• Refine Query
  • Source
  • Publication year
  • to
  • Language
  • 10
  • 2
  • 2
  • 2
  • Tagged with
  • 24
  • 12
  • 10
  • 8
  • 6
  • 6
  • 6
  • 5
  • 5
  • 5
  • 5
  • 4
  • 4
  • 4
  • 4
  • About
  • The Global ETD Search service is a free service for researchers to find electronic theses and dissertations. This service is provided by the Networked Digital Library of Theses and Dissertations.
    Our metadata is collected from universities around the world. If you manage a university/consortium/country archive and want to be added, details can be found on the NDLTD website.
1

Inflammatory mediators in normal and abnormal renal development

Cale, Catherine Mary January 1999 (has links)
No description available.
2

The crucial roles played by HNF1β during kidney development / Le rôle décisif du HNF1ß durant le développement rénal

Massa, Filippo Maria 14 November 2012 (has links)
HNF1beta est un facteur de transcription exprimé pendant le développement rénal dans le bourgeon urétéral et les dérivés épithéliaux du blastème métanéphrique. Les mutations de HNF1B/MODY5 représentent l’une des anomalies génétiques les plus fréquentes à l’origine des CAKUT (Congenital Abnormalities of Kidney and Urinary Tract). Afin d’étudier le rôle de HNF1beta au cours du développement rénal, j’ai utilisé un système CRE-LoxP pour inactiver Hnf1b dans différents compartiments et à différents temps au cours de la morphogenèse rénale chez la souris. Mes résultats ont montré que Hnf1b est nécessaire pour l’expression de gènes clés exprimés dans le bourgeon urétéral, incluant Wnt9b, Emx2, Pax2 et Lhx1.Leur absence est à l’origine de la formation de reins rudimentaires, caractérisés par un branchement anormal du bourgeon urétéral et une absence de transition mésenchyme-épithélium. D’autre part, l’inactivation spécifique de Hnf1b dans le blastème métanéphrique n’affecte pas les premiers dérivés épithéliaux mais conduit à la formation de précurseurs néphroniques anormaux. En l’absence de HNF1beta, les corps en S ne développent pas une protubérance de cellules épithéliales, à l’origine des tubules proximaux et des anses de Henlé. Au niveau moléculaire, ces anomalies sont associées à une diminution de l’expression de Dll1, un ligand de Notch, qui est directement contrôlé par HNF1beta. Cette diminution conduit à une activation défectueuse de la voie Notch dans le futur compartiment tubulaire du corps en virgule et du corps en S. Des résultats préliminaires concernant une inactivation très chimérique de Hnf1b dans le mésenchyme ont montré que dans ce modèle, les néphrons se forment normalement mais que les tubules sont plus courts et partiellement kystiques. Mes résultats ont montré que les modèles murins d’inactivation conditionnelle de Hnf1b récapitulent la plupart des anomalies observées chez les patients MODY5. L’ensemble de mes résultats améliore notre compréhension du programme génétique contrôlé par Hnf1b au cours du développement dont les anomalies peuvent conduire aux CAKUT chez les patients / HNF1β is a transcription factor expressed during nephrogenesis in the ureteric bud and in the tubular epithelia derived from the metanephric mesenchyme. Mutations in HNF1B/MODY5 represent one of the most prevalent human genetic defects responsible for Congenital Abnormalities of Kidney and Urinary Tract (CAKUT). To investigate the role played by HNF1beta during kidney development, I made use of a CRE-LoxP strategy to inactivate Hnf1b in different compartments and at different time points during mouse kidney morphogenesis. My results showed that Hnf1b is required for the expression of several key genes including Wnt9b, Emx2, Pax2 and Lhx. These genes are normally expressed in the ureteric bud and their absence leads to the formation of rudimentary kidneys, characterized by abnormal ureteric bud branching and lack of mesenchymal to epithelial transition. On the other hand, the specific inactivation of Hnf1b in the metanephric mesenchyme does not affect the early steps of epithelisation but leads to severe malformation of nephron precursors. In the absence of HNF1β, S-Shaped bodies lack the formation of a bulge of epithelial cells that normally give rise to Henle’s loops and proximal tubules. At the molecular level, these defects are associated with a decreased expression of Dll1, a Notch ligand that is directly controlled by HNF1β. This downregulation leads to a defective activation of the Notch pathway in the prospective tubular compartment of comma and S-shaped bodies. Preliminary results using a highly chimeric inactivation of Hnf1b in the mesenchyme showed that the spotted absence of Hnf1b does not prevent nephrogenesis. However, tubules are shorter and partially cystic. My results showed that murine models for Hnf1b deficiency recapitulates many of the malformations described in MODY5 patients. All together, my results improved our understanding of the genetic program controlled by HNF1β during kidney development whose dysfunction may lead to CAKUT in patients
3

Activated HH Signaling: Deleterious Lineage-dependent Effects on Nephrogenesis and Collecting Duct Formation

Staite, Marian Vicky 11 January 2011 (has links)
Hedgehog (HH) signaling controls renal development. Mutations in PTC1, the HH receptor, cause cancer in non-renal tissues. We hypothesized that constitutively active HH signaling is deleterious to renal development in mice with PTC1 deficiency targeted to the metanephric mesenchyme (MM)(Rarb2-Cre;Ptc1 loxP/-, termed Ptc1 mutants). Increased HH signaling in MM of mutant mice was confirmed by qRT-PCR for Ptc1. A decrease in NCAM-positive nephrogenic precursors at E13.5 and WT1-positive glomeruli at E18.5 was found. Increased cortical expression of Foxd1 was observed. At E13.5, a cluster of ectopic cells expressing Raldh2, Ptc2 and Bmp4 accumulated at the presumptive uretero-pelvic junction (UPJ). Magnetic resonance imaging demonstrated an increase in pelvic volume. Constitutive expression of GLI3 repressor via the Gli3Δ699 allele in Ptc1 mutants increased nephron number comparable to wild type mice and decreased pelvic volume compared to Ptc1 mutants. Thus repression of HH activity is required for proper nephrogenesis and patterning of the UPJ.
4

Activated HH Signaling: Deleterious Lineage-dependent Effects on Nephrogenesis and Collecting Duct Formation

Staite, Marian Vicky 11 January 2011 (has links)
Hedgehog (HH) signaling controls renal development. Mutations in PTC1, the HH receptor, cause cancer in non-renal tissues. We hypothesized that constitutively active HH signaling is deleterious to renal development in mice with PTC1 deficiency targeted to the metanephric mesenchyme (MM)(Rarb2-Cre;Ptc1 loxP/-, termed Ptc1 mutants). Increased HH signaling in MM of mutant mice was confirmed by qRT-PCR for Ptc1. A decrease in NCAM-positive nephrogenic precursors at E13.5 and WT1-positive glomeruli at E18.5 was found. Increased cortical expression of Foxd1 was observed. At E13.5, a cluster of ectopic cells expressing Raldh2, Ptc2 and Bmp4 accumulated at the presumptive uretero-pelvic junction (UPJ). Magnetic resonance imaging demonstrated an increase in pelvic volume. Constitutive expression of GLI3 repressor via the Gli3Δ699 allele in Ptc1 mutants increased nephron number comparable to wild type mice and decreased pelvic volume compared to Ptc1 mutants. Thus repression of HH activity is required for proper nephrogenesis and patterning of the UPJ.
5

Genetic and molecular mechanisms regulating mammalian nephron endowment

Perl, Alison 23 August 2022 (has links)
No description available.
6

Mechanisms of epithelial branching, nephrogenesis, and the role of the Rho-GTPase family in kidney development

Lindström, Nils Olof January 2009 (has links)
The metanephric kidney consists of two types of epithelia; the Wolffian duct-derived ureteric bud and the nephrogenic components that originate from mesenchymal-toepithelial transitions in the metanephric mesenchyme. The ureteric bud forms when inductive signals from the metanephric mesenchyme stimulates the evagination of an epithelial tube from the Wolffian duct into the mesenchyme. Reciprocal signalling between the ureteric bud and the metanephric mesenchyme regulates the branching of the ureteric bud and the induction of nephron formation. Inductive and inhibitory signalling of ureteric bud growth and branching has been shown by several protein families, however, the mechanical aspects of ureteric bud branching and nephrogenesis are largely unknown. I investigated the roles of Rac1-GTPase and Rho-kinase during kidney development. These proteins are important regulators of the cytoskeleton where Rac1 is a promoter of actin filament polymerisation and Rho-kinase directly stimulates the formation and contraction of actin-myosin stress fibres. Using a cell-permeable inhibitor, Rac1 was inhibited with no effects on nephron formation or subsequent segmentation and patterning. Inhibition of active Rac1 significantly reduced the level of ureteric bud branching and also resulted in lower proliferation rates. Rho-kinase was similarly targeted using two inhibitors. Rho-kinase inhibition had important effects on nephron formation and nephron maturation. Inhibition of Rhokinase resulted in decreased levels of nephron formation and severely morphologically abnormal nephrons. The formation of apical-basal polarity was disturbed as was the development of the visceral and parietal epithelia; precursors of the renal corpuscle. Inhibition of Rho-kinase led to abnormal formation of the proximal-distal axis and abnormal segmentation of the nephron. The effects of Rho-kinase inhibition were partially mimicked by direct targeting of actin-myosin contractions using a myosin-ATPase inhibitor. This demonstrated that Rho-kinase is necessary during multiple stages of nephrogenesis and maturation, at least in part, as a result of its ability to regulate actin-myosin contraction. These results show that Rac1 and Rho-kinase play important roles during several aspects of kidney development and highlights the significance of further investigating the mechanisms involved during kidney organogenesis.
7

The crucial roles played by HNF1β during kidney development

Massa, Filippo Maria 14 November 2012 (has links) (PDF)
HNF1β is a transcription factor expressed during nephrogenesis in the ureteric bud and in the tubular epithelia derived from the metanephric mesenchyme. Mutations in HNF1B/MODY5 represent one of the most prevalent human genetic defects responsible for Congenital Abnormalities of Kidney and Urinary Tract (CAKUT). To investigate the role played by HNF1beta during kidney development, I made use of a CRE-LoxP strategy to inactivate Hnf1b in different compartments and at different time points during mouse kidney morphogenesis. My results showed that Hnf1b is required for the expression of several key genes including Wnt9b, Emx2, Pax2 and Lhx. These genes are normally expressed in the ureteric bud and their absence leads to the formation of rudimentary kidneys, characterized by abnormal ureteric bud branching and lack of mesenchymal to epithelial transition. On the other hand, the specific inactivation of Hnf1b in the metanephric mesenchyme does not affect the early steps of epithelisation but leads to severe malformation of nephron precursors. In the absence of HNF1β, S-Shaped bodies lack the formation of a bulge of epithelial cells that normally give rise to Henle's loops and proximal tubules. At the molecular level, these defects are associated with a decreased expression of Dll1, a Notch ligand that is directly controlled by HNF1β. This downregulation leads to a defective activation of the Notch pathway in the prospective tubular compartment of comma and S-shaped bodies. Preliminary results using a highly chimeric inactivation of Hnf1b in the mesenchyme showed that the spotted absence of Hnf1b does not prevent nephrogenesis. However, tubules are shorter and partially cystic. My results showed that murine models for Hnf1b deficiency recapitulates many of the malformations described in MODY5 patients. All together, my results improved our understanding of the genetic program controlled by HNF1β during kidney development whose dysfunction may lead to CAKUT in patients
8

Advanced in vivo imaging of the interactions between vascular and renal development in the zebrafish pronephric kidney

Verdon, Rachel Faye January 2015 (has links)
BACKGROUND Heart disease and renal dysfunction are often mutually reinforcing conditions, although the factors underlying this relationship are not fully understood. Cardiac remodelling resulting from disease is partly caused by the reactivation of developmental programmes. By unravelling the mechanisms that drive kidney development and function, it may be possible to gain novel insight into remodelled kidney states that are linked to disease. In this study, we have investigated the interplay between renal and cardiovascular systems during nephrogenesis at the level of the blood filter. METHODOLOGY Owing to the optical transparency and rapid external development of the embryo, the zebrafish provides a research model for advanced imaging technologies, allowing us to visualise structures located deep within living specimens. Here, we combine deep-tissue live imaging and novel functional assays to study development and function of the pronephric kidney - the first and most basic kidney to form in the embryo. RESULTS Using two-photon excitation microscopy, we have successfully established methodology for performing, deep-tissue, time-lapse imaging in living embryos of the two primary cell types forming the kidney – endothelial and epithelial cells. Fluorescence angiograms were performed using supra-vital dye agents to visualise circulatory flow in relation to the pronephric vasculature and the process of blood-filtration. Observations from live imaging studies, supported by immunostaining, were used to create a comprehensive model of the developing glomerular morphology, and interactions at the endothelial-epithelial cell interface, where glomerular epithelial primordia merge around the vascular component of the pronephric kidney. To investigate renal function, we devised a novel assay of pronephric filtration, by tracking the accumulation of injected fluorescent tracers within the excreted filtrate of embryos. This allowed us to relate our time-lapse observations to maturation of the glomerulus, and the evolution of perm-selective function. Finally, we explored methods of mechanically obstructing blood-flow in order to investigate whether altered hemodynamic forces would influence pronephric development. We found that in those embryos with severely disrupted circulatory flow, the glomerular morphology was affected. CONCLUSIONS In summary, the combination of these techniques has allowed us to visualise the multi-cellular organisation of the pronephric kidney over time, which has previously been limited to primarily fixed-tissue approaches. A detailed model of pronephric development has been developed, which could ultimately be used to dissect the molecular mechanisms underlying embryonic kidney development.
9

Role of Wnt11 in kidney ontogenesis and development of renal organoid based models to identify candidate oncogenes

Xu, Q. (Qi) 22 May 2018 (has links)
Abstract In the kidney, Wnt is involved in ureteric bud branching and nephrogenesis. Wnt mutation may lead to specific developmental dysfunctions and diseases. As part of this thesis, I show that Wnt11 is expressed in the renal tubules, except for the ureteric epitheliums, and I examine the function of Wnt11 in renal tubule organization using the new C57Bl6 Wnt11-/- mouse model. Convoluted and dilated tubules were observed in the Wnt11 mutated kidneys that may cause glomerular cysts and kidney dysfunction. More specifically, a lack of Wnt11 in the kidney reduced Six2, Hoxd1, and Hox10 expression, which may have contributed to the anomalies in the kidney tubular system. Embryogenesis and carcinogenesis share molecular characteristics. Gene expression changes take place during development to meet the demands of the tissue formation, but ectopic expression of embryonic genes by deletion, SNPs, or epigenetic modification in adult may lead to cancer. The research carried out as part of this thesis identified genes that were differentially expressed in both induced metanephric mesenchymes (MM) and human ccRCC. Gene silencing of Bnip3, Gsn, Lgals3, Pax8, Cav1, Egfr, and Itgb2 mediated by siRNA inhibited the migration, viability and invasion capacity of Renca cells (RCC). Furthermore, by using the novel 3D in vitro MM-Renca co-culture setup, the result revealed that downregulation of Bnip3, Cav1 or Gsn in Renca cells partly rescued the RCC-mediated inhibition epithelial tubules formation during nephrogenesis. Altogether, the data demonstrates that renal ontogenesis control genes play a role both in normal kidney development and in kidney cancer. The 3D RCC-MM chimera organoids developed as part of the research may also serve as a novel model for kidney cancer study. / Tiivistelmä Wnt-viestit ohjaavat munuaisen kehityksen yhteydessä sekä virtsanjohtimen että munuaiskeräsen kasvua. Virhe Wnt-proteiinia tuottavassa geenissä johtaa puolestaan vakavaan kehityshäiriöön ja syöpään, jos geeni aktivoituu aikuisvaiheessa. Tässä väitöskirjassa osoitetaan, että Wnt11-geeni osallistuu alkion munuaisen epiteeliputkiston kehityksen säätelyyn. Wnt11-signaalin tehtävää tutkittiin munuaisen putkiston rakenteen synnyssä poistogeenisen C57Bl6-hiirimallin avulla. Wnt11-puutos häiritsi virtsan kokoojatiehyiden rakenteiden kehitystä. Tämä on mahdollinen syy siihen, että Wnt11-poistogeenisen hiiren munuaiseen kehittyy hiusverisuonikerästen laajentumia. Munuaisen toiminta on myös heikentynyt verrokkiin verrattuna. Munuaisen epiteeliputkien kehitykseen osallistuvien geenien Six1, Hoxd1 ja Hox10 aktiivisuus oli niin ikään heikentynyt Wnt11-signaloinnin puutoksen vuoksi. Nämä seikat voivat olla puutoksen aiheuttamien kudosrakenteellisten muutosten taustalla. Solun kasvusäätely on keskeinen tekijä sekä alkion että syövän kehityksessä. Prosessien taustalla ovat myös samankaltaiset molekyylit. Wnt-solusignaalit esimerkiksi säätelevät normaalin kehityksen aikana solujen itsesäätelyä, solujen jakautumista, solujen vaeltamista ja solujen invaasiota kudoksiin ja osallistuvat syövän syntyyn niiden viestinnän häiriintyessä. Kehitystä säätelevien geenien tuotteet ohjaavat solujen jakautumista, mikä on edellytys kudosrakenteen synnylle. Jos nämä geenit syystä tai toisesta aktivoituvat uudelleen aikuisvaiheen aikana, se voi johtaa syöpäkasvaimen syntyyn. Tässä työssä verrattiin keskenään munuaisen elinkehitykseen ja ihmisen syövän kasvuun osallistuvia geenejä. siRNA-välitteinen hiljennys geeneille Bnip3, Gsn, Lgals3, Pax8, Cav1, Egfr ja Itgb2 inhiboi Renca-solujen migraatiota, elinkykyä ja invaasiota. Lisäksi tulokset paljastivat, että käytettäessä uutta 3D in vitro MM Renca -kasvatusmenetelmää Bnip3-, Cav1- tai Gsn-geenien hiljennys Renca-soluissa osittain pelastaa RCC-välitteisen inhibition epiteeliputkissa nefrogeneesin aikana. Kaiken kaikkiaan työssä seulottiin uusin tavoin sekä ihmisen munuaissyöpään liittyviä geenejä että kehitettiin lisäksi uusia niin kutsuttuja kokeellisia funktionaalisia organoidimenetelmiä. Työn tulokset tarjoavat uuden strategian, jolla geenien osuutta munuaissyöpään voidaan tutkia seikkaperäisesti yhdessä 3D-kasvatusmenetelmien kanssa
10

Studies of kidney induction <em>in vitro</em> using gene expression profiling and novel tissue manipulation technique

Junttila, S. (Sanna) 05 December 2014 (has links)
Abstract For decades, the mammalian kidney has served as a model system for studying developmental processes, such as induced epithelialization, branching morphogenesis, and cell differentiations. The possibility to recapitulate and follow the renal organogenesis ex vivo in organ culture set-ups has provided a large amount of molecular and cellular information about sequential events during development. However, certain limitations remain when combining traditional organ culture set-ups with modern molecular technology. This thesis seeks to address these disadvantages. In the experimental part of the thesis, the traditional organ culture set-ups were studied, modified, and optimized to meet the needs of functional genetic screening. First, the traditional transfilter- induced nephrogenesis was characterized with a panel of nephron segment specific markers to reveal the differentiation level of in vitro developing mouse renal tissue. A comprehensive genome wide time course microarray analysis was also performed to in vitro- induced metanephric mesenchyme. Next, to improve the accessibility of genetic tools into the three- dimensional organ in culture, the classic kidney culture set-ups were modified to tolerate dissociation and re-aggregation before the induction of nephrogenesis. This step was achieved with the aid of preservative growth factors offering a 24- hour window to manipulate the genetic and cellular composition of the explant. The dissociation and re-aggregation per se had not particular effect on the progress of the nephron differentiation. Demonstrations of the addition and removal of cells, as well as a virus vector mediated gene knock in and knock down are presented. The gene expression data, together with the novel organ manipulation and culture techniques presented in this thesis, provide a useful guide and specific tools to further characterize the details of nephron development and differentiation in functional manner. / Tiivistelmä Nisäkkäiden munuainen on toiminut vuosikymmeniä mallielimenä tutkittaessa kehitysbiologisia tapahtumasarjoja, kuten epitelisaatiota, haaroittumismorfologiaa sekä solujen erilaistumista. Munuaisaihioita voidaan viljellä laboratorio-olosuhteissa, jolloin kehityksen aikaisia muutoksia päästään seuraamaan lähes reaaliaikaisesti. Perinteisten kudosviljelytekniikoiden tarjoamat mahdollisuudet solujen molekulaariseen muokkaukseen ovat kuitenkin varsin rajalliset. Tässä väitöskirjassa esitettävät tulokset pyrkivät osaltaan vähentämään näitä rajoitteita. Väitöskirjan kokeellisessa osassa tarkastellaan lähemmin klassista munuaiskudosviljelyä sekä esitetään siihen tehtyjä optimointeja, joiden avulla kudosviljelyä pyritään hyödyntämään geenien toiminnan tutkimuksessa. Aluksi perinteisellä tavalla reikäisen kalvon läpi indusoitu nefroni karakterisoitiin tarkasti hyödyntäen useita erilaistumista osoittavia merkkimolekyylejä. Lisäksi samalla tekniikalla tuotettujen munuaiskudosviljelmien geeniekspressiota tutkittiin mikrosiruanalyysillä. Klassisia kudosviljelytekniikoita muokattiin soveltuvammaksi moderneille geneettisille työkaluille. Munuaiskudos hajotettiin ensin solususpensioksi, jonka jälkeen solut muodostivat uudelleen kolmiulotteisen, kudosmaisen rakenteen. Hyödyntämällä suojaavia kasvutekijöitä, hajotus kyettiin tekemään jo ennen nefronien muodostumisen alkua. Näin saavutettin 24 tunnin aikaikkuna indusoimattoman kudoksen geneettiselle muokkaukselle. Väitöskirjassa esitelläänkin demonsrtaatiot solujen lisäämisestä ja poistamisesta sekä virusvälitteisestä geenin aktivoinnista ja hiljennyksestä hyödyntäen uutta kudosmanipulaatio ja –vilejelytekniikkaa. Nefronin kehityksen aikaisen geeniekspression kartoitus sekä tässä tutkimuksessa kehitetyt uudet kudosmanipulaatio ja -viljelytekniikat tarjoavat yhdessä työkaluja molekyylitason yksityiskohtaiseen tutkimiseen.

Page generated in 0.0596 seconds