Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 25
Filter
1.
Cancer Immunol Immunother ; 72(1): 1-20, 2023 Jan.
Article in English | MEDLINE | ID: mdl-35654889

ABSTRACT

Lactic acidosis has been reported in solid tumor microenvironment (TME) including glioblastoma (GBM). In TME, several signaling molecules, growth factors and metabolites have been identified to induce resistance to chemotherapy and to sustain immune escape. In the early phases of the disease, microglia infiltrates TME, contributing to tumorigenesis rather than counteracting its growth. Insulin-like Growth Factor Binding Protein 6 (IGFBP6) is expressed during tumor development, and it is involved in migration, immune-escape and inflammation, thus providing an attractive target for GBM therapy. Here, we aimed at investigating the crosstalk between lactate metabolism and IGFBP6 in TME and GBM progression. Our results show that microglia exposed to lactate or IGFBP6 significantly increased the Monocarboxylate transporter 1 (MCT1) expression together with genes involved in mitochondrial metabolism. We, also, observed an increase in the M2 markers and a reduction of inducible nitric oxide synthase (iNOS) levels, suggesting a role of lactate/IGFBP6 metabolism in immune-escape activation. GBM cells exposed to lactate also showed increased levels of IGFBP6 and vice-versa. Such a phenomenon was coupled with a IGFBP6-mediated sonic hedgehog (SHH) ignaling increase. We, finally, tested our hypothesis in a GBM zebrafish animal model, where we observed an increase in microglia cells and igfbp6 gene expression after lactate exposure. Our results were confirmed by the analysis of human transcriptomes datasets and immunohistochemical assay from human GBM biopsies, suggesting the existence of a lactate/IGFBP6 crosstalk in microglial cells, so that IGFBP6 expression is regulated by lactate production in GBM cells and in turn modulates microglia polarization.


Subject(s)
Brain Neoplasms , Glioblastoma , Animals , Humans , Glioblastoma/pathology , Microglia/metabolism , Insulin-Like Growth Factor Binding Protein 6/metabolism , Insulin-Like Growth Factor Binding Protein 6/therapeutic use , Lactic Acid/metabolism , Lactic Acid/therapeutic use , Tumor Microenvironment , Zebrafish/metabolism , Cell Line, Tumor , Hedgehog Proteins , Brain Neoplasms/pathology
2.
Nature ; 547(7663): 364-368, 2017 07 20.
Article in English | MEDLINE | ID: mdl-28693035

ABSTRACT

Polymodal thermo- and mechanosensitive two-pore domain potassium (K2P) channels of the TREK subfamily generate 'leak' currents that regulate neuronal excitability, respond to lipids, temperature and mechanical stretch, and influence pain, temperature perception and anaesthetic responses. These dimeric voltage-gated ion channel (VGIC) superfamily members have a unique topology comprising two pore-forming regions per subunit. In contrast to other potassium channels, K2P channels use a selectivity filter 'C-type' gate as the principal gating site. Despite recent advances, poor pharmacological profiles of K2P channels limit mechanistic and biological studies. Here we describe a class of small-molecule TREK activators that directly stimulate the C-type gate by acting as molecular wedges that restrict interdomain interface movement behind the selectivity filter. Structures of K2P2.1 (also known as TREK-1) alone and with two selective K2P2.1 (TREK-1) and K2P10.1 (TREK-2) activators-an N-aryl-sulfonamide, ML335, and a thiophene-carboxamide, ML402-define a cryptic binding pocket unlike other ion channel small-molecule binding sites and, together with functional studies, identify a cation-π interaction that controls selectivity. Together, our data reveal a druggable K2P site that stabilizes the C-type gate 'leak mode' and provide direct evidence for K2P selectivity filter gating.


Subject(s)
Potassium Channels, Tandem Pore Domain/agonists , Potassium Channels, Tandem Pore Domain/chemistry , Animals , Arachidonic Acid/chemistry , Arachidonic Acid/metabolism , Arachidonic Acid/pharmacology , Benzamides/chemistry , Benzamides/metabolism , Benzamides/pharmacology , Binding Sites/drug effects , HEK293 Cells , Humans , Ion Channel Gating/drug effects , Lipids , Mice , Models, Molecular , Pichia , Potassium Channels, Tandem Pore Domain/metabolism , Protein Conformation/drug effects , Sulfonamides/chemistry , Sulfonamides/metabolism , Sulfonamides/pharmacology , Thiophenes/chemistry , Thiophenes/metabolism , Thiophenes/pharmacology , Xenopus laevis
3.
Nature ; 552(7685): 426-429, 2017 12 21.
Article in English | MEDLINE | ID: mdl-29236684

ABSTRACT

Calcium-activated chloride channels (CaCCs) encoded by TMEM16A control neuronal signalling, smooth muscle contraction, airway and exocrine gland secretion, and rhythmic movements of the gastrointestinal system. To understand how CaCCs mediate and control anion permeation to fulfil these physiological functions, knowledge of the mammalian TMEM16A structure and identification of its pore-lining residues are essential. TMEM16A forms a dimer with two pores. Previous CaCC structural analyses have relied on homology modelling of a homologue (nhTMEM16) from the fungus Nectria haematococca that functions primarily as a lipid scramblase, as well as subnanometre-resolution electron cryo-microscopy. Here we present de novo atomic structures of the transmembrane domains of mouse TMEM16A in nanodiscs and in lauryl maltose neopentyl glycol as determined by single-particle electron cryo-microscopy. These structures reveal the ion permeation pore and represent different functional states. The structure in lauryl maltose neopentyl glycol has one Ca2+ ion resolved within each monomer with a constricted pore; this is likely to correspond to a closed state, because a CaCC with a single Ca2+ occupancy requires membrane depolarization in order to open (C.J.P. et al., manuscript submitted). The structure in nanodiscs has two Ca2+ ions per monomer and its pore is in a closed conformation; this probably reflects channel rundown, which is the gradual loss of channel activity that follows prolonged CaCC activation in 1 mM Ca2+. Our mutagenesis and electrophysiological studies, prompted by analyses of the structures, identified ten residues distributed along the pore that interact with permeant anions and affect anion selectivity, as well as seven pore-lining residues that cluster near pore constrictions and regulate channel gating. Together, these results clarify the basis of CaCC anion conduction.


Subject(s)
Anoctamin-1/chemistry , Anoctamin-1/ultrastructure , Calcium/chemistry , Calcium/pharmacology , Cryoelectron Microscopy , Ion Channel Gating/drug effects , Animals , Anions/chemistry , Anions/metabolism , Anoctamin-1/metabolism , Calcium/metabolism , Glucosides/chemistry , HEK293 Cells , Humans , Ion Transport/drug effects , Mice , Models, Molecular , Nanostructures/chemistry , Nanostructures/ultrastructure , Protein Conformation/drug effects
4.
Nat Chem Biol ; 14(9): 870-875, 2018 09.
Article in English | MEDLINE | ID: mdl-30061717

ABSTRACT

Amyloids adopt 'cross-ß' structures composed of long, twisted fibrils with ß-strands running perpendicular to the fibril axis. Recently, a toxic peptide was proposed to form amyloid-like cross-α structures in solution, with a planar bilayer-like assembly observed in the crystal structure. Here we crystallographically characterize designed peptides that assemble into spiraling cross-α amyloid-like structures, which resemble twisted ß-amyloid fibrils. The peptides form helical dimers, stabilized by packing of small and apolar residues, and the dimers further assemble into cross-α amyloid-like fibrils with superhelical pitches ranging from 170 Å to 200 Å. When a small residue that appeared critical for packing was converted to leucine, it resulted in structural rearrangement to a helical polymer. Fluorescently tagged versions of the designed peptides form puncta in mammalian cells, which recover from photobleaching with markedly different kinetics. These structural folds could be potentially useful for directing in vivo protein assemblies with predetermined spacing and stabilities.


Subject(s)
Amyloid/chemistry , Peptides/chemistry , Crystallography, X-Ray , Humans , Kinetics , Models, Molecular , Peptides/chemical synthesis , Protein Conformation
6.
Nat Chem Biol ; 10(6): 457-62, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24776929

ABSTRACT

cAMP mediates autonomic regulation of heart rate by means of hyperpolarization-activated cyclic nucleotide-gated (HCN) channels, which underlie the pacemaker current If. cAMP binding to the C-terminal cyclic nucleotide binding domain enhances HCN open probability through a conformational change that reaches the pore via the C-linker. Using structural and functional analysis, we identified a binding pocket in the C-linker of HCN4. Cyclic dinucleotides, an emerging class of second messengers in mammals, bind the C-linker pocket (CLP) and antagonize cAMP regulation of the channel. Accordingly, cyclic dinucleotides prevent cAMP regulation of If in sinoatrial node myocytes, reducing heart rate by 30%. Occupancy of the CLP hence constitutes an efficient mechanism to hinder ß-adrenergic stimulation on If. Our results highlight the regulative role of the C-linker and identify a potential drug target in HCN4. Furthermore, these data extend the signaling scope of cyclic dinucleotides in mammals beyond their first reported role in innate immune system.


Subject(s)
Cyclic AMP/metabolism , Cyclic GMP/analogs & derivatives , Dinucleoside Phosphates/metabolism , Hyperpolarization-Activated Cyclic Nucleotide-Gated Channels/metabolism , Ion Channel Gating/physiology , Muscle Proteins/metabolism , Potassium Channels/metabolism , Animals , Binding Sites , Blotting, Western , Crystallography, X-Ray , Cyclic GMP/chemistry , Cyclic GMP/metabolism , Dinucleoside Phosphates/chemistry , HEK293 Cells , High-Throughput Screening Assays , Humans , Hyperpolarization-Activated Cyclic Nucleotide-Gated Channels/genetics , Ion Channel Gating/drug effects , Ligands , Mice , Mice, Inbred C57BL , Molecular Docking Simulation , Molecular Structure , Muscle Proteins/genetics , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , Patch-Clamp Techniques , Potassium Channels/genetics , Sinoatrial Node/cytology , Sinoatrial Node/drug effects , Sinoatrial Node/metabolism , Small Molecule Libraries/chemistry , Small Molecule Libraries/pharmacology , Transfection
7.
Biochim Biophys Acta ; 1827(6): 793-805, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23541892

ABSTRACT

Voltage-dependent anion selective channel isoform1 maintains the permeability of the outer mitochondrial membrane. Its voltage-gating properties are relevant in bioenergetic metabolism and apoptosis. The N-terminal domain is suspected to be involved in voltage-gating, due to its peculiar localization. However this issue is still controversial. In this work we exchanged or deleted the ß-strands that take contact with the N-terminal domain. The exchange of the whole hVDAC1 ß-barrel with the homologous hVDAC3 ß-barrel produces a chimeric protein that, in reconstituted systems, loses completely voltage-dependence. hVDAC3 ß-barrel has most residues in common with hVDAC1, including V143 and L150 considered anchor points for the N-terminus. hVDAC1 mutants completely lacking either the ß-strand 9 or both ß-strands 9 and 10 were expressed, refolded and reconstituted in artificial bilayers. The mutants formed smaller pores. Molecular dynamics simulations of the mutant structure supported its ability to form smaller pores. The mutant lacking both ß-strands 9 and 10 showed a new voltage-dependence feature resulting in a fully asymmetric behavior. These data indicate that a network of ß-strands in the pore-walls, and not single residues, are required for voltage-gating in addition to the N-terminus.


Subject(s)
Voltage-Dependent Anion Channel 1/chemistry , Amino Acid Sequence , Membrane Potentials , Models, Molecular , Molecular Dynamics Simulation , Molecular Sequence Data , Protein Structure, Secondary , Recombinant Fusion Proteins/chemistry , Sequence Alignment , Voltage-Dependent Anion Channel 1/physiology
8.
iScience ; 27(6): 109853, 2024 Jun 21.
Article in English | MEDLINE | ID: mdl-38784007

ABSTRACT

The voltage-dependent anion-selective channel isoform 1 (VDAC1) is a pivotal component in cellular metabolism and apoptosis with a prominent role in many cancer types, offering a unique therapeutic intervention point. Through an in-silico-to-in-vitro approach we identified a set of VA molecules (VDAC Antagonists) that selectively bind to VDAC1 and display specificity toward cancer cells. Biochemical characterization showed that VA molecules can directly interact with VDAC1 with micromolar affinity by competing with the endogenous ligand NADH for a partially shared binding site. NADH displacement results in mitochondrial distress and reduced cell proliferation, especially when compared to non-cancerous cells. Experiments performed on organoids derived from intrahepatic cholangiocarcinoma patients demonstrated a dose-dependent reduction in cell viability upon treatment with VA molecules with lower impact on healthy cells than conventional treatments like gemcitabine. VA molecules are chemical entities representing promising candidates for further optimization and development as cancer therapy strategies through precise metabolic interventions.

9.
Appl Microbiol Biotechnol ; 97(16): 7325-36, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23584245

ABSTRACT

In Gram-negative bacteria, production of the signal molecule c-di-GMP by diguanylate cyclases (DGCs) is a key trigger for biofilm formation, which, in turn, is often required for the development of chronic bacterial infections. Thus, DGCs represent interesting targets for new chemotherapeutic drugs with anti-biofilm activity. We searched for inhibitors of the WspR protein, a Pseudomonas aeruginosa DGC involved in biofilm formation and production of virulence factors, using a set of microbiological assays developed in an Escherichia coli strain expressing the wspR gene. We found that azathioprine, an immunosuppressive drug used in the treatment of Crohn's disease, was able to inhibit WspR-dependent c-di-GMP biosynthesis in bacterial cells. However, in vitro enzymatic assays ruled out direct inhibition of WspR DGC activity either by azathioprine or by its metabolic derivative 2-amino-6-mercapto-purine riboside. Azathioprine is an inhibitor of 5-aminoimidazole-4-carboxamide ribotide (AICAR) transformylase, an enzyme involved in purine biosynthesis, which suggests that inhibition of c-di-GMP biosynthesis by azathioprine may be due to perturbation of intracellular nucleotide pools. Consistent with this hypothesis, WspR activity is abolished in an E. coli purH mutant strain, unable to produce AICAR transformylase. Despite its effect on WspR, azathioprine failed to prevent biofilm formation by P. aeruginosa; however, it affected production of extracellular structures in E. coli clinical isolates, suggesting efficient inhibition of c-di-GMP biosynthesis in this bacterium. Our results indicate that azathioprine can prevent biofilm formation in E. coli through inhibition of c-di-GMP biosynthesis and suggest that such inhibition might contribute to its anti-inflammatory activity in Crohn's disease.


Subject(s)
Azathioprine/metabolism , Cyclic GMP/analogs & derivatives , Escherichia coli/drug effects , Nucleotides/antagonists & inhibitors , Pseudomonas aeruginosa/drug effects , Anti-Bacterial Agents/metabolism , Biofilms/drug effects , Biofilms/growth & development , Cyclic GMP/biosynthesis , Escherichia coli/metabolism , Escherichia coli/physiology , Pseudomonas aeruginosa/metabolism , Pseudomonas aeruginosa/physiology
10.
Nat Commun ; 14(1): 5521, 2023 09 08.
Article in English | MEDLINE | ID: mdl-37684224

ABSTRACT

The second messenger cyclic AMP regulates many nuclear processes including transcription, pre-mRNA splicing and mitosis. While most functions are attributed to protein kinase A, accumulating evidence suggests that not all nuclear cyclic AMP-dependent effects are mediated by this kinase, implying that other effectors may be involved. Here we explore the nuclear roles of Exchange Protein Activated by cyclic AMP 1. We find that it enters the nucleus where forms reversible biomolecular condensates in response to cyclic AMP. This phenomenon depends on intrinsically disordered regions present at its amino-terminus and is independent of protein kinase A. Finally, we demonstrate that nuclear Exchange Protein Activated by cyclic AMP 1 condensates assemble at genomic loci on chromosome 6 in the proximity of Histone Locus Bodies and promote the transcription of a histone gene cluster. Collectively, our data reveal an unexpected mechanism through which cyclic AMP contributes to nuclear spatial compartmentalization and promotes the transcription of specific genes.


Subject(s)
Cyclic AMP , Histones , Histones/genetics , Cell Nucleus , Nuclear Proteins , Cyclic AMP-Dependent Protein Kinases
11.
bioRxiv ; 2023 Oct 18.
Article in English | MEDLINE | ID: mdl-37905049

ABSTRACT

K2P potassium channels regulate excitability by affecting cellular resting membrane potential in the brain, cardiovascular system, immune cells, and sensory organs. Despite their important roles in anesthesia, arrhythmia, pain, hypertension, sleep, and migraine, the ability to control K2P function remains limited. Here, we describe a chemogenetic strategy termed CATKLAMP (Covalent Activation of TREK family K+ channels to cLAmp Membrane Potential) that leverages the discovery of a site in the K2P modulator pocket that reacts with electrophile-bearing derivatives of a TREK subfamily small molecule activator, ML335, to activate the channel irreversibly. We show that the CATKLAMP strategy can be used to probe fundamental aspects of K2P function, as a switch to silence neuronal firing, and is applicable to all TREK subfamily members. Together, our findings exemplify a new means to alter K2P channel activity that should facilitate studies both molecular and systems level studies of K2P function and enable the search for new K2P modulators.

12.
Cell Prolif ; 56(4): e13388, 2023 Apr.
Article in English | MEDLINE | ID: mdl-36794373

ABSTRACT

Metabolic changes of malignant plasma cells (PCs) and adaptation to tumour microenvironment represent one of the hallmarks of multiple myeloma (MM). We previously showed that MM mesenchymal stromal cells are more glycolytic and produce more lactate than healthy counterpart. Hence, we aimed to explore the impact of high lactate concentration on metabolism of tumour PCs and its impact on the efficacy of proteasome inhibitors (PIs). Lactate concentration was performed by colorimetric assay on MM patient's sera. The metabolism of MM cell treated with lactate was assessed by seahorse and real time Polymerase Chain Reaction (PCR). Cytometry was used to evaluate mitochondrial reactive oxygen species (mROS), apoptosis and mitochondrial depolarization. Lactate concentration resulted increased in MM patient's sera. Therefore, PCs were treated with lactate and we observed an increase of oxidative phosphorylation-related genes, mROS and oxygen consumption rate. Lactate supplementation exhibited a significant reduction in cell proliferation and less responsive to PIs. These data were confirmed by pharmacological inhibition of monocarboxylate transporter 1 (MCT1) by AZD3965 which was able to overcame metabolic protective effect of lactate against PIs. Consistently, high levels of circulating lactate caused expansion of Treg and monocytic myeloid derived suppressor cells and such effect was significantly reduced by AZD3965. Overall, these findings showed that targeting lactate trafficking in TME inhibits metabolic rewiring of tumour PCs, lactate-dependent immune evasion and thus improving therapy efficacy.


Subject(s)
Multiple Myeloma , Symporters , Humans , Lactic Acid/metabolism , Proteasome Inhibitors/pharmacology , Multiple Myeloma/drug therapy , Symporters/genetics , Symporters/metabolism , Cell Line, Tumor , Tumor Microenvironment
13.
J Biol Chem ; 286(52): 44811-20, 2011 Dec 30.
Article in English | MEDLINE | ID: mdl-22006928

ABSTRACT

Hyperpolarization-activated cyclic nucleotide-gated (HCN) channels are dually activated by hyperpolarization and binding of cAMP to their cyclic nucleotide binding domain (CNBD). HCN isoforms respond differently to cAMP; binding of cAMP shifts activation of HCN2 and HCN4 by 17 mV but shifts that of HCN1 by only 2-4 mV. To explain the peculiarity of HCN1, we solved the crystal structures and performed a biochemical-biophysical characterization of the C-terminal domain (C-linker plus CNBD) of the three isoforms. Our main finding is that tetramerization of the C-terminal domain of HCN1 occurs at basal cAMP concentrations, whereas those of HCN2 and HCN4 require cAMP saturating levels. Therefore, HCN1 responds less markedly than HCN2 and HCN4 to cAMP increase because its CNBD is already partly tetrameric. This is confirmed by voltage clamp experiments showing that the right-shifted position of V(½) in HCN1 is correlated with its propensity to tetramerize in vitro. These data underscore that ligand-induced CNBD tetramerization removes tonic inhibition from the pore of HCN channels.


Subject(s)
Cyclic AMP/metabolism , Ion Channel Gating/physiology , Ion Channels/chemistry , Ion Channels/metabolism , Protein Multimerization/physiology , Animals , Cyclic AMP/chemistry , Cyclic AMP/genetics , Humans , Hyperpolarization-Activated Cyclic Nucleotide-Gated Channels , Ion Channels/genetics , Oocytes , Potassium Channels , Protein Isoforms/chemistry , Protein Isoforms/genetics , Protein Isoforms/metabolism , Protein Structure, Tertiary , Xenopus laevis
14.
Nat Struct Mol Biol ; 29(6): 537-548, 2022 06.
Article in English | MEDLINE | ID: mdl-35655098

ABSTRACT

Every voltage-gated ion channel (VGIC) has a pore domain (PD) made from four subunits, each comprising an antiparallel transmembrane helix pair bridged by a loop. The extent to which PD subunit structure requires quaternary interactions is unclear. Here, we present crystal structures of a set of bacterial voltage-gated sodium channel (BacNaV) 'pore only' proteins that reveal a surprising collection of non-canonical quaternary arrangements in which the PD tertiary structure is maintained. This context-independent structural robustness, supported by molecular dynamics simulations, indicates that VGIC-PD tertiary structure is independent of quaternary interactions. This fold occurs throughout the VGIC superfamily and in diverse transmembrane and soluble proteins. Strikingly, characterization of PD subunit-binding Fabs indicates that non-canonical quaternary PD conformations can occur in full-length VGICs. Together, our data demonstrate that the VGIC-PD is an autonomously folded unit. This property has implications for VGIC biogenesis, understanding functional states, de novo channel design, and VGIC structural origins.


Subject(s)
Voltage-Gated Sodium Channels , Molecular Conformation , Molecular Dynamics Simulation , Voltage-Gated Sodium Channels/chemistry , Voltage-Gated Sodium Channels/metabolism
15.
Biol Chem ; 392(7): 617-24, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21627534

ABSTRACT

General diffusion porins are passive transmembrane channels. We have explored the possibility to create artificial nanopores starting from natural ß-barrel structures. Structural elements of bacterial porins were used to build a series of artificial nanopores. The basic module was selected by multi-alignment of general diffusion porins. The sequence corresponded to a highly conserved motif containing two ß-strands, which was obtained from Escherichia coli OmpF. Dimeric to octameric repeats were obtained through cDNA recombinant technology. The hexameric repeat was used to test its properties. This protein was expressed, purified and reconstituted in the planar bilayer membranes. It was able to form channels in membranes with a conductance of 300 pS in 150 mm KCl and did not show any relevant voltage-dependence.


Subject(s)
Nanopores , Porins/biosynthesis , Porins/metabolism , Recombinant Proteins/biosynthesis , Recombinant Proteins/metabolism , Amino Acid Sequence , Computational Biology , Escherichia coli/genetics , Escherichia coli/metabolism , Molecular Sequence Data , Porins/genetics , Protein Multimerization , Protein Structure, Tertiary , Recombinant Proteins/genetics , Sequence Alignment
16.
Methods Enzymol ; 653: 151-188, 2021.
Article in English | MEDLINE | ID: mdl-34099170

ABSTRACT

K2P (KCNK) potassium channels form 'background' or 'leak' currents that are important for controlling cell excitability in the brain, cardiovascular system, and somatosensory neurons. K2P2.1 (TREK-1) is one of the founding members of this family and one of the first well-characterized polymodal ion channels capable of responding to a variety of physical and chemical gating cues. Of the six K2P subfamilies, the thermo-and mechano-sensitive TREK subfamily comprising K2P2.1 (TREK-1), K2P4.1 (TRAAK), and K2P10.1 (TREK-2) is the first to have structures determined for each subfamily member. These structural studies have revealed key architectural features that provide a framework for understanding how gating cues sensed by different channel elements converge on the K2P selectivity filter C-type gate. TREK family structural studies have also revealed numerous sites where small molecules or lipids bind and affect channel function. This rich structural landscape provides the framework for probing K2P function and for the development of new K2P-directed agents. Such molecules may be useful for affecting processes where TREK channels are important such as anesthesia, pain, arrythmia, ischemia, migraine, intraocular pressure, and lung injury. Production of high quality protein samples is key to addressing new questions about K2P function and pharmacology. Here, we present methods for producing pure K2P2.1 (TREK-1) suitable for advancing towards these goals through structural and biochemical studies.


Subject(s)
Potassium Channels, Tandem Pore Domain , Neurons , Potassium Channels, Tandem Pore Domain/genetics
17.
Cell Chem Biol ; 27(5): 511-524.e4, 2020 05 21.
Article in English | MEDLINE | ID: mdl-32059793

ABSTRACT

The trinuclear ruthenium amine ruthenium red (RuR) inhibits diverse ion channels, including K2P potassium channels, TRPs, the calcium uniporter, CALHMs, ryanodine receptors, and Piezos. Despite this extraordinary array, there is limited information for how RuR engages targets. Here, using X-ray crystallographic and electrophysiological studies of an RuR-sensitive K2P, K2P2.1 (TREK-1) I110D, we show that RuR acts by binding an acidic residue pair comprising the "Keystone inhibitor site" under the K2P CAP domain archway above the channel pore. We further establish that Ru360, a dinuclear ruthenium amine not known to affect K2Ps, inhibits RuR-sensitive K2Ps using the same mechanism. Structural knowledge enabled a generalizable design strategy for creating K2P RuR "super-responders" having nanomolar sensitivity. Together, the data define a "finger in the dam" inhibition mechanism acting at a novel K2P inhibitor binding site. These findings highlight the polysite nature of K2P pharmacology and provide a new framework for K2P inhibitor development.


Subject(s)
Coloring Agents/pharmacology , Potassium Channels, Tandem Pore Domain/antagonists & inhibitors , Ruthenium Compounds/pharmacology , Ruthenium Red/pharmacology , Amines/chemistry , Amines/pharmacology , Animals , Coloring Agents/chemistry , Crystallography, X-Ray , Mice , Molecular Docking Simulation , Potassium Channels, Tandem Pore Domain/chemistry , Potassium Channels, Tandem Pore Domain/metabolism , Ruthenium/chemistry , Ruthenium/pharmacology , Ruthenium Compounds/chemistry , Ruthenium Red/chemistry
18.
Cells ; 9(5)2020 05 17.
Article in English | MEDLINE | ID: mdl-32429588

ABSTRACT

Human sperm cells express different aquaporins (AQPs), AQP3, 7, 8, 11, which are localized both in the plasma membrane and in intracellular structures. Besides cell volume regulation and end stage of cytoplasm removal during sperm maturation, the role of AQPs extends also to reactive oxygen species (ROS) elimination. Moreover, oxidative stress has been shown to inhibit AQP-mediated H2O2 permeability. A decrease in AQPs functionality is related to a decrease in sperm cells number and motility. Here we investigate the possible effect of human Papillomavirus (HPV) on both expression and function of AQPs in human sperm cells of patients undergoing infertility couple evaluation. Stopped-flow light-scattering experiments demonstrated that HPV infection heavily reduced water permeability of sperm cells in normospermic samples. Confocal immunofluorescence experiments showed a colocalization of HPV L1 protein with AQP8 (Pearson's correlation coefficient of 0.61), confirmed by co-immunoprecipitation experiments. No interaction of HPV with AQP3 and AQP7 was observed. A 3D model simulation of L1 protein and AQP8 interaction was also performed. Present findings may suggest that HPV infection directly inhibits AQP8 functionality and probably makes sperm cells more sensitive to oxidative stress.


Subject(s)
Aquaporins/antagonists & inhibitors , Papillomavirus Infections/metabolism , Papillomavirus Infections/pathology , Spermatozoa/metabolism , Spermatozoa/virology , Aquaporins/chemistry , Aquaporins/metabolism , Capsid Proteins/metabolism , Cell Membrane Permeability , DNA, Viral/analysis , Ejaculation , Humans , Infertility, Male/pathology , Infertility, Male/virology , Male , Molecular Docking Simulation , Oncogene Proteins, Viral/metabolism , Osmosis , Papillomaviridae/genetics , Semen/metabolism , Spermatozoa/pathology , Water
19.
Sci Adv ; 6(44)2020 10.
Article in English | MEDLINE | ID: mdl-33127683

ABSTRACT

K2P potassium channels regulate cellular excitability using their selectivity filter (C-type) gate. C-type gating mechanisms, best characterized in homotetrameric potassium channels, remain controversial and are attributed to selectivity filter pinching, dilation, or subtle structural changes. The extent to which such mechanisms control C-type gating of innately heterodimeric K2Ps is unknown. Here, combining K2P2.1 (TREK-1) x-ray crystallography in different potassium concentrations, potassium anomalous scattering, molecular dynamics, and electrophysiology, we uncover unprecedented, asymmetric, potassium-dependent conformational changes that underlie K2P C-type gating. These asymmetric order-disorder transitions, enabled by the K2P heterodimeric architecture, encompass pinching and dilation, disrupt the S1 and S2 ion binding sites, require the uniquely long K2P SF2-M4 loop and conserved "M3 glutamate network," and are suppressed by the K2P C-type gate activator ML335. These findings demonstrate that two distinct C-type gating mechanisms can operate in one channel and underscore the SF2-M4 loop as a target for K2P channel modulator development.

20.
Sci Adv ; 5(6): eaax2650, 2019 06.
Article in English | MEDLINE | ID: mdl-31223657

ABSTRACT

Dinoflagelates and cyanobacteria produce saxitoxin (STX), a lethal bis-guanidinium neurotoxin causing paralytic shellfish poisoning. A number of metazoans have soluble STX-binding proteins that may prevent STX intoxication. However, their STX molecular recognition mechanisms remain unknown. Here, we present structures of saxiphilin (Sxph), a bullfrog high-affinity STX-binding protein, alone and bound to STX. The structures reveal a novel high-affinity STX-binding site built from a "proto-pocket" on a transferrin scaffold that also bears thyroglobulin domain protease inhibitor repeats. Comparison of Sxph and voltage-gated sodium channel STX-binding sites reveals a convergent toxin recognition strategy comprising a largely rigid binding site where acidic side chains and a cation-π interaction engage STX. These studies reveal molecular rules for STX recognition, outline how a toxin-binding site can be built on a naïve scaffold, and open a path to developing protein sensors for environmental STX monitoring and new biologics for STX intoxication mitigation.


Subject(s)
Carrier Proteins/metabolism , Saxitoxin/metabolism , Amino Acid Sequence , Animals , Binding Sites/drug effects , Binding Sites/physiology , Cell Line , Cyanobacteria/metabolism , Humans , Peptide Hydrolases/metabolism , Protease Inhibitors/pharmacology , Protein Binding/drug effects , Protein Binding/physiology , Rana catesbeiana , Sf9 Cells , Sodium Channels/metabolism , Thyroglobulin/metabolism , Transferrin/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL