Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Celia M. Santi is active.

Publication


Featured researches published by Celia M. Santi.


Neuron | 2003

The Sodium-Activated Potassium Channel Is Encoded by a Member of the Slo Gene Family

Alex Yuan; Celia M. Santi; Aguan Wei; Zhao-Wen Wang; Kelly Pollak; Michael L. Nonet; Leonard K. Kaczmarek; C. Michael Crowder; Lawrence Salkoff

Na(+)-activated potassium channels (K(Na)) have been identified in cardiomyocytes and neurons where they may provide protection against ischemia. We now report that K(Na) is encoded by the rSlo2 gene (also called Slack), the mammalian ortholog of slo-2 in C. elegans. rSlo2, heterologously expressed, shares many properties of native K(Na) including activation by intracellular Na(+), high conductance, and prominent subconductance states. In addition to activation by Na(+), we report that rSLO-2 channels are cooperatively activated by intracellular Cl(-), similar to C. elegans SLO-2 channels. Since intracellular Na(+) and Cl(-) both rise in oxygen-deprived cells, coactivation may more effectively trigger the activity of rSLO-2 channels in ischemia. In C. elegans, mutational and physiological analysis revealed that the SLO-2 current is a major component of the delayed rectifier. We demonstrate in C. elegans that slo-2 mutants are hypersensitive to hypoxia, suggesting a conserved role for the slo-2 gene subfamily.


FEBS Letters | 2010

The SLO3 sperm-specific potassium channel plays a vital role in male fertility

Celia M. Santi; Pablo Martínez-López; José Luis de la Vega-Beltrán; Alice Butler; Arturo Alisio; Alberto Darszon; Lawrence Salkoff

Here we show a unique example of male infertility conferred by a gene knockout of the sperm‐specific, pH‐dependent SLO3 potassium channel. In striking contrast to wild‐type sperm which undergo membrane hyperpolarization during capacitation, we found that SLO3 mutant sperm undergo membrane depolarization. Several defects in SLO3 mutant sperm are evident under capacitating conditions, including impaired motility, a bent “hairpin” shape, and failure to undergo the acrosome reaction (AR). The failure of AR is rescued by valinomycin which hyperpolarizes mutant sperm. Thus SLO3 is the principal potassium channel responsible for capacitation‐induced hyperpolarization, and membrane hyperpolarization is crucial to the AR.


The Journal of Neuroscience | 2006

Opposite Regulation of Slick and Slack K+ Channels by Neuromodulators

Celia M. Santi; Gonzalo Ferreira; Bo Yang; Valeswara Rao Gazula; Alice Butler; Aguan Wei; Leonard K. Kaczmarek; Lawrence Salkoff

Slick (Slo2.1) and Slack (Slo2.2) are two novel members of the mammalian Slo potassium channel gene family that may contribute to the resting potentials of cells and control their basal level of excitability. Slo2 channels have sensors that couple channel activity to the intracellular concentrations of Na+ and Cl− ions (Yuan et al., 2003). We now report that activity of both Slo2 channels is controlled by neuromodulators through Gαq-protein coupled receptors (GqPCRs) (the M1 muscarinic receptor and the mGluR1 metabotropic glutamate receptor). Experiments coexpressing channels and receptors in Xenopus oocytes show that Slo2.1 and Slo2.2 channels are modulated in opposite ways: Slo2.1 is strongly inhibited, whereas Slo2.2 currents are strongly activated through GqPCR stimulation. Differential regulation involves protein kinase C (PKC); application of the PKC activator PMA, to cells expressing channels but not receptors, inhibits Slo2.1 whole-cell currents and increases Slo2.2 currents. Synthesis of a chimera showed that the distal carboxyl region of Slo2.1 controls the sensitivity of Slo2.1 to PMA. Slo2 channels have widespread expression in brain (Bhattacharjee et al., 2002, 2005). Using immunocytochemical techniques, we show coexpression of Slo2 channels with the GqPCRs in cortical and hippocampal brain sections and in cultured hippocampal neurons. The differential control of these novel channels by neurotransmitters may elicit long-lasting increases or decreases in neuronal excitability and, because of their widespread distribution, may provide a mechanism to activate or repress electrical activity in many systems of the brain.


Biochemical and Biophysical Research Communications | 2009

Mouse sperm K+ currents stimulated by pH and cAMP possibly coded by Slo3 channels

Pablo Martínez-López; Celia M. Santi; Claudia L. Treviño; Ana Y. Ocampo-Gutiérrez; Juan José Acevedo; Arturo Alisio; Lawrence Salkoff; Alberto Darszon

Slo3 channels belong to the high conductance Slo K+ channel family. They are activated by voltage and intracellular alkalinization, and have a K+/Na+ permeability ratio (PK/PNa) of only approximately 5. Slo3 channels have only been found in mammalian sperm. Here we show that Slo3 channels expressed in Xenopus oocytes are also stimulated by elevated cAMP levels through PKA dependent phosphorylation. Capacitation, a maturational process required by mammalian sperm to enable them to fertilize eggs, involves intracellular alkalinization and an increase in cAMP. Our mouse sperm patch clamp recordings have revealed a K+ current that is time and voltage dependent, is activated by intracellular alkalinization, has a PK/PNa > or = 5, is weakly blocked by TEA and is very sensitive to Ba2+. This current is also stimulated by cAMP. All of these properties match those displayed by heterologously expressed Slo3 channels, suggesting that the native current we observe in sperm is indeed carried by Slo3 channels.


Zygote | 1998

Localisation of inositol trisphosphate and ryanodine receptors during mouse spermatogenesis: possible functional implications

Claudia L. Treviño; Celia M. Santi; Carmen Beltrán; Arturo Hernández-Cruz; Alberto Darszon; Hilda Lomelí

During spermatogenesis the activity of intracellular Ca(2+)-release channels is likely to play an important role in different specific cellular functions. Accordingly, messenger RNAs for the three inositol 1,4,5-trisphosphate receptor (IP3R) subtypes were found to be present throughout spermatogenesis. Immunocytochemical analysis revealed distinct distribution patterns of the mature IP3Rs during sperm differentiation. At early stages, IP3Rs are distributed throughout the cytoplasm, and as differentiation proceeds they become selectively localised to the Golgi complex. Consistently, spermatogonia underwent large intracellular Ca2+ release in response to thapsigargin (TG), while smaller responses were detected in late spermatocytes and spermatids. The distribution of IP3Rs and the larger Ca(2+)-release responses found in spermatogonia, suggest that IP3Rs may be involved in cell proliferation at this stage. This notion is supported by our observations in a spermatogenic cell line that depletion of intracellular Ca2+ pools using TG inhibits cell division, and that incubation with an IP3R-I antisense oligonucleotide completely inhibited proliferation. Furthermore, the three genes encoding ryanodine receptor proteins (RyRs) are expressed at all stages of spermatogenesis. However, immunocytochemical studies with specific antibodies against each of the RyR subtypes detected types 1 and 3 in spermatogenic cells and only type 3 in mature sperm. In contrast to IP3Rs, RyRs remain scattered in the cytoplasm throughout differentiation. Functional responses to caffeine and ryanodine were absent in spermatogenic cells and in mature sperm. These findings suggest that IP3Rs have significantly more important roles in spermatogenesis than RyRs, and that one of these roles is crucial for cell proliferation.


Molecular Human Reproduction | 2014

Membrane Hyperpolarization during Human Sperm Capacitation

Ignacio López-González; Paulina Torres-Rodríguez; O. Sánchez-Carranza; A. Solís-López; Celia M. Santi; Alberto Darszon; Claudia L. Treviño

Sperm capacitation is a complex and indispensable physiological process that spermatozoa must undergo in order to acquire fertilization capability. Spermatozoa from several mammalian species, including mice, exhibit a capacitation-associated plasma membrane hyperpolarization, which is necessary for the acrosome reaction to occur. Despite its importance, this hyperpolarization event has not been adequately examined in human sperm. In this report we used flow cytometry to show that a subpopulation of human sperm indeed undergo a plasma membrane hyperpolarization upon in vitro capacitation. This hyperpolarization correlated with two other well-characterized capacitation parameters, namely an increase in intracellular pH and Ca(2+) concentration, measured also by flow cytometry. We found that sperm membrane hyperpolarization was completely abolished in the presence of a high external K(+) concentration (60 mM), indicating the participation of K(+) channels. In order to identify, which of the potential K(+) channels were involved in this hyperpolarization, we used different K(+) channel inhibitors including charybdotoxin, slotoxin and iberiotoxin (which target Slo1) and clofilium (a more specific blocker for Slo3). All these K(+) channel antagonists inhibited membrane hyperpolarization to a similar extent, suggesting that both members of the Slo family may potentially participate. Two very recent papers recorded K(+) currents in human sperm electrophysiologically, with some contradictory results. In the present work, we show through immunoblotting that Slo3 channels are present in the human sperm membrane. In addition, we found that human Slo3 channels expressed in CHO cells were sensitive to clofilium (50 μM). Considered altogether, our data indicate that Slo1 and Slo3 could share the preponderant role in the capacitation-associated hyperpolarization of human sperm in contrast to what has been previously reported for mouse sperm, where Slo3 channels are the main contributors to the hyperpolarization event.


PLOS ONE | 2013

Ion permeabilities in mouse sperm reveal an external trigger for SLO3-dependent hyperpolarization.

Julio C. Chávez; José Luis de la Vega-Beltrán; Jessica Escoffier; Pablo E. Visconti; Claudia L. Treviño; Alberto Darszon; Lawrence Salkoff; Celia M. Santi

Unlike most cells of the body which function in an ionic environment controlled within narrow limits, spermatozoa must function in a less controlled external environment. In order to better understand how sperm control their membrane potential in different ionic conditions, we measured mouse sperm membrane potentials under a variety of conditions and at different external K+ concentrations, both before and after capacitation. Experiments were undertaken using both wild-type, and mutant mouse sperm from the knock-out strain of the sperm-specific, pH-sensitive, SLO3 K+ channel. Membrane voltage data were fit to the Goldman-Hodgkin-Katz equation. Our study revealed a significant membrane permeability to both K+ and Cl− before capacitation, as well as Na+. The permeability to both K+ and Cl− has the effect of preventing large changes in membrane potential when the extracellular concentration of either ion is changed. Such a mechanism may protect against undesired shifts in membrane potential in changing ionic environments. We found that a significant portion of resting membrane potassium permeability in wild-type sperm was contributed by SLO3 K+ channels. We also found that further activation of SLO3 channels was the essential mechanism producing membrane hyperpolarization under two separate conditions, 1) elevation of external pH prior to capacitation and 2) capacitating conditions. Both conditions produced a significant membrane hyperpolarization in wild-type which was absent in SLO3 mutant sperm. Hyperpolarization in both conditions may result from activation of SLO3 channels by raising intracellular pH; however, demonstrating that SLO3-dependent hyperpolarization is achieved by an alkaline environment alone shows that SLO3 channel activation might occur independently of other events associated with capacitation. For example sperm may undergo stages of membrane hyperpolarization when reaching alkaline regions of the female genital tract. Significantly, other events associated with sperm capacitation, occur in SLO3 mutant sperm and thus proceed independently of hyperpolarization.


Journal of Biological Chemistry | 2014

SLO3 K+ Channels Control Calcium Entry through CATSPER Channels in Sperm

Julio C. Chávez; Juan Ferreira; Alice Butler; José Luis De La Vega Beltrán; Claudia L. Treviño; Alberto Darszon; Lawrence Salkoff; Celia M. Santi

Background: SLO3 and CATSPER are two sperm-specific ion channels. Results: SLO3 K+ channels control Ca2+ entry through CATSPER channels. Conclusion: SLO3 control of CATSPER channel activity involves an intermediary step in which SLO3-dependent hyperpolarization may elicit internal alkalization via a voltage-dependent mechanism. Significance: Understanding the control of Ca2+ entry in sperm is crucial to understanding fertility; this study also reveals an unusual role for a K+ channel. Here we show how a sperm-specific potassium channel (SLO3) controls Ca2+ entry into sperm through a sperm-specific Ca2+ channel, CATSPER, in a totally unanticipated manner. The genetic deletion of either of those channels confers male infertility in mice. During sperm capacitation SLO3 hyperpolarizes the sperm, whereas CATSPER allows Ca2+ entry. These two channels may be functionally connected, but it had not been demonstrated that SLO3-dependent hyperpolarization is required for Ca2+ entry through CATSPER channels, nor has a functional mechanism linking the two channels been shown. In this study we show that Ca2+ entry through CATSPER channels is deficient in Slo3 mutant sperm lacking hyperpolarization; we also present evidence supporting the hypothesis that SLO3 channels activate CATSPER channels indirectly by promoting a rise in intracellular pH through a voltage-dependent mechanism. This mechanism may work through a Na+/H+ exchanger (sNHE) and/or a bicarbonate transporter, which utilizes the inward driving force of the Na+ gradient, rendering it intrinsically voltage-dependent. In addition, the sperm-specific Na+/H+ exchanger (sNHE) possess a putative voltage sensor that might be activated by membrane hyperpolarization, thus increasing the voltage sensitivity of internal alkalization.


Proceedings of the National Academy of Sciences of the United States of America | 2003

Dissection of K+ currents in Caenorhabditis elegans muscle cells by genetics and RNA interference.

Celia M. Santi; Alex Yuan; Gloria L. Fawcett; Zhao-Wen Wang; Alice Butler; Michael L. Nonet; Aguan Wei; P. Rojas; Lawrence Salkoff

GFP-promoter experiments have previously shown that at least nine genes encoding potassium channel subunits are expressed in Caenorhabditis elegans muscle. By using genetic, RNA interference, and physiological techniques we revealed the molecular identity of the major components of the outward K+ currents in body wall muscle cells in culture. We found that under physiological conditions, outward current is dominated by the products of only two genes, Shaker (Kv1) and Shal (Kv4), both expressing voltage-dependent potassium channels. Other channels may be held in reserve to respond to particular circumstances. Because GFP-promoter experiments indicated that slo-2 expression is prominent, we created a deletion mutant to identify the SLO-2 current in vivo. In both whole-cell and single-channel modes, in vivo SLO-2 channels were active only when intracellular Ca2+ and Cl- were raised above normal physiological conditions, as occurs during hypoxia. Under such conditions, SLO-2 is the largest outward current, contributing up to 87% of the total current. Other channels are present in muscle, but our results suggest that they are unlikely to contribute a large outward component under physiological conditions. However, they, too, may contribute currents conditional on other factors. Hence, the picture that emerges is of a complex membrane with a small number of household conductances functioning under normal circumstances, but with additional conductances that are activated during unusual circumstances.


Journal of Biological Chemistry | 2006

Mutant Analysis of the Shal (Kv4) Voltage-gated Fast Transient K+ Channel in Caenorhabditis elegans

Gloria L. Fawcett; Celia M. Santi; Alice Butler; Thanawath Harris; Manuel Covarrubias; Lawrence Salkoff

Shal (Kv4) α-subunits are the most conserved among the family of voltage-gated potassium channels. Previous work has shown that the Shal potassium channel subfamily underlies the predominant fast transient outward current in Drosophila neurons (Tsunoda, S., and Salkoff, L. (1995) J. Neurosci. 15, 1741–1754) and the fast transient outward current in mouse heart muscle (Guo, W., Jung, W. E., Marionneau, C., Aimond, F., Xu, H., Yamada, K. A., Schwarz, T. L., Demolombe, S., and Nerbonne, J. M. (2005) Circ. Res. 97, 1342–1350). We show that Shal channels also play a role as the predominant transient outward current in Caenorhabditis elegans muscle. Green fluorescent protein promoter experiments also revealed SHL-1 expression in a subset of neurons as well as in C. elegans body wall muscle and in male-specific diagonal muscles. The shl-1 (ok1168) null mutant removed all fast transient outward current from muscle cells. SHL-1 currents strongly resembled Shal currents in other species except that they were active in a more depolarized voltage range. We also determined that the remaining delayed-rectifier current in cultured myocytes was carried by the Shaker ortholog SHK-1. In shl-1 (ok1168) mutants there was a significant compensatory increase in the SHK-1 current. Male shl-1 (ok1168) animals exhibited reduced mating efficiency resulting from an apparent difficulty in locating the hermaphrodite vulva. SHL-1 channels are apparently important in fine-tuning complex behaviors, such as mating, that play a crucial role in the survival and propagation of the species.

Collaboration


Dive into the Celia M. Santi's collaboration.

Top Co-Authors

Avatar

Lawrence Salkoff

Washington University in St. Louis

View shared research outputs
Top Co-Authors

Avatar

Alice Butler

Washington University in St. Louis

View shared research outputs
Top Co-Authors

Avatar

Alberto Darszon

National Autonomous University of Mexico

View shared research outputs
Top Co-Authors

Avatar

Claudia L. Treviño

National Autonomous University of Mexico

View shared research outputs
Top Co-Authors

Avatar

Aguan Wei

Washington University in St. Louis

View shared research outputs
Top Co-Authors

Avatar

Pablo E. Visconti

University of Massachusetts Amherst

View shared research outputs
Top Co-Authors

Avatar

Juan Ferreira

Washington University in St. Louis

View shared research outputs
Top Co-Authors

Avatar

Gloria L. Fawcett

Washington University in St. Louis

View shared research outputs
Top Co-Authors

Avatar

Gonzalo Budelli

Washington University in St. Louis

View shared research outputs
Top Co-Authors

Avatar

Gonzalo Ferreira

Washington University in St. Louis

View shared research outputs
Researchain Logo
Decentralizing Knowledge