Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 20
Filter
1.
EMBO J ; 39(4): e102723, 2020 02 17.
Article in English | MEDLINE | ID: mdl-31880004

ABSTRACT

Cilia serve as cellular antennae that translate sensory information into physiological responses. In the sperm flagellum, a single chemoattractant molecule can trigger a Ca2+ rise that controls motility. The mechanisms underlying such ultra-sensitivity are ill-defined. Here, we determine by mass spectrometry the copy number of nineteen chemosensory signaling proteins in sperm flagella from the sea urchin Arbacia punctulata. Proteins are up to 1,000-fold more abundant than the free cellular messengers cAMP, cGMP, H+ , and Ca2+ . Opto-chemical techniques show that high protein concentrations kinetically compartmentalize the flagellum: Within milliseconds, cGMP is relayed from the receptor guanylate cyclase to a cGMP-gated channel that serves as a perfect chemo-electrical transducer. cGMP is rapidly hydrolyzed, possibly via "substrate channeling" from the channel to the phosphodiesterase PDE5. The channel/PDE5 tandem encodes cGMP turnover rates rather than concentrations. The rate-detection mechanism allows continuous stimulus sampling over a wide dynamic range. The textbook notion of signal amplification-few enzyme molecules process many messenger molecules-does not hold for sperm flagella. Instead, high protein concentrations ascertain messenger detection. Similar mechanisms may occur in other small compartments like primary cilia or dendritic spines.


Subject(s)
Arbacia/physiology , Chemotaxis , Proteomics , Signal Transduction , Animals , Arbacia/ultrastructure , Calcium/metabolism , Cilia/physiology , Cilia/ultrastructure , Cyclic GMP/metabolism , Electron Microscope Tomography , Flagella/physiology , Flagella/ultrastructure , Guanylate Cyclase/metabolism , Male , Mass Spectrometry , Spermatozoa/physiology , Spermatozoa/ultrastructure
2.
Proc Natl Acad Sci U S A ; 117(24): 13783-13791, 2020 06 16.
Article in English | MEDLINE | ID: mdl-32467169

ABSTRACT

Proton (H+) channels are special: They select protons against other ions that are up to a millionfold more abundant. Only a few proton channels have been identified so far. Here, we identify a family of voltage-gated "pacemaker" channels, HCNL1, that are exquisitely selective for protons. HCNL1 activates during hyperpolarization and conducts protons into the cytosol. Surprisingly, protons permeate through the channel's voltage-sensing domain, whereas the pore domain is nonfunctional. Key to proton permeation is a methionine residue that interrupts the series of regularly spaced arginine residues in the S4 voltage sensor. HCNL1 forms a tetramer and thus contains four proton pores. Unlike classic HCN channels, HCNL1 is not gated by cyclic nucleotides. The channel is present in zebrafish sperm and carries a proton inward current that acidifies the cytosol. Our results suggest that protons rather than cyclic nucleotides serve as cellular messengers in zebrafish sperm. Through small modifications in two key functional domains, HCNL1 evolutionarily adapted to a low-Na+ freshwater environment to conserve sperm's ability to depolarize.


Subject(s)
Zebrafish/metabolism , Amino Acid Sequence , Animals , Biological Transport , Male , Multigene Family , Protons , Spermatozoa/metabolism , Zebrafish/genetics
3.
Proc Natl Acad Sci U S A ; 117(39): 24359-24368, 2020 09 29.
Article in English | MEDLINE | ID: mdl-32938798

ABSTRACT

The mechanisms underlying sex determination are astonishingly plastic. Particularly the triggers for the molecular machinery, which recalls either the male or female developmental program, are highly variable and have evolved independently and repeatedly. Fish show a huge variety of sex determination systems, including both genetic and environmental triggers. The advent of sex chromosomes is assumed to stabilize genetic sex determination. However, because sex chromosomes are notoriously cluttered with repetitive DNA and pseudogenes, the study of their evolution is hampered. Here we reconstruct the birth of a Y chromosome present in the Atlantic herring. The region is tiny (230 kb) and contains only three intact genes. The candidate male-determining gene BMPR1BBY encodes a truncated form of a BMP1B receptor, which originated by gene duplication and translocation and underwent rapid protein evolution. BMPR1BBY phosphorylates SMADs in the absence of ligand and thus has the potential to induce testis formation. The Y region also contains two genes encoding subunits of the sperm-specific Ca2+ channel CatSper required for male fertility. The herring Y chromosome conforms with a characteristic feature of many sex chromosomes, namely, suppressed recombination between a sex-determining factor and genes that are beneficial for the given sex. However, the herring Y differs from other sex chromosomes in that suppression of recombination is restricted to an ∼500-kb region harboring the male-specific and sex-associated regions. As a consequence, any degeneration on the herring Y chromosome is restricted to those genes located in the small region affected by suppressed recombination.


Subject(s)
Fishes/genetics , Sex Chromosomes/genetics , Animals , Evolution, Molecular , Female , Fish Proteins/genetics , Fishes/physiology , Gene Duplication , Male , Reproduction
4.
J Am Chem Soc ; 143(18): 6981-6989, 2021 05 12.
Article in English | MEDLINE | ID: mdl-33905249

ABSTRACT

The function of proteins is linked to their conformations that can be resolved with several high-resolution methods. However, only a few methods can provide the temporal order of intermediates and conformational changes, with each having its limitations. Here, we combine pulsed electron-electron double resonance spectroscopy with a microsecond freeze-hyperquenching setup to achieve spatiotemporal resolution in the angstrom range and lower microsecond time scale. We show that the conformational change of the Cα-helix in the cyclic nucleotide-binding domain of the Mesorhizobium loti potassium channel occurs within about 150 µs and can be resolved with angstrom precision. Thus, this approach holds great promise for obtaining 4D landscapes of conformational changes in biomolecules.


Subject(s)
Electrons , Freezing , Mesorhizobium/chemistry , Potassium Channels/metabolism , Models, Molecular , Potassium Channels/chemistry , Protein Conformation , Spectrum Analysis , Time Factors
5.
J Biol Chem ; 294(11): 3853-3871, 2019 03 15.
Article in English | MEDLINE | ID: mdl-30662006

ABSTRACT

The nonlysosomal glucosylceramidase ß2 (GBA2) catalyzes the hydrolysis of glucosylceramide to glucose and ceramide. Mutations in the human GBA2 gene have been associated with hereditary spastic paraplegia (HSP), autosomal-recessive cerebellar ataxia (ARCA), and the Marinesco-Sjögren-like syndrome. However, the underlying molecular mechanisms are ill-defined. Here, using biochemistry, immunohistochemistry, structural modeling, and mouse genetics, we demonstrate that all but one of the spastic gait locus #46 (SPG46)-connected mutations cause a loss of GBA2 activity. We demonstrate that GBA2 proteins form oligomeric complexes and that protein-protein interactions are perturbed by some of these mutations. To study the pathogenesis of GBA2-related HSP and ARCA in vivo, we investigated GBA2-KO mice as a mammalian model system. However, these mice exhibited a high phenotypic variance and did not fully resemble the human phenotype, suggesting that mouse and human GBA2 differ in function. Whereas some GBA2-KO mice displayed a strong locomotor defect, others displayed only mild alterations of the gait pattern and no signs of cerebellar defects. On a cellular level, inhibition of GBA2 activity in isolated cerebellar neurons dramatically affected F-actin dynamics and reduced neurite outgrowth, which has been associated with the development of neurological disorders. Our results shed light on the molecular mechanism underlying the pathogenesis of GBA2-related HSP and ARCA and reveal species-specific differences in GBA2 function in vivo.


Subject(s)
Cerebellar Ataxia/metabolism , Locomotion/genetics , Loss of Function Mutation , Spastic Paraplegia, Hereditary/metabolism , beta-Glucosidase/metabolism , Animals , Biocatalysis , Cerebellar Ataxia/genetics , Glucosylceramidase , Humans , Mice , Mice, Knockout , Spastic Paraplegia, Hereditary/genetics , Species Specificity , beta-Glucosidase/antagonists & inhibitors , beta-Glucosidase/deficiency , beta-Glucosidase/genetics
6.
EMBO J ; 34(3): 379-92, 2015 Feb 03.
Article in English | MEDLINE | ID: mdl-25535245

ABSTRACT

Sperm guidance is controlled by chemical and physical cues. In many species, Ca(2+) bursts in the flagellum govern navigation to the egg. In Arbacia punctulata, a model system of sperm chemotaxis, a cGMP signaling pathway controls these Ca(2+) bursts. The underlying Ca(2+) channel and its mechanisms of activation are unknown. Here, we identify CatSper Ca(2+) channels in the flagellum of A. punctulata sperm. We show that CatSper mediates the chemoattractant-evoked Ca(2+) influx and controls chemotactic steering; a concomitant alkalization serves as a highly cooperative mechanism that enables CatSper to transduce periodic voltage changes into Ca(2+) bursts. Our results reveal intriguing phylogenetic commonalities but also variations between marine invertebrates and mammals regarding the function and control of CatSper. The variations probably reflect functional and mechanistic adaptations that evolved during the transition from external to internal fertilization.


Subject(s)
Calcium Channels/metabolism , Calcium Signaling/physiology , Chemotaxis/physiology , Evolution, Molecular , Membrane Potentials/physiology , Sea Urchins/metabolism , Animals , Calcium Channels/genetics , Male , Sea Urchins/genetics
7.
J Physiol ; 595(5): 1533-1546, 2017 03 01.
Article in English | MEDLINE | ID: mdl-27859356

ABSTRACT

KEY POINTS: In human sperm, proton flux across the membrane is controlled by the voltage-gated proton channel Hv1. We show that sperm harbour both Hv1 and an N-terminally cleaved isoform termed Hv1Sper. The pH-control of Hv1Sper and Hv1 is distinctively different. Hv1Sper and Hv1 can form heterodimers that combine features of both constituents. Cleavage and heterodimerization of Hv1 might represent an adaptation to the specific requirements of pH control in sperm. ABSTRACT: In human sperm, the voltage-gated proton channel Hv1 controls the flux of protons across the flagellar membrane. Here, we show that sperm harbour Hv1 and a shorter isoform, termed Hv1Sper. Hv1Sper is generated from Hv1 by removal of 68 amino acids from the N-terminus by post-translational proteolytic cleavage. The pH-dependent gating of the channel isoforms is distinctly different. In both Hv1 and Hv1Sper, the conductance-voltage relationship is determined by the pH difference across the membrane (∆pH). However, simultaneous changes in intracellular and extracellular pH that leave ΔpH constant strongly shift the activation curve of Hv1Sper but not that of Hv1, demonstrating that cleavage of the N-terminus tunes pH sensing in Hv1. Moreover, we show that Hv1 and Hv1Sper assemble as heterodimers that combine features of both constituents. We suggest that cleavage and heterodimerization of Hv1 represents an adaptation to the specific requirements of pH control in sperm.


Subject(s)
Ion Channel Gating/physiology , Ion Channels/physiology , Spermatozoa/physiology , Animals , Cell Line , HEK293 Cells , Humans , Hydrogen-Ion Concentration , Ion Channels/metabolism , Male , Mice, Inbred C57BL , Oocytes/physiology , Protein Processing, Post-Translational/drug effects , Respiratory Mucosa , Serine Proteases/metabolism , Serine Proteinase Inhibitors/pharmacology , Spermatozoa/drug effects , Spermatozoa/metabolism , Sulfones/pharmacology , Xenopus laevis
8.
J Biol Chem ; 288(5): 3381-93, 2013 Feb 01.
Article in English | MEDLINE | ID: mdl-23250757

ABSTRACT

GBA1 and GBA2 are both ß-glucosidases, which cleave glucosylceramide (GlcCer) to glucose and ceramide. GlcCer is a main precursor for higher order glycosphingolipids but might also serve as intracellular messenger. Mutations in the lysosomal GBA1 underlie Gaucher disease, the most common lysosomal storage disease in humans. Knocking out the non-lysosomal GBA2 in mice results in accumulation of GlcCer outside the lysosomes in various tissues (e.g. testis and liver) and impairs sperm development and liver regeneration. However, the underlying mechanisms are not well understood. To reveal the physiological function of GBA2 and, thereby, of the non-lysosomal GlcCer pool, it is important to characterize the localization of GBA2 and its activity in different tissues. Thus, we generated GBA2-specific antibodies and developed an assay that discriminates between GBA1 and GBA2 without the use of detergent. We show that GBA2 is not, as previously thought, an integral membrane protein but rather a cytosolic protein that tightly associates with cellular membranes. The interaction with the membrane, in particular with phospholipids, is important for its activity. GBA2 is localized at the ER and Golgi, which puts GBA2 in a key position for a lysosome-independent route of GlcCer-dependent signaling. Furthermore, our results suggest that GBA2 might affect the phenotype of Gaucher disease, because GBA2 activity is reduced in Gba1 knock-out fibroblasts and fibroblasts from a Gaucher patient. Our results provide the basis to understand the mechanism for GBA2 function in vivo and might help to unravel the role of GBA2 during pathogenesis of Gaucher disease.


Subject(s)
Endoplasmic Reticulum/enzymology , Golgi Apparatus/enzymology , Lysosomes/enzymology , Membrane Proteins/metabolism , beta-Glucosidase/metabolism , Animals , Antibody Specificity , Down-Regulation , Enzyme Assays , Fibroblasts/enzymology , Fluorescence , Glucosylceramidase , HEK293 Cells , Hippocampus/cytology , Humans , Mice , Neurons/cytology , Neurons/enzymology , Protein Binding , Protein Transport , beta-Glucosidase/immunology
9.
Nat Cell Biol ; 8(10): 1149-54, 2006 Oct.
Article in English | MEDLINE | ID: mdl-16964244

ABSTRACT

Eggs attract sperm by chemical factors, a process called chemotaxis. Sperm from marine invertebrates use cGMP signalling to transduce incident chemoattractants into changes in the Ca2+ concentration in the flagellum, which control the swimming behaviour during chemotaxis. The signalling pathway downstream of the synthesis of cGMP by a guanylyl cyclase is ill-defined. In particular, the ion channels that are involved in Ca2+ influx and their mechanisms of gating are not known. Using rapid voltage-sensitive dyes and kinetic techniques, we record the voltage response that is evoked by the chemoattractant in sperm from the sea urchin Arbacia punctulata. We show that the chemoattractant evokes a brief hyperpolarization followed by a sustained depolarization. The hyperpolarization is caused by the opening of K+-selective cyclic-nucleotide-gated (CNG) channels in the flagellum. Ca2+ influx commences at the onset of recovery from hyperpolarization. The voltage threshold of Ca2+ entry indicates the involvement of low-voltage-activated Ca(v) channels. These results establish a model of chemosensory transduction in sperm whereby a cGMP-induced hyperpolarization opens Ca(v) channels by a 'recovery-from-inactivation' mechanism and unveil an evolutionary kinship between transduction mechanisms in sperm and photoreceptors.


Subject(s)
Calcium Signaling/physiology , Cyclic GMP/metabolism , Ion Channel Gating , Ion Channels , Potassium/metabolism , Signal Transduction , Spermatozoa/metabolism , Animals , Arbacia/chemistry , Calcium/metabolism , Chemotaxis , Guanylate Cyclase/metabolism , Male
10.
Elife ; 92020 06 24.
Article in English | MEDLINE | ID: mdl-32579112

ABSTRACT

Compartmentalization of cellular signaling forms the molecular basis of cellular behavior. The primary cilium constitutes a subcellular compartment that orchestrates signal transduction independent from the cell body. Ciliary dysfunction causes severe diseases, termed ciliopathies. Analyzing ciliary signaling has been challenging due to the lack of tools to investigate ciliary signaling. Here, we describe a nanobody-based targeting approach for optogenetic tools in mammalian cells and in vivo in zebrafish to specifically analyze ciliary signaling and function. Thereby, we overcome the loss of protein function observed after fusion to ciliary targeting sequences. We functionally localized modifiers of cAMP signaling, the photo-activated adenylyl cyclase bPAC and the light-activated phosphodiesterase LAPD, and the cAMP biosensor mlCNBD-FRET to the cilium. Using this approach, we studied the contribution of spatial cAMP signaling in controlling cilia length. Combining optogenetics with nanobody-based targeting will pave the way to the molecular understanding of ciliary function in health and disease.


Subject(s)
Cilia/physiology , Optogenetics , Signal Transduction/physiology , Single-Domain Antibodies , Animals , Calcium/metabolism , Cell Line , Humans , Mice , Single-Cell Analysis
11.
Nat Neurosci ; 7(7): 705-10, 2004 Jul.
Article in English | MEDLINE | ID: mdl-15195096

ABSTRACT

An important mechanism by which vertebrate olfactory sensory neurons rapidly adapt to odorants is feedback modulation of the Ca(2+)-permeable cyclic nucleotide-gated (CNG) transduction channels. Extensive heterologous studies of homomeric CNGA2 channels have led to a molecular model of channel modulation based on the binding of calcium-calmodulin to a site on the cytoplasmic amino terminus of CNGA2. Native rat olfactory CNG channels, however, are heteromeric complexes of three homologous but distinct subunits. Notably, in heteromeric channels, we found no role for CNGA2 in feedback modulation. Instead, an IQ-type calmodulin-binding site on CNGB1b and a similar but previously unidentified site on CNGA4 are necessary and sufficient. These sites seem to confer binding of Ca(2+)-free calmodulin (apocalmodulin), which is then poised to trigger inhibition of native channels in the presence of Ca(2+).


Subject(s)
Calcium/metabolism , Calmodulin/metabolism , Ion Channels/metabolism , Olfactory Bulb/cytology , Olfactory Receptor Neurons/physiology , Amino Acid Motifs/physiology , Animals , Calcium Signaling , Cells, Cultured , Cyclic AMP/pharmacology , Cyclic Nucleotide-Gated Cation Channels , Embryo, Mammalian , Feedback, Physiological , Humans , Ion Channel Gating/physiology , Ion Channels/chemistry , Kidney , Membrane Potentials/physiology , Mutagenesis, Site-Directed , Patch-Clamp Techniques/methods , Rats , Rats, Sprague-Dawley , Recombinant Proteins/metabolism , Transfection/methods
12.
Nat Commun ; 9(1): 4611, 2018 11 05.
Article in English | MEDLINE | ID: mdl-30397200

ABSTRACT

Optogenetics enables manipulation of biological processes with light at high spatio-temporal resolution to control the behavior of cells, networks, or even whole animals. In contrast to the performance of excitatory rhodopsins, the effectiveness of inhibitory optogenetic tools is still insufficient. Here we report a two-component optical silencer system comprising photoactivated adenylyl cyclases (PACs) and the small cyclic nucleotide-gated potassium channel SthK. Activation of this 'PAC-K' silencer by brief pulses of low-intensity blue light causes robust and reversible silencing of cardiomyocyte excitation and neuronal firing. In vivo expression of PAC-K in mouse and zebrafish neurons is well tolerated, where blue light inhibits neuronal activity and blocks motor responses. In combination with red-light absorbing channelrhodopsins, the distinct action spectra of PACs allow independent bimodal control of neuronal activity. PAC-K represents a reliable optogenetic silencer with intrinsic amplification for sustained potassium-mediated hyperpolarization, conferring high operational light sensitivity to the cells of interest.


Subject(s)
Optogenetics/methods , Potassium Channels/genetics , Potassium Channels/metabolism , Potassium Channels/radiation effects , Silencer Elements, Transcriptional , Adenylyl Cyclases/genetics , Adenylyl Cyclases/metabolism , Adenylyl Cyclases/radiation effects , Animals , Animals, Genetically Modified , Channelrhodopsins/radiation effects , Gene Expression/genetics , Gene Expression/radiation effects , HEK293 Cells , Humans , Light , Mice , Models, Animal , Myocytes, Cardiac/metabolism , Neurons/metabolism , Neurons/radiation effects , Rhodopsin/pharmacology , Zebrafish
13.
Elife ; 42015 Dec 09.
Article in English | MEDLINE | ID: mdl-26650356

ABSTRACT

Calcium in the flagellum controls sperm navigation. In sperm of marine invertebrates and mammals, Ca(2+) signalling has been intensely studied, whereas for fish little is known. In sea urchin sperm, a cyclic nucleotide-gated K(+) channel (CNGK) mediates a cGMP-induced hyperpolarization that evokes Ca(2+) influx. Here, we identify in sperm of the freshwater fish Danio rerio a novel CNGK family member featuring non-canonical properties. It is located in the sperm head rather than the flagellum and is controlled by intracellular pH, but not cyclic nucleotides. Alkalization hyperpolarizes sperm and produces Ca(2+) entry. Ca(2+) induces spinning-like swimming, different from swimming of sperm from other species. The "spinning" mode probably guides sperm into the micropyle, a narrow entrance on the surface of fish eggs. A picture is emerging of sperm channel orthologues that employ different activation mechanisms and serve different functions. The channel inventories probably reflect adaptations to species-specific challenges during fertilization.


Subject(s)
Calcium Signaling , Cyclic Nucleotide-Gated Cation Channels/metabolism , Potassium/metabolism , Spermatozoa/physiology , Zebrafish/physiology , Animals , Male , Spermatozoa/drug effects
14.
Elife ; 3: e01438, 2014 Mar 26.
Article in English | MEDLINE | ID: mdl-24670955

ABSTRACT

Sperm are equipped with a unique set of ion channels that orchestrate fertilization. In mouse sperm, the principal K(+) current (IKSper) is carried by the Slo3 channel, which sets the membrane potential (Vm) in a strongly pHi-dependent manner. Here, we show that IKSper in human sperm is activated weakly by pHi and more strongly by Ca(2+). Correspondingly, Vm is strongly regulated by Ca(2+) and less so by pHi. We find that inhibitors of Slo3 suppress human IKSper, and we identify the Slo3 protein in the flagellum of human sperm. Moreover, heterologously expressed human Slo3, but not mouse Slo3, is activated by Ca(2+) rather than by alkaline pHi; current-voltage relations of human Slo3 and human IKSper are similar. We conclude that Slo3 represents the principal K(+) channel in human sperm that carries the Ca(2+)-activated IKSper current. We propose that, in human sperm, the progesterone-evoked Ca(2+) influx carried by voltage-gated CatSper channels is limited by Ca(2+)-controlled hyperpolarization via Slo3. DOI: http://dx.doi.org/10.7554/eLife.01438.001.


Subject(s)
Calcium/metabolism , Potassium Channels, Voltage-Gated/metabolism , Potassium/metabolism , Spermatozoa/drug effects , Spermatozoa/physiology , Flagella/chemistry , Humans , Hydrogen-Ion Concentration , Large-Conductance Calcium-Activated Potassium Channel alpha Subunits , Large-Conductance Calcium-Activated Potassium Channels , Male , Potassium Channels, Voltage-Gated/genetics , Spermatozoa/metabolism
15.
J Cell Biol ; 206(4): 541-57, 2014 Aug 18.
Article in English | MEDLINE | ID: mdl-25135936

ABSTRACT

Guanylyl cyclases (GCs), which synthesize the messenger cyclic guanosine 3',5'-monophosphate, control several sensory functions, such as phototransduction, chemosensation, and thermosensation, in many species from worms to mammals. The GC chemoreceptor in sea urchin sperm can decode chemoattractant concentrations with single-molecule sensitivity. The molecular and cellular underpinnings of such ultrasensitivity are not known for any eukaryotic chemoreceptor. In this paper, we show that an exquisitely high density of 3 × 10(5) GC chemoreceptors and subnanomolar ligand affinity provide a high ligand-capture efficacy and render sperm perfect absorbers. The GC activity is terminated within 150 ms by dephosphorylation steps of the receptor, which provides a means for precise control of the GC lifetime and which reduces "molecule noise." Compared with other ultrasensitive sensory systems, the 10-fold signal amplification by the GC receptor is surprisingly low. The hallmarks of this signaling mechanism provide a blueprint for chemical sensing in small compartments, such as olfactory cilia, insect antennae, or even synaptic boutons.


Subject(s)
Arbacia/metabolism , Cyclic GMP/biosynthesis , Guanylate Cyclase/metabolism , Receptors, Guanylate Cyclase-Coupled/metabolism , Spermatozoa/metabolism , Animals , Chemoreceptor Cells/metabolism , Chemotactic Factors/physiology , HEK293 Cells , Humans , Male , Phosphorylation , Protein Binding , Signal Transduction
16.
Sci Signal ; 5(232): ra48, 2012 Jul 10.
Article in English | MEDLINE | ID: mdl-22786723

ABSTRACT

Olfactory cyclic nucleotide-gated (CNG) ion channels are essential contributors to signal transduction of olfactory sensory neurons. The activity of the channels is controlled by the cyclic nucleotides guanosine 3',5'-monophosphate (cGMP) and adenosine 3',5'-monophosphate (cAMP). The olfactory CNG channels are composed of two CNGA2 subunits, one CNGA4 and one CNGB1b subunit, each containing a cyclic nucleotide-binding domain. Using patch-clamp fluorometry, we measured ligand binding and channel activation simultaneously and showed that cGMP activated olfactory CNG channels not only by binding to the two CNGA2 subunits but also by binding to the CNGA4 subunit. In a channel in which the CNGA2 subunits were compromised for ligand binding, cGMP binding to CNGA4 was sufficient to partly activate the channel. In contrast, in heterotetrameric channels, the CNGB1b subunit did not bind cGMP, but channels with this subunit showed activation by cAMP. Thus, the modulatory subunits participate actively in translating ligand binding to activation of heterotetrameric olfactory CNG channels and enable the channels to differentiate between cyclic nucleotides.


Subject(s)
Cyclic AMP/metabolism , Cyclic GMP/metabolism , Cyclic Nucleotide-Gated Cation Channels/metabolism , Olfactory Nerve/metabolism , Protein Subunits/metabolism , Signal Transduction/physiology , Animals , Fluorometry , Microscopy, Fluorescence , Patch-Clamp Techniques , Protein Binding , Rats
17.
Sci Signal ; 2(94): ra68, 2009 Oct 27.
Article in English | MEDLINE | ID: mdl-19861689

ABSTRACT

Sperm of the sea urchin Arbacia punctulata can respond to a single molecule of chemoattractant released by an egg. The mechanism underlying this extreme sensitivity is unknown. Crucial signaling events in the response of A. punctulata sperm to chemoattractant include the rapid synthesis of the intracellular messenger guanosine 3',5'-monophosphate (cGMP) and the ensuing membrane hyperpolarization that results from the opening of potassium-selective cyclic nucleotide-gated (CNGK) channels. Here, we use calibrated photolysis of caged cGMP to show that approximately 45 cGMP molecules are generated during the response to a single molecule of chemoattractant. The CNGK channel can respond to such small cGMP changes because it is exquisitely sensitive to cGMP and activated in a noncooperative fashion. Like voltage-activated Ca(v) and Na(v) channels, the CNGK polypeptide consists of four homologous repeat sequences. Disabling each of the four cyclic nucleotide-binding sites through mutagenesis revealed that binding of a single cGMP molecule to repeat 3 is necessary and sufficient to activate the CNGK channel. Thus, CNGK has developed a mechanism of activation that is different from the activation of other CNG channels, which requires the cooperative binding of several ligands and operates in the micromolar rather than the nanomolar range.


Subject(s)
Chemotaxis/physiology , Cyclic GMP/physiology , Cyclic Nucleotide-Gated Cation Channels/physiology , Spermatozoa/cytology , Amino Acid Sequence , Animals , Blotting, Western , Cloning, Molecular , Cyclic Nucleotide-Gated Cation Channels/chemistry , Cyclic Nucleotide-Gated Cation Channels/genetics , Hydrolysis , Immunohistochemistry , Male , Molecular Sequence Data , Protein Processing, Post-Translational , Sea Urchins , Sequence Homology, Amino Acid
18.
EMBO Rep ; 8(8): 749-55, 2007 Aug.
Article in English | MEDLINE | ID: mdl-17668006

ABSTRACT

Ion channels gated by cyclic nucleotides have crucial roles in neuronal excitability and signal transduction of sensory neurons. Here, we studied ligand binding of a cyclic nucleotide-activated K(+) channel from Mesorhizobium loti and its isolated cyclic nucleotide-binding domain. The channel and the binding domain alone bind cyclic AMP with similar affinity in a non-cooperative manner. The cAMP sensitivities of binding and activation coincide. Thus, each subunit in the tetrameric channel acts independently of the others. The binding and gating properties of the bacterial channel are distinctively different from those of eukaryotic cyclic nucleotide-gated channels.


Subject(s)
Alphaproteobacteria/metabolism , Bacterial Proteins/chemistry , Cyclic AMP/chemistry , Cyclic Nucleotide-Gated Cation Channels/chemistry , Potassium Channels/chemistry , Bacterial Proteins/genetics , Cyclic Nucleotide-Gated Cation Channels/genetics , Ligands , Potassium Channels/genetics , Protein Structure, Tertiary , Protein Subunits/chemistry , Spectrometry, Fluorescence
19.
Anal Chem ; 79(11): 4040-9, 2007 Jun 01.
Article in English | MEDLINE | ID: mdl-17487973

ABSTRACT

Fluorescence antibunching is a well-known technique for determining the number of independent emitters per molecule or molecular complex. It was rarely applied to autofluorescent proteins due to the necessity of collecting large numbers of fluorescence photons from a single molecule, which is usually impossible to achieve with rather photolabile autofluorescent proteins. Here, we measure fluorescence antibunching on molecules in solution, allowing us to accumulate data over a large number of molecules. We use that method for determining an average stoichiometry of molecular complexes. The proposed method is absolute in the sense that it does not need any calibration or referencing. We develop the necessary theoretical background and check the method on pure dye solutions and on molecular complexes with known stoichiometry.


Subject(s)
Spectrometry, Fluorescence/methods , Animals , Genes, Reporter/genetics , Glucosides/chemistry , Oligonucleotides/chemistry , Receptors, Glycine/chemistry , Receptors, Purinergic P2/chemistry , Receptors, Purinergic P2X , Solutions , Xenopus laevis
20.
Biochemistry ; 42(15): 4593-600, 2003 Apr 22.
Article in English | MEDLINE | ID: mdl-12693957

ABSTRACT

Proper control of intracellular free Ca(2+) is thought to involve subsets of proteins that co-localize to mediate coordinated Ca(2+) entry and Ca(2+) extrusion. The outer segments of vertebrate rod and cone photoreceptors present one example: Ca(2+) influx is exclusively mediated via cGMP-gated channels (CNG), whereas the Na(+)/Ca(2+)-K(+) exchanger (NCKX) is the only Ca(2+) extrusion protein present. In situ, a rod NCKX homodimer and a CNG heterotetramer are thought to be part of a single protein complex. However, NCKX-NCKX and NCKX-CNG interactions have been described so far only in bovine rod outer segment membranes. We have used thiol-specific cross-linking and co-immunoprecipitation to examine NCKX self-assembly and CNG-NCKX co-assembly after heterologous expression of either the rod or cone NCKX/CNG isoforms. Co-immunoprecipitation clearly demonstrated both NCKX homooligomerization and interactions between NCKX and CNG. The NCKX-NCKX and NCKX-CNG interactions were observed for both the rod and the cone isoforms. Thiol-specific cross-linking led to rod NCKX1 dimers and to cone NCKX2 adducts of an apparent molecular weight higher than that predicted for a NCKX2 dimer. The mass of the cross-link product critically depended on the location of the particular cysteine residue used by the cross-linker, and we cannot exclude that NCKX forms a higher oligomer. The NCKX-NCKX and NCKX-CNG interactions were not isoform-specific (i.e., rod NCKX could interact with cone NCKX, rod NCKX could interact with cone CNGA, and vice versa). Deletion of the two large hydrophilic loops from the NCKX protein did not abolish the NCKX oligomerization, suggesting that it is mediated by the highly conserved transmembrane spanning segments.


Subject(s)
Cyclic GMP/metabolism , Ion Channels/metabolism , Photoreceptor Cells, Vertebrate/metabolism , Sodium-Calcium Exchanger/metabolism , Animals , Calcium/metabolism , Cell Line , Chickens/metabolism , Cyclic Nucleotide-Gated Cation Channels , Humans , In Vitro Techniques , Insecta , Precipitin Tests , Sodium-Calcium Exchanger/biosynthesis
SELECTION OF CITATIONS
SEARCH DETAIL