Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 47
Filtrar
1.
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
2.
Hum Mol Genet ; 29(23): 3781-3792, 2021 02 04.
Artigo em Inglês | MEDLINE | ID: mdl-33305798

RESUMO

Heterozygous mutations in the human SOX9 gene cause the skeletal malformation syndrome campomelic dysplasia which in 75% of 46, XY individuals is associated with male-to-female sex reversal. Although studies in homozygous Sox9 knockout mouse models confirmed that SOX9 is critical for testis development, mice heterozygous for the Sox9-null allele were reported to develop normal testes. This led to the belief that the SOX9 dosage requirement for testis differentiation is different between humans, which often require both alleles, and mice, in which one allele is sufficient. However, in prior studies, gonadal phenotypes in heterozygous Sox9 XY mice were assessed only by either gross morphology, histological staining or analyzed on a mixed genetic background. In this study, we conditionally inactivated Sox9 in somatic cells of developing gonads using the Nr5a1-Cre mouse line on a pure C57BL/6 genetic background. Section and whole-mount immunofluorescence for testicular and ovarian markers showed that XY Sox9 heterozygous gonads developed as ovotestes. Quantitative droplet digital PCR confirmed a 50% reduction of Sox9 mRNA as well as partial sex reversal shown by an upregulation of ovarian genes. Our data show that haploinsufficiency of Sox9 can perturb testis development in mice, suggesting that mice may provide a more accurate model of human disorders/differences of sex development than previously thought.


Assuntos
Displasia Campomélica/patologia , Transtornos do Desenvolvimento Sexual/patologia , Gônadas/patologia , Heterozigoto , Fatores de Transcrição SOX9/fisiologia , Diferenciação Sexual , Fator Esteroidogênico 1/fisiologia , Animais , Displasia Campomélica/etiologia , Displasia Campomélica/metabolismo , Modelos Animais de Doenças , Transtornos do Desenvolvimento Sexual/etiologia , Transtornos do Desenvolvimento Sexual/metabolismo , Feminino , Regulação da Expressão Gênica no Desenvolvimento , Gônadas/metabolismo , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fenótipo
3.
Am J Pathol ; 192(5): 738-749, 2022 05.
Artigo em Inglês | MEDLINE | ID: mdl-35181335

RESUMO

Kidney organoids derived from pluripotent stem cells and epithelial organoids derived from adult tissue (tubuloids) have been used to study various kidney disorders with a strong genetic component, such as polycystic kidney disease, Wilms tumor, and congenital nephrotic syndrome. However, complex disorders without clear genetic associations, such as acute kidney injury and many forms of chronic kidney disease, are only just beginning to be investigated using these in vitro approaches. Although organoids are a reductionist model, they contain clinically relevant cell populations that may help to elucidate human-specific pathogenic mechanisms. Thus, organoids may complement animal disease models to accelerate the translation of laboratory proof-of-concept research into clinical practice. This review discusses whether kidney organoids and tubuloids are suitable models for the study of complex human kidney disease and highlights their advantages and limitations compared with monolayer cell culture and animal models.


Assuntos
Injúria Renal Aguda , Células-Tronco Pluripotentes , Insuficiência Renal Crônica , Animais , Diferenciação Celular , Feminino , Humanos , Rim , Masculino , Organoides
4.
Cell Mol Life Sci ; 79(6): 296, 2022 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-35570209

RESUMO

Polymyxin antibiotics are often used as a last-line defense to treat life-threatening Gram-negative pathogens. However, polymyxin-induced kidney toxicity is a dose-limiting factor of paramount importance and can lead to suboptimal treatment. To elucidate the mechanism and develop effective strategies to overcome polymyxin toxicity, we employed a whole-genome CRISPR screen in human kidney tubular HK-2 cells and identified 86 significant genes that upon knock-out rescued polymyxin-induced toxicity. Specifically, we discovered that knockout of the inwardly rectifying potassium channels Kir4.2 and Kir5.1 (encoded by KCNJ15 and KCNJ16, respectively) rescued polymyxin-induced toxicity in HK-2 cells. Furthermore, we found that polymyxins induced cell depolarization via Kir4.2 and Kir5.1 and a significant cellular uptake of polymyxins was evident. All-atom molecular dynamics simulations revealed that polymyxin B1 spontaneously bound to Kir4.2, thereby increasing opening of the channel, resulting in a potassium influx, and changes of the membrane potential. Consistent with these findings, small molecule inhibitors (BaCl2 and VU0134992) of Kir potassium channels reduced polymyxin-induced toxicity in cell culture and mouse explant kidney tissue. Our findings provide critical mechanistic information that will help attenuate polymyxin-induced nephrotoxicity in patients and facilitate the design of novel, safer polymyxins.


Assuntos
Canais de Potássio Corretores do Fluxo de Internalização , Animais , Humanos , Rim/metabolismo , Potenciais da Membrana , Camundongos , Polimixinas/metabolismo , Polimixinas/toxicidade , Potássio/metabolismo , Canais de Potássio Corretores do Fluxo de Internalização/genética , Canais de Potássio Corretores do Fluxo de Internalização/metabolismo
5.
Development ; 146(12)2019 06 12.
Artigo em Inglês | MEDLINE | ID: mdl-31118232

RESUMO

Recent advances in the generation of kidney organoids and the culture of primary nephron progenitors from mouse and human have been based on knowledge of the molecular basis of kidney development in mice. Although gene expression during kidney development has been intensely investigated, single cell profiling provides new opportunities to further subsect component cell types and the signalling networks at play. Here, we describe the generation and analysis of 6732 single cell transcriptomes from the fetal mouse kidney [embryonic day (E)18.5] and 7853 sorted nephron progenitor cells (E14.5). These datasets provide improved resolution of cell types and specific markers, including subdivision of the renal stroma and heterogeneity within the nephron progenitor population. Ligand-receptor interaction and pathway analysis reveals novel crosstalk between cellular compartments and associates new pathways with differentiation of nephron and ureteric epithelium cell types. We identify transcriptional congruence between the distal nephron and ureteric epithelium, showing that most markers previously used to identify ureteric epithelium are not specific. Together, this work improves our understanding of metanephric kidney development and provides a template to guide the regeneration of renal tissue.


Assuntos
Perfilação da Expressão Gênica , Regulação da Expressão Gênica no Desenvolvimento , Rim/embriologia , Receptor Cross-Talk , Análise de Célula Única/métodos , Algoritmos , Animais , Diferenciação Celular , Linhagem da Célula , Epitélio/embriologia , Rim/citologia , Ligantes , Camundongos , Camundongos Endogâmicos C57BL , Néfrons/embriologia , Organogênese , Transdução de Sinais , Células-Tronco/citologia , Transcriptoma , Ureter/embriologia
6.
Development ; 146(5)2019 03 07.
Artigo em Inglês | MEDLINE | ID: mdl-30846463

RESUMO

Kidney organoids have potential uses in disease modelling, drug screening and regenerative medicine. However, novel cost-effective techniques are needed to enable scaled-up production of kidney cell types in vitro We describe here a modified suspension culture method for the generation of kidney micro-organoids from human pluripotent stem cells. Optimisation of differentiation conditions allowed the formation of micro-organoids, each containing six to ten nephrons that were surrounded by endothelial and stromal populations. Single cell transcriptional profiling confirmed the presence and transcriptional equivalence of all anticipated renal cell types consistent with a previous organoid culture method. This suspension culture micro-organoid methodology resulted in a three- to fourfold increase in final cell yield compared with static culture, thereby representing an economical approach to the production of kidney cells for various biological applications.


Assuntos
Técnicas de Cultura de Células , Regulação da Expressão Gênica no Desenvolvimento , Rim/citologia , Células-Tronco Pluripotentes/citologia , Albuminas/metabolismo , Diferenciação Celular , Células Cultivadas , Doxorrubicina/farmacologia , Humanos , Néfrons/metabolismo , Organoides , Transdução de Sinais , Transcrição Gênica , Proteínas Wnt/metabolismo
7.
Nat Methods ; 16(1): 79-87, 2019 01.
Artigo em Inglês | MEDLINE | ID: mdl-30573816

RESUMO

The utility of human pluripotent stem cell-derived kidney organoids relies implicitly on the robustness and transferability of the protocol. Here we analyze the sources of transcriptional variation in a specific kidney organoid protocol. Although individual organoids within a differentiation batch showed strong transcriptional correlation, we noted significant variation between experimental batches, particularly in genes associated with temporal maturation. Single-cell profiling revealed shifts in nephron patterning and proportions of component cells. Distinct induced pluripotent stem cell clones showed congruent transcriptional programs, with interexperimental and interclonal variation also strongly associated with nephron patterning. Epithelial cells isolated from organoids aligned with total organoids at the same day of differentiation, again implicating relative maturation as a confounder. This understanding of experimental variation facilitated an optimized analysis of organoid-based disease modeling, thereby increasing the utility of kidney organoids for personalized medicine and functional genomics.


Assuntos
Rim/metabolismo , Organoides/metabolismo , Diferenciação Celular/genética , Células Clonais , Células Epiteliais/citologia , Perfilação da Expressão Gênica , Humanos , Células-Tronco Pluripotentes Induzidas/citologia , Células-Tronco Pluripotentes Induzidas/metabolismo , Rim/citologia , Nefropatias/genética , Nefropatias/patologia , Modelos Biológicos , Organoides/citologia , Reprodutibilidade dos Testes , Análise de Célula Única , Transcrição Gênica
8.
Kidney Int ; 100(4): 780-786, 2021 10.
Artigo em Inglês | MEDLINE | ID: mdl-34089762

RESUMO

For decades, measurements of kidney microanatomy using 2-dimensional sections has provided us with a detailed knowledge of kidney morphology under physiological and pathological conditions. However, the rapid development of tissue clearing methods in recent years, in combination with the development of novel 3-dimensional imaging modalities have provided new insights into kidney structure and function. This review article describes a range of novel insights into kidney development and disease obtained recently using these new methodological approaches. For example, in the developing kidney these approaches have provided new understandings of ureteric branching morphogenesis, nephron progenitor cell proliferation and commitment, interactions between ureteric tip cells and nephron progenitor cells, and the establishment of nephron segmentation. In whole adult mouse kidneys, tissue clearing combined with light sheet microscopy can image and quantify the total number of glomeruli, a major breakthrough in the field. Similar approaches have provided new insights into the structure of the renal vasculature and innervation, tubulointerstitial remodeling, podocyte loss and hypertrophy, cyst formation, the evolution of cellular crescents, and the structure of the glomerular filtration barrier. Many more advances in the understanding of kidney biology and pathology can be expected as additional clearing and imaging techniques are developed and adopted by more investigators.


Assuntos
Podócitos , Ureter , Animais , Rim/diagnóstico por imagem , Glomérulos Renais , Camundongos , Néfrons , Organogênese
9.
Proc Natl Acad Sci U S A ; 115(23): 5998-6003, 2018 06 05.
Artigo em Inglês | MEDLINE | ID: mdl-29784808

RESUMO

Nephrogenesis concludes by the 36th week of gestation in humans and by the third day of postnatal life in mice. Extending the nephrogenic period may reduce the onset of adult renal and cardiovascular disease associated with low nephron numbers. We conditionally deleted either Mtor or Tsc1 (coding for hamartin, an inhibitor of Mtor) in renal progenitor cells. Loss of one Mtor allele caused a reduction in nephron numbers; complete deletion led to severe paucity of glomeruli in the kidney resulting in early death after birth. By contrast, loss of one Tsc1 allele from renal progenitors resulted in a 25% increase in nephron endowment with no adverse effects. Increased progenitor engraftment rates ex vivo relative to controls correlated with prolonged nephrogenesis through the fourth postnatal day. Complete loss of both Tsc1 alleles in renal progenitors led to a lethal tubular lesion. The hamartin phenotypes are not dependent on the inhibitory effect of TSC on the Mtor complex but are dependent on Raptor.


Assuntos
Néfrons , Organogênese/fisiologia , Serina-Treonina Quinases TOR/metabolismo , Proteínas Supressoras de Tumor/metabolismo , Animais , Feminino , Masculino , Camundongos , Camundongos Transgênicos , Néfrons/química , Néfrons/citologia , Néfrons/crescimento & desenvolvimento , Néfrons/fisiologia , Serina-Treonina Quinases TOR/genética , Proteína 1 do Complexo Esclerose Tuberosa
10.
Development ; 144(6): 1087-1096, 2017 03 15.
Artigo em Inglês | MEDLINE | ID: mdl-28174247

RESUMO

Human pluripotent stem cells, after directed differentiation in vitro, can spontaneously generate complex tissues via self-organisation of the component cells. Self-organisation can also reform embryonic organ structure after tissue disruption. It has previously been demonstrated that dissociated embryonic kidneys can recreate component epithelial and mesenchymal relationships sufficient to allow continued kidney morphogenesis. Here, we investigate the timing and underlying mechanisms driving self-organisation after dissociation of the embryonic kidney using time-lapse imaging, high-resolution confocal analyses and mathematical modelling. Organotypic self-organisation sufficient for nephron initiation was observed within a 24 h period. This involved cell movement, with structure emerging after the clustering of ureteric epithelial cells, a process consistent with models of random cell movement with preferential cell adhesion. Ureteric epithelialisation rapidly followed the formation of ureteric cell clusters with the reformation of nephron-forming niches representing a later event. Disruption of P-cadherin interactions was seen to impair this ureteric epithelial cell clustering without affecting epithelial maturation. This understanding could facilitate improved regulation of patterning within organoids and facilitate kidney engineering approaches guided by cell-cell self-organisation.


Assuntos
Células Epiteliais/citologia , Rim/embriologia , Ureter/citologia , Animais , Caderinas/metabolismo , Adesão Celular , Agregação Celular , Diferenciação Celular , Linhagem da Célula , Movimento Celular , Simulação por Computador , Células Epiteliais/metabolismo , Camundongos , Modelos Biológicos , Morfogênese , Fatores de Tempo
11.
Kidney Int ; 93(3): 589-598, 2018 03.
Artigo em Inglês | MEDLINE | ID: mdl-29217079

RESUMO

The regulation of final nephron number in the kidney is poorly understood. Cessation of nephron formation occurs when the self-renewing nephron progenitor population commits to differentiation. Transcription factors within this progenitor population, such as SIX2, are assumed to control expression of genes promoting self-renewal such that homozygous Six2 deletion results in premature commitment and an early halt to kidney development. In contrast, Six2 heterozygotes were assumed to be unaffected. Using quantitative morphometry, we found a paradoxical 18% increase in ureteric branching and final nephron number in Six2 heterozygotes, despite evidence for reduced levels of SIX2 protein and transcript. This was accompanied by a clear shift in nephron progenitor identity with a distinct subset of downregulated progenitor genes such as Cited1 and Meox1 while other genes were unaffected. The net result was an increase in nephron progenitor proliferation, as assessed by elevated EdU (5-ethynyl-2'-deoxyuridine) labeling, an increase in MYC protein, and transcriptional upregulation of MYC target genes. Heterozygosity for Six2 on an Fgf20-/- background resulted in premature differentiation of the progenitor population, confirming that progenitor regulation is compromised in Six2 heterozygotes. Overall, our studies reveal a unique dose response of nephron progenitors to the level of SIX2 protein in which the role of SIX2 in progenitor proliferation versus self-renewal is separable.


Assuntos
Proliferação de Células/genética , Autorrenovação Celular/genética , Haploinsuficiência , Proteínas de Homeodomínio/genética , Morfogênese/genética , Néfrons/metabolismo , Células-Tronco/metabolismo , Fatores de Transcrição/genética , Animais , Proteínas Reguladoras de Apoptose , Fatores de Crescimento de Fibroblastos/deficiência , Fatores de Crescimento de Fibroblastos/genética , Regulação da Expressão Gênica no Desenvolvimento , Genótipo , Heterozigoto , Proteínas de Homeodomínio/metabolismo , Camundongos Knockout , Néfrons/embriologia , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Fenótipo , Proteínas Proto-Oncogênicas c-myc/genética , Proteínas Proto-Oncogênicas c-myc/metabolismo , Transdução de Sinais/genética , Transativadores/genética , Transativadores/metabolismo , Fatores de Transcrição/deficiência
12.
J Math Biol ; 76(7): 1673-1697, 2018 06.
Artigo em Inglês | MEDLINE | ID: mdl-29392399

RESUMO

The adult mammalian kidney has a complex, highly-branched collecting duct epithelium that arises as a ureteric bud sidebranch from an epithelial tube known as the nephric duct. Subsequent branching of the ureteric bud to form the collecting duct tree is regulated by subcellular interactions between the epithelium and a population of mesenchymal cells that surround the tips of outgrowing branches. The mesenchymal cells produce glial cell-line derived neurotrophic factor (GDNF), that binds with RET receptors on the surface of the epithelial cells to stimulate several subcellular pathways in the epithelium. Such interactions are known to be a prerequisite for normal branching development, although competing theories exist for their role in morphogenesis. Here we introduce the first agent-based model of ex vivo kidney uretic branching. Through comparison with experimental data, we show that growth factor-regulated growth mechanisms can explain early epithelial cell branching, but only if epithelial cell division depends in a switch-like way on the local growth factor concentration; cell division occurring only if the driving growth factor level exceeds a threshold. We also show how a recently-developed method, "Approximate Approximate Bayesian Computation", can be used to infer key model parameters, and reveal the dependency between the parameters controlling a growth factor-dependent growth switch. These results are consistent with a requirement for signals controlling proliferation and chemotaxis, both of which are previously identified roles for GDNF.


Assuntos
Rim/crescimento & desenvolvimento , Modelos Biológicos , Algoritmos , Animais , Teorema de Bayes , Proliferação de Células , Biologia Computacional , Simulação por Computador , Fator Neurotrófico Derivado de Linhagem de Célula Glial/metabolismo , Humanos , Rim/metabolismo , Conceitos Matemáticos , Camundongos , Camundongos Transgênicos , Morfogênese , Técnicas de Cultura de Órgãos , Transdução de Sinais , Análise de Sistemas
13.
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
15.
Dev Biol ; 404(2): 88-102, 2015 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-26116176

RESUMO

ROBO2 plays a key role in regulating ureteric bud (UB) formation in the embryo, with mutations in humans and mice leading to supernumerary kidneys. Previous studies have established that the number and position of UB outgrowths is determined by the domain of metanephric mesenchymal Gdnf expression, which is expanded anteriorly in Robo2 mouse mutants. To clarify how this phenotype arises, we used high-resolution 3D imaging to reveal an increase in the number of nephrogenic cord cells, leading to extension of the metanephric mesenchyme field in Robo2-null mouse embryos. Ex vivo experiments suggested a dependence of this effect on proliferative signals from the Wolffian duct. Loss of Robo2 resulted in a failure of the normal separation of the mesenchyme from the Wolffian duct/ureteric epithelium, suggesting that aberrant juxtaposition of these two compartments in Robo2-null mice exposes the mesenchyme to abnormally high levels of proliferative stimuli. Our data suggest a new model in which SLIT-ROBO signalling acts not by attenuating Gdnf expression or activity, but instead by limiting epithelial/mesenchymal interactions in the nascent metanephros and restricting the extent of the nephrogenic field. These insights illuminate the aetiology of multiplex kidney formation in human individuals with ROBO2 mutations.


Assuntos
Fator Neurotrófico Derivado de Linhagem de Célula Glial/genética , Proteínas de Homeodomínio/genética , Rim/embriologia , Receptores Imunológicos/genética , Fatores de Transcrição/genética , Ductos Mesonéfricos/embriologia , Animais , Linhagem Celular , Proliferação de Células , Células Epiteliais/citologia , Fator Neurotrófico Derivado de Linhagem de Célula Glial/biossíntese , Proteínas de Homeodomínio/biossíntese , Humanos , Peptídeos e Proteínas de Sinalização Intercelular/genética , Mesoderma/citologia , Mesoderma/embriologia , Camundongos , Camundongos Knockout , Proteínas do Tecido Nervoso/genética , Regiões Promotoras Genéticas/genética , Receptores Imunológicos/metabolismo , Fatores de Transcrição/biossíntese
16.
Nephrology (Carlton) ; 20(5): 312-4, 2015 May.
Artigo em Inglês | MEDLINE | ID: mdl-25619899

RESUMO

Kidney growth is dependent on functional interactions between mesenchymal nephron progenitors, the ureteric epithelium and surrounding stroma, which together make up the nephrogenic niche. Signalling between these populations regulates nephron progenitor maintenance, branching morphogenesis and nephron induction. Nephron endowment is sensitive to changes in the size of the nephron progenitor pool and to decreases in factors that promote branching morphogenesis. However, determining the morphogenic consequences of these disruptions in vivo has been challenging as quantitating kidney morphogenesis is hampered by the size, opacity and three-dimensional complexity of the tissue. The recent application of whole mount immunofluorescence and tissue clearing, coupled with multiscale imaging and quantitative analysis, has begun to give insights into the dynamics of kidney formation. This review focuses on how the quantitative nature of this approach has enabled mathematical modelling of cell cycle lengths, growth rates, cell number and branching rates and is advancing our understanding of kidney organogenesis.


Assuntos
Rim/embriologia , Organogênese/fisiologia , Ciclo Celular/fisiologia , Proliferação de Células/fisiologia , Humanos , Rim/citologia , Modelos Biológicos
17.
J Clin Invest ; 134(10)2024 Apr 16.
Artigo em Inglês | MEDLINE | ID: mdl-38625739

RESUMO

Renal interstitial fibrosis is an important mechanism in the progression of chronic kidney disease (CKD) to end-stage kidney disease. However, we lack specific treatments to slow or halt renal fibrosis. Ribosome profiling identified upregulation of a secreted micropeptide, C4orf48 (Cf48), in mouse diabetic nephropathy. Cf48 RNA and protein levels were upregulated in tubular epithelial cells in human and experimental CKD. Serum Cf48 levels were increased in human CKD and correlated with loss of kidney function, increasing CKD stage, and the degree of active interstitial fibrosis. Cf48 overexpression in mice accelerated renal fibrosis, while Cf48 gene deletion or knockdown by antisense oligonucleotides significantly reduced renal fibrosis in CKD models. In vitro, recombinant Cf48 (rCf48) enhanced TGF-ß1-induced fibrotic responses in renal fibroblasts and epithelial cells independently of Smad3 phosphorylation. Cellular uptake of Cf48 and its profibrotic response in fibroblasts operated via the transferrin receptor. RNA immunoprecipitation-sequencing identified Cf48 binding to mRNA of genes involved in the fibrotic response, including Serpine1, Acta2, Ccn2, and Col4a1. rCf48 binds to the 3'UTR of Serpine1 and increases mRNA half-life. We identify the secreted Cf48 micropeptide as a potential enhancer of renal fibrosis that operates as an RNA-binding peptide to promote the production of extracellular matrix.


Assuntos
Nefropatias Diabéticas , Fibrose , Proteínas do Tecido Nervoso , Insuficiência Renal Crônica , Animais , Humanos , Masculino , Camundongos , Regiões 3' não Traduzidas , Nefropatias Diabéticas/metabolismo , Nefropatias Diabéticas/patologia , Nefropatias Diabéticas/genética , Rim/metabolismo , Rim/patologia , Camundongos Knockout , Insuficiência Renal Crônica/metabolismo , Insuficiência Renal Crônica/patologia , Insuficiência Renal Crônica/genética , Proteínas de Ligação a RNA/genética , Proteínas de Ligação a RNA/metabolismo , Proteína Smad3/metabolismo , Proteína Smad3/genética , Fator de Crescimento Transformador beta1/metabolismo , Fator de Crescimento Transformador beta1/genética , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/metabolismo
18.
Hum Mol Genet ; 20(11): 2213-24, 2011 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-21427128

RESUMO

X-linked ATR-X (alpha thalassemia, mental retardation, X-linked) syndrome in males is characterized by mental retardation, facial dysmorphism, alpha thalassemia and urogenital abnormalities, including small testes. It is unclear how mutations in the chromatin-remodeling protein ATRX cause these highly specific clinical features, since ATRX is widely expressed during organ development. To investigate the mechanisms underlying the testicular defects observed in ATR-X syndrome, we generated ScAtrxKO (Sertoli cell Atrx knockout) mice with Atrx specifically inactivated in the supporting cell lineage (Sertoli cells) of the mouse testis. ScAtrxKO mice developed small testes and discontinuous tubules, due to prolonged G2/M phase and apoptosis of proliferating Sertoli cells during fetal life. Apoptosis might be a consequence of the cell cycle defect. We also found that the onset of spermatogenesis was delayed in postnatal mice, with a range of spermatogenesis defects evident in adult ScAtrxKO mice. ATRX and the androgen receptor (AR) physically interact in the testis and in the Sertoli cell line TM4 and co-operatively activate the promoter of Rhox5, an important direct AR target. We also demonstrate that ATRX directly binds to the Rhox5 promoter in TM4 cells. Finally, gene expression of Rhox5 and of another AR-dependent gene, Spinlw1, was reduced in ScAtrxKO testes. These data suggest that ATRX can directly enhance the expression of androgen-dependent genes through physical interaction with AR. Recruitment of ATRX by DNA sequence-specific transcription factors could be a general mechanism by which ATRX achieves tissue-specific transcriptional regulation which could explain the highly specific clinical features of ATR-X syndrome when ATRX is mutated.


Assuntos
Proliferação de Células , DNA Helicases/genética , Proteínas Nucleares/genética , Receptores Androgênicos/metabolismo , Células de Sertoli/metabolismo , Animais , Apoptose , Ciclo Celular , Células Cultivadas , Montagem e Desmontagem da Cromatina , DNA Helicases/metabolismo , Modelos Animais de Doenças , Feminino , Regulação da Expressão Gênica , Proteínas de Homeodomínio/genética , Proteínas de Homeodomínio/metabolismo , Imunoprecipitação , Masculino , Deficiência Intelectual Ligada ao Cromossomo X/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Mutação , Proteínas Nucleares/metabolismo , Regiões Promotoras Genéticas , Análise de Sequência de DNA , Testículo/anormalidades , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Proteína Nuclear Ligada ao X , Talassemia alfa/genética
19.
Biol Reprod ; 88(6): 143, 2013 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-23616593

RESUMO

MicroRNAs (miRNAs) have been shown to play key regulatory roles in a range of biological processes, including cell differentiation and development. To identify miRNAs that participate in gonad differentiation, a fundamental and tightly regulated developmental process, we examined miRNA expression profiles at the time of sex determination and during the early fetal differentiation of mouse testes and ovaries using high-throughput sequencing. We identified several miRNAs that were expressed in a sexually dimorphic pattern, including several members of the let-7 family, miR-378, and miR-140-3p. We focused our analysis on the most highly expressed, sexually dimorphic miRNA, miR-140-3p, and found that both miR-140-3p and its more lowly expressed counterpart, the previously annotated guide strand, miR-140-5p, are testis enriched and expressed in testis cords. Analysis of the miR-140-5p/miR-140-3p-null mouse revealed a significant increase in the number of Leydig cells in the developing XY gonad, strongly suggesting an important role for miR-140-5p/miR-140-3p in testis differentiation in mouse.


Assuntos
Diferenciação Celular/genética , Células Intersticiais do Testículo/citologia , MicroRNAs/metabolismo , Testículo/citologia , Animais , Contagem de Células , Células Intersticiais do Testículo/metabolismo , Masculino , Camundongos , Camundongos Knockout , MicroRNAs/genética , Testículo/embriologia , Testículo/metabolismo
20.
Dev Biol ; 360(1): 110-22, 2011 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-21963425

RESUMO

Nephron number in the mammalian kidney is known to vary dramatically, with postnatal renal function directly influenced by nephron complement. What determines final nephron number is poorly understood but nephron formation in the mouse kidney ceases within the first few days after birth, presumably due to the loss of all remaining nephron progenitors via epithelial differentiation. What initiates this event is not known. Indeed, whether nephron formation occurs in the same way at this time as during embryonic development has also not been examined. In this study, we investigate the key cellular compartments involved in nephron formation; the ureteric tip, cap mesenchyme and early nephrons; from postnatal day (P) 0 to 6 in the mouse. High resolution analyses of gene and protein expression indicate that loss of nephron progenitors precedes loss of ureteric tip identity, but show spatial shifts in the expression of cap mesenchyme genes during this time. In addition, cap mesenchymal volume and rate of proliferation decline prior to birth. Section-based 3D modeling and Optical Projection Tomography revealed a burst of ectopic nephron induction, with the formation of multiple (up to 5) nephrons per ureteric tip evident from P2. While the distal-proximal patterning of these nephrons occurred normally, their spatial relationship with the ureteric compartment was altered. We propose that this phase of nephron formation represents an acceleration of differentiation within the cap mesenchyme due to a displacement of signals within the nephrogenic niche.


Assuntos
Rim/crescimento & desenvolvimento , Néfrons/crescimento & desenvolvimento , Animais , Animais Recém-Nascidos , Ciclina D1/genética , Ciclina D1/metabolismo , Feminino , Regulação da Expressão Gênica no Desenvolvimento , Proteínas de Homeodomínio/genética , Proteínas de Homeodomínio/metabolismo , Imageamento Tridimensional , Rim/embriologia , Rim/fisiologia , Camundongos , Modelos Anatômicos , Modelos Biológicos , Néfrons/embriologia , Néfrons/fisiologia , Organogênese/genética , Organogênese/fisiologia , Gravidez , Tomografia Óptica , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Ureter/embriologia , Ureter/crescimento & desenvolvimento
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA