Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
PLoS One ; 10(2): e0117254, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25692292

RESUMO

Primary pancreatic carcinoma has an unfavourable prognosis and standard treatment strategies mostly fail in advanced cases. Virotherapy might overcome this resistance to current treatment modalities. However, data from clinical studies with oncolytic viruses, including replicating adenoviral (Ad) vectors, have shown only limited activity against pancreatic cancer and other carcinomas. Since pancreatic carcinomas have a complex tumor architecture and frequently a strong stromal compartment consisting of non-neoplastic cell types (mainly pancreatic stellate cells = hPSCs) and extracellular matrix, it is not surprising that Ad vectors replicating in neoplastic cells will likely fail to eradicate this aggressive tumor type. Because the TGFß receptor (TGFBR) is expressed on both neoplastic cells and hPSCs we inserted the TGFBR targeting peptide CKS17 into the hypervariable region 5 (HVR5) of the capsid protein hexon with the aim to generate a replicating Ad vector with improved activity in complex tumors. We demonstrated increased transduction of both pancreatic cancer cell lines and of hPSCs and enhanced cytotoxicity in co-cultures of both cell types. Surface plasmon resonance analysis demonstrated decreased binding of coagulation factor X to CKS17-modified Ad particles and in vivo biodistribution studies performed in mice indicated decreased transduction of hepatocytes. Thus, to increase activity of replicating Ad vectors we propose to relax tumor cell selectivity by genetic hexon-mediated targeting to the TGFBR (or other receptors present on both neoplastic and non-neoplastic cells within the tumor) to enable replication also in the stromal cell compartment of tumors, while abolishing hepatocyte transduction, and thereby increasing safety.


Assuntos
Adenoviridae/genética , Proteínas do Capsídeo/genética , Terapia Genética , Vetores Genéticos/genética , Vírus Oncolíticos/genética , Neoplasias Pancreáticas/patologia , Engenharia de Proteínas , Adenoviridae/fisiologia , Sequência de Aminoácidos , Animais , Transporte Biológico , Proteínas do Capsídeo/química , Proteínas do Capsídeo/metabolismo , Linhagem Celular Tumoral , Proteína de Membrana Semelhante a Receptor de Coxsackie e Adenovirus/metabolismo , Fator X/metabolismo , Vetores Genéticos/metabolismo , Hepatócitos/patologia , Hepatócitos/virologia , Humanos , Imunoglobulina M/metabolismo , Macrófagos/metabolismo , Camundongos , Dados de Sequência Molecular , Vírus Oncolíticos/fisiologia , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/terapia , Neoplasias Pancreáticas/virologia , Células Estreladas do Pâncreas/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Receptor do Fator de Crescimento Transformador beta Tipo II , Receptores de Fatores de Crescimento Transformadores beta/metabolismo , Células Estromais/virologia , Transdução Genética , Tropismo Viral , Replicação Viral
2.
Biol Chem ; 395(9): 977-90, 2014 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-24854540

RESUMO

The 15 members of the kallikrein-related serine peptidase (KLK) family have diverse tissue-specific expression profiles and roles in a range of cellular processes, including proliferation, migration, invasion, differentiation, inflammation and angiogenesis that are required in both normal physiology as well as pathological conditions. These roles require cleavage of a range of substrates, including extracellular matrix proteins, growth factors, cytokines as well as other proteinases. In addition, it has been clear since the earliest days of KLK research that cleavage of cell surface substrates is also essential in a range of KLK-mediated cellular processes where these peptidases are essentially acting as agonists and antagonists. In this review we focus on these KLK-regulated cell surface receptor systems including bradykinin receptors, proteinase-activated receptors, as well as the plasminogen activator, ephrins and their receptors, and hepatocyte growth factor/Met receptor systems and other plasma membrane proteins. From this analysis it is clear that in many physiological and pathological settings KLKs have the potential to regulate multiple receptor systems simultaneously; an important issue when these peptidases and substrates are targeted in disease.


Assuntos
Proteínas de Membrana/metabolismo , Receptores de Superfície Celular/metabolismo , Calicreínas Teciduais/metabolismo , Animais , Humanos , Cininas/metabolismo , Especificidade por Substrato
3.
Neoplasia ; 15(8): 975-88, 2013 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-23908597

RESUMO

We sought to identify fibroblast growth factor receptor 2 (FGFR2) kinase domain mutations that confer resistance to the pan-FGFR inhibitor, dovitinib, and explore the mechanism of action of the drug-resistant mutations. We cultured BaF3 cells overexpressing FGFR2 in high concentrations of dovitinib and identified 14 dovitinib-resistant mutations, including the N550K mutation observed in 25% of FGFR2(mutant) endometrial cancers (ECs). Structural and biochemical in vitro kinase analyses, together with BaF3 proliferation assays, showed that the resistance mutations elevate the intrinsic kinase activity of FGFR2. BaF3 lines were used to assess the ability of each mutation to confer cross-resistance to PD173074 and ponatinib. Unlike PD173074, ponatinib effectively inhibited all the dovitinib-resistant FGFR2 mutants except the V565I gatekeeper mutation, suggesting ponatinib but not dovitinib targets the active conformation of FGFR2 kinase. EC cell lines expressing wild-type FGFR2 were relatively resistant to all inhibitors, whereas EC cell lines expressing mutated FGFR2 showed differential sensitivity. Within the FGFR2(mutant) cell lines, three of seven showed marked resistance to PD173074 and relative resistance to dovitinib and ponatinib. This suggests that alternative mechanisms distinct from kinase domain mutations are responsible for intrinsic resistance in these three EC lines. Finally, overexpression of FGFR2(N550K) in JHUEM-2 cells (FGFR2(C383R)) conferred resistance (about five-fold) to PD173074, providing independent data that FGFR2(N550K) can be associated with drug resistance. Biochemical in vitro kinase analyses also show that ponatinib is more effective than dovitinib at inhibiting FGFR2(N550K). We propose that tumors harboring mutationally activated FGFRs should be treated with FGFR inhibitors that specifically bind the active kinase.


Assuntos
Benzimidazóis/farmacologia , Imidazóis/farmacologia , Mutação , Piridazinas/farmacologia , Pirimidinas/farmacologia , Quinolonas/farmacologia , Receptor Tipo 2 de Fator de Crescimento de Fibroblastos/genética , Trifosfato de Adenosina/metabolismo , Animais , Sítios de Ligação/genética , Biocatálise/efeitos dos fármacos , Linhagem Celular , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Resistencia a Medicamentos Antineoplásicos/genética , Eletroforese em Gel de Poliacrilamida , Neoplasias do Endométrio/genética , Neoplasias do Endométrio/metabolismo , Neoplasias do Endométrio/patologia , Feminino , Humanos , Concentração Inibidora 50 , Modelos Moleculares , Fosforilação/efeitos dos fármacos , Inibidores de Proteínas Quinases/farmacologia , Estrutura Terciária de Proteína , Receptor Tipo 2 de Fator de Crescimento de Fibroblastos/química , Receptor Tipo 2 de Fator de Crescimento de Fibroblastos/metabolismo
4.
Mol Cancer ; 11: 75, 2012 Oct 05.
Artigo em Inglês | MEDLINE | ID: mdl-23039341

RESUMO

BACKGROUND: Melanoma is the most lethal form of skin cancer, but recent advances in molecularly targeted agents against the Ras/Raf/MAPK pathway demonstrate promise as effective therapies. Despite these advances, resistance remains an issue, as illustrated recently by the clinical experience with vemurafenib. Such acquired resistance appears to be the result of parallel pathway activation, such as PI3K, to overcome single-agent inhibition. In this report, we describe the cytotoxicity and anti-tumour activity of the novel MEK inhibitor, E6201, in a broad panel of melanoma cell lines (n = 31) of known mutational profile in vitro and in vivo. We further test the effectiveness of combining E6201 with an inhibitor of PI3K (LY294002) in overcoming resistance in these cell lines. RESULTS: The majority of melanoma cell lines were either sensitive (IC50 < 500 nM, 24/31) or hypersensitive (IC50 < 100 nM, 18/31) to E6201. This sensitivity correlated with wildtype PTEN and mutant BRAF status, whereas mutant RAS and PI3K pathway activation were associated with resistance. Although MEK inhibitors predominantly exert a cytostatic effect, E6201 elicited a potent cytocidal effect on most of the sensitive lines studied, as evidenced by Annexin positivity and cell death ELISA. Conversely, E6201 did not induce cell death in the two resistant melanoma cell lines tested. E6201 inhibited xenograft tumour growth in all four melanoma cell lines studied to varying degrees, but a more pronounced anti-tumour effect was observed for cell lines that previously demonstrated a cytocidal response in vitro. In vitro combination studies of E6201 and LY294002 showed synergism in all six melanoma cell lines tested, as defined by a mean combination index < 1. CONCLUSIONS: Our data demonstrate that E6201 elicits a predominantly cytocidal effect in vitro and in vivo in melanoma cells of diverse mutational background. Resistance to E6201 was associated with disruption of PTEN and activation of downstream PI3K signalling. In keeping with these data we demonstrate that co-inhibition of MAPK and PI3K is effective in overcoming resistance inherent in melanoma.


Assuntos
Lactonas/farmacologia , Melanoma/tratamento farmacológico , PTEN Fosfo-Hidrolase/genética , Inibidores de Proteínas Quinases/farmacologia , Proteínas Proto-Oncogênicas B-raf/genética , Animais , Antineoplásicos/farmacologia , Sobrevivência Celular/efeitos dos fármacos , Cromonas/farmacologia , Resistencia a Medicamentos Antineoplásicos , Feminino , Humanos , MAP Quinase Quinase Quinases/antagonistas & inibidores , MAP Quinase Quinase Quinases/metabolismo , Melanoma/enzimologia , Melanoma/genética , Camundongos , Camundongos Nus , Quinases de Proteína Quinase Ativadas por Mitógeno/antagonistas & inibidores , Quinases de Proteína Quinase Ativadas por Mitógeno/metabolismo , Morfolinas/farmacologia , Mutação , PTEN Fosfo-Hidrolase/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Proto-Oncogênicas B-raf/metabolismo , Ensaios Antitumorais Modelo de Xenoenxerto
5.
J Biol Chem ; 286(49): 42303-42315, 2011 Dec 09.
Artigo em Inglês | MEDLINE | ID: mdl-21994943

RESUMO

Reciprocal interactions between Src family kinases (SFKs) and focal adhesion kinase (FAK) are critical during changes in cell attachment. Recently it has been recognized that another SFK substrate, CUB-domain-containing protein 1 (CDCP1), is differentially phosphorylated during these events. However, the molecular processes underlying SFK-mediated phosphorylation of CDCP1 are poorly understood. Here we identify a novel mechanism in which FAK tyrosine 861 and CDCP1-Tyr-734 compete as SFK substrates and demonstrate cellular settings in which SFKs switch between these sites. Our results show that stable CDCP1 expression induces robust SFK-mediated phosphorylation of CDCP1-Tyr-734 with concomitant loss of p-FAK-Tyr-861 in adherent HeLa cells. SFK substrate switching in these cells is dependent on the level of expression of CDCP1 and is also dependent on CDCP1-Tyr-734 but is independent of CDCP1-Tyr-743 and -Tyr-762. In HeLa CDCP1 cells, engagement of SFKs with CDCP1 is accompanied by an increase in phosphorylation of Src-Tyr-416 and a change in cell morphology to a fibroblastic appearance dependent on CDCP1-Tyr-734. SFK switching between FAK-Tyr-861 and CDCP1-Tyr-734 also occurs during changes in adhesion of colorectal cancer cell lines endogenously expressing these two proteins. Consistently, increased p-FAK-Tyr-861 levels and a more epithelial morphology are seen in colon cancer SW480 cells silenced for CDCP1. Unlike protein kinase Cδ, FAK does not appear to form a trimeric complex with Src and CDCP1. These data demonstrate novel aspects of the dynamics of SFK-mediated cell signaling that may be relevant during cancer progression.


Assuntos
Antígenos CD/química , Moléculas de Adesão Celular/química , Proteína-Tirosina Quinases de Adesão Focal/química , Proteínas de Neoplasias/química , Tirosina/química , Quinases da Família src/metabolismo , Antígenos de Neoplasias , Sítios de Ligação , Adesão Celular , Linhagem Celular Tumoral , Membrana Celular/metabolismo , Progressão da Doença , Fibroblastos/metabolismo , Inativação Gênica , Células HeLa , Humanos , Microscopia Confocal/métodos , Fosforilação
6.
J Biol Chem ; 285(34): 26162-73, 2010 Aug 20.
Artigo em Inglês | MEDLINE | ID: mdl-20551327

RESUMO

CUB-domain-containing protein 1 (CDCP1) is an integral membrane glycoprotein with potential as a marker and therapeutic target for a number of cancers. Here we examine mechanisms regulating cellular processing of CDCP1. By analyzing cell lines exclusively passaged non-enzymatically and through use of a panel of protease inhibitors, we demonstrate that full-length 135 kDa CDCP1 is post-translationally processed in a range of cell lines by a mechanism involving serine protease activity, generating a C-terminal 70-kDa fragment. Immunopurification and N-terminal sequencing of this cell-retained fragment and detailed mutagenesis, show that proteolytic processing of CDCP1 occurs at two sites, Arg-368 and Lys-369. We show that the serine protease matriptase is an efficient, but not essential, cellular processor of CDCP1 at Arg-368. Importantly, we also demonstrate that proteolysis induces tyrosine phosphorylation of 70-kDa CDCP1 and recruitment of Src and PKCdelta to this fragment. In addition, Western blot and mass spectroscopy analyses show that an N-terminal 65-kDa CDCP1 ectodomain is shed intact from the cell surface. These data provide new insights into mechanisms regulating CDCP1 and suggest that the biological role of this protein and, potentially, its function in cancer, may be mediated by both 70-kDa cell retained and 65-kDa shed fragments, as well as the full-length 135-kDa protein.


Assuntos
Antígenos CD/metabolismo , Moléculas de Adesão Celular/metabolismo , Proteínas de Neoplasias/metabolismo , Proteína Quinase C-delta/metabolismo , Serina Proteases/metabolismo , Quinases da Família src/metabolismo , Antígenos de Neoplasias , Linhagem Celular , Humanos , Glicoproteínas de Membrana , Fragmentos de Peptídeos/biossíntese , Fosforilação , Transporte Proteico , Tirosina/metabolismo
7.
Mol Cancer Res ; 7(8): 1197-211, 2009 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-19671673

RESUMO

The function of CUB domain-containing protein 1 (CDCP1), a recently described transmembrane protein expressed on the surface of hematopoietic stem cells and normal and malignant cells of different tissue origin, is not well defined. The contribution of CDCP1 to tumor metastasis was analyzed by using HeLa carcinoma cells overexpressing CDCP1 (HeLa-CDCP1) and a high-disseminating variant of prostate carcinoma PC-3 naturally expressing high levels of CDCP1 (PC3-hi/diss). CDCP1 expression rendered HeLa cells more aggressive in experimental metastasis in immunodeficient mice. Metastatic colonization by HeLa-CDCP1 was effectively inhibited with subtractive immunization-generated, CDCP1-specific monoclonal antibody (mAb) 41-2, suggesting that CDCP1 facilitates relatively late stages of the metastatic cascade. In the chick embryo model, time- and dose-dependent inhibition of HeLa-CDCP1 colonization by mAb 41-2 was analyzed quantitatively to determine when and where CDCP1 functions during metastasis. Quantitative PCR and immunohistochemical analyses indicated that CDCP1 facilitated tumor cell survival soon after vascular arrest. Live cell imaging showed that the function-blocking mechanism of mAb 41-2 involved enhancement of tumor cell apoptosis, confirmed by attenuation of mAb 41-2-mediated effects with the caspase inhibitor z-VAD-fmk. Under proapoptotic conditions in vitro, CDCP1 expression conferred HeLa-CDCP1 cells with resistance to doxorubicin-induced apoptosis, whereas ligation of CDCP1 with mAb 41-2 caused additional enhancement of the apoptotic response. The functional role of naturally expressed CDCP1 was shown by mAb 41-2-mediated inhibition of both experimental and spontaneous metastasis of PC3-hi/diss. These findings confirm that CDCP1 functions as an antiapoptotic molecule and indicate that during metastasis CDCP1 facilitates tumor cell survival likely during or soon after extravasation.


Assuntos
Antígenos CD/metabolismo , Moléculas de Adesão Celular/metabolismo , Membrana Celular/metabolismo , Proteínas de Neoplasias/metabolismo , Neoplasias/metabolismo , Neoplasias/patologia , Animais , Anticorpos Monoclonais/farmacologia , Antígenos de Neoplasias , Apoptose/efeitos dos fármacos , Forma Celular/efeitos dos fármacos , Embrião de Galinha , Modelos Animais de Doenças , Doxorrubicina/farmacologia , Células HeLa , Humanos , Cinética , Masculino , Camundongos , Metástase Neoplásica/patologia , Fosfotirosina/metabolismo , Neoplasias da Próstata/metabolismo , Neoplasias da Próstata/patologia , Ensaio Tumoral de Célula-Tronco
8.
IUBMB Life ; 61(7): 723-30, 2009 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-19514048

RESUMO

In the last few years dysregulated expression of the cell surface glycoprotein CUB domain-containing protein 1 (CDCP1) has been associated with several cancers and this cell surface molecule has been recognized both as a tumor marker and as a potential target to disrupt progression of cancer. Here we summarize what is known about CDCP1 including its structural features, expression in normal and cancerous tissues, and the in vitro experiments and studies in animal models that have provided the key insights into its potential role in tumor formation and metastasis in humans. We conclude by highlighting opportunities and challenges in targeting CDCP1 in cancer.


Assuntos
Antígenos CD/fisiologia , Biomarcadores Tumorais/fisiologia , Moléculas de Adesão Celular/fisiologia , Proteínas de Neoplasias/fisiologia , Neoplasias/fisiopatologia , Sequência de Aminoácidos , Antígenos de Neoplasias , Progressão da Doença , Células-Tronco Hematopoéticas/fisiologia , Dados de Sequência Molecular , Neoplasias/tratamento farmacológico , Fosforilação , Alinhamento de Sequência , Células-Tronco/fisiologia , Quinases da Família src/fisiologia
9.
J Gene Med ; 10(12): 1303-14, 2008 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-18837065

RESUMO

BACKGROUND: Chemical capsid modification of adenovirus vectors with synthetic polymers has been shown to aid in overcoming typical barriers for adenovirus vector-mediated gene transfer. Carbohydrate-based polymers for covalent modification of adenovirus vectors have been largely neglected so far. We utilized a reductive amination strategy to generate a novel class of adenovirus-based glycovectors with a mannan derivative. METHODS: Reductive amination to covalently couple polysaccharides to the capsid surface of adenovirus serotype 5-based vectors was investigated utilizing an oxidized derivative of mannan. After biochemical and physical characterization of mannanylated vectors, their performance was analysed in vitro in cell lines and primary human cells, and in vivo in mice after local and systemic vector injection. RESULTS: We describe the successful modification of adenovirus vectors with large polysaccharides by reductive amination. The particles were efficiently modified, physically intact and, importantly, detargeted from the natural Coxsackie and adenovirus receptor/integrin pathway in vitro. In addition, they exhibited significantly decreased transduction of muscle after local delivery and of liver after systemic delivery in mice. However, despite the modification of 60% of capsid surface amino groups, mannanylated particles were unable to evade neutralizing anti-Ad5 antibodies. CONCLUSIONS: Mannanylated vectors are a paradigm for a novel class of glycoviruses modified with large polysaccharides. Vector promiscuity as one of the important hurdles for Ad-mediated gene transfer could be significantly decreased in vivo, whereas mannanylated vectors were unable to escape from anti-adenovirus antibodies. Our studies provide a detailed analysis of mannan-modified Ad vectors and suggest further improvements for this novel class of glycovectors.


Assuntos
Adenoviridae/genética , Capsídeo/metabolismo , Vetores Genéticos/química , Vetores Genéticos/metabolismo , Polissacarídeos/metabolismo , Adenoviridae/metabolismo , Animais , Capsídeo/química , Células Cultivadas , Feminino , Técnicas de Transferência de Genes , Vetores Genéticos/genética , Glicosilação , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Modelos Biológicos , Oxirredução , Transgenes
10.
Mol Ther ; 16(11): 1813-24, 2008 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-18714309

RESUMO

Adenovirus (Ad) vector targeting requires presentation of specific ligands on the virion's surface. Geneti-chemical targeting is based on the genetic introduction of cysteine residues bearing reactive thiol groups into solvent-accessible capsomeres of the virion and subsequent chemical coupling of ligands. Here, we exploited this technology to modify the pIX capsomere with high-affinity ligands. Genetic introduction of C-terminal cysteines to pIX allowed for specific coupling of full-length proteins to the virion, while not affecting vector production. Direct comparison of the two high-affinity ligands receptor- associated protein (RAP) and transferrin (Tf) revealed that targeting after coupling of a high-affinity ligand to pIX presumably requires release of the ligand from its receptor after cell entry. In addition, data obtained by live cell imaging of labeled vector particles demonstrated that coupling of very large proteins to pIX can impair intracellular vector particle trafficking. Finally, we demonstrate that the geneti-chemical targeting technology is suitable for in vivo targeting to liver after intravenous injection. Our data provide significant insight into basic requirements for successful targeting of pIX-modified Ad vectors.


Assuntos
Adenoviridae/genética , Proteínas do Capsídeo/genética , Vetores Genéticos , Proteína Associada a Proteínas Relacionadas a Receptor de LDL/química , Proteínas Relacionadas a Receptor de LDL/metabolismo , Animais , Biotina/química , Proteínas do Capsídeo/química , Linhagem Celular , Cisteína/genética , Feminino , Humanos , Ligantes , Maleimidas/química , Camundongos , Camundongos Endogâmicos BALB C , Receptores da Transferrina/metabolismo , Transferrina/química , Transferrina/metabolismo , Vírion/genética
11.
Mol Immunol ; 45(5): 1398-404, 2008 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-17950458

RESUMO

B-NK is a C-type lectin with an immunorecptor tyrosine-based inhibition motif, that is located in the vicinity of the chicken MHC and that has been described as a potential chicken NK cell receptor. We have generated an epitope tagged B-NK version for immunization and for biochemical studies. B-NK was expressed as a heavily glycosylated, homodimeric, type II transmembrane protein. With the help of a newly developed B-NK specific mab, the tissue distribution of B-NK has been analyzed. In the blood, B-NK was mainly present on a small population of gammadelta T cells, whereas in spleen it was expressed by alphabeta T cells. Moreover, B-NK was also found on CD3(-)CD8(+) sorted splenocytes that were in vitro expanded by IL-2 and on embryonic splenocytes, both of which resemble chicken NK cells. In order to characterize cells expressing B-NK ligands, a BWZ.36 based reporter system was employed, that induced beta-galactosidase activity upon ligand binding. While potential B-NK ligands such as MHC class I or the C-type lectin B-lec did not induce any signal, a trypsin sensitive B-NK ligand was expressed on phorbol myristate or concanavalin A activated splenocytes, but not unstimulated splenocytes. In summary, B-NK is expressed by NK cells and T cell subsets, and it binds to a ligand on activated cells.


Assuntos
Células Matadoras Naturais/química , Receptores Imunológicos/análise , Receptores Imunológicos/metabolismo , Baço/citologia , Subpopulações de Linfócitos T/química , Animais , Galinhas , Células Matadoras Naturais/imunologia , Lectinas Tipo C/análise , Lectinas Tipo C/metabolismo , Ligantes , Ligação Proteica , Receptores de Antígenos de Linfócitos T alfa-beta , Receptores de Antígenos de Linfócitos T gama-delta , Transdução de Sinais , Baço/química , Subpopulações de Linfócitos T/imunologia
12.
Mol Ther ; 16(1): 154-62, 2008 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-17848961

RESUMO

Genetic vaccination with adenoviral (Ad) gene transfer vectors requires transduction of professional antigen-presenting cells. However, because the natural Ad receptors are expressed on many cell types, the Ad vectors currently in use are characterized by high promiscuity. In fact, the majority of injected Ad vector particles are likely to transduce non-target cells. We have analyzed various sizes of polyethylene glycol (PEG) molecules for vector particle detargeting, and our data provide evidence that the size of the PEG determines detargeting efficiency. With the use of appropriately large PEG molecules, vector particles were detargeted from muscle after local delivery and from liver after systemic delivery in mouse models. Surprisingly, fully detargeted PEGylated Ad vectors still induced strong cellular and humoral immune responses to vector-encoded transgene products. Also, injection of PEGylated and non-PEGylated vector particles resulted in similar kinetics of transgene product-specific cytotoxic immune responses, thereby suggesting that the same cell types were involved in their induction. Furthermore, we showed that PEGylated vectors evade neutralizing anti-Ad antibodies in vivo. This feature might help circumvent the recognized limitation imposed by the widespread occurrence of anti-Ad immunity in the human population. We suggest that PEGylated Ad particles with significantly reduced promiscuity may qualify as a novel and safe vector format for genetic vaccination.


Assuntos
Adenoviridae/genética , Anticorpos Antivirais/sangue , Vetores Genéticos/imunologia , Vetores Genéticos/metabolismo , Polietilenoglicóis/metabolismo , Transdução Genética , Vacinas de DNA/imunologia , Vacinas Virais/genética , Adenoviridae/imunologia , Animais , Anticorpos Antivirais/biossíntese , Linhagem Celular Tumoral , Feminino , Vetores Genéticos/administração & dosagem , Humanos , Tolerância Imunológica/genética , Células K562 , Camundongos , Camundongos Endogâmicos BALB C , Peso Molecular , Vacinas de DNA/administração & dosagem , Vacinas de DNA/genética , Vacinas Virais/administração & dosagem , Vacinas Virais/imunologia
13.
J Nat Prod ; 69(7): 995-1000, 2006 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-16872131

RESUMO

The fungus Emericella nidulans var. acristata was isolated as an endophyte from a Mediterranean green alga. Cultivation of this fungus yielded two new compounds, arugosins G (1) and H (2), together with the known metabolites 3-9. Arugosins (1-4) are benzophenone derivatives, biosynthetically related to the xanthones 5, 6, and 9. The indole alkaloid 7 displayed antitumor activity in a panel of 36 human tumor cell lines, exhibiting a mean IC(50) value of 5.5 microg/mL in an in vitro survival and proliferation assay. Furthermore, compounds 3 and 4 showed moderate antitumor activity toward individual tumor cell lines. None of compounds 1-8 exhibited any immunostimulatory activity assessed as the capacity to induce cytokines in PBMCs from healthy donors.


Assuntos
Fungos/química , Compostos Heterocíclicos com 3 Anéis/isolamento & purificação , Fenóis/isolamento & purificação , Bacillus megaterium/efeitos dos fármacos , Chlorella/efeitos dos fármacos , Clorófitas , Citocinas/efeitos dos fármacos , Ensaios de Seleção de Medicamentos Antitumorais , Escherichia coli/efeitos dos fármacos , Compostos Heterocíclicos com 3 Anéis/química , Compostos Heterocíclicos com 3 Anéis/farmacologia , Humanos , Concentração Inibidora 50 , Biologia Marinha , Mar Mediterrâneo , Fenóis/química , Fenóis/farmacologia , Células Tumorais Cultivadas , Xantonas/química , Xantonas/isolamento & purificação , Xantonas/farmacologia
14.
J Immunol ; 176(10): 6270-6, 2006 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-16670338

RESUMO

Pancreatic carcinoma, the fifth leading cause of cancer-related mortality, frequently presents the stress-inducible heat shock protein 70 (Hsp70) on the cell membrane. Therefore, we explored an immunological approach exploiting the efficacy of NK cells activated either with low dose IL-2 plus Hsp70-peptide TKDNNLLGRFELSG (TKD; IL-2/TKD) or with IL-2 alone in a xenograft pancreatic carcinoma model. An orthotopic injection of either 2.5 x 10(6) or 1 x 10(6) Colo357 cells in SCID/beige mice resulted in rapidly growing primary tumors and the development of hepatic metastases on days 5 and 10, respectively. In line with results of in vitro migration assays, these NK cells also had the capacity to infiltrate pancreatic tumors and liver metastases in tumor-bearing mice. In vitro, a combined treatment of NK cells with IL-2/TKD but neither of the two treatments alone causes a profound increase in the lytic capacity against Hsp70 membrane-positive Colo357 cells. In vivo, a single i.v. injection of these NK cells on day 15 post-tumor inoculation resulted in a significant reduction in tumor weights, a delayed onset of hepatic metastases, and a prolonged life expectancy. In contrast, identically treated T cells and NK cells treated with IL-2 alone were significantly less efficient in controlling pancreatic tumors and metastases. Most importantly, four repeated i.v. infusions of IL-2/TKD-activated NK cells eradicated primary tumors and prevented hepatic metastases. In summary, our mouse data have implicated that NK cells preactivated with IL-2/TKD might provide a novel therapeutic tool for the treatment of aggressive, Hsp70-positive pancreatic carcinoma.


Assuntos
Adenocarcinoma/imunologia , Proteínas de Choque Térmico HSP70/fisiologia , Interleucina-2/fisiologia , Células Matadoras Naturais/imunologia , Neoplasias Pancreáticas/imunologia , Adenocarcinoma/secundário , Transferência Adotiva , Animais , Linhagem Celular Tumoral , Movimento Celular/imunologia , Feminino , Humanos , Células Matadoras Naturais/transplante , Neoplasias Hepáticas Experimentais/imunologia , Neoplasias Hepáticas Experimentais/prevenção & controle , Neoplasias Hepáticas Experimentais/secundário , Ativação Linfocitária/imunologia , Masculino , Camundongos , Camundongos SCID , Transplante de Neoplasias , Neoplasias Pancreáticas/patologia , Transplante Heterólogo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...