Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 39
Filter
1.
Transl Res ; 255: 140-151, 2023 05.
Article in English | MEDLINE | ID: mdl-36563959

ABSTRACT

While natriuretic peptides (NPs) are primarily known for their renal and cardiovascular actions, NPs stimulate lipolysis in adipocytes and induce a thermogenic program in white adipose tissue (WAT) that resembles brown fat. The biologic effects of NPs are negatively regulated by the NP clearance receptor (NPRC), which binds and degrades NPs. Knockout (KO) of NPRC protects against diet induced obesity and improves insulin sensitivity in obese mice. To determine if pharmacologic blockade of NPRC enhanced the beneficial metabolic actions of NPs in type 2 diabetes, we blocked NP clearance in a mouse model of type 2 diabetes using the specific NPRC ligand ANP(4-23). We found that treatment with ANP(4-23) caused a significant decrease in body weight by increasing energy expenditure and reducing fat mass without a change in lean body mass. The decrease in fat mass was associated with a significant improvement in insulin sensitivity and reduced serum insulin levels. These beneficial effects were accompanied by a decrease in infiltrating macrophages in adipose tissue, and reduced expression of inflammatory markers in both serum and WAT. These data suggest that inhibiting NP clearance may be an effective pharmacologic approach to promote weight loss and enhance insulin sensitivity in type 2 diabetes. Optimizing the therapeutic approach may lead to useful therapies for obesity and type 2 diabetes.


Subject(s)
Diabetes Mellitus, Type 2 , Insulin Resistance , Mice , Animals , Diabetes Mellitus, Type 2/drug therapy , Diabetes Mellitus, Type 2/metabolism , Mice, Knockout , Natriuretic Peptides/metabolism , Natriuretic Peptides/therapeutic use , Obesity/metabolism , Weight Loss
2.
Physiol Rep ; 9(21): e15095, 2021 11.
Article in English | MEDLINE | ID: mdl-34755480

ABSTRACT

Glomerular podocytes play a key role in proteinuric diseases. Accumulating evidence suggests that cGMP signaling has podocyte protective effects. The major source of cGMP generation in podocytes is natriuretic peptides. The natriuretic peptide clearance receptor (NPRC) binds and degrades natriuretic peptides. As a result, NPRC inhibits natriuretic peptide-induced cGMP generation. To enhance cGMP generation in podocytes, we blocked natriuretic peptide clearance using the specific NPRC ligand ANP(4-23). We then studied the effects of NPRC blockade in both cultured podocytes and in a mouse transgenic (TG) model of focal segmental glomerulosclerosis (FSGS) created in our laboratory. In this model, a single dose of the podocyte toxin puromycin aminonucleoside (PAN) causes robust albuminuria in TG mice, but only mild disease in non-TG animals. We found that natriuretic peptides protected cultured podocytes from PAN-induced apoptosis, and that ANP(4-23) enhanced natriuretic peptide-induced cGMP generation in vivo. PAN-induced heavy proteinuria in vehicle-treated TG mice, and this increase in albuminuria was reduced by treatment with ANP(4-23). Treatment with ANP(4-23) also reduced the number of mice with glomerular injury and enhanced urinary cGMP excretion, but these differences were not statistically significant. Systolic BP was similar in vehicle and ANP(4-23)-treated mice. These data suggest that: 1. Pharmacologic blockade of NPRC may be useful for treating glomerular diseases such as FSGS, and 2. Treatment outcomes might be improved by optimizing NPRC blockade to inhibit natriuretic peptide clearance more effectively.


Subject(s)
Atrial Natriuretic Factor/therapeutic use , Glomerulosclerosis, Focal Segmental/drug therapy , Peptide Fragments/therapeutic use , Proteinuria/drug therapy , Receptors, Atrial Natriuretic Factor/metabolism , Animals , Apoptosis , Atrial Natriuretic Factor/pharmacology , Cell Line , Cyclic GMP/metabolism , Female , Male , Mice , Natriuretic Peptides/metabolism , Peptide Fragments/pharmacology , Podocytes/drug effects , Podocytes/metabolism
3.
JCI Insight ; 6(15)2021 08 09.
Article in English | MEDLINE | ID: mdl-34369383

ABSTRACT

The transcription factor Twist1 regulates several processes that could impact kidney disease progression, including epithelial cell differentiation and inflammatory cytokine induction. Podocytes are specialized epithelia that exhibit features of immune cells and could therefore mediate unique effects of Twist1 on glomerular disease. To study Twist1 functions in podocytes during proteinuric kidney disease, we employed a conditional mutant mouse in which Twist1 was selectively ablated in podocytes (Twist1-PKO). Deletion of Twist1 in podocytes augmented proteinuria, podocyte injury, and foot process effacement in glomerular injury models. Twist1 in podocytes constrained renal accumulation of monocytes/macrophages and glomerular expression of CCL2 and the macrophage cytokine TNF-α after injury. Deletion of TNF-α selectively from podocytes had no impact on the progression of proteinuric nephropathy. By contrast, the inhibition of CCL2 abrogated the exaggeration in proteinuria and podocyte injury accruing from podocyte Twist1 deletion. Collectively, Twist1 in podocytes mitigated urine albumin excretion and podocyte injury in proteinuric kidney diseases by limiting CCL2 induction that drove monocyte/macrophage infiltration into injured glomeruli. Myeloid cells, rather than podocytes, further promoted podocyte injury and glomerular disease by secreting TNF-α. These data highlight the capacity of Twist1 in the podocyte to mitigate glomerular injury by curtailing the local myeloid immune response.


Subject(s)
Chemokine CCL2/metabolism , Myeloid Cells/immunology , Podocytes/metabolism , Renal Insufficiency, Chronic , Tumor Necrosis Factor-alpha/metabolism , Twist-Related Protein 1/metabolism , Animals , Cell Differentiation , Gene Silencing , Immunity/immunology , Kidney Glomerulus/immunology , Kidney Glomerulus/injuries , Kidney Glomerulus/metabolism , Macrophages , Mice , Proteinuria/metabolism , Renal Insufficiency, Chronic/immunology , Renal Insufficiency, Chronic/metabolism , Renal Insufficiency, Chronic/pathology
4.
Cells ; 9(1)2019 12 23.
Article in English | MEDLINE | ID: mdl-31877991

ABSTRACT

Over a decade ago, mutations in the gene encoding TRPC6 (transient receptor potential cation channel, subfamily C, member 6) were linked to development of familial forms of nephrosis. Since this discovery, TRPC6 has been implicated in the pathophysiology of non-genetic forms of kidney disease including focal segmental glomerulosclerosis (FSGS), diabetic nephropathy, immune-mediated kidney diseases, and renal fibrosis. On the basis of these findings, TRPC6 has become an important target for the development of therapeutic agents to treat diverse kidney diseases. Although TRPC6 has been a major focus for drug discovery, more recent studies suggest that other TRPC family members play a role in the pathogenesis of glomerular disease processes and chronic kidney disease (CKD). This review highlights the data implicating TRPC6 and other TRPC family members in both genetic and non-genetic forms of kidney disease, focusing on TRPC3, TRPC5, and TRPC6 in a cell type (glomerular podocytes) that plays a key role in proteinuric kidney diseases.


Subject(s)
Kidney Diseases/metabolism , TRPC Cation Channels/metabolism , TRPC6 Cation Channel/metabolism , Diabetic Nephropathies/pathology , Fibrosis , Glomerulosclerosis, Focal Segmental/pathology , Humans , Kidney/pathology , Kidney Diseases/genetics , Kidney Diseases/physiopathology , Proteinuria/metabolism , Renal Insufficiency, Chronic/pathology , TRPC Cation Channels/genetics , TRPC Cation Channels/physiology , TRPC6 Cation Channel/genetics , TRPC6 Cation Channel/physiology
5.
Kidney Int ; 95(2): 321-332, 2019 02.
Article in English | MEDLINE | ID: mdl-30665571

ABSTRACT

Gain-of-function mutations in TRPC6 cause familial focal segmental glomerulosclerosis, and TRPC6 is upregulated in glomerular diseases including diabetic kidney disease. We studied the effect of systemic TRPC6 knockout in the Akita model of type 1 diabetes. Knockout of TRPC6 inhibited albuminuria in Akita mice at 12 and 16 weeks of age, but this difference disappeared by 20 weeks. Knockout of TRPC6 also reduced tubular injury in Akita mice; however, mesangial expansion was significantly increased. Hyperglycemia and blood pressure were similar between TRPC6 knockout and wild-type Akita mice, but knockout mice were more insulin resistant. In cultured podocytes, knockout of TRPC6 inhibited expression of the calcium/calcineurin responsive gene insulin receptor substrate 2 and decreased insulin responsiveness. Insulin resistance is reported to promote diabetic kidney disease independent of blood glucose levels. While the mechanisms are not fully understood, insulin activates both Akt2 and ERK, which inhibits apoptosis signal regulated kinase 1 (ASK1)-p38-induced apoptosis. In cultured podocytes, hyperglycemia stimulated p38 signaling and induced apoptosis, which was reduced by insulin and ASK1 inhibition and enhanced by Akt or ERK inhibition. Glomerular p38 signaling was increased in TRPC6 knockout Akita mice and was associated with enhanced expression of the p38 gene target cyclooxygenase 2. These data suggest that knockout of TRPC6 in Akita mice promotes insulin resistance and exacerbates glomerular disease independent of hyperglycemia.


Subject(s)
Diabetes Mellitus, Type 1/complications , Diabetic Nephropathies/pathology , Glomerular Mesangium/pathology , TRPC Cation Channels/metabolism , Animals , Apoptosis , Blood Glucose/analysis , Diabetes Mellitus, Type 1/blood , Diabetes Mellitus, Type 1/genetics , Diabetic Nephropathies/etiology , Disease Models, Animal , Humans , Insulin Receptor Substrate Proteins/metabolism , Insulin Resistance/genetics , MAP Kinase Kinase Kinase 5/metabolism , Mice , Mice, Knockout , Podocytes , TRPC Cation Channels/genetics , TRPC6 Cation Channel
6.
Sci Rep ; 8(1): 12286, 2018 08 16.
Article in English | MEDLINE | ID: mdl-30115939

ABSTRACT

Expression of a constitutively active Rho A (V14Rho) in podocytes in vivo induces albuminuria and foot process (FP) effacement. These effects may be mediated by the Rho A effector Rho kinase (ROK); but inhibition of ROK with Y27632 failed to attenuate albuminuria or FP effacement in V14Rho mice. ROK activates LIM kinases (LIMKs), which phosphorylate and inhibit the actin depolymerizing factor cofilin 1 (CFL1). Sustained phosphorylation of CFL1 is implicated in human nephrotic diseases, but Y27632 did not inhibit phosphorylation of CFL1 in vivo, despite effective ROK inhibition. CFL1 is also phosphorylated by testis-specific kinase 1 (TESK1) on the same serine residue. TESK1 was expressed in podocytes, and, similar to the in vivo situation, Y27632 had little effect on phospho-CFL1 (pCFL1) levels in cultured podocytes. In contrast, Y27632 reduced pCFL1 levels in TESK1 knockout (KO) cells. ROK inhibition enhanced podocyte motility but, the motility promoting effect of Y27632 was absent in TESK1 KO podocytes. Thus, TESK1 regulates podocyte cytoskeletal dynamics in glomerular podocytes and may play an important role in regulating glomerular filtration barrier integrity in glomerular disease processes.


Subject(s)
Cofilin 1/metabolism , Kidney Glomerulus/metabolism , Podocytes/metabolism , Protein Serine-Threonine Kinases/metabolism , Actin Cytoskeleton/metabolism , Amides/pharmacology , Animals , Cell Line, Transformed , Humans , Kidney Glomerulus/cytology , Kidney Glomerulus/enzymology , Mice , Mice, Transgenic , Phosphorylation , Podocytes/enzymology , Protein Kinase Inhibitors/pharmacology , Protein Serine-Threonine Kinases/genetics , Pyridines/pharmacology , rho-Associated Kinases/antagonists & inhibitors
7.
J Am Soc Nephrol ; 29(8): 2110-2122, 2018 08.
Article in English | MEDLINE | ID: mdl-30002222

ABSTRACT

BACKGROUND: We previously reported that mutations in the anillin (ANLN) gene cause familial forms of FSGS. ANLN is an F-actin binding protein that modulates podocyte cell motility and interacts with the phosphoinositide 3-kinase (PI3K) pathway through the slit diaphragm adaptor protein CD2-associated protein (CD2AP). However, it is unclear how the ANLN mutations cause the FSGS phenotype. We hypothesized that the R431C mutation exerts its pathogenic effects by uncoupling ANLN from CD2AP. METHODS: We conducted in vivo complementation assays in zebrafish to determine the effect of the previously identified missense ANLN variants, ANLNR431C and ANLNG618C during development. We also performed in vitro functional assays using human podocyte cell lines stably expressing wild-type ANLN (ANLNWT ) or ANLNR431C . RESULTS: Experiments in anln-deficient zebrafish embryos showed a loss-of-function effect for each ANLN variant. In human podocyte lines, expression of ANLNR431C increased cell migration, proliferation, and apoptosis. Biochemical characterization of ANLNR431C -expressing podocytes revealed hyperactivation of the PI3K/AKT/mTOR/p70S6K/Rac1 signaling axis and activation of mTOR-driven endoplasmic reticulum stress in ANLNR431C -expressing podocytes. Inhibition of mTOR, GSK-3ß, Rac1, or calcineurin ameliorated the effects of ANLNR431C . Additionally, inhibition of the calcineurin/NFAT pathway reduced the expression of endogenous ANLN and mTOR. CONCLUSIONS: The ANLNR431C mutation causes multiple derangements in podocyte function through hyperactivation of PI3K/AKT/mTOR/p70S6K/Rac1 signaling. Our findings suggest that the benefits of calcineurin inhibition in FSGS may be due, in part, to the suppression of ANLN and mTOR. Moreover, these studies illustrate that rational therapeutic targets for familial FSGS can be identified through biochemical characterization of dysregulated podocyte phenotypes.


Subject(s)
Glomerulosclerosis, Focal Segmental/genetics , Microfilament Proteins/genetics , Phosphatidylinositol 3-Kinases/genetics , Proto-Oncogene Proteins c-akt/genetics , TOR Serine-Threonine Kinases/metabolism , Animals , Apoptosis/genetics , Cell Movement/genetics , Cells, Cultured , Gene Expression Regulation , Glomerulosclerosis, Focal Segmental/pathology , Glomerulosclerosis, Focal Segmental/physiopathology , Humans , Mutation, Missense , Podocytes/metabolism , Sensitivity and Specificity , Signal Transduction , Zebrafish , rac1 GTP-Binding Protein/genetics
8.
Kidney Int ; 92(2): 283-285, 2017 08.
Article in English | MEDLINE | ID: mdl-28709595

ABSTRACT

Selective modulation of Rho GTPase activity in podocytes recapitulates characteristic features of human nephrosis. Using a mouse model, Robins et al. found that high levels of Rac1 activation in podocytes caused podocyte detachment and glomerulosclerosis. Podocyte Rac1 activity was enhanced in biopsy specimens from patients with nephrosis, and serum from this patient population activated Rac1 in cultured podocytes. These data provide a causal link between podocyte Rac1 activation and human nephrotic diseases.


Subject(s)
Glomerulosclerosis, Focal Segmental , Nephrosis , Nephrotic Syndrome , Podocytes , Humans , rac1 GTP-Binding Protein
9.
Am J Physiol Renal Physiol ; 313(2): F430-F439, 2017 Aug 01.
Article in English | MEDLINE | ID: mdl-28490532

ABSTRACT

Enhanced expression of cyclooxygenase 2 (COX2) in podocytes contributes to glomerular injury in diabetic kidney disease, but some basal level of podocyte COX2 expression might be required to promote podocyte attachment and/or survival. To investigate the role of podocyte COX2 expression in diabetic kidney disease, we deleted COX2 specifically in podocytes in a mouse model of Type 1 diabetes mellitus (Akita mice). Podocyte-specific knockout (KO) of COX2 did not affect renal morphology or albuminuria in nondiabetic mice. Albuminuria was significantly increased in wild-type (WT) and KO Akita mice compared with nondiabetic controls, and the increase in albuminuria was significantly greater in KO Akita mice compared with WT Akita mice at both 16 and 20 wk of age. At the 20-wk time point, mesangial expansion was also increased in WT and KO Akita mice compared with nondiabetic animals, and these histologic abnormalities were not improved by KO of COX2. Tubular injury was seen only in diabetic mice, but there were no significant differences between groups. Thus, KO of COX2 enhanced albuminuria and did not improve the histopathologic features of diabetic kidney disease. These data suggest that 1) KO of COX2 in podocytes does not ameliorate diabetic kidney disease in Akita mice, and 2) some basal level of podocyte COX2 expression in podocytes is necessary to attenuate the adverse effects of diabetes on glomerular filtration barrier function.


Subject(s)
Albuminuria/enzymology , Cyclooxygenase 2/deficiency , Diabetic Nephropathies/enzymology , Podocytes/enzymology , Albuminuria/genetics , Albuminuria/pathology , Albuminuria/urine , Animals , Biomarkers/blood , Biomarkers/urine , Blood Glucose/metabolism , Blood Pressure , Cyclooxygenase 2/genetics , Diabetic Nephropathies/genetics , Diabetic Nephropathies/pathology , Diabetic Nephropathies/urine , Disease Models, Animal , Eicosanoids/urine , Genetic Predisposition to Disease , Integrases/genetics , Intracellular Signaling Peptides and Proteins/genetics , Male , Membrane Proteins/genetics , Mice, 129 Strain , Mice, Knockout , Phenotype , Podocytes/ultrastructure , Promoter Regions, Genetic , Renin/metabolism , Severity of Illness Index
10.
Am J Physiol Renal Physiol ; 309(10): F807-20, 2015 Nov 15.
Article in English | MEDLINE | ID: mdl-26336162

ABSTRACT

Diabetic nephropathy (DN) is a serious complication of both type 1 and type 2 diabetes mellitus. The disease is now the most common cause of end-stage kidney disease (ESKD) in developed countries, and both the incidence and prevalence of diabetes mellitus is increasing worldwide. Current treatments are directed at controlling hyperglycemia and hypertension, as well as blockade of the renin angiotensin system with angiotensin-converting enzyme inhibitors (ACEIs), and angiotensin receptor blockers. Despite these therapies, DN progresses to ESKD in many patients. As a result, much interest is focused on developing new therapies. It has been over two decades since ACEIs were shown to have beneficial effects in DN independent of their blood pressure-lowering actions. Since that time, our understanding of disease mechanisms in DN has evolved. In this review, we summarize major cell signaling pathways implicated in the pathogenesis of diabetic kidney disease, as well as emerging treatment strategies. The goal is to identify promising targets that might be translated into therapies for the treatment of patients with diabetic kidney disease.


Subject(s)
Angiotensin Receptor Antagonists/pharmacology , Angiotensin-Converting Enzyme Inhibitors/pharmacology , Antihypertensive Agents/pharmacology , Diabetes Mellitus, Type 2/drug therapy , Diabetic Nephropathies/drug therapy , Kidney Failure, Chronic/drug therapy , Animals , Humans , Kidney Failure, Chronic/diagnosis
11.
J Clin Invest ; 125(5): 1913-26, 2015 May.
Article in English | MEDLINE | ID: mdl-25844902

ABSTRACT

Familial forms of focal segmental glomerulosclerosis (FSGS) have been linked to gain-of-function mutations in the gene encoding the transient receptor potential channel C6 (TRPC6). GPCRs coupled to Gq signaling activate TRPC6, suggesting that Gq-dependent TRPC6 activation underlies glomerular diseases. Here, we developed a murine model in which a constitutively active Gq α subunit (Gq(Q209L), referred to herein as GqQ>L) is specifically expressed in podocytes and examined the effects of this mutation in response to puromycin aminonucleoside (PAN) nephrosis. We found that compared with control animals, animals expressing GqQ>L exhibited robust albuminuria, structural features of FSGS, and reduced numbers of glomerular podocytes. Gq activation stimulated calcineurin (CN) activity, resulting in CN-dependent upregulation of TRPC6 in murine kidneys. Deletion of TRPC6 in GqQ>L-expressing mice prevented FSGS development and inhibited both tubular damage and podocyte loss induced by PAN nephrosis. Similarly, administration of the CN inhibitor FK506 reduced proteinuria and tubular injury but had more modest effects on glomerular pathology and podocyte numbers in animals with constitutive Gq activation. Moreover, these Gq-dependent effects on podocyte injury were generalizable to diabetic kidney disease, as expression of GqQ>L promoted albuminuria, mesangial expansion, and increased glomerular basement membrane width in diabetic mice. Together, these results suggest that targeting Gq/TRPC6 signaling may have therapeutic benefits for the treatment of glomerular diseases.


Subject(s)
GTP-Binding Protein alpha Subunits, Gq-G11/physiology , Glomerulosclerosis, Focal Segmental/genetics , TRPC Cation Channels/physiology , Albuminuria/chemically induced , Animals , Calcineurin/metabolism , Diabetes Mellitus, Type 1/complications , Diabetes Mellitus, Type 1/genetics , Diabetic Nephropathies/genetics , Diabetic Nephropathies/metabolism , Diabetic Nephropathies/pathology , GTP-Binding Protein alpha Subunits, Gq-G11/genetics , Gene Deletion , Genes, Reporter , Glomerulosclerosis, Focal Segmental/chemically induced , Glomerulosclerosis, Focal Segmental/pathology , HEK293 Cells , Humans , Kidney Glomerulus/pathology , Kidney Tubules/pathology , Mice , Mice, Mutant Strains , Mice, Transgenic , NFATC Transcription Factors/metabolism , Podocytes/metabolism , Point Mutation , Puromycin Aminonucleoside/toxicity , Recombinant Fusion Proteins/metabolism , Signal Transduction , TRPC Cation Channels/biosynthesis , TRPC Cation Channels/deficiency , TRPC Cation Channels/genetics , TRPC6 Cation Channel , Tacrolimus/pharmacology
12.
J Am Soc Nephrol ; 26(4): 831-43, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25145932

ABSTRACT

FSGS is a clinical disorder characterized by focal scarring of the glomerular capillary tuft, podocyte injury, and nephrotic syndrome. Although idiopathic forms of FSGS predominate, recent insights into the molecular and genetic causes of FSGS have enhanced our understanding of disease pathogenesis. Here, we report a novel missense mutation of the transcriptional regulator Wilms' Tumor 1 (WT1) as the cause of nonsyndromic, autosomal dominant FSGS in two Northern European kindreds from the United States. We performed sequential genome-wide linkage analysis and whole-exome sequencing to evaluate participants from family DUK6524. Subsequently, whole-exome sequencing and direct sequencing were performed on proband DNA from family DUK6975. We identified multiple suggestive loci on chromosomes 6, 11, and 13 in family DUK6524 and identified a segregating missense mutation (R458Q) in WT1 isoform D as the cause of FSGS in this family. The identical mutation was found in family DUK6975. The R458Q mutation was not found in 1600 control chromosomes and was predicted as damaging by in silico simulation. We depleted wt1a in zebrafish embryos and observed glomerular injury and filtration defects, both of which were rescued with wild-type but not mutant human WT1D mRNA. Finally, we explored the subcellular mechanism of the mutation in vitro. WT1(R458Q) overexpression significantly downregulated nephrin and synaptopodin expression, promoted apoptosis in HEK293 cells and impaired focal contact formation in podocytes. Taken together, these data suggest that the WT1(R458Q) mutation alters the regulation of podocyte homeostasis and causes nonsyndromic FSGS.


Subject(s)
Glomerulosclerosis, Focal Segmental/genetics , Microfilament Proteins/metabolism , WT1 Proteins/genetics , Adolescent , Adult , Animals , Cell Movement , Cell Survival , Exome , Female , Gene Expression Regulation , Gene Knockdown Techniques , Genetic Linkage , Glomerulosclerosis, Focal Segmental/metabolism , HEK293 Cells , Humans , Male , Mutation, Missense , Nephrosis/etiology , Nephrosis/metabolism , Podocytes/physiology , Sequence Analysis, DNA , WT1 Proteins/deficiency , Young Adult , Zebrafish , Zebrafish Proteins/deficiency
13.
Kidney Int ; 85(5): 1009-11, 2014 May.
Article in English | MEDLINE | ID: mdl-24786873

ABSTRACT

The role of the circulating renin-angiotensin system (RAS) in regulating systemic blood pressure and sodium balance is well established. More recently, researchers have turned their focus to the local generation of angiotensin II (Ang II) in specific tissues. Matsusaka et al. revisit the renal RAS and provide evidence that liver-derived angiotensinogen (AGT) is a major determinant of renal Ang II levels in a model of podocyte injury.


Subject(s)
Angiotensin II/metabolism , Angiotensinogen/metabolism , Kidney Diseases/metabolism , Kidney Tubules, Proximal/metabolism , Liver/metabolism , Podocytes/metabolism , Animals
14.
Biochem Biophys Res Commun ; 444(4): 622-7, 2014 Feb 21.
Article in English | MEDLINE | ID: mdl-24491571

ABSTRACT

To determine if augmenting podocyte injury promotes the development of advanced diabetic nephropathy (DN), we created mice that expressed the enzyme cytosine deaminase (CD) specifically in podocytes of diabetic Akita mice (Akita-CD mice). In these mice, treatment with the prodrug 5-flucytosine (5-FC) causes podocyte injury as a result of conversion to the toxic metabolite 5-fluorouracil (5-FU). We found that treatment of 4-5 week old Akita mice with 5-FC for 5 days caused robust albuminuria at 16 and 20 weeks of age compared to 5-FC treated Akita controls, which do not express CD (Akita CTLs). By 20 weeks of age, there was a significant increase in mesangial expansion in Akita-CD mice compared to Akita CTLs, which was associated with a variable increase in glomerular basement membrane (GBM) width and interstitial fibrosis. At 20 weeks of age, podocyte number was similarly reduced in both groups of Akita mice, and was inversely correlated with the albuminuria and mesangial expansion. Thus, enhancing podocyte injury early in the disease process promotes the development of prominent mesangial expansion, interstitial fibrosis, increased GBM thickness and robust albuminuria. These data suggest that podocytes play a key role in the development of advanced features of diabetic kidney disease.


Subject(s)
Diabetic Nephropathies/pathology , Kidney/pathology , Podocytes/pathology , Albuminuria/complications , Animals , Antimetabolites/adverse effects , Cytosine Deaminase/genetics , Diabetic Nephropathies/blood , Diabetic Nephropathies/enzymology , Diabetic Nephropathies/genetics , Disease Models, Animal , Flucytosine/adverse effects , Fluorouracil/adverse effects , Gene Expression , Kidney/drug effects , Kidney/enzymology , Kidney/metabolism , Mice , Podocytes/drug effects , Podocytes/enzymology , Podocytes/metabolism , Prodrugs/adverse effects
15.
Biochem Biophys Res Commun ; 425(2): 407-12, 2012 Aug 24.
Article in English | MEDLINE | ID: mdl-22846567

ABSTRACT

G protein-coupled receptor kinase interacting protein 2 (GIT2) is a signaling scaffold protein involved in the regulation of cytoskeletal structure, membrane trafficking, and G protein-coupled receptor internalization. Since dynamic cytoskeletal reorganization plays key roles both in osteoblast differentiation and in the maintenance of osteoclast polarity during bone resorption, we hypothesized that skeletal physiology would be altered in GIT2(-/-) mice. We found that adult GIT2(-/-) mice have decreased bone mineral density and bone volume in both the trabecular and cortical compartments. This osteopenia was associated with decreased numbers of mature osteoblasts, diminished osteoblastic activity, and increased marrow adiposity, suggesting a defect in osteoblast maturation. In vitro, mesenchymal stem cells derived from GIT2(-/-) mice exhibited impaired differentiation into osteoblasts and increased adipocyte differentiation, consistent with a role for GIT2 in mesenchymal stem cell fate determination. Despite elevated osteoclast inducing cytokines and osteoclast numbers, GIT2(-/-) mice also exhibit impaired bone resorption, consistent with a further role for GIT2 in regulating osteoclast function. Collectively, these findings underscore the importance of the cytoskeleton in both osteoblast and osteoclast function and demonstrate that GIT2 plays essential roles in skeletal metabolism, affecting both bone formation and bone resorption in vivo.


Subject(s)
Bone Resorption/genetics , Cell Cycle Proteins/metabolism , Cell Differentiation , Mesenchymal Stem Cells/cytology , Osteoblasts/cytology , Osteogenesis/genetics , Phosphoproteins/metabolism , Animals , Bone Density/genetics , Cell Count , Cell Cycle Proteins/genetics , Cytoskeleton/metabolism , Female , GTPase-Activating Proteins , Intercellular Signaling Peptides and Proteins , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Phosphoproteins/genetics
16.
PLoS One ; 7(4): e33942, 2012.
Article in English | MEDLINE | ID: mdl-22496773

ABSTRACT

Akita mice are a genetic model of type 1 diabetes. In the present studies, we investigated the phenotype of Akita mice on the FVB/NJ background and examined urinary nephrin excretion as a marker of kidney injury. Male Akita mice were compared with non-diabetic controls for functional and structural characteristics of renal and cardiac disease. Podocyte number and apoptosis as well as urinary nephrin excretion were determined in both groups. Male FVB/NJ Akita mice developed sustained hyperglycemia and albuminuria by 4 and 8 weeks of age, respectively. These abnormalities were accompanied by a significant increase in systolic blood pressure in 10-week old Akita mice, which was associated with functional, structural and molecular characteristics of cardiac hypertrophy. By 20 weeks of age, Akita mice developed a 10-fold increase in albuminuria, renal and glomerular hypertrophy and a decrease in the number of podocytes. Mild-to-moderate glomerular mesangial expansion was observed in Akita mice at 30 weeks of age. In 4-week old Akita mice, the onset of hyperglycemia was accompanied by increased podocyte apoptosis and enhanced excretion of nephrin in urine before the development of albuminuria. Urinary nephrin excretion was also significantly increased in albuminuric Akita mice at 16 and 20 weeks of age and correlated with the albumin excretion rate. These data suggest that: 1. FVB/NJ Akita mice have phenotypic characteristics that may be useful for studying the mechanisms of kidney and cardiac injury in diabetes, and 2. Enhanced urinary nephrin excretion is associated with kidney injury in FVB/NJ Akita mice and is detectable early in the disease process.


Subject(s)
Biomarkers/urine , Diabetes Complications/urine , Diabetes Mellitus, Type 1/physiopathology , Diabetic Nephropathies/urine , Kidney/injuries , Membrane Proteins/urine , Podocytes/pathology , Albuminuria/etiology , Animals , Blotting, Western , Diabetes Complications/diagnosis , Diabetes Complications/etiology , Diabetes Mellitus, Experimental/physiopathology , Diabetes Mellitus, Experimental/urine , Diabetes Mellitus, Type 1/urine , Diabetic Nephropathies/diagnosis , Diabetic Nephropathies/etiology , Disease Models, Animal , Hyperglycemia/etiology , Immunoenzyme Techniques , Kidney/pathology , Male , Mice , Mice, Inbred Strains , Phenotype , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction
17.
Circ Res ; 110(12): 1604-17, 2012 Jun 08.
Article in English | MEDLINE | ID: mdl-22534490

ABSTRACT

RATIONALE: Human clinical trials using type 1 angiotensin (AT(1)) receptor antagonists indicate that angiotensin II is a critical mediator of cardiovascular and renal disease. However, recent studies have suggested that individual tissue pools of AT(1) receptors may have divergent effects on target organ damage in hypertension. OBJECTIVE: We examined the role of AT(1) receptors on T lymphocytes in the pathogenesis of hypertension and its complications. METHODS AND RESULTS: Deficiency of AT(1) receptors on T cells potentiated kidney injury during hypertension with exaggerated renal expression of chemokines and enhanced accumulation of T cells in the kidney. Kidneys and purified CD4(+) T cells from "T cell knockout" mice lacking AT(1) receptors on T lymphocytes had augmented expression of Th1-associated cytokines including interferon-γ and tumor necrosis factor-α. Within T lymphocytes, the transcription factors T-bet and GATA-3 promote differentiation toward the Th1 and Th2 lineages, respectively, and AT(1) receptor-deficient CD4(+) T cells had enhanced T-bet/GATA-3 expression ratios favoring induction of the Th1 response. Inversely, mice that were unable to mount a Th1 response due to T-bet deficiency were protected from kidney injury in our hypertension model. CONCLUSIONS: The current studies identify an unexpected role for AT(1) receptors on T lymphocytes to protect the kidney in the setting of hypertension by favorably modulating CD4(+) T helper cell differentiation.


Subject(s)
CD4-Positive T-Lymphocytes/metabolism , Hypertension/metabolism , Kidney/metabolism , Receptor, Angiotensin, Type 1/physiology , Animals , CD4-Positive T-Lymphocytes/pathology , Cell Differentiation/immunology , Hypertension/pathology , Hypertension/prevention & control , Kidney/immunology , Kidney/pathology , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Knockout
18.
Kidney Int ; 81(11): 1075-85, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22278020

ABSTRACT

Podocytes are highly differentiated cells that play an important role in maintaining glomerular filtration barrier integrity; a function regulated by small GTPase proteins of the Rho family. To investigate the role of Rho A in podocyte biology, we created transgenic mice expressing doxycycline-inducible constitutively active (V14 Rho) or dominant-negative Rho A (N19 Rho) in podocytes. Specific induction of either Rho A construct in podocytes caused albuminuria and foot process effacement along with disruption of the actin cytoskeleton as evidenced by decreased expression of the actin-associated protein synaptopodin. The mechanisms of these adverse effects, however, appeared to be different. Active V14 Rho enhanced actin polymerization, caused a reduction in nephrin mRNA and protein levels, promoted podocyte apoptosis, and decreased endogenous Rho A levels. In contrast, the dominant-negative N19 Rho caused a loss of podocyte stress fibers, did not alter the expression of either nephrin or Rho A, and did not cause podocyte apoptosis. Thus, our findings suggest that Rho A plays an important role in maintaining the integrity of the glomerular filtration barrier under basal conditions, but enhancement of Rho A activity above basal levels promotes podocyte injury.


Subject(s)
Albuminuria/etiology , Glomerular Filtration Barrier/enzymology , Podocytes/enzymology , rhoA GTP-Binding Protein/metabolism , Actin Cytoskeleton/enzymology , Albuminuria/enzymology , Albuminuria/genetics , Albuminuria/pathology , Animals , Apoptosis , Gene Expression Regulation , Genotype , Glomerular Filtration Barrier/pathology , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice , Mice, Transgenic , Microfilament Proteins/genetics , Microfilament Proteins/metabolism , Mutation , Phenotype , Podocytes/pathology , RNA, Messenger/metabolism , Stress Fibers/enzymology , Time Factors , rhoA GTP-Binding Protein/genetics
19.
Mol Endocrinol ; 25(8): 1376-86, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21622531

ABSTRACT

To determine the role of Gq signaling and calcineurin (CN) activation in promoting apoptosis of glomerular podocytes, constitutively active Gq [Gq(+)] or CN [CN(+)] proteins were introduced into cultured podocytes using protein transduction by tagging the proteins with the transactivator of transcription peptide. To investigate the role of CN in promoting podocyte apoptosis in vivo, a genetic model of type 1 diabetes mellitus (Akita mice) was treated with the CN inhibitor FK506. In cultured podocytes, Gq(+) stimulated nuclear translocation of nuclear factor of activated T cells (NFAT) family members, activated an NFAT reporter construct, and enhanced podocyte apoptosis in a CN-dependent fashion. CN(+) similarly promoted podocyte apoptosis, and apoptosis induced by either angiotensin II or endothelin-1 was blocked by FK506. Induction of apoptosis required NFAT-induced gene transcription because apoptosis induced by either Gq(+) or CN(+) was blocked by an inhibitor that prevented CN-dependent NFAT activation without affecting CN phosphatase activity. Podocyte apoptosis was mediated, in part, by the NFAT-responsive gene cyclooxygenase 2 (COX2) and prostaglandin E(2) generation because apoptosis induced by Gq(+) was attenuated by either COX2 inhibition or blockade of the Gq-coupled E-series prostaglandins receptor. The findings appeared relevant to podocyte apoptosis in diabetic nephropathy because apoptosis was significantly reduced in Akita mice by treatment with FK506. These data suggest that Gq stimulates CN and promotes podocyte apoptosis both in vitro and in vivo. Apoptosis requires NFAT-dependent gene transcription and is mediated, in part, by CN-dependent COX2 induction, prostaglandin E(2) generation, and autocrine activation of the Gq-coupled E-series prostaglandins receptor.


Subject(s)
Apoptosis , Calcineurin/metabolism , Kidney Glomerulus/pathology , Podocytes/metabolism , Podocytes/pathology , Albuminuria/blood , Albuminuria/pathology , Animals , Apoptosis/drug effects , Blood Glucose/metabolism , Cyclooxygenase 2/metabolism , Densitometry , Dinoprostone/metabolism , GTP-Binding Protein alpha Subunits, Gq-G11/metabolism , Mice , Models, Biological , NFATC Transcription Factors/metabolism , Oligopeptides/pharmacology , Podocytes/drug effects , Podocytes/enzymology , Reactive Oxygen Species/metabolism , Signal Transduction/drug effects , Tacrolimus/pharmacology
20.
Mol Endocrinol ; 23(9): 1455-65, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19556343

ABSTRACT

Activation of Wnt signaling pathways causes release and stabilization of the transcription regulator beta-catenin from a destruction complex composed of axin and the adenomatous polyposis coli (APC) protein (canonical signaling pathway). Assembly of this complex is facilitated by a protein-protein interaction between APC and a regulator of G protein signaling (RGS) domain in axin. Because G protein-coupled receptor kinase 2 (GRK2) has a RGS domain that is closely related to the RGS domain in axin, we determined whether GRK2 regulated canonical signaling. We found that GRK2 inhibited Wnt1-induced activation of a reporter construct as well as reduced Wnt3a-dependent stabilization and nuclear translocation of beta-catenin. GRK2 enzymatic activity was required for this negative regulatory effect, and depletion of endogenous GRK2 using small interfering RNA enhanced canonical signaling. GRK2-dependent inhibition of canonical signaling is relevant to osteoblast (OB) biology because overexpression of GRK2 attenuated Wnt/beta-catenin signaling in calvarial OBs. Coimmunoprecipitation studies found that: 1) GRK2 bound APC; 2) The GRK2-APC interaction was promoted by GRK2 enzymatic activity; and 3) Deletion of the RGS domain in GRK2 prevented both the GRK2-APC interaction and GRK2-dependent inhibition of canonical signaling. These data suggest that: 1) GRK2 negatively regulates Wnt signaling; 2) GRK2-dependent inhibition of canonical signaling requires a protein-protein interaction between the RGS domain in GRK2 and APC; and 3) Enzymatic activity promotes the GRK2-APC interaction and is required for the negative regulatory effect on canonical signaling. We speculate that inhibiting GRK2 activity in bone-forming OBs might be a useful therapeutic strategy for increasing bone mass.


Subject(s)
G-Protein-Coupled Receptor Kinase 2/metabolism , Gene Expression Regulation, Enzymologic , Wnt Proteins/antagonists & inhibitors , Animals , Bone and Bones/metabolism , Cell Nucleus/metabolism , Culture Media, Conditioned , Humans , Mice , Models, Biological , Protein Interaction Mapping , Protein Structure, Tertiary , RNA, Small Interfering/metabolism , Signal Transduction , beta Catenin/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...