Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
Add more filters










Publication year range
1.
Sci Rep ; 13(1): 16699, 2023 10 04.
Article in English | MEDLINE | ID: mdl-37794029

ABSTRACT

Mucopolysaccharidosis type IIIB (MPS IIIB) is a rare and devastating childhood-onset lysosomal storage disease caused by complete loss of function of the lysosomal hydrolase α-N-acetylglucosaminidase. The lack of functional enzyme in MPS IIIB patients leads to the progressive accumulation of heparan sulfate throughout the body and triggers a cascade of neuroinflammatory and other biochemical processes ultimately resulting in severe mental impairment and early death in adolescence or young adulthood. The low prevalence and severity of the disease has necessitated the use of animal models to improve our knowledge of the pathophysiology and for the development of therapeutic treatments. In this study, we took a systematic approach to characterizing a classical mouse model of MPS IIIB. Using a series of histological, biochemical, proteomic and behavioral assays, we tested MPS IIIB mice at two stages: during the pre-symptomatic and early symptomatic phases of disease development, in order to validate previously described phenotypes, explore new mechanisms of disease pathology and uncover biomarkers for MPS IIIB. Along with previous findings, this study helps provide a deeper understanding of the pathology landscape of this rare disease with high unmet medical need and serves as an important resource to the scientific community.


Subject(s)
Mucopolysaccharidosis III , Humans , Mice , Animals , Young Adult , Adult , Child , Mucopolysaccharidosis III/genetics , Acetylglucosaminidase/genetics , Proteomics , Heparitin Sulfate , Hydrolases , Disease Models, Animal
2.
J Clin Invest ; 133(3)2023 02 01.
Article in English | MEDLINE | ID: mdl-36454649

ABSTRACT

Comprehensive cis-regulatory landscapes are essential for accurate enhancer prediction and disease variant mapping. Although cis-regulatory element (CRE) resources exist for most tissues and organs, many rare - yet functionally important - cell types remain overlooked. Despite representing only a small fraction of the heart's cellular biomass, the cardiac conduction system (CCS) unfailingly coordinates every life-sustaining heartbeat. To globally profile the mouse CCS cis-regulatory landscape, we genetically tagged CCS component-specific nuclei for comprehensive assay for transposase-accessible chromatin-sequencing (ATAC-Seq) analysis. Thus, we established a global CCS-enriched CRE database, referred to as CCS-ATAC, as a key resource for studying CCS-wide and component-specific regulatory functions. Using transcription factor (TF) motifs to construct CCS component-specific gene regulatory networks (GRNs), we identified and independently confirmed several specific TF sub-networks. Highlighting the functional importance of CCS-ATAC, we also validated numerous CCS-enriched enhancer elements and suggested gene targets based on CCS single-cell RNA-Seq data. Furthermore, we leveraged CCS-ATAC to improve annotation of existing human variants related to cardiac rhythm and nominated a potential enhancer-target pair that was dysregulated by a specific SNP. Collectively, our results established a CCS-regulatory compendium, identified novel CCS enhancer elements, and illuminated potential functional associations between human genomic variants and CCS component-specific CREs.


Subject(s)
Cell Nucleus , Chromatin , Heart Conduction System , Myocardial Contraction , Animals , Humans , Mice , Cell Nucleus/genetics , Chromatin/genetics , Gene Regulatory Networks , Myocardial Contraction/genetics , Myocardial Contraction/physiology , Transcription Factors/genetics , Heart Conduction System/physiology
4.
J Clin Invest ; 131(19)2021 10 01.
Article in English | MEDLINE | ID: mdl-34596051

ABSTRACT

The cardiac conduction system (CCS) ensures regular contractile function, and injury to any of its components can cause cardiac dysrhythmia. Although all cardiomyocytes (CMs) originate from common progenitors, the CCS is composed of biologically distinct cell types with unique functional and developmental characteristics. In contrast to ventricular cardiomyocytes, which continue to proliferate after birth, most CCS cells terminally exit the cell cycle during fetal development. Although the CCS should thus provide a poor substrate for postnatal injury repair, its regenerative capacity remains untested. Here, we describe a genetic system for ablating CMs that reside within the atrioventricular conduction system (AVCS). Adult mouse AVCS ablation resulted in regenerative failure characterized by persistent atrioventricular conduction defects and contractile dysfunction. In contrast, AVCS injury in neonatal mice led to recovery in a subset of these mice, thus providing evidence for CCS plasticity. Furthermore, CM proliferation did not appear to completely account for the observed functional recovery, suggesting that mechanisms regulating recovery from dysrhythmia are likely to be distinct from cardiac regeneration associated with ventricular injury. Taken together, we anticipate that our results will motivate further mechanistic studies of CCS plasticity and enable the exploration of rhythm restoration as an alternative therapeutic strategy.


Subject(s)
Atrioventricular Node/injuries , Myocytes, Cardiac/physiology , Regeneration/physiology , Animals , Atrioventricular Node/physiology , Cell Plasticity/physiology , Mice , Mice, Inbred C57BL
5.
Cephalalgia ; 41(5): 499-514, 2021 04.
Article in English | MEDLINE | ID: mdl-33626922

ABSTRACT

BACKGROUND: The clinical efficacy of migraine therapeutic agents directed towards the calcitonin-gene related peptide (CGRP) pathway has confirmed the key role of this axis in migraine pathogenesis. Three antibodies against CGRP - fremanezumab, galcanezumab and eptinezumab - and one antibody against the CGRP receptor, erenumab, are clinically approved therapeutics for the prevention of migraine. In addition, two small molecule CGRP receptor antagonists, ubrogepant and rimegepant, are approved for acute migraine treatment. Targeting either the CGRP ligand or receptor is efficacious for migraine treatment; however, a comparison of the mechanism of action of these therapeutic agents is lacking in the literature. METHODS: To gain insights into the potential differences between these CGRP pathway therapeutics, we compared the effect of a CGRP ligand antibody (fremanezumab), a CGRP receptor antibody (erenumab) and a CGRP receptor small molecule antagonist (telcagepant) using a combination of binding, functional and imaging assays. RESULTS: Erenumab and telcagepant antagonized CGRP, adrenomedullin and intermedin cAMP signaling at the canonical human CGRP receptor. In contrast, fremanezumab only antagonized CGRP-induced cAMP signaling at the human CGRP receptor. In addition, erenumab, but not fremanezumab, bound and internalized at the canonical human CGRP receptor. Interestingly, erenumab also bound and internalized at the human AMY1 receptor, a CGRP receptor family member. Both erenumab and telcagepant antagonized amylin-induced cAMP signaling at the AMY1 receptor while fremanezumab did not affect amylin responses. CONCLUSION: The therapeutic effect of agents targeting the CGRP ligand versus receptor for migraine prevention (antibodies) or acute treatment (gepants) may involve distinct mechanisms of action. These findings suggest that differing mechanisms could affect efficacy, safety, and/or tolerability in migraine patients.


Subject(s)
Antibodies, Monoclonal, Humanized/therapeutic use , Antibodies, Monoclonal/therapeutic use , Calcitonin Gene-Related Peptide Receptor Antagonists/therapeutic use , Calcitonin Gene-Related Peptide/immunology , Migraine Disorders/drug therapy , Migraine Disorders/prevention & control , Antibodies, Monoclonal/administration & dosage , Antibodies, Monoclonal, Humanized/administration & dosage , Azepines/therapeutic use , Calcitonin Gene-Related Peptide Receptor Antagonists/administration & dosage , Humans , Imidazoles/therapeutic use , Islet Amyloid Polypeptide , Receptors, Calcitonin Gene-Related Peptide
6.
Sci Rep ; 10(1): 4069, 2020 Feb 28.
Article in English | MEDLINE | ID: mdl-32107454

ABSTRACT

An amendment to this paper has been published and can be accessed via a link at the top of the paper.

7.
Cell Rep ; 27(12): 3486-3499.e6, 2019 06 18.
Article in English | MEDLINE | ID: mdl-31216470

ABSTRACT

Ectopic expression of transcription factors (TFs) can reprogram cell state. However, because of the large combinatorial space of possible TF cocktails, it remains difficult to identify TFs that reprogram specific cell types. Here, we develop Reprogram-Seq to experimentally screen thousands of TF cocktails for reprogramming performance. Reprogram-Seq leverages organ-specific cell-atlas data with single-cell perturbation and computational analysis to predict, evaluate, and optimize TF combinations that reprogram a cell type of interest. Focusing on the cardiac system, we perform Reprogram-Seq on MEFs using an undirected library of 48 cardiac factors and, separately, a directed library of 10 epicardial-related TFs. We identify a combination of three TFs, which efficiently reprogram MEFs to epicardial-like cells that are transcriptionally, molecularly, morphologically, and functionally similar to primary epicardial cells. Reprogram-Seq holds promise to accelerate the generation of specific cell types for regenerative medicine.


Subject(s)
Cellular Reprogramming/genetics , Computational Biology/methods , Pericardium/cytology , Pericardium/metabolism , Single-Cell Analysis/methods , Transcription Factors/metabolism , Activating Transcription Factor 3/genetics , Activating Transcription Factor 3/metabolism , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/metabolism , Cell Line , Embryonic Stem Cells/cytology , Embryonic Stem Cells/metabolism , Fibroblasts/cytology , Fibroblasts/metabolism , GATA6 Transcription Factor/genetics , GATA6 Transcription Factor/metabolism , Gene Expression Regulation, Developmental/genetics , Mice , RNA-Seq , Software , Transcription Factors/genetics
8.
Cell Rep ; 27(8): 2354-2369.e7, 2019 05 21.
Article in English | MEDLINE | ID: mdl-31116981

ABSTRACT

Gata4, Hand2, Mef2c, and Tbx5 (GHMT) can reprogram transduced fibroblasts into induced pacemaker-like myocytes (iPMs), but the underlying mechanisms remain obscure. Here, we explore the role of Hand2 in iPM formation by using a combination of transcriptome, genome, and biochemical assays. We found many shared transcriptional signatures between iPMs and the endogenous sinoatrial node (SAN), yet key regulatory networks remain missing. We demonstrate that Hand2 augments chromatin accessibility at loci involved in sarcomere organization, electrical coupling, and membrane depolarization. Focusing on an established cardiac Hand2 cistrome, we observe selective reorganization of chromatin accessibility to promote pacemaker-specific gene expression. Moreover, we identify a Hand2 cardiac subtype diversity (CSD) domain through biochemical analysis of the N terminus. By integrating our RNA-seq and ATAC-seq datasets, we highlight desmosome organization as a hallmark feature of iPM formation. Collectively, our results illuminate Hand2-dependent mechanisms that may guide future efforts to rationally improve iPM formation.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/metabolism , Cellular Reprogramming/genetics , Chromatin/metabolism , Desmosomes/metabolism , Gene Expression/genetics , Transcriptome/physiology
9.
PLoS One ; 14(4): e0214677, 2019.
Article in English | MEDLINE | ID: mdl-30939177

ABSTRACT

Recent studies have highlighted the extraordinary cell type diversity that exists within mammalian organs, yet the molecular drivers of such heterogeneity remain elusive. To address this issue, much attention has been focused on profiling the transcriptome and epigenome of individual cell types. However, standard cell type isolation methods based on surface or fluorescent markers remain problematic for cells residing within organs with significant connective tissue. Since the nucleus contains both genomic and transcriptomic information, the isolation of nuclei tagged in specific cell types (INTACT) method provides an attractive solution. Although INTACT has been successfully applied to plants, flies, zebrafish, frogs, and mouse brain and adipose tissue, broad use across mammalian organs remains challenging. Here we describe the PAN-INTACT method, which can be used to isolate cell type specific nuclei from fibrous mouse organs, which are particularly problematic. As a proof-of-concept, we demonstrate successful isolation of cell type-specific nuclei from the mouse heart, which contains substantial connective tissue and harbors multiple cell types, including cardiomyocytes, fibroblasts, endothelial cells, and epicardial cells. Compared to established techniques, PAN-INTACT allows more rapid isolation of cardiac nuclei to facilitate downstream applications. We show cell type-specific isolation of nuclei from the hearts of Nkx2-5Cre/+; R26Sun1-2xsf-GFP-6xmyc/+ mice, which we confirm by expression of lineage markers. Furthermore, we perform Assay for Transposase Accessible Chromatin (ATAC)-Seq to provide high-fidelity chromatin accessibility maps of Nkx2-5+ nuclei. To extend the applicability of PAN-INTACT, we also demonstrate successful isolation of Wt1+ podocytes from adult kidney. Taken together, our data suggest that PAN-INTACT is broadly applicable for profiling the transcriptional and epigenetic landscape of specific cell types. Thus, we envision that our method can be used to systematically probe mechanistic details of cell type-specific functions within individual organs of intact mice.


Subject(s)
Cell Nucleus/chemistry , Cell Separation/methods , Animals , Cell Nucleus/metabolism , Homeobox Protein Nkx-2.5/metabolism , Immunoprecipitation , Magnetics , Mice , Mice, Inbred C57BL , Myocardium/cytology , Myocardium/metabolism
10.
Sci Rep ; 9(1): 2106, 2019 02 14.
Article in English | MEDLINE | ID: mdl-30765799

ABSTRACT

The atrioventricular node (AVN) coordinates the timing of atrial and ventricular contraction to optimize cardiac performance. To study this critical function using mouse genetics, however, new reagents are needed that allow AVN-specific manipulation. Here we describe a novel Gjd3-CreEGFP mouse line that successfully recombines floxed alleles within the AVN beginning at E12.5. These mice have been engineered to express CreEGFP under the control of endogenous Gjd3 regulatory elements without perturbing native protein expression. Detailed histological analysis of Gjd3-CreEGFP mice reveals specific labeling of AVN cardiomyocytes and a subset of cardiac endothelial cells. Importantly, we show that Gjd3-CreEGFP mice have preserved cardiac mechanical and electrical function. In one application of our newly described mouse line, we provide a three-dimensional (3D) view of the AVN using tissue clearing combined with confocal microscopy. With this 3D model as a reference, we identify specific AVN sub-structures based on marker staining characteristics. In addition, we use our Gjd3-CreEGFP mice to guide microdissection of the AVN and construction of a single-cell atlas. Thus, our results establish a new transgenic tool for AVN-specific recombination, provide an updated model of AVN morphology, and describe a roadmap for exploring AVN cellular heterogeneity.


Subject(s)
Action Potentials , Atrioventricular Node/cytology , Atrioventricular Node/physiology , Connexins/physiology , Endothelial Cells/cytology , ErbB Receptors/metabolism , Myocytes, Cardiac/cytology , Animals , Endothelial Cells/metabolism , ErbB Receptors/genetics , Gene Knock-In Techniques , Heart Atria/cytology , Heart Atria/physiopathology , Integrases/metabolism , Mice , Myocytes, Cardiac/metabolism
11.
Dev Dyn ; 248(2): 173-188, 2019 02.
Article in English | MEDLINE | ID: mdl-30444277

ABSTRACT

BACKGROUND: Many human gene mutations have been linked to congenital heart disease (CHD), yet CHD remains a major health issue worldwide due in part to an incomplete understanding of the molecular basis for cardiac malformation. RESULTS: Here we identify the orthologous mouse Pou6f1 and zebrafish pouC as POU homeodomain transcription factors enriched in the developing heart. We find that pouC is a multi-functional transcriptional regulator containing separable activation, repression, protein-protein interaction, and DNA binding domains. Using zebrafish heart development as a model system, we demonstrate that pouC knockdown impairs cardiac morphogenesis and affects cardiovascular function. We also find that levels of pouC expression must be fine-tuned to enable proper heart formation. At the cellular level, we demonstrate that pouC knockdown disrupts atrioventricular canal (AVC) cardiomyocyte maintenance, although chamber myocyte specification remains intact. Mechanistically, we show that pouC binds a bmp4 intronic regulatory element to mediate transcriptional activation. CONCLUSIONS: Taken together, our study establishes pouC as a novel transcriptional input into the regulatory hierarchy that drives AVC morphogenesis in zebrafish. We anticipate that these findings will inform future efforts to explore functional conservation in mammals and potential association with atrioventricular septal defects in humans. Developmental Dynamics 248:173-188, 2019. © 2018 Wiley Periodicals, Inc.


Subject(s)
Bone Morphogenetic Protein 4/genetics , Gene Expression Regulation, Developmental , Heart Septum/growth & development , POU Domain Factors/physiology , Zebrafish Proteins/genetics , Zebrafish Proteins/physiology , Animals , Bone Morphogenetic Protein 4/metabolism , Heart/embryology , Heart/growth & development , Heart Septal Defects , Heart Septum/embryology , Mice , POU Domain Factors/metabolism , Protein Binding , Transcription Factors , Zebrafish , Zebrafish Proteins/metabolism
12.
PLoS One ; 12(3): e0174517, 2017.
Article in English | MEDLINE | ID: mdl-28358866

ABSTRACT

The cardiac conduction system (CCS) is composed of specialized cardiomyocytes that initiate and maintain cardiac rhythm. Any perturbation to the normal sequence of electrical events within the heart can result in cardiac arrhythmias. To understand how cardiac rhythm is established at the molecular level, several genetically modified mouse lines expressing Cre recombinase within specific CCS compartments have been created. In general, Cre driver lines have been generated either by homologous recombination of Cre into an endogenous locus or Cre expression driven by a randomly inserted transgene. However, haploinsufficiency of the endogenous gene compromises the former approach, while position effects negatively impact the latter. To address these limitations, we generated a Cre driver line for the ventricular conduction system (VCS) that preserves endogenous gene expression by targeting the Contactin2 (Cntn2) 3' untranslated region (3'UTR). Here we show that Cntn23'UTR-IRES-Cre-EGFP/+ mice recombine floxed alleles within the VCS and that Cre expression faithfully recapitulates the spatial distribution of Cntn2 within the heart. We further demonstrate that Cre expression initiates after birth with preservation of native Cntn2 protein. Finally, we show that Cntn23'UTR-IRES-Cre-EGFP/+ mice maintain normal cardiac mechanical and electrical function. Taken together, our results establish a novel VCS-specific Cre driver line without the adverse consequences of haploinsufficiency or position effects. We expect that our new mouse line will add to the accumulating toolkit of CCS-specific mouse reagents and aid characterization of the cell-autonomous molecular circuitry that drives VCS maintenance and function.


Subject(s)
Arrhythmias, Cardiac/genetics , Brugada Syndrome/genetics , Contactin 2/genetics , Heart Conduction System , 3' Untranslated Regions , Animals , Arrhythmias, Cardiac/physiopathology , Brugada Syndrome/physiopathology , Cardiac Conduction System Disease , Contactin 2/biosynthesis , Disease Models, Animal , Gene Targeting , Haploinsufficiency/genetics , Heart Ventricles/metabolism , Heart Ventricles/physiopathology , Homologous Recombination/genetics , Humans , Integrases/genetics , Mice , Mice, Transgenic , Myocytes, Cardiac/metabolism , Phenotype
13.
Mol Cell Biol ; 35(4): 649-61, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25487574

ABSTRACT

The cardiac conduction system coordinates electrical activation through a series of interconnected structures, including the atrioventricular node (AVN), the central connection point that delays impulse propagation to optimize cardiac performance. Although recent studies have uncovered important molecular details of AVN formation, relatively little is known about the transcriptional mechanisms that regulate AV delay, the primary function of the mature AVN. We identify here MyoR as a novel transcription factor expressed in Cx30.2(+) cells of the AVN. We show that MyoR specifically inhibits a Cx30.2 enhancer required for AVN-specific gene expression. Furthermore, we demonstrate that MyoR interacts directly with Gata4 to mediate transcriptional repression. Our studies reveal that MyoR contains two nonequivalent repression domains. While the MyoR C-terminal repression domain inhibits transcription in a context-dependent manner, the N-terminal repression domain can function in a heterologous context to convert the Hand2 activator into a repressor. In addition, we show that genetic deletion of MyoR in mice increases Cx30.2 expression by 50% and prolongs AV delay by 13%. Taken together, we conclude that MyoR modulates a Gata4-dependent regulatory circuit that establishes proper AV delay, and these findings may have wider implications for the variability of cardiac rhythm observed in the general population.


Subject(s)
Atrioventricular Node/metabolism , GATA4 Transcription Factor/metabolism , Signal Transduction/genetics , Transcription Factors/metabolism , Animals , Atrioventricular Node/cytology , Basic Helix-Loop-Helix Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/metabolism , Binding Sites , COS Cells , Chlorocebus aethiops , Connexins/genetics , Connexins/metabolism , Embryo, Mammalian , Female , GATA4 Transcription Factor/genetics , Gene Expression Regulation , Genes, Reporter , Heart Rate/physiology , Luciferases/genetics , Luciferases/metabolism , Male , Mice , Mice, Transgenic , Protein Binding , Protein Interaction Domains and Motifs , Transcription Factors/genetics , Transcription, Genetic , beta-Galactosidase/genetics , beta-Galactosidase/metabolism
14.
Development ; 141(22): 4267-78, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25344074

ABSTRACT

Various combinations of cardiogenic transcription factors, including Gata4 (G), Hand2 (H), Mef2c (M) and Tbx5 (T), can reprogram fibroblasts into induced cardiac-like myocytes (iCLMs) in vitro and in vivo. Given that optimal cardiac function relies on distinct yet functionally interconnected atrial, ventricular and pacemaker (PM) cardiomyocytes (CMs), it remains to be seen which subtypes are generated by direct reprogramming and whether this process can be harnessed to produce a specific CM of interest. Here, we employ a PM-specific Hcn4-GFP reporter mouse and a spectrum of CM subtype-specific markers to investigate the range of cellular phenotypes generated by reprogramming of primary fibroblasts. Unexpectedly, we find that a combination of four transcription factors (4F) optimized for Hcn4-GFP expression does not generate beating PM cells due to inadequate sarcomeric protein expression and organization. However, applying strict single-cell criteria to GHMT-reprogrammed cells, we observe induction of diverse cellular phenotypes, including those resembling immature forms of all three major cardiac subtypes (i.e. atrial, ventricular and pacemaker). In addition, we demonstrate that cells induced by GHMT are directly reprogrammed and do not arise from an Nxk2.5(+) progenitor cell intermediate. Taken together, our results suggest a remarkable degree of plasticity inherent to GHMT reprogramming and provide a starting point for optimization of CM subtype-specific reprogramming protocols.


Subject(s)
Cell Differentiation/physiology , Embryonic Induction/physiology , Fibroblasts/cytology , Heart/embryology , Myocytes, Cardiac/physiology , Transcription Factors/metabolism , Action Potentials/physiology , Analysis of Variance , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/metabolism , DNA Primers/genetics , Fibroblasts/metabolism , Fibroblasts/physiology , GATA4 Transcription Factor/genetics , GATA4 Transcription Factor/metabolism , Green Fluorescent Proteins/metabolism , Hyperpolarization-Activated Cyclic Nucleotide-Gated Channels/metabolism , Immunohistochemistry , MEF2 Transcription Factors/genetics , MEF2 Transcription Factors/metabolism , Mice , Myocytes, Cardiac/cytology , Real-Time Polymerase Chain Reaction , T-Box Domain Proteins/genetics , T-Box Domain Proteins/metabolism , Transcription Factors/genetics
15.
J Neurochem ; 121(3): 349-61, 2012 May.
Article in English | MEDLINE | ID: mdl-22309577

ABSTRACT

The nicotinic acetylcholine receptor (nAChR) ß3 subunit is thought to serve an accessory role in nAChR subtypes expressed in dopaminergic regions implicated in drug dependence and reward. When ß3 subunits are expressed in excess, they have a dominant-negative effect on function of selected nAChR subtypes. In this study, we show, in Xenopus oocytes expressing α2, α3 or α4 plus either ß2 or ß4 subunits, that in the presumed presence of similar amounts of each nAChR subunit, co-expression with wild-type ß3 subunits generally (except for α3*-nAChR) lowers amplitudes of agonist-evoked, inward peak currents by 20-50% without having dramatic effects (≤ 2-fold) on agonist potencies. By contrast, co-expression with mutant ß3(V9'S) subunits generally (except for α4ß2*-nAChR) increases agonist potencies, consistent with an expected gain-of-function effect. This most dramatically demonstrates formation of complexes containing three kinds of subunit. Moreover, for oocytes expressing nAChR containing any α subunit plus ß4 and ß3(V9'S) subunits, there is spontaneous channel opening sensitive to blockade by the open channel blocker, atropine. Collectively, the results indicate that ß3 subunits integrate into all of the studied receptor assemblies and suggest that natural co-expression with ß3 subunits can influence levels of expression and agonist sensitivities of several nAChR subtypes.


Subject(s)
Receptors, Nicotinic/physiology , Acetylcholine/pharmacology , Animals , Atropine/pharmacology , Cloning, Molecular , Data Interpretation, Statistical , Female , Humans , Ion Channel Gating/drug effects , Ion Channel Gating/physiology , Muscarinic Antagonists/pharmacology , Mutagenesis , Mutation/genetics , Nicotine/pharmacology , Nicotinic Agonists/pharmacology , Oocytes/metabolism , Patch-Clamp Techniques , Plasmids , RNA, Complementary/administration & dosage , RNA, Complementary/genetics , Receptors, Nicotinic/drug effects , Transcription, Genetic , Xenopus , Xenopus Proteins
16.
Cell Physiol Biochem ; 28(2): 239-50, 2011.
Article in English | MEDLINE | ID: mdl-21865731

ABSTRACT

BACKGROUND/AIMS: The effect of daily injections with genistein (naturally occurring phytoestrogen) on intestinal chloride (Cl(-)) secretion was measured with Ussing chamber short circuit current (I(sc), µA/cm(2)), in C57BL/6J male and female mice, using 600 mg/kg genistein/day (600G), 300 mg/kg genistein/day (300G), 150 mg/kg genistein/day (150G) or genistein-free vehicle control (0G) for 1- or 2-weeks. METHODS AND RESULTS: Injecting with 600G elicited significant increases in basal I(sc) in females after 1-week (ñ70 µA/cm(2), n=15, p < 0.05) and in males after 2-weeks (ñ80 µA/cm(2), n=5, p < 0.05) compared to their 0G counterparts. Chloride-free ringer significantly reduced basal I(sc) by 65% in 600G males and 72% in 600G females, suggesting that Cl(-) was the major anion comprising the genistein-stimulated secretion. The forskolin-stimulated (10 µM) I(sc) was significantly inhibited by the CFTR chloride channel inhibitors, glibenclamide (500 µM) and CFTR(inh)-172 (100 µM) in 600G males and females, suggesting some contribution by genistein-dependent CFTR-mediated Cl(-) secretion. We found no associated changes in intestinal morphology, nor change in total CFTR protein with 600G. There was a 5% increase in apical/subapical ratio in 600G males compared to controls (no change in females). CONCLUSION: These data suggest that male and female mice both exhibit increased Cl- secretion with 600G, however, the mechanisms mediating this are gender-dependent.


Subject(s)
Anticarcinogenic Agents/pharmacology , Chlorides/metabolism , Genistein/pharmacology , Intestine, Small/drug effects , Intestine, Small/metabolism , Animals , Anticarcinogenic Agents/administration & dosage , Anticarcinogenic Agents/blood , Colforsin/pharmacology , Cystic Fibrosis Transmembrane Conductance Regulator/antagonists & inhibitors , Cystic Fibrosis Transmembrane Conductance Regulator/metabolism , Female , Genistein/administration & dosage , Genistein/blood , Glyburide/pharmacology , Intestine, Small/pathology , Male , Mice , Mice, Inbred C57BL , Sex Factors
17.
J Biol Chem ; 286(44): 37976-37989, 2011 Nov 04.
Article in English | MEDLINE | ID: mdl-21832048

ABSTRACT

Despite the apparent function of naturally expressed mammalian α6*-nicotinic acetylcholine receptors (α6*-nAChR; where * indicates the known or possible presence of additional subunits), their functional and heterologous expression has been difficult. Here, we report that coexpression with wild-type ß3 subunits abolishes the small amount of function typically seen for all-human or all-mouse α6ß4*-nAChR expressed in Xenopus oocytes. However, levels of function and agonist potencies are markedly increased, and there is atropine-sensitive blockade of spontaneous channel opening upon coexpression of α6 and ß4 subunits with mutant ß3 subunits harboring valine-to-serine mutations at 9'- or 13'-positions. There is no function when α6 and ß2 subunits are expressed alone or in the presence of wild-type or mutant ß3 subunits. Interestingly, hybrid nAChR containing mouse α6 and human (h) ß4 subunits have function potentiated rather than suppressed by coexpression with wild-type hß3 subunits and potentiated further upon coexpression with hß3(V9'S) subunits. Studies using nAChR chimeric mouse/human α6 subunits indicated that residues involved in effects seen with hybrid nAChR are located in the α6 subunit N-terminal domain. More specifically, nAChR hα6 subunit residues Asn-143 and Met-145 are important for dominant-negative effects of nAChR hß3 subunits on hα6hß4-nAChR function. Asn-143 and additional residues in the N-terminal domain of nAChR hα6 subunits are involved in the gain-of-function effects of nAChR hß3(V9'S) subunits on α6ß2*-nAChR function. These studies illuminate the structural bases for effects of ß3 subunits on α6*-nAChR function and suggest that unique subunit interfaces involving the complementary rather than the primary face of α6 subunits are involved.


Subject(s)
Receptors, Nicotinic/chemistry , Receptors, Nicotinic/genetics , Amino Acid Sequence , Animals , Antidepressive Agents/therapeutic use , Dose-Response Relationship, Drug , Humans , Ion Channels/chemistry , Ligands , Mice , Molecular Sequence Data , Neurotransmitter Agents/metabolism , Point Mutation , Protein Structure, Tertiary , Sequence Homology, Amino Acid , Smoking Cessation
SELECTION OF CITATIONS
SEARCH DETAIL
...