Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
Add more filters










Publication year range
1.
Neurol Neuroimmunol Neuroinflamm ; 11(3): e200217, 2024 May.
Article in English | MEDLINE | ID: mdl-38547427

ABSTRACT

BACKGROUND AND OBJECTIVES: Epstein-Barr virus (EBV) has been strongly implicated in the pathogenesis of multiple sclerosis (MS). Despite this, there are no routinely used tests to measure cellular response to EBV. In this study, we analyzed the cellular response to EBV nuclear antigen-1 (EBNA-1) in people with MS (pwMS) using a whole blood assay. METHODS: This cross-sectional study took place in a dedicated MS clinic in a university hospital. We recruited healthy controls, people with epilepsy (PWE), and pwMS taking a range of disease-modifying treatments (DMTs) including natalizumab, anti-CD20 monoclonal antibodies (mAbs), dimethyl fumarate (DMF), and also treatment naïve. Whole blood samples were stimulated with commercially available PepTivator EBNA1 peptides and a control virus-cytomegalovirus (CMV) peptide. We recorded the cellular response to stimulation with both interferon gamma (IFN-γ) and interleukin-2 (IL-2). We also compared the cellular responses to EBNA1 with IgG responses to EBNA1, viral capsid antigen (VCA), and EBV viral load. RESULTS: We recruited 86 pwMS, with relapsing remitting MS, in this group, and we observed a higher level of cellular response recorded with IFN-γ (0.79 IU/mL ± 1.36) vs healthy controls (0.29 IU/mL ± 0.90, p = 0.0048) and PWE (0.17 IU/mL ± 0.33, p = 0.0088). Treatment with either anti-CD20 mAbs (0.28 IU/mL ± 0.57) or DMF (0.07 IU/mL ± 0.15) resulted in a cellular response equivalent to control levels or in PWE (p = 0.26). The results of recording IL-2 response were concordant with IFN-γ: with suppression also seen with anti-CD20 mAbs and DMF. By contrast, we did not record any differential effect of DMTs on the levels of IgG to either EBNA-1 or VCA. Nor did we observe differences in cellular response to cytomegalovirus between groups. DISCUSSION: This study demonstrates how testing and recording the cellular response to EBNA-1 in pwMS may be beneficial. EBNA-1 stimulation of whole blood samples produced higher levels of IFN-γ and IL-2 in pwMS compared with controls and PWE. In addition, we show a differential effect of currently available DMTs on this response. The functional assay deployed uses whole blood samples with minimal preprocessing suggesting that employment as a treatment response measure in clinical trials targeting EBV may be possible.


Subject(s)
Epstein-Barr Virus Nuclear Antigens , Herpesvirus 4, Human , Multiple Sclerosis , Humans , Antibodies, Viral , Antigens, Viral , Capsid Proteins , Cross-Sectional Studies , Epstein-Barr Virus Infections/complications , Epstein-Barr Virus Infections/immunology , Epstein-Barr Virus Nuclear Antigens/immunology , Herpesvirus 4, Human/immunology , Immunity, Cellular , Immunoglobulin G , Interferon-gamma , Interleukin-2 , Multiple Sclerosis/drug therapy , Multiple Sclerosis/virology
3.
Front Physiol ; 11: 230, 2020.
Article in English | MEDLINE | ID: mdl-32256387

ABSTRACT

Interstitial cells of Cajal (ICC) are pacemaker cells that generate electrical slow waves in gastrointestinal (GI) smooth muscles. Slow waves organize basic motor patterns, such as peristalsis and segmentation in the GI tract. Slow waves depend upon activation of Ca2+-activated Cl- channels (CaCC) encoded by Ano1. Slow waves consist of an upstroke depolarization and a sustained plateau potential that is the main factor leading to excitation-contraction coupling. The plateau phase can last for seconds in some regions of the GI tract. How elevated Ca2+ is maintained throughout the duration of slow waves, which is necessary for sustained activation of CaCC, is unknown. Modeling has suggested a role for Na+/Ca2+ exchanger (NCX) in regulating CaCC currents in ICC, so we tested this idea on murine intestinal ICC. ICC of small and large intestine express NCX isoforms. NCX3 is closely associated with ANO1 in ICC, as shown by immunoprecipitation and proximity ligation assays (PLA). KB-R7943, an inhibitor of NCX, increased CaCC current in ICC, suggesting that NCX, acting in Ca2+ exit mode, helps to regulate basal [Ca2+] i in these cells. Shifting NCX into Ca2+ entry mode by replacing extracellular Na+ with Li+ increased spontaneous transient inward currents (STICs), due to activation of CaCC. Stepping ICC from -80 to -40 mV activated slow wave currents that were reduced in amplitude and duration by NCX inhibitors, KB-R7943 and SN-6, and enhanced by increasing the NCX driving force. SN-6 reduced the duration of clustered Ca2+ transients that underlie the activation of CaCC and the plateau phase of slow waves. Our results suggest that NCX participates in slow waves as modeling has predicted. Dynamic changes in membrane potential and ionic gradients during slow waves appear to flip the directionality of NCX, facilitating removal of Ca2+ during the inter-slow wave interval and providing Ca2+ for sustained activation of ANO1 during the slow wave plateau phase.

4.
Am J Physiol Gastrointest Liver Physiol ; 311(6): G1037-G1046, 2016 12 01.
Article in English | MEDLINE | ID: mdl-27742704

ABSTRACT

Interstitial cells of Cajal (ICC) generate electrical slow waves by coordinated openings of ANO1 channels, a Ca2+-activated Cl- (CaCC) conductance. Efflux of Cl- during slow waves must be significant, as there is high current density during slow-wave currents and slow waves are of sufficient magnitude to depolarize the syncytium of smooth muscle cells and PDGFRα+ cells to which they are electrically coupled. We investigated how the driving force for Cl- current is maintained in ICC. We found robust expression of Slc12a2 (which encodes an Na+-K+-Cl- cotransporter, NKCC1) and immunohistochemical confirmation that NKCC1 is expressed in ICC. With the use of the gramicidin permeabilized-patch technique, which is reported to not disturb [Cl-]i, the reversal potential for spontaneous transient inward currents (ESTICs) was -10.5 mV. This value corresponds to the peak of slow waves when they are recorded directly from ICC in situ. Inhibition of NKCC1 with bumetanide shifted ESTICs to more negative potentials within a few minutes and reduced pacemaker activity. Bumetanide had no direct effects on ANO1 or CaV3.2 channels expressed in HEK293 cells or L-type Ca2+ currents. Reducing extracellular Cl- to 10 mM shifted ESTICs to positive potentials as predicted by the Nernst equation. The relatively rapid shift in ESTICs when NKCC1 was blocked suggests that significant changes in the transmembrane Cl- gradient occur during the slow-wave cycle, possibly within microdomains formed between endoplasmic reticulum and the plasma membrane in ICC. Recovery of Cl- via NKCC1 might have additional consequences on shaping the waveforms of slow waves via Na+ entry into microdomains.


Subject(s)
Action Potentials , Chlorides/metabolism , Interstitial Cells of Cajal/metabolism , Solute Carrier Family 12, Member 2/metabolism , Animals , Bumetanide/pharmacology , Calcium Channels, T-Type/metabolism , Cells, Cultured , HEK293 Cells , Humans , Interstitial Cells of Cajal/drug effects , Interstitial Cells of Cajal/physiology , Mice , Periodicity , Sodium Potassium Chloride Symporter Inhibitors/pharmacology , Solute Carrier Family 12, Member 2/genetics
5.
Am J Physiol Cell Physiol ; 311(3): C437-51, 2016 09 01.
Article in English | MEDLINE | ID: mdl-27413167

ABSTRACT

Anoctamin-1 (ANO1) is a Ca(2+)-activated Cl(-) channel expressed in many types of cells. Splice variants of ANO1 have been shown to influence the biophysical properties of conductance. It has been suggested that several new antagonists of ANO1 with relatively high affinity and selectivity might be useful for experimental and, potentially, therapeutic purposes. We investigated the effects of intracellular Ca(2+) concentration ([Ca(2+)]i) at 100-1,000 nM, a concentration range that might be achieved in cells during physiological activation of ANO1 channels, on blockade of ANO1 channels expressed in HEK-293 cells. Whole cell and excised patch configurations of the patch-clamp technique were used to perform tests on a variety of naturally occurring splice variants of ANO1. Blockade of ANO1 currents with aminophenylthiazole (T16Ainh-A01) was highly dependent on [Ca(2+)]i Increasing [Ca(2+)]i reduced the potency of this blocker. Similar Ca(2+)-dependent effects were also observed with benzbromarone. Experiments on excised, inside-out patches showed that the diminished potency of the blockers caused by intracellular Ca(2+) might involve a competitive interaction for a common binding site or repulsion of the blocking drugs by electrostatic forces at the cytoplasmic surface of the channels. The degree of interaction between the channel blockers and [Ca(2+)]i depends on the splice variant expressed. These experiments demonstrate that the efficacy of ANO1 antagonists depends on [Ca(2+)]i, suggesting a need for caution when ANO1 blockers are used to determine the role of ANO1 in physiological functions and in their use as therapeutic agents.


Subject(s)
Alternative Splicing/genetics , Calcium/metabolism , Chloride Channels/metabolism , Alternative Splicing/drug effects , Animals , Anoctamin-1 , Benzbromarone/pharmacology , Cell Line , Cytoplasm/metabolism , HEK293 Cells , Humans , Mice , Patch-Clamp Techniques/methods , Pyrimidines/pharmacology , Thiazoles/pharmacology
6.
Am J Physiol Cell Physiol ; 308(8): C608-20, 2015 Apr 15.
Article in English | MEDLINE | ID: mdl-25631870

ABSTRACT

Interstitial cells of Cajal (ICC) provide pacemaker activity in gastrointestinal muscles that underlies segmental and peristaltic contractions. ICC generate electrical slow waves that are due to large-amplitude inward currents resulting from anoctamin 1 (ANO1) channels, which are Ca(2+)-activated Cl(-) channels. We investigated the hypothesis that the Ca(2+) responsible for the stochastic activation of ANO1 channels during spontaneous transient inward currents (STICs) and synchronized activation of ANO1 channels during slow wave currents comes from intracellular Ca(2+) stores. ICC, obtained from the small intestine of Kit(+/copGFP) mice, were studied under voltage and current clamp to determine the effects of blocking Ca(2+) uptake into stores and release of Ca(2+) via inositol 1,4,5-trisphosphate (IP3)-dependent and ryanodine-sensitive channels. Cyclocpiazonic acid, thapsigargin, 2-APB, and xestospongin C inhibited STICs and slow wave currents. Ryanodine and tetracaine also inhibited STICs and slow wave currents. Store-active compounds had no direct effects on ANO1 channels expressed in human embryonic kidney-293 cells. Under current clamp, store-active drugs caused significant depolarization of ICC and reduced spontaneous transient depolarizations (STDs). After block of ryanodine receptors with ryanodine and tetracaine, repolarization did not restore STDs. ANO1 expressed in ICC has limited access to cytoplasmic Ca(2+) concentration, suggesting that pacemaker activity depends on Ca(2+) dynamics in restricted microdomains. Our data from studies of isolated ICC differ somewhat from studies on intact muscles and suggest that release of Ca(2+) from both IP3 and ryanodine receptors is important in generating pacemaker activity in ICC.


Subject(s)
Calcium/metabolism , Chloride Channels/metabolism , Endoplasmic Reticulum/metabolism , Interstitial Cells of Cajal/metabolism , Ryanodine Receptor Calcium Release Channel/metabolism , Animals , Anoctamin-1 , Calcium Channel Blockers/pharmacology , Calcium-Transporting ATPases/antagonists & inhibitors , Cells, Cultured , Chloride Channels/biosynthesis , Enzyme Activation , Enzyme Inhibitors/pharmacology , HEK293 Cells , Humans , Indoles/pharmacology , Inositol 1,4,5-Trisphosphate/chemistry , Intestine, Small/cytology , Macrocyclic Compounds/pharmacology , Membrane Potentials/drug effects , Mice , Muscle Contraction/physiology , Myocytes, Smooth Muscle/metabolism , Oxazoles/pharmacology , Patch-Clamp Techniques , Ryanodine/pharmacology , Thapsigargin/pharmacology
8.
BMC Mol Biol ; 12: 35, 2011 Aug 08.
Article in English | MEDLINE | ID: mdl-21824394

ABSTRACT

BACKGROUND: TMEM16A (Anoctamin 1; ANO1) is an eight transmembrane protein that functions as a calcium-activated chloride channel. TMEM16A in human exhibits alternatively spliced exons (6b, 13 and 15), which confer important roles in the regulation of channel function. Mouse Tmem16a is reported to consist of 25 exons that code for a 956 amino acid protein. In this study our aim was to provide details of mouse Tmem16a genomic structure and to investigate if Tmem16a transcript undergoes alternative splicing to generate channel diversity. RESULTS: We identified Tmem16a transcript variants consisting of alternative exons 6b, 10, 13, 14, 15 and 18. Our findings indicate that many of these exons are expressed in various combinations and that these splicing events are mostly conserved between mouse and human. In addition, we confirmed the expression of these exon variants in other mouse tissues. Additional splicing events were identified including a novel conserved exon 13b, tandem splice sites of exon 1 and 21 and two intron retention events. CONCLUSION: Our results suggest that Tmem16a gene is significantly more complex than previously described. The complexity is especially evident in the region spanning exons 6 through 16 where a number of the alternative splicing events are thought to affect calcium sensitivity, voltage dependence and the kinetics of activation and deactivation of this calcium-activated chloride channel. The identification of multiple Tmem16a splice variants suggests that alternative splicing is an exquisite mechanism that operates to diversify TMEM16A channel function in both physiological and pathophysiological conditions.


Subject(s)
Alternative Splicing , Chloride Channels/genetics , Chloride Channels/metabolism , Amino Acid Sequence , Animals , Anoctamin-1 , Computational Biology , Exons , Humans , Introns , Mice , Molecular Sequence Data
9.
J Physiol ; 589(Pt 18): 4565-82, 2011 Sep 15.
Article in English | MEDLINE | ID: mdl-21768263

ABSTRACT

Interstitial cells of Cajal (ICC) provide pacemaker activity and functional bridges between enteric motor nerve terminals and gastrointestinal smooth muscle cells. The ionic conductance(s) in ICC that are activated by excitatory neural inputs are unknown. Transgenic mice (Kit(copGFP/+)) with constitutive expression of a bright green fluorescent protein were used to investigate cellular responses of ICC to cholinergic stimulation. ICC displayed spontaneous transient inward currents (STICs) under voltage clamp that corresponded to spontaneous transient depolarizations (STDs) under current clamp. STICs reversed at 0 mV when E(Cl) = 0 mV and at -40 mV when E(Cl) was -40 mV, suggesting the STICs were due to a chloride conductance. Carbachol (CCh, 100 nm and 1 µm) induced a sustained inward current (depolarization in current clamp) and increased the amplitude and frequency of STICs and STDs. CCh responses were blocked by atropine (10 µm) or 4-DAMP (100 nm), an M(3) receptor antagonist. STDs were blocked by niflumic acid and 5-nitro-2-(3-phenylpropylamino)-benzoic acid (both 100 µm), and CCh had no effect in the presence of these drugs. The responses of intact circular muscles to CCh and stimulation of intrinsic excitatory nerves by electrical field stimulation (EFS) were also compared. CCh (1 µm) caused atropine-sensitive depolarization and increased the maximum depolarization of slow waves. Similar atropine-sensitive responses were elicited by stimulation of intrinsic excitatory neurons. Niflumic acid (100 µm) blocked responses to EFS but had minor effect on responses to exogenous CCh. These data suggest that different ionic conductances are responsible for electrical responses elicited by bath-applied CCh and cholinergic nerve stimulation.


Subject(s)
Chloride Channels/physiology , Interstitial Cells of Cajal/physiology , Muscarinic Agonists/pharmacology , Muscarinic Antagonists/pharmacology , Receptors, Muscarinic/physiology , Animals , Atropine/pharmacology , Carbachol/pharmacology , Chloride Channels/drug effects , Green Fluorescent Proteins/genetics , Interstitial Cells of Cajal/cytology , Interstitial Cells of Cajal/drug effects , Intestine, Small/cytology , Membrane Potentials/drug effects , Membrane Potentials/physiology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Models, Animal , Patch-Clamp Techniques , Piperidines/pharmacology , Receptors, Muscarinic/drug effects
10.
J Physiol ; 587(Pt 20): 4887-904, 2009 Oct 15.
Article in English | MEDLINE | ID: mdl-19687122

ABSTRACT

Interstitial cells of Cajal (ICC) generate pacemaker activity (slow waves) in gastrointestinal (GI) smooth muscles, but the mechanism(s) of pacemaker activity are controversial. Several conductances, such as Ca(2+)-activated Cl() channels (CaCC) and non-selective cation channels (NSCC) have been suggested to be involved in slow wave depolarization. We investigated the expression and function of a new class of CaCC, anoctamin 1 (ANO1), encoded by Tmem16a, which was discovered to be highly expressed in ICC in a microarray screen. GI muscles express splice variants of the Tmem16a transcript in addition to other paralogues of the Tmem16a family. ANO1 protein is expressed abundantly and specifically in ICC in all regions of the murine, non-human primate (Macaca fascicularis) and human GI tracts. CaCC blocking drugs, niflumic acid and 4,4-diisothiocyano-2,2-stillbene-disulfonic acid (DIDS) reduced the frequency and blocked slow waves in murine, primate, human small intestine and stomach in a concentration-dependent manner. Unitary potentials, small stochastic membrane depolarizations thought to underlie slow waves, were insensitive to CaCC blockers. Slow waves failed to develop by birth in mice homozygous for a null allele of Tmem16a (Tmem16a(tm1Bdh)(/tm1Bdh)) and did not develop subsequent to birth in organ culture, as in wildtype and heterozygous muscles. Loss of function of ANO1 did not inhibit the development of ICC networks that appeared structurally normal as indicated by Kit antibodies. These data demonstrate the fundamental role of ANO1 in the generation of slow waves in GI ICC.


Subject(s)
Gastrointestinal Motility/physiology , Gastrointestinal Tract/physiology , Interstitial Cells of Cajal/physiology , Membrane Proteins/metabolism , Muscle, Smooth/physiology , Neoplasm Proteins/metabolism , 4,4'-Diisothiocyanostilbene-2,2'-Disulfonic Acid/pharmacology , Animals , Anoctamin-1 , Chloride Channels , Cyclooxygenase Inhibitors/pharmacology , Gastrointestinal Motility/drug effects , Gastrointestinal Tract/cytology , Gastrointestinal Tract/drug effects , Gene Expression Regulation , Humans , Immunohistochemistry , Interstitial Cells of Cajal/cytology , Interstitial Cells of Cajal/drug effects , Macaca fascicularis , Membrane Proteins/genetics , Mice , Muscle, Smooth/cytology , Muscle, Smooth/drug effects , Neoplasm Proteins/genetics , Niflumic Acid/pharmacology , RNA/analysis , RNA/genetics , Reverse Transcriptase Polymerase Chain Reaction
11.
Biochem Biophys Res Commun ; 384(4): 476-81, 2009 Jul 10.
Article in English | MEDLINE | ID: mdl-19426717

ABSTRACT

Bestrophins form Ca2+-activated Cl- channels when they are expressed heterologously. Here we report the functional characterization of murine bestrophin 1 (mBest1). We isolated mBest1 transcript from mouse heart and analyzed the biophysical properties and expression of this channel protein using a tetracycline inducible system. mBest1 expression is localized at the membrane of transfected HEK cells, in agreement with its role as a channel. Whole-cell patch clamp experiments revealed a calcium sensitive, time independent chloride current. mBest1 current displayed slight voltage dependence, exhibited an anion permeability sequence of SCN- > I- > Cl- and was sensitive to DIDS and niflumic acid. Anion replacement studies were also performed on mBest2 and mBest3 and differences were observed in their relative permeability and slope conductance to SCN-. Our study provides the first characterization of the biophysical properties of mBest1 and a framework for the elucidation of the physiological role of bestrophins.


Subject(s)
Chloride Channels/physiology , Animals , Bestrophins , Cell Line , Cell Membrane/metabolism , Cell Membrane/physiology , Chloride Channels/genetics , Chloride Channels/metabolism , Membrane Potentials , Mice
12.
Am J Physiol Cell Physiol ; 295(6): C1610-24, 2008 Dec.
Article in English | MEDLINE | ID: mdl-18945938

ABSTRACT

Bestrophins are a novel family of proteins that encode calcium-activated chloride channels. In this study we establish that Bestrophin transcripts are expressed in the mouse and human heart. Native mBest3 protein expression and localization in heart was demonstrated by using a specific polyclonal mBest3 antibody. Immunostaining of isolated cardiac myocytes indicates that mBest3 is present at the membrane. Using the patch-clamp technique, we characterized the biophysical and pharmacological properties of mBest3 cloned from heart. Whole cell chloride currents were evoked in both HEK293 and COS-7 cells expressing mBest3 by elevation of intracellular calcium. mBest3 currents displayed a K(D) for Ca(2+) of approximately 175 nM. The calcium-activated chloride current was found to be time and voltage independent and displayed slight outward rectification. The anion permeability sequence of the channel was SCN(-)>I(-)>Cl(-), and the current was inhibited by niflumic acid and DIDS in the micromolar range. In addition, we generated a site-specific mutation (F80L) in the putative pore region of mBest3 that significantly altered the ion conduction and pharmacology of this channel. Our functional and mutational studies examining the biophysical properties of mBest3 indicate that it functions as a pore-forming chloride channel that is activated by physiological levels of calcium. This study reports novel findings regarding the molecular expression, tissue localization, and functional properties of mBest3 cloned from heart.


Subject(s)
Chloride Channels/physiology , Heart/physiology , Muscle Proteins/physiology , Amino Acid Sequence , Animals , Bestrophins , Blotting, Western , Humans , Mice , Mice, Inbred BALB C , Microscopy, Confocal , Molecular Sequence Data , Mutagenesis, Site-Directed , Patch-Clamp Techniques , Reverse Transcriptase Polymerase Chain Reaction , Sequence Homology, Amino Acid
13.
Can J Physiol Pharmacol ; 83(7): 541-56, 2005 Jul.
Article in English | MEDLINE | ID: mdl-16091780

ABSTRACT

Calcium-activated chloride channels (ClCa) are ligand-gated anion channels as they have been shown to be activated by a rise in intracellular Ca2+ concentration in various cell types including cardiac, skeletal and vascular smooth muscle cells, endothelial and epithelial cells, as well as neurons. Because ClCa channels are normally closed at resting, free intracellular Ca2+ concentration (approximately 100 nmol/L) in most cell types, they have generally been considered excitatory in nature, providing a triggering mechanism during signal transduction for membrane excitability, osmotic balance, transepithelial chloride movements, or fluid secretion. Unfortunately, the genes responsible for encoding this class of ion channels is still unknown. This review centers primarily on recent findings on the properties of these channels in smooth muscle cells. The first section discusses the functional significance and biophysical and pharmacological properties of ClCa channels in smooth muscle cells, and ends with a description of 2 candidate gene families (i.e., CLCA and Bestrophin) that are postulated to encode for these channels in various cell types. The second section provides a summary of recent findings demonstrating the regulation of native ClCa channels in vascular smooth muscle cells by calmodulin-dependent protein kinase II and calcineurin and how their fine tuning by these enzymes may influence vascular tone.


Subject(s)
Calcium/physiology , Chloride Channels/physiology , Muscle, Smooth/physiology , Animals , Chloride Channels/genetics , Gene Expression Regulation/physiology , Humans , Muscle, Smooth/cytology
14.
Br J Pharmacol ; 137(8): 1321-9, 2002 Dec.
Article in English | MEDLINE | ID: mdl-12466242

ABSTRACT

1. Motor innervation in the canine rectoanal region was examined in isolated strips of the circular muscle layer. Contractile responses to electrical field stimulation began at lower frequencies and were more persistent in the internal anal sphincter (IAS) than in the rectum. 2. Motor innervation to the IAS was almost exclusively sympathetic, since it was blocked by guanethidine (Guan 3 microM) while the response in the proximal rectum was approximately 50% muscarinic, and sensitive to the M(3) selective antagonist 4-diphenylacetoxy-N-methylpiperidine (4-DAMP, 0.1 microM) and 50% tachykinergic, and sensitive to the neurokinin 2 (NK(2)) receptor antagonist GR 94800 (1 microM). From IAS to rectum there was a gradual shift in the relative contribution of intrinsic and extrinsic neural innervation. 3. Responses to exogenously applied transmitters exhibited a similar pattern to that observed with motor innervation. Norepinephrine (NE) was most potent in the IAS and acetylcholine (ACh) and NK-A were most potent in the proximal rectum. The responses were inhibited by prazosin, 4-DAMP and GR 94800 respectively. 4. A gradient in the density of adrenergic alpha(1), muscarinic and NK(2) receptors also existed from IAS to rectum as determined by measuring the binding of [(3)H]-prazosin, [(3)H]-quinuclidinyl benzilate ([(3)H]-QNB and [(3)H]-SR-48968 to smooth muscle membranes. 5. In summary, these data suggest that the shift in motor innervation in the rectoanal region is achieved in part by changes in receptor populations available for activation by sympathetic and enteric motor neurons.


Subject(s)
Anal Canal/innervation , Motor Neurons/physiology , Receptors, Adrenergic, alpha-1/physiology , Receptors, Muscarinic/physiology , Receptors, Neurokinin-2/physiology , Rectum/innervation , Adrenergic alpha-1 Receptor Antagonists , Anal Canal/drug effects , Anal Canal/physiology , Animals , Dogs , Electric Stimulation/methods , Female , In Vitro Techniques , Male , Motor Neurons/drug effects , Muscle Contraction/drug effects , Muscle Contraction/physiology , Receptors, Neurokinin-2/antagonists & inhibitors , Rectum/drug effects , Rectum/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...