Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 19 de 19
1.
J Anat ; 233(2): 222-242, 2018 08.
Article En | MEDLINE | ID: mdl-29797482

Orofacial clefting represents the most common craniofacial birth defect. Cleft lip with or without cleft palate (CL/P) is genetically distinct from cleft palate only (CPO). Numerous transcription factors (TFs) regulate normal development of the midface, comprising the premaxilla, maxilla and palatine bones, through control of basic cellular behaviors. Within the Pbx family of genes encoding Three Amino-acid Loop Extension (TALE) homeodomain-containing TFs, we previously established that in the mouse, Pbx1 plays a preeminent role in midfacial morphogenesis, and Pbx2 and Pbx3 execute collaborative functions in domains of coexpression. We also reported that Pbx1 loss from cephalic epithelial domains, on a Pbx2- or Pbx3-deficient background, results in CL/P via disruption of a regulatory network that controls apoptosis at the seam of frontonasal and maxillary process fusion. Conversely, Pbx1 loss in cranial neural crest cell (CNCC)-derived mesenchyme on a Pbx2-deficient background results in CPO, a phenotype not yet characterized. In this study, we provide in-depth analysis of PBX1 and PBX2 protein localization from early stages of midfacial morphogenesis throughout development of the secondary palate. We further establish CNCC-specific roles of PBX TFs and describe the developmental abnormalities resulting from their loss in the murine embryonic secondary palate. Additionally, we compare and contrast the phenotypes arising from PBX1 loss in CNCC with those caused by its loss in the epithelium and show that CNCC-specific Pbx1 deletion affects only later secondary palate morphogenesis. Moreover, CNCC mutants exhibit perturbed rostro-caudal organization and broadening of the midfacial complex. Proliferation defects are pronounced in CNCC mutants at gestational day (E)12.5, suggesting altered proliferation of mutant palatal progenitor cells, consistent with roles of PBX factors in maintaining progenitor cell state. Although the craniofacial skeletal abnormalities in CNCC mutants do not result from overt patterning defects, osteogenesis is delayed, underscoring a critical role of PBX factors in CNCC morphogenesis and differentiation. Overall, the characterization of tissue-specific Pbx loss-of-function mouse models with orofacial clefting establishes these strains as unique tools to further dissect the complexities of this congenital craniofacial malformation. This study closely links PBX TALE homeodomain proteins to the variation in maxillary shape and size that occurs in pathological settings and during evolution of midfacial morphology.


Cranial Nerves/embryology , Homeodomain Proteins/physiology , Palate/embryology , Pre-B-Cell Leukemia Transcription Factor 1/physiology , Proto-Oncogene Proteins/physiology , Animals , Cleft Palate/genetics , Cranial Nerves/metabolism , Female , Mice , Mice, Transgenic , Palate/metabolism , Pregnancy
2.
Angiogenesis ; 21(2): 335-347, 2018 05.
Article En | MEDLINE | ID: mdl-29397483

The principal function of glomeruli is to filter blood through a highly specialized filtration barrier consisting of a fenestrated endothelium, the glomerular basement membrane and podocyte foot processes. Previous studies have uncovered a crucial role of endothelial a disintegrin and metalloprotease 10 (ADAM10) and Notch signaling in the development of glomeruli, yet the resulting defects have not been further characterized nor understood in the context of kidney development. Here, we used several different experimental approaches to analyze the kidneys and glomeruli from mice lacking ADAM10 in endothelial cells (A10ΔEC mice). Scanning electron microscopy of glomerular casts demonstrated enlarged vascular diameter and increased intussusceptive events in A10ΔEC glomeruli compared to controls. Consistent with these findings, genes known to regulate vessel caliber (Apln, AplnR and Vegfr3) are significantly upregulated in A10ΔEC glomeruli. Moreover, transmission electron microscopy revealed the persistence of diaphragms in the fenestrae of A10ΔEC glomerular endothelial cells, which was corroborated by the elevated expression of the protein PLVAP/PV-1, an integral component of fenestral diaphragms. Analysis of gross renal vasculature by light sheet microscopy showed no major alteration of the branching pattern, indicating a localized importance of ADAM10 in the glomerular endothelium. Since intussusceptions and fenestrae with diaphragms are normally found in developing, but not mature glomeruli, our results provide the first evidence for a crucial role of endothelial ADAM10, a key regulator of Notch signaling, in promoting the development and maturation of the glomerular vasculature.


ADAM10 Protein/metabolism , Amyloid Precursor Protein Secretases/metabolism , Endothelial Cells/metabolism , Kidney Glomerulus/metabolism , Membrane Proteins/metabolism , Receptors, Notch/metabolism , Signal Transduction/physiology , Animals , Endothelial Cells/ultrastructure , Kidney Glomerulus/blood supply , Kidney Glomerulus/ultrastructure , Mice , Mice, Transgenic
3.
Development ; 142(15): 2653-64, 2015 Aug 01.
Article En | MEDLINE | ID: mdl-26138478

The architecture of an organ's vascular bed subserves its physiological function and metabolic demands. However, the mechanisms underlying gross vascular patterning remain elusive. Using intravital dye labeling and 3D imaging, we discovered that systems-level vascular patterning in the kidney is dependent on the kinetics of vascular mural cell (VMC) differentiation. Conditional ablation of the TALE transcription factor Pbx1 in renal VMC progenitors in the mouse led to the premature upregulation of PDGFRß, a master initiator of VMC-blood vessel association. This precocious VMC differentiation resulted in nonproductive angiogenesis, abnormal renal arterial tree patterning and neonatal death consistent with kidney dysfunction. Notably, we establish that Pbx1 directly represses Pdgfrb, and demonstrate that decreased Pdgfrb dosage in conditional Pbx1 mutants substantially rescues vascular patterning defects and neonatal survival. These findings identify, for the first time, an in vivo transcriptional regulator of PDGFRß, and reveal a previously unappreciated role for VMCs in systems-level vascular patterning.


Cell Differentiation/physiology , Homeodomain Proteins/metabolism , Kidney/blood supply , Receptor, Platelet-Derived Growth Factor beta/metabolism , Transcription Factors/metabolism , Animals , Blotting, Western , Chromatin Immunoprecipitation , Electrophoresis, Polyacrylamide Gel , Flow Cytometry , Fluorescent Antibody Technique , Homeodomain Proteins/genetics , Imaging, Three-Dimensional , In Situ Hybridization , Kaplan-Meier Estimate , Kidney/cytology , Kinetics , Mice , Oligonucleotides/genetics , Pre-B-Cell Leukemia Transcription Factor 1 , Real-Time Polymerase Chain Reaction , Transcription Factors/genetics
4.
Semin Cell Dev Biol ; 36: 50-6, 2014 Dec.
Article En | MEDLINE | ID: mdl-25128732

The renal vascular bed has a stereotypic architecture that is essential for the kidney's role in excreting metabolic waste and regulating the volume and composition of body fluids. The kidney's excretory functions are dependent on the delivery of the majority of renal blood flow to the glomerular capillaries, which filter plasma removing from it metabolic waste, as well as vast quantities of solutes and fluids. The renal tubules reabsorb from the glomerular filtrate solutes and fluids required for homeostasis, while the post-glomerular capillary beds return these essential substances back into the systemic circulation. Thus, the kidney's regulatory functions are dependent on the close proximity or alignment of the post-glomerular capillary beds with the renal tubules. This review will focus on our current knowledge of the mechanisms controlling the embryonic development of the renal vasculature. An understanding of this process is critical for developing novel therapies to prevent vessel rarefaction and will be essential for engineering renal tissues suitable for restoring kidney function to the ever-increasing population of patients with end stage renal disease.


Kidney Glomerulus/blood supply , Kidney Tubules/blood supply , Kidney/blood supply , Kidney/embryology , Humans , Kidney Diseases/metabolism , Kidney Glomerulus/cytology , Kidney Glomerulus/embryology , Kidney Tubules/embryology , Neovascularization, Physiologic
5.
FASEB J ; 28(2): 730-9, 2014 Feb.
Article En | MEDLINE | ID: mdl-24189942

Renal pacemakers set the origin and frequency of the smooth muscle contractions that propel wastes from the kidney to the bladder. Although congenital defects impairing this peristalsis are a leading cause of pediatric renal failure, the mechanisms underlying renal pacemaker activity remain unknown. Using ratiometric optical mapping and video microscopy, we discovered that hyperpolarization-activated cation (HCN) channel block with the specific anatagonist ZD7288 (30 µm; IC50) abolished the pacemaker depolarizations that initiate murine upper urinary tract peristalsis. Optical mapping and immunohistochemistry indicate that pacemaker potentials are generated by cells expressing HCN isoform-3, and that HCN3(+) cells are coupled to definitive smooth muscle via gap junctions. Furthermore, we demonstrate that HCN3(+) cells coexpress T-type Ca(2+) (TTC) channels and that TTC channel inhibition with R(-)efonidipine or NNC55-0396 decreased contractile frequency in a dose-dependent manner. Collectively, these data demonstrate that HCN3(+)/TTC(+) cells are the pacemakers that set the origin and rate of upper urinary tract peristalsis. These results reveal a conserved mechanism controlling autorhythmicity in 2 distinct muscle types, as HCN and TTC channels also mediate cardiac pacemaker activity. Moreover, these findings have translational applications, including the development of novel diagnostics to detect fetal urinary tract motility defects prior to renal damage.-Hurtado, R., Bub, G., Herzlinger, D. A molecular signature of tissues with pacemaker activity in the heart and upper urinary tract involves coexpressed hyperpolarization-activated cation and T-type Ca(2+) channels.


Hyperpolarization-Activated Cyclic Nucleotide-Gated Channels/metabolism , Kidney/metabolism , Muscle, Smooth/metabolism , Animals , Dihydropyridines/pharmacology , Hyperpolarization-Activated Cyclic Nucleotide-Gated Channels/antagonists & inhibitors , Immunohistochemistry , Kidney/drug effects , Mice , Muscle Contraction/drug effects , Muscle, Smooth/drug effects , Nitrophenols/pharmacology , Organophosphorus Compounds/pharmacology , Peristalsis/drug effects , Urinary Tract/drug effects , Urinary Tract/metabolism
6.
Development ; 140(22): 4565-73, 2013 Nov.
Article En | MEDLINE | ID: mdl-24154527

Kidneys remove unwanted substances from the body and regulate the internal body environment. These functions are carried out by specialized cells (podocytes) that act as a filtration barrier between the internal milieu and the outside world, and by a series of tubules and ducts that process the filtrate and convey it to the outside. In the kidneys of amniote vertebrates, the filtration (podocyte) and tubular functions are tightly integrated into functional units called nephrons. The specification of the podocyte and tubular components of amniote nephrons is currently not well understood. The present study investigates podocyte and tubule differentiation in the avian mesonephric kidney, and presents several findings that refine our understanding of the initial events of nephron formation. First, well before the first morphological or molecular signs of nephron formation, mesonephric mesenchyme can be separated on the basis of morphology and the expression of the transcription factor Pod1 into dorsal and ventral components, which can independently differentiate in culture along tubule and podocyte pathways, respectively. Second, canonical Wnt signals, which are found in the nephric duct adjacent to the dorsal mesonephric mesenchyme and later in portions of the differentiating nephron, strongly inhibit podocyte but not tubule differentiation, suggesting that Wnt signaling plays an important role in the segmentation of the mesonephric mesenchyme into tubular and glomerular segments. The results are discussed in terms of their broader implications for models of nephron segmentation.


Body Patterning , Chickens/metabolism , Kidney Tubules/embryology , Kidney Tubules/metabolism , Podocytes/cytology , Wnt Signaling Pathway , Animals , Cell Differentiation , Chick Embryo , Kidney Glomerulus/cytology , Kidney Glomerulus/embryology , Kidney Tubules/cytology , Mesoderm/cytology , Mesoderm/embryology , Mesonephros/embryology , Models, Biological , Nephrons/cytology , Nephrons/metabolism , Podocytes/metabolism , Time Factors
7.
J Clin Invest ; 121(3): 836-8, 2011 Mar.
Article En | MEDLINE | ID: mdl-21339639

Mutations in GLI3, a component of the Sonic Hedgehog (Shh) signaling pathway, cause a variety of human developmental syndromes. In this issue of the JCI, Cain and colleagues show that tightly regulated GLI3 repressor activity is essential for Shh-dependent differentiation of upper urinary tract pacemaker cells and the efficient flow of urine from the kidney to the bladder. These results link defective pacemaker cell differentiation with hydronephrosis and provide a cellular basis for one of the abnormal renal defects observed in humans with the GLI3-linked disease Pallister-Hall syndrome.


Kruppel-Like Transcription Factors/physiology , Nerve Tissue Proteins/physiology , Urinary Tract/anatomy & histology , Urinary Tract/embryology , Animals , Cell Differentiation , Electrophysiology , Gene Expression Regulation, Developmental , Hedgehog Proteins/metabolism , Humans , Kidney/anatomy & histology , Models, Biological , Muscle Contraction , Muscle, Smooth/pathology , Pallister-Hall Syndrome/metabolism , Urinary Bladder/anatomy & histology , Zinc Finger Protein Gli3
8.
Kidney Int ; 77(6): 500-8, 2010 Mar.
Article En | MEDLINE | ID: mdl-20032965

Peristaltic waves of the ureteric smooth muscles move urine down from the kidney, a process that is commonly defective in congenital diseases. To study the mechanisms that control the initiation and direction of contractions, we used video microscopy and optical mapping techniques and found that electrical and contractile waves began in a region where the renal pelvis joined the connective tissue core of the kidney. Separation of this pelvis-kidney junction from more distal urinary tract segments prevented downstream peristalsis, indicating that it housed the trigger for peristalsis. Moreover, cells in the pelvis-kidney junction were found to express isoform 3 of the hyperpolarization-activated cation on channel family known to be required for initiating electrical activity in the brain and heart. Immunocytochemical and real-time PCR analyses found that hyperpolarization-activated cation-3 is expressed at the pelvis-kidney junction where electrical excitation and contractile waves originate. Inhibition of this channel caused a loss of electrical activity at the pelvis-kidney junction and randomized the origin of electrical activity in the urinary tract, thus markedly perturbing contractions. Collectively, our study demonstrates that hyperpolarization-activated cation-3 channels play a fundamental role in coordinating proximal-to-distal peristalsis of the upper urinary tract. This provides insight into the genetic causes of common inherited urinary tract disorders such as reflux and obstruction.


Cyclic Nucleotide-Gated Cation Channels/physiology , Kidney Pelvis/physiology , Potassium Channels/physiology , Ureter/physiology , Animals , Cations , Hyperpolarization-Activated Cyclic Nucleotide-Gated Channels , Kidney , Muscle Contraction/physiology , Muscle, Smooth/physiology , Pelvis , Peristalsis , Rats , Urinary Tract , Urinary Tract Physiological Phenomena
9.
Dev Biol ; 329(2): 169-75, 2009 May 15.
Article En | MEDLINE | ID: mdl-19272374

The development of most, if not all, tubular organs is dependent on signaling between epithelial and stromal progenitor populations. Most often, these lineages derive from different germ layers that are specified during gastrulation, well in advance of organ condensation. Thus, one of the first stages of organogenesis is the integration of distinct progenitor populations into a single embryonic rudiment. In contrast, the stromal and epithelial lineages controlling renal development are both believed to derive from the intermediate mesoderm and to be specified as the kidney develops. In this study we directly analyzed the lineage of renal epithelia and stroma in the developing chick embryo using two independent fate mapping techniques. Results of these experiments confirm the hypothesis that nephron epithelia derive from the intermediate mesoderm. Most importantly, we discovered that large populations of renal stroma originate in the paraxial mesoderm. Collectively, these studies suggest that the signals that subdivide mesoderm into intermediate and paraxial domains may play a role in specifying nephron epithelia and a renal stromal lineage. In addition, these fate mapping data indicate that renal development, like the development of all other tubular organs, is dependent on the integration of progenitors from different embryonic tissues into a single rudiment.


Kidney/embryology , Mesoderm , Stromal Cells/cytology , Animals , Body Patterning , Chick Embryo , Lac Operon
10.
J Urol ; 181(1): 401-7, 2009 Jan.
Article En | MEDLINE | ID: mdl-19010499

PURPOSE: Congenital ureteropelvic junction obstruction has been associated with aberrant ureteral smooth muscle organization. Recent evidence has shown that BMP4 may be involved in ureteral morphogenesis. We determined whether the disruption of BMP4 signaling results in abnormal smooth muscle investment of the ureter and ureteropelvic junction. MATERIALS AND METHODS: We used a Cre mediated Bmp4 knockout system to conditionally excise the Bmp4 gene in developing mouse embryos. Kidney rudiments were isolated from embryos at varying gestational ages from WT and conditional knockout mice. Metanephric kidney explants were cultured in the presence or absence of the BMP antagonist Noggin. Agarose beads pre-incubated with Gremlin, another BMP antagonist, were used for localized disruption of BMP signaling. Frozen sections and whole metanephric explants were then analyzed by immunofluorescence. RESULTS: Bmp4 gene excision resulted in a dose dependent loss of ureteral smooth muscle. Antagonism of BMP signaling inhibited ureteral smooth muscle investment in a dose dependent manner and was paralleled by a dose dependent decrease in the immediate downstream targets of BMP signaling, phosphorylated Smad1, 5 and 8. Localized antagonism of BMP resulted in the focal disruption of ureteral smooth muscle investment. CONCLUSIONS: We report that decreased BMP signaling, whether by the loss of BMP4 in vivo or direct antagonism in vitro, results in a gradual reduction of the normal, well organized coat of smooth muscle surrounding the ureter. Our results also suggest that this occurs via a direct Smad dependent pathway. This raises the possibility that abnormalities in BMP4 signaling may have a role in the development of congenital ureteropelvic junction obstruction.


Bone Morphogenetic Protein 4/physiology , Kidney Pelvis/embryology , Muscle, Smooth/embryology , Ureter/embryology , Ureteral Obstruction/etiology , Animals , Bone Morphogenetic Protein 4/antagonists & inhibitors , Bone Morphogenetic Protein 4/genetics , Carrier Proteins/pharmacology , Female , Intercellular Signaling Peptides and Proteins/pharmacology , Mice
11.
Dev Biol ; 326(1): 101-11, 2009 Feb 01.
Article En | MEDLINE | ID: mdl-19041859

Embryonic blood vessel formation is initially mediated through the sequential differentiation, migration, and assembly of endothelial cells (ECs). While many molecular signals that promote vascular development have been identified, little is known about suppressors of this process. In higher vertebrates, including birds and mammals, the vascular network forms throughout the embryonic disk with the exception of a region along the midline. We have previously shown that the notochord is responsible for the generation and maintenance of the avascular midline and that BMP antagonists expressed by this embryonic tissue, including Noggin and Chordin, can mimic this inhibitory role. Here we report that the notochord suppresses the generation of ECs from the mesoderm both in vivo and in vitro. We also report that the notochord diminishes the ability of mature ECs to organize into a primitive plexus. Furthermore, Noggin mimics notochord-based inhibition by preventing mesodermal EC generation and mature EC network formation. These findings suggest that the mesoderm surrounding the midline is competent to give rise to ECs and to form blood vessels, but that notochord derived-BMP antagonists suppress EC differentiation and maturation processes leading to inhibition of midline vessel formation.


Bone Morphogenetic Proteins/metabolism , Endothelial Cells/cytology , Notochord/physiology , Animals , Aorta/cytology , Bone Morphogenetic Proteins/antagonists & inhibitors , Carrier Proteins/metabolism , Cell Differentiation/physiology , Cells, Cultured , Chick Embryo , Endothelial Cells/physiology , Endothelium, Vascular/cytology , Endothelium, Vascular/physiology , Glycoproteins/metabolism , Humans , Intercellular Signaling Peptides and Proteins/metabolism , Mesoderm/cytology , Mesoderm/metabolism , Quail
12.
Gene Expr Patterns ; 7(6): 680-99, 2007 Jun.
Article En | MEDLINE | ID: mdl-17452023

Cataloguing gene expression during development of the genitourinary tract will increase our understanding not only of this process but also of congenital defects and disease affecting this organ system. We have developed a high-resolution ontology with which to describe the subcompartments of the developing murine genitourinary tract. This ontology incorporates what can be defined histologically and begins to encompass other structures and cell types already identified at the molecular level. The ontology is being used to annotate in situ hybridisation data generated as part of the Genitourinary Development Molecular Anatomy Project (GUDMAP), a publicly available data resource on gene and protein expression during genitourinary development. The GUDMAP ontology encompasses Theiler stage (TS) 17-27 of development as well as the sexually mature adult. It has been written as a partonomic, text-based, hierarchical ontology that, for the embryological stages, has been developed as a high-resolution expansion of the existing Edinburgh Mouse Atlas Project (EMAP) ontology. It also includes group terms for well-characterised structural and/or functional units comprising several sub-structures, such as the nephron and juxtaglomerular complex. Each term has been assigned a unique identification number. Synonyms have been used to improve the success of query searching and maintain wherever possible existing EMAP terms relating to this organ system. We describe here the principles and structure of the ontology and provide representative diagrammatic, histological, and whole mount and section RNA in situ hybridisation images to clarify the terms used within the ontology. Visual examples of how terms appear in different specimen types are also provided.


Gene Expression Regulation, Developmental , Mice/genetics , Urogenital System/growth & development , Animals , Clitoris/growth & development , Endoderm/physiology , Female , Male , Mesoderm/physiology , Mice/embryology , Mice/growth & development , Nephrons/embryology , Nephrons/growth & development , Penis/growth & development , Scrotum/growth & development , Sexual Maturation , Urogenital System/anatomy & histology
13.
Development ; 134(10): 1967-75, 2007 May.
Article En | MEDLINE | ID: mdl-17442697

Urinary tract morphogenesis requires the sub-division of the ureteric bud (UB) into the intra-renal collecting system and ureter, two tissues with unique structural and functional properties. In this report we investigate the cellular and molecular mechanisms that mediate their differentiation. Fate mapping experiments in the developing chick indicate that the UB is surrounded by two distinct mesenchymal populations: nephrogenic mesenchyme derived from the intermediate mesoderm and tailbud-derived mesoderm, which is selectively associated with the domain of the UB that differentiates into the ureter. Functional experiments utilizing murine metanephric kidney explants show that BMP4, a paracrine factor secreted by tailbud-derived mesenchyme, is required for ureter morphogenesis. Conversely, ectopic BMP4 signaling is sufficient to induce ureter morphogenesis in domains of the UB normally fated to differentiate into the intra-renal collecting system. Collectively, these results indicate that the border between the kidney and ureter forms where mesenchymal tissues originating in two different areas of the early embryo meet. These data raise the possibility that the susceptibility of this junction to congenital defects in humans, such as ureteral-pelvic obstructions, may be related to the complex morphogenetic movements that are required to integrate cells from these different lineages into a single functional structure.


Bone Morphogenetic Proteins/physiology , Gene Expression Regulation, Developmental , Signal Transduction , Ureter/embryology , Urinary Tract/embryology , Animals , Bone Morphogenetic Protein 4 , Bone Morphogenetic Proteins/metabolism , Cell Lineage , Chick Embryo , Epithelium/metabolism , Humans , In Situ Hybridization , Kidney/embryology , Mesoderm/metabolism , Microscopy, Fluorescence , RNA, Messenger/metabolism
14.
Development ; 132(17): 3847-57, 2005 Sep.
Article En | MEDLINE | ID: mdl-16049112

During kidney morphogenesis, the formation of nephrons begins when mesenchymal nephron progenitor cells aggregate and transform into epithelial vesicles that elongate and assume an S-shape. Cells in different regions of the S-shaped body subsequently differentiate into the morphologically and functionally distinct segments of the mature nephron. Here, we have used an allelic series of mutations to determine the role of the secreted signaling molecule FGF8 in nephrogenesis. In the absence of FGF8 signaling, nephron formation is initiated, but the nascent nephrons do not express Wnt4 or Lim1, and nephrogenesis does not progress to the S-shaped body stage. Furthermore, the nephron progenitor cells that reside in the peripheral zone, the outermost region of the developing kidney, are progressively lost. When FGF8 signaling is severely reduced rather than eliminated, mesenchymal cells differentiate into S-shaped bodies. However, the cells within these structures that normally differentiate into the tubular segments of the mature nephron undergo apoptosis, resulting in the formation of kidneys with severely truncated nephrons consisting of renal corpuscles connected to collecting ducts by an abnormally short tubular segment. Thus, unlike other FGF family members, which regulate growth and branching morphogenesis of the collecting duct system, Fgf8 encodes a factor essential for gene regulation and cell survival at distinct steps in nephrogenesis.


Fibroblast Growth Factors/metabolism , Gene Expression Regulation, Developmental/genetics , Nephrons/cytology , Nephrons/metabolism , Alleles , Animals , Cell Survival , Fibroblast Growth Factor 8 , Fibroblast Growth Factors/genetics , Mesoderm/metabolism , Mice , Mice, Transgenic , Nephrons/embryology , Phenotype , Signal Transduction , Spinal Cord/embryology , Spinal Cord/metabolism , Spinal Cord/pathology
15.
J Urol ; 173(1): 292-5, 2005 Jan.
Article En | MEDLINE | ID: mdl-15592099

PURPOSE: c-kit encodes a tyrosine kinase receptor that is required for the differentiation of a wide variety of cells during embryogenesis, including pacemaker cells of the gut. Functional expression of this tyrosine kinase receptor is required for gut peristalsis and c-kit expression has recently been documented in the adult murine urinary tract. In this study we analyzed the temporal onset of c-kit expression during ureter morphogenesis in vivo and determined if c-kit activity is essential for ureteral peristalsis in vitro. MATERIALS AND METHODS: The kidneys and ureters of gestational days 12.5 to 17.5 WT mice were isolated and frozen sections were prepared for analysis of c-kit, alpha-smooth muscle actin and uroplakin expression by immunocytochemical techniques. In addition, ureters were isolated from gestational day 15.5 mouse urogenital systems and cultured at the air/medium interface on 0.4 um pore polycarbonate membrane filters with Dulbecco's modified Eagle's medium/fetal calf serum in the presence or absence of antibodies that inhibit c-kit function. RESULTS: By gestational day 15.5 c-kit expression could be detected in a subset of renal epithelia and cells of the ureteropelvic adventitia. Prominent staining for c-kit was seen in the muscularis propria of the proximal ureter. In vitro studies demonstrated that isolated embryonic ureters acquire the ability to undergo unidirectional contractions after 3 days of culture, which is coincident with up-regulation of c-kit expression. Furthermore, incubation of isolated ureters with antibodies that neutralize c-kit activity markedly altered ureter morphology and peristalsis. CONCLUSIONS: We identified the initial expression and location of c-kit in the embryonic murine upper urinary tract. c-kit expression is up-regulated in the developing ureter prior to the ability of this tissue to undergo unidirectional contractions and c-kit function is required for the peristalsis in vitro.


Muscle Contraction/physiology , Proto-Oncogene Proteins c-kit/physiology , Ureter/physiology , Animals , Immunohistochemistry , Mice , Muscle, Smooth/physiology , Organ Culture Techniques , Up-Regulation/physiology , Ureter/embryology , Ureter/metabolism
16.
Dev Dyn ; 231(3): 601-8, 2004 Nov.
Article En | MEDLINE | ID: mdl-15376282

Mammalian kidney morphogenesis begins when the ureteric bud (UB) induces surrounding metanephric mesenchyme to differentiate into nephrons, the functional units of the mature organ. Although several genes required for this process have been identified, the mechanisms that control final nephron number and the localization of distinct tubular segments to cortical and medullary zones of the kidney remain poorly understood. This finding is due, in part, to the lack of quantitative studies describing the acquisition of mature renal structure. We have analyzed the following parameters of the developing murine kidney throughout embryogenesis: nephron and UB tip number, distance between UB branch points and total kidney, and cortical and medullary volume. Results of this morphometric analysis reveal previously unrecognized changes in the pattern of UB growth and rate of nephrogenesis. In addition, this morphometric index provides a much-needed reference for accurately describing renal patterning defects exhibited by genetically altered mice.


Embryonic Development , Kidney/embryology , Kidney/growth & development , Animals , Animals, Newborn , Calbindins , Female , Fluorescent Antibody Technique, Direct , Green Fluorescent Proteins/metabolism , Kidney Cortex/embryology , Kidney Cortex/growth & development , Kidney Glomerulus/embryology , Kidney Medulla/embryology , Kidney Medulla/growth & development , Male , Mesoderm/cytology , Mice , Mice, Transgenic , Models, Biological , Morphogenesis , Nephrons/embryology , Organ Size , Pregnancy , S100 Calcium Binding Protein G/metabolism , Ureter/embryology
17.
Proc Natl Acad Sci U S A ; 101(37): 13607-12, 2004 Sep 14.
Article En | MEDLINE | ID: mdl-15342908

In addition to the traditional renin-angiotensin system, a great deal of evidence favors the existence of numerous independent tissue-specific renin-angiotensin systems. We report that mast cells are an additional source of renin and constitute a unique extrarenal renin-angiotensin system. We use renin-specific antibodies to demonstrate that cardiac mast cells contain renin. Extending this observation to the human mast cell line HMC-1, we show that these mast cells also express renin. The HMC-1 renin RT-PCR product is 100% homologous to Homo sapiens renin. HMC-1 cells also contain renin protein, as demonstrated both by immunoblot and immunocytochemical analyses. Renin released from HMC-1 cells is active; furthermore, HMC-1 cells are able to synthesize renin. It is known that, in the heart, mast cells are found in the interstitium in close proximity to nerves and myocytes, which both express angiotensin II receptors. Inasmuch as myocardial interstitium contains angiotensinogen and angiotensin-converting enzyme, and because we were able to detect renin only in mast cells, we postulate that the release of renin from cardiac mast cells is the pivotal event triggering local formation of angiotensin II. Because of the ubiquity of mast cells, our results represent a unique paradigm for understanding local renin-angiotensin systems, not just in the heart, but in all tissues. Our findings provide a rationale for targeting mast cells in conjunction with renin-angiotensin system inhibitors in the management of angiotensin II-related dysfunctions.


Mast Cells/metabolism , Renin/metabolism , Animals , Cell Line , Female , Gene Expression , Humans , Kidney/cytology , Kidney/metabolism , Male , Microscopy, Confocal , Myocardium/cytology , Myocardium/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley
18.
Am J Physiol Renal Physiol ; 283(4): F799-809, 2002 Oct.
Article En | MEDLINE | ID: mdl-12217872

Renal epithelial cells derive from either cells of the metanephric mesenchyme or ureteric bud cells, but the origin of other renal cells is unclear. To test whether metanephric mesenchymal cells generate cells other than epithelial, we examined the developmental potential of a metanephric mesenchymal cell line (7.1.1 cells) and of primary cultures of metanephric mesenchymal cells. 7.1.1 Cells express both mesenchymal and epithelial markers and, on confluence, form well-defined monolayers expressing epithelial junctional proteins. However, 7.1.1 cells as well as primary cultures of metanephric mesenchymal cells also generate spindle-shaped cells that are positive for alpha-smooth muscle actin, indicating that they are myofibroblasts and/or smooth muscle; this differentiation pathway is inhibited by collagen IV and enhanced by fetal calf serum or transforming growth factor-beta(1). Transforming growth factor-beta(1) also induces expression of smooth muscle proteins, indicating that the cells differentiate into smooth muscle. 7.1.1 Cells as well as primary cultures of metanephric mesenchymal cells also express vascular endothelial growth factor receptor 2 and Tie-2, suggesting that the metanephric mesenchymal cells that generate epithelia may also differentiate into endothelial cells. The pluripotency of the 7.1.1 cells is self-renewing. The data suggest that the metanephric mesenchyme contains embryonic renal stem cells.


Kidney/cytology , Kidney/embryology , Mesoderm/cytology , Stem Cells/physiology , Animals , Cell Differentiation/physiology , Cell Line/physiology , Clone Cells/physiology , DNA Primers , Epithelial Cells/physiology , Female , Fibroblasts/physiology , Immunohistochemistry , Indicators and Reagents , Muscle, Smooth, Vascular/cytology , Muscle, Smooth, Vascular/physiology , Phenotype , Pregnancy , Rats , Reverse Transcriptase Polymerase Chain Reaction
...