Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
Add more filters











Publication year range
1.
J Pharmacol Exp Ther ; 369(3): 503-510, 2019 06.
Article in English | MEDLINE | ID: mdl-30894457

ABSTRACT

Underlying pathogenic mechanisms in chronic kidney disease (CKD) include chronic inflammation, oxidant stress, and matrix remodeling associated with dysregulated nuclear factor-κ B, nuclear factor-κ B, and SMAD signaling pathways, respectively. Important cytoprotective mechanisms activated by oxidative inflammatory conditions are mediated by nitrated fatty acids that covalently modify proteins to limit inflammation and oxidant stress. In the present study, we evaluated the effects of chronic treatment with CXA-10 (10-nitro-9(E)-octadec-9-enoic acid) in the uninephrectomized deoxycorticosterone acetate-high-salt mouse model of CKD. After 4 weeks of treatment, CXA-10 [2.5 millligrams per kilogram (mpk), p.o.] significantly attenuated increases in plasma cholesterol, heart weight, and kidney weight observed in the model without impacting systemic arterial blood pressure. CXA-10 also reduced albuminuria, nephrinuria, glomerular hypertrophy, and glomerulosclerosis in the model. Inflammatory MCP-1 and fibrosis (collagen, fibronectin, plasminogen activator inhibitor-1, and osteopontin) renal biomarkers were significantly reduced in the CXA-10 (2.5 mpk) group. The anti-inflammatory and antifibrotic effects, as well as glomerular protection, were not observed in the enalapril-treated group. Also, CXA-10 appears to exhibit hormesis as all protective effects observed in the low-dose group were absent in the high-dose group (12.5 mpk). Taken together, these findings demonstrate that, at the appropriate dose, the nitrated fatty acid CXA-10 exhibits anti-inflammatory and antifibrotic effects in the kidney and limits renal injury in a model of CKD.


Subject(s)
Cytoprotection/drug effects , Desoxycorticosterone Acetate/pharmacology , Kidney Diseases/chemically induced , Kidney Diseases/pathology , Kidney/drug effects , Kidney/pathology , Nitro Compounds/pharmacology , Oleic Acids/pharmacology , Salts/adverse effects , Animals , Desoxycorticosterone Acetate/pharmacokinetics , Kidney/metabolism , Kidney Diseases/metabolism , Male , Mice , Nitro Compounds/pharmacokinetics , Oleic Acids/pharmacokinetics , Oxidative Stress/drug effects , Tissue Distribution
2.
Physiol Rep ; 4(12)2016 Jun.
Article in English | MEDLINE | ID: mdl-27356569

ABSTRACT

Development of a disease-modifying therapy to treat autosomal dominant polycystic kidney disease (ADPKD) requires well-characterized preclinical models that accurately reflect the pathology and biochemical changes associated with the disease. Using a Pkd1 conditional knockout mouse, we demonstrate that subtly altering the timing and extent of Pkd1 deletion can have a significant impact on the origin and severity of kidney cyst formation. Pkd1 deletion on postnatal day 1 or 2 results in cysts arising from both the cortical and medullary regions, whereas deletion on postnatal days 3-8 results in primarily medullary cyst formation. Altering the extent of Pkd1 deletion by modulating the tamoxifen dose produces dose-dependent changes in the severity, but not origin, of cystogenesis. Limited Pkd1 deletion produces progressive kidney cystogenesis, accompanied by interstitial fibrosis and loss of kidney function. Cyst growth occurs in two phases: an early, rapid growth phase, followed by a later, slow growth period. Analysis of biochemical pathway changes in cystic kidneys reveals dysregulation of the cell cycle, increased proliferation and apoptosis, activation of Mek-Erk, Akt-mTOR, and Wnt-ß-catenin signaling pathways, and altered glycosphingolipid metabolism that resemble the biochemical changes occurring in human ADPKD kidneys. These pathways are normally active in neonatal mouse kidneys until repressed around 3 weeks of age; however, they remain active following Pkd1 deletion. Together, this work describes the key parameters to accurately model the pathological and biochemical changes associated with ADPKD in a conditional mouse model.


Subject(s)
Gene Deletion , Polycystic Kidney Diseases/genetics , TRPP Cation Channels/metabolism , Animals , Disease Models, Animal , Fibrosis , Kidney/metabolism , Kidney/pathology , MAP Kinase Signaling System , Mice , Polycystic Kidney Diseases/metabolism , Polycystic Kidney Diseases/pathology , TRPP Cation Channels/genetics , Wnt Signaling Pathway
3.
PLoS One ; 11(7): e0157288, 2016.
Article in English | MEDLINE | ID: mdl-27379382

ABSTRACT

BACKGROUND: The platelet-derived growth factor receptor ß (PDGFRß)+ perivascular cell activation becomes increasingly recognized as a main source of scar-associated kidney myofibroblasts and recently emerged as a new cellular therapeutic target. AIMS: In this regard, we first confirmed the presence of PDGFRß+ perivascular cells in a human case of end-stage aristolochic acid nephropathy (AAN) and thereafter we focused on the early fibrosis events of transforming growth factor ß (TGFß) inhibition in a rat model of AAN. MATERIALS AND METHODS: Neutralizing anti-TGFß antibody (1D11) and its control isotype (13C4) were administered (5 mg/kg, i.p.) at Days -1, 0, 2 and 4; AA (15 mg/kg, sc) was injected daily. RESULTS: At Day 5, 1D11 significantly suppressed p-Smad2/3 signaling pathway improving renal function impairment, reduced the score of acute tubular necrosis, peritubular capillaritis, interstitial inflammation and neoangiogenesis. 1D11 markedly decreased interstitial edema, disruption of tubular basement membrane loss of brush border, cytoplasmic edema and organelle ultrastructure alterations (mitochondrial disruption and endoplasmic reticulum edema) in proximal tubular epithelial cells. Moreover, 1D11 significantly inhibited p-PERK activation and attenuated dysregulation of unfolded protein response (UPR) pathways, endoplasmic reticulum and mitochondrial proteostasis in vivo and in vitro. CONCLUSIONS: The early inhibition of p-Smad2/3 signaling pathway improved acute renal function impairment, partially prevented epithelial-endothelial axis activation by maintaining PTEC proteostasis and reduced early PDGFRß+ pericytes-derived myofibroblasts accumulation.


Subject(s)
Acute Kidney Injury/metabolism , Mitochondrial Proteins/metabolism , Pericytes/metabolism , Receptor, Platelet-Derived Growth Factor beta/metabolism , Transforming Growth Factor beta/metabolism , Acute Kidney Injury/chemically induced , Acute Kidney Injury/prevention & control , Animals , Antibodies, Neutralizing/immunology , Antibodies, Neutralizing/pharmacology , Aristolochic Acids , Blotting, Western , Cell Line , Cells, Cultured , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Homeostasis/drug effects , Humans , Kidney Tubules, Proximal/drug effects , Kidney Tubules, Proximal/metabolism , Male , Models, Biological , Myofibroblasts/drug effects , Myofibroblasts/metabolism , Pericytes/drug effects , Rats, Wistar , Signal Transduction/drug effects , Smad Proteins/metabolism , Time Factors , Transforming Growth Factor beta/immunology
4.
Am J Physiol Renal Physiol ; 310(11): F1414-22, 2016 06 01.
Article in English | MEDLINE | ID: mdl-27076647

ABSTRACT

Polycystic kidney diseases (PKDs) are genetic diseases characterized by renal cyst formation with increased cell proliferation, apoptosis, and transition to a secretory phenotype at the expense of terminal differentiation. Despite recent progress in understanding PKD pathogenesis and the emergence of potential therapies, the key molecular mechanisms promoting cystogenesis are not well understood. Here, we demonstrate that mechanisms including endoplasmic reticulum stress, oxidative damage, and compromised mitochondrial function all contribute to nephronophthisis-associated PKD. Ca(2+)/calmodulin-dependent protein kinase II (CaMKII) is emerging as a critical mediator of these cellular processes. Therefore, we reasoned that pharmacological targeting of CaMKII may translate into effective inhibition of PKD in jck mice. Our data demonstrate that CaMKII is activated within cystic kidney epithelia in jck mice. Blockade of CaMKII with a selective inhibitor results in effective inhibition of PKD in jck mice. Mechanistic experiments in vitro and in vivo demonstrated that CaMKII inhibition relieves endoplasmic reticulum stress and oxidative damage and improves mitochondrial integrity and membrane potential. Taken together, our data support CaMKII inhibition as a new and effective therapeutic avenue for the treatment of cystic diseases.


Subject(s)
Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Endoplasmic Reticulum Stress/physiology , Kidney/metabolism , Mitochondria/metabolism , Oxidative Stress/physiology , Polycystic Kidney Diseases/metabolism , Animals , Mice
5.
Hum Mol Genet ; 25(11): 2245-2255, 2016 06 01.
Article in English | MEDLINE | ID: mdl-27053712

ABSTRACT

Polycystic kidney diseases (PKDs) comprise a subgroup of ciliopathies characterized by the formation of fluid-filled kidney cysts and progression to end-stage renal disease. A mechanistic understanding of cystogenesis is crucial for the development of viable therapeutic options. Here, we identify CDK5, a kinase active in post mitotic cells, as a new and important mediator of PKD progression. We show that long-lasting attenuation of PKD in the juvenile cystic kidneys (jck) mouse model of nephronophthisis by pharmacological inhibition of CDK5 using either R-roscovitine or S-CR8 is accompanied by sustained shortening of cilia and a more normal epithelial phenotype, suggesting this treatment results in a reprogramming of cellular differentiation. Also, a knock down of Cdk5 in jck cells using small interfering RNA results in significant shortening of ciliary length, similar to what we observed with R-roscovitine. Finally, conditional inactivation of Cdk5 in the jck mice significantly attenuates cystic disease progression and is associated with shortening of ciliary length as well as restoration of cellular differentiation. Our results suggest that CDK5 may regulate ciliary length by affecting tubulin dynamics via its substrate collapsin response mediator protein 2. Taken together, our data support therapeutic approaches aimed at restoration of ciliogenesis and cellular differentiation as a promising strategy for the treatment of renal cystic diseases.


Subject(s)
Cilia/drug effects , Cyclin-Dependent Kinase 5/genetics , Kidney Failure, Chronic/drug therapy , Polycystic Kidney Diseases/drug therapy , Animals , Cell Differentiation/drug effects , Cilia/pathology , Cyclin-Dependent Kinase 5/antagonists & inhibitors , Disease Models, Animal , Humans , Intercellular Signaling Peptides and Proteins/genetics , Intercellular Signaling Peptides and Proteins/metabolism , Kidney/drug effects , Kidney/pathology , Kidney Failure, Chronic/genetics , Kidney Failure, Chronic/pathology , Mice , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Polycystic Kidney Diseases/genetics , Polycystic Kidney Diseases/pathology , Purines/administration & dosage , Roscovitine , Tubulin/genetics , Tubulin/metabolism
6.
J Bone Miner Res ; 29(5): 1141-57, 2014.
Article in English | MEDLINE | ID: mdl-24166835

ABSTRACT

Altered bone turnover is a key pathologic feature of chronic kidney disease-mineral and bone disorder (CKD-MBD). Expression of TGF-ß1, a known regulator of bone turnover, is increased in bone biopsies from individuals with CKD. Similarly, TGF-ß1 mRNA and downstream signaling is increased in bones from jck mice, a model of high-turnover renal osteodystrophy. A neutralizing anti-TGF-ß antibody (1D11) was used to explore TGF-ß's role in renal osteodystrophy. 1D11 administration to jck significantly attenuated elevated serum osteocalcin and type I collagen C-telopeptides. Histomorphometric analysis indicated that 1D11 administration increased bone volume and suppressed the elevated bone turnover in a dose-dependent manner. These effects were associated with reductions in osteoblast and osteoclast surface areas. Micro-computed tomography (µCT) confirmed the observed increase in trabecular bone volume and demonstrated improvements in trabecular architecture and increased cortical thickness. 1D11 administration was associated with significant reductions in expression of osteoblast marker genes (Runx2, alkaline phosphatase, osteocalcin) and the osteoclast marker gene, Trap5. Importantly, in this model, 1D11 did not improve kidney function or reduce serum parathyroid hormone (PTH) levels, indicating that 1D11 effects on bone are independent of changes in renal or parathyroid function. 1D11 also significantly attenuated high-turnover bone disease in the adenine-induced uremic rat model. Antibody administration was associated with a reduction in pSMAD2/SMAD2 in bone but not bone marrow as assessed by quantitative immunoblot analysis. Immunostaining revealed pSMAD staining in osteoblasts and osteocytes but not osteoclasts, suggesting 1D11 effects on osteoclasts may be indirect. Immunoblot and whole genome mRNA expression analysis confirmed our previous observation that repression of Wnt/ß-catenin expression in bone is correlated with increased osteoclast activity in jck mice and bone biopsies from CKD patients. Furthermore, our data suggest that elevated TGF-ß may contribute to the pathogenesis of high-turnover disease partially through inhibition of ß-catenin signaling.


Subject(s)
Chronic Kidney Disease-Mineral and Bone Disorder/metabolism , Osteoclasts/metabolism , Transforming Growth Factor beta1/metabolism , Wnt Signaling Pathway , Animals , Antibodies, Neutralizing/pharmacology , Antigens, Differentiation/metabolism , Chronic Kidney Disease-Mineral and Bone Disorder/diagnostic imaging , Chronic Kidney Disease-Mineral and Bone Disorder/genetics , Chronic Kidney Disease-Mineral and Bone Disorder/pathology , Collagen Type I , Disease Models, Animal , Male , Mice , Osteocalcin/metabolism , Osteoclasts/pathology , Peptides , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley , Transforming Growth Factor beta1/antagonists & inhibitors , Transforming Growth Factor beta1/genetics , X-Ray Microtomography
7.
PLoS One ; 8(8): e70464, 2013.
Article in English | MEDLINE | ID: mdl-23940580

ABSTRACT

Recent findings indicate that elderly patients with acute kidney injury (AKI) have an increased incidence of progression to chronic kidney disease (CKD) due to incomplete recovery from an acute insult. In the current study, a co-morbid model of AKI was developed to better mimic the patient population and to investigate whether age exacerbates the fibrosis and inflammation that develop in the sequelae of progressive kidney disease following acute injury. Young (8-10 weeks) and aged (46-49 weeks) C57BL/6 mice were subjected to 30 min bilateral renal ischemia-reperfusion (I/R) to induce AKI. The aged animals have greater mortality and prolonged elevation of plasma creatinine correlating with less tubular epithelial cell proliferation compared to the young. Six weeks post-reperfusion, interstitial fibrosis is greater in aged kidneys based on picrosirius red staining and immunolocalization of cellular fibronectin, collagen III and collagen IV. Aged kidneys 6 weeks post-reperfusion also express higher levels of p53 and p21 compared to the young, correlating with greater increases in senescence associated (SA) ß-galactosidase, a known marker of cellular senescence. A higher influx of F4/80(+) macrophages and CD4(+) T lymphocytes is measured and is accompanied by increases in mRNA of monocyte chemoattractant protein-1 (MCP-1) and tumor necrosis factor-α (TNF-α). Importantly, microvascular density is significantly less, correlating with an increase in nitro-tyrosine, a marker of oxidative stress. Collectively, these data demonstrate that prolonged acute injury in the aged animals results in an accelerated progression of kidney disease in a chronic state.


Subject(s)
Acute Kidney Injury/pathology , Cellular Senescence/physiology , Fibrosis/pathology , Kidney/pathology , Reperfusion Injury/pathology , Acute Kidney Injury/metabolism , Animals , Chemokine CCL2/metabolism , Fibrosis/metabolism , Kidney/metabolism , Male , Mice , Mice, Inbred C57BL , Reperfusion Injury/metabolism , Tumor Necrosis Factor-alpha/metabolism
8.
Cell Cycle ; 11(21): 4040-6, 2012 Nov 01.
Article in English | MEDLINE | ID: mdl-23032260

ABSTRACT

Autosomal dominant polycystic kidney disease (ADPKD) and other forms of PKD are associated with dysregulated cell cycle and proliferation. Although no effective therapy for the treatment of PKD is currently available, possible mechanism-based approaches are beginning to emerge. A therapeutic intervention targeting aberrant cilia-cell cycle connection using CDK-inhibitor R-roscovitine showed effective arrest of PKD in jck and cpk models that are not orthologous to human ADPKD. To evaluate whether CDK inhibition approach will translate into efficacy in an orthologous model of ADPKD, we tested R-roscovitine and its derivative S-CR8 in a model with a conditionally inactivated Pkd1 gene (Pkd1 cKO). Similar to ADPKD, Pkd1 cKO mice developed renal and hepatic cysts. Treatment of Pkd1 cKO mice with R-roscovitine and its more potent and selective analog S-CR8 significantly reduced renal and hepatic cystogenesis and attenuated kidney function decline. Mechanism of action studies demonstrated effective blockade of cell cycle and proliferation and reduction of apoptosis. Together, these data validate CDK inhibition as a novel and effective approach for the treatment of ADPKD.


Subject(s)
Adenine/analogs & derivatives , Cyclin-Dependent Kinases/antagonists & inhibitors , Kidney Diseases, Cystic/drug therapy , Liver Diseases/drug therapy , Protein Kinase Inhibitors/therapeutic use , Purines/therapeutic use , Adenine/chemistry , Adenine/pharmacology , Adenine/therapeutic use , Animals , Apoptosis/drug effects , Cyclin-Dependent Kinases/metabolism , Disease Models, Animal , Humans , Kidney Diseases, Cystic/enzymology , Kidney Diseases, Cystic/pathology , Liver Diseases/enzymology , Liver Diseases/pathology , Mice , Mice, Knockout , Polycystic Kidney, Autosomal Dominant/drug therapy , Polycystic Kidney, Autosomal Dominant/enzymology , Polycystic Kidney, Autosomal Dominant/pathology , Protein Kinase C/deficiency , Protein Kinase C/genetics , Protein Kinase C/metabolism , Protein Kinase Inhibitors/chemistry , Protein Kinase Inhibitors/pharmacology , Purines/chemistry , Purines/pharmacology , Roscovitine
9.
Hum Mol Genet ; 21(15): 3397-407, 2012 Aug 01.
Article in English | MEDLINE | ID: mdl-22563011

ABSTRACT

Genetic forms of polycystic kidney diseases (PKDs), including nephronophthisis, are characterized by formation of fluid-filled cysts in the kidneys and progression to end-stage renal disease. No therapies are currently available to treat cystic diseases, making it imperative to dissect molecular mechanisms in search of therapeutic targets. Accumulating evidence suggests a pathogenic role for glucosylceramide (GlcCer) in multiple forms of PKD. It is not known, however, whether other structural glycosphingolipids (GSLs) or bioactive signaling sphingolipids (SLs) modulate cystogenesis. Therefore, we set out to address the role of a specific GSL (ganglioside GM3) and signaling SL (sphingosine-1-phosphate, S1P) in PKD progression, using the jck mouse model of nephronopthisis. To define the role of GM3 accumulation in cystogenesis, we crossed jck mice with mice carrying a targeted mutation in the GM3 synthase (St3gal5) gene. GM3-deficient jck mice displayed milder PKD, revealing a pivotal role for ganglioside GM3. Mechanistic changes in regulation of the cell-cycle machinery and Akt-mTOR signaling were consistent with reduced cystogenesis. Dramatic overexpression of sphingosine kinase 1 (Sphk1) mRNA in jck kidneys suggested a pathogenic role for S1P. Surprisingly, genetic loss of Sphk1 exacerbated cystogenesis and was associated with increased levels of GlcCer and GM3. On the other hand, increasing S1P accumulation through pharmacologic inhibition of S1P lyase had no effect on the progression of cystogenesis or kidney GSL levels. Together, these data suggest that genes involved in the SL metabolism may be modifiers of cystogenesis, and suggest GM3 synthase as a new anti-cystic therapeutic target.


Subject(s)
Phosphotransferases (Alcohol Group Acceptor)/genetics , Polycystic Kidney Diseases/genetics , Sialyltransferases/genetics , Animals , Disease Models, Animal , Glucosylceramides/metabolism , Glycosphingolipids/metabolism , Mice , Oncogene Protein v-akt/genetics , Oncogene Protein v-akt/metabolism , Phosphotransferases (Alcohol Group Acceptor)/metabolism , Polycystic Kidney Diseases/enzymology , Sialyltransferases/metabolism , Sphingosine/metabolism , TOR Serine-Threonine Kinases/metabolism
10.
Am J Physiol Regul Integr Comp Physiol ; 303(1): R57-69, 2012 Jul 01.
Article in English | MEDLINE | ID: mdl-22538513

ABSTRACT

This study examined the effects of anti-TGF-ß antibody (1D11) therapy in Dahl S (S) rats fed a 4% NaCl diet. Baseline renal expression of TGF-ß1 and the degree of injury were lower in female than male S rats maintained on a 0.4% NaCl diet. 4% NaCl diet increased mean arterial pressure (MAP), proteinuria, and renal injury to the same extent in both male and female S rats. Chronic treatment with 1D11 had renoprotective effects in both sexes. The ability of 1D11 to oppose the development of proteinuria when given alone or in combination with antihypertensive agents was further studied in 6-wk-old female S rats, since baseline renal injury was less than that seen in male rats. 1D11, diltiazem, and hydrochlorothiazide (HCT) attenuated the development of hypertension, proteinuria, and glomerular injury. 1D11 had no additional effect when given in combination with these antihypertensive agents. We also explored whether 1D11 could reverse renal injury in 9-wk-old male S rats with preexisting renal injury. MAP increased to 197 ± 4 mmHg and proteinuria rose to >300 mg/day after 3 wk on a 4% NaCl diet. Proteinuria was reduced by 30-40% in rats treated with 1D11, HCT, or captopril + 1D11, but the protective effect was lost in rats fed the 4% NaCl diet for 6 wk. Nevertheless, 1D11, HCT, and captopril + 1D11 still reduced renomedullary and cardiac fibrosis. These results indicate that anti-TGF-ß antibody therapy reduces renal and cardiac fibrosis and affords additional renoprotection when given in combination with various antihypertensive agents in Dahl S rats.


Subject(s)
Acute Kidney Injury/prevention & control , Antibodies, Anti-Idiotypic/immunology , Antibodies, Anti-Idiotypic/therapeutic use , Antihypertensive Agents/therapeutic use , Hypertension/prevention & control , Proteinuria/prevention & control , Transforming Growth Factor beta/immunology , Acute Kidney Injury/chemically induced , Acute Kidney Injury/pathology , Animals , Antibodies, Anti-Idiotypic/pharmacology , Antihypertensive Agents/pharmacology , Blood Pressure/drug effects , Blood Pressure/physiology , Captopril/pharmacology , Captopril/therapeutic use , Diltiazem/pharmacology , Diltiazem/therapeutic use , Disease Models, Animal , Female , Fibrosis , Hydrochlorothiazide/pharmacology , Hydrochlorothiazide/therapeutic use , Hypertension/chemically induced , Hypertension/physiopathology , Male , Proteinuria/chemically induced , Proteinuria/metabolism , Rats , Rats, Inbred Dahl , Sex Characteristics , Sodium Chloride, Dietary/adverse effects
11.
Kidney Int ; 79(11): 1236-43, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21368745

ABSTRACT

Primary focal segmental glomerulosclerosis (FSGS) is a disease with poor prognosis and high unmet therapeutic need. Here, we evaluated the safety and pharmacokinetics of single-dose infusions of fresolimumab, a human monoclonal antibody that inactivates all forms of transforming growth factor-ß (TGF-ß), in a phase I open-label, dose-ranging study. Patients with biopsy-confirmed, treatment-resistant, primary FSGS with a minimum estimated glomerular filtration rate (eGFR) of 25 ml/min per 1.73 m(2), and a urine protein to creatinine ratio over 1.8 mg/mg were eligible. All 16 patients completed the study in which each received one of four single-dose levels of fresolimumab (up to 4 mg/kg) and was followed for 112 days. Fresolimumab was well tolerated with pustular rash the only adverse event in two patients. One patient was diagnosed with a histologically confirmed primitive neuroectodermal tumor 2 years after fresolimumab treatment. Consistent with treatment-resistant FSGS, there was a slight decline in eGFR (median decline baseline to final of 5.85 ml/min per 1.73 m(2)). Proteinuria fluctuated during the study with the median decline from baseline to final in urine protein to creatinine ratio of 1.2 mg/mg with all three Black patients having a mean decline of 3.6 mg/mg. The half-life of fresolimumab was ∼14 days, and the mean dose-normalized Cmax and area under the curve were independent of dose. Thus, single-dose fresolimumab was well tolerated in patients with primary resistant FSGS. Additional evaluation in a larger dose-ranging study is necessary.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Glomerulosclerosis, Focal Segmental/drug therapy , Kidney/drug effects , Transforming Growth Factor beta/antagonists & inhibitors , Adult , Antibodies, Monoclonal/administration & dosage , Antibodies, Monoclonal/adverse effects , Antibodies, Monoclonal/pharmacokinetics , Antibodies, Monoclonal, Humanized , Biomarkers/urine , Biopsy , Creatinine/urine , Dose-Response Relationship, Drug , Europe , Female , Glomerular Filtration Rate/drug effects , Glomerulosclerosis, Focal Segmental/immunology , Glomerulosclerosis, Focal Segmental/pathology , Glomerulosclerosis, Focal Segmental/physiopathology , Humans , Infusions, Parenteral , Kidney/immunology , Kidney/pathology , Kidney/physiopathology , Male , Middle Aged , Proteinuria/drug therapy , Proteinuria/immunology , Transforming Growth Factor beta/immunology , Treatment Outcome , United States , Young Adult
12.
Nat Med ; 16(7): 788-92, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20562878

ABSTRACT

Polycystic kidney disease (PKD) represents a family of genetic disorders characterized by renal cystic growth and progression to kidney failure. No treatment is currently available for people with PKD, although possible therapeutic interventions are emerging. Despite genetic and clinical heterogeneity, PKDs have in common defects of cystic epithelia, including increased proliferation, apoptosis and activation of growth regulatory pathways. Sphingolipids and glycosphingolipids are emerging as major regulators of these cellular processes. We sought to evaluate the therapeutic potential for glycosphingolipid modulation as a new approach to treat PKD. Here we demonstrate that kidney glucosylceramide (GlcCer) and ganglioside GM3 levels are higher in human and mouse PKD tissue as compared to normal tissue, regardless of the causative mutation. Blockade of GlcCer accumulation with the GlcCer synthase inhibitor Genz-123346 effectively inhibits cystogenesis in mouse models orthologous to human autosomal dominant PKD (Pkd1 conditional knockout mice) and nephronophthisis (jck and pcy mice). Molecular analysis in vitro and in vivo indicates that Genz-123346 acts through inhibition of the two key pathways dysregulated in PKD: Akt protein kinase-mammalian target of rapamycin signaling and cell cycle machinery. Taken together, our data suggest that inhibition of GlcCer synthesis represents a new and effective treatment option for PKD.


Subject(s)
Dioxanes/pharmacology , Glucosylceramides/biosynthesis , Polycystic Kidney Diseases/metabolism , Pyrrolidines/pharmacology , Animals , Cell Cycle , Disease Models, Animal , G(M3) Ganglioside/metabolism , Glucosyltransferases/antagonists & inhibitors , Glycosphingolipids/metabolism , Humans , Mice , Mice, Knockout , Polycystic Kidney Diseases/drug therapy , Rats
13.
Kidney Int ; 75(5): 511-7, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19052533

ABSTRACT

While it is known that the arachidonic acid metabolite 20-hydroxyeicosatetraenoic acid (20-HETE) contributes to ischemic injury in the heart and brain, its role in kidney injury is unclear. Here we determined the effects on ischemia-reperfusion injury of the 20-HETE analogues, 20-hydroxyeicosa-5(Z), 14(Z)-dienoic acid (5,14-20-HEDE), and N-[20-hydroxyeicosa-5(Z),14(Z)-dienoyl]glycine (5,14-20-HEDGE), and of the inhibitor of 20-HETE synthesis N-hydroxy-N-(4-butyl-2 methylphenyl) formamidine (HET0016). Using Sprague-Dawley rats we found that while treatment with the inhibitor exacerbated renal injury, infusion of both 5,14-20-HEDE and 5,14-20-HEDGE significantly attenuated injury when compared to vehicle or inhibitor-treated rats. Medullary blood flow, measured by laser-Doppler flowmetry, decreased to half of the baseline one hour after reperfusion in the control rats, but 5,14-20-HEDGE completely prevented this. Treatment of control animals with 5,14-20-HEDGE increased urine output and sodium excretion without altering their mean arterial pressure or glomerular filtration rate. Our results suggest that 20-HETE analogues protect the kidney from ischemia-reperfusion injury by inhibiting renal tubular sodium transport and preventing the post-ischemic fall in medullary blood flow. Analogues of 20-HETE may be useful in the treatment of acute ischemic kidney injury.


Subject(s)
Hydroxyeicosatetraenoic Acids/pharmacology , Kidney Diseases/drug therapy , Reperfusion Injury/drug therapy , Animals , Hydroxyeicosatetraenoic Acids/chemistry , Kidney Medulla/blood supply , Kidney Tubules/metabolism , Protective Agents , Rats , Rats, Sprague-Dawley , Regional Blood Flow , Sodium/metabolism
14.
Am J Physiol Renal Physiol ; 292(4): F1291-301, 2007 Apr.
Article in English | MEDLINE | ID: mdl-17164399

ABSTRACT

In unilateral ureteral obstruction (UUO), the kidney is characterized by increased fibrosis and apoptosis. Both transforming growth factor-beta (TGF-beta) and ANG II have been implicated, and ANG II may mediate its effects through TGF-beta. Previous studies demonstrated amelioration of renal damage when either TGF-beta or ANG II has been individually targeted. In this study, we sought to determine whether combining 1D11 (monoclonal antibody to TGF-beta) and an ACE inhibitor, enalapril, would be more effective in UUO than either individual treatment, as has been shown in diabetic and glomerulonephritic models. Rats underwent UUO and were given either control monoclonal antibody, 1D11 or enalapril, or 1D11/enalapril combination, for 14 days. Kidneys were harvested and examined for fibrosis [trichrome; collagen (real-time PCR, Sircol assay) and fibroblast-specific protein expression (immunohistochemistry), apoptosis (TUNEL), macrophage infiltration (immunohistochemistry), and TGF-beta expression (real-time PCR and tubular localization with immunohistochemistry)]. UUO was found to induce fibrosis, apoptosis, macrophage infiltration, and TGF-beta expression in the obstructed kidney. Administration of either 1D11 or enalapril individually significantly decreased all these changes; when 1D11 and enalapril were combined, there was little additive effect, and the combination did not provide full protection against damage. The results demonstrate that, for the most part, combination therapy is not additive in UUO. This could be due to the continued presence of a physical obstruction or to biochemical differences between UUO and other renal disease models. Furthermore, it suggests that other targets may be amenable to pharmacological manipulation in UUO.


Subject(s)
Antibodies, Monoclonal/administration & dosage , Enalapril/administration & dosage , Transforming Growth Factor beta/immunology , Ureteral Obstruction/drug therapy , Animals , Apoptosis/physiology , Collagen/biosynthesis , Drug Therapy, Combination , Fibrosis , Immunohistochemistry , In Situ Nick-End Labeling , Kidney/drug effects , Kidney/metabolism , Kidney/pathology , Macrophage Activation/drug effects , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley
15.
Nature ; 444(7121): 949-52, 2006 Dec 14.
Article in English | MEDLINE | ID: mdl-17122773

ABSTRACT

Polycystic kidney diseases (PKDs) are primarily characterized by the growth of fluid-filled cysts in renal tubules leading to end-stage renal disease. Mutations in the PKD1 or PKD2 genes lead to autosomal dominant PKD (ADPKD), a slowly developing adult form. Autosomal recessive polycystic kidney disease results from mutations in the PKHD1 gene, affects newborn infants and progresses very rapidly. No effective treatment is currently available for PKD. A previously unrecognized site of subcellular localization was recently discovered for all proteins known to be disrupted in PKD: primary cilia. Because ciliary functions seem to be involved in cell cycle regulation, disruption of proteins associated with cilia or centrioles may directly affect the cell cycle and proliferation, resulting in cystic disease. We therefore reasoned that the dysregulated cell cycle may be the most proximal cause of cystogenesis, and that intervention targeted at this point could provide significant therapeutic benefit for PKD. Here we show that treatment with the cyclin-dependent kinase (CDK) inhibitor (R)-roscovitine does indeed yield effective arrest of cystic disease in jck and cpk mouse models of PKD. Continuous daily administration of the drug is not required to achieve efficacy; pulse treatment provides a robust, long-lasting effect, indicating potential clinical benefits for a lifelong therapy. Molecular studies of the mechanism of action reveal effective cell-cycle arrest, transcriptional inhibition and attenuation of apoptosis. We found that roscovitine is active against cysts originating from different parts of the nephron, a desirable feature for the treatment of ADPKD, in which cysts form in multiple nephron segments. Our results indicate that inhibition of CDK is a new and effective approach to the treatment of PKD.


Subject(s)
Cyclin-Dependent Kinases/antagonists & inhibitors , Disease Models, Animal , Polycystic Kidney Diseases/drug therapy , Purines/pharmacology , Purines/therapeutic use , Animals , Apoptosis/drug effects , Disease Progression , Kidney/drug effects , Kidney/pathology , Mice , Mice, Inbred C57BL , Mice, Mutant Strains , Polycystic Kidney Diseases/pathology , Purines/administration & dosage , Roscovitine , Time Factors
16.
Hypertension ; 45(4): 643-8, 2005 Apr.
Article in English | MEDLINE | ID: mdl-15723968

ABSTRACT

This study examined the role of transforming growth factor-beta (TGF-beta) in altering the glomerular permeability to albumin (P(alb)) during hypertension development in Dahl salt-sensitive (Dahl S) rats and whether TGF-beta acts by inhibiting the glomerular production of 20-HETE. The results indicate that the renal expression of TGF-beta doubles in Dahl S rats fed a high-salt diet for 7 days, and this is associated with a marked rise in P(alb) from 0.19+/-0.04 to 0.75+/-0.01 and changes in the ultrastructure of the glomerular filtration barrier. Chronic treatment of Dahl S rats with a TGF-beta neutralizing antibody prevented the increase in P(alb) and preserved the structure of glomerular capillaries. It had no effect on the rise in blood pressure produced by the high-salt diet. In other studies, preincubation of glomeruli isolated from Sprague Dawley rats with TGF-beta1 (10 ng/mL) for 15 minutes increased P(alb) from 0.01+/-0.01 to 0.60+/-0.02. This was associated with inhibition of the glomerular production of 20-HETE from 221+/-11 to 3.4+/-0.5 mug per 30 minutes per milligram of protein. Pretreatment of Sprague Dawley glomeruli with a stable analog of 20-HETE, 20-hydroxyeicosa-5(Z), 14(Z)-dienoic acid, reduced baseline P(alb) and opposed the effects of TGF-beta to increase P(alb). These studies indicate that upregulation of the glomerular formation of TGF-beta may contribute to the development of proteinuria and glomerular injury early in hypertension development in Dahl S rats by increasing P(alb) through inhibition of the glomerular production of 20-HETE.


Subject(s)
Hydroxyeicosatetraenoic Acids/antagonists & inhibitors , Hypertension/metabolism , Kidney Glomerulus/metabolism , Kidney Glomerulus/pathology , Serum Albumin, Bovine/pharmacokinetics , Transforming Growth Factor beta/metabolism , Animals , Hypertension/etiology , In Vitro Techniques , Kidney/metabolism , Microscopy, Electron , Permeability/drug effects , Rats , Rats, Inbred Dahl , Rats, Sprague-Dawley , Sodium Chloride, Dietary/administration & dosage , Sodium Chloride, Dietary/pharmacology , Transforming Growth Factor beta/pharmacology , Transforming Growth Factor beta1
17.
Am J Physiol Regul Integr Comp Physiol ; 283(3): R757-67, 2002 Sep.
Article in English | MEDLINE | ID: mdl-12185011

ABSTRACT

This study examined the role of transforming growth factor-beta (TGF-beta) in the development of hypertension and renal disease in 9-wk-old male Dahl salt-sensitive (Dahl S) rats fed an 8% NaCl diet for 3 wk. The rats received an intraperitoneal injection of a control or an anti-TGF-beta antibody (anti-TGF-beta Ab) every other day for 2 wk. Mean arterial pressure was significantly lower in Dahl S rats treated with anti-TGF-beta Ab (177 +/- 3 mmHg, n = 12) than in control rats (190 +/- 4 mmHg, n = 17). Anti-TGF-beta Ab therapy also reduced proteinuria from 226 +/- 20 to 154 +/- 16 mg/day. Renal blood flow, cortical blood flow, and creatinine clearance were not significantly different in control and treated rats; however, medullary blood flow was threefold higher in the treated rats than in the controls. Despite the reduction in proteinuria, the degree of glomerulosclerosis and renal hypertrophy was similar in control and anti-TGF-beta Ab-treated rats. Renal levels of TGF-beta1 and -beta2, alpha-actin, type III collagen, and fibronectin mRNA decreased in rats treated with anti-TGF-beta Ab. To examine whether an earlier intervention with anti-TGF-beta Ab would confer additional renoprotection, these studies were repeated in a group of 6-wk-old Dahl S rats. Anti-TGF-beta Ab therapy significantly reduced blood pressure, proteinuria, and the degree of glomerulosclerosis and renal medullary fibrosis in this group of rats. The results indicate that anti-TGF-beta Ab therapy reduces blood pressure, proteinuria, and the renal injury associated with hypertension.


Subject(s)
Antibodies, Monoclonal/pharmacology , Hypertension, Renal/therapy , Transforming Growth Factor beta/immunology , Anesthesia , Animals , Blood Pressure , Collagen Type III/genetics , Fibronectins/genetics , Glomerulosclerosis, Focal Segmental/pathology , Glomerulosclerosis, Focal Segmental/therapy , Hypertension, Renal/pathology , Kidney Glomerulus/pathology , Male , Proteinuria/therapy , Rats , Rats, Inbred Dahl , Renal Circulation , Sodium Chloride, Dietary/pharmacology , Transforming Growth Factor beta/genetics , Transforming Growth Factor beta1 , Transforming Growth Factor beta2
SELECTION OF CITATIONS
SEARCH DETAIL