Uncoupling protein-1: involvement in a novel pathway for beta -adrenergic, cAMP-mediated intestinal relaxation

Irina Shabalina1, Claes Wiklund2, Tore Bengtsson1, Anders Jacobsson1, Barbara Cannon1, and Jan Nedergaard1

1 Wenner-Gren Institute, Arrhenius Laboratories F3, Stockholm University, SE-106 91 Stockholm; and 2 Department of Anaesthesiology and Intensive Care Medicine, Karolinska Hospital and Institute, SE-171 76, Stockholm, Sweden


    ABSTRACT
TOP
ABSTRACT
INTRODUCTION
MATERIALS AND METHODS
RESULTS
DISCUSSION
REFERENCES

The pathway for adrenergic relaxation of smooth muscle is not fully understood. As mitochondrial uncoupling protein-1 (UCP1) expression has been reported in cells within the longitudinal smooth muscle layer of organs exhibiting peristalsis, we examined whether the absence of UCP1 affects adrenergic responsiveness. Intestinal (ileal) segments were obtained from UCP1-ablated mice and from wild-type mice (C57Bl/6, 129/SvPas, and outbred NMRI). In UCP1-containing mice, isoprenaline totally inhibited contractions induced by electrical field stimulation, but in intestine from UCP1-ablated mice, a significant residual contraction remained even at a high isoprenaline concentration; the segments were threefold less sensitive to isoprenaline. Also, when contraction was induced by carbachol, there was a residual isoprenaline-insensitive contraction. Similar results were obtained with the beta 3-selective agonist CL-316,243 and with the adenylyl cyclase stimulator forskolin. Thus the UCP1 reported to be expressed in the longitudinal muscle layer of the mouse intestine is apparently functional, and UCP1, presumably through uncoupling, may be involved in a novel pathway leading from increased cAMP levels to relaxation in organs exhibiting peristalsis.

carbachol; isoprenaline; forskolin


    INTRODUCTION
TOP
ABSTRACT
INTRODUCTION
MATERIALS AND METHODS
RESULTS
DISCUSSION
REFERENCES

SMOOTH MUSCLE TONE IS ACHIEVED by a balance between the actions of contraction- and relaxation-inducing agents. Intracellular signaling pathways for the corresponding processes have been proposed (for reviews see Refs. 38, 43, 53, 65, 73). The pathway from the muscarinic acetylcholine receptors to enhanced contraction clearly involves an increase in cytosolic Ca2+ levels. The corresponding pathway leading from stimulation of beta -adrenergic receptors and increases in cAMP to relaxation of phasic smooth muscle (36, 24, 73) is not fully understood. It involves a reduction in Ca2+ levels but also a reduction in the effectiveness of Ca2+ in invoking contraction; the mechanism behind this reduced effectiveness has not been clarified.

A novel possibility for explaining the reduced Ca2+ effectiveness has recently arisen unexpectedly based on observations originating from studies in an apparently unrelated field, that of nonshivering thermogenesis. The phenomenon of classic adaptive nonshivering thermogenesis is presently understood molecularly as being the outcome of the activity of the archetypal mitochondrial uncoupling protein-1 (UCP1; thermogenin) (reviewed in Refs. 16, 35, 51, 60) originally identified in brown adipose tissue (20, 40) and demonstrated to be the only protein able to mediate nonshivering thermogenesis (17). UCP1 functions as a regulated transporter for proton equivalents over the mitochondrial membrane, leading to energy being released as heat instead of being captured in ATP. Until recently, UCP1 has been considered to be expressed only in brown adipose tissue (3, 25), to the extent that its occurrence in a given adipose cell has become the established criterion distinguishing brown and white adipose tissue (4). Conversely, no indication of UCP1 expression in other tissues has existed that could not be understood as being caused by contamination of extracts with brown adipocytes or being explainable based on the existence of the recently identified closely related proteins UCP2 or UCP3 (2). However, in a recent investigation, Nibbelink et al. (52) have reported that UCP1 is expressed very specifically in cells that seem to be included among the longitudinal smooth muscle layer of a series of organs, all characterized by exhibiting peristaltic motion: stomach, eosophagus, small intestine, ureter origin, ductus deferens, and uterus. In the cells that apparently contain UCP1, the level of UCP1 protein was of a magnitude similar to that in brown adipocytes (52). UCP1 was not found in arterial walls; and although arterial walls contain smooth muscle cells, they do not demonstrate peristaltic movements.

Functionally, the UCP1 could be present in these organs for the same reason that it is in brown adipose tissue, i.e., for thermogenesis. However, in classic experiments, Depocas (8, 9) showed that norepinephrine-induced thermogenesis in cold-acclimated rats was unaffected by functional evisceration, demonstrating that at least at the systemic level, no contribution was observable from any thermogenic process in the abdomen. It is therefore an interesting possibility that, in these organs, UCP1 has a functional role that is not thermogenic. In brown adipose tissue, the activation of UCP1 (thermogenesis) is induced by beta -adrenergic receptors and increased cAMP (75, 76). In different organs exhibiting peristaltic motion, beta -adrenergic receptors also have a role: to induce relaxation/inhibition of contraction via an increase in cAMP levels (22, 24, 26, 36, 68, 73). Other relaxing agents [VIP, pituitary adenylate cyclase-activating polypeptide (PACAP)] have also been suggested as inducing relaxation through this cAMP-dependent pathway (for reviews see Refs. 43 and 53). Because activated UCP1 in the mitochondria of smooth muscle cells may functionally explain the lowered effectiveness of Ca2+ in inducing contraction observed under these conditions (see DISCUSSION), we have here utilized UCP1-ablated mice to examine whether a UCP1-dependent component exists in beta -adrenergically induced relaxation.


    MATERIALS AND METHODS
TOP
ABSTRACT
INTRODUCTION
MATERIALS AND METHODS
RESULTS
DISCUSSION
REFERENCES

Animals

The UCP1-ablated [UCP1(-/-)] mice were progeny of those described (10) in which the gene encoding UCP1 had been inactivated by homologous recombination with a deletion vector in which exon 2 and parts of exon 3 had been replaced by a neomycin resistance gene; in the brown adipose tissue of these mice, no UCP1 can be detected with polyclonal antibodies (10; and unpublished observations). The mice were bred in the institute and were phenotypically [e.g., hair color as well as body wt and growth rate on standard diet and at normal (24°C) animal house temperature] indistinguishable from mice of the C57Bl/6 strain, which were the donors of the blastocysts. Genetically, the UCP1(-/-) mice are infiltrated with 129/SvJ (from the embryonic stem cells) and 129/SvPas (to which the chimeras were bred). Thus for the present experiments, three types of UCP1-containing [UCPI(+/+)] mice were used: the C57Bl/6 strain (B&K Universal, Stockholm, Sweden), a 129/SvPas strain (Charles River, Cleon, France), and genetically outbred NMRI mice (B&K Universal). All animals were fed ad libitum (rat and mouse standard diet No. 1; BeeKay Feeds, B&K Universal), had free access to water, and were kept on a 12:12-h light-dark cycle. Mice of 8-to-10-wk age and of both sexes were acclimated (1 per cage) to 24°C for at least 2 wk before the start of the experiments.

