1
|
Wu W, Lan W, Jiao X, Wang K, Deng Y, Chen R, Zeng R, Li J. Pyroptosis in sepsis-associated acute kidney injury: mechanisms and therapeutic perspectives. Crit Care 2025; 29:168. [PMID: 40270016 PMCID: PMC12020238 DOI: 10.1186/s13054-025-05329-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2025] [Accepted: 02/21/2025] [Indexed: 04/25/2025] Open
Abstract
Sepsis-associated acute kidney injury (S-AKI) is a severe complication characterized by high morbidity and mortality, driven by multi-organ dysfunction. Recent evidence suggests that pyroptosis, a form of programmed cell death distinct from apoptosis and necrosis, plays a critical role in the pathophysiology of S-AKI. This review examines the mechanisms of pyroptosis, focusing on inflammasome activation (e.g., NLRP3), caspase-mediated processes, and the role of Gasdermin D in renal tubular damage. We also discuss the contributions of inflammatory mediators, oxidative stress, and potential therapeutic strategies targeting pyroptosis, including inflammasome inhibitors, caspase inhibitors, and anti-inflammatory therapies. Lastly, we highlight the clinical implications and challenges in translating these findings into effective treatments, underscoring the need for personalized medicine approaches in managing S-AKI.
Collapse
Affiliation(s)
- Wenyu Wu
- The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, 510405, China
- Guangdong Clinical Research Academy of Chinese Medicine, Guangzhou, 510405, China
| | - Wanning Lan
- The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, 510405, China
- The First Clinical Medical School of Guangzhou University of Chinese Medicine, Guangzhou, 510405, China
| | - Xin Jiao
- The Second Clinical Medical College of Guangzhou University of Chinese Medicine, Guangzhou, 510405, China
- The Second Affiliated Hospital of Guangzhou University of Chinese Medicine (Guangdong Provincial Hospital of Chinese Medicine), Guangzhou, 510120, China
| | - Kai Wang
- The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, 510405, China
- The First Clinical Medical School of Guangzhou University of Chinese Medicine, Guangzhou, 510405, China
| | - Yawen Deng
- The Second Clinical Medical College of Guangzhou University of Chinese Medicine, Guangzhou, 510405, China
- The Second Affiliated Hospital of Guangzhou University of Chinese Medicine (Guangdong Provincial Hospital of Chinese Medicine), Guangzhou, 510120, China
| | - Rui Chen
- The Second Affiliated Hospital of Guangzhou University of Chinese Medicine (Guangdong Provincial Hospital of Chinese Medicine), Guangzhou, 510120, China.
- Guangdong Provincial Key Laboratory of Research On Emergency in TCM, Guangzhou, Guangdong, China.
| | - Ruifeng Zeng
- The Second Affiliated Hospital of Guangzhou University of Chinese Medicine (Guangdong Provincial Hospital of Chinese Medicine), Guangzhou, 510120, China.
- Guangdong Provincial Key Laboratory of Research On Emergency in TCM, Guangzhou, Guangdong, China.
| | - Jun Li
- The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, 510405, China.
- The First Clinical Medical School of Guangzhou University of Chinese Medicine, Guangzhou, 510405, China.
| |
Collapse
|
2
|
Bishr A, Atwa AM, El-Mokadem BM, El-Din MN. Canagliflozin potentially promotes renal protection against glycerol-induced acute kidney injury by activating the AMPK/SIRT1/FOXO-3a/PGC-1α and Nrf2/HO-1 pathways. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025:10.1007/s00210-025-04017-x. [PMID: 40257493 DOI: 10.1007/s00210-025-04017-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/12/2024] [Accepted: 03/04/2025] [Indexed: 04/22/2025]
Abstract
The reno-protective potential of canagliflozin (Cana), an inhibitor of the sodium glucose-linked co-transporter-2 (SGLT-2), has been demonstrated in different models of kidney injury. However, its potential role in preventing glycerol (Gly)-induced acute kidney injury (AKI) remains to be divulged. Therefore, the aim of this study is to investigate the potential reno-protective effect of Cana and its underlying mechanism in a rat model of Gly-induced AKI. Rats were randomly allocated into five groups: normal, Gly, Gly pretreated with 10 mg/kg Cana, Gly pretreated with Cana 25 mg/kg, and normal pretreated with Cana 25 mg/kg for 14 consecutive days. Pretreatment with Cana improved renal structure and enhanced kidney functions manifested by reducing serum creatinine and blood urea nitrogen, as well as renal contents of neutrophil gelatinase-associated lipocalin and kidney injury molecule. Moreover, Cana signified its anti-inflammatory effect by reducing the Gly-induced elevation in renal contents of nuclear factor-κB and interleuκin-6. Additionally, Cana augmented the defense enzymatic antioxidants superoxide dismutase (SOD), manganese-SOD, and heme oxygenase-1, besides increasing the protein expression of the antioxidant transcription factor nuclear factor erythroid 2-related factor 2 to point for its ability to correct redox balance. Cana also upregulated the protein expression of the 5' adenosine monophosphate-activated protein kinase (AMPK), Sirtuin1 (SIRT1), Forkhead box protein O3 (FOXO-3a), and peroxisome proliferator-activated receptor-gamma coactivator 1α (PGC-1α), as well as the transcriptional activity of growth arrest and DNA damage-inducible protein alpha (GAAD45a). In conclusion, Cana demonstrated potentially novel reno-protective mechanisms and mitigated the consequences of AKI through its antioxidant and anti-inflammatory properties, partially by activating the AMPK/SIRT1/FOXO-3a/PGC-1α pathway.
Collapse
Affiliation(s)
- Abeer Bishr
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Ahram Canadian University, Giza, Egypt.
| | - Ahmed M Atwa
- Department of Pharmacology and Toxicology, Egyptian Russian University, Cairo, Egypt
- College of Pharmacy, Al-Ayen Iraqi University, AUIQ, An Nasiriyah, Iraq
| | - Bassant M El-Mokadem
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Egyptian Chinese University, Cairo, Egypt
| | - Mahmoud Nour El-Din
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, University of Sadat City, Menoufia, Egypt
| |
Collapse
|
3
|
Sun C, Zhao X, Wang X, Yu Y, Shi H, Tang J, Sun S, Zhu S. Astragalus Polysaccharide Mitigates Rhabdomyolysis-Induced Acute Kidney Injury via Inhibition of M1 Macrophage Polarization and the cGAS-STING Pathway. J Inflamm Res 2024; 17:11505-11527. [PMID: 39735897 PMCID: PMC11675321 DOI: 10.2147/jir.s494819] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2024] [Accepted: 12/05/2024] [Indexed: 12/31/2024] Open
Abstract
Purpose This study aimed to examine the impact of APS on acute kidney injury induced by rhabdomyolysis (RIAKI), exploring its association with macrophage M1 polarization and elucidating the underlying mechanisms. Methods C57BL/6J mice were randomly assigned to one of three groups: a normal control group, a RIAKI model group, and an APS treatment group. Techniques such as flow cytometry and immunofluorescence were employed to demonstrate that APS can inhibit the transition of renal macrophages to the M1 phenotype in RIAKI. Furthermore, the raw264.7 macrophage cell line was chosen and induced into the M1 phenotype to further examine the impact of APS on this model and elucidate the underlying mechanism. Results Administration of APS led to a significant decrease in UREA levels by 25.2% and CREA levels by 60.9% within the model group. Also, APS exhibited an inhibitory effect on the infiltration of M1 macrophages and the cGAS-STING pathway in kidneys within the RIAKI, subsequently leading to decreased serum concentrations of IL-1β, IL-6 and TNF-α by 44.5%, 12.9%, and 10.3%, respectively, consistent with the results of in vitro experiments. Furthermore, APS exhibited an anti-apoptotic effect on MPC5 cells when co-cultured with M1 macrophages. Conclusion Astragalus polysaccharide (APS) potentially mitigated rhabdomyolysis-induced renal damage by impeding the M1 polarization of macrophages. This inherent mechanism might involve the suppression of the cGAS-STING pathway activation within macrophages. Furthermore, APS could endow protective effects on podocytes through the inhibition of apoptosis.
Collapse
Affiliation(s)
- Chuanchuan Sun
- Department of Traditional Chinese Medicine, The First Affiliated Hospital of Jinan University, Guangzhou, People’s Republic of China
- Department of Nephrology, The First Affiliated Hospital of Jinan University, Guangzhou, People’s Republic of China
| | - Xinhai Zhao
- Department of Traditional Chinese Medicine, The First Affiliated Hospital of Jinan University, Guangzhou, People’s Republic of China
| | - Xianghong Wang
- Department of Endocrinology and Metabolism, Zhuhai People’s Hospital (Zhuhai Clinical Medical College of Jinan University), Zhuhai, People’s Republic of China
| | - Yeye Yu
- Department of Traditional Chinese Medicine, The First Affiliated Hospital of Jinan University, Guangzhou, People’s Republic of China
| | - Heng Shi
- Department of Gastroenterology, The Central Hospital of Shaoyang, Shaoyang, People’s Republic of China
| | - Jun Tang
- The Fifth Affiliated Hospital of Zunyi Medical University, Zhuhai(Zhuhai Sixth People’s Hospital), Zhuhai, People’s Republic of China
| | - Shengyun Sun
- Department of Traditional Chinese Medicine, The First Affiliated Hospital of Jinan University, Guangzhou, People’s Republic of China
| | - Shiping Zhu
- Department of Traditional Chinese Medicine, The First Affiliated Hospital of Jinan University, Guangzhou, People’s Republic of China
| |
Collapse
|
4
|
Sabra MS, Allam EAH, Hassanein KMA. Sildenafil and furosemide nanoparticles as a novel pharmacological treatment for acute renal failure in rats. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2024; 397:7865-7879. [PMID: 38748227 PMCID: PMC11449963 DOI: 10.1007/s00210-024-03128-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/26/2024] [Accepted: 04/28/2024] [Indexed: 10/04/2024]
Abstract
Hospitalized patients often develop acute renal failure (ARF), which causes severe morbidity and death. This research investigates the potential renoprotective benefits of sildenafil and furosemide in glycerol-induced ARF, and measures kidney function metrics in response to nanoparticle versions of these medications. Inducing ARF is commonly done by injecting 50% glycerol intramuscularly. Rats underwent a 24-h period of dehydration and starvation before slaughter for renal function testing. We investigated urine analysis, markers of oxidative stress, histology of kidney tissue, immunohistochemistry analysis of caspase-3 and interleukin-1 beta (IL-1 β), kidney injury molecule-1 (KIM-1), and neutrophil gelatinase-associated lipocalin (NGAL), which are specific indicators of kidney tissue damage. The results of our study showed that the combination of sildenafil and furosemide, using both traditional and nanoparticle formulations, had a greater protective effect on the kidneys compared to using either drug alone. The recovery of renal tissue indicators, serum markers, and urine markers, which are indicative of organ damage, provides evidence of improvement. This was also indicated by the reduction in KIM-1 and NGAL tubular expression. The immunohistochemistry tests showed that the combination therapy, especially with the nanoforms, greatly improved the damaged cellular changes in the kidneys, as shown by higher levels of caspase-3 and IL-1β. According to the findings, a glycerol-induced rat model demonstrates that sildenafil and furosemide, either alone or in combination, in conventional or nanoparticulate forms, improve ARF dysfunction. The synergistic nanoparticulate compositions show remarkable effectiveness. This observation highlights the possible therapeutic implications for ARF treatment.
Collapse
Affiliation(s)
- Mahmoud S Sabra
- Pharmacology Department, Faculty of Veterinary Medicine, Assiut University, Assiut, 71526, Egypt.
| | - Essmat A H Allam
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Assiut University, Assiut, 71526, Egypt
| | - Khaled M Ahmed Hassanein
- Pathology and Clinical Pathology Department, Faculty of Veterinary Medicine, Assiut University, Assiut, 71526, Egypt
| |
Collapse
|
5
|
Safari Samangani M, Mehri S, Aminifard T, Jafarian A, Yazdani PF, Hosseinzadeh H. Effect of verbascoside against acute kidney injury induced by rhabdomyolysis in rats. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2024; 397:7939-7950. [PMID: 38753047 DOI: 10.1007/s00210-024-03144-1] [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: 02/15/2024] [Accepted: 05/03/2024] [Indexed: 10/04/2024]
Abstract
Rhabdomyolysis is a pathological condition caused by muscle tissue degradation. In this condition, intracellular contents enter the bloodstream, and acute kidney injury (AKI) develops. Verbascoside (VB) is one of the most common phenylethanoid glycosides and has antioxidant and anti-inflammatory effects. This study investigated the effects of VB on AKI induced by rhabdomyolysis in rats. Male Wistar rats were divided into six groups (n = 6): (1) control group (normal saline), (2) 50% glycerol (10 ml/kg, IM, single injection, only on the first day), (3)-(5) 50% glycerol (same as group 2) + VB (30, 60, and 100 mg/kg, IP, 4 days), and (6) VB (100 mg/kg). Serum and kidney tissue samples were collected on day 5. Subsequently, serum creatinine (Cr), blood urea nitrogen (BUN), renal glutathione (GSH), malondialdehyde (MDA), lipocalin associated with neutrophil gelatinase (NGAL), tumor necrosis factor-alpha (TNF-α), and pathological changes were investigated. The injection of glycerol elevated levels of kidney damage markers, including Cr and BUN in serum, MDA, TNF-α, and NGAL, along with a reduction in GSH levels in the kidney tissue. The administration of VB (100 mg/kg) significantly lowered the levels of these markers, indicating the therapeutic effect of VB against AKI caused by rhabdomyolysis. Histopathological examinations revealed enhanced myoglobin cast formation and tubular necrosis in the glycerol group, which was reduced in rats that received VB, although this reduction did not reach statistical significance. VB can reduce rhabdomyolysis-induced AKI through its anti-inflammatory and antioxidant effects and decrease kidney damage severity.
Collapse
Affiliation(s)
- Maryam Safari Samangani
- Department of Pharmacodynamics and Toxicology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Soghra Mehri
- Department of Pharmacodynamics and Toxicology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
- Pharmaceutical Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Tahereh Aminifard
- Department of Pharmacodynamics and Toxicology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Amirhossein Jafarian
- Department of Pathology, Ghaem Hospital, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Pooneh Fallah Yazdani
- Department of Pharmacodynamics and Toxicology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Hossein Hosseinzadeh
- Department of Pharmacodynamics and Toxicology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran.
- Pharmaceutical Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
6
|
Kazeminia S, Eirin A. Role of mitochondria in endogenous renal repair. Clin Sci (Lond) 2024; 138:963-973. [PMID: 39076039 PMCID: PMC11410300 DOI: 10.1042/cs20231331] [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: 04/24/2024] [Revised: 07/03/2024] [Accepted: 07/15/2024] [Indexed: 07/31/2024]
Abstract
Renal tubules have potential to regenerate and repair after mild-to-moderate injury. Proliferation of tubular epithelial cells represents the initial step of this reparative process. Although for many years, it was believed that proliferating cells originated from a pre-existing intra-tubular stem cell population, there is now consensus that surviving tubular epithelial cells acquire progenitor properties to regenerate the damaged kidney. Scattered tubular-like cells (STCs) are dedifferentiated adult renal tubular epithelial cells that arise upon injury and contribute to renal self-healing and recovery by replacing lost neighboring tubular epithelial cells. These cells are characterized by the co-expression of the stem cell surface markers CD133 and CD24, as well as mesenchymal and kidney injury markers. Previous studies have shown that exogenous delivery of STCs ameliorates renal injury and dysfunction in murine models of acute kidney injury, underscoring the regenerative potential of this endogenous repair system. Although STCs contain fewer mitochondria than their surrounding terminally differentiated tubular epithelial cells, these organelles modulate several important cellular functions, and their integrity and function are critical to preserve the reparative capacity of STCs. Recent data suggest that the microenviroment induced by cardiovascular risk factors, such as obesity, hypertension, and renal ischemia may compromise STC mitochondrial integrity and function, limiting the capacity of these cells to repair injured renal tubules. This review summarizes current knowledge of the contribution of STCs to kidney repair and discusses recent insight into the key role of mitochondria in modulating STC function and their vulnerability in the setting of cardiovascular disease.
Collapse
Affiliation(s)
- Sara Kazeminia
- Department of Internal Medicine, Division of Nephrology and Hypertension, Mayo Clinic, Rochester, MN, U.S.A
| | - Alfonso Eirin
- Department of Internal Medicine, Division of Nephrology and Hypertension, Mayo Clinic, Rochester, MN, U.S.A
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN, U.S.A
| |
Collapse
|
7
|
Yang L, Shen Y, Li W, Zha B, Xu W, Ding H. Elevated plasma myoglobin level is closely associated with type 2 diabetic kidney disease. J Diabetes 2024; 16:e13508. [PMID: 38036859 PMCID: PMC10925879 DOI: 10.1111/1753-0407.13508] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/07/2023] [Revised: 10/31/2023] [Accepted: 11/12/2023] [Indexed: 12/02/2023] Open
Abstract
BACKGROUND Diabetic kidney disease (DKD) is the most frequent complication in patients with type 2 diabetes mellitus (T2DM). It causes a chronic and progressive decline in kidney function, and ultimately patients require renal replacement therapy. To date, an increasing number of clinical studies have been conducted to explore the potential and novel biomarkers, which can advance the diagnosis, estimate the prognosis, and optimize the therapeutic strategies at the early stage of DKD. In the current study, we sought to investigate the association of plasma myoglobin with DKD. METHODS A total of 355 T2DM patients with DKD and 710 T2DM patients without DKD were enrolled in this study. Laboratory parameters including blood cell count, hemoglobin A1c, biochemical parameters, and plasma myoglobin were recorded. Patients were classified on admission according to the tertile of myoglobin and clinical parameters were compared between the groups. Pearson correlation analysis, linear regression, logistic regression, receiver operating characteristics (ROC) analysis, and spline regression were performed. RESULTS Plasma myoglobin significantly increased in patients with DKD and was associated with renal function and inflammatory parameters. Plasma myoglobin was an independent risk factor for the development of DKD. The area under ROC curve of myoglobin was 0.831. Spline regression showed that there was a significant linear association between DKD incidence and a high level of plasma myoglobin when it exceeded 36.4 mg/mL. CONCLUSIONS This study shows that elevated plasma myoglobin level is closely associated with the development of kidney injury in patients with T2DM.
Collapse
Affiliation(s)
- Lin Yang
- Department of Nephrology, Shanghai Fifth People's HospitalFudan UniversityShanghaiChina
| | - Yan Shen
- Department of Endocrinology, Shanghai Fifth People's HospitalFudan UniversityShanghaiChina
| | - Wenxiao Li
- Department of Endocrinology, Shanghai Fifth People's HospitalFudan UniversityShanghaiChina
- Center of Community‐Based Health ResearchFudan UniversityShanghaiChina
- Jiangchuan Community Health Service CenterShanghaiChina
| | - Bingbing Zha
- Department of Endocrinology, Shanghai Fifth People's HospitalFudan UniversityShanghaiChina
| | - Wenjun Xu
- Department of NephrologyZhejiang Kaihua County Hospital of Chinese MedicineZhejiangChina
| | - Heyuan Ding
- Department of Endocrinology, Shanghai Fifth People's HospitalFudan UniversityShanghaiChina
| |
Collapse
|
8
|
Guerrero-Hue M, Vallejo-Mudarra M, García-Caballero C, Córdoba-David GM, Palomino-Antolín A, Herencia C, Vendrell-Casana B, Rubio-Navarro A, Egido J, Blanco-Colio LM, Moreno JA. Tweak/Fn14 system is involved in rhabdomyolysis-induced acute kidney injury. Biomed Pharmacother 2023; 169:115925. [PMID: 38007933 DOI: 10.1016/j.biopha.2023.115925] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2023] [Revised: 11/07/2023] [Accepted: 11/20/2023] [Indexed: 11/28/2023] Open
Abstract
BACKGROUND Rhabdomyolysis is a severe clinical syndrome associated to acute kidney injury (AKI) and chronic kidney disease (CKD). TWEAK/Fn14 signaling axis regulates renal inflammation and tubular cell death. However, the functional role of TWEAK/Fn14 in rhabdomyolysis remains unknown. METHODS Rhabdomyolysis was induced in wild-type, TWEAK- and Fn14-deficient mice or mice treated with TWEAK blocking antibody. Renal injury, inflammation, fibrosis and cell death were assessed. Additionally, we performed in vivo and in vitro studies to explore the possible signalling pathways involved in Fn14 regulation. FINDINGS Fn14 renal expression was increased in mice with rhabdomyolysis, correlating with decline of renal function. Mechanistically, myoglobin (Mb) induced Fn14 expression via ERK and p38 pathway, whereas Nrf2 activation diminished Mb-mediated Fn14 upregulation in cultured renal cells. TWEAK or Fn14 genetic depletion ameliorated rhabdomyolysis-associated loss of renal function, histological damage, tubular cell death, inflammation, and expression of both tubular and endothelial injury markers. Deficiency of TWEAK or Fn14 also decreased long-term renal inflammation and fibrosis in mice with rhabdomyolysis. Finally, pharmacological treatment with a blocking TWEAK antibody diminished the expression of acute renal injury markers and cell death and lessened residual kidney fibrosis and chronic inflammation in rhabdomyolysis. INTERPRETATION TWEAK/Fn14 axis participates in the pathogenesis of rhabdomyolysis-AKI and subsequent AKI-CKD transition. Blockade of this signaling pathway may represent a promising therapeutic strategy for reducing rhabdomyolysis-mediated renal injury. FUNDING Spanish Ministry of Science and Innovation, ISCIII and Junta de Andalucía.
Collapse
Affiliation(s)
- Melania Guerrero-Hue
- Maimonides Biomedical Research Institute of Cordoba (IMIBIC), Hospital Universitario Reina Sofía, Cordoba, Spain
| | - Mercedes Vallejo-Mudarra
- Maimonides Biomedical Research Institute of Cordoba (IMIBIC), Hospital Universitario Reina Sofía, Cordoba, Spain
| | - Cristina García-Caballero
- Maimonides Biomedical Research Institute of Cordoba (IMIBIC), Hospital Universitario Reina Sofía, Cordoba, Spain
| | - Gina Marcela Córdoba-David
- Renal, Vascular and Diabetes Research Lab, Fundación Instituto de Investigaciones Sanitarias-Fundación Jiménez Díaz, Autonoma University, Madrid, Spain
| | - Alejandra Palomino-Antolín
- Molecular Neuroinflammation and Neuronal Plasticity Research Laboratory, Hospital Universitario Santa Cristina, Instituto de Investigación Sanitaria-Hospital Universitario de la Princesa, Madrid, Spain; Instituto Teófilo Hernando, Departamento de Farmacología y Terapéutica, Facultad de Medicina, Autonoma University, Madrid, Spain
| | - Carmen Herencia
- Renal, Vascular and Diabetes Research Lab, Fundación Instituto de Investigaciones Sanitarias-Fundación Jiménez Díaz, Autonoma University, Madrid, Spain
| | - Beatriz Vendrell-Casana
- Maimonides Biomedical Research Institute of Cordoba (IMIBIC), Hospital Universitario Reina Sofía, Cordoba, Spain
| | - Alfonso Rubio-Navarro
- Laboratory of Advanced Therapies: Differentiation, Regeneration and Cancer (CTS-963). Center of Biomedical Research. University of Granada, Spain; Instituto de Investigación Biosanitaria de Granada (ibs.GRANADA), Granada, Spain; Excellence Research Unit "Modeling Nature" (MNat), University of Granada, Granada, Spain
| | - Jesús Egido
- Renal, Vascular and Diabetes Research Lab, Fundación Instituto de Investigaciones Sanitarias-Fundación Jiménez Díaz, Autonoma University, Madrid, Spain; Centre of Biomedical Research in Network of Diabetes and Metabolic Disease Associated (CIBERDEM), Madrid, Spain
| | - Luis Miguel Blanco-Colio
- Renal, Vascular and Diabetes Research Lab, Fundación Instituto de Investigaciones Sanitarias-Fundación Jiménez Díaz, Autonoma University, Madrid, Spain; Centre of Biomedical Research in Network of Cardiovascular Diseases (CIBERCV), Madrid, Spain
| | - Juan Antonio Moreno
- Maimonides Biomedical Research Institute of Cordoba (IMIBIC), Hospital Universitario Reina Sofía, Cordoba, Spain; Centre of Biomedical Research in Network of Cardiovascular Diseases (CIBERCV), Madrid, Spain; Department of Cell Biology, Physiology and Immunology, University of Cordoba, Cordoba, Spain.
| |
Collapse
|
9
|
Liang J, Liu Y. Animal Models of Kidney Disease: Challenges and Perspectives. KIDNEY360 2023; 4:1479-1493. [PMID: 37526653 PMCID: PMC10617803 DOI: 10.34067/kid.0000000000000227] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/17/2023] [Accepted: 07/24/2023] [Indexed: 08/02/2023]
Abstract
Kidney disease is highly prevalent and affects approximately 850 million people worldwide. It is also associated with high morbidity and mortality, and current therapies are incurable and often ineffective. Animal models are indispensable for understanding the pathophysiology of various kidney diseases and for preclinically testing novel remedies. In the last two decades, rodents continue to be the most used models for imitating human kidney diseases, largely because of the increasing availability of many unique genetically modified mice. Despite many limitations and pitfalls, animal models play an essential and irreplaceable role in gaining novel insights into the mechanisms, pathologies, and therapeutic targets of kidney disease. In this review, we highlight commonly used animal models of kidney diseases by focusing on experimental AKI, CKD, and diabetic kidney disease. We briefly summarize the pathological characteristics, advantages, and drawbacks of some widely used models. Emerging animal models such as mini pig, salamander, zebrafish, and drosophila, as well as human-derived kidney organoids and kidney-on-a-chip are also discussed. Undoubtedly, careful selection and utilization of appropriate animal models is of vital importance in deciphering the mechanisms underlying nephropathies and evaluating the efficacy of new treatment options. Such studies will provide a solid foundation for future diagnosis, prevention, and treatment of human kidney diseases.
Collapse
Affiliation(s)
- Jianqing Liang
- Division of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou, China
- State Key Laboratory of Organ Failure Research, National Clinical Research Center of Kidney Disease, Guangdong Provincial Institute of Nephrology, Guangzhou, China
| | - Youhua Liu
- Division of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou, China
- State Key Laboratory of Organ Failure Research, National Clinical Research Center of Kidney Disease, Guangdong Provincial Institute of Nephrology, Guangzhou, China
| |
Collapse
|
10
|
Mahmood YS, Kathem SH. Protective effect of citronellol in rhabdomyolysis-induced acute kidney injury in mice. J Med Life 2023; 16:1057-1061. [PMID: 37900084 PMCID: PMC10600660 DOI: 10.25122/jml-2023-0102] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2023] [Accepted: 05/03/2023] [Indexed: 10/31/2023] Open
Abstract
Acute kidney injury (AKI) is a serious pathophysiological event consequent to rhabdomyolysis. Inflammatory mechanisms play a role in the development of rhabdomyolysis-induced AKI. Citronellol (CT) is a naturally occurring monoterpene in essential oils of aromatic plant species. In this study, we explored the protective effects of citronellol on AKI resulting from glycerol-induced rhabdomyolysis. Rhabdomyolysis was induced by a single intramuscular injection of glycerol 50% (10mg/kg) in the thigh caudal muscle. Four groups of mice were assigned, including a control group, a group administered with glycerol to induce AKI as a model, a group treated with glycerol plus 50mg/kg CT, and a group treated with glycerol plus 100mg/kg CT. The renal function of mice from all groups was evaluated using kidney histopathological changes and kidney injury molecule-1 (KIM-1). Myoglobin levels were measured to detect rhabdomyolysis. Apoptosis was evaluated by renal cleaved caspase-3 and BAX levels. Both doses of citronellol (50mg/kg and 100mg/kg) significantly reduced KIM-1 mRNA expression and myoglobin levels compared to the glycerol group. In addition, citronellol resulted in lower cleaved caspase-3 and BAX in the renal tissue, indicating that citronellol exerted an anti-apoptotic effect in AKI. Citronellol showed a reno-protective effect against rhabdomyolysis-induced AKI, which may be attributed to its anti-apoptotic effects.
Collapse
Affiliation(s)
| | - Sarmed Hashim Kathem
- Department of Pharmacology and Toxicology, College of Pharmacy, University of Baghdad, Baghdad, Iraq
| |
Collapse
|
11
|
Administration of a single dose of lithium ameliorates rhabdomyolysis-associated acute kidney injury in rats. PLoS One 2023; 18:e0281679. [PMID: 36795689 PMCID: PMC9934413 DOI: 10.1371/journal.pone.0281679] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2022] [Accepted: 01/27/2023] [Indexed: 02/17/2023] Open
Abstract
Rhabdomyolysis is characterized by muscle damage and leads to acute kidney injury (AKI). Clinical and experimental studies suggest that glycogen synthase kinase 3β (GSK3β) inhibition protects against AKI basically through its critical role in tubular epithelial cell apoptosis, inflammation and fibrosis. Treatment with a single dose of lithium, an inhibitor of GSK3β, accelerated recovery of renal function in cisplatin and ischemic/reperfusion-induced AKI models. We aimed to evaluate the efficacy of a single dose of lithium in the treatment of rhabdomyolysis-induced AKI. Male Wistar rats were allocated to four groups: Sham, received saline 0.9% intraperitoneally (IP); lithium (Li), received a single IP injection of lithium chloride (LiCl) 80 mg/kg body weight (BW); glycerol (Gly), received a single dose of glycerol 50% 5 mL/kg BW intramuscular (IM); glycerol plus lithium (Gly+Li), received a single dose of glycerol 50% IM plus LiCl IP injected 2 hours after glycerol administration. After 24 hours, we performed inulin clearance experiments and collected blood / kidney / muscle samples. Gly rats exhibited renal function impairment accompanied by kidney injury, inflammation and alterations in signaling pathways for apoptosis and redox state balance. Gly+Li rats showed a remarkable improvement in renal function as well as kidney injury score, diminished CPK levels and an overstated decrease of renal and muscle GSK3β protein expression. Furthermore, administration of lithium lowered the amount of macrophage infiltrate, reduced NFκB and caspase renal protein expression and increased the antioxidant component MnSOD. Lithium treatment attenuated renal dysfunction in rhabdomyolysis-associated AKI by improving inulin clearance and reducing CPK levels, inflammation, apoptosis and oxidative stress. These therapeutic effects were due to the inhibition of GSK3β and possibly associated with a decrease in muscle injury.
Collapse
|
12
|
Abstract
Pyroptosis is a form of regulated cell death that is mediated by the membrane-targeting, pore-forming gasdermin family of proteins. Pyroptosis was initially described as a caspase 1- and inflammasome-dependent cell death pathway typified by the loss of membrane integrity and the secretion of cytokines such as IL-1β. However, gasdermins are now recognized as the principal effectors of this form of regulated cell death; activated gasdermins insert into cell membranes, where they form pores that result in the secretion of cytokines, alarmins and damage-associated molecular patterns and cause cell membrane rupture. It is now evident that gasdermins can be activated by inflammasome- and caspase-independent mechanisms in multiple cell types and that crosstalk occurs between pyroptosis and other cell death pathways. Although they are important for host antimicrobial defence, a growing body of evidence supports the notion that pyroptosis and gasdermins have pathological roles in cancer and several non-microbial diseases involving the gut, liver and skin. The well-documented roles of inflammasome activity and apoptosis pathways in kidney diseases suggests that gasdermins and pyroptosis may also be involved to some extent. However, despite some evidence for involvement of pyroptosis in the context of acute kidney injury and chronic kidney disease, our understanding of gasdermin biology and pyroptosis in the kidney remains limited.
Collapse
|
13
|
Guo W, Wang Y, Wu Y, Liu J, Li Y, Wang J, Ou S, Wu W. Integration of transcriptomics and metabolomics reveals the molecular mechanisms underlying the effect of nafamostat mesylate on rhabdomyolysis-induced acute kidney injury. Front Pharmacol 2022; 13:931670. [PMID: 36532745 PMCID: PMC9748812 DOI: 10.3389/fphar.2022.931670] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Accepted: 11/17/2022] [Indexed: 11/09/2023] Open
Abstract
Objective: To investigate the role and mechanisms of action of nafamostat mesylate (NM) in rhabdomyolysis-induced acute kidney injury (RIAKI). Methods: RIAKI rats were assigned into control group (CN), RIAKI group (RM), and NM intervention group (NM). Inflammatory cytokines and proenkephalin a 119-159 (PENKID) were assessed. Cell apoptosis and glutathione peroxidase-4 (GPX4) were detected using TUNEL assay and immunohistochemical staining. Mitochondrial membrane potential (MMP) was detected by JC-1 dye. The expression of genes and metabolites after NM intervention was profiled using transcriptomic and metabolomic analysis. The differentially expressed genes (DEGs) were validated using qPCR. The KEGG and conjoint analysis of transcriptome and metabolome were used to analyze the enriched pathways and differential metabolites. The transcription factors were identified based on the animal TFDB 3.0 database. Results: Serum creatinine, blood urea nitrogen, and PENKID were remarkably higher in the RM group and lower in the NM group compared to the CN group. Pro-inflammatory cytokines increased in the RM group and notably decreased following NM treatment compared to the CN group. Tubular pathological damages were markedly attenuated and renal cell apoptosis was reduced significantly in the NM group compared to the RM group. The expression of GPX4 was lower in the RM group compared to the CN group, and it increased significantly after NM treatment. A total of 294 DEGs were identified in the RM group compared with the NM group, of which 192 signaling pathways were enriched, and glutathione metabolism, IL-17 signaling, and ferroptosis-related pathways were the top-ranking pathways. The transcriptional levels of Anpep, Gclc, Ggt1, Mgst2, Cxcl13, Rgn, and Akr1c1 were significantly different between the NM and RM group. Gclc was the key gene contributing to NM-mediated renal protection in RIAKI. Five hundred and five DEGs were annotated. Compared with the RM group, most of the upregulated DEGs in the NM group belonged to Glutathione metabolism, whereas most of the downregulated DEGs were related to the transcription factor Cytokine-cytokine receptor interaction. Conclusion: NM protects the kidneys against RIAKI, which is mainly associated with NM mediated regulation of glutathione metabolism, inflammatory response, ferroptosis-related pathways, and the related key DEGs. Targeting these DEGs might emerge as a potential molecular therapy for RIAKI.
Collapse
Affiliation(s)
- Wenli Guo
- Metabolic Vascular Disease Key Laboratory, Sichuan Clinical Research Center for Nephropathy, Department of Nephrology, Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
- Department of Nephrology and Rheumatology, Sichuan Provincial People’s Hospital Qionglai Hospital, Medical Center Hospital Of Qionglai City. Chengdu, Sichuan, China
| | - Yu Wang
- Metabolic Vascular Disease Key Laboratory, Sichuan Clinical Research Center for Nephropathy, Department of Nephrology, Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
| | - Yuxuan Wu
- Metabolic Vascular Disease Key Laboratory, Sichuan Clinical Research Center for Nephropathy, Department of Nephrology, Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
| | - Jiang Liu
- Metabolic Vascular Disease Key Laboratory, Sichuan Clinical Research Center for Nephropathy, Department of Nephrology, Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
| | - Ying Li
- Metabolic Vascular Disease Key Laboratory, Sichuan Clinical Research Center for Nephropathy, Department of Nephrology, Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
| | - Jing Wang
- Metabolic Vascular Disease Key Laboratory, Sichuan Clinical Research Center for Nephropathy, Department of Nephrology, Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
| | - Santao Ou
- Metabolic Vascular Disease Key Laboratory, Sichuan Clinical Research Center for Nephropathy, Department of Nephrology, Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
| | - Weihua Wu
- Metabolic Vascular Disease Key Laboratory, Sichuan Clinical Research Center for Nephropathy, Department of Nephrology, Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
| |
Collapse
|
14
|
Madkour AH, Helal MG, Said E, Salem HA. Dose-dependent renoprotective impact of Lactoferrin against glycerol-induced rhabdomyolysis and acute kidney injury. Life Sci 2022; 302:120646. [PMID: 35595070 DOI: 10.1016/j.lfs.2022.120646] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2022] [Revised: 05/11/2022] [Accepted: 05/12/2022] [Indexed: 11/20/2022]
Abstract
Acute kidney injury (AKI) is a clinical disorder with a serious impact on the quality of patients' lives. Considering its increased worldwide prevalence, investigating novel therapeutic approaches for the management of AKI has been inevitable. Lactoferrin (LF), a glycoprotein belonging to the transferrin family, is known to play an important role in regulating iron homeostasis. This study aimed to evaluate the renoprotective effect of LF (30, 100, and 300 mg/kg orally) against glycerol (GLY)-induced rhabdomyolysis (RM) in rats. RM was induced by a single intramuscular injection of GLY 50% (10 mL/kg) after 24-h water deprivation in male Sprague-Dawley rats. LF administration conferred significant dose-dependent renoprotective impact against GLY-induced RM as evidenced by the decreased renal/somatic index and the significant improvement in renal functions as confirmed by the significant increase in creatinine clearance, decrease in serum creatinine and blood urea nitrogen, and improvement in albuminuria and proteinuria. Redox homeostasis was significantly restored in a dose-dependent manner as well. Moreover, serum interleukin-1β (IL-1β) was significantly decreased with a parallel significant decrease in renal NOD-like receptor family pyrin domain containing 3 (NLRP3) and thioredoxin interacting protein (TXNIP), kidney injury molecule-1 (KIM-1), caspase-3 expression, nuclear factor kappa B (NF-κB), cluster of differentiation (CD68) expression, and a significant increase in renal nuclear factor erythroid 2-related factor 2 (NRF2) expression. Ultimately, LF administration was associated with a significant amelioration of GLY-induced renal necrotic and inflammatory alterations. In conclusion, the observed dose-dependent nephroprotective effect of LF can be attributed to its modulatory impact on inflammatory/apoptotic/oxidative signaling.
Collapse
Affiliation(s)
- Ahmed H Madkour
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Mansoura University, 35516 Mansoura, Egypt
| | - Manar G Helal
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Mansoura University, 35516 Mansoura, Egypt
| | - Eman Said
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Mansoura University, 35516 Mansoura, Egypt; Faculty of Pharmacy, New Mansoura University, 7723730 New Mansoura, Egypt.
| | - Hatem A Salem
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Mansoura University, 35516 Mansoura, Egypt
| |
Collapse
|
15
|
Panahipour L, Cervantes LCC, Oladzad Abbasabadi A, Sordi MB, Kargarpour Z, Gruber R. Blocking of Caspases Exerts Anti-Inflammatory Effects on Periodontal Cells. LIFE (BASEL, SWITZERLAND) 2022; 12:life12071045. [PMID: 35888133 PMCID: PMC9316350 DOI: 10.3390/life12071045] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/03/2022] [Revised: 06/22/2022] [Accepted: 06/30/2022] [Indexed: 11/16/2022]
Abstract
Periodontitis is an inflammatory process that is associated with caspase activity. Caspases could thus become molecular targets for the modulation of the inflammatory response to harmful factors, such as lipopolysaccharides (LPS) and TNFα. Here, the impact of the pan-caspase inhibitor Z-VAD-FMK (carbobenzoxy-valyl-alanyl-aspartyl-[O-methyl]-fluoro-methyl ketone) on the modulation of the LPS-induced inflammatory response of murine RAW 264.7 cells and primary macrophages was examined. Moreover, the inflammatory responses of human gingival fibroblasts, HSC2 oral squamous carcinoma cells and murine ST2 mesenchymal fibroblasts when exposed to TNFα were studied. Data showed that Z-VAD-FMK significantly lowered the inflammatory response of RAW 264.7 cells and primary macrophages, as indicated by the expression of IL1 and IL6. In murine ST2 mesenchymal fibroblasts, the TNFα-induced expression of CCL2 and CCL5 was significantly reduced. In human gingival fibroblasts and HSC2 cells, Z-VAD-FMK considerably reduced the TNFα-induced expression of CXCL8 and CXCL10. These findings suggest that pharmacological blocking of caspases in an inflammatory environment lowers the expression of cytokines and chemokines in periodontal cells.
Collapse
Affiliation(s)
- Layla Panahipour
- Department of Oral Biology, University Clinic of Dentistry, Medical University of Vienna, Sensengasse 2a, 1090 Vienna, Austria; (L.P.); (L.C.C.C.); (A.O.A.); (M.B.S.); (Z.K.)
| | - Lara Cristina Cunha Cervantes
- Department of Oral Biology, University Clinic of Dentistry, Medical University of Vienna, Sensengasse 2a, 1090 Vienna, Austria; (L.P.); (L.C.C.C.); (A.O.A.); (M.B.S.); (Z.K.)
- Department of Diagnosis and Surgery, School of Dentistry, São Paulo State University (UNESP), Araçatuba, Sao Paulo 16015-050, Brazil
| | - Azarakhsh Oladzad Abbasabadi
- Department of Oral Biology, University Clinic of Dentistry, Medical University of Vienna, Sensengasse 2a, 1090 Vienna, Austria; (L.P.); (L.C.C.C.); (A.O.A.); (M.B.S.); (Z.K.)
| | - Mariane Beatriz Sordi
- Department of Oral Biology, University Clinic of Dentistry, Medical University of Vienna, Sensengasse 2a, 1090 Vienna, Austria; (L.P.); (L.C.C.C.); (A.O.A.); (M.B.S.); (Z.K.)
- Centre for Research on Dental Implants (CEPID), Department of Dentistry, Federal University of Santa Catarina (UFSC), Florianopolis 88040-900, Brazil
| | - Zahra Kargarpour
- Department of Oral Biology, University Clinic of Dentistry, Medical University of Vienna, Sensengasse 2a, 1090 Vienna, Austria; (L.P.); (L.C.C.C.); (A.O.A.); (M.B.S.); (Z.K.)
| | - Reinhard Gruber
- Department of Oral Biology, University Clinic of Dentistry, Medical University of Vienna, Sensengasse 2a, 1090 Vienna, Austria; (L.P.); (L.C.C.C.); (A.O.A.); (M.B.S.); (Z.K.)
- Department of Periodontology, School of Dental Medicine, University of Bern, Freiburgstrasse 7, 3010 Bern, Switzerland
- Austrian Cluster for Tissue Regeneration, Donaueschingenstraße 13, 1200 Vienna, Austria
- Correspondence:
| |
Collapse
|
16
|
Balzer MS, Doke T, Yang YW, Aldridge DL, Hu H, Mai H, Mukhi D, Ma Z, Shrestha R, Palmer MB, Hunter CA, Susztak K. Single-cell analysis highlights differences in druggable pathways underlying adaptive or fibrotic kidney regeneration. Nat Commun 2022; 13:4018. [PMID: 35821371 PMCID: PMC9276703 DOI: 10.1038/s41467-022-31772-9] [Citation(s) in RCA: 110] [Impact Index Per Article: 36.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Accepted: 07/01/2022] [Indexed: 01/14/2023] Open
Abstract
The kidney has tremendous capacity to repair after acute injury, however, pathways guiding adaptive and fibrotic repair are poorly understood. We developed a model of adaptive and fibrotic kidney regeneration by titrating ischemic injury dose. We performed detailed biochemical and histological analysis and profiled transcriptomic changes at bulk and single-cell level (> 110,000 cells) over time. Our analysis highlights kidney proximal tubule cells as key susceptible cells to injury. Adaptive proximal tubule repair correlated with fatty acid oxidation and oxidative phosphorylation. We identify a specific maladaptive/profibrotic proximal tubule cluster after long ischemia, which expresses proinflammatory and profibrotic cytokines and myeloid cell chemotactic factors. Druggability analysis highlights pyroptosis/ferroptosis as vulnerable pathways in these profibrotic cells. Pharmacological targeting of pyroptosis/ferroptosis in vivo pushed cells towards adaptive repair and ameliorates fibrosis. In summary, our single-cell analysis defines key differences in adaptive and fibrotic repair and identifies druggable pathways for pharmacological intervention to prevent kidney fibrosis.
Collapse
Affiliation(s)
- Michael S Balzer
- Renal, Electrolyte, and Hypertension Division, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
- Institute for Diabetes, Obesity and Metabolism, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Tomohito Doke
- Renal, Electrolyte, and Hypertension Division, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
- Institute for Diabetes, Obesity and Metabolism, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Ya-Wen Yang
- Renal, Electrolyte, and Hypertension Division, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
- Institute for Diabetes, Obesity and Metabolism, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Daniel L Aldridge
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Hailong Hu
- Renal, Electrolyte, and Hypertension Division, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
- Institute for Diabetes, Obesity and Metabolism, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Hung Mai
- Renal, Electrolyte, and Hypertension Division, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
- Institute for Diabetes, Obesity and Metabolism, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Dhanunjay Mukhi
- Renal, Electrolyte, and Hypertension Division, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
- Institute for Diabetes, Obesity and Metabolism, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Ziyuan Ma
- Renal, Electrolyte, and Hypertension Division, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
- Institute for Diabetes, Obesity and Metabolism, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Rojesh Shrestha
- Renal, Electrolyte, and Hypertension Division, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
- Institute for Diabetes, Obesity and Metabolism, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Matthew B Palmer
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Christopher A Hunter
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Katalin Susztak
- Renal, Electrolyte, and Hypertension Division, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA.
- Institute for Diabetes, Obesity and Metabolism, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA.
- Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA.
| |
Collapse
|
17
|
Liu Y, Yu M, Chen L, Liu J, Li X, Zhang C, Xiang X, Li X, Lv Q. Systemic Review of Animal Models Used in the Study of Crush Syndrome. Shock 2022; 57:469-478. [PMID: 35066515 DOI: 10.1097/shk.0000000000001911] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
ABSTRACT Crush syndrome (CS), also known as traumatic rhabdomyolysis, is the leading cause of death following extrication from structural collapse due to earthquakes. Due to the unfeasibility of human studies, animal models are used to study crush syndrome pathophysiology, including biochemistry and treatment regimes. The aim of this systematic literature review was to identify the differences and benefits of various animal models used in the study of CS and provide valuable information for design of future research. A systematic search was conducted in two methods: with the filters "(crush syndrome) AND (crush muscle injury)" and with the keywords "(crush syndrome) AND (animal model)" covering all articles in the PubMed databases. The search generated 378 articles. After screening abstracts, 91 articles were retrieved and read, then 11 repeated articles were removed and 2 reference papers were included. We finally reviewed 82 original articles. There appear to be two primary methods employed for inducing crush syndrome in animal models, which are chemically induced injury and physically induced injury. Chemical method mainly includes intramuscular (IM) injection of tissue extract solution and IM injection of 50% glycerine. Physical method can be classified into invasive and non-invasive physical compression by elasticated material, inflatable band and heavy load. Various species of animals have been used to study CS, including mice (13.4%), rats (68.3%), rabbits (11.0%), canines (4.9%), goats (1.2%), and pigs (1.2%). Small animals are suitable for researches exploring the mechanism of disease or drug efficacy while large animals can work better with clinical application-related researches. In regard to the choice of modeling method, compressing the certain muscle of animals by heavy things is superior to others to cause systemic trauma-related rhabdomyolysis signs. In addition, due to the significant burden of crush injuries on animals, further attention shall be paid to the selection of the most suitable anesthetics and appropriate analgesics.
Collapse
Affiliation(s)
- Yahua Liu
- Emergency Department, Chinese PLA General Hospital (The Third Center), Beijing, China
- Beijing Key Laboratory of Disaster Rescue Medicine, Beijing, China
| | - Mengyang Yu
- General Medicine Department, Chinese PLA General Hospital (The Third Center), Beijing, China
| | - Li Chen
- General Medicine Department, Chinese PLA general Hospital (The First Center), Beijing, China
| | - Jing Liu
- Pathology Department, Chinese PLA General Hospital (The Third Center), Beijing, China
| | - Xin Li
- Emergency Department, Chinese PLA General Hospital (The Third Center), Beijing, China
| | - Chengying Zhang
- General Medicine Department, Chinese PLA General Hospital (The Third Center), Beijing, China
| | - Xueyuan Xiang
- Urology, Chinese PLA General Hospital (The Third Center), Beijing, China
| | - Xiaoxue Li
- Beijing Key Laboratory of Disaster Rescue Medicine, Beijing, China
- Chinese PLA General Hospital (Innovative Medicine Division), Beijing, China
| | - Qi Lv
- Institute of Disaster Medicine, Tianjin University, Tianjin, China
- Tianjin Key Laboratory of Disaster Medicine Technology, Tianjin, China
| |
Collapse
|
18
|
Song Z, Gong Q, Guo J. Pyroptosis: Mechanisms and Links with Fibrosis. Cells 2021; 10:cells10123509. [PMID: 34944017 PMCID: PMC8700428 DOI: 10.3390/cells10123509] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2021] [Revised: 12/09/2021] [Accepted: 12/10/2021] [Indexed: 02/06/2023] Open
Abstract
Fibrosis is responsible for approximately 45% of deaths in the industrialized world and has been a major global healthcare burden. Excessive fibrosis is the primary cause of organ failure. However, there are currently no approved drugs available for the prevention or treatment of fibrosis-related diseases. It has become evident that fibrosis is characterized by inflammation. In a large number of studies of various organs in mice and humans, pyroptosis has been found to play a significant role in fibrosis. Pyroptosis is a form of programmed cell death mediated by the N-terminal fragment of cysteinyl aspartate-specific proteinase (caspase)-1-cleaved gasdermin D (GSDMD, producing GSDMD-N) that gives rise to inflammation via the release of some proinflammatory cytokines, including IL-1β, IL-18 and HMGB1. These cytokines can initiate the activation of fibroblasts. Inflammasomes, an important factor upstream of GSDMD, can activate caspase-1 to trigger the maturation of IL-1β and IL-18. Moreover, the inhibition of inflammasomes, proinflammatory cytokines and GSDMD can prevent the progression of fibrosis. This review summarizes the growing evidence indicating that pyroptosis triggers fibrosis, and highlights potential novel targets for antifibrotic therapies.
Collapse
Affiliation(s)
- Zihao Song
- Department of Immunology, School of Medicine, Yangtze University, Jingzhou 434023, China;
| | - Quan Gong
- Department of Immunology, School of Medicine, Yangtze University, Jingzhou 434023, China;
- Correspondence: (Q.G.); (J.G.)
| | - Jiawei Guo
- Department of Pharmacology, School of Medicine, Yangtze University, Jingzhou 434023, China
- Correspondence: (Q.G.); (J.G.)
| |
Collapse
|
19
|
Gupta K, Pandey S, Bagang N, Mehra K, Singh G. Trimetazidine an emerging paradigm in renal therapeutics: Preclinical and clinical insights. Eur J Pharmacol 2021; 913:174624. [PMID: 34774496 DOI: 10.1016/j.ejphar.2021.174624] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2021] [Revised: 10/30/2021] [Accepted: 11/03/2021] [Indexed: 10/19/2022]
Abstract
Trimetazidine (TMZ) is a well-known anti-ischemic agent used for the treatment of angina pectoris. In the past decades, the efficacy of this drug has been tested in a wide range of kidney injuries, including drug-induced nephrotoxicity (DIN), radio-contrast agent-induced nephropathy, and surgically induced renal ischemic injury. TMZhas renoprotective effects by attenuating oxidative stress, inflammatory cytokine release, maintaining oxygen and energy balance. Moreover, TMZ administration prevented kidney graft rejection in the porcine model by suppressing the infiltration of mononuclear cells, preserving mitochondrial functions, and maintaining Ca+ homeostasis. In DIN and diabetic kidney diseases,TMZ treatment prevents renal injury by inactivating immune cells, attenuating renal fibrosis, inflammation, apoptosis, and histological abnormalities. Interestingly, the clinical therapeutic efficacy of TMZ has also been documented in pre-existing kidney disease patients undergoing contrast exposure for diagnostic intervention. However, the mechanistic insights into the TMZ mediated renoprotective effects in other forms of renal injuries, including type-2 diabetes, drug-induced nephrotoxicity, and hypertension-induced chronic kidney diseases, remain uninvestigated and incomplete. Moreover, the clinical utility of TMZ as a renoprotective agent in radio-contrast-induced nephrotoxicity needs to be tested in a large patient population. Nevertheless, the available pieces of evidence suggest that TMZ is a promising and emerging renal therapy for the treatment and management of kidney diseases of variable etiologies. This review discusses the various pre-clinical and clinical findings and provides mechanistic insights into the TMZ mediated beneficial effects in various kidney diseases.
Collapse
Affiliation(s)
- Kirti Gupta
- Department of Pharmacy, Maharishi Markandeshwar Deemed to be University, Mullana, Ambala (Haryana), India
| | - Sneha Pandey
- Department of Pharmacology, Indo-Soviet Friendship College of Pharmacy, Moga, Punjab, India
| | - Newly Bagang
- Department of Pharmacology, Indo-Soviet Friendship College of Pharmacy, Moga, Punjab, India
| | - Kamalpreet Mehra
- Department of Pharmacy, Maharishi Markandeshwar Deemed to be University, Mullana, Ambala (Haryana), India
| | | |
Collapse
|
20
|
Wang Y, Li Y, Xu Y. Pyroptosis in Kidney Disease. J Mol Biol 2021; 434:167290. [PMID: 34626644 DOI: 10.1016/j.jmb.2021.167290] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2021] [Revised: 09/24/2021] [Accepted: 09/29/2021] [Indexed: 01/06/2023]
Abstract
In the last several decades, apoptosis interference has been considered clinically irrelevant in the context of renal injury. Recent discovery of programmed necrotic cell death, including necroptosis, ferroptosis, and pyroptosis refreshed our understanding of the role of cell death in kidney disease. Pyroptosis is characterized by a lytic pro- inflammatory type of cell death resulting from gasdermin-induced membrane permeabilization via activation of inflammatory caspases and inflammasomes. The danger-associated molecular patterns (DAMPs), alarmins and pro-inflammatory cytokines are released from pyroptotic cells in an uncontrolled manner, which provoke inflammation, resulting in secondary organ or tissue injuries. The caspases and inflammasome activation-related proteins and pore-forming effector proteins known as GSDMD and GSDME have been implicated in a variety of acute and chronic microbial and non-microbial kidney diseases. Here, we review the recent advances in pathological mechanisms of pyroptosis in kidney disease and highlight the potential therapeutic strategies in future.
Collapse
Affiliation(s)
- Yujia Wang
- Department of Nephrology, Blood Purification Research Center, the First Affiliated Hospital, Fujian Medical University, Fuzhou 350005, China
| | - Yinshuang Li
- Department of Nephrology, Blood Purification Research Center, the First Affiliated Hospital, Fujian Medical University, Fuzhou 350005, China
| | - Yanfang Xu
- Department of Nephrology, Blood Purification Research Center, the First Affiliated Hospital, Fujian Medical University, Fuzhou 350005, China.
| |
Collapse
|
21
|
Ni W, Zhang Y, Yin Z. The protective mechanism of Klotho gene-modified bone marrow mesenchymal stem cells on acute kidney injury induced by rhabdomyolysis. Regen Ther 2021; 18:255-267. [PMID: 34466631 PMCID: PMC8367782 DOI: 10.1016/j.reth.2021.07.003] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2021] [Revised: 06/21/2021] [Accepted: 07/13/2021] [Indexed: 01/09/2023] Open
Abstract
Background Studies have shown that the Klotho gene has tremendous potential for future therapeutic purposes in both acute and chronic kidney diseases (CKD). This study aimed to investigate the possible protective mechanisms of the Klotho gene against acute kidney injury (AKI) induced by rhabdomyolysis (RM). Methods In this study, bone marrow mesenchymal stem cells (BMSCs) were transfected with recombinant adenoviruses expressing the Klotho gene (BMSCs-Klotho) and by those expressing empty vector (BMSCs-EV). After successful transfection, we tested the proliferation, secretion and migration abilities of the BMSCs-Klotho compared with those of the BMSCs-EV and BMSCs. Then, 30 male C57BL/6 mice were examined, with 6 mice randomly assigned to the control group (PBS injected into the tail vein, CON) or one of the four treatment groups treated with either BMSCs-Klotho (AKI+BMSCs-Klotho), BMSCs-EV (AKI+BMSCs-EV), BMSCs (AKI+BMSCs) or PBS (AKI+PBS) after induction of RM. Seventy-two h after treatment, serum creatinine (SCr) and blood urea nitrogen (BUN) levels were obtained to assess renal function, and renal tissue was obtained to measure kidney tissue damage. Additionally, kidney protective mechanism-related indexes, such as EPO, IGF-1, KIM-1 and HIF-1, were analysed using Western blot analysis and immunohistochemistry. Results The results obtained showed that the proliferation, secretory and migration abilities of the BMSCs were significantly increased after transfection with the Klotho gene. Treatment with BMSCs-Klotho, BMSCs-EV or BMSCs improved renal function compared to treatment with PBS. However, the improvement observed in renal function in the BMSCs-Klotho group was better than that of the other groups. Histological analysis demonstrated that tissue damage was significantly decreased in the mice in the AKI+BMSCs-Klotho, AKI+BMSCs-EV or AKI+BMSCs groups compared to that in the mice in the AKI+PBS group. However, the best recovery was observed in the mice treated with BMSCs-Klotho concomitantly. Furthermore, the expression of protective factors erythropoietin (EPO) and insulin-like growth factor 1 (IGF-1) increased obviously, and the injury biomarkers kidney injury molecule 1 (KIM-1) and hypoxia inducible factor 1 (HIF-1) decreased notably in the group of BMSCs-Klotho, BMSCs-EV and BMSCs. Additionally, the levels of the aforementioned protein indicators in the AKI+BMSCs-Klotho group were not different from those in the CON group. Conclusion Klotho overexpression exerted positive effects on BMSCs and markedly promoted recovery from RM-induced AKI. These findings suggest that the overexpression of the Klotho gene might be a good candidate for further therapy for AKI in clinical trials.
Collapse
Affiliation(s)
- WenHui Ni
- Department of Renal Medicine, First People's Hospital of Zhangjiagang City, China
| | - Ying Zhang
- Department of Renal Medicine, Xuzhou Medical University Affiliated Hospital, China
| | - Zhongcheng Yin
- Department of Renal Medicine, Xuzhou Medical University Affiliated Hospital, China
| |
Collapse
|
22
|
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
|
23
|
Nicholas SB. Novel Anti-inflammatory and Anti-fibrotic Agents for Diabetic Kidney Disease-From Bench to Bedside. Adv Chronic Kidney Dis 2021; 28:378-390. [PMID: 34922694 DOI: 10.1053/j.ackd.2021.09.010] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2020] [Revised: 08/30/2021] [Accepted: 09/17/2021] [Indexed: 02/08/2023]
Abstract
Chronic low-grade inflammation, now coined by the new paradigm as "metaflammation" or "metainflammation", has been linked to chronic kidney disease and its progression. In diabetes, altered metabolism denotes factors associated with the metabolic syndrome and hyperglycemia, among others. The interplay among hyperglycemia, oxidative stress, and inflammation in the pathogenesis of diabetic kidney disease (DKD) has been broadly explored. Identification of mediators of inflammatory processes involving macrophage infiltration, production of inflammasomes, release of cytokines, and activation of pertinent signaling pathways including mitogen-activated protein kinase, Jun N-terminal kinase, Janus kinase (JAK)-signal transducer and activator of transcription (STAT) pathway (JAK/STAT), and apoptosis signal-regulating kinase 1 signaling mechanisms have enabled the development of therapeutic agents for DKD. This review describes the evidence supporting the contribution of the inflammatory response and fibrotic changes and focuses on selected, novel, promising drugs as well as repurposed drugs that have made it to phase 2, 3, or 4 of clinical trials in adults with type 2 diabetes mellitus and their potential to become an important part of our armamentarium to improve the management of DKD. Importantly, drugs that solely target inflammatory processes may be insufficient to fully optimize care of patients with DKD because of the complex nature of the disease.
Collapse
|
24
|
Marczak L, Idkowiak J, Tracz J, Stobiecki M, Perek B, Kostka-Jeziorny K, Tykarski A, Wanic-Kossowska M, Borowski M, Osuch M, Formanowicz D, Luczak M. Mass Spectrometry-Based Lipidomics Reveals Differential Changes in the Accumulated Lipid Classes in Chronic Kidney Disease. Metabolites 2021; 11:275. [PMID: 33925471 PMCID: PMC8146808 DOI: 10.3390/metabo11050275] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2021] [Accepted: 04/23/2021] [Indexed: 11/16/2022] Open
Abstract
Chronic kidney disease (CKD) is characterized by the progressive loss of functional nephrons. Although cardiovascular disease (CVD) complications and atherosclerosis are the leading causes of morbidity and mortality in CKD, the mechanism by which the progression of CVD accelerates remains unclear. To reveal the molecular mechanisms associated with atherosclerosis linked to CKD, we applied a shotgun lipidomics approach fortified with standard laboratory analytical methods and gas chromatography-mass spectrometry technique on selected lipid components and precursors to analyze the plasma lipidome in CKD and classical CVD patients. The MS-based lipidome profiling revealed the upregulation of triacylglycerols in CKD and downregulation of cholesterol/cholesteryl esters, sphingomyelins, phosphatidylcholines, phosphatidylethanolamines and ceramides as compared to CVD group and controls. We have further observed a decreased abundance of seven fatty acids in CKD with strong inter-correlation. In contrast, the level of glycerol was elevated in CKD in comparison to all analyzed groups. Our results revealed the putative existence of a functional causative link-the low cholesterol level correlated with lower estimated glomerular filtration rate and kidney dysfunction that supports the postulated "reverse epidemiology" theory and suggest that the lipidomic background of atherosclerosis-related to CKD is unique and might be associated with other cellular factors, i.e., inflammation.
Collapse
Affiliation(s)
- Lukasz Marczak
- Department of Natural Products Biochemistry, Institute of Bioorganic Chemistry Polish Academy of Sciences, 61-704 Poznan, Poland; (J.I.); (M.S.)
| | - Jakub Idkowiak
- Department of Natural Products Biochemistry, Institute of Bioorganic Chemistry Polish Academy of Sciences, 61-704 Poznan, Poland; (J.I.); (M.S.)
- Department of Analytical Chemistry, Faculty of Chemical Technology, University of Pardubice, 532 10 Pardubice, Czech Republic
| | - Joanna Tracz
- Department of Biomedical Proteomics, Institute of Bioorganic Chemistry Polish Academy of Sciences, 61-704 Poznan, Poland;
| | - Maciej Stobiecki
- Department of Natural Products Biochemistry, Institute of Bioorganic Chemistry Polish Academy of Sciences, 61-704 Poznan, Poland; (J.I.); (M.S.)
| | - Bartłomiej Perek
- Department of Cardiac Surgery and Transplantology, Poznan University of Medical Sciences, 61-001 Poznan, Poland;
| | - Katarzyna Kostka-Jeziorny
- Department of Hypertension, Angiology and Internal Disease, Poznan University of Medical Sciences, 61-001 Poznan, Poland; (K.K.-J.); (A.T.)
| | - Andrzej Tykarski
- Department of Hypertension, Angiology and Internal Disease, Poznan University of Medical Sciences, 61-001 Poznan, Poland; (K.K.-J.); (A.T.)
| | - Maria Wanic-Kossowska
- Department of Nephrology, Transplantology and Internal Medicine, Poznan University of Medical Sciences, 60-355 Poznan, Poland;
| | - Marcin Borowski
- Institute of Computing Science, Poznan University of Technology, 60-965 Poznan, Poland;
| | - Marcin Osuch
- Department of Molecular and Systems Biology, Institute of Bioorganic Chemistry Polish Academy of Sciences, 61-704 Poznan, Poland;
| | - Dorota Formanowicz
- Chair and Department of Medical Chemistry and Laboratory Medicine, Poznan University of Medical Sciences, 60-806 Poznan, Poland;
| | - Magdalena Luczak
- Department of Biomedical Proteomics, Institute of Bioorganic Chemistry Polish Academy of Sciences, 61-704 Poznan, Poland;
| |
Collapse
|
25
|
Moreno KGT, Gasparotto Junior A, Dos Santos AC, Palozi RAC, Guarnier LP, Marques AAM, Romão PVM, Lorençone BR, Cassemiro NS, Silva DB, Tirloni CAS, de Barros ME. Nephroprotective and antilithiatic activities of Costus spicatus (Jacq.) Sw.: Ethnopharmacological investigation of a species from the Dourados region, Mato Grosso do Sul State, Brazil. JOURNAL OF ETHNOPHARMACOLOGY 2021; 266:113409. [PMID: 32979411 DOI: 10.1016/j.jep.2020.113409] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/08/2020] [Revised: 09/09/2020] [Accepted: 09/18/2020] [Indexed: 06/11/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Costus spicatus (Jacq.) Sw., also known as "cana-do-brejo," is a species that is widely used in Brazilian traditional medicine for the treatment of kidney diseases. However, no studies have evaluated its nephroprotective and antilithiatic effects. AIM To investigate nephroprotective and antilithiatic effects of C. spicatus in a preclinical model of acute kidney injury (AKI) and in vitro nephrolithiasis. MATERIALS AND METHODS C. spicatus leaves were collected directly from the natural environment in the Dourados region, Mato Grosso do Sul State, Brazil. The ethanol-soluble fraction of C. spicatus (ESCS) was obtained by infusion. Phytochemical characterization was performed by liquid chromatography coupled to diode array detector and mass spectrometer (LC-DAD-MS). We assessed whether ESCS has acute or prolonged diuretic activity. The nephroprotective effects of ESCS were evaluated in a model of AKI that was induced by glycerol (10 ml/kg, intramuscularly) in Wistar rats. Different doses of ESCS (30, 100, and 300 mg/kg) were administered orally for 5 days before the induction of AKI. Urinary parameters were measured on days 1, 3, 5, and 7. Twenty-four hours after the last urine collection, blood samples were obtained for the biochemical analysis. Blood pressure levels, renal vascular reactivity, renal tissue redox status, and histopathological changes were measured. Antilithiatic effects were evaluated by in vitro crystallization. Calcium oxalate precipitation was induced by sodium oxalate in urine samples with ESCS at 0.05, 0.5, and 5 mg/ml. RESULTS From LC-DAD-MS analyses, flavonoids, saponins and other phenolic compounds were determined in the composition of ESCS. Significant reductions of the excretion of urinary total protein, creatinine, sodium, and potassium were observed in the AKI group, with significant histopathological damage (swelling, vacuolization, necrosis, and inflammatory infiltration) in the proximal convoluted tubule. Treatment with ESCS exerted a significant nephroprotective effect by increasing the urinary excretion of total protein, urea, creatinine, sodium, potassium, calcium, and chloride. All of the groups that were treated with ESCS exhibited a reduction of histopathological lesions and significant modulation of the tissue redox state. We also observed a concentration-dependent effect of ESCS on the crystallization of urinary crystals, with reductions of the size and proportion of monohydrated crystals. CONCLUSION The data suggest that C. spicatus has nephroprotective and antilithiatic effects, suggesting possible effectiveness in its traditional use.
Collapse
Affiliation(s)
- Karyne Garcia Tafarelo Moreno
- Laboratório de Urinálise, Faculdade de Ciências da Saúde, Universidade Federal da Grande Dourados, Dourados, MS, Brazil
| | - Arquimedes Gasparotto Junior
- Laboratório de Farmacologia Cardiovascular- LaFaC, Faculdade de Ciências da Saúde, Universidade Federal da Grande Dourados, Dourados, MS, Brazil.
| | - Ariany Carvalho Dos Santos
- Laboratório de Histopatologia, Faculdade de Ciências da Saúde, Universidade Federal da Grande Dourados, Dourados, MS, Brazil
| | - Rhanany Alan Calloi Palozi
- Laboratório de Farmacologia Cardiovascular- LaFaC, Faculdade de Ciências da Saúde, Universidade Federal da Grande Dourados, Dourados, MS, Brazil
| | - Lucas Pires Guarnier
- Laboratório de Farmacologia Cardiovascular- LaFaC, Faculdade de Ciências da Saúde, Universidade Federal da Grande Dourados, Dourados, MS, Brazil
| | - Aline Aparecida Macedo Marques
- Laboratório de Farmacologia Cardiovascular- LaFaC, Faculdade de Ciências da Saúde, Universidade Federal da Grande Dourados, Dourados, MS, Brazil
| | - Paulo Vitor Moreira Romão
- Laboratório de Farmacologia Cardiovascular- LaFaC, Faculdade de Ciências da Saúde, Universidade Federal da Grande Dourados, Dourados, MS, Brazil
| | - Bethânia Rosa Lorençone
- Laboratório de Farmacologia Cardiovascular- LaFaC, Faculdade de Ciências da Saúde, Universidade Federal da Grande Dourados, Dourados, MS, Brazil
| | - Nadla Soares Cassemiro
- Laboratório de Produtos Naturais e Espectrometria de Massas (LaPNEM), Faculdade de Ciências Farmacêuticas, Alimentos e Nutrição (FACFAN), Universidade Federal Do Mato Grosso Do Sul, Campo Grande, MS, Brazil
| | - Denise Brentan Silva
- Laboratório de Produtos Naturais e Espectrometria de Massas (LaPNEM), Faculdade de Ciências Farmacêuticas, Alimentos e Nutrição (FACFAN), Universidade Federal Do Mato Grosso Do Sul, Campo Grande, MS, Brazil
| | - Cleide Adriane Signor Tirloni
- Laboratório de Farmacologia Cardiovascular- LaFaC, Faculdade de Ciências da Saúde, Universidade Federal da Grande Dourados, Dourados, MS, Brazil
| | - Márcio Eduardo de Barros
- Laboratório de Urinálise, Faculdade de Ciências da Saúde, Universidade Federal da Grande Dourados, Dourados, MS, Brazil
| |
Collapse
|
26
|
Harmine, a natural β-carboline alkaloid, ameliorates apoptosis by decreasing the expression of caspase-3 in the kidney of diabetic male Wistar rats. GENE REPORTS 2020. [DOI: 10.1016/j.genrep.2020.100863] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
27
|
Song SJ, Kim SM, Lee SH, Moon JY, Hwang HS, Kim JS, Park SH, Jeong KH, Kim YG. Rhabdomyolysis-Induced AKI Was Ameliorated in NLRP3 KO Mice via Alleviation of Mitochondrial Lipid Peroxidation in Renal Tubular Cells. Int J Mol Sci 2020; 21:ijms21228564. [PMID: 33202867 PMCID: PMC7696646 DOI: 10.3390/ijms21228564] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2020] [Revised: 11/09/2020] [Accepted: 11/10/2020] [Indexed: 12/13/2022] Open
Abstract
Introduction: A recent study showed that early renal tubular injury is ameliorated in Nod-like receptor pyrin domain-containing protein 3 (NLRP3) KO mice with rhabdomyolysis-induced acute kidney injury (RIAKI). However, the precise mechanism has not been determined. Therefore, we investigated the role of NLRP3 in renal tubular cells in RIAKI. Methods: Glycerol-mediated RIAKI was induced in NLRP3 KO and wild-type (WT) mice. The mice were euthanized 24 h after glycerol injection, and both kidneys and plasma were collected. HKC-8 cells were treated with ferrous myoglobin to mimic a rhabdomyolytic environment. Results: Glycerol injection led to increase serum creatinine, aspartate aminotransferase (AST), and renal kidney injury molecule-1 (KIM-1) level; renal tubular necrosis; and apoptosis. Renal injury was attenuated in NLRP3 KO mice, while muscle damage and renal neutrophil recruitment did not differ between NLRP3 KO mice and WT mice. Following glycerin injection, increases in cleaved caspase-3, poly (ADP-ribose) polymerase (PARP), and a decrease in the glutathione peroxidase 4 (GPX-4) level were observed in the kidneys of mice with RIAKI, and these changes were alleviated in the kidneys of NLRP3 KO mice. NLRP3 was upregulated, and cell viability was suppressed in HKC-8 cells treated with ferrous myoglobin. Myoglobin-induced apoptosis and lipid peroxidation were significantly decreased in siNLRP3-treated HKC-8 cells compared to ferrous myoglobin-treated HKC-8 cells. Myoglobin reduced the mitochondrial membrane potential and increased mitochondrial fission and reactive oxygen species (ROS) and lipid peroxidation levels, which were restored to normal levels in NLRP3-depleted HKC-8 cells. Conclusions: NLRP3 depletion ameliorated renal tubular injury in a murine glycerol-induced acute kidney injury (AKI) model. A lack of NLRP3 improved tubular cell viability via attenuation of myoglobin-induced mitochondrial injury and lipid peroxidation, which might be the critical factor in protecting the kidney.
Collapse
|
28
|
Boudhabhay I, Poillerat V, Grunenwald A, Torset C, Leon J, Daugan MV, Lucibello F, El Karoui K, Ydee A, Chauvet S, Girardie P, Sacks S, Farrar CA, Garred P, Berthaud R, Le Quintrec M, Rabant M, de Lonlay P, Rambaud C, Gnemmi V, Fremeaux-Bacchi V, Frimat M, Roumenina LT. Complement activation is a crucial driver of acute kidney injury in rhabdomyolysis. Kidney Int 2020; 99:581-597. [PMID: 33137339 DOI: 10.1016/j.kint.2020.09.033] [Citation(s) in RCA: 56] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2020] [Revised: 09/01/2020] [Accepted: 09/25/2020] [Indexed: 12/17/2022]
Abstract
Rhabdomyolysis is a life-threatening condition caused by skeletal muscle damage with acute kidney injury being the main complication dramatically worsening the prognosis. Specific treatment for rhabdomyolysis-induced acute kidney injury is lacking and the mechanisms of the injury are unclear. To clarify this, we studied intra-kidney complement activation (C3d and C5b-9 deposits) in tubules and vessels of patients and mice with rhabdomyolysis-induced acute kidney injury. The lectin complement pathway was found to be activated in the kidney, likely via an abnormal pattern of Fut2-dependent cell fucosylation, recognized by the pattern recognition molecule collectin-11 and this proceeded in a C4-independent, bypass manner. Concomitantly, myoglobin-derived heme activated the alternative pathway. Complement deposition and acute kidney injury were attenuated by pre-treatment with the heme scavenger hemopexin. This indicates that complement was activated in a unique double-trigger mechanism, via the alternative and lectin pathways. The direct pathological role of complement was demonstrated by the preservation of kidney function in C3 knockout mice after the induction of rhabdomyolysis. The transcriptomic signature for rhabdomyolysis-induced acute kidney injury included a strong inflammatory and apoptotic component, which were C3/complement-dependent, as they were normalized in C3 knockout mice. The intra-kidney macrophage population expressed a complement-sensitive phenotype, overexpressing CD11b and C5aR1. Thus, our results demonstrate a direct pathological role of heme and complement in rhabdomyolysis-induced acute kidney injury. Hence, heme scavenging and complement inhibition represent promising therapeutic strategies.
Collapse
Affiliation(s)
- Idris Boudhabhay
- Centre de Recherche des Cordeliers, Institut National de la Santé et de la Recherche Médicale, Sorbonne Université, Université de Paris, Paris, France
| | - Victoria Poillerat
- Centre de Recherche des Cordeliers, Institut National de la Santé et de la Recherche Médicale, Sorbonne Université, Université de Paris, Paris, France
| | - Anne Grunenwald
- Centre de Recherche des Cordeliers, Institut National de la Santé et de la Recherche Médicale, Sorbonne Université, Université de Paris, Paris, France
| | - Carine Torset
- Centre de Recherche des Cordeliers, Institut National de la Santé et de la Recherche Médicale, Sorbonne Université, Université de Paris, Paris, France
| | - Juliette Leon
- Centre de Recherche des Cordeliers, Institut National de la Santé et de la Recherche Médicale, Sorbonne Université, Université de Paris, Paris, France
| | - Marie V Daugan
- Centre de Recherche des Cordeliers, Institut National de la Santé et de la Recherche Médicale, Sorbonne Université, Université de Paris, Paris, France
| | - Francesca Lucibello
- Institut National de la Santé et de la Recherche Médicale U932, Paris Sciences et Lettres University, Institut Curie, Paris, France
| | - Khalil El Karoui
- Service de Néphrologie et Transplantation Rénale, Hôpital Henri Mondor, Assistance Publique - Hôpitaux de Paris, Créteil, Paris, France
| | - Amandine Ydee
- Pathology Department, Lille University Hospital (Centre Hospitalier Universitaire), Pathology Institute, Institut National de la Santé et de la Recherche Médicale UMR-S1172 Lille, JPARC-Jean-Pierre Aubert Research Center, Team "Mucins, Epithelial Differentiation and Carcinogenesis," Lille University, Centre Hospitalier Universitaire Lille, Lille, France
| | - Sophie Chauvet
- Centre de Recherche des Cordeliers, Institut National de la Santé et de la Recherche Médicale, Sorbonne Université, Université de Paris, Paris, France; Department of Nephrology, Georges Pompidou European Hospital, Paris, France
| | - Patrick Girardie
- Intensive Care Department, Université de Lille, Centre Hospitalier Universitaire Lille, Lille, France
| | - Steven Sacks
- Medical Research Council Centre for Transplantation, Peter Gorer Department of Immunobiology, School of Immunology and Microbial Sciences, King's College London, London, UK
| | - Conrad A Farrar
- Medical Research Council Centre for Transplantation, Peter Gorer Department of Immunobiology, School of Immunology and Microbial Sciences, King's College London, London, UK
| | - Peter Garred
- Laboratory of Molecular Medicine, Department of Clinical Immunology, University of Copenhagen, Copenhagen, Denmark
| | - Romain Berthaud
- Department of Pediatric Nephrology, Necker Hospital, Assistance Publique - Hôpitaux de Paris, Paris, France
| | - Moglie Le Quintrec
- Department of Nephrology and Kidney Transplantation, Centre Hospitalier Universitaire de Montpellier, Montpellier, France
| | - Marion Rabant
- Department of Pathology, Necker Hospital, Assistance Publique - Hôpitaux de Paris (AP-HP), Paris, France
| | - Pascale de Lonlay
- Reference Centre for Metabolic Diseases, Necker-Enfants Malades Hospital, Imagine Institute, Université Paris-Descartes, Assistance Publique - Hôpitaux de Paris, Paris, France
| | - Caroline Rambaud
- Service Médecine Légale, Hôpital Raymond Poincaré, Assistance Publique - Hôpitaux de Paris, Garches, France
| | - Viviane Gnemmi
- Pathology Department, Lille University Hospital (Centre Hospitalier Universitaire), Pathology Institute, Institut National de la Santé et de la Recherche Médicale UMR-S1172 Lille, JPARC-Jean-Pierre Aubert Research Center, Team "Mucins, Epithelial Differentiation and Carcinogenesis," Lille University, Centre Hospitalier Universitaire Lille, Lille, France
| | - Veronique Fremeaux-Bacchi
- Centre de Recherche des Cordeliers, Institut National de la Santé et de la Recherche Médicale, Sorbonne Université, Université de Paris, Paris, France; Laboratory of Immunology, Hôpital Européen Georges Pompidou, Assistance Publique - Hôpitaux de Paris, Paris, France
| | - Marie Frimat
- University of Lille, U995-LIRIC-Lille Inflammation Research International Center, Lille, France; Department of Nephrology, Lille University Hospital, Centre Hospitalier Universitaire, Lille, France
| | - Lubka T Roumenina
- Centre de Recherche des Cordeliers, Institut National de la Santé et de la Recherche Médicale, Sorbonne Université, Université de Paris, Paris, France.
| |
Collapse
|
29
|
Alyaseer AAA, de Lima MHS, Braga TT. The Role of NLRP3 Inflammasome Activation in the Epithelial to Mesenchymal Transition Process During the Fibrosis. Front Immunol 2020; 11:883. [PMID: 32508821 PMCID: PMC7251178 DOI: 10.3389/fimmu.2020.00883] [Citation(s) in RCA: 83] [Impact Index Per Article: 16.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2020] [Accepted: 04/16/2020] [Indexed: 02/06/2023] Open
Abstract
Fibrosis is considered a complex form of tissue damage commonly present in the end stage of many diseases. It is also related to a high percentage of death, whose predominant characteristics are an excessive and abnormal deposition of fibroblasts and myofibroblasts -derived extracellular matrix (ECM) components. Epithelial-to-mesenchymal transition (EMT), a process in which epithelial cells gradually change to mesenchymal ones, is a major contributor in the pathogenesis of fibrosis. The key mediator of EMT is a multifunctional cytokine called transforming growth factor-β (TGF-β) that acts as the main inducer of the ECM assembly and remodeling through the phosphorylation of Smad2/3, which ultimately forms a complex with Smad4 and translocates into the nucleus. On the other hand, the bone morphogenic protein-7 (BMP-7), a member of the TGF family, reverses EMT by directly counteracting TGF-β induced Smad-dependent cell signaling. NLRP3 (NACHT, LRR, and PYD domains-containing protein 3), in turn, acts as cytosolic sensors of microbial and self-derived molecules and forms an immune complex called inflammasome in the context of inflammatory commitments. NLRP3 inflammasome assembly is triggered by extracellular ATP, reactive oxygen species (ROS), potassium efflux, calcium misbalance, and lysosome disruption. Due to its involvement in multiple diseases, NLRP3 has become one of the most studied pattern-recognition receptors (PRRs). Nevertheless, the role of NLRP3 in fibrosis development has not been completely elucidated. In this review, we described the relation of the previously mentioned fibrosis pathway with the NLRP3 inflammasome complex formation, especially EMT-related pathways. For now, it is suggested that the EMT happens independently from the oligomerization of the whole inflammasome complex, requiring just the presence of the NLRP3 receptor and the ASC protein to trigger the EMT events, and we will present different pieces of research that give controversial point of views.
Collapse
Affiliation(s)
| | | | - Tarcio Teodoro Braga
- Department of Pathology, Federal University of Parana, Curitiba, Brazil.,Instituto Carlos Chagas, Fiocruz-Parana, Curitiba, Brazil
| |
Collapse
|
30
|
AlBasher G, Alfarraj S, Alarifi S, Alkhtani S, Almeer R, Alsultan N, Alharthi M, Alotibi N, Al-Dbass A, Abdel Moneim AE. Nephroprotective Role of Selenium Nanoparticles Against Glycerol-Induced Acute Kidney Injury in Rats. Biol Trace Elem Res 2020; 194:444-454. [PMID: 31264127 DOI: 10.1007/s12011-019-01793-5] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/14/2019] [Accepted: 06/19/2019] [Indexed: 02/08/2023]
Abstract
Acute kidney injury (AKI) is a clinical syndrome associated with the incidence of rhabdomyolysis (RM). The current study was carried out to evaluate whether selenium nanoparticles (SeNPs) can protect against the glycerol-induced AKI model. Rats were distributed into four equal groups (n = 7): the control group (G1), SeNPs group (G2), AKI group (G3), and SeNPs+AKI group (G4). Rats in G1 were intramuscularly injected with physiological saline (0.9% NaCl). Rats in G2 were gavaged with SeNPs (0.1 mg/kg) for 14 days. Rats in G3 were intramuscularly injected with 50% glycerol (10 ml/kg). Rats in G4 were administered with SeNPs for 14 days and then injected with glycerol, as in G3. Glycerol-injected rats showed a significant increase in the kidney relative weight, as well as in the serum urea, creatinine, Kim-1, and renal malondialdehyde, nitric oxide, TNF-α, IL-1β, cytochrome c, Bax, and caspase-3 levels. In addition, a significant decrease in glutathione, glutathione peroxidase, glutathione reductase, superoxide dismutase, and catalase was recorded in the renal tissue. Selenium nanoparticles reduced the biochemical, molecular, and histological changes produced by glycerol. Overall, our results suggest that selenium nanoparticles could be used to protect against AKI development via antioxidant, anti-inflammatory, and anti-apoptotic activities.
Collapse
Affiliation(s)
- Gadah AlBasher
- Department of Zoology, College of Science, King Saud University, Riyadh, Saudi Arabia.
| | - Saleh Alfarraj
- Department of Zoology, College of Science, King Saud University, Riyadh, Saudi Arabia
| | - Saud Alarifi
- Department of Zoology, College of Science, King Saud University, Riyadh, Saudi Arabia
| | - Saad Alkhtani
- Department of Zoology, College of Science, King Saud University, Riyadh, Saudi Arabia
| | - Rafa Almeer
- Department of Zoology, College of Science, King Saud University, Riyadh, Saudi Arabia
| | - Nouf Alsultan
- Department of Zoology, College of Science, King Saud University, Riyadh, Saudi Arabia
| | - Mada Alharthi
- Department of Applied Medical Science, Collage of Applied Medical Science, Shaqra University, Riyadh, Saudi Arabia
| | - Nouf Alotibi
- Department of Chemistry, College of Sciences, King Saud University, Riyadh, Saudi Arabia
| | - Abeer Al-Dbass
- Department of Biochemistry, College of Science, King Saud University, Riyadh, Saudi Arabia
| | - Ahmed E Abdel Moneim
- Department of Zoology and Entomology, Faculty of Science, Helwan University, Cairo, Egypt
| |
Collapse
|
31
|
Zheng Y, Guan H, Zhou X, Xu Y, Fu C, Xiao J, Ye Z. The association of renal tubular inflammatory and injury markers with uric acid excretion in chronic kidney disease patients. Int Urol Nephrol 2020; 52:923-932. [PMID: 32232720 DOI: 10.1007/s11255-020-02447-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2019] [Accepted: 03/16/2020] [Indexed: 01/05/2023]
Abstract
AIM To investigate the correlation of renal tubular inflammatory and injury markers with renal uric acid excretion in chronic kidney disease (CKD) patients. METHODS Seventy-three patients with CKD were enrolled. Fasting blood and morning urine sample were collected for routine laboratory measurements. At the same time, 24 h of urine was collected for urine biochemistry analyses, and 10 ml was extracted from the 24-h urine sample to further detect renal tubular inflammatory and injury markers, including interleukin-18 (IL-18), interleukin 1β (IL-1β), neutrophil gelatinase-associated lipocalin (NGAL) and kidney injury molecule-1 (KIM-1). The patients were divided into three tertile groups according to their 24-h urinary uric acid (24-h UUA) levels (UUA1: 24-h UUA ≤ 393.12 mg; UUA2: 393.12 < 24-h UUA ≤ 515.76 mg; UUA3: 24-h UUA > 515.76 mg). The general clinical and biochemical indexes were compared. Multivariable linear regression models were used to test the association of IL-18/Urinary creatinine concentration (IL-18/CR), IL-1β/CR, NGAL/CR and KIM-1/CR with renal uric acid excretion indicators. RESULTS All of tested renal tubular inflammation- and injury-related urinary markers were negatively associated with 24-h UUA and UEUA, and the negative correlation still persisted after adjusting for multiple influencing factors including urinary protein and eGFR. Further group analyses showed that these makers were significantly higher in the UUA1 than in the UUA3 group. CONCLUSIONS Our findings suggest that markers of urinary interstitial inflammation and injury in CKD patients are significantly correlated with 24-h UUA and Urinary excretion of uric acid (UEUA), and those with high 24-h UUA have lower levels of these markers. Renal uric acid excretion may also reflect the inflammation and injury of renal tubules under certain conditions.
Collapse
Affiliation(s)
- Yuqi Zheng
- Department of Nephrology, Huadong Hospital Affiliated With Fudan University, No. 221 West Yan'an Road, Shanghai, 200040, P.R. China.,Shanghai Key Laboratory of Clinical Geriatric Medicine, No. 221 West Yan'an Road, Shanghai, 200040, P.R. China
| | - Haochen Guan
- Department of Nephrology, Huadong Hospital Affiliated With Fudan University, No. 221 West Yan'an Road, Shanghai, 200040, P.R. China.,Shanghai Key Laboratory of Clinical Geriatric Medicine, No. 221 West Yan'an Road, Shanghai, 200040, P.R. China
| | - Xun Zhou
- Department of Nephrology, Huadong Hospital Affiliated With Fudan University, No. 221 West Yan'an Road, Shanghai, 200040, P.R. China.,Shanghai Key Laboratory of Clinical Geriatric Medicine, No. 221 West Yan'an Road, Shanghai, 200040, P.R. China
| | - Ying Xu
- Department of Nephrology, Huadong Hospital Affiliated With Fudan University, No. 221 West Yan'an Road, Shanghai, 200040, P.R. China.,Shanghai Key Laboratory of Clinical Geriatric Medicine, No. 221 West Yan'an Road, Shanghai, 200040, P.R. China
| | - Chensheng Fu
- Department of Nephrology, Huadong Hospital Affiliated With Fudan University, No. 221 West Yan'an Road, Shanghai, 200040, P.R. China.,Shanghai Key Laboratory of Clinical Geriatric Medicine, No. 221 West Yan'an Road, Shanghai, 200040, P.R. China
| | - Jing Xiao
- Department of Nephrology, Huadong Hospital Affiliated With Fudan University, No. 221 West Yan'an Road, Shanghai, 200040, P.R. China. .,Shanghai Key Laboratory of Clinical Geriatric Medicine, No. 221 West Yan'an Road, Shanghai, 200040, P.R. China.
| | - Zhibin Ye
- Department of Nephrology, Huadong Hospital Affiliated With Fudan University, No. 221 West Yan'an Road, Shanghai, 200040, P.R. China. .,Shanghai Key Laboratory of Clinical Geriatric Medicine, No. 221 West Yan'an Road, Shanghai, 200040, P.R. China.
| |
Collapse
|
32
|
Noh MR, Jang HS, Kim J, Padanilam BJ. Renal Sympathetic Nerve-Derived Signaling in Acute and Chronic kidney Diseases. Int J Mol Sci 2020; 21:ijms21051647. [PMID: 32121260 PMCID: PMC7084190 DOI: 10.3390/ijms21051647] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2020] [Revised: 02/20/2020] [Accepted: 02/23/2020] [Indexed: 12/11/2022] Open
Abstract
The kidney is innervated by afferent sensory and efferent sympathetic nerve fibers. Norepinephrine (NE) is the primary neurotransmitter for post-ganglionic sympathetic adrenergic nerves, and its signaling, regulated through adrenergic receptors (AR), modulates renal function and pathophysiology under disease conditions. Renal sympathetic overactivity and increased NE level are commonly seen in chronic kidney disease (CKD) and are critical factors in the progression of renal disease. Blockade of sympathetic nerve-derived signaling by renal denervation or AR blockade in clinical and experimental studies demonstrates that renal nerves and its downstream signaling contribute to progression of acute kidney injury (AKI) to CKD and fibrogenesis. This review summarizes our current knowledge of the role of renal sympathetic nerve and adrenergic receptors in AKI, AKI to CKD transition and CKDand provides new insights into the therapeutic potential of intervening in its signaling pathways.
Collapse
Affiliation(s)
- Mi Ra Noh
- Department of Cellular and Integrative Physiology, University of Nebraska Medical Center, Omaha, NE 68198-5850, USA; (M.R.N.); (H.-S.J.); (J.K.)
| | - Hee-Seong Jang
- Department of Cellular and Integrative Physiology, University of Nebraska Medical Center, Omaha, NE 68198-5850, USA; (M.R.N.); (H.-S.J.); (J.K.)
| | - Jinu Kim
- Department of Cellular and Integrative Physiology, University of Nebraska Medical Center, Omaha, NE 68198-5850, USA; (M.R.N.); (H.-S.J.); (J.K.)
- Department of Anatomy, Jeju National University School of Medicine, Jeju 63243, Korea
- Interdisciplinary Graduate Program in Advanced Convergence Technology & Science, Jeju National University, Jeju 63243, Korea
| | - Babu J. Padanilam
- Department of Cellular and Integrative Physiology, University of Nebraska Medical Center, Omaha, NE 68198-5850, USA; (M.R.N.); (H.-S.J.); (J.K.)
- Department of Internal Medicine, Section of Nephrology, University of Nebraska Medical Center, Omaha, NE 68198-5850, USA
- Correspondence:
| |
Collapse
|
33
|
Pengrattanachot N, Cherngwelling R, Jaikumkao K, Pongchaidecha A, Thongnak L, Swe MT, Chatsudthipong V, Lungkaphin A. Atorvastatin attenuates obese-induced kidney injury and impaired renal organic anion transporter 3 function through inhibition of oxidative stress and inflammation. Biochim Biophys Acta Mol Basis Dis 2020; 1866:165741. [PMID: 32101757 DOI: 10.1016/j.bbadis.2020.165741] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2019] [Revised: 01/17/2020] [Accepted: 02/18/2020] [Indexed: 12/15/2022]
Abstract
An excessive consumption of high-fat diet can lead to the alterations of glucose and lipid metabolism, impaired insulin signaling and increased ectopic lipid accumulation resulting in renal lipotoxicity and subsequent renal dysfunction. Atorvastatin is a lipid-lowering drug in clinical treatment. Several studies have reported that atorvastatin has several significant pleiotropic effects including anti-inflammatory, antioxidant, and anti-apoptotic effects. However, the effects of atorvastatin on metabolic disturbance and renal lipotoxicity in obesity are not fully understood. In this study, obesity in rat was developed by high-fat diet (HFD) feeding for 16 weeks. After that, the HFD-fed rats were received either a vehicle (HF), atorvastatin (HFA) or vildagliptin (HFVIL), by oral gavage for 4 weeks. We found that HF rats showed insulin resistance, visceral fat expansion and renal lipid accumulation. Impaired renal function and renal organic anion transporter 3 (Oat3) function and expression were also observed in HF rats. The marked increases in MDA level, renal injury and NF-κB, TGF-β, NOX-4, PKC-α expression were demonstrated in HF rats. Atorvastatin or vildagliptin treatment attenuated insulin resistance and renal lipid accumulation-induced lipotoxicity in HFA and HFVIL rats. Moreover, the proteins involved in renal inflammation, fibrosis, oxidative stress and apoptosis were attenuated leading to improved renal Oat3 function and renal function in the treated groups. Interestingly, atorvastatin showed higher efficacy than vildagliptin in improving insulin resistance, renal lipid accumulation and in exerting renoprotective effects in obesity-induced renal injury and impaired renal Oat3 function.
Collapse
Affiliation(s)
| | - Rada Cherngwelling
- Department of Physiology, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Krit Jaikumkao
- Department of Radiologic Technology, Faculty of Associated Medical Sciences, Chiang Mai University, Chiang Mai, Thailand
| | - Anchalee Pongchaidecha
- Department of Physiology, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Laongdao Thongnak
- Department of Physiology, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Myat Theingi Swe
- Department of Physiology, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand; Department of Physiology, University of Medicine, Yangon, Myanmar
| | - Varanuj Chatsudthipong
- Research Center of Transport Protein for Medical Innovation, Faculty of Science, Mahidol University, Bangkok, Thailand
| | - Anusorn Lungkaphin
- Department of Physiology, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand; Functional Food Research Center for Well-being, Chiang Mai University, Chiang Mai, Thailand.
| |
Collapse
|
34
|
Abstract
Inflammasomes are multiprotein innate immune complexes that regulate caspase-dependent inflammation and cell death. Pattern recognition receptors, such as nucleotide-binding oligomerization domain (NOD)-like receptors and absent in melanoma 2 (AIM2)-like receptors, sense danger signals or cellular events to activate canonical inflammasomes, resulting in caspase 1 activation, pyroptosis and the secretion of IL-1β and IL-18. Non-canonical inflammasomes can be activated by intracellular lipopolysaccharides, toxins and some cell signalling pathways. These inflammasomes regulate the activation of alternative caspases (caspase 4, caspase 5, caspase 11 and caspase 8) that lead to pyroptosis, apoptosis and the regulation of other cellular pathways. Many inflammasome-related genes and proteins have been implicated in animal models of kidney disease. In particular, the NLRP3 (NOD-, LRR- and pyrin domain-containing 3) inflammasome has been shown to contribute to a wide range of acute and chronic microbial and non-microbial kidney diseases via canonical and non-canonical mechanisms that regulate inflammation, pyroptosis, apoptosis and fibrosis. In patients with chronic kidney disease, immunomodulation therapies targeting IL-1β such as canakinumab have been shown to prevent cardiovascular events. Moreover, findings in experimental models of kidney disease suggest that small-molecule inhibitors targeting NLRP3 and other inflammasome components are promising therapeutic agents.
Collapse
Affiliation(s)
- Takanori Komada
- Division of Inflammation Research, Center for Molecular Medicine, Jichi Medical University, Shimotsuke, Tochigi, Japan
| | - Daniel A Muruve
- Department of Medicine, Snyder Institute for Chronic Diseases, University of Calgary, Calgary, Alberta, Canada.
| |
Collapse
|
35
|
Research Progress on the Role of Inflammasomes in Kidney Disease. Mediators Inflamm 2020; 2020:8032797. [PMID: 32410864 PMCID: PMC7204206 DOI: 10.1155/2020/8032797] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2019] [Accepted: 01/16/2020] [Indexed: 12/12/2022] Open
Abstract
Inflammasomes are multimeric complexes composed of cytoplasmic sensors, apoptosis-associated speck-like protein containing a caspase activation and recruitment domain (ASC or PYCARD), and procaspase-1 and play roles in regulating caspase-dependent inflammation and cell death. Inflammasomes are assembled by sensing pathogen-associated molecular patterns (PAMPs) and damage-associated molecular patterns (DAMPs) and initiate inflammatory responses by activating caspase-1. Activated caspase-1 promotes the release of the inflammatory cytokines interleukin-1β (IL-1β) and IL-18 and eventually induces pyroptosis. Inflammasomes are closely related to kidney diseases. In particular, the NLRP3 (NACHT, LRR, and PYD domain-containing protein 3) inflammasome has been shown to cause acute and chronic kidney diseases by regulating canonical and noncanonical mechanisms of inflammation. Small-molecule inhibitors that target NLRP3 and other components of the inflammasome are potential options for the treatment of kidney-related diseases such as diabetic nephropathy. This article will focus on the research progress on inflammasomes and the key pathogenic roles of inflammasomes in the development and progression of kidney diseases and explore the potential of this intracellular inflammation to further prevent or block the development of the kidney disease.
Collapse
|
36
|
Nasir N, Raji S, Mustafa F, Rizvi TA, Al Natour Z, Hilal-Alnaqbi A, Al Ahmad M. Electrical detection of blood cells in urine. Heliyon 2019; 6:e03102. [PMID: 31909269 PMCID: PMC6938827 DOI: 10.1016/j.heliyon.2019.e03102] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2019] [Revised: 11/21/2019] [Accepted: 12/18/2019] [Indexed: 12/12/2022] Open
Abstract
Available methods for detecting blood in the urine (hematuria) can be problematic since results can be influenced by many factors in patients and in the lab settings, resulting in false positive or false negative results. This necessitates the development of new, accurate and easy-access methods that save time and effort. This study demonstrates a label-free and accurate method for detecting the presence of red and white blood cells (RBCs and WBCs) in urine by measuring the changes in the dielectric properties of urine upon increasing concentrations of both cell types. The current method could detect changes in the electrical properties of fresh urine over a short time interval, making this method suitable for detecting changes that cannot be recognized by conventional methods. Correcting for these changes enabled the detection of a minimum cell concentration of 102 RBCs per ml which is not possible by conventional methods used in the labs except for the semi-quantitative method that can detect 50 RBCs per ml, but it is a lengthy and involved procedure, not suitable for high volume labs. This ability to detect very small amount of both types of cells makes the proposed technique an attractive tool for detecting hematuria, the presence of which is indicative of problems in the excretory system.
Collapse
Affiliation(s)
- Nida Nasir
- Department of Electrical Engineering, College of Engineering, United Arab Emirates University (UAEU), Al Ain, 15551, United Arab Emirates.,Zayed Center for Health Sciences, United Arab Emirates University, United Arab Emirates
| | - Shaima Raji
- Department of Electrical Engineering, College of Engineering, United Arab Emirates University (UAEU), Al Ain, 15551, United Arab Emirates.,Zayed Center for Health Sciences, United Arab Emirates University, United Arab Emirates
| | - Farah Mustafa
- Department of Biochemistry, College of Medicine & Health Sciences, United Arab Emirates University (UAEU), Al Ain, 15551, United Arab Emirates.,Zayed Center for Health Sciences, United Arab Emirates University, United Arab Emirates
| | - Tahir A Rizvi
- Department of Microbiology and Immunology, College of Medicine & Health Sciences, United Arab Emirates University (UAEU), Al Ain, 15551, United Arab Emirates.,Zayed Center for Health Sciences, United Arab Emirates University, United Arab Emirates
| | - Zeina Al Natour
- Department of Electrical Engineering, College of Engineering, United Arab Emirates University (UAEU), Al Ain, 15551, United Arab Emirates.,Zayed Center for Health Sciences, United Arab Emirates University, United Arab Emirates
| | - Ali Hilal-Alnaqbi
- Department of Mechanical Engineering, College of Engineering, United Arab Emirates University (UAEU), Al Ain, 15551, United Arab Emirates.,Abu Dhabi Polytechnic, Abu Dhabi, 1114999, United Arab Emirates.,Zayed Center for Health Sciences, United Arab Emirates University, United Arab Emirates
| | - Mahmoud Al Ahmad
- Department of Electrical Engineering, College of Engineering, United Arab Emirates University (UAEU), Al Ain, 15551, United Arab Emirates.,Zayed Center for Health Sciences, United Arab Emirates University, United Arab Emirates
| |
Collapse
|
37
|
Kim YG, Kim SM, Kim KP, Lee SH, Moon JY. The Role of Inflammasome-Dependent and Inflammasome-Independent NLRP3 in the Kidney. Cells 2019; 8:cells8111389. [PMID: 31694192 PMCID: PMC6912448 DOI: 10.3390/cells8111389] [Citation(s) in RCA: 107] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2019] [Revised: 10/23/2019] [Accepted: 10/30/2019] [Indexed: 01/27/2023] Open
Abstract
Cytoplasmic nucleotide-binding oligomerization domain-like receptor protein 3 (NLRP3) forms an inflammasome with apoptosis-associated speck-like protein containing a CARD (ASC) and pro-caspase-1, which is followed by the cleavage of pro-caspase-1 to active caspase-1 and ultimately the activation of IL-1β and IL-18 and induction of pyroptosis in immune cells. NLRP3 activation in kidney diseases aggravates inflammation and subsequent fibrosis, and this effect is abrogated by genetic or pharmacologic deletion of NLRP3. Inflammasome-dependent NLRP3 mediates the progression of kidney diseases by escalating the inflammatory response in immune cells and the cross-talk between immune cells and renal nonimmune cells. However, recent studies have suggested that NLRP3 has several inflammasome-independent functions in the kidney. Inflammasome-independent NLRP3 regulates apoptosis in tubular epithelial cells by interacting with mitochondria and mediating mitochondrial reactive oxygen species production and mitophagy. This review will summarize the mechanisms by which NLRP3 functions in the kidney in both inflammasome-dependent and inflammasome-independent ways and the role of NLRP3 and NLRP3 inhibitors in kidney diseases.
Collapse
Affiliation(s)
- Yang Gyun Kim
- Division of Nephrology, Department of Internal Medicine, Kyung Hee University Medical School, Seoul 02447, Korea; (Y.G.K.); (S.-M.K.); (S.-H.L.)
| | - Su-Mi Kim
- Division of Nephrology, Department of Internal Medicine, Kyung Hee University Medical School, Seoul 02447, Korea; (Y.G.K.); (S.-M.K.); (S.-H.L.)
| | - Ki-Pyo Kim
- Division of Nephrology and Hypertension, Department of Internal Medicine, Inha University of Medicine, Incheon 22212, Korea;
| | - Sang-Ho Lee
- Division of Nephrology, Department of Internal Medicine, Kyung Hee University Medical School, Seoul 02447, Korea; (Y.G.K.); (S.-M.K.); (S.-H.L.)
| | - Ju-Young Moon
- Division of Nephrology, Department of Internal Medicine, Kyung Hee University Medical School, Seoul 02447, Korea; (Y.G.K.); (S.-M.K.); (S.-H.L.)
- Correspondence: ; Tel.: +82-2-440-6262
| |
Collapse
|
38
|
Wang S, Fan J, Mei X, Luan J, Li Y, Zhang X, Chen W, Wang Y, Meng G, Ju D. Interleukin-22 Attenuated Renal Tubular Injury in Aristolochic Acid Nephropathy via Suppressing Activation of NLRP3 Inflammasome. Front Immunol 2019; 10:2277. [PMID: 31616439 PMCID: PMC6768973 DOI: 10.3389/fimmu.2019.02277] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2019] [Accepted: 09/09/2019] [Indexed: 01/03/2023] Open
Abstract
Aristolochic acid nephropathy (AAN), as a rapidly progressive interstitial nephropathy due to excessive ingestion of aristolochia herbal medications, has recently raised considerable concerns among clinicians and researchers as its underlying pathogenic mechanisms are largely unclear. In the current study, we identified NLRP3 inflammasome activation as a novel pathological mechanism of AAN. We found that NLRP3 inflammasome was aberrantly activated both in vivo and in vitro after AA exposure. Blockade of IL-1β and NLRP3 inflammasome activation by IL-1Ra significantly attenuated renal tubular injury and function loss in AA-induced nephropathy. Moreover, NLRP3 or Caspase-1 deficiency protected against renal injury in the mouse model of acute AAN, suggesting that the NLRP3 signaling pathway was probably involved in the pathogenesis of AAN. We also found that administration of IL-22 could markedly attenuate renal tubular injury in AAN. Notably, IL-22 intervention significantly alleviated renal fibrosis and dysfunction in AA-induced nephropathy. Furthermore, IL-22 largely inhibited renal activation of NLRP3 inflammasome in AA-induced nephropathy. These results indicated that IL-22 ameliorated renal tubular injury in AAN through suppression of NLRP3 inflammasome activation. In summary, this study identified renal activation of NLRP3 inflammasome as a novel mechanism underlying the pathogenesis of AAN, thus providing a potential therapeutic strategy for AAN based on suppression of NLRP3 inflammasome activation.
Collapse
Affiliation(s)
- Shaofei Wang
- Minhang Hospital, Fudan University, Shanghai, China
- School of Pharmacy, Fudan University, Shanghai, China
| | - Jiajun Fan
- Minhang Hospital, Fudan University, Shanghai, China
- School of Pharmacy, Fudan University, Shanghai, China
| | - Xiaobin Mei
- Department of Nephrology, Changhai Hospital, Second Military Medical University, Shanghai, China
| | - Jingyun Luan
- Minhang Hospital, Fudan University, Shanghai, China
- School of Pharmacy, Fudan University, Shanghai, China
| | - Yubin Li
- Minhang Hospital, Fudan University, Shanghai, China
- School of Pharmacy, Fudan University, Shanghai, China
| | - Xuyao Zhang
- Minhang Hospital, Fudan University, Shanghai, China
- School of Pharmacy, Fudan University, Shanghai, China
| | - Wei Chen
- Minhang Hospital, Fudan University, Shanghai, China
- School of Pharmacy, Fudan University, Shanghai, China
| | - Yichen Wang
- Minhang Hospital, Fudan University, Shanghai, China
- School of Pharmacy, Fudan University, Shanghai, China
| | - Guangxun Meng
- Unit of Innate Immunity, Key Laboratory of Molecular Virology and Immunology, Institute Pasteur of Shanghai, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Dianwen Ju
- Minhang Hospital, Fudan University, Shanghai, China
- School of Pharmacy, Fudan University, Shanghai, China
| |
Collapse
|
39
|
Yin M, Jiang N, Guo L, Ni Z, Al-Brakati AY, Othman MS, Abdel Moneim AE, Kassab RB. Oleuropein suppresses oxidative, inflammatory, and apoptotic responses following glycerol-induced acute kidney injury in rats. Life Sci 2019; 232:116634. [PMID: 31279782 DOI: 10.1016/j.lfs.2019.116634] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2019] [Revised: 06/30/2019] [Accepted: 07/03/2019] [Indexed: 12/18/2022]
|
40
|
Van Avondt K, Nur E, Zeerleder S. Mechanisms of haemolysis-induced kidney injury. Nat Rev Nephrol 2019; 15:671-692. [PMID: 31455889 DOI: 10.1038/s41581-019-0181-0] [Citation(s) in RCA: 112] [Impact Index Per Article: 18.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/10/2019] [Indexed: 12/16/2022]
Abstract
Intravascular haemolysis is a fundamental feature of chronic hereditary and acquired haemolytic anaemias, including those associated with haemoglobinopathies, complement disorders and infectious diseases such as malaria. Destabilization of red blood cells (RBCs) within the vasculature results in systemic inflammation, vasomotor dysfunction, thrombophilia and proliferative vasculopathy. The haemoprotein scavengers haptoglobin and haemopexin act to limit circulating levels of free haemoglobin, haem and iron - potentially toxic species that are released from injured RBCs. However, these adaptive defence systems can fail owing to ongoing intravascular disintegration of RBCs. Induction of the haem-degrading enzyme haem oxygenase 1 (HO1) - and potentially HO2 - represents a response to, and endogenous defence against, large amounts of cellular haem; however, this system can also become saturated. A frequent adverse consequence of massive and/or chronic haemolysis is kidney injury, which contributes to the morbidity and mortality of chronic haemolytic diseases. Intravascular destruction of RBCs and the resulting accumulation of haemoproteins can induce kidney injury via a number of mechanisms, including oxidative stress and cytotoxicity pathways, through the formation of intratubular casts and through direct as well as indirect proinflammatory effects, the latter via the activation of neutrophils and monocytes. Understanding of the detailed pathophysiology of haemolysis-induced kidney injury offers opportunities for the design and implementation of new therapeutic strategies to counteract the unfavourable and potentially fatal effects of haemolysis on the kidney.
Collapse
Affiliation(s)
- Kristof Van Avondt
- Department of Immunopathology, Sanquin Research, and Landsteiner Laboratory, Academic Medical Centre, University of Amsterdam, Amsterdam, The Netherlands. .,Institute for Cardiovascular Prevention (IPEK), Ludwig Maximilian University of Munich, Munich, Germany.
| | - Erfan Nur
- Department of Haematology, Academic Medical Centre, University of Amsterdam, Amsterdam, The Netherlands
| | - Sacha Zeerleder
- Department of Immunopathology, Sanquin Research, and Landsteiner Laboratory, Academic Medical Centre, University of Amsterdam, Amsterdam, The Netherlands. .,Department of Haematology and Central Haematology Laboratory, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland. .,Department for BioMedical Research, University of Bern, Bern, Switzerland.
| |
Collapse
|
41
|
Dai W, Huang H, Si L, Hu S, Zhou L, Xu L, Deng Y. Melatonin prevents sepsis-induced renal injury via the PINK1/Parkin1 signaling pathway. Int J Mol Med 2019; 44:1197-1204. [PMID: 31432108 PMCID: PMC6713408 DOI: 10.3892/ijmm.2019.4306] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2018] [Accepted: 09/04/2018] [Indexed: 12/13/2022] Open
Abstract
Melatonin (N-acetyl-5-methoxytryptamine; MT) has been shown to have a protective effect against sepsis-induced renal injury, however, the mechanisms underlying the function of MT remain to be elucidated. Therefore, in the present study, the potential mechanisms underlying the preventive role of MT in sepsis-induced renal injury were investigated. Hematoxylin and eosin staining was used to detect the effect of MT on the reduction of renal tissue damage, and immunohistochemistry (IHC), ELISA and western blot analysis were performed to determine the influence of MT on the protein expression of PTEN-induced putative kinase 1 (PINK1), nucleotide-oligomerization binding domain and leucine-rich repeat pyrin domain-containing 3 (NLRP3), apoptosis-associated speck-like protein containing a C-terminal caspase recruitment domain (ASC1), interleukin (IL)-18, IL-1β, IL-6 and cleaved caspase-1. Finally, a TUNEL assay was used to compare the rate of apoptosis of renal tissues among the sham, cecal ligation and puncture (CLP), and CLP + MT groups. The extent of tissue damage in the CLP group was the highest and the extent of tissue damage in the sham group was the lowest. The IHC and western blot analysis showed that the sham group had the highest protein level of PINK1, whereas the CLP group had the lowest protein level of PINK1. By contrast, the sham group had the lowest protein level of NLRP, whereas the CLP group had the highest level of NLRP3. Furthermore, CLP treatment enhanced the protein expression of ASC1 and cleaved caspase-1, whereas the administration of MT reduced the protein expression of ASC1 and cleaved caspase-1 to a certain degree. Finally, the apoptotic rate was found to be the highest in the CLP group and the lowest in the sham group. Taken together, in evaluating the therapeutic effect of MT on sepsis-induced renal injury, the results of the present study showed that MT alleviated sepsis-induced renal injury by regulating the expression of PINK1, Parkin1, NLRP3, ASC and cleaved caspase-1 in rats.
Collapse
Affiliation(s)
- Wenling Dai
- Department of Critical Care Medicine, Yancheng City No. 1 People's Hospital, Yancheng, Jiangsu 224006, P.R. China
| | - Haifeng Huang
- Department of Clinical Laboratory, Sheyang Zhenyang Hospital, Yancheng, Jiangsu 224300, P.R. China
| | - Linjie Si
- Department of Critical Care Medicine, Yancheng City No. 1 People's Hospital, Yancheng, Jiangsu 224006, P.R. China
| | - Shi Hu
- Department of Critical Care Medicine, Yancheng City No. 1 People's Hospital, Yancheng, Jiangsu 224006, P.R. China
| | - Liangliang Zhou
- Department of Critical Care Medicine, Yancheng City No. 1 People's Hospital, Yancheng, Jiangsu 224006, P.R. China
| | - Lingling Xu
- Department of Clinical Laboratory, Yancheng City No. 1 People's Hospital, Yancheng, Jiangsu 224006, P.R. China
| | - Yijun Deng
- Department of Critical Care Medicine, Yancheng City No. 1 People's Hospital, Yancheng, Jiangsu 224006, P.R. China
| |
Collapse
|
42
|
Priante G, Gianesello L, Ceol M, Del Prete D, Anglani F. Cell Death in the Kidney. Int J Mol Sci 2019; 20:E3598. [PMID: 31340541 PMCID: PMC6679187 DOI: 10.3390/ijms20143598] [Citation(s) in RCA: 122] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2019] [Revised: 07/18/2019] [Accepted: 07/18/2019] [Indexed: 12/14/2022] Open
Abstract
Apoptotic cell death is usually a response to the cell's microenvironment. In the kidney, apoptosis contributes to parenchymal cell loss in the course of acute and chronic renal injury, but does not trigger an inflammatory response. What distinguishes necrosis from apoptosis is the rupture of the plasma membrane, so necrotic cell death is accompanied by the release of unprocessed intracellular content, including cellular organelles, which are highly immunogenic proteins. The relative contribution of apoptosis and necrosis to injury varies, depending on the severity of the insult. Regulated cell death may result from immunologically silent apoptosis or from immunogenic necrosis. Recent advances have enhanced the most revolutionary concept of regulated necrosis. Several modalities of regulated necrosis have been described, such as necroptosis, ferroptosis, pyroptosis, and mitochondrial permeability transition-dependent regulated necrosis. We review the different modalities of apoptosis, necrosis, and regulated necrosis in kidney injury, focusing particularly on evidence implicating cell death in ectopic renal calcification. We also review the evidence for the role of cell death in kidney injury, which may pave the way for new therapeutic opportunities.
Collapse
Affiliation(s)
- Giovanna Priante
- Kidney Histomorphology and Molecular Biology Laboratory, Clinical Nephrology, Department of Medicine - DIMED, University of Padua, via Giustiniani 2, 35128 Padova, Italy.
| | - Lisa Gianesello
- Kidney Histomorphology and Molecular Biology Laboratory, Clinical Nephrology, Department of Medicine - DIMED, University of Padua, via Giustiniani 2, 35128 Padova, Italy
| | - Monica Ceol
- Kidney Histomorphology and Molecular Biology Laboratory, Clinical Nephrology, Department of Medicine - DIMED, University of Padua, via Giustiniani 2, 35128 Padova, Italy
| | - Dorella Del Prete
- Kidney Histomorphology and Molecular Biology Laboratory, Clinical Nephrology, Department of Medicine - DIMED, University of Padua, via Giustiniani 2, 35128 Padova, Italy
| | - Franca Anglani
- Kidney Histomorphology and Molecular Biology Laboratory, Clinical Nephrology, Department of Medicine - DIMED, University of Padua, via Giustiniani 2, 35128 Padova, Italy
| |
Collapse
|
43
|
Guerrero-Hue M, García-Caballero C, Palomino-Antolín A, Rubio-Navarro A, Vázquez-Carballo C, Herencia C, Martín-Sanchez D, Farré-Alins V, Egea J, Cannata P, Praga M, Ortiz A, Egido J, Sanz AB, Moreno JA. Curcumin reduces renal damage associated with rhabdomyolysis by decreasing ferroptosis-mediated cell death. FASEB J 2019; 33:8961-8975. [PMID: 31034781 DOI: 10.1096/fj.201900077r] [Citation(s) in RCA: 184] [Impact Index Per Article: 30.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Acute kidney injury is a common complication of rhabdomyolysis. A better understanding of this syndrome may be useful to identify novel therapeutic targets because there is no specific treatment so far. Ferroptosis is an iron-dependent form of regulated nonapoptotic cell death that is involved in renal injury. In this study, we investigated whether ferroptosis is associated with rhabdomyolysis-mediated renal damage, and we studied the therapeutic effect of curcumin, a powerful antioxidant with renoprotective properties. Induction of rhabdomyolysis in mice increased serum creatinine levels, endothelial damage, inflammatory chemokines, and cytokine expression, alteration of redox balance (increased lipid peroxidation and decreased antioxidant defenses), and tubular cell death. Treatment with curcumin initiated before or after rhabdomyolysis induction ameliorated all these pathologic and molecular alterations. Although apoptosis or receptor-interacting protein kinase (RIPK)3-mediated necroptosis were activated in rhabdomyolysis, our results suggest a key role of ferroptosis. Thus, treatment with ferrostatin 1, a ferroptosis inhibitor, improved renal function in glycerol-injected mice, whereas no beneficial effects were observed with the pan-caspase inhibitor carbobenzoxy-valyl-alanyl-aspartyl-(O-methyl)-fluoromethylketone or in RIPK3-deficient mice. In cultured renal tubular cells, myoglobin (Mb) induced ferroptosis-sensitive cell death that was also inhibited by curcumin. Mechanistic in vitro studies showed that curcumin reduced Mb-mediated inflammation and oxidative stress by inhibiting the TLR4/NF-κB axis and activating the cytoprotective enzyme heme oxygenase 1. Our findings are the first to demonstrate the involvement of ferroptosis in rhabdomyolysis-associated renal damage and its sensitivity to curcumin treatment. Therefore, curcumin may be a potential therapeutic approach for patients with this syndrome.-Guerrero-Hue, M., García-Caballero, C., Palomino-Antolín, A., Rubio-Navarro, A., Vázquez-Carballo, C., Herencia, C., Martín-Sanchez, D., Farré-Alins, V., Egea, J., Cannata, P., Praga, M., Ortiz, A., Egido, J., Sanz, A. B., Moreno, J. A. Curcumin reduces renal damage associated with rhabdomyolysis by decreasing ferroptosis-mediated cell death.
Collapse
Affiliation(s)
- Melania Guerrero-Hue
- Renal, Vascular, and Diabetes Research Laboratory, Fundación Instituto de Investigaciones Sanitarias, Fundación Jiménez Díaz, Autónoma University, Madrid, Spain
| | - Cristina García-Caballero
- Renal, Vascular, and Diabetes Research Laboratory, Fundación Instituto de Investigaciones Sanitarias, Fundación Jiménez Díaz, Autónoma University, Madrid, Spain
| | - Alejandra Palomino-Antolín
- Instituto de Investigación Sanitaria (IIS), Hospital Universitario de la Princesa, Madrid, Spain.,Instituto Teófilo Hernando, Departament of Pharmacology and Therapeutics, Medicine Faculty, Autónoma University, Madrid, Spain
| | - Alfonso Rubio-Navarro
- Renal, Vascular, and Diabetes Research Laboratory, Fundación Instituto de Investigaciones Sanitarias, Fundación Jiménez Díaz, Autónoma University, Madrid, Spain
| | - Cristina Vázquez-Carballo
- Renal, Vascular, and Diabetes Research Laboratory, Fundación Instituto de Investigaciones Sanitarias, Fundación Jiménez Díaz, Autónoma University, Madrid, Spain
| | - Carmen Herencia
- Renal, Vascular, and Diabetes Research Laboratory, Fundación Instituto de Investigaciones Sanitarias, Fundación Jiménez Díaz, Autónoma University, Madrid, Spain
| | - Diego Martín-Sanchez
- Renal, Vascular, and Diabetes Research Laboratory, Fundación Instituto de Investigaciones Sanitarias, Fundación Jiménez Díaz, Autónoma University, Madrid, Spain
| | - Víctor Farré-Alins
- Instituto de Investigación Sanitaria (IIS), Hospital Universitario de la Princesa, Madrid, Spain.,Instituto Teófilo Hernando, Departament of Pharmacology and Therapeutics, Medicine Faculty, Autónoma University, Madrid, Spain
| | - Javier Egea
- Instituto de Investigación Sanitaria (IIS), Hospital Universitario de la Princesa, Madrid, Spain.,Instituto Teófilo Hernando, Departament of Pharmacology and Therapeutics, Medicine Faculty, Autónoma University, Madrid, Spain
| | - Pablo Cannata
- Pathology Department, Fundación Instituto de Investigaciones Sanitarias, Fundación Jiménez Díaz, Autónoma University, Madrid, Spain
| | - Manuel Praga
- Department of Nephrology, 12 de Octubre Hospital, Madrid, Spain
| | - Alberto Ortiz
- Renal, Vascular, and Diabetes Research Laboratory, Fundación Instituto de Investigaciones Sanitarias, Fundación Jiménez Díaz, Autónoma University, Madrid, Spain
| | - Jesús Egido
- Renal, Vascular, and Diabetes Research Laboratory, Fundación Instituto de Investigaciones Sanitarias, Fundación Jiménez Díaz, Autónoma University, Madrid, Spain.,Diabetes and Associated Metabolic Diseases Networking Biomedical Research Center (CIBERDEM), Madrid, Spain
| | - Ana Belén Sanz
- Renal, Vascular, and Diabetes Research Laboratory, Fundación Instituto de Investigaciones Sanitarias, Fundación Jiménez Díaz, Autónoma University, Madrid, Spain
| | - Juan Antonio Moreno
- Renal, Vascular, and Diabetes Research Laboratory, Fundación Instituto de Investigaciones Sanitarias, Fundación Jiménez Díaz, Autónoma University, Madrid, Spain.,Department of Cell Biology, Physiology, and Immunology, Maimonides Biomedical Research Institute of Cordoba (IMIBIC), University of Cordoba, Cordoba, Spain.,Biomedical Research Networking Center on Cardiovascular Diseases (CIBERCV), Madrid, Spain
| |
Collapse
|
44
|
Giuliani KTK, Kassianos AJ, Healy H, Gois PHF. Pigment Nephropathy: Novel Insights into Inflammasome-Mediated Pathogenesis. Int J Mol Sci 2019; 20:E1997. [PMID: 31018590 PMCID: PMC6514712 DOI: 10.3390/ijms20081997] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2019] [Revised: 04/17/2019] [Accepted: 04/17/2019] [Indexed: 01/05/2023] Open
Abstract
Pigment nephropathy is an acute decline in renal function following the deposition of endogenous haem-containing proteins in the kidneys. Haem pigments such as myoglobin and haemoglobin are filtered by glomeruli and absorbed by the proximal tubules. They cause renal vasoconstriction, tubular obstruction, increased oxidative stress and inflammation. Haem is associated with inflammation in sterile and infectious conditions, contributing to the pathogenesis of many disorders such as rhabdomyolysis and haemolytic diseases. In fact, haem appears to be a signalling molecule that is able to activate the inflammasome pathway. Recent studies highlight a pathogenic function for haem in triggering inflammatory responses through the activation of the nucleotide-binding domain-like receptor protein 3 (NLRP3) inflammasome. Among the inflammasome multiprotein complexes, the NLRP3 inflammasome has been the most widely characterized as a trigger of inflammatory caspases and the maturation of interleukin-18 and -1β. In the present review, we discuss the latest evidence on the importance of inflammasome-mediated inflammation in pigment nephropathy. Finally, we highlight the potential role of inflammasome inhibitors in the prophylaxis and treatment of pigment nephropathy.
Collapse
Affiliation(s)
- Kurt T K Giuliani
- Kidney Health Service, Royal Brisbane and Women's Hospital, Brisbane, QLD 4029, Australia.
- Conjoint Kidney Research Laboratory, Chemical Pathology-Pathology Queensland, Brisbane, QLD 4029, Australia.
- Faculty of Medicine, University of Queensland, Brisbane, QLD 4006, Australia.
| | - Andrew J Kassianos
- Kidney Health Service, Royal Brisbane and Women's Hospital, Brisbane, QLD 4029, Australia.
- Conjoint Kidney Research Laboratory, Chemical Pathology-Pathology Queensland, Brisbane, QLD 4029, Australia.
- Faculty of Medicine, University of Queensland, Brisbane, QLD 4006, Australia.
- Institute of Health and Biomedical Innovation/School of Biomedical Sciences, Queensland University of Technology, Brisbane, QLD 4059, Australia.
| | - Helen Healy
- Kidney Health Service, Royal Brisbane and Women's Hospital, Brisbane, QLD 4029, Australia.
- Conjoint Kidney Research Laboratory, Chemical Pathology-Pathology Queensland, Brisbane, QLD 4029, Australia.
- Faculty of Medicine, University of Queensland, Brisbane, QLD 4006, Australia.
| | - Pedro H F Gois
- Kidney Health Service, Royal Brisbane and Women's Hospital, Brisbane, QLD 4029, Australia.
- Conjoint Kidney Research Laboratory, Chemical Pathology-Pathology Queensland, Brisbane, QLD 4029, Australia.
- Faculty of Medicine, University of Queensland, Brisbane, QLD 4006, Australia.
| |
Collapse
|
45
|
Abd-Ellatif RN, Hegab II, Atef MM, Sadek MT, Hafez YM. Diacerein protects against glycerol-induced acute kidney injury: Modulating oxidative stress, inflammation, apoptosis and necroptosis. Chem Biol Interact 2019; 306:47-53. [PMID: 30974099 DOI: 10.1016/j.cbi.2019.04.008] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2019] [Revised: 03/31/2019] [Accepted: 04/05/2019] [Indexed: 12/18/2022]
Abstract
Necroptosis is suggested to have an important role in the pathogenesis of rhabdomyolysis induced acute kidney injury (AKI). In this study, the renoprotective effect of diacerein on glycerol-induced AKI was investigated. Twenty four male albino rats were included in this study and divided into four groups: (group I) saline control group, (group II) glycerol-treated group, (groups III&IV) diacerein + glycerol -treated groups (25 and 50 mg/kg/day) respectively. Renal malondialdehyde (MDA) level in addition to catalase and heme oxygenase (HO) activities were estimated. Comet assay and histopathological changes were evaluated. The levels of pro-apoptotic Bcl-2-associated X (Bax) protein, tumor necrosis factor alpha (TNF-α) and receptor-interacting serine/threonine-protein kinases 3 (RIPK3) were measured by ELISA. RIPK3 and mixed lineage kinase domain-like pseudokinase (MLKL) mRNA expression were assessed by real time PCR. Glycerol treatment caused significant renal histological abnormalities and functional impairment (increased urea and creatinine). Increased levels of renal MDA with concomitant decrease in renal catalase activity and significant DNA damage in comet assay were observed. High expression of RIPK3 and MLKL in the glycerol-treated group with marked elevation of Bax, TNF-α and RIPK3 levels and HO-1 activity were also documented. Diacerein treatment dependently attenuated glycerol induced structural and functional changes in kidney and significantly elicit reduction of renal tissue oxidative damage whereas it decreased renal expression of RIPK3 and MLKL, and decreased Bax, TNF-α and RIPK3 levels and HO-1 activity. CONCLUSION: These results demonstrated that diacerein might have potential application in the amelioration of AKI via its anti-oxidant, anti-inflammatory, anti-apoptotic and anti-necroptotic effects.
Collapse
Affiliation(s)
| | - Islam Ibrahim Hegab
- Department of Physiology, Faculty of Medicine, Tanta University, Tanta, Egypt
| | - Marwa Mohamed Atef
- Department of Medical Biochemistry, Faculty of Medicine, Tanta University, Tanta, Egypt.
| | - Mona Tayssir Sadek
- Department of Histology, Faculty of Medicine, Tanta University, Tanta, Egypt
| | - Yasser Mostafa Hafez
- Department of Internal Medicine, Faculty of Medicine, Tanta University, Tanta, Egypt
| |
Collapse
|
46
|
Chew STH, Hwang NC. Acute Kidney Injury After Cardiac Surgery: A Narrative Review of the Literature. J Cardiothorac Vasc Anesth 2019; 33:1122-1138. [DOI: 10.1053/j.jvca.2018.08.003] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/18/2018] [Indexed: 02/07/2023]
|
47
|
Amini N, Sarkaki A, Dianat M, Mard SA, Ahangarpour A, Badavi M. The renoprotective effects of naringin and trimetazidine on renal ischemia/reperfusion injury in rats through inhibition of apoptosis and downregulation of micoRNA-10a. Biomed Pharmacother 2019; 112:108568. [PMID: 30780111 DOI: 10.1016/j.biopha.2019.01.029] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2018] [Revised: 01/06/2019] [Accepted: 01/08/2019] [Indexed: 10/27/2022] Open
Abstract
Renal Ischemia-Reperfusion (IR) injury occurs due to circulatory shock and renal transplantation, leading to mortality and morbidity worldwide. The primary purpose of the current study was to evaluate the renoprotective effects of the naringin (NAR) and trimetazidine (TMZ) on IR injury, renal hemodynamics, antioxidant capacity, microRNA-10a, and expression of apoptosis factors. Forty rats were divided into five groups randomly: Sham, IR injury, (TMZ, 5 mg/kg), (NAR pretreatment, 100 mg/kg), and TMZ plus NAR. The sham group underwent the identical surgical procedure as the other groups, except for the application of clamps. After anesthesia, IR injury was induced by 45 min of ischemia, followed by reperfusion for 4 h. Tissue and blood samples were collected for evaluation of renal function, antioxidant activity and, biochemical and molecular parameters. Administration of the NAR, TMZ, and their combination decreased the plasma level of microRNA-10a, caspase-3, and Bcl-2 associated x protein (Bax) mRNA expression, but increased the B- cell lymphoma 2 (Bcl-2) mRNA expression in the kidney tissue. In addition, antioxidant activity, renal blood flow, creatinine clearance (CCr), and fractional excretion of sodium (FENa) were improved. The NAR, TMZ, and their combination can prevent renal I/R injury through promotion of the level of antioxidant enzymes, as well as decrease of microRNA-10a and anti-apoptosis properties. Our data also suggest that NAR, TMZ, or their combination might be beneficial as potent therapeutic factors against renal IR injury.
Collapse
Affiliation(s)
- Negin Amini
- Department of Physiology, School of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Alireza Sarkaki
- Department of Physiology, School of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran; The Persian Gulf Physiology Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Mahin Dianat
- Department of Physiology, School of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran; The Persian Gulf Physiology Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Seyyed Ali Mard
- Department of Physiology, School of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Akram Ahangarpour
- Department of Physiology, School of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Mohammad Badavi
- Department of Physiology, School of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran; The Persian Gulf Physiology Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran.
| |
Collapse
|
48
|
Anticoagulant Related Nephropathy Induced by Dabigatran. Case Rep Nephrol 2018; 2018:7381505. [PMID: 30631615 PMCID: PMC6304482 DOI: 10.1155/2018/7381505] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2018] [Accepted: 11/19/2018] [Indexed: 11/18/2022] Open
Abstract
We describe a case of biopsy-proven dabigatran related nephropathy in a patient without underlying IgA nephropathy. To date, dabigatran related nephropathy was only reported in patients with concurrent or undiagnosed IgA nephropathy, suggesting that it may predispose patients to dabigatran associated injury. The patient is an 81-year-old woman with multiple medical comorbidities, including nonvalvular atrial fibrillation, who was anticoagulated with dabigatran. She presented to hospital with acute kidney injury in the setting of volume overload. Her estimated glomerular filtration rate decreased from a baseline of 57 mL/min/1.73 m2 to 4 mL/min/1.73 m2, necessitating hemodialysis. Renal ultrasound findings, fractional excretion of sodium, and urinalysis suggested acute kidney injury. Renal biopsy showed acute tubular injury, tubular red blood cell casts, and an absence of active glomerulonephritis, similar to the pathological findings of warfarin related nephropathy. A diagnosis of anticoagulant related nephropathy secondary to dabigatran was therefore established. This case demonstrates that dabigatran, like warfarin, may increase tubular bleeding risk in patients, irrespective of underlying kidney or glomerular disease.
Collapse
|
49
|
Sun X, Luan Q, Qiu S. Valsartan prevents glycerol-induced acute kidney injury in male albino rats by downregulating TLR4 and NF-κB expression. Int J Biol Macromol 2018; 119:565-571. [DOI: 10.1016/j.ijbiomac.2018.07.149] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2018] [Revised: 07/22/2018] [Accepted: 07/23/2018] [Indexed: 12/11/2022]
|
50
|
van Swelm RPL, Vos M, Verhoeven F, Thévenod F, Swinkels DW. Endogenous hepcidin synthesis protects the distal nephron against hemin and hemoglobin mediated necroptosis. Cell Death Dis 2018; 9:550. [PMID: 29749404 PMCID: PMC5945780 DOI: 10.1038/s41419-018-0568-z] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2017] [Revised: 03/30/2018] [Accepted: 04/06/2018] [Indexed: 12/11/2022]
Abstract
Hemoglobinuria is associated with kidney injury in various hemolytic pathologies. Currently, there is no treatment available and its pathophysiology is not completely understood. Here we studied the potential detrimental effects of hemoglobin (Hb) exposure to the distal nephron (DN). Involvement of the DN in Hb kidney injury was suggested by the induction of renal hepcidin synthesis (p < 0.001) in mice repeatedly injected with intravenous Hb. Moreover, the hepcidin induction was associated with a decline in urinary kidney injury markers 24p3/NGAL and KIM1, suggesting a role for hepcidin in protection against Hb kidney injury. We demonstrated that uptake of Hb in the mouse cortical collecting duct cells (mCCDcl1) is mediated by multi-protein ligand receptor 24p3R, as indicated by a significant 90% reduction in Hb uptake (p < 0.001) after 24p3R silencing. Moreover, incubation of mCCDcl1 cells with Hb or hemin for 4 or 24 h resulted in hepcidin synthesis and increased mRNA expression of markers for oxidative, inflammatory and ER stress, but no cell death as indicated by apoptosis staining. A protective role for cellular hepcidin against Hb-induced injury was demonstrated by aggravation of oxidative, inflammatory and ER stress after 4 h Hb or hemin incubation in hepcidin silenced mCCDcl1 cells. Hepcidin silencing potentiated hemin-mediated cell death that could be diminished by co-incubation of Nec-1, suggesting that endogenous hepcidin prevents necroptosis. Combined, these results demonstrate that renal hepcidin synthesis protects the DN against hemin and hemoglobin-mediated injury.
Collapse
Affiliation(s)
- Rachel P L van Swelm
- Department of Laboratory Medicine, Radboud university medical center, Radboud Institute for Molecular Life Sciences, Nijmegen, The Netherlands.
| | - Madelon Vos
- Department of Laboratory Medicine, Radboud university medical center, Radboud Institute for Molecular Life Sciences, Nijmegen, The Netherlands
| | - Frank Verhoeven
- Department of Laboratory Medicine, Radboud university medical center, Radboud Institute for Molecular Life Sciences, Nijmegen, The Netherlands
| | - Frank Thévenod
- Institute of Physiology, Pathophysiology & Toxicology, Center for Biomedical Training and Research, University of Witten/Herdecke, Witten, Germany
| | - Dorine W Swinkels
- Department of Laboratory Medicine, Radboud university medical center, Radboud Institute for Molecular Life Sciences, Nijmegen, The Netherlands
| |
Collapse
|