Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
Add more filters










Publication year range
1.
J Biol Chem ; 297(3): 101079, 2021 09.
Article in English | MEDLINE | ID: mdl-34391780

ABSTRACT

Phosphorylation (activation) and dephosphorylation (deactivation) of the slit diaphragm proteins NEPHRIN and NEPH1 are critical for maintaining the kidney epithelial podocyte actin cytoskeleton and, therefore, proper glomerular filtration. However, the mechanisms underlying these events remain largely unknown. Here we show that NEPHRIN and NEPH1 are novel receptor proteins for hepatocyte growth factor (HGF) and can be phosphorylated independently of the mesenchymal epithelial transition receptor in a ligand-dependent fashion through engagement of their extracellular domains by HGF. Furthermore, we demonstrate SH2 domain-containing protein tyrosine phosphatase-2-dependent dephosphorylation of these proteins. To establish HGF as a ligand, purified baculovirus-expressed NEPHRIN and NEPH1 recombinant proteins were used in surface plasma resonance binding experiments. We report high-affinity interactions of NEPHRIN and NEPH1 with HGF, although NEPHRIN binding was 20-fold higher than that of NEPH1. In addition, using molecular modeling we constructed peptides that were used to map specific HGF-binding regions in the extracellular domains of NEPHRIN and NEPH1. Finally, using an in vitro model of cultured podocytes and an ex vivo model of Drosophila nephrocytes, as well as chemically induced injury models, we demonstrated that HGF-induced phosphorylation of NEPHRIN and NEPH1 is centrally involved in podocyte repair. Taken together, this is the first study demonstrating a receptor-based function for NEPHRIN and NEPH1. This has important biological and clinical implications for the repair of injured podocytes and the maintenance of podocyte integrity.


Subject(s)
Hepatocyte Growth Factor/metabolism , Membrane Proteins/metabolism , Animals , Cell Line , Glomerular Filtration Rate/physiology , Hepatocyte Growth Factor/physiology , Humans , Intercellular Junctions/metabolism , Kidney/pathology , Kidney Glomerulus/metabolism , Membrane Proteins/genetics , Mice , Peptides/metabolism , Phosphorylation , Podocytes/metabolism , Protein Binding/physiology , Signal Transduction/physiology
2.
Am J Physiol Renal Physiol ; 319(1): F63-F75, 2020 07 01.
Article in English | MEDLINE | ID: mdl-32463726

ABSTRACT

Diuretics and renin-angiotensin system blockers are often insufficient to control the blood pressure (BP) in salt-sensitive (SS) subjects. Abundant data support the proposal that the level of atrial natriuretic peptide may correlate with the pathogenesis of SS hypertension. We hypothesized here that increasing atrial natriuretic peptide levels with sacubitril, combined with renin-angiotensin system blockage by valsartan, can be beneficial for alleviation of renal damage in a model of SS hypertension, the Dahl SS rat. To induce a BP increase, rats were challenged with a high-salt 4% NaCl diet for 21 days, and chronic administration of vehicle or low-dose sacubitril and/or valsartan (75 µg/day each) was performed. Urine flow, Na+ excretion, and water consumption were increased on the high-salt diet compared with the starting point (0.4% NaCl) in all groups but remained similar among the groups at the end of the protocol. Upon salt challenge, we observed a mild decrease in systolic BP and urinary neutrophil gelatinase-associated lipocalin levels (indicative of alleviated tubular damage) in the valsartan-treated groups. Sacubitril, as well as sacubitril/valsartan, attenuated the glomerular filtration rate decline induced by salt. Alleviation of protein cast formation and lower renal medullary fibrosis were observed in the sacubitril/valsartan- and valsartan-treated groups, but not when sacubitril alone was administered. Interestingly, proteinuria was mildly mitigated only in rats that received sacubitril/valsartan. Further studies of the effects of sacubitril/valsartan in the setting of SS hypertension, perhaps involving a higher dose of the drug, are warranted to determine if it can interfere with the progression of the disease.


Subject(s)
Aminobutyrates/administration & dosage , Angiotensin Receptor Antagonists/administration & dosage , Blood Pressure/drug effects , Hypertension/drug therapy , Kidney Diseases/drug therapy , Tetrazoles/administration & dosage , Valsartan/administration & dosage , Aminobutyrates/therapeutic use , Angiotensin Receptor Antagonists/therapeutic use , Animals , Biphenyl Compounds , Drug Combinations , Hypertension/physiopathology , Kidney Diseases/physiopathology , Male , Rats , Rats, Inbred Dahl , Sodium Chloride, Dietary , Tetrazoles/therapeutic use , Valsartan/therapeutic use
3.
Kidney360 ; 1(3): 179-190, 2020 Mar.
Article in English | MEDLINE | ID: mdl-33490963

ABSTRACT

BACKGROUND: Autosomal dominant polycystic kidney disease is caused by genetic mutations in PKD1 or PKD2. Macrophages and their associated inflammatory cytokines promote cyst progression; however, transcription factors within macrophages that control cytokine production and cystic disease are unknown. METHODS: In these studies, we used conditional Pkd1 mice to test the hypothesis that macrophage-localized interferon regulatory factor-5 (IRF5), a transcription factor associated with production of cyst-promoting cytokines (TNFα, IL-6), is required for accelerated cyst progression in a unilateral nephrectomy (1K) model. Analyses of quantitative real-time PCR (qRT-PCR) and flow-cytometry data 3 weeks post nephrectomy, a time point before the onset of severe cystogenesis, indicate an accumulation of inflammatory infiltrating and resident macrophages in 1K Pkd1 mice compared with controls. qRT-PCR data from FACS cells at this time demonstrate that macrophages from 1K Pkd1 mice have increased expression of Irf5 compared with controls. To determine the importance of macrophage-localized Irf5 in cyst progression, we injected scrambled or IRF5 antisense oligonucleotide (ASO) in 1K Pkd1 mice and analyzed the effect on macrophage numbers, cytokine production, and renal cystogenesis 6 weeks post nephrectomy. RESULTS: Analyses of qRT-PCR and IRF5 ASO treatment significantly reduced macrophage numbers, Irf5 expression in resident-but not infiltrating-macrophages, and the severity of cystic disease. In addition, IRF5 ASO treatment in 1K Pkd1 mice reduced Il6 expression in resident macrophages, which was correlated with reduced STAT3 phosphorylation and downstream p-STAT3 target gene expression. CONCLUSIONS: These data suggest that Irf5 promotes inflammatory cytokine production in resident macrophages resulting in accelerated cystogenesis.


Subject(s)
Polycystic Kidney Diseases , Polycystic Kidney, Autosomal Dominant , Animals , Interferon Regulatory Factors/genetics , Kidney/metabolism , Macrophages/metabolism , Mice , Polycystic Kidney Diseases/metabolism , Polycystic Kidney, Autosomal Dominant/genetics
4.
Kidney Int ; 96(3): 656-673, 2019 09.
Article in English | MEDLINE | ID: mdl-31262488

ABSTRACT

Podocytes have limited ability to recover from injury. Here, we demonstrate that increased mitochondrial biogenesis, to meet the metabolic and energy demand of a cell, accelerates podocyte recovery from injury. Analysis of events induced during podocyte injury and recovery showed marked upregulation of peroxisome proliferator-activated receptor-γ coactivator-1α (PGC-1α), a transcriptional co-activator of mitochondrial biogenesis, and key components of the mitochondrial electron transport chain. To evaluate our hypothesis that increasing mitochondrial biogenesis enhanced podocyte recovery from injury, we treated injured podocytes with formoterol, a potent, specific, and long-acting ß2-adrenergic receptor agonist that induces mitochondrial biogenesis in vitro and in vivo. Formoterol increased mitochondrial biogenesis and restored mitochondrial morphology and the injury-induced changes to the organization of the actin cytoskeleton in podocytes. Importantly, ß2-adrenergic receptors were found to be present on podocyte membranes. Their knockdown attenuated formoterol-induced mitochondrial biogenesis. To determine the potential clinical relevance of these findings, mouse models of acute nephrotoxic serum nephritis and chronic (Adriamycin [doxorubicin]) glomerulopathy were used. Mice were treated with formoterol post-injury when glomerular dysfunction was established. Strikingly, formoterol accelerated the recovery of glomerular function by reducing proteinuria and ameliorating kidney pathology. Furthermore, formoterol treatment reduced cellular apoptosis and increased the expression of the mitochondrial biogenesis marker PGC-1α and multiple electron transport chain proteins. Thus, our results support ß2-adrenergic receptors as novel therapeutic targets and formoterol as a therapeutic compound for treating podocytopathies.


Subject(s)
Adrenergic beta-2 Receptor Agonists/pharmacology , Formoterol Fumarate/pharmacology , Glomerulonephritis/drug therapy , Mitochondria/drug effects , Podocytes/drug effects , Adrenergic beta-2 Receptor Agonists/therapeutic use , Animals , Apoptosis/drug effects , Cell Line , Disease Models, Animal , Doxorubicin/toxicity , Formoterol Fumarate/therapeutic use , Gene Knockdown Techniques , Glomerulonephritis/chemically induced , Glomerulonephritis/pathology , Humans , Mice , Mitochondria/metabolism , Organelle Biogenesis , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/metabolism , Podocytes/cytology , Podocytes/pathology , Receptors, Adrenergic, beta-2/genetics , Receptors, Adrenergic, beta-2/metabolism , Signal Transduction
5.
Kidney Int ; 96(1): 139-158, 2019 07.
Article in English | MEDLINE | ID: mdl-31097328

ABSTRACT

Transforming growth factor-ß (TGF-ß) is known to play a critical role in the pathogenesis of many progressive podocyte diseases. However, the molecular mechanisms regulating TGF-ß signaling in podocytes remain unclear. Using a podocyte-specific myosin (Myo)1c knockout, we demonstrate whether Myo1c is critical for TGF-ß-signaling in podocyte disease pathogenesis. Specifically, podocyte-specific Myo1c knockout mice were resistant to fibrotic injury induced by Adriamycin or nephrotoxic serum. Further, loss of Myo1c also protected from injury in the TGF-ß-dependent unilateral ureteral obstruction mouse model of renal interstitial fibrosis. Mechanistic analyses showed that loss of Myo1c significantly blunted TGF-ß signaling through downregulation of canonical and non-canonical TGF-ß pathways. Interestingly, nuclear rather than the cytoplasmic Myo1c was found to play a central role in controlling TGF-ß signaling through transcriptional regulation. Differential expression analysis of nuclear Myo1c-associated gene promoters showed that nuclear Myo1c targeted the TGF-ß responsive gene growth differentiation factor (GDF)-15 and directly bound to the GDF-15 promoter. Importantly, GDF15 was found to be involved in podocyte pathogenesis, where GDF15 was upregulated in glomeruli of patients with focal segmental glomerulosclerosis. Thus, Myo1c-mediated regulation of TGF-ß-responsive genes is central to the pathogenesis of podocyte injury. Hence, inhibiting this process may have clinical application in treating podocytopathies.


Subject(s)
Growth Differentiation Factor 15/genetics , Kidney Diseases/pathology , Myosin Type I/metabolism , Podocytes/pathology , Signal Transduction/genetics , Transforming Growth Factor beta/metabolism , Animals , Disease Models, Animal , Doxorubicin/toxicity , Female , Fibrosis , Gene Expression Regulation , Humans , Kidney Diseases/chemically induced , Male , Mice , Mice, Knockout , Myosin Type I/genetics , Podocytes/drug effects , Promoter Regions, Genetic , Transcription, Genetic
6.
Am J Physiol Renal Physiol ; 314(2): F210-F218, 2018 02 01.
Article in English | MEDLINE | ID: mdl-29021226

ABSTRACT

The intrarenal renin angiotensin system (RAS) is activated in polycystic kidney disease. We have recently shown in the Pkd1 mouse that Gen 2 antisense oligonucleotide (ASO), which suppresses angiotensinogen (Agt) synthesis, is efficacious in slowing kidney cyst formation compared with lisinopril. The aim of this current study was to determine 1) if unilateral nephrectomy accelerates cystogenesis in Pkd1 mice (as previously shown in cilia knockout mice) and 2) whether Agt ASO can slow the progression in this accelerated cystic mouse model. Adult Pkd1 conditional floxed allele mice expressing cre were administered tamoxifen, resulting in global knockout of Pkd1. Three weeks after tamoxifen injection, mice underwent left unilateral nephrectomy. Mice were then treated with Agt ASO (75 mg/kg per week) or aliskiren (20 mg/kg per day)+Agt ASO or control for 8 wk. Unilateral nephrectomy accelerated kidney cyst formation compared with nonnephrectomized mice. Both Agt ASO and Aliskiren+Agt ASO treatments significantly reduced plasma and urinary Agt levels. Blood pressure was lowest in Aliskiren+Agt ASO mice among all treatment groups, and the control group had the highest blood pressure. All mice developed significant kidney cysts at 8 wk after nephrectomy, but Agt ASO and Aliskiren+Agt ASO groups had fewer kidney cysts than controls. Renal pAkt, pS6 levels, and apoptosis were significantly suppressed in those receiving Agt ASO compared with controls. These results indicate that suppressing Agt using an ASO slowed the progression of accelerated cystic kidney disease induced by unilateral nephrectomy in Pkd1 mice by suppressing intrarenal RAS, mammalian target of rapamycin pathway, and cell proliferation.


Subject(s)
Amides/pharmacology , Angiotensinogen/metabolism , Fumarates/pharmacology , Kidney/drug effects , Polycystic Kidney, Autosomal Dominant/prevention & control , Renin-Angiotensin System/drug effects , Renin/antagonists & inhibitors , TRPP Cation Channels/metabolism , Angiotensinogen/genetics , Animals , Apoptosis/drug effects , Cell Proliferation/drug effects , Disease Models, Animal , Disease Progression , ErbB Receptors/metabolism , Female , Genetic Predisposition to Disease , Kidney/metabolism , Kidney/pathology , Male , Mice, Knockout , Nephrectomy , Oligonucleotides, Antisense/genetics , Oligonucleotides, Antisense/metabolism , Phenotype , Polycystic Kidney, Autosomal Dominant/genetics , Polycystic Kidney, Autosomal Dominant/metabolism , Polycystic Kidney, Autosomal Dominant/pathology , Renin/metabolism , Renin-Angiotensin System/genetics , TOR Serine-Threonine Kinases/metabolism , TRPP Cation Channels/deficiency , TRPP Cation Channels/genetics , Time Factors
7.
J Am Soc Nephrol ; 28(7): 2119-2132, 2017 Jul.
Article in English | MEDLINE | ID: mdl-28202497

ABSTRACT

Aminopeptidase A (APA) is expressed in glomerular podocytes and tubular epithelia and metabolizes angiotensin II (AngII), a peptide known to promote glomerulosclerosis. In this study, we tested whether APA expression changes in response to progressive nephron loss or whether APA exerts a protective role against glomerular damage and during AngII-mediated hypertensive kidney injury. At advanced stages of FSGS, fawn-hooded hypertensive rat kidneys exhibited distinctly increased APA staining in areas of intact glomerular capillary loops. Moreover, BALB/c APA-knockout (KO) mice injected with a nephrotoxic serum showed persistent glomerular hyalinosis and albuminuria 96 hours after injection, whereas wild-type controls achieved virtually full recovery. We then tested the effect of 4-week infusion of AngII (400 ng/kg per minute) in APA-KO and wild-type mice. Although we observed no significant difference in achieved systolic BP, AngII-treated APA-KO mice developed a significant rise in albuminuria not observed in AngII-treated wild-type mice along with increased segmental and global sclerosis and/or collapse of juxtamedullary glomeruli, microcystic tubular dilation, and tubulointerstitial fibrosis. In parallel, AngII treatment significantly increased the kidney AngII content and attenuated the expression of podocyte nephrin in APA-KO mice but not in wild-type controls. These data show that deficiency of APA increases susceptibility to glomerular injury in BALB/c mice. The augmented AngII-mediated kidney injury observed in association with increased intrarenal AngII accumulation in the absence of APA suggests a protective metabolizing role of APA in AngII-mediated glomerular diseases.


Subject(s)
Glutamyl Aminopeptidase/deficiency , Kidney Diseases/enzymology , Kidney Diseases/etiology , Kidney Glomerulus , Animals , Disease Susceptibility , Male , Mice , Mice, Knockout , Rats
8.
Article in English | MEDLINE | ID: mdl-31528834

ABSTRACT

BACKGROUND: Chronic in-center hemodialysis (HD) patients may experience more morbidity and mortality after the weekend. Since our Veterans Administration Hospital HD unit is closed on the weekend, non-traditional HD schedules were created. Some schedules contained a 4-day weekend compared to the usual 3-day weekend. We hypothesized that there are more frequent cardiovascular events (CVEs) and higher mortality after longer interdialytic intervals. METHODS: Patients (n=85) were placed on HD schedules as they became available. The usual interdialytic interval group consisted of patients dialyzing on Mon-Wed-Fri or Mon-Tue-Fri (longest interdialytic gap 3 days, n=29), and the long interdialytic interval group consisted of patients dialyzing on Mon-Wed-Thu, Mon-Tue-Thu, Tue-Wed-Fri, or Tue-Thu-Fri (longest interdialytic gap 4 days, n=56). RESULTS: All-cause mortality was not different between groups, and CVEs occurred more frequently in the usual interdialytic interval group (maybe due to higher mean potassium and phosphorus concentrations). However, within each group, a similar pattern of CVE occurrence as a function of time after dialysis was observed. Compared to CVEs occurring during the 2 days after HD (the lowest frequency), CVEs occurred 2-3 times more frequently during and immediately after HD and 5-7 times more frequently during the third and fourth days after HD. The greatest risk of CVE occurred during the fourth day after HD, which exists only in the long interdialytic interval group. CONCLUSION: In chronic HD patients, CVEs are most likely to occur after the longest interdialytic intervals.

9.
Physiol Rep ; 3(5)2015 May.
Article in English | MEDLINE | ID: mdl-25999403

ABSTRACT

The mechanism for early hypertension in polycystic kidney disease (PKD) has not been elucidated. One potential pathway that may contribute to the elevation in blood pressure in PKD is the activation of the intrarenal renin-angiotensin-system (RAS). For example, it has been shown that kidney cyst and cystic fluid contains renin, angiotensin II (AngII), and angiotensinogen (Agt). Numerous studies suggest that ciliary dysfunction plays an important role in PKD pathogenesis. However, it is unknown whether the primary cilium affects the intrarenal RAS in PKD. The purpose of this study was to determine whether loss of cilia or polycystin 1 (PC1) increases intrarenal RAS in mouse model of PKD. Adult Ift88 and Pkd1 conditional floxed allele mice with or without cre were administered tamoxifen to induce global knockout of the gene. Three months after tamoxifen injection, kidney tissues were examined by histology, immunofluorescence, western blot, and mRNA to assess intrarenal RAS components. SV40 immortalized collecting duct cell lines from hypomorphic Ift88 mouse were used to assess intrarenal RAS components in collecting duct cells. Mice without cilia and PC1 demonstrated increased kidney cyst formation, systolic blood pressure, prorenin, and kidney and urinary angiotensinogen levels. Interestingly immunofluorescence study of the kidney revealed that the prorenin receptor was localized to the basolateral membrane of principal cells in cilia (-) but not in cilia (+) kidneys. Collecting duct cAMP responses to AngII administration was greater in cilia (-) vs. cilia (+) cells indicating enhanced intrarenal RAS activity in the absence of cilia. These data suggest that in the absence of cilia or PC1, there is an upregulation of intrarenal RAS components and activity, which may contribute to elevated blood pressure in PKD.

10.
Am J Physiol Renal Physiol ; 309(1): F79-87, 2015 Jul 01.
Article in English | MEDLINE | ID: mdl-25904703

ABSTRACT

In polycystic kidney disease (PKD), the rate of cyst formation and disease progression is highly variable. The lack of predictability in disease progression may be due to additional environmental factors or pathophysiological processes called "third hits." Diabetes is a growing epidemic, and recent studies suggest that PKD patients may be at an increased risk for this disease. We sought to determine if hyperglycemia enhances the initiation and rate of cystogenesis. Tamoxifen was administered to adult Ift88 conditional floxed allele mice to induce cilia loss in the presence of Cre. Subsequent administration of streptozotocin resulted in equivalent hyperglycemia in cilia(+) and cilia(-) mice. Hyperglycemia with loss of cilia increased the rate of cyst formation and cell proliferation. Structural and functional alterations in the kidney, including focal glomerular foot process effacement, interstitial inflammation, formation of primitive renal tubules, polyuria, and increased proteinuria, were also observed in hyperglycemic cilia(-) mice. Gene array analysis indicated enhanced Wnt and epithelial-to-mesenchymal transition signaling in the kidney of hyperglycemic cilia(-) mice. These data show that hyperglycemia, in the absence of cilia, results in renal structural and functional damage and accelerates cystogenesis, suggesting that diabetes is a risk factor in the progression of PKD.


Subject(s)
Hyperglycemia/complications , Kidney/pathology , Polycystic Kidney Diseases/etiology , Animals , Cell Proliferation , Epithelial-Mesenchymal Transition , Female , Hemodynamics , Hyperglycemia/pathology , Hyperglycemia/physiopathology , Kidney Function Tests , Male , Mice, Knockout , Polycystic Kidney Diseases/pathology , Random Allocation , Wnt Proteins/metabolism
11.
Mol Endocrinol ; 29(6): 896-908, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25871850

ABSTRACT

Angiotensin II (AngII) plays a critical role in the regulation of vascular tone and blood pressure mainly via regulation of Ca(2+) mobilization. Several reports have implicated sphingosine kinase 1 (SK1)/sphingosine 1-phosphate (S1P) in the mobilization of intracellular Ca(2+) through a yet-undefined mechanism. Here we demonstrate that AngII-induces biphasic calcium entry in vascular smooth muscle cells, consisting of an immediate peak due to inositol tris-phosphate-dependent release of intracellular calcium, followed by a sustained transmembrane Ca(2+) influx through store-operated calcium channels (SOCs). Inhibition of SK1 attenuates the second phase of transmembrane Ca(2+) influx, suggesting a role for SK1 in AngII-dependent activation of SOC. Intracellular S1P triggers SOC-dependent Ca(2+) influx independent of S1P receptors, whereas external application of S1P stimulated S1P receptor-dependent Ca(2+) influx that is insensitive to inhibitors of SOCs, suggesting that the SK1/S1P axis regulates store-operated calcium entry via intracellular rather than extracellular actions. Genetic deletion of SK1 significantly inhibits both the acute hypertensive response to AngII in anaesthetized SK1 knockout mice and the sustained hypertensive response to continuous infusion of AngII in conscious animals. Collectively these data implicate SK1 as the missing link that connects the angiotensin AT1A receptor to transmembrane Ca(2+) influx and identify SOCs as a potential intracellular target for SK1.


Subject(s)
Calcium Channels/metabolism , Calcium/metabolism , Hypertension/enzymology , Hypertension/pathology , Phosphotransferases (Alcohol Group Acceptor)/antagonists & inhibitors , Angiotensin II , Animals , Blood Pressure/drug effects , Cell Membrane/drug effects , Cell Membrane/metabolism , Chronic Disease , Enzyme Activation/drug effects , Gene Deletion , HEK293 Cells , Humans , Hypertension/physiopathology , Lysophospholipids/biosynthesis , Lysophospholipids/pharmacology , Male , Mice, Inbred C57BL , Phosphotransferases (Alcohol Group Acceptor)/metabolism , Rats, Sprague-Dawley , Receptors, Lysosphingolipid/metabolism , Sphingosine/analogs & derivatives , Sphingosine/biosynthesis , Sphingosine/pharmacology
12.
PLoS One ; 9(10): e110083, 2014.
Article in English | MEDLINE | ID: mdl-25337950

ABSTRACT

Unopposed angiotensin (Ang) II-mediated cellular effects may lead to progressive glomerulosclerosis. While Ang-II can be locally generated in the kidneys, we previously showed that glomerular podocytes primarily convert Ang-I, the precursor of Ang-II, to Ang-(1-7) and Ang-(2-10), peptides that have been independently implicated in biological actions opposing those of Ang-II. Therefore, we hypothesized that Ang-(1-7) and Ang-(2-10) could be renoprotective in the fawn-hooded hypertensive rat, a model of focal segmental glomerulosclerosis. We evaluated the ability of 8-12 week-long intravenous administration of either Ang-(1-7) or Ang-(2-10) (100-400 ng/kg/min) to reduce glomerular injury in uni-nephrectomized fawn-hooded hypertensive rats, early or late in the disease. Vehicle-treated rats developed hypertension and lesions of focal segmental glomerulosclerosis. No reduction in glomerular damage was observed, as measured by either 24-hour urinary protein excretion or histological examination of glomerulosclerosis, upon Ang-(1-7) or Ang-(2-10) administration, regardless of peptide dose or disease stage. On the contrary, when given at 400 ng/kg/min, both peptides induced a further increase in systolic blood pressure. Content of Ang peptides was measured by parallel reaction monitoring in kidneys harvested at sacrifice. Exogenous administration of Ang-(1-7) and Ang-(2-10) did not lead to a significant increase in their corresponding intrarenal levels. However, the relative abundance of Ang-(1-7) with respect to Ang-II was increased in kidney homogenates of Ang-(1-7)-treated rats. We conclude that chronic intravenous administration of Ang-(1-7) or Ang-(2-10) does not ameliorate glomerular damage in a rat model of focal segmental glomerulosclerosis and may induce a further rise in blood pressure, potentially aggravating glomerular injury.


Subject(s)
Angiotensin II/administration & dosage , Angiotensin I/administration & dosage , Glomerulosclerosis, Focal Segmental/drug therapy , Hypertension/drug therapy , Peptide Fragments/administration & dosage , Podocytes/drug effects , Animals , Drug Administration Schedule , Glomerulosclerosis, Focal Segmental/complications , Glomerulosclerosis, Focal Segmental/metabolism , Glomerulosclerosis, Focal Segmental/pathology , HEK293 Cells , Humans , Hypertension/complications , Hypertension/metabolism , Hypertension/pathology , Injections, Intravenous , Male , Nephrectomy , Podocytes/metabolism , Podocytes/pathology , Radioligand Assay , Rats , Treatment Failure
13.
Nephrology (Carlton) ; 15(5): 544-8, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20649874

ABSTRACT

BACKGROUND: We tested the hypothesis that patterns of serum creatinine concentrations (S-cr) prior to percutaneous renal biopsy (PRB) predict the utility of PRB in safely making renal diagnoses, revealing treatable disease, and altering therapy in chronic kidney disease patients. METHODS: PRB specimens (170 patients) were assigned to 1 of 5 groups: S-cr never greater than 0.11 mM for at least 6 months prior to PRB (Group 1); S-cr greater than 0.11 mM but less than 0.18 mM during the 6 months prior to PRB (Groups 2); S-cr less than 0.18 mM during the 6 months prior to PRB but greater than 0.18 mM prior to these 6 months (Group 3); S-cr greater than 0.18 mM for less than 6 months prior to PRB (Group 4); S-cr greater than 0.18 mM for more than 6 months prior to PRB (Group 5). RESULTS: Histopathology chronicity score (0-9) increased with increasing group number: 2.1 (Group 1); 4.4 (Group 2); 4.5 (Group 3); 5.4 (Group 4); 7.0 (Group 5). Post-PRB bleeding was more common with increasing group number. New therapy was instituted after PRB most frequently in Group 4 (62%) and least frequently in Group 5 (24%). CONCLUSION: After more prolonged elevations of S-cr, PRB may be less safe and less likely to reveal treatable disease and opportunities for therapy.


Subject(s)
Biopsy/statistics & numerical data , Kidney Diseases/pathology , Kidney/pathology , Adult , Biomarkers/blood , Biopsy/adverse effects , Chi-Square Distribution , Chronic Disease , Creatinine/blood , Female , Hemorrhage/etiology , Humans , Kidney/metabolism , Kidney Diseases/blood , Kidney Diseases/therapy , Male , Middle Aged , Predictive Value of Tests , Retrospective Studies , Risk Assessment , Severity of Illness Index , South Carolina , Time Factors , Up-Regulation
14.
Hypertension ; 53(5): 790-7, 2009 May.
Article in English | MEDLINE | ID: mdl-19289651

ABSTRACT

Intraglomerular renin-angiotensin system enzyme activities have been examined previously using glomerular lysates and immune-based assays. However, preparation of glomerular extracts compromises the integrity of their anatomic architecture. In addition, antibody-based assays focus on angiotensin (Ang) II detection, ignoring the generation of other Ang I-derived metabolites, some of which may cross-react with Ang II. Therefore, our aim was to examine the metabolism of Ang I in freshly isolated intact glomeruli using matrix-assisted laser desorption ionization time of flight mass spectrometry as an analytic method. Glomeruli from male Sprague-Dawley rats were isolated by sieving and incubated in Krebs buffer in the presence of 1 micromol/L of Ang I for 15 to 90 minutes, with or without various peptidase inhibitors. Peptide sequences were confirmed by matrix-assisted laser desorption ionization time of flight tandem mass spectrometry or linear-trap-quadrupole mass spectrometry. Peaks were quantified using customized valine-(13)C(.15)N-labeled peptides as standards. The most prominent peaks resulting from Ang I cleavage were 899 and 1181 m/z, corresponding with Ang (1-7) and Ang (2-10), respectively. Smaller peaks for Ang II, Ang (1-9), and Ang (3-10) also were detected. The disappearance of Ang I was significantly reduced during inhibition of aminopeptidase A or neprilysin. In contrast, captopril did not alter Ang I degradation. Furthermore, during simultaneous inhibition of aminopeptidase A and neprilysin, the disappearance of Ang I was markedly attenuated compared with all of the other conditions. These results suggest that there is prominent intraglomerular conversion of Ang I to Ang (2-10) and Ang (1-7), mediated by aminopeptidase A and neprilysin, respectively. Formation of these alternative Ang peptides may be critical to counterbalance the local actions of Ang II. Enhancement of these enzymatic activities may constitute potential therapeutic targets for Ang II-mediated glomerular diseases.


Subject(s)
Angiotensin I/metabolism , Kidney Glomerulus/metabolism , Peptide Fragments/metabolism , Animals , Glutamyl Aminopeptidase/antagonists & inhibitors , Glutamyl Aminopeptidase/physiology , Male , Neprilysin/antagonists & inhibitors , Neprilysin/physiology , Rats , Rats, Sprague-Dawley , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization
15.
Am J Hypertens ; 22(2): 221-7, 2009 Feb.
Article in English | MEDLINE | ID: mdl-19039312

ABSTRACT

BACKGROUND: We compared the effects of the sulfhydryl-containing (thiol) antioxidant dithiothreitol (DTT), which disrupts disulfide bonds, on cell signaling through angiotensin II (AngII) Type 1 receptors (AT1Rs) and arginine vasopressin (AVP) V1 receptors (V1Rs). The AT1R contains two extracellular disulfides bonds but its ligand contains none, whereas the V1R contains no extracellular disufides bonds but its ligand contains 1. METHODS: We measured radioligand binding, intracellular calcium responses, and extracellular signal-regulated kinase phosphorylation in cultured rat aortic vascular smooth muscle cells and alterations in urine osmolality in intact rats. RESULTS: Preincubation of cells with DTT, a maneuver designed to target receptor disulfides, resulted in concentration-dependent decreases in specific (125)I-AngII binding to AT1Rs and acute angiotensin-stimulated intracellular calcium mobilization but no decreases in specific (125)I-AVP binding to V1Rs or AVP-stimulated intracellular calcium mobilization. In contrast, preincubation of the ligands with DTT followed by acute exposure to the cells, a maneuver designed to target ligand disulfides, blunted calcium mobilization to AVP robustly but to AngII only minimally. In intact rats, the increase in urine osmolality caused by subcutaneous injection with the AVP analogue desmopressin was significantly diminished when the analogue was preincubated with an excess of DTT. CONCLUSION: DTT inhibits cell signaling to AngII AT1Rs and AVP V1Rs, at least in part through disruption of disulfide linkages, but the pattern of response depends upon whether disulfides of ligand or receptor are targeted.


Subject(s)
Antioxidants/pharmacology , Dithiothreitol/pharmacology , Muscle, Smooth, Vascular/drug effects , Receptor, Angiotensin, Type 1/physiology , Receptors, Vasopressin/physiology , Signal Transduction/drug effects , Animals , Calcium/metabolism , Cells, Cultured , Deamino Arginine Vasopressin/chemistry , Deamino Arginine Vasopressin/pharmacology , Disulfides/metabolism , Diuresis/drug effects , Extracellular Signal-Regulated MAP Kinases/metabolism , Male , Muscle, Smooth, Vascular/cytology , Osmolar Concentration , Rats , Rats, Sprague-Dawley , Receptor, Angiotensin, Type 1/drug effects , Receptors, Vasopressin/drug effects , Urine/physiology
16.
Am J Physiol Renal Physiol ; 295(3): F811-7, 2008 Sep.
Article in English | MEDLINE | ID: mdl-18632797

ABSTRACT

The present study was performed to test the hypothesis that under normal physiological conditions and/or during augmentation of kinin levels, intrarenal kinins act on medullary bradykinin B(2) (BKB(2)) receptors to acutely increase papillary blood flow (PBF) and therefore Na(+) excretion. We determined the effect of acute inner medullary interstitial (IMI) BKB(2) receptor blockade on renal hemodynamics and excretory function in rats fed either a normal (0.23%)- or a low (0.08%)-NaCl diet. For each NaCl diet, two groups of rats were studied. Baseline renal hemodynamic and excretory function were determined during IMI infusion of 0.9% NaCl into the left kidney. The infusion was then either changed to HOE-140 (100 microg.kg(-1).h(-1), treated group) or maintained with 0.9% NaCl (time control group), and the parameters were again determined. In rats fed a normal-salt diet, HOE-140 infusion decreased left kidney Na(+) excretion (urinary Na(+) extraction rate) and fractional Na(+) excretion by 40 +/- 5% and 40 +/- 4%, respectively (P < 0.01), but did not alter glomerular filtration rate, inner medullary blood flow (PBF), or cortical blood flow. In rats fed a low-salt diet, HOE-140 infusion did not alter renal regional hemodynamics or excretory function. We conclude that in rats fed a normal-salt diet, kinins act tonically via medullary BKB(2) receptors to increase Na(+) excretion independent of changes in inner medullary blood flow.


Subject(s)
Kidney Medulla/metabolism , Kinins/metabolism , Receptor, Bradykinin B2/metabolism , Renal Circulation , Sodium/metabolism , Animals , Bradykinin/analogs & derivatives , Bradykinin/pharmacology , Bradykinin B2 Receptor Antagonists , Diet, Sodium-Restricted , Kidney Medulla/blood supply , Kidney Medulla/drug effects , Male , NG-Nitroarginine Methyl Ester/pharmacology , Rats , Rats, Sprague-Dawley , Sodium Chloride, Dietary/administration & dosage
17.
Am J Physiol Renal Physiol ; 291(4): F770-80, 2006 Oct.
Article in English | MEDLINE | ID: mdl-16609153

ABSTRACT

Marine elasmobranchs maintain internal osmolality higher than their external environment, resulting in an osmotic gradient for branchial water uptake. This gradient is markedly increased in low-salinity habitats. The subsequent increase in water uptake presents a challenge to volume homeostasis. The Atlantic stingray is a marine elasmobranch that inhabits a remarkable range of environmental salinities. We hypothesized that the ability of these stingrays to regulate fluid volume in low-salinity environments is due primarily to a renal glomerular and tubular functional reserve. We tested this hypothesis by measuring renal excretory function after a rapid and sustained 50% reduction in the osmolality of the external medium. Atlantic stingrays were maintained in harbor water [control salinity (CS) approximately 850 mosmol/kgH(2)O] for 1 wk. Rays were then either transferred to diluted harbor water [low salinity (LS) approximately 440 mosmol/kgH(2)O] or maintained in CS for a further 24 h. Renal excretory function was markedly higher in the rays subjected to low salinity. Glomerular filtration rate was threefold higher and urine flow rate ninefold higher in the LS group. The clearance of solute-free water was greater, and solute-free water comprised a significantly larger proportion of the urine output for the stingrays transferred to dilute harbor water. We conclude that 1) the kidneys of Atlantic stingrays have a remarkable glomerular and tubular functional reserve, and 2) the marked increase in renal function attenuates the increase in fluid volume when these fish move into low-salinity habitats.


Subject(s)
Kidney/physiology , Sodium Chloride/analysis , Urine/physiology , Acclimatization , Animals , Atlantic Ocean , Fresh Water , Kidney Function Tests , Osmolar Concentration , Seawater , Skates, Fish
18.
Am J Physiol Regul Integr Comp Physiol ; 291(3): R844-53, 2006 Sep.
Article in English | MEDLINE | ID: mdl-16614049

ABSTRACT

The cloning of cDNAs encoding facilitated urea transporters (UTs) from the kidneys of the elasmobranchs indicates that in these fish renal urea reabsorption occurs, at least in part, by passive processes. The previously described elasmobranch urea transporter clones from shark (shUT) and stingray (strUT-1) differ from each other primarily because of the COOH-terminus of the predicted strUT-1 translation product being extended by 51-amino acid residues compared with shUT. Previously, we noted multiple UT transcripts were present in stingray kidney. We hypothesized that a COOH terminally abbreviated UT isoform, homologous to shUT, would also be present in stingray kidney. Therefore, we used 5'/3' rapid amplification of cDNA ends to identify a 3'UTR-variant (strUT-1a) of the cDNA that encodes (strUT-1), as well as three, 3'UTR-variant cDNAs (strUT-2a,b,c) that encode a second phloretin-sensitive, urea transporter (strUT-2). The 5'UTR and the first 1,132 nucleotides of the predicted coding region of the strUT-2 cDNAs are identical to the strUT-1 cDNAs. The remainder of the coding region contains only five novel nucleotides. The strUT-2 cDNAs putatively encode a 379-amino acid protein, the first 377 amino acids identical to strUT-1 plus 2 additional amino acids. We conclude that 1) a second UT isoform is expressed in the Atlantic stingray and that this isoform is similar in size to the UT previously cloned from the kidney of the dogfish shark, and 2) at least five transcripts encoding the 2 stingray UTs are derived from a single gene product through alternative splicing and polyadenylation.


Subject(s)
Kidney/metabolism , Membrane Transport Proteins/genetics , Membrane Transport Proteins/metabolism , Skates, Fish/genetics , Skates, Fish/metabolism , Amino Acid Sequence , Animals , Cloning, Molecular , Gene Expression Regulation , Male , Membrane Transport Proteins/chemistry , Molecular Sequence Data , Urea Transporters
19.
BMC Genet ; 6: 40, 2005 Jul 18.
Article in English | MEDLINE | ID: mdl-16026620

ABSTRACT

BACKGROUND: Sitosterolemia is a recessively inherited disorder in humans that is associated with premature atherosclerotic disease. Mutations in ABCG5 or ABCG8, comprising the sitosterolemia locus, STSL, are now known to cause this disease. Three in-bred strains of rats, WKY, SHR and SHRSP, are known to be sitosterolemic, hypertensive and they carry a missense 'mutation' in a conserved residue of Abcg5, Gly583Cys. Since these rat strains are also know to carry mutations at other genetic loci and the extent of phytosterolemia is only moderate, it is important to verify that the mutations in Abcg5 are causative for phytosterolemia and whether they contribute to hypertension. METHODS: To investigate whether the missense change in Abcg5 is responsible for the sitosterolemia we performed a segregation analysis in 103 F2 rats from a SHR x SD cross. Additionally, we measured tail-cuff blood pressure and measured intestinal lipid transport to identify possible mechanisms whereby this mutation causes sitosterolemia. RESULTS: Segregation analysis showed that the inheritance of the Gly583Cys mutation Abcg5 segregated with elevated plant sterols and this pattern was recessive, proving that this genetic change is responsible for the sitosterolemia in these rat strains. Tail-cuff monitoring of blood pressure in conscious animals showed no significant differences between wild-type, heterozygous and homozygous mutant F2 rats, suggesting that this alteration may not be a significant determinant of hypertension in these rats on a chow diet. CONCLUSION: This study shows that the previously identified Gly583Cys change in Abcg5 in three hypertension-susceptible rats is responsible for the sitosterolemia, but may not be a major determinant of blood pressure in these rats.


Subject(s)
ATP-Binding Cassette Transporters/genetics , Lipoproteins/genetics , Mutation, Missense , Phytosterols/blood , ATP Binding Cassette Transporter, Subfamily G, Member 5 , Animals , Blood Pressure/genetics , Genes, Recessive , Inheritance Patterns , Rats , Rats, Inbred SHR , Sitosterols/blood
20.
J Am Soc Nephrol ; 16(8): 2346-53, 2005 Aug.
Article in English | MEDLINE | ID: mdl-15944340

ABSTRACT

Antioxidants seem to inhibit angiotensin II (Ang II) actions by consuming stimulated reactive oxygen species. An alternative hypothesis was investigated: Antioxidants that are also strong reducers of disulfide bonds inhibit the binding of Ang II to its surface receptors with consequent attenuation of signal transduction and cell action. Incubation of cultured vascular smooth muscle cells, which possess Ang II type 1a receptors, with the reducing agent n-acetylcysteine (NAC) for 1 h at 37 degrees C resulted in decreased Ang II radioligand binding in a concentration-dependent pattern. NAC removal restored Ang II binding within 30 min. Incubation with n-acetylserine, a nonreducing analogue of NAC, did not lower Ang II binding, and oxidized NAC was less effective than reduced NAC in lowering Ang II binding. NAC did not decrease Ang II type 1a receptor protein content. Other antioxidants regulated Ang II receptors differently: alpha-Lipoic acid lowered Ang II binding after 24 h, and vitamin E did not lower Ang II binding at all. NAC inhibited Ang II binding in cell membranes at 21 or 37 but not 4 degrees C. Dihydrolipoic acid (the reduced form of alpha-lipoic acid), which contains free sulfhydryl groups as NAC does, decreased Ang II receptor binding in cell membranes, whereas alpha-lipoic acid, which does not contain free sulfhydryl groups, did not. Ang II-stimulated inositol phosphate formation was decreased by preincubation with NAC (1 h) or alpha-lipoic acid (24 h) but not vitamin E. In conclusion, certain antioxidants that are reducing agents lower Ang II receptor binding, and Ang II-stimulated signal transduction is decreased in proportion to decreased receptor binding.


Subject(s)
Acetylcysteine/pharmacology , Muscle, Smooth, Vascular/cytology , Myocytes, Smooth Muscle/metabolism , Receptors, Angiotensin/metabolism , Animals , Antioxidants/chemistry , Antioxidants/pharmacology , Cell Membrane/metabolism , Disulfides , Dose-Response Relationship, Drug , Green Fluorescent Proteins/metabolism , Immunoblotting , Inositol Phosphates/chemistry , Kinetics , Male , Oxygen/chemistry , Phosphates/chemistry , Protein Binding , Radioligand Assay , Rats , Rats, Sprague-Dawley , Reactive Oxygen Species , Receptor, Angiotensin, Type 1/metabolism , Signal Transduction , Temperature , Thioctic Acid/analogs & derivatives , Thioctic Acid/chemistry , Time Factors , Vitamin E/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL
...