Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 39
Filtrar
1.
Blood ; 116(4): 575-83, 2010 Jul 29.
Artículo en Inglés | MEDLINE | ID: mdl-20430958

RESUMEN

Recombinant adenovirus-engineered dendritic cells (Ad.DCs) are potent immunologic adjuvants of antiviral and anticancer vaccines. The effectiveness of Ad.DC-based vaccines may depend on the ability of Ad.DCs to crosstalk with natural killer (NK) cells and to activate, polarize, and bridge innate and adaptive immunity. We investigated, for the first time, whether and how human Ad.DCs activate NK cells, and compared the Ad.DC function with that of immature DCs and matured DCs (mDCs). We found that adenovirus transduction and lipopolysaccharide/interferon-gamma-induced maturation increased expression of transmembrane tumor necrosis factor (TNF) and trans-presented (trans) interleukin-15 (IL-15) on DCs, leading to enhanced NK cell activation without enhancing DC susceptibility to NK cell-mediated killing. This crosstalk enhanced NK cell CD69 expression, interferon-gamma secretion, proliferation, and antitumor activities, with Ad.DCs being significantly more effective than immature DCs, but less effective than mDCs. The Ad.DC and mDC crosstalk with NK cells was largely prevented by physical separation of DCs and NK cells, and neutralization of total TNF and IL-15, but not by selective sequestration of soluble TNF. These findings demonstrate that both Ad.DCs and mDCs can efficiently promote innate immune functions by activation of NK cells through the cooperative activities of tmTNF and trans-IL-15 mediated by cell-to-cell contact.


Asunto(s)
Infecciones por Adenoviridae/inmunología , Comunicación Celular , Células Dendríticas/fisiología , Interleucina-15/fisiología , Células Asesinas Naturales/inmunología , Factor de Necrosis Tumoral alfa/fisiología , Animales , Comunicación Celular/inmunología , Diferenciación Celular/inmunología , Membrana Celular/metabolismo , Células Cultivadas , Células Dendríticas/inmunología , Células Dendríticas/metabolismo , Células Dendríticas/virología , Femenino , Humanos , Inmunidad Innata/inmunología , Interleucina-15/metabolismo , Células K562 , Células Asesinas Naturales/metabolismo , Células Asesinas Naturales/fisiología , Activación de Linfocitos/inmunología , Ratones , Ratones Endogámicos NOD , Ratones Noqueados , Ratones SCID , Unión Proteica/fisiología , Factor de Necrosis Tumoral alfa/metabolismo
2.
Mol Ther ; 19(4): 805-14, 2011 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-21189473

RESUMEN

HLA-A2 transgenic mice bearing established HLA-A2(neg) B16 melanomas were effectively treated by intratumoral (i.t.) injection of syngeneic dendritic cells (DCs) transduced to express high levels of interleukin (IL)-12, resulting in CD8(+) T cell-dependent antitumor protection. In this model, HLA-A2-restricted CD8(+) T cells do not directly recognize tumor cells and therapeutic benefit was associated with the crosspriming of HLA-A2-restricted type-1 CD8(+) T cells reactive against antigens expressed by stromal cells [i.e., pericytes and vascular endothelial cells (VEC)]. IL-12 gene therapy-induced CD8(+) T cells directly recognized HLA-A2(+) pericytes and VEC flow-sorted from B16 tumor lesions based on interferon (IFN)-γ secretion and translocation of the lytic granule-associated molecule CD107 to the T cell surface after coculture with these target cells. In contrast, these CD8(+) T effector cells failed to recognize pericytes/VEC isolated from the kidneys of tumor-bearing HHD mice. The tumor-associated stromal antigen (TASA)-derived peptides studied are evolutionarily conserved and could be recognized by CD8(+) T cells harvested from the blood of HLA-A2(+) normal donors or melanoma patients after in vitro stimulation. These TASA and their derivative peptides may prove useful in vaccine formulations against solid cancers, as well as, in the immune monitoring of HLA-A2(+) cancer patients receiving therapeutic interventions, such as IL-12 gene therapy.


Asunto(s)
Terapia Genética/métodos , Interleucina-12/metabolismo , Linfocitos T Citotóxicos/inmunología , Linfocitos T Citotóxicos/metabolismo , Animales , Linfocitos T CD8-positivos , Línea Celular Tumoral , Interleucina-12/genética , Melanoma Experimental/metabolismo , Ratones , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
3.
J Physiol ; 589(Pt 5): 1061-80, 2011 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-21224239

RESUMEN

Female rabbit hearts are more susceptible to torsade de pointes (TdP) in acquired long QT type 2 than males, in-part due to higher L-type Ca2+ current (ICa,L) at the base of the heart. In principle, higher Ca2+ influx via ICa,L should be balanced by higher efflux, perhaps mediated by parallel sex differences of sodium-calcium exchange (NCX) current (INCX). We now show that NCX1, like Cav1.2α, is greater at the base of female than male left ventricular epicardium and greater at the base than at the apex in both sexes. In voltage-clamp studies, inward (0, +20 mV, P < 0.04) and outward (-80, -60, -40, -20 mV, P < 0.01) INCX densities were significantly higher (1.5-2 fold) in female base compared to apex and male (base and apex) myocytes. Myocytes were incubated ±17ß-oestradiol (E2 = 1 nm) and INCX was measured on days 0, 1, 2 and 3. Inward and outward INCX decreased over 2 days in female base myocytes becoming similar to INCX at the apex. E2 incubation (24 h) increased NCX1 (50%) and INCX (∼3-fold at 60 mV) in female base but not endocardium, apex or in male base myocytes. INCX upregulation by E2 was blunted by an oestrogen receptor (ER) antagonist (fulvestrant, 1 µm), and inhibition of transcription (actinomycin D, 5 µg ml-1) or translation (cycloheximide, 20 µg ml-1). Dofetilide (an IKr blocker) induced early afterdepolarizations (EADs) in female base myocytes cultured for 1 day if incubated with E2, but not without E2 or with E2+KB-R4973 (an INCX inhibitor), E2+fulvestrant or E2 with apex myocytes. Thus, E2 upregulates NCX1 by a genomic mechanism mediated by ERs, and de novo mRNA and protein biosynthesis, in a sex- and region-dependent manner which contributes to the enhanced propensity to EADs and TdP in female hearts.


Asunto(s)
Estradiol/farmacología , Estrógenos/farmacología , Corazón/efectos de los fármacos , Miocardio/metabolismo , Miocitos Cardíacos/metabolismo , Intercambiador de Sodio-Calcio/metabolismo , Animales , Western Blotting , Cicloheximida/farmacología , Dactinomicina/farmacología , Electrofisiología , Estradiol/análogos & derivados , Antagonistas de Estrógenos/farmacología , Femenino , Fulvestrant , Inmunohistoquímica , Masculino , Miocardio/citología , Miocitos Cardíacos/citología , Miocitos Cardíacos/efectos de los fármacos , Fenetilaminas/farmacología , Inhibidores de la Síntesis de la Proteína/farmacología , Conejos , Sulfonamidas/farmacología
4.
Int J Cancer ; 129(9): 2158-70, 2011 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-21170961

RESUMEN

The multikinase inhibitor sunitinib malate (SUT) has been reported to reduce levels of myeloid suppressor cells and Treg cells in cancer patients, hypothetically diminishing intrinsic impediments for active immunization against tumor-associated antigens in such individuals. The goal of this study was to identify longitudinal immune molecular and cellular changes associated with tumor regression and disease-free status after the treatment of established day 7 s.c. MO5 (B16.OVA) melanomas with SUT alone (1 mg/day via oral gavage for 7 days), vaccination using ovalbumin (OVA) peptide-pulsed dendritic cell [vaccine (VAC)] alone, or the combination of SUT and VAC (SUT/VAC). We observed superior anti-tumor efficacy for SUT/VAC combination approaches, particularly when SUT was applied at the time of the initial vaccination or the VAC boost. Treatment effectiveness was associated with the acute loss of (and/or failure to recruit) cells bearing myeloid-derived suppressor cells or Treg phenotypes within the tumor microenvironment (TME) and the corollary, prolonged enhancement of Type-1 anti-OVA CD8(+) T cell responses in the tumor-draining lymph node and the TME. Enhanced Type-1 T cell infiltration of tumors was associated with treatment-induced expression of vascular cell adhesion molecule-1 (VCAM-1) and CXCR3 ligand chemokines in vascular/peri-vascular cells within the TME, with SUT/VAC therapy benefits conditionally negated upon adminsitration of CXCR3 or VCAM-1 blocking antibodies. These data support the ability of a short 7 day course of SUT to (re)condition the TME to become more receptive to the recruitment and prolonged therapeutic action of (VAC-induced) anti-tumor Tc1 cells.


Asunto(s)
Vacunas contra el Cáncer/inmunología , Indoles/farmacología , Melanoma Experimental/inmunología , Pirroles/farmacología , Animales , Anticuerpos Bloqueadores/administración & dosificación , Anticuerpos Bloqueadores/inmunología , Línea Celular Tumoral , Terapia Combinada , Células Dendríticas/inmunología , Epítopos de Linfocito T/inmunología , Femenino , Inmunoterapia Adoptiva , Indoles/administración & dosificación , Ganglios Linfáticos/inmunología , Melanoma Experimental/terapia , Ratones , Ratones Endogámicos C57BL , Pirroles/administración & dosificación , Receptores CXCR3/inmunología , Sunitinib , Linfocitos T/inmunología , Microambiente Tumoral/inmunología , Molécula 1 de Adhesión Celular Vascular/inmunología
5.
J Exp Med ; 202(7): 941-53, 2005 Oct 03.
Artículo en Inglés | MEDLINE | ID: mdl-16203865

RESUMEN

In addition to their cytotoxic activities, natural killer (NK) cells can have immunoregulatory functions. We describe a distinct "helper" differentiation pathway of human CD56+CD3- NK cells into CD56+/CD83+/CCR7+/CD25+ cells that display high migratory responsiveness to lymph node (LN)-associated chemokines, high ability to produce interferon-gamma upon exposure to dendritic cell (DC)- or T helper (Th) cell-related signals, and pronounced abilities to promote interleukin (IL)-12p70 production in DCs and the development of Th1 responses. This helper pathway of NK cell differentiation, which is not associated with any enhancement of cytolytic activity, is induced by IL-18, but not other NK cell-activating factors. It is blocked by prostaglandin (PG)E2, a factor that induces a similar CD83+/CCR7+/CD25+ LN-homing phenotype in maturing DCs. The current data demonstrate independent regulation of the "helper" versus "effector" pathways of NK cell differentiation and novel mechanisms of immunoregulation by IL-18 and PGE2.


Asunto(s)
Antígenos CD/inmunología , Diferenciación Celular/inmunología , Movimiento Celular/inmunología , Inmunoglobulinas/inmunología , Células Asesinas Naturales/citología , Glicoproteínas de Membrana/inmunología , Receptores de Quimiocina/inmunología , Linfocitos T Colaboradores-Inductores/citología , Línea Celular , Quimiotaxis/inmunología , Citocinas/inmunología , Células Dendríticas/inmunología , Células Dendríticas/metabolismo , Citometría de Flujo , Humanos , Interferón gamma/metabolismo , Interleucinas/metabolismo , Células Asesinas Naturales/inmunología , Ganglios Linfáticos/inmunología , Ganglios Linfáticos/metabolismo , Receptores CCR7 , Linfocitos T Colaboradores-Inductores/inmunología , Antígeno CD83
6.
Am J Respir Cell Mol Biol ; 41(1): 85-92, 2009 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-19097987

RESUMEN

Carbon monoxide (CO) is a biologically active molecule produced in the body by the stress-inducible enzyme, heme oxygenase. We have previously shown that CO suppresses fibrosis in a murine bleomycin model. To investigate the mechanisms by which CO opposes fibrogenesis, we performed gene expression profiling of fibroblasts treated with transforming growth factor-beta(1) and CO. The most highly differentially expressed categories of genes included those related to muscular system development and the small proline-rich family of proteins. We confirmed in vitro, and in an in vivo bleomycin model of lung fibrosis, that CO suppresses alpha-smooth muscle actin expression and enhances small proline-rich protein-1a expression. We further show that these effects of CO depend upon signaling via the extracellular signal-regulated kinase pathway. Our results demonstrate novel transcriptional targets for CO and further elucidate the mechanism by which CO suppresses fibrosis.


Asunto(s)
Actinas/metabolismo , Monóxido de Carbono/farmacología , Proteínas Ricas en Prolina del Estrato Córneo/metabolismo , Fibroblastos/efectos de los fármacos , Pulmón/efectos de los fármacos , Compuestos Organometálicos/farmacología , Fibrosis Pulmonar/prevención & control , Actinas/genética , Administración por Inhalación , Animales , Bleomicina , Desarrollo Óseo/efectos de los fármacos , Monóxido de Carbono/administración & dosificación , Muerte Celular/efectos de los fármacos , Movimiento Celular/efectos de los fármacos , Células Cultivadas , Proteínas Ricas en Prolina del Estrato Córneo/genética , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Fibroblastos/metabolismo , Perfilación de la Expresión Génica , Pulmón/metabolismo , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Masculino , Ratones , Ratones Endogámicos C57BL , Desarrollo de Músculos/efectos de los fármacos , Fibrosis Pulmonar/inducido químicamente , Fibrosis Pulmonar/genética , Fibrosis Pulmonar/metabolismo , Factores de Tiempo , Factor de Crecimiento Transformador beta1/metabolismo , Ubiquitinación/efectos de los fármacos
7.
Am J Pathol ; 173(2): 337-46, 2008 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-18583319

RESUMEN

The presence of activated fibroblasts or myofibroblasts represents a hallmark of progressive lung fibrosis. Because the transcriptional response of fibroblasts to transforming growth factor-beta(1) (TGF-beta(1)) is a determinant of disease progression, we investigated the role of the transcriptional regulator inhibitor of differentiation-1 (Id1) in the setting of lung fibrosis. Mice lacking the gene for Id1 had increased susceptibility to bleomycin-induced lung fibrosis, and fibroblasts lacking Id1 exhibited enhanced responses to TGF-beta(1). Because the effect of Id1 on fibrosis could not be explained by known mechanisms, we performed protein interaction screening and identified a novel binding partner for Id1, known as dead ringer-like-1 (Dril1). Dril1 shares structural similarities with Id1 and was recently implicated in TGF-beta(1) signaling during embryogenesis. To date, little is known about the function of Dril1 in humans. Although it has not been previously implicated in fibrotic disease, we found that Dril1 was highly expressed in lungs from patients with idiopathic pulmonary fibrosis and was regulated by TGF-beta(1) in human fibroblasts. Dril1 enhanced activation of TGF-beta(1) target genes, whereas Id1 decreased expression of these same molecules. Id1 inhibited DNA binding by Dril1, and the two proteins co-localized in vitro and in vivo, providing a potential mechanism for suppression of fibrosis by Id1 through inhibition of the profibrotic function of Dril1.


Asunto(s)
Fibroblastos/metabolismo , Proteína 1 Inhibidora de la Diferenciación/fisiología , Oncogenes/fisiología , Fibrosis Pulmonar/metabolismo , Transactivadores/fisiología , Factor de Crecimiento Transformador beta/fisiología , Animales , Bleomicina , Células Cultivadas , Proteínas de Unión al ADN , Humanos , Proteína 1 Inhibidora de la Diferenciación/genética , Pulmón/metabolismo , Pulmón/patología , Ratones , Ratones Noqueados , Unión Proteica , Fibrosis Pulmonar/inducido químicamente , Fibrosis Pulmonar/patología , Transactivadores/biosíntesis , Factores de Transcripción , Factor de Crecimiento Transformador beta/farmacología
8.
Am J Respir Crit Care Med ; 177(11): 1223-32, 2008 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-18356564

RESUMEN

RATIONALE: Ventilator-induced lung injury (VILI) leads to an unacceptably high mortality. In this regard, the antiinflammatory properties of inhaled carbon monoxide (CO) may provide a therapeutic option. OBJECTIVES: This study explores the mechanisms of CO-dependent protection in a mouse model of VILI. METHODS: Mice were ventilated (12 ml/kg, 1-8 h) with air in the absence or presence of CO (250 ppm). Airway pressures, blood pressure, and blood gases were monitored. Lung tissue was analyzed for inflammation, injury, and gene expression. Bronchoalveolar lavage fluid was analyzed for protein, cell and neutrophil counts, and cytokines. MEASUREMENTS AND MAIN RESULTS: Mechanical ventilation caused significant lung injury reflected by increases in protein concentration, total cell and neutrophil counts in the bronchoalveolar lavage fluid, as well as the induction of heme oxygenase-1 and heat shock protein-70 in lung tissue. In contrast, CO application prevented lung injury during ventilation, inhibited stress-gene up-regulation, and decreased lung neutrophil infiltration. These effects were preceded by the inhibition of ventilation-induced cytokine and chemokine production. Furthermore, CO prevented the early ventilation-dependent up-regulation of early growth response-1 (Egr-1). Egr-1-deficient mice did not sustain lung injury after ventilation, relative to wild-type mice, suggesting that Egr-1 acts as a key proinflammatory regulator in VILI. Moreover, inhibition of peroxysome proliferator-activated receptor (PPAR)-gamma, an antiinflammatory nuclear regulator, by GW9662 abolished the protective effects of CO. CONCLUSIONS: Mechanical ventilation causes profound lung injury and inflammatory responses. CO treatment conferred protection in this model dependent on PPAR-gamma and inhibition of Egr-1.


Asunto(s)
Antimetabolitos/uso terapéutico , Monóxido de Carbono/uso terapéutico , Respiración Artificial/efectos adversos , Síndrome de Dificultad Respiratoria/etiología , Síndrome de Dificultad Respiratoria/prevención & control , Resistencia de las Vías Respiratorias/fisiología , Animales , Presión Sanguínea/fisiología , Modelos Animales de Enfermedad , Proteína 1 de la Respuesta de Crecimiento Precoz/fisiología , Masculino , Ratones , Ratones Endogámicos C57BL , Infiltración Neutrófila/fisiología , PPAR gamma/fisiología , Síndrome de Dificultad Respiratoria/metabolismo
9.
Circ Res ; 98(2): 192-9, 2006 Feb 03.
Artículo en Inglés | MEDLINE | ID: mdl-16357303

RESUMEN

The farnesoid X receptor (FXR) is a member of the nuclear receptor superfamily that is highly expressed in liver, kidney, adrenals, and intestine. FXR may play an important role in the pathogenesis of cardiovascular diseases via regulating the metabolism and transport of cholesterol. In this study, we report that FXR is also expressed in rat pulmonary artery endothelial cells (EC), a "nonclassical" bile acid target tissue. FXR is functional in EC, as demonstrated by induction of its target genes such as small heterodimer partner (SHP) after treatment with chenodeoxycholic acid, a FXR agonist. Interestingly, activation of FXR in EC led to downregulation of endothelin (ET)-1 expression. Reporter assays showed that activation of FXR inhibited transcriptional activation of the human ET-1 gene promoter and also repressed the activity of a heterologous promoter driven by activator protein (AP)-1 response elements. Electrophoretic mobility-shift and chromatin immunoprecipitation assays indicated that FXR reduced the binding activity of AP-1 transcriptional factors, suggesting that FXR may suppress ET-1 expression via negatively interfering with AP-1 signaling. These studies suggest that FXR may play a role in endothelial homeostasis and may serve as a novel molecular target for manipulating ET-1 expression in vascular EC.


Asunto(s)
Proteínas de Unión al ADN/fisiología , Células Endoteliales/metabolismo , Endotelina-1/genética , Regulación de la Expresión Génica , Factores de Transcripción/fisiología , Animales , Células Cultivadas , Ácido Quenodesoxicólico/farmacología , Regulación hacia Abajo , Células Endoteliales/química , Humanos , Regiones Promotoras Genéticas , Arteria Pulmonar/química , Conejos , Ratas , Receptores Citoplasmáticos y Nucleares/genética , Elementos de Respuesta , Factor de Transcripción AP-1/fisiología , Activación Transcripcional
10.
Cancer Res ; 66(9): 4895-903, 2006 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-16651446

RESUMEN

Dendritic cells play significant roles in the development and maintenance of antitumor immune responses. Therapeutic recruitment of dendritic cells into the tumor microenvironment has the potential to result in enhanced antitumor T-cell cross-priming against a broad array of naturally processed and presented tumor-associated antigens. We have observed that the treatment of BALB/c mice bearing syngeneic CMS4 sarcomas with the combination of recombinant Flt3 ligand and recombinant granulocyte-macrophage colony-stimulating factor (GM-CSF) for five sequential days is sufficient to optimize the number of tumor-infiltrating dendritic cells (TIDC). However, despite the significant increase in the number of TIDCs, the therapeutic benefit of Flt3 ligand and GM-CSF treatment is minimal. Therapy-associated TIDCs do not exhibit a "suppressed" or "suppressor" phenotype in vitro, and their enhanced numbers in cytokine-treated mice were associated with increased levels of peripheral antitumor CD8(+) T effector cells and with an augmented population of CD8(+) tumor-infiltrating lymphocytes (TIL). These data suggest that Flt3 ligand + GM-CSF therapy of murine tumors fails at a mechanistic point that is downstream of specific T-cell priming by therapy-induced TIDCs and the recruitment of these T cells into the tumor microenvironment. Based on the enhanced infiltration of tumors by CD4(+)CD25(+) TIL in Flt3 ligand + GM-CSF-treated mice, this could reflect the dominant influence of regulatory T cells in situ.


Asunto(s)
Linfocitos T CD8-positivos/efectos de los fármacos , Células Dendríticas/efectos de los fármacos , Factor Estimulante de Colonias de Granulocitos y Macrófagos/farmacología , Inmunoterapia/métodos , Proteínas de la Membrana/farmacología , Neoplasias Experimentales/inmunología , Neoplasias Experimentales/terapia , Animales , Linfocitos T CD8-positivos/inmunología , Línea Celular Tumoral , Reacciones Cruzadas , Citocinas/inmunología , Células Dendríticas/inmunología , Femenino , Factor Estimulante de Colonias de Granulocitos y Macrófagos/inmunología , Linfocitos Infiltrantes de Tumor/efectos de los fármacos , Linfocitos Infiltrantes de Tumor/inmunología , Proteínas de la Membrana/inmunología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Bazo/efectos de los fármacos , Bazo/inmunología
11.
J Cereb Blood Flow Metab ; 27(2): 316-26, 2007 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-16736044

RESUMEN

The pathobiology of traumatic brain injury (TBI) includes activation of multiple caspases followed by cell death with a spectrum of apoptotic phenotypes. There are initiator (e.g. caspase-2, -8, and -9) and effector (e.g. caspase-3 and -7) caspases. Recently, caspase-2 and -8 have been shown to regulate cell death via provoking cytochrome c release from the mitochondria upstream of caspase-9. Here, we show that an intracerebral injection of the pan-caspase inhibitor boc-Aspartyl(OMe)-fluoromethylketone (BAF; 1 micromol) 1 min after TBI in rats reduces caspase-3-like activity, terminal deoxynucleotidyl transferase-mediated dUTP nick-end labeling (TUNEL) and tissue damage, and cytochrome c release in ipsilateral cortex at 24 h versus vehicle. To investigate whether either caspase-2 and/or caspase-8 activation may contribute to cytochrome release, the effect of BAF treatment on caspase-2 and caspase-8 proteolysis was also examined. boc-aspartyl(OMe)-fluoromethylketone treatment inhibited proteolysis of caspase-2 but not caspase-8 24 h after TBI in rats versus vehicle. However, BAF with or without nerve growth factor (12.5 ng/h x 14 days intracerebrally via osmotic pump) did not result in differences in motor function, Morris water maze performance, hippocampal neuron survival, nor contusion volume at 14 days. These data suggest that BAF treatment reduces acute cell death after TBI by inhibiting mitochondrial release of cytochrome c, possibly via a mechanism involving initiator caspases; however, BAF appears to delay cell death, rather than result in permanent protection.


Asunto(s)
Clorometilcetonas de Aminoácidos/farmacología , Lesiones Encefálicas/tratamiento farmacológico , Lesiones Encefálicas/patología , Encéfalo/patología , Citocromos c/metabolismo , Mitocondrias/enzimología , Fármacos Neuroprotectores , Animales , Western Blotting , Encéfalo/enzimología , Lesiones Encefálicas/enzimología , Caspasa 2/metabolismo , Caspasa 3/metabolismo , Caspasa 8/metabolismo , Supervivencia Celular/efectos de los fármacos , Etiquetado Corte-Fin in Situ , Masculino , Aprendizaje por Laberinto/fisiología , Mitocondrias/efectos de los fármacos , Factor de Crecimiento Nervioso/metabolismo , Proteínas del Tejido Nervioso/biosíntesis , Proteínas del Tejido Nervioso/aislamiento & purificación , Ratas , Ratas Sprague-Dawley
12.
J Neurotrauma ; 23(11): 1583-90, 2006 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-17115905

RESUMEN

The expression and proteolysis of caspase family proteins are involved in the initiation and execution of apoptosis, which has been reported to occur in human and experimental traumatic brain injury (TBI). Caspase-3, -6, and -7 belong to the group of executioner caspases, which are cleaved and activated at the late, irreversible stage of apoptosis. Our previous studies demonstrated roles for caspase-1, -3, and -8 in humans after severe TBI. Here we report expression of caspase-7 mRNA and protein in humans after TBI (n = 16) and control brain-bank tissue (n = 6). Semiquantitative reverse transcription polymerase chain reaction showed no differences between caspase-7 mRNA in TBI patients versus controls (73 +/- 24 vs. 85 +/- 56 relative optical density [ROD], respectively). In contrast, Western blot analysis showed increased pro-caspase-7 in TBI patients versus controls (214 +/- 30 vs. 1 +/- 1 ROD, respectively), as well as an increase in the approximately 20 kD proteolytic fragment in TBI patients versus controls (86 +/- 13 vs. 22 +/- 12 ROD, respectively), consistent with activation of caspase-7 after TBI in humans. Immunohistochemical analysis showed that cells expressing caspase-7 included astrocytes and neurons and possibly other glial cell types and infiltrated inflammatory cells. These data show that caspase-7 and its cleavage product are increased in human brain after TBI in many central nervous system, as well as noncentral nervous system, cell types. Thus, caspase-7 may play a role in the glial and inflammatory responses, and possibly neuronal death, after TBI in humans.


Asunto(s)
Lesiones Encefálicas/enzimología , Caspasa 7/metabolismo , Activación Enzimática/fisiología , Apoptosis/fisiología , Astrocitos/enzimología , Astrocitos/metabolismo , Western Blotting , Lesiones Encefálicas/patología , Femenino , Humanos , Inmunohistoquímica , Masculino , Persona de Mediana Edad , Neuronas/enzimología , Neuronas/metabolismo , ARN Mensajero/análisis , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
13.
Aesthet Surg J ; 26(4): 395-403, 2006.
Artículo en Inglés | MEDLINE | ID: mdl-19338921

RESUMEN

BACKGROUND: Surgical repair of the superficial fascial system (SFS) has been claimed to both increase wound strength and enhance surgical outcome through anchoring of deeper tissues. OBJECTIVE: The authors assessed the biomechanical properties of the SFS to determine whether repair of the SFS layer improved early and long-term postoperative wound strength. METHODS: Four complementary studies were conducted to study the dermis and SFS junctional architecture and connective tissue content: gross dissection using a dehydrating agent (Pen-Fix; Richard-Allan Scientific, Kalamazoo, MI), a histologic study with hemotoxylin and eosin staining, soft tissue radiography, and immunofluorescence staining. Freshly excised human abdominal and lower back/buttock tissues underwent a midline incision, followed by repair using dermal sutures only (DRM), dermal sutures plus SFS sutures (DRM/SFS) or repair of the SFS only (SFS). Fresh swine abdominal tissues were similarly excised and repaired. Biomechanical tests were undertaken to compare the ex vivo human and swine tissues. Three types of closure-dermal sutures only (DRM), dermal sutures plus permanent 0-braided nylon suture in the SFS (DRM/SFS/N), and dermal sutures plus absorbable 0-vicryl suture in the SFS (DRM/SFS/V) were also tested in an in vivo swine model. RESULTS: Immunofluorescence studies showed collagen and elastin content and ratios to be comparable in the dermis and SFS. In ex vivo studies of human abdominal and back tissues, cyclic creep did not vary significantly among the different types of repair. DRM/SFS repair had a significantly higher failure load than dermal repair alone in both human abdominal and back tissues. In the in vivo swine study, normal tissue had a significantly higher failure load than all repair groups. The wounds where SFS had been repaired in addition to dermis exhibited an increased tensile strength and, among these, the wounds closed with SFS repair with a nonabsorbable suture exhibited greater tensile strength compared to absorbable suture repair. However, no statistically significant difference was noted, due to the small sample size. CONCLUSIONS: We have determined, using an ex vivo model, that repair of the SFS layer in addition to dermis repair significantly increases the initial biomechanical strength of wound repair. This has the potential to decrease early wound dehiscence. In our in vivo model, the use of a nonabsorbable suture to approximate the SFS demonstrated a trend toward increased long-term wound strength. We believe our studies provide scientific data documenting that SFS is a key contributory strength layer in the early postoperative period, and is likely to be a strength layer even in the later stages of wound healing.

14.
Cancer Res ; 64(4): 1386-95, 2004 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-14973054

RESUMEN

Nitric oxide is a potent radiosensitizer of tumors, but its use clinically is limited by serious side effects when administered systemically. We have demonstrated previously that gene transfer of the inducible nitric oxide synthase gene (iNOS) into colorectal cancer cells enhances radiation-induced apoptosis in vitro. The objectives of this study were to further characterize the effects of iNOS gene transfer on the radiosensitivity of human colorectal cancer cells in vitro and tumors grown in athymic nude mice. Adenoviral gene transfer of iNOS (AdiNOS) into human colorectal cancer cell lines (HCT-116 and SNU-1040 cells) significantly enhanced the effects of radiation with sensitizing enhancement ratios (0.1) of 1.65 and 1.6, respectively. The radiation enhancement induced by iNOS was associated with increased iNOS expression and nitric oxide production and prevented by L-NIO, an enzymatic inhibitor of iNOS. AdiNOS treatment of HCT-116 tumors combined with radiation (2 Gy x three fractions) led to a 3.4-fold greater (P < 0.005) tumor growth delay compared with radiation (RT) alone. AdiNOS plus RT also caused significant (P < 0.01) tumor regression with 63% of tumors regressing compared with only 6% of tumors treated with RT. AdiNOS plus RT significantly (P < or = 0.001) increased the percentage of apoptotic cells (22 +/- 4%) compared with either tumors treated with control vector plus RT (9 +/- 1%), AdiNOS alone (9 +/- 3%), or no treatment (2 +/- 1%). These radiosensitizing effects of AdiNOS occurred at low infection efficiency (4% of tumor infected), indicating a significant bystander effect.


Asunto(s)
Neoplasias Colorrectales/irrigación sanguínea , Neoplasias Colorrectales/radioterapia , Terapia Genética , Óxido Nítrico Sintasa/genética , Tolerancia a Radiación , Adenoviridae/genética , Animales , Apoptosis , Ciclo Celular , Neoplasias Colorrectales/patología , Transferencia de Gen Horizontal , Humanos , Ratones , Ratones Desnudos , Óxido Nítrico/fisiología , Óxido Nítrico Sintasa de Tipo II
15.
Cancer Res ; 64(21): 8015-21, 2004 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-15520210

RESUMEN

Nitric oxide (NO) is a potent tumor radiosensitizer; however, its clinical use is limited by systemic side effects. We have demonstrated previously that gene transfer of the human inducible NO synthase (iNOS) gene into tumor cells and tumors induces high-output NO production that significantly enhances tumor radioresponsiveness, with no observed side effects. Notably, iNOS gene transfer enhances tumor radioresponsiveness via apoptotic cell death. Because NO and ionizing radiation are both known to promote p53-dependent apoptosis, we hypothesized that p53 activation might be a primary mechanism for the synergy of these two genotoxic stresses. We report that NO and ionizing radiation synergistically activate p53 in colorectal cancers grown in athymic mice by augmenting phosphorylation of p53 at serine 15. The effect of NO and ionizing radiation on tumor cell apoptosis and tumor radioresponsiveness is significantly reduced in p53 knockout isogenic cancer cell lines. Furthermore, the transfer of both p53 and iNOS genes into tumor cells lacking functional p53 enhanced their radioresponsiveness more than transfer of either gene alone.


Asunto(s)
Apoptosis , Neoplasias del Colon/terapia , Terapia Genética , Óxido Nítrico Sintasa/genética , Proteína p53 Supresora de Tumor/fisiología , Adenoviridae/genética , Animales , Línea Celular Tumoral , Neoplasias del Colon/química , Neoplasias del Colon/patología , Terapia Combinada , Humanos , Ratones , Ratones Desnudos , Óxido Nítrico Sintasa de Tipo II , Tolerancia a Radiación , Proteína p53 Supresora de Tumor/análisis
16.
Cancer Res ; 63(19): 6378-86, 2003 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-14559827

RESUMEN

Dendritic cells (DCs) were adenovirally engineered to constitutively and durably secrete the potent Th1-biasing cytokines interleukin (IL)-12 (AdIL12DC) and/or IL-18 (AdIL18DC) and evaluated for their ability to promote therapeutic antitumor immunity in murine sarcoma models. Injection of either AdIL12DC or AdIL18DC into day 7 CMS4 or MethA tumors resulted in tumor rejection or slowed tumor growth when compared with control cohorts. Importantly, intratumoral injection with DCs engineered to secrete both IL-12 and IL-18 (AdIL12/IL18DC) resulted in complete and the most acute rejection of any treatment group analyzed. This strategy was also effective in promoting the regression of contralateral, untreated tumors. Both CD4+ and CD8+ T cells were required for tumor rejection. CD8+ splenic T cells from mice treated with AdIL12/IL18DC produced the highest levels of IFN-gamma in response to tumor rechallenge in vitro and displayed the broadest repertoire of Tc1-type reactivity to acid-eluted, tumor-derived peptides among all treatment cohorts. This apparent enhancement in cross-presentation of tumor-associated epitopes in vivo may result from the increased capacity of engineered DCs to kill tumor cells, survive tumor-induced apoptosis, and present immunogenic MHC/tumor peptide complexes to T cells after intratumoral injection. In support of this hypothesis, cytokine gene-engineered DCs expressed higher levels of MHC and costimulatory molecules, as well as Fas ligand and membrane-bound tumor necrosis factor alpha, with the latter markers associated with elevated tumoricidal activity in vitro. Cytokine gene-engineered DCs appeared to have a survival advantage in situ when injected into tumor lesions, to be found in approximation with regions of tumor apoptosis, and to have the capacity to ingest apoptotic tumor bodies. These results support the ability of combined cytokine gene transfer to enhance multiple effector functions mediated by intralesionally injected DCs that may concertedly promote cross-priming and the accelerated immune-mediated rejection of tumors.


Asunto(s)
Células Dendríticas/inmunología , Inmunoterapia Adoptiva/métodos , Interleucina-12/inmunología , Interleucina-18/inmunología , Sarcoma Experimental/terapia , Células TH1/inmunología , Adenoviridae/genética , Animales , Proteínas Reguladoras de la Apoptosis , Linfocitos T CD8-positivos/inmunología , Células Dendríticas/metabolismo , Células Dendríticas/fisiología , Células Dendríticas/virología , Epítopos de Linfocito T/inmunología , Femenino , Interleucina-12/biosíntesis , Interleucina-12/genética , Interleucina-12/metabolismo , Interleucina-18/biosíntesis , Interleucina-18/genética , Interleucina-18/metabolismo , Linfotoxina-alfa/biosíntesis , Glicoproteínas de Membrana/biosíntesis , Ratones , Ratones Endogámicos BALB C , Sarcoma Experimental/inmunología , Ligando Inductor de Apoptosis Relacionado con TNF , Células TH1/metabolismo , Transducción Genética , Factor de Necrosis Tumoral alfa/biosíntesis
17.
Cancer Res ; 64(22): 8381-8, 2004 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-15548708

RESUMEN

The overexpression of the inhibitor of apoptosis protein, survivin, may provide tumor cells with a distinct survival advantage in situ; hence, therapeutic strategies have been designed to inhibit its expression. In this study, we ectopically expressed the interferon regulatory factor (IRF)-1 protein in the breast carcinoma cell lines MDA-MB-468 and SK-BR-3 using a recombinant adenovirus (Ad-IRF-1). By screening microarray analysis of cDNA from the human breast cancer cell line MDA-MB-468 infected with Ad-IRF-1, we observed a 15-fold down-regulation of the survivin gene when compared with uninfected cells. Consequently, we tested survivin expression in Ad-IRF-1-infected MDA-MB-468 and SK-BR-3 breast cancer cell lines. Immunoblotting analyses supported the contention that ectopic expression of the IRF-1 protein results in down-regulation of survivin protein expression that is independent of p53. In addition, Ad-IRF-1 infection of these human breast cancer cell lines induces the expression of p21. We also report that increased apoptosis is observed in tumor cells infected with Ad-IRF-1 compared with Ad-Psi5 mock-infected cells and that cell death is further augmented when the IRF-1-infected cells are cultured with Adriamycin. Moreover, in a xenogeneic mouse model of breast carcinoma, in vivo treatment of tumor-bearing mice with intratumoral Ad-IRF-1 injections results in tumor growth inhibition. In resected tumors from mice that had been treated with Ad-IRF-1, tumor cells that express the IRF-1 transgene have a predominant IRF-1-positive, survivin-negative phenotype. Collectively, these observations suggest that therapies designed to enhance IRF-1 expression within tumor cells may represent novel treatment strategies for breast cancer.


Asunto(s)
Apoptosis , Neoplasias de la Mama/patología , Proteínas de Unión al ADN/metabolismo , Proteínas Asociadas a Microtúbulos/metabolismo , Fosfoproteínas/metabolismo , Animales , Neoplasias de la Mama/metabolismo , Línea Celular Tumoral , Doxorrubicina/farmacología , Humanos , Inmunohistoquímica , Proteínas Inhibidoras de la Apoptosis , Factor 1 Regulador del Interferón , Ratones , Proteínas de Neoplasias , Survivin
18.
Shock ; 19(2): 117-22, 2003 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-12578118

RESUMEN

It has been previously shown that the inducible nitric oxide (NO) synthase (iNOS; NOS-2) is elevated after hemorrhage, and that iNOS-derived NO participates in the upregulation of inflammation as well as lung and liver injury postresuscitation from shock. The purpose of this study was to elucidate the time course of iNOS mRNA expression, as well as the cellular and subcellular localization of iNOS protein in the liver posthemorrhage in rats subjected to varying durations of hemorrhagic shock (HS; mean arterial blood pressure [MAP] = 40 mmHg) with or without resuscitation. Expression of iNOS mRNA in rat liver by real-time reverse transcriptase (RT)-PCR demonstrated iNOS upregulation in shocked animals as compared with their sham counterparts as early as 60 min after the initiation of hemorrhage. By 1 h of HS, iNOS protein was detectable in rat liver by immunofluorescence, and this expression increased with time. Immunofluorescence localized iNOS primarily to the hepatocytes, and in particular to hepatocytes in the centrilobular regions. This analysis, confirmed by immunoelectron microscopy, revealed that iNOS colocalizes with catalase, a peroxisomal marker. Furthermore, we determined that iNOS mRNA is detectable by RT-PCR in liver biopsies from human subjects with HS (MAP < 90 mmHg) associated with trauma (n = 18). In contrast, none of the seven nontrauma surgical patients studied had detectable iNOS mRNA in their livers. Collectively, these results suggest that hepatic iNOS expression, associated with peroxisomal localization, is an early molecular response to HS in experimental animals and possibly in human patients with trauma with HS.


Asunto(s)
Hígado/enzimología , Óxido Nítrico Sintasa/biosíntesis , Choque Hemorrágico/metabolismo , Animales , Biopsia , Catalasa/metabolismo , Citosol/metabolismo , Hepatocitos/metabolismo , Humanos , Hígado/patología , Masculino , Microscopía Confocal , Microscopía Inmunoelectrónica , Óxido Nítrico Sintasa de Tipo II , Peroxisomas/metabolismo , ARN Mensajero/metabolismo , Ratas , Ratas Sprague-Dawley , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Factores de Tiempo , Regulación hacia Arriba
19.
Heart Rhythm ; 2010 Dec 27.
Artículo en Inglés | MEDLINE | ID: mdl-21193060

RESUMEN

This article has been withdrawn at the request of the author(s) and/or editor. The Publisher apologizes for any inconvenience this may cause. The full Elsevier Policy on Article Withdrawal can be found at http://www.elsevier.com/locate/withdrawalpolicy.

20.
Cancer Res ; 70(22): 9041-52, 2010 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-21045144

RESUMEN

Little preclinical modeling currently exists to support the use of OX40 agonists as therapeutic agents in the setting of advanced cancers, as well as the mechanisms through which therapeutic efficacy is achieved. We show that treatment of mice bearing well-established day 17 sarcomas with a novel OX40 ligand-Fc fusion protein (OX40L-Fc) resulted in tumor regression or dormancy in the majority of treated animals. Unexpectedly, dendritic cells (DC) in the progressive tumor microenvironment (TME) acquire OX40 expression and bind fluorescently labeled OX40L-Fc. Furthermore, longitudinal analyses revealed that DCs become enriched in the tumor-draining lymph node (TDLN) of both wild-type and Rag-/- mice within 3 days after OX40L-Fc treatment. By day 7 after treatment, a significant expansion of CXCR3+ T effector cells was noted in the TDLN, and by day 10 after treatment, type 1 polarized T cells exhibiting a reactivated memory phenotype had accumulated in the tumors. High levels of CXCL9 (a CXCR3 ligand) and enhanced expression of VCAM-1 by vascular endothelial cells (VEC) were observed in the TME early after treatment with OX40L-Fc. Notably, these vascular alterations were maintained in Rag-/- mice, indicating that the OX40L-Fc-mediated activation of both DC and VEC occurs in a T-cell-independent manner. Collectively, these findings support a paradigm in which the stimulation of DC, T cells, and the tumor vasculature by an OX40 agonist dynamically orchestrates the activation, expansion, and recruitment of therapeutic T cells into established tumors.


Asunto(s)
Células Dendríticas/inmunología , Células Endoteliales/inmunología , Proteínas Recombinantes de Fusión/farmacología , Subgrupos de Linfocitos T/inmunología , Microambiente Tumoral/efectos de los fármacos , Animales , Línea Celular Tumoral , Células Dendríticas/metabolismo , Células Endoteliales/metabolismo , Femenino , Citometría de Flujo , Fragmentos Fc de Inmunoglobulinas/genética , Fragmentos Fc de Inmunoglobulinas/metabolismo , Estimación de Kaplan-Meier , Antígeno Ki-67/metabolismo , Ratones , Ratones de la Cepa 129 , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Microscopía Confocal , Molécula-1 de Adhesión Celular Endotelial de Plaqueta/metabolismo , Receptores CCR7/metabolismo , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Sarcoma Experimental/tratamiento farmacológico , Sarcoma Experimental/inmunología , Sarcoma Experimental/patología , Subgrupos de Linfocitos T/metabolismo , Microambiente Tumoral/inmunología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA