Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 7 de 7
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Oncotarget ; 7(11): 12718-30, 2016 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-26863637

RESUMO

Malignant gliomas have poor prognosis and urgently require new therapies. Activating Transcription Factor 5 (ATF5) is highly expressed in gliomas, and interference with its expression/function precipitates targeted glioma cell apoptosis in vitro and in vivo. We designed a novel deliverable truncated-dominant-negative (d/n) form of ATF5 fused to a cell-penetrating domain (Pen-d/n-ATF5-RP) that can be intraperitoneally/subcutaneously administered to mice harboring malignant gliomas generated; (1) by PDGF-B/sh-p53 retroviral transformation of endogenous neural progenitor cells; and (2) by human U87-MG xenografts. In vitro Pen-d/n-ATF5-RP entered into glioma cells and triggered massive apoptosis. In vivo, subcutaneously-administered Pen-d/n-ATF5-RP passed the blood brain barrier, entered normal brain and tumor cells, and then caused rapid selective tumor cell death. MRI verified elimination of retrovirus-induced gliomas within 8-21 days. Histopathology revealed growth-suppression of intracerebral human U87-MG cells xenografts. For endogenous PDGF-B gliomas, there was no recurrence or mortality at 6-12 months versus 66% mortality in controls at 6 months. Necropsy and liver-kidney blood enzyme analysis revealed no adverse effects on brain or other tissues. Our findings thus identify Pen-d/n-ATF5-RP as a potential therapy for malignant gliomas.


Assuntos
Fatores Ativadores da Transcrição/antagonistas & inibidores , Antineoplásicos/farmacologia , Neoplasias Encefálicas , Desenho de Fármacos , Glioma , Fatores Ativadores da Transcrição/farmacologia , Animais , Proteínas de Transporte/farmacologia , Linhagem Celular Tumoral , Peptídeos Penetradores de Células , Humanos , Camundongos , Peptídeos/farmacologia , Ensaios Antitumorais Modelo de Xenoenxerto
2.
PLoS One ; 10(12): e0145523, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26709509

RESUMO

Studies have suggested a link between the transforming growth factor beta 1 (TGF-ß1) signaling cascade and the stress-inducible activating transcription factor 3 (ATF3). We have demonstrated that triglyceride-rich lipoproteins (TGRL) lipolysis products activate MAP kinase stress associated JNK/c-Jun pathways resulting in up-regulation of ATF3, pro-inflammatory genes and induction of apoptosis in human aortic endothelial cells. Here we demonstrate increased release of active TGF-ß at 15 min, phosphorylation of Smad2 and translocation of co-Smad4 from cytosol to nucleus after a 1.5 h treatment with lipolysis products. Activation and translocation of Smad2 and 4 was blocked by addition of SB431542 (10 µM), a specific inhibitor of TGF-ß-activin receptor ALKs 4, 5, 7. Both ALK receptor inhibition and anti TGF-ß1 antibody prevented lipolysis product induced up-regulation of ATF3 mRNA and protein. ALK inhibition prevented lipolysis product-induced nuclear accumulation of ATF3. ALKs 4, 5, 7 inhibition also prevented phosphorylation of c-Jun and TGRL lipolysis product-induced p53 and caspase-3 protein expression. These findings demonstrate that TGRL lipolysis products cause release of active TGF-ß and lipolysis product-induced apoptosis is dependent on TGF-ß signaling. Furthermore, signaling through the stress associated JNK/c-Jun pathway is dependent on TGF-ß signaling suggesting that TGF-ß signaling is necessary for nuclear accumulation of the ATF3/cJun transcription complex and induction of pro-inflammatory responses.


Assuntos
Fator 3 Ativador da Transcrição/biossíntese , Células Endoteliais/metabolismo , Lipoproteínas/metabolismo , Receptores de Fatores de Crescimento Transformadores beta/metabolismo , Triglicerídeos/metabolismo , Transporte Ativo do Núcleo Celular , Animais , Aorta/metabolismo , Apoptose , Caspase 3/metabolismo , Células Cultivadas , Humanos , Lipólise , Lipoproteínas/genética , Camundongos , Fosforilação , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Transdução de Sinais , Proteína Smad2/metabolismo , Proteína Smad4/metabolismo , Estresse Fisiológico , Fator de Crescimento Transformador beta1/metabolismo , Triglicerídeos/genética , Proteína Supressora de Tumor p53/metabolismo
3.
Cell Med ; 4(1): 1-11, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-23152950

RESUMO

Mesenchymal stem cells (MSCs) derived from bone marrow (BM), adipose tissue (AT), umbilical cord blood (CB), and umbilical cord tissue (CT) are increasingly being used to treat equine inflammatory and degenerative lesions. MSCs modulate the immune system in part through mediator secretion. Animal species and MSC tissue of origin are both important determinants of MSC function. In spite of widespread clinical use, how equine MSCs function to heal tissues is fully unknown. In this study, MSCs derived from BM, AT, CB, and CT were compared for their ability to inhibit lymphocyte proliferation and secrete mediators in response to activation. Five MSC lines from each tissue were isolated. Lymphocyte proliferation was assessed in a mixed leukocyte reaction, and mediator secretion was determined by ELISA. Regardless of tissue of origin, quiescent MSCs did not alter lymphocyte proliferation or secrete mediators, except for transforming growth factor-ß (TGF-ß1). When stimulated, MSCs of all tissue types decreased lymphocyte proliferation, increased prostaglandin (PGE(2)) and interleukin-6 (IL-6) secretion, and decreased production of tumor necrosis factor-α (TNF-α) and interferon-γ (IFN-γ). BM-MSCs and CB-MSCs also produced nitric oxide (NO), while AT-MSCs and CT-MSCs did not. Equine MSCs did not produce indoleamine 2,3-dioxygenase (IDO). These data suggest that activated equine MSCs derived from BM, AT, CT, and CB secrete high concentration of mediators and are similar to MSCs from rodents and humans in their immunomodulatory profiles. These findings have implication for the treatment of inflammatory lesions dominated by activated lymphocytes and TNF-α and IFN-γ in vivo.

4.
Physiol Genomics ; 43(15): 917-29, 2011 Aug 16.
Artigo em Inglês | MEDLINE | ID: mdl-21652769

RESUMO

Epidemiologic studies associate exposure to ambient particulate matter (APM) with increased cardiovascular mortality. Since both pulmonary inflammation and systemic circulation of ultrafine particles are hypothesized as initiating cardiovascular effects, we examined responses of potential target cells in vitro. Human aortic endothelial cells (HAEC) were exposed to 10 µg/ml fine and ultrafine APM collected in an urban setting in summer 2006 or winter 2007 in the San Joaquin Valley, California. RNA isolated after 3 h was analyzed with high-density oligonucleotide arrays. Summer APM treatment affected genes involved in xenobiotic and oxidoreductase activity, transcription factors, and inflammatory responses in HAEC, while winter APM had a robust xenobiotic but lesser inflammatory response. Real-time polymerase chain reaction analysis confirmed that particulate matter (PM)-treated HAEC increased mRNA levels of xenobiotic response enzymes CYP1A1, ALDH1A3, and TIPARP and cellular stress response transcription factor ATF3. Inflammatory response genes included E-selectin, PTGS2, CXCL-2 (MIP-2α), and CCL-2 (MCP-1). Multiplex protein assays showed secretion of IL-6 and MCP-1 by HAEC. Since induction of CYP1A1 is mediated through the ligand-activated aryl hydrocarbon receptor (AhR), we demonstrated APM induced AhR nuclear translocation by immunofluorescence and Western blotting and activation of the AhR response element using a luciferase reporter construct. Inhibitor studies suggest differential influences of polycyclic aromatic hydrocarbon signaling, ROS-mediated responses and endotoxin alter stress and proinflammatory endothelial cell responses. Our findings demonstrate gene responses correlated with current concepts that systemic inflammation drives cardiovascular effects of particulate air pollution. We also demonstrate a unique pattern of gene responses related to xenobiotic metabolism in PM-exposed HAEC.


Assuntos
Células Endoteliais/efeitos dos fármacos , Células Endoteliais/metabolismo , Material Particulado/toxicidade , Apoptose/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Células Cultivadas , Quimiocina CCL2/genética , Quimiocina CCL2/metabolismo , Quimiocina CXCL2/genética , Citocromo P-450 CYP1A1/genética , Sistema Enzimático do Citocromo P-450/genética , Selectina E/genética , Humanos , Interleucina-6/metabolismo , Análise de Sequência com Séries de Oligonucleotídeos , Reação em Cadeia da Polimerase , Receptores de Hidrocarboneto Arílico/genética
5.
Mol Cancer Res ; 8(8): 1105-15, 2010 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-20663862

RESUMO

Glioblastoma multiforme is an extremely aggressive and clinically unresponsive form of cancer. Transformed neoplastic neural stem cells, resistant to chemotherapy and radiation therapy, are thought to be responsible for the initial tumor formation and the recurrence of disease following surgical resection. These stem cells express multidrug resistance markers along with CD133. We show that ectopic overexpression of CD133 in rat C6 glioma cells leads to significant reluctance to undergo apoptosis from camptothecin and doxorubicin. Although p53 was upregulated in CD133-overexpressing glioma cells treated with DNA-damaging agents, apoptosis seems to be p53 independent. At least one ABC transporter, rat P-glycoprotein/ABCB1, was upregulated by 62% in CD133(+) cells with a corresponding increase in activity. Thus, the combination of higher P-glycoprotein mRNA transcription and elevated transporter activity seems to contribute to the protection from cytotoxic reagents. In conclusion, previous investigators have reported that resilient cancer stem cells coexpress CD133 and ABC transporters with increased reluctance toward apoptosis. Our data suggest that CD133 may contribute to the observed resistance to apoptosis of CD133(+) cancer stem cells.


Assuntos
Antígenos CD/metabolismo , Apoptose , Resistencia a Medicamentos Antineoplásicos , Glioma/metabolismo , Glioma/patologia , Glicoproteínas/metabolismo , Células-Tronco Neoplásicas/metabolismo , Células-Tronco Neoplásicas/patologia , Peptídeos/metabolismo , Antígeno AC133 , Membro 1 da Subfamília B de Cassetes de Ligação de ATP/metabolismo , Animais , Antibióticos Antineoplásicos/farmacologia , Antineoplásicos Fitogênicos/farmacologia , Western Blotting , Camptotecina/farmacologia , Proliferação de Células , Doxorrubicina/farmacologia , Glioma/tratamento farmacológico , Masculino , Células-Tronco Neoplásicas/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Ratos , Ratos Sprague-Dawley , Células Tumorais Cultivadas , Proteína Supressora de Tumor p53/metabolismo , Proteína X Associada a bcl-2/metabolismo
6.
Proteomics ; 5(17): 4398-413, 2005 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-16222722

RESUMO

A single injection of monocrotaline produces a pulmonary insult in rats with a phenotype similar to human primary pulmonary hypertension. Although extensively used as a model, the mechanism(s) by which this chemical insult mimics a condition with genetic and environmental links remains an enigma, although formation of protein adducts has been implicated. Monocrotaline (MCT) is non-toxic and must undergo hepatic dehydrogenation to the soft electrophile monocrotaline pyrrole as prerequisite to damaging endothelial cells lining arterioles at remote pulmonary sites. In this report we extend our earlier investigation (J. Biol. Chem. 2000, 275, 29091-29099) by examining protein adducts to lower abundance adducts, a pI range not covered before, and subcellular localization of adduct-forming proteins associated with plasma membranes. Human pulmonary artery endothelial cells were exposed to [(14)C]MCT pyrrole (MCTP) and protein targets were identified using 2-DE with IPG 4-11. Adducted proteins were identified by pI, apparent molecular weight, and PMF using MALDI-TOF MS. Results of this study show that the majority of adducts form on proteins that contain reactive thiols in a CXXC motif, such as protein disulfide isomerase A(3) (ERp57), protein disulfide isomerase (PDI), and endothelial PDI. These same proteins were the major adduct-forming proteins associated with the plasma membrane. Other proteins found to be targets were thioredoxin, galectin-1, reticulocalbin 1 and 3, cytoskeletal tropomyosin, mitochondrial ATP synthase beta-chain, annexin A2 and cofilin-1. For the first time, MCTP adducts were observed on proteins not known to contain cysteine residues. However, known reactive proteins including nucleophosmin did not form detectable adducts, potentially indicating that MCTP did not reach the interior of nucleus to the same extent as other cellular sites. These findings suggest that molecular events underlying MCTP toxicity are initiated at the plasma membrane or readily accessible subcellular regions including the cytosol and membranes of the endoplasmic reticulum and mitochondria.


Assuntos
Cisteína , Endotélio Vascular/química , Monocrotalina/análogos & derivados , Isomerases de Dissulfetos de Proteínas/química , Proteínas/química , Artéria Pulmonar/química , Sequência de Aminoácidos , Células Cultivadas , Coloides , Reagentes de Ligações Cruzadas , Eletroforese em Gel Bidimensional , Endotélio Vascular/enzimologia , Proteínas de Choque Térmico/química , Proteínas de Choque Térmico/isolamento & purificação , Humanos , Dados de Sequência Molecular , Fragmentos de Peptídeos/química , Isomerases de Dissulfetos de Proteínas/isolamento & purificação , Proteínas/isolamento & purificação , Artéria Pulmonar/enzimologia , Dióxido de Silício , Tropomiosina/química , Tropomiosina/isolamento & purificação
7.
Proteomics ; 3(4): 479-95, 2003 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-12687615

RESUMO

Many aspects of the toxicity of xenobiotic compounds have been attributed to the consequences of covalent modification of specific proteins, but the nature and specificity of protein targets for classes of electrophilic toxins remain largely uncharacterized. For inhaled toxicants, the point of exposure or absorption lies with epithelial cells lining the pulmonary tree. In this study, abundant proteins in human bronchial epithelial cells that are arylated in vitro by two quinonoid compounds, 1,4-benzoquinone (BQ) and 1,4-naphthoquinone (NQ) have been detected using (14)C-labeled quinones and two-dimensional gel electrophoresis. These proteins were identified using matrix assisted laser desorption/ionization mass spectrometry for tryptic mass mapping followed by sequence database searching. Corroborative identification of protein targets was obtained from the apparent isoelectric points, molecular weights, and the use of antibody probes. There were subtle differences in the protein targets of BQ and NQ, but both associated with the following abundant proteins, nucleophosmin, galectin-1, probable protein disulfide isomerase, protein disulfide isomerase, 60 kDa heat shock protein, mitochondrial stress-70 protein, epithelial cell marker protein, and S100-type calcium binding protein A14. We further delineate the properties of these proteins that make them preferred targets and the evidence these adducts present for delivery of these quinones to subcellular compartments.


Assuntos
Benzoquinonas/farmacologia , Brônquios/efeitos dos fármacos , Brônquios/metabolismo , Células Epiteliais/efeitos dos fármacos , Naftoquinonas/farmacologia , Proteínas/metabolismo , Adolescente , Adulto , Sequência de Aminoácidos , Autorradiografia , Brônquios/citologia , Células Cultivadas , Eletroforese em Gel Bidimensional , Células Epiteliais/metabolismo , Feminino , Humanos , Masculino , Espectrometria de Massas , Oxirredução , Mapeamento de Peptídeos , Proteínas/química
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA