Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 1.166
Filter
Add more filters

Publication year range
1.
Cell ; 145(1): 39-53, 2011 Apr 01.
Article in English | MEDLINE | ID: mdl-21376383

ABSTRACT

Treatment of tuberculosis, a complex granulomatous disease, requires long-term multidrug therapy to overcome tolerance, an epigenetic drug resistance that is widely attributed to nonreplicating bacterial subpopulations. Here, we deploy Mycobacterium marinum-infected zebrafish larvae for in vivo characterization of antitubercular drug activity and tolerance. We describe the existence of multidrug-tolerant organisms that arise within days of infection, are enriched in the replicating intracellular population, and are amplified and disseminated by the tuberculous granuloma. Bacterial efflux pumps that are required for intracellular growth mediate this macrophage-induced tolerance. This tolerant population also develops when Mycobacterium tuberculosis infects cultured macrophages, suggesting that it contributes to the burden of drug tolerance in human tuberculosis. Efflux pump inhibitors like verapamil reduce this tolerance. Thus, the addition of this currently approved drug or more specific efflux pump inhibitors to standard antitubercular therapy should shorten the duration of curative treatment.


Subject(s)
Drug Tolerance , Macrophages/microbiology , Mycobacterium marinum/physiology , Mycobacterium tuberculosis/physiology , Tuberculosis/microbiology , Animals , Antitubercular Agents/therapeutic use , Bacterial Proteins/antagonists & inhibitors , Bacterial Proteins/metabolism , Disease Models, Animal , Granuloma/physiopathology , Humans , Larva/microbiology , Membrane Transport Modulators/pharmacology , Membrane Transport Proteins/metabolism , Mycobacterium Infections, Nontuberculous/drug therapy , Mycobacterium Infections, Nontuberculous/pathology , Mycobacterium Infections, Nontuberculous/physiopathology , Mycobacterium marinum/drug effects , Tuberculosis/drug therapy , Tuberculosis/pathology , Tuberculosis/physiopathology , Verapamil/pharmacology , Zebrafish/microbiology
2.
Nat Rev Neurosci ; 20(9): 514-532, 2019 09.
Article in English | MEDLINE | ID: mdl-31289380

ABSTRACT

Light-controllable tools provide powerful means to manipulate and interrogate brain function with relatively low invasiveness and high spatiotemporal precision. Although optogenetic approaches permit neuronal excitation or inhibition at the network level, other technologies, such as optopharmacology (also known as photopharmacology) have emerged that provide molecular-level control by endowing light sensitivity to endogenous biomolecules. In this Review, we discuss the challenges and opportunities of photocontrolling native neuronal signalling pathways, focusing on ion channels and neurotransmitter receptors. We describe existing strategies for rendering receptors and channels light sensitive and provide an overview of the neuroscientific insights gained from such approaches. At the crossroads of chemistry, protein engineering and neuroscience, optopharmacology offers great potential for understanding the molecular basis of brain function and behaviour, with promises for future therapeutics.


Subject(s)
Ion Channels/metabolism , Neurons/metabolism , Optogenetics/trends , Photochemical Processes , Receptors, G-Protein-Coupled/metabolism , Animals , Humans , Ion Channel Gating/drug effects , Ion Channel Gating/physiology , Ion Channels/antagonists & inhibitors , Ion Channels/chemistry , Membrane Transport Modulators/pharmacology , Neurons/chemistry , Neurons/drug effects , Optogenetics/methods , Photochemical Processes/drug effects , Receptors, G-Protein-Coupled/chemistry
3.
Proc Natl Acad Sci U S A ; 118(28)2021 07 13.
Article in English | MEDLINE | ID: mdl-34260402

ABSTRACT

Sinoatrial node myocytes (SAMs) act as cardiac pacemaker cells by firing spontaneous action potentials (APs) that initiate each heartbeat. The funny current (If) is critical for the generation of these spontaneous APs; however, its precise role during the pacemaking cycle remains unresolved. Here, we used the AP-clamp technique to quantify If during the cardiac cycle in mouse SAMs. We found that If is persistently active throughout the sinoatrial AP, with surprisingly little voltage-dependent gating. As a consequence, it carries both inward and outward current around its reversal potential of -30 mV. Despite operating at only 2 to 5% of its maximal conductance, If carries a substantial fraction of both depolarizing and repolarizing net charge movement during the firing cycle. We also show that ß-adrenergic receptor stimulation increases the percentage of net depolarizing charge moved by If, consistent with a contribution of If to the fight-or-flight increase in heart rate. These properties were confirmed by heterologously expressed HCN4 channels and by mathematical models of If Modeling further suggested that the slow rates of activation and deactivation of the HCN4 isoform underlie the persistent activity of If during the sinoatrial AP. These results establish a new conceptual framework for the role of If in pacemaking, in which it operates at a very small fraction of maximal activation but nevertheless drives membrane potential oscillations in SAMs by providing substantial driving force in both inward and outward directions.


Subject(s)
Biological Clocks/physiology , Electrophysiological Phenomena , Myocytes, Cardiac/physiology , Sinoatrial Node/physiology , Action Potentials/drug effects , Action Potentials/physiology , Animals , Biological Clocks/drug effects , Computer Simulation , Diastole/drug effects , Diastole/physiology , Electrophysiological Phenomena/drug effects , HEK293 Cells , Humans , Hyperpolarization-Activated Cyclic Nucleotide-Gated Channels/metabolism , Ivabradine/pharmacology , Membrane Transport Modulators/pharmacology , Mice, Inbred C57BL , Myocytes, Cardiac/drug effects , Sinoatrial Node/drug effects
4.
Am J Physiol Cell Physiol ; 321(6): C932-C946, 2021 12 01.
Article in English | MEDLINE | ID: mdl-34644122

ABSTRACT

Chloride transport across cell membranes is broadly involved in epithelial fluid transport, cell volume and pH regulation, muscle contraction, membrane excitability, and organellar acidification. The human genome encodes at least 53 chloride-transporting proteins with expression in cell plasma or intracellular membranes, which include chloride channels, exchangers, and cotransporters, some having broad anion specificity. Loss-of-function mutations in chloride transporters cause a wide variety of human diseases, including cystic fibrosis, secretory diarrhea, kidney stones, salt-wasting nephropathy, myotonia, osteopetrosis, hearing loss, and goiter. Although impactful advances have been made in the past decade in drug treatment of cystic fibrosis using small molecule modulators of the defective cystic fibrosis transmembrane conductance regulator (CFTR) chloride channel, other chloride channels and solute carrier proteins (SLCs) represent relatively underexplored target classes for drug discovery. New opportunities have emerged for the development of chloride transport modulators as potential therapeutics for secretory diarrheas, constipation, dry eye disorders, kidney stones, polycystic kidney disease, hypertension, and osteoporosis. Approaches to chloride transport-targeted drug discovery are reviewed herein, with focus on chloride channel and exchanger classes in which recent preclinical advances have been made in the identification of small molecule modulators and in proof of concept testing in experimental animal models.


Subject(s)
Antiporters/drug effects , Chloride Channels/drug effects , Chlorides/metabolism , Drug Design , Drug Discovery , Membrane Transport Modulators/pharmacology , Animals , Antiporters/genetics , Antiporters/metabolism , Chloride Channels/genetics , Chloride Channels/metabolism , Cystic Fibrosis Transmembrane Conductance Regulator/drug effects , Cystic Fibrosis Transmembrane Conductance Regulator/genetics , Cystic Fibrosis Transmembrane Conductance Regulator/metabolism , Humans , Ion Transport , Kinetics , Membrane Transport Modulators/chemistry , Mutation , Sulfate Transporters/drug effects , Sulfate Transporters/genetics , Sulfate Transporters/metabolism
5.
J Cell Physiol ; 236(7): 5235-5252, 2021 07.
Article in English | MEDLINE | ID: mdl-33368220

ABSTRACT

Intratympanic injection of gentamicin has proven to be an effective therapy for intractable vestibular dysfunction. However, most studies to date have focused on the cochlea, so little is known about the distribution and uptake of gentamicin by the counterpart of the auditory system, specifically vestibular hair cells (HCs). Here, with a combination of in vivo and in vitro approaches, we used a gentamicin-Texas Red (GTTR) conjugate to investigate the mechanisms of gentamicin vestibulotoxicity in the developing mammalian utricular HCs. In vivo, GTTR fluorescence was concentrated in the apical cytoplasm and the cellular membrane of neonatal utricular HCs, but scarce in the nucleus of HCs and supporting cells. Quantitative analysis showed the GTTR uptake by striolar HCs was significantly higher than that in the extrastriola. In addition, the GTTR fluorescence intensity in the striola was increased gradually from 1 to 8 days, peaking at 8-9 days postnatally. In vitro, utricle explants were incubated with GTTR and candidate uptake conduits, including mechanotransduction (MET) channels and endocytosis in the HC, were inhibited separately. GTTR uptake by HCs could be inhibited by quinine, a blocker of MET channels, under both normal and stressed conditions. Meanwhile, endocytic inhibition only reduced GTTR uptake in the CoCl2 hypoxia model. In sum, the maturation of MET channels mediated uptake of GTTR into vestibular HCs. Under stressed conditions, MET channels play a pronounced role, manifested by channel-dependent stress enhanced GTTR permeation, while endocytosis participates in GTTR entry in a more selective manner.


Subject(s)
Biological Transport/physiology , Gentamicins/pharmacology , Gentamicins/pharmacokinetics , Hair Cells, Auditory/metabolism , Saccule and Utricle/embryology , Animals , Endocytosis/drug effects , Female , Gentamicins/chemistry , Male , Membrane Transport Modulators/pharmacology , Mice , Mice, Inbred C57BL , Organ Culture Techniques , Quinine/pharmacology , Reactive Oxygen Species/metabolism , Saccule and Utricle/metabolism , Staining and Labeling , Vestibular Diseases/drug therapy , Vestibular Diseases/pathology , Xanthenes/chemistry
6.
Physiol Rev ; 94(2): 419-59, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24692353

ABSTRACT

TMEM16 proteins, also known as anoctamins, are involved in a variety of functions that include ion transport, phospholipid scrambling, and regulation of other membrane proteins. The first two members of the family, TMEM16A (anoctamin-1, ANO1) and TMEM16B (anoctamin-2, ANO2), function as Ca2+-activated Cl- channels (CaCCs), a type of ion channel that plays important functions such as transepithelial ion transport, smooth muscle contraction, olfaction, phototransduction, nociception, and control of neuronal excitability. Genetic ablation of TMEM16A in mice causes impairment of epithelial Cl- secretion, tracheal abnormalities, and block of gastrointestinal peristalsis. TMEM16A is directly regulated by cytosolic Ca2+ as well as indirectly by its interaction with calmodulin. Other members of the anoctamin family, such as TMEM16C, TMEM16D, TMEM16F, TMEM16G, and TMEM16J, may work as phospholipid scramblases and/or ion channels. In particular, TMEM16F (ANO6) is a major contributor to the process of phosphatidylserine translocation from the inner to the outer leaflet of the plasma membrane. Intriguingly, TMEM16F is also associated with the appearance of anion/cation channels activated by very high Ca2+ concentrations. Furthermore, a TMEM16 protein expressed in Aspergillus fumigatus displays both ion channel and lipid scramblase activity. This finding suggests that dual function is an ancestral characteristic of TMEM16 proteins and that some members, such as TMEM16A and TMEM16B, have evolved to a pure channel function. Mutations in anoctamin genes (ANO3, ANO5, ANO6, and ANO10) cause various genetic diseases. These diseases suggest the involvement of anoctamins in a variety of cell functions whose link with ion transport and/or lipid scrambling needs to be clarified.


Subject(s)
Chloride Channels/metabolism , Signal Transduction , Animals , Chloride Channels/chemistry , Chloride Channels/drug effects , Chloride Channels/genetics , Disease Models, Animal , Genetic Predisposition to Disease , Humans , Membrane Transport Modulators/pharmacology , Mutation , Protein Conformation , Signal Transduction/drug effects , Structure-Activity Relationship
7.
Am J Physiol Gastrointest Liver Physiol ; 321(2): G185-G199, 2021 08 01.
Article in English | MEDLINE | ID: mdl-34132108

ABSTRACT

Recent studies in our lab have shown that the KV7 channel activator, flupirtine, inhibits colonic epithelial Cl- secretion through effects on submucosal neurons of the enteric nervous system (ENS). We hypothesized that flupirtine would also stimulate Na+ absorption as a result of reduced secretory ENS input to the epithelium. To test this hypothesis, unidirectional 22Na+ fluxes were measured under voltage-clamped conditions. Pharmacological approaches using an Ussing-style recording chamber combined with immunofluorescence microscopy techniques were used to determine the effect of flupirtine on active Na+ transport in the rat colon. Flupirtine stimulated electroneutral Na+ absorption in partially seromuscular-stripped colonic tissues, while simultaneously inhibiting short-circuit current (ISC; i.e., Cl- secretion). Both of these effects were attenuated by pretreatment with the ENS inhibitor, tetrodotoxin. The Na+/H+ exchanger isoform 3 (NHE-3)-selective inhibitor, S3226, significantly inhibited flupirtine-stimulated Na+ absorption, whereas the NHE-2-selective inhibitor HOE-694 did not. NHE-3 localization near the apical membranes of surface epithelial cells was also more apparent in flupirtine-treated colon versus control. Flupirtine did not alter epithelial Na+ channel (ENaC)-mediated Na+ absorption in distal colonic tissues obtained from hyperaldosteronaemic rats and had no effect in the normal ileum but did stimulate Na+ absorption in the proximal colon. Finally, the parallel effects of flupirtine on ISC (Cl- secretion) and Na+ absorption were significantly correlated with each other. Together, these data indicate that flupirtine stimulates NHE-3-dependent Na+ absorption, likely as a result of reduced stimulatory input to the colonic epithelium by submucosal ENS neurons.NEW & NOTEWORTHY We present a novel mechanism regarding regulation of epithelial ion transport by enteric neurons. Activation of neuronal KV7 K+ channels markedly stimulates Na+ absorption and inhibits Cl- secretion across the colonic epithelium. This may be useful in developing new treatments for diarrheal disorders, such as irritable bowel syndrome with diarrhea (IBS-D).


Subject(s)
Aminopyridines/pharmacology , Colon/metabolism , Enteric Nervous System/metabolism , Intestinal Absorption , Sodium/metabolism , Animals , Colon/drug effects , Epithelial Sodium Channels/metabolism , Guanidines/pharmacology , Male , Membrane Transport Modulators/pharmacology , Methacrylates/pharmacology , Rats , Rats, Sprague-Dawley , Sodium-Hydrogen Exchangers/antagonists & inhibitors , Sodium-Hydrogen Exchangers/metabolism , Sulfones/pharmacology
8.
Am J Physiol Heart Circ Physiol ; 320(4): H1738-H1748, 2021 04 01.
Article in English | MEDLINE | ID: mdl-33635166

ABSTRACT

Emerging evidence suggests the exercise pressor reflex is exaggerated in early stage type 1 diabetes mellitus (T1DM). Piezo channels may play a role in this exaggeration, as blocking these channels attenuates the exaggerated pressor response to tendon stretch in T1DM rats. However, tendon stretch constitutes a different mechanical and physiological stimuli than that occurring during muscle contraction. Therefore, the purpose of this study was to determine the contribution of Piezo channels in evoking the pressor reflex during an intermittent muscle contraction in T1DM. In unanesthetized decerebrate rats, we compared the pressor and cardioaccelerator responses to intermittent muscle contraction before and after locally injecting grammostola spatulata mechanotoxin 4 (GsMTx-4, 0.25 µM) into the hindlimb vasculature. Although GsMTx-4 has a high potency for Piezo channels, it has also been suggested to block transient receptor potential cation (TRPC) channels. We, therefore, performed additional experiments to control for this possibility by also injecting SKF 96365 (10 µM), a TRPC channel blocker. We found that local injection of GsMTx-4, but not SKF 96365, attenuated the exaggerated peak pressor (ΔMAP before: 33 ± 3 mmHg, after: 22 ± 3 mmHg, P = 0.007) and pressor index (ΔBPi before: 668 ± 91 mmHg·s, after: 418 ± 81 mmHg·s, P = 0.021) response in streptozotocin (STZ) rats (n = 8). GsMTx-4 attenuated the exaggerated early onset pressor and the pressor response over time, which eliminated peak differences as well as those over time between T1DM and healthy controls. These data suggest that Piezo channels are an effective target to normalize the exercise pressor reflex in T1DM.NEW & NOTEWORTHY This is the first study to demonstrate that blocking Piezo channels is effective in ameliorating the exaggerated exercise pressor reflex evoked by intermittent muscle contraction, commonly occurring during physical activity, in T1DM. Thus, these findings suggest Piezo channels may serve as an effective therapeutic target to reduce the acute and prolonged cardiovascular strain that may occur during dynamic exercise in T1DM.


Subject(s)
Autonomic Nervous System/drug effects , Blood Pressure/drug effects , Cardiovascular System/innervation , Diabetes Mellitus, Experimental/drug therapy , Diabetes Mellitus, Type 1/drug therapy , Intercellular Signaling Peptides and Proteins/pharmacology , Membrane Transport Modulators/pharmacology , Muscle Contraction , Muscle, Skeletal/innervation , Reflex, Abnormal/drug effects , Spider Venoms/pharmacology , Animals , Autonomic Nervous System/metabolism , Autonomic Nervous System/physiopathology , Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Experimental/physiopathology , Diabetes Mellitus, Type 1/metabolism , Diabetes Mellitus, Type 1/physiopathology , Female , Heart Rate/drug effects , Ion Channels/antagonists & inhibitors , Ion Channels/metabolism , Male , Physical Conditioning, Animal , Rats, Sprague-Dawley , Time Factors
9.
Cell Physiol Biochem ; 55(S3): 14-45, 2021 Mar 03.
Article in English | MEDLINE | ID: mdl-33656309

ABSTRACT

Although ion channels are crucial in many physiological processes and constitute an important class of drug targets, much is still unclear about their function and possible malfunctions that lead to diseases. In recent years, computational methods have evolved into important and invaluable approaches for studying ion channels and their functions. This is mainly due to their demanding mechanism of action where a static picture of an ion channel structure is often insufficient to fully understand the underlying mechanism. Therefore, the use of computational methods is as important as chemical-biological based experimental methods for a better understanding of ion channels. This review provides an overview on a variety of computational methods and software specific to the field of ion-channels. Artificial intelligence (or more precisely machine learning) approaches are applied for the sequence-based prediction of ion channel family, or topology of the transmembrane region. In case sufficient data on ion channel modulators is available, these methods can also be applied for quantitative structureactivity relationship (QSAR) analysis. Molecular dynamics (MD) simulations combined with computational molecular design methods such as docking can be used for analysing the function of ion channels including ion conductance, different conformational states, binding sites and ligand interactions, and the influence of mutations on their function. In the absence of a three-dimensional protein structure, homology modelling can be applied to create a model of your ion channel structure of interest. Besides highlighting a wide range of successful applications, we will also provide a basic introduction to the most important computational methods and discuss best practices to get a rough idea of possible applications and risks.


Subject(s)
Artificial Intelligence , Ion Channels/chemistry , Membrane Transport Modulators/chemistry , Molecular Dynamics Simulation , Software , Animals , Binding Sites , Humans , Ion Channel Gating/drug effects , Ion Channels/agonists , Ion Channels/antagonists & inhibitors , Ligands , Membrane Transport Modulators/pharmacology , Models, Molecular , Protein Binding , Protein Conformation, alpha-Helical , Protein Conformation, beta-Strand , Protein Interaction Domains and Motifs , Quantitative Structure-Activity Relationship , Structural Homology, Protein
10.
Cell Physiol Biochem ; 55(S3): 87-107, 2021 Mar 06.
Article in English | MEDLINE | ID: mdl-33667333

ABSTRACT

Potassium channels of the tandem of two-pore-domain (K2P) family were among the last potassium channels cloned. However, recent progress in understanding their physiological relevance and molecular pharmacology revealed their therapeutic potential and thus these channels evolved as major drug targets against a large variety of diseases. However, after the initial cloning of the fifteen family members there was a lack of potent and/or selective modulators. By now a large variety of K2P channel modulators (activators and blockers) have been described, especially for TASK-1, TASK-3, TREK-1, TREK2, TRAAK and TRESK channels. Recently obtained crystal structures of K2P channels, alanine scanning approaches to map drug binding sites, in silico experiments with molecular dynamics simulations (MDs) combined with electrophysiological studies to reveal the mechanism of channel inhibition/activation, yielded a good understanding of the molecular pharmacology of these channels. Besides summarizing drugs that were identified to modulate K2P channels, the main focus of this article is on describing the differential binding sites and mechanisms of channel modulation that are utilized by the different K2P channel blockers and activators.


Subject(s)
Cardiac Conduction System Disease/drug therapy , Membrane Transport Modulators/pharmacology , Migraine Disorders/drug therapy , Potassium Channels, Tandem Pore Domain/metabolism , Potassium/metabolism , Action Potentials/drug effects , Action Potentials/physiology , Binding Sites , Cardiac Conduction System Disease/genetics , Cardiac Conduction System Disease/metabolism , Cardiac Conduction System Disease/pathology , Gene Expression , Humans , Ion Channel Gating/drug effects , Ion Transport , Ligands , Membrane Transport Modulators/chemistry , Membrane Transport Modulators/classification , Migraine Disorders/genetics , Migraine Disorders/metabolism , Migraine Disorders/pathology , Molecular Dynamics Simulation , Organ Specificity , Potassium Channels, Tandem Pore Domain/classification , Potassium Channels, Tandem Pore Domain/genetics , Protein Binding , Protein Isoforms/classification , Protein Isoforms/genetics , Protein Isoforms/metabolism , Protein Structure, Secondary
11.
Cell Physiol Biochem ; 55(S3): 108-130, 2021 May 28.
Article in English | MEDLINE | ID: mdl-34043299

ABSTRACT

Transient receptor potential vanilloid (TRPV) channels are part of the TRP channel superfamily and named after the first identified member TRPV1, that is sensitive to the vanillylamide capsaicin. Their overall structure is similar to the structure of voltage gated potassium channels (Kv) built up as homotetramers from subunits with six transmembrane helices (S1-S6). Six TRPV channel subtypes (TRPV1-6) are known, that can be subdivided into the thermoTRPV (TRPV1-4) and the Ca2+-selective TRPV channels (TRPV5, TRPV6). Contrary to Kv channels, TRPV channels are not primary voltage gated. All six channels have distinct properties and react to several endogenous ligands as well as different gating stimuli such as heat, pH, mechanical stress, or osmotic changes. Their physiological functions are highly diverse and subtype as well as tissue specific. In many tissues they serve as sensors for different pain stimuli (heat, pressure, pH) and contribute to the homeostasis of electrolytes, the maintenance of barrier functions and the development of macrophages. Due to their fundamental role in manifold physiological and pathophysiological processes, TRPV channels are promising targets for drug development. However, drugs targeting specific TRPV channels, that are suitable for drug therapy, are rare. Moreover, selective and potent compounds for further research at TRPV channels are often lacking. In this review different aspects of the structure, the different gating stimuli, the expression pattern, the physiological and pathophysiological roles as well as the modulating mechanisms of synthetic, natural and endogenous ligands are summarized.


Subject(s)
Analgesics/pharmacology , Antineoplastic Agents/pharmacology , Immunologic Factors/pharmacology , Membrane Transport Modulators/pharmacology , TRPV Cation Channels/metabolism , Analgesics/chemistry , Analgesics/classification , Antineoplastic Agents/chemistry , Antineoplastic Agents/classification , Binding Sites , Brain/cytology , Brain/drug effects , Brain/metabolism , Humans , Immunologic Factors/chemistry , Immunologic Factors/classification , Ion Channel Gating/drug effects , Ligands , Lung/cytology , Lung/drug effects , Lung/metabolism , Membrane Transport Modulators/chemistry , Membrane Transport Modulators/classification , Models, Molecular , Organ Specificity , Protein Binding , Protein Isoforms/agonists , Protein Isoforms/antagonists & inhibitors , Protein Isoforms/classification , Protein Isoforms/metabolism , Protein Structure, Secondary , Spleen/cytology , Spleen/drug effects , Spleen/metabolism , TRPV Cation Channels/agonists , TRPV Cation Channels/antagonists & inhibitors , TRPV Cation Channels/classification
12.
Cell Physiol Biochem ; 55(S3): 145-156, 2021 May 28.
Article in English | MEDLINE | ID: mdl-34043301

ABSTRACT

The population of regulatory T cells (Tregs) is critical for immunological self-tolerance and homeostasis. Proper ion regulation contributes to Treg lineage identity, regulation, and effector function. Identified ion channels include Ca2+ release-activated Ca2+, transient receptor potential, P2X, volume-regulated anion and K+ channels Kv1.3 and KCa3.1. Ion channel modulation represents a promising therapeutic approach for the treatment of autoimmune diseases such as rheumatoid arthritis and multiple sclerosis. This review summarizes studies with gene-targeted mice and pharmacological modulators affecting Treg number and function. Furthermore, participation of ion channels is illustrated and the power of future research possibilities is discussed.


Subject(s)
Arthritis, Rheumatoid/drug therapy , Calcium/metabolism , Gene Expression Regulation/drug effects , Membrane Transport Modulators/pharmacology , Multiple Sclerosis/drug therapy , T-Lymphocytes, Regulatory/drug effects , Animals , Arthritis, Rheumatoid/genetics , Arthritis, Rheumatoid/immunology , Arthritis, Rheumatoid/metabolism , Calcium/immunology , Calcium Release Activated Calcium Channels/genetics , Calcium Release Activated Calcium Channels/immunology , Calcium Signaling , Disease Models, Animal , Gene Expression Regulation/immunology , Humans , Intermediate-Conductance Calcium-Activated Potassium Channels/genetics , Intermediate-Conductance Calcium-Activated Potassium Channels/immunology , Kv1.3 Potassium Channel/genetics , Kv1.3 Potassium Channel/immunology , Membrane Proteins/genetics , Membrane Proteins/immunology , Membrane Transport Modulators/chemistry , Mice , Multiple Sclerosis/genetics , Multiple Sclerosis/immunology , Multiple Sclerosis/metabolism , Receptors, Purinergic P2X/genetics , Receptors, Purinergic P2X/immunology , Receptors, Purinergic P2X7/genetics , Receptors, Purinergic P2X7/immunology , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/pathology , Transient Receptor Potential Channels/genetics , Transient Receptor Potential Channels/immunology
13.
J Pharmacol Exp Ther ; 377(2): 293-304, 2021 05.
Article in English | MEDLINE | ID: mdl-33602875

ABSTRACT

In failing hearts, Na+/Ca2+ exchanger (NCX) overactivity contributes to Ca2+ depletion, leading to contractile dysfunction. Inhibition of NCX is expected to normalize Ca2+ mishandling, to limit afterdepolarization-related arrhythmias, and to improve cardiac function in heart failure (HF). SAR340835/SAR296968 is a selective NCX inhibitor for all NCX isoforms across species, including human, with no effect on the native voltage-dependent calcium and sodium currents in vitro. Additionally, it showed in vitro and in vivo antiarrhythmic properties in several models of early and delayed afterdepolarization-related arrhythmias. Its effect on cardiac function was studied under intravenous infusion at 250,750 or 1500 µg/kg per hour in dogs, which were either normal or submitted to chronic ventricular pacing at 240 bpm (HF dogs). HF dogs were infused with the reference inotrope dobutamine (10 µg/kg per minute, i.v.). In normal dogs, NCX inhibitor increased cardiac contractility (dP/dtmax) and stroke volume (SV) and tended to reduce heart rate (HR). In HF dogs, NCX inhibitor significantly and dose-dependently increased SV from the first dose (+28.5%, +48.8%, and +62% at 250, 750, and 1500 µg/kg per hour, respectively) while significantly increasing dP/dtmax only at 1500 (+33%). Furthermore, NCX inhibitor significantly restored sympathovagal balance and spontaneous baroreflex sensitivity (BRS) from the first dose and reduced HR at the highest dose. In HF dogs, dobutamine significantly increased dP/dtmax and SV (+68.8%) but did not change HR, sympathovagal balance, or BRS. Overall, SAR340835, a selective potent NCX inhibitor, displayed a unique therapeutic profile, combining antiarrhythmic properties, capacity to restore systolic function, sympathovagal balance, and BRS in HF dogs. NCX inhibitors may offer new therapeutic options for acute HF treatment. SIGNIFICANCE STATEMENT: HF is facing growing health and economic burden. Moreover, patients hospitalized for acute heart failure are at high risk of decompensation recurrence, and no current acute decompensated HF therapy definitively improved outcomes. A new potent, Na+/Ca2+ exchanger inhibitor SAR340835 with antiarrhythmic properties improved systolic function of failing hearts without creating hypotension, while reducing heart rate and restoring sympathovagal balance. SAR340835 may offer a unique and attractive pharmacological profile for patients with acute heart failure as compared with current inotrope, such as dobutamine.


Subject(s)
Heart Failure/drug therapy , Membrane Transport Modulators/therapeutic use , Sodium-Calcium Exchanger/antagonists & inhibitors , Vagus Nerve/drug effects , Animals , Baroreflex , Dogs , Heart/drug effects , Heart Rate , Membrane Transport Modulators/administration & dosage , Membrane Transport Modulators/pharmacology , Myocardial Contraction , Myocardium/metabolism , Swine
14.
J Pharmacol Exp Ther ; 377(1): 20-28, 2021 04.
Article in English | MEDLINE | ID: mdl-33431609

ABSTRACT

Treatment of chronic pain remains an unmet medical need. The neuronal voltage-gated potassium Kv7/KCNQ/M channel has been implicated as a therapeutic target for chronic pain. However, whether pharmacological activation of the Kv7 channel can alleviate pain remains elusive. In this study, we show that selective activation of native M-currents by a novel channel opener SCR2682 reduces repetitive firings of dorsal root ganglia (DRG) sensory neurons. Intraperitoneal administration of SCR2682 relieves mechanical allodynia and thermal hyperalgesia in rat models of pain induced by complete Freund's adjuvant (CFA) or spared nerve injury (SNI) in a dose-dependent manner without affecting locomotor activity. The antinociceptive efficacy of SCR2682 can be reversed by the channel-specific blocker XE991. Furthermore, SCR2682 increases Kv7.2/KCNQ2 mRNA and protein expression in DRG neurons from rats in the SNI model of neuropathic pain. Taken together, pharmacological activation of neuronal Kv7 channels by opener SCR2682 can alleviate pain in rats, thus possessing therapeutic potential for chronic pain or hyperexcitability-related neurologic disorders. SIGNIFICANCE STATEMENT: A novel voltage-gated potassium Kv7 channel opener SCR2682 inhibits action potential firings in dorsal root ganglia sensory neurons and exhibits efficacy in antinociception, thus possessing a developmental potential for treatment of chronic pain or epilepsy.


Subject(s)
Analgesics/therapeutic use , Chronic Pain/drug therapy , KCNQ2 Potassium Channel/metabolism , Membrane Transport Modulators/therapeutic use , Pyridines/therapeutic use , Action Potentials , Analgesics/pharmacology , Animals , Cells, Cultured , Ganglia, Spinal/drug effects , Ganglia, Spinal/metabolism , Ganglia, Spinal/physiology , KCNQ2 Potassium Channel/agonists , Male , Membrane Transport Modulators/pharmacology , Pyridines/pharmacology , Rats , Rats, Sprague-Dawley
15.
J Pharmacol Exp Ther ; 377(1): 75-85, 2021 04.
Article in English | MEDLINE | ID: mdl-33504590

ABSTRACT

Inflammatory bowel diseases (IBD) are chronic inflammatory diseases of the gastrointestinal tract arising from abnormal responses of the innate and adaptative immune systems. Interleukin (IL)-10-producing CD4+CD25+ regulatory T (Treg) cells play a protective role in the recovery phase of IBD. In the present study, the effects of the administration of the selective Ca2+-activated K+ channel KCa3.1 inhibitor TRAM-34 on disease activities were examined in chemically induced IBD model mice. IBD disease severity, as assessed by diarrhea, visible fecal blood, inflammation, and crypt damage in the colon, was significantly lower in mice administered 1 mg/kg TRAM-34 than in vehicle-administered mice. Quantitative real-time polymerase chain reaction examinations showed that IL-10 expression levels in the recovery phase were markedly increased by the inhibition of KCa3.1 in mesenteric lymph node (mLN) Treg cells of IBD model mice compared with vehicle-administered mice. Among several positive and negative transcriptional regulators (TRs) for IL-10, three positive TRs-E4BP4, KLF4, and Blimp1-were upregulated by the inhibition of KCa3.1 in the mLN Treg cells of IBD model mice. In mouse peripheral CD4+CD25+ Treg cells induced by lectin stimulation, IL-10 expression and secretion were enhanced by the treatment with TRAM-34, together with the upregulation of E4BP4, KLF4, and Blimp1. Collectively, the present results demonstrated that the pharmacological inhibition of KCa3.1 decreased IBD symptoms in the IBD model by increasing IL-10 production in peripheral Treg cells and that IL-10high Treg cells produced by the treatment with KCa3.1 inhibitor may contribute to efficient Treg therapy for chronic inflammatory disorders, including IBD. SIGNIFICANCE STATEMENT: Pharmacological inhibition of Ca2+-activated K+ channel KCa3.1 increased IL-10 expression in peripheral Treg cells, together with the upregulation of the transcriptional regulators of IL-10: Krüppel-like factor 4, E4 promoter-binding protein 4, and/or B lymphocyte-induced maturation protein 1. The manipulation of IL-10high-producing Treg cells by the pharmacological inhibition of KCa3.1 may be beneficial in the treatment of chronic inflammatory diseases such as inflammatory bowel disease.


Subject(s)
Inflammatory Bowel Diseases/metabolism , Interleukin-10/metabolism , Intermediate-Conductance Calcium-Activated Potassium Channels/antagonists & inhibitors , Membrane Transport Modulators/pharmacology , Pyrazoles/pharmacology , T-Lymphocytes, Regulatory/metabolism , Animals , Basic-Leucine Zipper Transcription Factors/genetics , Basic-Leucine Zipper Transcription Factors/metabolism , Female , Inflammatory Bowel Diseases/drug therapy , Inflammatory Bowel Diseases/pathology , Interleukin-10/genetics , Interleukin-2 Receptor alpha Subunit/genetics , Interleukin-2 Receptor alpha Subunit/metabolism , Intermediate-Conductance Calcium-Activated Potassium Channels/metabolism , Kruppel-Like Factor 4 , Kruppel-Like Transcription Factors/genetics , Kruppel-Like Transcription Factors/metabolism , Membrane Transport Modulators/administration & dosage , Membrane Transport Modulators/therapeutic use , Mice , Mice, Inbred C57BL , Positive Regulatory Domain I-Binding Factor 1/genetics , Positive Regulatory Domain I-Binding Factor 1/metabolism , Pyrazoles/administration & dosage , Pyrazoles/therapeutic use , T-Lymphocytes, Regulatory/drug effects
16.
Am J Physiol Regul Integr Comp Physiol ; 320(4): R563-R573, 2021 04 01.
Article in English | MEDLINE | ID: mdl-33085914

ABSTRACT

Transient receptor potential vanilloid 4 (TRPV4) channels exist on vascular endothelial cells and eccrine sweat gland secretory cells in human skin. Here, we assessed whether TRPV4 channels contribute to cutaneous vasodilation and sweating during whole body passive heat stress (protocol 1) and to cutaneous vasodilation during postocclusive reactive hyperemia and local thermal hyperemia (protocol 2). Intradermal microdialysis was employed to locally deliver pharmacological agents to forearm skin sites, where cutaneous vascular conductance (CVC) and sweat rate were assessed. In protocol 1 (12 young adults), CVC and sweat rate were increased by passive whole body heating, resulting in a body core temperature elevation of 1.2 ± 0.1°C. The elevated CVC and sweat rate assessed at sites treated with TRPV4 channel antagonist (either 200 µM HC-067047 or 125 µM GSK2193874) were not different from the vehicle control site (5% dimethyl sulfoxide). After whole body heating, the TRPV4 channel agonist (100 µM GSK1016790A) was administered to each skin site, eliciting elevations in CVC. Relative to control, this response was partly attenuated by both TRPV4 channel antagonists, confirming drug efficacy. In protocol 2 (10 young adults), CVC was increased following a 5-min arterial occlusion and during local heating from 33 to 42°C. These responses did not differ between the control and the TRPV4 channel antagonist sites (200 µM HC-067047). We show that TRPV4 channels are not required for regulating cutaneous vasodilation or sweating during a whole body passive heat stress. Furthermore, they are not required for regulating cutaneous vasodilation during postocclusive reactive hyperemia and local thermal hyperemia.


Subject(s)
Hyperemia/physiopathology , Hyperthermia/physiopathology , Membrane Transport Modulators/administration & dosage , Skin/blood supply , Sweating , TRPV Cation Channels/antagonists & inhibitors , Vasodilation , Adult , Female , Humans , Hyperemia/metabolism , Hyperthermia/metabolism , Leucine/administration & dosage , Leucine/analogs & derivatives , Male , Microdialysis , Morpholines/administration & dosage , Piperidines/administration & dosage , Pyrroles/administration & dosage , Quinolines/administration & dosage , Regional Blood Flow , Skin/metabolism , Sulfonamides/administration & dosage , TRPV Cation Channels/metabolism , Time Factors , Young Adult
17.
Toxicol Appl Pharmacol ; 429: 115704, 2021 10 15.
Article in English | MEDLINE | ID: mdl-34474082

ABSTRACT

Organic anion-transporting polypeptide 2B1 (OATP2B1) is a multispecific transporter mediating the cellular uptake of steroids and numerous drugs. OATP2B1 is abundantly expressed in the intestine and is also present in various tumors. Increased steroid hormone uptake by OATP2B1 has been suggested to promote progression of hormone dependent tumors. 13α-estrones are effective inhibitors of endogenous estrogen formation and are potential candidates to inhibit proliferation of hormone dependent cancers. Recently, we have identified a variety of 13α/ß-estrone-based inhibitors of OATP2B1. However, the nature of this interaction, whether these inhibitors are potential transported substrates of OATP2B1 and hence may be enriched in OATP2B1-overexpressing cells, has not yet been investigated. In the current study we explored the antiproliferative effect of the most effective OATP2B1 inhibitor 13α/ß-estrones in control and OATP2B1-overexpressing A431 carcinoma cells. We found an increased antiproliferative effect of 3-O-benzyl 13α/ß-estrones in both mock transfected and OATP2B1-overexpressing cells. However, C-2 halogenated 13α-estrones had a selective OATP2B1-mediated cell growth inhibitory effect. In order to demonstrate that increased sensitization can be attributed to OATP2B1-mediated cellular uptake, tritium labeled 2-bromo-13α-estrone was synthesized for direct transport measurements. These experiments revealed increased accumulation of [3H]2-bromo-13α-estrone due to OATP2B1 function. Our results indicate that C-2 halogenated 13α-estrones are good candidates in the design of anti-cancer drugs targeting OATP2B1.


Subject(s)
Antineoplastic Agents/pharmacology , Carcinoma, Squamous Cell/drug therapy , Cell Proliferation/drug effects , Estrone/pharmacology , Membrane Transport Modulators/pharmacology , Organic Anion Transporters/antagonists & inhibitors , Skin Neoplasms/drug therapy , Antineoplastic Agents/metabolism , Carcinoma, Squamous Cell/genetics , Carcinoma, Squamous Cell/metabolism , Carcinoma, Squamous Cell/pathology , Cell Line, Tumor , Estrone/analogs & derivatives , Estrone/metabolism , Humans , Membrane Transport Modulators/metabolism , Organic Anion Transporters/genetics , Organic Anion Transporters/metabolism , Skin Neoplasms/genetics , Skin Neoplasms/metabolism , Skin Neoplasms/pathology
18.
J Cardiovasc Pharmacol ; 77(4): 430-449, 2021 04 01.
Article in English | MEDLINE | ID: mdl-33416260

ABSTRACT

ABSTRACT: Establishing efficient perfusion into the myocardium is the main purpose in patients with acute coronary syndrome, but the process of reperfusion is not without risk and can damage the myocardium paradoxically. Unfortunately, there is no effective treatment for reperfusion injury, and efforts to find an efficient preventive approach are still ongoing. In the past 3 decades, there have been many successful animal studies on how to prevent reperfusion injury; nonetheless, translation to the clinical setting has almost always proven disappointing. In this article, we review clinical studies on the prevention of reperfusion injury in patients with acute coronary syndrome undergoing primary percutaneous coronary intervention in a pharmacologic-based approach. We categorize all the agents that are evaluated for the prevention of myocardial reperfusion injury based on their mechanisms of action into 5 groups: drugs that can reduce oxidative stress, drugs that can affect cellular metabolism, rheological agents that target microvascular obstruction, anti-inflammatory agents, and agents with mixed mechanisms of action. Then, review all the clinical studies of these agents in the setting of primary percutaneous coronary intervention. Finally, we will discuss the possible reasons for the failure in translation of studies into practice and propose potential solutions to overcome this problem.


Subject(s)
Acute Coronary Syndrome/therapy , Cardiovascular Agents/therapeutic use , Myocardial Reperfusion Injury/prevention & control , Percutaneous Coronary Intervention , Acute Coronary Syndrome/physiopathology , Anti-Inflammatory Agents/therapeutic use , Antioxidants/therapeutic use , Cardiovascular Agents/adverse effects , Humans , Membrane Transport Modulators/therapeutic use , Myocardial Reperfusion Injury/etiology , Myocardial Reperfusion Injury/physiopathology , Percutaneous Coronary Intervention/adverse effects , Risk Assessment , Risk Factors , Time Factors , Treatment Outcome
19.
Am J Respir Crit Care Med ; 201(8): 946-954, 2020 04 15.
Article in English | MEDLINE | ID: mdl-31898911

ABSTRACT

Rationale: Enhancing non-CFTR (cystic fibrosis transmembrane conductance regulator)-mediated anion secretion is an attractive therapeutic approach for the treatment of cystic fibrosis (CF) and other mucoobstructive diseases.Objectives: To determine the effects of TMEM16A potentiation on epithelial fluid secretion and mucociliary clearance.Methods: The effects of a novel low-molecular-weight TMEM16A potentiator (ETX001) were evaluated in human cell and animal models of airway epithelial function and mucus transport.Measurements and Main Results: Potentiating the activity of TMEM16A with ETX001 increased the Ca2+-activated Cl- channel activity and anion secretion in human bronchial epithelial (HBE) cells from patients with CF without impacting calcium signaling. ETX001 rapidly increased fluid secretion and airway surface liquid height in CF-HBE cells under both static conditions and conditions designed to mimic the shear stress associated with tidal breathing. In ovine models of mucus clearance (tracheal mucus velocity and mucociliary clearance), inhaled ETX001 was able to accelerate clearance both when CFTR function was reduced by administration of a pharmacological blocker and when CFTR was fully functional.Conclusions: Enhancing the activity of TMEM16A increases epithelial fluid secretion and enhances mucus clearance independent of CFTR function. TMEM16A potentiation is a novel approach for the treatment of patients with CF and non-CF mucoobstructive diseases.


Subject(s)
Anoctamin-1/drug effects , Cystic Fibrosis/metabolism , Epithelial Cells/drug effects , Membrane Transport Modulators/pharmacology , Mucociliary Clearance/drug effects , Mucus/drug effects , Administration, Inhalation , Animals , Anoctamin-1/metabolism , Bronchi/cytology , Calcium Signaling/drug effects , Cystic Fibrosis Transmembrane Conductance Regulator/antagonists & inhibitors , Cystic Fibrosis Transmembrane Conductance Regulator/genetics , Cystic Fibrosis Transmembrane Conductance Regulator/metabolism , Epithelial Cells/metabolism , Humans , Ion Transport/drug effects , Patch-Clamp Techniques , Respiration , Respiratory Mucosa/cytology , Sheep , Trachea/drug effects , Trachea/metabolism
20.
ScientificWorldJournal ; 2021: 5381993, 2021.
Article in English | MEDLINE | ID: mdl-34720766

ABSTRACT

Microbial infections remain a public health problem due to the upsurge of bacterial resistance. In this study, the antibacterial, antibiofilm, and efflux pump inhibitory activities of the stem bark of Acacia macrostachya, an indigenous African medicinal plant, were investigated. In traditional medicine, the plant is used in the treatment of microbial infections and inflammatory conditions. A crude methanol extract obtained by Soxhlet extraction was partitioned by column chromatography to obtain the petroleum ether, ethyl acetate, and methanol fractions. Antibacterial, efflux pump inhibition and antibiofilm formation activities were assessed by the high-throughput spot culture growth inhibition (HT-SPOTi), ethidium bromide accumulation, and the crystal violet retention assay, respectively. The minimum inhibitory concentrations (MICs) of the crude extract and major fractions ranged from 250 to ≥500 µg/mL. At a concentration of 3.9-250 µg/mL, all extracts demonstrated >80% inhibition of biofilm formation in S. aureus. In P. aeruginosa, the EtOAc fraction showed the highest antibiofilm activity (59-69%) while the pet-ether fraction was most active against E. coli biofilms (45-67%). Among the test samples, the crude extract, methanol, and ethyl acetate fractions showed remarkable efflux pump inhibition in S. aureus, E. coli, and P. aeruginosa. At ½ MIC, the methanol fraction demonstrated significant accumulation of EtBr in E. coli having superior efflux inhibition over the standard EPIs: chlorpromazine and verapamil. Tannins, flavonoids, triterpenoids, phytosterols, coumarins, and saponins were identified in preliminary phytochemical studies. Stigmasterol was identified in the EtOAc fraction. This study justifies the use of A. macrostachya in the treatment of infections in traditional medicine and highlights its potential as a source of bioactive compounds that could possibly interact with some resistance mechanisms in bacteria to combat antimicrobial resistance.


Subject(s)
Acacia , Anti-Bacterial Agents/pharmacology , Biofilms/drug effects , Plant Bark , Plant Extracts/pharmacology , Plant Stems , Anti-Bacterial Agents/isolation & purification , Biofilms/growth & development , Escherichia coli/drug effects , Escherichia coli/physiology , Humans , Membrane Transport Modulators/isolation & purification , Membrane Transport Modulators/pharmacology , Microbial Sensitivity Tests/methods , Plant Extracts/isolation & purification , Staphylococcus aureus/drug effects , Staphylococcus aureus/physiology
SELECTION OF CITATIONS
SEARCH DETAIL