Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
Más filtros

Bases de datos
Tipo del documento
Intervalo de año de publicación
1.
Int J Mol Sci ; 19(4)2018 Apr 13.
Artículo en Inglés | MEDLINE | ID: mdl-29652798

RESUMEN

In recent years many articles have underlined the key role of nanovesicles, i.e., exosomes, as information carriers among biological systems including cancer. Tumor-derived exosomes (TEXs) are key players in the dynamic crosstalk between cancer cells and the microenvironment while promote immune system control evasion. In fact, tumors are undoubtedly capable of silencing the immune response through multiple mechanisms, including the release of exosomes. TEXs have been shown to boost tumor growth and promote progression and metastatic spreading via suppression or stimulation of the immune response towards cancer cells. The advantage of immunotherapeutic treatment alone over combining immuno- and conventional therapy is currently debated. Understanding the role of tumor exosome-cargo is of crucial importance for our full comprehension of neoplastic immonosuppression and for the construction of novel therapies and vaccines based on (nano-) vesicles. Furthermore, to devise new anti-cancer approaches, diverse groups investigated the possibility of engineering TEXs by conditioning cancer cells’ own cargo. In this review, we summarize the state of art of TEX-based immunomodulation with a particular focus on the molecular function of non-coding family genes, microRNAs. Finally, we will report on recent efforts in the study of potential applications of engineered exosomes in cancer immunotherapy.


Asunto(s)
Exosomas/genética , Inmunoterapia/métodos , MicroARNs/genética , Neoplasias/terapia , Vacunas contra el Cáncer/uso terapéutico , Humanos , Nanopartículas/uso terapéutico , Neoplasias/genética , Microambiente Tumoral
2.
Nat Med ; 13(5): 613-8, 2007 May.
Artículo en Inglés | MEDLINE | ID: mdl-17468766

RESUMEN

Growing evidence indicates that microRNAs (miRNAs or miRs) are involved in basic cell functions and oncogenesis. Here we report that miR-133 has a critical role in determining cardiomyocyte hypertrophy. We observed decreased expression of both miR-133 and miR-1, which belong to the same transcriptional unit, in mouse and human models of cardiac hypertrophy. In vitro overexpression of miR-133 or miR-1 inhibited cardiac hypertrophy. In contrast, suppression of miR-133 by 'decoy' sequences induced hypertrophy, which was more pronounced than that after stimulation with conventional inducers of hypertrophy. In vivo inhibition of miR-133 by a single infusion of an antagomir caused marked and sustained cardiac hypertrophy. We identified specific targets of miR-133: RhoA, a GDP-GTP exchange protein regulating cardiac hypertrophy; Cdc42, a signal transduction kinase implicated in hypertrophy; and Nelf-A/WHSC2, a nuclear factor involved in cardiogenesis. Our data show that miR-133, and possibly miR-1, are key regulators of cardiac hypertrophy, suggesting their therapeutic application in heart disease.


Asunto(s)
Cardiomegalia/genética , MicroARNs/genética , Animales , Aorta Torácica/patología , Modelos Animales de Enfermedad , Humanos , Ratones , Ratones Transgénicos , Datos de Secuencia Molecular , Análisis de Secuencia por Matrices de Oligonucleótidos , Proteína Oncogénica v-akt/genética , Ratas
3.
Circulation ; 123(3): 282-91, 2011 Jan 25.
Artículo en Inglés | MEDLINE | ID: mdl-21220732

RESUMEN

BACKGROUND: Diabetes mellitus impairs endothelial cell (EC) function and postischemic reparative neovascularization by molecular mechanisms that are not fully understood. microRNAs negatively regulate the expression of target genes mainly by interaction in their 3' untranslated region. METHODS AND RESULTS: We found that microRNA-503 (miR-503) expression in ECs is upregulated in culture conditions mimicking diabetes mellitus (high D-glucose) and ischemia-associated starvation (low growth factors). Under normal culture conditions, lentivirus-mediated miR-503-forced expression inhibited EC proliferation, migration, and network formation on Matrigel (comparisons versus lentivirus.GFP control). Conversely, blocking miR-503 activity by either adenovirus-mediated transfer of a miR-503 decoy (Ad.decoymiR-503) or by antimiR-503 (antisense oligonucleotide) improved the functional capacities of ECs cultured under high D-glucose/low growth factors. We identified CCNE1 and cdc25A as direct miR-503 targets which are downregulated by high glucose/low growth factors in ECs. Next, we obtained evidence that miR-503 expression is increased in ischemic limb muscles of streptozotocin-diabetic mice and in ECs enriched from these muscles. Moreover, Ad.decoymiR-503 delivery to the ischemic adductor of diabetic mice corrected diabetes mellitus-induced impairment of postischemic angiogenesis and blood flow recovery. We finally investigated miR-503 and target gene expression in muscular specimens from the amputated ischemic legs of diabetic patients. As controls, calf biopsies of nondiabetic and nonischemic patients undergoing saphenous vein stripping were used. In diabetic muscles, miR-503 expression was remarkably higher, and it inversely correlated with cdc25 protein expression. Plasma miR-503 levels were also elevated in the diabetic individuals. CONCLUSIONS: Our data suggest miR-503 as a possible therapeutic target in diabetic patients with critical limb ischemia.


Asunto(s)
Diabetes Mellitus Experimental , Isquemia , MicroARNs/fisiología , Neovascularización Fisiológica/fisiología , Animales , Biopsia , Células Cultivadas , Diabetes Mellitus Experimental/genética , Diabetes Mellitus Experimental/patología , Diabetes Mellitus Experimental/fisiopatología , Células Endoteliales/citología , Células Endoteliales/fisiología , Extremidades/irrigación sanguínea , Expresión Génica/fisiología , Glucosa/farmacología , Humanos , Péptidos y Proteínas de Señalización Intercelular/farmacología , Isquemia/genética , Isquemia/patología , Isquemia/fisiopatología , Masculino , Ratones , Ratones Endogámicos , Músculo Esquelético/patología , Músculo Esquelético/fisiología , Venas Umbilicales/citología , Fosfatasas cdc25/genética
4.
Cell Death Dis ; 12(7): 636, 2021 06 21.
Artículo en Inglés | MEDLINE | ID: mdl-34155195

RESUMEN

Extracellular vesicles (EVs) and their cargo represent an intriguing source of cancer biomarkers for developing robust and sensitive molecular tests by liquid biopsy. Prostate cancer (PCa) is still one of the most frequent and deadly tumor in men and analysis of EVs from biological fluids of PCa patients has proven the feasibility and the unprecedented potential of such an approach. Here, we exploited an antibody-based proteomic technology, i.e. the Reverse-Phase Protein microArrays (RPPA), to measure key antigens and activated signaling in EVs isolated from sera of PCa patients. Notably, we found tumor-specific protein profiles associated with clinical settings as well as candidate markers for EV-based tumor diagnosis. Among others, PD-L1, ERG, Integrin-ß5, Survivin, TGF-ß, phosphorylated-TSC2 as well as partners of the MAP-kinase and mTOR pathways emerged as differentially expressed endpoints in tumor-derived EVs. In addition, the retrospective analysis of EVs from a 15-year follow-up cohort generated a protein signature with prognostic significance. Our results confirm that serum-derived EV cargo may be exploited to improve the current diagnostic procedures while providing potential prognostic and predictive information. The approach proposed here has been already applied to tumor entities other than PCa, thus proving its value in translational medicine and paving the way to innovative, clinically meaningful tools.


Asunto(s)
Biomarcadores de Tumor/sangre , Vesículas Extracelulares/metabolismo , Proteínas de Neoplasias/sangre , Neoplasias de la Próstata/sangre , Proteoma , Proteómica , Adulto , Anciano , Línea Celular Tumoral , Vesículas Extracelulares/ultraestructura , Humanos , Masculino , Persona de Mediana Edad , Valor Predictivo de las Pruebas , Neoplasias de la Próstata/ultraestructura , Análisis por Matrices de Proteínas , Reproducibilidad de los Resultados , Estudios Retrospectivos
5.
J Exp Med ; 200(10): 1257-66, 2004 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-15545353

RESUMEN

The mechanisms that control neural stem and progenitor cell survival are unknown. In several pathological conditions, death receptor (DR) ligands and inflammatory cytokines exert a deleterious effect on neurons, whereas primitive neural cells migrate and survive in the site of lesion. Here, we show that even in the presence of inflammatory cytokines, DRs are unable to generate death signals in primitive neural cells. Neural stem and progenitor cells did not express caspase 8, the presence of which is required for initiating the caspase cascade. However, exogenous or cytokine-mediated expression of caspase 8 was not sufficient to restore their DR sensitivity. Searching for molecules potentially able to block DR death-inducing signaling complex (DISC), we found that primitive neural cells expressed high levels of the death effector domain-containing protein PED (also known as PEA-15). PED localized in the DISC and prevented caspase 8 recruitment and activation. Moreover, lentiviral-mediated delivery of PED antisense DNA resulted in dramatic down-regulation of the endogenous gene expression and sensitization of primitive neural cells to apoptosis mediated by inflammatory cytokines and DRs. Thus, absence of caspase 8 and high expression of PED constitute two levels of protection from apoptosis induced by DRs and inflammatory cytokines in neural stem and progenitor cells.


Asunto(s)
Apoptosis/fisiología , Caspasas/metabolismo , Regulación de la Expresión Génica/fisiología , Células Madre Multipotentes/fisiología , Neuronas/fisiología , Fosfoproteínas/metabolismo , Receptores del Factor de Necrosis Tumoral/metabolismo , Proteínas Reguladoras de la Apoptosis , Caspasa 8 , Células Cultivadas , Cartilla de ADN , ADN sin Sentido/fisiología , Citometría de Flujo , Técnicas de Transferencia de Gen , Humanos , Immunoblotting , Inmunohistoquímica , Inmunoprecipitación , Péptidos y Proteínas de Señalización Intracelular , Microscopía Fluorescente , Células Madre Multipotentes/metabolismo , Fosfoproteínas/fisiología , Ribonucleasas/metabolismo
6.
Cell Death Dis ; 10(3): 201, 2019 02 27.
Artículo en Inglés | MEDLINE | ID: mdl-30814510

RESUMEN

The pressure towards innovation and creation of new model systems in regenerative medicine and cancer research has fostered the development of novel potential therapeutic applications. Kidney injuries provoke a high request of organ transplants making it the most demanding system in the field of regenerative medicine. Furthermore, renal cancer frequently threaten patients' life and aggressive forms still remain difficult to treat. Ethical issues related to the use of embryonic stem cells, has fueled research on adult, patient-specific pluripotent stem cells as a model for discovery and therapeutic development, but to date, normal and cancerous renal experimental models are lacking. Several research groups are focusing on the development of organoid cultures. Since organoids mimic the original tissue architecture in vitro, they represent an excellent model for tissue engineering studies and cancer therapy testing. We established normal and tumor renal cell carcinoma organoids previously maintained in a heterogeneous multi-clone stem cell-like enriching medium. Starting from adult normal kidney specimens, we were able to isolate and propagate organoid 3D-structures composed of both differentiated and undifferentiated cells while expressing nephron specific markers. Furthermore, we were capable to establish organoids derived from cancer tissues although with a success rate inferior to that of their normal counterpart. Cancer cultures displayed epithelial and mesenchymal phenotype while retaining tumor specific markers. Of note, tumor organoids recapitulated neoplastic masses when orthotopically injected into immunocompromised mice. Our data suggest an innovative approach of long-term establishment of normal- and cancer-derived renal organoids obtained from cultures of fleshly dissociated adult tissues. Our results pave the way to organ replacement pioneering strategies as well as to new models for studying drug-induced nephrotoxicity and renal diseases. Along similar lines, deriving organoids from renal cancer patients opens unprecedented opportunities for generation of preclinical models aimed at improving therapeutic treatments.


Asunto(s)
Riñón/patología , Organoides/metabolismo , Medicina de Precisión/métodos , Medicina Regenerativa/métodos , Insuficiencia Renal Crónica/terapia , Humanos , Insuficiencia Renal Crónica/patología
7.
Haematologica ; 93(12): 1899-902, 2008 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-18838478

RESUMEN

Resistance to chemotherapy-induced cell death represents a major obstacle in the treatment of acute myeloid leukemia. APRIL (A Proliferation Inducing Ligand) is a member of the tumor necrosis factor superfamily that plays a key role in normal B-cell development, while promoting survival and proliferation of malignant B cells. We investigated APRIL expression and activity in acute myeloid leukemia. We found that APRIL mRNA and protein, including the secreted form, are expressed in leukemic cells of patients with M0, M2 and M4 acute myeloid leukemia subtypes but not in normal hematopoietic progenitors. Retrovirus-mediated APRIL expression in normal hematopoietic progenitors confers resistance to chemotherapeutic drugs-induced apoptosis. Conversely, blocking APRIL function by recombinant soluble APRIL receptors increased chemotherapeutic drugs-induced cell adeath in acute myeloid leukemia cells. These results indicate that APRIL acts in an autocrine fashion to protect acute myeloid leukemia cells from drug-induced death and foresee a therapeutic potential of APRIL antagonists in the treatment of acute myeloid leukemia.


Asunto(s)
Antineoplásicos/farmacocinética , Resistencia a Antineoplásicos , Leucemia Mieloide Aguda/patología , Miembro 13 de la Superfamilia de Ligandos de Factores de Necrosis Tumoral/antagonistas & inhibidores , Comunicación Autocrina , Humanos , Leucemia Mieloide Aguda/tratamiento farmacológico , Células Tumorales Cultivadas
8.
J Exp Clin Cancer Res ; 37(1): 217, 2018 Sep 05.
Artículo en Inglés | MEDLINE | ID: mdl-30185225

RESUMEN

BACKGROUND: Clear cell RCC (ccRCC) accounts for approximately 75% of the renal cancer cases. Surgery treatment seems to be the best efficacious approach for the majority of patients. However, a consistent fraction (30%) of cases progress after surgery with curative intent. It is currently largely debated the use of adjuvant therapy for high-risk patients and the clinical and molecular parameters for stratifying beneficiary categories. In addition, the treatment of advanced forms lacks reliable driver biomarkers for the appropriated therapeutic choice. Thus, renal cancer patient management urges predictive molecular indicators and models for therapy-decision making. METHODS: Here, we developed and optimized new models and tools for ameliorating renal cancer patient management. We isolated from fresh tumor specimens heterogeneous multi-clonal populations showing epithelial and mesenchymal characteristics coupled to stem cell phenotype. These cells retained long lasting-tumor-propagating capacity provided a therapy monitoring approach in vitro and in vivo while being able to form parental tumors when orthotopically injected and serially transplanted in immunocompromised murine hosts. RESULTS: In line with recent evidence of multiclonal cancer composition, we optimized in vitro cultures enriched of multiple tumor-propagating populations. Orthotopic xenograft masses recapitulated morphology, grading and malignancy of parental cancers. High-grade but not the low-grade neoplasias, resulted in efficient serial transplantation in mice. Engraftment capacity paralleled grading and recurrence frequency advocating for a prognostic value of our developed model system. Therefore, in search of novel molecular indicators for therapy decision-making, we used Reverse-Phase Protein Arrays (RPPA) to analyze a panel of total and phosphorylated proteins in the isolated populations. Tumor-propagating cells showed several deregulated kinase cascades associated with grading, including angiogenesis and m-TOR pathways. CONCLUSIONS: In the era of personalized therapy, the analysis of tumor propagating cells may help improve prediction of disease progression and therapy assignment. The possibility to test pharmacological response of ccRCC stem-like cells in vitro and in orthotopic models may help define a pharmacological profiling for future development of more effective therapies. Likewise, RPPA screening on patient-derived populations offers innovative approach for possible prediction of therapy response.


Asunto(s)
Biomarcadores de Tumor/genética , Neoplasias Renales/genética , Recurrencia Local de Neoplasia/genética , Medicina de Precisión , Animales , Linaje de la Célula/genética , Modelos Animales de Enfermedad , Humanos , Neoplasias Renales/patología , Ratones , Recurrencia Local de Neoplasia/patología , Pronóstico , Ensayos Antitumor por Modelo de Xenoinjerto
9.
Mol Cell Oncol ; 3(4): e1109744, 2016 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-27652312

RESUMEN

It is clear that several prostate cancers remain indolent whereas others develop into advanced forms. There is a need to improve patient management by identifying biomarkers for personalized treatment. We demonstrated that miR-15/miR-16 loss, miR-21 upregulation, and deregulation of their target genes represent a promising predictive signature of poor patient prognosis.

10.
FASEB J ; 18(12): 1392-4, 2004 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-15231728

RESUMEN

Hematopoietic (Hem) and endothelial (End) lineages derive from a common progenitor cell, the hemangioblast: specifically, the human cord blood (CB) CD34+KDR+ cell fraction comprises primitive Hem and End cells, as well as hemangioblasts. In humans, the potential therapeutic role of Hem and End progenitors in ischemic heart disease is subject to intense investigation. Particularly, the contribution of these cells to angiogenesis and cardiomyogenesis in myocardial ischemia is not well established. In our studies, we induced myocardial infarct (MI) in the immunocompromised NOD-SCID mouse model, and monitored the effects of myocardial transplantation of human CB CD34+ cells on cardiac function. Specifically, we compared the therapeutic effect of unseparated CD34+ cells vs. PBS and mononuclear cells (MNCs); moreover, we compared the action of the CD34+KDR+ cell subfraction vs. the CD34+KDR- subset. CD34+ cells significantly improve cardiac function after MI, as compared with PBS/MNCs. Similar beneficial actions were obtained using a 2-log lower number of CD34+KDR+ cells, while the same number of CD34+KDR- cells did not have any effects. The beneficial effect of CD34+KDR+ cells may mostly be ascribed to their notable resistance to apoptosis and to their angiogenic action, since cardiomyogenesis was limited. Altogether, our results indicate that, within the CD34+ cell population, the CD34+KDR+ fraction is responsible for the improvement in cardiac hemodynamics and hence represents the candidate active CD34+ cell subset.


Asunto(s)
Antígenos CD34/metabolismo , Trasplante de Células , Infarto del Miocardio/fisiopatología , Infarto del Miocardio/terapia , Receptor 2 de Factores de Crecimiento Endotelial Vascular/metabolismo , Animales , Apoptosis , Comunicación Autocrina/efectos de los fármacos , Fusión Celular , Linaje de la Célula , Técnicas de Cocultivo , Medio de Cultivo Libre de Suero/farmacología , Sangre Fetal/citología , Sangre Fetal/metabolismo , Ventrículos Cardíacos/patología , Ventrículos Cardíacos/fisiopatología , Hemodinámica/fisiología , Humanos , Ratones , Ratones Endogámicos NOD , Ratones SCID , Infarto del Miocardio/patología , Factores de Tiempo , Factor A de Crecimiento Endotelial Vascular/metabolismo , Función Ventricular
11.
Oncoscience ; 2(11): 920-3, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26697526

RESUMEN

Prostate cancer remains the second leading cause of death in men. It is imperative to improve patient management in identifying bio-markers for personalized treatment. We demonstrated miR-15/miR-16 loss and miR-21 up-regulation and deregulation of their target genes, which represent a promising signature for ameliorating therapy assignment and risk assessment in prostate cancer.

12.
Biomed Res Int ; 2014: 146170, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25309903

RESUMEN

Prostate cancer (PCa) is one of the leading causes of cancer-related death in men. Despite considerable advances in prostate cancer early detection and clinical management, validation of new biomarkers able to predict the natural history of tumor progression is still necessary in order to reduce overtreatment and to guide therapeutic decisions. MicroRNAs are endogenous noncoding RNAs which offer a fast fine-tuning and energy-saving mechanism for posttranscriptional control of protein expression. Growing evidence indicate that these RNAs are able to regulate basic cell functions and their aberrant expression has been significantly correlated with cancer development. Therefore, detection of microRNAs in tumor tissues and body fluids represents a new tool for early diagnosis and patient prognosis prediction. In this review, we summarize current knowledge about microRNA deregulation in prostate cancer mainly focusing on the different clinical aspects of the disease. We also highlight the potential roles of microRNAs in PCa management, while also discussing several current challenges and needed future research.


Asunto(s)
Bases de Datos Genéticas , MicroARNs/uso terapéutico , Neoplasias de la Próstata/diagnóstico , Neoplasias de la Próstata/terapia , Medición de Riesgo , Progresión de la Enfermedad , Regulación Neoplásica de la Expresión Génica , Humanos , Masculino , Pronóstico , Neoplasias de la Próstata/genética , Neoplasias de la Próstata/patología
13.
Crit Rev Oncog ; 18(4): 303-15, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23614617

RESUMEN

Prostate cancer is one of the most common causes of cancer-related death. The management of prostate cancer patients has become increasingly complex, consequently calling on the need for identifying and validating prognostic and predictive biomarkers. Growing evidence indicates that microRNAs play a crucial role in the pathobiology of neoplastic diseases. The deregulation of the cellular "miRNome" in prostate cancer has been connected with multiple tumor-promoting activities such as aberrant activation of growth signals, anti-apoptotic effects, prometastatic mechanisms, alteration of the androgen receptor pathway, and regulation of the cancer stem cell phenotype. With the elucidation of molecular mechanisms controlled by microRNAs, investigations have been conducted in an attempt to exploit these molecules in the clinical setting. Moreover, the multifaceted biological activity of microRNAs makes them an attractive candidate as anticancer agents. This review summarizes the current knowledge on microRNA deregulation in prostate cancer, and the rationale underlying their exploitation as cancer biomarkers and therapeutics.


Asunto(s)
Biomarcadores de Tumor/genética , MicroARNs/fisiología , Neoplasias de la Próstata/diagnóstico , Neoplasias de la Próstata/terapia , Animales , Regulación Neoplásica de la Expresión Génica , Humanos , Masculino , Neoplasias de la Próstata/genética
14.
Cancer J ; 18(3): 253-61, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22647362

RESUMEN

The management of prostate cancer patients is rapidly changing. The extended survival seen in randomized phase III trials with new molecules has significantly enriched the therapeutic armamentarium, and ongoing clinical trials are assessing whether the integration of these active drugs within established therapeutic regimens results in a further benefit for patients. This complex scenario is raising the need for the identification and validation of biomarkers able to drive the decision-making process during the course of the disease. Compelling evidence has documented the role of microRNAs in cancer biology, and their multifaceted biological activity makes them an attractive candidate as diagnostic, prognostic, and predictive biomarkers. This review summarizes the current knowledge about microRNA deregulation in prostate cancer, how these molecules have been investigated in the clinical setting, and strategies investigators should consider for sharpening their potential.


Asunto(s)
Biomarcadores de Tumor , MicroARNs/genética , Metástasis de la Neoplasia/genética , Neoplasias de la Próstata/genética , Apoptosis/genética , Puntos de Control del Ciclo Celular/genética , Proliferación Celular , Perfilación de la Expresión Génica , Humanos , Masculino , MicroARNs/metabolismo , Invasividad Neoplásica/genética , Pronóstico , Neoplasias de la Próstata/diagnóstico , Neoplasias de la Próstata/terapia
15.
Recent Pat Cardiovasc Drug Discov ; 5(3): 156-61, 2010 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-20649511

RESUMEN

MicroRNAs (miRNAs) are a broad class of small non-coding RNAs that control expression of complementary target messenger RNAs. Dysregulation of miRNAs has been described in various disease states including cancer and cardiac disease. A particular miRNA that was consistently reported to be upregulated in both cancer and various forms of cardiovascular diseases is miR-21. MiR-21 exerts oncogenic activity and therefore is considered as an oncomir. In the cardiovascular system miR-21 is enriched in fibroblasts and contributes to the development of fibrosis and heart failure. MiR-21 therefore emerges as an interesting candidate for the development of therapeutic strategies against many forms of cancer as well as heart diseases. Indeed, treatment with anti-miR-21 oligonucleotides reduced breast cancer growth. Inhibition of miR-21 by synthetic miRNA antagonists (antagomirs) improved heart function in a cardiac disease model. The same beneficial effects were observed in miR-21 knockout mice subjected to pressure-overload of the left ventricle underlining the key role of miR-21 as a therapeutic target. We here overview the current patent situation about the therapeutic use of miR-21 modulation in cancer and cardiovascular disease.


Asunto(s)
Enfermedades Cardiovasculares/metabolismo , MicroARNs/metabolismo , Neoplasias/metabolismo , Animales , Enfermedades Cardiovasculares/tratamiento farmacológico , Humanos , Ratones , MicroARNs/antagonistas & inhibidores , Terapia Molecular Dirigida , Neoplasias/tratamiento farmacológico , Oligonucleótidos/farmacología , Regulación hacia Arriba
16.
Methods Mol Biol ; 614: 149-60, 2010.
Artículo en Inglés | MEDLINE | ID: mdl-20225042

RESUMEN

The manipulation of cell differentiation is important to create new sources for the treatment of degenerative diseases or solve cell depletion after aggressive therapy against cancer. In this chapter, the use of a tissue-specific promoter lentiviral vector to obtain a myocardial pure lineage from murine embryonic stem cells (mES) is described in detail. Since the cardiac isoform of troponin I gene product is not expressed in skeletal or other muscle types, short mouse cardiac troponin proximal promoter is used to drive reporter genes. Cells are infected simultaneously with two lentiviral vectors, the first expressing EGFP to monitor the transduction efficiency, and the other expressing a puromycin resistance gene to select the specific cells of interest. This technical approach describes a method to obtain a pure cardiomyocyte population and can be applied to other lineages of interest.


Asunto(s)
Diferenciación Celular , Vectores Genéticos , Lentivirus/genética , Animales , Células Madre Embrionarias/citología , Ratones , Regiones Promotoras Genéticas
17.
Endocr Relat Cancer ; 17(1): F1-17, 2010 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-19779034

RESUMEN

Despite much progress in prostate cancer management, new diagnostic, prognostic and therapeutic tools are needed to predict disease severity, choose among the available treatments and establish more effective therapies for advanced prostate cancer. In the last few years, compelling evidence has documented the role of microRNAs as new broad-spectrum oncogenes or tumour suppressor genes, thus their use as diagnostic, prognostic and therapeutic biomolecules is envisaged. This review extensively and critically summarizes the current knowledge about microRNA deregulation in prostate cancer disease, underlining present limits and future perspectives.


Asunto(s)
Adenocarcinoma/genética , Regulación Neoplásica de la Expresión Génica , MicroARNs/genética , Neoplasias de la Próstata/genética , ARN Neoplásico/genética , Adenocarcinoma/diagnóstico , Adenocarcinoma/terapia , Andrógenos , Transformación Celular Neoplásica/genética , Progresión de la Enfermedad , Perfilación de la Expresión Génica , Genes Supresores de Tumor , Humanos , Masculino , MicroARNs/análisis , Invasividad Neoplásica/genética , Proteínas de Neoplasias/biosíntesis , Proteínas de Neoplasias/genética , Neoplasias Hormono-Dependientes/diagnóstico , Neoplasias Hormono-Dependientes/genética , Neoplasias Hormono-Dependientes/terapia , Pronóstico , Neoplasias de la Próstata/diagnóstico , Neoplasias de la Próstata/terapia , Proteínas Proto-Oncogénicas c-myc/fisiología
20.
PLoS One ; 3(12): e4029, 2008.
Artículo en Inglés | MEDLINE | ID: mdl-19107213

RESUMEN

BACKGROUND: MiR-221 and miR-222 are two highly homologous microRNAs whose upregulation has been recently described in several types of human tumors, for some of which their oncogenic role was explained by the discovery of their target p27, a key cell cycle regulator. We previously showed this regulatory relationship in prostate carcinoma cell lines in vitro, underlying the role of miR-221/222 as inducers of proliferation and tumorigenicity. METHODOLOGY/PRINCIPAL FINDINGS: Here we describe a number of in vivo approaches confirming our previous data. The ectopic overexpression of miR-221 is able, per se, to confer a high growth advantage to LNCaP-derived tumors in SCID mice. Consistently, the anti-miR-221/222 antagomir treatment of established subcutaneous tumors derived from the highly aggressive PC3 cell line, naturally expressing high levels of miR-221/222, reduces tumor growth by increasing intratumoral p27 amount; this effect is long lasting, as it is detectable as long as 25 days after the treatment. Furthermore, we provide evidence in favour of a clinical relevance of the role of miR-221/222 in prostate carcinoma, by showing their general upregulation in patient-derived primary cell lines, where we find a significant inverse correlation with p27 expression. CONCLUSIONS/SIGNIFICANCE: These findings suggest that modulating miR-221/222 levels may have a therapeutic potential in prostate carcinoma.


Asunto(s)
Carcinoma/patología , Proliferación Celular/efectos de los fármacos , MicroARNs/antagonistas & inhibidores , Oligonucleótidos/farmacología , Neoplasias de la Próstata/patología , Anciano , Animales , Secuencia de Bases , Carcinoma/genética , Progresión de la Enfermedad , Regulación hacia Abajo/efectos de los fármacos , Regulación hacia Abajo/genética , Regulación hacia Abajo/fisiología , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Terapia Genética , Humanos , Masculino , Ratones , Ratones SCID , MicroARNs/genética , Persona de Mediana Edad , Antígeno Nuclear de Célula en Proliferación/genética , Neoplasias de la Próstata/genética , Células Tumorales Cultivadas , Ensayos Antitumor por Modelo de Xenoinjerto
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA