Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
Más filtros

Bases de datos
Tipo del documento
Intervalo de año de publicación
1.
Nucleic Acids Res ; 52(10): 5610-5623, 2024 Jun 10.
Artículo en Inglés | MEDLINE | ID: mdl-38554106

RESUMEN

The use of androgen receptor (AR) inhibitors in prostate cancer gives rise to increased cellular lineage plasticity resulting in resistance to AR-targeted therapies. In this study, we examined the chromatin landscape of AR-positive prostate cancer cells post-exposure to the AR inhibitor enzalutamide. We identified a novel regulator of cell plasticity, the homeobox transcription factor SIX2, whose motif is enriched in accessible chromatin regions after treatment. Depletion of SIX2 in androgen-independent PC-3 prostate cancer cells induced a switch from a stem-like to an epithelial state, resulting in reduced cancer-related properties such as proliferation, colony formation, and metastasis both in vitro and in vivo. These effects were mediated through the downregulation of the Wnt/ß-catenin signalling pathway and subsequent reduction of nuclear ß-catenin. Collectively, our findings provide compelling evidence that the depletion of SIX2 may represent a promising strategy for overcoming the cell plasticity mechanisms driving antiandrogen resistance in prostate cancer.


Asunto(s)
Benzamidas , Plasticidad de la Célula , Proteínas de Homeodominio , Nitrilos , Feniltiohidantoína , Neoplasias de la Próstata , Receptores Androgénicos , Vía de Señalización Wnt , beta Catenina , Animales , Humanos , Masculino , Ratones , Benzamidas/farmacología , beta Catenina/metabolismo , beta Catenina/genética , Línea Celular Tumoral , Plasticidad de la Célula/genética , Proliferación Celular/efectos de los fármacos , Resistencia a Antineoplásicos/genética , Regulación Neoplásica de la Expresión Génica , Proteínas de Homeodominio/metabolismo , Proteínas de Homeodominio/genética , Proteínas del Tejido Nervioso/metabolismo , Proteínas del Tejido Nervioso/genética , Nitrilos/farmacología , Células PC-3 , Feniltiohidantoína/farmacología , Feniltiohidantoína/análogos & derivados , Neoplasias de la Próstata/genética , Neoplasias de la Próstata/metabolismo , Neoplasias de la Próstata/patología , Neoplasias de la Próstata/tratamiento farmacológico , Receptores Androgénicos/metabolismo , Receptores Androgénicos/genética , Vía de Señalización Wnt/efectos de los fármacos
2.
Cytotherapy ; 19(6): 689-702, 2017 06.
Artículo en Inglés | MEDLINE | ID: mdl-28411126

RESUMEN

BACKGROUND: Adoptive T-cell therapy offers new options for cancer treatment. Clinical results suggest that T-cell persistence, depending on T-cell memory, improves efficacy. The use of interleukin (IL)-2 for in vitro T-cell expansion is not straightforward because it drives effector T-cell differentiation but does not promote the formation of T-cell memory. We have developed a cost-effective expansion protocol for chimeric antigen receptor (CAR) T cells with an early memory phenotype. METHODS: Lymphocytes were transduced with third-generation lentiviral vectors and expanded using CD3/CD28 microbeads. The effects of altering the IL-2 supplementation (0-300 IU/mL) and length of expansion (10-20 days) on the phenotype of the T-cell products were analyzed. RESULTS: High IL-2 levels led to a decrease in overall generation of early memory T cells by both decreasing central memory T cells and augmenting effectors. T memory stem cells (TSCM, CD95+CD45RO-CD45RA+CD27+) were present variably during T-cell expansion. However, their presence was not IL-2 dependent but was linked to expansion kinetics. CD19-CAR T cells generated in these conditions displayed in vitro antileukemic activity. In summary, production of CAR T cells without any cytokine supplementation yielded the highest proportion of early memory T cells, provided a 10-fold cell expansion and the cells were functionally potent. DISCUSSION: The number of early memory T cells in a T-cell preparation can be increased by simply reducing the amount of IL-2 and limiting the length of T-cell expansion, providing cells with potentially higher in vivo performance. These findings are significant for robust and cost-effective T-cell manufacturing.


Asunto(s)
Interleucina-2/farmacología , Receptores de Antígenos de Linfocitos T/metabolismo , Linfocitos T/fisiología , Técnicas de Cultivo de Célula , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Citocinas/genética , Humanos , Memoria Inmunológica , Interleucina-15/farmacología , Interleucina-2/metabolismo , Fenotipo , Receptores de Antígenos de Linfocitos T/genética , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Células Madre/citología , Células Madre/fisiología , Linfocitos T/efectos de los fármacos , Linfocitos T/inmunología
3.
Amino Acids ; 46(3): 689-700, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-23836421

RESUMEN

Spermidine/spermine N(1)-acetyltransferase (SSAT) regulates intracellular polyamine levels by catabolizing spermidine and spermine which are essential for cell proliferation and differentiation. Hematological characterization of SSAT overexpressing mice (SSAT mice) revealed enhanced myelopoiesis and thrombocytopoiesis leading to increased amounts of myeloid cells in bone marrow, peripheral blood, and spleen compared to wild-type animals. The level of SSAT activity in the bone marrow cells was associated with the bone marrow cellularity and spleen weight which both were significantly increased in SSAT mice. The result of bone marrow transplantations indicated that both the intrinsic SSAT overexpression of bone marrow cells and bone marrow microenvironment had an impact on the observed hematopoietic phenotype. The Lineage-negative Sca-1(+) c-Kit(+) hematopoietic stem cell (HSC) compartment in SSAT mice, showed enhanced proliferation, increased proportion of long-term HSCs and affected expression of transcription factors associated with lineage priming and myeloid differentiation. The proportions of common myeloid and megakaryocytic/erythroid progenitors were decreased and the proportion of granulocyte-macrophage progenitors was increased in SSAT bone marrow. The data suggest that SSAT overexpression and the concomitantly accelerated polyamine metabolism in hematopoietic cells and bone marrow microenvironment affect lineage commitment and lead to the development of a mouse myeloproliferative disease in SSAT mice.


Asunto(s)
Acetiltransferasas/genética , Hematopoyesis , Trastornos Mieloproliferativos/metabolismo , Poliaminas/metabolismo , Acetiltransferasas/metabolismo , Animales , Femenino , Perfilación de la Expresión Génica , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Trastornos Mieloproliferativos/enzimología , Trastornos Mieloproliferativos/patología , Reacción en Cadena en Tiempo Real de la Polimerasa
4.
Curr Opin Lipidol ; 24(2): 116-22, 2013 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-23314926

RESUMEN

PURPOSE OF REVIEW: Despite improved therapies, cardiovascular diseases are the leading cause of morbidity and mortality worldwide. Therefore, new therapeutic approaches are still needed. In the gene therapy field, RNA interference (RNAi) and regulation of microRNAs (miRNAs) have gained a lot of attention in addition to traditional overexpression based strategies. Here, recent findings in therapeutic gene silencing and modulation of small RNA expression related to atherogenesis and dyslipidemia are summarized. RECENT FINDINGS: Novel gene therapy approaches for the treatment of hyperlipidemia have been addressed. Antisense oligonucleotide and RNAi-based therapies against apolipoprotein B100 and proprotein convertase subtilisin/kexin type 9 have shown already efficacy in preclinical and clinical trials. In addition, several miRNAs dysregulated in atherosclerotic lesions and regulating cholesterol homeostasis have been found, which may represent novel targets for future therapies. SUMMARY: New therapies for lowering lipid levels are now being tested in clinical trials, and both antisense oligonucleotide and RNAi-based therapies have shown promising results in lowering cholesterol levels. However, the modulation of inflammatory component in atherosclerosis by gene therapy and targeting of the effects to plaques are still difficult challenges.


Asunto(s)
Aterosclerosis/terapia , Dislipidemias/terapia , Terapia Genética , Aterosclerosis/genética , HDL-Colesterol/genética , HDL-Colesterol/metabolismo , LDL-Colesterol/genética , LDL-Colesterol/metabolismo , Ensayos Clínicos como Asunto , Dislipidemias/genética , Epigénesis Genética , Humanos , MicroARNs/genética , MicroARNs/metabolismo , Neovascularización Patológica/genética , Neovascularización Patológica/metabolismo , Placa Aterosclerótica/genética , Placa Aterosclerótica/metabolismo , Interferencia de ARN , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo
5.
Commun Biol ; 7(1): 108, 2024 01 18.
Artículo en Inglés | MEDLINE | ID: mdl-38238517

RESUMEN

Treatment-induced neuroendocrine prostate cancer (t-NEPC) is a lethal subtype of castration-resistant prostate cancer resistant to androgen receptor (AR) inhibitors. Our study unveils that AR suppresses the neuronal development protein dihydropyrimidinase-related protein 5 (DPYSL5), providing a mechanism for neuroendocrine transformation under androgen deprivation therapy. Our unique CRPC-NEPC cohort, comprising 135 patient tumor samples, including 55 t-NEPC patient samples, exhibits a high expression of DPYSL5 in t-NEPC patient tumors. DPYSL5 correlates with neuroendocrine-related markers and inversely with AR and PSA. DPYSL5 overexpression in prostate cancer cells induces a neuron-like phenotype, enhances invasion, proliferation, and upregulates stemness and neuroendocrine-related markers. Mechanistically, DPYSL5 promotes prostate cancer cell plasticity via EZH2-mediated PRC2 activation. Depletion of DPYSL5 decreases proliferation, induces G1 phase cell cycle arrest, reverses neuroendocrine phenotype, and upregulates luminal genes. In conclusion, DPYSL5 plays a critical role in regulating prostate cancer cell plasticity, and we propose the AR/DPYSL5/EZH2/PRC2 axis as a driver of t-NEPC progression.


Asunto(s)
Neoplasias de la Próstata , Masculino , Humanos , Neoplasias de la Próstata/tratamiento farmacológico , Neoplasias de la Próstata/genética , Neoplasias de la Próstata/metabolismo , Antagonistas de Andrógenos , Próstata/patología , Hidrolasas , Proteínas Asociadas a Microtúbulos , Proteína Potenciadora del Homólogo Zeste 2/genética
7.
PLoS One ; 17(3): e0265948, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35358280

RESUMEN

MicroRNAs are well characterized in their role in silencing gene expression by targeting 3´-UTR of mRNAs in cytoplasm. However, recent studies have shown that miRNAs have a role in the regulation of genes in the nucleus, where they are abundantly located. We show here that in mouse endothelial cell line (C166), nuclear microRNA miR-466c participates in the regulation of vascular endothelial growth factor a (Vegfa) gene expression in hypoxia. Upregulation of Vegfa expression in response to hypoxia was significantly compromised after removal of miR-466c with CRISPR-Cas9 genomic deletion. We identified a promoter-associated long non-coding RNA on mouse Vegfa promoter and show that miR-466c directly binds to this transcript to modulate Vegfa expression. Collectively, these observations suggest that miR-466c regulates Vegfa gene transcription in the nucleus by targeting the promoter, and expands on our understanding of the role of miRNAs well beyond their canonical role.


Asunto(s)
MicroARNs , ARN Largo no Codificante , Factor A de Crecimiento Endotelial Vascular , Animales , Hipoxia/genética , Ratones , MicroARNs/genética , ARN Largo no Codificante/genética , ARN Mensajero , Factor A de Crecimiento Endotelial Vascular/metabolismo
8.
Cardiovasc Diabetol ; 10: 59, 2011 Jun 30.
Artículo en Inglés | MEDLINE | ID: mdl-21718508

RESUMEN

BACKGROUND: Lack of suitable mouse models has hindered the studying of diabetic macrovascular complications. We examined the effects of type 2 diabetes on coronary artery disease and cardiac function in hypercholesterolemic low-density lipoprotein receptor-deficient apolipoprotein B100-only mice (LDLR-/-ApoB100/100). METHODS AND RESULTS: 18-month-old LDLR-/-ApoB100/100 (n = 12), diabetic LDLR-/-ApoB100/100 mice overexpressing insulin-like growth factor-II (IGF-II) in pancreatic beta cells (IGF-II/LDLR-/-ApoB100/100, n = 14) and age-matched C57Bl/6 mice (n = 15) were studied after three months of high-fat Western diet. Compared to LDLR-/-ApoB100/100 mice, diabetic IGF-II/LDLR-/-ApoB100/100 mice demonstrated more calcified atherosclerotic lesions in aorta. However, compensatory vascular enlargement was similar in both diabetic and non-diabetic mice with equal atherosclerosis (cross-sectional lesion area ~60%) and consequently the lumen area was preserved. In coronary arteries, both hypercholesterolemic models showed significant stenosis (~80%) despite positive remodeling. Echocardiography revealed severe left ventricular systolic dysfunction and anteroapical akinesia in both LDLR-/-ApoB100/100 and IGF-II/LDLR-/-ApoB100/100 mice. Myocardial scarring was not detected, cardiac reserve after dobutamine challenge was preserved and ultrasructural changes revealed ischemic yet viable myocardium, which together with coronary artery stenosis and slightly impaired myocardial perfusion suggest myocardial hibernation resulting from chronic hypoperfusion. CONCLUSIONS: LDLR-/-ApoB100/100 mice develop significant coronary atherosclerosis, severe left ventricular dysfunction with preserved but diminished cardiac reserve and signs of chronic myocardial hibernation. However, the cardiac outcome is not worsened by type 2 diabetes, despite more advanced aortic atherosclerosis in diabetic animals.


Asunto(s)
Apolipoproteína B-100/metabolismo , Diabetes Mellitus Tipo 2/complicaciones , Angiopatías Diabéticas/complicaciones , Corazón/fisiopatología , Receptores de LDL/deficiencia , Disfunción Ventricular Izquierda/etiología , Disfunción Ventricular Izquierda/fisiopatología , Envejecimiento/fisiología , Animales , Apolipoproteína B-100/genética , Cardiotónicos/farmacología , Enfermedad de la Arteria Coronaria/etiología , Enfermedad de la Arteria Coronaria/fisiopatología , Diabetes Mellitus Tipo 2/genética , Diabetes Mellitus Tipo 2/metabolismo , Angiopatías Diabéticas/genética , Angiopatías Diabéticas/metabolismo , Dieta Alta en Grasa , Modelos Animales de Enfermedad , Dobutamina/farmacología , Femenino , Corazón/efectos de los fármacos , Hipercolesterolemia/complicaciones , Hipercolesterolemia/fisiopatología , Factor II del Crecimiento Similar a la Insulina/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Miocardio/patología , Miocardio/ultraestructura , Receptores de LDL/genética
9.
J Mater Chem B ; 8(2): 282-289, 2020 01 14.
Artículo en Inglés | MEDLINE | ID: mdl-31803886

RESUMEN

In this paper we describe a straightforward supramolecular strategy to encapsulate silicon phthalocyanine (SiPc) photosensitizers (PS) in polymeric micelles made of poly(ε-caprolactone)-b-methoxypoly(ethylene glycol) (PCL-PEG) block copolymers. While PCL-PEG micelles are promising nanocarriers based on their biocompatibility and biodegradability, the design of our new PS favors their encapsulation. In particular, they combine two axial benzoyl substituents, each of them carrying either three hydrophilic methoxy(triethylenoxy) chains (1), three hydrophobic dodecyloxy chains (3), or both kinds of chains (2). The SiPc derivatives 1 and 2 are therefore amphiphilic, with the SiPc unit contributing to the hydrophobic core, while lipophilicity increases along the series, making it possible to correlate the loading efficacy in PCL-PEG micelles with the hydrophobic/hydrophilic balance of the PS structure. This has led to a new kind of third-generation nano-PS that efficiently photogenerates 1O2, while preliminary in vitro experiments demonstrate an excellent cellular uptake and a promising PDT activity.


Asunto(s)
Indoles/química , Compuestos de Organosilicio/química , Fármacos Fotosensibilizantes/química , Portadores de Fármacos/química , Micelas , Poliésteres/química , Polietilenglicoles/química
10.
Genome Med ; 12(1): 99, 2020 11 20.
Artículo en Inglés | MEDLINE | ID: mdl-33218352

RESUMEN

BACKGROUND: Tight regulatory loops orchestrate commitment to B cell fate within bone marrow. Genetic lesions in this gene regulatory network underlie the emergence of the most common childhood cancer, acute lymphoblastic leukemia (ALL). The initial genetic hits, including the common translocation that fuses ETV6 and RUNX1 genes, lead to arrested cell differentiation. Here, we aimed to characterize transcription factor activities along the B-lineage differentiation trajectory as a reference to characterize the aberrant cell states present in leukemic bone marrow, and to identify those transcription factors that maintain cancer-specific cell states for more precise therapeutic intervention. METHODS: We compared normal B-lineage differentiation and in vivo leukemic cell states using single cell RNA-sequencing (scRNA-seq) and several complementary genomics profiles. Based on statistical tools for scRNA-seq, we benchmarked a workflow to resolve transcription factor activities and gene expression distribution changes in healthy bone marrow lymphoid cell states. We compared these to ALL bone marrow at diagnosis and in vivo during chemotherapy, focusing on leukemias carrying the ETV6-RUNX1 fusion. RESULTS: We show that lymphoid cell transcription factor activities uncovered from bone marrow scRNA-seq have high correspondence with independent ATAC- and ChIP-seq data. Using this comprehensive reference for regulatory factors coordinating B-lineage differentiation, our analysis of ETV6-RUNX1-positive ALL cases revealed elevated activity of multiple ETS-transcription factors in leukemic cells states, including the leukemia genome-wide association study hit ELK3. The accompanying gene expression changes associated with natural killer cell inactivation and depletion in the leukemic immune microenvironment. Moreover, our results suggest that the abundance of G1 cell cycle state at diagnosis and lack of differentiation-associated regulatory network changes during induction chemotherapy represent features of chemoresistance. To target the leukemic regulatory program and thereby overcome treatment resistance, we show that inhibition of ETS-transcription factors reduced cell viability and resolved pathways contributing to this using scRNA-seq. CONCLUSIONS: Our data provide a detailed picture of the transcription factor activities characterizing both normal B-lineage differentiation and those acquired in leukemic bone marrow and provide a rational basis for new treatment strategies targeting the immune microenvironment and the active regulatory network in leukemia.


Asunto(s)
Diferenciación Celular/genética , Proliferación Celular , Subunidad alfa 2 del Factor de Unión al Sitio Principal/genética , Leucemia/genética , Linfocitos/fisiología , Proteínas Proto-Oncogénicas c-ets/genética , Proteínas Represoras/genética , Médula Ósea , Línea Celular Tumoral , Niño , Subunidad alfa 2 del Factor de Unión al Sitio Principal/metabolismo , Sistemas de Liberación de Medicamentos , Regulación Neoplásica de la Expresión Génica , Predisposición Genética a la Enfermedad , Estudio de Asociación del Genoma Completo , Humanos , Leucemia/tratamiento farmacológico , Proteínas Proto-Oncogénicas c-ets/metabolismo , Proteínas Represoras/metabolismo , Factores de Transcripción , Transcriptoma , Translocación Genética , Proteína ETS de Variante de Translocación 6
11.
Front Cardiovasc Med ; 5: 16, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29594149

RESUMEN

Endothelial cell (EC) therapy may promote vascular growth or reendothelization in a variety of disease conditions. However, the production of a cell therapy preparation containing differentiated, dividing cells presenting typical EC phenotype, functional properties and chemokine profile is challenging. We focused on comparative analysis of seven small molecule-mediated differentiation protocols of ECs from human induced pluripotent stem cells. Differentiated cells showed a typical surface antigen pattern of ECs as characterized with flow cytometry analysis, functional properties, such as tube formation and ability to uptake acetylated LDL. Gene expression analysis by RNA sequencing revealed an efficient silencing of pluripotency genes and upregulation of genes related to cellular adhesion during differentiation. In addition, distinct patterns of transcription factor expression were identified during cellular reprogramming providing targets for more effective differentiation protocols in the future. Altogether, our results suggest that the most optimal EC differentiation protocol includes early inhibition of Rho-associated coiled-coil kinase and activation of cyclic AMP signaling, and inhibition of transforming growth factor beta signaling after mesodermal stage. These findings provide the first systematic characterization of the most potent signalling factors and small molecules used to generate ECs from human induced pluripotent stem cells and, consequently, this work improves the existing EC differentiation protocols and opens up new avenues for controlling cell fate for regenerative EC therapy.

12.
Eur J Pharm Biopharm ; 124: 63-72, 2018 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-29274374

RESUMEN

Photodynamic therapy (PDT) is an emerging method to treat light-accessible malignancies. To increase specificity and allow dose reduction, conjugates of photosensitizers (PS) with antibodies against tumor-associated antigens have been developed for photoimmunotherapy (PIT). However, so far it is unclear whether cellular internalization of these conjugates after binding affects PIT efficacy. The use of low molecular weight llama single domain antibodies (VHHs, nanobodies) for PIT is preferred above full size antibodies because of better tumor penetration. Therefore, we functionalized the VHH 7D12, directed against the epidermal growth factor receptor (EGFR), with a PS (IRDye700DX). To assess the impact of cellular internalization on activity, the VHHs were additionally conjugated to a cell-penetrating peptide (VHH[PS]-CPP). Here we show that upon illumination with near-infrared (NIR) light, both VHH[PS] and VHH[PS]-CPP conjugates specifically induce cell death of EGFR expressing cancer cell lines and of EGFR-expressing cells derived from surgically obtained ascites from patients with high-grade serous ovarian cancer. However, VHH[PS] conjugates were significantly more effective compared to internalizing VHH[PS]-CPP suggesting that cell surface association is required for optimal therapeutic activity.


Asunto(s)
Receptores ErbB/metabolismo , Inmunoconjugados/farmacología , Neoplasias Ováricas/tratamiento farmacológico , Fotoquimioterapia/métodos , Fármacos Fotosensibilizantes/farmacología , Anticuerpos de Dominio Único/metabolismo , Muerte Celular/efectos de los fármacos , Línea Celular Tumoral , Péptidos de Penetración Celular/química , Péptidos de Penetración Celular/metabolismo , Relación Dosis-Respuesta a Droga , Composición de Medicamentos , Endocitosis , Receptores ErbB/inmunología , Femenino , Humanos , Inmunoconjugados/química , Inmunoconjugados/inmunología , Inmunoconjugados/metabolismo , Nanomedicina/métodos , Neoplasias Ováricas/inmunología , Neoplasias Ováricas/metabolismo , Neoplasias Ováricas/patología , Fármacos Fotosensibilizantes/química , Fármacos Fotosensibilizantes/metabolismo , Anticuerpos de Dominio Único/química , Anticuerpos de Dominio Único/inmunología , Tecnología Farmacéutica/métodos
13.
Eur J Pharm Sci ; 107: 112-125, 2017 Sep 30.
Artículo en Inglés | MEDLINE | ID: mdl-28679107

RESUMEN

Selective elimination of macrophages by photodynamic therapy (PDT) is a new and promising therapeutic modality for the reduction of atherosclerotic plaques. m-Tetra(hydroxyphenyl)chlorin (mTHPC, or Temoporfin) may be suitable as photosensitizer for this application, as it is currently used in the clinic for cancer PDT. In the present study, mTHPC was encapsulated in polymeric micelles based on benzyl-poly(ε-caprolactone)-b-methoxy poly(ethylene glycol) (Ben-PCL-mPEG) using a film hydration method, with loading capacity of 17%. Because of higher lipase activity in RAW264.7 macrophages than in C166 endothelial cells, the former cells degraded the polymers faster, resulting in faster photosensitizer release and higher in vitro photocytotoxicity of mTHPC-loaded micelles in those macrophages. However, we observed release of mTHPC from the micelles in 30min in blood plasma in vitro which explains the observed similar in vivo pharmacokinetics of the mTHPC micellar formulation and free mTHPC. Therefore, we could not translate the beneficial macrophage selectivity from in vitro to in vivo. Nevertheless, we observed accumulation of mTHPC in atherosclerotic lesions of mice aorta's which is probably the result of binding to lipoproteins upon release from the micelles. Therefore, future experiments will be dedicated to increase the stability and thus allow accumulation of intact mTHPC-loaded Ben-PCL-mPEG micelles to macrophages of atherosclerotic lesions.


Asunto(s)
Enfermedades Cardiovasculares/tratamiento farmacológico , Mesoporfirinas/administración & dosificación , Micelas , Fármacos Fotosensibilizantes/administración & dosificación , Animales , Enfermedades Cardiovasculares/metabolismo , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/efectos de la radiación , Células Endoteliales/efectos de los fármacos , Células Endoteliales/metabolismo , Células Endoteliales/efectos de la radiación , Femenino , Luz , Mesoporfirinas/sangre , Mesoporfirinas/farmacocinética , Mesoporfirinas/uso terapéutico , Ratones , Ratones Endogámicos BALB C , Ratones Noqueados , Ratones Desnudos , Fotoquimioterapia , Fármacos Fotosensibilizantes/sangre , Fármacos Fotosensibilizantes/farmacocinética , Fármacos Fotosensibilizantes/uso terapéutico , Poliésteres/administración & dosificación , Poliésteres/farmacocinética , Poliésteres/uso terapéutico , Células RAW 264.7 , Oxígeno Singlete/química , Distribución Tisular
14.
Oncotarget ; 7(44): 71594-71607, 2016 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-27689404

RESUMEN

Diffuse gliomas are primary brain cancers that are characterised by infiltrative growth. Whereas high-grade glioma characteristically presents with perinecrotic neovascularisation, large tumor areas thrive on pre-existent vasculature as well. Clinical studies have revealed that pharmacological inhibition of the angiogenic process does not improve survival of glioblastoma patients. Direct targeting of tumor vessels may however still be an interesting therapeutic approach as it allows pinching off the blood supply to tumor cells. Such tumor vessel targeting requires the identification of tumor-specific vascular targeting agents (TVTAs).Here we describe a novel TVTA, C-C7, which we identified via in vivo biopanning of a llama nanobody phage display library in an orthotopic mouse model of diffuse glioma. We show that C-C7 recognizes a subpopulation of tumor blood vessels in glioma xenografts and clinical glioma samples. Additionally, C-C7 recognizes macrophages and activated endothelial cells in atherosclerotic lesions. By using C-C7 as bait in yeast-2-hybrid (Y2H) screens we identified dynactin-1-p150Glued as its binding partner. The interaction was confirmed by co-immunostainings with C-C7 and a commercial anti-dynactin-1-p150Glued antibody, and via co-immunoprecipitation/western blot studies. Normal brain vessels do not express dynactin-1-p150Glued and its expression is reduced under anti-VEGF therapy, suggesting that dynactin-1-p150Glued is a marker for activated endothelial cells.In conclusion, we show that in vivo phage display combined with Y2H screenings provides a powerful approach to identify tumor-targeting nanobodies and their binding partners. Using this combination of methods we identify dynactin-1-p150Glued as a novel targetable protein on activated endothelial cells and macrophages.


Asunto(s)
Neoplasias Encefálicas/irrigación sanguínea , Técnicas de Visualización de Superficie Celular/métodos , Complejo Dinactina/inmunología , Glioblastoma/irrigación sanguínea , Anticuerpos de Dominio Único/uso terapéutico , Animales , Neoplasias Encefálicas/terapia , Células Endoteliales/fisiología , Glioblastoma/terapia , Humanos , Inmunohistoquímica , Macrófagos/fisiología , Ratones , Técnicas del Sistema de Dos Híbridos
15.
Cardiovasc Res ; 99(4): 716-23, 2013 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-23756254

RESUMEN

AIMS: The role of vascular endothelial growth factor (VEGF-A) in atherogenesis has remained controversial. We addressed this by comparing the effects of adenoviral VEGF-A gene transfer on atherosclerosis and lipoproteins in ApoE(-/-), LDLR(-/-), LDLR(-/-)ApoE(-/-), and LDLR(-/-)ApoB(100/100) mice. METHODS AND RESULTS: After 4 weeks on western diet, systemic adenoviral gene transfer was performed with hVEGF-A or control vectors. Effects on atherosclerotic lesion area and composition, lipoprotein profiles, and plasma lipoprotein lipase (LPL) activity were examined. On day 4, VEGF-A induced alterations in lipoprotein profiles and a significant negative correlation was observed between plasma LPL activity and VEGF-A levels. One month after gene transfer, no changes in atherosclerosis were observed in LDLR(-/-) and LDLR(-/-)ApoB(100/100) models, whereas both ApoE(-/-) models displayed increased en face lesion areas in thoracic and abdominal aortas. VEGF-A also reduced LPL mRNA in heart and white adipose tissue, whereas Angptl4 was increased, potentially providing further mechanistic explanation for the findings. CONCLUSION: VEGF-A gene transfer induced pro-atherogenic changes in lipoprotein profiles in all models. As a novel finding, VEGF-A also reduced LPL activity, which might underlie the observed changes in lipid profiles. However, VEGF-A was observed to increase atherosclerosis only in the ApoE(-/-) background, clearly indicating some mouse model-specific effects.


Asunto(s)
Aterosclerosis/etiología , Terapia Genética , Hiperlipidemias/terapia , Lipoproteína Lipasa/metabolismo , Lipoproteínas/sangre , Factor A de Crecimiento Endotelial Vascular/genética , Adenoviridae/genética , Animales , Apolipoproteínas B/deficiencia , Apolipoproteínas B/fisiología , Modelos Animales de Enfermedad , Femenino , Hiperlipidemias/sangre , Hiperlipidemias/patología , Lípidos/sangre , Lipoproteína Lipasa/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Receptores de LDL/fisiología
16.
Cardiovasc Res ; 98(1): 107-15, 2013 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-23341579

RESUMEN

AIMS: The loss of nuclear factor E2-related factor 2 (Nrf2) has been shown to protect against atherogenesis in apoE-deficient mice. The mechanism by which Nrf2 deficiency affords atheroprotection in this model is currently unknown, but combined systemic and local vascular effects on lesion macrophages have been proposed. We investigated the effect of bone marrow-specific loss of Nrf2 on early atherogenesis in low-density lipoprotein (LDL) receptor-deficient (LDLR(-/-)) mice, and assessed the effect of Nrf2 on cellular accumulation of modified LDLs and the expression of inflammatory markers in macrophages. METHODS AND RESULTS: The effect of bone marrow-specific loss of Nrf2 on atherogenesis was studied using bone marrow transplantation of wild-type (WT) or Nrf2(-/-) bone marrow to LDLR(-/-) mice. Mice transplanted with Nrf2(-/-) bone marrow and fed a high-fat diet for 6 weeks exhibited significantly larger atherosclerotic lesions than WT bone marrow transplanted mice. Moreover, in thioglycollate-elicited Nrf2(-/-) macrophages, the uptake of acetylated and malondialdehyde-modified LDLs was increased in comparison with WT controls, with the concomitant increase in the expression of scavenger receptor A and toll-like receptor 4. In addition, the expression of pro-inflammatory monocyte chemoattractant protein-1 and interleukin-6 were increased in Nrf2(-/-) vs. WT macrophages. CONCLUSION: Nrf2 deficiency specific to bone marrow-derived cells aggravates atherosclerosis in LDLR(-/-) mice. Furthermore, the loss of Nrf2 in macrophages enhances foam cell formation and promotes the pro-inflammatory phenotype.


Asunto(s)
Aterosclerosis/etiología , Macrófagos/fisiología , Factor 2 Relacionado con NF-E2/fisiología , Animales , Quimiocina CCL2/genética , Colesterol/metabolismo , Femenino , Ratones , Ratones Endogámicos C57BL , Receptores de LDL/fisiología , Receptores Depuradores/análisis
17.
Cardiovasc Res ; 88(3): 530-8, 2010 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-20634212

RESUMEN

AIMS: Macrophage scavenger receptor A (SR-A) and class B scavenger receptor CD36 (CD36) contribute to foam cell formation and atherogenesis via uptake of modified lipoproteins. So far, the role of these scavenger receptors has been studied mainly using knockout models totally lacking these receptors. We studied the role of SR-A and CD36 in foam cell formation and atherogenesis by RNA interference (RNAi)-mediated silencing, which is a clinically feasible method to down-regulate the expression of these receptors. METHODS AND RESULTS: We constructed lentivirus vectors encoding short hairpin RNAs (shRNAs) against mouse SR-A and CD36. Decreased SR-A but not CD36 expression led to reduced foam cell formation caused by acetylated low-density lipoprotein (LDL) in mouse macrophages, whereas the uptake of oxidized LDL was not altered. More importantly, silencing of SR-A upregulates CD36 and vice versa. In LDL receptor-deficient apolipoprotein B100 (LDLR(-/-)ApoB(100/100)) mice kept on a western diet, silencing of either SR-A or CD36 in bone marrow cells led to a marked decrease (37.4 and 34.2%, respectively) in cross-sectional lesion area, whereas simultaneous silencing of both receptors was not effective. CONCLUSION: Our results suggest that silencing of either SR-A or CD36 alone reduces atherogenesis in mice. However, due to reciprocal upregulation, silencing of both SR-A and CD36 is not effective.


Asunto(s)
Aterosclerosis/prevención & control , Antígenos CD36/genética , Hiperlipidemias/complicaciones , Interferencia de ARN , Receptores Depuradores de Clase A/genética , Regulación hacia Arriba/genética , Animales , Apolipoproteínas B/genética , Apolipoproteínas B/metabolismo , Aterosclerosis/genética , Aterosclerosis/metabolismo , Antígenos CD36/metabolismo , Línea Celular , Modelos Animales de Enfermedad , Femenino , Células Espumosas/metabolismo , Células Espumosas/patología , Humanos , Hiperlipidemias/metabolismo , Hiperlipidemias/patología , Lentivirus/genética , Macrófagos/metabolismo , Macrófagos/patología , Masculino , Ratones , Ratones Noqueados , Receptores de LDL/genética , Receptores de LDL/metabolismo , Receptores Depuradores de Clase A/metabolismo
18.
Atherosclerosis ; 213(1): 122-8, 2010 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-20688330

RESUMEN

OBJECTIVE: Endothelial lipase (EL) is a new member of triacylglycerol lipase family that has been shown to decrease high-density lipoprotein (HDL) cholesterol levels leading to increased risk of atherosclerosis. Its expression is increased during inflammation and by inflammatory cytokines. Sulforaphane (SFN) is a naturally occurring isothiocyanate present in cruciferous vegetables that has antioxidant and anti-inflammatory effects. Nuclear factor (NF)-κB is one of the molecular targets for SFN-mediated protective effects. Our aim was therefore to assess whether SFN could impact on EL expression via modulation of NF-κB pathway. METHODS AND RESULTS: Quantitative PCR and Western blot results demonstrated that SFN inhibited tumor necrosis factor (TNF)-α-mediated induction of EL in human umbilical vein endothelial cells (HUVEC). Lentiviral transduction of HUVEC with mutated form of IκB-α (IκBM) as well as silencing of NF-κB subunit p65 using RNA interference revealed that TNF-α-mediated induction of EL is mediated through NF-κB pathway. In addition, a total of five NF-κB binding sites were found in LIPG gene, which encodes EL. SFN inhibited binding of NF-κB to these sites analyzed by chromatin immunoprecipitation (ChIP) and electrophoretic mobility shift assay (EMSA). SFN also inhibited TNF-α mediated phosphorylation of IκB kinase (IKK) 1/2 and IκB-α. CONCLUSIONS: Collectively, these results indicate that SFN inhibits EL expression via inhibition of NF-κB which may have a beneficial effect on HDL cholesterol levels.


Asunto(s)
Células Endoteliales/citología , Regulación Enzimológica de la Expresión Génica , Lipasa/antagonistas & inhibidores , FN-kappa B/metabolismo , Tiocianatos/farmacología , Antioxidantes/química , Aterosclerosis/metabolismo , Células Cultivadas , Humanos , Quinasa I-kappa B/metabolismo , Isotiocianatos/química , Lipasa/biosíntesis , Modelos Biológicos , Unión Proteica , Sulfóxidos , Venas Umbilicales/citología , Verduras
19.
J Gene Med ; 8(4): 433-41, 2006 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-16389634

RESUMEN

BACKGROUND: RNA interference (RNAi) is a post-transcriptional RNA degradation process, which has become a very useful tool in gene function studies and gene therapy applications. Long-term cellular expression of small interfering RNA (siRNA) molecules required for many gene therapy applications can be achieved by lentiviral vectors (LVs). The two most commonly used promoters to drive the short hairpin RNA (shRNA) expression are the human U6 small nuclear promoter (U6) and the human H1 promoter (H1). METHODS: We investigated whether there is any significant difference between the efficiencies of U6 and H1 in LV-mediated RNAi using green fluorescent protein (GFP) as a target gene by flow cytometry and real-time reverse-transcription polymerase chain reaction (RT-PCR) in endothelial cells. Also, we compared the efficiencies of U6 and H1 in the GFP transgenic mouse brain after stereotactic LV injection. RESULTS: We show that the U6 promoter is more efficient than H1 in GFP silencing in vitro, leading to 80% GFP knockdown at an average of one integrated vector genome per target cell genome. The silencing is persistent for several months. In addition, the U6 promoter is superior to H1 in vivo and leads to stable GFP knockdown in mouse brain for at least 9 months. CONCLUSIONS: These results show that LV-mediated RNAi is a powerful gene-silencing method for the long-term inhibition of gene expression in vitro and in vivo.


Asunto(s)
Encéfalo/metabolismo , Células Endoteliales/metabolismo , Regiones Promotoras Genéticas , Interferencia de ARN , Animales , Encéfalo/citología , Línea Celular , Expresión Génica , Gliceraldehído 3-Fosfato/metabolismo , Lentivirus/genética , Ratones , ARN Mensajero/metabolismo , Transducción Genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA