Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 26.940
Filter
1.
Development ; 151(13)2024 Jul 01.
Article in English | MEDLINE | ID: mdl-38958074

ABSTRACT

The bromodomain and extra-terminal (BET) family of proteins reads epigenetic histone acetylation marks on the genome and regulates the transcriptional machinery. In their study, Carole LaBonne and colleagues reveal the role of BET protein activity in the maintenance of pluripotency and establishment of the neural crest in Xenopus laevis. To know more about their work, we spoke to the first author Paul Huber and the corresponding author Carole LaBonne, Developmental and Stem Cell Biologist at Northwestern University.


Subject(s)
Xenopus laevis , Animals , History, 21st Century , Humans , History, 20th Century , Neural Crest/metabolism , Developmental Biology/history
2.
Proc Natl Acad Sci U S A ; 121(28): e2408346121, 2024 Jul 09.
Article in English | MEDLINE | ID: mdl-38968117

ABSTRACT

Xenopus embryos provide a favorable material to dissect the sequential steps that lead to dorsal-ventral (D-V) and anterior-posterior (A-P) cell differentiation. Here, we analyze the signaling pathways involved in this process using loss-of-function and gain-of-function approaches. The initial step was provided by Hwa, a transmembrane protein that robustly activates early ß-catenin signaling when microinjected into the ventral side of the embryo leading to complete twinned axes. The following step was the activation of Xenopus Nodal-related growth factors, which could rescue the depletion of ß-catenin and were themselves blocked by the extracellular Nodal antagonists Cerberus-Short and Lefty. During gastrulation, the Spemann-Mangold organizer secretes a cocktail of growth factor antagonists, of which the BMP antagonists Chordin and Noggin could rescue simultaneously D-V and A-P tissues in ß-catenin-depleted embryos. Surprisingly, this rescue occurred in the absence of any ß-catenin transcriptional activity as measured by ß-catenin activated Luciferase reporters. The Wnt antagonist Dickkopf (Dkk1) strongly synergized with the early Hwa signal by inhibiting late Wnt signals. Depletion of Sizzled (Szl), an antagonist of the Tolloid chordinase, was epistatic over the Hwa and Dkk1 synergy. BMP4 mRNA injection blocked Hwa-induced ectopic axes, and Dkk1 inhibited BMP signaling late, but not early, during gastrulation. Several unexpected findings were made, e.g., well-patterned complete embryonic axes are induced by Chordin or Nodal in ß-catenin knockdown embryos, dorsalization by Lithium chloride (LiCl) is mediated by Nodals, Dkk1 exerts its anteriorizing and dorsalizing effects by regulating late BMP signaling, and the Dkk1 phenotype requires Szl.


Subject(s)
Body Patterning , Intercellular Signaling Peptides and Proteins , Signal Transduction , Xenopus Proteins , beta Catenin , Animals , Body Patterning/genetics , Xenopus Proteins/metabolism , Xenopus Proteins/genetics , beta Catenin/metabolism , beta Catenin/genetics , Intercellular Signaling Peptides and Proteins/metabolism , Intercellular Signaling Peptides and Proteins/genetics , Xenopus laevis/embryology , Gene Expression Regulation, Developmental , Gastrulation , Nodal Protein/metabolism , Nodal Protein/genetics , Embryo, Nonmammalian/metabolism , Embryo, Nonmammalian/embryology , Organizers, Embryonic/metabolism , Glycoproteins
3.
J Gen Physiol ; 156(8)2024 Aug 05.
Article in English | MEDLINE | ID: mdl-38968404

ABSTRACT

We used voltage clamp fluorometry to probe the movement of the S4 helix in the voltage-sensing domain of the sea urchin HCN channel (spHCN) expressed in Xenopus oocytes. We obtained markedly different fluorescence responses with either ALEXA-488 or MTS-TAMRA covalently linked to N-terminal Cys332 of the S4 helix. With hyperpolarizing steps, ALEXA-488 fluorescence increased rapidly, consistent with it reporting the initial inward movement of S4, as previously described. In contrast, MTS-TAMRA fluorescence increased more slowly and its early phase correlated with that of channel opening. Additionally, a slow fluorescence component that tracked the development of the mode shift, or channel hysteresis, could be resolved with both labels. We quantitated this component as an increased deactivation tail current delay with concomitantly longer activation periods and found it to depend strongly on the presence of K+ ions in the pore. Using collisional quenching experiments and structural predictions, we established that ALEXA-488 was more exposed to solvent than MTS-TAMRA. We propose that components of S4 movement during channel activation can be kinetically resolved using different fluorescent probes to reveal distinct biophysical properties. Our findings underscore the need to apply caution when interpreting voltage clamp fluorometry data and demonstrate the potential utility of different labels to interrogate distinct biophysical properties of voltage-gated membrane proteins.


Subject(s)
Fluorescent Dyes , Xenopus laevis , Animals , Fluorescent Dyes/chemistry , Ion Channel Gating/physiology , Hyperpolarization-Activated Cyclic Nucleotide-Gated Channels/metabolism , Hyperpolarization-Activated Cyclic Nucleotide-Gated Channels/genetics , Hyperpolarization-Activated Cyclic Nucleotide-Gated Channels/chemistry , Oocytes/metabolism , Sea Urchins , Membrane Potentials/physiology
4.
Nat Commun ; 15(1): 5619, 2024 Jul 04.
Article in English | MEDLINE | ID: mdl-38965213

ABSTRACT

The sigma-1 receptor (σ1R) is a non-opioid membrane receptor, which responds to a diverse array of synthetic ligands to exert various pharmacological effects. Meanwhile, candidates for endogenous ligands of σ1R have also been identified. However, how endogenous ligands bind to σ1R remains unknown. Here, we present crystal structures of σ1R from Xenopus laevis (xlσ1R) bound to two endogenous neurosteroid ligands, progesterone (a putative antagonist) and dehydroepiandrosterone sulfate (DHEAS) (a putative agonist), at 2.15-3.09 Å resolutions. Both neurosteroids bind to a similar location in xlσ1R mainly through hydrophobic interactions, but surprisingly, with opposite binding orientations. DHEAS also forms hydrogen bonds with xlσ1R, whereas progesterone interacts indirectly with the receptor through water molecules near the binding site. Binding analyses are consistent with the xlσ1R-neurosteroid complex structures. Furthermore, molecular dynamics simulations and structural data reveal a potential water entry pathway. Our results provide insight into binding of two endogenous neurosteroid ligands to σ1R.


Subject(s)
Dehydroepiandrosterone Sulfate , Molecular Dynamics Simulation , Progesterone , Receptors, sigma , Sigma-1 Receptor , Xenopus laevis , Receptors, sigma/metabolism , Receptors, sigma/chemistry , Animals , Ligands , Binding Sites , Progesterone/metabolism , Progesterone/chemistry , Dehydroepiandrosterone Sulfate/metabolism , Dehydroepiandrosterone Sulfate/chemistry , Protein Binding , Crystallography, X-Ray , Neurosteroids/metabolism , Neurosteroids/chemistry , Hydrogen Bonding , Hydrophobic and Hydrophilic Interactions
5.
Toxins (Basel) ; 16(6)2024 Jun 03.
Article in English | MEDLINE | ID: mdl-38922152

ABSTRACT

Studies on the interaction sites of peptide toxins and ion channels typically involve site-directed mutations in toxins. However, natural mutant toxins exist among them, offering insights into how the evolutionary process has conserved crucial sequences for activities and molecular target selection. In this study, we present a comparative investigation using electrophysiological approaches and computational analysis between two alpha toxins from evolutionarily close scorpion species of the genus Tityus, namely, Tst3 and Ts3 from T. stigmurus and T. serrulatus, respectively. These toxins exhibit three natural substitutions near the C-terminal region, which is directly involved in the interaction between alpha toxins and Nav channels. Additionally, we characterized the activity of the Tst3 toxin on Nav1.1-Nav1.7 channels. The three natural changes between the toxins did not alter sensitivity to Nav1.4, maintaining similar intensities regarding their ability to alter opening probabilities, delay fast inactivation, and induce persistent currents. Computational analysis demonstrated a preference for the down conformation of VSD4 and a shift in the conformational equilibrium towards this state. This illustrates that the sequence of these toxins retained the necessary information, even with alterations in the interaction site region. Through electrophysiological and computational analyses, screening of the Tst3 toxin on sodium isoform revealed its classification as a classic α-NaTx with a broad spectrum of activity. It effectively delays fast inactivation across all tested isoforms. Structural analysis of molecular energetics at the interface of the VSD4-Tst3 complex further confirmed this effect.


Subject(s)
Scorpion Venoms , Scorpions , Scorpion Venoms/chemistry , Scorpion Venoms/genetics , Animals , Brazil , Humans , Xenopus laevis , Ion Channel Gating/drug effects , Amino Acid Sequence , Animals, Poisonous
6.
BMC Biotechnol ; 24(1): 44, 2024 Jun 26.
Article in English | MEDLINE | ID: mdl-38926833

ABSTRACT

BACKGROUND: Mammalian display is an appealing technology for therapeutic antibody development. Despite the advantages of mammalian display, such as full-length IgG display with mammalian glycosylation and its inherent ability to select antibodies with good biophysical properties, the restricted library size and large culture volumes remain challenges. Bxb1 serine integrase is commonly used for the stable genomic integration of antibody genes into mammalian cells, but presently lacks the efficiency required for the display of large mammalian display libraries. To increase the Bxb1 integrase-mediated stable integration efficiency, our study investigates factors that potentially affect the nuclear localization of Bxb1 integrase. METHODS: In an attempt to enhance Bxb1 serine integrase-mediated integration efficiency, we fused various nuclear localization signals (NLS) to the N- and C-termini of the integrase. Concurrently, we co-expressed multiple proteins associated with nuclear transport to assess their impact on the stable integration efficiency of green fluorescent protein (GFP)-encoding DNA and an antibody display cassette into the genome of Chinese hamster ovary (CHO) cells containing a landing pad for Bxb1 integrase-mediated integration. RESULTS: The nucleoplasmin NLS from Xenopus laevis, when fused to the C-terminus of Bxb1 integrase, demonstrated the highest enhancement in stable integration efficiency among the tested NLS fusions, exhibiting over a 6-fold improvement compared to Bxb1 integrase lacking an NLS fusion. Subsequent additions of extra NLS fusions to the Bxb1 integrase revealed an additional 131% enhancement in stable integration efficiency with the inclusion of two copies of C-terminal nucleoplasmin NLS fusions. Further improvement was achieved by co-expressing the Ran GTPase-activating protein (RanGAP). Finally, to validate the applicability of these findings to more complex proteins, the DNA encoding the membrane-bound clinical antibody abrilumab was stably integrated into the genome of CHO cells using Bxb1 integrase with two copies of C-terminal nucleoplasmin NLS fusions and co-expression of RanGAP. This approach demonstrated over 14-fold increase in integration efficiency compared to Bxb1 integrase lacking an NLS fusion. CONCLUSIONS: This study demonstrates that optimizing the NLS sequence fusion for Bxb1 integrase significantly enhances the stable genomic integration efficiency. These findings provide a practical approach for constructing larger libraries in mammalian cells through the stable integration of genes into a genomic landing pad.


Subject(s)
Cricetulus , Integrases , Nuclear Localization Signals , Animals , CHO Cells , Integrases/metabolism , Integrases/genetics , Nuclear Localization Signals/metabolism , Nuclear Localization Signals/genetics , Cell Nucleus/metabolism , Cell Nucleus/genetics , Serine/metabolism , Green Fluorescent Proteins/metabolism , Green Fluorescent Proteins/genetics , Cricetinae , Xenopus laevis/metabolism
7.
Development ; 151(13)2024 Jul 01.
Article in English | MEDLINE | ID: mdl-38884356

ABSTRACT

Neural crest cells are a stem cell population unique to vertebrate embryos that retains broad multi-germ layer developmental potential through neurulation. Much remains to be learned about the genetic and epigenetic mechanisms that control the potency of neural crest cells. Here, we examine the role that epigenetic readers of the BET (bromodomain and extra terminal) family play in controlling the potential of pluripotent blastula and neural crest cells. We find that inhibiting BET activity leads to loss of pluripotency at blastula stages and a loss of neural crest at neurula stages. We compare the effects of HDAC (an eraser of acetylation marks) and BET (a reader of acetylation) inhibition and find that they lead to similar cellular outcomes through distinct effects on the transcriptome. Interestingly, loss of BET activity in cells undergoing lineage restriction is coupled to increased expression of genes linked to pluripotency and prolongs the competence of initially pluripotent cells to transit to a neural progenitor state. Together these findings advance our understanding of the epigenetic control of pluripotency and the formation of the vertebrate neural crest.


Subject(s)
Neural Crest , Animals , Neural Crest/cytology , Neural Crest/metabolism , Epigenesis, Genetic , Gene Expression Regulation, Developmental , Xenopus Proteins/metabolism , Xenopus Proteins/genetics , Xenopus laevis/embryology , Blastula/metabolism , Blastula/cytology , Cell Differentiation , Pluripotent Stem Cells/cytology , Pluripotent Stem Cells/metabolism , Transcriptome/genetics
8.
Int J Mol Sci ; 25(11)2024 May 23.
Article in English | MEDLINE | ID: mdl-38891889

ABSTRACT

The sodium-glucose co-transporter-2 (SGLT2) inhibitor dapagliflozin is increasingly used in the treatment of diabetes and heart failure. Dapagliflozin has been associated with reduced incidence of atrial fibrillation (AF) in clinical trials. We hypothesized that the favorable antiarrhythmic outcome of dapagliflozin use may be caused in part by previously unrecognized effects on atrial repolarizing potassium (K+) channels. This study was designed to assess direct pharmacological effects of dapagliflozin on cloned ion channels Kv11.1, Kv1.5, Kv4.3, Kir2.1, K2P2.1, K2P3.1, and K2P17.1, contributing to IKur, Ito, IKr, IK1, and IK2P K+ currents. Human channels coded by KCNH2, KCNA5, KCND3, KCNJ2, KCNK2, KCNK3, and KCNK17 were heterologously expressed in Xenopus laevis oocytes, and currents were recorded using the voltage clamp technique. Dapagliflozin (100 µM) reduced Kv11.1 and Kv1.5 currents, whereas Kir2.1, K2P2.1, and K2P17.1 currents were enhanced. The drug did not significantly affect peak current amplitudes of Kv4.3 or K2P3.1 K+ channels. Biophysical characterization did not reveal significant effects of dapagliflozin on current-voltage relationships of study channels. In conclusion, dapagliflozin exhibits direct functional interactions with human atrial K+ channels underlying IKur, IKr, IK1, and IK2P currents. Substantial activation of K2P2.1 and K2P17.1 currents could contribute to the beneficial antiarrhythmic outcome associated with the drug. Indirect or chronic effects remain to be investigated in vivo.


Subject(s)
Benzhydryl Compounds , Glucosides , Sodium-Glucose Transporter 2 Inhibitors , Xenopus laevis , Humans , Glucosides/pharmacology , Sodium-Glucose Transporter 2 Inhibitors/pharmacology , Benzhydryl Compounds/pharmacology , Animals , Potassium Channels/metabolism , Oocytes/metabolism , Oocytes/drug effects , Sodium-Glucose Transporter 2/metabolism , Sodium-Glucose Transporter 2/genetics
9.
Commun Biol ; 7(1): 746, 2024 Jun 19.
Article in English | MEDLINE | ID: mdl-38898124

ABSTRACT

DNA methylation maintenance is essential for cell fate inheritance. In differentiated cells, this involves orchestrated actions of DNMT1 and UHRF1. In mice, the high-affinity binding of DPPA3 to the UHRF1 PHD finger regulates UHRF1 chromatin dissociation and cytosolic localization, which is required for oocyte maturation and early embryo development. However, the human DPPA3 ortholog functions during these stages remain unclear. Here, we report the structural basis for human DPPA3 binding to the UHRF1 PHD finger. The conserved human DPPA3 85VRT87 motif binds to the acidic surface of UHRF1 PHD finger, whereas mouse DPPA3 binding additionally utilizes two unique α-helices. The binding affinity of human DPPA3 for the UHRF1 PHD finger was weaker than that of mouse DPPA3. Consequently, human DPPA3, unlike mouse DPPA3, failed to inhibit UHRF1 chromatin binding and DNA remethylation in Xenopus egg extracts effectively. Our data provide novel insights into the distinct function and structure of human DPPA3.


Subject(s)
CCAAT-Enhancer-Binding Proteins , Ubiquitin-Protein Ligases , Animals , Humans , Mice , Amino Acid Sequence , CCAAT-Enhancer-Binding Proteins/metabolism , CCAAT-Enhancer-Binding Proteins/genetics , CCAAT-Enhancer-Binding Proteins/chemistry , Chromatin/metabolism , DNA Methylation , PHD Zinc Fingers/genetics , Protein Binding , Ubiquitin-Protein Ligases/metabolism , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/chemistry , Xenopus laevis/metabolism
10.
Mol Cells ; 47(6): 100076, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38825188

ABSTRACT

The actin-based cytoskeleton is considered a fundamental driving force for cell differentiation and development. Destrin (Dstn), a member of the actin-depolymerizing factor family, regulates actin dynamics by treadmilling actin filaments and increasing globular actin pools. However, the specific developmental roles of dstn have yet to be fully elucidated. Here, we investigated the physiological functions of dstn during early embryonic development using Xenopus laevis as an experimental model organism. dstn is expressed in anterior neural tissue and neural plate during Xenopus embryogenesis. Depleting dstn promoted morphants with short body axes and small heads. Moreover, dstn inhibition extended the neural plate region, impairing cell migration and distribution during neurulation. In addition to the neural plate, dstn knockdown perturbed neural crest cell migration. Our data suggest new insights for understanding the roles of actin dynamics in embryonic neural development, simultaneously presenting a new challenge for studying the complex networks governing cell migration involving actin dynamics.


Subject(s)
Cell Movement , Destrin , Embryonic Development , Xenopus laevis , Animals , Xenopus laevis/embryology , Xenopus laevis/metabolism , Destrin/metabolism , Destrin/genetics , Xenopus Proteins/metabolism , Xenopus Proteins/genetics , Neural Crest/metabolism , Neural Crest/embryology , Neural Crest/cytology , Neurogenesis , Neural Plate/metabolism , Neural Plate/embryology , Actins/metabolism , Gene Expression Regulation, Developmental
11.
Front Cell Infect Microbiol ; 14: 1394713, 2024.
Article in English | MEDLINE | ID: mdl-38836054

ABSTRACT

The rabies virus enters the nervous system by interacting with several molecular targets on host cells to modify behavior and trigger receptor-mediated endocytosis of the virion by poorly understood mechanisms. The rabies virus glycoprotein (RVG) interacts with the muscle acetylcholine receptor and the neuronal α4ß2 subtype of the nicotinic acetylcholine receptor (nAChR) family by the putative neurotoxin-like motif. Given that the neurotoxin-like motif is highly homologous to the α7 nAChR subtype selective snake toxin α-bungarotoxin (αBTX), other nAChR subtypes are likely involved. The purpose of this study is to determine the activity of the RVG neurotoxin-like motif on nAChR subtypes that are expressed in brain regions involved in rabid animal behavior. nAChRs were expressed in Xenopus laevis oocytes, and two-electrode voltage clamp electrophysiology was used to collect concentration-response data to measure the functional effects. The RVG peptide preferentially and completely inhibits α7 nAChR ACh-induced currents by a competitive antagonist mechanism. Tested heteromeric nAChRs are also inhibited, but to a lesser extent than the α7 subtype. Residues of the RVG peptide with high sequence homology to αBTX and other neurotoxins were substituted with alanine. Altered RVG neurotoxin-like peptides showed that residues phenylalanine 192, arginine 196, and arginine 199 are important determinants of RVG peptide apparent potency on α7 nAChRs, while serine 195 is not. The evaluation of the rabies ectodomain reaffirmed the observations made with the RVG peptide, illustrating a significant inhibitory impact on α7 nAChR with potency in the nanomolar range. In a mammalian cell culture model of neurons, we confirm that the RVG peptide binds preferentially to cells expressing the α7 nAChR. Defining the activity of the RVG peptide on nAChRs expands our understanding of basic mechanisms in host-pathogen interactions that result in neurological disorders.


Subject(s)
Glycoproteins , Rabies virus , Xenopus laevis , alpha7 Nicotinic Acetylcholine Receptor , alpha7 Nicotinic Acetylcholine Receptor/metabolism , Animals , Rabies virus/physiology , Rabies virus/metabolism , Humans , Glycoproteins/metabolism , Glycoproteins/genetics , Oocytes/metabolism , Viral Proteins/metabolism , Viral Proteins/genetics , Viral Envelope Proteins/metabolism , Viral Envelope Proteins/genetics , Host-Pathogen Interactions , Protein Binding , Rabies/metabolism , Rabies/virology , Acetylcholine/metabolism , Acetylcholine/pharmacology , Neurotoxins/metabolism , Neurotoxins/pharmacology
12.
Cell Mol Life Sci ; 81(1): 266, 2024 Jun 17.
Article in English | MEDLINE | ID: mdl-38880807

ABSTRACT

Acid-sensing ion channels (ASICs) are trimeric proton-gated cation channels that play a role in neurotransmission and pain sensation. The snake venom-derived peptides, mambalgins, exhibit potent analgesic effects in rodents by inhibiting central ASIC1a and peripheral ASIC1b. Despite their distinct species- and subtype-dependent pharmacology, previous structure-function studies have focussed on the mambalgin interaction with ASIC1a. Currently, the specific channel residues responsible for this pharmacological profile, and the mambalgin pharmacophore at ASIC1b remain unknown. Here we identify non-conserved residues at the ASIC1 subunit interface that drive differences in the mambalgin pharmacology from rat ASIC1a to ASIC1b, some of which likely do not make peptide binding interactions. Additionally, an amino acid variation below the core binding site explains potency differences between rat and human ASIC1. Two regions within the palm domain, which contribute to subtype-dependent effects for mambalgins, play key roles in ASIC gating, consistent with subtype-specific differences in the peptides mechanism. Lastly, there is a shared primary mambalgin pharmacophore for ASIC1a and ASIC1b activity, with certain peripheral peptide residues showing variant-specific significance for potency. Through our broad mutagenesis studies across various species and subtype variants, we gain a more comprehensive understanding of the pharmacophore and the intricate molecular interactions that underlie ligand specificity. These insights pave the way for the development of more potent and targeted peptide analogues required to advance our understating of human ASIC1 function and its role in disease.


Subject(s)
Acid Sensing Ion Channels , Elapid Venoms , Acid Sensing Ion Channels/metabolism , Acid Sensing Ion Channels/genetics , Acid Sensing Ion Channels/chemistry , Animals , Humans , Rats , Elapid Venoms/chemistry , Elapid Venoms/metabolism , Elapid Venoms/pharmacology , Elapid Venoms/genetics , Amino Acid Sequence , Binding Sites , Models, Molecular , Xenopus laevis , Peptides
13.
Methods Mol Biol ; 2796: 119-138, 2024.
Article in English | MEDLINE | ID: mdl-38856899

ABSTRACT

Ion channels comprise one of the largest targets for drug development and treatment and have been a subject of enduring fascination since first discovered in the 1950s. Over the past decades, thousands of publications have explored the cellular biology and molecular physiology of these proteins, and many channel structures have been determined since the late 1990s. Trying to connect the dots between ion channel function and structure, voltage clamp fluorometry (VCF) emerges as a powerful tool because it allows monitoring of the conformational rearrangements underlying the different functional states of the channel. This technique represents an elegant harmonization of molecular biology, electrophysiology, and fluorescence. In the following chapter, we will provide a concise guide to performing VCF on Xenopus laevis oocytes using the two-electrode voltage clamp (TEVC) modality. This is the most widely used configuration on Xenopus oocytes for its relative simplicity and demonstrated success in a number of different ion channels utilizing a variety of attached labels.


Subject(s)
Fluorometry , Ion Channels , Oocytes , Patch-Clamp Techniques , Xenopus laevis , Animals , Patch-Clamp Techniques/methods , Fluorometry/methods , Oocytes/metabolism , Ion Channels/metabolism , Ion Channel Gating
14.
J Gen Physiol ; 156(7)2024 Jul 01.
Article in English | MEDLINE | ID: mdl-38832889

ABSTRACT

Voltage-gated ion channels are responsible for the electrical excitability of neurons and cardiomyocytes. Thus, they are obvious targets for pharmaceuticals aimed to modulate excitability. Compounds activating voltage-gated potassium (KV) channels are expected to reduce excitability. To search for new KV-channel activators, we performed a high-throughput screen of 10,000 compounds on a specially designed Shaker KV channel. Here, we report on a large family of channel-activating compounds with a carboxyl (COOH) group as the common motif. The most potent COOH activators are lipophilic (4 < LogP <7) and are suggested to bind at the interface between the lipid bilayer and the channel's positively charged voltage sensor. The negatively charged form of the COOH-group compounds is suggested to open the channel by electrostatically pulling the voltage sensor to an activated state. Several of the COOH-group compounds also activate the therapeutically important KV7.2/7.3 channel and can thus potentially be developed into antiseizure drugs. The COOH-group compounds identified in this study are suggested to act via the same site and mechanism of action as previously studied COOH-group compounds, such as polyunsaturated fatty acids and resin acids, but distinct from sites for several other types of potassium channel-activating compounds.


Subject(s)
Ion Channel Gating , Animals , Ion Channel Gating/drug effects , Shaker Superfamily of Potassium Channels/metabolism , KCNQ2 Potassium Channel/metabolism , KCNQ2 Potassium Channel/agonists , Potassium Channels, Voltage-Gated/metabolism , Potassium Channels, Voltage-Gated/drug effects , KCNQ3 Potassium Channel/metabolism , Humans , Xenopus laevis
15.
Aquat Toxicol ; 272: 106975, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38824744

ABSTRACT

In this study, we investigated the effects of PVC microplastics (PVC-MPs) using two different animal models: the brittle star Ophiactis virens, and the African clawed frog Xenopus laevis. This is the first study using an environmental relevant sample of PVC-MPs obtained through mechanical fragmentation of a common PVC plumbing pipe. Exposure experiments on brittle star were performed on the adult stage for a duration of 14 days, while those on African clawed frog were performed on the embryogenic developmental stage according to the standardized FETAX protocol (Frog Embryo Teratogenesis Assay-Xenopus). For both models, different endpoints were analysed: mortality, developmental parameters, behavioural assays and histological analyses on target organs by optical and electronic microscopy. Results showed that the concentration of 0.1 µg mL-1 PVC do not cause any adverse effects in both models (common NOEC concentration), while exposure to 1 µg mL-1 PVC adversely affected at least one species (common LOEC concentration). In particular arm regeneration efficiency was the most affected parameters in O. virens leading to a significantly lower differentiation pattern at 1 µg mL-1 PVC. On the contrary, in X. laevis larvae histopathological analyses and behavioural tests were the most susceptible endpoints, exhibiting several abnormal figures and different swimming speed at 10 µg mL-1 PVC. Histopathological analyses revealed a higher abundance of degenerating cells, pyknotic nuclei and cellular debris in the gut of exposed larvae in respect to control. The comparative analyses performed in this work allowed to characterize the specificity of action of the PVC-MPs on the two species, underlining the importance of exploring a large spectrum of endpoints to offer adequate protection in the emerging fields of microplastic research.


Subject(s)
Microplastics , Polyvinyl Chloride , Water Pollutants, Chemical , Xenopus laevis , Animals , Polyvinyl Chloride/toxicity , Microplastics/toxicity , Water Pollutants, Chemical/toxicity , Embryo, Nonmammalian/drug effects , Larva/drug effects
16.
J Biol Chem ; 300(6): 107373, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38762183

ABSTRACT

Motile cilia on the cell surface produce fluid flows in the body and abnormalities in motile cilia cause primary ciliary dyskinesia. Dynein axonemal assembly factor 6 (DNAAF6), a causative gene of primary ciliary dyskinesia, was isolated as an interacting protein with La ribonucleoprotein 6 (LARP6) that regulates ciliogenesis in multiciliated cells (MCCs). In MCCs of Xenopus embryos, LARP6 and DNAAF6 were colocalized in biomolecular condensates termed dynein axonemal particles and synergized to control ciliogenesis. Moreover, tubulin alpha 1c-like mRNA encoding α-tubulin protein, that is a major component of ciliary axoneme, was identified as a target mRNA regulated by binding LARP6. While DNAAF6 was necessary for high α-tubulin protein expression near the apical side of Xenopus MCCs during ciliogenesis, its mutant, which abolishes binding with LARP6, was unable to restore the expression of α-tubulin protein near the apical side of MCCs in Xenopus DNAAF6 morphant. These results indicated that the binding of LARP6 and DNAAF6 in dynein axonemal particles regulates highly expressed α-tubulin protein near the apical side of Xenopus MCCs during ciliogenesis.


Subject(s)
Cilia , Ribonucleoproteins , Tubulin , Xenopus Proteins , Xenopus laevis , Cilia/metabolism , Animals , Ribonucleoproteins/metabolism , Ribonucleoproteins/genetics , Tubulin/metabolism , Xenopus Proteins/metabolism , Xenopus Proteins/genetics , Humans , SS-B Antigen , Autoantigens/metabolism , Autoantigens/genetics , Protein Binding , Axoneme/metabolism , RNA, Messenger/metabolism , RNA, Messenger/genetics
17.
J Neurosci ; 44(27)2024 Jul 03.
Article in English | MEDLINE | ID: mdl-38692734

ABSTRACT

Aberrant condensation and localization of the RNA-binding protein (RBP) fused in sarcoma (FUS) occur in variants of amyotrophic lateral sclerosis (ALS) and frontotemporal dementia (FTD). Changes in RBP function are commonly associated with changes in axonal cytoskeletal organization and branching in neurodevelopmental disorders. Here, we asked whether branching defects also occur in vivo in a model of FUS-associated disease. We use two reported Xenopus models of ALS/FTD (of either sex), the ALS-associated mutant FUS(P525L) and a mimic of hypomethylated FUS, FUS(16R). Both mutants strongly reduced axonal complexity in vivo. We also observed an axon looping defect for FUS(P525L) in the target area, which presumably arises due to errors in stop cue signaling. To assess whether the loss of axon complexity also had a cue-independent component, we assessed axonal cytoskeletal integrity in vitro. Using a novel combination of fluorescence and atomic force microscopy, we found that mutant FUS reduced actin density in the growth cone, altering its mechanical properties. Therefore, FUS mutants may induce defects during early axonal development.


Subject(s)
Amyotrophic Lateral Sclerosis , Axons , Frontotemporal Dementia , Mutation , RNA-Binding Protein FUS , RNA-Binding Protein FUS/genetics , RNA-Binding Protein FUS/metabolism , Axons/pathology , Axons/metabolism , Animals , Amyotrophic Lateral Sclerosis/genetics , Amyotrophic Lateral Sclerosis/pathology , Amyotrophic Lateral Sclerosis/metabolism , Frontotemporal Dementia/genetics , Frontotemporal Dementia/pathology , Frontotemporal Dementia/metabolism , Female , Male , Xenopus laevis , Growth Cones/metabolism , Humans , Disease Models, Animal
18.
Genetics ; 227(3)2024 Jul 08.
Article in English | MEDLINE | ID: mdl-38788202

ABSTRACT

Developmental and Epileptic Encephalopathies (DEE) are a genetically diverse group of severe, early onset seizure disorders. DEE are normally identified clinically in the first six months of life by the presence of frequent, difficult to control seizures and accompanying stalling or regression of development. DEE75 results from de novo mutations of the NEUROD2 gene that result in loss of activity of the encoded transcription factor, and the seizure phenotype was shown to be recapitulated in Xenopus tropicalis tadpoles. We used CRISPR/Cas9 to make a DEE75 model in Xenopus laevis, to further investigate the developmental etiology. NeuroD2.S CRISPR/Cas9 edited tadpoles were more active, swam faster on average, and had more seizures (C-shaped contractions resembling unprovoked C-start escape responses) than their sibling controls. Live imaging of Ca2+ signaling revealed prolongued, strong signals sweeping through the brain, indicative of neuronal hyperactivity. While the resulting tadpole brain appeared grossly normal, the blood-brain barrier (BBB) was found to be leakier than that of controls. Additionally, the TGFß antagonist Losartan was shown to have a short-term protective effect, reducing neuronal hyperactivity and reducing permeability of the BBB. Treatment of NeuroD2 CRISPant tadpoles with 5 mM Losartan decreased seizure events by more than 4-fold compared to the baseline. Our results support a model of DEE75 resulting from reduced NeuroD2 activity during vertebrate brain development, and indicate that a leaky BBB contributes to epileptogenesis.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors , Blood-Brain Barrier , Disease Models, Animal , Larva , Seizures , Xenopus Proteins , Xenopus laevis , Animals , Blood-Brain Barrier/metabolism , Larva/genetics , Seizures/genetics , Xenopus Proteins/genetics , Xenopus Proteins/metabolism , Basic Helix-Loop-Helix Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/metabolism , Brain/metabolism , Neurons/metabolism , Gene Knockdown Techniques , Epilepsy/genetics
19.
J Hazard Mater ; 472: 134462, 2024 Jul 05.
Article in English | MEDLINE | ID: mdl-38718506

ABSTRACT

The antiviral drugs favipiravir and oseltamivir are widely used to treat viral infections, including coronavirus 2019 (COVID-19), and their levels are expected to increase in the aquatic environment. In this study, the potential toxic and teratogenic effects of these drugs were evaluated using the frog embryo teratogenesis assay Xenopus (FETAX). In addition, glutathione S-transferase (GST), glutathione reductase (GR), catalase, carboxylesterase (CaE), and acetylcholinesterase (AChE) enzyme activities and malondialdehyde levels were measured as biochemical markers in embryos and tadpoles for comparative assessment of the sublethal effects of the test compounds. Prior to embryo exposure, drug concentrations in the exposure medium were measured with high-performance liquid chromatography. The 96-h median lethal concentration (LC50) was 137.9 and 32.3 mg/L for favipiravir and oseltamivir, respectively. The teratogenic index for favipiravir was 4.67. Both favipiravir and oseltamivir inhibited GR, CaE, and AChE activities in embryos, while favipiravir increased the GST and CaE activities in tadpoles. In conclusion, favipiravir, for which teratogenicity data are available in mammalian test organisms and human teratogenicity is controversial, inhibited Xenopus laevis embryo development and was teratogenic. In addition, sublethal concentrations of both drugs altered the biochemical responses in embryos and tadpoles, with differences between the developmental stages.


Subject(s)
Amides , Antiviral Agents , Embryo, Nonmammalian , Embryonic Development , Oseltamivir , Xenopus laevis , Animals , Antiviral Agents/toxicity , Oseltamivir/toxicity , Embryonic Development/drug effects , Amides/toxicity , Embryo, Nonmammalian/drug effects , Pyrazines/toxicity , COVID-19 , COVID-19 Drug Treatment , SARS-CoV-2/drug effects , Larva/drug effects , Teratogens/toxicity , Carboxylesterase/metabolism
20.
J Hazard Mater ; 472: 134535, 2024 Jul 05.
Article in English | MEDLINE | ID: mdl-38718515

ABSTRACT

As a frequently detected per- and polyfluoroalkyl substance in the environment, 6:6 perfluoroalkylhypophosphinic acid (6:6 PFPiA) is vulnerable to transformation in the liver of organisms, but the transformation in gut is still unclear. This study investigates the molecular mechanisms of 6:6 PFPiA transformation in the gut of Xenopus laevis upon a 28-day exposure in water. Before Day 16, a notable correlation (p = 0.03) was observed between the transformation product (PFHxPA) and cytochrome P450 (CYP450) enzyme concentration in gut. This suggests that CYP450 enzymes played an important role in the transformation of 6:6 PFPiA in the gut, which was verified by an in vitro incubation with gut tissues, and supported by the molecular docking results of 6:6 PFPiA binding with CYP450 enzymes. From the day 16, the CYP450 concentration in gut decreased by 31.3 % due to the damage caused by 6:6 PFPiA, leading to a decrease in the transformation capacity in gut, but the transformation rate was stronger than in liver. This was in contrast with the in vitro experiment, where transformation was stronger in liver. In the mean time, the abundance of Bacteroidota in gut increased, which released hydrolytic enzyme and then could participate in the transformation as well. This study reveals the potential of the gut in metabolizing environmental pollutants, and provides profound insights into the potential health risks caused by 6:6 PFPiA in organisms.


Subject(s)
Cytochrome P-450 Enzyme System , Gastrointestinal Microbiome , Xenopus laevis , Animals , Cytochrome P-450 Enzyme System/metabolism , Water Pollutants, Chemical/toxicity , Water Pollutants, Chemical/metabolism , Molecular Docking Simulation , Liver/enzymology , Liver/metabolism , Biotransformation , Organophosphorus Compounds/toxicity , Organophosphorus Compounds/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...