Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 31
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Nat Commun ; 15(1): 371, 2024 Jan 08.
Artículo en Inglés | MEDLINE | ID: mdl-38191531

RESUMEN

Aurora Kinase A (AURKA) promotes cell proliferation and is overexpressed in different types of polycystic kidney disease (PKD). To understand AURKA's role in regulating renal cyst development we conditionally deleted the gene in mouse models of Autosomal Dominant PKD (ADPKD) and Joubert Syndrome, caused by Polycystin 1 (Pkd1) and Inositol polyphosphate-5-phosphatase E (Inpp5e) mutations respectively. We show that while Aurka is dispensable for collecting duct development and homeostasis, its deletion prevents cyst formation in both disease models. Cross-comparison of transcriptional changes implicated AKT signaling in cyst prevention and we show that (i) AURKA and AKT physically interact, (ii) AURKA regulates AKT activity in a kinase-independent manner and (iii) inhibition of AKT can reduce disease severity. AKT activation also regulates Aurka expression, creating a feed-forward loop driving renal cystogenesis. We find that the AURKA kinase inhibitor Alisertib stabilises the AURKA protein, agonizing its cystogenic functions. These studies identify AURKA as a master regulator of renal cyst development in different types of PKD, functioning in-part via AKT.


Asunto(s)
Aurora Quinasa A , Quistes , Enfermedades Renales Poliquísticas , Riñón Poliquístico Autosómico Dominante , Animales , Ratones , Aurora Quinasa A/genética , Monoéster Fosfórico Hidrolasas , Enfermedades Renales Poliquísticas/genética , Enfermedades Renales Poliquísticas/prevención & control , Proteínas Proto-Oncogénicas c-akt/genética
2.
Kidney Int ; 105(4): 731-743, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38158181

RESUMEN

Autosomal Dominant Polycystic Kidney Disease (ADPKD) is a leading cause of kidney failure and is associated with substantial morbidity and mortality. Interstitial inflammation is attributed to the action of infiltrating macrophages and is a feature thought to aggravate disease progression. Here, we investigated the therapeutic potential of the anti-inflammatory IL37b cytokine as a treatment for ADPKD using genetic mouse models, demonstrating that transgenic expression of human IL37b reduced collecting duct cyst burden in both early and adult-onset ADPKD rodent models. Moreover, injection of recombinant human IL37b could also reduce cyst burden in early onset ADPKD mice, an observation not associated with increased macrophage number at early stages of cyst formation. Interestingly, transgenic IL37b expression also did not alter macrophage numbers in advanced disease. Whole kidney RNA-seq highlighted an IL37b-mediated upregulation of the interferon signaling pathway and single-cell RNA-seq established that these changes originate at least partly from kidney resident macrophages. We further found that blocking type I interferon signaling in mice expressing IL37b resulted in increased cyst number, confirming this as an important pathway by which IL37b exerts its beneficial effects. Thus, our studies show that IL37b promotes interferon signaling in kidney resident macrophages which suppresses cyst initiation, identifying this protein as a potential therapy for ADPKD.


Asunto(s)
Quistes , Riñón Poliquístico Autosómico Dominante , Ratones , Humanos , Animales , Riñón Poliquístico Autosómico Dominante/tratamiento farmacológico , Riñón Poliquístico Autosómico Dominante/genética , Inflamación/genética , Inflamación/complicaciones , Riñón/metabolismo , Quistes/complicaciones , Interleucinas , Interferones
3.
bioRxiv ; 2023 Dec 05.
Artículo en Inglés | MEDLINE | ID: mdl-38106143

RESUMEN

Low nephron number correlates with the development of hypertension and chronic kidney disease later in life. While intrauterine growth restriction caused by maternal low protein diet (LPD) is thought to be a significant cause of reduced nephron endowment in impoverished communities, its influence on the cellular and molecular processes which drive nephron formation are poorly understood. We conducted a comprehensive characterization of the impact of LPD on kidney development using tomographic and confocal imaging to quantify changes in branching morphogenesis and the cellular and morphological features of nephrogenic niches across development. These analyses were paired with single-cell RNA sequencing to dissect the transcriptional changes that LPD imposes during renal development. Differences in the expression of genes involved in metabolism were identified in most cell types we analyzed, yielding imbalances and shifts in cellular energy production. We further demonstrate that LPD impedes branching morphogenesis and significantly reduces the number of pretubular aggregates - the initial precursors to nephron formation. The most striking observation was that LPD changes the developmental trajectory of nephron progenitor cells, driving the formation of a partially committed cell population which likely reflects a failure of cells to commit to nephron formation and which ultimately reduces endowment. This unique profile of a fetal programming defect demonstrates that low nephron endowment arises from the pleiotropic impact of changes in branching morphogenesis and nephron progenitor cell commitment, the latter of which highlights a critical role for nutrition in regulating the cell fate decisions underpinning nephron endowment. Significance Statement: While a mother's diet and behavior can negatively impact the number of nephrons in the kidneys of her offspring, the root cellular and molecular drivers of these deficits have not been rigorously explored. In this study we use advanced imaging and gene expression analysis in mouse models to define how a maternal low protein diet, analogous to that of impoverished communities, results in reduced nephron endowment. We find that low protein diet has pleiotropic effects on metabolism and the normal programs of gene expression. These profoundly impact the process of branching morphogenesis necessary to establish niches for nephron generation and change cell behaviors which regulate how and when nephron progenitor cells commit to differentiation.

4.
Anat Rec (Hoboken) ; 303(10): 2578-2587, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-32790143

RESUMEN

Branching morphogenesis is an integral developmental mechanism central to the formation of a range of organs including the kidney, lung, pancreas and mammary gland. The ramified networks of epithelial tubules it establishes are critical for the processes of secretion, excretion and exchange mediated by these tissues. In the kidney, branching serves to establish the collecting duct system that transports urine from the nephrons into the renal pelvis, ureter and finally the bladder. Generally speaking, the formation of these networks in different organs begins with the specification and differentiation of simple bud-like organ anlage, which then undergo a process of elaboration, typically by bifurcation. This process is often governed by the interaction of progenitor cells at the tips of the epithelia with neighboring mesenchymal cell populations which direct the branching process and which often themselves differentiate to form part of the adult organ. In the kidney, the tips of ureteric bud elaborate through a dynamic cell signaling relationship with overlying nephron progenitor cell populations. These cells sequentially commit to differentiation and the resulting nephrons reintegrate with the ureteric epithelium as development progresses. This review will describe recent advances in understanding the how the elaboration of the ureteric bud is patterned and consider the extent to which this process is shared with other organs.


Asunto(s)
Diferenciación Celular/fisiología , Riñón/embriología , Organogénesis/fisiología , Humanos , Células Madre/fisiología
5.
Development ; 147(21)2020 06 22.
Artículo en Inglés | MEDLINE | ID: mdl-32439764

RESUMEN

Laminin alpha 5 (LAMA5) is a member of a large family of proteins that trimerise and then polymerise to form a central component of all basement membranes. Consequently, the protein plays an instrumental role in shaping the normal development of the kidney, skin, neural tube, lung and limb, and many other organs and tissues. Pathogenic mutations in some laminins have been shown to cause a range of largely syndromic conditions affecting the competency of the basement membranes to which they contribute. We report the identification of a mutation in the polymerisation domain of LAMA5 in a patient with a complex syndromic disease characterised by defects in kidney, craniofacial and limb development, and by a range of other congenital defects. Using CRISPR-generated mouse models and biochemical assays, we demonstrate the pathogenicity of this variant, showing that the change results in a failure of the polymerisation of α/ß/γ laminin trimers. Comparing these in vivo phenotypes with those apparent upon gene deletion in mice provides insights into the specific functional importance of laminin polymerisation during development and tissue homeostasis.


Asunto(s)
Discapacidades del Desarrollo/genética , Desarrollo Fetal , Laminina/genética , Mutación/genética , Polimerizacion , Secuencia de Aminoácidos , Animales , Animales Recién Nacidos , Preescolar , Discapacidades del Desarrollo/patología , Feto/embriología , Humanos , Hidronefrosis/patología , Recién Nacido , Riñón/anomalías , Riñón/embriología , Riñón/patología , Laminina/química , Pulmón/anomalías , Pulmón/embriología , Pulmón/patología , Masculino , Ratones , Dominios Proteicos , Síndrome
6.
Dev Biol ; 454(2): 156-169, 2019 10 15.
Artículo en Inglés | MEDLINE | ID: mdl-31242448

RESUMEN

Adamts18 encodes a secreted metalloprotease restricted to branch-tip progenitor pools directing the morphogenesis of multiple mammalian organs. Adamts18 was targeted to explore a potential role in branching morphogenesis. In the kidney, an arborized collecting system develops through extensive branching morphogenesis of an initial epithelial outgrowth of the mesonephric duct, the ureteric bud. Adamts18 mutants displayed a weakly penetrant phenotype: duplicated ureteric outgrowths forming enlarged, bi-lobed kidneys with an increased nephron endowment. In contrast, Adamts18 mutants showed a fully penetrant lung phenotype: epithelial growth was markedly reduced and early secondary branching scaled to the reduced length of the primary airways. Furthermore, there was a pronounced delay in the appearance of differentiated cell types in both proximal and distally positions of the developing airways. Adamts18 is closely related to Adamts16. In the kidney but not the lung, broad epithelial Adamts16 expression overlaps Adamts18 in branch tips. However, compound Adamts16/18 mutants displayed a comparable low penetrance duplicated ureteric phenotype, ruling out a possible role for Adamts16 as a functional modifier of the Adamts18 kidney phenotype. Given the predicted action of secreted Adamts18 metalloprotease, and broad expression of Adamts18 in branching organ systems, these findings suggest distinct requirements for matrix modelling in the morphogenesis of epithelial networks.


Asunto(s)
Proteínas ADAMTS/metabolismo , Organogénesis/fisiología , Proteínas ADAMTS/fisiología , Animales , Femenino , Regulación del Desarrollo de la Expresión Génica/genética , Riñón/citología , Riñón/embriología , Riñón/metabolismo , Pulmón/embriología , Pulmón/metabolismo , Masculino , Metaloproteasas/genética , Metaloproteasas/metabolismo , Ratones , Ratones Noqueados , Morfogénesis , Nefronas/metabolismo , Técnicas de Cultivo de Órganos/métodos , Uréter/metabolismo
7.
Elife ; 72018 12 05.
Artículo en Inglés | MEDLINE | ID: mdl-30516471

RESUMEN

A normal endowment of nephrons in the mammalian kidney requires a balance of nephron progenitor self-renewal and differentiation throughout development. Here, we provide evidence for a novel action of ureteric branch tip-derived Wnt11 in progenitor cell organization and interactions within the nephrogenic niche, ultimately determining nephron endowment. In Wnt11 mutants, nephron progenitors dispersed from their restricted niche, intermixing with interstitial progenitors. Nephron progenitor differentiation was accelerated, kidneys were significantly smaller, and the nephron progenitor pool was prematurely exhausted, halving the final nephron count. Interestingly, RNA-seq revealed no significant differences in gene expression. Live imaging of nephron progenitors showed that in the absence of Wnt11 they lose stable attachments to the ureteric branch tips, continuously detaching and reattaching. Further, the polarized distribution of several markers within nephron progenitors is disrupted. Together these data highlight the importance of Wnt11 signaling in directing nephron progenitor behavior which determines a normal nephrogenic program.


Asunto(s)
Polaridad Celular/genética , Regulación del Desarrollo de la Expresión Génica , Nefronas/metabolismo , Organogénesis/genética , Células Madre/metabolismo , Proteínas Wnt/genética , Animales , Diferenciación Celular , Movimiento Celular , Embrión de Mamíferos , Femenino , Genes Reporteros , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Queratina-8/genética , Queratina-8/metabolismo , Masculino , Ratones , Ratones Transgénicos , Nefronas/citología , Nefronas/crecimiento & desarrollo , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Transducción de Señal , Células Madre/citología , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Proteínas Wnt/metabolismo
8.
Elife ; 72018 07 31.
Artículo en Inglés | MEDLINE | ID: mdl-30063208

RESUMEN

Branching morphogenesis of the ureteric bud is integral to kidney development; establishing the collecting ducts of the adult organ and driving organ expansion via peripheral interactions with nephron progenitor cells. A recent study suggested that termination of tip branching within the developing kidney involved stochastic exhaustion in response to nephron formation, with such a termination event representing a unifying developmental process evident in many organs. To examine this possibility, we have profiled the impact of nephron formation and maturation on elaboration of the ureteric bud during mouse kidney development. We find a distinct absence of random branch termination events within the kidney or evidence that nephrogenesis impacts the branching program or cell proliferation in either tip or progenitor cell niches. Instead, organogenesis proceeds in a manner indifferent to the development of these structures. Hence, stochastic cessation of branching is not a unifying developmental feature in all branching organs.


Asunto(s)
Nefronas/embriología , Organogénesis , Animales , Proliferación Celular , Embrión de Mamíferos/metabolismo , Proteínas Fluorescentes Verdes/metabolismo , Ratones , Nefronas/citología , Uréter/embriología
9.
Development ; 144(23): 4377-4385, 2017 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-29038307

RESUMEN

Metanephric kidney development is orchestrated by the iterative branching morphogenesis of the ureteric bud. We describe an underlying patterning associated with the ramification of this structure and show that this pattern is conserved between developing kidneys, in different parts of the organ and across developmental time. This regularity is associated with a highly reproducible branching asymmetry that is consistent with locally operative growth mechanisms. We then develop a class of tip state models to represent elaboration of the ureteric tree and describe rules for 'half-delay' branching morphogenesis that describe almost perfectly the patterning of this structure. Spatial analysis suggests that the observed asymmetry may arise from mutual suppression of bifurcation, but not extension, between the growing ureteric tips, and demonstrates that disruption of patterning occurs in mouse mutants in which the distribution of tips on the surface of the kidney is altered. These findings demonstrate that kidney development occurs by way of a highly conserved reiterative pattern of asymmetric bifurcation that is governed by intrinsic and locally operative mechanisms.


Asunto(s)
Riñón/embriología , Morfogénesis/fisiología , Uréter/embriología , Proteínas Adaptadoras Transductoras de Señales/deficiencia , Proteínas Adaptadoras Transductoras de Señales/genética , Proteínas Adaptadoras Transductoras de Señales/fisiología , Animales , Tipificación del Cuerpo/genética , Tipificación del Cuerpo/fisiología , Proteína Morfogenética Ósea 7/deficiencia , Proteína Morfogenética Ósea 7/genética , Proteína Morfogenética Ósea 7/fisiología , Imagenología Tridimensional , Conceptos Matemáticos , Proteínas de la Membrana/deficiencia , Proteínas de la Membrana/genética , Proteínas de la Membrana/fisiología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Mutantes , Modelos Biológicos , Morfogénesis/genética , Mutación , Fenotipo , Fosfoproteínas/deficiencia , Fosfoproteínas/genética , Fosfoproteínas/fisiología , Factor de Crecimiento Transformador beta2/deficiencia , Factor de Crecimiento Transformador beta2/genética , Factor de Crecimiento Transformador beta2/fisiología
10.
Results Probl Cell Differ ; 60: 233-256, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28409348

RESUMEN

The kidney develops as an outgrowth of the epithelial nephric duct known as the ureteric bud, in a position specified by a range of rostral and caudal factors which serve to ensure two kidneys form in the appropriate positions in the body. At its simplest level, kidney development can be viewed as the process by which this single bud then undergoes a process of arborisation to form a complex connected network of ducts which will serve to drain urine from the nephrons in the adult organ. The process of bud elaboration is dictated by factors expressed by both the bud itself and by surrounding cells of the metanephric mesenchyme which control cell division and bifurcation. These cells play two critical roles. Firstly, they potentiate the ongoing elaboration of the ureteric tree: remove them and branching ceases. Secondly, they harbour progenitor cells which are fated to undergo their own process of tubulogenesis to form the nephrons of the adult organ. In this chapter, we will discuss how the ureteric bud arises in the developing embryo, how it undergoes branching, how we can measure and study this process and finally the likely relevance that this process has for our understanding of congenital and acquired kidney disease.


Asunto(s)
Riñón/embriología , Morfogénesis/fisiología , Animales , Humanos , Procesamiento de Imagen Asistido por Computador
11.
Nat Rev Nephrol ; 12(12): 754-767, 2016 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-27818506

RESUMEN

The mammalian kidney develops from a simple epithelial bud to an arborized network of tubules, which are fated to form the ureter, renal pelvis and collecting ducts. This process of ductal elaboration is achieved through an ancient developmental mechanism known as branching morphogenesis that is widely employed in glandular organs, the vasculature and lungs. It breaks up large solid tissues facilitating secretion, excretion and gas exchange, depending on the tissue. In the kidney, growth of the ureteric bud is driven by interactions between progenitor cells in the tips of the epithelial tree and their mesenchymal 'caps'. The cells of the cap mesenchyme give rise to nephrons; therefore, the interaction between these two cell populations is likely to be a critical driver of nephron number, which is determined during gestation. These cellular interactions are potentially affected by genetic mutations (congenital kidney diseases) and by changes in the fetal environment. Understanding the aetiology of congenital and acquired kidney diseases therefore requires a full appreciation of the processes involved in establishing the cellular architecture of the kidney and of the factors that affect the commitment of progenitor cells to form nephrons.


Asunto(s)
Enfermedades Renales/embriología , Riñón/embriología , Morfogénesis , Organogénesis , Animales , Humanos , Ratones , Morfogénesis/genética , Morfogénesis/fisiología , Nefronas/embriología , Organogénesis/genética , Organogénesis/fisiología , Células Madre , Uréter/embriología
12.
J Am Soc Nephrol ; 27(10): 3093-3104, 2016 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26975438

RESUMEN

Podocyte depletion is sufficient for the development of numerous glomerular diseases and can be absolute (loss of podocytes) or relative (reduced number of podocytes per volume of glomerulus). Commonly used methods to quantify podocyte depletion introduce bias, whereas gold standard stereologic methodologies are time consuming and impractical. We developed a novel approach for assessing podocyte depletion in whole glomeruli that combines immunofluorescence, optical clearing, confocal microscopy, and three-dimensional analysis. We validated this method in a transgenic mouse model of selective podocyte depletion, in which we determined dose-dependent alterations in several quantitative indices of podocyte depletion. This new approach provides a quantitative tool for the comprehensive and time-efficient analysis of podocyte depletion in whole glomeruli.


Asunto(s)
Recuento de Células/métodos , Tamaño de la Célula , Glomérulos Renales/citología , Podocitos/citología , Animales , Imagenología Tridimensional , Ratones
13.
Sci Rep ; 5: 15209, 2015 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-26469293

RESUMEN

Sexual dimorphism is a prominent feature of renal physiology and as a consequence, it differentially affects predisposition to many adult kidney diseases. Furthermore the left and right kidneys differ in terms of their position, size and involvement in congenital malformations of the urogenital tract. We set out to determine whether differences in the program of branching morphogenesis that establishes the basic architecture of the kidney were apparent with respect to either sex or laterality in mouse embryonic kidneys. This was achieved using a combination of optical projection tomography imaging and computational analysis of many spatial metrics describing the branched ureteric tree. We undertook a comprehensive assessment of twelve aspects of ureteric morphology across developmental time and we found no consistent differences between kidneys of different sexes or laterality. These results suggest that dimorphism is established after birth or at a physiological or cellular level that is not reflected in the morphology of the ureteric tree.


Asunto(s)
Riñón/embriología , Morfogénesis/fisiología , Animales , Desarrollo Embrionario , Femenino , Riñón/anatomía & histología , Masculino , Ratones , Ratones Endogámicos C57BL , Caracteres Sexuales , Tomografía Óptica , Uréter/anatomía & histología , Uréter/crecimiento & desarrollo
14.
Development ; 142(8): 1458-69, 2015 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-25790853

RESUMEN

Epigenetic mechanisms involved in the establishment of lung epithelial cell lineage identities during development are largely unknown. Here, we explored the role of the histone methyltransferase Ezh2 during lung lineage determination. Loss of Ezh2 in the lung epithelium leads to defective lung formation and perinatal mortality. We show that Ezh2 is crucial for airway lineage specification and alveolarization. Using optical projection tomography imaging, we found that branching morphogenesis is affected in Ezh2 conditional knockout mice and the remaining bronchioles are abnormal, lacking terminally differentiated secretory club cells. Remarkably, RNA-seq analysis revealed the upregulation of basal genes in Ezh2-deficient epithelium. Three-dimensional imaging for keratin 5 further showed the unexpected presence of a layer of basal cells from the proximal airways to the distal bronchioles in E16.5 embryos. ChIP-seq analysis indicated the presence of Ezh2-mediated repressive marks on the genomic loci of some but not all basal genes, suggesting an indirect mechanism of action of Ezh2. We found that loss of Ezh2 de-represses insulin-like growth factor 1 (Igf1) expression and that modulation of IGF1 signaling ex vivo in wild-type lungs could induce basal cell differentiation. Altogether, our work reveals an unexpected role for Ezh2 in controlling basal cell fate determination in the embryonic lung endoderm, mediated in part by repression of Igf1 expression.


Asunto(s)
Diferenciación Celular/fisiología , Factor I del Crecimiento Similar a la Insulina/metabolismo , Pulmón/citología , Pulmón/metabolismo , Complejo Represivo Polycomb 2/metabolismo , Animales , Diferenciación Celular/genética , Inmunoprecipitación de Cromatina , Proteína Potenciadora del Homólogo Zeste 2 , Citometría de Flujo , Factor I del Crecimiento Similar a la Insulina/genética , Queratina-5/genética , Queratina-5/metabolismo , Pulmón/embriología , Ratones , Complejo Represivo Polycomb 2/genética , Reacción en Cadena de la Polimerasa
15.
Mamm Genome ; 26(1-2): 57-79, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25552398

RESUMEN

Genetic background plays a dominant role in mammary gland development and breast cancer (BrCa). Despite this, the role of genetics is only partially understood. This study used strain-dependent variation in an inbred mouse mapping panel, to identify quantitative trait loci (QTL) underlying structural variation in mammary ductal development, and determined if these QTL correlated with genomic intervals conferring BrCa susceptibility in humans. For about half of the traits, developmental variation among the complete set of strains in this study was greater (P < 0.05) than that of previously studied strains, or strains in current common use for mammary gland biology. Correlations were also detected with previously reported variation in mammary tumor latency and metastasis. In-silico genome-wide association identified 20 mammary development QTL (Mdq). Of these, five were syntenic with previously reported human BrCa loci. The most significant (P = 1 × 10(-11)) association of the study was on MMU6 and contained the genes Plxna4, Plxna4os1, and Chchd3. On MMU5, a QTL was detected (P = 8 × 10(-7)) that was syntenic to a human BrCa locus on h12q24.5 containing the genes Tbx3 and Tbx5. Intersection of linked SNP (r(2) > 0.8) with genomic and epigenomic features, and intersection of candidate genes with gene expression and survival data from human BrCa highlighted several for further study. These results support the conclusion that mammary tumorigenesis and normal ductal development are influenced by common genetic factors and that further studies of genetically diverse mice can improve our understanding of BrCa in humans.


Asunto(s)
Neoplasias de la Mama/genética , Glándulas Mamarias Animales/crecimiento & desarrollo , Ratones Endogámicos/genética , Sitios de Carácter Cuantitativo/genética , Animales , Neoplasias de la Mama/fisiopatología , Mapeo Cromosómico , Simulación por Computador , Femenino , Estudio de Asociación del Genoma Completo , Técnicas Histológicas , Humanos , Ratones , Polimorfismo de Nucleótido Simple/genética , Especificidad de la Especie , Sintenía/genética , Tomografía Óptica
16.
J Theor Biol ; 365: 226-37, 2015 Jan 21.
Artículo en Inglés | MEDLINE | ID: mdl-25308508

RESUMEN

Bifurcating developmental branching morphogenesis gives rise to complex organs such as the lung and the ureteric tree of the kidney. However, a few quantitative methods or tools exist to compare and distinguish, at a structural level, the critical features of these important biological systems. Here we develop novel graph alignment techniques to quantify the structural differences of rooted bifurcating trees and demonstrate their application in the analysis of developing kidneys from in normal and mutant mice. We have developed two graph based metrics: graph discordance, which measures how well the graphs representing the branching structures of distinct trees graphs can be aligned or overlayed; and graph inclusion, which measures the degree of containment of a tree graph within another. To demonstrate the application of these approaches we first benchmark the discordance metric on a data set of 32 normal and 28Tgfß(+/-) mutant mouse ureteric trees. We find that the discordance metric better distinguishes control and mutant mouse kidneys than alternative metrics based on graph size and fingerprints - the distribution of tip depths. Using this metric we then show that the structure of the mutant trees follows the same pattern as the normal kidneys, but undergo a major delay in elaboration at later stages. Analysis of both controls and mutants using the inclusion metric gives strong support to the hypothesis that ureteric tree growth is stereotypic. Additionally, we present a new generalised multi-tree alignment algorithm that minimises the sum of pairwise graph discordance and which can be used to generate maximum consensus trees that represent the archetype for fixed developmental stages. These tools represent an advance in the analysis and quantification of branching patterns and will be invaluable in gaining a deeper understanding of the mechanisms that drive development. All code is being made available with documentation and example data with this publication.


Asunto(s)
Morfogénesis , Uréter/crecimiento & desarrollo , Animales , Riñón/crecimiento & desarrollo , Riñón/metabolismo , Ratones , Mutación/genética , Factor de Crecimiento Transformador beta2/metabolismo , Uréter/metabolismo
17.
Nat Protoc ; 9(12): 2859-79, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25411953

RESUMEN

Developmental branching morphogenesis establishes organ architecture, and it is driven by iterative interactions between epithelial and mesenchymal progenitor cell populations. We describe an approach for analyzing this interaction and how it contributes to organ development. After initial in vivo cell labeling with the nucleoside analog 5-ethynyl-2'-deoxyuridine (EdU) and tissue-specific antibodies, optical projection tomography (OPT) and confocal microscopy are used to image the developing organ. These imaging data then inform a second analysis phase that quantifies (using Imaris and Tree Surveyor software), models and integrates these events at a cell and tissue level in 3D space and across developmental time. The protocol establishes a benchmark for assessing the impact of genetic change or fetal environment on organogenesis that does not rely on ex vivo organ culture or section-based reconstruction. By using this approach, examination of two developmental stages for an organ such as the kidney can be undertaken by a postdoctoral-level researcher in 6 weeks, with a full developmental analysis in mouse achievable in 5 months.


Asunto(s)
Imagenología Tridimensional/métodos , Riñón/embriología , Riñón/crecimiento & desarrollo , Técnicas de Cultivo de Órganos/métodos , Animales , Proliferación Celular , Desoxiuridina/análogos & derivados , Riñón/citología , Ratones Endogámicos C57BL , Microscopía Confocal/métodos , Morfogénesis , Organogénesis , Programas Informáticos , Células Madre/citología , Tomografía Óptica/métodos
18.
Dev Cell ; 29(2): 188-202, 2014 Apr 28.
Artículo en Inglés | MEDLINE | ID: mdl-24780737

RESUMEN

Although kidneys of equal size can vary 10-fold in nephron number at birth, discovering what regulates such variation has been hampered by a lack of quantitative parameters defining kidney development. Here we report a comprehensive, quantitative, multiscale analysis of mammalian kidney development in which we measure changes in cell number, compartment volumes, and cellular dynamics across the entirety of organogenesis, focusing on two key nephrogenic progenitor populations: the ureteric epithelium and the cap mesenchyme. In doing so, we describe a discontinuous developmental program governed by dynamic changes in interactions between these key cellular populations occurring within a previously unappreciated structurally stereotypic organ architecture. We also illustrate the application of this approach to the detection of a subtle mutant phenotype. This baseline program of kidney morphogenesis provides a framework for assessing genetic and environmental developmental perturbation and will serve as a gold standard for the analysis of other organs.


Asunto(s)
Riñón/embriología , Nefronas/embriología , Uréter/embriología , Urotelio/embriología , Animales , Recuento de Células , Células Madre Embrionarias/fisiología , Femenino , Regulación del Desarrollo de la Expresión Génica , Riñón/citología , Riñón/fisiología , Masculino , Ratones , Ratones Endogámicos C57BL , Mutación , Nefronas/citología , Nefronas/fisiología , Fenotipo , Embarazo , Uréter/citología , Uréter/fisiología , Urotelio/citología , Urotelio/fisiología
19.
Exp Neurol ; 249: 8-19, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-23939410

RESUMEN

Purkinje neurons are a sensitive and specialised cell type important for fine motor movement and coordination. Purkinje cell damage manifests as motor incoordination and ataxia - a prominent feature of many human disorders including spinocerebellar ataxia and Huntington's disease. A correlation between Purkinje degeneration and excess cerebellar levels of tissue-type plasminogen activator (tPA) has been observed in multiple genetically-distinct models of ataxia. Here we show that Purkinje loss in a mouse model of Huntington's disease also correlates with a 200% increase in cerebellar tPA activity. That elevated tPA levels arise in a variety of ataxia models suggests that tPA is a common mediator of Purkinje damage. To address the specific contribution of tPA to cerebellar dysfunction we studied the T4 mice line that overexpresses murine tPA in postnatal neurons through the Thy1.2 gene promoter, which directs preferential expression to Purkinje cells within the cerebellum. Here we show that T4 mice develop signs of cerebellar damage within 10 weeks of birth including atrophy of Purkinje cell soma and dendrites, astrogliosis, reduced molecular layer volume and altered gait. In contrast, T4 mice displayed no evidence of microgliosis, nor any changes in interneuron density, nor alteration in the cerebellar granular neuron layer. Thus, excess tPA levels may be sufficient to cause targeted Purkinje cell degeneration and ataxia. We propose that elevated cerebellar tPA levels exert a common pathway of Purkinje cell damage. Therapeutically lowering cerebellar tPA levels may represent a novel means of preserving Purkinje cell integrity and motor coordination across a wide range of neurodegenerative diseases.


Asunto(s)
Ataxia/metabolismo , Ataxia/fisiopatología , Líquido Extracelular/metabolismo , Marcha/fisiología , Células de Purkinje/metabolismo , Activador de Tejido Plasminógeno/fisiología , Animales , Ataxia/enzimología , Líquido Extracelular/enzimología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Células de Purkinje/enzimología , Células de Purkinje/patología
20.
PLoS One ; 8(3): e58243, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23516451

RESUMEN

There is strong evidence from human and animal models that exposure to maternal hyperglycemia during in utero development can detrimentally affect fetal kidney development. Notwithstanding this knowledge, the precise effects of diabetic pregnancy on the key processes of kidney development are unclear due to a paucity of studies and limitations in previously used methodologies. The purpose of the present study was to elucidate the effects of hyperglycemia on ureteric branching morphogenesis and nephrogenesis using unbiased techniques. Diabetes was induced in pregnant C57Bl/6J mice using multiple doses of streptozotocin (STZ) on embryonic days (E) 6.5-8.5. Branching morphogenesis was quantified ex vivo using Optical Projection Tomography, and nephrons were counted using unbiased stereology. Maternal hyperglycemia was recognised from E12.5. At E14.5, offspring of diabetic mice demonstrated fetal growth restriction and a marked deficit in ureteric tip number (control 283.7 ± 23.3 vs. STZ 153.2 ± 24.6, mean ± SEM, p<0.01) and ureteric tree length (control 33.1 ± 2.6 mm vs. STZ 17.6 ± 2.7 mm, p = 0.001) vs. controls. At E18.5, fetal growth restriction was still present in offspring of STZ dams and a deficit in nephron endowment was observed (control 1246.2 ± 64.9 vs. STZ 822.4 ± 74.0, p<0.001). Kidney malformations in the form of duplex ureter and hydroureter were a common observation (26%) in embryos of diabetic pregnancy compared with controls (0%). Maternal insulin treatment from E13.5 normalised maternal glycaemia but did not normalise fetal weight nor prevent the nephron deficit. The detrimental effect of hyperglycemia on ureteric branching morphogenesis and, in turn, nephron endowment in the growth-restricted fetus highlights the importance of glycemic control in early gestation and during the initial stages of renal development.


Asunto(s)
Hiperglucemia/metabolismo , Morfogénesis , Nefronas/embriología , Embarazo en Diabéticas/metabolismo , Uréter/embriología , Animales , Glucemia/efectos de los fármacos , Diabetes Mellitus Experimental , Femenino , Desarrollo Fetal/efectos de los fármacos , Humanos , Hiperglucemia/inducido químicamente , Insulina/metabolismo , Insulina/farmacología , Ratones , Morfogénesis/efectos de los fármacos , Nefronas/efectos de los fármacos , Embarazo , Embarazo en Diabéticas/inducido químicamente , Efectos Tardíos de la Exposición Prenatal , Estreptozocina/efectos adversos , Uréter/efectos de los fármacos , Sistema Urinario/anomalías , Sistema Urinario/embriología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...