Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 82
Filtrar
1.
Mol Biol Rep ; 50(6): 5013-5020, 2023 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-37097539

RESUMO

BACKGROUND: Globally, congenital heart defect (CHD) is the most common congenital malformation, responsible for higher morbidity and mortality in the pediatric population. It is a complex multifactorial disease influenced by gene-environment and gene-gene interactions. The current study was the first attempt to study these polymorphisms in common clinical phenotypes of CHD in Pakistan and the association between maternal hypertension and diabetes with single nucleotide polymorphisms (SNPs) in children. METHODS: A total of 376 subjects were recruited in this current case-control study. Six variants from three genes were analyzed by cost-effective multiplex PCR and genotyped by minisequencing. Statistical analysis was done by GraphPad prism and Haploview. The association of SNPs and CHD was determined using logistic regression. RESULTS: The risk allele frequency was higher in cases as compared to healthy subjects, but the results were not significant for rs703752. However, stratification analysis suggested that rs703752 was significantly associated with the tetralogy of Fallot. The rs2295418 was significantly associated with maternal hypertension (OR = 16.41, p = 0.003), while a weak association was present between maternal diabetes and rs360057 (p = 0.08). CONCLUSION: In conclusion, variants in transcriptional and signaling genes were associated with Pakistani pediatric CHD patients that showed varied susceptibility between different clinical phenotypes of CHD. In addition, this study was the first report regarding the significant association between maternal hypertension and the LEFTY2 gene variant.


Assuntos
Diabetes Mellitus , Cardiopatias Congênitas , Hipertensão , Criança , Humanos , Paquistão , Estudos de Casos e Controles , Proteína Homeobox Nkx-2.5/genética , Cardiopatias Congênitas/genética , Diabetes Mellitus/genética , Hipertensão/genética , Polimorfismo de Nucleotídeo Único/genética , Predisposição Genética para Doença , Fatores de Determinação Direita-Esquerda/genética
2.
Nat Commun ; 13(1): 6101, 2022 10 15.
Artigo em Inglês | MEDLINE | ID: mdl-36243734

RESUMO

The hindered diffusion model postulates that the movement of a signaling molecule through an embryo is affected by tissue geometry and binding-mediated hindrance, but these effects have not been directly demonstrated in vivo. Here, we visualize extracellular movement and binding of individual molecules of the activator-inhibitor signaling pair Nodal and Lefty in live developing zebrafish embryos using reflected light-sheet microscopy. We observe that diffusion coefficients of molecules are high in extracellular cavities, whereas mobility is reduced and bound fractions are high within cell-cell interfaces. Counterintuitively, molecules nevertheless accumulate in cavities, which we attribute to the geometry of the extracellular space by agent-based simulations. We further find that Nodal has a larger bound fraction than Lefty and shows a binding time of tens of seconds. Together, our measurements and simulations provide direct support for the hindered diffusion model and yield insights into the nanometer-to-micrometer-scale mechanisms that lead to macroscopic signal dispersal.


Assuntos
Proteína Nodal , Peixe-Zebra , Animais , Difusão , Regulação da Expressão Gênica no Desenvolvimento , Fatores de Determinação Direita-Esquerda/genética , Proteína Nodal/metabolismo , Fator de Crescimento Transformador beta/metabolismo , Peixe-Zebra/genética , Proteínas de Peixe-Zebra/genética
3.
Nat Commun ; 13(1): 497, 2022 01 25.
Artigo em Inglês | MEDLINE | ID: mdl-35079017

RESUMO

Morphogens are signaling molecules that convey positional information and dictate cell fates during development. Although ectopic expression in model organisms suggests that morphogen gradients form through diffusion, little is known about how morphogen gradients are created and interpreted during mammalian embryogenesis due to the combined difficulties of measuring endogenous morphogen levels and observing development in utero. Here we take advantage of a human gastruloid model to visualize endogenous Nodal protein in living cells, during specification of germ layers. We show that Nodal is extremely short range so that Nodal protein is limited to the immediate neighborhood of source cells. Nodal activity spreads through a relay mechanism in which Nodal production induces neighboring cells to transcribe Nodal. We further show that the Nodal inhibitor Lefty, while biochemically capable of long-range diffusion, also acts locally to control the timing of Nodal spread and therefore of mesoderm differentiation during patterning. Our study establishes a paradigm for tissue patterning by an activator-inhibitor pair.


Assuntos
Blastocisto/metabolismo , Gástrula/metabolismo , Gastrulação/genética , Células-Tronco Embrionárias Humanas/metabolismo , Proteína Nodal/genética , Blastocisto/citologia , Linhagem Celular , Difusão , Imunofluorescência/métodos , Gástrula/citologia , Regulação da Expressão Gênica no Desenvolvimento , Técnicas de Inativação de Genes , Células-Tronco Embrionárias Humanas/citologia , Humanos , Hibridização in Situ Fluorescente/métodos , Fatores de Determinação Direita-Esquerda/genética , Fatores de Determinação Direita-Esquerda/metabolismo , Microscopia Confocal/métodos , Microscopia de Fluorescência/métodos , Proteína Nodal/metabolismo
4.
Genes (Basel) ; 12(10)2021 09 23.
Artigo em Inglês | MEDLINE | ID: mdl-34680872

RESUMO

Ulcerative colitis (UC) and Crohn's disease (CD) are characterized by an imbalance between pro-inflammatory and anti-inflammatory cytokines, interfering with the resolution of inflammation. Due to the crucial role of cytokines, new insights into their profiles in UC and CD would help to improve our understanding of pathogenesis and enable the development of new treatment modalities. We provide an expression profile of cytokines in UC and CD, using bioinformatics approach, and experimental validation of expression of the selected genes. We retrieved data and analyzed the cytokine gene expression profiles of UC and CD. From ten genes with inverse expression, common to CD and UC, BMP8B, LEFTY1 and INSL5 were selected for gene expression experimental validation. Experimentally, BMP8B and INSL5 were down-regulated in both CD and UC but followed the bioinformatics trend. The expression of genes LEFTY1 and BMP8B was statistically significant when comparing UC and CD in colon and the expression of gene LEFTY1 showed statistical significance when CD in ileum and colon were compared. Using the bioinformatics approach and experimental validation, we found differences in expression profiles between UC and CD for INSL5, LEFTY1 and BMP8B. These three promising candidate genes need to be further explored at different levels, such as DNA methylation and protein expression, to provide more evidence on their potential diagnostic role in CD and UC.


Assuntos
Proteínas Morfogenéticas Ósseas/genética , Colite Ulcerativa/genética , Doença de Crohn/genética , Insulina/genética , Fatores de Determinação Direita-Esquerda/genética , Proteínas/genética , Adulto , Biomarcadores/metabolismo , Colite Ulcerativa/diagnóstico , Colite Ulcerativa/patologia , Doença de Crohn/diagnóstico , Doença de Crohn/patologia , Diagnóstico Diferencial , Feminino , Perfilação da Expressão Gênica , Regulação da Expressão Gênica/genética , Humanos , Masculino , Análise em Microsséries , Pessoa de Meia-Idade
5.
J Cardiovasc Transl Res ; 14(4): 636-646, 2021 08.
Artigo em Inglês | MEDLINE | ID: mdl-33409963

RESUMO

Transforming growth factor-ß1 signaling pathways are known to involve in the development of post-infarction fibrosis, a process characterized by the aberrant activation, proliferation, and differentiation of fibroblasts, as well as the unbalanced turnover of extracellular matrix proteins. Recent studies have shown that Lefty1, a novel member of TGF-ß superfamily, acts as a brake on the TGF-ß signaling pathway in non-cardiac tissues. However, its role in myocardial infarction (MI)-induced fibrosis and left ventricular remodeling has not been fully elucidated. Here, for the first time, we reported that Lefty1 alleviated post-MI fibroblast proliferation, differentiation, and secretion through suppressing p-Smad2 and p-ERK1/2 signaling pathways in vivo and in vitro. In MI mice or TGF-ß1-treated neonatal rat cardiac fibroblasts (CFBs), the expression of Lefty1 was upregulated. Adenovirus-mediated overexpression of Lefty1 significantly attenuated TGF-ß1-induced CFBs' proliferation, differentiation, and collagen production. Using the adeno-associated virus approach, we confirmed that Lefty1 attenuates MI-induced cardiac injury, as evidenced by the decreased infarct size and preserved cardiac function. These results highlight the importance of Lefty1 in the prevention of post-MI fibrosis and may help identify potential targets for therapeutic intervention of cardiac fibrosis. Graphical abstract.


Assuntos
Fatores de Determinação Direita-Esquerda/metabolismo , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , Infarto do Miocárdio/metabolismo , Miócitos Cardíacos/metabolismo , Proteína Smad2/metabolismo , Animais , Diferenciação Celular , Proliferação de Células , Células Cultivadas , Colágeno/genética , Colágeno/metabolismo , Dependovirus/genética , Modelos Animais de Doenças , Fibrose , Vetores Genéticos , Fatores de Determinação Direita-Esquerda/genética , Masculino , Camundongos Endogâmicos C57BL , Infarto do Miocárdio/genética , Infarto do Miocárdio/patologia , Infarto do Miocárdio/fisiopatologia , Miócitos Cardíacos/efeitos dos fármacos , Miócitos Cardíacos/patologia , Fosforilação , Ratos Sprague-Dawley , Transdução de Sinais , Fator de Crescimento Transformador beta1/farmacologia , Função Ventricular Esquerda
6.
Semin Cell Dev Biol ; 110: 11-18, 2021 02.
Artigo em Inglês | MEDLINE | ID: mdl-32571625

RESUMO

The initial breaking of left-right (L-R) symmetry in the embryo is controlled by a motile-cilia-driven leftward fluid flow in the left-right organiser (LRO), resulting in L-R asymmetric gene expression flanking the LRO. Ultimately this results in left- but not right-sided activation of the Nodal-Pitx2 pathway in more lateral tissues. While aspects of the initial breaking event clearly vary between vertebrates, events in the Lateral Plate Mesoderm (LPM) are conserved through the vertebrate lineage. Evidence from model systems and humans highlights the role of cilia both in the initial symmetry breaking and in the ability of more lateral tissues to exhibit asymmetric gene expression. In this review we concentrate on the process of L-R determination in mouse and humans.


Assuntos
Padronização Corporal/genética , Cílios/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Mecanotransdução Celular/genética , Mesoderma/metabolismo , Animais , Cílios/ultraestrutura , Embrião de Mamíferos , Retroalimentação Fisiológica , Proteínas de Homeodomínio/genética , Proteínas de Homeodomínio/metabolismo , Humanos , Fatores de Determinação Direita-Esquerda/genética , Fatores de Determinação Direita-Esquerda/metabolismo , Mesoderma/crescimento & desenvolvimento , Mesoderma/ultraestrutura , Camundongos , Canais de Cátion TRPP/genética , Canais de Cátion TRPP/metabolismo , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Proteína Wnt3/genética , Proteína Wnt3/metabolismo , Proteína Homeobox PITX2
7.
Dev Biol ; 470: 21-36, 2021 02.
Artigo em Inglês | MEDLINE | ID: mdl-33197427

RESUMO

Nodal signaling is essential for mesoderm and endoderm formation, as well as neural plate induction and establishment of left-right asymmetry. However, the mechanisms controlling expression of Nodal pathway genes in these contexts are not fully known. Previously, we showed that Cdx1b induces expression of downstream Nodal signaling factors during early endoderm formation. In this study, we show that Cdx1b also regulates epithalamic asymmetry in zebrafish embryos by modulating expression of ndr2 and lft1. We first knocked down cdx1b with translation-blocking and splicing-blocking morpholinos (MOs). Most embryos injected with translation-blocking MOs showed absent ndr2, lft1 and pitx2c expression in the left dorsal diencephalon during segmentation and pharyngula stages accompanied by aberrant parapineal migration and habenular laterality at 72 â€‹h post fertilization (hpf). These defects were less frequent in embryos injected with splicing-blocking MO. To confirm the morphant phenotype, we next generated both zygotic (Z)cdx1b-/- and maternal zygotic (MZ)cdx1b-/- mutants by CRISPR-Cas9 mutagenesis. Expression of ndr2, lft1 and pitx2c was absent in the left dorsal diencephalon of a high proportion of MZcdx1b-/- mutants; however, aberrant dorsal diencephalic pitx2c expression patterns were observed at low frequency in Zcdx1b-/- mutant embryos. Correspondingly, dysregulated parapineal migration and habenular laterality were also observed in MZcdx1b-/- mutant embryos at 72 hpf. On the other hand, Kupffer's vesicle cilia length and number, expression pattern of spaw in the lateral plate mesoderm and pitx2c in the gut as well as left-right patterning of various visceral organs were not altered in MZcdx1b-/- mutants compared to wild-type embryos. Chromatin immunoprecipitation revealed that Cdx1b directly regulates ndr2 and lft1 expression. Furthermore, injection of cdx1b-vivo MO1 but not cdx1b-vivo 4 â€‹mm MO1 in the forebrain ventricle at 18 hpf significantly downregulated lft1 expression in the left dorsal diencephalon at 23-24 â€‹s stages. Together, our results suggest that Cdx1b regulates transcription of ndr2 and lft1 to maintain proper Nodal activity in the dorsal diencephalon and epithalamic asymmetry in zebrafish embryos.


Assuntos
Padronização Corporal/genética , Epitálamo/embriologia , Proteínas de Homeodomínio/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular/genética , Fatores de Determinação Direita-Esquerda/genética , Proteínas de Peixe-Zebra/genética , Proteínas de Peixe-Zebra/metabolismo , Peixe-Zebra/embriologia , Peixe-Zebra/genética , Animais , Movimento Celular , Diencéfalo/embriologia , Diencéfalo/metabolismo , Embrião não Mamífero/metabolismo , Epitálamo/metabolismo , Perfilação da Expressão Gênica , Regulação da Expressão Gênica no Desenvolvimento , Técnicas de Silenciamento de Genes , Habenula/embriologia , Coração/embriologia , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Fatores de Determinação Direita-Esquerda/metabolismo , Proteína Nodal/metabolismo , Glândula Pineal/citologia , Glândula Pineal/embriologia , Ligação Proteica , Transdução de Sinais , Peixe-Zebra/metabolismo
8.
Mol Carcinog ; 59(12): 1409-1419, 2020 12.
Artigo em Inglês | MEDLINE | ID: mdl-33111989

RESUMO

Glioblastomas (GBM) contain numerous hypoxic foci associated with a rare fraction of glioma stem cells (GSCs). Left-right determination factor (LEFTY) and Nodal, members of the transforming growth factor ß (TGF-ß) superfamily, have glycogen synthase kinase 3ß (GSK-3ß) phosphorylation motifs and are linked with stemness in human malignancies. Herein, we investigated the roles of LEFTY and Nodal in GBM hypoxic foci. In clinical samples, significantly higher expression of LEFTY, Nodal, phospho (p) GSK-3ß, pSmad2, and Nestin, as well as higher apoptotic and lower proliferation rates, were observed in nonpseudopalisading (non-Ps) perinecrotic lesions as compared to Ps and non-necrotic tumor lesions, with a positive correlation between LEFTY, Nodal, pGSK-3ß, or pSmad2 scores. In KS-1, a GBM cell line that lacks endogenous Nodal expression, treatment with the hypoxic mimetic CoCl2 increased LEFTY, pGSK-3ß, and pSmad2 levels, but decreased pAkt levels. Moreover, the promoter for LEFTY, but not Nodal, was activated by Smad2 or TGF-ß1, suggesting that overexpression of LEFTY and Nodal may be due to Akt-independent GSK-3ß inactivation, with or without cooperation of the TGF-ß1/Smad2 axis. LEFTY and Nodal overexpression increased proliferation rates and reduced susceptibility to CoCl2 -induced apoptosis, and increased the expression of epithelial-mesenchymal transition (EMT)/GSC-related markers. An increased ALDH1high population and more efficient spheroid formation was also observed in LEFTY-overexpressing cells. These findings suggest that LEFTY and Nodal may contribute to cell survival in non-Ps GBM perinecrotic lesions, leading to alterations in apoptosis, proliferation, or EMT/GCS features.


Assuntos
Neoplasias Encefálicas/metabolismo , Glioblastoma/metabolismo , Fatores de Determinação Direita-Esquerda/metabolismo , Proteína Nodal/metabolismo , Regulação para Cima , Adolescente , Adulto , Idoso , Neoplasias Encefálicas/genética , Hipóxia Celular , Linhagem Celular Tumoral , Sobrevivência Celular , Cobalto/efeitos adversos , Transição Epitelial-Mesenquimal , Feminino , Regulação Neoplásica da Expressão Gênica , Glioblastoma/genética , Humanos , Fatores de Determinação Direita-Esquerda/genética , Masculino , Pessoa de Meia-Idade , Proteína Nodal/genética , Fosforilação , Transdução de Sinais , Adulto Jovem
9.
Nat Cell Biol ; 20(9): 1032-1042, 2018 09.
Artigo em Inglês | MEDLINE | ID: mdl-30061678

RESUMO

Individuals can vary substantially in size, but the proportions of their body plans are often maintained. We generated smaller zebrafish by removing 30% of their cells at the blastula stages and found that these embryos developed into normally patterned individuals. Strikingly, the proportions of all germ layers adjusted to the new embryo size within 2 hours after cell removal. As Nodal-Lefty signalling controls germ-layer patterning, we performed a computational screen for scale-invariant models of this activator-inhibitor system. This analysis predicted that the concentration of the highly diffusive inhibitor Lefty increases in smaller embryos, leading to a decreased Nodal activity range and contracted germ-layer dimensions. In vivo studies confirmed that Lefty concentration increased in smaller embryos, and embryos with reduced Lefty levels or with diffusion-hindered Lefty failed to scale their tissue proportions. These results reveal that size-dependent inhibition of Nodal signalling allows scale-invariant patterning.


Assuntos
Blástula/metabolismo , Padronização Corporal , Fatores de Determinação Direita-Esquerda/metabolismo , Proteínas de Membrana/metabolismo , Proteínas de Peixe-Zebra/metabolismo , Animais , Animais Geneticamente Modificados , Padronização Corporal/genética , Regulação da Expressão Gênica no Desenvolvimento , Fatores de Determinação Direita-Esquerda/genética , Proteínas de Membrana/genética , Transdução de Sinais , Fatores de Tempo , Peixe-Zebra/embriologia , Peixe-Zebra/genética , Peixe-Zebra/metabolismo , Proteínas de Peixe-Zebra/genética
10.
Nat Commun ; 9(1): 1942, 2018 05 16.
Artigo em Inglês | MEDLINE | ID: mdl-29769531

RESUMO

The establishment of left-right (LR) asymmetry is fundamental to animal development, but the identification of a unifying mechanism establishing laterality across different phyla has remained elusive. A cilia-driven, directional fluid flow is important for symmetry breaking in numerous vertebrates, including zebrafish. Alternatively, LR asymmetry can be established independently of cilia, notably through the intrinsic chirality of the acto-myosin cytoskeleton. Here, we show that Myosin1D (Myo1D), a previously identified regulator of Drosophila LR asymmetry, is essential for the formation and function of the zebrafish LR organizer (LRO), Kupffer's vesicle (KV). Myo1D controls the orientation of LRO cilia and interacts functionally with the planar cell polarity (PCP) pathway component VanGogh-like2 (Vangl2), to shape a productive LRO flow. Our findings identify Myo1D as an evolutionarily conserved regulator of animal LR asymmetry, and show that functional interactions between Myo1D and PCP are central to the establishment of animal LR asymmetry.


Assuntos
Padronização Corporal/genética , Miosinas/genética , Proteínas de Peixe-Zebra/genética , Peixe-Zebra/genética , Animais , Animais Geneticamente Modificados , Polaridade Celular/genética , Cílios/genética , Cílios/metabolismo , Embrião não Mamífero/embriologia , Embrião não Mamífero/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Fatores de Determinação Direita-Esquerda/genética , Fatores de Determinação Direita-Esquerda/metabolismo , Mutação , Miosinas/metabolismo , Peixe-Zebra/embriologia , Peixe-Zebra/metabolismo , Proteínas de Peixe-Zebra/metabolismo
11.
Int J Mol Med ; 42(3): 1229-1236, 2018 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-29845221

RESUMO

Endometrial cancer (EC) is the most common gynecological tumor in developed countries with an increasing incidence. Left­right determination factor 2 (LEFTY2), a suppressor of cell proliferation and tumor growth, is a negative regulator of EC progression. The roles of LEFTY2 are emerging; however, the regulatory mechanisms of its expression have not been well understood. MicroRNA (miR)­215 as an oncogene serves an important role in tumorigenesis by regulating target genes. In the present study, it was demonstrated that overexpression of miR­215 promoted epithelial to mesenchymal transition (EMT), colony formation and DNA synthesis in EC HEC­1A cells and its expression was upregulated in EC tissues. Using online miR target prediction software, it was revealed that LEFTY2 is predicted as a target of miR­215. Using western blot analysis and immunofluorescence assays, it was demonstrated that overexpression of miR­215 markedly downregulated LEFTY2 protein expression levels in HEC­1A cells and LEFTY2 protein expression was downregulated in EC tissues, which was inversely correlated with miR­215 expression. Furthermore, the present study indicated that overexpression of LEFTY2 protein promoted mesenchymal to epithelial transition and sensitized HEC­1A cells to cisplatin treatment. In addition, it was revealed that the overexpression of LEFTY2 inhibited colony formation and DNA synthesis in HEC­1A cells. Thus, miR­215 may promote EMT and proliferation by regulating LEFTY2 in EC.


Assuntos
Neoplasias do Endométrio/metabolismo , Transição Epitelial-Mesenquimal/fisiologia , Regulação Neoplásica da Expressão Gênica/fisiologia , Fatores de Determinação Direita-Esquerda/metabolismo , MicroRNAs/metabolismo , Adulto , Idoso , Western Blotting , Proliferação de Células/genética , Proliferação de Células/fisiologia , Biologia Computacional , Neoplasias do Endométrio/genética , Transição Epitelial-Mesenquimal/genética , Feminino , Regulação Neoplásica da Expressão Gênica/genética , Humanos , Fatores de Determinação Direita-Esquerda/genética , MicroRNAs/genética , Pessoa de Meia-Idade
12.
Nucleic Acids Res ; 46(12): 6026-6040, 2018 07 06.
Artigo em Inglês | MEDLINE | ID: mdl-29733394

RESUMO

Sin3a is a core component of histone-deacetylation-activity-associated transcriptional repressor complex, playing important roles in early embryo development. Here, we reported that down-regulation of Sin3a led to the loss of embryonic stem cell (ESC) self-renewal and skewed differentiation into mesendoderm lineage. We found that Sin3a functioned as a transcriptional coactivator of the critical Nodal antagonist Lefty1 through interacting with Tet1 to de-methylate the Lefty1 promoter. Further studies showed that two amino acid residues (Phe147, Phe182) in the PAH1 domain of Sin3a are essential for Sin3a-Tet1 interaction and its activity in regulating pluripotency. Furthermore, genome-wide analyses of Sin3a, Tet1 and Pol II ChIP-seq and of 5mC MeDIP-seq revealed that Sin3a acted with Tet1 to facilitate the transcription of a set of their co-target genes. These results link Sin3a to epigenetic DNA modifications in transcriptional activation and have implications for understanding mechanisms underlying versatile functions of Sin3a in mouse ESCs.


Assuntos
Proteínas de Ligação a DNA/metabolismo , Células-Tronco Embrionárias/metabolismo , Proteínas Proto-Oncogênicas/metabolismo , Proteínas Repressoras/metabolismo , Ativação Transcricional , Animais , Diferenciação Celular , Linhagem Celular , Linhagem da Célula , Células-Tronco Embrionárias/citologia , Fatores de Determinação Direita-Esquerda/genética , Fatores de Determinação Direita-Esquerda/metabolismo , Camundongos , Proteína Nodal/metabolismo , Regiões Promotoras Genéticas , Domínios e Motivos de Interação entre Proteínas , Proteínas Repressoras/química , Proteínas Repressoras/genética , Complexo Correpressor Histona Desacetilase e Sin3
13.
Cell Death Dis ; 9(3): 302, 2018 02 21.
Artigo em Inglês | MEDLINE | ID: mdl-29467473

RESUMO

TRIM family members have been implicated in a variety of biological processes such as differentiation and development. We here found that Trim59 plays a critical role in early embryo development from blastocyst stage to gastrula. There existed delayed development and empty yolk sacs from embryonic day (E) 8.5 in Trim59-/- embryos. No viable Trim59-/- embryos were observed beyond E9.5. Trim59 deficiency affected primary germ layer formation at the beginning of gastrulation. At E6.5 and E7.5, the expression of primary germ layer formation-associated genes including Brachyury, lefty2, Cer1, Otx2, Wnt3, and BMP4 was reduced in Trim59-/- embryos. Homozygous mutant embryonic epiblasts were contracted and the mesoderm was absent. Trim59 could interact with actin- and myosin-associated proteins. Its deficiency disturbed F-actin polymerization during inner cell mass differentiation. Trim59-mediated polymerization of F-actin was via WASH K63-linked ubiquitination. Thus, Trim59 may be a critical regulator for early embryo development from blastocyst stage to gastrula through modulating F-actin assembly.


Assuntos
Proteínas de Transporte/genética , Proteínas de Transporte/metabolismo , Gástrula/embriologia , Gástrula/metabolismo , Actinas/química , Actinas/genética , Actinas/metabolismo , Animais , Blastocisto/metabolismo , Desenvolvimento Embrionário , Feminino , Proteínas Fetais/genética , Proteínas Fetais/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Peptídeos e Proteínas de Sinalização Intracelular , Fatores de Determinação Direita-Esquerda/genética , Fatores de Determinação Direita-Esquerda/metabolismo , Masculino , Mesoderma/embriologia , Mesoderma/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fatores de Transcrição Otx/genética , Fatores de Transcrição Otx/metabolismo , Polimerização , Proteínas com Domínio T/genética , Proteínas com Domínio T/metabolismo , Proteínas com Motivo Tripartido
14.
Nucleic Acids Res ; 46(1): 104-119, 2018 01 09.
Artigo em Inglês | MEDLINE | ID: mdl-29059375

RESUMO

In many organisms, transcriptional and post-transcriptional regulation of components of pathways or processes has been reported. However, to date, there are few reports of translational co-regulation of multiple components of a developmental signaling pathway. Here, we show that an RNA element which we previously identified as a dorsal localization element (DLE) in the 3'UTR of zebrafish nodal-related1/squint (ndr1/sqt) ligand mRNA, is shared by the related ligand nodal-related2/cyclops (ndr2/cyc) and the nodal inhibitors, lefty1 (lft1) and lefty2 mRNAs. We investigated the activity of the DLEs through functional assays in live zebrafish embryos. The lft1 DLE localizes fluorescently labeled RNA similarly to the ndr1/sqt DLE. Similar to the ndr1/sqt 3'UTR, the lft1 and lft2 3'UTRs are bound by the RNA-binding protein (RBP) and translational repressor, Y-box binding protein 1 (Ybx1), whereas deletions in the DLE abolish binding to Ybx1. Analysis of zebrafish ybx1 mutants shows that Ybx1 represses lefty1 translation in embryos. CRISPR/Cas9-mediated inactivation of human YBX1 also results in human NODAL translational de-repression, suggesting broader conservation of the DLE RNA element/Ybx1 RBP module in regulation of Nodal signaling. Our findings demonstrate translational co-regulation of components of a signaling pathway by an RNA element conserved in both sequence and structure and an RBP, revealing a 'translational regulon'.


Assuntos
Embrião não Mamífero/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Proteínas de Peixe-Zebra/genética , Peixe-Zebra/genética , Regiões 3' não Traduzidas/genética , Animais , Embrião não Mamífero/embriologia , Células HEK293 , Humanos , Peptídeos e Proteínas de Sinalização Intracelular/genética , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Fatores de Determinação Direita-Esquerda/genética , Fatores de Determinação Direita-Esquerda/metabolismo , Ligantes , Ligantes da Sinalização Nodal/genética , Ligantes da Sinalização Nodal/metabolismo , RNA/genética , RNA/metabolismo , Peixe-Zebra/embriologia , Peixe-Zebra/metabolismo , Proteínas de Peixe-Zebra/metabolismo
15.
Int J Mol Med ; 41(3): 1293-1304, 2018 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-29286065

RESUMO

Renal tubulointerstitial inflammation has an important role in fibrosis, which is the main pathogenetic alteration associated with chronic kidney disease (CKD). The left­right determination factor 1 (Lefty­1) gene pleiotropically and biologically regulates transforming growth factor, mitogen­activated protein kinase and other signaling pathways, and is considered to have a potential anti­inflammatory function. However, its role in renal tubulointerstitial inflammation, which is often a long­term consequence of renal fibrosis, is currently unknown. In the present study, the effects of adenovirus­mediated overexpression of Lefty­1 (Ad­Lefty­1­flag) on renal tubulointerstitial inflammation were determined using a mouse model of unilateral ureteral obstruction (UUO) and a rat renal tubular duct epithelial cell line (NRK­52E), which was treated with lipopolysaccharide (LPS). In vivo results indicated that the inflammatory response was increased in UUO mice, as evidenced by the increase in inflammatory cytokines and chemokines. Conversely, Lefty­1 significantly reversed the effects of UUO. Furthermore, the results of the in vitro study demonstrated that Lefty­1 significantly inhibited LPS­induced inflammatory marker expression in cultured NRK­52E cells via the nuclear factor (NF)­κB signaling pathway. These results suggested that Lefty­1 may ameliorate renal tubulointerstitial inflammation by suppressing NF­κB signaling. In conclusion, the findings of the present study indicated that Lefty­1 may be considered a potential novel therapeutic agent for inhibiting renal tubulointerstitial inflammation or even reversing the CKD process.


Assuntos
Anti-Inflamatórios/metabolismo , Inflamação/metabolismo , Inflamação/patologia , Túbulos Renais/metabolismo , Túbulos Renais/patologia , Fatores de Determinação Direita-Esquerda/metabolismo , NF-kappa B/metabolismo , Transdução de Sinais , Actinas/metabolismo , Adenoviridae/metabolismo , Animais , Morte Celular , Núcleo Celular/metabolismo , Colágeno/metabolismo , Fibronectinas/metabolismo , Mediadores da Inflamação/metabolismo , Fatores de Determinação Direita-Esquerda/genética , Lipopolissacarídeos , Macrófagos/metabolismo , Masculino , Camundongos Endogâmicos C57BL , Transporte Proteico , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Ratos , Proteína Smad7/metabolismo , Obstrução Ureteral/patologia
16.
Elife ; 62017 12 07.
Artigo em Inglês | MEDLINE | ID: mdl-29215332

RESUMO

Developmental signaling pathways often activate their own inhibitors. Such inhibitory feedback has been suggested to restrict the spatial and temporal extent of signaling or mitigate signaling fluctuations, but these models are difficult to rigorously test. Here, we determine whether the ability of the mesendoderm inducer Nodal to activate its inhibitor Lefty is required for development. We find that zebrafish lefty mutants exhibit excess Nodal signaling and increased specification of mesendoderm, resulting in embryonic lethality. Strikingly, development can be fully restored without feedback: Lethal patterning defects in lefty mutants can be rescued by ectopic expression of lefty far from its normal expression domain or by spatially and temporally uniform exposure to a Nodal inhibitor drug. While drug-treated mutants are less tolerant of mild perturbations to Nodal signaling levels than wild type embryos, they can develop into healthy adults. These results indicate that patterning without inhibitory feedback is functional but fragile.


Assuntos
Endoderma/embriologia , Retroalimentação , Fatores de Determinação Direita-Esquerda/metabolismo , Mesoderma/embriologia , Proteína Nodal/metabolismo , Transdução de Sinais , Proteínas de Peixe-Zebra/metabolismo , Animais , Técnicas de Inativação de Genes , Fatores de Determinação Direita-Esquerda/genética , Peixe-Zebra/embriologia , Proteínas de Peixe-Zebra/genética
17.
Cell Commun Signal ; 15(1): 56, 2017 12 21.
Artigo em Inglês | MEDLINE | ID: mdl-29268772

RESUMO

BACKGROUND: The left-right determination factor (LEFTY) is a novel member of the TGF-ß/Smad2 pathway and belongs to the premenstrual/menstrual repertoire in human endometrium, but little is known about its functional role in endometrial carcinomas (Em Cas). Herein, we focused on LEFTY expression and its association with progesterone therapy in Em Cas. METHODS: Regulation and function of LEFTY, as well as its associated molecules including Smad2, ovarian hormone receptors, GSK-3ß, and cell cycle-related factors, were assessed using clinical samples and cell lines of Em Cas. RESULTS: In clinical samples, LEFTY expression was positively correlated with estrogen receptor-α, but not progesterone receptor (PR), status, and was inversely related to phosphorylated (p) Smad2, cyclin A2, and Ki-67 levels. During progesterone therapy, expression of LEFTY, pSmad2, and pGSK-3ß showed stepwise increases, with significant correlations to morphological changes toward secretory features and decreased Ki-67 values. In Ishikawa cells, an Em Ca cell line that expresses PR, progesterone treatment reduced proliferation and induced increased expression of LEFTY and pGSK-3ß, although LEFTY promoter regions were inhibited by transfection of PR. Moreover, inhibition of GSK-3ß resulted in increased LEFTY expression through a decrease in its ubiquitinated form, suggesting posttranslational regulation of LEFTY protein via GSK-3ß suppression in response to progesterone. In addition, overexpression or knockdown of LEFTY led to suppression or enhancement of Smad2-dependent cyclin A2 expression, respectively. CONCLUSION: Upregulation of LEFTY may serve as a useful clinical marker for the therapeutic effects of progesterone for Em Cas, leading to inhibition of tumor cell proliferation through alteration in Smad2-dependent transcription of cyclin A2.


Assuntos
Neoplasias do Endométrio/tratamento farmacológico , Neoplasias do Endométrio/metabolismo , Fatores de Determinação Direita-Esquerda/metabolismo , Progesterona/metabolismo , Adulto , Ciclo Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Neoplasias do Endométrio/genética , Neoplasias do Endométrio/patologia , Endométrio/efeitos dos fármacos , Endométrio/metabolismo , Endométrio/patologia , Receptor alfa de Estrogênio/metabolismo , Feminino , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Humanos , Fatores de Determinação Direita-Esquerda/genética , Receptores de Progesterona/metabolismo , Transdução de Sinais/efeitos dos fármacos , Proteína Smad2/metabolismo , Adulto Jovem
18.
Nat Commun ; 8(1): 1492, 2017 11 14.
Artigo em Inglês | MEDLINE | ID: mdl-29138408

RESUMO

Anterior-posterior (A-P) polarity of mouse embryos is established by distal visceral endoderm (DVE) at embryonic day (E) 5.5. Lefty1 is expressed first at E3.5 in a subset of epiblast progenitor cells (L1epi cells) and then in a subset of primitive endoderm cells (L1dve cells) fated to become DVE. Here we studied how prospective DVE cells are selected. Lefty1 expression in L1epi and L1dve cells depends on Nodal signaling. A cell that experiences the highest level of Nodal signaling begins to express Lefty1 and becomes an L1epi cell. Deletion of Lefty1 alone or together with Lefty2 increased the number of prospective DVE cells. Ablation of L1epi or L1dve cells triggered Lefty1 expression in a subset of remaining cells. Our results suggest that selection of prospective DVE cells is both random and regulated, and that a fixed prepattern for the A-P axis does not exist before the blastocyst stage.


Assuntos
Embrião de Mamíferos/metabolismo , Endoderma/metabolismo , Fatores de Determinação Direita-Esquerda/genética , Proteína Nodal/metabolismo , Transdução de Sinais , Vísceras/metabolismo , Animais , Padronização Corporal , Cromossomos Artificiais Bacterianos , Embrião de Mamíferos/citologia , Regulação da Expressão Gênica no Desenvolvimento , Redes Reguladoras de Genes , Camundongos , Camundongos Transgênicos , Processos Estocásticos
19.
Endocrinology ; 158(11): 4076-4092, 2017 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-28938408

RESUMO

Successful pregnancy relies on dynamic control of cell signaling to achieve uterine receptivity and the necessary biological changes required for endometrial decidualization, embryo implantation, and fetal development. Glucocorticoids are master regulators of intracellular signaling and can directly regulate embryo implantation and endometrial remodeling during murine pregnancy. In immortalized human uterine cells, we have shown that glucocorticoids and estradiol (E2) coregulate thousands of genes. Recently, glucocorticoids and E2 were shown to coregulate the expression of Left-right determination factor 1 (LEFTY1), previously implicated in the regulation of decidualization. To elucidate the molecular mechanism by which glucocorticoids and E2 regulate the expression of LEFTY1, immortalized and primary human endometrial cells were evaluated for gene expression and receptor recruitment to regulatory regions of the LEFTY1 gene. Glucocorticoid administration induced expression of LEFTY1 messenger RNA and protein and recruitment of the glucocorticoid receptor (GR) and activated polymerase 2 to the promoter of LEFTY1. Glucocorticoid-mediated recruitment of GR was dependent on pioneer factors FOXA1 and FOXA2. E2 was found to antagonize glucocorticoid-mediated induction of LEFTY1 by reducing recruitment of GR, FOXA1, FOXA2, and activated polymerase 2 to the LEFTY1 promoter. Gene expression analysis identified several genes whose glucocorticoid-dependent induction required FOXA1 and FOXA2 in endometrial cells. These results suggest a molecular mechanism by which E2 antagonizes GR-dependent induction of specific genes by preventing the recruitment of the pioneer factors FOXA1 and FOXA2 in a physiologically relevant model.


Assuntos
Endométrio/efeitos dos fármacos , Endométrio/metabolismo , Glucocorticoides/farmacologia , Fator 3-alfa Nuclear de Hepatócito/fisiologia , Fator 3-beta Nuclear de Hepatócito/fisiologia , Receptores de Glucocorticoides/fisiologia , Células Cultivadas , Dexametasona/farmacologia , Implantação do Embrião/efeitos dos fármacos , Implantação do Embrião/genética , Estradiol/farmacologia , Feminino , Regulação da Expressão Gênica/efeitos dos fármacos , Fator 3-alfa Nuclear de Hepatócito/metabolismo , Fator 3-beta Nuclear de Hepatócito/metabolismo , Humanos , Fatores de Determinação Direita-Esquerda/genética , Regiões Promotoras Genéticas/efeitos dos fármacos , Receptores de Glucocorticoides/metabolismo , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/genética
20.
Mol Cancer Ther ; 16(5): 787-792, 2017 05.
Artigo em Inglês | MEDLINE | ID: mdl-28468864

RESUMO

The tumor microenvironment is a vital feature of oncogenesis and tumor progression. There are several parallels between cancer cells and early developmental stem cells, including their plasticity and signaling mechanisms. In early fetal development, Nodal is expressed for endodermal and mesodermal differentiation. This expression has been shown reemerge in the setting of epithelial cancers, such as breast and melanoma. High Nodal expression correlates to an aggressive tumor grade in these malignancies. Nodal signal begins with its interaction with its coreceptor, Cripto-1, leading to activation of Smad2/Smad3 and ultimately downstream transcription and translation. Lefty is the natural inhibitor of Nodal and controls Nodal signaling during fetal development. However, cancer cells lack the presence of Lefty, thus leading to uncontrolled tumor growth. Given this understanding, inhibition of the Nodal pathway offers a new novel therapeutic target in oncology. Mol Cancer Ther; 16(5); 787-92. ©2017 AACR.


Assuntos
Carcinogênese/genética , Terapia de Alvo Molecular , Neoplasias/genética , Proteína Nodal/genética , Diferenciação Celular/genética , Proteínas Ligadas por GPI/genética , Humanos , Peptídeos e Proteínas de Sinalização Intercelular/genética , Fatores de Determinação Direita-Esquerda/genética , Proteínas de Neoplasias/genética , Neoplasias/tratamento farmacológico , Transdução de Sinais , Proteína Smad2/genética , Proteína Smad3/genética , Microambiente Tumoral/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA