Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 5.294
Filtrar
Mais filtros

Eixos temáticos
Intervalo de ano de publicação
1.
Cell ; 171(1): 242-255.e27, 2017 Sep 21.
Artigo em Inglês | MEDLINE | ID: mdl-28938116

RESUMO

The morphogenesis of branched organs remains a subject of abiding interest. Although much is known about the underlying signaling pathways, it remains unclear how macroscopic features of branched organs, including their size, network topology, and spatial patterning, are encoded. Here, we show that, in mouse mammary gland, kidney, and human prostate, these features can be explained quantitatively within a single unifying framework of branching and annihilating random walks. Based on quantitative analyses of large-scale organ reconstructions and proliferation kinetics measurements, we propose that morphogenesis follows from the proliferative activity of equipotent tips that stochastically branch and randomly explore their environment but compete neutrally for space, becoming proliferatively inactive when in proximity with neighboring ducts. These results show that complex branched epithelial structures develop as a self-organized process, reliant upon a strikingly simple but generic rule, without recourse to a rigid and deterministic sequence of genetically programmed events.


Assuntos
Rim/crescimento & desenvolvimento , Glândulas Mamárias Humanas/crescimento & desenvolvimento , Modelos Biológicos , Morfogênese , Próstata/crescimento & desenvolvimento , Animais , Feminino , Humanos , Rim/embriologia , Masculino , Glândulas Mamárias Humanas/embriologia , Camundongos , Próstata/embriologia
2.
Nature ; 626(8001): 1084-1093, 2024 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-38355799

RESUMO

The house mouse (Mus musculus) is an exceptional model system, combining genetic tractability with close evolutionary affinity to humans1,2. Mouse gestation lasts only 3 weeks, during which the genome orchestrates the astonishing transformation of a single-cell zygote into a free-living pup composed of more than 500 million cells. Here, to establish a global framework for exploring mammalian development, we applied optimized single-cell combinatorial indexing3 to profile the transcriptional states of 12.4 million nuclei from 83 embryos, precisely staged at 2- to 6-hour intervals spanning late gastrulation (embryonic day 8) to birth (postnatal day 0). From these data, we annotate hundreds of cell types and explore the ontogenesis of the posterior embryo during somitogenesis and of kidney, mesenchyme, retina and early neurons. We leverage the temporal resolution and sampling depth of these whole-embryo snapshots, together with published data4-8 from earlier timepoints, to construct a rooted tree of cell-type relationships that spans the entirety of prenatal development, from zygote to birth. Throughout this tree, we systematically nominate genes encoding transcription factors and other proteins as candidate drivers of the in vivo differentiation of hundreds of cell types. Remarkably, the most marked temporal shifts in cell states are observed within one hour of birth and presumably underlie the massive physiological adaptations that must accompany the successful transition of a mammalian fetus to life outside the womb.


Assuntos
Animais Recém-Nascidos , Embrião de Mamíferos , Desenvolvimento Embrionário , Gástrula , Análise de Célula Única , Imagem com Lapso de Tempo , Animais , Feminino , Camundongos , Gravidez , Animais Recém-Nascidos/embriologia , Animais Recém-Nascidos/genética , Diferenciação Celular/genética , Embrião de Mamíferos/citologia , Embrião de Mamíferos/embriologia , Desenvolvimento Embrionário/genética , Gástrula/citologia , Gástrula/embriologia , Gastrulação/genética , Rim/citologia , Rim/embriologia , Mesoderma/citologia , Mesoderma/enzimologia , Neurônios/citologia , Neurônios/metabolismo , Retina/citologia , Retina/embriologia , Somitos/citologia , Somitos/embriologia , Fatores de Tempo , Fatores de Transcrição/genética , Transcrição Gênica , Especificidade de Órgãos/genética
3.
Cell ; 155(5): 1104-18, 2013 Nov 21.
Artigo em Inglês | MEDLINE | ID: mdl-24238962

RESUMO

Cellular senescence disables proliferation in damaged cells, and it is relevant for cancer and aging. Here, we show that senescence occurs during mammalian embryonic development at multiple locations, including the mesonephros and the endolymphatic sac of the inner ear, which we have analyzed in detail. Mechanistically, senescence in both structures is strictly dependent on p21, but independent of DNA damage, p53, or other cell-cycle inhibitors, and it is regulated by the TGF-ß/SMAD and PI3K/FOXO pathways. Developmentally programmed senescence is followed by macrophage infiltration, clearance of senescent cells, and tissue remodeling. Loss of senescence due to the absence of p21 is partially compensated by apoptosis but still results in detectable developmental abnormalities. Importantly, the mesonephros and endolymphatic sac of human embryos also show evidence of senescence. We conclude that the role of developmentally programmed senescence is to promote tissue remodeling and propose that this is the evolutionary origin of damage-induced senescence.


Assuntos
Senescência Celular , Desenvolvimento Embrionário , Saco Endolinfático/embriologia , Mesonefro/embriologia , Animais , Inibidor de Quinase Dependente de Ciclina p21/genética , Inibidor de Quinase Dependente de Ciclina p21/metabolismo , Embrião de Mamíferos/citologia , Embrião de Mamíferos/metabolismo , Saco Endolinfático/citologia , Feminino , Humanos , Rim/embriologia , Masculino , Mesonefro/citologia , Camundongos , Fosfatidilinositol 3-Quinases/metabolismo , Transdução de Sinais , Proteínas Smad/metabolismo , Fator de Crescimento Transformador beta/metabolismo
4.
Genes Dev ; 33(19-20): 1319-1345, 2019 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-31575677

RESUMO

There are now many reports of human kidney organoids generated via the directed differentiation of human pluripotent stem cells (PSCs) based on an existing understanding of mammalian kidney organogenesis. Such kidney organoids potentially represent tractable tools for the study of normal human development and disease with improvements in scale, structure, and functional maturation potentially providing future options for renal regeneration. The utility of such organotypic models, however, will ultimately be determined by their developmental accuracy. While initially inferred from mouse models, recent transcriptional analyses of human fetal kidney have provided greater insight into nephrogenesis. In this review, we discuss how well human kidney organoids model the human fetal kidney and how the remaining differences challenge their utility.


Assuntos
Rim/fisiologia , Modelos Biológicos , Organoides/fisiologia , Regulação da Expressão Gênica no Desenvolvimento , Humanos , Rim/citologia , Rim/embriologia , Rim/crescimento & desenvolvimento , Organoides/citologia
5.
Pflugers Arch ; 476(6): 963-974, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38563997

RESUMO

Complex interactions of the branching ureteric bud (UB) and surrounding mesenchymal cells during metanephric kidney development determine the final number of nephrons. Impaired nephron endowment predisposes to arterial hypertension and chronic kidney disease. In the kidney, extracellular matrix (ECM) proteins are usually regarded as acellular scaffolds or as the common histological end-point of chronic kidney diseases. Since only little is known about their physiological role in kidney development, we aimed for analyzing the expression and role of fibronectin. In mouse, fibronectin was expressed during all stages of kidney development with significant changes over time. At embryonic day (E) 12.5 and E13.5, fibronectin lined the UB epithelium, which became less pronounced at E16.5 and then switched to a glomerular expression in the postnatal and adult kidneys. Similar results were obtained in human kidneys. Deletion of fibronectin at E13.5 in cultured metanephric mouse kidneys resulted in reduced kidney sizes and impaired glomerulogenesis following reduced cell proliferation and branching of the UB epithelium. Fibronectin colocalized with alpha 8 integrin and fibronectin loss caused a reduction in alpha 8 integrin expression, release of glial-derived neurotrophic factor and expression of Wnt11, both of which are promoters of UB branching. In conclusion, the ECM protein fibronectin acts as a regulator of kidney development and is a determinant of the final nephron number.


Assuntos
Fibronectinas , Rim , Animais , Humanos , Camundongos , Proliferação de Células , Matriz Extracelular/metabolismo , Fibronectinas/metabolismo , Fibronectinas/genética , Fator Neurotrófico Derivado de Linhagem de Célula Glial/metabolismo , Fator Neurotrófico Derivado de Linhagem de Célula Glial/genética , Cadeias alfa de Integrinas , Integrinas/metabolismo , Integrinas/genética , Rim/metabolismo , Rim/embriologia , Camundongos Endogâmicos C57BL , Proteínas Wnt/metabolismo , Proteínas Wnt/genética
6.
Kidney Int ; 105(4): 844-864, 2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-38154558

RESUMO

Congenital anomalies of the kidney and urinary tract (CAKUT) are the predominant cause for chronic kidney disease below age 30 years. Many monogenic forms have been discovered due to comprehensive genetic testing like exome sequencing. However, disease-causing variants in known disease-associated genes only explain a proportion of cases. Here, we aim to unravel underlying molecular mechanisms of syndromic CAKUT in three unrelated multiplex families with presumed autosomal recessive inheritance. Exome sequencing in the index individuals revealed three different rare homozygous variants in FOXD2, encoding a transcription factor not previously implicated in CAKUT in humans: a frameshift in the Arabic and a missense variant each in the Turkish and the Israeli family with segregation patterns consistent with autosomal recessive inheritance. CRISPR/Cas9-derived Foxd2 knockout mice presented with a bilateral dilated kidney pelvis accompanied by atrophy of the kidney papilla and mandibular, ophthalmologic, and behavioral anomalies, recapitulating the human phenotype. In a complementary approach to study pathomechanisms of FOXD2-dysfunction-mediated developmental kidney defects, we generated CRISPR/Cas9-mediated knockout of Foxd2 in ureteric bud-induced mouse metanephric mesenchyme cells. Transcriptomic analyses revealed enrichment of numerous differentially expressed genes important for kidney/urogenital development, including Pax2 and Wnt4 as well as gene expression changes indicating a shift toward a stromal cell identity. Histology of Foxd2 knockout mouse kidneys confirmed increased fibrosis. Further, genome-wide association studies suggest that FOXD2 could play a role for maintenance of podocyte integrity during adulthood. Thus, our studies help in genetic diagnostics of monogenic CAKUT and in understanding of monogenic and multifactorial kidney diseases.


Assuntos
Estruturas Embrionárias , Fatores de Transcrição Forkhead , Nefropatias , Rim , Néfrons , Sistema Urinário , Anormalidades Urogenitais , Refluxo Vesicoureteral , Adulto , Animais , Humanos , Camundongos , Estudo de Associação Genômica Ampla , Rim/anormalidades , Rim/embriologia , Nefropatias/genética , Camundongos Knockout , Néfrons/embriologia , Fatores de Transcrição/genética , Anormalidades Urogenitais/genética , Refluxo Vesicoureteral/genética , Fatores de Transcrição Forkhead/deficiência , Fatores de Transcrição Forkhead/metabolismo
7.
Biochem Biophys Res Commun ; 722: 150155, 2024 08 30.
Artigo em Inglês | MEDLINE | ID: mdl-38795454

RESUMO

Runt-related transcription factor (RUNX) family members play critical roles in the development of multiple organs. Mammalian RUNX family members, consisting of RUNX1, RUNX2, and RUNX3, have distinct tissue-specific expression and function. In this study, we examined the spatiotemporal expression patterns of RUNX family members in developing kidneys and analyzed the role of RUNX1 during kidney development. In the developing mouse kidney, RUNX1 protein was strongly expressed in the ureteric bud (UB) tip and weakly expressed in the distal segment of the renal vesicle (RV), comma-shaped body (CSB), and S-shaped body (SSB). In contrast, RUNX2 protein was restricted to the stroma, and RUNX3 protein was only expressed in immune cells. We also analyzed the expression of RUNX family members in the cynomolgus monkey kidney. We found that expression patterns of RUNX2 and RUNX3 were conserved between rodents and primates, whereas RUNX1 was only expressed in the UB tip, not in the RV, CSB, or SSB of cynomolgus monkeys, suggesting a species differences. We further evaluated the roles of RUNX1 using two different conditional knockout mice: Runx1f/f:HoxB7-Cre and Runx1f/f:R26-CreERT2 and found no abnormalities in the kidney. Our findings showed that RUNX1, which is mainly expressed in the UB tip, is not essential for kidney development.


Assuntos
Subunidade alfa 2 de Fator de Ligação ao Core , Rim , Animais , Subunidade alfa 2 de Fator de Ligação ao Core/metabolismo , Subunidade alfa 2 de Fator de Ligação ao Core/genética , Rim/metabolismo , Rim/embriologia , Rim/crescimento & desenvolvimento , Camundongos , Macaca fascicularis , Regulação da Expressão Gênica no Desenvolvimento , Subunidade alfa 1 de Fator de Ligação ao Core/metabolismo , Subunidade alfa 1 de Fator de Ligação ao Core/genética , Subunidade alfa 3 de Fator de Ligação ao Core/metabolismo , Subunidade alfa 3 de Fator de Ligação ao Core/genética , Subunidades alfa de Fatores de Ligação ao Core/metabolismo , Subunidades alfa de Fatores de Ligação ao Core/genética , Camundongos Endogâmicos C57BL , Camundongos Knockout
8.
Biochem Biophys Res Commun ; 714: 149959, 2024 06 25.
Artigo em Inglês | MEDLINE | ID: mdl-38657443

RESUMO

Gestational diabetes mellitus (GDM) presents a substantial population health concern. Previous studies have revealed that GDM can ultimately influence nephron endowment. In this study, we established a GDM mouse model to investigate the embryological alterations and molecular mechanisms underlying the development of congenital anomalies of the kidney and urinary tract (CAKUT) affected by GDM. Our study highlights that GDM could contribute to the manifestation of CAKUT, with prevalent phenotypes characterized by isolated hydronephrosis and duplex kidney complicated with hydronephrosis in mice. Ectopic ureteric buds (UBs) and extended length of common nephric ducts (CNDs) were noted in the metanephric development stage. The expression of Ret and downstream p-ERK activity were enhanced in UBs, which indicated the alteration of RET/MAPK/ERK pathway may be one of the mechanisms contributing to the increased occurrence of CAKUT associated with GDM.


Assuntos
Diabetes Gestacional , Sistema de Sinalização das MAP Quinases , Proteínas Proto-Oncogênicas c-ret , Anormalidades Urogenitais , Refluxo Vesicoureteral , Animais , Feminino , Camundongos , Gravidez , Diabetes Gestacional/metabolismo , Rim/anormalidades , Rim/metabolismo , Rim/embriologia , Proteínas Proto-Oncogênicas c-ret/metabolismo , Proteínas Proto-Oncogênicas c-ret/genética , Sistema Urinário/anormalidades , Sistema Urinário/embriologia , Anormalidades Urogenitais/etiologia , Anormalidades Urogenitais/genética , Anormalidades Urogenitais/patologia
9.
Development ; 148(19)2021 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-34553766

RESUMO

The postnatal kidney is predominantly composed of nephron epithelia with the interstitial components representing a small proportion of the final organ, except in the diseased state. This is in stark contrast to the developing organ, which arises from the mesoderm and comprises an expansive stromal population with distinct regional gene expression. In many organs, the identity and ultimate function of an epithelium is tightly regulated by the surrounding stroma during development. However, although the presence of a renal stromal stem cell population has been demonstrated, the focus has been on understanding the process of nephrogenesis whereas the role of distinct stromal components during kidney morphogenesis is less clear. In this Review, we consider what is known about the role of the stroma of the developing kidney in nephrogenesis, where these cells come from as well as their heterogeneity, and reflect on how this information may improve human kidney organoid models.


Assuntos
Células-Tronco Embrionárias/metabolismo , Rim/embriologia , Animais , Diferenciação Celular , Células-Tronco Embrionárias/citologia , Regulação da Expressão Gênica no Desenvolvimento , Humanos , Rim/citologia , Rim/metabolismo , Organogênese
10.
Pediatr Res ; 95(7): 1754-1757, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38287105

RESUMO

BACKGROUND: The prorenin receptor (PRR) plays a critical role in ureteric bud (UB) branching morphogenesis. DOT1 Like (DOT1L), a histone methyltransferase specific for Histone 3 lysine 79 (H3K79), is important for differentiation of the UB-derived renal collecting duct cells. In this study, we tested whether DOT1L/H3 dimethyl K79 (H3m2K79) are regulated by PRR deletion in the UB and UB-derived collecting ducts in the embryonic mouse kidneys. METHODS: Mutant Hoxb7Cre+/PRRflox/flox (PRRUB-/-) and control PRRUB+/+, mice were studied on embryonic (E) day E17.5. DOT1L mRNA and protein expression in the kidney was examined by real-time qRT-PCR and immunohistochemistry, respectively. H3m2K79 protein expression was determined by immunohistochemistry and Western blot analysis. RESULTS: DOT1L mRNA levels were decreased in mutant compared to control mice (0.68 ± 0.06 vs. 1.0 ± 0.01, p < 0.01). DOT1L and H3m2K79 immunostaining was reduced in the mutant vs. control kidneys (Dot1: 0.62 ± 0.03 vs. 1.0 ± 0.01, p < 0.05; H3m2K79: 0.64 ± 0.04 vs.1.1 ± 0.01. p < 0.05.). Western blot analysis revealed decreased H3m2K79 protein levels in mutant compared to control kidneys (1.0 ± 0.06 vs. 1.5 ± 0.02, p < 0.05). CONCLUSION: Targeted deletion of the PRR in the UB and UB-derived collecting ducts results in reduced DOT1L gene/protein and H3m2K79 protein expression in the embryonic mouse metanephroi in vivo. IMPACT: The role of histone methylation in mediating the effect of the prorenin receptor on the ureteric bud branching (UB) morphogenesis and urine acidification during kidney development is unknown. We demonstrate that histone H3 lysine (K) 79 dimethylation by methyltransferase Dot1 is reduced in the embryonic kidney of mice that lack the prorenin receptor in the UB lineage.


Assuntos
Histona-Lisina N-Metiltransferase , Histonas , Receptor de Pró-Renina , Receptores de Superfície Celular , Ureter , Animais , Histona-Lisina N-Metiltransferase/metabolismo , Histona-Lisina N-Metiltransferase/genética , Camundongos , Histonas/metabolismo , Receptores de Superfície Celular/metabolismo , Receptores de Superfície Celular/genética , Ureter/embriologia , Ureter/metabolismo , Transdução de Sinais , Camundongos Knockout , Deleção de Genes , Metilação , Rim/metabolismo , Rim/embriologia , RNA Mensageiro/metabolismo , RNA Mensageiro/genética , Regulação da Expressão Gênica no Desenvolvimento , Túbulos Renais Coletores/metabolismo , Túbulos Renais Coletores/embriologia , Proteínas de Homeodomínio/metabolismo , Proteínas de Homeodomínio/genética , Estruturas Embrionárias
11.
Ann Vasc Surg ; 108: 65-75, 2024 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-38942378

RESUMO

BACKGROUND: During human morphogenesis, the definitive kidneys derive from the metanephros during Carnegie Stage 14 to 23. The pronephros and the mesonephros develop previously and successively to finally lead to the formation of the urinary tract. Renal vascularization, first described in 1912 by Félix using a "ladder theory" model, is highly variable and current available morphogenesis descriptions do not explain all reported anatomical variations. The aim of this work was to study the morphogenesis of the human metanephros and its vascularization by three-dimensional reconstructions of human embryos. METHODS: Histological sections of 23 human embryos from the Carnegie Collection and 5 human embryos from the French collection (Carnegie stages 14 to 23) were completely digitalized and reconstructed in three dimensions using specific softwares and then analyzed by descriptive method using manual annotation. RESULTS: In all studied embryos, the mesonephric arteries did not reach the metanephros irrespective to the position of the metanephros during its cranial ascent. Before the end of the cranial metanephros migration (15 embryos), at the level of the aorto-iliac bifurcation, a "primitive" vascularization was shown in 9 of them. The renal artery originated from the primitive iliac arteries for 8 embryos and from the inferior mesenteric artery in one embryo. Further, a capillary cluster emerging from the lateral wall of the aorta and extending toward the metanephros was found in 2 embryos (Carnegie stages 21 and 22). This may correspond to a phenomenon of neoangiogenesis responsible of the definitive renal artery. CONCLUSIONS: The present study reported the morphogenesis of human renal arteries between Carnegie stages 14 and 23 using an original method of tridimensional computerized reconstructions of historical human embryos. Some original findings, in contradiction with the original Felix's description, may explain the most frequently reported anatomical variations.


Assuntos
Imageamento Tridimensional , Mesonefro , Morfogênese , Artéria Renal , Humanos , Artéria Renal/embriologia , Mesonefro/embriologia , Idade Gestacional , Rim/irrigação sanguínea , Rim/embriologia , Modelos Anatômicos
12.
Arch Gynecol Obstet ; 310(3): 1451-1459, 2024 09.
Artigo em Inglês | MEDLINE | ID: mdl-38365961

RESUMO

BACKGROUND: The precise determination of gestational age is essential for effectively managing and prognosis of all pregnancies. Through careful biometry, timely interventions can be implemented, leading to positive outcomes for both the mother and fetus. In routine fetal biometry, parameters such as biparietal diameter (BPD), femur length (FL), head circumference (HC), and abdominal circumference (AC) have been traditionally used. This study aims to evaluate the usefulness of fetal kidney length (FKL) as a marker for fetal biometry. METHODOLOGY: This prospective, observational, and cross-sectional study was conducted in the Radiodiagnosis and Obstetrics and Gynaecology departments, including a diverse group of pregnant women from various socio-economic backgrounds, with adherence to ethical standards. Women with singleton pregnancies between 22 and 40 weeks of gestation who met the inclusion and exclusion criteria were examined through ultrasound. The data collected were subsequently analyzed. RESULT: In the current study, 280 participants with an average age of 26.71 ± 3.6 years were included. The agreement between the mean fetal kidney length and standard biometry parameters was almost perfect, with a strength of agreement exceeding 0.99. A strong and statistically significant positive correlation existed between fetal kidney length and the estimated gestational period calculated using DLMP/standard biometric measurements. CONCLUSION: Fetal kidney length is a reliable indicator of gestational age and can supplement standard biometric measurements to provide a more precise estimation of gestational age, especially in the later stages when obtaining such standard measurements may be challenging.


Assuntos
Biometria , Idade Gestacional , Rim , Ultrassonografia Pré-Natal , Humanos , Feminino , Gravidez , Estudos Transversais , Estudos Prospectivos , Adulto , Rim/diagnóstico por imagem , Rim/embriologia , Rim/anatomia & histologia , Adulto Jovem , Feto/diagnóstico por imagem , Feto/anatomia & histologia , Desenvolvimento Fetal , Tamanho do Órgão
13.
J Environ Sci (China) ; 145: 75-87, 2024 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-38844325

RESUMO

Prednisone is a synthetic glucocorticoid that is commonly used in both human and veterinary medication. Now, it is also recognized as an emerging environmental contaminant. Pregnant women may be exposed to prednisone actively or passively through multiple pathways and cause developmental toxicity to the fetus. However, the impact of prenatal prednisone exposure (PPE) on fetal kidney development remains unclear. In this study, pregnant mice were administered prednisone intragastrically during full-term pregnancy with different doses (0.25, 0.5, or 1 mg/(kg·day)), or at the dose of 1 mg/(kg·day) in different gestational days (GD) (GD0-9, GD10-18, or GD0-18). The pregnant mice were euthanized on GD18. HE staining revealed fetal kidney dysplasia, with an enlarged glomerular Bowman's capsule space and a reduced capillary network in the PPE groups. The expression of the podocyte and the mesangial cell marker genes was significantly reduced in the PPE groups. However, overall gene expression in renal tubules and collecting ducts were markedly increased. All of the above effects were more pronounced in high-dose, full-term pregnancy, and female fetuses. Studies on the mechanism of the female fetal kidney have revealed that PPE reduced the expression of Six2, increased the expression of Hnf1ß, Hnf4α, and Wnt9b, and inhibited the expression of glial cell line-derived neurotrophic factor (GDNF) and Notch signaling pathways. In conclusion, this study demonstrated that there is a sex difference in the developmental toxicity of PPE to the fetal kidney, and the time effect is manifested as full-term pregnancy > early pregnancy > mid-late pregnancy.


Assuntos
Rim , Prednisona , Feminino , Animais , Gravidez , Camundongos , Rim/efeitos dos fármacos , Rim/embriologia , Prednisona/toxicidade , Desenvolvimento Fetal/efeitos dos fármacos , Masculino , Efeitos Tardios da Exposição Pré-Natal/induzido quimicamente , Exposição Materna/efeitos adversos
14.
Kidney Int ; 103(1): 77-86, 2023 01.
Artigo em Inglês | MEDLINE | ID: mdl-36055600

RESUMO

The kidney is an essential organ that ensures bodily fluid homeostasis and removes soluble waste products from the organism. Nephrons, the functional units of the kidney, comprise a blood filter, the glomerulus or glomus, and an epithelial tubule that processes the filtrate from the blood or coelom and selectively reabsorbs solutes, such as sugars, proteins, ions, and water, leaving waste products to be eliminated in the urine. Genes coding for transporters are segmentally expressed, enabling the nephron to sequentially process the filtrate. The Xenopus embryonic kidney, the pronephros, which consists of a single large nephron, has served as a valuable model to identify genes involved in nephron formation and patterning. Therefore, the developmental patterning program that generates these segments is of great interest. Prior work has defined the gene expression profiles of Xenopus nephron segments via in situ hybridization strategies, but a comprehensive understanding of the cellular makeup of the pronephric kidney remains incomplete. Here, we carried out single-cell mRNA sequencing of the functional Xenopus pronephric nephron and evaluated its cellular composition through comparative analyses with previous Xenopus studies and single-cell mRNA sequencing of the adult mouse kidney. This study reconstructs the cellular makeup of the pronephric kidney and identifies conserved cells, segments, and associated gene expression profiles. Thus, our data highlight significant conservation in podocytes, proximal and distal tubule cells, and divergence in cellular composition underlying the capacity of each nephron to remove wastes in the form of urine, while emphasizing the Xenopus pronephros as a model for physiology and disease.


Assuntos
Rim , Néfrons , Animais , Camundongos , Regulação da Expressão Gênica no Desenvolvimento , Rim/embriologia , Glomérulos Renais/embriologia , Néfrons/embriologia , RNA Mensageiro/genética , Xenopus laevis/embriologia
15.
Development ; 147(21)2020 06 22.
Artigo em Inglês | MEDLINE | ID: mdl-32439764

RESUMO

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.


Assuntos
Deficiências do Desenvolvimento/genética , Desenvolvimento Fetal , Laminina/genética , Mutação/genética , Polimerização , Sequência de Aminoácidos , Animais , Animais Recém-Nascidos , Pré-Escolar , Deficiências do Desenvolvimento/patologia , Feto/embriologia , Humanos , Hidronefrose/patologia , Recém-Nascido , Rim/anormalidades , Rim/embriologia , Rim/patologia , Laminina/química , Pulmão/anormalidades , Pulmão/embriologia , Pulmão/patologia , Masculino , Camundongos , Domínios Proteicos , Síndrome
16.
Development ; 147(10)2020 05 22.
Artigo em Inglês | MEDLINE | ID: mdl-32444428

RESUMO

Over the past 5 years, several studies have begun to uncover the links between the classical signal transduction pathways and the physical mechanisms that are used to sculpt branched tissues. These advances have been made, in part, thanks to innovations in live imaging and reporter animals. With modern research tools, our conceptual models of branching morphogenesis are rapidly evolving, and the differences in branching mechanisms between each organ are becoming increasingly apparent. Here, we highlight four branched epithelia that develop at different spatial scales, within different surrounding tissues and via divergent physical mechanisms. Each of these organs has evolved to employ unique branching strategies to achieve a specialized final architecture.


Assuntos
Epitélio/metabolismo , Morfogênese/fisiologia , Transdução de Sinais/fisiologia , Animais , Feminino , Humanos , Rim/embriologia , Rim/crescimento & desenvolvimento , Rim/metabolismo , Pulmão/embriologia , Pulmão/crescimento & desenvolvimento , Pulmão/metabolismo , Glândulas Mamárias Animais/embriologia , Glândulas Mamárias Animais/crescimento & desenvolvimento , Glândulas Mamárias Animais/metabolismo , Glândulas Mamárias Humanas/embriologia , Glândulas Mamárias Humanas/crescimento & desenvolvimento , Glândulas Mamárias Humanas/metabolismo , Glândulas Salivares/embriologia , Glândulas Salivares/crescimento & desenvolvimento , Glândulas Salivares/metabolismo
17.
Development ; 147(4)2020 02 20.
Artigo em Inglês | MEDLINE | ID: mdl-32001436

RESUMO

Proper organ development depends on coordinated communication between multiple cell types. Retinoic acid (RA) is an autocrine and paracrine signaling molecule essential for the development of most organs, including the lung. Despite extensive work detailing effects of RA deficiency in early lung morphogenesis, little is known about how RA regulates late gestational lung maturation. Here, we investigate the role of the RA catabolizing protein Cyp26b1 in the lung. Cyp26b1 is highly enriched in lung endothelial cells (ECs) throughout development. We find that loss of Cyp26b1 leads to reduction of alveolar type 1 cells, failure of alveolar inflation and early postnatal lethality in mouse. Furthermore, we observe expansion of distal epithelial progenitors, but no appreciable changes in proximal airways, ECs or stromal populations. Exogenous administration of RA during late gestation partially mimics these defects; however, transcriptional analyses comparing Cyp26b1-/- with RA-treated lungs reveal overlapping, but distinct, responses. These data suggest that defects observed in Cyp26b1-/- lungs are caused by both RA-dependent and RA-independent mechanisms. This work reports crucial cellular crosstalk during lung development involving Cyp26b1-expressing endothelium and identifies a novel RA modulator in lung development.


Assuntos
Epitélio/embriologia , Pulmão/embriologia , Alvéolos Pulmonares/embriologia , Ácido Retinoico 4 Hidroxilase/genética , Ácido Retinoico 4 Hidroxilase/fisiologia , Animais , Sistemas CRISPR-Cas , Diferenciação Celular , Células Endoteliais/citologia , Células Epiteliais/citologia , Feminino , Regulação da Expressão Gênica no Desenvolvimento , Rim/embriologia , Camundongos , Camundongos Endogâmicos C57BL , Organogênese/efeitos dos fármacos , Gravidez , Prenhez , Transdução de Sinais , Células-Tronco/citologia , Tretinoína/farmacologia
18.
Circ Res ; 128(7): 887-907, 2021 04 02.
Artigo em Inglês | MEDLINE | ID: mdl-33793334

RESUMO

Renin cells are essential for survival perfected throughout evolution to ensure normal development and defend the organism against a variety of homeostatic threats. During embryonic and early postnatal life, they are progenitors that participate in the morphogenesis of the renal arterial tree. In adult life, they are capable of regenerating injured glomeruli, control blood pressure, fluid-electrolyte balance, tissue perfusion, and in turn, the delivery of oxygen and nutrients to cells. Throughout life, renin cell descendants retain the plasticity or memory to regain the renin phenotype when homeostasis is threatened. To perform all of these functions and maintain well-being, renin cells must regulate their identity and fate. Here, we review the major mechanisms that control the differentiation and fate of renin cells, the chromatin events that control the memory of the renin phenotype, and the major pathways that determine their plasticity. We also examine how chronic stimulation of renin cells alters their fate leading to the development of a severe and concentric hypertrophy of the intrarenal arteries and arterioles. Lastly, we provide examples of additional changes in renin cell fate that contribute to equally severe kidney disorders.


Assuntos
Hipertensão/etiologia , Rim/citologia , Renina/fisiologia , Animais , Arteríolas/embriologia , Pressão Sanguínea/fisiologia , Comunicação Celular , Diferenciação Celular , Plasticidade Celular , Cromatina/fisiologia , Montagem e Desmontagem da Cromatina/fisiologia , Conexinas/fisiologia , Homeostase , Humanos , Integrinas/fisiologia , Sistema Justaglomerular/citologia , Rim/irrigação sanguínea , Rim/embriologia , Glomérulos Renais/fisiologia , Camundongos , MicroRNAs/fisiologia , Fenótipo , Regeneração/fisiologia , Artéria Renal , Renina/metabolismo , Sistema Renina-Angiotensina/fisiologia , Células-Tronco/fisiologia , Equilíbrio Hidroeletrolítico
19.
Genes Dev ; 29(5): 467-82, 2015 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-25737276

RESUMO

Wilms' tumor, or nephroblastoma, is the most common pediatric renal cancer. The tumors morphologically resemble embryonic kidneys with a disrupted architecture and are associated with undifferentiated metanephric precursors. Here, we discuss genetic and epigenetic findings in Wilms' tumor in the context of renal development. Many of the genes implicated in Wilms' tumorigenesis are involved in the control of nephron progenitors or the microRNA (miRNA) processing pathway. Whereas the first group of genes has been extensively studied in normal development, the second finding suggests important roles for miRNAs in general-and specific miRNAs in particular-in normal kidney development that still await further analysis. The recent identification of Wilms' tumor cancer stem cells could provide a framework to integrate these pathways and translate them into new or improved therapeutic interventions.


Assuntos
Regulação da Expressão Gênica no Desenvolvimento , Neoplasias Renais/genética , Rim/embriologia , Organogênese/genética , Tumor de Wilms/genética , Animais , Epigênese Genética/genética , Regulação Neoplásica da Expressão Gênica , Humanos , Rim/patologia , MicroRNAs/genética
20.
Dev Biol ; 477: 98-116, 2021 09.
Artigo em Inglês | MEDLINE | ID: mdl-34000274

RESUMO

Chronic kidney disease (CKD) and end stage renal disease (ESRD) are increasingly frequent and devastating conditions that have driven a surge in the need for kidney transplantation. A stark shortage of organs has fueled interest in generating viable replacement tissues ex vivo for transplantation. One promising approach has been self-organizing organoids, which mimic developmental processes and yield multicellular, organ-specific tissues. However, a recognized roadblock to this approach is that many organoid cell types fail to acquire full maturity and function. Here, we comprehensively assess the vasculature in two distinct kidney organoid models as well as in explanted embryonic kidneys. Using a variety of methods, we show that while organoids can develop a wide range of kidney cell types, as previously shown, endothelial cells (ECs) initially arise but then rapidly regress over time in culture. Vasculature of cultured embryonic kidneys exhibit similar regression. By contrast, engraftment of kidney organoids under the kidney capsule results in the formation of a stable, perfused vasculature that integrates into the organoid. This work demonstrates that kidney organoids offer a promising model system to define the complexities of vascular-nephron interactions, but the establishment and maintenance of a vascular network present unique challenges when grown ex vivo.


Assuntos
Endotélio Vascular/embriologia , Rim/irrigação sanguínea , Rim/embriologia , Organogênese , Organoides/embriologia , Animais , Células Cultivadas , Células Endoteliais , Endotélio Vascular/citologia , Feminino , Humanos , Rim/citologia , Masculino , Camundongos , Organoides/transplante , RNA-Seq , Técnicas de Cultura de Tecidos
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa