Next Article in Journal
Bayesian Information-Theoretic Calibration of Radiotherapy Sensitivity Parameters for Informing Effective Scanning Protocols in Cancer
Next Article in Special Issue
Airway Epithelial Dysfunction in Asthma: Relevant to Epidermal Growth Factor Receptors and Airway Epithelial Cells
Previous Article in Journal
Physical Activity in 6.5-Year-Old Children Born Extremely Preterm
Previous Article in Special Issue
Virus-Induced Asthma Exacerbations: SIRT1 Targeted Approach
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Hypercapnia: An Aggravating Factor in Asthma

1
Division of Pulmonary and Critical Care Medicine, Northwestern University, Chicago, IL 60611, USA
2
Department of Allergology and Respiratory Medicine, University of Showa School of Medicine, Tokyo 142-0064, Japan
*
Author to whom correspondence should be addressed.
J. Clin. Med. 2020, 9(10), 3207; https://doi.org/10.3390/jcm9103207
Submission received: 12 September 2020 / Revised: 1 October 2020 / Accepted: 2 October 2020 / Published: 5 October 2020
(This article belongs to the Special Issue New Frontiers in Asthma Treatment & Management)

Abstract

:
Asthma is a common chronic respiratory disorder with relatively good outcomes in the majority of patients with appropriate maintenance therapy. However, in a small minority, patients can experience severe asthma with respiratory failure and hypercapnia, necessitating intensive care unit admission. Hypercapnia occurs due to alveolar hypoventilation and insufficient removal of carbon dioxide (CO2) from the blood. Although mild hypercapnia is generally well tolerated in patients with asthma, there is accumulating evidence that elevated levels of CO2 can act as a gaso-signaling molecule, triggering deleterious effects in various organs such as the lung, skeletal muscles and the innate immune system. Here, we review recent advances on pathophysiological response to hypercapnia and discuss potential detrimental effects of hypercapnia in patients with asthma.

1. Introduction

Asthma is a chronic disease characterized by reversible airway obstruction caused by bronchial smooth muscle contraction, airway inflammation and increased secretions, and is currently estimated that more than 330 million people are afflicted with this disease worldwide [1,2]. The natural history of asthma is punctuated by acute exacerbations, most of which respond to conventional treatment using inhaled bronchodilators and corticosteroids, and oxygen. However, deterioration or failure to respond to these measures sometimes leads to severe respiratory failure. Approximately 5 to 10% of asthmatic patients experience a severe asthma attack each year and, of those who are admitted to hospital, 10% require intensive care unit (ICU) admission [3]. Ten to twenty-six percent of cases with acute severe asthma present to the emergency department with hypercapnia [3]. In-hospital mortality rates for patients with severe asthma who require ICU admission is 3.2 to 9.8%, with higher mortality rates seen in those patients who require invasive ventilation [4]. Hypercapnia is associated with the institution of mechanical ventilation and greater risk for in-hospital death in acute severe asthma [5,6].
Hypercapnia, an elevation in the arterial carbon dioxide (CO2) tension, is a complication of inadequate alveolar gas exchange in patients with severe acute and chronic lung diseases [7] including asthma [5,6,8,9,10,11,12,13,14,15,16,17]. It has been initially reported that hypercapnia was innocuous or even protective in mechanically ventilated patients with severe asthma [9,10], acute lung injury and acute respiratory distress syndrome (ARDS) [18,19,20,21], where the concepts of “permissive” and even “therapeutic” hypercapnia have been proposed for the mechanically ventilated patients. The lower tidal volumes during protective ventilation can lead to hypercapnia and an associated drop in pH resulting in hypercapnic respiratory acidosis that has been reported as a protective effect via the inhibition of the nuclear factor-κB (NF-κB) pathway, a pivotal transcription activator in inflammation and injury [7,22]. However, in recent years, it has become increasingly evident that elevated CO2 acts as a gaso-signaling molecule, resulting in deleterious effects in various organs such as the lung [23,24,25,26,27,28,29] and skeletal muscles [30,31,32] as well as innate immunity system [25,29,33,34,35,36,37]. In the lung, recent studies reported that high levels of CO2 activate specific gene expression [26,38,39,40] and signal transduction pathways with adverse consequences on alveolar fluid clearance through Na, K-ATPase endocytosis via intracellular calcium- or extracellular signal-regulated kinase (ERK)-mediated AMP-activated protein kinase (AMPK)/protein kinase C-ζ/c-Jun-N-Terminal Kinase (JNK) signaling or soluble adenylyl cyclase-mediated protein kinase A-Iα signaling [24,41,42,43,44,45,46] and epithelial cell repair via AMPK-mediated Rac1-GTPase signaling, NF-κB pathways or miR-183-regulated mitochondrial dysfunction [28,47,48,49]. In addition, a secondary analysis of three prospective non-interventional cohort studies of ARDS patients receiving mechanically ventilation reported that severe hypercapnia was independently associated with higher ICU mortality [23]. These recent reports have led to the re-evaluation of the potential complexity of hypercapnia effects, and are stimulating more research to better understand its biologic effects. Here, we review recent advances on pathophysiological response to hypercapnia; CO2 sensing, CO2-dependent regulation of respiration and signaling events initiated by hypercapnia, and discuss the relevance of these data to patients with asthma and hypercapnia.

2. CO2 Sensing and Respiration

Cells possess the ability to sense and respond to changes in concentration of gaseous molecules through evolutionarily conserved pathways [50]. CO2 is a small non-polar molecule and produced in the mitochondria of eukaryotic cells during oxidative phosphorylation and its physiological levels in mammalian tissues (~5%) [7] are significantly higher than those found in the atmosphere (~0.04%) [51,52]. CO2 is thought to traverse biological cell membranes via passive diffusion, depending upon the transmembrane concentration gradient of CO2 and the lipid/water partition behavior of the gas [53]. However, the discovery of the effect of cholesterol on CO2 permeability and of protein channels used by CO2 to cross membranes such as aquaporins (AQPs) [54,55] and rhesus proteins [56] has challenged this view. Functionally, AQP1, AQP4-M23, AQP5 and AQP6 seem to effect high permeability for CO2 [55]. In the lung, AQP1 is expressed in microvascular endothelia, AQP3 and AQP4 in airway epithelia, and AQP5 in type I alveolar epithelial cells and a subset of airway epithelial cells [54]. Once inside the cell, CO2 very rapidly equilibrates with its hydrated form, H2CO3, which in turn dissociates into H+ and HCO3 catalyzed by carbonic anhydrases [57].
CO2 + H2O ⇌ H2CO3 ⇌ H+ + HCO3
Increased partial pressure CO2 (pCO2) in the blood, which occurs due to insufficient removal of CO2 (alveolar hypoventilation), can give rise to elevated pCO2 in the cerebrospinal fluid and result in elevated H+ concentrations, cerebrospinal fluid acidification. Multiple sites within the central nervous system are capable of sensing and eliciting rapid adaptive responses to these changes, which results in alteration in the rate and depth of breathing.

2.1. Central CO2 Chemosensing

Acute chemosensing of CO2 is a complex process involving integration of multiple brain regions, effector channels and molecules. Neurons detect changes in CO2/H+ and modulate the CO2-chemosensory regulation of respiration. Several regions in the brain, particularly medulla oblongata, medullary raphe and cerebellum in the brain stem, have been described as potential area of interest [58,59,60,61,62]. A recent report suggests important crosstalk between the carotid body and central chemosensing regions of the brain that determines the respiratory response to altered CO2 level [63]. Much of the work in this area has been reviewed by Cummins et al. [50]. Here, we review key molecular mechanisms of the brain that are involved in CO2 chemosensitivity to elicit the change in respiratory rate.

2.1.1. pH-Sensitive Ion Channel

The ability to acutely sense and respond to elevated CO2 levels occurs via a physiological adaptation to reflect acid/base balance in the blood. The pH is a major effector of CO2-dependent signaling in the brain. The Twik-related acid-sensing potassium (K+) (TASK) channels are members of the background K2P channel family that facilitate selective K+ leak and contribute to the negative resting membrane potential in cells [64]. TASK-1, -2 and -3 channels play a role in CO2-dependent regulation of breathing. TASK-1 and -3 channels display acid sensitivity and are widely expressed in known chemosensing regions in the brain [65]. Catecholaminergic neurons in the locus coeuruleus have also been proposed to contribute to the ventilatory response to hypercapnia [66]. Specifically, several of transient receptor potential channels (TRPC), in particular TRPC 5, are sensitive to pH and enriched in chemosensory regions of the brain.

2.1.2. CO2-Sensitive Connexin Protein

Huckstepp et al., reported adenosine triphosphate (ATP) released from brain slices derived from the ventral surface of the medulla oblongata in response to elevated CO2 levels, independently of extracellular acidification [67]. Connexin hemi-channels including connexin 26 in the medulla oblongata were also reported as contributing to the ATP release in known chemosensory regions. Subsequent studies revealed a role for inward rectifying K+ channels [67], participating in hyperpolarization of excitable cells and CO2-dependent inhibition during hypercapnia. A recent study reported that the chemo-sensitivity of connexin 26 linked directly to a CO2-dependent posttranslational modification of the channel, independently of pH changes [68]. Molecular CO2 can bind to Lys125 on connexin 26 forming a carbamate bridge between Lys125 and a neighboring residue, Arg104. The CO2-dependent modification causes a structural change in the gap junction, which facilitates altered connexin-dependent signaling (e.g., ATP release). This study identified central chemo-sensitivity to elevated CO2 mediated not only by indirect changes in pH but directly by high CO2-dependent modifications.

3. Hypercapnia in Asthmatic Patients

The pH-modulating effects of hypercapnia can be attenuated via bicarbonate reabsorption by the kidneys [69]. However, during acute hypercapnia, the buffering capacity of the blood is not sufficient to handle the excess CO2, resulting in acute respiratory acidosis (pH < 7.35). Levels of partial pressure of CO2 in arterial blood (PaCO2) among asthmatic patients experiencing severe exacerbations varies considerably. Scala reported that hypercapnia occurred in 10 to 26% of cases presenting to the emergency department with greater airway obstruction, higher respiratory rate and pulsus paradoxus [3]. The medical literature reports cases of severe hypercapnia in asthmatic patients with values of PaCO2 reaching 202 and 218 mmHg (pH value, 6.68 and 6.90, respectively) in two 24 and 28 year-old women during severe status asthmaticus [12], 208 mmHg (pH = 6.73) in a 35 year-old woman during an episode of near fatal asthma [13] and 175 mmHg (pH = 6.99) in a 33 year-old woman during severe status asthma [17]. Pediatric reports are scarce, but an eight year-old boy during an episode of near fatal asthma had PaCO2 values of 293 mmHg (pH = 6.77) [14], a two year-old girl with status asthmaticus had the values of 238 mmHg (pH = 6.71) [16] and an eleven year-old boy with status asthmaticus with 187 mmHg and pH of 6.84 [15].
In patients with asthma, the presence of hypercapnia reflects more severe airflow obstruction and more severe chronic asthma conditions [8,11]. Mountain et al., reported that acute asthmatic patients with hypercapnia (mean PaCO2 value, 53.6 mmHg) were more likely to require maintenance therapy with β-adrenergic agents and corticosteroids, and were less likely to have been discharged from an emergency room visit [11]. In a secondary analysis of data from a clinical database, the Intensive Care National Audit and Research Centre (ICNARC), hypercapnia was shown to be associated with severe asthmatic patients requiring mechanical ventilation and greater risk for in-hospital death after adjusting for Acute Physiology and Chronic Health Evaluation (APACHE) II score [5]. Stow et al., reported that in asthmatic patients admitted to Australian ICUs from 1996 to 2003 non-survivors who were not ventilated in the first 24 h had a higher PaCO2 level, but no difference of arterial oxygen tension, than those who did survive (mean PaCO2 value, 74.1 vs. 54.3 mmHg) [6]. Asthmatic patients presenting with worsening hypercapnia and respiratory acidosis require intubation and the need for ventilatory assistance. In mechanically ventilated patients with asthma, “permissive” hypercapnia is the currently recommended strategy for severe asthma with the goals of minimizing barotrauma/volutrauma [9,10,70,71]. Elsayegh et al., reported that the peak value of PaCO2 on the first day of mechanical ventilation with the “permissive” hypercapnia averaged 67 mm Hg and exceeded 100 mmHg in 12% of cases (the highest PaCO2 value, 159 mmHg) [72]. With this approach, decreasing the respiratory rate and tidal volume as well as increasing the inspiratory flow rate leads to an increase in expiratory time and decrease of the dynamic hyperinflation.

4. Detrimental Effects of Hypercapnia in Asthma

The symptoms and physiologic consequences of hypercapnia are significant. A series of adaptive mechanisms are activated in vital organs such as brain and heart to preserve tissue oxygenation and perfusion, in particular by preservation and defense of intracellular pH. The injurious effects of hypercapnia on the central nervous and cardiovascular systems are well documented [70]. Hypercapnia results in cerebrovascular vasodilatation leading to an increase in intracranial pressure by increasing the blood volume in the brain [70,73]. There have been reports in patients with severe asthma developing cerebral edema and subarachnoid hemorrhage as a complication of hypercapnia or “permissive hypercapnia” [15,17,74]. The myocardial response to hypercapnia is characterized by impairment in contractility due to acute respiratory acidosis, which is reversible [70,75]. Accumulating scientific evidence points to the role of high CO2 on the lung airways, innate immunity and adipogenesis, which could contribute to the disease pathogenesis and progression of asthma.

4.1. Lung Airways

The predominant feature of asthma is shortness of breath or dyspnea due to the excessive constriction of the airway smooth muscles. As such, relieving airway smooth muscle constriction is a therapeutic target of asthma management. Elevated CO2 levels are reported to modulate the tone of lung airways, which is in a dynamic equilibrium between excitatory and inhibitory mechanisms. Lung airway cells sense and respond to changes in CO2 levels via specific mechanisms of the vagus reflexes, molecular CO2 and pH effects. The effects of hypercapnia on the airways and airway smooth muscle is complicated. There are reports attesting to it causing increased airway contractility [26,76,77,78,79,80,81,82,83,84,85] or airway relaxation [86,87,88,89,90,91,92,93,94,95,96,97]. We review recent advances in our understanding of how elevated CO2 conditions modulate the airway tone, focusing on the effects of hypercapnia and respiratory acidosis.

4.1.1. Hypercapnia

Airway tone is regulated by interaction of the sympathetic and parasympathetic pathways [82,98] where the stimulation of vagal efferent nerves can increase bronchoconstriction [82,98,99,100]. Evidence suggesting that changes in CO2 levels in the blood affect the airway tone was first reported in 1892 [76]. Einthoven described that inhalation of high concentrations of carbonic acid (CO2-rich mixtures) caused bronchoconstriction in dogs, which was confirmed in various models of normoxic hypercapnia-exposed dogs [77,78,79,80,81] and cats [82,83]. The hypercapnia-induced bronchoconstriction was abolished by blocking the vagus nerve and understood to be dependent on the integrity of vagal conduction [76,77,78,79,80,82,83]. In healthy human volunteers, it has been reported that inhalation of high CO2 concentrations decreases specific airway or pulmonary conductance [84,85]. The increases in airway resistance during high CO2 exposure were initially interpreted as extrathoracic airway narrowing [84] such as larynx narrowing [85], because the hypercapnic effect was not blocked by atropine or β12 adrenergic receptor agonists. However, the direct studies of laryngeal resistance during high CO2 exposure indicated no change in animal models [101] and normal human subjects [102]. Furthermore, several reports of bronchoconstriction in the hypercapnia-exposed animal models [78,80,82] revealed that the blockage of the vagus nerve did not entirely abolish the bronchoconstrictor response to the high CO2 exposure, suggesting that other mechanisms contribute to the hypercapnia-mediated airway constriction. More recently, we have reported that high CO2 acts as a signaling molecule to increase smooth muscle contraction in mouse and human airway smooth muscle cells [26]. We found that high concentrations of CO2, independently of hypoxia and extracellular pH, increased acetylcholine-induced cell contraction dependent on CO2 dose and exposure time in cell culture systems. In a murine model, the exposure to normoxic hypercapnia increased acetylcholine-induced airway contraction in precision lung cut slices as well as airway resistance. Furthermore, we found that, in a small cohort of patients with chronic obstructive pulmonary disease (COPD), patients with hypercapnia (PaCO2 > 45 mmHg) had higher airway resistance, which improved after correction of hypercapnia (Figure 1).
Our study also provided insights into the molecular mechanisms by which high CO2 levels promote airway smooth muscle cell contractility via calcium-calpain signaling. The signaling was mediated by caspase-7, which by cleaving the transcription factor myocyte-specific enhancer factor 2D (MEF2D), leads to downregulation of the microRNA-133a (miR-133a) and consequent upregulation of Ras homolog family member (Rho) A and myosin light-chain (MLC) phosphorylation. Our data suggest that elevated CO2 levels activate specific signal transduction pathways in airway smooth muscle cells, which results in deleterious effects on the airway tone, leading to bronchoconstriction. Taken together, these more recent reports suggest that hypercapnia promotes airway constriction by activating the vagus nerve and high CO2-responsive signal transduction pathways to worsen airway obstruction in patients with severe asthma.

4.1.2. Respiratory Acidosis

Hypercapnia has been also reported to lead to airway relaxation [86,87,88,89,90,91,92,93,94,95,96,97]. Inhalation of high CO2 concentrations initially decreased airway constriction as well as the isolated bronchial ring tension caused by drugs such as 5-hydroxytryptamine [88,89,90]. It also reversed the airway constriction associated with pulmonary artery occlusion in ventilated animal models [86,88]. In humans, the administration of high CO2 relaxed the constriction of airways in a patient with unilateral pulmonary artery occlusion [87] and young asthmatic adults with hyperventilation [91] or exercise-induced bronchoconstriction [91,92]. These effects of hypercapnia were not mediated by the nerve reflexes and are understood to be due to changes in extracellular/intracellular pH levels, possibly acute respiratory acidosis in airway smooth muscle cells. Several in vitro studies reported that respiratory or normocapnic (metabolic) acidosis caused a reversible reduction in active tension of bronchial rings [89,90,93,94]. Extracellular pH can alter airway smooth muscle tone by changing the levels of intracellular pH and calcium (Ca2+) [94,95,103]. Intracellular acidification has been reported to decrease intracellular Ca2+ levels through voltage-dependent Ca2+ channels in the K+-induced contractile model, thereby inhibiting airway smooth muscle cell contraction [96]. We have reported that airway smooth muscle relaxation occurred during acute hypercapnia, but it was early, modest and transient [26]. As such, we reason that elevated CO2 levels may have a transient relaxing effect on contracted airways due to the decrease in pH, i.e., respiratory acidosis. There are no reports describing that hypercapnia and acute respiratory acidosis improved airway contractility or obstruction during acute exacerbation in patients with severe asthma.

4.2. Innate Immunity

Respiratory infection is one of the risk factors for development and exacerbation in patients with asthma [104,105,106,107]. Recent studies have reported that viral and/or bacterial infections were observed in 70% of adult inpatients with an asthma exacerbation [106]. Viral and bacterial super-infection is an important determinant of severe acute exacerbations and was more likely to result in hospital readmission following severe acute exacerbation [104]. Hypercapnia has been reported to be associated with increased mortality in hospitalized patients with community-acquired pneumonia [108] and in patients with cystic fibrosis awaiting lung transplantation [109]. In transcriptomic analyses of hypercapnia in model organisms, exposure to normoxic hypercapnia altered the expression of innate immune system genes in Caenorhabditis elegans [110] and Drosophila melanogaster [111]. In adult flies and the Drosophila S2 cell line, hypercapnia suppressed induction of genes involved in specific antimicrobial peptides such as diptericin that are regulated by Relish, an orthologue of the mammalian transcription factor NF-κB [111]. Transcriptomic studies in mouse neonatal lung tissue and human bronchial epithelial cells reported that hypercapnia altered the expression of components of the innate immune system [38,39]. It downregulated the expression of inflammatory mediator genes including interferons, interleukins, chemokines and tumor necrosis factor (TNF) in the neonatal lung [38]. In human bronchial epithelial cells, hypercapnic respiratory acidosis resulted in downregulation of genes related to the interleukin 6 (IL-6) receptor and chemokines [39]. Hypercapnia selectively inhibited the expression of IL-6 and TNF, and decreased phagocytosis in human and mouse alveolar macrophages [33]. Fitzpatrick et al., reported that in patients with moderate and severe asthma alveolar macrophage phagocytosis was decreased by more than 50% compared with that seen in control subjects and the impairment of phagocytosis was associated with poorly controlled asthma [112]. Hypercapnia also inhibited Beclin 1 activity by increases in Bcl-2 and Bcl-xL expression, and prevented autophagy and bacterial killing in human macrophages [36]. Furthermore, hypercapnia led to inhibition of the canonical NF-κB pathway while promoting activation of the noncanonical NF-κB component IKKα/RelB/p100, whose function is largely anti-inflammatory and immunosuppressive [34,35,113]. In these in vitro studies, the high CO2-induced inhibitions of cytokine gene expression, phagocytosis, autophagy and NF-κB signaling was independent of pH effects. Contrastingly, acidosis is known to impair the function of immune cells [114], including alveolar macrophages [115]. Thus, hypercapnia might modulate innate immunity and host defense via pH-independent or -dependent mechanisms. In mice exposed to normoxic hypercapnia, high CO2 levels decreased IL-6 and TNF expression in the lung during the early phase of Pseudomonas aeruginosa infection, and inhibited the phagocytosis of bacteria and generation of reactive oxygen species by lung neutrophils, resulting in increased burden of Pseudomonas aeruginosa in the lungs and other organs and higher mortality [25]. More recently, Casalino-Matsuda et al., has reported that hypercapnia increased virus-induced lung injury and mortality in mice infected with influenza A virus and hypercapnia [29]. They observed that elevated CO2 levels increased influenza A virus replication and inhibited antiviral gene and protein expression in macrophages. Interestingly, both in vivo studies showed similar reversibility of hypercapnia-induced defects in antiviral and antibacterial immunity [25,29]. There have been several reports suggesting that hypercapnia activates the renin-angiotensin system and angiotensin-converting enzyme 2 (ACE2) expression [116,117,118], which is identified as a receptor for the spike protein of SARS-CoV-2 and facilitating the viral entry into target cells [118]. It has not yet been reported that hypercapnia affects the pathogenesis of SARS-CoV-2 infection, but these findings in the studies of hypercapnia may also provide new insights into the understanding of the SARS-CoV-2 infection mechanisms. Elevated CO2 level should be taken into consideration as a potential risk factor to cause and worsen any infections in patients with severe asthma.

4.3. Adipogenesis

Asthma and obstructive sleep apnea (OSA) have been reported to coexist and contribute to an overlap syndrome where a bidirectional relationship may negatively affect the other condition [119,120,121]. A recent meta-analysis has revealed that the prevalence of OSA and OSA risk in adult asthmatic patients is 50% and 27.5%, respectively, and the odds of having OSA or OSA risk is 2.64 and 3.73 times higher in asthmatic patients than in non-asthmatic patients [122]. Asthmatic patients with OSA had significantly higher body mass index (BMI) in comparison with non-asthmatic patients [122]. Redline et al., reported that a 1 kg/m2 increase in BMI above the mean for age and sex translated to a 12% increase in risk of OSA [123], suggesting that OSA is related to obesity which is known to be prevalent in patients with severe asthma [124,125,126]. Obese patients with asthma have more severe disease with increased medication use [126] and a 4.6-fold higher risk of hospitalization as compared to the non-obese patients with asthma [127]. Obesity exacerbates OSA via several mechanisms; (1) neck adiposity decreases the size of the upper airway lumen, (2) abdominal adiposity decreases lung volumes and chest wall compliance, and increases airway resistance, (3) obesity-associated leptin resistance decreases ventilatory drive and response to hypercapnia [128,129]. Recently, Kikuchi et al., reported that either intermittent or sustained exposure to hypercapnia, independently of acidosis and oxygenation levels, promoted adipogenesis in visceral and subcutaneous preadipocytes [130]. The mechanisms by which hypercapnia induced adipogenesis lead to increased production of cyclic adenosine monophosphate (cAMP) via soluble adenylyl cyclase and activation of protein kinase A and exchanger protein directly activated by cAMP (EPAC). This, in turn, activates proadipogenic transcription factors, such as cAMP response element binding protein (CREB), CCAAT/enhancer binding protein β (C/EBP-β), and peroxisome proliferator–activated receptor γ (PPAR-γ). In addition, plasma leptin levels contribute to an aberrant hypercapnic ventilatory response in obese patients. Leptin is produced by adipocytes and its levels in serum correlate positively with total body fat mass [131]. In animal models, it has been reported that leptin-deficient mice show a blunted ventilator response to hypercapnia, suggesting that leptin can act as the respiratory stimulus [132,133]. In contrast, obese patients show a relative deficiency of leptin in the cerebrospinal fluid as compared with lean controls [134], suggesting a failure of central feedback mechanisms, “leptin resistance”. These data are supported by evidence linking hyperleptinemia and reduced respiratory drive and hypercapnic response to leptin resistance of the respiratory center [135]. Collectively, these reports suggest a maladaptive cycle between hypoventilation, hypercapnia and increased fat mass, leading to the progression of obesity and OSA, thus contributing to asthma severity.

5. Conclusions

Hypercapnic respiratory failure is a hallmark of severe asthma. As reviewed above, elevated CO2 levels are rapidly sensed by chemosensing regions of the brain that regulate the respiratory drive, however, there are many reports suggesting that hypercapnia is also sensed by non-excitable cells and has significant effects on cellular and tissue functions. Recent discoveries suggest that hypercapnia increases airway contractility, impairs the innate immune response, and promotes adipogenesis, which likely underlies, at least in part, the negative effects of elevated CO2 in patients with asthma (Figure 2). Mechanical ventilation with “permissive hypercapnia” for severe asthma is currently an accepted therapeutic strategy [22,70]. The “permissive hypercapnia” approach is based on observational reports from the 1980s to 1990s [9,10,18,21] and is generally well tolerated for short periods of time if oxygenation is preserved and severe respiratory acidosis is avoided [70]. However, more recent studies challenge the “permissive hypercapnia” approach in view of new evidence suggesting that hypercapnia is harmful [23,26,27,136,137]. In recent years, noninvasive positive pressure ventilation (NPPV) aimed at correcting elevated PaCO2 values has been shown to be beneficial in patients with obstructive lung diseases and hypercapnia [26,136,137,138,139]. The institution of NPPV in hypercapnic patients with COPD improved outcomes: mortality, pulmonary function and health related quality of life [26,136,137]. In patients with asthma, a recent analysis of a national database documented increasing use of NPPV for life-threatening asthma and a concomitant decrease in use of invasive mechanical ventilation [140]. There is growing evidence reporting beneficial effects of NPPV by reducing hypercapnia, which is associated with improved pulmonary function and reduction in in-hospital mortality in asthmatic patients [138,139]. Recent preclinical and clinical studies of hypercapnia describe the mechanisms that underlie the benefits of reducing hypercapnia. Our review has summarized the data that could provide a guidance for re-assessment of the current paradigms of treatment and management in patients with asthma and hypercapnia.

Author Contributions

Conceptualization, M.S.; writing—original draft preparation, M.S.; writing—review and editing, M.S., T.H., J.I.S. All authors have read and agreed to the submitted version of the manuscript.

Funding

This review was supported in part by the US National Institutes of Health (HL-147070).

Acknowledgments

The authors thank Lynn C. Welch for support and insightful discussions. T.H. is funded in part by JSPS KAKENHI (grant number: 18K10654) and by the Environment Research and Technology Development Fund (year 2019 to 2021) of the Environmental Restoration and Conservation Agency (ERCA) of Japan.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. GINA 2020. Available online: www.ginasthma.org (accessed on 11 September 2020).
  2. World Health Organization. Asthma. Available online: https://www.who.int/news-room/q-a-detail/asthma (accessed on 11 September 2020).
  3. Scala, R. Noninvasive ventilation in severe acute asthma? Still far from the truth. Respir. Care 2010, 55, 630–637. [Google Scholar] [PubMed]
  4. Green, E.; Jain, P.; Bernoth, M. Noninvasive ventilation for acute exacerbations of asthma: A systematic review of the literature. Aust. Crit. Care 2017, 30, 289–297. [Google Scholar] [CrossRef] [PubMed]
  5. Gupta, D.; Keogh, B.; Chung, K.F.; Ayres, J.G.; Harrison, D.A.; Goldfrad, C.; Brady, A.R.; Rowan, K. Characteristics and outcome for admissions to adult, general critical care units with acute severe asthma: A secondary analysis of the ICNARC Case Mix Programme Database. Crit. Care 2004, 8, R112. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  6. Stow, P.J.; Pilcher, D.; Wilson, J.; George, C.; Bailey, M.; Higlett, T.; Bellomo, R.; Hart, G.K.; the Australian & New Zealand Intensive Care Society Adult Patient Database Management Committee. Improved outcomes from acute severe asthma in Australian intensive care units (1996–2003). Thorax 2007, 62, 842–847. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  7. Shigemura, M.; Lecuona, E.; Sznajder, J.I. Effects of hypercapnia on the lung. J. Physiol. 2017, 595, 2431–2437. [Google Scholar] [CrossRef] [Green Version]
  8. McFadden, E.R., Jr.; Lyons, H.A. Arterial-blood gas tension in asthma. N. Engl. J. Med. 1968, 278, 1027–1032. [Google Scholar] [CrossRef]
  9. Menitove, S.M.; Goldring, R.M. Combined ventilator and bicarbonate strategy in the management of status asthmaticus. Am. J. Med. 1983, 74, 898–901. [Google Scholar] [CrossRef]
  10. Darioli, R.; Perret, C. Mechanical controlled hypoventilation in status asthmaticus. Am. Rev. Respir. Dis. 1984, 129, 385–387. [Google Scholar]
  11. Mountain, R.D.; Sahn, S.A. Clinical features and outcome in patients with acute asthma presenting with hypercapnia. Am. Rev. Respir. Dis. 1988, 138, 535–539. [Google Scholar] [CrossRef]
  12. Mutlu, G.M.; Factor, P.; Schwartz, D.E.; Sznajder, J.I. Severe status asthmaticus: Management with permissive hypercapnia and inhalation anesthesia. Crit. Care Med. 2002, 30, 477–480. [Google Scholar] [CrossRef] [Green Version]
  13. Adnet, F.; Plaisance, P.; Borron, S.W.; Levy, A.; Payen, D. Prolonged severe hypercapnia complicating near fatal asthma in a 35-year-old woman. Intensive Care Med. 1998, 24, 1335–1338. [Google Scholar] [CrossRef] [PubMed]
  14. Mazzeo, A.T.; Spada, A.; Pratico, C.; Lucanto, T.; Santamaria, L.B. Hypercapnia: What is the limit in paediatric patients? A case of near-fatal asthma successfully treated by multipharmacological approach. Paediatr. Anaesth. 2004, 14, 596–603. [Google Scholar] [CrossRef] [PubMed]
  15. Edmunds, S.M.; Harrison, R. Subarachnoid hemorrhage in a child with status asthmaticus: Significance of permissive hypercapnia. Pediatr. Crit. Care Med. 2003, 4, 100–103. [Google Scholar] [CrossRef] [PubMed]
  16. Masuda, Y.; Tatsumi, H.; Goto, K.; Imaizumi, H.; Yoshida, S.; Kimijima, T.; Yamakage, M. Treatment of life-threatening hypercapnia with isoflurane in an infant with status asthmaticus. J. Anesth. 2014, 28, 610–612. [Google Scholar] [CrossRef] [PubMed]
  17. Zender, H.O.; Eggimann, P.; Bulpa, P.; Chevrolet, J.C.; Jolliet, P. Quadriparesia following permissive hypercapnia and inhalational anesthesia in a patient with severe status asthmaticus. Intensive Care Med. 1996, 22, 1001. [Google Scholar] [CrossRef]
  18. Hickling, K.G.; Walsh, J.; Henderson, S.; Jackson, R. Low mortality rate in adult respiratory distress syndrome using low-volume, pressure-limited ventilation with permissive hypercapnia: A prospective study. Crit. Care Med. 1994, 22, 1568–1578. [Google Scholar] [CrossRef]
  19. Laffey, J.G.; Kavanagh, B.P. Carbon Dioxide and the Critically Ill—Too Little of a Good Thing? Lancet 1999, 354, 1283–1286. [Google Scholar] [CrossRef]
  20. ARDS Network. Ventilation with lower tidal volumes as compared with traditional tidal volumes for acute lung injury and the acute respiratory distress syndrome. The Acute Respiratory Distress Syndrome Network. N. Engl. J. Med. 2000, 342, 1301–1308. [Google Scholar] [CrossRef]
  21. Amato, M.B.; Barbas, C.S.; Medeiros, D.M.; Magaldi, R.B.; Schettino, G.P.; Lorenzi-Filho, G.; Kairalla, R.A.; Deheinzelin, D.; Munoz, C.; Oliveira, R.; et al. Effect of a protective-ventilation strategy on mortality in the acute respiratory distress syndrome. N. Engl. J. Med. 1998, 338, 347–354. [Google Scholar] [CrossRef]
  22. Contreras, M.; Masterson, C.; Laffey, J.G. Permissive hypercapnia: What to remember. Curr. Opin. Anaesthesiol. 2015, 28, 26–37. [Google Scholar] [CrossRef]
  23. Nin, N.; Muriel, A.; Penuelas, O.; Brochard, L.; Lorente, J.A.; Ferguson, N.D.; Raymondos, K.; Rios, F.; Violi, D.A.; Thille, A.W.; et al. Severe hypercapnia and outcome of mechanically ventilated patients with moderate or severe acute respiratory distress syndrome. Intensive Care Med. 2017, 43, 200–208. [Google Scholar] [CrossRef] [PubMed]
  24. Vadasz, I.; Dada, L.A.; Briva, A.; Trejo, H.E.; Welch, L.C.; Chen, J.; Toth, P.T.; Lecuona, E.; Witters, L.A.; Schumacker, P.T.; et al. AMP-activated protein kinase regulates CO2-induced alveolar epithelial dysfunction in rats and human cells by promoting Na,K-ATPase endocytosis. J. Clin. Investig. 2008, 118, 752–762. [Google Scholar] [CrossRef] [PubMed]
  25. Gates, K.L.; Howell, H.A.; Nair, A.; Vohwinkel, C.U.; Welch, L.C.; Beitel, G.J.; Hauser, A.R.; Sznajder, J.I.; Sporn, P.H. Hypercapnia impairs lung neutrophil function and increases mortality in murine pseudomonas pneumonia. Am. J. Respir. Cell Mol. Biol. 2013, 49, 821–828. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  26. Shigemura, M.; Lecuona, E.; Angulo, M.; Homma, T.; Rodriguez, D.A.; Gonzalez-Gonzalez, F.J.; Welch, L.C.; Amarelle, L.; Kim, S.J.; Kaminski, N.; et al. Hypercapnia increases airway smooth muscle contractility via caspase-7-mediated miR-133a-RhoA signaling. Sci. Transl. Med. 2018, 10, eaat1662. [Google Scholar] [CrossRef] [Green Version]
  27. Bharat, A.; Graf, N.; Mullen, A.; Kanter, J.; Andrei, A.C.; Sporn, P.H.; DeCamp, M.M.; Sznajder, J.I. Pleural Hypercarbia After Lung Surgery Is Associated with Persistent Alveolopleural Fistulae. Chest 2016, 149, 220–227. [Google Scholar] [CrossRef] [Green Version]
  28. Bharat, A.; Angulo, M.; Sun, H.; Akbarpour, M.; Alberro, A.; Cheng, Y.; Shigemura, M.; Berdnikov, S.; Welch, L.C.; Kanter, J.A.; et al. High CO2 Levels Impair Lung Wound Healing. Am. J. Respir. Cell Mol. Biol. 2020, 63, 244–254. [Google Scholar] [CrossRef]
  29. Casalino-Matsuda, S.M.; Chen, F.; Gonzalez-Gonzalez, F.J.; Nair, A.; Dib, S.; Yemelyanov, A.; Gates, K.L.; Budinger, G.R.S.; Beitel, G.J.; Sporn, P.H.S. Hypercapnia Suppresses Macrophage Antiviral Activity and Increases Mortality of Influenza A Infection via Akt1. J. Immunol. 2020, 205, 489–501. [Google Scholar] [CrossRef]
  30. Jaitovich, A.; Angulo, M.; Lecuona, E.; Dada, L.A.; Welch, L.C.; Cheng, Y.; Gusarova, G.; Ceco, E.; Liu, C.; Shigemura, M.; et al. High CO2 levels cause skeletal muscle atrophy via AMP-activated kinase (AMPK), FoxO3a protein, and muscle-specific Ring finger protein 1 (MuRF1). J. Biol. Chem. 2015, 290, 9183–9194. [Google Scholar] [CrossRef] [Green Version]
  31. Korponay, T.C.; Balnis, J.; Vincent, C.E.; Singer, D.V.; Chopra, A.; Adam, A.P.; Ginnan, R.; Singer, H.A.; Jaitovich, A. High CO2 Downregulates Skeletal Muscle Protein Anabolism via AMP-activated Protein Kinase alpha2-mediated Depressed Ribosomal Biogenesis. Am. J. Respir. Cell Mol. Biol. 2020, 62, 74–86. [Google Scholar] [CrossRef]
  32. Balnis, J.; Korponay, T.C.; Jaitovich, A. AMP-Activated Protein Kinase (AMPK) at the Crossroads Between CO2 Retention and Skeletal Muscle Dysfunction in Chronic Obstructive Pulmonary Disease (COPD). Int. J. Mol. Sci. 2020, 21, 955. [Google Scholar] [CrossRef] [Green Version]
  33. Wang, N.; Gates, K.L.; Trejo, H.; Favoreto, S., Jr.; Schleimer, R.P.; Sznajder, J.I.; Beitel, G.J.; Sporn, P.H. Elevated CO2 selectively inhibits interleukin-6 and tumor necrosis factor expression and decreases phagocytosis in the macrophage. FASEB J. 2010, 24, 2178–2190. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  34. Cummins, E.P.; Oliver, K.M.; Lenihan, C.R.; Fitzpatrick, S.F.; Bruning, U.; Scholz, C.C.; Slattery, C.; Leonard, M.O.; McLoughlin, P.; Taylor, C.T. NF-kappaB links CO2 sensing to innate immunity and inflammation in mammalian cells. J. Immunol. 2010, 185, 4439–4445. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  35. Oliver, K.M.; Lenihan, C.R.; Bruning, U.; Cheong, A.; Laffey, J.G.; McLoughlin, P.; Taylor, C.T.; Cummins, E.P. Hypercapnia induces cleavage and nuclear localization of RelB protein, giving insight into CO2 sensing and signaling. J. Biol. Chem. 2012, 287, 14004–14011. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  36. Casalino-Matsuda, S.M.; Nair, A.; Beitel, G.J.; Gates, K.L.; Sporn, P.H. Hypercapnia Inhibits Autophagy and Bacterial Killing in Human Macrophages by Increasing Expression of Bcl-2 and Bcl-xL. J. Immunol. 2015, 194, 5388–5396. [Google Scholar] [CrossRef] [Green Version]
  37. Lu, Z.; Casalino-Matsuda, S.M.; Nair, A.; Buchbinder, A.; Budinger, G.R.S.; Sporn, P.H.S.; Gates, K.L. A role for heat shock factor 1 in hypercapnia-induced inhibition of inflammatory cytokine expression. FASEB J. 2018, 32, 3614–3622. [Google Scholar] [CrossRef] [Green Version]
  38. Li, G.; Zhou, D.; Vicencio, A.G.; Ryu, J.; Xue, J.; Kanaan, A.; Gavrialov, O.; Haddad, G.G. Effect of carbon dioxide on neonatal mouse lung: A genomic approach. J. Appl. Physiol. 2006, 101, 1556–1564. [Google Scholar] [CrossRef] [Green Version]
  39. Casalino-Matsuda, S.M.; Wang, N.; Ruhoff, P.T.; Matsuda, H.; Nlend, M.C.; Nair, A.; Szleifer, I.; Beitel, G.J.; Sznajder, J.I.; Sporn, P.H.S. Hypercapnia Alters Expression of Immune Response, Nucleosome Assembly and Lipid Metabolism Genes in Differentiated Human Bronchial Epithelial Cells. Sci. Rep. 2018, 8, 13508. [Google Scholar] [CrossRef]
  40. Shigemura, M.; Lecuona, E.; Angulo, M.; Dada, L.A.; Edwards, M.B.; Welch, L.C.; Casalino-Matsuda, S.M.; Sporn, P.H.S.; Vadasz, I.; Helenius, I.T.; et al. Elevated CO2 regulates the Wnt signaling pathway in mammals, Drosophila melanogaster and Caenorhabditis elegans. Sci. Rep. 2019, 9, 18251. [Google Scholar] [CrossRef] [Green Version]
  41. Briva, A.; Vadasz, I.; Lecuona, E.; Welch, L.C.; Chen, J.; Dada, L.A.; Trejo, H.E.; Dumasius, V.; Azzam, Z.S.; Myrianthefs, P.M.; et al. High CO2 levels impair alveolar epithelial function independently of pH. PLoS ONE 2007, 2, e1238. [Google Scholar] [CrossRef]
  42. Vadasz, I.; Dada, L.A.; Briva, A.; Helenius, I.T.; Sharabi, K.; Welch, L.C.; Kelly, A.M.; Grzesik, B.A.; Budinger, G.R.; Liu, J.; et al. Evolutionary conserved role of c-Jun-N-terminal kinase in CO2-induced epithelial dysfunction. PLoS ONE 2012, 7, e46696. [Google Scholar] [CrossRef]
  43. Welch, L.C.; Lecuona, E.; Briva, A.; Trejo, H.E.; Dada, L.A.; Sznajder, J.I. Extracellular signal-regulated kinase (ERK) participates in the hypercapnia-induced Na, K-ATPase downregulation. FEBS Lett. 2010, 584, 3985–3989. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  44. Dada, L.A.; Trejo Bittar, H.E.; Welch, L.C.; Vagin, O.; Deiss-Yehiely, N.; Kelly, A.M.; Baker, M.R.; Capri, J.; Cohn, W.; Whitelegge, J.P.; et al. High CO2 Leads to Na,K-ATPase Endocytosis via c-Jun Amino-Terminal Kinase-Induced LMO7b Phosphorylation. Mol. Cell. Biol. 2015, 35, 3962–3973. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  45. Lecuona, E.; Sun, H.; Chen, J.; Trejo, H.E.; Baker, M.A.; Sznajder, J.I. Protein kinase A-Ialpha regulates Na,K-ATPase endocytosis in alveolar epithelial cells exposed to high CO(2) concentrations. Am. J. Respir. Cell Mol. Biol. 2013, 48, 626–634. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  46. Kryvenko, V.; Wessendorf, M.; Morty, R.E.; Herold, S.; Seeger, W.; Vagin, O.; Dada, L.A.; Sznajder, J.I.; Vadasz, I. Hypercapnia Impairs Na,K-ATPase Function by Inducing Endoplasmic Reticulum Retention of the beta-Subunit of the Enzyme in Alveolar Epithelial Cells. Int. J. Mol. Sci. 2020, 21, 1467. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  47. Doerr, C.H.; Gajic, O.; Berrios, J.C.; Caples, S.; Abdel, M.; Lymp, J.F.; Hubmayr, R.D. Hypercapnic acidosis impairs plasma membrane wound resealing in ventilator-injured lungs. Am. J. Respir. Crit. Care Med. 2005, 171, 1371–1377. [Google Scholar] [CrossRef] [Green Version]
  48. O’Toole, D.; Hassett, P.; Contreras, M.; Higgins, B.D.; McKeown, S.T.; McAuley, D.F.; O’Brien, T.; Laffey, J.G. Hypercapnic acidosis attenuates pulmonary epithelial wound repair by an NF-kappaB dependent mechanism. Thorax 2009, 64, 976–982. [Google Scholar] [CrossRef] [Green Version]
  49. Vohwinkel, C.U.; Lecuona, E.; Sun, H.; Sommer, N.; Vadász, I.; Chandel, N.S.; Sznajder, J.I. Elevated CO2 levels cause mitochondrial dysfunction and impair cell proliferation. J. Biol. Chem. 2011, 286, 37067–37076. [Google Scholar] [CrossRef] [Green Version]
  50. Cummins, E.P.; Strowitzki, M.J.; Taylor, C.T. Mechanisms and Consequences of Oxygen and Carbon Dioxide Sensing in Mammals. Physiol. Rev. 2020, 100, 463–488. [Google Scholar] [CrossRef]
  51. Monastersky, R. Global carbon dioxide levels near worrisome milestone. Nature 2013, 497, 13–14. [Google Scholar] [CrossRef] [Green Version]
  52. The Keeling Curve. Available online: https://scripps.ucsd.edu/programs/keelingcurve/ (accessed on 11 September 2020).
  53. Missner, A.; Pohl, P. 110 years of the Meyer-Overton rule: Predicting membrane permeability of gases and other small compounds. ChemPhysChem 2009, 10, 1405–1414. [Google Scholar] [CrossRef] [Green Version]
  54. Verkman, A.S. Role of aquaporins in lung liquid physiology. Respir. Physiol. Neurobiol. 2007, 159, 324–330. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  55. Musa-Aziz, R.; Chen, L.M.; Pelletier, M.F.; Boron, W.F. Relative CO2/NH3 selectivities of AQP1, AQP4, AQP5, AmtB, and RhAG. Proc. Natl. Acad. Sci. USA 2009, 106, 5406–5411. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  56. Endeward, V.; Cartron, J.P.; Ripoche, P.; Gros, G. RhAG protein of the Rhesus complex is a CO2 channel in the human red cell membrane. FASEB J. 2008, 22, 64–73. [Google Scholar] [CrossRef] [PubMed]
  57. Casey, J.R.; Grinstein, S.; Orlowski, J. Sensors and regulators of intracellular pH. Nat. Rev. Mol. Cell Biol. 2010, 11, 50–61. [Google Scholar] [CrossRef]
  58. Mitchell, R.A. Respiratory chemosensitivity in the medulla oblongata. J. Physiol. 1969, 202, 3P–4P. [Google Scholar]
  59. Mulkey, D.K.; Stornetta, R.L.; Weston, M.C.; Simmons, J.R.; Parker, A.; Bayliss, D.A.; Guyenet, P.G. Respiratory control by ventral surface chemoreceptor neurons in rats. Nat. Neurosci. 2004, 7, 1360–1369. [Google Scholar] [CrossRef]
  60. Richerson, G.B.; Wang, W.; Tiwari, J.; Bradley, S.R. Chemosensitivity of serotonergic neurons in the rostral ventral medulla. Respir. Physiol. 2001, 129, 175–189. [Google Scholar] [CrossRef]
  61. Wang, W.; Richerson, G.B. Development of chemosensitivity of rat medullary raphe neurons. Neuroscience 1999, 90, 1001–1011. [Google Scholar] [CrossRef]
  62. Xu, F.; Frazier, D.T. Role of the cerebellar deep nuclei in respiratory modulation. Cerebellum 2002, 1, 35–40. [Google Scholar] [CrossRef]
  63. Smith, C.A.; Blain, G.M.; Henderson, K.S.; Dempsey, J.A. Peripheral chemoreceptors determine the respiratory sensitivity of central chemoreceptors to CO2: Role of carotid body CO2. J. Physiol. 2015, 593, 4225–4243. [Google Scholar] [CrossRef] [Green Version]
  64. Bayliss, D.A.; Barhanin, J.; Gestreau, C.; Guyenet, P.G. The role of pH-sensitive TASK channels in central respiratory chemoreception. Pflugers Arch. 2015, 467, 917–929. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  65. Lesage, F.; Barhanin, J. Molecular physiology of pH-sensitive background K2P channels. Physiology 2011, 26, 424–437. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  66. Cui, N.; Zhang, X.; Tadepalli, J.S.; Yu, L.; Gai, H.; Petit, J.; Pamulapati, R.T.; Jin, X.; Jiang, C. Involvement of TRP channels in the CO2 chemosensitivity of locus coeruleus neurons. J. Neurophysiol. 2011, 105, 2791–2801. [Google Scholar] [CrossRef] [PubMed]
  67. Huckstepp, R.T.; id Bihi, R.; Eason, R.; Spyer, K.M.; Dicke, N.; Willecke, K.; Marina, N.; Gourine, A.V.; Dale, N. Connexin hemichannel-mediated CO2-dependent release of ATP in the medulla oblongata contributes to central respiratory chemosensitivity. J. Physiol. 2010, 588, 3901–3920. [Google Scholar] [CrossRef]
  68. Meigh, L.; Greenhalgh, S.A.; Rodgers, T.L.; Cann, M.J.; Roper, D.I.; Dale, N. CO2 directly modulates connexin 26 by formation of carbamate bridges between subunits. Elife 2013, 2, e01213. [Google Scholar] [CrossRef] [Green Version]
  69. Sullivan, W.J.; Dorman, P.J. The renal response to chronic respiratory acidosis. J. Clin. Investig. 1955, 34, 268–276. [Google Scholar] [CrossRef] [Green Version]
  70. Feihl, F.; Perret, C. Permissive hypercapnia. How permissive should we be? Am. J. Respir. Crit. Care Med. 1994, 150, 1722–1737. [Google Scholar] [CrossRef]
  71. Louie, S.; Morrissey, B.M.; Kenyon, N.J.; Albertson, T.E.; Avdalovic, M. The critically ill asthmatic—From ICU to discharge. Clin. Rev. Allergy Immunol. 2012, 43, 30–44. [Google Scholar] [CrossRef]
  72. Elsayegh, D.; Saito, S.; Eden, E.; Shapiro, J. Increasing severity of status asthmaticus in an urban medical intensive care unit. J. Hosp. Med. 2008, 3, 206–211. [Google Scholar] [CrossRef]
  73. Lumb, A.B. Nunn’s Applied Respiratory Physiology; foreword by Ronald Pearl; Elsevier: Edinburgh, UK, 2017. [Google Scholar]
  74. Rodrigo, C.; Rodrigo, G. Subarachnoid hemorrhage following permissive hypercapnia in a patient with severe acute asthma. Am. J. Emerg. Med. 1999, 17, 697–699. [Google Scholar] [CrossRef]
  75. Tang, W.C.; Weil, M.H.; Gazmuri, R.J.; Bisera, J.; Rackow, E.C. Reversible impairment of myocardial contractility due to hypercarbic acidosis in the isolated perfused rat heart. Crit. Care Med. 1991, 19, 218–224. [Google Scholar] [CrossRef] [PubMed]
  76. Einthoven, W. Ueber die Wirkung der Bronchialmuskeln, nach einer neuen Methode untersucht, und über Asthma nervosum. Arch. Gesamte Physiol. Menschen Tiere 1892, 51, 367–445. [Google Scholar] [CrossRef]
  77. De Burgh Daly, M.; Lambertsen, D.C.; Schweitzer, A. The effects upon the bronchial musculature of altering the oxygen and carbon dioxide tensions of the blood perfusing the brain. J. Physiol. 1953, 119, 292–314. [Google Scholar] [CrossRef] [PubMed]
  78. Loofbourrow, G.N.; Wood, W.B.; Baird, I.L. Tracheal constriction in the dog. Am. J. Physiol. 1957, 191, 411–415. [Google Scholar] [CrossRef] [PubMed]
  79. Nadel, J.A.; Widdicombe, J.G. Effect of changes in blood gas tensions and carotid sinus pressure on tracheal volume and total lung resistance to airflow. J. Physiol. 1962, 163, 13–33. [Google Scholar] [CrossRef] [Green Version]
  80. Green, M.; Widdicombe, J.G. The effects of ventilation of dogs with different gas mixtures on airway calibre and lung mechanics. J. Physiol. 1966, 186, 363–381. [Google Scholar] [CrossRef] [Green Version]
  81. Kondo, T.; Kobayashi, I.; Hayama, N.; Tazaki, G.; Ohta, Y. Role of cholinergic neural transmission on airway resistance in the dog. J. Auton. Nerv. Syst. 2000, 80, 64–70. [Google Scholar] [CrossRef]
  82. Dixon, W.E. Contributions to the physiology of the lungs: Part I. The bronchial muscles, their innervation, and the action of drugs upon them. J. Physiol. 1903, 29, 97–173. [Google Scholar] [CrossRef]
  83. Iscoe, S.; Fisher, J.T. Bronchomotor responses to hypoxia and hypercapnia in decerebrate cats. J. Appl. Physiol. 1995, 78, 117–123. [Google Scholar] [CrossRef]
  84. Sterling, G.M. The mechanism of decreased specific airway conductance in man during hypercapnia caused by inhalation of 7 percent CO2. Clin. Sci. 1969, 37, 539–548. [Google Scholar]
  85. Rodarte, J.R.; Hyatt, R.E. Effect of acute exposure to CO2 on lung mechanics in normal man. Respir. Physiol. 1973, 17, 135–145. [Google Scholar] [CrossRef]
  86. Severinghaus, J.W.; Swenson, E.W.; Finley, T.N.; Lategola, M.T.; Williams, J. Unilateral hypoventilation produced in dogs by occluding one pulmonary artery. J. Appl. Physiol. 1961, 16, 53–60. [Google Scholar] [CrossRef] [PubMed]
  87. Darke, C.S.; Astin, T.W. Differential ventilation in unilateral pulmonary artery occlusion. Thorax 1972, 27, 480–486. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  88. Astin, T.W.; Barer, G.R.; Shaw, J.W.; Warren, P.M. The action of carbon dioxide on constricted airways. J. Physiol. 1973, 235, 607–623. [Google Scholar] [CrossRef]
  89. Sterling, G.M.; Holst, P.E.; Nadel, J.A. Effect of CO2 and pH on bronchoconstriction caused by serotonin vs. acetylcholine. J. Appl. Physiol. 1972, 32, 39–43. [Google Scholar] [CrossRef]
  90. Duckles, S.P.; Rayner, M.D.; Nadel, J.A. Effects of CO2 and pH on drug-induced contractions of airway smooth muscle. J. Pharmacol. Exp. Ther. 1974, 190, 472–481. [Google Scholar]
  91. Fisher, H.K.; Holton, P.; Buxton, R.S.; Nadel, J.A. Resistance to breathing during exercise-induced asthma attacks. Am. Rev. Respir. Dis. 1970, 101, 885–896. [Google Scholar]
  92. Fisher, H.K.; Hansen, T.A. Site of action of inhaled 6 per cent carbon dioxide in the lungs of asthmatic subjects before and after exercise. Am. Rev. Respir. Dis. 1976, 114, 861–870. [Google Scholar]
  93. Stephens, N.L.; Meyers, J.L.; Cherniack, R.M. Oxygen, carbon dioxide, H+ ion, and bronchial length-tension relationships. J. Appl. Physiol. 1968, 25, 376–383. [Google Scholar] [CrossRef]
  94. Croxton, T.L.; Lande, B.; Hirshman, C.A. Role of intracellular pH in relaxation of porcine tracheal smooth muscle by respiratory gases. Am. J. Physiol. 1995, 268, L207–L213. [Google Scholar] [CrossRef]
  95. Twort, C.H.; Cameron, I.R. Effects of PCO2, pH and extracellular calcium on contraction of airway smooth muscle from rats. Respir. Physiol. 1986, 66, 259–267. [Google Scholar] [CrossRef]
  96. Yamakage, M.; Lindeman, K.S.; Hirshman, C.A.; Croxton, T.L. Intracellular pH regulates voltage-dependent Ca2+ channels in porcine tracheal smooth muscle cells. Am. J. Physiol. 1995, 268, L642–L646. [Google Scholar] [CrossRef] [PubMed]
  97. El Mays, T.Y.; Saifeddine, M.; Choudhury, P.; Hollenberg, M.D.; Green, F.H. Carbon dioxide enhances substance P-induced epithelium-dependent bronchial smooth muscle relaxation in Sprague-Dawley rats. Can. J. Physiol. Pharmacol. 2011, 89, 513–520. [Google Scholar] [CrossRef] [PubMed]
  98. Cabezas, G.A.; Graf, P.D.; Nadel, J.A. Sympathetic versus parasympathetic nervous regulation of airways in dogs. J. Appl. Physiol. 1971, 31, 651–655. [Google Scholar] [CrossRef]
  99. Colebatch, H.J.; Halmagyi, D.F. Effect of Vagotomy and Vagal Stimulation on Lung Mechanics and Circulation. J. Appl. Physiol. 1963, 18, 881–887. [Google Scholar] [CrossRef]
  100. Woolcock, A.J.; Macklem, P.T.; Hogg, J.C.; Wilson, N.J.; Nadel, J.A.; Frank, N.R.; Brain, J. Effect of vagal stimulation on central and peripheral airways in dogs. J. Appl. Physiol. 1969, 26, 806–813. [Google Scholar] [CrossRef]
  101. Campbell, C.J.; Murtagh, J.A.; Raber, C.F. Chemical Agents and Reflex Control of Laryngeal Glottis. Ann. Otol. Rhinol. Laryngol. 1963, 72, 589–604. [Google Scholar] [CrossRef]
  102. Spann, R.W.; Hyatt, R.E. Factors affecting upper airway resistance in conscious man. J. Appl. Physiol. 1971, 31, 708–712. [Google Scholar] [CrossRef]
  103. Yamakage, M.; Kohro, S.; Yamauchi, M.; Namiki, A. The effects of extracellular pH on intracellular pH, Ca2+ and tension of canine tracheal smooth muscle strips. Life Sci. 1995, 56, PL175–PL180. [Google Scholar] [CrossRef]
  104. Wark, P.A.; Tooze, M.; Powell, H.; Parsons, K. Viral and bacterial infection in acute asthma and chronic obstructive pulmonary disease increases the risk of readmission. Respirology 2013, 18, 996–1002. [Google Scholar] [CrossRef]
  105. Oliver, B.G.; Robinson, P.; Peters, M.; Black, J. Viral infections and asthma: An inflammatory interface? Eur. Respir. J. 2014, 44, 1666–1681. [Google Scholar] [CrossRef] [PubMed]
  106. Iikura, M.; Hojo, M.; Koketsu, R.; Watanabe, S.; Sato, A.; Chino, H.; Ro, S.; Masaki, H.; Hirashima, J.; Ishii, S.; et al. The importance of bacterial and viral infections associated with adult asthma exacerbations in clinical practice. PLoS ONE 2015, 10, e0123584. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  107. Resiliac, J.; Grayson, M.H. Epidemiology of Infections and Development of Asthma. Immunol. Allergy Clin. N. Am. 2019, 39, 297–307. [Google Scholar] [CrossRef] [PubMed]
  108. Laserna, E.; Sibila, O.; Aguilar, P.R.; Mortensen, E.M.; Anzueto, A.; Blanquer, J.M.; Sanz, F.; Rello, J.; Marcos, P.J.; Velez, M.I.; et al. Hypocapnia and hypercapnia are predictors for ICU admission and mortality in hospitalized patients with community-acquired pneumonia. Chest 2012, 142, 1193–1199. [Google Scholar] [CrossRef] [Green Version]
  109. Belkin, R.A.; Henig, N.R.; Singer, L.G.; Chaparro, C.; Rubenstein, R.C.; Xie, S.X.; Yee, J.Y.; Kotloff, R.M.; Lipson, D.A.; Bunin, G.R. Risk factors for death of patients with cystic fibrosis awaiting lung transplantation. Am. J. Respir. Crit. Care Med. 2006, 173, 659–666. [Google Scholar] [CrossRef] [Green Version]
  110. Sharabi, K.; Hurwitz, A.; Simon, A.J.; Beitel, G.J.; Morimoto, R.I.; Rechavi, G.; Sznajder, J.I.; Gruenbaum, Y. Elevated CO2 levels affect development, motility, and fertility and extend life span in Caenorhabditis elegans. Proc. Natl. Acad. Sci. USA 2009, 106, 4024–4029. [Google Scholar] [CrossRef] [Green Version]
  111. Helenius, I.T.; Krupinski, T.; Turnbull, D.W.; Gruenbaum, Y.; Silverman, N.; Johnson, E.A.; Sporn, P.H.; Sznajder, J.I.; Beitel, G.J. Elevated CO2 suppresses specific Drosophila innate immune responses and resistance to bacterial infection. Proc. Natl. Acad. Sci. USA 2009, 106, 18710–18715. [Google Scholar] [CrossRef] [Green Version]
  112. Fitzpatrick, A.M.; Holguin, F.; Teague, W.G.; Brown, L.A. Alveolar macrophage phagocytosis is impaired in children with poorly controlled asthma. J. Allergy Clin. Immunol. 2008, 121, 1372–1378. [Google Scholar] [CrossRef] [Green Version]
  113. Keogh, C.E.; Scholz, C.C.; Rodriguez, J.; Selfridge, A.C.; von Kriegsheim, A.; Cummins, E.P. Carbon dioxide-dependent regulation of NF-kappaB family members RelB and p100 gives molecular insight into CO2-dependent immune regulation. J. Biol. Chem. 2017, 292, 11561–11571. [Google Scholar] [CrossRef] [Green Version]
  114. Lardner, A. The effects of extracellular pH on immune function. J. Leukoc. Biol. 2001, 69, 522–530. [Google Scholar]
  115. Lang, C.J.; Dong, P.; Hosszu, E.K.; Doyle, I.R. Effect of CO2 on LPS-induced cytokine responses in rat alveolar macrophages. Am. J. Physiol. Lung Cell Mol. Physiol. 2005, 289, L96–L103. [Google Scholar] [CrossRef] [PubMed]
  116. Wood, C.E.; Kane, C.; Raff, H. Peripheral chemoreceptor control of fetal renin responses to hypoxia and hypercapnia. Circ. Res. 1990, 67, 722–732. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  117. Yilin, Z.; Yandong, N.; Faguang, J. Role of angiotensin-converting enzyme (ACE) and ACE2 in a rat model of smoke inhalation induced acute respiratory distress syndrome. Burns 2015, 41, 1468–1477. [Google Scholar] [CrossRef] [PubMed]
  118. Zamai, L. The Yin and Yang of ACE/ACE2 Pathways: The Rationale for the Use of Renin-Angiotensin System Inhibitors in COVID-19 Patients. Cells 2020, 9, 1704. [Google Scholar] [CrossRef] [PubMed]
  119. Larsson, L.G.; Lindberg, A.; Franklin, K.A.; Lundback, B. Symptoms related to obstructive sleep apnoea are common in subjects with asthma, chronic bronchitis and rhinitis in a general population. Respir. Med. 2001, 95, 423–429. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  120. Prasad, B.; Nyenhuis, S.M.; Weaver, T.E. Obstructive sleep apnea and asthma: Associations and treatment implications. Sleep Med. Rev. 2014, 18, 165–171. [Google Scholar] [CrossRef] [PubMed]
  121. Min, Y.Z.; Subbarao, P.; Narang, I. The Bidirectional Relationship Between Asthma and Obstructive Sleep Apnea: Which Came First? J. Pediatr. 2016, 176, 10–16. [Google Scholar] [CrossRef] [PubMed]
  122. Kong, D.L.; Qin, Z.; Shen, H.; Jin, H.Y.; Wang, W.; Wang, Z.F. Association of Obstructive Sleep Apnea with Asthma: A Meta-Analysis. Sci. Rep. 2017, 7, 4088. [Google Scholar] [CrossRef] [Green Version]
  123. Redline, S.; Tishler, P.V.; Schluchter, M.; Aylor, J.; Clark, K.; Graham, G. Risk factors for sleep-disordered breathing in children. Associations with obesity, race, and respiratory problems. Am. J. Respir. Crit. Care Med. 1999, 159, 1527–1532. [Google Scholar] [CrossRef]
  124. Beuther, D.A.; Sutherland, E.R. Overweight, obesity, and incident asthma: A meta-analysis of prospective epidemiologic studies. Am. J. Respir. Crit. Care Med. 2007, 175, 661–666. [Google Scholar] [CrossRef] [Green Version]
  125. Barros, R.; Moreira, P.; Padrao, P.; Teixeira, V.H.; Carvalho, P.; Delgado, L.; Moreira, A. Obesity increases the prevalence and the incidence of asthma and worsens asthma severity. Clin. Nutr. 2017, 36, 1068–1074. [Google Scholar] [CrossRef] [PubMed]
  126. Taylor, B.; Mannino, D.; Brown, C.; Crocker, D.; Twum-Baah, N.; Holguin, F. Body mass index and asthma severity in the National Asthma Survey. Thorax 2008, 63, 14–20. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  127. Mosen, D.M.; Schatz, M.; Magid, D.J.; Camargo, C.A., Jr. The relationship between obesity and asthma severity and control in adults. J. Allergy Clin. Immunol. 2008, 122, 507–511. [Google Scholar] [CrossRef] [PubMed]
  128. Jordan, A.S.; McSharry, D.G.; Malhotra, A. Adult obstructive sleep apnoea. Lancet 2014, 383, 736–747. [Google Scholar] [CrossRef] [Green Version]
  129. Piper, A. Obesity hypoventilation syndrome: Weighing in on therapy options. Chest 2016, 149, 856–868. [Google Scholar]
  130. Kikuchi, R.; Tsuji, T.; Watanabe, O.; Yamaguchi, K.; Furukawa, K.; Nakamura, H.; Aoshiba, K. Hypercapnia Accelerates Adipogenesis: A Novel Role of High CO2 in Exacerbating Obesity. Am. J. Respir. Cell Mol. Biol. 2017, 57, 570–580. [Google Scholar] [CrossRef]
  131. Considine, R.V.; Sinha, M.K.; Heiman, M.L.; Kriauciunas, A.; Stephens, T.W.; Nyce, M.R.; Ohannesian, J.P.; Marco, C.C.; McKee, L.J.; Bauer, T.L.; et al. Serum immunoreactive-leptin concentrations in normal-weight and obese humans. N. Engl. J. Med. 1996, 334, 292–295. [Google Scholar] [CrossRef]
  132. Tankersley, C.; Kleeberger, S.; Russ, B.; Schwartz, A.; Smith, P. Modified control of breathing in genetically obese (ob/ob) mice. J. Appl. Physiol. 1996, 81, 716–723. [Google Scholar] [CrossRef]
  133. O’Donnell, C.P.; Schaub, C.D.; Haines, A.S.; Berkowitz, D.E.; Tankersley, C.G.; Schwartz, A.R.; Smith, P.L. Leptin prevents respiratory depression in obesity. Am. J. Respir. Crit. Care Med. 1999, 159, 1477–1484. [Google Scholar] [CrossRef]
  134. Caro, J.F.; Kolaczynski, J.W.; Nyce, M.R.; Ohannesian, J.P.; Opentanova, I.; Goldman, W.H.; Lynn, R.B.; Zhang, P.L.; Sinha, M.K.; Considine, R.V. Decreased cerebrospinal-fluid/serum leptin ratio in obesity: A possible mechanism for leptin resistance. Lancet 1996, 348, 159–161. [Google Scholar] [CrossRef]
  135. Campo, A.; Fruhbeck, G.; Zulueta, J.J.; Iriarte, J.; Seijo, L.M.; Alcaide, A.B.; Galdiz, J.B.; Salvador, J. Hyperleptinaemia, respiratory drive and hypercapnic response in obese patients. Eur. Respir. J. 2007, 30, 223–231. [Google Scholar] [CrossRef] [PubMed]
  136. Köhnlein, T.; Windisch, W.; Köhler, D.; Drabik, A.; Geiseler, J.; Hartl, S.; Karg, O.; Laier-Groeneveld, G.; Nava, S.; Schönhofer, B.; et al. Non-invasive positive pressure ventilation for the treatment of severe stable chronic obstructive pulmonary disease: A prospective, multicentre, randomised, controlled clinical trial. Lancet Respir. Med. 2014, 2, 698–705. [Google Scholar] [CrossRef]
  137. Murphy, P.B.; Rehal, S.; Arbane, G.; Bourke, S.; Calverley, P.M.A.; Crook, A.M.; Dowson, L.; Duffy, N.; Gibson, G.J.; Hughes, P.D.; et al. Effect of Home Noninvasive Ventilation With Oxygen Therapy vs Oxygen Therapy Alone on Hospital Readmission or Death After an Acute COPD Exacerbation: A Randomized Clinical Trial. JAMA 2017, 317, 2177–2186. [Google Scholar] [CrossRef] [PubMed]
  138. Fernandez, M.M.; Villagra, A.; Blanch, L.; Fernandez, R. Non-invasive mechanical ventilation in status asthmaticus. Intensive Care Med. 2001, 27, 486–492. [Google Scholar] [CrossRef] [PubMed]
  139. Tomii, K.; Seo, R.; Tachikawa, R.; Harada, Y.; Murase, K.; Kaji, R.; Takeshima, Y.; Hayashi, M.; Nishimura, T.; Ishihara, K. Impact of noninvasive ventilation (NIV) trial for various types of acute respiratory failure in the emergency department; decreased mortality and use of the ICU. Respir. Med. 2009, 103, 67–73. [Google Scholar] [CrossRef] [Green Version]
  140. Nanchal, R.; Kumar, G.; Majumdar, T.; Taneja, A.; Patel, J.; Dagar, G.; Jacobs, E.R.; Whittle, J. Utilization of mechanical ventilation for asthma exacerbations: Analysis of a national database. Respir. Care 2014, 59, 644–653. [Google Scholar] [CrossRef]
Figure 1. Hypercapnia increases airway and respiratory resistance in patients with chronic obstructive pulmonary disease (COPD). (A) Comparison of airway resistance (sRtot) measured by plethysmographic assessment between normocapnic and hypercapnic patients. (B) Comparison of respiratory resistance measured by impulse oscillometry (IOS) between normocapnic and hypercapnic patients. Values of R5, R20, and R5-R20 indicate total, proximal, and peripheral respiratory resistance, respectively. (C) Changes of respiratory resistance in hypercapnic patients. Reproduced from [26]. Copyright © 2018 American Association for the Advancement of Science.
Figure 1. Hypercapnia increases airway and respiratory resistance in patients with chronic obstructive pulmonary disease (COPD). (A) Comparison of airway resistance (sRtot) measured by plethysmographic assessment between normocapnic and hypercapnic patients. (B) Comparison of respiratory resistance measured by impulse oscillometry (IOS) between normocapnic and hypercapnic patients. Values of R5, R20, and R5-R20 indicate total, proximal, and peripheral respiratory resistance, respectively. (C) Changes of respiratory resistance in hypercapnic patients. Reproduced from [26]. Copyright © 2018 American Association for the Advancement of Science.
Jcm 09 03207 g001
Figure 2. Schematic summarizing potential detrimental effects of hypercapnia in patients with severe asthma. Recent discoveries indicate that hypercapnia increases airway contractility, impairs the innate immune response, and promotes adipogenesis, which likely underlies the negative effects of elevated carbon dioxide (CO2) on airway contraction, respiratory infections and obesity in patients with asthma.
Figure 2. Schematic summarizing potential detrimental effects of hypercapnia in patients with severe asthma. Recent discoveries indicate that hypercapnia increases airway contractility, impairs the innate immune response, and promotes adipogenesis, which likely underlies the negative effects of elevated carbon dioxide (CO2) on airway contraction, respiratory infections and obesity in patients with asthma.
Jcm 09 03207 g002

Share and Cite

MDPI and ACS Style

Shigemura, M.; Homma, T.; Sznajder, J.I. Hypercapnia: An Aggravating Factor in Asthma. J. Clin. Med. 2020, 9, 3207. https://doi.org/10.3390/jcm9103207

AMA Style

Shigemura M, Homma T, Sznajder JI. Hypercapnia: An Aggravating Factor in Asthma. Journal of Clinical Medicine. 2020; 9(10):3207. https://doi.org/10.3390/jcm9103207

Chicago/Turabian Style

Shigemura, Masahiko, Tetsuya Homma, and Jacob I Sznajder. 2020. "Hypercapnia: An Aggravating Factor in Asthma" Journal of Clinical Medicine 9, no. 10: 3207. https://doi.org/10.3390/jcm9103207

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop