Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 108
Filtrar
1.
Osteoarthritis Cartilage ; 27(12): 1831-1840, 2019 12.
Artigo em Inglês | MEDLINE | ID: mdl-31536814

RESUMO

OBJECTIVE: Synovium contains multipotent progenitor/stromal cells (MPCs) with potential to participate in cartilage repair. Understanding the identity of these MPCs will allow their therapeutic potential to be fully exploited. Hence this study aimed to identify primary synovial MPCs and characterize them in the context of cartilage regeneration. METHODS: Primary MPC/MPC-subset specific markers in synovium were identified by FACS analysis of uncultured cells. MPC-subsets from human synovium obtained from patients undergoing total knee arthroplasty were FACS sorted, cultured, immunophenotyped and chondrogenically differentiated. The anatomical localization of MPCs in synovium was examined using immunohistochemistry. Finally, the presence of these MPC subsets in healthy synovium obtained from human organ donors was examined. RESULTS: A combination of CD45, CD31, CD73 and CD90 can isolate two distinct MPC-subsets in synovium. These MPC-subsets, freshly isolated from synovium, did not express CD45 or CD31, but expressed CD73. Additionally, a sub-population of CD73+ cells also expressed CD90. CD45-CD31-CD73+CD90- cells were significantly more chondrogenic than CD45-CD31-CD73+CD90+ cells in the presence of TGFß1. Interestingly, reduced chondrogenic ability of CD73+CD90+ cells could be reversed by the addition of BMP2, showing discrete chondrogenic factor requirements by distinct cell-subsets. In addition, these MPCs had distinct anatomical localization; CD73 was expressed both in intimal and sub-intimal region while CD90 was enriched in the sub-intimal region. We further demonstrated that these subsets are also present in healthy synovium. CONCLUSIONS: We provide indications that primary MPCs in synovial intima and sub-intima are phenotypically and functionally distinct with different chondrogenic properties.


Assuntos
Cartilagem Articular/fisiologia , Diferenciação Celular/fisiologia , Condrogênese/fisiologia , Células-Tronco Multipotentes/metabolismo , Osteoartrite do Joelho , Regeneração/fisiologia , 5'-Nucleotidase/metabolismo , Idoso , Idoso de 80 Anos ou mais , Estudos de Casos e Controles , Moléculas de Adesão Celular/metabolismo , Feminino , Citometria de Fluxo , Proteínas Ligadas por GPI/metabolismo , Humanos , Imuno-Histoquímica , Imunofenotipagem , Antígenos Comuns de Leucócito/metabolismo , Masculino , Pessoa de Meia-Idade , Molécula-1 de Adesão Celular Endotelial a Plaquetas/metabolismo , Receptores de Quimiocinas/metabolismo , Receptores de Fatores de Crescimento/metabolismo , Membrana Sinovial/citologia , Antígenos Thy-1/metabolismo
2.
J Cell Mol Med ; 16(8): 1827-39, 2012 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-21973026

RESUMO

Although mesenchymal stromal cells (MSCs) have been applied clinically to treat cardiac diseases, it is unclear how and to which extent transplanted MSCs exert their beneficial effects. To address these questions, pre-clinical MSC administrations are needed for which pigs appear to be the species of choice. This requires the use of porcine cells to prevent immune rejection. However, it is currently unknown to what extent porcine MSCs (pMSCs) resemble human MSCs (hMSCs). Aim of this study was to compare MSC from porcine bone marrow (BM) with human cells for phenotype, multi-lineage differentiation potential, immune-modulatory capacity and the effect on cardiac function after transplantation in a mouse model of myocardial infarction. Flow cytometric analysis revealed that pMSC expressed surface antigens also found on hMSC, including CD90, MSCA-1 (TNAP/W8B2 antigen), CD44, CD29 and SLA class I. Clonogenic outgrowth was significantly enriched following selection of CD271+ cells from BM of human and pig (129 ± 29 and 1961 ± 485 fold, respectively). hMSC and pMSC differentiated comparably into the adipogenic, osteogenic or chondrogenic lineages, although pMSC formed fat much faster than hMSC. Immuno-modulation, an important feature of hMSC, was clearly demonstrated for pMSC when co-cultured with porcine peripheral blood cells stimulated with PMA and pIL-2. Finally, pMSC transplantation after myocardial infarction attenuated adverse remodelling to a similar extent as hMSC when compared to control saline injection. These findings demonstrate that pMSCs have comparable characteristics and functionality with hMSCs, making reliable extrapolation of pre-clinical pMSC studies into a clinical setting very well possible.


Assuntos
Diferenciação Celular , Imunomodulação/imunologia , Transplante de Células-Tronco Mesenquimais , Células-Tronco Mesenquimais/citologia , Miocárdio/patologia , Adipócitos/citologia , Adipócitos/metabolismo , Adipogenia , Animais , Antígenos CD/metabolismo , Células da Medula Óssea/citologia , Proliferação de Células , Separação Celular , Condrogênese , Citometria de Fluxo , Testes de Função Cardíaca , Humanos , Imunofenotipagem , Masculino , Camundongos , Camundongos SCID , Osteoblastos/citologia , Osteoblastos/metabolismo , Osteogênese , Fenótipo , Sus scrofa , Linfócitos T/citologia , Linfócitos T/metabolismo
3.
J Exp Med ; 167(5): 1645-58, 1988 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-2896757

RESUMO

We have shown in a murine model system for cytomegalovirus (CMV) disease in the immunocompromised host that CMV infection interferes with the earliest detectable step in hemopoiesis, the generation of the stem cell CFU-S-I, and thereby prevents the autoreconstitution of bone marrow after sublethal irradiation. The antihemopoietic effect could not be ascribed to a direct infection of stem cells. The failure in hemopoiesis was prevented by adoptive transfer of antiviral CD8+ T lymphocytes and could be overcome by syngeneic bone marrow transplantation. CD8+ T lymphocytes and bone marrow cells both mediated survival, although only CD8+ T lymphocytes were able to limit virus multiplication in host tissues. We concluded that not the cytopathic effect of virus replication in host tissues, but the failure in hemopoiesis, is the primary cause of death in murine CMV disease.


Assuntos
Infecções por Citomegalovirus/patologia , Hematopoese , Células-Tronco Hematopoéticas/patologia , Síndromes de Imunodeficiência/complicações , Animais , Transplante de Medula Óssea , Diferenciação Celular , Citomegalovirus/fisiologia , Infecções por Citomegalovirus/complicações , Feminino , Transplante de Células-Tronco Hematopoéticas , Imunização Passiva , Camundongos , Camundongos Endogâmicos BALB C , Quimera por Radiação , Linfócitos T Reguladores/imunologia , Linfócitos T Reguladores/transplante , Replicação Viral
4.
EBioMedicine ; 54: 102678, 2020 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-32272438

RESUMO

BACKGROUND: Overriding the differentiation blockage in acute myeloid leukemia (AML) is the most successful mode-of-action in leukemia therapy - now curing the vast majority of patients with acute promyelocytic leukemia (APL) using all-trans retinoic acid (ATRA)-based regimens. Similar approaches in other leukemia subtypes, such as IDH1/2-mutated AML, are under active investigation. We herein present successful release of the differentiation blockage upon treatment with the natural (-)-Δ9-Tetrahydrocannabinol isomer dronabinol in vitro and in vivo. METHODS: Cellular maturation and differentiation were followed in two patients employing whole genome methylation profiling, proteome analyses, NGS deep sequencing and multispectral imaging flow cytometry. For functional studies lentiviral OGT knock-down in vitro and ex vivo cell models were created to evaluate proliferative, apoptotic and differentiating effects of OGT in acute leukemia. FINDINGS: In here, we provide molecular evidence that dronbinol is capable to override the differentiation blockage of acute leukemia blasts at the state of the leukemia-initiating clone. We further identify the O-linked ß-N-acetyl glucosamine (O-GlcNAc) transferase (OGT) to be crucial in this process. OGT is a master regulator enzyme adding O-GlcNAc to serine or threonine residues in a multitude of target proteins. Aberrant O-GlcNAc modification is implicated in pathologies of metabolic, neurodegenerative and autoimme diseases as well as cancers. We provide evidence that dronabinol induces transcription of OGT via epigenetic hypomethylation of the transcription start site (TSS). A lentiviral OGT-knock out approach proves the central role of OGT exerting antileukemic efficacy via a dual-mechanism of action: High concentrations of dronabinol result in induction of apoptosis, whereas lower concentrations drive cellular maturation. Most intriguingly, overriding of the differentiation blockage of acute leukemia blasts is validated in vivo following two patients treated with dronabinol. INTERPRETATION: In conclusion, we provide evidence for overcoming the differentiation blockage in acute leukemia in subentities beyond promyelocytic and IDH1/2-mutated leukemia and thereby identify O-GlcNAcylation as a novel (drugable) field for future leukemia research. FUNDING: Unrestricted grant support by the IZKF Program of the Medical Faculty Tübingen (MMS) and Brigitte Schlieben-Lange Program as well as the Margarete von Wrangell Program of the Ministry of Science, Research and the Arts, Baden-Württemberg, Germany (KKS) and Athene Program of the excellence initiative University of Tübingen (KKS).


Assuntos
Epigênese Genética , Hematopoese , Leucemia Promielocítica Aguda/genética , N-Acetilglucosaminiltransferases/genética , Apoptose , Células Cultivadas , Metilação de DNA , Dronabinol/uso terapêutico , Reposicionamento de Medicamentos , Humanos , Isocitrato Desidrogenase/genética , Células Jurkat , Leucemia Promielocítica Aguda/sangue , Leucemia Promielocítica Aguda/tratamento farmacológico , Masculino , N-Acetilglucosaminiltransferases/metabolismo , Psicotrópicos/uso terapêutico , Sítio de Iniciação de Transcrição , Adulto Jovem
5.
Cytotherapy ; 11(2): 245-55, 2009.
Artigo em Inglês | MEDLINE | ID: mdl-19152153

RESUMO

From 4 to 5 April 2008, international experts met for the second time in Tubingen, Germany, to present and discuss the latest proceedings in research on non-hematopoietic stem cells (NHSC). This report presents issues of basic research including characterization, isolation, good manufacturing practice (GMP)-like production and imaging as well as clinical applications focusing on the regenerative and immunomodulatory capacities of NHSC.


Assuntos
Células-Tronco Adultas/citologia , Pesquisa Biomédica , Células-Tronco Embrionárias/citologia , Imunoterapia Adotiva , Neoplasias/terapia , Células-Tronco Adultas/fisiologia , Pesquisa Biomédica/ética , Pesquisa Biomédica/métodos , Pesquisa Biomédica/tendências , Técnicas de Cultura de Células , Diferenciação Celular , Movimento Celular , Transdiferenciação Celular , Diagnóstico por Imagem , Células-Tronco Embrionárias/fisiologia , Perfilação da Expressão Gênica , Alemanha , Mobilização de Células-Tronco Hematopoéticas , Humanos , Medicina Regenerativa/tendências , Nicho de Células-Tronco
6.
J Cell Biol ; 131(1): 243-9, 1995 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-7559781

RESUMO

Differentiation and proliferation of hematopoietic progenitors take place in the bone marrow and is a tightly controlled process. Cell adhesion molecules of the integrin and immunoglobulin families have been shown to be involved in these processes, but almost nothing was known about the involvement of the cadherin family in the hematopoietic system. A PCR screening of RNA of human bone marrow mononuclear cells with specific primers for classical cadherins revealed that E-cadherin, which is mainly expressed by cells of epithelial origin, is also expressed by bone marrow cells. Western blot analysis and immunofluorescence staining of bone marrow sections confirmed this unexpected finding. A more detailed analysis using immunoaffinity columns and dual color flow cytometry showed that the expression of E-cadherin is restricted to defined maturation stages of the erythropoietic lineage. Erythroblasts and normoblasts express E-cadherin, mature erythrocytes do not. A functional role of E-cadherin in the differentiation process of the erythroid lineage was indicated by antibody-inhibition studies. The addition of anti-E-cadherin antibody to bone marrow mononuclear cultures containing exogeneous erythropoietin drastically diminished the formation of erythropoietic cells. These data suggest a non-anticipated expression and function of E-cadherin in one defined hematopoietic cell lineage.


Assuntos
Caderinas/fisiologia , Células Precursoras Eritroides/fisiologia , Sequência de Bases , Células da Medula Óssea , Diferenciação Celular/fisiologia , Separação Celular , Células Cultivadas/fisiologia , Primers do DNA , Citometria de Fluxo , Humanos , Dados de Sequência Molecular , Monócitos/fisiologia , Reação em Cadeia da Polimerase , RNA Mensageiro/análise
7.
J Cell Biol ; 108(2): 737-46, 1989 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-2645304

RESUMO

In the mammalian host, the unicellular flagellate Trypanosoma brucei is covered by a dense surface coat that consists of a single species of macromolecule, the membrane form of the variant surface glycoprotein (mfVSG). After uptake by the insect vector, the tsetse fly, bloodstream-form trypanosomes differentiate to procyclic forms in the fly midgut. Differentiation is characterized by the loss of the mfVSG coat and the acquisition of a new surface glycoprotein, procyclin. In this study, the change in surface glycoprotein composition during differentiation was investigated in vitro. After triggering differentiation, a rapid increase in procyclin-specific mRNA was observed. In contrast, there was a lag of several hours before procyclin could be detected. Procyclin was incorporated and uniformly distributed in the surface coat. The VSG coat was subsequently shed. For a single cell, it took 12-16 h to express a maximum level of procyclin at the surface while the loss of the VSG coat required approximately 4 h. The data are discussed in terms of the possible molecular arrangement of mfVSG and procyclin at the cell surface. Molecular modeling data suggest that a (Asp-Pro)2 (Glu-Pro)22-29 repeat in procyclin assumes a cylindrical shape 14-18 nm in length and 0.9 nm in diameter. This extended shape would enable procyclin to interdigitate between the mfVSG molecules during differentiation, exposing epitopes beyond the 12-15-nm-thick VSG coat.


Assuntos
Regulação da Expressão Gênica , Trypanosoma brucei brucei/crescimento & desenvolvimento , Glicoproteínas Variantes de Superfície de Trypanosoma/metabolismo , Sequência de Aminoácidos , Animais , Diferenciação Celular , Sondas de DNA , Eletroforese em Gel de Poliacrilamida , Citometria de Fluxo , Imunofluorescência , Cinética , Microscopia Eletrônica , Dados de Sequência Molecular , Estrutura Molecular , Hibridização de Ácido Nucleico , Conformação Proteica , RNA Mensageiro/biossíntese , Trypanosoma brucei brucei/genética , Trypanosoma brucei brucei/metabolismo
8.
Int J Immunopathol Pharmacol ; 21(1): 11-21, 2008.
Artigo em Inglês | MEDLINE | ID: mdl-18336727

RESUMO

IgE-dependent activation of basophils is associated with upregulation of several surface molecules. We recently identified the surface enzyme aminopeptidase N (CD13) as a novel activation antigen on human basophils. In the present study, we asked whether CD13 can be employed as a novel marker of allergen-induced activation of basophils in allergic individuals. Patients allergic to major allergens from grass pollen (Phl p 1, Phl p 5), birch pollen (Bet v 1), or house dust mites (Der p 2), were examined. Blood basophils were exposed to various concentrations of recombinant allergens for 15 minutes, and examined for expression of CD13 by multicolor flow cytometry. The allergen-induced upregulation of CD13 was compared with allergen-dependent increases in expression of CD63 and CD203c. Exposure to recombinant allergens resulted in an increase in expression of CD13 on basophils in all sensitized individuals, whereas no increase in CD13 was seen in healthy controls. The effects of the recombinant allergens on CD13-expression were dose- and time-dependent, were not observed in the absence of extracellular calcium, and were counteracted by preincubation of basophils with the PI3-kinase-targeting drugs staurosporin and LY294002. There was a good correlation between allergen-induced upregulation of CD13, CD63, and CD203c on basophils. In aggregate, our data show that recombinant allergens promote expression of CD13 on basophils in sensitized individuals. The functional significance and diagnostic implications of this observation remain to be determined.


Assuntos
Alérgenos/imunologia , Basófilos/imunologia , Antígenos CD13/sangue , Hipersensibilidade/imunologia , Adolescente , Adulto , Antígenos CD/sangue , Basófilos/enzimologia , Basófilos/fisiologia , Cálcio/fisiologia , Feminino , Humanos , Interleucina-3/farmacologia , Masculino , Pessoa de Meia-Idade , Fosfatidilinositol 3-Quinases/fisiologia , Diester Fosfórico Hidrolases/sangue , Glicoproteínas da Membrana de Plaquetas , Pirofosfatases/sangue , Proteínas Recombinantes/imunologia , Tetraspanina 30
9.
Int J Immunopathol Pharmacol ; 21(4): 797-806, 2008.
Artigo em Inglês | MEDLINE | ID: mdl-19144265

RESUMO

The ectoenzyme E-NPP3 (CD203c) has recently been identified as a novel activation-linked cell surface antigen on basophils. In the present study, we examined expression of CD203c on normal mast cells (MC)and bone marrow (bm) MC derived from 85 patients with systemic mastocytosis (SM), including cases with indolent SM (ISM, n=72), SM with associated clonal hematologic non-MC-lineage disease (SM-AHNMD, n=6), aggressive SM (ASM, n=3), and mast cell leukemia (MCL, n=4). Surface expression of CD203c was analyzed by multicolor flow cytometry. In patients with SM, bm MC expressed significantly higher amounts of CD203c compared to normal bm MC (median MFI in controls: 260 versus median MFI in SM: 516, p<0.05). Slightly lower amounts of CD203c were detected on MC in SM-AHNMD and ASM compared to ISM. To demonstrate CD203c expression in MC at the mRNA level, neoplastic MC were highly enriched by cell sorting, and were found to express CD203c mRNA in RT-PCR analysis. Cross-linking of the IgE receptor on MC resulted in a substantial upregulation of CD203c, whereas the KIT-ligand stem cell factor (SCF) showed no significant effects. In conclusion, CD203c is a novel activation-linked surface antigen on MC that is upregulated in response to IgE receptor cross-linking and is overexpressed on neoplastic MC in patients with SM.


Assuntos
Mastócitos/imunologia , Mastocitose/imunologia , Neoplasias/imunologia , Diester Fosfórico Hidrolases/imunologia , Pirofosfatases/imunologia , Receptores de IgE/imunologia , Regulação para Cima , Sequência de Bases , Primers do DNA , Citometria de Fluxo , Humanos , Diester Fosfórico Hidrolases/genética , Pirofosfatases/genética , RNA Mensageiro/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa
10.
Int J Immunopathol Pharmacol ; 20(2): 267-78, 2007.
Artigo em Inglês | MEDLINE | ID: mdl-17624239

RESUMO

We recently identified the ectoenzyme CD203c as a novel basophil activation antigen that is upregulated in response to FcepsilonRI cross-linkage. We investigated the effects of various interleukins (ILs) on expression of CD203c on blood basophils using an antibody against CD203c and flow cytometry. Of all cytokines tested, only IL-3 was found to upregulate expression of CD203c on basophils above baseline levels. The effects of IL-3 were dose- and time-dependent (EC(50): 0.1-1 ng/ml) without differences observed between healthy and allergic donors. Whereas anti-IgE induced maximum upregulation of CD203c within 15 minutes, the IL-3-induced upregulation showed a maximum after 180 minutes. IgE-receptor cross-linking resulted in enhanced expression of both CD63 and CD203c, whereas IL-3 enhanced the levels of CD203c without promoting expression of CD63. The IL-3-induced upregulation of CD203c was also observed in highly enriched basophils and was counteracted by a blocking antibody against the alpha chain of the IL-3 receptor (CD123). The IL-3-induced upregulation of CD203c was also found to depend on the presence of calcium. To analyze signaling pathways involved in IL-3-induced upregulation of CD203c, pharmacologic inhibitors were applied. The PI3-kinase inhibitors, wortmannin and LY294002 counteracted the IL-3-induced expression of CD203c, whereas MEK- and PKC inhibitors showed no effects. In conclusion, IL-3 upregulates expression of CD203c on basophils through a specific receptor and via a PI3-kinase-dependent signaling-pathway. Compared to FcepsilonRI-mediated cell activation, IL-3-induced upregulation of CD203c is a late(r) event and is not accompanied by upregulation of CD63.


Assuntos
Basófilos/imunologia , Basófilos/metabolismo , Betula/imunologia , Interleucina-3/fisiologia , Diester Fosfórico Hidrolases/genética , Pólen/imunologia , Pirofosfatases/genética , Rinite Alérgica Sazonal/imunologia , Antígenos CD/biossíntese , Antígenos CD/genética , Células Cultivadas , Regulação da Expressão Gênica/fisiologia , Humanos , Diester Fosfórico Hidrolases/biossíntese , Glicoproteínas da Membrana de Plaquetas/biossíntese , Glicoproteínas da Membrana de Plaquetas/genética , Pirofosfatases/biossíntese , Tetraspanina 30
11.
Leukemia ; 31(2): 459-469, 2017 02.
Artigo em Inglês | MEDLINE | ID: mdl-27435001

RESUMO

Antibody-dependent cellular cytotoxicity (ADCC) of natural killer (NK) cells largely contributes to the success of monoclonal antibody (mAb) treatment in cancer. As no antibodies are clinically available for immunotherapy of myeloid leukemias (MLs), we aimed to develop an Fc-optimized CD133 mAb for induction of NK ADCC against MLs. When comparing different available CD133 mAbs, no difference was observed with regard to binding to primary chronic myeloid leukemia cells. However, clone 293C3 recognized acute myeloid leukemia (AML) cells in a substantially higher percentage of patient cases and was thus chosen to generate chimeric mAbs with either wild-type Fc part (293C3-WT) or a variant containing amino-acid exchanges (S239D/I332E) to enhance affinity to CD16 on NK cells (293C3-SDIE). In vitro, treatment with 293C3-SDIE significantly enhanced activation, degranulation and lysis of primary CD133-positive AML cells by allogeneic and autologous NK cells as compared with its wild-type counterpart. In line with the observed lower expression levels of CD133 on healthy cells compared with malignant hematopoietic cells, 293C3-SDIE caused no relevant toxicity towards committed hematopoietic progenitor cells. In a NOD.Cg-PrkdcscidIL2rgtmWjl/Sz xenotransplantation model, 293C3-SDIE facilitated elimination of patient AML cells by human NK cells. Thus, 293C3-SDIE constitutes an attractive immunotherapeutic compound, in particular for elimination of minimal residual disease in the context of allogeneic stem cell transplantation in AML.


Assuntos
Antígeno AC133/imunologia , Anticorpos Monoclonais/imunologia , Anticorpos Monoclonais/farmacologia , Fragmentos Fc das Imunoglobulinas/imunologia , Células Matadoras Naturais/imunologia , Células Matadoras Naturais/metabolismo , Leucemia Mieloide Aguda/imunologia , Leucemia Mieloide Aguda/metabolismo , Animais , Citotoxicidade Celular Dependente de Anticorpos , Degranulação Celular/imunologia , Citocinas/metabolismo , Citotoxicidade Imunológica/imunologia , Epitopos/imunologia , Xenoenxertos , Humanos , Ativação Linfocitária/imunologia , Camundongos
12.
BMC Cancer ; 6: 195, 2006 Jul 21.
Artigo em Inglês | MEDLINE | ID: mdl-16859559

RESUMO

BACKGROUND: The chemokine stromal derived factor-1 (SDF-1 or CXCL12) and its receptor CXCR4 have been demonstrated to be crucial for the homing of stem cells and prostate cancers to the marrow. While screening prostate cancers for CXCL12-responsive adhesion molecules, we identified CD164 (MGC-24) as a potential regulator of homing. CD164 is known to function as a receptor that regulates stem cell localization to the bone marrow. RESULTS: Using prostate cancer cell lines, it was demonstrated that CXCL12 induced both the expression of CD164 mRNA and protein. Functional studies demonstrated that blocking CD164 on prostate cancer cell lines reduced the ability of these cells to adhere to human bone marrow endothelial cells, and invade into extracellular matrices. Human tissue microarrays stained for CD164 demonstrated a positive correlation with prostate-specific antigen levels, while its expression was negatively correlated with the expression of androgen receptor. CONCLUSION: Our findings suggest that CD164 may participate in the localization of prostate cancer cells to the marrow and is further evidence that tumor metastasis and hematopoietic stem cell trafficking may involve similar processes.


Assuntos
Neoplasias da Medula Óssea/secundário , Endolina/metabolismo , Metástase Neoplásica/fisiopatologia , Neoplasias da Próstata/patologia , Neoplasias da Medula Óssea/fisiopatologia , Adesão Celular , Quimiocina CXCL12 , Quimiocinas CXC/fisiologia , Perfilação da Expressão Gênica , Células-Tronco Hematopoéticas/fisiologia , Humanos , Masculino , Antígeno Prostático Específico , Células Tumorais Cultivadas
13.
Cancer Res ; 53(18): 4424-31, 1993 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-7689935

RESUMO

The function of the steel factor receptor, p145c-kit, in patient-derived acute myeloblastic leukemia (AML) cells was investigated. Steel factor stimulation of AML cells coexpressing p145c-kit and the progenitor cell antigen CD34 resulted in complete receptor down-regulation, a marked decrease of CD34 antigen expression, and the induction of the granulocyte lineage antigen CD15. These changes in surface marker expression paralleled morphological differentiation to granulated blasts and promyelocytes. Interestingly, the same phenotype was achieved by IL-3 stimulation of AML cells. p145c-kit extracellular domain-specific antibodies had either blocking or enhancing effects on ligand binding, receptor phosphorylation and down-regulation, and induction of cell proliferation. Correlations of these phenomena with distinct effects of antibody stimulation on cell substrate phosphorylation provide clues for the dissection of the p145c-kit signal and the analysis of its relevance for AML.


Assuntos
Leucemia Mieloide Aguda/patologia , Proteínas Proto-Oncogênicas/fisiologia , Animais , Anticorpos Monoclonais/imunologia , Diferenciação Celular , Regulação para Baixo , Fatores de Crescimento de Células Hematopoéticas/fisiologia , Humanos , Interleucina-3/farmacologia , Leucemia Mieloide Aguda/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Peso Molecular , Proteínas Proto-Oncogênicas/análise , Proteínas Proto-Oncogênicas c-kit , Fator de Células-Tronco
14.
Cancer Res ; 57(4): 696-701, 1997 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-9044847

RESUMO

Immunotherapies directed to the proto-oncogene product HER-2/neu, which is overexpressed on a subset of breast and other carcinomas, currently receive considerable attention. We have investigated cell-mediated effector mechanisms of HER-2/neu antibodies against breast cancer cell lines. Compared to unfractionated control blood, whole blood from patients during granulocyte colony-stimulating factor (G-CSF) treatment exhibits significantly enhanced lysis (P < 0.001) of SK-BR-3 cells in the presence of HER-2/neu antibody 520C9. The extent of tumor cell killing correlated positively (r = 0.74) to polymorphonuclear neutrophil (PMN) blood counts. Fractionation of whole blood into plasma, mononuclear cells, and PMNs showed major killing capacity to reside in the granulocyte fraction. PMNs were efficiently cytolytic with a panel of HER-2/neu antibodies and against various breast cancer cell lines. Experiments with blocking antibodies to Fc(gamma)R documented Fc(gamma)RII (CD32) as the major trigger molecule for monoclonal antibody 502C9-mediated cytotoxicity. Killing via 520C9 was significantly influenced by an allotypic polymorphism of Fc(gamma)RIIa, the CD32 molecule expressed on PMNs. In reverse antibody-dependent cell-mediated cytotoxicity experiments with a panel of HER-2/neu-directed bispecific antibodies, Fc(gamma)RIII (CD16) proved to be an efficient trigger molecule in blood from healthy volunteers. During G-CSF treatment, however, Fc(gamma)RI (CD64)-expressed on monocytes and G-CSF primed, but not on healthy donor PMNs-became the predominant cytotoxic trigger molecule. Thus, G-CSF application increased effector cell numbers for HER-2/neu-directed immunotherapy, and G-CSF primed PMNs proved particularly effective with a [HER-2/neu x Fc(gamma)RI] bispecific antibody. These findings support clinical trials with HER-2/neu-directed antibodies in combination with G-CSF in breast cancer patients overexpressing HER-2/neu.


Assuntos
Anticorpos Biespecíficos/uso terapêutico , Neoplasias da Mama/terapia , Fator Estimulador de Colônias de Granulócitos/uso terapêutico , Granulócitos/imunologia , Imunoterapia/métodos , Receptor ErbB-2/imunologia , Receptores de IgG/imunologia , Citotoxicidade Celular Dependente de Anticorpos/imunologia , Neoplasias da Mama/imunologia , Neoplasias da Mama/metabolismo , Feminino , Humanos , Imunidade Celular , Proto-Oncogene Mas , Receptor ErbB-2/metabolismo , Células Tumorais Cultivadas
15.
Cancer Res ; 61(18): 6846-50, 2001 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-11559560

RESUMO

The epithelial mucin MUC1 is overexpressed on the cell surface of many epithelial malignancies as well as on some B-cell lymphomas and multiple myelomas. Recently, we identified two HLA-A2-restricted T-cell epitopes derived from the MUC1 protein. To further extend the potential application of these peptides, we analyzed the expression of MUC1 on blast cells from patients with acute myelogenous leukemia (AML; n = 43) and several other hematological malignancies including acute lymphoblastic leukemia (n = 24), chronic lymphocytic leukemia (n = 36), hairy cell leukemia (n = 9), follicular lymphoma (n = 7), and multiple myeloma (n = 12). Using reverse transcription-PCR and MUC1-specific monoclonal antibodies, MUC1 expression was found in 67% of AML samples and 92% of myeloma samples. To analyze the presentation of MUC1 peptides by primary AML blasts, we induced MUC1-specific CTLs in vitro using peptide-pulsed dendritic cells from HLA-A2+ healthy donors as antigen-presenting cells. These CTLs efficiently lysed in an antigen-specific and HLA-A2-restricted manner not only target cells pulsed with the antigenic peptide but also tumor cell lines including multiple myeloma cells and primary AML blasts that constitutively expressed both MUC1 and HLA-A2. The specificity of the CTLs was confirmed in a cold target inhibition assay. Our data demonstrate that MUC1-derived peptides are tumor antigens in AML and several other hematological malignancies that could potentially be used for immunotherapeutic approaches.


Assuntos
Neoplasias Hematológicas/imunologia , Mucina-1/imunologia , Linfócitos T Citotóxicos/imunologia , Anticorpos Monoclonais/imunologia , Especificidade de Anticorpos , Apresentação de Antígeno/imunologia , Células Dendríticas/imunologia , Epitopos de Linfócito T/imunologia , Antígeno HLA-A2/imunologia , Humanos , Leucemia Mieloide Aguda/imunologia , Ativação Linfocitária/imunologia , Mucina-1/biossíntese , Mieloma Múltiplo/imunologia
16.
Biochim Biophys Acta ; 733(1): 117-23, 1983 Aug 24.
Artigo em Inglês | MEDLINE | ID: mdl-6882750

RESUMO

We have investigated the membrane-damaging effect of phallolysin on liposomes varying in phospholipid composition, net charge and physical constitution. Liposomes were prepared from lipids extracted from bovine or human erythrocyte ghosts. The liposomes composed of bovine lipids (the intact cell showing little sensitivity to phallolysin) were found comparably sensitive to those prepared from lipids of human red cells (these cells being of high sensitivity). In addition, artificial mixtures of lipids were used for the preparation of liposomes, consisting of (a) negatively charged phospholipids such as dicetyl phosphate or phosphatidylserine, (b) cholesterol, and (c) either sphingomyelin (as the major component of erythrocytes from ruminants) or phosphatidylcholine (as the major component of erythrocytes from non-ruminants). Again, we found only little difference in the susceptibilities of sphingomyelin- and phosphatidylcholine-containing liposomes. On the other hand, the susceptibility depended on the presence of phospholipids with negative net charges. Omittance of phosphatidylcholine or dicetyl phosphate, or replacement by the positively charged stearylamine, decreased the susceptibility by a factor of more than 20. Finally, we prepared liposomes from dicetyl phosphate, cholesterol and phosphatidylcholine in two physical states: large unilamellar and smaller multilamellar liposomes. The unilamellar liposomes were about 10-times more sensitive to phallolysin. We conclude: (1) Phallolysin damages phospholipid-membranes in the absence of receptor proteins, but high concentrations of the toxin are required. (2) Membrane damage takes place with liposomes containing phosphatidylcholine as well as those containing sphingomyelin. (3) Phallolysin damages only liposomes containing phospholipids with a negative net charge.


Assuntos
Amanitinas/toxicidade , Citotoxinas/toxicidade , Lipossomos , Animais , Bovinos , Membrana Eritrocítica/efeitos dos fármacos , Membrana Eritrocítica/ultraestrutura , Humanos , Cinética , Lipídeos de Membrana/sangue , Microscopia Eletrônica , Modelos Biológicos , Fosfolipídeos/sangue , Relação Estrutura-Atividade
17.
J Clin Oncol ; 17(5): 1535-44, 1999 May.
Artigo em Inglês | MEDLINE | ID: mdl-10334541

RESUMO

PURPOSE: The expression of the carcinoma-associated mucin MUC-1 is thought to be restricted to epithelial cells and is used for micrometastatic tumor cell detection in patients with solid tumors, including those with breast cancer. Little is known, however, about the expression of MUC-1 epitopes in normal hematopoietic cells. MATERIALS AND METHODS: MUC-1 expression was analyzed by flow cytometry and immunocytology on bone marrow (BM) mononuclear cells and purified CD34+ cells from healthy volunteers, using different anti-MUC-1-specific monoclonal antibodies. In addition, Western blotting of MUC-1 proteins was performed. RESULTS: Surprisingly, 2% to 10% of normal human BM mononuclear cells expressed MUC-1, as defined by the anti-MUC-1 antibodies BM-2 (2E11), BM-7, 12H12, MAM-6, and HMFG-1. In contrast, two antibodies recognizing the BM-8 and the HMFG-2 epitopes of MUC-1 were not detected. MUC-1+ cells from normal BM consisted primarily of erythroblasts and normoblasts. In agreement with this, normal CD34+ cells cultured in vitro to differentiate into the erythroid lineage showed a strong MUC-1 expression on day 7 proerythroblasts. Western blotting of these cells confirmed that the reactive species is the known high molecular weight MUC-1 protein. CONCLUSION: Our data demonstrate that some MUC-1 epitopes are expressed on normal BM cells and particularly on cells of the erythroid lineage. Hence the application of anti-MUC-1 antibodies for disseminated tumor cell detection in BM or peripheral blood progenitor cells may provide false-positive results, and only carefully evaluated anti-MUC-1 antibodies (eg, HMFG-2) might be selected. Furthermore, MUC-1-targeted immunotherapy in cancer patients might be hampered by the suppression of erythropoiesis.


Assuntos
Medula Óssea/imunologia , Epitopos/análise , Células-Tronco Hematopoéticas/imunologia , Mucina-1/imunologia , Anticorpos Monoclonais , Citometria de Fluxo , Humanos
18.
Leukemia ; 5(10): 854-60, 1991 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-1720490

RESUMO

A monoclonal antibody (17F11) was raised by immunization of a Balb/c mouse with leukemic blasts from a patient with acute non-lymphocytic leukemia (ANLL). This antibody recognizes most leukemic blasts of myeloid but not of lymphoid lineage and no peripheral blood cells. By screening NIH-3T3 fibroblasts transfected with the human proto-oncogene c-kit (NIH-3T3/hckit) it could be shown that 17F11 specifically recognizes the gene product P145c-kit. Immunofluorescence analysis on normal hemopoietic cells revealed that 17F11 weakly stains 1-3% of bone marrow mononuclear cells (BMMNC). By FACS sorting and colony assays it could be shown that granulocyte--macrophage progenitor cells could be enriched 10-20-fold, granulocyte progenitors 50-80-fold, and erythroid and multipotential progenitor cells 15-20-fold, in the 17F11 positive fraction. Double fluorescence analysis revealed that P145c-kit is co-expressed on 40-60% of the CD34 positive BMMNC. Finally, these data show that P145c-kit is expressed on blast cells from most patients with ANLL (26/30) and chronic myeloid leukemia in blast crisis (7/9), but is absent on blasts from patients with acute lymphoblastic leukemia expressing the T-, B-lineage, or common ALL phenotypes.


Assuntos
Antígenos de Neoplasias/genética , Antígenos de Superfície/genética , Células-Tronco Hematopoéticas/imunologia , Leucemia Mieloide Aguda/genética , Proteínas Proto-Oncogênicas/genética , Proto-Oncogenes/genética , Animais , Anticorpos Monoclonais/imunologia , Antígenos CD/fisiologia , Antígenos CD34 , Antígenos de Neoplasias/imunologia , Antígenos de Superfície/imunologia , Medula Óssea/imunologia , Medula Óssea/fisiologia , Células da Medula Óssea , Células Cultivadas , Expressão Gênica , Células-Tronco Hematopoéticas/fisiologia , Humanos , Leucemia Mieloide Aguda/imunologia , Leucemia Mieloide Aguda/patologia , Camundongos , Camundongos Endogâmicos BALB C , Proto-Oncogene Mas , Proteínas Proto-Oncogênicas c-kit , Células Tumorais Cultivadas
19.
Leukemia ; 8(2): 258-63, 1994 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-7508533

RESUMO

Antigenic profiles in AML that have generally accepted prognostic significance, and allow treatment stratification, have not yet been defined. In a previous report of Ashman et al., the proto-oncogene c-kit defined by binding of the moab YB5.B8 was expressed on about one third of AML cases, mainly of the undifferentiated FAB-subtypes and associated with poor prognosis and overall survival. In this study, the moab 17F11 also directed against the c-kit structure stained 41/47 AML and 6/8 CML blast specimens, whereas all investigated 40 ALL samples were c-kit negative. c-kit was not restricted to any particular, undifferentiated FAB-subtype, but found in 9/9 AML-M0/M1, 18/19 AML-M2, 0/1 AML-M3, 11/13 AML-M4 and 3/5 AML-M5 subtypes. Immunophenotypical analysis showed no restriction of c-kit expression to immature, CD34+ precursors, but c-kit was also expressed on CD4+ CD34- precursor cells differentiating towards the monocyte lineage. In addition, multi-color labelings revealed an extraordinary heterogeneity of concomitant antigen expression on c-kit+ cells 10/36 c-kit+ CD34+ samples expressing CD56 and 16/36 c-kit+ CD34+ samples being CD7 positive; two c-kit+ CD34+ specimens carried the B-cell antigen CD19. In correlation to clinical outcome c-kit expression as single parameter was not predictive for poor response to therapy and short survival as previously suggested.


Assuntos
Expressão Gênica , Leucemia Mieloide Aguda/genética , Proteínas Proto-Oncogênicas/genética , Proto-Oncogenes/genética , Receptores Proteína Tirosina Quinases/genética , Receptores de Fator Estimulador de Colônias/genética , Idoso , Anticorpos Monoclonais/imunologia , Antígenos de Neoplasias/análise , Humanos , Imunofenotipagem , Leucemia Mieloide Aguda/imunologia , Leucemia Mieloide Aguda/mortalidade , Pessoa de Meia-Idade , Leucemia-Linfoma Linfoblástico de Células Precursoras/genética , Prognóstico , Proto-Oncogene Mas , Proteínas Proto-Oncogênicas/análise , Proteínas Proto-Oncogênicas c-kit , Receptores Proteína Tirosina Quinases/análise , Receptores de Fator Estimulador de Colônias/análise , Taxa de Sobrevida
20.
Leukemia ; 9(11): 1910-20, 1995 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-7475283

RESUMO

The effect of interleukin 10 (IL-10) on proliferation and cytokine secretion by acute myelogenous leukemia (AML) blast cells was investigated in vitro. IL-10 inhibited spontaneous AML blast proliferation for a majority of patients, whereas in the presence of exogenous growth factors (granulocyte-stimulating factor, G-CSF; granulocyte-macrophage colony-stimulating factor, GM-CSF; interleukin 3) the IL-10 effect on blast proliferation showed a wide variation depending on the individual AML patient. IL-10 seemed to cause an irreversible inhibitory effect on AML blasts, as inhibition could also be demonstrated when IL-10 was present only during the initial preincubation of the leukemia cells. IL-10 also inhibited AML blast colony formation. However, independent of the effect on AML blast proliferation, IL-10 decrease cytokine secretion from AML blast cells for all patients, as demonstrated for IL-1 alpha, IL-1 beta, tumor necrosis factor-alpha, GM-CSF and interleukin 6. IL-10 did not inhibit development of apoptosis in AML blasts cultured in vitro. Expression of complement receptors and capability to adhere and internalize bacteria by AML blasts were not altered by IL-10.


Assuntos
Citocinas/metabolismo , Interleucina-10/farmacologia , Leucemia Mieloide Aguda/patologia , Idoso , Apoptose/efeitos dos fármacos , Crise Blástica/patologia , Diferenciação Celular/efeitos dos fármacos , Divisão Celular/efeitos dos fármacos , Feminino , Humanos , Imunofenotipagem , Ativação Linfocitária/efeitos dos fármacos , Masculino , Pessoa de Meia-Idade , Células Tumorais Cultivadas
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa