Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Br J Cancer ; 99(10): 1635-43, 2008 Nov 18.
Artigo em Inglês | MEDLINE | ID: mdl-19002182

RESUMO

Expression of the epithelial cell adhesion molecule EpCAM is upregulated in a variety of carcinomas. This antigen is therefore explored in tumour diagnosis, and clinical trials have been initiated to examine EpCAM-based therapies. Notably, the possible intracellular effects and signalling pathways triggered by EpCAM-specific antibodies are unknown. Here, we show treatment of the mouse lung carcinoma cell line A2C12, of the human lung carcinoma cell line A549 and the human colorectal cell line Caco-2 with the monoclonal EpCAM antibody G8.8 to cause dose dependently an increase in cell proliferation, as determined by the MTS and the 5'-bromo-2'-deoxyuridine (BrdU) labelling assay. Furthermore, a genome-wide approach identified networks of regulated genes, most notably cell cycle regulators, upon treatment with an EpCAM-specific antibody. Indeed, changes in the expression of cell cycle regulators agreed well with the BrdU labelling data, and an analysis of differentially expressed genes revealed the processes with the strongest over-representation of modulated genes, for example, cell cycle, cell death, cellular growth and proliferation, and cancer. These data suggest that EpCAM is involved in signal transduction triggering several intracellular signalling pathways. Knowing EpCAM signalling pathways might lead to a reassessment of EpCAM-based therapies.


Assuntos
Anticorpos Monoclonais/farmacologia , Antígenos de Neoplasias/metabolismo , Moléculas de Adesão Celular/metabolismo , Proteínas de Ciclo Celular/genética , Expressão Gênica/efeitos dos fármacos , Animais , Proteínas de Ciclo Celular/metabolismo , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Neoplasias Colorretais , Modelos Animais de Doenças , Molécula de Adesão da Célula Epitelial , Perfilação da Expressão Gênica , Genoma , Humanos , Neoplasias Pulmonares , Camundongos , Camundongos Transgênicos , Análise de Sequência com Séries de Oligonucleotídeos
2.
Br J Cancer ; 89(3): 564-72, 2003 Aug 04.
Artigo em Inglês | MEDLINE | ID: mdl-12888831

RESUMO

Specific ligands of the peripheral benzodiazepine receptor (PBR) are known to induce apoptosis and cell cycle arrest in oesophageal cancer cells. However, the underlying mechanisms are still unknown. Here, we investigated the transcriptional alterations and activation of protein kinases in response to PBR-specific ligands. Using cDNA arrays, we examined the transcriptional effects of the PBR-specific ligand FGIN-1-27 in two oesophageal cancer cell lines, KYSE-140 (squamous cell carcinoma) and OE-33 (adenocarcinoma). In oesophageal cancer cells, FGIN-1-27 induced extensive changes in the expression of genes involved in the regulation of apoptosis and cell cycle. Both in oesophageal cancer cell lines (KYSE-140, OE-33) we observed a strong upregulation of the growth arrest and DNA-damage-inducible genes, gadd45 and gadd153, in response to PBR ligands. gadd genes are known to be induced by p38MAPK activation. Using Western blotting we detected a time- and dose-dependent phosphorylation of p38MAPK, which was found to be functionally involved in gadd induction, apoptosis, and cell cycle arrest. In conclusion, our data indicate that PBR-specific ligands cause apoptosis and cell cycle arrest by activation of the p38MAPK pathway and induction of gadd45 and gadd153.


Assuntos
Apoptose , Carcinoma de Células Escamosas/genética , Carcinoma de Células Escamosas/patologia , Ciclo Celular , Neoplasias Esofágicas/genética , Neoplasias Esofágicas/patologia , Regulação Neoplásica da Expressão Gênica , Proteínas Quinases Ativadas por Mitógeno/farmacologia , Proteínas , Receptores de GABA-A/metabolismo , Western Blotting , Proteínas Estimuladoras de Ligação a CCAAT/biossíntese , Dano ao DNA , Humanos , Ácidos Indolacéticos/farmacologia , Peptídeos e Proteínas de Sinalização Intracelular , Ligantes , Análise de Sequência com Séries de Oligonucleotídeos , Fosforilação , Biossíntese de Proteínas , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transdução de Sinais , Fator de Transcrição CHOP , Fatores de Transcrição/biossíntese , Células Tumorais Cultivadas , Proteínas Quinases p38 Ativadas por Mitógeno , Proteínas GADD45
3.
Br J Cancer ; 88(8): 1217-22, 2003 Apr 22.
Artigo em Inglês | MEDLINE | ID: mdl-12698187

RESUMO

Patients with (previous) head and neck cancer (HNC) are at high risk for developing second squamous cell cancer of the oesophagus. The role of cyclooxygenase-2 (COX-2) in oesophageal squamous carcinogenesis has not yet been investigated in this high-risk group. Therefore, this study examined COX-2 mRNA and protein expression in oesophageal biopsies and resected tissues of 44 HNC patients. The evaluation covered 55 oesophageal tissue samples (18 invasive oesophageal squamous cell cancers, four high- and eight low-grade dysplasias, 25 normal squamous epithelia) from the 44 patients. mRNA levels of COX-2 were measured by real-time PCR using a LightCycler. COX-2 protein expression was studied immunohistochemically and graded by a staining score. COX-2 mRNA was detected in all samples, and its levels correlated positively with the immunohistochemical staining score (P<0.05). COX-2 expression was upregulated during oesophageal squamous carcinogenesis in HNC patients, that is COX-2 expression increased significantly from normal oesophageal squamous epithelium to low- and high-grade dysplasia and finally to invasive squamous cell cancer (P<0.001). Our findings suggest that COX-2 upregulation contributes to oesophageal squamous carcinogenesis in HNC patients. Prospective studies are needed to evaluate the chemopreventive potential of COX-2 inhibitors in this high-risk group.


Assuntos
Carcinoma de Células Escamosas/enzimologia , Neoplasias Esofágicas/enzimologia , Neoplasias de Cabeça e Pescoço/enzimologia , Isoenzimas/genética , Prostaglandina-Endoperóxido Sintases/genética , Sequência de Bases , Carcinoma de Células Escamosas/genética , Ciclo-Oxigenase 2 , Primers do DNA , Neoplasias Esofágicas/genética , Regulação Enzimológica da Expressão Gênica/genética , Regulação Neoplásica da Expressão Gênica/genética , Neoplasias de Cabeça e Pescoço/genética , Humanos , Proteínas de Membrana , RNA Mensageiro/genética , Transcrição Gênica/genética
4.
Int J Colorectal Dis ; 18(3): 239-47, 2003 May.
Artigo em Inglês | MEDLINE | ID: mdl-12673490

RESUMO

BACKGROUND AND AIMS: Photodynamic therapy (PDT) is a new treatment modality for early esophageal neoplasia. With two absorption maxima in the visible light range (550 and 588 nm) hypericin is a very promising photosensitizer for PDT with incoherent light sources. We studied the effects of photosensitizing hypericin in both primary cell cultures and cell lines (squamous: Kyse-140 and adenocarcinoma: OE-33) of human esophageal cancer using an incoherent white light source. MATERIALS AND METHODS: Esophageal cancer cells were preincubated (4-24 h) with hypericin (10 nM-1 micro M) and then irradiated with a light energy dose of 30 J/cm(2). RESULTS: Hypericin showed strong phototoxic effects and induced apoptosis in a dose-dependent fashion. The IC(50) value of hypericin phototoxicity was approximately 30 nM in both squamous and adenocarcinoma cells. In the concentrations used nonphotoactivated hypericin showed no toxic or apoptotic potency. The phototoxicity of hypericin was compared to that of delta-aminolevulinic acid (5-ALA), which is already being used for photodynamic therapy of gastrointestinal cancer. 5-ALA produced similar phototoxic effects but at a much higher dose (IC(50) 182+/-8 micro M in Kyse-140 and 308+/-40 micro M in OE-33 cells). Moreover, 5-ALA did not induce apoptosis to a relevant extent. CONCLUSION: Hypericin is a very promising new photosensitizer for innovative photodynamic therapy of esophageal cancer. Both the well known clinical safety of hypericin and the lower costs of broad band light sources argue in favor of clinical trials.


Assuntos
Adenocarcinoma/tratamento farmacológico , Antineoplásicos/uso terapêutico , Carcinoma de Células Escamosas/tratamento farmacológico , Neoplasias Esofágicas/tratamento farmacológico , Perileno/análogos & derivados , Perileno/farmacologia , Fotoquimioterapia/métodos , Fármacos Fotossensibilizantes/uso terapêutico , Ácido Aminolevulínico/uso terapêutico , Antracenos , Apoptose/efeitos dos fármacos , Relação Dose-Resposta à Radiação , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Fatores de Tempo , Células Tumorais Cultivadas/efeitos dos fármacos
5.
Br J Cancer ; 86(4): 636-44, 2002 Feb 12.
Artigo em Inglês | MEDLINE | ID: mdl-11870549

RESUMO

Extracellular ATP is known to inhibit growth of various tumours by activating specific purinergic receptors (P2-receptors). Since the therapy of advanced oesophageal cancer is unsatisfying, new therapeutic approaches are mandatory. Here, we investigated the functional expression and potential antiproliferative effects of P2-purinergic receptors in human oesophageal cancer cells. Prolonged incubation of primary cell cultures of human oesophageal cancers as well as of the squamous oesophageal cancer cell line Kyse-140 with ATP or its stable analogue ATP gamma S dose-dependently inhibited cell proliferation. This was due to both an induction of apoptosis and cell cycle arrest. The expression of P2-receptors was examined by RT-PCR, immunocytochemistry, and [Ca(2+)](i)-imaging. Application of various extracellular nucleotides dose-dependently increased [Ca(2+)](i). The rank order of potency was ATP=UTP>ATP gamma S>ADP=UDP. 2-methylthio-ATP and alpha,beta-methylene-ATP had no effects on [Ca(2+)](i). Complete cross-desensitization between ATP and UTP was observed. Moreover, the phospholipase C inhibitor U73122 dose-dependently reduced the ATP triggered [Ca(2+)](i) signal. The pharmacological features strongly suggest the functional expression of G-protein coupled P2Y(2)-receptors in oesophageal squamous cancer cells. P2Y(2)-receptors are involved in the antiproliferative actions of extracellular nucleotides. Thus, P2Y(2)-receptors are promising target proteins for innovative approaches in oesophageal cancer therapy.


Assuntos
Carcinoma de Células Escamosas/metabolismo , Divisão Celular/efeitos dos fármacos , Neoplasias Esofágicas/metabolismo , Nucleotídeos/farmacologia , Receptores Purinérgicos P2/metabolismo , Células Tumorais Cultivadas/efeitos dos fármacos , Apoptose/efeitos dos fármacos , Cálcio/metabolismo , Carcinoma de Células Escamosas/patologia , Caspase 3 , Caspases/metabolismo , Ciclo Celular/efeitos dos fármacos , Primers do DNA/química , Neoplasias Esofágicas/patologia , Imunofluorescência , Humanos , Marcação In Situ das Extremidades Cortadas , Receptores Purinérgicos P2Y2 , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Sódio/metabolismo
6.
Br J Cancer ; 85(11): 1771-80, 2001 Nov 30.
Artigo em Inglês | MEDLINE | ID: mdl-11742501

RESUMO

The peripheral benzodiazepine receptor (PBR) has been implicated in growth control of various tumour models. Although colorectal cancers were found to overexpress PBR, the functional role of PBR in colorectal cancer growth has not been addressed to date. Using primary cell cultures of human colorectal cancers and the human colorectal carcinoma cell lines HT29, LS174T, and Colo320 DM we studied the involvement of PBR in the growth control and apoptosis of colorectal cancers. Both mRNA and protein expression of PBR were detected by RT-PCR and flow cytometry. Using confocal laser scanning microscopy and immunohistochemistry the PBR was localized in the mitochondria. The specific PBR ligands FGIN-1-27, PK 11195, or Ro5-4864 inhibited cell proliferation dose-dependently. FGIN-1-27 decreased the mitochondrial membrane potential, which indicates an early event in apoptosis. Furthermore, FGIN-1-27, PK 11195 or Ro5-4864 increased caspase-3 activity. In addition to their apoptosis-inducing effects, PBR ligands induced cell cycle arrest in the G(1)/G(0)-phase. Thus, our data demonstrate a functional involvement of PBR in colorectal cancer growth and qualify the PBR as a possible target for innovative therapeutic approaches in colorectal cancer.


Assuntos
Apoptose/efeitos dos fármacos , Ciclo Celular/efeitos dos fármacos , Neoplasias Colorretais/patologia , Ligantes , Receptores de GABA-A/genética , Adulto , Idoso , Benzodiazepinonas/metabolismo , Benzodiazepinonas/farmacologia , Ciclo Celular/fisiologia , Neoplasias Colorretais/genética , Neoplasias Colorretais/fisiopatologia , Relação Dose-Resposta a Droga , Fase G1/efeitos dos fármacos , Fase G1/fisiologia , Regulação Neoplásica da Expressão Gênica , Células HT29 , Humanos , Ácidos Indolacéticos/metabolismo , Ácidos Indolacéticos/farmacologia , Membranas Intracelulares/efeitos dos fármacos , Membranas Intracelulares/fisiologia , Isoquinolinas/metabolismo , Isoquinolinas/farmacologia , Potenciais da Membrana/efeitos dos fármacos , Pessoa de Meia-Idade , Mitocôndrias/química , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/fisiologia , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Receptores de GABA-A/metabolismo , Fase de Repouso do Ciclo Celular/efeitos dos fármacos , Fase de Repouso do Ciclo Celular/fisiologia , Células Tumorais Cultivadas/efeitos dos fármacos
7.
Biochemistry ; 40(34): 10213-22, 2001 Aug 28.
Artigo em Inglês | MEDLINE | ID: mdl-11513599

RESUMO

The peripheral benzodiazepine receptor (PBR) is composed of three subunits with molecular masses of 18, 30, and 32 kDa. Many physiological functions have been attributed to the PBR, including regulation of steroidogenesis. Furthermore, the PBR itself is under hormonal regulation. In the current study, we investigated the role of female gonadal sex hormones in the regulation of PBR expression in steroidogenic and nonsteroidogenic tissues. To accomplish this, adult female rats were pharmacologically castrated using chronic administration of the gonadotropin-releasing hormone agonist decapeptyl (triptorelin-D-Trp(6)-LHRH). Half of these rats received 17beta-estradiol as hormone replacement, while a control group received daily injections of vehicle only. We found that PBR binding capacity dropped by 40 and 48% in ovaries and adrenals, respectively, following decapeptyl administration, as opposed to no change in the kidney. This down-regulation of PBR densities was prevented by estradiol replacement. We did not find evidence for transcriptional, posttranscriptional, and translational mechanisms in this decapeptyl-induced down-regulation. In contrast, immunoprecipitation of the PBR complex, using antibodies against the 18- and 32-kDa subunits of the complex, demonstrated that there were changes in PBR subunit interactions, consistent with the down-regulation of PBR binding capacity. These findings represent a novel hormone-dependent posttranslational regulatory mechanism.


Assuntos
Glândulas Suprarrenais/metabolismo , Regulação para Baixo/fisiologia , Estradiol/farmacologia , Ovário/metabolismo , Receptores de GABA-A/genética , Receptores de GABA-A/metabolismo , Pamoato de Triptorrelina/farmacologia , Animais , Regulação para Baixo/efeitos dos fármacos , Feminino , Isoquinolinas/farmacocinética , Rim/metabolismo , Cinética , Luteolíticos/farmacologia , Peso Molecular , Especificidade de Órgãos , Progesterona/sangue , Subunidades Proteicas , Ratos , Ratos Sprague-Dawley , Receptores de GABA-A/química , Transcrição Gênica/efeitos dos fármacos
8.
Int J Colorectal Dis ; 16(3): 154-66, 2001 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-11459289

RESUMO

Extracellular nucleotides induce apoptosis and inhibit growth of colorectal cancer cells. To understand the underlying signaling pathways, we investigated the role of nucleotide-sensitive P2 receptors and focused on the receptor-mediated signaling of intracellular Ca2+ and cyclic adenosine monophosphate (cAMP) in two colorectal carcinoma cell lines (HT29, Colo320 DM). Expression and functionality of P2 receptor subtypes evaluated by RT-PCR and [Ca2+]i imaging revealed that solely metabotropic P2 receptors of the subtype P2Y2 were expressed on a functional level in both cell lines. Short-term stimulation of P2Y2 receptors caused Ca2+ mobilization from intracellular stores and a subsequent transmembrane Ca2+ influx. The receptor-induced [Ca2+]i elevation was shown to increase basal-stimulated [cAMP]i moderately and to potentiate forskolin-stimulated [cAMP]i vigorously, since the effects were dose-dependently inhibited by preloading the cells with the [Ca2+]i chelator BAPTA. In contrast, activation of protein kinase C (PKC) did not contribute to a receptor-mediated rise in [cAMP]i, since the PKC inhibitor staurosporine completely failed to reduce P2Y2 receptor-induced increases in [cAMP]i. Prolonged application of P2Y2 receptor agonists induced a time-dependent increase in apoptosis (up to 50% above control values) in both cell lines and caused dose-dependent inhibition of cell proliferation of up to 85% (Colo320 DM) or 64% (HT29). Chelating [Ca2+]i with BAPTA almost completely abolished P2Y2 receptor-induced cell death. Rises in [cAMP]i elicited by either forskolin or cAMP derivatives inhibited growth in both cell lines, too. In line with the potentiating effect of P2Y2 receptors on forskolin-stimulated [cAMP]i increases, costimulation with forskolin and P2Y2 receptor agonists led to synergistic antiproliferative effects. Moreover, a synergistic growth inhibition was observed when coincubating the cells with the P2Y2 receptor agonist ATP and the cytostatic drug 5-fluorouracil, which forms the basis for most currently applied chemotherapeutic regimes in colorectal cancer treatment. Our results demonstrate the growth inhibitory potency of P2Y2 receptors in colorectal carcinoma cells. Receptor-induced [Ca2+]i signaling appears to play a major role in the observed antiproliferative and apoptosis-inducing effects.


Assuntos
Apoptose , Cálcio/metabolismo , Neoplasias Colorretais/patologia , AMP Cíclico/metabolismo , Receptores Purinérgicos P2/fisiologia , Trifosfato de Adenosina/farmacologia , Antimetabólitos Antineoplásicos/uso terapêutico , Divisão Celular/efeitos dos fármacos , Neoplasias Colorretais/metabolismo , Fluoruracila/uso terapêutico , Humanos , Receptores Purinérgicos P2/metabolismo , Receptores Purinérgicos P2Y2 , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Células Tumorais Cultivadas/efeitos dos fármacos , Uridina Trifosfato/farmacologia
9.
Mol Biol Cell ; 10(10): 3067-79, 1999 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-10512851

RESUMO

Haptokinetic cell migration across surfaces is mediated by adhesion receptors including beta1 integrins and CD44 providing adhesion to extracellular matrix (ECM) ligands such as collagen and hyaluronan (HA), respectively. Little is known, however, about how such different receptor systems synergize for cell migration through three-dimensionally (3-D) interconnected ECM ligands. In highly motile human MV3 melanoma cells, both beta1 integrins and CD44 are abundantly expressed, support migration across collagen and HA, respectively, and are deposited upon migration, whereas only beta1 integrins but not CD44 redistribute to focal adhesions. In 3-D collagen lattices in the presence or absence of HA and cross-linking chondroitin sulfate, MV3 cell migration and associated functions such as polarization and matrix reorganization were blocked by anti-beta1 and anti-alpha2 integrin mAbs, whereas mAbs blocking CD44, alpha3, alpha5, alpha6, or alphav integrins showed no effect. With use of highly sensitive time-lapse videomicroscopy and computer-assisted cell tracking techniques, promigratory functions of CD44 were excluded. 1) Addition of HA did not increase the migratory cell population or its migration velocity, 2) blocking of the HA-binding Hermes-1 epitope did not affect migration, and 3) impaired migration after blocking or activation of beta1 integrins was not restored via CD44. Because alpha2beta1-mediated migration was neither synergized nor replaced by CD44-HA interactions, we conclude that the biophysical properties of 3-D multicomponent ECM impose more restricted molecular functions of adhesion receptors, thereby differing from haptokinetic migration across surfaces.


Assuntos
Movimento Celular , Matriz Extracelular/metabolismo , Receptores de Hialuronatos/metabolismo , Integrinas/metabolismo , Melanoma/metabolismo , Proteínas de Membrana/metabolismo , Anticorpos Monoclonais/farmacologia , Sulfatos de Condroitina/metabolismo , Colágeno/metabolismo , Imunofluorescência , Humanos , Ácido Hialurônico/metabolismo , Processamento de Imagem Assistida por Computador , Glicoproteínas de Membrana , Microscopia de Vídeo , Complexo Glicoproteico GPIb-IX de Plaquetas , Receptores de Colágeno , Células Tumorais Cultivadas
10.
Cancer Res ; 57(10): 2061-70, 1997 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-9158006

RESUMO

The three-step model of cell migration consisting of protrusion of a leading lamella, attachment to the substrate, and contraction of the cell body is well established for fibroblasts migrating across planar surfaces. However, it is not resolved to what extent the migration of cancer cells in a 3-dimensional tissue environment follows similar principles. Here, we present evidence that the migration of highly invasive MV3 melanoma cells in 3-dimensional collagen matrices follows the three-step concept of migration but also results in characteristic reorganization of the extracellular matrix. After incorporation in the lattice, MV3 cells spontaneously developed a slow type of migration (mean velocity, 0.19 microm/min), leading to alignment of collagen fibers at attachment sites, as detected from unfixed and fixed samples by confocal reflection contrast in combination with immunofluorescence staining. In the process of migration, the formation of focal clusters or stripes of alpha2 and beta1 integrins colocalized with binding sites to collagen fibrils at the leading as well as the trailing edge. In contrast, CD44 was nonclustered and redistributed toward the rear end of the cell. At detachment sites, dynamic fiber traction, localized fiber disruption, and the release of cell surface determinants, including alpha2beta1 integrins and CD44, resulted in circumscribed matrix reorganization. Not infrequently, these emerging tube-like paths of least resistance bordered by a dense fiber network facilitated the reorientation and contact guidance of proximate MV3 cells to migrate along the preexisting path. In conclusion, the migration of MV3 cells in 3-dimensional collagen lattices resulted in dynamic tissue reorganization and receptor shedding the consequences of which were directly visualized by combining confocal reflection imaging with immunofluorescence.


Assuntos
Colágeno/fisiologia , Colágeno/ultraestrutura , Matriz Extracelular/fisiologia , Matriz Extracelular/ultraestrutura , Receptores de Hialuronatos/fisiologia , Integrinas/fisiologia , Melanoma/patologia , Animais , Bovinos , Adesão Celular/fisiologia , Comunicação Celular/fisiologia , Movimento Celular/fisiologia , Colágeno/metabolismo , Matriz Extracelular/metabolismo , Humanos , Receptores de Hialuronatos/metabolismo , Processamento de Imagem Assistida por Computador , Integrinas/metabolismo , Microscopia Confocal , Microscopia de Vídeo , Receptores de Colágeno
11.
Cancer Lett ; 118(2): 173-80, 1997 Oct 14.
Artigo em Inglês | MEDLINE | ID: mdl-9459207

RESUMO

Active cellular locomotion is a feature of such diverse cell types as lymphocytes and cancer cells. The locomotory phenotype of a cell should ultimately reflect the biochemical basis of different migratory strategies. We investigated the locomotory behavior of five epithelial cell lines and one non-epithelial human cell-line as well as human CD4+ T lymphocytes in a three-dimensional collagen type I matrix using time-lapse video microscopy and computer assisted cell-tracking. Migration velocity was up to 70 times lower in tumor cells (0.1-0.3 microm/min) as compared to T lymphocytes (7-7.5 microm/min), whereas the percentage of spontaneously active cells was up to twice as high in tumor cells (80-90%) in comparison to T lymphocytes (54%). Persistence, i.e. the degree of roaming, varied appreciably between the different cell types. In conclusion, velocity and persistence may describe distinct migration strategies in different cell types, i.e. discerning T cell migration from tumor cell invasion.


Assuntos
Linfócitos T/citologia , Células Tumorais Cultivadas/patologia , Movimento Celular , Colágeno , Matriz Extracelular , Humanos , Microscopia Confocal , Gravação em Vídeo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA