Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 27
Filter
Add more filters










Publication year range
1.
Clin Sci (Lond) ; 138(10): 599-614, 2024 May 22.
Article in English | MEDLINE | ID: mdl-38739452

ABSTRACT

AIM: Acute kidney injury (AKI) increases the risk for progressive chronic kidney disease (CKD). MicroRNA (miR)-486-5p protects against kidney ischemia-reperfusion (IR) injury in mice, although its long-term effects on the vasculature and development of CKD are unknown. We studied whether miR-486-5p would prevent the AKI to CKD transition in rat, and affect vascular function. METHODS: Adult male rats were subjected to bilateral kidney IR followed by i.v. injection of liposomal-packaged miR-486-5p (0.5 mg/kg). Kidney function and histologic injury were assessed after 24 h and 10 weeks. Kidney endothelial protein levels were measured by immunoblot and immunofluorescence, and mesenteric artery reactivity was determined by wire myography. RESULTS: In rats with IR, miR-486-5p blocked kidney endothelial cell increases in intercellular adhesion molecule-1 (ICAM-1), reduced neutrophil infiltration and histologic injury, and normalized plasma creatinine (P<0.001). However, miR-486-5p attenuated IR-induced kidney endothelial nitric oxide synthase (eNOS) expression (P<0.05). At 10 weeks, kidneys from rats with IR alone had decreased peritubular capillary density and increased interstitial collagen deposition (P<0.0001), and mesenteric arteries showed impaired endothelium-dependent vasorelaxation (P<0.001). These changes were inhibited by miR-486-5p. Delayed miR-486-5p administration (96 h, 3 weeks after IR) had no impact on kidney fibrosis, capillary density, or endothelial function. CONCLUSION: In rats, administration of miR-486-5p early after kidney IR prevents injury, and protects against CKD development and systemic endothelial dysfunction. These protective effects are associated with inhibition of endothelial ICAM-1 and occur despite reduction in eNOS. miR-486-5p holds promise for the prevention of ischemic AKI and its complications.


Subject(s)
Acute Kidney Injury , Intercellular Adhesion Molecule-1 , Kidney , MicroRNAs , Rats, Sprague-Dawley , Renal Insufficiency, Chronic , Reperfusion Injury , Animals , MicroRNAs/metabolism , MicroRNAs/genetics , Male , Acute Kidney Injury/prevention & control , Acute Kidney Injury/metabolism , Acute Kidney Injury/genetics , Acute Kidney Injury/pathology , Renal Insufficiency, Chronic/prevention & control , Renal Insufficiency, Chronic/metabolism , Renal Insufficiency, Chronic/genetics , Renal Insufficiency, Chronic/pathology , Kidney/pathology , Kidney/blood supply , Kidney/metabolism , Reperfusion Injury/prevention & control , Reperfusion Injury/metabolism , Intercellular Adhesion Molecule-1/metabolism , Intercellular Adhesion Molecule-1/genetics , Nitric Oxide Synthase Type III/metabolism , Rats , Disease Models, Animal , Disease Progression , Endothelial Cells/metabolism
2.
Can J Kidney Health Dis ; 10: 20543581231207146, 2023.
Article in English | MEDLINE | ID: mdl-37881406

ABSTRACT

Background: Angiotensin-converting enzyme 2 (ACE2) hydrolyzes angiotensin (Ang) II to Ang-(1-7), promoting vasodilatation, and inhibiting oxidative stress and inflammation. Plasma membrane ACE2 is the receptor for all known SARS-CoV-2 (severe acute respiratory syndrome coronavirus 2) viral variants. In COVID-19 infection, soluble ACE2 variants may act as decoys to bind and neutralize the coronavirus, reducing its tissue infectivity. Furthermore, soluble ACE2 variants have been proposed as potential therapeutics for kidney disease and hypertensive disorders. Objective: Soluble ACE2 variants conjugated to human Fc domains and selected for high-potency viral SARS-CoV-2 neutralization were prepared and evaluated for ACE2 activity in vitro. Lead candidates were then tested for systemic ACE2 activity, stability, and effects on blood pressure and albuminuria in mice with Ang II-induced hypertension. Methods: ACE2 activity of 10 soluble ACE2 variants was first assessed in cell-free conditions using a fluorogenic substrate, or by Ang II hydrolysis to Ang-(1-7). Hypertension was induced in male or female mice by implantation of osmotic minipumps containing Ang II. Two lead ACE2 variants were injected intravenously (i.v.) into hypertensive mice, followed by measurements of blood pressure (tail-cuff plethysmography), albuminuria, and tissue ACE2 activity and protein (immunoblots). Results: Soluble ACE2-Fc variants demonstrated significant ACE2 enzymatic activity, with kinetics comparable with human recombinant ACE2. In hypertensive mice, single dose i.v. injection of ACE2-Fc variant K (10 mg/kg) significantly decreased systolic blood pressure at 24 hours, with partial lowering sustained to 48 hours, and tendency to reduce albuminuria at 72 hours. By contrast, ACE2-Fc variant I had no effect on blood pressure or albuminuria in hypertensive mice; ACE2-Fc variant K was detected by immunoblotting in plasma, kidney, heart, lung, liver, and spleen lysates 72 hours after injection, associated with significantly increased ACE2 activity in all tissues except kidney and spleen. Angiotensin-converting enzyme 2-Fc variant I had no effect on plasma ACE2 activity. Conclusions: Soluble ACE2-Fc variant K reduces blood pressure and tends to lower albuminuria in hypertensive mice. Furthermore, soluble ACE2-Fc variant K has prolonged tissue retention, associated with increased tissue ACE2 activity. The results support further studies directed at the therapeutic potential of soluble ACE2-Fc variant K for cardiovascular and kidney protection.


Contexte: L'enzyme de conversion de l'angiotensine 2 (ACE2) hydrolyse l'angiotensine (Ang) II en angiotensine (Ang)-(1-7), ce qui favorise la vasodilatation et inhibe le stress oxydatif et l'inflammation. L'ACE2 de la membrane plasmique est le récepteur de tous les variants connus du SARS-COV-2. Dans les cas d'infection à la COVID-19, les variants solubles de l'ACE2 peuvent agir comme leurres pour lier et neutraliser le coronavirus, et réduire ainsi son infectiosité dans les tissus. Des variants solubles de l'ACE2 ont également été proposés comme agents thérapeutiques potentiels pour l'insuffisance rénale et les troubles liés à l'hypertension. Objectif: Des variants solubles de l'ACE2 conjugués au domaine Fc humain ont été sélectionnés pour leur fort potentiel neutralisant du virus SARS-COV-2, puis préparés et évalués pour la mesure de l'activité de l'ACE2 in vitro. Les meilleurs candidats ont ensuite été testés chez des souris souffrant d'hypertension induite par l'Ang II afin de mesurer l'activité d'ACE2, ainsi que leur stabilité et leurs effets sur la pression artérielle et l'albuminurie. Méthodologie: L'activité de 10 variants solubles de l'ACE2 a d'abord été évaluée en conditions acellulaires à l'aide d'un substrat fluorogène, ou par hydrolyse de l'Ang II en Ang-(1-7). L'hypertension a été induite chez des souris mâles ou femelles par l'implantation de minipompes osmotiques contenant de l'Ang II. Deux des meilleurs variants de l'ACE2 ont été injectés par voie intraveineuse (i.v.) à des souris hypertendues, puis des mesures de la pression artérielle (pléthysmographie par manchon caudal), de l'albuminurie, de l'activité de l'ACE2 dans les tissus et des protéines (immunobuvardage) ont été effectuées. Résultats: Les variants solubles ACE2-Fc ont montré une activité enzymatique significative, avec une cinétique comparable à celle de l'ACE2 recombinante humaine. Chez les souris hypertendues, l'injection i.v. d'une dose unique (10 mg/kg) du variant K ACE2-Fc a abaissé significativement la pression artérielle systolique après 24 heures­une réduction partielle s'étant prolongée jusqu'à 48 heures­et a montré une tendance à réduire l'albuminurie après 72 heures. En revanche, le variant I ACE2-Fc n'a eu aucun effet sur la pression artérielle ou l'albuminurie des souris hypertendues. Après 72 heures, le variant K ACE2-Fc a été détecté par immunobuvardage dans le plasma, ainsi que dans des lysats de reins, de cœur, de poumon, de foie et de rate, ce qui a été associé à une augmentation significative de l'activité de l'ACE2 dans tous les tissus sauf dans les reins et la rate. Le variant I ACE2-Fc n'a montré aucun effet sur l'activité de l'ACE2 dans le plasma. Conclusion: Le variant soluble K ACE2-Fc abaisse la pression artérielle et tend à diminuer l'albuminurie chez les souris hypertendues. Il présente en outre une rétention tissulaire prolongée, laquelle est associée à une plus grande activité de l'ACE2 dans les tissus. Ces résultats appuient d'autres études portant sur le potentiel thérapeutique du variant soluble K ACE2-Fc dans la protection cardiovasculaire et rénale.

3.
Kidney Int ; 100(3): 597-612, 2021 09.
Article in English | MEDLINE | ID: mdl-34181969

ABSTRACT

Acute kidney injury (AKI) carries high morbidity and mortality, and effective treatments are lacking. Preclinical models support involvement of micro-RNAs (miRs) in AKI pathogenesis, although effects on the kidney transcriptome are unclear. We previously showed that injection of cord blood endothelial colony forming cell-derived exosomes, enriched in miR-486-5p, prevented ischemic AKI in mice. To further define this, we studied direct effects of miR-486-5p in mice with kidney ischemia-reperfusion injury. RNA-Seq was used to compare the impact of miR-486-5p and exosomes on the transcriptome of proximal tubules and kidney endothelial cells 24 hours after ischemia-reperfusion. In mice with AKI, injection of miR-486-5p mimic increased its levels in proximal tubules and endothelial cells, and improved plasma creatinine, histological injury, neutrophil infiltration, and apoptosis. Additionally, miR-486-5p inhibited expression of its target phosphatase and tensin homolog, and activated protein kinase B. In proximal tubules, miR-486-5p or exosomes reduced expression of genes associated with ischemic injury and the tumor necrosis factor (TNF) pathway, and altered distinct apoptotic genes. In endothelial cells, genes associated with metabolic processes were altered by miR-486-5p or exosomes, although TNF pathway genes were not affected. Thus, our results suggest that miR-486-5p may have therapeutic potential in AKI.


Subject(s)
Acute Kidney Injury , MicroRNAs , Reperfusion Injury , Acute Kidney Injury/genetics , Acute Kidney Injury/prevention & control , Animals , Apoptosis , Endothelial Cells , Ischemia , Kidney , Mice , MicroRNAs/genetics , Reperfusion Injury/genetics , Reperfusion Injury/prevention & control , Transcriptome
4.
Clin Sci (Lond) ; 134(14): 1887-1909, 2020 07 31.
Article in English | MEDLINE | ID: mdl-32662516

ABSTRACT

Female sex protects against development of acute kidney injury (AKI). While sex hormones may be involved in protection, the role of differential gene expression is unknown. We conducted gene profiling in male and female mice with or without kidney ischemia-reperfusion injury (IRI). Mice underwent bilateral renal pedicle clamping (30 min), and tissues were collected 24 h after reperfusion. RNA-sequencing (RNA-Seq) was performed on proximal tubules (PTs) and kidney endothelial cells. Female mice were resistant to ischemic injury compared with males, determined by plasma creatinine and neutrophil gelatinase-associated lipocalin (NGAL), histologic scores, neutrophil infiltration, and extent of apoptosis. Sham mice had sex-specific gene disparities in PT and endothelium, and male mice showed profound gene dysregulation with ischemia-reperfusion compared with females. After ischemia PTs from females exhibited smaller increases compared with males in injury-associated genes lipocalin-2 (Lcn2), hepatitis A virus cellular receptor 1 (Havcr1), and keratin 18 (Krt18), and no up-regulation of SRY-Box transcription factor 9 (Sox9) or keratin 20 (Krt20). Endothelial up-regulation of adhesion molecules and cytokines/chemokines occurred in males, but not females. Up-regulated genes in male ischemic PTs were linked to tumor necrosis factor (TNF) and Toll-like receptor (TLR) pathways, while female ischemic PTs showed up-regulated genes in pathways related to transport. The data highlight sex-specific gene expression differences in male and female PTs and endothelium before and after ischemic injury that may underlie disparities in susceptibility to AKI.


Subject(s)
Acute Kidney Injury/metabolism , Endothelial Cells/metabolism , Kidney Tubules, Proximal/metabolism , Reperfusion Injury/metabolism , Sex Characteristics , Acute Kidney Injury/genetics , Animals , Female , Gene Expression Profiling , Male , Mice , Reperfusion Injury/genetics , Sequence Analysis, RNA
5.
Lab Invest ; 100(3): 414-425, 2020 03.
Article in English | MEDLINE | ID: mdl-31527829

ABSTRACT

Prostaglandin E2 receptor EP1 (PGE2/EP1) promotes diabetic renal injury, and EP1 receptor deletion improves hyperfiltration, albuminuria, and fibrosis. The role of EP1 receptors in hypertensive kidney disease (HKD) remains controversial. We examined the contribution of EP1 receptors to HKD. EP1 null (EP1-/-) mice were bred with hypertensive TTRhRen mice (Htn) to evaluate kidney function and injury at 24 weeks. EP1 deletion had no effect on elevation of systolic blood pressure in Htn mice (HtnEP1-/-) but resulted in pronounced albuminuria and reduced FITC-inulin clearance, compared with Htn or wild-type (WT) mice. Ultrastructural injury to podocytes and glomerular endothelium was prominent in HtnEP1-/- mice; including widened subendothelial space, subendothelial lucent zones and focal lifting of endothelium from basement membrane, with focal subendothelial cell debris. Cortex COX2 mRNA was increased by EP1 deletion. Glomerular EP3 mRNA was reduced by EP1 deletion, and EP4 by Htn and EP1 deletion. In WT mice, PGE2 increased chloride reabsorption via EP1 in isolated perfused thick ascending limb (TAL), but PGE2 or EP1 deletion did not affect vasopressin-mediated chloride reabsorption. In WT and Htn mouse inner medullary collecting duct (IMCD), PGE2 inhibited vasopressin-water transport, but not in EP1-/- or HtnEP1-/- mice. Overall, EP1 mediated TAL and IMCD transport in response to PGE2 is unaltered in Htn, and EP1 is protective in HKD.


Subject(s)
Hypertension, Renal , Podocytes , Receptors, Prostaglandin E, EP1 Subtype , Animals , Disease Models, Animal , Endothelial Cells/cytology , Endothelial Cells/metabolism , Endothelial Cells/pathology , Gene Deletion , Glomerular Filtration Rate/genetics , Hypertension, Renal/metabolism , Hypertension, Renal/pathology , Kidney Glomerulus/metabolism , Kidney Glomerulus/pathology , Male , Mice , Mice, Transgenic , Podocytes/cytology , Podocytes/metabolism , Podocytes/pathology , Receptors, Prostaglandin E, EP1 Subtype/genetics , Receptors, Prostaglandin E, EP1 Subtype/metabolism
6.
Sci Rep ; 8(1): 16320, 2018 11 05.
Article in English | MEDLINE | ID: mdl-30397255

ABSTRACT

Endothelial colony forming cell (ECFC)-derived exosomes protect mice against ischemic kidney injury, via transfer of microRNA-(miR)-486-5p. Mechanisms mediating exosome recruitment to tissues are unclear. We hypothesized that ECFC exosomes target ischemic kidneys, involving interaction between exosomal CXC chemokine receptor type 4 (CXCR4) and stromal cell-derived factor (SDF)-1α. Ischemia-reperfusion was induced in mice by bilateral renal vascular clamp, with intravenous infusion of exosomes at reperfusion. Optical imaging determined exosome biodistribution, and miR-486-5p was measured by real-time PCR. Human umbilical vein endothelial cells (HUVECs) were cultured to study the CXCR4/SDF-1α interaction. Targeting of administered exosomes to ischemic kidneys was detected 30 min and 4 hrs after reperfusion. Exosomes increased miR-486-5p levels only in kidneys, within proximal tubules, glomeruli, and endothelial cells. Uptake of fluorescently-labeled exosomes into HUVECs, and exosomal transfer of miR-486-5p were enhanced by hypoxia, effects blocked by neutralizing antibody to SDF-1α or by the CXCR4 inhibitor plerixafor. Infusion of ECFC exosomes prevented ischemic kidney injury in vivo, an effect that was not observed when exosomes were pre-incubated with plerixafor. These data indicate that ECFC exosomes selectively target the kidneys after ischemic injury, with rapid cellular transfer of miR486-5p. Targeting of exosomes may involve interaction of CXCR4 with endothelial cell SDF-1α.


Subject(s)
Chemokine CXCL12/metabolism , Endothelial Cells/pathology , Exosomes/metabolism , Ischemia/metabolism , Ischemia/pathology , Kidney/blood supply , Receptors, CXCR4/metabolism , Animals , Ischemia/genetics , Kidney/pathology , Ligands , Mice , MicroRNAs/genetics , Protein Binding
7.
Lab Invest ; 98(3): 360-370, 2018 03.
Article in English | MEDLINE | ID: mdl-29251736

ABSTRACT

PGE2 regulates glomerular hemodynamics, renin secretion, and tubular transport. This study examined the contribution of PGE2 EP1 receptors to sodium and water homeostasis. Male EP1-/- mice were bred with hypertensive TTRhRen mice (Htn) to evaluate blood pressure and kidney function at 8 weeks of age in four groups: wildtype (WT), EP1-/-, Htn, HtnEP1-/-. Blood pressure and water balance were unaffected by EP1 deletion. COX1 and mPGE2 synthase were increased and COX2 was decreased in mice lacking EP1, with increases in EP3 and reductions in EP2 and EP4 mRNA throughout the nephron. Microdissected proximal tubule sglt1, NHE3, and AQP1 were increased in HtnEP1-/-, but sglt2 was increased in EP1-/- mice. Thick ascending limb NKCC2 was reduced in the cortex but increased in the medulla. Inner medullary collecting duct (IMCD) AQP1 and ENaC were increased, but AVP V2 receptors and urea transporter-1 were reduced in all mice compared to WT. In WT and Htn mice, PGE2 inhibited AVP-water transport and increased calcium in the IMCD, and inhibited sodium transport in cortical collecting ducts, but not in EP1-/- or HtnEP1-/- mice. Amiloride (ENaC) and hydrochlorothiazide (pendrin inhibitor) equally attenuated the effect of PGE2 on sodium transport. Taken together, the data suggest that EP1 regulates renal aquaporins and sodium transporters, attenuates AVP-water transport and inhibits sodium transport in the mouse collecting duct, which is mediated by both ENaC and pendrin-dependent pathways.


Subject(s)
Dinoprostone/metabolism , Hypertension/metabolism , Kidney Tubules, Collecting/metabolism , Receptors, Prostaglandin E, EP1 Subtype/metabolism , Sodium/metabolism , Animals , Aquaporins/metabolism , Blood Pressure , Calcium/metabolism , Glomerular Filtration Rate , Male , Mice , Prostaglandin-E Synthases/metabolism , Prostaglandin-Endoperoxide Synthases/metabolism , Sodium-Hydrogen Exchanger 3/metabolism , Solute Carrier Family 12, Member 1/metabolism
8.
Am J Physiol Renal Physiol ; 312(2): F335-F342, 2017 Feb 01.
Article in English | MEDLINE | ID: mdl-27733369

ABSTRACT

The relationship between the renal renin-angiotensin aldosterone system (RAAS) and cardiorenal pathophysiology is unclear. Our aims were to assess 1) levels of urinary RAAS components and 2) the association between RAAS components and HbA1c, the urine albumin/creatinine ratio (ACR), estimated glomerular filtration rate (eGFR), and blood pressure (BP) in otherwise healthy adolescents with type 1 diabetes mellitus (TID) vs. healthy controls (HC). Urinary angiotensinogen and angtionsin-converting enzyme (ACE) 2 levels, activity of ACE and ACE2, BP, HbA1c, ACR, and eGFR were measured in 65 HC and 194 T1D from the Adolescent Type 1 Diabetes Cardio-Renal Intervention Trial (AdDIT). Urinary levels of all RAAS components were higher in T1D vs. HC (P < 0.0001). Higher HbA1c was associated with higher urinary angiotensinogen, ACE2, and higher activity of ACE and ACE2 (P < 0.0001, P = 0.0003, P = 0.003, and P = 0.007 respectively) in T1D. Higher ACR (within the normal range) was associated with higher urinary angiotensinogen (P < 0.0001) and ACE activity (P = 0.007), but not with urinary ACE2 activity or ACE2 levels. These observations were absent in HC. Urinary RAAS components were not associated with BP or eGFR in T1D or HC. Otherwise healthy adolescents with T1D exhibit higher levels of urinary RAAS components compared with HC. While levels of all urinary RAAS components correlate with HbA1c in T1D, only urinary angiotensinogen and ACE activity correlate with ACR, suggesting that these factors reflect an intermediary pathogenic link between hyperglycemia and albuminuria within the normal range.


Subject(s)
Blood Pressure/physiology , Diabetes Mellitus, Type 1/physiopathology , Kidney/physiopathology , Renin-Angiotensin System/physiology , Adolescent , Albuminuria/metabolism , Angiotensinogen/urine , Biomarkers/metabolism , Creatinine/urine , Diabetes Mellitus, Type 1/metabolism , Female , Glomerular Filtration Rate/physiology , Humans , Kidney/metabolism , Male , Peptidyl-Dipeptidase A/urine
9.
Kidney Int ; 90(6): 1238-1250, 2016 12.
Article in English | MEDLINE | ID: mdl-27650731

ABSTRACT

Administration of human cord blood endothelial colony-forming cells (ECFCs) or their exosomes protects mice against kidney ischemia/reperfusion injury. Here we studied the microRNA (miRNA) content of ECFC exosomes and the role of miRNA transfer in kidney and endothelial cell protection. ECFC exosomes were enriched in miR-486-5p, which targets the phosphatase and tensin homolog (PTEN) and the Akt pathway. In cultured endothelial cells exposed to hypoxia, incubation with ECFC exosomes increased miR-486-5p, decreased PTEN, and stimulated Akt phosphorylation. Exposure of hypoxic endothelial cells to conditioned medium from ECFCs pretreated with anti-miR-486-5p blocked increases in miR-486-5p and phosphorylated Akt, restored expression of PTEN, and enhanced apoptosis. Coculture of endothelial cells with ECFCs enhanced endothelial miR-486-5p levels. Targeting of PTEN by miR-486-5p was observed in endothelial cells, and PTEN knockdown blocked apoptosis. In mice with ischemic kidney injury, infusion of ECFC exosomes induced potent functional and histologic protection, associated with increased kidney miR-486-5p levels, decreased PTEN, and activation of Akt. Infusion of exosomes from ECFCs transfected with anti-miR-486-5p had no protective effect. Thus, delivery of ECFC exosomes reduces ischemic kidney injury via transfer of miR-486-5p targeting PTEN. Exosomes enriched in miR-486-5p could represent a therapeutic tool in acute kidney injury.


Subject(s)
Acute Kidney Injury/metabolism , Exosomes/metabolism , MicroRNAs/metabolism , PTEN Phosphohydrolase/metabolism , Reperfusion Injury/metabolism , Animals , Apoptosis , Cells, Cultured , Endothelial Cells/physiology , Human Umbilical Vein Endothelial Cells , Humans , Male , Mice
10.
Front Pharmacol ; 7: 146, 2016.
Article in English | MEDLINE | ID: mdl-27313531

ABSTRACT

Angiotensin-converting enzyme 2 (ACE2) degrades angiotensin (Ang) II to Ang-(1-7), and protects against diabetic renal injury. Soluble ACE2 fragments are shed from the proximal tubule, and appear at high levels in the urine with diabetes. High glucose-induced shedding of ACE2 from proximal tubular cells is mediated by the enzyme "a disintegrin and metalloproteinase-17″ (ADAM17). Here, we investigated the mechanism for constitutive shedding of ACE2. Mouse proximal tubular cells were cultured and ACE2 shedding into the media was assessed by enzyme activity assay and immunoblot analysis. Cells were incubated with pharmacologic inhibitors, or transfected with silencing (si) RNA. Incubation of proximal tubular cells with increasing concentrations of D-glucose stimulated ACE2 shedding, which peaked at 16 mM, while L-glucose (osmotic control) had no effect on shedding. In cells maintained in 7.8 mM D-glucose, ACE2 shedding was significantly inhibited by the pan-protein kinase C (PKC) competitive inhibitor sotrastaurin, but not by an inhibitor of ADAM17. Incubation of cells with the PKC-α and -ß1-specific inhibitor Go6976, the PKC ß1 and ß2-specific inhibitor ruboxistaurin, inhibitors of matrix metalloproteinases-2,-8, and -9, or an inhibitor of ADAM10 (GI250423X) had no effect on basal ACE2 shedding. By contrast, the PKC-δ inhibitor rottlerin significantly inhibited both constitutive and high glucose-induced ACE2 shedding. Transfection of cells with siRNA directed against PKC-δ reduced ACE2 shedding by 20%, while knockdown of PKC-ε was without effect. These results indicate that constitutive shedding of ACE2 from proximal tubular cells is mediated by PKC-δ, which is also linked to high glucose-induced shedding. Targeting PKC-δ may preserve membrane-bound ACE2 in proximal tubule in disease states and diminish Ang II-stimulated adverse signaling.

11.
Lab Invest ; 95(9): 1044-55, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26121313

ABSTRACT

Renal prostaglandin (PG) E2 regulates salt and water transport, and affects disease processes via EP1-4 receptors, but its role in the proximal tubule (PT) is unknown. Our study investigates the effects of PGE2 on mouse PT fluid reabsorption, and its role in growth, sodium transporter expression, fibrosis, and oxidative stress in a mouse PT cell line (MCT). To determine which PGE2 EP receptors are expressed in MCT, qPCR for EP1-4 was performed on cells stimulated for 24 h with PGE2 or transforming growth factor beta (TGFß), a known mediator of PT injury in kidney disease. EP1 and EP4 were detected in MCT, but EP2 and EP3 are not expressed. EP1 was increased by PGE2 and TGFß, but EP4 was unchanged. To confirm the involvement of EP1 and EP4, sulprostone (SLP, EP1/3 agonist), ONO8711 (EP1 antagonist), and EP1 and EP4 siRNA were used. We first show that PGE2, SLP, and TGFß reduced H(3)-thymidine and H(3)-leucine incorporation. The effects on cell-cycle regulators were examined by western blot. PGE2 increased p27 via EP1 and EP4, but TGFß increased p21; PGE2-induced p27 was attenuated by TGFß. PGE2 and SLP reduced cyclinE, while TGFß increased cyclinD1, an effect attenuated by PGE2 administration. Na-K-ATPase α1 (NaK) was increased by PGE2 via EP1 and EP4. TGFß had no effect on NaK. Additionally, PGE2 and TGFß increased fibronectin levels, reaching 12-fold upon co-stimulation. EP1 siRNA abrogated PGE2-fibronectin. PGE2 also increased ROS generation, and ONO-8711 blocked PGE2-ROS. Finally, PGE2 significantly increased fluid reabsorption by 31 and 46% in isolated perfused mouse PT from C57BL/6 and FVB mice, respectively, and this was attenuated in FVB-EP1 null mice. Altogether PGE2 acting on EP1 and EP4 receptors may prove to be important mediators of PT injury, and salt and water transport.


Subject(s)
Dinoprostone/pharmacology , Kidney Tubules, Proximal/physiology , Receptors, Prostaglandin E, EP1 Subtype/metabolism , Receptors, Prostaglandin E, EP4 Subtype/metabolism , Renal Reabsorption/drug effects , Acridines , Analysis of Variance , Animals , Blotting, Western , Bridged Bicyclo Compounds/pharmacology , Caproates/pharmacology , Cyclin D1/metabolism , Cyclin E/metabolism , Dinoprostone/analogs & derivatives , Dinoprostone/metabolism , Kidney Tubules, Proximal/drug effects , Mice , Mice, Inbred C57BL , Oxidative Stress/drug effects , RNA, Small Interfering/pharmacology , Receptors, Prostaglandin E, EP1 Subtype/agonists , Receptors, Prostaglandin E, EP1 Subtype/antagonists & inhibitors , Transforming Growth Factor beta/pharmacology
12.
Am J Pathol ; 185(3): 729-40, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25625676

ABSTRACT

Angiotensin-(1-7) is a ligand for the Mas receptor and may protect against tissue injury associated with renin-angiotensin system activation. We determined the effects of endogenous or exogenous angiotensin-(1-7) in mice with unilateral ureteral obstruction (UUO). Mice with UUO were treated with or without the angiotensin-(1-7) antagonist A779 or with 6, 24, or 62 µg/kg per hour exogenous angiotensin-(1-7). After 10 days, kidneys were harvested for histology, immunoblots, and measurement of NADPH oxidase. Compared with controls, A779 treatment significantly increased fibronectin, transforming growth factor-ß, and α-smooth muscle actin expression in obstructed kidneys and enhanced tubulointerstitial injury, apoptosis, and NADPH oxidase. Unexpectedly, administration of angiotensin-(1-7) to mice with UUO caused injury in obstructed kidneys compared with controls and increased macrophage infiltration. In obstructed kidneys from mice with gene deletion of Mas (Mas(-/-)), apoptosis and macrophage infiltration were increased compared with wild-type mice. Angiotensin-(1-7) (but not A779) further increased apoptosis and macrophage influx in obstructed kidneys from Mas(-/-) mice, compared with untreated Mas(-/-) mice. These data indicate that endogenous angiotensin-(1-7) protects against kidney injury in UUO. In mice with or without the Mas receptor, however, delivery of exogenous angiotensin-(1-7) worsens kidney damage. The results suggest dose-dependent effects of angiotensin-(1-7) in the kidney in UUO, with endogenous angiotensin-(1-7) promoting repair pathways via interaction with Mas and higher amounts exacerbating injury.


Subject(s)
Angiotensin II/analogs & derivatives , Angiotensin I/therapeutic use , Peptide Fragments/therapeutic use , Ureter/drug effects , Ureteral Obstruction/drug therapy , Actins/metabolism , Angiotensin I/antagonists & inhibitors , Angiotensin I/pharmacology , Angiotensin II/pharmacology , Angiotensin II/therapeutic use , Animals , Fibronectins/metabolism , Male , Mice , Muscle, Smooth/drug effects , Muscle, Smooth/metabolism , Peptide Fragments/antagonists & inhibitors , Peptide Fragments/pharmacology , Transforming Growth Factor beta/metabolism , Ureter/metabolism , Ureteral Obstruction/metabolism
14.
Can J Physiol Pharmacol ; 92(8): 703-6, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24920267

ABSTRACT

Angiotensin-converting enzyme 2 (ACE2) is expressed in the kidney and may be renoprotective. We determined whether urinary ACE2 enzyme activity and protein levels (ELISA), as well as angiotensinogen and ACE, are elevated during clamped euglycemia (4-6 mmol·L(-1)) in patients with uncomplicated type 1 diabetes (T1D, n = 58) compared with normoglycemic controls (n = 21). We also measured the effect of clamped hyperglycemia (9-11 mmol·L(-1)) on each urinary factor in T1D patients. Urinary ACE2 activity and protein levels were higher during clamped euglycemia in T1D compared with the controls (p < 0.0001). In contrast, urinary angiotensinogen levels (p = 0.27) and ACE excretion (p = 0.68) did not differ. In response to clamped hyperglycemia in T1D, urinary ACE2 protein decreased (p < 0.0001), whereas urinary ACE2 activity as well as angiotensinogen and ACE levels remained unchanged. Urinary ACE2 activity and protein expression are increased in T1D patients prior to the onset of clinical complications. Further work is required to determine the functional role of urinary ACE2 in early T1D.


Subject(s)
Diabetes Mellitus, Type 1/urine , Peptidyl-Dipeptidase A/urine , Adult , Angiotensin-Converting Enzyme 2 , Angiotensinogen/metabolism , Case-Control Studies , Cohort Studies , Female , Glucose Clamp Technique , Humans , Male , Peptidyl-Dipeptidase A/metabolism
15.
PLoS One ; 9(1): e85958, 2014.
Article in English | MEDLINE | ID: mdl-24454948

ABSTRACT

Angiotensin-converting enzyme 2 (ACE2) is highly expressed in the kidney proximal tubule, where it cleaves angiotensin (Ang) II to Ang-(1-7). Urinary ACE2 levels increase in diabetes, suggesting that ACE2 may be shed from tubular cells. The aim of this study was to determine if ACE2 is shed from proximal tubular cells, to characterize ACE2 fragments, and to study pathways for shedding. Studies involved primary cultures of mouse proximal tubular cells, with ACE2 activity measured using a synthetic substrate, and analysis of ACE2 fragments by immunoblots and mass spectrometry. The culture media from mouse proximal tubular cells demonstrated a time-dependent increase in ACE2 activity, suggesting constitutive ACE2 shedding. ACE2 was detected in media as two bands at ∼ 90 kDa and ∼ 70 kDa on immunoblots. By contrast, full-length ACE2 appeared at ∼ 100 kDa in cell lysates or mouse kidney cortex. Mass spectrometry of the two deglycosylated fragments identified peptides matching mouse ACE2 at positions 18-706 and 18-577, respectively. The C-terminus of the 18-706 peptide fragment contained a non-tryptic site, suggesting that Met(706) is a candidate ACE2 cleavage site. Incubation of cells in high D-glucose (25 mM) (and to a lesser extent Ang II) for 48-72 h increased ACE2 activity in the media (p<0.001), an effect blocked by inhibition of a disintegrin and metalloproteinase (ADAM)17. High D-glucose increased ADAM17 activity in cell lysates (p<0.05). These data indicate that two glycosylated ACE2 fragments are constitutively shed from mouse proximal tubular cells. ACE2 shedding is stimulated by high D-glucose, at least partly via an ADAM17-mediated pathway. The results suggest that proximal tubular shedding of ACE2 may increase in diabetes, which could enhance degradation of Ang II in the tubular lumen, and increase levels of Ang-(1-7).


Subject(s)
Kidney Tubules, Proximal/enzymology , Peptidyl-Dipeptidase A/metabolism , ADAM Proteins/metabolism , ADAM17 Protein , Amino Acid Sequence , Angiotensin II/physiology , Angiotensin-Converting Enzyme 2 , Animals , Catalytic Domain , Cells, Cultured , Culture Media, Conditioned , Glucose/physiology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Molecular Sequence Data , Peptidyl-Dipeptidase A/chemistry , Primary Cell Culture , Proteolysis
16.
PLoS One ; 7(5): e37649, 2012.
Article in English | MEDLINE | ID: mdl-22629438

ABSTRACT

Angiotensin-converting enzyme 2 (ACE2) is expressed in the kidney and may be a renoprotective enzyme, since it converts angiotensin (Ang) II to Ang-(1-7). ACE2 has been detected in urine from patients with chronic kidney disease. We measured urinary ACE2 activity and protein levels in renal transplant patients (age 54 yrs, 65% male, 38% diabetes, n = 100) and healthy controls (age 45 yrs, 26% male, n = 50), and determined factors associated with elevated urinary ACE2 in the patients. Urine from transplant subjects was also assayed for ACE mRNA and protein. No subjects were taking inhibitors of the renin-angiotensin system. Urinary ACE2 levels were significantly higher in transplant patients compared to controls (p = 0.003 for ACE2 activity, and p≤0.001 for ACE2 protein by ELISA or western analysis). Transplant patients with diabetes mellitus had significantly increased urinary ACE2 activity and protein levels compared to non-diabetics (p<0.001), while ACE2 mRNA levels did not differ. Urinary ACE activity and protein were significantly increased in diabetic transplant subjects, while ACE mRNA levels did not differ from non-diabetic subjects. After adjusting for confounding variables, diabetes was significantly associated with urinary ACE2 activity (p = 0.003) and protein levels (p<0.001), while female gender was associated with urinary mRNA levels for both ACE2 and ACE. These data indicate that urinary ACE2 is increased in renal transplant recipients with diabetes, possibly due to increased shedding from tubular cells. Urinary ACE2 could be a marker of renal renin-angiotensin system activation in these patients.


Subject(s)
Diabetes Mellitus/urine , Kidney Transplantation , Peptidyl-Dipeptidase A/urine , Adult , Aged , Angiotensin II/urine , Angiotensin-Converting Enzyme 2 , Female , Humans , Kidney/metabolism , Male , Middle Aged , RNA, Messenger/urine , Renin-Angiotensin System/physiology , Sex Factors
17.
Kidney Int ; 82(3): 292-303, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22475818

ABSTRACT

Angiotensin-converting enzyme 2 (ACE2) degrades angiotensin II to angiotensin-(1-7) and is expressed in podocytes. Here we overexpressed ACE2 in podocytes in experimental diabetic nephropathy using transgenic methods where a nephrin promoter drove the expression of human ACE2. Glomeruli from these mice had significantly increased mRNA, protein, and activity of ACE2 compared to wild-type mice. Male mice were treated with streptozotocin to induce diabetes. After 16 weeks, there was no significant difference in plasma glucose levels between wild-type and transgenic diabetic mice. Urinary albumin was significantly increased in wild-type diabetic mice at 4 weeks, whereas albuminuria in transgenic diabetic mice did not differ from wild-type nondiabetic mice. However, this effect was transient and by 16 weeks both transgenic and nontransgenic diabetic mice had similar rates of proteinuria. Compared to wild-type diabetic mice, transgenic diabetic mice had an attenuated increase in mesangial area, decreased glomerular area, and a blunted decrease in nephrin expression. Podocyte numbers decreased in wild-type diabetic mice at 16 weeks, but were unaffected in transgenic diabetic mice. At 8 weeks, kidney cortical expression of transforming growth factor-ß1 was significantly inhibited in transgenic diabetic mice as compared to wild-type diabetic mice. Thus, the podocyte-specific overexpression of human ACE2 transiently attenuates the development of diabetic nephropathy.


Subject(s)
Diabetic Nephropathies/enzymology , Diabetic Nephropathies/genetics , Peptidyl-Dipeptidase A/genetics , Peptidyl-Dipeptidase A/metabolism , Podocytes/enzymology , Angiotensin-Converting Enzyme 2 , Animals , Blood Pressure , Diabetic Nephropathies/pathology , Diabetic Nephropathies/physiopathology , Glomerular Filtration Rate , Glomerular Mesangium/enzymology , Glomerular Mesangium/pathology , Humans , Male , Mice , Mice, Transgenic , Podocytes/pathology , RNA, Messenger/genetics , RNA, Messenger/metabolism , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Transforming Growth Factor beta1/metabolism , Up-Regulation
18.
J Clin Endocrinol Metab ; 96(11): 3517-24, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21880804

ABSTRACT

CONTEXT: Women with a history of severe preeclampsia are at an increased risk for the development of vascular disease. OBJECTIVE: We hypothesized that abnormalities in the renin-angiotensin system (RAS) may be a predisposing factor. DESIGN AND SETTING: Physiological assessments were conducted at an academic center. PARTICIPANTS: Sixteen women with previous severe preeclampsia (PPE) were compared with nine previously pregnant controls (PPC) and 11 never-pregnant controls (NPC). INTERVENTIONS: Baseline circulating components of the RAS and expression of angiotensin (ANG) II type I (AT1) and type II (AT2) receptors in the skin were assessed along with the response to simulated orthostatic stress using incremental lower-body negative pressure (LBNP: -15, -25, and -40 mm Hg) and a graded ANG II infusion (1 and 3 ng/kg · min). MAIN OUTCOME MEASURES: Response to LBNP and ANG II was evaluated. RESULTS: RAS components were not different between previously pregnant groups, but were decreased compared with NPC subjects. In response to LBNP, there were significant increases in RAS components in all three groups, but the response to this stimulus was significantly lower and delayed in PPE subjects. Despite the blunted rise in circulating RAS mediators in PPE subjects, their blood pressure was maintained in 88% compared with only 33 and 55% in the PPC and NPC groups, respectively (P = 0.014). All three groups responded to the graded ANG II infusion with an increase in blood pressure that was significantly more pronounced in PPE subjects (P = 0.037) correlating with AT1/AT2 receptor expression. CONCLUSIONS: Alterations in the RAS in formerly preeclamptic patients may contribute to future vascular disease.


Subject(s)
Postpartum Period , Pre-Eclampsia/physiopathology , Renin-Angiotensin System/physiology , Stress, Physiological/physiology , Adult , Angiotensins/blood , Female , Humans , Pre-Eclampsia/blood , Pregnancy , Receptor, Angiotensin, Type 1/metabolism , Receptors, Angiotensin/metabolism , Renin/blood , Skin/metabolism , Vascular Diseases/etiology
19.
Am J Physiol Renal Physiol ; 298(6): F1523-32, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20357030

ABSTRACT

Angiotensin-converting enzyme 2 (ACE2) is expressed at high levels in the kidney and converts angiotensin II (ANG II) to ANG-(1-7). We studied the effects of ACE2 inhibition and ANG-(1-7) in the (5/6) nephrectomy ((5/6) Nx) mouse model of chronic kidney disease (CKD). Male FVB mice underwent sham surgery (Sham) or (5/6) Nx and were administered either vehicle, the ACE2 inhibitor MLN-4760 (MLN), the AT(1) receptor antagonist losartan, MLN plus losartan, or ANG-(1-7) for 4 wk. In (5/6) Nx mice with or without MLN, kidney cortical ACE2 protein expression was significantly decreased at 4 wk, compared with Sham. Inhibition of ACE2 caused a decrease in renal cortical ACE2 activity. Kidney cortical ACE expression and activity did not differ between groups of mice. In (5/6) Nx mice treated with MLN, kidney levels of ANG II were significantly increased, compared with Sham. (5/6) Nx induced a mild but insignificant increase in blood pressure (BP), a 50% reduction in FITC-inulin clearance, and a significant increase in urinary albumin excretion. ACE2 inhibition in (5/6) Nx mice did not affect BP or FITC-inulin clearance but significantly increased albuminuria compared with (5/6) Nx alone, an effect reversed by losartan. Treatment of (5/6) Nx mice with ANG-(1-7) increased kidney and plasma levels of ANG-(1-7) but did not alter BP, FITC-inulin clearance, or urinary albumin excretion, and it increased relative mesangial area. These data indicate that kidney ACE2 is downregulated in the early period after (5/6) Nx. Inhibition of ACE2 in (5/6) Nx mice increases albuminuria via an AT(1) receptor-dependent mechanism, independent of BP. In contrast, ANG-(1-7) does not affect albuminuria after (5/6) Nx. We propose that endogenous ACE2 is renoprotective in CKD.


Subject(s)
Angiotensin I/metabolism , Kidney Diseases/enzymology , Kidney/enzymology , Peptide Fragments/metabolism , Peptidyl-Dipeptidase A/metabolism , Albuminuria/enzymology , Albuminuria/physiopathology , Angiotensin I/administration & dosage , Angiotensin II/blood , Angiotensin II Type 1 Receptor Blockers/administration & dosage , Angiotensin-Converting Enzyme 2 , Angiotensin-Converting Enzyme Inhibitors/administration & dosage , Animals , Blood Pressure , Body Weight , Chronic Disease , Disease Models, Animal , Glomerular Filtration Rate , Hematocrit , Imidazoles/administration & dosage , Infusion Pumps , Injections, Subcutaneous , Inulin/analogs & derivatives , Kidney/drug effects , Kidney/pathology , Kidney/physiopathology , Kidney Diseases/drug therapy , Kidney Diseases/pathology , Kidney Diseases/physiopathology , Leucine/administration & dosage , Leucine/analogs & derivatives , Losartan/administration & dosage , Male , Mice , Nephrectomy , Organ Size , Peptide Fragments/administration & dosage , Proto-Oncogene Mas , Proto-Oncogene Proteins/metabolism , Receptor, Angiotensin, Type 1/metabolism , Receptors, G-Protein-Coupled/metabolism , Time Factors
20.
Nephrol Dial Transplant ; 24(6): 1766-73, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19144997

ABSTRACT

BACKGROUND: In the diabetic kidney, stimulation of mitogen-activated protein kinases (MAPKs) leads to extracellular matrix protein synthesis. In the proximal tubule, angiotensin-(1-7) [Ang-(1-7)] blocks activation of MAPKs by angiotensin II. We studied the effect of Ang-(1-7) on signalling responses in LLC-PK(1) cells in normal (5 mM) or high (25 mM) glucose. METHODS: The p38 MAPK was assayed by immunoblot, Src homology 2-containing protein-tyrosine phosphatase-1 (SHP-1) activity was measured after immunoprecipitation, cell protein synthesis was determined by [(3)H]-leucine incorporation and transforming growth factor-beta1 (TGF-beta1), fibronectin and collagen IV were assayed by immunoblots and/or ELISA. RESULTS: High glucose stimulated p38 MAPK. This response was inhibited by Ang-(1-7) in a concentration-dependent fashion, an effect reversed by the receptor Mas antagonist A-779. Ang-(1-7) increased SHP-1 activity, via the receptor Mas. An inhibitor of tyrosine phosphatase, phenylarsine oxide, reversed the inhibitory effect of Ang-(1-7) on high glucose-stimulated p38 MAPK. Ang-(1-7) inhibited high glucose-stimulated protein synthesis, and blocked the stimulatory effect of glucose on TGF-beta1. Conversely, Ang-(1-7) had no effect on glucose-stimulated synthesis of fibronectin or collagen IV. CONCLUSIONS: These data indicate that in proximal tubular cells, binding of Ang-(1-7) to the receptor Mas stimulates SHP-1, associated with the inhibition of glucose-stimulated p38 MAPK. Ang-(1-7) selectively inhibits glucose-stimulated protein synthesis and TGF-beta1. In diabetic nephropathy, Ang-(1-7) may partly counteract the profibrotic effects of high glucose.


Subject(s)
Angiotensin I/pharmacology , Glucose/physiology , Kidney Tubules, Proximal/drug effects , Kidney Tubules, Proximal/metabolism , Peptide Fragments/pharmacology , Protein Tyrosine Phosphatases/metabolism , Animals , Collagen Type IV/metabolism , Diabetic Nephropathies/drug therapy , Diabetic Nephropathies/metabolism , Diabetic Nephropathies/pathology , Enzyme Activation/drug effects , Fibronectins/metabolism , Humans , Kidney Tubules, Proximal/pathology , LLC-PK1 Cells , Protein Tyrosine Phosphatase, Non-Receptor Type 6/metabolism , Signal Transduction/drug effects , Swine , Transforming Growth Factor beta1/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...