Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 39
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
J Physiol ; 589(Pt 5): 1061-80, 2011 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-21224239

RESUMO

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.


Assuntos
Estradiol/farmacologia , Estrogênios/farmacologia , Coração/efeitos dos fármacos , Miocárdio/metabolismo , Miócitos Cardíacos/metabolismo , Trocador de Sódio e Cálcio/metabolismo , Animais , Western Blotting , Cicloeximida/farmacologia , Dactinomicina/farmacologia , Eletrofisiologia , Estradiol/análogos & derivados , Antagonistas de Estrogênios/farmacologia , Feminino , Fulvestranto , Imuno-Histoquímica , Masculino , Miocárdio/citologia , Miócitos Cardíacos/citologia , Miócitos Cardíacos/efeitos dos fármacos , Fenetilaminas/farmacologia , Inibidores da Síntese de Proteínas/farmacologia , Coelhos , Sulfonamidas/farmacologia
2.
Int J Cancer ; 129(9): 2158-70, 2011 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-21170961

RESUMO

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.


Assuntos
Vacinas Anticâncer/imunologia , Indóis/farmacologia , Melanoma Experimental/imunologia , Pirróis/farmacologia , Animais , Anticorpos Bloqueadores/administração & dosagem , Anticorpos Bloqueadores/imunologia , Linhagem Celular Tumoral , Terapia Combinada , Células Dendríticas/imunologia , Epitopos de Linfócito T/imunologia , Feminino , Imunoterapia Adotiva , Indóis/administração & dosagem , Linfonodos/imunologia , Melanoma Experimental/terapia , Camundongos , Camundongos Endogâmicos C57BL , Pirróis/administração & dosagem , Receptores CXCR3/imunologia , Sunitinibe , Linfócitos T/imunologia , Microambiente Tumoral/imunologia , Molécula 1 de Adesão de Célula Vascular/imunologia
3.
Mol Ther ; 19(4): 805-14, 2011 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-21189473

RESUMO

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.


Assuntos
Terapia Genética/métodos , Interleucina-12/metabolismo , Linfócitos T Citotóxicos/imunologia , Linfócitos T Citotóxicos/metabolismo , Animais , Linfócitos T CD8-Positivos , Linhagem Celular Tumoral , Interleucina-12/genética , Melanoma Experimental/metabolismo , Camundongos , Reação em Cadeia da Polimerase Via Transcriptase Reversa
4.
Cancer Res ; 70(22): 9041-52, 2010 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-21045144

RESUMO

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.


Assuntos
Células Dendríticas/imunologia , Células Endoteliais/imunologia , Proteínas Recombinantes de Fusão/farmacologia , Subpopulações de Linfócitos T/imunologia , Microambiente Tumoral/efeitos dos fármacos , Animais , Linhagem Celular Tumoral , Células Dendríticas/metabolismo , Células Endoteliais/metabolismo , Feminino , Citometria de Fluxo , Fragmentos Fc das Imunoglobulinas/genética , Fragmentos Fc das Imunoglobulinas/metabolismo , Estimativa de Kaplan-Meier , Antígeno Ki-67/metabolismo , Camundongos , Camundongos da Linhagem 129 , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Microscopia Confocal , Molécula-1 de Adesão Celular Endotelial a Plaquetas/metabolismo , Receptores CCR7/metabolismo , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Sarcoma Experimental/tratamento farmacológico , Sarcoma Experimental/imunologia , Sarcoma Experimental/patologia , Subpopulações de Linfócitos T/metabolismo , Microambiente Tumoral/imunologia
5.
J Orthop Res ; 28(12): 1600-7, 2010 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-20973062

RESUMO

Intervertebral disc degeneration (IDD) is a common and debilitating disorder that results in reduced flexibility of the spine, pain, and reduced mobility. Risk factors for IDD include age, genetic predisposition, injury, and other environmental factors such as smoking. Loss of proteoglycans (PGs) contributes to IDD with advancing age. Currently there is a lack of a model for rapid investigation of disc aging and evaluation of therapeutic interventions. Here we examined progression of disc aging in a murine model of a human progeroid syndrome caused by deficiency of the DNA repair endonuclease, ERCC1-XPF (Ercc1(-/Δ) mice). The ERCC1-deficient mice showed loss of disc height and degenerative structural changes in their vertebral bodies similar to those reported for old rodents. Compared to their wild-type littermates, Ercc1(-/Δ) mice also exhibit other age-related IDD characteristics, including premature loss of disc PG, reduced matrix PG synthesis, and enhanced apoptosis and cell senescence. Finally, the onset of age-associated disc pathologies was further accelerated in Ercc1(-/Δ) mice following chronic treatment with the chemotherapeutic agent mechlorethamine. These results demonstrate that Ercc1(-/Δ) mice represent an accurate and rapid model of disc aging and provide novel evidence that DNA damage negatively impacts PG synthesis.


Assuntos
Envelhecimento/patologia , Proteínas de Ligação a DNA/deficiência , Endonucleases/deficiência , Degeneração do Disco Intervertebral/patologia , Disco Intervertebral/patologia , Progéria/patologia , Animais , Apoptose , Senescência Celular , Modelos Animais de Doenças , Degeneração do Disco Intervertebral/induzido quimicamente , Mecloretamina/toxicidade , Camundongos , Proteoglicanas/metabolismo
6.
Blood ; 116(4): 575-83, 2010 Jul 29.
Artigo em Inglês | MEDLINE | ID: mdl-20430958

RESUMO

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.


Assuntos
Infecções por Adenoviridae/imunologia , Comunicação Celular , Células Dendríticas/fisiologia , Interleucina-15/fisiologia , Células Matadoras Naturais/imunologia , Fator de Necrose Tumoral alfa/fisiologia , Animais , Comunicação Celular/imunologia , Diferenciação Celular/imunologia , Membrana Celular/metabolismo , Células Cultivadas , Células Dendríticas/imunologia , Células Dendríticas/metabolismo , Células Dendríticas/virologia , Feminino , Humanos , Imunidade Inata/imunologia , Interleucina-15/metabolismo , Células K562 , Células Matadoras Naturais/metabolismo , Células Matadoras Naturais/fisiologia , Ativação Linfocitária/imunologia , Camundongos , Camundongos Endogâmicos NOD , Camundongos Knockout , Camundongos SCID , Ligação Proteica/fisiologia , Fator de Necrose Tumoral alfa/metabolismo
7.
Heart Rhythm ; 2010 Dec 27.
Artigo em Inglês | MEDLINE | ID: mdl-21193060

RESUMO

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.

8.
PLoS One ; 4(10): e7470, 2009 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-19829715

RESUMO

Dendritic cells (DC) play a pivotal role in transmission and dissemination of HIV-1. Earlier studies reported that DC present at the site of infection trap virus particles via DC-SIGN and transfer the virus to the interacting naïve T cells. This prompted us to ask the question whether DC could acquire virus from infected T cells during DC-T cell interaction. To address this, we investigated the likely transfer of virus from HIV-1 infected T cells to DC and the underlying mechanisms involved. Results indicate that DC acquire virus from infected T cells via antigen uptake mechanism and this results in infection of DC with expression of proteins directed by viral DNA. Further studies with HIV-1 lacking the Env protein also resulted in infection of DC. The use of antibodies against DC-SIGN and DC-SIGN-R ruled out a role for receptor in the infection of DC. Additional data show that DC infection is directly correlated with the ability of DC to take up antigen from infected T cells. Overall, these studies provide evidence to suggest that HIV-1, besides infecting immune cells, also utilizes immunological mechanism(s) to acquire and disseminate virus.


Assuntos
Células Dendríticas/virologia , Infecções por HIV/metabolismo , Infecções por HIV/virologia , HIV-1/metabolismo , Linfócitos T/virologia , Linfócitos T CD4-Positivos/virologia , Moléculas de Adesão Celular/metabolismo , Proteínas de Fluorescência Verde/química , Humanos , Células Jurkat , Lectinas Tipo C/metabolismo , Leucócitos Mononucleares/citologia , Receptor de Manose , Lectinas de Ligação a Manose/metabolismo , Microscopia de Fluorescência/métodos , Fenótipo , Plasmídeos/metabolismo , Receptores de Superfície Celular/metabolismo , Linfócitos T/metabolismo
9.
Am J Respir Cell Mol Biol ; 41(1): 85-92, 2009 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-19097987

RESUMO

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.


Assuntos
Actinas/metabolismo , Monóxido de Carbono/farmacologia , Proteínas Ricas em Prolina do Estrato Córneo/metabolismo , Fibroblastos/efeitos dos fármacos , Pulmão/efeitos dos fármacos , Compostos Organometálicos/farmacologia , Fibrose Pulmonar/prevenção & controle , Actinas/genética , Administração por Inalação , Animais , Bleomicina , Desenvolvimento Ósseo/efeitos dos fármacos , Monóxido de Carbono/administração & dosagem , Morte Celular/efeitos dos fármacos , Movimento Celular/efeitos dos fármacos , Células Cultivadas , Proteínas Ricas em Prolina do Estrato Córneo/genética , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Fibroblastos/metabolismo , Perfilação da Expressão Gênica , Pulmão/metabolismo , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Desenvolvimento Muscular/efeitos dos fármacos , Fibrose Pulmonar/induzido quimicamente , Fibrose Pulmonar/genética , Fibrose Pulmonar/metabolismo , Fatores de Tempo , Fator de Crescimento Transformador beta1/metabolismo , Ubiquitinação/efeitos dos fármacos
10.
Cancer Res ; 68(19): 8076-84, 2008 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-18829566

RESUMO

Bone marrow-derived dendritic cells engineered using recombinant adenovirus to secrete high levels of IL-12p70 dramatically inhibited the growth of established CMS4 sarcomas in BALB/c mice after intratumoral administration. An analysis of splenic CD8(+) T cells in regressor mice revealed a strong, complex reactivity pattern against high-performance liquid chromatography (HPLC)-resolved peptides isolated by acid elution from single-cell suspensions of surgically resected CMS4 lesions. Mass spectrometry analyses defined two major overlapping peptide species that derive from the murine hemoglobin-beta (HBB) protein within the most stimulatory HPLC fractions. Although cultured CMS4 tumor cells failed to express HBB mRNA based on reverse transcription-PCR analyses, prophylactic vaccination of BALB/c mice with vaccines containing HBB peptides promoted specific CD8(+) T-cell responses that protected mice against a subsequent challenge with CMS4 or unrelated syngeneic (HBB(neg)) tumors of divergent histology (sarcoma, carcinomas of the breast or colon). In situ imaging suggested that vaccines limit or destabilize tumor-associated vascular structures, potentially by promoting immunity against HBB+ vascular pericytes. Importantly, there were no untoward effects of vaccination with the HBB peptide on peripheral RBC numbers, RBC hemoglobin content, or vascular structures in the brain or eye.


Assuntos
Linfócitos T CD8-Positivos/imunologia , Proliferação de Células , Hemoglobinas/imunologia , Imunidade Celular/fisiologia , Neoplasias/terapia , Animais , Linfócitos T CD8-Positivos/fisiologia , Vacinas Anticâncer/imunologia , Vacinas Anticâncer/uso terapêutico , Células Dendríticas/imunologia , Células Dendríticas/transplante , Feminino , Hemoglobinas/antagonistas & inibidores , Imunoterapia Adotiva/métodos , Camundongos , Camundongos Endogâmicos BALB C , Neoplasias/imunologia , Neoplasias/patologia , Fragmentos de Peptídeos/imunologia , Fragmentos de Peptídeos/uso terapêutico , Transplante Isogênico/imunologia , Carga Tumoral/imunologia , Células Tumorais Cultivadas
11.
Am J Pathol ; 173(2): 337-46, 2008 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-18583319

RESUMO

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.


Assuntos
Fibroblastos/metabolismo , Proteína 1 Inibidora de Diferenciação/fisiologia , Oncogenes/fisiologia , Fibrose Pulmonar/metabolismo , Transativadores/fisiologia , Fator de Crescimento Transformador beta/fisiologia , Animais , Bleomicina , Células Cultivadas , Proteínas de Ligação a DNA , Humanos , Proteína 1 Inibidora de Diferenciação/genética , Pulmão/metabolismo , Pulmão/patologia , Camundongos , Camundongos Knockout , Ligação Proteica , Fibrose Pulmonar/induzido quimicamente , Fibrose Pulmonar/patologia , Transativadores/biossíntese , Fatores de Transcrição , Fator de Crescimento Transformador beta/farmacologia
12.
Am J Respir Crit Care Med ; 177(11): 1223-32, 2008 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-18356564

RESUMO

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.


Assuntos
Antimetabólitos/uso terapêutico , Monóxido de Carbono/uso terapêutico , Respiração Artificial/efeitos adversos , Síndrome do Desconforto Respiratório/etiologia , Síndrome do Desconforto Respiratório/prevenção & controle , Resistência das Vias Respiratórias/fisiologia , Animais , Pressão Sanguínea/fisiologia , Modelos Animais de Doenças , Proteína 1 de Resposta de Crescimento Precoce/fisiologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Infiltração de Neutrófilos/fisiologia , PPAR gama/fisiologia , Síndrome do Desconforto Respiratório/metabolismo
13.
Autophagy ; 4(1): 88-90, 2008 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-17957135

RESUMO

Autophagy is a homeostatic process for recycling of proteins and organelles, that increases during times of nutrient deprivation and is regulated by reactive oxygen species. We reported that autophagy can also be induced after traumatic brain injury (TBI) in mice.1 Specifically, autophagosomes and multilamellar bodies were frequently observed in cell processes and axons in injured brain regions by electron microscopy, and lipidated microtubule-associated protein light chain 3 (LC3-II), was increased after TBI vs. controls. To determine if antioxidants could reduce autophagy, separate mice were treated with the antioxidant ?-glutamylcysteinyl ethyl ester (GCEE). Treatment with GCEE preserved total antioxidant reserves, reduced LC3-II in injured brains, and improved both behavioral and histological outcome after TBI. Here we report that LC3-II and autophagosomes were detectable in brain tissue from humans after TBI. Taken together, we show that autophagy occurs after both experimental and clinical TBI, and that oxidative stress contributes to overall neuropathology after TBI in mice, at least in part by initiating or influencing autophagy.


Assuntos
Autofagia/fisiologia , Lesões Encefálicas , Encéfalo/patologia , Estado Terminal , Animais , Antioxidantes/metabolismo , Proteínas Reguladoras de Apoptose/metabolismo , Proteína Beclina-1 , Lesões Encefálicas/patologia , Lesões Encefálicas/fisiopatologia , Dipeptídeos/metabolismo , Humanos , Proteínas de Membrana/metabolismo , Camundongos , Proteínas Associadas aos Microtúbulos/metabolismo , Neurônios/citologia , Neurônios/metabolismo , Estresse Oxidativo
14.
J Cereb Blood Flow Metab ; 27(2): 316-26, 2007 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-16736044

RESUMO

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.


Assuntos
Clorometilcetonas de Aminoácidos/farmacologia , Lesões Encefálicas/tratamento farmacológico , Lesões Encefálicas/patologia , Encéfalo/patologia , Citocromos c/metabolismo , Mitocôndrias/enzimologia , Fármacos Neuroprotetores , Animais , Western Blotting , Encéfalo/enzimologia , Lesões Encefálicas/enzimologia , Caspase 2/metabolismo , Caspase 3/metabolismo , Caspase 8/metabolismo , Sobrevivência Celular/efeitos dos fármacos , Marcação In Situ das Extremidades Cortadas , Masculino , Aprendizagem em Labirinto/fisiologia , Mitocôndrias/efeitos dos fármacos , Fator de Crescimento Neural/metabolismo , Proteínas do Tecido Nervoso/biossíntese , Proteínas do Tecido Nervoso/isolamento & purificação , Ratos , Ratos Sprague-Dawley
15.
J Neurotrauma ; 23(11): 1583-90, 2006 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-17115905

RESUMO

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.


Assuntos
Lesões Encefálicas/enzimologia , Caspase 7/metabolismo , Ativação Enzimática/fisiologia , Apoptose/fisiologia , Astrócitos/enzimologia , Astrócitos/metabolismo , Western Blotting , Lesões Encefálicas/patologia , Feminino , Humanos , Imuno-Histoquímica , Masculino , Pessoa de Meia-Idade , Neurônios/enzimologia , Neurônios/metabolismo , RNA Mensageiro/análise , Reação em Cadeia da Polimerase Via Transcriptase Reversa
16.
Cancer Res ; 66(9): 4895-903, 2006 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-16651446

RESUMO

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.


Assuntos
Linfócitos T CD8-Positivos/efeitos dos fármacos , Células Dendríticas/efeitos dos fármacos , Fator Estimulador de Colônias de Granulócitos e Macrófagos/farmacologia , Imunoterapia/métodos , Proteínas de Membrana/farmacologia , Neoplasias Experimentais/imunologia , Neoplasias Experimentais/terapia , Animais , Linfócitos T CD8-Positivos/imunologia , Linhagem Celular Tumoral , Reações Cruzadas , Citocinas/imunologia , Células Dendríticas/imunologia , Feminino , Fator Estimulador de Colônias de Granulócitos e Macrófagos/imunologia , Linfócitos do Interstício Tumoral/efeitos dos fármacos , Linfócitos do Interstício Tumoral/imunologia , Proteínas de Membrana/imunologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Baço/efeitos dos fármacos , Baço/imunologia
17.
Circ Res ; 98(2): 192-9, 2006 Feb 03.
Artigo em Inglês | MEDLINE | ID: mdl-16357303

RESUMO

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.


Assuntos
Proteínas de Ligação a DNA/fisiologia , Células Endoteliais/metabolismo , Endotelina-1/genética , Regulação da Expressão Gênica , Fatores de Transcrição/fisiologia , Animais , Células Cultivadas , Ácido Quenodesoxicólico/farmacologia , Regulação para Baixo , Células Endoteliais/química , Humanos , Regiões Promotoras Genéticas , Artéria Pulmonar/química , Coelhos , Ratos , Receptores Citoplasmáticos e Nucleares/genética , Elementos de Resposta , Fator de Transcrição AP-1/fisiologia , Ativação Transcricional
18.
Aesthet Surg J ; 26(4): 395-403, 2006.
Artigo em Inglês | MEDLINE | ID: mdl-19338921

RESUMO

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.

19.
J Exp Med ; 202(7): 941-53, 2005 Oct 03.
Artigo em Inglês | MEDLINE | ID: mdl-16203865

RESUMO

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.


Assuntos
Antígenos CD/imunologia , Diferenciação Celular/imunologia , Movimento Celular/imunologia , Imunoglobulinas/imunologia , Células Matadoras Naturais/citologia , Glicoproteínas de Membrana/imunologia , Receptores de Quimiocinas/imunologia , Linfócitos T Auxiliares-Indutores/citologia , Linhagem Celular , Quimiotaxia/imunologia , Citocinas/imunologia , Células Dendríticas/imunologia , Células Dendríticas/metabolismo , Citometria de Fluxo , Humanos , Interferon gama/metabolismo , Interleucinas/metabolismo , Células Matadoras Naturais/imunologia , Linfonodos/imunologia , Linfonodos/metabolismo , Receptores CCR7 , Linfócitos T Auxiliares-Indutores/imunologia , Antígeno CD83
20.
Mol Ther ; 12(3): 510-8, 2005 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-15953766

RESUMO

Pulmonary endothelium plays an important role in the maintenance of normal pulmonary physiology and its dysfunction is involved in a number of pulmonary diseases. Correction of endothelial dysfunction via antisense oligodeoxynucleotides (ODN) is dependent on the development of a delivery vehicle that can efficiently deliver the ODN to pulmonary endothelium with minimal toxicity. To this end, we have developed a novel lipidic vector that is highly efficient in targeted delivery of ODN to pulmonary endothelium. This is based on a method that utilizes an ionizable aminolipid (1,2-dioleoyl-3-dimethylammonium propane) and an ethanol-containing buffer system for encapsulating large quantities of polyanionic ODN in lipid vesicles. An endothelium-specific antibody (273-34A) is incorporated into the lipid vesicles via a distearoylphosphatidylethanolamine-poly(ethylene glycol) spacer. The 273-34A antibody efficiently mediated delivery of ODN to mouse lung endothelial cells in vitro and in vivo. Furthermore, systemic administration of this formulation is associated with minimal hematological toxicities and induces little acute change in systemic and pulmonary hemodynamics. These results provide a basis for lipid-mediated delivery of ODN for the treatment of pulmonary diseases. They also suggest the utility of this approach as a research tool to characterize the function of genes in the pulmonary endothelium.


Assuntos
Células Endoteliais/metabolismo , Técnicas de Transferência de Genes , Pulmão/citologia , Oligonucleotídeos/genética , Animais , Regulação para Baixo , Sistemas de Liberação de Medicamentos , Vetores Genéticos , Técnicas In Vitro , Molécula 1 de Adesão Intercelular/biossíntese , Metabolismo dos Lipídeos , Lipopolissacarídeos/química , Lipossomos/metabolismo , Pulmão/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Microscopia de Fluorescência , Oligonucleotídeos/química , Fatores de Tempo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...