21 results on '"Hyink DP"'
Search Results
2. Role of SIRT1 in HIV-associated kidney disease.
- Author
-
Wang X, Liu R, Zhang W, Hyink DP, Das GC, Das B, Li Z, Wang A, Yuan W, Klotman PE, Lee K, and He JC
- Subjects
- AIDS-Associated Nephropathy complications, AIDS-Associated Nephropathy metabolism, Animals, Humans, Kidney metabolism, Kidney Glomerulus metabolism, Kidney Glomerulus virology, Mice, Podocytes metabolism, Renal Insufficiency, Chronic complications, Renal Insufficiency, Chronic pathology, Renal Insufficiency, Chronic virology, Transcription Factor RelA metabolism, AIDS-Associated Nephropathy virology, Renal Insufficiency, Chronic metabolism, Sirtuin 1 metabolism
- Abstract
Human immunodeficiency virus (HIV) infection of kidney cells can lead to HIV-associated nephropathy (HIVAN) and aggravate the progression of other chronic kidney diseases. Thus, a better understanding of the mechanisms of HIV-induced kidney cell injury is needed for effective therapy against HIV-induced kidney disease progression. We have previously shown that the acetylation and activation of key inflammatory regulators, NF-κB p65 and STAT3, were increased in HIVAN kidneys. Here, we demonstrate the key role of sirtuin 1 (SIRT1) deacetylase in the regulation of NF-κB and STAT3 activity in HIVAN. We found that SIRT1 expression was reduced in the glomeruli of human and mouse HIVAN kidneys and that HIV-1 gene expression was associated with reduced SIRT1 expression and increased acetylation of NF-κB p65 and STAT3 in cultured podocytes. Interestingly, SIRT1 overexpression, in turn, reduced the expression of negative regulatory factor in podocytes stably expressing HIV-1 proviral genes, which was associated with inactivation of NF-κB p65 and a reduction in HIV-1 long terminal repeat promoter activity. In vivo, the administration of the small-molecule SIRT1 agonist BF175 or inducible overexpression of SIRT1 specifically in podocytes markedly attenuated albuminuria, kidney lesions, and expression of inflammatory markers in Tg26 mice. Finally, we showed that the reduction in SIRT1 expression by HIV-1 is in part mediated through miR-34a expression. Together, our data provide a new mechanism of SIRT1 regulation and its downstream effects in HIV-1-infected kidney cells and indicate that SIRT1/miR-34a are potential drug targets to treat HIV-related kidney disease.
- Published
- 2020
- Full Text
- View/download PDF
3. Extracellular vesicles from human bone marrow mesenchymal stem cells repair organ damage caused by cadmium poisoning in a medaka model.
- Author
-
Matsukura T, Inaba C, Weygant EA, Kitamura D, Janknecht R, Matsumoto H, Hyink DP, Kashiwada S, and Obara T
- Subjects
- Animals, Bone Marrow Cells metabolism, Cadmium Poisoning metabolism, Cells, Cultured, Extracellular Vesicles metabolism, Humans, Kidney Tubules, Proximal metabolism, Kidney Tubules, Proximal pathology, Mesenchymal Stem Cells metabolism, Mitochondria metabolism, Mitochondria pathology, Oryzias, Podocytes metabolism, Podocytes pathology, Cadmium Poisoning therapy, Extracellular Vesicles transplantation, Mesenchymal Stem Cell Transplantation methods
- Abstract
Treatment modalities for kidney disease caused by long-term exposure to heavy metals, such as cadmium (Cd), are limited. Often, chronic, long-term environmental exposure to heavy metal is not recognized in the early stages; therefore, chelation therapy is not an effective option. Extracellular vesicles (EVs) derived from stem cells have been demonstrated to reduce disease pathology in both acute and chronic kidney disease models. To test the ability of EVs derived from human bone marrow mesenchymal stem cells (hBM-MSCs) to treat Cd damage, we generated a Cd-exposed medaka model. This model develops heavy metal-induced cell damage in various organs and tissues, and shows decreased overall survival. Intravenous injection of highly purified EVs from hBM-MSCs repaired the damage to apical and basolateral membranes and mitochondria of kidney proximal tubules, glomerular podocytes, bone deformation, and improved survival. Our system also serves as a model with which to study age- and sex-dependent cell injuries of organs caused by various agents and diseases. The beneficial effects of EVs on the tissue repair process, as shown in our novel Cd-exposed medaka model, may open new broad avenues for interventional strategies., (© 2019 The Authors. Physiological Reports published by Wiley Periodicals, Inc. on behalf of The Physiological Society and the American Physiological Society.)
- Published
- 2019
- Full Text
- View/download PDF
4. Epigenetic regulation of RCAN1 expression in kidney disease and its role in podocyte injury.
- Author
-
Li H, Zhang W, Zhong F, Das GC, Xie Y, Li Z, Cai W, Jiang G, Choi J, Sidani M, Hyink DP, Lee K, Klotman PE, and He JC
- Subjects
- AIDS-Associated Nephropathy virology, Animals, Biopsy, Calcium-Binding Proteins, Cells, Cultured, DNA Methylation drug effects, DNA Modification Methylases antagonists & inhibitors, DNA-Binding Proteins, Datasets as Topic, Decitabine pharmacology, Disease Models, Animal, Gene Knockout Techniques, Genome, Human genetics, Glucose metabolism, HIV-1, Humans, Intracellular Signaling Peptides and Proteins metabolism, Kidney Glomerulus pathology, Mice, Mice, Knockout, Muscle Proteins metabolism, NFATC Transcription Factors metabolism, Podocytes virology, Primary Cell Culture, Up-Regulation, AIDS-Associated Nephropathy pathology, Diabetic Nephropathies pathology, Epigenesis, Genetic, Intracellular Signaling Peptides and Proteins genetics, Muscle Proteins genetics, Podocytes pathology
- Abstract
Mounting evidence suggests that epigenetic modification is important in kidney disease pathogenesis. To determine whether epigenetic regulation is involved in HIV-induced kidney injury, we performed genome-wide methylation profiling and transcriptomic profiling of human primary podocytes infected with HIV-1. Comparison of DNA methylation and RNA sequencing profiles identified several genes that were hypomethylated with corresponding upregulated RNA expression in HIV-infected podocytes. Notably, we found only one hypermethylated gene with corresponding downregulated RNA expression, namely regulator of calcineurin 1 (RCAN1). Further, we found that RCAN1 RNA expression was suppressed in glomeruli in human diabetic nephropathy, IgA nephropathy, and lupus nephritis, and in mouse models of HIV-associated nephropathy and diabetic nephropathy. We confirmed that HIV infection or high glucose conditions suppressed RCAN1 expression in cultured podocytes. This suppression was alleviated upon pretreatment with DNA methyltransferase inhibitor 5-Aza-2'-deoxycytidine, suggesting that RCAN1 expression is epigenetically suppressed in the context of HIV infection and diabetic conditions. Mechanistically, increased expression of RCAN1 decreased HIV- or high glucose-induced nuclear factor of activated T cells (NFAT) transcriptional activity. Increased RCAN1 expression also stabilized actin cytoskeleton organization, consistent with the inhibition of the calcineurin pathway. In vivo, knockout of RCAN1 aggravated albuminuria and podocyte injury in mice with Adriamycin-induced nephropathy. Our findings suggest that epigenetic suppression of RCAN1 aggravates podocyte injury in the setting of HIV infection and diabetic nephropathy., (Copyright © 2018 International Society of Nephrology. All rights reserved.)
- Published
- 2018
- Full Text
- View/download PDF
5. Proteomics analysis of the non-muscle myosin heavy chain IIa-enriched actin-myosin complex reveals multiple functions within the podocyte.
- Author
-
Hays T, Ma'ayan A, Clark NR, Tan CM, Teixeira A, Teixeira A, Choi JW, Burdis N, Jung SY, Bajaj AO, O'Malley BW, He JC, Hyink DP, and Klotman PE
- Subjects
- Actins biosynthesis, Animals, Cytoskeletal Proteins biosynthesis, Cytoskeletal Proteins metabolism, Gene Expression Regulation, Humans, Membrane Proteins biosynthesis, Membrane Proteins metabolism, Mice, Molecular Motor Proteins biosynthesis, Multiprotein Complexes metabolism, Myosin Heavy Chains biosynthesis, Podocytes metabolism, Protein Interaction Maps, nef Gene Products, Human Immunodeficiency Virus metabolism, Actins metabolism, Molecular Motor Proteins metabolism, Myosin Heavy Chains metabolism, Proteomics
- Abstract
MYH9 encodes non-muscle myosin heavy chain IIA (NMMHCIIA), the predominant force-generating ATPase in non-muscle cells. Several lines of evidence implicate a role for MYH9 in podocytopathies. However, NMMHCIIA's function in podocytes remains unknown. To better understand this function, we performed immuno-precipitation followed by mass-spectrometry proteomics to identify proteins interacting with the NMMHCIIA-enriched actin-myosin complexes. Computational analyses revealed that these proteins belong to functional networks including regulators of cytoskeletal organization, metabolism and networks regulated by the HIV-1 gene nef. We further characterized the subcellular localization of NMMHCIIA within podocytes in vivo, and found it to be present within the podocyte major foot processes. Finally, we tested the effect of loss of MYH9 expression in podocytes in vitro, and found that it was necessary for cytoskeletal organization. Our results provide the first survey of NMMHCIIA-enriched actin-myosin-interacting proteins within the podocyte, demonstrating the important role of NMMHCIIA in organizing the elaborate cytoskeleton structure of podocytes. Our characterization of NMMHCIIA's functions goes beyond the podocyte, providing important insights into its general molecular role.
- Published
- 2014
- Full Text
- View/download PDF
6. Inhibition of Notch pathway attenuates the progression of human immunodeficiency virus-associated nephropathy.
- Author
-
Sharma M, Magenheimer LK, Home T, Tamano KN, Singhal PC, Hyink DP, Klotman PE, Vanden Heuvel GB, and Fields TA
- Subjects
- AIDS-Associated Nephropathy drug therapy, AIDS-Associated Nephropathy pathology, Amyloid Precursor Protein Secretases antagonists & inhibitors, Animals, Cell Dedifferentiation drug effects, Cell Dedifferentiation physiology, Cell Division drug effects, Cell Division physiology, Cell Line, Transformed, Dibenzazepines pharmacology, Dipeptides pharmacology, Disease Models, Animal, Disease Progression, Humans, Kidney drug effects, Kidney pathology, Kidney physiology, Ligands, Mice, Mice, Inbred Strains, Mice, Transgenic, Podocytes cytology, Podocytes drug effects, Proto-Oncogene Proteins antagonists & inhibitors, Receptor, Notch1 antagonists & inhibitors, Receptor, Notch4, Receptors, Notch antagonists & inhibitors, Signal Transduction drug effects, Up-Regulation drug effects, Up-Regulation physiology, AIDS-Associated Nephropathy metabolism, Podocytes metabolism, Proto-Oncogene Proteins metabolism, Receptor, Notch1 metabolism, Receptors, Notch metabolism, Signal Transduction physiology
- Abstract
The Notch pathway is an evolutionarily conserved signaling cascade that is critical in kidney development and has also been shown to play a pathogenetic role in a variety of kidney diseases. We have previously shown that the Notch signaling pathway is activated in human immunodeficiency virus-associated nephropathy (HIVAN) as well as in a rat model of the disease. In this study, we examined Notch signaling in the well established Tg26 mouse model of HIVAN. Notch signaling components were distinctly upregulated in the kidneys of these mice as well as in immortalized podocytes derived from these mice. Notch1 and Notch4 were upregulated in the Tg26 glomeruli, and Notch4 was also expressed in tubules. Notch ligands Jagged1, Jagged2, Delta-like1, and Delta-like 4 were all upregulated in the tubules of Tg26 mice, but glomeruli showed minimal expression of Notch ligands. To examine a potential pathogenetic role for Notch in HIVAN, Tg26 mice were treated with GSIXX, a gamma secretase inhibitor that blocks Notch signaling. Strikingly, GSIXX treatment resulted in significant improvement in both histological kidney injury scores and renal function. GSIXX-treated Tg26 mice also showed diminished podocyte proliferation and dedifferentiation, cellular hallmarks of the disease. Moreover, GSIXX blocked podocyte proliferation in vitro induced by HIV proteins Nef and Tat. These studies suggest that Notch signaling can promote HIVAN progression and that Notch inhibition may be a viable treatment strategy for HIVAN.
- Published
- 2013
- Full Text
- View/download PDF
7. Glomerular MYH9 expression is reduced by HIV-1.
- Author
-
Hays T, D'Agati VD, Garellek JA, Warren T, Trubin ME, Hyink DP, He JC, and Klotman PE
- Subjects
- AIDS-Associated Nephropathy metabolism, Animals, Disease Models, Animal, HIV Infections metabolism, HIV Infections physiopathology, Humans, Mice, Mice, Transgenic, Molecular Motor Proteins metabolism, Myosin Heavy Chains metabolism, Podocytes metabolism, Real-Time Polymerase Chain Reaction, AIDS-Associated Nephropathy physiopathology, HIV-1 metabolism, Nonmuscle Myosin Type IIA metabolism, Podocytes pathology
- Abstract
Background: The continuing disease burden of HIV-associated nephropathy (HIVAN) warrants better elucidation of its pathogenic mechanisms. Given that loss of MYH9 function causes a Mendelian renal disease, we hypothesized that renal expression of MYH9 is down-regulated by HIV-1 in HIVAN pathogenesis., Method and Results: Using immunofluorescence, we determined that glomerular expression of MYH9 was reduced in the kidneys of HIV-1 transgenic mice. We further determined that Myh9 expression was reduced in HIV-1 transgenic podocytes, statistically significantly at the protein level, and that MYH9 expression was significantly reduced at protein and message level in human podocytes transduced with HIV-1. In analyzing expression in human tissue, we confirmed that MYH9 is abundantly expressed in glomeruli, and podocytes specifically. Finally, we found that MYH9 expression was significantly reduced in human glomeruli in the setting of HIVAN., Conclusion: We conclude that the podocyte host response to HIV-1 includes down-regulation of MYH9 expression, and hypothesize that this down-regulation might play a role in the pathogenesis of HIVAN.
- Published
- 2012
- Full Text
- View/download PDF
8. Protein kinase-X interacts with Pin-1 and Polycystin-1 during mouse kidney development.
- Author
-
Li X, Hyink DP, Radbill B, Sudol M, Zhang H, Zheleznova NN, and Wilson PD
- Subjects
- Animals, Cell Adhesion genetics, Cell Movement genetics, Female, Mice, Morphogenesis genetics, NIMA-Interacting Peptidylprolyl Isomerase, Organ Culture Techniques, Pregnancy, Protein Serine-Threonine Kinases, Cyclic AMP-Dependent Protein Kinases metabolism, Kidney cytology, Kidney embryology, Peptidylprolyl Isomerase metabolism, Protein Kinases metabolism, TRPP Cation Channels metabolism
- Abstract
The regulation of epithelial branching morphogenesis by bone morphogenetic protein-7 depends, in part, on functionally defined cyclic adenosine monophosphate (cAMP)-dependent protein kinases. We previously identified protein kinase-X (PRKX), a cAMP-dependent kinase, as a regulator of epithelial morphogenesis during kidney development and found that it binds to and phosphorylates Polycystin-1. Overexpression of PRKX stimulates renal epithelial cell migration, tubulogenesis, ureteric bud branching, and glomerular induction in embryonic mouse kidney explants in organ cultures. Here we determined the physiological functions of endogenous PRKX. Knockdown by siRNA of PRKX gene expression in a human fetal collecting tubule (HFCT) cell line exceeded 70% and resulted in decreased cell migration and increased adhesion of the cells to a collagen I matrix. In embryonic mouse kidney explants, the same degree of knockdown decreased ureteric bud branching and glomerular induction. Because PRKX BAG-3 PIN-1 and MAGI-1 are all expressed in ureteric bud derivatives, we tested for interactions among them and found that PRKX binds to all three proteins through its WW domain as determined by TransSignal domain arrays, and it coimmunoprecipitated with Pin-1 in HFCT cell lysates. These studies suggest that Polycystin-1 and Pin-1 may mediate the function of PRKX in kidney development.
- Published
- 2009
- Full Text
- View/download PDF
9. Protein kinase X (PRKX) can rescue the effects of polycystic kidney disease-1 gene (PKD1) deficiency.
- Author
-
Li X, Burrow CR, Polgar K, Hyink DP, Gusella GL, and Wilson PD
- Subjects
- Animals, Cell Adhesion, Cell Line, Cell Movement, Cell Shape, Humans, Kidney metabolism, Mice, Organ Culture Techniques, Phosphorylation, Polycystic Kidney, Autosomal Dominant metabolism, Protein Binding, Protein Serine-Threonine Kinases genetics, RNA, Messenger genetics, Protein Serine-Threonine Kinases metabolism, TRPP Cation Channels metabolism
- Abstract
Autosomal dominant polycystic kidney disease (ADPKD) is a common, genetically determined developmental disorder of the kidney that is characterized by cystic expansion of renal tubules and is caused by truncating mutations and haplo-insufficiency of the PKD1 gene. Several defects in cAMP-mediated proliferation and ion secretion have been detected in ADPKD cyst-lining epithelia. Unlike the ubiquitous PKA, the cAMP-dependent CREB-kinase, Protein Kinase X (PRKX) is developmentally regulated, tissue restricted and induces renal epithelial cell migration, and tubulogenesis in vitro as well as branching morphogenesis of ureteric bud in developing kidneys. The possibility of functional interactions between PKD1-encoded polycystin-1 and PRKX was suggested by the renal co-distribution of PRKX and polycystin-1 and the binding and phosphorylation of the C-terminal of polycystin-1 by PRKX at S4166 in vitro. Early consequences of PKD1 mutation include increased tubule epithelial cell-matrix adhesion, decreased migration, reduced ureteric bud branching and aberrant renal tubule dilation. To determine whether PRKX might counteract the adverse effects of PKD1 mutation, human ADPKD epithelial cell lines were transfected with constitutively active PRKX and shown to rescue characteristic adhesion and migration defects. In addition, the co-injection of constitutively active PRKX with inhibitory pMyr-EGFP-PKD1 into the ureteric buds of mouse embryonic kidneys in organ culture resulted in restoration of normal branching morphogenesis without cystic tubular dilations. These results suggest that PRKX can restore normal function to PKD1-deficient kidneys and have implications for the development of preventative therapy for ADPKD.
- Published
- 2008
- Full Text
- View/download PDF
10. Inhibition of HER-2(neu/ErbB2) restores normal function and structure to polycystic kidney disease (PKD) epithelia.
- Author
-
Wilson SJ, Amsler K, Hyink DP, Li X, Lu W, Zhou J, Burrow CR, and Wilson PD
- Subjects
- Cell Movement, Cloning, Molecular, Epithelial Cells metabolism, Epithelial Cells pathology, ErbB Receptors metabolism, Humans, Immunoblotting, Polycystic Kidney Diseases metabolism, Polycystic Kidney Diseases pathology, Polycystic Kidney Diseases therapy, RNA, Small Interfering genetics, Receptor, ErbB-2 antagonists & inhibitors, Reference Values, Reverse Transcriptase Polymerase Chain Reaction, Polycystic Kidney Diseases genetics, Receptor, ErbB-2 genetics
- Abstract
Autosomal Dominant Polycystic Kidney Disease (ADPKD) is a very common lethal monogenetic disease with significant morbidities and a high likelihood of progression to renal failure for which there is no proven disease-specific therapy currently available for clinical use. Human ADPKD cystic epithelia have proliferative abnormalities mediated by EGFR over-expression and mispolarization leading autocrine response to EGF family ligands. We now show that apical localization of EGFR complexes in normal fetal and ADPKD epithelia is associated with heterodimerization of EGFR(HER-1) with HER-2(neu/ErbB2), while basal membrane localization in normal adult renal epithelia is associated with EGFR(HER-1) homodimers. Since ADPKD epithelial cells have reduced migratory function, this was used as a bioassay to evaluate the ability of compounds to rescue the aberrant human ADPKD phenotype. General tyrosine kinase inhibition by herbimycin and specific inhibition of HER-2(neu/ErbB2) by AG825 or pretreatment with ErbB2 siRNA reversed the migration defect of ADPKD epithelia. Selective inhibition of EGFR(HER-1) showed partial rescue. Increased ADPKD cell migration after inhibition of p38MAP kinase but not of PI3-kinase implicated p38MAPK downstream of HER-2(neu/ErbB2) stimulation. Daily administration of AG825 to PKD1 null heterozygous mice significantly inhibited the development of renal cysts. These studies implicate HER2(neu/ErbB2) as an effector of apical EGFR complex mispolarization and that its inhibition should be considered a candidate for clinical therapy of ADPKD.
- Published
- 2006
- Full Text
- View/download PDF
11. Image processing software for 3D light microscopy.
- Author
-
Clendenon JL, Byars JM, and Hyink DP
- Subjects
- Animals, Image Processing, Computer-Assisted instrumentation, Image Processing, Computer-Assisted methods, Imaging, Three-Dimensional, Microscopy, Software
- Abstract
Advances in microscopy now enable researchers to easily acquire multi-channel three-dimensional (3D) images and 3D time series (4D). However, processing, analyzing, and displaying this data can often be difficult and time- consuming. We discuss some of the software tools and techniques that are available to accomplish these tasks., (Copyright 2006 S. Karger AG, Basel.)
- Published
- 2006
- Full Text
- View/download PDF
12. Protein kinase X activates ureteric bud branching morphogenesis in developing mouse metanephric kidney.
- Author
-
Li X, Hyink DP, Polgar K, Gusella GL, Wilson PD, and Burrow CR
- Subjects
- Animals, Cells, Cultured, Female, Gene Expression Regulation, Developmental, Immunohistochemistry, In Situ Hybridization, In Vitro Techniques, Kidney pathology, Mice, Organogenesis genetics, RNA, Messenger analysis, Sensitivity and Specificity, Ureter pathology, Kidney embryology, Organogenesis physiology, Protein Kinases genetics, Ureter embryology
- Abstract
The human protein kinase X (PRKX) gene was identified previously as a cAMP-dependent serine/threonine kinase that is aberrantly expressed in autosomal dominant polycystic disease kidneys and normally expressed in fetal kidneys. The PRKX kinase belongs to a serine/threonine kinase family that is phylogenetically and functionally distinct from classical protein kinase A kinases. Expression of PRKX activates cAMP-dependent renal epithelial cell migration and tubular morphogenesis in cell culture, suggesting that it might regulate branching growth of the collecting duct system in the fetal kidney. With the use of a mouse embryonic kidney organ culture system that recapitulates early kidney development in vitro, it is demonstrated that lentiviral vector-driven expression of a constitutively active, cAMP-independent PRKX in the ureteric bud epithelium stimulates branching morphogenesis and results in a 2.5-fold increase in glomerular number. These results suggest that PRKX stimulates epithelial branching morphogenesis by activating cell migration and support a role for this kinase in the regulation of nephrogenesis and of collecting system development in the fetal kidney.
- Published
- 2005
- Full Text
- View/download PDF
13. Disruption of polycystin-1 function interferes with branching morphogenesis of the ureteric bud in developing mouse kidneys.
- Author
-
Polgar K, Burrow CR, Hyink DP, Fernandez H, Thornton K, Li X, Gusella GL, and Wilson PD
- Subjects
- Animals, Cell Line, Gene Expression Regulation, Developmental, Genetic Vectors, Humans, Mice, Morphogenesis, Polycystic Kidney, Autosomal Dominant embryology, Polycystic Kidney, Autosomal Dominant genetics, Proteins genetics, Proteins physiology, Recombinant Fusion Proteins genetics, TRPP Cation Channels, Kidney embryology, Proteins antagonists & inhibitors, Ureter embryology
- Abstract
The polycystic kidney disease (PKD1) gene-encoded protein, polycystin-1, is developmentally regulated, with highest expression levels seen in normal developing kidneys, where it is distributed in a punctate pattern at the basal surface of ureteric bud epithelia. Overexpression in ureteric epithelial cell membranes of an inhibitory pMyr-GFP-PKD1 fusion protein via a retroviral (VVC) delivery system and microinjection into the ureteric bud lumen of embryonic day 11 mouse metanephric kidneys resulted in disrupted branching morphogenesis. Using confocal quantitative analysis, significant reductions were measured in the numbers of ureteric bud branch points and tips, as well as in the total ureteric bud length, volume and area, while significant increases were seen as dilations of the terminal branches, where significant increases in outer diameter and volumes were measured. Microinjection of an activating 5TM-GFP-PKD1 fusion protein had an opposite effect and showed significant increases in ureteric bud length and area. These are the first studies to experimentally manipulate polycystin-1 expression by transduction in the embryonic mouse kidney and suggest that polycystin-1 plays a critical role in the regulation of epithelial morphogenesis during renal development.
- Published
- 2005
- Full Text
- View/download PDF
14. Midkine promotes selective expansion of the nephrogenic mesenchyme during kidney organogenesis.
- Author
-
Qiu L, Hyink DP, Gans WH, Amsler K, Wilson PD, and Burrow CR
- Abstract
During kidney development, the growth and development of the stromal and nephrogenic mesenchyme cell populations and the ureteric bud epithelium is tightly coupled through intricate reciprocal signaling mechanisms between these three tissue compartments. Midkine, a target gene activated by retinoid signaling in the metanephros, encodes a secreted polypeptide with mitogenic and anti-apoptotic activities in a wide variety of cell types. Using immmunohistochemical methods we demonstrated that Midkine is found in the uninduced mesenchyme at the earliest stages of metanephric kidney development and only subsequently concentrated in the ureteric bud epithelium and basement membrane. The biological effects of purified recombinant Midkine were analyzed in metanephric organ culture experiments carried out in serum-free defined media. These studies revealed that Midkine selectively promoted the overgrowth of the Pax-2 and N-CAM positive nephrogenic mesenchymal cells, failed to stimulate expansion of the stromal compartment and suppressed branching morphogenesis of the ureteric bud. Midkine suppressed apoptosis and stimulated cellular proliferation of the nephrogenic mesenchymal cells, and was capable of maintaining the viability of isolated mesenchymes cultured in the absence of the ureteric bud. These results suggest that Midkine may regulate the balance of epithelial and stromal progenitor cell populations of the metanephric mesenchyme during renal organogenesis.
- Published
- 2004
- Full Text
- View/download PDF
15. The receptor tyrosine kinase regulator Sprouty1 is a target of the tumor suppressor WT1 and important for kidney development.
- Author
-
Gross I, Morrison DJ, Hyink DP, Georgas K, English MA, Mericskay M, Hosono S, Sassoon D, Wilson PD, Little M, and Licht JD
- Subjects
- Adaptor Proteins, Signal Transducing, Animals, Binding Sites, Blotting, Northern, Chromatin metabolism, Cloning, Molecular, Drosophila, Genes, Reporter, Immunohistochemistry, Kidney metabolism, Kidney Glomerulus embryology, Mice, Models, Genetic, NIH 3T3 Cells, Oligonucleotides chemistry, Oligonucleotides, Antisense pharmacology, Precipitin Tests, Promoter Regions, Genetic, Reverse Transcriptase Polymerase Chain Reaction, Tissue Distribution, Transcription, Genetic, Transfection, Up-Regulation, Gene Expression Regulation, Developmental, Kidney embryology, Membrane Proteins metabolism, Membrane Proteins physiology, Phosphoproteins metabolism, Phosphoproteins physiology, WT1 Proteins metabolism
- Abstract
WT1 encodes a transcription factor involved in kidney development and tumorigenesis. Using representational difference analysis, we identified a new set of WT1 targets, including a homologue of the Drosophila receptor tyrosine kinase regulator, sprouty. Sprouty1 was up-regulated in cell lines expressing wild-type but not mutant WT1. WT1 bound to the endogenous sprouty1 promoter in vivo and directly regulated sprouty1 through an early growth response gene-1 binding site. Expression of Sprouty1 and WT1 overlapped in the developing metanephric mesenchyme, and Sprouty1, like WT1, plays a key role in the early steps of glomerulus formation. Disruption of Sprouty1 expression in embryonic kidney explants by antisense oligonucleotides reduced condensation of the metanephric mesenchyme, leading to a decreased number of glomeruli. In addition, sprouty1 was expressed in the ureteric tree and antisense-treated ureteric trees had cystic lumens. Therefore, sprouty1 represents a physiologically relevant target gene of WT1 during kidney development.
- Published
- 2003
- Full Text
- View/download PDF
16. Expression of the urate transporter/channel is developmentally regulated in human kidneys.
- Author
-
Hyink DP, Rappoport JZ, Wilson PD, and Abramson RG
- Subjects
- Adolescent, Adult, Amino Acid Sequence, Animals, Blotting, Western, Carrier Proteins chemistry, Carrier Proteins metabolism, Cell Membrane metabolism, Child, Child, Preschool, Cytoplasm metabolism, Female, Gestational Age, Humans, Immunohistochemistry, Infant, Infant, Newborn, Kidney embryology, Kidney Cortex chemistry, Kidney Cortex metabolism, Kidney Tubules, Proximal metabolism, Kidney Tubules, Proximal ultrastructure, Microvilli enzymology, Middle Aged, Molecular Sequence Data, Nephrons chemistry, Nephrons metabolism, Oocytes metabolism, Oocytes ultrastructure, Organic Cation Transport Proteins, Rats, Recombinant Proteins metabolism, Transfection, Xenopus laevis, Carrier Proteins genetics, Gene Expression Regulation, Developmental, Kidney growth & development, Kidney metabolism, Organic Anion Transporters
- Abstract
Recombinant protein prepared from cDNA cloned from rat kidney and its human homolog function as urate transporter/channels in lipid bilayers. Using the antibody (anti-uricase) that detected the rat cDNA clone, we now demonstrate that normal human kidneys contain an immunoreactive protein of identical size to that in rat kidney (36-37 kDa), presumably the human urate transporter/channel (hUAT). The amount of hUAT in kidney homogenates increases progressively from 13 wk of gestation to the early postnatal period. During gestation, hUAT expression is confined to the cytoplasm of proximal tubules of Stage III and/or IV nephrons. However, at 1 yr of age hUAT is primarily located subapically and within brush borders of proximal tubules. Xenopus laevis oocytes and differentiated A6 cells injected with cRNA and transfected with cDNA of hUAT, respectively, demonstrated a similar pattern: hUAT is not detected in oocytes but is abundantly expressed in cytoplasm and plasma membranes of A6 cells. These data imply that different developmental factors regulate the initiation of cytoplasmic hUAT expression and subsequent insertion into human proximal tubule brush-border membranes.
- Published
- 2001
- Full Text
- View/download PDF
17. WT1 expression induces features of renal epithelial differentiation in mesenchymal fibroblasts.
- Author
-
Hosono S, Luo X, Hyink DP, Schnapp LM, Wilson PD, Burrow CR, Reddy JC, Atweh GF, and Licht JD
- Subjects
- 3T3 Cells, Animals, Cell Line, Transformed, Epithelial Cells cytology, Epithelial Cells metabolism, Fibroblasts cytology, Fibroblasts metabolism, Insulin-Like Growth Factor II genetics, Insulin-Like Growth Factor II metabolism, Integrins genetics, Integrins metabolism, Kidney cytology, Mesoderm cytology, Mice, Wilms Tumor pathology, Cell Differentiation, Genes, Wilms Tumor, Integrin alpha Chains, Kidney metabolism, Mesoderm metabolism, Up-Regulation
- Abstract
The WT1 tumor suppressor gene, implicated in hereditofamilial and sporadic Wilms' tumor, is required for normal renal development and is up-regulated during the mesenchymal-epithelial transition. NIH3T3 fibroblasts overexpressing WT1 were less proliferative, larger in size and more firmly attached to tissue culture plastic, suggesting an alteration of their state of differentiation. These cells were studied in vivo by subcutaneous injection into nude mice. The resulting tumors exhibited epithelioid histopathology and formed desmosome-like structures. Molecular analyses of these WT1 expressing fibroblasts grown in culture and in nude mice revealed significant alterations in the expression of many kidney epithelial markers. These studies indicate that WT1 expression can initiate features of a program of epithelial differentiation consistent with a prominent role for WT1 in the mesenchymal epithelial transition that occurs during renal development. Through this work we identified a number of novel target genes for the WT1 transcription factor, including uvomorulin, integrin alpha8 and perlecan, and suggest that WTI may activate the IGF-II gene, also implicated in the development of Wilms' tumor.
- Published
- 1999
- Full Text
- View/download PDF
18. Origins and formation of microvasculature in the developing kidney.
- Author
-
Abrahamson DR, Robert B, Hyink DP, St John PL, and Daniel TO
- Subjects
- Animals, Capillaries growth & development, Humans, Microcirculation physiology, Renal Circulation physiology, Kidney blood supply, Kidney embryology
- Abstract
Regulation of microvessel assembly in the developing kidney is not known and may occur through vasculogenic, angiogenic, or both processes. To examine this question, we grafted rat and mice embryonic (E) day 12 (E12) kidneys, which have only a rudimentary vasculature, into anterior eye chambers of mouse and rat hosts. Species-specific, monoclonal anti-basement membrane antibodies showed that glomerular basement membranes, mesangial matrices, and microvessel basement membranes were always derived from the graft. When wild-type E12 mouse kidneys were grafted into anterior chambers of ROSA26 mice, in which the beta-galactosidase transgene is expressed ubiquitously, glomerular and microvascular endothelial cells stemmed from the graft, even after maintenance of kidneys in organ culture for 6 days before grafting. Immunolabeling with antibodies against the vascular endothelial growth factor (VEGF) receptor, Flk1, the EphB1 receptor, and its ligand, ephrin-B1, labeled discrete mesenchymal cells in embryonic and newborn kidney cortex, as well as developing microvessel and glomerular endothelium. In adult kidneys, Flk1 labeled glomeruli weakly, other vascular structures were unlabeled. When wild-type E12 kidneys were grafted under renal capsules of adult ROSA26 hosts, endothelium developing within the graft again came from the implanted kidney. In contrast, when E12 kidneys were grafted into renal cortices of newborns, glomeruli within grafts now contained host-derived endothelium. Similarly, when ROSA26 E12 kidneys were implanted into newborn wild-type hosts, chimeric vessels containing graft- and host-derived endothelium were seen in nearby host tissue. Our results indicate that cells capable of forming the entire microvascular tree of grafted metanephroi are already present in the E12 kidney. We hypothesize that Flk1/VEGF and EphB1/ephrin-B1 mediate renal endothelial mitosis-motility and cell guidance-aggregation behavior, respectively.
- Published
- 1998
- Full Text
- View/download PDF
19. Evidence that embryonic kidney cells expressing flk-1 are intrinsic, vasculogenic angioblasts.
- Author
-
Robert B, St John PL, Hyink DP, and Abrahamson DR
- Subjects
- Animals, Anterior Chamber surgery, Embryo, Mammalian cytology, Embryo, Mammalian metabolism, Embryo, Mammalian physiology, Fetal Tissue Transplantation, Fluorescent Antibody Technique, Kidney cytology, Kidney metabolism, Kidney Cortex surgery, Lectins, Mice, Mice, Inbred C57BL, Mice, Transgenic, Organ Culture Techniques, Receptors, Vascular Endothelial Growth Factor, Kidney embryology, Neovascularization, Physiologic, Plant Lectins, Receptor Protein-Tyrosine Kinases metabolism, Receptors, Growth Factor metabolism
- Abstract
Renal glomerular capillary tufts have been believed to arise from angiogenic ingrowth of extrinsic vessels. We found, however, that when embryonic day 12 (E12) mouse kidneys were maintained in culture for 6 days and then grafted into anterior eye chambers of adult transgenic ROSA26 host mice (which carry the beta-galactosidase transgene), glomerular endothelial cells within the grafts were predominantly of intrinsic, kidney origin. To identify potential endothelial precursors, we immunolabled kidneys with antibodies against the vascular endothelial growth factor receptor, flk-1. Numerous discrete cells expressing flk-1 were scattered throughout the nephrogenic mesenchyme of both E12 and newborn kidneys, and with development these cells became concentrated in microvessels, glomerular vascular clefts, and glomerular tufts. In adults, flk-1 was weakly expressed in glomeruli but absent elsewhere. To examine abilities of flk-1-positive cells to establish glomeruli, E12 kidneys were grafted into kidney cortices of adult and newborn ROSA26 hosts. Grafts into adults resulted in few glomeruli containing host-derived endothelium, whereas a majority of glomeruli grafted into newborns contained host cells. Cells of graft origin were found in vessels forming in renal cortices of newborn hosts, but not in adults. Our findings indicate that embryonic kidney cells expressing flk-1 are angioblasts that create microvessels and glomeruli by vasculogenesis.
- Published
- 1996
- Full Text
- View/download PDF
20. Endogenous origin of glomerular endothelial and mesangial cells in grafts of embryonic kidneys.
- Author
-
Hyink DP, Tucker DC, St John PL, Leardkamolkarn V, Accavitti MA, Abrass CK, and Abrahamson DR
- Subjects
- Animals, Antibodies, Monoclonal, Antibody Specificity, Cell Line, Extracellular Matrix physiology, Lectins, Mice, Mice, Inbred Strains, Rats, Rats, Sprague-Dawley, Renal Circulation, Species Specificity, Transplantation, Heterologous, Urothelium cytology, Fetal Tissue Transplantation, Glomerular Mesangium cytology, Kidney Glomerulus cytology, Kidney Transplantation, Plant Lectins
- Abstract
To address origins of glomerular endothelial and mesangial cells in embryonic mammalian kidneys, we established interspecies grafts between rats and mice, in which fetal kidneys were implanted into the anterior eye chamber of adult hosts. After 5-7 days, hosts bearing grafts received intravenous injections with species-specific monoclonal antibodies (MAbs) to matrix components. In all cases, glomerular basement membranes and mesangial matrices labeled solely for donor-derived matrix. Additionally, microvessel extracellular matrices within grafts were usually of donor origin. To examine directly the origin of glomerular endothelial and mesangial cells, we grafted embryonic gestational days 11-12 (E11-12) kidneys from normal mice into anterior eye chambers of host reverse-orientation splice acceptor 26 mice, which are transgenic animals that express beta-galactosidase in every cell. When grafts were developed for beta-galactosidase activity, host cells were seen in peripheral vessels, but the majority of glomerular endothelial cells were of donor, not host, origin. Where host-derived-endothelial cells were found in glomeruli, donor endothelial cells were present as well. Mesangial cells were always of donor origin. When E11 mouse kidneys were labeled with the endothelial cell-specific Bandeiraea simplicifolia isolectin B4, we determined that endothelial cells are present from the inception of metanephrogenesis. Together, the evidence shows that cells of endogenous kidney origin were almost entirely responsible for development of the glomerular microvasculature in oculo. External vessels from the host, although important for graft maintenance, were not major contributors to the glomerulus.
- Published
- 1996
- Full Text
- View/download PDF
21. Origin of the glomerular vasculature in the developing kidney.
- Author
-
Hyink DP and Abrahamson DR
- Subjects
- Animals, Endothelium, Vascular ultrastructure, Growth Substances physiology, Kidney Glomerulus embryology, Kidney Glomerulus ultrastructure, Organ Culture Techniques, Endothelium, Vascular embryology, Kidney Glomerulus blood supply
- Abstract
Mechanisms regulating the establishment of the glomerular endothelium and mesangium during glomerulogenesis are not understood. In this article, we discuss two different blood vessel growth processes: vasculogenesis and angiogenesis, with particular attention on how these processes might operate in the developing kidney. Results from metanephric organ cultures and interspecies grafts are also interpreted with an emphasis on generation of the renal microvasculature. Among the several growth factors identified in the metanephros, many (eg, fibroblast growth factor [FGF], vascular endothelial growth factor [VEGF], platelet-derived growth factor [PDGF], epidermal growth factor/transforming growth factor-alpha [EGF/TGF-alpha], hepatocyte growth factor/scatter factor [HGF/SF], and insulinlike growth factor [IGF]) have angiogenic properties, and these are discussed in relation to formation of the glomerulus. How extracellular matrices and proteases might be involved in vascularization are also considered.
- Published
- 1995
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.