Tissue Collection

Routinely on each experimental day, one wild-type and one UCP1(-/-) mouse (weighing 20-22 g) were anesthetized for 1-2 min by a mixture of 79% CO2-21% O2 and killed by cervical dislocation. Ileum (~10 cm) was removed 2 cm above the ileocaecal junction and immediately placed in preoxygenated physiological salt solution composed of (in mM) 149 Na+, 2.9 K+, 1.8 Ca2+, 0.5 Mg2+, 144 Cl-, 23.8 HCO<UP><SUB>3</SUB><SUP>−</SUP></UP>, 0.4 H2PO<UP><SUB>4</SUB><SUP>−</SUP></UP>, and 5.5 glucose and aerated with 5% CO2 in O2. Adjacent mesentery was gently cut away from the ileum segments. Ileal segments of ~1.0-cm length were cut, and suture needles were used to pass silicone-treated silk thread (5-0 size) through a single wall on each end of the ileal segment.

Organ Bath Studies

Routinely, eight ileal segments (4 from each type of mice) were mounted vertically in 25-ml water-jacketed organ baths made of glass (Section of Engineering, Mayo Clinic) filled with the salt solution detailed above, maintained at 37°C, and bubbled continuously with 5% CO2 in O2. During the experiments, the solution in the baths was changed at 20- to 30-min intervals by superfusion of aerated solution preheated to 37°C. The segment preparations were given an initial isometric load of 2.5 mN (250 mg weight) and were allowed to equilibrate for 2-3 h before the experiments were started. We observed that the time required to obtain stable responses to electrical field stimulation (EFS) (see EFS) appeared to be longer in segments from UCP1(-/-) mice, but as experiments were routinely performed in parallel with segments from wild-type and UCP1(-/-) mice, the preincubation time was the same. The motor activity was recorded isometrically by Grass force-displacement transducers (FT03) and tracings were made on a polygraph (model 79, Grass Instruments) (72).

EFS

Isolated ileal segment preparations were submitted to EFS by use of a direct current amplifier (Section of Engineering, Mayo Clinic) triggered by a stimulator (model S44, Grass Medical Instruments) via two parallel platinum electrodes (10 × 50 mm) 8 mm apart. This type of stimulation leads to contraction; this contraction may be due to a direct electrical effect on the muscles or due to stimulation of the nerves (that may be both excitatory and inhibitory for contraction). In methodological preexperiments, pulse duration response, and voltage response curves were, therefore, produced to determine optimal stimulation variables and to identify the nerve-mediated stimulation (that is sensitive to Na+ channel block). For the pulse duration response study, EFS was applied at 18 V, 3 Hz in 5-s trains every 2 min, and the pulse duration was increased gradually from 0.01 to 5.0 ms. This was repeated in the presence of 0.3 µM of the fast neuronal sodium channel blocker tetrodotoxin. At 0.2-ms pulse duration, the responses were maximum, with negligible responses remaining in the presence of tetrodotoxin. Because no response was observed when voltage-sensitive Na+ channels were blocked, these responses were regarded as nerve mediated, and this stimulation duration was used throughout the study. For the voltage-response study, the EFS was applied at various voltages (5, 10, 18, 30 V) at 3 Hz in 5-s trains every 2 min. Pulse duration was increased gradually from 0.1 to 3.0 ms at each voltage. Responses to EFS with 0.2 ms and 18 V were regarded to be (supra)maximal and were used in this study. Thus, for the experiments reported here, EFS of 0.2-ms pulse duration, 3-Hz frequency, and 15-V amplitude (as measured in the tissue baths, 18 V at the output) were applied in 5-s trains every 2 min.

Effects of Relaxing Agents on EFS-Evoked Contraction

Ileal segments were exposed to EFS as described above for 30-60 min before agonists were added. The mean of the amplitudes of the stable responses to EFS (the last 4 before additions) for each segment was considered as the control value and set to 100%. Investigated agents (as indicated in the relevant figure legends) were added cumulatively to the tissue baths during the pause between contractions. The mean of the amplitudes of the two contractions after the addition of each drug concentration was measured and expressed in percentage of the control response. On the basis of these normalized segment responses, the mean values from all segments (3-4) from each mouse were calculated.

Effects of Relaxing Agents on Carbachol-Induced Contraction

Ileal segments were contracted submaximally with 1 µM of the acetylcholinesterase-resistent muscarinic receptor agonist carbachol. The contraction reached a stable level within 8 min and maintained a stable plateau for >30 min when not further affected (not shown). To investigate the relaxant action, specified agonists (as indicated in the figure legends) were added cumulatively during this plateau contraction to the organ baths with 4 min allowed for stabilization of the relaxation response to each concentration. At the end of each cumulative dose-response curve, the ileal segments were maximally relaxed with 15 µM of the nonspecific smooth muscle relaxant papaverine. Total carbachol-induced contraction was defined as the difference between the stable carbachol value and the value after papaverine and was set to 100% for each segment, and all responses were expressed as a percentage of this carbachol response. On the basis of these normalized segment responses, the mean values from all segments (3-4) from each mouse were calculated. After full relaxant response had been obtained, tissue was rinsed at 10-min intervals for <= 30 min, and a new series of responses was recorded in the same manner.

Data Analysis

Mean dose-response data obtained from each mouse as described above (mean of 3-4 segments) were analyzed for simple Michaelis-Menten kinetics with the curve-fit option of the KaleidaGraph application using the equation V(x) = 100% - Vmax · [x/(IC50 + x)] where x is the concentration of the substance investigated. The maximum volume (Vmax) and IC50 values thus generated for each mouse were used for statistical analysis with Student's unpaired t-test.

Chemicals

Isoproterenol hydrochloride (isoprenaline), papaverine hydrochloride, tetrodotoxin, carbamylcholine chloride (carbachol), and forskolin were purchased from Sigma (St. Louis, MO). CL-316,243 was a gift from E. Danforth. Isoprenaline was dissolved in 0.1 mM ascorbic acid. All other drugs were dissolved in distilled water, except forskolin, which was dissolved in DMSO. Drugs were added to the baths in volumes <1% of the total bath volume. The final concentration of DMSO itself (0.5%) showed no significant biological effects.


    RESULTS
TOP
ABSTRACT
INTRODUCTION
MATERIALS AND METHODS
RESULTS
DISCUSSION
REFERENCES

A UCP1-Dependent Component in Isoprenaline-Induced Intestinal Relaxation

EFS. In wild-type mice, EFS induced stable and reproducible contractions in segments of small intestine (Fig. 1A); according to our methodological preexperiments (see MATERIALS AND METHODS), these contractions represented the outcome of nervous stimulation of the segments. The general beta -adrenergic agonist isoprenaline dose dependently and reversibly inhibited these contractile responses to EFS; at high isoprenaline concentrations, the contractile responses were virtually abolished (Fig. 1A). Data for seven mice are compiled in Fig. 2A. As seen, isoprenaline was consistently able to fully inhibit the electrically induced contraction in wild-type mice: at 1 µM isoprenaline, only 2 ± 1% remained (mean value from 7 mice), and based on Michaelis-Menten analysis of data points, the remaining contraction at infinite isoprenaline concentration was -2 ± 2%, with an IC50 for isoprenaline of 23 ± 3 nM (mean ± SE from 7 animals, each individually analyzed as illustrated for the mean values in Fig. 2A).


View larger version (42K):
[in this window]
[in a new window]
 
Fig. 1.   Effect of the beta -adrenoceptor agonist isoprenaline on electrical field stimulation (EFS)-induced contractile responses of ileal segments from mitochondrial uncoupling protein-1 {UCP1; UCP1-containing [UCP1(+/+)]; A} and UCP1-ablated (-/-) (B) mice. Ileal segments were isolated in parallel from UCP1(+/+) and UCP1(-/-) mice, and their contractile activity was monitored as described in MATERIALS AND METHODS. Filled circles indicate the application of EFS. Isoprenaline was added cumulatively to the indicated concentrations (log M) to the organ bath. At the start of the "recovery," the organ bath was rinsed with fresh physiological solution.



View larger version (12K):
[in this window]
[in a new window]
 
Fig. 2.   Dose-response curves for isoprenaline-induced inhibition of EFS-induced contraction in ileal segments from UCP1(+/+) and UCP1(-/-) mice. Experiments were performed principally as those exemplified in Fig. 1. A: comparison between responses in C57Bl/6 UCP1(+/+) () and UCP1(-/-) (open circle ) mice. Points are means ± SE from 7 mice of each type. Dose-response curves were drawn for best fit to simple Michaelis-Menten kinetics; mean values from similar analyses of the response of each mouse are those given in the text. B: data points obtained from 2 129/SvPas mice (black-triangle or black-down-triangle ) and from 2 NMRI mice ( or black-diamond ); the dose-response curves shown are those from A for ileal segments from UCP1(+/+) (C57BL/6) and UCP1(-/-) (dashed line) mice. C: the UCP1-dependent part of the isoprenaline-induced inhibition of contraction. Points were obtained by subtracting the corresponding values in A and the curve by subtracting the 2 curves in A.

As illustrated in Fig. 1B, EFS could induce phasic contractions also in ileal segments from UCP1(-/-) mice; there were no observable differences in contractile amplitude or basal muscle tone between the segments from UCP1(+/+) and UCP1(-/-) mice (6.9 ± 0.9 mN per 1-cm segment in wild-type segments and 6.3 ± 0.7 mN per segment from UCP1(-/-) mice; means from 7 animals in each group). As seen, although isoprenaline was able to diminish the EFS-induced contractions dose dependently and reversibly also in the segments from UCP1(-/-) mice, it did not totally eliminate these contractions even at the highest concentrations shown; even a 10-fold higher concentration (10 µM) (in independent experiments) was not able to fully eliminate the contractions (not shown). Data from seven UCP1(-/-) mice examined in this way are compiled in Fig. 2A. As seen, at 1 µM isoprenaline there was still a significant contraction (15 ± 4%). Moreover, analysis of the dose-response curves for Michaelis-Menten kinetics indicated a clear residual, isoprenaline-insensitive component in the intestine from UCP1(-/-) mice: even at infinite isoprenaline concentration, 10 ± 4% (P < 0.02 vs. wild type; Student's unpaired t-test) of the contraction remained (cf. also the results in Figs. 3-5 for this residual contraction). Furthermore, the intestinal segments from UCP1(-/-) mice were more than threefold less sensitive to isoprenaline, demonstrating an IC50 of 81 ± 18 nM (vs. the 23 ± 3 nM in the wild type; P < 0.01).


View larger version (18K):
[in this window]
[in a new window]
 
Fig. 3.   Isoprenaline-induced relaxation of carbachol-induced contraction of ileal segments from UCP1(+/+) and UCP1(-/-) mice. Original traces obtained from UCP1(+/+) (C57BL/6) (A) and UCP1(-/-) (B) mice are shown. C: dose-response curves for isoprenaline-induced relaxation of carbachol-induced contraction in ileal segments from UCP1(+/+) and UCP1(-/-) mice. Values are means ± SE from 5 mice of each type. Curves were drawn for best fit to Michaelis-Menten kinetics, with the Hill coefficient set to 0.6.



View larger version (17K):
[in this window]
[in a new window]
 
Fig. 4.   Effect of the beta 3-adrenoceptor agonist CL-316,243 on EFS-induced (A) and carbachol-induced (B) contractile responses of ileal segments from UCP1(+/+) and UCP1(-/-) mice. A: EFS-induced contraction. Experiments were performed and results were analyzed as in Figs. 1 and 2. Points are means ± SE from 5 mice of each type. Dose-response curves were calculated as in Fig. 2. B: carbachol-induced contraction. Experiments were performed and results were analyzed as in Fig. 3. Points are means ± SE of 5 mice of each type. Dose-response curves were calculated as in Fig. 3.



View larger version (15K):
[in this window]
[in a new window]
 
Fig. 5.   Forskolin effects on EFS-induced contractile responses of ileal segments from UCP1(+/+) and UCP1(-/-) mice. Experiments were performed and results were analyzed as in Figs. 1 and 2. Points are means ± SE from 3 mice of each type. Dose-response curves were calculated as in Fig. 2.

It was of importance to confirm that UCP1(+/+) strains other than the C57Bl/6 behaved qualitatively and quantitatively similarly under the experimental conditions utilized. In Fig. 2B, we have, therefore, compiled data from other UCP1(+/+) mice strains and show them together with the curve fits from Fig. 2A. Data from the 129/SvPas strain (part of the genetic background of the UCP1(-/-) mouse), as well as from outbred NMRI mice, are shown. As seen, these other UCP1(+/+) strains behaved indistinguishably from the UCP1(+/+) C57Bl/6 mice used as wild type in Figs. 1 and 2A, both in demonstrating a high isoprenaline sensitivity and especially in reaching full isoprenaline-induced inhibition of EFS-induced contractions. There was thus no indication that the inability of isoprenaline to fully inhibit contraction in the intestines from the UCP1(-/-) mice, and the lower isoprenaline sensitivity observed in these intestines, was strainrelated; rather, these features seem to be consistent differences between UCP1(+/+) mice and UCP1(-/-) mice.

In Fig. 2C, we have plotted the difference between the response to isoprenaline in intestine from UCP1(+/+) and UCP1(-/-) mice. This difference may be considered to be the UCP1-dependent component in the isoprenaline-induced inhibition of intestinal contraction. As seen, this component reached maximally as much as 30% of the total response to isoprenaline (and of total relaxation) at ~0.1 µM isoprenaline, with an apparent IC50 in the low nanomolar range.

Carbachol-induced contraction. Stimulation of intestine from UCP1(+/+) mice with the stable acetylcholine analog carbachol led, as expected (19), to persistent intestinal contraction, as exemplified in Fig. 3A. Addition of increasing concentrations of isoprenaline led, again as expected (19), to full relaxation of the carbachol-induced contraction, to the extent that addition of the general relaxant papaverine did not elicit any further relaxation. As compiled in Fig. 3C, full relaxation (-4 ± 3%) was always reached at infinite isoprenaline concentrations, and isoprenaline had an IC50 of 27 ± 9 nM (n = 5), i.e., a value identical to its IC50 value for inhibition of EFS-stimulated contractions.

As exemplified in Fig. 3B, carbachol could also induce a stable contraction in ileal segments from UCP1-ablated mice, and there was no observable difference between the carbachol-induced contractile force in intestine from UCP1(+/+) and UCP1(-/-) mice (5.5 ± 0.5 mN per segment vs. 5.2 ± 0.3 mN; n = 6). However, isoprenaline was unable to fully relax intestine from UCP1(-/-) mice, whereas papaverine still induced full relaxation. From the compilation in Fig. 3C, it is seen that the residual relaxation was 7 ± 1% (P < 0.03 vs. wild type) even at infinite isoprenaline concentration, with an isoprenaline affinity (IC50) of 37 ± 9 nM. Thus a qualitative difference existed between intestine from UCP1(+/+) and UCP1(-/-) mice, irrespective of whether the contraction was induced by EFS (probably through release of endogenous acetylcholine from the nerves) or by added carbachol.

beta 3-Adrenergic Stimulation

Besides being prominent in brown and white adipose tissue (15), beta 3-adrenoceptors are also expressed in the intestine (11, 23, 66). Although beta 1- and beta 2-adrenoceptors are also expressed in the intestine (26, 54, 61), beta -adrenergically induced relaxation in the intestine is mainly mediated by the beta 3-adrenoceptors (23, 37). Similarly, the beta 3-receptors are also the beta -receptors mainly responsible for norepinephrine-induced thermogenesis (75, 76) and for norepinephrine-induced UCP1 gene expression (59) in brown adipocytes. We therefore also examined the influence of UCP1-ablation on beta 3-adrenoceptor-mediated relaxation.

The beta 3-specific agonist CL-316,243 was not fully as efficient as the general beta -agonist isoprenaline in inhibiting EFS-induced intestinal contraction; a residual response of 22 ± 1% remained in wild-type mice (Fig. 4A) even at infinite isoprenaline concentrations (mean from 5 mice). The affinity (IC50) for CL-316,243 was 19 ± 4 nM (Fig. 4A). When CL-316,243 was used to inhibit EFS-induced contraction in intestine from UCP1(-/-) mice, the residual contraction was significantly greater (41 ± 6%; P < 0.04) than in wild-type mice; the IC50 was 21 ± 6 nM (Fig. 4A). [We examined whether there was any effect of UCP1 ablation on the expression of the beta 3-receptors in the intestine, principally performed as in (1), but found no difference in expression (not shown)].

Similar observations were made when relaxation of carbachol-precontracted ileal segments was induced with CL-316,243 (Fig. 4B). Here the residual contraction was 16 ± 3% in intestine from wild-type mice [principally in agreement with earlier data (11, 23)], and as high as 35 ± 6% in intestine from UCP1(-/-) mice (P < 0.03; means ± SE from 5 mice); the IC50 was 33 ± 9 in the wild-type and 34 ± 8 in the UCP1(-/-) mice.

Thus the absence of UCP1 resulted in a similar degree of lack of relaxation potency, whether the relaxation was induced by a general beta -agonist or by a beta 3-selective agonist.

cAMP-Induced Relaxation Involves a UCP1-Dependent Component

To investigate whether the UCP1-dependent relaxation necessitated direct beta -receptor involvement, we examined whether such a UCP1-dependent component would also be visible if the relaxation was induced downstream from the receptor. beta -Adrenergically induced relaxation [and relaxation through other receptors (43, 53)] is further mediated by cyclic nucleotides (5) such as cGMP, but especially cAMP (68), and relaxation can, therefore, be induced by the adenylyl cyclase activator forskolin (27, 57, 61).

Thus, as expected, in wild-type mice, forskolin could inhibit EFS-induced contractions, and at infinite forskolin concentrations, these contractions were fully eliminated (-1 ± 3% remaining) (Fig. 5). In contrast, as seen in Fig. 5, in ileal segments from UCP1(-/-) mice, forskolin was only able to partly inhibit the EFS-induced contractions. Even when tested in independent experiments at 100 µM forskolin concentration, full inhibition was not obtained (not shown), and the Michaelis-Menten analysis indicates that even at infinite forskolin concentrations 11 ± 3% of the contractions would be remaining (P < 0.05; n = 3). Forskolin also had a nearly threefold lower apparent affinity in these segments [IC50 was 159 ± 32 nM in intestine from UCP1(+/+) mice and 393 ± 92 nM in intestine from UCP1(-/-) mice (P < 0.05, n = 3) (Fig. 5)].

It was therefore concluded that the difference between intestine from UCP1(+/+) and UCP1(-/-) mice was not due to an alteration in beta -receptor function but was located in the pathway between cAMP and muscle relaxation. Correspondingly, the presence of the UCP1-dependent relaxation was not dependent on the beta -receptor itself being stimulated; rather, any receptor, the function of which is mediated via cAMP, would be expected to demonstrate an UCP1-dependent component.


    DISCUSSION
TOP
ABSTRACT
INTRODUCTION
MATERIALS AND METHODS
RESULTS
DISCUSSION
REFERENCES

In the present investigation, we have demonstrated the existence of a UCP1-dependent component in the pathway for beta -adrenergically induced relaxation in intestine. Whereas the presence of this component may be an indirect consequence of the absence of UCP1 in brown adipose tissue, it is more likely that it is explainable by the absence of UCP1 in intestinal cells. Thus a UCP1-dependent mechanism involved in the transduction pathway from beta -adrenergic receptor stimulation and cAMP increases to intestinal relaxation may exist. A hypothesis for a functional role of UCP1 in contraction/relaxation control is presented below. In extension of these studies, it may be envisaged that the existence of a UCP1-dependent component in relaxation may not be restricted to adrenergic control of intestinal contraction but may be of significance for the understanding of the function of other organs apparently expressing UCP1 and exhibiting peristalsis.

beta -Adrenergically Induced Relaxation Versus Other Relaxing Agents

We have here focused on the significance of UCP1 for the mediation of beta -adrenergically induced relaxation in the ileal segments. As the effect was seen both when the segments were stimulated electrically and when they were directly stimulated with carbachol, it is likely that the effects observed are not localized to the nerves but rather to (some of) the smooth muscle cells themselves. Furthermore, because a UCP1-dependent component was also observed when we increased cAMP levels in these segments by direct stimulation of adenylyl cyclase (Fig. 5), the implication is that all intestinal relaxations mediated via an increase in cAMP, not only those induced by beta -adrenergic receptors (23, 36), may have a significant UCP1-dependent fraction, including relaxations induced by VIP (50) and PACAP (28, 43).

Functional Consequences of Absence of UCP1 in the Intestine

For evident reasons, all dedicated investigations of phenotypical alterations in UCP1(-/-) mice have, as yet, been related to thermogenesis and metabolism. Data presented here demonstrate that the absence of UCP1 may affect the function of the intestine. There are no prior formal reports on such phenotypes, but an indication we have had of possible gastrointestinal effects of a lack of UCP1 is an occasional observation of a phenomenon of rectal prolapse, occurring in a few rather old UCP1(-/-) mice; we have not seen a similar phenomenon in any wild-type mice. We have also observed that the postmortem relaxation of the intestines occurs measurably slower in intestine from UCP1(-/-) mice (not detailed), and we noted during the experiments that the force of the response to EFS was variable during a longer time in intestinal segments from UCP1(-/-) mice. The significance of alterations in intestinal relaxation under normal conditions may, however, be limited. It is generally assumed that the degree of sympathetic control of the gut is normally low, and is only activated in defense reactions, i.e., during stress (69), inflammation (45, 55) and e.g., in postoperative ileus (12, 29). Manifestations of the consequences of the absence of the UCP1-dependent part of relaxation may thus be obvious only under such conditions. It is also clear from our observations that the presence of UCP1 is not an absolute necessity for the functioning of the intestine, in contrast to the absolute necessity of UCP1 for competent brown adipose tissue function (17, 21, 46, 47, 49). This is in accordance with the fact that UCP1 is not found in the genome of any nonmammalian organism. Thus the presence of UCP1-dependent relaxation is apparently a specific and additional mammalian feature, the survival value of which is presently unknown.

It may be important to consider whether the existence of the UCP1-dependent adrenergically induced intestinal relaxation demonstrated here challenges the conclusions from earlier studies on the phenotype of the UCP1(-/-) mice. However, the conclusions of studies published to date (7, 10, 17, 21, 46-49) would not seem to be influenced by this new aspect: the reported effects are adequately understood based on a functional defect in brown adipose tissue alone. However, the situation may be different regarding the analysis of so-called brown-adipose-tissue-deficient animals (13, 18, 41, 44). In these transgenic animals, diphtheria toxin is expressed under the UCP1 promotor. Thus any cell that normally expresses UCP1 will also express diphtheria toxin and this toxin will kill the cell. This means that the intestinal cells expressing UCP1 will be eliminated, which may have broader consequences than the partial deterioration of their function reported here. Thus certain phenotypic features of the so-called brown adipose tissue-deficient mice may be ascribable to e.g., intestinal effects.

Mechanism of beta -Adrenergically Induced Intestitinal Relaxation

The signaling system leading from acetylcholine stimulation of muscarinic receptors to contraction of smooth muscle cells is fairly well understood (43, 53, 65). However, the mechanism for the relaxation pathway is still unclear (73). cAMP-induced phosphorylation of specific proteins, such as telokin (42, 70), has been discussed, but the further effect of these phosphorylations is also unknown. On the basis of the experiments reported here, together with what is known about UCP1 function in brown adipose tissue (reviewed in Refs. 16, 35, 51, 60), a first, hypothetical model for a novel pathway can be formulated (Fig. 6).


View larger version (37K):
[in this window]
[in a new window]
 
Fig. 6.   Possible functional role of UCP1 in mediation of relaxation in smooth muscle of organs with peristaltic motion. Acetylcholine (AcCh) stimulation of muscarinic receptors (M) leads to an increase in cytosolic Ca2+ levels that stimulates contraction; the contraction is dependent on ATP produced in the mitochondria. Norepinephrine (NE) binds to beta 3-adrenergic receptors (beta ), leading to adenylyl cyclase (AC) activation and thus to an increase in cytosolic cAMP levels that can result in activation of UCP1 in the mitochondrial membrane. This may result in mitochondrial uncoupling, i.e., a significant fraction of the proton gradient may be diverted from the ATP-synthase and instead pass through UCP1, resulting in lower ATP levels and thus possibly in a diminished response to muscarinic stimulation.

In the contraction process induced by acetylcholine binding to the muscarinic receptors and intracellularly mediated via an increase in cytosolic Ca2+ levels, ATP is used (31, 32, 65). This ATP is of mitochondrial origin. Adrenergic stimulation through beta -receptors can be expected to lead to an increase in cAMP levels, and this could subsequently lead to activation of UCP1. This would lead to mitochondrial uncoupling, i.e., some of the protons pumped out from the mitochondria by the respiratory chain would return to the mitochondrial matrix through UCP1, i.e., without passing through the ATP synthase, and less ATP would thus be formed. If this process is of sufficient magnitude, the ATP/ADP ratio in the cells will decrease, and less ATP will, therefore, be available for the contraction process. Thus a graded counteraction of induced contraction would be caused by adrenergic stimulation, although the Ca2+ level may be unaffected, i.e., a given level of Ca2+ may be less effective in inducing contraction, as has been found earlier (73). The fact that relaxation takes place, although not to the same extent, in the absence of UCP1, implies that other mechanisms are responsible for the major part of the relaxation process, but the UCP1-dependent relaxation would seem to have a higher sensitivity to beta -adrenergic stimulation/cAMP levels.

In brown adipocytes, the link between increased cAMP levels and the associated increase in protein kinase A activity (14) is associated with phosphorylation (activation) of hormone-sensitive lipase (63) and perilipin (6), leading to stimulated hydrolysis of triglycerides, and the released fatty acids, either in themselves or in some metabolite form, activate UCP1 (16, 35, 51, 60). Stores of triglyceride are apparently not observable in the longitudinal muscle layer of the intestine, and there are no reports that hormone-sensitive lipase or perilipin are expressed in this layer (although this may be due to the very small fraction of total tissue that constitutes the cells of interest). In the apparent absence of the established pathway for UCP1 activation, an interesting question as to the nature of the intracellular physiological activator of UCP1 in these cells arises. However, the mitochondria containing UCP1 are only a very small fraction of all mitochondria in intestinal tissue, and to examine the function of UCP1 in intestinal mitochondrial preparations is probably unfeasible, due to high dilution by mitochondria from other parts of the tissue.

Perspectives

The present studies were restricted to analyzing the effect of UCP1 ablation on relaxation of ileal segments. Evidently, the question may be raised as to whether similar mechanisms are involved in the relaxation of other types of smooth muscle. Because UCP1 is not present in the smooth muscle layers of blood vessels (52), the mechanism discussed here is not relevant in the control of vessel contractility and thus not for blood pressure in general. This distinction may be paralleled by the presence of distinct forms of myosin in vascular and peristaltic smooth muscle (30, 64).

However, the results presented here may be of interest concerning the function of other organs in which UCP1 has been indicated: stomach, eosophagus, ureter origin, epididymus, ductus deferens, and uterus. It is noteworthy that in most of these organs, a cAMP-mediated relaxation is seen [uterus (39, 56); vas deferens (27), urinary bladder (58), detrusor (57), distal colon (33), ileum (61), esophagus, and ureter (54)]. It is reasonable to assume that this relaxation could partly be UCP1-dependent, as demonstrated here for the ileal segments. Furthermore, it is also noteworthy that these organs all exhibit peristaltic motion. Regulated phasic contractile activity is coordinated, at least in the intestine, by the pacemaker cells, the intestinal cells of Cajal (reviewed in Refs. 62, 67), which are smooth muscle cells characterized e.g., by a high mitochondrial content (71). Similar pacemaker cells have at least been identified in the ureter (34) and discussed concerning the uterus (74). It is therefore not unlikely that UCP1 expression, cAMP-mediated relaxation, pacemaker cells, and peristalsis may be functionally linked in several organs, in addition to the intestine directly studied here.

Peristalsis, in addition to being required for the handling of food through the gastrointestinal system, is important in such functions as semen transfer, uterine function, and urine production. In the absence of dedicated observations and experiments, it may not be unexpected that no other phenotypic alterations have as yet been reported in UCP1(-/-) mice than those related to thermogenesis and now intestinal relaxation. However, the present observations may stimulate new types of dedicated studies of phenotypic manifestations of UCP1 ablation. Establishment of the significance of UCP1 in these contexts adds new challenges to the study of this unique mammalian protein.


    ACKNOWLEDGEMENTS

We thank Anette Ebberyd and Annelie Brolinson for technical help, Martin Nedergaard for animal observations, and Dana Hutchinson and L. P. Kozak for advice and contributions.


    FOOTNOTES

This study was supported by a grant from The Swedish Science Council.

Address for reprint requests and other correspondence: J. Nedergaard, Wenner-Gren Institute, Arrhenius Laboratories F3, Stockholm University, SE-106 91 Stockholm, Sweden (E-mail: jan{at}metabol.su.se).

The costs of publication of this article were defrayed in part by the payment of page charges. The article must therefore be hereby marked "advertisement" in accordance with 18 U.S.C. Section 1734 solely to indicate this fact.

August 7, 2002;10.1152/ajpgi.00193.2002

Received 21 May 2002; accepted in final form 30 July 2002.


    REFERENCES
TOP
ABSTRACT
INTRODUCTION
MATERIALS AND METHODS
RESULTS
DISCUSSION
REFERENCES

1.   Bengtsson, T, Redegren K, Strosberg AD, Nedergaard J, and Cannon B. Down-regulation of beta 3-adrenoreceptor gene expression in brown fat cells is transient and recovery is dependent upon a short-lived protein factor. J Biol Chem 271: 33366-33375, 1996[Abstract/Free Full Text].

2.   Bouillaud, F, Couplan E, Pecqueur C, and Ricquier D. Homologues of the uncoupling protein from brown adipose tissue (UCP1): UCP2, UCP3, BMCP1 and UCP4. Biochim Biophys Acta 1504: 107-119, 2001[ISI][Medline].

3.   Bouillaud, F, Ricquier D, Thibault J, and Weissenbach J. Molecular approach to thermogenesis in brown adipose tissue: cDNA cloning of the mitochondrial uncoupling protein. Proc Natl Acad Sci USA 82: 445-448, 1985[Abstract].

4.   Cannon, B, Matthias A, Golozoubova V, Ohlson KBE, Andersson U, Jacobsson A, and Nedergaard J. Unifying and distinguishing features of brown and white adipose tissues: UCP1 versus other UCPs. In: Progress in Obesity Research 8, edited by Ailhaud G, and Guy-Grand B.. London: Libbey, 1999, p. 13-26.

5.   Carvajal, JA, Germain AM, Huidobro-Toro JP, and Weiner CP. Molecular mechanism of cGMP-mediated smooth muscle relaxation. J Cell Physiol 184: 409-420, 2000[ISI][Medline].

6.   Chaudhry, A, and Granneman JG. Differential regulation of functional responses by beta -adrenergic receptor subtypes in brown adipocytes. Am J Physiol Regul Integr Comp Physiol 277: R147-R153, 1999[Abstract/Free Full Text].

7.   Commins, SP, Watson PM, Frampton IC, and Gettys TW. Leptin selectively reduces white adipose tissue in mice via a UCP1-dependent mechanism in brown adipose tissue. Am J Physiol Endocrinol Metab 280: E372-E373, 2001[Abstract/Free Full Text].

8.   Depocas, F. Chemical thermogenesis in the functionally eviscerated cold-acclimated rat. Can J Biochem Physiol 36: 691-699, 1958[ISI].

9.   Depocas, F. The calorigenic response of cold-acclimated white rats to infused noradrenaline. Can J Biochem Physiol 38: 107-114, 1960[ISI].

10.   Enerbäck, S, Jacobsson A, Simpson EM, Guerra C, Yamashita H, Harper ME, and Kozak LP. Mice lacking mitochondrial uncoupling protein are cold-sensitive but not obese. Nature 387: 90-94, 1997[ISI][Medline].

11.   Evans, BA, Papaioannou M, Anastasopoulos F, and Summers RJ. Differential regulation of beta 3-adrenoceptors in gut and adipose tissue of genetically obese (ob/ob) C57BL/6J-mice. Br J Pharmacol 124: 763-771, 1998[Abstract].

12.   Ferraz, AA, Wanderley GJ, Santos MA, Jr, Mathias CA, Araujo JG, Jr, and Ferraz EM. Effects of propranolol on human postoperative ileus. Dig Surg 18: 305-310, 2001[ISI][Medline].

13.   Frederich, RC, Hamann A, Anderson S, Lollmann B, Lowell BB, and Flier JS. Leptin levels reflect body lipid content in mice: evidence for diet-induced resistance to leptin action. Nat Med 1: 1311-1314, 1995[ISI][Medline].

14.   Fredriksson, JM, Thonberg H, Ohlson KBE, Ohba K, Cannon B, and Nedergaard J. Analysis of inhibition by H89 of UCP1 gene expression and thermogenesis indicates protein kinase A mediation of beta 3-adrenergic signaling rather than beta 3-adrenoceptor antagonism by H89. Biochim Biophys Acta 1538: 206-217, 2001[ISI][Medline].

15.   Galitsky, J, Carpéné C, Bousquet-Melou A, Berlan M, and Lafontan M. Differential activation of beta 1-, beta 2- and beta 3-adrenoceptors by catecholamines in white and brown adipocytes. Fundam Clin Pharmacol 9: 324-331, 1995[ISI][Medline].

16.   Garlid, KD, Jaburek M, Jezek P, and Varecha M. How do uncoupling proteins uncouple? Biochim Biophys Acta 1459: 383-389, 2000[ISI][Medline].

17.   Golozoubova, V, Hohtola E, Matthias A, Jacobsson A, Cannon B, and Nedergaard J. Only UCP1 can mediate adaptive nonshivering thermogenesis in the cold. FASEB J 15: 2048-2050, 2001[Abstract/Free Full Text].

18.   Hamann, A, Benecke H, Le Marchand-Brustel Y, Susulic VS, Lowell BB, and Flier JS. Characterization of insulin resistance and NIDDM in transgenic mice with reduced brown fat. Diabetes 44: 1266-1273, 1995[Abstract].

19.   Hardman, JG, Gilman AG, and Limbird LE. Goodman and Gilman's The Pharmacological Basis of Therapeutics (9th ed.). New York: McGraw-Hill, 1996.

20.   Heaton, GM, Wagenvoord RJ, Kemp JA, and Nicholls DG. Brown-adipose-tissue mitochondria: photoaffinity labelling of the regulatory site of energy dissipation. Eur J Biochem 82: 515-521, 1978[Abstract].

21.   Hofmann, WE, Liu X, Bearden CM, Harper ME, and Kozak LP. Effects of genetic background on thermoregulation and fatty acid-induced uncoupling of mitochondria in UCP1-deficient mice. J Biol Chem 276: 12460-12465, 2001[Abstract/Free Full Text].

22.   Horinouchi, T, and Koike K. Further characterization of beta 3-adrenoceptors in the guinea pig gastric fundus: stereoselectivity, beta -adrenoceptor alkylation, and structure-activity relationship. Can J Physiol Pharmacol 79: 985-995, 2001[ISI][Medline].

23.   Hutchinson, DS, Evans BA, and Summers RJ. beta 1-Adrenoceptors compensate for beta 3-adrenoceptors in ileum from beta 3-adrenoceptor knock-out mice. Br J Pharmacol 132: 433-442, 2001[Abstract/Free Full Text].

24.   Hutchinson, DS, Evans BA, and Summers RJ. beta 3-Adrenoceptor regulation and relaxation responses in mouse ileum. Br J Pharmacol 129: 1251-1259, 2000[Abstract/Free Full Text].

25.   Jacobsson, A, Stadler U, Glotzer MA, and Kozak LP. Mitochondrial uncoupling protein from mouse brown fat. Molecular cloning, genetic mapping, and mRNA expression. J Biol Chem 260: 16250-16254, 1985[Abstract/Free Full Text].

26.   Johnson, M. The beta -adrenoceptor. Am J Respir Crit Care Med 158: S146-S153, 1998[Abstract/Free Full Text].

27.   Kato, K, Furuya K, Tsutsui I, Ozaki T, and Yamagishi S. Cyclic AMP-mediated inhibition of noradrenaline-induced contraction and Ca2+ influx in guinea-pig vas deferens. Exp Physiol 85: 387-398, 2000[Abstract].

28.   Katsoulis, S, and Schmidt WE. Role of PACAP in the regulation of gastrointestinal motility. Ann NY Acad Sci 805: 364-378, 1996[ISI][Medline].

29.   Kehlet, H. Postoperative ileus. Gut 47, Suppl 4: iv85-iv86, 2000[Free Full Text]; discussion iv87.

30.   Kelley, CA, Takahashi M, Yu JH, and Adelstein RS. An insert of seven amino acids confers functional differences between smooth muscle myosins from the intestines and vasculature. J Biol Chem 268: 12848-12854, 1993[Abstract/Free Full Text].

31.   Khromov, A, Somlyo AV, and Somlyo AP. MgADP promotes a catch-like state developed through force-calcium hysteresis in tonic smooth muscle. Biophys J 75: 1926-1934, 1998[Abstract/Free Full Text].

32.   Khromov, AS, Somlyo AP, and Somlyo AV. Photolytic release of MgADP reduces rigor force in smooth muscle. Biophys J 80: 1905-1914, 2001[Abstract/Free Full Text].

33.   Kishi, M, Takeuchi T, Katayama H, Yamazaki Y, Nishio H, Hata F, and Takewaki T. Involvement of cyclic AMP-PKA pathway in VIP-induced, charybdotoxin-sensitive relaxation of longitudinal muscle of the distal colon of Wistar-ST rats. Br J Pharmacol 129: 140-146, 2000[Abstract/Free Full Text].

34.   Klemm, MF, Exintaris B, and Lang RJ. Identification of the cells underlying pacemaker activity in the guinea-pig upper urinary tract. J Physiol 519: 867-884, 1999[Abstract/Free Full Text].

35.   Klingenberg, M, and Echtay KS. Uncoupling proteins: the issues from a biochemist point of view. Biochim Biophys Acta 1504: 128-143, 2001[ISI][Medline].

36.   Koike, K, Horinouchi T, and Takayanagi I. Signal transduction pathway involved in beta 3-adrenoceptor-mediated relaxation in guinea pig taenia caecum. Jpn J Pharmacol 68: 41-46, 1995[ISI][Medline].

37.   Koike, K, and Takayanagi I. Beta3-adrenoceptor mechanisms in guinea-pig taenia caecum. Life Sci 62: 1503-1507, 1998[ISI][Medline].

38.   Kuemmerle, JF, Murthy KS, and Makhlouf GM. Longitudinal smooth muscle of the mammalian intestine. A model for Ca2+ signaling by cADPR. Cell Biochem Biophys 28: 31-44, 1998[Medline].

39.   Kuhn, R, Uckert S, Stief CG, Truss MC, Lietz B, Bischoff E, Schramm M, and Jonas U. Relaxation of human ureteral smooth muscle in vitro by modulation of cyclic nucleotide-dependent pathways. Urol Res 28: 110-115, 2000[ISI][Medline].

40.   Lin, CS, and Klingenberg M. Isolation of the uncoupling protein from brown adipose tissue mitochondria. FEBS Lett 113: 299-303, 1980[ISI][Medline].

41.   Lowell, BB, Susulic VS, Hamann A, Lawitts JA, Himms-Hagen J, Boyer BB, Kozak LP, and Flier JS. Development of obesity in transgenic mice after genetic ablation of brown adipose tissue. Nature 366: 740-742, 1993[ISI][Medline].

42.   MacDonald, JA, Walker LA, Nakamoto RK, Gorenne I, Somlyo AV, Somlyo AP, and Haystead TA. Phosphorylation of telokin by cyclic nucleotide kinases and the identification of in vivo phosphorylation sites in smooth muscle. FEBS Lett 479: 83-88, 2000[ISI][Medline].

43.   Makhlouf, GM, and Murthy KS. Signal transduction in gastrointestinal smooth muscle. Cell Signal 9: 269-276, 1997[ISI][Medline].

44.   Mantzoros, CS, Frederich RC, Qu D, Lowell BB, Maratos-Flier E, and Flier JS. Severe leptin resistance in brown fat-deficient uncoupling protein promoter-driven diphtheria toxin A mice despite suppression of hypothalamic neuropeptide Y and circulating corticosterone concentrations. Diabetes 47: 230-238, 1998[Abstract].

45.   Martinolle, JP, Garcia-Villar R, More J, and Bueno L. Evidence for mast cell, leukotriene and nitric oxide involvement in the regulation of the adrenoceptor number of inflamed small intestine in guinea pigs. Neurogastroenterol Motil 7: 187-195, 1995[ISI][Medline].

46.   Matthias, A, Jacobsson A, Cannon B, and Nedergaard J. The bioenergetics of brown fat mitochondria from UCP1-ablated mice. UCP1 is not involved in fatty acid-induced de-energization. J Biol Chem 274: 28150-28160, 1999[Abstract/Free Full Text].

47.   Matthias, A, Ohlson KEB, Fredriksson JM, Jacobsson A, Nedergaard J, and Cannon B. Thermogenic responses in brown-fat cells are fully UCP1-dependent: UCP2 or UCP3 do not substitute for UCP1 in adrenergically or fatty-acid induced thermogenesis. J Biol Chem 275: 25073-25081, 2000[Abstract/Free Full Text].

48.   Monemdjou, S, Hofmann WE, Kozak LP, and Harper ME. Increased mitochondrial proton leak in skeletal muscle mitochondria of UCP1-deficient mice. Am J Physiol Endocrinol Metab 279: E941-E946, 2000[Abstract/Free Full Text].

49.   Monemdjou, S, Kozak LP, and Harper ME. Mitochondrial proton leak in brown adipose tissue mitochondria of Ucp1-deficient mice is GDP insensitive. Am J Physiol Endocrinol Metab 276: E1073-E1082, 1999[Abstract/Free Full Text].

50.   Murthy, KS, Grider JR, Jin JG, and Makhlouf GM. Interplay of VIP and nitric oxide in the regulation of neuromuscular function in the gut. Ann NY Acad Sci 805: 355-362, 1996[ISI][Medline].

51.   Nedergaard, J, Golozoubova V, Matthias A, Asadi A, Jacobsson A, and Cannon B. UCP1: the only protein able to mediate adaptive non-shivering thermogenesis and metabolic inefficiency. Biochim Biophys Acta 1504: 82-106, 2001[ISI][Medline].

52.   Nibbelink, M, Moulin K, Arnaud E, Duval C, Penicaud L, and Casteilla L. Brown fat UCP1 is specifically expressed in uterine longitudinal smooth muscle cells. J Biol Chem 276: 47291-47295, 2001[Abstract/Free Full Text].

53.   Olsson, C, and Holmgren S. The control of gut motility. Comp Biochem Physiol A Mol Integr Physiol 128: 481-503, 2001[ISI][Medline].

54.   Oostendorp, J, Preitner F, Moffat J, Jimenez M, Giacobino JP, Molenaar P, and Kaumann AJ. Contribution of beta -adrenoceptor subtypes to relaxation of colon and oesophagus and pacemaker activity of ureter in wild-type and beta 3-adrenoceptor knockout mice. Br J Pharmacol 130: 747-758, 2000[Abstract/Free Full Text].

55.   Pol, O, Valle L, Ferrer I, and Puig MM. The inhibitory effects of alpha(2)-adrenoceptor agonists on gastrointestinal transit during croton oil-induced intestinal inflammation. Br J Pharmacol 119: 1649-1655, 1996[Abstract].

56.   Price, SA, and Bernal AL. Uterine quiescence: the role of cyclic AMP. Exp Physiol 86: 265-272, 2001[Abstract].

57.   Qiu, Y, Kraft P, Craig EC, Liu X, and Haynes-Johnson D. Cyclic nucleotide phosphodiesterases in rabbit detrusor smooth muscle. Urology 59: 145-149, 2002[ISI][Medline].

58.   Qiu, Y, Kraft P, Craig EC, Liu X, and Haynes-Johnson D. Identification and functional study of phosphodiesterases in rat urinary bladder. Urol Res 29: 388-392, 2001[ISI][Medline].

59.   Rehnmark, S, Néchad M, Herron D, Cannon B, and Nedergaard J. alpha - and beta -Adrenergic induction of the expression of the uncoupling protein thermogenin in brown adipocytes differentiated in culture. J Biol Chem 265: 16464-16471, 1990[Abstract/Free Full Text].

60.   Rial, E, and Gonzalez-Barroso MM. Physiological regulation of the transport activity in the uncoupling proteins UCP1 and UCP2. Biochim Biophys Acta 1504: 70-81, 2001[ISI][Medline].

61.   Roberts, SJ, Papaioannou M, Evans BA, and Summers RJ. Characterization of beta -adrenoceptor mediated smooth muscle relaxation and the detection of mRNA for beta 1-, beta 2- and beta 3-adrenoceptors in rat ileum. Br J Pharmacol 127: 949-961, 1999[Abstract/Free Full Text].

62.   Sanders, KM, Ordog T, Koh SD, and Ward SM. A novel pacemaker mechanism drives gastrointestinal rhythmicity. News Physiol Sci 15: 291-298, 2000[Abstract/Free Full Text].

63.   Shih, MF, and Taberner PV. Selective activation of brown adipocyte hormone-sensitive lipase and cAMP production in the mouse by beta 3-adrenoceptor agonists. Biochem Pharmacol 50: 601-608, 1995[ISI][Medline].

64.   Somlyo, AP. Myosin isoforms in smooth muscle: how may they affect function and structure? J Muscle Res Cell Motil 14: 557-563, 1993[ISI][Medline].

65.   Somlyo, AP, and Somlyo AV. Signal transduction and regulation in smooth muscle. Nature 372: 231-236, 1994[ISI][Medline].

66.   Strosberg, AD, and Pietri-Rouxel F. Function and regulation of the beta 3-adrenoceptor. Trends Pharmacol Sci 17: 373-381, 1996[ISI][Medline].

67.   Thomsen, L, Robinson TL, Lee JC, Farraway LA, Hughes MJ, Andrews DW, and Huizinga JD. Interstitial cells of Cajal generate a rhythmic pacemaker current. Nat Med 4: 848-851, 1998[ISI][Medline].

68.   Torphy, TJ. beta -Adrenoceptors, cAMP and airway smooth muscle relaxation: challenges to the dogma. Trends Pharmacol Sci 15: 370-374, 1994[ISI][Medline].

69.   Tsukada, F, Sugawara M, Sawamura K, Ohuchi Y, Kohno H, and Ohkubo Y. Beta3-adrenoceptor is involved in the inhibition of small intestinal motility due to restraint stress in rats. Biol Pharm Bull 24: 995-997, 2001[ISI][Medline].

70.   Walker, LA, MacDonald JA, Liu X, Nakamoto RK, Haystead TA, Somlyo AV, and Somlyo AP. Site-specific phosphorylation and point mutations of telokin modulate its Ca2+-desensitizing effect in smooth muscle. J Biol Chem 276: 24519-24524, 2001[Abstract/Free Full Text].

71.   Ward, SM, Ordog T, Koh SD, Baker SA, Jun JY, Amberg G, Monaghan K, and Sanders KM. Pacemaking in interstitial cells of Cajal depends upon calcium handling by endoplasmic reticulum and mitochondria. J Physiol 525: 355-361, 2000[Abstract/Free Full Text].

72.   Wiklund, CU, Lim S, Lindsten U, and Lindahl SG. Relaxation by sevoflurane, desflurane and halothane in the isolated guinea-pig trachea via inhibition of cholinergic neurotransmission. Br J Anaesth 83: 422-429, 1999[Abstract/Free Full Text].

73.   Woodrum, DA, and Brophy CM. The paradox of smooth muscle physiology. Mol Cell Endocrinol 177: 135-143, 2001[ISI][Medline].

74.   Wray, S, Kupittayanant S, Shmygol A, Smith RD, and Burdyga T. The physiological basis of uterine contractility: a short review. Exp Physiol 86: 239-246, 2001[Abstract].

75.   Zhao, J, Cannon B, and Nedergaard J. alpha 1-Adrenergic stimulation potentiates the thermogenic action of beta 3-adrenoceptor-generated cAMP in brown fat cells. J Biol Chem 272: 32847-32856, 1997[Abstract/Free Full Text].

76.   Zhao, J, Cannon B, and Nedergaard J. Thermogenesis is beta 3- but not beta 1-adrenergically mediated in rat brown fat cells, even after cold acclimation. Am J Physiol Regul Integr Comp Physiol 275: R2002-R2011, 1998[Abstract/Free Full Text].


Am J Physiol Gastrointest Liver Physiol 283(5):G1107-G1116
0193-1857/02 $5.00 Copyright © 2002 the American Physiological Society