Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
Add more filters










Publication year range
1.
Physiol Rep ; 12(13): e16129, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38955668

ABSTRACT

Cardiotrophin-1 (CT-1), a member of the interleukin (IL)-6 cytokine family, has renoprotective effects in mouse models of acute kidney disease and tubulointerstitial fibrosis, but its role in glomerular disease is unknown. To address this, we used the mouse model of nephrotoxic nephritis to test the hypothesis that CT-1 also has a protective role in immune-mediated glomerular disease. Using immunohistochemistry and analysis of single-cell RNA-sequencing data of isolated glomeruli, we demonstrate that CT-1 is expressed in the glomerulus in male mice, predominantly in parietal epithelial cells and is downregulated in mice with nephrotoxic nephritis. Furthermore, analysis of data from patients revealed that human glomerular disease is also associated with reduced glomerular CT-1 transcript levels. In male mice with nephrotoxic nephritis and established proteinuria, administration of CT-1 resulted in reduced albuminuria, prevented podocyte loss, and sustained plasma creatinine, compared with mice administered saline. CT-1 treatment also reduced fibrosis in the kidney cortex, peri-glomerular macrophage accumulation and the kidney levels of the pro-inflammatory mediator complement component 5a. In conclusion, CT-1 intervention therapy delays the progression of glomerular disease in mice by preserving kidney function and inhibiting renal inflammation and fibrosis.


Subject(s)
Cytokines , Kidney Glomerulus , Mice, Inbred C57BL , Animals , Male , Cytokines/metabolism , Cytokines/genetics , Mice , Kidney Glomerulus/metabolism , Kidney Glomerulus/pathology , Disease Models, Animal , Humans , Fibrosis , Glomerulonephritis/metabolism , Glomerulonephritis/pathology , Glomerulonephritis/drug therapy
2.
Sci Rep ; 12(1): 12172, 2022 07 16.
Article in English | MEDLINE | ID: mdl-35842494

ABSTRACT

Plasma ultrafiltration in the kidney occurs across glomerular capillaries, which are surrounded by epithelial cells called podocytes. Podocytes have a unique shape maintained by a complex cytoskeleton, which becomes disrupted in glomerular disease resulting in defective filtration and albuminuria. Lack of endogenous thymosin ß4 (TB4), an actin sequestering peptide, exacerbates glomerular injury and disrupts the organisation of the podocyte actin cytoskeleton, however, the potential of exogenous TB4 therapy to improve podocyte injury is unknown. Here, we have used Adriamycin (ADR), a toxin which injures podocytes and damages the glomerular filtration barrier leading to albuminuria in mice. Through interrogating single-cell RNA-sequencing data of isolated glomeruli we demonstrate that ADR injury results in reduced levels of podocyte TB4. Administration of an adeno-associated viral vector encoding TB4 increased the circulating level of TB4 and prevented ADR-induced podocyte loss and albuminuria. ADR injury was associated with disorganisation of the podocyte actin cytoskeleton in vitro, which was ameliorated by treatment with exogenous TB4. Collectively, we propose that systemic gene therapy with TB4 prevents podocyte injury and maintains glomerular filtration via protection of the podocyte cytoskeleton thus presenting a novel treatment strategy for glomerular disease.


Subject(s)
Kidney Diseases , Podocytes , Albuminuria , Animals , Cells, Cultured , Doxorubicin , Genetic Therapy , Kidney Glomerulus , Mice , Thymosin
3.
J Am Soc Nephrol ; 32(7): 1713-1732, 2021 Jul.
Article in English | MEDLINE | ID: mdl-34049963

ABSTRACT

BACKGROUND: Accumulation of extracellular matrix in organs and tissues is a feature of both aging and disease. In the kidney, glomerulosclerosis and tubulointerstitial fibrosis accompany the decline in function, which current therapies cannot address, leading to organ failure. Although histologic and ultrastructural patterns of excess matrix form the basis of human disease classifications, a comprehensive molecular resolution of abnormal matrix is lacking. METHODS: Using mass spectrometry-based proteomics, we resolved matrix composition over age in mouse models of kidney disease. We compared the changes in mice with a global characterization of human kidneymatrix during aging and to existing kidney disease datasets to identify common molecular features. RESULTS: Ultrastructural changes in basement membranes are associated with altered cell adhesion and metabolic processes and with distinct matrix proteomes during aging and kidney disease progression in mice. Within the altered matrix, basement membrane components (laminins, type IV collagen, type XVIII collagen) were reduced and interstitial matrix proteins (collagens I, III, VI, and XV; fibrinogens; and nephronectin) were increased, a pattern also seen in human kidney aging. Indeed, this signature of matrix proteins was consistently modulated across all age and disease comparisons, and the increase in interstitial matrix was also observed in human kidney disease datasets. CONCLUSIONS: This study provides deep molecular resolution of matrix accumulation in kidney aging and disease, and identifies a common signature of proteins that provides insight into mechanisms of response to kidney injury and repair.

4.
Proc Natl Acad Sci U S A ; 117(26): 15137-15147, 2020 06 30.
Article in English | MEDLINE | ID: mdl-32554502

ABSTRACT

RNA modifications play a fundamental role in cellular function. Pseudouridylation, the most abundant RNA modification, is catalyzed by the H/ACA small ribonucleoprotein (snoRNP) complex that shares four core proteins, dyskerin (DKC1), NOP10, NHP2, and GAR1. Mutations in DKC1, NOP10, or NHP2 cause dyskeratosis congenita (DC), a disorder characterized by telomere attrition. Here, we report a phenotype comprising nephrotic syndrome, cataracts, sensorineural deafness, enterocolitis, and early lethality in two pedigrees: males with DKC1 p.Glu206Lys and two children with homozygous NOP10 p.Thr16Met. Females with heterozygous DKC1 p.Glu206Lys developed cataracts and sensorineural deafness, but nephrotic syndrome in only one case of skewed X-inactivation. We found telomere attrition in both pedigrees, but no mucocutaneous abnormalities suggestive of DC. Both mutations fall at the dyskerin-NOP10 binding interface in a region distinct from those implicated in DC, impair the dyskerin-NOP10 interaction, and disrupt the catalytic pseudouridylation site. Accordingly, we found reduced pseudouridine levels in the ribosomal RNA (rRNA) of the patients. Zebrafish dkc1 mutants recapitulate the human phenotype and show reduced 18S pseudouridylation, ribosomal dysregulation, and a cell-cycle defect in the absence of telomere attrition. We therefore propose that this human disorder is the consequence of defective snoRNP pseudouridylation and ribosomal dysfunction.


Subject(s)
Cataract/genetics , Cell Cycle Proteins/genetics , Enterocolitis/genetics , Hearing Loss, Sensorineural/genetics , Nephrotic Syndrome/genetics , Nuclear Proteins/genetics , Ribonucleoproteins, Small Nucleolar/genetics , Animals , Child , Female , Genetic Predisposition to Disease , Humans , Longevity , Male , Models, Molecular , Molecular Dynamics Simulation , Mutation , Pedigree , Protein Conformation , RNA, Ribosomal/genetics , Zebrafish
5.
Methods Mol Biol ; 2067: 25-39, 2020.
Article in English | MEDLINE | ID: mdl-31701443

ABSTRACT

With the advances in next-generation sequencing and rapid filtering of candidate variants in diseased patients, it has been increasingly important to develop translatable in vivo models to study genetic changes. This allows for functional validation of pathogenic mutations and establishes a system to understand the etiology of disease. Due to the ease of genetic manipulation and rapid ex utero development, the zebrafish has become a valuable resource to study important biological processes, including nephrogenesis. The development and function of the zebrafish pronephros are akin to that of mammals. As such, they offer a tractable model to study kidney disease, especially diabetic nephropathy. However, in order to study kidney dysfunction in zebrafish it is imperative that an appropriate readout is available. The appearance of macro-proteins in patient's urine is indicative of defective kidney function. In this technical chapter, we describe the in vivo use of fluorescently tagged dextrans of different molecular weights to reveal the integrity of the zebrafish glomerular filtration barrier.


Subject(s)
Glomerular Filtration Barrier/pathology , Pronephros/pathology , Animals , Animals, Genetically Modified , Dextrans/chemistry , Dextrans/metabolism , Diabetic Nephropathies/diagnosis , Diabetic Nephropathies/genetics , Diabetic Nephropathies/pathology , Diabetic Nephropathies/urine , Disease Models, Animal , Embryo, Nonmammalian/physiology , Female , Fluorescent Dyes/chemistry , Fluorescent Dyes/metabolism , Genes, Reporter/genetics , Glomerular Filtration Barrier/physiology , Kidney Glomerulus/metabolism , Kidney Glomerulus/pathology , Male , Pronephros/metabolism , Zebrafish , Zebrafish Proteins/genetics , Zebrafish Proteins/metabolism
6.
Elife ; 82019 12 06.
Article in English | MEDLINE | ID: mdl-31808745

ABSTRACT

Heterogeneity of lymphatic vessels during embryogenesis is critical for organ-specific lymphatic function. Little is known about lymphatics in the developing kidney, despite their established roles in pathology of the mature organ. We performed three-dimensional imaging to characterize lymphatic vessel formation in the mammalian embryonic kidney at single-cell resolution. In mouse, we visually and quantitatively assessed the development of kidney lymphatic vessels, remodeling from a ring-like anastomosis under the nascent renal pelvis; a site of VEGF-C expression, to form a patent vascular plexus. We identified a heterogenous population of lymphatic endothelial cell clusters in mouse and human embryonic kidneys. Exogenous VEGF-C expanded the lymphatic population in explanted mouse embryonic kidneys. Finally, we characterized complex kidney lymphatic abnormalities in a genetic mouse model of polycystic kidney disease. Our study provides novel insights into the development of kidney lymphatic vasculature; a system which likely has fundamental roles in renal development, physiology and disease.


Subject(s)
Kidney/metabolism , Lymphangiogenesis/genetics , Lymphatic Vessels/metabolism , Polycystic Kidney Diseases/genetics , Animals , Gene Expression Regulation, Developmental , Genetic Heterogeneity , Humans , Kidney/embryology , Kinetics , Lymphatic Vessels/embryology , Mammals/embryology , Mammals/genetics , Mammals/metabolism , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Polycystic Kidney Diseases/embryology , Polycystic Kidney Diseases/metabolism , Spatio-Temporal Analysis , Vascular Endothelial Growth Factor C/genetics , Vascular Endothelial Growth Factor C/metabolism
7.
JACC Basic Transl Sci ; 4(1): 15-26, 2019 Feb.
Article in English | MEDLINE | ID: mdl-30847415

ABSTRACT

The authors hypothesized that despite similar cardiovascular event rates, the improved cardiovascular survival from sodium glucose transporter 2 (SGLT2) inhibition, seen clinically, could be via a direct cytoprotective effect, including protection against myocardial ischemia/reperfusion injury. Langendorff-perfused hearts, from diabetic and nondiabetic rats, fed long-term for 4 weeks with canagliflozin, had lower infarct sizes; this being the first demonstration of canagliflozin's cardioprotective effect against ischemia/reperfusion injury in both diabetic and nondiabetic animals. By contrast, direct treatment of isolated nondiabetic rat hearts with canagliflozin, solubilized in the isolated Langendorff perfusion buffer, had no impact on infarct size. This latter study demonstrates that the infarct-sparing effect of long-term treatment with canagliflozin results from either a glucose-independent effect or up-regulation of cardiac prosurvival pathways. These results further suggest that SGLT2 inhibitors could be repurposed as novel cardioprotective interventions in high-risk cardiovascular patients irrespective of diabetic status.

8.
J Pathol ; 246(4): 485-496, 2018 12.
Article in English | MEDLINE | ID: mdl-30125361

ABSTRACT

Planar cell polarity (PCP) pathways control the orientation and alignment of epithelial cells within tissues. Van Gogh-like 2 (Vangl2) is a key PCP protein that is required for the normal differentiation of kidney glomeruli and tubules. Vangl2 has also been implicated in modifying the course of acquired glomerular disease, and here, we further explored how Vangl2 impacts on glomerular pathobiology in this context. Targeted genetic deletion of Vangl2 in mouse glomerular epithelial podocytes enhanced the severity of not only irreversible accelerated nephrotoxic nephritis but also lipopolysaccharide-induced reversible glomerular damage. In each proteinuric model, genetic deletion of Vangl2 in podocytes was associated with an increased ratio of active-MMP9 to inactive MMP9, an enzyme involved in tissue remodelling. In addition, by interrogating microarray data from two cohorts of renal patients, we report increased VANGL2 transcript levels in the glomeruli of individuals with focal segmental glomerulosclerosis, suggesting that the molecule may also be involved in certain human glomerular diseases. These observations support the conclusion that Vangl2 modulates glomerular injury, at least in part by acting as a brake on MMP9, a potentially harmful endogenous enzyme. © 2018 The Authors. The Journal of Pathology published by John Wiley & Sons Ltd on behalf of Pathological Society of Great Britain and Ireland.


Subject(s)
Cell Polarity , Glomerulosclerosis, Focal Segmental/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Kidney Glomerulus/metabolism , Membrane Proteins/metabolism , Nephrosis, Lipoid/metabolism , Nerve Tissue Proteins/metabolism , Podocytes/metabolism , Adult , Animals , Case-Control Studies , Cells, Cultured , Disease Models, Animal , Enzyme Activation , Female , Glomerulosclerosis, Focal Segmental/genetics , Glomerulosclerosis, Focal Segmental/pathology , Glomerulosclerosis, Focal Segmental/physiopathology , Humans , Intracellular Signaling Peptides and Proteins/genetics , Kidney Glomerulus/pathology , Kidney Glomerulus/physiopathology , Male , Matrix Metalloproteinase 9/metabolism , Membrane Proteins/genetics , Mice, Inbred C57BL , Mice, Knockout , Middle Aged , Nephrosis, Lipoid/genetics , Nephrosis, Lipoid/pathology , Nephrosis, Lipoid/physiopathology , Nerve Tissue Proteins/deficiency , Nerve Tissue Proteins/genetics , Podocytes/pathology , Signal Transduction , Young Adult
9.
Cell Death Discov ; 4: 13, 2018 Dec.
Article in English | MEDLINE | ID: mdl-29531810

ABSTRACT

Kidney function is directly linked to the number of nephrons which are generated until 32-36 weeks gestation in humans. Failure to make nephrons during development leads to congenital renal malformations, whilst nephron loss in adulthood occurs in progressive renal disease. Therefore, an understanding of the molecular processes which underlie human nephron development may help design new treatments for renal disease. Mesenchyme to epithelial transition (MET) is critical for forming nephrons, and molecular pathways which control rodent MET have been identified. However, we do not know whether they are relevant in human kidney development. In this study, we isolated mesenchymal cell lines derived from human first trimester kidneys in monolayer culture and investigated their differentiation potential. We found that the mesenchymal cells could convert into osteogenic, but not adipogenic or endothelial lineages. Furthermore, addition of lithium chloride led to MET which was accompanied by increases in epithelial (CDH1) and tubular (ENPEP) markers and downregulation of renal progenitor (SIX2, EYA1, CD133) and mesenchymal markers (HGF, CD24). Prior to phenotypic changes, lithium chloride altered Wnt signalling with elevations in AXIN2, GSK3ß phosphorylation and ß-catenin. Collectively, these studies provide the first evidence that lithium-induced Wnt activation causes MET in human kidneys. Therapies targeting Wnts may be critical in the quest to regenerate nephrons for human renal diseases.

10.
Dis Model Mech ; 10(4): 409-423, 2017 04 01.
Article in English | MEDLINE | ID: mdl-28237967

ABSTRACT

Lung diseases impose a huge economic and health burden worldwide. A key aspect of several adult lung diseases, such as idiopathic pulmonary fibrosis (IPF) and chronic obstructive pulmonary disease (COPD), including emphysema, is aberrant tissue repair, which leads to an accumulation of damage and impaired respiratory function. Currently, there are few effective treatments available for these diseases and their incidence is rising. The planar cell polarity (PCP) pathway is critical for the embryonic development of many organs, including kidney and lung. We have previously shown that perturbation of the PCP pathway impairs tissue morphogenesis, which disrupts the number and shape of epithelial tubes formed within these organs during embryogenesis. However, very little is known about the role of the PCP pathway beyond birth, partly because of the perinatal lethality of many PCP mouse mutant lines. Here, we investigate heterozygous Looptail (Lp) mice, in which a single copy of the core PCP gene, Vangl2, is disrupted. We show that these mice are viable but display severe airspace enlargement and impaired adult lung function. Underlying these defects, we find that Vangl2Lp/+ lungs exhibit altered distribution of actin microfilaments and abnormal regulation of the actin-modifying protein cofilin. In addition, we show that Vangl2Lp/+ lungs exhibit many of the hallmarks of tissue damage, including an altered macrophage population, abnormal elastin deposition and elevated levels of the elastin-modifying enzyme, Mmp12, all of which are observed in emphysema. In vitro, disruption of VANGL2 impairs directed cell migration and reduces the rate of repair following scratch wounding of human alveolar epithelial cells. Moreover, using population data from a birth cohort of young adults, all aged 31, we found evidence of an interactive effect between VANGL2 and smoking on lung function. Finally, we show that PCP genes VANGL2 and SCRIB are significantly downregulated in lung tissue from patients with emphysema. Our data reveal an important novel role for the PCP pathway in adult lung homeostasis and repair and shed new light on the genetic factors which may modify destructive lung diseases such as emphysema.


Subject(s)
Aging/pathology , Cell Polarity , Homeostasis , Lung/pathology , Nerve Tissue Proteins/genetics , Wound Healing , A549 Cells , Actin Cytoskeleton/metabolism , Animals , Cell Movement , Down-Regulation/genetics , Elastin/metabolism , Embryo, Mammalian/pathology , Gene Knockdown Techniques , Heterozygote , Humans , Lung/embryology , Lung/physiopathology , Macrophages/pathology , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice , Models, Biological , Mutation/genetics , Phenotype , Polymorphism, Genetic , Pulmonary Disease, Chronic Obstructive/genetics , Smoking/adverse effects , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/metabolism
11.
Kidney Int ; 90(6): 1274-1284, 2016 12.
Article in English | MEDLINE | ID: mdl-27597235

ABSTRACT

The mammalian kidney contains nephrons comprising glomeruli and tubules joined to ureteric bud-derived collecting ducts. It has a characteristic bean-like shape, with near-complete rostrocaudal symmetry around the hilum. Here we show that Celsr1, a planar cell polarity (PCP) gene implicated in neural tube morphogenesis, is required for ureteric tree growth in early development and later in gestation prevents tubule overgrowth. We also found an interaction between Celsr1 and Vangl2 (another PCP gene) in ureteric tree growth, most marked in the caudal compartment of the kidneys from compound heterozygous mutant mice with a stunted rump. Furthermore, these genes together are required for the maturation of glomeruli. Interestingly, we demonstrated patients with CELSR1 mutations and spina bifida can have significant renal malformations. Thus, PCP genes are important in mammalian kidney development and have an unexpected role in rostrocaudal patterning during organogenesis.


Subject(s)
Cell Polarity/genetics , Kidney/embryology , Nerve Tissue Proteins/physiology , Receptors, G-Protein-Coupled/physiology , Spinal Dysraphism/pathology , Animals , Humans , Kidney/pathology , Mice, Inbred C3H
12.
Kidney Int ; 90(5): 1056-1070, 2016 11.
Article in English | MEDLINE | ID: mdl-27575556

ABSTRACT

Glomerular disease is characterized by morphologic changes in podocyte cells accompanied by inflammation and fibrosis. Thymosin ß4 regulates cell morphology, inflammation, and fibrosis in several organs and administration of exogenous thymosin ß4 improves animal models of unilateral ureteral obstruction and diabetic nephropathy. However, the role of endogenous thymosin ß4 in the kidney is unknown. We demonstrate that thymosin ß4 is expressed prominently in podocytes of developing and adult mouse glomeruli. Global loss of thymosin ß4 did not affect healthy glomeruli, but accelerated the severity of immune-mediated nephrotoxic nephritis with worse renal function, periglomerular inflammation, and fibrosis. Lack of thymosin ß4 in nephrotoxic nephritis led to the redistribution of podocytes from the glomerular tuft toward the Bowman capsule suggesting a role for thymosin ß4 in the migration of these cells. Thymosin ß4 knockdown in cultured podocytes also increased migration in a wound-healing assay, accompanied by F-actin rearrangement and increased RhoA activity. We propose that endogenous thymosin ß4 is a modifier of glomerular injury, likely having a protective role acting as a brake to slow disease progression.


Subject(s)
Glomerulonephritis/metabolism , Podocytes/metabolism , Thymosin/metabolism , Animals , Cell Movement , Cells, Cultured , Cytoskeleton/metabolism , Fibrosis , Glomerulonephritis/pathology , Kidney Glomerulus/pathology , Macrophages , Male , Mice, Inbred C57BL , Mice, Knockout
13.
J Am Soc Nephrol ; 27(9): 2771-83, 2016 09.
Article in English | MEDLINE | ID: mdl-26892346

ABSTRACT

FSGS is a CKD with heavy proteinuria that eventually progresses to ESRD. Hereditary forms of FSGS have been linked to mutations in the transient receptor potential cation channel, subfamily C, member 6 (TRPC6) gene encoding a nonselective cation channel. Most of these TRPC6 mutations cause a gain-of-function phenotype, leading to calcium-triggered podocyte cell death, but the underlying molecular mechanisms are unclear. We studied the molecular effect of disease-related mutations using tridimensional in silico modeling of tetrameric TRPC6. Our results indicated that G757 is localized in a domain forming a TRPC6-TRPC6 interface and predicted that the amino acid exchange G757D causes local steric hindrance and disruption of the channel complex. Notably, functional characterization of model interface domain mutants suggested a loss-of-function phenotype. We then characterized 19 human FSGS-related TRPC6 mutations, the majority of which caused gain-of-function mutations. However, five mutations (N125S, L395A, G757D, L780P, and R895L) caused a loss-of-function phenotype. Coexpression of wild-type TRPC6 and TRPC6 G757D, mimicking heterozygosity observed in patients, revealed a dominant negative effect of TRPC6 G757D. Our comprehensive analysis of human disease-causing TRPC6 mutations reveals loss of TRPC6 function as an additional concept of hereditary FSGS and provides molecular insights into the mechanism responsible for the loss-of-function phenotype of TRPC6 G757D in humans.


Subject(s)
Glomerulosclerosis, Focal Segmental/genetics , Mutation , TRPC Cation Channels/genetics , DNA Mutational Analysis , Glomerulosclerosis, Focal Segmental/physiopathology , Humans , TRPC6 Cation Channel
14.
J Am Soc Nephrol ; 27(1): 69-77, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26038530

ABSTRACT

Polycystic kidney diseases (PKD) are genetic disorders characterized by progressive epithelial cyst growth leading to destruction of normally functioning renal tissue. Current therapies have focused on the cyst epithelium, and little is known about how the blood and lymphatic microvasculature modulates cystogenesis. Hypomorphic Pkd1(nl/nl) mice were examined, showing that cystogenesis was associated with a disorganized pericystic network of vessels expressing platelet/endothelial cell adhesion molecule 1 and vascular endothelial growth factor receptor 3 (VEGFR3). The major ligand for VEGFR3 is VEGFC, and there were lower levels of Vegfc mRNA within the kidneys during the early stages of cystogenesis in 7-day-old Pkd1(nl/nl) mice. Seven-day-old mice were treated with exogenous VEGFC for 2 weeks on the premise that this would remodel both the VEGFR3(+) pericystic vascular network and larger renal lymphatics that may also affect the severity of PKD. Treatment with VEGFC enhanced VEGFR3 phosphorylation in the kidney, normalized the pattern of the pericystic network of vessels, and widened the large lymphatics in Pkd1(nl/nl) mice. These effects were associated with significant reductions in cystic disease, BUN and serum creatinine levels. Furthermore, VEGFC administration reduced M2 macrophage pericystic infiltrate, which has been implicated in the progression of PKD. VEGFC administration also improved cystic disease in Cys1(cpk/cpk) mice, a model of autosomal recessive PKD, leading to a modest but significant increase in lifespan. Overall, this study highlights VEGFC as a potential new treatment for some aspects of PKD, with the possibility for synergy with current epithelially targeted approaches.


Subject(s)
Polycystic Kidney Diseases/drug therapy , Vascular Endothelial Growth Factor C/therapeutic use , Animals , Mice , Polycystic Kidney Diseases/etiology , Vascular Endothelial Growth Factor C/physiology
15.
J Am Soc Nephrol ; 26(12): 3021-34, 2015 Dec.
Article in English | MEDLINE | ID: mdl-25896609

ABSTRACT

Glomerular disease often features altered histologic patterns of extracellular matrix (ECM). Despite this, the potential complexities of the glomerular ECM in both health and disease are poorly understood. To explore whether genetic background and sex determine glomerular ECM composition, we investigated two mouse strains, FVB and B6, using RNA microarrays of isolated glomeruli combined with proteomic glomerular ECM analyses. These studies, undertaken in healthy young adult animals, revealed unique strain- and sex-dependent glomerular ECM signatures, which correlated with variations in levels of albuminuria and known predisposition to progressive nephropathy. Among the variation, we observed changes in netrin 4, fibroblast growth factor 2, tenascin C, collagen 1, meprin 1-α, and meprin 1-ß. Differences in protein abundance were validated by quantitative immunohistochemistry and Western blot analysis, and the collective differences were not explained by mutations in known ECM or glomerular disease genes. Within the distinct signatures, we discovered a core set of structural ECM proteins that form multiple protein-protein interactions and are conserved from mouse to man. Furthermore, we found striking ultrastructural changes in glomerular basement membranes in FVB mice. Pathway analysis of merged transcriptomic and proteomic datasets identified potential ECM regulatory pathways involving inhibition of matrix metalloproteases, liver X receptor/retinoid X receptor, nuclear factor erythroid 2-related factor 2, notch, and cyclin-dependent kinase 5. These pathways may therefore alter ECM and confer susceptibility to disease.


Subject(s)
Extracellular Matrix/genetics , Extracellular Matrix/metabolism , Kidney Diseases/genetics , Kidney Glomerulus/metabolism , Albuminuria/genetics , Albuminuria/metabolism , Animals , Collagen Type I/genetics , Collagen Type I/metabolism , Cyclin-Dependent Kinase 5/metabolism , Extracellular Matrix/ultrastructure , Female , Fibroblast Growth Factor 2/genetics , Fibroblast Growth Factor 2/metabolism , Genetic Predisposition to Disease , Glomerular Basement Membrane/ultrastructure , Kidney Diseases/metabolism , Liver X Receptors , Male , Matrix Metalloproteinases/metabolism , Metalloendopeptidases/genetics , Metalloendopeptidases/metabolism , Mice , Mice, Inbred Strains , NF-E2-Related Factor 2/metabolism , Nerve Growth Factors/genetics , Nerve Growth Factors/metabolism , Netrins , Oligonucleotide Array Sequence Analysis , Orphan Nuclear Receptors/metabolism , RNA/analysis , Sex Factors , Signal Transduction , Tenascin/genetics , Tenascin/metabolism
16.
J Am Soc Nephrol ; 25(1): 33-42, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24009238

ABSTRACT

Vascular growth factors play an important role in maintaining the structure and integrity of the glomerular filtration barrier. In healthy adult glomeruli, the proendothelial survival factors vascular endothelial growth factor-A (VEGF-A) and angiopoietin-1 are constitutively expressed in glomerular podocyte epithelia. We demonstrate that this milieu of vascular growth factors is altered in streptozotocin-induced type 1 diabetic mice, with decreased angiopoietin-1 levels, VEGF-A upregulation, decreased soluble VEGF receptor-1 (VEGFR1), and increased VEGFR2 phosphorylation. This was accompanied by marked albuminuria, nephromegaly, hyperfiltration, glomerular ultrastructural alterations, and aberrant angiogenesis. We subsequently hypothesized that restoration of angiopoietin-1 expression within glomeruli might ameliorate manifestations of early diabetic glomerulopathy. Podocyte-specific inducible repletion of angiopoietin-1 in diabetic mice caused a 70% reduction of albuminuria and prevented diabetes-induced glomerular endothelial cell proliferation; hyperfiltration and renal morphology were unchanged. Furthermore, angiopoietin-1 repletion in diabetic mice increased Tie-2 phosphorylation, elevated soluble VEGFR1, and was paralleled by a decrease in VEGFR2 phosphorylation and increased endothelial nitric oxide synthase Ser(1177) phosphorylation. Diabetes-induced nephrin phosphorylation was also reduced in mice with angiopoietin-1 repletion. In conclusion, targeted angiopoietin-1 therapy shows promise as a renoprotective tool in the early stages of diabetic kidney disease.


Subject(s)
Angiopoietin-1/metabolism , Diabetic Nephropathies/metabolism , Diabetic Nephropathies/therapy , Molecular Targeted Therapy , Angiopoietin-1/deficiency , Angiopoietin-1/genetics , Angiopoietin-2/genetics , Angiopoietin-2/metabolism , Animals , Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Experimental/pathology , Diabetes Mellitus, Experimental/therapy , Diabetic Nephropathies/pathology , Humans , Intracellular Signaling Peptides and Proteins/deficiency , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Male , Membrane Proteins/deficiency , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice , Mice, Inbred C57BL , Mice, Inbred CBA , Mice, Mutant Strains , Podocytes/metabolism , Podocytes/pathology , RNA, Messenger/genetics , RNA, Messenger/metabolism , Signal Transduction , Vascular Endothelial Growth Factor Receptor-1/metabolism , Vascular Endothelial Growth Factor Receptor-2/metabolism
17.
Kidney Int ; 83(6): 1118-29, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23447063

ABSTRACT

Normally, the glomerular filtration barrier almost completely excludes circulating albumin from entering the urine. Genetic variation and both pre- and postnatal environmental factors may affect albuminuria in humans. Here we determine whether glomerular gene expression in mouse strains with naturally occurring variations in albuminuria would allow identification of proteins deregulated in relatively 'leaky' glomeruli. Albuminuria increased in female B6 to male B6 to female FVB/N to male FVB/N mice, whereas the number of glomeruli/kidney was the exact opposite. Testosterone administration led to increased albuminuria in female B6 but not female FVB/N mice. A common set of 39 genes, many expressed in podocytes, were significantly differentially expressed in each of the four comparisons: male versus female B6 mice, male versus female FVB/N mice, male FVB/N versus male B6 mice, and female FVB/N versus female B6 mice. The transcripts encoded proteins involved in oxidation/reduction reactions, ion transport, and enzymes involved in detoxification. These proteins may represent novel biomarkers and even therapeutic targets for early kidney and cardiovascular disease.


Subject(s)
Albuminuria/etiology , Kidney Glomerulus/metabolism , Testosterone/metabolism , Albuminuria/genetics , Albuminuria/pathology , Albuminuria/urine , Animals , Blood Pressure , Cells, Cultured , Female , Gene Expression Profiling , Gene Expression Regulation , Genotype , Glomerular Filtration Barrier/metabolism , Kidney Glomerulus/pathology , Male , Mice , Mice, Inbred C57BL , Permeability , Phenotype , Podocytes/metabolism , RNA, Messenger/metabolism , Sex Factors , Species Specificity
18.
PLoS One ; 8(2): e56273, 2013.
Article in English | MEDLINE | ID: mdl-23409162

ABSTRACT

Cardiovascular disease (CVD) is increasingly recognised as a complication of childhood chronic kidney disease (CKD) even in the absence of diabetes and hypertension. We hypothesized that an alteration in angiopoietin-1 and -2, growth factors which regulate endothelial and vascular function could be involved. We report that the endothelial survival factor, angiopoietin-1 is low in children with pre-dialysis CKD whereas the pro-inflammatory angiopoietin-2 is elevated in children on dialysis. In dialysis patients, angiopoietin-2 positively correlated with time on dialysis, systolic blood pressure, and carotid artery intima media thickness. Elevated angiopoietin-2 levels in dialysis versus pre-dialysis CKD patients were also associated with an anti-angiogenic (high soluble VEGFR-1 and low VEGF-A) and pro-inflammatory (high urate, E-selectin, P-selectin and VCAM-1) milieu. Ang-2 was immunodetected in arterial biopsy samples whilst the expression of VEGF-A was significantly downregulated in dialysis patients. Serum urate correlated with angiopoietin-2 levels in dialysis patients and addition of uric acid was able to induce rapid release of angiopoietin-2 from cultured endothelial cells. Thus, angiopoietin-2 is a marker for cardiovascular disease in children on chronic dialysis and may act as an anti-angiogenic and pro-inflammatory effector in this context. The possibility that the release of angiopoietin-2 from endothelia is mediated by urate should be explored further.


Subject(s)
Angiopoietin-2/blood , Cardiovascular Diseases/complications , Renal Dialysis , Renal Insufficiency, Chronic/blood , Renal Insufficiency, Chronic/therapy , Adolescent , Angiopoietin-2/metabolism , Arteries/metabolism , Arteries/pathology , Biomarkers/blood , Cell Adhesion Molecules/blood , Cell Adhesion Molecules/chemistry , Child , Female , Human Umbilical Vein Endothelial Cells/drug effects , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Male , Renal Insufficiency, Chronic/pathology , Solubility , Time Factors , Uric Acid/pharmacology , Vascular Endothelial Growth Factor A/blood , Vascular Endothelial Growth Factor Receptor-1/blood
19.
Nephrol Dial Transplant ; 27(6): 2355-64, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22146311

ABSTRACT

BACKGROUND: Recently, we identified a microduplication in chromosomal band 1q21.1 encompassing the CHD1L/ALC1 gene encoding a chromatin-remodelling enzyme in congenital anomalies of the kidneys and urinary tract (CAKUT) patient. METHODS: To explore the role of CHD1L in CAKUT, we screened 85 CAKUT patients for mutations in the CHD1L gene and performed functional analyses of the three heterozygous missense variants detected. In addition, we quantitatively determined CHD1L expression in multiple human fetal and adult tissues and analysed expression of CHD1L protein in human embryonal, adult and hydronephrotic kidney sections. RESULTS: Two of three novel heterozygous missense variants identified in three patients were not found in >400 control chromosomes. All variants lead to amino acid substitutions in or near the CHD1L macro domain, a poly-ADP-ribose (PAR)-binding module interacting with PAR polymerase 1 (PARP1), and showed decreased interaction with PARP1 by pull-down assay of transfected cell lysates. Quantitative messenger RNA analysis demonstrated high CHD1L expression in human fetal kidneys, and levels were four times higher than in adult kidneys. In the human embryo at 7-11 weeks gestation, CHD1L immunolocalized in the early ureteric bud and the S- and comma-shaped bodies, critical stages of kidney development. In normal postnatal sections, CHD1L was expressed in the cytoplasm of tubular cells in all tubule segments. CHD1L expression appeared higher in the hydronephrotic kidney of one patient with a hypofunctional CHD1L variant than in normal kidneys, recapitulating high fetal levels. CONCLUSION: Our data suggest that CHD1L plays a role in kidney development and may be a new candidate gene for CAKUT.


Subject(s)
Congenital Abnormalities/genetics , DNA Helicases/genetics , DNA-Binding Proteins/genetics , Gene Expression Regulation, Developmental , Kidney/abnormalities , Mutation/genetics , Urinary Tract/abnormalities , Adult , Blotting, Western , Cells, Cultured , Child , Child, Preschool , DNA Helicases/metabolism , DNA-Binding Proteins/metabolism , Female , Fetus , Fluorescent Antibody Technique , Follow-Up Studies , Humans , Immunoenzyme Techniques , Immunoprecipitation , Infant , Infant, Newborn , Kidney/embryology , Kidney/metabolism , Male , Pedigree , Poly (ADP-Ribose) Polymerase-1 , Poly(ADP-ribose) Polymerases/genetics , Poly(ADP-ribose) Polymerases/metabolism , Prognosis , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Urinary Tract/embryology , Urinary Tract/metabolism
20.
Pediatr Nephrol ; 27(6): 991-8, 2012 Jun.
Article in English | MEDLINE | ID: mdl-21993971

ABSTRACT

BACKGROUND: Fraser syndrome (FS) features renal agenesis and cystic kidneys. Mutations of FRAS1 (Fraser syndrome 1)and FREM2 (FRAS1-related extracellular matrix protein 2)cause FS. They code for basement membrane proteins expressed in metanephric epithelia where they mediate epithelial/mesenchymal signalling. Little is known about whether and where these molecules are expressed in more mature kidneys. METHODS: In healthy and congenital polycystic kidney (cpk)mouse kidneys we sought Frem2 expression using a LacZ reporter gene and quantified Fras family transcripts. Fras1 immunohistochemistry was undertaken in cystic kidneys from cpk mice and PCK (Pkhd1 mutant) rats (models of autosomal recessive polycystic kidney disease) and in wildtype metanephroi rendered cystic by dexamethasone. RESULTS: Nascent nephrons transiently expressed Frem2 in both tubule and podocyte epithelia. Maturing and adult collecting ducts also expressed Frem2. Frem2 was expressed in cpk cystic epithelia although Frem2 haploinsufficiency did not significantly modify cystogenesis in vivo. Fras1 transcripts were significantly upregulated, and Frem3 downregulated, in polycystic kidneys versus the non-cystic kidneys of littermates. Fras1 was immunodetected in cpk, PCK and dexamethasone-induced cystepithelia. CONCLUSIONS: These descriptive results are consistent with the hypothesis that Fras family molecules play diverse roles in kidney epithelia. In future, this should be tested by conditional deletion of FS genes in nephron segments and collecting ducts.


Subject(s)
Extracellular Matrix Proteins/genetics , Fraser Syndrome/genetics , Membrane Proteins/genetics , Nephrons/metabolism , Polycystic Kidney, Autosomal Recessive/genetics , Animals , Dexamethasone/pharmacology , Disease Models, Animal , Embryo Culture Techniques , Extracellular Matrix Proteins/metabolism , Fraser Syndrome/metabolism , Fraser Syndrome/pathology , Gene Expression Regulation , Genes, Reporter , Immunohistochemistry , Lac Operon , Membrane Proteins/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , Mutation , Nephrons/drug effects , Nephrons/embryology , Nephrons/pathology , Polycystic Kidney, Autosomal Recessive/metabolism , Polycystic Kidney, Autosomal Recessive/pathology , Rats , Receptors, Cell Surface/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...