Background

The alpha2-adrenergic agonist dexmedetomidine alters global cerebral blood flow (CBF). However, few studies have investigated the action of dexmedetomidine on the cerebral microcirculation. This investigation examined the effects of dexmedetomidine on (1) regional CBF in the rat cerebral cortex using laser-Doppler flowmetry and (2) on pial arteriolar diameter.

Methods

Halothane-anesthetized rats were fitted with instruments to measure CBF as determined by laser-Doppler flow (CBFldf) or to measure pial arteriolar diameter by preparing a cranial hollow deepened until a translucent plate of skull remained, thereby maintaining the integrity of the cranial vault. In both groups, 20 microg/kg dexmedetomidine was infused intravenously. Thirty minutes later, the mean arterial pressure was restored to control values with an infusion of phenylephrine (0.5 to 5 microg/kg/min).

Results

Administration of dexmedetomidine was associated with decreases in end-tidal and arterial carbon dioxide. The CBFldf and pial arteriolar diameter were measured during normocapnia (controlled carbon dioxide) and during dexmedetomidine-induced hypocapnia. Intravenous administration of dexmedetomidine significantly decreased systemic arterial pressure concurrent with a decrease in CBFldf (22% in normocapnic animals, 36% in hypocapnic animals). Restoration of mean arterial pressure increased CBFldf in normocapnic but not in hypocapnic animals. Similarly, dexmedetomidine significantly reduced pial vessel diameter in both normocapnic (9%) and hypocapnic animals (17%). However, vessel diameters remained decreased in the normocapnic and hypocapnic animals after the mean arterial pressure was restored.

Conclusions

These results suggest a modulation of cerebral vascular autoregulation by dexmedetomidine which may be mediated, in part, by alterations in carbon dioxide. Dexmedetomidine may have a direct action on the cerebral vessels to reduce the CBF during normo- or hypocapnia. The differences between CBFldf and pial arteriole responses to restoration of mean arterial pressure may reflect the difference in measurement techniques because laser-Doppler measurements reflect the net effect of several arterial segments on microvascular perfusion, whereas diameter measurements specifically examined individual pial arterioles, suggesting that dexmedetomidine vasoconstriction in the cerebral vasculature may be differentially and regionally mediated.

DEXMEDETOMIDINE, a highly selective alpha2-adrenergic agonist, possesses potentially useful perianesthetic properties, including sedation, analgesia, anxiolysis, muscle relaxation, hemodynamic stability, and a reduction in the mean alveolar concentration of inhalational anesthetics, [1–6]all mediated through specific presynaptic and postsynaptic alpha2-adrenergic receptors within the central nervous system. [1–3,5–7]Other alpha2-adrenergic agonists decrease the cerebral blood flow (CBF) in humans and animals. [8–10]Studies suggest that dexmedetomidine may produce significant reductions in CBF without specifically influencing the cerebral metabolic rate for oxygen. [11–13]Dexmedetomidine-induced reductions in CBF have also been observed in the presence of changes in the pressures of arterial carbon dioxide and oxygen. [14,15]Most previous studies have examined the action of dexmedetomidine on global CBF. Studies have suggested that certain volatile anesthetics may differentially effect global and regional CBF. [16]Thus it may be critically important to distinguish between specific regional erythrocyte flow and global CBF.

Laser-Doppler flowmetry (CBFldf) measures CBF to highly localized regions of the brain, [17–19]in a minimally invasive manner, and continuously monitors microvascular perfusion of tissues. Pial arteriolar diameter changes also may be valid indicators of local cerebral perfusion responses. [20,21] 

Previous in vitro studies from this laboratory [22]showed that dexmedetomidine vasoconstricted canine middle cerebral artery vessels. In addition, recent in vivo canine studies showed a significant dexmedetomidine-induced constriction of pial vessels. [23]However, studies have not fully compared the effects of dexmedetomidine on regional CBF, autoregulation, and vessel diameter in the cerebral microcirculation. The purpose of the present investigation was to identify the specific actions of dexmedetomidine on (1) autoregulation of regional CBF in the rat cerebral cortex using laser-Doppler flowmetry and on (2) pial arteriolar diameter by direct microscopic observation through a thinned cranium. These studies were performed during alterations in systemic arterial pressure (SAP) and also during normocapnia and dexmedetomidine-induced hypocapnia. We hypothesized that dexmedetomidine would similarly alter pial vessel diameter and CBFldfin anesthetized rats.

Animal Preparation

All studies were approved by the Animal Use and Care Committee of the Medical College of Wisconsin and were in accordance with National Institutes of Health guidelines (Guide for the Care and Use of Laboratory Animals, DHEW [DHHS], NIH publication no. 85–23, revised 1985).

Anesthesia was induced in 33 adult, male Sprague-Dawley rats (weighing 250–450 g) by an intraperitoneal injection of pentobarbital (65 mg/kg; Sigma Chemical Co., St. Louis, MO). After a midline ventral neck dissection, the trachea was isolated and cannulated with polyethylene tubing (PE 240, Intramedic[registered sign], Becton Dickinson, Parsippany, NJ). The femoral artery and vein were cannulated with polyethylene catheters (PE 50, Intramedic[registered sign]) filled with heparinized saline to transduce SAP, maintain fluids, and administer drugs, respectively. Intermittent positive-pressure ventilation with 70% nitrogen and 30% oxygen was instituted (model 707; Harvard Apparatus, South Natick, MA). The inspired gases were mixed using a calibrated 3-tube flowmeter (Linde; Union Carbide, Somerset, NJ). Anesthesia was maintained with 1 mean alveolar concentration halothane. Gallamine (80 mg/kg given intraperitoneally; Sigma Chemical) was used to maintain muscle paralysis. Inspired and expired oxygen, carbon dioxide, and volatile anesthetic concentrations were measured continuously (POET II; TM Criticare Systems, Milwaukee, WI). Arterial blood gas tensions were intermittently determined using a blood gas analyzer (Radiometer ABL3; Copenhagen, Denmark). Body temperature was continuously monitored using a rectal thermistor and maintained at 37 +/- 0.2 [degree sign] Celsius with a water-circulated heating pad (model 73 ATA; YSI, Yellow Springs, OH). Cranial/pericranial temperature was not monitored, but the cranial vault remained intact without breaching the transcranial bone or the dura in all experiments. End-tidal carbon dioxide (FETCO2), mean systemic arterial blood pressure (MAP), systolic SAP, diastolic SAP, cerebral blood flow (CBFldf), and the concentration of anesthetic vapor were displayed continuously and recorded on an eight-channel polygraph (Astro-Med, West Warwick, RI).

The rats were positioned prone and their heads secured in a stereotaxic apparatus (model 900; David Kopf, Tujunga, CA). After removal of a 2-cm2area of scalp and connective tissue, a cranial hollow, 2 mm in diameter, was drilled over the right parietal cerebral cortex, approximately 3 mm posterior to the bregma and 3 mm to the right of the midline. The drilling was done with the aid of a stereomicroscope (Stereo Zoom 5; Cambridge Instruments, Division of Leica, Inc., Deerfield, IL) using a low-speed (0–8,000 rpm) air drill (model Rhino XP; Midwest Dental, Des Plaines, IL), and the surface was simultaneously cooled by continuous air flow through a suction tip. The cranial hollow was deepened until a translucent plate of skull remained, as described previously by Hudetz et al. [18] 

Measurement of Cerebral Blood Flow by Laser-Doppler Flow (Groups I sub A and I sub B)

In 18 rats, microvascular perfusion was monitored throughout the study using a laser-Doppler flowmeter (PF3; Perimed, Stockholm, Sweden) and flowprobe (PF 316 dental probe; Perimed) with a tip diameter of 1 mm. Using a micromanipulator, the probe was lowered and positioned within the burr hole. Mineral oil was applied to improve optical coupling between the probe tip and brain tissue. Care was taken to avoid positioning the probe over large, visible vessels, which were also easily identified by extraordinarily high flow values. Once established, the probe position was not altered for the duration of the experimental protocol.

All animals were allowed to stabilize for 1 h during 1 mean alveolar concentration halothane administration and normocapnia (FETCO2, 34–37 mmHg). After baseline control measurements of CBFLDF, MAP, SAP, diastolic SAP, and FETCO2, dexmedetomidine (20 micro gram/kg; Farmos Group, Turku, Finland), dissolved in 1 ml normal saline, was infused intravenously during a 15-min period. Subsequently, a 15-min period was allowed for equilibration after dexmedetomidine infusion and measurements were repeated. The MAP was restored to control values in the next 20–25 min using a titrated infusion of phenylephrine (0.5–5 micro gram [center dot] kg sup -1 [center dot] min sup -1; Sigma Chemical Co., St. Louis, MO), and all parameters were again measured at 50% and 100% restoration of MAP. Experiments were successfully completed in 12 rats (groups IAand IB). Preliminary experiments suggested that during dexmedetomidine infusion there was a decrease in the FETCO22 values. Consequently, in eight animals, the ventilation was continuously adjusted to maintain normocapnia (group I sub A, n = 8), whereas in four animals, no adjustments in ventilation were made, and thus the FETCO2was allowed to vary (group I sub B, n = 4).

Measurement of Pial Arteriolar Diameter (Groups II sub A and II sub B)

Fifteen rats were used to examine the effects of dexmedetomidine on the cerebral microvasculature by direct microscopic observation through a thinned cranium. A cranial burr hole was made to a maximum depth but without completely penetrating the skull. The cranial hollow was cleaned and dried by vacuum, filled with a drop of mineral oil, and sealed with a quartz cover slip (Fisher Scientific, Pittsburgh, PA). Pial arteries were visualized using a dark-field metallurgical microscope (model BHMJ; Olympus, Tokyo, Japan) equipped with a 50-W halogen lamp and a vertical epi-illuminator. The incident light was filtered using an infrared filter to minimize heating of the tissue. A 20x ultralong working distance, infinity-corrected lens (ULWD Neo SPlan 20, 0.40; Olympus) was used with a 2.5x TV projection lens. Pial arteries were differentiated from pial veins by lighter color, differences in the branching pattern, and the presence of pulsatile flow. The microscopic image was monitored using a CCD video camera (model VDC-2524; Sanyo, Osaka, Japan), which was continuously displayed and video recorded (model HR-D780V; JVC, Yokohama, Japan).

The experimental protocol was similar to that used in groups IAand IB. Briefly, control video recordings of the microvessels were made. Dexmedetomidine was infused and recording of the microvessels was repeated, phenylephrine administered, and recordings again made. Experiments were successfully completed in ten rats (groups IIAand IIB). In a manner similar to the CBFldfflow experiments of groups IAand IB, a decrease in FETCO2was observed with infusions of dexmedetomidine. Consequently, FETCO2was controlled within normocapnic limits in six rats (group II sub A, n = 6), whereas in four rats (group IIB, n = 4) ventilation was not adjusted.

Using a JVC professional video recorder (model BR-S525U) the recorded video images were examined, selected at specified intervals, and grabbed using an image capturer board (Spectrum NTSC; Redlake Corp., Morgan Hill, CA). Hard copies of selected video images were made using the video graphics printer (UP-860; Sony Corp., Teaneck, NJ). Vessel diameter was measured for the digitized image contrast-enhanced images using Image I software (National Institutes of Health, Bethesda, MD). In each image, seven to eight lines were drawn perpendicular to the length of the vessel and across its opposite edges, and these distances were averaged and calibrated to give an average vessel diameter.

Dexmedetomidine, End-tidal Gas, and Arterial Gas Tensions (Group III)

To confirm that changes in expired end-tidal gas concentrations reflected systemic arterial gas tensions, seven additional adult, male Sprague-Dawley rats (group III) were prepared in a manner similar to that of groups I and II. The animals were anesthetized, ventilated, fitted with monitoring instruments, and positioned in an identical manner. The scalp was not incised and burr holes were not made. The experimental protocol was identical to that of groups IBand IIB. A 0.25 to 0.5 ml sample of arterial blood was analyzed for pH and partial pressures of carbon dioxide and oxygen during control conditions, at the conclusion of dexmedetomidine infusion (15 min), 15 min after completion of dexmedetomidine infusion (30 min), and after restoration (100%) of SAP with phenylephrine.

Statistical Analysis

All data are expressed as means +/- SEM. All CBFldfvalues were normalized to percentages of control. Statistical analysis of data within and between groups during control conditions and during dexmedetomidine infusion, after infusion, and during phenylephrine administration was performed with a repeated measures analysis of variance and Duncan's modification of the t test, using the SAS procedure GLM software (SAS Institute, Cary, NC). Changes from control values were considered significant when the probability value was <0.05.

Dexmedetomidine and Laser-Doppler Flow Determination of Cerebral Blood Flow

During control conditions with 1 mean alveolar concentration halothane equilibration and normocapnia, baseline MAP was 102 +/- 3 mmHg and CBFldfwas 156 +/- 6 perfusion units in group IA(normocapnia)(Table 1, Figure 1). Values were similar in group I sub B (hypocapnia) with MAP and CBFldfof 109 +/- 3 mmHg and 163 +/- 10 perfusion units, respectively (Table 2, Figure 1). Subsequent to initiating the dexmedetomidine infusion, a transient, nonsignificant increase in MAP was observed in most animals. This was followed by a progressive and significant decrease in MAP in animals in groups IAand IB. Thirty minutes after initiating dexmedetomidine infusion, MAP decreased by 32% in group IArats (102 +/- 3 to 69 +/- 6 mmHg) and by 36% in group IBrats (109 +/- 3 to 70 +/- 2 mmHg). During the dexmedetomidine infusion, a significant reduction in FETCO2was observed in group IBrats (30 +/- 2 mmHg 30 min after dexmedetomidine infusion compared with 35 +/- 0 in control), whereas in group IAthe ventilation was adjusted to keep the FETCO2within normocapnic limits.

Table 1. Effects of Dexmedetomidine during Normocapnia (Group IA, N = 8) 

Table 1. Effects of Dexmedetomidine during Normocapnia (Group IA, N = 8) 
Table 1. Effects of Dexmedetomidine during Normocapnia (Group IA, N = 8) 

Figure 1. The effect of dexmedetomidine (20 micro gram/kg) on laser-Doppler flow during normocapnia and hypocapnia and after restoration of systemic arterial pressure with phenylephrine. % refers to 50% or 100% restoration of control mean arterial pressure. *Significantly different (P < 0.05) compared with control values.

Figure 1. The effect of dexmedetomidine (20 micro gram/kg) on laser-Doppler flow during normocapnia and hypocapnia and after restoration of systemic arterial pressure with phenylephrine. % refers to 50% or 100% restoration of control mean arterial pressure. *Significantly different (P < 0.05) compared with control values.

Close modal

Table 2. Effects of Dexmedetomidine during Hypocapnia (Group IB, N = 4) 

Table 2. Effects of Dexmedetomidine during Hypocapnia (Group IB, N = 4) 
Table 2. Effects of Dexmedetomidine during Hypocapnia (Group IB, N = 4) 

After starting the infusion of dexmedetomidine, significant decreases in CBFldfvalues were observed in group IAand IBanimals. The decreases were not significantly different among the normocapnic groups and hypocapnic groups. Thirty minutes after dexmedetomidine infusion, the decrease in CBFldfwas 22% in group I sub A rats (156 +/- 6 to 121 +/- 12 perfusion units) and 36% in group I sub B rats (163 +/- 10 to 104 +/- 10 perfusion units). When MAP was restored with phenylephrine infusion, the CBFldfreturned to control values in group IArats (normocapnic; 156 +/- 14 perfusion units compared with 156 +/- 6 at control) but remained significantly decreased in group IBrats (hypocapnic; 127 +/- 9 perfusion units compared with 163 +/- 10 at control).

Dexmedetomidine and Pial Artery Diameter

The MAP at control was 105 mmHg in normocapnic animals (group IIA) and hypocapnic animals (group IIB;Table 3and Table 4, respectively). Baseline vessel diameter was 59 +/- 4 micro meter in group IIAanimals, whereas in group IIBanimals the vessel diameter was 72 +/- 7 micro meter. Figure 2shows the vessel diameters (as percentages of baseline control). There were no differences in vessel diameter (raw or normalized data) between groups during control or at any time during the experimental protocol. Similar to the observation in group I animals, the infusion of dexmedetomidine resulted in a significant decrease in MAP. Thirty minutes after starting dexmedetomidine infusion, the percentage decrease in MAP was 40% in group IIA(105 +/- 1 to 63 +/- 6 mmHg) and 40%(105 +/- 3 to 63 +/- 2 mmHg) in group IIB. In addition, a significant decrease in FET sub CO2was observed during dexmedetomidine infusion in the group IIBrats (30 +/- 1 mmHg 30 min after dexmedetomidine infusion compared with 35 +/- 0 mmHg at control), whereas the FETCO2was maintained at control levels in the group IIAanimals.

Table 3. Effects of Dexmedetomidine during Normocapnia (Group IIA, N = 6) 

Table 3. Effects of Dexmedetomidine during Normocapnia (Group IIA, N = 6) 
Table 3. Effects of Dexmedetomidine during Normocapnia (Group IIA, N = 6) 

Table 4. Effects of Dexmedetomidine during Hypocapnia (Group IIB, N = 4) 

Table 4. Effects of Dexmedetomidine during Hypocapnia (Group IIB, N = 4) 
Table 4. Effects of Dexmedetomidine during Hypocapnia (Group IIB, N = 4) 

Figure 2. The effect of dexmedetomidine (20 micro gram/kg) on pial arteriolar diameter during normocapnia and hypocapnia and after restoration of systemic arterial pressure with phenylephrine. % refers to 50% or 100% restoration of control mean arterial pressure. *Significantly different (P < 0.05) compared with control values.

Figure 2. The effect of dexmedetomidine (20 micro gram/kg) on pial arteriolar diameter during normocapnia and hypocapnia and after restoration of systemic arterial pressure with phenylephrine. % refers to 50% or 100% restoration of control mean arterial pressure. *Significantly different (P < 0.05) compared with control values.

Close modal

With dexmedetomidine infusion, a significant decrease in vessel diameter was observed in both group IIAand IIBrats. Within 15 min of dexmedetomidine infusion, there was a significant decrease in vessel diameter in both the normocapnic group (53 +/- 4 compared with 59 +/- 4; 10% of control) and the hypocapnic group (61 +/- 10 compared with 72 +/- 7; 17% of control;Figure 3). Thirty minutes after dexmedetomidine infusion, the vessel diameter remained significantly decreased by 16% of control in group IIBrats (62 +/- 12 compared with 72 +/- 7 micro meter at control; P < 0.05) but not in group IIArats (56 +/- 5 compared with 59 +/- 4 micro meter at control; P = not significant). When MAP was restored to control values with phenylephrine infusion, the significant decreases in vessel diameter persisted in group IIB(hypocapnic) animals (60 +/- 11 compared with 72 +/- 7 micro meter at control) and reappeared in group IIA(normocapnic) animals (53 +/- 4 compared with 59 +/- 4 micro meter at control).

Figure 3. The effect of dexmedetomidine on pial arteriolar diameter during control, immediately after dexmedetomidine (20 micro gram/kg) infusion, 30 min after infusion (post), and after restoration of systemic arterial pressure (phenylephrine). The measured diameter of the vessel is labeled in white. Note the decrease in diameter after dexmedetomidine infusion.

Figure 3. The effect of dexmedetomidine on pial arteriolar diameter during control, immediately after dexmedetomidine (20 micro gram/kg) infusion, 30 min after infusion (post), and after restoration of systemic arterial pressure (phenylephrine). The measured diameter of the vessel is labeled in white. Note the decrease in diameter after dexmedetomidine infusion.

Close modal

Dexmedetomidine, End-tidal Gas, and Arterial Gas Tensions

(Table 5) summarizes the effects of dexmedetomidine on end-tidal carbon dioxide concentrations and concurrent systemic arterial pH, carbon dioxide, and oxygen concentrations. As in groups IBand IIB, dexmedetomidine reduced end-tidal carbon dioxide concentrations (36 +/- 0 to 31 +/- 1 mmHg; control compared with 30 min after dexmedetomidine; P < 0.05) with a concurrent decrease in the arterial partial pressure of carbon dioxide (36 +/- 0 to 30 +/- 2 mmHg; control compared with 30 min after dexmedetomidine; P < 0.05) without significantly altering pH or arterial oxygen partial pressures.

Table 5. Effects of Dexmedetomidine on Arterial Blood Gas Tensions (Group III, N = 7) 

Table 5. Effects of Dexmedetomidine on Arterial Blood Gas Tensions (Group III, N = 7) 
Table 5. Effects of Dexmedetomidine on Arterial Blood Gas Tensions (Group III, N = 7) 

This study examined the specific effects of dexmedetomidine on regional CBF in the rat cerebral cortex using laser-Doppler flowmetry and the effects on pial arterial diameter. In the present investigation, dexmedetomidine produced significant decreases in CBFldfand in pial vessel diameter. The slow intravenous infusion of dexmedetomidine that we used in these experiments transiently increased then progressively and significantly reduced SAP, which is characteristic of alpha2-adrenergic agonists. Restoration of SAP by phenylephrine returned CBFldfto near control values in normocapnic rats but not in hypocapnic rats. In contrast, after restoration of MAP to control values, pial vessel diameter remained significantly decreased in both normocapnic and hypocapnic animals.

The alpha2-adrenergic agonists, including clonidine and dexmedetomidine, produce multiple actions within the central nervous system and reduce the dose requirements for anesthetics and narcotics. [1–6,24]Initially a transient increase in SAP occurs due to peripheral vascular alpha2-adrenoceptor-mediated vasoconstriction. [25,26]These agents alter systemic hemodynamics through a central sympatholytic and parasympathomimetic action via specific alpha2-adrenergic receptors at bulbar vasomotor centers, with contributions from supracollicular centers. [7,27,28]This central nervous system action decreases arterial pressure which, in the present investigation, was reversed with phenylephrine, which has no intrinsic effects on CBF. [29] 

Alpha2-Adrenergic receptors are widely distributed within the cerebral vasculature, [30]and specific cerebral vasoconstrictive responses produced by activation of these receptors has been proposed. [31]Clonidine may significantly reduce CBF in both humans and animals [9–12]by stimulating postsynaptic vascular alpha sub 2 -adrenergic receptors. Previous studies have suggested that dexmedetomidine may also reduce CBF, in both isoflurane- and halothane-anesthetized dogs, without specifically influencing the metabolic rate for oxygen. [11–13]This suggests that dexmedetomidine acts on specific brain regions with dense populations of alpha2-receptors to directly vasoconstrict rather than indirectly vasoconstrict secondary to cerebral metabolic depressant effects. However, in vitro studies have shown an alpha2-adrenergic receptor-mediated vasodilatation of rat middle cerebral arteries that involved [32]nitric oxide and a pertussis toxin-sensitive G protein as mediators. Thus vascular actions, altered sympathetic nerve or regional cerebral activity, and vessel specific actions all may influence the responses seen to alpha2-adrenergic agonists.

The commonly used cerebral venous outflow technique to measure CBF requires extensive surgical preparation with possible cerebrovascular compromise. [11,12]Laser-Doppler flow determination of CBF is a sensitive and reliable method to measure regional CBF with a fast response time. To place the CBFldfprobe, most studies use a cranial window technique. [17,20]However, the present studies used a minimally invasive model to monitor perfusion in the rat brain cortex without requiring craniotomy. [18]The CBFldfprobe was applied over a small area of thinned cranium, without invading the tissues to be analyzed. The remaining cranium constitutes an effective barrier for gas diffusion, helps isolate the cerebral tissues from the atmosphere, minimizes or eliminates cerebral hyperemia, maintains local cerebral temperature, and preserves the intracranial pressure.

It has been suggested that CBF is maintained at nearly constant values despite wide variations in MAP by adjustments in cerebral vascular resistance. [33]Autoregulation of CBFldfhas been observed between MAPs of 60–140 mmHg. [19]A linear relation between CBFldfand FETCO2levels has also been demonstrated. [17,18]Previous studies with volatile anesthetics, including halothane, have demonstrated dose-related increases in CBFldfin rats. [34]While persisted, it was attenuated at low concentrations of halothane. Therefore, the use of halothane as the baseline anesthetic, and of pentobarbital as the initial anesthetic, may represent a potential limitation of the present investigation and may have affected the results obtained after dexmedetomidine administration, possibly by attenuating vascular responsiveness.

The finding that dexmedetomidine produced significant decreases in CBFldfdespite an initial transient increase in MAP is consistent with previous studies in which a decrease in CBF was observed with dexmedetomidine with either no change or an increase in associated MAP. [11,12]This suggests that dexmedetomidine may have a direct action on the cerebral vessels to reduce the CBF. The alterations in the CBFldfseen with decreases and increases in MAP, in the autoregulatory range of 60–140 mmHg, suggest that dexmedetomidine might disrupt the autoregulatory mechanisms of the cerebral circulation. As expected, the decrease in CBFldfwas more pronounced in the presence of hypocapnia. The cause of the hypocapnic response produced by dexmedetomidine, in animals in which ventilation was not adjusted, may represent an increase in the depth of anesthesia in these animals already anesthetized with only 1 mean alveolar concentration of halothane. [1–3]Previous studies have shown a species-specific decrease in FETCO2after administration of dexmedetomidine. [4,25,35] 

Various methods have been used in the past to study diameter changes in cerebral microcirculation. Simultaneous measurements of pial diameter and CBFldfhave also been performed. [17,20]Most previous studies describe the closed cranial window preparation for visualizing the cerebral cortex. [21]Although this preparation preserves the cerebral microenvironment, the pial vessels may be injured when bone and dura are lifted. To obviate this, we measured microvessel diameter without breaking the integrity of the cranium. Similar to the measurement of CBFldf, the pial diameters were also measured through the thinned cranial bone of rats with a fairly minimal surgical procedure. To our knowledge, this is the first time cerebrovascular behavior has been studied by direct microscopy through the thinned intact cranial bone of adult rats.

Potential limitations of the current technique include a possible erroneous simultaneous measurement of cortical, dural, and cranial bone flow rather than purely cortical microvascular flow and assessment of a single flow point rather than flow distribution under the cranial hollow. Extreme care was taken in each experiment to visualize cranial bone blood vessels during the microdissection of the cranial hollow. Vessels were encountered during drilling but not near the bottom when pial vessels were clearly visible. The bone vessels and pial vessels have well-distinguishable structures and, therefore, cannot be mistaken. In addition, the density of dural vessels and their blood flow is low. Furthermore, the photon migration path distribution in brain tissue is banana shaped between the laser emitting and receiving fibers and, therefore, the measurement should be more sensitive to subsurface vessels. Only a single point measurement of cortical flow was obtained, and because the probe was always positioned at the site with the lowest reading, a higher flow would have been present at the other sites beneath the cranial hollow, although we did not do a systematic search in this investigation. An additional limitation of the present technique is the difficulty in performing topical pharmacologic studies, through a burr hole with an intact cranium in contrast to open preparations.

Studies evaluating the responsiveness of the cerebral vasculature to adrenergic-related agents have produced conflicting results. Norepinephrine has been shown to constrict pial vessels by stimulating alpha2-adrenoceptors in pigs, [36]but it also may differentially affect the cerebral arteries, with constriction in rostral regions and dilatation in caudal regions of the bovine brain, resulting in an altered blood flow distribution. [37]Norepinephrine has also been shown to have no effect on cerebral arterioles under baseline conditions. [38] 

Clonidine may exert cerebrovascular constrictive effects in vivo [39]and in vitro, [37]probably mediated by its actions on cerebral arterial alpha2-receptors. However, clonidine also may inhibit neurosympathetic cerebroarterial constriction by activating prejunctional alpha2-adrenoceptors. [40]A direct cerebral vasoconstrictor action has been proposed for dexmedetomidine due to its ability to decrease CBF. [11–13]Coughlan et al. [22]showed dexmedetomidine-induced direct vasoconstriction of canine middle cerebral arteries in vitro. Recently, Ishiyama et al. [23]showed that topical application of dexmedetomidine constricted pial arteries and veins. Although the vasoconstrictive effects of dexmedetomidine appear to be mediated by activating alpha2-adrenoceptors, opposing vasodilatory effects may also occur when adenosine triphosphate-sensitive K sup + channels areactivated. [23]Similar to its effect on the CBF, the cerebrovascularconstrictive effects of dexmedetomidine do not appear to be metabolically mediated. It is also probably not influenced by nitric oxide release because the nitric oxide synthase inhibitor L-NAME did not attenuate dexmedetomidine-induced constriction of cerebral arteries. [22,23] 

In the present study, we found that the infusion of dexmedetomidine initially caused cerebral vasoconstriction despite reductions in MAP. This suggests that dexmedetomidine directly vasoconstricts the pial vessels, similar to the observed in vitro effects. [22]Presumably, dexmedetomidine might affect the autoregulation of pial arterioles. Although the initial vasoconstriction after dexmedetomidine infusion was followed by vasodilatation as the MAP decreased further, the vessel diameters did not return to control values during either normocapnia or hypocapnia. Restoration of MAP then restored CBFldfto control levels during controlled normocapnia, but not during hypocapnia, after dexmedetomidine.

These results are consistent with a direct vasoconstrictor effect of dexmedetomidine overriding the cerebrovascular pressure autoregulation. The differences in the response patterns between CBFldfand pial vessel responses to restoration of MAP may reflect the difference in measurement techniques or the specific vascular segments whose responses are assessed. Changes in CBF are believed to be accompanied by adjustments in the cerebral arteriolar diameter. Haberi et al. [17]found a linear relation between CBFldfresponses and arteriolar diameter responses to vasoconstrictor and vasodilator stimuli but suggest that diameter measurements, which are highly localized, may not precisely reflect events in other parts of the arterial tree. On the other hand, Ngai et al. [20]found that pial arteriolar diameter changes parallel CBFldfresponses during sciatic nerve stimulation and suggest that arteriolar diameter changes can be considered valid indicators of local cerebral perfusion responses. However, it is still unclear whether surface vessel diameter changes may quantitatively predict flow responses in the cerebral cortex. Although flow within an arteriole has been linked proportionally to its internal diameter, [41]the distribution of cerebrovascular resistance may not be in a steady state in the vascular network. The region studied and the magnitude and direction of response of parenchymal vessels monitored by CBFldfmay be different than that of pial arterioles.

In conclusion, this investigation shows the ability of dexmedetomidine to significantly affect the cerebromicrovascular system. During spontaneous hypocapnia and controlled normocapnia, dexmedetomidine decreased SAP, CBFldf, and pial arteriolar diameter. Restoration of SAP did not reverse the dexmedetomidine-induced decrease in pial arteriolar diameter, but CBFldfmeasurements of CBF did return to control levels during normocapnia but not during hypocapnia. Further investigations are needed to fully delineate the differential actions of dexmedetomidine on various vascular segments of the cerebral circulation.

The authors thank Ines Krolo for performing supplementary experiments, David Schwabe for providing technical assistance, and Angela M. Barnes for preparing the manuscript.

1.
Segal IS, Vickery RG, Walton JK, Doze VA, Maze M: Dexmedetomidine diminishes halothane anesthetic requirement in rats through a postsynaptic alpha sub 2 adrenergic receptor. Anesthesiology 1988; 69:818-23.
2.
Jaakola ML, Salonen M, Lehtinen R, Scheinin H: The analgesic actions of dexmedetomidine-a novel alpha-2 adrenoceptor agonist-in healthy volunteers. Pain 1991; 46:281-5.
3.
Doze VA, Chen BX, Maze M: Dexmedetomidine produces a hypnotic-anesthetic action in rats via activation of central alpha sub 2 adrenoceptors. Anesthesiology 1989; 71:75-9.
4.
Zornow MH: Ventilatory, hemodynamic and sedative effects of the alpha 2 adrenergic agonist, dexmedetomidine. Neuropharmacology 1991; 30:1065-71.
5.
Proctor LT, Schmeling WT, Warltier DC: Premedication with oral dexmedetomidine alters hemodynamic actions of intravenous anesthetic agents in chronically instrumented dogs. Anesthesiology 1992; 77:554-62.
6.
Bloor BC, Schmeling WT: Cardiovascular effects of alpha sub 2 -adrenoceptors. Anaesth Pharmacol Rev 1993; 1:246-62.
7.
Unnerstall JR, Kopajtic TA, Kuhar MJ: Distribution of alpha-2 agonist binding sites in the rat and human central nervous system: analysis of some functional, anatomic correlates of the pharmacologic effects of clonidine and related adrenergic agents. Brain Res Rev 1984; 7:69-101.
8.
James IM, Larbi E, Zaimis E: The effect of the acute intravenous administration of clonidine on cerebral blood flow in man. Br J Pharmacol 1970; 39:198P-9P.
9.
Greene CS, Cretler DD, Cervenka K, McCoy CE, Brown FD, Murphy MB: Cerebral blood flow during the acute therapy of severe hypertension with oral clonidine. Am J Emerg Med 1990; 8:293-6.
10.
Kanawati IS, Yaksh TL, Anderson RE, Marsh RW: Effects of clonidine on cerebral blood flow and the response to arterial CO sub 2. J Cereb Blood Flow Metab 1986; 6:358-65.
11.
Karlsson BR, Forsman M, Roald OK, Heier MS, Steen PA: Effect of dexmedetomidine, a selective and potent alpha-2 agonist, or cerebral blood flow and oxygen consumption during halothane anesthesia in dogs. Anesth Analg 1990; 71:125-9.
12.
Zornow MH, Fleischer JE, Scheller MS, Nakakinmura K, Drummond JC: Dexmedetomidine, and alpha-2-adrenergic agonist, decreases cerebral blood flow in the isoflurane-anesthetized dog. Anesth Analg 1990; 70:623-30.
13.
McPherson RW, Kirsch JR, Traystman RJ: Inhibition of nitric oxide synthetase does not affect alpha 2-adrenergic-mediated cerebral vasoconstriction. Anesth Analg 1994; 78:67-72.
14.
McPherson RW, Koehler RC, Traystman RJ: Hypoxia, alpha-2-adrenergic, and nitric oxide-dependent interactions on canine cerebral blood flow. Am J Physiol 1994; 226:H476-H82.
15.
Fale A, Kirsch JR, McPherson RW: alpha sub 2 -adrenergic agonist effects on normocapnic and hypercapnic cerebral blood flow in the dog are anesthetic dependent. Anesth Analg 1994; 79:892-8.
16.
Hansen TD, Warner DS, Todd MM: Distribution of cerebral blood flow during halothane versus isoflurane anesthesia in rats. Anesthesiology 1988; 69:332-7.
17.
Haberl RL, Heizer ML, Marmarou A, Ellis EF: Laser-Doppler assessment of brain microcirculation: Effect of systemic alterations. Am J Physiol 1989; 256:H1247-H54.
18.
Hudetz AG, Roman RJ, Harder DR: Spontaneous flow occillations in the cerebral cortex during acute changes in mean arterial pressure. J Cereb Blood Flow Metab 1992; 12:491-9.
19.
Florence G, Seylaz J: Rapid autoregulation of cerebral blood flow: A laser-doppler flowmetry study. J Cereb Blood Flow Metab 1992; 12:674-80.
20.
Ngai AC, Meno JR, Winn HR: Simultaneous measurements of pial arteriolar diameter and laser-doppler flow during somatosensory stimulation. J Cereb Blood Flow Metabol 1995; 15:124-7.
21.
Morii S, Ngai AC, Winn HR: Reactivity of rat pial arterioles and venules to adenosine and carbon dioxide: With detailed description of the closed cranial window technique in rats. J Cereb Blood Flow Metab 1986; 6:34-41.
22.
Coughlan MG, Lee JG, Bosnjak ZJ, Schmeling WT, Kampine JP, Warltier DC: Direct coronary and cerebral vascular responses to dexmedetomidine. Anesthesiology 1992; 77:998-1006.
23.
Ishiyama T, Dohi S, Iida H, Watanabe Y, Shimonaka H: Mechanisms of dexmedetomidine-induced cerebrovascular effects in canine in vivo experiments. Anesth Analg 1995; 81:1208-15.
24.
Flacke JW, Bloor BC, Flacke WE, Wong D, Dazza S, Stead SW, Laus H: Reduced narcotic requirement by clonidine with improved hemodynamic and adrenergic stability in patients undergoing coronary bypass surgery. Anesthesiology 1987; 67:11-9.
25.
Schmeling WT, Kampine JP, Roerig DL, Warltier DC: The effects of the stereoisomers of the alpha sub 2 -adrenergic agonist medetomidine on systemic and coronary hemodynamics in conscious dogs. Anesthesiology 1991; 75:499-511.
26.
Smet G, Hoobler SW, Sanbar S, Julius S: Clinical observations on a new antihypertensive drug, 2-(2,6-dichlorphenylamine-2-imidazoline hydrochloride). Am Heart J 1969; 77:473-8.
27.
Schmitt H, Schmitt H: Localization of the hypotensive effect of 2-(2,6,-dichlorphenylamino)-2-imidazoline hydrochloride (St-155, Catapresan). Eur J Pharmacol 1969; 6:8-12.
28.
Tadepalli AS, Mills E: Contribution of supracollicular structures of the brain to the cerebral depression of cardiovascular function by clonidine. J Pharmacol Exp Ther 1978; 205:693-701.
29.
Edvinsson L, Lacombe P, Owman CH, Reynier-Rebuffel AM, Seylaz J: Quantitative changes in regional cerebral blood flow of rats induced by alpha- and beta adrenergic stimulants. Acta Physiol Scand 1979; 107:289-96.
30.
Skarby T: Pharmacological properties of prejunctional alpha-adrenoceptors in isolated feline middle cerebral arteries: comparison with the postjunctional alpha-adrenoceptors. Acta Physiol Scand 1984; 122:165-74.
31.
Edvinsson L, Mackenzie ET, Robert JP, Skarby T, Young AR: Cerebrovascular responses to haemorrhagic hypotension in anaesthetized cats: Effects of alpha-adrenoceptor antagonists. Acta Physiol Scand 1985; 123:317-23.
32.
Bryan RM, Eichler MY, Swafford MWG, Johnson TD, Suresh MS, Childres WF: Stimulation of alpha sub 2 -adrenoceptors dilates the rat middle cerebral artery. Anesthesiology 1996; 85:82-90.
33.
Hernandez MJ, Brennan RW, Bowman GS: Cerebral blood flow autoregulation in the rat. Stroke 1978; 9:150-5.
34.
Lee JG, Hudetz AG, Smith JJ, Hillard CJ, Bosnjak ZJ, Kampine JP: The effects of halothane and isoflurane on cerebrocortical microcirculation and autoregulation as assessed by laser doppler flowmetry. Anesth Analg 1994; 79:58-65.
35.
Ward DS: Respiratory effects of alpha sub 2 -adrenoceptor agonists. Anaesth Pharmacol Rev 1993; 1:263-7.
36.
Busija DW, Leffler CW: Exogenous norepinephrine constricts cerebral arterioles via alpha sub 2 -adrenoceptors in newborn pigs. J Cereb Blood Metab 1987; 7:184-8.
37.
Ayajiki K, Toda N: Isolated bovine cerebral arteries from rostral and caudal regions: Distinct responses to adrenoceptor agonists. Eur J Pharmacol 1990; 191:417-25.
38.
Bauknight Jr. GC, Faraci FM, Heistad DD: Endothelium-derived relaxing factor modulates noradrenergic constriction of cerebral arterioles in rabbits. Stroke 1992; 23:1522-5.
39.
Busija DW, Leffler CW: Post junctional alpha 2-adrenerceptors in pial arteries of anesthetized newborn pigs. Dev Pharmacol Ther 1987; 10:36-46.
40.
Reichl R, Walland A: Inhibition of neurosympathetic cerebroarterial constriction by clonidine in cats. Eur J Pharmacol 1980; 68:349-57.
41.
Mayrovitz HN, Roy J: Microvascular blood flow: Evidence indicating a cubic dependence on arteriolar diameter. Am J Physiol 1983; 243:H1031-H8.