Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
Add more filters










Publication year range
1.
Sci Rep ; 11(1): 19605, 2021 10 01.
Article in English | MEDLINE | ID: mdl-34599260

ABSTRACT

Increasing evidence suggests that single in kidney states (e.g., kidney transplantation and living donation) progressive glomerulosclerosis limits kidney lifespan. Modeling shows that post-nephrectomy compensatory glomerular volume (GV) increase drives podocyte depletion and hypertrophic stress resulting in proteinuria and glomerulosclerosis, implying that GV increase could serve as a therapeutic target to prevent progression. In this report we examine how Angiotensin Converting Enzyme inhibition (ACEi), started before uninephrectomy can reduce compensatory GV increase in wild-type Fischer344 rats. An unbiased computer-assisted method was used for morphometric analysis. Urine Insulin-like growth factor-1 (IGF-1), the major diver of body and kidney growth, was used as a readout. In long-term (40-week) studies of uni-nephrectomized versus sham-nephrectomized rats a 2.2-fold increase in GV was associated with reduced podocyte density, increased proteinuria and glomerulosclerosis. Compensatory GV increase was largely prevented by ACEi started a week before but not after uni-nephrectomy with no measurable impact on long-term eGFR. Similarly, in short-term (14-day) studies, ACEi started a week before uni-nephrectomy reduced both GV increase and urine IGF-1 excretion. Thus, timing of ACEi in relation to uni-nephrectomy had significant impact on post-nephrectomy "compensatory" glomerular growth and outcomes that could potentially be used to improve kidney transplantation and live kidney donation outcomes.


Subject(s)
Angiotensin-Converting Enzyme Inhibitors/pharmacology , Glomerulosclerosis, Focal Segmental/prevention & control , Hypertrophy/prevention & control , Nephrectomy/adverse effects , Solitary Kidney/pathology , Angiotensin-Converting Enzyme Inhibitors/administration & dosage , Animals , Hypertrophy/drug therapy , Insulin-Like Growth Factor I/urine , Kidney Glomerulus/pathology , Male , Podocytes , Proteinuria , Rats, Inbred F344
2.
JCI Insight ; 1(7)2016.
Article in English | MEDLINE | ID: mdl-27280173

ABSTRACT

BACKGROUND: Kidney function decreases with age. A potential mechanistic explanation for kidney and allograft half-life has evolved through the realization that linear reduction in glomerular podocyte density could drive progressive glomerulosclerosis to impact both native kidney and allograft half-lives. METHODS: Predictions from podometrics (quantitation of podocyte parameters) were tested using independent pathologic, functional, and outcome data for native kidneys and allografts derived from published reports and large registries. RESULTS: With age, native kidneys exponentially develop glomerulosclerosis, reduced renal function, and end-stage kidney disease, projecting a finite average kidney life span. The slope of allograft failure rate versus age parallels that of reduction in podocyte density versus age. Quantitative modeling projects allograft half-life at any donor age, and rate of podocyte detachment parallels the observed allograft loss rate. CONCLUSION: Native kidneys are designed to have a limited average life span of about 100-140 years. Allografts undergo an accelerated aging-like process that accounts for their unexpectedly short half-life (about 15 years), the observation that older donor age is associated with shorter allograft half-life, and the fact that long-term allograft survival has not substantially improved. Podometrics provides potential readouts for these processes, thereby offering new approaches for monitoring and intervention. FUNDING: National Institutes of Health.

3.
J Am Soc Nephrol ; 26(12): 3162-78, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26038526

ABSTRACT

Kidney aging is associated with an increasing proportion of globally scarred glomeruli, decreasing renal function, and exponentially increasing ESRD prevalence. In model systems, podocyte depletion causes glomerulosclerosis, suggesting age-associated glomerulosclerosis could be caused by a similar mechanism. We measured podocyte number, size, density, and glomerular volume in 89 normal kidney samples from living and deceased kidney donors and normal poles of nephrectomies. Podocyte nuclear density decreased with age due to a combination of decreased podocyte number per glomerulus and increased glomerular volume. Compensatory podocyte cell hypertrophy prevented a change in the proportion of tuft volume occupied by podocytes. Young kidneys had high podocyte reserve (podocyte density >300 per 10(6) µm(3)), but by 70-80 years of age, average podocyte nuclear density decreased to, <100 per 10(6) µm(3), with corresponding podocyte hypertrophy. In older age podocyte detachment rate (urine podocin mRNA-to-creatinine ratio) was higher than at younger ages and podocytes were stressed (increased urine podocin-to-nephrin mRNA ratio). Moreover, in older kidneys, proteinaceous material accumulated in the Bowman space of glomeruli with low podocyte density. In a subset of these glomeruli, mass podocyte detachment events occurred in association with podocytes becoming binucleate (mitotic podocyte catastrophe) and subsequent wrinkling of glomerular capillaries, tuft collapse, and periglomerular fibrosis. In kidneys of young patients with underlying glomerular diseases similar pathologic events were identified in association with focal global glomerulosclerosis. Podocyte density reduction with age may therefore directly lead to focal global glomerulosclerosis, and all progressive glomerular diseases can be considered superimposed accelerators of this underlying process.


Subject(s)
Aging/pathology , Glomerulosclerosis, Focal Segmental/pathology , Podocytes/pathology , Adolescent , Adult , Age Factors , Aged , Aged, 80 and over , Cell Count , Cell Nucleus/pathology , Child , Child, Preschool , Humans , Hypertrophy/pathology , Middle Aged , Organ Size , Young Adult
4.
Nephrol Dial Transplant ; 27(11): 4079-87, 2012 Nov.
Article in English | MEDLINE | ID: mdl-22863839

ABSTRACT

BACKGROUND: Proteinuria and/or albuminuria are widely used for noninvasive assessment of kidney diseases. However, proteinuria is a nonspecific marker of diverse forms of kidney injury, physiologic processes and filtration of small proteins of monoclonal and other pathologic processes. The opportunity to develop new glomerular disease biomarkers follows the realization that the degree of podocyte depletion determines the degree of glomerulosclerosis, and if persistent, determines the progression to end-stage kidney disease (ESKD). Podocyte cell lineage-specific mRNAs can be recovered in urine pellets of model systems and in humans. In model systems, progressive glomerular disease is associated with decreased nephrin mRNA steady-state levels compared with podocin mRNA. Thus, the urine podocin:nephrin mRNA ratio (PNR) could serve as a useful progression biomarker. The use of podocyte-specific transcript ratios also circumvents many problems inherent to urine assays. METHODS: To test this hypothesis, the human diphtheria toxin receptor (hDTR) rat model of progression was used to evaluate potentially useful urine mRNA biomarkers. We compared histologic progression parameters (glomerulosclerosis score, interstitial fibrosis score and percent of podocyte depletion) with clinical biomarkers [serum creatinine, systolic blood pressure (BP), 24-h urine volume, 24-h urine protein excretion and the urine protein:creatinine ratio(PCR)] and with the novel urine mRNA biomarkers. RESULTS: The PNR correlated with histologic outcome as well or better than routine clinical biomarkers and other urine mRNA biomarkers in the model system with high specificity and sensitivity, and a low coefficient of assay variation. CONCLUSIONS: We concluded that the PNR, used in combination with proteinuria, will be worth testing for its clinical diagnostic and decision-making utility.


Subject(s)
Intracellular Signaling Peptides and Proteins/metabolism , Kidney Diseases/metabolism , Kidney Glomerulus/metabolism , Membrane Proteins/metabolism , Podocytes/metabolism , Proteinuria/metabolism , RNA, Messenger/urine , Animals , Biomarkers/metabolism , Biomarkers/urine , Humans , Intracellular Signaling Peptides and Proteins/urine , Kidney Diseases/pathology , Kidney Glomerulus/pathology , Membrane Proteins/urine , Podocytes/pathology , Proteinuria/pathology , Rats , Rats, Inbred F344 , Real-Time Polymerase Chain Reaction
5.
J Gerontol A Biol Sci Med Sci ; 67(12): 1358-64, 2012 Dec.
Article in English | MEDLINE | ID: mdl-22843670

