Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
1.
Development ; 141(1): 17-27, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24284212

ABSTRACT

Forkhead transcription factors are essential for diverse processes in early embryonic development and organogenesis. Foxd1 is required during kidney development and its inactivation results in failure of nephron progenitor cell differentiation. Foxd1 is expressed in interstitial cells adjacent to nephron progenitor cells, suggesting an essential role for the progenitor cell niche in nephrogenesis. To better understand how cortical interstitial cells in general, and FOXD1 in particular, influence the progenitor cell niche, we examined the differentiation states of two progenitor cell subtypes in Foxd1(-/-) tissue. We found that although nephron progenitor cells are retained in a primitive CITED1-expressing compartment, cortical interstitial cells prematurely differentiate. To identify pathways regulated by FOXD1, we screened for target genes by comparison of Foxd1 null and wild-type tissues. We found that the gene encoding the small leucine-rich proteoglycan decorin (DCN) is repressed by FOXD1 in cortical interstitial cells, and we show that compound genetic inactivation of Dcn partially rescues the failure of progenitor cell differentiation in the Foxd1 null. We demonstrate that DCN antagonizes BMP/SMAD signaling, which is required for the transition of CITED1-expressing nephron progenitor cells to a state that is primed for WNT-induced epithelial differentiation. On the basis of these studies, we propose a mechanism for progenitor cell retention in the Foxd1 null in which misexpressed DCN produced by prematurely differentiated interstitial cells accumulates in the extracellular matrix, inhibiting BMP7-mediated transition of nephron progenitor cells to a compartment in which they can respond to epithelial induction signals.


Subject(s)
Decorin/metabolism , Forkhead Transcription Factors/metabolism , Nephrons/embryology , Organogenesis/physiology , Stem Cells/metabolism , Animals , Apoptosis Regulatory Proteins , Bone Morphogenetic Protein 7/antagonists & inhibitors , Bone Morphogenetic Protein 7/metabolism , Cell Differentiation , Cell Line , Decorin/biosynthesis , Decorin/genetics , Forkhead Transcription Factors/genetics , Gene Expression Profiling , Lymphoid Enhancer-Binding Factor 1/biosynthesis , Lymphoid Enhancer-Binding Factor 1/metabolism , Mice , Mice, Inbred ICR , Mice, Knockout , NIH 3T3 Cells , Nephrons/growth & development , Nephrons/metabolism , Nuclear Proteins/biosynthesis , Nuclear Proteins/metabolism , Signal Transduction , Smad Proteins/antagonists & inhibitors , Smad Proteins/metabolism , Trans-Activators/biosynthesis , Trans-Activators/metabolism , Transcriptome/genetics
2.
Kidney Int ; 90(2): 373-388, 2016 08.
Article in English | MEDLINE | ID: mdl-27344203

ABSTRACT

A substantial portion of the human population is affected by urogenital birth defects resulting from a failure in ureter development. Although recent research suggests roles for several genes in facilitating the ureter/bladder connection, the underlying molecular mechanisms remain poorly understood. Signaling via Eph receptor tyrosine kinases is involved in several developmental processes. Here we report that impaired Eph/Ephrin signaling in genetically modified mice results in severe hydronephrosis caused by defective ureteric bud induction, ureter maturation, and translocation. Our data imply that ureter translocation requires apoptosis in the urogenital sinus and inhibition of proliferation in the common nephric duct. These processes were disturbed in EphA4/EphB2 compound knockout mice and were accompanied by decreased ERK-2 phosphorylation. Using a set of Eph, Ephrin, and signaling-deficient mutants, we found that during urogenital development, different modes of Eph/Ephrin signaling occur at several sites with EphrinB2 and EphrinA5 acting in concert. Thus, Eph/Ephrin signaling should be considered in the etiology of congenital kidney and urinary tract anomalies.


Subject(s)
Ephrin-A5/metabolism , Ephrin-B2/metabolism , Hydronephrosis/genetics , Receptor, EphA4/metabolism , Receptor, EphB2/metabolism , Urogenital Abnormalities/genetics , Animals , Apoptosis , Humans , Hydronephrosis/metabolism , Kidney/embryology , Mice , Mice, Inbred C57BL , Mice, Knockout , Mitogen-Activated Protein Kinase 1/metabolism , Organ Culture Techniques , Organogenesis/genetics , Phosphorylation , Receptor, EphA4/genetics , Receptor, EphB2/genetics , Signal Transduction , Ureter/embryology , Urogenital Abnormalities/metabolism
3.
Proc Natl Acad Sci U S A ; 110(12): 4640-5, 2013 Mar 19.
Article in English | MEDLINE | ID: mdl-23487745

ABSTRACT

Embryonic nephron progenitor cells are segregated in molecularly distinct compartments of unknown function. Our study reveals an integral role for bone morphogenetic protein-SMAD in promoting transition of progenitors from the primitive Cbp/p300-interacting transactivator 1 expressing (CITED1+) compartment to the uniquely sine oculis-related homeobox 2 expressing (SIX2-only) compartment where they become inducible by wingless-type mouse mammary tumor virus integration site family member (WNT)/ß-catenin signaling. Significantly, CITED1(+) cells are refractory to WNT/ß-catenin induction. We propose a model in which the primitive CITED1(+) compartment is refractory to induction by WNT9b/ß-catenin, ensuring maintenance of undifferentiated progenitor cells for future nephrogenesis. Bone morphogenetic protein 7-SMAD is then required for transition to a distinct compartment in which cells become inducible by WNT9b/ß-catenin, allowing them to progress toward epithelialization.


Subject(s)
Cell Differentiation/physiology , Epithelial Cells/metabolism , Nephrons/metabolism , Stem Cells/metabolism , Wnt Signaling Pathway/physiology , Animals , Apoptosis Regulatory Proteins , Bone Morphogenetic Protein 7/genetics , Bone Morphogenetic Protein 7/metabolism , Cell Line , Epithelial Cells/cytology , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Mice , Mice, Knockout , Nephrons/cytology , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Smad Proteins/genetics , Smad Proteins/metabolism , Stem Cells/cytology , Trans-Activators/genetics , Trans-Activators/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism , Wnt Proteins/genetics , Wnt Proteins/metabolism , beta Catenin/genetics , beta Catenin/metabolism
4.
Development ; 138(23): 5099-112, 2011 Dec.
Article in English | MEDLINE | ID: mdl-22031548

ABSTRACT

Recent studies indicate that nephron progenitor cells of the embryonic kidney are arranged in a series of compartments of an increasing state of differentiation. The earliest progenitor compartment, distinguished by expression of CITED1, possesses greater capacity for renewal and differentiation than later compartments. Signaling events governing progression of nephron progenitor cells through stages of increasing differentiation are poorly understood, and their elucidation will provide key insights into normal and dysregulated nephrogenesis, as well as into regenerative processes that follow kidney injury. In this study, we found that the mouse CITED1(+) progenitor compartment is maintained in response to receptor tyrosine kinase (RTK) ligands that activate both FGF and EGF receptors. This RTK signaling function is dependent on RAS and PI3K signaling but not ERK. In vivo, RAS inactivation by expression of sprouty 1 (Spry1) in CITED1(+) nephron progenitors results in loss of characteristic molecular marker expression and in increased death of progenitor cells. Lineage tracing shows that surviving Spry1-expressing progenitor cells are impaired in their subsequent epithelial differentiation, infrequently contributing to epithelial structures. These findings demonstrate that the survival and developmental potential of cells in the earliest embryonic nephron progenitor cell compartment are dependent on FGF/EGF signaling through RAS.


Subject(s)
Cell Differentiation/physiology , Epidermal Growth Factor/metabolism , Fibroblast Growth Factors/metabolism , Nephrons/embryology , Signal Transduction/physiology , Adaptor Proteins, Signal Transducing , Animals , Apoptosis Regulatory Proteins , Cell Lineage , Cells, Cultured , Galactosides , In Situ Nick-End Labeling , Indoles , Membrane Proteins/metabolism , Mice , Microscopy, Fluorescence , Nephrons/cytology , Nuclear Proteins/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Phosphoproteins/metabolism , Polymerase Chain Reaction , Receptor Protein-Tyrosine Kinases/metabolism , Trans-Activators/metabolism , ras Proteins/metabolism
5.
Avian Dis ; 57(1): 22-8, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23678725

ABSTRACT

While real-time-polymerase chain reaction (RT PCR) has been used as a rapid test for detection of Salmonella Enteritidis in recent years, little research has been done to assess the feasibility of pooling poultry environmental samples with a Salmonella Enteritidis-specific RT PCR assay. Therefore the objective of this study was to compare RT PCR Salmonella Enteritidis detection in individual and pooled (in groups of two, three, and four) poultry environmental drag swab samples to traditional cultural methods. The drag swabs were collected from poultry facilities previously confirmed positive for Salmonella Enteritidis and were cultured according to National Poultry Improvement Plan guidelines. Initial, Salmonella Enteritidis-specific RT PCR assay threshold cycle cutoff values of < or = 36, < or = 30, and < or = 28 were evaluated in comparison to culture. The average limit of detection of the RT PCR assay was 2.4 x 10(3) colony-forming units (CFUs)/ml, which corresponded to an average threshold cycle value of 36.6. Before enrichment, samples inoculated with concentrations from 10(2) to 10(5) CFUs/ml were detected by RT PCR, while after enrichment, samples inoculated from 10(0) to 10(5) CFUs/ml were detected by RT PCR. Threshold cycle cutoff values were used in the subsequent field trial from which Salmonella Enteritidis was cultured in 7 of 208 environmental samples (3.4%). Individual samples were 99.0%, 100%, and 100% in agreement with the RT PCR at threshold cycle (C(t)) cutoff values of < or = 36, < or = 30, and < or = 28 respectively. The agreement for pooled samples also followed the same trend with highest agreement at C(t) < or = 28 (pool of 2 = 100.0%, pool of 3 = 100.0%, pool of 4 = 100.0%), midrange agreement at C(t) < or = 30 (pool of 2 = 99.0%, pool of 3 = 100.0%, pool of 4 = 100.0%), and lowest agreement at C(t) < or = 36 (pool of 2 = 98.1%, pool of 3 = 97.1%, pool of 4 = 98.1%). In conclusion, regardless of the level of pooling after tetrathionate enrichment, sensitivity was very good, and results would be comparable to what would have been found with individual culture or individual RT PCR at C(t) < or = 36.


Subject(s)
Chickens/microbiology , Colony Count, Microbial/methods , Environmental Microbiology , Real-Time Polymerase Chain Reaction/methods , Salmonella enteritidis/isolation & purification , Animals , Housing, Animal , Limit of Detection , ROC Curve , Salmonella enteritidis/genetics , Salmonella enteritidis/growth & development , Sensitivity and Specificity
6.
Development ; 136(21): 3557-66, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19793891

ABSTRACT

The iterative formation of nephrons during embryonic development relies on continual replenishment of progenitor cells throughout nephrogenesis. Defining molecular mechanisms that maintain and regulate this progenitor pool is essential to understanding nephrogenesis in developmental and regenerative contexts. Maintenance of nephron progenitors is absolutely dependent on BMP7 signaling, and Bmp7-null mice exhibit rapid loss of progenitors. However, the signal transduction machinery operating downstream of BMP7 as well as the precise target cell remain undefined. Using a novel primary progenitor isolation system, we have investigated signal transduction and biological outcomes elicited by BMP7. We find that BMP7 directly and rapidly activates JNK signaling in nephron progenitors resulting in phosphorylation of Jun and ATF2 transcription factors. This signaling results in the accumulation of cyclin D3 and subsequent proliferation of PAX2(+) progenitors, inversely correlating with the loss of nephron progenitors seen in the Bmp7-null kidney. Activation of Jun and ATF2 is severely diminished in Bmp7-null kidneys, providing an important in vivo correlate. BMP7 thus promotes proliferation directly in nephron progenitors by activating the JNK signaling circuitry.


Subject(s)
Bone Morphogenetic Protein 7/metabolism , Kidney/embryology , Nephrons/cytology , Stem Cells/cytology , Activating Transcription Factor 2 , Animals , Cell Proliferation , Kidney/cytology , MAP Kinase Kinase 4/metabolism , Mesoderm/metabolism , Mice , PAX2 Transcription Factor/metabolism , Smad Proteins/metabolism
7.
J Med Entomol ; 59(1): 380-383, 2022 01 12.
Article in English | MEDLINE | ID: mdl-34642751

ABSTRACT

Sex is a factor influencing development in many insect species, but is not widely studied in forensically important blow flies. If sex influences blow fly development, knowing the sex of a larva from a corpse can increase precision in estimates of that larva's age. The improved prediction of larval age will make estimates of time since death using entomological evidence better. Larvae lack sexually dimorphic morphological characteristics, so sex is not immediately known visually. To generate sexually dimorphic reference growth curves, a subsample must be large enough to ensure enough males and females are present for comparison. Using two entire age Chrysomya megacephala (Fabricius) cohorts, we evaluated the minimum sample number needed to have enough individuals of both sexes for comparison using 95% prediction intervals. Through a simulation of three trials of 1000 random replicates, we determined that a sample size of 19 would prevent any instance of a comparison not occurring because of insufficient sampling from one sex. As the current method for molecular sex determination can be expensive, we also compared how the results of various subsampling percentages compare those of the entire age cohorts. We found that subsampling at least 50% of an entire cohort leads to almost identical results in comparison to the entire age cohort. Together, these findings will help establish guidelines for generating sex-specific reference growth curves. A uniform approach to generating these sex-specific growth curves will lead to more consistency in age estimates made from them.


Subject(s)
Calliphoridae/growth & development , Forensic Entomology/methods , Animals , Larva/growth & development , Sex Factors
8.
Am J Physiol Renal Physiol ; 299(6): F1320-7, 2010 Dec.
Article in English | MEDLINE | ID: mdl-20861081

ABSTRACT

Follistatin-like 1 (FSTL1) is a secreted protein with homology to both Follistatin and the SPARC/BM40 family of matricellular proteins. In this study, we sought to determine the expression patterns of Fstl1 and its cognate receptor Dip2a in the adult, and to assess the consequences of Fstl1 inactivation on development and homeostasis of the kidney. We find that FSTL1 circulates at high levels in both the human and the mouse and that it is also locally expressed in the loop of Henle in the kidney. To begin to understand the in vivo functions of Fstl1, we generated a mouse mutant using a genetrap approach. The hypomorphic Fstl1 genetrap strain displays a strong reduction in FSTL1 expression at the protein level, but it does not show overt developmental defects. FSTL1 has previously been implicated in diverse disease processes as a regulator of inflammatory cytokine expression, and we therefore evaluated the response of the genetrap strain to cisplatin-mediated acute kidney injury, a disease model with highly cytokine-dependent pathology. We find that although TNF-α and Il6 levels are unchanged relative to wild-type, renal Il-1ß expression is increased in genetrap mice following cisplatin treatment. Furthermore, histopatological analysis, expression of the tissue injury marker Havcr1, and measurement of serum creatinine demonstrate that reduction of Fstl1 expression sensitizes the kidney to acute cisplatin nephrotoxicity, suggesting a role for FSTL1-mediated Il-1ß suppression in protection of the kidney from acute nephrotoxic injury.


Subject(s)
Acute Kidney Injury/metabolism , Follistatin-Related Proteins/physiology , Interleukin-1beta/biosynthesis , Kidney/metabolism , Acute Kidney Injury/chemically induced , Adult , Animals , Cisplatin/toxicity , Creatinine/blood , Follistatin-Related Proteins/genetics , HEK293 Cells , Hepatitis A Virus Cellular Receptor 1 , Humans , Kidney/drug effects , Membrane Proteins/biosynthesis , Mice , Nerve Tissue Proteins/biosynthesis , Nuclear Proteins , Receptors, Cell Surface/biosynthesis
9.
J Am Soc Nephrol ; 20(5): 1020-31, 2009 May.
Article in English | MEDLINE | ID: mdl-19357253

ABSTRACT

Stimulation of the bone morphogenetic protein (BMP) pathway protects the kidney from acute and chronic injury. Numerous regulators in the kidney control BMP signaling, offering many targets for therapeutic manipulation. Here, we screened for modulators of BMP signaling in the ischemia-sensitive S3 segment and found that Chordin-like 1 is expressed in this segment of both the mouse and human nephron. Chordin-like 1 specifically antagonizes BMP7, which is expressed in the neighboring distal nephron, and this depends on the presence of the protein Twisted gastrulation. Upon ischemia-induced degeneration of the S3 segment, we observed a reduction in Chordin-like 1 expression coincident with intense BMP signaling in tubules of the recovering kidney. Restored expression accompanied proximal tubule epithelia redifferentiation, again coincident with decreased BMP signaling. We propose that Chordin-like 1 reduces BMP7 signaling in healthy proximal tubules, and the loss of this activity upon sloughing of injured epithelia promotes BMP7 signaling in repopulating, dedifferentiated epithelia. As regenerating epithelia differentiate, Chordin-like 1 is again expressed, antagonizing BMP7. These data suggest a mechanism for dynamic regulation of renoprotective BMP7 signaling in the S3 segment of the proximal tubule.


Subject(s)
Bone Morphogenetic Protein 7/physiology , Eye Proteins/genetics , Ischemia/physiopathology , Kidney Tubules, Proximal/physiology , Kidney/injuries , Nerve Tissue Proteins/genetics , Proteins/physiology , Reperfusion Injury/physiopathology , Animals , Eye Proteins/physiology , Gene Expression Regulation , Humans , Kidney Medulla/physiology , Kidney Medulla/physiopathology , Kidney Tubules/injuries , Kidney Tubules/physiology , Kidney Tubules, Proximal/injuries , Mice , Nerve Tissue Proteins/physiology , Proteins/genetics , Regeneration
10.
Am J Physiol Renal Physiol ; 297(3): F809-15, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19535568

ABSTRACT

Long-term pulse chase experiments previously identified a sizable population of BrdU-retaining cells within the renal papilla. The origin of these cells has been unclear, and in this work we test the hypothesis that they become quiescent early during the course of kidney development and organ growth. Indeed, we find that BrdU-retaining cells of the papilla can be labeled only by pulsing with BrdU from embryonic (E) day 11.25 to postnatal (P) day 7, the approximate period of kidney development in the mouse. BrdU signal in the cortex and outer medulla is rapidly diluted by cellular proliferation during embryonic development and juvenile growth, whereas cells within the papilla differentiate and exit the cell cycle during organogenesis. Indeed, by E17.5, little or no active proliferation can be seen in the distal papilla, indicating maturation of this structure in a distal-to-proximal manner during organogenesis. We conclude that BrdU-retaining cells of the papilla represent a population of cells that quiesce during embryonic development and localize within a region of the kidney that matures early. We therefore propose that selective papillary retention of BrdU arises through a combination of regionalized slowing of, and exit from, the cell cycle within the papilla during the period of ongoing kidney development, and extensive proliferative growth of the juvenile kidney resulting in dilution of BrdU below the detection level in extra-papillary regions.


Subject(s)
Bromodeoxyuridine/administration & dosage , Cell Proliferation , Kidney Cortex/cytology , Kidney Medulla/cytology , Staining and Labeling/methods , Animals , Animals, Newborn , Cell Cycle , Cell Differentiation , Female , Gestational Age , Injections, Intraperitoneal , Kidney Cortex/embryology , Kidney Cortex/growth & development , Kidney Medulla/embryology , Kidney Medulla/growth & development , Mice , Mice, Inbred ICR , Pregnancy
11.
Dev Biol ; 311(2): 679-90, 2007 Nov 15.
Article in English | MEDLINE | ID: mdl-17936743

ABSTRACT

Bmp7 is expressed in numerous tissues throughout development and is required for morphogenesis of the eye, hindlimb and kidney. In this study we show that the majority if not all of the cis-regulatory sequence governing expression at these anatomical sites during development is present in approximately 20 kb surrounding exon 1. In eye, limb and kidney, multiple distinct enhancer elements drive Bmp7 expression within each organ. In the eye, the elements driving expression in the pigmented epithelium and iris are spatially separated. In the kidney, Bmp7 expression in collecting ducts and nephron progenitors is driven by separate enhancer elements. Similarly, limb mesenchyme and apical ectodermal ridge expression are governed by separate elements. Although enhancers for pigmented epithelium, nephrogenic mesenchyme and apical ectodermal ridge are distributed across the approximately 20 kb region, an element of approximately 480 base pairs within intron 1 governs expression within the developing iris, collecting duct system of the kidney and limb mesenchyme. This element is remarkably conserved both in sequence and position in the Bmp7 locus between different vertebrates, ranging from Xenopus tropicalis to Homo sapiens, demonstrating that there is strong selective pressure for Bmp7 expression at these tissue sites. Furthermore, we show that the frog enhancer functions appropriately in transgenic mice. Interestingly, the intron 1 element cannot be found in the Bmp7 genes of vertebrates such as Danio rerio and Takifugu rubripes indicating that this modification of the Bmp7 gene might have arisen during the adaptation from aquatic to terrestrial life. Mutational analysis demonstrates that the enhancer activity of the intron 1 element is entirely dependent on the presence of a 10 base pair site within the intron 1 enhancer containing a predicted binding site for the FOXD3 transcription factor.


Subject(s)
Bone Morphogenetic Proteins , Enhancer Elements, Genetic , Extremities , Eye , Kidney , Transforming Growth Factor beta , Animals , Base Sequence , Bone Morphogenetic Protein 7 , Bone Morphogenetic Proteins/genetics , Bone Morphogenetic Proteins/metabolism , DNA Mutational Analysis , Exons , Extremities/anatomy & histology , Extremities/embryology , Eye/anatomy & histology , Eye/embryology , Eye/metabolism , Forkhead Transcription Factors/genetics , Forkhead Transcription Factors/metabolism , Gene Expression Regulation, Developmental , Genes, Reporter , Humans , Introns , Kidney/anatomy & histology , Kidney/embryology , Kidney/metabolism , Mice , Mice, Inbred CBA , Mice, Transgenic , Molecular Sequence Data , Morphogenesis , Mutagenesis, Site-Directed , Repressor Proteins/genetics , Repressor Proteins/metabolism , Sequence Alignment , Sequence Homology, Nucleic Acid , Transforming Growth Factor beta/genetics , Transforming Growth Factor beta/metabolism , Xenopus Proteins
12.
BMC Dev Biol ; 8: 86, 2008 Sep 18.
Article in English | MEDLINE | ID: mdl-18801194

ABSTRACT

BACKGROUND: Bone morphogenetic proteins (BMPs) regulate essential processes during organogenesis, and a functional understanding of these secreted proteins depends on identification of their target cells. In this study, we generate a transgenic reporter for organogenesis studies that we use to define BMP pathway activation in the developing kidney. RESULTS: Mouse strains reporting on BMP pathway activation were generated by transgenically expressing beta-galactosidase under the control of BMP responsive elements from Id1. Reporter expression corresponds well with immunoassays for pathway activation in all organs studied, validating the model. Using these reporters we have generated a detailed map of cellular targets of BMP signaling in the developing kidney. We find that SMAD dependent BMP signaling is active in collecting duct trunks, but not tips. Furthermore, glomerular endothelial cells, and proximal nephron tubules from the renal vesicle stage onward show pathway activation. Surprisingly, little activation is detected in the nephrogenic zone of the kidney, and in organ culture BMP treatment fails to activate SMAD dependent BMP signaling in nephron progenitor cells. In contrast, signaling is efficiently induced in collecting duct tips. CONCLUSION: Transgenic reporters driven by control elements from BMP responsive genes such as Id1 offer significant advantages in sensitivity and consistency over immunostaining for studies of BMP pathway activation. They also provide opportunities for analysis of BMP signaling in organ and primary cell cultures subjected to experimental manipulation. Using such a reporter, we made the surprising finding that SMAD dependent BMP signaling is inactive in nephron progenitors, and that these cells are refractory to activation by applied growth factors. Furthermore, we find that the BMP pathway is not normally active in collecting duct tips, but that it can be ectopically activated by BMP treatment, offering a possible explanation for the inhibitory effects of BMP treatment on collecting duct growth and branching.


Subject(s)
Bone Morphogenetic Proteins/physiology , Gene Expression Regulation, Developmental/physiology , Gene Targeting , Genes, Reporter/physiology , Kidney/embryology , Organogenesis/physiology , Signal Transduction/physiology , Animals , Female , Kidney/metabolism , Mice , Mice, Inbred C57BL , Mice, Inbred ICR , Mice, Transgenic , Smad1 Protein/biosynthesis , Smad1 Protein/genetics , Smad5 Protein/biosynthesis , Smad5 Protein/genetics , Smad8 Protein/biosynthesis , Smad8 Protein/genetics , beta-Galactosidase/biosynthesis , beta-Galactosidase/genetics
13.
Biotechniques ; 44(4): 547-9, 2008 Apr.
Article in English | MEDLINE | ID: mdl-18476819

ABSTRACT

Cellular interactions in development of the kidney are used as a model of reciprocal inductive events between epithelium and mesenchyme. Time- and labor-intensive methods have been developed to study this phenomenon. For example, in mice, the targeted disruption of genes in vivo has been used to modify the genetic program directing kidney development. However, gene targeting is a resource-intensive approach and alternative strategies for gene and protein modification in the kidney need to be developed. Herein, we have developed an efficient system for the delivery of antisense morpholino to alter normal protein expression. We describe the use of Endo-Porter to effectively deliver morpholinos to all parts and regions of the kidney explant. Also, we definitively show via confocal microscopy and Western blot analysis that the use of Endo-Porter in delivering antisense morpholinos is robust throughout the entire kidney explant, providing efficient suppression of protein expression. This method saves time and cost when compared with targeted disruption and is an improvement upon previous kidney organ culture methods.


Subject(s)
Drug Delivery Systems , Kidney/cytology , Peptides , Animals , Fluorescein/metabolism , Fluorescent Dyes/metabolism , Mice , Mice, Inbred ICR , Models, Biological , Neoplasm Proteins/antagonists & inhibitors , Oligonucleotides, Antisense/chemistry , Organ Culture Techniques , RNA, Messenger/metabolism , Time Factors
14.
Gene Expr Patterns ; 7(4): 491-500, 2007 Feb.
Article in English | MEDLINE | ID: mdl-17129766

ABSTRACT

Follistatin-like 1 (Fstl1) is a distantly related homolog of the Activin and Bone Morphogenetic Protein antagonist Follistatin. Interestingly, this molecule also has homology with the extracellular matrix modifying protein BM-40/SPARC/osteonectin. Previous studies in chick have identified Fstl1 as a regulator of early mesoderm patterning, somitogenesis, myogenesis and neural development. In this study, we determine the developmental expression pattern of Fstl1 in the mouse. We find that Fstl1 is ubiquitously expressed in the early embryo, and that expression becomes regionalized later during development. In the majority of tissues, Fstl1 is strongly expressed in the mesenchymal component and excluded from the epithelium. Notable exceptions include the central nervous system, in which Fstl1 expression is entirely absent with the exception of the choroid plexi and floor plate, the lung, in which Fstl1 expression can be seen in airway epithelia and the kidney, in which collecting ducts and nascent nephron epithelia express the highest levels of Fstl1.


Subject(s)
Follistatin-Related Proteins/genetics , Gene Expression Regulation, Developmental , Kidney/embryology , Lung/embryology , Organogenesis , Amino Acid Sequence , Animals , Embryo, Mammalian/metabolism , Embryonic Development , Extremities/embryology , Gastrointestinal Tract/embryology , Gonads/embryology , In Situ Hybridization , Kidney/metabolism , Lung/metabolism , Mice , Mice, Inbred ICR , Molecular Sequence Data , Nervous System/embryology , Nervous System/metabolism
15.
J Vis Exp ; (50)2011 Apr 22.
Article in English | MEDLINE | ID: mdl-21540822

ABSTRACT

Embryonic development of the kidney has been extensively studied both as a model for epithelial-mesenchymal interaction in organogenesis and to gain understanding of the origins of congenital kidney disease. More recently, the possibility of steering naïve embryonic stem cells toward nephrogenic fates has been explored in the emerging field of regenerative medicine. Genetic studies in the mouse have identified several pathways required for kidney development, and a global catalog of gene transcription in the organ has recently been generated http://www.gudmap.org/, providing numerous candidate regulators of essential developmental functions. Organogenesis of the rodent kidney can be studied in organ culture, and many reports have used this approach to analyze outcomes of either applying candidate proteins or knocking down the expression of candidate genes using siRNA or morpholinos. However, the applicability of organ culture to the study of signaling that regulates stem/progenitor cell differentiation versus renewal in the developing kidney is limited as cultured organs contain a compact extracellular matrix limiting diffusion of macromolecules and virus particles. To study the cell signaling events that influence the stem/progenitor cell niche in the kidney we have developed a primary cell system that establishes the nephrogenic zone or progenitor cell niche of the developing kidney ex vivo in isolation from the epithelial inducer of differentiation. Using limited enzymatic digestion, nephrogenic zone cells can be selectively liberated from developing kidneys at E17.5. Following filtration, these cells can be cultured as an irregular monolayer using optimized conditions. Marker gene analysis demonstrates that these cultures contain a distribution of cell types characteristic of the nephrogenic zone in vivo, and that they maintain appropriate marker gene expression during the culture period. These cells are highly accessible to small molecule and recombinant protein treatment, and importantly also to viral transduction, which greatly facilitates the study of candidate stem/progenitor cell regulator effects. Basic cell biological parameters such as proliferation and cell death as well as changes in expression of molecular markers characteristic of nephron stem/progenitor cells in vivo can be successfully used as experimental outcomes. Ongoing work in our laboratory using this novel primary cell technique aims to uncover basic mechanisms governing the regulation of self-renewal versus differentiation in nephron stem/progenitor cells.


Subject(s)
Cytological Techniques/methods , Embryonic Stem Cells/cytology , Kidney/cytology , Kidney/embryology , Animals , Embryo, Mammalian/cytology , Mice
16.
Trends Biotechnol ; 27(11): 612-4, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19765844

ABSTRACT

Research and development connect technology and innovation to product design. However, the term is often used to refer to only a subset of the necessary disciplines to the exclusion of technical operations. Here, we argue that the importance of technical operations is undeniable, offering possible solutions by drawing on lessons from outdated biotherapeutics production methods and highlighting advances in the field.


Subject(s)
Biomedical Technology/methods , Technology, Pharmaceutical/methods , Animals , Biomedical Technology/economics , Biomedical Technology/legislation & jurisprudence , Quality Control , Research Design/legislation & jurisprudence , Technology, Pharmaceutical/economics , Technology, Pharmaceutical/legislation & jurisprudence
17.
Mech Dev ; 126(5-6): 337-49, 2009.
Article in English | MEDLINE | ID: mdl-19268530

ABSTRACT

Branching morphogenesis is a developmental process characteristic of many organ systems. Specifically, during renal branching morphogenesis, its been postulated that the final number of nephrons formed is one key clinical factor in the development of hypertension in adulthood. As it has been established that BMPs regulate, in part, renal activity of p38 MAP kinase (p38(MAPK)) and it has demonstrated that the cytoplasmic protein Neurotrophin Receptor MAGE homologue (NRAGE) augments p38(MAPK) activation, it was hypothesized that a decrease in the expression of NRAGE during renal branching would result in decreased branching of the UB that correlated with changes in p38(MAPK) activation. To verify this, the expression of NRAGE was reduced in ex vivo kidney explants cultures using antisense morpholino. Morpholino treated ex vivo kidney explants expression were severely stunted in branching, a trait that was rescued with the addition of exogenous GDNF. Renal explants also demonstrated a precipitous drop in p38(MAPK) activation that too was reversed in the presence of recombinant GDNF. RNA profiling of NRAGE diminished ex vivo kidney explants resulted in altered expression of GDNF, Ret, BMP7 and BMPRIb mRNAs. Our results suggested that in early kidney development NRAGE might have multiple roles during renal branching morphogenesis through association with both the BMP and GDNF signaling pathways.


Subject(s)
Kidney/embryology , Morphogenesis , Neoplasm Proteins/metabolism , Animals , Apoptosis/drug effects , Bone Morphogenetic Proteins/metabolism , Cell Proliferation/drug effects , Enzyme Activation/drug effects , Gene Expression Regulation, Developmental/drug effects , Glial Cell Line-Derived Neurotrophic Factor/pharmacology , Homeodomain Proteins/metabolism , Immunoprecipitation , Kidney/cytology , Kidney/drug effects , Kidney/enzymology , Mice , Mice, Transgenic , Models, Biological , Morphogenesis/drug effects , Oligonucleotides, Antisense/pharmacology , Organ Culture Techniques , Phosphorylation/drug effects , Signal Transduction/drug effects , Ureter/drug effects , Ureter/embryology , Ureter/enzymology , p38 Mitogen-Activated Protein Kinases/metabolism
18.
J Neurobiol ; 62(1): 134-47, 2005 Jan.
Article in English | MEDLINE | ID: mdl-15452851

ABSTRACT

Growth cone navigation is guided by extrinsic environmental proteins, called guidance cues. Many in vitro studies have characterized growth cone turning up and down gradients of soluble guidance cues. Although previous studies have shown that axonal elongation rates can be regulated by gradients of surface-bound molecules, there are no convincing demonstrations of growth cones turning to migrate up a surface-bound gradient of an adhesive ligand or guidance cue. In order to test this mode of axonal guidance, we used a photo-immobilization technique to create grids and gradients of an adhesive laminin peptide on polystyrene culture dish surfaces. Chick embryo dorsal root ganglia (DRGs) were placed on peptide grid patterns containing surface-bound gradients of the IKVAV-containing peptide. DRG growth cones followed a path of surface-bound peptide to the middle of a perpendicularly oriented gradient with a 25% concentration difference across 30 microm. The majority of growth cones turned and migrated up the gradient, turning until they were oriented directly up the gradient. Growth cones slowed their migration when they encountered the gradient, but growth cone velocity returned to the previous rate after turning up or down the gradient. This resembles in vivo situations where growth cones slow at a choice point before changing the direction of axonal extension. Thus, these results support the hypothesis that mechanisms of axonal guidance include growth cone orientation by gradients of surface-bound adhesive molecules and guidance cues.


Subject(s)
Cell Differentiation/physiology , Chemotaxis/physiology , Ganglia, Spinal/embryology , Growth Cones/metabolism , Laminin/metabolism , Animals , Cell Adhesion/drug effects , Cell Adhesion/physiology , Cell Communication/physiology , Cell Differentiation/drug effects , Chemotactic Factors/metabolism , Chemotactic Factors/pharmacology , Chemotaxis/drug effects , Chick Embryo , Cues , Ganglia, Spinal/cytology , Ganglia, Spinal/metabolism , Growth Cones/drug effects , Growth Cones/ultrastructure , Laminin/pharmacology , Lasers , Nerve Growth Factors/metabolism , Nerve Growth Factors/pharmacology , Neural Cell Adhesion Molecules/metabolism , Peptides/metabolism , Peptides/pharmacology , Photic Stimulation
SELECTION OF CITATIONS
SEARCH DETAIL