Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 27
Filtrar
Más filtros










Intervalo de año de publicación
1.
Cells ; 12(3)2023 01 29.
Artículo en Inglés | MEDLINE | ID: mdl-36766786

RESUMEN

Triple negative breast cancer (TNBC) is an aggressive breast cancer subtype for which no effective targeted therapies are available. Growing evidence suggests that chemotherapy-resistant cancer cells with stem-like properties (CSC) may repopulate the tumor. The androgen receptor (AR) is expressed in up to 50% of TNBCs, and AR inhibition decreases CSC and tumor initiation. Runt-related transcription factor 1 (RUNX1) correlates with poor prognosis in TNBC and is regulated by the AR in prostate cancer. Our group has shown that RUNX1 promotes TNBC cell migration and regulates tumor gene expression. We hypothesized that RUNX1 is regulated by the AR and that both may work together in TNBC CSC to promote disease recurrence following chemotherapy. Chromatin immunoprecipitation sequencing (ChIP-seq) experiments in MDA-MB-453 revealed AR binding to RUNX1 regulatory regions. RUNX1 expression is upregulated by dihydrotestosterone (DHT) in MDA-MB-453 and in an AR+-TNBC HCI-009 patient-derived xenograft (PDX) tumors (p < 0.05). RUNX1 is increased in a CSC-like experimental model in MDA-MB-453 and SUM-159PT cells (p < 0.05). Inhibition of RUNX1 transcriptional activity reduced the expression of CSC markers. Interestingly, RUNX1 inhibition reduced cell viability and enhanced paclitaxel and enzalutamide sensitivity. Targeting RUNX1 may be an attractive strategy to potentiate the anti-tumor effects of AR inhibition, specifically in the slow-growing CSC-like populations that resist chemotherapy which lead to metastatic disease.


Asunto(s)
Neoplasias de la Mama Triple Negativas , Humanos , Línea Celular Tumoral , Subunidad alfa 2 del Factor de Unión al Sitio Principal/genética , Recurrencia Local de Neoplasia , Receptores Androgénicos/metabolismo , Neoplasias de la Mama Triple Negativas/tratamiento farmacológico , Neoplasias de la Mama Triple Negativas/genética , Neoplasias de la Mama Triple Negativas/patología , Femenino
2.
Oncogene ; 40(43): 6195-6209, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-34545187

RESUMEN

Canonical Wnt/ß-catenin signaling is an established regulator of cellular state and its critical contributions to tumor initiation, malignant tumor progression and metastasis formation have been demonstrated in various cancer types. Here, we investigated how the binding of ß-catenin to the transcriptional coactivators B-cell CLL/lymphoma 9 (Bcl9) and Bcl9-Like (Bcl9L) affected mammary gland carcinogenesis in the MMTV-PyMT transgenic mouse model of metastatic breast cancer. Conditional knockout of both Bcl9 and Bcl9L resulted into tumor cell death. In contrast, disrupting the interaction of Bcl9/Bcl9L with ß-catenin, either by deletion of their HD2 domains or by a point mutation in the N-terminal domain of ß-catenin (D164A), diminished primary tumor growth and tumor cell proliferation and reduced tumor cell invasion and lung metastasis. In comparison, the disruption of HD1 domain-mediated binding of Bcl9/Bcl9L to Pygopus had only moderate effects. Interestingly, interfering with the ß-catenin-Bcl9/Bcl9L-Pygo chain of adapters only partially impaired the transcriptional response of mammary tumor cells to Wnt3a and TGFß treatments. Together, the results indicate that Bcl9/Bcl9L modulate but are not critically required for canonical Wnt signaling in its contribution to breast cancer growth and malignant progression, a notion consistent with the "just-right" hypothesis of Wnt-driven tumor progression.


Asunto(s)
Neoplasias de la Mama/patología , Proteínas de Unión al ADN/metabolismo , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Factores de Transcripción/metabolismo , beta Catenina/metabolismo , Animales , Neoplasias de la Mama/genética , Neoplasias de la Mama/metabolismo , Línea Celular Tumoral , Proteínas de Unión al ADN/genética , Progresión de la Enfermedad , Transición Epitelial-Mesenquimal , Femenino , Humanos , Ratones , Invasividad Neoplásica , Metástasis de la Neoplasia , Trasplante de Neoplasias , Factores de Transcripción/genética , Vía de Señalización Wnt , beta Catenina/genética
3.
Proc Natl Acad Sci U S A ; 118(34)2021 08 24.
Artículo en Inglés | MEDLINE | ID: mdl-34408016

RESUMEN

During malignant progression, epithelial cancer cells dissolve their cell-cell adhesion and gain invasive features. By virtue of its dual function, ß-catenin contributes to cadherin-mediated cell-cell adhesion, and it determines the transcriptional output of Wnt signaling: via its N terminus, it recruits the signaling coactivators Bcl9 and Pygopus, and via the C terminus, it interacts with the general transcriptional machinery. This duality confounds the simple loss-of-function analysis of Wnt signaling in cancer progression. In many cancer types including breast cancer, the functional contribution of ß-catenin's transcriptional activities, as compared to its adhesion functions, to tumor progression has remained elusive. Employing the mouse mammary tumor virus (MMTV)-PyMT mouse model of metastatic breast cancer, we compared the complete elimination of ß-catenin with the specific ablation of its signaling outputs in mammary tumor cells. Notably, the complete lack of ß-catenin resulted in massive apoptosis of mammary tumor cells. In contrast, the loss of ß-catenin's transcriptional activity resulted in a reduction of primary tumor growth, tumor invasion, and metastasis formation in vivo. These phenotypic changes were reflected by stalled cell cycle progression and diminished epithelial-mesenchymal transition (EMT) and cell migration of breast cancer cells in vitro. Transcriptome analysis revealed subsets of genes which were specifically regulated by ß-catenin's transcriptional activities upon stimulation with Wnt3a or during TGF-ß-induced EMT. Our results uncouple the signaling from the adhesion function of ß-catenin and underline the importance of Wnt/ß-catenin-dependent transcription in malignant tumor progression of breast cancer.


Asunto(s)
Adhesión Celular/fisiología , Neoplasias Mamarias Animales/metabolismo , Transducción de Señal/fisiología , Proteína Wnt3A/metabolismo , beta Catenina/metabolismo , Animales , Apoptosis , Ciclo Celular , Movimiento Celular , Transición Epitelial-Mesenquimal/efectos de los fármacos , Femenino , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Neoplasias Mamarias Animales/genética , Ratones , Ratones Transgénicos , Invasividad Neoplásica , Metástasis de la Neoplasia , Transcriptoma , Factor de Crecimiento Transformador beta/farmacología , Proteína Wnt3A/genética , beta Catenina/genética
4.
Cancer Res ; 80(17): 3631-3648, 2020 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-32586983

RESUMEN

Pygopus 2 (Pygo2) is a coactivator of Wnt/ß-catenin signaling that can bind bi- or trimethylated lysine 4 of histone-3 (H3K4me2/3) and participate in chromatin reading and writing. It remains unknown whether the Pygo2-H3K4me2/3 association has a functional relevance in breast cancer progression in vivo. To investigate the functional relevance of histone-binding activity of Pygo2 in malignant progression of breast cancer, we generated a knock-in mouse model where binding of Pygo2 to H3K4me2/3 was rendered ineffective. Loss of Pygo2-histone interaction resulted in smaller, differentiated, and less metastatic tumors, due, in part, to decreased canonical Wnt/ß-catenin signaling. RNA- and ATAC-sequencing analyses of tumor-derived cell lines revealed downregulation of TGFß signaling and upregulation of differentiation pathways such as PDGFR signaling. Increased differentiation correlated with a luminal cell fate that could be reversed by inhibition of PDGFR activity. Mechanistically, the Pygo2-histone interaction potentiated Wnt/ß-catenin signaling, in part, by repressing the expression of Wnt signaling antagonists. Furthermore, Pygo2 and ß-catenin regulated the expression of miR-29 family members, which, in turn, repressed PDGFR expression to promote dedifferentiation of wild-type Pygo2 mammary epithelial tumor cells. Collectively, these results demonstrate that the histone binding function of Pygo2 is important for driving dedifferentiation and malignancy of breast tumors, and loss of this binding activates various differentiation pathways that attenuate primary tumor growth and metastasis formation. Interfering with the Pygo2-H3K4me2/3 interaction may therefore serve as an attractive therapeutic target for metastatic breast cancer. SIGNIFICANCE: Pygo2 represents a potential therapeutic target in metastatic breast cancer, as its histone-binding capability promotes ß-catenin-mediated Wnt signaling and transcriptional control in breast cancer cell dedifferentiation, EMT, and metastasis.


Asunto(s)
Desdiferenciación Celular/fisiología , Regulación Neoplásica de la Expresión Génica/fisiología , Histonas/metabolismo , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Neoplasias Mamarias Experimentales/patología , Animales , Progresión de la Enfermedad , Femenino , Técnicas de Sustitución del Gen , Ratones , Ratones Endogámicos C57BL
5.
Cancer Res ; 78(16): 4497-4511, 2018 08 15.
Artículo en Inglés | MEDLINE | ID: mdl-29748375

RESUMEN

R-spondin3 (RSPO3) is a member of a family of secreted proteins that enhance Wnt signaling pathways in diverse processes, including cancer. However, the role of RSPO3 in mammary gland and breast cancer development remains unclear. In this study, we show that RSPO3 is expressed in the basal stem cell-enriched compartment of normal mouse mammary glands but is absent from committed mature luminal cells in which exogenous RSPO3 impairs lactogenic differentiation. RSPO3 knockdown in basal-like mouse mammary tumor cells reduced canonical Wnt signaling, epithelial-to-mesenchymal transition-like features, migration capacity, and tumor formation in vivo Conversely, RSPO3 overexpression, which was associated with some LGR and RUNX factors, highly correlated with the basal-like subtype among patients with breast cancer. Thus, we identified RSPO3 as a novel key modulator of breast cancer development and a potential target for treatment of basal-like breast cancers.Significance: These findings identify RSPO3 as a potential therapetuic target in basal-like breast cancers.Graphical Abstract: http://cancerres.aacrjournals.org/content/canres/78/16/4497/F1.large.jpg Cancer Res; 78(16); 4497-511. ©2018 AACR.


Asunto(s)
Neoplasias de la Mama/genética , Mama/metabolismo , Neoplasias Mamarias Animales/genética , Trombospondinas/genética , Animales , Mama/patología , Neoplasias de la Mama/patología , Diferenciación Celular/genética , Línea Celular Tumoral , Proliferación Celular/genética , Subunidades alfa del Factor de Unión al Sitio Principal/genética , Células Epiteliales/patología , Transición Epitelial-Mesenquimal/genética , Femenino , Regulación Neoplásica de la Expresión Génica , Humanos , Neoplasias Mamarias Animales/patología , Ratones , Receptores Acoplados a Proteínas G/genética , Vía de Señalización Wnt/genética
6.
Oncotarget ; 8(51): 88475-88487, 2017 Oct 24.
Artículo en Inglés | MEDLINE | ID: mdl-29179450

RESUMEN

Angiotensin (Ang) II, the main effector peptide of the renin-angiotensin system, has been implicated in multiple aspects of cancer progression such as proliferation, migration, invasion, angiogenesis and metastasis. Ang-(1-7), is a biologically active heptapeptide, generated predominantly from AngII by the enzymatic activity of angiotensin converting enzyme 2. Previous studies have shown that Ang-(1-7) counterbalances AngII actions in different pathophysiological settings. In this study, we have analysed the impact of Ang-(1-7) on AngII-induced pro-tumorigenic features on normal murine mammary epithelial cells NMuMG and breast cancer cells MDA-MB-231. AngII stimulated the activation of the survival factor AKT in NMuMG cells mainly through the AT1 receptor. This PI3K/AKT pathway activation also promoted epithelial-mesenchymal transition (EMT). Concomitant treatment of NMuMG cells with AngII and Ang-(1-7) completely abolished EMT features induced by AngII. Furthermore, Ang-(1-7) abrogated AngII induced migration and invasion of the MDA-MB-231 cells as well as pro-angiogenic events such as the stimulation of MMP-9 activity and VEGF expression. Together, these results demonstrate for the first time that Ang-(1-7) counteracts tumor aggressive signals stimulated by AngII in breast cancer cells emerging the peptide as a potential therapy to prevent breast cancer progression.

7.
Oncotarget ; 7(6): 6552-65, 2016 Feb 09.
Artículo en Inglés | MEDLINE | ID: mdl-26735887

RESUMEN

Runx1 participation in epithelial mammary cells is still under review. Emerging data indicates that Runx1 could be relevant for breast tumor promotion. However, to date no studies have specifically evaluated the functional contribution of Runx1 to control gene expression in mammary epithelial tumor cells. It has been described that Runx1 activity is defined by protein context interaction. Interestingly, Foxp3 is a breast tumor suppressor gene. Here we show that endogenous Runx1 and Foxp3 physically interact in normal mammary cells and this interaction blocks Runx1 transcriptional activity. Furthermore we demonstrate that Runx1 is able to bind to R-spondin 3 (RSPO3) and Gap Junction protein Alpha 1 (GJA1) promoters. This binding upregulates Rspo3 oncogene expression and downregulates GJA1 tumor suppressor gene expression in a Foxp3-dependent manner. Moreover, reduced Runx1 transcriptional activity decreases tumor cell migration properties. Collectively, these data provide evidence of a new mechanism for breast tumor gene expression regulation, in which Runx1 and Foxp3 physically interact to control mammary epithelial cell gene expression fate. Our work suggests for the first time that Runx1 could be involved in breast tumor progression depending on Foxp3 availability.


Asunto(s)
Adenocarcinoma/metabolismo , Neoplasias de la Mama/metabolismo , Conexina 43/metabolismo , Subunidad alfa 2 del Factor de Unión al Sitio Principal/metabolismo , Factores de Transcripción Forkhead/metabolismo , Regulación Neoplásica de la Expresión Génica , Trombospondinas/metabolismo , Adenocarcinoma/genética , Adenocarcinoma/patología , Animales , Apoptosis , Western Blotting , Neoplasias de la Mama/genética , Neoplasias de la Mama/patología , Movimiento Celular , Proliferación Celular , Inmunoprecipitación de Cromatina , Femenino , Técnica del Anticuerpo Fluorescente , Humanos , Técnicas para Inmunoenzimas , Inmunoprecipitación , Ratones , Ratones Endogámicos BALB C , Microscopía Confocal , Microscopía Fluorescente , Regiones Promotoras Genéticas/genética , Células Tumorales Cultivadas , Cicatrización de Heridas , Ensayos Antitumor por Modelo de Xenoinjerto
8.
J Exp Med ; 209(11): 1985-2000, 2012 Oct 22.
Artículo en Inglés | MEDLINE | ID: mdl-23027923

RESUMEN

Kaposi's sarcoma (KS), a multifocal vascular neoplasm linked to human herpesvirus-8 (HHV-8/KS-associated herpesvirus [KSHV]) infection, is the most common AIDS-associated malignancy. Clinical management of KS has proven to be challenging because of its prevalence in immunosuppressed patients and its unique vascular and inflammatory nature that is sustained by viral and host-derived paracrine-acting factors primarily released under hypoxic conditions. We show that interactions between the regulatory lectin galectin-1 (Gal-1) and specific target N-glycans link tumor hypoxia to neovascularization as part of the pathogenesis of KS. Expression of Gal-1 is found to be a hallmark of human KS but not other vascular pathologies and is directly induced by both KSHV and hypoxia. Interestingly, hypoxia induced Gal-1 through mechanisms that are independent of hypoxia-inducible factor (HIF) 1α and HIF-2α but involved reactive oxygen species-dependent activation of the transcription factor nuclear factor κB. Targeted disruption of Gal-1-N-glycan interactions eliminated hypoxia-driven angiogenesis and suppressed tumorigenesis in vivo. Therapeutic administration of a Gal-1-specific neutralizing mAb attenuated abnormal angiogenesis and promoted tumor regression in mice bearing established KS tumors. Given the active search for HIF-independent mechanisms that serve to couple tumor hypoxia to pathological angiogenesis, our findings provide novel opportunities not only for treating KS patients but also for understanding and managing a variety of solid tumors.


Asunto(s)
Galectina 1/metabolismo , Neovascularización Patológica/metabolismo , Polisacáridos/metabolismo , Sarcoma de Kaposi/metabolismo , Animales , Anticuerpos Monoclonales/farmacología , Anticuerpos Neutralizantes/farmacología , Hipoxia de la Célula , Línea Celular Tumoral , Células Cultivadas , Galectina 1/genética , Galectina 1/inmunología , Regulación Neoplásica de la Expresión Génica , Células HEK293 , Herpesvirus Humano 8/fisiología , Interacciones Huésped-Patógeno , Humanos , Hipoxia , Immunoblotting , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Desnudos , Neovascularización Patológica/genética , Neovascularización Patológica/prevención & control , Unión Proteica/efectos de los fármacos , Interferencia de ARN , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Sarcoma de Kaposi/tratamiento farmacológico , Sarcoma de Kaposi/genética , Ensayos Antitumor por Modelo de Xenoinjerto
9.
Glycobiology ; 22(10): 1374-86, 2012 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-22752006

RESUMEN

Mechanisms accounting for the protection of the fetal semi-allograft from maternal immune cells remain incompletely understood. In previous studies, we showed that galectin-1 (Gal1), an immunoregulatory glycan-binding protein, hierarchically triggers a cascade of tolerogenic events at the mouse fetomaternal interface. Here, we show that Gal1 confers immune privilege to human trophoblast cells through the modulation of a number of regulatory mechanisms. Gal1 was mainly expressed in invasive extravillous trophoblast cells of human first trimester and term placenta in direct contact with maternal tissue. Expression of Gal1 by the human trophoblast cell line JEG-3 was primarily controlled by progesterone and pro-inflammatory cytokines and impaired T-cell responses by limiting T cell viability, suppressing the secretion of Th1-type cytokines and favoring the expansion of CD4(+)CD25(+)FoxP3(+) regulatory T (T(reg)) cells. Targeted inhibition of Gal1 expression through antibody (Ab)-mediated blockade, addition of the specific disaccharide lactose or retroviral-mediated siRNA strategies prevented these immunoregulatory effects. Consistent with a homeostatic role of endogenous Gal1, patients with recurrent pregnancy loss showed considerably lower levels of circulating Gal1 and had higher frequency of anti-Gal1 auto-Abs in their sera compared with fertile women. Thus, endogenous Gal1 confers immune privilege to human trophoblast cells by triggering a broad tolerogenic program with potential implications in threatened pregnancies.


Asunto(s)
Aborto Habitual/inmunología , Galectina 1/inmunología , Trofoblastos/inmunología , Línea Celular , Supervivencia Celular/inmunología , Citocinas/inmunología , Galectina 1/antagonistas & inhibidores , Galectina 1/biosíntesis , Humanos , Progesterona/farmacología , Linfocitos T/citología , Linfocitos T/inmunología , Trofoblastos/citología
10.
J Cell Physiol ; 227(4): 1721-30, 2012 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-21688264

RESUMEN

Glucocorticoids influence post-natal mammary gland development by sequentially controlling cell proliferation, differentiation, and apoptosis. In the mammary gland, it has been demonstrated that glucocorticoid treatment inhibits epithelial apoptosis in post-lactating glands. In this study, our first goal was to identify new glucocorticoid target genes that could be involved in generating this effect. Expression profiling, by microarray analysis, revealed that expression of several cell-cycle control genes was altered by dexamethasone (DEX) treatment after lactation. Importantly, it was determined that not only the exogenous synthetic hormone, but also the endogenous glucocorticoids regulated the expression of these genes. Particularly, we found that the expression of cell cycle inhibitors p21CIP1, p18INK4c, and Atm was differentially regulated by glucocorticoids through the successive stages of mammary gland development. In undifferentiated cells, DEX treatment induced their expression and reduced cell proliferation, while in differentiated cells this hormone repressed expression of those cell cycle inhibitors and promoted survival. Therefore, differentiation status determined the effect of glucocorticoids on mammary cell fate. Particularly, we have determined that p21CIP1 inhibition would mediate the activity of these hormones in differentiated mammary cells because over-expression of this protein blocked DEX-induced apoptosis protection. Together, our data suggest that the multiple roles played by glucocorticoids in mammary gland development and function might be at least partially due to the alternative roles that these hormones play on the expression of cell cycle regulators.


Asunto(s)
Dexametasona/farmacología , Glucocorticoides/farmacología , Glándulas Mamarias Animales/citología , Glándulas Mamarias Animales/efectos de los fármacos , Animales , Proteínas de la Ataxia Telangiectasia Mutada , Ciclo Celular/efectos de los fármacos , Ciclo Celular/genética , Proteínas de Ciclo Celular/genética , Diferenciación Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Inhibidor p18 de las Quinasas Dependientes de la Ciclina/genética , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/genética , Proteínas de Unión al ADN/genética , Células Epiteliales/citología , Células Epiteliales/efectos de los fármacos , Células Epiteliales/metabolismo , Femenino , Regulación del Desarrollo de la Expresión Génica/efectos de los fármacos , Lactancia/efectos de los fármacos , Lactancia/genética , Glándulas Mamarias Animales/crecimiento & desarrollo , Glándulas Mamarias Animales/metabolismo , Ratones , Ratones Endogámicos BALB C , Proteínas Serina-Treonina Quinasas/genética , Proteínas Supresoras de Tumor/genética
11.
BMC Cell Biol ; 10: 55, 2009 Jul 17.
Artículo en Inglés | MEDLINE | ID: mdl-19615079

RESUMEN

BACKGROUND: Shortly after weaning, a complex multi-step process that leads to massive epithelial apoptosis is triggered by tissue local factors in the mouse mammary gland. Several reports have demonstrated the relevance of mechanical stress to induce adaptive responses in different cell types. Interestingly, these signaling pathways also participate in mammary gland involution. Then, it has been suggested that cell stretching caused by milk accumulation after weaning might be the first stimulus that initiates the complete remodeling of the mammary gland. However, no previous report has demonstrated the impact of mechanical stress on mammary cell physiology. To address this issue, we have designed a new practical device that allowed us to evaluate the effects of radial stretching on mammary epithelial cells in culture. RESULTS: We have designed and built a new device to analyze the biological consequences of applying mechanical stress to cells cultured on flexible silicone membranes. Subsequently, a geometrical model that predicted the percentage of radial strain applied to the elastic substrate was developed. By microscopic image analysis, the adjustment of these calculations to the actual strain exerted on the attached cells was verified. The studies described herein were all performed in the HC11 non-tumorigenic mammary epithelial cell line, which was originated from a pregnant BALB/c mouse. In these cells, as previously observed in other tissue types, mechanical stress induced ERK1/2 phosphorylation and c-Fos mRNA and protein expression. In addition, we found that mammary cell stretching triggered involution associated cellular events as Leukemia Inhibitory Factor (LIF) expression induction, STAT3 activation and AKT phosphorylation inhibition. CONCLUSION: Here, we show for the first time, that mechanical strain is able to induce weaning-associated events in cultured mammary epithelial cells. These results were obtained using a new practical and affordable device specifically designed for such a purpose. We believe that our results indicate the relevance of mechanical stress among the early post-lactation events that lead to mammary gland involution.


Asunto(s)
Células Epiteliales/citología , Células Epiteliales/metabolismo , Glándulas Mamarias Animales/citología , Estrés Mecánico , Animales , Línea Celular , Femenino , Expresión Génica , Factor Inhibidor de Leucemia/genética , Factor Inhibidor de Leucemia/metabolismo , Ratones , Ratones Endogámicos BALB C , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Fosforilación , Embarazo , Proteínas Proto-Oncogénicas c-fos/genética , Proteínas Proto-Oncogénicas c-fos/metabolismo , ARN Mensajero/genética , Factor de Transcripción STAT3/metabolismo
12.
Cancer Immunol Immunother ; 56(4): 491-9, 2007 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-16900348

RESUMEN

Tumors escape from immune surveillance by producing immunosuppressive cytokines and proapototic factors, including TGF-beta and galectin-1 (Gal-1). Since immunosuppressive mechanisms might act in concert to confer tumor-immune privilege, we investigated the potential cross talk between TGF-beta and Gal-1 in highly metastatic mammary adenocarcinoma (LM3) cells. While Gal-1 treatment was not capable of regulating TGF-beta synthesis, a pronounced and dose-dependent increase in Gal-1 expression was observed when tumor cells were treated with TGF-beta(1. )This effect was also observed in the murine lung adenocarcinoma LP07 and in the human breast adenocarcinoma MCF-7 cell lines. TGF-beta1-mediated upregulation of Gal-1 expression was specifically mediated by TbetaRI and TbetaRII, since it was abrogated when LM3 cells were infected with retroviral vectors expressing the dominant negative forms of these receptors. In addition, gal-1 gene sequence analysis revealed the presence of three putative binding sites for Smad4 and Smad3 transcription factors, consistent with the ability of TGF-beta(1) to trigger a Smad-dependent signaling pathway in these cells. Thus, TGF-beta(1) may trigger a Smad-dependent pathway to control Gal-1 expression, suggesting that distinct mechanisms might cooperate in tilting the balance toward an immunosuppressive environment at the tumor site.


Asunto(s)
Adenocarcinoma/inmunología , Galectina 1/biosíntesis , Neoplasias Mamarias Experimentales/inmunología , Factor de Crecimiento Transformador beta1/metabolismo , Escape del Tumor/fisiología , Adenocarcinoma/metabolismo , Animales , Western Blotting , Línea Celular Tumoral , Femenino , Técnica del Anticuerpo Fluorescente , Galectina 1/genética , Galectina 1/inmunología , Humanos , Neoplasias Mamarias Experimentales/metabolismo , Ratones , Receptor Cross-Talk/inmunología , Proteínas Smad/inmunología , Proteínas Smad/metabolismo , Factor de Crecimiento Transformador beta1/inmunología
13.
Medicina (B Aires) ; 66(4): 357-62, 2006.
Artículo en Español | MEDLINE | ID: mdl-16977975

RESUMEN

Recent evidence indicates that protein-glycan interactions play a critical role in different events associated with the physiology of T-cell responses including thymocyte maturation, T-cell activation, lymphocyte migration and T-cell apoptosis. Glycans decorating T-cell surface glycoproteins can modulate T-cell physiology by specifically interacting with endogenous lectins including selectins and galectins. These endogenous lectins are capable of recognizing sugar structures localized on T-cell surface glycoproteins and trigger different signal transduction pathways leading to differentiation, proliferation, cell cycle regulation or apoptosis. Protein-carbohydrate interactions may be controlled at different levels, including regulated expression of lectins during T-cell maturation and differentiation and the spatio-temporal regulation of glycosyltransferases and glycosidases, which create and modify sugar structures present in T-cell surface glycoproteins. This article briefly reviews the mechanisms by which protein-carbohydrate interactions modulate immunological processes such as T-cell activation, migration and apoptosis.


Asunto(s)
Polisacáridos/metabolismo , Proteínas/metabolismo , Linfocitos T/fisiología , Apoptosis , Comunicación Celular , Galectinas/química , Galectinas/inmunología , Galectinas/metabolismo , Glicosilación , Glicosiltransferasas , Humanos , Polisacáridos/química , Polisacáridos/inmunología , Unión Proteica/inmunología , Proteínas/química , Proteínas/inmunología , Selectinas/química , Selectinas/inmunología , Selectinas/metabolismo
14.
Medicina (B.Aires) ; 66(4): 357-362, 2006.
Artículo en Español | LILACS | ID: lil-449006

RESUMEN

Recent evidence indicates that protein-glycan interactions play a critical role in different events associated with the physiology of T-cell responses including thymocyte maturation, T-cell activation, lymphocyte migration and T-cell apoptosis. Glycans decorating T-cell surface glycoproteins can modulate T-cell physiology by specifically interacting with endogenous lectins including selectins and galectins. These endogenous lectins are capable of recognizing sugar structures localized on T-cell surface glycoproteins and trigger different signal transduction pathways leading to differentiation, proliferation, cell cycle regulation or apoptosis. Protein-carbohydrate interactions may be controlled at different levels, including regulated expression of lectins during T-cell maturation and differentiation and the spatio-temporal regulation of glycosyltransferases and glycosidases, which create and modify sugar structures present in T-cell surface glycoproteins. This article briefly reviews the mechanisms by which protein-carbohydrate interactions modulate immunological processes such as T-cell activation, migration and apoptosis.


Las interacciones entre proteínas y glicanos juegan un papel fundamental en numerosos eventos de la regulación de la fisiología del sistema inmune, como maduración tímica, activación, migración y apoptosis de células T. Los carbohidratos son capaces de modular la fisiología linfocitaria a través de la interacción específica con lectinas endógenas como selectinas y galectinas. Estas lectinas endógenas son capaces de reconocer estructuras sacarídicas localizadas en glicoproteínas de la superficie celular y regular procesos tan diversos como proliferación, diferenciación y ciclo celular. Existen diversos niveles de control de la interacción entre lectinas y azúcares; en primer lugar podemos mencionar la expresión regulada de estas lectinas durante el desarrollo de una respuesta inmune, y en segundo lugar la regulación espacio-temporal de la actividad de glicosiltranferasas y glicosidasas cuya función es crear y modificar los azúcares específicos para estas lectinas. Existen evidencias de que la expresión y actividad de estas enzimas se regulan en forma positiva o negativa durante diferentes eventos del desarrollo, ejecución y finalización de la respuesta inmune. En este artículo se analizarán los mecanismos a través de los cuales las interacciones entre lectinas con sus carbohidratos específicos modulan en forma específica diversos procesos fisiológicos, como maduración de timocitos, migración linfocitaria, activación y diferenciación de células T y apoptosis.


Asunto(s)
Humanos , Linfocitos T/fisiología , Polisacáridos/metabolismo , Proteínas/metabolismo , Apoptosis , Comunicación Celular , Glicosilación , Glicosiltransferasas , Galectinas/química , Galectinas/inmunología , Galectinas/metabolismo , Unión Proteica/inmunología , Polisacáridos/química , Polisacáridos/inmunología , Proteínas/química , Proteínas/inmunología , Selectinas/química , Selectinas/inmunología , Selectinas/metabolismo
15.
Medicina (B.Aires) ; 66(4): 357-362, 2006.
Artículo en Español | BINACIS | ID: bin-123215

RESUMEN

Recent evidence indicates that protein-glycan interactions play a critical role in different events associated with the physiology of T-cell responses including thymocyte maturation, T-cell activation, lymphocyte migration and T-cell apoptosis. Glycans decorating T-cell surface glycoproteins can modulate T-cell physiology by specifically interacting with endogenous lectins including selectins and galectins. These endogenous lectins are capable of recognizing sugar structures localized on T-cell surface glycoproteins and trigger different signal transduction pathways leading to differentiation, proliferation, cell cycle regulation or apoptosis. Protein-carbohydrate interactions may be controlled at different levels, including regulated expression of lectins during T-cell maturation and differentiation and the spatio-temporal regulation of glycosyltransferases and glycosidases, which create and modify sugar structures present in T-cell surface glycoproteins. This article briefly reviews the mechanisms by which protein-carbohydrate interactions modulate immunological processes such as T-cell activation, migration and apoptosis.(AU)


Las interacciones entre proteínas y glicanos juegan un papel fundamental en numerosos eventos de la regulación de la fisiología del sistema inmune, como maduración tímica, activación, migración y apoptosis de células T. Los carbohidratos son capaces de modular la fisiología linfocitaria a través de la interacción específica con lectinas endógenas como selectinas y galectinas. Estas lectinas endógenas son capaces de reconocer estructuras sacarídicas localizadas en glicoproteínas de la superficie celular y regular procesos tan diversos como proliferación, diferenciación y ciclo celular. Existen diversos niveles de control de la interacción entre lectinas y azúcares; en primer lugar podemos mencionar la expresión regulada de estas lectinas durante el desarrollo de una respuesta inmune, y en segundo lugar la regulación espacio-temporal de la actividad de glicosiltranferasas y glicosidasas cuya función es crear y modificar los azúcares específicos para estas lectinas. Existen evidencias de que la expresión y actividad de estas enzimas se regulan en forma positiva o negativa durante diferentes eventos del desarrollo, ejecución y finalización de la respuesta inmune. En este artículo se analizarán los mecanismos a través de los cuales las interacciones entre lectinas con sus carbohidratos específicos modulan en forma específica diversos procesos fisiológicos, como maduración de timocitos, migración linfocitaria, activación y diferenciación de células T y apoptosis. (AU)


Asunto(s)
Humanos , Polisacáridos/metabolismo , Proteínas/metabolismo , Linfocitos T/fisiología , Apoptosis , Comunicación Celular , Galectinas/química , Galectinas/inmunología , Galectinas/metabolismo , Glicosilación , Glicosiltransferasas , Polisacáridos/química , Polisacáridos/inmunología , Unión Proteica/inmunología , Proteínas/química , Proteínas/inmunología , Selectinas/química , Selectinas/inmunología , Selectinas/metabolismo
16.
Medicina (B.Aires) ; 66(4): 357-362, 2006.
Artículo en Español | BINACIS | ID: bin-119221

RESUMEN

Recent evidence indicates that protein-glycan interactions play a critical role in different events associated with the physiology of T-cell responses including thymocyte maturation, T-cell activation, lymphocyte migration and T-cell apoptosis. Glycans decorating T-cell surface glycoproteins can modulate T-cell physiology by specifically interacting with endogenous lectins including selectins and galectins. These endogenous lectins are capable of recognizing sugar structures localized on T-cell surface glycoproteins and trigger different signal transduction pathways leading to differentiation, proliferation, cell cycle regulation or apoptosis. Protein-carbohydrate interactions may be controlled at different levels, including regulated expression of lectins during T-cell maturation and differentiation and the spatio-temporal regulation of glycosyltransferases and glycosidases, which create and modify sugar structures present in T-cell surface glycoproteins. This article briefly reviews the mechanisms by which protein-carbohydrate interactions modulate immunological processes such as T-cell activation, migration and apoptosis.(AU)


Las interacciones entre proteínas y glicanos juegan un papel fundamental en numerosos eventos de la regulación de la fisiología del sistema inmune, como maduración tímica, activación, migración y apoptosis de células T. Los carbohidratos son capaces de modular la fisiología linfocitaria a través de la interacción específica con lectinas endógenas como selectinas y galectinas. Estas lectinas endógenas son capaces de reconocer estructuras sacarídicas localizadas en glicoproteínas de la superficie celular y regular procesos tan diversos como proliferación, diferenciación y ciclo celular. Existen diversos niveles de control de la interacción entre lectinas y azúcares; en primer lugar podemos mencionar la expresión regulada de estas lectinas durante el desarrollo de una respuesta inmune, y en segundo lugar la regulación espacio-temporal de la actividad de glicosiltranferasas y glicosidasas cuya función es crear y modificar los azúcares específicos para estas lectinas. Existen evidencias de que la expresión y actividad de estas enzimas se regulan en forma positiva o negativa durante diferentes eventos del desarrollo, ejecución y finalización de la respuesta inmune. En este artículo se analizarán los mecanismos a través de los cuales las interacciones entre lectinas con sus carbohidratos específicos modulan en forma específica diversos procesos fisiológicos, como maduración de timocitos, migración linfocitaria, activación y diferenciación de células T y apoptosis. (AU)


Asunto(s)
Humanos , Polisacáridos/metabolismo , Proteínas/metabolismo , Linfocitos T/fisiología , Apoptosis , Comunicación Celular , Galectinas/química , Galectinas/inmunología , Galectinas/metabolismo , Glicosilación , Glicosiltransferasas , Polisacáridos/química , Polisacáridos/inmunología , Unión Proteica/inmunología , Proteínas/química , Proteínas/inmunología , Selectinas/química , Selectinas/inmunología , Selectinas/metabolismo
17.
Cancer Cell ; 5(3): 241-51, 2004 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-15050916

RESUMEN

Despite the existence of tumor-specific immune cells, most tumors have devised strategies to avoid immune attack. We demonstrate here that galectin-1 (Gal-1), a negative regulator of T cell activation and survival, plays a pivotal role in promoting escape from T cell-dependent immunity, thus conferring immune privilege to tumor cells. Blockade of immunosuppressive Gal-1 in vivo promotes tumor rejection and stimulates the generation of a tumor-specific T cell-mediated response in syngeneic mice, which are then able to resist subsequent challenge with wild-type Gal-1-sufficient tumors. Our data indicate that Gal-1 signaling in activated T cells constitutes an important mechanism of tumor-immune escape and that blockade of this inhibitory signal can allow for and potentiate effective immune responses against tumor cells, with profound implications for cancer immunotherapy.


Asunto(s)
Galectina 1/metabolismo , Regulación Neoplásica de la Expresión Génica/fisiología , Linfocitos T Citotóxicos/metabolismo , Animales , Antígenos CD4/inmunología , Antígenos CD4/metabolismo , Antígenos CD8/inmunología , Antígenos CD8/metabolismo , Supervivencia Celular , Galectina 1/inmunología , Humanos , Melanoma Experimental/inmunología , Melanoma Experimental/metabolismo , Ratones , Microscopía Fluorescente , Transducción de Señal , Linfocitos T Citotóxicos/inmunología , Células Tumorales Cultivadas
18.
Mol Cell Biochem ; 267(1-2): 177-85, 2004 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-15663199

RESUMEN

Recent evidence has implicated galectins and their carbohydrate ligands as novel regulators of T-cell homeostasis. Galectin-1 (Gal-1), a member of this family, inhibits clonal expansion, induces apoptosis of antigen-primed T lymphocytes and suppresses the development of T-cell-mediated autoimmune diseases in vivo. Because the beta-galactoside-binding protein is expressed in activated but not resting T cells, it has been hypothesized that Gal-1-induced apoptosis may constitute an autocrine suicide mechanism to eliminate activated T cells contributing to the termination of an effector immune response. We undertook this study to investigate the signals and intracellular pathways leading to Gal-1 expression during T-cell activation. When T cells were stimulated either with anti-CD3 or anti-CD28 monoclonal antibody plus PMA in the presence of accessory cells, a sustained up-regulation of Gal-1 was observed, reaching a plateau between days 3 and 5 following CD3 engagement or costimulation through CD28. Investigation of the signal transduction events involved in this process revealed a role for Lck and Fyn kinases, since the Src kinase inhibitor PP1 inhibited the up-regulated expression of Gal-1 following T-cell activation. Downstream signaling routes involve mitogen-activated protein kinase (MAPK) kinase (MEK)1/extracellular signal-regulated kinase (ERK) and p38 MAPK, as Gal-1 expression was prevented by U0126 and SB202190. In addition, expression of Gal-1 involves interleukin (IL)-2-dependent signaling routes triggered by p70S6 kinase, as it could be inhibited by rapamycin. This is the first demonstration of the intracellular pathways that control activation-induced expression of Gal-1, which may reveal potential targets for immune intervention to modulate expression of this beta-galactoside-binding protein in pathological disorders.


Asunto(s)
Galectina 1/metabolismo , Regulación de la Expresión Génica/inmunología , Proteína Tirosina Quinasa p56(lck) Específica de Linfocito/metabolismo , MAP Quinasa Quinasa 1/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Proteínas Quinasas S6 Ribosómicas 70-kDa/metabolismo , Linfocitos T/metabolismo , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo , Familia-src Quinasas/metabolismo , Anticuerpos Monoclonales/farmacología , Western Blotting , Butadienos/farmacología , Proliferación Celular , Electroforesis en Gel de Poliacrilamida , Inhibidores Enzimáticos/farmacología , Galectina 1/genética , Humanos , Imidazoles/farmacología , Interleucina-2/metabolismo , Leucocitos Mononucleares/efectos de los fármacos , Activación de Linfocitos , Modelos Biológicos , Nitrilos/farmacología , Proteínas Proto-Oncogénicas c-fyn , Piridinas/farmacología , Transducción de Señal , Sirolimus/farmacología , Acetato de Tetradecanoilforbol/farmacología , Factores de Tiempo , Regulación hacia Arriba
19.
Gastroenterology ; 124(5): 1381-94, 2003 May.
Artículo en Inglés | MEDLINE | ID: mdl-12730878

RESUMEN

BACKGROUND & AIMS: Uncontrolled T-cell activation plays a critical role in the pathogenesis of inflammatory bowel diseases. Therefore, pharmacologic strategies directed to restore the normal responsiveness of the immune system by deleting inappropriately activated T cells could be efficacious in the treatment of these pathologic conditions. Galectin-1 is an endogenous lectin expressed in lymphoid organs that plays a role in the maintenance of central and peripheral tolerance. The aim of the present study was to evaluate the therapeutic effects of galectin-1 on T-helper cell type 1-mediated experimental colitis induced by intrarectal administration of 2,4,6-trinitrobenzene sulfonic acid (TNBS) in mice. METHODS: Cells and tissues from mice with TNBS colitis receiving treatment with several doses of human recombinant galectin-1 (hrGAL-1) were analyzed for morphology, cytokine production, and apoptosis. RESULTS: Prophylactic and therapeutic administration of rhGAL-1 resulted in a striking improvement in the clinical and histopathologic aspects of the disease. hrGAL-1 reduced the number of hapten-activated spleen T cells, decreased inflammatory cytokine production, and profoundly reduced the ability of lamina propria T cells to produce IFN gamma in vitro. Moreover, hrGAL-1 led to the appearance of apoptotic mononuclear cells in colon tissue when administered in vivo and induced selective apoptosis of TNBS-activated lamina propria T cells in vitro. CONCLUSION: Collectively, these data show that hrGAL-1 exerts protective and immunomodulatory activity in TNBS-induced colitis and it might be effective in the treatment of inflammatory bowel diseases.


Asunto(s)
Colitis/tratamiento farmacológico , Galectina 1/farmacología , Animales , Linfocitos T CD4-Positivos/efectos de los fármacos , Linfocitos T CD4-Positivos/metabolismo , Colitis/inmunología , Colitis/patología , Modelos Animales de Enfermedad , Galectina 1/genética , Expresión Génica/fisiología , Interferón gamma/metabolismo , Interleucina-1/metabolismo , Interleucina-12/metabolismo , Ratones , Ratones Endogámicos BALB C , Proteínas Recombinantes/farmacología , Bazo/citología , Ácido Trinitrobencenosulfónico , Factor de Necrosis Tumoral alfa/metabolismo
20.
Biol Reprod ; 68(1): 51-9, 2003 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-12493695

RESUMEN

Galectin-1, a highly conserved beta-galactoside-binding protein, induces apoptosis of activated T cells and suppresses the development of autoimmunity and chronic inflammation. To gain insight regarding the potential role of galectin-1 as a novel mechanism of immune privilege, we investigated expression and ultrastructural localization of galectin-1 in rat testis. Galectin-1 expression was assessed by Western blot analysis and immunocytochemical localization in testes obtained from rats aged from 9 to 60 days. Expression of this carbohydrate-binding protein was developmentally regulated, and its immunolabeling exhibited a stage-specific pattern throughout the spermatogenic process. Immunogold staining using the anti-galectin-1 antibody revealed the typical Sertoli cell profile in the seminiferous epithelium, mainly at stages X-II. During spermiation (stages VI-VIII), a strong labeling was observed at the luminal pole of seminiferous epithelium, localized on apical stalks of Sertoli cells, on heads of mature spermatids, and on bodies of residual cytoplasm. Moreover, spermatozoa released into the lumen showed a strong immunostaining. Following spermiation (stage VIII), galectin-1 expression was restored at the basal portion of Sertoli cells and progressively spread out through the whole cells as differentiation of germinal cells proceeded. Immunoelectron microscopy confirmed distribution of galectin-1 in nuclei and cytoplasmic projections of Sertoli cells and on heads and tails of late spermatids and residual bodies. Surface localization of galectin-1 was evidenced in spermatozoa from caput epididymis. Thus, the regulated expression of galectin-1 during the spermatogenic cycle suggests a novel role for this immunosuppressive lectin in reproductive biology.


Asunto(s)
Galectina 1/metabolismo , Espermatogénesis/fisiología , Testículo/metabolismo , Animales , Apoptosis , Western Blotting , Epidídimo/citología , Epidídimo/metabolismo , Galactósidos/metabolismo , Inmunohistoquímica , Masculino , Microscopía Inmunoelectrónica , Ratas , Ratas Wistar , Espermatozoides/metabolismo , Espermatozoides/ultraestructura , Testículo/crecimiento & desarrollo , Testículo/ultraestructura
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA