Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
J Physiol ; 593(20): 4589-601, 2015 Oct 15.
Article in English | MEDLINE | ID: mdl-26227063

ABSTRACT

In the enteric nervous systems, NO is released from nitrergic neurons as a major inhibitory neurotransmitter. NO acts via NO-sensitive guanylyl cyclase (NO-GC), which is found in different gastrointestinal (GI) cell types including smooth muscle cells (SMCs) and interstitial cells of Cajal (ICC). The precise mechanism of nitrergic signalling through these two cell types to regulate colonic spontaneous contractions is not fully understood yet. In the present study we investigated the impact of endogenous and exogenous NO on colonic contractile motor activity using mice lacking nitric oxide-sensitive guanylyl cyclase (NO-GC) globally and specifically in SMCs and ICC. Longitudinal smooth muscle of proximal colon from wild-type (WT) and knockout (KO) mouse strains exhibited spontaneous contractile activity ex vivo. WT and smooth muscle-specific guanylyl cyclase knockout (SMC-GCKO) colon showed an arrhythmic contractile activity with varying amplitudes and frequencies. In contrast, colon from global and ICC-specific guanylyl cyclase knockout (ICC-GCKO) animals showed a regular contractile rhythm with constant duration and amplitude of the rhythmic contractions. Nerve blockade (tetrodotoxin) or specific blockade of NO signalling (L-NAME, ODQ) did not significantly affect contractions of GCKO and ICC-GCKO colon whereas the arrhythmic contractile patterns of WT and SMC-GCKO colon were transformed into uniform motor patterns. In contrast, the response to electric field-stimulated neuronal NO release was similar in SMC-GCKO and global GCKO. In conclusion, our results indicate that basal enteric NO release acts via myenteric ICC to influence the generation of spontaneous contractions whereas the effects of elevated endogenous NO are mediated by SMCs in the murine proximal colon.


Subject(s)
Colon/physiology , Interstitial Cells of Cajal/physiology , Muscle Contraction/physiology , Muscle, Smooth/physiology , Nitric Oxide/physiology , Animals , Guanylate Cyclase/genetics , Guanylate Cyclase/physiology , Male , Mice, Inbred C57BL , Mice, Knockout , Myocytes, Smooth Muscle/physiology , Receptors, Cytoplasmic and Nuclear/genetics , Receptors, Cytoplasmic and Nuclear/physiology , Signal Transduction , Soluble Guanylyl Cyclase
2.
J Physiol ; 593(2): 403-14, 2015 Jan 15.
Article in English | MEDLINE | ID: mdl-25630261

ABSTRACT

Oesophageal achalasia is a disease known to result from reduced relaxation of the lower oesophageal sphincter (LES). Nitric oxide (NO) is one of the main inhibitory transmitters. NO-sensitive guanylyl cyclase (NO-GC) acts as the key target of NO and, by the generation of cGMP, mediates nitrergic relaxation in the LES. To date, the exact mechanism of nitrergic LES relaxation is still insufficiently elucidated. To clarify the role of NO-GC in LES relaxation, we used cell-specific knockout (KO) mouse lines for NO-GC. These include mice lacking NO-GC in smooth muscle cells (SMC-GCKO), in interstitial cells of Cajal (ICC-GCKO) and in both SMC/ICC (SMC/ICC-GCKO). We applied oesophageal manometry to study the functionality of LES in vivo. Isometric force studies were performed to monitor LES responsiveness to exogenous NO and electric field stimulation of intrinsic nerves in vitro. Cell-specific expression/deletion of NO-GC was monitored by immunohistochemistry. Swallowing-induced LES relaxation is strongly reduced by deletion of NO-GC in ICC. Basal LES tone is affected by NO-GC deletion in either SMC or ICC. Lack of NO-GC in both cells leads to a complete interruption of NO-induced relaxation and, therefore, to an achalasia-like phenotype similar to that seen in global GCKO mice. Our data indicate that regulation of basal LES tone is based on a dual mechanism mediated by NO-GC in SMC and ICC whereas swallow-induced LES relaxation is mainly regulated by nitrergic mechanisms in ICC.


Subject(s)
Esophageal Sphincter, Lower/metabolism , Guanylate Cyclase/metabolism , Interstitial Cells of Cajal/metabolism , Muscle Relaxation , Nitric Oxide/metabolism , Receptors, Cytoplasmic and Nuclear/metabolism , Animals , Esophageal Sphincter, Lower/cytology , Esophageal Sphincter, Lower/physiology , Guanylate Cyclase/genetics , Interstitial Cells of Cajal/physiology , Isometric Contraction , Mice , Mice, Inbred C57BL , Myocytes, Smooth Muscle/metabolism , Myocytes, Smooth Muscle/physiology , Receptors, Cytoplasmic and Nuclear/genetics , Soluble Guanylyl Cyclase
3.
Am J Physiol Gastrointest Liver Physiol ; 307(1): G98-106, 2014 Jul 01.
Article in English | MEDLINE | ID: mdl-24833707

ABSTRACT

Nitric oxide (NO) is a major inhibitory neurotransmitter in the gastrointestinal (GI) tract. Its main effector, NO-sensitive guanylyl cyclase (NO-GC), is expressed in several GI cell types, including smooth muscle cells (SMC), interstitial cells of Cajal (ICC), and fibroblast-like cells. Up to date, the interplay between neurons and these cells to initiate a nitrergic inhibitory junction potential (IJP) is unclear. Here, we investigate the origin of the nitrergic IJP in murine fundus and colon. IJPs were determined in fundus and colon SMC of mice lacking NO-GC globally (GCKO) and specifically in SMC (SM-GCKO), ICC (ICC-GCKO), and both SMC/ICC (SM/ICC-GCKO). Nitrergic IJP was abolished in ICC-GCKO fundus and reduced in SM-GCKO fundus. In the colon, the amplitude of nitrergic IJP was reduced in ICC-GCKO, whereas nitrergic IJP in SM-GCKO was reduced in duration. These results were corroborated by loss of the nitrergic IJP in global GCKO. In conclusion, our results prove the obligatory role of NO-GC in ICC for the initiation of an IJP. NO-GC in SMC appears to enhance the nitrergic IJP, resulting in a stronger and prolonged hyperpolarization in fundus and colon SMC, respectively. Thus NO-GC in both cell types is mandatory to induce a full nitrergic IJP. Our data from the colon clearly reveal the nitrergic IJP to be biphasic, resulting from individual inputs of ICC and SMC.


Subject(s)
Colon/innervation , Gastric Fundus/innervation , Interstitial Cells of Cajal/metabolism , Neural Inhibition , Nitrergic Neurons/metabolism , Nitric Oxide/metabolism , Synaptic Transmission , Animals , Guanylate Cyclase/genetics , Guanylate Cyclase/metabolism , Inhibitory Postsynaptic Potentials , Mice , Mice, Inbred C57BL , Mice, Knockout , Myocytes, Smooth Muscle/metabolism , Time Factors
5.
J Physiol ; 591(21): 5365-75, 2013 Nov 01.
Article in English | MEDLINE | ID: mdl-24018948

ABSTRACT

The action of nitric oxide (NO) to stimulate NO-sensitive guanylyl cyclase (NO-GC), followed by production of cGMP, and eventually to cause smooth muscle relaxation is well known. In the lower urinary tract (LUT), in contrast to the cardiovascular system and the gastrointestinal tract, specific localization in combination with function of NO-GC has not been investigated to date. Consequently, little is known about the mechanisms regulating relaxation of the lower urinary tract in general and the role of NO-GC-expressing cells in particular. To study the distribution and function of NO-GC in the murine lower urinary tract, we used internal urethral sphincter and bladder detrusor from global (GCKO) and smooth muscle cell-specific (SM-GCKO) NO-GC knock-out mice for immunohistochemical analyses and organ bath experiments. In urethral sphincter, NO-GC-positive immunofluorescence was confined to smooth muscle cells (SMCs). Deletion of NO-GC in SMCs abolished NO-induced relaxation. In bladder detrusor, exposure to NO did not cause relaxation although immunohistochemistry uncovered the existence of NO-GC in the tissue. In contrast to the urethral sphincter, expression of NO-GC in bladder detrusor was limited to platelet-derived growth factor receptor α (PDGFRα)-positive interstitial cells. In conclusion, NO-GC found in SMCs of the urethral sphincter mediates NO-induced relaxation; bladder detrusor is unique as NO-GC is not expressed in SMCs and, thus, NO does not induce relaxation. Nevertheless, NO-GC expression was found in PDGFRα-positive interstitial cells of the murine bladder with an as yet unknown function. Further investigation is needed to clarify the role of NO-GC in the detrusor.


Subject(s)
Guanylate Cyclase/metabolism , Receptors, Cytoplasmic and Nuclear/metabolism , Urethra/metabolism , Urinary Bladder/metabolism , Animals , Guanylate Cyclase/genetics , Mice , Mice, Inbred C57BL , Muscle Relaxation , Myocytes, Smooth Muscle/enzymology , Receptors, Cytoplasmic and Nuclear/genetics , Soluble Guanylyl Cyclase , Urethra/cytology , Urinary Bladder/cytology
6.
Br J Pharmacol ; 170(2): 317-27, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23763290

ABSTRACT

BACKGROUND AND PURPOSE: Nitric oxide (NO) is known to activate NO-sensitive guanylyl cyclase (NO-GC) and to elicit cGMP production. However, NO has also been proposed to induce cGMP-independent effects. It is accepted practice to use specific NO-GC inhibitors, such as ODQ or NS2028, to assess cGMP-dependent NO effects. Consequently, NO-induced reactions seen in the presence of these inhibitors commonly serve as an affirmation of cGMP independence. EXPERIMENTAL APPROACH: We evaluated the use of ODQ to discriminate between cGMP-dependent and cGMP-independent NO effects. NO-GC-expressing HEK cells, platelets and tissues from wild type (WT) and NO-GC-deficient mice (GCKO) were used. KEY RESULTS: NO donors led to accumulation of cGMP in platelets and GC-expressing HEK cells and induced phosphorylation of the vasodilator-stimulated phosphoprotein in platelets; both effects were reduced by ODQ. High concentrations of NO donors, however, overrode the inhibitory effect of ODQ. Correspondingly, ODQ inhibited but did not fully eliminate NO-induced relaxation of aorta and fundus from WT mice. Relaxation induced by endogenously released NO was fully or partially inhibited by ODQ in fundus and aorta, respectively. In aorta and fundus of GCKO mice NO-induced relaxation was absent and served as standard for complete NO-GC inhibition. CONCLUSIONS AND IMPLICATIONS: High NO concentrations can overcome the inhibitory effect of ODQ on NO-GC. Smooth muscle relaxation induced by NO donors/endogenously released NO in the presence of ODQ in WT was absent in GCKO animals indicating involvement of NO-GC. Accordingly, NO-induced effects in the presence of ODQ do not necessarily prove cGMP independence.


Subject(s)
Cyclic GMP/metabolism , Guanylate Cyclase/metabolism , Nitric Oxide/metabolism , Oxadiazoles/pharmacology , Adult , Animals , Aorta/drug effects , Aorta/metabolism , Female , Guanylate Cyclase/genetics , HEK293 Cells , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Muscle Relaxation/drug effects , Muscle Relaxation/physiology , Muscle, Smooth/drug effects , Muscle, Smooth/metabolism , Nitric Oxide Donors/pharmacology , Oxazines/pharmacology , Signal Transduction/drug effects , Signal Transduction/physiology
7.
Methods Mol Biol ; 1020: 63-72, 2013.
Article in English | MEDLINE | ID: mdl-23709026

ABSTRACT

Radioimmunoassay is an established method to determine the amount of a specific substance in a given cell or tissue sample. Commercially available RIA or Elisa are very cost intensive. Here, we describe the generation of radioactive cGMP tracer and the quantification of cGMP. Although working with radioactive material requires experience and care, this method is very sensitive and rather cheap, once it is established.


Subject(s)
Cyclic GMP/chemistry , Radioimmunoassay/methods , Charcoal/analysis , Charcoal/chemistry , Cyclic GMP/analysis , Halogenation , Humans
8.
Gastroenterology ; 145(1): 188-196, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23528627

ABSTRACT

BACKGROUND & AIMS: It is not clear how nitric oxide (NO) released from enteric neurons relaxes gastrointestinal (GI) smooth muscle. In analogy to the vascular system, NO might directly induce relaxation of smooth muscle cells (SMCs) by acting on its receptor, NO-sensitive guanylyl cyclase (NO-GC). Alternatively, intermediate cells, such as the interstitial cells of Cajal (ICCs), might detect nitrergic signals to indirectly regulate smooth muscle tone, and thereby regulate the motor function of the GI tract. We investigated the role of ICCs and SMCs in nitrergic relaxation using mice with cell-specific disruption of the gene encoding the ß1 subunit of NO-GC (GUCY1B3). METHODS: We created mice that lack NO-GC specifically in SMCs (SM-guanylyl cyclase knockout [GCKO]), ICCs (ICC-GCKO), or both (SM/ICC-GCKO). We investigated the effects of exogenous and endogenous NO on murine fundus using isometric force studies. Total gut transit time was measured to monitor the functional consequences of NO-GC deletion on GI motility in vivo. RESULTS: NO-GC is expressed in ICC and SMC. Deletion of the NO receptor from SMCs incompletely reduced NO-induced fundus relaxation, which was hardly affected after ICC-specific deletion. Gut transit time did not change in SM-GCKO or ICC-GCKO mice compared with control mice. However, nitrergic relaxation was not observed in SM/ICC-GCKO mice, which had increased gut transit time compared with controls. CONCLUSIONS: In mice, NO-GC is the only NO receptor to relax the fundus; deletion of NO-GC from the combination of SMCs and ICCs blocks nitrergic signaling. Therefore, ICCs and SMCs jointly mediate the relaxant effect of enteric NO.


Subject(s)
Gastric Fundus/physiology , Guanylate Cyclase/physiology , Interstitial Cells of Cajal/physiology , Myocytes, Smooth Muscle/physiology , Nitric Oxide/physiology , Signal Transduction/physiology , Animals , Electric Stimulation , Gastrointestinal Motility , Mice , Muscle Relaxation/drug effects , Nifedipine/pharmacology
9.
J Physiol ; 591(2): 491-502, 2013 Jan 15.
Article in English | MEDLINE | ID: mdl-23129789

ABSTRACT

Nitric oxide (NO) and cGMP have been shown to be important mediators of penile erection. Erectile dysfunction may result from reduced or non-functional signal transduction within this cascade. There is, however, some inconsistency in the available data as mice lacking NO synthases (endothelial and neuronal nitric oxide synthase, or both) appear to be fertile whereas mice deficient in cGMP-dependent protein kinase I (PKGI) suffer from erectile dysfunction. To clarify this discrepancy we performed studies on mice lacking the NO receptor NO-sensitive guanylyl cyclase (NO-GC). In addition, we generated cell-specific NO-GC knockout (KO) lines to investigate the function of NO in individual cell types. NO-GC was specifically deleted in smooth muscle or endothelial cells (SM-guanylyl cyclase knockout (SM-GCKO) and EC-GCKO, respectively) and these KO lines were compared with total knockouts (GCKO) and wild-type animals. We investigated expression of NO-GC, NO-induced relaxation of corpus cavernosum smooth muscle and their ability to generate offspring. NO-GC-positive immunostaining was detected in smooth muscle and endothelial cells of murine corpus cavernosum but not in interstitial cells of Cajal. NO released from NO donors as well as from nitrergic neurons failed to relax precontracted corpus cavernosum from GCKO mice in organ bath experiments. Similar results were obtained in corpus cavernosum from SM-GCKO mice whereas deletion of NO-GC in endothelial cells did not affect relaxation. The lack of NO-induced relaxation in GCKO animals was not compensated for by guanosine 3,5-cyclic monophosphate (cGMP) signalling. To our surprise, GCKO males were fertile although their ability to produce offspring was decreased. Our data show that deletion of NO-GC specifically in smooth muscle cells abolishes NO-induced corpus cavernosum relaxation but does not lead to infertility.


Subject(s)
Erectile Dysfunction/metabolism , Fertility/genetics , Guanylate Cyclase/metabolism , Receptors, Cytoplasmic and Nuclear/metabolism , Animals , Cyclic GMP/metabolism , Endothelial Cells/metabolism , Erectile Dysfunction/genetics , Erectile Dysfunction/physiopathology , Gene Deletion , Guanylate Cyclase/genetics , Interstitial Cells of Cajal/metabolism , Male , Mice , Mice, Knockout , Muscle Relaxation , Muscle, Smooth/metabolism , Neurons/metabolism , Nitric Oxide/metabolism , Penis/cytology , Receptors, Cytoplasmic and Nuclear/genetics , Soluble Guanylyl Cyclase
10.
Mol Nutr Food Res ; 56(11): 1627-35, 2012 Nov.
Article in English | MEDLINE | ID: mdl-22965487

ABSTRACT

SCOPE: Based on the studies suggesting that active transport mechanisms contribute to the absorption of flavonoids into human intestinal Caco-2 cells, we here used the structurally similar fluorescent rhodamine 123 to test a possible influence of flavonoids on its uptake. METHODS AND RESULTS: Rhodamine absorption displayed saturation kinetics with a K(m) of 1.1 µM and a pH-optimum of 8.5 and was stimulated by flavone four-fold in its V(max) . Ring C of the other 16 flavonoids tested turned out to be of special importance in order to act as potent inhibitors for rhodamine transport, with a positive charge there, as present in the anthocyanidins, or a 2,3 double bond together with an aromatic ring fused to position 2, as present in flavones and flavonols, being essential structural requirements. Flavone-stimulated rhodamine uptake was unaffected by classical substrates of organic cation transporters or inhibitors of adenosine triphosphate (ATP)-dependent efflux pumps. Also, inhibitors of mitogen-activated protein kinases or tyrosine kinases did not influence the transport, whose stimulation, however, was essentially dependent on the simultaneous presence of flavone. The existence of a flavone-activated apical flavonoid transporter in Caco-2 cells was finally associated with the potently diminished transepithelial apical to basolateral fluxes of (14) C-kaempferol in the presence of competing unlabeled flavonoid substrates. CONCLUSION: In conclusion, flavone activates an as yet unidentified transporter for flavonoids in the apical membrane of Caco-2 cells.


Subject(s)
Carrier Proteins/metabolism , Flavonoids/pharmacology , ATP Binding Cassette Transporter, Subfamily B/antagonists & inhibitors , ATP Binding Cassette Transporter, Subfamily B/metabolism , Absorption , Anthocyanins/pharmacology , Caco-2 Cells/drug effects , Caco-2 Cells/metabolism , Cell Membrane/metabolism , Chlorides/metabolism , Flavones/pharmacology , Guanidine/pharmacology , Humans , Hydrogen-Ion Concentration , Kaempferols/chemistry , Kaempferols/metabolism , Kinetics , MAP Kinase Signaling System/drug effects , Organic Cation Transport Proteins/antagonists & inhibitors , Protein-Tyrosine Kinases/metabolism , Rhodamines/pharmacokinetics , Sodium/metabolism , Verapamil/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...