Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 27
1.
Int J Biol Sci ; 19(16): 5145-5159, 2023.
Article En | MEDLINE | ID: mdl-37928264

Mitochondrial dysfunction plays a pivotal role in diabetic kidney disease initiation and progression. PTEN-induced serine/threonine kinase 1 (PINK1) is a core organizer of mitochondrial quality control; however, its function in diabetic kidney disease remains controversial. Here, we aimed to investigate the pathophysiological roles of PINK1 in diabetic tubulopathy, focusing on its effects on mitochondrial homeostasis and tubular cell necroptosis, which is a specialized form of regulated cell death. PINK1-knockout mice showed more severe diabetes-induced tubular injury, interstitial fibrosis, and albuminuria. The expression of profibrotic cytokines significantly increased in the kidneys of diabetic Pink1-/- mice, which eventually culminated in aggravated interstitial fibrosis. Additionally, the knockdown of PINK1 in HKC-8 cells upregulated the fibrosis-associated proteins, and these effects were rescued by PINK1 overexpression. PINK1 deficiency was also associated with exaggerated hyperglycemia-induced mitochondrial dysfunction and defective mitophagic activity, whereas PINK1 overexpression ameliorated these negative effects and restored mitochondrial homeostasis. Mitochondrial reactive oxygen species triggered tubular cell necroptosis under hyperglycemic conditions, which was aggravated by PINK1 deficiency and improved by its overexpression. In conclusion, PINK1 plays a pivotal role in suppressing mitochondrial dysfunction and tubular cell necroptosis under high glucose conditions and exerts protective effects in diabetic kidney disease.


Diabetes Mellitus , Diabetic Nephropathies , Mice , Animals , Diabetic Nephropathies/genetics , Diabetic Nephropathies/metabolism , Necroptosis/genetics , Protein Kinases/genetics , Protein Kinases/metabolism , Mitochondria/metabolism , Mice, Knockout , Fibrosis , Diabetes Mellitus/metabolism
2.
ACS Omega ; 7(48): 43492-43498, 2022 Dec 06.
Article En | MEDLINE | ID: mdl-36506148

The adsorption of peptides and proteins on hydrophobic solid surfaces has received considerable research attention owing to their wide applications to biocompatible nanomaterials and nanodevices, such as biosensors and cell adhesion materials with reduced nanomaterial toxicity. However, fundamental understandings about physicochemical hydrophobic interactions between peptides and hydrophobic solid surfaces are still unknown. In this study, we investigate the effect of secondary structures on adsorption energies between peptides and hydrophobic solid surfaces via experimental and theoretical analyses using surface-assisted laser desorption/ionization-time-of-flight (SALDI-TOF) and molecular dynamics (MD) simulations. The hydrophobic interactions between peptides and hydrophobic solid surfaces measured via SALDI-TOF and MD simulations indicate that the hydrophobic interaction of peptides with random coil structures increased more than that of peptides with an α-helix structure when polar amino acids are replaced with hydrophobic amino acids. Additionally, our study sheds new light on the fundamental understanding of the hydrophobic interaction between hydrophobic solid surfaces and peptides that have diverse secondary structures.

3.
FASEB J ; 36(5): e22272, 2022 05.
Article En | MEDLINE | ID: mdl-35436011

The transition of fibroblasts into myofibroblasts is a crucial step in kidney fibrosis. However, the biological processes involved in this transdifferentiation are incompletely understood. In this study, we discovered that the midbody plays a role in the fibroblast-myofibroblast transition by mediating TGF-ß/Smad signaling. Combining bulk RNA-seq, histology, and the western blot of unilateral ureteral obstruction kidneys, we demonstrated that the pathway related to microtubules is implicated in kidney fibrosis, and the blocking of microtubule dynamics by colchicine improved kidney fibrosis. Subsequently, to explore microtubule-based organelles in detail, we cultured NRK-49F (rat kidney fibroblast cell line) and HKC-8 (human proximal tubule cell line) under transforming growth factor-ß1 (TGF-ß1) stimulation, which caused deciliation in both cell lines during epithelial-mesenchymal and fibroblast-myofibroblast transition. We identified another microtubule-based organelle, the midbody, whose formation is promoted by TGF-ß1 in fibroblasts as a result of proliferation in contrast to tubular cells. Notably, TGF-ß receptors were present in the midbody of both cell lines. In TGF-ß1-treated fibroblasts, colchicine or Hedgehog pathway inhibitor 4 impaired the midbody formation, and attenuated the upregulation of canonical TGF-ß/Smad signaling and α-SMA expression. These findings offer novel insight into the midbody as an active organelle involved in fibroblast-myofibroblast transition by mediating TGF-ß/Smad signaling, which could be a potential therapeutic target.


Kidney Diseases , Myofibroblasts , Animals , Colchicine/pharmacology , Epithelial-Mesenchymal Transition , Female , Fibroblasts/metabolism , Fibrosis , Hedgehog Proteins/metabolism , Humans , Kidney Diseases/pathology , Male , Myofibroblasts/metabolism , Rats , Signal Transduction/physiology , Transforming Growth Factor beta1/metabolism
4.
Front Immunol ; 12: 656632, 2021.
Article En | MEDLINE | ID: mdl-34177898

Urine has been regarded as a good resource based on the assumption that urine can directly reflect the state of the allograft or ongoing injury in kidney transplantation. Previous studies, suggesting the usefulness of urinary mRNA as a biomarker of acute rejection, imply that urinary mRNA mirrors the transcriptional activity of the kidneys. We selected 14 data-driven candidate genes through a meta-analysis and measured the candidate genes using quantitative PCR without pre-amplification in the cross-sectional specimens from Korean kidney transplant patients. Expression of 9/14 genes (CXCL9, CD3ϵ, IP-10, LCK, C1QB, PSMB9, Tim-3, Foxp3, and FAM26F) was significantly different between acute rejection and stable graft function with normal pathology and long-term graft survival in 103 training samples. CXCL9 was also distinctly expressed in allografts with acute rejection in in situ hybridization analysis. This result, consistent with the qPCR result, implies that urinary mRNA could reflect the magnitude of allograft injury. We developed an AR prediction model with the urinary mRNAs by a binary logistic regression and the AUC of the model was 0.89 in the training set. The model was validated in 391 independent samples, and the AUC value yielded 0.84 with a fixed manner. In addition, the decision curve analysis indicated a range of reasonable threshold probabilities for biopsy. Therefore, we suggest the urine mRNA signature could be used as a non-invasive monitoring tool of acute rejection for clinical application and could help determine whether to perform a biopsy in a recipient with increased creatinine.


Allografts/immunology , Biomarkers , Graft Rejection/diagnosis , Graft Rejection/etiology , Kidney Transplantation/adverse effects , Liquid Biopsy/methods , RNA, Messenger/genetics , Acute Disease , Adult , Biomarkers/urine , Cell-Free Nucleic Acids , Female , Humans , Immunohistochemistry , Kidney Transplantation/methods , Male , Middle Aged , ROC Curve , Reproducibility of Results
5.
Int J Mol Sci ; 22(4)2021 Feb 05.
Article En | MEDLINE | ID: mdl-33562802

Renal ischemia-reperfusion injury (IRI) is involved in the majority of clinical conditions that manifest as renal function deterioration; however, specific treatment for this type of injury is still far from clinical use. Since Toll-like receptor (TLR)-mediated signaling is a key mediator of IRI, we examined the effect of a multiple-TLR-blocking peptide named TLR-inhibitory peptide 1 (TIP1), which exerts the strongest action on TLR4, on renal IRI. We subjected C57BL/6 mice to 23 min of renal pedicle clamping preceded by intraperitoneal injection with a vehicle or TIP1. Sham control mice underwent flank incision only. Mouse kidneys were harvested after 24 h of reperfusion for histology, western blot, RT-PCR, and flow cytometry analysis. Pretreatment with TIP1 lowered the magnitude of elevated plasma creatinine levels and attenuated tubular injury. TIP1 treatment also reduced mRNA expression of inflammatory cytokines and decreased apoptotic cells and oxidative stress in post-ischemic kidneys. In kidneys pretreated with TIP1, the infiltration of macrophages and T helper 17 cells was less abundant than those in the IRI only group. These results suggest that TIP1 has a potential beneficial effect in attenuating the degree of kidney damage induced by IRI.


Acute Kidney Injury/prevention & control , Cell-Penetrating Peptides/administration & dosage , Reperfusion Injury/prevention & control , Signal Transduction/drug effects , Acute Kidney Injury/genetics , Acute Kidney Injury/metabolism , Animals , Cell-Penetrating Peptides/pharmacology , Creatinine/blood , Cytokines/genetics , Disease Models, Animal , Gene Expression Regulation/drug effects , Male , Mice , Mice, Inbred C57BL , Oxidative Stress/drug effects , Reperfusion Injury/genetics , Reperfusion Injury/metabolism , Toll-Like Receptors/genetics , Toll-Like Receptors/metabolism
7.
Int J Mol Sci ; 21(22)2020 Nov 13.
Article En | MEDLINE | ID: mdl-33202867

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.


Acute Kidney Injury , Kidney Tubules , Lipid Peroxidation , Mitochondria , NLR Family, Pyrin Domain-Containing 3 Protein/deficiency , Rhabdomyolysis , Acute Kidney Injury/etiology , Acute Kidney Injury/genetics , Acute Kidney Injury/metabolism , Acute Kidney Injury/pathology , Animals , Kidney Tubules/metabolism , Kidney Tubules/pathology , Mice , Mice, Knockout , Mitochondria/genetics , Mitochondria/metabolism , Mitochondria/pathology , Mitochondrial Dynamics/genetics , Myoglobin/genetics , Myoglobin/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Reactive Oxygen Species/metabolism , Rhabdomyolysis/complications , Rhabdomyolysis/genetics , Rhabdomyolysis/metabolism , Rhabdomyolysis/pathology
8.
J Korean Med Sci ; 35(36): e305, 2020 Sep 14.
Article En | MEDLINE | ID: mdl-32924342

BACKGROUND: Oxidative stress induced by chronic hyperglycemia is recognized as a significant mechanistic contributor to the development of diabetic kidney disease (DKD). Nonphagocytic nicotinamide adenine dinucleotide phosphate oxidase 4 (Nox4) is a major source of reactive oxygen species (ROS) in many cell types and in the kidney tissue of diabetic animals. We designed this study to explore the therapeutic potential of chloroquine (CQ) and amodiaquine (AQ) for inhibiting mitochondrial Nox4 and diabetic tubular injury. METHODS: Human renal proximal tubular epithelial cells (hRPTCs) were cultured in high-glucose media (30 mM D-glucose), and diabetes was induced with streptozotocin (STZ, 50 mg/kg i.p. for 5 days) in male C57BL/6J mice. CQ and AQ were administered to the mice via intraperitoneal injection for 14 weeks. RESULTS: CQ and AQ inhibited mitochondrial Nox4 and increased mitochondrial mass in hRPTCs under high-glucose conditions. Reduced mitochondrial ROS production after treatment with the drugs resulted in decreased endoplasmic reticulum (ER) stress, suppressed inflammatory protein expression and reduced cell apoptosis in hRPTCs under high-glucose conditions. Notably, CQ and AQ treatment diminished Nox4 activation and ER stress in the kidneys of STZ-induced diabetic mice. In addition, we observed attenuated inflammatory protein expression and albuminuria in STZ-induced diabetic mice after CQ and AQ treatment. CONCLUSION: We substantiated the protective actions of CQ and AQ in diabetic tubulopathy associated with reduced mitochondrial Nox4 activation and ER stress alleviation. Further studies exploring the roles of mitochondrial Nox4 in the pathogenesis of DKD could suggest new therapeutic targets for patients with DKD.


Amodiaquine/pharmacology , Chloroquine/pharmacology , Endoplasmic Reticulum Stress/drug effects , Mitochondria/metabolism , NADPH Oxidase 4/metabolism , Amodiaquine/chemistry , Amodiaquine/metabolism , Amodiaquine/therapeutic use , Animals , Apoptosis/drug effects , Cells, Cultured , Chloroquine/chemistry , Chloroquine/metabolism , Chloroquine/therapeutic use , Diabetes Mellitus, Experimental/chemically induced , Diabetes Mellitus, Experimental/drug therapy , Diabetes Mellitus, Experimental/pathology , Diabetes Mellitus, Type 1/pathology , Glucose/pharmacology , Humans , Kidney Tubules, Proximal/cytology , Kidney Tubules, Proximal/metabolism , Male , Mice , Mice, Inbred C57BL , NADPH Oxidase 4/antagonists & inhibitors , Reactive Oxygen Species/metabolism
9.
J Clin Med ; 9(8)2020 Aug 12.
Article En | MEDLINE | ID: mdl-32806730

Approximately 5% of patients with IgA nephropathy (IgAN) exhibit mild mesangial lesions with acute onset nephrotic syndrome and diffuse foot process effacement representative of minimal change disease (MCD). It is not clear whether these unusual cases of IgAN with MCD (IgAN-MCD) are variant types of IgAN or coincidental deposition of IgA in patients with MCD. In a retrospective multicenter cohort study of 18 hospitals in Korea, we analyzed 46 patients with IgAN-MCD. Patients with endocapillary proliferation, segmental sclerosis, and crescent were excluded, and the clinical features and prognosis of IgAN-MCD were compared with those of pure MCD. In addition, we performed galactose-deficient IgA1 (KM55) staining to characterize IgAN-MCD. Among the 21,697 patients with glomerulonephritis enrolled in the database, 46 patients (0.21%) were diagnosed with IgAN-MCD, and 1610 patients (7.4%) with pure MCD. The 46 patients with IgAN-MCD accounted for 0.6% of primary IgAN patients (n = 7584). There was no difference in prognosis between patients with IgAN-MCD and those with only MCD. IgA and KM55 showed double positivity in all patients with IgAN-MCD (n = 4) or primary IgAN (n = 5) under double immunofluorescent staining. However, in four patients with lupus nephritis, mesangial IgA was deposited, but galactose-deficient-IgA1 (Gd-IgA1) was not. These findings suggest that IgAN-MCD is a dual glomerulopathy in which MCD was superimposed on possibly indolent IgAN. We confirmed by KM55 staining that IgAN-MCD is true IgAN, enabling better characterizations of the disease. Furthermore, IgAN-MCD shows a good prognosis when treated according to the usual MCD treatment modality.

10.
Am J Physiol Renal Physiol ; 317(4): F767-F780, 2019 10 01.
Article En | MEDLINE | ID: mdl-31390268

We examined the effects of empagliflozin, a selective inhibitor of Na+-glucose cotransporter 2, on mitochondrial quality control and autophagy in renal tubular cells in a diabetic environment in vivo and in vitro. Human renal proximal tubular cells (hRPTCs) were incubated under high-glucose conditions. Diabetes was induced with streptozotocin in male C57BL/6J mice. Improvements in mitochondrial biogenesis and balanced fusion-fission protein expression were noted in hRPTCs after treatment with empagliflozin in high-glucose media. Empagliflozin also increased autophagic activities in renal tubular cells in the high-glucose environment, which was accompanied with mammalian target of rapamycin inhibition. Moreover, reduced mitochondrial reactive oxygen species production and decreased apoptotic and fibrotic protein expression were observed in hRPTCs after treatment with empagliflozin, even in the hyperglycemic circumstance. Importantly, empagliflozin restored AMP-activated protein kinase-α phosphorylation and normalized levels of AMP-to-ATP ratios in hRPTCs subjected to a high-glucose environment, which suggests the way that empagliflozin is involved in mitochondrial quality control. Empagliflozin effectively suppressed Na+-glucose cotransporter 2 expression and ameliorated renal morphological changes in the kidneys of streptozotocin-induced diabetic mice. Electron microscopy analysis showed that mitochondrial fragmentation was decreased and 8-hydroxy-2'-deoxyguanosine content was low in renal tubular cells of empagliflozin treatment groups compared with those of the diabetic control group. We suggest one mechanism related to the renoprotective actions of empagliflozin, which reverse mitochondrial dynamics and autophagy.


Autophagy/drug effects , Benzhydryl Compounds/therapeutic use , Diabetic Nephropathies/drug therapy , Diabetic Nephropathies/pathology , Glucosides/therapeutic use , Mitochondria/drug effects , Mitochondria/pathology , Sodium-Glucose Transporter 2 Inhibitors/therapeutic use , Adenosine Monophosphate/metabolism , Adenosine Triphosphate/metabolism , Animals , Apoptosis Regulatory Proteins/antagonists & inhibitors , Cell Line , Diabetes Mellitus, Experimental/drug therapy , Diabetes Mellitus, Experimental/genetics , Diabetes Mellitus, Experimental/pathology , Gene Expression/drug effects , Humans , Kidney Tubules, Proximal/drug effects , Kidney Tubules, Proximal/pathology , Male , Mice , Mice, Inbred C57BL , Organelle Biogenesis , Reactive Oxygen Species/metabolism , Sodium-Glucose Transporter 2/metabolism
11.
Am J Physiol Renal Physiol ; 317(3): F670-F682, 2019 09 01.
Article En | MEDLINE | ID: mdl-31339773

The intratubular renin-angiotensin system (RAS) is thought to play an essential role in hypertensive renal disease, but information regarding sex-related differences in this system is limited. The present study investigated sex differences in the intratubular RAS in two-kidney, one-clip (2K1C) rats. A 2.5-mm clip was placed on the left renal artery of Sprague-Dawley rats, and rats were euthanized 3 or 5 wk after the operation. Systolic blood pressure increased in 2K1C rats in both sexes but was significantly higher in male rats than in female rats, and an antihypertensive effect was not observed in 2K1C ovariectomized (OVX) female rats. Compared with male 2K1C rats, intratubular angiotensin-converting enzyme (ACE) and ANG II were repressed, and intratubular ACE2, angiotensin (1-7), and Mas receptor were increased in both kidneys in female 2K1C rats 5 wk after surgery. Comparison with male and female rats and intratubular mRNA levels of ACE and ANG II type 1 receptor were augmented in OVX female rats, regardless of the clipping surgery 3 wk postoperation. ANG II type 2 receptor was upregulated in female rats with or without OVX; thus, the ANG II type 1-to-type 2 receptor ratio was higher in male rats than in female rats. In conclusion, female rats were protected from hypertensive renal and cardiac injury after renal artery clipping. An increase in the intratubular nonclassic RAS [ACE2/angiotensin (1-7)/Mas receptor] and a decrease in the ANG II type 1-to-type 2 receptor ratio could limit the adverse effects of the classic RAS during renovascular hypertension in female rats, and estrogen is suggested to play a primary role in the regulation of intratubular RAS components.


Blood Pressure , Estrogens/metabolism , Hypertension/metabolism , Kidney Tubules/metabolism , Renal Artery/surgery , Renin-Angiotensin System , Angiotensin I/metabolism , Angiotensin II/metabolism , Angiotensin-Converting Enzyme 2 , Animals , Constriction , Disease Models, Animal , Female , Hypertension/etiology , Hypertension/genetics , Hypertension/physiopathology , Hypertrophy, Left Ventricular/etiology , Hypertrophy, Left Ventricular/metabolism , Hypertrophy, Left Ventricular/physiopathology , Kidney Tubules/physiopathology , Macrophages/metabolism , Male , Ovariectomy , Peptide Fragments/metabolism , Peptidyl-Dipeptidase A/genetics , Peptidyl-Dipeptidase A/metabolism , Proto-Oncogene Mas , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , Rats, Sprague-Dawley , Receptor, Angiotensin, Type 1/genetics , Receptor, Angiotensin, Type 1/metabolism , Receptor, Angiotensin, Type 2/metabolism , Receptors, G-Protein-Coupled/genetics , Receptors, G-Protein-Coupled/metabolism , Renal Artery/physiopathology , Sex Factors , Signal Transduction
12.
J Proteome Res ; 18(7): 2803-2812, 2019 07 05.
Article En | MEDLINE | ID: mdl-31244212

During aging, the kidney undergoes functional and physiological changes that are closely affiliated with chronic kidney disease (CKD). There is increasing evidence supporting the role of lipid or lipid-derived mediators in the pathogenesis of CKD and other aging-related diseases. To understand the role of lipids in various metabolic processes during kidney aging, we conducted matrix-assisted laser desorption/ionization-imaging mass spectrometry (MALDI-IMS) analysis in kidneys harvested from young (2 months old, n = 3) and old mice (24 months old, n = 3). MALDI-IMS analysis showed an increase in ceramide level and a decrease in sphingomyelin (SM) and phosphatidylcholine (PC) levels in kidneys of old mice. The increased expression of cPLA2 and SMPD1 protein in aged kidney was confirmed by immunohistochemistry and Western blot analysis. Our MALDI-IMS data showed the altered distribution of lipids in aged kidney as indicative of aging-related functional changes of the kidney. Combined analysis of MALDI-IMS and IHC confirmed lipidomic changes and expression levels of responsible enzymes as well as morphological changes.


Aging , Kidney/chemistry , Lipidomics/methods , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization/methods , Animals , Ceramides/metabolism , Immunohistochemistry , Kidney/diagnostic imaging , Mice , Phosphatidylcholines/metabolism , Phospholipases A2/metabolism , Sphingomyelin Phosphodiesterase/metabolism , Sphingomyelins/metabolism
13.
Sci Rep ; 9(1): 1854, 2019 02 12.
Article En | MEDLINE | ID: mdl-30755622

Endocan is a water-soluble proteoglycan exclusively secreted by vascular endothelium. Endocan levels may be elevated in kidney transplant recipients experiencing antibody-mediated rejection (ABMR), which is characterized by vascular inflammation in transplanted kidney. We evaluated the clinical relevance of endocan as markers of microvascular inflammation in patients who underwent kidney transplantation. Plasma and urinary endocan levels were measured in 203 kidney transplant recipients and were compared across different etiologies of allograft dysfunction and various pathologic scores. Both plasma and urinary endocan levels were significantly higher in patients with acute ABMR than those in patients with normal pathology, acute tubular necrosis (ATN), acute pyelonephritis, BK virus associated nephropathy (BKVN), and T-cell mediated rejection (TCMR). Patients with chronic active ABMR also exhibited significantly higher plasma and urinary endocan levels than patients with long-term graft survival. Scores of glomerulitis and peritubular capillaritis, which are typical features of microvascular inflammation, were significantly elevated in patients with higher plasma and/or urinary endocan levels. Furthermore, plasma and urinary endocan levels could effectively discriminate ABMR from ATN, BKVN, and TCMR. Finally, patients exhibiting high urinary and plasma endocan levels in acute ABMR group showed significantly worse renal survival. Altogether, plasma and urinary endocan levels may serve as potential markers of microvascular inflammation in kidney transplant recipients.


Inflammation/immunology , Kidney Failure, Chronic/surgery , Kidney Transplantation , Microcirculation/immunology , Neoplasm Proteins/blood , Neoplasm Proteins/urine , Proteoglycans/blood , Proteoglycans/urine , Adult , Area Under Curve , Biopsy , Female , Graft Rejection , Humans , Male , Middle Aged , Necrosis , Polyomavirus Infections/metabolism , Pyelonephritis/immunology , ROC Curve , Retrospective Studies , T-Lymphocytes/cytology , Transplant Recipients , Treatment Outcome , Tumor Virus Infections/immunology
14.
Free Radic Biol Med ; 131: 40-49, 2019 02 01.
Article En | MEDLINE | ID: mdl-30476538

Binding of epidermal growth factor (EGF) to its cell surface receptor induces production of H2O2, which serves as an intracellular messenger. We have shown that exogenous H2O2 reversibly inactivates the phosphatidylinositol 4-phosphate [PtdIns(4)P] phosphatase Sac1 (suppressor of actin 1) at the Golgi complex of mammalian cells by oxidizing its catalytic cysteine residue and thereby increases both the amount of Golgi PtdIns(4)P and the rate of protein secretion. Here we investigated the effects of EGF on Sac1 oxidation and PtdIns(4)P abundance at the Golgi in A431 cells. EGF induced a transient increase in Golgi PtdIns(4)P as well as a transient oxidation of Sac1 in a manner dependent on elevation of the intracellular Ca2+ concentration and on H2O2. Oxidation of Sac1 occurred at the Golgi, as revealed with the use of the Golgi-confined Sac1-K2A mutant. Knockdown of Duox enzymes implicated these Ca2+-dependent members of the NADPH oxidase family as the major source of H2O2 for Sac1 oxidation. Expression of a Golgi-targeted H2O2 probe revealed transient EGF-induced H2O2 production at this organelle. Our findings have thus uncovered a previously unrecognized EGF signaling pathway that links intracellular Ca2+ mobilization to events at the Golgi including Duox activation, H2O2 production, Sac1 oxidation, and PtdIns(4)P accumulation.


Calcium/metabolism , Golgi Apparatus/drug effects , Hydrogen Peroxide/metabolism , Membrane Proteins/genetics , Phosphatidylinositol Phosphates/metabolism , Cell Line, Tumor , Dual Oxidases/antagonists & inhibitors , Dual Oxidases/genetics , Dual Oxidases/metabolism , Epithelial Cells/cytology , Epithelial Cells/drug effects , Epithelial Cells/metabolism , ErbB Receptors/pharmacology , Gene Expression Regulation , Golgi Apparatus/metabolism , Humans , Membrane Proteins/antagonists & inhibitors , Membrane Proteins/metabolism , Oxidation-Reduction , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Signal Transduction
15.
Am J Nephrol ; 48(6): 456-464, 2018.
Article En | MEDLINE | ID: mdl-30472702

BACKGROUND: Peritoneal fibrosis is a devastating complication of peritoneal dialysis. However, its precise mechanism is unclear, and specific treatments have not yet been established. Recent evidence suggests that the sonic hedgehog (SHH) signaling pathway is involved in tissue fibrogenesis. Drugs that inhibit this pathway are emerging in the field of anti-fibrosis therapy. Itraconazole, an anti-fungal agent, was also recently recognized as an inhibitor of the SHH signaling pathway. In this study, we used a mouse model to investigate whether the SHH signaling pathway is involved in the development of peritoneal fibrosis and the effects of itraconazole on peritoneal fibrosis. METHODS: Peritoneal fibrosis was induced by intraperitoneal (IP) injection of 0.1% chlorhexidine gluconate (CG) solution every other day for 4 weeks, with or without itraconazole treatment (20 mg/kg, IP injection on a daily basis). Male C57BL/6 mice were divided into 4 groups: saline group, saline plus itraconazole group, CG group, and CG plus itraconazole group. Isotonic saline was administered intraperitoneally to the control group. The peritoneal tissues were evaluated for histological changes, expression of fibrosis markers, and the main components of the SHH signaling pathway. RESULTS: Peritoneal thickening was evident in the CG group and was significantly decreased by itraconazole administration (80.4 ± 7.7 vs. 28.2 ± 3.8 µm, p < 0.001). The expression of the following SHH signaling pathway components was upregulated in the CG group and suppressed by itraconazole treatment: SHH, patched, smoothened, and glioma-associated oncogene transcription factor 1. The IP injection of CG solution increased the expression of fibrosis markers such as α-smooth muscle actin and transforming growth factor-ß1 in the peritoneal tissues. Itraconazole treatment significantly decreased the expression of these markers. CONCLUSION: Our study provides the first evidence that the SHH signaling pathway may be implicated in peritoneal fibrosis. It also demonstrates that itraconazole treatment has protective effects on peritoneal fibrosis through the regulation of the SHH signaling pathway. These findings suggest that blockage of the SHH signaling pathway is a potential therapeutic strategy for peritoneal fibrosis.


Hedgehog Proteins/metabolism , Itraconazole/pharmacology , Peritoneal Dialysis/adverse effects , Peritoneal Fibrosis/drug therapy , Signal Transduction/drug effects , Animals , Chlorhexidine/administration & dosage , Chlorhexidine/analogs & derivatives , Chlorhexidine/toxicity , Disease Models, Animal , Humans , Injections, Intraperitoneal , Itraconazole/therapeutic use , Male , Mice , Mice, Inbred C57BL , Peritoneal Fibrosis/chemically induced , Peritoneal Fibrosis/pathology , Peritoneum/drug effects , Peritoneum/pathology , Treatment Outcome
16.
Front Immunol ; 9: 2563, 2018.
Article En | MEDLINE | ID: mdl-30483252

The NOD-like receptor family, pyrin domain containing-3 (NLRP3) inflammasome has been implicated in renal inflammation and fibrosis. However, the biological function of inflammasome-independent NLRP3 in non-immune cells is still unclear. We evaluated the role of inflammasome-independent NLRP3 in renal tubular cells and assessed the value of NLRP3 as a therapeutic target for acute kidney injury (AKI). Various renal tubular cell lines and primary cultured tubular cells from NLRP3 knockout (KO) mice were used for in vitro studies. We also tested the role of tubular NLRP3 in AKI with a unilateral ureter obstruction model (UUO). Hypoxia induced significant increase of NLRP3 independent of ASC, caspase-1, and IL-1ß. NLRP3 in renal tubular cells relocalized from the cytosol to the mitochondria during hypoxia and bound to mitochondrial antiviral signal protein (MAVS). The deletion of NLRP3 or MAVS in renal tubular cells attenuated mitochondrial reactive oxygen species (ROS) production and depolarization of the mitochondrial membrane potentials under hypoxia. In response to UUO, NLRP3 KO mice showed less fibrosis, apoptosis, and ROS injury than wild type (WT) mice. Compared with WT kidney, mitophagy was up-regulated in NLRP3 KO kidney relative to the baseline and it was protective against AKI. Our results indicate that inflammasome-independent NLRP3 in renal tubular cells plays important role in mitochondrial ROS production and injury by binding to MAVS after hypoxic injury. This mitochondrial regulation in the absence of NLRP3 increases autophagy and attenuates apoptosis after UUO. We suggest that inflammasome-independent NLRP3 could be a therapeutic target of AKI to prevent the progression of chronic kidney disease.


Acute Kidney Injury/metabolism , Adaptor Proteins, Signal Transducing/metabolism , Hypoxia/metabolism , Kidney Tubules/metabolism , Mitochondria/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Acute Kidney Injury/immunology , Adaptor Proteins, Signal Transducing/genetics , Animals , Apoptosis , Autophagy , Cells, Cultured , Disease Models, Animal , Humans , Hypoxia/immunology , Inflammasomes/metabolism , Kidney Tubules/pathology , Membrane Potential, Mitochondrial , Mice , Mice, Knockout , NLR Family, Pyrin Domain-Containing 3 Protein/genetics , Reactive Oxygen Species/metabolism , Ureter/surgery
17.
Biomed Res Int ; 2018: 3421568, 2018.
Article En | MEDLINE | ID: mdl-30225249

Thymosin ß4 (Tß4) treatment was known to show the potential therapeutic effects on diabetic complications. This study was performed to determine if Tß4 expression is changed in both serum and tissues under diabetic conditions and can be a serum biomarker. Type 1 diabetic mice were induced in C57/BL6J mice by intraperitoneal injection of streptozotocin (STZ) at a dose of 50 mg/kg body weight. The mice were sacrificed at 16 weeks after STZ injection. Tissues and plasmas were obtained to determine the expression levels of Tß4 using ELISA, real time RT-PCR, and immunohistochemistry. The average serum glucose level was increased to approximately 400 mg/dL beginning 2 weeks after the five injections of STZ and lasting for at least 13 weeks until sacrifice. The plasma and tissue levels of Tß4 in the age-matched control mice were not significantly different from those of the diabetic mice. In conclusion, the Tß4 expression level in the plasmas and tissues of diabetic mice was not affected by diabetic conditions. It indirectly suggests that the therapeutic effect of Tß4 on diabetic complications is due to its regenerative effects on damaged tissue but not to the changed expression level of Tß4 in plasma and tissues of diabetes.


Diabetes Mellitus, Experimental/blood , Thymosin/metabolism , Animals , Injections, Intraperitoneal , Mice , Streptozocin , Thymosin/therapeutic use
18.
Sci Rep ; 8(1): 8774, 2018 06 08.
Article En | MEDLINE | ID: mdl-29884802

We investigated the effects of chloroquine (CQ) and amodiaquine (AQ) on AMPK phosphorylation in renal tubular cells in a diabetic environment in vivo and in vitro. We also examined whether CQ- or AQ-mediated AMPK activity restoration attenuated diabetic tubulopathy by normalizing mitochondrial fragmentation. Human renal proximal epithelial cells (HKC8) were incubated in high-glucose conditions. Diabetes was induced with streptozotocin in male C57/BL6J mice. Treatment with CQ or AQ abolished high-glucose-induced phospho-AMPK and phosph-PGC1α down-regulation in HKC8 cells. Improvements in functional mitochondrial mass and balanced fusion/fission protein expression were observed in HKC8 cells after treatment with CQ or AQ in high-glucose conditions. Moreover, decreased mitochondrial ROS production and reduced apoptotic and fibrotic protein expression were noted in HKC8 cells after treatment with CQ or AQ, even in high-glucose conditions. CQ and AQ treatment effectively mitigated albuminuria and renal histopathologic changes and increased AMPK activity in the kidneys of diabetic mice. Electron microscopy analysis showed that mitochondrial fragmentation was decreased, and 8-OHdG content was low in the renal tubular cells of the CQ and AQ treatment groups compared with those of the diabetic control group. Our results suggest that CQ and AQ may be useful treatments for patients with diabetic kidney disease.


AMP-Activated Protein Kinases/metabolism , Amodiaquine/therapeutic use , Chloroquine/therapeutic use , Diabetes Mellitus, Experimental/complications , Diabetic Nephropathies/drug therapy , Enzyme Activators/therapeutic use , Animals , Antimalarials/therapeutic use , Cell Line , Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Experimental/pathology , Diabetic Nephropathies/metabolism , Diabetic Nephropathies/pathology , Glucose/metabolism , Humans , Kidney Tubules/drug effects , Kidney Tubules/metabolism , Kidney Tubules/pathology , Male , Mice, Inbred C57BL , Mitochondria/drug effects , Mitochondria/metabolism , Mitochondria/pathology , Phosphorylation/drug effects
19.
Sci Rep ; 7(1): 16782, 2017 12 01.
Article En | MEDLINE | ID: mdl-29196746

Diabetes is a risk factor for acute kidney injury (AKI) and chronic kidney disease (CKD). Diabetic patients are easy to progress to CKD after AKI. Currently, activation of fibrotic signalling including transforming growth factor-ß1 (TGF-ß1) is recognized as a key mechanism in CKD. Here, we investigated the influence of diabetes on CKD progression after AKI by using a unilateral renal ischaemia-reperfusion injury (IRI) model in diabetic mice. IRI induced extensive tubular injury, fibrosis and lymphocyte recruitment at 3 weeks after IRI, irrespective of diabetes. However, diabetes showed sustained tubular injury and markedly increased fibrosis and lymphocyte recruitment compared with non-diabetes at 5 week after IRI. The mRNAs and proteins related to TGF-ß1 and sonic hedgehog (Shh) signalling were significantly higher in diabetic versus non-diabetic IRI kidneys. During the in vitro study, the hyperglycaemia induced the activation of TGF-ß1 and Shh signalling and also increased profibrogenic phenotype change. However, hyperglycaemic control with insulin did not improve the progression of renal fibrosis and the activation of TGF-ß1 and Shh signalling. In conclusion, diabetes promotes CKD progression of AKI via activation of the TGF-ß1 and Shh signalling pathways, but insulin treatment was not enough for preventing the progression of renal fibrosis.


Acute Kidney Injury/complications , Diabetes Mellitus, Experimental/complications , Renal Insufficiency, Chronic/etiology , Reperfusion Injury/complications , Signal Transduction , Acute Kidney Injury/genetics , Acute Kidney Injury/metabolism , Animals , Cell Line , Diabetes Mellitus, Experimental/genetics , Diabetes Mellitus, Experimental/metabolism , Disease Models, Animal , Disease Progression , Gene Expression Regulation , Hedgehog Proteins/genetics , Hedgehog Proteins/metabolism , Humans , Male , Mice , Renal Insufficiency, Chronic/genetics , Renal Insufficiency, Chronic/metabolism , Reperfusion Injury/genetics , Reperfusion Injury/metabolism , Streptozocin , Transforming Growth Factor beta1/genetics , Transforming Growth Factor beta1/metabolism
20.
J Diabetes Res ; 2017: 6483572, 2017.
Article En | MEDLINE | ID: mdl-28409163

Purpose. In this study, we investigated the effect of PGC1α activators on mitochondrial fusion, fission, and autophagic quality control in renal tubular cells in a diabetic environment in vivo and in vitro. We also examined whether the upregulation of PGC1α attenuates diabetic tubulopathy by normalizing mitochondrial homeostasis. Methods. HKC8 cells were subjected to high-glucose conditions (30 mM D-glucose). Diabetes was induced with streptozotocin (STZ, 50 mg/kg i.p. for 5 days) in male C57/BL6J mice. AICAR or metformin was used as a PGC1α activator. Results. Treatment with the PGC1α activators AICAR and metformin improved functional mitochondrial mass in HKC8 cells in high-glucose conditions. Moreover, in renal proximal tubular cells, increased PGC1α activity correlated with the reversal of changes in Drp1, Mfn1, and LC3-II protein expression in a high-glucose environment. Normalized mitochondrial life cycles resulted in low ROS production and reduced apoptosis. AICAR and metformin treatment effectively mitigated albuminuria and renal histopathology and decreased the expression of TGFß1 and αSMA in the kidneys of diabetic mice. Conclusions. Our results demonstrate that increases in PGC1α activity improve diabetic tubulopathy by modulating mitochondrial dynamics and autophagy.


Aminoimidazole Carboxamide/analogs & derivatives , Autophagy/drug effects , Diabetes Mellitus, Experimental/metabolism , Diabetic Nephropathies/metabolism , Hypoglycemic Agents/pharmacology , Kidney Tubules, Proximal/drug effects , Metformin/pharmacology , Mitochondrial Dynamics/drug effects , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/drug effects , Ribonucleotides/pharmacology , Aminoimidazole Carboxamide/pharmacology , Animals , Apoptosis/drug effects , Cell Line , Glucose/pharmacology , Humans , Kidney Tubules, Proximal/cytology , Male , Mice , Sweetening Agents/pharmacology
...