Skip to main content

Impact of hyperoxia on the gut during critical illnesses

Abstract

Molecular oxygen is typically delivered to patients via oxygen inhalation or extracorporeal membrane oxygenation (ECMO), potentially resulting in systemic hyperoxia from liberal oxygen inhalation or localized hyperoxia in the lower body from peripheral venoarterial (VA) ECMO. Consequently, this exposes the gastrointestinal tract to excessive oxygen levels. Hyperoxia can trigger organ damage due to the overproduction of reactive oxygen species and is associated with increased mortality. The gut and gut microbiome play pivotal roles in critical illnesses and even small variations in oxygen levels can have a dramatic influence on the physiology and ecology of gut microbes. Here, we reviewed the emerging preclinical evidence which highlights how excessive inhaled oxygen can provoke diffuse villous damage, barrier dysfunction in the gut, and gut dysbiosis. The hallmark of this dysbiosis includes the expansion of oxygen-tolerant pathogens (e.g., Enterobacteriaceae) and the depletion of beneficial oxygen-intolerant microbes (e.g., Muribaculaceae). Furthermore, we discussed potential impact of oxygen on the gut in various underlying critical illnesses involving inspiratory oxygen and peripheral VA-ECMO. Currently, the available findings in this area are somewhat controversial, and a consensus has not yet to be reached. It appears that targeting near-physiological oxygenation levels may offer a means to avoid hyperoxia-induced gut injury and hypoxia-induced mesenteric ischemia. However, the optimal oxygenation target may vary depending on special clinical conditions, including acute hypoxia in adults and neonates, as well as particular patients undergoing gastrointestinal surgery or VA-ECMO support. Last, we outlined the current challenges and the need for future studies in this area. Insights into this vital ongoing research can assist clinicians in optimizing oxygenation for critically ill patients.

Background

The administration of supplemental oxygen has become one of the most commonly utilized therapies in the intensive care units (ICUs) worldwide [1, 2]. Molecular oxygen has been recognized as a “friend and foe” in treatment of critically ill patients since its discovery in the eighteenth century [3]. On the one hand, it is important for mitochondrial aerobic respiration in treating hypoxic patients; on the other hand, supplemental oxygen (e.g., oxygen inhalation and ECMO) may expose patients to supraphysiological oxygen levels, leading to arterial hyperoxia (defined as arterial oxygen partial pressure (PaO2) > 100 mmHg) [4, 5]. Arterial hyperoxia can provoke multiple organ injuries including the lung, retia, heart, and gut [6,7,8,9] and contribute to significantly increased mortality [10, 11].

The gastrointestinal tract is lined with a single-cell layer epithelium which has a surface area of 30 m2, roughly equivalent to half the size of a standard badminton court [12]. The gut houses trillions of diverse and dynamic microorganisms, termed gut microbiome (GM) [13]. Among these microorganisms, the predominant members consist of obligate anaerobes, including the classes Clostridia (Phylum Firmicutes) and Bacteroidia (Phylum Bacteroidetes), which together constitute more than 90% of the entire gut microbes [14]. It is important to note that the intestinal lumen represents an oxygen-deprived environment with an extremely low oxygen tension level, typically measuring less than 1 mmHg. This condition can be altered by the introduction of oxygen through inhalation [15]. Such an increase in oxygen availability has the potential to disrupt the stability of gut-associated microbial community and may lead to an uncontrolled expansion of oxygen-tolerant Enterobacteriaceae (phylum Proteobacteria), which is commonly recognized as a marker of dysbiosis [16, 17].

The gut and GM play key roles in the development, maintenance, and outcomes of sepsis and multiple organ dysfunction syndrome (MODS) [12, 18]. In this review, we outlined the impact of hyperoxia on the gut and GM in the context of oxygen inhalation. VA-ECMO is increasingly being used for oxygenation and circulatory support in patients with cardiogenic shock and cardiac arrest [19]. Typically, intensivists employ a peripheral cannulation strategy to facilitate rapid cannulation and provide prompt hemodynamic assistance during episodes of cardiogenic shock, with femoro-femoral VA-ECMO being the preferred approach. Additionally, we also discussed the potential repercussions of hyperoxia on the gut during peripheral VA-ECMO, as it may directly expose the intra-abdominal organs to severe hyperoxia due to dual circulation [4].

Hyperoxia during oxygen inhalation and peripheral VA-ECMO

Oxygen inhalation is typically administered with either a conservative or liberal approach to normalize arterial hypoxia with the goal of achieving either a low or a high level of oxygenation [20]. The oxygen-ICU trial was a pivotal study that unveiled significant clinical harm associated with liberal oxygen administration, including higher mortality and increased instances of shock, liver failure, and bacteremia [20]. This evidence has led to updated clinical practice guidelines emphasizing a more conservative approach to supplemental oxygen and has spurred numerous randomized trials to define the optimal regimen for a given clinical condition [21].

Figure 1 illustrates the three fundamental subtypes of hyperoxia (alveolar, whole-body, and lower-body hyperoxia). These subtypes are based on the pathophysiology of traditional oxygen inhalation (conservative vs. liberal oxygen therapy) and peripheral VA-ECMO. Conservative oxygen inhalation involves administering oxygen at the lowest feasible inspired fraction of oxygen (FiO2 = 21–40%) for patients with gas exchange impairment [20]. This approach results in an elevation of oxygen tension in the bronchi, alveoli, and arterial blood, ultimately leading to mild alveolar hyperoxia and normoxemia (PaO2 = 70–100 mmHg) [20, 22]. Alternatively, liberal oxygen inhalation aims to maintain the FiO2 at least 40%, resulting in moderate to severe alveolar hyperoxia and mild whole-body hyperoxia (PaO2 = 100–150 mmHg) [20]. During peripheral VA-ECMO support, desaturated blood is drawn from the inferior vena cava by a centrifugal pump, saturated within an oxygenator, and reperfused in the retrograde direction back to the aorta, leading to lower-body hyperoxia [4]. The Extracorporeal Life Support Organization (ELSO) recommends targeting a post-oxygenator partial pressure of oxygen (PPOSTO2) around 150 mmHg during VA-ECMO [23]. However, moderate hyperoxia (PaO2 = 150–300 mmHg) is still observed in 30% and severe hyperoxia (PaO2 = 300–500 mmHg) in 20% of cardiogenic shock patients during VA-ECMO support [24, 25], and these conditions are associated with increased mortality [24, 26].

Fig. 1
figure 1

Subtypes of hyperoxia of oxygen inhalation (conservative vs. liberal oxygen therapy) and peripheral VA-ECMO; A Conservative oxygen therapy leads to mild alveolar hyperoxia and normoxemia; B Liberal oxygen therapy leads to moderate or severe alveolar hyperoxia and mild whole-body hyperoxia; C Peripheral VA-ECMO may lead to moderate or severe lower-body hyperoxia; PO2: partial pressure of oxygen; FiO2: inspired oxygen fraction; FSO2: sweep gas oxygen fraction; PPOSTO2: post-oxygenator oxygen partial pressure

While alveolar hyperoxia is usually the primary concern during traditional oxygen inhalation [5, 27], an elevated whole-body PaO2 induced by liberal inspiratory oxygen may cause hyperoxia in extrapulmonary organs, including the gastrointestinal tract and GM [28, 29]. Moreover, the potential harm is directly related to hyperoxemia severity, and extreme hyperoxemia (i.e., PaO2 > 300 mmHg) is associated with more harm in critically ill populations [5]. During peripheral VA-ECMO, the lower body is exposed to even more severe arterial hyperoxia than during oxygen inhalation, so hyperoxic injury of the gastrointestinal tract is more likely, potentially altering the GM composition [30].

Biological effects of intestinal hyperoxia

Molecular oxygen is vital for mitochondrial respiration and thus for cellular and tissue homeostasis. However, mitochondrial respiration also results in production of reactive oxygen species (ROS) like superoxide anion, hydroxyl radical, and hydrogen peroxide, which increase at higher oxygen concentrations [5]. These ROS have both beneficial and detrimental effects. On the one hand, ROS are involved in various enzymatic pathways, signal transduction cascades, and reparative/protective processes such as the “respiratory burst” associated with phagocytosis. However, ROS production rate surpassing the endogenous antioxidant capacity of the cell can result in oxidative damage to nucleic acids, proteins, and lipids, leading to membrane failure, mutations, and activation of programmed cell death pathways [3]. 8-hydroxy-2′-deoxyguanosine (8-OHdG) is one of the predominant forms of ROS-induced oxidative lesions, and has therefore been widely used as a biomarker for oxidative injury [31]. An animal study elucidated that hyperoxia provoked intestinal oxidative injury by increasing the level of 8-OHdG in a time- and dose-dependent manner [6].

Intestinal ischemia/reperfusion (I/R) is a common feature during sepsis and MODS [32]. Hyperoxia following ischemia and reoxygenation is due in part to the reduced affinity of hemoglobin for oxygen under elevated carbon dioxide partial pressure and/or low blood pH, termed the Bohr effect [33]. The ensuing elevation in tissue oxygen results in ROS generation and reperfusion-induced tissue damage [33]. However, there is still controversy over whether hyperoxia has beneficial of harmful effects on the intestinal I/R [34, 35]. In support of this pathomechanism, administration of superoxide dismutase, an enzyme that scavenges superoxide radicals, attenuated the increased intestinal capillary permeability induced by regional ischemia in cats [36]. Other studies reported that hyperoxia reduced intestinal vascular constriction [28] and alleviated small bowel injury after I/R [34].

Gut injury and dysbiosis in critical illness

Over the past five decades, both clinical and experimental animal studies have highlighted the pivotal role of gut dysfunction and GM dysbiosis in critical illnesses [18]. For instance, gastrointestinal symptoms during the initial week of intensive care, especially gastrointestinal bleeding and absence of bowel sounds, were identified as independent predictors of higher 28-day mortality [37]. Similarly, intestinal fatty acid-binding protein (I-FABP), a known marker of enterocyte injury, was independently associated with shock and 28-day mortality in critically ill patients [38]. Early enteral nutrition promotes recovery in critically ill patients by preserving the homeostasis of gut barrier and GM [39]. A prevailing theory posits that gut failure increases the risk of adverse clinical outcomes by promoting sepsis and MODS [40].

Multiple studies have also reported that the GM can serve as a predictive factor for patients’ susceptibility to diseases and leveraging its potential holds promise in prevention and modulation of critical illnesses [18, 41,42,43]. Critical illnesses often lead to the depletion of “health-promoting” microbes and an increase in pathogenic microbes (dysbiosis) [13, 18]. Critically ill patients frequently exhibit significantly lower microbial diversity and robustness than healthy individuals. Further, opportunistic pathogens, including Enterobacteriaceae, Staphylococcus, Enterococcus, and Candida albicans, have been found to flourish in the gut of critically ill patients [13, 18, 44], while beneficial obligate anaerobes were reduced in abundance, particularly Faecalibacterium prausnitzii (an indicator of a healthy colonic microbiota), Lactobacillus, and Bifidobacterium [13, 18]. Probiotics and fecal microbial transplantation (FMT) have been demonstrated to improve the condition of patients with recurrent Clostridioides difficile infection [45, 46]. Similarly, a recent meta-analysis suggested that ingestion of probiotics or synbiotics during critical illness can reduce ventilator-associated pneumonia, healthcare-associated pneumonia, and length of stay in the ICU and hospital, although there may not be substantial effects on mortality [47].

Animal studies

Hyperoxia provokes gut injury and barrier dysfunction

Recent research has provided further substantiation of hyperoxia-induced gut injury in murine models through oxygen inhalation [6, 48,49,50]. This injury manifests as intestinal histopathological abnormalities, including mucosal atrophy (e.g., villus shortening), enterocyte death, reduced Paneth cells and goblet cells, and infiltration of polymorphonuclear leukocytes and macrophages [49, 51]. Both neonatal and adult exposure to hyperoxia disrupts the integrity of gut barrier, facilitating the translocation of lipopolysaccharides (LPS) from the gut bacteria into the bloodstream, ultimately resulting in endotoxemia [6, 51, 52]. Hyperoxia further exacerbates the cascade of gut inflammation by increasing the levels of pro-inflammatory cytokines (TNF-α, IL-1β, IFN-γ) and decreasing anti-inflammatory cytokines (IL-10, IL-17D) [49, 53]. Additionally, hyperoxia elevated the level of chemokine C-X-C motif ligand 1 in the gut epithelium, acting as a potent neutrophil chemoattractant and indicating the presence of intestinal neutrophilia [6].

Hyperoxia-induced gut injury is influenced by various signaling pathways, including toll-like receptor (TLR)-4, TNF, nuclear factor-κB (NF-κB), and nuclear factor erythroid 2-related factor 2 (Nrf2) pathways [6, 49]. TLR-4 is one of the receptors involved in innate immunity activated by LPS from Gram-negative bacteria during hyperoxia [6]. Nrf2, acting as a master regulator of redox homeostasis, is upregulated by ROS and playing a crucial role in cellular protection by inducing the expression of antioxidant genes [53]. In hyperoxic conditions, NF-κB becomes activated through TLR-4 and TNF pathways, leading to the production of inflammatory cytokines, enterocyte death, and the suppression of Nrf2 [50]. Supplementation of N-acetylcysteine, a potent antioxidant, mitigates hyperoxia-induced gut injury by inhibiting ROS production [52]. In summary, oxidative stress, immunity responses, and inflammation collectively contribute to oxygen-induced gut injury (Fig. 2).

Fig. 2
figure 2

Hyperoxia provokes gut injury and dysbiosis; LPS: lipopolysaccharide; TNF-α: tumor necrosis factor-alpha; TNFR: tumor necrosis factor receptor; NF-κB: nuclear factor-κB; Nrf2: nuclear factor erythroid 2-related factor 2; TLR-4: toll-like receptor-4; NAC: N-acetylcysteine; ROS: reactive oxygen species; DHA: docosahexaenoic acid; TJ: tight junction

Hyperoxia provokes gut dysbiosis and metabolic disorders

Inhalation of oxygen, leading to hyperoxia, has been extensively investigated in preclinical studies to understand its impact on the gut microbiome [29, 54,55,56]. Oxygen inhalation has consistently demonstrated a significant increase in Firmicutes/Bacteroidetes ratio in both neonatal and adult mice [57, 58]. Exposure to hyperoxia, both in neonatal and adult mice, has been linked to a marked expansion of oxygen-tolerant microbes, particularly pathogenic Proteobacteria, Enterobacteriaceae, and Staphylococcus [6, 29, 56]. Concurrently, it has led to a decrease in the relative abundance of oxygen-intolerant microbes like Bacteroidetes and Muribaculaceae, as well as Lactobacillus in mice [6, 55, 57]. Notably, even a brief exposure to hyperoxia (72 h) in adult mice resulted in significant alterations in gut microbiome, marked by the depletion of Ruminococcaceae [29]. Muribaculaceae, Lactobacillus, and Ruminococcaceae are beneficial microbes which can produce short chain fatty acids (SCFAs) with anti-inflammatory properties [59,60,61]. Hyperoxia may reprogram the metabolic pathways in the gut producing the abnormal metabolites [9]. Our recent metabolomics analysis revealed that oxygen inhalation suppressed the metabolism of fecal polyunsaturated fatty acids (PUFAs) in mice, resulting in reduced levels of linoleic acids and α-linolenic acids, along with their secondary metabolites [57]. Supplemental docosahexaenoic acid (DHA), a beneficial PUFA, may limit inflammation and apoptosis involved in hyperoxia-induced intestinal injury in neonatal mice [62]. These findings collectively suggest the presence of gut dysbiosis and metabolic disorders induced by hyperoxia [9, 49] (Fig. 2).

Cross talk between hyperoxia-induced gut dysbiosis and distant organ injuries

The GM contributes to the development and function of remote organs, including the lungs through the “gut–lung axis” and brain through the “gut–brain axis” [63]. Hyperoxia can disrupt GM homeostasis and proper signaling through these axes, contributing to gut, lung, and brain dysfunction [6, 54, 64]. For instance, hyperoxia was reported to promote bacterial translocation from the gut to the lungs in neonatal mice [65]. Oxygen inhalation also decreased the relative abundance of intestinal Lactobacillus, while administration of Lactobacillus attenuated hyperoxia-induced lung injury in murine models [55, 66]. Alternatively, oral administration of antimicrobial peptides restored gut dysbiosis and alleviated lung injury in mice exposed to hyperoxia [56]. Variations in gut bacterial communities correlated with variations in lung inflammation among hyperoxia-exposed mice [29]. Adopting a high-fiber diet or supplementation of the gut microbe metabolite acetate (SCFA) increased the abundance of Bacteroides, thereby mitigating gut dysbiosis and attenuating acute lung injury in hyperoxia-exposed mice [67]. In addition, it was reported that the TLR-4 pathway may contribute to the deleterious effects of hyperoxia-induced gut dysbiosis on lung development [64]. Hyperoxia exposure during the first week of life and ensuing gut dysbiosis were also implicated in brain dysfunction at adolescence in mice [54]. Collectively, these preclinical studies suggest that intestinal inflammation, dysbiosis, and metabolic disturbances contribute to oxygen-induced lung injury and brain dysfunction through gut–lung and gut–brain axes [9] (Fig. 3).

Fig. 3
figure 3

Dual circulation, mixing zone, and intestinal hyperoxia in peripheral VA-ECMO; PP: pulse pressure; ABG: arterial blood gas; LVEF: left ventricular ejection fraction; Mixing zone 1: ascending aorta; 2: aortic arch; 3: descending aorta; 4: thoracic/abdominal aorta

Clinical studies

A literature search was conducted in the PubMed database using the following keywords: “oxygen therapy,” “hyperoxia,” “hyperoxemia,” “anastomotic leakage,” “necrotizing enterocolitis,” and “ECMO.” Table 1 presents the key clinical studies that investigated the impact of hyperoxia on the intestinal morbidity. Notably, there is a dearth of human studies focusing on the effects of hyperoxia during VA-ECMO on the gut. Consequently, this discussion delves into the correlation between intestinal hyperoxia and dual circulation during peripheral VA-ECMO.

Table 1 Impact of hyperoxia on gut-associated morbidity in clinical studies

Lower vs. higher oxygenation targets and intestinal morbidity in adults

The recent LOCO2 trial, a multicenter randomized trial, studied liberal oxygen therapy (higher oxygenation targets) or conservative oxygen therapy (lower oxygenation targets) in patients with acute respiratory distress syndrome (ARDS) [68]. The trial was prematurely halted due to increased 90-day mortality and concerns about five episodes of mesenteric ischemia, all occurring in the conservative oxygen group [1]. However, subsequent analysis in the HOT-ICU trail revealed no significant differences in 90-day mortality or incidence of new episodes of intestinal ischemia between the lower and higher oxygenation target groups in the broader ICU population [69]. Further, the HOT-ICU trial included a sample approximately 15-fold larger than the LOCO2 trial, suggesting that the unequal distribution of intestinal ischemia between groups in the LOCO2 trial occurred by chance [70]. In addition, the increased incidence of mesenteric ischemia in the LOCO2 trial may be attributed to the lower oxygenation target (a peripheral oxygen saturation, SpO2 = 88%) in the conservative arm compared to that in the HOT-ICU trial (SpO2 = 90%) [1].

While the current studies on liberal oxygen inhalation do not report gut-associated adverse events [71], the possibility of subclinical enterocyte injury, such as elevated serum level of I-FABP, cannot be entirely ruled out. The sensitivity of small bowel to reduced oxygen delivery is well established owning to its unique blood supply characteristics [72]. Therefore, special attention should be given to the potential occurrence of intestinal ischemia, particularly in patients more susceptible to hypoxia, such as those with ARDS, when prescribing conservative oxygen therapy [1].

Lower vs. higher oxygenation targets and intestinal morbidity in infants

Necrotizing enterocolitis (NEC) is among the most common and devastating diseases in infants [73]. The Neonatal Oxygen Prospective Meta-analysis Collaboration, established in 2003 [74], was a joint effort involving investigators from five distinct randomized clinical trials: SUPPORT trial [75], the three BOOST II trials conducted in the UK, Australia, and New Zealand [76,77,78], and the COT trial [79]. These five studies, with a total sample size of almost 5000 infants, were designed to compare the effects of a lower oxygen saturation target (SpO2 = 85–89%) versus a higher target (SpO2 = 91–95%) on death or major disability as the primary outcome. According to the results, a lower oxygen saturation target significantly increased the mortality rate and the ratio of NEC patients requiring surgery [74]. NEC is more likely to occur in the colon, which has a higher ischemic tendency because of its dependence on the most distal branches of the mesenteric vascular supply [80]. However, the clinically appropriate oxygen saturation range in the intestines for preterm infants is unknown and may vary with gestational and postnatal age [81].

Perioperative hyperoxia and gastrointestinal surgery

Resection and anastomosis of the digestive tract is a commonly carried out procedure during gastrointestinal surgery [82]. Anastomotic leakage is a major postoperative complication which can result in local collections and sepsis and increase the length of hospital stay and mortality in patients [83, 84]. Tissue oxygen tension is often low in wounds and colorectal anastomoses that may reduce tissue healing and lead to a high risk of surgical site infection (SSI) and anastomotic leakage [85,86,87]. In 2016, the World Health Organization (WHO) recommended supplementing 80% oxygen during anesthesia and, if feasible, for 2 to 6 h after surgery to reduce SSI risk [88]. However, these WHO guidelines have met criticism due to the weakness of the underlying analyses [89]. Further, two updated meta-analyses and further clinical trials with large sample sizes did not detect any benefit of perioperative hyperoxia, but instead observed an increased frequency of pulmonary complications [90,91,92]. A recent meta-analysis did find reduced SSI and anastomotic leakage incidences in the pooled high FiO2 arm, but this finding should be interpreted with caution due to low quality of evidence presented in the individual studies [93]. Moreover, the double-blinded randomized PROXI trial of 1400 patients found that administration of 80% oxygen did not result in a significant change in the risks of SSI and anastomotic leakage after abdominal surgery compared to 30% oxygen [94].

Given this emerging evidence for the detrimental effects of hyperoxia on the gut [49], even with exposure for a brief 24-h period [6], perioperative hyperoxia may not be suitable during gastrointestinal surgery. In fact, the World Federation of Societies of Anesthesiologists recommends conservative perioperative oxygen inhalation (FiO2 = 30–40%) for intubated patients during general anesthesia and a normal SpO2 above 93% postoperatively [89, 95].

Dual circulation during peripheral VA-ECMO and intestinal hyperoxia

Peripheral VA-ECMO creates a dual-circulation hemodynamic pattern with competitive blood flows from both the heart and VA-ECMO contributing to circulation and oxygenation [4]. The mixing zone is defined as the point where these two flows intersect and the specific location of this mixing zone is contingent on the ratio of native cardiac output (NCO) to ECMO blood flow [96]. As NCO increases while ECMO support decreases, the mixing zone shifts distally along the aorta [96]. Arterial blood gas (ABG), pulse pressure (PP), and point-of-care ultrasound are valuable parameters for NCO assessment [96,97,98]. Using these methods, the mixing zone can be approximately localized to the ascending aorta if the NCO/ECMO flow ratio is 1:4, to the aortic arch if 1:3, the descending aorta if 1:2, and thoracic aorta/abdominal aorta if 1:1 [99]. The mesenteric circulation is primarily supplied by the celiac trunk and superior and inferior mesenteric arteries [100]. Celiac trunk mixing (50%/50% from the NCO/ECMO) occurs when the NCO/ECMO flow ratio is 1:1 [99]. These findings demonstrate that patients on VA-ECMO support are frequently exposed to prolonged hyperoxia of the gastrointestinal tract (Fig. 4).

Fig. 4
figure 4

Cross talk between hyperoxia-induced gut dysbiosis and distant organ injuries

Current challenges and future studies

Most preclinical studies have reported marked hyperoxia-induced gut injury and dysbiosis after exposing healthy animals to inhaled oxygen with a very high FiO2 (e.g., > 80%) [29, 48, 56]. However, this condition rarely occurs in clinical practice when oxygen inhalation is prescribed for hypoxic patients [21]. Therefore, it may be more fruitful to assess the impact of hyperoxia on the gastrointestinal tract in animal disease models (e.g., mice with acute lung injury) and other clinical conditions such as ARDS.

In contrast to inhaled oxygen, VA-ECMO directly exposes the human gastrointestinal tract to hyperoxia, potentially inducing gut injury and dysbiosis [4, 30]. Further animal model and clinical studies are needed to assess the frequency, severity, and risk factors for this condition. Most recent studies have detected arterial hyperoxia by measuring ABGs in samples extracted from the right arm as per the institutional ECMO protocol, primarily due to concerns regarding Harlequin syndrome [25, 101, 102]. The syndrome describes when NCO recovery coexists with respiratory failure, an antegrade ejection by the left ventricle of relatively deoxygenated blood hinders the retrograde oxygenated blood flow of the VA-ECMO in the aortic arch resulting in upper-body hypoxia [103]. As a result of dual circulation, however, the PaO2 measured at the right arm may underestimate the oxygen burden in the lower body [30]. We therefore suggest that future studies adopt PPOSTO2 act as a metric for hyperoxia assessment and concomitantly measure markers of gut injury (e.g., I-FABP), dysbiosis (e.g., Enterobacteriaceae) and metabolic disorders (e.g., SCFAs) to assess the potential negative impact of intestinal hyperoxia.

The impact of hyperoxia on the gut and GM appears to vary according to the clinical situation. While hyperbaric hyperoxia may be associated with improved anastomotic healing and a decreased risk of inflammatory bowel disease flares [86, 104], excess normobaric hyperoxia has harmful effects on the gut [6]. Maturation of oxygenation is a dynamic process in the developing intestines of infants and differs from adults [81]. Future studies should thus address how a particular oxygenation target can be set and achieved in specific age and illness groups, and how the particular target influences the gut and GM [1].

Finally, emerging novel technologies may help clinicians improve the regulation of intestinal oxygenation during oxygen therapy. Abdominal near-infrared spectroscopy can be used to measure intestinal oxygenation changes and to diagnose NEC in preterm infants [105, 106]. Several devices have also been developed to continuously monitor the PPOSTO2 during VA-ECMO support [4]. The reliability of these devices for reducing lower-body hyperoxia and minimizing hyperoxic injury to the intestines must be confirmed.

Conclusions

Recent animal studies highlight the risk of gut injury and dysbiosis induced by excessive oxygen inhalation. While the effects of hyperoxia on gut integrity and GM composition may vary across clinical settings and patient conditions, lower oxygenation targets may increase the risk of mesenteric ischemia and NEC in adults (SpO2 = 88–92%) and infants (SpO2 = 85–89%), respectively. Further, perioperative hyperoxia (FiO2 = 80%) may not reduce the risk for SSI and anastomotic leaks during gastrointestinal surgery, so a lower oxygenation target (FiO2 = 30–40%) may be more rational. Extreme intestinal hyperoxia (PaO2 ≥ 300 mmHg) during VA-ECMO is likely to contribute to gut injury and dysbiosis, and further studies are needed. It appears that targeting oxygenation within a normal range may help to avoid both hyperoxia-induced gut injury and hypoxia-induced mesenteric ischemia among most ICU patients. In addition, we suggest that PaO2 should be carefully titrated for specific clinical situations and contexts.

Availability of data and materials

Not applicable.

Abbreviations

VA-ECMO:

Venoarterial-extracorporeal membrane oxygenation

ROS:

Radical oxygen species

GM:

Gut microbiome

ICUs:

Intensive care units

ELSO:

Extracorporeal Life Support Organization

PPOSTO2 :

Post-oxygenator oxygen partial pressure

MODS:

Multiple organ dysfunction syndrome

FiO2 :

Inspired oxygen fraction

PO2 :

Oxygen partial pressure

PaO2 :

Arterial oxygen partial pressure

SpO2 :

Peripheral oxygen saturation

NCO:

Native cardiac output

ABG:

Arterial blood gas

PP:

Pulse pressure

I-FABP:

Intestinal-fatty acid-binding protein

ARDS:

Acute respiratory distress syndrome

NEC:

Necrotizing enterocolitis

SSI :

Surgical site infection

WHO:

World health organization

I/R:

Intestinal ischemia/reperfusion

SCFAs :

Short chain fatty acids

PUFAs:

Polyunsaturated fatty acids

8-OHdG:

8-Hydroxy-2′-deoxyguanosine

References

  1. Angus DC. Oxygen therapy for the critically Ill. N Engl J Med. 2020;382:1054–6.

    Article  PubMed  Google Scholar 

  2. Hochberg CH, Semler MW, Brower RG. Oxygen toxicity in critically Ill adults. Am J Respir Crit Care Med. 2021;204(6):632–41.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. Damiani E, Donati A, Girardis M. Oxygen in the critically ill: friend or foe. Curr Opin Anaesthesiol. 2018;31:129–35.

    Article  CAS  PubMed  Google Scholar 

  4. Winiszewski H, Guinot PG, Schmidt M, et al. Optimizing PO(2) during peripheral veno-arterial ECMO: a narrative review. Crit Care. 2022;26(1):226.

    Article  PubMed  PubMed Central  Google Scholar 

  5. Singer M, Young PJ, Laffey JG, Asfar P, Taccone FS, Skrifvars MB, et al. Dangers of hyperoxia. Crit Care. 2021;25:440.

    Article  PubMed  PubMed Central  Google Scholar 

  6. Li Y, Tao Y, Xu J, He Y, Zhang W, Jiang Z, et al. Hyperoxia provokes time- and dose-dependent gut injury and endotoxemia and alters gut microbiome and transcriptome in mice. Front Med (Lausanne). 2021;8: 732039.

    Article  PubMed  Google Scholar 

  7. Helmerhorst H, Schouten L, Wagenaar G, Juffermans NP, Roelofs J, Schultz MJ, et al. Hyperoxia provokes a time- and dose-dependent inflammatory response in mechanically ventilated mice, irrespective of tidal volumes. Intensive Care Med Exp. 2017;5:27.

    Article  PubMed  PubMed Central  Google Scholar 

  8. Rincon F, Kang J, Maltenfort M, Vibbert M, Urtecho J, Athar MK, et al. Association between hyperoxia and mortality after stroke: a multicenter cohort study. Crit Care Med. 2014;42:387–96.

    Article  PubMed  Google Scholar 

  9. Sun T, Yu H, Li D, Zhang H, Fu J. Emerging role of metabolic reprogramming in hyperoxia-associated neonatal diseases. Redox Biol. 2023;66: 102865.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Chu DK, Kim LH, Young PJ, Zamiri N, Almenawer SA, Jaeschke R, et al. Mortality and morbidity in acutely ill adults treated with liberal versus conservative oxygen therapy (IOTA): a systematic review and meta-analysis. Lancet. 2018;391:1693–705.

    Article  PubMed  Google Scholar 

  11. Damiani E, Adrario E, Girardis M, Romano R, Pelaia P, Singer M, et al. Arterial hyperoxia and mortality in critically ill patients: a systematic review and meta-analysis. Crit Care. 2014;18:711.

    Article  PubMed  PubMed Central  Google Scholar 

  12. Otani S, Coopersmith CM. Gut integrity in critical illness. J Intensive Care. 2019;7:17.

    Article  PubMed  PubMed Central  Google Scholar 

  13. Corriero A, Gadaleta RM, Puntillo F, Inchingolo F, Moschetta A, Brienza N. The central role of the gut in intensive care. Crit Care. 2022;26:379.

    Article  PubMed  PubMed Central  Google Scholar 

  14. Eckburg PB, Bik EM, Bernstein CN, Purdom E, Dethlefsen L, Sargent M, et al. Diversity of the human intestinal microbial flora. Science. 2005;308:1635–8.

    Article  ADS  PubMed  PubMed Central  Google Scholar 

  15. Albenberg L, Esipova TV, Judge CP, et al. Correlation between intraluminal oxygen gradient and radial partitioning of intestinal microbiota. Gastroenterology. 2014;147(5):1055-63.e8.

    Article  PubMed  Google Scholar 

  16. Rivera-Chávez F, Lopez CA, Bäumler AJ. Oxygen as a driver of gut dysbiosis. Free Radic Biol Med. 2017;105:93–101.

    Article  PubMed  Google Scholar 

  17. Schmidt TM, Kao JY. A little O2 may go a long way in structuring the GI microbiome. Gastroenterology. 2014;147:956–9.

    Article  PubMed  Google Scholar 

  18. Dickson RP. The microbiome and critical illness. Lancet Respir Med. 2016;4(1):59–72.

    Article  PubMed  Google Scholar 

  19. Rao P, Khalpey Z, Smith R, Burkhoff D, Kociol RD. Venoarterial extracorporeal membrane oxygenation for cardiogenic shock and cardiac arrest. Circ Heart Fail. 2018;11: e004905.

    Article  PubMed  Google Scholar 

  20. Girardis M, Busani S, Damiani E, Donati A, Rinaldi L, Marudi A, et al. Effect of conservative vs conventional oxygen therapy on mortality among patients in an intensive care unit: the oxygen-ICU randomized clinical trial. JAMA. 2016;316:1583–9.

    Article  CAS  PubMed  Google Scholar 

  21. Siemieniuk R, Chu DK, Kim LH, Güell-Rous MR, Alhazzani W, Soccal PM, et al. Oxygen therapy for acutely ill medical patients: a clinical practice guideline. BMJ. 2018;363: k4169.

    Article  PubMed  Google Scholar 

  22. Dushianthan A, Bracegirdle L, Cusack R, Cumpstey AF, Postle AD, Grocott M. Alveolar hyperoxia and exacerbation of lung injury in critically Ill SARS-CoV-2 pneumonia. Med Sci (Basel). 2023;11(4):70.

    CAS  PubMed  Google Scholar 

  23. Lorusso R, Shekar K, MacLaren G, Schmidt M, Pellegrino V, Meyns B, et al. ELSO interim guidelines for venoarterial extracorporeal membrane oxygenation in adult cardiac patients. ASAIO J. 2021;67:827–44.

    Article  PubMed  Google Scholar 

  24. Premraj L, Brown A, Fraser JF, Pellegrino V, Pilcher D, Burrell A. Oxygenation during venoarterial extracorporeal membrane oxygenation: physiology, current evidence, and a pragmatic approach to oxygen titration. Crit Care Med. 2023 .

  25. Tigano S, Caruso A, Liotta C, et al. Exposure to severe hyperoxemia worsens survival and neurological outcome in patients supported by veno-arterial extracorporeal membrane oxygenation: a meta-analysis. Resuscitation. 2023: 110071.

  26. Jentzer JC, Miller PE, Alviar C, Yalamuri S, Bohman JK, Tonna JE. Exposure to arterial hyperoxia during extracorporeal membrane oxygenator support and mortality in patients with cardiogenic shock. Circ Heart Fail. 2023;16(4): e010328.

    Article  CAS  PubMed  Google Scholar 

  27. Davis WB, Rennard SI, Bitterman PB, Crystal RG. Pulmonary oxygen toxicity. Early reversible changes in human alveolar structures induced by hyperoxia. N Engl J Med. 1983;309:878–83.

    Article  CAS  PubMed  Google Scholar 

  28. Smit B, Smulders YM, Eringa EC, Oudemans-van Straaten HM, Girbes A, Wever KE, et al. Effects of hyperoxia on vascular tone in animal models: systematic review and meta-analysis. Crit Care. 2018;22:189.

    Article  PubMed  PubMed Central  Google Scholar 

  29. Ashley SL, Sjoding MW, Popova AP, Cui TX, Hoostal MJ, Schmidt TM, et al. Lung and gut microbiota are altered by hyperoxia and contribute to oxygen-induced lung injury in mice. Sci Transl Med. 2020;12:eaau9959.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Winiszewski H, Piton G, Perrotti A, Capellier G. Hyperoxemia and veno-arterial extracorporeal membrane oxygenation: Do not forget the gut. Crit Care Med. 2018;46(1):e98–9.

    Article  PubMed  Google Scholar 

  31. Valavanidis A, Vlachogianni T, Fiotakis C. 8-hydroxy-2’ -deoxyguanosine (8-OHdG): a critical biomarker of oxidative stress and carcinogenesis. J Environ Sci Health C Environ Carcinog Ecotoxicol Rev. 2009;27(2):120–39.

    Article  CAS  PubMed  Google Scholar 

  32. Ma Y, Yang X, Chatterjee V, Wu MH, Yuan SY. The gut-lung axis in systemic inflammation. Role of mesenteric lymph as a conduit. Am J Respir Cell Mol Biol. 2021;64(1):19–28.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Wolbarsht ML, Fridovich I. Hyperoxia during reperfusion is a factor in reperfusion injury. Free Radic Biol Med. 1989;6(1):61–2.

    Article  CAS  PubMed  Google Scholar 

  34. Sukhotnik I, Brod V, Lurie M, Rahat MA, Shnizer S, Lahat N, et al. The effect of 100% oxygen on intestinal preservation and recovery following ischemia-reperfusion injury in rats. Crit Care Med. 2009;37:1054–61.

    Article  CAS  PubMed  Google Scholar 

  35. Schoenberg MH, Beger HG. Reperfusion injury after intestinal ischemia. Crit Care Med. 1993;21:1376–86.

    Article  CAS  PubMed  Google Scholar 

  36. Granger DN, Rutili G, McCord JM. Superoxide radicals in feline intestinal ischemia. Gastroenterology. 1981;81(1):22–9.

    Article  CAS  PubMed  Google Scholar 

  37. Reintam Blaser A, Poeze M, Malbrain ML, Björck M, Oudemans-van Straaten HM, Starkopf J. Gastrointestinal symptoms during the first week of intensive care are associated with poor outcome: a prospective multicentre study. Intensive Care Med. 2013;39(5):899–909.

    Article  PubMed  PubMed Central  Google Scholar 

  38. Piton G, Belon F, Cypriani B, Regnard J, Puyraveau M, Manzon C, et al. Enterocyte damage in critically ill patients is associated with shock condition and 28-day mortality. Crit Care Med. 2013;41(9):2169–76.

    Article  PubMed  Google Scholar 

  39. Moron R, Galvez J, Colmenero M, Anderson P, Cabeza J, Rodriguez-Cabezas ME. The importance of the microbiome in critically Ill patients: role of nutrition. Nutrients. 2019;11(12):3002.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Assimakopoulos SF, Triantos C, Thomopoulos K, et al. Gut-origin sepsis in the critically ill patient: pathophysiology and treatment. Infection. 2018;46(6):751–60.

    Article  CAS  PubMed  Google Scholar 

  41. Adelman MW, Woodworth MH, Langelier C, Busch LM, Kempker JA, Kraft CS, et al. The gut microbiome’s role in the development, maintenance, and outcomes of sepsis. Crit Care. 2020;24:278.

    Article  PubMed  PubMed Central  Google Scholar 

  42. Haak BW, Wiersinga WJ. The role of the gut microbiota in sepsis. Lancet Gastroenterol Hepatol. 2017;2:135–43.

    Article  PubMed  Google Scholar 

  43. Panigrahi P, Parida S, Nanda NC, Satpathy R, Pradhan L, Chandel DS, et al. A randomized synbiotic trial to prevent sepsis among infants in rural India. Nature. 2017;548:407–12.

    Article  ADS  CAS  PubMed  Google Scholar 

  44. McDonald D, Ackermann G, Khailova L, Baird C, Heyland D, Kozar R, et al. Extreme dysbiosis of the microbiome in critical illness. mSphere. 2016; 1(4)

  45. Quraishi MN, Widlak M, Bhala N, et al. Systematic review with meta-analysis: the efficacy of faecal microbiota transplantation for the treatment of recurrent and refractory Clostridium difficile infection. Aliment Pharmacol Ther. 2017;46(5):479–93.

    Article  CAS  PubMed  Google Scholar 

  46. Feuerstadt P, Louie TJ, Lashner B, Wang E, Diao L, Bryant JA, et al. SER-109, an oral microbiome therapy for recurrent clostridioides difficile infection. N Engl J Med. 2022;386:220–9.

    Article  CAS  PubMed  Google Scholar 

  47. Sharif S, Greer A, Skorupski C, Hao Q, Johnstone J, Dionne JC, et al. Probiotics in critical illness: a systematic review and meta-analysis of randomized controlled trials. Crit Care Med. 2022;50:1175–86.

    Article  PubMed  Google Scholar 

  48. Chou HC, Chen CM. Neonatal hyperoxia disrupts the intestinal barrier and impairs intestinal function in rats. Exp Mol Pathol. 2017;102:415–21.

    Article  CAS  PubMed  Google Scholar 

  49. Wang HC, Chou HC, Chen CM. Molecular mechanisms of hyperoxia-induced neonatal intestinal injury. Int J Mol Sci. 2023;24:4366.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. Chou HC, Chen CM. Cathelicidin attenuates hyperoxia-induced intestinal injury through inhibition of NF-κB activity in newborn rats. Exp Mol Pathol. 2020;113: 104269.

    Article  CAS  PubMed  Google Scholar 

  51. Chen CM, Chou HC. Hyperoxia disrupts the intestinal barrier in newborn rats. Exp Mol Pathol. 2016;101:44–9.

    Article  CAS  PubMed  Google Scholar 

  52. Liu DY, Lou WJ, Zhang DY, Sun SY. ROS plays a role in the neonatal rat intestinal barrier damages induced by hyperoxia. Biomed Res Int. 2020;2020:8819195.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  53. Liu X, Zhang D, Cai Q, Liu D, Sun S. Involvement of nuclear factor erythroid 2-related factor 2 in neonatal intestinal interleukin-17D expression in hyperoxia. Int J Mol Med. 2020;46(4):1423–32.

    CAS  PubMed  PubMed Central  Google Scholar 

  54. Lo YC, Chen KY, Chou HC, Lin IH, Chen CM. Neonatal hyperoxia induces gut dysbiosis and behavioral changes in adolescent mice. J Chin Med Assoc. 2021;84(3):290–8.

    Article  CAS  PubMed  Google Scholar 

  55. Chen CM, Yang Y, Chou HC, Lin S. Intranasal administration of Lactobacillus johnsonii attenuates hyperoxia-induced lung injury by modulating gut microbiota in neonatal mice. J Biomed Sci. 2023;30(1):57.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  56. Abdelgawad A, Nicola T, Martin I, et al. Antimicrobial peptides modulate lung injury by altering the intestinal microbiota. Microbiome. 2023;11(1):226.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  57. Cai Y, Luo Y, Dai N, Yang Y, He Y, Chen H, et al. Functional metagenomic and metabolomics analysis of gut dysbiosis induced by hyperoxia. Front Microbiol. 2023;14:1197970.

    Article  PubMed  PubMed Central  Google Scholar 

  58. Chen CM, Yang Y, Chou HC. Maternal antibiotic exposure disrupts microbiota and exacerbates hyperoxia-induced lung injury in neonatal mice. Pediatr Res. 2021;90(4):776–83.

    Article  CAS  PubMed  Google Scholar 

  59. Wu T, Yu Q, Luo Y, Dai Z, Zhang Y, Wang C, et al. Whole-grain highland barley attenuates atherosclerosis associated with NLRP3 inflammasome pathway and gut microbiota in ApoE(-/-) mice. Nutrients. 2023;15:4186.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  60. Lee J, d’Aigle J, Atadja L, Quaicoe V, Honarpisheh P, Ganesh BP, et al. Gut microbiota-derived short-chain fatty acids promote poststroke recovery in aged mice. Circ Res. 2020;127:453–65.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  61. Li H, Xiang Y, Zhu Z, Wang W, Jiang Z, Zhao M, et al. Rifaximin-mediated gut microbiota regulation modulates the function of microglia and protects against CUMS-induced depression-like behaviors in adolescent rat. J Neuroinflammation. 2021;18:254.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  62. Li N, Ma L, Liu X, Shaw L, Li Calzi S, Grant MB, et al. Arginyl-glutamine dipeptide or docosahexaenoic acid attenuates hyperoxia-induced small intestinal injury in neonatal mice. J Pediatr Gastroenterol Nutr. 2012;54:499–504.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  63. Budden KF, Gellatly SL, Wood DL, et al. Emerging pathogenic links between microbiota and the gut-lung axis. Nat Rev Microbiol. 2017;15(1):55–63.

    Article  CAS  PubMed  Google Scholar 

  64. Wedgwood S, Gerard K, Halloran K, Hanhauser A, Monacelli S, Warford C, et al. Intestinal dysbiosis and the developing lung: the role of toll-like receptor 4 in the gut-lung axis. Front Immunol. 2020;11:357.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  65. Chen CM, Chou HC, Yang Y, Su EC, Liu YR. Predicting hyperoxia-induced lung injury from associated intestinal and lung dysbiosis in neonatal mice. Neonatology. 2021;118(2):163–73.

    Article  CAS  PubMed  Google Scholar 

  66. Wedgwood S, Warford C, Agvatisiri SR, Thai PN, Chiamvimonvat N, Kalanetra KM, et al. The developing gut-lung axis: postnatal growth restriction, intestinal dysbiosis, and pulmonary hypertension in a rodent model. Pediatr Res. 2020;87(3):472–9.

    Article  CAS  PubMed  Google Scholar 

  67. Chu SJ, Tang SE, Pao HP, Wu SY, Liao WI. A high-fiber diet or dietary supplementation of acetate attenuate hyperoxia-induced acute lung injury. Nutrients. 2022;14(24):5231.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  68. Barrot L, Asfar P, Mauny F, Winiszewski H, Montini F, Badie J, et al. Liberal or conservative oxygen therapy for acute respiratory distress syndrome. N Engl J Med. 2020;382:999–1008.

    Article  CAS  PubMed  Google Scholar 

  69. Schjørring OL, Klitgaard TL, Perner A, Wetterslev J, Lange T, Siegemund M, et al. Lower or higher oxygenation targets for acute hypoxemic respiratory failure. N Engl J Med. 2021;384:1301–11.

    Article  PubMed  Google Scholar 

  70. Young PJ. Effect of oxygen therapy on mortality in the ICU. N Engl J Med. 2021;384:1361–3.

    Article  CAS  PubMed  Google Scholar 

  71. Klitgaard TL, Schjørring OL, Nielsen FM, Meyhoff CS, Perner A, Wetterslev J, et al. Higher versus lower fractions of inspired oxygen or targets of arterial oxygenation for adults admitted to the intensive care unit. Cochrane Database Syst Rev. 2023;9:CD012631.

    PubMed  Google Scholar 

  72. Lundgren O. The circulation of the small bowel mucosa. Gut. 1974;15:1005–13.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  73. Neu J, Walker WA. Necrotizing enterocolitis. N Engl J Med. 2011;364:255–64.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  74. Askie LM, Darlow BA, Finer N, Schmidt B, Stenson B, Tarnow-Mordi W, et al. Association between oxygen saturation targeting and death or disability in extremely preterm infants in the neonatal oxygenation prospective meta-analysis collaboration. JAMA. 2018;319:2190–201.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  75. Vaucher YE, Peralta-Carcelen M, Finer NN, Carlo WA, Gantz MG, Walsh MC, et al. Neurodevelopmental outcomes in the early CPAP and pulse oximetry trial. N Engl J Med. 2012;367:2495–504.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  76. Darlow BA, Marschner SL, Donoghoe M, et al. Randomized controlled trial of oxygen saturation targets in very preterm infants: two year outcomes. J Pediatr. 2014;165(1):30-35.e2.

    Article  PubMed  Google Scholar 

  77. Tarnow-Mordi W, Stenson B, Kirby A, et al. Outcomes of two trials of oxygen-saturation targets in preterm infants. N Engl J Med. 2016;374(8):749–60.

    Article  CAS  PubMed  Google Scholar 

  78. Stenson BJ, Tarnow-Mordi WO, Darlow BA, Simes J, Juszczak E, Askie L, et al. Oxygen saturation and outcomes in preterm infants. N Engl J Med. 2013;368:2094–104.

    Article  PubMed  Google Scholar 

  79. Schmidt B, Whyte RK, Asztalos EV, Moddemann D, Poets C, Rabi Y, et al. Effects of targeting higher vs lower arterial oxygen saturations on death or disability in extremely preterm infants: a randomized clinical trial. JAMA. 2013;309:2111–20.

    Article  CAS  PubMed  Google Scholar 

  80. van der Heide M, Mebius MJ, Bos AF, Roofthooft M, Berger R, Hulscher J, et al. Hypoxic/ischemic hits predispose to necrotizing enterocolitis in (near) term infants with congenital heart disease: a case control study. BMC Pediatr. 2020;20:553.

    Article  PubMed  PubMed Central  Google Scholar 

  81. Dotinga BM, Mintzer JP, Moore JE, Hulscher J, Bos AF, Kooi E. Maturation of intestinal oxygenation: a review of mechanisms and clinical implications for preterm neonates. Front Pediatr. 2020;8:354.

    Article  PubMed  PubMed Central  Google Scholar 

  82. Goulder F. Bowel anastomoses: the theory, the practice and the evidence base. World J Gastrointest Surg. 2012;4:208–13.

    Article  PubMed  PubMed Central  Google Scholar 

  83. Reducing surgical site infections in low-income and middle-income countries (FALCON): a pragmatic, multicentre, stratified, randomised controlled trial. Lancet. 2021;398:1687–99.

  84. Hajjar R, Gonzalez E, Fragoso G, Oliero M, Alaoui AA, Calvé A, et al. Gut microbiota influence anastomotic healing in colorectal cancer surgery through modulation of mucosal proinflammatory cytokines. Gut. 2023;72:1143–54.

    Article  CAS  PubMed  Google Scholar 

  85. Hopf HW, Hunt TK, West JM, Blomquist P, Goodson WH 3rd, Jensen JA, et al. Wound tissue oxygen tension predicts the risk of wound infection in surgical patients. Arch Surg. 1997;132:997–1004.

    Article  CAS  PubMed  Google Scholar 

  86. Makanyengo SO, Carroll GM, Goggins BJ, Smith SR, Pockney PG, Keely S. Systematic review on the influence of tissue oxygenation on gut microbiota and anastomotic healing. J Surg Res. 2020;249:186–96.

    Article  CAS  PubMed  Google Scholar 

  87. Hunt TK, Hopf HW. High inspired oxygen fraction and surgical site infection. JAMA. 2009;302:1588–9.

    Article  CAS  PubMed  Google Scholar 

  88. Allegranzi B, Zayed B, Bischoff P, Kubilay NZ, de Jonge S, de Vries F, et al. New WHO recommendations on intraoperative and postoperative measures for surgical site infection prevention: an evidence-based global perspective. Lancet Infect Dis. 2016;16:e288–303.

    Article  PubMed  Google Scholar 

  89. Hedenstierna G, Meyhoff CS, Perchiazzi G, Larsson A, Wetterslev J, Rasmussen LS. Modification of the world health organization global guidelines for prevention of surgical site infection is needed. Anesthesiology. 2019;131:765–8.

    Article  PubMed  Google Scholar 

  90. Mattishent K, Thavarajah M, Sinha A, Peel A, Egger M, Solomkin J, et al. Safety of 80% vs 30–35% fraction of inspired oxygen in patients undergoing surgery: a systematic review and meta-analysis. Br J Anaesth. 2019;122:311–24.

    Article  PubMed  Google Scholar 

  91. de Jonge S, Egger M, Latif A, Loke YK, Berenholtz S, Boermeester M, et al. Effectiveness of 80% vs 30–35% fraction of inspired oxygen in patients undergoing surgery: an updated systematic review and meta-analysis. Br J Anaesth. 2019;122:325–34.

    Article  PubMed  Google Scholar 

  92. Staehr-Rye AK, Meyhoff CS, Scheffenbichler FT, Vidal Melo MF, Gätke MR, Walsh JL, et al. High intraoperative inspiratory oxygen fraction and risk of major respiratory complications. Br J Anaesth. 2017;119:140–9.

    Article  CAS  PubMed  Google Scholar 

  93. Kuh JH, Jung WS, Lim L, Yoo HK, Ju JW, Lee HJ, et al. The effect of high perioperative inspiratory oxygen fraction for abdominal surgery on surgical site infection: a systematic review and meta-analysis. Sci Rep. 2023;13:15599.

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  94. Meyhoff CS, Wetterslev J, Jorgensen LN, Henneberg SW, Høgdall C, Lundvall L, et al. Effect of high perioperative oxygen fraction on surgical site infection and pulmonary complications after abdominal surgery: the PROXI randomized clinical trial. JAMA. 2009;302:1543–50.

    Article  CAS  PubMed  Google Scholar 

  95. Meyhoff CS, Fonnes S, Wetterslev J, Jorgensen LN, Rasmussen LS. WHO Guidelines to prevent surgical site infections. Lancet Infect Dis. 2017;17:261–2.

    Article  PubMed  Google Scholar 

  96. Asija R, Fried JA, Siddall EC, et al. How I manage differential gas exchange in peripheral venoarterial extracorporeal membrane oxygenation. Crit Care. 2023;27(1):408.

    Article  PubMed  PubMed Central  Google Scholar 

  97. Mourad M, Eliet J, Zeroual N, Saour M, Sentenac P, Manna F, et al. Pulse pressure and end-tidal carbon dioxide for monitoring low native cardiac output during veno-arterial ECLS: a prospective observational study. Crit Care. 2020;24(1):569.

    Article  PubMed  PubMed Central  Google Scholar 

  98. Hussey PT, von Mering G, Nanda NC, Ahmed MI, Addis DR. Echocardiography for extracorporeal membrane oxygenation. Echocardiography. 2022;39(2):339–70.

    Article  PubMed  PubMed Central  Google Scholar 

  99. Stevens MC, Callaghan FM, Forrest P, Bannon PG, Grieve SM. Flow mixing during peripheral veno-arterial extra corporeal membrane oxygenation—a simulation study. J Biomech. 2017;55:64–70.

    Article  CAS  PubMed  Google Scholar 

  100. Salim H, Ozgur O, Erman K, et al. Collateral circulation develops in stenosis of the celiac trunk and superior mesenteric artery. Surg Radiol Anat. 2023;45(4):479–86.

    Article  PubMed  Google Scholar 

  101. Wilson J, Fisher R, Caetano F, et al. Managing Harlequin Syndrome in VA-ECMO - do not forget the right ventricle. Perfusion. 2022;37(5):526–9.

    Article  PubMed  Google Scholar 

  102. Winiszewski H, Piton G, Capellier G. Early hyperoxia and 28-day mortality in patients on venoarterial ECMO support for refractory cardiogenic shock: discussion about potential confounding factors. Crit Care. 2022;26(1):313.

    Article  PubMed  PubMed Central  Google Scholar 

  103. Honore PM, Barreto Gutierrez L, Kugener L, Redant S, Attou R, Gallerani A, et al. Risk of harlequin syndrome during bi-femoral peripheral VA-ECMO: should we pay more attention to the watershed or try to change the venous cannulation site. Crit Care. 2020;24:450.

    Article  PubMed  PubMed Central  Google Scholar 

  104. Singh AK, Jha DK, Jena A, Kumar-M P, Sebastian S, Sharma V. Hyperbaric oxygen therapy in inflammatory bowel disease: a systematic review and meta-analysis. Eur J Gastroenterol Hepatol. 2021;33:e564–73.

    Article  CAS  PubMed  Google Scholar 

  105. Thomas RA, Ballard MR, Aladangady N, Banerjee J. Abdominal Near Infrared Spectroscopy can be reliably used to measure splanchnic oxygenation changes in preterm infants. J Perinatol. 2023;43:716–21.

    Article  CAS  PubMed  Google Scholar 

  106. Palleri E, van der Heide M, Hulscher J, Bartocci M, Wester T, Kooi E. Clinical usefulness of splanchnic oxygenation in predicting necrotizing enterocolitis in extremely preterm infants: a cohort study. BMC Pediatr. 2023;23:336.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

The authors express their gratitude for the financial supports received.

Funding

This study was supported by the National Natural Science Foundation of China (82160370, 81960024), Science and Technology Program of Guizhou Province (QIANKEHEZHICHEN[2022]YIBAN179, QIANKEHEZHICHEN S[2020]2319, QIANKEHEJICHU[2021]YIBAN441), Zunyi Science and Technology Planning Project [Zun yi Ke He HZ ZI (2023) 207], and Educational Department of Guizhou Province [Qianjiaoji 2023(020)].

Author information

Authors and Affiliations

Authors

Contributions

ND, YL, YT and JG reviewed the literature, performed the study, and contributed to manuscript drafting. YH, YC, and HQ contributed to interpreting the findings and manuscript drafting. XF, TC, MC, and ZX reviewed the literature and were responsible for important intellectual content in the manuscript. All authors issued final approval for the version to be submitted.

Corresponding authors

Correspondence to Xiaoyun Fu, Miao Chen or Zhouxiong Xing.

Ethics declarations

Ethics approval and consent to participate

Not applicable.

Consent for publication

Written informed consent for the publication of this work was obtained from all participants.

Competing interests

The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.

Additional information

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Dai, N., Gu, J., Luo, Y. et al. Impact of hyperoxia on the gut during critical illnesses. Crit Care 28, 66 (2024). https://0-doi-org.brum.beds.ac.uk/10.1186/s13054-024-04848-9

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://0-doi-org.brum.beds.ac.uk/10.1186/s13054-024-04848-9

Keywords