Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 27
Filter
Add more filters










Publication year range
1.
Ecol Evol ; 14(5): e11383, 2024 May.
Article in English | MEDLINE | ID: mdl-38803606

ABSTRACT

Predator non-consumptive effects (NCE) can alter prey foraging time and habitat use, potentially reducing fitness. Prey can mitigate NCEs by increasing vigilance, chewing-vigilance synchronization, and spatiotemporal avoidance of predators. We quantified the relationship between Mexican wolf (Canis lupus baileyi) predation risk and elk (Cervus canadensis) behavior. We conducted behavioral observations on adult female elk and developed predation risk indices using GPS collar data from Mexican wolves, locations of elk killed by wolves, and landscape covariates. We compared a priori models to determine the best predictors of adult female behavior and multitasking. Metrics that quantified both spatial and temporal predation risk were the most predictive. Vigilance was positively associated with increased predation risk. The effect of predation risk on foraging and resting differed across diurnal periods. During midday when wolf activity was lower, the probability of foraging increased while resting decreased in high-risk areas. During crepuscular periods when elk and wolves were most active, increased predation risk was associated with increased vigilance and slight decreases in foraging. Our results suggest elk are temporally avoiding predation risk from Mexican wolves by trading resting for foraging, a trade-off often not evaluated in behavioral studies. Probability of multitasking depended on canopy openness and an interaction between maternal period and predation risk; multitasking decreased prior to parturition and increased post parturition in high-risk areas. Openness was inversely related to multitasking. These results suggest adult female elk are altering the type of vigilance used depending on resource availability/quality, current energetic needs, and predation risk. Our results highlight potentially important, but often-excluded behaviors and trade-offs prey species may use to reduce the indirect effects of predation and contribute additional context to our understanding of predator-prey dynamics.

3.
Mol Cell ; 59(4): 685-97, 2015 Aug 20.
Article in English | MEDLINE | ID: mdl-26257285

ABSTRACT

We developed Split DamID (SpDamID), a protein complementation version of DamID, to mark genomic DNA bound in vivo by interacting or juxtapositioned transcription factors. Inactive halves of DAM (DNA adenine methyltransferase) were fused to protein pairs to be queried. Either direct interaction between proteins or proximity enabled DAM reconstitution and methylation of adenine in GATC. Inducible SpDamID was used to analyze Notch-mediated transcriptional activation. We demonstrate that Notch complexes label RBP sites broadly across the genome and show that a subset of these complexes that recruit MAML and p300 undergo changes in chromatin accessibility in response to Notch signaling. SpDamID differentiates between monomeric and dimeric binding, thereby allowing for identification of half-site motifs used by Notch dimers. Motif enrichment of Notch enhancers coupled with SpDamID reveals co-targeting of regulatory sequences by Notch and Runx1. SpDamID represents a sensitive and powerful tool that enables dynamic analysis of combinatorial protein-DNA transactions at a genome-wide level.


Subject(s)
DNA/genetics , Molecular Probe Techniques , Receptors, Notch/physiology , Animals , Base Sequence , Binding Sites , Cell Line , DNA/metabolism , Enhancer Elements, Genetic , Mice, Transgenic , Molecular Sequence Data , Protein Binding
4.
Development ; 142(6): 1193-202, 2015 Mar 15.
Article in English | MEDLINE | ID: mdl-25725069

ABSTRACT

We have previously described the creation and analysis of a Notch1 activity-trap mouse line, Notch1 intramembrane proteolysis-Cre6MT or N1IP::Cre(LO), that marked cells experiencing relatively high levels of Notch1 activation. Here, we report and characterize a second line with improved sensitivity (N1IP::Cre(HI)) to mark cells experiencing lower levels of Notch1 activation. This improvement was achieved by increasing transcript stability and by restoring the native carboxy terminus of Cre, resulting in a five- to tenfold increase in Cre activity. The magnitude of this effect probably impacts Cre activity in strains with carboxy-terminal Ert2 fusion. These two trap lines and the related line N1IP::Cre(ERT2) form a complementary mapping tool kit to identify changes in Notch1 activation patterns in vivo as the consequence of genetic or pharmaceutical intervention, and illustrate the variation in Notch1 signal strength from one tissue to the next and across developmental time.


Subject(s)
Cell Line/physiology , Receptor, Notch1/metabolism , Signal Transduction/physiology , Animals , Blotting, Western , Cell Line/metabolism , Fibroblasts , Galactosides , Gene Knock-In Techniques , Immunohistochemistry , Indoles , Integrases/genetics , Integrases/metabolism , Mice , Receptor, Notch1/genetics , Transfection
5.
Cytoskeleton (Hoboken) ; 71(3): 195-209, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24520051

ABSTRACT

The Abelson (Abl) non-receptor tyrosine kinase regulates the cytoskeleton during multiple stages of neural development, from neurulation, to the articulation of axons and dendrites, to synapse formation and maintenance. We previously showed that Abl is genetically linked to the microtubule (MT) plus end tracking protein (+TIP) CLASP in Drosophila. Here we show in vertebrate cells that Abl binds to CLASP and phosphorylates it in response to serum or PDGF stimulation. In vitro, Abl phosphorylates CLASP with a Km of 1.89 µM, indicating that CLASP is a bona fide substrate. Abl-phosphorylated tyrosine residues that we detect in CLASP by mass spectrometry lie within previously mapped F-actin and MT plus end interaction domains. Using purified proteins, we find that Abl phosphorylation modulates direct binding between purified CLASP2 with both MTs and actin. Consistent with these observations, Abl-induced phosphorylation of CLASP2 modulates its localization as well as the distribution of F-actin structures in spinal cord growth cones. Our data suggest that the functional relationship between Abl and CLASP2 is conserved and provides a means to control the CLASP2 association with the cytoskeleton.


Subject(s)
Actins/metabolism , Microtubule-Associated Proteins/metabolism , Microtubules/metabolism , Proto-Oncogene Proteins c-abl/metabolism , Actin Cytoskeleton/drug effects , Actin Cytoskeleton/metabolism , Amino Acid Sequence , Animals , COS Cells , Cell Adhesion/drug effects , Chlorocebus aethiops , Growth Cones/drug effects , Growth Cones/metabolism , HEK293 Cells , Humans , Microtubule-Associated Proteins/chemistry , Microtubules/drug effects , Molecular Sequence Data , Phosphorylation/drug effects , Phosphotyrosine/metabolism , Platelet-Derived Growth Factor/pharmacology , Protein Binding/drug effects , Signal Transduction/drug effects , Subcellular Fractions/drug effects , Subcellular Fractions/metabolism , Substrate Specificity/drug effects , Xenopus
6.
Development ; 141(2): 346-54, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24353058

ABSTRACT

Mesangial cells are specialized pericyte/smooth muscle cells that surround and constrain the vascular network within the glomerulus of the kidney. They are derived from the stromal mesenchyme, a progenitor population distinct from nephron stem cells. Whether mesangial cells have a distinct origin from vascular smooth muscle cells (VSMCs) and the pathways that govern their specification are unknown. Here we show that Notch signaling in stromal progenitors is essential for mesangial cell formation but is dispensable for the smooth muscle and interstitial cell lineages. Deletion of RBPjk, the common DNA-binding partner of all active Notch receptors, with Foxd1(tgCre) results in glomerular aneurysm and perinatal death from kidney failure. This defect occurs early in glomerular development as stromal-derived, desmin-positive cells fail to coalesce near forming nephrons and thus do not invade the vascular cleft of the S-shaped body. This is in contrast to other mutants in which the loss of the mesangium was due to migration defects, and suggests that loss of Notch signaling results in a failure to specify this population from the stroma. Interestingly, Pdgfrb-positive VSMCs do not enter the vascular cleft and cannot rescue the mesangial deficiency. Notch1 and Notch2 act redundantly through γ-secretase and RBPjk in this process, as individual mutants have mesangial cells at birth. Together, these data demonstrate a unique origin of mesangial cells and demonstrate a novel, redundant function for Notch receptors in mesangial cell specification, proliferation or survival during kidney development.


Subject(s)
Embryonic Stem Cells/cytology , Embryonic Stem Cells/metabolism , Kidney/embryology , Kidney/metabolism , Mesangial Cells/cytology , Mesangial Cells/metabolism , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/metabolism , Receptor, Notch1/metabolism , Receptor, Notch2/metabolism , Amyloid Precursor Protein Secretases/metabolism , Animals , Cell Differentiation , Cell Lineage , Female , Forkhead Transcription Factors/metabolism , Immunoglobulin J Recombination Signal Sequence-Binding Protein/metabolism , Kidney/cytology , Mice , Mice, Mutant Strains , Mice, Transgenic , Myocytes, Smooth Muscle/cytology , Myocytes, Smooth Muscle/metabolism , Pregnancy , Receptor, Notch1/genetics , Receptor, Notch2/genetics , Receptor, Platelet-Derived Growth Factor beta/metabolism , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Signal Transduction
7.
Dev Cell ; 25(6): 585-98, 2013 Jun 24.
Article in English | MEDLINE | ID: mdl-23806616

