Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 110
Filter
1.
Metabolomics ; 15(10): 131, 2019 10 01.
Article in English | MEDLINE | ID: mdl-31576432

ABSTRACT

INTRODUCTION: Shiga toxin 2a (Stx2a) induces hemolytic uremic syndrome (STEC HUS) by targeting glomerular endothelial cells (GEC). OBJECTIVES: We investigated in a metabolomic analysis the response of a conditionally immortalized, stable glomerular endothelial cell line (ciGEnC) to Stx2a stimulation as a cell culture model for STEC HUS. METHODS: CiGEnC were treated with tumor necrosis factor-(TNF)α, Stx2a or sequentially with TNFα and Stx2a. We performed a metabolomic high-throughput screening by lipid- or gas chromatography and subsequent mass spectrometry. Metabolite fold changes in stimulated ciGEnC compared to untreated cells were calculated. RESULTS: 320 metabolites were identified and investigated. In response to TNFα + Stx2a, there was a predominant increase in intracellular free fatty acids and amino acids. Furthermore, lipid- and protein derived pro-inflammatory mediators, oxidative stress and an augmented intracellular energy turnover were increased in ciGEnC. Levels of most biochemicals related to carbohydrate metabolism remained unchanged. CONCLUSION: Stimulation of ciGEnC with TNFα + Stx2a is associated with profound metabolic changes indicative of increased inflammation, oxidative stress and energy turnover.


Subject(s)
Endothelial Cells/drug effects , Endothelial Cells/metabolism , Kidney Glomerulus/cytology , Metabolomics , Shiga Toxin 2/pharmacology , Cell Count , Cell Survival/drug effects , Cells, Cultured , Endothelial Cells/cytology , Humans , Inflammation/chemically induced , Inflammation/metabolism , Inflammation/pathology , Lipopolysaccharides , Multivariate Analysis , Shiga Toxin 2/metabolism
2.
Diabetes ; 68(1): 172-187, 2019 01.
Article in English | MEDLINE | ID: mdl-30389746

ABSTRACT

Elevated levels of vascular endothelial growth factor (VEGF) A are thought to cause glomerular endothelial cell (GEnC) dysfunction and albuminuria in diabetic nephropathy. We hypothesized that VEGFC could counteract these effects of VEGFA to protect the glomerular filtration barrier and reduce albuminuria. Isolated glomeruli were stimulated ex vivo with VEGFC, which reduced VEGFA- and type 2 diabetes-induced glomerular albumin solute permeability (Ps'alb). VEGFC had no detrimental effect on glomerular function in vivo when overexpression was induced locally in podocytes (podVEGFC) in otherwise healthy mice. Further, these mice had reduced glomerular VEGFA mRNA expression, yet increased glomerular VEGF receptor heterodimerization, indicating differential signaling by VEGFC. In a model of type 1 diabetes, the induction of podVEGFC overexpression reduced the development of hypertrophy, albuminuria, loss of GEnC fenestrations and protected against altered VEGF receptor expression. In addition, VEGFC protected against raised Ps'alb by endothelial glycocalyx disruption in glomeruli. In summary, VEGFC reduced the development of diabetic nephropathy, prevented VEGF receptor alterations in the diabetic glomerulus, and promoted both glomerular protection and endothelial barrier function. These important findings highlight a novel pathway for future investigation in the treatment of diabetic nephropathy.


Subject(s)
Diabetic Nephropathies/metabolism , Animals , Blotting, Western , Cells, Cultured , Diabetes Mellitus, Type 1/genetics , Diabetes Mellitus, Type 1/metabolism , Diabetic Nephropathies/genetics , Fluorescent Antibody Technique , Genotype , Humans , Immunoprecipitation , Podocytes/metabolism , Receptors, Vascular Endothelial Growth Factor/genetics , Receptors, Vascular Endothelial Growth Factor/metabolism , Vascular Endothelial Growth Factor Receptor-3/genetics , Vascular Endothelial Growth Factor Receptor-3/metabolism
3.
Front Immunol ; 9: 3008, 2018.
Article in English | MEDLINE | ID: mdl-30619356

ABSTRACT

Atypical hemolytic uremic syndrome (aHUS) is a severe disease characterized by microvascular endothelial cell (EC) lesions leading to thrombi formation, mechanical hemolysis and organ failure, predominantly renal. Complement system overactivation is a hallmark of aHUS. To investigate this selective susceptibility of the microvascular renal endothelium to complement attack and thrombotic microangiopathic lesions, we compared complement and cyto-protection markers on EC, from different vascular beds, in in vitro and in vivo models as well as in patients. No difference was observed for complement deposits or expression of complement and coagulation regulators between macrovascular and microvascular EC, either at resting state or after inflammatory challenge. After prolonged exposure to hemolysis-derived heme, higher C3 deposits were found on glomerular EC, in vitro and in vivo, compared with other EC in culture and in mice organs (liver, skin, brain, lungs and heart). This could be explained by a reduced complement regulation capacity due to weaker binding of Factor H and inefficient upregulation of thrombomodulin (TM). Microvascular EC also failed to upregulate the cytoprotective heme-degrading enzyme heme-oxygenase 1 (HO-1), normally induced by hemolysis products. Only HUVEC (Human Umbilical Vein EC) developed adaptation to heme, which was lost after inhibition of HO-1 activity. Interestingly, the expression of KLF2 and KLF4-known transcription factors of TM, also described as possible transcription modulators of HO-1- was weaker in micro than macrovascular EC under hemolytic conditions. Our results show that the microvascular EC, and especially glomerular EC, fail to adapt to the stress imposed by hemolysis and acquire a pro-coagulant and complement-activating phenotype. Together, these findings indicate that the vulnerability of glomerular EC to hemolysis is a key factor in aHUS, amplifying complement overactivation and thrombotic microangiopathic lesions.


Subject(s)
Atypical Hemolytic Uremic Syndrome/immunology , Complement C3/immunology , Heme Oxygenase-1/metabolism , Heme/immunology , Kidney Glomerulus/immunology , Animals , Atypical Hemolytic Uremic Syndrome/blood , Atypical Hemolytic Uremic Syndrome/pathology , Biopsy , Complement Activation , Disease Models, Animal , Disease Susceptibility/immunology , Endothelial Cells/immunology , Endothelium, Vascular/cytology , Endothelium, Vascular/immunology , Female , Heme/metabolism , Hemolysis/immunology , Human Umbilical Vein Endothelial Cells , Humans , Kidney Glomerulus/blood supply , Kidney Glomerulus/cytology , Kidney Glomerulus/pathology , Kruppel-Like Factor 4 , Membrane Proteins/metabolism , Mice , Mice, Inbred C57BL , Microvessels/cytology , Microvessels/immunology , Primary Cell Culture , Thrombomodulin/metabolism , Up-Regulation
4.
Postgrad Med J ; 93(1106): 717-718, 2017 12.
Article in English | MEDLINE | ID: mdl-28778952

Subject(s)
Medicine , Humans
5.
Sci Rep ; 6: 35671, 2016 10 24.
Article in English | MEDLINE | ID: mdl-27774996

ABSTRACT

Glucocorticoids are steroids that reduce inflammation and are used as immunosuppressive drugs for many diseases. They are also the mainstay for the treatment of minimal change nephropathy (MCN), which is characterised by an absence of inflammation. Their mechanisms of action remain elusive. Evidence suggests that immunomodulatory drugs can directly act on glomerular epithelial cells or 'podocytes', the cell type which is the main target of injury in MCN. To understand the nature of glucocorticoid effects on non-immune cell functions, we generated RNA sequencing data from human podocyte cell lines and identified the genes that are significantly regulated in dexamethasone-treated podocytes compared to vehicle-treated cells. The upregulated genes are of functional relevance to cytoskeleton-related processes, whereas the downregulated genes mostly encode pro-inflammatory cytokines and growth factors. We observed a tendency for dexamethasone-upregulated genes to be downregulated in MCN patients. Integrative analysis revealed gene networks composed of critical signaling pathways that are likely targeted by dexamethasone in podocytes.


Subject(s)
Epithelial Cells/drug effects , Gene Regulatory Networks/genetics , Glucocorticoids/genetics , Podocytes/metabolism , RNA/genetics , Signal Transduction/genetics , Cells, Cultured , Dexamethasone/pharmacology , Down-Regulation/drug effects , Down-Regulation/genetics , Epithelial Cells/metabolism , Gene Regulatory Networks/drug effects , Humans , Podocytes/drug effects , Sequence Analysis, RNA/methods , Signal Transduction/drug effects , Up-Regulation/drug effects , Up-Regulation/genetics
6.
Nat Microbiol ; 1: 16004, 2016 Feb 22.
Article in English | MEDLINE | ID: mdl-27572168

ABSTRACT

Middle East respiratory syndrome coronavirus (MERS-CoV) causes sporadic zoonotic disease and healthcare-associated outbreaks in human. MERS is often complicated by acute respiratory distress syndrome (ARDS) and multi-organ failure(1,2). The high incidence of renal failure in MERS is a unique clinical feature not often found in other human coronavirus infections(3,4). Whether MERS-CoV infects the kidney and how it triggers renal failure are not understood(5,6). Here, we demonstrated renal infection and apoptotic induction by MERS-CoV in human ex vivo organ culture and a nonhuman primate model. High-throughput analysis revealed that the cellular genes most significantly perturbed by MERS-CoV have previously been implicated in renal diseases. Furthermore, MERS-CoV induced apoptosis through upregulation of Smad7 and fibroblast growth factor 2 (FGF2) expression in both kidney and lung cells. Conversely, knockdown of Smad7 effectively inhibited MERS-CoV replication and protected cells from virus-induced cytopathic effects. We further demonstrated that hyperexpression of Smad7 or FGF2 induced a strong apoptotic response in kidney cells. Common marmosets infected by MERS-CoV developed ARDS and disseminated infection in kidneys and other organs. Smad7 and FGF2 expression were elevated in the lungs and kidneys of the infected animals. Our results provide insights into the pathogenesis of MERS-CoV and host targets for treatment.


Subject(s)
Apoptosis , Coronavirus Infections/pathology , Fibroblast Growth Factor 2/metabolism , Kidney/pathology , Lung/pathology , Middle East Respiratory Syndrome Coronavirus/pathogenicity , Smad7 Protein/metabolism , Animals , Callithrix , Cytopathogenic Effect, Viral , Disease Models, Animal , Host-Pathogen Interactions , Humans , Organ Culture Techniques
7.
Clin Exp Nephrol ; 20(5): 679-688, 2016 Oct.
Article in English | MEDLINE | ID: mdl-26715508

ABSTRACT

BACKGROUND: Fibrin deposition within glomeruli is commonly seen in kidney biopsy specimens, suggesting enhanced coagulant activity. Tissue factor (TF) is a coagulation factor which is also related to various biological effects, and TF is upregulated by hypoxia in cancer cells. Recently, hypoxic podocyte injury has been proposed, therefore, we investigated TF expression in hypoxia. METHODS: Conditionally immortalized human podocytes were differentiated and treated under hypoxic or normoxic conditions. mRNA expressions of TF and tissue factor pathway inhibitor (TFPI) were analyzed by quantitative RT-PCR. Protein levels of TF and TFPI were tested by enzyme-linked immunosorbent assay. We employed small interfering RNA (siRNA) to temporary knockdown early growth response protein 1 (Egr-1), hypoxia-inducible factor-1α (HIF-1α) and TF. The expression of CD2-associated protein (CD2AP) mRNA and phalloidin staining was examined to assess podocyte injury. RESULTS: Hypoxia increased mRNA expression of TF (6 h: 2.3 ± 0.05 fold, p < 0.001, 24 h: 5.6 ± 2.4 fold, p < 0.05) and suppressed TFPI (6 h: 0.54 ± 0.04 fold, p < 0.05, 24 h: 0.24 ± 0.06 fold, p < 0.001) compared with normoxia. Similarly, protein levels of TF were increased and TFPI were decreased. Egr-1 siRNA did not change TF mRNA expression. Pyrrolidine dithiocarbamate (PDTC), a nuclear factor kappa B (NF-κB) inhibitor, significantly reduced hypoxia induced TF expression, and HIF-1α knockdown further increased TF. Hypoxia resulted in decreased CD2AP and actin reorganization in podocytes, and these changes were attenuated by TF siRNA. CONCLUSION: Hypoxia increased the expression of TF in human podocytes NF-κB dependently. TF may have a critical role in the hypoxic podocyte injury.


Subject(s)
NF-kappa B/metabolism , Oxygen/metabolism , Podocytes/metabolism , Thromboplastin/metabolism , Actin Cytoskeleton/metabolism , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Cell Hypoxia , Cell Line , Cobalt/pharmacology , Cytoskeletal Proteins/genetics , Cytoskeletal Proteins/metabolism , Early Growth Response Protein 1/genetics , Early Growth Response Protein 1/metabolism , Fluorescent Antibody Technique , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Lipoproteins/genetics , Lipoproteins/metabolism , NF-kappa B/antagonists & inhibitors , Phalloidine/metabolism , Podocytes/drug effects , Podocytes/pathology , Pyrrolidines/pharmacology , RNA Interference , RNA, Messenger/genetics , RNA, Messenger/metabolism , Signal Transduction , Thiocarbamates/pharmacology , Thromboplastin/genetics , Time Factors , Transfection , Up-Regulation
8.
Clin Sci (Lond) ; 128(12): 883-93, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25626449

ABSTRACT

Minimal change nephropathy (MCN) is the third most common cause of primary nephrotic syndrome in adults. Most patients with MCN respond to corticosteroid therapy, but relapse is common. In children, steroid-dependent patients are often given alternative agents to spare the use of steroids and to avoid the cumulative steroid toxicity. In this respect, levamisole has shown promise due to its ability to effectively maintain remission in children with steroid-sensitive or steroid-dependent nephrotic syndrome. Despite clinical effectiveness, there is a complete lack of molecular evidence to explain its mode of action and there are no published reports on the use of this compound in adult patients. We studied the effectiveness of levamisole in a small cohort of adult patients and also tested the hypothesis that levamisole's mode of action is attributable to its direct effects on podocytes. In the clinic, we demonstrate that in our adult patients, cohort levamisole is generally well tolerated and clinically useful. Using conditionally immortalized human podocytes, we show that levamisole is able to induce expression of glucocorticoid receptor (GR) and to activate GR signalling. Furthermore, levamisole is able to protect against podocyte injury in a puromycin aminonucleoside (PAN)-treated cell model. In this model the effects of levamisole are blocked by the GR antagonist mifepristone (RU486), suggesting that GR signalling is a critical target of levamisole's action. These results indicate that levamisole is effective in nephrotic syndrome in adults, as well as in children, and point to molecular mechanisms for this drug's actions in podocyte diseases.


Subject(s)
Glucocorticoids/therapeutic use , Levamisole/therapeutic use , Nephrotic Syndrome/drug therapy , Adolescent , Adult , Cells, Cultured/drug effects , Drug Therapy, Combination , Female , Humans , Levamisole/adverse effects , Levamisole/antagonists & inhibitors , Levamisole/pharmacology , Male , Middle Aged , Mifepristone/pharmacology , Nephrotic Syndrome/metabolism , Nephrotic Syndrome/pathology , Off-Label Use , Podocytes/drug effects , Podocytes/metabolism , Prednisolone/therapeutic use , Puromycin Aminonucleoside/antagonists & inhibitors , Puromycin Aminonucleoside/pharmacology , Receptors, Glucocorticoid/metabolism , Signal Transduction/drug effects , Young Adult
9.
Cell Signal ; 27(3): 460-9, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25542307

ABSTRACT

In the present study, we evaluated the effect of short term hyperglycemia on renal lesions in a mouse model (Tg26) of HIV-associated nephropathy (HIVAN). Control and Tg26 mice in groups (n=6) were administered either normal saline (FVBN or Tg) or streptozotocin (FVBN+STZ or Tg26+STZ). After two weeks, biomarkers were collected and kidneys were harvested. FVBN+ STZ and Tg26+STZ displayed elevated serum glucose levels when compared to FVBN and Tg26 respectively. Tg26+STZ displayed elevated (P<0.05) blood urea nitrogen (BUN) levels (P<0.05) and enhanced (P<0.01) proteinuria when compared to Tg26. Tg26+STZ displayed enhanced (P<0.001) number of sclerotic glomeruli and microcysts vs. Tg26. Renal tissues of Tg26 displayed down regulation of vitamin D receptor (VDR) expression and enhanced Ang II production when compared to FVBN mice. Hyperglycemia exacerbated down regulation of VDR and production of Ang II in FVBN and Tg mice. Hyperglycemia increased kidney cell reactive oxygen species (ROS) production and oxidative DNA damage in both FVBN and Tg26 mice. In in vitro studies, HIV down regulated podocyte VDR expression and also enhanced renin angiotensin system activation. In addition, both glucose and HIV stimulated kidney cell ROS generation and DNA damage and compromised DNA repair; however, tempol (superoxide dismutase mimetic), losartan (Ang II blocker) and EB1089 (VDR agonist) provided protection against DNA damaging effects of glucose and HIV. These findings indicated that glucose activated the RAS and inflicted oxidative stress-mediated DNA damage via down regulation of kidney cell VDR expression in HIV milieu both in vivo and in vitro.


Subject(s)
AIDS-Associated Nephropathy/pathology , Hyperglycemia/pathology , Kidney Glomerulus/metabolism , Receptors, Calcitriol/metabolism , AIDS-Associated Nephropathy/metabolism , Angiotensinogen/genetics , Angiotensinogen/metabolism , Animals , Cells, Cultured , Down-Regulation/drug effects , Glucose/pharmacology , HEK293 Cells , HIV-1/metabolism , Humans , Hyperglycemia/chemically induced , Hyperglycemia/metabolism , Kidney Glomerulus/cytology , Kidney Glomerulus/drug effects , Mice , Mice, Transgenic , Podocytes/cytology , Podocytes/drug effects , Podocytes/metabolism , Proteinuria/etiology , Reactive Oxygen Species/metabolism , Receptors, Calcitriol/genetics , Streptozocin/toxicity , gag Gene Products, Human Immunodeficiency Virus/genetics , gag Gene Products, Human Immunodeficiency Virus/metabolism , pol Gene Products, Human Immunodeficiency Virus/genetics , pol Gene Products, Human Immunodeficiency Virus/metabolism
10.
J Am Soc Nephrol ; 26(2): 339-48, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25012173

ABSTRACT

Although APOL1 gene variants are associated with nephropathy in African Americans, little is known about APOL1 protein synthesis, uptake, and localization in kidney cells. To address these questions, we examined APOL1 protein and mRNA localization in human kidney and human kidney-derived cell lines. Indirect immunofluorescence microscopy performed on nondiseased nephrectomy cryosections from persons with normal kidney function revealed that APOL1 protein was markedly enriched in podocytes (colocalized with synaptopodin and Wilms' tumor suppressor) and present in lower abundance in renal tubule cells. Fluorescence in situ hybridization detected APOL1 mRNA in glomeruli (podocytes and endothelial cells) and tubules, consistent with endogenous synthesis in these cell types. When these analyses were extended to renal-derived cell lines, quantitative RT-PCR did not detect APOL1 mRNA in human mesangial cells; however, abundant levels of APOL1 mRNA were observed in proximal tubule cells and glomerular endothelial cells, with lower expression in podocytes. Western blot analysis revealed corresponding levels of APOL1 protein in these cell lines. To explain the apparent discrepancy between the marked abundance of APOL1 protein in kidney podocytes observed in cryosections versus the lesser abundance in podocyte cell lines, we explored APOL1 cellular uptake. APOL1 protein was taken up readily by human podocytes in vitro but was not taken up efficiently by mesangial cells, glomerular endothelial cells, or proximal tubule cells. We hypothesize that the higher levels of APOL1 protein in human cryosectioned podocytes may reflect both endogenous protein synthesis and APOL1 uptake from the circulation or glomerular filtrate.


Subject(s)
Apolipoproteins/metabolism , Kidney Glomerulus/metabolism , Kidney Tubules, Proximal/metabolism , Kidney/metabolism , Lipoproteins, HDL/metabolism , Mesangial Cells/metabolism , RNA, Messenger/metabolism , Apolipoprotein L1 , Biopsy , Cell Line , Cells, Cultured , Epithelial Cells/metabolism , Epithelial Cells/pathology , Humans , In Vitro Techniques , Kidney/pathology , Kidney/surgery , Kidney Glomerulus/pathology , Kidney Tubules, Proximal/pathology , Mesangial Cells/pathology , Microscopy, Fluorescence , Nephrectomy , Podocytes/metabolism , Podocytes/pathology
11.
FASEB J ; 28(11): 4686-99, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25122554

ABSTRACT

The endothelial surface glycocalyx is a hydrated mesh in which proteoglycans are prominent. It is damaged in diseases associated with elevated levels of tumor necrosis factor α (TNF-α). We investigated the mechanism of TNF-α-induced disruption of the glomerular endothelial glycocalyx. We used conditionally immortalized human glomerular endothelial cells (GEnCs), quantitative PCR arrays, Western blotting, immunoprecipitation, immunofluorescence, and dot blots to examine the effects of TNF-α. TNF-α induced syndecan 4 (SDC4) mRNA up-regulation by 2.5-fold, whereas cell surface SDC4 and heparan sulfate (HS) were reduced by 36 and 30%, respectively, and SDC4 and sulfated glycosaminoglycan in the culture medium were increased by 52 and 65%, respectively, indicating TNF-α-induced shedding. Small interfering (siRNA) knockdown of SDC4 (by 52%) caused a corresponding loss of cell surface HS of similar magnitude (38%), and immunoprecipitation demonstrated that SDC4 and HS are shed as intact proteoglycan ectodomains. All of the effects of TNF-α on SDC4 and HS were abrogated by the metalloproteinase (MMP) inhibitor batimastat. Also abrogated was the associated 37% increase in albumin passage across GEnC monolayers. Specific MMP9 knockdown by siRNA similarly blocked TNF-α effects. SDC4 is the predominant HS proteoglycan in the GEnC glycocalyx. TNF-α-induced MMP9-mediated shedding of SDC4 is likely to contribute to the endothelial glycocalyx disruption observed in diabetes and inflammatory states.


Subject(s)
Glycocalyx/metabolism , Matrix Metalloproteinase 9/metabolism , Syndecan-4/metabolism , Tumor Necrosis Factor-alpha/pharmacology , Cell Membrane , Cells, Cultured , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Gene Expression/physiology , Gene Knockdown Techniques , Humans , Matrix Metalloproteinase 9/genetics , Proteoglycans/metabolism
12.
Am J Physiol Renal Physiol ; 307(3): F326-36, 2014 Aug 01.
Article in English | MEDLINE | ID: mdl-24899058

ABSTRACT

Development of higher rates of nondiabetic glomerulosclerosis (GS) in African Americans has been attributed to two coding sequence variants (G1 and G2) in the APOL1 gene. To date, the cellular function and the role of APOL1 variants (Vs) in GS are still unknown. In this study, we examined the effects of overexpressing wild-type (G0) and kidney disease risk variants (G1 and G2) of APOL1 in human podocytes using a lentivirus expression system. Interestingly, G0 inflicted podocyte injury only at a higher concentration; however, G1 and G2 promoted moderate podocyte injury at lower and higher concentrations. APOL1Vs expressing podocytes displayed diffuse distribution of both Lucifer yellow dye and cathepsin L as manifestations of enhanced lysosomal membrane permeability (LMP). Chloroquine attenuated the APOL1Vs-induced increase in podocyte injury, consistent with targeting lysosomes. The chloride channel blocker DIDS prevented APOL1Vs- induced injury, indicating a role for chloride influx in osmotic swelling of lysosomes. Direct exposure of noninfected podocytes with conditioned media from G1- and G2-expressing podocytes also induced injury, suggesting a contributory role of the secreted component of G1 and G2 as well. Adverse host factors (AHFs) such as hydrogen peroxide, hypoxia, TNF-α, and puromycin aminonucleoside augmented APOL1- and APOL1Vs-induced podocyte injury, while the effect of human immunodeficiency virus (HIV) on podocyte injury was overwhelming under conditions of APOLVs expression. We conclude that G0 and G1 and G2 APOL1 variants have the potential to induce podocyte injury in a manner which is further augmented by AHFs, with HIV infection being especially prominent.


Subject(s)
Apolipoproteins/genetics , Apolipoproteins/metabolism , Genetic Variation/genetics , Lipoproteins, HDL/genetics , Lipoproteins, HDL/metabolism , Lysosomes/physiology , Podocytes/metabolism , Podocytes/pathology , 4,4'-Diisothiocyanostilbene-2,2'-Disulfonic Acid/pharmacology , Actins/metabolism , Black or African American/ethnology , Black or African American/genetics , Apolipoprotein L1 , Cells, Cultured , Chloride Channels/antagonists & inhibitors , Chloride Channels/drug effects , Chloroquine/pharmacology , Genetic Predisposition to Disease/ethnology , Genetic Predisposition to Disease/genetics , Glomerulosclerosis, Focal Segmental/ethnology , Glomerulosclerosis, Focal Segmental/genetics , Humans , Necrosis/physiopathology , Permeability , Podocytes/drug effects
13.
J Am Soc Nephrol ; 25(5): 953-66, 2014 May.
Article in English | MEDLINE | ID: mdl-24436469

ABSTRACT

The glomerular basement membrane (GBM) is a specialized extracellular matrix (ECM) compartment within the glomerulus that contains tissue-restricted isoforms of collagen IV and laminin. It is integral to the capillary wall and therefore, functionally linked to glomerular filtration. Although the composition of the GBM has been investigated with global and candidate-based approaches, the relative contributions of glomerular cell types to the production of ECM are not well understood. To characterize specific cellular contributions to the GBM, we used mass spectrometry-based proteomics to analyze ECM isolated from podocytes and glomerular endothelial cells in vitro. These analyses identified cell type-specific differences in ECM composition, indicating distinct contributions to glomerular ECM assembly. Coculture of podocytes and endothelial cells resulted in an altered composition and organization of ECM compared with monoculture ECMs, and electron microscopy revealed basement membrane-like ECM deposition between cocultured cells, suggesting the involvement of cell-cell cross-talk in the production of glomerular ECM. Notably, compared with monoculture ECM proteomes, the coculture ECM proteome better resembled a tissue-derived glomerular ECM dataset, indicating its relevance to GBM in vivo. Protein network analyses revealed a common core of 35 highly connected structural ECM proteins that may be important for glomerular ECM assembly. Overall, these findings show the complexity of the glomerular ECM and suggest that both ECM composition and organization are context-dependent.


Subject(s)
Extracellular Matrix Proteins/physiology , Extracellular Matrix/physiology , Kidney Glomerulus/physiology , Receptor Cross-Talk/physiology , Cells, Cultured , Coculture Techniques , Culture Media, Conditioned , Extracellular Matrix/metabolism , Extracellular Matrix/ultrastructure , Extracellular Matrix Proteins/biosynthesis , Humans , Kidney Glomerulus/metabolism , Kidney Glomerulus/ultrastructure , Phenotype , Podocytes/physiology , Protein Interaction Maps
14.
Am J Pathol ; 183(2): 604-16, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23770346

ABSTRACT

Damage to endothelial glycocalyx impairs vascular barrier function and may contribute to progression of chronic vascular disease. An early indicator is microalbuminuria resulting from glomerular filtration barrier damage. We investigated the contributions of hyaluronic acid (HA) and chondroitin sulfate (CS) to glomerular microvascular endothelial cell (GEnC) glycocalyx and examined whether these are modified by vascular endothelial growth factors A and C (VEGFA and VEGFC). HA and CS were imaged on GEnCs and their resynthesis was examined. The effect of HA and CS on transendothelial electrical resistance (TEER) and labeled albumin flux across monolayers was assessed. Effects of VEGFA and VEGFC on production and charge characteristics of glycosaminoglycan (GAG) were examined via metabolic labeling and liquid chromatography. GAG shedding was quantified using Alcian Blue. NDST2 expression was examined using real-time PCR. GEnCs expressed HA and CS in the glycocalyx. CS contributed to the barrier to both ion (TEER) and protein flux across the monolayer; HA had only a limited effect. VEGFC promoted HA synthesis and increased the charge density of synthesized GAGs. In contrast, VEGFA induced shedding of charged GAGs. CS plays a role in restriction of macromolecular flux across GEnC monolayers, and VEGFA and VEGFC differentially regulate synthesis, charge, and shedding of GAGs in GEnCs. These observations have important implications for endothelial barrier regulation in glomerular and other microvascular beds.


Subject(s)
Chondroitin Sulfates/metabolism , Glycosaminoglycans/metabolism , Hyaluronic Acid/metabolism , Kidney Glomerulus/blood supply , Vascular Endothelial Growth Factor A/physiology , Vascular Endothelial Growth Factor C/physiology , Cells, Cultured , Endothelial Cells/metabolism , Glycocalyx/metabolism , Humans , Kidney Glomerulus/metabolism , Microvessels/metabolism , Real-Time Polymerase Chain Reaction
16.
Am J Physiol Renal Physiol ; 305(3): F343-54, 2013 Aug 01.
Article in English | MEDLINE | ID: mdl-23678040

ABSTRACT

Oxidative stress has been implicated to contribute to HIV-induced kidney cell injury; however, the role of p53, a modulator of oxidative stress, has not been evaluated in the development of HIV-associated nephropathy (HIVAN). We hypothesized that mammalian target of rapamycin (mTOR) may be critical for the induction of p53-mediated oxidative kidney cell injury in HIVAN. To test our hypothesis, we evaluated the effect of an mTOR inhibitor, rapamycin, on kidney cell p53 expression, downstream signaling, and kidney cell injury in both in vivo and in vitro studies. Inhibition of the mTOR pathway resulted in downregulation of renal tissue p53 expression, associated downstream signaling, and decreased number of sclerosed glomeruli, tubular microcysts, and apoptosed and 8-hydroxy deoxyguanosine (8-OHdG)-positive (+ve) cells in Tg26 mice. mTOR inhibition not only attenuated kidney cell expression of p66ShcA and phospho-p66ShcA but also reactivated the redox-sensitive stress response program in the form of enhanced expression of manganese superoxide dismutase (MnSOD) and catalase. In in vitro studies, the mTOR inhibitor also provided protection against HIV-induced podocyte apoptosis. Moreover, mTOR inhibition downregulated HIV-induced podocyte (HP/HIV) p53 expression. Since HP/HIV silenced for mTOR displayed a lack of expression of p53 as well as attenuated podocyte apoptosis, this suggests that mTOR is critical for kidney cell p53 activation and associated oxidative kidney cell injury in the HIV milieu.


Subject(s)
AIDS-Associated Nephropathy/pathology , Acute Kidney Injury/etiology , Acute Kidney Injury/pathology , HIV Infections/complications , HIV Infections/pathology , Oxidative Stress/physiology , TOR Serine-Threonine Kinases/physiology , Tumor Suppressor Protein p53/physiology , 8-Hydroxy-2'-Deoxyguanosine , Animals , Apoptosis/physiology , Catalase/metabolism , Deoxyguanosine/analogs & derivatives , Deoxyguanosine/metabolism , Gene Silencing , Humans , Immunohistochemistry , In Situ Nick-End Labeling , Mice , Mice, Transgenic , Podocytes/pathology , Rats , Real-Time Polymerase Chain Reaction , Signal Transduction/physiology , Superoxide Dismutase/metabolism
17.
Pediatr Nephrol ; 28(9): 1803-12, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23689904

ABSTRACT

BACKGROUND: Minimal change disease (MCD) is the most common cause of nephrotic syndrome in children and is associated with the expression of CD80 in podocytes and the increased excretion of CD80 in urine. We hypothesized that serum from patients with MCD might stimulate CD80 expression in cultured podocytes. METHODS: Sera and peripheral blood mononuclear cells (PBMCs) were collected from subjects with MCD in relapse and remission and from normal controls. Immortalized human podocytes were incubated with culture media containing patient sera or supernatants from patient and control PBMC cultures. CD80 expression was measured by quantitative PCR and western blot analysis. RESULTS: Sera collected from patients with MCD in relapse, but not in remission, significantly increased CD80 expression (mean ± standard deviation: 1.8 ± 0.7 vs. 0.8 ± 0.2; p < 0.004) and CD80 protein secretion by podocytes (p < 0.05 between relapse and normal controls). No such CD80 increase was observed when podocytes were incubated with supernatants of PBMC cultures from patients in relapse. CONCLUSIONS: Sera from MCD patients in relapse, but not in remission, stimulated CD80 expression in cultured podocytes. Identifying this factor in sera could provide insights into the pathogenesis of this disorder. No role in CD80 expression by podocytes was found for cytokines released by PBMCs.


Subject(s)
B7-1 Antigen/biosynthesis , Nephrosis, Lipoid/metabolism , Podocytes/metabolism , Adolescent , Anti-Inflammatory Agents/therapeutic use , Blotting, Western , Cells, Cultured , Child , Child, Preschool , Cytokines/metabolism , Dose-Response Relationship, Drug , Enzyme-Linked Immunosorbent Assay , Female , Glyceraldehyde 3-Phosphate Dehydrogenase (NADP+)/metabolism , Humans , Kidney Function Tests , Male , Monocytes/metabolism , Nephrosis, Lipoid/blood , Nephrosis, Lipoid/drug therapy , Prednisone/therapeutic use , RNA/biosynthesis , RNA/genetics , RNA/isolation & purification , Real-Time Polymerase Chain Reaction , Recurrence , Serum , Young Adult
18.
Exp Cell Res ; 319(13): 2073-2080, 2013 Aug 01.
Article in English | MEDLINE | ID: mdl-23611955

ABSTRACT

Recent studies suggested that miRNAs are involved in the development of the pathogenesis of HIV-associated nephropathy (HIVAN). Rapamycin, a widely used mTOR inhibitor, has been demonstrated to slow down the progression of HIVAN. However, the role of miRNA in the regulation of these processes has not been investigated so far. In the current study, we have used a microarray-based approach in combination with real-time PCR to profile the miRNA expression patterns in rapamycin-treated HIVAN mice (Tg26). Our results demonstrated that 19 miRNAs belonging to 13 different families expressed differentially in renal tissues of rapamycin-receiving Tg26 mice when compared to Tg26 mice-receiving saline only. The patterns of miRNAs expression in rapamycin-receiving Tg26 mice took a reverse turn. These miRNAs were classified into 8 functional categories. In in vitro studies, we examined the expression of specific miRNAs in HIV-1 transduced human podocytes (HIV/HPs). HIV/HPs displayed attenuation of expression of miR-99a, -100a, -199a and miR-200, whereas, rapamycin inhibited this effect of HIV. These findings suggest that rapamycin-mediated up-regulation of specific miRNAs could contribute to amelioration of renal lesions in HIVAN mice.


Subject(s)
AIDS-Associated Nephropathy/genetics , MicroRNAs/genetics , Sirolimus/pharmacology , AIDS-Associated Nephropathy/pathology , AIDS-Associated Nephropathy/prevention & control , Animals , Cells, Cultured , Disease Progression , Drug Evaluation, Preclinical , Female , Gene Expression Regulation/drug effects , HIV-1/physiology , HeLa Cells , Humans , Immunosuppressive Agents/pharmacology , Male , Mice , Mice, Transgenic
19.
PLoS One ; 8(3): e55748, 2013.
Article in English | MEDLINE | ID: mdl-23555556

ABSTRACT

Morphine has been reported to accelerate the progression of chronic kidney disease. However, whether morphine affects slit diaphragm (SD), the major constituent of glomerular filtration barrier, is still unclear. In the present study, we examined the effect of morphine on glomerular filtration barrier in general and podocyte integrity in particular. Mice were administered either normal saline or morphine for 72 h, then urine samples were collected and kidneys were subsequently isolated for immunohistochemical studies and Western blot. For in vitro studies, human podocytes were treated with morphine and then probed for the molecular markers of slit diaphragm. Morphine-receiving mice displayed a significant increase in albuminuria and showed effacement of podocyte foot processes. In both in vivo and in vitro studies, the expression of synaptopodin, a molecular marker for podocyte integrity, and the slit diaphragm constituting molecules (SDCM), such as nephrin, podocin, and CD2-associated protein (CD2AP), were decreased in morphine-treated podocytes. In vitro studies indicated that morphine modulated podocyte expression of SDCM through opiate mu (MOR) and kappa (KOR) receptors. Since morphine also enhanced podocyte oxidative stress, the latter seems to contribute to decreased SDCM expression. In addition, AKT, p38, and JNK pathways were involved in morphine-induced down regulation of SDCM in human podocytes. These findings demonstrate that morphine has the potential to alter the glomerular filtration barrier by compromising the integrity of podocytes.


Subject(s)
Albuminuria/metabolism , Morphine/adverse effects , Narcotics/adverse effects , Podocytes/metabolism , Adaptor Proteins, Signal Transducing/biosynthesis , Albuminuria/chemically induced , Albuminuria/pathology , Animals , Cell Line, Transformed , Cytoskeletal Proteins/biosynthesis , Gene Expression Regulation/drug effects , Glomerular Filtration Rate/drug effects , Humans , Intracellular Signaling Peptides and Proteins/biosynthesis , MAP Kinase Kinase 4/metabolism , MAP Kinase Signaling System/drug effects , Membrane Proteins/biosynthesis , Mice , Morphine/pharmacology , Narcotics/pharmacology , Podocytes/pathology , Proto-Oncogene Proteins c-akt/metabolism , Receptors, Opioid, kappa/biosynthesis , Receptors, Opioid, mu/biosynthesis , Renal Insufficiency, Chronic/metabolism , Renal Insufficiency, Chronic/pathology , p38 Mitogen-Activated Protein Kinases/metabolism
20.
PLoS One ; 8(2): e55852, 2013.
Article in English | MEDLINE | ID: mdl-23457483

ABSTRACT

Reactive oxygen species (ROS) play a key role in the pathogenesis of proteinuria in glomerular diseases like diabetic nephropathy. Glomerular endothelial cell (GEnC) glycocalyx covers the luminal aspect of the glomerular capillary wall and makes an important contribution to the glomerular barrier. ROS are known to depolymerise glycosaminoglycan (GAG) chains of proteoglycans, which are crucial for the barrier function of GEnC glycocalyx. The aim of this study is to investigate the direct effects of ROS on the structure and function of GEnC glycocalyx using conditionally immortalised human GEnC. ROS were generated by exogenous hydrogen peroxide. Biosynthesis and cleavage of GAG chains was analyzed by radiolabelling (S(35) and (3)H-glucosamine). GAG chains were quantified on GEnC surface and in the cell supernatant using liquid chromatography and immunofluorescence techniques. Barrier properties were estimated by measuring trans-endothelial passage of albumin. ROS caused a significant loss of WGA lectin and heparan sulphate staining from the surface of GEnC. This lead to an increase in trans-endothelial albumin passage. The latter could be inhibited by catalase and superoxide dismutase. The effect of ROS on GEnC was not mediated via the GAG biosynthetic pathway. Quantification of radiolabelled GAG fractions in the supernatant confirmed that ROS directly caused shedding of HS GAG. This finding is clinically relevant and suggests a mechanism by which ROS may cause proteinuria in clinical conditions associated with high oxidative stress.


Subject(s)
Endothelial Cells/metabolism , Glycocalyx/metabolism , Glycosaminoglycans/metabolism , Kidney Glomerulus/metabolism , Reactive Oxygen Species/metabolism , Albumins/metabolism , Cell Line , Cell Survival , Humans , Wheat Germ Agglutinins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...