Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Haematologica ; 108(11): 3001-3010, 2023 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-37259567

RESUMEN

Azacitidine (Aza) combined with donor lymphocyte infusions (DLI) is an established treatment for relapse of myeloid malignancies after allogeneic transplantation. Based on its immunomodulatory and anti-leukemic properties we considered Lenalidomide (Lena) to act synergistically with Aza/DLI to improve outcome. We, therefore, prospectively investigated tolerability and efficacy of this combination as first salvage therapy for adults with post-transplant relapse of acute myeloid leukemia, myelodysplastic syndromes and chronic myelomonocytic leukemia. Patients were scheduled for eight cycles Aza (75 mg/m2 day 1-7), Lena (2.5 or 5 mg, days 1-21) and up to three DLI with increasing T-cell dosages (0.5×106-1.5×107 cells/kg). Primary endpoint was safety, while secondary endpoints included response, graft-versus-host disease (GvHD) and overall survival (OS). Fifty patients with molecular (52%) or hematological (48%) relapse of myelodysplastic syndromes (n=24), acute myeloid leukemia (n=23) or chronic myelomonocytic leukemia (n=3) received a median of seven (range, 1-8) cycles including 14 patients with 2.5 mg and 36 with 5 mg Lena daily dosage. Concomitantly, 34 patients (68%) received at least one DLI. Overall response rate was 56% and 25 patients (50%) achieved complete remission being durable in 80%. Median OS was 21 months and 1-year OS rate 65% with no impact of type of or time to relapse and Lena dosages. Treatment was well tolerated indicated by febrile neutropenia being the only grade ≥3 non-hematologic adverse event in >10% of patients and modest acute (grade 2-4 24%) and chronic (moderate/severe 28%) GvHD incidences. In summary, Lena can be safely added to Aza/DLI without excess of GvHD and toxicity. Its significant anti-leukemic activity suggests that this combination is a novel salvage option for post-transplant relapse (clinicaltrials gov. Identifier: NCT02472691).


Asunto(s)
Enfermedad Injerto contra Huésped , Trasplante de Células Madre Hematopoyéticas , Leucemia Mieloide Aguda , Leucemia Mielomonocítica Crónica , Síndromes Mielodisplásicos , Adulto , Humanos , Azacitidina/uso terapéutico , Lenalidomida , Leucemia Mielomonocítica Crónica/terapia , Leucemia Mielomonocítica Crónica/complicaciones , Transfusión de Linfocitos/efectos adversos , Síndromes Mielodisplásicos/patología , Trasplante Homólogo/efectos adversos , Enfermedad Crónica , Enfermedad Injerto contra Huésped/diagnóstico , Enfermedad Injerto contra Huésped/etiología , Enfermedad Injerto contra Huésped/tratamiento farmacológico , Linfocitos T/patología , Recurrencia , Trasplante de Células Madre Hematopoyéticas/efectos adversos
2.
Stem Cells ; 39(9): 1270-1284, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-34013984

RESUMEN

Acute myeloid leukemia (AML) is characterized by an expansion of leukemic cells and a simultaneous reduction of normal hematopoietic precursors in the bone marrow (BM) resulting in hematopoietic insufficiency, but the underlying mechanisms are poorly understood in humans. Assuming that leukemic cells functionally inhibit healthy CD34+ hematopoietic stem and progenitor cells (HSPC) via humoral factors, we exposed healthy BM-derived CD34+ HSPC to cell-free supernatants derived from AML cell lines as well as from 24 newly diagnosed AML patients. Exposure to AML-derived supernatants significantly inhibited proliferation, cell cycling, colony formation, and differentiation of healthy CD34+ HSPC. RNA sequencing of healthy CD34+ HSPC after exposure to leukemic conditions revealed a specific signature of genes related to proliferation, cell-cycle regulation, and differentiation, thereby reflecting their functional inhibition on a molecular level. Experiments with paired patient samples showed that these inhibitory effects are markedly related to the immunomagnetically enriched CD34+ leukemic cell population. Using PCR, ELISA, and RNA sequencing, we detected overexpression of TGFß1 in leukemic cells on the transcriptional and protein level and, correspondingly, a molecular signature related to TGFß1 signaling in healthy CD34+ HSPC. This inhibitory effect of TGFß1 on healthy hematopoiesis was functionally corrobated and could be pharmacologically reverted by SD208, an inhibitor of TGFß receptor 1 signaling. Overall, these data indicate that leukemic cells induce functional inhibition of healthy CD34+ HSPC, at least in part, through TGFß1, suggesting that blockage of this pathway may improve hematopoiesis in AML.


Asunto(s)
Células Madre Hematopoyéticas , Leucemia Mieloide Aguda , Antígenos CD34/metabolismo , Médula Ósea/metabolismo , Hematopoyesis , Células Madre Hematopoyéticas/metabolismo , Humanos , Leucemia Mieloide Aguda/genética
3.
Mar Drugs ; 17(9)2019 Sep 05.
Artículo en Inglés | MEDLINE | ID: mdl-31491907

RESUMEN

There is a variety of antineoplastic drugs that are based on natural compounds from ecological niches with high evolutionary pressure. We used two cell lines (Jurkat J16 and Ramos) in a screening to assess 300 different naturally occurring compounds with regard to their antineoplastic activity. The results of the compounds 4,6-dibromo-2-(2',4'-dibromophenoxy)phenol (P01F03), 4,5,6-tribromo-2-(2',4'-dibromophenoxy)phenol (P01F08), and 5-epi-nakijinone Q (P03F03) prompted us to perform further research. Using viability and apoptosis assays on the cell lines of primary human leukemic and normal hematopoietic cells, we found that P01F08 induced apoptosis in the cell lines at IC50 values between 1.61 and 2.95 µM after 72 h. IC50 values of peripheral blood mononuclear cells (PBMNCs) from healthy donors were higher, demonstrating that the cytotoxicity in the cell lines reached 50%, while normal PBMNCs were hardly affected. The colony-forming unit assay showed that the hematopoietic progenitor cells were not significantly affected in their growth by P01F08 at a concentration of 3 µM. P01F08 showed a 3.2-fold lower IC50 value in primary leukemic cells [acute myeloid leukemia (AML)] compared to the PBMNC of healthy donors. We could confirm the antineoplastic effect of 5-epi-nakijinone Q (P03F03) on the cell lines via the induction of apoptosis but noted a similarly strong cytotoxic effect on normal PBMNCs.


Asunto(s)
Antineoplásicos/uso terapéutico , Fenol/uso terapéutico , Adulto , Anciano , Apoptosis/efectos de los fármacos , Línea Celular Tumoral , Femenino , Células HL-60 , Células Madre Hematopoyéticas/efectos de los fármacos , Humanos , Células Jurkat , Leucemia Mieloide Aguda/tratamiento farmacológico , Leucocitos Mononucleares/efectos de los fármacos , Masculino , Persona de Mediana Edad , Células THP-1
4.
Sci Rep ; 9(1): 12028, 2019 08 19.
Artículo en Inglés | MEDLINE | ID: mdl-31427693

RESUMEN

Graphene quantum dots (GQDs) are a promising next generation nanomaterial with manifold biomedical applications. For real world applications, comprehensive studies on their influence on the functionality of primary human cells are mandatory. Here, we report the effects of GQDs on the transcriptome of CD34+ hematopoietic stem cells after an incubation time of 36 hours. Of the 20 800 recorded gene expressions, only one, namely the selenoprotein W, 1, is changed by the GQDs in direct comparison to CD34+ hematopoietic stem cells cultivated without GQDs. Only a meta analysis reveals that the expression of 1171 genes is weakly affected, taking into account the more prominent changes just by the cell culture. Eight corresponding, weakly affected signaling pathways are identified, which include, but are not limited to, the triggering of apoptosis. These results suggest that GQDs with sizes in the range of a few nanometers hardly influence the CD34+ cells on the transcriptome level after 36 h of incubation, thereby demonstrating their high usability for in vivo studies, such as fluorescence labeling or delivery protocols, without strong effects on the functional status of the cells.


Asunto(s)
Regulación de la Expresión Génica , Grafito , Células Madre Hematopoyéticas/metabolismo , Puntos Cuánticos , Antígenos CD34/metabolismo , Apoptosis/genética , Materiales Biocompatibles , Supervivencia Celular , Perfilación de la Expresión Génica , Humanos , Nanoestructuras , Transcriptoma
5.
Int J Cancer ; 139(10): 2343-52, 2016 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-27389073

RESUMEN

New drugs for the treatment of multiple myeloma (MM) comprise immunomodulatory substances such as lenalidomide and related compounds. While lenalidomide has found its way into first-line treatment as well as into relapse therapy, little is known about lenalidomide effects on normal hematopoietic stem and progenitor cells (HSPCs). In this study, we investigated whether HSPCs are influenced by lenalidomide on a phenotypic, functional and gene expression level. For that purpose, samples from patients with MM were obtained who underwent equivalent first-line treatment including induction therapy, cytotoxic stem cell mobilization and high-dose melphalan therapy followed by autologous blood stem cell transplantation and a subsequent uniform lenalidomide consolidation treatment within a prospective clinical trial. We found that after six months of lenalidomide therapy, the number of CD34(+) HSPCs decreased. Additionally, lenalidomide affects the numerical composition of hematopoietic cells in the bone marrow while it does not affect long-term HSPC proliferation in vitro. We found a significant amplification of fetal hemoglobin (HbF) expression on a transcriptional level and can confirm a stimulated erythropoiesis on a phenotypic level. These effects were accompanied by silencing of the TGF-ß signaling pathway on the gene expression and protein level that is known to be amplified in active MM. However, these pleiotropic effects gave no evidence for mutagenic potential. In conclusion, lenalidomide does not exert long-term effects on proliferation of HSPCs but instead promotes erythropoiesis by shifting hemoglobin expression toward HbF and by silencing the TGF-ß signaling pathway.


Asunto(s)
Eritropoyesis/efectos de los fármacos , Mieloma Múltiple/sangre , Mieloma Múltiple/tratamiento farmacológico , Mielopoyesis/efectos de los fármacos , Talidomida/análogos & derivados , Inhibidores de la Angiogénesis/uso terapéutico , Médula Ósea/efectos de los fármacos , Quimioterapia de Consolidación , Hemoglobina Fetal/metabolismo , Células Madre Hematopoyéticas/efectos de los fármacos , Humanos , Lenalidomida , Persona de Mediana Edad , Talidomida/uso terapéutico
6.
Clin Hemorheol Microcirc ; 61(2): 111-8, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26410866

RESUMEN

INTRODUCTION: Phthalates are a group of synthetic plasticizers that are ubiquitous environmental pollutants with toxic and endocrine disrupting characteristics. DEHP is the most commonly used plasticizer in the world and is still applied to stem cell transfusion bags used for storage of hematopoietic stem and progenitor cells (CD34+ HSPC), which are transferred during stem cell transplantation. Here we examined the effect of DEHP on vitality of CD34+ HSPC as well as stem cell specific properties like migration and differentiation capacity - both important for successful stem cell transplantations. MATERIAL AND METHODS: CD34+ HSPC were incubated for 24 h and 72 h with DEHP concentrations ranging from 1 µg/ml to 250 µg/ml. DEHP was diluted in DMSO. Migration rate was analyzed along an SDF-1α gradient using Transwell migration inserts. Differentiation of CD34+ HSPC was investigated after two weeks in methylcellulose with colony stimulating factors. Apoptosis rate was measured via Annexin V and 7-AAD staining. RESULTS: 24 h of incubation with 10 µg/ml DEHP led to a significant (p <  0.01) decrease in migration rate of CD34+ HSPC (70.70% ± 7.53% ) with a minimum migration rate of 48.33% ± 6.72% in relation to control after incubation with 100 µg/ml DEHP for 72 h. Incubation with the highest tested DEHP concentrations (50 and 100 µg/ml) significantly (p <  0.05) altered colony formation rate and cell type distribution. Apoptosis rate of CD34+ HSPC significantly (p <  0.05) increased after incubation with concentrations of 10 µg/ml DEHP for 24 h (1.46 ± 0.19) with a maximum apoptosis rate of 2.71 ± 0.66 after 24 h incubation with the highest DEHP concentration (250 µg/ml) in relation to control. CONCLUSIONS: As shown, DEHP takes impact on migration rate, apoptosis rate, and differentiation of CD34+ HSPC. As these are functions with an important role in stem cell transplantations, the usage of DEHP-free stem cell transfusion bags should be considered.


Asunto(s)
Dietilhexil Ftalato/efectos adversos , Células Madre Hematopoyéticas/efectos de los fármacos , Plastificantes/efectos adversos , Antígenos CD34 , Apoptosis/efectos de los fármacos , Diferenciación Celular/efectos de los fármacos , Movimiento Celular/efectos de los fármacos , Dietilhexil Ftalato/farmacología , Humanos , Plastificantes/farmacología , Células Madre/efectos de los fármacos
7.
Clin Hemorheol Microcirc ; 58(1): 241-6, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25267456

RESUMEN

INTRODUCTION: Softeners like phthalate esters are ubiquitous in the environment and have been detected in transfusion bags, though there is only a limited amount of studies on the effect of phthalates on blood cells. This study seeks to determine effects on cell migration of human promyelocytic leukemia cells (HL-60) incubated with di(2-ethylhexyl)phthalate (DEHP) at concentrations found in blood bags. MATERIAL AND METHODS: HL-60 cells were incubated with DEHP concentrations ranging from 0.1 µg/ml to 1000 µg/ml, diluted in DMSO, over 24 h, 48 h, and 72 h. Migration rate was analyzed along an SDF-1α gradient using Transwell migration inserts. RESULTS: Of the applied concentrations 100 µg/ml, 250 µg/ml, 500 µg/ml, and 1000 µg/ml showed a significant decrease in migration rates relative to DMSO control at all measuring points (p < 0.05), with relative migration rates between 37.87 % for 100 µg/ml and 25.34 % for 1000 µg/ml relative to DMSO after 24 h of stimulation and 19.73 % for 100 µg/ml and 14.69 % for 1000 µg/ml respectively after 72 h of incubation. CONCLUSION: Our results indicate HL-60 to be a suitable in vitro model for examining effects of DEHP on the migration of blood and nucleated cells at concentrations found in blood bags.


Asunto(s)
Movimiento Celular/efectos de los fármacos , Dietilhexil Ftalato/farmacología , Plastificantes/farmacología , Apoptosis , Supervivencia Celular , Quimiocina CXCL12/metabolismo , Relación Dosis-Respuesta a Droga , Células HL-60 , Humanos
8.
Exp Hematol ; 41(9): 823-831.e2, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-23660069

RESUMEN

Homing and engraftment of hematopoietic stem and progenitor cells (HSPCs) during bone marrow transplantation are critically dependent on integrins such as ß1-integrin. In the present study, we show that ß1-integrin and the tetraspanin CD63 form a cell surface receptor complex for the soluble serum protein tissue inhibitor of metalloproteinases-1 (TIMP-1) on human CD34⁺ HSPCs. Through binding to this receptor complex, TIMP-1 activates ß1-integrin, increases adhesion and migration of human CD34⁺ cells, and protects these cells from induced apoptosis. TIMP-1 stimulation in murine bone marrow mononuclear cells also promotes migration and adhesion; this is associated with augmented homing of murine mononuclear cells and of murine LSK⁺ cells during bone marrow transplantation. These results not only indicate that TIMP-1 is conducive to HSPC homing; they also identify CD63 and ß1-integrin as a TIMP-1 receptor complex on HSPCs.


Asunto(s)
Trasplante de Médula Ósea , Movimiento Celular , Supervivencia de Injerto , Células Madre Hematopoyéticas/metabolismo , Inhibidor Tisular de Metaloproteinasa-1/metabolismo , Animales , Adhesión Celular , Femenino , Humanos , Integrina beta1/metabolismo , Masculino , Ratones , Tetraspanina 30/metabolismo , Trasplante Homólogo
9.
Mol Cell Proteomics ; 12(5): 1272-80, 2013 May.
Artículo en Inglés | MEDLINE | ID: mdl-23382103

RESUMEN

Bleeding complications are a significant clinical problem in patients with myelodysplastic syndromes even at sufficient platelet counts (>50,000/µl). However, the underlying pathology of this hemorrhagic diathesis is still unknown. Here, we analyzed the platelet proteome of patients with myelodysplastic syndromes by quantitative two-dimensional difference gel electrophoresis followed by mass spectrometric protein identification. Proteins identified with lower concentrations, such as Talin-1, Vinculin, Myosin-9, Filmain-A, and Actin play critical roles in integrin αIIbß3 signaling and thus platelet aggregation. Despite normal agonist receptor expression, calcium flux, and granule release upon activation, the activation capacity of integrin αIIbß3 was diminished in myelodysplastic syndrome platelets. Förster resonance energy transfer analysis showed a reduced co-localization of Talin-1 to the integrin's ß3-subunit, which is required for receptor activation and fibrinogen binding. In addition, platelet spreading on immobilized fibrinogen was incomplete, and platelet aggregation assays confirmed a general defect in integrin-dependent platelet aggregation in patients with myelodysplastic syndromes. Our data provide novel aspects on the molecular pathology of impaired platelet function in myelodysplastic syndromes and suggest a mechanism of defective integrin αIIbß3 signaling that may contribute to the hemorrhagic diathesis observed in these patients.


Asunto(s)
Plaquetas/metabolismo , Integrinas/fisiología , Síndromes Mielodisplásicos/fisiopatología , Agregación Plaquetaria , Proteoma/metabolismo , Adulto , Anciano , Anciano de 80 o más Años , Ácido Araquidónico/fisiología , Adhesión Celular , Células Cultivadas , Colágeno/fisiología , Femenino , Humanos , Masculino , Persona de Mediana Edad , Síndromes Mielodisplásicos/metabolismo , Mapas de Interacción de Proteínas , Receptores de Superficie Celular/metabolismo , Transducción de Señal , Adulto Joven
10.
Blood ; 120(13): 2620-30, 2012 Sep 27.
Artículo en Inglés | MEDLINE | ID: mdl-22517906

RESUMEN

Multiple myeloma (MM) is a clonal plasma cell disorder frequently accompanied by hematopoietic impairment. We show that hematopoietic stem and progenitor cells (HSPCs), in particular megakaryocyte-erythrocyte progenitors, are diminished in the BM of MM patients. Genomic profiling of HSPC subsets revealed deregulations of signaling cascades, most notably TGFß signaling, and pathways involved in cytoskeletal organization, migration, adhesion, and cell-cycle regulation in the patients. Functionally, proliferation, colony formation, and long-term self-renewal were impaired as a consequence of activated TGFß signaling. In accordance, TGFß levels in the BM extracellular fluid were elevated and mesenchymal stromal cells (MSCs) had a reduced capacity to support long-term hematopoiesis of HSPCs that completely recovered on blockade of TGFß signaling. Furthermore, we found defective actin assembly and down-regulation of the adhesion receptor CD44 in MM HSPCs functionally reflected by impaired migration and adhesion. Still, transplantation into myeloma-free NOG mice revealed even enhanced engraftment and normal differentiation capacities of MM HSPCs, which underlines that functional impairment of HSPCs depends on MM-related microenvironmental cues and is reversible. Taken together, these data implicate that hematopoietic suppression in MM emerges from the HSPCs as a result of MM-related microenvironmental alterations.


Asunto(s)
Antígenos CD34/metabolismo , Biomarcadores/metabolismo , Médula Ósea/patología , Células Madre Hematopoyéticas/patología , Células Progenitoras de Megacariocitos y Eritrocitos/patología , Mieloma Múltiple/metabolismo , Mieloma Múltiple/patología , Animales , Western Blotting , Médula Ósea/metabolismo , Estudios de Casos y Controles , Adhesión Celular , Ciclo Celular , Diferenciación Celular , Movimiento Celular , Proliferación Celular , Células Cultivadas , Femenino , Citometría de Flujo , Técnica del Anticuerpo Fluorescente , Perfilación de la Expresión Génica , Células Madre Hematopoyéticas/metabolismo , Humanos , Técnicas para Inmunoenzimas , Masculino , Células Progenitoras de Megacariocitos y Eritrocitos/metabolismo , Células Madre Mesenquimatosas/metabolismo , Células Madre Mesenquimatosas/patología , Ratones , Ratones Endogámicos NOD , Mieloma Múltiple/genética , Análisis de Secuencia por Matrices de Oligonucleótidos , ARN Mensajero/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transducción de Señal
11.
Ann Hematol ; 91(2): 173-81, 2012 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-21717102

RESUMEN

Acute myeloid leukemia (AML) is a heterogeneous hematological malignancy. Treatment of patients suffering from high-risk AML as defined by clinical parameters, cytogenetics, and/or molecular analyses is often unsuccessful. OSI-461 is a pro-apoptotic compound that has been proposed as a novel therapeutic option for patients suffering from solid tumors like prostate or colorectal carcinoma. But little is known about its anti-proliferative potential in AML. Hence, we treated bone marrow derived CD34(+) selected blast cells from 20 AML patients and the five AML cell lines KG-1a, THP-1, HL-60, U-937, and MV4-11 with the physiologically achievable concentration of 1 µM OSI-461 or equal amounts of DMSO as a control. Following incubation with OSI-461, we found a consistent induction of apoptosis and an accumulation of cells in the G2/M phase of the cell cycle. In addition, we demonstrate that the OSI-461 mediated anti-proliferative effects observed in AML are associated with the induction of the pro-apoptotic cytokine mda-7/IL-24 and activation of the growth arrest and DNA-damage inducible genes (GADD) 45α and 45γ. Furthermore, OSI-461 treated leukemia cells did not regain their proliferative potential for up to 8 days after cessation of treatment following the initial 48 h treatment period with 1 µM OSI-461. This indicates sufficient targeting of the leukemia-initiating cells in our in vitro experiments through OSI-461. The AML samples tested in this study included samples from patients who were resistant to conventional chemotherapy and/or had FLT3-ITD mutations demonstrating the high potential of OSI-461 in human AML.


Asunto(s)
Apoptosis/efectos de los fármacos , Leucemia Mieloide Aguda/tratamiento farmacológico , Leucemia Mieloide Aguda/patología , Sulindac/análogos & derivados , Ciclo Celular/efectos de los fármacos , Línea Celular Tumoral/efectos de los fármacos , Ensayos Clínicos como Asunto , Expresión Génica/efectos de los fármacos , Humanos , Inhibidores de Fosfodiesterasa/farmacología , Inhibidores de Fosfodiesterasa/uso terapéutico , Sulindac/farmacología , Sulindac/uso terapéutico
12.
Apoptosis ; 16(9): 889-901, 2011 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-21739277

RESUMEN

Acute myeloid leukemia is a heterogeneous disease with varying genetic and molecular pathologies. Non-steroidal anti-inflammatory drugs (NSAIDs) have been proven to possess significant anti-proliferative potential in various cancer cells in vitro and in vivo. Hence, treatment with these agents can be utilized to study disease specific anti-proliferative pathways. In this study, a total number of 42 bone marrow derived CD34(+) selected de novo AML patient samples and the AML cell lines THP-1 and HL-60 were treated with the NSAIDs Sulindac sulfide and Diclofenac. We analyzed viability, apoptosis, differentiation and addressed the molecular mechanisms involved. We found a consistent induction of apoptosis and to some extent an increased myeloid differentiation capacity in NSAID treated AML cells. Comprehensive protein and gene expression profiling of Diclofenac treated AML cells revealed transcriptional activation of GADD45α and its downstream MAPK/JNK pathway as well as increased protein levels of the caspase-3 precursor. This pointed towards a role of the c-Jun NH(2)-terminal kinase (JNK) in NSAID mediated apoptosis that we found indeed to be dependent on JNK activity as addition of a specific JNK-inhibitor abrogated apoptosis. Furthermore, the AP-1 transcription factor family members' c-Jun, JunB and Fra-2 were transcriptionally activated in NSAID treated AML cells and re-expression of these transcription factors led to activation of GADD45α with induction of apoptosis. Mechanistically, we demonstrate that NSAIDs induce apoptosis in AML through a novel pathway involving increased expression of AP-1 heterodimers, which by itself is sufficient to induce GADD45α expression with consecutive activation of JNK and induction of apoptosis.


Asunto(s)
Apoptosis , Diclofenaco/uso terapéutico , Leucemia Mieloide Aguda/tratamiento farmacológico , Sulindac/análogos & derivados , Factor de Transcripción AP-1/metabolismo , Antiinflamatorios no Esteroideos/uso terapéutico , Caspasa 3/metabolismo , Proteínas de Ciclo Celular/metabolismo , Diferenciación Celular , Supervivencia Celular , Clonación Molecular , Citometría de Flujo , Antígeno 2 Relacionado con Fos/metabolismo , Perfilación de la Expresión Génica/métodos , Regulación Neoplásica de la Expresión Génica , Vectores Genéticos , Células HL-60 , Humanos , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Leucemia Mieloide Aguda/genética , Leucemia Mieloide Aguda/metabolismo , Proteínas Nucleares/metabolismo , Proteínas Proto-Oncogénicas c-jun/metabolismo , Interferencia de ARN , Espectrometría de Masa por Láser de Matriz Asistida de Ionización Desorción/métodos , Sulindac/uso terapéutico , Activación Transcripcional
13.
Prostaglandins Other Lipid Mediat ; 94(1-2): 25-33, 2011 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-21167292

RESUMEN

Cytochrome P450 epoxygenases (CYP450) have been recently shown to promote malignant progression. Here we investigated the mRNA and protein expression and potential clinical relevance of CYP2C9 in esophageal cancer. Highest expression was detected in esophageal adenocarcinoma (EAC; n=78) and adjacent esophageal mucosa (NEM; n=79). Levels of CYP2C9 in EAC and NEM were significantly higher compared to esophageal squamous cell carcinoma (ESCC; n=105). Early tumor stages and well-differentiated tumors showed a significantly higher CYP2C9 expression compared to progressed tumors. Moreover, CYP2C9 expression was correlated to high Ki-67 labeling indices in EAC and Ki-67 positive tumor cells in EAC and ESCC. Selective inhibition of CYP2C9 decreased tumor cell proliferation (KYSE30, PT1590 and OE19) in vitro, which was abolished by 11,12-epoxyeicosatrienoic acid (11,12-EET). Cell-cycle analysis using FACS revealed that inhibition of CYP2C9 leads to a G0/G1 phase cell-cycle arrest. CYP2C9 seems to be relevant for early esophageal cancer development by promoting tumor cell proliferation. Pharmacological inhibition of CYP2C9 might contribute to a more efficient therapy in CYP2C9 highly expressing esophageal cancers.


Asunto(s)
Hidrocarburo de Aril Hidroxilasas/metabolismo , Carcinoma de Células Escamosas/enzimología , Carcinoma de Células Escamosas/patología , Proliferación Celular , Neoplasias Esofágicas/enzimología , Neoplasias Esofágicas/patología , Ácido 8,11,14-Eicosatrienoico/análogos & derivados , Ácido 8,11,14-Eicosatrienoico/farmacología , Hidrocarburo de Aril Hidroxilasas/genética , Línea Celular Tumoral , Citocromo P-450 CYP2C9 , Progresión de la Enfermedad , Fase G1 , Humanos , Inmunohistoquímica , Fase de Descanso del Ciclo Celular
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...