ABSTRACT

Notch2, but not Notch1, plays indispensable roles in kidney organogenesis, and Notch2 haploinsufficiency is associated with Alagille syndrome. We proposed that proximal nephron fates are regulated by a threshold that requires nearly all available free Notch intracellular domains (NICDs) but could not identify the mechanism that explains why Notch2 (N2) is more important than Notch1 (N1). By generating mice that swap their ICDs, we establish that the overall protein concentration, expression domain, or ICD amino acid composition does not account for the differential requirement of these receptors. Instead, we find that the N2 extracellular domain (NECD) increases Notch protein localization to the cell surface during kidney development and is cleaved more efficiently upon ligand binding. This context-specific asymmetry in NICD release efficiency is further enhanced by Fringe. Our results indicate that an elevated N1 surface level could compensate for the loss of N2 signal in specific cell contexts.


Subject(s)
Alagille Syndrome/metabolism , Kidney/embryology , Kidney/metabolism , Membrane Proteins/metabolism , Receptor, Notch2/metabolism , Alagille Syndrome/genetics , Alagille Syndrome/pathology , Animals , Epithelial Cells/cytology , Epithelial Cells/physiology , Extracellular Space/metabolism , Gene Expression Regulation, Developmental , Gene Knock-In Techniques , HEK293 Cells , Humans , Kidney/cytology , Ligands , Membrane Proteins/chemistry , Membrane Proteins/genetics , Mice , Mice, Mutant Strains , Mutant Chimeric Proteins/chemistry , Mutant Chimeric Proteins/genetics , Mutant Chimeric Proteins/metabolism , Nephrons/cytology , Nephrons/embryology , Nephrons/metabolism , Podocytes/cytology , Podocytes/physiology , Protein Structure, Tertiary/physiology , Receptor, Notch1/chemistry , Receptor, Notch1/genetics , Receptor, Notch1/metabolism , Receptor, Notch2/chemistry , Receptor, Notch2/genetics
8.
Adv Exp Med Biol ; 727: 99-113, 2012.
Article in English | MEDLINE | ID: mdl-22399342

ABSTRACT

The kidney is the body's filter, responsible for the removal of metabolic waste and the excretion or reabsorption of electrolytes to control blood composition and pH balance. The functional unit of this filter is the nephron, whose segmented architecture has been largely conserved in form and function throughout eukaryotic evolution. Not surprisingly, the core developmental pathways that regulate the formation of the nephron have also been conserved. In particular, the Notch signaling pathway functions in both primitive and advanced nephrons to pattern domains required for the kidney's diverse functions. In this chapter, we will discuss the role that Notch plays in directing cell fate decisions during embryonic development of the pronephros and metanephros. We will go on to discuss the later role of Notch signaling as a cyst-suppressor and the consequences of aberrant or absent Notch activity in disease and cancer. The work discussed here highlights the fundamental importance of Notch during development and homeostasis of the kidney and underlies the need for mechanistic understanding of its role towards the treatment of human disease.


Subject(s)
Kidney Diseases/metabolism , Kidney Diseases/pathology , Kidney/cytology , Kidney/metabolism , Receptors, Notch/metabolism , Signal Transduction , Animals , Humans
9.
Neoplasia ; 14(12): 1153-63, 2012 Dec.
Article in English | MEDLINE | ID: mdl-23308048

ABSTRACT

Hepatoblastoma, the most common pediatric liver cancer, consists of epithelial mixed embryonal/fetal (EMEF) and pure fetal histologic subtypes, with the latter exhibiting a more favorable prognosis. Few embryonal histology markers that yield insight into the biologic basis for this prognostic discrepancy exist. CBP/P-300 interacting transactivator 1 (CITED1), a transcriptional co-activator, is expressed in the self-renewing nephron progenitor population of the developing kidney and broadly in its malignant analog, Wilms tumor (WT). In this current study, CITED1 expression is detected in mouse embryonic liver initially on post-coitum day 10.5 (e10.5), begins to taper by e14.5, and is undetectable in e18.5 and adult livers. CITED1 expression is detected in regenerating murine hepatocytes following liver injury by partial hepatectomy and 3,5-diethoxycarbonyl-1,4-dihydrocollidine. Importantly, while CITED1 is undetectable in normal human adult livers, 36 of 41 (87.8%) hepatoblastoma specimens express CITED1, where it is enriched in EMEF specimens compared to specimens of pure fetal histology. CITED1 overexpression in Hep293TT human hepatoblastoma cells induces cellular proliferation and upregulates the Wnt inhibitors Kringle containing transmembrane protein 1 (KREMEN1) and CXXC finger protein 4 (CXXC4). CITED1 mRNA expression correlates with expression of CXXC4 and KREMEN1 in clinical hepatoblastoma specimens. These data show that CITED1 is expressed during a defined time course of liver development and is no longer expressed in the adult liver but is upregulated in regenerating hepatocytes following liver injury. Moreover, as in WT, this embryonic marker is reexpressed in hepatoblastoma and correlates with embryonal histology. These findings identify CITED1 as a novel marker of hepatic progenitor cells that is re-expressed following liver injury and in embryonic liver tumors.


Subject(s)
Hepatoblastoma/metabolism , Liver Neoplasms/metabolism , Liver Regeneration , Liver/metabolism , Nuclear Proteins/metabolism , Trans-Activators/metabolism , Transcription Factors/metabolism , Wnt Signaling Pathway , Animals , Apoptosis Regulatory Proteins , Cell Line, Tumor , Cell Proliferation , DNA-Binding Proteins/metabolism , Embryo, Mammalian/metabolism , Hepatoblastoma/genetics , Humans , Liver/injuries , Liver Neoplasms/genetics , Membrane Proteins/metabolism , Mice , Mutation , RNA, Messenger/metabolism , Time Factors , beta Catenin/genetics
10.
Development ; 138(19): 4245-54, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21852398

ABSTRACT

The primary excretory organ in vertebrates is the kidney, which is responsible for blood filtration, solute homeostasis and pH balance. These functions are carried out by specialized epithelial cells organized into tubules called nephrons. Each of these cell types arise during embryonic development from a mesenchymal stem cell pool through a process of mesenchymal-to-epithelial transition (MET) that requires sequential action of specific Wnt signals. Induction by Wnt9b directs cells to exit the stem cell niche and express Wnt4, which is both necessary and sufficient for the formation of epithelia. Without either factor, MET fails, nephrons do not form and newborn mice die owing to kidney failure. Ectopic Notch activation in stem cells induces mass differentiation and exhaustion of the stem cell pool. To investigate whether this reflected an interaction between Notch and Wnt, we employed a novel gene manipulation strategy in cultured embryonic kidneys. We show that Notch activation is capable of inducing MET in the absence of both Wnt4 and Wnt9b. Following MET, the presence of Notch directs cells primarily to the proximal tubule fate. Only nephron stem cells have the ability to undergo MET in response to Wnt or Notch, as activation in the closely related stromal mesenchyme has no inductive effect. These data demonstrate that stem cells for renal epithelia are uniquely poised to undergo MET, and that Notch activation can replace key inductive Wnt signals in this process. After MET, Notch provides an instructive signal directing cells towards the proximal tubule lineage at the expense of other renal epithelial fates.


Subject(s)
Gene Expression Regulation, Developmental , Nephrons/embryology , Receptors, Notch/metabolism , Stem Cells/cytology , Wnt Proteins/metabolism , Wnt4 Protein/metabolism , Animals , Cell Dedifferentiation , Cell Lineage , Epithelial Cells/cytology , Mesoderm/metabolism , Mice , Models, Biological , Podocytes/cytology
11.
Cold Spring Harb Protoc ; 2011(1): pdb.prot5558, 2011 Jan 01.
Article in English | MEDLINE | ID: mdl-21205858

ABSTRACT

The study of organogenesis in mammals allows investigation of a wide variety of basic cell biological processes in the context of the intact organ. This has become especially important in the age of genetics, as the consequences of gene deletion or mutation in the mouse can be directly linked to human congenital abnormalities. The ability to culture some organs ex vivo during development has emerged as an important tool to understand how tissues are constructed and the signaling pathways that regulate these processes. It has been especially useful in organs that grow via branching morphogenic mechanisms, such as the lung and kidney. Here we demonstrate isolation, ex vivo growth, and fluorescent immunostaining of mouse embryonic day 12.5 (E12.5) kidneys. To demonstrate nephron formation using live imaging, we have isolated and cultured kidneys from mice carrying a green fluorescent protein (GFP) transgene driven by the Hes 1 promoter, which is expressed early in the developing nephron. We also provide a protocol for robust imaging of multiple kidney structures in the whole-mount setting. These techniques serve as a basic platform for the analysis of branching morphogenesis and nephron formation in genetic mouse models or in response to exogenous factors, such as agonists or inhibitors, which can be directly added to the culture medium.


Subject(s)
Fluorescent Antibody Technique/methods , Kidney/embryology , Kidney/growth & development , Organ Culture Techniques/methods , Staining and Labeling/methods , Animals , Fluorescent Dyes/metabolism , Kidney/metabolism , Mice , Morphogenesis , Organogenesis
12.
Proc Natl Acad Sci U S A ; 107(21): 9879-84, 2010 May 25.
Article in English | MEDLINE | ID: mdl-20457918

ABSTRACT

Mutations in leucine-rich repeat kinase 2 (LRRK2) are the most common genetic cause of Parkinson's disease. LRRK2 is a large protein containing a small GTPase domain and a kinase domain, but its physiological role is unknown. To identify the normal function of LRRK2 in vivo, we generated two independent lines of germ-line deletion mice. The dopaminergic system of LRRK2(-/-) mice appears normal, and numbers of dopaminergic neurons and levels of striatal dopamine are unchanged. However, LRRK2(-/-) kidneys, which suffer the greatest loss of LRRK compared with other organs, develop striking accumulation and aggregation of alpha-synuclein and ubiquitinated proteins at 20 months of age. The autophagy-lysosomal pathway is also impaired in the absence of LRRK2, as indicated by accumulation of lipofuscin granules as well as altered levels of LC3-II and p62. Furthermore, loss of LRRK2 dramatically increases apoptotic cell death, inflammatory responses, and oxidative damage. Collectively, our findings show that LRRK2 plays an essential and unexpected role in the regulation of protein homeostasis during aging, and suggest that LRRK2 mutations may cause Parkinson's disease and cell death via impairment of protein degradation pathways, leading to alpha-synuclein accumulation and aggregation over time.


Subject(s)
Aging , Apoptosis , Protein Serine-Threonine Kinases/metabolism , Ubiquitination , alpha-Synuclein/metabolism , Animals , Autophagy , Dopamine/metabolism , Homeostasis , Kidney Diseases/genetics , Kidney Diseases/metabolism , Kidney Diseases/pathology , Leucine-Rich Repeat Serine-Threonine Protein Kinase-2 , Lysosomes/metabolism , Mice , Mice, Knockout , Oxidation-Reduction , Protein Serine-Threonine Kinases/deficiency
13.
Dev Biol ; 337(2): 386-95, 2010 Jan 15.
Article in English | MEDLINE | ID: mdl-19914235

ABSTRACT

We previously determined that Notch2, and not Notch1, was required for forming proximal nephron segments. The dominance of Notch2 may be conserved in humans, since Notch2 mutations occur in Alagille syndrome (ALGS) 2 patients, which includes renal complications. To test whether mutations in Notch1 could increase the severity of renal complications in ALGS, we inactivated conditional Notch1 and Notch2 alleles in mice using a Six2-GFP::Cre. This BAC transgene is expressed mosaically in renal epithelial progenitors but uniformly in cells exiting the progenitor pool to undergo mesenchymal-to-epithelial transition. Although delaying Notch2 inactivation had a marginal effect on nephron numbers, it created a sensitized background in which the inactivation of Notch1 severely compromised nephron formation, function, and survival. These and additional observations indicate that Notch1 in concert with Notch2 contributes to the morphogenesis of renal vesicles into S-shaped bodies in a RBP-J-dependent manner. A significant implication is that elevating Notch1 activity could improve renal functions in ALGS2 patients. As proof of principle, we determined that conditional inactivation of Mint, an inhibitor of Notch-RBP-J interaction, resulted in a moderate rescue of Notch2 null kidneys, implying that temporal blockage of Notch signaling inhibitors downstream of receptor activation may have therapeutic benefits for ALGS patients.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Gene Dosage/genetics , Nephrons/embryology , Nephrons/metabolism , Nerve Tissue Proteins/metabolism , Receptor, Notch1/metabolism , Animals , Cell Survival , Epithelial Cells/cytology , Epithelial Cells/metabolism , Green Fluorescent Proteins/metabolism , Homeodomain Proteins/metabolism , Immunoglobulin J Recombination Signal Sequence-Binding Protein/metabolism , Integrases/metabolism , Kidney Glomerulus/cytology , Kidney Glomerulus/metabolism , Kidney Tubules, Proximal/cytology , Kidney Tubules, Proximal/metabolism , Mesoderm/cytology , Mesoderm/metabolism , Mice , Mosaicism , PAX3 Transcription Factor , Paired Box Transcription Factors/metabolism , Receptor, Notch2/metabolism , Repressor Proteins/metabolism , Stem Cells/cytology , Stem Cells/metabolism , Transcription Factors/metabolism , Transcription, Genetic , Transgenes/genetics
14.
BMC Med Inform Decis Mak ; 9: 44, 2009 Oct 08.
Article in English | MEDLINE | ID: mdl-19814826

ABSTRACT

BACKGROUND: Advances in technology and the scientific understanding of disease processes are presenting new opportunities to improve health through individualized approaches to patient management referred to as personalized medicine. Future health care strategies that deploy genomic technologies and molecular therapies will bring opportunities to prevent, predict, and pre-empt disease processes but will be dependent on knowledge management capabilities for health care providers that are not currently available. A key cornerstone to the potential application of this knowledge will be effective use of electronic health records. In particular, appropriate clinical use of genomic test results and molecularly-targeted therapies present important challenges in patient management that can be effectively addressed using electronic clinical decision support technologies. DISCUSSION: Approaches to shaping future health information needs for personalized medicine were undertaken by a work group of the American Health Information Community. A needs assessment for clinical decision support in electronic health record systems to support personalized medical practices was conducted to guide health future development activities. Further, a suggested action plan was developed for government, researchers and research institutions, developers of electronic information tools (including clinical guidelines, and quality measures), and standards development organizations to meet the needs for personalized approaches to medical practice. In this article, we focus these activities on stakeholder organizations as an operational framework to help identify and coordinate needs and opportunities for clinical decision support tools to enable personalized medicine. SUMMARY: This perspective addresses conceptual approaches that can be undertaken to develop and apply clinical decision support in electronic health record systems to achieve personalized medical care. In addition, to represent meaningful benefits to personalized decision-making, a comparison of current and future applications of clinical decision support to enable individualized medical treatment plans is presented. If clinical decision support tools are to impact outcomes in a clear and positive manner, their development and deployment must therefore consider the needs of the providers, including specific practice needs, information workflow, and practice environment.


Subject(s)
Decision Support Systems, Clinical , Information Management/trends , Medical Records Systems, Computerized/trends , Practice Management, Medical/organization & administration , Precision Medicine/trends , Humans , Practice Management, Medical/trends
15.
Dev Biol ; 333(1): 78-89, 2009 Sep 01.
Article in English | MEDLINE | ID: mdl-19576203

ABSTRACT

Identification of multipotent cardiac progenitors has provided important insights into the mechanisms of myocardial lineage specification, yet has done little to clarify the origin of the endocardium. Despite its essential role in heart development, characterization of the endocardial lineage has been limited by the lack of specific markers of this early vascular subpopulation. To distinguish endocardium from other vasculature, we generated an NFATc1-nuc-LacZ BAC transgenic mouse line capable of labeling this specific endothelial subpopulation at the earliest stages of cardiac development. To further characterize endocardiogenesis, embryonic stem cells (ESCs) derived from NFATc1-nuc-LacZ blastocysts were utilized to demonstrate that endocardial differentiation in vitro recapitulates the close temporal-spatial relationship observed between myocardium and endocardium seen in vivo. Endocardium is specified as a cardiac cell lineage, independent from other vascular populations, responding to BMP and Wnt signals that enhance cardiomyocyte differentiation. Furthermore, a population of Flk1+ cardiovascular progenitors, distinct from hemangioblast precursors, represents a mesodermal precursor of the endocardial endothelium, as well as other cardiovascular lineages. Taken together, these studies emphasize that the endocardium is a unique cardiac lineage and provides further evidence that endocardium and myocardium are derived from a common precursor.


Subject(s)
Cell Lineage/physiology , Embryonic Stem Cells/physiology , Endocardium/embryology , Endothelial Cells/physiology , Multipotent Stem Cells/physiology , Myocytes, Cardiac/physiology , Vascular Endothelial Growth Factor Receptor-2/metabolism , Animals , Antigens, Differentiation/metabolism , Cell Differentiation/physiology , Cells, Cultured , Endocardium/cytology , Endocardium/physiology , Endothelial Cells/cytology , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/physiology , Mesoderm/cytology , Mesoderm/embryology , Mice , Mice, Transgenic , Multipotent Stem Cells/cytology , Muscle, Smooth, Vascular/cytology , Muscle, Smooth, Vascular/embryology , Myocytes, Cardiac/cytology , Myocytes, Smooth Muscle/cytology , Myocytes, Smooth Muscle/physiology , NFATC Transcription Factors/genetics
16.
J Am Soc Nephrol ; 20(4): 777-86, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19297558

ABSTRACT

A number of studies have shown that placental insufficiency affects embryonic patterning of the kidney and leads to a decreased number of functioning nephrons in adulthood; however, there is circumstantial evidence that placental insufficiency may also affect renal medullary growth, which could account for cases of unexplained renal medullary dysplasia and for abnormalities in renal function among infants who had experienced intrauterine growth retardation. We observed that mice with late gestational placental insufficiency associated with genetic loss of Cited1 expression in the placenta had renal medullary dysplasia. This was not caused by lower urinary tract obstruction or by defects in branching of the ureteric bud during early nephrogenesis but was associated with decreased tissue oxygenation and increased apoptosis in the expanding renal medulla. Loss of placental Cited1 was required for Cited1 mutants to develop renal dysplasia, and this was not dependent on alterations in embryonic Cited1 expression. Taken together, these findings suggest that renal medullary dysplasia in Cited1 mutant mice is a direct consequence of decreased tissue oxygenation resulting from placental insufficiency.


Subject(s)
Kidney Diseases/genetics , Nuclear Proteins/deficiency , Nuclear Proteins/genetics , Placental Insufficiency/genetics , Trans-Activators/deficiency , Transcription Factors/deficiency , Transcription Factors/genetics , Animals , Apoptosis Regulatory Proteins , Female , Fetal Growth Retardation/genetics , Gene Expression , Humans , Infant , Kidney/embryology , Kidney Diseases/pathology , Kidney Medulla/embryology , Kidney Medulla/pathology , Mice , Mice, Inbred C57BL , Placenta/physiology , Pregnancy , Trans-Activators/genetics , Transcription, Genetic
17.
Dev Biol ; 313(1): 234-45, 2008 Jan 01.
Article in English | MEDLINE | ID: mdl-18061157

ABSTRACT

Classic tissue recombination and in vitro lineage tracing studies suggest that condensed metanephric mesenchyme (MM) gives rise to nephronic epithelium of the adult kidney. However, these studies do not distinguish between cap mesenchyme and pre-tubular aggregates comprising the condensed MM, nor do they establish whether these cells have self-renewing capacity. To address these questions, we generated Cited1-CreER(T2) BAC transgenic mice, which express tamoxifen-regulated Cre recombinase exclusively in the cap mesenchyme. Fate mapping was performed by crossing these mice with the Rosa26R(LacZ) reporter line and evaluating the location and cellular characteristics of LacZ positive cells at different time points following tamoxifen injection. These studies confirmed expected results from previous in vitro analysis of MM cell fate, and provide in vivo evidence that the cap mesenchyme does not contribute to collecting duct epithelium in the adult. Furthermore, by exploiting the temporally regulated Cre recombinase, these studies show that nephronic epithelium arising at different stages of nephrogenesis has distinct spatial distribution in the adult kidney, and demonstrate for the first time that the cap mesenchyme includes a population of self-renewing epithelial progenitor cells.


Subject(s)
Cell Lineage , Epithelial Cells/cytology , Kidney/cytology , Kidney/embryology , Mesoderm/cytology , Stem Cells/cytology , Animals , Apoptosis Regulatory Proteins , Humans , Mice , Mice, Transgenic , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Trans-Activators/genetics , Trans-Activators/metabolism
18.
Drug Discov Today ; 12(17-18): 717-24, 2007 Sep.
Article in English | MEDLINE | ID: mdl-17826684

ABSTRACT

Aberrant protein kinase signaling is a hallmark of many human diseases including cancer, diabetes, and neurological disorders. Kinase inhibitors have shown to be successful at treating some of these diseases, implying that understanding kinase signaling pathways may lead to additional, non-kinase drug targets. However, identifying substrates of protein kinases is difficult due to the universality of the chemical mechanism kinases utilize and the ability of multiple kinases to phosphorylate the same protein substrates. In this review, we explore the advantages and disadvantages of several techniques for identifying kinase substrates. Once putative substrates are identified, their validation as physiological substrates remains a major challenge. We propose three criteria for confirming the physiological relevance of a putative substrate's interaction with a kinase.


Subject(s)
Antineoplastic Agents/pharmacology , Neoplasms/enzymology , Protein Kinase Inhibitors/pharmacology , Protein Kinases/physiology , Animals , Antibodies, Monoclonal/pharmacology , Antibodies, Monoclonal/therapeutic use , Antibodies, Monoclonal, Humanized , Antineoplastic Agents/therapeutic use , Benzamides , Drug Delivery Systems , Humans , Imatinib Mesylate , Lapatinib , Neoplasms/drug therapy , Phosphoproteins/metabolism , Phosphorylation , Piperazines/pharmacology , Piperazines/therapeutic use , Protein Kinase Inhibitors/therapeutic use , Proteomics , Pyrimidines/pharmacology , Pyrimidines/therapeutic use , Quinazolines/pharmacology , Quinazolines/therapeutic use , Signal Transduction , Trastuzumab
19.
Biochemistry ; 46(41): 11614-20, 2007 Oct 16.
Article in English | MEDLINE | ID: mdl-17892306

ABSTRACT

Abl family kinases have been implicated in the regulation of cell morphogenesis and migration, but the molecular mechanisms through which they operate are not fully elucidated. We applied the bump-hole technique, pioneered by Shokat and colleagues, to identify direct substrates of Abl and the Abl-related gene (Arg) kinases. This technique required the engineering of Abl/Arg to utilize an unnatural ATP analogue as a phospho-donor. Mutation of T334A and T361A in Abl and Arg, respectively, altered their nucleotide specificity and allowed them to utilize N6-benzyl-ATP as a phospho-donor. These mutations did not affect the catalytic activity or protein substrate specificity of Abl and Arg. An unexpected high level of background labeling necessitated further optimization of this approach. Dialysis, pretreatment with a broad-spectrum Ser/Thr kinase inhibitor, K-252a, and purification of phosphotyrosine-containing proteins allowed for definitive identification of putative substrates. Using mass spectrometry, we identified eight putative substrates. One of these putative substrates, myosin IIB, can be phosphorylated in vivo by Arg. Our results indicate that the bump-hole technique can be used to identify Abl family kinase substrates and suggests that myosin IIB may be regulated by tyrosine phosphorylation.


Subject(s)
Nonmuscle Myosin Type IIB/metabolism , Protein-Tyrosine Kinases/genetics , Protein-Tyrosine Kinases/metabolism , Animals , Brain/enzymology , Carbazoles/pharmacology , Enzyme Inhibitors/pharmacology , Indole Alkaloids/pharmacology , Kinetics , Mice , Mice, Knockout , Mutagenesis, Site-Directed , Polymerase Chain Reaction , Protein-Tyrosine Kinases/chemistry , Protein-Tyrosine Kinases/deficiency , Recombinant Proteins/antagonists & inhibitors , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Substrate Specificity
20.
Neoplasia ; 9(7): 589-600, 2007 Jul.
Article in English | MEDLINE | ID: mdl-17710162

ABSTRACT

Wilms' tumors, or nephroblastomas, are thought to arise from abnormal postnatal retention and dysregulated differentiation of nephrogenic progenitor cells that originate as a condensed metanephric mesenchyme within embryonic kidneys. We have previously shown that the transcriptional regulator CITED1 (CBP/p300-interacting transactivators with glutamic acid [E]/aspartic acid [D]-rich C-terminal domain) is expressed exclusively in these nephrogenic progenitor cells and is downregulated as they differentiate to form nephronic epithelia. In the current study, we show that CITED1 expression persists in blastemal cell populations of both experimental rat nephroblastomas and human Wilms' tumors, and that primary human Wilms' tumors presenting with disseminated disease show the highest level of CITED1 expression. Unlike the predominantly cytoplasmic subcellular localization of CITED1 in the normal developing kidney, CITED1 is clearly detectable in the nuclear compartment of Wilms' tumor blastema. These findings indicate that CITED1 is a marker of primitive blastema in Wilms' tumors and suggest that persistent expression and/or altered subcellular localization of CITED1 in the condensed metanephric mesenchyme could play a role in Wilms' tumor initiation and pathogenesis.


Subject(s)
Biomarkers, Tumor/metabolism , Kidney Neoplasms/metabolism , Kidney/metabolism , Nuclear Proteins/metabolism , Transcription Factors/metabolism , Wilms Tumor/metabolism , Animals , Apoptosis Regulatory Proteins , Biomarkers, Tumor/analysis , Biomarkers, Tumor/genetics , Humans , Kidney/embryology , Kidney/pathology , Kidney Neoplasms/genetics , Kidney Neoplasms/pathology , Neoplasm Staging , Nuclear Proteins/analysis , Nuclear Proteins/genetics , RNA, Messenger/analysis , RNA, Messenger/metabolism , Rats , Rats, Inbred Strains , Trans-Activators , Transcription Factors/analysis , Transcription Factors/genetics , Wilms Tumor/genetics , Wilms Tumor/pathology
SELECTION OF CITATIONS
SEARCH DETAIL
...