ABSTRACT

Kidney function declines with age in the majority of the population. Although very few older people progress to end stage, the consequences of doing so are burdensome for the patient and very expensive for the society. Although some of the observed decline is likely due to changes in the vasculature, much is associated with the development of age-associated glomerulosclerosis. This article will review the well-established structural and functional changes in the glomerulus with age. The role of calorie restriction in modifying age-related pathology will be discussed. The importance of the podocyte as a critical cell in the aging process is considered using animal models and human biopsy material. Newer data on changes in gene expression driven by nuclear factor kappa beta (NFkB) and possible changes in biology in the glomerulus are discussed. The relationship between pathways involved in aging and the decline in kidney function is reviewed. There is speculation on the significance of these changes in relation to normal and pathological aging.


Subject(s)
Aging/physiology , Kidney Glomerulus/physiology , Animals , Caloric Restriction , Ceruloplasmin/metabolism , Disease Progression , Gene Expression Regulation/physiology , Glomerular Basement Membrane/physiology , Glomerular Filtration Rate/physiology , Humans , Insulin-Like Growth Factor I/physiology , Kidney Failure, Chronic/physiopathology , Kidney Glomerulus/anatomy & histology , Podocytes/physiology
6.
J Am Soc Nephrol ; 23(8): 1351-63, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22773827

ABSTRACT

Podocyte depletion leads to glomerulosclerosis, but whether an impaired capacity of podocytes to respond to hypertrophic stress also causes glomerulosclerosis is unknown. We generated transgenic Fischer 344 rats that express a dominant negative AA-4E-BP1 transgene driven by the podocin promoter; a member of the mammalian target of rapamycin complex 1 (mTORC1) pathway, 4E-BP1 modulates cap-dependent translation, which is a key determinant of a cell's hypertrophic response to nutrients and growth factors. AA-4E-BP1 rat podocytes expressed the transgene and had normal kidney histology and protein excretion at 100 g of body weight but developed ESRD by 12 months. Proteinuria and glomerulosclerosis were linearly related to both increasing body weight and transgene dose. Uni-nephrectomy reduced the body weight at which proteinuria first developed by 40%-50%. The initial histologic manifestation of disease was the appearance of bare areas of glomerular basement membrane from the pulling apart of podocyte foot processes, followed by adhesions to the Bowman capsule. Morphometric analysis confirmed the mismatch between glomerular tuft volume and total podocyte volume (number × size) per tuft in relation to weight gain and nephrectomy. Proteinuria and glomerulosclerosis did not develop if dietary calorie restriction prevented weight gain and glomerular enlargement. In summary, failure of podocytes to match glomerular tuft growth in response to growth signaling through the mTORC1 pathway can trigger proteinuria, glomerulosclerosis, and progression to ESRD. Reducing body weight and glomerular growth may be useful adjunctive therapies to slow or prevent progression to ESRD.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Glomerulosclerosis, Focal Segmental/etiology , Kidney Glomerulus/growth & development , Phosphoproteins/metabolism , Podocytes/physiology , Weight Gain , Adaptor Proteins, Signal Transducing/genetics , Animals , Caloric Restriction , Cell Cycle Proteins , Glomerulosclerosis, Focal Segmental/prevention & control , Heterozygote , Homozygote , Humans , Hypertrophy , Male , Nephrectomy , Phosphoproteins/genetics , Podocytes/pathology , Proteinuria/etiology , Proteinuria/prevention & control , Rats , Rats, Inbred F344 , Rats, Transgenic
7.
Kidney Int ; 81(1): 40-55, 2012 Jan.
Article in English | MEDLINE | ID: mdl-21937979

ABSTRACT

Podocyte depletion is a major mechanism driving glomerulosclerosis. Progression is the process by which progressive glomerulosclerosis leads to end stage kidney disease (ESKD). In order to determine mechanisms contributing to persistent podocyte loss, we used a human diphtheria toxin transgenic rat model. After initial diphtheria toxin-induced podocyte injury (over 30% loss in 4 weeks), glomeruli became destabilized, resulting in continued autonomous podocyte loss causing global podocyte depletion (ESKD) by 13 weeks. This was monitored by urine mRNA analysis and by quantitating podocytes in glomeruli. Similar patterns of podocyte depletion were found in the puromycin aminonucleoside and 5/6 nephrectomy rat models of progressive end-stage disease. Angiotensin II blockade (combined enalapril and losartan) restabilized the glomeruli, and prevented continuous podocyte loss and progression to ESKD. Discontinuing angiotensin II blockade resulted in recurrent glomerular destabilization, podocyte loss, and progression to ESKD. Reduction in blood pressure alone did not reduce proteinuria or prevent podocyte loss from destabilized glomeruli. The protective effect of angiotensin II blockade was entirely accounted for by reduced podocyte loss. Thus, an initiating event resulting in a critical degree of podocyte depletion can destabilize glomeruli and initiate a superimposed angiotensin II-dependent podocyte loss process that accelerates progression resulting in eventual global podocyte depletion and ESKD. These events can be monitored noninvasively in real-time through urine mRNA assays.


Subject(s)
Angiotensin II/metabolism , Kidney Failure, Chronic/metabolism , Kidney Failure, Chronic/pathology , Kidney Glomerulus/metabolism , Kidney Glomerulus/pathology , Podocytes/metabolism , Podocytes/pathology , Angiotensin II/antagonists & inhibitors , Animals , Antihypertensive Agents/pharmacology , Disease Models, Animal , Humans , Intracellular Signaling Peptides and Proteins/genetics , Kidney Failure, Chronic/drug therapy , Kidney Failure, Chronic/genetics , Male , Membrane Proteins/genetics , Podocytes/drug effects , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats , Rats, Inbred F344 , Rats, Sprague-Dawley , Rats, Transgenic
8.
J Am Soc Nephrol ; 21(4): 587-97, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20150534

ABSTRACT

The peak prevalence of ESRD from glomerulosclerosis occurs at 70 to 79 years. To understand why old glomeruli are prone to failure, we analyzed the Fischer 344 rat model of aging under ad libitum-fed (rapid aging) and calorie-restricted (slowed aging) conditions. All glomerular cells contained genes whose expression changed "linearly" during adult life from 2 to 24 months: mesangial cells (e.g., MMP9), endothelial cells (e.g., ICAM and VCAM), parietal epithelial cells (e.g., ceruloplasmin), and podocytes (e.g., nephrin and prepronociceptin). Patterns of aging glomerular gene expression closely resembled atherosclerosis, including activation of endothelial cells, epithelial cells, and macrophages, as well as proinflammatory pathways related to cell adhesion, chemotaxis, blood coagulation, oxidoreductases, matrix metalloproteinases, and TGF-beta activation. We used a nonbiased data-mining approach to identify NFkappaB as the likely transcriptional regulator of these events. We confirmed NFkappaB activation by two independent methods: translocation of NFkappaB p50 to glomerular nuclei and ChIP assays demonstrating NFkappaB p50 binding to the kappaB motif of target genes in old versus young glomeruli. These data suggest that old glomeruli exhibit NFkappaB-associated up-regulation of a proinflammatory, procoagulable, and profibrotic phenotype compared with young glomeruli; these distinctions could explain their enhanced susceptibility to failure. Furthermore, these results provide a potential mechanistic explanation for the close relationship between ESRD and atherosclerotic organ failure as two parallel arms of age-associated NFkappaB-driven processes.


Subject(s)
Blood Coagulation , Inflammation/etiology , Kidney Glomerulus/pathology , NF-kappa B/physiology , Age Factors , Animals , Fibrosis/etiology , Gene Expression Regulation , Male , NF-kappa B/genetics , Rats , Rats, Inbred F344
9.
Am J Physiol Renal Physiol ; 297(1): F138-44, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19403647

ABSTRACT

Glomerular capillary filtration barrier characteristics are determined in part by the slit-pore junctions of glomerular podocytes. Protein tyrosine phosphatase receptor-O (PTPro) is a transmembrane protein expressed on the apical surface of podocyte foot processes. Tyrosine phosphorylation of podocyte proteins including nephrin may control the filtration barrier. To determine whether PTPro activity is required to maintain glomerular macromolecular permeability, albumin permeability (P(alb)) was studied after incubation of glomeruli from normal animals with a series of monoclonal (mAb) and polyclonal antibodies. Reagents included mAbs to rabbit and rat PTPro and polyclonal rabbit immune IgG to rat PTPro. mAb 4C3, specific to the amino acid core of PTPro, decreased its phosphatase activity and increased P(alb) of rabbit glomeruli in a time- and concentration-dependent manner. In contrast, mAb P8E7 did not diminish phosphatase activity and did not alter P(alb). Preincubation of 4C3 with PTPro extracellular domain fusion protein blocked glomerular binding and abolished permeability activity. In parallel experiments, P(alb) of rat glomeruli was increased by two mAbs (1B4 and 1D1) or by polyclonal anti-rat PTPro. We conclude that PTPro interaction with specific antibodies acutely increases P(alb). The identity of the normal ligand for PTPro and of its substrate, as well as the mechanism by which phosphatase activity of this receptor affects the filtration barrier, remain to be determined.


Subject(s)
Albumins/metabolism , Antibodies, Monoclonal/pharmacology , Cell Membrane Permeability/drug effects , Kidney Glomerulus/metabolism , Receptor-Like Protein Tyrosine Phosphatases, Class 3/immunology , Albuminuria/metabolism , Animals , Disease Models, Animal , Dose-Response Relationship, Drug , Glomerular Filtration Rate/drug effects , Kidney Glomerulus/drug effects , Podocytes/drug effects , Podocytes/metabolism , Protein Binding , Protein Structure, Tertiary/drug effects , Rabbits , Rats , Receptor-Like Protein Tyrosine Phosphatases, Class 3/metabolism , Time Factors
10.
J Am Soc Nephrol ; 20(5): 1041-52, 2009 May.
Article in English | MEDLINE | ID: mdl-19389856

ABSTRACT

Because loss of podocytes associates with glomerulosclerosis, monitoring podocyte loss by measuring podocyte products in urine may be clinically useful. To determine whether a single episode of podocyte injury would cause persistent podocyte loss, we induced limited podocyte depletion using a diphtheria toxin receptor (hDTR) transgenic rat. We monitored podocyte loss by detecting nephrin and podocin mRNA in urine particulates with quantitative reverse transcriptase-PCR. Aquaporin 2 mRNA served as a kidney reference gene to account for variable kidney contribution to RNA amount and quality. We found that a single injection of diphtheria toxin resulted in an initial peak of proteinuria and podocyte mRNAs (podocin and nephrin) followed 8 d later by a second peak of proteinuria and podocyte mRNAs that were podocin positive but nephrin negative. Proteinuria that persisted for months correlated with podocin-positive, nephrin-negative mRNAs in urine. Animals with persistent podocyte mRNA in urine progressed to ESRD with global podocyte depletion and interstitial scarring. Podocytes in ectatic tubules expressed podocalyxin and podocin proteins but not nephrin, compatible with detached podocytes' having an altered phenotype. Parallel human studies showed that biopsy-proven glomerular injury associated with increased urinary podocin:aquaporin 2 and nephrin:aquaporin 2 molar ratios. We conclude that a single episode of podocyte injury can trigger glomerular destabilization, resulting in persistent podocyte loss and an altered phenotype of podocytes recovered from urine. Podocyte mRNAs in urine may be a useful clinical tool for the diagnosis and monitoring of glomerular diseases.


Subject(s)
Kidney Cortex Necrosis/pathology , Kidney Diseases/genetics , Kidney Diseases/pathology , Kidney Glomerulus/pathology , Podocytes/pathology , RNA, Messenger/urine , Animals , Animals, Genetically Modified , Aquaporin 2/genetics , Biomarkers , Disease Progression , Female , Heparin-binding EGF-like Growth Factor , Humans , Intercellular Signaling Peptides and Proteins/genetics , Kidney Cortex Necrosis/genetics , Kidney Failure, Chronic/genetics , Kidney Failure, Chronic/pathology , Male , Proteinuria/genetics , Proteinuria/pathology , RNA, Messenger/genetics , Rats , Reference Values
11.
J Am Soc Nephrol ; 17(5): 1382-7, 2006 May.
Article in English | MEDLINE | ID: mdl-16597684

ABSTRACT

Biologic aging is accelerated by high-calorie intake, increased free radical production, and oxidation of key biomolecules. Fischer 344 rats that are maintained on an ad libitum diet develop oxidant injury and age-associated glomerulosclerosis by 24 mo. Calorie restriction prevents both oxidant injury and glomerulosclerosis. Ceruloplasmin (Cp) is a copper-containing ferroxidase that functions as an antioxidant in part by oxidizing toxic ferrous iron to nontoxic ferric iron. Glomerular Cp mRNA and protein expression were measured in ad libitum-fed and calorie-restricted rats at ages 2, 6, 17, and 24 mo. In ad libitum-fed rats, Cp mRNA expression increased six-fold (P < 0.01) and protein expression increased five-fold (P = 0.01) between 2 and 24 mo of age. In calorie-restricted rats, Cp mRNA expression increased three-fold (P < 0.01) and protein expression increased 1.6-fold (NS) between 2 and 24 mo of age. Both the cell-associated alternately spliced variant and secreted variants of Cp were expressed. Immunofluorescent analysis showed that Cp was expressed by the parietal epithelial cells that line the inner aspect of Bowman's capsule in the glomerulus. Cp also was present in urine, particularly of old ad libitum-fed rats with high tissue Cp expression. Cp expression by Bowman's capsule epithelial cells therefore occurred in direct proportion to known levels of oxidant activity (older age and high-calorie diet) and is secreted into the urine. It is suggested that Cp expression at this site may be part of the repertoire of the glomerular parietal epithelial cell to protect the glomerular podocytes and the downstream nephron from toxic effects of filtered molecules, including ferrous iron.


Subject(s)
Aging/metabolism , Ceruloplasmin/metabolism , Energy Intake/physiology , Epithelial Cells/metabolism , Kidney Glomerulus/metabolism , Animals , Antioxidants/metabolism , Cells, Cultured , Ceruloplasmin/urine , Rats , Rats, Inbred F344
12.
J Am Soc Nephrol ; 16(10): 2953-66, 2005 Oct.
Article in English | MEDLINE | ID: mdl-16120818

ABSTRACT

Whether podocyte depletion could cause the glomerulosclerosis of aging in Fischer 344 rats at ages 2, 6, 17, and 24 mo was evaluated. Ad libitum-fed rats developed proteinuria and glomerulosclerosis by 24 mo, whereas calorie-restricted rats did not. No evidence of age-associated progressive linear loss of podocytes from glomeruli was found. Rather, ad libitum-fed rats developed glomerular enlargement over time. To accommodate the increased glomerular volume, podocytes principally underwent hypertrophy, whereas other glomerular cells underwent hyperplasia. Stages of hypertrophy through which podocytes pass en route to podocyte loss and glomerulosclerosis were identified: Stage 1, normal podocyte; stage 2, nonstressed podocyte hypertrophy; stage 3, "adaptive" podocyte hypertrophy manifest by changes in synthesis of structural components (e.g., desmin) but maintenance of normal function; stage 4, "decompensated" podocyte hypertrophy relative to total glomerular volume manifest by reduced production of key machinery necessary for normal podocyte function (e.g., Wilms' tumor 1 protein [WT1], transcription factor pod1, nephrin, glomerular epithelial protein 1, podocalyxin, vascular endothelial growth factor, and alpha5 type IV collagen) and associated with widened foot processes and decreased filter efficiency (proteinuria); and stage 5, podocyte numbers decrease in association with focal segmental glomerulosclerosis. In contrast, in calorie-restricted rats, glomerular enlargement was minor, significant podocyte hypertrophy did not occur, podocyte machinery was unchanged, there was no proteinuria, and glomerulosclerosis did not develop. Glomerular enlargement therefore was associated with podocyte hypertrophy rather than hyperplasia. Hypertrophy above a certain threshold was associated with podocyte stress and then failure, culminating in reduced podocyte numbers in sclerotic glomeruli. This process could be prevented by calorie restriction.


Subject(s)
Caloric Restriction , Glomerulosclerosis, Focal Segmental/etiology , Glomerulosclerosis, Focal Segmental/prevention & control , Kidney Glomerulus/pathology , Podocytes/pathology , Adaptation, Physiological , Age Factors , Animals , Cell Count , Energy Intake , Hypertrophy , Podocytes/physiology , Rats , Rats, Inbred F344
13.
J Am Soc Nephrol ; 16(10): 2941-52, 2005 Oct.
Article in English | MEDLINE | ID: mdl-16107576

ABSTRACT

Glomerular injury and proteinuria in diabetes (types 1 and 2) and IgA nephropathy is related to the degree of podocyte depletion in humans. For determining the causal relationship between podocyte depletion and glomerulosclerosis, a transgenic rat strain in which the human diphtheria toxin receptor is specifically expressed in podocytes was developed. The rodent homologue does not act as a diphtheria toxin (DT) receptor, thereby making rodents resistant to DT. Injection of DT into transgenic rats but not wild-type rats resulted in dose-dependent podocyte depletion from glomeruli. Three stages of glomerular injury caused by podocyte depletion were identified: Stage 1, 0 to 20% depletion showed mesangial expansion, transient proteinuria and normal renal function; stage 2, 21 to 40% depletion showed mesangial expansion, capsular adhesions (synechiae), focal segmental glomerulosclerosis, mild persistent proteinuria, and normal renal function; and stage 3, >40% podocyte depletion showed segmental to global glomerulosclerosis with sustained high-grade proteinuria and reduced renal function. These pathophysiologic consequences of podocyte depletion parallel similar degrees of podocyte depletion, glomerulosclerosis, and proteinuria seen in diabetic glomerulosclerosis. This model system provides strong support for the concept that podocyte depletion could be a major mechanism driving glomerulosclerosis and progressive loss of renal function in human glomerular diseases.


Subject(s)
Glomerulosclerosis, Focal Segmental/etiology , Podocytes/drug effects , Podocytes/physiology , Receptors, Cell Surface/biosynthesis , Animals , Cell Count , Diphtheria Toxin/pharmacology , Heparin-binding EGF-like Growth Factor , Humans , Intercellular Signaling Peptides and Proteins , Rats , Rats, Inbred F344 , Receptors, Cell Surface/genetics , Transgenes
14.
J Am Soc Nephrol ; 14(10): 2484-93, 2003 Oct.
Article in English | MEDLINE | ID: mdl-14514726

ABSTRACT

Podocyte loss and glomerular hypertrophy are associated with development of glomerulosclerosis, suggesting that there may be a maximal area for each podocyte in terms of its capacity to support and maintain the glomerular filter. This study hypothesized that exceeding this maximal threshold will result in mesangial expansion and glomerulosclerosis. It may therefore be useful to measure podocyte number, glomerular volume, and glomerular volume per podocyte in clinical biopsy samples. An approach that uses thick and thin histologic sections cut from paraffin-embedded tissue to measure Wilms' tumor-1 protein-positive podocyte nuclear number and glomerular tuft area was studied. A rat model of aging has been used to track changes in glomerular podocyte number, glomerular volume per podocyte, and glomerular volume. Implications for clinical use of these variables are discussed.


Subject(s)
Carrier Proteins/metabolism , Cell Count/methods , DNA-Binding Proteins/metabolism , Kidney Glomerulus/cytology , Microtomy/methods , Nuclear Proteins/metabolism , Aging , Animals , Biomarkers , Cell Nucleus/metabolism , Kidney Cortex/cytology , Kidney Cortex/metabolism , Kidney Glomerulus/metabolism , Rats , Rats, Inbred BN
SELECTION OF CITATIONS
SEARCH DETAIL
...