24 resultados para Amphiregulin


Relevância:

20.00% 20.00%

Publicador:

Resumo:

Abstract Ovarian hormones are key regulators of postnatal mammary gland development and are linked to breast carcinogenesis. In particular, estrogens induce mammary epithelial cells to proliferate at the onset of puberty, leading to the elongation of the rudimental ductal tree into the fatty stromal tissue. Elucidating the molecular events underlying estrogen mitogenic activity in the mammary gland is of value in understanding how the deregulation of this signalling pathway can lead to breast tumorigenesis. Our lab has recently shown that estrogen induces mammary proliferation via epithelial estrogen receptor alpha (ERα) by a paracrine mechanism. Based on the finding that several EGF receptor (EGFR) ligands are able to substitute for estrogens and that amphiregulin (Areg), one of these ligands, is required during mammary development, we have hypothesized that Areg is a key mediator of estrogen induced paracrine signalling during ductal morphogenesis. Our analysis of the Areg -/- mice mammary phenotype reveals that epithelial Areg is required at the onset of puberty for epithelial proliferation, terminal end bud (TEB) formation and, subsequently, ductal elongation. Hormonal stimulation experiments show that among the EGFR ligands, only Arég is specifically controlled by estrogen at the transcriptional level, via ERα, in the mammary gland. Moreover, Areg is required for the estrogen-induced mammotrophic effects of epithelial proliferation and ductal elongation. We have shown that ectopic Areg expression in ERα -/- mammary epithelial cells is sufficient to induce ductal morphogenesis. Our transplantations experiment show that when Areg -/- cells are in the presence of wt cells they contribute to all aspects of ductal development, suggesting that this growth factor acts in a paracrine fashion. Moreover, this result shows that Areg -/- epithelial cells are not intrinsically impaired in proliferation. Our transplantation experiment carried out under physiological conditions confirmed previous reports showing that stromal EGFR is needed for ductal morphogenesis. This suggests that estrogen-driven Areg signalling involves an epithelium-stroma crosstalk. Thus, these data confirmed our hypothesis of Areg being an important estrogen mediator during ductal morphogenesis. Résumé Les hormones ovariennes, régulatrices clés du développement post-natal de la glande mammaire, sont également liées à la carcinogénèse du sein. En particulier, l'oestrogène induit la division des cellules épithéliales au début de la puberté. Cette prolifération amène à l'élongation du réseau canalaire rudimentaire et permet l'invasion du compartiment stromal. L'élucidation des mécanismes moléculaires responsables de l'activité mitogénique de l'oestrogène dans la glande mammaire est précieuse pour une meilleure compréhension du développement du cancer du sein. Notre laboratoire a récemment démontré que l'cestrogène induit la prolifération des cellules épithéliales par un signal paracrine, grâce au récepteur à l'oestrogène alpha (ERα). En se basant sur le fait que plusieurs ligands du récepteur à l'EGF (EGFR) sont capables de se substituer à l'cestrogène et d'induire la prolifération épithéliale et qu'amphiregulin (Areg), un de ces ligands, est essentielle au développement de la glande mammaire, nous avons émi l'hypothèse que Areg est un médiateur essentiel du signal paracrine induit par l'oestrogène pendant la croissance du système canalaire. Nos analyses phénotypiques des glandes mammaires issues de souris transgéniques Areg -/- démontrent que cette protéine est indispensable à la prolifération des cellules épithéliales mammaires au début de la puberté et à la formation des bourgeons terminaux qui conduisent à l'élongation des canaux. Nos expériences de stimulations hormonales démontrent que, parmi l'ensemble des ligands du EGFR, seule Areg est contrôlée au niveau transcriptionnel par l'cestrogène dans la glande mammaire, ceci via le récepteur ERα. De plus, Areg est essentielle pour le effets mammotrophique induit par l'cestrogène, à savoir la prolifération épithéliál et la croissance du système canalaire. Par ailleurs, l'expression ectopique d'Areg dans des cellules epithéliales mammaires de souris transgéniques ERα -/- est suffisante pour permettre la formation du réseau canalaire. En présence de cellules normales, les cellules dépourvues du gène d'Areg contribuent à la formation des canaux. Cette expérience suggère que ce facteur de croissance agit de manière paracrine. De plus, ce résultat montre que les cellules épithéliales Areg -/- conservent leur potentiel prolifératif. Nos expériences de transplantation, réalisées dans des conditions physiologiques, ont confirmé des précédentes études qui montraient que le récepteur stromal à l'EGF est nécessaire pour la morphogénèse du système canalaire. Ceci suggère que la voie de signalisation activée par l'oestrogène et dépendante d' implique une communication entre l'épithélium et le stroma. Ainsi, ces résultats valident notre hypothèse puisqu'ils confirment Areg en tant que médiateur majeur de l'oestrogène dans la morphogénèse du système canalaire.

Relevância:

20.00% 20.00%

Publicador:

Resumo:

Conselho Nacional de Desenvolvimento Científico e Tecnológico (CNPq)

Relevância:

20.00% 20.00%

Publicador:

Resumo:

Fundação de Amparo à Pesquisa do Estado de São Paulo (FAPESP)

Relevância:

20.00% 20.00%

Publicador:

Resumo:

The barrier surfaces of the skin, lung, and intestine are constantly exposed to environmental stimuli that can result in inflammation and tissue damage. Interleukin (IL)-33-dependent group 2 innate lymphoid cells (ILC2s) are enriched at barrier surfaces and have been implicated in promoting inflammation; however, the mechanisms underlying the tissue-protective roles of IL-33 or ILC2s at surfaces such as the intestine remain poorly defined. Here we demonstrate that, following activation with IL-33, expression of the growth factor amphiregulin (AREG) is a dominant functional signature of gut-associated ILC2s. In the context of a murine model of intestinal damage and inflammation, the frequency and number of AREG-expressing ILC2s increases following intestinal injury and genetic disruption of the endogenous AREG-epidermal growth factor receptor (EGFR) pathway exacerbated disease. Administration of exogenous AREG limited intestinal inflammation and decreased disease severity in both lymphocyte-sufficient and lymphocyte-deficient mice, revealing a previously unrecognized innate immune mechanism of intestinal tissue protection. Furthermore, treatment with IL-33 or transfer of ILC2s ameliorated intestinal disease severity in an AREG-dependent manner. Collectively, these data reveal a critical feedback loop in which cytokine cues from damaged epithelia activate innate immune cells to express growth factors essential for ILC-dependent restoration of epithelial barrier function and maintenance of tissue homeostasis.

Relevância:

10.00% 10.00%

Publicador:

Resumo:

Dermcidin (DCD) is a human gene mapped to chromosome 12q13 region, which is co-amplified with multiple oncogenes with a well-established role in the growth, survival and progression of breast cancers. Here, we present a summary of a DNA microarray-based study that identified the genes that are up- and down-regulated in a human MDA-361 pLKO control clone and three clones expressing short hairpin RNA against three different regions of DCD mRNA. A list of 235 genes was differentially expressed among independent clones (> 3-fold change and P < 0.005). The gene expression of 208 was reduced and of 27 was increased in the three DCD-RNAi clones compared to pLKO control clone. The expression of 77 genes (37%) encoding for enzymes involved in amino acid metabolism, glucose metabolism and oxidoreductase activity and several genes required for cell survival and DNA repair were decreased. The expression of EGFR/ErbB-1 gene, an important predictor of outcome in breast cancer, was reduced together with the genes for betacellulin and amphiregulin, two known ligands of EGFR/ErbB receptors. Many of the 27 genes up-regulated by DCD-RNAi expression have not yet been fully characterized; among those with known function, we identified the calcium-calmodulin-dependent protein kinase-II delta and calcineurin A alpha. We compared 132 up-regulated and 12 down-regulated genes in our dataset with those genes up- and down-regulated by inhibitors targeting various signaling pathway components. The analysis showed that the genes in the DCD pathway are aligned with those functionally influenced by the drugs sirolimus, LY-294002 and wortmannin. Therefore, DCD may exert its function by activating the PI3K/AKT/mTOR signaling pathway. Together, these bioinformatic approaches suggest the involvement of DCD in the regulation of genes for breast cancer cell metabolism, proliferation and survival.

Relevância:

10.00% 10.00%

Publicador:

Resumo:

Behçet's disease (BD) is a complex disease with genetic and environmental risk factors implicated in its etiology; however, its pathophysiology is poorly understood. To decipher BD's genetic underpinnings, we combined gene expression profiling with pathway analysis and association studies. We compared the gene expression profiles in peripheral blood mononuclear cells (PBMCs) of 15 patients and 14 matched controls using Affymetrix microarrays and found that the neuregulin signaling pathway was over-represented among the differentially expressed genes. The Epiregulin (EREG), Amphiregulin (AREG), and Neuregulin-1 (NRG1) genes of this pathway stand out as they are also among the top differentially expressed genes. Twelve haplotype tagging SNPs at the EREG-AREG locus and 15 SNPs in NRG1 found associated in at least one published BD genome-wide association study were tested for association with BD in a dataset of 976 Iranian patients and 839 controls. We found a novel association with BD for the rs6845297 SNP located downstream of EREG, and replicated three associations at NRG1 (rs4489285, rs383632, and rs1462891). Multifactor dimensionality reduction analysis indicated the existence of epistatic interactions between EREG and NRG1 variants. EREG-AREG and NRG1, which are members of the epidermal growth factor (EGF) family, seem to modulate BD susceptibility through main effects and gene–gene interactions. These association findings support a role for the EGF/ErbB signaling pathway inBD pathogenesis that warrants further investigation and highlight the importance of combining genetic and genomic approaches to dissect the genetic architecture of complex diseases.

Relevância:

10.00% 10.00%

Publicador:

Resumo:

Ductal growth of the mammary gland occurs in two distinct stages. The first round of branching morphogenesis occurs during embryogenesis, and the second round commences at the onset of puberty. Currently, relatively little is known about the genetic networks that control the initial phases of ductal expansion, which, unlike pubertal development, proceeds independent of hormonal input in female mice. Here we identify NF-κB downstream of the TNF-like ligand ectodysplasin (Eda) as a unique regulator of embryonic and prepubertal ductal morphogenesis. Loss of Eda, or inhibition of NF-κB, led to smaller ductal trees with fewer branches. On the other hand, overexpression of Eda caused a dramatic NF-κB-dependent phenotype in both female and male mice characterized by precocious and highly increased ductal growth and branching that correlated with enhanced cell proliferation. We have identified several putative transcriptional target genes of Eda/NF-κB, including PTHrP, Wnt10a, and Wnt10b, as well as Egf family ligands amphiregulin and epigen. We developed a mammary bud culture system that allowed us to manipulate mammary development ex vivo and found that recombinant PTHrP, Wnt3A, and Egf family ligands stimulate embryonic branching morphogenesis, suggesting that these pathways may cooperatively mediate the effects of Eda.

Relevância:

10.00% 10.00%

Publicador:

Resumo:

A woman's risk of breast cancer is strongly affected by her reproductive history. The hormonal milieu is also a key determinant of the course of the disease. Combining mouse genetics with tissue recombination techniques, we have established that the female reproductive hormones, estrogens, progesterone, and prolactin, act sequentially on the mammary epithelium to trigger distinct developmental steps. The hormones impinge directly on a subset of luminal mammary epithelial cells that express the respective hormone receptors and act as sensor cells translating and amplifying systemic signals into local stimuli. Local signaling is stage and age specific. During puberty, estrogens promote proliferation using the EGF family member, amphiregulin, as essential paracrine mediator. In adulthood, progesterone, rather than estrogen, is the major inducer of stem cell activation and cell proliferation of the mammary epithelium. Hormonal signaling modulates crucial developmental pathways that impinge on mammary stem cell populations, while Notch signaling, by inhibiting p63, is central to mammary cell fate determination. Cell proliferation occurs in two waves. The first results from direct stimulation of the small fraction of hormone receptor positive cells. It is followed by a second wave of progesterone-induced proliferation involving mostly hormone receptor negative cells, in which RANKL is a key mediator. A model in which repeated activation of paracrine signaling by progesterone with resulting stem cell activation promotes breast carcinogenesis is proposed.

Relevância:

10.00% 10.00%

Publicador:

Resumo:

PURPOSE: The EGF receptor (EGFR) is overexpressed in the majority of metastatic castration-resistant prostate cancers (mCRPC) and might represent a valid therapeutic target. The combination of docetaxel and cetuximab, the monoclonal antibody against EGFR, has not been tested in patients with prostate cancer. EXPERIMENTAL DESIGN: Patients with mCRPC progressing during or within 90 days after at least 12 weeks of docetaxel were included in this phase II trial. Treatment consisted of docetaxel (75 mg/m(2) every 3 weeks or 35 mg/m(2) on days 1, 8, 15 every 4 weeks) in combination with cetuximab (400 mg/m(2) on day 1 and then 250 mg/m(2) weekly). The primary endpoint was progression-free survival (PFS) at 12 weeks defined as the absence of prostate-specific antigen (PSA), radiographic, or clinical progression. Evaluation of known biomarkers of response and resistance to cetuximab (EGFR, PTEN, amphiregulin, epiregulin) was conducted. RESULTS: Thirty-eight patients were enrolled at 15 Swiss centers. Median age was 68 years and median PSA was 212 ng/mL. PFS at 12 weeks was 34% [95% confidence interval (CI), 19%-52%], PFS at 24 weeks was 20%, and median overall survival (OS) was 13.3 months (95% CI, 7.3-15.4). Seven patients (20%) had a confirmed ≥ 50% and 11 patients (31%) a confirmed ≥ 30% PSA decline. About 47% of enrolled patients experienced grade 3 and 8% grade 4 toxicities. A significantly improved PFS was found in patients with overexpression of EGFR and persistent activity of PTEN. CONCLUSIONS: EGFR inhibition with cetuximab might improve the outcome of patients with mCRPC. A potential correlation between EGFR overexpression, persistent expression of PTEN, and EGFR inhibition should be investigated further.

Relevância:

10.00% 10.00%

Publicador:

Resumo:

Abstract : Breast cancer incidence rates have increased over the past hundred years, in particular, in Western industrial countries and they continue to rise worldwide. Breast cancer risk has been linked to life exposure to endogenous and exogenous estrogens, and there is increasing concern that exposure to endocrine disruptors which are increasingly accumulating in our environment may also have a role. Using the mouse as model, I have analyzed the physiological role of estrogen signaling in mammary gland development. I have shown that estrogen signaling through the estrogen receptor alpha (ERα) in the mammary epithelium is required for ductal morphogenesis during puberty. Moreover, I have demonstrated that estrogens induce proliferation of mammary epithelial cells through a paracrine mechanism. The presence of estrogen signaling is essential cell intrinsically via ERα or ERβ for the terminal differentiation into milk secreting cells during pregnancy. Furthermore, I have examined how perinatal exposure to the estrogenic plasticizer bisphenol A (BPA) found ubiquitously in consumer goods such as baby bottles formula and beverage containers affects the normal mammary gland development and possibly predispose the mammary gland to tumorigenesis. I have found that C57b16 mice that were exposed, via their drinking water, to several BPA doses ranging from 0.025µg/kg/day to 250µg/kg/day exhibits delayed terminal end bud formation and consequently the ductal outgrowth. Later in life, the mice that were exposed in utero to BPA displayed an increased number of mammary epithelial cells. Acute exposure of 3-week-old mice to BPA can alter gene expression levels of an important estrogen target gene, amphiregulin. Taken together these data are compatible with a scenario in which perinatal BPA exposure may alter mammary gland development by affecting developmental signaling pathways. Résumé : Les taux d'incidence des cancers du sein ont augmenté au cours des cent dernières années en particulier dans les pays industriels occidentaux et ils continuent d'augmenter dans le monde entier. Le risque du cancer du sein a été corrélé à l'exposition au cours de la vie aux oestrogènes endogènes et exogènes. Il y a une préoccupation croissante concernant l'exposition aux perturbateurs endocriniens qui ne cessent de s'accumulent dans notre environnement et qui peuvent également avoir un rôle dans l'augmentation des cancers du sein. En utilisant le modèle de souris, j'ai analysé le rôle physiologique de la voie de signalisation à l'oestrogène dans le développement mammaire. J'ai prouvé que l'oestrogène par l'intermédiaire de son récepteur alpha (ERα) est indispensable dans l'épithélium pour la morphogénèse du système canalaire pendant la puberté. De plus, j'ai démontré que les oestrogènes induisent la prolifération des cellules épithéliales mammaires par un mécanisme paracrine. La présence de la voie de signalisation à l'oestrogène est essentielle de manière intrinsèque à la cellule par l'intermédiaire d'ERα ou ERβ pour la différentiation terminale des cellules épithéliales en cellules sécrétrices de lait pendant la grossesse. En outre, j'ai examiné comment l'exposition périnatale au bisphénol A (BPA), un plastifiant présentant des propriétés ostrogéniques et omniprésent dans divers produits d'usage courant tels que les biberons des bébés et les récipients en plastique, affecte le développement de la glande mammaire et prédispose probablement celle-ci à la tumorigénèse. J'ai constaté que l'exposition périnatale à BPA retarde la formation des bourgeons terminaux et par conséquent la croissance du système canalaire. Plus tard dans la vie, les souris qui ont été exposées dans l'utérus au BPA ont montré un plus grand nombre de cellules épithéliales mammaires. L'exposition aiguë de souris âgées de 3 semaines au BPA perturbe le niveau d'expression d'un gène cible important de l'oestrogène, l'amphiregulin. Ces données sont compatibles avec un scénario dans lequel l'exposition périnatale au BPA peut changer le développement de la glande mammaire en affectant des voies de signalisation développementales.

Relevância:

10.00% 10.00%

Publicador:

Resumo:

Recent findings have implicated Fas/Fas ligand (FasL) in mediating the death of keratinocytes in spongiotic lesions. We asked whether dying keratinocytes could potentially initiate a protective response of the skin to limit the destruction of the epidermis in the spongiotic areas. In addition to apoptosis, treatment of keratinocyte cultures in vitro with FasL triggers a profound phoshorylation of the epidermal growth factor receptor (EGFR) and of its downstream effectors ERK and protein kinase B (PKB/Akt). Using a variety of inhibitors and blocking antibodies, we demonstrated that: (i) apoptosis is required for the generation of the signal(s) leading to the activation of EGFR, ERK, and Akt; (ii) the activation of EGFR, ERK, and Akt by FasL is indeed mediated by its bona fide receptor Fas; (iii) the activation of EGFR is essential for the subsequent activation of ERK and Akt; and (iv) apoptotic keratinocytes secrete soluble EGFR ligands (including amphiregulin) that are processed from membrane-bound proligand forms by metalloproteinase(s). Our findings demonstrate a potential mechanism for the restriction and repair of spongiotic damage in eczemas.

Relevância:

10.00% 10.00%

Publicador:

Resumo:

Intrinsic resistance to the epidermal growth factor receptor (EGFR; HER1) tyrosine kinase inhibitor (TKI) gefitinib, and more generally to EGFR TKIs, is a common phenomenon in breast cancer. The availability of molecular criteria for predicting sensitivity to EGFR-TKIs is, therefore, the most relevant issue for their correct use and for planning future research. Though it appears that in non-small-cell lung cancer (NSCLC) response to gefitinib is directly related to the occurrence of specific mutations in the EGFR TK domain, breast cancer patients cannot be selected for treatment with gefitinib on the same basis as such EGFR mutations have beenreported neither in primary breast carcinomas nor in several breast cancer cell lines. Alternatively, there is a generalagreement on the hypothesis that the occurrence of molecular alterations that activate transduction pathways downstreamof EGFR (i.e., MEK1/MEK2 - ERK1/2 MAPK and PI-3'K - AKT growth/survival signaling cascades) significantly affect the response to EGFR TKIs in breast carcinomas. However,there are no studies so far addressing a role of EGF-related ligands as intrinsic breast cancer cell modulators of EGFR TKIefficacy. We recently monitored gene expression profiles andsub-cellular localization of HER-1/-2/-3/-4 related ligands (i.e., EGF, amphiregulin, transforming growth factor-α, ß-cellulin,epiregulin and neuregulins) prior to and after gefitinib treatment in a panel of human breast cancer cell lines. First, gefitinibinduced changes in the endogenous levels of EGF-related ligands correlated with the natural degree of breast cancer cellsensitivity to gefitinib. While breast cancer cells intrinsically resistant to gefitinib (IC50 ≥15 μM) markedly up-regulated(up to 600 times) the expression of genes codifying for HERspecific ligands, a significant down-regulation (up to 106 times)of HER ligand gene transcription was found in breast cancer cells intrinsically sensitive to gefitinib (IC50 ≤1 μM). Second,loss of HER1 function differentially regulated the nuclear trafficking of HER-related ligands. While gefitinib treatment induced an active import and nuclear accumulation of the HER ligand NRG in intrinsically gefitinib-resistant breastcancer cells, an active export and nuclear loss of NRG was observed in intrinsically gefitinib-sensitive breast cancer cells.In summary, through in vitro and pharmacodynamic studies we have learned that, besides mutations in the HER1 gene,oncogenic changes downstream of HER1 are the key players regulating gefitinib efficacy in breast cancer cells. It now appears that pharmacological inhibition of HER1 functionalso leads to striking changes in both the gene expression and the nucleo-cytoplasmic trafficking of HER-specific ligands,and that this response correlates with the intrinsic degree of breast cancer sensitivity to the EGFR TKI gefitinib. Therelevance of this previously unrecognized intracrine feedback to gefitinib warrants further studies as cancer cells could bypassthe antiproliferative effects of HER1-targeted therapeutics without a need for the overexpression and/or activation of other HER family members and/or the activation of HER-driven downstream signaling cascades