI, Frolenkov GI, Sanchez D, James PS, Freedman N,
PMID: 12506128 Lab MJ, Jones R, Klenerman D, Korchev YE (2006) Imaging
98. Perez-Reyes E, Lee JH (2013) Ins and outs of T-channel structure proteins in membranes of living cells by high-resolution scanning ion
function. Pflugers Arch. PubMed PMID: 24337909 conductance microscopy. Angew Chem Int Ed Engl 45:22122216
99. Ren D, Navarro B, Perez G, Jackson AC, Hsu S, Shi Q, Tilly JL, 116. Son WY, Lee JH, Han CT (2000) Acrosome reaction of human
Clapham DE (2001) A sperm ion channel required for sperm spermatozoa is mainly mediated by alpha1H T-type calcium chan-
motility and male fertility. Nature 413(6856):603609 nels. Mol Hum Reprod 6:893897, PMID: 11006317
100. Ren D, Xia J (2010) Calcium signaling through CatSper channels in 117. Son WY, Han CT, Lee JH, Jung KY, Lee HM, Choo YK (2002)
mammalian fertilization. Physiology (Bethesda) 25:165175. doi: Developmental expression patterns of alpha1H T-type Ca2+ chan-
10.1152/physiol.00049.2009, PMID: 20551230 nels during spermatogenesis and organogenesis in mice. Dev
101. Reyes JG, Herrera E, Lobos L, Salas K, Lagos N, Jorquera Growth Differ 44:181190, PMID: 12060068
RA, Labarca P, Benos DJ (2002) Dynamics of intracellular 118. Stamboulian S, Kim D, Shin HS, Ronjat M, De Waard M, Arnoult C
calcium induced by lactate and glucose in rat pachytene (2004) Biophysical and pharmacological characterization of spermato-
spermatocytes and round spermatids. Reproduction 123(5): genic T-type calcium current in mice lacking the CaV3.1 (alpha1G)
701710, PMID: 12006098 calcium channel: CaV3.2 (alpha1H) is the main functional calcium
102. Roldan ER, Murase T, Shi QX (1994) Exocytosis in spermatozoa in channel in wild-type spermatogenic cells. J Cell Physiol 200:116124
response to progesterone and zona pellucida. Science 266:1578 119. Strnker T, Goodwin N, Brenker C, Kashikar ND, Weyand I, Seifert
1581, PMID: 7985030 R, Kaupp UB (2011) The CatSper channel mediates progesterone-
103. Sakata Y, Saegusa H, Zong S, Osanai M, Murakoshi T, Shimizu Y, induced Ca2+ influx in human sperm. Nature 471(7338):382386.
Noda T, Aso T, Tanabe T (2002) Ca(v)2.3 (alpha1E) Ca2+ channel doi:10.1038/nature09769, PMID: 21412338
participates in the control of sperm function. FEBS Lett 516:229 120. Su W, Mruk DD, Cheng CY (2013) Regulation of actin dynamics
233, PMID: 11959138 and protein trafficking during spermatogenesisinsights into a
104. Sarantopoulos C, McCallum JB, Kwok WM, Hogan Q (2004) Beta- complex process. Crit Rev Biochem Mol Biol 48(2):153172. doi:
escin diminishes voltage-gated calcium current rundown in perfo- 10.3109/10409238.2012.758084, PMID: 23339542
rated patch-clamp recordings from rat primary afferent neurons. J 121. Suarez SS, Varosi SM, Dai X (1993) Intracellular calcium increases
Neurosci Methods 139(1):6168, PMID: 15351522 with hyperactivation in intact, moving hamster sperm and oscillates
105. Snchez-Crdenas C, Guerrero A, Trevio CL, Hernndez-Cruz A, with the flagellar beat cycle. Proc Natl Acad Sci U S A 90:4660
Darszon A (2012) Acute slices of mice testis seminiferous tubules 4664, PMID: 8506314
unveil spontaneous and synchronous Ca2+ oscillations in germ cell 122. Takao D, Kamimura S (2008) FRAP analysis of molecular diffusion
clusters. Biol Reprod. 18;87(4):92. doi: 10.1095/biolreprod.112. inside sea-urchin spermatozoa. J Exp Biol 211(Pt 22):35943600.
100255. PMID: 22914313 doi:10.1242/jeb.021923, PMID: 18978224
106. Sanchez D, Labarca P, Darszon A (2001) Sea urchin sperm cation- 123. Tao J, Wu Y, Chen J, Zhu H, Li S (2005) Effects of urocortin on T-
selective channels directly modulated by cAMP. FEBS Lett 503: type calcium currents in mouse spermatogenic cells. Biochem
111115, PMID: 11513865 Biophys Res Commun 329(2):743748, PMID: 15737649
107. Santi CM, Darszon A, Hernndez-Cruz A (1996) A 124. Tao J, Zhang Y, Li S, Sun W, Soong TW (2009) Tyrosine kinase-
dihydropyridine-sensitive T-type Ca2+ current is the main Ca2+ independent inhibition by genistein on spermatogenic T-type calci-
current carrier in mouse primary spermatocytes. Am J Physiol um channels attenuates mouse sperm motility and acrosome reac-
271:C1583C1593, PMID: 8944642 tion. Cell Calcium 45(2):133143. doi:10.1016/j.ceca.2008.07.004,
108. Santi CM, Martnez-Lpez P, de la Vega-Beltrn JL, Butler A, PMID: 18789523
Alisio A, Darszon A, Salkoff L (2010) The SLO3 sperm-specific 125. Trevio CL, Felix R, Castellano LE, Gutierrez C, Rodriguez D,
potassium channel plays a vital role in male fertility. FEBS Lett 584: Pacheco J, Lopez-Gonzalez I, Gomora JC, Tsutsumi V, Hernandez-
10411046. doi:10.1016/j.febslet.2010.02.005, PMID: 20138882 Cruz A, Fiordelisio T, Scaling AL, Darszon A (2004) Expression
109. Sakata Y, Saegusa H, Zong S, Osanai M, Murakoshi T, Shimizu Y, and differential cell distribution of low-threshold Ca(2+) channels in
Noda T, Aso T, Tanabe T (2001) Analysis of Ca(2+) currents in mammalian male germ cells and sperm. FEBS Lett 563:8792,
spermatocytes from mice lacking Ca(v)2.3 (alpha(1E)) Ca(2+) PMID: 15063728
channel. Biochem Biophys Res Commun 288:10321036, PMID: 126. Vacquier VD (1998) Evolution of gamete recognition proteins.
11689014 Science 281:19951998, PMID: 9748153
110. Schackmann RW (1989) Ionic regulation of the sea urchin sperm 127. Visconti PE, Kopf GS (1998) Regulation of protein phos-
acrosome reaction and stimulation by egg-derived peptides. In: phorylation during sperm capacitation. Biol Reprod 59:16,
Schatte H, Schatten G (eds) The cell biology of fertilization. PMID: 9674985
Academic, San Diego, pp 328 128. Visconti PE, Krapf D, de la Vega-Beltrn JL, Acevedo JJ, Darszon
111. Schreiber M, Wei A, Yuan A, Gaut J, Saito M, Salkoff L (1998) A (2011) Ion channels, phosphorylation and mammalian sperm
Slo3, a novel pH-sensitive K+ channel from mammalian spermato- capacitation. Asian J Androl 13(3):395405. doi:10.1038/aja.
cytes. J Biol Chem 273:35093516, PMID: 9452476 2010.69, PMID: 21540868
112. Senatore A, Zhorov BS, Spafford JD (2012) Cav3 Ttype calcium 129. Visconti PE (2012) Sperm bioenergetics in a nutshell. Biol Reprod.
channels. WIREs Membr Transp Signal 1:467491. doi:10.1002/ 28;87(3):72. PMID: 22914312
wmts.41 130. Wang CS, Gao XH, Cheng H, Gong W, Zhang XC, Cheng J, Gao R,
113. Servin-Vences MR, Tatsu Y, Ando H, Guerrero A, Yumoto N, Xiao H (2006) [Effects of flunarizine on T-type calcium channels in
Darszon A, Nishigaki T (2012) A caged progesterone analog alters mouse spermatogenic cells]. ZhonghuaNan Ke Xue. 12(7):594-597,
intracellular Ca 2+ and flagellar bending in human sperm. 601. Chinese. PMID: 16894933
Pflugers Arch - Eur J Physiol (2014) 466:819831 831

131. Wang Q, Lu L, Gao X, Wang C, Wang J, Cheng J, Gao R, Xiao H Endocrinol. Oct 27;7:119. doi: 10.1186/1477-7827-7-119. PMID:
(2011) Effects of raloxifene on voltage-dependent T-type Ca2+ 19860887
channels in mouse spermatogenic cells. Pharmacology 87(12): 139. Yanagimachi R (1994) Mammalian fertilization. In: Knobile E,
7080. doi:10.1159/000321726, PMID: 21228613 Neill JD (eds) The physiology of reproduction, vol 1. Raven, New
132. Wassarman PM, Litscher ES (2008) Mammalian fertilization is York, pp 189317
dependent on multiple membrane fusion events. Methods Mol Biol 140. Yang C, Zeng XH, Zhou Y, Xia XM, Lingle CJ (2011) LRRC52
475:99113. doi:10.1007/978-1-59745-250-2_6, PMID: 18979240 (leucine-rich-repeat-containing protein 52), a testis-specific auxilia-
133. Wennemuth G, Westenbroek RE, Xu T, Hille B, Babcock DF ry subunit of the alkalization-activated Slo3 channel. Proc Natl
(2000) CaV2.2 and CaV2.3 (N- and R-type) Ca2+ channels in Acad Sci U S A 108(48):1941919424. doi:10.1073/pnas.
depolarization-evoked entry of Ca2+ into mouse sperm. J Biol 1111104108, PMID: 22084117
Chem 275(28):2121021217, PMID: 10791962 141. Yoshida M (2003) Mechanism of chemotaxis of the ascidian sper-
134. Wolgemuth DJ, Manterola M, Vasileva A (2013) Role of cyclins in matozoa. Zoolog Sci 20:14991500, PMID: 14960802
controlling progression of mammalian spermatogenesis. Int J Dev 142. Zapata O, Ralston J, Beltran C, Parys JB, Chen JL, Longo FJ,
Biol 57(24):159168. doi:10.1387/ijdb.130047av, PMID: Darszon A (1997) Inositol triphosphate receptors in sea urchin
23784826 sperm. Zygote 5:355364, PMID: 9563683
135. Wood CD, Darszon A, Whitaker M (2003) Speract induces calcium 143. Zeng XH, Yang C, Kim ST, Lingle CJ, Xia XM (2011) Deletion of
oscillations in the sperm tail. J Cell Biol 161(1):89101, PMID: the Slo3 gene abolishes alkalization-activated K+ current in mouse
12695500 spermatozoa. Proc Natl Acad Sci U S A 108(14):58795884. doi:
136. Wood CD, Nishigaki T, Furuta T, Baba SA, Darszon A (2005) Real- 10.1073/pnas.1100240108, PMID: 21427226
time analysis of the role of Ca(2+) in flagellar movement and 144. Zeng XH, Navarro B, Xia XM, Clapham DE, Lingle CJ (2013)
motility in single sea urchin sperm. J Cell Biol 169:725731, Simultaneous knockout of Slo3 and CatSper1 abolishes all
PMID: 15928204 alkalization- and voltage-activated current in mouse spermatozoa.
137. Xiao H, Zhang XC, Zhang L, Dai XQ, Gong W, Cheng J, Gao R, J Gen Physiol 142(3):305313. doi:10.1085/jgp.201311011, PMID:
Wang X (2006) Fenvalerate modifies T-type Ca2+ channels in 23980198
mouse spermatogenic cells. Reprod Toxicol 21(1):4853, PMID: 145. Zhang Y, Jiang X, Snutch TP, Tao J (2013) Modulation of low-
16039827 voltage-activated T-type Ca2+ channels. Biochim Biophys Acta
138. Xia J, Ren D (2009) The BSA-induced Ca2+ influx during sperm 1828(7):15501559. doi:10.1016/j.bbamem.2012.08.032, PMID:
capacitation is CATSPER channel-dependent. Reprod Biol 22975282

You might also like