Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 243
Filtrar
2.
Kidney Int ; 102(1): 108-120, 2022 07.
Artigo em Inglês | MEDLINE | ID: mdl-35341793

RESUMO

Oxidative metabolism in mitochondria regulates cellular differentiation and gene expression through intermediary metabolites and reactive oxygen species. Its role in kidney development and pathogenesis is not completely understood. Here we inactivated ubiquinone-binding protein QPC, a subunit of mitochondrial complex III, in two types of kidney progenitor cells to investigate the role of mitochondrial electron transport in kidney homeostasis. Inactivation of QPC in sine oculis-related homeobox 2 (SIX2)-expressing cap mesenchyme progenitors, which give rise to podocytes and all nephron segments except collecting ducts, resulted in perinatal death from severe kidney dysplasia. This was characterized by decreased proliferation of SIX2 progenitors and their failure to differentiate into kidney epithelium. QPC inactivation in cap mesenchyme progenitors induced activating transcription factor 4-mediated nutritional stress responses and was associated with a reduction in kidney tricarboxylic acid cycle metabolites and amino acid levels, which negatively impacted purine and pyrimidine synthesis. In contrast, QPC inactivation in ureteric tree epithelial cells, which give rise to the kidney collecting system, did not inhibit ureteric differentiation, and resulted in the development of functional kidneys that were smaller in size. Thus, our data demonstrate that mitochondrial oxidative metabolism is critical for the formation of cap mesenchyme-derived nephron segments but dispensable for formation of the kidney collecting system. Hence, our studies reveal compartment-specific needs for metabolic reprogramming during kidney development.


Assuntos
Complexo III da Cadeia de Transporte de Elétrons , Rim , Néfrons , Organogênese , Podócitos , Aminoácidos/deficiência , Diferenciação Celular , Complexo III da Cadeia de Transporte de Elétrons/metabolismo , Feminino , Humanos , Rim/embriologia , Rim/metabolismo , Mesoderma/metabolismo , Néfrons/metabolismo , Organogênese/genética , Podócitos/metabolismo , Gravidez , Ureter/embriologia
3.
J Pathol ; 252(3): 290-303, 2020 11.
Artigo em Inglês | MEDLINE | ID: mdl-32715478

RESUMO

Development of the mammalian kidney is orchestrated by reciprocal interactions of stromal and nephrogenic mesenchymal cells with the ureteric bud epithelium. Previous work showed that the transcription factor Wilms tumor 1 (WT1) acts in the nephrogenic lineage to maintain precursor cells, to drive the epithelial transition of aggregating precursors into a renal vesicle and to specify and maintain the podocyte fate. However, WT1 is expressed not only in the nephrogenic lineage but also transiently in stromal progenitors in the renal cortex. Here we report that specific deletion of Wt1 in the stromal lineage using the Foxd1cre driver line results at birth in cryptorchidism and hypoplastic kidneys that harbour fewer and enlarged ureteric bud tips and display an expansion of capsular stroma into the cortical region. In vivo and ex vivo analysis at earlier stages revealed that stromal loss of Wt1 reduces stromal proliferation, and delays and alters branching morphogenesis, resulting in a variant architecture of the collecting duct tree with an increase of single at the expense of bifurcated ureteric bud tips. Molecular analysis identified a transient reduction of Aldh1a2 expression and of retinoic acid signalling activity in stromal progenitors, and of Ret in ureteric bud tips. Administration of retinoic acid partly rescued the branching defects of mutant kidneys in culture. We propose that WT1 maintains retinoic acid signalling in the cortical stroma, which, in turn, assures proper levels and dynamics of Ret expression in the ureteric bud tips, and thus normal ramification of the ureteric tree. © 2020 The Authors. The Journal of Pathology published by John Wiley & Sons, Ltd. on behalf of The Pathological Society of Great Britain and Ireland.


Assuntos
Criptorquidismo/embriologia , Criptorquidismo/genética , Deleção de Genes , Regulação da Expressão Gênica no Desenvolvimento , Rim/embriologia , Ureter/embriologia , Proteínas WT1/genética , Animais , Biomarcadores/metabolismo , Criptorquidismo/metabolismo , Rim/anormalidades , Rim/metabolismo , Masculino , Camundongos , Organogênese/genética , Ureter/anormalidades , Ureter/metabolismo , Proteínas WT1/metabolismo
4.
J Pediatr Urol ; 15(3): 240.e1-240.e7, 2019 May.
Artigo em Inglês | MEDLINE | ID: mdl-30850354

RESUMO

The etiology of ureterovesical junction obstruction (UVJO) and ureteropelvic junction obstruction (UPJO) is obscure with an adynamic narrow segment causing the obstruction. In this study, the authors compared interstitial cells of Cajal (ICC) and collagen-to-muscle ratio (CM ratio) between UVJO, UPJO, and fetal ureters to investigate whether a maturational arrest of the fetal ureter could explain both clinical pathologies. METHODS: Group 1 (control) involved specimens of the normal ureter (nephrectomy for trauma/tumor; n = 20), while group 2, specimens of UVJO (n = 14); group 2 was further divided into group 2a, the dilated megaureter above UVJO, and group 2b, UVJO narrow segment; group 3, UPJO narrow segment excised during pyeloplasty (n = 31); and group 4, normal fetal ureters (n = 12). The specimens were analyzed for ICC using immunohistochemistry and CM ratio on Masson's trichrome (stains collagen in blue and muscle in red). RESULTS: The median ICC/10 high-power field was 16.1 (8.3) in the normal and 17.3 (7.9) in the dilated segment of the megaureter, with no significant difference, but was significantly less in the narrow segment of UVJO at 4.5 (2.0), narrow segment of UPJO at 5.1 (2.3), and fetal ureter at 5.0 (2.3). The median CM ratio was 0.75 (0.29) in the normal and 0.65 (0.2) in the dilated segment of the megaureter, with no significant difference between them (figure), but was significantly higher in the narrow segment of UVJO at 3.0 (0.8), narrow segment of UPJO at 2.5 (0.71), and fetal ureter at 3.1 (0.61). Overall UVJO, UPJO, and fetal ureter segment had significantly less ICC density and more collagen compared with the normal ureter (P < 0.001 by Mann-Whitney U test). DISCUSSION: There are conflicting reports on the etiopathogenesis of UVJO and UPJO, with several authors showing decreased ICC and increased collagen in the narrow segment. In this study, the authors found that the pathological changes at UVJ and UPJ segments resemble fetal ureter morphology. We also found that in fetal ureters, as the gestation progressed, there was an increase in the ICC density/smooth muscle, whereas the collagen content decreased. While the entire ureter has uniform embryological origin, it essentially remains an epithelial tube until the late gestation. The maturational process involves differentiation of smooth muscles cells/ICC to establish the peristaltic machinery required to functionally connect the ureter at both ends. This process, probably, starts at the mid ureter during fetal life and extends toward the UPJ and UVJ, and its failure, probably, results in UPJO or UVJO. The study's limitations are small numbers, and further larger studies are required to validate this hypothesis.


Assuntos
Feto/patologia , Pelve Renal/patologia , Ureter/embriologia , Ureter/patologia , Obstrução Ureteral/patologia , Bexiga Urinária/patologia , Pré-Escolar , Colágeno/análise , Feminino , Humanos , Lactente , Células Intersticiais de Cajal/patologia , Gravidez , Estudos Prospectivos , Ureter/química
5.
J Pediatr Surg ; 53(12): 2502-2506, 2018 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-30503249

RESUMO

BACKGROUND: The kidney develops from an intimate interaction between the ureteric bud and the metanephric mass. We attempted to differentially stain the derivatives of the ureteric bud and the metanephric mass in ovine fetuses. METHODS: After appropriate approval, 47 fetal lambs' kidneys at 50 (4), 60 (6), 70 (5), 80 (4), 100 (10), 110 (8), 145 (10) days' gestation (term is 140-145 days) were obtained. After confirming the pregnancy, the sheep were anesthetized, and the fetuses sacrificed. The fetal kidneys were prepared for histological examination, using immunostaining for ß-catenin, Laminin, CK34ßE12, CK7, E-cadherin, and EMA. RESULTS: In the nephrogenic zone, positive staining was only seen for ß-catenin and Laminin. Areas with linear ß-catenin expression increased with increasing gestational age, whereas cytoplasmic granular expression in the nephrogenic zone diminished. At 50 days, Laminin-positive cells appeared in the ureteric bud epithelial cells, but not in the proximal tubule epithelium. They were found only in the immature collecting duct at 60 days. CONCLUSION: We have shown that the distribution of ß-catenin and Laminin positive-stained cells initially appearing in the ureteric bud changes with gestational age. Further studies may help inform the optimal timing of fetal shunt insertion in obstructive uropathy.


Assuntos
Rim/embriologia , Ovinos/embriologia , Ureter/embriologia , Animais , Caderinas/metabolismo , Feminino , Feto/embriologia , Imuno-Histoquímica , Queratina-7/metabolismo , Queratinas/metabolismo , Rim/metabolismo , Laminina/metabolismo , Mucina-1/metabolismo , Gravidez , Ureter/metabolismo , Via de Sinalização Wnt , beta Catenina/metabolismo
6.
J Am Soc Nephrol ; 29(4): 1198-1209, 2018 04.
Artigo em Inglês | MEDLINE | ID: mdl-29436516

RESUMO

Nonobstructive hydronephrosis, defined as dilatation of the renal pelvis with or without dilatation of the ureter, is the most common antenatal abnormality detected by fetal ultrasound. Yet, the etiology of nonobstructive hydronephrosis is poorly defined. We previously demonstrated that defective development of urinary tract pacemaker cells (utPMCs) expressing hyperpolarization-activated cyclic nucleotide-gated channel 3 (HCN3) and the stem cell marker cKIT causes abnormal ureteric peristalsis and nonobstructive hydronephrosis. However, further investigation of utPMC development and function is limited by lack of knowledge regarding the embryonic derivation, development, and molecular apparatus of these cells. Here, we used lineage tracing in mice to identify cells that give rise to utPMCs. Neural crest cells (NCCs) indelibly labeled with tdTomato expressed HCN3 and cKIT. Furthermore, purified HCN3+ and cKIT+ utPMCs were enriched in Sox10 and Tfap-2α, markers of NCCs. Sequencing of purified RNA from HCN3+ cells revealed enrichment of a small subset of RNAs, including RNA encoding protein kinase 2ß (PTK2ß), a Ca2+-dependent tyrosine kinase that regulates ion channel activity in neurons. Immunofluorescence analysis in situ revealed PTK2ß expression in NCCs as early as embryonic day 12.5 and in HCN3+ and cKIT+ utPMCs as early as embryonic day 15.5, with sustained expression in HCN3+ utPMCs until postnatal week 8. Pharmacologic inhibition of PTK2ß in murine pyeloureteral tissue explants inhibited contraction frequency. Together, these results demonstrate that utPMCs are derived from NCCs, identify new markers of utPMCs, and demonstrate a functional contribution of PTK2ß to utPMC function.


Assuntos
Quinase 2 de Adesão Focal/fisiologia , Regulação da Expressão Gênica no Desenvolvimento , Células Intersticiais de Cajal/enzimologia , Pelve Renal/fisiologia , Crista Neural/enzimologia , Peristaltismo/fisiologia , Ureter/fisiologia , Animais , Antígenos de Diferenciação/análise , Quinase 2 de Adesão Focal/biossíntese , Quinase 2 de Adesão Focal/genética , Genes Reporter , Idade Gestacional , Hidronefrose/enzimologia , Hidronefrose/fisiopatologia , Canais Disparados por Nucleotídeos Cíclicos Ativados por Hiperpolarização/análise , Células Intersticiais de Cajal/fisiologia , Pelve Renal/citologia , Pelve Renal/embriologia , Pelve Renal/crescimento & desenvolvimento , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Crista Neural/fisiologia , Canais de Potássio/análise , Proteínas Proto-Oncogênicas c-kit/análise , RNA Mensageiro/biossíntese , Fatores de Transcrição SOXE/análise , Transdução de Sinais , Fator de Transcrição AP-2/análise , Ureter/citologia , Ureter/embriologia , Ureter/crescimento & desenvolvimento
7.
J Am Soc Nephrol ; 29(3): 775-783, 2018 03.
Artigo em Inglês | MEDLINE | ID: mdl-29326158

RESUMO

The epithelial Wolffian duct (WD) inserts into the cloaca (primitive bladder) before metanephric kidney development, thereby establishing the initial plumbing for eventual joining of the ureters and bladder. Defects in this process cause common anomalies in the spectrum of congenital anomalies of the kidney and urinary tract (CAKUT). However, developmental, cellular, and molecular mechanisms of WD-cloaca fusion are poorly understood. Through systematic analysis of early WD tip development in mice, we discovered that a novel process of spatiotemporally regulated apoptosis in WD and cloaca was necessary for WD-cloaca fusion. Aberrant RET tyrosine kinase signaling through tyrosine (Y) 1062, to which PI3K- or ERK-activating proteins dock, or Y1015, to which PLCγ docks, has been shown to cause CAKUT-like defects. Cloacal apoptosis did not occur in RetY1062F mutants, in which WDs did not reach the cloaca, or in RetY1015F mutants, in which WD tips reached the cloaca but did not fuse. Moreover, inhibition of ERK or apoptosis prevented WD-cloaca fusion in cultures, and WD-specific genetic deletion of YAP attenuated cloacal apoptosis and WD-cloacal fusion in vivo Thus, cloacal apoptosis requires direct contact and signals from the WD tip and is necessary for WD-cloacal fusion. These findings may explain the mechanisms of many CAKUT.


Assuntos
Apoptose/genética , Cloaca/embriologia , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Proteínas Proto-Oncogênicas c-ret/genética , Anormalidades Urogenitais/genética , Ductos Mesonéfricos/embriologia , Proteínas Adaptadoras de Transdução de Sinal/genética , Animais , Proteínas de Ciclo Celular , Cloaca/anormalidades , Cloaca/metabolismo , Rim/embriologia , Sistema de Sinalização das MAP Quinases , Camundongos , Mutação , Fosfoproteínas/genética , Proteínas Proto-Oncogênicas c-ret/metabolismo , Ureter/embriologia , Ductos Mesonéfricos/anormalidades , Ductos Mesonéfricos/metabolismo , Proteínas de Sinalização YAP
8.
J Am Soc Nephrol ; 29(2): 532-544, 2018 02.
Artigo em Inglês | MEDLINE | ID: mdl-29109083

RESUMO

Intrinsic ureteropelvic junction obstruction is the most common cause of congenital hydronephrosis, yet the underlying pathogenesis is undefined. Hedgehog proteins control morphogenesis by promoting GLI-dependent transcriptional activation and inhibiting the formation of the GLI3 transcriptional repressor. Hedgehog regulates differentiation and proliferation of ureteric smooth muscle progenitor cells during murine kidney-ureter development. Histopathologic findings of smooth muscle cell hypertrophy and stroma-like cells, consistently observed in obstructing tissue at the time of surgical correction, suggest that Hedgehog signaling is abnormally regulated during the genesis of congenital intrinsic ureteropelvic junction obstruction. Here, we demonstrate that constitutively active Hedgehog signaling in murine intermediate mesoderm-derived renal progenitors results in hydronephrosis and failure to develop a patent pelvic-ureteric junction. Tissue obstructing the ureteropelvic junction was marked as early as E13.5 by an ectopic population of cells expressing Ptch2, a Hedgehog signaling target. Constitutive expression of GLI3 repressor in Ptch1-deficient mice rescued ectopic Ptch2 expression and obstructive hydronephrosis. Whole transcriptome analysis of isolated Ptch2+ cells revealed coexpression of genes characteristic of stromal progenitor cells. Genetic lineage tracing indicated that stromal cells blocking the ureteropelvic junction were derived from intermediate mesoderm-derived renal progenitors and were distinct from the smooth muscle or epithelial lineages. Analysis of obstructive ureteric tissue resected from children with congenital intrinsic ureteropelvic junction obstruction revealed a molecular signature similar to that observed in Ptch1-deficient mice. Together, these results demonstrate a Hedgehog-dependent mechanism underlying mammalian intrinsic ureteropelvic junction obstruction.


Assuntos
Proteínas Hedgehog/genética , Hidronefrose/genética , Proteínas do Tecido Nervoso/genética , Receptor Patched-1/genética , Receptor Patched-2/genética , Transdução de Sinais , Obstrução Ureteral/genética , Proteína Gli3 com Dedos de Zinco/genética , Aldeído Oxirredutases/genética , Animais , Linhagem da Célula , Criança , Feminino , Fatores de Transcrição Forkhead/genética , Expressão Gênica , Proteínas Hedgehog/metabolismo , Humanos , Hidronefrose/congênito , Hidronefrose/patologia , Hibridização In Situ , Pelve Renal/embriologia , Pelve Renal/metabolismo , Masculino , Mesoderma/embriologia , Mesoderma/metabolismo , Camundongos , Camundongos Knockout , Proteínas do Tecido Nervoso/metabolismo , Células-Tronco/metabolismo , Fatores de Transcrição/genética , Transcrição Gênica , Transcriptoma , Regulação para Cima , Ureter/embriologia , Ureter/metabolismo , Obstrução Ureteral/congênito , Obstrução Ureteral/patologia , Proteína Gli3 com Dedos de Zinco/metabolismo
9.
Development ; 144(24): 4704-4719, 2017 12 15.
Artigo em Inglês | MEDLINE | ID: mdl-29158444

RESUMO

Kidney development depends crucially on proper ureteric bud branching giving rise to the entire collecting duct system. The transcription factor HNF1B is required for the early steps of ureteric bud branching, yet the molecular and cellular events regulated by HNF1B are poorly understood. We report that specific removal of Hnf1b from the ureteric bud leads to defective cell-cell contacts and apicobasal polarity during the early branching events. High-resolution ex vivo imaging combined with a membranous fluorescent reporter strategy show decreased mutant cell rearrangements during mitosis-associated cell dispersal and severe epithelial disorganization. Molecular analysis reveals downregulation of Gdnf-Ret pathway components and suggests that HNF1B acts both upstream and downstream of Ret signaling by directly regulating Gfra1 and Etv5 Subsequently, Hnf1b deletion leads to massively mispatterned ureteric tree network, defective collecting duct differentiation and disrupted tissue architecture, which leads to cystogenesis. Consistently, mRNA-seq analysis shows that the most impacted genes encode intrinsic cell-membrane components with transporter activity. Our study uncovers a fundamental and recurring role of HNF1B in epithelial organization during early ureteric bud branching and in further patterning and differentiation of the collecting duct system in mouse.


Assuntos
Polaridade Celular/genética , Fator 1-beta Nuclear de Hepatócito/genética , Túbulos Renais Coletores/embriologia , Ureter/embriologia , Anormalidades Urogenitais/embriologia , Anormalidades Urogenitais/genética , Animais , Adesão Celular/genética , Células Cultivadas , Proteínas de Ligação a DNA/metabolismo , Regulação para Baixo/genética , Fator Neurotrófico Derivado de Linhagem de Célula Glial/metabolismo , Receptores de Fator Neurotrófico Derivado de Linhagem de Célula Glial/metabolismo , Fator 1-beta Nuclear de Hepatócito/metabolismo , Camundongos , Camundongos Knockout , Proteínas Nucleares/metabolismo , Técnicas de Cultura de Órgãos , Fator de Transcrição PAX2/biossíntese , Transdução de Sinais/genética , Fatores de Transcrição/metabolismo , Ubiquitina-Proteína Ligases
10.
Sci Rep ; 7(1): 14803, 2017 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-29093497

RESUMO

The differentiated cell types of the mature ureter arise from the distal ureteric bud epithelium and its surrounding mesenchyme. Uncommitted epithelial cells first become intermediate cells from which both basal and superficial cells develop. Mesenchymal progenitors give rise to separated layers of adventitial fibrocytes, smooth muscle cells and lamina propria fibrocytes. How progenitor expansion and differentiation are balanced is poorly understood. Here, we addressed the role of retinoic acid (RA) signaling in these programs. Using expression analysis of components and target genes, we show that pathway activity is restricted to the mesenchymal and epithelial progenitor pools. Inhibition of RA signaling in ureter explant cultures resulted in tissue hypoplasia with a relative expansion of smooth muscle cells at the expense of lamina propria fibroblasts in the mesenchyme, and of superficial cells at the expense of intermediate cells in the ureteric epithelium. Administration of RA led to a slight reduction of smooth muscle cells, and almost completely prevented differentiation of intermediate cells into basal and superficial cells. We identified cellular programs and transcriptional targets of RA signaling that may account for this activity. We conclude that RA signaling is required and sufficient to maintain mesenchymal and epithelial progenitors in early ureter development.


Assuntos
Embrião de Mamíferos/embriologia , Células Epiteliais/metabolismo , Células-Tronco Mesenquimais/metabolismo , Transdução de Sinais/efeitos dos fármacos , Tretinoína/farmacologia , Ureter/embriologia , Animais , Regulação da Expressão Gênica no Desenvolvimento/efeitos dos fármacos , Camundongos , Camundongos Transgênicos , Miócitos de Músculo Liso/metabolismo
11.
Pediatr Res ; 82(6): 1022-1029, 2017 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-29135976

RESUMO

BackgroundFibroblast growth factor receptor 2 (Fgfr2) deletion from murine peri-Wolffian duct stroma (ST) results in aberrant ureteric bud induction, abnormal ureteral insertion into the bladder, and high rates of vesicoureteral reflux (VUR). It is unclear which receptor docking protein(s) is/are responsible for Fgfr2 actions in these tissues. We investigated whether the docking protein, fibroblast receptor substrate 2α (Frs2α), had a role in peri-Wolffian duct ST similar to Fgfr2.MethodsWe conditionally deleted Frs2α in peri-Wolffian duct ST with a Tbx18cre mouse line (Frs2αST-/-). We assessed for ureteric induction defects and alterations in downstream targets mediating defects. We performed euthanized cystograms and assessed ureter-bladder junctions by three-dimensional (3D) reconstructions.ResultsEmbryonic day (E) 11.5 Frs2αST-/- embryos had many displaced ureteric bud induction sites when compared with controls. E11.0 Frs2αST-/- embryos had decreased Bmp4 expression and signaling, which can cause abnormal ureteric bud induction. Postnatal day 1 (P1) and P30 Frs2αST-/- mice had higher VUR rates and grades vs. CONTROLS: Mutant refluxing ureters that inserted improperly into the bladder had shortened intravesicular tunnels (IVTs) when compared with controlsConclusionFrs2αST-/- embryos have aberrant ureteric induction sites, improper ureteral insertion, shortened intravesicular lengths, and VUR. Induction site defects appear secondary to reduced Bmp4 expression, similar to Fgfr2 mutants.


Assuntos
Proteínas de Membrana/genética , Ureter/embriologia , Refluxo Vesicoureteral/genética , Ductos Mesonéfricos/metabolismo , Animais , Apoptose , Proteína Morfogenética Óssea 4/genética , Proliferação de Células , Camundongos , Camundongos Knockout , Ureter/patologia
12.
Development ; 144(22): 4148-4158, 2017 11 15.
Artigo em Inglês | MEDLINE | ID: mdl-28993400

RESUMO

Kidney collecting system development requires integrin-dependent cell-extracellular matrix interactions. Integrins are heterodimeric transmembrane receptors consisting of α and ß subunits; crucial integrins in the kidney collecting system express the ß1 subunit. The ß1 cytoplasmic tail has two NPxY motifs that mediate functions by binding to cytoplasmic signaling and scaffolding molecules. Talins, scaffolding proteins that bind to the membrane proximal NPxY motif, are proposed to activate integrins and to link them to the actin cytoskeleton. We have defined the role of talin binding to the ß1 proximal NPxY motif in the developing kidney collecting system in mice that selectively express a Y-to-A mutation in this motif. The mice developed a hypoplastic dysplastic collecting system. Collecting duct cells expressing this mutation had moderate abnormalities in cell adhesion, migration, proliferation and growth factor-dependent signaling. In contrast, mice lacking talins in the developing ureteric bud developed kidney agenesis and collecting duct cells had severe cytoskeletal, adhesion and polarity defects. Thus, talins are essential for kidney collecting duct development through mechanisms that extend beyond those requiring binding to the ß1 integrin subunit NPxY motif.


Assuntos
Integrina beta1/metabolismo , Morfogênese , Talina/metabolismo , Ureter/citologia , Ureter/embriologia , Junções Aderentes/metabolismo , Motivos de Aminoácidos , Animais , Sítios de Ligação , Adesão Celular , Membrana Celular/metabolismo , Polaridade Celular , Regulação da Expressão Gênica no Desenvolvimento , Integrina beta1/química , Túbulos Renais Coletores/citologia , Túbulos Renais Coletores/embriologia , Camundongos Endogâmicos C57BL , Mutação/genética , Proteínas de Junções Íntimas/genética , Proteínas de Junções Íntimas/metabolismo , Ureter/metabolismo
13.
Development ; 144(23): 4377-4385, 2017 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-29038307

RESUMO

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.


Assuntos
Rim/embriologia , Morfogênese/fisiologia , Ureter/embriologia , Proteínas Adaptadoras de Transdução de Sinal/deficiência , Proteínas Adaptadoras de Transdução de Sinal/genética , Proteínas Adaptadoras de Transdução de Sinal/fisiologia , Animais , Padronização Corporal/genética , Padronização Corporal/fisiologia , Proteína Morfogenética Óssea 7/deficiência , Proteína Morfogenética Óssea 7/genética , Proteína Morfogenética Óssea 7/fisiologia , Imageamento Tridimensional , Conceitos Matemáticos , Proteínas de Membrana/deficiência , Proteínas de Membrana/genética , Proteínas de Membrana/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Mutantes , Modelos Biológicos , Morfogênese/genética , Mutação , Fenótipo , Fosfoproteínas/deficiência , Fosfoproteínas/genética , Fosfoproteínas/fisiologia , Fator de Crescimento Transformador beta2/deficiência , Fator de Crescimento Transformador beta2/genética , Fator de Crescimento Transformador beta2/fisiologia
14.
Hum Mol Genet ; 26(18): 3553-3563, 2017 09 15.
Artigo em Inglês | MEDLINE | ID: mdl-28655168

RESUMO

Heterozygous loss of Bmp4 results both in humans and mice in severe malformation of the urinary tract. These defects have at least partially been attributed to loss of expression of Bmp4 in the ureteric mesenchyme, yet the cellular and molecular function of this signal as well as its effector pathways in this tissue have remained incompletely resolved. Here, we show that mice with a conditional deletion of Bmp4 in the ureteric mesenchyme exhibited hydroureter and hydronephrosis at newborn stages due to functional and physical ureter obstruction. Proliferation in both the mesenchymal and epithelial progenitor pools was severely reduced and smooth muscle cell and urothelial differentiation programs were not activated. Epithelial expression of P-ERK1/2, P-AKT and P-P38, and mesenchymal expression of P-SMAD1/5/9, P-P38 and P-AKT were abrogated. Pharmacological inhibition and activation experiments in ureter cultures defined AKT as the most relevant downstream effector for epithelial and mesenchymal proliferation as well as for epithelial differentiation. Epithelial proliferation and differentiation were also influenced by P-38 and ERK1/2, while SMAD signaling, together with AKT and P-38, were required for smooth muscle cell differentiation. Our analysis suggests that BMP4 is the signal that couples the proliferation and differentiation programs in the epithelial and mesenchymal tissue compartments of the developing ureter by different downstream effectors, most importantly AKT and SMAD.


Assuntos
Proteína Morfogenética Óssea 4/metabolismo , Ureter/metabolismo , Animais , Proteína Morfogenética Óssea 4/genética , Diferenciação Celular/genética , Proliferação de Células , Células Epiteliais/metabolismo , Feminino , Regulação da Expressão Gênica no Desenvolvimento/genética , Masculino , Mesoderma/metabolismo , Camundongos , Miócitos de Músculo Liso/metabolismo , Organogênese/genética , Gravidez , Transdução de Sinais/genética , Ureter/embriologia
15.
J Am Soc Nephrol ; 28(1): 250-259, 2017 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-27432740

RESUMO

The renal vasculature is integral to the physiologic function of the kidneys in regulating hemodynamics of the body and maintaining organ health. The close inter-relationship of capillaries and the renal epithelium is key to renal physiology, but how renal tubules regulate capillary development remains unclear. Our previous work showed that Wnt7b is expressed in the ureteric trunk epithelium and activates canonical Wnt signaling in the surrounding medullary interstitium, where the capillaries reside. In this study, we showed by immunofluorescence that the target interstitial cells of Wnt7b/canonical Wnt signaling are mural cells of periureteric bud capillaries in the nascent renal medulla of embryonic mice. Genetic ablation of Wnt7b enhanced the proliferation of Wnt7b target mural cells, an effect that associated with decreased expression of PDGFRß and p57kip2, a cyclin-dependent kinase inhibitor, in these cells. Furthermore, Wnt7b regulated lumen formation of the capillary endothelium in the renal medulla. In the absence of Wnt7b signaling, the periureteric bud medullary capillaries displayed narrower lumens lined with less flattened endothelial cells and a significantly increased presence of luminal endothelial cell-cell junctions, a transient configuration in the forming blood vessels in the controls. Moreover, the absence of Wnt7b led to greatly diminished levels of vascular endothelial (VE)-cadherin at the cell surface in these blood vessels. VE-cadherin is essential for blood vessel lumen formation; thus, Wnt7b may regulate lumen formation through modulation of VE-cadherin localization. Overall, these results indicate a novel role of Wnt7b signaling and the ureteric bud epithelium in renal medullary capillary development.


Assuntos
Capilares/embriologia , Medula Renal/irrigação sanguínea , Organogênese , Proteínas Proto-Oncogênicas/fisiologia , Transdução de Sinais , Ureter/embriologia , Proteínas Wnt/fisiologia , Animais , Epitélio/embriologia , Camundongos
16.
J Am Soc Nephrol ; 28(6): 1792-1801, 2017 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-28028137

RESUMO

The mammalian ureter consists of a mesenchymal wall composed of smooth muscle cells and surrounding fibrocytes of the tunica adventitia and the lamina propria and an inner epithelial lining composed of layers of basal, intermediate, and superficial cells. How these cell types arise from multipotent progenitors is poorly understood. Here, we performed marker analysis, cell proliferation assays, and genetic lineage tracing to define the lineage relations and restrictions of the mesenchymal and epithelial cell types in the developing and mature mouse ureter. At embryonic day (E) 12.5, the mesenchymal precursor pool began to subdivide into an inner and outer compartment that began to express markers of smooth muscle precursors and adventitial fibrocytes, respectively, by E13.5. Smooth muscle precursors further diversified into lamina propria cells directly adjacent to the ureteric epithelium and differentiated smooth muscle cells from E16.5 onwards. Uncommitted epithelial progenitors of the ureter differentiated into intermediate cells at E14.5. After stratification into two layers at E15.5 and three cell layers at E18.5, intermediate cells differentiated into basal cells and superficial cells. In homeostasis, proliferation of all epithelial and mesenchymal cell types remained low but intermediate cells still gave rise to basal cells, whereas basal cells divided only into basal cells. These studies provide a framework to further determine the molecular mechanisms of cell differentiation in the tissues of the developing ureter.


Assuntos
Linhagem da Célula/fisiologia , Músculo Liso/citologia , Ureter/embriologia , Animais , Diferenciação Celular , Células Epiteliais , Mesoderma/citologia , Camundongos , Ureter/citologia
17.
Am J Physiol Renal Physiol ; 312(3): F407-F417, 2017 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-28031172

RESUMO

The prorenin receptor (PRR) is a receptor for renin and prorenin, and an accessory subunit of the vacuolar proton pump H+-ATPase. Renal branching morphogenesis, defined as growth and branching of the ureteric bud (UB), is essential for mammalian kidney development. Previously, we demonstrated that conditional ablation of the PRR in the UB in PRRUB-/- mice causes severe defects in UB branching, resulting in marked kidney hypoplasia at birth. Here, we investigated the UB transcriptome using whole genome-based analysis of gene expression in UB cells, FACS-isolated from PRRUB-/-, and control kidneys at birth (P0) to determine the primary role of the PRR in terminal differentiation and growth of UB-derived collecting ducts. Three genes with expression in UB cells that previously shown to regulate UB branching morphogenesis, including Wnt9b, ß-catenin, and Fgfr2, were upregulated, whereas the expression of Wnt11, Bmp7, Etv4, and Gfrα1 was downregulated. We next demonstrated that infection of immortalized UB cells with shPRR in vitro or deletion of the UB PRR in double-transgenic PRRUB-/-/BatGal+ mice, a reporter strain for ß-catenin transcriptional activity, in vivo increases ß-catenin activity in the UB epithelia. In addition to UB morphogenetic genes, the functional groups of differentially expressed genes within the downregulated gene set included genes involved in molecular transport, metabolic disease, amino acid metabolism, and energy production. Together, these data demonstrate that UB PRR performs essential functions during UB branching and collecting duct morphogenesis via control of a hierarchy of genes that control UB branching and terminal differentiation of the collecting duct cells.


Assuntos
Túbulos Renais Coletores/metabolismo , Morfogênese , Receptores de Superfície Celular/metabolismo , Ureter/metabolismo , Proteínas Wnt/metabolismo , Via de Sinalização Wnt , beta Catenina/metabolismo , Animais , Animais Recém-Nascidos , Proteína Morfogenética Óssea 7/genética , Proteína Morfogenética Óssea 7/metabolismo , Diferenciação Celular , Linhagem da Célula , Separação Celular/métodos , Biologia Computacional , Citometria de Fluxo , Perfilação da Expressão Gênica/métodos , Regulação da Expressão Gênica no Desenvolvimento , Redes Reguladoras de Genes , Genótipo , Receptores de Fator Neurotrófico Derivado de Linhagem de Célula Glial/genética , Receptores de Fator Neurotrófico Derivado de Linhagem de Célula Glial/metabolismo , Túbulos Renais Coletores/embriologia , Camundongos Knockout , Fenótipo , Proteínas Proto-Oncogênicas c-ets/genética , Proteínas Proto-Oncogênicas c-ets/metabolismo , Receptor Tipo 2 de Fator de Crescimento de Fibroblastos/genética , Receptor Tipo 2 de Fator de Crescimento de Fibroblastos/metabolismo , Receptores de Superfície Celular/deficiência , Receptores de Superfície Celular/genética , Transcriptoma , Ureter/embriologia , Proteínas Wnt/genética , beta Catenina/genética , Receptor de Pró-Renina
18.
Nat Commun ; 7: 12309, 2016 08 02.
Artigo em Inglês | MEDLINE | ID: mdl-27480037

RESUMO

Branching morphogenesis is a complex biological process common to the development of most epithelial organs. Here we demonstrate that NF2, LATS1/2 and YAP play a critical role in branching morphogenesis in the mouse kidney. Removal of Nf2 or Lats1/2 from the ureteric bud (UB) lineage causes loss of branching morphogenesis that is rescued by loss of one copy of Yap and Taz, and phenocopied by YAP overexpression. Mosaic analysis demonstrates that cells with high YAP expression have reduced contribution to UB tips, similar to Ret(-/-) cells, and that YAP suppresses RET signalling and tip identity. Conversely, Yap/Taz UB-deletion leads to cyst-like branching and expansion of UB tip markers, suggesting a shift towards tip cell identity. Based on these data we propose that NF2 and the Hippo pathway locally repress YAP/TAZ activity in the UB to promote subsequent splitting of the tip to allow branching morphogenesis.


Assuntos
Rim/metabolismo , Morfogênese/genética , Neurofibromina 2/genética , Proteínas Serina-Treonina Quinases/genética , Transdução de Sinais/genética , Proteínas Adaptadoras de Transdução de Sinal/genética , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Animais , Proteínas de Ciclo Celular , Via de Sinalização Hippo , Rim/embriologia , Camundongos Knockout , Camundongos Transgênicos , Neurofibromina 2/metabolismo , Fosfoproteínas/genética , Fosfoproteínas/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas Supressoras de Tumor/genética , Proteínas Supressoras de Tumor/metabolismo , Ureter/embriologia , Ureter/metabolismo , Proteínas de Sinalização YAP
19.
Dev Biol ; 418(2): 297-306, 2016 10 15.
Artigo em Inglês | MEDLINE | ID: mdl-27346698

RESUMO

Morphogenesis of the mammalian kidney requires reciprocal interactions between two cellular domains at the periphery of the developing organ: the tips of the epithelial ureteric tree and adjacent regions of cap mesenchyme. While the presence of the cap mesenchyme is essential for ureteric branching, how it is specifically maintained at the tips is unclear. Using ex vivo timelapse imaging we show that cells of the cap mesenchyme are highly motile. Individual cap mesenchyme cells move within and between cap domains. They also attach and detach from the ureteric tip across time. Timelapse tracks collected for >800 cells showed evidence that this movement was largely stochastic, with cell autonomous migration influenced by opposing attractive, repulsive and cell adhesion cues. The resulting swarming behaviour maintains a distinct cap mesenchyme domain while facilitating dynamic remodelling in response to underlying changes in the tip.


Assuntos
Rim/citologia , Rim/embriologia , Células-Tronco Mesenquimais/citologia , Ureter/citologia , Ureter/embriologia , Animais , Adesão Celular/fisiologia , Movimento Celular/fisiologia , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Rim/metabolismo , Células-Tronco Mesenquimais/fisiologia , Camundongos , Camundongos Transgênicos , Microscopia de Fluorescência , Morfogênese/fisiologia , Técnicas de Cultura de Órgãos , Processos Estocásticos , Imagem com Lapso de Tempo
20.
PLoS One ; 11(1): e0147171, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-26794322

RESUMO

The Wnts can be considered as candidates for the Congenital Anomaly of Kidney and Urinary Tract, CAKUT diseases since they take part in the control of kidney organogenesis. Of them Wnt5a is expressed in ureteric bud (UB) and its deficiency leads to duplex collecting system (13/90) uni- or bilateral kidney agenesis (10/90), hypoplasia with altered pattern of ureteric tree organization (42/90) and lobularization defects with partly fused ureter trunks (25/90) unlike in controls. The UB had also notably less tips due to Wnt5a deficiency being at E15.5 306 and at E16.5 765 corresponding to 428 and 1022 in control (p<0.02; p<0.03) respectively. These changes due to Wnt5a knock out associated with anomalies in the ultrastructure of the UB daughter epithelial cells. The basement membrane (BM) was malformed so that the BM thickness increased from 46.3 nm to 71.2 nm (p<0.01) at E16.5 in the Wnt5a knock out when compared to control. Expression of a panel of BM components such as laminin and of type IV collagen was also reduced due to the Wnt5a knock out. The P4ha1 gene that encodes a catalytic subunit of collagen prolyl 4-hydroxylase I (C-P4H-I) in collagen synthesis expression and the overall C-P4H enzyme activity were elevated by around 26% due to impairment in Wnt5a function from control. The compound Wnt5a+/-;P4ha1+/- embryos demonstrated Wnt5a-/- related defects, for example local hyperplasia in the UB tree. A R260H WNT5A variant was identified from renal human disease cohort. Functional studies of the consequence of the corresponding mouse variant in comparison to normal ligand reduced Wnt5a-signalling in vitro. Together Wnt5a has a novel function in kidney organogenesis by contributing to patterning of UB derived collecting duct development contributing putatively to congenital disease.


Assuntos
Membrana Basal/patologia , Células Epiteliais/citologia , Túbulos Renais Coletores/patologia , Ureter/embriologia , Ureter/metabolismo , Anormalidades Urogenitais/fisiopatologia , Refluxo Vesicoureteral/fisiopatologia , Proteínas Wnt/fisiologia , Adolescente , Animais , Membrana Basal/metabolismo , Células Cultivadas , Criança , Embrião de Mamíferos/citologia , Embrião de Mamíferos/metabolismo , Células Epiteliais/metabolismo , Feminino , Humanos , Técnicas Imunoenzimáticas , Hibridização In Situ , Túbulos Renais Coletores/metabolismo , Camundongos , Camundongos Knockout , Morfogênese , Mutação/genética , Conformação Proteica , RNA Mensageiro/genética , Reação em Cadeia da Polimerase em Tempo Real , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Proteínas Wnt/química , Proteína Wnt-5a , Proteína Wnt4/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA