Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 30
Filter
Add more filters










Publication year range
1.
Sci Rep ; 11(1): 22434, 2021 11 17.
Article in English | MEDLINE | ID: mdl-34789782

ABSTRACT

The kidney is a complex organ composed of more than 30 terminally differentiated cell types that all are required to perform its numerous homeostatic functions. Defects in kidney development are a significant cause of chronic kidney disease in children, which can lead to kidney failure that can only be treated by transplant or dialysis. A better understanding of molecular mechanisms that drive kidney development is important for designing strategies to enhance renal repair and regeneration. In this study, we profiled gene expression in the developing mouse kidney at embryonic day 14.5 at single-cell resolution. Consistent with previous studies, clusters with distinct transcriptional signatures clearly identify major compartments and cell types of the developing kidney. Cell cycle activity distinguishes between the "primed" and "self-renewing" sub-populations of nephron progenitors, with increased expression of the cell cycle-related genes Birc5, Cdca3, Smc2 and Smc4 in "primed" nephron progenitors. In addition, augmented expression of cell cycle related genes Birc5, Cks2, Ccnb1, Ccnd1 and Tuba1a/b was detected in immature distal tubules, suggesting cell cycle regulation may be required for early events of nephron patterning and tubular fusion between the distal nephron and collecting duct epithelia.


Subject(s)
Cell Cycle/genetics , Cell Differentiation/genetics , Gene Expression Regulation, Developmental , Kidney Tubules, Distal/embryology , Sequence Analysis, RNA/methods , Single-Cell Analysis/methods , Transcriptome , Animals , Female , Mice , Pregnancy
2.
Croat Med J ; 60(6): 521-531, 2019 Dec 31.
Article in English | MEDLINE | ID: mdl-31894918

ABSTRACT

AIM: To explore the spatial and temporal expression patterns of DAB1 and Reelin in the developing and postnatal healthy human kidneys as potential determinants of kidney development. METHODS: Paraffin-embedded fetal kidney tissue between the 13/14th and 38th developmental weeks (dw) and postnatal tissue at 1.5 and 7 years were stained with DAB1 and Reelin antibodies by double immunofluorescence. RESULTS: During the fetal kidney development and postnatal period, DAB1 and Reelin showed specific spatial expression pattern and diverse fluorescence intensity. During the fetal period, DAB1 was strongly expressed in the distal convoluted tubules (DCT), with strong reactivity, and diversely in the proximal convoluted tubules (PCT) and glomeruli. In the postnatal period, DAB1 expression decreased. The strongest Reelin expression in early fetal stages was observed in the PCT. In the postnatal period, Reelin expression decreased dramatically in all observed structures. These two markers were colocalized during early developmental stages, mostly in PCT, DCT, and podocytes. CONCLUSION: The appearance of DAB1 and Reelin during fetal kidney development confirms their potential significant role in the formation of kidney structure or function. High DAB1 expression in the DCT implies its regulatory role in tubular formation or function maintenance during development. Reelin was highly expressed in human kidneys at early fetal stages, mostly in the PCT, while at later fetal stages and postnatal period its expression decreased.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Cell Adhesion Molecules, Neuronal/metabolism , Extracellular Matrix Proteins/metabolism , Kidney/embryology , Kidney/growth & development , Nerve Tissue Proteins/metabolism , Serine Endopeptidases/metabolism , Child , Fetal Development , Gestational Age , Humans , Infant , Kidney/metabolism , Kidney Tubules, Distal/embryology , Kidney Tubules, Distal/metabolism , Kidney Tubules, Proximal/embryology , Kidney Tubules, Proximal/metabolism , Podocytes/metabolism , Reelin Protein
3.
Elife ; 72018 08 10.
Article in English | MEDLINE | ID: mdl-30095068

ABSTRACT

Wingless/Wnts are signalling molecules, traditionally considered to pattern tissues as long-range morphogens. However, more recently the spread of Wingless was shown to be dispensable in diverse developmental contexts in Drosophila and vertebrates. Here we demonstrate that release and spread of Wingless is required to pattern the proximo-distal (P-D) axis of Drosophila Malpighian tubules. Wingless signalling, emanating from the midgut, directly activates odd skipped expression several cells distant in the proximal tubule. Replacing Wingless with a membrane-tethered version that is unable to diffuse from the Wingless producing cells results in aberrant patterning of the Malpighian tubule P-D axis and development of short, deformed ureters. This work directly demonstrates a patterning role for a released Wingless signal. As well as extending our understanding about the functional modes by which Wnts shape animal development, we anticipate this mechanism to be relevant to patterning epithelial tubes in other organs, such as the vertebrate kidney.


Subject(s)
Body Patterning , Drosophila Proteins/metabolism , Drosophila melanogaster/physiology , Gene Expression Regulation, Developmental , Kidney Tubules, Distal/physiology , Kidney Tubules, Proximal/physiology , Wnt1 Protein/metabolism , Animals , Drosophila Proteins/genetics , Drosophila melanogaster/embryology , Embryo, Nonmammalian/cytology , Embryo, Nonmammalian/physiology , Kidney Tubules, Distal/embryology , Kidney Tubules, Proximal/embryology , Morphogenesis , Wnt Signaling Pathway , Wnt1 Protein/genetics
4.
Angiogenesis ; 21(3): 617-634, 2018 08.
Article in English | MEDLINE | ID: mdl-29627966

ABSTRACT

The kidney vasculature facilitates the excretion of wastes, the dissemination of hormones, and the regulation of blood chemistry. To carry out these diverse functions, the vasculature is regionalized within the kidney and along the nephron. However, when and how endothelial regionalization occurs remains unknown. Here, we examine the developing kidney vasculature to assess its 3-dimensional structure and transcriptional heterogeneity. First, we observe that endothelial cells (ECs) grow coordinately with the kidney bud as early as E10.5, and begin to show signs of specification by E13.5 when the first arteries can be identified. We then focus on how ECs pattern and remodel with respect to the developing nephron and collecting duct epithelia. ECs circumscribe nephron progenitor populations at the distal tips of the ureteric bud (UB) tree and form stereotyped cruciform structures around each tip. Beginning at the renal vesicle (RV) stage, ECs form a continuous plexus around developing nephrons. The endothelial plexus envelops and elaborates with the maturing nephron, becoming preferentially enriched along the early distal tubule. Lastly, we perform transcriptional and immunofluorescent screens to characterize spatiotemporal heterogeneity in the kidney vasculature and identify novel regionally enriched genes. A better understanding of development of the kidney vasculature will help instruct engineering of properly vascularized ex vivo kidneys and evaluate diseased kidneys.


Subject(s)
Embryo, Mammalian/embryology , Endothelial Cells/metabolism , Kidney Tubules, Distal/embryology , Organogenesis/physiology , Renal Artery/embryology , Renal Veins , Animals , Embryo, Mammalian/cytology , Endothelial Cells/cytology , Fetal Stem Cells/metabolism , Fluorescent Antibody Technique/methods , Kidney Tubules, Distal/cytology , Mice , Renal Artery/cytology , Renal Veins/growth & development , Renal Veins/metabolism , Transcription, Genetic/physiology , Urethra/cytology , Urethra/embryology
5.
Kidney Int ; 87(6): 1125-40, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25651362

ABSTRACT

MicroRNAs, activated by the enzyme Dicer1, control post-transcriptional gene expression. Dicer1 has important roles in the epithelium during nephrogenesis, but its function in stromal cells during kidney development is unknown. To study this, we inactivated Dicer1 in renal stromal cells. This resulted in hypoplastic kidneys, abnormal differentiation of the nephron tubule and vasculature, and perinatal mortality. In mutant kidneys, genes involved in stromal cell migration and activation were suppressed as were those involved in epithelial and endothelial differentiation and maturation. Consistently, polarity of the proximal tubule was incorrect, distal tubule differentiation was diminished, and elongation of Henle's loop attenuated resulting in lack of inner medulla and papilla in stroma-specific Dicer1 mutants. Glomerular maturation and capillary loop formation were abnormal, whereas peritubular capillaries, with enhanced branching and increased diameter, formed later. In Dicer1-null renal stromal cells, expression of factors associated with migration, proliferation, and morphogenic functions including α-smooth muscle actin, integrin-α8, -ß1, and the WNT pathway transcriptional regulator LEF1 were reduced. Dicer1 mutation in stroma led to loss of expression of distinct microRNAs. Of these, miR-214, -199a-5p, and -199a-3p regulate stromal cell functions ex vivo, including WNT pathway activation, migration, and proliferation. Thus, Dicer1 activity in the renal stromal compartment regulates critical stromal cell functions that, in turn, regulate differentiation of the nephron and vasculature during nephrogenesis.


Subject(s)
Cell Differentiation/genetics , DEAD-box RNA Helicases/physiology , Neovascularization, Physiologic/genetics , Nephrons/embryology , Ribonuclease III/physiology , Actins/metabolism , Animals , Capillaries/embryology , Cell Movement/genetics , Cell Proliferation/genetics , DEAD-box RNA Helicases/genetics , DEAD-box RNA Helicases/metabolism , Female , Gene Expression , Integrin alpha Chains/metabolism , Kidney Glomerulus/blood supply , Kidney Glomerulus/cytology , Kidney Glomerulus/embryology , Kidney Tubules/blood supply , Kidney Tubules/cytology , Kidney Tubules/embryology , Kidney Tubules, Distal/blood supply , Kidney Tubules, Distal/cytology , Kidney Tubules, Distal/embryology , Kidney Tubules, Proximal/blood supply , Kidney Tubules, Proximal/cytology , Kidney Tubules, Proximal/embryology , Loop of Henle/blood supply , Loop of Henle/cytology , Loop of Henle/embryology , Mice , MicroRNAs/genetics , Nephrons/abnormalities , Nephrons/cytology , Organogenesis/genetics , Podocytes/physiology , Ribonuclease III/genetics , Ribonuclease III/metabolism , Stromal Cells/physiology , Transcriptome , Ureter/abnormalities , Wnt Signaling Pathway/genetics
6.
J Am Soc Nephrol ; 23(10): 1682-90, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22904347

ABSTRACT

Formation of a functional renal network requires the interconnection of two epithelial tubes: the nephron, which arises from kidney mesenchyme, and the collecting system, which originates from the branching ureteric epithelium. How this connection occurs, however, is incompletely understood. Here, we used high-resolution image analysis in conjunction with genetic labeling of epithelia to visualize and characterize this process. Although the focal absence of basal lamina from renal vesicle stages ensures that both epithelial networks are closely apposed, we found that a patent luminal interconnection is not established until S-shaped body stages. Precursor cells of the distal nephron in the interconnection zone exhibit a characteristic morphology consisting of ill-defined epithelial junctional complexes but without expression of mesenchymal markers such as vimentin and Snai2. Live-cell imaging revealed that before luminal interconnection, distal cells break into the lumen of the collecting duct epithelium, suggesting that an invasive behavior is a key step in the interconnection process. Furthermore, loss of distal cell identity, which we induced by activating the Notch pathway, prevented luminal interconnection. Taken together, these data support a model in which establishing the distal identity of nephron precursor cells closest to the nascent collecting duct epithelium leads to an active cell invasion, which in turn contributes to a patent tubular interconnection between the nephron and collecting duct epithelia.


Subject(s)
Kidney/embryology , Animals , Cell Movement , Embryonic Stem Cells/cytology , Embryonic Stem Cells/metabolism , Female , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Imaging, Three-Dimensional , Kidney/metabolism , Kidney Tubules, Distal/embryology , Kidney Tubules, Distal/metabolism , Male , Mice , Mice, Transgenic , Microscopy, Confocal , Nephrons/embryology , Nephrons/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism , Zonula Occludens-1 Protein/metabolism
7.
Histochem Cell Biol ; 134(4): 355-69, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20853169

ABSTRACT

The kidney has an intrinsic ability to repair itself when injured. Epithelial cells of distal tubules may participate in regeneration. Stem cell marker, TRA-1-60 is linked to pluripotency in human embryonic stem cells and is lost upon differentiation. TRA-1-60 expression was mapped and quantified in serial sections of human foetal, adult and diseased kidneys. In 8- to 10-week human foetal kidney, the epitope was abundantly expressed on ureteric bud and structures derived therefrom including collecting duct epithelium. In adult kidney inner medulla/papilla, comparisons with reactivity to epithelial membrane antigen, aquaporin-2 and Tamm-Horsfall protein, confirmed extensive expression of TRA-1-60 in cells lining collecting ducts and thin limb of the loop of Henle, which may be significant since the papillae were proposed to harbour slow cycling cells involved in kidney homeostasis and repair. In the outer medulla and cortex there was rare, sporadic expression in tubular cells of the collecting ducts and nephron, with positive cells confined to the thin limb and thick ascending limb and distal convoluted tubules. Remarkably, in cortex displaying tubulo-interstitial injury, there was a dramatic increase in number of TRA-1-60 expressing individual cells and in small groups of cells in distal tubules. Dual staining showed that TRA-1-60 positive cells co-expressed Pax-2 and Ki-67, markers of tubular regeneration. Given the localization in foetal kidney and the distribution patterns in adults, it is tempting to speculate that TRA-1-60 may identify a population of cells contributing to repair of distal tubules in adult kidney.


Subject(s)
Antigens, Surface/metabolism , Kidney/embryology , Kidney/metabolism , Nephritis, Interstitial/metabolism , Proteoglycans/metabolism , Stem Cells/metabolism , Adult , Aged , Biomarkers/metabolism , Female , Fetus/metabolism , Fluorescent Antibody Technique , Gene Expression Regulation, Developmental , Humans , Immunohistochemistry , Ki-67 Antigen/metabolism , Kidney Diseases/metabolism , Kidney Diseases/pathology , Kidney Medulla/embryology , Kidney Medulla/metabolism , Kidney Tubules, Distal/embryology , Kidney Tubules, Distal/metabolism , Male , Middle Aged , Nephritis, Interstitial/pathology , Nephrons/embryology , Nephrons/metabolism , PAX2 Transcription Factor/metabolism , Pluripotent Stem Cells , Regeneration , Up-Regulation
8.
Rev Med Chir Soc Med Nat Iasi ; 112(1): 203-7, 2008.
Article in Romanian | MEDLINE | ID: mdl-18677927

ABSTRACT

UNLABELLED: We have looked for electron microscopy aspects tissue fragments of human nephrons harvested from fetuses of 7,5-19 weeks to show ultrastructure aspects of the nephron during development. MATERIAL AND METHODS: We have used 9 fragments from human embryos; Four cases were aged of 7,5, 8, 12 and 17 weeks. The other 5 cases were between 18 and 19 weeks. Tissue fragments were fixed in glutaraldehyde at 4 degrees C and processed by the classic technique. RESULTS: The electron microscopy study shows the evolutive steps of the human nephron (stages I-IV), stages met for the 12-19 weeks embryo. For the cases under this age the meta-nephrogenic blastema was dominant. In the IVth development stage, the cells in the proximal tubule show a higher development degree than the distal segments. CONCLUSION: These observations indicate that during early development the proximal segment is better developed and probably more functional than the distal segments.


Subject(s)
Microscopy, Electron , Nephrons/embryology , Nephrons/ultrastructure , Fetal Development , Fetus , Gestational Age , Humans , Kidney Tubules, Distal/embryology , Kidney Tubules, Distal/ultrastructure , Kidney Tubules, Proximal/embryology , Kidney Tubules, Proximal/ultrastructure
9.
Am J Physiol Regul Integr Comp Physiol ; 293(2): R759-65, 2007 Aug.
Article in English | MEDLINE | ID: mdl-17567711

ABSTRACT

Low birth weight humans and rats exposed to a low-protein diet in utero have reduced bone mineral content. Renal calcium loss during the period of rapid skeletal growth is associated with bone loss. Because young rats exposed to low protein display altered renal function, we tested the hypothesis that renal calcium excretion is perturbed in this model. Pregnant Wistar rats were fed isocalorific diets containing either 18% (control) or 9% (low) protein throughout gestation. Using standard renal clearance techniques, Western blotting for renal calcium transport proteins, and assays for Na(+)-K(+)-ATPase activity and serum calcitropic hormones, we characterized calcium handling in 4-wk-old male offspring. Histomorphometric analyses of femurs revealed a reduction in trabecular bone mass in low-protein rats. Renal calcium (control vs. low protein: 10.4 +/- 2.1 vs. 27.6 +/- 4.5 nmol x min(-1) x 100 g body wt(-1); P < 0.01) and sodium excretion were increased, but glomerular filtration rate was reduced in low-protein animals. Total plasma calcium was reduced in low-protein rats (P < 0.01), but ionized calcium, serum calcitropic hormone concentrations, and total body calcium did not differ. There was no significant change in plasma membrane Ca(2+)-ATPase pump, epithelial calcium channel, or calbindin-D(28K) expression in low-protein rat kidneys. However, Na(+)-K(+)-ATPase activity was 36% lower (P < 0.05) in low-protein rats. These data suggest that the hypercalciuria of low-protein rats arises through a reduction in passive calcium reabsorption in the proximal tubule rather than active distal tubule uptake. This may contribute to the reduction in bone mass observed in this model.


Subject(s)
Calcium/metabolism , Dietary Proteins/pharmacology , Kidney Tubules, Distal/metabolism , Kidney Tubules, Proximal/metabolism , Prenatal Exposure Delayed Effects , Animals , Biological Transport, Active/physiology , Blotting, Western , Bone and Bones/embryology , Bone and Bones/metabolism , Calbindins , Calcium Channels/metabolism , Diet, Protein-Restricted , Female , Fetal Nutrition Disorders/metabolism , Glomerular Filtration Rate , Hypercalciuria/metabolism , Kidney Tubules, Distal/embryology , Kidney Tubules, Proximal/embryology , Male , Pregnancy , Rats , Rats, Wistar , S100 Calcium Binding Protein G/metabolism , Sodium-Hydrogen Exchanger 3 , Sodium-Hydrogen Exchangers/metabolism , Sodium-Potassium-Exchanging ATPase/metabolism , TRPV Cation Channels/metabolism
10.
J Am Soc Nephrol ; 18(4): 1121-9, 2007 Apr.
Article in English | MEDLINE | ID: mdl-17314325

ABSTRACT

Pax genes are important regulators of kidney development. In the mouse, homozygous Pax2 inactivation results in renal agenesis, a phenotype that has largely precluded the analysis of Pax gene function during metanephric kidney development. To address this later function, kidney development was analyzed in embryos that were compound heterozygous for Pax2 and for Pax8, a closely related member of the Pax gene family. Both genes are coexpressed in differentiating nephrons and collecting ducts. At the morphological level, Pax2(+/-)Pax8(+/-) metanephric kidneys are severely hypodysplastic and characterized by a reduction in ureter tips and nephron number in comparison with wild-type or Pax2(+/-) kidneys. In developing nephrons, the molecular analysis of Pax2(+/-)Pax8(+/-) kidneys reveals a strong reduction in the expression levels of Lim1, a key regulator of nephron differentiation, accompanied by an increase in apoptosis. At a more mature stage, the reduction of Pax2/8 gene dosage severely affects distal tubule formation, revealing a role for Pax genes in the differentiation of specific nephron segments. At the ureter tips, the expression of Wnt11, a target of glial cell-derived neurotrophic factor-Ret signaling, is significantly reduced, whereas the expression levels of Ret and GDNF remain normal. Together, these results demonstrate a crucial role for Pax2 and Pax8 in nephron differentiation and branching morphogenesis of the metanephros.


Subject(s)
Kidney/embryology , Morphogenesis , Nephrons/embryology , PAX2 Transcription Factor/physiology , Paired Box Transcription Factors/physiology , Animals , Cell Differentiation , Cell Survival , Gene Expression Regulation, Developmental , Kidney Tubules, Distal/embryology , Mice , Mice, Inbred C3H , PAX2 Transcription Factor/genetics , PAX8 Transcription Factor , Paired Box Transcription Factors/genetics , Wnt Proteins/genetics
11.
Development ; 133(23): 4737-48, 2006 Dec.
Article in English | MEDLINE | ID: mdl-17079272

ABSTRACT

Duct epithelial structure is an essential feature of many internal organs, including exocrine glands and the kidney. The ducts not only mediate fluid transfer but also help to maintain homeostasis. For instance, fluids and solutes are resorbed from or secreted into the primary fluid flowing through the lumen of the ducts in the exocrine glands and kidneys. The molecular mechanism underlying the functional maturation of these ducts remains largely unknown. Here, we show that a grainyhead-related transcription factor, CP2-like 1 (CP2L1), is required for the maturation of the ducts of the salivary gland and kidney. In the mouse, Cp2l1 is specifically expressed in the developing ducts of a number of exocrine glands, including the salivary gland, as well as in those of the kidney. In Cp2l1-deficient mice, the expression of genes directly involved in functional maturation of the ducts was specifically reduced in both the salivary gland and kidney, indicating that Cp2l1 is required for the differentiation of duct cells. Furthermore, the composition of saliva and urine was abnormal in these mice. These results indicate that Cp2l1 expression is required for normal duct development in both the salivary gland and kidney.


Subject(s)
Kidney/embryology , Repressor Proteins/metabolism , Salivary Ducts/embryology , Animals , Base Sequence , DNA Primers/genetics , Female , Gene Expression Regulation, Developmental , Kidney/metabolism , Kidney Tubules, Collecting/embryology , Kidney Tubules, Collecting/metabolism , Kidney Tubules, Distal/embryology , Kidney Tubules, Distal/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Organ Culture Techniques , Repressor Proteins/genetics , Saliva/chemistry , Salivary Ducts/metabolism , Urine/chemistry
12.
Development ; 133(15): 2961-71, 2006 Aug.
Article in English | MEDLINE | ID: mdl-16818449

ABSTRACT

Notch signaling has been shown to play a role in cell fate decisions in the Xenopus pronephros anlagen. Here, we show that the Xenopus Hairy-related transcription factor (HRT) gene XHRT1, and the Hairy/Enhancer of split (HES) genes Xhairy1, Xhairy2b, esr9 and esr10, have distinct restricted dynamic expression patterns during pronephros development, and that their expression is regulated by Notch. XHRT1, which is the earliest and strongest gene expressed in the pronephric region, is initially transcribed predominantly in the forming glomus, where it is downregulated by antisense morpholino oligonucleotide inhibition of xWT1. Later, it is activated in the most dorsoanterior part of the pronephros anlagen that gives rise to the proximal tubules. In agreement with this dynamic expression profile, we found that early activation of Notch favors glomus, whereas only later activation promotes proximal tubule formation. We show that, among the bHLH-O factors tested, only XHRT1 efficiently inhibits distal tubule and duct formation, and that only its translational inhibition causes a reduction of the expression of proximal tubule and glomus markers. Using domain swap experiments, we found that the XHRT1 C-terminal region is crucial for its activity. Together, our results provide evidence that XHRT1 plays an important role in glomerular development and early proximodistal patterning that is distinct from those of the other pronephric bHLH repressors.


Subject(s)
Kidney Glomerulus/embryology , Transcription Factors/genetics , Xenopus Proteins/genetics , Xenopus/embryology , Animals , Body Patterning , Embryo, Nonmammalian/physiology , Gene Expression Regulation, Developmental , In Situ Hybridization , Kidney Tubules, Distal/embryology , Protein Biosynthesis , Xenopus/genetics
13.
Kidney Int ; 68(4): 1643-7, 2005 Oct.
Article in English | MEDLINE | ID: mdl-16164641

ABSTRACT

BACKGROUND: Preterm neonates and renal transplant patients frequently develop nephrocalcinosis. Experimental studies revealed that crystal retention in the distal nephron, a process that may lead to nephrocalcinosis, is limited to proliferating/regenerating tubular cells expressing hyaluronan and osteopontin at their luminal surface. Fetal and transplant kidneys contain proliferating and/or regenerating cells since nephrogenesis is not completed until 36 weeks of gestation, while ischemia and nephrotoxic immunosuppressants may lead to injury and repair in renal transplants. This prompted us to investigate the expression of hyaluronan and osteopontin and to correlate this to the appearance of tubular calcifications both in fetal/preterm and transplanted kidneys. METHODS: Sections of fetal/preterm kidneys and protocol biopsies of transplanted kidneys (12 and 24 weeks posttransplantation from the same patients) were stained for osteopontin, hyaluronan, and calcifications (von Kossa). RESULTS: Hyaluronan and osteopontin were expressed at the luminal surface of the epithelial cells lining the distal tubules of all fetal kidneys at birth and in all kidney graft protocol biopsies 12 and 24 weeks posttransplantation. In 7 out of 18 surviving (at least 4 days) preterm neonates crystal retention developed. In renal allografts a striking increase (from 2/10 to 6/10) in tubular crystal retention between 12 and 24 weeks posttransplantation was observed. In addition, crystals were selectively retained in distal renal tubules containing cells with hyaluronan and osteopontin at their luminal surface. CONCLUSION: The results of this study show that luminal expression of hyaluronan and osteopontin preceded renal distal tubular retention of crystals in preterm neonates and renal transplant patients. We propose that the presence of this crystal binding phenotype may play a general role in renal calcification processes.


Subject(s)
Kidney Transplantation , Kidney Tubules, Distal/metabolism , Kidney Tubules, Distal/pathology , Nephrocalcinosis/metabolism , Nephrocalcinosis/pathology , Cell Division , Chronic Disease , Crystallization , Cyclosporine/adverse effects , Graft Rejection/metabolism , Graft Rejection/pathology , Humans , Hyaluronic Acid/metabolism , Immunosuppressive Agents/adverse effects , Infant, Newborn , Infant, Premature , Ischemia/metabolism , Ischemia/pathology , Kidney Tubules, Distal/embryology , Nephrons/embryology , Nephrons/metabolism , Nephrons/pathology , Osteopontin , Sialoglycoproteins/metabolism
14.
Kidney Int ; 68(2): 487-96, 2005 Aug.
Article in English | MEDLINE | ID: mdl-16014025

ABSTRACT

BACKGROUND: Ontogeny and cellular distribution of vasopressin receptors in the kidney are key factors determining the role of vasopressin in renal physiology. Expression of vasopressin V(2) receptor (V(2)R) mRNA and the immunoreactive protein in rat kidney were investigated. METHODS: An antiserum directed to epitope TLD25 of the rat V(2)R sequence was characterized by Western blotting. Expression of V(2)R mRNA was assessed by reverse transcription-polymerase chain reaction (RT-PCR), and on protein level by immunohistochemistry. RESULTS: Specificity of the antiserum was documented by Western blots from cells expressing a fusion protein of V(2)R and GFP. Using lysates of rat kidney and of native cell lines expressing V(2)R but not V(1)R, our antiserum to peptide TLD25 revealed a major band of 55 kD corresponding to the monomeric form of V(2)R, and a band of 110 kD most likely representing the homodimeric form of the receptor. This highly specific antiserum allowed us to localize the V(2)R in thick ascending limbs, distal convoluted and connecting tubules, and in collecting ducts. During ontogeny, immunoreactivity was first observed at the luminal membrane on prenatal day 20, emerging at the basolateral side from postnatal day 5 on. RT-PCR demonstrated V(2)R transcripts from prenatal day 18 to gradually increasing thereafter. CONCLUSION: Expression of V(2)R is first detectable in the late embryonic stage of rat ontogeny starting from day E18 and gradually increasing with kidney maturation. In the adult kidney, V(2)R is differentially distributed in the various nephron segments.


Subject(s)
Nephrons/embryology , Nephrons/physiology , Receptors, Vasopressin/genetics , Receptors, Vasopressin/metabolism , Age Factors , Animals , Antibody Specificity , Cell Membrane/metabolism , Female , Gene Expression Regulation, Developmental , Gestational Age , Immunohistochemistry , Kidney Tubules, Collecting/embryology , Kidney Tubules, Collecting/physiology , Kidney Tubules, Distal/embryology , Kidney Tubules, Distal/physiology , Loop of Henle/embryology , Loop of Henle/physiology , Male , Pregnancy , RNA, Messenger/analysis , Rats , Rats, Sprague-Dawley , Receptors, Vasopressin/immunology
15.
Development ; 130(19): 4751-9, 2003 Oct.
Article in English | MEDLINE | ID: mdl-12925600

ABSTRACT

This study identifies a role for the gene for the POU transcription factor Brn1 in distal tubule formation and function in the mammalian kidney. Normal development of Henle's loop (HL), the distal convoluted tubule and the macula densa was severely retarded in Brn1-deficient mice. In particular, elongation and differentiation of the developing HL was affected. In the adult kidney, Brn1 was detected only in the thick ascending limb (TAL) of HL. In addition, the expression of a number of TAL-specific genes was reduced in the Brn1+/- kidney, including Umod, Nkcc2/Slc12a1, Bsnd, Kcnj1 and Ptger3. These results suggest that Brn1 is essential for both the development and function of the nephron in the kidney.


Subject(s)
Kidney Tubules, Distal/embryology , Kidney Tubules, Distal/metabolism , Neuropeptides/metabolism , Potassium Channels, Inwardly Rectifying , Trans-Activators/metabolism , Animals , Animals, Newborn , Chloride Channels , Female , Gene Dosage , Kidney Tubules, Distal/cytology , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice , Mice, Knockout , Mucoproteins/genetics , Mucoproteins/metabolism , Nerve Tissue Proteins , Neuropeptides/genetics , POU Domain Factors , Potassium Channels/genetics , Potassium Channels/metabolism , Pregnancy , Receptors, Prostaglandin E/genetics , Receptors, Prostaglandin E/metabolism , Receptors, Prostaglandin E, EP3 Subtype , Renal Insufficiency , Sodium-Potassium-Chloride Symporters/genetics , Sodium-Potassium-Chloride Symporters/metabolism , Solute Carrier Family 12, Member 1 , Trans-Activators/genetics , Uromodulin
16.
Anat Embryol (Berl) ; 205(5-6): 441-52, 2002 Oct.
Article in English | MEDLINE | ID: mdl-12382147

ABSTRACT

Glucose is an essential molecule for most mammalian cells, and is particularly important during fetal development, when cells are rapidly dividing and differentiating. In rats, GLUT1 is present at high levels in most fetal tissues, with levels decreasing after birth. We used immunohistochemistry to localise GLUT12 protein, a recently identified member of the sugar transporter family, and GLUT1 during rat fetal development. GLUT12 staining was observed in heart muscle from gestational days 15 to 21. GLUT12 staining in skeletal muscle increased from gestational days 17 to 21, and GLUT12 was also detected in brown adipose tissue. The expression of GLUT12 in insulin-responsive tissues supports a potential role for GLUT12 in the provision of glucose to these tissues before the appearance of GLUT4. GLUT12 protein was also expressed in fetal chondrocytes from gestational day 15 onward, in kidney distal tubules and collecting ducts from day 19, and in lung bronchioles from day 19. The specific pattern of expression observed in the rat fetus suggests that GLUT12 may be important in hexose delivery to developing tissues.


Subject(s)
Kidney Tubules, Distal/chemistry , Kidney Tubules, Distal/embryology , Monosaccharide Transport Proteins/analysis , Myocardium/chemistry , Animals , Bronchi/chemistry , Bronchi/embryology , Chondrocytes/chemistry , Female , Glucose Transport Proteins, Facilitative , Glucose Transporter Type 1 , Heart/embryology , Kidney Tubules, Collecting/chemistry , Kidney Tubules, Collecting/embryology , Muscle, Skeletal/chemistry , Muscle, Skeletal/embryology , Pregnancy , Rats , Rats, Sprague-Dawley
17.
Connect Tissue Res ; 37(1-2): 53-60, 1998.
Article in English | MEDLINE | ID: mdl-9643646

ABSTRACT

Tubulointerstitial nephritis antigen (TIN-ag) is a 58 kDa glycoprotein restricted within the kidney to basement membranes underlying the epithelium of Bowman's capsule and proximal and distal tubules. Autoantibody formation against this component has been described in association with primary immune-mediated tubulointerstital nephritis, membranous nephropathy and anti-glomerular basement membrane nephritis. In the present report, the ontogeny of this protein was studied in human fetal kidney tissue by immunohistochemical analysis of immature and developing nephrons using a panel of monoclonal and polyclonal antibodies. TIN-ag is first detected in basement membranes underlying the epithelium of Bowman's capsule of early capillary loop stage glomeruli and the primitive proximal tubule. No detectable expression is observed in the basement membranes of the branching ureteric bud, nephrogenic vesicle, or comma shape and s-shape stages of nephrogenic development. Increased staining of the proximal tubular basement membrane is associated with outgrowth of the primitive tubule from the urinary pole of the developing glomerulus. In more mature fetal tubules, TIN-ag expression closely resembles that of previously reported observations in mature tissue where it is present in high amounts in the basement membranes of proximal tubules, and to a lesser extent in Bowman's capsule and distal tubules. Our results suggest that TIN-ag expression is developmentally regulated in a precise spatial and temporal pattern throughout nephrogenesis.


Subject(s)
Cell Adhesion Molecules/biosynthesis , Membrane Glycoproteins/biosynthesis , Nephrons/embryology , Telomere-Binding Proteins , Antibodies, Monoclonal/metabolism , Antigens, Surface , Collagen/metabolism , Fetus , Fluorescent Antibody Technique , Humans , Kidney Glomerulus/cytology , Kidney Glomerulus/embryology , Kidney Glomerulus/immunology , Kidney Tubules, Distal/cytology , Kidney Tubules, Distal/embryology , Kidney Tubules, Distal/immunology , Mucin-1/metabolism , Nephrons/cytology , Nephrons/immunology , Ureter/cytology , Ureter/embryology , Ureter/immunology
18.
J Biol Chem ; 272(21): 13725-30, 1997 May 23.
Article in English | MEDLINE | ID: mdl-9153225

ABSTRACT

The hepatocyte nuclear factor-3 (HNF-3)/fork head homolog (HFH) proteins are an extensive family of transcription factors, which share homology in the winged helix DNA binding domain. Members of the HFH/winged helix family have been implicated in cell fate determination during pattern formation, in organogenesis, and in cell-type-specific gene expression. In this study we isolated a full-length HFH-3 cDNA clone from a human kidney library which encoded a 351-amino acid protein containing a centrally located winged helix DNA binding domain. We demonstrate that HFH-3 is a potent transcriptional activator requiring 138 C-terminal residues for activity. We used in situ hybridization to demonstrate that HFH-3 expression is restricted to the epithelium of the renal distal convoluted tubules. We determined the HFH-3 DNA binding consensus sequence by in vitro DNA binding site selection using recombinant HFH-3 protein and used this consensus sequence to identify putative HFH-3 target genes expressed there. These putative HFH-3 target genes include the Na/K-ATPase, Na/H and anion exchangers, E-cadherin, and mineralocorticoid receptor genes as well as genes for the transcription factors HNF-1, vHNF-1, and HNF-4.


Subject(s)
DNA-Binding Proteins , Kidney Tubules, Distal/chemistry , Trans-Activators/biosynthesis , Aging/genetics , Aging/metabolism , Amino Acid Sequence , Animals , Base Sequence , Binding Sites , Consensus Sequence , DNA/metabolism , DNA, Complementary/isolation & purification , Epithelium/chemistry , Forkhead Transcription Factors , Gene Library , Humans , In Situ Hybridization , Kidney Tubules, Distal/embryology , Kidney Tubules, Distal/metabolism , Mice , Molecular Sequence Data , Rats , Recombinant Proteins/metabolism , Trans-Activators/genetics , Trans-Activators/metabolism , Transcription, Genetic
19.
J Anat ; 183 ( Pt 1): 141-7, 1993 Aug.
Article in English | MEDLINE | ID: mdl-7505779

ABSTRACT

The human kidney develops from 2 embryonic tissues, the ureteric bud and the metanephric blastema. The site in the adult renal distal tubule corresponding to the junction between these tissues has never been established unequivocally and is usually said to be the union between the collecting duct and the connecting piece, based on microdissection evidence. We have examined kidneys from 21 human fetuses of various ages using an immunohistological method for substances related to the ABO blood group system, various cytokeratins including those detected by the monoclonal antibody PKK2, and Tamm-Horsfall protein. The ureteric bud and connecting piece expressed the type 1 precursor chain of ABO antigens mostly early in gestation, the H antigen of the ABO system mostly later in gestation, and cytokeratins detected by PKK2. The induced nephrons after the S-shaped body stage expressed Tamm-Horsfall protein. In the adult renal tubule, distal from the macula densa, it was already known that there is a sharp junction between the segment expressing Tamm-Horsfall protein and the more distal segment that expresses the H antigen and cytokeratins detected by PKK2. The finding that the ureteric bud and connecting piece express the same antigens as this segment while the S-shaped body eventually expresses Tamm-Horsfall protein is consistent with the concept that (1) the connecting piece arises from the ureteric bud, not the S-shaped body, and (2) the junction of ureteric bud derivatives and metanephric blastema derivatives is on the distal side of the macula densa at the distal end of Tamm-Horsfall staining.


Subject(s)
Kidney Tubules, Distal/embryology , ABO Blood-Group System/analysis , Humans , Immunoenzyme Techniques , Isoantigens/analysis , Keratins/analysis , Kidney/chemistry , Kidney/embryology , Kidney Cortex/chemistry , Kidney Tubules, Distal/chemistry , Mucoproteins/analysis , Uromodulin
20.
Endocrinology ; 133(1): 3-10, 1993 Jul.
Article in English | MEDLINE | ID: mdl-8319578

ABSTRACT

The insulin receptor-related receptor (IRR) demonstrates striking structural homology to the insulin receptor (IR) and insulin-like growth factor-I receptor (IGFR), suggesting that IRR is a member of the IR family. However, the endogenous ligand and biological role for this "orphan" receptor are unknown. To identify potential sites of action for the IRR, in situ hybridization was employed to reveal cellular patterns of IRR gene expression in the developing and adult rat and in the adult human kidney. From embryonic days 15-20, IRR mRNA is most abundant in sensory neurons of the trigeminal and dorsal root ganglia and, to a lesser extent, neurons of the autonomic system. IRR gene expression diminishes in the majority of sensory neurons postnatally, but remains abundant in a subpopulation of adult rat trigeminal and dorsal root ganglion neurons. IRR mRNA is localized in peripheral autonomic ganglia localized in the adrenal medulla and renal hilum in the adult. From birth to maturity, IRR mRNA is abundant in renal epithelial cells focally localized in the distal tubule in both rat and human kidney. The specificity of this pattern of IRR gene expression was demonstrated by hybridization of serial tissue sections with two different nonoverlapping cRNA probes. Nonspecific signal, as measured by IRR sense probe hybridization, was negligible. This highly restricted pattern of IRR gene expression is in marked contrast to the very widespread pattern of gene expression demonstrated by the IR and IGFR. This study showed that IRR, IR, and IGFR mRNAs were colocalized in some sensory neurons, suggesting the possibility for hybrid receptor formation in these cells. In summary, IRR gene expression is focally localized in sensory and autonomic neurons and renal distal tubule cells. These observations suggest that the IRR, in contrast to the related IR and IGFR, serves a narrow cell-specific role.


Subject(s)
Gene Expression , Kidney Tubules, Distal/innervation , Neurons/metabolism , RNA, Messenger/metabolism , Receptor, Insulin/genetics , Animals , Epithelium/metabolism , Female , In Situ Hybridization , Kidney Tubules, Distal/embryology , Kidney Tubules, Distal/metabolism , Neurons, Afferent/metabolism , RNA Probes , RNA, Antisense , Rats , Rats, Sprague-Dawley , Sympathetic Nervous System/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...