Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
1.
Acta Neuropathol Commun ; 7(1): 137, 2019 08 26.
Artigo em Inglês | MEDLINE | ID: mdl-31451106

RESUMO

Retinoblastoma is the most common intraocular malignancy in children. We previously found that the ACVR1C/SMAD2 pathway is significantly upregulated in invasive retinoblastoma samples from patients. Here we studied the role of an ACVR1C ligand, Nodal, in regulating growth and metastatic dissemination in retinoblastoma. Inhibition of Nodal using multiple short hairpin (shRNAs) in WERI Rb1 and Y79 retinoblastoma cell cultures reduced growth by more than 90%, as determined by CCK-8 growth assay. Proliferation was also significantly inhibited, as found by Ki67 assay. These effects were paralleled by inhibition in the phosphorylation of the downstream effector SMAD2, as well as induction of apoptosis, as we observed more than three-fold increase in the percentage of cells positive for cleaved-caspase-3 or expressing cleaved-PARP1. Importantly, we found that downregulation of Nodal potently suppressed invasion in vitro, by 50 to 80%, as determined by transwell invasion assay (p = 0.02). Using an orthotopic model of retinoblastoma in zebrafish, we found 34% reduction in the ability of the cells to disseminate outside the eye, when Nodal was knocked down by shRNA (p = 0.0003). These data suggest that Nodal plays an important role in promoting growth, proliferation and invasion in retinoblastoma, and can be considered a new therapeutic target for both primary tumor growth and metastatic progression.


Assuntos
Progressão da Doença , Regulação para Baixo/fisiologia , Proteína Nodal/biossíntese , Neoplasias da Retina/metabolismo , Retinoblastoma/metabolismo , Animais , Proliferação de Células/fisiologia , Humanos , Camundongos , Invasividade Neoplásica/genética , Invasividade Neoplásica/patologia , Proteína Nodal/genética , Neoplasias da Retina/genética , Neoplasias da Retina/patologia , Retinoblastoma/genética , Retinoblastoma/patologia , Ensaios Antitumorais Modelo de Xenoenxerto/métodos , Peixe-Zebra
2.
Mol Reprod Dev ; 86(11): 1652-1662, 2019 11.
Artigo em Inglês | MEDLINE | ID: mdl-31418961

RESUMO

To gain further insight in the mechanisms of the embryo-maternal dialog in the oviduct, expression of members of the transforming growth factor-ß superfamily, NODAL, its inhibitor, LEFTY2, and their coreceptor, CFC1, were studied in the oviduct of 3-day post copula (3 dpc) females with and without embryos (E and NE), pseudopregnant rats (SP3), and in 3-day embryos. Nodal transcripts in SP3 oviducts showed a steady-state relative abundance when compared with proestrus stage and the 3 dpc. In contrast, Lefty2 and Cfc1 relative abundance levels in proestrus and 3 dpc were higher. When comparing E with NE oviducts, Nodal and Lefty2 expression levels decreased, while Cfc1 expression increased in the presence of embryos. Nodal messenger RNA (mRNA) was observed in the embryo, but Lefty2 and Cfc1 transcripts were not found. In addition, an increase in Lefty2 expression coincided with increased levels of matrix metalloproteinases 9 mRNA and protein in the oviduct and in the oviductal fluid, respectively. These observations have shed new light on the relevance of the NODAL/LEFTY2 pathway in the oviduct during early embryo development and the role of the embryo in modulating this pathway.


Assuntos
Tubas Uterinas/metabolismo , Regulação da Expressão Gênica/fisiologia , Fatores de Determinação Direita-Esquerda/biossíntese , Proteína Nodal/biossíntese , Gravidez/fisiologia , Transdução de Sinais/fisiologia , Animais , Embrião de Mamíferos , Feminino , Ratos , Ratos Wistar
3.
Development ; 144(24): 4694-4703, 2017 12 15.
Artigo em Inglês | MEDLINE | ID: mdl-29122841

RESUMO

Correct patterning of left-right (LR) asymmetry is essential during the embryonic development of bilaterians. Hedgehog (Hh) signaling is known to play a role in LR asymmetry development of mouse, chicken and sea urchin embryos by regulating Nodal expression. In this study, we report a novel regulatory mechanism for Hh in LR asymmetry development of amphioxus embryos. Our results revealed that Hh-/- embryos abolish Cerberus (Cer) transcription, with bilaterally symmetric expression of Nodal, Lefty and Pitx In consequence, Hh-/- mutants duplicated left-side structures and lost right-side characters, displaying an abnormal bilaterally symmetric body plan. These LR defects in morphology and gene expression could be rescued by Hh mRNA injection. Our results indicate that Hh participates in amphioxus LR patterning by controlling Cer gene expression. Curiously, however, upregulation of Hh signaling failed to alter the Cer expression pattern or LR morphology in amphioxus embryos, indicating that Hh might not provide an asymmetric cue for Cer expression. In addition, Hh is required for mouth opening in amphioxus, hinting at a homologous relationship between amphioxus and vertebrate mouth development.


Assuntos
Padronização Corporal/genética , Regulação da Expressão Gênica no Desenvolvimento , Proteínas Hedgehog/genética , Anfioxos/embriologia , Boca/embriologia , Animais , Animais Geneticamente Modificados/embriologia , Técnicas de Inativação de Genes , Proteínas Hedgehog/metabolismo , Peptídeos e Proteínas de Sinalização Intercelular/biossíntese , Fatores de Determinação Direita-Esquerda/biossíntese , Proteína Nodal/biossíntese , Fatores de Transcrição Box Pareados/biossíntese , Transdução de Sinais , Transcrição Gênica/genética
4.
PLoS Genet ; 12(6): e1006070, 2016 06.
Artigo em Inglês | MEDLINE | ID: mdl-27272319

RESUMO

During mammalian development, left-right (L-R) asymmetry is established by a cilia-driven leftward fluid flow within a midline embryonic cavity called the node. This 'nodal flow' is detected by peripherally-located crown cells that each assemble a primary cilium which contain the putative Ca2+ channel PKD2. The interaction of flow and crown cell cilia promotes left side-specific expression of Nodal in the lateral plate mesoderm (LPM). Whilst the PKD2-interacting protein PKD1L1 has also been implicated in L-R patterning, the underlying mechanism by which flow is detected and the genetic relationship between Polycystin function and asymmetric gene expression remains unknown. Here, we characterize a Pkd1l1 mutant line in which Nodal is activated bilaterally, suggesting that PKD1L1 is not required for LPM Nodal pathway activation per se, but rather to restrict Nodal to the left side downstream of nodal flow. Epistasis analysis shows that Pkd1l1 acts as an upstream genetic repressor of Pkd2. This study therefore provides a genetic pathway for the early stages of L-R determination. Moreover, using a system in which cultured cells are supplied artificial flow, we demonstrate that PKD1L1 is sufficient to mediate a Ca2+ signaling response after flow stimulation. Finally, we show that an extracellular PKD domain within PKD1L1 is crucial for PKD1L1 function; as such, destabilizing the domain causes L-R defects in the mouse. Our demonstration that PKD1L1 protein can mediate a response to flow coheres with a mechanosensation model of flow sensation in which the force of fluid flow drives asymmetric gene expression in the embryo.


Assuntos
Padronização Corporal/genética , Cílios/genética , Proteínas de Membrana/genética , Mesoderma/metabolismo , Proteína Nodal/genética , Canais de Cátion TRPP/genética , Animais , Embrião de Mamíferos/citologia , Regulação da Expressão Gênica no Desenvolvimento , Peptídeos e Proteínas de Sinalização Intercelular/genética , Mesoderma/embriologia , Camundongos , Camundongos Endogâmicos C3H , Camundongos Transgênicos , Proteína Nodal/biossíntese , Estrutura Terciária de Proteína , Canais de Cátion TRPP/antagonistas & inibidores
5.
Cancer Metastasis Rev ; 35(1): 21-39, 2016 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-26951550

RESUMO

The transforming growth factor beta (TGFß) superfamily member Nodal is an established regulator of early embryonic development, with primary roles in endoderm induction, left-right asymmetry, and primitive streak formation. Nodal signals through TGFß family receptors at the plasma membrane and induces signaling cascades leading to diverse transcriptional regulation. While conceptually simple, the regulation of Nodal and its molecular effects are profoundly complex and context dependent. Pioneering work by developmental biologists has characterized the signaling pathways, regulatory components, and provided detailed insight into the mechanisms by which Nodal mediates changes at the cellular and organismal levels. Nodal is also an important factor in maintaining pluripotency of embryonic stem cells through regulation of core transcriptional programs. Collectively, this work has led to an appreciation for Nodal as a powerful morphogen capable of orchestrating multiple cellular phenotypes. Although Nodal is not active in most adult tissues, its reexpression and signaling have been linked to multiple types of human cancer, and Nodal has emerged as a driver of tumor growth and cellular plasticity. In vitro and in vivo experimental evidence has demonstrated that inhibition of Nodal signaling reduces cancer cell aggressive characteristics, while clinical data have established associations with Nodal expression and patient outcomes. As a result, there is great interest in the potential targeting of Nodal activity in a therapeutic setting for cancer patients that may provide new avenues for suppressing tumor growth and metastasis. In this review, we evaluate our current understanding of the complexities of Nodal function in cancer and highlight recent experimental evidence that sheds light on the therapeutic potential of its inhibition.


Assuntos
Terapia de Alvo Molecular , Neoplasias/genética , Proteína Nodal/genética , Fator de Crescimento Transformador beta/genética , Regulação Neoplásica da Expressão Gênica , Humanos , Neoplasias/patologia , Neoplasias/terapia , Proteína Nodal/biossíntese , Transdução de Sinais
6.
Dev Biol ; 401(2): 236-48, 2015 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-25744724

RESUMO

Species-specific symmetry-breaking events at the left-right organizer (LRO) drive an evolutionarily-conserved cascade of gene expression in the lateral plate mesoderm that is required for the asymmetric positioning of organs within the body cavity. The mechanisms underlying the transfer of the left and right laterality information from the LRO to the lateral plate mesoderm are poorly understood. Here, we investigate the role of Claudin-10, a tight junction protein, in facilitating the transfer of left-right identity from the LRO to the lateral plate mesoderm. Claudin-10 is asymmetrically expressed on the right side of the chick LRO, Hensen's node. Gain- and loss-of-function studies demonstrated that right-sided expression of Claudin-10 is essential for normal rightward heart tube looping, the first morphological asymmetry during organogenesis. Manipulation of Claudin-10 expression did not perturb asymmetric gene expression at Hensen's node, but did disrupt asymmetric gene expression in the lateral plate mesoderm. Bilateral expression of Claudin-10 at Hensen's node prevented expression of Nodal, Lefty-2 and Pitx2c in the left lateral plate mesoderm, while morpholino knockdown of Claudin-10 inhibited expression of Snail1 in the right lateral plate mesoderm. We also determined that amino acids that are predicted to affect ion selectivity and protein interactions that bridge Claudin-10 to the actin cytoskeleton were essential for its left-right patterning function. Collectively, our data demonstrate a novel role for Claudin-10 during the transmission of laterality information from Hensen's node to both the left and right sides of the embryo and demonstrate that tight junctions have a critical role during the relay of left-right patterning cues from Hensen's node to the lateral plate mesoderm.


Assuntos
Padronização Corporal/genética , Claudinas/metabolismo , Mesoderma/metabolismo , Organizadores Embrionários/metabolismo , Junções Íntimas/metabolismo , Citoesqueleto de Actina/metabolismo , Animais , Embrião de Galinha , Claudinas/biossíntese , Claudinas/genética , Expressão Gênica , Regulação da Expressão Gênica no Desenvolvimento , Técnicas de Silenciamento de Genes , Coração/embriologia , Fatores de Determinação Direita-Esquerda/biossíntese , Morfolinos/genética , Proteína Nodal/biossíntese , Organogênese/genética , Transdução de Sinais/genética , Fatores de Transcrição da Família Snail , Fatores de Transcrição/biossíntese , Proteínas de Peixe-Zebra/biossíntese
7.
Cell Death Dis ; 6: e1688, 2015 Mar 12.
Artigo em Inglês | MEDLINE | ID: mdl-25766327

RESUMO

Both fibroblast growth factor 9 (Fgf9) and Kit Ligand (Kl) signal through tyrosine kinase receptors, yet they exert opposite effects on meiotic differentiation in postnatal spermatogonia, Fgf9 acting as a meiosis-inhibiting substance and Kl acting as a promoter of the differentiation process. To understand the molecular mechanisms that might underlie this difference, we tried to dissect the intracellular signaling elicited by these two growth factors. We found that both Fgf9 and Kl stimulate Erk1/2 activation in Kit+ (differentiating) spermatogonia, even though with different time courses, whereas Kl, but not Fgf9, elicits activation of the Pi3k-Akt pathway. Sustained Erk1/2 activity promoted by Fgf9 is required for induction of the autocrine Cripto-Nodal-Smad2/3 signaling loop in these cells. Nodal signaling, in turn, is essential to mediate Fgf9 suppression of the meiotic program, including inhibition of Stra8 and Scp3 expression and induction of the meiotic gatekeeper Nanos2. On the contrary, sustained activation of the Pi3k-Akt pathway is required for the induction of Stra8 expression elicited by Kl and retinoic acid. Moreover, we found that Kl treatment impairs Nodal mRNA expression and Fgf9-mediated Nanos2 induction, reinforcing the antagonistic effect of these two growth factors on the meiotic fate of male germ cells.


Assuntos
Fator 9 de Crescimento de Fibroblastos/biossíntese , Proteína Nodal/biossíntese , Proteína Smad2/biossíntese , Fator de Células-Tronco/biossíntese , Diferenciação Celular/genética , Fator 9 de Crescimento de Fibroblastos/genética , Regulação da Expressão Gênica no Desenvolvimento , Células Germinativas/crescimento & desenvolvimento , Células Germinativas/metabolismo , Humanos , Sistema de Sinalização das MAP Quinases/genética , Masculino , Meiose/genética , Óxido Nítrico Sintase Tipo II/biossíntese , Óxido Nítrico Sintase Tipo II/genética , Proteína Nodal/genética , Fosfatidilinositol 3-Quinases/genética , Transdução de Sinais , Espermatogônias/crescimento & desenvolvimento , Espermatogônias/metabolismo
8.
Pancreatology ; 15(2): 156-61, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25708930

RESUMO

BACKGROUND: Nodal (nodal growth differentiation factor) and its inhibitor Lefty (left right determination factor), which are ligands of the TGF (transforming growth factor) ß superfamily, are responsible for the determination of left-right asymmetry in vertebrates. Nodal/Lefty signaling has been suggested to play a role in the development of metastatic melanoma and breast cancer. However, it remains unclear whether this pathway is also involved in human pancreatic ductal adenocarcinoma (PDAC). METHODS: Pancreatic cancer patient specimens with clinical data (n = 54) were used to investigate the clinical significance of Nodal-Lefty signaling. A set of in vitro assays were carried out in a human pancreatic cancer cell line (Colo-357) to assess the functional relevance of Nodal-Lefty signaling. RESULTS: Nodal was absent in the human normal pancreas, while Lefty was present in islet cells. Though Nodal and Lefty expression were found in cancer cells at various expression levels, the cancer-associated tubular complexes were particularly positive for Lefty. Survival analysis revealed that high expression of Nodal correlated with reduced patient survival (median survival 17.8 vs 33.0 months, p = 0.013). Cultured pancreatic cancer cell lines expressed Nodal and Lefty at different levels. In vitro functional assays revealed that treatment with human recombinant Nodal inhibited cell growth and increased invasion of Colo-357 pancreatic cancer cells whereas no effect was found upon treatment with recombinant Lefty. CONCLUSION: Nodal-Lefty signaling might be involved in the pathogenesis of PDAC as Nodal expression marks a subtype of PDAC with unfavorable prognosis.


Assuntos
Proteína Nodal/biossíntese , Neoplasias Pancreáticas/mortalidade , Idoso , Biomarcadores Tumorais , Carcinogênese , Linhagem Celular Tumoral , Proliferação de Células , Feminino , Humanos , Fatores de Determinação Direita-Esquerda/biossíntese , Fatores de Determinação Direita-Esquerda/genética , Masculino , Pessoa de Meia-Idade , Invasividade Neoplásica/patologia , Proteína Nodal/genética , Neoplasias Pancreáticas/diagnóstico , RNA Interferente Pequeno , Análise de Sobrevida , Fator de Crescimento Transformador beta/fisiologia
9.
Mol Med Rep ; 12(1): 587-94, 2015 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-25672326

RESUMO

Expression of the nodal gene is high in a number of tumor cell types and may promote tumor growth. The expression of lefty, an inhibitor of nodal is often reduced in tumor cells. To the best of our knowledge, few studies have investigated the expression of nodal and lefty in renal cell carcinoma (RCC) cells. In the present study, quantitative polymerase chain reaction assays demonstrated that the level of nodal expression in RCC cells was high compared with that of adjacent non-tumor tissue cells, while the opposite pattern was observed for the level of lefty expression. Furthermore, lefty overexpression in RCC cells inhibited the expression of nodal. Nodal overexpression promoted RCC cell proliferation and invasion, and inhibited RCC cell apoptosis. Nodal downregulation and lefty overexpression led to similar observations: The inhibition of RCC cell proliferation and invasion, and the promotion of RCC cell apoptosis. The results of the present study suggested that the expression of nodal promoted RCC growth by activating the smad and extracellular signal-regulated kinases 1/2 pathways. The expression of lefty in RCC cells was lower than that in adjacent non-tumor cells, which may result in the overexpression of nodal, thereby promoting the growth of RCC. The results of the present study may therefore be useful for the development of novel biomarkers for RCC tumor diagnosis, and suggest a potential target gene for the treatment of RCC.


Assuntos
Carcinoma de Células Renais/genética , Proliferação de Células/genética , Fatores de Determinação Direita-Esquerda/biossíntese , Proteína Nodal/biossíntese , Adulto , Idoso , Apoptose/genética , Carcinoma de Células Renais/patologia , Linhagem Celular , MAP Quinases Reguladas por Sinal Extracelular/genética , Feminino , Regulação Neoplásica da Expressão Gênica , Humanos , Fatores de Determinação Direita-Esquerda/genética , Sistema de Sinalização das MAP Quinases , Masculino , Pessoa de Meia-Idade , Invasividade Neoplásica/genética , Proteína Nodal/genética
10.
Oncotarget ; 6(3): 1490-506, 2015 Jan 30.
Artigo em Inglês | MEDLINE | ID: mdl-25557170

RESUMO

Metastasis is the major cause for the high mortality rate of pancreatic cancer. Human embryonic stem cell (hESC) associated genes frequently correlate with malignant disease progression. Recent studies have demonstrated that the embryonic protein Nodal, which plays a critical role during embryonic development, is re-expressed in several types of tumors and promotes cancers progression. However, little is known about the role of Nodal in pancreatic cancer. Here, we show that Nodal expression is upregulated in human pancreatic cancer tissues. Moreover, Nodal expression levels correlate well with the grade of pancreatic cancer differentiation. In addition, we present clear evidence that Nodal induces signal transduction through the Smad2/3-dependent pathway in vitro. Furthermore, we show that Nodal promotes pancreatic cancer cell migration and invasion, induces epithelial-mesenchymal transition (EMT) and enhances the expression of matrix metalloproteinase-2 (MMP2) and CXC chemokine receptor 4 (CXCR4). Using an in vivo liver metastasis model of pancreatic cancer, we observed that blocking Nodal signaling activity with the small-molecule inhibitor SB431542 decreases the number and size of liver metastases. Taken together, our results suggest that Nodal overexpression induces a metastatic phenotype in pancreatic cancer cells, and that targeting Nodal signaling may be a promising therapeutic strategy for pancreatic cancer.


Assuntos
Carcinoma Ductal Pancreático/metabolismo , Carcinoma Ductal Pancreático/patologia , Proteína Nodal/biossíntese , Neoplasias Pancreáticas/metabolismo , Neoplasias Pancreáticas/patologia , Proteína Smad2/metabolismo , Proteína Smad3/metabolismo , Animais , Carcinoma Ductal Pancreático/genética , Diferenciação Celular/fisiologia , Linhagem Celular Tumoral , Movimento Celular/fisiologia , Feminino , Técnicas de Silenciamento de Genes , Xenoenxertos , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Metástase Neoplásica , Proteína Nodal/genética , Proteína Nodal/metabolismo , Neoplasias Pancreáticas/genética , Fenótipo , Transdução de Sinais , Proteína Smad2/genética , Proteína Smad3/genética , Regulação para Cima
11.
PLoS Genet ; 10(8): e1004548, 2014 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-25121599

RESUMO

In chordates, neural induction is the first step of a complex developmental process through which ectodermal cells acquire a neural identity. In ascidians, FGF-mediated neural induction occurs at the 32-cell stage in two blastomere pairs, precursors respectively of anterior and posterior neural tissue. We combined molecular embryology and cis-regulatory analysis to unveil in the ascidian Ciona intestinalis the remarkably simple proximal genetic network that controls posterior neural fate acquisition downstream of FGF. We report that the combined action of two direct FGF targets, the TGFß factor Nodal, acting via Smad- and Fox-binding sites, and the transcription factor Otx suffices to trigger ascidian posterior neural tissue formation. Moreover, we found that this strategy is conserved in the distantly related ascidian Phallusia mammillata, in spite of extreme sequence divergence in the cis-regulatory sequences involved. Our results thus highlight that the modes of gene regulatory network evolution differ with the evolutionary scale considered. Within ascidians, developmental regulatory networks are remarkably robust to genome sequence divergence. Between ascidians and vertebrates, major fate determinants, such as Otx and Nodal, can be co-opted into different networks. Comparative developmental studies in ascidians with divergent genomes will thus uncover shared ascidian strategies, and contribute to a better understanding of the diversity of developmental strategies within chordates.


Assuntos
Evolução Molecular , Redes Reguladoras de Genes , Neurogênese/genética , Proteína Nodal/genética , Fatores de Transcrição Otx/genética , Animais , Sítios de Ligação , Blastômeros , Sistema Nervoso Central/crescimento & desenvolvimento , Ciona intestinalis/genética , Ciona intestinalis/crescimento & desenvolvimento , Gástrula/crescimento & desenvolvimento , Regulação da Expressão Gênica no Desenvolvimento , Proteína Nodal/biossíntese , Fatores de Transcrição Otx/biossíntese
12.
Stem Cell Reports ; 1(3): 248-65, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-24319661

RESUMO

Cell-fate decisions and pluripotency are dependent on networks of key transcriptional regulators. Recent reports demonstrated additional functions of pluripotency-associated factors during early lineage commitment. The T-box transcription factor TBX3 has been implicated in regulating embryonic stem cell self-renewal and cardiogenesis. Here, we show that TBX3 is dynamically expressed during specification of the mesendoderm lineages in differentiating embryonic stem cells (ESCs) in vitro and in developing mouse and Xenopus embryos in vivo. Forced TBX3 expression in ESCs promotes mesendoderm specification by directly activating key lineage specification factors and indirectly by enhancing paracrine Nodal/Smad2 signaling. TBX3 loss-of-function analyses in the Xenopus underline its requirement for mesendoderm lineage commitment. Moreover, we uncovered a functional redundancy between TBX3 and Tbx2 during Xenopus gastrulation. Taken together, we define further facets of TBX3 actions and map TBX3 as an upstream regulator of the mesendoderm transcriptional program during gastrulation.


Assuntos
Diferenciação Celular , Células-Tronco Embrionárias/citologia , Gastrulação/genética , Mesoderma/crescimento & desenvolvimento , Proteínas com Domínio T/biossíntese , Animais , Linhagem da Célula , Regulação da Expressão Gênica no Desenvolvimento/genética , Mesoderma/metabolismo , Camundongos , Proteína Nodal/biossíntese , Proteína Nodal/genética , Proteína Smad2/genética , Proteínas com Domínio T/genética , Xenopus
13.
Hypertension ; 61(4): 864-72, 2013 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-23424236

RESUMO

Preeclampsia is a major disorder of pregnancy and a leading cause of maternal and perinatal morbidity and mortality. MicroRNAs are small noncoding RNAs that regulate gene expression posttranscriptionally. In this study, we examined the expression of miR-376c and found that miR-376c levels were downregulated in both placental and plasma samples collected from preeclamptic patients, when compared with the normal pregnant women at the same gestational stage. Overexpression of miR-376c induced trophoblast cell proliferation, migration, and invasion in HTR8/SVneo cells and promoted placental explant outgrowth. In contrast, inhibition of endogenous miR-376c resulted in a decrease in trophoblast cell invasion and placental explant outgrowth. We identified activin receptor-like kinase 5 (ALK5), a type I receptor for transforming growth factor-ß, and ALK7, a type I receptor for Nodal, as targets of miR-376c. Overexpression of miR-376c repressed transforming growth factor-ß and Nodal functions, whereas overexpression of ALK5 and ALK7 reversed the effects of miR-376c. These results demonstrate that miR-376c inhibits both ALK5 and ALK7 expression to impair transforming growth factor-ß/Nodal signaling, leading to increases in cell proliferation and invasion. An unbalanced Nodal/transforming growth factor-ß and miR-376c expression may lead to the development of preeclampsia.


Assuntos
Regulação da Expressão Gênica , MicroRNAs/genética , Proteína Nodal/genética , Pré-Eclâmpsia/genética , RNA/genética , Fator de Crescimento Transformador beta/genética , Trofoblastos/metabolismo , Proliferação de Células , Feminino , Humanos , MicroRNAs/metabolismo , Proteína Nodal/biossíntese , Placenta/citologia , Placenta/metabolismo , Pré-Eclâmpsia/metabolismo , Gravidez , Transdução de Sinais/genética , Fator de Crescimento Transformador beta/biossíntese , Trofoblastos/citologia
14.
Mol Biol Cell ; 22(24): 4809-21, 2011 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-22031289

RESUMO

Low oxygen (O(2)) levels characterize the microenvironment of both stem cells and rapidly growing tumors. Moreover, hypoxia is associated with the maintenance of stem cell-like phenotypes and increased invasion, angiogenesis and metastasis in cancer patients. Metastatic cancers, such as breast cancer and melanoma, aberrantly express the embryonic morphogen Nodal, and the presence of this protein is correlated with metastatic disease. In this paper, we demonstrate that hypoxia induces Nodal expression in melanoma and breast cancer cells concomitant with increased cellular invasion and angiogenic phenotypes. Of note, Nodal expression remains up-regulated up to 48 h following reoxygenation. The oxygen-mediated regulation of Nodal expression occurs via a combinatorial mechanism. Within the first 24 h of exposure to low O(2), there is an increase in protein stability. This increase in stability is accompanied by an induction of transcription, mediated by the HIF-1α-dependent activation of Notch-responsive elements in the node-specific enhancer of the Nodal gene locus. Finally, Nodal expression is maintained upon reoxygenation by a canonical SMAD-dependent feed-forward mechanism. This work provides insight into the O(2)-mediated regulation of Nodal, a key stem cell-associated factor, and reveals that Nodal may be a target for the treatment and prevention of hypoxia-induced tumor progression.


Assuntos
Neoplasias da Mama/metabolismo , Regulação Neoplásica da Expressão Gênica , Melanoma/metabolismo , Proteínas de Neoplasias/biossíntese , Proteína Nodal/biossíntese , Oxigênio/metabolismo , Neoplasias da Mama/genética , Neoplasias da Mama/patologia , Hipóxia Celular/genética , Linhagem Celular Tumoral , Feminino , Loci Gênicos , Humanos , Subunidade alfa do Fator 1 Induzível por Hipóxia/genética , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Masculino , Melanoma/genética , Melanoma/patologia , Proteínas de Neoplasias/genética , Proteína Nodal/genética , Estabilidade Proteica , Elementos de Resposta , Proteínas Smad/genética , Proteínas Smad/metabolismo , Fatores de Tempo
15.
Cancer Res ; 70(24): 10340-50, 2010 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-21159651

RESUMO

Metastatic melanoma is an aggressive skin cancer associated with poor prognosis. The reactivation of the embryonic morphogen Nodal in metastatic melanoma has previously been shown to regulate the aggressive behavior of these tumor cells. During the establishment of left-right asymmetry in early vertebrate development, Nodal expression is specifically regulated by a Notch signaling pathway. We hypothesize that a similar relationship between Notch and Nodal may be reestablished in melanoma. In this study, we investigate whether cross talk between the Notch and Nodal pathways can explain the reactivation of Nodal in aggressive metastatic melanoma cells. We show a molecular link between Notch and Nodal signaling in the aggressive melanoma cell line MV3 via the activity of an RBPJ-dependent Nodal enhancer element. We show a precise correlation between Notch4 and Nodal expression in multiple aggressive cell lines but not poorly aggressive cell lines. Surprisingly, Notch4 is specifically required for expression of Nodal in aggressive cells and plays a vital role both in the balance of cell growth and in the regulation of the aggressive phenotype. In addition, Notch4 function in vasculogenic mimicry and anchorage-independent growth in vitro is due in part to Notch4 regulation of Nodal. This study identifies an important role for cross talk between Notch4 and Nodal in metastatic melanoma, placing Notch4 upstream of Nodal, and offers a potential molecular target for melanoma therapy.


Assuntos
Melanoma/metabolismo , Proteína Nodal/metabolismo , Proteínas Proto-Oncogênicas/metabolismo , Receptores Notch/metabolismo , Apoptose/fisiologia , Processos de Crescimento Celular/fisiologia , Linhagem Celular Tumoral , Humanos , Melanoma/irrigação sanguínea , Melanoma/genética , Melanoma/patologia , Neovascularização Patológica/metabolismo , Neovascularização Patológica/patologia , Proteína Nodal/biossíntese , Proteína Nodal/genética , Proteínas Proto-Oncogênicas/antagonistas & inibidores , Proteínas Proto-Oncogênicas/biossíntese , RNA Interferente Pequeno/administração & dosagem , RNA Interferente Pequeno/genética , Receptor Cross-Talk , Receptor Notch4 , Receptores Notch/antagonistas & inibidores , Receptores Notch/biossíntese , Transdução de Sinais , Transfecção
16.
Mod Pathol ; 23(9): 1209-14, 2010 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-20495543

RESUMO

Nodal, a potent embryonic morphogen in the transforming growth factor-beta family, is a proposed key regulator of melanoma tumorigenicity. However, there has been no systematic study of Nodal expression in melanocytic lesions. We investigated Nodal expression by immunohistochemistry in 269 melanocytic lesions, including compound nevi, dysplastic nevi, congenital nevi, Spitz nevi, melanoma in situ, malignant melanoma including the variant desmoplastic melanoma, and metastatic melanoma. We found that the Nodal expression was significantly increased in malignant lesions (including melanoma in situ, malignant melanoma, and metastatic melanoma) compared with compound nevi, Spitz nevi, and dysplastic nevi. Surprisingly, congenital nevi expressed a level of Nodal comparable with malignant lesions, whereas desmoplastic melanoma showed lower expression than nondesmoplastic malignant melanoma (P<0.05). Deep melanoma (Breslow depth >1 mm) displayed a higher percentage of Nodal-positive tumor cells than did superficial melanoma (Breslow depth < or =1 mm), although there was no statistical difference in the overall staining intensity (P=0.18). Melanomas in situ showed a lower level of Nodal expression than did deep melanomas and metastatic melanomas (P<0.05). The low expression of Nodal in normal and dysplastic nevi, and its increasing expression with the progression of malignant lesions, are suggestive of a role for Nodal in melanoma progression.


Assuntos
Melanoma/metabolismo , Melanoma/patologia , Proteína Nodal/biossíntese , Neoplasias Cutâneas/metabolismo , Neoplasias Cutâneas/patologia , Biomarcadores Tumorais/análise , Síndrome do Nevo Displásico/metabolismo , Síndrome do Nevo Displásico/patologia , Humanos , Imuno-Histoquímica , Nevo Pigmentado/metabolismo , Nevo Pigmentado/patologia , Lesões Pré-Cancerosas/metabolismo , Lesões Pré-Cancerosas/patologia , Fator de Crescimento Transformador beta/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA