Anda di halaman 1dari 5

Protective Effects of Hypercapnic Acidosis on

Ventilator-induced Lung Injury


ALAIN F. BROCCARD, JOHN R. HOTCHKISS, CHRISTINE VANNAY, MICHELE MARKERT, ALAIN SAUTY,
FRANÇOIS FEIHL, and MARIE-DENISE SCHALLER
Division of Intensive Care, Department of Internal Medicine and Central Laboratory for Clinical Chemistry, University Hospital (CHUV),
Lausanne, Switzerland

To investigate whether respiratory acidosis modulates ventilator- (5–8). Second, surfactant production or function may be mod-
induced lung injury (VILI), we perfused (constant flow) 21 isolated ulated by acid–base status (9–11). Third, hypercapnia appears
sets of normal rabbit lungs, ventilated them for 20 min (pressure to downregulate the endogenous production of nitric oxide
controlled ventilation [PCV]  15 cm H2O) (Baseline) with an in- (NO) in the lung (12), an effect of potential relevance, consid-
spired CO2 fraction adjusted for the partial pressure of CO2 in the ering the dual cytoxoxiccytoprotective potential of NO (13).
perfusate (PCO2  40 mm Hg), and then randomized them into Additionally, respiratory acidosis protects rabbit lungs ex vivo
three groups. Group A (control: n  7) was ventilated with PCV  and in vivo against ischemia–reperfusion injury (14, 15). It is
15 cm H2O for three consecutive 20-min periods (T1, T2, T3). In
unknown whether such protection would extend to VILI.
Group B (high PCVnormocapnia; n  7), PCV was given at 20
With these considerations in mind, we designed the present
(T1), 25 (T2), and 30 (T3) cm H2O. The targeted PCO2 was 40 mm
study to investigate the possible impact of hypercapnic acido-
Hg in Groups A and B. Group C (high PCVhypercapnia; n  7) was
ventilated in the same way as Group B, but the targeted PCO2 was
sis on the development of VILI in an isolated, perfused rabbit
 70 to 100 mm Hg. The changes (from Baseline to T3) in weight lung preparation.
gain (WG: g) and in the ultrafiltration coefficient (Kf  grmin
cm H2O100g) and the protein and hemoglobin concentrations in METHODS
bronchoalveolar lavage fluid (BALF) were used to assess injury. Care, techniques, and procedures used in the study were approved by
Group B experienced a significantly greater WG (14.85  5.49 the animal care committee of our institution.
[mean  SEM] g) and Kf (1.40  0.49 gmincm H2O100 g) than
did either Group A (WG  0.70  0.43; Kf  0.01  0.03) or Isolated Perfused Lung Preparation
Group C (WG  5.27  2.03 g; Kf  0.25  0.12 gmincm H2O We used a previously described, isolated, perfused lung preparation
100 g). BALF protein and hemoglobin concentrations (gL) were (New Zealand rabbits) (16, 17). No inhibitor of cyclooxygenase was
higher in Group B (11.98  3.78 gL and 1.82  0.40 gL, respec- added to the perfusate. Perfusion was set at a constant flow of 300 ml
tively) than in Group A (2.92  0.75 gL and 0.38  0.15 gL) or min and left atrial pressure (Pla) at 6 mm Hg (referenced to the top of
Group C (5.71  1.88 gL and 1.19  0.32 gL). We conclude that the lung). Initial ventilation (Veolar ventilator; Hamilton Medical,
respiratory acidosis decreases the severity of VILI in this model. Rhazüns, Switzerland) was set in the pressure controlled mode (pressure
controlled ventilation [PCV]), with a positive end-expiratory pressure
Keywords: respiratory acidosis; hypercapnia; mechanical ventilation; (PEEP) of 3 cm H2O, a respiratory rate (RR) of 20 breathsmin, and an
acute lung injury; rabbits inspiratory time fraction of 0.33. The ventilator was connected to two
gas tanks: the first tank (O2: 40%; CO2: 5%; N2: 55%) was connected to
Recent clinical trials have indicated that ventilatory strategies the air inlet and the second (O2: 40%; CO2: 25%; N2: 35%) to the oxygen
aimed at limiting ventilator-induced lung injury (VILI) may inlet. Thus, adjustment of the ventilator FIO2 knob allowed titration of
have a positive impact on the outcome of the acute respiratory the inspired CO2 fraction (FIO2) to the desired perfusate PCO2.
distress syndrome (1, 2). These strategies impose limits on tidal
volume (VT) and inflation pressure, and may result in hyper- Protocol
capnic acidosis. Until recently, the latter condition was mainly The experimental protocol consisted of four successive 20-min peri-
viewed as a side effect to be either tolerated (permissive hyper- ods. During the first period (Baseline), lungs were ventilated at 15 cm
capnia) or treated by increasing the rate of mechanical breath- H2O peak airway pressure (Paw) with normocapnia (perfusate PCO2 
ing or by giving a bicarbonate infusion, or with techniques such 40  5 [mean  SEM] mm Hg). Subsequently, the preparations were
randomized into three groups and studied for three additional periods
as tracheal gas insufflation or extracorporeal CO2 removal (3).
(T1, T2, and T3). In the control group (C, n  7), peak Paw and perfu-
However, hypercapnia and acidosis have myriads of effects sate PCO2 were kept at the Baseline values. In the high-pressure nor-
at the cellular level, several of which might influence the de- mocapnic group (HPNC, n  7), peak Paw was raised in increments of
velopment of VILI. First, it is now believed that mechanical 5 cm H2O at the beginning of each new period to a maximum value of
stretch induces lung injury in part by activating various intra- 30 cm H2O during T3. The targeted arterial CO2 tension (PaCO2 and
cellular signaling cascades (4), some of them having demon- pH were the same as in the C group. In the high-pressure hypercapnic
strated a dependence on pH or partial pressure of CO2 (PCO2) group (HPHC), n  7], peak Paw was increased as in the HPNC
group, but the targeted PaCO2 was between 70 and 100 mm Hg.

Measurements
(Received in original form July 13, 2000 and in revised form May 2, 2001)
Supported by the Central Hospitalier Universitarium Vandais. Dr. Hotchkiss was Paw, flow, VT, pulmonary artery pressure (Ppa), Pla, and pulmonary
supported by an American Heart Association Scientist Development Grant, Re- capillary pressure (Pcp) were measured as previously described (16).
gions Hospital, St. Paul, Minnesota, and the Centre Hospitalier Universitaire Vau- Three blood gas analyses were performed during each period. At Base-
dois, Lausanne, Switzerland. line and T3, plasma nitrite  nitrate (NOx) concentrations were as-
Correspondence and requests for reprints should be addressed to A. Broccard, M.D., sayed (18, 19). Edema formation (weight gain [WG] from the beginning
Division des Soins Intensifs, Départment de Médecine. BH10-982, University Hospital to the end of each period) was used to calculate changes in WG from
(CHUV), CH-1011 Lausanne, Switzerland. E-mail: alain.broccard@chuv.hospvd.ch Baseline (WG at T1,T2,T3 as WG[Tx]  WG[Baseline]). The ultrafil-
Am J Respir Crit Care Med Vol 164. pp 802–806, 2001 tration coefficient (Kf) was determined at the end of each period (16),
Internet address: www.atsjournals.org and changes in Kf from Baseline (Kf at T1,T2,T3) were calculated as
Broccard, Hotchkiss, Vannay, et al.: Lung Injury and Mechanical Ventilation 803

TABLE 1. CHARACTERISTICS OF THE GROUPS AT BASELINE

Control High-Pressure High-Pressure


Normocapnic Normocapnic Hypercapnic
Variables at Baseline Group (C) Group (HPNC) Group (HPHC)

Rabbit weight, kg 2.8  0.1 3.1  0.1 2.9  0.2


Lung weight, gr 16  1 16  1 16  1
Ischemic time, min 23  1 23  2 23  2
Perfusate hematocrit, % 6.1  0.3 6.3  0.2 6.3  0.3
Pla, mm Hg 6.6  0.1 6.5  0.2 6.8  0.2
Ppa, mm Hg 12.7  0.7 11.5  1.2 12.9  1.0
Pcp, mm Hg 9.1  0.6 8.7  0.5 8.4  0.4
Pulmonary vascular resistance, dynes  sec1  cm5 1,284  173 1,219  326 1,298  284
Lung compliance, ml/cm H2O 4.3  0.2 4.5  0.4 5.1  0.5
PaO2, mm Hg 233  4 220  6 228  6
PaCO2, mm Hg 42  5 36  6 35  5
pH 7.26  0.03 7.35  0.05 7.32  0.05
W, g 1.35  0.3 0.55  0.2 1.37  0.7
Kf, g/min/cm H2O/100g 0.18  0.03* 0.11  0.02 0.08  0.02

Definition of abbreviations: W  weight gain; Kf  ultrafiltration coefficient; PaCO2  arterial carbon dioxide tension; PaO2  arterial ox-
ygen tension; Pcp  pulmonary capillary pressure; Pla  left atrial pressure; Ppa  pulmonary artery pressure.
* p  0.04 Control group versus HPHC group.

Kf (Tx)  Kf (Baseline) (17). When these other measurements were Statistics


completed, recovered fluid from a bronchoalveolar lavage (BAL) of
the left lung was divided into two aliquots to measure the BAL hemo- The mean values of variables measured only once were compared
globin level (HbBAL)(20), and, after centrifugation, the supernatant among groups with the Kruskal–Wallis analysis of variance (ANOVA)
protein (bicinchonic acid assay, [BCA]; Pierce, Rockford, IL) and NOx on ranks; the Student–Newman–Keuls’ adjustment was performed for
concentrations (18, 19). NOx was used to track lung NO production. multiple comparisons. Variables for which one measurement was
The lung wet weight (WWL)dry weight (DWL) ratio was measured with made in each experimental period were analyzed with a repeated
lung tissue pieces sampled from each of the right lung lobes. measures ANOVA. Correlations were examined through linear re-

TABLE 2. CHARACTERISTICS OF STUDY GROUPS AFTER RANDOMIZATION AT THE BEGINNING OF EACH


PERIOD OF VENTILATION

Control High-Pressure High-Pressure


Normocapnic Normocapnic Hypercapnic
Time Variables Group Group Group

T1 Peak alveolar pressure, cm H2O 15.1  0.3 20.2  0.2* 20.7  0.2*
Total PEEP, cm H2O 2.4  0.2 2.4  0.2 2.7  0.2
VT, ml 52  4 75  13* 73  8*
Pla, mm Hg 6.9  0.2 6.4  0.3 6.8  0.2
Ppa , mm Hg 12.7  0.7 12.5  1.1 12.8  0.9
Ppa-Pla, mm Hg 3.9  0.7 4.6  0.3 3.9  1.0
PaO2, (mm Hg) 230  3 229  4 240  3
PaCO2, mm Hg 28  0.6† 36  4.7† 64  6.5
pH 7.38  0.01† 7.32  0.04† 7.12  0.04
T2 Peak alveolar pressure, cm H2O 15.1 0.2 25.2  0.2* 25.1  0.2*
Total PEEP, cm H2O 2.4  0.3 2.4  0.2 2.7  0.3
VT, ml 54  3 80  9* 83  8*
Pla, mm Hg 6.9  0.1 6.6  0.4 6.9  0.3
Ppa , mm Hg 11.8  1 14.1  1 13.5  1
Ppa-Pla, mm Hg 4.0  0.6 5.9  1.7 4.0  0.9
PaO2, mm Hg 229  5 233  6 244  4
PaCO2, mm Hg 40  3† 34  3† 93  7
pH 7.3  0.02† 7.3  0.03† 6.98  0.02
T3 Peak alveolar pressure (cm H2O) 15.1  0.2 30.1  0.1* 30.0  0.2*
Total PEEP, cm H2O 2.2  0.2 2.4  0.2 2.7  0.2
VT, ml 52  2 84  10* 100  10*
Pla, mm Hg 6.9  0.1 6.5  0.3 6.9  0.3
Ppa , mm Hg 11.9  0.7 16.7  1.4* 15.7  0.8*
Ppa-Pla, mm Hg 4.9  0.7 9.5  1.2‡ 6.1  0.9
PaO2, mm Hg 234  5 230  7 247  3
PaCO2, mm Hg 37  3† 45  6† 105  6
pH 7.29  0.02† 7.29  0.04† 6.93  0.02

Definition of abbreviations: PaCO2  arterial carbon dioxide tension; PaO2  arterial oxygen tension; PEEP  positive end-expiratory pres-
sure; Pla  left artrial pressure; Ppa  pulmonary artery pressure; Ppa-Pla  pulmonary artery pressure minus left atrial pressure measured
with continuous positive airway pressure.
* p 0.05 versus Control group.

p 0.05 versus High-Pressure Hypercapnic group.

versus the two other groups.
804 AMERICAN JOURNAL OF RESPIRATORY AND CRITICAL CARE MEDICINE VOL 164 2001

Figure 1. Indices of lung injury. *p


0.05 within-group comparison (T3 versus
T1), p 0.05 for between-group compar-
ison at the same time; † versus C group.

versus C group and HPHC group.

gression analysis. The level of


was set at 0.05. In the RESULTS sec- T1 to T3 (p  0.04 and p  0.25, respectively). In the HPNC
tion, all data are summarized as mean  SEM. group, both WG (p  0.01) and Kf (p  0.005) increased
significantly over time. At the end of the protocol (T3), WG
RESULTS and Kf were significantly greater in the HPNC than in the
HPHC or C groups (Figures 1a and 1b; WG: p 0.001; Kf
The three study groups had similar characteristics at Baseline, p  0.01). The HPHC group differed significantly from the C
as summarized in Table 1. In particular, the initial WG did not group for WG (p  0.03) but not for Kf (p  0.27).
differ among the groups. The Baseline Kf was higher in the C At the end of the experimental period, the WWLDWL ratio
group than in the HPHC group (Table 1), but this difference was higher in the HPNC than in the HPHC or C groups (p
was small, and values in all three groups were well within the 0.005; HPNC group  8.33  0.25; HPHC group  7.19  0.18; C
normal range for rabbit lungs (21). The ischemic time (21  1 group  6.85  0.34). The BALF protein concentration was
[mean  SEM] min), the initial hematocrit (6.2  0.1%), the higher in the HPNC group than in the C group (p 0.05) (Figure
initial perfusate pH (7.30  0.03), and the perfusate tempera- 1c), whereas BALF hemoglobin levels were higher in the two ac-
ture (36.5  0.1 C) did not differ among the groups. After tive treatment groups than in the C group (p 0.05) (Figure 1d).
randomization, the controlled variables for the three groups The BALF protein concentration was higher in the HPNC than
differed at each stage of the study, as intended (Table 2). in the HPHC group (p 0.05), with a similar but statistically in-
significant trend noted for BALF hemoglobin (p  0.24).
Indices of Lung Injury
Neither edema formation (WG: Figure 1a) nor permeability NOx in BALF and Perfusate
(Kf: Figure 1b) increased significantly over time in the C The overall mean circulating NOx was 41.2  3.1 ml/L at Base-
group. In the HPHC group, WG but not Kf increased from line and 58.8  3.3 ml/L at T3 (p 0.01). The level of NOx in

Figure 2. NOx concentration (a) change over time in per-


fusate and (b) at the end in BALF. *p 0.05 versus C and
HPHC groups, † p 0.05 versus C group.
Broccard, Hotchkiss, Vannay, et al.: Lung Injury and Mechanical Ventilation 805

the perfusate increased over time in all groups. This change tures, we are confident that the mechanical stress imposed on
was less marked in the HPHC group than in the other two the lungs ventilated with high pressures was sufficient to pro-
groups, a difference that did not reach statistical significance duce injury, at least in normocapnia.
(Figure 2a). The level of NOx in the BALF, measured at the
end of the experimental period, was highest in the HPNC
group, intermediate in the HPHC group, and lowest in the C Possible Mechanisms of Protection Against VILI Afforded by
group (Figure 2b, p 0.05 for all comparisons). BALF NOx Respiratory Acidosis
correlated significantly with all indices of injury: Kf (r  0.65, The study data clearly indicate a protective effect of hypercap-
p 0.001); BALF hemoglobin (r  0.62, p 0.01); and BALF nic acidosis against VILI in this model. Indeed, with the possi-
protein (r  0.7, p 0.001). ble exception of BALF hemoglobin, all markers of lung injury
that were noted to be increased in the HPNC group were
Hemodynamic Data blunted in the HPHC group (Figure 1). The study was prima-
At Baseline, there were no significant differences in vascular rily conducted to establish this fact. It was not designed to sort
pressures among the study groups (Table 1). Throughout the out the relative contributions of PCO2 and pH. Nor did it spe-
experiment, Pla was adequately controlled at values around cifically address the possible mechanisms of protection, for
the targeted value (Table 2). After randomization, the mean which, nevertheless, a few clues exist in the data.
pulmonary artery pressure (Ppa) measured during tidal venti- Hemodynamic factors. In the course of experimentation,
lation remained close to the Baseline value and similar in all the Ppa and vascular resistance increased less in the HPHC
groups until T2. At the beginning of T3, it became higher in the than in the HPNC group (Table 2). This difference might have
HPNC and HPHC groups than in the C group (Table 2). Mean been due to hypercapnia per se, which is a known vasodilator
Ppa differed among all groups only at the end of T3 and was in the pulmonary circulation (22). In the presence of lung in-
highest in the HPNC, intermediate in the HPHC, and lowest in jury, differences in Ppa may have a significant impact on
the C group (data not shown, p 0.05 for all comparisons). edema formation and possibly on injury. Nevertheless, several
Pcp measured under apneic conditions with a continuous posi- arguments run against a fundamental role of hemodynamic
tive airway pressure (CPAP) of 5 cm H2O was similar among factors in the lung-protective effects of respiratory acidosis.
groups from Baseline (Table 1) to the end of the study [9.2  First, although hypercapnia largely blunted the formation of
0.2 cm H2O (C group); 9.8  0.4 cm H2O (HPNC group); 8.5  edema (Figure 1a), it had no influence on capillary pressure
0.5 cm H2O (HPHC group); p  0.1]. Pulmonary vascular re- throughout the experiment. Second, the hemodynamic differ-
sistance (as reflected by Ppa minus Pla under conditions of ence between the HPNC and HPHC groups was present only
constant lung perfusion, interrupted ventilation, and a CPAP after significant injury developed (end of T3) and was limited
of 5 cm H2O), remained practically stable and comparable to a modestly higher Ppa in the HPNC group (19.6  1.1 mm
among groups until the end of T2 (Table 2). At the end of T3, Hg compared with 16.5  0.8 mm Hg; p 0.05). Respiratory
by contrast, this variable was significantly higher in the HPNC acidosis per se had no discernible effect on the vascular tone
group than in the other two groups (Table 2) (p 0.05). At prior to injury, probably because the CO2-mediated vasodila-
this time, pulmonary vascular resistance correlated with WG tion in this condition is offset by acidosis-mediated vasocon-
(r  0.58, p  0.005) and Kf (r  0.5, p  0.02). striction (22) (i.e., at T1 and T2; Table 2). These consider-
ations converge to indicate that pulmonary hypertension was
most likely a marker rather than a cause of VILI in this model,
DISCUSSION as also documented by other investigators (23).
Possible role of NO. We found that circulating NOx tended
Our main finding was that respiratory acidosis decreased the
to increase less from T1 to T3 in the HPHC group than in the
severity of VILI in this model. Hemodynamic effects of the
other two study groups (Figure 2a). In addition, BALF NOx
acid–base status were unlikely to account for this observation,
at the end of the experimental period was lower in the HPHC
because they were modest and only apparent after significant
group than in the HPNC group (Figure 2b), and correlated
lung injury developed. Our data raise the hypothesis that re-
with all indices of injury. Since BAL and circulating NOx
spiratory acidosis could reduce VILI by attenuating the in-
track the formation of NO within the lung (24), our results
creased NO production arising from high-pressure mechanical
suggest that the latter may have been blunted by respiratory
ventilation.
acidosis. Indeed, depressed NO production by high airway
concentrations of CO2 has been reported by other investiga-
Adequacy of the Experimental Model tors, both in vivo and in isolated perfused rabbit lungs (12).
The validity of the present experiments depends crucially on That BALF NOx was higher in the HPNC than in the C group
the actual induction of VILI in the group of isolated lungs would be consistent with recent data indicating that lung
ventilated with high pressure in normocapnia (HPNC). Injury stretch may stimulate the production of NO by the airway mu-
was assessed by measurement of edema formation (WG) cosa (25, 26). At the cellular level, NO may be both protective
and altered lung permeability to markers covering a wide and cytotoxic, the net result being highly dependent on the
range of sizes (i.e., water [Kf], protein [BALF protein con- particular pathophysiologic state (13). Reflecting this com-
centration], and red blood cells [BALF hemoglobin concen- plexity, inhalation of NO was protective in a model of oxidant-
tration]). Each of these measurements taken in isolation has induced lung injury (27), whereas the endogenous production
limitations for assessing injury. However, all of them signifi- of NO was deleterious to the lung in experimental endotox-
cantly differed in the HPNC and the C groups at the end of emia (28).
the experiment (Figure 1). In our previous study of the same If the cytotoxic potential of NO predominated under the
model, injurious ventilatory patterns induced these same mul- conditions of the present study, our results might fit into the
tiple abnormalities, which correlated closely with histologic following scenario: (1) stretch-induced overproduction of NO
scores of both alveolar and extraalveolar hemorrhage (17). To contributed to VILI; and (2) hypercapnic acidosis was protec-
the extent that extravasation of red blood cells is indicative of tive by interfering with this mechanism. Further experiments
either anatomic or severe functional disruption of lung struc- will be required to test this hypothesis.
806 AMERICAN JOURNAL OF RESPIRATORY AND CRITICAL CARE MEDICINE VOL 164 2001

Limitations of the Study Naoki K, Kudo H, Yamaguchi K. Hyperoxia and hypercapnic acidosis
differentially alter nuclear factor-kappa B activation in human pulmo-
The model used in the study has important limitations that are nary artery endothelial cells. Adv Exp Med Biol 1999;471:265–270.
discussed elsewhere (16), and characteristics that may prevent 8. Xu L, Fidler IJ. Acidic pH-induced elevation in interleukin 8 expression
direct extrapolation of our results to the intact animal, and by human ovarian carcinoma cells. Cancer Res 2000;60:4610–4616.
certainly to patients. For instance, we kept blood flow con- 9. Shepard JW Jr., Dolan GF, Yu SY. Factors regulating lamellar body vol-
stant. In the intact animal, respiratory acidosis enhances both ume density of type II pneumocytes in excised dog lungs. J Appl Phys-
sympathetic activity and catecholamine secretion, which tend iol 1982;53:555–562.
10. Chander A. Regulation of lung surfactant secretion by intracellular pH.
to increase cardiac output and pulmonary vascular resistance Am J Physiol 1989;257:L354–L360.
and pressure (3). This could partly offset the beneficial effect 11. Zhu S, Basiouny KF, Crow JP, Matalon S. Carbon dioxide enhances ni-
of hypercapnia on VILI. In addition, assuming that reduced tration of surfactant protein A by activated alveolar macrophages.
NO production is an important mechanism by which respira- Am J Physiol (Lung Cell Mol Physiol) 2000;278:L1025–L1031.
tory acidosis ameliorates VILI, the net result may be either 12. Adding LC, Agvald P, Persson MG, Gustafsson LE. Regulation of pul-
beneficial or detrimental according to the underlying type of monary nitric oxide by carbon dioxide is intrinsic to the lung. Acta
Physiol Scand 1999;167:167–174.
lung insult requiring mechanical ventilation. Although hyper- 13. Liaudet L, Soriano FG, Szabo C. Biology of nitric oxide signaling. Crit
capnia has been reported to be associated with an improved Care Med 2000;28:N37–N52.
outcome (1, 29, 30) and to cause no serious complications, the 14. Shibata K, Cregg N, Engelberts D, Takeuchi A, Fedorko L, Kavanagh
improved outcome could be entirely due to other aspects of BP. Hypercapnic acidosis may attenuate acute lung injury by inhibition
the ventilatory strategy used in the studies in which this was of endogenous xanthine oxidase. Am J Respir Crit Care Med 1998;158:
found, and not to hypercapnia per se. Moreover, whether hy- 1578–1584.
15. Laffey JG, Tanaka M, Engelberts D, Luo X, Yuan S, Keith TA, Post M,
percapnia resulting from hypoventilation and hypercapnia re- Lindsay T, Kavanagh BP. Therapeutic hypercapnia reduces pulmo-
sulting from inhaled CO2 may provide equivalent lung protec- nary and systemic injury following in vivo lung reperfusion. Am J
tion is still unknown. Respir Crit Care Med 2000;162:2287–2294.
16. Broccard AF, Hotchkiss JR, Suzuki S, Olson D, Marini JJ. Effects of
Conclusions mean airway pressure and tidal excursion on lung injury induced by
Two recent studies by Kavanagh and colleagues demonstrated mechanical ventilation in an isolated perfused rabbit lung model. Crit
Care Med 1999;27:1533–1541.
the lung-protective potential of hypercapnic acidosis in an is- 17. Broccard AF, Hotchkiss JR, Kuwayama N, Olson DA, Jamal S, Wangens-
chemia–reperfusion model of lung injury (14, 31). Our results teen DO, Marini JJ. Consequences of vascular flow on lung injury induced
show that the protection afforded by hypercapnic acidosis is by mechanical ventilation. Am J Respir Crit Care Med 1998;157:1935–1942.
not limited to ischemia–reperfusion injury, but also applies to 18. Liaudet L, Fishman D, Markert M, Perret C, Feihl F. L-canavanine im-
VILI. This is of great interest, since lung-protective ventila- proves organ function and tissue adenosine triphosphate levels in ro-
tory strategies tend to induce respiratory acidosis. Respiratory dent endotoxemia. Am J Respir Crit Care Med 1997;155:1643–1648.
19. Green LC, Wagner DA, Glogowski J, Skipper PL, Wishnok JS, Tannen-
acidosis may worsen gas exchange in patients with acute respi- baum SR. Analysis of nitrate, nitrite, and [15N]nitrate in biological flu-
ratory distress syndrome (32) and may have undesirable ef- ids. Anal Biochem 1982;126:131–138.
fects on organs other than the lungs, as reviewed elsewhere 20. Kenneth A, Tait Malloy E, Tait Malloy H. Microdetermination of oxy-
(3). If confirmed in intact animals, our results would, however, hemoglobin, methemoglobin, and sulfhemoglobin in a single sample
indicate that hypercapnic acidosis should not necessarily be of blood. J Biol Chem 2000;126:665–672.
viewed as an undesirable side effect to be tolerated, but rather 21. Hernandez LA, Peevy KJ, Moise AA, Parker JC. Chest wall restriction
limits high airway pressure-induced lung injury in young rabbits. J
as a potentially useful adjunct to current strategies for lung Appl Physiol 1989;66:2364–2368.
protection during mechanical ventilation. Our results also re- 22. Brimioulle S, Lejeune P, Vachiery JL, Leeman M, Melot C, Naeije R.
inforce the concept that medical interventions designed to re- Effects of acidosis and alkalosis on hypoxic pulmonary vasoconstric-
store normal physiologic parameters may be counterproduc- tion in dogs. Am J Physiol 1990;258:H347–H353.
tive in critically ill patients, at least under certain clinical 23. Parker JC, Gillespie MN, Taylor AE, Martin SL. Capillary filtration co-
circumstances. efficient, vascular resistance, and compliance in isolated mouse lungs.
J Appl Physiol 1999;87:1421–1427.
Acknowledgment : The authors thank Camille Anglada, technician in the 24. Su WY, Day BJ, Kang BH, Crapo JD, Huang YC, Chang LY. Lung epi-
Division of Pathophysiology, Department of Internal Medicine, University thelial cell-released nitric oxide protects against PMN-mediated cell
Hospital, Lausanne, Switzerland, for the time and technical assistance he injury. Am J Physiol 1996;271:L581–L586.
provided to us. They are grateful to Terramed, Inc. and Hamilton Medical, 25. Carlin RE, Ferrario L, Boyd JT, Camporesi EM, McGraw DJ, Hakim
Rhazüns, Switzerland, for lending us a Veolar ventilator for this study. TS. Determinants of nitric oxide in exhaled gas in the isolated rabbit
lung. Am J Respir Crit Care Med 1997;155:922–927.
26. Adding LC, Bannenberg GL, Gustafsson LE. Gadolinium chloride inhi-
References bition of pulmonary nitric oxide production and effects on pulmonary
1. Amato MB, Barbas CS, Medeiros DM, Magaldi RB, Schettino GP, circulation in the rabbit. Pharmacol Toxicol 1998;83:8–15.
Lorenzi-Filho G, Kairalla RA, Deheinzelin D, Munoz C, Oliveira R, 27. Kavanagh BP, Mouchawar A, Goldsmith J, Pearl RG. Effects of inhaled
et al. Effect of a protective-ventilation strategy on mortality in the NO and inhibition of endogenous NO synthesis in oxidant-induced
acute respiratory distress syndrome. N Engl J Med 1998;338:347–354. acute lung injury. J Appl Physiol 1994;76:1324–1329.
2. The Acute Respiratory Distress Syndrome Network. Ventilation with 28. Matsuo N. The role of intrapulmonary nitric oxide generation in the de-
lower tidal volumes as compared with traditional tidal volumes for velopment of adult respiratory distress syndrome. Surg Today 1999;29:
acute lung injury and the acute respiratory distress syndrome. N Engl 1068–1074.
J Med 2000;342:1301–1308. 29. Darioli R, Perret C. Mechanical controlled hypoventilation in status
3. Feihl F, Perret C. Permissive hypercapnia: how permissive should we asthmaticus. Am Rev Respir Dis 1984;129:385–387.
be? Am J Respir Crit Care Med 1994;150:1722–1737. 30. Hickling KG, Henderson SJ, Jackson R. Low mortality associated with low-
4. Dos Santos CC, Slutsky AS. Invited review: mechanisms of ventilator- volume, pressure limited ventilation with permissive hypercapnia in severe
induced lung injury: a perspective. J Appl Physiol 2000;89:1645–1655. adult respiratory distress syndrome. Intensive Care. Med 1990;16:372–377.
5. Miura M, Takayama K, Okada J. Neuronal expression of Fos protein in the 31. Laffey JG, Engelberts D, Kavanagh BP. Buffering hypercapnic acidosis
rat brain after baroreceptor stimulation. J Auton Nerv Syst 1994; 50:31–43. worsens acute lung injury. Am J Respir Crit Care Med 2000;161:141–146.
6. Kuo NT, Agani FH, Haxhiu MA, Chang CH. A possible role for protein 32. Feihl F, Eckert P, Brimioulle S, Jacobs O, Schaller MD, Melot C, Naeije
kinase C in CO2H-induced c-fos mRNA expression in PC12 cells. R. Permissive hypercapnia impairs pulmonary gas exchange in the
Respir Physiol 1998;111:127–135. acute respiratory distress syndrome. Am J Respir Crit Care Med 2000;
7. Takeshita K, Suzuki Y, Nishio K, Aoki T, Takeuchi O, Toda K, Sato N, 162:209–215.

Anda mungkin juga menyukai