1
|
Gheitasi I, Akbari G, Savari F. Physiological and cellular mechanisms of ischemic preconditioning microRNAs-mediated in underlying of ischemia/reperfusion injury in different organs. Mol Cell Biochem 2025; 480:855-868. [PMID: 39001984 DOI: 10.1007/s11010-024-05052-7] [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: 03/28/2024] [Accepted: 06/10/2024] [Indexed: 07/15/2024]
Abstract
Ischemia-reperfusion (I/R) injury, as a pathological phenomenon, takes place when blood supply to an organ is disrupted and then aggravated during restoration of blood flow. Ischemic preconditioning (IPC) is a potent method for attenuating subsequent events of IR damage in numerous organs. IPC protocol is determined by a brief and sequential time periods of I/R before the main ischemia. MicroRNAs are endogenous non-coding RNAs that regulate post-transcriptionally target mRNA translation via degrading it and/or suppressing protein synthesis. This review introduces the physiological and cellular mechanisms of ischemic preconditioning microRNAs-mediated after I/R insult in different organs such as the liver, kidney, heart, brain, and intestine. Data of this review have been collected from the scientific articles published in databases such as Science Direct, Scopus, PubMed, Web of Science, and Scientific Information Database from 2000 to 2023. Based on these literature studies, IPC/IR intervention can affect cellular mechanisms including oxidative stress, apoptosis, angiogenesis, and inflammation through up-regulation or down-regulation of multiple microRNAs and their target genes.
Collapse
Affiliation(s)
- Izadpanah Gheitasi
- Department of Physiology, Faculty of Medicine, Yasuj University of Medical Sciences, Yasuj, Iran
| | - Ghaidafeh Akbari
- Department of Physiology, Faculty of Medicine, Yasuj University of Medical Sciences, Yasuj, Iran.
| | - Feryal Savari
- Department of Medical Basic Sciences, Shoushtar Faculty of Medical Sciences, Shoushtar, Iran.
| |
Collapse
|
2
|
Li H, Wang S, An S, Gao B, Wu D, Li Y. Hydrogen sulphide reduces renal ischemia-reperfusion injury by enhancing autophagy and reducing oxidative stress. Nephrology (Carlton) 2024; 29:645-654. [PMID: 39075751 DOI: 10.1111/nep.14368] [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: 03/13/2024] [Revised: 06/11/2024] [Accepted: 07/02/2024] [Indexed: 07/31/2024]
Abstract
AIM Renal ischemia-reperfusion injury (IRI) is a major cause of acute kidney injury. Hydrogen sulphide (H2S) exerts a protective effect in renal IRI. The present study was carried out to investigate the effects of exogenous H2S on renal IRI by regulating autophagy in mice. METHODS Mice were randomly assigned to control, IRI and NaHS (an H2S donor, 28, 56 and 100 μmol/kg) groups. Renal IRI was induced by clamping the bilateral renal pedicles with non-traumatic arterial clamp for 45 min and then reperfused for 24 h. Mice were administered intraperitoneally with NaHS 20 min prior to renal ischemia. Sham group mice underwent the same procedures without clamping. Serum and kidney tissues were harvested 24 h after reperfusion for functional, histological, oxidative stress, and autophagic determination. RESULTS Compared with the control group, the concentrations of serum creatinine (Scr), blood urea nitrogen (BUN), and malondialdehyde (MDA), the protein levels of LC3II/I, Beclin-1 and P62, as well as the number of autophagosomes were significantly increased, but the activity of superoxide dismutase (SOD) was decreased after renal IRI. NaHS pre-treatment dramatically attenuated renal IRI-induced renal dysfunction, histological changes, MDA concentration and p62 expression in a dose-dependent manner. However, NaHS increased the SOD activity and the protein levels of LC3II/I and Beclin-1. CONCLUSION These results indicate that exogenous H2S protects the kidney from IRI through enhancement of autophagy and reduction of oxidative stress. Novel H2S donors could be developed in the treatment of renal IRI.
Collapse
Affiliation(s)
- Hui Li
- Joint National Laboratory of Antibody Drug Engineering, Henan University, Kaifeng, Henan, China
| | - Shuaiwei Wang
- International Laboratory for Sepsis Research, Huaihe Hospital, Henan University, Kaifeng, Henan, China
| | - Shuangshuang An
- Joint National Laboratory of Antibody Drug Engineering, Henan University, Kaifeng, Henan, China
| | - Biao Gao
- Kaifeng Central Hospital, Kaifeng, Henan, China
| | - Dongdong Wu
- School of Basic Medical Sciences, Henan University, Kaifeng, Henan, China
| | - Yanzhang Li
- School of Basic Medical Sciences, Henan University, Kaifeng, Henan, China
| |
Collapse
|
3
|
Golmohammadi M, Ivraghi MS, Hasan EK, Huldani H, Zamanian MY, Rouzbahani S, Mustafa YF, Al-Hasnawi SS, Alazbjee AAA, Khalajimoqim F, Khalaj F. Protective effects of pioglitazone in renal ischemia-reperfusion injury (RIRI): focus on oxidative stress and inflammation. Clin Exp Nephrol 2024; 28:955-968. [PMID: 38935212 DOI: 10.1007/s10157-024-02525-3] [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: 04/03/2024] [Accepted: 06/01/2024] [Indexed: 06/28/2024]
Abstract
BACKGROUND Renal ischemia-reperfusion injury (RIRI) is a critical phenomenon that compromises renal function and is the most serious health concern related to acute kidney injury (AKI). Pioglitazone (Pio) is a known agonist of peroxisome proliferator-activated receptor-gamma (PPAR-γ). PPAR-γ is a nuclear receptor that regulates genes involved in inflammation, metabolism, and cellular differentiation. Activation of PPAR-γ is associated with antiinflammatory and antioxidant effects, which are relevant to the pathophysiology of RIRI. This study aimed to investigate the protective effects of Pio in RIRI, focusing on oxidative stress and inflammation. METHODS We conducted a comprehensive literature search using electronic databases, including PubMed, ScienceDirect, Web of Science, Scopus, and Google Scholar. RESULTS The results of this study demonstrated that Pio has antioxidant, anti-inflammatory, and anti-apoptotic activities that counteract the consequences of RIRI. The study also discussed the underlying mechanisms, including the modulation of various pathways such as TNF-α, NF-κB signaling systems, STAT3 pathway, KIM-1 and NGAL pathways, AMPK phosphorylation, and autophagy flux. Additionally, the study presented a summary of various animal studies that support the potential protective effects of Pio in RIRI. CONCLUSION Our findings suggest that Pio could protect the kidneys from RIRI by improving antioxidant capacity and decreasing inflammation. Therefore, these findings support the potential of Pio as a therapeutic strategy for preventing RIRI in different clinical conditions.
Collapse
Affiliation(s)
- Maryam Golmohammadi
- School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, 1988873554, Iran
| | | | | | - Huldani Huldani
- Department of Physiology, Faculty of Medicine Lambung, Mangkurat University, South Kalimantan, Banjarmasin, Indonesia
| | - Mohammad Yasin Zamanian
- Urology and Nephrology Research Center, Hamadan University of Medical Sciences, Hamadan, Iran.
- Department of Physiology, Hamadan University of Medical Sciences, Hamadan, 6718773654, Iran.
- Department of Pharmacology and Toxicology, School of Pharmacy, Hamadan University of Medical Sciences, Hamadan, 6718773654, Iran.
| | - Shiva Rouzbahani
- Miller School of Medicine, Bascom Palmer Eye Institute, University of Miami, Miami, FL, USA
- Department of Community Medicine and Family Physician, School of Medicine, Isfahan University of Medical Sciences, Hezar Jarib Blvd, Isfahan, Iran
| | - Yasser Fakri Mustafa
- Department of Pharmaceutical Chemistry, College of Pharmacy, University of Mosul, Mosul, 41001, Iraq
| | | | | | - Faranak Khalajimoqim
- Department of Pharmacology and Toxicology, School of Pharmacy, Hamadan University of Medical Sciences, Hamadan, 6718773654, Iran
| | - Fattaneh Khalaj
- Digestive Diseases Research Center, Shariati Hospital, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
4
|
Jeong K, Je J, Dusabimana T, Kim H, Park SW. Early Growth Response 1 Contributes to Renal IR Injury by Inducing Proximal Tubular Cell Apoptosis. Int J Mol Sci 2023; 24:14295. [PMID: 37762598 PMCID: PMC10532368 DOI: 10.3390/ijms241814295] [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: 08/31/2023] [Revised: 09/17/2023] [Accepted: 09/18/2023] [Indexed: 09/29/2023] Open
Abstract
Renal ischemia-reperfusion (IR) causes acute kidney injury due to oxidative stress, tubular inflammation, and apoptosis. Early growth response 1 (Egr-1) is a transcription factor belonging to the immediate early gene family and is known to regulate cell proliferation, differentiation, and survival. Egr-1 expression is induced during renal IR; however, its pathogenic role and underlying mechanisms remain elusive. Here, we investigated the function of Egr-1 during renal IR using C57BL/6 mice and cultured renal proximal tubular HK-2 cells. Egr-1 expression increased immediately, 1-4 h after IR, whereas plasma creatinine and oxidative stress increased progressively over 24 h after IR. Egr-1 overexpression showed greater increases in plasma creatinine, renal tubular injury, and apoptosis than in the control after IR. Egr-1 overexpression also showed significant neutrophil infiltration and increased pro-inflammatory cytokines (TNF-α, MIP-2, and IL-6) after IR. Consistently, proximal tubular HK-2 cells showed immediate induction of Egr-1 at 1 h after hypoxia and reoxygenation, where its downstream target, p53, was also increased. Interestingly, Egr-1 overexpression enhanced p53 levels and tubular apoptosis, while the knockdown of Egr-1 reduced p53 levels and tubular apoptosis after H2O2 treatment. Egr-1 was recruited to the p53 promoter, which activates p53 transcription, and Egr-1 induction occurred through Erk/JNK signaling kinases, as the specific inhibitors blocked its expression. Taken together, these results show that Egr-1 is upregulated in proximal tubular cells and contributes to renal IR injury by inducing tubular apoptosis, mediated by p53 transcriptional activation. Thus, Egr-1 could be a potential therapeutic target for renal IR injury.
Collapse
Affiliation(s)
- Kyuho Jeong
- Department of Biochemistry, Dongguk University College of Medicine, Gyeongju 38066, Republic of Korea;
| | - Jihyun Je
- Department of Pharmacology, Institute of Medical Science, Gyeongsang National University College of Medicine, Jinju 52727, Republic of Korea; (J.J.); (T.D.)
- Antiaging Bio Cell Factory-Regional Leading Research Center (ABC-RLRC), Gyeongsang National University, Jinju 52828, Republic of Korea
| | - Theodomir Dusabimana
- Department of Pharmacology, Institute of Medical Science, Gyeongsang National University College of Medicine, Jinju 52727, Republic of Korea; (J.J.); (T.D.)
- Antiaging Bio Cell Factory-Regional Leading Research Center (ABC-RLRC), Gyeongsang National University, Jinju 52828, Republic of Korea
| | - Hwajin Kim
- Department of Pharmacology, Institute of Medical Science, Gyeongsang National University College of Medicine, Jinju 52727, Republic of Korea; (J.J.); (T.D.)
- Antiaging Bio Cell Factory-Regional Leading Research Center (ABC-RLRC), Gyeongsang National University, Jinju 52828, Republic of Korea
| | - Sang Won Park
- Department of Pharmacology, Institute of Medical Science, Gyeongsang National University College of Medicine, Jinju 52727, Republic of Korea; (J.J.); (T.D.)
- Antiaging Bio Cell Factory-Regional Leading Research Center (ABC-RLRC), Gyeongsang National University, Jinju 52828, Republic of Korea
- Department of Convergence Medical Science, Gyeongsang National University Graduate School, Jinju 52727, Republic of Korea
| |
Collapse
|
5
|
Ortega-Trejo JA, Bobadilla NA. Is Renal Ischemic Preconditioning an Alternative to Ameliorate the Short- and Long-Term Consequences of Acute Kidney Injury? Int J Mol Sci 2023; 24:ijms24098345. [PMID: 37176051 PMCID: PMC10178892 DOI: 10.3390/ijms24098345] [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: 03/14/2023] [Revised: 04/21/2023] [Accepted: 04/26/2023] [Indexed: 05/15/2023] Open
Abstract
Acute kidney injury (AKI) is a global health problem and has recently been recognized as a risk factor for developing chronic kidney disease (CKD). Unfortunately, there are no effective treatments to reduce or prevent AKI, which results in high morbidity and mortality rates. Ischemic preconditioning (IPC) has emerged as a promising strategy to prevent, to the extent possible, renal tissue from AKI. Several studies have used this strategy, which involves short or long cycles of ischemia/reperfusion (IR) prior to a potential fatal ischemic injury. In most of these studies, IPC was effective at reducing renal damage. Since the first study that showed renoprotection due to IPC, several studies have focused on finding the best strategy to activate correctly and efficiently reparative mechanisms, generating different modalities with promising results. In addition, the studies performing remote IPC, by inducing an ischemic process in distant tissues before a renal IR, are also addressed. Here, we review in detail existing studies on IPC strategies for AKI pathophysiology and the proposed triggering mechanisms that have a positive impact on renal function and structure in animal models of AKI and in humans, as well as the prospects and challenges for its clinical application.
Collapse
Affiliation(s)
- Juan Antonio Ortega-Trejo
- Molecular Physiology Unit, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Mexico City 04510, Mexico
| | - Norma A Bobadilla
- Molecular Physiology Unit, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Mexico City 04510, Mexico
- Department of Nephrology and Mineral Metabolism, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City 14080, Mexico
| |
Collapse
|
6
|
Meng F, Ke J, Guo F, Yan J, Wang L. DhHP-6 alleviates inflammation and reduces vascular permeability by eliminating reactive oxygen species. Free Radic Res 2023; 57:325-337. [PMID: 37533406 DOI: 10.1080/10715762.2023.2243030] [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: 01/16/2023] [Revised: 07/21/2023] [Accepted: 07/21/2023] [Indexed: 08/04/2023]
Abstract
Inflammation is a defensive immune response to external stimuli. However, uncontrolled inflammation may cause potential damage to the host. Therefore, timely control of uncontrolled inflammation is particularly important. Previous studies have found that small molecules with antioxidant activity, such as peroxidase mimic enzymes, can inhibit the development of inflammation. DhHP-6 is a new peptide mimic of peroxidase previously designed by our laboratory. Here, we explored its anti-inflammatory activity in vitro and in vivo. Our results showed that treatment with DhHP-6 significantly reduced the production of reactive oxygen species (ROS), NO, IL-6, and TNF-α in RAW264.7 cells induced by lipopolysaccharides (LPS); in addition, it also blocked the phosphorylation of extracellularly regulated kinase 1 and 2 (ERK1/2) and ribosomal s6 kinase 1 (RSK1), thereby blocking the phosphorylation and degradation of IκBα, and inhibiting the nuclear translocation of p65. Interestingly, treatment with DhHP-6 blocked the phosphorylation of ERK1/2 and myosin light chain kinase (MLCK) in HUVECs induced by LPS. Finally, we found that DhHP-6 treatment significantly reduced the infiltration of immune cells in balloon model rats. Therefore, we believe that DhHP-6 is a potent inhibitor of inflammation.
Collapse
Affiliation(s)
- Fanwei Meng
- Key Laboratory for Molecular Enzymology and Engineering, Ministry of Education, Jilin University, Changchun, China
- School of Life Sciences, Jilin University, Changchun, China
- Jilin Collaborative Innovation Center for Antibody Engineering, Jilin Medical University, Jilin, China
| | - Junfeng Ke
- Key Laboratory for Molecular Enzymology and Engineering, Ministry of Education, Jilin University, Changchun, China
- School of Life Sciences, Jilin University, Changchun, China
| | - Feng Guo
- Key Laboratory for Molecular Enzymology and Engineering, Ministry of Education, Jilin University, Changchun, China
- School of Life Sciences, Jilin University, Changchun, China
| | - Jiaqing Yan
- Hospital of Stomatology, Jilin University, Changchun, China
| | - Liping Wang
- Key Laboratory for Molecular Enzymology and Engineering, Ministry of Education, Jilin University, Changchun, China
- School of Life Sciences, Jilin University, Changchun, China
- Engineering Laboratory for AIDS Vaccine, Jilin University, Changchun, China
| |
Collapse
|
7
|
Bagheri A, Radman G, Aria N, Rezaei F, Khajenouri M, Ghiabi S, Bagheri Y. The Effects of Quercetin on Apoptosis and Antioxidant Activity in a Renal Ischemia/Reperfusion Injury Animal Model. Drug Res (Stuttg) 2023. [PMID: 36972618 DOI: 10.1055/a-1999-7600] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/29/2023]
Abstract
Renal ischemia-reperfusion injury (IRI) is considered as one of the most prevalent causes of acute kidney injury (AKI), which can happen in various clinical situations including hypovolemic shock, injury, thrombo-embolism, and after a kidney transplant. This paper aims to evaluate the reno-protective effects of Quercetin in induced ischemia/reperfusion injury by regulating apoptosis-related proteins, inflammatory cytokines, MMP-2, MMP-9, and nuclear factor kappa-light-chain-enhancer inactivated B cells (NF-kB) in rats. The male Wistar rats (n=32) were randomly divided into Sham, untreated IR, and Quercetin-treated IR (gavage and intraperitoneal). Quercetin was given orally and intraperitoneally one hour before inducing ischemia-reperfusion injury . After reperfusion, blood samples and kidneys were collected to assess renal function and inflammatory cytokines, apoptotic signaling proteins, and antioxidants. Urea, creatinine, and MDA levels improved in Quercetin-treated groups with different administration methods. In addition, the activities of other antioxidant in Quercetin-treated rats were higher than those in the IR group. Further, Quercetin inhibited NF-kB signaling, apoptosis-associated factors and produced matrix metalloproteinase protein in the kidneys of rats. Based on the findings, the antioxidant, anti-inflammatory, and anti-apoptotic effects of the Quercetin diminished renal ischemia-reperfusion injury in the rats significantly. It is suggested that a single dosage of Quercetin have a reno-protective impact in the case of renal I/R injury.
Collapse
Affiliation(s)
- Amin Bagheri
- Department of Urology, Sina Hospital, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Ghazal Radman
- Molecular Medicine Research Center, Hormozgan Health Institute, Hormozgan University of Medical Sciences, Bandar Abbas, Iran
- Network of Immunity in Infection, Malignancy and Autoimmunity (NIIMA), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Negar Aria
- Department of Microbiology, School of Biology, College of Science, University of Tehran, Tehran, Iran
| | - Fatemeh Rezaei
- Faculty of Pharmacy in Hradec Kralove, Charles University, Prague, Czech Republic
| | - Mohammad Khajenouri
- Department of Biology, Faculty of Science, Ferdowsi University of Mashhad, Mashhad, Iran
| | - Shamim Ghiabi
- Department of Medical Chemistry, Faculty of Pharmacy, Tehran Medical Sciences Islamic Azad University, Tehran, Iran
| | - Yasin Bagheri
- Kidney Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
8
|
Zarnitz L, Doorschodt BM, Ernst L, Hosseinnejad A, Edgworth E, Fechter T, Theißen A, Djudjaj S, Boor P, Rossaint R, Tolba RH, Bleilevens C. Taurine as Antioxidant in a Novel Cell- and Oxygen Carrier-Free Perfusate for Normothermic Machine Perfusion of Porcine Kidneys. Antioxidants (Basel) 2023; 12:antiox12030768. [PMID: 36979015 PMCID: PMC10045130 DOI: 10.3390/antiox12030768] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2023] [Revised: 03/14/2023] [Accepted: 03/17/2023] [Indexed: 03/30/2023] Open
Abstract
Donor organ-shortage has resulted in the increased use of marginal grafts; however, normothermic machine perfusion (NMP) holds the potential for organ viability assessment and restoration of marginal grafts prior to transplantation. Additionally, cell-, oxygen carrier-free and antioxidants-supplemented solutions could potentially prevent adverse effects (transfusion reactions, inflammation, hemolysis), associated with the use of autologous packed red blood cell (pRBC)-based perfusates. This study compared 6 h NMP of porcine kidneys, using an established pRBC-based perfusate (pRBC, n = 7), with the novel cell- and oxygen carrier-free organ preservation solution Ecosol, containing taurine (Ecosol, n = 7). Despite the enhanced tissue edema and tubular injury in the Ecosol group, related to a suboptimal molecular mass of polyethylene glycol as colloid present in the solution, functional parameters (renal blood flow, intrarenal resistance, urinary flow, pH) and oxygenation (arterial pO2, absence of hypoxia-inducible factor 1-alpha) were similar to the pRBC group. Furthermore, taurine significantly improved the antioxidant capacity in the Ecosol group, reflected in decreased lactate dehydrogenase, urine protein and tubular vacuolization compared to pRBC. This study demonstrates the feasibility of 6 h NMP using a taurine containing, cell- and oxygen carrier-free perfusate, achieving a comparable organ quality to pRBC perfused porcine kidneys.
Collapse
Affiliation(s)
- Laura Zarnitz
- Department of Anesthesiology, Medical Faculty, University Hospital RWTH Aachen, 52074 Aachen, Germany
| | - Benedict M Doorschodt
- Institute for Laboratory Animal Science and Experimental Surgery, Faculty of Medicine, RWTH Aachen University, 52074 Aachen, Germany
| | - Lisa Ernst
- Institute for Laboratory Animal Science and Experimental Surgery, Faculty of Medicine, RWTH Aachen University, 52074 Aachen, Germany
| | - Aisa Hosseinnejad
- DWI-Leibniz-Institute for Interactive Materials e.V., 52056 Aachen, Germany
| | - Eileen Edgworth
- Department of Anesthesiology, Medical Faculty, University Hospital RWTH Aachen, 52074 Aachen, Germany
| | - Tamara Fechter
- Department of Anesthesiology, Medical Faculty, University Hospital RWTH Aachen, 52074 Aachen, Germany
| | - Alexander Theißen
- Department of Anesthesiology, Medical Faculty, University Hospital RWTH Aachen, 52074 Aachen, Germany
| | - Sonja Djudjaj
- Institute of Pathology & Division of Nephrology, Medical Faculty, RWTH Aachen, 52074 Aachen, Germany
| | - Peter Boor
- Institute of Pathology & Division of Nephrology, Medical Faculty, RWTH Aachen, 52074 Aachen, Germany
| | - Rolf Rossaint
- Department of Anesthesiology, Medical Faculty, University Hospital RWTH Aachen, 52074 Aachen, Germany
| | - René H Tolba
- Institute for Laboratory Animal Science and Experimental Surgery, Faculty of Medicine, RWTH Aachen University, 52074 Aachen, Germany
| | - Christian Bleilevens
- Department of Anesthesiology, Medical Faculty, University Hospital RWTH Aachen, 52074 Aachen, Germany
| |
Collapse
|
9
|
Ahmad A. Prophylactic Treatment with Hydrogen Sulphide Can Prevent Renal Ischemia-Reperfusion Injury in L-NAME Induced Hypertensive Rats with Cisplatin-Induced Acute Renal Failure. Life (Basel) 2022; 12:1819. [PMID: 36362975 PMCID: PMC9695289 DOI: 10.3390/life12111819] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2022] [Revised: 11/01/2022] [Accepted: 11/03/2022] [Indexed: 08/26/2023] Open
Abstract
(Background and Objectives): Renal ischemia perfusion injury is one of the major issues in kidney transplant. The aim of the study was to investigate the hypothesis that prophylactic treatment-with a hydrogen sulphide donor to an acute renal failure case of hypertensive rats-can minimize the ischemia reperfusion injury of the kidney which is beneficial for kidney transplant. To check this hypothesis, the present study was designed to investigate the effect of chronic administration of a hydrogen sulphide (H2S) donor and sodium hydrosulfide (NaHS) on nuclear factor kappa B (NF-kB) and inter cellular adhesion molecule-1 (ICAM-1) concentration in non-renal failure (NRF) and acute renal failure (ARF) rats in the ischemia-reperfusion injury (IRI) model of the kidney in both normotensive WKY and hypertensive rats (L-nitro arginine methyl ester (L-NAME-induced); (Materials and Methods): A total number of 48 Sprague-Dawley rats were recruited into eight groups each consisting of six animals. Each of these eight groups was used to measure systemic and renal parameters, H2S, antioxidant parameters in plasma, plasma concentration of NF-kB and ICAM-1 and renal cortical blood pressure. ARF was induced by single intraperitoneal (i.p.) cisplatin injection (5 mg/kg). Hypertension was induced by oral administration of L-NAME in drinking water for four weeks at 40 mg/kg/day. NaHS was administered (i.p) at 56 µmol/kg for five weeks while dL-propargylglycine (PAG), a H2S generation inhibitor, was administered as a single intra-peritoneal injection (50 mg/kg). An acute surgical experiment was performed for the induction of renal ischemia for 30 min by renal artery clamping followed by reperfusion for three hours; (Results): Chronic administration of NaHS attenuated the severity of ARF in both normotensive and hypertensive animals (L-NAME) along with lowering the blood pressure in hypertensive groups. NaHS improved the oxidative stress parameters such as total superoxide dismutase (T-SOD), glutathione (GSH) and reduced the malondialdehyde (MDA) concentration along with reduction of NF-kB and ICAM-1 following renal IRI; Conclusions: These findings demonstrate that H2S not only reduced the severity of cisplatin induced ARF but also reduced the severity of renal IRI by upregulating antioxidants along with decreased concentrations of NF-kB and ICAM-1 in normotensive and L-NAME induced hypertensive rats.
Collapse
Affiliation(s)
- Ashfaq Ahmad
- Department of Pharmacy Practice, College of Pharmacy, University of Hafr Al Batin, Hafr Al Batin 39524, Saudi Arabia
| |
Collapse
|
10
|
2-Mercaptoethanol protects against DNA double-strand breaks after kidney ischemia and reperfusion injury through GPX4 upregulation. Pharmacol Rep 2022; 74:1041-1053. [PMID: 35989399 DOI: 10.1007/s43440-022-00403-x] [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: 05/02/2022] [Revised: 07/06/2022] [Accepted: 07/26/2022] [Indexed: 10/25/2022]
Abstract
BACKGROUND Kidney ischemia reperfusion injury (IRI) is characterized by tubular cell death. DNA double-strand breaks is one of the major sources of tubular cell death induced by IRI. 2-Mercaptoethanol (2-ME) is protective against DNA double-strand breaks derived from calf thymus and bovine embryo. Here, we sought to determine whether treatment with 2-ME attenuated DNA double-strand breaks, resulting in reduced kidney dysfunction and structural damage in IRI. METHODS Kidney IRI or sham-operation in mice was carried out. The mice were treated with 2-ME, Ras-selective lethal 3, or vehicle. Kidney function, tubular injury, DNA damage, antioxidant enzyme expression, and DNA damage response (DDR) kinases activation were assessed. RESULTS Treatment with 2-ME significantly attenuated kidney dysfunction, tubular injury, and DNA double-strand breaks after IRI. Among DDR kinases, IRI induced phosphorylation of ataxia telangiectasia mutated (ATM) and ataxia telangiectasia and Rad3 related (ATR), but IRI reduced phosphorylation of other DDR kinases including ataxia telangiectasia and Rad3 related, checkpoint kinase 1 (Chk1), Chk2, and Chinese hamster cells 1 (XRCC1). Treatment with 2-ME enhanced phosphorylation of ATM and ATM-mediated effector kinases in IRI-subjected kidneys, suggesting that 2-ME activates ATM-mediated DDR signaling pathway. Furthermore, 2-ME dramatically upregulated glutathione peroxidase 4 (GPX4) in IRI-subjected kidneys. Inhibition of GPX4 augmented adverse IRI consequences including kidney dysfunction, tubular injury, DNA double-strand breaks, and inactivation of ATM-mediated DDR signaling pathway after IRI in 2-ME-treated kidneys. CONCLUSIONS We have demonstrated that exogenous 2-ME protects against DNA double-strand breaks after kidney IRI through GPX4 upregulation and ATM activation.
Collapse
|
11
|
Barakat M, Hussein AM, Salama MF, Awadalla A, Barakat N, Serria M, El-Shafey M, El-Sherbiny M, El Adl MA. Possible Underlying Mechanisms for the Renoprotective Effect of Retinoic Acid-Pretreated Wharton's Jelly Mesenchymal Stem Cells against Renal Ischemia/Reperfusion Injury. Cells 2022; 11:1997. [PMID: 35805083 PMCID: PMC9266019 DOI: 10.3390/cells11131997] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2022] [Revised: 06/18/2022] [Accepted: 06/20/2022] [Indexed: 11/18/2022] Open
Abstract
Objectives: The current work investigated the effect of Wharton jelly mesenchymal stem cells (WJ-MSCs) pretreated with all-trans-retinoic acid (ATRA) on renal ischemia in rats and the possible role of oxidative stress, apoptotic and Wnt/β-Catenin signaling pathways, and inflammatory cytokines in their effects. Methods: The study included 90 male Sprague Dawley rats that were allocated to five groups (n = 18 rats): (I) Sham-operated group (right nephrectomy was performed); (II) Ischemia/reperfusion injury (IRI) group, a sham group with 45-min renal ischemia on the left kidney; (III) ATRA group, an ischemic group with an intravenous (i.v.) administration of ATRA 10 µM, 10 min post-surgery); (IV) WJ-MSCs group, an IRI group with an i.v. administration of 150 µL containing 7 × 106 WJ-MSCs, 10 min post-surgery; (V) WJ-MSCs + ATRA group, an IRI group with an i.v. administration of 150 µL of 7 × 106 WJ-MSCs pretreated with 10 µM ATRA. At the end of the experiments, serum creatinine, BUN micro-albuminuria (MAU), urinary protein, markers of redox state in the left kidney (MDA, CAT, SOD, and GSH), and the expression of Bax, IL-6, HIF-1α, Wnt7B, and β-catenin genes at the level of mRNA as well as for immunohistochemistry for NFkB and β-Catenin markers were analyzed. Results: The current study found that 45-min of renal ischemia resulted in significant impairment of kidney function (evidenced by the increase in serum creatinine, BUN, and urinary proteins) and deterioration of the kidney morphology, which was associated with a significant increase in redox state (evidenced by an increase in MDA and a decrease in GSH, SOD, and CAT), and a significant increase in inflammatory and apoptotic processes (evidenced by an increase in Bax and IL-6, NFkB, Wnt7B, β-catenin and HIF-1α) in kidney tissues (p < 0.05). On the other hand, treatment with ATRA, WJ-MSCs, or a combination of both, caused significant improvement in kidney function and morphology, which was associated with significant attenuation of oxidative stress, apoptotic markers, and inflammatory cytokines (IL6 and NFkB) with the upregulation of HIF-1α and β-catenin in kidney tissues (p < 0.05). Moreover, the renoprotective effect of WJ-MSCs pretreated with ATRA was more potent than WJ-MSCs alone. Conclusions: It is concluded that preconditioning of WJ-MSCs with ATRA may enhance their renoprotective effect. This effect could be due to the upregulation of the beta-catenin/Wnt pathway and attenuation of apoptosis, inflammation, and oxidative stress.
Collapse
Affiliation(s)
- Mai Barakat
- Department of Biochemistry, Faculty of Veterinary Medicine, Mansoura University, Mansoura 35516, Egypt; (M.B.); (M.F.S.); (M.A.E.A.)
- Institute of Global Public Health and Human Ecology, School of Science and Engineering, American University, Cairo 11835, Egypt
| | - Abdelaziz M. Hussein
- Department of Medical Physiology, Faculty of Medicine, Mansoura University, Mansoura 35516, Egypt
| | - Mohamed F. Salama
- Department of Biochemistry, Faculty of Veterinary Medicine, Mansoura University, Mansoura 35516, Egypt; (M.B.); (M.F.S.); (M.A.E.A.)
| | - Amira Awadalla
- Urology and Nephrology Center, Mansoura University, Mansoura 35516, Egypt; (A.A.); (N.B.)
| | - Nashwa Barakat
- Urology and Nephrology Center, Mansoura University, Mansoura 35516, Egypt; (A.A.); (N.B.)
| | - Mohamed Serria
- Department of Biochemistry, Faculty of Medicine, Mansoura University, Mansoura 35516, Egypt;
| | - Mohamed El-Shafey
- Department of Anatomy and Embryology, Faculty of Medicine, Mansoura University, Mansoura 35516, Egypt;
- Physiological Sciences Department, Fakeeh College for Medical Sciences, Jeddah 21461, Saudi Arabia
| | - Mohamed El-Sherbiny
- Department of Basic Medical Sciences, College of Medicine, AlMaarefa University, Riyadh 71666, Saudi Arabia;
| | - Mohamed A. El Adl
- Department of Biochemistry, Faculty of Veterinary Medicine, Mansoura University, Mansoura 35516, Egypt; (M.B.); (M.F.S.); (M.A.E.A.)
| |
Collapse
|
12
|
Vascular Endothelial Dysfunction in the Thoracic Aorta of Rats with Ischemic Acute Kidney Injury: Contribution of Indoxyl Sulfate. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:7547269. [PMID: 35251481 PMCID: PMC8896937 DOI: 10.1155/2022/7547269] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Revised: 01/21/2022] [Accepted: 02/09/2022] [Indexed: 01/04/2023]
Abstract
Chronic kidney disease (CKD) and cardiovascular disease are known to be linked, and the involvement of indoxyl sulfate (IS), a type of uremic toxin, has been suggested as one of the causes. It is known that IS induces vascular dysfunction through overproduction of reactive oxygen species (ROS). On the other hand, the involvement of IS in the vascular dysfunction associated with acute kidney injury (AKI) is not fully understood. Therefore, we investigated this issue using the thoracic aorta of rats with ischemic AKI. Ischemic AKI was induced by occlusion of the left renal artery and vein for 45 min, followed by reperfusion 2 weeks after contralateral nephrectomy. One day after reperfusion, there was marked deterioration in renal function evidenced by an increase in plasma creatinine. Furthermore, blood IS levels increased markedly due to worsening renal function. Seven days and 28 days after reperfusion, blood IS levels decreased with the improvement in renal function. Of note, acetylcholine-induced vasorelaxation deteriorated over time after reperfusion, contradicting the recovery of renal function. In addition, 28 days after reperfusion, we observed a significant increase in ROS production in the vascular tissue. Next, we administered AST-120, a spherical adsorbent charcoal, after reperfusion to assess whether the vascular endothelial dysfunction associated with the ischemic AKI was due to a temporary increase in blood IS levels. AST-120 reduced the temporary increase in blood IS levels after reperfusion without influencing renal function, but did not restore the impaired vascular reactivity. Thus, in ischemic AKI, we confirmed that the vascular endothelial function of the thoracic aorta is impaired even after the recovery of kidney injury, probably with excessive ROS production. IS, which increases from ischemia to early after reperfusion, may not be a major contributor to the vascular dysfunction associated with ischemic AKI.
Collapse
|
13
|
Dun RL, Lan TY, Tsai J, Mao JM, Shao YQ, Hu XH, Zhu WJ, Qi GC, Peng Y. Protective Effect of Melatonin for Renal Ischemia-Reperfusion Injury: A Systematic Review and Meta-Analysis. Front Physiol 2022; 12:791036. [PMID: 35095558 PMCID: PMC8793910 DOI: 10.3389/fphys.2021.791036] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2021] [Accepted: 12/16/2021] [Indexed: 12/21/2022] Open
Abstract
Background: Renal ischemia-reperfusion (I/R) injury is one of the major causes related to acute kidney damage. Melatonin has been shown as a powerful antioxidant, with many animal experiments have been designed to evaluate the therapeutic effect of it to renal I/R injury. Objectives: This systematic review aimed to assess the therapeutic effect of melatonin for renal I/R injury in animal models. Methods and Results: The PubMed, Web of Science, Embase, and Science Direct were searched for animal experiments applying melatonin to treat renal I/R injury to February 2021. Thirty-one studies were included. The pooled analysis showed a greater reduction of blood urea nitrogen (BUN) (21 studies, weighted mean difference (WMD) = −30.00 [−42.09 to −17.91], p < 0.00001), and serum creatinine (SCr) (20 studies, WMD = −0.91 [−1.17 to −0.66], p < 0.00001) treated with melatonin. Subgroup analysis suggested that multiple administration could reduce the BUN compared with control. Malondialdehyde and myeloperoxidase were significantly reduced, meanwhile, melatonin significantly improved the activity of glutathione, as well as superoxide dismutase. The possible mechanism for melatonin to treat renal I/R injury is inhibiting endoplasmic reticulum stress, apoptosis, inflammation, autophagy, and fibrillation in AKI to chronic kidney disease. Conclusions: From the available data of small animal studies, this systematic review demonstrated that melatonin could improve renal function and antioxidative effects to cure renal I/R injury through, then multiple administration of melatonin might be more appropriate. Nonetheless, extensive basic experiments are need to study the mechanism of melatonin, then well-designed randomized controlled trials to explore the protective effect of melatonin.
Collapse
Affiliation(s)
- Rong-liang Dun
- Urology Surgery, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Tian-ying Lan
- Nephrology, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Jennifer Tsai
- Urology Surgery, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Jian-min Mao
- Urology Surgery, Shanghai Seventh People's Hospital, Shanghai, China
| | - Yi-qun Shao
- Urology Surgery, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Xiao-hua Hu
- Urology Surgery, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Wen-jing Zhu
- Urology Surgery, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Guang-chong Qi
- Urology Surgery, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yu Peng
- Urology Surgery, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- *Correspondence: Yu Peng
| |
Collapse
|
14
|
Liu Z, Li Y, Li C, Yu L, Chang Y, Qu M. Delivery of coenzyme Q10 with mitochondria-targeted nanocarrier attenuates renal ischemia-reperfusion injury in mice. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2021; 131:112536. [PMID: 34857313 DOI: 10.1016/j.msec.2021.112536] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/14/2021] [Revised: 10/18/2021] [Accepted: 11/02/2021] [Indexed: 10/19/2022]
Abstract
Ischemia-reperfusion (I/R) injury causes high morbidity, mortality, and healthcare costs. I/R induces acute kidney injury through exacerbating the mitochondrial damage and increasing inflammatory and oxidative responses. Here, we developed the mitochondria-targeted nanocarrier to delivery of Coenzyme Q10 (CoQ10) for renal I/R treatment in animal model. The mitochondria-targeted TPP CoQ10 nanoparticles (T-NPCoQ10) were synthesized through ABC miktoarm polymers method and characterized by dynamic light scattering (DLS) and transmission electron microscopy (TEM). The I/R mouse model and oxygen-glucose deprivation/reperfusion (D/R) model were created to examine the role of T-NPCoQ10 on renal I/R. Mitochondrial DNA damage, apoptosis, and inflammatory cytokines were measured in I/R injury mice. Plasma creatinine, urea nitrogen, tubular injury score was tested to assess the renal function. T-NPCoQ10 nanoparticles could be delivered to renal mitochondria preciously and efficiently. T-NPCoQ10 administration attenuated oxidative injury in both cell and animal models significantly, alleviated mtDNA damage, suppressed inflammatory and apoptotic responses, and improved renal function. The mitochondria specific CoQ10 delivery provided a precious and efficient method for protecting inflammatory and oxidative responses of I/R-induced renal damage.
Collapse
Affiliation(s)
- Zhaohui Liu
- Department of Anesthesiology, Cangzhou Central Hospital, Teaching Hospital of Tianjin Medical University, Cangzhou 061000, Hebei, China.
| | - Yan Li
- Department of Anesthesiology, Cangzhou Central Hospital, Teaching Hospital of Tianjin Medical University, Cangzhou 061000, Hebei, China
| | - Chunlei Li
- Department of Anesthesiology, Cangzhou Central Hospital, Teaching Hospital of Tianjin Medical University, Cangzhou 061000, Hebei, China
| | - Lili Yu
- Department of Anesthesiology, Cangzhou Central Hospital, Teaching Hospital of Tianjin Medical University, Cangzhou 061000, Hebei, China
| | - Yulin Chang
- Department of Anesthesiology, Cangzhou Central Hospital, Teaching Hospital of Tianjin Medical University, Cangzhou 061000, Hebei, China
| | - Min Qu
- Department of Anesthesiology, Cangzhou Central Hospital, Teaching Hospital of Tianjin Medical University, Cangzhou 061000, Hebei, China
| |
Collapse
|
15
|
Jeong JY, Na KR, Shin JA, Suh KS, Kim JJ, Lee KW, Choi DE. Collecting Duct-Specific CR6-Interacting Factor-1-Deletion Aggravates Renal Inflammation and Fibrosis Induced by Unilateral Ureteral Obstruction. Int J Mol Sci 2021; 22:11699. [PMID: 34769136 PMCID: PMC8584192 DOI: 10.3390/ijms222111699] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Revised: 10/24/2021] [Accepted: 10/26/2021] [Indexed: 01/17/2023] Open
Abstract
Although inflammation and fibrosis, which are key mechanisms of chronic kidney disease, are associated with mitochondrial damage, little is known about the effects of mitochondrial damage on the collecting duct in renal inflammation and fibrosis. To generate collecting duct-specific mitochondrial injury mouse models, CR6-interacting factor-1 (CRIF1) flox/flox mice were bred with Hoxb7-Cre mice. We evaluated the phenotype of these mice. To evaluate the effects on unilateral ureteral obstruction (UUO)-induced renal injury, we divided the mice into the following four groups: a CRIF1flox/flox (wild-type (WT)) group, a CRIF1flox/flox-Hob7 Cre (CRIF1-KO) group, a WT-UUO group, and a CRIF1-KO UUO group. We evaluated the blood and urine chemistries, inflammatory and fibrosis markers, light microscopy, and electron microscopy of the kidneys. The inhibition of Crif1 mRNA in mIMCD cells reduced oxygen consumption and membrane potential. No significant differences in blood and urine chemistries were observed between WT and CRIF1-KO mice. In UUO mice, monocyte chemoattractant protein-1 and osteopontin expression, number of F4/80 positive cells, transforming growth factor-β and α-smooth muscle actin staining, and Masson's trichrome staining were significantly higher in the kidneys of CRIF1-KO mice compared with the kidneys of WT mice. In sham mice, urinary 8-hydroxydeoxyguanosine (8-OHDG) was higher in CRIF1-KO mice than in WT mice. Moreover, CRIF1-KO sham mice had increased 8-OHDG-positive cell recruitment compared with WT-sham mice. CRIF1-KO-UUO kidneys had increased recruitment of 8-OHDG-positive cells compared with WT-UUO kidneys. In conclusion, collecting duct-specific mitochondrial injury increased oxidative stress. Oxidative stress associated with mitochondrial damage may aggravate UUO-induced renal injury.
Collapse
Affiliation(s)
- Jin Young Jeong
- Department of Nephrology, Chungnam National University School of Medicine, Daejeon 35015, Korea; (J.Y.J.); (K.R.N.); (J.-J.K.); (K.W.L.)
| | - Ki Ryang Na
- Department of Nephrology, Chungnam National University School of Medicine, Daejeon 35015, Korea; (J.Y.J.); (K.R.N.); (J.-J.K.); (K.W.L.)
| | - Jin Ah Shin
- Department of Medical Science, Chungnam National University School of Medicine, Daejeon 35015, Korea;
| | - Kwang-Sun Suh
- Department of Pathology, Chungnam National University School of Medicine, Daejeon 35015, Korea;
| | - Jwa-Jin Kim
- Department of Nephrology, Chungnam National University School of Medicine, Daejeon 35015, Korea; (J.Y.J.); (K.R.N.); (J.-J.K.); (K.W.L.)
| | - Kang Wook Lee
- Department of Nephrology, Chungnam National University School of Medicine, Daejeon 35015, Korea; (J.Y.J.); (K.R.N.); (J.-J.K.); (K.W.L.)
| | - Dae Eun Choi
- Department of Nephrology, Chungnam National University School of Medicine, Daejeon 35015, Korea; (J.Y.J.); (K.R.N.); (J.-J.K.); (K.W.L.)
- Department of Medical Science, Chungnam National University School of Medicine, Daejeon 35015, Korea;
| |
Collapse
|
16
|
Bausys A, Maneikyte J, Leber B, Weber J, Feldbacher N, Strupas K, Dschietzig TB, Schemmer P, Stiegler P. Custodiol ® Supplemented with Synthetic Human Relaxin Decreases Ischemia-Reperfusion Injury after Porcine Kidney Transplantation. Int J Mol Sci 2021; 22:11417. [PMID: 34768845 PMCID: PMC8583819 DOI: 10.3390/ijms222111417] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2021] [Revised: 10/17/2021] [Accepted: 10/19/2021] [Indexed: 01/04/2023] Open
Abstract
Objective. Ischemia-reperfusion injury (IRI) is inevitable after kidney transplantation (KT), impairing outcomes. Relaxin-2 (RLX) is a promising insulin-related peptide hormone that protects against renal IRI in rodents, although large animal models are needed before RLX can be tested in a human setting. Methods. In this blinded, randomized, and placebo-controlled experimental study kidneys from 19 donor pigs were retrieved after perfusion with Custodiol® ± RLX (5 or 20 nmol/L) and underwent static cold storage (SCS) for 24 and 48 h, respectively. Subsequently, KT was performed after unilateral right nephrectomy. Study outcomes included markers for kidney function, oxidative stress, lipid peroxidation, and endothelial cell damage. PCR analysis for oxidative stress and apoptosis-related gene panels as well as immunohistochemistry were performed. Results. RLX upregulated SOD2 and NFKB expression to 135% (p = 0.042) and 125% (p = 0.019), respectively, while RIPK1 expression was downregulated to 82% (p = 0.016) of corresponding controls. Further RLX significantly downregulated RIPK1 and MLKL expression and decreased the number of Caspase 3- and MPO-positive cells in grafts after SCS. Conclusions. RLX supplemented Custodiol® significantly decreased IRI via both antioxidant and anti-apoptotic mechanisms. Clinical trials are warranted to implement synthetic human RLX as a novel additive to preservation solutions against IRI.
Collapse
Affiliation(s)
- Augustinas Bausys
- General, Visceral and Transplant Surgery, Department of Surgery, Medical University of Graz, 8036 Graz, Austria; (A.B.); (J.M.); (J.W.); (N.F.); (P.S.); (P.S.)
- Faculty of Medicine, Vilnius University, 01513 Vilnius, Lithuania;
- Department of Abdominal Surgery, National Cancer Institute, 10224 Vilnius, Lithuania
| | - Juste Maneikyte
- General, Visceral and Transplant Surgery, Department of Surgery, Medical University of Graz, 8036 Graz, Austria; (A.B.); (J.M.); (J.W.); (N.F.); (P.S.); (P.S.)
- Faculty of Medicine, Vilnius University, 01513 Vilnius, Lithuania;
| | - Bettina Leber
- General, Visceral and Transplant Surgery, Department of Surgery, Medical University of Graz, 8036 Graz, Austria; (A.B.); (J.M.); (J.W.); (N.F.); (P.S.); (P.S.)
| | - Jennifer Weber
- General, Visceral and Transplant Surgery, Department of Surgery, Medical University of Graz, 8036 Graz, Austria; (A.B.); (J.M.); (J.W.); (N.F.); (P.S.); (P.S.)
| | - Nicole Feldbacher
- General, Visceral and Transplant Surgery, Department of Surgery, Medical University of Graz, 8036 Graz, Austria; (A.B.); (J.M.); (J.W.); (N.F.); (P.S.); (P.S.)
| | - Kestutis Strupas
- Faculty of Medicine, Vilnius University, 01513 Vilnius, Lithuania;
| | - Thomas Bernd Dschietzig
- Relaxera GmbH & Co. KG, 64625 Bensheim, Germany;
- MHB Medizinische Hochschule Brandenburg, 16816 Neuruppin, Germany
| | - Peter Schemmer
- General, Visceral and Transplant Surgery, Department of Surgery, Medical University of Graz, 8036 Graz, Austria; (A.B.); (J.M.); (J.W.); (N.F.); (P.S.); (P.S.)
| | - Philipp Stiegler
- General, Visceral and Transplant Surgery, Department of Surgery, Medical University of Graz, 8036 Graz, Austria; (A.B.); (J.M.); (J.W.); (N.F.); (P.S.); (P.S.)
| |
Collapse
|
17
|
Hashmi SF, Rathore HA, Sattar MA, Johns EJ, Gan CY, Chia TY, Ahmad A. Hydrogen Sulphide Treatment Prevents Renal Ischemia-Reperfusion Injury by Inhibiting the Expression of ICAM-1 and NF-kB Concentration in Normotensive and Hypertensive Rats. Biomolecules 2021; 11:1549. [PMID: 34680182 PMCID: PMC8534271 DOI: 10.3390/biom11101549] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2021] [Revised: 10/13/2021] [Accepted: 10/14/2021] [Indexed: 01/13/2023] Open
Abstract
Our main objective was to investigate the effect of chronic administration of hydrogen sulphide donor (sodium hydrosulphide) on the expression of intercellular adhesion molecule-1 (ICAM-1) and concentration of nuclear factor-kappa B (NF-kB) in a renal ischemia-reperfusion injury (IRI) model of WKY and L-nitro-arginine-methyl-ester (L-NAME)-induced hypertensive rats. Sodium hydrosulphide (NaHS) was administered intraperitoneally (i.p.) for 35 days while cystathionine gamma lyase (CSE) inhibitor dL-propargylglycine (PAG) was administered at a single dose of 50 mg/kg. Animals were anesthetised using sodium pentobarbitone (60 mg/kg) and then prepared to induce renal ischemia by clamping the left renal artery for 30 min followed by 3 h of reperfusion. Pre-treatment with NaHS improved the renal functional parameters in both WKY and L-NAME-induced hypertensive rats along with reduction of blood pressure in hypertensive groups. Oxidative stress markers like malondialdehyde (MDA), total superoxide dismutase (T-SOD) and glutathione (GSH) were also improved by NaHS treatment following renal IRI. Levels of ICAM-1 and NF-kB concentration were reduced by chronic treatment with NaHS and increased by PAG administration after renal IRI in plasma and kidney. Treatment with NaHS improved tubular morphology and glomerulus hypertrophy. Pre-treatment with NaHS reduced the degree of renal IRI by potentiating its antioxidant and anti-inflammatory mechanism, as evidenced by decreased NF-kB concentration and downregulation of ICAM-1 expression.
Collapse
Affiliation(s)
- Syed F. Hashmi
- School of Pharmaceutical Sciences, Universiti Sains Malaysia, Penang 11800, Malaysia; (S.F.H.); (H.A.R.); (M.A.S.)
| | - Hassaan Anwer Rathore
- School of Pharmaceutical Sciences, Universiti Sains Malaysia, Penang 11800, Malaysia; (S.F.H.); (H.A.R.); (M.A.S.)
| | - Munavvar A. Sattar
- School of Pharmaceutical Sciences, Universiti Sains Malaysia, Penang 11800, Malaysia; (S.F.H.); (H.A.R.); (M.A.S.)
| | - Edward J. Johns
- Department of Physiology, University College Cork, T12 K8AF Cork, Ireland;
| | - Chee-Yuen Gan
- Analytical Biochemistry Research Centre (ABrC), Universiti Sains Malaysia (USM), Lebuh Bukit Jambul, Penang 11900, Malaysia;
| | - Tan Yong Chia
- Analytical Biochemistry Research Centre (ABrC), Universiti Sains Malaysia (USM), Lebuh Bukit Jambul, Penang 11900, Malaysia;
| | - Ashfaq Ahmad
- School of Pharmaceutical Sciences, Universiti Sains Malaysia, Penang 11800, Malaysia; (S.F.H.); (H.A.R.); (M.A.S.)
- Department of Pharmacy Practice, College of Pharmacy, University of Hafr Al-Batin, Hafr Al-Batin 31991, Saudi Arabia
| |
Collapse
|
18
|
Abstract
The phenomenon of ischemic preconditioning was discovered in 1986 in experiments with the heart, and then it was observed in almost all organs, the kidneys included. This phenomenon is underlain by conditioning of the tissues with short ischemia/reperfusion cycles intended for subsequent exposure to pathological ischemia. Despite the kidneys are not viewed as so vital organs as the brain or the heart, the acute ischemic injury to kidneys is a widespread pathology responsible for the yearly death of almost 2 million patients, while the number of patients with chronic kidney disease is estimated as hundreds of millions or nearly 10% adult population the world over. Currently, it is believed that adaptation of the kidneys to ischemia by preconditioning is the most effective way to prevent the development of acute kidney injury, so deep insight into its molecular mechanisms will be a launch pad for creating the nephroprotective therapy by elevating renal tolerance to oxygen deficiency. This review focuses on the key signaling pathways of kidney ischemic preconditioning, the potential pharmacological mimetics of its key elements, and the limitations of this therapeutic avenue associated with age-related decline of ischemic tolerance of the kidneys.
Collapse
|
19
|
Yan LJ. NADH/NAD + Redox Imbalance and Diabetic Kidney Disease. Biomolecules 2021; 11:biom11050730. [PMID: 34068842 PMCID: PMC8153586 DOI: 10.3390/biom11050730] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2021] [Revised: 05/11/2021] [Accepted: 05/12/2021] [Indexed: 12/11/2022] Open
Abstract
Diabetic kidney disease (DKD) is a common and severe complication of diabetes mellitus. If left untreated, DKD can advance to end stage renal disease that requires either dialysis or kidney replacement. While numerous mechanisms underlie the pathogenesis of DKD, oxidative stress driven by NADH/NAD+ redox imbalance and mitochondrial dysfunction have been thought to be the major pathophysiological mechanism of DKD. In this review, the pathways that increase NADH generation and those that decrease NAD+ levels are overviewed. This is followed by discussion of the consequences of NADH/NAD+ redox imbalance including disruption of mitochondrial homeostasis and function. Approaches that can be applied to counteract DKD are then discussed, which include mitochondria-targeted antioxidants and mimetics of superoxide dismutase, caloric restriction, plant/herbal extracts or their isolated compounds. Finally, the review ends by pointing out that future studies are needed to dissect the role of each pathway involved in NADH-NAD+ metabolism so that novel strategies to restore NADH/NAD+ redox balance in the diabetic kidney could be designed to combat DKD.
Collapse
Affiliation(s)
- Liang-Jun Yan
- Department of Pharmaceutical Sciences, College of Pharmacy, University of North Texas Health Science Center, Fort Worth, TX 76107, USA
| |
Collapse
|
20
|
Tang C, Cai J, Yin XM, Weinberg JM, Venkatachalam MA, Dong Z. Mitochondrial quality control in kidney injury and repair. Nat Rev Nephrol 2021; 17:299-318. [PMID: 33235391 PMCID: PMC8958893 DOI: 10.1038/s41581-020-00369-0] [Citation(s) in RCA: 292] [Impact Index Per Article: 73.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/19/2020] [Indexed: 01/30/2023]
Abstract
Mitochondria are essential for the activity, function and viability of eukaryotic cells and mitochondrial dysfunction is involved in the pathogenesis of acute kidney injury (AKI) and chronic kidney disease, as well as in abnormal kidney repair after AKI. Multiple quality control mechanisms, including antioxidant defence, protein quality control, mitochondrial DNA repair, mitochondrial dynamics, mitophagy and mitochondrial biogenesis, have evolved to preserve mitochondrial homeostasis under physiological and pathological conditions. Loss of these mechanisms may induce mitochondrial damage and dysfunction, leading to cell death, tissue injury and, potentially, organ failure. Accumulating evidence suggests a role of disturbances in mitochondrial quality control in the pathogenesis of AKI, incomplete or maladaptive kidney repair and chronic kidney disease. Moreover, specific interventions that target mitochondrial quality control mechanisms to preserve and restore mitochondrial function have emerged as promising therapeutic strategies to prevent and treat kidney injury and accelerate kidney repair. However, clinical translation of these findings is challenging owing to potential adverse effects, unclear mechanisms of action and a lack of knowledge of the specific roles and regulation of mitochondrial quality control mechanisms in kidney resident and circulating cell types during injury and repair of the kidney.
Collapse
Affiliation(s)
- Chengyuan Tang
- Department of Nephrology, Hunan Key Laboratory of Kidney Disease and Blood Purification, The Second Xiangya Hospital at Central South University, Changsha, China
| | - Juan Cai
- Department of Nephrology, Hunan Key Laboratory of Kidney Disease and Blood Purification, The Second Xiangya Hospital at Central South University, Changsha, China
| | - Xiao-Ming Yin
- Department of Pathology and Laboratory Medicine, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Joel M. Weinberg
- Department of Medicine, University of Michigan Medical Center, Ann Arbor, MI, USA
| | - Manjeri A. Venkatachalam
- Department of Pathology, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Zheng Dong
- Department of Nephrology, Hunan Key Laboratory of Kidney Disease and Blood Purification, The Second Xiangya Hospital at Central South University, Changsha, China.,Department of Cellular Biology and Anatomy, Medical College of Georgia at Augusta University and Charlie Norwood VA Medical Center, Augusta, GA, USA.,
| |
Collapse
|
21
|
Jung HY, Oh SH, Ahn JS, Oh EJ, Kim YJ, Kim CD, Park SH, Kim YL, Cho JH. NOX1 Inhibition Attenuates Kidney Ischemia-Reperfusion Injury via Inhibition of ROS-Mediated ERK Signaling. Int J Mol Sci 2020; 21:ijms21186911. [PMID: 32967113 PMCID: PMC7554761 DOI: 10.3390/ijms21186911] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2020] [Revised: 09/13/2020] [Accepted: 09/14/2020] [Indexed: 02/07/2023] Open
Abstract
The protective effects of nicotinamide adenine dinucleotide phosphate (NADPH) oxidase (NOX) 1 inhibition against kidney ischemia-reperfusion injury (IRI) remain uncertain. The bilateral kidney pedicles of C57BL/6 mice were clamped for 30 min to induce IRI. Madin–Darby Canine Kidney (MDCK) cells were incubated with H2O2 (1.4 mM) for 1 h to induce oxidative stress. ML171, a selective NOX1 inhibitor, and siRNA against NOX1 were treated to inhibit NOX1. NOX expression, oxidative stress, apoptosis assay, and mitogen-activated protein kinase (MAPK) pathway were evaluated. The kidney function deteriorated and the production of reactive oxygen species (ROS), including intracellular H2O2 production, increased due to IRI, whereas IRI-mediated kidney dysfunction and ROS generation were significantly attenuated by ML171. H2O2 evoked the changes in oxidative stress enzymes such as SOD2 and GPX in MDCK cells, which was mitigated by ML171. Treatment with ML171 and transfection with siRNA against NOX1 decreased the upregulation of NOX1 and NOX4 induced by H2O2 in MDCK cells. ML171 decreased caspase-3 activity, the Bcl-2/Bax ratio, and TUNEL-positive tubule cells in IRI mice and H2O2-treated MDCK cells. Among the MAPK pathways, ML171 affected ERK signaling by ERK phosphorylation in kidney tissues and tubular cells. NOX1-selective inhibition attenuated kidney IRI via inhibition of ROS-mediated ERK signaling.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Jang-Hee Cho
- Correspondence: ; Tel.: +82-10-6566-7551; Fax: +82-53-426-2046
| |
Collapse
|
22
|
Desoky EAE, Sakr AM, Alhefnawy M, Omran M, Abdalla MMH, Shahin AS, Ali MM. Renal protective effect of N-acetylcysteine with stepwise ramping voltage against extracorporeal shock wave lithotripsy-induced renal injury: a prospective randomized trial. Int Urol Nephrol 2020; 52:2261-2267. [PMID: 32729099 DOI: 10.1007/s11255-020-02580-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2020] [Accepted: 07/20/2020] [Indexed: 11/29/2022]
Abstract
PURPOSE To evaluate the role of combination of N-acetylcysteine with stepwise ramping voltage in renal protection against the ischemic, vascular and oxidative effects of extracorporeal shock wave lithotripsy. PATIENTS AND METHODS A prospective randomized trial on 164 adult patients scheduled for ESWL for single renal stones. Patients with radio-lucent stones, diabetes, hypertension, febrile UTI, and preoperative albuminuria were excluded from the study. Patients were randomized into one of four groups. Group A patients received maximal fixed voltage of ESWL. Group B patients received stepwise ramping voltage of ESWL. Group C patients received fixed maximal voltage with N-acetylcysteine (NAC) 600 mg/bid from 48 h before to 24 h after the procedure. Group D patients received gradual ramping voltage with NAC. Urinary β2-microglobulin, 24 h albumin and N-acetyl-β-D-glucosaminidase/creatinine ratio at 1 day and 5 days post-ESWL and the stone free rate at 2 weeks were measured. RESULTS Group D was the only group that showed no significant difference pre and post ESWL in urinary albumin, β2-microglobulin and N-acetyl-β-D-glucosaminidase/creatinine ratio. Post hoc analysis revealed no significant difference between group B and group C in albumin, β2-microglobulin N-acetyl-β-D-glucosaminidase/creatinine ratio, but both of them had significantly lower levels than group A and significantly higher levels than group D. There was no statistically significant difference between all groups in the stone free rate at 2 weeks. CONCLUSION N-acetylcysteine protects the kidney against ESWL-induced renal injuries especially if combined with stepwise ramping voltage.
Collapse
Affiliation(s)
- Esam A E Desoky
- Urology Department, Faculty of Medicine, Zagazig University, Zagazig, Egypt
| | - Ahmed M Sakr
- Urology Department, Faculty of Medicine, Zagazig University, Zagazig, Egypt
| | - Mohamed Alhefnawy
- Urology Department, Faculty of Medicine, Benha University, Benha, Egypt
| | - Mohamed Omran
- Urology Department, Faculty of Medicine, Zagazig University, Zagazig, Egypt
| | | | - Ashraf S Shahin
- Urology Department, Faculty of Medicine, Zagazig University, Zagazig, Egypt
| | - Maged M Ali
- Urology Department, Faculty of Medicine, Zagazig University, Zagazig, Egypt.
| |
Collapse
|
23
|
Rajaram RD, Dissard R, Jaquet V, de Seigneux S. Potential benefits and harms of NADPH oxidase type 4 in the kidneys and cardiovascular system. Nephrol Dial Transplant 2020; 34:567-576. [PMID: 29931336 DOI: 10.1093/ndt/gfy161] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2018] [Indexed: 12/21/2022] Open
Abstract
The main function of NADPH oxidases is to catalyse the formation of reactive oxygen species (ROS). NADPH oxidase 4 (NOX4) is expressed at high levels in kidney tubular cells, and at lower levels in endothelial cells, cardiomyocytes and other cell types under physiological conditions. NOX4 is constitutively active producing hydrogen peroxide (H2O2) as the prevalent ROS detected, whereas other NOX isoforms present in the renal and cardiovascular systems (i.e. NOX1, NOX2 and NOX5) generate superoxide radical anions as main products. Pharmacological inhibition of NOX4 has received enormous attention for its potential therapeutic benefit in fibrotic disease and nephropathologies. Ongoing clinical trials are testing this approach in humans. Diabetes elevates NOX4 expression in podocytes and mesangial cells, which was shown to damage glomeruli leading to podocyte loss, mesangial cell hypertrophy and matrix accumulation. Consequently, NOX4 represents an interesting therapeutic target in diabetic nephropathy. On the contrary, experiments using NOX4-deficient mice have shown that NOX4 is cytoprotective in tubular cells, cardiomyocytes, endothelial cells and vascular smooth muscle cells, and has a metabolism-regulating role when these cells are subjected to injury. Mice with systemic NOX4 deletion are more susceptible to acute and chronic tubular injury, heart failure and atherosclerosis. Overall, the current literature suggests a detrimental role of increased NOX4 expression in mesangial cells and podocytes during diabetic nephropathy, but a cytoprotective role of this enzyme in other cellular types where it is expressed endogenously. We review here the recent evidence on the role of NOX4 in the kidneys and cardiovascular system. With the emergence of pharmacological NOX4 inhibitors in clinical trials, caution should be taken in identifying potential side effects in patients prone to acute kidney injury and cardiovascular disease.
Collapse
Affiliation(s)
- Renuga D Rajaram
- Laboratory of Nephrology, Service of Nephrology, Departments of Internal Medicine Specialties and Cell Physiology and Metabolism, University of Geneva, Geneva, Switzerland
| | - Romain Dissard
- Laboratory of Nephrology, Service of Nephrology, Departments of Internal Medicine Specialties and Cell Physiology and Metabolism, University of Geneva, Geneva, Switzerland
| | - Vincent Jaquet
- Department of Pathology and Immunology, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Sophie de Seigneux
- Laboratory of Nephrology, Service of Nephrology, Departments of Internal Medicine Specialties and Cell Physiology and Metabolism, University of Geneva, Geneva, Switzerland
| |
Collapse
|
24
|
Comparative study on protective effect of hydrogen rich saline and adipose-derived stem cells on hepatic ischemia-reperfusion and hepatectomy injury in swine. Biomed Pharmacother 2019; 120:109453. [PMID: 31561069 DOI: 10.1016/j.biopha.2019.109453] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2019] [Revised: 09/08/2019] [Accepted: 09/09/2019] [Indexed: 01/25/2023] Open
Abstract
AIM To compare and evaluate the hepatoprotective effect of liver parenchyma injection of ADSCs and portal vein injection of HRS in laparoscopic hepatic ischemia reperfusion combined with hepatectomy injury in miniature pigs. METHODS Eighteen miniature pigs were randomly assigned to IRI group, HRS group and ADSCs group. HRS was injected through the portal vein 10 min before reperfusion, 1 d, 2 d, and 3 d after surgery. ADSCs were injected into liver parenchyma after hepatectomy. The serum and liver tissue samples were collected at different time points (preoperative, and postoperative at 1 d, 3 d and 7 d). RESULTS Compared with the IRI group, both ADSCs and HRS groups can promote liver function recovery, reduce oxidative stress, reduce inflammation, and promote liver regeneration. Compared with HRS, ALT and TBIL in ADSCs group were significantly decreased at 3 d, and AST was significantly reduced at 1 d. The activities of SOD and GSH-Px in ADSCs group were significantly higher than that in HRS group, but the MDA level in HRS group was markedly lower than that in ADSCs group at 1 d. IL-1β was significantly lower in the ADSCs group than in the HRS group at 1 day after operation. The expressions of HGF and PCNA were significantly higher than that in the HRS group at 3 day after surgery. CONCLUSION Our study has demonstrated that HRS and ADSCs have significant hepatoprotective effects in miniature pigs after HIRI and hepatectomy injury. However, liver parenchyma injection of ADSCs is more beneficial to the recovery of liver function than portal vein injection of HRS.
Collapse
|
25
|
Miyazaki T, Gharib SA, Hsu YWA, Xu K, Khodakivskyi P, Kobayashi A, Paragas J, Klose AD, Francis KP, Dubikovskaya E, Page-McCaw PS, Barasch J, Paragas N. Cell-specific image-guided transcriptomics identifies complex injuries caused by ischemic acute kidney injury in mice. Commun Biol 2019; 2:326. [PMID: 31508501 PMCID: PMC6718519 DOI: 10.1038/s42003-019-0571-7] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2018] [Accepted: 08/09/2019] [Indexed: 01/20/2023] Open
Abstract
The kidney's inherent complexity has made identifying cell-specific pathways challenging, particularly when temporally associating them with the dynamic pathophysiology of acute kidney injury (AKI). Here, we combine renal cell-specific luciferase reporter mice using a chemoselective luciferin to guide the acquisition of cell-specific transcriptional changes in C57BL/6 background mice. Hydrogen peroxide generation, a common mechanism of tissue damage, was tracked using a peroxy-caged-luciferin to identify optimum time points for immunoprecipitation of labeled ribosomes for RNA-sequencing. Together, these tools revealed a profound impact of AKI on mitochondrial pathways in the collecting duct. In fact, targeting the mitochondria with an antioxidant, ameliorated not only hydrogen peroxide generation, but also significantly reduced oxidative stress and the expression of the AKI biomarker, LCN2. This integrative approach of coupling physiological imaging with transcriptomics and drug testing revealed how the collecting duct responds to AKI and opens new venues for cell-specific predictive monitoring and treatment.
Collapse
Affiliation(s)
- Tomoaki Miyazaki
- 1Division of Nephrology, Department of Medicine, University of Washington, Seattle, WA 98195 USA
- 2Division of Nephrology, Department of Medicine, Showa University, Yokohama, Japan
| | - Sina A Gharib
- 3Computational Medicine Core, Center for Lung Biology, University of Washington, Seattle, WA 98195 USA
| | - Yun-Wei A Hsu
- 1Division of Nephrology, Department of Medicine, University of Washington, Seattle, WA 98195 USA
| | - Katherine Xu
- 4Renal Division, Department of Medicine, Columbia University, New York, NY 10027 USA
| | - Pavlo Khodakivskyi
- 5Institute of Chemical Sciences and Engineering, Swiss Federal Institute of Technology of Lausanne (EPFL), Lausanne, Switzerland
| | - Akio Kobayashi
- 1Division of Nephrology, Department of Medicine, University of Washington, Seattle, WA 98195 USA
| | | | | | | | - Elena Dubikovskaya
- 5Institute of Chemical Sciences and Engineering, Swiss Federal Institute of Technology of Lausanne (EPFL), Lausanne, Switzerland
| | - Patrick S Page-McCaw
- 9Division of Nephrology, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN 37232 USA
| | - Jonathan Barasch
- 4Renal Division, Department of Medicine, Columbia University, New York, NY 10027 USA
| | - Neal Paragas
- 1Division of Nephrology, Department of Medicine, University of Washington, Seattle, WA 98195 USA
| |
Collapse
|
26
|
Moon D, Kim J. Cyclosporin A aggravates hydrogen peroxide-induced cell death in kidney proximal tubule epithelial cells. Anat Cell Biol 2019; 52:312-323. [PMID: 31598361 PMCID: PMC6773893 DOI: 10.5115/acb.18.192] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2018] [Revised: 03/07/2019] [Accepted: 03/12/2019] [Indexed: 12/22/2022] Open
Abstract
Cyclosporin A (CsA) does not only exert a toxic effect on kidney parenchymal cells, but also protects them against necrotic cell death by inhibiting opening of mitochondrial permeability transition pore. However, whether CsA plays a role in hydrogen peroxide-induced kidney proximal tubular cell death is currently unclear. In the present study, treatment with CsA further increased apoptosis and necrosis in HK-2 human kidney proximal tubule epithelial cells during exposure to hydrogen peroxide. In addition, hydrogen peroxide-induced p53 activation and BH3 interacting-domain death agonist (BID) expression were higher in CsA-treated cells than those in non-treated cells, whereas hydrogen peroxide-induced activation of mitogen-activated protein kinases including p38, c-Jun N-terminal kinase, and extracellular signal-regulated kinase and activation of protein kinase B were not significantly altered by treatment with CsA. In oxidant-antioxidant system, reactive oxygen species (ROS) production induced by hydrogen peroxide was further enhanced by treatment with CsA. However, expression levels of antioxidant enzymes including manganese superoxide dismutase, copper/zinc superoxide dismutase, and catalase were not altered by treatment with hydrogen peroxide or CsA. Treatment with CsA further enhanced mitochondrial membrane potential induced by exposure to hydrogen peroxide, although it did not alter endoplasmic reticulum stress based on expression of glucose-regulated protein 78 and 94. Taken together, these data suggest that CsA can aggravate hydrogen peroxide-induced cell death through p53 activation, BID expression, and ROS production.
Collapse
Affiliation(s)
- Daeun Moon
- Interdisciplinary Graduate Program in Advanced Convergence Technology & Science, Jeju National University, Jeju, Korea
| | - Jinu Kim
- Interdisciplinary Graduate Program in Advanced Convergence Technology & Science, Jeju National University, Jeju, Korea.,Department of Anatomy, Jeju National University School of Medicine, Jeju, Korea
| |
Collapse
|
27
|
The potential protective effects of erythropoietin and estrogen on renal ischemia reperfusion injury in ovariectomized rats. ALEXANDRIA JOURNAL OF MEDICINE 2019. [DOI: 10.1016/j.ajme.2015.12.001] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
|
28
|
Zhang X, Wang Y, Shen W, Ma S, Chen W, Qi R. Rosa rugosa flavonoids alleviate myocardial ischemia reperfusion injury in mice by suppressing JNK and p38 MAPK. Microcirculation 2018; 24. [PMID: 28597598 DOI: 10.1111/micc.12385] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2017] [Accepted: 06/01/2017] [Indexed: 12/13/2022]
Abstract
OBJECTIVE Although Rosa rugosa has been applied for preventing coronary artery disease, the pharmacological mechanism is little explored. In this study, the effects and mechanisms of Rosa rugosa flavonoids (RRF) on myocardial ischemia reperfusion injury (MIRI) were investigated. METHODS Mice were pretreated by intragastric administration of 600 mg/kg RRF for 7 days. Then MIRI was induced by 45 minutes coronary artery ligation and 3 hours reperfusion. Myocardial infarct size (MIS) and histopathology, activities of myocardial enzymes, and effects of RRF on inflammation and apoptosis were evaluated. RESULTS Pretreating the mice with RRF significantly reduced MIS and inhibited activity of plasma myocardial enzymes. Activity of the enzymes associated with anti-oxidation, SOD, and TEAC, and mRNA expression of NOX2 were significantly elevated. RRF pretreatment significantly decreased the translocation of p65 from the cytoplasm into the nucleus and reduced the expression of the pro-inflammatory cytokines, IL-6 and IL-1β. RRF pretreatment also significantly prevented the expression of caspase-3 and Bax, and increased the expression of Bcl-2. And RRF inhibited the phosphorylation of JNK and p38 MAPK. CONCLUSIONS RRF significantly inhibited MIRI through anti-oxidative, anti-inflammatory, and anti-apoptosis effects, and mechanisms were associated with its inhibition on phosphorylation of JNK and p38 MAPK.
Collapse
Affiliation(s)
- Xuehui Zhang
- Shihezi University College of Pharmacy/Key Laboratory of Xinjiang Endemic Phytomedicine Resources Ministry of Education, Xinjiang, China.,Peking University Institute of Cardiovascular Sciences, Peking University Health Science Center, Peking University, Beijing, China.,Key Laboratory of Molecular Cardiovascular Sciences, Ministry of Education, Beijing, China
| | - Yuhui Wang
- Peking University Institute of Cardiovascular Sciences, Peking University Health Science Center, Peking University, Beijing, China.,Key Laboratory of Molecular Cardiovascular Sciences, Ministry of Education, Beijing, China
| | - Wanli Shen
- Shihezi University College of Pharmacy/Key Laboratory of Xinjiang Endemic Phytomedicine Resources Ministry of Education, Xinjiang, China.,Peking University Institute of Cardiovascular Sciences, Peking University Health Science Center, Peking University, Beijing, China.,Key Laboratory of Molecular Cardiovascular Sciences, Ministry of Education, Beijing, China
| | - Shangzhi Ma
- Shihezi University College of Pharmacy/Key Laboratory of Xinjiang Endemic Phytomedicine Resources Ministry of Education, Xinjiang, China.,Peking University Institute of Cardiovascular Sciences, Peking University Health Science Center, Peking University, Beijing, China.,Key Laboratory of Molecular Cardiovascular Sciences, Ministry of Education, Beijing, China
| | - Wen Chen
- Shihezi University College of Pharmacy/Key Laboratory of Xinjiang Endemic Phytomedicine Resources Ministry of Education, Xinjiang, China
| | - Rong Qi
- Shihezi University College of Pharmacy/Key Laboratory of Xinjiang Endemic Phytomedicine Resources Ministry of Education, Xinjiang, China.,Peking University Institute of Cardiovascular Sciences, Peking University Health Science Center, Peking University, Beijing, China.,Key Laboratory of Molecular Cardiovascular Sciences, Ministry of Education, Beijing, China
| |
Collapse
|
29
|
Cai Z, Shi T, Zhuang R, Fang H, Jiang X, Shao Y, Zhou H. Protective effect of N-acetylcysteine activated carbon release microcapsule on myocardial ischemia-reperfusion injury in rats. Exp Ther Med 2017; 15:1809-1818. [PMID: 29434769 PMCID: PMC5776512 DOI: 10.3892/etm.2017.5653] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2016] [Accepted: 05/05/2017] [Indexed: 12/21/2022] Open
Abstract
With the development of science and technology, and development of artery bypass, methods such as cardiopulmonary cerebral resuscitation have been practiced in recent years. Despite this, some methods fail to promote or recover the function of tissues and organs, and in some cases, may aggravate dysfunction and structural damage to tissues. The latter is typical of ischemia-reperfusion (IR) injury. Lipid peroxidation mediated by free radicals is an important process of myocardial IR injury. Myocardial IR has been demonstrated to induce the formation of large numbers of free radicals in rats, which promotes the peroxidation of lipids within unsaturated fatty acids in the myocardial cell membrane. Markers of lipid peroxidation include malondialdehyde, superoxide dismutase and lactic dehydrogenase. Recent studies have demonstrated that N-acetylcysteine (NAC) is able to dilate blood vessels, prevent oxidative damage, improve immunity, inhibit apoptosis and the inflammatory response and promote glutathione synthesis in cells. NAC also improves the systolic function of myocardial cells and cardiac function, prevents myocardial apoptosis, protects ventricular remodeling and vascular remodeling, reduces opiomelanocortin levels in the serum and increases the content of nitric oxide in the serum, thus improving vascular endothelial function. Therefore, NAC has potent pharmacological activity; however, the relatively fast metabolism of NAC, along with its large clinical dose and low bioavailability, limit its applications. The present study combined NAC with medicinal activated carbons, and prepared N-acetylcysteine activated carbon sustained-release microcapsules (ACNACs) to overcome the limitations of NAC. It was demonstrated that ACNACs exerted greater effective protective effects than NAC alone on myocardial IR injury in rats.
Collapse
Affiliation(s)
- Zhaobin Cai
- Department of Cardiology, The Xixi Hospital of Hangzhou Affiliated to Zhejiang University of Traditional Chinese Medicine, Hangzhou, Zhejiang 310023, P.R. China
| | - Tingting Shi
- Department of Pharmaceutical Preparation, The Xixi Hospital of Hangzhou Affiliated to Zhejiang University of Traditional Chinese Medicine, Hangzhou, Zhejiang 310023, P.R. China
| | - Rangxiao Zhuang
- Department of Pharmaceutical Preparation, The Xixi Hospital of Hangzhou Affiliated to Zhejiang University of Traditional Chinese Medicine, Hangzhou, Zhejiang 310023, P.R. China
| | - Hongying Fang
- Department of Pharmaceutical Preparation, The Xixi Hospital of Hangzhou Affiliated to Zhejiang University of Traditional Chinese Medicine, Hangzhou, Zhejiang 310023, P.R. China
| | - Xiaojie Jiang
- Department of Pharmaceutical Preparation, The Xixi Hospital of Hangzhou Affiliated to Zhejiang University of Traditional Chinese Medicine, Hangzhou, Zhejiang 310023, P.R. China
| | - Yidan Shao
- Department of Pharmaceutical Preparation, The Xixi Hospital of Hangzhou Affiliated to Zhejiang University of Traditional Chinese Medicine, Hangzhou, Zhejiang 310023, P.R. China
| | - Hongping Zhou
- Department of Pharmacy, Hangzhou Children's Hospital, Hangzhou, Zhejiang 310014, P.R. China
| |
Collapse
|
30
|
Sánchez-Ramos C, Prieto I, Tierrez A, Laso J, Valdecantos MP, Bartrons R, Roselló-Catafau J, Monsalve M. PGC-1α Downregulation in Steatotic Liver Enhances Ischemia-Reperfusion Injury and Impairs Ischemic Preconditioning. Antioxid Redox Signal 2017; 27:1332-1346. [PMID: 28269997 DOI: 10.1089/ars.2016.6836] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
AIMS Liver steatosis is associated with mitochondrial dysfunction and elevated reactive oxygen species (ROS) levels together with enhanced sensitivity to ischemia-reperfusion (IR) injury and limited response to preconditioning protocols. Here, we sought to determine whether the downregulation in the steatotic liver of peroxisome proliferator-activated receptor γ co-activator 1α (PGC-1α), a master regulator of mitochondrial metabolism and ROS that is known to play a role in liver metabolic control, could be responsible for the sensitivity of the steatotic liver to ischemic damage. RESULTS PGC-1α was induced in normal liver after exposure to an IR protocol, which was concomitant with an increase in the levels of antioxidant proteins. By contrast, its induction was severely blunted in the steatotic liver, resulting in a modest induction of antioxidant proteins. Livers of PGC-1α-/- mice on a chow diet were normal, but they exhibited an enhanced sensitivity to IR injury and also a lack of response to ischemic preconditioning (IPC), a phenotype that recapitulated the features of the steatotic liver in terms of liver damage, although the inflammatory response differed between both models. Utilizing an in vitro model of IPC, we found that PGC-1α expression was downregulated in hepatic cells cultured at 1% O2; whereas it was induced after reoxygenation (3% O2), and it was responsible for the recovery of antioxidant gene expression after the ischemic period. Innovation & Conclusion: PGC-1α plays an important role in the protection against IR injury in the liver, which is likely associated with its capacity to induce antioxidant gene expression. Antioxid. Redox Signal. 27, 1332-1346.
Collapse
Affiliation(s)
| | - Ignacio Prieto
- 1 Instituto de Investigaciones Biomédicas "Alberto Sols" (CSIC-UAM) , Madrid, Spain
| | - Alberto Tierrez
- 2 Fundación Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC) , Madrid, Spain
| | - Javier Laso
- 2 Fundación Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC) , Madrid, Spain
| | - M Pilar Valdecantos
- 3 Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERdem) , ISCIII, Madrid, Spain
| | - Ramon Bartrons
- 4 Unitat de Bioquímica i Biologia Molecular, Departament de Ciències Fisiològiques, Campus de Bellvitge, IDIBELL-Universitat de Barcelona , Hospitalet, Spain
| | - Joan Roselló-Catafau
- 5 Experimental Hepatic Ischemia-Reperfusion Unit, Institut d'Investigacions Biomèdiques de Barcelona (CSIC) , Barcelona, Spain
| | - María Monsalve
- 1 Instituto de Investigaciones Biomédicas "Alberto Sols" (CSIC-UAM) , Madrid, Spain
| |
Collapse
|
31
|
Chen J, Ren J, Loo WTY, Hao L, Wang M. Lysyl oxidases expression and histopathological changes of the diabetic rat nephron. Mol Med Rep 2017; 17:2431-2441. [PMID: 29207131 PMCID: PMC5783488 DOI: 10.3892/mmr.2017.8182] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2017] [Accepted: 11/06/2017] [Indexed: 02/05/2023] Open
Abstract
Diabetic nephropathy (DN) is a major complication of diabetes, the accumulation of extracellular matrix (ECM) is considered an indication of nephropathological changes. Lysyl oxidases (LOXs) are also associated with ECM. However, the majority of studies on LOXs have focused on their potential role in renal fibrogenesis and there has no examination of LOXs expression or the correlation with histopathological changes of DN, including glomerular basement membrane (GBM) thickening and glomerulosclerosis. In this study, the association between histological changes and LOXs was explored using a type 2 diabetes model of male Zucker diabetic fatty rats. The expression of LOX and lysyl oxidase-like 1 to 3 (LOXL1 to 3) levels were evaluated by immunohistochemical staining. The expression levels of LOX and LOXL2 in the kidney tissue in the diabetic group were significantly higher compared with those of the control group, but LOXL1 and LOXL3 expression levels were not significantly different between the two groups. These results indicated that LOXL2 and LOX may be critical factors involved in the progression of DN.
Collapse
Affiliation(s)
- Jun Chen
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Jie Ren
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Wings T Y Loo
- Department of Rehabilitation Sciences, The Hong Kong Polytechnic University, Kowloon, Hong Kong 999077, P.R. China
| | - Liang Hao
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Min Wang
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| |
Collapse
|
32
|
Tong F, Zhou X. The Nrf2/HO-1 Pathway Mediates the Antagonist Effect of L-Arginine On Renal Ischemia/Reperfusion Injury in Rats. Kidney Blood Press Res 2017; 42:519-529. [PMID: 28854440 DOI: 10.1159/000480362] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2017] [Accepted: 06/16/2017] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND/AIMS Ischemia/reperfusion (I/R) is the most common cause of acute renal injury. I/R-induced oxidative stress is involved in the development of acute renal injury, which can be reversed by supplementation with L-arginine, a precursor of nitric oxide (NO). This study was conducted to evaluate alterations in the expression of transcription factors [nuclear factor kappa B (NF-κB), nuclear factor-E2-related factor-2 (Nrf2), and heme oxygenase 1 (HO-1)] and heat shock protein 70 (HSP70) in the kidney of I/R-induced injury rats. METHODS Sprague-Dawley (SD) rats were subjected to bilateral renal ischemia for 45 min followed by reperfusion for 24 h. Group 1, Sham; group 2, I/R; group 3, L-arginine; and group 4, L-arginine+zinc protoporphyrin (ZnPP). The levels of serum creatinine (Scr), blood urea nitrogen (BUN), serum nitric oxide (NO), histic malondialdehyde (MDA) and reactive oxygen species (ROS) and superoxide dismutase (SOD) activity were determined, and the expression levels of Nrf2, HO-1, NF-κB, and HSP70 were evaluated. RESULTS The treatment of rats with L-arginine produced a significant reduction in the levels of BUN, Scr, MDA and a significant enhancement in the level of NO and in the activity of SOD compared to renal I/R groups. The expression levels of Nrf2, HO-1, and HSP70 were strongly increased, and the expression of NF-κB and production of ROS were significantly decreased in the L-arginine group compared to that of the I/R group. ZnPP increased renal damage and displayed effects opposite to those of L-arginine. CONCLUSION These findings suggested that L-arginine/NO reduces renal dysfunction associated with I/R of the kidney and may act as a trigger to regulate the NF-κB, HSP70 and Nrf2/HO-1 signaling cascades.
Collapse
|
33
|
Dai H, Wang M, Patel PN, Kalogeris T, Liu Y, Durante W, Korthuis RJ. Preconditioning with the BK Ca channel activator NS-1619 prevents ischemia-reperfusion-induced inflammation and mucosal barrier dysfunction: roles for ROS and heme oxygenase-1. Am J Physiol Heart Circ Physiol 2017; 313:H988-H999. [PMID: 28822969 DOI: 10.1152/ajpheart.00620.2016] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/13/2016] [Revised: 07/25/2017] [Accepted: 08/07/2017] [Indexed: 12/12/2022]
Abstract
Activation of large-conductance Ca2+-activated K+ (BKCa) channels evokes cell survival programs that mitigate intestinal ischemia and reperfusion (I/R) inflammation and injury 24 h later. The goal of the present study was to determine the roles of reactive oxygen species (ROS) and heme oxygenase (HO)-1 in delayed acquisition of tolerance to I/R induced by pretreatment with the BKCa channel opener NS-1619. Superior mesentery arteries were occluded for 45 min followed by reperfusion for 70 min in wild-type (WT) or HO-1-null (HO-1-/-) mice that were pretreated with NS-1619 or saline vehicle 24 h earlier. Intravital microscopy was used to quantify the numbers of rolling and adherent leukocytes. Mucosal permeability, tumor necrosis factor-α (TNF-α) levels, and HO-1 activity and expression in jejunum were also determined. I/R induced leukocyte rolling and adhesion, increased intestinal TNF-α levels, and enhanced mucosal permeability in WT mice, effects that were largely abolished by pretreatment with NS-1619. The anti-inflammatory and mucosal permeability-sparing effects of NS-1619 were prevented by coincident treatment with the HO-1 inhibitor tin protoporphyrin-IX or a cell-permeant SOD mimetic, Mn(III)tetrakis (4-benzoic acid) porphyrin (MnTBAP), in WT mice. NS-1619 also increased jejunal HO-1 activity in WT animals, an effect that was attenuated by treatment with the BKCa channel antagonist paxilline or MnTBAP. I/R also increased postischemic leukocyte rolling and adhesion and intestinal TNF-α levels in HO-1-/- mice to levels comparable to those noted in WT animals. However, NS-1619 was ineffective in preventing these effects in HO-1-deficient mice. In summary, our data indicate that NS-1619 induces the development of an anti-inflammatory phenotype and mitigates postischemic mucosal barrier disruption in the small intestine by a mechanism that may involve ROS-dependent HO-1 activity.NEW & NOTEWORTHY Antecedent treatment with the large-conductance Ca2+-activated K+ channel opener NS-1619 24 h before ischemia-reperfusion limits postischemic tissue injury by an oxidant-dependent mechanism. The present study shows that NS-1619-induced oxidant production prevents ischemia-reperfusion-induced inflammation and mucosal barrier disruption in the small intestine by provoking increases in heme oxygenase-1 activity.
Collapse
Affiliation(s)
- Hongyan Dai
- Department of Medical Pharmacology and Physiology and Dalton Cardiovascular Research Center, University of Missouri School of Medicine, Columbia, Missouri
| | - Meifang Wang
- Department of Medical Pharmacology and Physiology and Dalton Cardiovascular Research Center, University of Missouri School of Medicine, Columbia, Missouri
| | - Parag N Patel
- Department of Medical Pharmacology and Physiology and Dalton Cardiovascular Research Center, University of Missouri School of Medicine, Columbia, Missouri
| | - Theodore Kalogeris
- Department of Medical Pharmacology and Physiology and Dalton Cardiovascular Research Center, University of Missouri School of Medicine, Columbia, Missouri
| | - Yajun Liu
- Department of Medical Pharmacology and Physiology and Dalton Cardiovascular Research Center, University of Missouri School of Medicine, Columbia, Missouri
| | - William Durante
- Department of Medical Pharmacology and Physiology and Dalton Cardiovascular Research Center, University of Missouri School of Medicine, Columbia, Missouri
| | - Ronald J Korthuis
- Department of Medical Pharmacology and Physiology and Dalton Cardiovascular Research Center, University of Missouri School of Medicine, Columbia, Missouri
| |
Collapse
|
34
|
Sekijima M, Sahara H, Miki K, Villani V, Ariyoshi Y, Iwanaga T, Tomita Y, Yamada K. Hydrogen sulfide prevents renal ischemia-reperfusion injury in CLAWN miniature swine. J Surg Res 2017; 219:165-172. [PMID: 29078877 DOI: 10.1016/j.jss.2017.05.123] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2017] [Revised: 05/17/2017] [Accepted: 05/25/2017] [Indexed: 11/17/2022]
Abstract
BACKGROUND Hydrogen sulfide (H2S) has recently been reported to demonstrate both antiinflammatory and cytoprotective effects; however, its efficacy has not been well documented in large animal models. In this study, we examined whether the administration of H2S offers cytoprotective effects on renal ischemia-reperfusion injury (IRI) in a preclinical miniature swine model. METHODS Major histocompatibility complex-inbred, CLAWN miniature swine (n = 9) underwent a right nephrectomy, followed by induction of a 120-min period of warm ischemia via placement of clamps on the left renal artery and vein. Group 1 (n = 3) underwent renal ischemia without H2S administration. Groups 2 (n = 3) and 3 (n = 3) received Na2S (prodrug of H2S) 10 min before reperfusion of the ischemic kidneys followed by a 30-min of Na2S postreperfusion intravenously (group 2) or selective administration of Na2S via the left renal artery (group 3). IRI was assessed by kidney biopsies, levels of inflammatory cytokines in sera and kidney tissue. RESULTS Animals in group 1 had significantly higher serum creatinine levels compared with animals in groups 2 and 3 (P < 0.01). Histology showed severe tubular damage with TUNEL-positive cells in group 1 on postoperative day 2 compared with mild damage in group 2 and minimal damage in group 3. Furthermore, levels of inflammatory cytokines in both serum (interleukin-6 [IL-6], tumor necrosis factor-α, and high-mobility group box 1) and renal tissue (IL-1 and IL-6) in group 3 were markedly lower than in group 2, suggesting beneficial effects of selective Na2S administration. CONCLUSIONS Na2S administration, especially via an organ selective approach, appears to potentially offer cytoprotective and antiinflammatory effects following renal IRI.
Collapse
Affiliation(s)
- Mitsuhiro Sekijima
- Division of Organ Replacement and Xenotransplantation Surgery, Center for Advanced Biomedical Science and Swine Research, Kagoshima University, Kagoshima, Japan
| | - Hisashi Sahara
- Division of Organ Replacement and Xenotransplantation Surgery, Center for Advanced Biomedical Science and Swine Research, Kagoshima University, Kagoshima, Japan
| | - Katsuyuki Miki
- Division of Organ Replacement and Xenotransplantation Surgery, Center for Advanced Biomedical Science and Swine Research, Kagoshima University, Kagoshima, Japan; The 3rd Department of the Surgery, Kidney Center, Tokyo Women's Medical University, Tokyo, Japan
| | - Vincenzo Villani
- Transplantation Biology Research Center Laboratory, Center for Transplantation Sciences, Massachusetts General Hospital, Boston, Massachusetts
| | - Yuichi Ariyoshi
- Division of Organ Replacement and Xenotransplantation Surgery, Center for Advanced Biomedical Science and Swine Research, Kagoshima University, Kagoshima, Japan
| | - Takehiro Iwanaga
- Division of Organ Replacement and Xenotransplantation Surgery, Center for Advanced Biomedical Science and Swine Research, Kagoshima University, Kagoshima, Japan
| | - Yusuke Tomita
- Division of Organ Replacement and Xenotransplantation Surgery, Center for Advanced Biomedical Science and Swine Research, Kagoshima University, Kagoshima, Japan
| | - Kazuhiko Yamada
- Division of Organ Replacement and Xenotransplantation Surgery, Center for Advanced Biomedical Science and Swine Research, Kagoshima University, Kagoshima, Japan.
| |
Collapse
|
35
|
Xu Y, Zhou H, Zhu Q. The Impact of Microbiota-Gut-Brain Axis on Diabetic Cognition Impairment. Front Aging Neurosci 2017; 9:106. [PMID: 28496408 PMCID: PMC5406474 DOI: 10.3389/fnagi.2017.00106] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2016] [Accepted: 03/31/2017] [Indexed: 12/11/2022] Open
Abstract
Progressive cognitive dysfunction is a central characteristic of diabetic encephalopathy (DE). With an aging population, the incidence of DE is rising and it has become a major threat that seriously affects public health. Studies within this decade have indicated the important role of risk factors such as oxidative stress and inflammation on the development of cognitive impairment. With the recognition of the two-way communication between gut and brain, recent investigation suggests that “microbiota-gut-brain axis” also plays a pivotal role in modulating both cognition function and endocrine stability. This review aims to systemically elucidate the underlying impact of diabetes on cognitive impairment.
Collapse
Affiliation(s)
- Youhua Xu
- Faculty of Chinese Medicine, Macau University of Science and TechnologyTaipa, Macau.,State Key Laboratory of Quality Research in Chinese Medicine (Macau University of Science and Technology)Taipa, Macau
| | - Hua Zhou
- Faculty of Chinese Medicine, Macau University of Science and TechnologyTaipa, Macau.,State Key Laboratory of Quality Research in Chinese Medicine (Macau University of Science and Technology)Taipa, Macau.,Laboratory for Bioassay and Molecular Pharmacology of Chinese Medicines, Macau Institute for Applied Research in Medicine and HealthTaipa, Macau
| | - Quan Zhu
- Faculty of Chinese Medicine, Macau University of Science and TechnologyTaipa, Macau.,State Key Laboratory of Quality Research in Chinese Medicine (Macau University of Science and Technology)Taipa, Macau.,Laboratory for Bioassay and Molecular Pharmacology of Chinese Medicines, Macau Institute for Applied Research in Medicine and HealthTaipa, Macau.,Guangdong Consun Pharmaceutical Group, Institute of Consun Co. for Chinese Medicine in Kidney DiseasesGuangzhou, China
| |
Collapse
|
36
|
NADPH oxidase 4 deficiency increases tubular cell death during acute ischemic reperfusion injury. Sci Rep 2016; 6:38598. [PMID: 27924932 PMCID: PMC5141508 DOI: 10.1038/srep38598] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2016] [Accepted: 11/10/2016] [Indexed: 12/13/2022] Open
Abstract
NADPH oxidase 4 (NOX4) is highly expressed in kidney proximal tubular cells. NOX4 constitutively produces hydrogen peroxide, which may regulate important pro-survival pathways. Renal ischemia reperfusion injury (IRI) is a classical model mimicking human ischemic acute tubular necrosis. We hypothesized that NOX4 plays a protective role in kidney IRI. In wild type (WT) animals subjected to IRI, NOX4 protein expression increased after 24 hours. NOX4 KO (knock-out) and WT littermates mice were subjected to IRI. NOX4 KO mice displayed decreased renal function and more severe tubular apoptosis, decreased Bcl-2 expression and higher histologic damage scores compared to WT. Activation of NRF2 was decreased in NOX4 KO mice in response to IRI. This was related to decreased KEAP1 oxidation leading to decreased NRF2 stabilization. This resulted in decreased glutathione levels. In vitro silencing of NOX4 in cells showed an enhanced propensity to apoptosis, with reduced expression of NRF2, glutathione content and Bcl-2 expression, similar to cells derived from NOX4 KO mice. Overexpression of a constitutively active form of NRF2 (caNRF2) in NOX4 depleted cells rescued most of this phenotype in cultured cells, implying that NRF2 regulation by ROS issued from NOX4 may play an important role in its anti-apoptotic property.
Collapse
|
37
|
Bayir Y, Cadirci E, Polat B, Kilic Baygutalp N, Albayrak A, Karakus E, Un H, Keles MS, Kocak Ozgeris FB, Toktay E, Karaca M, Halici Z. Aliskiren - a promising strategy for ovarian ischemia/reperfusion injury protection in rats via RAAS. Gynecol Endocrinol 2016; 32:675-683. [PMID: 26939623 DOI: 10.3109/09513590.2016.1153055] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
The aim of this study was to evaluate the effects of aliskiren, direct renin inhibitor, as an antioxidant and tissue protective agent and evaluate the molecular, biochemical, and histopathological changes in experimental ischemia and ischemia/reperfusion injury in rat ovaries. Forty-eight female rats were randomly divided into eight groups: in Group 1, only sham operation was performed. Group 2 received 100 mg/kg aliskiren and sham operated. In Group 3, 3 h-period of bilateral ovarian ischemia was applied. Group 4 received a 3-h period of ischemia followed by 3 h of reperfusion. Groups 5 and 6 received 50 and 100 mg/kg, respectively, of aliskiren and bilateral ovarian ischemia was applied (after a 3-h period of ischemia, both ovaries were surgically removed). To Groups 7 and 8, 50 and 100 mg/kg of aliskiren were administered, respectively, and the induction of ischemia was performed. At the end of a 3-h period of ischemia, bilateral vascular clips were removed, and 3 h of reperfusion continued. After the experiments, IL-1β, IL-6, TNF-α, and iNOS mRNA expressions and SOD, GSH, MDA, renin, and angiotensin-II levels were determined and histopathological changes were examined in rat ovaries. Aliskiren treatment normalized excessive changes in cytokine and oxidative stress markers in both ischemia and ischemia/reperfusion injury. Histopathologically, treatment with aliskiren ameliorated the development of ischemia and/or ischemia/reperfusion tissue injury. This study concluded that aliskiren treatment is effective in reversing ischemia and/or ischemia/reperfusion induced ovary damage via the improvement of oxidative stress, reduction of inflammation, and suppression of the renin-angiotensin aldosterone system.
Collapse
Affiliation(s)
- Yasin Bayir
- a Department of Biochemistry , Faculty of Pharmacy, Ataturk University , Erzurum , Turkey
| | - Elif Cadirci
- b Department of Pharmacology , Faculty of Medicine, Ataturk University , Erzurum , Turkey
| | - Beyzagul Polat
- c Department of Pharmacology , Faculty of Pharmacy, Ataturk University , Erzurum , Turkey
| | - Nurcan Kilic Baygutalp
- a Department of Biochemistry , Faculty of Pharmacy, Ataturk University , Erzurum , Turkey
| | - Abdulmecit Albayrak
- b Department of Pharmacology , Faculty of Medicine, Ataturk University , Erzurum , Turkey
| | - Emre Karakus
- d Department of Pharmacology , Faculty of Veterinary, Ataturk University , Erzurum , Turkey
| | - Harun Un
- e Department of Biochemistry , Faculty of Pharmacy, Agri Ibrahim CecenUniversity , Agri , Turkey
| | - Mevlut Sait Keles
- f Department of Medical Biochemistry , Faculty of Medicine, Ataturk University , Erzurum , Turkey
| | | | - Erdem Toktay
- g Department of Histology and Embryology , Faculty of Medicine, Kafkas University , Kars , Turkey , and
| | - Mehmet Karaca
- h Department of Obstetrics and Gynecology , Education and Research Hospital , Antalya , Turkey
| | - Zekai Halici
- b Department of Pharmacology , Faculty of Medicine, Ataturk University , Erzurum , Turkey
| |
Collapse
|
38
|
Azizi F, Seifi B, Kadkhodaee M, Ahghari P. Administration of hydrogen sulfide protects ischemia reperfusion-induced acute kidney injury by reducing the oxidative stress. Ir J Med Sci 2016; 185:649-654. [PMID: 26141462 DOI: 10.1007/s11845-015-1328-z] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2015] [Accepted: 06/20/2015] [Indexed: 01/03/2023]
Abstract
BACKGROUND Renal ischemia-reperfusion injury (IRI) is a major cause of acute kidney injury. Hydrogen sulfide (H2S) has been known as a novel gaseous signaling molecule. AIMS The aim of this study was to investigate whether the efficacy of H2S in protecting against renal IRI is through its antioxidative effect. METHOD In this study, rats were randomized into Sham, IR, or sodium hydrosulfide (NaHS, an H2S donor) groups. To establish a model of renal IRI, both renal arteries were occluded for 55 min and then declamped to allow reperfusion for 24 h. Rats in the NaHS group received intraperitoneal injections of 75 μmol/kg NaHS 10 min before the onset of ischemia and immediately after the onset of reperfusion. Sham group underwent laparotomy without cross-clamping of renal pedicles. After reperfusion, plasma and renal tissue samples were collected for functional, histological, and oxidative stress evaluation. RESULTS The IR group exhibited significant rise in plasma creatinine, blood urea nitrogen (BUN), renal malondialdehyde (MDA) concentration, and significant reduction of renal superoxide dismutase (SOD) activity. Treatment with NaHS reduced the levels of plasma creatinine, BUN, renal MDA concentration, and increased SOD activity in the kidneys. NaHS improved renal histological changes in comparison to IR group. CONCLUSION Our data demonstrated that H2S can protect against renal IRI and that its therapeutic effects may be mediated by reducing oxidative stress.
Collapse
Affiliation(s)
- F Azizi
- Department of Neurosciences and Addiction, School of Advanced in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - B Seifi
- Department of Physiology, Faculty of Medicine, School of Medicine, Tehran University of Medical Sciences, Poorsina Ave., Tehran, Iran.
| | - M Kadkhodaee
- Department of Physiology, Faculty of Medicine, School of Medicine, Tehran University of Medical Sciences, Poorsina Ave., Tehran, Iran
| | - P Ahghari
- Department of Physiology, School of Medicine, Hamedan University of Medical Sciences, Hamadan, Iran
| |
Collapse
|
39
|
Wen J, Shu Y, Zhang W. ROS, P53, and ischemic acute kidney injury in diabetic models. Kidney Int 2016; 88:198-9. [PMID: 26126099 DOI: 10.1038/ki.2015.130] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Affiliation(s)
- Jiagen Wen
- 1] Department of Clinical Pharmacology, Xiangya Hospital, Central South University, ChangSha, China [2] Institute of Clinical Pharmacology, Central South University, ChangSha, China [3] Hunan Key Laboratory of Pharmacogenetics, ChangSha, China
| | - Yan Shu
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Maryland, Baltimore, Maryland, USA
| | - Wei Zhang
- 1] Department of Clinical Pharmacology, Xiangya Hospital, Central South University, ChangSha, China [2] Institute of Clinical Pharmacology, Central South University, ChangSha, China [3] Hunan Key Laboratory of Pharmacogenetics, ChangSha, China
| |
Collapse
|
40
|
Li Z, Deng X, Kang Z, Wang Y, Xia T, Ding N, Yin Y. Elevation of miR-21, through targeting MKK3, may be involved in ischemia pretreatment protection from ischemia-reperfusion induced kidney injury. J Nephrol 2016; 29:27-36. [PMID: 26149640 DOI: 10.1007/s40620-015-0217-x] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2015] [Accepted: 06/19/2015] [Indexed: 12/24/2022]
Abstract
BACKGROUND Ischemia-reperfusion (IR) causes acute kidney injury (AKI), and ischemia pretreatment may exert protection. Mitogen-activated protein kinase kinase 3 (MKK3), which is involved in the signal transduction pathway in IR-induced injury, is a potential target of miR-21. We aimed to verify the targeting regulation of miR-21 on MKK3 and to explore the effects of miR-21-mediated MKK3 expression changes in AKI. METHODS Vectors containing the MKK3 3'UTR and mutated MKK3-3U-M were constructed and co-transfected with nonsense miR, miR-21-5p mimics or inhibitor in HEK293 cells. Gene expressions were detected by dual luciferase reporter assay. The effects of miR-21 on mRNA and protein of MKK3 were investigated in HK-2 cells. Male C57BL/6J mice were treated with ischemic preconditioning (IPC) and IR. Kidney functions were assessed through monitoring serum creatinine (Scr) and blood urea nitrogen (BUN). Pathological changes were observed and scored with histological samples of kidney. Expression levels of miR-21, MKK3, interleukin (IL)-6, tumor necrosis factor (TNF)-α before and after IPC and IR were examined by real-time polymerase chain reaction and/or immunohistochemistry. RESULTS miR-21 regulated the expression of MKK3 via 3'UTR. Following IR, MKK3, IL-6 and TNF-α levels were increased. Scr, BUN and pathological injuries were aggravated, and miR-21 expression was increased. IPC increased miR-21 levels ahead of IR and inhibited the increases in MKK3, IL-6 and TNF-α levels and the aggravation of Scr, BUN and pathological injuries. CONCLUSIONS miR-21 targets MKK3 in vivo and in vitro, inhibiting the downstream factors IL-6 and TNF-α. Therefore, miR-21 might be involved in protection of IPC against IR of the kidney.
Collapse
Affiliation(s)
- Zhihui Li
- Department of Nephrology of Hunan Children's Hospital, Hunan Institute for Pediatric Research, 86 Ziyuan Road, Changsha, 410007, Hunan, People's Republic of China.
- Academy of Pediatrics of University of South China, 86 Ziyuan Road, Changsha, 410007, Hunan, People's Republic of China.
| | - Xu Deng
- Department of Nephrology of Hunan Children's Hospital, Hunan Institute for Pediatric Research, 86 Ziyuan Road, Changsha, 410007, Hunan, People's Republic of China
- Academy of Pediatrics of University of South China, 86 Ziyuan Road, Changsha, 410007, Hunan, People's Republic of China
| | - Zhijuan Kang
- Department of Nephrology of Hunan Children's Hospital, Hunan Institute for Pediatric Research, 86 Ziyuan Road, Changsha, 410007, Hunan, People's Republic of China
- Academy of Pediatrics of University of South China, 86 Ziyuan Road, Changsha, 410007, Hunan, People's Republic of China
| | - Ying Wang
- Department of Nephrology of Hunan Children's Hospital, Hunan Institute for Pediatric Research, 86 Ziyuan Road, Changsha, 410007, Hunan, People's Republic of China
- Academy of Pediatrics of University of South China, 86 Ziyuan Road, Changsha, 410007, Hunan, People's Republic of China
| | - Tuanhong Xia
- Department of Nephrology of Hunan Children's Hospital, Hunan Institute for Pediatric Research, 86 Ziyuan Road, Changsha, 410007, Hunan, People's Republic of China
- Academy of Pediatrics of University of South China, 86 Ziyuan Road, Changsha, 410007, Hunan, People's Republic of China
| | - Niu Ding
- Department of Nephrology of Hunan Children's Hospital, Hunan Institute for Pediatric Research, 86 Ziyuan Road, Changsha, 410007, Hunan, People's Republic of China
- Academy of Pediatrics of University of South China, 86 Ziyuan Road, Changsha, 410007, Hunan, People's Republic of China
| | - Yan Yin
- Department of Nephrology of Hunan Children's Hospital, Hunan Institute for Pediatric Research, 86 Ziyuan Road, Changsha, 410007, Hunan, People's Republic of China
| |
Collapse
|
41
|
Kierulf-Lassen C, Kristensen MLV, Birn H, Jespersen B, Nørregaard R. No Effect of Remote Ischemic Conditioning Strategies on Recovery from Renal Ischemia-Reperfusion Injury and Protective Molecular Mediators. PLoS One 2015; 10:e0146109. [PMID: 26720280 PMCID: PMC4697851 DOI: 10.1371/journal.pone.0146109] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2015] [Accepted: 12/14/2015] [Indexed: 01/31/2023] Open
Abstract
Ischemia-reperfusion injury (IRI) is the major cause of acute kidney injury. Remote ischemic conditioning (rIC) performed as brief intermittent sub-lethal ischemia and reperfusion episodes in a distant organ may protect the kidney against IRI. Here we investigated the renal effects of rIC applied either prior to (remote ischemic preconditioning; rIPC) or during (remote ischemic perconditioning; rIPerC) sustained ischemic kidney injury in rats. The effects were evaluated as differences in creatinine clearance (CrCl) rate, tissue tubular damage marker expression, and potential kidney recovery mediators. One week after undergoing right-sided nephrectomy, rats were randomly divided into four groups: sham (n = 7), ischemia and reperfusion (IR; n = 10), IR+rIPC (n = 10), and IR+rIPerC (n = 10). The rIC was performed as four repeated episodes of 5-minute clamping of the infrarenal aorta followed by 5-minute release either before or during 37 minutes of left renal artery clamping representing the IRI. Urine and blood were sampled prior to ischemia as well as 3 and 7 days after reperfusion. The kidney was harvested for mRNA and protein isolation. Seven days after IRI, the CrCl change from baseline values was similar in the IR (δ: 0.74 mL/min/kg [-0.45 to 1.94]), IR+rIPC (δ: 0.21 mL/min/kg [-0.75 to 1.17], p > 0.9999), and IR+rIPerC (δ: 0.41 mL/min/kg [-0.43 to 1.25], p > 0.9999) groups. Kidney function recovery was associated with a significant up-regulation of phosphorylated protein kinase B (pAkt), extracellular regulated kinase 1/2 (pERK1/2), and heat shock proteins (HSPs) pHSP27, HSP32, and HSP70, but rIC was not associated with any significant differences in tubular damage, inflammatory, or fibrosis marker expression. In our study, rIC did not protect the kidney against IRI. However, on days 3-7 after IRI, all groups recovered renal function. This was associated with pAkt and pERK1/2 up-regulation and increased HSP expression at day 7.
Collapse
Affiliation(s)
- Casper Kierulf-Lassen
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
- Department of Renal Medicine, Aarhus University Hospital, Aarhus, Denmark
- * E-mail:
| | | | - Henrik Birn
- Department of Renal Medicine, Aarhus University Hospital, Aarhus, Denmark
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
| | - Bente Jespersen
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
- Department of Renal Medicine, Aarhus University Hospital, Aarhus, Denmark
| | - Rikke Nørregaard
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| |
Collapse
|
42
|
Kim JI, Noh MR, Kim KY, Jang HS, Kim HY, Park KM. Methionine sulfoxide reductase A deficiency exacerbates progression of kidney fibrosis induced by unilateral ureteral obstruction. Free Radic Biol Med 2015. [PMID: 26210777 DOI: 10.1016/j.freeradbiomed.2015.07.018] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Methionine sulfoxide reductase A (MsrA), which stereospecifically catalyzes the reduction of methionine-S-sulfoxide, is an important reactive oxygen species (ROS) scavenger. Tissue fibrosis is a maladaptive repair process following injury, associated with oxidative stress. In this study, we investigated the role of MsrA in unilateral ureteral obstruction (UUO)-induced kidney fibrosis and its underlying mechanisms by using MsrA gene-deleted mice (MsrA(-/-)). MsrA deletion increased collagen deposition in the interstitium and the expression of collagen III and α-smooth muscle actin in the UUO kidneys, indicating that MsrA deficiency exacerbated the progression of UUO-induced kidney fibrosis. UUO reduced the kidney expression of MsrA, MsrB1, and MsrB2, thereby decreasing MsrA and MsrB activity. UUO increased hydrogen peroxide and lipid peroxidation levels and the ratio of oxidized glutathione (GSSG) to total glutathione (GSH) in the kidneys. The UUO-induced elevations in the levels of these oxidative stress markers and leukocyte markers were much higher in the MsrA(-/-) than in the MsrA(+/+) kidneys, the latter suggesting that the exacerbated kidney fibrosis in MsrA(-/-) mice was associated with enhanced inflammatory responses. Collectively, our data suggest that MsrA plays a protective role in the progression of UUO-induced kidney fibrosis via suppression of fibrotic responses caused by oxidative stress and inflammation.
Collapse
Affiliation(s)
- Jee In Kim
- Department of Molecular Medicine and MRC, Keimyung University School of Medicine, Daegu 705-717, Republic of Korea
| | - Mi Ra Noh
- Department of Anatomy and BK21 Plus Program, Kyungpook National University School of Medicine, Daegu 700-422, Republic of Korea
| | - Ki Young Kim
- Department of Biochemistry and Molecular Biology, Yeungnam University College of Medicine, Daegu 705-717, Republic of Korea
| | - Hee-Seong Jang
- Department of Anatomy and BK21 Plus Program, Kyungpook National University School of Medicine, Daegu 700-422, Republic of Korea
| | - Hwa-Young Kim
- Department of Biochemistry and Molecular Biology, Yeungnam University College of Medicine, Daegu 705-717, Republic of Korea.
| | - Kwon Moo Park
- Department of Anatomy and BK21 Plus Program, Kyungpook National University School of Medicine, Daegu 700-422, Republic of Korea.
| |
Collapse
|
43
|
Kapitsinou PP, Haase VH. Molecular mechanisms of ischemic preconditioning in the kidney. Am J Physiol Renal Physiol 2015; 309:F821-34. [DOI: 10.1152/ajprenal.00224.2015] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2015] [Accepted: 08/21/2015] [Indexed: 12/26/2022] Open
Abstract
More effective therapeutic strategies for the prevention and treatment of acute kidney injury (AKI) are needed to improve the high morbidity and mortality associated with this frequently encountered clinical condition. Ischemic and/or hypoxic preconditioning attenuates susceptibility to ischemic injury, which results from both oxygen and nutrient deprivation and accounts for most cases of AKI. While multiple signaling pathways have been implicated in renoprotection, this review will focus on oxygen-regulated cellular and molecular responses that enhance the kidney's tolerance to ischemia and promote renal repair. Central mediators of cellular adaptation to hypoxia are hypoxia-inducible factors (HIFs). HIFs play a crucial role in ischemic/hypoxic preconditioning through the reprogramming of cellular energy metabolism, and by coordinating adenosine and nitric oxide signaling with antiapoptotic, oxidative stress, and immune responses. The therapeutic potential of HIF activation for the treatment and prevention of ischemic injuries will be critically examined in this review.
Collapse
Affiliation(s)
- Pinelopi P. Kapitsinou
- Departments of Medicine, Anatomy and Cell Biology, and the Kidney Institute, University of Kansas Medical Center, Kansas City, Kansas
| | - Volker H. Haase
- Departments of Medicine, Cancer Biology, and Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, Tennessee; and
- Medicine and Research Services, Department of Veterans Affairs Hospital, Tennessee Valley Healthcare System, Nashville, Tennessee
| |
Collapse
|
44
|
Liu L, Liu C, Hou L, Lv J, Wu F, Yang X, Ren S, Ji W, Wang M, Chen L. Protection against ischemia/reperfusion‑induced renal injury by co‑treatment with erythropoietin and sodium selenite. Mol Med Rep 2015; 12:7933-40. [PMID: 26647839 PMCID: PMC4758319 DOI: 10.3892/mmr.2015.4426] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2014] [Accepted: 09/09/2015] [Indexed: 01/07/2023] Open
Abstract
Ischemia/reperfusion injury (IRI) has lzong been an area of concern and focus of investigations. Erythropoietin (EPO) exhibits multiple protective effects, and selenium is an antioxidant trace element in the body, however, there have been no reports concerning the effects of EPO combined with sodium selenite on IRI. In the present study, a mouse model of renal IRI (RIRI) was pre–treated with EPO and sodium selenite to determine the most appropriate combination ratio of the two for further investigation. The results revealed that EPO and sodium selenite had synergistic protective effects in RIRI. EPO was identified as the predominant treatment component, with sodium selenite serving as an adjuvant, and combination treatment was markedly more effective, compared with treatment with either drug alone. The optimal ratio of treatment was 10:1 (10 IU EPO: 1 µg sodium selenite). The results indicated that RIRI markedly induced renal injury, as evidenced by elevated levels of blood urea nitrogen (BUN), as well as higher pathological scores, based on hematoxylin and eosin staining. Pre–treatment with EPO and sodium selenite significantly decreased serum expression levels of BUN and malonaldehyde, and increased the expression levels of superoxide dismutase, glutathione peroxidase and nitric oxide (NO), compared with the model group. Furthermore, co treatment with EPO and sodium selenite upregulated the protein expression levels of phosphatidylinositol 3 kinase (PI3K) in renal tissue samples. Together, the results suggested that co administration of EPO and sodium selenite effectively ameliorates IRI induced renal injury by reducing oxidative stress and activating the PI3K/NO signaling pathway.
Collapse
Affiliation(s)
- Lu Liu
- Department of Clinical Medicine, College of Clinical Medicine, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi 710061, P.R. China
| | - Chao Liu
- Department of Clinical Medicine, College of Clinical Medicine, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi 710061, P.R. China
| | - Lan Hou
- Department of Clinical Medicine, College of Clinical Medicine, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi 710061, P.R. China
| | - Juan Lv
- Department of Pharmacology, College of Basic Medicine, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi 710061, P.R. China
| | - Fang Wu
- Department of Clinical Medicine, College of Clinical Medicine, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi 710061, P.R. China
| | - Xuefei Yang
- Department of Pharmacology, College of Basic Medicine, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi 710061, P.R. China
| | - Shuting Ren
- Department of Pathology, College of Basic Medicine, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi 710061, P.R. China
| | - Wenjun Ji
- Department of Pharmacology, College of Basic Medicine, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi 710061, P.R. China
| | - Meng Wang
- Department of Pharmacology, College of Basic Medicine, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi 710061, P.R. China
| | - Lina Chen
- Department of Pharmacology, College of Basic Medicine, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi 710061, P.R. China
| |
Collapse
|
45
|
Kierulf-Lassen C, Nieuwenhuijs-Moeke GJ, Krogstrup NV, Oltean M, Jespersen B, Dor FJMF. Molecular Mechanisms of Renal Ischemic Conditioning Strategies. Eur Surg Res 2015; 55:151-83. [PMID: 26330099 DOI: 10.1159/000437352] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2015] [Accepted: 07/02/2015] [Indexed: 11/19/2022]
Abstract
Ischemia-reperfusion injury is the leading cause of acute kidney injury in a variety of clinical settings such as renal transplantation and hypovolemic and/or septic shock. Strategies to reduce ischemia-reperfusion injury are obviously clinically relevant. Ischemic conditioning is an inherent part of the renal defense mechanism against ischemia and can be triggered by short periods of intermittent ischemia and reperfusion. Understanding the signaling transduction pathways of renal ischemic conditioning can promote further clinical translation and pharmacological advancements in this era. This review summarizes research on the molecular mechanisms underlying both local and remote ischemic pre-, per- and postconditioning of the kidney. The different types of conditioning strategies in the kidney recruit similar powerful pro-survival mechanisms. Likewise, renal ischemic conditioning mobilizes many of the same protective signaling pathways as in other organs, but differences are recognized.
Collapse
|
46
|
Impellizzeri D, Bruschetta G, Ahmad A, Crupi R, Siracusa R, Di Paola R, Paterniti I, Prosdocimi M, Esposito E, Cuzzocrea S. Effects of palmitoylethanolamide and silymarin combination treatment in an animal model of kidney ischemia and reperfusion. Eur J Pharmacol 2015; 762:136-49. [DOI: 10.1016/j.ejphar.2015.05.010] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2015] [Revised: 04/21/2015] [Accepted: 05/13/2015] [Indexed: 12/31/2022]
|
47
|
Kim UJ, Won R, Lee KH. Neuroprotective effects of okadaic acid following oxidative injury in organotypic hippocampal slice culture. Brain Res 2015; 1618:241-8. [DOI: 10.1016/j.brainres.2015.05.039] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2015] [Revised: 05/22/2015] [Accepted: 05/29/2015] [Indexed: 10/23/2022]
|
48
|
Tang C, He L, Liu J, Dong Z. Mitophagy: Basic Mechanism and Potential Role in Kidney Diseases. KIDNEY DISEASES 2015; 1:71-9. [PMID: 27536667 DOI: 10.1159/000381510] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/25/2015] [Revised: 03/03/2015] [Indexed: 01/17/2023]
Abstract
BACKGROUND Mitochondria play fundamental roles in cellular metabolism, signaling, and viability. Disruption of mitochondria not only leads to dysfunction of the organelles but also activates mechanisms of cell injury and death, contributing to the pathogenesis of various diseases. SUMMARY Removal of damaged mitochondria is therefore crucial for cellular homeostasis and survival. Mitophagy, the selective elimination of mitochondria via autophagy, is an important mechanism of mitochondrial quality control in physiological and pathological conditions. Defects in mitophagy have been implicated in a variety of human disorders, including both acute and chronic kidney diseases. However, the role and regulatory mechanisms of mitophagy in kidney cells and tissues remain largely unknown. KEY MESSAGE This review provides updated information on mitophagy and suggests a potential role of mitophagy in renal pathophysiology.
Collapse
Affiliation(s)
- Chengyuan Tang
- Department of Nephrology, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Liyu He
- Department of Nephrology, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Jing Liu
- Department of Nephrology, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Zheng Dong
- Department of Nephrology, The Second Xiangya Hospital, Central South University, Changsha, China; Department of Cellular Biology and Anatomy, Medical College of Georgia at Georgia Regents University and Charlie Norwood VA Medical Center, Augusta, Ga., USA
| |
Collapse
|
49
|
Ferjani H, Achour A, Bacha H, Abid S. Tacrolimus and mycophenolate mofetil associations. Hum Exp Toxicol 2015; 34:1119-32. [DOI: 10.1177/0960327115569812] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Gastrointestinal risk factors after organ transplantation are prevalent, due to the chronic use of immunosuppressant. The immunosuppressive drugs such as tacrolimus/mycophenolate mofetil (TAC/MMF) association are the most commonly used therapy. TAC and MMF have been implicated in gastrotoxicity, but their direct effects, alone and combined, on intestinal cells are not completely elucidated. This study investigated the effect of TAC and MMF alone and combined on human colon carcinoma cells. Our results demonstrated that TAC and MMF individually inhibit clearly cells proliferation, enhanced free radicals, lipid peroxidation production, induced DNA lesions and reduced mitochondrial membrane potential. In this study, we also showed that the two molecules TAC and MMF combined at high concentrations amplified the cell damage. Furthermore, the TAC (5 µM) prevented cell death induced by MMF (half maximal inhibitory concentration (IC50)). Also, MMF (50 µM) induced cytoprotection in HCT116 cells against TAC (IC50) toxicity. Our findings provide additional evidence that oxidative damage is the major contribution of TAC and MMF combined toxicities. In fact, MMF and TAC exert a gastroprotective effect by modulating reactive oxygen species production. These data underscore the pleiotropic effect of TAC and MMF on HCT116 cells that play a preventive and critical role on intestinal function.
Collapse
Affiliation(s)
- H Ferjani
- Laboratory of Research on Biologically Compatible Compounds, Faculty of Dentistry, Monastir, Tunisia
| | - A Achour
- Department of Nephrology, Dialysis and Transplant, University Hospital of Sahloul, Sousse, Tunisia
| | - H Bacha
- Laboratory of Research on Biologically Compatible Compounds, Faculty of Dentistry, Monastir, Tunisia
| | - S Abid
- Laboratory of Research on Biologically Compatible Compounds, Faculty of Dentistry, Monastir, Tunisia
| |
Collapse
|
50
|
Angiotensin II removes kidney resistance conferred by ischemic preconditioning. BIOMED RESEARCH INTERNATIONAL 2014; 2014:602149. [PMID: 25243156 PMCID: PMC4163347 DOI: 10.1155/2014/602149] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/30/2014] [Revised: 07/06/2014] [Accepted: 08/06/2014] [Indexed: 01/13/2023]
Abstract
Ischemic preconditioning (IPC) by ischemia/reperfusion (I/R) renders resistance to the kidney. Strong IPC triggers kidney fibrosis, which is involved in angiotensin II (AngII) and its type 1 receptor (AT1R) signaling. Here, we investigated the role of AngII/AT1R signal pathway in the resistance of IPC kidneys to subsequent I/R injury. IPC of kidneys was generated by 30 minutes of bilateral renal ischemia and 8 days of reperfusion. Sham-operation was performed to generate control (non-IPC) mice. To examine the roles of AngII and AT1R in IPC kidneys to subsequent I/R, IPC kidneys were subjected to either 30 minutes of bilateral kidney ischemia or sham-operation following treatment with AngII, losartan (AT1R blocker), or AngII plus losartan. IPC kidneys showed fibrotic changes, decreased AngII, and increased AT1R expression. I/R dramatically increased plasma creatinine concentrations in non-IPC mice, but not in IPC mice. AngII treatment in IPC mice resulted in enhanced morphological damage, oxidative stress, and inflammatory responses, with functional impairment, whereas losartan treatment reversed these effects. However, AngII treatment in non-IPC mice did not change I/R-induced injury. AngII abolished the resistance of IPC kidneys to subsequent I/R via the enhancement of oxidative stress and inflammatory responses, suggesting that the AngII/AT1R signaling pathway is associated with outcome in injury-experienced kidney.
Collapse
|