Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 20 de 48
Filtrar
1.
J Neurophysiol ; 129(3): 591-608, 2023 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-36651913

RESUMEN

Detection of sounds is a fundamental function of the auditory system. Although studies of auditory cortex have gained substantial insight into detection performance using behaving animals, previous subcortical studies have mostly taken place under anesthesia, in passively listening animals, or have not measured performance at threshold. These limitations preclude direct comparisons between neuronal responses and behavior. To address this, we simultaneously measured auditory detection performance and single-unit activity in the inferior colliculus (IC) and cochlear nucleus (CN) in macaques. The spontaneous activity and response variability of CN neurons were higher than those observed for IC neurons. Signal detection theoretic methods revealed that the magnitude of responses of IC neurons provided more reliable estimates of psychometric threshold and slope compared with the responses of single CN neurons. However, pooling small populations of CN neurons provided reliable estimates of psychometric threshold and slope, suggesting sufficient information in CN population activity. Trial-by-trial correlations between spike count and behavioral response emerged 50-75 ms after sound onset for most IC neurons, but for few neurons in the CN. These results highlight hierarchical differences between neurometric-psychometric correlations in CN and IC and have important implications for how subcortical information could be decoded.NEW & NOTEWORTHY The cerebral cortex is widely recognized to play a role in sensory processing and decision-making. Accounts of the neural basis of auditory perception and its dysfunction are based on this idea. However, significantly less attention has been paid to midbrain and brainstem structures in this regard. Here, we find that subcortical auditory neurons represent stimulus information sufficient for detection and predict behavioral choice on a trial-by-trial basis.


Asunto(s)
Corteza Auditiva , Núcleo Coclear , Colículos Inferiores , Animales , Colículos Inferiores/fisiología , Percepción Auditiva/fisiología , Corteza Auditiva/fisiología , Núcleo Coclear/fisiología , Neuronas/fisiología , Estimulación Acústica , Vías Auditivas/fisiología
2.
Nat Med ; 8(8): 841-9, 2002 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-12091880

RESUMEN

The mechanism by which angiogenic factors recruit bone marrow (BM)-derived quiescent endothelial and hematopoietic stem cells (HSCs) is not known. Here, we report that functional vascular endothelial growth factor receptor-1 (VEGFR1) is expressed on human CD34(+) and mouse Lin(-)Sca-1(+)c-Kit(+) BM-repopulating stem cells, conveying signals for recruitment of HSCs and reconstitution of hematopoiesis. Inhibition of VEGFR1, but not VEGFR2, blocked HSC cell cycling, differentiation and hematopoietic recovery after BM suppression, resulting in the demise of the treated mice. Placental growth factor (PlGF), which signals through VEGFR1, restored early and late phases of hematopoiesis following BM suppression. PlGF enhanced early phases of BM recovery directly through rapid chemotaxis of VEGFR1(+) BM-repopulating and progenitor cells. The late phase of hematopoietic recovery was driven by PlGF-induced upregulation of matrix metalloproteinase-9, mediating the release of soluble Kit ligand. Thus, PlGF promotes recruitment of VEGFR1(+) HSCs from a quiescent to a proliferative BM microenvironment, favoring differentiation, mobilization and reconstitution of hematopoiesis.


Asunto(s)
Hematopoyesis/fisiología , Células Madre Hematopoyéticas/metabolismo , Proteínas Gestacionales/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Proteínas Tirosina Quinasas Receptoras/metabolismo , Animales , Antimetabolitos Antineoplásicos/farmacología , Separación Celular , Trasplante de Células , Quimiotaxis , Femenino , Fluorouracilo/farmacología , Células Madre Hematopoyéticas/efectos de los fármacos , Humanos , Masculino , Metaloproteinasa 9 de la Matriz/metabolismo , Ratones , Ratones Endogámicos , Factor de Crecimiento Placentario , Receptores de Factores de Crecimiento/metabolismo , Receptores de Factores de Crecimiento Endotelial Vascular , Quimera por Trasplante , Trasplante Heterólogo , Receptor 1 de Factores de Crecimiento Endotelial Vascular
3.
Nat Med ; 10(1): 64-71, 2004 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-14702636

RESUMEN

The molecular pathways involved in the differentiation of hematopoietic progenitors are unknown. Here we report that chemokine-mediated interactions of megakaryocyte progenitors with sinusoidal bone marrow endothelial cells (BMECs) promote thrombopoietin (TPO)-independent platelet production. Megakaryocyte-active cytokines, including interleukin-6 (IL-6) and IL-11, did not induce platelet production in thrombocytopenic, TPO-deficient (Thpo(-/-)) or TPO receptor-deficient (Mpl(-/-)) mice. In contrast, megakaryocyte-active chemokines, including stromal-derived factor-1 (SDF-1) and fibroblast growth factor-4 (FGF-4), restored thrombopoiesis in Thpo(-/-) and Mpl(-/-) mice. FGF-4 and SDF-1 enhanced vascular cell adhesion molecule-1 (VCAM-1)- and very late antigen-4 (VLA-4)-mediated localization of CXCR4(+) megakaryocyte progenitors to the vascular niche, promoting survival, maturation and platelet release. Disruption of the vascular niche or interference with megakaryocyte motility inhibited thrombopoiesis under physiological conditions and after myelosuppression. SDF-1 and FGF-4 diminished thrombocytopenia after myelosuppression. These data suggest that TPO supports progenitor cell expansion, whereas chemokine-mediated interaction of progenitors with the bone marrow vascular niche allows the progenitors to relocate to a microenvironment that is permissive and instructive for megakaryocyte maturation and thrombopoiesis. Progenitor-active chemokines offer a new strategy to restore hematopoiesis in a clinical setting.


Asunto(s)
Médula Ósea/irrigación sanguínea , Quimiocinas/fisiología , Células Madre Hematopoyéticas/citología , Trombopoyesis/fisiología , Animales , Antígenos CD , Cadherinas/fisiología , Adhesión Celular/fisiología , Movimiento Celular/fisiología , Megacariocitos/citología , Ratones , Proteínas Oncogénicas/genética , Proteínas Oncogénicas/fisiología , Receptores CXCR4/fisiología , Receptores de Citocinas/genética , Receptores de Citocinas/fisiología , Receptores de Trombopoyetina , Trombopoyetina/genética , Trombopoyetina/fisiología
4.
Nat Med ; 8(8): 831-40, 2002 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-12091877

RESUMEN

The therapeutic potential of placental growth factor (PlGF) and its receptor Flt1 in angiogenesis is poorly understood. Here, we report that PlGF stimulated angiogenesis and collateral growth in ischemic heart and limb with at least a comparable efficiency to vascular endothelial growth factor (VEGF). An antibody against Flt1 suppressed neovascularization in tumors and ischemic retina, and angiogenesis and inflammatory joint destruction in autoimmune arthritis. Anti-Flt1 also reduced atherosclerotic plaque growth and vulnerability, but the atheroprotective effect was not attributable to reduced plaque neovascularization. Inhibition of VEGF receptor Flk1 did not affect arthritis or atherosclerosis, indicating that inhibition of Flk1-driven angiogenesis alone was not sufficient to halt disease progression. The anti-inflammatory effects of anti-Flt1 were attributable to reduced mobilization of bone marrow-derived myeloid progenitors into the peripheral blood; impaired infiltration of Flt1-expressing leukocytes in inflamed tissues; and defective activation of myeloid cells. Thus, PlGF and Flt1 constitute potential candidates for therapeutic modulation of angiogenesis and inflammation.


Asunto(s)
Anticuerpos Monoclonales/uso terapéutico , Arteriosclerosis/tratamiento farmacológico , Artritis Experimental/tratamiento farmacológico , Neovascularización Patológica/tratamiento farmacológico , Neovascularización Fisiológica , Proteínas Gestacionales/farmacología , Proteínas Proto-Oncogénicas/inmunología , Proteínas Tirosina Quinasas Receptoras/inmunología , Animales , Anticuerpos Monoclonales/farmacología , Arteriosclerosis/inmunología , Arteriosclerosis/patología , Artritis Experimental/patología , Vasos Sanguíneos/citología , Vasos Sanguíneos/efectos de los fármacos , Vasos Sanguíneos/patología , Modelos Animales de Enfermedad , Factores de Crecimiento Endotelial/farmacología , Femenino , Células Madre Hematopoyéticas/efectos de los fármacos , Miembro Posterior/irrigación sanguínea , Humanos , Isquemia/tratamiento farmacológico , Isquemia/patología , Articulaciones/patología , Linfocinas/farmacología , Ratones , Ratones Desnudos , Isquemia Miocárdica/tratamiento farmacológico , Isquemia Miocárdica/patología , Isquemia Miocárdica/fisiopatología , Neovascularización Fisiológica/efectos de los fármacos , Factor de Crecimiento Placentario , Proteínas Gestacionales/genética , Proteínas Gestacionales/fisiología , Proteínas Proto-Oncogénicas/metabolismo , Ratas , Proteínas Tirosina Quinasas Receptoras/metabolismo , Receptores de Factores de Crecimiento/inmunología , Receptores de Factores de Crecimiento/metabolismo , Receptores de Factores de Crecimiento Endotelial Vascular , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Factor A de Crecimiento Endotelial Vascular , Receptor 1 de Factores de Crecimiento Endotelial Vascular , Factores de Crecimiento Endotelial Vascular
5.
J Exp Med ; 195(12): 1575-84, 2002 Jun 17.
Artículo en Inglés | MEDLINE | ID: mdl-12070285

RESUMEN

The vascular endothelial growth factor (VEGF) receptor fetal liver kinase 1 (flk1; VEGFR-2, KDR) is an endothelial cell-specific receptor tyrosine kinase that mediates physiological and pathological angiogenesis. We hypothesized that an active immunotherapy approach targeting flk1 may inhibit tumor angiogenesis and metastasis. To test this hypothesis, we first evaluated whether immune responses to flk1 could be elicited in mice by immunization with dendritic cells pulsed with a soluble flk1 protein (DC-flk1). This immunization generated flk1-specific neutralizing antibody and CD8+ cytotoxic T cell responses, breaking tolerance to self-flk1 antigen. Tumor-induced angiogenesis was suppressed in immunized mice as measured in an alginate bead assay. Development of pulmonary metastases was strongly inhibited in DC-flk1-immunized mice challenged with B16 melanoma or Lewis lung carcinoma cells. DC-flk1 immunization also significantly prolonged the survival of mice challenged with Lewis lung tumors. Thus, an active immunization strategy that targets an angiogenesis-related antigen on endothelium can inhibit angiogenesis and may be a useful approach for treating angiogenesis-related diseases.


Asunto(s)
Carcinoma Pulmonar de Lewis/irrigación sanguínea , Melanoma Experimental/irrigación sanguínea , Metástasis de la Neoplasia/prevención & control , Neovascularización Patológica/prevención & control , Proteínas Tirosina Quinasas Receptoras/inmunología , Receptores de Factores de Crecimiento/inmunología , Animales , Carcinoma Pulmonar de Lewis/patología , Femenino , Inmunoterapia , Melanoma Experimental/patología , Ratones , Ratones Endogámicos C57BL , Receptores de Factores de Crecimiento Endotelial Vascular
6.
Bioorg Med Chem ; 17(2): 731-40, 2009 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-19101155

RESUMEN

A series of arylphthalazine derivatives were synthesized and evaluated as antagonists of VEGF receptor II (VEGFR-2). IM-094482 57, which was prepared in two steps from commercially available starting materials, was found to be a potent inhibitor of VEGFR-2 in enzymatic, cellular and mitogenic assays (comparable activity to ZD-6474). Additionally, 57 inhibited the related receptor, VEGF receptor I (VEGFR-1), and showed excellent exposure when dosed orally to female CD-1 mice.


Asunto(s)
Ftalazinas/farmacocinética , Receptor 2 de Factores de Crecimiento Endotelial Vascular/antagonistas & inhibidores , Administración Oral , Animales , Disponibilidad Biológica , Femenino , Isoquinolinas/síntesis química , Isoquinolinas/farmacocinética , Ratones , Ratones Endogámicos , Ftalazinas/administración & dosificación , Ftalazinas/síntesis química , Piperidinas , Quinazolinas , Receptor 1 de Factores de Crecimiento Endotelial Vascular/antagonistas & inhibidores
7.
J Clin Invest ; 115(11): 2992-3006, 2005 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-16224539

RESUMEN

The molecular and cellular pathways that support the maintenance and stability of tumor neovessels are not well defined. The efficacy of microtubule-disrupting agents, such as combretastatin A4 phosphate (CA4P), in inducing rapid regression of specific subsets of tumor neovessels has opened up new avenues of research to identify factors that support tumor neoangiogenesis. Herein, we show that CA4P selectively targeted endothelial cells, but not smooth muscle cells, and induced regression of unstable nascent tumor neovessels by rapidly disrupting the molecular engagement of the endothelial cell-specific junctional molecule vascular endothelial-cadherin (VE-cadherin) in vitro and in vivo in mice. CA4P increases endothelial cell permeability, while inhibiting endothelial cell migration and capillary tube formation predominantly through disruption of VE-cadherin/beta-catenin/Akt signaling pathway, thereby leading to rapid vascular collapse and tumor necrosis. Remarkably, stabilization of VE-cadherin signaling in endothelial cells with adenovirus E4 gene or ensheathment with smooth muscle cells confers resistance to CA4P. CA4P synergizes with low and nontoxic doses of neutralizing mAbs to VE-cadherin by blocking assembly of neovessels, thereby inhibiting tumor growth. These data suggest that the microtubule-targeting agent CA4P selectively induces regression of unstable tumor neovessels, in part through disruption of VE-cadherin signaling. Combined treatment with anti-VE-cadherin agents in conjunction with microtubule-disrupting agents provides a novel synergistic strategy to selectively disrupt assembly and induce regression of nascent tumor neovessels, with minimal toxicity and without affecting normal stabilized vasculature.


Asunto(s)
Antineoplásicos Fitogénicos/farmacología , Cadherinas/fisiología , Endotelio Vascular/efectos de los fármacos , Melanoma Experimental/irrigación sanguínea , Melanoma Experimental/tratamiento farmacológico , Neovascularización Patológica/tratamiento farmacológico , Transducción de Señal/efectos de los fármacos , Estilbenos/farmacología , Animales , Capilares/crecimiento & desarrollo , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Técnicas de Cocultivo , Células Endoteliales/efectos de los fármacos , Endotelio Vascular/metabolismo , Endotelio Vascular/patología , Femenino , Humanos , Melanoma Experimental/metabolismo , Ratones , Ratones Endogámicos C57BL , Neovascularización Patológica/metabolismo , beta Catenina/fisiología
8.
J Clin Invest ; 112(5): 659-69, 2003 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-12952915

RESUMEN

Gonadotropins induce ovarian follicle growth that is coincident with increased follicular vasculature, suggesting a role of angiogenesis in follicle development. Functional studies performed in nonhuman primates show that administration of substances that inactivate VEGF block the development and function of preovulatory follicles as demonstrated by histological analysis or hormone measurements. Blockage of function of VEGF receptor 2 (VEGFR-2) alters follicular hormone secretion, suggesting that the intraovarian effect of VEGF might be mediated by this receptor. The specific mechanism by which follicular development was blocked in these previous studies remains unclear, however. Here we characterize the intraovarian role of VEGFR-2 activity on follicular development by choosing a model in which active feedback is absent, the prepuberally hypophysectomized mouse. Hypophysectomy prevents advanced follicle growth and maturation; however, follicle development to the preovulatory stage can be stimulated by administration of gonadotropins. We report that exogenously administered gonadotropins are unable to drive follicle development to the preovulatory stage in the presence of antiangiogenic agent, VEGFR-2-neutralizing Ab's. This inhibition of follicular development is caused by arrests to both angiogenesis and antrum formation. We conclude that the intraovarian VEGF/VEGFR-2 pathway is critical for gonadotropin-dependent angiogenesis and follicular development.


Asunto(s)
Gonadotropinas Equinas/farmacología , Neovascularización Fisiológica , Folículo Ovárico/irrigación sanguínea , Folículo Ovárico/crecimiento & desarrollo , Receptor 2 de Factores de Crecimiento Endotelial Vascular/fisiología , Animales , División Celular , Factores de Crecimiento Endotelial/fisiología , Estradiol/biosíntesis , Femenino , Hipofisectomía , Péptidos y Proteínas de Señalización Intercelular/fisiología , Linfocinas/fisiología , Ratones , Progesterona/biosíntesis , Ratas , Factor A de Crecimiento Endotelial Vascular , Factores de Crecimiento Endotelial Vascular
9.
Cancer Res ; 65(4): 1294-305, 2005 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-15735015

RESUMEN

Inhibition of vascular endothelial growth factor (VEGF) signaling, a key regulator of tumor angiogenesis, through blockade of VEGF receptor (VEGFR)-2 by the monoclonal antibody DC101 inhibits angiogenesis, tumor growth, and invasion. In a surface xenotransplant assay on nude mice using a high-grade malignant squamous cell carcinoma cell line (A-5RT3), we show that DC101 causes vessel regression and normalization as well as stromal maturation resulting in a reversion to a noninvasive tumor phenotype. Vessel regression is followed by down-regulation of expression of both VEGFR-2 and VEGFR-1 on endothelial cells and increased association of alpha-smooth muscle actin-positive cells with small vessels indicating their normalization, which was further supported by a regular ultrastructure. The phenotypic regression of an invasive carcinoma to a well-demarcated dysplastic squamous epithelium is accentuated by the establishment of a clearly structured epithelial basement membrane and the accumulation of collagen bundles in the stabilized connective tissue. This normalization of the tumor-stroma border coincided with down-regulated expression of the stromal matrix metalloproteinases 9 and 13, which supposedly resulted in attenuated turnover of extracellular matrix components permitting their structural organization. Thus, in this mouse model of a human squamous cell carcinoma cell line, blockade of VEGF signaling resulted in the reversion of the epithelial tumor phenotype through stromal normalization, further substantiating the crucial role of stromal microenvironment in regulating the tumor phenotype.


Asunto(s)
Carcinoma de Células Escamosas/irrigación sanguínea , Carcinoma de Células Escamosas/enzimología , Colagenasas/biosíntesis , Metaloproteinasa 9 de la Matriz/biosíntesis , Receptor 2 de Factores de Crecimiento Endotelial Vascular/antagonistas & inhibidores , Animales , Anticuerpos Monoclonales/farmacología , Carcinoma de Células Escamosas/patología , Carcinoma de Células Escamosas/terapia , Procesos de Crecimiento Celular/fisiología , Línea Celular Tumoral , Humanos , Metaloproteinasa 13 de la Matriz , Inhibidores de la Metaloproteinasa de la Matriz , Ratones , Ratones Desnudos , Trasplante de Neoplasias , Neovascularización Patológica/enzimología , Neovascularización Patológica/terapia , Fenotipo , Células del Estroma/enzimología , Células del Estroma/patología , Trasplante Heterólogo , Receptor 2 de Factores de Crecimiento Endotelial Vascular/inmunología
10.
Cancer Res ; 65(8): 3185-92, 2005 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-15833849

RESUMEN

Induction of neoangiogenesis plays an important role in the pathogenesis of multiple myeloma. However, the mechanism by which expression of vascular endothelial growth factor (VEGF)-A and its receptors modulate the interaction of multiple myeloma cells with stromal cells is not known. Here, we describe a novel in vitro coculture system using fetal bone stromal cells as a feeder layer, which facilitates the survival and growth of human primary multiple myeloma cells. We show that stromal-dependent paracrine VEGF-A signaling promotes proliferation of human primary multiple myeloma cells. Primary multiple myeloma cells only expressed functional VEGF receptor (VEGFR)-1, but not VEGFR-2 or VEGFR-3. VEGFR-1 expression was detected in the cytoplasm and the nuclei of proliferating multiple myeloma cells. Inhibition of VEGFR-1 abrogated multiple myeloma cell proliferation and motility, suggesting that the functional interaction of VEGF-A with its cognate receptor is essential for the growth of primary multiple myeloma cells. Collectively, our results suggest that stromal-dependent paracrine and intracrine VEGF-A/VEGFR-1 signaling contributes to human primary multiple myeloma cell growth and therefore, VEGFR-1 blockade is a potential therapeutic strategy for the treatment of multiple myeloma.


Asunto(s)
Movimiento Celular/fisiología , Mieloma Múltiple/patología , Factor A de Crecimiento Endotelial Vascular/fisiología , Receptor 1 de Factores de Crecimiento Endotelial Vascular/fisiología , Huesos/citología , Huesos/metabolismo , Procesos de Crecimiento Celular/fisiología , Núcleo Celular/metabolismo , Supervivencia Celular/fisiología , Técnicas de Cocultivo , Citoplasma/metabolismo , Humanos , Mieloma Múltiple/metabolismo , Células del Estroma/citología , Células del Estroma/metabolismo , Células Tumorales Cultivadas , Factor A de Crecimiento Endotelial Vascular/metabolismo , Receptor 1 de Factores de Crecimiento Endotelial Vascular/antagonistas & inhibidores , Receptor 1 de Factores de Crecimiento Endotelial Vascular/metabolismo
11.
IEEE Trans Neural Syst Rehabil Eng ; 25(8): 1202-1210, 2017 08.
Artículo en Inglés | MEDLINE | ID: mdl-27740490

RESUMEN

This study investigated the feasibility of predicting intrinsically caused trips (ICTs) in individuals with stroke. Gait kinematics collected from 12 individuals with chronic stroke, who demonstrated ICTs in treadmill walking, were analyzed. A prediction algorithm based on the outlier principle was employed. Sequential forward selection (SFS) and minimum-redundancy-maximum-relevance (mRMR) were used separately to identify the precursors for accurate ICT prediction. The results showed that it was feasible to predict ICTs around 50-260 ms before ICTs occurred in the swing phase by monitoring lower limb kinematics during the preceding stance phase. Both SFS and mRMR were effective in identifying the precursors of ICTs. For 9 out of the 12 subjects, the paretic lower limb's shank orientation in the sagittal plane and the vertical velocity of the paretic foot's center of gravity were important in predicting ICTs accurately; the averaged area under receiver operating characteristic curve achieved 0.95 and above. For the other three subjects, kinematics of the less affected limb or proximal joints in the paretic side were identified as the precursors to an ICT, potentially due to the variations of neuromotor deficits among stroke survivors. Although additional engineering efforts are still needed to address the challenges in making our design clinically practical, the outcome of this study may lead to further proactive engineering mechanisms for ICT avoidance and therefore reduce the risk of falls in individuals with stroke.


Asunto(s)
Accidentes por Caídas/prevención & control , Trastornos Neurológicos de la Marcha/diagnóstico , Trastornos Neurológicos de la Marcha/fisiopatología , Monitoreo Ambulatorio/métodos , Reconocimiento de Normas Patrones Automatizadas/métodos , Rehabilitación de Accidente Cerebrovascular/métodos , Adulto , Anciano , Anciano de 80 o más Años , Estudios de Factibilidad , Femenino , Marcha , Trastornos Neurológicos de la Marcha/etiología , Humanos , Masculino , Persona de Mediana Edad , Reproducibilidad de los Resultados , Sensibilidad y Especificidad , Accidente Cerebrovascular/complicaciones , Accidente Cerebrovascular/fisiopatología
12.
Hear Res ; 344: 1-12, 2017 02.
Artículo en Inglés | MEDLINE | ID: mdl-27770624

RESUMEN

Detection thresholds for auditory stimuli (signals) increase in the presence of maskers. Natural environments contain maskers/distractors that can have a wide range of spatiotemporal properties relative to the signal. While these parameters have been well explored psychophysically in humans, they have not been well explored in animal models, and their neuronal underpinnings are not well understood. As a precursor to the neuronal measurements, we report the effects of systematically varying the spatial and temporal relationship between signals and noise in macaque monkeys (Macaca mulatta and Macaca radiata). Macaques detected tones masked by noise in a Go/No-Go task in which the spatiotemporal relationships between the tone and noise were systematically varied. Masked thresholds were higher when the masker was continuous or gated on and off simultaneously with the signal, and lower when the continuous masker was turned off during the signal. A burst of noise caused higher masked thresholds if it completely temporally overlapped with the signal, whereas partial overlap resulted in lower thresholds. Noise durations needed to be at least 100 ms before significant masking could be observed. Thresholds for short duration tones were significantly higher when the onsets of signal and masker coincided compared to when the signal was presented during the steady state portion of the noise (overshoot). When signal and masker were separated in space, masked signal detection thresholds decreased relative to when the masker and signal were co-located (spatial release from masking). Masking release was larger for azimuthal separations than for elevation separations. These results in macaques are similar to those observed in humans, suggesting that the specific spatiotemporal relationship between signal and masker determine threshold in natural environments for macaques in a manner similar to humans. These results form the basis for future investigations of neuronal correlates and mechanisms of masking.


Asunto(s)
Conducta Animal , Señales (Psicología) , Ruido/efectos adversos , Enmascaramiento Perceptual , Percepción de la Altura Tonal , Detección de Señal Psicológica , Localización de Sonidos , Estimulación Acústica , Animales , Audiometría , Vías Auditivas/fisiología , Umbral Auditivo , Macaca mulatta , Macaca radiata , Masculino , Modelos Animales , Periodicidad , Psicoacústica , Factores de Tiempo
13.
Clin Cancer Res ; 11(13): 4934-40, 2005 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-16000592

RESUMEN

PURPOSE: Inhibition of angiogenesis can influence tumor cell invasion and metastasis. We previously showed that blockade of vascular endothelial growth factor receptor-2 (VEGFR-2) with the monoclonal antibody DC101 inhibited intracerebral glioblastoma growth but caused increased tumor cell invasion along the preexistent vasculature. In the present study, we attempted to inhibit glioma cell invasion using a monoclonal antibody against the epidermal growth factor receptor (EGFR), which in the context of human glioblastomas, has been implicated in tumor cell invasion. In addition, we analyzed whether blockade of vascular endothelial (VE)-cadherin as a different antiangiogenic target could also inhibit glioblastoma angiogenesis and growth. EXPERIMENTAL DESIGNS: Nude mice who received intracerebral glioblastoma xenografts were treated using monoclonal antibodies against VEGFR-2 (DC101), EGFR (C225), and VE-cadherin (E4G10) either alone or in different combinations. RESULTS: Increased tumor cell invasion provoked by DC101 monotherapy was inhibited by 50% to 66% by combined treatment with C225 and DC101. C225 inhibited glioblastoma cell migration in vitro, but had no effect on the volume of the main tumor mass or on tumor cell proliferation or apoptosis in vivo, either alone or in combination with DC101. The anti-VE-cadherin monoclonal antibody E4G10 was a weaker inhibitor of tumor angiogenesis and growth than DC101, and also caused a weaker increase in tumor cell invasion. CONCLUSIONS: Inhibition of angiogenesis achieved by blocking either VEGFR-2 or VE-cadherin can cause increased glioma cell invasion in an orthotopic model. Increased tumor cell invasion induced by potent inhibition of angiogenesis with DC101 could be inhibited by simultaneous blockade of EGFR.


Asunto(s)
Anticuerpos Monoclonales/farmacología , Glioblastoma/tratamiento farmacológico , Neovascularización Patológica/prevención & control , Animales , Anticuerpos Monoclonales/administración & dosificación , Anticuerpos Monoclonales/uso terapéutico , Antígenos CD , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Apoptosis/efectos de los fármacos , Cadherinas/inmunología , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Endotelio Vascular/metabolismo , Receptores ErbB/inmunología , Femenino , Glioblastoma/irrigación sanguínea , Glioblastoma/patología , Humanos , Ratones , Ratones SCID , Invasividad Neoplásica , Neovascularización Patológica/patología , Factor de Crecimiento Transformador alfa/farmacología , Receptor 2 de Factores de Crecimiento Endotelial Vascular/inmunología , Ensayos Antitumor por Modelo de Xenoinjerto/métodos
14.
Cancer Res ; 63(23): 8264-70, 2003 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-14678984

RESUMEN

Intermittent bolus-contrast power Doppler ultrasound was used for noninvasive, quantitative monitoring of tumor perfusion during antiangiogenic therapy. Subcutaneous heterotransplants of human squamous cell carcinoma cells in nude mice were treated with a blocking antibody to vascular endothelial growth factor receptor 2 (DC101) and repeatedly examined at weekly intervals. Using replenishment kinetics of microbubbles (Levovist) tumor vascularization, including capillary blood flow, was clearly visualized by this dynamic ultrasound method allowing the determination of a comprehensive functional status of tumor vascularization (blood volume, blood flow, perfusion, and mean blood velocity) in all examined tumors. DC101 treatment decreased tumor blood flow (-64%) and volume (-73%) compared with untreated controls (+409% and +185%, respectively). Regression of functional vessel parameters was observed early well before reduction of tumor size. The treatment-related amount of reduction in tumor volume was directly correlated for the initial tumor blood flow before start of therapy and the perfusion calculated at the preceding examination. The vessel density (immunofluorescence staining with CD31 antibody at different time points) showed an excellent correlation with the calculated relative blood volume (k = 0.84, P < 0.01), thereby validating intermittent sonography as a useful monitoring method. We conclude that intermittent sonography is a promising tool for comprehensive monitoring of antiangiogenic or proangiogenic therapies, especially during early stages of treatment, thus yielding information regarding a prospective evaluation of therapy effects beyond the follow up of tumor size.


Asunto(s)
Carcinoma de Células Escamosas/irrigación sanguínea , Carcinoma de Células Escamosas/terapia , Neovascularización Patológica/diagnóstico por imagen , Neovascularización Patológica/terapia , Ultrasonografía Doppler/métodos , Animales , Anticuerpos Monoclonales/administración & dosificación , Carcinoma de Células Escamosas/diagnóstico por imagen , Quimioterapia del Cáncer por Perfusión Regional/métodos , Humanos , Ratones , Ratones Desnudos , Receptor 2 de Factores de Crecimiento Endotelial Vascular/antagonistas & inhibidores , Receptor 2 de Factores de Crecimiento Endotelial Vascular/inmunología , Ensayos Antitumor por Modelo de Xenoinjerto
15.
Cancer Res ; 62(10): 2731-5, 2002 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-12019144

RESUMEN

A number of recent preclinical studies have sparked interest in the concept of exploiting conventional chemotherapeutic drugs as antiangiogenics. Such antiangiogenic activity is achieved or optimized by metronomic-dosing protocols in which the drug is given at comparatively low doses using a frequent schedule of administration (e.g., once to three times per week) with no breaks, particularly when combined with an endothelial cell-specific antiangiogenic drug. The use of p.o. chemotherapeutic drugs is particularly suitable for this type of treatment strategy. We tested one such drug, cyclophosphamide (CTX), in a protocol wherein the drug was administered to mice at low doses, of approximately 10-40 mg/kg on a daily basis through the drinking water. CTX is typically given p.o. to patients, but it has almost always been injected when treating preclinical mouse tumor models. We found p.o. CTX to be a safe and convenient treatment with significant antitumor efficacy. Growth delays were observed for human orthotopic breast or ectopic colon cancer xenografts in nude or SCID mice. Established PC3 human prostate tumor xenografts could be induced to almost fully regress, remaining virtually nonpalpable for > or =2 months of continuous therapy, after which tumors began to grow progressively. These re-emergent tumors were not found to be drug resistant when tested in new hosts, using the same treatment protocol. Regression of spontaneously arising, late-stage pancreatic islet cell carcinomas in Rip Tag transgenic mice was also observed. The effects of continuous p.o. CTX treatment were enhanced significantly in an orthotopic, metastatic breast cancer xenograft model when used in combination with an antivascular endothelial growth factor receptor-2 blocking antibody. Maximum tolerated dose levels established for other mouse strains proved highly toxic to SCID mice, whereas daily p.o. low-dose regimens of CTX were well tolerated. Taken together, the results demonstrate the feasibility of delivering CTX in a p.o. metronomic chemotherapy regimen, which proved safe, reasonably efficacious, and potentially applicable to chronic treatment. Such a regimen may be particularly well suited for integration with antiangiogenic drugs.


Asunto(s)
Inhibidores de la Angiogénesis/administración & dosificación , Antineoplásicos Alquilantes/administración & dosificación , Neoplasias de la Mama/tratamiento farmacológico , Ciclofosfamida/administración & dosificación , Administración Oral , Animales , Anticuerpos/administración & dosificación , Neoplasias de la Mama/irrigación sanguínea , Neoplasias de la Mama/patología , División Celular/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Ingestión de Líquidos , Femenino , Células HT29/efectos de los fármacos , Humanos , Masculino , Ratones , Ratones Desnudos , Ratones SCID , Neoplasias de la Próstata/irrigación sanguínea , Neoplasias de la Próstata/tratamiento farmacológico , Neoplasias de la Próstata/patología , Proteínas Tirosina Quinasas Receptoras/inmunología , Receptores de Factores de Crecimiento/inmunología , Receptores de Factores de Crecimiento Endotelial Vascular , Células Tumorales Cultivadas , Ensayos Antitumor por Modelo de Xenoinjerto
16.
Cancer Res ; 62(9): 2567-75, 2002 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-11980651

RESUMEN

Vascular endothelial-cadherin (VE-cadherin) is an endothelial cell-specific adhesion molecule that is localized exclusively at cell-cell contacts referred to as adherens junctions. VE-cadherin-mediated adhesion is crucial for proper assembly of vascular structures during angiogenesis as well as for maintenance of a normal vascular integrity. We have shown previously that a monoclonal antibody (BV13) to VE-cadherin not only inhibits the formation of vascular tubes during tumor angiogenesis but also disrupts adherens junctions of normal vasculature with a concomitant increase in vascular permeability. The goal of the current studies was to block VE-cadherin function during angiogenesis without disrupting existing junctions on normal endothelium. Using in vitro screening assays to test for functional blocking of adherens junction formation and in vivo assays to detect antibody effects on vascular permeability in normal tissues, we have identified a novel blocking antibody (E4G10) that inhibits VE-cadherin function during angiogenesis but does not disrupt existing adherens junctions on normal vasculature. E4G10 inhibited formation of vascular tubes in vivo in the Matrigel plug and corneal micropocket assays. E4G10 also inhibited tumor growth in three models of mouse and human tumors via an antiangiogenic mechanism. Examination of normal mouse and tumor tissues showed that E4G10 bound to endothelial cells in a subset of tumor vasculature but not to normal vasculature. Bromodeoxyuridine labeling experiments showed that E4G10 specifically targeted a subset of tumor endothelium that is undergoing active cell proliferation, which likely reflects the activated, angiogenic endothelium. These findings indicate that VE-cadherin can be selectively targeted during states of pathological angiogenesis, despite its ubiquitous distribution throughout the entire vasculature. Our data also suggest that antibody E4G10 recognizes VE-cadherin epitopes that are only accessible on endothelial cells forming new adherens junctions, such as in angiogenic tumor vasculature.


Asunto(s)
Inhibidores de la Angiogénesis/farmacología , Anticuerpos Monoclonales/farmacología , Cadherinas/inmunología , Endotelio Vascular/inmunología , Neoplasias Cutáneas/irrigación sanguínea , Neoplasias Cutáneas/tratamiento farmacológico , Uniones Adherentes/efectos de los fármacos , Uniones Adherentes/inmunología , Inhibidores de la Angiogénesis/inmunología , Inhibidores de la Angiogénesis/metabolismo , Animales , Anticuerpos Monoclonales/inmunología , Anticuerpos Monoclonales/metabolismo , Antígenos CD , Permeabilidad Capilar/inmunología , División Celular/inmunología , Córnea/efectos de los fármacos , Endotelio Vascular/citología , Endotelio Vascular/efectos de los fármacos , Endotelio Vascular/metabolismo , Inhibidores de Crecimiento/inmunología , Inhibidores de Crecimiento/farmacología , Humanos , Ratones , Ratones Desnudos , Neovascularización Patológica/tratamiento farmacológico , Neovascularización Fisiológica/efectos de los fármacos , Neoplasias Cutáneas/inmunología , Ensayos Antitumor por Modelo de Xenoinjerto
17.
Cancer Res ; 63(24): 8912-21, 2003 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-14695208

RESUMEN

The insulin-like growth factor I receptor (IGF-IR) is overexpressed in many diverse tumor types and is a critical signaling molecule for tumor cell proliferation and survival. Therapeutic strategies targeting the IGF-IR may therefore be effective broad-spectrum anticancer agents. Through screening of a Fab phage display library, we have generated a fully human antibody (A12) that binds to the IGF-IR with high affinity (4.11 x 10(-11) M) and inhibits ligand binding with an IC(50) of 0.6-1 nM. Antibody-mediated blockade of ligand binding to the IGF-IR inhibited downstream signaling of the two major insulin-like growth factor (IGF) pathways, mitogen-activated protein kinase and phosphatidylinositol 3'-kinase/Akt, in MCF7 human breast cancer cells. As a result, the mitogenic and proliferative potential of IGF-I and IGF-II were significantly reduced. A12 did not block insulin binding to the insulin receptor but could block binding to atypical IGF-IR in MCF7 cells. In addition, A12 was shown to induce IGF-IR internalization and degradation on specific binding to tumor cells, resulting in a significant reduction in cell surface receptor density. In xenograft tumor models in vivo, IGF-IR blockade by A12 was shown to occur rapidly, resulting in significant growth inhibition of breast, renal, and pancreatic tumors. Histological analysis of tumor sections demonstrated a marked increase in apoptotic tumor cells in antibody-treated animals. These results demonstrate that A12 possesses strong antitumor activity in vitro and in vivo and may therefore be an effective therapeutic candidate for the treatment of cancers that are dependent on IGF-IR signaling for growth and survival.


Asunto(s)
Anticuerpos Monoclonales/farmacología , Receptor IGF Tipo 1/antagonistas & inhibidores , Animales , Especificidad de Anticuerpos , Neoplasias de la Mama/terapia , División Celular/efectos de los fármacos , Femenino , Humanos , Factor I del Crecimiento Similar a la Insulina/antagonistas & inhibidores , Factor I del Crecimiento Similar a la Insulina/metabolismo , Ligandos , Ratones , Ratones Desnudos , Biblioteca de Péptidos , Fosforilación , Receptor IGF Tipo 1/inmunología , Receptor IGF Tipo 1/metabolismo , Proteínas Recombinantes/farmacología , Transducción de Señal/efectos de los fármacos , Ensayos Antitumor por Modelo de Xenoinjerto
18.
Mol Cancer Ther ; 4(3): 369-79, 2005 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-15767546

RESUMEN

Platelet-derived growth factor receptor alpha (PDGFRalpha) is a type III receptor tyrosine kinase that is expressed on a variety of tumor types. A neutralizing monoclonal antibody to human PDGFRalpha, which did not cross-react with the beta form of the receptor, was generated. The fully human antibody, termed 3G3, has a Kd of 40 pmol/L and blocks both PDGF-AA and PDGF-BB ligands from binding to PDGFRalpha. In addition to blocking ligand-induced cell mitogenesis and receptor autophosphorylation, 3G3 inhibited phosphorylation of the downstream signaling molecules Akt and mitogen-activated protein kinase. This inhibition was seen in both transfected and tumor cell lines expressing PDGFRalpha. The in vivo antitumor activity of 3G3 was tested in human glioblastoma (U118) and leiomyosarcoma (SKLMS-1) xenograft tumor models in athymic nude mice. Antibody 3G3 significantly inhibited the growth of U118 (P=0.0004) and SKLMS-1 (P <0.0001) tumors relative to control. These data suggest that 3G3 may be useful for the treatment of tumors that express PDGFRalpha.


Asunto(s)
Anticuerpos Monoclonales/química , Receptor alfa de Factor de Crecimiento Derivado de Plaquetas/metabolismo , Animales , Becaplermina , Bioensayo , Línea Celular Tumoral , Relación Dosis-Respuesta Inmunológica , Citometría de Flujo , Humanos , Cinética , Ligandos , Sistema de Señalización de MAP Quinasas , Ratones , Ratones Desnudos , Ratones Transgénicos , Trasplante de Neoplasias , Fosforilación , Factor de Crecimiento Derivado de Plaquetas/química , Unión Proteica , Proteínas Serina-Treonina Quinasas/metabolismo , Estructura Terciaria de Proteína , Proteínas Proto-Oncogénicas/metabolismo , Proteínas Proto-Oncogénicas c-akt , Proteínas Proto-Oncogénicas c-sis , Receptor alfa de Factor de Crecimiento Derivado de Plaquetas/inmunología , Factores de Tiempo , Transfección
19.
Annu Int Conf IEEE Eng Med Biol Soc ; 2016: 5038-5041, 2016 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-28269400

RESUMEN

Above knee amputees exhibit a higher risk of falling than able-bodied people, so the capacity to recover from trips (a major cause of unintentional falls) is critical for these amputees to prevent fall-related injuries. Although trip recovery approaches using powered prostheses have been proposed, the effectiveness of these approaches has not been evaluated with varied trip-related disturbance levels. Here, we conducted a simulation study to understand the relationship between trip-related disturbance levels and environmental factors. This knowledge could clarify the design space as well as guide design and evaluation techniques of future trip recovery approaches.


Asunto(s)
Accidentes por Caídas/prevención & control , Simulación por Computador , Ambiente , Humanos
20.
Oncogene ; 22(56): 9097-106, 2003 Dec 08.
Artículo en Inglés | MEDLINE | ID: mdl-14663488

RESUMEN

The potential for disease-specific targeting and low toxicity profiles have made monoclonal antibodies attractive therapeutic drug candidates. Antibody-mediated target cell killing is frequently associated with immune effector mechanisms such as antibody-directed cellular cytotoxicity, but they can also be induced by apoptotic processes. Antibody-directed mechanisms, including antigen crosslinking, activation of death receptors, and blockade of ligand-receptor growth or survival pathways, can elicit the induction of apoptosis in targeted cells. Depending on their mechanism of action, monoclonal antibodies can induce targeted cell-specific killing alone or can enhance target cell susceptibility to chemo- or radiotherapeutics by effecting the modulation of antiapoptotic pathways. This review will focus on the mechanisms by which antibodies are capable of eliciting programmed cell death either directly or indirectly within tumor cells.


Asunto(s)
Anticuerpos Monoclonales/uso terapéutico , Antineoplásicos/uso terapéutico , Apoptosis/efectos de los fármacos , Animales , Anticuerpos Monoclonales/farmacología , Antineoplásicos/farmacología , División Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Humanos , Inmunotoxinas/uso terapéutico , Modelos Biológicos , Transducción de Señal
SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda