Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
Add more filters










Publication year range
1.
Proc Natl Acad Sci U S A ; 119(35): e2116505119, 2022 08 30.
Article in English | MEDLINE | ID: mdl-35994650

ABSTRACT

Albuminuria is a hallmark of glomerular disease of various etiologies. It is not only a symptom of glomerular disease but also a cause leading to glomerulosclerosis, interstitial fibrosis, and eventually, a decline in kidney function. The molecular mechanism underlying albuminuria-induced kidney injury remains poorly defined. In our genetic model of nephrotic syndrome (NS), we have identified CHOP (C/EBP homologous protein)-TXNIP (thioredoxin-interacting protein) as critical molecular linkers between albuminuria-induced ER dysfunction and mitochondria dyshomeostasis. TXNIP is a ubiquitously expressed redox protein that binds to and inhibits antioxidant enzyme, cytosolic thioredoxin 1 (Trx1), and mitochondrial Trx2. However, very little is known about the regulation and function of TXNIP in NS. By utilizing Chop-/- and Txnip-/- mice as well as 68Ga-Galuminox, our molecular imaging probe for detection of mitochondrial reactive oxygen species (ROS) in vivo, we demonstrate that CHOP up-regulation induced by albuminuria drives TXNIP shuttling from nucleus to mitochondria, where it is required for the induction of mitochondrial ROS. The increased ROS accumulation in mitochondria oxidizes Trx2, thus liberating TXNIP to associate with mitochondrial nod-like receptor protein 3 (NLRP3) to activate inflammasome, as well as releasing mitochondrial apoptosis signal-regulating kinase 1 (ASK1) to induce mitochondria-dependent apoptosis. Importantly, inhibition of TXNIP translocation and mitochondrial ROS overproduction by CHOP deletion suppresses NLRP3 inflammasome activation and p-ASK1-dependent mitochondria apoptosis in NS. Thus, targeting TXNIP represents a promising therapeutic strategy for the treatment of NS.


Subject(s)
Albuminuria , Carrier Proteins , Kidney , Mitochondria , Nephrotic Syndrome , Thioredoxins , Transcription Factor CHOP , Albuminuria/complications , Albuminuria/genetics , Albuminuria/prevention & control , Animals , Apoptosis , Carrier Proteins/metabolism , Cell Nucleus/metabolism , Gene Deletion , Inflammasomes/metabolism , Kidney/metabolism , Kidney/pathology , MAP Kinase Kinase Kinase 5/metabolism , Mice , Mitochondria/metabolism , Mitochondrial Proteins/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Nephrotic Syndrome/complications , Nephrotic Syndrome/genetics , Nephrotic Syndrome/pathology , Nephrotic Syndrome/prevention & control , Reactive Oxygen Species/metabolism , Thioredoxins/metabolism , Transcription Factor CHOP/deficiency , Transcription Factor CHOP/genetics , Transcription Factor CHOP/metabolism
2.
FASEB J ; 34(2): 2087-2104, 2020 02.
Article in English | MEDLINE | ID: mdl-31907991

ABSTRACT

Proteinuria is associated with renal function decline and cardiovascular mortality. This association may be attributed in part to alterations of Klotho expression induced by albuminuria, yet the underlying mechanisms are unclear. The presence of albumin decreased Klotho expression in the POD-ATTAC mouse model of proteinuric kidney disease as well as in kidney epithelial cell lines. This downregulation was related to both decreased Klotho transcription and diminished protein half-life, whereas cleavage by ADAM proteases was not modified. The regulation was albumin specific since it was neither observed in the analbuminemic Col4α3-/- Alport mice nor induced by exposure of kidney epithelial cells to purified immunoglobulins. Albumin induced features of ER stress in renal tubular cells with ATF3/ATF4 activation. ATF3 and ATF4 induction downregulated Klotho through altered transcription mediated by their binding on the Klotho promoter. Inhibiting ER stress with 4-PBA decreased the effect of albumin on Klotho protein levels without altering mRNA levels, thus mainly abrogating the increased protein degradation. Taken together, albuminuria decreases Klotho expression through increased protein degradation and decreased transcription mediated by ER stress induction. This implies that modulating ER stress may improve proteinuria-induced alterations of Klotho expression, and hence renal and extrarenal complications associated with Klotho loss.


Subject(s)
Activating Transcription Factor 3/metabolism , Albuminuria/metabolism , Down-Regulation , Endoplasmic Reticulum Stress , Glucuronidase/biosynthesis , Kidney Tubules/metabolism , Transcription, Genetic , Activating Transcription Factor 3/genetics , Albuminuria/genetics , Albuminuria/pathology , Animals , Autoantigens/genetics , Autoantigens/metabolism , Collagen Type IV/genetics , Collagen Type IV/metabolism , Glucuronidase/genetics , Humans , Kidney Tubules/pathology , Klotho Proteins , Mice , Mice, Knockout
3.
Am J Physiol Renal Physiol ; 311(1): F120-30, 2016 07 01.
Article in English | MEDLINE | ID: mdl-27147675

ABSTRACT

Alport syndrome is a familial kidney disease caused by defects in the collagen type IV network of the glomerular basement membrane. Lack of collagen-α3α4α5(IV) changes the glomerular basement membrane morphologically and functionally, rendering it leaky to albumin and other plasma proteins. Filtered albumin has been suggested to be a cause of the glomerular and tubular injuries observed at advanced stages of Alport syndrome. To directly investigate the role that albumin plays in the progression of disease in Alport syndrome, we generated albumin knockout (Alb(-/-)) mice to use as a tool for removing albuminuria as a component of kidney disease. Mice lacking albumin were healthy and indistinguishable from control littermates, although they developed hypertriglyceridemia. Dyslipidemia was observed in Alb(+/-) mice, which displayed half the normal plasma albumin concentration. Alb mutant mice were bred to collagen-α3(IV) knockout (Col4a3(-/-)) mice, which are a model for human Alport syndrome. Lack of circulating and filtered albumin in Col4a3(-/-);Alb(-/-) mice resulted in dramatically improved kidney disease outcomes, as these mice lived 64% longer than did Col4a3(-/-);Alb(+/+) and Col4a3(-/-);Alb(+/-) mice, despite similar blood pressures and serum triglyceride levels. Further investigations showed that the absence of albumin correlated with reduced transforming growth factor-ß1 signaling as well as reduced tubulointerstitial, glomerular, and podocyte pathology. We conclude that filtered albumin is injurious to kidney cells in Alport syndrome and perhaps in other proteinuric kidney diseases, including diabetic nephropathy.


Subject(s)
Albumins/metabolism , Kidney Diseases/metabolism , Nephritis, Hereditary/metabolism , Albumins/deficiency , Albumins/genetics , Animals , Autoantigens/genetics , Autoantigens/metabolism , Blood Pressure , Collagen Type IV/biosynthesis , Collagen Type IV/genetics , Collagen Type IV/metabolism , Disease Progression , Kidney/pathology , Kidney Diseases/etiology , Kidney Diseases/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Nephritis, Hereditary/complications , Nephritis, Hereditary/pathology , Survival Analysis , Transforming Growth Factor beta1/biosynthesis , Triglycerides/blood
4.
J Clin Invest ; 125(6): 2307-16, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25915582

ABSTRACT

Podocytes are specialized epithelial cells in the kidney glomerulus that play important structural and functional roles in maintaining the filtration barrier. Nephrotic syndrome results from a breakdown of the kidney filtration barrier and is associated with proteinuria, hyperlipidemia, and edema. Additionally, podocytes undergo changes in morphology and internalize plasma proteins in response to this disorder. Here, we used fluid-phase tracers in murine models and determined that podocytes actively internalize fluid from the plasma and that the rate of internalization is increased when the filtration barrier is disrupted. In cultured podocytes, the presence of free fatty acids (FFAs) associated with serum albumin stimulated macropinocytosis through a pathway that involves FFA receptors, the Gß/Gγ complex, and RAC1. Moreover, mice with elevated levels of plasma FFAs as the result of a high-fat diet were more susceptible to Adriamycin-induced proteinuria than were animals on standard chow. Together, these results support a model in which podocytes sense the disruption of the filtration barrier via FFAs bound to albumin and respond by enhancing fluid-phase uptake. The response to FFAs may function in the development of nephrotic syndrome by amplifying the effects of proteinuria.


Subject(s)
Albumins/metabolism , Fatty Acids/metabolism , Nephrotic Syndrome/metabolism , Pinocytosis , Podocytes/metabolism , Proteinuria/metabolism , Animals , Antibiotics, Antineoplastic/adverse effects , Antibiotics, Antineoplastic/pharmacology , Cell Line, Transformed , Dietary Fats/adverse effects , Dietary Fats/pharmacology , Doxorubicin/adverse effects , Doxorubicin/pharmacology , Fatty Acids/genetics , Mice , Mice, Inbred BALB C , Mice, Knockout , Nephrotic Syndrome/chemically induced , Nephrotic Syndrome/genetics , Nephrotic Syndrome/pathology , Neuropeptides/genetics , Neuropeptides/metabolism , Podocytes/pathology , Proteinuria/chemically induced , Proteinuria/genetics , Proteinuria/pathology , rac1 GTP-Binding Protein/genetics , rac1 GTP-Binding Protein/metabolism
5.
Proc Natl Acad Sci U S A ; 108(37): 15348-53, 2011 Sep 13.
Article in English | MEDLINE | ID: mdl-21876163

ABSTRACT

Pierson syndrome is a congenital nephrotic syndrome with ocular and neurological defects caused by mutations in LAMB2, the gene encoding the basement membrane protein laminin ß2 (Lamß2). It is the kidney glomerular basement membrane (GBM) that is defective in Pierson syndrome, as Lamß2 is a component of laminin-521 (LM-521; α5ß2γ1), the major laminin in the mature GBM. In both Pierson syndrome and the Lamb2(-/-) mouse model for this disease, laminin ß1 (Lamß1), a structurally similar homolog of Lamß2, is marginally increased in the GBM, but it fails to fully compensate for the loss of Lamß2, leading to the filtration barrier defects and nephrotic syndrome. Here we generated several lines of Lamß1 transgenic mice and used them to show that podocyte-specific Lamß1 expression in Lamb2(-/-) mice abrogates the development of nephrotic syndrome, correlating with a greatly extended lifespan. In addition, the more Lamß1 was expressed, the less urinary albumin was excreted. Transgenic Lamß1 expression increased the level of Lamα5 in the GBM of rescued mice, consistent with the desired increased deposition of laminin-511 (α5ß1γ1) trimers. Ultrastructural analysis revealed occasional knob-like subepithelial GBM thickening but intact podocyte foot processes in aged rescued mice. These results suggest the possibility that up-regulation of LAMB1 in podocytes, should it become achievable, would likely lessen the severity of nephrotic syndrome in patients carrying LAMB2 mutations.


Subject(s)
Abnormalities, Multiple/pathology , Eye Abnormalities/pathology , Laminin/metabolism , Nephrotic Syndrome/prevention & control , Podocytes/metabolism , Pupil Disorders/pathology , Abnormalities, Multiple/physiopathology , Animals , Capillaries/metabolism , Capillaries/pathology , Capillaries/ultrastructure , Disease Models, Animal , Eye Abnormalities/physiopathology , Glomerular Basement Membrane/metabolism , Glomerular Basement Membrane/pathology , Glomerular Basement Membrane/ultrastructure , Glomerular Filtration Rate , Humans , Infant , Laminin/deficiency , Mice , Mice, Transgenic , Myasthenic Syndromes, Congenital , Nephrotic Syndrome/pathology , Nephrotic Syndrome/physiopathology , Podocytes/pathology , Pupil Disorders/physiopathology , Survival Analysis , Time Factors
6.
Am J Physiol Renal Physiol ; 300(3): F811-20, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21209007

ABSTRACT

Dystroglycan (DG or DAG1) is considered a critical link between the basement membrane and the cytoskeleton in multiple tissues. DG consists of two subunits, an extracellular α-subunit that binds laminin and other basement membrane components, and a transmembrane ß-subunit. DG-null mouse embryos die during early embryogenesis because DG is required for Reichert's membrane formation. DG also forms an integral part of the dystrophin-glycoprotein complex in muscle. Although no human DG mutations have been reported, multiple forms of muscular dystrophy have been linked to DG glycosylation defects, and targeted deletion of muscle DG causes muscular dystrophy in mice. Moreover, DG is widely distributed in endothelial and epithelial cells, including those in the kidney. There has therefore been significant interest in DG's role in the kidney, especially in podocytes. Previous reports suggested that DG's disturbance in podocytes might cause glomerular filtration barrier abnormalities. To fully understand DG's contribution to nephrogenesis and kidney function, we used a conditional DG allele and a variety of Cre mice to systematically delete DG from podocytes, ureteric bud, metanephric mesenchyme, and then from the whole kidney. Surprisingly, none of these conditional deletions resulted in significant morphological or functional abnormalities in the kidney. Furthermore, DG-deficient podocytes did not show increased susceptibility to injury, and DG-deficient kidneys did not show delayed recovery. Integrins are therefore likely the primary extracellular matrix receptors in renal epithelia.


Subject(s)
Acute Kidney Injury/metabolism , Dystroglycans/metabolism , Kidney/embryology , Kidney/physiology , Acute Kidney Injury/physiopathology , Animals , Dystroglycans/genetics , Epithelial Cells/metabolism , Integrases/genetics , Integrases/metabolism , Integrin alpha3/genetics , Integrin alpha3/metabolism , Kidney Glomerulus/metabolism , Mice , Mice, Inbred C57BL , Mice, Inbred CBA , Mice, Transgenic , Models, Animal , Podocytes/metabolism
7.
J Am Soc Nephrol ; 21(10): 1657-66, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20651158

ABSTRACT

Mutant forms of TRPC6 can activate NFAT-dependent transcription in vitro via calcium influx and activation of calcineurin. The same TRPC6 mutants can cause FSGS, but whether this involves an NFAT-dependent mechanism is unknown. Here, we generated mice that allow conditional induction of NFATc1. Mice with NFAT activation in nascent podocytes in utero developed proteinuria and glomerulosclerosis postnatally, resembling FSGS. NFAT activation in adult mice also caused progressive proteinuria and FSGS. Ultrastructural studies revealed podocyte foot process effacement and deposition of extracellular matrix. NFAT activation did not initially affect expression of podocin, synaptopodin, and nephrin but reduced their expression as glomerular injury progressed. In contrast, we observed upregulation of Wnt6 and Fzd9 in the mutant glomeruli before the onset of significant proteinuria, suggesting a potential role for Wnt signaling in the pathogenesis of NFAT-induced podocyte injury and FSGS. These results provide in vivo evidence for the involvement of NFAT signaling in podocytes, proteinuria, and glomerulosclerosis. Furthermore, this study suggests that NFAT activation may be a key intermediate step in the pathogenesis of mutant TRPC6-mediated FSGS and that suppression of NFAT activity may contribute to the antiproteinuric effects of calcineurin inhibitors.


Subject(s)
Glomerulosclerosis, Focal Segmental/metabolism , NFATC Transcription Factors/metabolism , Podocytes/metabolism , Animals , Apoptosis , Cell Proliferation , Disease Models, Animal , Doxycycline , Glomerulosclerosis, Focal Segmental/pathology , Kidney Glomerulus/ultrastructure , Mice , Proteinuria/metabolism , Signal Transduction , TRPC Cation Channels/metabolism , TRPC6 Cation Channel , Weaning
8.
Am J Physiol Renal Physiol ; 297(6): F1566-74, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19794110

ABSTRACT

The functional role of glomerular parietal epithelial cells (PECs) remains poorly understood. To test the hypothesis that PECs form an impermeable barrier to filtered protein through the formation of tight junctions (TJ), studies were performed in normal animals and in the anti-glomerular basement membrane (GBM) model of crescentic nephritis. Electron microscopy showed well-defined TJ between PECs in normal mice, which no longer could be identified when these cells became extensively damaged or detached from their underlying Bowman's basement membrane. The TJ proteins claudin-1, zonula occludens-1, and occludin stained positive in PECs; however, staining decreased in anti-GBM disease. To show that these events were associated with increased permeability across the PEC-Bowman's basement membrane barrier, control and diseased animals were injected intravenously with either Texas red-conjugated dextran (3 kDa) or ovalbumin (45 kDa) tracers. As expected, both tracers were readily filtered across the glomerular filtration barrier and taken up by proximal tubular cells. However, when the glomerular filtration barrier was injured in anti-GBM disease, tracers were taken up by podocytes and PECs. Moreover, tracers were also detected between PECs and the underlying Bowman's basement membrane, and in many instances were detected in the extraglomerular space. We propose that together with its underlying Bowman's basement membrane, the TJ of PECs serve as a second barrier to protein. When disturbed following PEC injury, the increase in permeability of this layer to filtered protein is a mechanism underlying periglomerular inflammation characteristic of anti-GBM disease.


Subject(s)
Anti-Glomerular Basement Membrane Disease/complications , Kidney Glomerulus/metabolism , Nephritis/etiology , Nephritis/metabolism , Aging/metabolism , Animals , Basement Membrane/metabolism , Basement Membrane/pathology , Bowman Capsule/metabolism , Bowman Capsule/pathology , Cell Line , Claudin-1 , Embryonic Development , Epithelial Cells/metabolism , Glomerular Filtration Rate , Humans , Kidney Glomerulus/embryology , Kidney Glomerulus/pathology , Membrane Proteins/metabolism , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Microscopy, Electron , Occludin , PAX8 Transcription Factor , Paired Box Transcription Factors/metabolism , Permeability , Phosphoproteins/metabolism , Podocytes/metabolism , Rats , Tight Junctions/metabolism , Tight Junctions/ultrastructure , Zonula Occludens-1 Protein
9.
Curr Opin Nephrol Hypertens ; 18(3): 226-32, 2009 May.
Article in English | MEDLINE | ID: mdl-19374010

ABSTRACT

PURPOSE OF REVIEW: The nephrology community lacks a unified view of protein sieving through the glomerular capillary wall. The glomerular capillary wall consists of three distinct but closely interacting layers: the fenestrated endothelium, with its glycocalyx; the podocytes, with their interdigitated foot processes and slit diaphragms; and the intervening glomerular basement membrane. Proteinuria is associated with abnormalities in any one layer, suggesting that each contributes to the glomerular filtration barrier (GFB). Proteinuria can also be induced in the context of a normal glomerular capillary wall. Here, we review some classic studies as well as some newer concepts and present competing hypotheses about the GFB. RECENT FINDINGS: Two almost forgotten concepts have recently emerged. One group has challenged the exquisite selectivity of the GFB to albumin and suggested that proteinuria is the result of abnormal tubular uptake. There has also been a reemphasis on diffusion through the glomerular basement membrane as the driving force behind macromolecular filtration. New evidence suggests that the endothelial glycocalyx is an important charge-selective barrier. SUMMARY: We suggest viewing the GFB as a dynamic rather than as a rigid barrier, requiring three healthy layers and a hemodynamic steady state. Multiple challenges to studying the endothelium, the tubular handling of albumin, and the role of hemodynamic forces will require new tools, new hypotheses, and open minds.


Subject(s)
Kidney Glomerulus/physiology , Animals , Endothelium/physiology , Filtration , Glomerular Basement Membrane/metabolism , Humans , Kidney Tubules/metabolism , Podocytes/metabolism , Proteins/metabolism
11.
Genesis ; 47(1): 1-6, 2009 Jan.
Article in English | MEDLINE | ID: mdl-18942111

ABSTRACT

Pax3-Cre (P3Pro-Cre) transgenic mice have been used for conditional gene deletion and/or lineage tracing in derivatives of neural crest, neural tube, metanephric mesenchyme, and ureteric mesenchyme. However, the extent of its expression in skeletal muscle has not been reported. We investigated the expression of P3Pro-Cre in the skeletal muscle lineage using the R26R reporter and found an unexpected rostrocaudal gradient of expression. By X-gal staining, head, neck, forelimb, diaphragm, and most of the chest wall muscles did not show evidence of Cre expression, whereas all muscle groups posterior of the diaphragm stained blue. Intercostal muscles exhibited a rostrocaudal gradient of staining. The consistency of this expression pattern was demonstrated by using P3Pro-Cre to mutate a conditional dystroglycan allele. The result was loss of dystroglycan from caudal muscles, which exhibited the histological signs of muscle fiber injury and regeneration characteristic of muscular dystrophy. The lack of dystroglycan in regenerating myofibers suggests that the P3Pro-Cre transgene is active in satellite cells and/or in their precursors. In contrast, rostral muscles, including feeding and breathing muscles, maintained dystroglycan expression and were spared from disease. Accordingly, the mutants were viable for over a year. Its unique gradient of activity makes the P3Pro-Cre transgene a previously unappreciated yet powerful tool for manipulating gene expression in skeletal muscle and its precursors.


Subject(s)
Cell Lineage , Gene Expression Regulation, Developmental , Integrases/genetics , Muscle, Skeletal/cytology , Muscle, Skeletal/metabolism , Paired Box Transcription Factors/genetics , Transgenes/genetics , Animals , Integrases/metabolism , Mice , Mice, Transgenic , Muscle, Skeletal/growth & development , PAX3 Transcription Factor , Paired Box Transcription Factors/metabolism
12.
J Cell Biol ; 182(6): 1201-15, 2008 Sep 22.
Article in English | MEDLINE | ID: mdl-18794334

ABSTRACT

A prominent feature of synaptic maturation at the neuromuscular junction (NMJ) is the topological transformation of the acetylcholine receptor (AChR)-rich postsynaptic membrane from an ovoid plaque into a complex array of branches. We show here that laminins play an autocrine role in promoting this transformation. Laminins containing the alpha4, alpha5, and beta2 subunits are synthesized by muscle fibers and concentrated in the small portion of the basal lamina that passes through the synaptic cleft at the NMJ. Topological maturation of AChR clusters was delayed in targeted mutant mice lacking laminin alpha5 and arrested in mutants lacking both alpha4 and alpha5. Analysis of chimeric laminins in vivo and of mutant myotubes cultured aneurally demonstrated that the laminins act directly on muscle cells to promote postsynaptic maturation. Immunohistochemical studies in vivo and in vitro along with analysis of targeted mutants provide evidence that laminin-dependent aggregation of dystroglycan in the postsynaptic membrane is a key step in synaptic maturation. Another synaptically concentrated laminin receptor, Bcam, is dispensable. Together with previous studies implicating laminins as organizers of presynaptic differentiation, these results show that laminins coordinate post- with presynaptic maturation.


Subject(s)
Autocrine Communication/physiology , Laminin/metabolism , Neuromuscular Junction/physiology , Acetylcholinesterase/metabolism , Animals , Cells, Cultured , Dystroglycans/genetics , Dystroglycans/metabolism , Humans , Laminin/genetics , Mice , Mice, Knockout , Mice, Transgenic , Muscle Fibers, Skeletal/cytology , Muscle Fibers, Skeletal/metabolism , Muscle, Skeletal/cytology , Muscle, Skeletal/metabolism , Receptors, Cholinergic/genetics , Receptors, Cholinergic/metabolism , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Transgenes
13.
J Am Soc Nephrol ; 19(11): 2150-8, 2008 Nov.
Article in English | MEDLINE | ID: mdl-18776121

ABSTRACT

MicroRNAs (miRNAs) regulate gene expression by binding the 3' untranslated region of mRNAs. To define their role in glomerular function, miRNA biogenesis was disrupted in mouse podocytes using a conditional Dicer allele. Mutant mice developed proteinuria by 3 wk after birth and progressed rapidly to end-stage kidney disease. Podocyte pathology included effacement, vacuolization, and hypertrophy with crescent formation. Despite normal expression of WT1, podocytes underwent dedifferentiation, exemplified by cytoskeletal disruption with early transcriptional downregulation of synaptopodin. These abnormalities differed from Cd2ap(-/-) mice, indicating they were not a general consequence of glomerular disease. Glomerular labeling of ezrin, moesin, and gelsolin was altered at 3 wk, but expression of nestin and alpha-actinin was unchanged. Abnormal cell proliferation or apoptosis was not responsible for the glomerular injury. Mutant podocytes were incapable of synthesizing mature miRNA, as revealed by their loss of miR-30a. In contrast, expression of glomerular endothelial and mesangial cell miRNAs (miR-126 and miR-145, respectively) was unchanged. These findings demonstrate a critical role for miRNA in glomerular function and suggest a pathway that may participate in the pathogenesis of kidney diseases of podocyte origin. The unique architecture of podocytes may make them especially susceptible to cytoskeletal alterations initiated by aberrant miRNA dynamics.


Subject(s)
DEAD-box RNA Helicases/deficiency , Endoribonucleases/deficiency , Kidney Diseases/etiology , Podocytes/enzymology , Animals , Apoptosis , Cell Differentiation , Cell Proliferation , Cytoskeleton/metabolism , DEAD-box RNA Helicases/genetics , DEAD-box RNA Helicases/metabolism , Endoribonucleases/genetics , Endoribonucleases/metabolism , Kidney Diseases/enzymology , Kidney Diseases/genetics , Kidney Diseases/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , MicroRNAs/biosynthesis , MicroRNAs/genetics , Podocytes/pathology , RNA Processing, Post-Transcriptional , Ribonuclease III
14.
Dev Biol ; 316(2): 288-301, 2008 Apr 15.
Article in English | MEDLINE | ID: mdl-18328474

ABSTRACT

Integrins are transmembrane heteromeric receptors that mediate interactions between cells and extracellular matrix (ECM). beta1, the most abundantly expressed integrin subunit, binds at least 12 alpha subunits. beta1 containing integrins are highly expressed in the glomerulus of the kidney; however their role in glomerular morphogenesis and maintenance of glomerular filtration barrier integrity is poorly understood. To study these questions we selectively deleted beta1 integrin in the podocyte by crossing beta1(flox/flox) mice with podocyte specific podocin-cre mice (pod-Cre), which express cre at the time of glomerular capillary formation. We demonstrate that podocyte abnormalities are visualized during glomerulogenesis of the pod-Cre;beta1(flox/flox) mice and proteinuria is present at birth, despite a grossly normal glomerular basement membrane. Following the advent of glomerular filtration there is progressive podocyte loss and the mice develop capillary loop and mesangium degeneration with little evidence of glomerulosclerosis. By 3 weeks of age the mice develop severe end stage renal failure characterized by both tubulointerstitial and glomerular pathology. Thus, expression of beta1 containing integrins by the podocyte is critical for maintaining the structural integrity of the glomerulus.


Subject(s)
Gene Expression Regulation , Integrin beta1/genetics , Kidney Glomerulus/growth & development , Podocytes/physiology , Aging , Animals , Animals, Newborn , DNA Primers , In Situ Hybridization , Kidney Glomerulus/anatomy & histology , Kidney Glomerulus/physiology , Mice , Mice, Knockout , Mice, Transgenic , Polymerase Chain Reaction , Proteinuria/genetics
15.
Am J Pathol ; 171(1): 139-52, 2007 Jul.
Article in English | MEDLINE | ID: mdl-17591961

ABSTRACT

Glomerular charge selectivity has been attributed to anionic heparan sulfate proteoglycans (HSPGs) in the glomerular basement membrane (GBM). Agrin is the predominant GBM-HSPG, but evidence that it contributes to the charge barrier is lacking, because newborn agrin-deficient mice die from neuromuscular defects. To study agrin in adult kidney, a new conditional allele was used to generate podocyte-specific knockouts. Mutants were viable and displayed no renal histopathology up to 9 months of age. Perlecan, a HSPG normally confined to the mesangium in mature glomeruli, did not appear in the mutant GBM, which lacked heparan sulfate. Moreover, GBM agrin was found to be derived primarily from podocytes. Polyethyleneimine labeling of fetal kidneys revealed anionic sites along both laminae rarae of the GBM that became most prominent along the subepithelial aspect at maturity; labeling was greatly reduced along the subepithelial aspect in agrin-deficient and conditional knockout mice. Despite this severe charge disruption, the glomerular filtration barrier was not compromised, even when challenged with bovine serum albumin overload. We conclude that agrin is not required for establishment or maintenance of GBM architecture. Although agrin contributes significantly to the anionic charge to the GBM, both it and its charge are not needed for glomerular permselectivity. This calls into question whether charge selectivity is a feature of the GBM.


Subject(s)
Agrin/physiology , Cell Membrane Permeability , Glomerular Basement Membrane/physiology , Podocytes/metabolism , Agrin/genetics , Animals , Anion Transport Proteins/metabolism , Electrophysiology , Kidney/anatomy & histology , Kidney/diagnostic imaging , Kidney Glomerulus/anatomy & histology , Kidney Glomerulus/ultrastructure , Mice , Mice, Knockout , Mutation , Ultrasonography
16.
J Clin Invest ; 116(8): 2272-9, 2006 Aug.
Article in English | MEDLINE | ID: mdl-16886065

ABSTRACT

Primary defects in either podocytes or the glomerular basement membrane (GBM) cause proteinuria, a fact that complicates defining the barrier to albumin. Laminin beta2 (LAMB2) is a GBM component required for proper functioning of the glomerular filtration barrier. To investigate the GBM's role in glomerular filtration, we characterized GBM and overlying podocyte architecture in relation to development and progression of proteinuria in Lamb2-/- mice, which model Pierson syndrome, a rare congenital nephrotic syndrome. We found ectopic deposition of several laminins and mislocalization of anionic sites in the GBM, which together suggest that the Lamb2-/- GBM is severely disorganized, although it is ultrastructurally intact. Importantly, albuminuria was detectable shortly after birth and preceded podocyte foot process effacement and loss of slit diaphragms by at least 7 days. Expression and localization of slit diaphragm and foot process-associated proteins appeared normal at early stages. GBM permeability to the electron-dense tracer ferritin was dramatically elevated in Lamb2-/- mice, even before widespread foot process effacement. Increased ferritin permeability was not observed in nephrotic CD2-associated protein-null (Cd2ap-/-) mice, which have a primary podocyte defect. Together these data show that the GBM serves as a barrier to protein in vivo and that the glomerular slit diaphragm alone is not sufficient to prevent the passage of albumin into the urinary space.


Subject(s)
Albuminuria/physiopathology , Basement Membrane/physiology , Kidney Glomerulus/physiology , Laminin/deficiency , Podocytes/pathology , Proteinuria/genetics , Animals , Basement Membrane/ultrastructure , Disease Models, Animal , Ferritins/metabolism , Kidney Glomerulus/ultrastructure , Laminin/genetics , Laminin/physiology , Mice , Mice, Knockout , Microscopy, Electron, Scanning , Nephrotic Syndrome/genetics , Nephrotic Syndrome/pathology
17.
J Biol Chem ; 281(28): 19688-99, 2006 Jul 14.
Article in English | MEDLINE | ID: mdl-16690620

ABSTRACT

Alpha(v)beta8 integrin expression is restricted primarily to kidney, brain, and placenta. Targeted alpha(v) or beta8 deletion is embryonic lethal due to defective placenta and brain angiogenesis, precluding investigation of kidney alpha(v)beta8 function. We find that kidney beta8 is localized to glomerular mesangial cells, and expression is decreased in mouse models of glomerulosclerosis, suggesting that beta8 regulates normal mesangial cell differentiation. To interrogate beta8 signaling pathways, yeast two-hybrid and co-precipitation studies demonstrated beta8 interaction with Rho guanine nucleotide dissociation inhibitor-1 (GDI). Selective beta8 stimulation enhanced beta8-GDI interaction as well as Rac1 (but not RhoA) activation and lamellipodia formation. Mesangial cells from itgb8-/- mice backcrossed to a genetic background that permitted survival, or gdi-/- mice, which develop glomerulosclerosis, demonstrated RhoA (but not Rac1) activity and alpha-smooth muscle actin assembly, which characterizes mesangial cell myofibroblast transformation in renal disease. To determine whether Rac1 directly modulates RhoA-associated myofibroblast differentiation, mesangial cells were transduced with inhibitory Rac peptide fused to human immunodeficiency virus-Tat, resulting in enhanced alpha-smooth muscle actin organization. We conclude that the beta8 cytosolic tail in mesangial cells organizes a signaling complex that culminates in Rac1 activation to mediate wild-type differentiation, whereas decreased beta8 activation shifts mesangial cells toward a RhoA-dependent myofibroblast phenotype.


Subject(s)
Fibroblasts/cytology , Guanine Nucleotide Dissociation Inhibitors/metabolism , Integrin beta Chains/metabolism , rac1 GTP-Binding Protein/metabolism , Animals , Cell Differentiation , Fibroblasts/metabolism , Glomerulosclerosis, Focal Segmental/metabolism , Integrin alphaV/metabolism , Integrin beta Chains/chemistry , Kidney/metabolism , Mice , Mice, Transgenic , Phenotype , Protein Binding , Signal Transduction
18.
J Am Soc Nephrol ; 17(5): 1334-44, 2006 May.
Article in English | MEDLINE | ID: mdl-16611717

ABSTRACT

Alterations in glomerular podocyte cell-cell and cell-matrix contacts are key events in progressive glomerular failure. Integrin-linked kinase (ILK) has been implicated in podocyte cell-matrix interaction and is induced in proteinuria. For evaluation of ILK function in vivo, mice with a Cre-mediated podocyte-specific ILK inactivation were generated. These mice seemed normal at birth but developed progressive focal segmental glomerulosclerosis and died in terminal renal failure. The first ultrastructural lesions that are seen at onset of albuminuria are glomerular basement membrane (GBM) alterations with a significant increase in true harmonic mean GBM thickness. Podocyte foot process effacement and loss of slit diaphragm followed with progression to unselective proteinuria. No significant reduction of slit membrane molecules (podocin and nephrin), key GBM components (fibronectin, laminins, and collagen IV isoforms), or podocyte integrins could be observed at onset of proteinuria. However, alpha3-integrins were relocalized into a granular pattern along the GBM, consistent with altered integrin-mediated matrix assembly in ILK-deficient podocytes. As the increased GBM thickness precedes structural podocyte lesions and key components of the GBM were expressed at comparable levels to controls, these data suggest an essential role of ILK for the close interconnection of GBM structure and podocyte function.


Subject(s)
Cell Membrane/metabolism , Cell Membrane/pathology , Glomerulosclerosis, Focal Segmental/metabolism , Glomerulosclerosis, Focal Segmental/pathology , Podocytes/metabolism , Protein Serine-Threonine Kinases/metabolism , Animals , Cells, Cultured , Mice , Mice, Knockout , Mice, Transgenic , Protein Serine-Threonine Kinases/genetics , Survival Analysis , Survival Rate
19.
Development ; 133(5): 967-75, 2006 Mar.
Article in English | MEDLINE | ID: mdl-16452099

ABSTRACT

Pierson syndrome is a recently defined disease usually lethal within the first postnatal months and caused by mutations in the gene encoding laminin beta2 (LAMB2). The hallmarks of Pierson syndrome are congenital nephrotic syndrome accompanied by ocular abnormalities, including microcoria (small pupils), with muscular and neurological developmental defects also present. Lamb2(-/-) mice are a model for Pierson syndrome; they exhibit defects in the kidney glomerular barrier, in the development and organization of the neuromuscular junction, and in the retina. Lamb2(-/-) mice fail to thrive and die very small at 3 weeks of age, but to what extent the kidney and neuromuscular defects each contribute to this severe phenotype has been obscure, though highly relevant to understanding Pierson syndrome. To investigate this, we generated transgenic mouse lines expressing rat laminin beta2 either in muscle or in glomerular epithelial cells (podocytes) and crossed them onto the Lamb2(-/-) background. Rat beta2 was confined in skeletal muscle to synapses and myotendinous junctions, and in kidney to the glomerular basement membrane. In transgenic Lamb2(-/-) mice, beta2 deposition in only glomeruli prevented proteinuria but did not ameliorate the severe phenotype. By contrast, beta2 expression in only muscle restored synaptic architecture and led to greatly improved health, but the mice died from kidney disease at 1 month. Rescue of both glomeruli and synapses was associated with normal weight gain, fertility and lifespan. We conclude that muscle defects in Lamb2(-/-) mice are responsible for the severe failure to thrive phenotype, and that renal replacement therapy alone will be an inadequate treatment for Pierson syndrome.


Subject(s)
Glomerular Basement Membrane/abnormalities , Kidney Diseases/genetics , Laminin/genetics , Muscle, Skeletal/abnormalities , Muscular Diseases/genetics , Animals , Glomerular Basement Membrane/pathology , Kidney Diseases/pathology , Laminin/analysis , Laminin/metabolism , Mice , Mice, Knockout , Mice, Transgenic , Muscle, Skeletal/pathology , Muscular Diseases/pathology , Mutation , Neuromuscular Junction/chemistry , Neuromuscular Junction/metabolism , Neuromuscular Junction/ultrastructure , Podocytes/chemistry , Podocytes/metabolism , Rats , Synapses/chemistry , Synapses/metabolism , Syndrome
20.
Kidney Int ; 66(3): 1029-35, 2004 Sep.
Article in English | MEDLINE | ID: mdl-15327396

ABSTRACT

BACKGROUND: ROP-Os/+ mice are born with oligosyndactyly and oligonephronia and develop renal dysfunction, which includes renal tubular epithelial cell (RTC) Fas-dependent apoptosis and tubular atrophy. MRL/lpr mice harbor a Fas-inactivating mutation and develop glomerulonephritis, whereas mice expressing lpr on a C3H background demonstrate no renal phenotype. We hypothesized that crossing ROP-Os/+ with CH3-lpr/lpr mice would rescue the Os/+ renal phenotype by reducing Fas-dependent RTC apoptosis. METHODS: ROP-Os/+ mice were intercrossed with C3H-lpr/lpr mice and F(2) generation animals were phenotyped by kidney weight, serum creatinine, and albuminuria. Kidney sections were scored for histopathology and apoptosis. Univariate and multivariate analyses were used to examine additive effects of Os and lpr on renal phenotype. RESULTS: By 16 weeks, F(2)Os/+ lpr/lpr mice developed significantly more albuminuria, glomerulosclerosis, and interstitial inflammation compared to Os/++/+ mice. Glomerular cell apoptosis was increased in Os/+ lpr/lpr compared to Os/++/+ mice, with no significant difference in RTC apoptosis. A statistically significant Os-lpr effect on renal phenotype was demonstrated by multivariate analysis, which exceeded the combined independent effects if Os and lpr, indicating a biologic interaction exists between Os and lpr. CONCLUSION: Os/+ mice with a superimposed lpr mutation displayed a more severe renal phenotype, rather than phenotype rescue, suggesting that Fas pathway activation is necessary to delete cells resulting from Os-dependent injury. We further propose that an Os-lpr gene interaction and/or mixed ROP-C3H genetic background regulated the renal phenotype, consistent with the concept that chronic renal disease pathogenesis reflects effects of multiple nephropathy susceptibility alleles.


Subject(s)
Lupus Nephritis/genetics , Lupus Nephritis/physiopathology , Syndactyly/genetics , Animals , Apoptosis , Body Weight , Epithelial Cells/pathology , Epithelial Cells/physiology , Female , Genotype , Kidney Glomerulus/pathology , Kidney Glomerulus/physiopathology , Kidney Tubules/pathology , Kidney Tubules/physiopathology , Lupus Nephritis/complications , Male , Mice , Mice, Inbred C3H , Mice, Inbred MRL lpr , Organ Size , Phenotype , Syndactyly/complications , Toes/abnormalities , fas Receptor/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...