Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
1.
Development ; 144(3): 464-478, 2017 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-28049660

RESUMEN

Intestinal smooth muscle cells (iSMCs) are a crucial component of the adult gastrointestinal tract and support intestinal differentiation, peristalsis and epithelial homeostasis during development. Despite these crucial roles, the origin of iSMCs and the mechanisms responsible for their differentiation and function remain largely unknown in vertebrates. Here, we demonstrate that iSMCs arise from the lateral plate mesoderm (LPM) in a stepwise process. Combining pharmacological and genetic approaches, we show that TGFß/Alk5 signaling drives the LPM ventral migration and commitment to an iSMC fate. The Alk5-dependent induction of zeb1a and foxo1a is required for this morphogenetic process: zeb1a is responsible for driving LPM migration around the gut, whereas foxo1a regulates LPM predisposition to iSMC differentiation. We further show that TGFß, zeb1a and foxo1a are tightly linked together by miR-145 In iSMC-committed cells, TGFß induces the expression of miR-145, which in turn is able to downregulate zeb1a and foxo1a The absence of miR-145 results in only a slight reduction in the number of iSMCs, which still express mesenchymal genes but fail to contract. Together, our data uncover a cascade of molecular events that govern distinct morphogenetic steps during the emergence and differentiation of vertebrate iSMCs.


Asunto(s)
Intestinos/citología , Miocitos del Músculo Liso/citología , Animales , Animales Modificados Genéticamente , Diferenciación Celular/genética , Diferenciación Celular/fisiología , Proteína Forkhead Box O1/genética , Proteína Forkhead Box O1/metabolismo , Regulación del Desarrollo de la Expresión Génica , Técnicas de Silenciamiento del Gen , Mucosa Intestinal/metabolismo , Intestinos/embriología , Mesodermo/citología , Mesodermo/embriología , Mesodermo/metabolismo , Modelos Biológicos , Morfogénesis , Miocitos del Músculo Liso/metabolismo , Regiones Promotoras Genéticas , Transducción de Señal , Factor de Crecimiento Transformador beta/metabolismo , Pez Cebra/embriología , Pez Cebra/genética , Pez Cebra/metabolismo , Proteínas de Pez Cebra/genética , Proteínas de Pez Cebra/metabolismo , Homeobox 1 de Unión a la E-Box con Dedos de Zinc/genética , Homeobox 1 de Unión a la E-Box con Dedos de Zinc/metabolismo
2.
Blood ; 125(14): 2245-53, 2015 Apr 02.
Artículo en Inglés | MEDLINE | ID: mdl-25678499

RESUMEN

We recently described morgana as an essential protein able to regulate centrosome duplication and genomic stability, by inhibiting ROCK. Here we show that morgana (+/-) mice spontaneously develop a lethal myeloproliferative disease resembling human atypical chronic myeloid leukemia (aCML), preceded by ROCK hyperactivation, centrosome amplification, and cytogenetic abnormalities in the bone marrow (BM). Moreover, we found that morgana is underexpressed in the BM of patients affected by atypical CML, a disorder of poorly understood molecular basis, characterized by nonrecurrent cytogenetic abnormalities. Morgana is also underexpressed in the BM of a portion of patients affected by Philadelphia-positive CML (Ph(+) CML) caused by the BCR-ABL oncogene, and in this condition, morgana underexpression predicts a worse response to imatinib, the standard treatment for Ph(+) CML. Thus, morgana acts as an oncosuppressor with different modalities: (1) Morgana underexpression induces centrosome amplification and cytogenetic abnormalities, and (2) in Ph(+) CML, it synergizes with BCR-ABL signaling, reducing the efficacy of imatinib treatment. Importantly, ROCK inhibition in the BM of patients underexpressing morgana restored the efficacy of imatinib to induce apoptosis, suggesting that ROCK inhibitors, combined with imatinib treatment, can overcome suboptimal responses in patients in which morgana is underexpressed.


Asunto(s)
Benzamidas/farmacología , Proteínas Portadoras/fisiología , Resistencia a Antineoplásicos/genética , Proteínas de Fusión bcr-abl/metabolismo , Leucemia Mielógena Crónica BCR-ABL Positiva/patología , Cromosoma Filadelfia , Piperazinas/farmacología , Pirimidinas/farmacología , Quinasas Asociadas a rho/antagonistas & inhibidores , Animales , Apoptosis , Western Blotting , Médula Ósea/metabolismo , Médula Ósea/patología , Proliferación Celular , Citometría de Flujo , Proteínas de Fusión bcr-abl/genética , Humanos , Mesilato de Imatinib , Técnicas para Inmunoenzimas , Leucemia Mielógena Crónica BCR-ABL Positiva/genética , Leucemia Mielógena Crónica BCR-ABL Positiva/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Chaperonas Moleculares , Inhibidores de Proteínas Quinasas/farmacología , ARN Mensajero/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Células Tumorales Cultivadas , Quinasas Asociadas a rho/genética , Quinasas Asociadas a rho/metabolismo
3.
Cytokine ; 98: 42-50, 2017 10.
Artículo en Inglés | MEDLINE | ID: mdl-28579221

RESUMEN

The transcription factor signal transducer and activator of transcription (STAT) 3 is activated downstream of cytokines, growth factors and oncogenes to mediate their functions under both physiological and pathological conditions. In particular, aberrant/unrestrained STAT3 activity is detected in a wide variety of tumors, driving multiple pro-oncogenic functions. For that, STAT3 is widely considered as an oncogene and is the object of intense translational studies. One of the distinctive features of this factor is however, its ability to elicit different and sometimes contrasting effects under different conditions. In particular, STAT3 activities have been shown to be either pro-oncogenic or tumor-suppressive according to the tumor aetiology/mutational landscape, suggesting that the molecular bases underlining its functions are still incompletely understood. Here we discuss some of the properties that may provide the bases to explain STAT3 heterogeneous functions, and in particular how post-translational modifications contribute shaping its sub-cellular localization and activities, the cross talk between these activities and cell metabolic conditions, and finally how its functions can control the behaviour of both tumor and tumor microenvironment cell populations.


Asunto(s)
Neoplasias/fisiopatología , Factor de Transcripción STAT3/genética , Factor de Transcripción STAT3/metabolismo , Animales , Autofagia , Línea Celular Tumoral , Metabolismo Energético , Humanos , Ratones , Procesamiento Proteico-Postraduccional , Transducción de Señal , Microambiente Tumoral
4.
Haematologica ; 100(6): 720-9, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-25795718

RESUMEN

Feline leukemia virus subgroup C receptor 1 (Flvcr1) encodes two heme exporters: FLVCR1a, which localizes to the plasma membrane, and FLVCR1b, which localizes to mitochondria. Here, we investigated the role of the two Flvcr1 isoforms during erythropoiesis. We showed that, in mice and zebrafish, Flvcr1a is required for the expansion of committed erythroid progenitors but cannot drive their terminal differentiation, while Flvcr1b contributes to the expansion phase and is required for differentiation. FLVCR1a-down-regulated K562 cells have defective proliferation, enhanced differentiation, and heme loading in the cytosol, while FLVCR1a/1b-deficient K562 cells show impairment in both proliferation and differentiation, and accumulate heme in mitochondria. These data support a model in which the coordinated expression of Flvcr1a and Flvcr1b contributes to control the size of the cytosolic heme pool required to sustain metabolic activity during the expansion of erythroid progenitors and to allow hemoglobinization during their terminal maturation. Consistently, reduction or increase of the cytosolic heme rescued the erythroid defects in zebrafish deficient in Flvcr1a or Flvcr1b, respectively. Thus, heme export represents a tightly regulated process that controls erythropoiesis.


Asunto(s)
Diferenciación Celular/fisiología , Eritropoyesis/fisiología , Hemo/metabolismo , Líquido Intracelular/metabolismo , Proteínas de Transporte de Membrana/fisiología , Receptores Virales/fisiología , Secuencia de Aminoácidos , Animales , Humanos , Células K562 , Ratones , Ratones Noqueados , Ratones Transgénicos , Datos de Secuencia Molecular , Pez Cebra
5.
Int J Cancer ; 135(9): 1997-2003, 2014 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-24500994

RESUMEN

The transcription factor Signal Transducer and Activator of Transcription (STAT)3 has been considered as a potential anticancer target since its first description as an oncogene in 1999, recently leading to STAT3 inhibitors been brought to clinical trial for the treatment of solid tumors. However, the past 14 years of intense basic research have uncovered novel STAT3-mediated pathways that could affect the outcome of the designed therapies while at the same time help designing function-specific inhibitors. Particularly intriguing are the recent findings that suggest profound implications of STAT3 with the regulation of cellular metabolism in both canonical, that is transcriptional, and non-canonical ways. Here, after a short description of the main known features of STAT3 signaling and function, we review the recent literature on the role of STAT3 in regulating cellular metabolism and discuss the potential consequences on the therapeutic approaches currently under clinical experimentation.


Asunto(s)
Antineoplásicos/uso terapéutico , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Neoplasias/tratamiento farmacológico , Factor de Transcripción STAT3/antagonistas & inhibidores , Animales , Humanos , Neoplasias/metabolismo , Neoplasias/patología , Factor de Transcripción STAT3/metabolismo
6.
Breast Cancer Res ; 14(5): R137, 2012 Oct 26.
Artículo en Inglés | MEDLINE | ID: mdl-23098208

RESUMEN

INTRODUCTION: Intrinsic plasticity of breast carcinoma cells allows them to undergo a transient and reversible conversion into mesenchymal cells to disseminate into distant organs, where they can re-differentiate to an epithelial-like status to form a cohesive secondary mass. The p130Cas scaffold protein is overexpressed in human ER+ and HER2+ breast cancer where it contributes to cancer progression, invasion and resistance to therapy. However, its role in regulating mesenchymal aggressive breast cancer cells remains to be determined. The aim of this study was to investigate the molecular and functional involvement of this adaptor protein in breast cancer cell plasticity. METHODS: We used silencing strategies and rescue experiments to evaluate phenotypic and biochemical changes from mesenchymal to epithelial traits in breast tumor cell lines. In the mouse A17 cell model previously related to mesenchymal cancer stem cells and basal-like breast cancer, we biochemically dissected the signaling pathways involved and performed functional in vivo tumor growth ability assays. The significance of the signaling platform was assessed in a human setting through the use of specific inhibitors in aggressive MDA-MB-231 subpopulation LM2-4175 cells. To evaluate the clinical relevance of the results, we analyzed publicly available microarray data from the Netherlands Cancer Institute and from the Koo Foundation Sun Yat-Sen Cancer Center. RESULTS: We show that p130Cas silencing induces loss of mesenchymal features, by downregulating Vimentin, Snail, Slug and Twist transcriptional factors, resulting in the acquirement of epithelial-like traits. Mechanistically, p130Cas controls Cyclooxygenase-2 transcriptional expression, which in turn contributes to p130Cas-dependent maintenance of mesenchymal phenotype. This cascade of events also compromises in vivo tumor growth through inhibition of cell signaling controlling cell cycle progression. c-Src and JNK kinases are sequential players in p130Cas/ Cyclooxygenase-2 axis and their pharmacological inhibition is sufficient to downregulate Cyclooxygenase-2 leading to an epithelial phenotype. Finally, in silico microarray data analysis indicates that p130Cas and Cyclooxygenase-2 concomitant overexpression predicts poor survival and high probability of breast tumor recurrence. CONCLUSIONS: Overall, these data identify a new p130Cas/Cyclooxygenase-2 axis as a crucial element in the control of breast tumor plasticity, opening new therapeutic strategies leading to inhibition of these pathways in aggressive breast carcinoma.


Asunto(s)
Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Proteína Sustrato Asociada a CrK/metabolismo , Ciclooxigenasa 2/metabolismo , Animales , Neoplasias de la Mama/genética , Proteína Tirosina Quinasa CSK , Línea Celular Tumoral , Proteína Sustrato Asociada a CrK/genética , Ciclooxigenasa 2/genética , Femenino , Regulación Neoplásica de la Expresión Génica , Silenciador del Gen , Humanos , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Ratones , Modelos Biológicos , Fenotipo , Carácter Cuantitativo Heredable , Familia-src Quinasas/metabolismo
7.
Mol Ther Methods Clin Dev ; 18: 62-72, 2020 Sep 11.
Artículo en Inglés | MEDLINE | ID: mdl-32577433

RESUMEN

Myocarditis can lead to autoimmune disease, dilated cardiomyopathy, and heart failure, which is modeled in the mouse by cardiac myosin immunization (experimental autoimmune myocarditis [EAM]). Signal transducer and activator of transcription 3 (STAT3) systemic inhibition exerts both preventive and therapeutic effects in EAM, and STAT3 constitutive activation elicits immune-mediated myocarditis dependent on complement C3 and correlating with activation of the STAT3-interleukin 6 (IL-6) axis in the liver. Thus, liver-specific STAT3 inhibition may represent a therapeutic option, allowing to bypass the heart toxicity, predicted by systemic STAT3 inhibition. We therefore decided to explore the effectiveness of silencing liver Stat3 and C3 in preventing EAM onset and/or the recovery of cardiac functions. We first show that complement C3 and C5 genetic depletion significantly prevents the onset of spontaneous myocarditis, supporting the complement cascade as a viable target. In order to interfere with complement production and STAT3 activity specifically in the liver, we took advantage of liver-specific Stat3 or C3 small interfering (si)RNA nanoparticles, demonstrating that both siRNAs can significantly prevent myocarditis onset and improve the recovery of heart functions in EAM. Our data demonstrate that liver-specific Stat3/C3 siRNAs may represent a therapeutic option for autoimmune myocarditis and suggest that complement levels and activation might be predictive of progression to dilated cardiomyopathy.

8.
Cell Death Differ ; 26(5): 932-942, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-30042492

RESUMEN

STAT3 is an oncogenic transcription factor exerting its functions both as a canonical transcriptional activator and as a non-canonical regulator of energy metabolism and mitochondrial functions. While both activities are required for cell transformation downstream of different oncogenic stimuli, they rely on different post-translational activating events, namely phosphorylation on either Y705 (nuclear activities) or S727 (mitochondrial functions). Here, we report the discovery of the unexpected STAT3 localization to the endoplasmic reticulum (ER), from where it modulates ER-mitochondria Ca2+ release by interacting with the Ca2+ channel IP3R3 and facilitating its degradation. The release of Ca2+ is of paramount importance for life/death cell decisions, as excessive Ca2+ causes mitochondrial Ca2+ overload, the opening of the mitochondrial permeability transition pore, and the initiation of the intrinsic apoptotic program. Indeed, STAT3 silencing enhances ER Ca2+ release and sensitivity to apoptosis following oxidative stress in STAT3-dependent mammary tumor cells, correlating with increased IP3R3 levels. Accordingly, basal-like mammary tumors, which frequently display constitutively active STAT3, show an inverse correlation between IP3R3 and STAT3 protein levels. These results suggest that STAT3-mediated IP3R3 downregulation in the ER crucially contributes to its anti-apoptotic functions via modulation of Ca2+ fluxes.


Asunto(s)
Apoptosis/genética , Señalización del Calcio/genética , Receptores de Inositol 1,4,5-Trifosfato/genética , Factor de Transcripción STAT3/genética , Calcio/metabolismo , Muerte Celular/genética , Núcleo Celular/genética , Núcleo Celular/metabolismo , Retículo Endoplásmico/genética , Estrés del Retículo Endoplásmico/genética , Metabolismo Energético/genética , Expresión Génica/genética , Humanos , Mitocondrias/genética , Mitocondrias/metabolismo , Proteolisis
9.
Cancer Res ; 63(13): 3688-94, 2003 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-12839960

RESUMEN

Dendritic cells (DCs) need activation for the priming of antigen-specific immune responses. Recently activated DCs were described to prime in vitro strong T helper cell type 1 (Th(1)) responses, whereas at later time points, the same cells preferentially prime Th(2) cells [Langenkemp, A. et al., Nat. Immunol. 1: 311-316, 2000]. Because the immune response against cancer strongly depends on CTLs of Th(1)-like phenotype (Tc(1)), we verified here whether the kinetics of DCs activation also impacted on in vivo priming of tumor-specific CTLs. After pulsing with the CTL epitope TRP-2(180-188), bone-marrow-derived DCs, exposed to lipopolysaccharide (LPS) for 8 h (8hDC), elicited a more powerful Tc(1) response in C57BL/6 mice than did untreated DCs, or DCs exposed to LPS for 48 h (48hDC). Indeed, 8hDCs were the most potent protective and therapeutic vaccine against B16 melanoma. Despite a higher expression of MHC and costimulatory molecules by 48hDCs, 8hDCs and 48hDCs showed comparable allostimulatory and migration potential, and susceptibility to CTL-mediated apoptosis. However, 8hDCs exhibited a significantly higher interleukin (IL)-12 production potential. Release of IL-12 was necessary to induce potent Tc(1) cells, because DCs from IL-12p40(-/-) mice, irrespective of their maturation level, generated low CTL responses, comparable with 48hDCs and 0hDCs from wild-type animals. Our data are relevant for the design of DC-based vaccines.


Asunto(s)
Citotoxicidad Inmunológica , Células Dendríticas/inmunología , Activación de Linfocitos/inmunología , Melanoma Experimental/inmunología , Linfocitos T/inmunología , Animales , Apoptosis/inmunología , Células de la Médula Ósea/citología , Células de la Médula Ósea/inmunología , Células Dendríticas/patología , Femenino , Inmunización , Inmunofenotipificación , Interleucina-12/deficiencia , Interleucina-12/inmunología , Cinética , Melanoma Experimental/patología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Linfocitos T/patología , Células TH1/inmunología , Células Th2/inmunología
10.
Cancer Res ; 64(11): 3767-71, 2004 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-15172982

RESUMEN

Susceptibility of dendritic cells (DCs) to tumor-induced apoptosis reduces their efficacy in cancer therapy. Here we show that delivery within exponentially growing B16 melanomas of DCs treated ex vivo with nitric oxide (NO), released by the NO donor (z)-1-[2-(2-aminoethyl)-N-(2-ammonioethyl)amino]diazen-1-ium-1,2-diolate (DETA-NO), significantly reduced tumor growth, with cure of 37% of animals. DETA-NO-treated DCs became resistant to tumor-induced apoptosis because DETA-NO prevented tumor-induced changes in the expression of Bcl-2, Bax, and Bcl-xL; activation of caspase-9; and a reduction in the mitochondrial membrane potential. DETA-NO also increased DC cytotoxic activity against tumor cells and DC ability to trigger T-lymphocyte proliferation. All of the effects of DETA-NO were mediated through cGMP generation. NO and NO-generating drugs may therefore be used to increase the anticancer efficacy of DCs.


Asunto(s)
Células Dendríticas/efectos de los fármacos , Células Dendríticas/inmunología , Inmunoterapia Adoptiva/métodos , Melanoma Experimental/terapia , Óxido Nítrico/farmacología , Animales , Apoptosis/inmunología , Caspasa 9 , Caspasas/metabolismo , GMP Cíclico/metabolismo , Activación Enzimática , Femenino , Prueba de Cultivo Mixto de Linfocitos , Melanoma Experimental/inmunología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Donantes de Óxido Nítrico/farmacología , Triazenos/farmacología
11.
Sci Signal ; 9(459): ra124, 2016 12 20.
Artículo en Inglés | MEDLINE | ID: mdl-27999173

RESUMEN

Mobilization of neutrophils from the bone marrow determines neutrophil blood counts and thus is medically important. Balanced neutrophil mobilization from the bone marrow depends on the retention-promoting chemokine CXCL12 and its receptor CXCR4 and the egression-promoting chemokine CXCL2 and its receptor CXCR2. Both pathways activate the small guanosine triphosphatase Rac, leaving the role of this signaling event in neutrophil retention and egression ambiguous. On the assumption that active Rac determines persistent directional cell migration, we generated a mathematical model to link chemokine-mediated Rac modulation to neutrophil egression time. Our computer simulation indicated that, in the bone marrow, where the retention signal predominated, egression time strictly depended on the time it took Rac to return to its basal activity (namely, adaptation). This prediction was validated in mice lacking the Rac inhibitor ArhGAP15. Neutrophils in these mice showed prolonged Rac adaptation and cell-autonomous retention in the bone marrow. Our model thus demonstrates that mobilization in the presence of two spatially defined opposing chemotactic cues strictly depends on inhibitors shaping the time course of signal adaptation. Furthermore, our findings might help to find new modes of intervention to treat conditions characterized by excessively low or high circulating neutrophils.


Asunto(s)
Médula Ósea/enzimología , Neutrófilos/enzimología , Transducción de Señal/fisiología , Proteínas de Unión al GTP rac/metabolismo , Animales , Quimiocina CXCL12/genética , Quimiocina CXCL12/metabolismo , Proteínas Activadoras de GTPasa/genética , Proteínas Activadoras de GTPasa/metabolismo , Ratones , Ratones Noqueados , Receptores CXCR4/genética , Receptores CXCR4/metabolismo , Proteínas de Unión al GTP rac/genética
12.
Front Oncol ; 5: 121, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26106584

RESUMEN

Signal transducer and activator of transcription (STAT)3 mediates the signaling downstream of cytokine and growth factor receptors, regulating the expression of target genes. It is constitutively phosphorylated on tyrosine (Y-P) in many tumors, where its transcriptional activity can induce a metabolic switch toward aerobic glycolysis and down-regulate mitochondrial activity, a prominent metabolic feature of most cancer cells, correlating with reduced production of ROS, delayed senescence, and protection from apoptosis. STAT3 can, however, also localize to mitochondria, where its serine-phosphorylated (S-P) form preserves mitochondrial oxidative phosphorylation and controls the opening of the mitochondrial permeability transition pore, also promoting survival and resistance to apoptosis in response to specific signals/oncogenes such as RAS. Thus, downstream of different signals, both nuclear, Y-P STAT3, and mitochondrial, S-P STAT3, can act by promoting cell survival and reducing ROS production. Here, we discuss these properties in the light of potential connections between STAT3-driven alterations of mitochondrial metabolism and the development of drug resistance in cancer patients.

13.
Oncotarget ; 6(10): 7851-65, 2015 Apr 10.
Artículo en Inglés | MEDLINE | ID: mdl-25797262

RESUMEN

As widely recognized, tumor growth entails a close and complex cross-talk among cancer cells and the surrounding tumor microenvironment. We recently described the human RNASET2 gene as one key player of such microenvironmental cross-talk. Indeed, the protein encoded by this gene is an extracellular RNase which is able to control cancer growth in a non-cell autonomous mode by inducing a sustained recruitment of immune-competent cells belonging to the monocyte/macrophage lineage within a growing tumor mass. Here, we asked whether this oncosuppressor gene is sensitive to stress challenges and whether it can trigger cell-intrinsic processes as well. Indeed, RNASET2 expression levels were consistently found to increase following stress induction. Moreover, changes in RNASET2 expression levels turned out to affect several cancer-related parameters in vitro in an ovarian cancer cell line model. Of note, a remarkable rearrangement of the actin cytoskeleton organization, together with changes in cell adhesion and motility, emerged as putative mechanisms by which such cell-autonomous role could occur. Altogether, these biological features allow to put forward the hypothesis that the RNASET2 protein can act as a molecular barrier for limiting the damages and tissue remodeling events occurring during the earlier step of cell transformation.


Asunto(s)
Neoplasias Ováricas/genética , Ribonucleasas/genética , Proteínas Supresoras de Tumor/genética , Animales , Procesos de Crecimiento Celular/fisiología , Hipoxia de la Célula/fisiología , Línea Celular Tumoral , Movimiento Celular/fisiología , Femenino , Células HeLa , Xenoinjertos , Humanos , Masculino , Ratones , Ratones Desnudos , Neoplasias Ováricas/metabolismo , Neoplasias Ováricas/patología , Ribonucleasas/biosíntesis , Ribonucleasas/metabolismo , Transducción de Señal , Microambiente Tumoral , Proteínas Supresoras de Tumor/biosíntesis , Proteínas Supresoras de Tumor/metabolismo
14.
J Hematol Oncol ; 8: 64, 2015 Jun 09.
Artículo en Inglés | MEDLINE | ID: mdl-26054961

RESUMEN

BACKGROUND: Upon inflammation, myeloid cell generation in the bone marrow (BM) is broadly enhanced by the action of induced cytokines which are produced locally and at multiple sites throughout the body. METHODS: Using microarray studies, we found that Angptl4 is upregulated in the BM during systemic inflammation. RESULTS: Recombinant murine Angptl4 (rmAngptl4) stimulated the proliferation of myeloid colony-forming units (CFUs) in vitro. Upon repeated in vivo injections, rmAngptl4 increased BM progenitor cell frequency and this was paralleled by a relative increase in phenotypically defined granulocyte-macrophage progenitors (GMPs). Furthermore, in vivo treatment with rmAngptl4 resulted in elevated platelet counts in steady-state mice while allowing a significant acceleration of reconstitution of platelets after myelosuppressive therapy. The administration of rmAngptl4 increased the number of CD61(+)CD41(low)-expressing megakaryocytes (MK) in the BM of steady-state and in the spleen of transplanted mice. Furthermore, rmAngptl4 improved the in vitro differentiation of immature MKs from hematopoietic stem and progenitor cells. Mechanistically, using a signal transducer and activator of transcription 3 (STAT3) reporter knockin model, we show that rmAngptl4 induces de novo STAT3 expression in immature MK which could be important for the effective expansion of MKs after myelosuppressive therapy. CONCLUSION: Whereas the definitive role of Angptl4 in mediating the effects of lipopolysaccharide (LPS) on the BM has to be demonstrated by further studies involving multiple cytokine knockouts, our data suggest that Angptl4 plays a critical role during hematopoietic, especially megakaryopoietic, reconstitution following stem cell transplantation.


Asunto(s)
Angiopoyetinas/metabolismo , Plaquetas/metabolismo , Médula Ósea/metabolismo , Células Mieloides/metabolismo , Proteína 4 Similar a la Angiopoyetina , Animales , Femenino , Humanos , Ratones , Células Mieloides/citología , Factor de Transcripción STAT3/metabolismo , Regulación hacia Arriba
15.
J Vis Exp ; (89)2014 Jul 07.
Artículo en Inglés | MEDLINE | ID: mdl-25046434

RESUMEN

High levels of reactive oxygen species (ROS) may cause a change of cellular redox state towards oxidative stress condition. This situation causes oxidation of molecules (lipid, DNA, protein) and leads to cell death. Oxidative stress also impacts the progression of several pathological conditions such as diabetes, retinopathies, neurodegeneration, and cancer. Thus, it is important to define tools to investigate oxidative stress conditions not only at the level of single cells but also in the context of whole organisms. Here, we consider the zebrafish embryo as a useful in vivo system to perform such studies and present a protocol to measure in vivo oxidative stress. Taking advantage of fluorescent ROS probes and zebrafish transgenic fluorescent lines, we develop two different methods to measure oxidative stress in vivo: i) a "whole embryo ROS-detection method" for qualitative measurement of oxidative stress and ii) a "single-cell ROS detection method" for quantitative measurements of oxidative stress. Herein, we demonstrate the efficacy of these procedures by increasing oxidative stress in tissues by oxidant agents and physiological or genetic methods. This protocol is amenable for forward genetic screens and it will help address cause-effect relationships of ROS in animal models of oxidative stress-related pathologies such as neurological disorders and cancer.


Asunto(s)
Estrés Oxidativo/fisiología , Pez Cebra/embriología , Pez Cebra/metabolismo , Animales , Animales Modificados Genéticamente , Embrión no Mamífero/metabolismo , Femenino , Masculino , Especies Reactivas de Oxígeno/metabolismo
16.
Cancers (Basel) ; 6(3): 1579-96, 2014 Jul 31.
Artículo en Inglés | MEDLINE | ID: mdl-25089666

RESUMEN

STAT3 mediates cytokine and growth factor receptor signalling, becoming transcriptionally active upon tyrosine 705 phosphorylation (Y-P). Constitutively Y-P STAT3 is observed in many tumors that become addicted to its activity, and STAT3 transcriptional activation is required for tumor transformation downstream of several oncogenes. We have recently demonstrated that constitutively active STAT3 drives a metabolic switch towards aerobic glycolysis through the transcriptional induction of Hif-1α and the down-regulation of mitochondrial activity, in both MEF cells expressing constitutively active STAT3 (Stat3C/C) and STAT3-addicted tumor cells. This novel metabolic function is likely involved in mediating pre-oncogenic features in the primary Stat3C/C MEFs such as resistance to apoptosis and senescence and rapid proliferation. Moreover, it strongly contributes to the ability of primary Stat3C/C MEFs to undergo malignant transformation upon spontaneous immortalization, a feature that may explain the well known causative link between STAT3 constitutive activity and tumor transformation under chronic inflammatory conditions. Taken together with the recently uncovered role of STAT3 in regulating energy metabolism from within the mitochondrion when phosphorylated on Ser 727, these data place STAT3 at the center of a hub regulating energy metabolism under different conditions, in most cases promoting cell survival, proliferation and malignant transformation even though with distinct mechanisms.

17.
EMBO Mol Med ; 5(4): 572-90, 2013 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-23460527

RESUMEN

Myocarditis, often triggered by viral infection, may lead to heart auto-immunity and dilated cardiomyopathy. What determines the switch between disease resolution and progression is however incompletely understood. We show that pharmacological inhibition of STAT3, the main mediator of IL-6 signalling and of Th17-cell differentiation, protects mice from the development of Experimental Auto-immune Myocarditis reducing liver production of the complement component C3, and can act therapeutically when administered at disease peak. Further, we demonstrate that STAT3 is sufficient when constitutively active for triggering the onset of immune-mediated myocarditis, involving enhanced complement C3 production and IL-6 signalling amplification in the liver. Disease development can be prevented by C3 depletion and IL-6 receptor neutralization. This appears to be relevant to disease pathogenesis in humans, since acute myocarditis patients display significantly elevated circulating IL-6 and C3 levels and activated heart STAT3. Thus, aberrant IL-6/STAT3-mediated induction of liver acute phase response genes including C3, which occurs as a consequence of pre-existing inflammatory conditions, might represent an important factor determining the degree of myocarditis and its clinical outcome.


Asunto(s)
Cardiomiopatía Dilatada/inmunología , Miocarditis/inmunología , Factor de Transcripción STAT3/inmunología , Animales , Linfocitos T CD4-Positivos/microbiología , Cardiomiopatía Dilatada/genética , Complemento C3/inmunología , Progresión de la Enfermedad , Femenino , Humanos , Interleucina-6/genética , Interleucina-6/inmunología , Hígado/inmunología , Masculino , Ratones , Ratones Endogámicos BALB C , Miocarditis/genética , Factor de Transcripción STAT3/genética , Células Th17/inmunología
18.
Front Biosci (Landmark Ed) ; 17(6): 2306-26, 2012 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-22652781

RESUMEN

Interleukin-6 (IL-6) is a pleiotropic cytokine involved in the regulation of the cross talk between haematopoietic/immune cells and stromal cells, including the onset and resolution of inflammation, responses to infection, tissue remodelling and cancer. It is produced, among others, by fibroblasts, endothelial cells, macrophages and lymphocytes. IL-6 can interact with both membrane-bound and soluble forms of its ligand-binding receptor, the IL-6Ralpha, triggering signalling via dimerization of gp130, the signalling subunit of the IL-6 receptor complex. This leads to the activation of the JAK/STAT pathway and mainly culminates in the activation of the STAT3 transcription factor. Both IL-6 and STAT3 have recently emerged as main regulators of the differentiation and function of Th17 cells, via a positive feedback loop enhancing expression and/or activation of IL-6 itself, IL-17 and STAT3. Dysregulated IL-6 production and signalling are associated with chronic inflammatory diseases, auto-immunity and cancer, and are the object of intense translational research as promising therapeutic targets.


Asunto(s)
Autoinmunidad/inmunología , Interleucina-17/inmunología , Interleucina-6/inmunología , Factor de Transcripción STAT3/inmunología , Animales , Artritis Reumatoide/etiología , Artritis Reumatoide/inmunología , Enfermedades Autoinmunes/etiología , Enfermedades Autoinmunes/inmunología , Humanos , Ratones , Modelos Inmunológicos , Esclerosis Múltiple/etiología , Esclerosis Múltiple/inmunología , Miocarditis/etiología , Miocarditis/inmunología , Neoplasias/etiología , Neoplasias/inmunología , Transducción de Señal/inmunología , Células Th17/inmunología
19.
Horm Mol Biol Clin Investig ; 10(1): 217-25, 2012 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-25436678

RESUMEN

UNLABELLED: Abstract Background: The pro-oncogenic transcription factor STAT3 is constitutively active in tumours of many different origins, which often become addicted to its activity. STAT3 is believed to contribute to the initial survival of pre-cancerous cells as well as to hyper-proliferation and, later, metastasis. MATERIALS AND METHODS: To evaluate the contribution of enhanced STAT3 activation in a controlled model system, we generated knock-in mice in which a mutant constitutively active Stat3C allele replaces the endogenous wild-type allele and analysed its contribution to breast tumorigenesis. Moreover, we generated Stat3C/C MEF cells and analysed their gene expression and metabolic profiles. RESULTS: Constitutively active STAT3 could enhance the tumorigenic power of the rat Neu oncogene in MMTV-Neu transgenic mice and trigger the production of earlier onset and more invasive mammary tumours. Tumour-derived cell lines displayed higher migrating, invading and metastatic ability and showed disrupted distribution of cell-cell junction markers. These features were mediated by STAT3-dependent over-expression of the C-terminal tensin-like (Cten) focal adhesion protein. Moreover, STAT3C alone was able to induce aerobic glycolysis and down-regulate mitochondrial activity, both in primary fibroblasts and in STAT3-dependent tumour cell lines, acting via both HIF-1α-dependent and independent mechanisms. CONCLUSIONS: STAT3 can induce a metabolic switch that predisposes cells to aberrant survival, enhanced proliferation and, finally, tumour transformation. Later, enhanced Cten expression contributes to tissue infiltration and metastasis. While not excluding the contribution of many other tumour-specific STAT3 target genes, our data provide a unifying explanation of several pro-oncogenic STAT3 activities.

20.
J Clin Invest ; 122(12): 4569-79, 2012 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-23187127

RESUMEN

Feline leukemia virus subgroup C receptor 1 (FLVCR1) is a cell membrane heme exporter that maintains the balance between heme levels and globin synthesis in erythroid precursors. It was previously shown that Flvcr1-null mice died in utero due to a failure of erythropoiesis. Here, we identify Flvcr1b, a mitochondrial Flvcr1 isoform that promotes heme efflux into the cytoplasm. Flvcr1b overexpression promoted heme synthesis and in vitro erythroid differentiation, whereas silencing of Flvcr1b caused mitochondrial heme accumulation and termination of erythroid differentiation. Furthermore, mice lacking the plasma membrane isoform (Flvcr1a) but expressing Flvcr1b had normal erythropoiesis, but exhibited hemorrhages, edema, and skeletal abnormalities. Thus, FLVCR1b regulates erythropoiesis by controlling mitochondrial heme efflux, whereas FLVCR1a expression is required to prevent hemorrhages and edema. The aberrant expression of Flvcr1 isoforms may play a role in the pathogenesis of disorders characterized by an imbalance between heme and globin synthesis.


Asunto(s)
Diferenciación Celular , Eritrocitos/fisiología , Proteínas de Transporte de Membrana/fisiología , Proteínas Mitocondriales/fisiología , Receptores Virales/fisiología , Anomalías Múltiples/genética , Animales , Enfermedades del Desarrollo Óseo/genética , Encéfalo/metabolismo , Edema/genética , Eritrocitos/metabolismo , Eritropoyesis , Exones , Expresión Génica , Técnicas de Silenciamiento del Gen , Células HEK293 , Células HeLa , Hemo/metabolismo , Hemorragia/genética , Humanos , Proteínas de Transporte de Membrana/genética , Proteínas de Transporte de Membrana/metabolismo , Ratones , Ratones de la Cepa 129 , Ratones Endogámicos C57BL , Ratones Noqueados , Mitocondrias/metabolismo , Proteínas Mitocondriales/genética , Proteínas Mitocondriales/metabolismo , Especificidad de Órganos , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Isoformas de Proteínas/fisiología , Receptores Virales/genética , Receptores Virales/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA