1
|
Xu X, Song H, Zhang L, Chen C, Zhang X, Liu Y, Li C, Fu Q. Effects of Coatings on Antioxidant Enzyme Activities, Histopathology, and Transcriptome Profiles of Kidney Tissue in Larimichthys crocea. Genes (Basel) 2025; 16:392. [PMID: 40282352 PMCID: PMC12026950 DOI: 10.3390/genes16040392] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2025] [Revised: 03/23/2025] [Accepted: 03/27/2025] [Indexed: 04/29/2025] Open
Abstract
Background: As an innovative approach to deep-sea aquaculture, fish farm vessels offer a dual benefit by alleviating the pressure on offshore fishing resources while providing an additional high-quality protein source. However, the potential impacts of vessel coatings on farmed fish remain poorly understood. Methods: In this study, to investigate the effects of vessel coatings on the large yellow croaker (Larimichthys crocea), we established four experimental groups with coating concentrations at 1-fold, 10-fold, 20-fold, and 80-fold levels. Antioxidant enzyme activities in kidney tissues were measured across all groups, while histological and transcriptome analyses were specifically conducted for the 1-fold and 80-fold concentration groups. Results: Firstly, significant alterations in antioxidant enzyme activity were observed in the 80-fold concentration group. Moreover, histological analysis demonstrated more severe pathological changes in kidney tissue at the higher concentration, including interstitial hemorrhage and tubular epithelial cell fatty degeneration. In addition, we identified 11,902 differentially expressed genes (DEGs) by high-throughput sequencing. KEGG pathway enrichment analysis revealed that the DEGs were predominantly involved in critical biological processes, including endoplasmic reticulum protein processing, oxidative phosphorylation, cytokine-cytokine receptor interactions, cell cycle regulation, DNA replication, and PPAR signaling pathways. Finally, the validation of nine selected DEGs through quantitative real-time PCR (qRT-PCR) showed significant correlation with RNA-Seq data, confirming the reliability of our transcriptome analysis. Conclusions: This study provides preliminary insights into the antioxidant stress response mechanisms of L. crocea to coating exposure and establishes a theoretical foundation for optimizing healthy fish farming practices in aquaculture vessels.
Collapse
Affiliation(s)
- Xuan Xu
- School of Marine Science and Engineering, Qingdao Agricultural University, Qingdao 266109, China; (X.X.); (C.C.); (X.Z.); (Y.L.); (C.L.)
- Qingdao Conson Oceantec Valley Development Co., Ltd., Qingdao 266237, China; (H.S.); (L.Z.)
| | - Huayu Song
- Qingdao Conson Oceantec Valley Development Co., Ltd., Qingdao 266237, China; (H.S.); (L.Z.)
| | - Lu Zhang
- Qingdao Conson Oceantec Valley Development Co., Ltd., Qingdao 266237, China; (H.S.); (L.Z.)
| | - Chonghui Chen
- School of Marine Science and Engineering, Qingdao Agricultural University, Qingdao 266109, China; (X.X.); (C.C.); (X.Z.); (Y.L.); (C.L.)
| | - Xiaoxu Zhang
- School of Marine Science and Engineering, Qingdao Agricultural University, Qingdao 266109, China; (X.X.); (C.C.); (X.Z.); (Y.L.); (C.L.)
| | - Yiying Liu
- School of Marine Science and Engineering, Qingdao Agricultural University, Qingdao 266109, China; (X.X.); (C.C.); (X.Z.); (Y.L.); (C.L.)
| | - Chao Li
- School of Marine Science and Engineering, Qingdao Agricultural University, Qingdao 266109, China; (X.X.); (C.C.); (X.Z.); (Y.L.); (C.L.)
| | - Qiang Fu
- School of Marine Science and Engineering, Qingdao Agricultural University, Qingdao 266109, China; (X.X.); (C.C.); (X.Z.); (Y.L.); (C.L.)
- Qingdao Conson Oceantec Valley Development Co., Ltd., Qingdao 266237, China; (H.S.); (L.Z.)
| |
Collapse
|
2
|
Yan J, Kim H, Kim B, Piao H, Jang JY, Kang TK, Lee W, Kim D, Jo S, Shin D, Abuzar SMD, Kim ML, Yang J, Jon S. Synthetic Bilirubin-Based Nanomedicine Protects Against Renal Ischemia/Reperfusion Injury Through Antioxidant and Immune-Modulating Activity. Adv Healthc Mater 2025; 14:e2403846. [PMID: 39846887 PMCID: PMC11912105 DOI: 10.1002/adhm.202403846] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2024] [Revised: 01/09/2025] [Indexed: 01/24/2025]
Abstract
Renal ischemia/reperfusion injury (IRI) is a common form of acute kidney injury. The basic mechanism underlying renal IRI is acute inflammation, where oxidative stress plays an important role. Although bilirubin exhibits potent reactive oxygen species (ROS)-scavenging properties, its clinical application is hindered by problems associated with solubility, stability, and toxicity. In this study, BX-001N, a synthetic polyethylene glycol-conjugated bilirubin 3α nanoparticle is developed and assessed its renoprotective effects in renal IRI. Intravenous administration of BX-001N led to increase uptake in the kidneys with minimal migration to the brain after IRI. Peri-IRI BX-001N administration improves renal function and attenuates renal tissue injury and tubular apoptosis to a greater extent than free bilirubin on day 1 after IRI. BX-001N suppressed renal infiltration of inflammatory cells and reduced expression of TNF-α and MCP-1. Furthermore, BX-001N increases renal tubular regeneration on day 3 and suppresses renal fibrosis on day 28. BX-001N decreases the renal expressions of dihydroethidium, malondialdehyde, and nitrotyrosine after IRI. In conclusion, BX-001N, the first Good Manufacturing Practice-grade synthetic bilirubin-based nanomedicine attenuates acute renal injury and chronic fibrosis by suppressing ROS generation and inflammation after IRI. It shows adequate safety profiles and holds promise as a new therapy for renal IRI.
Collapse
Affiliation(s)
- Ji‐Jing Yan
- Division of NephrologyDepartment of Internal MedicineYonsei University College of MedicineSeoul03722Republic of Korea
- The Research Institute for TransplantationYonsei University College of MedicineSeoul03722Republic of Korea
| | - Hyunjin Kim
- BILIX.Co., Ltd.YonginGyeonggi‐do16942Republic of Korea
| | - Bomin Kim
- Division of NephrologyDepartment of Internal MedicineYonsei University College of MedicineSeoul03722Republic of Korea
- The Research Institute for TransplantationYonsei University College of MedicineSeoul03722Republic of Korea
| | - Honglin Piao
- Division of NephrologyDepartment of Internal MedicineYonsei University College of MedicineSeoul03722Republic of Korea
- The Research Institute for TransplantationYonsei University College of MedicineSeoul03722Republic of Korea
| | - Joon Young Jang
- Division of NephrologyDepartment of Internal MedicineYonsei University College of MedicineSeoul03722Republic of Korea
- The Research Institute for TransplantationYonsei University College of MedicineSeoul03722Republic of Korea
| | - Tae Kyeom Kang
- Natural Product Research CenterKorea Institute of Science & TechnologyGangneungGangwon‐do25451Republic of Korea
| | - Wook‐Bin Lee
- Natural Product Research CenterKorea Institute of Science & TechnologyGangneungGangwon‐do25451Republic of Korea
| | - Dohyeon Kim
- Department of Biological SciencesKAIST Institute for the BioCenturyCenter for Precision Bio‐NanomedicineKorea Advanced Institute of Science and TechnologyDaejeon34141Republic of Korea
| | - Seunghyun Jo
- BILIX.Co., Ltd.YonginGyeonggi‐do16942Republic of Korea
| | | | | | - Myung L. Kim
- BILIX.Co., Ltd.YonginGyeonggi‐do16942Republic of Korea
| | - Jaeseok Yang
- Division of NephrologyDepartment of Internal MedicineYonsei University College of MedicineSeoul03722Republic of Korea
- The Research Institute for TransplantationYonsei University College of MedicineSeoul03722Republic of Korea
| | - Sangyong Jon
- Department of Biological SciencesKAIST Institute for the BioCenturyCenter for Precision Bio‐NanomedicineKorea Advanced Institute of Science and TechnologyDaejeon34141Republic of Korea
| |
Collapse
|
3
|
Güneş I, Dursun AD, Özdemir Ç, Küçük A, Sezen ŞC, Arslan M, Özer A. Effects of Cerium Oxide on Kidney and Liver Tissue Damage in an Experimental Myocardial Ischemia-Reperfusion Model of Distant Organ Damage. MEDICINA (KAUNAS, LITHUANIA) 2024; 60:2044. [PMID: 39768923 PMCID: PMC11728079 DOI: 10.3390/medicina60122044] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/17/2024] [Revised: 11/25/2024] [Accepted: 12/06/2024] [Indexed: 01/16/2025]
Abstract
Background and Objectives: Ischemia-reperfusion (I/R) injury is a process in which impaired perfusion is restored by restoring blood flow and tissue recirculation. Nanomedicine uses cutting-edge technologies that emerge from interdisciplinary influences. In the literature, there are very few in vivo and in vitro studies on how cerium oxide (CeO2) affects systemic anti-inflammatory response and inflammation. Therefore, in our study, we aimed to investigate whether CeO2 administration has a protective effect against myocardial I/R injury in the liver and kidneys. Materials and Methods: Twenty-four rats were randomly divided into four groups after obtaining approval from an ethics committee. A control (group C), cerium oxide (group CO), IR (group IR), and Cerium oxide-IR (CO-IR group) groups were formed. Intraperitoneal CeO2 was administered at a dose of 0.5 mg/kg 30 min before left thoracotomy and left main coronary (LAD) ligation, and myocardial muscle ischemia was induced for 30 min. After LAD ligation was removed, reperfusion was performed for 120 min. All rats were euthanized using ketamine, and blood was collected. Liver and kidney tissue samples were evaluated histopathologically. Serum AST (aspartate aminotransferase), ALT (alanine aminotransaminase), GGT (gamma-glutamyl transferase), glucose, TOS (Total Oxidant Status), and TAS (Total Antioxidant Status) levels were also measured. Results: Necrotic cell and mononuclear cell infiltration in the liver parenchyma of rats in the IR group was observed to be significantly increased compared to the other groups. Hepatocyte degeneration was greater in the IR group compared to groups C and CO. Vascular vacuolization and hypertrophy, tubular degeneration, and necrosis were increased in the kidney tissue of the IR group compared to the other groups. Tubular dilatation was significantly higher in the IR group than in the C and CO groups. TOS was significantly higher in all groups than in the IR group (p < 0.0001, p < 0.0001, and p = 0.006, respectively). However, TAS level was lower in the IR group than in the other groups (p = 0.002, p = 0.020, and p = 0.031, respectively). Renal and liver histopathological findings decreased significantly in the CO-IR group compared to the IR group. A decrease in the TOS level and an increase in the TAS level were found compared to the IR group. The AST, ALT, GGT, and Glucose levels are shown. Conclusions: CeO2 administered before ischemia-reperfusion reduced oxidative stress and ameliorated IR-induced damage in distant organs. We suggest that CeO2 exerts protective effects in the myocardial IR model.
Collapse
Affiliation(s)
- Işın Güneş
- Department of Anesthesiology and Reanimation, Erciyes University Faculty of Medicine, Kayseri 38039, Turkey;
| | - Ali Doğan Dursun
- Department of Physiology, Atılım University Faculty of Medicine, Ankara 06560, Turkey;
- Vocational School of Health Services, Atilim University, Cankaya, Ankara 06805, Turkey
- Home Care Services, Medicana International Ankara Hospital, Cankaya, Ankara 06520, Turkey
| | - Çağrı Özdemir
- Department of Anesthesiology and Reanimation, Gazi University Faculty of Medicine, Ankara 06560, Turkey;
| | - Ayşegül Küçük
- Department of Physiology, Kutahya Health Sciences University Faculty of Medicine, Kutahya 43100, Turkey;
| | - Şaban Cem Sezen
- Department of Histology and Embryology, Kırıkkale University Faculty of Medicine, Kırıkkale 71000, Turkey;
| | - Mustafa Arslan
- Department of Anesthesiology and Reanimation, Gazi University Faculty of Medicine, Ankara 06560, Turkey;
- Application and Research Centre for Life Sciences, Gazi University, Ankara 06560, Turkey
- Centre for Laboratory Animal Breeding and Experimental Research (GÜDAM), Gazi University, Ankara 06560, Turkey
| | - Abdullah Özer
- Department Cardiovascular Surgery, Gazi University Faculty of Medicine, Ankara 06560, Turkey;
| |
Collapse
|
4
|
Jiang L, Wang S, Tan Y, Su T. Postpartum Renal Cortical Necrosis: A Case Series. Kidney Med 2024; 6:100892. [PMID: 39314861 PMCID: PMC11417324 DOI: 10.1016/j.xkme.2024.100892] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/25/2024] Open
Abstract
Rationale & Objective Postpartum renal cortical necrosis (postpartum RCN) is a severe form of obstetric acute kidney injury. This study aimed to identify clinicopathologic features in Chinese postpartum RCN cases to determine how pathologic findings may contribute to the treatment and prognosis. Study Design Single-center, case series. Setting & Participants Twelve patients with postpartum RCN had kidney biopsies at Peking University First Hospital between 2014 and 2021. The diagnosis of postpartum RCN was made according to typical magnetic resonance imaging or pathologic features. Clinical, laboratory, and pathologic data were compared between patients with estimated glomerular filtration rate <30 (poor outcome) and ≥30 mL/min/1.73 m2 after 6 months. Observations All patients with postpartum RCN presented with stage 3 acute kidney injury attributed to a probable atypical hemolytic uremic syndrome. Pregnancy terminations occurred at a median gestational age of 35.5 weeks. Kidney biopsy was performed from 18 days to 4 months from delivery. On biopsy, hemoglobin, platelet count, and lactate dehydrogenase levels had been restored to 137 g/L, 214 × 109/L, and 231.50 ± 65.01 U/L, respectively. Four patients exhibited poor outcome, demonstrating higher schistocyte count, serum creatinine, and mean arterial pressure at onset. Pathologically, glomerular segmental sclerosis was prevalent. The "not otherwise specified" variant was the most common type, followed by collapsing variant, cellular variant, and tip variant. Patients with poor kidney outcome had more glomerular coagulative necrosis, capillary thrombosis, extensive cortical coagulative necrosis, and pronounced arteriole/artery lesions including increased interlobular arteriole intimal edema and fibrin thrombosis, but a lower occurrence of segmental sclerosis. Limitations Limited sample size and retrospective design. Conclusions We identified key pathologic features in patients with postpartum RCN and atypical hemolytic uremic syndrome, highlighting the necessity for more effective therapeutic options. There is a clear demand for noninvasive biomarkers that can accurately track disease progression and inform treatment duration for long-term outcomes improvement.
Collapse
Affiliation(s)
- Lei Jiang
- Renal Division, Department of Medicine, Peking University First Hospital, Beijing, China
- Institute of Nephrology, Peking University, Beijing, China
- Renal Pathology Center, Institute of Nephrology, Peking University, Beijing, China
| | - Suxia Wang
- Renal Pathology Center, Institute of Nephrology, Peking University, Beijing, China
- Laboratory of Electron Microscopy, Pathological Center, Peking University First Hospital, Beijing, China
| | - Ying Tan
- Renal Division, Department of Medicine, Peking University First Hospital, Beijing, China
- Institute of Nephrology, Peking University, Beijing, China
- Renal Pathology Center, Institute of Nephrology, Peking University, Beijing, China
| | - Tao Su
- Renal Division, Department of Medicine, Peking University First Hospital, Beijing, China
- Institute of Nephrology, Peking University, Beijing, China
- Renal Pathology Center, Institute of Nephrology, Peking University, Beijing, China
| |
Collapse
|
5
|
Kapisiz A, Kaya C, Eryilmaz S, Karabulut R, Turkyilmaz Z, Inan MA, Gulbahar O, Sonmez K. Protective effects of lupeol in rats with renal ischemia‑reperfusion injury. Exp Ther Med 2024; 28:313. [PMID: 38911048 PMCID: PMC11190881 DOI: 10.3892/etm.2024.12602] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Accepted: 05/02/2024] [Indexed: 06/25/2024] Open
Abstract
Acute kidney injury (AKI) caused by ischemia and, exogenous or endogenous nephrotoxic agents poses a serious health issue. AKI is seen in 1% of all hospital admissions, 2-5% of hospitalizations and 67% of intensive care unit (ICU) patients. The in-hospital mortality rates for AKI is 40-50, and >50% for ICU patients. Ischemia-reperfusion (I/R) injury in the kidney can activate inflammatory responses and oxidative stress, resulting in AKI. The common endpoint in acute tubular necrosis is a cellular insult secondary to ischemia or direct toxins, which results in effacement of brush border, cell death and decreased function of tubular cells. The aim of the present study was to assess if the reported antioxidant and anti-inflammatory agent lupeol can exert any effects against renal I/R damage. In total, 24 Wistar Albino rats were randomly assigned into four groups of 6, namely Sham, lupeol, ischemia and therapy groups. In the lupeol group, intraperitoneal administration of 100 mg/kg lupeol was given 1 h before laparotomy, whilst only laparotomy was conducted in the sham group. The renal arteries of both kidneys were clamped for 45 min, 1 h after either intraperitoneal saline injection (in the ischemia group) or 100 mg/kg lupeol application (in the therapy group). The blood samples and renal tissues of all rats were collected after 24 h. In blood samples, blood urea nitrogen (BUN) was measured by the urease enzymatic method, and creatinine was measured by the kinetic Jaffe method. Using ELISA method, TNF-α and IL-6 levels were measured in the blood samples, whereas malondialdehyde (MDA), glutathione (GSH), caspase-3 levels were measured in kidney tissues. In addition, kidney histopathological analysis was performed by evaluating the degree of degeneration, tubular dilatation, interstitial lymphocyte infiltration, protein cylinders, necrosis and loss of brush borders. It was determined that renal damage occurred due to higher BUN, creatinine, MDA, TNF-α and caspase-3 values observed in the kidney tissues and blood samples of rats in ischemia group compared with the Sham group. Compared with those in the ischemia group, rats in the therapy group exhibited increased levels of GSH and reduced levels of BUN, TNF-α, MDA. Furthermore, the ischemia group also had reduced histopathological damage scores. Although differences in creatinine, IL-6 and caspase-3 levels were not statistically significant, they were markedly reduced in the treatment group. Taken together, these findings suggest that lupeol can prevent kidney damage as mainly evidenced by the reduced histopathological damage scores, decreased levels of oxidative stress and reduced levels of inflammatory markers. These properties may allow lupeol to be used in the treatment of AKI.
Collapse
Affiliation(s)
- Alparslan Kapisiz
- Department of Pediatric Surgery, Gazi University Faculty of Medicine, Yenimahalle, 06500 Ankara, Turkey
| | - Cem Kaya
- Department of Pediatric Surgery, Gazi University Faculty of Medicine, Yenimahalle, 06500 Ankara, Turkey
| | - Sibel Eryilmaz
- Department of Pediatric Surgery, Gazi University Faculty of Medicine, Yenimahalle, 06500 Ankara, Turkey
| | - Ramazan Karabulut
- Department of Pediatric Surgery, Gazi University Faculty of Medicine, Yenimahalle, 06500 Ankara, Turkey
| | - Zafer Turkyilmaz
- Department of Pediatric Surgery, Gazi University Faculty of Medicine, Yenimahalle, 06500 Ankara, Turkey
| | - Mehmet Arda Inan
- Department of Pathology, Gazi University Faculty of Medicine, Yenimahalle, 06500 Ankara, Turkey
| | - Ozlem Gulbahar
- Department of Biochemistry, Gazi University Faculty of Medicine, Yenimahalle, 06500 Ankara, Turkey
| | - Kaan Sonmez
- Department of Pediatric Surgery, Gazi University Faculty of Medicine, Yenimahalle, 06500 Ankara, Turkey
| |
Collapse
|
6
|
Li P, Huang Z, Duan X, Wang T, Yang S, Jiang D, Li J. PET image-guided kidney injury theranostics enabled by a bipyramidal DNA framework. Biomater Sci 2024; 12:2086-2095. [PMID: 38439626 DOI: 10.1039/d3bm01575k] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/06/2024]
Abstract
Understanding the pharmacokinetic profiles of nanomaterials in living organisms is essential for their application in disease treatment. Bipyramidal DNA frameworks (BDFs) are a type of DNA nanomaterial that have shown prospects in the fields of molecular imaging and therapy. To serve as a reference for disease-related studies involving the BDF, we constructed a 68Ga-BDF and employed positron emission tomography (PET) imaging to establish its pharmacokinetic model in healthy mice. Our investigation revealed that the BDF was primarily eliminated from the body via the urinary system. Ureteral obstruction could significantly alter the metabolism of the urinary system. By utilizing the established pharmacokinetic model, we sensitively observed distinct imaging indicators in unilateral ureteral obstruction and acute kidney injury (a complication of ureteral obstruction) mouse models. Furthermore, we observed that the BDF showed therapeutic effects in an AKI model. We believe that the established pharmacokinetic model and unique renal excretion characteristics of the BDF will provide researchers with more information for studying kidney diseases.
Collapse
Affiliation(s)
- Pinghui Li
- Inner Mongolia Medical University, Hohhot 010050, China
| | - Zhidie Huang
- Inner Mongolia Medical University, Hohhot 010050, China
| | - Xiaoyan Duan
- Department of Nuclear Medicine, The Affiliated Hospital of Inner Mongolia Medical University, Hohhot 010050, China.
- Inner Mongolia Key Laboratory of Molecular Imaging, Hohhot 010050, China
| | - Tao Wang
- Department of Nuclear Medicine, The Affiliated Hospital of Inner Mongolia Medical University, Hohhot 010050, China.
- Inner Mongolia Key Laboratory of Molecular Imaging, Hohhot 010050, China
| | - Shaowen Yang
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China.
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan 430022, China
| | - Dawei Jiang
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China.
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan 430022, China
| | - Jianbo Li
- Department of Nuclear Medicine, The Affiliated Hospital of Inner Mongolia Medical University, Hohhot 010050, China.
- Inner Mongolia Key Laboratory of Molecular Imaging, Hohhot 010050, China
| |
Collapse
|
7
|
Han X, Bi L, Yan J, Song P, Wang Y, Wang X, Wu Y, Ding X, Zhang H, Wang Y, Li X. Mesoscale size-promoted targeted therapy for acute kidney injury through combined RONS scavenging and inflammation alleviation strategy. Mater Today Bio 2024; 25:101002. [PMID: 38420141 PMCID: PMC10900835 DOI: 10.1016/j.mtbio.2024.101002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Revised: 02/13/2024] [Accepted: 02/13/2024] [Indexed: 03/02/2024] Open
Abstract
Acute kidney injury (AKI) is a heterogeneous, high-mortality clinical syndrome with diverse pathogenesis and prognosis, but it lacks the effective therapy clinically. Its pathogenesis is associated with production of reactive oxygen/nitrogen species and infiltration of inflammatory cells. To overcome these pathogenic factors and improve the therapeutic efficiency, we synthesized triptolide-loaded mesoscale polydopamine melanin-mimetic nanoparticles (MeNP4TP) as the antioxidant plus anti-inflammatory therapeutic platform to synergistically scavenge reactive oxygen/nitrogen species (RONS), inhibit the activity of macrophages and dendritic cells, and generate Treg cells for AKI therapy. It was demonstrated that mesoscale size was beneficial for MeNP4TP to specifically accumulate at renal tubule cells, and MeNP4TP could significantly attenuate oxidative stress, reduce proinflammatory immune cells in renal, and repair renal function in cisplatin-induced AKI mouse model. MeNP4TP might be a potential candidate to inhibit oxidative damages and inflammatory events in AKI.
Collapse
Affiliation(s)
- Xiaoqing Han
- Department of Urology, First Hospital of Jilin University, Changchun, 130021, China
- Laboratory of Chemical Biology, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, China
| | - Luopeng Bi
- Department of Urology, First Hospital of Jilin University, Changchun, 130021, China
| | - Jiao Yan
- Laboratory of Chemical Biology, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, China
| | - Panpan Song
- Laboratory of Chemical Biology, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, China
- University of Science and Technology of China, Hefei, Anhui, 230026, China
| | - Yanjing Wang
- Laboratory of Chemical Biology, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, China
- University of Science and Technology of China, Hefei, Anhui, 230026, China
| | - Xingbo Wang
- Laboratory of Chemical Biology, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, China
- University of Science and Technology of China, Hefei, Anhui, 230026, China
| | - Yunyun Wu
- School of Chemistry and Life Science, Changchun University of Technology, Changchun, 130012, China
| | - Xiaobo Ding
- Department of Radiology, First Hospital of Jilin University, Changchun, 130021, China
| | - Haiyuan Zhang
- Laboratory of Chemical Biology, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, China
- University of Science and Technology of China, Hefei, Anhui, 230026, China
| | - Yanbo Wang
- Department of Urology, First Hospital of Jilin University, Changchun, 130021, China
| | - Xi Li
- School of Chemistry and Life Science, Changchun University of Technology, Changchun, 130012, China
| |
Collapse
|
8
|
Rashid H, Jali A, Akhter MS, Abdi SAH. Molecular Mechanisms of Oxidative Stress in Acute Kidney Injury: Targeting the Loci by Resveratrol. Int J Mol Sci 2023; 25:3. [PMID: 38203174 PMCID: PMC10779152 DOI: 10.3390/ijms25010003] [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: 09/26/2023] [Revised: 10/31/2023] [Accepted: 11/07/2023] [Indexed: 01/12/2024] Open
Abstract
Reactive oxygen species are a group of cellular molecules that stand as double-edged swords, their good and bad being discriminated by a precise balance. Several metabolic reactions in the biological system generate these molecules that interact with cellular atoms to regulate functions ranging from cell homeostasis to cell death. A prooxidative state of the cell concomitant with decreased clearance of such molecules leads to oxidative stress, which contributes as a prime pathophysiological mechanism in various diseases including renal disorders, such as acute kidney injury. However, targeting the generation of oxidative stress in renal disorders by an antioxidant, resveratrol, is gaining considerable therapeutic importance and is known to improve the condition in preclinical studies. This review aims to discuss molecular mechanisms of oxidative stress in acute kidney injury and its amelioration by resveratrol. The major sources of data were PubMed and Google Scholar, with studies from the last five years primarily included, with significant earlier data also considered. Mitochondrial dysfunction, various enzymatic reactions, and protein misfolding are the major sources of reactive oxygen species in acute kidney injury, and interrupting these loci of generation or intersection with other cellular components by resveratrol can mitigate the severity of the condition.
Collapse
Affiliation(s)
- Hina Rashid
- Department of Pharmacology and Toxicology, College of Pharmacy, Jazan University, Jizan 45142, Saudi Arabia
| | - Abdulmajeed Jali
- Department of Pharmacology and Toxicology, College of Pharmacy, Jazan University, Jizan 45142, Saudi Arabia
| | - Mohammad Suhail Akhter
- Department of Medical Laboratory Technology, College of Applied Medical Sciences, Jazan University, Jizan 45142, Saudi Arabia
| | - Sayed Aliul Hasan Abdi
- Department of Pharmacy, Faculty of Clinical Pharmacy, Al Baha University, Al Baha 65711, Saudi Arabia
| |
Collapse
|
9
|
Gharaie S, Lee K, Noller K, Lo EK, Miller B, Jung HJ, Newman-Rivera AM, Kurzhagen JT, Singla N, Welling PA, Fan J, Cahan P, Noel S, Rabb H. Single cell and spatial transcriptomics analysis of kidney double negative T lymphocytes in normal and ischemic mouse kidneys. Sci Rep 2023; 13:20888. [PMID: 38017015 PMCID: PMC10684868 DOI: 10.1038/s41598-023-48213-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2023] [Accepted: 11/23/2023] [Indexed: 11/30/2023] Open
Abstract
T cells are important in the pathogenesis of acute kidney injury (AKI), and TCR+CD4-CD8- (double negative-DN) are T cells that have regulatory properties. However, there is limited information on DN T cells compared to traditional CD4+ and CD8+ cells. To elucidate the molecular signature and spatial dynamics of DN T cells during AKI, we performed single-cell RNA sequencing (scRNA-seq) on sorted murine DN, CD4+, and CD8+ cells combined with spatial transcriptomic profiling of normal and post AKI mouse kidneys. scRNA-seq revealed distinct transcriptional profiles for DN, CD4+, and CD8+ T cells of mouse kidneys with enrichment of Kcnq5, Klrb1c, Fcer1g, and Klre1 expression in DN T cells compared to CD4+ and CD8+ T cells in normal kidney tissue. We validated the expression of these four genes in mouse kidney DN, CD4+ and CD8+ T cells using RT-PCR and Kcnq5, Klrb1, and Fcer1g genes with the NIH human kidney precision medicine project (KPMP). Spatial transcriptomics in normal and ischemic mouse kidney tissue showed a localized cluster of T cells in the outer medulla expressing DN T cell genes including Fcer1g. These results provide a template for future studies in DN T as well as CD4+ and CD8+ cells in normal and diseased kidneys.
Collapse
Affiliation(s)
- Sepideh Gharaie
- Department of Medicine, Johns Hopkins University, School of Medicine, Ross 965, 720 Rutland Ave, Baltimore, MD, 21205, USA
| | - Kyungho Lee
- Department of Medicine, Johns Hopkins University, School of Medicine, Ross 965, 720 Rutland Ave, Baltimore, MD, 21205, USA
| | - Kathleen Noller
- Department of Biomedical Engineering, Johns Hopkins University, School of Medicine, Baltimore, MD, 21205, USA
| | - Emily K Lo
- Department of Biomedical Engineering, Johns Hopkins University, School of Medicine, Baltimore, MD, 21205, USA
| | - Brendan Miller
- Department of Biomedical Engineering, Johns Hopkins University, School of Medicine, Baltimore, MD, 21205, USA
| | - Hyun Jun Jung
- Department of Medicine, Johns Hopkins University, School of Medicine, Ross 965, 720 Rutland Ave, Baltimore, MD, 21205, USA
| | - Andrea M Newman-Rivera
- Department of Medicine, Johns Hopkins University, School of Medicine, Ross 965, 720 Rutland Ave, Baltimore, MD, 21205, USA
| | - Johanna T Kurzhagen
- Department of Medicine, Johns Hopkins University, School of Medicine, Ross 965, 720 Rutland Ave, Baltimore, MD, 21205, USA
| | - Nirmish Singla
- Department of Urology, Johns Hopkins University, School of Medicine, Baltimore, MD, 21205, USA
| | - Paul A Welling
- Department of Medicine, Johns Hopkins University, School of Medicine, Ross 965, 720 Rutland Ave, Baltimore, MD, 21205, USA
- Department of Physiology, Johns Hopkins University, School of Medicine, Baltimore, MD, 21205, USA
| | - Jean Fan
- Department of Biomedical Engineering, Johns Hopkins University, School of Medicine, Baltimore, MD, 21205, USA
| | - Patrick Cahan
- Department of Biomedical Engineering, Johns Hopkins University, School of Medicine, Baltimore, MD, 21205, USA
| | - Sanjeev Noel
- Department of Medicine, Johns Hopkins University, School of Medicine, Ross 965, 720 Rutland Ave, Baltimore, MD, 21205, USA
| | - Hamid Rabb
- Department of Medicine, Johns Hopkins University, School of Medicine, Ross 965, 720 Rutland Ave, Baltimore, MD, 21205, USA.
| |
Collapse
|
10
|
Janfeshan S, Masjedi F, Karimi Z. Protective effects of limb remote ischemic per-conditioning on the heart injury induced by renal ischemic-reperfusion through the interaction of the apelin with the RAS/iNOS pathway. BIOIMPACTS : BI 2023; 14:27567. [PMID: 38505676 PMCID: PMC10945303 DOI: 10.34172/bi.2023.27567] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/14/2022] [Revised: 01/28/2023] [Accepted: 03/13/2023] [Indexed: 03/21/2024]
Abstract
Introduction Remote ischemic conditioning upregulates endogenous protective pathways in response to ischemia-reperfusion injury. This study tested the hypothesis that limb remote ischemic per- conditioning (RIPerC) exerts cardioprotective effects via the renin-angiotensin system (RAS)/inducible nitric oxide synthase (iNOS)/apelin pathway. Methods Renal ischemia-reperfusion injury (I/R) was induced by bilateral occlusion of the renal pedicles for 60 minutes, followed by 24 hours of reperfusion; sham-operated rats served as controls. RIPerC was induced by four cycles (5 minutes) of limb ischemia-reperfusion along with bilateral renal ischemia. The functional disturbance was evaluated by renal (BUN and creatinine) and cardiac (troponin I and lactate dehydrogenase) injury biomarkers. Results Renal I/R injury increased renal and cardiac injury biomarkers that were reduced in the RIPerC group. Histopathological findings of the kidney and heart were also suggestive of amelioration injury-induced changes in the RIPerC group. Assessment of cardiac electrophysiology revealed that RIPerC ameliorated the decline in P wave duration without significantly affecting other cardiac electrophysiological changes. Further, renal I/R injury increased the plasma (322.40±34.01 IU/L), renal (8.27±1.10 mIU/mg of Protein), and cardiac (68.28±10.28 mIU/mg of protein) angiotensin-converting enzyme (ACE) activities in association with elevations in the plasma and urine nitrite (25.47±2.01 & 16.62±3.05 μmol/L) and nitrate (15.47±1.33 & 5.01±0.96 μmol/L) levels; these changes were reversed by RIPerC. Further, renal ischemia-reperfusion injury significantly (P=0.047) decreased the renal (but not cardiac) apelin mRNA expression, while renal and cardiac ACE2 (P<0.05) and iNOS (P=0.043) mRNA expressions were significantly increased compared to the sham group; these effects were largely reversed by RIPerC. Conclusion Our results indicated that RIPerC protects the heart against renal ischemia- reperfusion injury, likely via interaction of the apelin with the RAS/iNOS pathway.
Collapse
Affiliation(s)
- Sahar Janfeshan
- Shiraz Nephro-Urology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Fatemeh Masjedi
- Shiraz Nephro-Urology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Zeinab Karimi
- Shiraz Nephro-Urology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| |
Collapse
|
11
|
Huang F, Ren X, Yuan B, Yang W, Xu L, Zhang J, Zhang H, Geng M, Li X, Zhang F, Xu J, Zhu W, Ren S, Meng L, Lu S. Systemic Mutation of Ncf1 Ameliorates Obstruction-Induced Renal Fibrosis While Macrophage-Rescued NCF1 Further Alleviates Renal Fibrosis. Antioxid Redox Signal 2023. [PMID: 37392014 DOI: 10.1089/ars.2022.0195] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 07/02/2023]
Abstract
Aims: NCF1, a subunit of the NADPH oxidase 2 (NOX2), first described the expression in neutrophils and macrophages and participated in the pathogenesis from various systems. However, there are controversial findings on the role of NCF1 in different kinds of kidney diseases. In this study, we aim to pinpoint the specific role of NCF1 in the progression of renal fibrosis induced by obstruction. Results: In this study, NCF1 expression was upregulated in kidney biopsies of chronic kidney disease patients. The expression level of all subunits of the NOX2 complex was also significantly increased in the unilateral ureteral obstruction (UUO) kidney. Then, we used wild-type mice and Ncf1 mutant mice (Ncf1m1j mice) to perform UUO-induced renal fibrosis. Results demonstrated that Ncf1m1j mice exhibited mild renal fibrosis but increased macrophages count and CD11b+Ly6Chi macrophage proportion. Next, we compared the renal fibrosis degree between Ncf1m1j mice and Ncf1 macrophage-rescued mice (Ncf1m1j.Ncf1Tg-CD68 mice). We found that rescuing NCF1 expression in macrophages further alleviated renal fibrosis and decreased macrophage infiltration in the UUO kidney. In addition, flow cytometry data showed fewer CD11b+Ly6Chi macrophages in the kidney of the Ncf1m1j.Ncf1Tg-CD68 group than the Ncf1m1j group. Innovation: We first used the Ncf1m1j mice and Ncf1m1j.Ncf1Tg-CD68 mice to detect the role of NCF1 in the pathological process of renal fibrosis induced by obstruction. Also, we found that NCF1 expressed in different cell types exerts opposing effects on obstructive nephropathy. Conclusion: Taken together, our findings support that systemic mutation of Ncf1 ameliorates renal fibrosis induced by obstruction, and rescuing NCF1 in macrophages further alleviates renal fibrosis.
Collapse
Affiliation(s)
- Fumeng Huang
- Institute of Molecular and Translational Medicine (IMTM), and Department of Biochemistry and Molecular Biology, Xi'an Jiaotong University Health Science Center, Xi'an, China
- Key Laboratory of Environment and Genes Related to Diseases, Xi'an Jiaotong University, Ministry of Education, Xi'an, China
| | - Xiaomin Ren
- Institute of Molecular and Translational Medicine (IMTM), and Department of Biochemistry and Molecular Biology, Xi'an Jiaotong University Health Science Center, Xi'an, China
- Department of Hematology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Bingyu Yuan
- Institute of Molecular and Translational Medicine (IMTM), and Department of Biochemistry and Molecular Biology, Xi'an Jiaotong University Health Science Center, Xi'an, China
| | - Wenbo Yang
- Institute of Molecular and Translational Medicine (IMTM), and Department of Biochemistry and Molecular Biology, Xi'an Jiaotong University Health Science Center, Xi'an, China
| | - Lexuan Xu
- Institute of Molecular and Translational Medicine (IMTM), and Department of Biochemistry and Molecular Biology, Xi'an Jiaotong University Health Science Center, Xi'an, China
| | - Jing Zhang
- Institute of Molecular and Translational Medicine (IMTM), and Department of Biochemistry and Molecular Biology, Xi'an Jiaotong University Health Science Center, Xi'an, China
| | - Haonan Zhang
- Institute of Molecular and Translational Medicine (IMTM), and Department of Biochemistry and Molecular Biology, Xi'an Jiaotong University Health Science Center, Xi'an, China
| | - Manman Geng
- National Joint Engineering Research Center of Biodiagnostics and Biotherapy, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Xiaowei Li
- National Joint Engineering Research Center of Biodiagnostics and Biotherapy, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Fujun Zhang
- Institute of Molecular and Translational Medicine (IMTM), and Department of Biochemistry and Molecular Biology, Xi'an Jiaotong University Health Science Center, Xi'an, China
| | - Jing Xu
- Institute of Molecular and Translational Medicine (IMTM), and Department of Biochemistry and Molecular Biology, Xi'an Jiaotong University Health Science Center, Xi'an, China
| | - Wenhua Zhu
- Institute of Molecular and Translational Medicine (IMTM), and Department of Biochemistry and Molecular Biology, Xi'an Jiaotong University Health Science Center, Xi'an, China
| | - Shuting Ren
- Department of Pathology, School of Basic Medical Sciences, Health Science Center, Xi'an Jiaotong University, Xi'an, China
| | - Liesu Meng
- Institute of Molecular and Translational Medicine (IMTM), and Department of Biochemistry and Molecular Biology, Xi'an Jiaotong University Health Science Center, Xi'an, China
- Key Laboratory of Environment and Genes Related to Diseases, Xi'an Jiaotong University, Ministry of Education, Xi'an, China
- National Joint Engineering Research Center of Biodiagnostics and Biotherapy, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Shemin Lu
- Institute of Molecular and Translational Medicine (IMTM), and Department of Biochemistry and Molecular Biology, Xi'an Jiaotong University Health Science Center, Xi'an, China
- Key Laboratory of Environment and Genes Related to Diseases, Xi'an Jiaotong University, Ministry of Education, Xi'an, China
- National Joint Engineering Research Center of Biodiagnostics and Biotherapy, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| |
Collapse
|
12
|
Wruck W, Genfi AKA, Adjaye J. Natural Products in Renal-Associated Drug Discovery. Antioxidants (Basel) 2023; 12:1599. [PMID: 37627594 PMCID: PMC10451693 DOI: 10.3390/antiox12081599] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2023] [Accepted: 08/08/2023] [Indexed: 08/27/2023] Open
Abstract
The global increase in the incidence of kidney failure constitutes a major public health problem. Kidney disease is classified into acute and chronic: acute kidney injury (AKI) is associated with an abrupt decline in kidney function and chronic kidney disease (CKD) with chronic renal failure for more than three months. Although both kidney syndromes are multifactorial, inflammation and oxidative stress play major roles in the diversity of processes leading to these kidney malfunctions. Here, we reviewed various publications on medicinal plants with antioxidant and anti-inflammatory properties with the potential to treat and manage kidney-associated diseases in rodent models. Additionally, we conducted a meta-analysis to identify gene signatures and associated biological processes perturbed in human and mouse cells treated with antioxidants such as epigallocatechin gallate (EGCG), the active ingredient in green tea, and the mushroom Ganoderma lucidum (GL) and in kidney disease rodent models. We identified EGCG- and GL-regulated gene signatures linked to metabolism; inflammation (NRG1, E2F1, NFKB1 and JUN); ion signalling; transport; renal processes (SLC12A1 and LOX) and VEGF, ERBB and BDNF signalling. Medicinal plant extracts are proving to be effective for the prevention, management and treatment of kidney-associated diseases; however, more detailed characterisations of their targets are needed to enable more trust in their application in the management of kidney-associated diseases.
Collapse
Affiliation(s)
- Wasco Wruck
- Institute for Stem Cell Research and Regenerative Medicine, Medical Faculty, Heinrich Heine University, Moorenstr. 5, 40225 Düsseldorf, Germany
| | - Afua Kobi Ampem Genfi
- Department of Biochemistry and Molecular Biology, Faculty of Biosciences, University for Development Studies, Nyankpala P.O. Box TL 1882, Ghana
| | - James Adjaye
- Institute for Stem Cell Research and Regenerative Medicine, Medical Faculty, Heinrich Heine University, Moorenstr. 5, 40225 Düsseldorf, Germany
- EGA Institute for Women's Health, Zayed Centre for Research into Rare Diseases in Children (ZCR), University College London (UCL), 20 Guilford Street, London WC1N 1DZ, UK
| |
Collapse
|
13
|
DiKun KM, Gudas LJ. Vitamin A and retinoid signaling in the kidneys. Pharmacol Ther 2023; 248:108481. [PMID: 37331524 PMCID: PMC10528136 DOI: 10.1016/j.pharmthera.2023.108481] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Revised: 05/18/2023] [Accepted: 06/14/2023] [Indexed: 06/20/2023]
Abstract
Vitamin A (VA, retinol) and its metabolites (commonly called retinoids) are required for the proper development of the kidney during embryogenesis, but retinoids also play key roles in the function and repair of the kidney in adults. Kidneys filter 180-200 liters of blood per day and each kidney contains approximately 1 million nephrons, which are often referred to as the 'functional units' of the kidney. Each nephron consists of a glomerulus and a series of tubules (proximal tubule, loop of Henle, distal tubule, and collecting duct) surrounded by a network of capillaries. VA is stored in the liver and converted to active metabolites, most notably retinoic acid (RA), which acts as an agonist for the retinoic acid receptors ((RARs α, β, and γ) to regulate gene transcription. In this review we discuss some of the actions of retinoids in the kidney after injury. For example, in an ischemia-reperfusion model in mice, injury-associated loss of proximal tubule (PT) differentiation markers occurs, followed by re-expression of these differentiation markers during PT repair. Notably, healthy proximal tubules express ALDH1a2, the enzyme that metabolizes retinaldehyde to RA, but transiently lose ALDH1a2 expression after injury, while nearby myofibroblasts transiently acquire RA-producing capabilities after injury. These results indicate that RA is important for renal tubular injury repair and that compensatory mechanisms exist for the generation of endogenous RA by other cell types upon proximal tubule injury. ALDH1a2 levels also increase in podocytes, epithelial cells of the glomeruli, after injury, and RA promotes podocyte differentiation. We also review the ability of exogenous, pharmacological doses of RA and receptor selective retinoids to treat numerous kidney diseases, including kidney cancer and diabetic kidney disease, and the emerging genetic evidence for the importance of retinoids and their receptors in maintaining or restoring kidney function after injury. In general, RA has a protective effect on the kidney after various types of injuries (eg. ischemia, cytotoxic actions of chemicals, hyperglycemia related to diabetes). As more research into the actions of each of the three RARs in the kidney is carried out, a greater understanding of the actions of vitamin A is likely to lead to new insights into the pathology of kidney disorders and the development of new therapies for kidney diseases.
Collapse
Affiliation(s)
- Krysta M DiKun
- Department of Pharmacology, Weill Cornell Medical College of Cornell University, New York, NY, USA; New York Presbyterian Hospital, New York, NY, USA; Weill Cornell Graduate School of Medical Sciences, New York, NY, USA
| | - Lorraine J Gudas
- Department of Pharmacology, Weill Cornell Medical College of Cornell University, New York, NY, USA; Department of Urology, Weill Cornell Medicine, New York, NY, USA; New York Presbyterian Hospital, New York, NY, USA; Weill Cornell Graduate School of Medical Sciences, New York, NY, USA.
| |
Collapse
|
14
|
Dominguez JH, Xie D, Kelly KJ. Renal, but not platelet or skin, extracellular vesicles decrease oxidative stress, enhance nascent peptide synthesis, and protect from ischemic renal injury. Am J Physiol Renal Physiol 2023; 325:F164-F176. [PMID: 37318988 PMCID: PMC10393335 DOI: 10.1152/ajprenal.00321.2022] [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: 01/03/2023] [Revised: 06/08/2023] [Accepted: 06/09/2023] [Indexed: 06/17/2023] Open
Abstract
Acute kidney injury (AKI) is deadly and expensive, and specific, effective therapy remains a large unmet need. We have demonstrated the beneficial effects of transplanted adult tubular cells and extracellular vesicles (EVs; exosomes) derived from those renal cells on experimental ischemic AKI, even when administered after renal failure is established. To further examine the mechanisms of benefit with renal EVs, we tested the hypothesis that EVs from other epithelia or platelets (a rich source of EVs) might be protective, using a well-characterized ischemia-reperfusion model. When given after renal failure was present, renal EVs, but not those from skin or platelets, markedly improved renal function and histology. The differential effects allowed us to examine the mechanisms of benefit with renal EVs. We found significant decreases in oxidative stress postischemia in the renal EV-treated group with preservation of renal superoxide dismutase and catalase as well as increases in anti-inflammatory interleukin-10. In addition, we propose a novel mechanism of benefit: renal EVs enhanced nascent peptide synthesis following hypoxia in cells and in postischemic kidneys. Although EVs have been used therapeutically, these results serve as "proof of principle" to examine the mechanisms of injury and protection.NEW & NOTEWORTHY Acute kidney injury is common and deadly, yet the only approved treatment is dialysis. Thus, a better understanding of injury mechanisms and potential therapies is needed. We found that organ-specific, but not extrarenal, extracellular vesicles improved renal function and structure postischemia when given after renal failure occurred. Oxidative stress was decreased and anti-inflammatory interleukin-10 increased with renal, but not skin or platelet, exosomes. We also propose enhanced nascent peptide synthesis as a novel protective mechanism.
Collapse
Affiliation(s)
- Jesus H. Dominguez
- Division of Nephrology and Hypertension, Department of Medicine, Indiana University School of Medicine, Indianapolis, Indiana, United States
- Roudebush Veterans Affairs Medical Center, Indianapolis, Indiana, United States
| | - Danhui Xie
- Division of Nephrology and Hypertension, Department of Medicine, Indiana University School of Medicine, Indianapolis, Indiana, United States
| | - K. J. Kelly
- Division of Nephrology and Hypertension, Department of Medicine, Indiana University School of Medicine, Indianapolis, Indiana, United States
- Roudebush Veterans Affairs Medical Center, Indianapolis, Indiana, United States
| |
Collapse
|
15
|
Mo H, Yu H, Jiang Z, Chen Y, Yao M, Liu K, Li Y, Yu J, Wang L. Molecular cloning, tissues distribution, and function analysis of thioredoxin-like protein-1 (TXNL1) in Chinese giant salamanders Andrias davidianus. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2023; 144:104691. [PMID: 36967023 DOI: 10.1016/j.dci.2023.104691] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Revised: 02/23/2023] [Accepted: 03/22/2023] [Indexed: 06/05/2023]
Abstract
Thioredoxin-like protein-1 (TXNL1) is the member of thioredoxin superfamily, a family of thiol oxidoreductases. TXNL1 plays an important role in scavenging ROS and the maintenance of cellular redox balance. However, its physiological functions in Andrias davidianus have not been well understood. In the present study, the full-length cDNA encoding thioredoxin-like protein-1 (AdTXNL1) of A. davidianus was cloned, the mRNA tissue distribution was analyzed, and the function was characterized. The Adtxnl1 cDNA contained an open reading frame (ORF) of 870 bp encoding a polypeptide of 289 amino acids with the N-terminal TRX domain, a Cys34-Ala35-Pro36-Cys37 (CAPC) motif, and the C-terminal proteasome-interacting thioredoxin domain (PITH). The mRNA of AdTXNL1 was expressed in a wide range of tissues, with the highest level in the liver. The transcript level of AdTXNL1 was significantly up-regulated post Aeromonas hydrophila challenge in liver tissue. Moreover, the recombinant AdTXNL1 protein was produced and purified, and used to investigate the antioxidant activity. In the insulin disulfide reduction assay, rAdTXNL1 exhibited strong antioxidant capability. Altogether, the thioredoxin-like protein-1 may be involved in reduction/oxidation (redox) balance and as an important immunological gene in A. davidianus.
Collapse
Affiliation(s)
- Haolin Mo
- College of Animal Science and Technology, Northwest A&F University, Yangling, 712100, China
| | - Huixia Yu
- College of Animal Science and Technology, Northwest A&F University, Yangling, 712100, China
| | - Zebin Jiang
- College of Animal Science and Technology, Northwest A&F University, Yangling, 712100, China
| | - Yongqing Chen
- College of Animal Science and Technology, Northwest A&F University, Yangling, 712100, China
| | - Mingxing Yao
- College of Animal Science and Technology, Northwest A&F University, Yangling, 712100, China
| | - Kexin Liu
- College of Animal Science and Technology, Northwest A&F University, Yangling, 712100, China
| | - Yang Li
- College of Animal Science and Technology, Northwest A&F University, Yangling, 712100, China
| | - Jiajia Yu
- College of Animal Science and Technology, Northwest A&F University, Yangling, 712100, China
| | - Lixin Wang
- College of Animal Science and Technology, Northwest A&F University, Yangling, 712100, China.
| |
Collapse
|
16
|
Wali HA, Alabdulwahed MA, Al-Hussain GY, Alabbad MA, Al-Khalaf LH, Alhumaidi SS, Alhussain K, Alomair SM, Almulhim AS. Assessment of knowledge, attitude, and practices of acute kidney injury incidence with co-administration of piperacillin/tazobactam and vancomycin among healthcare workers: A cross-sectional study. Saudi Pharm J 2023; 31:904-910. [PMID: 37234343 PMCID: PMC10205774 DOI: 10.1016/j.jsps.2023.04.020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2023] [Accepted: 04/18/2023] [Indexed: 05/27/2023] Open
Abstract
Background No studies have identified a link between acute kidney injury (AKI) incidence due to the co-administration of vancomycin and piperacillin/tazobactam (VPT) and healthcare providers' knowledge, attitudes, and practices. We aimed to (1) assess the knowledge, attitudes, and practices towards AKI due to VPT co-administration among healthcare providers in Saudi Arabia, and (2) examine the relationship between healthcare providers' knowledge and attitudes about AKI due to VPT co-administration and their practices. Methods This cross-sectional study was conducted between February 2022 and April 2022. Healthcare providers, including physicians, pharmacists, and nurses, were included in the study population. The correlation coefficient assessed the relationship between knowledge, attitude, and practice. Spearman's rho was used as a test statistic. Results Of the invited healthcare providers, 192 responded to the survey. A significant difference in knowledge was found among healthcare providers for two variables: the definition of AKI (p < 0.001) and appropriate management of AKI due to VPT (p = 0.002). Physicians were found to rely less on the most common causative organisms of infection to guide empirical antibiotic therapy (p < 0.001). In addition, physicians were less likely to switch piperacillin/tazobactam to cefepime or meropenem in combination with vancomycin with AKI incidence (p = 0.001). A positive attitude towards the perceived AKI risk with VPT was positively correlated with avoiding using VPT unless no alternatives were available (Rho = 0.336) and taking protective measures when using VPT (Rho = 0.461). Conclusion Deviation has been observed in the knowledge, attitudes, and practices of AKI incidence with the co-administration of piperacillin/tazobactam and vancomycin among healthcare workers. Interventions at the organizational level are recommended to guide best practices.
Collapse
Affiliation(s)
- Haytham A. Wali
- Department of Pharmacy Practice, College of Clinical Pharmacy, King Faisal University, Al-Ahsa, Saudi Arabia
| | - Mohammed A. Alabdulwahed
- Department of Pharmacy Practice, College of Clinical Pharmacy, King Faisal University, Al-Ahsa, Saudi Arabia
| | - Ghaida Y. Al-Hussain
- Department of Pharmacy Practice, College of Clinical Pharmacy, King Faisal University, Al-Ahsa, Saudi Arabia
- Department of Pharmacy Services, Al-Ahsa Hospital, Al-Ahsa, Saudi Arabia
| | - Meaid A. Alabbad
- Department of Pharmacy Practice, College of Clinical Pharmacy, King Faisal University, Al-Ahsa, Saudi Arabia
| | - Lojain H. Al-Khalaf
- Department of Pharmacy Practice, College of Clinical Pharmacy, King Faisal University, Al-Ahsa, Saudi Arabia
| | - Sarah S. Alhumaidi
- Department of Pharmacy Practice, College of Clinical Pharmacy, King Faisal University, Al-Ahsa, Saudi Arabia
| | - Khalid Alhussain
- Department of Pharmacy Practice, College of Clinical Pharmacy, King Faisal University, Al-Ahsa, Saudi Arabia
| | - Sufyan M. Alomair
- Department of Pharmacy Practice, College of Clinical Pharmacy, King Faisal University, Al-Ahsa, Saudi Arabia
| | - Abdulaziz S. Almulhim
- Department of Pharmacy Practice, College of Clinical Pharmacy, King Faisal University, Al-Ahsa, Saudi Arabia
| |
Collapse
|
17
|
Huang Y, Chen K, Liu L, Ma H, Zhang X, Tan K, Li Y, Liu Y, Liu C, Wang H, Zhang XD. Single Atom-Engineered NIR-II Gold Clusters with Ultrahigh Brightness and Stability for Acute Kidney Injury. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2023:e2300145. [PMID: 37058089 DOI: 10.1002/smll.202300145] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/05/2023] [Revised: 02/21/2023] [Indexed: 06/19/2023]
Abstract
Near-infrared-II (NIR-II) imaging has shown great potential for monitoring the pathological progression and deep tissue imaging but is limited to present unmet NIR-II agent. Present fluorophores show a promising prospect for NIR-II imaging, but brightness and photostability are still highly challenging during real-time monitoring. In this work, atom-engineered NIR-II Au24 Cd1 clusters with ultrahigh brightness, stability, and photostability are developed via single atomic Cd doping. Single atom Cd substitutions contribute to Cd 4d state in HOMO and redistribution of energy level near the gap, exhibiting 56-fold fluorescence enhancement of Au24 Cd1 clusters. Meanwhile, single atomic Cd reinforces CdAu bond energy, formation energy, and stabilized cluster structure, leading to persistent stability for up to 1 month without decay, as well as excellent photostability of 1 h without photobleaching, much longer than clinically approved indocyanine green (<5 min). In vivo imaging shows gold clusters can monitor acute kidney injury (AKI) even after 72 h of injury, enabling evaluating progression at a very long window. Meanwhile, the bioactive gold clusters can alleviate AKI-induced oxidative stress damage and acute neuroinflammation. Single atom-engineered gold clusters exhibit molecular tracking and diagnostic prospect in kidney-related diseases.
Collapse
Affiliation(s)
- You Huang
- Tianjin Key Laboratory of Brain Science and Neural Engineering, Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin, 300072, China
| | - Ke Chen
- Tianjin Key Laboratory of Brain Science and Neural Engineering, Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin, 300072, China
| | - Ling Liu
- Tianjin Key Laboratory of Brain Science and Neural Engineering, Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin, 300072, China
| | - Huizhen Ma
- Department of Physics and Tianjin Key Laboratory of Low Dimensional Materials Physics and Preparing Technology, School of Sciences, Tianjin University, Tianjin, 300350, China
| | - Xiaoning Zhang
- Tianjin Key Laboratory of Brain Science and Neural Engineering, Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin, 300072, China
| | - Kexin Tan
- Tianjin Key Laboratory of Brain Science and Neural Engineering, Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin, 300072, China
| | - Yuan Li
- Department of Physics and Tianjin Key Laboratory of Low Dimensional Materials Physics and Preparing Technology, School of Sciences, Tianjin University, Tianjin, 300350, China
| | - Ying Liu
- Department of Physics and Tianjin Key Laboratory of Low Dimensional Materials Physics and Preparing Technology, School of Sciences, Tianjin University, Tianjin, 300350, China
| | - Changlong Liu
- Department of Physics and Tianjin Key Laboratory of Low Dimensional Materials Physics and Preparing Technology, School of Sciences, Tianjin University, Tianjin, 300350, China
| | - Hao Wang
- Tianjin Key Laboratory of Brain Science and Neural Engineering, Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin, 300072, China
| | - Xiao-Dong Zhang
- Tianjin Key Laboratory of Brain Science and Neural Engineering, Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin, 300072, China
| |
Collapse
|
18
|
Chen S, Wang Y, Liu Y, Bai L, Li F, Wu Y, Xie X, Zhang N, Zeng C, Zhang L, Wang X. Investigating the effect of dehydromiltirone on septic AKI using a network pharmacology method, molecular docking, and experimental validation. Front Pharmacol 2023; 14:1145675. [PMID: 37007048 PMCID: PMC10050741 DOI: 10.3389/fphar.2023.1145675] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2023] [Accepted: 02/27/2023] [Indexed: 03/17/2023] Open
Abstract
Acute kidney injury (AKI) is a severe and frequent complication of sepsis that occurs in intensive care units with inflammation and rapid decline in renal function as the main pathological features. Systemic inflammation, microvascular dysfunction, and tubule injury are the main causes of sepsis-induced AKI (SI-AKI). The high prevalence and death rate from SI-AKI is a great challenge for clinical treatment worldwide. However, in addition to hemodialysis, there is no effective drug to improve renal tissue damage and alleviate the decline in kidney function. We conducted a network pharmacological analysis of Salvia miltiorrhiza (SM), a traditional Chinese medicine, which is widely used for the treatment of kidney disease. Then, we combined molecular docking and a dynamics simulation to screen for the active monomer dehydromiltirone (DHT) that has therapeutic effects on SI-AKI and investigated its potential mechanism of action through experimental validation. The components and targets of SM were obtained by searching the database, and 32 overlapping genes were screened by intersection analysis with AKI targets. GO and KEGG data showed that the functions of a common gene were closely related to oxidative stress, mitochondrial function, and apoptosis. The molecular docking results combined with molecular dynamics simulations provide evidence for a binding model between DHT and cyclooxygenase-2 (COX2), both of which are mainly driven by van der Waals interactions and a hydrophobic effect. In vivo, we found that mice pretreated with an intraperitoneal injection of DHT (20 mg/kg/d) for 3 days ameliorated CLP surgery-induced renal function loss and renal tissue damage and inhibited inflammatory mediators IL-6, IL-1β, TNF-α, and MCP-1 production. In vitro, the DHT pretreatment decreased LPS-induced expression of COX2, inhibited cell death and oxidative stress, alleviated mitochondrial dysfunction, and restrained apoptosis in HK-2 cells. Our research indicates that the renal preventive effect of DHT is related to maintaining mitochondrial dynamic balance, restoring mitochondrial oxidative phosphorylation, and inhibiting cell apoptosis. The findings in this study provide a theoretical basis and a novel method for the clinical therapy of SI-AKI.
Collapse
Affiliation(s)
- Sijia Chen
- Department of Nephrology, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yanzhe Wang
- Department of Nephrology, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yuyuan Liu
- Department of Nephrology, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Suzhou, Jiangsu, China
| | - Linnan Bai
- Department of Nephrology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Fengqin Li
- Department of Nephrology, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yue Wu
- Department of Nephrology, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xinmiao Xie
- Department of Nephrology, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Nan Zhang
- Department of Nephrology, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Chuchu Zeng
- Department of Nephrology, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ling Zhang
- Department of Obstetrics and Gynecology, Chengdu Women’s and Children’s Central Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan, China
- *Correspondence: Xiaoxia Wang, , Ling Zhang,
| | - Xiaoxia Wang
- Department of Nephrology, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- *Correspondence: Xiaoxia Wang, , Ling Zhang,
| |
Collapse
|
19
|
Chang SN, Park JG, Kang SC. Therapeutic propensity of ginsenosides Rg1 and Rg3 in rhabdomyolysis-induced acute kidney injury and renohepatic crosstalk in rats. Int Immunopharmacol 2023; 115:109602. [PMID: 36580761 DOI: 10.1016/j.intimp.2022.109602] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2022] [Revised: 12/10/2022] [Accepted: 12/12/2022] [Indexed: 12/28/2022]
Abstract
BACKGROUND Ginseng is a traditional herbal medicine used for thousands of years in Southeast Asian countries because of its medicinal properties. Ginsenosides Rg1 and Rg3 have demonstrated therapeutic properties against a broad spectrum of diseases. PURPOSE Here in this study, we investigated the therapeutic efficacy of Rg1 and Rg3 in alleviating glycerol-induced acute kidney injury, also known as rhabdomyolysis-induced acute kidney injury (RAKI). METHODS AKI was induced in male Wistar rats through intramuscular injection of 10 mL/kg glycerol and simultaneous oral treatment of ginsenosides Rg1 and Rg3 for 3 days. We also evaluated the therapeutic potential of Rg1 and Rg3 on human embryonic kidney epithelial (HEK-293). Cell viability and LDH assay were performed on HEK-293 cells to evaluate the toxicity of Rg1 and Rg3. Evaluation of important kidney damage markers such as creatinine and blood urea nitrogen (BUN) was carried out at different time points from the rat serum. Histopathological analysis was performed on kidney tissues. We also performed experiments such as ELISA assay, immunohistochemistry, immunofluorescence staining, COMET assay, western blotting, TUNEL assay, and flow cytometry to obtain results. RESULTS Rg1 and Rg3 significantly downregulated the expression of kidney damage markers such as creatinine and BUN in a dose-dependent manner. Histopathological analysis revealed damage across the glomerulus, tubules, and collecting duct rendering the kidney dysfunctional in glycerol treatment groups. However, Rg1 and Rg3 treated groups showed a significant reduction in tubular necrosis at both 10 and 20 mg/kg. There was also a sharp downregulation of oxidative and ER stress markers. Additionally, we observed nuclear translocation of Nrf2 which were more prominent in kidney tissues. Rg1 and Rg3 were also able to mitigate apoptotic cell death in vitro and in vivo evaluated through immunofluorescence staining for p53, TUNEL assay, flow cytometry, and immunoblotting for intrinsic apoptosis markers. CONCLUSION In summary, we conclude that Rg1 and Rg3 exhibited natural therapeutic remedy against AKI.
Collapse
Affiliation(s)
- Sukkum Ngullie Chang
- Department of Biotechnology, Daegu University, Gyeongsan, Gyeongbuk 38453, Republic of Korea.
| | - Jae Gyu Park
- Advanced Bio Convergence Center (ABCC), Pohang Technopark Foundation, Pohang 37668, Republic of Korea.
| | - Sun Chul Kang
- Department of Biotechnology, Daegu University, Gyeongsan, Gyeongbuk 38453, Republic of Korea.
| |
Collapse
|
20
|
Punica granatum L. Polyphenolic Extract as an Antioxidant to Prevent Kidney Injury in Metabolic Syndrome Rats. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2023; 2023:6144967. [PMID: 36644578 PMCID: PMC9836814 DOI: 10.1155/2023/6144967] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/19/2022] [Revised: 11/21/2022] [Accepted: 11/28/2022] [Indexed: 01/07/2023]
Abstract
Introduction Obesity and metabolic syndrome (MetS) constitute a rapidly increasing health problem and contribute to the development of multiple comorbidities like acute and chronic kidney disease. Insulin resistance, inappropriate lipolysis, and excess of free fatty acids (FFAs) are associated with glomerulus hyperfiltration and atherosclerosis. The important component of MetS, oxidative stress, is also involved in the destabilization of kidney function and the progression of kidney injury. Natural polyphenols have the ability to reduce the harmful effect of reactive oxygen and nitrogen species (ROS/RNS). Extract derived from Punica granatum L. is rich in punicalagin that demonstrates positive effects in MetS and its associated diseases. The aim of the study was to investigate the effect of bioactive substances of pomegranate peel to kidney damage associated with the MetS. Methods In this study, we compared biomarkers of oxidative stress in kidney tissue of adult male Zucker Diabetic Fatty (ZDF) rats with MetS and healthy controls that were treated with Punica granatum L. extract at a dose of 100 or 200 mg/kg. Additionally, we evaluated the effect of polyphenolic extract on kidney injury markers and remodeling. The concentration of ROS/RNS, oxLDL, glutathione (GSH), kidney injury molecule-1 (KIM-1), neutrophil gelatinase-associated lipocalin (NGAL), metalloproteinase 2 and 9 (MMP-2, MMP-9), and the activity of superoxide dismutase (SOD) and catalase (CAT) were measured. Results The data showed significant differences in oxidative stress markers between treated and untreated MetS rats. ROS/RNS levels, oxLDL concentration, and SOD activity were lower, whereas CAT activity was higher in rats with MetS receiving polyphenolic extract. After administration of the extract, markers for kidney injury (NGAL, KIM-1) decreased. Conclusion Our study confirmed the usefulness of pomegranate polyphenols in the treatment of MetS and the prevention of kidney damage. However, further, more detailed research is required to establish the mechanism of polyphenol protection.
Collapse
|
21
|
Zhu L, Luo M, Zhang Y, Fang F, Li M, An F, Zhao D, Zhang J. Free radical as a double-edged sword in disease: Deriving strategic opportunities for nanotherapeutics. Coord Chem Rev 2023. [DOI: 10.1016/j.ccr.2022.214875] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
|
22
|
Gadekar GJ, Bhandare PA, Bandawane DD. Amelioration of 5-Fluorouracil Induced Nephrotoxicity by Acacia catechu through Overcoming Oxidative Damage and Inflammation in Wistar Rats. Cardiovasc Hematol Disord Drug Targets 2023; 23:189-201. [PMID: 37946347 DOI: 10.2174/011871529x274030231102065433] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2023] [Revised: 10/03/2023] [Accepted: 10/13/2023] [Indexed: 11/12/2023]
Abstract
AIM The research intended to explore the possible nephroprotective potential of the ethyl acetate fraction derived from Acacia catechu leaves against nephrotoxicity brought about by 5-fluorouracil (5-FU) in Wistar rats. BACKGROUND While possessing strong anticancer properties, 5-FU is hindered in its therapeutic application due to significant organ toxicity linked to elevated oxidative stress and inflammation. OBJECTIVE The study is undertaken to conduct an analysis of the ethyl acetate fraction of A. catechu leaves both in terms of quality and quantity, examining its impact on different biochemical and histopathological parameters within the context of 5-FU-induced renal damage in rats and elucidation of the mechanism behind the observed outcomes. METHODOLOGY Intraperitoneal injection of 5-FU at a dosage of 20 mg/kg/day over 5 days was given to induce nephrotoxicity in rats. The evaluation of nephrotoxicity involved quantifying serum creatinine, urea, uric acid, and electrolyte concentrations. Furthermore, superoxide dismutase, catalase antioxidant enzymes, and TNF-α concentration in serum were also measured. RESULTS 5-FU injection led to the initiation of oxidative stress within the kidneys, leading to modifications in renal biomarkers (including serum creatinine, urea, uric acid, and Na+, K+ levels), and a reduction in antioxidant enzymes namely superoxide dismutase and catalase. Notably, the presence of the inflammatory cytokine TNF-α was significantly elevated due to 5-FU. Microscopic examination of renal tissue revealed tubular degeneration and congestion. However, treatment involving the ethyl acetate fraction derived from A. catechu leaves effectively and dose-dependently reversed the changes observed in renal biomarkers, renal antioxidant enzymes, inflammatory mediators, and histopathological features, bringing them closer to normal conditions. The observed recuperative impact was mainly attributed to the antioxidant and antiinflammatory properties of the fraction. CONCLUSION The ethyl acetate fraction of A. catechu leaves exhibited a mitigating influence on the renal impairment caused by 5-FU, showcasing its potential as a nephroprotective agent capable of preventing and ameliorating 5-FU-induced nephrotoxicity.
Collapse
Affiliation(s)
- Gayatri Jaising Gadekar
- Department of Pharmacology, P. E. Society's Modern College of Pharmacy, Nigdi, Pune- 44, India
| | | | - Deepti Dinesh Bandawane
- Department of Pharmacology, P. E. Society's Modern College of Pharmacy, Nigdi, Pune- 44, India
| |
Collapse
|
23
|
A Hydrodistillate of Gynostemma pentaphyllum and Damulin B Prevent Cisplatin-Induced Nephrotoxicity In Vitro and In Vivo via Regulation of AMPKα1 Transcription. Nutrients 2022; 14:nu14234997. [PMID: 36501027 PMCID: PMC9737728 DOI: 10.3390/nu14234997] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2022] [Revised: 11/21/2022] [Accepted: 11/22/2022] [Indexed: 11/26/2022] Open
Abstract
The clinical application of cisplatin, one of the most effective chemotherapeutic agents used to treat various cancers, has been limited by the risk of adverse effects, notably nephrotoxicity. Despite intensive research for decades, there are no effective approaches for alleviating cisplatin nephrotoxicity. This study aimed to investigate the protective effects and potential mechanisms of a Gynostemma pentaphyllum leaves hydrodistillate (GPHD) and its major component, damulin B, against cisplatin-induced nephrotoxicity in vitro and in vivo. A hydro-distillation method can extract large amounts of components within a short period of time using non-toxic, environmentally friendly solvent. We found that the levels of AMP-activated protein kinase α1 (AMPKα1), reactive oxygen species (ROS), and apoptosis were tightly associated with each other in HEK293 cells treated with cisplatin. We demonstrated that AMPKα1 acted as an anti-oxidant factor and that ROS generated by cisplatin suppressed the expression of AMPKα1 at the transcriptional level, thereby resulting in induction of apoptosis. Treatment with GPHD or damulin B effectively prevented cisplatin-induced apoptosis of HEK293 cells and cisplatin-induced acute kidney injury in mice by suppressing oxidative stress and maintaining AMPKα1 levels. Therefore, our study suggests that GPHD and damulin B may serve as prospective adjuvant agents against cisplatin-induced nephrotoxicity.
Collapse
|
24
|
Nath KA, Singh RD, Croatt AJ, Adams CM. Heme Proteins and Kidney Injury: Beyond Rhabdomyolysis. KIDNEY360 2022; 3:1969-1979. [PMID: 36514409 PMCID: PMC9717624 DOI: 10.34067/kid.0005442022] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/16/2022] [Accepted: 09/23/2022] [Indexed: 11/07/2022]
Abstract
Heme proteins, the stuff of life, represent an ingenious biologic strategy that capitalizes on the biochemical versatility of heme, and yet is one that avoids the inherent risks to cellular vitality posed by unfettered and promiscuously reactive heme. Heme proteins, however, may be a double-edged sword because they can damage the kidney in certain settings. Although such injury is often viewed mainly within the context of rhabdomyolysis and the nephrotoxicity of myoglobin, an increasing literature now attests to the fact that involvement of heme proteins in renal injury ranges well beyond the confines of this single disease (and its analog, hemolysis); indeed, through the release of the defining heme motif, destabilization of intracellular heme proteins may be a common pathway for acute kidney injury, in general, and irrespective of the underlying insult. This brief review outlines current understanding regarding processes underlying such heme protein-induced acute kidney injury (AKI) and chronic kidney disease (CKD). Topics covered include, among others, the basis for renal injury after the exposure of the kidney to and its incorporation of myoglobin and hemoglobin; auto-oxidation of myoglobin and hemoglobin; destabilization of heme proteins and the release of heme; heme/iron/oxidant pathways of renal injury; generation of reactive oxygen species and reactive nitrogen species by NOX, iNOS, and myeloperoxidase; and the role of circulating cell-free hemoglobin in AKI and CKD. Also covered are the characteristics of the kidney that render this organ uniquely vulnerable to injury after myolysis and hemolysis, and pathobiologic effects emanating from free, labile heme. Mechanisms that defend against the toxicity of heme proteins are discussed, and the review concludes by outlining the therapeutic strategies that have arisen from current understanding of mechanisms of renal injury caused by heme proteins and how such mechanisms may be interrupted.
Collapse
Affiliation(s)
- Karl A. Nath
- Division of Nephrology and Hypertension, Department of Medicine, Mayo Clinic, Rochester, Minnesota
| | - Raman Deep Singh
- Division of Nephrology and Hypertension, Department of Medicine, Mayo Clinic, Rochester, Minnesota
| | - Anthony J. Croatt
- Division of Nephrology and Hypertension, Department of Medicine, Mayo Clinic, Rochester, Minnesota
| | - Christopher M. Adams
- Division of Endocrinology, Metabolism and Nutrition, Department of Medicine, Mayo Clinic Rochester, Minnesota
| |
Collapse
|
25
|
Chen Z, Qi F, Qiu W, Wu C, Zong M, Ge M, Xu D, You Y, Zhu Y, Zhang Z, Lin H, Shi J. Hydrogenated Germanene Nanosheets as an Antioxidative Defense Agent for Acute Kidney Injury Treatment. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2022; 9:e2202933. [PMID: 36202760 PMCID: PMC9685437 DOI: 10.1002/advs.202202933] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Revised: 08/12/2022] [Indexed: 06/16/2023]
Abstract
Acute kidney injury (AKI) is a sudden kidney dysfunction caused by aberrant reactive oxygen species (ROS) metabolism that results in high clinical mortality. The rapid development of ROS scavengers provides new opportunities for AKI treatment. Herein, the use of hydrogen-terminated germanene (H-germanene) nanosheets is reported as an antioxidative defense nanoplatform against AKI in mice. The simulation results show that 2D H-germanene can effectively scavenge ROS through free radical adsorption and subsequent redox reactions. In particular, the H-germanene exhibits high accumulation in injured kidneys, thereby offering a favorable opportunity for treating renal diseases. In the glycerol-induced murine AKI model, H-germanene delivers robust antioxidative protection against ROS attack to maintain normal kidney function indicators without negative influence in vivo. This positive in vivo antioxidative defense in living animals demonstrates that the present H-germanene nanoplatform is a powerful antioxidant against AKI and various anti-inflammatory diseases.
Collapse
Affiliation(s)
- Zhixin Chen
- State Key Laboratory of High Performance Ceramics and Superfine MicrostructuresShanghai Institute of CeramicsChinese Academy of SciencesResearch Unit of Nanocatalytic Medicine in Specific Therapy for Serious DiseaseChinese Academy of Medical Sciences (2021RU012)Shanghai200050P. R. China
- Center of Materials Science and Optoelectronics EngineeringUniversity of Chinese Academy of SciencesBeijing100049P. R. China
| | - Fenggang Qi
- State Key Laboratory of High Performance Ceramics and Superfine MicrostructuresShanghai Institute of CeramicsChinese Academy of SciencesResearch Unit of Nanocatalytic Medicine in Specific Therapy for Serious DiseaseChinese Academy of Medical Sciences (2021RU012)Shanghai200050P. R. China
- Center of Materials Science and Optoelectronics EngineeringUniversity of Chinese Academy of SciencesBeijing100049P. R. China
| | - Wujie Qiu
- State Key Laboratory of High Performance Ceramics and Superfine MicrostructuresShanghai Institute of CeramicsChinese Academy of SciencesResearch Unit of Nanocatalytic Medicine in Specific Therapy for Serious DiseaseChinese Academy of Medical Sciences (2021RU012)Shanghai200050P. R. China
| | - Chenyao Wu
- State Key Laboratory of High Performance Ceramics and Superfine MicrostructuresShanghai Institute of CeramicsChinese Academy of SciencesResearch Unit of Nanocatalytic Medicine in Specific Therapy for Serious DiseaseChinese Academy of Medical Sciences (2021RU012)Shanghai200050P. R. China
- Center of Materials Science and Optoelectronics EngineeringUniversity of Chinese Academy of SciencesBeijing100049P. R. China
| | - Ming Zong
- Department of Clinical LaboratoryShanghai East HospitalTongji University School of MedicineShanghai200120P. R. China
| | - Min Ge
- State Key Laboratory of High Performance Ceramics and Superfine MicrostructuresShanghai Institute of CeramicsChinese Academy of SciencesResearch Unit of Nanocatalytic Medicine in Specific Therapy for Serious DiseaseChinese Academy of Medical Sciences (2021RU012)Shanghai200050P. R. China
- Center of Materials Science and Optoelectronics EngineeringUniversity of Chinese Academy of SciencesBeijing100049P. R. China
| | - Deliang Xu
- State Key Laboratory of High Performance Ceramics and Superfine MicrostructuresShanghai Institute of CeramicsChinese Academy of SciencesResearch Unit of Nanocatalytic Medicine in Specific Therapy for Serious DiseaseChinese Academy of Medical Sciences (2021RU012)Shanghai200050P. R. China
- Center of Materials Science and Optoelectronics EngineeringUniversity of Chinese Academy of SciencesBeijing100049P. R. China
| | - Yanling You
- State Key Laboratory of High Performance Ceramics and Superfine MicrostructuresShanghai Institute of CeramicsChinese Academy of SciencesResearch Unit of Nanocatalytic Medicine in Specific Therapy for Serious DiseaseChinese Academy of Medical Sciences (2021RU012)Shanghai200050P. R. China
- Center of Materials Science and Optoelectronics EngineeringUniversity of Chinese Academy of SciencesBeijing100049P. R. China
| | - Ya‐Xuan Zhu
- State Key Laboratory of High Performance Ceramics and Superfine MicrostructuresShanghai Institute of CeramicsChinese Academy of SciencesResearch Unit of Nanocatalytic Medicine in Specific Therapy for Serious DiseaseChinese Academy of Medical Sciences (2021RU012)Shanghai200050P. R. China
- Shanghai Tenth People's HospitalShanghai Frontiers Science Center of Nanocatalytic MedicineSchool of Medicine Tongji UniversityShanghai200331P. R. China
| | - Zhimin Zhang
- State Key Laboratory of High Performance Ceramics and Superfine MicrostructuresShanghai Institute of CeramicsChinese Academy of SciencesResearch Unit of Nanocatalytic Medicine in Specific Therapy for Serious DiseaseChinese Academy of Medical Sciences (2021RU012)Shanghai200050P. R. China
- Center of Materials Science and Optoelectronics EngineeringUniversity of Chinese Academy of SciencesBeijing100049P. R. China
| | - Han Lin
- State Key Laboratory of High Performance Ceramics and Superfine MicrostructuresShanghai Institute of CeramicsChinese Academy of SciencesResearch Unit of Nanocatalytic Medicine in Specific Therapy for Serious DiseaseChinese Academy of Medical Sciences (2021RU012)Shanghai200050P. R. China
- Shanghai Tenth People's HospitalShanghai Frontiers Science Center of Nanocatalytic MedicineSchool of Medicine Tongji UniversityShanghai200331P. R. China
| | - Jianlin Shi
- State Key Laboratory of High Performance Ceramics and Superfine MicrostructuresShanghai Institute of CeramicsChinese Academy of SciencesResearch Unit of Nanocatalytic Medicine in Specific Therapy for Serious DiseaseChinese Academy of Medical Sciences (2021RU012)Shanghai200050P. R. China
- Center of Materials Science and Optoelectronics EngineeringUniversity of Chinese Academy of SciencesBeijing100049P. R. China
- Shanghai Tenth People's HospitalShanghai Frontiers Science Center of Nanocatalytic MedicineSchool of Medicine Tongji UniversityShanghai200331P. R. China
| |
Collapse
|
26
|
Chen Z, Qi F, Qiu W, Wu C, Zong M, Ge M, Xu D, You Y, Zhu Y, Zhang Z, Lin H, Shi J. Hydrogenated Germanene Nanosheets as an Antioxidative Defense Agent for Acute Kidney Injury Treatment. ADVANCED SCIENCE 2022; 9. [DOI: doi.org/10.1002/advs.202202933] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Indexed: 09/08/2023]
Abstract
AbstractAcute kidney injury (AKI) is a sudden kidney dysfunction caused by aberrant reactive oxygen species (ROS) metabolism that results in high clinical mortality. The rapid development of ROS scavengers provides new opportunities for AKI treatment. Herein, the use of hydrogen‐terminated germanene (H‐germanene) nanosheets is reported as an antioxidative defense nanoplatform against AKI in mice. The simulation results show that 2D H‐germanene can effectively scavenge ROS through free radical adsorption and subsequent redox reactions. In particular, the H‐germanene exhibits high accumulation in injured kidneys, thereby offering a favorable opportunity for treating renal diseases. In the glycerol‐induced murine AKI model, H‐germanene delivers robust antioxidative protection against ROS attack to maintain normal kidney function indicators without negative influence in vivo. This positive in vivo antioxidative defense in living animals demonstrates that the present H‐germanene nanoplatform is a powerful antioxidant against AKI and various anti‐inflammatory diseases.
Collapse
Affiliation(s)
- Zhixin Chen
- State Key Laboratory of High Performance Ceramics and Superfine Microstructures Shanghai Institute of Ceramics Chinese Academy of Sciences Research Unit of Nanocatalytic Medicine in Specific Therapy for Serious Disease Chinese Academy of Medical Sciences (2021RU012) Shanghai 200050 P. R. China
- Center of Materials Science and Optoelectronics Engineering University of Chinese Academy of Sciences Beijing 100049 P. R. China
| | - Fenggang Qi
- State Key Laboratory of High Performance Ceramics and Superfine Microstructures Shanghai Institute of Ceramics Chinese Academy of Sciences Research Unit of Nanocatalytic Medicine in Specific Therapy for Serious Disease Chinese Academy of Medical Sciences (2021RU012) Shanghai 200050 P. R. China
- Center of Materials Science and Optoelectronics Engineering University of Chinese Academy of Sciences Beijing 100049 P. R. China
| | - Wujie Qiu
- State Key Laboratory of High Performance Ceramics and Superfine Microstructures Shanghai Institute of Ceramics Chinese Academy of Sciences Research Unit of Nanocatalytic Medicine in Specific Therapy for Serious Disease Chinese Academy of Medical Sciences (2021RU012) Shanghai 200050 P. R. China
| | - Chenyao Wu
- State Key Laboratory of High Performance Ceramics and Superfine Microstructures Shanghai Institute of Ceramics Chinese Academy of Sciences Research Unit of Nanocatalytic Medicine in Specific Therapy for Serious Disease Chinese Academy of Medical Sciences (2021RU012) Shanghai 200050 P. R. China
- Center of Materials Science and Optoelectronics Engineering University of Chinese Academy of Sciences Beijing 100049 P. R. China
| | - Ming Zong
- Department of Clinical Laboratory Shanghai East Hospital Tongji University School of Medicine Shanghai 200120 P. R. China
| | - Min Ge
- State Key Laboratory of High Performance Ceramics and Superfine Microstructures Shanghai Institute of Ceramics Chinese Academy of Sciences Research Unit of Nanocatalytic Medicine in Specific Therapy for Serious Disease Chinese Academy of Medical Sciences (2021RU012) Shanghai 200050 P. R. China
- Center of Materials Science and Optoelectronics Engineering University of Chinese Academy of Sciences Beijing 100049 P. R. China
| | - Deliang Xu
- State Key Laboratory of High Performance Ceramics and Superfine Microstructures Shanghai Institute of Ceramics Chinese Academy of Sciences Research Unit of Nanocatalytic Medicine in Specific Therapy for Serious Disease Chinese Academy of Medical Sciences (2021RU012) Shanghai 200050 P. R. China
- Center of Materials Science and Optoelectronics Engineering University of Chinese Academy of Sciences Beijing 100049 P. R. China
| | - Yanling You
- State Key Laboratory of High Performance Ceramics and Superfine Microstructures Shanghai Institute of Ceramics Chinese Academy of Sciences Research Unit of Nanocatalytic Medicine in Specific Therapy for Serious Disease Chinese Academy of Medical Sciences (2021RU012) Shanghai 200050 P. R. China
- Center of Materials Science and Optoelectronics Engineering University of Chinese Academy of Sciences Beijing 100049 P. R. China
| | - Ya‐Xuan Zhu
- State Key Laboratory of High Performance Ceramics and Superfine Microstructures Shanghai Institute of Ceramics Chinese Academy of Sciences Research Unit of Nanocatalytic Medicine in Specific Therapy for Serious Disease Chinese Academy of Medical Sciences (2021RU012) Shanghai 200050 P. R. China
- Shanghai Tenth People's Hospital Shanghai Frontiers Science Center of Nanocatalytic Medicine School of Medicine Tongji University Shanghai 200331 P. R. China
| | - Zhimin Zhang
- State Key Laboratory of High Performance Ceramics and Superfine Microstructures Shanghai Institute of Ceramics Chinese Academy of Sciences Research Unit of Nanocatalytic Medicine in Specific Therapy for Serious Disease Chinese Academy of Medical Sciences (2021RU012) Shanghai 200050 P. R. China
- Center of Materials Science and Optoelectronics Engineering University of Chinese Academy of Sciences Beijing 100049 P. R. China
| | - Han Lin
- State Key Laboratory of High Performance Ceramics and Superfine Microstructures Shanghai Institute of Ceramics Chinese Academy of Sciences Research Unit of Nanocatalytic Medicine in Specific Therapy for Serious Disease Chinese Academy of Medical Sciences (2021RU012) Shanghai 200050 P. R. China
- Shanghai Tenth People's Hospital Shanghai Frontiers Science Center of Nanocatalytic Medicine School of Medicine Tongji University Shanghai 200331 P. R. China
| | - Jianlin Shi
- State Key Laboratory of High Performance Ceramics and Superfine Microstructures Shanghai Institute of Ceramics Chinese Academy of Sciences Research Unit of Nanocatalytic Medicine in Specific Therapy for Serious Disease Chinese Academy of Medical Sciences (2021RU012) Shanghai 200050 P. R. China
- Center of Materials Science and Optoelectronics Engineering University of Chinese Academy of Sciences Beijing 100049 P. R. China
- Shanghai Tenth People's Hospital Shanghai Frontiers Science Center of Nanocatalytic Medicine School of Medicine Tongji University Shanghai 200331 P. R. China
| |
Collapse
|
27
|
Singh RD, Croatt AJ, Ackerman AW, Grande JP, Trushina E, Salisbury JL, Christensen TA, Adams CM, Tchkonia T, Kirkland JL, Nath KA. Prominent Mitochondrial Injury as an Early Event in Heme Protein-Induced Acute Kidney Injury. KIDNEY360 2022; 3:1672-1682. [PMID: 36514726 PMCID: PMC9717657 DOI: 10.34067/kid.0004832022] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/20/2022] [Accepted: 08/15/2022] [Indexed: 01/12/2023]
Abstract
Background Mitochondrial injury occurs in and underlies acute kidney injury (AKI) caused by ischemia-reperfusion and other forms of renal injury. However, to date, a comprehensive analysis of this issue has not been undertaken in heme protein-induced AKI (HP-AKI). We examined key aspects of mitochondrial function, expression of proteins relevant to mitochondrial quality control, and mitochondrial ultrastructure in HP-AKI, along with responses to heme in renal proximal tubule epithelial cells. Methods The long-established murine glycerol model of HP-AKI was examined at 8 and 24 hours after HP-AKI. Indices of mitochondrial function (ATP and NAD+), expression of proteins relevant to mitochondrial dynamics, mitochondrial ultrastructure, and relevant gene/protein expression in heme-exposed renal proximal tubule epithelial cells in vitro were examined. Results ATP and NAD+ content and the NAD+/NADH ratio were all reduced in HP-AKI. Expression of relevant proteins indicate that mitochondrial biogenesis (PGC-1α, NRF1, and TFAM) and fusion (MFN2) were impaired, as was expression of key proteins involved in the integrity of outer and inner mitochondrial membranes (VDAC, Tom20, and Tim23). Conversely, marked upregulation of proteins involved in mitochondrial fission (DRP1) occurred. Ultrastructural studies, including novel 3D imaging, indicate profound changes in mitochondrial structure, including mitochondrial fragmentation, mitochondrial swelling, and misshapen mitochondrial cristae; mitophagy was also observed. Exposure of renal proximal tubule epithelial cells to heme in vitro recapitulated suppression of PGC-1α (mitochondrial biogenesis) and upregulation of p-DRP1 (mitochondrial fission). Conclusions Modern concepts pertaining to AKI apply to HP-AKI. This study validates the investigation of novel, clinically relevant therapies such as NAD+-boosting agents and mitoprotective agents in HP-AKI.
Collapse
Affiliation(s)
- Raman Deep Singh
- Division of Nephrology and Hypertension, Department of Medicine, Mayo Clinic Rochester, Minnesota
| | - Anthony J. Croatt
- Division of Nephrology and Hypertension, Department of Medicine, Mayo Clinic Rochester, Minnesota
| | - Allan W. Ackerman
- Division of Nephrology and Hypertension, Department of Medicine, Mayo Clinic Rochester, Minnesota
| | - Joseph P. Grande
- Department of Laboratory Medicine and Pathology, Mayo Clinic Rochester, Minnesota
| | - Eugenia Trushina
- Department of Neurology, Mayo Clinic Rochester, Minnesota
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic Rochester, Minnesota
| | - Jeffrey L. Salisbury
- Microscopy and Cell Analysis Core Facility, Mayo Clinic Rochester, Minnesota
- Department of Biochemistry and Molecular Biology, Mayo Clinic Rochester, Minnesota
| | | | - Christopher M. Adams
- Division of Endocrinology, Diabetes, Metabolism and Nutrition, Department of Medicine, Mayo Clinic Rochester, Minnesota
| | - Tamara Tchkonia
- Department of Physiology and Biomedical Engineering, Mayo Clinic Rochester, Minnesota
| | - James L. Kirkland
- Department of Physiology and Biomedical Engineering, Mayo Clinic Rochester, Minnesota
- Department of General Internal Medicine, Department of Medicine, Mayo Clinic Rochester, Minnesota
| | - Karl A. Nath
- Division of Nephrology and Hypertension, Department of Medicine, Mayo Clinic Rochester, Minnesota
| |
Collapse
|
28
|
Ouattar H, Zouirech O, Kara M, Assouguem A, Almutairi SM, Al-Hemaid FM, Rasheed RA, Ullah R, Abbasi AM, Aouane M, Mikou K. In Vitro Study of the Phytochemical Composition and Antioxidant, Immunostimulant, and Hemolytic Activities of Nigella sativa (Ranunculaceae) and Lepidium sativum Seeds. Molecules 2022; 27:5946. [PMID: 36144678 PMCID: PMC9505328 DOI: 10.3390/molecules27185946] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2022] [Revised: 09/03/2022] [Accepted: 09/07/2022] [Indexed: 11/16/2022] Open
Abstract
The Moroccan flora abounds and is an important reserve of medicinal plants. Nigella sativa and Lepidium sativum are plants that are widely used in traditional medicine for their multiple therapeutic properties. The current study aims to highlight the biological activities that can justify and valorize the use of these plants. Flavonoids, total phenols, condensed tannins, and sugars were determined. The biological activities tested were antioxidant by determining the IC50 (defined as the concentration of an antioxidant required to decrease the initial concentration by 50%; inversely related to the antioxidant capacity), hemagglutination, and hemolytic activities. Phytochemical quantification of the seed extracts indicated that the total phenol content was largely similar for both plants and in the order of 10 mg GAE (Gallic acid equivalent)/g. On the other hand, L. sativum seeds registered a higher content of flavonoids (3.09 ± 0.04 mg QE (quercetin equivalent)/g) as compared to Nigella saliva (0.258 ± 0.058). Concerning condensed tannins, N. saliva seeds present a higher amount with a value of 7.2 ± 0.025 mg/g as compared to L. sativum (1.4 ± 0.22 mg/g). Concerning the total sugar content, L. sativum shows a higher content (67.86 ± 0.87 mg/g) as compared to N. sativa (58.17 ± 0.42 mg/g); it is also richer in mucilage with a content of 240 mg as compared to 8.2 mg for N. saliva. Examination of the antioxidant activity using a DPPH (2.2-diphenyl 1-pycrilhydrazyl) test revealed that the EButOH (n-butanol extract) and EAE (ethyl acetate extract) extracts were the most active, with IC50 values of 48.7 and 50.65 μg/mL for the N. sativa extracts and 15.7 and 52.64 μg/mL for the L. sativum extracts, respectively. The results of the hemagglutination activity of the different extracts of the two plants prepared in the PBS (phosphate-buffered saline) medium showed significant agglutination for the L. sativum extract (1/50) compared to the N. sativa extract (1/20). An evaluation of the hemolytic effect of the crude extract of the studied seeds on erythrocytes isolated from rat blood incubated in PBS buffer compared to the total hemolysis induced by distilled water showed a hemolysis rate of 54% for Nigella sativa and 34% for L. sativum. In conclusion, the two plants studied in the current work exhibited high antioxidant potential, which could explain their beneficial properties.
Collapse
Affiliation(s)
- Hafssa Ouattar
- Laboratory of Natural Resources and Sustainable Development, Faculty of Sciences, University of Ibn Tofail, P.O. Box 133, Kenitra 14000, Morocco
| | - Otmane Zouirech
- Laboratory of Natural Substances, Pharmacology, Environment, Modeling, Health and Quality of Life (SNAMOPEQ), Faculty of Sciences Dhar El Mahraz, University Sidi Mohamed Ben Abdellah, P.O. Box 3000, Fez 30000, Morocco
| | - Mohammed Kara
- Laboratory of Biotechnology, Conservation and Valorisation of Naturals Resources (LBCVNR), Faculty of Sciences Dhar El Mehraz, Sidi Mohamed Ben Abdellah University, P.O. Box 1796 Atlas, Fez 30000, Morocco
| | - Amine Assouguem
- Laboratory of Functional Ecology and Environment, Faculty of Sciences and Technology, Sidi Mohamed Ben Abdellah University, Imouzzer Street, P.O. Box 2202, Fez 30000, Morocco
- Laboratory of Applied Organic Chemistry, Faculty of Sciences and Technology, Sidi Mohamed Ben Abdellah University, Imouzzer Street, P.O. Box 2202, Fez 30000, Morocco
| | - Saeedah Musaed Almutairi
- Department of Botany and Microbiology, College of Science, King Saud University, P.O. Box 2455, Riyadh 11451, Saudi Arabia
| | - Fahad M. Al-Hemaid
- Department of Botany and Microbiology, College of Science, King Saud University, P.O. Box 2455, Riyadh 11451, Saudi Arabia
| | - Rabab Ahmed Rasheed
- Histology and Cell Biology Department, Faculty of Medicine, King Salman International University, El Tor 46612, Egypt
| | - Riaz Ullah
- Department of Pharmacognosy, College of Pharmacy, King Saud University, P.O. Box 2455, Riyadh 11451, Saudi Arabia
| | - Arshad Mehmood Abbasi
- Department of Environmental Sciences, COMSATS University Islamabad, Abbottabad Campus, Abbottabad 22060, Pakistan or
- University of Gastronomic Sciences, Piazza Vittorio Emanuele II, 9, 12042 Pollenzo, Italy
| | - Mahjoub Aouane
- Laboratory of Natural Resources and Sustainable Development, Faculty of Sciences, University of Ibn Tofail, P.O. Box 133, Kenitra 14000, Morocco
| | - Karima Mikou
- Laboratory of Functional Ecology and Environment, Faculty of Sciences and Technology, Sidi Mohamed Ben Abdellah University, Imouzzer Street, P.O. Box 2202, Fez 30000, Morocco
| |
Collapse
|
29
|
Saritas H, Demirel HH, Bulbul A, Gorucu F, Uğurlu Z, Koc Y, Demirkan I, Saritas ZK. Myrtus Communis (Myrtle Tree: leaf and body) extract in Rat Renal Ischemia/Reperfusion injury. JOURNAL OF CLINICAL AND EXPERIMENTAL INVESTIGATIONS 2022. [DOI: 10.29333/jcei/12410] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
|
30
|
Ashkar F, Bhullar KS, Wu J. The Effect of Polyphenols on Kidney Disease: Targeting Mitochondria. Nutrients 2022; 14:nu14153115. [PMID: 35956292 PMCID: PMC9370485 DOI: 10.3390/nu14153115] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2022] [Revised: 07/26/2022] [Accepted: 07/27/2022] [Indexed: 02/01/2023] Open
Abstract
Mitochondrial function, including oxidative phosphorylation (OXPHOS), mitochondrial biogenesis, and mitochondria dynamics, are essential for the maintenance of renal health. Through modulation of mitochondrial function, the kidneys are able to sustain or recover acute kidney injury (AKI), chronic kidney disease (CKD), nephrotoxicity, nephropathy, and ischemia perfusion. Therapeutic improvement in mitochondrial function in the kidneys is related to the regulation of adenosine triphosphate (ATP) production, free radicals scavenging, decline in apoptosis, and inflammation. Dietary antioxidants, notably polyphenols present in fruits, vegetables, and plants, have attracted attention as effective dietary and pharmacological interventions. Considerable evidence shows that polyphenols protect against mitochondrial damage in different experimental models of kidney disease. Mechanistically, polyphenols regulate the mitochondrial redox status, apoptosis, and multiple intercellular signaling pathways. Therefore, this review attempts to focus on the role of polyphenols in the prevention or treatment of kidney disease and explore the molecular mechanisms associated with their pharmacological activity.
Collapse
Affiliation(s)
| | | | - Jianping Wu
- Correspondence: ; Tel.: +1-780-492-6885; Fax: +1-780-492-8524
| |
Collapse
|
31
|
Sun D, Cui S, Ma H, Zhu P, Li N, Zhang X, Zhang L, Xuan L, Li J. Salvianolate ameliorates renal tubular injury through the Keap1/Nrf2/ARE pathway in mouse kidney ischemia-reperfusion injury. JOURNAL OF ETHNOPHARMACOLOGY 2022; 293:115331. [PMID: 35489662 DOI: 10.1016/j.jep.2022.115331] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Revised: 04/19/2022] [Accepted: 04/25/2022] [Indexed: 06/14/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Acute kidney injury (AKI) is a common clinical disease characterized by rapid loss of renal function. Salvianolate is a prescribed Chinese medicine derived from traditional Chinese medicine Salvia miltiorrhiza bunge that possesses many pharmacological effects, the active components extracted from Salvia miltiorrhiza bunge have been proved to protect ischemia-reperfusion (I/R)-AKI. AIM OF THE STUDY This study aims to validate the therapeutic effect of SAL on I/R-AKI, and explore its potential pharmacological mechanism. MATERIALS AND METHODS Mice were pretreated with/without salvianolate (10, 30, and 90 mg/kg) before renal ischemia-reperfusion operation. Serum creatinine, BUN, and H&E staining were performed to evaluate renal function. Immunofluorescence analysis was conducted to measure renal tubular injury including inflammatory factors and peroxide level. Apoptosis of the kidney tissues was determined by TUNEL assay. Keap1-Nrf2-ARE and apoptosis signaling pathways were measured by Western blot, RT-PCR, and YO-PRO-1 staining in kidneys or NRK52E cells. RESULTS Pretreatment with SAL effectively alleviated renal function and ameliorated epithelial tubular injury, oxidative stress, and inflammatory response. Furthermore, the mechanistic study demonstrated that the SAL exerts anti-apoptotic effects through activation of the Keap1-Nrf2-ARE signaling pathway in renal tubular cells. CONCLUSION These findings indicate the therapeutic benefit of salvianolate in the protection of renal injury from ischemia-reperfusion, and strengthen the evidence for the AKI treatment strategy by the anti-oxidative stress response, suggesting that SAL may be a potential agent for the treatment of AKI.
Collapse
Affiliation(s)
- Dan Sun
- School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, China; State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
| | - Shichao Cui
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
| | - Haijian Ma
- Department of Pharmacology, School of Pharmacy, Fudan University, Shanghai, China
| | - Pengfei Zhu
- The First Clinical Medical School, Nanjing University of Chinese Medicine, Nanjing, China
| | - Ni Li
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
| | - Xinwen Zhang
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
| | - Lina Zhang
- Shanghai Green Valley Pharmaceutical Co.,Ltd, Shanghai, China
| | - Lijiang Xuan
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
| | - Jingya Li
- School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, China; State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China.
| |
Collapse
|
32
|
Bilgiç S, Özgöçmen M, Ozer MK. Thymoquinone ameliorates amikacin induced oxidative damage in rat brain tissue. Biotech Histochem 2022; 98:38-45. [PMID: 35811486 DOI: 10.1080/10520295.2022.2087905] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022] Open
Abstract
We investigated the potential neuroprotective effects of thymoquinone (TQ) on amikacin (AK) induced oxidative damage in rat brain. We used 21 male rats divided randomly into three equal groups. The control group was injected intraperitoneally (i.p.) with 0.5 ml 0.9% aqueous NaCl and given 1 ml 0.9% aqueous NaCl orally. The AK group was administered 1.2 g/kg aqueous AK i.p. as a single dose on the day 3 of the study. The AK + TQ group was given a single 1.2 g/kg dose of AK i.p. on the day 3 of the study plus 40 mg/kg/day TQ by oral gavage daily. Treatment with TQ increased serum ferritin and decreased serum calcium levels significantly. TQ also decreased NADPH oxidase-2, NADPH oxidase-4, and caspase-3 levels. Decreased malondialdehyde (MDA) levels and increased superoxide dismutase (SOD) and catalase (CAT) activities were detected in the AK + TQ group compared to the AK group. TQ administration inhibited lipid peroxide formation and blocked oxidative reactions, which reduced the MDA level and increased SOD and CAT activities induced by AK. Oxidative damage caused by AK was ameliorated by TQ treatment owing to its antioxidative and anti-apoptotic effects. TQ may be a potential therapeutic agent for reducing the severity of AK induced oxidative damage to the brain.
Collapse
Affiliation(s)
- Sedat Bilgiç
- Department of Medical Biochemistry, Vocational School of Health Services, University of Adıyaman, Adıyaman, Turkey
| | - Meltem Özgöçmen
- Department of Histology, and Embryology, Faculty of Medicine, Suleyman Demirel University, Isparta, Turkey
| | - Mehmet Kaya Ozer
- Department of Pharmacology, Faculty of Medicine, Adıyaman University, Adıyaman, Turkey
| |
Collapse
|
33
|
KAÇMAZ F, OZCAN O, ARPACI A, AYAZ E, BAYRAKTAR HS, GÖRÜR S. Investigation of the Effects of Lipoic Acid and Dihydrolipoate on Experimental Renal Ischemia-Reperfusion Model. MUSTAFA KEMAL ÜNIVERSITESI TIP DERGISI 2022. [DOI: 10.17944/mkutfd.1012649] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022] Open
|
34
|
Dase J, Rasyid H, Masadah R, Cangara MH, Bukhari A, Dwiyanti R, Hatta M. Analysis of mRNA and protein kidney injury Molecule-1 (KIM-1) expression in a kidney model during the initiation phase of ischemia reperfusion injury. Ann Med Surg (Lond) 2022; 75:103373. [PMID: 35242323 PMCID: PMC8857411 DOI: 10.1016/j.amsu.2022.103373] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2022] [Revised: 02/01/2022] [Accepted: 02/10/2022] [Indexed: 10/27/2022] Open
Abstract
BACKGROUND Kidney injury molecule-1 (KIM-1) is a transmembrane glycoprotein expressed predominantly on the proximal tubular epithelium. OBJECTIVE We wanted to see if there was a critical time for increased tubular damage and its related biomarker, KIM-1 mRNA, and protein expressions during the first 24 h of ischemia-reperfusion injury. METHOD An Experimental research used five male Rattus Norvegicus rats in each group. Bulldog clamp was used to clamp renal arteries and veins to create renal ischemia. Immunohistochemistry was used for the analysis of KIM-1 protein expression. While Tubular Injury Score was examined by Histopathology. RT-PCR was used for KIM-1 mRNA expression. RESULTS Tubular Injury Score (TIS) was significantly higher in ischemia than control. TIS remained similar after IR 30 min, peaked at IR 2 h, and decreased to the level of IR 30 min at IR 24 h.The KIM-1 mRNA expression was also higher in ischemia than in control. Similarly, KIM-1 mRNA expression increased more after IR 30 min, IR 2 h, and IR 24 h.The KIM-1 protein expression was higher in ischemia than in control. KIM-1 protein increased more after IR 30 min, IR for 2 h, and remained similar at IR for 24 h.KIM-1 mRNA and protein expressions at IR 2 h were significantly different compared to ischemia but not significantly different compared to that in IR 24 h. CONCLUSIONS KIM-1 mRNA and protein expressions increased within 24 h IR with the critical time was in the 2 h IR.
Collapse
Affiliation(s)
- Jerny Dase
- Departement of Forensic & Medicolegal Faculty Medicine Hasanuddin University, Dr. Wahidin Sudirohusodo Hospital, Makassar, Indonesia
| | - Haerani Rasyid
- Nephrology and Hypertension Division, Department of Internal Medicine, Faculty of Medicine, Hasanuddin University, Makassar, Indonesia
| | - Rina Masadah
- Departement of Anatomical Pathology, Faculty Medicine Hasanuddin University, Makassar, Indonesia
| | - Muhammad Husni Cangara
- Departement of Anatomical Pathology, Faculty Medicine Hasanuddin University, Makassar, Indonesia
| | - Agussalim Bukhari
- Department of Nutritional Sciences, Faculty of Medicine, Hasanuddin University, Makassar, Indonesia
| | - Ressy Dwiyanti
- Departement of Microbiology, Faculty Medicine Universitas Tadulako, Palu, Indonesia
| | - Mochammad Hatta
- Department of Microbiology, Faculty Medicine Universitas Hasanuddin Makassar, Indonesia
| |
Collapse
|
35
|
Tungalag T, Yoo YJ, Tae HJ, Yang DK. Olanzapine-Induced Therapeutic Hypothermia Attenuates Renal Injury in Rats after Asphyxial Cardiac Arrest and Resuscitation. Antioxidants (Basel) 2022; 11:antiox11030443. [PMID: 35326094 PMCID: PMC8944495 DOI: 10.3390/antiox11030443] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2022] [Revised: 02/17/2022] [Accepted: 02/21/2022] [Indexed: 01/25/2023] Open
Abstract
Return of spontaneous circulation (ROSC) through cardiopulmonary resuscitation (CPR) after cardiac arrest (CA) causes post-cardiac arrest syndrome (PCAS) due to dysfunction in various organs, which provokes acute kidney injury because of renal ischemia-reperfusion injury. Therapeutic hypothermia (TH) can reduce PCAS after CA and ROSC. However, it needs to be more sophisticated and effective. Hence, we aimed to elucidate the protective effects of olanzapine-induced TH against renal injury in asphyxial CA-induced rats. Every rat’s body temperature was maintained at 33 °C for 6 h after administering olanzapine post-CA and ROSC. Olanzapine-induced TH dramatically increased the survival rate of the rats and ameliorated renal tissue damage. Moreover, it suppressed oxidative stress responses through preservation of mitochondrial function and endoplasmic reticulum stress as the main contributor of oxidative stress. Notably, these actions of olanzapine-induced TH were mediated through the Sirt3-related signaling pathway, including the maintenance of Sirt3 and FOXO3a protein expression and the activation of AMPKα and superoxide dismutase 1 (SOD2, a mitochondrial antioxidant). This study is the first to disclose the protective effects of olanzapine-induced TH against renal injury after CA and ROSC, suggesting that olanzapine-induced TH could be utilized for treating CA followed by ROSC.
Collapse
Affiliation(s)
- Tsendsuren Tungalag
- Department of Veterinary Pharmacology and Toxicology, College of Veterinary Medicine, Jeonbuk National University, Iksan 54596, Jeollabuk-do, Korea;
| | - Yeo-Jin Yoo
- Department of Veterinary Anatomy and Toxicology, College of Veterinary Medicine and Bio-Safety Research Institute, Jeonbuk National University, Iksan 54596, Jeollabuk-do, Korea;
| | - Hyun-Jin Tae
- Department of Veterinary Anatomy and Toxicology, College of Veterinary Medicine and Bio-Safety Research Institute, Jeonbuk National University, Iksan 54596, Jeollabuk-do, Korea;
- Correspondence: (H.-J.T.); (D.K.Y.)
| | - Dong Kwon Yang
- Department of Veterinary Pharmacology and Toxicology, College of Veterinary Medicine, Jeonbuk National University, Iksan 54596, Jeollabuk-do, Korea;
- Correspondence: (H.-J.T.); (D.K.Y.)
| |
Collapse
|
36
|
Intravenous Administration of Cisplatin with Magnesium Sulfate Supplement May Prevent Kidney Toxicity in Rats: The Role of Gender and Magnesium Sulfate Dose. Int J Nephrol 2022; 2022:1218222. [PMID: 35223098 PMCID: PMC8866029 DOI: 10.1155/2022/1218222] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2021] [Accepted: 01/22/2022] [Indexed: 11/17/2022] Open
Abstract
Background. Cisplatin (CP) is widely used to treat various kinds of malignancies, but to avoid its side effects of nephrotoxicity and hypomagnesemia, magnesium supplementation is a subject of debate. The current study was designed to determine the protective role of intravenous magnesium sulfate (MgSO4) against intravenous administration of CP in male and female rats. Method. In this case-control experimental study, 80 Wistar male and female rats in 12 groups of experiments were subjected to receive intravenous administration of CP accompanied with intravenous infusion of different doses (1, 3, and 10 mg/ml solution) of MgSO4 and were compared with the control groups. Results. CP administration increased blood urea nitrogen (BUN), creatinine (Cr), kidney tissue damage score (KTDS), and kidney weight (KW), and they were attenuated by the mid-dose of MgSO4 supplementation in female rats. However, in male rats, the increase of Cr, BUN, KTDS, and KW induced by CP was ameliorated by low, mid-, and high doses of MgSO4 supplements. The levels of these markers were significantly different between male and female rats in the mid-dose of MgSO4-treated group (BUN:
, Cr:
, KTDS:
, and KW:
). CP reduced clearance of Cr (ClCr) in both male and female rats significantly compared to the control group of saline alone (Pmale = 0.002 and Pfemale = 0.001), and the mid- and high doses of MgSO4 supplements improved ClCr in female rats. There were also sex differences in ClCr in mid- (
) and high (
) doses of MgSO4-treated groups. CP accompanied with the mid-dose of MgSO4 supplement reduced the KTDS (Pmale = 0.04 and Pfemale = 0.004) and KW (Pmale = 0.002 and Pfemale = 0.042) in both male and female rats significantly when compared with the CP-alone-treated group, while there were also significant differences between the sexes (KTDS:
and KW:
). CP accompanied with three different doses of MgSO4 supplements did not improve the serum levels of lactate dehydrogenase, urine level of sodium, malondialdehyde, urine flow, and nitrite statistically when compared with the CP-alone-treated group. Conclusion. The renal protective effect of MgSO4 could be dose and gender related.
Collapse
|
37
|
Hou X, Shi J, Zhang J, Wang Z, Zhang S, Li R, Jiang W, Huang T, Guo J, Shang W. Treatment of Acute Kidney Injury Using a Dual Enzyme Embedded Zeolitic Imidazolate Frameworks Cascade That Catalyzes In Vivo Reactive Oxygen Species Scavenging. Front Bioeng Biotechnol 2022; 9:800428. [PMID: 35059388 PMCID: PMC8764232 DOI: 10.3389/fbioe.2021.800428] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2021] [Accepted: 12/01/2021] [Indexed: 12/12/2022] Open
Abstract
Significant advances have been made in recent years for the utilization of natural enzymes with antioxidant properties to treat acute kidney injury (AKI). However, these enzymes have been of limited clinical utility because of their limited cellular uptake, poor pharmacokinetic properties, and suboptimal stability. We employed a novel biomimetic mineralization approach to encapsulate catalase (CAT) and superoxide dismutase (SOD) in a zeolitic imidazolate framework-8 (ZIF-8). Next, this SOD@CAT@ZIF-8 complex was anchored with MPEG2000-COOH to yield an MPEG2000-SOD@CAT@ZIF-8 (PSCZ) composite. The composite was then used as a stable tool with antioxidant properties for the integrated cascade-based treatment of AKI, remarkably improved intracellular enzyme delivery. This dual-enzyme-embedded metal-organic framework could effectively scavenge reactive oxygen species. In conclusion, the ZIF-8-based "armor plating" represents an effective means of shielding enzymes with improved therapeutic utility to guide the precision medicine-based treatment of AKI.
Collapse
Affiliation(s)
- Xinyue Hou
- Department of Kidney Transplantation, The First Affiliated Hospital of Zhengzhou University, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, China.,Department of Molecular Pathology, Application Center for Precision Medicine, The Second Affiliated Hospital of Zhengzhou University, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, China
| | - Jianxiang Shi
- Department of Molecular Pathology, Application Center for Precision Medicine, The Second Affiliated Hospital of Zhengzhou University, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, China
| | - Jie Zhang
- Department of Kidney Transplantation, The First Affiliated Hospital of Zhengzhou University, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, China
| | - Zhigang Wang
- Department of Kidney Transplantation, The First Affiliated Hospital of Zhengzhou University, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, China
| | - Sen Zhang
- Department of Molecular Pathology, Application Center for Precision Medicine, The Second Affiliated Hospital of Zhengzhou University, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, China
| | - Ruifeng Li
- Department of Molecular Pathology, Application Center for Precision Medicine, The Second Affiliated Hospital of Zhengzhou University, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, China
| | - Wei Jiang
- Department of Molecular Pathology, Application Center for Precision Medicine, The Second Affiliated Hospital of Zhengzhou University, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, China
| | - Tingting Huang
- College of Chemistry, Jilin University, Changchun, China
| | - Jiancheng Guo
- Department of Molecular Pathology, Application Center for Precision Medicine, The Second Affiliated Hospital of Zhengzhou University, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, China
| | - Wenjun Shang
- Department of Kidney Transplantation, The First Affiliated Hospital of Zhengzhou University, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, China
| |
Collapse
|
38
|
Zhu YL, Huang J, Chen XY, Xie J, Yang Q, Wang JF, Deng XM. Senkyunolide I alleviates renal Ischemia-Reperfusion injury by inhibiting oxidative stress, endoplasmic reticulum stress and apoptosis. Int Immunopharmacol 2021; 102:108393. [PMID: 34857480 DOI: 10.1016/j.intimp.2021.108393] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2021] [Revised: 11/14/2021] [Accepted: 11/19/2021] [Indexed: 12/19/2022]
Abstract
BACKGROUND Ligusticum striatum DC. is traditionally used to treat ischemic diseases because of its potent effect against blood stasis and thrombosis, including various cardiovascular, cerebral and renal diseases. Senkyunolide I (SEI), which is the major active phthalide ingredient of Ligusticum striatum DC., is mainly distributed in kidney and has been shown to attenuate ischemia reperfusion injury in liver. However, the underlying effect of SEI against renal ischemia-reperfusion injury (IRI) remain unclear. METHODS Renal ischemia reperfusion mice model was established by clamping bilateral renal pedicles. In vitro oxidative stress model was induced by H2O2. Level of blood urea nitrogen (BUN) and serum creatinine (SCr) was tested for in vivo model evaluation, while cell viability was tested using CCK8 to evaluate in vitro model. SEI solution containing 1% DMSO was injected intraperitoneally in the I/R group, while normal saline containing 1% DMSO injected in the Sham group. Reduced glutathione (GSH) solution containing 1% DMSO was used as a positive control. RESULTS SEI protected renal function and structural integrity. It reversed the I/R-induced elevation of BUN, SCr levels and renal pathological injury. The secretion of proinflammatory cytokines including TNF-α and IL-6 was inhibited, and the renal apoptosis was attenuated by SEI. In addition, SEI played a protective role by reducing the production of reactive oxidative species (ROS), as shown by the elevated expression of antioxidant proteins including Nrf2, HO-1, NQO1, and reduced expression of endoplasmic reticulum stress (ERS) related proteins including GRP78 and CHOP. It also attenuated HK2 cell injury in an in vitro model induced by H2O2. CONCLUSIONS SEI alleviates renal injury induced by ischemia reperfusion with anti-inflammatory, anti-endoplasmic reticulum stress, anti-oxidative and anti-apoptotic effect.
Collapse
Affiliation(s)
- Ya-Lin Zhu
- Faculty of Anesthesiology, Changhai Hospital, Naval Medical University, Shanghai, China
| | - Jie Huang
- Faculty of Anesthesiology, Changhai Hospital, Naval Medical University, Shanghai, China
| | - Xue-Ying Chen
- Faculty of Anesthesiology, Changhai Hospital, Naval Medical University, Shanghai, China
| | - Jian Xie
- Faculty of Anesthesiology, Changhai Hospital, Naval Medical University, Shanghai, China
| | - Qing Yang
- Faculty of Anesthesiology, Changhai Hospital, Naval Medical University, Shanghai, China
| | - Jia-Feng Wang
- Faculty of Anesthesiology, Changhai Hospital, Naval Medical University, Shanghai, China.
| | - Xiao-Ming Deng
- Faculty of Anesthesiology, Changhai Hospital, Naval Medical University, Shanghai, China.
| |
Collapse
|
39
|
Varol S, Öktem F, Koçyiğit A, Doğan Demir A, Karataş E, Aydın M, Göknar N, İpekçi T. The impact of Technetium-99m dimercapto-succinic acid scintigraphy on DNA damage and oxidative stress in children. Int J Clin Pract 2021; 75:e14810. [PMID: 34487588 DOI: 10.1111/ijcp.14810] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/19/2021] [Revised: 08/10/2021] [Accepted: 09/03/2021] [Indexed: 01/01/2023] Open
Abstract
BACKGROUND Technetium-99m-dimercapto succinic acid (Tc-99m DMSA) scintigraphy is a commonly used imaging modality in children with urological abnormalities. The radiopharmaceuticals, which have the effects of ionising radiation, are used in this method. This study aimed to investigate the impact of the Tc-99m DMSA scan on renal oxidative stress and mononuclear leukocyte (MNL) DNA damage. METHODS Children, who were followed up by paediatric nephrology at Bezmialem Vakif University and underwent Tc-99m DMSA scintigraphy between April 2015 and January 2016 with the indication of detection of renal scars, were included in this study. The exclusion criteria were nephrolithiasis, history of premature birth and recent urinary tract infection 3 months prior to scintigraphy or antibiotic use in the last 1 month. 3 mL heparinised blood samples were obtained just before, immediately after and 1 week after the scintigraphy. MNL DNA damage, total antioxidant status (TAS) and total oxidant status (TOS) were measured in the blood samples. The oxidative stress index (OSI) was calculated. Spot urine samples were obtained from each patient before and within 3 days after performing the scintigraphy. TAS/Creatinine (TAS/Cr), TOS/Creatinine (TOS/Cr) and N-acetyl-glucosaminidase/creatinine (NAG/Cr) levels were measured in the urine samples. RESULTS Twenty-seven children were evaluated. The values between TAS, TOS and OSI levels in serum samples at baseline, immediately after and 1 week after the scintigraphy (P = .105, P = .913, and P = .721, respectively) showed no statistically significant difference. The levels of TAS/Cr, TOS/Cr, NAG/Cr ratios and OSI, which were evaluated from urine samples before and within 3 days after the scintigraphy scan were also similar (P = .391, P = .543, P = .819 and P = .179, respectively). The levels of DNA damage only increased following scintigraphy scan and decreased a week later (P < .05). CONCLUSIONS The effect of Tc-99m DMSA scintigraphy is insufficient to create oxidative damage, but it can cause DNA damage via the direct impact of ionising radiation which can be repaired again in a short time.
Collapse
Affiliation(s)
- Selçuk Varol
- Department of Pediatrics, Faculty of Medicine, Bezmialem Vakif University, Istanbul, Turkey
- Department of Pediatrics, Yeşilhisar State Hospital, Kayseri, Turkey
| | - Faruk Öktem
- Department of Pediatric Nephrology, Faculty of Medicine, Bezmialem Vakif University, Istanbul, Turkey
- Department of Pediatric Nephrology, ADAMS Compassionate Healthcare Network, Chantilly, VA, USA
| | - Abdurrahim Koçyiğit
- Department of Biochemistry, Faculty of Medicine, Bezmialem Vakif University, Istanbul, Turkey
| | - Ayşegül Doğan Demir
- Department of Pediatrics, Faculty of Medicine, Bezmialem Vakif University, Istanbul, Turkey
| | - Ersin Karataş
- Department of Biochemistry, Faculty of Medicine, Bezmialem Vakif University, Istanbul, Turkey
- Department of Moleculer Biology and Genetics, Faculty of Basic Sciences, Gebze Technical University, Kocaeli, Turkey
| | - Mehmet Aydın
- Department of Nuclear Medicine, Faculty of Medicine, Bezmialem Vakıf University, İstanbul, Turkey
| | - Nilüfer Göknar
- Department of Pediatric Nephrology, Faculty of Medicine, Bezmialem Vakif University, Istanbul, Turkey
- Department of Pediatric Nephrology, Faculty of Medicine, İstanbul Medeniyet University, İstanbul, Turkey
| | - Tümay İpekçi
- Urology Department, Medical Faculty Alanya Practice and Research Center, Başkent University, Antalya, Turkey
| |
Collapse
|
40
|
Luo Z, Liu Y, Tang Z, Liu J, Xu X, Li M, Dai Y. Quantitative Evaluation of Renal Cortex Perfusion Using Contrast-Enhanced Ultrasound Imaging Parameters in Ischemia-Reperfusion Injury in Rabbits. ULTRASOUND IN MEDICINE & BIOLOGY 2021; 47:3253-3262. [PMID: 34400032 DOI: 10.1016/j.ultrasmedbio.2021.07.013] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/19/2021] [Revised: 06/18/2021] [Accepted: 07/16/2021] [Indexed: 06/13/2023]
Abstract
The aim of this study was to evaluate blood perfusion of the renal cortex during ischemia-reperfusion (I/R) injury using quantitative contrast-enhanced ultrasound (CEUS) parameters. In this experiment, 24 rabbits were randomly divided into four groups (N = 6): sham-operated group, 24-h post-operation for I/R injury group (24-h I/R), 3-d post-operation for I/R injury group (3-d I/R) and 5 d post-operation for I/R injury group (5 d I/R). All quantitative CEUS parameters were monitored and included the gradient from the start frame to the peak frame (Grad), area under the curve (Area), time-to-peak (TTP), difference between B(intercept intensity at t=0) and A(the intensity attenuation t= 0) and arrival time (AT). Subsequently, we analyzed the changes in these parameters, as well as the correlation between changes in CEUS parameters and pathological parameters. AT and TTP values peaked 3 d after I/R surgery, which correlated with the most significant pathological changes at the same time point. These parameters (such as AT, TTP and Grad) may be useful in dynamically monitoring the severity of tissue damage at the early stage of I/R injury.
Collapse
Affiliation(s)
- Zhijian Luo
- Department of Ultrasound, Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan Province, China
| | - Yulu Liu
- Department of Medical Imaging, Southwest Medical University, Luzhou, Sichuan Province, China
| | - Ziyi Tang
- Department of Medical Imaging, Southwest Medical University, Luzhou, Sichuan Province, China
| | - Jialing Liu
- Department of Medical Imaging, Southwest Medical University, Luzhou, Sichuan Province, China
| | - Xuemei Xu
- Department of Medical Imaging, Southwest Medical University, Luzhou, Sichuan Province, China
| | - Mingxing Li
- Department of Ultrasound, Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan Province, China
| | - Yan Dai
- Department of Nuclear Medicine, Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan Province, China; Department of Pharmacy, Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan Province, China; Department of Ultrasound, Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan Province, China.
| |
Collapse
|
41
|
Fernandes SM, Watanabe M, Vattimo MDFF. Inflammation: improving understanding to prevent or ameliorate kidney diseases. J Venom Anim Toxins Incl Trop Dis 2021; 27:e20200162. [PMID: 34712277 PMCID: PMC8525891 DOI: 10.1590/1678-9199-jvatitd-2020-0162] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2020] [Accepted: 04/12/2021] [Indexed: 11/21/2022] Open
Abstract
Inflammatory processes are believed to play an important role in immune response to maintain tissue homeostasis by activating cellular signaling pathways and releasing inflammatory mediators in the injured tissue. Although acute inflammation can be considered protective, an uncontrolled inflammation may evolve to tissue damage, leading to chronic inflammatory diseases. Inflammation can be considered the major factor involved in the pathological progression of acute and chronic kidney diseases. Functional characteristics of this organ increase its vulnerability to developing various forms of injuries, including acute kidney injury (AKI) and chronic kidney disease (CKD). In view of translational research, several discoveries should be considered regarding the pathogenesis of the inflammatory process, which results in the validation of biomarkers for early detection of kidney diseases. Biomarkers enable the identification of proinflammatory mediators in kidney affections, based on laboratory research applied to clinical practice. Some inflammatory molecules can be useful biomarkers for the detection and diagnosis of kidney diseases, such as neutrophil gelatinase-associated lipocalin, kidney injury molecule-1 and interleukin 18.
Collapse
Affiliation(s)
- Sheila Marques Fernandes
- Animal Model Experimental Laboratory (LEMA), School of Nursing (EEUSP), University of São Paulo (USP), São Paulo, SP, Brazil
| | - Mirian Watanabe
- Animal Model Experimental Laboratory (LEMA), School of Nursing (EEUSP), University of São Paulo (USP), São Paulo, SP, Brazil.,Health Sciences and Wellbeing (CISBEM), University Center of United Metropolitan Colleges, São Paulo, SP, Brazil
| | | |
Collapse
|
42
|
Zhang DY, Tu T, Younis MR, Zhu KS, Liu H, Lei S, Qu J, Lin J, Huang P. Clinically translatable gold nanozymes with broad spectrum antioxidant and anti-inflammatory activity for alleviating acute kidney injury. Theranostics 2021; 11:9904-9917. [PMID: 34815794 PMCID: PMC8581429 DOI: 10.7150/thno.66518] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2021] [Accepted: 10/01/2021] [Indexed: 01/15/2023] Open
Abstract
Rationale: Acute kidney injury (AKI) is associated with aberrant generation of oxidative species and inflammation, leading to high mortality of in-hospitalized patients. Although N-acetylcysteine (NAC) showed positive effects in alleviating contrast-induced AKI, the clinical applications are strongly restrained due to the low bioavailability, low renal accumulation, short renal retention time, and high dosage-induced toxicity. Methods: We addressed the clinical dilemma of NAC by developing ultrasmall gold nanoclusters (1-2 nm) capped with NAC (denoted as Au NCs-NAC) as a nanozyme-based antioxidant defense system for AKI alleviation. Rhabdomyolysis-induced AKI mice model was developed, and the same dose of free NAC (as a control) and NAC onto Au NCs (Au NCs-NAC) was used for in vivo investigation of AKI restoration. Results: The as-developed gold nanozyme exhibited high bioavailability and good physicochemical stability as compared to NAC. Meanwhile, Au NCs-NAC showed broad-spectrum antioxidant activity of Au NCs-NAC, offering in vitro renoprotective effects, as well as macrophages by relieving inflammation under hydrogen peroxide or lipopolysaccharide stimulation. Notably, owing to the smaller size than kidney threshold (5.5 nm), Au NCs-NAC displayed preferential renal enrichment (< 2 h) and longer retention (> 24 h) in AKI mice as revealed by fluorescence imaging, thereby largely enhancing the restoration of renal function in AKI mice than free NAC by protecting the kidneys from oxidative injury and inflammation without systemic toxicity, as demonstrated by tissues staining, inflammatory cytokines and biomarkers detection, and mice survival rate. Conclusion: Owing to the synergistic anti-inflammatory/antioxidative effects, and enhanced bioavailability and renal accumulation/retention, Au NCs-NAC displayed far superior therapeutic performance than NAC alone. This work will facilitate the development of high-performance antioxidative nanoplatforms, as well as overcome the clinical limitations of small molecular drugs for AKI treatment and other inflammatory diseases.
Collapse
Affiliation(s)
- Dong-Yang Zhang
- Marshall Laboratory of Biomedical Engineering, International Cancer Center, Laboratory of Evolutionary Theranostics (LET), School of Biomedical Engineering, Shenzhen University Health Science Center, Shenzhen 518060, China
- Key Laboratory of Optoelectronic Devices and Systems of Ministry of Education and Guangdong Province, College of Optoelectronic Engineering, Shenzhen University, Shenzhen 518060, China
| | - Tianhui Tu
- Marshall Laboratory of Biomedical Engineering, International Cancer Center, Laboratory of Evolutionary Theranostics (LET), School of Biomedical Engineering, Shenzhen University Health Science Center, Shenzhen 518060, China
| | - Muhammad Rizwan Younis
- Marshall Laboratory of Biomedical Engineering, International Cancer Center, Laboratory of Evolutionary Theranostics (LET), School of Biomedical Engineering, Shenzhen University Health Science Center, Shenzhen 518060, China
- Key Laboratory of Optoelectronic Devices and Systems of Ministry of Education and Guangdong Province, College of Optoelectronic Engineering, Shenzhen University, Shenzhen 518060, China
| | - Kathy S. Zhu
- Marshall Laboratory of Biomedical Engineering, International Cancer Center, Laboratory of Evolutionary Theranostics (LET), School of Biomedical Engineering, Shenzhen University Health Science Center, Shenzhen 518060, China
- National Clinical Research Center for Oral Diseases, National Engineering Laboratory for Digital and Material Technology of Stomatology, Beijing Key Laboratory of Oral Digital Medicine, Peking University School and Hospital of Stomatology, Beijing 100081, China
| | - Hengke Liu
- Marshall Laboratory of Biomedical Engineering, International Cancer Center, Laboratory of Evolutionary Theranostics (LET), School of Biomedical Engineering, Shenzhen University Health Science Center, Shenzhen 518060, China
| | - Shan Lei
- Marshall Laboratory of Biomedical Engineering, International Cancer Center, Laboratory of Evolutionary Theranostics (LET), School of Biomedical Engineering, Shenzhen University Health Science Center, Shenzhen 518060, China
- Key Laboratory of Optoelectronic Devices and Systems of Ministry of Education and Guangdong Province, College of Optoelectronic Engineering, Shenzhen University, Shenzhen 518060, China
| | - Junle Qu
- Key Laboratory of Optoelectronic Devices and Systems of Ministry of Education and Guangdong Province, College of Optoelectronic Engineering, Shenzhen University, Shenzhen 518060, China
| | - Jing Lin
- Marshall Laboratory of Biomedical Engineering, International Cancer Center, Laboratory of Evolutionary Theranostics (LET), School of Biomedical Engineering, Shenzhen University Health Science Center, Shenzhen 518060, China
| | - Peng Huang
- Marshall Laboratory of Biomedical Engineering, International Cancer Center, Laboratory of Evolutionary Theranostics (LET), School of Biomedical Engineering, Shenzhen University Health Science Center, Shenzhen 518060, China
| |
Collapse
|
43
|
Li Y, Hepokoski M, Gu W, Simonson T, Singh P. Targeting Mitochondria and Metabolism in Acute Kidney Injury. J Clin Med 2021; 10:3991. [PMID: 34501442 PMCID: PMC8432487 DOI: 10.3390/jcm10173991] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Revised: 08/20/2021] [Accepted: 08/25/2021] [Indexed: 12/21/2022] Open
Abstract
Acute kidney injury (AKI) significantly contributes to morbidity and mortality in critically ill patients. AKI is also an independent risk factor for the development and progression of chronic kidney disease. Effective therapeutic strategies for AKI are limited, but emerging evidence indicates a prominent role of mitochondrial dysfunction and altered tubular metabolism in the pathogenesis of AKI. Therefore, a comprehensive, mechanistic understanding of mitochondrial function and renal metabolism in AKI may lead to the development of novel therapies in AKI. In this review, we provide an overview of current state of research on the role of mitochondria and tubular metabolism in AKI from both pre-clinical and clinical studies. We also highlight current therapeutic strategies which target mitochondrial function and metabolic pathways for the treatment of AKI.
Collapse
Affiliation(s)
- Ying Li
- Division of Nephrology and Hypertension, University of California San Diego, San Diego, CA 92093, USA;
- VA San Diego Healthcare System, San Diego, CA 92161, USA;
| | - Mark Hepokoski
- VA San Diego Healthcare System, San Diego, CA 92161, USA;
- Division of Pulmonary, Critical Care and Sleep Medicine, University of California San Diego, San Diego, CA 92093, USA; (W.G.); (T.S.)
| | - Wanjun Gu
- Division of Pulmonary, Critical Care and Sleep Medicine, University of California San Diego, San Diego, CA 92093, USA; (W.G.); (T.S.)
| | - Tatum Simonson
- Division of Pulmonary, Critical Care and Sleep Medicine, University of California San Diego, San Diego, CA 92093, USA; (W.G.); (T.S.)
| | - Prabhleen Singh
- Division of Nephrology and Hypertension, University of California San Diego, San Diego, CA 92093, USA;
- VA San Diego Healthcare System, San Diego, CA 92161, USA;
| |
Collapse
|
44
|
Zhao X, Wang L, Li J, Peng L, Tang C, Zha X, Ke K, Yang M, Su B, Yang W. Redox-Mediated Artificial Non-Enzymatic Antioxidant MXene Nanoplatforms for Acute Kidney Injury Alleviation. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2021; 8:e2101498. [PMID: 34272933 PMCID: PMC8456282 DOI: 10.1002/advs.202101498] [Citation(s) in RCA: 51] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/11/2021] [Revised: 05/23/2021] [Indexed: 02/05/2023]
Abstract
Acute kidney injury (AKI), as a common oxidative stress-related renal disease, causes high mortality in clinics annually, and many other clinical diseases, including the pandemic COVID-19, have a high potential to cause AKI, yet only rehydration, renal dialysis, and other supportive therapies are available for AKI in the clinics. Nanotechnology-mediated antioxidant therapy represents a promising therapeutic strategy for AKI treatment. However, current enzyme-mimicking nanoantioxidants show poor biocompatibility and biodegradability, as well as non-specific ROS level regulation, further potentially causing deleterious adverse effects. Herein, the authors report a novel non-enzymatic antioxidant strategy based on ultrathin Ti3 C2 -PVP nanosheets (TPNS) with excellent biocompatibility and great chemical reactivity toward multiple ROS for AKI treatment. These TPNS nanosheets exhibit enzyme/ROS-triggered biodegradability and broad-spectrum ROS scavenging ability through the readily occurring redox reaction between Ti3 C2 and various ROS, as verified by theoretical calculations. Furthermore, both in vivo and in vitro experiments demonstrate that TPNS can serve as efficient antioxidant platforms to scavenge the overexpressed ROS and subsequently suppress oxidative stress-induced inflammatory response through inhibition of NF-κB signal pathway for AKI treatment. This study highlights a new type of therapeutic agent, that is, the redox-mediated non-enzymatic antioxidant MXene nanoplatforms in treatment of AKI and other ROS-associated diseases.
Collapse
Affiliation(s)
- Xing Zhao
- College of Polymer Science and EngineeringSichuan UniversityState Key Laboratory of Polymer Materials EngineeringChengduSichuan610065China
| | - Li‐Ya Wang
- Department of NephrologyMed‐X Center for ManufacturingWest China HospitalSichuan UniversityChengdu610041China
| | - Jia‐Meng Li
- Department of NephrologyMed‐X Center for ManufacturingWest China HospitalSichuan UniversityChengdu610041China
| | - Li‐Mei Peng
- College of Polymer Science and EngineeringSichuan UniversityState Key Laboratory of Polymer Materials EngineeringChengduSichuan610065China
| | - Chun‐Yan Tang
- College of Polymer Science and EngineeringSichuan UniversityState Key Laboratory of Polymer Materials EngineeringChengduSichuan610065China
| | - Xiang‐Jun Zha
- College of Polymer Science and EngineeringSichuan UniversityState Key Laboratory of Polymer Materials EngineeringChengduSichuan610065China
| | - Kai Ke
- College of Polymer Science and EngineeringSichuan UniversityState Key Laboratory of Polymer Materials EngineeringChengduSichuan610065China
| | - Ming‐Bo Yang
- College of Polymer Science and EngineeringSichuan UniversityState Key Laboratory of Polymer Materials EngineeringChengduSichuan610065China
| | - Bai‐Hai Su
- Department of NephrologyMed‐X Center for ManufacturingWest China HospitalSichuan UniversityChengdu610041China
- The First People's Hospital of Shuangliu DistrictChengdu610200China
| | - Wei Yang
- College of Polymer Science and EngineeringSichuan UniversityState Key Laboratory of Polymer Materials EngineeringChengduSichuan610065China
| |
Collapse
|
45
|
Jibin K, Victor M, Saranya G, Santhakumar H, Murali V, Maiti KK, Jayasree RS. Nanohybrids of Magnetically Intercalated Optical Metamaterials for Magnetic Resonance/Raman Imaging and In Situ Chemodynamic/Photothermal Therapy. ACS APPLIED BIO MATERIALS 2021; 4:5742-5752. [PMID: 35006723 DOI: 10.1021/acsabm.1c00510] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Target-specific reactive oxygen species (ROS)-based cancer treatments with high therapeutic efficacy and minimal side effects have been identified recently as a potentially effective cancer management strategy. Herein, we report the fabrication of a targeted nanotheranostic agent built on an iron oxide nanoparticle-decorated graphene-gold hybrid [plasmonic magnetic nanoprobe (PMNP)] for self-guided magnetic resonance (MR)/surface-enhanced Raman scattering imaging and photothermal therapy (PTT)/chemodynamic therapy (CDT). In the presence of glutathione, which is abundant in the tumor environment, the iron oxide nanoparticles undergo in situ reduction, which in turn generates hydroxyl radicals via a Fenton reaction to realize targeted destruction of tumor cells. Moreover, the localized production of heat benefited from the near-infrared absorption of the PMNP accelerates the intratumoral ROS generation process, with a synergistic effect of CDT/PTT. Furthermore, the probe offers an accurate visualization of the intracellular localization of the material through SERS/MR dual imaging channels. In view of the advantages offered by the tumor-specific stimuli-responsive nature of the probe, the PMNP presents as an effective tool for cancer management.
Collapse
Affiliation(s)
- Kunnumpurathu Jibin
- Division of Biophotonics and Imaging, Department of Biomaterial Sciences and Technology, Biomedical Technology Wing, Sree Chitra Tirunal Institute for Medical Sciences and Technology (SCTIMST), Trivandrum 695012, India
| | - Marina Victor
- Division of Biophotonics and Imaging, Department of Biomaterial Sciences and Technology, Biomedical Technology Wing, Sree Chitra Tirunal Institute for Medical Sciences and Technology (SCTIMST), Trivandrum 695012, India
| | - Giridharan Saranya
- Chemical Science & Technology Division, CSIR-National Institute for Interdisciplinary Science & Technology (NIIST) and Academy of Scientific and Innovative Research (AcSIR), CSIR-NIIST, Thiruvananthapuram, 695019 Kerala, India
| | - Hema Santhakumar
- Division of Biophotonics and Imaging, Department of Biomaterial Sciences and Technology, Biomedical Technology Wing, Sree Chitra Tirunal Institute for Medical Sciences and Technology (SCTIMST), Trivandrum 695012, India
| | - Vishnupriya Murali
- Chemical Science & Technology Division, CSIR-National Institute for Interdisciplinary Science & Technology (NIIST) and Academy of Scientific and Innovative Research (AcSIR), CSIR-NIIST, Thiruvananthapuram, 695019 Kerala, India
| | - Kaustabh K Maiti
- Chemical Science & Technology Division, CSIR-National Institute for Interdisciplinary Science & Technology (NIIST) and Academy of Scientific and Innovative Research (AcSIR), CSIR-NIIST, Thiruvananthapuram, 695019 Kerala, India
| | - Ramapurath S Jayasree
- Division of Biophotonics and Imaging, Department of Biomaterial Sciences and Technology, Biomedical Technology Wing, Sree Chitra Tirunal Institute for Medical Sciences and Technology (SCTIMST), Trivandrum 695012, India
| |
Collapse
|
46
|
Jawad A, Yoo YJ, Cho JH, Yoon JC, Tian W, Islam MS, Lee EY, Shin HY, Kim SE, Kim K, Ahn D, Park BY, Kim IS, Lee JH, Tae HJ. Therapeutic hypothermia effect on asphyxial cardiac arrest-induced renal ischemia/reperfusion injury via change of Nrf2/HO-1 levels. Exp Ther Med 2021; 22:1031. [PMID: 34373717 PMCID: PMC8343472 DOI: 10.3892/etm.2021.10463] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Accepted: 06/16/2021] [Indexed: 01/08/2023] Open
Abstract
The present study aimed to investigate the renoprotective effect of therapeutic hypothermia (TH) on renal ischemia-reperfusion injury (RI/RI) induced by asphyxial cardiac arrest (CA) in rats. A total of 48 male rats were randomly divided into five groups: i) Sham (n=6); ii) Normothermia + CA (Normo.) (n=14); iii) Normo. and 2 h of TH after return of spontaneous circulation (ROSC) (n=12); iv) Normo. and 4 h of TH after ROSC (n=9); and v) Normo. and 6 h of TH after ROSC (n=7). All rats except the Sham group underwent asphyxia CA and were sacrificed 1 day after ROSC. The survival rate increased from 42.8% in the Normo. group to 50, 66.6 and 85.7% in the groups with 2, 4 and 6 h of TH after CA, respectively. TH attenuated the histopathological changes of the renal tissues following ROSC and the levels of blood urea nitrogen, serum creatinine and malondialdehyde in renal tissues. On immunohistochemistry, the relative optical density of nuclear erythroid-related factor-2 (Nrf2) and heme oxygenase (HO-1) expression in renal tissues increased in the Normo. group compared with that in the Sham group and exhibited further significant increases at 6 h of TH after ROSC. In conclusion, TH attenuated renal injury and increased the expression of Nrf2 and HO-1 in a TH treatment time-dependent manner.
Collapse
Affiliation(s)
- Ali Jawad
- Department of Veterinary Medicine and Bio-Safety Research Institute, Jeonbuk National University, Iksan, Jeollabuk-do 54696, Republic of Korea
| | - Yeo-Jin Yoo
- Department of Veterinary Medicine and Bio-Safety Research Institute, Jeonbuk National University, Iksan, Jeollabuk-do 54696, Republic of Korea
| | - Jeong-Hwi Cho
- Department of Veterinary Medicine and Bio-Safety Research Institute, Jeonbuk National University, Iksan, Jeollabuk-do 54696, Republic of Korea
| | - Jae Chol Yoon
- Department of Emergency Medicine, Research Institute of Clinical Medicine of Jeonbuk National University, Jeonju, Jeollabuk-do 54907, Republic of Korea
| | - Weishun Tian
- Department of Veterinary Medicine and Bio-Safety Research Institute, Jeonbuk National University, Iksan, Jeollabuk-do 54696, Republic of Korea
| | - Mohammad Sadikul Islam
- Department of Veterinary Medicine and Bio-Safety Research Institute, Jeonbuk National University, Iksan, Jeollabuk-do 54696, Republic of Korea
| | - Eui-Yong Lee
- Department of Veterinary Medicine and Bio-Safety Research Institute, Jeonbuk National University, Iksan, Jeollabuk-do 54696, Republic of Korea
| | - Ha-Young Shin
- Department of Veterinary Medicine and Bio-Safety Research Institute, Jeonbuk National University, Iksan, Jeollabuk-do 54696, Republic of Korea
| | - So Eun Kim
- Department of Emergency Medicine, Research Institute of Clinical Medicine of Jeonbuk National University, Jeonju, Jeollabuk-do 54907, Republic of Korea
| | - Kyunghwa Kim
- Department of Thoracic and Cardiovascular Surgery, Research Institute of Clinical Medicine of Jeonbuk National University-Biomedical Research Institute of Jeonbuk National University Hospital, Jeonju, Jeollabuk-do 54907, Republic of Korea
| | - Dongchoon Ahn
- Department of Veterinary Medicine and Bio-Safety Research Institute, Jeonbuk National University, Iksan, Jeollabuk-do 54696, Republic of Korea
| | - Byung-Yong Park
- Department of Veterinary Medicine and Bio-Safety Research Institute, Jeonbuk National University, Iksan, Jeollabuk-do 54696, Republic of Korea
| | - In-Shik Kim
- Department of Veterinary Medicine and Bio-Safety Research Institute, Jeonbuk National University, Iksan, Jeollabuk-do 54696, Republic of Korea
| | - Jun Ho Lee
- Department of Anesthesiology and Pain Medicine, Research Institute of Clinical Medicine of Jeonbuk National University-Biomedical Research Institute of Jeonbuk National University Hospital, Jeonju, Jeollabuk-do 54907, Republic of Korea
| | - Hyun-Jin Tae
- Department of Veterinary Medicine and Bio-Safety Research Institute, Jeonbuk National University, Iksan, Jeollabuk-do 54696, Republic of Korea
| |
Collapse
|
47
|
Tseng WC, Lee PY, Tsai MT, Chang FP, Chen NJ, Chien CT, Hung SC, Tarng DC. Hypoxic mesenchymal stem cells ameliorate acute kidney ischemia-reperfusion injury via enhancing renal tubular autophagy. Stem Cell Res Ther 2021; 12:367. [PMID: 34183058 PMCID: PMC8240301 DOI: 10.1186/s13287-021-02374-x] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2021] [Accepted: 05/09/2021] [Indexed: 12/29/2022] Open
Abstract
BACKGROUND Acute kidney injury (AKI) is an emerging global healthcare issue without effective therapy yet. Autophagy recycles damaged organelles and helps maintain tissue homeostasis in acute renal ischemia-reperfusion (I/R) injury. Hypoxic mesenchymal stem cells (HMSCs) represent an innovative cell-based therapy in AKI. Moreover, the conditioned medium of HMSCs (HMSC-CM) rich in beneficial trophic factors may serve as a cell-free alternative therapy. Nonetheless, whether HMSCs or HMSC-CM mitigate renal I/R injury via modulating tubular autophagy remains unclear. METHODS Renal I/R injury was induced by clamping of the left renal artery with right nephrectomy in male Sprague-Dawley rats. The rats were injected with either PBS, HMSCs, or HMSC-CM immediately after the surgery and sacrificed 48 h later. Renal tubular NRK-52E cells subjected to hypoxia-reoxygenation (H/R) injury were co-cultured with HMSCs or treated with HMSC-CM to assess the regulatory effects of HSMCs on tubular autophagy and apoptosis. The association of tubular autophagy gene expression and renal recovery was also investigated in patients with ischemic AKI. RESULT HMSCs had a superior anti-oxidative effect in I/R-injured rat kidneys as compared to normoxia-cultured mesenchymal stem cells. HMSCs further attenuated renal macrophage infiltration and inflammation, reduced tubular apoptosis, enhanced tubular proliferation, and improved kidney function decline in rats with renal I/R injury. Moreover, HMSCs suppressed superoxide formation, reduced DNA damage and lipid peroxidation, and increased anti-oxidants expression in renal tubular epithelial cells during I/R injury. Co-culture of HMSCs with H/R-injured NRK-52E cells also lessened tubular cell death. Mechanistically, HMSCs downregulated the expression of pro-inflammatory interleukin-1β, proapoptotic Bax, and caspase 3. Notably, HMSCs also upregulated the expression of autophagy-related LC3B, Atg5 and Beclin 1 in renal tubular cells both in vivo and in vitro. Addition of 3-methyladenine suppressed the activity of autophagy and abrogated the renoprotective effects of HMSCs. The renoprotective effect of tubular autophagy was further validated in patients with ischemic AKI. AKI patients with higher renal LC3B expression were associated with better renal recovery. CONCLUSION The present study describes that the enhancing effect of MSCs, and especially of HMCSs, on tissue autophagy can be applied to suppress renal tubular apoptosis and attenuate renal impairment during renal I/R injury in the rat. Our findings provide further mechanistic support to HMSCs therapy and its investigation in clinical trials of ischemic AKI.
Collapse
Affiliation(s)
- Wei-Cheng Tseng
- Division of Nephrology, Department of Medicine, Taipei Veterans General Hospital, 201, Section 2, Shih-Pai Road, Taipei, 11217, Taiwan.,Faculty of Medicine, School of Medicine, National Yang-Ming University, Taipei, Taiwan.,Institute of Clinical Medicine, School of Medicine, National Yang-Ming University, Taipei, Taiwan.,Center for Intelligent Drug Systems and Smart Bio-devices (IDS2B), National Chiao-Tung University, Hsinchu, Taiwan.,Institute of Clinical Medicine, School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Pei-Ying Lee
- Holistic Education Center, Tzu Chi University of Science and Technology, Hualien, Taiwan
| | - Ming-Tsun Tsai
- Division of Nephrology, Department of Medicine, Taipei Veterans General Hospital, 201, Section 2, Shih-Pai Road, Taipei, 11217, Taiwan.,Faculty of Medicine, School of Medicine, National Yang-Ming University, Taipei, Taiwan.,Institute of Clinical Medicine, School of Medicine, National Yang-Ming University, Taipei, Taiwan.,Institute of Clinical Medicine, School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Fu-Pang Chang
- Faculty of Medicine, School of Medicine, National Yang-Ming University, Taipei, Taiwan.,Institute of Clinical Medicine, School of Medicine, National Yang-Ming University, Taipei, Taiwan.,Institute of Clinical Medicine, School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan.,Department of Pathology and Laboratory Medicine, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Nien-Jung Chen
- Institute of Clinical Medicine, School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan.,Institute of Microbiology and Immunology, School of Medicine, National Yang-Ming University, Taipei, Taiwan
| | - Chiang-Ting Chien
- Department of Life Science, School of Life Science, National Taiwan Normal University, Taipei, Taiwan
| | - Shih-Chieh Hung
- Institute of Clinical Medicine, School of Medicine, National Yang-Ming University, Taipei, Taiwan. .,Institute of Clinical Medicine, School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan. .,Integrative Stem Cell Center, Department of Orthopedics, and Institute of New Drug Development, New Drug Development Center, China Medical University, Taichung, Taiwan. .,Institute of Biomedical Sciences, Academia Sinica, 128, Section 2, Academia Road, Taipei, 11529, Taiwan.
| | - Der-Cherng Tarng
- Division of Nephrology, Department of Medicine, Taipei Veterans General Hospital, 201, Section 2, Shih-Pai Road, Taipei, 11217, Taiwan. .,Faculty of Medicine, School of Medicine, National Yang-Ming University, Taipei, Taiwan. .,Institute of Clinical Medicine, School of Medicine, National Yang-Ming University, Taipei, Taiwan. .,Center for Intelligent Drug Systems and Smart Bio-devices (IDS2B), National Chiao-Tung University, Hsinchu, Taiwan. .,Institute of Clinical Medicine, School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan. .,Department and Institute of Physiology, School of Medicine, National Yang-Ming University, Taipei, Taiwan. .,Department of Biological Science and Technology, College of Biological Science and Technology, National Chiao-Tung University, Hsinchu, Taiwan.
| |
Collapse
|
48
|
Abstract
The kidney is a highly metabolic organ that requires substantial adenosine triphosphate for the active transport required to maintain water and solute reabsorption. Aberrations in energy availability and energy utilization can lead to cellular dysfunction and death. Mitochondria are essential for efficient energy production. The pathogenesis of acute kidney injury is complex and varies with different types of injury. However, multiple distinct acute kidney injury syndromes share a common dysregulation of energy metabolism. Pathways of energy metabolism and mitochondrial dysfunction are emerging as critical drivers of acute kidney injury and represent new potential targets for treatment. This review shows the basic metabolic pathways that all cells depend on for life; describes how the kidney optimizes those pathways to meet its anatomic, physiologic, and metabolic needs; summarizes the importance of metabolic and mitochondrial dysfunction in acute kidney injury; and analyzes the mitochondrial processes that become dysregulated in acute kidney injury including mitochondrial dynamics, mitophagy, mitochondrial biogenesis, and changes in mitochondrial energy metabolism.
Collapse
Affiliation(s)
- Amanda J Clark
- Division of Nephrology, Boston Children's Hospital, Harvard Medical School, Boston, MA
| | - Samir M Parikh
- Division of Nephrology, Center for Vascular Biology Research, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA.
| |
Collapse
|
49
|
Meade RD, Akerman AP, Notley SR, McGinn R, Poirier P, Gosselin P, Kenny GP. Physiological factors characterizing heat-vulnerable older adults: A narrative review. ENVIRONMENT INTERNATIONAL 2020; 144:105909. [PMID: 32919284 DOI: 10.1016/j.envint.2020.105909] [Citation(s) in RCA: 152] [Impact Index Per Article: 30.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/16/2020] [Revised: 05/24/2020] [Accepted: 06/17/2020] [Indexed: 05/26/2023]
Abstract
More frequent and intense periods of extreme heat (heatwaves) represent the most direct challenge to human health posed by climate change. Older adults are particularly vulnerable, especially those with common age-associated chronic health conditions (e.g., cardiovascular disease, hypertension, obesity, type 2 diabetes, chronic kidney disease). In parallel, the global population is aging and age-associated disease rates are on the rise. Impairments in the physiological responses tasked with maintaining homeostasis during heat exposure have long been thought to contribute to increased risk of health disorders in older adults during heatwaves. As such, a comprehensive overview of the provisional links between age-related physiological dysfunction and elevated risk of heat-related injury in older adults would be of great value to healthcare officials and policy makers concerned with protecting heat-vulnerable sectors of the population from the adverse health impacts of heatwaves. In this narrative review, we therefore summarize our current understanding of the physiological mechanisms by which aging impairs the regulation of body temperature, hemodynamic stability and hydration status. We then examine how these impairments may contribute to acute pathophysiological events common during heatwaves (e.g., heatstroke, major adverse cardiovascular events, acute kidney injury) and discuss how age-associated chronic health conditions may exacerbate those impairments. Finally, we briefly consider the importance of physiological research in the development of climate-health programs aimed at protecting heat-vulnerable individuals.
Collapse
Affiliation(s)
- Robert D Meade
- Human and Environmental Physiology Research Unit, School of Human Kinetics, University of Ottawa, Ottawa, Ontario, Canada
| | - Ashley P Akerman
- Human and Environmental Physiology Research Unit, School of Human Kinetics, University of Ottawa, Ottawa, Ontario, Canada
| | - Sean R Notley
- Human and Environmental Physiology Research Unit, School of Human Kinetics, University of Ottawa, Ottawa, Ontario, Canada
| | - Ryan McGinn
- Department of Anesthesiology and Pain Medicine, The Ottawa Hospital, University of Ottawa, Ottawa, ON, Canada
| | - Paul Poirier
- Institut Universitaire de Cardiologie et de Pneumologie de Québec, Université Laval, Québec, Québec, Canada
| | - Pierre Gosselin
- Institut National de Santé Publique du Québec and Université Laval, Québec, Québec, Canada
| | - Glen P Kenny
- Human and Environmental Physiology Research Unit, School of Human Kinetics, University of Ottawa, Ottawa, Ontario, Canada; Clinical Epidemiology Program, Ottawa Hospital Research Institute, Ottawa, Ontario, Canada.
| |
Collapse
|
50
|
Wetzel MD, Stanley K, Wang WW, Maity S, Madesh M, Reeves WB, Awad AS. Selective inhibition of arginase-2 in endothelial cells but not proximal tubules reduces renal fibrosis. JCI Insight 2020; 5:142187. [PMID: 32956070 PMCID: PMC7566719 DOI: 10.1172/jci.insight.142187] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Accepted: 09/02/2020] [Indexed: 01/10/2023] Open
Abstract
Fibrosis is the final common pathway in the pathophysiology of most forms of chronic kidney disease (CKD). As treatment of renal fibrosis still remains largely supportive, a refined understanding of the cellular and molecular mechanisms of kidney fibrosis and the development of novel compounds are urgently needed. Whether arginases play a role in the development of fibrosis in CKD is unclear. We hypothesized that endothelial arginase-2 (Arg2) promotes the development of kidney fibrosis induced by unilateral ureteral obstruction (UUO). Arg2 expression and arginase activity significantly increased following renal fibrosis. Pharmacologic blockade or genetic deficiency of Arg2 conferred kidney protection following renal fibrosis, as reflected by a reduction in kidney interstitial fibrosis and fibrotic markers. Selective deletion of Arg2 in endothelial cells (Tie2Cre/Arg2fl/fl) reduced the level of fibrosis after UUO. In contrast, selective deletion of Arg2 specifically in proximal tubular cells (Ggt1Cre/Arg2fl/fl) failed to reduce renal fibrosis after UUO. Furthermore, arginase inhibition restored kidney nitric oxide (NO) levels, oxidative stress, and mitochondrial function following UUO. These findings indicate that endothelial Arg2 plays a major role in renal fibrosis via its action on NO and mitochondrial function. Blocking Arg2 activity or expression could be a novel therapeutic approach for prevention of CKD.
Collapse
|