Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 77
Filtrar
1.
Proc Natl Acad Sci U S A ; 120(15): e2300309120, 2023 04 11.
Artigo em Inglês | MEDLINE | ID: mdl-37011209

RESUMO

Calmodulin (CaM) regulates many ion channels to control calcium entry into cells, and mutations that alter this interaction are linked to fatal diseases. The structural basis of CaM regulation remains largely unexplored. In retinal photoreceptors, CaM binds to the CNGB subunit of cyclic nucleotide-gated (CNG) channels and, thereby, adjusts the channel's Cyclic guanosine monophosphate (cGMP) sensitivity in response to changes in ambient light conditions. Here, we provide the structural characterization for CaM regulation of a CNG channel by using a combination of single-particle cryo-electron microscopy and structural proteomics. CaM connects the CNGA and CNGB subunits, resulting in structural changes both in the cytosolic and transmembrane regions of the channel. Cross-linking and limited proteolysis-coupled mass spectrometry mapped the conformational changes induced by CaM in vitro and in the native membrane. We propose that CaM is a constitutive subunit of the rod channel to ensure high sensitivity in dim light. Our mass spectrometry-based approach is generally relevant for studying the effect of CaM on ion channels in tissues of medical interest, where only minute quantities are available.


Assuntos
Calmodulina , Canais de Cátion Regulados por Nucleotídeos Cíclicos , Canais de Cátion Regulados por Nucleotídeos Cíclicos/genética , Canais de Cátion Regulados por Nucleotídeos Cíclicos/metabolismo , Calmodulina/metabolismo , Ativação do Canal Iônico/fisiologia , Microscopia Crioeletrônica , Cálcio/metabolismo , Nucleotídeos Cíclicos/farmacologia , GMP Cíclico/metabolismo
2.
EMBO J ; 39(4): e102363, 2020 02 17.
Artigo em Inglês | MEDLINE | ID: mdl-31957048

RESUMO

Navigation of sperm in fluid flow, called rheotaxis, provides long-range guidance in the mammalian oviduct. The rotation of sperm around their longitudinal axis (rolling) promotes rheotaxis. Whether sperm rolling and rheotaxis require calcium (Ca2+ ) influx via the sperm-specific Ca2+ channel CatSper, or rather represent passive biomechanical and hydrodynamic processes, has remained controversial. Here, we study the swimming behavior of sperm from healthy donors and from infertile patients that lack functional CatSper channels, using dark-field microscopy, optical tweezers, and microfluidics. We demonstrate that rolling and rheotaxis persist in CatSper-deficient human sperm. Furthermore, human sperm undergo rolling and rheotaxis even when Ca2+ influx is prevented. Finally, we show that rolling and rheotaxis also persist in mouse sperm deficient in both CatSper and flagellar Ca2+ -signaling domains. Our results strongly support the concept that passive biomechanical and hydrodynamic processes enable sperm rolling and rheotaxis, rather than calcium signaling mediated by CatSper or other mechanisms controlling transmembrane Ca2+ flux.


Assuntos
Hidrodinâmica , Motilidade dos Espermatozoides , Espermatozoides/fisiologia , Animais , Fenômenos Biomecânicos , Cálcio/metabolismo , Canais de Cálcio/genética , Canais de Cálcio/metabolismo , Sinalização do Cálcio , Humanos , Masculino , Camundongos , Proteínas de Plasma Seminal/genética , Proteínas de Plasma Seminal/metabolismo
3.
EMBO J ; 39(4): e102723, 2020 02 17.
Artigo em Inglês | MEDLINE | ID: mdl-31880004

RESUMO

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.


Assuntos
Arbacia/fisiologia , Quimiotaxia , Proteômica , Transdução de Sinais , Animais , Arbacia/ultraestrutura , Cálcio/metabolismo , Cílios/fisiologia , Cílios/ultraestrutura , GMP Cíclico/metabolismo , Tomografia com Microscopia Eletrônica , Flagelos/fisiologia , Flagelos/ultraestrutura , Guanilato Ciclase/metabolismo , Masculino , Espectrometria de Massas , Espermatozoides/fisiologia , Espermatozoides/ultraestrutura
4.
Proc Natl Acad Sci U S A ; 117(24): 13783-13791, 2020 06 16.
Artigo em Inglês | MEDLINE | ID: mdl-32467169

RESUMO

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.


Assuntos
Peixe-Zebra/metabolismo , Sequência de Aminoácidos , Animais , Transporte Biológico , Masculino , Família Multigênica , Prótons , Espermatozoides/metabolismo , Peixe-Zebra/genética
5.
Proc Natl Acad Sci U S A ; 117(39): 24359-24368, 2020 09 29.
Artigo em Inglês | MEDLINE | ID: mdl-32938798

RESUMO

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.


Assuntos
Peixes/genética , Cromossomos Sexuais/genética , Animais , Evolução Molecular , Feminino , Proteínas de Peixes/genética , Peixes/fisiologia , Duplicação Gênica , Masculino , Reprodução
6.
J Struct Biol ; 214(1): 107828, 2022 03.
Artigo em Inglês | MEDLINE | ID: mdl-34971760

RESUMO

The recently reported structure of the human CNGA1/CNGB1 CNG channel in the open state (Xue et al., 2021a) shows that one CNGA1 and one CNGB1 subunit do not open the central hydrophobic gate completely upon cGMP binding. This is different from what has been reported for CNGA homomeric channels (Xue et al., 2021b; Zheng et al., 2020). In seeking to understand how this difference is due to the presence of the CNGB1 subunit, we find that the deposited density map (Xue et al., 2021a) (EMDB 24465) contains an additional density not reported in the images of the original publication. This additional density fits well the structure of calmodulin (CaM), and it unambiguously connects the newly identified D-helix of CNGB1 to one of the CNGA1 helices (A1R) participating in the coiled-coil region. Interestingly, the CNGA1 subunit that engages in the interaction with this additional density is the one that, together with CNGB1, does not open completely the central gate. The sequence of the D-helix of CNGB1 contains a known CaM-binding site of exquisitely high affinity - named CaM2 (Weitz et al., 1998) -, and thus the presence of CaM in that region is not surprising. The mechanism through which CaM reduces currents across the membrane by acting on the native channel (Bauer, 1996; Hsu and Molday, 1993; Weitz et al., 1998) remains unclear. We suggest that the presence of CaM may explain the partially open central gate reported by Xue et al. (2021a). The structure of the open and closed states of the CNGA1/CNGB1 channel may be different with and without CaM present.


Assuntos
Calmodulina , Canais de Cátion Regulados por Nucleotídeos Cíclicos , Sítios de Ligação , Calmodulina/metabolismo , Microscopia Crioeletrônica , Canais de Cátion Regulados por Nucleotídeos Cíclicos/química , Canais de Cátion Regulados por Nucleotídeos Cíclicos/metabolismo , Humanos , Células Fotorreceptoras Retinianas Bastonetes/metabolismo
7.
J Am Chem Soc ; 143(18): 6981-6989, 2021 05 12.
Artigo em Inglês | MEDLINE | ID: mdl-33905249

RESUMO

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.


Assuntos
Elétrons , Congelamento , Mesorhizobium/química , Canais de Potássio/metabolismo , Modelos Moleculares , Canais de Potássio/química , Conformação Proteica , Análise Espectral , Fatores de Tempo
8.
EMBO J ; 34(3): 379-92, 2015 Feb 03.
Artigo em Inglês | MEDLINE | ID: mdl-25535245

RESUMO

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.


Assuntos
Canais de Cálcio/metabolismo , Sinalização do Cálcio/fisiologia , Quimiotaxia/fisiologia , Evolução Molecular , Potenciais da Membrana/fisiologia , Ouriços-do-Mar/metabolismo , Animais , Canais de Cálcio/genética , Masculino , Ouriços-do-Mar/genética
9.
J Physiol ; 595(5): 1533-1546, 2017 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-27859356

RESUMO

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.


Assuntos
Ativação do Canal Iônico/fisiologia , Canais Iônicos/fisiologia , Espermatozoides/fisiologia , Animais , Linhagem Celular , Células HEK293 , Humanos , Concentração de Íons de Hidrogênio , Canais Iônicos/metabolismo , Masculino , Camundongos Endogâmicos C57BL , Oócitos/fisiologia , Processamento de Proteína Pós-Traducional/efeitos dos fármacos , Mucosa Respiratória , Serina Proteases/metabolismo , Inibidores de Serina Proteinase/farmacologia , Espermatozoides/efeitos dos fármacos , Espermatozoides/metabolismo , Sulfonas/farmacologia , Xenopus laevis
10.
Nature ; 471(7338): 382-6, 2011 Mar 17.
Artigo em Inglês | MEDLINE | ID: mdl-21412338

RESUMO

In the oviduct, cumulus cells that surround the oocyte release progesterone. In human sperm, progesterone stimulates a Ca(2+) increase by a non-genomic mechanism. The Ca(2+) signal has been proposed to control chemotaxis, hyperactivation and acrosomal exocytosis of sperm. However, the underlying signalling mechanism has remained mysterious. Here we show that progesterone activates the sperm-specific, pH-sensitive CatSper Ca(2+) channel. We found that both progesterone and alkaline pH stimulate a rapid Ca(2+) influx with almost no latency, incompatible with a signalling pathway involving metabotropic receptors and second messengers. The Ca(2+) signals evoked by alkaline pH and progesterone are inhibited by the Ca(v) channel blockers NNC 55-0396 and mibefradil. Patch-clamp recordings from sperm reveal an alkaline-activated current carried by mono- and divalent ions that exhibits all the hallmarks of sperm-specific CatSper Ca(2+) channels. Progesterone substantially enhances the CatSper current. The alkaline- and progesterone-activated CatSper current is inhibited by both drugs. Our results resolve a long-standing controversy over the non-genomic progesterone signalling. In human sperm, either the CatSper channel itself or an associated protein serves as the non-genomic progesterone receptor. The identification of CatSper channel blockers will greatly facilitate the study of Ca(2+) signalling in sperm and help to define further the physiological role of progesterone and CatSper.


Assuntos
Canais de Cálcio/metabolismo , Sinalização do Cálcio/efeitos dos fármacos , Cálcio/metabolismo , Progesterona/farmacologia , Espermatozoides/efeitos dos fármacos , Espermatozoides/metabolismo , Alprostadil/farmacologia , Benzimidazóis/farmacologia , Bloqueadores dos Canais de Cálcio/farmacologia , AMP Cíclico , Ciclopropanos/farmacologia , Condutividade Elétrica , Humanos , Concentração de Íons de Hidrogênio , Masculino , Mibefradil/farmacologia , Naftalenos/farmacologia , Técnicas de Patch-Clamp , Progesterona/metabolismo
11.
Biophys J ; 111(8): 1668-1678, 2016 Oct 18.
Artigo em Inglês | MEDLINE | ID: mdl-27760354

RESUMO

Ligand-protein binding processes are essential in biological systems. A well-studied system is the binding of cyclic adenosine monophosphate to the cyclic nucleotide binding domain of the bacterial potassium channel MloK1. Strikingly, the measured on-rate for cyclic adenosine monophosphate binding is two orders of magnitude slower than a simple Smoluchowski diffusion model would suggest. To resolve this discrepancy and to characterize the ligand-binding path in structural and energetic terms, we calculated 1100 ligand-binding molecular dynamics trajectories and tested two scenarios: In the first scenario, the ligand transiently binds to the protein surface and then diffuses along the surface into the binding site. In the second scenario, only ligands that reach the protein surface in the vicinity of the binding site proceed into the binding site. Here, a binding funnel, which increasingly confines the translational as well as the rotational degrees of freedom, determines the binding pathways and limits the on-rate. From the simulations, we identified five surface binding states and calculated the rates between these surface binding states, the binding site, and the bulk. We find that the transient binding of the ligands to the surface binding states does not affect the on-rate, such that this effect alone cannot explain the observed low on-rate. Rather, by quantifying the translational and rotational degrees of freedom and by calculating the binding committor, our simulations confirmed the existence of a binding funnel as the main bottleneck. Direct binding via the binding funnel dominates the binding kinetics, and only ∼10% of all ligands proceed via the surface into the binding site. The simulations further predict an on-rate between 15 and 40µs-1(mol/l)-1, which agrees with the measured on-rate.


Assuntos
Proteínas de Bactérias/química , Proteínas de Bactérias/metabolismo , AMP Cíclico/metabolismo , Simulação de Dinâmica Molecular , Canais de Potássio/química , Canais de Potássio/metabolismo , Ligação Proteica , Domínios Proteicos , Rotação
12.
EMBO J ; 31(7): 1654-65, 2012 Apr 04.
Artigo em Inglês | MEDLINE | ID: mdl-22354039

RESUMO

The sperm-specific CatSper channel controls the intracellular Ca(2+) concentration ([Ca(2+)](i)) and, thereby, the swimming behaviour of sperm. In humans, CatSper is directly activated by progesterone and prostaglandins-female factors that stimulate Ca(2+) influx. Other factors including neurotransmitters, chemokines, and odorants also affect sperm function by changing [Ca(2+)](i). Several ligands, notably odorants, have been proposed to control Ca(2+) entry and motility via G protein-coupled receptors (GPCRs) and cAMP-signalling pathways. Here, we show that odorants directly activate CatSper without involving GPCRs and cAMP. Moreover, membrane-permeable analogues of cyclic nucleotides that have been frequently used to study cAMP-mediated Ca(2+) signalling also activate CatSper directly via an extracellular site. Thus, CatSper or associated protein(s) harbour promiscuous binding sites that can host various ligands. These results contest current concepts of Ca(2+) signalling by GPCR and cAMP in mammalian sperm: ligands thought to activate metabotropic pathways, in fact, act via a common ionotropic mechanism. We propose that the CatSper channel complex serves as a polymodal sensor for multiple chemical cues that assist sperm during their voyage across the female genital tract.


Assuntos
Canais de Cálcio/metabolismo , Sinalização do Cálcio/fisiologia , Feromônios/metabolismo , Espermatozoides/metabolismo , Aldeídos/farmacologia , Benzimidazóis/farmacologia , Bloqueadores dos Canais de Cálcio/farmacologia , Sinalização do Cálcio/efeitos dos fármacos , AMP Cíclico/metabolismo , Ciclopropanos/farmacologia , Humanos , Masculino , Mibefradil/farmacologia , Naftalenos/farmacologia , Receptores Acoplados a Proteínas G/metabolismo , Espermatozoides/efeitos dos fármacos
13.
EMBO Rep ; 15(7): 758-65, 2014 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-24820036

RESUMO

Synthetic endocrine disrupting chemicals (EDCs), omnipresent in food, household, and personal care products, have been implicated in adverse trends in human reproduction, including infertility and increasing demand for assisted reproduction. Here, we study the action of 96 ubiquitous EDCs on human sperm. We show that structurally diverse EDCs activate the sperm-specific CatSper channel and, thereby, evoke an intracellular Ca(2+) increase, a motility response, and acrosomal exocytosis. Moreover, EDCs desensitize sperm for physiological CatSper ligands and cooperate in low-dose mixtures to elevate Ca(2+) levels in sperm. We conclude that EDCs interfere with various sperm functions and, thereby, might impair human fertilization.


Assuntos
Disruptores Endócrinos/farmacologia , Espermatozoides/efeitos dos fármacos , Espermatozoides/fisiologia , Acrossomo/metabolismo , Potenciais de Ação/efeitos dos fármacos , Ligação Competitiva , Cálcio/metabolismo , Canais de Cálcio/metabolismo , Sinalização do Cálcio/efeitos dos fármacos , Disruptores Endócrinos/química , Exocitose/efeitos dos fármacos , Humanos , Ligantes , Masculino , Ligação Proteica , Motilidade dos Espermatozoides/efeitos dos fármacos
15.
Trends Biochem Sci ; 36(1): 55-64, 2011 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-20729090

RESUMO

Ion channels gated by cyclic nucleotides serve multiple functions in sensory signaling in diverse cell types ranging from neurons to sperm. Newly discovered members from bacteria and marine invertebrates provide a wealth of structural and functional information on this channel family. A hallmark of classical tetrameric cyclic-nucleotide-gated channels is their cooperative activation by binding of several ligands. By contrast, the new members seem to be uncooperative, and binding of a single ligand molecule suffices to open these channels. These new findings provide a fresh look at the mechanism of allosteric activation of ion channels.


Assuntos
Canais de Cátion Regulados por Nucleotídeos Cíclicos/metabolismo , Animais , Canais de Cátion Regulados por Nucleotídeos Cíclicos/química , Ativação do Canal Iônico , Ligantes , Ligação Proteica
16.
Nat Rev Neurosci ; 11(3): 188-200, 2010 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-20145624

RESUMO

Vertebrates and insects have evolved complex repertoires of chemosensory receptors to detect and distinguish odours. With a few exceptions, vertebrate chemosensory receptors belong to the family of G protein-coupled receptors that initiate a cascade of cellular signalling events and thereby electrically excite the neuron. Insect receptors, which are structurally and genetically unrelated to vertebrate receptors, are a complex of two distinct molecules that serves both as a receptor for the odorant and as an ion channel that is gated by binding of the odorant. Metabotropic signalling in vertebrates provides a rich panoply of positive and negative regulation, whereas ionotropic signalling in insects enhances processing speed.


Assuntos
Insetos/fisiologia , Condutos Olfatórios/fisiologia , Neurônios Receptores Olfatórios/fisiologia , Receptores Odorantes/fisiologia , Vertebrados/fisiologia , Animais , Odorantes , Feromônios , Transdução de Sinais/fisiologia , Olfato/fisiologia
17.
J Biol Chem ; 288(5): 3381-93, 2013 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-23250757

RESUMO

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.


Assuntos
Retículo Endoplasmático/enzimologia , Complexo de Golgi/enzimologia , Lisossomos/enzimologia , Proteínas de Membrana/metabolismo , beta-Glucosidase/metabolismo , Animais , Especificidade de Anticorpos , Regulação para Baixo , Ensaios Enzimáticos , Fibroblastos/enzimologia , Fluorescência , Glucosilceramidase , Células HEK293 , Hipocampo/citologia , Humanos , Camundongos , Neurônios/citologia , Neurônios/enzimologia , Ligação Proteica , Transporte Proteico , beta-Glucosidase/imunologia
18.
Proc Natl Acad Sci U S A ; 108(15): 6121-6, 2011 Apr 12.
Artigo em Inglês | MEDLINE | ID: mdl-21430265

RESUMO

Cyclic nucleotide-sensitive ion channels, known as HCN and CNG channels, are activated by binding of ligands to a domain (CNBD) located on the cytoplasmic side of the channel. The underlying mechanisms are not well understood. To elucidate the gating mechanism, structures of both the ligand-free and -bound CNBD are required. Several crystal structures of the CNBD from HCN2 and a bacterial CNG channel (MloK1) have been solved. However, for HCN2, the cAMP-free and -bound state did not reveal substantial structural rearrangements. For MloK1, structural information for the cAMP-free state has only been gained from mutant CNBDs. Moreover, in the crystal, the CNBD molecules form an interface between dimers, proposed to be important for allosteric channel gating. Here, we have determined the solution structure by NMR spectroscopy of the cAMP-free wild-type CNBD of MloK1. A comparison of the solution structure of cAMP-free and -bound states reveals large conformational rearrangement on ligand binding. The two structures provide insights on a unique set of conformational events that accompany gating within the ligand-binding site.


Assuntos
Alphaproteobacteria/metabolismo , AMP Cíclico/química , Canais de Cátion Regulados por Nucleotídeos Cíclicos/química , Cristalografia por Raios X , Canais de Cátion Regulados por Nucleotídeos Cíclicos/genética , Mutação , Ressonância Magnética Nuclear Biomolecular , Estrutura Terciária de Proteína
19.
Biophys J ; 104(1): 63-74, 2013 Jan 08.
Artigo em Inglês | MEDLINE | ID: mdl-23332059

RESUMO

Many receptors and ion channels are activated by ligands. One key question concerns the binding mechanism. Does the ligand induce conformational changes in the protein via the induced-fit mechanism? Or does the protein preexist as an ensemble of conformers and the ligand selects the most complementary one, via the conformational selection mechanism? Here, we study ligand binding of a tetrameric cyclic nucleotide-gated channel from Mesorhizobium loti and of its monomeric binding domain (CNBD) using rapid mixing, mutagenesis, and structure-based computational biology. Association rate constants of ∼10(7) M(-1) s(-1) are compatible with diffusion-limited binding. Ligand binding to the full-length CNG channel and the isolated CNBD differ, revealing allosteric control of the CNBD by the effector domain. Finally, mutagenesis of allosteric residues affects only the dissociation rate constant, suggesting that binding follows the induced-fit mechanism. This study illustrates the strength of combining mutational, kinetic, and computational approaches to unravel important mechanistic features of ligand binding.


Assuntos
Canais de Cátion Regulados por Nucleotídeos Cíclicos/metabolismo , Mesorhizobium/metabolismo , Receptores de Superfície Celular/metabolismo , Regulação Alostérica , Arginina , Proteínas de Bactérias/química , Canais de Cátion Regulados por Nucleotídeos Cíclicos/química , Canais de Cátion Regulados por Nucleotídeos Cíclicos/genética , Análise Mutacional de DNA , Cinética , Ligantes , Modelos Moleculares , Proteínas Mutantes/química , Proteínas Mutantes/metabolismo , Ligação Proteica , Estrutura Terciária de Proteína , Eletricidade Estática
20.
Nat Cell Biol ; 8(10): 1149-54, 2006 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-16964244

RESUMO

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.


Assuntos
Sinalização do Cálcio/fisiologia , GMP Cíclico/metabolismo , Ativação do Canal Iônico , Canais Iônicos , Potássio/metabolismo , Transdução de Sinais , Espermatozoides/metabolismo , Animais , Arbacia/química , Cálcio/metabolismo , Quimiotaxia , Guanilato Ciclase/metabolismo , Masculino
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA