Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
2.
PLoS Genet ; 18(6): e1010240, 2022 06.
Artigo em Inglês | MEDLINE | ID: mdl-35704566

RESUMO

Assessing the role of the WT1 transcription factor (WT1) during early gonad differentiation and its impact on adult sex development has been difficult due to the complete gonadal agenesis and embryonic lethality exhibited by Wt1KO mouse models. Here, we generated Wt1LoxP/GFP;Wt1Cre mice, the first Wt1KO mouse model that reaches adulthood with a dramatically reduced Wt1 expression during early gonadogenesis. Wt1LoxP/GFP;Wt1Cre mice lacked mature gonads and displayed genital tracts containing both male and female genital structures and ambiguous genitalia. We found that WT1 is necessary for the activation of both male and female sex-determining pathways, as embryonic mutant gonads failed to upregulate the expression of the genes specific for each genetic programme. The gonads of Wt1LoxP/GFP;Wt1Cre mice showed a lack of production of Sertoli and pre-granulosa cells and a reduced number of germ cells. NR5A1 and the steroidogenic genes expression was modulated differently in XY and XX Wt1LoxP/GFP;Wt1Cre gonads, explaining the mutant phenotypes. Further studies of the XX Wt1LoxP/GFP;Wt1Cre gonads revealed that deletion of WT1 at an early stage impaired the differentiation of several cell types including somatic cells and the ovarian epithelium. Through the characterisation of this Wt1KO mouse model, we show that the deletion of Wt1 during early gonadogenesis produces dramatic defects in adult sex development.


Assuntos
Gônadas , Diferenciação Sexual , Animais , Diferenciação Celular/genética , Feminino , Gônadas/metabolismo , Masculino , Camundongos , Ovário/metabolismo , Diferenciação Sexual/genética , Desenvolvimento Sexual , Testículo/metabolismo , Proteínas WT1/genética , Proteínas WT1/metabolismo
3.
Biomed Pharmacother ; 144: 112349, 2021 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-34700229

RESUMO

Membranous nephropathy (MN) is the most common cause of nephrotic syndrome in adults without diabetes. Primary MN has been associated with circulating antibodies against native podocyte antigens, including phospholipase A2 receptor (PLA2R); however, precision therapy targeting the signaling cascade of PLA2R activation is lacking. Both PLA2R and the mammalian target of rapamycin (mTOR) exist in podocytes, but the interplay between these two proteins and their roles in MN warrants further exploration. This study aimed to investigate the crosstalk between PLA2R activation and mTOR signaling in a human podocyte cell line. We demonstrated that podocyte apoptosis was induced by Group IB secretory phospholipase A2 (sPLA2IB) in a concentration- and time-dependent manner via upregulation of phosphoinositide 3-kinase (PI3K), protein kinase B (AKT), and mTOR, and inhibited by rapamycin or LY294002. Furthermore, aberrant activation of the PI3K/AKT/mTOR pathway triggers both extrinsic (caspase-8 and caspase-3) and intrinsic (Bcl-2-associated X protein [BAX], B-cell lymphoma 2 [BCL-2], cytochrome c, caspase-9, and caspase-3) apoptotic cascades in podocytes. The therapeutic implications of our findings are that strategies to reduce PLA2R activation and PI3K/AKT/mTOR pathway inhibition in PLA2R-activated podocytes help protect podocytes from apoptosis. The therapeutic potential of rapamycin shown in this study provides cellular evidence supporting the repurposing of rapamycin for MN treatment.


Assuntos
Apoptose/efeitos dos fármacos , Glomerulonefrite Membranosa/tratamento farmacológico , Inibidores de MTOR/farmacologia , Fosfatidilinositol 3-Quinase/metabolismo , Podócitos/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-akt/metabolismo , Receptores da Fosfolipase A2/metabolismo , Sirolimo/farmacologia , Serina-Treonina Quinases TOR/antagonistas & inibidores , Proteínas Reguladoras de Apoptose/metabolismo , Linhagem Celular , Ativação Enzimática , Glomerulonefrite Membranosa/enzimologia , Glomerulonefrite Membranosa/patologia , Humanos , Podócitos/enzimologia , Podócitos/patologia , Transdução de Sinais , Serina-Treonina Quinases TOR/metabolismo
4.
Dis Model Mech ; 14(1)2021 01 26.
Artigo em Inglês | MEDLINE | ID: mdl-33735101

RESUMO

Congenital diaphragmatic hernia (CDH) is a relatively common developmental defect with considerable mortality and morbidity. Formation of the diaphragm is a complex process that involves several cell types, each with different developmental origins. Owing to this complexity, the aetiology of CDH is not well understood. The pleuroperitoneal folds (PPFs) and the posthepatic mesenchymal plate (PHMP) are transient structures that are essential during diaphragm development. Using several mouse models, including lineage tracing, we demonstrate the heterogeneous nature of the cells that make up the PPFs. The conditional deletion of Wilms tumor 1 homolog (Wt1) in the non-muscle mesenchyme of the PPFs results in CDH. We show that the fusion of the PPFs and the PHMP to form a continuous band of tissue involves movements of cells from both sources. The PPFs of mutant mice fail to fuse with the PHMP and exhibit increased RALDH2 (also known as ALDH1A2) expression. However, no changes in the expression of genes (including Snai1, Snai2, Cdh1 and Vim) implicated in epithelial-to-mesenchymal transition are observed. Additionally, the mutant PPFs lack migrating myoblasts and muscle connective tissue fibroblasts (TCF4+/GATA4+), suggesting possible interactions between these cell types. Our study demonstrates the importance of the non-muscle mesenchyme in development of the diaphragm.


Assuntos
Diafragma/patologia , Regulação da Expressão Gênica no Desenvolvimento , Mesoderma/metabolismo , Animais , Tecido Conjuntivo , Modelos Animais de Doenças , Feminino , Fibroblastos/metabolismo , Deleção de Genes , Perfilação da Expressão Gênica , Hérnias Diafragmáticas Congênitas/genética , Masculino , Camundongos , Desenvolvimento Muscular , Fatores de Tempo , Transgenes , Proteínas WT1/metabolismo
5.
Development ; 146(20)2019 10 17.
Artigo em Inglês | MEDLINE | ID: mdl-31624071

RESUMO

The epicardium plays a crucial role in embryonic heart development and adult heart repair; however, the molecular events underlying its maturation remain unknown. Wt1, one of the main markers of the embryonic epicardium, is essential for epicardial development and function. Here, we analyse the transcriptomic profile of epicardial-enriched cells at different stages of development and from control and epicardial-specific Wt1 knockout (Wt1KO) mice. Transcriptomic and cell morphology analyses of epicardial cells from epicardial-specific Wt1KO mice revealed a defect in the maturation process of the mutant epicardium, including sustained upregulation of Bmp4 expression and the inability of mutant epicardial cells to transition into a mature squamous phenotype. We identified Bmp4 as a transcriptional target of Wt1, thus providing a molecular basis for the retention of the cuboidal cell shape observed in the Wt1KO epicardium. Accordingly, inhibition of the Bmp4 signalling pathway both ex vivo and in vivo rescued the cuboidal phenotype of the mutant epicardium. Our findings indicate the importance of the cuboidal-to-squamous transition in epicardial maturation, a process regulated by Wt1.


Assuntos
Proteína Morfogenética Óssea 4/metabolismo , Proteína Morfogenética Óssea 4/farmacologia , Pericárdio/citologia , Pericárdio/metabolismo , Proteínas WT1/metabolismo , Animais , Forma Celular/efeitos dos fármacos , Forma Celular/genética , Células Cultivadas , Feminino , Citometria de Fluxo , Regulação da Expressão Gênica no Desenvolvimento/efeitos dos fármacos , Regulação da Expressão Gênica no Desenvolvimento/genética , Coração/efeitos dos fármacos , Masculino , Camundongos , Camundongos Knockout , Microscopia Eletrônica de Varredura , Microscopia Eletrônica de Transmissão , Miocárdio/metabolismo , Miocárdio/ultraestrutura , Pericárdio/efeitos dos fármacos , Pericárdio/ultraestrutura , Proteínas WT1/genética
6.
Cells ; 8(9)2019 09 14.
Artigo em Inglês | MEDLINE | ID: mdl-31540085

RESUMO

Empagliflozin is known to retard the progression of kidney disease in diabetic patients. However, the underlying mechanism is incompletely understood. High glucose induces oxidative stress in renal tubules, eventually leading to mitochondrial damage. Here, we investigated whether empagliflozin exhibits protective functions in renal tubules via a mitochondrial mechanism. We used human proximal tubular cell (PTC) line HK-2 and employed western blotting, terminal deoxynucleotidyl transferase dUTP nick end labelling assay, fluorescence staining, flow cytometry, and enzyme-linked immunosorbent assay to investigate the impact of high glucose and empagliflozin on cellular apoptosis, mitochondrial morphology, and functions including mitochondrial membrane potential (MMP), reactive oxygen species (ROS) production, and adenosine triphosphate (ATP) generation. We found that PTCs were susceptible to high glucose-induced mitochondrial fragmentation, and empagliflozin ameliorated this effect via the regulation of mitochondrial fission (FIS1 and DRP1) and fusion (MFN1 and MFN2) proteins. Empagliflozin reduced the high glucose-induced cellular apoptosis and improved mitochondrial functions by restoring mitochondrial ROS production, MMP, and ATP generation. Our results suggest that empagliflozin may protect renal PTCs from high glucose-mediated injuries through a mitochondrial mechanism. This could be one of the novel mechanisms explaining the benefits demonstrated in EMPA-REG OUTCOME trial.


Assuntos
Glucose/farmacologia , Mitocôndrias/efeitos dos fármacos , Apoptose/efeitos dos fármacos , Células Cultivadas , Humanos , Mitocôndrias/metabolismo , Mitocôndrias/patologia , Estresse Oxidativo/efeitos dos fármacos
7.
Science ; 364(6438): 328-329, 2019 04 26.
Artigo em Inglês | MEDLINE | ID: mdl-31023910
8.
Br J Cancer ; 119(12): 1508-1517, 2018 12.
Artigo em Inglês | MEDLINE | ID: mdl-30374123

RESUMO

BACKGROUND: The Wilms' tumour protein (WT1), which influences tumour development and angiogenesis, is a promising therapeutic target in breast cancer. We hypothesised that WT1 expression would vary in endothelial cells in distinct sub-classifications of breast cancer. METHODS: WT1 expression and vascular density were quantified by immunohistochemical analysis of human (n = 57) and murine breast cancers. Human tumours were sub-classified by histopathological grade, ER status and HER2 enrichment. RESULTS: WT1 was identified in endothelial (and epithelial and smooth muscle) cells in tumours and tumour-free tissues (controls) from patients and mice with breast cancer. WT1 expression was higher in tumours than in controls, but this was not due to increased endothelial WT1. Vascular WT1 in cancers decreased as histopathological grade increased. WT1 was higher in ER-positive versus ER-negative cancers. Strikingly, reduced WT1 expression in controls correlated with an increased Nottingham Prognostic Index score. CONCLUSIONS: Expression of WT1 is increased in breast cancers but this is not limited to the vascular compartment. The association between reduced WT1 in tumour-free tissue and poor prognosis suggests a protective role for WT1 in the healthy breast.


Assuntos
Neoplasias da Mama/patologia , Proteínas WT1/análise , Animais , Neoplasias da Mama/irrigação sanguínea , Neoplasias da Mama/química , Neoplasias da Mama/mortalidade , Feminino , Humanos , Camundongos , Gradação de Tumores , Receptor ErbB-2/análise , Receptores de Estrogênio/análise , Proteínas WT1/fisiologia
9.
Adipocyte ; 6(3): 205-216, 2017 07 03.
Artigo em Inglês | MEDLINE | ID: mdl-28949833

RESUMO

The excessive expansion of white adipose tissue underlies the global obesity epidemic. However, not all fat is equal, and the impact of heterogeneity on the development and expansion of different adipose depots is becoming increasingly apparent. Two mechanisms are responsible for the growth of adipose tissue: hyperplasia (increasing adipocyte number) and hypertrophy (increasing adipocyte size). The former relies on the differentiation of adipocyte stem cells, which reside within the adipose stromal vascular fraction. Many differences in gene expression, adipogenesis, and the response to obesogenic stimuli have been described when comparing adipose stem cells from different depots. Considering that there is disparity in the pathogenicity of the depots, understanding this heterogeneity has clinically relevant implications. Here we review the current knowledge surrounding such differences, in the context of development, expansion and therapeutics. Moreover, given the importance of these differences, we suggest that careful consideration for the precise methodologies used, is essential if we are to truly understand the physiologically relevant consequences of this heterogeneity.


Assuntos
Adipócitos Brancos/citologia , Tecido Adiposo Branco/fisiologia , Adipócitos/citologia , Adipócitos Brancos/metabolismo , Adipogenia/fisiologia , Adipocinas/genética , Adipocinas/metabolismo , Tecido Adiposo/metabolismo , Tecido Adiposo Marrom/citologia , Tecido Adiposo Branco/citologia , Adiposidade/fisiologia , Animais , Diferenciação Celular/genética , Humanos , Camundongos , Obesidade/metabolismo , Células-Tronco/metabolismo , Células-Tronco/fisiologia
11.
Methods Mol Biol ; 1467: 73-80, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27417960

RESUMO

Adult homeostasis is dependent on normal Wt1 expression. Loss of Wt1 expression in adult mice causes rapid loss of the mesenchymal tissues, fat and bone, amongst other phenotypes. Bone and bone marrow mesenchymal stromal cells can be studied by cell isolation and expansion. The stemness of these cells can then be characterized by carrying out a colony-forming unit-fibroblast assay and observing clonogenic capabilities.


Assuntos
Células da Medula Óssea/citologia , Osso e Ossos/citologia , Separação Celular/métodos , Ensaio de Unidades Formadoras de Colônias/métodos , Animais , Contagem de Células , Células Cultivadas , Fibroblastos , Células-Tronco Mesenquimais/citologia , Camundongos , Células-Tronco/citologia
12.
Methods Mol Biol ; 1467: 81-91, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27417961

RESUMO

The current global obesity epidemic has triggered increased interest in adipose tissue biology. A major area of attention for many is adipose tissue development. A greater understanding of adipocyte ontogeny could be highly beneficial in answering questions about obesity-associated disease. Recent work has shown that a proportion of mature adipocytes in visceral white adipose tissue are derived from Wt1-expressing adipocyte precursor cells. These adipocyte precursor cells reside within the adipose tissue itself, and are a constituent of the stromal vascular fraction (SVF), along with other, non-adipogenic, cell types. Crucially, heterogeneity exists within the adipocyte precursor population, with only a proportion of cells expressing Wt1. Moreover, it appears that this difference in the precursor cells may influence the mature adipocytes, with Wt1-lineage-positive adipocytes having fewer, larger lipid droplets than the Wt1-lineage negative. Using fluorescence-activated cell sorting, based on specific marker profiles, it is possible to isolate the adipocyte precursor cells from the SVF. Subsequently, this population can be divided into Wt1-expressing and non-expressing fractions, therefore permitting further analysis of the two cell populations, and the mature adipocytes derived from them. In this chapter we outline a method by which adipocyte precursor cells can be isolated, and how, using a specific mouse model, Wt1-expressing and non-expressing cells can be separated.


Assuntos
Adipócitos/citologia , Separação Celular/métodos , Citometria de Fluxo/métodos , Proteínas Repressoras/metabolismo , Adipócitos/metabolismo , Tecido Adiposo Branco/citologia , Animais , Diferenciação Celular , Proliferação de Células , Células Cultivadas , Camundongos , Proteínas WT1
13.
PLoS One ; 11(2): e0148105, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-26828722

RESUMO

Gene function in stem cell maintenance is often tested by inducing deletion via the Cre-loxP system. However, controls for Cre and other variables are frequently not included. Here we show that when cultured in the presence of 4-OH tamoxifen, bone and marrow cells containing the CreERT2 construct have a reduced colony forming ability. Inactive CreERT2 recombinase, however, has the opposite effect. Young female marrow cells containing the inactive CreERT2 construct grew more colonies than cells lacking the construct altogether. Young female control marrow cells (i.e., negative for CreERT2) also produced significantly greater colony numbers when cultured with 4-OH tamoxifen, compared with the ethanol vehicle control. In conclusion, we report that the use of the Cre-loxP system is inadvisable in combination with CFU-F assays, and that appropriate controls should be in place to extend the future use of Cre-loxP in alternate assays.


Assuntos
Integrases/metabolismo , Caracteres Sexuais , Tamoxifeno/farmacologia , Animais , Células da Medula Óssea/efeitos dos fármacos , Células da Medula Óssea/metabolismo , Diferenciação Celular/efeitos dos fármacos , Células Cultivadas , Ensaio de Unidades Formadoras de Colônias , Feminino , Masculino , Camundongos
14.
Proc Natl Acad Sci U S A ; 113(3): 656-61, 2016 Jan 19.
Artigo em Inglês | MEDLINE | ID: mdl-26739565

RESUMO

Recent reports suggest that mammalian embryonic coronary endothelium (CoE) originates from the sinus venosus and ventricular endocardium. However, the contribution of extracardiac cells to CoE is thought to be minor and nonsignificant for coronary formation. Using classic (Wt1(Cre)) and previously undescribed (G2-Gata4(Cre)) transgenic mouse models for the study of coronary vascular development, we show that extracardiac septum transversum/proepicardium (ST/PE)-derived endothelial cells are required for the formation of ventricular coronary arterio-venous vascular connections. Our results indicate that at least 20% of embryonic coronary arterial and capillary endothelial cells derive from the ST/PE compartment. Moreover, we show that conditional deletion of the ST/PE lineage-specific Wilms' tumor suppressor gene (Wt1) in the ST/PE of G2-Gata4(Cre) mice and in the endothelium of Tie2(Cre) mice disrupts embryonic coronary transmural patterning, leading to embryonic death. Taken together, our results demonstrate that ST/PE-derived endothelial cells contribute significantly to and are required for proper coronary vascular morphogenesis.


Assuntos
Vasos Coronários/embriologia , Embrião de Mamíferos/citologia , Células Endoteliais/citologia , Septos Cardíacos/citologia , Pericárdio/citologia , Animais , Biomarcadores/metabolismo , Linhagem da Célula , Vasos Coronários/citologia , Desenvolvimento Embrionário , Elementos Facilitadores Genéticos/genética , Transição Epitelial-Mesenquimal , Fator de Transcrição GATA4/metabolismo , Deleção de Genes , Genes Reporter , Proteínas de Fluorescência Verde/metabolismo , Integrases/metabolismo , Camundongos , Modelos Biológicos , Miócitos Cardíacos/citologia , Miócitos Cardíacos/metabolismo , Fenótipo , Proteínas WT1/metabolismo
15.
Adipocyte ; 4(3): 217-21, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26257994

RESUMO

One major gap in adipocyte biology has been a lack of understanding of the developmental origins of the different visceral white adipose tissue (WAT) depots and subcutaneous WAT. In a recent study we showed that most visceral WAT but no subcutaneous WAT arises from cells expressing the Wilms' tumor 1 (Wt1) gene late in mouse gestation.(1) Wt1 continues to be expressed in visceral WAT progenitors into adult life. We also showed that visceral WAT is lined by a mesothelium and provided evidence that this structure is the source of adipocytes. Our study also adds to the growing body of evidence that there is heterogeneity in the visceral progenitors, such that there are Wt1-expressing and non-expressing subsets, the relative proportions of which vary between depots. This raises the enticing prospect that the adipocytes arising from these progenitor subsets may have different properties and our preliminary data support this notion. Finally, evidence from our study and one from Spiegelman's group(2) suggests that Wt1 is not just a marker but regulates visceral WAT identity and the progenitor population. We discuss the implications of this work and some of the questions and future directions that arise from it.

16.
Hum Mol Genet ; 24(19): 5464-74, 2015 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-26173456

RESUMO

Homozygous loss of function (HLOF) variants provide a valuable window on gene function in humans, as well as an inventory of the human genes that are not essential for survival and reproduction. All humans carry at least a few HLOF variants, but the exact number of inactivated genes that can be tolerated is currently unknown­as are the phenotypic effects of losing function for most human genes. Here, we make use of 1432 whole exome sequences from five European populations to expand the catalogue of known human HLOF mutations; after stringent filtering of variants in our dataset, we identify a total of 173 HLOF mutations, 76 (44%) of which have not been observed previously. We find that population isolates are particularly well suited to surveys of novel HLOF genes because individuals in such populations carry extensive runs of homozygosity, which we show are enriched for novel, rare HLOF variants. Further, we make use of extensive phenotypic data to show that most HLOFs, ascertained in population-based samples, appear to have little detectable effect on the phenotype. On the contrary, we document several genes directly implicated in disease that seem to tolerate HLOF variants. Overall HLOF genes are enriched for olfactory receptor function and are expressed in testes more often than expected, consistent with reduced purifying selection and incipient pseudogenisation.


Assuntos
Mutação , População Branca/genética , Exoma , Frequência do Gene , Homozigoto , Humanos , Fenótipo , Seleção Genética
17.
Intravital ; 4(1): e1055430, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-28243514

RESUMO

The use of confocal and multi-photon microscopy for intra-vital cancer imaging has impacted on our understanding of cancer cell behavior and interaction with the surrounding tumor microenvironment in vivo. However, many studies to-date rely on the use fluorescent dyes or genetically encoded probes that enable visualization of a structure or cell population of interest, but do not illuminate the complexity of the surrounding tumor microenvironment. Here, we show that multi-modal microscopy combining 2-photon fluorescence with CARS can begin to address this deficit, enabling detailed imaging of the tumor niche without the need for additional labeling. This can be performed on live tumor-bearing animals through optical observation windows, permitting real-time and longitudinal imaging of dynamic processes within the tumor niche.

18.
Nat Cell Biol ; 16(4): 367-75, 2014 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-24609269

RESUMO

Fuelled by the obesity epidemic, there is considerable interest in the developmental origins of white adipose tissue (WAT) and the stem and progenitor cells from which it arises. Whereas increased visceral fat mass is associated with metabolic dysfunction, increased subcutaneous WAT is protective. There are six visceral fat depots: perirenal, gonadal, epicardial, retroperitoneal, omental and mesenteric, and it is a subject of much debate whether these have a common developmental origin and whether this differs from that for subcutaneous WAT. Here we show that all six visceral WAT depots receive a significant contribution from cells expressing Wt1 late in gestation. Conversely, no subcutaneous WAT or brown adipose tissue arises from Wt1-expressing cells. Postnatally, a subset of visceral WAT continues to arise from Wt1-expressing cells, consistent with the finding that Wt1 marks a proportion of cell populations enriched in WAT progenitors. We show that all visceral fat depots have a mesothelial layer like the visceral organs with which they are associated, and provide several lines of evidence that Wt1-expressing mesothelium can produce adipocytes. These results reveal a major ontogenetic difference between visceral and subcutaneous WAT, and pinpoint the lateral plate mesoderm as a major source of visceral WAT. They also support the notion that visceral WAT progenitors are heterogeneous, and suggest that mesothelium is a source of adipocytes.


Assuntos
Adipócitos/metabolismo , Tecido Adiposo Marrom/metabolismo , Tecido Adiposo Branco/metabolismo , Proteínas WT1/metabolismo , Adipócitos/citologia , Tecido Adiposo Marrom/citologia , Tecido Adiposo Marrom/embriologia , Tecido Adiposo Branco/citologia , Tecido Adiposo Branco/embriologia , Animais , Antineoplásicos Hormonais/farmacologia , Linhagem da Célula/genética , Técnicas de Introdução de Genes , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Mesoderma/citologia , Mesoderma/metabolismo , Camundongos , Tamoxifeno/farmacologia , Proteínas WT1/genética
19.
Hum Mol Genet ; 22(25): 5083-95, 2013 Dec 20.
Artigo em Inglês | MEDLINE | ID: mdl-23900076

RESUMO

The embryonic epicardium is an important source of cardiovascular precursor cells and paracrine factors that are required for adequate heart formation. Signaling pathways regulated by WT1 that promote heart development have started to be described; however, there is little information on signaling pathways regulated by WT1 that could act in a negative manner. Transcriptome analysis of Wt1KO epicardial cells reveals an unexpected role for WT1 in repressing the expression of interferon-regulated genes that could be involved in a negative regulation of heart morphogenesis. Here, we showed that WT1 is required to repress the expression of the chemokines Ccl5 and Cxcl10 in epicardial cells. We observed an inverse correlation of Wt1 and the expression of Cxcl10 and Ccl5 during epicardium development. Chemokine receptor analyses of hearts from Wt1(gfp/+) mice demonstrate the differential expression of their chemokine receptors in GFP(+) epicardial enriched cells and GFP(-) cells. Functional assays demonstrate that CXCL10 and CCL5 inhibit epicardial cells migration and the proliferation of cardiomyocytes respectively. WT1 regulates the expression levels of Cxcl10 and Ccl5 in epicardial cells directly and indirectly through increasing the levels of IRF7. As epicardial cell reactivation after a myocardial damage is linked with WT1 expression, the present work has potential implications in adult heart repair.


Assuntos
Quimiocina CCL5/biossíntese , Quimiocina CXCL10/biossíntese , Coração/crescimento & desenvolvimento , Pericárdio/crescimento & desenvolvimento , Proteínas WT1/genética , Animais , Quimiocina CCL5/genética , Quimiocina CXCL10/genética , Perfilação da Expressão Gênica , Regulação da Expressão Gênica no Desenvolvimento , Técnicas de Inativação de Genes , Fator Regulador 7 de Interferon/metabolismo , Camundongos , Miocárdio/metabolismo , Miócitos Cardíacos/metabolismo , Pericárdio/citologia , Receptores de Quimiocinas/antagonistas & inibidores , Receptores de Quimiocinas/metabolismo , Transdução de Sinais , Proteínas WT1/biossíntese
20.
Trends Genet ; 28(10): 515-24, 2012 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-22658804

RESUMO

From both the fundamental and clinical perspectives, there is growing interest in mesenchymal cells and the mechanisms that regulate the two-way switch between mesenchymal and epithelial states. Here, we review recent findings showing that the Wilms' tumor gene (Wt1) is a key regulator of mesenchyme maintenance and the mesenchyme to epithelial balance in the development of certain mesodermal organs. We summarize recent experiments demonstrating, unexpectedly, that Wt1 is also essential for the integrity or function of multiple adult tissues, mainly, we argue, through regulating mesenchymal cells. We also discuss growing evidence that implicates Wt1 in tissue repair and regeneration. Drawing on these findings, we highlight the similarities between Wt1-expressing cells in different tissues. We believe that future studies aimed at elucidating the mechanisms underlying the functions of Wt1 in adult cells will reveal key cell types, pathways, and molecules regulating adult tissue homeostasis and repair.


Assuntos
Homeostase , Mesoderma/metabolismo , Neoplasias/metabolismo , Proteínas WT1/metabolismo , Animais , Regulação da Expressão Gênica no Desenvolvimento , Regulação Neoplásica da Expressão Gênica , Humanos , Proteínas WT1/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...