Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 114
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Proc Natl Acad Sci U S A ; 119(41): e2204758119, 2022 10 11.
Artículo en Inglés | MEDLINE | ID: mdl-36191215

RESUMEN

Obesity is associated with an increased risk of, and a poor prognosis for, postmenopausal (PM) breast cancer (BC). Our goal was to determine whether diet-induced obesity (DIO) promotes 1) shorter tumor latency, 2) an escape from tumor dormancy, and 3) an acceleration of tumor growth and to elucidate the underlying mechanism(s). We have developed in vitro assays and PM breast tumor models complemented by a noninvasive imaging system to detect vascular invasion of dormant tumors and have used them to determine whether obesity promotes the escape from breast tumor dormancy and tumor growth by facilitating the switch to the vascular phenotype (SVP) in PM BC. Obese mice had significantly higher tumor frequency, higher tumor volume, and lower overall survival compared with lean mice. We demonstrate that DIO exacerbates mammary gland hyperplasia and neoplasia, reduces tumor latency, and increases tumor frequency via an earlier acquisition of the SVP. DIO establishes a local and systemic proangiogenic and inflammatory environment via the up-regulation of lipocalin-2 (LCN2), vascular endothelial growth factor (VEGF), and basic fibroblast growth factor (bFGF) that may promote the escape from tumor dormancy and tumor progression. In addition, we show that targeting neovascularization via a multitargeted receptor tyrosine kinase inhibitor, sunitinib, can delay the acquisition of the SVP, thereby prolonging tumor latency, reducing tumor frequency, and increasing tumor-free survival, suggesting that targeting neovascularization may be a potential therapeutic strategy in obesity-associated PM BC progression. This study establishes the link between obesity and PM BC and, for the first time to our knowledge, bridges the dysfunctional neovascularization of obesity with the earliest stages of tumor development.


Asunto(s)
Factor 2 de Crecimiento de Fibroblastos , Neoplasias Mamarias Experimentales , Menopausia , Obesidad , Factor A de Crecimiento Endotelial Vascular , Animales , Femenino , Factor 2 de Crecimiento de Fibroblastos/metabolismo , Lipocalina 2 , Neoplasias Mamarias Experimentales/fisiopatología , Ratones , Ratones Endogámicos C57BL , Ratones Obesos , Neovascularización Patológica/patología , Obesidad/genética , Inhibidores de Proteínas Quinasas , Sunitinib , Factor A de Crecimiento Endotelial Vascular/metabolismo , Factores de Crecimiento Endotelial Vascular/metabolismo
2.
Mol Cell Proteomics ; 21(1): 100176, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-34774759

RESUMEN

Urologic chronic pelvic pain syndrome (UCPPS) is a condition of unknown etiology characterized by pelvic pain and urinary frequency and/or urgency. As the proximal fluid of this syndrome, urine is an ideal candidate sample matrix for an unbiased study of UCPPS. In this study, a large, discovery-phase, TMT-based quantitative urinary proteomics analysis of 244 participants was performed. The participants included patients with UCPPS (n = 82), healthy controls (HC) (n = 94), and disparate chronic pain diseases, termed positive controls (PC) (n = 68). Using training and testing cohorts, we identified and validated a small and distinct set of proteins that distinguished UCPPS from HC (n = 9) and UCPPS from PC (n = 3). The validated UCPPS: HC proteins were predominantly extracellular matrix/extracellular matrix modifying or immunomodulatory/host defense in nature. Significantly varying proteins in the UCPPS: HC comparison were overrepresented by the members of several dysregulated biological processes including decreased immune cell migration, decreased development of epithelial tissue, and increased bleeding. Comparison with the PC cohort enabled the evaluation of UCPPS-specific upstream regulators, contrasting UCPPS with other conditions that cause chronic pain. Specific to UCPPS were alterations in the predicted signaling of several upstream regulators, including alpha-catenin, interleukin-6, epidermal growth factor, and transforming growth factor beta 1, among others. These findings advance our knowledge of the etiology of UCPPS and inform potential future clinical translation into a diagnostic panel for UCPPS.


Asunto(s)
Dolor Crónico , Enfermedad Crónica , Humanos , Dolor Pélvico/diagnóstico , Dolor Pélvico/etiología , Proteómica , Síndrome
3.
Proc Natl Acad Sci U S A ; 116(37): 18295-18303, 2019 09 10.
Artículo en Inglés | MEDLINE | ID: mdl-31451668

RESUMEN

Triple-negative breast cancer (TNBC), which has the highest mortality rate of all breast cancer, is in urgent need of a therapeutic that hinders the spread and growth of cancer cells. CRISPR genome editing holds the promise of a potential cure for many genetic diseases, including TNBC; however, its clinical translation is being challenged by the lack of safe and effective nonviral delivery systems for in vivo therapeutic genome editing. Here we report the synthesis and application of a noncationic, deformable, and tumor-targeted nanolipogel system (tNLG) for CRISPR genome editing in TNBC tumors. We have demonstrated that tNLGs mediate a potent CRISPR knockout of Lipocalin 2 (Lcn2), a known breast cancer oncogene, in human TNBC cells in vitro and in vivo. The loss of Lcn2 significantly inhibits the migration and the mesenchymal phenotype of human TNBC cells and subsequently attenuates TNBC aggressiveness. In an orthotopic TNBC model, we have shown that systemically administered tNLGs mediated >81% CRISPR knockout of Lcn2 in TNBC tumor tissues, resulting in significant tumor growth suppression (>77%). Our proof-of-principle results provide experimental evidence that tNLGs can be used as a safe, precise, and effective delivery approach for in vivo CRISPR genome editing in TNBC.


Asunto(s)
Portadores de Fármacos/química , Sistemas de Liberación de Medicamentos/métodos , Edición Génica , Nanopartículas/química , Neoplasias de la Mama Triple Negativas/tratamiento farmacológico , Neoplasias de la Mama Triple Negativas/genética , Animales , Sistemas CRISPR-Cas , Línea Celular Tumoral , Movimiento Celular , Modelos Animales de Enfermedad , Femenino , Regulación Neoplásica de la Expresión Génica , Técnicas de Silenciamiento del Gen , Terapia Genética/métodos , Vectores Genéticos , Humanos , Molécula 1 de Adhesión Intercelular/genética , Lípidos/química , Lipocalina 2/genética , Liposomas/química , Ratones , Ratones Desnudos
4.
Adv Funct Mater ; 31(44)2021 Oct 26.
Artículo en Inglés | MEDLINE | ID: mdl-34924915

RESUMEN

To date, the scaled-up manufacturing and efficient drug loading of exosomes are two existing challenges limiting the clinical translation of exosome-based drug delivery. Herein, we developed a facile magnetic extrusion method for preparing endosome-derived vesicles, also known as exosome mimetics (EMs), which share the same biological origin and similar morphology, composition, and biofunctions with native exosomes. The high yield and consistency of this magnetic extrusion method help to overcome the manufacturing bottleneck in exosome research. Moreover, the proposed standardized multi-step method readily facilitates the ammonium sulfate gradient approach to actively load chemodrugs such as doxorubicin into EMs. The engineered EMs developed and tested here exhibit comparable drug delivery properties as do native exosomes and potently inhibit tumor growth by delivering doxorubicin in an orthotopic breast tumor model. These findings demonstrate that EMs can be prepared in a facile and scaled-up manner as a promising biological nanomedicine for cancer drug delivery.

5.
J Urol ; 205(2): 514-523, 2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-33026902

RESUMEN

PURPOSE: We analyzed a series of novel noninvasive urinary biomarkers for their ability to objectively monitor the longitudinal clinical status of patients with urological chronic pelvic pain syndrome. MATERIALS AND METHODS: Baseline, 6 and 12-month urine samples were collected (216) and used to quantify vascular endothelial growth factor, vascular endothelial growth factor (VEGF) receptor 1 (R1), neutrophil gelatinase associated lipocalin (NGAL), matrix metalloproteinase-2, matrix metalloproteinase (MMP)-9, and MMP-9/NGAL complex by enzyme-linked immunosorbent assays. Patient symptom changes were classified as improved, stable or worse using a functional clustering algorithm. Proportional odds models were used to evaluate the association between symptom change and urinary biomarkers. RESULTS: Across all sampled participants, longitudinal decreases in normalized VEGF concentration (pg/µg) were associated with pain severity improvement, and decreases in MMP-9, NGAL and VEGF-R1 concentration (pg/ml) as well as NGAL normalized concentration were associated with improved urinary symptoms. Longitudinal decreases in normalized VEGF-R1 were associated with pain improvement in patients with moderate widespreadness, no bladder symptoms and no painful filling. Lower baseline normalized VEGF-R1 concentration was associated with pain improvement in patients with pelvic pain only. Higher baseline MMP-9/NGAL levels were associated with pain and urinary improvement across all participants. Moreover, longitudinal increases in MMP-2 concentration was associated with improved pain in men and patients with painful filling. CONCLUSIONS: Our results suggest these urinary biomarkers may be useful in monitoring urological chronic pelvic pain syndrome symptom changes with respect to both urinary severity and pain severity. With further testing, they may represent objective biological measures of urological chronic pelvic pain syndrome progression and/or resolution while also providing insight into the pathophysiology of urological chronic pelvic pain syndrome.


Asunto(s)
Dolor Crónico/orina , Dolor Pélvico/orina , Enfermedades Urológicas/orina , Biomarcadores/orina , Femenino , Humanos , Estudios Longitudinales , Masculino , Síndrome
6.
Int J Mol Sci ; 21(11)2020 May 28.
Artículo en Inglés | MEDLINE | ID: mdl-32481745

RESUMEN

Breast cancer brain metastasis is a major clinical challenge and is associated with a dismal prognosis. Understanding the mechanisms underlying the early stages of brain metastasis can provide opportunities to develop efficient diagnostics and therapeutics for this significant clinical challenge. We have previously reported that breast cancer-derived extracellular vesicles (EVs) breach the blood-brain barrier (BBB) via transcytosis and can promote brain metastasis. Here, we elucidate the functional consequences of EV transport across the BBB. We demonstrate that brain metastasis-promoting EVs can be internalized by astrocytes and modulate the behavior of these cells to promote extracellular matrix remodeling in vivo. We have identified protein and miRNA signatures in these EVs that can lead to the interaction of EVs with astrocytes and, as such, have the potential to serve as targets for development of diagnostics and therapeutics for early detection and therapeutic intervention in breast cancer brain metastasis.


Asunto(s)
Astrocitos/metabolismo , Barrera Hematoencefálica , Neoplasias de la Mama/metabolismo , Vesículas Extracelulares/metabolismo , MicroARNs/genética , Proteína de Unión al GTP cdc42/metabolismo , Animales , Neoplasias de la Mama/genética , Línea Celular Tumoral , Análisis por Conglomerados , Medios de Cultivo Condicionados/metabolismo , Endocitosis , Matriz Extracelular/metabolismo , Femenino , Humanos , Ratones , Ratones Desnudos , Metástasis de la Neoplasia , Pronóstico , Proteómica , Inhibidor Tisular de Metaloproteinasa-2/metabolismo
7.
Cytometry A ; 95(8): 843-853, 2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-31294926

RESUMEN

A rapid, high-throughput, and quantitative method for cell entry route characterization is still lacking in nanomedicine research. Here, we report the application of imaging flow cytometry for quantitatively analyzing cell entry routes of actively targeted nanomedicines. We first engineered ICAM1 antibody-directed fusogenic nanoliposomes (ICAM1-FusoNLPs) and ICAM1 antibody-directed endocytic nanolipogels (ICAM1-EndoNLGs) featuring highly similar surface properties but different cell entry routes: receptor-mediated membrane fusion and receptor-mediated endocytosis, respectively. By using imaging flow cytometry, we characterized their intracellular delivery into human breast cancer MDA-MB-231 cells. We found that ICAM1-FusoNLPs mediated a 2.8-fold increased cell uptake of fluorescent payload, FITC-dextran, with a 2.4-fold increased intracellular distribution area in comparison with ICAM1-EndoNLGs. We also investigated the effects of incubation time and endocytic inhibitors on the cell entry routes of ICAM1-FusoNLP and ICAM1-EndoNLG. Our results indicate that receptor-mediated membrane fusion is a faster and more efficient cell entry route than receptor-mediated endocytosis, bringing with it a significant therapeutic benefit in a proof-of-principle nanomedicine-mediated siRNA transfection experiment. Our studies suggest that cell entry route may be an important design parameter to be considered in the development of next-generation nanomedicines. © 2019 International Society for Advancement of Cytometry.


Asunto(s)
Endocitosis/genética , Citometría de Flujo , Molécula 1 de Adhesión Intercelular/ultraestructura , Liposomas/química , Anticuerpos/química , Línea Celular Tumoral , Humanos , Molécula 1 de Adhesión Intercelular/química , Molécula 1 de Adhesión Intercelular/genética , Liposomas/ultraestructura , Nanomedicina , ARN Interferente Pequeño/química , ARN Interferente Pequeño/ultraestructura , Internalización del Virus
8.
Small ; 14(18): e1703493, 2018 05.
Artículo en Inglés | MEDLINE | ID: mdl-29468837

RESUMEN

Template synthesis represents an important class of nanofabrication methods. Herein, recent advances in nanomaterial preparation by extrusion through nanoporous membranes that preserve the template membrane without sacrificing it, which is termed as "non-sacrificing template synthesis," are reviewed. First, the types of nanoporous membranes used in nanoporous membrane extrusion applications are introduced. Next, four common nanoporous membrane extrusion strategies: vesicle extrusion, membrane emulsification, precipitation extrusion, and biological membrane extrusion, are examined. These methods have been utilized to prepare a wide range of nanomaterials, including liposomes, emulsions, nanoparticles, nanofibers, and nanotubes. The principle and historical context of each specific technology are discussed, presenting prominent examples and evaluating their positive and negative features. Finally, the current challenges and future opportunities of nanoporous membrane extrusion methods are discussed.

9.
Pediatr Res ; 83(5): 982-992, 2018 05.
Artículo en Inglés | MEDLINE | ID: mdl-29342131

RESUMEN

BackgroundHutchinson-Gilford progeria syndrome (HGPS) is an ultra-rare, fatal, segmental premature aging syndrome caused by the aberrant lamin A protein, progerin. The protein farnesyltransferase inhibitor, lonafarnib, ameliorates some aspects of cardiovascular and bone disease.MethodsWe performed a prospective longitudinal survey of plasma proteins in 24 children with HGPS (an estimated 10% of the world's population at the time) at baseline and on lonafarnib therapy, compared with age- and gender-matched controls using a multi-analyte, microsphere-based immunofluorescent assay.ResultsThe mean levels for 23/66 (34.8%) proteins were significantly lower and 7/66 (10.6%) were significantly higher in HGPS samples compared with those in controls (P≤0.05). Six proteins whose concentrations were initially lower normalized with lonafarnib therapy: interleukins 1α, 7, and 13, beta-2 microglobulin, C-reactive protein, and myoglobin. Alpha-2 macroglobulin, a protease inhibitor associated with stroke, was elevated at baseline and subsequently normalized with lonafarnib therapy.ConclusionThis is the first study to employ a multi-analyte array platform in HGPS. Novel potential biomarkers identified in this study should be further validated by correlations with clinical disease status, especially proteins associated with cardiovascular disease and those that normalized with lonafarnib therapy.


Asunto(s)
Proteínas Sanguíneas/análisis , Piperidinas/uso terapéutico , Progeria/sangre , Progeria/tratamiento farmacológico , Piridinas/uso terapéutico , Adolescente , Biomarcadores/sangre , Proteína C-Reactiva/análisis , Estudios de Casos y Controles , Niño , Preescolar , Inhibidores Enzimáticos/farmacología , Femenino , Humanos , Interleucina-13/sangre , Interleucina-1alfa/sangre , Interleucina-7/sangre , Lamina Tipo A , Estudios Longitudinales , Masculino , Mutación , Mioglobina/sangre , Piperidinas/sangre , Estudios Prospectivos , Piridinas/sangre , Microglobulina beta-2/sangre
10.
Cytometry A ; 91(5): 424-432, 2017 05.
Artículo en Inglés | MEDLINE | ID: mdl-28314083

RESUMEN

The switch of tumor cells from a dormant, non-angiogenic phenotype to an active, angiogenic phenotype is a critical step in early cancer progression. To date, relatively little is known about the cellular behaviors of angiogenic and non-angiogenic tumor cell phenotypes. In this study, holographic imaging cytometry, a quantitative phase imaging (QPI) technique was used to continuously and non-invasively analyze, quantify, and compare a panel of fundamental cellular behaviors of angiogenic and non-angiogenic human osteosarcoma cells (KHOS) in a simple and economical way. Results revealed that angiogenic KHOS cells (KHOS-A) have significantly higher cell motility speeds than their non-angiogenic counterpart (KHOS-N) while no difference in their cell proliferation rates and cell cycle lengths were observed. KHOS-A cells were also found to have significantly smaller cell areas and greater cell optical thicknesses when compared with the non-angiogenic KHOS-N cells. No difference in average cell volumes was observed. These studies demonstrate that the morphology and behavior of angiogenic and non-angiogenic cells can be continuously, efficiently, and non-invasively monitored using a simple, quantitative, and economical system that does not require tedious and time-consuming assays to provide useful information about tumor dormancy. © 2017 International Society for Advancement of Cytometry.


Asunto(s)
Movimiento Celular/genética , Citometría de Flujo/métodos , Holografía/métodos , Neovascularización Patológica/genética , Osteosarcoma/genética , Línea Celular Tumoral , Linaje de la Célula/genética , Proliferación Celular/genética , Humanos , Osteosarcoma/patología
11.
BJU Int ; 120(1): 130-142, 2017 07.
Artículo en Inglés | MEDLINE | ID: mdl-28263447

RESUMEN

OBJECTIVE: To examine a series of candidate markers for urological chronic pelvic pain syndrome (UCPPS), selected based on their proposed involvement in underlying biological processes so as to provide new insights into pathophysiology and suggest targets for expanded clinical and mechanistic studies. METHODS: Baseline urine samples from Multidisciplinary Approach to the Study of Chronic Pelvic Pain (MAPP) Research Network study participants with UCPPS (n = 259), positive controls (PCs; chronic pain without pelvic pain, n = 107) and healthy controls (HCs, n = 125) were analysed for the presence of proteins that are suggested in the literature to be associated with UCPPS. Matrix metalloproteinase (MMP)-2, MMP-9, MMP-9/neutrophil gelatinase-associated lipocalin (NGAL) complex (also known as Lipocalin 2), vascular endothelial growth factor (VEGF), VEGF receptor 1 (VEGF-R1) and NGAL were assayed and quantitated using mono-specific enzyme-linked immunosorbent assays for each protein. Log-transformed concentration (pg/mL or ng/mL) and concentration normalized to total protein (pg/µg) values were compared among the UCPPS, PC and HC groups within sex using the Student's t-test, with P values adjusted for multiple comparisons. Multivariable logistic regression and receiver-operating characteristic curves assessed the utility of the biomarkers in distinguishing participants with UCPPS and control participants. Associations of protein with symptom severity were assessed by linear regression. RESULTS: Significantly higher normalized concentrations (pg/µg) of VEGF, VEGF-R1 and MMP-9 in men and VEGF concentration (pg/mL) in women were associated with UCPPS vs HC. These proteins provided only marginal discrimination between UCPPS participants and HCs. In men with UCCPS, pain severity was significantly positively associated with concentrations of MMP-9 and MMP-9/NGAL complex, and urinary severity was significantly positively associated with MMP-9, MMP-9/NGAL complex and VEGF-R1. In women with UCPPS, pain and urinary symptom severity were associated with increased normalized concentrations of MMP-9/NGAL complex, while pain severity alone was associated with increased normalized concentrations of VEGF, and urinary severity alone was associated with increased normalized concentrations of MMP-2. Pain severity in women with UCPPS was significantly positively associated with concentrations of all biomarkers except NGAL, and urinary severity with all concentrations except VEGF-R1. CONCLUSION: Altered levels of MMP-9, MMP-9/NGAL complex and VEGF-R1 in men, and all biomarkers in women, were associated with clinical symptoms of UCPPS. None of the evaluated candidate markers usefully discriminated UCPPS patients from controls. Elevated VEGF, MMP-9 and VEGF-R1 levels in men and VEGF levels in women may provide potential new insights into the pathophysiology of UCPPS.


Asunto(s)
Metaloproteinasa 9 de la Matriz/metabolismo , Dolor Pélvico/fisiopatología , Dolor Pélvico/psicología , Sistema Urinario/patología , Enfermedades Urológicas/metabolismo , Factor A de Crecimiento Endotelial Vascular/metabolismo , Adulto , Biomarcadores/metabolismo , Investigación Biomédica , Dolor Crónico , Ensayo de Inmunoadsorción Enzimática , Femenino , Humanos , Comunicación Interdisciplinaria , Masculino , Proyectos de Investigación , Síndrome , Estados Unidos , Enfermedades Urológicas/fisiopatología
12.
Proc Natl Acad Sci U S A ; 111(41): 14710-5, 2014 Oct 14.
Artículo en Inglés | MEDLINE | ID: mdl-25267626

RESUMEN

Triple negative breast cancers (TNBCs) have a high mortality rate owing to aggressive proliferation and metastasis and a lack of effective therapeutic options. Herein, we describe the overexpression of intercellular adhesion molecule-1 (ICAM-1) in human TNBC cell lines and tissues, and demonstrate that ICAM-1 is a potential molecular target and biomarker for TNBC therapy and diagnosis. We synthesized ICAM-1 antibody-conjugated iron oxide nanoparticles (ICAM-IONPs) as a magnetic resonance imaging (MRI) probe to evaluate tumor targeting. Quantitative analysis of ICAM-1 surface expression predicted the targeting capability of ICAM-IONPs to TNBC cells. MRI of the TNBC xenograft tumor after systemic administration of ICAM-IONPs, coupled with iron quantification and histology, demonstrated a significant and sustained MRI contrast enhancement and probe accumulation in tumors with ICAM-1 overexpression relative to control. Identification of ICAM-1 as a TNBC target and biomarker may lead to the development of a new strategy and platform for addressing a critical gap in TNBC patient care.


Asunto(s)
Molécula 1 de Adhesión Intercelular/metabolismo , Terapia Molecular Dirigida , Neoplasias de la Mama Triple Negativas/metabolismo , Neoplasias de la Mama Triple Negativas/terapia , Animales , Biomarcadores de Tumor/metabolismo , Línea Celular Tumoral , Femenino , Compuestos Férricos/química , Humanos , Imagen por Resonancia Magnética , Ratones , Nanopartículas/química , Nanopartículas/ultraestructura , Ensayos Antitumor por Modelo de Xenoinjerto
13.
FASEB J ; 29(4): 1371-82, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25550468

RESUMEN

We have previously identified a zinc finger transcription factor, ZNF24 (zinc finger protein 24), as a novel inhibitor of tumor angiogenesis and have demonstrated that ZNF24 exerts this effect by repressing the transcription of VEGF in breast cancer cells. Here we focused on the role of ZNF24 in modulating the angiogenic potential of the endothelial compartment. Knockdown of ZNF24 by siRNA in human primary microvascular endothelial cells (ECs) led to significantly decreased cell migration and invasion compared with control siRNA. ZNF24 knockdown consistently led to significantly impaired VEGF receptor 2 (VEGFR2) signaling and decreased levels of matrix metalloproteinase-2 (MMP-2), with no effect on levels of major regulators of MMP-2 activity such as the tissue inhibitors of metalloproteinases and MMP-14. Moreover, silencing ZNF24 in these cells led to significantly decreased EC proliferation. Quantitative PCR array analyses identified multiple cell cycle regulators as potential ZNF24 downstream targets which may be responsible for the decreased proliferation in ECs. In vivo, knockdown of ZNF24 specifically in microvascular ECs led to significantly decreased formation of functional vascular networks. Taken together, these results demonstrate that ZNF24 plays an essential role in modulating the angiogenic potential of microvascular ECs by regulating the proliferation, migration, and invasion of these cells.


Asunto(s)
Células Endoteliales/citología , Células Endoteliales/metabolismo , Factores de Transcripción de Tipo Kruppel/metabolismo , Neovascularización Fisiológica , Movimiento Celular , Proliferación Celular , Células Cultivadas , Regulación del Desarrollo de la Expresión Génica , Técnicas de Silenciamiento del Gen , Humanos , Factores de Transcripción de Tipo Kruppel/antagonistas & inhibidores , Factores de Transcripción de Tipo Kruppel/genética , Metaloproteinasa 2 de la Matriz/genética , Metaloproteinasa 2 de la Matriz/metabolismo , Neovascularización Fisiológica/genética , ARN Mensajero/genética , ARN Mensajero/metabolismo , ARN Interferente Pequeño/genética , Transducción de Señal , Receptor 2 de Factores de Crecimiento Endotelial Vascular/metabolismo
14.
J Biol Chem ; 289(20): 14301-9, 2014 May 16.
Artículo en Inglés | MEDLINE | ID: mdl-24692560

RESUMEN

In the course of conducting a series of studies whose goal was to discover novel endogenous angiogenesis inhibitors, we have purified matrilin-1 (MATN-1) and have demonstrated, for the first time, that it inhibits neovascularization both in vitro and in vivo. Proteins were extracted from cartilage using a 2 m NaCl, 0.01 m HEPES buffer at 4 °C, followed by concentration of the extract. The concentrate was fractionated by size exclusion chromatography, and fractions were then screened for their ability to inhibit capillary endothelial cell (EC) proliferation in vitro. Fractions containing EC inhibitory activity were pooled and further purified by cation exchange chromatography. The resulting fractions from this step were then screened to isolate the antiangiogenic activity in vitro. This activity was identified by tandem mass spectrometry as being MATN-1. Human MATN-1 was cloned and expressed in Pichia pastoris and purified to homogeneity. Purified recombinant MATN-1, along with purified native protein, was shown to inhibit angiogenesis in vivo using the chick chorioallantoic membrane assay by the inhibition of capillary EC proliferation and migration. Finally, using a MATN-1-deficient mouse, we showed that angiogenesis during fracture healing was significantly higher in MATN-1(-/-) mice compared with the wild type mice as demonstrated by in vivo imaging and by elevated expression of angiogenesis markers including PECAM1, VEGFR, and VE-cadherin.


Asunto(s)
Inhibidores de la Angiogénesis/metabolismo , Proteínas Matrilinas/metabolismo , Neovascularización Fisiológica , Secuencia de Aminoácidos , Inhibidores de la Angiogénesis/química , Inhibidores de la Angiogénesis/genética , Inhibidores de la Angiogénesis/farmacología , Animales , Bovinos , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Pollos , Células Endoteliales/citología , Células Endoteliales/efectos de los fármacos , Fracturas Óseas/metabolismo , Fracturas Óseas/fisiopatología , Técnicas de Inactivación de Genes , Humanos , Masculino , Proteínas Matrilinas/química , Proteínas Matrilinas/genética , Proteínas Matrilinas/farmacología , Ratones , Datos de Secuencia Molecular , Neovascularización Fisiológica/efectos de los fármacos , Tibia/lesiones , Cicatrización de Heridas
15.
Biochem Biophys Res Commun ; 464(1): 126-32, 2015 Aug 14.
Artículo en Inglés | MEDLINE | ID: mdl-26086095

RESUMEN

Class 3 semaphorins were discovered as a family of axon guidance molecules, but are now known to be involved in diverse biologic processes. In this study, we investigated the anti-angiogenic potential of SEMA3E and SEMA3F (SEMA3E&F) in infantile hemangioma (IH). IH is a common vascular tumor that involves both vasculogenesis and angiogenesis. Our lab has identified and isolated hemangioma stem cells (HemSC), glucose transporter 1 positive (GLUT1(+)) endothelial cells (designated as GLUT1(sel) cells) based on anti-GLUT1 magnetic beads selection and GLUT1-negative endothelial cells (named HemEC). We have shown that these types of cells play important roles in hemangiogenesis. We report here that SEMA3E inhibited HemEC migration and proliferation while SEMA3F was able to suppress the migration and proliferation in all three types of cells. Confocal microscopy showed that stress fibers in HemEC were reduced by SEMA3E&F and that stress fibers in HemSC were decreased by SEMA3F, which led to cytoskeletal collapse and loss of cell motility in both cell types. Additionally, SEMA3E&F were able to inhibit vascular endothelial growth factor (VEGF)-induced sprouts in all three types of cells. Further, SEMA3E&F reduced the level of p-VEGFR2 and its downstream p-ERK in HemEC. These results demonstrate that SEMA3E&F inhibit IH cell proliferation and suppress the angiogenic activities of migration and sprout formation. SEMA3E&F may have therapeutic potential to treat or prevent growth of highly proliferative IH.


Asunto(s)
Células Endoteliales/efectos de los fármacos , Regulación Neoplásica de la Expresión Génica , Proteínas de la Membrana/farmacología , Células Madre Neoplásicas/efectos de los fármacos , Neovascularización Patológica/prevención & control , Proteínas del Tejido Nervioso/farmacología , Semaforinas/farmacología , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Células Endoteliales/metabolismo , Células Endoteliales/patología , Quinasas MAP Reguladas por Señal Extracelular/antagonistas & inhibidores , Quinasas MAP Reguladas por Señal Extracelular/genética , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Hemangioma/irrigación sanguínea , Hemangioma/genética , Hemangioma/patología , Humanos , Lactante , Neoplasias de Tejido Vascular/irrigación sanguínea , Neoplasias de Tejido Vascular/genética , Neoplasias de Tejido Vascular/patología , Células Madre Neoplásicas/metabolismo , Células Madre Neoplásicas/patología , Proteínas Recombinantes/farmacología , Transducción de Señal , Células Tumorales Cultivadas , Factor A de Crecimiento Endotelial Vascular/antagonistas & inhibidores , Factor A de Crecimiento Endotelial Vascular/genética , Factor A de Crecimiento Endotelial Vascular/metabolismo , Receptor 2 de Factores de Crecimiento Endotelial Vascular/antagonistas & inhibidores , Receptor 2 de Factores de Crecimiento Endotelial Vascular/genética , Receptor 2 de Factores de Crecimiento Endotelial Vascular/metabolismo
16.
Am J Pathol ; 184(7): 2099-110, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24952430

RESUMEN

Obesity and metabolic syndrome are linked to an increased prevalence of breast cancer among postmenopausal women. A common feature of obesity, metabolic syndrome, and a Western diet rich in saturated fat is a high level of circulating cholesterol. Epidemiological reports investigating the relationship between high circulating cholesterol levels, cholesterol-lowering drugs, and breast cancer are conflicting. Here, we modeled this complex condition in a well-controlled, preclinical animal model using innovative isocaloric diets. Female severe combined immunodeficient mice were fed a low-fat/no-cholesterol diet and then randomized to four isocaloric diet groups: low-fat/no-cholesterol diet, with or without ezetimibe (cholesterol-lowering drug), and high-fat/high-cholesterol diet, with or without ezetimibe. Mice were implanted orthotopically with MDA-MB-231 cells. Breast tumors from animals fed the high-fat/high-cholesterol diet exhibited the fastest progression. Significant differences in serum cholesterol level between groups were achieved and maintained throughout the study; however, no differences were observed in intratumoral cholesterol levels. To determine the mechanism of cholesterol-induced tumor progression, we analyzed tumor proliferation, apoptosis, and angiogenesis and found a significantly greater percentage of proliferating cells from mice fed the high-fat/high-cholesterol diet. Tumors from hypercholesterolemic animals displayed significantly less apoptosis compared with the other groups. Tumors from high-fat/high-cholesterol mice had significantly higher microvessel density compared with tumors from the other groups. These results demonstrate that hypercholesterolemia induces angiogenesis and accelerates breast tumor growth in vivo.


Asunto(s)
Hipercolesterolemia/patología , Neoplasias Mamarias Experimentales/patología , Neovascularización Patológica/patología , Animales , Apoptosis , Azetidinas/farmacología , Línea Celular Tumoral , Proliferación Celular , Colesterol/sangre , Colesterol en la Dieta/efectos adversos , Dieta Alta en Grasa/efectos adversos , Ezetimiba , Femenino , Humanos , Ratones SCID , Trasplante de Neoplasias
17.
Blood ; 121(4): 707-15, 2013 Jan 24.
Artículo en Inglés | MEDLINE | ID: mdl-23212515

RESUMEN

VEGF is a key regulator of normal and pathologic angiogenesis. Although many trans-activating factors of VEGF have been described, the transcriptional repression of VEGF remains much less understood. We have previously reported the identification of a SCAN domain-containing C2H2 zinc finger protein, ZNF24, that represses the transcription of VEGF. In the present study, we identify the mechanism by which ZNF24 represses VEGF transcription. Using reporter gene and electrophoretic mobility shift assays, we identify an 11-bp fragment of the proximal VEGF promoter as the ZNF24-binding site that is essential for ZNF24-mediated repression. We demonstrate in 2 in vivo models the potent inhibitory effect of ZNF24 on the vasculature. Expression of human ZNF24 induced in vivo vascular defects consistent with those induced by VEGF knockdown using a transgenic zebrafish model. These defects could be rescued by VEGF overexpression. Overexpression of ZNF24 in human breast cancer cells also inhibited tumor angiogenesis in an in vivo tumor model. Analyses of human breast cancer tissues showed that ZNF24 and VEGF levels were inversely correlated in malignant compared with normal tissues. These data demonstrate that ZNF24 represses VEGF transcription through direct binding to an 11-bp fragment of the VEGF proximal promoter and that it functions as a negative regulator of tumor growth by inhibiting angiogenesis.


Asunto(s)
Vasos Sanguíneos/metabolismo , Factores de Transcripción de Tipo Kruppel/metabolismo , Proteínas Represoras/metabolismo , Factor A de Crecimiento Endotelial Vascular/genética , Animales , Sitios de Unión , Neoplasias de la Mama/genética , Neoplasias de la Mama/metabolismo , Línea Celular Tumoral , Femenino , Regulación de la Expresión Génica , Humanos , Factores de Transcripción de Tipo Kruppel/genética , Neovascularización Patológica/genética , Neovascularización Patológica/metabolismo , Regiones Promotoras Genéticas , Unión Proteica , Proteínas Represoras/genética , Activación Transcripcional , Factor A de Crecimiento Endotelial Vascular/metabolismo , Pez Cebra
18.
BMC Cancer ; 15: 259, 2015 Apr 11.
Artículo en Inglés | MEDLINE | ID: mdl-25884438

RESUMEN

BACKGROUND: The objective of this study was to discover and to validate novel noninvasive biomarkers that distinguish between benign prostate hyperplasia (BPH) and localized prostate cancer (PCa), thereby helping to solve the diagnostic dilemma confronting clinicians who treat these patients. METHODS: Quantitative iTRAQ LC/LC/MS/MS analysis was used to identify proteins that are differentially expressed in the urine of men with BPH compared with those who have localized PCa. These proteins were validated in 173 urine samples from patients diagnosed with BPH (N = 83) and PCa (N = 90). Multivariate logistic regression analysis was used to identify the predictive biomarkers. RESULTS: Three proteins, ß2M, PGA3, and MUC3 were identified by iTRAQ and validated by immunoblot analyses. Univariate analysis demonstrated significant elevations in urinary ß2M (P < 0.001), PGA3 (P = 0.006), and MUC3 (P = 0.018) levels found in the urine of PCa patients. Multivariate logistic regression analysis revealed AUC values ranging from 0.618 for MUC3 (P = 0.009), 0.625 for PGA3 (P < 0.008), and 0.668 for ß2M (P < 0.001). The combination of all three demonstrated an AUC of 0.710 (95% CI: 0.631 - 0.788, P < 0.001); diagnostic accuracy improved even more when these data were combined with PSA categories (AUC = 0.812, (95% CI: 0.740 - 0.885, P < 0.001). CONCLUSIONS: Urinary ß2M, PGA3, and MUC3, when analyzed alone or when multiplexed with clinically defined categories of PSA, may be clinically useful in noninvasively resolving the dilemma of effectively discriminating between BPH and localized PCa.


Asunto(s)
Adenocarcinoma/diagnóstico , Biomarcadores de Tumor/orina , Hiperplasia Prostática/diagnóstico , Neoplasias de la Próstata/diagnóstico , Neoplasias de la Próstata/orina , Adenocarcinoma/orina , Anciano , Diagnóstico Diferencial , Humanos , Masculino , Persona de Mediana Edad , Hiperplasia Prostática/orina , Proteoma/metabolismo , Curva ROC
19.
FASEB J ; 27(1): 45-50, 2013 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-22982376

RESUMEN

Lipocalin 2 (Lcn2), a member of the lipocalin family, is up-regulated in a variety of epithelial cancers. We have previously reported that Lcn2 induces the epithelial to mesenchymal transition in breast cancer through the estrogen receptor α/Slug axis and that it is a potential noninvasive biomarker of this disease. Here, we report the novel finding that Lcn2 regulates breast cancer angiogenesis. Vascular endothelial growth factor (VEGF), a key angiogenic activator, was significantly increased with Lcn2 expression in MCF-7 human breast cancer cells as well as in an angiogenic line derived from MDA-MB-436 cells. Treatment with a VEGF-neutralizing antibody demonstrates that VEGF is essential for the angiogenic activity of Lcn2. We further demonstrate that Lcn2-induced VEGF is mediated through hypoxia-inducible factor 1α (HIF-1α) and that Lcn2 regulates HIF-1α through extracellular signal-regulated kinase (Erk). The regulation of HIF-1α and VEGF by Lcn2 was also demonstrated in the aggressive MDA-MB-231 cell line. Using the mouse corneal pocket assay, we found that Lcn2 significantly enhanced the angiogenesis induced by VEGF. Taken together, these results are the first to demonstrate that Lcn2 promotes angiogenesis in vitro and in vivo and suggest a novel mechanism through which Lcn2 may promote tumor progression.


Asunto(s)
Neoplasias de la Mama/irrigación sanguínea , Lipocalinas/fisiología , Neovascularización Patológica/fisiopatología , Secuencia de Bases , Línea Celular Tumoral , Cartilla de ADN , Femenino , Humanos , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Regulación hacia Arriba/fisiología , Factor A de Crecimiento Endotelial Vascular/metabolismo
20.
Mol Pharm ; 11(3): 755-65, 2014 Mar 03.
Artículo en Inglés | MEDLINE | ID: mdl-24467226

RESUMEN

Because breast cancer patient survival inversely correlates with metastasis, we engineered vehicles to inhibit both the C-X-C chemokine receptor type 4 (CXCR4) and lipocalin-2 (Lcn2) mediated migratory pathways. pH-responsive liposomes were designed to protect and trigger the release of Lcn2 siRNA. Liposomes were modified with anti-CXCR4 antibodies to target metastatic breast cancer (MBC) cells and block migration along the CXCR4-CXCL12 axis. This synergistic approach--coupling the CXCR4 axis blockade with Lcn2 silencing--significantly reduced migration in triple-negative human breast cancer cells (88% for MDA-MB-436 and 92% for MDA-MB-231). The results suggested that drug delivery vehicles engineered to attack multiple migratory pathways may effectively slow progression of MBC.


Asunto(s)
Proteínas de Fase Aguda/antagonistas & inhibidores , Anticuerpos Monoclonales/farmacología , Movimiento Celular , Regulación Neoplásica de la Expresión Génica , Lipocalinas/antagonistas & inhibidores , Proteínas Proto-Oncogénicas/antagonistas & inhibidores , ARN Interferente Pequeño/genética , Receptores CXCR4/antagonistas & inhibidores , Neoplasias de la Mama Triple Negativas/terapia , Proteínas de Fase Aguda/genética , Proteínas de Fase Aguda/metabolismo , Apoptosis , Western Blotting , Proliferación Celular , Terapia Combinada , Sistemas de Liberación de Medicamentos , Sinergismo Farmacológico , Femenino , Técnica del Anticuerpo Fluorescente , Humanos , Concentración de Iones de Hidrógeno , Lipocalina 2 , Lipocalinas/genética , Lipocalinas/metabolismo , Liposomas , Proteínas Proto-Oncogénicas/genética , Proteínas Proto-Oncogénicas/metabolismo , ARN Interferente Pequeño/administración & dosificación , Receptores CXCR4/inmunología , Receptores CXCR4/metabolismo , Transducción de Señal , Neoplasias de la Mama Triple Negativas/secundario , Células Tumorales Cultivadas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA