1
|
Elbaki BTA, Sameh H, El-Haleem MRA, Abd-Elsattar AA. Possible protective effect of quercetin on lung injury induced by skeletal muscle ischemia reperfusion (IR) injury of adult male albino rats: Histological and biochemical study. J Mol Histol 2024; 56:48. [PMID: 39699779 DOI: 10.1007/s10735-024-10303-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2024] [Accepted: 10/13/2024] [Indexed: 12/20/2024]
Abstract
When a lower limb is injured, the most delicate organs that are at risk of harm are the lungs. Among the flavonoids, quercetin is a significant component that is found in apples and onions in the highest proportions. Numerous biological actions, including as anti-inflammatory, antioxidant, and anti-cancer properties, have been linked to quercetin. To investigate the impact of quercetin on lung injury induced by skeletal muscle ischemia reperfusion (IR) injury. Three equal groups of twenty-four adult rats were used: control, Ischemia-reperfusion (IR) group and IR group treated with quercetin. Rats in (IR) group were exposed to ischemia by ligation of femoral artery for 2h then after removal of the clamp, reperfusion was estabilished for another 24h. IR group treated with quercetin, rats were underwent hind limb IR as described in group II then were given quercetin that was administered at a dose of 20mg/kg intraperitoneally. Measurement of cytokines in serum, MDA in tissue homogenate and VEGF in serum and tissue homogrnate in addition to mRNA expression level and detection of protein level of both sirtuin-1(SIRT1) and NF-κB were assessed at the end of experiment. Histological and immunohistochemical assessment of the lungs were also carried. IR group showed notable rise of inflammatory cytokines such as IL-1β, IL-6 and TNF-α in addition to high level of VEGF and MDA in IR group when compared to the IR group treated with quercetin. Also, gene expression and protein level of SIRT1 were reduced while NF-κB mRNA expression and protein level were significantly upregulated in IR group compared to IR group treated with quercetin. Histologically, IR group indicated marked histological alterations of lung tissue. Also, IR showed strong brownish cytoplasmic immunostaining for iNOS and abundance of Ki67-positive cells. These alterations were significantly reversed in IR group treated with quercetin. Biochemical and immunohistochemical findings of this study demonstrate that quercetin administration have protective effects against lung injury induced by lower limb IR.
Collapse
Affiliation(s)
- Bassant T Abd Elbaki
- Department of Medical Histology and Cell Biology, Faculty of Medicine, Zagazig University, Zagazig, Egypt
| | - Hend Sameh
- Department of Medical Biochemistry, Faculty of Medicine, Zagazig University, Zagazig, Egypt
| | - Manal R Abd El-Haleem
- Department of Histology and Cell Biology, Faculty of Medicine, Suez University, Suez, Egypt
| | - Alyaa A Abd-Elsattar
- Department of Medical Histology and Cell Biology, Faculty of Medicine, Zagazig University, Zagazig, Egypt.
| |
Collapse
|
2
|
Terinte-Balcan G, Lebraud E, Zuber J, Anglicheau D, Ismail G, Rabant M. Deciphering the Complexity of the Immune Cell Landscape in Kidney Allograft Rejection. Transpl Int 2024; 37:13835. [PMID: 39722854 PMCID: PMC11668586 DOI: 10.3389/ti.2024.13835] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2024] [Accepted: 11/26/2024] [Indexed: 12/28/2024]
Abstract
While the Banff classification dichotomizes kidney allograft rejection based on the localization of the cells in the different compartments of the cortical kidney tissue [schematically interstitium for T cell mediated rejection (TCMR) and glomerular and peritubular capillaries for antibody-mediated rejection (AMR)], there is a growing evidences that subtyping the immune cells can help refine prognosis prediction and treatment tailoring, based on a better understanding of the pathophysiology of kidney allograft rejection. In the last few years, multiplex IF techniques and automatic counting systems as well as transcriptomics studies (bulk, single-cell and spatial techniques) have provided invaluable clues to further decipher the complex puzzle of rejection. In this review, we aim to better describe the inflammatory infiltrates that occur during the course of kidney transplant rejection (active AMR, chronic active AMR and acute and chronic active TCMR). We also discuss minor components of the inflammatory response (mastocytes, eosinophils, neutrophils, follicular dendritic cells). We conclude by discussing whether the over simplistic dichotomy between AMR and TCMR, currently used in clinical routine, remains relevant given the great diversity of immune actors involved in rejections.
Collapse
Affiliation(s)
- George Terinte-Balcan
- Nephrology department, “Carol Davila” University of Medicine and Pharmacy, Bucharest, Romania
- Department of Pathology, Necker-Enfants Malades Hospital, Assistance Publique—Hopitaux de Paris, Paris, France
- Centre National de la Recherche Scientifique (CNRS), Inserm U1151, Institut Necker-Enfants Malades, Université Paris Cité, Paris, France
| | - Emilie Lebraud
- Centre National de la Recherche Scientifique (CNRS), Inserm U1151, Institut Necker-Enfants Malades, Université Paris Cité, Paris, France
| | - Julien Zuber
- Department of Kidney and Metabolic Diseases, Transplantation and Clinical Immunology, Necker Hospital, Assistance Publique Hôpitaux de Paris (AP-HP), Paris, France
| | - Dany Anglicheau
- Centre National de la Recherche Scientifique (CNRS), Inserm U1151, Institut Necker-Enfants Malades, Université Paris Cité, Paris, France
- Department of Kidney and Metabolic Diseases, Transplantation and Clinical Immunology, Necker Hospital, Assistance Publique Hôpitaux de Paris (AP-HP), Paris, France
| | - Gener Ismail
- Nephrology department, “Carol Davila” University of Medicine and Pharmacy, Bucharest, Romania
- Department of Nephrology, Fundeni Clinical Institute, Bucharest, Romania
| | - Marion Rabant
- Department of Pathology, Necker-Enfants Malades Hospital, Assistance Publique—Hopitaux de Paris, Paris, France
- Centre National de la Recherche Scientifique (CNRS), Inserm U1151, Institut Necker-Enfants Malades, Université Paris Cité, Paris, France
| |
Collapse
|
3
|
Wu W, Meng F, Zhang H, Tian H, Zhang X. Neutrophil PPIF exacerbates lung ischemia-reperfusion injury after lung transplantation by promoting calcium overload-induced neutrophil extracellular traps formation. Int Immunopharmacol 2024; 142:113051. [PMID: 39236457 DOI: 10.1016/j.intimp.2024.113051] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2024] [Revised: 08/24/2024] [Accepted: 08/29/2024] [Indexed: 09/07/2024]
Abstract
Lung ischemia-reperfusion (I/R) injury is the main risk factor for primary graft dysfunction and patient death after lung transplantation (LTx). It is widely accepted that the main pathological mechanism of lung I/R injury are calcium overload, oxygen free radical explosion and neutrophil-mediated damage, which leading to the lack of effective treatment options. The aim of this study was to further explore the mechanisms of lung I/R injury after LTx and to provide potential therapeutic strategies. Our bioinformatics analysis revealed that the neutrophil extracellular traps (NETs) formation was closely involved in lung I/R injury after LTx, which was accompanied by up-regulation of peptidylprolyl isomerase F (PPIF) and peptidyl arginine deiminase 4 (PADI4). We further established an orthotopic LTx mouse model to simulate lung I/R injury in vivo, and found that PPIF and PADI4 inhibitors effectively reduced neutrophil infiltration, NETs formation, inflammatory response, and lung I/R injury. In the neutrophil model induced by HL-60 cell line in vitro, we found that PPIF inhibitor cyclosporin A (Cys A) better alleviated calcium overload induced inflammatory response, reactive oxygen species content and NETs formation. Further study demonstrated that interfering with neutrophil PPIF protected mitochondrial function by alleviating store-operated calcium entry (SOCE) during calcium overload and played the above positive role. On this basis, we found that the reduction of calcium content in neutrophils was accompanied by the inhibition of calcineurin (CN) and nuclear factor of activated T cells (NFAT). In conclusion, our findings suggested that neutrophil PPIF could serve as a novel biomarker and potential therapeutic target of lung I/R injury after LTx, which provided new clues for its treatment by inhibiting calcium overload-induced NETs formation.
Collapse
Affiliation(s)
- Wensi Wu
- Department of Thoracic Surgery, Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Fanqing Meng
- Department of Anesthesiology, Jinan Maternity and Child Care Hospital, Jinan, China
| | - Huiying Zhang
- Department of Thoracic Surgery, Qilu Hospital of Shandong University, Jinan, China
| | - Hui Tian
- Department of Thoracic Surgery, Qilu Hospital of Shandong University, Jinan, China.
| | - Xiaojun Zhang
- Department of Anesthesiology, Qilu Hospital of Shandong University, Jinan, China.
| |
Collapse
|
4
|
Kollareth DJM, Sharma AK. Precision cut lung slices: an innovative tool for lung transplant research. Front Immunol 2024; 15:1504421. [PMID: 39669559 PMCID: PMC11634892 DOI: 10.3389/fimmu.2024.1504421] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Accepted: 11/11/2024] [Indexed: 12/14/2024] Open
Abstract
Lung ischemia-reperfusion injury (IRI), a common complication after lung transplantation (LTx), plays a crucial role in both primary graft dysfunction (PGD) and chronic lung allograft dysfunction (CLAD) thereby adversely impacting the clinical outcomes in these patient cohorts. Lung IRI is characterized by several molecular events including immune cell infiltration, reactive oxygen species (ROS) generation, calcium overload, inflammation and various forms of cell death pathways. Currently, no therapeutic agents are available to clinically prevent lung IRI. While animal and cell culture models are highly valuable in understanding the pathophysiology of lung IRI, they may not completely recapitulate the complexity of human lung tissue pathology. This limitation necessitates the requirement for developing innovative preclinical human research tools that can supplement available scientific modalities. Emerging evidence suggests that precision-cut lung slices (PCLS) have become an indispensable tool in scientific research to study lung biology in an ex vivo tissue system. Recent studies using PCLS have investigated lung diseases including asthma, chronic obstructive pulmonary disease (COPD), and lung cancer. Although PCLS can be successfully employed to determine the deleterious events in the pathogenesis of lung IRI, including cell-cell interactions as well as hallmarks of inflammation and oxidative stress-dependent pathways, detailed studies employing PCLS to decipher these molecular events in post-LTx injury are currently limited. This review focuses on the applicability and unexplored potential of PCLS as a powerful tool in lung IRI research for understanding the pathophysiology and consequent development of new therapeutic modalities.
Collapse
Affiliation(s)
| | - Ashish K. Sharma
- Department of Surgery, University of Florida, Gainesville, FL, United States
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, University of Florida, Gainesville, FL, United States
- Department of Pharmacology and Therapeutics, University of Florida, Gainesville, FL, United States
| |
Collapse
|
5
|
Van Slambrouck J, Loopmans S, Prisciandaro E, Barbarossa A, Kortleven P, Feys S, Vandervelde CM, Jin X, Cenik I, Moermans K, Fieuws S, Provoost AL, Willems A, De Leyn P, Van Veer H, Depypere L, Jansen Y, Pirenne J, Neyrinck A, Weynand B, Vanaudenaerde B, Carmeliet G, Vos R, Van Raemdonck D, Ghesquière B, Van Weyenbergh J, Ceulemans LJ. The effect of rewarming ischemia on tissue transcriptome and metabolome signatures: A clinical observational study in lung transplantation. J Heart Lung Transplant 2024:S1053-2498(24)01905-3. [PMID: 39486771 DOI: 10.1016/j.healun.2024.10.020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2024] [Revised: 09/17/2024] [Accepted: 10/21/2024] [Indexed: 11/04/2024] Open
Abstract
BACKGROUND In lung transplantation (LuTx), various ischemic phases exist, yet the rewarming ischemia time (RIT) during implantation has often been overlooked. During RIT, lungs are deflated and exposed to the body temperature in the recipient's chest cavity. Our prior clinical findings demonstrated that prolonged RIT increases the risk of primary graft dysfunction. However, the molecular mechanisms of rewarming ischemic injury in this context remain unexplored. We aimed to characterize the rewarming ischemia phase during LuTx by measuring organ temperature and comparing transcriptome and metabolome profiles in tissue obtained at the end versus the start of implantation. METHODS In a clinical observational study, 34 double-LuTx with ice preservation were analyzed. Lung core and surface temperature (n = 65 and 55 lungs) were measured during implantation. Biopsies (n = 59 lungs) were wedged from right middle lobe and left lingula at start and end of implantation. Tissue transcriptomic and metabolomic profiling were performed. RESULTS Temperature increased rapidly during implantation, reaching core/surface temperatures of 21.5°C/25.4°C within 30 minutes. Transcriptomics showed increased proinflammatory signaling and oxidative stress at the end of implantation. Upregulation of NLRP3 and NFKB1 correlated with RIT. Metabolomics indicated elevated levels of amino acids, hypoxanthine, uric acid, and cysteineglutathione disulfide alongside decreased levels of glucose and carnitines. Arginine, tyrosine, and 1-carboxyethylleucine showed a correlation with incremental RIT. CONCLUSIONS The final rewarming ischemia phase in LuTx involves rapid organ rewarming, accompanied by transcriptomic and metabolomic changes indicating proinflammatory signaling and disturbed cell metabolism. Limiting implantation time and cooling of the lung represent potential interventions to alleviate rewarming ischemic injury.
Collapse
Affiliation(s)
- Jan Van Slambrouck
- Department of Thoracic Surgery, University Hospitals Leuven, Leuven, Belgium; Department of Chronic Diseases and Metabolism, Laboratory of Respiratory Diseases and Thoracic Surgery (BREATHE), KU Leuven, Leuven, Belgium
| | - Shauni Loopmans
- Department of Cellular and Molecular Medicine, Laboratory of Applied Mass Spectrometry, KU Leuven, Leuven, Belgium; Center for Cancer Biology, Metabolomics Core Facility Leuven, VIB, Leuven, Belgium
| | - Elena Prisciandaro
- Department of Thoracic Surgery, University Hospitals Leuven, Leuven, Belgium; Department of Chronic Diseases and Metabolism, Laboratory of Respiratory Diseases and Thoracic Surgery (BREATHE), KU Leuven, Leuven, Belgium
| | - Annalisa Barbarossa
- Department of Thoracic Surgery, University Hospitals Leuven, Leuven, Belgium; Department of Chronic Diseases and Metabolism, Laboratory of Respiratory Diseases and Thoracic Surgery (BREATHE), KU Leuven, Leuven, Belgium
| | - Phéline Kortleven
- Department of Chronic Diseases and Metabolism, Laboratory of Respiratory Diseases and Thoracic Surgery (BREATHE), KU Leuven, Leuven, Belgium; Department of Pharmaceutical and Pharmacological Sciences, Molecular Virology and Gene Therapy, KU Leuven, Leuven, Belgium
| | - Simon Feys
- Department of Microbiology, Immunology and Transplantation, Laboratory of Clinical Infectious and Inflammatory Disorders, KU Leuven, Leuven, Belgium; Department of Medical Intensive Care, University Hospitals Leuven, Leuven, Belgium
| | - Christelle M Vandervelde
- Department of Thoracic Surgery, University Hospitals Leuven, Leuven, Belgium; Department of Chronic Diseases and Metabolism, Laboratory of Respiratory Diseases and Thoracic Surgery (BREATHE), KU Leuven, Leuven, Belgium
| | - Xin Jin
- Department of Chronic Diseases and Metabolism, Laboratory of Respiratory Diseases and Thoracic Surgery (BREATHE), KU Leuven, Leuven, Belgium
| | - Ismail Cenik
- Department of Thoracic Surgery, University Hospitals Leuven, Leuven, Belgium
| | - Karen Moermans
- Department of Chronic Diseases and Metabolism, Laboratory of Clinical and Experimental Endocrinology, KU Leuven, Leuven, Belgium
| | - Steffen Fieuws
- Department of Public Health, Interuniversity Center for Biostatistics and Statistical Bioinformatics, KU Leuven, Leuven, Belgium
| | - An-Lies Provoost
- Department of Thoracic Surgery, University Hospitals Leuven, Leuven, Belgium; Department of Chronic Diseases and Metabolism, Laboratory of Respiratory Diseases and Thoracic Surgery (BREATHE), KU Leuven, Leuven, Belgium
| | - Anton Willems
- Department of Cellular and Molecular Medicine, Laboratory of Applied Mass Spectrometry, KU Leuven, Leuven, Belgium; Center for Cancer Biology, Metabolomics Core Facility Leuven, VIB, Leuven, Belgium
| | - Paul De Leyn
- Department of Thoracic Surgery, University Hospitals Leuven, Leuven, Belgium; Department of Chronic Diseases and Metabolism, Laboratory of Respiratory Diseases and Thoracic Surgery (BREATHE), KU Leuven, Leuven, Belgium
| | - Hans Van Veer
- Department of Thoracic Surgery, University Hospitals Leuven, Leuven, Belgium; Department of Chronic Diseases and Metabolism, Laboratory of Respiratory Diseases and Thoracic Surgery (BREATHE), KU Leuven, Leuven, Belgium
| | - Lieven Depypere
- Department of Thoracic Surgery, University Hospitals Leuven, Leuven, Belgium; Department of Chronic Diseases and Metabolism, Laboratory of Respiratory Diseases and Thoracic Surgery (BREATHE), KU Leuven, Leuven, Belgium
| | - Yanina Jansen
- Department of Thoracic Surgery, University Hospitals Leuven, Leuven, Belgium; Department of Chronic Diseases and Metabolism, Laboratory of Respiratory Diseases and Thoracic Surgery (BREATHE), KU Leuven, Leuven, Belgium
| | - Jacques Pirenne
- Department of Microbiology, Immunology and Transplantation, Laboratory of Abdominal Transplantation, KU Leuven, Leuven, Belgium; Department of Abdominal Transplant Surgery, University Hospitals Leuven, Leuven, Belgium
| | - Arne Neyrinck
- Department of Cardiovascular Sciences, Anesthesiology and Algology, KU Leuven, Leuven, Belgium; Department of Anesthesiology, University Hospitals Leuven, Leuven, Belgium
| | - Birgit Weynand
- Department of Pathology, University Hospitals Leuven, Leuven, Belgium; Department of Imaging and Pathology, Laboratory of Translational Cell & Tissue Research, KU Leuven, Leuven, Belgium
| | - Bart Vanaudenaerde
- Department of Chronic Diseases and Metabolism, Laboratory of Respiratory Diseases and Thoracic Surgery (BREATHE), KU Leuven, Leuven, Belgium
| | - Geert Carmeliet
- Department of Chronic Diseases and Metabolism, Laboratory of Clinical and Experimental Endocrinology, KU Leuven, Leuven, Belgium
| | - Robin Vos
- Department of Chronic Diseases and Metabolism, Laboratory of Respiratory Diseases and Thoracic Surgery (BREATHE), KU Leuven, Leuven, Belgium; Department of Respiratory Diseases, University Hospitals Leuven, Leuven, Belgium
| | - Dirk Van Raemdonck
- Department of Thoracic Surgery, University Hospitals Leuven, Leuven, Belgium; Department of Chronic Diseases and Metabolism, Laboratory of Respiratory Diseases and Thoracic Surgery (BREATHE), KU Leuven, Leuven, Belgium
| | - Bart Ghesquière
- Department of Cellular and Molecular Medicine, Laboratory of Applied Mass Spectrometry, KU Leuven, Leuven, Belgium; Center for Cancer Biology, Metabolomics Core Facility Leuven, VIB, Leuven, Belgium
| | - Johan Van Weyenbergh
- Department of Microbiology, Immunology and Transplantation, Laboratory of Clinical and Epidemiological Virology, Rega Institute, KU Leuven, Leuven, Belgium
| | - Laurens J Ceulemans
- Department of Thoracic Surgery, University Hospitals Leuven, Leuven, Belgium; Department of Chronic Diseases and Metabolism, Laboratory of Respiratory Diseases and Thoracic Surgery (BREATHE), KU Leuven, Leuven, Belgium.
| |
Collapse
|
6
|
Kaur G, Wang Q, Tjitropranoto A, Unwalla H, Rahman I. Cold ischemia time alters cell-type specific senescence leading to loss of cellular integrity in mouse lungs. Exp Lung Res 2024; 50:184-198. [PMID: 39427288 PMCID: PMC11513191 DOI: 10.1080/01902148.2024.2414974] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2024] [Revised: 09/11/2024] [Accepted: 10/01/2024] [Indexed: 10/22/2024]
Abstract
Purpose: Ischemia-reperfusion injury (IRI) is a major challenge in lung transplantation often causing graft dysfunction and chronic airway illnesses in recipients. To prevent potential transplant related complications, strict guidelines were put in place to choose viable donor lungs with minimal risk of IRI. These regulations deem most of the donor organs unfit for transplant which then are donated for research to understand the mechanisms of health and diseases in human. However, resected organs that are being transported undergo cold ischemia that can negatively affect the tissue architecture and other cellular functions under study. Thus, it is important to assess how cold ischemia time (CIT) affects the physiological mechanism. In this respect, we are interested in studying how CIT affects cellular senescence in normal aging and various pulmonary pathologies. We thus hypothesized that prolonged CIT exhibits cell-type specific changes in lung cellular senescence in mice. Methods: Lung lobes from C57BL/6J (n = 5-8) mice were harvested and stored in UW Belzer cold storage solution for 0, 4-, 9-, 12-, 24-, and 48-h CIT. Lung cellular senescence was determined using fluorescence (C12FdG) assay and co-immunolabelling was performed to identify changes in individual cell types. Results: We found a rapid decline in the overall lung cellular senescence after 4-h of CIT in our study. Co-immunolabelling revealed the endothelial cells to be most affected by cold ischemia, demonstrating significant decrease in the endothelial cell senescence immediately after harvest. Annexin V-PI staining further revealed a prominent increase in the number of necrotic cells at 4-h CIT, thus suggesting that most of the cells undergo cell death within a few hours of cold ischemic injury. Conclusions: We thus concluded that CIT significantly lowers the cellular senescence in lung tissues and must be considered as a confounding factor for mechanistic studies in the future.
Collapse
Affiliation(s)
- Gagandeep Kaur
- Department of Environmental Medicine, University of Rochester Medical Center, Rochester, NY
| | - Qixin Wang
- Department of Environmental Medicine, University of Rochester Medical Center, Rochester, NY
| | - Ariel Tjitropranoto
- Department of Environmental Medicine, University of Rochester Medical Center, Rochester, NY
| | - Hoshang Unwalla
- Department of Cellular and Molecular Medicine, Herbert Wertheim College of Medicine, Florida International University, Miami, FL, USA
| | - Irfan Rahman
- Department of Environmental Medicine, University of Rochester Medical Center, Rochester, NY
| |
Collapse
|
7
|
KOCAK S, GUNER I, YAMAN MO, YILMAZ TEKIZ, MEYDANLI EEGUZEL, YELMEN N, SAHIN G. Alpha B-crystallin Ameliorates Imbalance of Redox Homeostasis, Inflammation and Apoptosis in an Acute Lung Injury Model with Rats. Medeni Med J 2024; 39:211-220. [PMID: 39350576 PMCID: PMC11572206 DOI: 10.4274/mmj.galenos.2024.82274] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2024] [Accepted: 09/09/2024] [Indexed: 11/20/2024] Open
Abstract
Objective Ischemia-reperfusion (IR) of the aorta is a significant contributor to the development of postoperative acute lung damage after abdominal aortic surgery. The aim of the present study was to examine the effect of alpha B-crystallin, a small heat shock protein (known as HspB5), on lung injury induced by abdominal aortic IR in rats. Methods Male Sprague-Dawley rats were divided into three groups: control, ischemia-reperfusion (IR, 90 min ischemia and 180 min reperfusion), and alpha B-crystallin +IR. Alpha B-crystallin (50 μg/100 g) was intraperitoneally administered 1 h before IR. Lung tissue samples were obtained for histological and biochemical analyses of oxidative stress and cytokine and apoptosis parameters in plasma, lung tissues, and bronchoalveolar lavage (BAL) fluid. Results The levels of malondialdehyde, reactive oxygen species, total oxidant status, tumor necrosis factor-alpha (TNF-α), interleukin-1 beta (IL-1β), nuclear factor kappa B (NFKβ), caspase-9 (CASP-9), 8-hydroxy-2'-deoxyguanosine, total antioxidant status, superoxide dismutase, and interleukin-10 levels in lung tissues, plasma, and BAL fluid (p<0.05 versus control) increased in Aortic IR. However, alpha B-crystallin significantly reduced the lung tissue levels of oxidative, inflamatuvar, and apoptotic parameters in the plasma, lung tissues, and BAL fluid (p<0.05 versus aortic IR). Histopathological results showed that alpha B-crystallin ameliorated the morphological changes related to lung injury (p<0.001). Conclusion Alpha B-crystallin substantially restored disrupted the redox balance, inflammation, and apoptotic parameters in rats exposed to IR. The cytoprotective effect of alpha B-crystallin on redox balance might be attributed to improved lung injury.
Collapse
Affiliation(s)
- Seda KOCAK
- Kirsehir Ahi Evran University Faculty of Medicine, Department of Physiology, Kirsehir, Türkiye
| | - Ibrahim GUNER
- Tekirdag Namik Kemal University Faculty of Medicine, Department of Physiology, Tekirdag, Türkiye
| | - Muhittin Onur YAMAN
- Istanbul University-Cerrahpasa, Cerrahpasa Faculty of Medicine, Department of Physiology, Istanbul, Türkiye
| | - Tugba EKIZ YILMAZ
- Istanbul University-Cerrahpasa, Cerrahpasa Faculty of Medicine, Department of Histology and Embriology, Istanbul, Türkiye
| | - Emine Elif GUZEL MEYDANLI
- Istanbul University-Cerrahpasa, Cerrahpasa Faculty of Medicine, Department of Histology and Embriology, Istanbul, Türkiye
| | - Nermin YELMEN
- Istanbul University-Cerrahpasa, Cerrahpasa Faculty of Medicine, Department of Physiology, Istanbul, Türkiye
| | - Gulderen SAHIN
- Istanbul Aydin University Faculty of Medicine, Department of Physiology, Istanbul, Türkiye
| |
Collapse
|
8
|
Eggenhofer E, Proneth B. Ferroptosis Inhibition: A Key Opportunity for the Treatment of Ischemia/Reperfusion Injury in Liver Transplantation. Transplantation 2024:00007890-990000000-00874. [PMID: 39294870 DOI: 10.1097/tp.0000000000005199] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/21/2024]
Abstract
The outcome after liver transplantation has improved in recent years, which can be attributed to superior storage and transportation conditions of the organs, as well as better peri- and postoperative management and advancements in surgical techniques. Nevertheless, there is an increasing discrepancy between the need for organs and their availability. Consequently, the mortality rate on the waiting list is high and continues to rise. One way of counteracting this trend is to increase the use of "expanded criteria donors." This means that more and more donors will be included, especially those who are older and having additional comorbidities (eg, steatosis). A major complication of any transplantation is the occurrence of ischemia/reperfusion injury (IRI), which often leads to liver dysfunction and failure. However, there have been various promising approaches to minimize IRI in recent years, but an effective and clinically applicable method to achieve a better outcome for patients after liver transplantation is still missing. Thereby, the so-called marginal organs are predominantly affected by IRI; thus, it is crucial to develop suitable and effective treatment options for patients. Recently, regulated cell death mechanisms, particularly ferroptosis, have been implicated to play a major role in IRI, including the liver. Therefore, inhibiting this kind of cell death modality presents a promising therapeutic approach for the management of this yet untreatable condition. Thus, this review provides an overview of the role of ferroptosis in liver IRI and transplantation and discusses possible therapeutic solutions based on ferroptosis inhibition to restrain IRI in marginal organs (especially steatosis and donation after circulatory death organs).
Collapse
Affiliation(s)
- Elke Eggenhofer
- Department of Surgery, University Hospital Regensburg, Regensburg, Germany
| | - Bettina Proneth
- Institute of Metabolism and Cell Death, Helmholtz Munich, Neuherberg, Germany
| |
Collapse
|
9
|
Wu SY, Chu SJ, Tang SE, Pao HP, Huang KL, Liao WI. Monomethyl fumarate attenuates lung Ischemia/Reperfusion injury by disrupting the GAPDH/Siah1 signaling cascade. Int Immunopharmacol 2024; 137:112488. [PMID: 38889510 DOI: 10.1016/j.intimp.2024.112488] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2024] [Revised: 06/10/2024] [Accepted: 06/12/2024] [Indexed: 06/20/2024]
Abstract
Monomethyl fumarate (MMF), a potent anti-inflammatory agent used to treat multiple sclerosis, has demonstrated efficacy in various inflammatory and ischemia/reperfusion (IR) models; however, its impact on IR-induced acute lung injury (ALI) has not been explored. We investigated, for the first time, whether MMF attenuates lung IR injury through inhibition of the GAPDH/Siah1 signaling pathway. Rats were subjected to IR injury using an isolated perfused lung model, and proximity ligation assays were employed to evaluate the presence and distribution of the GAPDH/Siah1 complex. In vitro studies involved pretreating human primary alveolar epithelial cells (HPAECs) with MMF and/or inducing GAPDH overexpression or silencing, followed by exposure to hypoxia-reoxygenation. The findings revealed significantly reduced lung damage indicators, including edema, proinflammatory cytokines, oxidative stress and apoptosis, in MMF-treated rats. Notably, MMF treatment inhibited GAPDH/Siah1 complex formation and nuclear translocation, indicating that disruption of the GAPDH/Siah1 cascade was the primary cause of these improvements. Our in vitro studies on pretreated HPAECs corroborate these in vivo findings, further strengthening this interpretation. Our study results suggest that the protective effects of MMF against lung IR injury may be attributed, at least in part, to its ability to disrupt the GAPDH/Siah1 signaling cascade, thereby attenuating inflammatory and apoptotic responses. Given these encouraging results, MMF has emerged as a promising therapeutic candidate for the management of lung IR injury.
Collapse
Affiliation(s)
- Shu-Yu Wu
- Institute of Aerospace and Undersea Medicine, National Defense Medical Center, Taipei, Taiwan; School of Pharmacy, National Defense Medical Center, Taipei, Taiwan
| | - Shi-Jye Chu
- Division of Rheumatology, Immunology and Allergy, Department of Internal Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Shih-En Tang
- Institute of Aerospace and Undersea Medicine, National Defense Medical Center, Taipei, Taiwan; Division of Pulmonary and Critical Care, Department of Internal Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Hsin-Ping Pao
- The Graduate Institute of Medical Sciences, National Defense Medical Center, Taipei, Taiwan
| | - Kun-Lun Huang
- Institute of Aerospace and Undersea Medicine, National Defense Medical Center, Taipei, Taiwan; The Graduate Institute of Medical Sciences, National Defense Medical Center, Taipei, Taiwan
| | - Wen-I Liao
- Department of Emergency Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan.
| |
Collapse
|
10
|
Gao M, Zhu X, Gao X, Yang H, Li H, Du Y, Gao J, Chen Z, Dong H, Wang B, Zhang L. Kaempferol mitigates sepsis-induced acute lung injury by modulating the SphK1/S1P/S1PR1/MLC2 signaling pathway to restore the integrity of the pulmonary endothelial cell barrier. Chem Biol Interact 2024; 398:111085. [PMID: 38823539 DOI: 10.1016/j.cbi.2024.111085] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2024] [Revised: 05/19/2024] [Accepted: 05/29/2024] [Indexed: 06/03/2024]
Abstract
Sepsis-induced acute lung injury (SALI) is the common complication of sepsis, resulting in high incidence and mortality rates. The primary pathogenesis of SALI is the interplay between acute inflammation and endothelial barrier damage. Studies have shown that kaempferol (KPF) has anti-sepsis properties. Sphingosine kinase 1 (SphK1)/sphingosine-1-phosphate (S1P) signaling pathway's significance in acute lung damage and S1P receptor 1 (S1PR1) agonists potential in myosin light chain 2 (MLC2) phosphorylation are documented. Whether KPF can regulate the SphK1/S1P/S1PR1/MLC2 signaling pathway to protect the lung endothelial barrier remains unclear. This study investigates the KPF's therapeutic effects and molecular mechanisms in repairing endothelial cell barrier damage in both LPS-induced sepsis mice and human umbilical vein endothelial cells (HUVECs). KPF significantly reduced lung tissue damage and showed anti-inflammatory effects by decreasing IL-6 and TNF-α synthesis in the sepsis mice model. Further, KPF administration can reduce the high permeability of the LPS-induced endothelial cell barrier and alleviate lung endothelial cell barrier injury. Mechanistic studies showed that KPF pretreatment can suppress MLC2 hyperphosphorylation and decrease SphK1, S1P, and S1PR1 levels. The SphK1/S1P/S1PR1/MLC2 signaling pathway controls the downstream proteins linked to endothelial barrier damage, and the Western blot (WB) showed that KPF raised the protein levels. These proteins include zonula occludens (ZO)-1, vascular endothelial (VE)-cadherin and Occludin. The present work revealed that in mice exhibiting sepsis triggered by LPS, KPF strengthened the endothelial barrier and reduced the inflammatory response. The SphK1/S1P/S1PR1/MLC2 pathway's modulation is the mechanism underlying this impact.
Collapse
Affiliation(s)
- Meijuan Gao
- School of Traditional Chinese Medicine, Binzhou Medical University, Yantai, 264003, PR China
| | - Xuan Zhu
- Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, School of Pharmacy, Yantai University, Yantai, 264005, PR China
| | - XiaoJin Gao
- Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, School of Pharmacy, Yantai University, Yantai, 264005, PR China
| | - Hui Yang
- School of Traditional Chinese Medicine, Binzhou Medical University, Yantai, 264003, PR China
| | - Haixia Li
- Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, School of Pharmacy, Yantai University, Yantai, 264005, PR China
| | - Yuan Du
- Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, School of Pharmacy, Yantai University, Yantai, 264005, PR China
| | - Jing Gao
- Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, School of Pharmacy, Yantai University, Yantai, 264005, PR China
| | - Zhuoxi Chen
- School of Traditional Chinese Medicine, Binzhou Medical University, Yantai, 264003, PR China
| | - Hanpeng Dong
- Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, School of Pharmacy, Yantai University, Yantai, 264005, PR China
| | - Binsheng Wang
- School of Traditional Chinese Medicine, Binzhou Medical University, Yantai, 264003, PR China.
| | - Leiming Zhang
- School of Traditional Chinese Medicine, Binzhou Medical University, Yantai, 264003, PR China.
| |
Collapse
|
11
|
Dai P, He J, Wei Y, Xu M, Zhao J, Zhou X, Tang H. High Dose of Estrogen Protects the Lungs from Ischemia-Reperfusion Injury by Downregulating the Angiotensin II Signaling Pathway. Inflammation 2024; 47:1248-1261. [PMID: 38386131 DOI: 10.1007/s10753-024-01973-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2023] [Revised: 12/21/2023] [Accepted: 01/09/2024] [Indexed: 02/23/2024]
Abstract
We explored the sex difference in lung ischemia-reperfusion injury (LIRI) and the role and mechanism of estrogen (E2) and angiotensin II (Ang II) in LIRI. We established a model of LIRI in mice. E2, Ang II, E2 inhibitor (fulvestrant), and angiotensin II receptor blocker (losartan) were grouped for treatment. The lung wet/dry weight ratio, natural killer (NK) cells (by flow cytometry), neutrophils (by flow cytometry), expression of key proteins (by Western blot, immunohistochemistry, ELISA, and immunofluorescence), and expression of related protein mRNA (by qPCR) were detected. The ultrastructure of the alveolar epithelial cells was observed by transmission electron microscopy. We found that E2 and Ang II played an important role in the progression of LIRI. The two signaling pathways showed obvious antagonism, and E2 regulates LIRI in the different sexes by downregulating Ang II, leading to a better prognosis. E2 and losartan reduced the inflammatory cell infiltration in lung tissue and key inflammatory factors in serum while fulvestrant and Ang II had the opposite effect. The protective effect of E2 was related with AKT, p38, COX2, and HIF-1α.
Collapse
Affiliation(s)
- Peng Dai
- Department of Thoracic Surgery, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
| | - Jutong He
- Department of Thoracic Surgery, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
| | - Yanhong Wei
- Department of Rheumatology and Immunology, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
| | - Ming Xu
- Department of Thoracic Surgery, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
| | - Jinping Zhao
- Department of Thoracic Surgery, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China.
| | - Xuefeng Zhou
- Department of Thoracic Surgery, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China.
| | - Hexiao Tang
- Department of Thoracic Surgery, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China.
| |
Collapse
|
12
|
Bagheri SM, Allahtavakoli M, Hakimizadeh E. Neuroprotective effect of ischemic postconditioning against hyperperfusion and its mechanisms of neuroprotection. JOURNAL OF RESEARCH IN MEDICAL SCIENCES : THE OFFICIAL JOURNAL OF ISFAHAN UNIVERSITY OF MEDICAL SCIENCES 2024; 29:31. [PMID: 39239075 PMCID: PMC11376715 DOI: 10.4103/jrms.jrms_341_22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/06/2022] [Revised: 05/13/2023] [Accepted: 06/27/2023] [Indexed: 09/07/2024]
Abstract
Background In recent years, stroke and ischemia-reperfusion injury has motivated researchers to find new ways to reduce the complications. Although reperfusion is essential for brain survival, it is like a double-edged sword that may cause further damage to the brain. Ischemic postconditioning (IPostC) refers to the control of blood flow in postischemia-reperfusion that can reduce ischemia-reperfusion injuries. Materials and Methods Articles were collected by searching for the terms: Ischemic postconditioning and neuroprotective and ischemic postconditioning and hyperperfusion. Suitable articles were collected from electronic databases, including ISI Web of Knowledge, Medline/PubMed, ScienceDirect, Embase, Scopus, Biological Abstract, Chemical Abstract, and Google Scholar. Results New investigations show that IPostC has protection against hyperperfusion by reducing the amount of blood flow during reperfusion and thus reducing infarction volume, preventing the blood-brain barrier damage, and reducing the rate of apoptosis through the activation of innate protective systems. Numerous mechanisms have been suggested for IPostC, which include reduction of free radical production, apoptosis, inflammatory factors, and activation of endogenous protective pathways. Conclusion It seems that postconditioning can prevent damage to the brain by reducing the flow and blood pressure caused by hyperperfusion. It can protect the brain against damages such as stroke and hyperperfusion by activating various endogenous protection systems. In the present review article, we tried to evaluate both useful aspects of IPostC, neuroprotective effects, and fight against hyperperfusion.
Collapse
Affiliation(s)
- Seyyed Majid Bagheri
- Physiology-Pharmacology Research Center, Research Institute of Basic Medical Sciences, Rafsanjan University of Medical Sciences, Rafsanjan, Iran
| | - Mohammad Allahtavakoli
- Physiology-Pharmacology Research Center, Research Institute of Basic Medical Sciences, Rafsanjan University of Medical Sciences, Rafsanjan, Iran
- Department of Physiology and Pharmacology, School of Medicine, Rafsanjan University of Medical Sciences, Rafsanjan, Iran
| | - Elham Hakimizadeh
- Physiology-Pharmacology Research Center, Research Institute of Basic Medical Sciences, Rafsanjan University of Medical Sciences, Rafsanjan, Iran
| |
Collapse
|
13
|
Wang J, Sun J, Yu H, Hu C, Wu J, Hu C. Donor Inhalation of Nebulized Dexmedetomidine Alleviates Ischemia-Reperfusion Injury in Rat Lung Transplantation. Pharmacology 2024; 109:293-304. [PMID: 38806015 DOI: 10.1159/000539528] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2023] [Accepted: 05/23/2024] [Indexed: 05/30/2024]
Abstract
INTRODUCTION The occurrence of lung ischemia-reperfusion injury (LIRI) after lung transplantation results in primary graft dysfunction (PGD) in more than 50% of cases, which seriously affects the prognosis of recipients. Currently, donor lung protection is the focus of research on improving graft survival in lung transplant recipients. Dexmedetomidine (Dex) is a widely used general anesthesia adjuvant in clinical practice to alleviate ischemia-reperfusion injury in the lungs, liver, heart, kidneys, and brain. However, intravenous infusion of Dex can cause negative effects on the cardiovascular system. Inhaling nebulized Dex can directly act on the alveolar tissue and alleviate its cardiovascular inhibitory effect by reducing drug intake. This study aimed to investigate the effect of donor nebulized Dex inhalation on LIRI after lung transplantation in rats. METHODS We randomly divided the male Sprague-Dawley rats into donor rats and recipient rats, and allowed the donor rats to inhale nebulized Dex or physiological saline 15 min before surgery. The donor lung was refrigerated for 8 h before each single-lung transplant. After 2 h of reperfusion of the transplanted lung, serum and transplanted lung tissue were collected. The wet-to-dry weight ratio of the lung tissue was measured, arterial blood gas was detected, and histopathology changes, oxidative stress, inflammatory reactions, and apoptosis were evaluated. RESULTS Pretransplant inhalation of Dex through the donor's lung reduced the injury of the transplanted lung, increased the levels of malondialdehyde and myeloperoxidase, and decreased the levels of superoxide dismutase and glutathione in the lung tissue. Moreover, nebulized Dex inhalation of the donor lung inhibited LIRI-induced tumor necrosis factor-α, interleukin-6, and inducible nitric oxide synthase expression and also suppressed nuclear factor kappa B phosphorylation. Nebulized Dex inhalation reduced the rate of cell apoptosis in the transplanted lung tissue by inhibiting the upregulation of Bax, downregulation of Bcl-2, and increase in caspase-3 lysis caused by LIRI. CONCLUSION Inhalation of atomized Dex is a potential donor lung protection strategy, which can be used to reduce LIRI after lung transplantation and may be helpful to improve the occurrence of PGD and prognosis of lung transplant recipients.
Collapse
Affiliation(s)
- Jing Wang
- Department of Anesthesiology, Wuxi People's Hospital Affiliated to Nanjing Medical University, Wuxi, China,
- Department of Anesthesiology, The Third People's Hospital of Chengdu, Affiliated Hospital of Southwest Jiaotong University, Chengdu, China,
| | - Jiaojiao Sun
- Wuxi School of Medicine, Jiangnan University, Wuxi, China
| | - Huizhi Yu
- Department of Anesthesiology, The First Affiliated Hospital of Ningbo University, Ningbo, China
| | - Chunlan Hu
- Department of Anesthesiology, Wuxi People's Hospital Affiliated to Nanjing Medical University, Wuxi, China
| | - Jinbo Wu
- Department of Anesthesiology, Wuxi People's Hospital Affiliated to Nanjing Medical University, Wuxi, China
| | - Chunxiao Hu
- Department of Transplantation Anesthesiology, Wuxi People's Hospital Affiliated to Nanjing Medical University, Wuxi, China
| |
Collapse
|
14
|
Liu T, Wei H, Zhang L, Ma C, Wei Y, Jiang T, Li W. Metformin attenuates lung ischemia-reperfusion injury and necroptosis through AMPK pathway in type 2 diabetic recipient rats. BMC Pulm Med 2024; 24:237. [PMID: 38745191 PMCID: PMC11094932 DOI: 10.1186/s12890-024-03056-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2023] [Accepted: 05/08/2024] [Indexed: 05/16/2024] Open
Abstract
BACKGROUND Diabetes mellitus (DM) can aggravate lung ischemia-reperfusion (I/R) injury and is a significant risk factor for recipient mortality after lung transplantation. Metformin protects against I/R injury in a variety of organs. However, the effect of metformin on diabetic lung I/R injury remains unclear. Therefore, this study aimed to observe the effect and mechanism of metformin on lung I/R injury following lung transplantation in type 2 diabetic rats. METHODS Sprague-Dawley rats were randomly divided into the following six groups: the control + sham group (CS group), the control + I/R group (CIR group), the DM + sham group (DS group), the DM + I/R group (DIR group), the DM + I/R + metformin group (DIRM group) and the DM + I/R + metformin + Compound C group (DIRMC group). Control and diabetic rats underwent the sham operation or left lung transplantation operation. Lung function, alveolar capillary permeability, inflammatory response, oxidative stress, necroptosis and the p-AMPK/AMPK ratio were determined after 24 h of reperfusion. RESULTS Compared with the CIR group, the DIR group exhibited decreased lung function, increased alveolar capillary permeability, inflammatory responses, oxidative stress and necroptosis, but decreased the p-AMPK/AMPK ratio. Metformin improved the function of lung grafts, decreased alveolar capillary permeability, inflammatory responses, oxidative stress and necroptosis, and increased the p-AMPK/AMPK ratio. In contrast, the protective effects of metformin were abrogated by Compound C. CONCLUSIONS Metformin attenuates lung I/R injury and necroptosis through AMPK pathway in type 2 diabetic lung transplant recipient rats.
Collapse
Affiliation(s)
- Tianhua Liu
- Department of Anesthesiology, Hei Long Jiang Province Key Laboratory of Research on Anesthesiology and Critical Care Medicine, Second Affiliated Hospital of Harbin Medical University, NO.246, Xuefu Road, Nangang District, Harbin, Heilongjiang Province, 150081, China
| | - Hong Wei
- Department of Anesthesiology, Sixth Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Lijuan Zhang
- Department of Anesthesiology, Hei Long Jiang Province Key Laboratory of Research on Anesthesiology and Critical Care Medicine, Second Affiliated Hospital of Harbin Medical University, NO.246, Xuefu Road, Nangang District, Harbin, Heilongjiang Province, 150081, China
| | - Can Ma
- Department of Anesthesiology, Hei Long Jiang Province Key Laboratory of Research on Anesthesiology and Critical Care Medicine, Second Affiliated Hospital of Harbin Medical University, NO.246, Xuefu Road, Nangang District, Harbin, Heilongjiang Province, 150081, China
| | - Yuting Wei
- Department of Anesthesiology, Hei Long Jiang Province Key Laboratory of Research on Anesthesiology and Critical Care Medicine, Second Affiliated Hospital of Harbin Medical University, NO.246, Xuefu Road, Nangang District, Harbin, Heilongjiang Province, 150081, China
| | - Tao Jiang
- Department of Anesthesiology, Hei Long Jiang Province Key Laboratory of Research on Anesthesiology and Critical Care Medicine, Second Affiliated Hospital of Harbin Medical University, NO.246, Xuefu Road, Nangang District, Harbin, Heilongjiang Province, 150081, China
| | - Wenzhi Li
- Department of Anesthesiology, Hei Long Jiang Province Key Laboratory of Research on Anesthesiology and Critical Care Medicine, Second Affiliated Hospital of Harbin Medical University, NO.246, Xuefu Road, Nangang District, Harbin, Heilongjiang Province, 150081, China.
| |
Collapse
|
15
|
Cenik I, Van Slambrouck J, Provoost AL, Barbarossa A, Vanluyten C, Boelhouwer C, Vanaudenaerde BM, Vos R, Pirenne J, Van Raemdonck DE, Ceulemans LJ. Controlled Hypothermic Storage for Lung Preservation: Leaving the Ice Age Behind. Transpl Int 2024; 37:12601. [PMID: 38694492 PMCID: PMC11062243 DOI: 10.3389/ti.2024.12601] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Accepted: 03/06/2024] [Indexed: 05/04/2024]
Abstract
Controlled hypothermic storage (CHS) is a recent advance in lung transplantation (LTx) allowing preservation at temperatures higher than those achieved with traditional ice storage. The mechanisms explaining the benefits of CHS compared to conventional static ice storage (SIS) remain unclear and clinical data on safety and feasibility of lung CHS are limited. Therefore, we aimed to provide a focus review on animal experiments, molecular mechanisms, CHS devices, current clinical experience, and potential future benefits of CHS. Rabbit, canine and porcine experiments showed superior lung physiology after prolonged storage at 10°C vs. ≤4°C. In recent molecular analyses of lung CHS, better protection of mitochondrial health and higher levels of antioxidative metabolites were observed. The acquired insights into the underlying mechanisms and development of CHS devices allowed clinical application and research using CHS for lung preservation. The initial findings are promising; however, further data collection and analysis are required to draw more robust conclusions. Extended lung preservation with CHS may provide benefits to both recipients and healthcare personnel. Reduced time pressure between procurement and transplantation introduces flexibility allowing better decision-making and overnight bridging by delaying transplantation to daytime without compromising outcome.
Collapse
Affiliation(s)
- Ismail Cenik
- Department of Thoracic Surgery, University Hospitals Leuven, Leuven, Belgium
- Laboratory of Respiratory Diseases and Thoracic Surgery (BREATHE), Department of Chronic Diseases and Metabolism, KU Leuven, Leuven, Belgium
| | - Jan Van Slambrouck
- Department of Thoracic Surgery, University Hospitals Leuven, Leuven, Belgium
- Laboratory of Respiratory Diseases and Thoracic Surgery (BREATHE), Department of Chronic Diseases and Metabolism, KU Leuven, Leuven, Belgium
| | - An-Lies Provoost
- Department of Thoracic Surgery, University Hospitals Leuven, Leuven, Belgium
- Laboratory of Respiratory Diseases and Thoracic Surgery (BREATHE), Department of Chronic Diseases and Metabolism, KU Leuven, Leuven, Belgium
| | - Annalisa Barbarossa
- Department of Thoracic Surgery, University Hospitals Leuven, Leuven, Belgium
- Laboratory of Respiratory Diseases and Thoracic Surgery (BREATHE), Department of Chronic Diseases and Metabolism, KU Leuven, Leuven, Belgium
| | - Cedric Vanluyten
- Department of Thoracic Surgery, University Hospitals Leuven, Leuven, Belgium
- Laboratory of Respiratory Diseases and Thoracic Surgery (BREATHE), Department of Chronic Diseases and Metabolism, KU Leuven, Leuven, Belgium
| | - Caroline Boelhouwer
- Department of Thoracic Surgery, University Hospitals Leuven, Leuven, Belgium
- Laboratory of Respiratory Diseases and Thoracic Surgery (BREATHE), Department of Chronic Diseases and Metabolism, KU Leuven, Leuven, Belgium
| | | | - Robin Vos
- Laboratory of Respiratory Diseases and Thoracic Surgery (BREATHE), Department of Chronic Diseases and Metabolism, KU Leuven, Leuven, Belgium
- Department of Pulmonology, University Hospitals Leuven, Leuven, Belgium
| | - Jacques Pirenne
- Abdominal Transplant Surgery, University Hospitals Leuven, Leuven, Belgium
- Immunology and Transplantation, Department of Microbiology, KU Leuven, Leuven, Belgium
| | - Dirk E. Van Raemdonck
- Department of Thoracic Surgery, University Hospitals Leuven, Leuven, Belgium
- Laboratory of Respiratory Diseases and Thoracic Surgery (BREATHE), Department of Chronic Diseases and Metabolism, KU Leuven, Leuven, Belgium
| | - Laurens J. Ceulemans
- Department of Thoracic Surgery, University Hospitals Leuven, Leuven, Belgium
- Laboratory of Respiratory Diseases and Thoracic Surgery (BREATHE), Department of Chronic Diseases and Metabolism, KU Leuven, Leuven, Belgium
| |
Collapse
|
16
|
Özgür Y, Işık R, Orhan B, Berçik İnal B, Özer T, Altun Ş, Özcan B. Tocilizumab Reduces Lung Injury in a Rat Lung Ischemia and Reperfusion Model. THORACIC RESEARCH AND PRACTICE 2024; 25:62-67. [PMID: 38454201 PMCID: PMC11114182 DOI: 10.5152/thoracrespract.2024.23061] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/25/2023] [Accepted: 12/22/2023] [Indexed: 03/09/2024]
Abstract
OBJECTIVE In this study, the effect of tocilizumab (TCZ) on lung tissue in lung ischemia-reperfusion (I/R) injury in rats was investigated. MATERIAL AND METHODS A total of 24 Wistar rats were divided into 4 equal groups, with 6 rats in each group: Left lung I/R was applied to I/R groups. In the I/R groups, the left lung hilum was clamped for 45 minutes, and then the clamp was removed and reperfused for 120 minutes. In the TCZ groups, 4 mg/kg and 8 mg/kg of TCZ were administered intraperitoneally to the rats 30 minutes before surgery. RESULTS The tumor necrosis factor-alpha mean value was not statistically significant between the groups (P = .091). Statistically significant results were observed between group I/R-TCZ (8 mg/kg) and group I/R for catalase. (P = .005). Statistically significant results were observed between group I/R-TCZ (8 mg/kg) and group I/R for malondialdehyde. (P = .009). The difference in total ischemia score between group I/R-TCZ (4 mg/kg) and group I/R-TCZ (8 mg/kg) and group I/R was statistically significant (P < .001). In terms of alveolar hemorrhage, there was a statistically significant difference between group I/R-TCZ (4 mg/kg) and group I/R-TCZ (8 mg/kg) and group I/R (P = .01 and P = .002, respectively). There was a statistically significant difference between group I/R-TCZ (8 mg/kg) and group I/R in terms of neutrophil accumulation (P = .01). In terms of interstitial edema, there was a statistically significant difference between group I/R-TCZ (4 mg/kg) and group I/R-TCZ (8 mg/kg) and group I/R (P = .006 and P = .001, respectively). In terms of pulmonary edema, there was a statistically significant difference between group I/R-TCZ (4 mg/kg) and group I/R-TCZ (8 mg/kg) and group I/R (P = .01 and P = .009, respectively). CONCLUSION Lung tissue may be affected by I/R injury and this damage can be reversed with the use of TCZ.
Collapse
Affiliation(s)
- Yücel Özgür
- Bahçelievler State Hospital Anesthesiology and Reanimation Clinic, İstanbul, Turkey
| | - Reyhan Işık
- Department of Medical Biochemistry, Health Sciences University İstanbul Training and Research Hospital, İstanbul, Turkey
| | - Bağnu Orhan
- Department of Medical Biochemistry, Health Sciences University İstanbul Training and Research Hospital, İstanbul, Turkey
| | - Berrin Berçik İnal
- Department of Medical Biochemistry, Health Sciences University İstanbul Training and Research Hospital, İstanbul, Turkey
| | - Tanıl Özer
- Department of Cardiovascular Surgery, Koşuyolu High Specialization Education And Research Hospital, İstanbul, Turkey
| | - Şenel Altun
- Department of Casdiovascular Surgery, Bahçelievler State Hospital, İstanbul, Turkey
| | - Burcu Özcan
- Department of Pathology, Health Sciences University İstanbul Training and Research Hospital, İstanbul, Turkey
| |
Collapse
|
17
|
Kim H, Suh GJ, Kwon WY, Kim KS, Jung YS, Kim T, Park H. Kallistatin deficiency exacerbates neuronal damage after cardiac arrest. Sci Rep 2024; 14:4279. [PMID: 38383562 PMCID: PMC10881987 DOI: 10.1038/s41598-024-54415-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2023] [Accepted: 02/13/2024] [Indexed: 02/23/2024] Open
Abstract
The purpose of study was to evaluate that kallistatin deficiency causes excessive production of reactive oxygen species and exacerbates neuronal injury after cardiac arrest. For in vitro study, kallistatin knockdown human neuronal cells were given ischemia-reperfusion injury, and the oxidative stress and apoptosis were evaluated. For clinical study, cardiac arrest survivors admitted to the ICU were divided into the good (CPC 1-2) and poor (CPC 3-5) 6-month neurological outcome groups. The serum level of kallistatin, Nox-1, H2O2 were measured. Nox-1 and H2O2 levels were increased in the kallistatin knockdown human neuronal cells with ischemia-reperfusion injury (p < 0.001) and caspase-3 was elevated and apoptosis was promoted (SERPINA4 siRNA: p < 0.01). Among a total of 62 cardiac arrest survivors (16 good, 46 poor), serum kallistatin were lower, and Nox-1 were higher in the poor neurological group at all time points after admission to the ICU (p = 0.013 at admission; p = 0.020 at 24 h; p = 0.011 at 72 h). At 72 h, H2O2 were higher in the poor neurological group (p = 0.038). Kallistatin deficiency exacerbates neuronal ischemia-reperfusion injury and low serum kallistatin levels were associated with poor neurological outcomes in cardiac arrest survivors.
Collapse
Affiliation(s)
- Hayoung Kim
- Department of Emergency Medicine, Seoul National University Hospital, Seoul, Republic of Korea
- Research Center for Disaster Medicine, Seoul National University Medical Research Center, Seoul, Republic of Korea
| | - Gil Joon Suh
- Department of Emergency Medicine, Seoul National University Hospital, Seoul, Republic of Korea.
- Research Center for Disaster Medicine, Seoul National University Medical Research Center, Seoul, Republic of Korea.
- Department of Emergency Medicine, Seoul National University College of Medicine, Seoul, Republic of Korea.
| | - Woon Yong Kwon
- Department of Emergency Medicine, Seoul National University Hospital, Seoul, Republic of Korea
- Research Center for Disaster Medicine, Seoul National University Medical Research Center, Seoul, Republic of Korea
- Department of Emergency Medicine, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Kyung Su Kim
- Department of Emergency Medicine, Seoul National University Hospital, Seoul, Republic of Korea
- Research Center for Disaster Medicine, Seoul National University Medical Research Center, Seoul, Republic of Korea
| | - Yoon Sun Jung
- Department of Critical Care Medicine, Seoul National University Hospital, Seoul, Republic of Korea
| | - Taegyun Kim
- Department of Emergency Medicine, Seoul National University Hospital, Seoul, Republic of Korea
- Research Center for Disaster Medicine, Seoul National University Medical Research Center, Seoul, Republic of Korea
- Department of Emergency Medicine, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Heesu Park
- Department of Emergency Medicine, Seoul National University Hospital, Seoul, Republic of Korea
- Research Center for Disaster Medicine, Seoul National University Medical Research Center, Seoul, Republic of Korea
| |
Collapse
|
18
|
Steinkühler T, Yang S, Hu MA, Jainandunsing JS, Jager NM, Erasmus ME, Struys MMRF, Bosch DJ, van Meurs M, Jabaudon M, Richard D, Timens W, Leuvenink HGD, Nieuwenhuijs-Moeke GJ. Ex Vivo Optimization of Donor Lungs with Inhaled Sevoflurane during Normothermic Ex Vivo Lung Perfusion (VITALISE): A Pilot and Feasibility Study in Sheep. Int J Mol Sci 2024; 25:2413. [PMID: 38397090 PMCID: PMC10888671 DOI: 10.3390/ijms25042413] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Revised: 02/09/2024] [Accepted: 02/15/2024] [Indexed: 02/25/2024] Open
Abstract
Volatile anesthetics have been shown in different studies to reduce ischemia reperfusion injury (IRI). Ex vivo lung perfusion (EVLP) facilitates graft evaluation, extends preservation time and potentially enables injury repair and improvement of lung quality. We hypothesized that ventilating lungs with sevoflurane during EVLP would reduce lung injury and improve lung function. We performed a pilot study to test this hypothesis in a slaughterhouse sheep DCD model. Lungs were harvested, flushed and stored on ice for 3 h, after which EVLP was performed for 4 h. Lungs were ventilated with either an FiO2 of 0.4 (EVLP, n = 5) or FiO2 of 0.4 plus sevoflurane at a 2% end-tidal concentration (Cet) (S-EVLP, n = 5). Perfusate, tissue samples and functional measurements were collected and analyzed. A steady state of the target Cet sevoflurane was reached with measurable concentrations in perfusate. Lungs in the S-EVLP group showed significantly better dynamic lung compliance than those in the EVLP group (p = 0.003). Oxygenation capacity was not different in treated lungs for delta partial oxygen pressure (PO2; +3.8 (-4.9/11.1) vs. -11.7 (-12.0/-3.2) kPa, p = 0.151), but there was a trend of a better PO2/FiO2 ratio (p = 0.054). Perfusate ASAT levels in S-EVLP were significantly reduced compared to the control group (198.1 ± 93.66 vs. 223.9 ± 105.7 IU/L, p = 0.02). We conclude that ventilating lungs with sevoflurane during EVLP is feasible and could be useful to improve graft function.
Collapse
Affiliation(s)
- Timo Steinkühler
- Department of Anesthesiology, University Medical Center Groningen, 9713 GZ Groningen, The Netherlands
| | - Shuqi Yang
- Department of Anesthesiology, University Medical Center Groningen, 9713 GZ Groningen, The Netherlands
| | - Michiel A. Hu
- Department of Thoracic Surgery, University Medical Center Groningen, 9713 GZ Groningen, The Netherlands
| | - Jayant S. Jainandunsing
- Department of Anesthesiology, University Medical Center Groningen, 9713 GZ Groningen, The Netherlands
| | - Neeltina M. Jager
- Department of Anesthesiology, University Medical Center Groningen, 9713 GZ Groningen, The Netherlands
| | - Michiel E. Erasmus
- Department of Thoracic Surgery, University Medical Center Groningen, 9713 GZ Groningen, The Netherlands
| | - Michel M. R. F. Struys
- Department of Anesthesiology, University Medical Center Groningen, 9713 GZ Groningen, The Netherlands
- Department of Basic and Applied Medical Sciences, Ghent University, 9000 Ghent, Belgium
| | - Dirk J. Bosch
- Department of Anesthesiology, University Medical Center Groningen, 9713 GZ Groningen, The Netherlands
| | - Matijs van Meurs
- Department of Critical Care, University Medical Center Groningen, 9713 GZ Groningen, The Netherlands
| | - Matthieu Jabaudon
- Department of Perioperative Medicine, University Hospital Clermont-Ferrand, 63001 Clermont-Ferrand, France
- Institute of Genetics, Reproduction & Development, University Clermont Auvergne, 63001 Clermont-Ferrand, France
- National Institute of Health and Medical Research (INSERM), National Center for Scientific Research (CNRS), 75794 Paris, France
| | - Damien Richard
- Department of Pharmacology and Toxicology, University Hospital Clermont-Ferrand, University Clermont Auvergne, 63001 Clermont-Ferrand, France
| | - Wim Timens
- Department of Pathology and Medical Biology, University Medical Center Groningen, 9713 GZ Groningen, The Netherlands
| | - Henri G. D. Leuvenink
- Department of Surgery, University Medical Center Groningen, 9713 GZ Groningen, The Netherlands
| | | |
Collapse
|
19
|
Yu H, Wang J, Liu M, Hu C, Sun J, Xu B, Lu S, Huang D, Pang Q, Hu C. Metformin alleviates lung ischemia-reperfusion injury in a rat lung transplantation model. Exp Lung Res 2024; 50:15-24. [PMID: 38317565 DOI: 10.1080/01902148.2023.2301615] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2023] [Accepted: 12/28/2023] [Indexed: 02/07/2024]
Abstract
Background: Lung ischemia-reperfusion injury (LIRI) is among the complications observed after lung transplantation and is associated with morbidity and mortality. Preconditioning of the donor lung before organ retrieval may improve organ quality after transplantation. We investigated whether preconditioning with metformin (Met) ameliorates LIRI after lung transplantation. Methods: Twenty Lewis rats were randomly divided into the sham, LIRI, and Met groups. The rats in the LIRI and Met groups received saline and Met, respectively, via oral gavage. Subsequently, a donor lung was harvested and kept in cold storage for 8 h. The LIRI and Met groups then underwent left lung transplantation. After 2 h of reperfusion, serum and transplanted lung tissues were examined. Results: The partial pressure of oxygen (PaO2) was greater in the Met group than in the LIRI group. In the Met group, wet-to-dry (W/D) weight ratios, inflammatory factor levels, oxidative stress levels and apoptosis levels were notably decreased. Conclusions: Met protects against ischemia-reperfusion injury after lung transplantation in rats, and its therapeutic effect is associated with its anti-inflammatory, antioxidative, and antiapoptotic properties.
Collapse
Affiliation(s)
- Huizhi Yu
- Department of Anesthesiology, Wuxi People's Hospital Affiliated to Nanjing Medical University, Wuxi, Jiangsu, China
- Department of Anesthesiology, The First Affiliated Hospital of Ningbo University, Ningbo, Zhejiang, China
| | - Jing Wang
- Department of Anesthesiology, Wuxi People's Hospital Affiliated to Nanjing Medical University, Wuxi, Jiangsu, China
| | - Mingzhao Liu
- Department of Lung Transplant Center, Wuxi People's Hospital Affiliated to Nanjing Medical University, Wuxi, Jiangsu, China
| | - Chunlan Hu
- Department of Anesthesiology, Wuxi People's Hospital Affiliated to Nanjing Medical University, Wuxi, Jiangsu, China
| | - Jiaojiao Sun
- Department of Physiopathology, Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu, China
| | - Bo Xu
- Department of Anesthesiology, Wuxi People's Hospital Affiliated to Nanjing Medical University, Wuxi, Jiangsu, China
| | - Shunmei Lu
- Department of Anesthesiology, Wuxi People's Hospital Affiliated to Nanjing Medical University, Wuxi, Jiangsu, China
| | - Dongxiao Huang
- Department of Anesthesiology, Wuxi People's Hospital Affiliated to Nanjing Medical University, Wuxi, Jiangsu, China
| | - Qingfeng Pang
- Department of Physiopathology, Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu, China
| | - Chunxiao Hu
- Department of Transplant Anesthesiology, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi, Jiangsu, China
| |
Collapse
|
20
|
Zhan Y, Li L, Guo C, Zhang Y, Zhao L, Tao Z, Zhang H, Chen S. MicroRNA-141-3p reduces pulmonary hypoxia/reoxygenation injury through suppression of Beclin-1-dependent autophagy. Aging (Albany NY) 2024; 16:1352-1373. [PMID: 38261732 PMCID: PMC10866419 DOI: 10.18632/aging.205430] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2023] [Accepted: 11/06/2023] [Indexed: 01/25/2024]
Abstract
Alterations in autophagy are involved in pulmonary hypoxia/reoxygenation (H/R)-induced injury. Here, we intended to explain the function of microRNA-141-3p (miR-141-3p) in regulating autophagy under the H/R condition. Rat pulmonary microvascular endothelial cells (PMVECs) were applied for H/R cell model establishment, followed by tracing of autophagy formation. SIRT1 plays a critical role in controlling the lifespan of yeast, flies, and mice. Interaction between SIRT1 and Beclin-1, an indicator protein for autophagy, and between miR-141-3p and SIRT1 was assayed with their roles in PMVEC injury. Autophagy of PMVECs was activated after hypoxia treatment and further activated after H/R treatment. The binding of miR-141-3p and SIRT1 was verified. In H/R-treated PMVECs, the binding of miR-141-3p and SIRT1 was reduced. Furthermore, SIRT1 acted as a deacetylase to stabilize the Beclin-1 protein, promoting autophagy and PMVEC injury. H/R rat models were established, and in vivo, experiments further confirmed that miR-141-3p regulated autophagy and lung injury in H/R rats through SIRT1/Beclin-1 axis. The current study highlighted that reduced miR-141-3p in H/R-treated PMVECs promoted deacetylation of Beclin-1 by SIRT1, thus causing PMVEC injury.
Collapse
Affiliation(s)
- Yanping Zhan
- Department of Anesthesiology, The First Affiliated Hospital of Nanchang University, Nanchang 330006, P.R. China
| | - Lei Li
- Department of Anesthesiology, The First Affiliated Hospital of Nanchang University, Nanchang 330006, P.R. China
| | - Chen Guo
- Jiangxi Maternal and Child Health Hospital, Nanchang 330006, P.R. China
| | - Yang Zhang
- Department of Anesthesiology, The First Affiliated Hospital of Nanchang University, Nanchang 330006, P.R. China
| | - Lili Zhao
- Department of Anesthesiology, The First Affiliated Hospital of Nanchang University, Nanchang 330006, P.R. China
| | - Zhe Tao
- Department of Anesthesiology, The First Affiliated Hospital of Nanchang University, Nanchang 330006, P.R. China
| | - Hua Zhang
- Nanchang University, Nanchang 330006, P.R. China
| | - Shibiao Chen
- Department of Anesthesiology, The First Affiliated Hospital of Nanchang University, Nanchang 330006, P.R. China
| |
Collapse
|
21
|
Zhao S, Hu Y, Yang B, Zhang L, Xu M, Jiang K, Liu Z, Wu M, Huang Y, Li P, Liang SJ, Sun X, Hide G, Lun ZR, Wu Z, Shen J. The transplant rejection response involves neutrophil and macrophage adhesion-mediated trogocytosis and is regulated by NFATc3. Cell Death Dis 2024; 15:75. [PMID: 38242872 PMCID: PMC10798984 DOI: 10.1038/s41419-024-06457-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2023] [Revised: 01/04/2024] [Accepted: 01/08/2024] [Indexed: 01/21/2024]
Abstract
The anti-foreign tissue (transplant rejection) response, mediated by the immune system, has been the biggest obstacle to successful organ transplantation. There are still many enigmas regarding this process and some aspects of the underlying mechanisms driving the immune response against foreign tissues remain poorly understood. Here, we found that a large number of neutrophils and macrophages were attached to the graft during skin transplantation. Furthermore, both types of cells could autonomously adhere to and damage neonatal rat cardiomyocyte mass (NRCM) in vitro. We have demonstrated that Complement C3 and the receptor CR3 participated in neutrophils/macrophages-mediated adhesion and damage this foreign tissue (NRCM or skin grafts). We have provided direct evidence that the damage to these tissues occurs by a process referred to as trogocytosis, a damage mode that has never previously been reported to directly destroy grafts. We further demonstrated that this process can be regulated by NFAT, in particular, NFATc3. This study not only enriches an understanding of host-donor interaction in transplant rejection, but also provides new avenues for exploring the development of novel immunosuppressive drugs which prevent rejection during transplant therapy.
Collapse
Affiliation(s)
- Siyu Zhao
- Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, 510080, Guangdong, China
- Key Laboratory of Tropical Disease Control (Sun Yat-Sen University), Ministry of Education, Guangzhou, 510080, Guangdong, China
- Provincial Engineering Technology Research Center for Biological Vector Control, Guangzhou, 510080, Guangdong, China
| | - Yunyi Hu
- Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, 510080, Guangdong, China
- Key Laboratory of Tropical Disease Control (Sun Yat-Sen University), Ministry of Education, Guangzhou, 510080, Guangdong, China
- Provincial Engineering Technology Research Center for Biological Vector Control, Guangzhou, 510080, Guangdong, China
| | - Bicheng Yang
- The Andrology Department, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, China
| | - Lichao Zhang
- Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, 510080, Guangdong, China
- Key Laboratory of Tropical Disease Control (Sun Yat-Sen University), Ministry of Education, Guangzhou, 510080, Guangdong, China
- Provincial Engineering Technology Research Center for Biological Vector Control, Guangzhou, 510080, Guangdong, China
| | - Meiyining Xu
- Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, 510080, Guangdong, China
- Key Laboratory of Tropical Disease Control (Sun Yat-Sen University), Ministry of Education, Guangzhou, 510080, Guangdong, China
- Provincial Engineering Technology Research Center for Biological Vector Control, Guangzhou, 510080, Guangdong, China
| | - Kefeng Jiang
- Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, 510080, Guangdong, China
- Key Laboratory of Tropical Disease Control (Sun Yat-Sen University), Ministry of Education, Guangzhou, 510080, Guangdong, China
- Provincial Engineering Technology Research Center for Biological Vector Control, Guangzhou, 510080, Guangdong, China
| | - Zhun Liu
- Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, 510080, Guangdong, China
- Key Laboratory for Stem Cells and Tissue Engineering (Sun Yat-Sen University), Ministry of Education, Guangzhou, 510080, Guangdong, China
| | - Mingrou Wu
- Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, 510080, Guangdong, China
- Key Laboratory of Tropical Disease Control (Sun Yat-Sen University), Ministry of Education, Guangzhou, 510080, Guangdong, China
- Provincial Engineering Technology Research Center for Biological Vector Control, Guangzhou, 510080, Guangdong, China
| | - Yun Huang
- Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, 510080, Guangdong, China
- Key Laboratory of Tropical Disease Control (Sun Yat-Sen University), Ministry of Education, Guangzhou, 510080, Guangdong, China
- Provincial Engineering Technology Research Center for Biological Vector Control, Guangzhou, 510080, Guangdong, China
| | - Peipei Li
- Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, 510080, Guangdong, China
- Key Laboratory of Tropical Disease Control (Sun Yat-Sen University), Ministry of Education, Guangzhou, 510080, Guangdong, China
- Provincial Engineering Technology Research Center for Biological Vector Control, Guangzhou, 510080, Guangdong, China
| | - Si-Jia Liang
- Department of Pharmacology, Cardiac and Cerebral Vascular Research Center, Sun Yat-sen University, 74 Zhongshan 2 Rd, Guangzhou, 510080, China
| | - Xi Sun
- Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, 510080, Guangdong, China
- Key Laboratory of Tropical Disease Control (Sun Yat-Sen University), Ministry of Education, Guangzhou, 510080, Guangdong, China
- Provincial Engineering Technology Research Center for Biological Vector Control, Guangzhou, 510080, Guangdong, China
| | - Geoff Hide
- Biomedical Research and Innovation Centre, School of Science, Engineering and Environment, University of Salford, Salford, M5 4WT, UK
| | - Zhao-Rong Lun
- Biomedical Research and Innovation Centre, School of Science, Engineering and Environment, University of Salford, Salford, M5 4WT, UK
- State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-Sen University, Guangzhou, 510275, China
| | - Zhongdao Wu
- Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, 510080, Guangdong, China
- Key Laboratory of Tropical Disease Control (Sun Yat-Sen University), Ministry of Education, Guangzhou, 510080, Guangdong, China
- Provincial Engineering Technology Research Center for Biological Vector Control, Guangzhou, 510080, Guangdong, China
| | - Jia Shen
- Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, 510080, Guangdong, China.
- Key Laboratory of Tropical Disease Control (Sun Yat-Sen University), Ministry of Education, Guangzhou, 510080, Guangdong, China.
- Provincial Engineering Technology Research Center for Biological Vector Control, Guangzhou, 510080, Guangdong, China.
| |
Collapse
|
22
|
Bejeshk MA, Najafipour H, Khaksari M, Nematollahi MH, Rajizadeh MA, Dabiri S, Beik A, Samareh-Fekri M, Sepehri G. Preparation and Evaluation of Preventive Effects of Inhalational and Intraperitoneal Injection of Myrtenol Loaded Nano-Niosomes on Lung Ischemia-Reperfusion Injury in Rats. J Pharm Sci 2024; 113:85-94. [PMID: 37931787 DOI: 10.1016/j.xphs.2023.11.002] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Revised: 11/01/2023] [Accepted: 11/01/2023] [Indexed: 11/08/2023]
Abstract
INTRODUCTION Ischemia-reperfusion injury (IRI) is directly related to forming reactive oxygen species, endothelial cell injury, increased vascular permeability, and the activation of neutrophils and cytokines. Niosomes are nanocarriers and an essential part of drug delivery systems. We aimed to investigate the effects of myrtenol's inhaled and intraperitoneal niosomal form, compared to its simple form, on lung ischemia reperfusion injury (LIRI). MATERIAL AND METHOD Wistar rats were divided into ten groups. Simple and niosomal forms of myrtenol were inhaled or intraperitoneally injected daily for one week prior to LIRI. We evaluated oxidative stress, apoptotic, and inflammatory indices, nitric oxide, inducible nitric oxide synthase (iNOS), endothelial nitric oxide synthase (eNOS) and histopathological indices. RESULTS Pretreatment with simple and niosomal forms of myrtenol significantly inhibited the indices of pulmonary edema, pro-inflammatory cytokines and proteins, oxidant agents, nitric oxide, iNOS, apoptotic proteins, congestion of capillaries, neutrophil infiltration, and bleeding in the alveoli. Furthermore, myrtenol increased anti-inflammatory cytokines, anti-oxidants agents, eNOS, anti-apoptotic proteins and the survival time of animals. The niosomal form of myrtenol showed a more ameliorative effect than its simple form. CONCLUSION The results showed the superior protective effect of the inhalation of myrtenol niosomal form against LIRI compared to its simple form and systemic use.
Collapse
Affiliation(s)
- Mohammad Abbas Bejeshk
- Department of Physiology and Pharmacology, Afzalipour Medical Faculty, Kerman university of Medical Sciences, Kerman, Iran; Physiology Research Center, Institute of Neuropharmacology, Kerman University of Medical Sciences, Kerman, Iran
| | - Hamid Najafipour
- Department of Physiology and Pharmacology, Afzalipour Medical Faculty, Kerman university of Medical Sciences, Kerman, Iran; Cardiovascular Research Center, Institute of Basic and Clinical Physiology Sciences, Kerman University of Medical Sciences, Kerman, Iran
| | - Mohammad Khaksari
- Department of Physiology and Pharmacology, Afzalipour Medical Faculty, Kerman university of Medical Sciences, Kerman, Iran; Endocrinology and Metabolism Research Center, Institute of Basic and Clinical Physiology Sciences, Kerman University of Medical Sciences, Kerman, Iran
| | | | - Mohammad Amin Rajizadeh
- Department of Physiology and Pharmacology, Afzalipour Medical Faculty, Kerman university of Medical Sciences, Kerman, Iran; Physiology Research Center, Institute of Neuropharmacology, Kerman University of Medical Sciences, Kerman, Iran
| | - Shahriar Dabiri
- Pathology and Stem Cells Research Center, Department of Pathology, School of Medicine, Kerman University of Medical Science, Kerman, Iran
| | - Ahmad Beik
- Department of Physiology and Pharmacology, Afzalipour Medical Faculty, Kerman university of Medical Sciences, Kerman, Iran; Cardiovascular Research Center, Institute of Basic and Clinical Physiology Sciences, Kerman University of Medical Sciences, Kerman, Iran
| | - Mitra Samareh-Fekri
- Physiology Research Center, Institute of Neuropharmacology, Kerman University of Medical Sciences, Kerman, Iran
| | - Gholamreza Sepehri
- Department of Physiology and Pharmacology, Afzalipour Medical Faculty, Kerman university of Medical Sciences, Kerman, Iran; Neuroscience Research Center, Kerman University of Medical Sciences, Kerman, Iran.
| |
Collapse
|
23
|
Ren Z, Zheng Z, Feng X. Role of gut microbes in acute lung injury/acute respiratory distress syndrome. Gut Microbes 2024; 16:2440125. [PMID: 39658851 PMCID: PMC11639474 DOI: 10.1080/19490976.2024.2440125] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/04/2024] [Revised: 10/31/2024] [Accepted: 12/04/2024] [Indexed: 12/12/2024] Open
Abstract
Acute lung injury (ALI) is an acute, diffuse inflammatory lung condition triggered by factors of severe infections, trauma, shock, burns, ischemia-reperfusion, and mechanical ventilation. It is primarily characterized by refractory hypoxemia and respiratory distress. The more severe form, acute respiratory distress syndrome (ARDS), can progress to multi-organ failure and has a high mortality rate. Despite extensive research, the exact pathogenesis of ALI and ARDS remains complex and not fully understood. Recent advancements in studying the gut microecology of patients have revealed the critical role that gut microbes play in ALI/ARDS onset and progression. While the exact mechanisms are still under investigation, evidence increasingly points to the influence of gut microbes and their metabolites on ALI/ARDS. This review aims to summarize the role of gut microbes and their metabolites in ALI/ARDS caused by various triggers. Moreover, it explores potential mechanisms and discusses how gut microbe-targeting interventions might offer new clinical strategies for the treatment of ALI/ARDS.
Collapse
Affiliation(s)
- Zixuan Ren
- Shandong Provincial Key Laboratory for Rheumatic Disease and Translational Medicine, The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan Hospital, Jinan, Shandong, China
- School of Clinical and Basic Medical Sciences, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Zhihuan Zheng
- Shandong Provincial Key Laboratory for Rheumatic Disease and Translational Medicine, The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan Hospital, Jinan, Shandong, China
- School of Clinical and Basic Medical Sciences, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Xiujing Feng
- Shandong Provincial Key Laboratory for Rheumatic Disease and Translational Medicine, The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan Hospital, Jinan, Shandong, China
- School of Clinical and Basic Medical Sciences, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, China
- Key Laboratory of Endocrine Glucose & Lipids Metabolism and Brain Aging, Ministry of Education, Department of Endocrinology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| |
Collapse
|
24
|
Zhang Z, Li X, Guo J, He B, Wu L, Yang R, Li X, Fang D, Yang X, Yang D, Wang F, Tang M, Han Y, Jose PA, Wang H, Zeng C. β-aminoisobutyrics acid, a metabolite of BCAA, activates the AMPK/Nrf-2 pathway to prevent ferroptosis and ameliorates lung ischemia-reperfusion injury. Mol Med 2023; 29:164. [PMID: 38049750 PMCID: PMC10696792 DOI: 10.1186/s10020-023-00729-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2023] [Accepted: 09/17/2023] [Indexed: 12/06/2023] Open
Abstract
BACKGROUND Lung ischemia-reperfusion (I/R) injury is a serious clinical problem without effective treatment. Enhancing branched-chain amino acids (BCAA) metabolism can protect against cardiac I/R injury, which may be related to bioactive molecules generated by BCAA metabolites. L-β-aminoisobutyric acid (L-BAIBA), a metabolite of BCAA, has multi-organ protective effects, but whether it protects against lung I/R injury is unclear. METHODS To assess the protective effect of L-BAIBA against lung I/R injury, an animal model was generated by clamping the hilum of the left lung, followed by releasing the clamp in C57BL/6 mice. Mice with lung I/R injury were pre-treated or post-treated with L-BAIBA (150 mg/kg/day), given by gavage or intraperitoneal injection. Lung injury was assessed by measuring lung edema and analyzing blood gases. Inflammation was assessed by measuring proinflammatory cytokines in bronchoalveolar lavage fluid (BALF), and neutrophil infiltration of the lung was measured by myeloperoxidase activity. Molecular biological methods, including western blot and immunofluorescence, were used to detect potential signaling mechanisms in A549 and BEAS-2B cells. RESULTS We found that L-BAIBA can protect the lung from I/R injury by inhibiting ferroptosis, which depends on the up-regulation of the expressions of GPX4 and SLC7A11 in C57BL/6 mice. Additionally, we demonstrated that the Nrf-2 signaling pathway is key to the inhibitory effect of L-BAIBA on ferroptosis in A549 and BEAS-2B cells. L-BAIBA can induce the nuclear translocation of Nrf-2. Interfering with the expression of Nrf-2 eliminated the protective effect of L-BAIBA on ferroptosis. A screening of potential signaling pathways revealed that L-BAIBA can increase the phosphorylation of AMPK, and compound C can block the Nrf-2 nuclear translocation induced by L-BAIBA. The presence of compound C also blocked the protective effects of L-BAIBA on lung I/R injury in C57BL/6 mice. CONCLUSIONS Our study showed that L-BAIBA protects against lung I/R injury via the AMPK/Nrf-2 signaling pathway, which could be a therapeutic target.
Collapse
Affiliation(s)
- Ziyue Zhang
- Department of Cardiology, Daping Hospital, The Third Military Medical University (Army Medical University), Chongqing, P. R. China
- Key Laboratory of Geriatric Cardiovascular and Cerebrovascular Disease Research, Chongqing Key Laboratory for Hypertension Research, Chongqing Cardiovascular Clinical Research Center, Ministry of Education of China, Chongqing Institute of Cardiology, Chongqing, P. R. China
- Outpatient Department, Hospital of PLA, Hanzhong, Shanxi, 96608, P. R. China
| | - Xingbing Li
- Department of Cardiology, Daping Hospital, The Third Military Medical University (Army Medical University), Chongqing, P. R. China
- Key Laboratory of Geriatric Cardiovascular and Cerebrovascular Disease Research, Chongqing Key Laboratory for Hypertension Research, Chongqing Cardiovascular Clinical Research Center, Ministry of Education of China, Chongqing Institute of Cardiology, Chongqing, P. R. China
- Department of Cardiology, Chongqing Hospital of Traditional Chinese Medicine, Chongqing, P. R. China
| | - Jingwen Guo
- Department of Cardiology, Daping Hospital, The Third Military Medical University (Army Medical University), Chongqing, P. R. China
- Key Laboratory of Geriatric Cardiovascular and Cerebrovascular Disease Research, Chongqing Key Laboratory for Hypertension Research, Chongqing Cardiovascular Clinical Research Center, Ministry of Education of China, Chongqing Institute of Cardiology, Chongqing, P. R. China
| | - Bo He
- Department of Cardiology, Daping Hospital, The Third Military Medical University (Army Medical University), Chongqing, P. R. China
- Key Laboratory of Geriatric Cardiovascular and Cerebrovascular Disease Research, Chongqing Key Laboratory for Hypertension Research, Chongqing Cardiovascular Clinical Research Center, Ministry of Education of China, Chongqing Institute of Cardiology, Chongqing, P. R. China
| | - Lianpan Wu
- Department of Cardiology, Daping Hospital, The Third Military Medical University (Army Medical University), Chongqing, P. R. China
- Key Laboratory of Geriatric Cardiovascular and Cerebrovascular Disease Research, Chongqing Key Laboratory for Hypertension Research, Chongqing Cardiovascular Clinical Research Center, Ministry of Education of China, Chongqing Institute of Cardiology, Chongqing, P. R. China
| | - Rongpei Yang
- Department of Cardiology, Daping Hospital, The Third Military Medical University (Army Medical University), Chongqing, P. R. China
- Key Laboratory of Geriatric Cardiovascular and Cerebrovascular Disease Research, Chongqing Key Laboratory for Hypertension Research, Chongqing Cardiovascular Clinical Research Center, Ministry of Education of China, Chongqing Institute of Cardiology, Chongqing, P. R. China
| | - Xingyue Li
- Key Laboratory of Geriatric Cardiovascular and Cerebrovascular Disease Research, Chongqing Key Laboratory for Hypertension Research, Chongqing Cardiovascular Clinical Research Center, Ministry of Education of China, Chongqing Institute of Cardiology, Chongqing, P. R. China
| | - Dandong Fang
- Department of Cardiology, Daping Hospital, The Third Military Medical University (Army Medical University), Chongqing, P. R. China
- Key Laboratory of Geriatric Cardiovascular and Cerebrovascular Disease Research, Chongqing Key Laboratory for Hypertension Research, Chongqing Cardiovascular Clinical Research Center, Ministry of Education of China, Chongqing Institute of Cardiology, Chongqing, P. R. China
| | - XiaoLi Yang
- Department of Cardiology, Daping Hospital, The Third Military Medical University (Army Medical University), Chongqing, P. R. China
- Key Laboratory of Geriatric Cardiovascular and Cerebrovascular Disease Research, Chongqing Key Laboratory for Hypertension Research, Chongqing Cardiovascular Clinical Research Center, Ministry of Education of China, Chongqing Institute of Cardiology, Chongqing, P. R. China
| | - Donghai Yang
- Department of Cardiology, Daping Hospital, The Third Military Medical University (Army Medical University), Chongqing, P. R. China
- Key Laboratory of Geriatric Cardiovascular and Cerebrovascular Disease Research, Chongqing Key Laboratory for Hypertension Research, Chongqing Cardiovascular Clinical Research Center, Ministry of Education of China, Chongqing Institute of Cardiology, Chongqing, P. R. China
| | - Fengxian Wang
- Department of Cardiology, Daping Hospital, The Third Military Medical University (Army Medical University), Chongqing, P. R. China
- Key Laboratory of Geriatric Cardiovascular and Cerebrovascular Disease Research, Chongqing Key Laboratory for Hypertension Research, Chongqing Cardiovascular Clinical Research Center, Ministry of Education of China, Chongqing Institute of Cardiology, Chongqing, P. R. China
| | - Ming Tang
- Department of Cardiology, Daping Hospital, The Third Military Medical University (Army Medical University), Chongqing, P. R. China
- Key Laboratory of Geriatric Cardiovascular and Cerebrovascular Disease Research, Chongqing Key Laboratory for Hypertension Research, Chongqing Cardiovascular Clinical Research Center, Ministry of Education of China, Chongqing Institute of Cardiology, Chongqing, P. R. China
| | - Yu Han
- Department of Cardiology, Daping Hospital, The Third Military Medical University (Army Medical University), Chongqing, P. R. China
- Key Laboratory of Geriatric Cardiovascular and Cerebrovascular Disease Research, Chongqing Key Laboratory for Hypertension Research, Chongqing Cardiovascular Clinical Research Center, Ministry of Education of China, Chongqing Institute of Cardiology, Chongqing, P. R. China
| | - Pedro A Jose
- Division of Renal Diseases & Hypertension, Department of Medicine, Department of Physiology/Pharmacology, The George Washington University School of Medicine & Health Sciences, Washington, DC, USA
| | - Hongyong Wang
- Department of Cardiology, Daping Hospital, The Third Military Medical University (Army Medical University), Chongqing, P. R. China.
- Key Laboratory of Geriatric Cardiovascular and Cerebrovascular Disease Research, Chongqing Key Laboratory for Hypertension Research, Chongqing Cardiovascular Clinical Research Center, Ministry of Education of China, Chongqing Institute of Cardiology, Chongqing, P. R. China.
| | - Chunyu Zeng
- Department of Cardiology, Daping Hospital, The Third Military Medical University (Army Medical University), Chongqing, P. R. China.
- Key Laboratory of Geriatric Cardiovascular and Cerebrovascular Disease Research, Chongqing Key Laboratory for Hypertension Research, Chongqing Cardiovascular Clinical Research Center, Ministry of Education of China, Chongqing Institute of Cardiology, Chongqing, P. R. China.
- State Key Laboratory of Trauma, Burns and Combined Injury, Daping Hospital, The Third Military Medical University (Army Medical University), Chongqing, P. R. China.
- Cardiovascular Research Center of Chongqing College, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Chongqing, P. R. China.
| |
Collapse
|
25
|
Qian J, Xu Z, Yin M, Qin Z, Pinhu L. Bioinformatics analyses of immune-related genes and immune infiltration associated with lung ischemia-reperfusion injury. Transpl Immunol 2023; 81:101926. [PMID: 37652362 DOI: 10.1016/j.trim.2023.101926] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2023] [Revised: 08/21/2023] [Accepted: 08/27/2023] [Indexed: 09/02/2023]
Abstract
BACKGROUND Ischemia-reperfusion injury (IRI) is a significant complication that can occur following lung transplantation and is known to contribute to poor prognosis. Our research aimed to investigate the potential molecular targets and mechanisms involved in lung IRI (LIRI), in order to improve our understanding of this condition. METHOD We downloaded gene expression datasets (GSE127003 and GSE18995) linked to LIRI from the GEO database. Using WGCNA, we identified LIRI-related modules. Functional enrichment analyses were performed on the modules showing significant correlation with LIRI. Core immune-related genes (IRGs) were identified and validated using the GSE18995 dataset. A rat LIRI model was established to validate the expression changes of core IRGs. The LIRI groups were subjected to 60 min of warm ischemia followed by 120 min of reperfusion. Additionally, the xCell algorithm was used to characterize the immune landscape and analyze the relationships between hub IRGs and infiltrating immune cells. RESULTS A total of 483 genes from the turquoise module were identified through WGCNA, with a predominant enrichment in immune- and inflammation-related pathways. Three IRGs (PTGS2, CCL2, and RELB) were found to be up-regulated after reperfusion in both GSE127003 and GSE18995 datasets, and this was further confirmed using the rat LIRI model. The xCell analysis revealed that immune score, CD8+ naive T cells, eosinophils, neutrophils, NK cells, and Tregs were upregulated after reperfusion. PTGS2, CCL2, and RELB showed positive correlations with CD8+ naive T cells, monocytes, neutrophils, and Tregs. CONCLUSION PTGS2, CCL2, and RELB were found to be potential biomarkers for LIRI. Immune and microenvironment scores were higher after reperfusion compared to before reperfusion. PTGS2, CCL2, and RELB appear to play a crucial role in the development of LIRI and may contribute to it by increasing the number of immune cells. Our findings offer new perspectives on potential treatment targets and the pathogenesis of LIRI.
Collapse
Affiliation(s)
- Jing Qian
- Department of Cardiothoracic Intensive Care Medicine, The First Affiliated Hospital of Guangxi Medical University, Nanning 530021, China
| | - Zhanyu Xu
- Department of Thoracic Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning 530021, China
| | - Mingjing Yin
- Department of Emergency, The First Affiliated Hospital of Guangxi Medical University, Nanning 530021, China
| | - Zhidan Qin
- Department of Critical Care Medicine, The First Affiliated Hospital of Guangxi Medical University, Nanning 530021, China
| | - Liao Pinhu
- Department of Emergency, The First Affiliated Hospital of Guangxi Medical University, Nanning 530021, China.
| |
Collapse
|
26
|
Wu C, Xu J, Zhang Z, Wei D, Xu Y, Zhao Y. The Effects of IL-23/IL-18-Polarized Neutrophils on Renal Ischemia-Reperfusion Injury and Allogeneic-Skin-Graft Rejection in Mice. Biomedicines 2023; 11:3148. [PMID: 38137369 PMCID: PMC10740676 DOI: 10.3390/biomedicines11123148] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2023] [Revised: 10/26/2023] [Accepted: 11/20/2023] [Indexed: 12/24/2023] Open
Abstract
Neutrophils display heterogeneity and plasticity with different subgroups and immune-regulatory functions under various surrounding conditions. Neutrophils induced by IL-23/IL-18 (referred to N(IL-23+IL-18) neutrophils) have a unique gene-expression profile, with highly expressing IL-17, MHC-II, and costimulatory molecules. The adoptive transfer of N(IL-23+IL-18) neutrophils significantly increased the pathogenesis in a renal ischemia-reperfusion injury mouse model. N(IL-23+IL-18) neutrophils directly and efficiently induced allogeneic T cell proliferation in vitro. N(IL-23+IL-18) neutrophils enhanced the syngeneic T cell response to allogeneic antigens in mixed-lymphocyte reaction assays. The adoptive transfer of the donor or host N(IL-23+IL-18) neutrophils significantly enhanced the antidonor antibody production in an allogeneic-skin-transplanted mouse model, accompanied by increased Tfh cells in the spleens. Therefore, the neutrophil subset induced by IL-23/IL-18 promotes tissue injury and antidonor humoral response in the allogeneic transplantation mouse model.
Collapse
Affiliation(s)
- Changhong Wu
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100045, China; (C.W.); (J.X.); (Y.X.)
- University of Chinese Academy of Sciences, Beijing 101408, China
| | - Jinglin Xu
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100045, China; (C.W.); (J.X.); (Y.X.)
- University of Chinese Academy of Sciences, Beijing 101408, China
| | - Zhaoqi Zhang
- CAS Key Laboratory of Quantitative Engineering Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China; (Z.Z.); (D.W.)
| | - Dong Wei
- CAS Key Laboratory of Quantitative Engineering Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China; (Z.Z.); (D.W.)
| | - Yanan Xu
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100045, China; (C.W.); (J.X.); (Y.X.)
| | - Yong Zhao
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100045, China; (C.W.); (J.X.); (Y.X.)
- CAS Key Laboratory of Quantitative Engineering Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China; (Z.Z.); (D.W.)
- Faculty of Synthetic Biology, Shenzhen Institute of Advanced Technology, Shenzhen 518055, China
| |
Collapse
|
27
|
Beyoglu MA, Sahin MF, Turkkan S, Yazicioglu A, Akbulut ED, Neselioglu S, Erel O, Yekeler E. Dynamic Thiol-Disulfide Homeostasis in Lung Transplant Recipients. EXP CLIN TRANSPLANT 2023; 21:841-847. [PMID: 34981712 DOI: 10.6002/ect.2021.0360] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
OBJECTIVES In this study, we investigated dynamic thiol-disulfide homeostasis as a new indicator of oxidative stress in lung transplant recipients. In addition, we compared dynamic thiol-disulfide homeostasis parameters according to transplant indication and time after transplant. MATERIALS AND METHODS This study had a single-center, observational, randomized design. In terms of transplant indications, lung transplant recipients were grouped as chronic obstructive pulmonary disease, interstitial lung disease, bronchiectasis, and other indications. To make comparisons based on time after transplant, lung transplant recipients were categorized into the following groups: >6 and ≤24 months, >24 and ≤48 months, >48 and ≤72 months, and >72 months. A fully automated spectrophotometric technique was used to measure dynamic thiol-disulfide homeostasis in fasting blood samples. RESULTS Our study included 34 lung transplant recipients and 36 healthy volunteers. Native thiol (P = .005) and total thiol levels (P = .06) were lower in lung transplant recipients. Disulfide levels were similar. Disulfide-to-native thiol (P = .027) and disulfide-to-total thiol ratios (P = .027) were significantly higher in lung transplant recipients. Native thiol-to-total thiol ratios were lower in lung transplant recipients (P = .027). When we examined patients according to transplant indication, no statistically significant differences were found in dynamic thiol-disulfide homeostasis parameters, except for total thiol and disulfide levels. We also found no significant differences when we examined dynamic thiol-disulfide homeostasis parameters according to time after transplant. CONCLUSIONS Thiol-related antioxidant activity is significantly reduced after lung transplant, regardless of indication and transplant time. Ensuring oxidative balance in lung transplant recipients with an antioxidant supplement regimen can prevent damage from oxidative stress.
Collapse
Affiliation(s)
- Muhammet Ali Beyoglu
- From the Department of General Thoracic Surgery and Lung Transplantation, Ankara City Hospital, University of Health Sciences, Ankara, Turkey
| | | | | | | | | | | | | | | |
Collapse
|
28
|
Al-Subu AM, Long MT, Nelson KL, Amond KL, Lasarev MR, Ferrazzano PA, Lushaj EB, Anagnostopoulos PV. Risk of Hypovitaminosis and Vitamin C Deficiency in Pediatric Patients Undergoing Cardiopulmonary Bypass. Pediatr Cardiol 2023; 44:1487-1494. [PMID: 37498330 DOI: 10.1007/s00246-023-03243-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/12/2023] [Accepted: 07/14/2023] [Indexed: 07/28/2023]
Abstract
Vitamin C levels are known rapidly decrease in adult critical illness. Vitamin C scavenges free radicals, provides critical protection of the endothelial barrier, and improves endothelial responsiveness to catecholamines. Children with congenital heart disease and undergoing cardiac surgery might be at increased risk for low circulating vitamin C levels. A prospective single-center observational study investigated perioperative changes in vitamin C levels in critically ill Children who underwent congenital heart surgery using CPB. Vitamin C serum levels were collected preoperatively and postoperatively (upon admission to the ICU, 24 and 72 h). Linear mixed-effect model was used to estimate mean circulating concentration of vitamin C and to estimate changes in concentration over time. Primary outcome was change in circulating levels of vitamin C before and after CPB. Secondary outcomes were hospital length of stay (LOS), acute kidney injury (AKI), and illness severity. Forty-one patients with a median age of 4.5 [interquartile range (IQR) 2.6-65.6] months at the time of surgery were consented and enrolled. Median CPB duration was 130 [90-175] minutes, and hospital LOS was 9.1 [5.2-19] days. Mean vitamin C levels (μmol/L) before CPB, at PICU admission, 24 h, and 72 h were 82.0 (95% CI 73.4-90.7), 53.4 (95% CI 44.6,62.0), 55.1 (95% CI 46.3,63.8), and 59.2 (95% CI 50.3,68.1), respectively. Upon postoperative admission to the PICU, vitamin C levels decreased by 28.7 (95% CI 20.6-36.8; p < 0.001) μmol/L, whereas levels at 24 and 72 h recovered and did not differ substantially from concentrations reported upon PICU admission (p > 0.15). Changes in vitamin C concentration were not associated with CPB time, STAT mortality category, age, or PIM3. Three patients had post-CPB hypovitaminosis C or vitamin C deficiency. Reduction in vitamin C levels was not associated with hospital LOS (p = 0.673). A 25 μmol/L decrease in vitamin C levels upon PICU admission was associated with developing AKI (aOR = 3.65; 95% CI 1.01-18.0, p = 0.049). Pediatric patients undergoing cardiac surgery with CPB showed decreased vitamin C levels during the immediate postoperative period. Effects of hypovitaminosis C and vitamin C deficiency in this population remain unclear.
Collapse
Affiliation(s)
- Awni M Al-Subu
- Division of Pediatric Critical Care Medicine, Department of Pediatrics, University of Wisconsin School of Medicine and Public Health, 600 Highland Ave Rm H6/535, Madison, WI, 53792, USA.
| | - Micah T Long
- Division of Critical Care, Department of Anesthesiology, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - Kari L Nelson
- Department of Surgery, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - Kate L Amond
- Department of Surgery, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - Michael R Lasarev
- Department of Biostatistics and Medical Informatics, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - Peter A Ferrazzano
- Division of Pediatric Critical Care Medicine, Department of Pediatrics, University of Wisconsin School of Medicine and Public Health, 600 Highland Ave Rm H6/535, Madison, WI, 53792, USA
| | - Entela B Lushaj
- Department of Surgery, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - Petros V Anagnostopoulos
- Department of Surgery, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| |
Collapse
|
29
|
Hu CY, Li GY, Li CT. Thiopental sodium attenuates hypoxia/reoxygenation-induced injury in osteoblasts by modulating AKT signaling. In Vitro Cell Dev Biol Anim 2023; 59:528-535. [PMID: 37556024 DOI: 10.1007/s11626-023-00801-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Accepted: 07/14/2023] [Indexed: 08/10/2023]
Abstract
Thiopental sodium (TPTS) is a barbiturate general anesthetic, while its effects on hypoxia/reoxygenation (H/R)-induced injury are still unclear. This study aimed to investigate whether TPTS exerts protective effects against the H/R-induced osteoblast cell injury and explore the underlying mechanisms. Osteoblast cell injury model was induced by the H/R condition, which was treated with or without TPTS. Cell viability and lactate dehydrogenase (LDH) release were determined by the corresponding commercial kits. The levels of oxidative stress were determined in the experimental groups. Cell apoptosis and Caspase-3 activities were determined by propidium iodide staining and substrate-based assay, respectively. Western blotting and qRT-PCR were performed to measure the mRNA and protein levels, respectively. Treatment with TPTS was able to increase cell viability and reduce LDH release in H/R-induced osteoblasts. Additionally, TPTS regulated oxidative stress in H/R-induced osteoblasts by suppressing malondialdehyde (MDA) and reactive oxygen species (ROS) as well as boosting superoxide dismutase (SOD). TPTS was able to suppress cell apoptosis by suppressing Caspase-3 activity and cleavage. TPTS exerted protective effects against cell injury and apoptosis induced by the H/R conditions, which were associated with its regulation of Akt signaling. Moreover, TPTS induced osteoblast differentiation under the H/R condition. In summary, TPTS attenuates H/R-induced injury in osteoblasts by regulating AKT signaling.
Collapse
Affiliation(s)
- Chuan-Yu Hu
- Department of Orthopedics Ward 4, Dongzhimen Hospital Beijing University of Chinese Medicine, Beijing, 100700, China
| | - Guo-Yan Li
- Department of Anesthesiology, Dongzhimen Hospital Beijing University of Chinese Medicine, Beijing, 100700, China.
| | - Chun-Tian Li
- Department of Acupuncture, Dongzhimen Hospital Beijing University of Chinese Medicine, Beijing, 100700, China
| |
Collapse
|
30
|
Liu X, Pan B, Wang X, Xu J, Wang X, Song Z, Zhang E, Wang F, Wang W. Ischemia/reperfusion-activated ferroptosis in the early stage triggers excessive inflammation to aggregate lung injury in rats. Front Med (Lausanne) 2023; 10:1181286. [PMID: 37425328 PMCID: PMC10327590 DOI: 10.3389/fmed.2023.1181286] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2023] [Accepted: 06/05/2023] [Indexed: 07/11/2023] Open
Abstract
Objective Lung ischemia/reperfusion injury (LIRI) is a clinical syndrome of acute lung injury that occurs after lung transplantation or remote organ ischemia. Ferroptosis and inflammation are involved in the pathogenesis of LIRI according to the results of several studies on animal models. However, the interactive mechanisms between ferroptosis and inflammation contributing to LIRI remain unclear. Methods HE staining and indicators of oxidative stress were used to evaluated the lung injury. The reactive oxygen species (ROS) level was examined by DHE staining. The quantitative Real-time PCR (qRT-PCR) and western blot analysis were employed to detect the level of inflammation and ferroptosis, and deferoxamine (DFO) was used to assess the importance of ferroptosis in LIRI and its effect on inflammation. Results In the present study, the link of ferroptosis with inflammation was evaluated at reperfusion 30-, 60- and 180-minute time points, respectively. As the results at reperfusion 30-minute point shown, the pro-ferroptotic indicators, especially cyclooxygenase (COX)-2 and acyl-CoA synthetase long-chain family member 4 (ACSL4), were upregulated while the anti-ferroptotic factors glutathione peroxidase 4 (GPX4), cystine-glumate antiporter (XCT) and ferritin heavy chain (FTH1) were downregulated. Meanwhile, the increased level of interleukin (IL)-6, tumor necrosis factor alpha (TNF-α) and IL-1β were observed beginning at reperfusion 60-minute point but mostly activated at reperfusion 180-minute point. Furthermore, deferoxamine (DFO) was employed to block ferroptosis, which can alleviate lung injury. Expectedly, the survival rate of rats was increased and the lung injury was mitigated containing the improvement of type II alveolar cells ultrastructure and ROS production. In addition, at the reperfusion 180-minute point, the inflammation was observed to be dramatically inhibited after DFO administration as verified by IL-6, TNF-α and IL-1β detection. Conclusion These findings suggest that ischemia/reperfusion-activated ferroptosis plays an important role as the trigger for inflammation to further deteriorate lung damages. Inhibiting ferroptosis may have therapeutic potential for LIRI in clinical practice.
Collapse
Affiliation(s)
- Xiujie Liu
- School of Basic Medical Science, Wenzhou Medical University, Wenzhou, China
- Institute of Ischemia/Reperfusion Injury, Wenzhou Medical University, Wenzhou, China
| | - Binhui Pan
- Nephrology Department, Wenzhou Central Hospital, Wenzhou, China
| | - Xiaoting Wang
- School of Basic Medical Science, Wenzhou Medical University, Wenzhou, China
- Institute of Ischemia/Reperfusion Injury, Wenzhou Medical University, Wenzhou, China
| | - Junpeng Xu
- School of Basic Medical Science, Wenzhou Medical University, Wenzhou, China
- Institute of Ischemia/Reperfusion Injury, Wenzhou Medical University, Wenzhou, China
| | - Xinyu Wang
- School of Basic Medical Science, Wenzhou Medical University, Wenzhou, China
- Institute of Ischemia/Reperfusion Injury, Wenzhou Medical University, Wenzhou, China
| | - Zhengyang Song
- School of Basic Medical Science, Wenzhou Medical University, Wenzhou, China
- Institute of Ischemia/Reperfusion Injury, Wenzhou Medical University, Wenzhou, China
| | - Eryao Zhang
- Department of Gastroenterology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| | - Fangyan Wang
- School of Basic Medical Science, Wenzhou Medical University, Wenzhou, China
- Institute of Ischemia/Reperfusion Injury, Wenzhou Medical University, Wenzhou, China
| | - Wantie Wang
- School of Basic Medical Science, Wenzhou Medical University, Wenzhou, China
- Institute of Ischemia/Reperfusion Injury, Wenzhou Medical University, Wenzhou, China
| |
Collapse
|
31
|
Zhang L, Wang S, Zhang Y, Li F, Yu C. Sulforaphane alleviates lung ischemia‑reperfusion injury through activating Nrf‑2/HO‑1 signaling. Exp Ther Med 2023; 25:265. [PMID: 37206558 PMCID: PMC10189751 DOI: 10.3892/etm.2023.11964] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2022] [Accepted: 03/10/2023] [Indexed: 05/21/2023] Open
Abstract
Oxidative stress and inflammation are both involved in the pathogenesis of lung ischemia-reperfusion (I/R) injury. Sulforaphane (SFN) is a natural product with cytoprotective, anti-inflammatory, and antioxidant properties. The present study hypothesized that SFN may protect against lung I/R injury via the regulation of antioxidant and anti-inflammatory-related pathways. A rat model of lung I/R injury was established, and rats were randomly divided into 3 groups: Sham group, I/R group, and SFN group. It was shown that SFN protected against a pathological inflammatory response via inhibition of neutrophil accumulation and in the reduction of the serum levels of the pro-inflammatory cytokines, IL-6, IL-1β, and TNF-α. SFN treatment also significantly inhibited lung reactive oxygen species production, decreased the levels of 8-OH-dG and malondialdehyde, and reversed the decrease in the antioxidant activities of the enzymes catalase, superoxide dismutase, and glutathione peroxidase in the lungs of the I/R treated rats. In addition, SFN ameliorated I/R-induced lung apoptosis in rats by suppressing Bax and cleaved caspase-3 levels and increased Bcl-2 expression. Furthermore, SFN treatment activated an Nrf2-related antioxidant pathway, as indicated by the increased nuclear transfer of Nrf2 and the downstream HO-1 and NADPH quinone oxidoreductase-1. In conclusion, these findings suggested that SFN protected against I/R-induced lung lesions in rats via activation of the Nrf2/HO-1 pathway and the accompanied anti-inflammatory and anti-apoptotic effects.
Collapse
Affiliation(s)
- Liang Zhang
- Department of Respiratory and Critical Care Medicine, Yantaishan Hospital, Yantai, Shandong 264001, P.R. China
| | - Shuxian Wang
- Department of Respiratory, Yantai Beihai Hospital, Yantai, Shandong 265701, P.R. China
| | - Ying Zhang
- Department of Emergency, Tai'an Central Hospital, Tai'an, Shandong 271000, P.R. China
| | - Fenghuan Li
- Department of Respiratory and Critical Care Medicine, Yantaishan Hospital, Yantai, Shandong 264001, P.R. China
| | - Chaoxiao Yu
- Department of Respiratory and Critical Care Medicine, Yantaishan Hospital, Yantai, Shandong 264001, P.R. China
- Correspondence to: Dr Chaoxiao Yu, Department of Respiratory and Critical Care Medicine, Yantaishan Hospital, 10,087 Keji Road, Laishan, Yantai, Shandong 264001, P.R. China
| |
Collapse
|
32
|
Gokbulut P, Kuskonmaz SM, Koc G, Onder CE, Yumusak N, Erel O, Nural AS, Culha C. Evaluation of the effects of empagliflozin on acute lung injury in rat intestinal ischemia-reperfusion model. J Endocrinol Invest 2023; 46:1017-1026. [PMID: 36495440 DOI: 10.1007/s40618-022-01978-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/21/2022] [Accepted: 11/30/2022] [Indexed: 12/14/2022]
Abstract
BACKGROUND Empagliflozin is a selective sodium-glucose co-transporter (SGLT2) inhibitor that is approved for the treatment of type 2 diabetes. The beneficial effects of empagliflozin on other organ systems including the heart and kidneys have been proven. The aim of this study is to evaluate the role of empagliflozin on acute lung injury induced by intestinal ischemia-reperfusion (I/R). MATERIALS AND METHODS A total of 27 male Wistar albino rats were divided into three groups: sham, I/R, and I/R + empagliflozin; each group containing nine animals. Sham group rats underwent laparotomy without I/R injury. Rats in the I/R group underwent laparotomy, 1 h of after ischemia-reperfusion injury (superior mesenteric artery ligation was followed by 2 h of reperfusion). Rats in I/R were given empagliflozin (30 mg/kg) by gastric gavage for 7 days before the ischemia-reperfusion injury. All animals were killed at the end of reperfusion and lung tissue samples were obtained for immunohistochemical staining and histopathological investigation in all groups. RESULTS Serum glucose, AST, ALT, creatinine, native thiol, total thiol, and disulfide levels and disulfide-native thiol, disulfide-total thiol, and native thiol-total thiol ratios as well as the IMA levels were analyzed and compared among the groups. While intestinal I/R significantly increases serum aspartate aminotransferase (AST), alanine aminotransferase (ALT), and creatinine levels; did not cause any change in homeostasis parameters and IMA level. Empagliflozin treatment had no significant effect on biochemical parameters. Empagliflozin treatment induced a significant decrease in positive immunostaining for IL-1, IL-6, TNF-alpha, caspase 3, caspase 8, and caspase 9 compared to the I/R group in lung tissue samples. Intestinal I/R caused severe histopathological injury including edema, hemorrhage, increased thickness of the alveolar wall, and infiltration of inflammatory cells into alveolar spaces. Empagliflozin treatment significantly attenuated the severity of intestinal I/R injury. CONCLUSIONS It was concluded that empagliflozin treatment may have beneficial effects in acute lung injury, and, therefore, has the potential for clinical use.
Collapse
Affiliation(s)
- P Gokbulut
- Department of Endocrinology, Ankara Training and Research Hospital, Hacettepe Neighborhood Ulucanlar Caddesi No: 89 Altindag, 06230, Ankara, Turkey.
| | - S M Kuskonmaz
- Department of Endocrinology, Ankara Training and Research Hospital, Hacettepe Neighborhood Ulucanlar Caddesi No: 89 Altindag, 06230, Ankara, Turkey
| | - G Koc
- Department of Endocrinology, Ankara Training and Research Hospital, Hacettepe Neighborhood Ulucanlar Caddesi No: 89 Altindag, 06230, Ankara, Turkey
| | - C E Onder
- Ömer Halisdemir University Training and Research Hospital, Nigde, Turkey
| | - N Yumusak
- Faculty of Veterinary Medicine, Harran University, Sanlıurfa, Turkey
| | - O Erel
- Department of Biochemistry, Ankara City Hospital, Ankara, Turkey
| | - A S Nural
- Department of Biochemistry, Ankara City Hospital, Ankara, Turkey
| | - C Culha
- Department of Endocrinology, Ankara Training and Research Hospital, Hacettepe Neighborhood Ulucanlar Caddesi No: 89 Altindag, 06230, Ankara, Turkey
| |
Collapse
|
33
|
Park H, Jung YS, Suh GJ, Kwon WY, Kim KS, Kim T, Kim H, Shin J. Combination Therapy of Niacin and Apocynin Attenuates Lung Injury During Sepsis in Rats. J Surg Res 2023; 285:51-58. [PMID: 36640610 DOI: 10.1016/j.jss.2022.12.020] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2022] [Revised: 12/06/2022] [Accepted: 12/25/2022] [Indexed: 01/13/2023]
Abstract
INTRODUCTION Oxidative stress contributes to tissue injury through reactive oxygen species-dependent signaling pathways during sepsis. We studied therapeutic benefits of the combination therapy of niacin, which increased reduced glutathione levels, and apocynin, which suppressed reduced nicotinamide adenine dinucleotide phosphate (NADPH) oxidase (Nox) activity, in septic rats. MATERIALS AND METHODS Polymicrobial sepsis was induced through cecal ligation and puncture (CLP) with antibiotics in male Sprague-Dawley rats (n = 189). The rats were randomly divided into sham, CLP, CLP + niacin, CLP + apocynin, and CLP + niacin + apocynin groups. Six hours after CLP, vehicle, niacin (360 mg/kg through the orogastric tube), and/or apocynin (20 mg/kg through intraperitoneal injection) were administered. The occurrence of mortality for 72 h after CLP was observed. Next, a separate set of animals was euthanized at 24 h post-CLP for lung tissue analyses. RESULTS Combination therapy with niacin and apocynin significantly improved survival in rats with sepsis (75.0% versus 28.8%, P = 0.006) but monotherapy with niacin or apocynin did not. Monotherapy with niacin and apocynin appeared to increase NADPH levels and decrease Nox levels and activity, respectively, but failed to show statistical significances. However, combination therapy significantly decreased Nox levels and activity, increased NADPH and glutathione levels, decreased intranuclear nuclear factor-κB (NF-κB) p65 levels, reduced inflammatory cytokine expression and malondialdehyde levels, and attenuated histological lung injuries. CONCLUSIONS Combination therapy with niacin and apocynin synergistically attenuated lung injuries and improved survival in rats with sepsis through niacin-induced glutathione redox cycle activation and apocynin-induced Nox suppression.
Collapse
Affiliation(s)
- Heesu Park
- Department of Emergency Medicine, Seoul National University Hospital, Seoul, Republic of Korea
| | - Yoon Sun Jung
- Department of Critical Care Medicine, Seoul National University Hospital, Seoul, Republic of Korea.
| | - Gil Joon Suh
- Department of Emergency Medicine, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Woon Yong Kwon
- Department of Emergency Medicine, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Kyung Su Kim
- Department of Emergency Medicine, Seoul National University Hospital, Seoul, Republic of Korea
| | - Taegyun Kim
- Department of Emergency Medicine, Seoul National University Hospital, Seoul, Republic of Korea
| | - Hayoung Kim
- Department of Emergency Medicine, Seoul National University Hospital, Seoul, Republic of Korea
| | - Jieun Shin
- Department of Emergency Medicine, Seoul National University Hospital, Seoul, Republic of Korea
| |
Collapse
|
34
|
Zhang X, Yu S, Li X, Wen X, Liu S, Zu R, Ren H, Li T, Yang C, Luo H. Research progress on the interaction between oxidative stress and platelets: Another avenue for cancer? Pharmacol Res 2023; 191:106777. [PMID: 37080257 DOI: 10.1016/j.phrs.2023.106777] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/04/2023] [Revised: 04/06/2023] [Accepted: 04/18/2023] [Indexed: 04/22/2023]
Abstract
Oxidative stress (OS) is a chemical imbalance between an oxidant and an antioxidant, causing damage to redox signaling and control or causing molecular damage. Unbalanced oxidative metabolism can produce excessive reactive oxygen species (ROS). These excess ROS can cause drastic changes in platelet metabolism and further affect platelet function. It will also lead to an increase in platelet procoagulant phenotype and cell apoptosis, which will increase the risk of thrombosis. The creation of ROS and subsequent platelet activation, adhesion, and recruitment are then further encouraged in an auto-amplifying loop by ROS produced from platelets. Meanwhile, cancer cells produce a higher concentration of ROS due to their fast metabolism and high proliferation rate. However, excessive ROS can result in damage to and modification of cellular macromolecules. The formation of cancer and its progression is strongly associated with oxidative stress and the resulting oxidative damage. In addition, platelets are an important part of the tumor microenvironment, and there is a significant cross-communication between platelets and cancer cells. Cancer cells alter the activation status of platelets, their RNA spectrum, proteome, and other properties. The "cloaking" of cancer cells by platelets providing physical protection,avoiding destruction from shear stress and the attack of immune cells, promoting tumor cell invasion.We explored the vicious circle interaction between ROS, platelets, and cancer in this review, and we believe that ROS can play a stimulative role in tumor growth and metastasis through platelets.
Collapse
Affiliation(s)
- Xingmei Zhang
- Department of Clinical Laboratory, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, Chengdu, 610041 China; College of Medical Technology, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, 610042, China
| | - Sisi Yu
- Department of Clinical Laboratory, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, Chengdu, 610041 China
| | - Xiaobo Li
- Molecular Diagnostic Laboratory of Department of Microbiology and Immunology, 3201 Hospital Affiliated to Medical College of Xi'an Jiaotong University, Hanzhong 723099, China
| | - Xiaoxia Wen
- College of Medical Technology, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, 610042, China
| | - Shan Liu
- College of Medical Technology, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, 610042, China
| | - Ruiling Zu
- Department of Clinical Laboratory, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, Chengdu, 610041 China
| | - Hanxiao Ren
- College of Medical Technology, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, 610042, China
| | - Tian Li
- School of Basic Medicine, Fourth Military Medical University, Xi'an 710032, China.
| | - Chaoguo Yang
- College of Medical Technology, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, 610042, China.
| | - Huaichao Luo
- Department of Clinical Laboratory, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, Chengdu, 610041 China.
| |
Collapse
|
35
|
miR-125b-5p in adipose derived stem cells exosome alleviates pulmonary microvascular endothelial cells ferroptosis via Keap1/Nrf2/GPX4 in sepsis lung injury. Redox Biol 2023; 62:102655. [PMID: 36913799 PMCID: PMC10023991 DOI: 10.1016/j.redox.2023.102655] [Citation(s) in RCA: 96] [Impact Index Per Article: 48.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2023] [Revised: 02/20/2023] [Accepted: 03/02/2023] [Indexed: 03/11/2023] Open
Abstract
BACKGROUND Sepsis is a fatal disease with a high rate of morbidity and mortality, during which acute lung injury is the earliest and most serious complication. Injury of pulmonary microvascular endothelial cells (PMVECs) induced by excessive inflammation plays an important role in sepsis acute lung injury. This study is meant to explore the protective effect and mechanism of ADSCs exosomes on excessive inflammation PMVECs injury. RESULTS We successfully isolated ADSCs exosomes, the characteristic of which were confirmed. ADSCs exosomes reduced excessive inflammatory response induced ROS accumulation and cell injury in PMVECs. Besides, ADSCs exosomes inhibited excessive inflammatory response induced ferroptosis while upregulated expression of GPX4 in PMVECs. And further GPX4 inhibition experiments revealed that ADSCs exosomes alleviated inflammatory response induced ferroptosis via upregulating GPX4. Meanwhile, ADSCs exosomes could increase the expression and nucleus translocation of Nrf2, while decrease the expression of Keap1. miRNA analysis and further inhibition experiments verified that specific delivery of miR-125b-5p by ADSCs exosomes inhibited Keap1 and alleviated ferroptosis. In CLP induced sepsis model, ADSCs exosomes could relieve the lung tissue injury and reduced the death rate. Besides, ADSCs exosomes alleviated oxidative stress injury and ferroptosis of lung tissue, while remarkably increase expression of Nrf2 and GPX4. CONCLUSION Collectively, we illustrated a novel potentially therapeutic mechanism that miR-125b-5p in ADSCs exosomes could alleviate the inflammation induced PMVECs ferroptosis in sepsis induced acute lung injury via regulating Keap1/Nrf2/GPX4 expression, hence improve the acute lung injury in sepsis.
Collapse
|
36
|
Tian Y, Zhu CL, Li P, Li HR, Liu Q, Deng XM, Wang JF. Nicotinamide Mononucleotide Attenuates LPS-Induced Acute Lung Injury With Anti-Inflammatory, Anti-Oxidative and Anti-Apoptotic Effects. J Surg Res 2023; 283:9-18. [PMID: 36347171 DOI: 10.1016/j.jss.2022.09.030] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Revised: 08/23/2022] [Accepted: 09/18/2022] [Indexed: 11/07/2022]
Abstract
INTRODUCTION Nicotinamide mononucleotide (NMN) is a nucleotide that is commonly recognized for its role as an intermediate of nicotinamide adenine dinucleotide (NAD+) biosynthesis with multiple pharmacological effects. The purpose of this study was to evaluate the protective effect of nicotinamide mononucleotide (NMN) against lipopolysaccharide (LPS)-induced acute lung injury (ALI). METHODS We investigated the effect of NMN on ALI-induced inflammatory response, oxidative stress, and cell apoptosis. The ALI mouse model was performed by injecting LPS intratracheally at a dose of 10 mg/kg in 50 μL saline. Flow cytometry was used to detect neutrophil infiltration in bronchoalveolar lavage fluid (BALF), and ELISA was used to detect the contents of inflammatory cytokines TNF-α, IL-1β and IL-6 in BALF. Oxidative stress was evaluated by determining the superoxide dismutase (SOD) activity and malondialdehyde (MDA) content in lung tissue. ROS formation was analyzed by immunofluorescence. Western blotting was performed to detect apoptotic levels and p38MAPK/NF-κB phosphorylation levels in lung tissue. RESULTS In the ALI mouse model, NMN showed a significant therapeutic effect compared to the LPS group. NMN attenuated the pathological damage and cell apoptosis in lung tissue, decreased the levels of TNF-α, IL-1β, and IL-6 in BALF, and reduced the number of total cells and neutrophils in BALF. In addition, NMN attenuated the LPS-induced elevation of dry-to-wet ratio, MDA content, p38 MAPK and p65 NF-κB phosphorylation levels, and the SOD activity was increased by NMN treatment. CONCLUSIONS In conclusion, the present study showed that NMN exerted a protective effect on LPS-induced ALI with anti-inflammatory, antioxidative, and antiapoptotic effects.
Collapse
Affiliation(s)
- Ye Tian
- Department of Anesthesiology, The Sixth Medical Centre of General Hospital of PLA, Beijing, China
| | - Cheng-Long Zhu
- Changhai Hospital, Naval Medical University, Shanghai, China
| | - Peng Li
- Changhai Hospital, Naval Medical University, Shanghai, China
| | - Hui-Ru Li
- Changhai Hospital, Naval Medical University, Shanghai, China
| | - Qiang Liu
- Changhai Hospital, Naval Medical University, Shanghai, China
| | - Xiao-Ming Deng
- Changhai Hospital, Naval Medical University, Shanghai, China.
| | - Jia-Feng Wang
- Changhai Hospital, Naval Medical University, Shanghai, China.
| |
Collapse
|
37
|
Altan M, Yaman MO, Kervancıoğlu G, Kılıç A, Demirci EK, Bozdoğan Polat SH, Karadeniz Z, Güner I, Yelmen N, Şahin G. Aortic ischemia-reperfusion injury and potency of fluoxetine. IRANIAN JOURNAL OF BASIC MEDICAL SCIENCES 2023; 26:301-307. [PMID: 36865048 PMCID: PMC9922374 DOI: 10.22038/ijbms.2023.65974.14508] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 06/08/2022] [Accepted: 12/07/2022] [Indexed: 03/04/2023]
Abstract
Objectives Due to cross-clamping of the aorta during aortic aneurysm surgeries, ischemia-reperfusion (IR) develops, and it may cause damage to the aorta itself or even to remote organs by oxidative stress or inflammation. Fluoxetine (FLX) which might be used in the preoperative period for its tranquilizing effect also has antioxidant effects in short-term use. The purpose of our study is to examine whether FLX protects aorta tissue, against the damage caused by IR. Materials and Methods Three groups of Wistar rats were formed randomly. 1) Control group (sham-operated), 2) IR group (60 min ischemia+120 min perfusion), and 3) FLX+IR group (FLX dose was 20 mg/kg for 3 days IP before IR). At the end of each procedure, aorta samples were collected, and oxidant-antioxidant, anti-inflammatory, and anti-apoptotic status of the aorta were evaluated. Histological examinations of the samples were provided. Results Levels of LOOH, MDA, ROS, TOS, MPO, TNFα, IL-1β, IL-6, NF-kB, MMP-9, caspase-9, 8-OHdG, NO, and HA were found to be significantly increased in the IR group compared with control (P<0.05) and SOD, GSH, TAS, and IL-10 levels were significantly lower (P<0.05). FLX significantly decreased LOOH, MDA, ROS, TOS, MPO, TNFα, IL-1β, IL-6, NF-kB, MMP-9, caspase-9, 8-OHdG, NO, and HA levels in the FLX+IR group compared with IR group (P<0.05) and increased IL-10, SOD, GSH, and TAS (P<0.05). FLX administration prevented the deterioration of aortic tissue damage. Conclusion Our study is the first study that demonstrates FLX-mediated suppression of IR injury in the infrarenal abdominal aorta by antioxidant, anti-inflammatory, and anti-apoptotic properties.
Collapse
Affiliation(s)
- Mehmet Altan
- Istanbul University-Cerrahpaşa, Cerrahpasa Medical Faculty, Department of Physiology, Istanbul, Turkey
| | - Muhittin Onur Yaman
- Istanbul University-Cerrahpaşa, Vocational School of Health Sciences, Istanbul, Turkey
| | - Gülnaz Kervancıoğlu
- Istanbul Aydın University, Medical Faculty, Department of Histology & Embryology, Istanbul, Turkey
| | - Aysu Kılıç
- Bezmialem University, Medical Faculty, Department of Physiology, Istanbul, Turkey
| | - Elif Kervancıoğlu Demirci
- Istanbul University, Istanbul Faculty of Medicine, Department of Histology & Embryology, Istanbul, Turkey
| | | | - Zeliha Karadeniz
- Istanbul Aydın University, Medical Faculty, Department of Obstetrics & Gynecology, Istanbul, Turkey
| | - Ibrahim Güner
- Tekirdağ Namık Kemal University, Medical Faculty, Department of Physiology, Tekirdağ, Turkey
| | - Nermin Yelmen
- Istanbul University-Cerrahpaşa, Cerrahpasa Medical Faculty, Department of Physiology, Istanbul, Turkey
| | - Gülderen Şahin
- Istanbul Aydın University, Medical Faculty, Department of Physiology, Istanbul, Turkey,Corresponding author: Gülderen Şahin. Florya Yerleşkesi, Besyol Mah. Inonu Cad. No: 38, Sefakoy–Kuçukcekmece,Istanbul, Turkey. Tel: +90 532 393 21 18;
| |
Collapse
|
38
|
Liu MY, Ju YN, Jia BW, Sun XK, Qiu L, Liu HY, Xu GX, Tai QH, Tan J, Gao W. Inhibition of DNA methylation attenuates lung ischemia-reperfusion injury after lung transplantation. J Int Med Res 2023; 51:3000605231153587. [PMID: 36756846 PMCID: PMC9912569 DOI: 10.1177/03000605231153587] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/10/2023] Open
Abstract
OBJECTIVE DNA methylation plays an important role in inflammation and oxidative stress. This study aimed to investigate the effect of inhibiting DNA methylation on lung ischemia-reperfusion injury (LIRI). METHODS We adopted a completely random design for our study. Thirty-two rats were randomized into the sham, LIRI, azathioprine (AZA), and pluripotin (SC1) groups. The rats in the LIRI, AZA, and SC1 groups received left lung transplantation and intravenous injection of saline, AZA, and SC1, respectively. After 24 hours of reperfusion, histological injury, the arterial oxygen partial pressure to fractional inspired oxygen ratio, the wet/dry weight ratio, protein and cytokine concentrations in lung tissue, and DNA methylation in lung tissue were evaluated. The pulmonary endothelium that underwent hypoxemia and reoxygenation was treated with AZA or SC1. Endothelial apoptosis, chemokines, reactive oxygen species, nuclear factor-κB, and apoptotic proteins in the endothelium were studied. RESULTS Inhibition of DNA methylation by AZA attenuated lung injury, inflammation, and the oxidative stress response, but SC1 aggravated LIRI injury. AZA significantly improved endothelial function, suppressed apoptosis and necrosis, reduced chemokines, and inhibited nuclear factor-κB. CONCLUSIONS Inhibition of DNA methylation ameliorates LIRI and apoptosis and improves pulmonary function via the regulation of inflammation and oxidative stress.
Collapse
Affiliation(s)
- Ming-yuan Liu
- Department of Anesthesiology, The Second Affiliated Hospital of
Harbin Medical University, Harbin, Heilongjiang, China
| | - Ying-nan Ju
- Department of Intensive Care Unit, The Third Affiliated Hospital
of Harbin Medical University, Harbin, Heilongjiang, China
| | - Bao-wei Jia
- Department of Anesthesiology, The Second Affiliated Hospital of
Harbin Medical University, Harbin, Heilongjiang, China
| | - Xi-kun Sun
- Department of Anesthesiology, The Second Affiliated Hospital of
Harbin Medical University, Harbin, Heilongjiang, China
| | - Lin Qiu
- Department of Anesthesiology, The Second Affiliated Hospital of
Harbin Medical University, Harbin, Heilongjiang, China
| | - Heng-yu Liu
- Department of Anesthesiology, The Second Affiliated Hospital of
Harbin Medical University, Harbin, Heilongjiang, China
| | - Guang-xiao Xu
- Department of Anesthesiology, The Second Affiliated Hospital of
Harbin Medical University, Harbin, Heilongjiang, China
| | - Qi-hang Tai
- Department of Anesthesiology, The Second Affiliated Hospital of
Harbin Medical University, Harbin, Heilongjiang, China
| | - Jing Tan
- Department of Anesthesiology, The Second Affiliated Hospital of
Harbin Medical University, Harbin, Heilongjiang, China
| | - Wei Gao
- Department of Anesthesiology, The Second Affiliated Hospital of
Harbin Medical University, Harbin, Heilongjiang, China,Wei Gao, Department of Anesthesiology, The
Second Affiliated Hospital of Harbin Medical University, 246 Xuefu Road, Harbin,
Heilongjiang 150081, China.
| |
Collapse
|
39
|
Pre-Treatment of Transplant Donors with Hydrogen Sulfide to Protect against Warm and Cold Ischemia-Reperfusion Injury in Kidney and Other Transplantable Solid Organs. Int J Mol Sci 2023; 24:ijms24043518. [PMID: 36834928 PMCID: PMC9963309 DOI: 10.3390/ijms24043518] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2023] [Revised: 02/01/2023] [Accepted: 02/07/2023] [Indexed: 02/12/2023] Open
Abstract
Ischemia-reperfusion injury (IRI), a pathological condition resulting from prolonged cessation and subsequent restoration of blood flow to a tissue, is an inevitable consequence of solid organ transplantation. Current organ preservation strategies, such as static cold storage (SCS), are aimed at reducing IRI. However, prolonged SCS exacerbates IRI. Recent research has examined pre-treatment approaches to more effectively attenuate IRI. Hydrogen sulfide (H2S), the third established member of a family of gaseous signaling molecules, has been shown to target the pathophysiology of IRI and thus appears to be a viable candidate that can overcome the transplant surgeon's enemy. This review discusses pre-treatment of renal grafts and other transplantable organs with H2S to mitigate transplantation-induced IRI in animal models of transplantation. In addition, ethical principles of pre-treatment and potential applications of H2S pre-treatment in the prevention of other IRI-associated conditions are discussed.
Collapse
|
40
|
Mitochondrial transplant after ischemia reperfusion promotes cellular salvage and improves lung function during ex-vivo lung perfusion. J Heart Lung Transplant 2023; 42:575-584. [PMID: 36707296 DOI: 10.1016/j.healun.2023.01.002] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2022] [Revised: 12/02/2022] [Accepted: 01/04/2023] [Indexed: 01/13/2023] Open
Abstract
BACKGROUND In lung transplantation, ischemia-reperfusion injury associated with mitochondrial damage can lead to graft rejection. Intact, exogenous mitochondria provide a unique treatment option to salvage damaged cells within lung tissue. METHODS We developed a novel method to freeze and store allogeneic mitochondria isolated from porcine heart tissue. Stored mitochondria were injected into a model of induced ischemia-reperfusion injury using porcine ex-vivo lung perfusion. Treatment benefits to immune modulation, antioxidant defense, and cellular salvage were evaluated. These findings were corroborated in human lungs undergoing ex-vivo lung perfusion. Lung tissue homogenate and primary lung endothelial cells were then used to address underlying mechanisms. RESULTS Following cold ischemia, mitochondrial transplant reduced lung pulmonary vascular resistance and tissue pro-inflammatory signaling and cytokine secretion. Further, exogenous mitochondria reduced reactive oxygen species by-products and promoted glutathione synthesis, thereby salvaging cell viability. These results were confirmed in a human model of ex-vivo lung perfusion wherein transplanted mitochondria decreased tissue oxidative and inflammatory signaling, improving lung function. We demonstrate that transplanted mitochondria induce autophagy and suggest that bolstered autophagy may act upstream of the anti-inflammatory and antioxidant benefits. Importantly, chemical inhibitors of the MEK autophagy pathway blunted the favorable effects of mitochondrial transplant. CONCLUSIONS These data provide direct evidence that mitochondrial transplant improves cellular health and lung function when administered during ex-vivo lung perfusion and suggest the mechanism of action may be through promotion of cellular autophagy. Data herein contribute new insights into the therapeutic potential of mitochondrial transplant to abate ischemia-reperfusion injury during lung transplant, and thus reduce graft rejection.
Collapse
|
41
|
Anti-Oxidant and Pro-Oxidant Effects of Peroxiredoxin 6: A Potential Target in Respiratory Diseases. Cells 2023; 12:cells12010181. [PMID: 36611974 PMCID: PMC9818991 DOI: 10.3390/cells12010181] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Revised: 12/26/2022] [Accepted: 12/30/2022] [Indexed: 01/03/2023] Open
Abstract
Peroxiredoxin 6 (PRDX6) is widely distributed in several organs, especially the lungs. The role of PRDX6 in oxidative stress is controversial and even contradictory, as indicated by research conducted over the past 20 years. PRDX6 has anti-oxidant or pro-oxidant effects on oxidative stress in different diseases. It can even exhibit both anti-oxidant and pro-oxidant effects in the same disease. These findings are attributed to the fact that PRDX6 is a multifunctional enzyme. The peroxidase and phospholipase A2 activity of PRDX6 is closely related to its anti-oxidant and pro-oxidant effects, which leads to the conflicting regulatory effects of PRDX6 on oxidative stress in respiratory diseases. Moreover, PRDX6 interacts with multiple redox signaling pathways to interfere with cell proliferation and apoptosis. PRDX6 has become a new target in respiratory disease research due to its important regulatory role in oxidative stress. In this paper, the role of PRDX6 in oxidative stress in respiratory diseases and the research progress in targeting PRDX6 are reviewed.
Collapse
|
42
|
Iguidbashian KG, Robison J, Khailova L, Jaggers J, Ing R, Lawson S, Osorio Lujan SM, Klawitter J, Davidson JA. Changes in infant porcine pulmonary tissue oxylipins induced by cardiopulmonary bypass. Pediatr Res 2022; 92:1274-1281. [PMID: 35681098 DOI: 10.1038/s41390-022-02125-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/27/2021] [Revised: 03/30/2022] [Accepted: 05/17/2022] [Indexed: 11/09/2022]
Abstract
BACKGROUND Oxylipins are metabolites derived from fatty acids such as arachidonic acid (AA) and are key mediators in inflammation, host defense, and tissue injury. Serum oxylipins increase in adults after cardiopulmonary bypass (CPB) but tissue-level changes are poorly defined. The objective of this study was to characterize pulmonary tissue oxylipins in an infant porcine model of CPB with deep hypothermic circulatory arrest (DHCA). METHODS Infant pigs underwent CPB with DHCA. Controls received anesthesia only. Right upper and lower lobes of the lung underwent oxylipin analysis via liquid chromatography-tandem mass spectrometry. One-way ANOVA was utilized to assess differences in oxylipin concentrations across groups, followed by pairwise comparisons. RESULTS AA and multiple AA metabolites via cytochrome P450 (CYP450), lipoxygenase (LOX), and cyclooxygenase (COX) pathways were significantly increased in the upper and lower lobe of pigs exposed to CPB/DHCA as compared to controls. Multiple prostaglandin metabolites produced via COX were also significantly elevated in the lower lobes of control animals. CONCLUSIONS CPB/DHCA induces a significant increase in pulmonary tissue AA, with subsequent metabolism via COX, LOX, and CYP450 pathways. Interestingly, prostaglandins were also elevated in the lower lobes of the controls, suggesting a mechanism separate from CPB/DHCA. Future oxylipin studies are needed to better understand CPB-induced acute lung injury. IMPACT CPB/DHCA and, to a lesser extent, lung region influence pulmonary tissue-level AA metabolite production. Inflammatory mediator AA metabolites have been noted in previous studies to increase following CPB; however, this is the first study to look at pulmonary tissue-level differences following CPB/DHCA. Increases in many AA metabolites, including LOX- and CYP450-derived products, were seen in both upper and lower lobe of piglets following CPB/DHCA. COX-derived prostaglandin metabolites were increased not only in CPB upper and lower lobe but also in mechanically ventilated control lower lobe, suggesting an additional, separate mechanism from CPB/DCHA.
Collapse
Affiliation(s)
- Kelsey G Iguidbashian
- Department of Pediatrics, University of Colorado/Children's Hospital of Colorado, Aurora, CO, USA
| | - Justin Robison
- Department of Pediatrics, Washington University in St. Louis, St. Louis, MO, USA
| | - Ludmila Khailova
- Department of Pediatrics, University of Colorado/Children's Hospital of Colorado, Aurora, CO, USA
| | - James Jaggers
- Department of Surgery, University of Colorado, Aurora, CO, USA
| | - Richard Ing
- Department of Anesthesiology, University of Colorado, Aurora, CO, USA
| | - Scott Lawson
- Heart Institute, Children's Hospital Colorado, Aurora, CO, USA
| | - Suzanne M Osorio Lujan
- Department of Pediatrics, University of Colorado/Children's Hospital of Colorado, Aurora, CO, USA
| | - Jelena Klawitter
- Department of Anesthesiology, University of Colorado, Aurora, CO, USA
| | - Jesse A Davidson
- Department of Pediatrics, University of Colorado/Children's Hospital of Colorado, Aurora, CO, USA.
| |
Collapse
|
43
|
Veraar C, Kirschner E, Schwarz S, Jaksch P, Hoetzenecker K, Tschernko E, Dworschak M, Ankersmit HJ, Moser B. Follistatin-like 1 and Biomarkers of Neutrophil Activation Are Associated with Poor Short-Term Outcome after Lung Transplantation on VA-ECMO. BIOLOGY 2022; 11:biology11101475. [PMID: 36290379 PMCID: PMC9598172 DOI: 10.3390/biology11101475] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/13/2022] [Revised: 09/17/2022] [Accepted: 10/06/2022] [Indexed: 11/06/2022]
Abstract
The investigation of biomarkers associated with undesired outcome following lung transplantation (LuTX) is essential for a better understanding of the underlying pathophysiology, an earlier identification of susceptible recipients and the development of targeted therapeutic options. We therefore determined the longitudinal perioperative course of putative cytokines related to neutrophil activation (chemokine CC motif ligand 4 (CCL-4), interleukin (IL)-23 and Lipocalin 2 (LCN2)) and a cytokine that has been implicated in graft-versus-host disease (Follistatin-like 1 (FSTL1)) in 42 consecutive patients undergoing LuTX. We plotted receiver-operating curves (ROC) to assess the predictive power of the measured cytokines for short-term outcomes namely primary graft dysfunction (PGD), early complications requiring extracorporeal membrane oxygenation (ECMO), and a high postoperative sequential organ failure assessment (SOFA). All cytokines increased immediately after surgery. ROC analyses determined significant associations between CCL4 and a high SOFA score (area under the curve (AUC) 0.74 (95%CI:0.5−0.9; p < 0.05), between LCN2 and postoperative ECMO support (AUC 0.73 (95%CI:0.5−0.9; p < 0.05), and between FSTL1 and PGD (AUC 0.70 (95%CI:0.5−0.9; p < 0.05). The serum concentrations of the neutrophil-derived cytokines LCN2 and CCL4 as well as FSTL1 were all related to poor outcome after LuTX. The specific predictive power, however, still has to be assessed in larger trials. The potential role of FSTL1 as a biomarker in the development of PGD could be of great interest particularly since this protein appears to play a crucial role in allograft tolerance.
Collapse
Affiliation(s)
- Cecilia Veraar
- Department of Anesthesiology, Intensive Care Medicine and Pain Medicine, Division of Cardiothoracic and Vascular Anesthesia and Intensive Care Medicine, Medical University of Vienna, 1090 Vienna, Austria
- Applied Immunology Laboratory, Medical University of Vienna, 1090 Vienna, Austria
- Correspondence:
| | - Enzo Kirschner
- Department of Thoracic Surgery, Medical University of Vienna, 1090 Vienna, Austria
| | - Stefan Schwarz
- Department of Thoracic Surgery, Medical University of Vienna, 1090 Vienna, Austria
| | - Peter Jaksch
- Department of Thoracic Surgery, Medical University of Vienna, 1090 Vienna, Austria
| | - Konrad Hoetzenecker
- Department of Thoracic Surgery, Medical University of Vienna, 1090 Vienna, Austria
| | - Edda Tschernko
- Department of Anesthesiology, Intensive Care Medicine and Pain Medicine, Division of Cardiothoracic and Vascular Anesthesia and Intensive Care Medicine, Medical University of Vienna, 1090 Vienna, Austria
| | - Martin Dworschak
- Department of Anesthesiology, Intensive Care Medicine and Pain Medicine, Division of Cardiothoracic and Vascular Anesthesia and Intensive Care Medicine, Medical University of Vienna, 1090 Vienna, Austria
| | - Hendrik J. Ankersmit
- Applied Immunology Laboratory, Medical University of Vienna, 1090 Vienna, Austria
- Department of Thoracic Surgery, Medical University of Vienna, 1090 Vienna, Austria
| | - Bernhard Moser
- Department of Thoracic Surgery, Medical University of Vienna, 1090 Vienna, Austria
| |
Collapse
|
44
|
Downregulation of SIRT3 Aggravates Lung Ischemia Reperfusion Injury by Increasing Mitochondrial Fission and Oxidative Stress through HIF-1α-Dependent Mechanisms. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:9041914. [PMID: 36211825 PMCID: PMC9537006 DOI: 10.1155/2022/9041914] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/09/2022] [Revised: 04/11/2022] [Accepted: 05/27/2022] [Indexed: 12/06/2022]
Abstract
Lung ischemia-reperfusion injury (LIRI) is a severe multifaceted pathological condition that can lead to poor patient outcome where oxidative stress and the resulting inflammatory response can trigger and exacerbate tissue damage in LIRI patients. Sirtuin3 (SIRT3), a member of the sirtuin family, protects against oxidative stress-related diseases. However, it remains unclear if and how SIRT3 alleviates lung injury induced by ischemia/reperfusion (I/R). Our previous study showed that lung tissue structures were severely damaged at 6 h after lung I/R in mice, however, repair of the injured lung tissue was significant at 24 h. In this study, we found that both SIRT3 mRNA and protein levels were markedly increased at 24 h after lung I/R in vivo. Meanwhile, inhibition of SIRT3 aggravated lung injury and inflammation, augmented mitochondrial fission and oxidative stress and increased Hypoxia-inducible factor-1α (HIF-1α) expression in vivo. The results suggest that SIRT3 may be an upstream regulator of HIF-1α expression. Knockdown of SIRT3 resulted in excessive mitochondrial fission and increased oxidative stress in vitro, and we found that knocking down the expression of HIF-1α alleviated these changes. This suggests that the SIRT3-HIF-1α signaling pathway is involved in regulating mitochondrial function and oxidative stress. Furthermore, inhibition of dynamin-related protein 1 (Drp-1) by the inhibitor of mitophagy, Mdivi-1, blocked mitochondrial fission and alleviated oxidative stress in vitro. Taken together, our results demonstrated that downregulation of SIRT3 aggravates LIRI by increasing mitochondrial fission and oxidative stress. Activation of SIRT3 inhibits mitochondrial fission and this mechanism may serve as a new therapeutic strategy to treat LIRI.
Collapse
|
45
|
Huang R, Shi Q, Zhang S, Lin H, Han C, Qian X, Huang Y, Ren X, Sun J, Feng N, Xia C, Shi M. Inhibition of the cGAS-STING Pathway Attenuates Lung Ischemia/Reperfusion Injury via Regulating Endoplasmic Reticulum Stress in Alveolar Epithelial Type II Cells of Rats. J Inflamm Res 2022; 15:5103-5119. [PMID: 36091334 PMCID: PMC9462969 DOI: 10.2147/jir.s365970] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2022] [Accepted: 08/26/2022] [Indexed: 11/23/2022] Open
Abstract
Purpose Endoplasmic reticulum stress (ERS) plays an important role in the pathogenesis of lung ischemia/reperfusion (I/R) injury. Cyclic GMP-AMP synthase (cGAS) is a cytosol dsDNA sensor, coupling with downstream stimulator of interferon genes (STING) located in the ER, which involves innate immune responses. The aim of our present study was to investigate the effects of cGAS on lung I/R injury via regulating ERS. Methods We used Sprague-Dawley rats to make the lung I/R model by performing left hilum occlusion-reperfusion surgery. cGAS-specific inhibitor RU.521, STING agonist SR-717, and 4-phenylbutyric acid (4-PBA), the ERS inhibitor, were intraperitoneally administered in rats. Double immunofluorescent staining was applied to detect the colocalization of cGAS or BiP, an ERS protein, with alveolar epithelial type II cells (AECIIs) marker. We used transmission electron microscopy to examine the ultrastructure of ER and mitochondria. Apoptosis and oxidative stress in the lungs were assessed, respectively. The profiles of pulmonary edema and lung tissue injury were evaluated. And the pulmonary ventilation function was measured using a spirometer system. Results In lung I/R rats, the cGAS-STING pathway was upregulated, which implied they were activated. After cGAS-STING pathway was inhibited or activated in lung I/R rats, the ERS was alleviated after cGAS was inhibited, while when STING was activated after lung I/R, ERS was aggravated in the AECIIs, these results suggested that cGAS-STING pathway might trigger ERS responses. Furthermore, activation of cGAS-STING pathway induced increased apoptosis, inflammation, and oxidative stress via regulating ERS and therefore resulted in pulmonary edema and pathological injury in the lungs of I/R rats. Inhibition of cGAS-STING pathway attenuated ERS, therefore attenuated lung injury and promoted pulmonary ventilation function in I/R rats. Conclusion Inhibition of the cGAS-STING pathway attenuates lung ischemia/reperfusion injury via alleviating endoplasmic reticulum stress in alveolar epithelial type II cells of rats.
Collapse
Affiliation(s)
- Renhui Huang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Fudan University, Shanghai, 200032, People's Republic of China.,Department of Clinical Medicine, Shanghai Medical College, Fudan University, Shanghai, 200032, People's Republic of China
| | - Qi Shi
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Fudan University, Shanghai, 200032, People's Republic of China
| | - Shutian Zhang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Fudan University, Shanghai, 200032, People's Republic of China.,Department of Clinical Medicine, Shanghai Medical College, Fudan University, Shanghai, 200032, People's Republic of China
| | - Hong Lin
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Fudan University, Shanghai, 200032, People's Republic of China.,Department of Clinical Medicine, Shanghai Medical College, Fudan University, Shanghai, 200032, People's Republic of China
| | - Chengzhi Han
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Fudan University, Shanghai, 200032, People's Republic of China.,Department of Clinical Medicine, Shanghai Medical College, Fudan University, Shanghai, 200032, People's Republic of China
| | - Xinyi Qian
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Fudan University, Shanghai, 200032, People's Republic of China
| | - Yijun Huang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Fudan University, Shanghai, 200032, People's Republic of China
| | - Xiaorong Ren
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Fudan University, Shanghai, 200032, People's Republic of China
| | - Jiayuan Sun
- Department of Respiratory Endoscopy, Department of Respiratory and Critical Care Medicine, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, 200030, People's Republic of China
| | - Nana Feng
- Department of Respiratory and Critical Medicine, Shanghai Eighth People's Hospital Affiliated to Jiangsu University, Shanghai, People's Republic of China
| | - Chunmei Xia
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Fudan University, Shanghai, 200032, People's Republic of China
| | - Meng Shi
- Department of Thoracic and Cardiovascular Surgery, Huashan Hospital, Affiliated with Fudan University, Shanghai, 200040, People's Republic of China
| |
Collapse
|
46
|
Gao M, Zhang Z, Lai K, Deng Y, Zhao C, Lu Z, Geng Q. Puerarin: A protective drug against ischemia-reperfusion injury. Front Pharmacol 2022; 13:927611. [PMID: 36091830 PMCID: PMC9449408 DOI: 10.3389/fphar.2022.927611] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2022] [Accepted: 07/08/2022] [Indexed: 11/25/2022] Open
Abstract
Ischemia-reperfusion (I/R) is a pathological process that occurs in numerous organs throughout the human body and is frequently associated with severe cellular damage and death. Puerarin is an isoflavone compound extracted from the root of Pueraria lobata and has pharmacological effects such as dilating cerebral vessels and anti-free radical generation in cerebral ischemic tissues. With the deepening of experimental research and clinical research on puerarin, it has been found that puerarin has a protective effect on ischemia-reperfusion injury (IRI) of the heart, brain, spinal cord, lung, intestine and other organs. In summary, puerarin has a vast range of pharmacological effects and significant protective effects, and it also has obvious advantages in the clinical protection of patients with organ IRI. With the deepening of experimental pharmacological research and clinical research, it is expected to be an effective drug for IRI treatment. In this review, we summarize the current knowledge of the protective effect of puerarin on I/R organ injury and its possible underlying molecular mechanisms.
Collapse
Affiliation(s)
- Minglang Gao
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan, China
| | - Ziyao Zhang
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan, China
| | - Kai Lai
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan, China
| | - Yu Deng
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan, China
| | - Chuanbing Zhao
- Department of Pancreatic Surgery, Renmin Hospital of Wuhan University, Wuhan, China
| | - Zilong Lu
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan, China
| | - Qing Geng
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan, China
| |
Collapse
|
47
|
Phenolic Acids from Fructus Chebulae Immaturus Alleviate Intestinal Ischemia-Reperfusion Injury in Mice through the PPARα/NF-κB Pathway. Molecules 2022; 27:molecules27165227. [PMID: 36014464 PMCID: PMC9415796 DOI: 10.3390/molecules27165227] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2022] [Revised: 08/05/2022] [Accepted: 08/11/2022] [Indexed: 12/13/2022] Open
Abstract
Intestinal ischemia/reperfusion (II/R) injury is a common life-threatening complication with high morbidity and mortality. Chebulae Fructus Immaturus, the unripe fruit of Terminalia chebula Retz., also known as “Xiqingguo” or “Tibet Olive” in China, has been widely used in traditional Tibetan medicine throughout history. The phenolic acids’ extract of Chebulae Fructus Immaturus (XQG for short) has exhibited strong antioxidative, anti-inflammation, anti-apoptosis, and antibacterial activities. However, whether XQG can effectively ameliorate II/R injuries remains to be clarified. Our results showed that XQG could effectively alleviate II/R-induced intestinal morphological damage and intestinal barrier injury by decreasing the oxidative stress, inflammatory response, and cell death. Transcriptomic analysis further revealed that the main action mechanism of XQG protecting against II/R injury was involved in activating PPARα and inhibiting the NF-κB-signaling pathway. Our study suggests the potential usage of XQG as a new candidate to alleviate II/R injury.
Collapse
|
48
|
Targeting vascular inflammation through emerging methods and drug carriers. Adv Drug Deliv Rev 2022; 184:114180. [PMID: 35271986 PMCID: PMC9035126 DOI: 10.1016/j.addr.2022.114180] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2021] [Revised: 02/18/2022] [Accepted: 03/04/2022] [Indexed: 12/16/2022]
Abstract
Acute inflammation is a common dangerous component of pathogenesis of many prevalent conditions with high morbidity and mortality including sepsis, thrombosis, acute respiratory distress syndrome (ARDS), COVID-19, myocardial and cerebral ischemia-reperfusion, infection, and trauma. Inflammatory changes of the vasculature and blood mediate the course and outcome of the pathology in the tissue site of insult, remote organs and systemically. Endothelial cells lining the luminal surface of the vasculature play the key regulatory functions in the body, distinct under normal vs. pathological conditions. In theory, pharmacological interventions in the endothelial cells might enable therapeutic correction of the overzealous damaging pro-inflammatory and pro-thrombotic changes in the vasculature. However, current agents and drug delivery systems (DDS) have inadequate pharmacokinetics and lack the spatiotemporal precision of vascular delivery in the context of acute inflammation. To attain this level of precision, many groups design DDS targeted to specific endothelial surface determinants. These DDS are able to provide specificity for desired tissues, organs, cells, and sub-cellular compartments needed for a particular intervention. We provide a brief overview of endothelial determinants, design of DDS targeted to these molecules, their performance in experimental models with focus on animal studies and appraisal of emerging new approaches. Particular attention is paid to challenges and perspectives of targeted therapeutics and nanomedicine for advanced management of acute inflammation.
Collapse
|
49
|
Ex Vivo Lung Perfusion with β-Nicotinamide Adenine Dinucleotide (NAD+) Improves Ischemic Lung Function. Antioxidants (Basel) 2022; 11:antiox11050843. [PMID: 35624707 PMCID: PMC9137530 DOI: 10.3390/antiox11050843] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Revised: 04/20/2022] [Accepted: 04/20/2022] [Indexed: 12/31/2022] Open
Abstract
Ischemia-reperfusion injury compromises short- and long-term outcomes after lung transplantation. The scarce existing data on NAD+ suggest effects on hypoxia-induced vasoconstriction, on reactive oxygen species and on tampering inflammation. We exposed rat lungs to 14 h of cold ischemic storage and perfused them in a rat ex vivo lung perfusion (EVLP) system for 4 h. A control group (n = 6) was compared to groups receiving 100 µM (n = 6) or 200 µM NAD+ (n = 6) in the preservation solution and groups receiving 200 µM (n = 4) or 2000 µM (n = 6) NAD+ every 30 min in the perfusate, starting at 1 h of EVLP. Compared to the control, significant effects were only achieved in the 2000 µM NAD+ group. During the 4 h of EVLP, we monitored higher vascular flow, lower mean pulmonary arterial pressure and increased oxygenation capacity. Tissue inflammation estimated with the myeloperoxidase assay was lower in the 2000 µM NAD+ group. We observed higher levels of anti-inflammatory IL-10, higher anti-inflammatory IL-6/IL-10 ratios and lower levels of pro-inflammatory IL-12 and IL-18 as well as a trend of more anti-inflammatory IFNy in the 2000 µM NAD+ perfusate. In the bronchoalveolar lavage, the pro-inflammatory levels of IL-1α and IL-1β were lower in the 2000 µM NAD+ group. NAD+ administered during EVLP is a promising agent with both anti-inflammatory properties and the ability to improve ischemic lung function.
Collapse
|
50
|
Maeda A, Kogata S, Toyama C, Lo PC, Okamatsu C, Yamamoto R, Masahata K, Kamiyama M, Eguchi H, Watanabe M, Nagashima H, Okuyama H, Miyagawa S. The Innate Cellular Immune Response in Xenotransplantation. Front Immunol 2022; 13:858604. [PMID: 35418992 PMCID: PMC8995651 DOI: 10.3389/fimmu.2022.858604] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Accepted: 02/23/2022] [Indexed: 01/02/2023] Open
Abstract
Xenotransplantation is very attractive strategy for addressing the shortage of donors. While hyper acute rejection (HAR) caused by natural antibodies and complement has been well defined, this is not the case for innate cellular xenogeneic rejection. An increasing body of evidence suggests that innate cellular immune responses contribute to xenogeneic rejection. Various molecular incompatibilities between receptors and their ligands across different species typically have an impact on graft outcome. NK cells are activated by direct interaction as well as by antigen dependent cellular cytotoxicity (ADCC) mechanisms. Macrophages are activated through various mechanisms in xenogeneic conditions. Macrophages recognize CD47 as a "marker of self" through binding to SIRPα. A number of studies have shown that incompatibility of porcine CD47 against human SIRPα contributes to the rejection of xenogeneic target cells by macrophages. Neutrophils are an early responder cell that infiltrates xenogeneic grafts. It has also been reported that neutrophil extracellular traps (NETs) activate macrophages as damage-associated pattern molecules (DAMPs). In this review, we summarize recent insights into innate cellular xenogeneic rejection.
Collapse
Affiliation(s)
- Akira Maeda
- Department of Promotion for Blood and Marrow Transplantation, Aichi Medical University School of Medicine, Nagakute, Japan.,Department of Pediatric Surgery, Osaka University Graduate School of Medicine, Suita, Japan
| | - Shuhei Kogata
- Department of Pediatric Surgery, Osaka University Graduate School of Medicine, Suita, Japan
| | - Chiyoshi Toyama
- Department of Pediatric Surgery, Osaka University Graduate School of Medicine, Suita, Japan
| | - Pei-Chi Lo
- Department of Pediatric Surgery, Osaka University Graduate School of Medicine, Suita, Japan
| | - Chizu Okamatsu
- Department of Pediatric Surgery, Osaka University Graduate School of Medicine, Suita, Japan
| | - Riho Yamamoto
- Department of Pediatric Surgery, Osaka University Graduate School of Medicine, Suita, Japan
| | - Kazunori Masahata
- Department of Pediatric Surgery, Osaka University Graduate School of Medicine, Suita, Japan
| | - Masafumi Kamiyama
- Department of Pediatric Surgery, Osaka University Graduate School of Medicine, Suita, Japan
| | - Hiroshi Eguchi
- Department of Pediatric Surgery, Osaka University Graduate School of Medicine, Suita, Japan
| | - Masahito Watanabe
- International Institute for Bio-Resource Research, Meiji University, Kawasaki, Japan
| | - Hiroshi Nagashima
- International Institute for Bio-Resource Research, Meiji University, Kawasaki, Japan
| | - Hiroomi Okuyama
- Department of Pediatric Surgery, Osaka University Graduate School of Medicine, Suita, Japan
| | - Shuji Miyagawa
- Department of Pediatric Surgery, Osaka University Graduate School of Medicine, Suita, Japan.,International Institute for Bio-Resource Research, Meiji University, Kawasaki, Japan
| |
Collapse
|