Skip to main content
Erschienen in: Respiratory Research 1/2022

Open Access 01.12.2022 | Review

The effects of ischaemic conditioning on lung ischaemia–reperfusion injury

verfasst von: Dimitrios Vlastos, Mohamed Zeinah, George Ninkovic-Hall, Stefanos Vlachos, Agni Salem, Athanasios Asonitis, Hemangi Chavan, Lazaros Kalampalikis, Abdullah Al Shammari, José María Alvarez Gallesio, Aina Pons, Ioanna Andreadou, Ignatios Ikonomidis

Erschienen in: Respiratory Research | Ausgabe 1/2022

Abstract

Ischaemia–reperfusion injury (IRI) encompasses the deleterious effects on cellular function and survival that result from the restoration of organ perfusion. Despite their unique tolerance to ischaemia and hypoxia, afforded by their dual (pulmonary and bronchial) circulation as well as direct oxygen diffusion from the airways, lungs are particularly susceptible to IRI (LIRI). LIRI may be observed in a variety of clinical settings, including lung transplantation, lung resections, cardiopulmonary bypass during cardiac surgery, aortic cross-clamping for abdominal aortic aneurysm repair, as well as tourniquet application for orthopaedic operations. It is a diagnosis of exclusion, manifesting clinically as acute lung injury (ALI) or acute respiratory distress syndrome (ARDS). Ischaemic conditioning (IC) signifies the original paradigm of treating IRI. It entails the application of short, non-lethal ischemia and reperfusion manoeuvres to an organ, tissue, or arterial territory, which activates mechanisms that reduce IRI. Interestingly, there is accumulating experimental and preliminary clinical evidence that IC may ameliorate LIRI in various pathophysiological contexts. Considering the detrimental effects of LIRI, ranging from ALI following lung resections to primary graft dysfunction (PGD) after lung transplantation, the association of these entities with adverse outcomes, as well as the paucity of protective or therapeutic interventions, IC holds promise as a safe and effective strategy to protect the lung. This article aims to provide a narrative review of the existing experimental and clinical evidence regarding the effects of IC on LIRI and prompt further investigation to refine its clinical application.
Hinweise

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Abkürzungen
AAA
Abdominal aortic aneurysm
ALI
Acute lung injury
ARDS
Acute respiratory distress syndrome
ATP
Adenosine triphosphate
(A-a) DO2
Alveolar-arterial oxygen gradient
BAL
Bronchoalveolar lavage
CCL5
Chemokine ligand 5
Cd
Dynamic lung compliance
CGRP
Calcitonin gene-related peptide
CPAP
Continuous positive airway pressure
Cs
Static lung compliance
EBC
Exhaled breath condensate
ET-1
Endothelin-1
FIO2
Fraction of inspired oxygen
HPV
Hypoxic pulmonary vasoconstriction
hsCRP
Hypersensitive C-reactive protein
IC
Ischaemic conditioning
IFN-γ
Interferon-gamma
IL
Interleukin
IRI
Ischaemia–reperfusion injury
LIRI
Lung ischaemia–reperfusion injury
MCP-1
Monocyte chemoattractant protein-1
MDA
Malondialdehyde
MIP-2
Macrophage inflammatory protein-2
MOF
Multiorgan failure
mPAP
Mean pulmonary artery pressure
MPO
Myeloperoxidase
OLV
One lung ventilation
PaCO2
Arterial carbon dioxide tension
PaO2
Arterial oxygen tension
PAP
Pulmonary artery pressure
Paw
Airway pressure
PEEP
Positive end-expiratory pressure
PGD
Primary graft dysfunction
PMN
Polymorphonuclear leucocyte
PvO2
Mixed venous oxygen tension
PVR
Pulmonary vascular resistance
PVRI
Pulmonary vascular resistance index
P/F ratio
Ratio of the partial pressure of oxygen to the inspired fraction of oxygen
Raw
Airway resistance
RCT
Randomised control trial
RIC
Remote ischemic conditioning
ROS
Reactive oxygen species
sICAM-1
Serum soluble intercellular adhesion molecule-1
SOD
Superoxide dismutase
SvO2
Venous oxygen saturation
TBARS
Thiobarbituric acid reactive substances
TLC
Total lung capacity
TNF-a
Tumour necrosis factor-alpha
TXB2
Thromboxane B2
VCAM-1
Vascular cell adhesion molecule-1
VSD
Ventricular septal defect
V/Q mismatch
Ventilation-perfusion mismatch

Background

Ischaemia–reperfusion injury (IRI) encompasses the deleterious effects on cellular function and survival that result from the restoration of organ perfusion [1]. Counterintuitively, IRI further aggravates ischaemic organ damage as the degree of injury after reperfusion surpasses that caused by ischaemia per se [2]. It is mediated by sterile inflammation, enhanced oxidative stress and coagulation, endothelial dysfunction, and activation of cellular death pathways [1, 2]. Crucially, it is a systemic process with the potential to evoke distant organ injury and progress to multiple organ dysfunction syndrome [2]. Despite their unique tolerance to ischaemia and hypoxia, afforded by their dual (pulmonary and bronchial) circulation as well as direct oxygen diffusion from the airways [3], lungs are particularly susceptible to IRI (LIRI) [4]. Importantly, the cessation of ventilation leads to functional impairments similar to those induced by hypoperfusion [3], to which it is also interrelated by way of hypoxic pulmonary vasoconstriction (HPV) [5]. LIRI may be observed in a variety of clinical settings, including lung transplantation [4, 6], lung resections [7, 8], cardiopulmonary bypass (CPB) during cardiac surgery [912], aortic cross-clamping for abdominal aortic aneurysm (AAA) repair [13], as well as tourniquet application for orthopaedic operations [14]. It culminates in the breakdown of lung endothelial and epithelial barriers, leading to pulmonary oedema with attendant gas exchange impairment [4] and increased pulmonary vascular resistance resulting in pulmonary hypertension [15].
Ischaemic conditioning (IC) signifies the original paradigm of treating IRI. It entails the application of short, non-lethal ischemia and reperfusion manoeuvres to an organ, tissue, or arterial territory, which activates mechanisms that reduce IRI [16]. The concept of IC has several temporal and anatomical variations. In specific, the protective ischaemic stimulus may be applied before, during, or following the index reperfusion episode (pre-, per-, and post-conditioning respectively) with similar beneficial effects [16]. Furthermore, this intervention has systemic protective properties which are exploited by remote ischemic conditioning (RIC): biochemical and neuronal mechanisms confer protection to organs distant to the conditioning stimulus [16]. There is accumulating experimental and clinical evidence that IC may ameliorate LIRI in various pathophysiological contexts. In specific, it reduces the underlying oxidative stress and sterile inflammation in animal [17] and human models [7]. This is translated into decreased histologic damage [18], reduced pulmonary oedema [19], and improved respiratory function [8, 19] and pulmonary vascular haemodynamics [10, 20]. Considering the detrimental effects of LIRI, ranging from acute lung injury (ALI) following lung resections [15] to primary graft dysfunction (PGD) after lung transplantation [4], the association of these entities with adverse outcomes [4, 21], as well as the paucity of protective or therapeutic clinical interventions [4], ischaemic conditioning holds promise as a safe and effective strategy to protect the lung. This article aims to provide a review of the existing experimental and clinical evidence regarding the effects of IC on LIRI.

The pathophysiology of lung ischaemia reperfusion injury

Enhanced oxidative stress appears to play a prominent role in the pathophysiology of LIRI [22]. Ischaemia, in the presence or not of apnoea as determined by ventilatory manoeuvres, creates a hypoxic environment with inhibited mechanotransduction in the arterioles and capillaries [23]. This triggers the production of reactive oxygen species (ROS) by endothelial cells, macrophages, and other immune cells [24]. The lung antioxidative mechanisms are overwhelmed upon reperfusion, resulting in an imbalance between ROS production and clearance [22]. Eventually, direct oxidative damage ensues with carbonylation of proteins and peroxynitration of proteins, lipids, and DNA [25]. Furthermore, ROS instigate a robust innate immune response by activating alveolar macrophages, which in turn release proinflammatory cytokines, including interleukin (IL)-8, -12, 18, and tumour necrosis factor (TNF)-a [26]. Additionally, neutrophils are recruited [26], and in concert with macrophages further enhance ROS generation; thus, a self-perpetuating cycle of oxidative stress enhancement is created [27, 28]. These activated leucocytes transmigrate into the extravascular space and cause increased microvascular permeability, thrombosis, oedema, and parenchymal cell death, by way of proteases, elastases, and ROS production [2]. Adherence of neutrophils to the endothelium further promotes the formation of gaps between the endothelial cells [6]. Moreover, these inflammatory cascades trigger platelet aggregation and coagulation, leading to formation of microthrombi and microvascular constriction [29]. The attendant activation of vasoactive agents, including thromboxane A2 and platelet activating factor, further promotes oedema formation [29, 30]. LIRI is also characterised by apoptotic phenomena, in part initiated by inflammatory cytokines (including IL-1β, -2, -8, and TNF-a) and enhanced by the release of proapoptotic factors due to mitochondrial rupture [31]. More specifically, the hypoxia-induced adenosine triphosphate (ATP) deficiency induces dysfunction of ATP-dependent ion pumps that results in mitochondrial calcium overload [29]. This leads to increased permeability of the mitochondrial transition pores and swelling, eventually leading to rupture [31]. Type II epithelial cells play a key role as both victim and as culprit of LIRI, as their reperfusion-induced dysfunction leads to impaired production and composition of pulmonary surfactant [32]. Crucially, the above described prooxidative and inflammatory signalling may exert systemic effects: on the one hand, distant IRI is known to cause pulmonary injury [33]; on the other hand, LIRI induces remote organ inflammatory and oxidative damage [34], while unilateral lung reperfusion may lead to similar deleterious processes in the contralateral, non-ischaemic lung [35] (Fig. 1).

The clinical impact of lung ischaemia reperfusion injury

The end result of LIRI is disruption of the alveolar-capillary barrier, causing non-cardiogenic pulmonary oedema and ventilation-perfusion (V/Q) [5]. Total and extravascular lung water is increased, causing gas exchange and lung mechanics impairment, with decreased arterial oxygen tension (PaO2) [36], increased airway pressures and increased alveolar-arterial oxygen gradient [(A-a) DO2] [37]. In addition, the attendant defective surfactant production and composition leads to reduced static (Cs) and dynamic lung compliance (Cd), while further increasing (A-a) DO2 [38]. Moreover, pulmonary vascular resistance (PVR) is increased up to three-fold following reperfusion, mainly due to pulmonary precapillary vasoconstriction [39]. Thus, LIRI is further compounded by pulmonary hypertension, which might also additionally hydrostatically promote the formation of pulmonary oedema [40].
In the absence of specific diagnostic criteria, LIRI is a diagnosis of exclusion, manifesting clinically as ALI or acute respiratory distress syndrome (ARDS) [41]. It has a detrimental impact in various clinical scenarios, particularly following lung transplantation: LIRI leads to PGD, which is the major cause of both short- and long-term morbidity and mortality in this setting [15, 42]. Importantly, the incidence of severe (grade 3) PGD within 72 h of transplantation is approximately 30% [42], while PGD is also associated with late graft rejection, which is the primary mortality aetiology beyond 1 year of the procedure [43, 44]. ALI/ARDS is frequent following lung resections, with a reported incidence of 0.88%, 2.96%, and 7.9% following sublobar, lobar/bilobar resection, and pneumonectomy respectively [21]. The associated mortality is considerable, ranging from 22% in sublobar resections to 50% following pneumonectomy [21]. LIRI is also a major source of morbidity and mortality after cardiac surgery, as it contributes to the development of ARDS, with an incidence of up to 20% and a mortality reaching 80% [45]. Interestingly, hepatosplanchnic IRI in aortic surgery has been found to cause lung injury, with increased pulmonary leak index (PLI) in up to 74% of the patients, especially in cases involving clamping of major aortic branches and in direct correlation with the aortic clamping time [46]. ALI also complicates 30–50% of major trauma cases, with an associated mortality of 10% depending on the severity of lung dysfunction [47].

The protective effects of ischemic conditioning on lung ischaemia reperfusion injury—effects on oxidative stress and inflammation

Experimental evidence

One of the critical mechanisms mediating the protective effects of IC against LIRI is the alleviation of oxidative stress and inflammation. Li et al. were among the first to demonstrate this, in a canine model of lung transplantation (in situ LIRI). Specifically, IC by donor pulmonary hilar occlusion/reperfusion before transplantation was associated with reduced infiltration of the transplanted lung interstitium by polymorphonuclear leukocytes (PMNs) and reduced blood levels of malondialdehyde (MDA) and thromboxane B2 (TXB2) following reperfusion; on the contrary, superoxide dismutase (SOD) levels were higher, denoting a preserved antioxidant reserve [48]. Similarly, in a rat model experiment, IC decreased thiobarbituric acid reactive substances (TBARS) after lung storage, reflecting the decreased MDA levels [49]. Focusing on the fluid shifts caused by enhanced oxidative stress and inflammation, Gasparri et al. showed that 15, but not 5, minutes of transient lung ischaemia before lung preservation mitigated lung oedema upon reperfusion [50]. Soncul et al. utilised isolated lungs mounted on a modified Langendorff perfusion apparatus, where IC was related with reduced tissue and perfusate MDA levels [51]. In a similar IC protocol, tissue MDA levels were reduced, glutathione levels increased, intra-alveolar oedema and capillary congestion were prevented, while type II alveolar and endothelial cells were preserved [52]. Li et al. applied IC before sustained lung ischemia–reperfusion to rabbits. Lung MDA levels were lower and SOD levels were higher in the preconditioned lungs, with an attendant reduction in lung oedema and alveolar damage [53]. Friedrich et al. applied IC on a canine model of LIRI, whereby the reperfusion insult was preceded by either a single 5-min occlusion with a 15-min reperfusion period or two successive 10-min ischemia–reperfusion stimuli, and was followed by bronchoalveolar lavage (BAL). Interestingly, the former protocol resulted in reduced BAL fluid protein and TNF-a content, while the latter had no effect, highlighting the importance of the IC stimulus duration [54]. Jun et al. elucidated the genetic background of IC: conditioning of the donor lung resulted in downregulation of a vast array of inflammatory and immune mediator genes, including IL-1, IL-2, IL-3, IL-6, IL-15, TNF-a, intercellular adhesion molecule-2 (ICAM-2), vascular cell adhesion molecule-1 (VCAM-1), and activated leukocyte adhesion molecules [55].
Remote ischemic conditioning (RIC) appears to have similar effects to local IC on in situ LIRI, as demonstrated by Song et al.: 6 cycles of 10-s aortic occlusion/reperfusion protected from LIRI induced by lung hilar clamping. In more detail, alveoli were preserved with less neutrophilic infiltration and oedema, the increase in lung wet-to-dry weight ratio was prevented, while plasma IL-6, TNF-a, and ROS levels were reduced [17]. Waldow et al. similarly concluded that RIC prevented the IL-1β increase and abrogated the lung macrophage infiltration induced by LIRI [20]. RIC by hepatic hilar clamping also ameliorated the increase in IL-6 and TNF-a, reduced myeloperoxidase (MPO) activity (reflecting the respective lung neutrophil accumulation) and the BAL fluid leucocyte levels, in parallel with inhibited alveolar damage and reduced wet-to-dry lung ratio; apoptotic cascades were attenuated, with decreased cleaved caspase-3 expression and fewer apoptotic nuclei [19]. An interesting study by Zhou et al. highlighted the protection conferred against CPB-induced LIRI: RIC alleviated intra-alveolar neutrophil infiltration and alveolar wall thickening, reduced BAL fluid protein levels and lung wet-to-dry weight ratio, in parallel with increased anti-inflammatory IL-4 and IL-10 [56].
Besides in situ LIRI, conditioning exerts protection in cases of remote organ reperfusion. In the setting of aortic occlusion and reperfusion, RIC was associated with reduced alveolar oedema, congestion, and neutrophil infiltration [18, 57]. Similar benefits have been derived from hepatosplanchnic conditioning, which abrogated the P-selectin upregulation caused by IRI, with an attendant reduction in alveolar and perivascular neutrophil infiltration, reduced MDA levels, and preserved vascular permeability [58]. Likewise, hepatic or mesenteric conditioning reduced inflammatory cell infiltration in animals undergoing hepatosplanchnic reperfusion [59]. In concordance with these findings, Meng et al. showed that mesenteric conditioning decreased plasma and lung tissue TNF-a and IL-6 levels, in contrast with an increase in IL-10 levels, SOD and glutathione peroxidase activity. Moreover, endothelial and alveolar epithelial architecture were preserved, as was the integrity of type II alveolar cell mitochondria, with concurrently diminished wet-to-dry lung weight ratio and pulmonary microvascular dysfunction [60]. However, dos Santos et al. did not reveal any histologic preservation by IC; interestingly, mesenteric IRI caused only minimal lung injury, thereby narrowing the margins for demonstrating a protective effect [61].
Harkin et al. investigated the effects of IC on limb reperfusion-induced LIRI, by way of external iliac artery occlusion/reperfusion. IC attenuated the increase in plasma IL-6 and phagocytic priming, without affecting the TNF-a levels; lung tissue MPO increase was also diminished, as was the elevation of weight-to-dry weight ratio [62]. A similar study focused on unilateral lower limb ischaemia, utilising a left lower limb tourniquet. RIC resulted in decreased plasma TBARS, as well as reduced PMN infiltration and MPO activity in the lung, while protecting from alveolar and interstitial oedema, and alveolar haemorrhage [63].
Haemorrhagic shock resuscitation is hampered by multiorgan failure (MOF), which is the commonest cause of death after severe trauma [64]. The role of systemic inflammation and oxidative stress is pivotal [65], while respiratory failure is recognised among the commonest and deadliest complications thereof [64]. Jan et al. studied the effects of lower limb tourniquet occlusion/reperfusion on a rat model of haemorrhagic shock. IC significantly reduced plasma IL-6 levels, in parallel with diminished lung IL-6, PGE2, MDA, and BAL fluid protein concentration. The levels of macrophage inflammatory protein-2 (MIP-2), MPO activity, PMN-to-alveoli and wet-to-dry weight ratios were also decreased, in association with mitigated alveolar wall oedema, haemorrhagic changes, vascular congestion, and PMN infiltration [66]. In a comparable study, RIC inhibited the rise in plasma TNF-a levels, as well as the lung TNF-a mRNA and protein expression following shock resuscitation. Lung MPO activity and protein leakage into BAL fluid were similarly reduced [67].
The suspension of ventilation, in concert with the resultant HPV, result in a hypoxic and hypoperfused lung environment that instigates LIRI [3, 5]. Preliminary evidence in this context have been contradictory. Bergmann et al. exploited RIC in a swine model of one lung ventilation (OLV). On the one hand, lung TNF-a and BAL fluid leucocyte levels were reduced; on the other hand, serum IL-1β and IL-8 were not affected, while microhaemorrhage and alveolar oedema of the ventilated lung were enhanced [68].
The marked heterogenicity of experimental IC protocols obviates reaching a safe conclusion with regards to their differential efficacy. Up to six cycles of ischaemia/reperfusion, with individual cycle ischaemic durations ranging from 10 s to 15 min have been successfully utilised. Thus, it may be inferred that a cumulative ischaemic stimulus of 1 and up to 30 min may confer biochemical protection from LIRI, although the most commonly applied protocols comprised three or four 5- or 10-min ischaemia–reperfusion cycles (Tables 1, 3; Fig. 2).
Table 1
Experimental studies of the ischaemic conditioning/remote ischaemic conditioning effects on lung ischaemia reperfusion injury
 
Model
Ischaemic conditioning protocol
Oxidative stress and inflammation
Respiratory function and pulmonary haemodynamics
Song et al. [17]
Rat lung in situ
Six aortic 10-s/10-s cycles
Reduced plasma IL-6, TNF-a, ROS, reduced alveolar PMN infiltration and oedema, reduced lung wet-to-dry ratio
Increased PaO2, reduced PaCO2
Dorsa et al. [18]
Rat abdominal aortic occlusion
Three 2-min/2-min abdominal aortic cycles
Reduced alveolar oedema, congestion, and PMN infiltration
NA
Luo et al. [19]
Rat lung in situ
Four 5-min/5 min hepatic hilar cycles
Reduced plasma IL-6 and TNF-a, reduced lung MPO activity and BAL fluid WBC count, reduced alveolar damage and lung wet-to-dry ratio, reduced lung caspase-3 expression and apoptotic nuclei
Increased PaO2, reduced PaCO2
Waldow et al. [20]
Porcine lung in situ
Three 5-min/5-min femoral arterial cycles
Reduced plasma IL-1β and macrophage count, reduced lung macrophage infiltration, plasma IL-6 and ROS not affected
Increased PaO2 and PvO2, reduced PAP and PVR
Li et al. [48]
Canine lung transplantation’
One 10-min-15-min donor lung cycle
Reduced lung PMN infiltration, reduced serum MDA and TXB2, increased SOD
Increased PvO2, reduced mPAP
Du et al. [49]
Rat lung transplantation
One 5 min/10-min donor lung cycle
Reduced TBARS
Increased PaO2, decreased PaCO2
Gasparri et al. [50]
Rabbit lung preservation
One lung 5-min/10-min vs three 5-min/10-min vs five 3-min/6-min cycles
Reduced lung oedema
Increased veno-arterial PO2 gradient observed following the conditioning protocols of 15-min total ischaemic stimulus
Soncul et al. [51]
Guinea pig lung preservation
Two lung 5-min/5 min cycles
Reduced lung tissue and perfusate MDA, reduced glutathione
Reduced PAP
Kandilci et al. [52]
Rat lung preservation
Two lung 5-min/5 min cycles
Reduced tissue MDA and glutathione, reduced intra-alveolar oedema and capillary congestion, type II alveolar and endothelial cell preservation
Reduced PAP
Li et al. [53]
Rabbit lung in situ
One lung 10-min/15-min cycle
Reduced lung MDA, lung oedema and alveolar damage, increased lung SOD
Increased PaO2, decreased mPAP
Friedrich et al. [54]
Canine lung in situ
One lung 5-min/15-min vs two 10-min/10-min cycles
Reduced BAL fluid protein and TNF-a following the 5-min ischaemic stimulus conditioning protocol
Increased PaO2 and PvO2, increased Cd, following the 5-min ischaemic stimulus conditioning protocol; neutral effects on PVR following both protocols
Jun et al. [55]
Rat lung transplantation
Three donor lung 5-min/5-min cycles
Inflammatory and immune mediator genes downregulation
NA
Zhou et al. [56]
Rat cardiopulmonary bypass
Three 5-min/5-min hind limb cycles
Reduced alveolar PMN infiltration and wall thickening, reduced BAL fluid protein, reduced lung wet-to-dry ratio, increased serum IL-4 and -10
Increased TLC and Cd, reduced Raw
Akahane et al. [57]
Rat abdominal aortic occlusion
Three 2-min/2-min abdominal aortic cycles
Reduced lung PMN infiltration and interstitial oedema
NA
Peralta et al. [58]
Rat hepatic ischaemia–reperfusion
One 10-min/10-min hepatic cycle
Reduced P-selectin upregulation, reduced lung PMN infiltration and MDA, preserved vascular permeability
NA
Neto et al. [59]
Rat hepatic/splanchnic ischaemia–reperfusion
One 10-min/10-min hepatic hilar/mesenteric arterial cycle
Reduced lung inflammatory cell infiltration
NA
Meng et al. [60]
Mouse splanchnic ischaemia–reperfusion
Three 30-s/30-s mesenteric arterial cycles
Reduced plasma and lung TNF-a and IL-6, increased plasma and lung IL-10, increased lung SOD and glutathione peroxidase activity, preserved endothelial and alveolar architecture
NA
Dos Santos et al. [61]
Rat splanchnic ischaemia–reperfusion
Two 2-min/2-min vs four 30-s/30-s mesenteric arterial cycles
Neutral effects on histological findings
NA
Harkin et al. [62]
Porcine limb ischaemia–reperfusion
Three 5-min/5-min external iliac arterial cycles
Reduced plasma IL-6 and phagocytic priming, reduced lung MPO and wet-to-dry weight ratio, neutral effect on plasma TNF-a
Increased PaO2, reduced (A-a) DO2, reduced mPAP
Olguner et al. [63]
Rat lower limb ischaemia–reperfusion
Three 10-min/10-min lower limb tourniquet cycles
Reduced plasma TBARS, reduced lung PMN infiltration and MPO activity, reduced lung oedema and alveolar haemorrhage
NA
Jan et al. [66]
Rat haemorrhagic shock
Three 10-min/10-min lower limb tourniquet cycles
Reduced plasma IL-6, reduced lung IL-6, PGE2, and MDA, reduced BAL fluid protein, reduced lung MIP-2, MPO activity, PMN-to-alveoli and wet-to-dry weight ratio, reduced histological injury
Increased PaO2, reduced (A-a) DO2, increased pH and BE
Leung et al. [67]
Mouse haemorrhagic shock
Four 5-min/5-min lower limb tourniquet cycles
Reduced plasma TNF-a, reduced lung TNF-a mRNA and protein expression, reduced lung MPO activity and BAL fluid protein
NA
Bergmann et al. [68]
Swine one lung ventilation
Three 5-min/5-min hind limb tourniquet cycles
Reduced lung TNF-a and BAL fluid WBC, neutral effect on serum IL-1β and -8, enhanced lung microhaemorrhage and alveolar oedema
Reduced oxygenation index, increased SvO2
Featherstone et al. [75]
Rat lung preservation
One lung 5-min/5-min vs one 10-min/5-min vs two 5-min/5-min lung cycles
NA
Increased lung compliance, neutral effects on oxygenation and PVR observed following all conditioning protocols
Kharbanda et al. [76]
Swine cardiopulmonary bypass
Four 5-min/5-min hind limb tourniquet cycles
NA
Reduced Raw and ventilation pressures, neutral effect on PVR
Xia et al. [77]
Sheep coronary arterial occlusion
Three 5-min/5-min iliac arterial cycles
NA
Increased PaO2 and P/F ratio, decreased PVR and PAP
(A-a) DO2 alveolar-arterial oxygen gradient, BAL bronchoalveolar lavage, Cd dynamic lung compliance, IC Ischaemic conditioning, IL interleukin, IRI ischaemia–reperfusion injury, MDA malondialdehyde, MIP-2 macrophage inflammatory protein-2, mPAP mean pulmonary artery pressure, MPO myeloperoxidase, NA not applicable, PaCO2 arterial carbon dioxide tension, PaO2 arterial oxygen tension, PAP pulmonary artery pressure, PMN polymorphonuclear leucocyte, PvO2 mixed venous oxygen tension, PVR pulmonary vascular resistance, P/F ratio ratio of the partial pressure of oxygen to the inspired fraction of oxygen, Raw airway resistance, RIC remote ischemic conditioning, ROS reactive oxygen species, SOD superoxide dismutase, SvO2 venous oxygen saturation, TBARS thiobarbituric acid reactive substances, TLC total lung capacity, TNF-a tumour necrosis factor-alpha, TXB2 thromboxane B2

Clinical evidence

The majority of clinical data pertaining to the investigation of IC effects on LIRI has been obtained in the context of CPB-induced IRI. Forty patients undergoing valve replacement were randomised to an IC or control group. Analysis of pulmonary vein blood showed that IC by way of aortic occlusion/reperfusion mitigated the increase in MDA, PMN, and TBX2 levels, while increasing SOD. Similarly, calcitonin gene-related peptide (CGRP) levels in coronary sinus blood were enhanced, denoting the activation of the associated anti-oxidant pathway, in parallel with reduced pulmonary oedema, haemorrhage, and PMN infiltration [10]. RIC has similar properties, as exemplified by Jin et al. who utilised upper limb and thigh cuff inflation/deflation in a randomised control trial (RCT) of 241 patients undergoing valvular replacement: conditioning reduced the levels of serum soluble intercellular adhesion molecule-1 (sICAM-1), endothelin-1 (ET-1), and MDA, while increasing NO concentration [69]. In an RCT of 60 infants undergoing ventricular septal defect (VSD) repair, Zhou et al. applied upper limb cuff inflation/deflation in the intervention group. Postoperatively, serum IL-6, -8, -10, and TNF-a levels were reduced, while coronary sinus MDA was reduced and SOD increased in preconditioned infants [12]. Nonetheless, these findings are not unequivocal. Lower limb cuff inflation/deflation was applied to children undergoing surgical repair of congenital heart defects, but the realised lung protection could not be associated with systemic inflammation, as IL-6, -8, -10, and TNF-a levels were not affected by RIC [70]. Similarly, Hu et al. conducted an RCT that included 201 patients undergoing valve replacement, whereby RIC did not affect hypersensitive C-reactive protein (hsCRP) levels [11].
IC has been utilised in patients undergoing thoracic surgical operations. Chen et al. have been among the first to implement preconditioning in patients undergoing pneumonectomy: in a small study of 20 patients, a single pulmonary artery occlusion/reperfusion manoeuvre resulted in increased CGRP and SOD levels [71]. Li et al. randomised 216 patients undergoing elective lung resection to either a RIC, or a standard thoracic surgical treatment arm. Upper limb cuff inflation/deflation significantly reduced IL-6, TNF-a, and MDA levels post-operatively [7]. An analogous RCT was performed in 55 patients undergoing lobectomy for lung cancer, whose blood and exhaled breath condensate (EBC) were examined to determine the levels of oxidative stress. RIC mitigated oxidative stress, as denoted by reduced 8-isoprostane, cumulative nitrate and nitrite, hyperoxide, and acidity in the EBC samples of preconditioned patients; blood 8-isoprostane as well as cumulative nitrate and nitrite were also reduced in the RIC arm [8]. These findings were not reproduced in the pilot RCT by Lin et al. who randomised 60 patients undergoing bilateral sequential lung transplantation to a RIC or a standard treatment arm: IL-2, -6, -8, -10, TNF-a, interferon-gamma (IFN-γ), interferon gamma-induced protein 10, monocyte chemoattractant protein-1 (MCP-1), and chemokine ligand 5 (CCL5) did not differ between the two study groups [72].
Aortic cross-clamping during open abdominal aortic aneurysm repair (AAA) is known to trigger a systemic inflammatory response with an attendant accentuation of oxidative stress [33]; these cascades may progress to MOF, which underlies up to 25% of peri-operative deaths in this setting [73]. Against this background, the effects of RIC in the form of upper limb cuff inflation/deflation were evaluated in an RCT of 62 patients undergoing open AAA repair. RIC attenuated the post-operative increase in IL-6, TNF-a, and MDA levels, while increasing the activity of SOD [13]. Limb reperfusion may also induce MOF through comparable mechanisms [74]. Lin et al. studied 30 patients undergoing lower extremity surgery, necessitating sustained thigh tourniquet application. When this was preceded by IC, the rise in plasma IL-6, -8, and MDA after limb reperfusion was mitigated [14].
Similar to experimental investigations, clinical IC protocols have not been standardised. However, three 5-min ischaemia–reperfusion cycles have been most commonly utilised and may confer a biochemically determined protection from LIRI in a variety of clinical settings, although some studies have shown a neutral effect (Tables 2, 3; Fig. 2).
Table 2
Clinical studies of the ischaemic conditioning/remote ischaemic conditioning effects on lung ischaemia reperfusion injury
Study
Clinical setting
Ischaemic conditioning protocol
Oxidative stress and inflammation
Respiratory function and pulmonary haemodynamics
Li et al. [7]
Lung resection
Three 5-min/5-min arm cuff cycles
Reduced serum IL-6, TNF-a, and MDA
Increased P/F and a/A ratio, reduced (A-a) DO2, reduced ALI incidence, increased Cs and Cd
García-de-la-Asunción et al. [8]
Lobectomy
Three 5-min/5-min arm cuff cycles
Reduced EBC 8-isoprostane, nitrates and nitrites, hyperoxide, and acidity, reduced blood 8-isoprostane, nitrates and nitrites, neutral effect on CRP
Increased PaO2, P/F and a/A ratio, decreased (A-a) DO2 and RI
Li et al. [10]
Valve replacement (cardiopulmonary bypass)
Two 3-min/2-min aortic cycles
Reduced pulmonary vein MDA, PMN, and TBX2, increased SOD, increased coronary sinus CGRP, reduced lung oedema, haemorrhage, and PMN infiltration
Reduced PVRI and mPAP, increased PaO2, reduced pulmonary complications (atelectasis, pneumonitis, pneumothorax)
Hu et al. [11]
Valve replacement (cardiopulmonary bypass)
Three 5-min/5-min thigh cycles
Neutral effect on serum hsCRP
Reduced ALI incidence, neutral effect on A-aO2
Zhou et al. [12]
Infantile ventricular septal defect repair (cardiopulmonary bypass)
Three 5-min/5-min arm cuff cycles
Reduced serum IL-6, -8, -10, and TNF-a, reduced coronary sinus MDA, increased coronary sinus SOD
Reduced RI, increased Cs and Cd
Li et al. [13]
Abdominal aortic aneurysm repair
Three 5-min/5-min arm cuff cycles
Reduced plasma IL-6, TNF-a, and MDA, increased SOD
Increased a/A ratio, reduced (A-a) DO2 and RI, increased Cs and Cd
Lin et al. [14]
Lower limb surgery
Three 5-min/5-min thigh tourniquet cycles
Reduced plasma IL-6, -8, and MDA
Increased PaO2 and a/A ratio, reduced (A-a) DO2 and RI
Jin et al. [69]
Valve replacement (cardiopulmonary bypass)
Two 5-min/5-min arm and thigh cuff cycles
Reduced serum sICAM-1, ET-1, and MDA, increased NO
Reduced (A-a) DO2, RI, ALI incidence
Cheung et al. [70]
Children congenital heart defect repair (cardiopulmonary bypass)
Four 5-min/5-min thigh cuff cycles
Neutral effect on serum IL-6, -8, -10, and TNF-a
Reduced Paw, neutral effect on oxygenation and compliance
Chen et al. [71]
Pneumonectomy
One 10-min/10-min pulmonary arterial cycle
Increased serum CGRP and SOD
Increased PvO2
Lin et al. [72]
Lung transplantation
Three 5-min/5-min lower limb cuff cycles
Neutral effect on IL-2, -6, -8, -10, TNF-a, IFN-γ, interferon gamma-induced protein 10, MCP-1, and CCL5
Trend for decreased PGD and biopsy-proven rejection risk, increased P/F ratio in restrictive lung disease group
Min et al. [78]
Valve replacement (cardiopulmonary bypass)
Four 5-min/5-min arm cuff cycles
NA
Increased P/F ratio, reduced need for mechanical ventilation > 48 h, neutral effect on Cs and Cd
Hong et al. [79]
Off-pump coronary artery bypass grafting
Four 5-min/5-min lower limb cuff cycles applied twice
NA
Neutral effect on oxygenation and duration of mechanical ventilation
Kim et al. [80]
Valvular heart surgery (cardiopulmonary bypass)
Three 10-min/10-min lower limb cuff cycles applied twice
NA
Neutral effect on respiratory function and outcomes
Rahman et al. [81]
Coronary artery bypass grafting (cardiopulmonary bypass)
Three 5-min/5-min arm cuff cycles
NA
Neutral effect on respiratory function and outcomes
Lee et al. [82]
Infantile ventricular septal defect repair (cardiopulmonary bypass)
Four 5-min/5-min thigh cuff cycles
NA
Neutral effect on respiratory function and outcomes
ALI acute lung injury, (A-a) DO2 alveolar-arterial oxygen gradient, CCL5 chemokine ligand 5, Cd dynamic lung compliance, CGRP calcitonin gene-related peptide, Cs static lung compliance, EBC exhaled breath condensate, ET-1 endothelin-1, hsCRP hypersensitive C-reactive protein, IC Ischaemic conditioning, IFN-γ interferon-gamma, IL interleukin, IRI ischaemia–reperfusion injury, MCP-1 monocyte chemoattractant protein-1, MDA malondialdehyde, mPAP mean pulmonary artery pressure, NA not applicable, PaO2 arterial oxygen tension, PAP pulmonary artery pressure, Paw airway pressure, PGD primary graft dysfunction, PMN polymorphonuclear leucocyte, PvO2 mixed venous oxygen tension, PVRI pulmonary vascular resistance index, P/F ratio ratio of the partial pressure of oxygen to the inspired fraction of oxygen, Raw airway resistance, RIC remote ischemic conditioning, sICAM-1 serum soluble intercellular adhesion molecule-1, SOD superoxide dismutase, TNF-a tumour necrosis factor-alpha
Table 3
Summary of the protective effects of ischaemic conditioning from lung ischaemia–reperfusion injury
Ischaemic conditioning protective effects from lung ischaemia reperfusion injury
Studies
↓ Leukocyte emigration
Li et al. [10]; Song et al. [17]; Dorsa et al. [18]; Luo et al. [19]; Waldow et al. [20]; Li et al. [48]; Zhou et al. [56]; Akahane et al. [57]; Peralta et al. [58]; Neto et al. [59]; Harkin et al. [62]; Olguner et al. [63]; Jan et al. [66]; Leung et al. [67]; Bergmann et al. [68]; Jin et al. [69]
↓ Oxidative stress
Li et al. [7]; García-de-la-Asunción et al. [8]; Li et al. [10]; Zhou et al. [12]; Li et al. [13]; Lin et al. [14]; Song et al. [17]; Li et al. [48]; Du et al. [49]; Soncul et al. [51]; Kandilci et al. [52]; Li et al. [53]; Peralta et al. [58]; Meng et al. [60]; Olguner et al. [63]; Jan et al. [66]; Jin et al. [69]; Chen et al. [71];
↓ Inflammatory/
↑Anti-inflammatory cytokines
Zhou et al. [12]; Li et al. [7]; Li et al. [13]; Lin et al. [14]; Song et al. [17]; Luo et al. [19]; Waldow et al. [20]; Zhou et al. [56]; Meng et al. [60]; Leung et al. [67]; Bergmann et al. [68]
Inflammatory genes downregulation
Jun et al. [55]
↓ Pulmonary vasoconstrictors
Li et al. [10]; Li et al. [48]; Jin et al. [69]
↓ Non-cardiogenic pulmonary oedema
Li et al. [10]; Song et al. [17]; Dorsa et al. [18]; Luo et al. [19]; Gasparri et al. [50]; Kandilci et al. [52]; Li et al. [53]; Friedrich et al. [54]; Zhou et al. [56]; Akahane et al. [57]; Peralta et al. [58]; Meng et al. [60]; Harkin et al. [62]; Olguner et al. [63]; Jan et al. [66]
Improved gas exchange
Li et al. [7]; García-de-la-Asunción et al. [8]; Li et al. [10]; Zhou et al. [12]; Li et al. [13]; Lin et al. [14]; Song et al. [17]; Luo et al. [19]; Waldow et al. [20]; Li et al. [48]; Du et al. [49]; Li et al. [53]; Friedrich et al. [54]; Harkin et al. [62]; Jan et al. [66]; Bergmann et al. [68]; Jin et al. [69]; Chen et al. [71]; Lin et al. [72]; Xia et al. [77]; Min et al. [78]
Improved lung mechanics
Li et al. [7]; Zhou et al. [12]; Li et al. [13]; Friedrich et al. [54]; Cheung et al. [70]; Featherstone et al. [75]; Kharbanda et al. [76]
Improved pulmonary haemodynamics
Li et al. [10]; Waldow et al. [20]; Li et al. [48]; Soncul et al. [51]; Kandilci et al. [52]; Li et al. [53]; Harkin et al. [62]; Xia et al. [77]

The protective effects of ischemic conditioning on lung ischaemia reperfusion injury—effects on respiratory function and pulmonary haemodynamics

Experimental evidence

The oxidative stress and inflammatory response alleviation conferred by IC may be functionally translated into improved respiratory function and pulmonary haemodynamics. In the canine transplantation model studied by Li et al. donor lung IC was associated with increased mixed venous oxygen tension (PvO2) and reduced mean pulmonary artery pressure (mPAP) following reperfusion [48]. Du et al. also provided evidence of improved gas exchange, as denoted by higher arterial oxygen (PaO2) and decreased carbon dioxide tension (PaCO2) levels [49]. Similar oxygenation improvement has been demonstrated in the form of increased veno-arterial oxygen pressure gradients [50], while Soncul et al. concluded that IC ameliorated the increase in pulmonary artery pressure (PAP) caused by LIRI [51]. IC also preserved pulmonary arterial endothelial function in the study of Kandilci et al., reflected on decreased pulmonary perfusion pressures in response to histamine [52]. Further expanding the evidence above, Li et al. showed that PO2 is increased and mPAP decreased by IC [53]. Data of improved Cd have also been provided, in parallel with higher PaO2 and PvO2 levels; however, in the same study pulmonary vascular resistance (PVR) was not significantly affected [54]. Featherstone et al. reported analogous improvement in lung compliance; nonetheless, oxygenation and PVR did not differ between the preconditioned and the control groups [75].
RIC exerts comparable protective effects: gas exchange was significantly improved in the context of an in situ LIRI model, with increased PaO2 and decreased PaCO2, in the studies of Luo et al. [19] and Song et al. [17]. Waldow et al. showed that RIC improved PaO2 and PvO2, in parallel with a reduction in PAP and PVR [20]. In a setting of CPB-induced LIRI, evidence of superior total lung capacity (TLC) and Cd, as well as reduced airway resistance (Raw) was provided [56]. Comparably, Raw was lower upon exposure to CPB with a resultant decrease in ventilation pressures and a trend for lung compliance improvement in the study of Kharbanda et al. [76]. In an interesting study of coronary occlusion/reperfusion, simulating an off-pump coronary artery bypass stimulus, RIC increased PaO2 and P/F ratio, while decreasing PVR and PAP [77].
Investigating LIRI within the realms of remote reperfusion injury, Harkin et al. showed an improvement in PaO2 and (A-a) DO2 with an attendant reduction in mPAP following limb reperfusion in preconditioned animals [62]. Jan et al. demonstrated similar protective properties after resuscitation from haemorrhagic shock, with increased PaO2 and decreased (A-a) DO2, with an attendant amelioration of acidosis, as pH and base excess were increased by IC [66]. Interestingly, Bergmann et al. highlighted the improved respiratory function conferred by IC during OLV: oxygenation index [fraction of inspired oxygen (FIO2) * mean airway pressure (Paw mean)/PaO2)] was lower after RIC, with an attendant increase in venous oxygen saturation (SvO2) [68].
In summary, most experimental studies have reported improvements in gas exchange, respiratory mechanics, and pulmonary haemodynamics by IC. Interestingly, local conditioning appears to exert similar effects to RIC, both in settings of in situ as well as remote reperfusion injury. Thus, the systemic nature of the underlying processes has been consistently demonstrated (Tables 1, 3; Fig. 2).

Clinical evidence

There is accumulating clinical evidence delineating the effects of IC against LIRI in several pathophysiological contexts. Li et al. investigated the application if IC in patients exposed to CPB and revealed that preconditioning reduced PVRI and mPAP, while increasing PaO2; importantly, there was an attendant reduction in pulmonary complications (lobar collapse, pneumonitis, pneumothorax) and mechanical ventilation time [10]. In a similar cohort of patients, RIC conferred a decrease in (A-a) DO2, respiratory index [(A-a) DO2/PaO2], as well as the incidence of ALI [69]. Zhou et al. also concluded that RIC improved RI and decreased static (Cs) and dynamic lung compliance (Cd) [12], while the incidence of ALI was reduced in patients undergoing RIC during their valve replacement, despite the fact that (A-a) DO2 was not affected [11]. Moreover, RIC applied both before and after CBP in patients undergoing valvular replacement improved P/F ratio and reduced the incidence of mechanical ventilation beyond 48 h; Cs and Cd remained unchanged [78]. However, Cheung et al. did not reproduce any improvements in either oxygenation or compliance; nonetheless, Raw was reduced in preconditioned patients [70]. Hong et al. also showed a neutral effect of RIC on oxygenation and mechanical ventilation duration; however, the included patients underwent off-pump CABG, which obviates the exposure to CPB [79]. Additional studies have shown a lack of improvement in respiratory indices or outcomes, but no inflicted harm [8082].
The results of the studies focusing on patients undergoing lung resections uniformly support the notion that IC confers significant protection from LIRI. RIC abrogated the deleterious effects of OLV on respiratory function, as denoted by improvements in P/F ratio, (A-a) DO2, and arterial-alveolar oxygen tension ratio (a/A ratio), with an attendant decreased ALI incidence; Cs and Cd were also significantly increased [7]. Concordant evidence was provided by García-de-la-Asunción et al., who showed an improvement in PaO2, (A-a) DO2, P/F and a/A ratio, and RI [8]. Chen et al. also demonstrated increased pulmonary venous oxygen tension in patients undergoing pneumonectomy [71]. In an important pilot RCT of RIC within the context of lung transplantation by Lin et al., preconditioning was associated with a trend for decreased PGD and biopsy-proven rejection risk, while P/F ratio was significantly increased in the subgroup of patients with restrictive lung disease [72]. RIC protects from the lung dysfunction triggered by splanchnic IRI, as shown by the RCT of Lin et al., whereby preconditioned patients undergoing AAA repair benefited from higher a/A ratio, lower (A-a) DO2 and RI, as well as improved Cs and Cd [13]. Comparable effects have been demonstrated following lower limb reperfusion: RIC resulted in improved PaO2, a/A ratio, (A-a) DO2, and RI [14].
Thus, despite the contradictory evidence provided by studies of cardiac surgery patients, the majority of data obtained in clinical settings that entail in situ lung, as well as hepatosplanchnic and limb reperfusion underlines the beneficial effects obtained from IC: respiratory function and ventilation mechanics may improve, while respiratory complications may be averted (Tables 2, 3; Fig. 2).

Conclusions

LIRI has a detrimental effect on respiratory function and pulmonary haemodynamics, with dismal consequences for patient prognosis in a variety of clinical settings. The abundance of experimental evidence revealing the beneficial effects of IC, both on the underlying inflammatory and oxidative cascades, as well as the resultant functional derangements is being gradually complemented by encouraging clinical data. Given its promising preliminary results, safety, and ease of application, IC appears to be an intervention worth of further investigation and a useful addition to our deficient armamentarium against LIRI.

Acknowledgements

Nil.

Declarations

Not applicable.
Not applicable.

Competing interests

The authors declare that they do not have any competing interests.
Open AccessThis 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.

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Literatur
1.
Zurück zum Zitat Cowled P, Fitridge R. Pathophysiology of reperfusion injury. In: Fitridge R, Thompson M, editors. Mechanisms of vascular disease: a reference book for vascular specialists. Adelaide (AU): University of Adelaide Press; 2011. p. 18. Cowled P, Fitridge R. Pathophysiology of reperfusion injury. In: Fitridge R, Thompson M, editors. Mechanisms of vascular disease: a reference book for vascular specialists. Adelaide (AU): University of Adelaide Press; 2011. p. 18.
2.
Zurück zum Zitat Eltzschig HK, Collard CD. Vascular ischaemia and reperfusion injury. Br Med Bull. 2004;70:71–86.CrossRef Eltzschig HK, Collard CD. Vascular ischaemia and reperfusion injury. Br Med Bull. 2004;70:71–86.CrossRef
3.
Zurück zum Zitat Allison RC, Kyle J, Adkins WK, Prasad VR, McCord JM, Taylor AE. Effect of ischemia reperfusion or hypoxia reoxygenation on lung vascular permeability and resistance. J Appl Physiol. 1985;1990(69):597–603. Allison RC, Kyle J, Adkins WK, Prasad VR, McCord JM, Taylor AE. Effect of ischemia reperfusion or hypoxia reoxygenation on lung vascular permeability and resistance. J Appl Physiol. 1985;1990(69):597–603.
4.
Zurück zum Zitat Laubach VE, Sharma AK. Mechanisms of lung ischemia-reperfusion injury. Curr Opin Organ Transplant. 2016;21:246–52.CrossRef Laubach VE, Sharma AK. Mechanisms of lung ischemia-reperfusion injury. Curr Opin Organ Transplant. 2016;21:246–52.CrossRef
5.
Zurück zum Zitat Marshall BE, Marshall C, Benumof J, Saidman LJ. Hypoxic pulmonary vasoconstriction in dogs: effects of lung segment size an oxygen tension. J Appl Physiol. 1981;51:1543–51.CrossRef Marshall BE, Marshall C, Benumof J, Saidman LJ. Hypoxic pulmonary vasoconstriction in dogs: effects of lung segment size an oxygen tension. J Appl Physiol. 1981;51:1543–51.CrossRef
6.
Zurück zum Zitat Talaie T, DiChiacchio L, Prasad NK, et al. Ischemia-reperfusion injury in the transplanted lung: a literature review. Transplant Direct. 2021;7: e652.CrossRef Talaie T, DiChiacchio L, Prasad NK, et al. Ischemia-reperfusion injury in the transplanted lung: a literature review. Transplant Direct. 2021;7: e652.CrossRef
7.
Zurück zum Zitat Li C, Xu M, Wu Y, Li YS, Huang WQ, Liu KX. Limb remote ischemic preconditioning attenuates lung injury after pulmonary resection under propofol-remifentanil anesthesia: a randomized controlled study. Anesthesiology. 2014;121:249–59.CrossRef Li C, Xu M, Wu Y, Li YS, Huang WQ, Liu KX. Limb remote ischemic preconditioning attenuates lung injury after pulmonary resection under propofol-remifentanil anesthesia: a randomized controlled study. Anesthesiology. 2014;121:249–59.CrossRef
8.
Zurück zum Zitat García-de-la-Asunción J, Bruno L, Perez-Griera J, et al. Remote ischemic preconditioning decreases oxidative lung damage after pulmonary lobectomy: a single-center randomized, double-blind. Controlled Trial Anesth Analg. 2017;125:499–506.CrossRef García-de-la-Asunción J, Bruno L, Perez-Griera J, et al. Remote ischemic preconditioning decreases oxidative lung damage after pulmonary lobectomy: a single-center randomized, double-blind. Controlled Trial Anesth Analg. 2017;125:499–506.CrossRef
9.
Zurück zum Zitat Suzuki T. Additional lung-protective perfusion techniques during cardiopulmonary bypass. Ann Thorac Cardiovasc Surg. 2010;16:150–5. Suzuki T. Additional lung-protective perfusion techniques during cardiopulmonary bypass. Ann Thorac Cardiovasc Surg. 2010;16:150–5.
10.
Zurück zum Zitat Li G, Chen S, Lu E, Luo W. Cardiac ischemic preconditioning improves lung preservation in valve replacement operations. Ann Thorac Surg. 2001;71:631–5.CrossRef Li G, Chen S, Lu E, Luo W. Cardiac ischemic preconditioning improves lung preservation in valve replacement operations. Ann Thorac Surg. 2001;71:631–5.CrossRef
11.
Zurück zum Zitat Hu Q, Luo W, Huang L, Huang R, Chen R, Gao Y. Multiorgan protection of remote ischemic perconditioning in valve replacement surgery. J Surg Res. 2016;200:13–20.CrossRef Hu Q, Luo W, Huang L, Huang R, Chen R, Gao Y. Multiorgan protection of remote ischemic perconditioning in valve replacement surgery. J Surg Res. 2016;200:13–20.CrossRef
12.
Zurück zum Zitat Zhou W, Zeng D, Chen R, et al. Limb ischemic preconditioning reduces heart and lung injury after an open heart operation in infants. Pediatr Cardiol. 2010;31:22–9.CrossRef Zhou W, Zeng D, Chen R, et al. Limb ischemic preconditioning reduces heart and lung injury after an open heart operation in infants. Pediatr Cardiol. 2010;31:22–9.CrossRef
13.
Zurück zum Zitat Li C, Li YS, Xu M, et al. Limb remote ischemic preconditioning for intestinal and pulmonary protection during elective open infrarenal abdominal aortic aneurysm repair: a randomized controlled trial. Anesthesiology. 2013;118:842–52.CrossRef Li C, Li YS, Xu M, et al. Limb remote ischemic preconditioning for intestinal and pulmonary protection during elective open infrarenal abdominal aortic aneurysm repair: a randomized controlled trial. Anesthesiology. 2013;118:842–52.CrossRef
14.
Zurück zum Zitat Lin LN, Wang LR, Wang WT, et al. Ischemic preconditioning attenuates pulmonary dysfunction after unilateral thigh tourniquet-induced ischemia-reperfusion. Anesth Analg. 2010;111:539–43.CrossRef Lin LN, Wang LR, Wang WT, et al. Ischemic preconditioning attenuates pulmonary dysfunction after unilateral thigh tourniquet-induced ischemia-reperfusion. Anesth Analg. 2010;111:539–43.CrossRef
15.
Zurück zum Zitat den Hengst WA, Gielis JF, Lin JY, Van Schil PE, De Windt LJ, Moens AL. Lung ischemia-reperfusion injury: a molecular and clinical view on a complex pathophysiological process. Am J Physiol Heart Circ Physiol. 2010;299:H1283–99.CrossRef den Hengst WA, Gielis JF, Lin JY, Van Schil PE, De Windt LJ, Moens AL. Lung ischemia-reperfusion injury: a molecular and clinical view on a complex pathophysiological process. Am J Physiol Heart Circ Physiol. 2010;299:H1283–99.CrossRef
16.
Zurück zum Zitat Heusch G, Bøtker HE, Przyklenk K, Redington A, Yellon D. Remote ischemic conditioning. J Am Coll Cardiol. 2015;65:177–95.CrossRef Heusch G, Bøtker HE, Przyklenk K, Redington A, Yellon D. Remote ischemic conditioning. J Am Coll Cardiol. 2015;65:177–95.CrossRef
17.
Zurück zum Zitat Song SQ, Gan HL, Zhang JQ, Feng L, Sun JC, Wang SX. Post-conditioning through lower limb ischemia-reperfusion can alleviate lung ischemia-reperfusion injury. Int J Clin Exp Med. 2015;8:14953–61. Song SQ, Gan HL, Zhang JQ, Feng L, Sun JC, Wang SX. Post-conditioning through lower limb ischemia-reperfusion can alleviate lung ischemia-reperfusion injury. Int J Clin Exp Med. 2015;8:14953–61.
18.
Zurück zum Zitat Dorsa RC, Pontes JC, Antoniolli AC, et al. Effect of remote ischemic postconditioning in inflammatory changes of the lung parenchyma of rats submitted to ischemia and reperfusion. Rev Bras Cir Cardiovasc. 2015;30:353–9. Dorsa RC, Pontes JC, Antoniolli AC, et al. Effect of remote ischemic postconditioning in inflammatory changes of the lung parenchyma of rats submitted to ischemia and reperfusion. Rev Bras Cir Cardiovasc. 2015;30:353–9.
19.
Zurück zum Zitat Luo N, Liu J, Chen Y, Li H, Hu Z, Abbott GW. Remote ischemic preconditioning STAT3-dependently ameliorates pulmonary ischemia/reperfusion injury. PLoS ONE. 2018;13: e0196186.CrossRef Luo N, Liu J, Chen Y, Li H, Hu Z, Abbott GW. Remote ischemic preconditioning STAT3-dependently ameliorates pulmonary ischemia/reperfusion injury. PLoS ONE. 2018;13: e0196186.CrossRef
20.
Zurück zum Zitat Waldow T, Alexiou K, Witt W, et al. Protection against acute porcine lung ischemia/reperfusion injury by systemic preconditioning via hind limb ischemia. Transpl Int. 2005;18:198–205.CrossRef Waldow T, Alexiou K, Witt W, et al. Protection against acute porcine lung ischemia/reperfusion injury by systemic preconditioning via hind limb ischemia. Transpl Int. 2005;18:198–205.CrossRef
21.
Zurück zum Zitat Dulu A, Pastores SM, Park B, Riedel E, Rusch V, Halpern NA. Prevalence and mortality of acute lung injury and ARDS after lung resection. Chest. 2006;130:73–8.CrossRef Dulu A, Pastores SM, Park B, Riedel E, Rusch V, Halpern NA. Prevalence and mortality of acute lung injury and ARDS after lung resection. Chest. 2006;130:73–8.CrossRef
22.
Zurück zum Zitat Pak O, Sydykov A, Kosanovic D, et al. Lung ischaemia-reperfusion injury: the role of reactive oxygen species. Adv Exp Med Biol. 2017;967:195–225.CrossRef Pak O, Sydykov A, Kosanovic D, et al. Lung ischaemia-reperfusion injury: the role of reactive oxygen species. Adv Exp Med Biol. 2017;967:195–225.CrossRef
23.
Zurück zum Zitat Lansman JB. Endothelial mechanosensors. Going with the flow. Nature. 1988;331:481–2.CrossRef Lansman JB. Endothelial mechanosensors. Going with the flow. Nature. 1988;331:481–2.CrossRef
24.
Zurück zum Zitat Zhao G, Al-Mehdi AB, Fisher AB. Anoxia-reoxygenation versus ischemia in isolated rat lungs. Am J Physiol. 1997;273:L1112–7. Zhao G, Al-Mehdi AB, Fisher AB. Anoxia-reoxygenation versus ischemia in isolated rat lungs. Am J Physiol. 1997;273:L1112–7.
25.
Zurück zum Zitat Chen X, Chen H, Deng R, Shen J. Pros and cons of current approaches for detecting peroxynitrite and their applications. Biomed J. 2014;37:120–6.CrossRef Chen X, Chen H, Deng R, Shen J. Pros and cons of current approaches for detecting peroxynitrite and their applications. Biomed J. 2014;37:120–6.CrossRef
26.
Zurück zum Zitat Schröder K. NADPH oxidases in redox regulation of cell adhesion and migration. Antioxid Redox Signal. 2014;20:2043–58.CrossRef Schröder K. NADPH oxidases in redox regulation of cell adhesion and migration. Antioxid Redox Signal. 2014;20:2043–58.CrossRef
27.
Zurück zum Zitat Mortaz E, Alipoor SD, Adcock IM, Mumby S, Koenderman L. Update on neutrophil function in severe inflammation. Front Immunol. 2018;9:2171.CrossRef Mortaz E, Alipoor SD, Adcock IM, Mumby S, Koenderman L. Update on neutrophil function in severe inflammation. Front Immunol. 2018;9:2171.CrossRef
28.
Zurück zum Zitat Short JD, Downs K, Tavakoli S, Asmis R. Protein thiol redox signaling in monocytes and macrophages. Antioxid Redox Signal. 2016;25:816–35.CrossRef Short JD, Downs K, Tavakoli S, Asmis R. Protein thiol redox signaling in monocytes and macrophages. Antioxid Redox Signal. 2016;25:816–35.CrossRef
29.
Zurück zum Zitat Ferrari RS, Andrade CF. Oxidative stress and lung ischemia-reperfusion injury. Oxid Med Cell Longev. 2015;2015: 590987.CrossRef Ferrari RS, Andrade CF. Oxidative stress and lung ischemia-reperfusion injury. Oxid Med Cell Longev. 2015;2015: 590987.CrossRef
30.
Zurück zum Zitat Dixon JT, Gozal E, Roberts AM. Platelet-mediated vascular dysfunction during acute lung injury. Arch Physiol Biochem. 2012;118:72–82.CrossRef Dixon JT, Gozal E, Roberts AM. Platelet-mediated vascular dysfunction during acute lung injury. Arch Physiol Biochem. 2012;118:72–82.CrossRef
31.
Zurück zum Zitat Kalogeris T, Bao Y, Korthuis RJ. Mitochondrial reactive oxygen species: a double edged sword in ischemia/reperfusion vs preconditioning. Redox Biol. 2014;2:702–14.CrossRef Kalogeris T, Bao Y, Korthuis RJ. Mitochondrial reactive oxygen species: a double edged sword in ischemia/reperfusion vs preconditioning. Redox Biol. 2014;2:702–14.CrossRef
32.
Zurück zum Zitat Novick RJ, Gehman KE, Ali IS, Lee J. Lung preservation: the importance of endothelial and alveolar type II cell integrity. Ann Thorac Surg. 1996;62:302–14.CrossRef Novick RJ, Gehman KE, Ali IS, Lee J. Lung preservation: the importance of endothelial and alveolar type II cell integrity. Ann Thorac Surg. 1996;62:302–14.CrossRef
33.
Zurück zum Zitat Bown MJ, Nicholson ML, Bell PR, Sayers RD. Cytokines and inflammatory pathways in the pathogenesis of multiple organ failure following abdominal aortic aneurysm repair. Eur J Vasc Endovasc Surg. 2001;22:485–95.CrossRef Bown MJ, Nicholson ML, Bell PR, Sayers RD. Cytokines and inflammatory pathways in the pathogenesis of multiple organ failure following abdominal aortic aneurysm repair. Eur J Vasc Endovasc Surg. 2001;22:485–95.CrossRef
34.
Zurück zum Zitat Esme H, Fidan H, Koken T, Solak O. Effect of lung ischemia–reperfusion on oxidative stress parameters of remote tissues. Eur J Cardiothorac Surg. 2006;29:294–8.CrossRef Esme H, Fidan H, Koken T, Solak O. Effect of lung ischemia–reperfusion on oxidative stress parameters of remote tissues. Eur J Cardiothorac Surg. 2006;29:294–8.CrossRef
35.
Zurück zum Zitat Palazzo R, Hamvas A, Shuman T, Kaiser L, Cooper J, Schuster DP. Injury in nonischemic lung after unilateral pulmonary ischemia with reperfusion. J Appl Physiol. 1985;1992(72):612–20. Palazzo R, Hamvas A, Shuman T, Kaiser L, Cooper J, Schuster DP. Injury in nonischemic lung after unilateral pulmonary ischemia with reperfusion. J Appl Physiol. 1985;1992(72):612–20.
36.
Zurück zum Zitat Jurmann MJ, Dammenhayn L, Schaefers HJ, Haverich A. Pulmonary reperfusion injury: evidence for oxygen-derived free radical mediated damage and effects of different free radical scavengers. Eur J Cardiothorac Surg. 1990;4:665–70.CrossRef Jurmann MJ, Dammenhayn L, Schaefers HJ, Haverich A. Pulmonary reperfusion injury: evidence for oxygen-derived free radical mediated damage and effects of different free radical scavengers. Eur J Cardiothorac Surg. 1990;4:665–70.CrossRef
37.
Zurück zum Zitat Qayumi AK, Jamieson WR, Godin DV, Lam S, Ko KM, Germann E, Van den Broek J. Response to allopurinol pretreatment in a swine model of heart-lung transplantation. J Invest Surg. 1990;3:331–40.CrossRef Qayumi AK, Jamieson WR, Godin DV, Lam S, Ko KM, Germann E, Van den Broek J. Response to allopurinol pretreatment in a swine model of heart-lung transplantation. J Invest Surg. 1990;3:331–40.CrossRef
38.
Zurück zum Zitat Sievers HH, Freund-Kaas C, Eleftheriadis S, et al. Lung protection during total cardiopulmonary bypass by isolated lung perfusion: preliminary results of a novel perfusion strategy. Ann Thorac Surg. 2002;74:1167–72; discussion 1172.CrossRef Sievers HH, Freund-Kaas C, Eleftheriadis S, et al. Lung protection during total cardiopulmonary bypass by isolated lung perfusion: preliminary results of a novel perfusion strategy. Ann Thorac Surg. 2002;74:1167–72; discussion 1172.CrossRef
39.
Zurück zum Zitat Löckinger A, Schütte H, Walmrath D, Seeger W, Grimminger F. Protection against gas exchange abnormalities by pre-aerosolized PGE1, iloprost and nitroprusside in lung ischemia-reperfusion. Transplantation. 2001;71:185–93.CrossRef Löckinger A, Schütte H, Walmrath D, Seeger W, Grimminger F. Protection against gas exchange abnormalities by pre-aerosolized PGE1, iloprost and nitroprusside in lung ischemia-reperfusion. Transplantation. 2001;71:185–93.CrossRef
40.
Zurück zum Zitat Zamora CA, Baron DA, Heffner JE. Thromboxane contributes to pulmonary hypertension in ischemia-reperfusion lung injury. J Appl Physiol. 1985;1993(74):224–9. Zamora CA, Baron DA, Heffner JE. Thromboxane contributes to pulmonary hypertension in ischemia-reperfusion lung injury. J Appl Physiol. 1985;1993(74):224–9.
41.
Zurück zum Zitat Goudarzi BM, Bonvino S. Critical care issues in lung and heart transplantation. Crit Care Clin. 2003;19:209–31.CrossRef Goudarzi BM, Bonvino S. Critical care issues in lung and heart transplantation. Crit Care Clin. 2003;19:209–31.CrossRef
42.
Zurück zum Zitat Porteous MK, Diamond JM, Christie JD. Primary graft dysfunction: lessons learned about the first 72 h after lung transplantation. Curr Opin Organ Transplant. 2015;20:506–14.CrossRef Porteous MK, Diamond JM, Christie JD. Primary graft dysfunction: lessons learned about the first 72 h after lung transplantation. Curr Opin Organ Transplant. 2015;20:506–14.CrossRef
43.
Zurück zum Zitat Fiser SM, Tribble CG, Long SM, et al. Ischemia-reperfusion injury after lung transplantation increases risk of late bronchiolitis obliterans syndrome. Ann Thorac Surg. 2002;73:1041–7; discussion 1047-8.CrossRef Fiser SM, Tribble CG, Long SM, et al. Ischemia-reperfusion injury after lung transplantation increases risk of late bronchiolitis obliterans syndrome. Ann Thorac Surg. 2002;73:1041–7; discussion 1047-8.CrossRef
44.
Zurück zum Zitat Kreisel D, Krupnick AS, Puri V, et al. Short- and long-term outcomes of 1000 adult lung transplant recipients at a single center. J Thorac Cardiovasc Surg. 2011;141:215–22.CrossRef Kreisel D, Krupnick AS, Puri V, et al. Short- and long-term outcomes of 1000 adult lung transplant recipients at a single center. J Thorac Cardiovasc Surg. 2011;141:215–22.CrossRef
45.
Zurück zum Zitat Stephens RS, Shah AS, Whitman GJ. Lung injury and acute respiratory distress syndrome after cardiac surgery. Ann Thorac Surg. 2013;95:1122–9.CrossRef Stephens RS, Shah AS, Whitman GJ. Lung injury and acute respiratory distress syndrome after cardiac surgery. Ann Thorac Surg. 2013;95:1122–9.CrossRef
46.
Zurück zum Zitat Cornet AD, Kingma SD, Trof RJ, Wisselink W, Groeneveld AB. Hepatosplanchnic ischemia/reperfusion is a major determinant of lung vascular injury after aortic surgery. J Surg Res. 2009;157:48–54.CrossRef Cornet AD, Kingma SD, Trof RJ, Wisselink W, Groeneveld AB. Hepatosplanchnic ischemia/reperfusion is a major determinant of lung vascular injury after aortic surgery. J Surg Res. 2009;157:48–54.CrossRef
47.
Zurück zum Zitat MacCallum NS, Evans TW. Epidemiology of acute lung injury. Curr Opin Crit Care. 2005;11:43–9.CrossRef MacCallum NS, Evans TW. Epidemiology of acute lung injury. Curr Opin Crit Care. 2005;11:43–9.CrossRef
48.
Zurück zum Zitat Li G, Chen S, Lou W, Lu E. Protective effects of ischemic preconditioning on donor lung in canine lung transplantation. Chest. 1998;113:1356–9.CrossRef Li G, Chen S, Lou W, Lu E. Protective effects of ischemic preconditioning on donor lung in canine lung transplantation. Chest. 1998;113:1356–9.CrossRef
49.
Zurück zum Zitat Du ZY, Hicks M, Winlaw D, Spratt P, Macdonald P. Ischemic preconditioning enhances donor lung preservation in the rat. J Heart Lung Transplant. 1996;15:1258–67. Du ZY, Hicks M, Winlaw D, Spratt P, Macdonald P. Ischemic preconditioning enhances donor lung preservation in the rat. J Heart Lung Transplant. 1996;15:1258–67.
50.
Zurück zum Zitat Gasparri RI, Jannis NC, Flameng WJ, Lerut TE, Van Raemdonck DE. Ischemic preconditioning enhances donor lung preservation in the rabbit. Eur J Cardiothorac Surg. 1999;16:639–46.CrossRef Gasparri RI, Jannis NC, Flameng WJ, Lerut TE, Van Raemdonck DE. Ischemic preconditioning enhances donor lung preservation in the rabbit. Eur J Cardiothorac Surg. 1999;16:639–46.CrossRef
51.
Zurück zum Zitat Soncul H, Oz E, Kalaycioglu S. Role of ischemic preconditioning on ischemia-reperfusion injury of the lung. Chest. 1999;115:1672–7.CrossRef Soncul H, Oz E, Kalaycioglu S. Role of ischemic preconditioning on ischemia-reperfusion injury of the lung. Chest. 1999;115:1672–7.CrossRef
52.
Zurück zum Zitat Kandilci HB, Gumusel B, Topaloglu E, et al. Effects of ischemic preconditioning on rat lung: role of nitric oxide. Exp Lung Res. 2006;32:287–303.CrossRef Kandilci HB, Gumusel B, Topaloglu E, et al. Effects of ischemic preconditioning on rat lung: role of nitric oxide. Exp Lung Res. 2006;32:287–303.CrossRef
53.
Zurück zum Zitat Li G, Chen S, Lu E, Hu T. Protective effects of ischemic preconditioning on lung ischemia reperfusion injury: an in-vivo rabbit study. Thorac Cardiovasc Surg. 1999;47:38–41.CrossRef Li G, Chen S, Lu E, Hu T. Protective effects of ischemic preconditioning on lung ischemia reperfusion injury: an in-vivo rabbit study. Thorac Cardiovasc Surg. 1999;47:38–41.CrossRef
54.
Zurück zum Zitat Friedrich I, Spillner J, Lu EX, et al. Ischemic pre-conditioning of 5 minutes but not of 10 minutes improves lung function after warm ischemia in a canine model. J Heart Lung Transplant. 2001;20:985–95.CrossRef Friedrich I, Spillner J, Lu EX, et al. Ischemic pre-conditioning of 5 minutes but not of 10 minutes improves lung function after warm ischemia in a canine model. J Heart Lung Transplant. 2001;20:985–95.CrossRef
55.
Zurück zum Zitat Jun N, Ke J, Gang C, Lin C, Jinsong L, Jianjun W. The protective effect of ischemic preconditioning associated with altered gene expression profiles in rat lung after reperfusion. J Surg Res. 2011;168:281–93.CrossRef Jun N, Ke J, Gang C, Lin C, Jinsong L, Jianjun W. The protective effect of ischemic preconditioning associated with altered gene expression profiles in rat lung after reperfusion. J Surg Res. 2011;168:281–93.CrossRef
56.
Zurück zum Zitat Zhou X, Jiang R, Dong Y, Wang L. Remote ischemic preconditioning attenuates cardiopulmonary bypass-induced lung injury. PLoS ONE. 2017;12: e0189501.CrossRef Zhou X, Jiang R, Dong Y, Wang L. Remote ischemic preconditioning attenuates cardiopulmonary bypass-induced lung injury. PLoS ONE. 2017;12: e0189501.CrossRef
57.
Zurück zum Zitat Akahane HGK, Gomes RZ, Paludo KS, Linhares F, Lopes L. The influence of allopurinol and post-conditioning on lung injuries induced by lower-limb ischemia and reperfusion in Wistar rats. Acta Cir Bras. 2017;32:746–54.CrossRef Akahane HGK, Gomes RZ, Paludo KS, Linhares F, Lopes L. The influence of allopurinol and post-conditioning on lung injuries induced by lower-limb ischemia and reperfusion in Wistar rats. Acta Cir Bras. 2017;32:746–54.CrossRef
58.
Zurück zum Zitat Peralta C, Fernández L, Panés J, et al. Preconditioning protects against systemic disorders associated with hepatic ischemia-reperfusion through blockade of tumor necrosis factor-induced P-selectin up-regulation in the rat. Hepatology. 2001;33:100–13.CrossRef Peralta C, Fernández L, Panés J, et al. Preconditioning protects against systemic disorders associated with hepatic ischemia-reperfusion through blockade of tumor necrosis factor-induced P-selectin up-regulation in the rat. Hepatology. 2001;33:100–13.CrossRef
59.
Zurück zum Zitat Thomaz Neto FJ, Koike MK, AbrahãoMde S, et al. Ischemic preconditioning attenuates remote pulmonary inflammatory infiltration of diabetic rats with an intestinal and hepatic ischemia-reperfusion injury. Acta Cir Bras. 2013;28:174–8.CrossRef Thomaz Neto FJ, Koike MK, AbrahãoMde S, et al. Ischemic preconditioning attenuates remote pulmonary inflammatory infiltration of diabetic rats with an intestinal and hepatic ischemia-reperfusion injury. Acta Cir Bras. 2013;28:174–8.CrossRef
60.
Zurück zum Zitat Meng QT, Cao C, Wu Y, et al. Ischemic post-conditioning attenuates acute lung injury induced by intestinal ischemia-reperfusion in mice: role of Nrf2. Lab Invest. 2016;96:1087–104.CrossRef Meng QT, Cao C, Wu Y, et al. Ischemic post-conditioning attenuates acute lung injury induced by intestinal ischemia-reperfusion in mice: role of Nrf2. Lab Invest. 2016;96:1087–104.CrossRef
61.
Zurück zum Zitat Santos CH, Aydos RD, Nogueira Neto E, et al. Evaluation of pulmonary reperfusion injury in rats undergoing mesenteric ischemia and reperfusion and protective effect of postconditioning on this process. Braz J Cardiovasc Surg. 2015;30:533–7. Santos CH, Aydos RD, Nogueira Neto E, et al. Evaluation of pulmonary reperfusion injury in rats undergoing mesenteric ischemia and reperfusion and protective effect of postconditioning on this process. Braz J Cardiovasc Surg. 2015;30:533–7.
62.
Zurück zum Zitat Harkin DW, Barros D’Sa AA, McCallion K, Hoper M, Campbell FC. Ischemic preconditioning before lower limb ischemia–reperfusion protects against acute lung injury. J Vasc Surg. 2002;35:1264–73.CrossRef Harkin DW, Barros D’Sa AA, McCallion K, Hoper M, Campbell FC. Ischemic preconditioning before lower limb ischemia–reperfusion protects against acute lung injury. J Vasc Surg. 2002;35:1264–73.CrossRef
63.
Zurück zum Zitat Olguner C, Koca U, Kar A, et al. Ischemic preconditioning attenuates the lipid peroxidation and remote lung injury in the rat model of unilateral lower limb ischemia reperfusion. Acta Anaesthesiol Scand. 2006;50:150–5.CrossRef Olguner C, Koca U, Kar A, et al. Ischemic preconditioning attenuates the lipid peroxidation and remote lung injury in the rat model of unilateral lower limb ischemia reperfusion. Acta Anaesthesiol Scand. 2006;50:150–5.CrossRef
64.
Zurück zum Zitat Regel G, Grotz M, Weltner T, Sturm JA, Tscherne H. Pattern of organ failure following severe trauma. World J Surg. 1996;20:422–9.CrossRef Regel G, Grotz M, Weltner T, Sturm JA, Tscherne H. Pattern of organ failure following severe trauma. World J Surg. 1996;20:422–9.CrossRef
65.
Zurück zum Zitat Jarrar D, Chaudry IH, Wang P. Organ dysfunction following hemorrhage and sepsis: mechanisms and therapeutic approaches (review). Int J Mol Med. 1999;4:575–83. Jarrar D, Chaudry IH, Wang P. Organ dysfunction following hemorrhage and sepsis: mechanisms and therapeutic approaches (review). Int J Mol Med. 1999;4:575–83.
66.
Zurück zum Zitat Jan WC, Chen CH, Tsai PS, Huang CJ. Limb ischemic preconditioning mitigates lung injury induced by haemorrhagic shock/resuscitation in rats. Resuscitation. 2011;82:760–6.CrossRef Jan WC, Chen CH, Tsai PS, Huang CJ. Limb ischemic preconditioning mitigates lung injury induced by haemorrhagic shock/resuscitation in rats. Resuscitation. 2011;82:760–6.CrossRef
67.
Zurück zum Zitat Leung CH, Caldarone CA, Wang F, et al. Remote ischemic conditioning prevents lung and liver injury after hemorrhagic shock/resuscitation: potential role of a humoral plasma factor. Ann Surg. 2015;261:1215–25.CrossRef Leung CH, Caldarone CA, Wang F, et al. Remote ischemic conditioning prevents lung and liver injury after hemorrhagic shock/resuscitation: potential role of a humoral plasma factor. Ann Surg. 2015;261:1215–25.CrossRef
68.
Zurück zum Zitat Bergmann A, Schilling T, Hedenstierna G, et al. Pulmonary effects of remote ischemic preconditioning in a porcine model of ventilation-induced lung injury. Respir Physiol Neurobiol. 2019;259:111–8.CrossRef Bergmann A, Schilling T, Hedenstierna G, et al. Pulmonary effects of remote ischemic preconditioning in a porcine model of ventilation-induced lung injury. Respir Physiol Neurobiol. 2019;259:111–8.CrossRef
69.
Zurück zum Zitat Jin X, Wang L, Li L, Zhao X. Protective effect of remote ischemic pre-conditioning on patients undergoing cardiac bypass valve replacement surgery: a randomized controlled trial. Exp Ther Med. 2019;17:2099–106. Jin X, Wang L, Li L, Zhao X. Protective effect of remote ischemic pre-conditioning on patients undergoing cardiac bypass valve replacement surgery: a randomized controlled trial. Exp Ther Med. 2019;17:2099–106.
70.
Zurück zum Zitat Cheung MM, Kharbanda RK, Konstantinov IE, et al. Randomized controlled trial of the effects of remote ischemic preconditioning on children undergoing cardiac surgery: first clinical application in humans. J Am Coll Cardiol. 2006;47:2277–82.CrossRef Cheung MM, Kharbanda RK, Konstantinov IE, et al. Randomized controlled trial of the effects of remote ischemic preconditioning on children undergoing cardiac surgery: first clinical application in humans. J Am Coll Cardiol. 2006;47:2277–82.CrossRef
71.
Zurück zum Zitat Chen S, Li G, Long L. Clinical research of ischemic preconditioning on lung protection. Hunan Yi Ke Da Xue Xue Bao. 1999;24:357–9. Chen S, Li G, Long L. Clinical research of ischemic preconditioning on lung protection. Hunan Yi Ke Da Xue Xue Bao. 1999;24:357–9.
72.
Zurück zum Zitat Lin E, Snell GI, Levvey BJ, et al. Safety, feasibility, and effect of remote ischemic conditioning in patients undergoing lung transplantation. J Heart Lung Transplant. 2014;33:1139–48.CrossRef Lin E, Snell GI, Levvey BJ, et al. Safety, feasibility, and effect of remote ischemic conditioning in patients undergoing lung transplantation. J Heart Lung Transplant. 2014;33:1139–48.CrossRef
73.
Zurück zum Zitat Sayers RD, Thompson MM, Nasim A, Healey P, Taub N, Bell PR. Surgical management of 671 abdominal aortic aneurysms: a 13 year review from a single centre. Eur J Vasc Endovasc Surg. 1997;13:322–7.CrossRef Sayers RD, Thompson MM, Nasim A, Healey P, Taub N, Bell PR. Surgical management of 671 abdominal aortic aneurysms: a 13 year review from a single centre. Eur J Vasc Endovasc Surg. 1997;13:322–7.CrossRef
74.
Zurück zum Zitat Yassin MM, Harkin DW, Barros D’Sa AA, Halliday MI, Rowlands BJ. Lower limb ischemia-reperfusion injury triggers a systemic inflammatory response and multiple organ dysfunction. World J Surg. 2002;26:115–21.CrossRef Yassin MM, Harkin DW, Barros D’Sa AA, Halliday MI, Rowlands BJ. Lower limb ischemia-reperfusion injury triggers a systemic inflammatory response and multiple organ dysfunction. World J Surg. 2002;26:115–21.CrossRef
75.
Zurück zum Zitat Featherstone RL, Chambers DJ, Kelly FJ. Ischemic preconditioning enhances recovery of isolated rat lungs after hypothermic preservation. Ann Thorac Surg. 2000;69:237–42.CrossRef Featherstone RL, Chambers DJ, Kelly FJ. Ischemic preconditioning enhances recovery of isolated rat lungs after hypothermic preservation. Ann Thorac Surg. 2000;69:237–42.CrossRef
76.
Zurück zum Zitat Kharbanda RK, Li J, Konstantinov IE, et al. Remote ischaemic preconditioning protects against cardiopulmonary bypass-induced tissue injury: a preclinical study. Heart. 2006;92:1506–11.CrossRef Kharbanda RK, Li J, Konstantinov IE, et al. Remote ischaemic preconditioning protects against cardiopulmonary bypass-induced tissue injury: a preclinical study. Heart. 2006;92:1506–11.CrossRef
77.
Zurück zum Zitat Xia Z, Herijgers P, Nishida T, Ozaki S, Wouters P, Flameng W. Remote preconditioning lessens the deterioration of pulmonary function after repeated coronary artery occlusion and reperfusion in sheep. Can J Anaesth. 2003;50:481–8.CrossRef Xia Z, Herijgers P, Nishida T, Ozaki S, Wouters P, Flameng W. Remote preconditioning lessens the deterioration of pulmonary function after repeated coronary artery occlusion and reperfusion in sheep. Can J Anaesth. 2003;50:481–8.CrossRef
78.
Zurück zum Zitat Min JJ, Bae JY, Kim TK, et al. Pulmonary protective effects of remote ischaemic preconditioning with postconditioning in patients undergoing cardiac surgery involving cardiopulmonary bypass: a substudy of the remote ischaemic preconditioning with postconditioning outcome trial. Heart Lung Circ. 2016;25:484–92.CrossRef Min JJ, Bae JY, Kim TK, et al. Pulmonary protective effects of remote ischaemic preconditioning with postconditioning in patients undergoing cardiac surgery involving cardiopulmonary bypass: a substudy of the remote ischaemic preconditioning with postconditioning outcome trial. Heart Lung Circ. 2016;25:484–92.CrossRef
79.
Zurück zum Zitat Hong DM, Jeon Y, Lee CS, et al. Effects of remote ischemic preconditioning with postconditioning in patients undergoing off-pump coronary artery bypass surgery–randomized controlled trial. Circ J. 2012;76:884–90.CrossRef Hong DM, Jeon Y, Lee CS, et al. Effects of remote ischemic preconditioning with postconditioning in patients undergoing off-pump coronary artery bypass surgery–randomized controlled trial. Circ J. 2012;76:884–90.CrossRef
80.
Zurück zum Zitat Kim JC, Shim JK, Lee S, Yoo YC, Yang SY, Kwak YL. Effect of combined remote ischemic preconditioning and postconditioning on pulmonary function in valvular heart surgery. Chest. 2012;142:467–75.CrossRef Kim JC, Shim JK, Lee S, Yoo YC, Yang SY, Kwak YL. Effect of combined remote ischemic preconditioning and postconditioning on pulmonary function in valvular heart surgery. Chest. 2012;142:467–75.CrossRef
81.
Zurück zum Zitat Rahman IA, Mascaro JG, Steeds RP, et al. Remote ischemic preconditioning in human coronary artery bypass surgery: from promise to disappointment? Circulation. 2010;122:S53–9.CrossRef Rahman IA, Mascaro JG, Steeds RP, et al. Remote ischemic preconditioning in human coronary artery bypass surgery: from promise to disappointment? Circulation. 2010;122:S53–9.CrossRef
82.
Zurück zum Zitat Lee JH, Park YH, Byon HJ, Kim HS, Kim CS, Kim JT. Effect of remote ischaemic preconditioning on ischaemic-reperfusion injury in pulmonary hypertensive infants receiving ventricular septal defect repair. Br J Anaesth. 2012;108:223–8.CrossRef Lee JH, Park YH, Byon HJ, Kim HS, Kim CS, Kim JT. Effect of remote ischaemic preconditioning on ischaemic-reperfusion injury in pulmonary hypertensive infants receiving ventricular septal defect repair. Br J Anaesth. 2012;108:223–8.CrossRef
Metadaten
Titel
The effects of ischaemic conditioning on lung ischaemia–reperfusion injury
verfasst von
Dimitrios Vlastos
Mohamed Zeinah
George Ninkovic-Hall
Stefanos Vlachos
Agni Salem
Athanasios Asonitis
Hemangi Chavan
Lazaros Kalampalikis
Abdullah Al Shammari
José María Alvarez Gallesio
Aina Pons
Ioanna Andreadou
Ignatios Ikonomidis
Publikationsdatum
01.12.2022
Verlag
BioMed Central
Erschienen in
Respiratory Research / Ausgabe 1/2022
Elektronische ISSN: 1465-993X
DOI
https://doi.org/10.1186/s12931-022-02288-z

Weitere Artikel der Ausgabe 1/2022

Respiratory Research 1/2022 Zur Ausgabe

Leitlinien kompakt für die Innere Medizin

Mit medbee Pocketcards sicher entscheiden.

Seit 2022 gehört die medbee GmbH zum Springer Medizin Verlag

Notfall-TEP der Hüfte ist auch bei 90-Jährigen machbar

26.04.2024 Hüft-TEP Nachrichten

Ob bei einer Notfalloperation nach Schenkelhalsfraktur eine Hemiarthroplastik oder eine totale Endoprothese (TEP) eingebaut wird, sollte nicht allein vom Alter der Patientinnen und Patienten abhängen. Auch über 90-Jährige können von der TEP profitieren.

Niedriger diastolischer Blutdruck erhöht Risiko für schwere kardiovaskuläre Komplikationen

25.04.2024 Hypotonie Nachrichten

Wenn unter einer medikamentösen Hochdrucktherapie der diastolische Blutdruck in den Keller geht, steigt das Risiko für schwere kardiovaskuläre Ereignisse: Darauf deutet eine Sekundäranalyse der SPRINT-Studie hin.

Bei schweren Reaktionen auf Insektenstiche empfiehlt sich eine spezifische Immuntherapie

Insektenstiche sind bei Erwachsenen die häufigsten Auslöser einer Anaphylaxie. Einen wirksamen Schutz vor schweren anaphylaktischen Reaktionen bietet die allergenspezifische Immuntherapie. Jedoch kommt sie noch viel zu selten zum Einsatz.

Therapiestart mit Blutdrucksenkern erhöht Frakturrisiko

25.04.2024 Hypertonie Nachrichten

Beginnen ältere Männer im Pflegeheim eine Antihypertensiva-Therapie, dann ist die Frakturrate in den folgenden 30 Tagen mehr als verdoppelt. Besonders häufig stürzen Demenzkranke und Männer, die erstmals Blutdrucksenker nehmen. Dafür spricht eine Analyse unter US-Veteranen.

Update Innere Medizin

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.