Sign in to use this feature.

Years

Between: -

Subjects

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Journals

Article Types

Countries / Regions

Search Results (78)

Search Parameters:
Keywords = lung ischemia-reperfusion injury

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
27 pages, 1490 KB  
Review
Damage-Associated Molecular Patterns in Perioperative Anesthesia Care: A Clinical Perspective
by Wiriya Maisat and Koichi Yuki
Anesth. Res. 2026, 3(1), 1; https://doi.org/10.3390/anesthres3010001 (registering DOI) - 20 Dec 2025
Abstract
Damage-associated molecular patterns (DAMPs) are endogenous molecules released during cellular stress or injury that trigger sterile inflammation. In perioperative settings, common triggers include surgical trauma, ischemia–reperfusion injury, cardiopulmonary bypass, blood transfusion, and mechanical ventilation. When released extracellularly, DAMPs activate innate immune receptors such [...] Read more.
Damage-associated molecular patterns (DAMPs) are endogenous molecules released during cellular stress or injury that trigger sterile inflammation. In perioperative settings, common triggers include surgical trauma, ischemia–reperfusion injury, cardiopulmonary bypass, blood transfusion, and mechanical ventilation. When released extracellularly, DAMPs activate innate immune receptors such as Toll-like receptors (TLRs) and the receptor for advanced glycation end products (RAGE), initiating signaling cascades that amplify inflammation, disrupt endothelial integrity, and promote coagulation and metabolic imbalance. This sterile inflammatory response may extend local tissue injury into systemic organ dysfunction, manifesting clinically as acute lung injury, acute kidney injury, myocardial dysfunction, disseminated intravascular coagulation, and perioperative neurocognitive disorders. Recognizing the central role of DAMPs reframes these complications as predictable consequences of endogenous danger signaling rather than solely as results of infection or hemodynamic instability. This understanding supports the use of established strategies such as protective ventilation and restrictive transfusion to minimize DAMP release. Emerging evidence also suggests that anesthetic agents may influence DAMP-mediated inflammation: propofol and dexmedetomidine appear to exert anti-inflammatory effects, whereas volatile anesthetics show variable results. Although clinical data remain limited, anesthetic choice and perioperative management may significantly affect systemic inflammatory burden and recovery. Future research validating DAMPs as biomarkers and therapeutic targets may inform precision anesthetic strategies aimed at modulating sterile inflammation, ultimately enhancing perioperative outcome. Full article
Show Figures

Graphical abstract

14 pages, 3259 KB  
Article
Therapeutic Effects of Argon Inhalation on Lung Ischemia–Reperfusion Injury in CLAWN Miniature Swine
by Takehiro Iwanaga, Masayoshi Okumi, Yuichi Ariyoshi, Kazuhiro Takeuchi, Akira Kondo, Mitsuhiro Sekijima, Yurika Ichinari, Akira Shimizu and Hisashi Sahara
J. Clin. Med. 2025, 14(24), 8821; https://doi.org/10.3390/jcm14248821 - 12 Dec 2025
Viewed by 229
Abstract
Background: Noble gases, such as argon, have been observed to exhibit cytoprotective effects. The non-anesthetic properties, abundance, and cost-effectiveness of argon suggest its clinical potential. While its efficacy in mitigating ischemia–reperfusion injury has been demonstrated in cellular and small animal models, data [...] Read more.
Background: Noble gases, such as argon, have been observed to exhibit cytoprotective effects. The non-anesthetic properties, abundance, and cost-effectiveness of argon suggest its clinical potential. While its efficacy in mitigating ischemia–reperfusion injury has been demonstrated in cellular and small animal models, data on its effects in large animals remain limited. This study evaluated the effects of argon inhalation on pulmonary ischemia–reperfusion injury in miniature swine with potential applications in transplantation. Methods: The left bronchial and pulmonary artery and veins were clamped for 90 min, and then the clamps were released to induce lung ischemia–reperfusion injury in 10 CLAWN miniature swine. The argon group (n = 5) inhaled a mixture of 30% oxygen and 70% argon for 360 min, whereas the control group (n = 5) inhaled a mixture of 30% oxygen and 70% nitrogen for an equivalent duration. Lung function was evaluated using chest X-ray, lung biopsies, and blood gas analysis. Results: The PaO2/FiO2 ratio significantly decreased in the control group 2 h post-reperfusion (568 ± 12 to 272 ± 39 mmHg), but was better preserved in the argon group (562 ± 17 to 430 ± 48 mmHg). Blood gas from the left pulmonary vein showed a superior PvO2/FiO2 ratio in the argon group (331 ± 40 vs. 186 ± 17 mmHg at 2 h; 519 ± 19 vs. 292 ± 33 mmHg at 2 days). Chest X-ray revealed reduced infiltration in the left lung. The lung biopsy histological scores improved in the argon group at 2 h and 2 days. Serum superoxide dismutase analysis and tissue TUNEL assays suggested that antioxidant and anti-apoptotic mechanisms, respectively, were involved. Conclusions: Perioperative argon inhalation attenuates ischemia–reperfusion injury in swine lungs, likely via anti-apoptotic and antioxidant effects. Full article
Show Figures

Figure 1

20 pages, 3437 KB  
Article
Early and Mid-Term Results of Solid Organ Transplantation After Circulatory Death: A 4-Year Single Centre Experience
by Antonella Galeone, Marilena Casartelli Liviero, Alex Borin, Rostand Emmanuel Nguefouet Momo, Leonardo Gottin, Francesco Onorati, Irene Maffei, Marco Schiavon, Paolo Persona, Tiziano Menon, Luigino Boschiero, Alessandro Antonelli, Giovanni Battista Luciani and Amedeo Carraro
Medicina 2025, 61(12), 2126; https://doi.org/10.3390/medicina61122126 - 28 Nov 2025
Viewed by 298
Abstract
Background and Objectives: The use of controlled donation after circulatory death (cDCD) donors has significantly increased during the past decades and successfully expanded the donors’ pool. However, warm ischemia may have detrimental effects on graft function. Italian Law requires a no-touch period [...] Read more.
Background and Objectives: The use of controlled donation after circulatory death (cDCD) donors has significantly increased during the past decades and successfully expanded the donors’ pool. However, warm ischemia may have detrimental effects on graft function. Italian Law requires a no-touch period of at least 20 min, which is much longer compared to the 5 min accepted in most European countries. Materials and Methods This is an Italian single-centre retrospective review of all cDCD procedures performed from April 2021 to June 2025. Patients with severe brain injury undergoing withdrawal of life-sustaining therapy (WLST) were considered for cDCD. After cardiac arrest and a no-touch period of 20 min, organ reperfusion was performed using abdominal or thoraco-abdominal normothermic regional perfusion (NRP) through femoral vessels cannulation. The primary endpoint was 30-day graft survival; secondary endpoints included: incidence of primary non-function (PNF) and non-anastomotic biliary stricture (NAS) in liver transplantation, PNF and delayed graft function (DGF) in kidney transplantation, primary graft dysfunction (PGD) in heart and lung transplantation, and recipient’s survival. Results: A total of 52 patients, 33 (63%) males, median age 74 (65–79) years, underwent WLST during the study period and were included in the cDCD program. Median functional warm ischemic time (WIT), total WIT, asystolic phase, and NRP duration were 37 (34–40), 40 (37–42), 24 (23–26), and 192 (166–212) min, respectively. A total of 123 organs (46 livers, 61 kidneys, 8 hearts, and 8 lungs) were considered suitable for transplantation, procured, and successfully transplanted in 115 recipients. We report the early and mid-term outcomes of 84 recipients, including 41 liver recipients, 32 kidney recipients, and 8 heart recipients transplanted at the Azienda Ospedaliera Universitaria Integrata of Verona, and 3 lung recipients transplanted at the Azienda Ospedale Università of Padova. The 30-day graft survival was 95% in liver recipients, 97% in kidney recipients, and 100% in heart and lung recipients. PNF was observed in two liver recipients, and PGD in two lung recipients. DGF was recorded in 3 (9%) kidney recipients. Six recipients died during the follow-up, and the mean survival time was 3.9 ± 0.1 years. Conclusions: Solid organ transplantation using cDCD donors is feasible and provides excellent early and mid-term results despite longer donor asystolic times. Larger data and longer follow-up are necessary to confirm these promising results. Full article
Show Figures

Figure 1

20 pages, 3348 KB  
Article
Efficiency of Calcium Fructoborate-Loaded Novel Natural Niosomes Compared to Traditional Liposomes and Niosomes in Rat Ischemia–Reperfusion Injury Model
by Kubra Vardar, Nilufer Kara, Nuri Murat Ozayman, Kubilay Gocucu, Sirin Funda Eren, Metin Plevneli, Ismail Aslan and Mehmet Necmettin Atsu
Pharmaceutics 2025, 17(11), 1434; https://doi.org/10.3390/pharmaceutics17111434 - 6 Nov 2025
Viewed by 639
Abstract
Background/Objectives: Liposomes and niosomes are established drug delivery systems, some of which have received FDA approval and demonstrated therapeutic efficacy. This study investigates a novel niosome formulation, utilizing two natural food-derived components, as a cost-effective alternative to traditional nanocarriers. The active pharmaceutical ingredient, [...] Read more.
Background/Objectives: Liposomes and niosomes are established drug delivery systems, some of which have received FDA approval and demonstrated therapeutic efficacy. This study investigates a novel niosome formulation, utilizing two natural food-derived components, as a cost-effective alternative to traditional nanocarriers. The active pharmaceutical ingredient, calcium fructoborate (CF), possesses notable anti-inflammatory properties. The study aims to evaluate the efficacy of this novel natural niosome (NN) system, in comparison to existing nanocarrier formulations, in an ischemia–reperfusion (I/R) pain model. Methods: An acute ischemia/reperfusion injury model was employed to induce pain in 36 rats. The efficacy of the following treatments was assessed: standard CF, liposomal CF, niosomal CF, and natural niosomal CF. Efficacy was determined by quantifying the treatments’ ability to mitigate inflammation and oxidative stress in the kidneys, lungs, heart, and liver, and by evaluating potential organ damage through histopathological analysis. Results: The NN treatment significantly reduced malondialdehyde (MDA) and tumor necrosis factor-alpha (TNF-α) levels in the kidneys and liver compared to the other treatments (p < 0.05). In the kidney, NN treatment also significantly decreased creatinine levels relative to the other treatments (p < 0.01). The histopathological analysis of kidney tissue revealed that NN treatment attenuated tubular dilation, interstitial inflammation, and epithelial thinning. In the heart, liposomal treatment significantly increased MDA levels (p < 0.05) and decreased sialic acid levels (p < 0.05); however, no significant differences were observed in troponin levels (p > 0.05). In the lung, no significant differences in MDA, lactate, TNF-α, or sialic acid levels were detected among the treatment groups (p > 0.05). Conclusions: The natural niosome drug delivery system demonstrates potential as a therapeutic intervention for protecting and improving kidney and liver health. While liposomal treatment exhibited some adverse effects, it effectively suppressed inflammation. This study provides a foundation for future research and positions the NN drug delivery system as a promising, cost-effective alternative for inflammation-associated pathologies. Full article
(This article belongs to the Section Drug Delivery and Controlled Release)
Show Figures

Figure 1

16 pages, 5554 KB  
Article
Acute Kidney Injury Induces Lung Damage via Mitochondrial DAMPs by Activating TREM-1 and cGAS-STING Pathways
by Zhi Tian, Runze Ni, Nadezhda N. Zheleznova, Diane Allen-Gipson, Lei Wang, Vijay Subramanian, Kiran Dhanireddy, Sarah Y. Yuan, Nohely Hernandez Soto, Jose D. Herazo-Maya, Kristof Williams, Isabella Lozonschi, Andrew Bedard, Gabrielle Morrison and Ruisheng Liu
Cells 2025, 14(21), 1716; https://doi.org/10.3390/cells14211716 - 31 Oct 2025
Viewed by 897
Abstract
Acute kidney injury (AKI) is a leading cause of distant organ dysfunction among critically ill patients. Mitochondrial dysfunction is considered a key factor driving the damage after renal ischemia–reperfusion (IR) injury. Damaged mitochondria release mitochondrial damage-associated molecular patterns (mtDAMPs) into the cytosol, which [...] Read more.
Acute kidney injury (AKI) is a leading cause of distant organ dysfunction among critically ill patients. Mitochondrial dysfunction is considered a key factor driving the damage after renal ischemia–reperfusion (IR) injury. Damaged mitochondria release mitochondrial damage-associated molecular patterns (mtDAMPs) into the cytosol, which initiate a systemic inflammatory response. To better understand the underlying mechanism, mice were challenged with 30 min of bilateral renal ischemia followed by 24 h of reperfusion. The cytokine profiling in mouse lung tissues revealed that TREM-1 was significantly increased. Western Blot (WB) analysis demonstrated that the cGAS and STING pathway was increased in AKI mice. Transmission electron microscopy (TEM) images indicated that the mtDAMPs were released from damaged kidney mitochondria. Injection of mtDAMPs into mice induced an inflammatory response in the lungs similar to that induced by AKI. Mouse macrophages and lung epithelial cells were utilized to verify if inhibition of the TREM-1 and cGAS-STING pathways reduces mtDAMP-induced lung injury. Electric Cell-substrate Impedance Sensing (ECIS) results demonstrated that inhibiting the TREM-1 and cGAS-STING pathways significantly increased cell proliferation and migration while reducing mtDAMP-induced cytotoxicity. In conclusion, our findings suggest that targeting TREM-1 and cGAS-STING has the potential to attenuate acute lung injury in IR-AKI. Full article
(This article belongs to the Section Tissues and Organs)
Show Figures

Graphical abstract

34 pages, 976 KB  
Review
Lung Ischemia–Reperfusion Injury in Lung Transplant Surgery: Where Do We Stand?
by Lawek Berzenji, Jeroen M. H. Hendriks, Stijn E. Verleden, Suresh Krishan Yogeswaran, Wen Wen, Patrick Lauwers, Geert Verleden, Rudi De Paep, Pieter Mertens, Inez Rodrigus, Dirk Adriaensen and Paul Van Schil
Antioxidants 2025, 14(11), 1295; https://doi.org/10.3390/antiox14111295 - 28 Oct 2025
Viewed by 1839
Abstract
Lung ischemia–reperfusion injury (LIRI) remains a major contributor to perioperative morbidity and mortality in thoracic surgery, especially for lung transplantations, where it is one of the principal drivers of primary graft dysfunction (PGD). Although substantial advances have been made in surgical technique, donor [...] Read more.
Lung ischemia–reperfusion injury (LIRI) remains a major contributor to perioperative morbidity and mortality in thoracic surgery, especially for lung transplantations, where it is one of the principal drivers of primary graft dysfunction (PGD). Although substantial advances have been made in surgical technique, donor management, and perioperative care, LIRI continues to pose a significant clinical challenge. Mechanistically, LIRI reflects a combined pathology of oxidative stress, endothelial and glycocalyx disruption, innate immune activation, mitochondrial dysfunction, and regulated cell death, resulting in loss of alveolar–capillary barrier integrity and gas exchange failure. Current management is phase-specific and multimodal, spanning donor care and preservation, controlled reperfusion and lung-protective ventilation, and pharmacological treatments. Treatment candidates that target oxidative stress and inflammatory cascades (e.g., antioxidants, complement and adenosine pathways, mesenchymal stromal cell products, and dipeptidyl-peptidase-4 inhibition) show promise, yet translation into a clinical scenario remains difficult. Increasing evidence supports endothelial-preserving and mitochondria-sparing strategies, rigorous perioperative bundles, and biomarker-guided trials to move from pathophysiology to practice. Ultimately, addressing LIRI requires an integrated, multidisciplinary approach that spans surgical, anesthetic, and pharmacologic domains, with the goal of improving both early outcomes and long-term graft survival in lung transplant patients. Full article
Show Figures

Figure 1

17 pages, 2190 KB  
Article
Lidocaine Attenuates miRNA Dysregulation and Kinase Signaling Activation in a Porcine Model of Lung Ischemia/Reperfusion Injury
by Alberto Alonso, Sergio D. Paredes, Agustín Turrero, Lisa Rancan, Ignacio Garutti, Carlos Simón and Elena Vara
Int. J. Mol. Sci. 2025, 26(21), 10385; https://doi.org/10.3390/ijms262110385 - 25 Oct 2025
Viewed by 368
Abstract
Ischemia/reperfusion (I/R) injury is a major complication in lung transplantation. Recent evidence suggests that mitogen-activated protein kinases (MAPKs) such as p-38 mitogen-activated protein kinase (p-38 MAPK) and extracellular signal-regulated kinase (ERK), along with functionally related kinases like phosphoinositide 3-kinase (PI3K) and protein kinase [...] Read more.
Ischemia/reperfusion (I/R) injury is a major complication in lung transplantation. Recent evidence suggests that mitogen-activated protein kinases (MAPKs) such as p-38 mitogen-activated protein kinase (p-38 MAPK) and extracellular signal-regulated kinase (ERK), along with functionally related kinases like phosphoinositide 3-kinase (PI3K) and protein kinase B (AKT), contribute to I/R pathophysiology by mediating inflammatory and stress-response signaling. MicroRNAs (miRNAs) also play a regulatory role in these processes. Lidocaine has demonstrated anti-inflammatory activity in several tissues; however, its ability to modulate miRNA expression and kinase activation in the lung is not yet fully understood. This study investigated the involvement of these signaling molecules in lung I/R injury and evaluated the modulatory effect of intravenous lidocaine in a porcine lung auto-transplantation model. Eighteen large white pigs were assigned to sham-operated (n = 6), control (lung auto-transplantation, n = 6), or lidocaine-treated (n = 6) groups. Lidocaine was administered as a 1.5 mg/kg bolus followed by a continuous infusion (1.5 mg·kg−1·h−1). Lung biopsies were collected before ischemia, before reperfusion, and at 30- and 60-min post-reperfusion to assess total and phosphorylated levels of p-38 MAPK, ERK, PI3K, and AKT (Thr308, Ser473), along with miR-126, miR-142-5p, miR-152, and miR-155 expression. I/R increased p-38 MAPK and AKT, and enhanced phosphorylation of all four kinases. miRNA levels were also upregulated. Lidocaine partially or completely attenuated these changes. These findings support a role for these molecular pathways in lung I/R injury and suggest that lidocaine may offer protective effects through their modulation. Full article
(This article belongs to the Special Issue New Molecular Insights into Ischemia/Reperfusion: 2nd Edition)
Show Figures

Graphical abstract

49 pages, 1461 KB  
Review
Kidneys on the Frontline: Nephrologists Tackling the Wilds of Acute Kidney Injury in Trauma Patients—From Pathophysiology to Early Biomarkers
by Merita Rroji, Marsida Kasa, Nereida Spahia, Saimir Kuci, Alfred Ibrahimi and Hektor Sula
Diagnostics 2025, 15(19), 2438; https://doi.org/10.3390/diagnostics15192438 - 25 Sep 2025
Cited by 1 | Viewed by 3979
Abstract
Acute kidney injury (AKI) is a frequent and severe complication in trauma patients, affecting up to 28% of intensive care unit (ICU) admissions and contributing significantly to morbidity, mortality, and long-term renal impairment. Trauma-related AKI (TRAKI) arises from diverse mechanisms, including hemorrhagic shock, [...] Read more.
Acute kidney injury (AKI) is a frequent and severe complication in trauma patients, affecting up to 28% of intensive care unit (ICU) admissions and contributing significantly to morbidity, mortality, and long-term renal impairment. Trauma-related AKI (TRAKI) arises from diverse mechanisms, including hemorrhagic shock, ischemia–reperfusion injury, systemic inflammation, rhabdomyolysis, nephrotoxicity, and complex organ crosstalk involving the brain, lungs, and abdomen. Pathophysiologically, TRAKI involves early disruption of the glomerular filtration barrier, tubular epithelial injury, and renal microvascular dysfunction. Inflammatory cascades, oxidative stress, immune thrombosis, and maladaptive repair mechanisms mediate these injuries. Trauma-related rhabdomyolysis and exposure to contrast agents or nephrotoxic drugs further exacerbate renal stress, particularly in patients with pre-existing comorbidities. Traditional markers such as serum creatinine (sCr) are late indicators of kidney damage and lack specificity. Emerging structural and stress response biomarkers—such as neutrophil gelatinase-associated lipocalin (NGAL), kidney injury molecule 1 (KIM-1), liver-type fatty acid-binding protein (L-FABP), interleukin-18 (IL-18), C-C motif chemokine ligand 14 (CCL14), Dickkopf-3 (DKK3), and the U.S. Food and Drug Administration (FDA)-approved tissue inhibitor of metalloproteinases-2 × insulin-like growth factor-binding protein 7 (TIMP-2 × IGFBP-7)—allow earlier detection of subclinical AKI and better predict progression and the need for renal replacement therapy. Together, functional indices like urinary sodium and fractional potassium excretion reflect early microcirculatory stress and add clinical value. In parallel, risk stratification tools, including the Renal Angina Index (RAI), the McMahon score, and the Haines model, enable the early identification of high-risk patients and help tailor nephroprotective strategies. Together, these biomarkers and risk models shift from passive AKI recognition to proactive, personalized management. A new paradigm that integrates biomarker-guided diagnostics and dynamic clinical scoring into trauma care promises to reduce AKI burden and improve renal outcomes in this critically ill population. Full article
(This article belongs to the Special Issue Advances in Nephrology)
Show Figures

Graphical abstract

19 pages, 3632 KB  
Article
Levosimendan Pretreatment Attenuates Mesenteric Artery Ischemia/Reperfusion Injury and Multi-Organ Damage in Rats
by Zoran Matković, Milica Gajić Bojić, Uglješa Maličević, Aleksandra Krivokuća, Nebojša Mandić-Kovačević, Snežana Uletilović, Ljiljana Amidžić, Sanja Jovičić, Maja Barudžija, Miloš P. Stojiljković, Radoslav Gajanin, Sergej Bolevich and Ranko Škrbić
Int. J. Mol. Sci. 2025, 26(18), 9131; https://doi.org/10.3390/ijms26189131 - 18 Sep 2025
Viewed by 758
Abstract
Acute mesenteric ischemia (AMI) is a life-threatening condition characterised by oxidative stress, inflammation, apoptosis, and necrosis of intestinal epithelial cells. Different drugs with vasoactive, antioxidant, and anti-inflammatory properties have been used to treat AMI. Levosimendan is a drug with proven anti-ischemic effects used [...] Read more.
Acute mesenteric ischemia (AMI) is a life-threatening condition characterised by oxidative stress, inflammation, apoptosis, and necrosis of intestinal epithelial cells. Different drugs with vasoactive, antioxidant, and anti-inflammatory properties have been used to treat AMI. Levosimendan is a drug with proven anti-ischemic effects used in the management of acute congestive heart failure. This study evaluated the protective effects of levosimendan pretreatment on intestinal, as well as lung, heart, and kidney tissue in a rat model of mesenteric artery ischemia/reperfusion (I/R) injury. Male Wistar rats (N = 24) were divided into four groups: control, I/R, levosimendan (LS) 1 mg/kg i.p, and LS + I/R (1 mg/kg i.p. 30 min before injury). I/R by itself caused elevation of oxidative markers (thyobarbituric acid reactive species (TBARS), hydrogen peroxide (H2O2), super oxide anjon radical (O2), and nitrogen dioxide (NO2)), induced inflammation (macrophage infiltration and Interleukin-6 (IL-6) production), and apoptosis (nuclear factor kappa light-chain enhancer of activated B cells (NF-κB), cleaved caspase-3 (CC3), and terminal deoxy-nucleotidyl transferase (TdT)-mediated dUTP nick end labelling (TUNEL)). Levosimendan pretreatment significantly reduced oxidative stress markers and enhanced antioxidant defences (catalase (CAT), reduced glutathione (GSH), and superoxide dismutase (SOD)). Histological analysis revealed reduced mucosal damage and preserved goblet cells in intestinal tissue. Similar protective effects of levosimendan were observed in other organs such as lung, heart, and kidney. Immunohistochemistry showed reduced epithelial apoptosis and upregulation of antioxidant and anti-inflammatory proteins. These findings highlight levosimendan’s ability to protect mesenteric I/R tissue injury and multi-organ damage by suppressing oxidative stress, inflammation, and apoptosis, emphasising its therapeutic potential in clinical settings. Full article
(This article belongs to the Section Molecular Pathology, Diagnostics, and Therapeutics)
Show Figures

Figure 1

20 pages, 1142 KB  
Review
The Therapeutic Potential of Garlic-Derived Organic Polysulfides for Ischemia-Reperfusion Injury
by Chunlei Wang, Ning Han, Caiyun Mao, Jiaxu Chen, Nana Cheng, Jieyou Zhao, Yunjia Song and Xutao Sun
Int. J. Mol. Sci. 2025, 26(17), 8257; https://doi.org/10.3390/ijms26178257 - 26 Aug 2025
Cited by 2 | Viewed by 1650
Abstract
Ischemia-reperfusion (I/R) injury refers to the exacerbation of tissue or organ damage upon the restoration of blood flow after an ischemic event. Despite its widespread clinical occurrence, therapeutic interventions for I/R injury remain limited in efficacy, presenting a significant challenge in modern medicine. [...] Read more.
Ischemia-reperfusion (I/R) injury refers to the exacerbation of tissue or organ damage upon the restoration of blood flow after an ischemic event. Despite its widespread clinical occurrence, therapeutic interventions for I/R injury remain limited in efficacy, presenting a significant challenge in modern medicine. Garlic, traditionally consumed as a food, has gained considerable attention for its medicinal properties. Numerous animal studies have shown that garlic-derived organic polysulfides significantly improve nerve function scores post-I/R, reduce infarct size, mitigate inflammatory responses, and inhibit cellular apoptosis. Thus, understanding the role of garlic-derived organic polysulfides in I/R injury may unveil novel therapeutic targets. This review explores the protective effects and mechanisms of garlic-derived organic polysulfides on I/R injury in various organs, including the brain, spinal cord, myocardium, lungs, liver, kidneys, and testes, highlighting their potential in advancing treatment strategies for affected patients. Full article
(This article belongs to the Section Bioactives and Nutraceuticals)
Show Figures

Graphical abstract

18 pages, 6039 KB  
Article
Neutrophil Gelatinase-Associated Lipocalin: A Shared Early Biomarker of Remote Organ Dysfunction in Blast-Induced Extremity Trauma
by Cassie J. Rowe, Uloma Nwaolu, Philip J. Spreadborough and Thomas A. Davis
Int. J. Mol. Sci. 2025, 26(16), 7794; https://doi.org/10.3390/ijms26167794 - 12 Aug 2025
Viewed by 816
Abstract
Polytrauma is a critical global health concern characterized by immune dysregulation and a high risk of multiple organ dysfunction syndrome (MODS). Early molecular mechanisms linking trauma severity to organ injury are poorly understood. We used two rat blast-polytrauma models: a tourniquet-induced ischemia/reperfusion injury [...] Read more.
Polytrauma is a critical global health concern characterized by immune dysregulation and a high risk of multiple organ dysfunction syndrome (MODS). Early molecular mechanisms linking trauma severity to organ injury are poorly understood. We used two rat blast-polytrauma models: a tourniquet-induced ischemia/reperfusion injury (tIRI) model and a non-ischemia/reperfusion injury (non-IRI) model. Naïve animals served as controls. RT-qPCR of 120 inflammatory genes in the lung, kidney, and liver, combined with STRING protein–protein interaction analysis, revealed distinct yet overlapping inflammatory gene signatures across all the organs. A core set of genes (Il6, Lbp, Nos2, and Lcn2) was consistently upregulated, indicating shared inflammatory pathways. Transcriptomic responses were most pronounced in the tIRI group, with greater magnitude and altered temporal dynamics, uniquely amplifying pro-inflammatory cytokines, immune cell activators, chemokines, and tissue damage markers. Lipocalin-2 (Lcn2/NGAL) emerged as a shared hub gene across all the organs within 24 h post-injury. Its expression significantly correlated with MODS activity and adverse outcomes, independent of the injury model. At 168 h, Lcn2 expression correlated with increased liver damage and NGAL levels correlated with tissue trauma severity. These findings elucidate distinct pro-inflammatory mediators and networks underlying secondary organ dysfunction, highlighting NGAL as a potential universal biomarker of trauma-induced inflammation and MODS activity, suggesting it as a therapeutic target. Full article
(This article belongs to the Section Molecular Biology)
Show Figures

Figure 1

15 pages, 3987 KB  
Article
Cardioprotective Effects of Bosentan in Rats Subjected to Lung Ischemia–Reperfusion Injury
by Şevki Mustafa Demiröz, Ayşegül Küçük, Esra Tekin, Sibel Söylemez, Hanife Yılmaz, Şaban Cem Sezen, Muharrem Atlı, Hüseyin Demirtaş, Abdullah Özer, Yusuf Ünal and Mustafa Arslan
Medicina 2025, 61(7), 1298; https://doi.org/10.3390/medicina61071298 - 18 Jul 2025
Viewed by 789
Abstract
Objective: This study aimed to investigate the cardioprotective effects of bosentan, an endothelin receptor antagonist, in a rat model of lung ischemia–reperfusion (I/R) injury, with a focus on myocardial tissue involvement. Methods: Twenty-four male Wistar rats were randomly assigned to four [...] Read more.
Objective: This study aimed to investigate the cardioprotective effects of bosentan, an endothelin receptor antagonist, in a rat model of lung ischemia–reperfusion (I/R) injury, with a focus on myocardial tissue involvement. Methods: Twenty-four male Wistar rats were randomly assigned to four groups: sham, bosentan, I/R, and I/R + bosentan. Lung I/R injury was induced by hilar clamping for 45 min, followed by 60 min of reperfusion. Bosentan (30 mg/kg) was administered intraperitoneally 30 min prior to the procedure. Myocardial tissue was evaluated histopathologically for structural disorganization, inflammation, fibrosis, and edema. TGF-β1 protein levels in myocardial tissue were compared across the groups using β-actin as the loading control. ELISA was used to quantify ET-1, NF-κB, and p53 levels, while spectrophotometric analysis was employed to assess MDA levels and the activities of SOD and CAT enzymes in heart tissue. Results: The I/R group exhibited significant myocardial disorganization, inflammation, and interstitial edema compared to the sham and bosentan groups. Bosentan treatment markedly ameliorated these histopathological alterations. Additionally, the I/R group showed elevated levels of ET-1, NF-κB, p53, and MDA, along with reduced SOD and CAT activities; these changes were significantly attenuated by bosentan administration. Bosentan treatment significantly reduced myocardial ET-1 levels (from 136.88 ± 5.02 to 120.18 ± 2.67 nmol/g, p = 0.003), NF-κB levels (from 0.87 ± 0.04 to 0.51 ± 0.03 ng/mg, p = 0.002), and TGF-β1 expression (from 1.72 ± 0.10 to 0.91 ± 0.08 relative units, p = 0.001). Moreover, bosentan increased antioxidant enzyme activities, elevating SOD levels from 21.45 ± 1.23 to 32.67 ± 1.45 U/mg protein (p = 0.001) and CAT levels from 15.22 ± 0.98 to 25.36 ± 1.12 U/mg protein (p = 0.002). Conclusions: Bosentan exerts cardioprotective effects in rats subjected to lung I/R injury by reducing myocardial damage, inflammation, and oxidative stress. These findings suggest that bosentan may serve as a potential therapeutic agent for preventing remote organ injury associated with pulmonary I/R. Full article
(This article belongs to the Section Cardiology)
Show Figures

Figure 1

23 pages, 1524 KB  
Review
Primary Graft Dysfunction in Lung Transplantation: An Overview of the Molecular Mechanisms
by Jitte Jennekens, Sue A. Braithwaite, Bart Luijk, Niels P. van der Kaaij, Nienke Vrisekoop, Saskia C. A. de Jager and Linda M. de Heer
Int. J. Mol. Sci. 2025, 26(14), 6776; https://doi.org/10.3390/ijms26146776 - 15 Jul 2025
Cited by 1 | Viewed by 1773
Abstract
Primary graft dysfunction (PGD) remains a major complication after lung transplantation. Donor lung ischemia followed by reperfusion drives oxidative stress and inflammatory responses. The pathophysiology is influenced by various donor-, procedure-, and recipient-related factors, which complicates the identification of biomarkers for evaluation of [...] Read more.
Primary graft dysfunction (PGD) remains a major complication after lung transplantation. Donor lung ischemia followed by reperfusion drives oxidative stress and inflammatory responses. The pathophysiology is influenced by various donor-, procedure-, and recipient-related factors, which complicates the identification of biomarkers for evaluation of donor lung injury or therapeutic interventions to minimize PGD. This review provides an overview of the molecular pathways that contribute to PGD pathophysiology, including those involved in loss of endothelial–epithelial membrane integrity, neutrophil infiltration, and the development of pulmonary edema. Full article
Show Figures

Figure 1

20 pages, 5004 KB  
Article
Maresin1 Alleviates Ischemia Reperfusion Injury After Lung Transplantation by Inhibiting Ferroptosis via the PKA-Hippo-YAP Signaling Pathway
by Peng Deng, You Wu, Li Wan, Xiangfu Sun and Quanchao Sun
Biomedicines 2025, 13(7), 1594; https://doi.org/10.3390/biomedicines13071594 - 30 Jun 2025
Cited by 2 | Viewed by 1215
Abstract
Background: Lung ischemia reperfusion injury (LIRI) is a severe complication after lung transplantation (LT). Ferroptosis contributes to the pathogenesis of LIRI. Maresin1 (MaR1) is an endogenous pro-resolving lipid mediator that exerts protective effects against multiorgan diseases. However, the role and mechanism of [...] Read more.
Background: Lung ischemia reperfusion injury (LIRI) is a severe complication after lung transplantation (LT). Ferroptosis contributes to the pathogenesis of LIRI. Maresin1 (MaR1) is an endogenous pro-resolving lipid mediator that exerts protective effects against multiorgan diseases. However, the role and mechanism of MaR1 in the ferroptosis of LIRI after LT need to be further investigated. Methods: A mouse LT model and a pulmonary vascular endothelial cell line after hypoxia reoxygenation (H/R) culture were established in our study. Histological morphology and inflammatory cytokine levels predicted the severity of LIRI. Cell viability and cell injury were determined by CCK-8 and LDH assays. Ferroptosis biomarkers, including Fe2+, MDA, 4-HNE, and GSH, were assessed by relevant assay kits. Transferrin receptor (TFRC) and Acyl-CoA Synthetase Long Chain Family Member 4 (ACSL4) protein levels were examined by western blotting. In vitro, lipid peroxide levels were detected by DCFH-DA staining and flow cytometry analysis. The ultrastructure of mitochondria was imaged using transmission electron microscopy. Furthermore, the potential mechanism by which MaR1 regulates ferroptosis was explored and verified with signaling pathway inhibitors using Western blotting. Results: MaR1 protected mice from LIRI after LTx, which was reversed by the ferroptosis agonist Sorafenib in vivo. MaR1 administration decreased Fe2+, MDA, 4-HNE, TFRC, and ACSL4 contents, increased GSH levels, and ameliorated mitochondrial ultrastructural injury after LTx. In vitro, Sorafenib resulted in lower cell viability and worsened cell injury and enhanced the hallmarks of ferroptosis after H/R culture, which was rescued by MaR1 treatment. Mechanistically, the protein kinase A and YAP inhibitors partly blocked the effects of MaR1 on ferroptosis inhibition and LIRI protection. Conclusions: This study revealed that MaR1 alleviates LIRI and represses ischemia reperfusion-induced ferroptosis via the PKA-Hippo-YAP signaling pathway, which may offer a promising theoretical basis for the clinical application of organ protection after LTx. Full article
(This article belongs to the Special Issue The Role of Iron in Human Diseases)
Show Figures

Figure 1

33 pages, 2003 KB  
Review
Acute Compartment Syndrome and Intra-Abdominal Hypertension, Decompression, Current Pharmacotherapy, and Stable Gastric Pentadecapeptide BPC 157 Solution
by Predrag Sikiric, Sven Seiwerth, Anita Skrtic, Mario Staresinic, Sanja Strbe, Antonia Vuksic, Suncana Sikiric, Dinko Bekic, Toni Penovic, Dominik Drazenovic, Tomislav Becejac, Marijan Tepes, Zrinko Madzar, Luka Novosel, Lidija Beketic Oreskovic, Ivana Oreskovic, Mirjana Stupnisek, Alenka Boban Blagaic and Ivan Dobric
Pharmaceuticals 2025, 18(6), 866; https://doi.org/10.3390/ph18060866 - 10 Jun 2025
Cited by 1 | Viewed by 2667
Abstract
In this study, pharmacotherapies of abdominal compartment syndrome (ACS) and intra-abdominal hypertension (IAH) in animal studies were reviewed from the perspective of ACS/IAH as failed cytoprotection issues, as non-specific injuries, and from the point of view of the cytoprotection concept as resolution. Therefore, [...] Read more.
In this study, pharmacotherapies of abdominal compartment syndrome (ACS) and intra-abdominal hypertension (IAH) in animal studies were reviewed from the perspective of ACS/IAH as failed cytoprotection issues, as non-specific injuries, and from the point of view of the cytoprotection concept as resolution. Therefore, this review challenges the unresolved theoretical and practical issues of severe multiorgan failure, acknowledged significance in clinics, and resolving outcomes (i.e., open abdomen). Generally, the reported agents not aligned with cytoprotection align with current pharmacotherapy limitations and have (non-)confirmed effectiveness, mostly in only one organ, mild/moderate IAH, prophylactic application, and provide only a tentative resolution. Contrarily, stable gastric pentadecapeptide BPC 157 therapy, as a novel and relevant cytoprotective mediator having pleiotropic beneficial effects, simultaneously resolves many targets, resolving established disturbances, specifically compression/ischemia (grade III and grade IV), and decompression/advanced reperfusion. BPC 157 therapy rapidly activates collateral bypassing pathways, and, in ACS and IAH, and later, in reperfusion, there is a “bypassing key” (i.e., azygos vein direct blood flow delivery). This serves to counteract multiorgan and vessel failure, including lesions and hemorrhages in the brain, heart, lung, liver, kidney and gastrointestinal tract, thrombosis, peripherally and centrally, intracranial (superior sagittal sinus), portal and caval hypertension and aortal hypotension, occlusion/occlusion-like syndrome, advanced Virchow triad circumstances, and free radical formation acting as a membrane stabilizer and free radical scavenger. Likewise, not only in ACS/IAH resolving, but also in other occlusion/occlusion-like syndromes, this “bypassing key” could be an effect of the essential endothelial cytoprotective capacity of BPC 157 and a particular modulatory effect on the NO-system, and a rescuing impact on vasomotor tone. Full article
(This article belongs to the Section Pharmacology)
Show Figures

Graphical abstract

Back to TopTop