1
|
Afsar B, Afsar RE, Caliskan Y, Lentine KL. Mineralocorticoid receptor blockage in kidney transplantation: too much of a good thing or not? Int Urol Nephrol 2024:10.1007/s11255-024-04256-6. [PMID: 39470940 DOI: 10.1007/s11255-024-04256-6] [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: 07/03/2024] [Accepted: 10/21/2024] [Indexed: 11/01/2024]
Abstract
Although, kidney transplantation (KT) is the best treatment option for patients with end-stage kidney disease, long-term complications including chronic kidney allograft disease (CKAD) and major adverse cardiovascular events (MACE) are common. To decrease these complications new therapeutic options are necessary. Mineralocorticoid receptor antagonists (MRAs) are one of the promising drugs in this context. In the general population, MRAs had favorable effects on blood pressure regulation, MACE, proteinuria and progression of chronic kidney disease. In the context of KT, there are limited studies showing beneficial effects such as reducing proteinuria and oxidative stress. In this review, we performed a narrative review to assess the use and impact of MRAs in kidney transplant recipients. We found that in KTRs, MRAs are safe and they have favorable or neutral impact on blood pressure, glomerular filtration rate, urinary protein/albumin excretion, and oxidative stress. No data was found regarding major cardiovascular adverse events.
Collapse
Affiliation(s)
- Baris Afsar
- School of Medicine, Division of Nephrology, Saint Louis University, SSM Health Saint Louis University Hospital, St. Louis, MO, USA.
| | - Rengin Elsurer Afsar
- School of Medicine, Division of Nephrology, Saint Louis University, SSM Health Saint Louis University Hospital, St. Louis, MO, USA
| | - Yasar Caliskan
- School of Medicine, Division of Nephrology, Saint Louis University, SSM Health Saint Louis University Hospital, St. Louis, MO, USA
| | - Krista L Lentine
- School of Medicine, Division of Nephrology, Saint Louis University, SSM Health Saint Louis University Hospital, St. Louis, MO, USA
| |
Collapse
|
2
|
Izemrane D, Benziane A, Makrelouf M, Hamdis N, Rabia SH, Boudjellaba S, Baz A, Benaziza D. Living donors kidney transplantation and oxidative stress: Nitric oxide as a predictive marker of graft function. PLoS One 2024; 19:e0307824. [PMID: 39312562 PMCID: PMC11419388 DOI: 10.1371/journal.pone.0307824] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2023] [Accepted: 07/10/2024] [Indexed: 09/25/2024] Open
Abstract
BACKGROUND Glomerular filtration rate is the best indicator of renal function and a predictor of graft and patient survival after kidney transplantation. METHODS In a single-centre prospective analysis, we assessed the predictive performances of 4 oxidative stress biomarkers in estimating graft function at 6 months and 1 year after kidney transplantation from living donors. Blood samples were achieved on days (D-1, D1, D2, D3, D6 and D8), months (M1, M3 and M6) and after one year (1Y). For donors, a blood sample was collected on D-1. Malondialdehyde (MDA), nitric oxide (NO), glutathione s-transferase (GST), myeloperoxydase (MPO), and creatinine (Cr) were measured by spectrophotometric essays. The estimated glomerular filtration rate by the modification of diet in renal disease equation (MDRD-eGFR) was used to assess renal function in 32 consecutive donor-recipient pairs. Pearson's and Spearman's correlations have been applied to filter out variables and covariables that can be used to build predictive models of graft function at six months and one year. The predictive performances of NO and MPO were tested by multivariable stepwise linear regression to estimate glomerular filtration rate at six months. RESULTS Three models with the highest coefficients of determination stand out, combining the two variables nitric oxide at day 6 and an MDRD-eGFR variable at day 6 or MDRD-eGFR at day 21 or MDRD-eGFR at 3 months, associated for the first two models or not for the third model with donor age as a covariable (P = 0.000, r2 = 0.599, r2adj = 0.549; P = 0.000, r2 = 0.548, r2adj = 0.497; P = 0.000, r2 = 0.553, r2adj = 0.517 respectively). CONCLUSION Quantification of nitric oxide at day six could be useful in predicting graft function at six months in association with donor age and the estimated glomerular filtration rate in recipient at day 6, day 21 and 3 months after transplantation.
Collapse
Affiliation(s)
- Djamila Izemrane
- Laboratory of Biology and Animal Physiology, Higher Normal School, Kouba, Algiers, Algeria
- National Higher Veterinary School, Issad Abbes, Oued Smar, Algiers, Algeria
| | - Ali Benziane
- Department of Nephrology-Hemodialysis and Transplantation, Lamine Debaghine University Hospital, Bab El Oued, Algiers, Algeria
| | - Mohamed Makrelouf
- Central Biology Laboratory, Lamine Debaghine University Hospital, Bab El Oued, Algiers, Algeria
| | - Nacim Hamdis
- Laboratory of Food Technology Research, Faculty of Engineering Sciences-University M’Hamed Bougara, City Frantz Fanon, Boumerdes, Algeria
| | - Samia Hadj Rabia
- Laboratory of Biology and Animal Physiology, Higher Normal School, Kouba, Algiers, Algeria
- Department of Nuclear Applications, Nuclear Research Center, Sebala, Algiers, Algeria
| | - Sofiane Boudjellaba
- National Higher Veterinary School, Issad Abbes, Oued Smar, Algiers, Algeria
- Laboratory of Research Management of Local Animal Resources (GRAL), National Higher Veterinary School, Issad Abbes, Oued Smar, Algiers, Algeria
| | - Ahsene Baz
- Laboratory of Biology and Animal Physiology, Higher Normal School, Kouba, Algiers, Algeria
| | - Djamila Benaziza
- Laboratory of Biology and Animal Physiology, Higher Normal School, Kouba, Algiers, Algeria
| |
Collapse
|
3
|
Chen H, Buzdar JA, Riaz R, Fouad D, Ahmed N, Shah QA, Chen S. Bovine lactoferrin alleviates aflatoxin B1 induced hepatic and renal injury in broilers by mediating Nrf2 signaling pathway. Poult Sci 2024; 103:104316. [PMID: 39383667 PMCID: PMC11492589 DOI: 10.1016/j.psj.2024.104316] [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: 07/10/2024] [Revised: 09/03/2024] [Accepted: 09/05/2024] [Indexed: 10/11/2024] Open
Abstract
Aflatoxin B1 (AFB1) a mycotoxin found in chicken feed that possess a global hazard to poultry health. However different potent compounds like bovine lactoferrin (bLF) may prove to be protective effects against AFB1. This study aims to explore the protective effect of bLF against AFB1-induced injury in the liver and kidney in broiler. For this purpose, 600 broilers chicks were randomly alienated into 5 groups (n = 120 each): negative control; positive control (3 mg/kg AFB1), and bLF high, medium, and low dosage groups (600 mg/kg, 300 mg/kg, and 150 mg/kg, respectively). The results highlight that AFB1 toxicity in birds exhibited low feed intake, reduction in weight gain, and a decrease in FCR while, bLF regulated these adverse effects. Meanwhile, AFB1 group showed higher levels of alanine transaminase (ALT) and aspartate aminotransferase (AST) and lower levels of superoxide dismutase (SOD) and glutathione (GSHpx) in liver, while urea and creatinine were decline in kidney. Supplementation with bLF effectively controlled these biomarkers and control the negative effects of toxicity. Furthermore, hematoxylin and eosin (H&E) staining exhibited normal morphological structures within liver and kidney in the bLF treated groups, while degenerative changes were observed in AFB1 group. Similarly, bLF, decreased oxidative stress and thus prevented apoptosis in the liver and kidney cells of the birds. Whereas, mRNA level of mitochondrial apoptosis related gene including Bcl-2 (Bak and Bax), caspase-3 and caspase-9 was upregulated, while bcl2 gene were downregulated in AFB1 group. Dietary supplementation of bLF effectively normalizes the expression of these genes. AFB1 exposed birds shown to decrease gene expression level of the crucial component of Nrf2 pathway, responsible to regulate antioxidant defense. Interestingly, bLF reverse these detrimental effects of and restore the normal expression levels of Nrf2 pathway. Conclusively, our findings demonstrate that bLF mitigates the detrimental effects of AFB1, besides regulation of the apoptosis-related genes via mitochondrial pathways. These findings validate that the bLF (600 mg/kg) could be used as protective agent against AFB1-induced liver and kidney damage.
Collapse
Affiliation(s)
- Hong Chen
- Department of Basic Veterinary Medicine, College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi Province, 712100, China
| | - Jameel Ahmed Buzdar
- Department of Basic Veterinary Science, Faculty of Veterinary & Animal Science, Lasbela University of Agriculture, Water and Marine Sciences, Uthal, 90150, Baluchistan, Pakistan
| | - Roshan Riaz
- Department of Animal Nutrition and Nutritional Diseases, Faculty of Veterinary Medicine, Kafkas University, Kars, 36100, Türkiye
| | - Dalia Fouad
- Zoology Department, College of Science, King Saud University, 11451, Riyadh, Saudi Arabia
| | - Nisar Ahmed
- Department of Basic Veterinary Science, Faculty of Veterinary & Animal Science, Lasbela University of Agriculture, Water and Marine Sciences, Uthal, 90150, Baluchistan, Pakistan
| | - Qurban Ali Shah
- Department of Basic Veterinary Science, Faculty of Veterinary & Animal Science, Lasbela University of Agriculture, Water and Marine Sciences, Uthal, 90150, Baluchistan, Pakistan
| | - Shulin Chen
- Department of Basic Veterinary Medicine, College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi Province, 712100, China.
| |
Collapse
|
4
|
Wang J, Wang J, Lu C, Wang Y, Bi H, Zheng J, Ding X. ISL1-overexpressing BMSCs attenuate renal ischemia-reperfusion injury by suppressing apoptosis and oxidative stress through the paracrine action. Cell Mol Life Sci 2024; 81:312. [PMID: 39066917 PMCID: PMC11335236 DOI: 10.1007/s00018-024-05354-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2024] [Revised: 06/29/2024] [Accepted: 07/07/2024] [Indexed: 07/30/2024]
Abstract
Ischemia-reperfusion injury (IRI) is a major event in renal transplantation, leading to adverse outcomes. Bone marrow mesenchymal stem cells (BMSCs) are novel promising therapeutics for repairing kidney injuries. The therapeutic efficacy of BMSCs with ISL1 overexpression in renal IRI and its underlying mechanism need to be investigated. The unilateral renal IRI rat model was established to mimic clinical acute kidney injury. Rats were injected with PBS, BMSCs-Scrambled or BMSCs-ISL1 via the tail vein at the timepoint of reperfusion, and then sacrificed after 24 h of reperfusion. The administration of BMSCs-ISL1 significantly improved renal function, inhibited tubular cells apoptosis, inflammation, oxidative stress in rats. In vitro, HKC cells subjected to H2O2 stimulation were pretreated with the conditioned medium (CM) of BMSCs-Scrambled or BMSCs-ISL1. The pretreatment of ISL1-CM attenuated apoptosis and oxidative stress induced by H2O2 in HKC cells. Our proteomic data suggested that haptoglobin (Hp) was one of the secretory proteins in ISL1-CM. Subsequent experiments confirmed that Hp was the important paracrine factor from BMSCs-ISL1 that exerted anti-apoptotic and antioxidant functions. Mechanistically, Hp played a cytoprotective role via the inhibition of ERK signaling pathway, which could be abrogated by Ro 67-7476, the ERK phosphorylation agonist. The results suggested that paracrine action may be the main mechanism for BMSCs-ISL1 to exert protective effects. As an important anti-apoptotic and antioxidant factor in ISL1-CM, Hp may serve as a new therapeutic agent for treating IRI, providing new insights for overcoming the long-term adverse effects of stem cell therapy.
Collapse
Affiliation(s)
- Jiale Wang
- Department of Renal Transplantation, Hospital of Nephrology, The First Affiliated Hospital of Xi'an Jiaotong University, 277 Yanta Western Rd, Xi'an, Shaanxi, 710061, China
| | - Jingwen Wang
- Department of Renal Transplantation, Hospital of Nephrology, The First Affiliated Hospital of Xi'an Jiaotong University, 277 Yanta Western Rd, Xi'an, Shaanxi, 710061, China
| | - Cuinan Lu
- Department of Renal Transplantation, Hospital of Nephrology, The First Affiliated Hospital of Xi'an Jiaotong University, 277 Yanta Western Rd, Xi'an, Shaanxi, 710061, China
| | - Ying Wang
- Department of Renal Transplantation, Hospital of Nephrology, The First Affiliated Hospital of Xi'an Jiaotong University, 277 Yanta Western Rd, Xi'an, Shaanxi, 710061, China
| | - Huanjing Bi
- Department of Renal Transplantation, Hospital of Nephrology, The First Affiliated Hospital of Xi'an Jiaotong University, 277 Yanta Western Rd, Xi'an, Shaanxi, 710061, China
| | - Jin Zheng
- Department of Renal Transplantation, Hospital of Nephrology, The First Affiliated Hospital of Xi'an Jiaotong University, 277 Yanta Western Rd, Xi'an, Shaanxi, 710061, China
| | - Xiaoming Ding
- Department of Renal Transplantation, Hospital of Nephrology, The First Affiliated Hospital of Xi'an Jiaotong University, 277 Yanta Western Rd, Xi'an, Shaanxi, 710061, China.
| |
Collapse
|
5
|
Wang H, Li Y, Cao X, Niu H, Li X, Wang J, Yang J, Xu C, Wang H, Wan S, Li K, Fu S, Yang L. MELATONIN ATTENUATES RENAL ISCHEMIA-REPERFUSION INJURY BY REGULATING MITOCHONDRIAL DYNAMICS AND AUTOPHAGY THROUGH AMPK/DRP1. Shock 2024; 62:74-84. [PMID: 38713551 DOI: 10.1097/shk.0000000000002330] [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: 05/09/2024]
Abstract
ABSTRACT Ischemia-reperfusion injury (IRI) often stems from an imbalance between mitochondrial dynamics and autophagy. Melatonin mitigates IRI by regulating mitochondrial dynamics. However, the precise molecular mechanism underlying the role of melatonin in reducing IRI through modulating mitochondrial dynamics remains elusive. The objective of this study was to investigate whether pretreatment with melatonin before IRI confers protective effects by modulating mitochondrial dynamics and mitophagy. Melatonin pretreatment was administered to HK-2 cells and live rats before subjecting them to hypoxia-reoxygenation or IRI, respectively. Cells and rat kidney models were evaluated for markers of oxidative stress, autophagy, mitochondrial dynamics, and the expression of adenosine 5'-monophosphate (AMP)-activated protein kinase (AMPK) and phospho-AMPKα (P-AMPK). After renal IRI, increased mitochondrial fission and autophagy were observed, accompanied by exacerbated cellular oxidative stress injury and aggravated mitochondrial dysfunction. Nevertheless, melatonin pretreatment inhibited mitochondrial fission, promoted mitochondrial fusion, and attenuated autophagy levels. This intervention was correlated with a notable reduction in oxidative stress injury and remarkable restoration of mitochondrial functionality. Ischemia-reperfusion injury led to a decline in P-AMPK levels, whereas melatonin pretreatment increased the level of P-AMPK levels. Silencing AMPK with small interfering RNA exacerbated mitochondrial damage, and in this context, melatonin pretreatment did not alleviate mitochondrial fission or autophagy levels but resulted in sustained oxidative stress damage. Collectively, these findings indicate that melatonin pretreatment shields the kidneys from IRI by mitigating excessive mitochondrial fission, moderating autophagy levels, and preserving appropriate mitochondrial fission, all in an AMPK-dependent manner.
Collapse
Affiliation(s)
- Huabin Wang
- Department of Urology, Institute of Urology, Gansu Urological Clinical Center, Lanzhou University Second Hospital, Lanzhou, China
| | - Yi Li
- Department of Anesthesiology, Lanzhou University Second Hospital, Lanzhou, China
| | - Xichao Cao
- The Second Clinical Medical College of Lanzhou University, Lanzhou, China
| | - Heping Niu
- Department of Urology, Institute of Urology, Gansu Urological Clinical Center, Lanzhou University Second Hospital, Lanzhou, China
| | - Xiaoran Li
- Department of Urology, Institute of Urology, Gansu Urological Clinical Center, Lanzhou University Second Hospital, Lanzhou, China
| | - Jirong Wang
- Department of Urology, Institute of Urology, Gansu Urological Clinical Center, Lanzhou University Second Hospital, Lanzhou, China
| | - Jianwei Yang
- Department of Urology, Institute of Urology, Gansu Urological Clinical Center, Lanzhou University Second Hospital, Lanzhou, China
| | - Changhong Xu
- Department of Urology, Institute of Urology, Gansu Urological Clinical Center, Lanzhou University Second Hospital, Lanzhou, China
| | - Hailong Wang
- Department of Urology, Institute of Urology, Gansu Urological Clinical Center, Lanzhou University Second Hospital, Lanzhou, China
| | - Shun Wan
- Department of Urology, Institute of Urology, Gansu Urological Clinical Center, Lanzhou University Second Hospital, Lanzhou, China
| | - Kunpeng Li
- Department of Urology, Institute of Urology, Gansu Urological Clinical Center, Lanzhou University Second Hospital, Lanzhou, China
| | - Shengjun Fu
- Department of Urology, Institute of Urology, Gansu Urological Clinical Center, Lanzhou University Second Hospital, Lanzhou, China
| | - Li Yang
- Department of Urology, Institute of Urology, Gansu Urological Clinical Center, Lanzhou University Second Hospital, Lanzhou, China
| |
Collapse
|
6
|
Cao W, Zhang J, Yu S, Gan X, An R. N-acetylcysteine regulates oxalate induced injury of renal tubular epithelial cells through CDKN2B/TGF-β/SMAD axis. Urolithiasis 2024; 52:46. [PMID: 38520518 DOI: 10.1007/s00240-023-01527-2] [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: 07/25/2023] [Accepted: 12/26/2023] [Indexed: 03/25/2024]
Abstract
This study was aimed to investigate the preventive effects of N-acetyl-L-cysteine (NAC) against renal tubular cell injury induced by oxalate and stone formation and further explore the related mechanism. Transcriptome sequencing combined with bioinformatics analysis were performed to identify differentially expressed gene (DEG) and related pathways. HK-2 cells were pretreated with or without antioxidant NAC/with or silencing DEG before exposed to sodium oxalate. Then, the cell viability, oxidative biomarkers of superoxidase dismutase (SOD) and malondialdehyde (MDA), apoptosis and cell cycle were measured through CCK8, ELISA and flow cytometry assay, respectively. Male SD rats were separated into control group, hyperoxaluria (HOx) group, NAC intervention group, and TGF-β/SMAD pathway inhibitor group. After treatment, the structure changes and oxidative stress and CaOx crystals deposition were evaluated in renal tissues by H&E staining, immunohistochemical and Pizzolato method. The expression of TGF-β/SMAD pathway related proteins (TGF-β1, SMAD3 and SMAD7) were determined by Western blot in vivo and in vitro. CDKN2B is a DEG screened by transcriptome sequencing combined with bioinformatics analysis, and verified by qRT-PCR. Sodium oxalate induced declined HK-2 cell viability, in parallel with inhibited cellular oxidative stress and apoptosis. The changes induced by oxalate in HK-2 cells were significantly reversed by NAC treatment or the silencing of CDKN2B. The cell structure damage and CaOx crystals deposition were observed in kidney tissues of HOx group. Meanwhile, the expression levels of SOD and 8-OHdG were detected in kidney tissues of HOx group. The changes induced by oxalate in kidney tissues were significantly reversed by NAC treatment. Besides, expression of SMAD7 was significantly down-regulated, while TGF-β1 and SMAD3 were accumulated induced by oxalate in vitro and in vivo. The expression levels of TGF-β/SMAD pathway related proteins induced by oxalate were reversed by NAC. In conclusion, we found that NAC could play an anti-calculus role by mediating CDKN2B/TGF-β/SMAD axis.
Collapse
Affiliation(s)
- Wei Cao
- Department of Urology, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, 150081, China
| | - Jingbo Zhang
- Department of Urology, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, 150081, China
| | - Shiliang Yu
- Department of Urology, The First Affiliated Hospital of Harbin Medical University, 23 YouZheng Street, HarbinHarbin, Heilongjiang, 150001, China
| | - Xiuguo Gan
- Department of Urology, The First Affiliated Hospital of Harbin Medical University, 23 YouZheng Street, HarbinHarbin, Heilongjiang, 150001, China
| | - Ruihua An
- Department of Urology, The First Affiliated Hospital of Harbin Medical University, 23 YouZheng Street, HarbinHarbin, Heilongjiang, 150001, China.
| |
Collapse
|
7
|
Zhang CY, Yang M. Anti-oxidative stress treatment and current clinical trials. World J Hepatol 2024; 16:294-299. [PMID: 38495278 PMCID: PMC10941751 DOI: 10.4254/wjh.v16.i2.294] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/02/2023] [Revised: 01/08/2024] [Accepted: 02/05/2024] [Indexed: 02/27/2024] Open
Abstract
Oxidative stress disturbs the balance between the production of reactive oxygen species (ROS) and the detoxification biological process. It plays an important role in the development and progression of many chronic diseases. Upon exposure to oxidative stress or the inducers of ROS, the cellular nucleus undergoes some biological processes via different signaling pathways, such as stress adaption through the forkhead box O signaling pathway, inflammatory response through the IκB kinase/nuclear factor-κB signaling pathway, hypoxic response via the hypoxia-inducible factor/prolyl hydroxylase domain proteins pathway, DNA repair or apoptosis through the p53 signaling pathway, and antioxidant response through the Kelch-like ECH-associated protein 1/nuclear factor E2-related factor 2 signaling pathway. These processes are involved in many diseases. Therefore, oxidative stress has gained more attraction as a targeting process for disease treatment. Meanwhile, anti-oxidative stress agents have been widely explored in pre-clinical trials. However, only limited clinical trials are performed to evaluate the efficacy of anti-oxidative stress agents or antioxidants in diseases. In this letter, we further discuss the current clinical trials related to anti-oxidative stress treatment in different diseases. More pre-clinical studies and clinical trials are expected to use anti-oxidative stress strategies as disease treatment or dietary supplementation to improve disease treatment outcomes.
Collapse
Affiliation(s)
- Chun-Ye Zhang
- Bond Life Sciences Center, University of Missouri, Columbia, MO 65211, United States
| | - Ming Yang
- Department of Surgery, University of Missouri, Columbia, MO 65212, United States.
| |
Collapse
|
8
|
Szentimrei R, Lőrincz H, Szentpéteri A, Varga VE, Seres I, Varga É, Nemes B, Harangi M, Paragh G. Assessment of amino-terminal C-type natriuretic peptide serum level and its correlation with high-density lipoprotein structure and function in patients with end stage renal disease before and after kidney transplantation. Chem Biol Interact 2023; 385:110749. [PMID: 37802408 DOI: 10.1016/j.cbi.2023.110749] [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: 07/14/2023] [Revised: 09/28/2023] [Accepted: 10/04/2023] [Indexed: 10/10/2023]
Abstract
We aimed to investigate serum amino-terminal C-type natriuretic peptide (NT-proCNP) and its relationship with quantitative and qualitative HDL-parameters in patients with end-stage renal disease (ESRD) before, then 1 and 6 months after kidney transplantation (TX). Seventy patients (47 males, 23 females, mean age 51.7 ± 12.4 years) were enrolled in a prospective follow-up study. We examined serum creatinine, C-reactive protein, procalcitonin, fasting glucose and lipid parameters before, then 1 and 6 months after TX. High-density lipoprotein- (HDL)-associated paraoxonase-1 (PON1) paraoxonase and arylesterase activities were measured spectrophotometrically. Lipoprotein subfractions were determined by Lipoprint. NT-proCNP and oxidized low-density lipoprotein (oxLDL) levels were measured by ELISA. Mean NT-proCNP was 45.8 ± 21.9 pmol/L before renal transplantation and decreased markedly 1 month and 6 months after transplantation (5.3 ± 2.5 and 7.7 ± 4.9 pmol/L, respectively, P = 1 × 10-4). During the 6 months' follow-up, PON1 arylesterase, paraoxonase and salt-stimulated paraoxonase activities improved. NT-proCNP positively correlated with procalcitonin and creatinine and negatively with GFR, LDL-cholesterol (LDL-C) and HDL-cholesterol (HDL-C). There was a negative correlation between serum NT-proCNP and PON1 arylesterase activity. According to the multiple regression analysis, the best predicting variables of NT-proCNP were serum procalcitonin, creatinine and PON1 arylesterase activity. NT-proCNP might be a novel link between HDL dysfunction and impaired vascular function in ESRD, but not after kidney transplantation. Further studies in larger populations are needed to clarify the exact role of NT-proCNP in the risk prediction for cardiovascular comorbidities and complications in ESRD.
Collapse
Affiliation(s)
- Réka Szentimrei
- Division of Metabolic Diseases, Department of Internal Medicine, Faculty of Medicine, University of Debrecen, Debrecen, Hungary; Doctoral School of Health Sciences, Faculty of Medicine, University of Debrecen, Debrecen, Hungary.
| | - Hajnalka Lőrincz
- Division of Metabolic Diseases, Department of Internal Medicine, Faculty of Medicine, University of Debrecen, Debrecen, Hungary.
| | - Anita Szentpéteri
- Division of Metabolic Diseases, Department of Internal Medicine, Faculty of Medicine, University of Debrecen, Debrecen, Hungary.
| | - Viktória Evelin Varga
- Division of Metabolic Diseases, Department of Internal Medicine, Faculty of Medicine, University of Debrecen, Debrecen, Hungary.
| | - Ildikó Seres
- Division of Metabolic Diseases, Department of Internal Medicine, Faculty of Medicine, University of Debrecen, Debrecen, Hungary.
| | - Éva Varga
- Department of Internal Medicine and Hematology, Semmelweis University, Budapest, Hungary.
| | - Balázs Nemes
- Institute of Surgery, Department of Organ Transplantation, University of Debrecen, Debrecen, Hungary.
| | - Mariann Harangi
- Division of Metabolic Diseases, Department of Internal Medicine, Faculty of Medicine, University of Debrecen, Debrecen, Hungary.
| | - György Paragh
- Division of Metabolic Diseases, Department of Internal Medicine, Faculty of Medicine, University of Debrecen, Debrecen, Hungary.
| |
Collapse
|
9
|
Piko N, Bevc S, Hojs R, Ekart R. The Role of Oxidative Stress in Kidney Injury. Antioxidants (Basel) 2023; 12:1772. [PMID: 37760075 PMCID: PMC10525550 DOI: 10.3390/antiox12091772] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2023] [Revised: 09/13/2023] [Accepted: 09/15/2023] [Indexed: 09/29/2023] Open
Abstract
Acute kidney injury and chronic kidney disease are among the most common non-communicable diseases in the developed world, with increasing prevalence. Patients with acute kidney injury are at an increased risk of developing chronic kidney disease. One of kidney injury's most common clinical sequelae is increased cardiovascular morbidity and mortality. In recent years, new insights into the pathophysiology of renal damage have been made. Oxidative stress is the imbalance favoring the increased generation of ROS and/or reduced body's innate antioxidant defense mechanisms and is of pivotal importance, not only in the development and progression of kidney disease but also in understanding the enhanced cardiovascular risk in these patients. This article summarizes and emphasizes the role of oxidative stress in acute kidney injury, various forms of chronic kidney disease, and also in patients on renal replacement therapy (hemodialysis, peritoneal dialysis, and after kidney transplant). Additionally, the role of oxidative stress in the development of drug-related nephrotoxicity and also in the development after exposure to various environmental and occupational pollutants is presented.
Collapse
Affiliation(s)
- Nejc Piko
- Department of Dialysis, Clinic for Internal Medicine, University Medical Centre, 2000 Maribor, Slovenia;
| | - Sebastjan Bevc
- Department of Nephrology, Clinic for Internal Medicine, University Medical Centre, 2000 Maribor, Slovenia; (S.B.); (R.H.)
- Medical Faculty, University of Maribor, 2000 Maribor, Slovenia
| | - Radovan Hojs
- Department of Nephrology, Clinic for Internal Medicine, University Medical Centre, 2000 Maribor, Slovenia; (S.B.); (R.H.)
- Medical Faculty, University of Maribor, 2000 Maribor, Slovenia
| | - Robert Ekart
- Department of Dialysis, Clinic for Internal Medicine, University Medical Centre, 2000 Maribor, Slovenia;
- Medical Faculty, University of Maribor, 2000 Maribor, Slovenia
| |
Collapse
|
10
|
Nanodrugs alleviate acute kidney injury: Manipulate RONS at kidney. Bioact Mater 2023; 22:141-167. [PMID: 36203963 PMCID: PMC9526023 DOI: 10.1016/j.bioactmat.2022.09.021] [Citation(s) in RCA: 38] [Impact Index Per Article: 38.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Revised: 08/12/2022] [Accepted: 09/19/2022] [Indexed: 02/06/2023] Open
Abstract
Currently, there are no clinical drugs available to treat acute kidney injury (AKI). Given the high prevalence and high mortality rate of AKI, the development of drugs to effectively treat AKI is a huge unmet medical need and a research hotspot. Although existing evidence fully demonstrates that reactive oxygen and nitrogen species (RONS) burst at the AKI site is a major contributor to AKI progression, the heterogeneity, complexity, and unique physiological structure of the kidney make most antioxidant and anti-inflammatory small molecule drugs ineffective because of the lack of kidney targeting and side effects. Recently, nanodrugs with intrinsic kidney targeting through the control of size, shape, and surface properties have opened exciting prospects for the treatment of AKI. Many antioxidant nanodrugs have emerged to address the limitations of current AKI treatments. In this review, we systematically summarized for the first time about the emerging nanodrugs that exploit the pathological and physiological features of the kidney to overcome the limitations of traditional small-molecule drugs to achieve high AKI efficacy. First, we analyzed the pathological structural characteristics of AKI and the main pathological mechanism of AKI: hypoxia, harmful substance accumulation-induced RONS burst at the renal site despite the multifactorial initiation and heterogeneity of AKI. Subsequently, we introduced the strategies used to improve renal targeting and reviewed advances of nanodrugs for AKI: nano-RONS-sacrificial agents, antioxidant nanozymes, and nanocarriers for antioxidants and anti-inflammatory drugs. These nanodrugs have demonstrated excellent therapeutic effects, such as greatly reducing oxidative stress damage, restoring renal function, and low side effects. Finally, we discussed the challenges and future directions for translating nanodrugs into clinical AKI treatment. AKI is a common clinical acute syndrome with high morbidity and mortality but without effective clinical drug available. Hypoxia and accumulation of toxic substances are key pathological features of various heterogeneous AKI. Excessive RONS is the core of the pathological mechanism of AKI. The development of nanodrugs is expected to achieve successful treatment in AKI.
Collapse
|
11
|
Melis N, Rubera I, Giraud S, Cougnon M, Duranton C, Poet M, Jarretou G, Thuillier R, Counillon L, Hauet T, Pellerin L, Tauc M, Pisani DF. Renal Ischemia Tolerance Mediated by eIF5A Hypusination Inhibition Is Regulated by a Specific Modulation of the Endoplasmic Reticulum Stress. Cells 2023; 12:cells12030409. [PMID: 36766751 PMCID: PMC9913814 DOI: 10.3390/cells12030409] [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: 12/06/2022] [Revised: 01/18/2023] [Accepted: 01/21/2023] [Indexed: 01/27/2023] Open
Abstract
Through kidney transplantation, ischemia/reperfusion is known to induce tissular injury due to cell energy shortage, oxidative stress, and endoplasmic reticulum (ER) stress. ER stress stems from an accumulation of unfolded or misfolded proteins in the lumen of ER, resulting in the unfolded protein response (UPR). Adaptive UPR pathways can either restore protein homeostasis or can turn into a stress pathway leading to apoptosis. We have demonstrated that N1-guanyl-1,7-diamineoheptane (GC7), a specific inhibitor of eukaryotic Initiation Factor 5A (eIF5A) hypusination, confers an ischemic protection of kidney cells by tuning their metabolism and decreasing oxidative stress, but its role on ER stress was unknown. To explore this, we used kidney cells pretreated with GC7 and submitted to either warm or cold anoxia. GC7 pretreatment promoted cell survival in an anoxic environment concomitantly to an increase in xbp1 splicing and BiP level while eiF2α phosphorylation and ATF6 nuclear level decreased. These demonstrated a specific modulation of UPR pathways. Interestingly, the pharmacological inhibition of xbp1 splicing reversed the protective effect of GC7 against anoxia. Our results demonstrated that eIF5A hypusination inhibition modulates distinctive UPR pathways, a crucial mechanism for the protection against anoxia/reoxygenation.
Collapse
Affiliation(s)
- Nicolas Melis
- Laboratory of Cellular and Molecular Biology, Center for Cancer Research, National Cancer Institute, Bethesda, MD 20892, USA
| | - Isabelle Rubera
- Université Côte d’Azur, CNRS, LP2M, 06108 Nice, France
- Laboratories of Excellence Ion Channel Science and Therapeutics, 06103 Nice, France
| | - Sebastien Giraud
- INSERM U1313, IRMETIST, Université de Poitiers et CHU de Poitiers, 86000 Poitiers, France
| | - Marc Cougnon
- Université Côte d’Azur, CNRS, LP2M, 06108 Nice, France
- Laboratories of Excellence Ion Channel Science and Therapeutics, 06103 Nice, France
| | - Christophe Duranton
- Université Côte d’Azur, CNRS, LP2M, 06108 Nice, France
- Laboratories of Excellence Ion Channel Science and Therapeutics, 06103 Nice, France
| | - Mallorie Poet
- Université Côte d’Azur, CNRS, LP2M, 06108 Nice, France
- Laboratories of Excellence Ion Channel Science and Therapeutics, 06103 Nice, France
| | - Gisèle Jarretou
- Université Côte d’Azur, CNRS, LP2M, 06108 Nice, France
- Laboratories of Excellence Ion Channel Science and Therapeutics, 06103 Nice, France
| | - Raphaël Thuillier
- INSERM U1313, IRMETIST, Université de Poitiers et CHU de Poitiers, 86000 Poitiers, France
| | - Laurent Counillon
- Université Côte d’Azur, CNRS, LP2M, 06108 Nice, France
- Laboratories of Excellence Ion Channel Science and Therapeutics, 06103 Nice, France
| | - Thierry Hauet
- INSERM U1313, IRMETIST, Université de Poitiers et CHU de Poitiers, 86000 Poitiers, France
| | - Luc Pellerin
- INSERM U1313, IRMETIST, Université de Poitiers et CHU de Poitiers, 86000 Poitiers, France
| | - Michel Tauc
- Université Côte d’Azur, CNRS, LP2M, 06108 Nice, France
- Laboratories of Excellence Ion Channel Science and Therapeutics, 06103 Nice, France
| | - Didier F. Pisani
- Université Côte d’Azur, CNRS, LP2M, 06108 Nice, France
- Laboratories of Excellence Ion Channel Science and Therapeutics, 06103 Nice, France
- Correspondence:
| |
Collapse
|
12
|
Simona MS, Alessandra V, Emanuela C, Elena T, Michela M, Fulvia G, Vincenzo S, Ilaria B, Federica M, Eloisa A, Massimo A, Maristella G. Evaluation of Oxidative Stress and Metabolic Profile in a Preclinical Kidney Transplantation Model According to Different Preservation Modalities. Int J Mol Sci 2023; 24:ijms24021029. [PMID: 36674540 PMCID: PMC9861050 DOI: 10.3390/ijms24021029] [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: 11/30/2022] [Revised: 01/02/2023] [Accepted: 01/03/2023] [Indexed: 01/07/2023] Open
Abstract
This study addresses a joint nuclear magnetic resonance (NMR) and electron paramagnetic resonance (EPR) spectroscopy approach to provide a platform for dynamic assessment of kidney viability and metabolism. On porcine kidney models, ROS production, oxidative damage kinetics, and metabolic changes occurring both during the period between organ retrieval and implantation and after kidney graft were examined. The 1H-NMR metabolic profile—valine, alanine, acetate, trimetylamine-N-oxide, glutathione, lactate, and the EPR oxidative stress—resulting from ischemia/reperfusion injury after preservation (8 h) by static cold storage (SCS) and ex vivo machine perfusion (HMP) methods were monitored. The functional recovery after transplantation (14 days) was evaluated by serum creatinine (SCr), oxidative stress (ROS), and damage (thiobarbituric-acid-reactive substances and protein carbonyl enzymatic) assessments. At 8 h of preservation storage, a significantly (p < 0.0001) higher ROS production was measured in the SCS vs. HMP group. Significantly higher concentration data (p < 0.05−0.0001) in HMP vs. SCS for all the monitored metabolites were found as well. The HMP group showed a better function recovery. The comparison of the areas under the SCr curves (AUC) returned a significantly smaller (−12.5 %) AUC in the HMP vs. SCS. EPR-ROS concentration (μmol·g−1) from bioptic kidney tissue samples were significantly lower in HMP vs. SCS. The same result was found for the NMR monitored metabolites: lactate: −59.76%, alanine: −43.17%; valine: −58.56%; and TMAO: −77.96%. No changes were observed in either group under light microscopy. In conclusion, a better and more rapid normalization of oxidative stress and functional recovery after transplantation were observed by HMP utilization.
Collapse
Affiliation(s)
- Mrakic-Sposta Simona
- Institute of Clinical Physiology, National Research Council (IFC-CNR), 20159 Milano, Italy
| | - Vezzoli Alessandra
- Institute of Clinical Physiology, National Research Council (IFC-CNR), 20159 Milano, Italy
- Correspondence: (V.A.); (G.M.)
| | - Cova Emanuela
- Department of Molecular Medicine, IRCCS Foundation Policlinico San Matteo, 27100 Pavia, Italy
| | - Ticcozzelli Elena
- Department of Surgery, IRCCS Foundation Policlinico San Matteo, 27100 Pavia, Italy
| | - Montorsi Michela
- Department of Human Sciences and Promotion of the Quality of Life, San Raffaele Roma Open University, 00166 Roma, Italy
| | - Greco Fulvia
- Institute of Chemical Sciences and Technologies “G. Natta”, National Research Council (SCITEC-CNR), 20133 Milan, Italy
| | - Sepe Vincenzo
- Department of Molecular Medicine, IRCCS Foundation Policlinico San Matteo, 27100 Pavia, Italy
| | - Benzoni Ilaria
- Department of Surgery, IRCCS Foundation Policlinico San Matteo, 27100 Pavia, Italy
| | - Meloni Federica
- Section of Pneumology, Department of Internal Medicine, University of Pavia, 27100 Pavia, Italy
| | - Arbustini Eloisa
- Centre for Inherited Cardiovascular Diseases, IRCCS Foundation Policlinico San Matteo, 27100 Pavia, Italy
| | - Abelli Massimo
- Department of Surgery, IRCCS Foundation Policlinico San Matteo, 27100 Pavia, Italy
| | - Gussoni Maristella
- Institute of Chemical Sciences and Technologies “G. Natta”, National Research Council (SCITEC-CNR), 20133 Milan, Italy
- Correspondence: (V.A.); (G.M.)
| |
Collapse
|
13
|
Zhang K, Huang Q, Peng L, Lin S, Liu J, Zhang J, Li C, Zhai S, Xu Z, Wang S. The multifunctional roles of autophagy in the innate immune response: Implications for regulation of transplantation rejection. Front Cell Dev Biol 2022; 10:1007559. [PMID: 36619861 PMCID: PMC9810636 DOI: 10.3389/fcell.2022.1007559] [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: 07/30/2022] [Accepted: 11/04/2022] [Indexed: 12/24/2022] Open
Abstract
Organ transplantation is the main treatment for end-stage organ failure, which has rescued tens of thousands of lives. Immune rejection is the main factor affecting the survival of transplanted organs. How to suppress immune rejection is an important goal of transplantation research. A graft first triggers innate immune responses, leading to graft inflammation, tissue injury and cell death, followed by adaptive immune activation. At present, the importance of innate immunity in graft rejection is poorly understood. Autophagy, an evolutionarily conserved intracellular degradation system, is proven to be involved in regulating innate immune response following graft transplants. Moreover, there is evidence indicating that autophagy can regulate graft dysfunction. Although the specific mechanism by which autophagy affects graft rejection remains unclear, autophagy is involved in innate immune signal transduction, inflammatory response, and various forms of cell death after organ transplantation. This review summarizes how autophagy regulates these processes and proposes potential targets for alleviating immune rejection.
Collapse
Affiliation(s)
- Kunli Zhang
- Institute of Animal Health, Guangdong Academy of Agricultural Sciences, Guangdong Provincial Key Laboratory of Livestock Disease Prevention Guangdong Province, Scientific Observation and Experiment Station of Veterinary Drugs and Diagnostic Techniques of Guangdong Province, Ministry of Agriculture and Rural Affairs, Guangzhou, China
| | - Qiuyan Huang
- State Key Laboratory of Livestock and Poultry Breeding, Guangdong Key Laboratory of Animal Breeding and Nutrition, Institute of Animal Science, Guangdong Academy of Agricultural Sciences, Guangzhou, China
| | - Laru Peng
- Guangzhou Laboratory, Guangzhou International BioIsland, Guangzhou, China
| | - Sen Lin
- Sericultural & Agri-Food Research Institute, Guangdong Academy of Agricultural Sciences, Guangzhou, China
| | - Jie Liu
- Guangdong Yantang Dairy Co, Ltd, Guangzhou, China
| | - Jianfeng Zhang
- Institute of Animal Health, Guangdong Academy of Agricultural Sciences, Guangdong Provincial Key Laboratory of Livestock Disease Prevention Guangdong Province, Scientific Observation and Experiment Station of Veterinary Drugs and Diagnostic Techniques of Guangdong Province, Ministry of Agriculture and Rural Affairs, Guangzhou, China,Maoming Branch, Guangdong Laboratory for Lingnan Modern Agriculture, Maoming, China
| | - Chunling Li
- Institute of Animal Health, Guangdong Academy of Agricultural Sciences, Guangdong Provincial Key Laboratory of Livestock Disease Prevention Guangdong Province, Scientific Observation and Experiment Station of Veterinary Drugs and Diagnostic Techniques of Guangdong Province, Ministry of Agriculture and Rural Affairs, Guangzhou, China
| | - Shaolun Zhai
- Institute of Animal Health, Guangdong Academy of Agricultural Sciences, Guangdong Provincial Key Laboratory of Livestock Disease Prevention Guangdong Province, Scientific Observation and Experiment Station of Veterinary Drugs and Diagnostic Techniques of Guangdong Province, Ministry of Agriculture and Rural Affairs, Guangzhou, China
| | - Zhihong Xu
- Institute of Animal Health, Guangdong Academy of Agricultural Sciences, Guangdong Provincial Key Laboratory of Livestock Disease Prevention Guangdong Province, Scientific Observation and Experiment Station of Veterinary Drugs and Diagnostic Techniques of Guangdong Province, Ministry of Agriculture and Rural Affairs, Guangzhou, China,*Correspondence: Zhihong Xu, ; Sutian Wang,
| | - Sutian Wang
- State Key Laboratory of Livestock and Poultry Breeding, Guangdong Key Laboratory of Animal Breeding and Nutrition, Institute of Animal Science, Guangdong Academy of Agricultural Sciences, Guangzhou, China,Maoming Branch, Guangdong Laboratory for Lingnan Modern Agriculture, Maoming, China,*Correspondence: Zhihong Xu, ; Sutian Wang,
| |
Collapse
|
14
|
Hauet T, Pisani DF. New Strategies Protecting from Ischemia/Reperfusion. Int J Mol Sci 2022; 23:ijms232415867. [PMID: 36555508 PMCID: PMC9779635 DOI: 10.3390/ijms232415867] [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: 12/06/2022] [Accepted: 12/12/2022] [Indexed: 12/15/2022] Open
Abstract
This Special Issue aims to highlight new avenues in the management of Ischemia/Reperfusion (I/R) injury [...].
Collapse
Affiliation(s)
- Thierry Hauet
- INSERM U1313, IRMETIST, Université de Poitiers et CHU de Poitiers, 86021 Poitiers, France
| | | |
Collapse
|
15
|
Lepoittevin M, Giraud S, Kerforne T, Allain G, Thuillier R, Hauet T. How to improve results after DCD (donation after circulation death). Presse Med 2022; 51:104143. [PMID: 36216034 DOI: 10.1016/j.lpm.2022.104143] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/31/2021] [Accepted: 09/29/2022] [Indexed: 11/09/2022] Open
Abstract
The shortage of organs for transplantation has led health professionals to look for alternative sources of donors. One of the avenues concerns donors who have died after circulatory arrest. This is a special situation because the organs from these donors are exposed to warm ischaemia-reperfusion lesions that are unavoidable during the journey of the organs from the donor to the moment of transplantation in the recipient. We will address and discuss the key issues from the perspective of team organization, legislation and its evolution, and the ethical framework. In a second part, the avenues to improve the quality of organs will be presented following the itinerary of the organs between the donor and the recipient. The important moments from the point of view of therapeutic strategy will be put into perspective. New connections between key players involved in pathophysiological mechanisms and implications for innate immunity and injury processes are among the avenues to explore. Technological developments to improve the quality of organs from these recipients will be analyzed, such as perfusion techniques with new modalities of temperatures and oxygenation. New molecules are being investigated for their potential role in protecting these organs and an analysis of potential prospects will be proposed. Finally, the important perspectives that seem to be favored will be discussed in order to reposition the use of deceased donors after circulatory arrest. The use of these organs has become a routine procedure and improving their quality and providing the means for their evaluation is absolutely inevitable.
Collapse
Affiliation(s)
- Maryne Lepoittevin
- Unité UMR U1082, F-86000 Poitiers, France; Faculté de Médecine et de Pharmacie, Université de Poitiers, F-86000 Poitiers, France
| | - Sébastien Giraud
- Unité UMR U1082, F-86000 Poitiers, France; Service de Biochimie, Pôle Biospharm, Centre Hospitalier Universitaire, 2 rue de la Milétrie, CS 90577, 86021 Poitiers Cedex, France
| | - Thomas Kerforne
- Unité UMR U1082, F-86000 Poitiers, France; Faculté de Médecine et de Pharmacie, Université de Poitiers, F-86000 Poitiers, France; CHU Poitiers, Service de Réanimation Chirurgie Cardio-Thoracique et Vasculaire, Coordination des P.M.O., F-86021 Poitiers, France
| | - Géraldine Allain
- Unité UMR U1082, F-86000 Poitiers, France; Faculté de Médecine et de Pharmacie, Université de Poitiers, F-86000 Poitiers, France; CHU Poitiers, Service de Chirurgie Cardiothoracique et Vasculaire, F-86021 Poitiers, France
| | - Raphaël Thuillier
- Unité UMR U1082, F-86000 Poitiers, France; Faculté de Médecine et de Pharmacie, Université de Poitiers, F-86000 Poitiers, France; Service de Biochimie, Pôle Biospharm, Centre Hospitalier Universitaire, 2 rue de la Milétrie, CS 90577, 86021 Poitiers Cedex, France
| | - Thierry Hauet
- Unité UMR U1082, F-86000 Poitiers, France; Faculté de Médecine et de Pharmacie, Université de Poitiers, F-86000 Poitiers, France; Fédération Hospitalo-Universitaire « Survival Optimization in Organ Transplantation », CHU de Poitiers, 2 rue de la Milétrie - CS 90577, 86021 Poitiers Cedex, France.
| |
Collapse
|
16
|
Zhou X, Chen Q, Guo C, Su Y, Guo H, Cao M, Liu Z, Zhang D, Diao N, Fan H, Chen D. CD44 Receptor-Targeted and Reactive Oxygen Species-Responsive H 2S Donor Micelles Based on Hyaluronic Acid for the Therapy of Renal Ischemia/Reperfusion Injury. ACS OMEGA 2022; 7:42339-42346. [PMID: 36440107 PMCID: PMC9686187 DOI: 10.1021/acsomega.2c05407] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/22/2022] [Accepted: 10/31/2022] [Indexed: 06/16/2023]
Abstract
For the therapy attenuating renal ischemia-reperfusion (IR) injury, a novel drug delivery system was urgently needed, which could precisely deliver drugs to the pathological renal tissue. Here, we have prepared new nanomaterials with a reactive oxygen species (ROS)-responsive hydrogen sulfide (H2S) donor and hyaluronic acid that targets CD44 receptor. The novel material was synthesized and characterized via related experiments. Then, rapamycin was loaded, which inhibited kidney damage. In the in vitro study, we found that the micelles had ROS-responsiveness, biocompatibility, and cell penetration. In addition, the experimental results showed that the intracellular H2S concentration after administration was threefold higher than that of the control group. The western blot assay revealed that they have anti-inflammatory effects via H2S donor blocking the NF-κB signaling pathway. Consequently, the rising CD44 receptor-targeting and ROS-sensitive H2S donor micelles would provide a promising way for renal IR injury. This work provides a strategy for improving ischemia/reperfusion injury for pharmaceuticals.
Collapse
Affiliation(s)
- Xiudi Zhou
- Collaborative
Innovation Center of Advanced Drug Delivery System and Biotech Drugs,
School of Pharmacy, Yantai University, Yantai264005, P. R. China
- Department
of Pharmacy, Binzhou People’s Hospital
Affiliated to Shandong First Medical University, China, Binzhou256600, P. R. China
| | - Qiang Chen
- Collaborative
Innovation Center of Advanced Drug Delivery System and Biotech Drugs,
School of Pharmacy, Yantai University, Yantai264005, P. R. China
| | - Chunjing Guo
- College
of Marine Life Science, Ocean University
of China, Qingdao266003, P. R. China
| | - Yanguo Su
- Collaborative
Innovation Center of Advanced Drug Delivery System and Biotech Drugs,
School of Pharmacy, Yantai University, Yantai264005, P. R. China
| | - Huimin Guo
- Collaborative
Innovation Center of Advanced Drug Delivery System and Biotech Drugs,
School of Pharmacy, Yantai University, Yantai264005, P. R. China
| | - Min Cao
- Collaborative
Innovation Center of Advanced Drug Delivery System and Biotech Drugs,
School of Pharmacy, Yantai University, Yantai264005, P. R. China
| | - Zhongxin Liu
- Collaborative
Innovation Center of Advanced Drug Delivery System and Biotech Drugs,
School of Pharmacy, Yantai University, Yantai264005, P. R. China
| | - Dandan Zhang
- Collaborative
Innovation Center of Advanced Drug Delivery System and Biotech Drugs,
School of Pharmacy, Yantai University, Yantai264005, P. R. China
| | - Ningning Diao
- Collaborative
Innovation Center of Advanced Drug Delivery System and Biotech Drugs,
School of Pharmacy, Yantai University, Yantai264005, P. R. China
| | - Huaying Fan
- Collaborative
Innovation Center of Advanced Drug Delivery System and Biotech Drugs,
School of Pharmacy, Yantai University, Yantai264005, P. R. China
| | - Daquan Chen
- Collaborative
Innovation Center of Advanced Drug Delivery System and Biotech Drugs,
School of Pharmacy, Yantai University, Yantai264005, P. R. China
| |
Collapse
|
17
|
Perera MR, Sinclair JH. The Human Cytomegalovirus β2.7 Long Non-Coding RNA Prevents Induction of Reactive Oxygen Species to Maintain Viral Gene Silencing during Latency. Int J Mol Sci 2022; 23:ijms231911017. [PMID: 36232315 PMCID: PMC9569889 DOI: 10.3390/ijms231911017] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2022] [Revised: 09/06/2022] [Accepted: 09/16/2022] [Indexed: 11/16/2022] Open
Abstract
Human cytomegalovirus (HCMV) is a significant source of disease for the immunosuppressed and immunonaive. The treatment of HCMV is made more problematic by viral latency, a lifecycle stage in which the virus reduces its own gene expression and produces no infectious virus. The most highly expressed viral gene during HCMV latency is the viral β2.7 long non-coding RNA. Although we have recently shown that the β2.7 lncRNA lowers levels of reactive oxygen species (ROS) during infection in monocytes, how this impacts latency is unclear. We now show that β2.7 is important for establishing and maintaining HCMV latency by aiding the suppression of viral lytic gene expression and that this is directly related to its ability to quench reactive oxygen species (ROS). Consistent with this, we also find that exogenous inducers of ROS cause reactivation of latent HCMV. These effects can be compensated by treatment with an antioxidant to lower ROS levels. Finally, we show that ROS-mediated reactivation is independent of myeloid differentiation, but instead relies on NF-κB activation. Altogether, these results reveal a novel factor that is central to the complex process that underpins HCMV latency. These findings may be of particular relevance in the transplant setting, in which transplanted tissue/organs are subject to very high ROS levels, and HCMV reactivation poses a significant threat.
Collapse
|
18
|
Zhou Q, He X, Zhao X, Fan Q, Lai S, Liu D, He H, He M. Ginsenoside Rg1 Ameliorates Acute Renal Ischemia/Reperfusion Injury via Upregulating AMPK α1 Expression. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:3737137. [PMID: 36092159 PMCID: PMC9458375 DOI: 10.1155/2022/3737137] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/16/2022] [Revised: 06/29/2022] [Accepted: 07/29/2022] [Indexed: 11/17/2022]
Abstract
Acute renal ischemia/reperfusion (I/R) injury often occurs during kidney transplantation and other kidney surgeries, and the molecular mechanism involves oxidative stress. We hypothesized that ginsenoside Rg1 (Rg1), a saponin derived from ginseng, would protect the renal tissue against acute renal I/R injury by upregulating 5' adenosine monophosphate-activated protein kinase α1 (AMPKα1) expression and inhibiting oxidative stress. The models of acute anoxia/reoxygenation (A/R) damage in normal rat kidney epithelial cell lines (NRK-52E) and acute renal I/R injury in mice were constructed. The results revealed that pretreatment with 25 μM Rg1 significantly increased NRK-52E viability, decreased lactate dehydrogenase (LDH) activity and apoptosis, suppressed reactive oxygen species generation and oxidative stress, stabilized mitochondrial membrane potential and reduced mitochondria permeability transition pore openness, decreased adenosine monophosphate/adenosine triphosphate ratio, and upregulated the expression of AMPKα1, cytochrome b-c1 complex subunit 2, NADH dehydrogenase (ubiquinone) 1 beta subcomplex subunit 8, and B-cell lymphoma 2, while downregulating BCL2-associated X protein expression. The effects of Rg1 pretreatment were similar to those of pAD/Flag-AMPKα1. After acute renal I/R injury, serum creatinine, blood urea nitrogen, LDH activity, and oxidative stress in renal tissue significantly increased. Rg1 pretreatment upregulated AMPKα1 expression, which protects against acute renal I/R injury by maintaining renal function homeostasis, inhibiting oxidative stress, and reducing apoptosis. Compound C, a specific inhibitor of AMPK, reversed the effects of Rg1. In summary, Rg1 pretreatment upregulated AMPKα1 expression, inhibited oxidative stress, maintained mitochondrial function, improved energy metabolism, reduced apoptosis, and ultimately protected renal tissue against acute renal I/R injury.
Collapse
Affiliation(s)
- Qing Zhou
- Institute of Cardiovascular Diseases, Jiangxi Academy of Clinical Medical Sciences, The First Affiliated Hospital of Nanchang University, Nanchang 330006, China
| | - Xinlan He
- Jiangxi Provincial Key Laboratory of Basic Pharmacology, Nanchang University School of Pharmaceutical Science, Nanchang 330006, China
| | - Xiaoyu Zhao
- Jiangxi Provincial Key Laboratory of Basic Pharmacology, Nanchang University School of Pharmaceutical Science, Nanchang 330006, China
| | - Qigui Fan
- Jiangxi Provincial Key Laboratory of Basic Pharmacology, Nanchang University School of Pharmaceutical Science, Nanchang 330006, China
| | - Songqing Lai
- Institute of Cardiovascular Diseases, Jiangxi Academy of Clinical Medical Sciences, The First Affiliated Hospital of Nanchang University, Nanchang 330006, China
| | - Dan Liu
- Jiangxi Provincial Key Laboratory of Basic Pharmacology, Nanchang University School of Pharmaceutical Science, Nanchang 330006, China
| | - Huan He
- Jiangxi Provincial Key Laboratory of Basic Pharmacology, Nanchang University School of Pharmaceutical Science, Nanchang 330006, China
| | - Ming He
- Institute of Cardiovascular Diseases, Jiangxi Academy of Clinical Medical Sciences, The First Affiliated Hospital of Nanchang University, Nanchang 330006, China
- Jiangxi Provincial Key Laboratory of Basic Pharmacology, Nanchang University School of Pharmaceutical Science, Nanchang 330006, China
| |
Collapse
|
19
|
Ritiu SA, Rogobete AF, Sandesc D, Bedreag OH, Papurica M, Popovici SE, Toma D, Ivascu RI, Velovan R, Garofil DN, Corneci D, Bratu LM, Pahontu EM, Pistol A. The Impact of General Anesthesia on Redox Stability and Epigenetic Inflammation Pathways: Crosstalk on Perioperative Antioxidant Therapy. Cells 2022; 11:1880. [PMID: 35741011 PMCID: PMC9221536 DOI: 10.3390/cells11121880] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2022] [Revised: 05/31/2022] [Accepted: 06/07/2022] [Indexed: 02/07/2023] Open
Abstract
Worldwide, the prevalence of surgery under general anesthesia has significantly increased, both because of modern anesthetic and pain-control techniques and because of better diagnosis and the increased complexity of surgical techniques. Apart from developing new concepts in the surgical field, researchers and clinicians are now working on minimizing the impact of surgical trauma and offering minimal invasive procedures due to the recent discoveries in the field of cellular and molecular mechanisms that have revealed a systemic inflammatory and pro-oxidative impact not only in the perioperative period but also in the long term, contributing to more difficult recovery, increased morbidity and mortality, and a negative financial impact. Detailed molecular and cellular analysis has shown an overproduction of inflammatory and pro-oxidative species, responsible for augmenting the systemic inflammatory status and making postoperative recovery more difficult. Moreover, there are a series of changes in certain epigenetic structures, the most important being the microRNAs. This review describes the most important molecular and cellular mechanisms that impact the surgical patient undergoing general anesthesia, and it presents a series of antioxidant therapies that can reduce systemic inflammation.
Collapse
Affiliation(s)
- Stelian Adrian Ritiu
- Clinic of Anaesthesia and Intensive Care, Emergency County Hospital “Pius Brînzeu”, 300723 Timișoara, Romania; (S.A.R.); (D.S.); (O.H.B.); (M.P.); (S.E.P.); (D.T.); (R.V.)
- Faculty of Medicine, “Victor Babeș” University of Medicine and Pharmacy, 300041 Timișoara, Romania;
| | - Alexandru Florin Rogobete
- Clinic of Anaesthesia and Intensive Care, Emergency County Hospital “Pius Brînzeu”, 300723 Timișoara, Romania; (S.A.R.); (D.S.); (O.H.B.); (M.P.); (S.E.P.); (D.T.); (R.V.)
- Faculty of Medicine, “Victor Babeș” University of Medicine and Pharmacy, 300041 Timișoara, Romania;
- Anaesthesia and Intensive Care Research Center (CCATITM), “Victor Babeș” University of Medicine and Pharmacy, 300041 Timișoara, Romania
| | - Dorel Sandesc
- Clinic of Anaesthesia and Intensive Care, Emergency County Hospital “Pius Brînzeu”, 300723 Timișoara, Romania; (S.A.R.); (D.S.); (O.H.B.); (M.P.); (S.E.P.); (D.T.); (R.V.)
- Faculty of Medicine, “Victor Babeș” University of Medicine and Pharmacy, 300041 Timișoara, Romania;
- Anaesthesia and Intensive Care Research Center (CCATITM), “Victor Babeș” University of Medicine and Pharmacy, 300041 Timișoara, Romania
| | - Ovidiu Horea Bedreag
- Clinic of Anaesthesia and Intensive Care, Emergency County Hospital “Pius Brînzeu”, 300723 Timișoara, Romania; (S.A.R.); (D.S.); (O.H.B.); (M.P.); (S.E.P.); (D.T.); (R.V.)
- Faculty of Medicine, “Victor Babeș” University of Medicine and Pharmacy, 300041 Timișoara, Romania;
- Anaesthesia and Intensive Care Research Center (CCATITM), “Victor Babeș” University of Medicine and Pharmacy, 300041 Timișoara, Romania
| | - Marius Papurica
- Clinic of Anaesthesia and Intensive Care, Emergency County Hospital “Pius Brînzeu”, 300723 Timișoara, Romania; (S.A.R.); (D.S.); (O.H.B.); (M.P.); (S.E.P.); (D.T.); (R.V.)
- Faculty of Medicine, “Victor Babeș” University of Medicine and Pharmacy, 300041 Timișoara, Romania;
- Anaesthesia and Intensive Care Research Center (CCATITM), “Victor Babeș” University of Medicine and Pharmacy, 300041 Timișoara, Romania
| | - Sonia Elena Popovici
- Clinic of Anaesthesia and Intensive Care, Emergency County Hospital “Pius Brînzeu”, 300723 Timișoara, Romania; (S.A.R.); (D.S.); (O.H.B.); (M.P.); (S.E.P.); (D.T.); (R.V.)
- Faculty of Medicine, “Victor Babeș” University of Medicine and Pharmacy, 300041 Timișoara, Romania;
| | - Daiana Toma
- Clinic of Anaesthesia and Intensive Care, Emergency County Hospital “Pius Brînzeu”, 300723 Timișoara, Romania; (S.A.R.); (D.S.); (O.H.B.); (M.P.); (S.E.P.); (D.T.); (R.V.)
- Faculty of Medicine, “Victor Babeș” University of Medicine and Pharmacy, 300041 Timișoara, Romania;
| | - Robert Iulian Ivascu
- Faculty of Medicine, “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania; (R.I.I.); (D.C.); (A.P.)
- Clinic of Anaesthesia and Intensive Care, Central Military Emergency Hospital “Dr. Carol Davila”, 010242 Bucharest, Romania
| | - Raluca Velovan
- Clinic of Anaesthesia and Intensive Care, Emergency County Hospital “Pius Brînzeu”, 300723 Timișoara, Romania; (S.A.R.); (D.S.); (O.H.B.); (M.P.); (S.E.P.); (D.T.); (R.V.)
- Faculty of Medicine, “Victor Babeș” University of Medicine and Pharmacy, 300041 Timișoara, Romania;
| | - Dragos Nicolae Garofil
- Faculty of Medicine, “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania; (R.I.I.); (D.C.); (A.P.)
| | - Dan Corneci
- Faculty of Medicine, “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania; (R.I.I.); (D.C.); (A.P.)
- Clinic of Anaesthesia and Intensive Care, Central Military Emergency Hospital “Dr. Carol Davila”, 010242 Bucharest, Romania
| | - Lavinia Melania Bratu
- Faculty of Medicine, “Victor Babeș” University of Medicine and Pharmacy, 300041 Timișoara, Romania;
| | - Elena Mihaela Pahontu
- Faculty of Pharmacy, “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania;
| | - Adriana Pistol
- Faculty of Medicine, “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania; (R.I.I.); (D.C.); (A.P.)
| |
Collapse
|
20
|
5-Methyltetrahydrofolate Attenuates Oxidative Stress and Improves Kidney Function in Acute Kidney Injury through Activation of Nrf2 and Antioxidant Defense. Antioxidants (Basel) 2022; 11:antiox11061046. [PMID: 35739943 PMCID: PMC9219715 DOI: 10.3390/antiox11061046] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Revised: 05/10/2022] [Accepted: 05/16/2022] [Indexed: 02/04/2023] Open
Abstract
Oxidative stress is a major mediator of adverse outcomes in acute kidney injury (AKI). Deficiency of micronutrients, such as folate, is common in AKI. Our previous study reported that AKI impaired kidney reabsorption of folate and decreased plasma folate level in rats. The present study investigated the effect of 5-methyltetrahydrofolate (5-MTHF), a biologically active form of folate/folic acid, on AKI-impaired kidney function and oxidative stress. Sprague-Dawley rats developed AKI after kidney ischemia (45 min) and reperfusion (24 h). Injection of 5-MTHF (3 µg/kg body weight) improved kidney function and attenuated oxidative stress with a restoration of glutathione and a reduction of lipid peroxidation in the kidney. Injection of 5-MTHF activated transcription factor Nrf2 and increased the expression of glutathione synthesizing enzymes, superoxide dismutase-1 and heme oxygenase-1 in the kidney. Simulated ischemia-reperfusion through hypoxia-reoxygenation increased oxidative stress in proximal tubular cells. Incubation of cells with 5-MTHF alleviated cell injury and increased antioxidant enzyme expression and intracellular glutathione levels. Inhibition of Nrf2 expression through siRNA transfection abolished the effect of 5-MTHF against oxidative stress. These results suggest that low-dose folic acid can improve kidney function through activation of Nrf2 and restoration of antioxidant defence. Micronutrient supplements may improve clinical outcomes in AKI.
Collapse
|
21
|
Lepoittevin M, Giraud S, Kerforne T, Barrou B, Badet L, Bucur P, Salamé E, Goumard C, Savier E, Branchereau J, Battistella P, Mercier O, Mussot S, Hauet T, Thuillier R. Preservation of Organs to Be Transplanted: An Essential Step in the Transplant Process. Int J Mol Sci 2022; 23:ijms23094989. [PMID: 35563381 PMCID: PMC9104613 DOI: 10.3390/ijms23094989] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Revised: 04/25/2022] [Accepted: 04/27/2022] [Indexed: 12/23/2022] Open
Abstract
Organ transplantation remains the treatment of last resort in case of failure of a vital organ (lung, liver, heart, intestine) or non-vital organ (essentially the kidney and pancreas) for which supplementary treatments exist. It remains the best alternative both in terms of quality-of-life and life expectancy for patients and of public health expenditure. Unfortunately, organ shortage remains a widespread issue, as on average only about 25% of patients waiting for an organ are transplanted each year. This situation has led to the consideration of recent donor populations (deceased by brain death with extended criteria or deceased after circulatory arrest). These organs are sensitive to the conditions of conservation during the ischemia phase, which have an impact on the graft’s short- and long-term fate. This evolution necessitates a more adapted management of organ donation and the optimization of preservation conditions. In this general review, the different aspects of preservation will be considered. Initially done by hypothermia with the help of specific solutions, preservation is evolving with oxygenated perfusion, in hypothermia or normothermia, aiming at maintaining tissue metabolism. Preservation time is also becoming a unique evaluation window to predict organ quality, allowing repair and/or optimization of recipient choice.
Collapse
Affiliation(s)
- Maryne Lepoittevin
- Biochemistry Department, CHU Poitiers, 86021 Poitiers, France; (M.L.); (S.G.); (R.T.)
- Faculty of Medicine and Pharmacy, University of Poitiers, 86073 Poitiers, France;
- INSERM U1313, IRMETIST, 86021 Poitiers, France; (B.B.); (L.B.)
| | - Sébastien Giraud
- Biochemistry Department, CHU Poitiers, 86021 Poitiers, France; (M.L.); (S.G.); (R.T.)
- Faculty of Medicine and Pharmacy, University of Poitiers, 86073 Poitiers, France;
- INSERM U1313, IRMETIST, 86021 Poitiers, France; (B.B.); (L.B.)
| | - Thomas Kerforne
- Faculty of Medicine and Pharmacy, University of Poitiers, 86073 Poitiers, France;
- INSERM U1313, IRMETIST, 86021 Poitiers, France; (B.B.); (L.B.)
- Cardio-Thoracic and Vascular Surgery Intensive Care Unit, Coordination of P.M.O., CHU Poitiers, 86021 Poitiers, France
| | - Benoit Barrou
- INSERM U1313, IRMETIST, 86021 Poitiers, France; (B.B.); (L.B.)
- Sorbonne Université Campus Pierre et Marie Curie, Faculté de Médecine, 75005 Paris, France
- Service Médico-Chirurgical de Transplantation Rénale, APHP, Hôpital Pitié-Salpêtrière, 75013 Paris, France
- Société Francophone de Transplantation et de l’Ecole Francophone pour le Prélèvement Multi-Organes, 75013 Paris, France; (P.B.); (E.S.); (C.G.); (E.S.); (J.B.); (P.B.); (O.M.); (S.M.)
| | - Lionel Badet
- INSERM U1313, IRMETIST, 86021 Poitiers, France; (B.B.); (L.B.)
- Société Francophone de Transplantation et de l’Ecole Francophone pour le Prélèvement Multi-Organes, 75013 Paris, France; (P.B.); (E.S.); (C.G.); (E.S.); (J.B.); (P.B.); (O.M.); (S.M.)
- Faculté de Médecine, Campus Lyon Santé Est, Université Claude Bernard, 69622 Lyon, France
- Service d’Urologie et Transplantation, Hospices Civils de Lyon, Hôpital Edouard-Herriot, 69003 Lyon, France
| | - Petru Bucur
- Société Francophone de Transplantation et de l’Ecole Francophone pour le Prélèvement Multi-Organes, 75013 Paris, France; (P.B.); (E.S.); (C.G.); (E.S.); (J.B.); (P.B.); (O.M.); (S.M.)
- Service de Chirurgie Digestive et Endocrinienne, Transplantation Hépatique, CHU de Tours, 37170 Chambray les Tours, France
- Groupement d’Imagerie Médicale, CHU de Tours, 37000 Tours, France
- University Hospital Federation SUPORT Tours Poitiers Limoges, 86021 Poitiers, France
| | - Ephrem Salamé
- Société Francophone de Transplantation et de l’Ecole Francophone pour le Prélèvement Multi-Organes, 75013 Paris, France; (P.B.); (E.S.); (C.G.); (E.S.); (J.B.); (P.B.); (O.M.); (S.M.)
- Service de Chirurgie Digestive et Endocrinienne, Transplantation Hépatique, CHU de Tours, 37170 Chambray les Tours, France
- Groupement d’Imagerie Médicale, CHU de Tours, 37000 Tours, France
- University Hospital Federation SUPORT Tours Poitiers Limoges, 86021 Poitiers, France
| | - Claire Goumard
- Société Francophone de Transplantation et de l’Ecole Francophone pour le Prélèvement Multi-Organes, 75013 Paris, France; (P.B.); (E.S.); (C.G.); (E.S.); (J.B.); (P.B.); (O.M.); (S.M.)
- Service de Chirurgie Digestive, Hépato-Bilio-Pancréatique et Transplantation Hépatique, APHP, Hôpital Pitié-Salpêtrière, 75013 Paris, France
| | - Eric Savier
- Société Francophone de Transplantation et de l’Ecole Francophone pour le Prélèvement Multi-Organes, 75013 Paris, France; (P.B.); (E.S.); (C.G.); (E.S.); (J.B.); (P.B.); (O.M.); (S.M.)
- Service de Chirurgie Digestive, Hépato-Bilio-Pancréatique et Transplantation Hépatique, APHP, Hôpital Pitié-Salpêtrière, 75013 Paris, France
| | - Julien Branchereau
- Société Francophone de Transplantation et de l’Ecole Francophone pour le Prélèvement Multi-Organes, 75013 Paris, France; (P.B.); (E.S.); (C.G.); (E.S.); (J.B.); (P.B.); (O.M.); (S.M.)
- Service d’Urologie et de Transplantation, CHU de Nantes, 44000 Nantes, France
| | - Pascal Battistella
- Société Francophone de Transplantation et de l’Ecole Francophone pour le Prélèvement Multi-Organes, 75013 Paris, France; (P.B.); (E.S.); (C.G.); (E.S.); (J.B.); (P.B.); (O.M.); (S.M.)
- Service de Cardiologie et Maladies Vasculaires, CHU de Montpellier, CEDEX 5, 34295 Montpellier, France
| | - Olaf Mercier
- Société Francophone de Transplantation et de l’Ecole Francophone pour le Prélèvement Multi-Organes, 75013 Paris, France; (P.B.); (E.S.); (C.G.); (E.S.); (J.B.); (P.B.); (O.M.); (S.M.)
- Service de Chirurgie Thoracique et Cardio-Vasculaire, Centre Chirurgical Marie LANNELONGUE, 92350 Le Plessis Robinson, France
| | - Sacha Mussot
- Société Francophone de Transplantation et de l’Ecole Francophone pour le Prélèvement Multi-Organes, 75013 Paris, France; (P.B.); (E.S.); (C.G.); (E.S.); (J.B.); (P.B.); (O.M.); (S.M.)
- Service de Chirurgie Thoracique et Cardio-Vasculaire, Centre Chirurgical Marie LANNELONGUE, 92350 Le Plessis Robinson, France
| | - Thierry Hauet
- Biochemistry Department, CHU Poitiers, 86021 Poitiers, France; (M.L.); (S.G.); (R.T.)
- Faculty of Medicine and Pharmacy, University of Poitiers, 86073 Poitiers, France;
- INSERM U1313, IRMETIST, 86021 Poitiers, France; (B.B.); (L.B.)
- Société Francophone de Transplantation et de l’Ecole Francophone pour le Prélèvement Multi-Organes, 75013 Paris, France; (P.B.); (E.S.); (C.G.); (E.S.); (J.B.); (P.B.); (O.M.); (S.M.)
- University Hospital Federation SUPORT Tours Poitiers Limoges, 86021 Poitiers, France
- Correspondence:
| | - Raphael Thuillier
- Biochemistry Department, CHU Poitiers, 86021 Poitiers, France; (M.L.); (S.G.); (R.T.)
- Faculty of Medicine and Pharmacy, University of Poitiers, 86073 Poitiers, France;
- INSERM U1313, IRMETIST, 86021 Poitiers, France; (B.B.); (L.B.)
| |
Collapse
|
22
|
Oxidative Stress and Ischemia/Reperfusion Injury in Kidney Transplantation: Focus on Ferroptosis, Mitophagy and New Antioxidants. Antioxidants (Basel) 2022; 11:antiox11040769. [PMID: 35453454 PMCID: PMC9024672 DOI: 10.3390/antiox11040769] [Citation(s) in RCA: 42] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Revised: 04/06/2022] [Accepted: 04/08/2022] [Indexed: 12/11/2022] Open
Abstract
Although there has been technical and pharmacological progress in kidney transplant medicine, some patients may experience acute post-transplant complications. Among the mechanisms involved in these conditions, ischemia/reperfusion (I/R) injury may have a primary pathophysiological role since it is one of the leading causes of delayed graft function (DGF), a slow recovery of the renal function with the need for dialysis (generally during the first week after transplantation). DGF has a significant social and economic impact as it is associated with prolonged hospitalization and the development of severe complications (including acute rejection). During I/R injury, oxidative stress plays a major role activating several pathways including ferroptosis, an iron-driven cell death characterized by iron accumulation and excessive lipid peroxidation, and mitophagy, a selective degradation of damaged mitochondria by autophagy. Ferroptosis may contribute to the renal damage, while mitophagy can have a protective role by reducing the release of reactive oxygen species from dysfunctional mitochondria. Deep comprehension of both pathways may offer the possibility of identifying new early diagnostic noninvasive biomarkers of DGF and introducing new clinically employable pharmacological strategies. In this review we summarize all relevant knowledge in this field and discuss current antioxidant pharmacological strategies that could represent, in the next future, potential treatments for I/R injury.
Collapse
|
23
|
Chen T, Niu L, Wang L, Zhou Q, Zhao X, Lai S, He X, He H, He M. Ferulic acid protects renal tubular epithelial cells against anoxia/reoxygenation injury mediated by AMPKα1. Free Radic Res 2022; 56:173-184. [PMID: 35382666 DOI: 10.1080/10715762.2022.2062339] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
Anoxia/reoxygenation (A/R) injury causes dysfunction of rat renal tubular epithelial cells (NRK-52E), which is associated with excess reactive oxygen species (ROS) generation and eventually leads to apoptosis. Ferulic acid (FA), a phenolic acid, which is abundant in fruits and vegetables. FA possesses the properties of scavenging free radicals and cytoprotection against oxygen stress. In the study, the protective effects of FA against NRK-52E cells damage induced by A/R were explored and confirmed the role of AMP-activated protein kinaseα1 (AMPKα1). We found that after NRK-52E cells suffered A/R damage, FA pretreatment increased the cell viability and decreased LDH activity in culture medium in a concentration-dependent manner, the activities of endogenous antioxidant enzymes such as glutathione peroxidase, superoxide dismutase and catalase improved, intracellular ROS generation and malondialdehyde contents mitigated. In addition, pretreatment of 75 μM FA ameliorated mitochondrial dysfunction by A/R-injury and ultimately decreased apoptosis (25.3 ± 0.61 vs 12.1 ± 0.60), which was evidenced by preventing the release of cytochrome c from mitochondria to the cytoplasm. 75 μM FA pretreatment also significantly upregulated AMPKα1 expression (3.16 ± 0.18 folds) and phosphorylation (2.56 ± 0.13 folds). However, compound C, a specific AMPK inhibitor, significantly attenuated FA pretreatment's effects, as mentionedabove. These results firstly clarified that FA pretreatment attenuated NRK-52E cell damage induced by A/R via upregulating AMPKα1 expression and phosphorylation.
Collapse
Affiliation(s)
- Tianpeng Chen
- Institute of Cardiovascular Diseases, Jiangxi Academy of Clinical Medical Sciences, the First Affiliated Hospital of Nanchang University, Nanchang 330006, China
| | - Li Niu
- Jiangxi Provincial Key Laboratory of Basic Pharmacology, Nanchang University School of Pharmaceutical Science, Nanchang 330006, China
| | - Liang Wang
- Department of rehabilitation, the First Affiliated Hospital of Nanchang University, Nanchang 330006, China
| | - Qing Zhou
- Jiangxi Provincial Key Laboratory of Basic Pharmacology, Nanchang University School of Pharmaceutical Science, Nanchang 330006, China
| | - Xiaoyu Zhao
- Jiangxi Provincial Key Laboratory of Basic Pharmacology, Nanchang University School of Pharmaceutical Science, Nanchang 330006, China
| | - Songqing Lai
- Institute of Cardiovascular Diseases, Jiangxi Academy of Clinical Medical Sciences, the First Affiliated Hospital of Nanchang University, Nanchang 330006, China
| | - Xinlan He
- Jiangxi Provincial Key Laboratory of Basic Pharmacology, Nanchang University School of Pharmaceutical Science, Nanchang 330006, China
| | - Huan He
- Jiangxi Provincial Key Laboratory of Basic Pharmacology, Nanchang University School of Pharmaceutical Science, Nanchang 330006, China
| | - Ming He
- Institute of Cardiovascular Diseases, Jiangxi Academy of Clinical Medical Sciences, the First Affiliated Hospital of Nanchang University, Nanchang 330006, China
| |
Collapse
|
24
|
Verstraeten L, Jochmans I. Sense and Sensibilities of Organ Perfusion as a Kidney and Liver Viability Assessment Platform. Transpl Int 2022; 35:10312. [PMID: 35356401 PMCID: PMC8958413 DOI: 10.3389/ti.2022.10312] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Accepted: 01/26/2022] [Indexed: 12/13/2022]
Abstract
Predicting organ viability before transplantation remains one of the most challenging and ambitious objectives in transplant surgery. Waitlist mortality is high while transplantable organs are discarded. Currently, around 20% of deceased donor kidneys and livers are discarded because of “poor organ quality”, Decisions to discard are still mainly a subjective judgement since there are only limited reliable tools predictive of outcome available. Organ perfusion technology has been posed as a platform for pre-transplant organ viability assessment. Markers of graft injury and function as well as perfusion parameters have been investigated as possible viability markers during ex-situ hypothermic and normothermic perfusion. We provide an overview of the available evidence for the use of kidney and liver perfusion as a tool to predict posttransplant outcomes. Although evidence shows post-transplant outcomes can be predicted by both injury markers and perfusion parameters during hypothermic kidney perfusion, the predictive accuracy is too low to warrant clinical decision making based upon these parameters alone. In liver, further evidence on the usefulness of hypothermic perfusion as a predictive tool is needed. Normothermic perfusion, during which the organ remains fully metabolically active, seems a more promising platform for true viability assessment. Although we do not yet fully understand “on-pump” organ behaviour at normothermia, initial data in kidney and liver are promising. Besides the need for well-designed (registry) studies to advance the field, the catch-22 of selection bias in clinical studies needs addressing.
Collapse
Affiliation(s)
- Laurence Verstraeten
- Lab of Abdominal Transplantation, Transplantation Research Group, Department of Microbiology, Immunology and Transplantation, KU Leuven, Leuven, Belgium
| | - Ina Jochmans
- Lab of Abdominal Transplantation, Transplantation Research Group, Department of Microbiology, Immunology and Transplantation, KU Leuven, Leuven, Belgium
- Department of Abdominal Transplantation, University Hospitals Leuven, Leuven, Belgium
- *Correspondence: Ina Jochmans,
| |
Collapse
|
25
|
Melis N, Carcy R, Rubera I, Cougnon M, Duranton C, Tauc M, Pisani DF. Akt Inhibition as Preconditioning Treatment to Protect Kidney Cells against Anoxia. Int J Mol Sci 2021; 23:ijms23010152. [PMID: 35008578 PMCID: PMC8745656 DOI: 10.3390/ijms23010152] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2021] [Revised: 11/15/2021] [Accepted: 12/21/2021] [Indexed: 11/16/2022] Open
Abstract
Lesions issued from the ischemia/reperfusion (I/R) stress are a major challenge in human pathophysiology. Of human organs, the kidney is highly sensitive to I/R because of its high oxygen demand and poor regenerative capacity. Previous studies have shown that targeting the hypusination pathway of eIF5A through GC7 greatly improves ischemic tolerance and can be applied successfully to kidney transplants. The protection process correlates with a metabolic shift from oxidative phosphorylation to glycolysis. Because the protein kinase B Akt is involved in ischemic protective mechanisms and glucose metabolism, we looked for a link between the effects of GC7 and Akt in proximal kidney cells exposed to anoxia or the mitotoxic myxothiazol. We found that GC7 treatment resulted in impaired Akt phosphorylation at the Ser473 and Thr308 sites, so the effects of direct Akt inhibition as a preconditioning protocol on ischemic tolerance were investigated. We evidenced that Akt inhibitors provide huge protection for kidney cells against ischemia and myxothiazol. The pro-survival effect of Akt inhibitors, which is reversible, implied a decrease in mitochondrial ROS production but was not related to metabolic changes or an antioxidant defense increase. Therefore, the inhibition of Akt can be considered as a preconditioning treatment against ischemia.
Collapse
Affiliation(s)
- Nicolas Melis
- Laboratory of Cellular and Molecular Biology, Center for Cancer Research, National Cancer Institute, Bethesda, MD 20892, USA;
| | - Romain Carcy
- Université Côte d’Azur, CNRS, LP2M, 06103 Nice, France; (R.C.); (I.R.); (M.C.); (C.D.); (M.T.)
- CHU Nice, Hôpital Pasteur 2, Service de Réanimation Polyvalente et Service de Réanimation des Urgences Vitales, 06103 Nice, France
- Laboratories of Excellence Ion Channel Science and Therapeutics, 06103 Nice, France
| | - Isabelle Rubera
- Université Côte d’Azur, CNRS, LP2M, 06103 Nice, France; (R.C.); (I.R.); (M.C.); (C.D.); (M.T.)
- Laboratories of Excellence Ion Channel Science and Therapeutics, 06103 Nice, France
| | - Marc Cougnon
- Université Côte d’Azur, CNRS, LP2M, 06103 Nice, France; (R.C.); (I.R.); (M.C.); (C.D.); (M.T.)
- Laboratories of Excellence Ion Channel Science and Therapeutics, 06103 Nice, France
| | - Christophe Duranton
- Université Côte d’Azur, CNRS, LP2M, 06103 Nice, France; (R.C.); (I.R.); (M.C.); (C.D.); (M.T.)
- Laboratories of Excellence Ion Channel Science and Therapeutics, 06103 Nice, France
| | - Michel Tauc
- Université Côte d’Azur, CNRS, LP2M, 06103 Nice, France; (R.C.); (I.R.); (M.C.); (C.D.); (M.T.)
- Laboratories of Excellence Ion Channel Science and Therapeutics, 06103 Nice, France
| | - Didier F. Pisani
- Université Côte d’Azur, CNRS, LP2M, 06103 Nice, France; (R.C.); (I.R.); (M.C.); (C.D.); (M.T.)
- Laboratories of Excellence Ion Channel Science and Therapeutics, 06103 Nice, France
- Correspondence:
| |
Collapse
|