Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
1.
Haematologica ; 108(11): 3001-3010, 2023 11 01.
Article in English | MEDLINE | ID: mdl-37259567

ABSTRACT

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).


Subject(s)
Graft vs Host Disease , Hematopoietic Stem Cell Transplantation , Leukemia, Myeloid, Acute , Leukemia, Myelomonocytic, Chronic , Myelodysplastic Syndromes , Adult , Humans , Azacitidine/therapeutic use , Lenalidomide , Leukemia, Myelomonocytic, Chronic/therapy , Leukemia, Myelomonocytic, Chronic/complications , Lymphocyte Transfusion/adverse effects , Myelodysplastic Syndromes/pathology , Transplantation, Homologous/adverse effects , Chronic Disease , Graft vs Host Disease/diagnosis , Graft vs Host Disease/etiology , Graft vs Host Disease/drug therapy , T-Lymphocytes/pathology , Recurrence , Hematopoietic Stem Cell Transplantation/adverse effects
2.
Stem Cells ; 39(9): 1270-1284, 2021 09.
Article in English | MEDLINE | ID: mdl-34013984

ABSTRACT

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.


Subject(s)
Hematopoietic Stem Cells , Leukemia, Myeloid, Acute , Antigens, CD34/metabolism , Bone Marrow/metabolism , Hematopoiesis , Hematopoietic Stem Cells/metabolism , Humans , Leukemia, Myeloid, Acute/genetics
3.
Mar Drugs ; 17(9)2019 Sep 05.
Article in English | MEDLINE | ID: mdl-31491907

ABSTRACT

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.


Subject(s)
Antineoplastic Agents/therapeutic use , Phenol/therapeutic use , Adult , Aged , Apoptosis/drug effects , Cell Line, Tumor , Female , HL-60 Cells , Hematopoietic Stem Cells/drug effects , Humans , Jurkat Cells , Leukemia, Myeloid, Acute/drug therapy , Leukocytes, Mononuclear/drug effects , Male , Middle Aged , THP-1 Cells
4.
Sci Rep ; 9(1): 12028, 2019 08 19.
Article in English | MEDLINE | ID: mdl-31427693

ABSTRACT

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.


Subject(s)
Gene Expression Regulation , Graphite , Hematopoietic Stem Cells/metabolism , Quantum Dots , Antigens, CD34/metabolism , Apoptosis/genetics , Biocompatible Materials , Cell Survival , Gene Expression Profiling , Humans , Nanostructures , Transcriptome
5.
Int J Cancer ; 139(10): 2343-52, 2016 Nov 15.
Article in English | MEDLINE | ID: mdl-27389073

ABSTRACT

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.


Subject(s)
Erythropoiesis/drug effects , Multiple Myeloma/blood , Multiple Myeloma/drug therapy , Myelopoiesis/drug effects , Thalidomide/analogs & derivatives , Angiogenesis Inhibitors/therapeutic use , Bone Marrow/drug effects , Consolidation Chemotherapy , Fetal Hemoglobin/metabolism , Hematopoietic Stem Cells/drug effects , Humans , Lenalidomide , Middle Aged , Thalidomide/therapeutic use
6.
Clin Hemorheol Microcirc ; 61(2): 111-8, 2015.
Article in English | MEDLINE | ID: mdl-26410866

ABSTRACT

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.


Subject(s)
Diethylhexyl Phthalate/adverse effects , Hematopoietic Stem Cells/drug effects , Plasticizers/adverse effects , Antigens, CD34 , Apoptosis/drug effects , Cell Differentiation/drug effects , Cell Movement/drug effects , Diethylhexyl Phthalate/pharmacology , Humans , Plasticizers/pharmacology , Stem Cells/drug effects
7.
Clin Hemorheol Microcirc ; 58(1): 241-6, 2014.
Article in English | MEDLINE | ID: mdl-25267456

ABSTRACT

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.


Subject(s)
Cell Movement/drug effects , Diethylhexyl Phthalate/pharmacology , Plasticizers/pharmacology , Apoptosis , Cell Survival , Chemokine CXCL12/metabolism , Dose-Response Relationship, Drug , HL-60 Cells , Humans
8.
Exp Hematol ; 41(9): 823-831.e2, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23660069

ABSTRACT

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.


Subject(s)
Bone Marrow Transplantation , Cell Movement , Graft Survival , Hematopoietic Stem Cells/metabolism , Tissue Inhibitor of Metalloproteinase-1/metabolism , Animals , Cell Adhesion , Female , Humans , Integrin beta1/metabolism , Male , Mice , Tetraspanin 30/metabolism , Transplantation, Homologous
9.
Mol Cell Proteomics ; 12(5): 1272-80, 2013 May.
Article in English | MEDLINE | ID: mdl-23382103

ABSTRACT

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.


Subject(s)
Blood Platelets/metabolism , Integrins/physiology , Myelodysplastic Syndromes/physiopathology , Platelet Aggregation , Proteome/metabolism , Adult , Aged , Aged, 80 and over , Arachidonic Acid/physiology , Cell Adhesion , Cells, Cultured , Collagen/physiology , Female , Humans , Male , Middle Aged , Myelodysplastic Syndromes/metabolism , Protein Interaction Maps , Receptors, Cell Surface/metabolism , Signal Transduction , Young Adult
10.
Blood ; 120(13): 2620-30, 2012 Sep 27.
Article in English | MEDLINE | ID: mdl-22517906

ABSTRACT

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.


Subject(s)
Antigens, CD34/metabolism , Biomarkers/metabolism , Bone Marrow/pathology , Hematopoietic Stem Cells/pathology , Megakaryocyte-Erythroid Progenitor Cells/pathology , Multiple Myeloma/metabolism , Multiple Myeloma/pathology , Animals , Blotting, Western , Bone Marrow/metabolism , Case-Control Studies , Cell Adhesion , Cell Cycle , Cell Differentiation , Cell Movement , Cell Proliferation , Cells, Cultured , Female , Flow Cytometry , Fluorescent Antibody Technique , Gene Expression Profiling , Hematopoietic Stem Cells/metabolism , Humans , Immunoenzyme Techniques , Male , Megakaryocyte-Erythroid Progenitor Cells/metabolism , Mesenchymal Stem Cells/metabolism , Mesenchymal Stem Cells/pathology , Mice , Mice, Inbred NOD , Multiple Myeloma/genetics , Oligonucleotide Array Sequence Analysis , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction
11.
Ann Hematol ; 91(2): 173-81, 2012 Feb.
Article in English | MEDLINE | ID: mdl-21717102

ABSTRACT

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.


Subject(s)
Apoptosis/drug effects , Leukemia, Myeloid, Acute/drug therapy , Leukemia, Myeloid, Acute/pathology , Sulindac/analogs & derivatives , Cell Cycle/drug effects , Cell Line, Tumor/drug effects , Clinical Trials as Topic , Gene Expression/drug effects , Humans , Phosphodiesterase Inhibitors/pharmacology , Phosphodiesterase Inhibitors/therapeutic use , Sulindac/pharmacology , Sulindac/therapeutic use
12.
Apoptosis ; 16(9): 889-901, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21739277

ABSTRACT

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.


Subject(s)
Apoptosis , Diclofenac/therapeutic use , Leukemia, Myeloid, Acute/drug therapy , Sulindac/analogs & derivatives , Transcription Factor AP-1/metabolism , Anti-Inflammatory Agents, Non-Steroidal/therapeutic use , Caspase 3/metabolism , Cell Cycle Proteins/metabolism , Cell Differentiation , Cell Survival , Cloning, Molecular , Flow Cytometry , Fos-Related Antigen-2/metabolism , Gene Expression Profiling/methods , Gene Expression Regulation, Neoplastic , Genetic Vectors , HL-60 Cells , Humans , JNK Mitogen-Activated Protein Kinases/metabolism , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/metabolism , Nuclear Proteins/metabolism , Proto-Oncogene Proteins c-jun/metabolism , RNA Interference , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization/methods , Sulindac/therapeutic use , Transcriptional Activation
13.
Prostaglandins Other Lipid Mediat ; 94(1-2): 25-33, 2011 Feb.
Article in English | MEDLINE | ID: mdl-21167292

ABSTRACT

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.


Subject(s)
Aryl Hydrocarbon Hydroxylases/metabolism , Carcinoma, Squamous Cell/enzymology , Carcinoma, Squamous Cell/pathology , Cell Proliferation , Esophageal Neoplasms/enzymology , Esophageal Neoplasms/pathology , 8,11,14-Eicosatrienoic Acid/analogs & derivatives , 8,11,14-Eicosatrienoic Acid/pharmacology , Aryl Hydrocarbon Hydroxylases/genetics , Cell Line, Tumor , Cytochrome P-450 CYP2C9 , Disease Progression , G1 Phase , Humans , Immunohistochemistry , Resting Phase, Cell Cycle
SELECTION OF CITATIONS
SEARCH DETAIL
...