Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 491
1.
Am J Physiol Cell Physiol ; 326(5): C1423-C1436, 2024 May 01.
Article En | MEDLINE | ID: mdl-38497113

Nicotinamide adenine dinucleotide (NAD+) is a pivotal coenzyme, essential for cellular reactions, metabolism, and mitochondrial function. Depletion of kidney NAD+ levels and reduced de novo NAD+ synthesis through the tryptophan-kynurenine pathway are linked to acute kidney injury (AKI), whereas augmenting NAD+ shows promise in reducing AKI. We investigated de novo NAD+ biosynthesis using in vitro, ex vivo, and in vivo models to understand its role in AKI. Two-dimensional (2-D) cultures of human primary renal proximal tubule epithelial cells (RPTECs) and HK-2 cells showed limited de novo NAD+ synthesis, likely due to low pathway enzyme gene expression. Using three-dimensional (3-D) spheroid culture model improved the expression of tubular-specific markers and enzymes involved in de novo NAD+ synthesis. However, de novo NAD+ synthesis remained elusive in the 3-D spheroid culture, regardless of injury conditions. Further investigation revealed that 3-D cultured cells could not metabolize tryptophan (Trp) beyond kynurenine (KYN). Intriguingly, supplementation of 3-hydroxyanthranilic acid into RPTEC spheroids was readily incorporated into NAD+. In a human precision-cut kidney slice (PCKS) ex vivo model, de novo NAD+ synthesis was limited due to substantially downregulated kynurenine 3-monooxygenase (KMO), which is responsible for KYN to 3-hydroxykynurenine conversion. KMO overexpression in RPTEC 3-D spheroids successfully reinstated de novo NAD+ synthesis from Trp. In addition, in vivo study demonstrated that de novo NAD+ synthesis is intact in the kidney of the healthy adult mice. Our findings highlight disrupted tryptophan-kynurenine NAD+ synthesis in in vitro cellular models and an ex vivo kidney model, primarily attributed to KMO downregulation.NEW & NOTEWORTHY Nicotinamide adenine dinucleotide (NAD+) is essential in regulating mitochondrial function. Reduced NAD+ synthesis through the de novo pathway is associated with acute kidney injury (AKI). Our study reveals a disruption in de novo NAD+ synthesis in proximal tubular models, but not in vivo, attributed to downregulation of enzyme kynurenine 3-monooxygenase (KMO). These findings highlight a crucial role of KMO in governing de novo NAD+ biosynthesis within the kidney, shedding light on potential AKI interventions.


Epithelial Cells , Kidney Tubules, Proximal , Kynurenine 3-Monooxygenase , NAD , Tryptophan , Animals , Humans , Mice , Acute Kidney Injury/metabolism , Acute Kidney Injury/pathology , Acute Kidney Injury/enzymology , Cell Line , Cells, Cultured , Epithelial Cells/metabolism , Kidney Tubules, Proximal/metabolism , Kynurenine/metabolism , Kynurenine 3-Monooxygenase/metabolism , Kynurenine 3-Monooxygenase/genetics , Mice, Inbred C57BL , NAD/metabolism , NAD/biosynthesis , Tryptophan/metabolism
2.
BMC Pediatr ; 22(1): 429, 2022 07 20.
Article En | MEDLINE | ID: mdl-35854249

BACKGROUND: The 1-year cumulative incidence of AKI reportedly is high (52%) in pediatric neoplastic disorders. About half of these events occur within 2 weeks. However, subclinical AKI episodes may remain unrecognized by the conventional creatinine-based approaches. We investigated the diagnostic value of urinary N-acetyl-ß-D-glucosaminidase (uNAG) as an early marker of acute kidney injury (AKI). METHODS: In our retrospective study, 33 children with neoplastic disorders were inculded who had serial uNAG tests (at least 5 samples/patient) with a total of 367 uNAG measurements. Renal function was determined by cystatin-C and creatinine based GFR, and relative increase of uNAG index (uNAGRI). We focused on detecting both clinical and subclinical AKI episodes (according to Biomarker-Guided Risk Assessment using pRIFLE criteria and /or elevated uNAG levels) and the incidence of chronic kidney damage. RESULTS: Sixty episodes in 26 patients, with positivity at least in one parameter of kidney panel, were identified during the observation period. We detected 18/60 clinical and 12/60 subclinical renal episodes. In 27/60 episodes only uNAG values was elevated with no therapeutic consequence at presentation. Two patients were detected with decreased initial creatinine levels with 3 "silent" AKI. In 13 patients, modest elevation of uNAG persisted suggesting mild, reversible tubular damage, while chronic tubuloglomerular injury occurred in 5 patients. Based on ROC analysis for the occurence of AKI, uNAGRI significantly indicated the presence of AKI, the sensitivity and specificity are higher than the changes of GFRCreat. Serial uNAG measurements are recommended for  the reduction of the great amount of false positive uNAG results, often due to overhydratation. CONCLUSION: Use of Biomarker-guided Risk Assessment for AKI identified 1.5 × more clinical and subclinical AKI episodes than with creatinine alone in our pediatric cancer patients. Based on the ROC curve for the occurence of AKI, uNAGRI has relatively high sensitivity and specificity comparable to changes of GFRCysC. The advantage of serial uNAG measurements is to decrease the number of false positive results. TRIAL REGISTRATION: The consent to participate is not applicable because it was not reqired for ethical approval and it is a retrospectiv study.


Acute Kidney Injury , Neoplasms , Acetylglucosaminidase/urine , Acute Kidney Injury/diagnosis , Acute Kidney Injury/enzymology , Acute Kidney Injury/urine , Biomarkers/urine , Child , Creatinine/urine , Early Detection of Cancer , Humans , Neoplasms/diagnosis , Neoplasms/urine , Retrospective Studies
3.
Int J Mol Sci ; 22(21)2021 Nov 02.
Article En | MEDLINE | ID: mdl-34769337

Ischemia/reperfusion injury (IRI) is a complex pathophysiological process characterized by blood circulation disorder caused by various factors, such as traumatic shock, surgery, organ transplantation, and thrombus. Severe metabolic dysregulation and tissue structure destruction are observed upon restoration of blood flow to the ischemic tissue. The kidney is a highly perfused organ, sensitive to ischemia and reperfusion injury, and the incidence of renal IRI has high morbidity and mortality. Several studies showed that infiltration of inflammatory cells, apoptosis, and angiogenesis are important mechanisms involved in renal IRI. Despite advances in research, effective therapies for renal IRI are lacking. Recently it has been demonstrated the role of KYP2047, a selective inhibitor of prolyl oligopeptidase (POP), in the regulation of inflammation, apoptosis, and angiogenesis. Thus, this research focused on the role of POP in kidney ischemia/reperfusion (KI/R). An in vivo model of KI/R was performed and mice were subjected to KYP2047 treatment (intraperitoneal, 0.5, 1 and 5 mg/kg). Histological analysis, Masson's trichrome and periodic acid shift (PAS) staining, immunohistochemical and Western blots analysis, real-time PCR (RT-PCR) and ELISA were performed on kidney samples. Moreover, serum creatinine and blood urea nitrogen (BUN) were quantified. POP-inhibition by KYP2047 treatment, only at the doses of 1 and 5 mg/kg, significantly reduced renal injury and collagen amount, regulated inflammation through canonical and non-canonical NF-κB pathway, and restored renal function. Moreover, KYP2047 modulated angiogenesis markers, such as TGF-ß and VEGF, also slowing down apoptosis. Interestingly, treatment with KYP2047 modulated PP2A activity. Thus, these findings clarified the role of POP inhibition in AKI, also offering novel therapeutic target for renal injury after KI/R.


Acute Kidney Injury/prevention & control , Ischemia/complications , Proline/analogs & derivatives , Prolyl Oligopeptidases/antagonists & inhibitors , Reperfusion Injury/complications , Acute Kidney Injury/enzymology , Acute Kidney Injury/etiology , Acute Kidney Injury/pathology , Animals , Apoptosis , Creatinine/metabolism , Male , Mice , Proline/pharmacology , Signal Transduction
4.
Int Immunopharmacol ; 100: 108097, 2021 Nov.
Article En | MEDLINE | ID: mdl-34521024

Zearalenone (ZEA) is a secondary metabolite produced by fungi such as Fusarium and Fusarium flavum, which is classified as a mycotoxin. Crops and feed in a humid surrounding are widely polluted by ZEA, which further endangering the healthful aquaculture of poultry and even human health. Up to now, prevention and cure of mycotoxicosis is still a crucial subject of poultry husbandry. Baicalin (BAI) is a flavonoid refined from dried roots of Scutellaria baicalensis possessing the function of hepatoprotective, anti-inflammatory, anti-oxidant, and anti-atherosclerotic efficacies.etc. But whether Baicalin also has a protective effect against ZEA intoxication is unclear. Therefore, the aim of this study was to establish a model of ZEA-induced toxic injury in chicks, and then to investigate the way in which Baicalin plays a protective role in the mechanism of ZEA-induced liver and kidney injury in chicks. The results exhibit that Baicalin could not only significantly decrease aspartate aminotransferase (AST) , alanine aminotransferase (ALT) and creatinine (Cre) levels in serum, but also ameliorate ZEA-induced pathologic changes of liver and kidney. Baicalin could also significantly regulate ZEA-induced the changes of catalase (CAT) , malondialdehyde (MDA) , total sulfhydryl group , except for glutathione peroxidase (GSH-px) , and inhibit the mRNA levels of inflammatory cytokines tumor necrosis factor-α (TNF-α) , interleukin-1ß (IL-1ß) and cyclooxygenase-2 (COX-2) with caspase-3 and caspase-11 in the caspase signaling pathway , meanwhile inhibit the cell apoptosis in immunohistochemistry. In summary, we successfully established a model of ZEA-induced liver injury in chicks, and confirm that Baicalin can reduce ZEA-induced liver and kidney injury in chicks. The mechanism of these effects is via inhibiting inflammation, oxidative stress and apoptosis, which also indicates the potential applicability of Baicalin for the prevention and treatment of ZEA-induced toxicity in chicks.


Acute Kidney Injury/prevention & control , Anti-Inflammatory Agents/pharmacology , Antioxidants/pharmacology , Apoptosis/drug effects , Caspases/metabolism , Chemical and Drug Induced Liver Injury/prevention & control , Cytokines/metabolism , Flavonoids/pharmacology , Inflammation Mediators/metabolism , Kidney/drug effects , Liver/drug effects , Oxidative Stress/drug effects , Acute Kidney Injury/enzymology , Acute Kidney Injury/immunology , Acute Kidney Injury/pathology , Animals , Caspases/genetics , Chemical and Drug Induced Liver Injury/enzymology , Chemical and Drug Induced Liver Injury/immunology , Chemical and Drug Induced Liver Injury/pathology , Chickens , Cytokines/genetics , Disease Models, Animal , Kidney/immunology , Kidney/metabolism , Kidney/pathology , Liver/enzymology , Liver/immunology , Liver/pathology , Signal Transduction , Zearalenone
5.
Inflammation ; 44(6): 2323-2332, 2021 Dec.
Article En | MEDLINE | ID: mdl-34585338

Acute kidney injury (AKI) is one of the most common diseases in patients treated in intensive care units. This study was intended to explore the underlying mechanism by which ulinastatin (UTI) influenced the inflammation and apoptosis of renal tubular epithelial cells, HK-2.The effects of UTI on the cell viability of HK-2 cells were first measured by MTT and lactate dehydrogenase (LDH) detection kit. The apoptosis and inflammation of HK-2 cells were then determined by TUNEL, western blot, ELISA, and RT-qPCR. Then, the proteins in the Toll-like receptor 4 (TLR4)/nuclear factor κB (NF-κB) and nuclear factor erythroid 2-related factor 2 (Nrf2)/Heme oxygenase 1 (HO-1) signaling pathways were measured by western blot for confirming the relationship between UTI and these pathways. Finally, Nrf-2 inhibitor ML385 and TLR4 activator CCL-34 were respectively used on LPS-induced HK-2 cells exposed to UTI for the conduction of gain-of-function and loss-of-function assays.UTI treatment boosted the cell viability of HK-2 cells damaged by LPS. Furthermore, UTI exposure cut down the apoptosis rate and inhibited the expression inflammatory factors of HK-2 cells induced by LPS. UTI treatment decreased the expression of proteins in the TLR4/NF-κB pathway, increased the HO-1 expression, and prompted the translocation of Nrf2 from the cytoplasm to the nucleus. The alleviated effects of UTI on inflammation and apoptosis LPS-induced HK-2 cells were abolished by ML385 and TLR4, respectively.UTI attenuates LPS-induced inflammation and inhibits endoplasmic reticulum stress-induced apoptosis in renal tubular epithelial cells by regulating TLR4/NF-κB and Nrf2/HO-1 pathways.


Acute Kidney Injury/prevention & control , Anti-Inflammatory Agents/pharmacology , Apoptosis/drug effects , Endoplasmic Reticulum Stress/drug effects , Epithelial Cells/drug effects , Glycoproteins/pharmacology , Heme Oxygenase-1/metabolism , Kidney Tubules/drug effects , NF-E2-Related Factor 2/metabolism , NF-kappa B/metabolism , Nephritis/prevention & control , Toll-Like Receptor 4/metabolism , Acute Kidney Injury/enzymology , Acute Kidney Injury/immunology , Acute Kidney Injury/pathology , Cell Line , Epithelial Cells/enzymology , Epithelial Cells/immunology , Epithelial Cells/pathology , Humans , Inflammation Mediators/metabolism , Kidney Tubules/enzymology , Kidney Tubules/immunology , Kidney Tubules/pathology , Lipopolysaccharides/toxicity , Nephritis/enzymology , Nephritis/immunology , Nephritis/pathology , Signal Transduction
6.
Front Immunol ; 12: 685523, 2021.
Article En | MEDLINE | ID: mdl-34335587

Recent studies have shown that autophagy upregulation can attenuate sepsis-induced acute kidney injury (SAKI). The tumor suppressor p53 has emerged as an autophagy regulator in various forms of acute kidney injury (AKI). Our previous studies showed that p53 acetylation exacerbated hemorrhagic shock-induced AKI and lipopolysaccharide (LPS)-induced endothelial barrier dysfunction. However, the role of p53-regulated autophagy in SAKI has not been examined and requires clarification. In this study, we observed the dynamic changes of autophagy in renal tubular epithelial cells (RTECs) and verified the protective effects of autophagy activation on SAKI. We also examined the changes in the protein expression, intracellular distribution (nuclear and cytoplasmic), and acetylation/deacetylation levels of p53 during SAKI following cecal ligation and puncture (CLP) or LPS treatment in mice and in a LPS-challenged human RTEC cell line (HK-2 cells). After sepsis stimulation, the autophagy levels of RTECs increased temporarily, followed by a sharp decrease. Autophagy inhibition was accompanied by an increased renal tubular injury score. By contrast, autophagy agonists could reduce renal tubular damage following sepsis. Surprisingly, the expression of p53 protein in both the renal cortex and HK-2 cells did not significantly change following sepsis stimulation. However, the translocation of p53 from the nucleus to the cytoplasm increased, and the acetylation of p53 was enhanced. In the mechanistic study, we found that the induction of p53 deacetylation, due to either the resveratrol/quercetin -induced activation of the deacetylase Sirtuin 1 (Sirt1) or the mutation of the acetylated lysine site in p53, promoted RTEC autophagy and alleviated SAKI. In addition, we found that acetylated p53 was easier to bind with Beclin1 and accelerated its ubiquitination-mediated degradation. Our study underscores the importance of deacetylated p53-mediated RTEC autophagy in future SAKI treatments.


Acute Kidney Injury/enzymology , Autophagy/drug effects , Kidney Tubules/enzymology , Sepsis/complications , Tumor Suppressor Protein p53/metabolism , Acetylation , Acute Kidney Injury/etiology , Acute Kidney Injury/pathology , Acute Kidney Injury/prevention & control , Animals , Beclin-1/metabolism , Cell Line , Disease Models, Animal , Humans , Kidney Tubules/drug effects , Kidney Tubules/ultrastructure , Lipopolysaccharides/pharmacology , Mice , Mice, Inbred C57BL , Sepsis/metabolism , Sirtuin 1/genetics , Sirtuin 1/metabolism , Survival Analysis , Tumor Suppressor Protein p53/genetics
7.
Heart Surg Forum ; 24(3): E506-E511, 2021 Jun 10.
Article En | MEDLINE | ID: mdl-34173741

BACKGROUND: Aspartate aminotransferase (AST) to alanine aminotransferase (ALT) ratio (AST/ALT) frequently is used in the diagnosis and prognosis of liver diseases, however it is also used in the diagnosis and prognosis of many other diseases, such as myocardial infarction, acute ischemic stroke, and peripheral artery disease. Acute kidney injury (AKI) is one of the most important complications after cardiac surgery and is one of the main causes of morbidity and mortality. The purpose of the study was to analyze the relationship between AST to ALT and AKI after isolated coronary artery bypass graft surgery (CABG). METHODS: We retrospectively reviewed the prospectively collected data of 253 adult patients, who underwent isolated CABG surgery with normal renal function (baseline serum creatinine value <1.4 mg/dL). Preoperative (T0) and postoperative day 1 and day 3 (T1 and T2) serum AST and ALT levels were analyzed, and AST/ALT was calculated. A preoperative AST/ALT of 1.22 was found to be the best cutoff point for predicting postoperative AKI. Kidney injury was interpreted, according to RIFLE classification. The effect of AST to ALT ratio on AKI after CABG was determined using logistic regression analysis, and the results were expressed as odds ratio (OR) with a 95% confidence interval (CI). A P value < .05 was considered statistically significant. RESULTS: Postoperative AKI occurred in 40 patients (15.8%). On logistic regression analysis, higher AST/ALT both preoperatively and postoperatively were associated with an increased incidence of postoperative AKI (T0: OR, 3.983; 95% CI, 1.940-8.180, P < .001, T1: OR, 2.760; 95% CI, 1.381-5.515, P = .004, T2: OR, 2.515; 95% CI, 1.195-5.294, P = .015). CONCLUSION: Preoperative and postoperative elevated AST to ALT ratio seems to be associated with an increased incidence of AKI after elective isolated CABG surgery.


Acute Kidney Injury/enzymology , Alanine Transaminase/blood , Aspartate Aminotransferases/blood , Coronary Artery Bypass/adverse effects , Acute Kidney Injury/epidemiology , Acute Kidney Injury/etiology , Cardiac Surgical Procedures/adverse effects , Female , Humans , Incidence , Male , Middle Aged , Postoperative Complications/enzymology , Postoperative Complications/epidemiology , Prognosis , Retrospective Studies , Risk Factors , Turkey/epidemiology
8.
Int J Mol Sci ; 22(5)2021 Feb 25.
Article En | MEDLINE | ID: mdl-33669091

Oxidative stress and inflammation play important roles in the pathophysiology of acute kidney injury (AKI). Transient receptor potential ankyrin 1 (TRPA1) is a Ca2+-permeable ion channel that is sensitive to reactive oxygen species (ROS). The role of TRPA1 in AKI remains unclear. In this study, we used human and animal studies to assess the role of renal TRPA1 in AKI and to explore the regulatory mechanism of renal TRPA1 in inflammation via in vitro experiments. TRPA1 expression increased in the renal tubular epithelia of patients with AKI. The severity of tubular injury correlated well with tubular TRPA1 or 8-hydroxy-2'-deoxyguanosine expression. In an animal model, renal ischemia-reperfusion injury (IR) increased tubular TRPA1 expression in wild-type (WT) mice. Trpa1-/- mice displayed less IR-induced tubular injury, oxidative stress, inflammation, and dysfunction in kidneys compared with WT mice. In the in vitro model, TRPA1 expression increased in renal tubular cells under hypoxia-reoxygenation injury (H/R) conditions. We demonstrated that H/R evoked a ROS-dependent TRPA1 activation, which elevated intracellular Ca2+ level, increased NADPH oxidase activity, activated MAPK/NF-κB signaling, and increased IL-8. Renal tubular TRPA1 may serve as an oxidative stress sensor and a crucial regulator in the activation of signaling pathways and promote the subsequent transcriptional regulation of IL-8. These actions might be evident in mice with IR or patients with AKI.


Acute Kidney Injury/metabolism , Deoxyguanosine/metabolism , Kidney Tubules/metabolism , NF-kappa B/metabolism , Oxidative Stress/genetics , Reperfusion Injury/metabolism , TRPA1 Cation Channel/metabolism , Acute Kidney Injury/enzymology , Acute Kidney Injury/genetics , Adult , Animals , Calcium/metabolism , Cell Line , Deoxyguanosine/analogs & derivatives , Disease Models, Animal , Epithelium/metabolism , Epithelium/pathology , Humans , Immunohistochemistry , Interleukin-8/metabolism , Kidney Tubules/cytology , Kidney Tubules/enzymology , Kidney Tubules/pathology , MAP Kinase Signaling System/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , NADP/metabolism , Reactive Oxygen Species/metabolism , TRPA1 Cation Channel/genetics
9.
Int J Mol Sci ; 22(4)2021 Feb 18.
Article En | MEDLINE | ID: mdl-33670516

The incidence of kidney disease is rising, constituting a significant burden on the healthcare system and making identification of new therapeutic targets increasingly urgent. The heme oxygenase (HO) system performs an important function in the regulation of oxidative stress and inflammation and, via these mechanisms, is thought to play a role in the prevention of non-specific injuries following acute renal failure or resulting from chronic kidney disease. The expression of HO-1 is strongly inducible by a wide range of stimuli in the kidney, consequent to the kidney's filtration role which means HO-1 is exposed to a wide range of endogenous and exogenous molecules, and it has been shown to be protective in a variety of nephropathological animal models. Interestingly, the positive effect of HO-1 occurs in both hemolysis- and rhabdomyolysis-dominated diseases, where the kidney is extensively exposed to heme (a major HO-1 inducer), as well as in non-heme-dependent diseases such as hypertension, diabetic nephropathy or progression to end-stage renal disease. This highlights the complexity of HO-1's functions, which is also illustrated by the fact that, despite the abundance of preclinical data, no drug targeting HO-1 has so far been translated into clinical use. The objective of this review is to assess current knowledge relating HO-1's role in the kidney and its potential interest as a nephroprotection agent. The potential therapeutic openings will be presented, in particular through the identification of clinical trials targeting this enzyme or its products.


Acute Kidney Injury/metabolism , Heme Oxygenase-1/metabolism , Heme/metabolism , Kidney Diseases/metabolism , Oxidative Stress , Acute Kidney Injury/enzymology , Acute Kidney Injury/pathology , Animals , Humans , Kidney/cytology , Kidney/enzymology , Kidney/metabolism , Kidney Diseases/enzymology , Kidney Diseases/pathology , Kidney Tubules/cytology , Kidney Tubules/enzymology , Kidney Tubules/metabolism , Protective Agents/metabolism
10.
Toxicol Appl Pharmacol ; 418: 115492, 2021 05 01.
Article En | MEDLINE | ID: mdl-33722665

Cisplatin is a commonly used anti-cancer drug, but it induces nephrotoxicity. As a water-soluble vitamin B family member, nicotinamide (NAM) was recently demonstrated to have beneficial effects for renal injury, but its underlying mechanism remains largely unclear. Here, we suggest that NAM may exert protective effects against cisplatin-induced acute kidney injury (AKI) mainly via suppressing the poly ADP-ribose polymerase 1 (PARP1)/p53 pathway. In our experiment, NAM protected against cisplatin-induced apoptosis both in cultured renal proximal tubular cells and AKI in mice. Mechanistically, NAM suppressed the expression and activation of p53, a known mediator of cisplatin-induced AKI. Upstream of p53, NAM attenuated the induction of γ-H2AX, a hallmark of DNA damage response. Interestingly, PARP1 was activated in cisplatin AKI and this activation was inhibited by NAM. Pharmacological inhibition of PARP1 with PJ34 significantly ameliorated p53 activation and cisplatin-induced cell death in RPTCs and AKI in mice. Thus, NAM may protect against cisplatin-induced AKI by suppressing the PARP1/p53 pathway.


Acute Kidney Injury/prevention & control , Cisplatin , Kidney Tubules, Proximal/drug effects , Niacinamide/pharmacology , Poly (ADP-Ribose) Polymerase-1/antagonists & inhibitors , Poly(ADP-ribose) Polymerase Inhibitors/pharmacology , Tumor Suppressor Protein p53/metabolism , Acute Kidney Injury/chemically induced , Acute Kidney Injury/enzymology , Acute Kidney Injury/pathology , Animals , Apoptosis/drug effects , Cell Line , Disease Models, Animal , Histones/metabolism , Kidney Tubules, Proximal/enzymology , Kidney Tubules, Proximal/pathology , Male , Mice, Inbred C57BL , Phosphoproteins/metabolism , Poly (ADP-Ribose) Polymerase-1/metabolism , Rats , Signal Transduction
11.
Mol Cell Endocrinol ; 529: 111257, 2021 06 01.
Article En | MEDLINE | ID: mdl-33781839

The observation that all components of the renin angiotensin system (RAS) are expressed in the kidney and the fact that intratubular angiotensin (Ang) II levels greatly exceed the plasma concentration suggest that the synthesis of renal Ang II occurs independently of the circulating RAS. One of the main components of this so-called intrarenal RAS is angiotensin-converting enzyme (ACE). Although the role of ACE in renal disease is demonstrated by the therapeutic effectiveness of ACE inhibitors in treating several conditions, the exact contribution of intrarenal versus systemic ACE in renal disease remains unknown. Using genetically modified mouse models, our group demonstrated that renal ACE plays a key role in the development of several forms of hypertension. Specifically, although ACE is expressed in different cell types within the kidney, its expression in renal proximal tubular cells is essential for the development of high blood pressure. Besides hypertension, ACE is involved in several other renal diseases such as diabetic kidney disease, or acute kidney injury even when blood pressure is normal. In addition, studies suggest that ACE might mediate at least part of its effect through mechanisms that are independent of the Ang I conversion into Ang II and involve other substrates such as N-acetyl-seryl-aspartyl-lysyl-proline (AcSDKP), Ang-(1-7), and bradykinin, among others. In this review, we summarize the recent advances in understanding the contribution of intrarenal ACE to different pathological conditions and provide insight into the many roles of ACE besides the well-known synthesis of Ang II.


Acute Kidney Injury/enzymology , Angiotensin I/metabolism , Diabetic Nephropathies/enzymology , Hypertension/enzymology , Peptide Fragments/metabolism , Peptidyl-Dipeptidase A/metabolism , Renin-Angiotensin System/genetics , Acute Kidney Injury/genetics , Acute Kidney Injury/pathology , Angiotensin I/genetics , Angiotensin II/genetics , Angiotensin II/metabolism , Animals , Blood Pressure/genetics , Bradykinin/metabolism , Diabetic Nephropathies/genetics , Diabetic Nephropathies/pathology , Gene Expression Regulation , Humans , Hypertension/genetics , Hypertension/pathology , Kidney/enzymology , Kidney/pathology , Mice , Oligopeptides/metabolism , Peptide Fragments/genetics , Peptidyl-Dipeptidase A/genetics , Signal Transduction , Water-Electrolyte Balance/genetics
12.
Am J Physiol Renal Physiol ; 320(5): F706-F718, 2021 05 01.
Article En | MEDLINE | ID: mdl-33719570

Cellular metabolic rates in the kidney are critical for maintaining normal renal function. In a hypoxic milieu, cells rely on glycolysis to meet energy needs, resulting in the generation of pyruvate and NADH. In the absence of oxidative phosphorylation, the continuation of glycolysis is dependent on the regeneration of NAD+ from NADH accompanied by the fermentation of pyruvate to lactate. This reaction is catalyzed by lactate dehydrogenase (LDH) isoform A (LDHA), whereas LDH isoform B (LDHB) catalyzes the opposite reaction. LDH is widely used as a potential injury marker as it is released from damaged cells into the urine and serum; however, the precise isoform-specific cellular localization of the enzyme along the nephron has not been characterized. By combining immunohistochemistry results and single-cell RNA-sequencing data on healthy mouse kidneys, we identified that LDHA is primarily expressed in proximal segments, whereas LDHB is expressed in the distal parts of the nephron. In vitro experiments in mouse and human renal proximal tubule cells showed an increase in LDHA following hypoxia with no change in LDHB. Using immunofluorescence, we observed that the overall expression of both LDHA and LDHB proteins decreased following renal ischemia-reperfusion injury as well as in the adenine-diet-induced model of chronic kidney disease. Single-nucleus RNA-sequencing analyses of kidneys following ischemia-reperfusion injury revealed a significant decline in the number of cells expressing detectable levels of Ldha and Ldhb; however, cells that were positive showed increased average expression postinjury, which subsided during the recovery phase. These data provide information on the cell-specific expression of LDHA and LDHB in the normal kidney as well as following acute and chronic kidney disease.NEW & NOTEWORTHY Cellular release of lactate dehydrogenase (LDH) is being used as an injury marker; however, the exact localization of LDH within the nephron remains unclear. We show that LDH isoform A is expressed proximally, whereas isoform B is expressed distally. Both subunit expressions were significantly altered in models of acute kidney injury and chronic kidney disease. Our study provides new insights into basal and postinjury renal lactate metabolism.


Acute Kidney Injury/enzymology , Kidney/enzymology , L-Lactate Dehydrogenase/metabolism , Renal Insufficiency, Chronic/enzymology , Acute Kidney Injury/genetics , Acute Kidney Injury/pathology , Animals , Biomarkers/metabolism , Cell Hypoxia , Cells, Cultured , Disease Models, Animal , Gene Expression Regulation, Enzymologic , Humans , Isoenzymes , Kidney/pathology , L-Lactate Dehydrogenase/genetics , Male , Mice, Inbred C57BL , Renal Insufficiency, Chronic/genetics , Renal Insufficiency, Chronic/pathology , Time Factors
13.
Cell Death Dis ; 12(2): 217, 2021 02 26.
Article En | MEDLINE | ID: mdl-33637691

Our previous studies showed that silent mating-type information regulation 2 homologue-1 (SIRT1, a deacetylase) upregulation could attenuate sepsis-induced acute kidney injury (SAKI). Upregulated SIRT1 can deacetylate certain autophagy-related proteins (Beclin1, Atg5, Atg7 and LC3) in vitro. However, it remains unclear whether the beneficial effect of SIRT1 is related to autophagy induction and the underlying mechanism of this effect is also unknown. In the present study, caecal ligation and puncture (CLP)-induced mice, and an LPS-challenged HK-2 cell line were established to mimic a SAKI animal model and a SAKI cell model, respectively. Our results demonstrated that SIRT1 activation promoted autophagy and attenuated SAKI. SIRT1 deacetylated only Beclin1 but not the other autophagy-related proteins in SAKI. SIRT1-induced autophagy and its protective effect against SAKI were mediated by the deacetylation of Beclin1 at K430 and K437. Moreover, two SIRT1 activators, resveratrol and polydatin, attenuated SAKI in CLP-induced septic mice. Our study was the first to demonstrate the important role of SIRT1-induced Beclin1 deacetylation in autophagy and its protective effect against SAKI. These findings suggest that pharmacologic induction of autophagy via SIRT1-mediated Beclin1 deacetylation may be a promising therapeutic approach for future SAKI treatment.


Acute Kidney Injury/enzymology , Autophagy , Beclin-1/metabolism , Kidney Tubules, Proximal/enzymology , Sepsis/complications , Sirtuin 1/metabolism , Acetylation , Acute Kidney Injury/etiology , Acute Kidney Injury/pathology , Acute Kidney Injury/prevention & control , Animals , Autophagy/drug effects , Cell Line , Disease Models, Animal , Enzyme Activation , Enzyme Activators/pharmacology , Glucosides/pharmacology , Humans , Kidney Tubules, Proximal/drug effects , Kidney Tubules, Proximal/ultrastructure , Male , Mice, Inbred C57BL , Resveratrol/pharmacology , Sepsis/microbiology , Signal Transduction , Sirtuin 1/genetics , Stilbenes/pharmacology , Time Factors
14.
J Biochem Mol Toxicol ; 35(4): e22692, 2021 Apr.
Article En | MEDLINE | ID: mdl-33404076

Folic acid (FA)-induced acute kidney injury (AKI) is a commonly used model in experimental animals for studying renal injury. This study aimed to investigate the probable protecting impact of nicorandil against FA-induced renal dysfunction. A mouse model was executed by a single injection of FA (250 mg/kg). Nicorandil was orally administrated in two doses (50 and 100 mg/kg) for 10 days. Nicorandil repressed the progression of FA-induced AKI as evidenced by the improvement of histopathological alterations and the substantial decrease of serum levels of creatinine, urea, blood urea nitrogen, malondialdehyde (MDA), and urinary protein levels. Moreover, nicorandil resulted in a profound reduction in oxidative stress as manifested by decreased MDA and increased reduced glutathione and superoxide dismutase in renal tissue. Notably, nicorandil suppressed FA-induced inflammation; it reduced renal levels of nuclear factor-κB, tumor necrosis factor-α, and interleukin-6. Furthermore, nicorandil decreased renal levels of nitric oxide, inducible nitric oxide synthase, and increased endothelial nitric oxide synthase. Lastly, nicorandil efficiently decreased expression of the proapoptotic protein (Bax) and caspase 3. Nicorandil confers dose-dependent protection against FA-induced AKI by alleviating oxidative stress, inflammation besides modulating nitric oxide synthase and reducing apoptosis.


Acute Kidney Injury/drug therapy , Acute Kidney Injury/enzymology , Folic Acid/adverse effects , Nicorandil/pharmacology , Nitric Oxide Synthase Type III/metabolism , Nitric Oxide Synthase Type II/metabolism , Acute Kidney Injury/chemically induced , Animals , Dose-Response Relationship, Drug , Folic Acid/pharmacology , Male , Mice
15.
Toxicol Mech Methods ; 31(1): 67-72, 2021 Jan.
Article En | MEDLINE | ID: mdl-32981412

Twenty-five male Wistar rats (140-170 g) were partitioned into 5 groups (n = 5). 2.5 mg/kg, 5 mg/kg, 10 mg/kg and 20 mg/kg of combine Tartrazine and Erythrosine (T+E; 50:50) were administered for 23 days. Serum urea and creatinine, gene expression and profiling of pro-inflammatory cytokine (Tumor Necrosis Factor- α gene), Caspase-9 and Kidney injury molecule-1 (KIM-1) and histomorphological examination of the kidney were investigated. The fold change of relative gene expression of TNF-α gene showed significantly (p < 0.05) up-regulation in all the treated rats except for the 10 mg/kg T+E treated rats when compared to control rats. Casp-9 and KIM-1 genes were significantly (p < 0.05) up-regulated in low dose treatment (2.5 mg/kg T+E and 5 mg/kg T+E) and down-regulated in high dose treatment (10 mg/kg T+E and 20 mg/kg T+E). However, there was significant (p < 0.05) increase in serum urea concentration in the rats treated with 5 mg/kg T+E and 20 mg/kg T+E while the rats treated with 10 mg/kg T+E indicated a significant (p < 0.05) decrease. Conversely, serum creatinine concentration indicated significant (p < 0.05) increase in10mg/kg T+E and 20 mg/kg T+E treated rats versus the control. From the histomorphological examination of the kidney, there was hypertrophy of the glomeruli in relation to the size of Bowman's capsule in the 10 mg/kg T+E and 20 mg/kg T+E treated rats. Kidney function was impaired as evident in up-regulation of TNF-α gene, KIM-1 gene, and serum urea and creatinine concentration with down-regulation of Casp-9 gene. The combined treatment also tampers with the architecture of the kidney.


Acute Kidney Injury/chemically induced , Caspase 9/metabolism , Cell Adhesion Molecules/metabolism , Coloring Agents/toxicity , Erythrosine/toxicity , Kidney/drug effects , Tartrazine/toxicity , Tumor Necrosis Factor-alpha/metabolism , Acute Kidney Injury/enzymology , Acute Kidney Injury/genetics , Acute Kidney Injury/pathology , Animals , Caspase 9/genetics , Cell Adhesion Molecules/genetics , Creatinine/blood , Dose-Response Relationship, Drug , Gene Expression Regulation , Kidney/enzymology , Kidney/pathology , Male , Rats, Wistar , Tumor Necrosis Factor-alpha/genetics , Urea/blood
16.
Int Immunopharmacol ; 90: 107123, 2021 Jan.
Article En | MEDLINE | ID: mdl-33168411

Sepsis is a life-threatening condition which affects multiple organs including the kidney. Sepsis-induced acute kidney injury (AKI) is a major health burden throughout the globe. Pathogenesis of sepsis-induced AKI is complex; however, it involves both innate and adaptive immune cells such as B cells, T cells, dendritic cells (DCs), macrophages, and neutrophils. Bruton's tyrosine kinase (BTK) is reportedly involved in inflammatory and oxidative signaling in different immune cells, however its contribution with respect to sepsis-induced AKI has not been delineated. This study attempted to investigate the role of BTK and its inhibition on oxidizing enzymes NADPH oxidase (NOX-2) and inducible nitric oxide synthase (iNOS) in DCs, neutrophils, and B cells during AKI. Our data reveal that BTK is activated in DCs, neutrophils, and B cells which causes an increase in AKI associated biochemical markers such as serum creatinine/blood urea nitrogen, renal myeloperoxidase activity, and histopathological disturbances in renal tubular structures. Activation of BTK causes upregulation of NOX-2/iNOS/nitrotyrosine in these immune cells and kidney. Treatment with BTK inhibitor, Ibrutinib causes attenuation in AKI associated dysfunction in biochemical parameters (serum creatinine/blood urea nitrogen, renal myeloperoxidase activity) and oxidative stress in immune cells and kidney (iNOS/NOX2/lipid peroxides/nitrotyrosine/protein carbonyls). In summary, the current investigation reveals a compelling role of BTK signaling in sepsis-induced AKI which is evident from amelioration of AKI associated renal dysfunction after its inhibition.


Acute Kidney Injury/prevention & control , Adenine/analogs & derivatives , Agammaglobulinaemia Tyrosine Kinase/antagonists & inhibitors , B-Lymphocytes/drug effects , Dendritic Cells/drug effects , Enzyme Inhibitors/pharmacology , Kidney/drug effects , Neutrophils/drug effects , Oxidative Stress/drug effects , Piperidines/pharmacology , Sepsis/drug therapy , Acute Kidney Injury/enzymology , Acute Kidney Injury/immunology , Acute Kidney Injury/pathology , Adenine/pharmacology , Agammaglobulinaemia Tyrosine Kinase/metabolism , Animals , B-Lymphocytes/enzymology , B-Lymphocytes/immunology , Dendritic Cells/enzymology , Dendritic Cells/immunology , Disease Models, Animal , Kidney/enzymology , Kidney/immunology , Kidney/pathology , Male , Mice, Inbred BALB C , NADPH Oxidase 2/metabolism , Neutrophils/enzymology , Neutrophils/immunology , Nitric Oxide Synthase Type II/metabolism , Sepsis/enzymology , Sepsis/immunology , Signal Transduction
17.
Gastroenterology ; 160(5): 1725-1740.e2, 2021 04.
Article En | MEDLINE | ID: mdl-33309778

BACKGROUND & AIMS: We recently showed that alcoholic hepatitis (AH) is characterized by dedifferentiation of hepatocytes and loss of mature functions. Glucose metabolism is tightly regulated in healthy hepatocytes. We hypothesize that AH may lead to metabolic reprogramming of the liver, including dysregulation of glucose metabolism. METHODS: We performed integrated metabolomic and transcriptomic analyses of liver tissue from patients with AH or alcoholic cirrhosis or normal liver tissue from hepatic resection. Focused analyses of chromatin immunoprecipitation coupled to DNA sequencing was performed. Functional in vitro studies were performed in primary rat and human hepatocytes and HepG2 cells. RESULTS: Patients with AH exhibited specific changes in the levels of intermediates of glycolysis/gluconeogenesis, the tricarboxylic acid cycle, and monosaccharide and disaccharide metabolism. Integrated analysis of the transcriptome and metabolome showed the used of alternate energetic pathways, metabolite sinks and bottlenecks, and dysregulated glucose storage in patients with AH. Among genes involved in glucose metabolism, hexokinase domain containing 1 (HKDC1) was identified as the most up-regulated kinase in patients with AH. Histone active promoter and enhancer markers were increased in the HKDC1 genomic region. High HKDC1 levels were associated with the development of acute kidney injury and decreased survival. Increased HKDC1 activity contributed to the accumulation of glucose-6-P and glycogen in primary rat hepatocytes. CONCLUSIONS: Altered metabolite levels and messenger RNA expression of metabolic enzymes suggest the existence of extensive reprogramming of glucose metabolism in AH. Increased HKDC1 expression may contribute to dysregulated glucose metabolism and represents a novel biomarker and therapeutic target for AH.


Cell Dedifferentiation , Energy Metabolism , Gene Expression Profiling , Glucose/metabolism , Hepatitis, Alcoholic/enzymology , Hepatocytes/enzymology , Hexokinase/metabolism , Liver/enzymology , Metabolomics , Acute Kidney Injury/enzymology , Acute Kidney Injury/genetics , Adaptation, Physiological , Animals , Europe , Female , Gene Expression Regulation, Enzymologic , Glucose-6-Phosphate/metabolism , Glycogen/metabolism , Hep G2 Cells , Hepatitis, Alcoholic/genetics , Hepatitis, Alcoholic/pathology , Hepatocytes/pathology , Hexokinase/genetics , Humans , Liver/pathology , Male , Metabolome , Middle Aged , Rats, Wistar , Transcriptome , United States
18.
Clin Biochem ; 88: 49-55, 2021 Feb.
Article En | MEDLINE | ID: mdl-33307060

OBJECTIVES: All patients who undergo cardiopulmonary bypass (CPB) experience some degree of ischemia reperfusion injury (IRI). Severe IRI-induced acute kidney injury (AKI) occurs in approximately 1-2% of patients undergoing CPB. Previous studies using activity-based protein profiling of urine identified group XV phospholipase A2, PLA2G15/LPLA2, as potentially associated with patients who develop AKI post CPB. The present study examined urinary PLA2G15/LPLA2 activity during CPB and in the near postoperative period for associations with subsequent AKI development. DESIGN & METHODS: Samples were collected in a nested case controlled cohort of 21 patients per group who either did (AKI) or did not (non-AKI) develop AKI post-operatively. Serum and urine samples from each patient before, during and after CPB were assayed for PLA2G15/LPLA2 activity. RESULTS: Urine activity significantly increased during the intra operative period. In contrast the activities in paired sera were markedly decreased during CPB. There was no correlation between the serum and urine activity levels of patients. There were no significant differences in activity levels of PLA2G15/LPLA2 in the urine or sera from patients that did and did not develop AKI. CONCLUSIONS: The lack of correlation between serum and urine activity levels suggests that the rapid intraoperative increases in PLA2G15/LPLA2 activity may originate from the kidney and as such offer an intraoperative indicator of early renal response to CPB associated stressors.


Acute Kidney Injury/enzymology , Acyltransferases/blood , Acyltransferases/urine , Cardiopulmonary Bypass , Phospholipases A2/blood , Phospholipases A2/urine , Acute Kidney Injury/etiology , Acute Kidney Injury/pathology , Aged , Biomarkers/blood , Biomarkers/urine , Cardiopulmonary Bypass/adverse effects , Case-Control Studies , Cohort Studies , Female , Humans , Intraoperative Period , Male , Middle Aged , Postoperative Complications/enzymology , Postoperative Complications/etiology , Postoperative Complications/pathology , Postoperative Period , Risk Factors
19.
Am J Physiol Renal Physiol ; 320(1): F61-F73, 2021 01 01.
Article En | MEDLINE | ID: mdl-33196323

Oxidative stress is a key concept in basic, translational, and clinical research to understand the pathophysiology of various disorders, including cardiovascular and renal diseases. Although attempts to directly reduce oxidative stress with redox-active substances have until now largely failed to prove clinical benefit, indirect approaches to combat oxidative stress enzymatically have gained further attention as potential therapeutic strategies. The pantetheinase Vanin-1 is expressed on kidney proximal tubular cells, and its reaction product cysteamine is described to negatively affect redox homeostasis by inhibiting the replenishment of cellular antioxidative glutathione stores. Vanin-1-deficient mice were shown to be protected against oxidative stress damage. The aim of this study was to elucidate whether pharmacological inhibition of Vanin-1 protects mice from oxidative stress-related acute or chronic kidney injury as well. By studying renal ischemia-reperfusion injury in Col4α3-/- (Alport syndrome) mice and in vitro hypoxia-reoxygenation in human proximal tubular cells we found that treatment with a selective and potent Vanin-1 inhibitor resulted in ample inhibition of enzymatic activity in vitro and in vivo. However, surrogate parameters of metabolic and redox homeostasis were only partially and insufficiently affected. Consequently, apoptosis and reactive oxygen species level in tubular cells as well as overall kidney function and fibrotic processes were not improved by Vanin-1 inhibition. We thus conclude that Vanin-1 functionality in the context of cardiovascular diseases needs further investigation and the biological relevance of pharmacological Vanin-1 inhibition for the treatment of kidney diseases remains to be proven.


Acute Kidney Injury/prevention & control , Amidohydrolases/antagonists & inhibitors , Enzyme Inhibitors/pharmacology , Kidney Tubules, Proximal/drug effects , Nephritis, Hereditary/prevention & control , Oxidative Stress/drug effects , Renal Insufficiency, Chronic/prevention & control , Reperfusion Injury/prevention & control , Acute Kidney Injury/enzymology , Acute Kidney Injury/genetics , Acute Kidney Injury/pathology , Amidohydrolases/genetics , Amidohydrolases/metabolism , Animals , Apoptosis/drug effects , Autoantigens/genetics , Autoantigens/metabolism , Cell Line , Collagen Type IV/genetics , Collagen Type IV/metabolism , Disease Models, Animal , Enzyme Inhibitors/pharmacokinetics , Fibrosis , GPI-Linked Proteins/antagonists & inhibitors , GPI-Linked Proteins/genetics , GPI-Linked Proteins/metabolism , Humans , Kidney Tubules, Proximal/enzymology , Kidney Tubules, Proximal/pathology , Male , Mice, Inbred C57BL , Mice, Knockout , Nephritis, Hereditary/enzymology , Nephritis, Hereditary/genetics , Nephritis, Hereditary/pathology , Renal Insufficiency, Chronic/enzymology , Renal Insufficiency, Chronic/genetics , Renal Insufficiency, Chronic/pathology , Reperfusion Injury/enzymology , Reperfusion Injury/genetics , Reperfusion Injury/pathology
20.
Am J Physiol Renal Physiol ; 319(6): F1054-F1066, 2020 12 01.
Article En | MEDLINE | ID: mdl-33135478

In the present study, we demonstrated the marked activity of SW033291, an inhibitor of 15-hydoxyprostaglandin dehydrogenase (15-PGDH), in preventing acute kidney injury (AKI) in a murine model of ischemia-reperfusion injury. AKI due to ischemic injury represents a significant clinical problem. PGE2 is vasodilatory in the kidney, but it is rapidly degraded in vivo due to catabolism by 15-PGDH. We investigated the potential of SW033291, a potent and specific 15-PGDH inhibitor, as prophylactic treatment for ischemic AKI. Prophylactic administration of SW033291 significantly increased renal tissue PGE2 levels and increased post-AKI renal blood flow and renal arteriole area. In parallel, prophylactic SW033291 decreased post-AKI renal morphology injury scores and tubular apoptosis and markedly reduced biomarkers of renal injury that included blood urea nitrogen, creatinine, neutrophil gelatinase-associated lipocalin, and kidney injury molecule-1. Prophylactic SW033291 also reduced post-AKI induction of proinflammatory cytokines, high-mobility group box 1, and malondialdehyde. Protective effects of SW033291 were mediated by PGE2 signaling, as they could be blocked by pharmacological inhibition of PGE2 synthesis. Consistent with activation of PGE2 signaling, SW033291 induced renal levels of both EP4 receptors and cAMP, along with other vasodilatory effectors, including AMP, adenosine, and the adenosine A2A receptor. The protective effects of SW0333291 could largely be achieved with a single prophylactic dose of the drug. Inhibition of 15-PGDH may thus represent a novel strategy for prophylaxis of ischemic AKI in multiple clinical settings, including renal transplantation and cardiovascular surgery.


Acute Kidney Injury/prevention & control , Adenosine/metabolism , Dinoprostone/metabolism , Enzyme Inhibitors/pharmacology , Hydroxyprostaglandin Dehydrogenases/antagonists & inhibitors , Kidney/blood supply , Kidney/drug effects , Pyridines/pharmacology , Receptor, Adenosine A2A/metabolism , Receptors, Prostaglandin E, EP4 Subtype/metabolism , Renal Circulation/drug effects , Reperfusion Injury/prevention & control , Thiophenes/pharmacology , Vasodilation/drug effects , Acute Kidney Injury/enzymology , Acute Kidney Injury/pathology , Acute Kidney Injury/physiopathology , Animals , Blood Flow Velocity , Disease Models, Animal , Hydroxyprostaglandin Dehydrogenases/metabolism , Kidney/enzymology , Kidney/pathology , Male , Mice, Inbred C57BL , Reperfusion Injury/enzymology , Reperfusion Injury/pathology , Reperfusion Injury/physiopathology , Signal Transduction
...