Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
1.
J Am Soc Nephrol ; 31(8): 1729-1745, 2020 08.
Article in English | MEDLINE | ID: mdl-32576600

ABSTRACT

BACKGROUND: Progression of CKD in type 2 diabetes, despite dual inhibition of sodium-glucose transporter-2 and the renin-angiotensin system, remains a concern. Bromoindirubin-3'-oxime (BIO), previously reported to promote podocyte survival and regeneration, is a candidate additional drug to elicit renoprotective effects beyond therapy with metformin, ramipril, and empagliflozin (MRE). Evaluating a drug with standard therapeutics more closely mimics the clinical setting than evaluating the drug alone. METHODS: Uninephrectomized BKS-Lepr-/- (db/db) mice treated with or without MRE served as a model of progressive CKD in type 2 diabetes. Mice on or off MRE were randomized to only 4 weeks of add-on BIO or vehicle. The primary end point was slope of GFR (ΔGFR). RESULTS: Four weeks of MRE treatment alone did not affect ΔGFR, but significantly attenuated hyperglycemia, albuminuria, and glomerulosclerosis and increased podocyte filtration slit density, as assessed by STED super-resolution microscopy upon tissue clearing. BIO alone improved albuminuria, podocyte density in superficial and juxtamedullary nephrons, and podocyte filtration slit density. MRE+BIO combination therapy had additive protective effects on ΔGFR, glomerulosclerosis, podocyte density in juxtamedullary nephrons, and filtration slit density. CONCLUSIONS: Add-on treatment with BIO for only 4 weeks attenuates progression of CKD beyond MRE therapy in mice with type 2 diabetes. Additional drug combinations may help to further delay ESKD in type 2 diabetes.


Subject(s)
Benzhydryl Compounds/administration & dosage , Diabetes Mellitus, Type 2/drug therapy , Diabetic Nephropathies/prevention & control , Glucosides/administration & dosage , Indoles/therapeutic use , Metformin/administration & dosage , Oximes/therapeutic use , Ramipril/administration & dosage , Animals , Diabetes Mellitus, Type 2/physiopathology , Diabetic Nephropathies/physiopathology , Disease Progression , Drug Therapy, Combination , Glomerular Filtration Rate/drug effects , Glycogen Synthase Kinase 3 beta/antagonists & inhibitors , Kidney/drug effects , Mice , Podocytes/drug effects
2.
Curr Opin Nephrol Hypertens ; 29(3): 302-309, 2020 05.
Article in English | MEDLINE | ID: mdl-32132388

ABSTRACT

PURPOSE OF REVIEW: Crescents are classical histopathological lesions found in severe forms of rapidly progressive glomerulonephritis, also referred to as crescentic glomerulonephritis (CGN). Crescent formation is a consequence of diverse upstream pathomechanisms and unraveling these mechanisms is of great interest for improving the management of patients affected by CGN. Thus, in this review, we provide an update on the latest insight into the understanding on how crescents develop and how they resolve. RECENT FINDINGS: Cellular crescents develop from activated parietal epithelial cells (PECs) residing along Bowman's capsule and their formation has as a consequence the decline in glomerular filtration rate (GFR). Cellular crescents can be reversible, but when multilevel growth of PECs associate with an epithelial--mesenchymal transition-like change in cell phenotype, fibrous crescents form, and crescents become irreversible also in terms of GFR recovery. Different molecular pathways trigger the activation of PECs and are a prime therapeutics target in CGN. First, crescent formation requires also vascular injury causing ruptures in the glomerular basement membrane that trigger plasmatic coagulation within Bowman's space. This vascular necrosis can be triggered by different upstream mechanisms, such as small vessel vasculitides, immune complex glomerulonephritis, anti-GBM disease, and C3 glomerulonephritis, that all share complement activation but involve diverse upstream immune mechanisms outside the kidney accessible for therapeutic intervention. SUMMARY: Knowing the upstream mechanisms that triggered crescent formation provides a tool for the development of therapeutic interventions for CGN.


Subject(s)
Glomerulonephritis/etiology , Kidney Glomerulus/pathology , Epithelial Cells/physiology , Glomerular Filtration Rate , Glomerulonephritis/pathology , Glomerulonephritis/physiopathology , Humans , Necrosis
3.
Int J Cardiol ; 275: 59-64, 2019 Jan 15.
Article in English | MEDLINE | ID: mdl-30314840

ABSTRACT

OBJECTIVES: The authors sought to evaluate cardiac activity of angiotensin-converting enzyme (ACE) and ACE2 after heart transplantation (HT) and its relation with acute rejection (AR) and chronic allograft vasculopathy (CAV). BACKGROUND: The renin-angiotensin system is altered in heart failure and HT. However, ACE and ACE2 activities in post-HT acute and chronic rejection have not been previously studied. METHODS: HT patients (n = 45) were included when appropriate serial endomyocardial biopsies (EMB) and coronary angiography were available for analysis. In 21 patients, three post-HT time points were selected for CAV study in EMB tissue: basal (0-3 wks), second (2-3 months) and third (4-5 months). At 10 years post-HT, CAV was evaluated by coronary angiography (CA) and patients were grouped by degree of CAV: 0-1, non-CAV (n = 15) and 2-3, CAV (n = 6). For the AR study, 28 HT patients with evidence of one EMB rejection at grade 3 and two EMB grade 1A and/or 1B rejections were selected. RESULTS: Post-HT, ACE2 activity was increased in the CAV group, compared to non-CAV. Patients with AR showed increased ACE, but not ACE2, activity. CONCLUSIONS: Our results suggest that early post-HT cardiac ACE2 activity may have an important role in CAV development. In contrast, ACE activity was increased in AR. The renin-angiotensin system seems to be altered after HT and strategies to balance the system may be useful.


Subject(s)
Graft Rejection/enzymology , Heart Transplantation/adverse effects , Myocardium/enzymology , Peptidyl-Dipeptidase A/metabolism , Acute Disease , Adult , Angiotensin-Converting Enzyme 2 , Biomarkers/metabolism , Biopsy , Chronic Disease , Coronary Angiography , Female , Follow-Up Studies , Graft Rejection/diagnosis , Heart Failure/surgery , Humans , Male , Middle Aged , Myocardium/pathology , Pilot Projects , Prognosis , Retrospective Studies , Severity of Illness Index , Time Factors , Transplantation, Homologous
4.
Front Immunol ; 9: 2173, 2018.
Article in English | MEDLINE | ID: mdl-30319631

ABSTRACT

The long pentraxin 3 (PTX3) exerts a variety of regulatory functions in acute and chronic tissue inflammation. In particular, PTX3 acts as an opsonin for a variety of pathogens and endogenous particles. We hypothesized that PTX3 would exhibit opsonin-like functions toward calcium oxalate crystals, too, and inhibit crystal growth. This process is fundamental in kidney stone disease as well as in hyperoxaluria-related nephrocalcinosis, the paradigmatic cause of chronic kidney disease (CKD) in children with primary hyperoxaluria type I due to genetic defects in oxalate metabolism. Direct effects of PTX3 on calcium oxalate crystals were investigated in chemico by adding recombinant PTX3 to supersaturated calcium and oxalate solutions. PTX3, but not isomolar concentrations of albumin, dose-dependently inhibited crystal growth. In vivo, the PTX3 protein was undetectable in tubular epithelial cells and urine of wild-type mice under physiological conditions. However, its levels increased within 3 weeks of feeding an oxalate-rich diet, an exposure inducing hyperoxaluria-related nephrocalcinosis and CKD in selected mouse strains (male and female C57BL/6N and male Balb/c mice) but not in others (male and female 129SV and CD-1, male and female Balb/c mice). Genetic ablation of ptx3 in nephrocalcinosis un-susceptible B6;129 mice was sufficient to raise the oxalate nephropathy phenotype observed in susceptible strains. We conclude that PTX3 is an endogenous inhibitor of calcium oxalate crystal growth. This mechanism limits hyperoxaluria-related nephrocalcinosis, e.g., in primary or secondary hyperoxaluria, and potentially also in the more prevalent kidney stone disease.


Subject(s)
C-Reactive Protein/immunology , Hyperoxaluria/complications , Nephrocalcinosis/immunology , Renal Insufficiency, Chronic/immunology , Serum Amyloid P-Component/immunology , Animals , C-Reactive Protein/genetics , Calcium Oxalate/immunology , Calcium Oxalate/urine , Disease Models, Animal , Female , Humans , Hyperoxaluria/urine , Kidney Tubules/immunology , Kidney Tubules/pathology , Male , Mice , Mice, 129 Strain , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Transgenic , Nephrocalcinosis/pathology , Nephrocalcinosis/urine , Recombinant Proteins/immunology , Recombinant Proteins/metabolism , Renal Insufficiency, Chronic/pathology , Renal Insufficiency, Chronic/urine , Serum Amyloid P-Component/genetics
5.
Lab Invest ; 98(9): 1237-1249, 2018 09.
Article in English | MEDLINE | ID: mdl-29884907

ABSTRACT

Angiotensin-converting enzyme (ACE) and ACE2 play a critical role in the renin-angiotensin system (RAS) by altering angiotensin II (ANGII) levels, thus governing its deleterious effects. Both enzymes are altered by sex and diabetes, and play an important role in the development of diabetic nephropathy (DN). Importantly, previous evidence in diabetic and ACE2-deficient (ACE2KO) males suggest a sex-dependent crosstalk between renal ACE and ACE2. In the present work, we aimed to study the sex-specific susceptibility to diabetes and direct infusion of ANGII in kidney disease progression, with a special focus on its link to ACE2 and ACE. In our mouse model, ANGII promoted hypertension, albuminuria, reduced glomerular filtration, and glomerular histological alterations. ANGII adverse effects were accentuated by diabetes and ACE2 deficiency, in a sex-dependent fashion: ACE2 deficiency accentuated ANGII-induced hypertension, albuminuria, and glomerular hypertrophy in diabetic females, whereas in diabetic males exacerbated ANGII-mediated glomerular hypertrophy, mesangial expansion, and podocyte loss. At the molecular level, ANGII downregulated renal ACE gene and enzymatic activity levels, as well as renin gene expression in ACE2KO mice. Interestingly, male sex and diabetes accentuated this effect. Here we show sex dimorphism in the severity of diabetes- and ANGII-related renal lesions, and demonstrate that ACE2- and ACE-related compensatory mechanisms are sex-specific. Supporting our previous findings, the modulation and ANGII-mediated crosstalk between ACE2 and ACE in DN progression was more evident in males. This work increases the understanding of the sex-specific role of ACE2 and ACE in DN, reinforcing the necessity of more personalized treatments targeting RAS.


Subject(s)
Angiotensin II/metabolism , Diabetic Nephropathies/metabolism , Peptidyl-Dipeptidase A/metabolism , Angiotensin II/administration & dosage , Angiotensin-Converting Enzyme 2 , Animals , Diabetes Mellitus, Experimental/metabolism , Diabetic Nephropathies/etiology , Diabetic Nephropathies/genetics , Disease Progression , Feedback, Physiological , Female , Fibrosis , Kidney/metabolism , Kidney/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Peptidyl-Dipeptidase A/deficiency , Peptidyl-Dipeptidase A/genetics , Renin-Angiotensin System/physiology , Sex Characteristics
6.
Atherosclerosis ; 253: 135-143, 2016 10.
Article in English | MEDLINE | ID: mdl-27615597

ABSTRACT

BACKGROUND AND AIMS: Circulating Angiotensin Converting Enzyme 2 (ACE2) activity in chronic kidney disease (CKD) patients without previous history of cardiovascular disease (CVD) has been associated with classical risk factors (older age, diabetes and male gender). Furthermore, silent atherosclerosis has been described as a pathological link between CKD and CVD. We analyzed baseline ACE2 activity in non-dialysis CKD stages 3-5 (CKD3-5) patients as a biomarker of renal progression, silent atherosclerosis and CV events after 2 years of follow-up. METHODS: Prospective study of 1458 CKD3-5 subjects without any previous CV event included in the Spanish multicenter NEFRONA study. Association between baseline circulating ACE2 activity and renal parameters, carotid/femoral echography, atheromatous disease, ankle-brachial index, intima-media thickness, need of renal replacement therapy, cardiovascular events and mortality at 24 months of follow-up were analyzed. RESULTS: Patients with an increase in the number of territories with plaques at 24 months showed significantly higher levels of baseline ACE2 activity as compared to stable patients (29.6 (20.6-47.6)RFU/µL/h versus 35.7 (24.5-56), p < 0.001). Multivariate linear regression analysis showed that male gender, pathological ankle-brachial index and progressive silent atherosclerosis defined as an increased number of territories with plaques at 24 months were associated with increased baseline ACE2 activity. Male gender, older age, diabetes, smoking and increased baseline circulating ACE2 were independent predictors of atherosclerosis at 24 months of follow-up. CONCLUSIONS: In CKD3-5 patients, higher circulating ACE2 activity at baseline is associated with higher risk for silent atherosclerosis, suggesting that ACE2 may serve as a biomarker to predict CV risk before CVD is established.


Subject(s)
Atherosclerosis/blood , Biomarkers/blood , Peptidyl-Dipeptidase A/blood , Renal Insufficiency, Chronic/blood , Adult , Aged , Angiotensin-Converting Enzyme 2 , Ankle Brachial Index , Carotid Arteries/pathology , Carotid Intima-Media Thickness , Diabetes Complications/blood , Female , Femoral Artery/pathology , Follow-Up Studies , Glomerular Filtration Rate , Humans , Kidney Function Tests , Male , Middle Aged , Prospective Studies , Renal Insufficiency, Chronic/mortality , Risk Factors , Sensitivity and Specificity
7.
Am J Physiol Renal Physiol ; 310(6): F534-46, 2016 Mar 15.
Article in English | MEDLINE | ID: mdl-26697977

ABSTRACT

Circulating and renal activity of angiotensin-converting enzyme 2 (ACE2) is increased in non-obese diabetic (NOD) mice. Because paricalcitol has been reported to protect against diabetic nephropathy, we investigated the role of paricalcitol in modulating ACE2 in these mice. In addition, renal ADAM17, a metalloprotease implied in ACE2 shedding, was assessed. NOD female and non-diabetic control mice were studied for 21 days after diabetes onset and divided into various treatment groups. Diabetic animals received either vehicle; 0.4 or 0.8 µg/kg paricalcitol, aliskiren, or a combination of paricalcitol and aliskiren. We then studied the effect of paricalcitol on ACE2 expression in proximal tubular epithelial cells. Paricalcitol alone or in combination with aliskiren resulted in significantly reduced circulating ACE2 activity in NOD mice but there were no changes in urinary albumin excretion. Serum renin activity was significantly decreased in mice that received aliskiren but no effect was found when paricalcitol was used alone. Renal content of ADAM17 was significantly decreased in animals that received a high dose of paricalcitol. Renal and circulating oxidative stress (quantified by plasma H2O2 levels and immunolocalization of nitrotyrosine) were reduced in high-dose paricalcitol-treated mice compared with non-treated diabetic mice. In culture, paricalcitol incubation resulted in a significant increase in ACE2 expression compared with nontreated cells. In NOD mice with type 1 diabetes, paricalcitol modulates ACE2 activity, ADAM17, and oxidative stress renal content independently from the glycemic profile and urinary albumin excretion. In tubular cells, paricalcitol may modulate ACE2 by blocking its shedding. In the early stage of diabetic nephropathy, paricalcitol treatment counterbalances the effect of diabetes on circulating ACE2 activity. Our results suggest that additional use of paricalcitol may be beneficial in treating patients with diabetes under standard therapeutic strategies.


Subject(s)
ADAM Proteins/metabolism , Diabetic Nephropathies/prevention & control , Ergocalciferols/therapeutic use , Kidney/drug effects , Peptidyl-Dipeptidase A/blood , ADAM17 Protein , Angiotensin-Converting Enzyme 2 , Animals , Blood Pressure , Diabetes Mellitus, Experimental , Drug Evaluation, Preclinical , Ergocalciferols/pharmacology , Female , Kidney/metabolism , Mice, Inbred NOD , Oxidative Stress/drug effects , Proteinuria/prevention & control , Random Allocation , Renin/metabolism
8.
J Clin Med ; 4(6): 1171-92, 2015 May 25.
Article in English | MEDLINE | ID: mdl-26239552

ABSTRACT

Diabetic kidney disease (DKD) remains the most common cause of chronic kidney disease and multiple therapeutic agents, primarily targeted at the renin-angiotensin system, have been assessed. Their only partial effectiveness in slowing down progression to end-stage renal disease, points out an evident need for additional effective therapies. In the context of diabetes, endothelin-1 (ET-1) has been implicated in vasoconstriction, renal injury, mesangial proliferation, glomerulosclerosis, fibrosis and inflammation, largely through activation of its endothelin A (ETA) receptor. Therefore, endothelin receptor antagonists have been proposed as potential drug targets. In experimental models of DKD, endothelin receptor antagonists have been described to improve renal injury and fibrosis, whereas clinical trials in DKD patients have shown an antiproteinuric effect. Currently, its renoprotective effect in a long-time clinical trial is being tested. This review focuses on the localization of endothelin receptors (ETA and ETB) within the kidney, as well as the ET-1 functions through them. In addition, we summarize the therapeutic benefit of endothelin receptor antagonists in experimental and human studies and the adverse effects that have been described.

9.
Nephrol Dial Transplant ; 30(7): 1176-85, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25813276

ABSTRACT

BACKGROUND: Patients with cardiovascular (CV) disease have an increased circulating angiotensin-converting enzyme 2 (ACE2) activity, but there is little information about changes in ACE2 in chronic kidney disease (CKD) patients without history of CV disease. We examined circulating ACE2 activity in CKD patients at stages 3-5 (CKD3-5) and in dialysis (CKD5D) without any history of CV disease. METHODS: Circulating ACE2 activity was measured in human ethylenediamine-tetraacetic acid (EDTA)-plasma samples from the NEFRONA study (n = 2572): control group (CONT) (n = 568), CKD3-5 (n = 1458) and CKD5D (n = 546). Different clinical and analytical variables such as gender; age; history of diabetes mellitus (DM), dyslipidemia and hypertension; glycaemic, renal, lipid and anaemia profiles; vitamin D analogues treatment and antihypertensive treatments (angiotensin-converting enzyme inhibitor and angiotensin receptor blockade) were analysed. Circulating ACE2 and ACE activities were measured using modified fluorimetric assay for EDTA-plasma samples, where zinc chloride was added to recover enzymatic activity. RESULTS: In CKD3-5 and CKD5D, significant decrease in circulating ACE2 activity was observed when compared with CONT, but no differences were found between CKD3-5 and CKD5 when performing paired case-control studies. By multivariate linear regression analysis, male gender and advanced age were identified as independent predictors of ACE2 activity in all groups. Diabetes was identified as independent predictor of ACE2 activity in CKD3-5. Significant increase in the activity of circulating ACE was found in CKD3-5 and CKD5D when compared with CONT and in CKD5D when compared with CKD3-5. By multiple regression analysis, female gender and younger age were identified as independent predictors of ACE activity in CONT and CKD3-5. Diabetes was also identified as an independent predictor of ACE activity in CKD3-5 patients. CONCLUSIONS: Circulating ACE2 and ACE activities can be measured in human EDTA-plasma samples with zinc added to recover enzymatic activity. In a CKD population without previous history of CV disease, ACE2 activity from human EDTA-plasma samples directly correlated with the classical CV risk factors namely older age, diabetes and male gender. Our data suggest that circulating ACE2 is altered in CKD patients at risk for CV event.


Subject(s)
Cardiovascular Diseases/diagnosis , Peptidyl-Dipeptidase A/blood , Renal Insufficiency, Chronic/complications , Angiotensin-Converting Enzyme 2 , Biomarkers/blood , Cardiovascular Diseases/blood , Cardiovascular Diseases/etiology , Case-Control Studies , Diabetes Mellitus/physiopathology , Female , Humans , Hypertension/physiopathology , Male , Middle Aged , Renal Dialysis , Renal Insufficiency, Chronic/pathology
SELECTION OF CITATIONS
SEARCH DETAIL
...