1
|
Guven C, Koçak S, Karadag A, Aydın H, Yalcin A, Turk A. Adropin as a protective agent against renal ischemia-reperfusion injury induced by suprarenal aortic cross-clamping in rats. BMC Nephrol 2025; 26:154. [PMID: 40140781 PMCID: PMC11948768 DOI: 10.1186/s12882-025-04087-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2024] [Accepted: 03/20/2025] [Indexed: 03/28/2025] Open
Abstract
BACKGROUND The development of protective therapeutic strategies against acute kidney injury associated with suprarenal aneurysms, renal artery occlusive disease, and suprarenal aortic reconstruction is of paramount importance. Adropin is a peptide hormone that has been shown to protect vascular endothelial cells and reduce oxidative stress, apoptosis, and inflammation. Therefore, in addition to its metabolic and vascular effects, adropin has potential as a therapeutic agent in renal ischemia-reperfusion injury. This study aims to investigate the protective effects of adropine on kidney ischemia-reperfusion (IR) injury under the suprarenal aortic cross clamp. METHODS Male Sprague Dawley rats were divided into six groups, with seven rats in each group for the study design. The control and ischemia reperfusion (IR) induced groups were designated as the two groups while the other four groups (TR1 to TR4 ) were administered varying doses of adropin at 0.5 mg/kg, 1 mg/kg, 1.5 mg/kg, and 2 mg/kg for each group. After a 60 min ischemic period, a 24-hour reperfusion period was implemented to assess the outcomes of adropin treatment on renal IR. Histopathological analysis was performed in conjunction with determination of apoptosis, and malondialdehyde (MDA) levels. In addition, serum concentrations of adropin, superoxide dismutase (SOD), glutathione peroxidase (GSH-Px), as well as endothelial nitric oxide synthase (eNOS) were measured in order to further define the biochemical reactions of the treatment. RESULTS MDA levels were significantly elevated in the IR group compared to the control group, while the activities of eNOS, SOD, and GSH-Px enzymes were significantly decreased (P < 0.05). MDA levels in the treatment groups were lower than those in the IR group, whereas eNOS, SOD, and GSH-Px levels were higher (P < 0.05). Statistically, the lowest adropin levels were observed in the IR group, while the highest levels were noted in the TR4 group (P < 0.05). Histopathological examination revealed a reduction in tissue damage in the treatment groups compared to the IR group. CONCLUSION The histological and biochemical findings from this study indicate that adropin provides protective effects against renal ischemia-reperfusion injury in a dose-dependent manner.
Collapse
Affiliation(s)
- Cengiz Guven
- Faculty of Medicine, Department of Cardiovascular Surgery, Adiyaman University, Adiyaman, Turkey
| | - Seda Koçak
- Faculty of Medicine, Department of Physiology, Kırşehir Ahi Evran University, Kırşehir, Turkey.
| | - Abdullah Karadag
- Faculty of Medicine, Department of Physiology, Adiyaman University, Adiyaman, Turkey
| | - Hasan Aydın
- Faculty of Pharmacy, Department of Pharmaceutical Toxicology, Adiyaman University, Adiyaman, Turkey
| | - Alper Yalcin
- Faculty of Medicine, Department of Histology and Embryology, Kahramanmaraş Sütçü Imam University, Adiyaman, Turkey
| | - Ahmet Turk
- Faculty of Medicine, Department of Histology and Embryology, Adiyaman University, Adıyaman, Turkey
| |
Collapse
|
2
|
Pushpakumar S, Juin SK, Almarshood H, Gondim DD, Ouseph R, Sen U. Diallyl Trisulfide Attenuates Ischemia-Reperfusion-Induced ER Stress and Kidney Dysfunction in Aged Female Mice. Cells 2025; 14:420. [PMID: 40136669 PMCID: PMC11941362 DOI: 10.3390/cells14060420] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2024] [Revised: 03/06/2025] [Accepted: 03/07/2025] [Indexed: 03/27/2025] Open
Abstract
Ischemia-reperfusion injury (IRI) is a common cause of acute kidney injury (AKI) in the aging population. Gender studies show that aging is associated with loss of protection from AKI in the female population. While ER stress contributes to IRI-induced AKI in the young, ER regulation during IR in the aged kidney is unclear. Because current evidence suggests hydrogen sulfide (H2S) modulates ER stress, we investigated whether exogenous supplementation of diallyl trisulfide (DATS), an H2S donor, mitigates AKI in aged female kidneys. Wild-type (WT, C57BL/6J) mice aged 75-78 weeks were treated with or without DATS before and after renal IRI. IRI increased ER stress proteins, inflammation, and fibrosis markers in the IRI kidney compared to the control. DATS mitigated ER stress, and reduced inflammation and fibrosis markers in the IRI kidney. Further, IRI kidneys demonstrated reduced blood flow, vascularity, angiogenesis, increased resistive index (RI), and reduced function. DATS treatment upregulated PI3K, AKT, p-mTOR, and pMAPK signaling to stimulate angiogenesis, which improved vascular density, blood flow, and renal function. Together, our results suggest that DATS rescues the aged female kidney IRI by modulating ER stress and upregulation of angiogenesis.
Collapse
Affiliation(s)
- Sathnur Pushpakumar
- Department of Physiology, University of Louisville School of Medicine, Louisville, KY 40202, USA
| | - Subir Kumar Juin
- Department of Microbiology and Immunology, University of Louisville School of Medicine, Louisville, KY 40202, USA;
| | - Hebah Almarshood
- Department of Physiology, University of Louisville School of Medicine, Louisville, KY 40202, USA
| | - Dibson Dibe Gondim
- Department of Pathology, University of Louisville School of Medicine, Louisville, KY 40202, USA
| | - Rosemary Ouseph
- Division of Nephrology & Hypertension, University of Louisville School of Medicine, Louisville, KY 40202, USA
| | - Utpal Sen
- Department of Physiology, University of Louisville School of Medicine, Louisville, KY 40202, USA
| |
Collapse
|
3
|
Balci Ç, Özcan MS, Aşci H, Karabacak P, Kuruşçu O, Taner R, Özmen Ö, Tepebaşi MY, İlhan İ, Çömlekçi S. Radiofrequency Electromagnetic and Pulsed Magnetic Fields Protected the Kidney Against Lipopolysaccharide-Induced Acute Systemic Inflammation, Oxidative Stress, and Apoptosis by Regulating the IL-6/HIF1α/eNOS and Bcl2/Bax/Cas-9 Pathways. MEDICINA (KAUNAS, LITHUANIA) 2025; 61:238. [PMID: 40005355 PMCID: PMC11857468 DOI: 10.3390/medicina61020238] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/26/2024] [Revised: 01/25/2025] [Accepted: 01/27/2025] [Indexed: 02/27/2025]
Abstract
Background/Objectives: Sepsis-associated acute kidney injury caused by lipopolysaccharide (LPS) is related to hypoxia, amplification of the inflammatory response, oxidative stress, mitochondrial dysfunction, and apoptosis. This study aims to explore the protective effects of a radiofrequency electromagnetic field (RF-EMF) and a pulsed magnetic field (PMF) on acute kidney injury in rats. Materials and methods: Forty female Wistar albino rats were randomly divided into five groups (each containing eight rats): control, LPS, RF-EMF, PMF, and RF-EMF + PMF groups. Six hours after LPS application, blood and tissues were removed for histopathological, immunohistochemical, biochemical, and genetic analysis. Results: Histopathological findings, caspase-3, inducible nitric oxide synthase and tumor necrosis factor-alpha immunoexpressions, total oxidant status and oxidative stress index levels, and interleukin-6, hypoxia-inducible factor alpha, Bcl-2-associated X protein, and caspase 9 gene expression in kidney tissue and blood urine nitrogen and creatinine levels in blood were increased, whereas endothelial nitric oxide synthase and B-cell lymphoma 2 gene expression were decreased in the LPS groups. Both RF-EMF and PMF reversed all these findings and recovered renal tissues. Conclusions: Noninvasive, nontoxic, low-cost PMF and RF-EMF, both single and combined, have been demonstrated to have renoprotective anti-inflammatory, antioxidant, and antiapoptotic effects.
Collapse
Affiliation(s)
- Çağrı Balci
- Department of Anesthesiology and Reanimation, Faculty of Medicine, Suleyman Demirel University, 32260 Isparta, Turkey; (Ç.B.); (P.K.); (O.K.)
| | - Mustafa S. Özcan
- Department of Anesthesiology and Reanimation, Faculty of Medicine, Suleyman Demirel University, 32260 Isparta, Turkey; (Ç.B.); (P.K.); (O.K.)
| | - Halil Aşci
- Department of Pharmacology, Faculty of Medicine, Suleyman Demirel University, 32260 Isparta, Turkey;
| | - Pınar Karabacak
- Department of Anesthesiology and Reanimation, Faculty of Medicine, Suleyman Demirel University, 32260 Isparta, Turkey; (Ç.B.); (P.K.); (O.K.)
| | - Oya Kuruşçu
- Department of Anesthesiology and Reanimation, Faculty of Medicine, Suleyman Demirel University, 32260 Isparta, Turkey; (Ç.B.); (P.K.); (O.K.)
| | - Rümeysa Taner
- Department of Bioengineering, Institute of Science, Suleyman Demirel University, 32260 Isparta, Turkey; (R.T.); (S.Ç.)
| | - Özlem Özmen
- Department of Pathology, Faculty of Veterinary Medicine, Burdur Mehmet Akif Ersoy University, 15200 Burdur, Turkey;
| | - Muhammet Y. Tepebaşi
- Department of Medical Genetics, Faculty of Medicine, Suleyman Demirel University, 32260 Isparta, Turkey;
| | - İlter İlhan
- Department of Biochemistry, Faculty of Medicine, Suleyman Demirel University, 32260 Isparta, Turkey;
| | - Selçuk Çömlekçi
- Department of Bioengineering, Institute of Science, Suleyman Demirel University, 32260 Isparta, Turkey; (R.T.); (S.Ç.)
- Department of Electronics and Communication Engineering, Faculty of Engineering, Suleyman Demirel University, 32260 Isparta, Turkey
| |
Collapse
|
4
|
Troise D, Allegra C, Cirolla LA, Mercuri S, Infante B, Castellano G, Stallone G. Exploring Potential Complement Modulation Strategies for Ischemia-Reperfusion Injury in Kidney Transplantation. Antioxidants (Basel) 2025; 14:66. [PMID: 39857400 PMCID: PMC11761266 DOI: 10.3390/antiox14010066] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2024] [Revised: 01/03/2025] [Accepted: 01/06/2025] [Indexed: 01/27/2025] Open
Abstract
The complement system plays a crucial role in regulating the inflammatory responses in kidney transplantation, potentially contributing to early decline in kidney function. Ischemia-reperfusion injury (IRI) is among the factors affecting graft outcomes and a primary contributor to delayed graft function. Complement activation, particularly the alternative pathway, participates in the pathogenesis of IRI, involving all kidney compartments. In particular, tubular epithelial cells often acquire a dysfunctional phenotype that can exacerbate complement activation and kidney damage. Currently, complement-modulating drugs are under investigation for the treatment of kidney diseases. Many of these drugs have shown potential therapeutic benefits, but no effective clinical treatments for renal IRI have been identified yet. In this review, we will explore drugs that target complement factors, complement receptors, and regulatory proteins, aiming to highlight their potential value in improving the management of renal IRI.
Collapse
Affiliation(s)
- Dario Troise
- Nephrology, Dialysis and Transplantation Unit, Advanced Research Center on Kidney Aging (A.R.K.A.), Department of Medical and Surgical Sciences, University of Foggia, 71122 Foggia, Italy
- Division of Renal Medicine and Baxter Novum, Department of Clinical Science, Intervention and Technology, Karolinska Institutet, 141 52 Stockholm, Sweden
| | - Costanza Allegra
- Nephrology, Dialysis and Transplantation Unit, Advanced Research Center on Kidney Aging (A.R.K.A.), Department of Medical and Surgical Sciences, University of Foggia, 71122 Foggia, Italy
| | - Luciana Antonia Cirolla
- Nephrology, Dialysis and Transplantation Unit, Advanced Research Center on Kidney Aging (A.R.K.A.), Department of Medical and Surgical Sciences, University of Foggia, 71122 Foggia, Italy
| | - Silvia Mercuri
- Nephrology, Dialysis and Transplantation Unit, Advanced Research Center on Kidney Aging (A.R.K.A.), Department of Medical and Surgical Sciences, University of Foggia, 71122 Foggia, Italy
| | - Barbara Infante
- Nephrology, Dialysis and Transplantation Unit, Advanced Research Center on Kidney Aging (A.R.K.A.), Department of Medical and Surgical Sciences, University of Foggia, 71122 Foggia, Italy
| | - Giuseppe Castellano
- Unit of Nephrology, Dialysis and Transplantation, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico di Milano, 20122 Milan, Italy
| | - Giovanni Stallone
- Nephrology, Dialysis and Transplantation Unit, Advanced Research Center on Kidney Aging (A.R.K.A.), Department of Medical and Surgical Sciences, University of Foggia, 71122 Foggia, Italy
| |
Collapse
|
5
|
Janosevic D, De Luca T, Eadon MT. The Kidney Precision Medicine Project and Single-Cell Biology of the Injured Proximal Tubule. THE AMERICAN JOURNAL OF PATHOLOGY 2025; 195:7-22. [PMID: 39332674 PMCID: PMC11686451 DOI: 10.1016/j.ajpath.2024.09.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/26/2024] [Revised: 08/29/2024] [Accepted: 09/11/2024] [Indexed: 09/29/2024]
Abstract
Single-cell RNA sequencing (scRNA-seq) has led to major advances in our understanding of proximal tubule subtypes in health and disease. The proximal tubule serves essential functions in overall homeostasis, but pathologic or physiological perturbations can affect its transcriptomic signature and corresponding tasks. These alterations in proximal tubular cells are often described within a scRNA-seq atlas as cell states, which are pathophysiological subclassifications based on molecular and morphologic changes in a cell's response to that injury compared with its native state. This review describes the major cell states defined in the Kidney Precision Medicine Project's scRNA-seq atlas. It then identifies the overlap between the Kidney Precision Medicine Project and other seminal works that may use different nomenclature or cluster proximal tubule cells at different resolutions to define cell state subtypes. The goal is for the reader to understand the key transcriptomic markers of important cellular injury and regeneration processes across this highly dynamic and evolving field.
Collapse
Affiliation(s)
- Danielle Janosevic
- Division of Nephrology, Indiana University School of Medicine, Indianapolis, Indiana
| | - Thomas De Luca
- Division of Nephrology, Indiana University School of Medicine, Indianapolis, Indiana
| | - Michael T Eadon
- Division of Nephrology, Indiana University School of Medicine, Indianapolis, Indiana.
| |
Collapse
|
6
|
Lyu D, Fu S. Association between platelet count and neonatal acute kidney injury: a cohort study using the medical information mart for intensive care III database. J Matern Fetal Neonatal Med 2024; 37:2379910. [PMID: 39043458 DOI: 10.1080/14767058.2024.2379910] [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: 10/31/2023] [Accepted: 06/04/2024] [Indexed: 07/25/2024]
Abstract
OBJECTIVE A decrease in platelet count has been reported to be associated with several neonatal inflammatory diseases, including sepsis and necrotizing enterocolitis; while its association with neonatal acute kidney injury (AKI) has not been reported. This study aims to explore the association between platelet count and neonatal AKI. METHODS This was a retrospective cohort study based on the Medical Information Mart for Intensive Care III (MIMIC-III) database. Data were extracted based on baseline characteristics, comorbidities, vital signs, laboratory parameters, and intervention measures. Logistic regression analysis was used to assess the association between platelet count and AKI, and results were shown as odds ratios (OR) with 95% confidence intervals (CI). RESULTS A total of 1,576 neonates were finally included in the analysis. After adjusting birth weight, sepsis, patent ductus arteriosus, hematocrit, percentage of neutrophils, and vasopressor use, we found that platelet count in the lowest quartile (Q1) was significantly associated with the higher odds of AKI than platelet count in the highest quartile (Q4) (OR = 1.70, 95% CI: 1.01-2.87). CONCLUSIONS Low platelet count was associated with the high odds of AKI in the neonatal intensive care unit (NICU), indicating that platelet count might be a biomarker for neonatal AKI. Large-scale multicenter studies should be performed to verify the results.
Collapse
Affiliation(s)
- Dianyi Lyu
- The First College of Clinical Medical Science, China Three Gorges University, Yichang, P.R. China
- Department of Pediatrics, Yichang Central People's Hospital, Yichang, P.R. China
| | - Shufang Fu
- The First College of Clinical Medical Science, China Three Gorges University, Yichang, P.R. China
- Department of Pediatrics, Yichang Central People's Hospital, Yichang, P.R. China
| |
Collapse
|
7
|
Choi JY, Kwon H, Kim H, Hong KT, Ma Y, Koh KN, Yun S, Yoo KH, Song SH, Im HJ, Kim JH, Kang HJ. Novel genomic variants influencing methotrexate delayed clearance in pediatric patients with acute lymphoblastic leukemia. Front Pharmacol 2024; 15:1480657. [PMID: 39611166 PMCID: PMC11603417 DOI: 10.3389/fphar.2024.1480657] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2024] [Accepted: 10/24/2024] [Indexed: 11/30/2024] Open
Abstract
Background Methotrexate (MTX) is the primary drug used in the treatment of pediatric acute lymphoblastic leukemia (ALL). However, some patients exhibit delayed clearance of high-dose (HD) MTX, which induces severe nephrotoxicity, mucositis, hepatotoxicity, and neurotoxicity. We sought to identify relevant variants associated with delayed clearance of HD-MTX in pediatric patients with ALL. Methods Whole-exome sequencing of germline DNA was performed in 51 Korean pediatric patients with ALL. A total of 341 HD-MTX infusion data points from 51 patients were analyzed. MTX levels and laboratory measurements reflecting toxicity outcomes were obtained. Correlations between peak serum MTX levels at 24 h and toxicity outcomes were assessed. Analyses were performed to identify variants affecting delayed MTX clearance. Results The 24 h MTX level strongly correlated with the subsequent creatinine (Cr) level. Moreover, rs2229866 in contactin 2 (CNTN2), rs200687372 in myotubularin Related Protein 9 (MTMR9), rs777260512 in polymerase iota (POLI), rs16954698 in polycystic kidney disease 1-like 2 (PKD1L2), rs117765468 in NSE1 Homolog, SMC5-SMC6 Complex Component (NSMCE1), and rs1800956 in endoglin (ENG) were identified as candidate variants associated with delayed MTX clearance. In particular, ENG rs1800956 was significantly associated with delayed MTX clearance in all analyses and PKD1L2 rs16954698 was replicated in an external dataset (phs000637.v1.p1) from the Database of Genotypes and Phenotypes (dbGaP). Conclusion This is the first whole-exome sequencing-based analysis of delayed MTX clearance in pediatric patients with ALL. ENG rs1800956 and PKD1L2 rs16954698 were found to be potentially influential variants associated with delayed MTX clearance. These findings provide insights into HD-MTX-induced nephrotoxicity and may contribute to reducing adverse reactions through treatment modification.
Collapse
Affiliation(s)
- Jung Yoon Choi
- Department of Pediatrics, Seoul National University Children’s Hospital, Seoul National University College of Medicine, Seoul, Republic of Korea
- Seoul National University Cancer Research Institute, Seoul, Republic of Korea
| | - Hoshik Kwon
- Seoul National University Biomedical Informatics (SNUBI), Division of Biomedical Informatics, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Hyery Kim
- Department of Pediatrics, Asan Medical Center Children’s Hospital, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Kyung Taek Hong
- Department of Pediatrics, Seoul National University Children’s Hospital, Seoul National University College of Medicine, Seoul, Republic of Korea
- Seoul National University Cancer Research Institute, Seoul, Republic of Korea
| | - Youngeun Ma
- Department of Pediatrics, Seoul Metropolitan Children’s Hospital, Seoul, Republic of Korea
| | - Kyung-Nam Koh
- Department of Pediatrics, Asan Medical Center Children’s Hospital, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Sunmin Yun
- Seoul National University Biomedical Informatics (SNUBI), Division of Biomedical Informatics, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Keon Hee Yoo
- Department of Pediatrics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Sang Hoon Song
- Department of Laboratory Medicine, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Ho Joon Im
- Department of Pediatrics, Asan Medical Center Children’s Hospital, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Ju Han Kim
- Seoul National University Biomedical Informatics (SNUBI), Division of Biomedical Informatics, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Hyoung Jin Kang
- Department of Pediatrics, Seoul National University Children’s Hospital, Seoul National University College of Medicine, Seoul, Republic of Korea
- Seoul National University Cancer Research Institute, Seoul, Republic of Korea
| |
Collapse
|
8
|
Wei Q, Huang J, Livingston MJ, Wang S, Dong G, Xu H, Zhou J, Dong Z. Pseudogene GSTM3P1 derived long non-coding RNA promotes ischemic acute kidney injury by target directed microRNA degradation of kidney-protective mir-668. Kidney Int 2024; 106:640-657. [PMID: 39074555 PMCID: PMC11416318 DOI: 10.1016/j.kint.2024.06.027] [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: 10/17/2023] [Revised: 05/21/2024] [Accepted: 06/17/2024] [Indexed: 07/31/2024]
Abstract
Long non-coding RNAs (lncRNAs) are a group of epigenetic regulators that have been implicated in kidney diseases including acute kidney injury (AKI). However, very little is known about the specific lncRNAs involved in AKI and the mechanisms underlying their pathologic roles. Here, we report a new lncRNA derived from the pseudogene GSTM3P1, which mediates ischemic AKI by interacting with and promoting the degradation of mir-668, a kidney-protective microRNA. GSTM3P1 and its mouse orthologue Gstm2-ps1 were induced by hypoxia in cultured kidney proximal tubular cells. In mouse kidneys, Gstm2-ps1 was significantly upregulated in proximal tubules at an early stage of ischemic AKI. This transient induction of Gstm2-ps1 depends on G3BP1, a key component in stress granules. GSTM3P1 overexpression increased kidney proximal tubular apoptosis after ATP depletion, which was rescued by mir-668. Notably, kidney proximal tubule-specific knockout of Gstm2-ps1 protected mice from ischemic AKI, as evidenced by improved kidney function, diminished tubular damage and apoptosis, and reduced kidney injury biomarker (NGAL) induction. To test the therapeutic potential, Gstm2-ps1 siRNAs were introduced into cultured mouse proximal tubular cells or administered to mice. In cultured cells, Gstm2-ps1 knockdown suppressed ATP depletion-associated apoptosis. In mice, Gstm2-ps1 knockdown ameliorated ischemic AKI. Mechanistically, both GSTM3P1 and Gstm2-ps1 possessed mir-668 binding sites and downregulated the mature form of mir-668. Specifically, GSTM3P1 directly bound to mature mir-668 to induce its decay via target-directed microRNA degradation. Thus, our results identify GSTM3P1 as a novel lncRNA that promotes kidney tubular cell death in AKI by binding mir-668 to inducing its degradation.
Collapse
Affiliation(s)
- Qingqing Wei
- Department of Cellular Biology and Anatomy, Medical College of Georgia, Augusta University, Augusta, Georgia, USA.
| | - Jing Huang
- Department of Cellular Biology and Anatomy, Medical College of Georgia, Augusta University, Augusta, Georgia, USA; Department of Nephrology, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| | - Man Jiang Livingston
- Department of Cellular Biology and Anatomy, Medical College of Georgia, Augusta University, Augusta, Georgia, USA
| | - Shixuan Wang
- Department of Cellular Biology and Anatomy, Medical College of Georgia, Augusta University, Augusta, Georgia, USA
| | - Guie Dong
- Department of Cellular Biology and Anatomy, Medical College of Georgia, Augusta University, Augusta, Georgia, USA
| | - Hongyan Xu
- Department of Biostatistics, Data Science and Epidemiology, School of Public Health, Augusta University, Augusta, Georgia, USA
| | - Jiliang Zhou
- Department of Pharmacology and Toxicology, Medical College of Georgia, Augusta University, Augusta, Georgia, USA
| | - Zheng Dong
- Department of Cellular Biology and Anatomy, Medical College of Georgia, Augusta University, Augusta, Georgia, USA; Charlie Norwood VA Medical Center, Augusta, Georgia, USA.
| |
Collapse
|
9
|
Hu Y, Lei Y, Yu M, Zhang Y, Huang X, Zhang G, Deng Q. Ultrasound super-resolution imaging for the assessment of renal allograft dysfunction: A pilot study. Heliyon 2024; 10:e36515. [PMID: 39247269 PMCID: PMC11380004 DOI: 10.1016/j.heliyon.2024.e36515] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Revised: 08/16/2024] [Accepted: 08/16/2024] [Indexed: 09/10/2024] Open
Abstract
Background The purpose of this study was to examine the feasibility and practical application of ultrasound (US) super-resolution imaging (SRI) in evaluating microvasculature and measuring renal allograft function. Methods Sixteen consecutive patients who received kidney transplants were prospectively enrolled. The patients were assigned as: normal allograft function (n = 6), and allograft malfunction (n = 10). Localizing each potential contrast signal resulted in super-resolution images (SRI). SRI was utilized to assess micro-vessel density (MVD) and microvascular flow rate, whereas contrast-enhanced (CE) US images were statistically processed to get the time to peak (TTP) and peak intensity. Logistic regression was utilized to evaluate their relationship. Results US SRI may be utilized effectively on allografts to show microvasculature with significantly higher resolution than typical color Doppler flow and CEUS pictures. In the multivariate analysis, MVD and TTP were significant US markers of renal allograft failure (p = 0.031 and p = 0.045). The combination of MVD and TTP produced an AUC of 0.783 (p < 0.05) for allograft dysfunction. Conclusions SRI can accurately portray the microvasculature of renal allografts, while MVD and TTP are appropriate US markers for assessing renal allograft failure.
Collapse
Affiliation(s)
- Yugang Hu
- Department of Ultrasound Imaging, Renmin Hospital of Wuhan University, Wuhan, 430061, China
| | - Yumeng Lei
- Department of Medical Ultrasound, China Resources & Wisco General Hospital, Wuhan University of Science and Technology, Wuhan, 430080, China
| | - Meihui Yu
- Department of Medical Ultrasound, China Resources & Wisco General Hospital, Wuhan University of Science and Technology, Wuhan, 430080, China
| | - Yao Zhang
- Department of Ultrasound Imaging, Renmin Hospital of Wuhan University, Wuhan, 430061, China
| | - Xingyue Huang
- Department of Ultrasound Imaging, Renmin Hospital of Wuhan University, Wuhan, 430061, China
| | - Ge Zhang
- Department of Medical Ultrasound, China Resources & Wisco General Hospital, Wuhan University of Science and Technology, Wuhan, 430080, China
| | - Qing Deng
- Department of Ultrasound Imaging, Renmin Hospital of Wuhan University, Wuhan, 430061, China
| |
Collapse
|
10
|
Nigussie EW, Abera EG, Woldemariam MB. Burden and determinants of renal dysfunction and in-hospital mortality among acute stroke patients in Ethiopia: A hospital-based observational study. Medicine (Baltimore) 2024; 103:e39140. [PMID: 39058854 PMCID: PMC11272293 DOI: 10.1097/md.0000000000039140] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/05/2024] [Accepted: 07/09/2024] [Indexed: 07/28/2024] Open
Abstract
Stroke, a leading global cause of mortality and neurological impairment, is often complicated by renal failure, exacerbating in-hospital risks and mortality. Limited understanding exists regarding renal failure prevalence in Ethiopian acute stroke patients. This study examines renal function abnormalities in acute stroke patients at Jimma Medical Center (JMC). A hospital-based cross-sectional study was conducted at JMC from December 5, 2023, to March 15, 2024. A structured data collection tool was developed after comprehensive review of pertinent literature, encompassing variables pertinent to the study objectives. Following data quality assurance, information was coded and inputted into EpiData version 3.1, subsequently analyzed using Statistical Package for Social Sciences (SPSS) version 26.0. Multivariable logistic regression analysis was performed to adjust for confounding variables, with statistical significance set at P < .05. The mean age of participants was 60.5 ± 15.5 years, with 129 (64.5%) being male. Forty-five participants (22.5%, 95% confidence interval [CI] = 16.9, 28.9) exhibited renal dysfunction. Advanced age (≥70 years), hypertension, diabetes mellitus (DM), cardiac disease, history of transient ischemic attack (TIA)/stroke, and hemorrhagic stroke type were identified as significant predictors of renal dysfunction among hospitalized stroke patients. The mortality rate was 3.7 times higher in stroke patients with renal dysfunction compared to those with normal renal function (adjusted odds ratio [AOR] = 3.7, 95% CI: 1.41, 6.22). Renal function abnormalities were prevalent among hospitalized acute stroke patients, emphasizing the significance of renal dysfunction as a frequent comorbidity. Older age, hypertension, DM, cardiac disease, history of TIA/stroke, and hemorrhagic stroke type emerged as statistically significant predictors of renal dysfunction. Furthermore, renal dysfunction was identified as a significant predictor of in-hospital mortality following stroke.
Collapse
Affiliation(s)
| | - Eyob Girma Abera
- Department of Public Health, Jimma University, Jimma, Oromia, Ethiopia
- Jimma University Clinical Trial Unit, Jimma, Oromia, Ethiopia
| | | |
Collapse
|
11
|
Patel NS, Herzog I, Dunn C, Merchant AM. Impact of Operative Approach on Acute Kidney Injury Risk Prediction Models for Colectomy. J Surg Res 2024; 299:224-236. [PMID: 38776578 DOI: 10.1016/j.jss.2024.04.026] [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: 09/24/2023] [Revised: 04/07/2024] [Accepted: 04/21/2024] [Indexed: 05/25/2024]
Abstract
INTRODUCTION Acute kidney injury (AKI) is a serious postoperative complication associated with increased morbidity and mortality. Identifying patients at risk for AKI is important for risk stratification and management. This study aimed to develop an AKI risk prediction model for colectomy and determine if the operative approach (laparoscopic versus open) alters the influence of predictive factors through an interaction term analysis. METHODS The American College of Surgeons National Surgical Quality Improvement Program database was analyzed from 2005 to 2019. Patients undergoing laparoscopic and open colectomy were identified and propensity score matched. Multivariable logistic regression identified significant preoperative demographic, comorbidity, and laboratory value predictors of AKI. The predictive ability of a baseline model consisting of these variables was compared to a proposed model incorporating interaction terms between operative approach and predictor variables using the likelihood ratio test, c-statistic, and Brier score. Shapley Additive Explanations values assessed relative importance of significant predictors. RESULTS 252,372 patients were included in the analysis. Significant AKI predictors were hypertension, age, sex, race, body mass index, smoking, diabetes, preoperative sepsis, Congestive heart failure, preoperative creatinine, preoperative albumin, and operative approach (P < 0.001). The proposed model with interaction terms had improved predictive ability per the likelihood ratio test (P < 0.05) but had no statistically significant interaction terms. C-statistic and Brier scores did not improve. Shapley Additive Explanations analysis showed hypertension had the highest importance. The importance of age and diabetes showed some variation between operative approaches. CONCLUSIONS While the inclusion of interaction terms collectively improved AKI prediction, no individual operative approach interaction terms were significant. Including operative approach interactions may enhance predictive ability of AKI risk models for colectomy.
Collapse
Affiliation(s)
| | - Isabel Herzog
- Rutgers New Jersey Medical School, Newark, New Jersey
| | - Colin Dunn
- Department of Surgery, Good Samaritan Hospital, San Jose, California
| | - Aziz M Merchant
- Rutgers New Jersey Medical School, Newark, New Jersey; Division of General and Minimally Invasive Surgery, Department of Surgery, Hackensack Meridian School of Medicine, JFK Hackensack Meridian Medical Center, Edison, New Jersey.
| |
Collapse
|
12
|
De Beule J, De Craemer S, Verstraeten L, Ghesquière B, Jochmans I. Ischemia-induced Metabolic Patterns Associate With Kidney Function During Normothermic Kidney Perfusion: A Preclinical Study. Ann Surg 2024; 280:156-164. [PMID: 37870241 DOI: 10.1097/sla.0000000000006137] [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: 10/24/2023]
Abstract
OBJECTIVE To investigate whether ischemia alters donor kidney metabolism and whether these changes are associated with organ function. BACKGROUND An unmet need in kidney transplantation is the ability to predict posttransplant organ function before transplantation. Key to such viability testing is a profound understanding of the organ's complex biochemistry and how ischemia, inevitable during the transplantation process, influences this. METHODS First, metabolic changes in perfusate glucose, lactate, and 20 amino acids, induced by no, 1 hour of warm, or 22 hours of cold ischemia, were investigated during 4-hour perfusion of pig kidneys with autologous whole blood (n = 6/group), simulating the ischemia-reperfusion phase of transplantation. Next, we confirmed similar metabolic changes during normothermic preservation of pigs (n = 3/group; n = 4 for cold ischemia) and discarded human kidneys (n = 6) perfused with a red blood cell-based perfusate. RESULTS At 2 hours of perfusion with autologous whole blood, abundances of 17/20 amino acids were significantly different between groups, reflecting the type of ischemia. Amino acid changes at 15 minutes and 2 hours of perfusion correlated with future kidney function during perfusion. Similar metabolic patterns were observed during perfusion preservation of pig and discarded human donor kidneys, suggesting an opportunity to assess kidney viability before transplantation. CONCLUSIONS Perfusate metabolite changes during normothermic kidney perfusion represent a unique noninvasive opportunity to assess graft viability. These findings now need validation in transplant studies.
Collapse
Affiliation(s)
- Julie De Beule
- Department of Microbiology, Immunology, and Transplantation, Laboratory of Abdominal Transplantation, Transplantation Research Group, KU Leuven, Leuven, Belgium
- Center for Cancer Biology, Metabolomics Core Facility Leuven, VIB, Leuven, Belgium
| | - Sam De Craemer
- Center for Cancer Biology, Metabolomics Core Facility Leuven, VIB, Leuven, Belgium
| | - Laurence Verstraeten
- Department of Microbiology, Immunology, and Transplantation, Laboratory of Abdominal Transplantation, Transplantation Research Group, KU Leuven, Leuven, Belgium
- Center for Cancer Biology, Metabolomics Core Facility Leuven, VIB, Leuven, Belgium
| | - Bart Ghesquière
- Center for Cancer Biology, Metabolomics Core Facility Leuven, VIB, Leuven, Belgium
- Department of Cellular and Molecular Medicine, Laboratory of Applied Mass Spectrometry, KU Leuven, Leuven, Belgium
| | - Ina Jochmans
- Department of Microbiology, Immunology, and Transplantation, Laboratory of Abdominal Transplantation, Transplantation Research Group, KU Leuven, Leuven, Belgium
- Department of Abdominal Transplant Surgery, University Hospitals Leuven, Leuven, Belgium
| |
Collapse
|
13
|
Huang X, Zhang Y, Zhou Q, Deng Q. Value of Ultrasound Super-Resolution Imaging for the Assessment of Renal Microcirculation in Patients with Acute Kidney Injury: A Preliminary Study. Diagnostics (Basel) 2024; 14:1192. [PMID: 38893718 PMCID: PMC11171740 DOI: 10.3390/diagnostics14111192] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2024] [Revised: 05/31/2024] [Accepted: 05/31/2024] [Indexed: 06/21/2024] Open
Abstract
The present study aimed to explore the clinical applicability of ultrasound super-resolution imaging (US SRI) for assessing renal microcirculation in patients with acute kidney injury (AKI). A total of 62 patients with sepsis were enrolled in the present study-38 with AKI and 24 control patients-from whom renal ultrasounds and clinical data were obtained. SonoVue contrast (1.5 mL) was administered through the elbow vein and contrast-enhanced ultrasound (CEUS) images were obtained on a Mindray Resona A20 ultrasound unit for 2 min. The renal perfusion time-intensity curve (TIC) was analyzed and, after 15 min, additional images were obtained to create a microscopic blood flow map. Microvascular density (MVD) was calculated and its correlation with serum creatinine (Scr) levels was analyzed. There were significant differences in heart rate, Scr, blood urea nitrogen, urine volume at 24 h, and glomerular filtration rate between the two groups (p < 0.01), whereas other characteristics, such as renal morphology, did not differ significantly between the AKI group and control group (p > 0.05). The time to peak and mean transit times of the renal cortex in the AKI group were prolonged compared to those in the control group (p < 0.01), while the peak intensity and area under the TIC were lower than those in the control group (p < 0.05). The MVD of the renal cortex in the AKI group was lower than that in the control group (18.46 ± 5.90% vs. 44.93 ± 11.65%; p < 0.01) and the MVD in the AKI group showed a negative correlation with Scr (R = -0.84; p < 0.01). Based on the aforementioned results, US SRI can effectively assess renal microcirculation in patients with AKI and is a noninvasive technique for the diagnosis of AKI and quantitative evaluation of renal microcirculation.
Collapse
Affiliation(s)
| | | | - Qing Zhou
- Department of Ultrasound, Renmin Hospital of Wuhan University, Wuhan 430060, China; (X.H.); (Y.Z.)
| | - Qing Deng
- Department of Ultrasound, Renmin Hospital of Wuhan University, Wuhan 430060, China; (X.H.); (Y.Z.)
| |
Collapse
|
14
|
Bostancı H, Erel S, Küçük A, Kip G, Sezen ŞC, Gokgoz S, Atlı M, Aktepe F, Dikmen K, Arslan M, Kavutçu M. Dexmedetomidine's Effects on the Livers and Kidneys of Rats with Pancreatic Ischemia-Reperfusion Injury. Drug Des Devel Ther 2024; 18:1785-1797. [PMID: 38828020 PMCID: PMC11141764 DOI: 10.2147/dddt.s441773] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2023] [Accepted: 05/13/2024] [Indexed: 06/05/2024] Open
Abstract
Objective Pancreatic surgeries inherently cause ischemia-reperfusion (IR) injury, affecting not only the pancreas but also distant organs. This study was conducted to explore the potential use of dexmedetomidine, a sedative with antiapoptotic, anti-inflammatory, and antioxidant properties, in mitigating the impacts of pancreatic IR on kidney and liver tissues. Methods A total of 24 rats were randomly divided into four groups: control (C), dexmedetomidine (D), ischemia reperfusion (IR), and dexmedetomidine ischemia reperfusion (D-IR). Pancreatic ischemia was induced in the IR and D-IR groups. Dexmedetomidine was administered intraperitoneally to the D and D-IR groups. Liver and kidney tissue samples were subjected to microscopic examinations after hematoxylin and eosin staining. The levels of thiobarbituric acid reactive substances (TBARS), aryllesterase (AES), catalase (CAT), and glutathione S-transferase (GST) enzyme activity were assessed in liver and kidney tissues. The serum levels of aspartate aminotransferase (AST), alanine aminotransferase (ALT), blood urea nitrogen (BUN), and creatinine were measured. Results A comparison of the groups revealed that the IR group exhibited significantly elevated TBARS (p < 0.0001), AES (p = 0.004), and CAT enzyme activity (p < 0.0001) levels in the liver and kidney compared to groups C and D. Group D-IR demonstrated notably reduced histopathological damage (p < 0.05) and low TBARS (p < 0.0001), AES (p = 0.004), and CAT enzyme activity (p < 0.0001) in the liver and kidney as well as low AST and ALT activity levels (p < 0.0001) in the serum compared to the IR group. Conclusion The preemptive administration of dexmedetomidine before pancreatic IR provides significant protection to kidney and liver tissues, as evidenced by the histopathological and biochemical parameters in this study. The findings underscored the potential therapeutic role of dexmedetomidine in mitigating the multiorgan damage associated with pancreatic surgeries.
Collapse
Affiliation(s)
- Hasan Bostancı
- Gazi University Faculty of Medicine, Department of General Surgery, Ankara, Turkey
| | - Selin Erel
- Gazi University Faculty of Medicine Department of Anesthesiology and Reanimation, Ankara, Turkey
| | - Ayşegül Küçük
- Kutahya Health Sciences University Faculty of Medicine, Department of Physiology, Kutahya, Turkey
| | - Gülay Kip
- Gazi University Faculty of Medicine Department of Anesthesiology and Reanimation, Ankara, Turkey
| | - Şaban Cem Sezen
- Kırıkkale University Faculty of Medicine, Department of Histology and Embryology, Kırıkkale, Turkey
| | - Seda Gokgoz
- Gazi University Faculty of Medicine, Department of Medical Biochemistry, Ankara, Turkey
| | - Muharrem Atlı
- Kırıkkale University Faculty of Medicine, Department of Histology and Embryology, Kırıkkale, Turkey
| | - Feyza Aktepe
- Gazi University Faculty of Medicine Department of Anesthesiology and Reanimation, Ankara, Turkey
| | - Kursat Dikmen
- Gazi University Faculty of Medicine, Department of General Surgery, Ankara, Turkey
| | - Mustafa Arslan
- Gazi University Faculty of Medicine Department of Anesthesiology and Reanimation, Ankara, Turkey
- Gazi University, Life Sciences Application and Research Center, Ankara, Turkey
- Gazi University, Laboratory Animal Breeding and Experimental Research Center (GUDAM), Ankara, Turkey
| | - Mustafa Kavutçu
- Gazi University Faculty of Medicine, Department of Medical Biochemistry, Ankara, Turkey
| |
Collapse
|
15
|
Saadh MJ, Mahdi MS, Allela OQB, Alazzawi TS, Ubaid M, Rakhimov NM, Athab ZH, Ramaiah P, Chinnasamy L, Alsaikhan F, Farhood B. Critical role of miR-21/exosomal miR-21 in autophagy pathway. Pathol Res Pract 2024; 257:155275. [PMID: 38643552 DOI: 10.1016/j.prp.2024.155275] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/28/2024] [Revised: 03/22/2024] [Accepted: 03/26/2024] [Indexed: 04/23/2024]
Abstract
Activation of autophagy, a process of cellular stress response, leads to the breakdown of proteins, organelles, and other parts of the cell in lysosomes, and can be linked to several ailments, such as cancer, neurological diseases, and rare hereditary syndromes. Thus, its regulation is very carefully monitored. Transcriptional and post-translational mechanisms domestically or in whole organisms utilized to control the autophagic activity, have been heavily researched. In modern times, microRNAs (miRNAs) are being considered to have a part in post-translational orchestration of the autophagic activity, with miR-21 as one of the best studied miRNAs, it is often more than expressed in cancer cells. This regulatory RNA is thought to play a major role in a plethora of processes and illnesses including growth, cancer, cardiovascular disease, and inflammation. Different studies have suggested that a few autophagy-oriented genes, such as PTEN, Rab11a, Atg12, SIPA1L2, and ATG5, are all targeted by miR-21, indicating its essential role in the regulation.
Collapse
Affiliation(s)
- Mohamed J Saadh
- Faculty of Pharmacy, Middle East University, Amman 11831, Jordan
| | | | | | - Tuqa S Alazzawi
- College of dentist, National University of Science and Technology, Dhi Qar, Iraq
| | | | - Nodir M Rakhimov
- Department of Oncology, Samarkand State Medical University, 18 Amir Temur Street, Samarkand, Uzbekistan; Department of Oncology, Tashkent State Dental Institute, Tashkent, Uzbekistan
| | - Zainab H Athab
- Department of Pharmacy, Al-Zahrawi University College, Karbala, Iraq
| | | | | | - Fahad Alsaikhan
- College of Pharmacy, Prince Sattam Bin Abdulaziz University, Alkharj, Saudi Arabia jSchool of Pharmacy, Ibn Sina National College for Medical Studies, Jeddah, Saudi Arabia.
| | - Bagher Farhood
- Department of Medical Physics and Radiology, Faculty of Paramedical Sciences, Kashan University of Medical Sciences, Kashan, Iran.
| |
Collapse
|
16
|
Nakamura E, Aoki T, Endo Y, Kazmi J, Hagiwara J, Kuschner CE, Yin T, Kim J, Becker LB, Hayashida K. Organ-Specific Mitochondrial Alterations Following Ischemia-Reperfusion Injury in Post-Cardiac Arrest Syndrome: A Comprehensive Review. Life (Basel) 2024; 14:477. [PMID: 38672748 PMCID: PMC11050834 DOI: 10.3390/life14040477] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2024] [Revised: 04/01/2024] [Accepted: 04/03/2024] [Indexed: 04/28/2024] Open
Abstract
BACKGROUND Mitochondrial dysfunction, which is triggered by systemic ischemia-reperfusion (IR) injury and affects various organs, is a key factor in the development of post-cardiac arrest syndrome (PCAS). Current research on PCAS primarily addresses generalized mitochondrial responses, resulting in a knowledge gap regarding organ-specific mitochondrial dynamics. This review focuses on the organ-specific mitochondrial responses to IR injury, particularly examining the brain, heart, and kidneys, to highlight potential therapeutic strategies targeting mitochondrial dysfunction to enhance outcomes post-IR injury. METHODS AND RESULTS We conducted a narrative review examining recent advancements in mitochondrial research related to IR injury. Mitochondrial responses to IR injury exhibit considerable variation across different organ systems, influenced by unique mitochondrial structures, bioenergetics, and antioxidative capacities. Each organ demonstrates distinct mitochondrial behaviors that have evolved to fulfill specific metabolic and functional needs. For example, cerebral mitochondria display dynamic responses that can be both protective and detrimental to neuronal activity and function during ischemic events. Cardiac mitochondria show vulnerability to IR-induced oxidative stress, while renal mitochondria exhibit a unique pattern of fission and fusion, closely linked to their susceptibility to acute kidney injury. This organ-specific heterogeneity in mitochondrial responses requires the development of tailored interventions. Progress in mitochondrial medicine, especially in the realms of genomics and metabolomics, is paving the way for innovative strategies to combat mitochondrial dysfunction. Emerging techniques such as mitochondrial transplantation hold the potential to revolutionize the management of IR injury in resuscitation science. CONCLUSIONS The investigation into organ-specific mitochondrial responses to IR injury is pivotal in the realm of resuscitation research, particularly within the context of PCAS. This nuanced understanding holds the promise of revolutionizing PCAS management, addressing the unique mitochondrial dysfunctions observed in critical organs affected by IR injury.
Collapse
Affiliation(s)
- Eriko Nakamura
- Laboratory for Critical Care Physiology, Feinstein Institutes for Medical Research, Northwell Health System, Manhasset, NY 11030, USA; (E.N.); (T.A.); (Y.E.); (J.K.); (J.H.); (C.E.K.); (T.Y.); (J.K.); (L.B.B.)
| | - Tomoaki Aoki
- Laboratory for Critical Care Physiology, Feinstein Institutes for Medical Research, Northwell Health System, Manhasset, NY 11030, USA; (E.N.); (T.A.); (Y.E.); (J.K.); (J.H.); (C.E.K.); (T.Y.); (J.K.); (L.B.B.)
| | - Yusuke Endo
- Laboratory for Critical Care Physiology, Feinstein Institutes for Medical Research, Northwell Health System, Manhasset, NY 11030, USA; (E.N.); (T.A.); (Y.E.); (J.K.); (J.H.); (C.E.K.); (T.Y.); (J.K.); (L.B.B.)
| | - Jacob Kazmi
- Laboratory for Critical Care Physiology, Feinstein Institutes for Medical Research, Northwell Health System, Manhasset, NY 11030, USA; (E.N.); (T.A.); (Y.E.); (J.K.); (J.H.); (C.E.K.); (T.Y.); (J.K.); (L.B.B.)
| | - Jun Hagiwara
- Laboratory for Critical Care Physiology, Feinstein Institutes for Medical Research, Northwell Health System, Manhasset, NY 11030, USA; (E.N.); (T.A.); (Y.E.); (J.K.); (J.H.); (C.E.K.); (T.Y.); (J.K.); (L.B.B.)
| | - Cyrus E. Kuschner
- Laboratory for Critical Care Physiology, Feinstein Institutes for Medical Research, Northwell Health System, Manhasset, NY 11030, USA; (E.N.); (T.A.); (Y.E.); (J.K.); (J.H.); (C.E.K.); (T.Y.); (J.K.); (L.B.B.)
- Department of Emergency Medicine, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, NY 11549, USA
| | - Tai Yin
- Laboratory for Critical Care Physiology, Feinstein Institutes for Medical Research, Northwell Health System, Manhasset, NY 11030, USA; (E.N.); (T.A.); (Y.E.); (J.K.); (J.H.); (C.E.K.); (T.Y.); (J.K.); (L.B.B.)
| | - Junhwan Kim
- Laboratory for Critical Care Physiology, Feinstein Institutes for Medical Research, Northwell Health System, Manhasset, NY 11030, USA; (E.N.); (T.A.); (Y.E.); (J.K.); (J.H.); (C.E.K.); (T.Y.); (J.K.); (L.B.B.)
| | - Lance B. Becker
- Laboratory for Critical Care Physiology, Feinstein Institutes for Medical Research, Northwell Health System, Manhasset, NY 11030, USA; (E.N.); (T.A.); (Y.E.); (J.K.); (J.H.); (C.E.K.); (T.Y.); (J.K.); (L.B.B.)
- Department of Emergency Medicine, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, NY 11549, USA
| | - Kei Hayashida
- Laboratory for Critical Care Physiology, Feinstein Institutes for Medical Research, Northwell Health System, Manhasset, NY 11030, USA; (E.N.); (T.A.); (Y.E.); (J.K.); (J.H.); (C.E.K.); (T.Y.); (J.K.); (L.B.B.)
- Department of Emergency Medicine, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, NY 11549, USA
| |
Collapse
|
17
|
Kharazmi F, Hosseini-Dastgerdi H, Pourshanazari AA, Nematbakhsh M. The denervation or activation of renal sympathetic nerve and renal blood flow. JOURNAL OF RESEARCH IN MEDICAL SCIENCES : THE OFFICIAL JOURNAL OF ISFAHAN UNIVERSITY OF MEDICAL SCIENCES 2023; 28:76. [PMID: 38152073 PMCID: PMC10751519 DOI: 10.4103/jrms.jrms_216_23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Revised: 07/09/2023] [Accepted: 07/17/2023] [Indexed: 12/29/2023]
Abstract
The denervation or activation of the sympathetic nerve in the kidney can affect renal hemodynamics. The sympathetic nervous system regulates the physiological functions of the kidneys. Stimulation of sympathetic efferent nerves affects various parameters related to renal hemodynamics, including sodium excretion, renin secretion, and renal blood flow (RBF). Hence, renal sympathetic fibers may also play an essential role in regulating systemic vascular resistance and controlling blood pressure. In the absence of renal nerves, the hemodynamics response to stimuli is negligible or absent. The effect of renal sympathetic denervation on RBF is dependent on several factors such as interspecies differences, the basic level of nerve activity in the vessels or local density of adrenergic receptor in the vascular bed. The role of renal denervation has been investigated therapeutically in hypertension and related disorders. Hence, the dynamic impact of renal nerves on RBF enables using RBF dynamic criteria as a marker for renal denervation therapy.
Collapse
Affiliation(s)
- Fatemeh Kharazmi
- Water and Electrolytes Research Center, Isfahan University of Medical Sciences, Isfahan, Iran
- Department of Physiology, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Hajaralsadat Hosseini-Dastgerdi
- Water and Electrolytes Research Center, Isfahan University of Medical Sciences, Isfahan, Iran
- Department of Physiology, Isfahan University of Medical Sciences, Isfahan, Iran
| | | | - Mehdi Nematbakhsh
- Water and Electrolytes Research Center, Isfahan University of Medical Sciences, Isfahan, Iran
- Department of Physiology, Isfahan University of Medical Sciences, Isfahan, Iran
| |
Collapse
|
18
|
Gomchok D, Ge RL, Wuren T. Platelets in Renal Disease. Int J Mol Sci 2023; 24:14724. [PMID: 37834171 PMCID: PMC10572297 DOI: 10.3390/ijms241914724] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Revised: 09/18/2023] [Accepted: 09/26/2023] [Indexed: 10/15/2023] Open
Abstract
Kidney disease is a major global health concern, affecting millions of people. Nephrologists have shown interest in platelets because of coagulation disorders caused by renal diseases. With a better understanding of platelets, it has been found that these anucleate and abundant blood cells not only play a role in hemostasis, but also have important functions in inflammation and immunity. Platelets are not only affected by kidney disease, but may also contribute to kidney disease progression by mediating inflammation and immune effects. This review summarizes the current evidence regarding platelet abnormalities in renal disease, and the multiple effects of platelets on kidney disease progression. The relationship between platelets and kidney disease is still being explored, and further research can provide mechanistic insights into the relationship between thrombosis, bleeding, and inflammation related to kidney disease, and elucidate targeted therapies for patients with kidney disease.
Collapse
Affiliation(s)
- Drolma Gomchok
- Research Center for High Altitude Medicine, School of Medicine, Qinghai University, Xining 810001, China; (D.G.); (R.-L.G.)
| | - Ri-Li Ge
- Research Center for High Altitude Medicine, School of Medicine, Qinghai University, Xining 810001, China; (D.G.); (R.-L.G.)
- Key Laboratory for Application for High Altitude Medicine, Qinghai University, Xining 810001, China
| | - Tana Wuren
- Research Center for High Altitude Medicine, School of Medicine, Qinghai University, Xining 810001, China; (D.G.); (R.-L.G.)
- Key Laboratory for Application for High Altitude Medicine, Qinghai University, Xining 810001, China
| |
Collapse
|
19
|
Morgan AM, Ogaly HA, Kamel S, Rashad MM, Hassanen EI, Ibrahim MA, Galal MK, Yassin AM, Dulmani SAA, Al-Zahrani FA, Hussien AM. Protective effects of N-acetyl-l-cysteine against penconazole-triggered hepatorenal toxicity in adult rats. J Vet Res 2023; 67:459-469. [PMID: 37786839 PMCID: PMC10541664 DOI: 10.2478/jvetres-2023-0039] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2022] [Accepted: 06/26/2023] [Indexed: 10/04/2023] Open
Abstract
Introduction Penconazole (PEN) is a widely applied triazole fungicide. This study sought to define the efficacy of N-acetyl-l-cysteine (NAC) in mitigating PEN-triggered hepatorenal toxicity in rats. Material and Methods Twenty-eight adult male albino Wistar rats were assigned to four groups: a normal control (NC), a PEN group, a NAC group and a PEN+NAC group. Administration of PEN (50 mg/kg body weight (b.w.) every 2 days) and NAC (150 mg/kg b.w., daily) took place via oral gavage for 10 days. Results Effective amelioration by NAC of PEN-induced liver and kidney dysfunction was indicated by a significant reduction in the circulating liver and kidney markers (aspartate aminotransferase, alanine aminotransferase, urea and creatinine). Attenuation of PEN-induced oxidative stress and lipid peroxidation in liver and kidney tissues was evident in a significant reduction in malondialdehyde and enhanced total antioxidant capacity. Moreover, NAC significantly reduced the histopathological alterations and the expression of tumour necrosis factor α in liver and kidney tissue. Furthermore, NAC maintained the messenger RNA levels of nuclear factor erythroid 2-related factor 2 (Nrf2), haem oxygenase 1, and Kelch-like erythroid cell-derived protein 1 and prevented nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB) protein upregulation caused by PEN. Conclusion N-acetyl-1-cysteine protected against PEN-induced hepatorenal oxidative damage and inflammatory response via activation of Nrf2 and inhibition of NF-κB pathways.
Collapse
Affiliation(s)
| | - Hanan A. Ogaly
- Chemistry Department, Faculty of Science, King Khalid University, Abha 62421, Abha High City, Saudi Arabia
| | - Shaimaa Kamel
- Biochemistry and Molecular Biology Department, 12211Giza, Egypt
| | - Maha M. Rashad
- Biochemistry and Molecular Biology Department, 12211Giza, Egypt
| | - Eman I. Hassanen
- Pathology Department, Faculty of Veterinary Medicine, Cairo University, 12211Giza, Egypt
| | | | - Mona K. Galal
- Biochemistry and Molecular Biology Department, 12211Giza, Egypt
| | - Aya M. Yassin
- Biochemistry and Molecular Biology Department, 12211Giza, Egypt
| | - Sharah A. Al Dulmani
- Chemistry Department, Faculty of Science, King Khalid University, Abha 62421, Abha High City, Saudi Arabia
| | - Fatimah A.M. Al-Zahrani
- Chemistry Department, Faculty of Science, King Khalid University, Abha 62421, Abha High City, Saudi Arabia
| | | |
Collapse
|
20
|
Bordoni L, Kristensen AM, Sardella D, Kidmose H, Pohl L, Krag SRP, Schiessl IM. Longitudinal tracking of acute kidney injury reveals injury propagation along the nephron. Nat Commun 2023; 14:4407. [PMID: 37479698 PMCID: PMC10362041 DOI: 10.1038/s41467-023-40037-y] [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: 01/17/2023] [Accepted: 07/10/2023] [Indexed: 07/23/2023] Open
Abstract
Acute kidney injury (AKI) is an important risk factor for chronic kidney disease (CKD), but the underlying mechanisms of failed tubule repair and AKI-CKD transition are incompletely understood. In this study, we aimed for dynamic tracking of tubule injury and remodeling to understand if focal injury upon AKI may spread over time. Here, we present a model of AKI, in which we rendered only half of the kidney ischemic. Using serial intravital 2-photon microscopy and genetic identification of cycling cells, we tracked dynamic tissue remodeling in post- and non-ischemic kidney regions simultaneously and over 3 weeks. Spatial and temporal analysis of cycling cells relative to initial necrotic cell death demonstrated pronounced injury propagation and expansion into non-necrotic tissue regions, which predicted tubule atrophy with epithelial VCAM1 expression. In summary, our longitudinal analyses of tubule injury, remodeling, and fate provide important insights into AKI pathology.
Collapse
Affiliation(s)
- Luca Bordoni
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
- GliaLab and Letten Centre, Division of Anatomy, Department of Molecular Medicine, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway
| | | | - Donato Sardella
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
| | - Hanne Kidmose
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
| | - Layla Pohl
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
| | | | | |
Collapse
|
21
|
McElliott MC, Al-Suraimi A, Telang AC, Ference-Salo JT, Chowdhury M, Soofi A, Dressler GR, Beamish JA. High-throughput image analysis with deep learning captures heterogeneity and spatial relationships after kidney injury. Sci Rep 2023; 13:6361. [PMID: 37076596 PMCID: PMC10115810 DOI: 10.1038/s41598-023-33433-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Accepted: 04/12/2023] [Indexed: 04/21/2023] Open
Abstract
Recovery from acute kidney injury can vary widely in patients and in animal models. Immunofluorescence staining can provide spatial information about heterogeneous injury responses, but often only a fraction of stained tissue is analyzed. Deep learning can expand analysis to larger areas and sample numbers by substituting for time-intensive manual or semi-automated quantification techniques. Here we report one approach to leverage deep learning tools to quantify heterogenous responses to kidney injury that can be deployed without specialized equipment or programming expertise. We first demonstrated that deep learning models generated from small training sets accurately identified a range of stains and structures with performance similar to that of trained human observers. We then showed this approach accurately tracks the evolution of folic acid induced kidney injury in mice and highlights spatially clustered tubules that fail to repair. We then demonstrated that this approach captures the variation in recovery across a robust sample of kidneys after ischemic injury. Finally, we showed markers of failed repair after ischemic injury were correlated both spatially within and between animals and that failed repair was inversely correlated with peritubular capillary density. Combined, we demonstrate the utility and versatility of our approach to capture spatially heterogenous responses to kidney injury.
Collapse
Affiliation(s)
- Madison C McElliott
- Division of Nephrology, Department of Internal Medicine, University of Michigan, 1500 E. Medical Center Drive, SPC 5364, Ann Arbor, MI, 48109, USA
| | - Anas Al-Suraimi
- Division of Nephrology, Department of Internal Medicine, University of Michigan, 1500 E. Medical Center Drive, SPC 5364, Ann Arbor, MI, 48109, USA
| | - Asha C Telang
- Division of Nephrology, Department of Internal Medicine, University of Michigan, 1500 E. Medical Center Drive, SPC 5364, Ann Arbor, MI, 48109, USA
| | - Jenna T Ference-Salo
- Division of Nephrology, Department of Internal Medicine, University of Michigan, 1500 E. Medical Center Drive, SPC 5364, Ann Arbor, MI, 48109, USA
| | - Mahboob Chowdhury
- Division of Nephrology, Department of Internal Medicine, University of Michigan, 1500 E. Medical Center Drive, SPC 5364, Ann Arbor, MI, 48109, USA
| | - Abdul Soofi
- Department of Pathology, University of Michigan, Ann Arbor, MI, USA
| | | | - Jeffrey A Beamish
- Division of Nephrology, Department of Internal Medicine, University of Michigan, 1500 E. Medical Center Drive, SPC 5364, Ann Arbor, MI, 48109, USA.
| |
Collapse
|
22
|
Lee B, Kang W, Oh SH, Cho S, Shin I, Oh EJ, Kim YJ, Ahn JS, Yook JM, Jung SJ, Lim JH, Kim YL, Cho JH, Oh WY. In vivo imaging of renal microvasculature in a murine ischemia-reperfusion injury model using optical coherence tomography angiography. Sci Rep 2023; 13:6396. [PMID: 37076541 PMCID: PMC10115874 DOI: 10.1038/s41598-023-33295-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2022] [Accepted: 04/11/2023] [Indexed: 04/21/2023] Open
Abstract
Optical coherence tomography angiography (OCTA) provides three-dimensional structural and semiquantitative imaging of microvasculature in vivo. We developed an OCTA imaging protocol for a murine kidney ischemia-reperfusion injury (IRI) model to investigate the correlation between renal microvascular changes and ischemic damage. Mice were divided into mild and moderate IRI groups according to the duration of ischemia (10 and 35 mins, respectively). Each animal was imaged at baseline; during ischemia; and at 1, 15, 30, 45, and 60 mins after ischemia. Amplitude decorrelation OCTA images were constructed with 1.5-, 3.0-, and 5.8-ms interscan times, to calculate the semiquantitative flow index in the superficial (50-70 μm) and the deep (220-340 μm) capillaries of the renal cortex. The mild IRI group showed no significant flow index change in both the superfial and the deep layers. The moderate IRI group showed a significantly decreased flow index from 15 and 45 mins in the superficial and deep layers, respectively. Seven weeks after IRI induction, the moderate IRI group showed lower kidney function and higher collagen deposition than the mild IRI group. OCTA imaging of the murine IRI model revealed changes in superficial blood flow after ischemic injury. A more pronounced decrease in superficial blood flow than in deep blood flow was associated with sustained dysfunction after IRI. Further investigation on post-IRI renal microvascular response using OCTA may improve our understanding of the relationship between the degree of ischemic insult and kidney function.
Collapse
Affiliation(s)
- ByungKun Lee
- Department of Mechanical Engineering, KAIST, Daejeon, Republic of Korea
- KI for Health Science and Technology, KAIST, Daejeon, Republic of Korea
| | - Woojae Kang
- Department of Mechanical Engineering, KAIST, Daejeon, Republic of Korea
- KI for Health Science and Technology, KAIST, Daejeon, Republic of Korea
| | - Se-Hyun Oh
- Division of Nephrology, Department of Internal Medicine, Kyungpook National University Hospital, Daegu, Republic of Korea
- Cell and Matrix Research Institute, Kyungpook National University, Daegu, Republic of Korea
| | - Seungwan Cho
- Department of Mechanical Engineering, KAIST, Daejeon, Republic of Korea
- KI for Health Science and Technology, KAIST, Daejeon, Republic of Korea
| | - Inho Shin
- Department of Mechanical Engineering, KAIST, Daejeon, Republic of Korea
- KI for Health Science and Technology, KAIST, Daejeon, Republic of Korea
| | - Eun-Joo Oh
- Division of Nephrology, Department of Internal Medicine, Kyungpook National University Hospital, Daegu, Republic of Korea
| | - You-Jin Kim
- Division of Nephrology, Department of Internal Medicine, Kyungpook National University Hospital, Daegu, Republic of Korea
- Cell and Matrix Research Institute, Kyungpook National University, Daegu, Republic of Korea
| | - Ji-Sun Ahn
- Division of Nephrology, Department of Internal Medicine, Kyungpook National University Hospital, Daegu, Republic of Korea
| | - Ju-Min Yook
- Division of Nephrology, Department of Internal Medicine, Kyungpook National University Hospital, Daegu, Republic of Korea
| | - Soo-Jung Jung
- Division of Nephrology, Department of Internal Medicine, Kyungpook National University Hospital, Daegu, Republic of Korea
- Cell and Matrix Research Institute, Kyungpook National University, Daegu, Republic of Korea
| | - Jeong-Hoon Lim
- Division of Nephrology, Department of Internal Medicine, Kyungpook National University Hospital, Daegu, Republic of Korea
- Cell and Matrix Research Institute, Kyungpook National University, Daegu, Republic of Korea
| | - Yong-Lim Kim
- Division of Nephrology, Department of Internal Medicine, Kyungpook National University Hospital, Daegu, Republic of Korea
- Cell and Matrix Research Institute, Kyungpook National University, Daegu, Republic of Korea
| | - Jang-Hee Cho
- Division of Nephrology, Department of Internal Medicine, Kyungpook National University Hospital, Daegu, Republic of Korea.
- Cell and Matrix Research Institute, Kyungpook National University, Daegu, Republic of Korea.
| | - Wang-Yuhl Oh
- Department of Mechanical Engineering, KAIST, Daejeon, Republic of Korea.
- KI for Health Science and Technology, KAIST, Daejeon, Republic of Korea.
| |
Collapse
|
23
|
Ultrasound localization microscopy of the human kidney allograft on a clinical ultrasound scanner. Kidney Int 2023; 103:930-935. [PMID: 36841476 DOI: 10.1016/j.kint.2023.01.027] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2022] [Revised: 01/18/2023] [Accepted: 01/27/2023] [Indexed: 02/27/2023]
Abstract
Chronic kidney disease is a major medical problem, causing more than a million deaths each year worldwide. Peripheral kidney microvascular damage characterizes most chronic kidney diseases, yet noninvasive and quantitative diagnostic tools to measure this are lacking. Ultrasound Localization Microscopy (ULM) can assess tissue microvasculature with unprecedented resolution. Here, we optimized methods on 35 kidney transplants and studied the feasibility of ULM in seven human kidney allografts with a standard low frame rate ultrasound scanner to access microvascular damage. Interlobar, arcuate, cortical radial vessels, and part of the medullary organization were visible on ULM density maps. The medullary vasa recta can be seen but are not as clear as the cortical vessels. Acquisition parameters were derived from Contrast-Enhanced Ultrasound examinations by increasing the duration of the recorded clip at the same plane. ULM images were compared with Color Doppler, Advanced Dynamic Flow, and Superb Microvascular Imaging with a contrast agent. Despite some additional limitations due to movement and saturation artifacts, ULM identified vessels two to four times thinner compared with Doppler modes. The mean ULM smallest analyzable vessel cross section was 0.3 ± 0.2 mm in the seven patients. Additionally, ULM was able to provide quantitative information on blood velocities in the cortical area. Thus, this proof-of-concept study has shown ULM to be a promising imaging technique for qualitative and quantitative microvascular assessment. Imaging native kidneys in patients with kidney diseases will be needed to identify their ULM biomarkers.
Collapse
|
24
|
Wang S, Wang Y, Lai X, Sun J, Hu M, Chen M, Li C, Xu F, Fan C, Liu X, Song Y, Chen G, Deng Y. Minimalist Nanocomplex with Dual Regulation of Endothelial Function and Inflammation for Targeted Therapy of Inflammatory Vascular Diseases. ACS NANO 2023; 17:2761-2781. [PMID: 36719043 DOI: 10.1021/acsnano.2c11058] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/18/2023]
Abstract
Vascular disorders, characterized by vascular endothelial dysfunction combined with inflammation, are correlated with numerous fatal diseases, such as coronavirus disease-19 and atherosclerosis. Achieving vascular normalization is an urgent problem that must be solved when treating inflammatory vascular diseases. Inspired by the vascular regulatory versatility of nitric oxide (NO) produced by endothelial nitric oxide synthase (eNOS) catalyzing l-arginine (l-Arg), the eNOS-activating effects of l-Arg, and the powerful anti-inflammatory and eNOS-replenishing effects of budesonide (BUD), we constructed a bi-prodrug minimalist nanoplatform co-loaded with BUD and l-Arg via polysialic acid (PSA) to form BUD-l-Arg@PSA. This promoted vascular normalization by simultaneously regulating vascular endothelial dysfunction and inflammation. Mediated by the special affinity between PSA and E-selectin, which is highly expressed on the surface of activated endothelial cells (ECs), BUD-l-Arg@PSA selectively accumulated in activated ECs, targeted eNOS expression and activation, and promoted NO production. Consequently, the binary synergistic regulation of the NO/eNOS signaling pathway occurred and improved vascular endothelial function. NO-induced nuclear factor-kappa B alpha inhibitor (IκBα) stabilization and BUD-induced nuclear factor-kappa B (NF-κB) response gene site occupancy achieved dual-site blockade of the NF-κB signaling pathway, thereby reducing the inflammatory response and inhibiting the infiltration of inflammation-related immune cells. In a renal ischemia-reperfusion injury mouse model, BUD-l-Arg@PSA reduced acute injury. In an atherosclerosis mouse model, BUD-l-Arg@PSA decreased atherosclerotic plaque burden and improved vasodilation. This represents a revolutionary therapeutic strategy for inflammatory vascular diseases.
Collapse
Affiliation(s)
- Shuo Wang
- College of Pharmacy, Shenyang Pharmaceutical University, Shenyang110016, China
| | - Yuequan Wang
- Department of Pharmaceutics, Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang110016, China
| | - Xiaoxue Lai
- College of Pharmacy, Shenyang Pharmaceutical University, Shenyang110016, China
| | - Jianwen Sun
- Key Laboratory of Structure-Based Drug Design and Discovery of Ministry of Education, Shenyang Pharmaceutical University, Shenyang110016, China
| | - Miao Hu
- College of Pharmacy, Shenyang Pharmaceutical University, Shenyang110016, China
| | - Meng Chen
- College of Pharmacy, Shenyang Pharmaceutical University, Shenyang110016, China
| | - Cong Li
- College of Pharmacy, Shenyang Pharmaceutical University, Shenyang110016, China
| | - Feng Xu
- College of Pharmacy, Shenyang Pharmaceutical University, Shenyang110016, China
| | - Chuizhong Fan
- College of Pharmacy, Shenyang Pharmaceutical University, Shenyang110016, China
| | - Xinrong Liu
- College of Pharmacy, Shenyang Pharmaceutical University, Shenyang110016, China
| | - Yanzhi Song
- College of Pharmacy, Shenyang Pharmaceutical University, Shenyang110016, China
| | - Guoliang Chen
- Key Laboratory of Structure-Based Drug Design and Discovery of Ministry of Education, Shenyang Pharmaceutical University, Shenyang110016, China
| | - Yihui Deng
- College of Pharmacy, Shenyang Pharmaceutical University, Shenyang110016, China
| |
Collapse
|
25
|
Desita EAN, Arfian N, Setyaningsih WAW, Sari DCR. Calcitriol attenuates vascular remodeling in association with alteration of ppET-1/ET BR/eNOS and ET AR expression in acute and chronic phases of kidney ischemia-reperfusion injury in mice. Can J Physiol Pharmacol 2023; 101:8-17. [PMID: 36223656 DOI: 10.1139/cjpp-2022-0130] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Kidney ischemia-reperfusion injury (IRI) causes acute kidney injury with increasing risk of maladaptive repair through endothelin-1 (ET-1)/endothelin type A receptor (ETAR) signaling. Calcitriol shows renoprotection in kidney fibrosis, however, its effects on vasoactive substances expression and vascular remodeling following kidney IRI remain unclear. This research aimed to investigate Calcitriol's effects on preproendothelin-1 (ppET-1), ETAR, endothelial nitric oxide synthase (eNOS) mRNA expression and vascular remodeling in acute and chronic phases of kidney IRI in mice. Twenty-five male Swiss mice were randomly divided into five groups (n = 5): SO (sham-operated), IR3 (3 day kidney IRI), IR12 (12 day kidney IRI), IRD3 (3 day kidney IRI + Calcitriol 0.5 µg/kg body weight (BW)/day), and IRD12 (12 day kidney IRI + Calcitriol 0.5 µg/kg BW/day). Ischemia-reperfusion injury groups underwent bilateral renal pedicles clamping for 30 min, then reperfusion. Kidneys were harvested for Sirius Red staining to observe interstitial fibrosis and vascular remodeling, polymerase chain reaction to quantify ppET-1, endothelin type B receptor (ETBR), eNOS mRNA expression, and Western blotting to quantify ETAR protein expression. Calcitriol treatment in both phases of kidney IRI showed lower serum creatinine and ETAR protein expression, while higher eNOS and ETBR mRNA expression than IRI-only groups. Furthermore, ppET-1 mRNA expression was higher in IRD3 than IR3, but lower in IRD12 than IR12. Calcitriol also prevented vascular remodeling as indicated by lower wall thickness and higher lumen/wall area ratio than IRI-only groups.
Collapse
Affiliation(s)
- Eryna Ayu Nugra Desita
- Department of Anatomy, Faculty of Medicine, Public Health, and Nursing, Universitas Gadjah Mada, Sleman, DI Yogyakarta, 55281, Indonesia
| | - Nur Arfian
- Department of Anatomy, Faculty of Medicine, Public Health, and Nursing, Universitas Gadjah Mada, Sleman, DI Yogyakarta, 55281, Indonesia
| | - Wiwit Ananda Wahyu Setyaningsih
- Department of Anatomy, Faculty of Medicine, Public Health, and Nursing, Universitas Gadjah Mada, Sleman, DI Yogyakarta, 55281, Indonesia
| | - Dwi Cahyani Ratna Sari
- Department of Anatomy, Faculty of Medicine, Public Health, and Nursing, Universitas Gadjah Mada, Sleman, DI Yogyakarta, 55281, Indonesia
| |
Collapse
|
26
|
Ramezanzadeh E, Fallah Arzpeyma S, Vakilpour A, Abedi M, Hassanipour S. Endothelial function assessment by flow-mediated dilation of the brachial artery in acute kidney injury and chronic kidney disease. CASPIAN JOURNAL OF INTERNAL MEDICINE 2023; 14:668-675. [PMID: 38024183 PMCID: PMC10646350 DOI: 10.22088/cjim.14.4.668] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Figures] [Subscribe] [Scholar Register] [Received: 06/01/2022] [Revised: 07/24/2022] [Accepted: 10/15/2022] [Indexed: 12/01/2023]
Abstract
Background Endothelial dysfunction has a significant role in the pathogenesis of cardiovascular events in patients with kidney dysfunction. The present study aimed to compare the level of endothelial dysfunction in patients with chronic kidney disease (CKD) and acute kidney injury (AKI) by brachial artery flow-mediated dilation (FMD) technique. Also, we sought to find whether this non-invasive technique may assist in accurately distinguishing the acute or chronic nature of kidney failure in patients presenting with uremia for the first time. Methods Demographic and medical characteristics, and laboratory and renal ultrasonography data of the patients with AKI and CKD were collected and compared with a control group. Brachial artery FMD was measured using a Toshiba aplio 300 device with a 7.5 MHz linear probe. Results In a total of 175 patients with a mean (SD) age of 55.96(15.54) years, FMD% was significantly lower in the CKD and AKI patients compared to the control group (Mean±SD: 16.28%±10.52%), 16.28 %±4.35%, and 24.24±5.71, respectively, p<0.001). Among the different causes of AKI, contrast-induced nephropathy (10.78%±1.75%), volume depletion (14.87%±1.22%), and post-renal AKI (15.96%±1.54%) had the lowest levels of FMD. Also, a significant correlation between FMD and eGFR (r=0.26, P<0.001), serum Hb (r=0.18, P=0.013), Na (r=0.19, P=0.011), BUN (r=-0.21, P=0.005) and Cr (r=- 0.13, P=0.084) was reported. Conclusion Compared to the control group, CKD and AKI patients showed greater levels of endothelial dysfunction as evidenced by lower brachial artery FMD. However, the FMD technique did not appear to be a practical method in differentiating CKD and AKI in patients presenting with uremia for the first time.
Collapse
Affiliation(s)
- Elham Ramezanzadeh
- Razi Clinic Research Development Center, Gilan University of Medical Sciences, Rasht, Iran
| | - Sima Fallah Arzpeyma
- Razi Clinic Research Development Center, Gilan University of Medical Sciences, Rasht, Iran
| | - Azin Vakilpour
- Cardiovascular Diseases Research Center, Department of Cardiology, Heshmat Hospital, School of Medicine, Gilan University of Medical Sciences, Rasht, Iran
| | - Mohammadsadegh Abedi
- Razi Clinic Research Development Center, Gilan University of Medical Sciences, Rasht, Iran
| | - Soheil Hassanipour
- Cardiovascular Diseases Research Center, Department of Cardiology, Heshmat Hospital, School of Medicine, Gilan University of Medical Sciences, Rasht, Iran
| |
Collapse
|
27
|
Wang Y, Li X, Xu X, Yu J, Chen X, Cao X, Zou J, Shen B, Ding X. Clec7a expression in inflammatory macrophages orchestrates progression of acute kidney injury. Front Immunol 2022; 13:1008727. [PMID: 36189317 PMCID: PMC9520532 DOI: 10.3389/fimmu.2022.1008727] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Accepted: 09/02/2022] [Indexed: 12/04/2022] Open
Abstract
Acute kidney injury (AKI) is associated with high risk of mortality, post-disease renal fibrosis, kidney dysfunction and renal failure. Renal macrophages play a key role in the pathogenesis (M1 subpopulation), healing and remodeling (M2 subpopulation) in AKI and, thus, have been a promising target for clinical treatment of AKI. Here, in a mouse renal ischemia/reperfusion injury (IRI) model for AKI, we showed that renal macrophages could be further classified into Clec7a+ M1 macrophages, Clec7a- M1 macrophages, Clec7a+ M2 macrophages and Clec7a- M2 macrophages, representing distinct macrophage populations with different functionality. Interestingly, Clec7a+ M1 macrophages exhibited potent pro-inflammatory and phagocytic effects compared to Clec7a- M1 macrophages, while Clec7a- M2 macrophages exhibited better proliferating and migrating potential, which is critical for their role in tissue repairing after injury. These data from mice were further strengthened by bioinformatics analyses using published database. In vivo, combined expression of Clec7a in M1 macrophages and depletion of Clec7a in M2 macrophages significantly improved the renal function after IRI-AKI. Together, our data suggest that Clec7a is crucial for the fine regulation of macrophage phenotype during AKI and could be a novel target for boosting clinical therapy.
Collapse
Affiliation(s)
- Yaqiong Wang
- Department of Nephrology, Zhongshan Hospital, Fudan University, Shanghai, China
- Shanghai Medical Center of Kidney, Shanghai, China
- Department of Nephrology, Shanghai Institute of Kidney and Dialysis, Shanghai, China
- Shanghai Key Laboratory of Kidney and Blood Purification, Shanghai, China
- Hemodialysis Quality Control Center of Shanghai, Shanghai, China
| | - Xianzhe Li
- Department of Nephrology, Zhongshan Hospital, Fudan University, Shanghai, China
- Shanghai Medical Center of Kidney, Shanghai, China
- Department of Nephrology, Shanghai Institute of Kidney and Dialysis, Shanghai, China
- Shanghai Key Laboratory of Kidney and Blood Purification, Shanghai, China
- Hemodialysis Quality Control Center of Shanghai, Shanghai, China
| | - Xialian Xu
- Department of Nephrology, Zhongshan Hospital, Fudan University, Shanghai, China
- Shanghai Medical Center of Kidney, Shanghai, China
- Department of Nephrology, Shanghai Institute of Kidney and Dialysis, Shanghai, China
- Shanghai Key Laboratory of Kidney and Blood Purification, Shanghai, China
- Hemodialysis Quality Control Center of Shanghai, Shanghai, China
| | - Jinbo Yu
- Department of Nephrology, Zhongshan Hospital, Fudan University, Shanghai, China
- Shanghai Medical Center of Kidney, Shanghai, China
- Department of Nephrology, Shanghai Institute of Kidney and Dialysis, Shanghai, China
- Shanghai Key Laboratory of Kidney and Blood Purification, Shanghai, China
- Hemodialysis Quality Control Center of Shanghai, Shanghai, China
| | - Xiaohong Chen
- Department of Nephrology, Zhongshan Hospital, Fudan University, Shanghai, China
- Shanghai Medical Center of Kidney, Shanghai, China
- Department of Nephrology, Shanghai Institute of Kidney and Dialysis, Shanghai, China
- Shanghai Key Laboratory of Kidney and Blood Purification, Shanghai, China
- Hemodialysis Quality Control Center of Shanghai, Shanghai, China
| | - Xuesen Cao
- Department of Nephrology, Zhongshan Hospital, Fudan University, Shanghai, China
- Shanghai Medical Center of Kidney, Shanghai, China
- Department of Nephrology, Shanghai Institute of Kidney and Dialysis, Shanghai, China
- Shanghai Key Laboratory of Kidney and Blood Purification, Shanghai, China
- Hemodialysis Quality Control Center of Shanghai, Shanghai, China
| | - Jianzhou Zou
- Department of Nephrology, Zhongshan Hospital, Fudan University, Shanghai, China
- Shanghai Medical Center of Kidney, Shanghai, China
- Department of Nephrology, Shanghai Institute of Kidney and Dialysis, Shanghai, China
- Shanghai Key Laboratory of Kidney and Blood Purification, Shanghai, China
- Hemodialysis Quality Control Center of Shanghai, Shanghai, China
| | - Bo Shen
- Department of Nephrology, Zhongshan Hospital, Fudan University, Shanghai, China
- Shanghai Medical Center of Kidney, Shanghai, China
- Department of Nephrology, Shanghai Institute of Kidney and Dialysis, Shanghai, China
- Shanghai Key Laboratory of Kidney and Blood Purification, Shanghai, China
- Hemodialysis Quality Control Center of Shanghai, Shanghai, China
- *Correspondence: Xiaoqiang Ding, ; Bo Shen,
| | - Xiaoqiang Ding
- Department of Nephrology, Zhongshan Hospital, Fudan University, Shanghai, China
- Shanghai Medical Center of Kidney, Shanghai, China
- Department of Nephrology, Shanghai Institute of Kidney and Dialysis, Shanghai, China
- Shanghai Key Laboratory of Kidney and Blood Purification, Shanghai, China
- Hemodialysis Quality Control Center of Shanghai, Shanghai, China
- *Correspondence: Xiaoqiang Ding, ; Bo Shen,
| |
Collapse
|
28
|
Wazir S, Abbas M, Ratanasrimetha P, Zhang C, Hariharan S, Puttarajappa CM. Preoperative blood pressure and risk of delayed graft function in deceased donor kidney transplantation. Clin Transplant 2022; 36:e14776. [PMID: 35821617 DOI: 10.1111/ctr.14776] [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: 01/10/2022] [Revised: 05/05/2022] [Accepted: 07/08/2022] [Indexed: 11/28/2022]
Abstract
BACKGROUND In kidney transplantation, delayed graft function (DGF) is associated with increased morbidity and a higher risk of graft failure. Prior research suggests that chronic hypotension increases DGF risk, but the relationship of preoperative blood pressure to DGF is unclear. METHODS In this single center study of adult deceased donor kidney transplant recipients transplanted between 2015 and 2019, we evaluated the question of whether preoperative mean arterial pressure (MAP) affected DGF risk. Additionally, we investigated whether the risk of DGF was moderated by certain donor and recipient characteristics. For recipient characteristics associated with increased DGF risk and preoperative MAP, we performed a mediation analysis to estimate the proportion of DGF risk mediated through preoperative MAP. RESULTS Among 562 deceased donor kidney recipients, DGF risk decreased as preoperative MAP increased, with a 2% lower risk per 1 mm Hg increase in MAP. This increased risk was similar, with no statistically significant interaction effect between preoperative MAP and donor (donation after circulatory death) and recipient characteristics (diabetes, body mass index, and use of anti-hypertensive medications). Preoperative MAP was negativity correlated with recipient BMI and duration of pre transplant dialysis. On mediation analysis, MAP accounted for 12% and 16% of the DGF risk associated with recipient BMI and pre-transplant dialysis duration, respectively. CONCLUSION In deceased donor kidney transplantation, each 1 mm Hg increase in preoperative MAP was associated with 2% lower DGF risk. Preoperative MAP was influenced by recipient BMI and dialysis duration, and likely contributes to some of the high DGF risk from obesity and long dialysis vintage.
Collapse
Affiliation(s)
- Shoaib Wazir
- Department of Medicine, Renal-Electrolyte Division, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Muhammad Abbas
- Department of Medicine, Renal-Electrolyte Division, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Praveen Ratanasrimetha
- Department of Medicine, Renal-Electrolyte Division, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Casey Zhang
- School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Sundaram Hariharan
- Department of Medicine, Renal-Electrolyte Division, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Chethan M Puttarajappa
- Department of Medicine, Renal-Electrolyte Division, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| |
Collapse
|
29
|
Ghajar-Rahimi G, Agarwal A. Endothelial KLF11 as a Nephroprotectant in AKI. KIDNEY360 2022; 3:1302-1305. [PMID: 36176668 PMCID: PMC9416841 DOI: 10.34067/kid.0003422022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/09/2022] [Accepted: 06/14/2022] [Indexed: 06/16/2023]
Affiliation(s)
- Gelare Ghajar-Rahimi
- Division of Nephrology, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama
- Nephrology Research and Training Center, University of Alabama at Birmingham, Birmingham, Alabama
- Division of Cardiothoracic Surgery, Department of Surgery, University of Alabama at Birmingham, Birmingham, Alabama
| | - Anupam Agarwal
- Division of Nephrology, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama
- Birmingham Veterans Administration Health Care Services, Birmingham, Alabama
| |
Collapse
|
30
|
Basile DP. Crystals or His(stones): Rethinking AKI in Tumor Lysis Syndrome. J Am Soc Nephrol 2022; 33:1055-1057. [PMID: 35641305 PMCID: PMC9161801 DOI: 10.1681/asn.2022040425] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022] Open
Affiliation(s)
- David P Basile
- Department of Anatomy, Cell Biology, and Physiology, Indiana University of Medicine, Indianapolis, Indiana
| |
Collapse
|
31
|
McLarnon SR, Wilson K, Patel B, Sun J, Sartain CL, Mejias CD, Musall JB, Sullivan JC, Wei Q, Chen JK, Hyndman KA, Marshall B, Yang H, Fogo AB, O’Connor PM. Lipopolysaccharide Pretreatment Prevents Medullary Vascular Congestion following Renal Ischemia by Limiting Early Reperfusion of the Medullary Circulation. J Am Soc Nephrol 2022; 33:769-785. [PMID: 35115326 PMCID: PMC8970460 DOI: 10.1681/asn.2021081089] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2021] [Accepted: 01/16/2022] [Indexed: 11/03/2022] Open
Abstract
BACKGROUND Vascular congestion of the renal medulla-trapped red blood cells in the medullary microvasculature-is a hallmark finding at autopsy in patients with ischemic acute tubular necrosis. Despite this, the pathogenesis of vascular congestion is not well defined. METHODS In this study, to investigate the pathogenesis of vascular congestion and its role in promoting renal injury, we assessed renal vascular congestion and tubular injury after ischemia reperfusion in rats pretreated with low-dose LPS or saline (control). We used laser Doppler flowmetry to determine whether pretreatment with low-dose LPS prevented vascular congestion by altering renal hemodynamics during reperfusion. RESULTS We found that vascular congestion originated during the ischemic period in the renal venous circulation. In control animals, the return of blood flow was followed by the development of congestion in the capillary plexus of the outer medulla and severe tubular injury early in reperfusion. Laser Doppler flowmetry indicated that blood flow returned rapidly to the medulla, several minutes before recovery of full cortical perfusion. In contrast, LPS pretreatment prevented both the formation of medullary congestion and its associated tubular injury. Laser Doppler flowmetry in LPS-pretreated rats suggested that limiting early reperfusion of the medulla facilitated this protective effect, because it allowed cortical perfusion to recover and clear congestion from the large cortical veins, which also drain the medulla. CONCLUSIONS Blockage of the renal venous vessels and a mismatch in the timing of cortical and medullary reperfusion results in congestion of the outer medulla's capillary plexus and promotes early tubular injury after renal ischemia. These findings indicate that hemodynamics during reperfusion contribute to the renal medulla's susceptibility to ischemic injury.
Collapse
Affiliation(s)
- Sarah R. McLarnon
- Department of Physiology, Medical College of Georgia at Augusta University, Augusta, Georgia
| | - Katie Wilson
- Department of Physiology, Medical College of Georgia at Augusta University, Augusta, Georgia
| | - Bansari Patel
- Department of Physiology, Medical College of Georgia at Augusta University, Augusta, Georgia
| | - Jingping Sun
- Department of Physiology, Medical College of Georgia at Augusta University, Augusta, Georgia
| | - Christina L. Sartain
- Department of Physiology, Medical College of Georgia at Augusta University, Augusta, Georgia
| | - Christopher D. Mejias
- Department of Physiology, Medical College of Georgia at Augusta University, Augusta, Georgia
| | - Jacqueline B. Musall
- Department of Physiology, Medical College of Georgia at Augusta University, Augusta, Georgia
| | - Jennifer C. Sullivan
- Department of Physiology, Medical College of Georgia at Augusta University, Augusta, Georgia
| | - Qingqing Wei
- Department of Cell Biology and Anatomy, Medical College of Georgia at Augusta University, Augusta, Georgia
| | - Jian-Kang Chen
- Department of Cell Biology and Anatomy, Medical College of Georgia at Augusta University, Augusta, Georgia
| | - Kelly A. Hyndman
- Cardio-Renal Physiology and Medicine, Division of Nephrology, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama
| | - Brendan Marshall
- Department of Cell Biology and Anatomy, Medical College of Georgia at Augusta University, Augusta, Georgia
| | - Haichun Yang
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Agnes B. Fogo
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Paul M. O’Connor
- Department of Physiology, Medical College of Georgia at Augusta University, Augusta, Georgia
| |
Collapse
|
32
|
Becker F, Kebschull L, Rieger C, Mohr A, Heitplatz B, Van Marck V, Hansen U, Ansari J, Reuter S, Strücker B, Pascher A, Brockmann JG, Castor T, Alexander JS, Gavins FNE. Bryostatin-1 Attenuates Ischemia-Elicited Neutrophil Transmigration and Ameliorates Graft Injury after Kidney Transplantation. Cells 2022; 11:cells11060948. [PMID: 35326400 PMCID: PMC8946580 DOI: 10.3390/cells11060948] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2022] [Revised: 03/04/2022] [Accepted: 03/09/2022] [Indexed: 12/19/2022] Open
Abstract
Ischemia reperfusion injury (IRI) is a form of sterile inflammation whose severity determines short- and long-term graft fates in kidney transplantation. Neutrophils are now recognized as a key cell type mediating early graft injury, which activates further innate immune responses and intensifies acquired immunity and alloimmunity. Since the macrolide Bryostatin-1 has been shown to block neutrophil transmigration, we aimed to determine whether these findings could be translated to the field of kidney transplantation. To study the effects of Bryostatin-1 on ischemia-elicited neutrophil transmigration, an in vitro model of hypoxia and normoxia was equipped with human endothelial cells and neutrophils. To translate these findings, a porcine renal autotransplantation model with eight hours of reperfusion was used to study neutrophil infiltration in vivo. Graft-specific treatment using Bryostatin-1 (100 nM) was applied during static cold storage. Bryostatin-1 dose-dependently blocked neutrophil activation and transmigration over ischemically challenged endothelial cell monolayers. When applied to porcine renal autografts, Bryostatin-1 reduced neutrophil graft infiltration, attenuated histological and ultrastructural damage, and improved renal function. Our novel findings demonstrate that Bryostatin-1 is a promising pharmacological candidate for graft-specific treatment in kidney transplantation, as it provides protection by blocking neutrophil infiltration and attenuating functional graft injury.
Collapse
Affiliation(s)
- Felix Becker
- Department of General, Visceral and Transplant Surgery, University Hospital Münster, 48149 Münster, Germany; (F.B.); (L.K.); (C.R.); (A.M.); (B.S.); (A.P.); (J.G.B.)
| | - Linus Kebschull
- Department of General, Visceral and Transplant Surgery, University Hospital Münster, 48149 Münster, Germany; (F.B.); (L.K.); (C.R.); (A.M.); (B.S.); (A.P.); (J.G.B.)
| | - Constantin Rieger
- Department of General, Visceral and Transplant Surgery, University Hospital Münster, 48149 Münster, Germany; (F.B.); (L.K.); (C.R.); (A.M.); (B.S.); (A.P.); (J.G.B.)
| | - Annika Mohr
- Department of General, Visceral and Transplant Surgery, University Hospital Münster, 48149 Münster, Germany; (F.B.); (L.K.); (C.R.); (A.M.); (B.S.); (A.P.); (J.G.B.)
| | - Barbara Heitplatz
- Gerhard Domagk Institute of Pathology, University Hospital Münster, 48149 Münster, Germany; (B.H.); (V.V.M.)
| | - Veerle Van Marck
- Gerhard Domagk Institute of Pathology, University Hospital Münster, 48149 Münster, Germany; (B.H.); (V.V.M.)
| | - Uwe Hansen
- Department of Molecular Medicine, Institute for Musculoskeletal Medicine, University Hospital Münster, 48149 Münster, Germany;
| | - Junaid Ansari
- Department of Neurology, Louisiana State University Health Sciences Center, Shreveport, LA 71130, USA;
| | - Stefan Reuter
- Division of General Internal Medicine, Nephrology and Rheumatology, Department of Medicine D, University Hospital of Münster, 48149 Münster, Germany;
| | - Benjamin Strücker
- Department of General, Visceral and Transplant Surgery, University Hospital Münster, 48149 Münster, Germany; (F.B.); (L.K.); (C.R.); (A.M.); (B.S.); (A.P.); (J.G.B.)
| | - Andreas Pascher
- Department of General, Visceral and Transplant Surgery, University Hospital Münster, 48149 Münster, Germany; (F.B.); (L.K.); (C.R.); (A.M.); (B.S.); (A.P.); (J.G.B.)
| | - Jens G. Brockmann
- Department of General, Visceral and Transplant Surgery, University Hospital Münster, 48149 Münster, Germany; (F.B.); (L.K.); (C.R.); (A.M.); (B.S.); (A.P.); (J.G.B.)
| | | | - J. Steve Alexander
- Department of Neurology, Louisiana State University Health Sciences Center, Shreveport, LA 71130, USA;
- Department of Molecular and Cellular Physiology, Louisiana State University Health Sciences Center, Shreveport, LA 71130, USA
- Correspondence: (J.S.A.); (F.N.E.G.)
| | - Felicity N. E. Gavins
- Department of Life Sciences, Centre for Inflammation Research and Translational Medicine (CIRTM), Brunel University London, Uxbridge UB8 3PH, UK
- Correspondence: (J.S.A.); (F.N.E.G.)
| |
Collapse
|
33
|
Vascular Endothelial Dysfunction in the Thoracic Aorta of Rats with Ischemic Acute Kidney Injury: Contribution of Indoxyl Sulfate. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:7547269. [PMID: 35251481 PMCID: PMC8896937 DOI: 10.1155/2022/7547269] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Revised: 01/21/2022] [Accepted: 02/09/2022] [Indexed: 01/04/2023]
Abstract
Chronic kidney disease (CKD) and cardiovascular disease are known to be linked, and the involvement of indoxyl sulfate (IS), a type of uremic toxin, has been suggested as one of the causes. It is known that IS induces vascular dysfunction through overproduction of reactive oxygen species (ROS). On the other hand, the involvement of IS in the vascular dysfunction associated with acute kidney injury (AKI) is not fully understood. Therefore, we investigated this issue using the thoracic aorta of rats with ischemic AKI. Ischemic AKI was induced by occlusion of the left renal artery and vein for 45 min, followed by reperfusion 2 weeks after contralateral nephrectomy. One day after reperfusion, there was marked deterioration in renal function evidenced by an increase in plasma creatinine. Furthermore, blood IS levels increased markedly due to worsening renal function. Seven days and 28 days after reperfusion, blood IS levels decreased with the improvement in renal function. Of note, acetylcholine-induced vasorelaxation deteriorated over time after reperfusion, contradicting the recovery of renal function. In addition, 28 days after reperfusion, we observed a significant increase in ROS production in the vascular tissue. Next, we administered AST-120, a spherical adsorbent charcoal, after reperfusion to assess whether the vascular endothelial dysfunction associated with the ischemic AKI was due to a temporary increase in blood IS levels. AST-120 reduced the temporary increase in blood IS levels after reperfusion without influencing renal function, but did not restore the impaired vascular reactivity. Thus, in ischemic AKI, we confirmed that the vascular endothelial function of the thoracic aorta is impaired even after the recovery of kidney injury, probably with excessive ROS production. IS, which increases from ischemia to early after reperfusion, may not be a major contributor to the vascular dysfunction associated with ischemic AKI.
Collapse
|
34
|
Lei S, Zhang G, Zhu B, Long X, Jiang Z, Liu Y, Hu D, Sheng Z, Zhang Q, Wang C, Gao Z, Zheng H, Ma T. In Vivo Ultrasound Localization Microscopy Imaging of the Kidney's Microvasculature With Block-Matching 3-D Denoising. IEEE TRANSACTIONS ON ULTRASONICS, FERROELECTRICS, AND FREQUENCY CONTROL 2022; 69:523-533. [PMID: 34727030 DOI: 10.1109/tuffc.2021.3125010] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
Structural abnormalities and functional changes of renal microvascular networks play a significant pathophysiologic role in the occurrence of kidney diseases. Super-resolution ultrasound imaging has been successfully utilized to visualize the microvascular network and provide valuable diagnostic information. To prevent the burst of microbubbles, a lower mechanical index (MI) is generally used in ultrasound localization microscopy (ULM) imaging. However, high noise levels lead to incorrect signal localizations in relatively low-MI settings and deep tissue. In this study, we implemented a block-matching 3-D (BM3D) image-denoising method, after the application of singular value decomposition filtering, to further suppress the noise at various depths. The in vitro flow-phantom results show that the BM3D method helps the significant reduction of the error localizations, thus improving the localization accuracy. In vivo rhesus macaque experiments help conclude that the BM3D method improves the resolution more than other image-based denoising techniques, such as the nonlocal means method. The obtained clutter-filtered images with fewer incorrect localizations can enable robust ULM imaging, thus helping in establishing an effective diagnostic tool.
Collapse
|
35
|
Mutchler SM, Hasan M, Kohan DE, Kleyman TR, Tan RJ. Deletion of the Gamma Subunit of ENaC in Endothelial Cells Does Not Protect against Renal Ischemia Reperfusion Injury. Int J Mol Sci 2021; 22:ijms222010914. [PMID: 34681576 PMCID: PMC8535410 DOI: 10.3390/ijms222010914] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2021] [Revised: 10/01/2021] [Accepted: 10/04/2021] [Indexed: 11/16/2022] Open
Abstract
Acute kidney injury due to renal ischemia-reperfusion injury (IRI) may lead to chronic or end stage kidney disease. A greater understanding of the cellular mechanisms underlying IRI are required to develop therapeutic options aimed at limiting or reversing damage from IRI. Prior work has shown that deletion of the α subunit of the epithelial Na+ channel (ENaC) in endothelial cells protects from IRI by increasing the availability of nitric oxide. While canonical ENaCs consist of an α, β, and γ subunit, there is evidence of non-canonical ENaC expression in endothelial cells involving the α subunit. We therefore tested whether the deletion of the γ subunit of ENaC also protects mice from IRI to differentiate between these channel configurations. Mice with endothelial-specific deletion of the γ subunit and control littermates were subjected to unilateral renal artery occlusion followed by 48 h of reperfusion. No significant difference was noted in injury between the two groups as assessed by serum creatinine and blood urea nitrogen, levels of specific kidney injury markers, and histological examination. While deletion of the γ subunit did not alter infiltration of immune cells or cytokine message, it was associated with an increase in levels of total and phosphorylated endothelial nitric oxide synthase (eNOS) in the injured kidneys. Our studies demonstrate that even though deletion of the γ subunit of ENaC may allow for greater activation of eNOS, this is not sufficient to prevent IRI, suggesting the protective effects of α subunit deletion may be due, in part, to other mechanisms.
Collapse
Affiliation(s)
- Stephanie M. Mutchler
- Department of Medicine, University of Pittsburgh, Pittsburgh, PA 15261, USA; (S.M.M.); (R.J.T.)
| | - Mahpara Hasan
- Department of Biological Sciences, Carnegie Mellon University, Pittsburgh, PA 15213, USA;
| | - Donald E. Kohan
- Department of Medicine, University of Utah, Salt Lake City, UT 84112, USA;
| | - Thomas R. Kleyman
- Departments of Medicine, Cell Biology, and Pharmacology and Chemical Biology, University of Pittsburgh, Pittsburgh, PA 15261, USA
- Correspondence:
| | - Roderick J. Tan
- Department of Medicine, University of Pittsburgh, Pittsburgh, PA 15261, USA; (S.M.M.); (R.J.T.)
| |
Collapse
|
36
|
Netrebenko AS, Gureev VV, Pokrovskii MV, Gureeva AV, Tsuverkalova YM, Rozhkov IS. Assessment of the Nephroprotective Properties of the Erythropoietin Mimetic Peptide and Infliximab in Kidney Ischemia-Reperfusion Injury in Rats. ARCHIVES OF RAZI INSTITUTE 2021; 76:995-1004. [PMID: 35096335 DOI: 10.22092/ari.2021.355849.1728] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 08/07/2021] [Accepted: 09/09/2021] [Indexed: 10/13/2022]
Abstract
Chronic kidney disease (CKD) or acute kidney injury (AKI) causes impaired kidney function, leading to cognitive impairment, neuropathy, and cerebrovascular disease. Due to kidney damage, toxins stay in the blood rather than leaving the body through the urine, and brain function is affected by kidney-brain interaction. The present study aimed to investigate the protective effects of erythropoietin mimetic peptide (pHBSP) and infliximab on ischemic renal reperfusion injury. The experiment was performed on 70 white male Wistar laboratory rats which received recombinant erythropoietin, pHBSP, and infliximab. Under anesthesia, traumatic vascular clamps were applied to the left renal pedicle for 40 min, and nephrectomy was performed on the right. Functional tests and laboratory tests were performed 5 min and 24 h after the reperfusion. Thereafter, 24 h after the surgery, the plasma creatinine and urea levels in the sham-operated animals were obtained at 45.9±0.8 mmol/L and 6.7±0.2 mmol/L, respectively. Plasma creatinine and urea levels in the control group animals were 102.63±3.6 mmol/L and 21.80±1.29 mmol/L, respectively. The administration of pHBSP and infliximab to the animals with ischemia-reperfusion kidney injury has a pronounced nephroprotective effect, as compared to erythropoietin. There was a significant decrease in blood levels of creatinine and urea, improvement of microcirculation in the kidney, normalization of glomerular filtration rate, and fractional sodium excretion. The results of the study demonstrated pointed to the prospects of pHBSP and infliximab administration in ischemia-reperfusion kidney injury and justified the feasibility of further research in this field.
Collapse
Affiliation(s)
- A S Netrebenko
- Belgorod National Research University, 85 Pobeda St., Belgorod, 308015, Russia
| | - V V Gureev
- Belgorod National Research University, 85 Pobeda St., Belgorod, 308015, Russia
| | - M V Pokrovskii
- Belgorod National Research University, 85 Pobeda St., Belgorod, 308015, Russia
| | - A V Gureeva
- Belgorod National Research University, 85 Pobeda St., Belgorod, 308015, Russia
| | - Y M Tsuverkalova
- Belgorod National Research University, 85 Pobeda St., Belgorod, 308015, Russia
| | - I S Rozhkov
- Belgorod National Research University, 85 Pobeda St., Belgorod, 308015, Russia
| |
Collapse
|
37
|
de Oliveira NA, Cardoso SC, Barbosa DA, da Fonseca CD. Acute kidney injury caused by venomous animals: inflammatory mechanisms. J Venom Anim Toxins Incl Trop Dis 2021; 27:20200189. [PMID: 34512738 PMCID: PMC8394371 DOI: 10.1590/1678-9199-jvatitd-2020-0189] [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: 01/09/2021] [Accepted: 04/15/2021] [Indexed: 01/06/2023] Open
Abstract
Either bites or stings of venomous animals comprise relevant public health problems in tropical countries. Acute kidney injury (AKI) induced by animal toxins is related to worse prognostic and outcomes. Being one the most important pathways to induce AKI following envenoming due to animal toxins, inflammation is an essential biological response that eliminates pathogenic bacteria and repairs tissue after injury. However, direct nephrotoxicity (i.e. apoptotic and necrotic mechanisms of toxins), pigmenturia (i.e. rhabdomyolysis and hemolysis), anaphylactic reactions, and coagulopathies could contribute to the renal injury. All these mechanisms are closely integrated, but inflammation is a distinct process. Hence, it is important to improve our understanding on inflammation mechanisms of these syndromes to provide a promising outlook to reduce morbidity and mortality. This literature review highlights the main scientific evidence of acute kidney injury induced by bites or stings from venomous animals and their inflammatory mechanisms. It included observational, cross-sectional, case-control and cohort human studies available up to December 2019. Descriptors were used according to Medical Subject Headings (MeSH), namely: “Acute kidney injury” or “Venom” and “Inflammation” on Medline/Pubmed and Google Scholar; “Kidney disease” or “Acute kidney injury” on Lilacs and SciELO. The present review evidenced that, among the described forms of renal inflammation, it can occur either directly or indirectly on renal cells by means of intravascular, systemic and endothelial hemolysis, activation of inflammatory pathway, as well as direct action of venom cytotoxic components on kidney structures.
Collapse
Affiliation(s)
- Naila Albertina de Oliveira
- Department of Nursing, Institute of Health Sciences, Paulista University (Unip), Jundiaí, SP, Brazil.,Graduate Program in Nursing, School of Nursing, Federal University of São Paulo (Unifesp), São Paulo, SP, Brazil
| | | | - Dulce Aparecida Barbosa
- Department of Clinical and Surgical Nursing, School of Nursing, Federal University of São Paulo (Unifesp), São Paulo, SP, Brazil
| | - Cassiane Dezoti da Fonseca
- Department of Clinical and Surgical Nursing, School of Nursing, Federal University of São Paulo (Unifesp), São Paulo, SP, Brazil
| |
Collapse
|
38
|
Lan S, Yang B, Migneault F, Turgeon J, Bourgault M, Dieudé M, Cardinal H, Hickey MJ, Patey N, Hébert MJ. Caspase-3-dependent peritubular capillary dysfunction is pivotal for the transition from acute to chronic kidney disease after acute ischemia-reperfusion injury. Am J Physiol Renal Physiol 2021; 321:F335-F351. [PMID: 34338031 DOI: 10.1152/ajprenal.00690.2020] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2020] [Accepted: 07/21/2021] [Indexed: 11/22/2022] Open
Abstract
Ischemia-reperfusion injury (IRI) is a major risk factor for chronic renal failure. Caspase-3, an effector responsible for apoptosis execution, is activated within the peritubular capillary (PTC) in the early stage of IRI-induced acute kidney injury (AKI). Recently, we showed that caspase-3-dependent microvascular rarefaction plays a key role in fibrosis development after mild renal IRI. Here, we further characterized the role of caspase-3 in microvascular dysfunction and progressive renal failure in both mild and severe AKI, by performing unilateral renal artery clamping for 30/60 min with contralateral nephrectomy in wild-type (C57BL/6) or caspase-3-/- mice. In both forms of AKI, caspase-3-/- mice showed better long-term outcomes despite worse initial tubular injury. After 3 wk, they showed reduced PTC injury, decreased PTC collagen deposition and α-smooth muscle actin expression, and lower tubular injury scores compared with wild-type animals. Caspase-3-/- mice with severe IRI also showed better preservation of long-term renal function. Intravital imaging and microcomputed tomography revealed preserved PTC permeability and better terminal capillary density in caspase-3-/- mice. Collectively, these results demonstrate the pivotal importance of caspase-3 in regulating long-term renal function after IRI and establish the predominant role of PTC dysfunction as a major contributor to progressive renal dysfunction.NEW & NOTEWORTHY Our findings demonstrate the pivotal importance of caspase-3 in regulating renal microvascular dysfunction, fibrogenesis, and long-term renal impairment after acute kidney injury induced by ischemia-reperfusion injury. Furthermore, this study establishes the predominant role of peritubular capillary integrity as a major contributor to progressive renal dysfunction after ischemia-reperfusion injury.
Collapse
Affiliation(s)
- Shanshan Lan
- Research Centre, Centre hospitalier de l'Université de Montréal, Montreal, Quebec, Canada
- Canadian Donation Transplant Research Program, Edmonton, Alberta, Canada
- Université de Montréal, Montreal, Quebec, Canada
| | - Bing Yang
- Research Centre, Centre hospitalier de l'Université de Montréal, Montreal, Quebec, Canada
- Canadian Donation Transplant Research Program, Edmonton, Alberta, Canada
- Université de Montréal, Montreal, Quebec, Canada
| | - Francis Migneault
- Research Centre, Centre hospitalier de l'Université de Montréal, Montreal, Quebec, Canada
- Canadian Donation Transplant Research Program, Edmonton, Alberta, Canada
| | - Julie Turgeon
- Research Centre, Centre hospitalier de l'Université de Montréal, Montreal, Quebec, Canada
- Canadian Donation Transplant Research Program, Edmonton, Alberta, Canada
| | - Maude Bourgault
- Research Centre, Centre hospitalier de l'Université de Montréal, Montreal, Quebec, Canada
| | - Mélanie Dieudé
- Research Centre, Centre hospitalier de l'Université de Montréal, Montreal, Quebec, Canada
- Canadian Donation Transplant Research Program, Edmonton, Alberta, Canada
- Université de Montréal, Montreal, Quebec, Canada
| | - Héloïse Cardinal
- Research Centre, Centre hospitalier de l'Université de Montréal, Montreal, Quebec, Canada
- Canadian Donation Transplant Research Program, Edmonton, Alberta, Canada
- Université de Montréal, Montreal, Quebec, Canada
| | - Michael J Hickey
- Centre for Inflammatory Diseases, Monash University Department of Medicine, Monash Medical Centre, Clayton, Victoria, Australia
| | - Natacha Patey
- Research Centre, Centre hospitalier de l'Université de Montréal, Montreal, Quebec, Canada
- Université de Montréal, Montreal, Quebec, Canada
- Department of Pathology, Centre Hospitalier Universitaire Sainte-Justine, Université de Montréal, Montreal, Quebec, Canada
| | - Marie-Josée Hébert
- Research Centre, Centre hospitalier de l'Université de Montréal, Montreal, Quebec, Canada
- Canadian Donation Transplant Research Program, Edmonton, Alberta, Canada
- Université de Montréal, Montreal, Quebec, Canada
| |
Collapse
|
39
|
Gao F, Qian M, Liu G, Ao W, Dai D, Yin C. USP10 alleviates sepsis-induced acute kidney injury by regulating Sirt6-mediated Nrf2/ARE signaling pathway. JOURNAL OF INFLAMMATION-LONDON 2021; 18:25. [PMID: 34412625 PMCID: PMC8375185 DOI: 10.1186/s12950-021-00291-7] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/16/2021] [Accepted: 08/06/2021] [Indexed: 02/08/2023]
Abstract
Background Severe sepsis, a major health problem worldwide, has become one of the leading causes of death in ICU patients. Further study on the pathogenesis and treatment of acute kidney injury (AKI) is of great significance to reduce high mortality rate of sepsis. In this study, the mechanism by which ubiquitin specific peptidase 10 (USP10) reduces sepsis-induced AKI was investigated. Ligation and perforation of cecum (CLP) was employed to establish C57BL/6 mouse models of sepsis. Hematoxylin-eosin (H&E) staining was performed to detect renal injury. The concentrations of serum creatinine (Cr), urea nitrogen (BUN) and cystatin C (Cys C) were determined using a QuantiChrom™ Urea Assay kit. RT-qPCR and western blot were conducted to assess the USP10 expression level. DHE staining was used to detect reactive oxygen species (ROS) levels. H2O2, MDA and SOD levels were assessed using corresponding colorimetric kits. Western blot was used to examine the expression levels of Bcl-2, Bax, cleaved caspase-3, Sirt6, Nrf2 and HO-1. MTT assay was used to determine cell viability, whereas TUNEL staining and flow cytometry were used to assess cell apoptosis. Results In this study, we found that USP10 was decreased in CLP-induced mouse renal tissues. We identified that USP10 alleviated renal dysfunction induced by CLP. Moreover, USP10 was found to reduce oxidative stress, and abated LPS-induced renal tubular epithelial cell injury and apoptosis. Finally, we discovered that USP10 promoted activation of the NRF2/HO-1 pathway through SIRT6 and attenuated LPS-induced renal tubular epithelial cell injury. Conclusions This study found that USP10 activates the NRF2/ARE signaling through SIRT6. USP10 alleviates sepsis-induced renal dysfunction and reduces renal tubular epithelial cell apoptosis and oxidative stress.
Collapse
Affiliation(s)
- Fei Gao
- Department of Critical Care Medicine, Affiliated Hospital of Zunyi Medical University, 563003, Zunyi City, Guizhou Province, China
| | - Mingjiang Qian
- Department of Critical Care Medicine, The Second Affiliated Hospital of Zunyi Medical University, Intersection of Xinlong Avenue and Xinpu Avenue, Xinpu New District, Honghuagang District, 563000, Zunyi City, Guizhou Province, China.
| | - Guoyue Liu
- Department of Critical Care Medicine, The Second Affiliated Hospital of Zunyi Medical University, Intersection of Xinlong Avenue and Xinpu Avenue, Xinpu New District, Honghuagang District, 563000, Zunyi City, Guizhou Province, China
| | - Wanping Ao
- Department of Critical Care Medicine, Affiliated Hospital of Zunyi Medical University, 563003, Zunyi City, Guizhou Province, China
| | - Dahua Dai
- Department of Critical Care Medicine, Affiliated Hospital of Zunyi Medical University, 563003, Zunyi City, Guizhou Province, China
| | - Cunzhi Yin
- Department of Critical Care Medicine, The Second Affiliated Hospital of Zunyi Medical University, Intersection of Xinlong Avenue and Xinpu Avenue, Xinpu New District, Honghuagang District, 563000, Zunyi City, Guizhou Province, China
| |
Collapse
|
40
|
Thapa K, Singh TG, Kaur A. Cyclic nucleotide phosphodiesterase inhibition as a potential therapeutic target in renal ischemia reperfusion injury. Life Sci 2021; 282:119843. [PMID: 34298037 DOI: 10.1016/j.lfs.2021.119843] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2021] [Revised: 07/10/2021] [Accepted: 07/13/2021] [Indexed: 12/19/2022]
Abstract
AIMS Ischemia/reperfusion (I/R) occurs in renal artery stenosis, partial nephrectomy and most commonly during kidney transplantation. It brings serious consequences such as DGF (Delayed Graft Function) or organ dysfunction leading to renal failure and ultimate death. There is no effective therapy to handle the consequences of Renal Ischemia/Reperfusion (I/R) injury. Cyclic nucleotides, cAMP and cGMP are the important second messengers that stimulate intracellular signal transduction for cell survival in response to growth factors and peptide hormones in normal tissues and in kidneys plays significant role that involves vascular tone regulation, inflammation and proliferation of parenchymal cells. Renal ischemia and subsequent reperfusion injury stimulate signal transduction pathways involved in oxidative stress, inflammation, alteration in renal blood flow leading to necrosis and apoptosis of renal cell. MATERIALS AND METHODS An extensive literature review of various search engines like PubMed, Medline, Bentham, Scopus, and EMBASE (Elsevier) databases was carried out. To understand the functioning of Phosphodiesterases (PDEs) and its pharmacological modulation in Renal Ischemia-Reperfusion Injury. KEY FINDINGS Current therapeutic options may not be enough to treat renal I/R injury in group of patients and therefore, the current review has discussed the general characteristics and physiology of PDEs and preclinical-studies defining the relationship between PDEs expression in renal injury due to I/R and its outcome on renal function. SIGNIFICANCE The role of PDE inhibitors in renal I/R injury and the clinical status of drugs for various renal diseases have been summarized in this review.
Collapse
Affiliation(s)
- Komal Thapa
- Chitkara College of Pharmacy, Chitkara University, 140401 Punjab, India; School of Pharmacy, Himachal Pradesh, India
| | | | - Amarjot Kaur
- Chitkara College of Pharmacy, Chitkara University, 140401 Punjab, India
| |
Collapse
|
41
|
Viñas JL, Spence M, Porter CJ, Douvris A, Gutsol A, Zimpelmann JA, Campbell PA, Burns KD. micro-RNA-486-5p protects against kidney ischemic injury and modifies the apoptotic transcriptome in proximal tubules. Kidney Int 2021; 100:597-612. [PMID: 34181969 DOI: 10.1016/j.kint.2021.05.034] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2020] [Revised: 05/18/2021] [Accepted: 05/20/2021] [Indexed: 02/08/2023]
Abstract
Acute kidney injury (AKI) carries high morbidity and mortality, and effective treatments are lacking. Preclinical models support involvement of micro-RNAs (miRs) in AKI pathogenesis, although effects on the kidney transcriptome are unclear. We previously showed that injection of cord blood endothelial colony forming cell-derived exosomes, enriched in miR-486-5p, prevented ischemic AKI in mice. To further define this, we studied direct effects of miR-486-5p in mice with kidney ischemia-reperfusion injury. RNA-Seq was used to compare the impact of miR-486-5p and exosomes on the transcriptome of proximal tubules and kidney endothelial cells 24 hours after ischemia-reperfusion. In mice with AKI, injection of miR-486-5p mimic increased its levels in proximal tubules and endothelial cells, and improved plasma creatinine, histological injury, neutrophil infiltration, and apoptosis. Additionally, miR-486-5p inhibited expression of its target phosphatase and tensin homolog, and activated protein kinase B. In proximal tubules, miR-486-5p or exosomes reduced expression of genes associated with ischemic injury and the tumor necrosis factor (TNF) pathway, and altered distinct apoptotic genes. In endothelial cells, genes associated with metabolic processes were altered by miR-486-5p or exosomes, although TNF pathway genes were not affected. Thus, our results suggest that miR-486-5p may have therapeutic potential in AKI.
Collapse
Affiliation(s)
- Jose L Viñas
- Division of Nephrology, Department of Medicine, Kidney Research Centre, Ottawa Hospital Research Institute, University of Ottawa, Ottawa, Ontario, Canada
| | - Matthew Spence
- Division of Nephrology, Department of Medicine, Kidney Research Centre, Ottawa Hospital Research Institute, University of Ottawa, Ottawa, Ontario, Canada
| | - Christopher J Porter
- Ottawa Bioinformatics Core Facility, the Sprott Centre for Stem Cell Research, Ottawa Hospital Research Institute, Ottawa, Ontario, Canada
| | - Adrianna Douvris
- Division of Nephrology, Department of Medicine, Kidney Research Centre, Ottawa Hospital Research Institute, University of Ottawa, Ottawa, Ontario, Canada
| | - Alex Gutsol
- Division of Nephrology, Department of Medicine, Kidney Research Centre, Ottawa Hospital Research Institute, University of Ottawa, Ottawa, Ontario, Canada
| | - Joseph A Zimpelmann
- Division of Nephrology, Department of Medicine, Kidney Research Centre, Ottawa Hospital Research Institute, University of Ottawa, Ottawa, Ontario, Canada
| | - Pearl A Campbell
- Regenerative Medicine Program, the Sprott Centre for Stem Cell Research, Ottawa Hospital Research Institute, Ottawa, Ontario, Canada
| | - Kevin D Burns
- Division of Nephrology, Department of Medicine, Kidney Research Centre, Ottawa Hospital Research Institute, University of Ottawa, Ottawa, Ontario, Canada.
| |
Collapse
|
42
|
Ischemia-Reperfusion Injuries Assessment during Pancreas Preservation. Int J Mol Sci 2021; 22:ijms22105172. [PMID: 34068301 PMCID: PMC8153272 DOI: 10.3390/ijms22105172] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2021] [Revised: 04/25/2021] [Accepted: 05/10/2021] [Indexed: 12/20/2022] Open
Abstract
Maintaining organ viability between donation and transplantation is of critical importance for optimal graft function and survival. To date in pancreas transplantation, static cold storage (SCS) is the most widely practiced method of organ preservation. The first experiments in ex vivo perfusion of the pancreas were performed at the beginning of the 20th century. These perfusions led to organ oedema, hemorrhage, and venous congestion after revascularization. Despite these early hurdles, a number of factors now favor the use of perfusion during preservation: the encouraging results of HMP in kidney transplantation, the development of new perfusion solutions, and the development of organ perfusion machines for the lung, heart, kidneys and liver. This has led to a resurgence of research in machine perfusion for whole organ pancreas preservation. This review highlights the ischemia-reperfusion injuries assessment during ex vivo pancreas perfusion, both for assessment in pre-clinical experimental models as well for future use in the clinic. We evaluated perfusion dynamics, oedema assessment, especially by impedance analysis and MRI, whole organ oxygen consumption, tissue oxygen tension, metabolite concentrations in tissue and perfusate, mitochondrial respiration, cell death, especially by histology, total cell free DNA, caspase activation, and exocrine and endocrine assessment.
Collapse
|
43
|
Karimi F, Nematbakhsh M. Mas Receptor Blockade Promotes Renal Vascular Response to Ang II after Partial Kidney Ischemia/Reperfusion in a Two-Kidney-One-Clip Hypertensive Rats Model. Int J Nephrol 2021; 2021:6618061. [PMID: 33986960 PMCID: PMC8079216 DOI: 10.1155/2021/6618061] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2020] [Revised: 03/18/2021] [Accepted: 04/12/2021] [Indexed: 12/26/2022] Open
Abstract
BACKGROUND Partial kidney ischemia-reperfusion (IR) injury is the principal cause of acute kidney injury. The renin-angiotensin system (RAS) and hypertension also may be influenced by renal IR injury. In two models of partial renal IR with and without ischemia preconditioning (IPC) and using Mas receptor (MasR) blockade, A779 or its vehicle, the renal vascular responses to angiotensin II (Ang II) administration in two-kidney-one-clip (2K1C) hypertensive rats were determined. METHODS Thirty-seven 2K1C male Wistar rats with systolic blood pressure ≥150 mmHg were randomly divided into three groups; sham, IR, and IPC + IR. The animals in the sham group underwent surgical procedures except partial IR. The rats in the IR group underwent 45 min partial kidney ischemia, and the animals in the IPC + IR group underwent two 5 min cycles of partial kidney ischemia followed by 10 min reperfusion and partial kidney ischemia for 45 min. The renal vascular responses to graded Ang II (30, 100, 300, and 1000 ng kg-1.min-1) infusion using A779 or its vehicle were measured at constant renal perfusion pressure. RESULTS Four weeks after 2K1C implementation, the intravenous infusion of graded Ang II resulted in dose-related increases in mean arterial pressure (MAP) (P dose < 0.0001) that was not different significantly between the groups. No significant differences were detected between the groups in renal blood flow (RBF) or renal vascular resistance (RVR) responses to Ang II infusion when MasR was not blocked. However, by MasR blockade, these responses were increased in IR and IPC + IR groups that were significantly different from the sham group (P < 0.05). For example, infusion of Ang II at dose 1000 ng kg-1.min-1 resulted in decreased RBF percentage change (RBF%) from the baseline to 17.5 ± 1.9%, 39.7 ± 3.8%, and 31.0 ± 3.4% in sham, IR, and IPC + IR, respectively. CONCLUSION These data revealed the important role of MasR after partial kidney IR in the responses of RBF and RVR to Ang II administration in 2K1C hypertensive rats.
Collapse
Affiliation(s)
- Farzaneh Karimi
- Water and Electrolytes Research Center, Isfahan University of Medical Sciences, Isfahan, Iran
- Department of Physiology, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Mehdi Nematbakhsh
- Water and Electrolytes Research Center, Isfahan University of Medical Sciences, Isfahan, Iran
- Department of Physiology, Isfahan University of Medical Sciences, Isfahan, Iran
- Isfahan MN Institute of Basic and Applied Sciences Research, Isfahan, Iran
| |
Collapse
|
44
|
Doreille A, Azzi F, Larivière-Beaudoin S, Karakeussian-Rimbaud A, Trudel D, Hébert MJ, Dieudé M, Patey N, Cardinal H. Acute Kidney Injury, Microvascular Rarefaction, and Estimated Glomerular Filtration Rate in Kidney Transplant Recipients. Clin J Am Soc Nephrol 2021; 16:415-426. [PMID: 33648972 PMCID: PMC8011007 DOI: 10.2215/cjn.07270520] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2020] [Accepted: 01/14/2021] [Indexed: 02/04/2023]
Abstract
BACKGROUND AND OBJECTIVES Animal studies suggest that microvascular rarefaction is a key factor in the acute kidney disease to CKD transition. Hence, delayed graft function appears as a unique human model of AKI to further explore the role of microvascular rarefaction in kidney transplant recipients. Here, we assessed whether delayed graft function is associated with peritubular capillary loss and evaluated the association between this loss and long-term kidney graft function. DESIGN, SETTING, PARTICIPANTS, & MEASUREMENTS This observational, retrospective cohort study included 61 participants who experienced delayed graft function and 130 who had immediate graft function. We used linear regression models to evaluate associations between delayed graft function and peritubular capillary density expressed as the percentage of efficient cortical area occupied by peritubular capillaries in pre- and post-transplant graft biopsies. eGFRs 1 and 3 years post-transplant were secondary outcomes. RESULTS Post-transplant biopsies were performed at a median of 113 days (interquartile range, 101-128) after transplantation. Peritubular capillary density went from 15.4% to 11.5% in patients with delayed graft function (median change, -3.7%; interquartile range, -6.6% to -0.8%) and from 19.7% to 15.1% in those with immediate graft function (median change, -4.5%; interquartile range, -8.0% to -0.8%). Although the unadjusted change in peritubular capillary density was similar between patients with and without delayed graft function, delayed graft function was associated with more peritubular capillary loss in the multivariable analysis (adjusted difference in change, -2.9%; 95% confidence interval, -4.0 to -1.8). Pretransplant peritubular capillary density and change in peritubular capillary density were associated with eGFR 1 and 3 years post-transplantation. CONCLUSIONS Perioperative AKI is associated with lower density in peritubular capillaries before transplantation and with loss of peritubular capillaries following transplantation. Lower peritubular capillary density is linked to lower long-term eGFR.
Collapse
Affiliation(s)
- Alice Doreille
- Centre de recherche du Centre hospitalier de l’Université de Montréal, Immunopathology axis, Montreal, Quebec, Canada,Faculté de Médecine, Université Paris-Sud, Paris, France
| | - Féryel Azzi
- Institut du cancer de Montréal, Montreal, Quebec, Canada
| | - Stéphanie Larivière-Beaudoin
- Centre de recherche du Centre hospitalier de l’Université de Montréal, Immunopathology axis, Montreal, Quebec, Canada,Canadian Donation and Transplantation Research Program, Edmonton, Alberta, Canada
| | - Annie Karakeussian-Rimbaud
- Centre de recherche du Centre hospitalier de l’Université de Montréal, Immunopathology axis, Montreal, Quebec, Canada,Canadian Donation and Transplantation Research Program, Edmonton, Alberta, Canada
| | - Dominique Trudel
- Institut du cancer de Montréal, Montreal, Quebec, Canada,Pathology Department, Centre hospitalier de l’Université de Montréal, Montreal, Quebec, Canada
| | - Marie-Josée Hébert
- Centre de recherche du Centre hospitalier de l’Université de Montréal, Immunopathology axis, Montreal, Quebec, Canada,Canadian Donation and Transplantation Research Program, Edmonton, Alberta, Canada,Nephrology Department, Centre hospitalier de l’Université de Montréal, Montreal, Quebec, Canada
| | - Mélanie Dieudé
- Centre de recherche du Centre hospitalier de l’Université de Montréal, Immunopathology axis, Montreal, Quebec, Canada,Canadian Donation and Transplantation Research Program, Edmonton, Alberta, Canada
| | - Natacha Patey
- Centre de recherche du Centre hospitalier de l’Université de Montréal, Immunopathology axis, Montreal, Quebec, Canada,Pathology Department, Sainte-Justine Hospital, Montreal, Quebec, Canada
| | - Héloïse Cardinal
- Centre de recherche du Centre hospitalier de l’Université de Montréal, Immunopathology axis, Montreal, Quebec, Canada,Canadian Donation and Transplantation Research Program, Edmonton, Alberta, Canada,Nephrology Department, Centre hospitalier de l’Université de Montréal, Montreal, Quebec, Canada
| |
Collapse
|
45
|
Drummer C, Saaoud F, Shao Y, Sun Y, Xu K, Lu Y, Ni D, Atar D, Jiang X, Wang H, Yang X. Trained Immunity and Reactivity of Macrophages and Endothelial Cells. Arterioscler Thromb Vasc Biol 2021; 41:1032-1046. [PMID: 33380171 PMCID: PMC7904591 DOI: 10.1161/atvbaha.120.315452] [Citation(s) in RCA: 56] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2020] [Accepted: 11/14/2020] [Indexed: 12/15/2022]
Abstract
Innate immune cells can develop exacerbated immunologic response and long-term inflammatory phenotype following brief exposure to endogenous or exogenous insults, which leads to an altered response towards a second challenge after the return to a nonactivated state. This phenomenon is known as trained immunity (TI). TI is not only important for host defense and vaccine response but also for chronic inflammations such as cardiovascular and metabolic diseases such as atherosclerosis. TI can occur in innate immune cells such as monocytes/macrophages, natural killer cells, endothelial cells (ECs), and nonimmune cells, such as fibroblast. In this brief review, we analyze the significance of TI in ECs, which are also considered as innate immune cells in addition to macrophages. TI can be induced by a variety of stimuli, including lipopolysaccharides, BCG (bacillus Calmette-Guerin), and oxLDL (oxidized low-density lipoprotein), which are defined as risk factors for cardiovascular and metabolic diseases. Furthermore, TI in ECs is functional for inflammation effectiveness and transition to chronic inflammation. Rewiring of cellular metabolism of the trained cells takes place during induction of TI, including increased glycolysis, glutaminolysis, increased accumulation of tricarboxylic acid cycle metabolites and acetyl-coenzyme A production, as well as increased mevalonate synthesis. Subsequently, this leads to epigenetic remodeling, resulting in important changes in chromatin architecture that enables increased gene transcription and enhanced proinflammatory immune response. However, TI pathways and inflammatory pathways are separated to ensure memory stays when inflammation undergoes resolution. Additionally, reactive oxygen species play context-dependent roles in TI. Therefore, TI plays significant roles in EC and macrophage pathology and chronic inflammation. However, further characterization of TI in ECs and macrophages would provide novel insights into cardiovascular disease pathogenesis and new therapeutic targets. Graphic Abstract: A graphic abstract is available for this article.
Collapse
Affiliation(s)
- Charles Drummer
- Cardiovascular Research Center, Centers for Inflammation, Translational and Clinical Lung Research and Thrombosis Research, Lewis Katz School of Medicine at Temple University, Philadelphia, PA 19140, USA
| | - Fatma Saaoud
- Cardiovascular Research Center, Centers for Inflammation, Translational and Clinical Lung Research and Thrombosis Research, Lewis Katz School of Medicine at Temple University, Philadelphia, PA 19140, USA
| | - Ying Shao
- Cardiovascular Research Center, Centers for Inflammation, Translational and Clinical Lung Research and Thrombosis Research, Lewis Katz School of Medicine at Temple University, Philadelphia, PA 19140, USA
| | - Yu Sun
- Cardiovascular Research Center, Centers for Inflammation, Translational and Clinical Lung Research and Thrombosis Research, Lewis Katz School of Medicine at Temple University, Philadelphia, PA 19140, USA
| | - Keman Xu
- Cardiovascular Research Center, Centers for Inflammation, Translational and Clinical Lung Research and Thrombosis Research, Lewis Katz School of Medicine at Temple University, Philadelphia, PA 19140, USA
| | - Yifan Lu
- Cardiovascular Research Center, Centers for Inflammation, Translational and Clinical Lung Research and Thrombosis Research, Lewis Katz School of Medicine at Temple University, Philadelphia, PA 19140, USA
| | - Dong Ni
- Cardiovascular Research Center, Centers for Inflammation, Translational and Clinical Lung Research and Thrombosis Research, Lewis Katz School of Medicine at Temple University, Philadelphia, PA 19140, USA
| | - Diana Atar
- Cardiovascular Research Center, Centers for Inflammation, Translational and Clinical Lung Research and Thrombosis Research, Lewis Katz School of Medicine at Temple University, Philadelphia, PA 19140, USA
| | - Xiaohua Jiang
- Cardiovascular Research Center, Centers for Inflammation, Translational and Clinical Lung Research and Thrombosis Research, Lewis Katz School of Medicine at Temple University, Philadelphia, PA 19140, USA
- Metabolic Disease Research, Lewis Katz School of Medicine at Temple University, Philadelphia, PA 19140, USA
| | - Hong Wang
- Metabolic Disease Research, Lewis Katz School of Medicine at Temple University, Philadelphia, PA 19140, USA
- Departments of Pharmacology, Microbiology and Immunology, Lewis Katz School of Medicine at Temple University, Philadelphia, PA 19140, USA
| | - Xiaofeng Yang
- Cardiovascular Research Center, Centers for Inflammation, Translational and Clinical Lung Research and Thrombosis Research, Lewis Katz School of Medicine at Temple University, Philadelphia, PA 19140, USA
- Metabolic Disease Research, Lewis Katz School of Medicine at Temple University, Philadelphia, PA 19140, USA
- Departments of Pharmacology, Microbiology and Immunology, Lewis Katz School of Medicine at Temple University, Philadelphia, PA 19140, USA
| |
Collapse
|
46
|
Feng W, Remedies CE, Obi IE, Aldous SR, Meera SI, Sanders PW, Inscho EW, Guan Z. Restoration of afferent arteriolar autoregulatory behavior in ischemia-reperfusion injury in rat kidneys. Am J Physiol Renal Physiol 2021; 320:F429-F441. [PMID: 33491564 PMCID: PMC7988813 DOI: 10.1152/ajprenal.00500.2020] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2020] [Revised: 12/23/2020] [Accepted: 01/12/2021] [Indexed: 12/11/2022] Open
Abstract
Renal autoregulation is critical in maintaining stable renal blood flow (RBF) and glomerular filtration rate (GFR). Renal ischemia-reperfusion (IR)-induced kidney injury is characterized by reduced RBF and GFR. The mechanisms contributing to renal microvascular dysfunction in IR have not been fully determined. We hypothesized that increased reactive oxygen species (ROS) contributed to impaired renal autoregulatory capability in IR rats. Afferent arteriolar autoregulatory behavior was assessed using the blood-perfused juxtamedullary nephron preparation. IR was induced by 60 min of bilateral renal artery occlusion followed by 24 h of reperfusion. Afferent arterioles from sham rats exhibited normal autoregulatory behavior. Stepwise increases in perfusion pressure caused pressure-dependent vasoconstriction to 65 ± 3% of baseline diameter (13.2 ± 0.4 μm) at 170 mmHg. In contrast, pressure-mediated vasoconstriction was markedly attenuated in IR rats. Baseline diameter averaged 11.7 ± 0.5 µm and remained between 90% and 101% of baseline over 65-170 mmHg, indicating impaired autoregulatory function. Acute antioxidant administration (tempol or apocynin) to IR kidneys for 20 min increased baseline diameter and improved autoregulatory capability, such that the pressure-diameter profiles were indistinguishable from those of sham kidneys. Furthermore, the addition of polyethylene glycol superoxide dismutase or polyethylene glycol-catalase to the perfusate blood also restored afferent arteriolar autoregulatory responsiveness in IR rats, indicating the involvement of superoxide and/or hydrogen peroxide. IR elevated mRNA expression of NADPH oxidase subunits and monocyte chemoattractant protein-1 in renal tissue homogenates, and this was prevented by tempol pretreatment. These results suggest that ROS accumulation, likely involving superoxide and/or hydrogen peroxide, impairs renal autoregulation in IR rats in a reversible fashion.NEW & NOTEWORTHY Renal ischemia-reperfusion (IR) leads to renal microvascular dysfunction manifested by impaired afferent arteriolar autoregulatory efficiency. Acute administration of scavengers of reactive oxygen species, polyethylene glycol-superoxide dismutase, or polyethylene glycol-catalase following renal IR restored afferent arteriolar autoregulatory capability in IR rats, indicating that renal IR led to reversible impairment of afferent arteriolar autoregulatory capability. Intervention with antioxidant treatment following IR may improve outcomes in patients by preserving renovascular autoregulatory function and potentially preventing the progression to chronic kidney disease after acute kidney injury.
Collapse
Affiliation(s)
- Wenguang Feng
- Division of Nephrology, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama
| | - Colton E Remedies
- Division of Nephrology, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama
| | - Ijeoma E Obi
- Division of Nephrology, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama
| | - Stephen R Aldous
- Division of Nephrology, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama
| | - Samia I Meera
- Division of Nephrology, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama
| | - Paul W Sanders
- Division of Nephrology, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama
- Department of Veterans Affairs Medical Center, Birmingham, Alabama
| | - Edward W Inscho
- Division of Nephrology, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama
| | - Zhengrong Guan
- Division of Nephrology, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama
| |
Collapse
|
47
|
Renal Tubular Epithelial TRPA1 Acts as An Oxidative Stress Sensor to Mediate Ischemia-Reperfusion-Induced Kidney Injury through MAPKs/NF-κB Signaling. Int J Mol Sci 2021; 22:ijms22052309. [PMID: 33669091 PMCID: PMC7956664 DOI: 10.3390/ijms22052309] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2021] [Revised: 02/19/2021] [Accepted: 02/23/2021] [Indexed: 12/19/2022] Open
Abstract
Oxidative stress and inflammation play important roles in the pathophysiology of acute kidney injury (AKI). Transient receptor potential ankyrin 1 (TRPA1) is a Ca2+-permeable ion channel that is sensitive to reactive oxygen species (ROS). The role of TRPA1 in AKI remains unclear. In this study, we used human and animal studies to assess the role of renal TRPA1 in AKI and to explore the regulatory mechanism of renal TRPA1 in inflammation via in vitro experiments. TRPA1 expression increased in the renal tubular epithelia of patients with AKI. The severity of tubular injury correlated well with tubular TRPA1 or 8-hydroxy-2'-deoxyguanosine expression. In an animal model, renal ischemia-reperfusion injury (IR) increased tubular TRPA1 expression in wild-type (WT) mice. Trpa1-/- mice displayed less IR-induced tubular injury, oxidative stress, inflammation, and dysfunction in kidneys compared with WT mice. In the in vitro model, TRPA1 expression increased in renal tubular cells under hypoxia-reoxygenation injury (H/R) conditions. We demonstrated that H/R evoked a ROS-dependent TRPA1 activation, which elevated intracellular Ca2+ level, increased NADPH oxidase activity, activated MAPK/NF-κB signaling, and increased IL-8. Renal tubular TRPA1 may serve as an oxidative stress sensor and a crucial regulator in the activation of signaling pathways and promote the subsequent transcriptional regulation of IL-8. These actions might be evident in mice with IR or patients with AKI.
Collapse
|
48
|
Matjuda EN, Engwa GA, Sewani-Rusike CR, Nkeh-Chungag BN. An Overview of Vascular Dysfunction and Determinants: The Case of Children of African Ancestry. Front Pediatr 2021; 9:769589. [PMID: 34956981 PMCID: PMC8709476 DOI: 10.3389/fped.2021.769589] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/02/2021] [Accepted: 11/08/2021] [Indexed: 12/13/2022] Open
Abstract
The balance between dilatory and constrictive factors is important as it keeps blood vessels in a homeostatic state. However, altered physiological processes as a result of obesity, hypertension, oxidative stress, and other cardiovascular risk factors may lead to vascular damage, causing an imbalance of vasoactive factors. Over time, the sustained imbalance of these vasoactive factors may lead to vascular dysfunction, which can be assessed by non-invasive methods, such as flow-mediated dilation, pulse wave velocity, flow-mediated slowing, retinal vessel analysis, peripheral vascular reactivity, and carotid intima-media thickness assessment. Although there is increasing prevalence of cardiovascular risk factors (obesity and hypertension) in children in sub-Saharan Africa, little is known about how this may affect vascular function. This review focuses on vasoactive factors implicated in vascular (dys)function, highlighting the determinants and consequences of vascular dysfunction. It further describes the non-invasive methods used for vascular (dys)function assessments and, last, describes the impact of cardiovascular risk factors on vascular dysfunction in children of African ancestry.
Collapse
Affiliation(s)
- Edna N Matjuda
- Department of Human Biology, Faculty of Health Sciences, Walter Sisulu University PBX1, Mthatha, South Africa
| | - Godwill Azeh Engwa
- Department of Biological and Environmental Sciences, Faculty of Natural Sciences, Walter Sisulu University PBX1, Mthatha, South Africa
| | - Constance R Sewani-Rusike
- Department of Human Biology, Faculty of Health Sciences, Walter Sisulu University PBX1, Mthatha, South Africa
| | - Benedicta N Nkeh-Chungag
- Department of Biological and Environmental Sciences, Faculty of Natural Sciences, Walter Sisulu University PBX1, Mthatha, South Africa
| |
Collapse
|
49
|
Sex diversity in proximal tubule and endothelial gene expression in mice with ischemic acute kidney injury. Clin Sci (Lond) 2020; 134:1887-1909. [PMID: 32662516 DOI: 10.1042/cs20200168] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2020] [Revised: 07/09/2020] [Accepted: 07/10/2020] [Indexed: 12/22/2022]
Abstract
Female sex protects against development of acute kidney injury (AKI). While sex hormones may be involved in protection, the role of differential gene expression is unknown. We conducted gene profiling in male and female mice with or without kidney ischemia-reperfusion injury (IRI). Mice underwent bilateral renal pedicle clamping (30 min), and tissues were collected 24 h after reperfusion. RNA-sequencing (RNA-Seq) was performed on proximal tubules (PTs) and kidney endothelial cells. Female mice were resistant to ischemic injury compared with males, determined by plasma creatinine and neutrophil gelatinase-associated lipocalin (NGAL), histologic scores, neutrophil infiltration, and extent of apoptosis. Sham mice had sex-specific gene disparities in PT and endothelium, and male mice showed profound gene dysregulation with ischemia-reperfusion compared with females. After ischemia PTs from females exhibited smaller increases compared with males in injury-associated genes lipocalin-2 (Lcn2), hepatitis A virus cellular receptor 1 (Havcr1), and keratin 18 (Krt18), and no up-regulation of SRY-Box transcription factor 9 (Sox9) or keratin 20 (Krt20). Endothelial up-regulation of adhesion molecules and cytokines/chemokines occurred in males, but not females. Up-regulated genes in male ischemic PTs were linked to tumor necrosis factor (TNF) and Toll-like receptor (TLR) pathways, while female ischemic PTs showed up-regulated genes in pathways related to transport. The data highlight sex-specific gene expression differences in male and female PTs and endothelium before and after ischemic injury that may underlie disparities in susceptibility to AKI.
Collapse
|
50
|
Kim IY, Park YK, Song SH, Seong EY, Lee DW, Bae SS, Lee SB. Akt1 is involved in tubular apoptosis and inflammatory response during renal ischemia-reperfusion injury. Mol Biol Rep 2020; 47:9511-9520. [PMID: 33247386 DOI: 10.1007/s11033-020-06021-1] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2020] [Accepted: 11/19/2020] [Indexed: 01/14/2023]
Abstract
Renal ischemia-reperfusion injury (IRI) is one of the major causes of acute kidney injury (AKI). Although Akt is involved in renal IRI, it is unclear as to which Akt isoform plays an important role in renal IRI. In this study, we investigated the role of Akt1 in renal IRI. We subjected the C57BL/6 male mice to unilateral IRI with contralateral nephrectomy. Two days after IRI, IRI-kidneys were harvested. The mice were divided into four groups: wild type (WT) IRI, Akt1-/- IRI, WT sham, and Akt1-/- sham. We found that Akt1, not Akt2 or Akt3, was markedly activated in WT IRI than in WT sham mice. The histologic damage score and serum creatinine level significantly increased in WT IRI mice, the increase being the highest in Akt1-/- IRI mice. The number of TdT-mediated dUTP nick-end labeling (TUNEL)-positive tubular cells and expression of cleaved caspase-3/Bax were higher in Akt1-/- IRI mice than in WT IRI mice. The expression of Bcl-2 was lower in Akt1-/- IRI mice than in WT IRI mice. The expression of tumor necrosis factor-α/interleukin-6/interleukin-1β and number of F4/80-positive macrophages were markedly higher in Akt1-/- IRI than in WT IRI mice. The expression of phosphorylated nuclear factor-κB p65 was also higher in Akt1-/- IRI mice than in WT IRI mice. Our results show that Akt1 deletion exacerbates kidney damage as it increases tubular apoptosis and inflammatory response during renal IRI. Akt1 could be a potential therapeutic target for developing treatments against IRI-induced AKI.
Collapse
Affiliation(s)
- Il Young Kim
- Department of Internal Medicine, Pusan National University School of Medicine, Yangsan, Republic of Korea.,Research Institute for Convergence of Biomedical Science and Technology, Pusan National University Yangsan Hospital, Yangsan, Republic of Korea
| | - Yeon Kyeong Park
- Research Institute for Convergence of Biomedical Science and Technology, Pusan National University Yangsan Hospital, Yangsan, Republic of Korea
| | - Sang Heon Song
- Department of Internal Medicine, Pusan National University School of Medicine, Yangsan, Republic of Korea.,Medical Research Institute, Pusan National University Hospital, Busan, Republic of Korea
| | - Eun Young Seong
- Department of Internal Medicine, Pusan National University School of Medicine, Yangsan, Republic of Korea.,Medical Research Institute, Pusan National University Hospital, Busan, Republic of Korea
| | - Dong Won Lee
- Department of Internal Medicine, Pusan National University School of Medicine, Yangsan, Republic of Korea.,Research Institute for Convergence of Biomedical Science and Technology, Pusan National University Yangsan Hospital, Yangsan, Republic of Korea
| | - Sun Sik Bae
- MRC for Ischemic Tissue Regeneration, Medical Research Institute, and Department of Pharmacology, Pusan National University School of Medicine, Yangsan, Republic of Korea
| | - Soo Bong Lee
- Department of Internal Medicine, Pusan National University School of Medicine, Yangsan, Republic of Korea. .,Research Institute for Convergence of Biomedical Science and Technology, Pusan National University Yangsan Hospital, Yangsan, Republic of Korea.
| |
Collapse
|