Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
1.
Haematologica ; 105(4): 937-950, 2020 04.
Artigo em Inglês | MEDLINE | ID: mdl-31248967

RESUMO

Glutathione peroxidase 4 (GPX4) is unique as it is the only enzyme that can prevent detrimental lipid peroxidation in vivo by reducing lipid peroxides to the respective alcohols thereby stabilizing oxidation products of unsaturated fatty acids. During reticulocyte maturation, lipid peroxidation mediated by 15-lipoxygenase in humans and rabbits and by 12/15-lipoxygenase (ALOX15) in mice was considered the initiating event for the elimination of mitochondria but is now known to occur through mitophagy. Yet, genetic ablation of the Alox15 gene in mice failed to provide evidence for this hypothesis. We designed a different genetic approach to tackle this open conundrum. Since either other lipoxygenases or non-enzymatic autooxidative mechanisms may compensate for the loss of Alox15, we asked whether ablation of Gpx4 in the hematopoietic system would result in the perturbation of reticulocyte maturation. Quantitative assessment of erythropoiesis indices in the blood, bone marrow (BM) and spleen of chimeric mice with Gpx4 ablated in hematopoietic cells revealed anemia with an increase in the fraction of erythroid precursor cells and reticulocytes. Additional dietary vitamin E depletion strongly aggravated the anemic phenotype. Despite strong extramedullary erythropoiesis reticulocytes failed to mature and accumulated large autophagosomes with engulfed mitochondria. Gpx4-deficiency in hematopoietic cells led to systemic hepatic iron overload and simultaneous severe iron demand in the erythroid system. Despite extremely high erythropoietin and erythroferrone levels in the plasma, hepcidin expression remained unchanged. Conclusively, perturbed reticulocyte maturation in response to Gpx4 loss in hematopoietic cells thus causes ineffective erythropoiesis, a phenotype partially masked by dietary vitamin E supplementation.


Assuntos
Eritropoese , Ferro , Fosfolipídeo Hidroperóxido Glutationa Peroxidase/genética , Reticulócitos , Vitamina E , Animais , Homeostase , Camundongos , Coelhos
2.
Am J Physiol Lung Cell Mol Physiol ; 315(1): L87-L101, 2018 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-29644893

RESUMO

Mesenchymal stromal cells (MSCs) are released into the airways of preterm infants following lung injury. These cells display a proinflammatory phenotype and are associated with development of severe bronchopulmonary dysplasia (BPD). We aimed to characterize the functional properties of MSCs obtained from tracheal aspirates of 50 preterm infants who required invasive ventilation. Samples were separated by disease severity. The increased proliferative capacity of MSCs was associated with longer duration of mechanical ventilation and higher severity of BPD. Augmented growth depended on nuclear accumulation of NFκBp65 and was accompanied by reduced expression of cytosolic α-smooth muscle actin (α-SMA). The central role of NF-κB signaling was confirmed by inhibition of IκBα phosphorylation. The combined score of proliferative capacity, accumulation of NFκBp65, and expression of α-SMA was used to predict the development of severe BPD with an area under the curve (AUC) of 0.847. We mimicked the clinical situation in vitro, and stimulated MSCs with IL-1ß and TNF-α. Both cytokines induced similar and persistent changes as was observed in MSCs obtained from preterm infants with severe BPD. RNA interference was employed to investigate the mechanistic link between NFκBp65 accumulation and alterations in phenotype. Our data indicate that determining the phenotype of resident pulmonary MSCs represents a promising biomarker-based approach. The persistent alterations in phenotype, observed in MSCs from preterm infants with severe BPD, were induced by the pulmonary inflammatory response. NFκBp65 accumulation was identified as a central regulatory mechanism. Future preclinical and clinical studies, aimed to prevent BPD, should focus on phenotype changes in pulmonary MSCs.


Assuntos
Displasia Broncopulmonar/metabolismo , Recém-Nascido Prematuro , Células-Tronco Mesenquimais/metabolismo , Transdução de Sinais , Traqueia/metabolismo , Fator de Transcrição RelA/metabolismo , Displasia Broncopulmonar/patologia , Feminino , Humanos , Lactente , Recém-Nascido , Interleucina-1beta/metabolismo , Masculino , Células-Tronco Mesenquimais/patologia , Traqueia/patologia , Fator de Necrose Tumoral alfa/metabolismo
3.
J Mol Med (Berl) ; 95(2): 169-180, 2017 02.
Artigo em Inglês | MEDLINE | ID: mdl-27576916

RESUMO

Early onset infection (EOI) in preterm infants <32 weeks gestational age (GA) is associated with a high mortality rate and the development of severe acute and long-term complications. The pathophysiology of EOI is not fully understood and clinical and laboratory signs of early onset infections in this patient cohort are often not conclusive. Thus, the aim of this study was to identify signatures characterizing preterm infants with EOI by using genome-wide gene expression (GWGE) analyses from umbilical arterial blood of preterm infants. This prospective cohort study was conducted in preterm infants <32 weeks GA. GWGE analyses using CodeLink human microarrays were performed from umbilical arterial blood of preterm infants with and without EOI. GWGE analyses revealed differential expression of 292 genes in preterm infants with EOI as compared to infants without EOI. Infants with EOI could be further differentiated into two subclasses and were distinguished by the magnitude of the expression of genes involved in both neutrophil and T cell activation. A hallmark activity for both subclasses of EOI was a common suppression of genes involved in natural killer (NK) cell function, which was independent from NK cell numbers. Significant results were recapitulated in an independent validation cohort. Gene expression profiling may enable early and more precise diagnosis of EOI in preterm infants. KEY MESSAGE: Gene expression (GE) profiling at birth characterizes preterm infants with EOI. GE analysis indicates dysregulation of NK cell activity. NK cell activity at birth may be a useful marker to improve early diagnosis of EOI.


Assuntos
Perfilação da Expressão Gênica , Doenças do Prematuro/diagnóstico , Recém-Nascido Prematuro , Infecções/diagnóstico , Idade de Início , Antígenos de Diferenciação de Linfócitos T/genética , Biomarcadores/sangue , Estudos de Coortes , Diagnóstico Precoce , Estudo de Associação Genômica Ampla , Humanos , Recém-Nascido , Doenças do Prematuro/genética , Infecções/genética , Células Matadoras Naturais/metabolismo , Subfamília C de Receptores Semelhantes a Lectina de Células NK/genética , Subfamília D de Receptores Semelhantes a Lectina de Células NK/genética , Neutrófilos/metabolismo , Estudos Prospectivos , RNA/sangue , Linfócitos T/metabolismo
4.
Int J Oncol ; 49(1): 99-110, 2016 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-27177126

RESUMO

Tumor side population (SP) cells display stem-like properties that can be modulated by treatment with the calcium channel blocker verapamil. Verapamil can enhance the cytotoxic effects of chemotherapeutic drugs and multidrug resistance by targeting the transport function of the P-glycoprotein (P-gp). This study focused on the therapeutic potential of verapamil on stem-like SP tumor cells, and further investigated its chemosensitizing effects using L3.6pl and AsPC-1 pancreatic carcinoma models. As compared to parental L3.6pl cells (0.9±0.22%), L3.6pl gemcitabine-resistant cells (L3.6plGres) showed a significantly higher percentage of SP cells (5.38±0.99%) as detected by Hoechst 33342/FACS assays. The L3.6plGres SP cells showed stable gemcitabine resistance, enhanced colony formation ability and increased tumorigenicity. Verapamil effectively inhibited L3.6plGres and AsPC-1 SP cell proliferation in vitro. A pro-apoptotic effect of verapamil was observed in L3.6pl cells, but not in L3.6plGres cells, which was linked to their differential expression of P-gp and equilibrative nucleoside transporter-1 (ENT-1). In an orthotopic pancreatic cancer mouse model, both low and high dose verapamil was shown to substantially reduce L3.6plGres-SP cell tumor growth and metastasis, enhance tumor apoptosis, and reduce microvascular density.


Assuntos
Membro 1 da Subfamília B de Cassetes de Ligação de ATP/biossíntese , Resistencia a Medicamentos Antineoplásicos/genética , Transportador Equilibrativo 1 de Nucleosídeo/biossíntese , Neoplasias Pancreáticas/tratamento farmacológico , Verapamil/administração & dosagem , Animais , Apoptose/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Transportador Equilibrativo 1 de Nucleosídeo/genética , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Humanos , Camundongos , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/patologia , Células da Side Population/efeitos dos fármacos , Ensaios Antitumorais Modelo de Xenoenxerto
5.
Target Oncol ; 10(4): 535-48, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-25639539

RESUMO

Our preliminary studies identified a small population side population (SP) cells in pancreatic cancer cells with stem cell-like properties, which were able to induce fast and aggressive tumor formation in nude mice. Gene expression analysis showed a significant difference in the expression of more than 1,300 genes in SP cells, among which a highly significant difference in microRNA expression of miR-21 and miR-221 between SP and NSP cells was identified. SP cells were identified and characterized by flow cytometry using Hoechst 33342 dye staining from a highly metastatic human pancreatic cancer cell line (L3.6pl). Antagomir transfection was performed using miRNA-21 and miRNA-221 antisense oligonucleotides (ASOs) and followed by detection of cell apoptosis, cell cycle progression, chemosensitivity, and invasion. Sorted SP cells from gemcitabine-resistant L3.6pl cells (L3.6pl(Gres)-SP) cells were orthotopically implanted in nude mice with or without miRNA-21 and miRNA-221 ASOs mono- and combination therapy. The administration of antagomir-21 and antagomir-221 significantly reduced the SP cell fraction, decreased SP cell differentiation, and downstream gene regulation, and thereby induced reduction of L3.6pl cell proliferation, invasion, and chemoresistance against gemcitabine and 5-Fluorouracil. Combination of ASOs therapy against miRNA-21 and miRNA-221 significantly inhibited primary tumor growth and metastasis compared to single antagomir treatment, especially, in L3.6plGres-SP-induced pancreatic tumor growth in vivo. These findings further indicate that the inhibition of miR-21 and miR-221 appear particularly suitable to target stem-like subpopulations and address their specific biological function to promote tumor progression in pancreatic cancer.


Assuntos
MicroRNAs/antagonistas & inibidores , Células-Tronco Neoplásicas/fisiologia , Oligonucleotídeos Antissenso/administração & dosagem , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/terapia , Animais , Carcinogênese/genética , Diferenciação Celular/genética , Linhagem Celular Tumoral , Proliferação de Células/genética , Resistencia a Medicamentos Antineoplásicos , Humanos , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , MicroRNAs/genética , MicroRNAs/metabolismo , Metástase Neoplásica , Células-Tronco Neoplásicas/patologia , Oligonucleotídeos Antissenso/genética , Neoplasias Pancreáticas/patologia , Transfecção , Regulação para Cima , Ensaios Antitumorais Modelo de Xenoenxerto
6.
Target Oncol ; 10(2): 215-27, 2015 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-24950733

RESUMO

Cancer stem cells (CSCs) have been proposed to underlie the initiation and maintenance of tumor growth and the development of chemoresistance in solid tumors. The identification and role of these important cells in pancreatic cancer remains controversial. Here, we isolate side population (SP) cells from the highly aggressive and metastatic human pancreatic cancer cell line L3.6pl and evaluate their potential role as models for CSCs. SP cells were isolated following Hoechst 33342 staining of L3.6pl cells. SP, non-SP, and unsorted L3.6pl cells were orthotopically xenografted into the pancreas of nude mice and tumor growth observed. RNA was analyzed by whole genome array and pathway mapping was performed. Drug resistant variants of L3.6pl were developed and examined for SP proportions and evaluated for surface expression of known CSC markers. A distinct SP with the ability to self-renew and differentiate into non-SP cells was isolated from L3.6pl (0.9 % ± 0.22). SP cells showed highly tumorigenic and metastatic characteristics after orthotopic injection. Transcriptomic analysis identified modulation of gene networks linked to tumorigenesis, differentiation, and metastasization in SP cells relative to non-SP cells. Wnt, NOTCH, and EGFR signaling pathways associated with tumor stem cells were altered in SP cells. When cultured with increasing concentrations of gemcitabine, the proportion of SP cells, ABCG2(+), and CD24(+) cells were significantly enriched, whereas 5-fluorouracil (5-FU) treatment lowered the percentage of SP cells. SP cells were distinct from cells positive for previously postulated pancreatic CSC markers. The Hoechst-induced side population in L3.6pl cells comprises a subset of tumor cells displaying aggressive growth and metastasization, increased gemcitabine-, but not 5-FU resistance. The cells may act as a partial model for CSC biology.


Assuntos
Adenocarcinoma/tratamento farmacológico , Antimetabólitos Antineoplásicos/farmacologia , Desoxicitidina/análogos & derivados , Resistencia a Medicamentos Antineoplásicos , Fluoruracila/farmacologia , Células-Tronco Neoplásicas/efeitos dos fármacos , Neoplasias Pancreáticas/tratamento farmacológico , Células da Side Population/efeitos dos fármacos , Adenocarcinoma/genética , Adenocarcinoma/metabolismo , Adenocarcinoma/secundário , Animais , Biomarcadores Tumorais/genética , Biomarcadores Tumorais/metabolismo , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Separação Celular , Desoxicitidina/farmacologia , Resistencia a Medicamentos Antineoplásicos/genética , Perfilação da Expressão Gênica/métodos , Regulação Neoplásica da Expressão Gênica , Redes Reguladoras de Genes , Humanos , Masculino , Camundongos Endogâmicos BALB C , Camundongos Nus , Células-Tronco Neoplásicas/metabolismo , Células-Tronco Neoplásicas/patologia , Análise de Sequência com Séries de Oligonucleotídeos , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/metabolismo , Neoplasias Pancreáticas/patologia , Fenótipo , Células da Side Population/metabolismo , Células da Side Population/patologia , Transdução de Sinais/efeitos dos fármacos , Fatores de Tempo , Ensaios Antitumorais Modelo de Xenoenxerto , Gencitabina
7.
Stem Cells Dev ; 23(2): 180-92, 2014 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-24021093

RESUMO

Dye-effluxing side population (SP) cells can be resistant to chemotherapy and are thought to resemble cancer stem cells. We characterized the relevance of the SP subpopulation in esophageal cancer cell lines and their relation to chemotherapy resistance and metastasis. The SP subpopulation was detected using Hoechst 33342 staining in five esophageal cancer cell lines OE19, OE21, OE33, PT1590, and LN1590. CTx-resistant cell lines were developed after long-term exposure to 5-fluorouracil (5-FU) and cisplatin and validated by analysis of resistance markers, thymidylate synthase and ERCC1. While neither LN1590 nor PT1590 had detectable SP cells, OE19, OE21, and OE33 cells were found to contain varying levels of SP cells. With increasing duration of 5-FU or cisplatin therapy, the SP subpopulation substantially emerged in PT1590 and LN1590. OE19-SP cells displayed significant higher tumorigenicity than OE19- non-SP (NSP) cells after subcutaneous tumor cell injection in vivo. SP cells isolated from OE19 and OE19/5-FUres were subsequently analyzed by an epithelial-to-mesenchymal transition (EMT) polymerase chain reaction array. Interestingly, the SP fraction of OE19/5-FUres showed a dramatic upregulation of EMT-related genes compared to the SP fraction of OE19. Our results provide evidence that (1) the proportion of SP cells is different in esophageal cancer, (2) SP cells exhibit stem cell properties and are associated to chemotherapy resistance, and (3) long-term CTx selects for SP cells with an upregulated EMT gene profile, which might be the source of systemic disease relapse. Further investigations are necessary to ideally target these EMT-associated SP cells in esophageal cancer.


Assuntos
Carcinoma de Células Escamosas/patologia , Resistencia a Medicamentos Antineoplásicos , Neoplasias Esofágicas/patologia , Metástase Neoplásica/patologia , Células-Tronco Neoplásicas/citologia , Células da Side Population/citologia , Subfamília B de Transportador de Cassetes de Ligação de ATP , Membro 1 da Subfamília B de Cassetes de Ligação de ATP/biossíntese , Membro 2 da Subfamília G de Transportadores de Cassetes de Ligação de ATP , Transportadores de Cassetes de Ligação de ATP/biossíntese , Adenocarcinoma/tratamento farmacológico , Adenocarcinoma/patologia , Animais , Antimetabólitos/farmacologia , Antineoplásicos/farmacologia , Carcinoma de Células Escamosas/tratamento farmacológico , Diferenciação Celular , Linhagem Celular Tumoral , Cisplatino/farmacologia , Proteínas de Ligação a DNA/biossíntese , Endonucleases/biossíntese , Transição Epitelial-Mesenquimal , Neoplasias Esofágicas/tratamento farmacológico , Carcinoma de Células Escamosas do Esôfago , Fluoruracila/farmacologia , Regulação Neoplásica da Expressão Gênica , Humanos , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Proteína 2 Associada à Farmacorresistência Múltipla , Proteínas Associadas à Resistência a Múltiplos Medicamentos/biossíntese , Proteínas de Neoplasias/biossíntese , Timidilato Sintase/biossíntese , beta Catenina/biossíntese
8.
Cancer Res ; 73(21): 6494-503, 2013 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-24030978

RESUMO

Carbonic anhydrase XII (CA XII) is a membrane-tethered cell surface enzyme that is highly expressed on many human tumor cells. Carbonic anhydrase members in this class of exofacial molecules facilitate tumor metabolism by facilitating CO2 venting and intracellular pH regulation. Accordingly, inhibition of exofacial CAs has been proposed as a general therapeutic strategy to target cancer. The recent characterization of 6A10, the first CA XII-specific inhibitory monoclonal antibody, offered an opportunity to evaluate this strategy with regard to CA XII-mediated catalysis. Using functional assays, we showed that 6A10 inhibited exofacial CA activity in CA XII-expressing cancer cells. 6A10 reduced spheroid growth in vitro under culture conditions where CA XII was active (i.e., alkaline pH) and where its catalytic activity was likely rate-limiting (i.e., restricted extracellular HCO3-supply). These in vitro results argued that the antibody exerted its growth-retarding effect by acting on the catalytic process, rather than on antigen binding per se. Notably, when administered in a mouse xenograft model of human cancer, 6A10 exerted a significant delay on tumor outgrowth. These results corroborate the notion that exofacial CA is critical for cancer cell physiology and they establish the immunotherapeutic efficacy of targeting CA XII using an inhibitory antibody.


Assuntos
Anticorpos Monoclonais/farmacologia , Inibidores da Anidrase Carbônica/farmacologia , Anidrases Carbônicas/química , Proliferação de Células/efeitos dos fármacos , Neoplasias/prevenção & controle , Animais , Apoptose/efeitos dos fármacos , Western Blotting , Anidrases Carbônicas/metabolismo , Catálise , Membrana Celular/efeitos dos fármacos , Membrana Celular/metabolismo , Imunofluorescência , Humanos , Concentração de Íons de Hidrogênio , Imunoprecipitação , Subunidade alfa de Receptor de Interleucina-2/fisiologia , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID , Neoplasias/enzimologia , Neoplasias/imunologia , Esferoides Celulares , Células Tumorais Cultivadas
9.
Mol Med ; 19: 54-61, 2013 Apr 30.
Artigo em Inglês | MEDLINE | ID: mdl-23552725

RESUMO

Trifunctional bispecific antibodies (trAbs) used in tumor immunotherapy have the unique ability to recruit T cells toward antigens on the tumor cell surface and, moreover, to activate accessory cells through their immunoglobulin Fc region interacting with activating Fcγ receptors. This scenario gives rise to additional costimulatory signals required for T cell-mediated tumor cell destruction and induction of an immunologic memory. Here we show in an in vitro system that most effective trAb-dependent T-cell activation and tumor cell elimination are achieved in the presence of dendritic cells (DCs). On the basis of these findings, we devise a novel approach of cancer immunotherapy that combines the specific advantages of trAbs with those of DC-based vaccination. Simultaneous delivery of trAbs and in vitro differentiated DCs resulted in a markedly improved tumor rejection in a murine melanoma model compared with monotherapy.


Assuntos
Anticorpos Biespecíficos/farmacologia , Células Dendríticas/imunologia , Imunoterapia , Melanoma/terapia , Linfócitos T/imunologia , Animais , Linhagem Celular Tumoral , Células Cultivadas , Técnicas de Cocultura , Feminino , Melanoma/imunologia , Camundongos , Camundongos Endogâmicos C57BL
10.
Cancer Res ; 72(16): 3958-66, 2012 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-22745368

RESUMO

A major goal of tumor immunotherapy is the induction of long-lasting systemic T-cell immunity. Bispecific antibodies (bsAbs) that lack the immunoglobulin Fc region confer T-cell-mediated killing of tumor cells but do not induce long-term memory. In contrast, trifunctional bsAbs comprise an appropriate Fc region and, therefore, not only recruit T cells but also accessory cells that bear activating Fcγ receptors (FcγR), providing additional T-cell-activating signals and securing presentation of tumor-derived antigens to T cells. In this study, we show that trifunctional bsAbs induce a polyvalent T-cell response and, therefore, a vaccination effect. Mice were treated with melanoma cells and with a trifunctional bsAb directed against the melanoma target antigen ganglioside GD2 in addition to murine CD3. The trifunctional bsAb activated dendritic cells and induced a systemic immune response that was not replicated by treatment with the F(ab')2-counterpart lacking the Fc region. Restimulation of spleen and lymph node cells in vitro yielded T-cell lines that specifically produced interferon-γ in response to tumor. In addition, trifunctional bsAb-induced T cells recognized various specific peptides derived from melanoma-associated antigens. Moreover, these polyvalent responses proved to be tumor-suppressive and could not be induced by the corresponding bsF(ab')2-fragment. Taken together, our findings provide preclinical proof of concept that trifunctional bsAbs can induce tumor-specific T cells with defined antigen specificity.


Assuntos
Anticorpos Biespecíficos/farmacologia , Vacinas Anticâncer/farmacologia , Epitopos de Linfócito T/imunologia , Melanoma Experimental/terapia , Linfócitos T/imunologia , Animais , Anticorpos Biespecíficos/imunologia , Complexo CD3/imunologia , Vacinas Anticâncer/imunologia , Feminino , Gangliosídeos/imunologia , Ativação Linfocitária , Melanoma Experimental/imunologia , Camundongos , Camundongos Endogâmicos C57BL
11.
Exp Neurol ; 234(1): 208-19, 2012 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-22230665

RESUMO

The interaction of human mesenchymal stem cells (hMSCs) and tumor cells has been investigated in various contexts. HMSCs are considered as cellular treatment vectors based on their capacity to migrate towards a malignant lesion. However, concerns about unpredictable behavior of transplanted hMSCs are accumulating. In malignant gliomas, the recruitment mechanism is driven by glioma-secreted factors which lead to accumulation of both, tissue specific stem cells as well as bone marrow derived hMSCs within the tumor. The aim of the present work was to study specific cellular interactions between hMSCs and glioma cells in vitro. We show, that glioma cells as well as hMSCs differentially express connexins, and that they interact via gap-junctional coupling. Besides this so-called functional syncytium formation, we also provide evidence of cell fusion events (structural syncytium). These complex cellular interactions led to an enhanced migration and altered proliferation of both, tumor and mesenchymal stem cell types in vitro. The presented work shows that glioma cells display signs of functional as well as structural syncytium formation with hMSCs in vitro. The described cellular phenomena provide new insight into the complexity of interaction patterns between tumor cells and host cells. Based on these findings, further studies are warranted to define the impact of a functional or structural syncytium formation on malignant tumors and cell based therapies in vivo.


Assuntos
Conexina 43/metabolismo , Expressão Gênica/fisiologia , Células Gigantes/fisiologia , Glioma/fisiopatologia , Células-Tronco Mesenquimais/fisiologia , Aminoácidos , Linhagem Celular Tumoral , Movimento Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Técnicas de Cocultura , Conexina 43/genética , Meios de Cultivo Condicionados/farmacologia , Dextranos , Expressão Gênica/efeitos dos fármacos , Células Gigantes/efeitos dos fármacos , Células Gigantes/patologia , Glioma/patologia , Humanos , RNA Mensageiro/metabolismo , Rodaminas , Frações Subcelulares
12.
Clin Exp Med ; 12(1): 25-30, 2012 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-21519961

RESUMO

The hedgehog (Hh) signaling pathway has been shown to be activated in the cancer stem cells of several tumor entities. The Hh inhibitor GDC-0449 has been proven to be effective in some cancers but not yet in lung cancer. We aimed at investigating whether GDC-0449 is effective in the lung cancer cell lines HCC (adenocarcinoma) and H1339 (small-cell-lung carcinoma), whether in these cell lines stem cell-like side populations (SPs) can be identified, and whether possible effects of GDC-0449 are mediated via SPs. SPs were identified by spectrum shift and decreased fluorescence after staining with 2.5 µg/ml Hoechst 33342. Expression of proteins was quantified by immunofluorescence. GDC-0449 (25 and 50 µM) inhibited concentration-dependent cell growth in HCC and H1339 cells. Further, the inhibitory effects of cisplatin on cell growth were augmented. In HCC and H1339 cell lines, SPs of 0.57 and 0.46% could be identified, respectively. SP, but not non-SP, cells were able to repopulate the original tumor population. The Hh receptor smoothened was detectable in SP but not in non-SP cells, showing the activation of the Hh pathway only in SPs. GDC-0449 considerably reduced SPs in HCC and H1339 cells. We demonstrate for the first time that GDC-0449 effectively reduces cell growth in lung cancer cell lines. This effect is mediated by the inhibition of stem cell-like SPs.


Assuntos
Anilidas/metabolismo , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Proteínas Hedgehog/antagonistas & inibidores , Neoplasias Pulmonares/metabolismo , Piridinas/metabolismo , Transdução de Sinais , Antineoplásicos/farmacologia , Protocolos de Quimioterapia Combinada Antineoplásica/farmacologia , Benzimidazóis/metabolismo , Linhagem Celular Tumoral , Cisplatino/farmacologia , Citometria de Fluxo , Fluorescência , Imunofluorescência , Humanos , Neoplasias Pulmonares/patologia , Carcinoma de Pequenas Células do Pulmão/metabolismo , Carcinoma de Pequenas Células do Pulmão/patologia , Fatores de Tempo
13.
Am J Pathol ; 179(1): 436-51, 2011 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-21703422

RESUMO

Tissue dendritic cells (DCs) may influence the progression of renal cell carcinoma (RCC) by regulating the functional capacity of antitumor effector cells. DCs and their interaction with T cells were analyzed in human RCC and control kidney tissues. The frequency of CD209(+) DCs in RCCs was found to be associated with an unfavorable T(H)1 cell balance in the tissue and advanced tumor stages. The CD209(+) DCs in RCC were unusual because most of them co-expressed macrophage markers (CD14, CD163). The phenotype of these enriched-in-renal-carcinoma DCs (ercDCs) could be reiterated in vitro by carcinoma-secreted factors (CXCL8/IL-8, IL-6, and vascular endothelial growth factor). ErcDCs resembled conventional DCs in costimulatory molecule expression and antigen cross-presentation. They did not suppress cognate cytotoxic T-lymphocyte function and did not cause CD3ζ down-regulation, FOXP3 induction, or T-cell apoptosis in situ or in vitro; thus, they are different from classic myeloid-derived suppressor cells. ErcDCs secreted high levels of metalloproteinase 9 and used T-cell crosstalk to increase tumor-promoting tumor necrosis factor α and reduce chemokines relevant for T(H)1-polarized lymphocyte recruitment. This modulation of the tumor environment exerted by ercDCs suggests an immunologic mechanism by which tumor control can fail without involving cytotoxic T-lymphocyte inhibition. Pharmacologic targeting of the deviated DC differentiation could improve the efficacy of immunotherapy against RCC.


Assuntos
Carcinoma de Células Renais/imunologia , Células Dendríticas/imunologia , Neoplasias Renais/imunologia , Linfócitos T Citotóxicos/imunologia , Linfócitos T/imunologia , Adulto , Idoso , Idoso de 80 Anos ou mais , Apoptose , Western Blotting , Carcinoma de Células Renais/metabolismo , Carcinoma de Células Renais/patologia , Estudos de Casos e Controles , Diferenciação Celular , Movimento Celular , Proliferação de Células , Quimiocinas/metabolismo , Apresentação Cruzada , Citocinas/metabolismo , Células Dendríticas/metabolismo , Células Dendríticas/patologia , Endocitose , Feminino , Citometria de Fluxo , Imunofluorescência , Humanos , Técnicas Imunoenzimáticas , Interleucina-6/metabolismo , Neoplasias Renais/metabolismo , Neoplasias Renais/patologia , Masculino , Pessoa de Meia-Idade , Fagocitose , RNA Mensageiro/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Linfócitos T/metabolismo , Linfócitos T/patologia , Linfócitos T Citotóxicos/metabolismo , Linfócitos T Citotóxicos/patologia , Células Th1/imunologia , Células Th1/metabolismo , Células Th1/patologia , Células Tumorais Cultivadas
14.
Cancer Immunol Immunother ; 60(5): 649-58, 2011 May.
Artigo em Inglês | MEDLINE | ID: mdl-21298264

RESUMO

The carbonic anhydrases (CAs) constitute a family of almost ubiquitous enzymes of significant importance for many physiological and pathological processes. CAs reversely catalyse the conversion of CO(2) + H(2)O to HCO(3) (-) and H(+), thereby contributing to the regulation of intracellular pH. Above all, CAs are of key importance for cells that perform glycolysis that inevitably leads to the intracellular accumulation of lactate. CA XII is a plasma membrane-associated isoform of the enzyme, which is induced by hypoxia and oestrogen and, consequently, expressed at high levels on various types of cancer and, intriguingly, on cancer stem cells. The enzyme is directly involved in tumour progression, and its inhibition has an anti-tumour effect. Apart from its role in carcinogenesis, the enzyme contributes to various other diseases like glaucoma and arteriosclerotic plaques, among others. CA XII is therefore regarded as promising target for specific therapies. We have now generated the first monoclonal antibody (6A10) that binds to the catalytic domain of CA XII on vital tumour cells and inhibits CA XII enzyme activity at nanomolar concentrations and thus much more effective than acetazolamide. In vitro results demonstrate that inhibition of CA XII by 6A10 inhibits the growth of tumour cells in 3-dimensional structures. In conclusion, we generated the first specific and efficient biological inhibitor of tumour-associated CA XII. This antibody may serve as a valuable tool for in vivo diagnosis and adjuvant treatment of different types of cancer.


Assuntos
Inibidores da Anidrase Carbônica/farmacologia , Anidrases Carbônicas/imunologia , Neoplasias/enzimologia , Neoplasias/imunologia , Animais , Anticorpos Monoclonais/metabolismo , Inibidores da Anidrase Carbônica/química , Anidrases Carbônicas/genética , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Citometria de Fluxo , Imunofluorescência , Expressão Gênica , Concentração de Íons de Hidrogênio , Proteínas de Membrana/imunologia , Ratos
16.
Cancer Res ; 69(13): 5331-9, 2009 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-19509230

RESUMO

Human mesenchymal stem cells (hMSC) aid in tissue maintenance and repair by differentiating into specialized cell types. Due to this ability, hMSC are currently being evaluated for cell-based therapies of tissue injury and degenerative diseases. However, extensive expansion ex vivo is a prerequisite to obtain the cell numbers required for human cell-based therapy protocols. Recent studies indicate that hMSC may contribute to cancer development and progression either by acting as cancer-initiating cells or through interactions with stromal elements. If spontaneous transformation ex vivo occurs, this may jeopardize the use of hMSC as therapeutic tools. Whereas murine MSC readily undergo spontaneous transformation, there are conflicting reports about spontaneous transformation of hMSC. We have addressed this controversy in a two-center study by growing bone marrow-derived hMSC in long-term cultures (5-106 weeks). We report for the first time spontaneous malignant transformation to occur in 45.8% (11 of 24) of these cultures. In comparison with hMSC, the transformed mesenchymal cells (TMC) showed a significantly increased proliferation rate and altered morphology and phenotype. In contrast to hMSC, TMC grew well in soft agar assays and were unable to undergo complete differentiation. Importantly, TMC were highly tumorigenic, causing multiple fast-growing lung deposits when injected into immunodeficient mice. We conclude that spontaneous malignant transformation may represent a biohazard in long-term ex vivo expansion of hMSC. On the other hand, this spontaneous transformation process may represent a unique model for studying molecular pathways initiating malignant transformation of hMSC.


Assuntos
Células da Medula Óssea/citologia , Transformação Celular Neoplásica , Células-Tronco Mesenquimais/citologia , Adolescente , Adulto , Animais , Células da Medula Óssea/patologia , Técnicas de Cultura de Células/métodos , Diferenciação Celular , Divisão Celular , Citometria de Fluxo , Humanos , Cinética , Células-Tronco Mesenquimais/patologia , Camundongos , Pessoa de Meia-Idade , Fenótipo , Valores de Referência , Especificidade da Espécie , Fatores de Tempo , Adulto Jovem
17.
Transgenic Res ; 18(1): 71-87, 2009 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-18819014

RESUMO

Murine embryonic stem cells (mESCs) inoculated at passage P13 with the mycoplasma species M. hominis, M. fermentans and M. orale and cultured over 20 passages showed reduced growth rate and viability (P < 0.0001) compared to control mESCs. Spectral karyotypic analysis of mycoplasma-infected mESCs showed a number of non-clonal chromosomal aberrations which increased with the duration of infection. The differentiation status of the infected mESCs was most affected at passage P13+6 where the infection was strongest and 46.3% of the mESCs expressed both POU5F1 and SSEA-1 markers whereas 84.8% of control mESCs expressed both markers. The percentage of germline chimeras from mycoplasma-infected mESCs was examined after blastocyst injection and embryo transfer to suitable recipients at different passages and, compared to the respective control group, was most affected at passage P13+5 (50% vs. 90%; P < 0.07). Further reductions were obtained at the same passage in the percentage of litters born (50% vs. 100%; P < 0.07) and in the percentage of pups born (22% vs. 45%; P < 0.001). Thirty three chimeras (39.8%) obtained from blastocyst injection with mycoplasma-infected mESCs showed reduced body weight (P < 0.0001), nasal discharge, osteoarthropathia, and cachexia. Flow cytometric analysis of plasma from chimeras produced with mycoplasma-infected mESCs revealed statistically significant differences in the proportions of T-cells and increased levels of IgG1 (P < 0.001), IgG2a (P < 0.05) and IgM (P < 0.05), anti-DNA antibodies (P < 0.05) and rheumatoid factor (P < 0.01). The present data indicate that mycoplasma contamination of mESCs affects various cell parameters, germline transmission, and postnatal development of the resulting chimeras.


Assuntos
Quimera/fisiologia , Embrião de Mamíferos/citologia , Células-Tronco Embrionárias/microbiologia , Células Germinativas/fisiologia , Mycoplasma/fisiologia , Animais , Biomarcadores/análise , Blastocisto/microbiologia , Blastocisto/fisiologia , Diferenciação Celular , Sobrevivência Celular , Quimera/microbiologia , Células-Tronco Embrionárias/citologia , Células-Tronco Embrionárias/metabolismo , Feminino , Fibroblastos/citologia , Imunoglobulina G/metabolismo , Cariotipagem , Masculino , Camundongos , Mycoplasma/isolamento & purificação , Gravidez
18.
PLoS One ; 4(12): e8539, 2009 Dec 31.
Artigo em Inglês | MEDLINE | ID: mdl-20046882

RESUMO

A chimeric HLA-DR4-H2-E (DR4) homozygous transgenic mouse line spontaneously develops diverse hematological malignancies with high frequency (70%). The majority of malignancies were distributed equally between T and B cell neoplasms and included lymphoblastic T cell lymphoma (LTCL), lymphoblastic B cell lymphoma (LBCL), diffuse large B cell lymphoma (DLBCL), the histiocyte/T cell rich variant of DLBCL (DLBCL-HA/T cell rich DLBCL), splenic marginal zone lymphoma (SMZL), follicular B cell lymphoma (FBL) and plasmacytoma (PCT). Most of these neoplasms were highly similar to human diseases. Also, some non-lymphoid malignancies such as acute myeloid leukemia (AML) and histiocytic sarcoma were found. Interestingly, composite lymphomas, including Hodgkin-like lymphomas, were also detected that had CD30(+) Hodgkin/Reed-Sternberg (H/RS)-like cells, representing a tumor type not previously described in mice. Analysis of microdissected H/RS-like cells revealed their origin as germinal center B cells bearing somatic hypermutations and, in some instances, crippled mutations, as described for human Hodgkin lymphoma (HL). Transgene integration in an oncogene was excluded as an exclusive driving force of tumorigenesis and age-related lymphoma development suggests a multi-step process. Thus, this DR4 line is a useful model to investigate common molecular mechanisms that may contribute to important neoplastic diseases in man.


Assuntos
Quimerismo , Neoplasias Hematológicas/imunologia , Neoplasias Hematológicas/patologia , Antígenos de Histocompatibilidade Classe II/imunologia , Doença de Hodgkin/imunologia , Doença de Hodgkin/patologia , Animais , Sequência de Bases , Antígeno Ki-1/metabolismo , Linfoma de Células B/patologia , Linfoma de Células T/patologia , Camundongos , Camundongos Transgênicos , Dados de Sequência Molecular , Células de Reed-Sternberg/patologia , Análise de Sobrevida , Transgenes/genética
19.
J Transl Med ; 5: 16, 2007 Mar 14.
Artigo em Inglês | MEDLINE | ID: mdl-17359532

RESUMO

BACKGROUND: Dendritic cells (DC) pulsed with tumor-derived antigenic material have widely been used in antitumor vaccination protocols. However, the optimal strategy of DC loading has not yet been established. Our aim was to define requirements of optimal DC vaccines in terms of in vivo protection in a murine B-cell lymphoma model. METHODS: We compare various loading reagents including whole parental and modified tumor cells and a single tumor-specific antigen, namely the lymphoma idiotype (Id). Bone marrow-derived DC were pulsed in vitro and used for therapy of established A20 lymphomas. RESULTS: We show that a vaccine with superior antitumor efficacy can be generated when DC are loaded with whole modified tumor cells which provide both (i) antigenic polyvalency and (ii) receptor-mediated antigen internalization. Uptake of cellular material was greatly enhanced when the tumor cells used for DC pulsing were engineered to express an anti-Fc receptor immunoglobulin specificity. Upon transfer of these DC, established tumor burdens were eradicated in 50% of mice. By contrast, pulsing DC with unmodified lymphoma cells or with the lymphoma Id, even when it was endowed with the anti-Fc receptor binding arm, was far less effective. A specific humoral anti-Id response could be detected, particularly following delivery of Id protein-pulsed DC, but it was not predictive of tumor protection. Instead a T-cell response was pivotal for successful tumor protection. Interaction of the transferred DC with CD8+ T lymphocytes seemed to play a role for induction of the immune response but was dispensable when DC had received an additional maturation stimulus. CONCLUSION: Our analyses show that the advantages of specific antigen redirection and antigenic polyvalency can be combined to generate DC-based vaccines with superior antitumor efficacy. This mouse model may provide information for the standardization of DC-based vaccination protocols.


Assuntos
Vacinas Anticâncer/imunologia , Células Dendríticas/imunologia , Linfoma/imunologia , Animais , Proliferação de Células , Endocitose , Fluoresceínas , Imunidade/imunologia , Linfoma/patologia , Camundongos , Camundongos Endogâmicos BALB C , Microscopia Confocal , Succinimidas , Linfócitos T/citologia , Linfócitos T/imunologia
20.
Cancer Immunol Immunother ; 55(5): 547-57, 2006 May.
Artigo em Inglês | MEDLINE | ID: mdl-16044254

RESUMO

The modulated expression of MHC class I on tumour tissue is well documented. Although the effect of MHC class I expression on the tumorigenicity and immunogenicity of MHC class I negative tumour cell lines has been rigorously studied, less is known about the validity of gene transfer and selection in cell lines with a mixed MHC class I phenotype. To address this issue we identified a C26 cell subline that consists of distinct populations of MHC class I (H-2D/K) positive and negative cells. Transient transfection experiments using liposome-based transfer showed a lower transgene expression in MHC class I negative cells. In addition, MHC class I negative cells were more sensitive to antibiotic selection. This led to the generation of fully MHC class I positive cell lines. In contrast to C26 cells, all transfectants were rejected in vivo and induced protection against the parental tumour cells in rechallenge experiments. Tumour cell specificity of the immune response was demonstrated in in vitro cytokine secretion and cytotoxicity assays. Transfectants expressing CD40 ligand and hygromycin phosphotransferase were not more immunogenic than cells expressing hygromycin resistance alone. We suggest that the MHC class I positive phenotype of the C26 transfectants had a bearing on their immunogenicity, because selected MHC class I positive cells were more immunogenic than parental C26 cells and could induce specific anti-tumour immune responses. These data demonstrate that the generation of tumour cell transfectants can lead to the selection of subpopulations that show an altered phenotype compared to the parental cell line and display altered immunogenicity independent of selection marker genes or other immune modulatory genes. Our results show the importance of monitoring gene transfer in the whole tumour cell population, especially for the evaluation of in vivo therapies targeted to heterogeneous tumour cell populations.


Assuntos
Expressão Gênica , Técnicas de Transferência de Genes , Terapia Genética , Antígenos de Histocompatibilidade Classe I/imunologia , Neoplasias Experimentais/imunologia , Animais , Linhagem Celular Tumoral , Ensaio de Imunoadsorção Enzimática , Citometria de Fluxo , Antígenos de Histocompatibilidade Classe I/genética , Camundongos , Camundongos Endogâmicos BALB C , Camundongos SCID , Transplante de Neoplasias , Transdução Genética , Transfecção
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA