Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
1.
Am J Physiol Renal Physiol ; 314(4): F501-F516, 2018 04 01.
Article in English | MEDLINE | ID: mdl-29187371

ABSTRACT

The erythropoietin receptor (EpoR) is widely expressed but its renoprotective action is unexplored. To examine the role of EpoR in vivo in the kidney, we induced acute kidney injury (AKI) by ischemia-reperfusion in mice with different EpoR bioactivities in the kidney. EpoR bioactivity was reduced by knockin of wild-type human EpoR, which is hypofunctional relative to murine EpoR, and a renal tubule-specific EpoR knockout. These mice had lower EPO/EpoR activity and lower autophagy flux in renal tubules. Upon AKI induction, they exhibited worse renal function and structural damage, more apoptosis at the acute stage (<7 days), and slower recovery with more tubulointerstitial fibrosis at the subacute stage (14 days). In contrast, mice with hyperactive EpoR signaling from knockin of a constitutively active human EpoR had higher autophagic flux, milder kidney damage, and better renal function at the acute stage but, surprisingly, worse tubulointerstitial fibrosis and renal function at the subacute stage. Either excess or deficient EpoR activity in the kidney was associated with abnormal peritubular capillaries and tubular hypoxia, creating a "U-shaped" relationship. The direct effects of EpoR on tubular cells were confirmed in vitro by a hydrogen peroxide model using primary cultured proximal tubule cells with different EpoR activities. In summary, normal erythropoietin (EPO)/EpoR signaling in renal tubules provides defense against renal tubular injury maintains the autophagy-apoptosis balance and peritubular capillary integrity. High and low EPO/EpoR bioactivities both lead to vascular defect, and high EpoR activity overides the tubular protective effects in AKI recovery.


Subject(s)
Acute Kidney Injury/metabolism , Capillaries/metabolism , Erythropoietin/metabolism , Kidney Tubules, Proximal/blood supply , Kidney Tubules, Proximal/metabolism , Neovascularization, Physiologic , Receptors, Erythropoietin/metabolism , Acute Kidney Injury/genetics , Acute Kidney Injury/pathology , Acute Kidney Injury/physiopathology , Animals , Apoptosis , Autophagy , Capillaries/pathology , Capillaries/physiopathology , Cell Hypoxia , Cells, Cultured , Disease Models, Animal , Fibrosis , Humans , Kidney Tubules, Proximal/pathology , Kidney Tubules, Proximal/physiopathology , Mice, 129 Strain , Mice, Transgenic , Receptors, Erythropoietin/deficiency , Receptors, Erythropoietin/genetics , Signal Transduction
2.
Lab Invest ; 98(8): 1105-1121, 2018 08.
Article in English | MEDLINE | ID: mdl-29858580

ABSTRACT

Cisplatin (CP)-induced nephrotoxicity is widely accepted as a model for acute kidney injury (AKI). Although cisplatin-induced chronic kidney disease (CKD) in rodent has been reported, the role of phosphate in the cisplatin-induced CKD progression is not described. In this study, we gave a single peritoneal injection of CP followed by high (2%) phosphate diet for 20 weeks. High dose CP (20 mg/Kg) led to high mortality; whereas a lower dose (10 mg/Kg) resulted in a full spectrum of AKI with tubular necrosis, azotemia, and 0% mortality 7 days after CP injection. After consuming a high phosphate diet, mice developed CKD characterized by low creatinine clearance, interstitial fibrosis, hyperphosphatemia, high plasma PTH and FGF23, low plasma 1,25(OH)2 Vitamin D3 and αKlotho, and classic uremic cardiovasculopathy. The CP model was robust in demonstrating the effect of aging, sexual dimorphism, and dietary phosphate on AKI and also AKI-to-CKD progression. Finally, we used the CP-high phosphate model to examine previously validated methods of genetically manipulated high αKlotho and therapy using exogenous soluble αKlotho protein supplementation. In this CP CKD model, αKlotho mitigated CKD progression, improved mineral homeostasis, and ameliorated cardiovascular disease. Taken together, CP and high phosphate nephrotoxicity is a reproducible and technically very simple model for the study of AKI, AKI-to-CKD progression, extrarenal complications of CKD, and for evaluation of therapeutic efficacy.


Subject(s)
Acute Kidney Injury/chemically induced , Cisplatin/toxicity , Kidney/drug effects , Renal Insufficiency, Chronic/chemically induced , Acute Kidney Injury/blood , Acute Kidney Injury/metabolism , Animals , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/toxicity , Cisplatin/administration & dosage , Disease Progression , Dose-Response Relationship, Drug , Female , Fibroblast Growth Factor-23 , Fibroblast Growth Factors/blood , Kidney/metabolism , Kidney/pathology , Male , Mice, 129 Strain , Mice, Transgenic , Phosphates/administration & dosage , Phosphates/toxicity , Rats, Sprague-Dawley , Renal Insufficiency, Chronic/blood , Renal Insufficiency, Chronic/metabolism
3.
Kidney Int ; 91(5): 1104-1114, 2017 05.
Article in English | MEDLINE | ID: mdl-28131398

ABSTRACT

α-Klotho is highly expressed in the kidney, and its extracellular domain is cleaved and released into the circulation. Chronic kidney disease (CKD) is a state of α-Klotho deficiency, which exerts multiple negative systemic effects on numerous organs including the cardiovascular system. Since acute kidney injury (AKI) greatly escalates the risk of CKD development, we explored the effect of α-Klotho on prevention and treatment on post-AKI to CKD progression and cardiovascular disease. Therein, ischemia reperfusion injury-induced AKI was followed by early administration of recombinant α-Klotho or vehicle starting one day and continued for four days after kidney injury (CKD prevention protocol). A CKD model was generated by unilateral nephrectomy plus contralateral ischemia reperfusion injury. Late administration of α-Klotho in this model was started four weeks after injury and sustained for 12 weeks (CKD treatment protocol). The prevention protocol precluded AKI to CKD progression and protected the heart from cardiac remodeling in the post-AKI model. One important effect of exogenous α-Klotho therapy was the restoration of endogenous α-Klotho levels long after the cessation of exogenous α-Klotho therapy. The treatment protocol still effectively improved renal function and attenuated cardiac remodeling in CKD, although these parameters did not completely return to normal. In addition, α-Klotho administration also attenuated high phosphate diet-induced renal and cardiac fibrosis, and improved renal and cardiac function in the absence of pre-existing renal disease. Thus, recombinant α-Klotho protein is safe and efficacious, and might be a promising prophylactic or therapeutic option for prevention or retardation of AKI-to-CKD progression and uremic cardiomyopathy.


Subject(s)
Acute Kidney Injury/drug therapy , Cardiomyopathies/drug therapy , Glucuronidase/metabolism , Glucuronidase/therapeutic use , Renal Insufficiency, Chronic/drug therapy , Acute Kidney Injury/blood , Acute Kidney Injury/complications , Acute Kidney Injury/metabolism , Animals , Biological Therapy/methods , Cardiomyopathies/etiology , Disease Models, Animal , Disease Progression , Drug Evaluation, Preclinical , Female , Fibrosis , Glucuronidase/administration & dosage , Humans , Injections, Intraperitoneal , Kidney/metabolism , Kidney/pathology , Klotho Proteins , Male , Mice , Myocardium/pathology , Recombinant Proteins/therapeutic use , Renal Insufficiency, Chronic/blood , Renal Insufficiency, Chronic/complications , Renal Insufficiency, Chronic/metabolism , Uremia/complications
4.
J Am Soc Nephrol ; 27(8): 2331-45, 2016 Aug.
Article in English | MEDLINE | ID: mdl-26701976

ABSTRACT

AKI confers increased risk of progression to CKD. αKlotho is a cytoprotective protein, the expression of which is reduced in AKI, but the relationship of αKlotho expression level to AKI progression to CKD has not been studied. We altered systemic αKlotho levels by genetic manipulation, phosphate loading, or aging and examined the effect on long-term outcome after AKI in two models: bilateral ischemia-reperfusion injury and unilateral nephrectomy plus contralateral ischemia-reperfusion injury. Despite apparent initial complete recovery of renal function, both types of AKI eventually progressed to CKD, with decreased creatinine clearance, hyperphosphatemia, and renal fibrosis. Compared with wild-type mice, heterozygous αKlotho-hypomorphic mice (αKlotho haploinsufficiency) progressed to CKD much faster, whereas αKlotho-overexpressing mice had better preserved renal function after AKI. High phosphate diet exacerbated αKlotho deficiency after AKI, dramatically increased renal fibrosis, and accelerated CKD progression. Recombinant αKlotho administration after AKI accelerated renal recovery and reduced renal fibrosis. Compared with wild-type conditions, αKlotho deficiency and overexpression are associated with lower and higher autophagic flux in the kidney, respectively. Upregulation of autophagy protected kidney cells in culture from oxidative stress and reduced collagen 1 accumulation. We propose that αKlotho upregulates autophagy, attenuates ischemic injury, mitigates renal fibrosis, and retards AKI progression to CKD.


Subject(s)
Acute Kidney Injury/complications , Autophagy/physiology , Disease Progression , Receptors, Cell Surface/physiology , Renal Insufficiency, Chronic/etiology , Animals , Autophagy/drug effects , Glucuronidase , Klotho Proteins , Mice , Receptors, Cell Surface/therapeutic use , Renal Insufficiency, Chronic/prevention & control
5.
J Am Soc Nephrol ; 27(1): 79-90, 2016 Jan.
Article in English | MEDLINE | ID: mdl-25977312

ABSTRACT

αKlotho is a multifunctional protein highly expressed in the kidney. Soluble αKlotho is released through cleavage of the extracellular domain from membrane αKlotho by secretases to function as an endocrine/paracrine substance. The role of the kidney in circulating αKlotho production and handling is incompletely understood, however. Here, we found higher αKlotho concentration in suprarenal compared with infrarenal inferior vena cava in both rats and humans. In rats, serum αKlotho concentration dropped precipitously after bilateral nephrectomy or upon treatment with inhibitors of αKlotho extracellular domain shedding. Furthermore, the serum half-life of exogenous αKlotho in anephric rats was four- to five-fold longer than that in normal rats, and exogenously injected labeled recombinant αKlotho was detected in the kidney and in urine of rats. Both in vivo (micropuncture) and in vitro (proximal tubule cell line) studies showed that αKlotho traffics from the basal to the apical side of the proximal tubule via transcytosis. Thus, we conclude that the kidney has dual roles in αKlotho homeostasis, producing and releasing αKlotho into the circulation and clearing αKlotho from the blood into the urinary lumen.


Subject(s)
Glucuronidase/metabolism , Kidney/metabolism , Animals , Glucuronidase/blood , Humans , Klotho Proteins , Rats
6.
PLoS One ; 12(9): e0181848, 2017.
Article in English | MEDLINE | ID: mdl-28886014

ABSTRACT

C-reactive protein (CRP), was recently reported to be closely associated with poor renal function in patients with acute kidney injury (AKI), but whether CRP is pathogenic or a mere biomarker in AKI remains largely unclear. Impaired autophagy is known to exacerbate renal ischemia-reperfusion injury (IRI). We examined whether the pathogenic role of CRP in AKI is associated with reduction of autophagy. We mated transgenic rabbit CRP over-expressing mice (Tg-CRP) with two autophagy reporter mouse lines, Tg-GFP-LC3 mice (LC3) and Tg-RFP-GFP-LC3 mice (RG-LC3) respectively to generate Tg-CRP-GFP-LC3 mice (PLC3) and Tg-CRP-RFP-GFP-LC3 mice (PRG-LC3). AKI was induced by IRI. Compared with LC3 mice, PLC3 mice developed more severe kidney damage after IRI. Renal tubules were isolated from LC3 mice at baseline for primary culture. OKP cells were transiently transfected with GFP-LC3 plasmid. CRP addition exacerbated lactate dehydrogenase release from both cell types. Immunoblots showed lower LC-3 II/I ratios and higher levels of p62, markers of reduced autophagy flux, in the kidneys of PLC3 mice compared to LC3 mice after IRI, and in primary cultured renal tubules and OKP cells treated with CRP and H2O2 compared to H2O2 alone. Immunohistochemistry showed much fewer LC-3 punctae, and electron microscopy showed fewer autophagosomes in kidneys of PLC3 mice compared to LC3 mice after IRI. Similarly, CRP addition reduced GFP-LC3 punctae induced by H2O2 in primary cultured proximal tubules and in GFP-LC3 plasmid transfected OKP cells. Rapamycin, an autophagy inducer, rescued impaired autophagy and reduced renal injury in vivo. In summary, it was suggested that CRP be more than mere biomarker in AKI, and render the kidney more susceptible to ischemic/oxidative injury, which is associated with down-regulating autophagy flux.


Subject(s)
Acute Kidney Injury/etiology , Autophagy/genetics , C-Reactive Protein/genetics , Gene Expression , Reperfusion Injury/complications , Reperfusion Injury/genetics , Acute Kidney Injury/pathology , Animals , Autophagy/drug effects , Beclin-1/metabolism , C-Reactive Protein/metabolism , Disease Models, Animal , Epithelial Cells/metabolism , Humans , Kidney Tubules/metabolism , Mice , Protein Binding , Proto-Oncogene Proteins c-bcl-2/metabolism , Rabbits , Reperfusion Injury/pathology , Severity of Illness Index , Sirolimus/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL