Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 87
Filtrar
Mais filtros

Bases de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
Cell ; 158(6): 1293-1308, 2014 Sep 11.
Artigo em Inglês | MEDLINE | ID: mdl-25215488

RESUMO

Fat (Ft) cadherins are enormous cell adhesion molecules that function at the cell surface to regulate the tumor-suppressive Hippo signaling pathway and planar cell polarity (PCP) tissue organization. Mutations in Ft cadherins are found in a variety of tumors, and it is presumed that this is due to defects in either Hippo signaling or PCP. Here, we show Drosophila Ft functions in mitochondria to directly regulate mitochondrial electron transport chain integrity and promote oxidative phosphorylation. Proteolytic cleavage releases a soluble 68 kDa fragment (Ft(mito)) that is imported into mitochondria. Ft(mito) binds directly to NADH dehydrogenase ubiquinone flavoprotein 2 (Ndufv2), a core component of complex I, stabilizing the holoenzyme. Loss of Ft leads to loss of complex I activity, increases in reactive oxygen species, and a switch to aerobic glycolysis. Defects in mitochondrial activity in ft mutants are independent of Hippo and PCP signaling and are reminiscent of the Warburg effect.


Assuntos
Caderinas/metabolismo , Moléculas de Adesão Celular/metabolismo , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/metabolismo , Mitocôndrias/metabolismo , Sequência de Aminoácidos , Animais , Moléculas de Adesão Celular/química , Polaridade Celular , Proteínas de Drosophila/química , Complexo de Proteínas da Cadeia de Transporte de Elétrons/metabolismo , Complexo I de Transporte de Elétrons/metabolismo , Olho/crescimento & desenvolvimento , Genes Supressores de Tumor , Humanos , MAP Quinase Quinase 4/metabolismo , Dados de Sequência Molecular , Transporte Proteico , Espécies Reativas de Oxigênio/metabolismo , Asas de Animais/crescimento & desenvolvimento
2.
PLoS Biol ; 20(10): e3001811, 2022 10.
Artigo em Inglês | MEDLINE | ID: mdl-36215313

RESUMO

Nuclear envelope membrane proteins (NEMPs) are a conserved family of nuclear envelope (NE) proteins that reside within the inner nuclear membrane (INM). Even though Nemp1 knockout (KO) mice are overtly normal, they display a pronounced splenomegaly. This phenotype and recent reports describing a requirement for NE openings during erythroblasts terminal maturation led us to examine a potential role for Nemp1 in erythropoiesis. Here, we report that Nemp1 KO mice show peripheral blood defects, anemia in neonates, ineffective erythropoiesis, splenomegaly, and stress erythropoiesis. The erythroid lineage of Nemp1 KO mice is overrepresented until the pronounced apoptosis of polychromatophilic erythroblasts. We show that NEMP1 localizes to the NE of erythroblasts and their progenitors. Mechanistically, we discovered that NEMP1 accumulates into aggregates that localize near or at the edge of NE openings and Nemp1 deficiency leads to a marked decrease of both NE openings and ensuing enucleation. Together, our results for the first time demonstrate that NEMP1 is essential for NE openings and erythropoietic maturation in vivo and provide the first mouse model of defective erythropoiesis directly linked to the loss of an INM protein.


Assuntos
Membrana Nuclear , Esplenomegalia , Camundongos , Animais , Eritroblastos/metabolismo , Núcleo Celular/metabolismo , Eritropoese/genética , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Camundongos Knockout
3.
Proc Natl Acad Sci U S A ; 119(29): e2203257119, 2022 07 19.
Artigo em Inglês | MEDLINE | ID: mdl-35858299

RESUMO

How did cells of early metazoan organisms first organize themselves to form a body axis? The canonical Wnt pathway has been shown to be sufficient for induction of axis in Cnidaria, a sister group to Bilateria, and is important in bilaterian axis formation. Here, we provide experimental evidence that in cnidarian Hydra the Hippo pathway regulates the formation of a new axis during budding upstream of the Wnt pathway. The transcriptional target of the Hippo pathway, the transcriptional coactivator YAP, inhibits the initiation of budding in Hydra and is regulated by Hydra LATS. In addition, we show functions of the Hippo pathway in regulation of actin organization and cell proliferation in Hydra. We hypothesize that the Hippo pathway served as a link between continuous cell division, cell density, and axis formation early in metazoan evolution.


Assuntos
Via de Sinalização Hippo , Hydra , Morfogênese , Animais , Padronização Corporal , Hydra/genética , Hydra/crescimento & desenvolvimento , Hydra/metabolismo , Morfogênese/genética , Transcrição Gênica , Proteínas de Sinalização YAP/metabolismo
4.
Genes Dev ; 30(24): 2696-2709, 2016 12 15.
Artigo em Inglês | MEDLINE | ID: mdl-28087714

RESUMO

Disruption of apical-basal polarity is implicated in developmental disorders and cancer; however, the mechanisms connecting cell polarity proteins with intracellular signaling pathways are largely unknown. We determined previously that membrane-associated guanylate kinase (MAGUK) protein discs large homolog 5 (DLG5) functions in cell polarity and regulates cellular proliferation and differentiation via undefined mechanisms. We report here that DLG5 functions as an evolutionarily conserved scaffold and negative regulator of Hippo signaling, which controls organ size through the modulation of cell proliferation and differentiation. Affinity purification/mass spectrometry revealed a critical role of DLG5 in the formation of protein assemblies containing core Hippo kinases mammalian ste20 homologs 1/2 (MST1/2) and Par-1 polarity proteins microtubule affinity-regulating kinases 1/2/3 (MARK1/2/3). Consistent with this finding, Hippo signaling is markedly hyperactive in mammalian Dlg5-/- tissues and cells in vivo and ex vivo and in Drosophila upon dlg5 knockdown. Conditional deletion of Mst1/2 fully rescued the phenotypes of brain-specific Dlg5 knockout mice. Dlg5 also interacts genetically with Hippo effectors Yap1/Taz Mechanistically, we show that DLG5 inhibits the association between MST1/2 and large tumor suppressor homologs 1/2 (LATS1/2), uses its scaffolding function to link MST1/2 with MARK3, and inhibits MST1/2 kinase activity. These data reveal a direct connection between cell polarity proteins and Hippo, which is essential for proper development of multicellular organisms.


Assuntos
Polaridade Celular/genética , Regulação da Expressão Gênica no Desenvolvimento/genética , Proteínas de Membrana/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Transdução de Sinais/genética , Proteínas Supressoras de Tumor/metabolismo , Animais , Células Cultivadas , Drosophila/embriologia , Drosophila/enzimologia , Drosophila/genética , Deleção de Genes , Técnicas de Silenciamento de Genes , Células HEK293 , Humanos , Proteínas de Membrana/genética , Camundongos , Camundongos Knockout , Ligação Proteica , Domínios Proteicos , Proteínas Serina-Treonina Quinases/genética , Proteômica , Interferência de RNA , Proteínas Supressoras de Tumor/genética
5.
Proc Natl Acad Sci U S A ; 117(32): 19310-19320, 2020 08 11.
Artigo em Inglês | MEDLINE | ID: mdl-32727892

RESUMO

Fat, Fat-like, and Dachsous family cadherins are giant proteins that regulate planar cell polarity (PCP) and cell adhesion in bilaterians. Their evolutionary origin can be traced back to prebilaterian species, but their ancestral function(s) are unknown. We identified Fat-like and Dachsous cadherins in Hydra, a member of phylum Cnidaria a sister group of bilaterian. We found Hydra does not possess a true Fat homolog, but has homologs of Fat-like (HyFatl) and Dachsous (HyDs) that localize at the apical membrane of ectodermal epithelial cells and are planar polarized perpendicular to the oral-aboral axis of the animal. Using a knockdown approach we found that HyFatl is involved in local cell alignment and cell-cell adhesion, and that reduction of HyFatl leads to defects in tissue organization in the body column. Overexpression and knockdown experiments indicate that the intracellular domain (ICD) of HyFatl affects actin organization through proline-rich repeats. Thus, planar polarization of Fat-like and Dachsous cadherins has ancient, prebilaterian origins, and Fat-like cadherins have ancient roles in cell adhesion, spindle orientation, and tissue organization.


Assuntos
Caderinas/metabolismo , Polaridade Celular , Hydra/citologia , Animais , Caderinas/genética , Adesão Celular , Hydra/classificação , Hydra/genética , Hydra/metabolismo , Filogenia , Fuso Acromático/genética , Fuso Acromático/metabolismo
6.
Nat Rev Mol Cell Biol ; 11(6): 404-13, 2010 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-20461097

RESUMO

Signal transduction pathways interact at various levels to define tissue morphology, size and differentiation during development. Understanding the mechanisms by which these pathways collude has been greatly enhanced by recent insights into how shared components are independently regulated and how the activity of one system is contextualized by others. Traditionally, it has been assumed that the components of signalling pathways show pathway fidelity and act with a high degree of autonomy. However, as illustrated by the Wnt and Hippo pathways, there is increasing evidence that components are often shared between multiple pathways and other components talk to each other through multiple mechanisms.


Assuntos
Transdução de Sinais , Adipócitos/metabolismo , Animais , Regulação da Expressão Gênica no Desenvolvimento , Humanos , Ligação Proteica , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas Wnt/metabolismo
8.
Dev Dyn ; 249(4): 523-542, 2020 04.
Artigo em Inglês | MEDLINE | ID: mdl-31747096

RESUMO

BACKGROUND: Normal skeletal development, in particular ossification, joint formation and shape features of condyles, depends on appropriate mechanical input from embryonic movement but it is unknown how such physical stimuli are transduced to alter gene regulation. Hippo/Yes-Associated Protein (YAP) signalling has been shown to respond to the physical environment of the cell and here we specifically investigate the YAP effector of the pathway as a potential mechanoresponsive mediator in the developing limb skeleton. RESULTS: We show spatial localization of YAP protein and of pathway target gene expression within developing skeletal rudiments where predicted biophysical stimuli patterns and shape are affected in immobilization models, coincident with the period of sensitivity to movement, but not coincident with the expression of the Hippo receptor Fat4. Furthermore, we show that under reduced mechanical stimulation, in immobile, muscle-less mouse embryos, this spatial localization is lost. In culture blocking YAP reduces chondrogenesis but the effect differs depending on the timing and/or level of YAP reduction. CONCLUSIONS: These findings implicate YAP signalling, independent of Fat4, in the transduction of mechanical signals during key stages of skeletal patterning in the developing limb, in particular endochondral ossification and shape emergence, as well as patterning of tissues at the developing synovial joint.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Proteínas de Ciclo Celular/metabolismo , Extremidades/embriologia , Esqueleto/embriologia , Proteínas Adaptadoras de Transdução de Sinal/genética , Animais , Caderinas/genética , Caderinas/metabolismo , Proteínas de Ciclo Celular/genética , Feminino , Masculino , Camundongos , Transdução de Sinais/genética , Transdução de Sinais/fisiologia , Esqueleto/citologia , Esqueleto/metabolismo , Proteínas de Sinalização YAP
9.
Dev Biol ; 450(1): 23-33, 2019 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-30858024

RESUMO

Development of an organism requires accurate coordination between the growth of a tissue and orientation of cells within the tissue. The large cadherin Fat has been shown to play a role in both of these processes. Fat is involved in the establishment of planar cell polarity and regulates growth through the Hippo pathway, a developmental cascade that controls proliferation and apoptosis. Both Fat and the Hippo pathway are known to regulate transcription of four-jointed, although the nature of this regulation is unknown. In this study, we test whether Fat affects four-jointed transcription via or independently of Hippo pathway. Our analysis of the four-jointed regulatory region reveals a 1.2 kb element that functions as an enhancer for graded expression of Four-jointed in the eye imaginal disc. Within this enhancer element, we identify a 20 bp fragment that is critical for regulation by Fat but not by Hippo. Our findings suggest that Fat and the Hippo pathway control four-jointed expression independently of each other and none of the transcription factors known to function downstream of the Hippo pathway are required to regulate four-jointed expression through the 1.2 kb element.


Assuntos
Moléculas de Adesão Celular , Proteínas de Drosophila , Elementos Facilitadores Genéticos , Regulação da Expressão Gênica no Desenvolvimento , Peptídeos e Proteínas de Sinalização Intracelular , Glicoproteínas de Membrana , Proteínas Serina-Treonina Quinases , Transdução de Sinais , Transcrição Gênica , Animais , Moléculas de Adesão Celular/genética , Moléculas de Adesão Celular/metabolismo , Proteínas de Drosophila/biossíntese , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Drosophila melanogaster , Genes Reporter , Peptídeos e Proteínas de Sinalização Intracelular/genética , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Glicoproteínas de Membrana/biossíntese , Glicoproteínas de Membrana/genética , Proteínas Serina-Treonina Quinases/genética , Proteínas Serina-Treonina Quinases/metabolismo
10.
J Cell Sci ; 131(13)2018 07 04.
Artigo em Inglês | MEDLINE | ID: mdl-29777038

RESUMO

Extracellular forces transmitted through the cytoskeleton can deform the cell nucleus. Large nuclear deformations increase the risk of disrupting the integrity of the nuclear envelope and causing DNA damage. The mechanical stability of the nucleus defines its capability to maintain nuclear shape by minimizing nuclear deformation and allowing strain to be minimized when deformed. Understanding the deformation and recovery behavior of the nucleus requires characterization of nuclear viscoelastic properties. Here, we quantified the decoupled viscoelastic parameters of the cell membrane, cytoskeleton, and the nucleus. The results indicate that the cytoskeleton enhances nuclear mechanical stability by lowering the effective deformability of the nucleus while maintaining nuclear sensitivity to mechanical stimuli. Additionally, the cytoskeleton decreases the strain energy release rate of the nucleus and might thus prevent shape change-induced structural damage to chromatin.


Assuntos
Núcleo Celular/química , Linhagem Celular , Membrana Celular/química , Membrana Celular/genética , Membrana Celular/metabolismo , Núcleo Celular/genética , Núcleo Celular/metabolismo , Forma do Núcleo Celular , Citoesqueleto/química , Citoesqueleto/genética , Citoesqueleto/metabolismo , Humanos , Membrana Nuclear/química , Membrana Nuclear/genética , Membrana Nuclear/metabolismo , Estresse Mecânico
11.
J Pathol ; 249(1): 114-125, 2019 09.
Artigo em Inglês | MEDLINE | ID: mdl-31038742

RESUMO

Autosomal Dominant Polycystic Kidney Disease is characterised by the development of fluid-filled cysts in the kidneys which lead to end-stage renal disease (ESRD). In the majority of cases, the disease is caused by a mutation in the Pkd1 gene. In a previous study, we demonstrated that renal injury can accelerate cyst formation in Pkd1 knock-out (KO) mice. In that study, we found that after injury four-jointed (Fjx1), an upstream regulator of planar cell polarity and the Hippo pathway, was aberrantly expressed in Pkd1 KO mice compared to WT. Therefore, we hypothesised a role for Fjx1 in injury/repair and cyst formation. We generated single and double deletion mice for Pkd1 and Fjx1, and we induced toxic renal injury using the nephrotoxic compound 1,2-dichlorovinyl-cysteine. We confirmed that nephrotoxic injury can accelerate cyst formation in Pkd1 mutant mice. This caused Pkd1 KO mice to reach ESRD significantly faster; unexpectedly, double KO mice survived significantly longer. Cyst formation was comparable in both models, but we found significantly less fibrosis and macrophage infiltration in double KO mice. Taken together, these data suggest that Fjx1 disruption protects the cystic kidneys against kidney failure by reducing inflammation and fibrosis. Moreover, we describe, for the first time, an interesting (yet unidentified) mechanism that partially discriminates cyst growth from fibrogenesis. © 2019 The Authors. The Journal of Pathology published by John Wiley & Sons Ltd on behalf of Pathological Society of Great Britain and Ireland.


Assuntos
Injúria Renal Aguda/complicações , Peptídeos e Proteínas de Sinalização Intercelular/deficiência , Falência Renal Crônica/etiologia , Rim/metabolismo , Rim Policístico Autossômico Dominante/complicações , Injúria Renal Aguda/induzido quimicamente , Injúria Renal Aguda/genética , Injúria Renal Aguda/metabolismo , Animais , Cisteína/análogos & derivados , Modelos Animais de Doenças , Progressão da Doença , Fibrose , Mediadores da Inflamação/metabolismo , Peptídeos e Proteínas de Sinalização Intercelular/genética , Rim/patologia , Falência Renal Crônica/genética , Falência Renal Crônica/metabolismo , Masculino , Camundongos Knockout , Mutação , Rim Policístico Autossômico Dominante/genética , Rim Policístico Autossômico Dominante/metabolismo , Canais de Cátion TRPP/genética , Fatores de Tempo , Via de Sinalização Wnt
12.
Biochem Soc Trans ; 47(3): 839-845, 2019 06 28.
Artigo em Inglês | MEDLINE | ID: mdl-31189732

RESUMO

The activity of any bacterial promoter is generally supposed to be set by its base sequence and the different transcription factors that bind in the local vicinity. Here, we review recent data indicating that the activity of the Escherichia coli lac operon promoter also depends upon its chromosomal location. Factors that affect promoter activity include the binding of nucleoid-associated proteins to neighbouring sequences, supercoiling and the activity of neighbouring promoters. We suggest that many bacterial promoters might be susceptible to similar position-dependent effects and we review recent data showing that the expression of mobile genes encoding antibiotic-resistance determinants is also location-dependent, both when carried on a bacterial chromosome or a conjugative plasmid.


Assuntos
Efeitos da Posição Cromossômica , Resistência Microbiana a Medicamentos/genética , Escherichia coli/genética , Regiões Promotoras Genéticas , Cromossomos Bacterianos , Elementos de DNA Transponíveis , Óperon Lac , Plasmídeos , Transcrição Gênica
13.
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
14.
Kidney Int ; 93(5): 1142-1153, 2018 05.
Artigo em Inglês | MEDLINE | ID: mdl-29459093

RESUMO

Congenital abnormalities of the kidney and the urinary tract (CAKUT) belong to the most common birth defects in human, but the molecular basis for the majority of CAKUT patients remains unknown. Here we show that the transcription factor SOX11 is a crucial regulator of kidney development. SOX11 is expressed in both mesenchymal and epithelial components of the early kidney anlagen. Deletion of Sox11 in mice causes an extension of the domain expressing Gdnf within rostral regions of the nephrogenic cord and results in duplex kidney formation. On the molecular level SOX11 directly binds and regulates a locus control region of the protocadherin B cluster. At later stages of kidney development, SOX11 becomes restricted to the intermediate segment of the developing nephron where it is required for the elongation of Henle's loop. Finally, mutation analysis in a cohort of patients suffering from CAKUT identified a series of rare SOX11 variants, one of which interferes with the transactivation capacity of the SOX11 protein. Taken together these data demonstrate a key role for SOX11 in normal kidney development and may suggest that variants in this gene predispose to CAKUT in humans.


Assuntos
Rim/anormalidades , Mutação , Fatores de Transcrição SOXC/genética , Ureter/anormalidades , Anormalidades Urogenitais/genética , Refluxo Vesicoureteral/genética , Animais , Caderinas/genética , Caderinas/metabolismo , Proliferação de Células , Modelos Animais de Doenças , Feminino , Regulação da Expressão Gênica no Desenvolvimento , Estudos de Associação Genética , Predisposição Genética para Doença , Fator Neurotrófico Derivado de Linhagem de Célula Glial/genética , Fator Neurotrófico Derivado de Linhagem de Célula Glial/metabolismo , Humanos , Rim/metabolismo , Masculino , Camundongos Knockout , Morfogênese , Fenótipo , Fatores de Risco , Fatores de Transcrição SOXC/deficiência , Ureter/metabolismo , Anormalidades Urogenitais/metabolismo , Anormalidades Urogenitais/patologia , Refluxo Vesicoureteral/metabolismo , Refluxo Vesicoureteral/patologia
15.
Development ; 142(15): 2696-703, 2015 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-26243870

RESUMO

Despite the high occurrence of congenital abnormalities of the lower urinary tract in humans, the molecular, cellular and morphological aspects of their development are still poorly understood. Here, we use a conditional knockout approach to inactivate within the nephric duct (ND) lineage the two effectors of the Hippo pathway, Yap and Taz. Deletion of Yap leads to hydronephrotic kidneys with blind-ending megaureters at birth. In Yap mutants, the ND successfully migrates towards, and contacts, the cloaca. However, close analysis reveals that the tip of the Yap(-/-) ND forms an aberrant connection with the cloaca and does not properly insert into the cloaca, leading to later detachment of the ND from the cloaca. Taz deletion from the ND does not cause any defect, but analysis of Yap(-/-);Taz(-/-) NDs indicates that both genes play partially redundant roles in ureterovesical junction formation. Aspects of the Yap(-/-) phenotype resemble hypersensitivity to RET signaling, including excess budding of the ND, increased phospho-ERK and increased expression of Crlf1, Sprouty1, Etv4 and Etv5. Importantly, the Yap(ND) (-/-) ND phenotype can be largely rescued by reducing Ret gene dosage. Taken together, these results suggest that disrupting Yap/Taz activities enhances Ret pathway activity and contributes to pathogenesis of lower urinary tract defects in human infants.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Morfogênese/fisiologia , Fosfoproteínas/metabolismo , Proteínas Proto-Oncogênicas c-ret/metabolismo , Transdução de Sinais/fisiologia , Sistema Urinário/embriologia , Proteínas Adaptadoras de Transdução de Sinal/genética , Animais , Bromodesoxiuridina , Proteínas de Ciclo Celular , Imunofluorescência , Galactosídeos , Técnicas de Inativação de Genes , Humanos , Imuno-Histoquímica , Hibridização In Situ , Marcação In Situ das Extremidades Cortadas , Indóis , Camundongos , Fosfoproteínas/genética , Transativadores , Proteínas de Sinalização YAP
16.
Development ; 142(15): 2564-73, 2015 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-26116661

RESUMO

Regulation of the balance between progenitor self-renewal and differentiation is crucial to development. In the mammalian kidney, reciprocal signalling between three lineages (stromal, mesenchymal and ureteric) ensures correct nephron progenitor self-renewal and differentiation. Loss of either the atypical cadherin FAT4 or its ligand Dachsous 1 (DCHS1) results in expansion of the mesenchymal nephron progenitor pool, called the condensing mesenchyme (CM). This has been proposed to be due to misregulation of the Hippo kinase pathway transcriptional co-activator YAP. Here, we use tissue-specific deletions to prove that FAT4 acts non-autonomously in the renal stroma to control nephron progenitors. We show that loss of Yap from the CM in Fat4-null mice does not reduce the expanded CM, indicating that FAT4 regulates the CM independently of YAP. Analysis of Six2(-/-);Fat4(-/-) double mutants demonstrates that excess progenitors in Fat4 mutants are dependent on Six2, a crucial regulator of nephron progenitor self-renewal. Electron microscopy reveals that cell organisation is disrupted in Fat4 mutants. Gene expression analysis demonstrates that the expression of Notch and FGF pathway components are altered in Fat4 mutants. Finally, we show that Dchs1, and its paralogue Dchs2, function in a partially redundant fashion to regulate the number of nephron progenitors. Our data support a model in which FAT4 in the stroma binds to DCHS1/2 in the mouse CM to restrict progenitor self-renewal.


Assuntos
Caderinas/metabolismo , Diferenciação Celular/fisiologia , Néfrons/ultraestrutura , Transdução de Sinais/fisiologia , Células-Tronco/citologia , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Análise de Variância , Animais , Proteínas de Ciclo Celular , Linhagem da Célula/fisiologia , Imunofluorescência , Perfilação da Expressão Gênica , Via de Sinalização Hippo , Immunoblotting , Hibridização In Situ , Marcação In Situ das Extremidades Cortadas , Camundongos , Camundongos Knockout , Microscopia Eletrônica , Fosfoproteínas/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Análise de Sequência de RNA , Proteínas de Sinalização YAP
17.
Development ; 142(16): 2781-91, 2015 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-26209645

RESUMO

Mammalian brain development requires coordination between neural precursor proliferation, differentiation and cellular organization to create the intricate neuronal networks of the adult brain. Here, we examined the role of the atypical cadherins Fat1 and Fat4 in this process. We show that mutation of Fat1 in mouse embryos causes defects in cranial neural tube closure, accompanied by an increase in the proliferation of cortical precursors and altered apical junctions, with perturbations in apical constriction and actin accumulation. Similarly, knockdown of Fat1 in cortical precursors by in utero electroporation leads to overproliferation of radial glial precursors. Fat1 interacts genetically with the related cadherin Fat4 to regulate these processes. Proteomic analysis reveals that Fat1 and Fat4 bind different sets of actin-regulating and junctional proteins. In vitro data suggest that Fat1 and Fat4 form cis-heterodimers, providing a mechanism for bringing together their diverse interactors. We propose a model in which Fat1 and Fat4 binding coordinates distinct pathways at apical junctions to regulate neural progenitor proliferation, neural tube closure and apical constriction.


Assuntos
Encéfalo/embriologia , Caderinas/metabolismo , Proliferação de Células/fisiologia , Células-Tronco Neurais/fisiologia , Tubo Neural/embriologia , Animais , Western Blotting , Caderinas/genética , Técnicas de Silenciamento de Genes , Camundongos , Microscopia de Fluorescência , Tubo Neural/metabolismo , beta-Galactosidase
18.
J Am Soc Nephrol ; 28(3): 852-861, 2017 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-27647853

RESUMO

In the kidney, formation of the functional filtration units, the nephrons, is essential for postnatal life. During development, mesenchymal progenitors tightly regulate the balance between self-renewal and differentiation to give rise to all nephron epithelia. Here, we investigated the functions of the Hippo pathway serine/threonine-protein kinases Lats1 and Lats2, which phosphorylate and inhibit the transcriptional coactivators Yap and Taz, in nephron progenitor cells. Genetic deletion of Lats1 and Lats2 in nephron progenitors of mice led to disruption of nephrogenesis, with an accumulation of spindle-shaped cells in both cortical and medullary regions of the kidney. Lineage-tracing experiments revealed that the cells that accumulated in the interstitium derived from nephron progenitor cells and expressed E-cadherin as well as vimentin, a myofibroblastic marker not usually detected after mesenchymal-to-epithelial transition. The accumulation of these interstitial cells associated with collagen deposition and ectopic expression of the myofibroblastic markers vimentin and α-smooth-muscle actin in developing kidneys. Although these myofibroblastic cells had high Yap and Taz accumulation in the nucleus concomitant with a loss of phosphorylated Yap, reduction of Yap and/or Taz expression levels completely rescued the Lats1/2 phenotype. Taken together, our results demonstrate that Lats1/2 kinases restrict Yap/Taz activities to promote nephron progenitor cell differentiation in the mammalian kidney. Notably, our data also show that myofibroblastic cells can differentiate from nephron progenitors.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/fisiologia , Miofibroblastos/fisiologia , Néfrons/citologia , Fosfoproteínas/fisiologia , Proteínas Serina-Treonina Quinases/fisiologia , Células-Tronco/fisiologia , Fatores de Transcrição/fisiologia , Proteínas Supressoras de Tumor/fisiologia , Aciltransferases , Animais , Proteínas de Ciclo Celular , Diferenciação Celular , Epitélio , Camundongos , Fosforilação , Proteínas de Sinalização YAP
19.
Educ Prim Care ; 29(3): 174-177, 2018 05.
Artigo em Inglês | MEDLINE | ID: mdl-29303045

RESUMO

Background A '1-hour protected supervision model' is well established for Psychiatry trainees. This model is also extended to GP trainees who are on placement in psychiatry. AIM: To explore the experiences of the '1-hour protected supervision model' for GP trainees in psychiatry placements in the UK. Methods Using a mixed methods approach, an anonymous online questionnaire was sent to GP trainees in the North West of England who had completed a placement in Psychiatry between February and August 2015. Results Discussing clinical cases whilst using the e-portfolio was the most useful learning event in this model. Patient care can potentially improve if a positive relationship develops between trainee/supervisor, which is impacted by the knowledge of this model at the start of the placement. Trainees found that clinical pressures were impacting on the occurrence of supervision. Conclusion The model works best when both GP trainees and their supervisors understand the model. The most frequently used and educationally beneficial aspect for GP trainees in psychiatry is the exploration of clinical cases using the learning portfolio as an educational tool. For effective delivery of this model of supervision, organisations must reflect on the balance between service delivery and allowing the supervisor and trainee adequate time for it to occur.


Assuntos
Clínicos Gerais/educação , Psiquiatria/educação , Competência Clínica , Humanos , Internato e Residência/métodos , Inquéritos e Questionários , Reino Unido
20.
PLoS Genet ; 10(9): e1004624, 2014 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-25210733

RESUMO

Neuronal differentiation is exquisitely controlled both spatially and temporally during nervous system development. Defects in the spatiotemporal control of neurogenesis cause incorrect formation of neural networks and lead to neurological disorders such as epilepsy and autism. The mTOR kinase integrates signals from mitogens, nutrients and energy levels to regulate growth, autophagy and metabolism. We previously identified the insulin receptor (InR)/mTOR pathway as a critical regulator of the timing of neuronal differentiation in the Drosophila melanogaster eye. Subsequently, this pathway has been shown to play a conserved role in regulating neurogenesis in vertebrates. However, the factors that mediate the neurogenic role of this pathway are completely unknown. To identify downstream effectors of the InR/mTOR pathway we screened transcriptional targets of mTOR for neuronal differentiation phenotypes in photoreceptor neurons. We identified the conserved gene unkempt (unk), which encodes a zinc finger/RING domain containing protein, as a negative regulator of the timing of photoreceptor differentiation. Loss of unk phenocopies InR/mTOR pathway activation and unk acts downstream of this pathway to regulate neurogenesis. In contrast to InR/mTOR signalling, unk does not regulate growth. unk therefore uncouples the role of the InR/mTOR pathway in neurogenesis from its role in growth control. We also identified the gene headcase (hdc) as a second downstream regulator of the InR/mTOR pathway controlling the timing of neurogenesis. Unk forms a complex with Hdc, and Hdc expression is regulated by unk and InR/mTOR signalling. Co-overexpression of unk and hdc completely suppresses the precocious neuronal differentiation phenotype caused by loss of Tsc1. Thus, Unk and Hdc are the first neurogenic components of the InR/mTOR pathway to be identified. Finally, we show that Unkempt-like is expressed in the developing mouse retina and in neural stem/progenitor cells, suggesting that the role of Unk in neurogenesis may be conserved in mammals.


Assuntos
Diferenciação Celular/genética , Proteínas de Ligação a DNA/genética , Proteínas de Drosophila/genética , Drosophila/genética , Drosophila/metabolismo , Regulação da Expressão Gênica , Neurônios/citologia , Neurônios/metabolismo , Serina-Treonina Quinases TOR/metabolismo , Animais , Encéfalo/metabolismo , Linhagem Celular , Proliferação de Células , Proteínas de Drosophila/metabolismo , Mutação , Células Fotorreceptoras/citologia , Células Fotorreceptoras/metabolismo , Ligação Proteica , Interferência de RNA , Retina/metabolismo , Transdução de Sinais
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA