Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 19 de 19
Filtrer
1.
Cell Signal ; 104: 110588, 2023 04.
Article de Anglais | MEDLINE | ID: mdl-36621728

RÉSUMÉ

The receptor tyrosine kinase orphan receptor 1 (ROR1) is a receptor for WNT5A and related Wnt proteins, that play an important role during embryonic development by regulating cell migration, cell polarity, neural patterning, and organogenesis. ROR1 exerts these functions by transducing signals from the Wnt secreted glycoproteins to the intracellular Wnt/PCP and Wnt/Ca++ pathways. Investigations in adult human cells, particularly cancer cells, have demonstrated that besides these two pathways, the WNT5A/ROR1 axis can activate a number of signaling pathways, including the PI3K/AKT, MAPK, NF-κB, STAT3, and Hippo pathways. Moreover, ROR1 is aberrantly expressed in cancer and was associated with tumor progression and poor survival by promoting cell proliferation, survival, invasion, epithelial to mesenchymal transition, and metastasis. Consequently, numerous therapeutic tools to target ROR1 are currently being evaluated in cancer patients. In this review, we will provide a detailed description of the signaling pathways regulated by ROR1 in cancer and their impact in tumor progression.


Sujet(s)
Transition épithélio-mésenchymateuse , Tumeurs , Grossesse , Femelle , Humains , Phosphatidylinositol 3-kinases/métabolisme , Transduction du signal , Tumeurs/génétique , Protéines de type Wingless/métabolisme , Lignée cellulaire tumorale , Récepteurs orphelins de type récepteur à tyrosine kinase/génétique , Récepteurs orphelins de type récepteur à tyrosine kinase/métabolisme
2.
J Cell Commun Signal ; 17(1): 75-88, 2023 Mar.
Article de Anglais | MEDLINE | ID: mdl-35723796

RÉSUMÉ

Receptor tyrosine kinase-like orphan receptor 2 (ROR2) is a protein with important functions during embryogenesis that is dysregulated in human cancer. An intriguing feature of this receptor is that it plays opposite roles in different tumor types either promoting or inhibiting tumor progression. Understanding the complex role of this receptor requires a more profound exploration of both the altered biological and molecular mechanisms. Here, we describe that ROR2 promotes Epithelial-Mesenchymal Transition (EMT) by inducing cadherin switch and the upregulation of the transcription factors ZEB1, Twist, Slug, Snail, and HIF1A, together with a mesenchymal phenotype and increased migration. We show that ROR2 activates both p38 and ERK mitogen-activated protein kinase pathways independently of Wnt5a. Further, we demonstrated that the upregulation of EMT-related proteins depends on the hyperactivation of the ERK pathway far above the typical high constitutive activity observed in melanoma. In addition, ROR2 also promoted ERK phosphorylation, EMT, invasion, and necrosis in xenotransplanted mice. ROR2 also associates with EMT in tumor samples from melanoma patients where analysis of large cohorts revealed that increased ROR2 levels are linked to EMT signatures. This important role of ROR2 translates into melanoma patient' s prognosis since elevated ROR2 levels reduced overall survival and distant metastasis-free survival of patients with lymph node metastasis. In sum, these results demonstrate that ROR2 contributes to melanoma progression by inducing EMT and necrosis and can be an attractive therapeutic target for melanoma.

3.
Cancer Cell Int ; 22(1): 288, 2022 Sep 20.
Article de Anglais | MEDLINE | ID: mdl-36127680

RÉSUMÉ

Receptor tyrosine kinase-like orphan receptor 2 (ROR2) is a receptor for the Wnt5a ligand that was shown to play a dual role in cancer. ROR2 was shown to either suppress or promote tumor progression in different tumor types by regulating the same biological processes (i.e. proliferation, invasion) in opposite ways. We have recently observed that ROR2 plays multiple and somewhat contradictory roles in melanoma where it impairs cell proliferation but promotes migration, EMT and chemoresistance. In the present article, ROR2 is proposed to be a major driver of "phenotype switching" in melanoma that can tilt the cellular behavior toward proliferative or invasive phenotypes. This function of ROR2 has therapeutic implications since it would provide an opportunity for targeting specific phenotypes such as invasive and drug-resistant ones by inhibiting ROR2.

4.
Med Oncol ; 39(10): 141, 2022 Jul 14.
Article de Anglais | MEDLINE | ID: mdl-35834068

RÉSUMÉ

Several diagnostic and prognostic markers for melanoma have been identified in last few years. However, their actual contribution to melanoma progression have not been investigated in detail. This study was aimed to identify genes, biological processes, and signaling pathways implicated in melanoma progression by applying bioinformatics analysis. We identified nine differentially expressed genes (DEGs) (IL36RN, KRT6A, KRT6B, KRT16, S100A7, SPRR1A, SPRR1B, SPRR2B, and KLK7) that were upregulated in primary melanoma compared with metastatic melanoma in all five datasets analyzed. All these genes except IL36RN, both form a protein-protein interaction network and have cellular functions associated with constitutive processes of keratinocytes. Thus, they were generically termed Epidermal Development and Cornification (EDC) genes. The differential expression of these genes in primary and metastatic melanoma was confirmed in the TCGA-SKCM cohort. High expression of the EDC genes correlated with reduced tumor thickness in primary melanoma and shorter survival in metastatic melanoma. Analysis of DEGs from primary melanoma patients displaying high or low expression of all eight EDC revealed that the upregulated genes are enriched in biological process related to cell migration, extracellular matrix organization, invasion, and Epithelial-Mesenchymal Transition. Further analysis of enriched curated oncogenic genesets together with RPPA data of phosphorylated proteins revealed the activation of MEK, ATF2, and EGFR pathways in tumors displaying high expression of EDC genes. Thus, EDC genes may contribute to melanoma progression by promoting the activation of MEK, ATF2, and EGFR pathways together with biological processes associated with tumor aggressiveness.


Sujet(s)
Mélanome , Tumeurs cutanées , Biologie informatique , Récepteurs ErbB/génétique , Régulation de l'expression des gènes tumoraux , Humains , Interleukines/métabolisme , Mélanome/génétique , Mélanome/anatomopathologie , Mitogen-Activated Protein Kinase Kinases/génétique , Mitogen-Activated Protein Kinase Kinases/métabolisme , Tumeurs cutanées/génétique , Tumeurs cutanées/anatomopathologie
6.
Cell Mol Biol Lett ; 27(1): 23, 2022 Mar 08.
Article de Anglais | MEDLINE | ID: mdl-35260073

RÉSUMÉ

BACKGROUND: ROR2 is a tyrosine-kinase receptor whose expression is dysregulated in many human diseases. In cancer, ROR2 stimulates proliferation, survival, migration, and metastasis, and is associated with more aggressive tumor stages. The purpose of this work is to study the role of ROR2 in the chemoresistance of melanoma. METHODS: Gain- and loss-of-function experiments were used to study the biological function of ROR2 in melanoma. Cell death induced by chemotherapeutic drugs and BH-3 mimetics was evaluated using crystal violet cytotoxicity assays and annexin V/propidium iodide staining. Western blots were used to evaluate the expression of proteins implicated in cell death. The differences observed between cells with manipulation of ROR2 levels and control cells were evaluated using both Student's t-test and ANOVA. RESULTS: We describe that ROR2 contributes to tumor progression by enhancing the resistance of melanoma cells to both chemotherapeutic drugs and BH-3 mimetics. We demonstrate that ROR2 reduced cell death upon treatment with cisplatin, dacarbazine, lomustine, camptothecin, paclitaxel, ABT-737, TW-37, and venetoclax. This effect was mediated by the inhibition of apoptosis. In addition, we investigated the molecular mechanisms implicated in this role of ROR2. We identified the MDM2/p53 pathway as a novel target of ROR2 since ROR2 positively regulates MDM2 levels, thus leading to p53 downregulation. We also showed that ROR2 also upregulates Mcl-1 and Bcl2-xL while it negatively regulates Bax and Bid expression. The effect of ROR2 on the expression of these proteins is mediated by the hyperactivation of ERK. CONCLUSIONS: These results demonstrate that ROR2 contributes to melanoma progression by inhibiting apoptosis and increasing chemoresistance. These results not only position ROR2 as a marker of chemoresistance but also support its use as a novel therapeutic target in cancer.


Sujet(s)
Extracellular Signal-Regulated MAP Kinases , Mélanome , Protéines proto-oncogènes c-bcl-2 , Récepteurs orphelins de type récepteur à tyrosine kinase , Protéine p53 suppresseur de tumeur , Apoptose , Lignée cellulaire tumorale , Cisplatine/pharmacologie , Résistance aux médicaments antinéoplasiques , Extracellular Signal-Regulated MAP Kinases/métabolisme , Humains , Mélanome/traitement médicamenteux , Protéines proto-oncogènes c-bcl-2/métabolisme , Récepteurs orphelins de type récepteur à tyrosine kinase/génétique , Protéine p53 suppresseur de tumeur/génétique , Protéine p53 suppresseur de tumeur/métabolisme
7.
Crit Rev Oncol Hematol ; 170: 103595, 2022 Feb.
Article de Anglais | MEDLINE | ID: mdl-35032666

RÉSUMÉ

ROR1 and ROR2 are Wnt receptors that are critical for ß-catenin-independent Wnt pathways and have been linked to processes driving tumor progression, such as cell proliferation, survival, invasion, and therapy resistance. Both receptors have garnered interest as potential therapeutic targets since they are largely absent in adult tissue, are overexpressed in several cancers, and, as members of the receptor tyrosine kinase family, are easier to target than all other components of the pathway. Unlike ROR1 which always promotes tumorigenesis, ROR2 has a very complex role in cancer acting either to promote or inhibit tumor progression in different tumor types. In the present article, we summarize the findings on ROR2 expression in cancer patients and its impact on clinical outcome. Further, we review the biological processes and signaling pathways regulated by ROR2 that explain its dual role in cancer. Finally, we describe the ongoing strategies to target ROR2 in cancer.


Sujet(s)
Récepteurs orphelins de type récepteur à tyrosine kinase , Voie de signalisation Wnt , Carcinogenèse/génétique , Lignée cellulaire tumorale , Prolifération cellulaire , Humains , Récepteurs orphelins de type récepteur à tyrosine kinase/génétique , Récepteurs orphelins de type récepteur à tyrosine kinase/métabolisme
8.
J Biomed Sci ; 28(1): 76, 2021 Nov 13.
Article de Anglais | MEDLINE | ID: mdl-34774050

RÉSUMÉ

BACKGROUND: Receptor tyrosine kinase-like orphan receptor 2 (ROR2) is a Wnt5a receptor aberrantly expressed in cancer that was shown to either suppress or promote carcinogenesis in different tumor types. Our goal was to study the role of ROR2 in melanoma. METHODS: Gain and loss-of-function strategies were applied to study the biological function of ROR2 in melanoma. Proliferation assays, flow cytometry, and western blotting were used to evaluate cell proliferation and changes in expression levels of cell-cycle and proliferation markers. The role of ROR2 in tumor growth was assessed in xenotransplantation experiments followed by immunohistochemistry analysis of the tumors. The role of ROR2 in melanoma patients was assessed by analysis of clinical data from the Leeds Melanoma Cohort. RESULTS: Unlike previous findings describing ROR2 as an oncogene in melanoma, we describe that ROR2 prevents tumor growth by inhibiting cell-cycle progression and the proliferation of melanoma cells. The effect of ROR2 is mediated by inhibition of Akt phosphorylation and activity which, in turn, regulates the expression, phosphorylation, and localization of major cell-cycle regulators including cyclins (A, B, D, and E), CDK1, CDK4, RB, p21, and p27. Xenotransplantation experiments demonstrated that ROR2 also reduces proliferation in vivo, resulting in inhibition of tumor growth. In agreement with these findings, a higher ROR2 level favors thin and non-ulcerated primary melanomas with reduced mitotic rate and better prognosis. CONCLUSION: We conclude that the expression of ROR2 slows down the growth of primary tumors and contributes to prolonging melanoma survival. Our results demonstrate that ROR2 has a far more complex role than originally described.


Sujet(s)
Cycle cellulaire , Prolifération cellulaire , Mélanome/génétique , Protéines proto-oncogènes c-akt/génétique , Récepteurs orphelins de type récepteur à tyrosine kinase/génétique , Protéines proto-oncogènes c-akt/métabolisme
9.
Cancer Metastasis Rev ; 39(3): 933-952, 2020 09.
Article de Anglais | MEDLINE | ID: mdl-32435939

RÉSUMÉ

Wnt5a is the prototypical activator of the non-canonical Wnt pathways, and its overexpression has been implicated in the progression of several tumor types by promoting cell motility, invasion, EMT, and metastasis. Recent evidences have revealed a novel role of Wnt5a in the phosphorylation of the NF-κB subunit p65 and the activation of the NF-κB pathway in cancer cells. In this article, we review the molecular mechanisms and mediators defining a Wnt5a/NF-κB signaling pathway and propose that the aberrant expression of Wnt5a in some tumors drives a Wnt5a/NF-κB/IL-6/STAT3 positive feedback loop that amplifies the effects of Wnt5a. The evidences discussed here suggest that Wnt5a has a double effect on the tumor microenvironment. First, it activates an autocrine ROR1/Akt/p65 pathway that promotes inflammation and chemotaxis of immune cells. Then, Wnt5a activates a TLR/MyD88/p50 pathway exclusively in myelomonocytic cells promoting the synthesis of the anti-inflammatory cytokine IL-10 and a tolerogenic phenotype. As a result of these mechanisms, Wnt5a plays a negative role on immune cell function that contributes to an immunosuppressive tumor microenvironment and would contribute to resistance to immunotherapy. Finally, we summarized the development of different strategies targeting either Wnt5a or the Wnt5a receptor ROR1 that can be helpful for cancer therapy by contributing to generate a more immunostimulatory tumor microenvironment.


Sujet(s)
Tumeurs/immunologie , Voie de signalisation Wnt/immunologie , Protéine Wnt-5a/immunologie , Animaux , Humains , Tolérance immunitaire , Inflammation/immunologie , Inflammation/métabolisme , Inflammation/anatomopathologie , Facteur de transcription NF-kappa B/immunologie , Facteur de transcription NF-kappa B/métabolisme , Métastase tumorale , Tumeurs/métabolisme , Tumeurs/anatomopathologie , Microenvironnement tumoral/immunologie , Protéine Wnt-5a/métabolisme
10.
Cancers (Basel) ; 13(1)2020 Dec 30.
Article de Anglais | MEDLINE | ID: mdl-33396645

RÉSUMÉ

The anti-apoptotic proteins from the Bcl-2 family are important therapeutic targets since they convey resistance to anticancer regimens. Despite the suspected functional redundancy among the six proteins of this subfamily, both basic studies and therapeutic approaches have focused mainly on BCL2, Bcl-xL, and MCL1. The role of BCL2L10, another member of this group, has been poorly studied in cancer and never has been in melanoma. We describe here that BCL2L10 is abundantly and frequently expressed both in melanoma cell lines and tumor samples. We established that BCL2L10 expression is driven by STAT3-mediated transcription, and by using reporter assays, site-directed mutagenesis, and ChIP analysis, we identified the functional STAT3 responsive elements in the BCL2L10 promoter. BCL2L10 is a pro-survival factor in melanoma since its expression reduced the cytotoxic effects of cisplatin, dacarbazine, and ABT-737 (a BCL2, Bcl-xL, and Bcl-w inhibitor). Meanwhile, both genetic and pharmacological inhibition of BCL2L10 sensitized melanoma cells to cisplatin and ABT-737. Finally, BCL2L10 inhibited the cell death upon combination treatments of PLX-4032, a BRAF inhibitor, with ABT-737 or cisplatin. In summary, we determined that BCL2L10 is expressed in melanoma and contributes to cell survival. Hence, targeting BCL2L10 may enhance the clinical efficacy of other therapies for malignant melanoma.

11.
Cells ; 8(9)2019 09 10.
Article de Anglais | MEDLINE | ID: mdl-31510045

RÉSUMÉ

Wnt5a signaling has been implicated in the progression of cancer by regulating multiple cellular processes, largely migration and invasion, epithelial-mesenchymal transition (EMT), and metastasis. Since Wnt5a signaling has also been involved in inflammatory processes in infectious and inflammatory diseases, we addressed the role of Wnt5a in regulating NF-κB, a pivotal mediator of inflammatory responses, in the context of cancer. The treatment of melanoma cells with Wnt5a induced phosphorylation of the NF-κB subunit p65 as well as IKK phosphorylation and IκB degradation. By using cDNA overexpression, RNA interference, and dominant negative mutants we determined that ROR1, Dvl2, and Akt (from the Wnt5a pathway) and TRAF2 and RIP (from the NF-κB pathway) are required for the Wnt5a/NF-κB crosstalk. Wnt5a also induced p65 nuclear translocation and increased NF-κB activity as evidenced by reporter assays and a NF-κB-specific upregulation of RelB, Bcl-2, and Cyclin D1. Further, stimulation of melanoma cells with Wnt5a increased the secretion of cytokines and chemokines, including IL-6, IL-8, IL-11, and IL-6 soluble receptor, MCP-1, and TNF soluble receptor I. The inhibition of endogenous Wnt5a demonstrated that an autocrine Wnt5a loop is a major regulator of the NF-κB pathway in melanoma. Taken together, these results indicate that Wnt5a activates the NF-κB pathway and has an immunomodulatory effect on melanoma through the secretion of cytokines and chemokines.


Sujet(s)
Mélanome/métabolisme , Facteur de transcription NF-kappa B/métabolisme , Protéine Wnt-5a/métabolisme , Communication autocrine , Lignée cellulaire tumorale , Chimiokines/métabolisme , Cytokines/métabolisme , Protéines Dishevelled/métabolisme , Humains , I-kappa B Kinase/métabolisme , Protéines I-kappa B/métabolisme , Facteur de transcription NF-kappa B/antagonistes et inhibiteurs , Phosphorylation , Protéines proto-oncogènes c-akt/métabolisme , Récepteurs orphelins de type récepteur à tyrosine kinase/métabolisme , Transduction du signal , Facteur-2 associé aux récepteurs de TNF/métabolisme , Facteur de transcription RelA/métabolisme
12.
Cell Biosci ; 9: 3, 2019.
Article de Anglais | MEDLINE | ID: mdl-30622697

RÉSUMÉ

BACKGROUND: The PI3K/Akt and the STAT3 pathways are functionally associated in many tumor types. Both in vitro and in vivo studies have revealed that either biochemical or genetic manipulation of the STAT3 pathway activity induce changes in the same direction in Akt activity. However, the implicated mechanism has been poorly characterized. Our goal was to characterize the precise mechanism linking STAT3 with the activity of Akt and other AGC kinases in cancer using melanoma cells as a model. RESULTS: We show that active STAT3 is constitutively bound to the PDK1 promoter and positively regulate PDK1 transcription through two STAT3 responsive elements. Transduction of WM9 and UACC903 melanoma cells with STAT3-small hairpin RNA decreased both PDK1 mRNA and protein levels. STAT3 knockdown also induced a decrease of the phosphorylation of AGC kinases Akt, PKC, and SGK. The inhibitory effect of STAT3 silencing on Akt phosphorylation was restored by HA-PDK1. Along this line, HA-PDK1 expression significantly blocked the cell death induced by dacarbazine plus STAT3 knockdown. This effect might be mediated by Bcl2 proteins since HA-PDK1 rescued Bcl2, Bcl-XL, and Mcl1 levels that were down-regulated upon STAT3 silencing. CONCLUSIONS: We show that PDK1 is a transcriptional target of STAT3, linking STAT3 pathway with AGC kinases activity in melanoma. These data provide further rationale for the ongoing effort to therapeutically target STAT3 and PDK1 in melanoma and, possibly, other malignancies.

13.
Mol Cell Biol ; 36(2): 320-9, 2016 01 15.
Article de Anglais | MEDLINE | ID: mdl-26552704

RÉSUMÉ

Sphingosine-1-phosphate (S1P) is a bioactive lipid mediator that regulates many processes in inflammation and cancer. S1P is a ligand for five G-protein-coupled receptors, S1PR1 to -5, and also has important intracellular actions. Previously, we showed that intracellular S1P is involved in tumor necrosis factor alpha (TNF)-induced NF-κB activation in melanoma cell lines that express filamin A (FLNA). Here, we show that extracellular S1P activates NF-κB only in melanoma cells that lack FLNA. In these cells, S1P, but not TNF, promotes IκB kinase (IKK) and p65 phosphorylation, IκBα degradation, p65 nuclear translocation, and NF-κB reporter activity. NF-κB activation induced by S1P was mediated via S1PR1 and S1PR2. Exogenous S1P enhanced the phosphorylation of protein kinase Cδ (PKCδ), and its downregulation reduced S1P-induced the phosphorylation of IKK and p65. In addition, silencing of Bcl10 also inhibited S1P-induced IKK phosphorylation. Surprisingly, S1P reduced Akt activation in melanoma cells that express FLNA, whereas in the absence of FLNA, high phosphorylation levels of Akt were maintained, enabling S1P-mediated NF-κB signaling. In accord, inhibition of Akt suppressed S1P-mediated IKK and p65 phosphorylation and degradation of IκBα. Hence, these results support a negative role of FLNA in S1P-mediated NF-κB activation in melanoma cells through modulation of Akt.


Sujet(s)
Filamines/immunologie , Lysophospholipides/immunologie , Mélanome/immunologie , Facteur de transcription NF-kappa B/immunologie , Protéines proto-oncogènes c-akt/immunologie , Transduction du signal , Sphingosine/analogues et dérivés , Lignée cellulaire tumorale , Humains , Protein kinase C-delta/immunologie , Récepteurs aux lysosphingolipides/immunologie , Sphingosine/immunologie , Récepteurs de la sphingosine-1-phosphate
14.
Mol Carcinog ; 55(11): 1772-1785, 2016 Nov.
Article de Anglais | MEDLINE | ID: mdl-26509654

RÉSUMÉ

The Receptor tyrosine kinase-like Orphan Receptor 1 (ROR1) is primarily expressed by neural crest cells during embryogenesis. Following a complete downregulation after birth, ROR1 was shown to re-express in various types of cancers. Little is known about ROR1 expression and function in melanoma. Here we show that ROR1 is aberrantly expressed in both melanoma cell lines and tumors and that its expression associates with poor Post-Recurrence Survival of melanoma. Using gain- and loss-of-function approaches we found that ROR1 enhances both anchorage-dependent and -independent growth of melanoma cells. In addition, ROR1 decreases cell adhesion and increases cell motility and migration. Mechanistically, ROR1 was found to induce upregulation of Akt and the mesenquimal markers N-cadherin and vimentin. The regulation of N-cadherin by ROR1 relies on both Akt dependent and independent mechanisms. ROR1 does not affect Wnt canonical pathway but was found to be engaged in a positive feedback loop with Wnt5a. In summary, we show that ROR1 contributes to melanoma progression and is a candidate biomarker of poor prognosis. Although further studies are needed to confirm this possibility, the present work indicates that ROR1 is a good prospective target for melanoma cancer therapy. © 2015 Wiley Periodicals, Inc.


Sujet(s)
Cadhérines/métabolisme , Mélanome/anatomopathologie , Phosphatidylinositol 3-kinases/métabolisme , Protéines proto-oncogènes c-akt/métabolisme , Récepteurs orphelins de type récepteur à tyrosine kinase/génétique , Récepteurs orphelins de type récepteur à tyrosine kinase/métabolisme , Adhérence cellulaire , Lignée cellulaire tumorale , Mouvement cellulaire , Prolifération cellulaire , Régulation de l'expression des gènes tumoraux , Humains , Mélanome/génétique , Mélanome/métabolisme , Études prospectives , Transduction du signal , Analyse de survie
15.
Pigment Cell Melanoma Res ; 24(5): 902-21, 2011 Oct.
Article de Anglais | MEDLINE | ID: mdl-21914141

RÉSUMÉ

Recent discoveries have increased our comprehension of the molecular signaling events critical for melanoma development and progression. Many oncogenes driving melanoma have been identified, and most of them exert their oncogenic effects through the activation of the RAF/MEK/ERK mitogen-activated protein kinase (MAPK) pathway. The c-Jun N-terminal kinase (JNK) and p38 MAPK pathways are also important in melanoma, but their precise role is not clear yet. This review summarizes our current knowledge on the role of the three main MAPK pathways, extracellular regulated kinase (ERK), JNK, and p38, and their impact on melanoma biology. Although the results obtained with BRAF inhibitors in melanoma patients are impressive, several mechanisms of acquired resistance have emerged. To overcome this obstacle constitutes the new challenge in melanoma therapy. Given the major role that MAPKs play in melanoma, understanding their functions and the interconnection among them and with other signaling pathways represents a step forward toward this goal.


Sujet(s)
Système de signalisation des MAP kinases , Mélanome/enzymologie , Mélanome/anatomopathologie , Mitogen-Activated Protein Kinases/métabolisme , Stress physiologique , Humains
16.
Nat Rev Cancer ; 10(1): 65-76, 2010 01.
Article de Anglais | MEDLINE | ID: mdl-20029425

RÉSUMÉ

Cooperation among transcription factors is central for their ability to execute specific transcriptional programmes. The AP1 complex exemplifies a network of transcription factors that function in unison under normal circumstances and during the course of tumour development and progression. This Perspective summarizes our current understanding of the changes in members of the AP1 complex and the role of ATF2 as part of this complex in tumorigenesis.


Sujet(s)
Facteur de transcription ATF-2/métabolisme , Réseaux de régulation génique , Tumeurs/métabolisme , Facteur de transcription AP-1/métabolisme , Facteur de transcription ATF-2/génétique , Animaux , Humains , Famille multigénique , Tumeurs/génétique , Liaison aux protéines , Protéines proto-oncogènes c-jun/génétique , Protéines proto-oncogènes c-jun/métabolisme , Facteur de transcription AP-1/génétique
17.
Biochem Biophys Res Commun ; 301(4): 819-24, 2003 Feb 21.
Article de Anglais | MEDLINE | ID: mdl-12589786

RÉSUMÉ

The variable genes of monoclonal antibody (mAb) B10, specific for the C-terminal region of the eukaryotic ribosomal P protein, have been cloned as a single-chain Fv fragment (scFv) and expressed in Escherichia coli. The primary sequence of the variable regions of the B10 antibody, together with a detailed characterization of the reactive residues of the antigen, allowed the construction of a model of the paratope-epitope interaction, giving a first insight into the binding mechanisms of anti-P autoantibodies to their target peptides. The mAb and scFv could be useful for extensive P protein detection since both recognize the highly conserved motif DDxGF.


Sujet(s)
Région variable d'immunoglobuline/génétique , Protéines ribosomiques/immunologie , Motifs d'acides aminés , Séquence d'acides aminés , Animaux , Anticorps monoclonaux/composition chimique , Anticorps monoclonaux/génétique , Sites de fixation des anticorps/génétique , ADN/génétique , Gènes d'immunoglobuline , Région variable d'immunoglobuline/composition chimique , Souris , Modèles moléculaires , Données de séquences moléculaires , Protéines de protozoaire/immunologie , Similitude de séquences d'acides aminés , Trypanosoma cruzi/immunologie
18.
Rev. argent. cardiol ; 66(2): 127-37, mar.-abr. 1998. graf
Article de Espagnol | LILACS | ID: lil-224560

RÉSUMÉ

En la búsqueda de marcadores serológicos de daño miocárdico activo en pacientes con infección crónica chagásica, se demuestra que el suero de aquéllos que presentan cardiomiopatía manifiesta reconoce las regiones C-terminales de las proteínas ribosomales P del Trypanosoma cruzi. En este estudio se demuestra que la infección no induce la respuesta autoinmune anti P que caracteriza a los pacientes con lupus, sino origina una respuesta anti P característica, expresada por los niveles elevados de anticuerpos contra las regiones polianiónicas presentes en dichas proteínas. Los anticuerpos anti P chagásicos son responsables del efecto estimulante de la IgG de los pacientes con cardiomiopatía chagásica crónica sobre los receptores ß1 adrenérgicos cardíacos, razón por la cual podrían participar en la patogenia de algunas manifestaciones de la cardiomiopatía chagásica crónica, en particular en las arritmias ventriculares. Los resultados de este trabajo indican que la actividad funcional (y patogenicidad) sobre el tejido miocárdico de los anticuerpos generados contra antígenos intracelulares del parásito resultan de su capacidad para reaccionar en forma cruzada con proteínas de la membrana celular cardíaca. Esta demostración obliga a replantear la utilización de quimioterápicos tripanomicidas eficientes y tolerados a largo plazo para anular la producción de esos anticuerpos


Sujet(s)
Humains , Anticorps , Lupus érythémateux disséminé , Cardiomyopathie associée à la maladie de Chagas , Récepteurs bêta-1 adrénergiques , Marqueurs biologiques/analyse , Protéines ribosomiques , Trypanocides/administration et posologie
19.
Rev. argent. cardiol ; 66(2): 127-37, mar.-abr. 1998. graf
Article de Espagnol | BINACIS | ID: bin-17227

RÉSUMÉ

En la búsqueda de marcadores serológicos de daño miocárdico activo en pacientes con infección crónica chagásica, se demuestra que el suero de aquéllos que presentan cardiomiopatía manifiesta reconoce las regiones C-terminales de las proteínas ribosomales P del Trypanosoma cruzi. En este estudio se demuestra que la infección no induce la respuesta autoinmune anti P que caracteriza a los pacientes con lupus, sino origina una respuesta anti P característica, expresada por los niveles elevados de anticuerpos contra las regiones polianiónicas presentes en dichas proteínas. Los anticuerpos anti P chagásicos son responsables del efecto estimulante de la IgG de los pacientes con cardiomiopatía chagásica crónica sobre los receptores ß1 adrenérgicos cardíacos, razón por la cual podrían participar en la patogenia de algunas manifestaciones de la cardiomiopatía chagásica crónica, en particular en las arritmias ventriculares. Los resultados de este trabajo indican que la actividad funcional (y patogenicidad) sobre el tejido miocárdico de los anticuerpos generados contra antígenos intracelulares del parásito resultan de su capacidad para reaccionar en forma cruzada con proteínas de la membrana celular cardíaca. Esta demostración obliga a replantear la utilización de quimioterápicos tripanomicidas eficientes y tolerados a largo plazo para anular la producción de esos anticuerpos (AU)


Sujet(s)
Humains , Anticorps , Cardiomyopathie associée à la maladie de Chagas , Récepteurs bêta-1 adrénergiques , Lupus érythémateux disséminé , Protéines ribosomiques , Marqueurs biologiques/analyse , Trypanocides/administration et posologie
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE