Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Semin Cancer Biol ; 97: 42-49, 2023 12.
Artículo en Inglés | MEDLINE | ID: mdl-37926346

RESUMEN

Disruption of oxygen homeostasis, resulting from an imbalance between O2 supply and demand during malignant proliferation, leads to the development of hypoxic tumor microenvironments that promote the acquisition of aggressive cancer cell phenotypes linked to metastasis and patient mortality. In this review, the mechanistic links between tumor hypoxia and metastatic progression are presented. Current status and perspectives of targeting hypoxia signaling pathways as a strategy to halt cancer cell metastatic activities are emphasized.


Asunto(s)
Hipoxia , Hipoxia Tumoral , Humanos , Hipoxia de la Célula
2.
Trends Cancer ; 8(9): 771-784, 2022 09.
Artículo en Inglés | MEDLINE | ID: mdl-35637155

RESUMEN

The lymphatic system is a major component of the tumor microenvironment, wherein lymphovascular remodeling occurs as a response to low O2 (hypoxia), resulting in metastatic dissemination and patient mortality. Further to this notion, recent data have brought forth an expanded view of lymphatics, beyond a 'passthrough' system for cancer cells (CCs) onto an immune effector function, crucially determining targeted (immuno)therapeutic responses. We hereby provide a novel view of how hypoxia-driven mechanisms of lymphatic remodeling and immunotolerance can be actively co-opted by CCs tilting the immunological balance away or in favor of suppressive tumor and metastatic microenvironments. We hypothesize that specific combinatorial approaches targeting hypoxia signaling pathways might be utilized to reverse this imbalance, to amplify responses to targeted immunotherapies for patients with cancer.


Asunto(s)
Neoplasias , Microambiente Tumoral , Hipoxia de la Célula , Humanos , Hipoxia , Inmunoterapia , Neoplasias/metabolismo
3.
EMBO Mol Med ; 12(9): e11416, 2020 09 07.
Artículo en Inglés | MEDLINE | ID: mdl-32686360

RESUMEN

Conventional maximum-tolerated dose (MTD) chemotherapy relies on periodic, massive cancer cell ablation events followed by treatment-free intermissions, stereotypically resulting in resistance, relapse, and mortality. Furthermore, MTD chemotherapy can promote metastatic dissemination via activation of a transcriptional program dependent on hypoxia-inducible factor (HIF)-1α and (HIF)-2α (hereafter referred to as HIFα). Instead, frequent low-dose metronomic (LDM) chemotherapy displays less adverse effects while preserving significant pre-clinical anticancer activity. Consequently, we hereby compared the effect of MTD or LDM chemotherapy upon HIFα in models of advanced, metastatic colon and breast cancer. Our results revealed that LDM chemotherapy could offset paralog-specific, MTD-dependent HIFα induction in colon cancers disseminating to the liver and lungs, while limiting HIFα and hypoxia in breast cancer lung metastases. Moreover, we assessed the translational significance of HIFα activity in colorectal and breast TCGA/microarray data, by developing two compact, 11-gene transcriptomic signatures allowing the stratification/identification of patients likely to benefit from LDM and/or HIFα-targeting therapies. Altogether, these results suggest LDM chemotherapy as a potential maintenance strategy to stave off HIFα induction within the intra-metastatic tumor microenvironment.


Asunto(s)
Neoplasias de la Mama , Neoplasias Pulmonares , Neoplasias de la Mama/tratamiento farmacológico , Femenino , Humanos , Dosis Máxima Tolerada , Recurrencia Local de Neoplasia , Microambiente Tumoral
4.
Cancer Lett ; 487: 74-84, 2020 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-32470491

RESUMEN

Hypoxia is a universal feature of solid cancers caused by a mismatch between cellular oxygen supply and consumption. To meet the increased demand for oxygen, hypoxic cancer cells (CCs) induce a multifaceted process known as angiogenesis, wherein new vessels are formed by the sprouting of pre-existing ones. In addition to providing oxygen for growth and an exit route for dissemination, angiogenic vessels and factors are co-opted by CCs to enable the generation of an immunotolerant, hypoxic tumor microenvironment, leading to therapeutic failure and mortality. In this review, we discuss how hypoxia-inducible factors (HIFs), the mechanistic target of rapamycin (mTOR), and the unfolded protein response (UPR) control angiogenic factors serving both vascular and immunomodulatory functions in the tumor microenvironment. Possible therapeutic strategies, wherein targeting oxygen sensing might enhance anti-angiogenic and immunologically-mediated anti-cancer responses, are suggested.


Asunto(s)
Subunidad alfa del Factor 1 Inducible por Hipoxia/genética , Neoplasias/genética , Serina-Treonina Quinasas TOR/genética , Microambiente Tumoral/genética , Respuesta de Proteína Desplegada/genética , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Vasos Sanguíneos/crecimiento & desarrollo , Vasos Sanguíneos/patología , Regulación Neoplásica de la Expresión Génica/genética , Redes Reguladoras de Genes/genética , Redes Reguladoras de Genes/inmunología , Humanos , Neoplasias/inmunología , Neoplasias/patología , Neovascularización Patológica/genética , Neovascularización Patológica/inmunología , Neovascularización Patológica/patología , Hipoxia Tumoral/genética , Hipoxia Tumoral/inmunología , Microambiente Tumoral/inmunología , Respuesta de Proteína Desplegada/inmunología
5.
Adv Exp Med Biol ; 1136: 71-85, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31201717

RESUMEN

Hypoxia (low O2) is a ubiquitous feature of solid cancers, arising as a mismatch between cellular O2 supply and consumption. Hypoxia is associated to metastatic disease and mortality owing to its ability to stimulate the formation of blood (angiogenesis) and lymphatic vessels (lymphangiogenesis), thereby allowing cancer cells to escape the unfavorable tumor microenvironment and disseminate into secondary sites. This review outlines molecular mechanisms by which intratumoral hypoxia regulates the expression of motogenic and mitogenic factors that induce angiogenesis and lymphangiogenesis, whilst discussing their implications for metastatic cancers.


Asunto(s)
Linfangiogénesis , Vasos Linfáticos , Neoplasias/patología , Hipoxia Tumoral , Humanos , Metástasis Linfática , Neovascularización Patológica , Microambiente Tumoral
6.
Am J Physiol Regul Integr Comp Physiol ; 315(6): R1072-R1084, 2018 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-30183339

RESUMEN

Hypoxia (low O2) is a ubiquitous microenvironmental factor promoting cancer progression, metastasis, and mortality, owing to the ability of cancer cells to co-opt physiological angiogenic responses. Notwithstanding, the pathophysiological induction of angiogenesis results in an abnormal tumor vasculature, further aggravating hypoxia in a feedforward loop that limits the efficacy of molecular targeted therapies. Recent studies suggest that, besides their canonical roles, angiogenic factors promote a panoply of immunosuppressive effects in the tumor microenvironment. Therefore, intratumoral hypoxia emerges as a hitherto unrecognized mechanism evolutionarily repurposing angiogenic molecules as (patho)physiological immunomodulators. On the other hand, antiangiogenic therapies could be aimed at impeding both tumor growth and immunotolerance toward cancer cells, a beneficial effect that can be countered if hypoxia signaling pathways are left unchecked, leading to therapeutic failure. This review summarizes evidence supporting the hypothesis that hypoxia acts as a common pathophysiological mechanism of resistance to immunotherapeutic and antiangiogenic agents while proposing potential strategies to curtail resistance and mortality in patients bearing solid malignancies.


Asunto(s)
Inductores de la Angiogénesis/farmacología , Inhibidores de la Angiogénesis/farmacología , Evasión Inmune , Neovascularización Patológica/inmunología , Microambiente Tumoral/efectos de los fármacos , Animales , Humanos , Hipoxia/metabolismo
7.
Trends Cell Biol ; 28(2): 128-142, 2018 02.
Artículo en Inglés | MEDLINE | ID: mdl-29191366

RESUMEN

Molecular oxygen (O2) is a universal electron acceptor that enables ATP synthesis through mitochondrial respiration in all metazoans. Consequently, hypoxia (low O2) has arisen as an organizing principle for cellular evolution, metabolism, and (patho)biology, eliciting a remarkable panoply of metabolic adaptations that trigger transcriptional, translational, post-translational, and epigenetic responses to determine cellular fitness. In this review we summarize current and emerging cell-autonomous molecular mechanisms that induce hypoxic metabolic reprogramming in health and disease.


Asunto(s)
Reprogramación Celular/fisiología , Metabolismo Energético/fisiología , Oxígeno/metabolismo , Animales , Hipoxia de la Célula/fisiología , Humanos , Transducción de Señal/fisiología
8.
Trends Cancer ; 3(7): 529-541, 2017 07.
Artículo en Inglés | MEDLINE | ID: mdl-28718406

RESUMEN

Hypoxia (low O2) is a pathobiological hallmark of solid cancers, resulting from the imbalance between cellular O2 consumption and availability. Hypoxic cancer cells (CCs) stimulate blood vessel sprouting (angiogenesis), aimed at restoring O2 delivery to the expanding tumor masses through the activation of a transcriptional program mediated by hypoxia-inducible factors (HIFs). Here, we review recent data suggesting that the efficacy of antiangiogenic (AA) therapies is limited in some circumstances by HIF-dependent compensatory responses to increased intratumoral hypoxia. In lieu of this evidence, we discuss the potential of targeting HIFs as a strategy to overcome these instances of AA therapy resistance.


Asunto(s)
Inhibidores de la Angiogénesis/uso terapéutico , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/antagonistas & inhibidores , Neoplasias/tratamiento farmacológico , Neovascularización Patológica/tratamiento farmacológico , Inhibidores de Topoisomerasa/uso terapéutico , Inhibidores de la Angiogénesis/farmacología , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , ADN-Topoisomerasas de Tipo I/metabolismo , Reposicionamiento de Medicamentos/métodos , Reposicionamiento de Medicamentos/tendencias , Resistencia a Antineoplásicos/efectos de los fármacos , Humanos , Hipoxia/patología , Terapia Molecular Dirigida/métodos , Nanoconjugados , Neoplasias/irrigación sanguínea , Neoplasias/patología , Oligonucleótidos/farmacología , Oligonucleótidos/uso terapéutico , Oligonucleótidos Antisentido/farmacología , Oligonucleótidos Antisentido/uso terapéutico , Transducción de Señal/efectos de los fármacos , Inhibidores de Topoisomerasa/farmacología , Resultado del Tratamiento
9.
Biochim Biophys Acta Rev Cancer ; 1868(1): 239-245, 2017 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-28526262

RESUMEN

Dissemination of breast cancer cells (BCCs) to distant sites (metastasis) is the ultimate cause of mortality in patients with breast cancer. Hypoxia (low O2) is a microenvironmental hallmark of most solid cancers arising as a mismatch between cellular O2 consumption and supply. Hypoxic selection of BCCs triggers molecular and cellular adaptations dependent upon hypoxia-inducible factors (HIFs), a family of evolutionarily conserved transcriptional activators that coordinate the expression of numerous genes controlling each step of the metastatic process. In this review, we summarize current advances in the understanding of HIF-driven molecular mechanisms that promote BCC metastatic dissemination and patient mortality. In addition, we discuss the clinical and therapeutic implications of HIF targeting in breast cancers.


Asunto(s)
Neoplasias de la Mama/patología , Hipoxia de la Célula/fisiología , Metástasis de la Neoplasia/patología , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Neoplasias de la Mama/metabolismo , Femenino , Humanos , Consumo de Oxígeno/fisiología , Activación Transcripcional/fisiología , Microambiente Tumoral/fisiología
10.
Adv Drug Deliv Rev ; 109: 45-62, 2017 01 15.
Artículo en Inglés | MEDLINE | ID: mdl-27771366

RESUMEN

Hypoxia (low O2) is an essential microenvironmental driver of phenotypic diversity in human solid cancers. Hypoxic cancer cells hijack evolutionarily conserved, O2- sensitive pathways eliciting molecular adaptations that impact responses to radiotherapy, tumor recurrence and patient survival. In this review, we summarize the radiobiological, genetic, epigenetic and metabolic mechanisms orchestrating oncogenic responses to hypoxia. In addition, we outline emerging hypoxia- targeting strategies that hold promise for individualized cancer therapy in the context of radiotherapy and drug delivery.


Asunto(s)
Hipoxia de la Célula/efectos de la radiación , Terapia Molecular Dirigida , Neoplasias/metabolismo , Neoplasias/radioterapia , Oxígeno/metabolismo , Hipoxia de la Célula/genética , Epigénesis Genética , Humanos , Neoplasias/genética , Neoplasias/patología
11.
Trends Cancer ; 2(12): 758-770, 2016 12.
Artículo en Inglés | MEDLINE | ID: mdl-28741521

RESUMEN

Intratumoral hypoxia (reduced O2 availability) is a common finding in human cancer and leads to increased activity of hypoxia-inducible factors (HIFs), which regulate the expression of genes that contribute to angiogenesis, metabolic reprogramming, extracellular matrix remodeling, epithelial-mesenchymal transition, motility, invasion, metastasis, cancer stem cell maintenance, immune evasion, and resistance to chemotherapy and radiation therapy. Conventional anticancer therapies target well-oxygenated and proliferating cancer cells, whereas there are no approved therapies that target hypoxic cancer cells, despite growing clinical and experimental evidence indicating that intratumoral hypoxia is a critical microenvironmental factor driving cancer progression. In this review, our current understanding of the consequences of HIF activity and the translational potential of targeting HIFs for cancer therapy are discussed.


Asunto(s)
Factor 1 Inducible por Hipoxia/metabolismo , Neoplasias/metabolismo , Neoplasias/patología , Animales , Transición Epitelial-Mesenquimal , Humanos , Células Madre Neoplásicas/metabolismo , Células Madre Neoplásicas/patología , Neovascularización Patológica
12.
Front Pharmacol ; 4: 13, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23408731

RESUMEN

HIF1α and NFkB are two transcription factors very frequently activated in tumors and involved in tumor growth, progression, and resistance to chemotherapy. In fact, HIF1α and NFkB together regulate transcription of over a thousand genes that, in turn, control vital cellular processes such as adaptation to the hypoxia, metabolic reprograming, inflammatory reparative response, extracellular matrix digestion, migration and invasion, adhesion, etc. Because of this wide involvement they could control in an integrated manner the origin of the malignant phenotype. Interestingly, hypoxia and inflammation have been sequentially bridged in tumors by the discovery that alarmin receptors genes such as RAGE, P2X7, and some TLRs, are activated by HIF1α; and that, in turn, alarmin receptors strongly activate NFkB and proinflammatory gene expression, evidencing all the hallmarks of the malignant phenotype. Recently, a large number of drugs have been identified that inhibit one or both transcription factors with promising results in terms of controlling tumor progression. In addition, many of these molecules are natural compounds or off-label drugs already used to cure other pathologies. Some of them are undergoing clinical trials and soon they will be used alone or in combination with standard anti-tumoral agents to achieve a better treatment of tumors with reduction of metastasis formation and, more importantly, with a net increase in survival. This review highlights the central role of HIF1α activated in hypoxic regions of the tumor, of NFkB activation and proinflammatory gene expression in transformed cells to understand their progression toward malignancy. Different molecules and strategies to inhibit these transcription factors will be reviewed. Finally, the central role of a new class of deacetylases called Sirtuins in regulating HIF1α and NFkB activity will be outlined.

13.
J Clin Invest ; 123(1): 189-205, 2013 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-23318994

RESUMEN

Metastasis involves critical interactions between cancer and stromal cells. Intratumoral hypoxia promotes metastasis through activation of hypoxia-inducible factors (HIFs). We demonstrate that HIFs mediate paracrine signaling between breast cancer cells (BCCs) and mesenchymal stem cells (MSCs) to promote metastasis. In a mouse orthotopic implantation model, MSCs were recruited to primary breast tumors and promoted BCC metastasis to LNs and lungs in a HIF-dependent manner. Coculture of MSCs with BCCs augmented HIF activity in BCCs. Additionally, coculture induced expression of the chemokine CXCL10 in MSCs and the cognate receptor CXCR3 in BCCs, which was augmented by hypoxia. CXCR3 expression was blocked in cocultures treated with neutralizing antibody against CXCL10. Conversely, CXCL10 expression was blocked in MSCs cocultured with BCCs that did not express CXCR3 or HIFs. MSC coculture did not enhance the metastasis of HIF-deficient BCCs. BCCs and MSCs expressed placental growth factor (PGF) and its cognate receptor VEGFR1, respectively, in a HIF-dependent manner, and CXCL10 expression by MSCs was dependent on PGF expression by BCCs. PGF promoted metastasis of BCCs and also facilitated homing of MSCs to tumors. Thus, HIFs mediate complex and bidirectional paracrine signaling between BCCs and MSCs that stimulates breast cancer metastasis.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Neoplasias de la Mama/metabolismo , Neoplasias Pulmonares/metabolismo , Células Madre Mesenquimatosas/metabolismo , Proteínas de Neoplasias/metabolismo , Transducción de Señal , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Neoplasias de la Mama/genética , Neoplasias de la Mama/patología , Línea Celular Tumoral , Quimiocina CXCL10/biosíntesis , Quimiocina CXCL10/genética , Femenino , Humanos , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/patología , Neoplasias Pulmonares/secundario , Células Madre Mesenquimatosas/patología , Ratones , Ratones SCID , Metástasis de la Neoplasia , Proteínas de Neoplasias/genética , Trasplante de Neoplasias , Receptores CXCR3/biosíntesis , Receptores CXCR3/genética , Trasplante Heterólogo
14.
Proc Natl Acad Sci U S A ; 109(40): E2707-16, 2012 Oct 02.
Artículo en Inglés | MEDLINE | ID: mdl-23012449

RESUMEN

Lymphatic dissemination from the primary tumor is a major mechanism by which breast cancer cells access the systemic circulation, resulting in distant metastasis and mortality. Numerous studies link activation of hypoxia-inducible factor 1 (HIF-1) with tumor angiogenesis, metastasis, and patient mortality. However, the role of HIF-1 in lymphatic dissemination is poorly understood. In this study, we show that HIF-1 promotes lymphatic metastasis of breast cancer by direct transactivation of the gene encoding platelet-derived growth factor B (PDGF-B), which has proliferative and chemotactic effects on lymphatic endothelial cells. Lymphangiogenesis and lymphatic metastasis in mice bearing human breast cancer orthografts were blocked by administration of the HIF-1 inhibitor digoxin or the tyrosine kinase inhibitor imatinib. Immunohistochemical analysis of human breast cancer biopsies demonstrated colocalization of HIF-1α and PDGF-B, which were correlated with lymphatic vessel area and histological grade. Taken together, these data provide experimental support for breast cancer clinical trials targeting HIF-1 and PDGF-B.


Asunto(s)
Neoplasias de la Mama/fisiopatología , Factor 1 Inducible por Hipoxia/fisiología , Metástasis Linfática/fisiopatología , Proteínas Proto-Oncogénicas c-sis/metabolismo , Activación Transcripcional/fisiología , Benzamidas , Neoplasias de la Mama/metabolismo , Hipoxia de la Célula/fisiología , Línea Celular Tumoral , Inmunoprecipitación de Cromatina , Digoxina/farmacología , Femenino , Humanos , Factor 1 Inducible por Hipoxia/antagonistas & inhibidores , Mesilato de Imatinib , Immunoblotting , Inmunohistoquímica , Luciferasas , Linfangiogénesis/efectos de los fármacos , Piperazinas/farmacología , Pirimidinas/farmacología , ARN Interferente Pequeño/metabolismo , Activación Transcripcional/efectos de los fármacos
15.
Carcinogenesis ; 32(8): 1167-75, 2011 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-21642357

RESUMEN

The role of hypoxia in regulating tumor progression is still controversial. Here, we demonstrate that, similarly to what previously observed by us in human prostate and breast tumor samples, hypoxia increases expression of the receptor for advanced glycation end products (RAGE) and the purinergic receptor P2X7 (P2X7R). The role of hypoxia was shown by the fact that hypoxia-inducible factor (HIF)-1α silencing downregulated RAGE and P2X7R protein levels as well as nuclear factor-kappaB (NF-κB) expression. In contrast, NF-κB silencing reduced P2X7R expression without affecting RAGE protein levels or nuclear accumulation of HIF-1α. Treatment of hypoxic tumor cells with HMGB1 and BzATP ligands, respectively, of RAGE and P2X7R, activated a signaling pathway that, through Akt and Erk phosphorylation, determines nuclear accumulation of NF-κB and increases cell invasion. Inhibition of Akt by SH5 and Erk by INH1 prevented both nuclear translocation of NF-κB and cell invasion. Moreover, silencing RAGE and P2X7R abolished nuclear accumulation of NF-κB as well as cell invasion without affecting HIF-1α stabilization. Once in the nucleus, NF-κB would contribute to cell survival and invasion under hypoxia, by maintaining RAGE and P2X7R expression levels and matrix metalloproteinases 2 and 9 synthesis. These results show that, hypoxia can upregulate expression levels of membrane receptors that, by binding extracellular molecules eventually released by necrotic cells, contribute to the increased invasiveness of transformed tumor cells. Moreover, these observations strengthen our working hypothesis that upregulation of damage-associated molecular patterns receptors by HIF-1α represents the crucial event bridging hypoxia and inflammation in obtaining the malignant phenotype.


Asunto(s)
Neoplasias de la Mama/patología , Movimiento Celular , Núcleo Celular/metabolismo , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , FN-kappa B/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Receptores Inmunológicos/metabolismo , Receptores Purinérgicos P2X7/metabolismo , Neoplasias de la Mama/metabolismo , Línea Celular Tumoral , Femenino , Humanos , Hipoxia , Subunidad alfa del Factor 1 Inducible por Hipoxia/antagonistas & inhibidores , Subunidad alfa del Factor 1 Inducible por Hipoxia/genética , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Quinasa I-kappa B/antagonistas & inhibidores , Quinasa I-kappa B/genética , Quinasa I-kappa B/metabolismo , Immunoblotting , Metaloproteinasa 2 de la Matriz/metabolismo , Metaloproteinasa 9 de la Matriz/metabolismo , FN-kappa B/genética , Fosforilación , Transporte de Proteínas , ARN Interferente Pequeño/genética , Receptor para Productos Finales de Glicación Avanzada , Receptores Inmunológicos/antagonistas & inhibidores , Receptores Inmunológicos/genética , Receptores Purinérgicos P2X7/química , Receptores Purinérgicos P2X7/genética , Transducción de Señal
16.
Cancer Sci ; 101(4): 1014-23, 2010 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-20151982

RESUMEN

The role of tumor cells in synthesizing pro-inflammatory molecules is still controversial. Here we report that hypoxic treatment of the MCF-7 human mammary adenocarcinoma cell line induced activation of hypoxia-inducible factor 1alpha (HIF-1alpha) and nuclear factor-kappa B (NF-kappaB). Importantly, hypoxia regulated expression of alarmin receptors such as the receptor for advanced glycation end products (RAGE) and the purinoreceptor (P2X7R), and up-regulated inflammatory response (IR) genes such as the inducible enzymes nitric oxide synthase (NOS2), cycloxygenase (COX2), and the acute-phase protein pentraxin-3 (PTX3). Hypoxia also stimulated chemokine (C-X-C motif) receptor 4 (CXCR4) mRNA synthesis. In fact, the CXCR4 ligand stromal-derived factor-1alpha (SDF-1alpha) increased invasion and migration of hypoxic MCF-7 cells. Inhibition of HIF-1alpha by chetomin and NF-kappaB by parthenolide reduced mRNA and protein expression of the studied molecules and prevented invasion of hypoxic MCF-7 cells. Moreover, solid invasive mammary tumor microenvironment was analyzed after laser-capture microdissection (LCMD) comparing tumor versus host normal tissue. Nuclear translocation of HIF-1alpha and NF-kappaB and up-regulation of IR, CXCR4, estrogen receptor alpha (ERalpha), and epithelial growth factor receptor (EGFR) was observed in tumor but not in host normal tissue in the absence of a local inflammatory leukocyte infiltrate. We conclude that under hypoxic conditions MCF-7 cells acquire a pro-inflammatory phenotype, and that solid human mammary carcinoma evidenced a similar activation of HIF-1alpha, NF-kappaB, and IR genes in malignant tumor cells as compared to the normal host tissues. We suggest a role for IR activation in the malignant progression of transformed cells.


Asunto(s)
Neoplasias de la Mama/genética , Neoplasias de la Mama/metabolismo , Hipoxia de la Célula/genética , Anciano , Neoplasias de la Mama/inmunología , Línea Celular Tumoral , Femenino , Humanos , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Inflamación/genética , Persona de Mediana Edad , FN-kappa B/antagonistas & inhibidores , FN-kappa B/genética , FN-kappa B/metabolismo , Invasividad Neoplásica/genética , Regulación hacia Arriba
17.
J Cell Physiol ; 223(2): 359-68, 2010 May.
Artículo en Inglés | MEDLINE | ID: mdl-20112292

RESUMEN

Survival strategies adopted by tumor cells in response to a hypoxic stress include activation of hypoxia-inducible factor 1 (HIF-1) and autophagy. However, the importance and the function of each molecular response is not well defined. In the present study, we investigated invasiveness, migration, matrix metalloproteinases (MMPs) activity, and cell survival of MDA-MB-231 cells under normoxia, hypoxia, and hypoxia/reoxygenation (H/R). Moreover, to assess the importance of hypoxia and autophagy on the parameters studied, cells were either left untreated or treated with Chetomin (a selective inhibitor of HIF-1alpha) or trifluoperazine (TFP, an activator of autophagy). We found that hypoxia and H/R stimulated invasiveness and migration of MDA-MB-231 cells with an increased MMP-2 activity. Chetomin and TFP differently regulated the cellular behavior under the oxygenation conditions studied. In fact, Chetomin was most effective in inhibiting cell invasion, MMPs activity, and cell survival under hypoxia but not normoxia or H/R. By contrast, TFP inhibition of cell invasion, migration, and cell survival was independent from oxygenation conditions. TFP-induced autophagy was inhibited by light chain protein 3 (LC3) silencing or 3-methyladenine (3MA) treatment. In fact, LC3-silenced cells were able to invade in the presence of TFP without any GATE16 processing and p62 degradation. Immunofluorescence assay showed that LC3 silencing inhibited TFP-induced autophagosome formation. However, we also showed that both TPF treatment and LC3 silencing caused cytoskeleton impairments suggesting a possible interaction between LC3 and cytoskeleton components. In conclusion, our study shows that hypoxia and autophagy by acting on common (HIF-1alpha) or separate (MMPs, cytoskeleton) targets differently regulate cell invasion, MMPs activity, and survival.


Asunto(s)
Autofagia/fisiología , Invasividad Neoplásica/fisiopatología , Neoplasias/metabolismo , Neoplasias/fisiopatología , Adenina/análogos & derivados , Adenina/farmacología , Autofagia/efectos de los fármacos , Hipoxia de la Célula/fisiología , Línea Celular Tumoral , Ensayos de Migración Celular , Movimiento Celular/fisiología , Supervivencia Celular/fisiología , Citoesqueleto/metabolismo , Disulfuros/farmacología , Antagonistas de Dopamina/farmacología , Matriz Extracelular/enzimología , Femenino , Humanos , Subunidad alfa del Factor 1 Inducible por Hipoxia/antagonistas & inhibidores , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Alcaloides Indólicos/farmacología , Metaloproteinasa 2 de la Matriz/metabolismo , Metaloproteinasas de la Matriz/metabolismo , Proteínas Asociadas a Microtúbulos/genética , Consumo de Oxígeno/fisiología , Fagosomas/efectos de los fármacos , Fagosomas/metabolismo , Interferencia de ARN , Trifluoperazina/farmacología
18.
Autophagy ; 4(8): 1042-53, 2008 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-18927491

RESUMEN

Adaptation to hypoxia through activation of the hypoxia inducible factor-1 (HIF-1) is crucial for tumor cells survival. Here we describe the antitumoral effects of the new molecule CR 3294 on tumor cells in the presence of hypoxia. Treatment of the breast carcinoma cell line MDA-MB-231 with CR 3294 in 1% O(2) resulted in an in vivo and in vitro inhibition of tumor growth. CR 3294 induced accumulation of autophagosomes in hypoxic MDA-MB-231 cells as assessed by both transmission electron microscopy (TEM) and the autophagic marker LC3-II. TEM analysis revealed the presence of invaginations of the cytoplasm into the nucleus. Autophagosomes were present in such invaginations. Moreover, CR 3294 inhibited both the DNA binding of HIF-1alpha and VEGF mRNA synthesis. Immunoprecipitation and immunofluorescence studies showed an interaction between LC3 and HIF-1alpha. We next detailed the effect of inhibitors and activators of autophagy on both HIF-1alpha and LC3. In particular, 3 methyladenine (3MA) and wortmannin, two macroautophagic inhibitors, prevented both the decrease of HIF-1alpha protein levels and LC3 processing in cells treated with CR 3294. Bafilomycin and leupeptin, inhibitors of lysosomes, prevented HIF-1alpha decrease without affecting LC3 processing. By contrast, treating hypoxic MDA-MB-231 cells with trifluoperazine (TFP) or serum withdrawal (SW), two activators of autophagy, diminished HIF-1alpha levels and stimulated LC3 processing. These results indicate that activation of the autophagic pathway in hypoxic cells by the new molecule CR 3294, as well as by TFP or SW, can have potentially important implications for cancer treatment.


Asunto(s)
Amidinas/farmacología , Antiinflamatorios no Esteroideos/farmacología , Antineoplásicos/farmacología , Autofagia/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Neoplasias/metabolismo , Tiourea/análogos & derivados , Adenina/análogos & derivados , Adenina/farmacología , Amidinas/química , Androstadienos/farmacología , Animales , Antiinflamatorios no Esteroideos/química , Antineoplásicos/química , Hipoxia de la Célula , Línea Celular Tumoral , Humanos , Ratones , Ratones Transgénicos , Proteínas Asociadas a Microtúbulos/metabolismo , Neoplasias/ultraestructura , Fagosomas/metabolismo , Fagosomas/ultraestructura , Tiourea/química , Tiourea/farmacología , Trifluoperazina/farmacología , Factor A de Crecimiento Endotelial Vascular/genética , Factor A de Crecimiento Endotelial Vascular/metabolismo , Wortmanina
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...