Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 30
Filter
1.
Immunity ; 47(4): 789-802.e9, 2017 10 17.
Article in English | MEDLINE | ID: mdl-29045907

ABSTRACT

Inhibitors of the receptor tyrosine kinase c-MET are currently used in the clinic to target oncogenic signaling in tumor cells. We found that concomitant c-MET inhibition promoted adoptive T cell transfer and checkpoint immunotherapies in murine cancer models by increasing effector T cell infiltration in tumors. This therapeutic effect was independent of tumor cell-intrinsic c-MET dependence. Mechanistically, c-MET inhibition impaired the reactive mobilization and recruitment of neutrophils into tumors and draining lymph nodes in response to cytotoxic immunotherapies. In the absence of c-MET inhibition, neutrophils recruited to T cell-inflamed microenvironments rapidly acquired immunosuppressive properties, restraining T cell expansion and effector functions. In cancer patients, high serum levels of the c-MET ligand HGF correlated with increasing neutrophil counts and poor responses to checkpoint blockade therapies. Our findings reveal a role for the HGF/c-MET pathway in neutrophil recruitment and function and suggest that c-MET inhibitor co-treatment may improve responses to cancer immunotherapy in settings beyond c-MET-dependent tumors.


Subject(s)
Immunotherapy/methods , Neoplasms, Experimental/therapy , Neutrophils/immunology , Proto-Oncogene Proteins c-met/immunology , Animals , Cell Line, Tumor , Cell Proliferation/genetics , Gene Expression Profiling/methods , Gene Expression Regulation, Neoplastic/immunology , Interferon-gamma/immunology , Interferon-gamma/metabolism , Kaplan-Meier Estimate , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Neoplasms, Experimental/immunology , Neoplasms, Experimental/metabolism , Neutrophils/metabolism , Proto-Oncogene Proteins c-met/genetics , Proto-Oncogene Proteins c-met/metabolism , Signal Transduction/genetics , Signal Transduction/immunology , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Tumor Microenvironment/genetics , Tumor Microenvironment/immunology
2.
Blood ; 129(4): 460-472, 2017 01 26.
Article in English | MEDLINE | ID: mdl-27683414

ABSTRACT

Epithelial-to-mesenchymal-transition (EMT) is critical for normal embryogenesis and effective postnatal wound healing, but is also associated with cancer metastasis. SNAIL, ZEB, and TWIST families of transcription factors are key modulators of the EMT process, but their precise roles in adult hematopoietic development and homeostasis remain unclear. Here we report that genetic inactivation of Zeb2 results in increased frequency of stem and progenitor subpopulations within the bone marrow (BM) and spleen and that these changes accompany differentiation defects in multiple hematopoietic cell lineages. We found no evidence that Zeb2 is critical for hematopoietic stem cell self-renewal capacity. However, knocking out Zeb2 in the BM promoted a phenotype with several features that resemble human myeloproliferative disorders, such as BM fibrosis, splenomegaly, and extramedullary hematopoiesis. Global gene expression and intracellular signal transduction analysis revealed perturbations in specific cytokine and cytokine receptor-related signaling pathways following Zeb2 loss, especially the JAK-STAT and extracellular signal-regulated kinase pathways. Moreover, we detected some previously unknown mutations within the human Zeb2 gene (ZFX1B locus) from patients with myeloid disease. Collectively, our results demonstrate that Zeb2 controls adult hematopoietic differentiation and lineage fidelity through widespread modulation of dominant signaling pathways that may contribute to blood disorders.


Subject(s)
Cytokines/genetics , Epithelial-Mesenchymal Transition/genetics , Hematopoiesis, Extramedullary/genetics , Homeodomain Proteins/genetics , Primary Myelofibrosis/genetics , Repressor Proteins/genetics , Splenomegaly/genetics , Adult , Animals , Base Sequence , Bone Marrow/metabolism , Bone Marrow/pathology , Cell Differentiation , Cell Lineage/genetics , Cytokines/metabolism , Gene Expression Regulation , Humans , Janus Kinases/genetics , Janus Kinases/metabolism , Mice , Mice, Knockout , Mitogen-Activated Protein Kinase 1/genetics , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/genetics , Mitogen-Activated Protein Kinase 3/metabolism , Mutation , Primary Myelofibrosis/metabolism , Primary Myelofibrosis/pathology , Repressor Proteins/deficiency , STAT Transcription Factors/genetics , STAT Transcription Factors/metabolism , Signal Transduction , Spleen/metabolism , Spleen/pathology , Splenomegaly/metabolism , Splenomegaly/pathology , Stem Cells/metabolism , Stem Cells/pathology , Transcription, Genetic , Zinc Finger E-box Binding Homeobox 2
3.
Arch Gynecol Obstet ; 297(2): 281-284, 2018 02.
Article in English | MEDLINE | ID: mdl-29110117

ABSTRACT

PURPOSE: The diagnosis of acute promyelocytic leukemia (APL) in pregnancy is an uncommon, life-threatening emergency. Choice of treatment and management of complications are challenging. METHODS: We report the case of a patient with diagnosis of APL at gestational age (GA) 24 + 4. We describe the interdisciplinary management during pregnancy and delivery and provide a 2-year follow-up. Existing reports on APL in pregnancy are summarized. RESULTS: Single-agent induction therapy with all-trans retinoic acid (ATRA) was started and resulted in normalization of blood cell counts after 32 days. Vaginal delivery of a healthy baby occurred at GA 34 + 4. Consolidation therapy consisted of four courses of ATRA and arsenic trioxide (ATO). Less than 100 cases of APL in pregnancy are published. Misdiagnosis as HELLP syndrome with fatal outcome may occur. Combination therapies (ATRA-plus anthracyclines) were used in the majority of reports. CONCLUSIONS: Diagnosis and treatment of APL during pregnancy continues to be a challenge requiring interdisciplinary team work. Single-agent ATRA therapy may evolve as a safe and less-toxic treatment modality.


Subject(s)
Antineoplastic Agents/therapeutic use , Arsenic Trioxide/therapeutic use , Leukemia, Promyelocytic, Acute/drug therapy , Pregnancy Complications, Neoplastic/drug therapy , Tretinoin/therapeutic use , Adult , Antineoplastic Agents/adverse effects , Arsenic Trioxide/administration & dosage , Arsenicals , Female , Gestational Age , Humans , Leukemia, Promyelocytic, Acute/diagnosis , Neoadjuvant Therapy , Pregnancy , Pregnancy Complications, Neoplastic/diagnosis , Treatment Outcome , Tretinoin/administration & dosage
4.
Tumour Biol ; 36(4): 2725-35, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25487614

ABSTRACT

We have recently shown that staurosporine mediates the conversion of small cell lung carcinoma (SCLC) cells into a neuron-like process-bearing phenotype. Here, we have extended these studies to the staurosporine analogs K252a, lestaurtinib, PKC412, stauprimide, and UCN-01 and analyzed their influence on process extension, cell cycle distribution, and induction of polyploidy in four SCLC cell lines. In GLC-2 cells, all compounds provoked extensive process formation with the exception of PKC412 that showed no response. In H1184 cells, process formation was predominantly induced by staurosporine and, to lesser extent, in lestaurtinib-, stauprimide-, and UCN-01-treated cells. In the presence of K252a or PKC412, cells became bipolar and spindle shaped or showed pronounced cell flattening. In GLC-36 and SCLC-24H cells, only cell flattening was detectable. Process formation was reversible upon drug removal as shown for GLC-2 and H1184 cells. Fluorescence-activated cell sorting (FACS) and fluorescence in situ hybridization (FISH) analysis indicated the induction of polyploidy in all staurosporine and in two out of four stauprimide-treated SCLC cell lines. For other staurosporine analogs, polyploidy was observed only in UCN-01-treated GLC-36 cells and in K252a-treated H1184 and GLC-36 cells. The presence of staurosporine or its analogs did not alter the constitutive activation pattern of the canonical Akt/PI3K or MEK/extracellular signal-regulated kinase (ERK)1/2 signaling pathways nor could we detect an influence of stauprimide application on the expression level of the c-Myc oncogene. These data demonstrate that in SCLC cells, albeit a higher substrate specificity, staurosporine analogs can induce staurosporine-comparable effects.


Subject(s)
Cell Cycle/drug effects , Cell Proliferation/drug effects , Small Cell Lung Carcinoma/drug therapy , Staurosporine/administration & dosage , Carbazoles/administration & dosage , Cell Line, Tumor , Furans , Humans , Indole Alkaloids/administration & dosage , Polyploidy , Signal Transduction/drug effects , Small Cell Lung Carcinoma/pathology , Staurosporine/analogs & derivatives
5.
Blood ; 117(21): 5620-30, 2011 May 26.
Article in English | MEDLINE | ID: mdl-21355089

ABSTRACT

Zeb2 (Sip1/Zfhx1b) is a member of the zinc-finger E-box-binding (ZEB) family of transcriptional repressors previously demonstrated to regulate epithelial-to-mesenchymal transition (EMT) processes during embryogenesis and tumor progression. We found high Zeb2 mRNA expression levels in HSCs and hematopoietic progenitor cells (HPCs), and examined Zeb2 function in hematopoiesis through a conditional deletion approach using the Tie2-Cre and Vav-iCre recombination mouse lines. Detailed cellular analysis demonstrated that Zeb2 is dispensable for hematopoietic cluster and HSC formation in the aorta-gonadomesonephros region of the embryo, but is essential for normal HSC/HPC differentiation. In addition, Zeb2-deficient HSCs/HPCs fail to properly colonize the fetal liver and/or bone marrow and show enhanced adhesive properties associated with increased ß1 integrin and Cxcr4 expression. Moreover, deletion of Zeb2 resulted in embryonic (Tie2-Cre) and perinatal (Vav-icre) lethality due to severe cephalic hemorrhaging and decreased levels of angiopoietin-1 and, subsequently, improper pericyte coverage of the cephalic vasculature. These results reveal essential roles for Zeb2 in embryonic hematopoiesis and are suggestive of a role for Zeb2 in hematopoietic-related pathologies in the adult.


Subject(s)
Cell Differentiation , Embryo, Mammalian/cytology , Embryo, Mammalian/metabolism , Epithelial-Mesenchymal Transition , Hematopoiesis/physiology , Hematopoietic Stem Cells/cytology , Homeodomain Proteins/physiology , Repressor Proteins/physiology , Animals , Cadherins/metabolism , Cell Movement , Female , Flow Cytometry , Genes, Lethal , Hematopoietic Stem Cells/metabolism , Integrases/metabolism , Male , Mice , Mice, Knockout , Zinc Finger E-box Binding Homeobox 2 , Zinc Fingers
6.
Mol Med ; 18: 1197-208, 2012 Oct 24.
Article in English | MEDLINE | ID: mdl-22801793

ABSTRACT

Ovarian cancers are heterogeneous and contain stemlike cells that are able to self-renew and are responsible for sustained tumor growth. Metastasis in the peritoneal cavity occurs more frequently in ovarian cancer than in other malignancies, but the underlying mechanism remains largely unknown. We have identified that ovarian cancer stemlike cells (CSCs), which were defined as side population (SP) cells, were present in patients' ascitic fluid and mesenchymally transformed cell lines, ES-2 and HO-8910PM. SP cells, which were sorted from both cell lines and implanted into immunocompromised mice, were localized to the xenografted tumor boundary. In addition, SP cells exhibited an epithelial phenotype and showed a distinct gene expression profile with reduced expression of cell adhesion molecules (CAMs), indicating that SP cells exert an important role in ovarian cancer progression on the basis of their delicate interaction with the surrounding microenvironment and anatomical localization in tumors. In contrast, non-SP cells exhibited a more mesenchymal phenotype and showed more increased invasive potential than SP cells. This heterogeneity was observed as an endogenous transformation via the epithelial-mesenchymal transition (EMT) process. Inhibition of the EMT process by Snail1 silencing reduced the SP cell frequency, and affected their invasive capacity and engraftment. These findings illustrate the interplay between epithelial ovarian CSCs and the EMT, and exert a link to explain tumor heterogeneity and its necessity for ovarian cancer maintenance, metastasis and progression.


Subject(s)
Cell Lineage , Cell Transformation, Neoplastic/pathology , Epithelial-Mesenchymal Transition , Mesoderm/pathology , Neoplasm Transplantation , Neoplasms, Glandular and Epithelial/pathology , Neoplastic Stem Cells/pathology , Ovarian Neoplasms/pathology , Animals , Ascitic Fluid/drug effects , Ascitic Fluid/pathology , Biomarkers, Tumor/metabolism , Carcinoma, Ovarian Epithelial , Cell Adhesion/drug effects , Cell Differentiation/drug effects , Cell Differentiation/genetics , Cell Line, Tumor , Cell Lineage/drug effects , Cell Proliferation/drug effects , Cell Transformation, Neoplastic/drug effects , Cell Transformation, Neoplastic/genetics , Epithelial-Mesenchymal Transition/drug effects , Epithelial-Mesenchymal Transition/genetics , Female , Gene Expression Profiling , Gene Expression Regulation, Neoplastic/drug effects , Humans , Injections, Subcutaneous , Mesoderm/drug effects , Mice , Mice, Inbred BALB C , Neoplasm Invasiveness , Neoplasms, Glandular and Epithelial/genetics , Neoplastic Stem Cells/drug effects , Ovarian Neoplasms/genetics , Side-Population Cells/drug effects , Side-Population Cells/pathology , Snail Family Transcription Factors , Transcription Factors/metabolism , Transforming Growth Factor beta1/pharmacology , Xenograft Model Antitumor Assays
8.
Nature ; 443(7110): 421-6, 2006 Sep 28.
Article in English | MEDLINE | ID: mdl-16957735

ABSTRACT

Stem-cell ageing is thought to contribute to altered tissue maintenance and repair. Older humans experience increased bone marrow failure and poorer haematologic tolerance of cytotoxic injury. Haematopoietic stem cells (HSCs) in older mice have decreased per-cell repopulating activity, self-renewal and homing abilities, myeloid skewing of differentiation, and increased apoptosis with stress. Here we report that the cyclin-dependent kinase inhibitor p16INK4a, the level of which was previously noted to increase in other cell types with age, accumulates and modulates specific age-associated HSC functions. Notably, in the absence of p16INK4a, HSC repopulating defects and apoptosis were mitigated, improving the stress tolerance of cells and the survival of animals in successive transplants, a stem-cell-autonomous tissue regeneration model. Inhibition of p16INK4a may ameliorate the physiological impact of ageing on stem cells and thereby improve injury repair in aged tissue.


Subject(s)
Cellular Senescence , Cyclin-Dependent Kinase Inhibitor p16/metabolism , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/metabolism , Aging , Animals , Apoptosis , Basic Helix-Loop-Helix Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/metabolism , Bone Marrow Transplantation , Cell Count , Cell Proliferation , Cyclin-Dependent Kinase Inhibitor p16/genetics , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Mice , Transcription Factor HES-1
10.
Anal Cell Pathol (Amst) ; 2018: 1754085, 2018.
Article in English | MEDLINE | ID: mdl-30406001

ABSTRACT

Cultivation of A549 non-small-cell lung carcinoma (NSCLC) cells in the presence of staurosporine (SSP) leads to a reduction or a lack of proliferation in a concentration-dependent manner. This inhibition of proliferation is accompanied by the generation of polyploid giant cancer cells (PGCCs) that are characterized by cell flattening, increased cell size, polyploidy, and polynucleation as determined by crystal violet staining, BrdU and DiI labelling, and flow cytometry as well as video time-lapse analysis. Continuous SSP treatment of A549 cells can preserve PGCCs for at least two months in a resting state. Upon removal of SSP, A549 PGCCs restart to divide and exhibit a proliferation pattern and cellular morphology indistinguishable from cells where PGCCs originally derived from. Thus, SSP-treated A549 cells represent a simple and reliable experimental model for the reversible generation of PGCCs and their subsequent experimental analysis.


Subject(s)
Carcinoma, Non-Small-Cell Lung/pathology , Giant Cells/pathology , Lung Neoplasms/pathology , Polyploidy , Staurosporine/adverse effects , A549 Cells , Bromodeoxyuridine/metabolism , Cell Proliferation/drug effects , Cell Size/drug effects , Giant Cells/drug effects , Humans
11.
Sci Rep ; 8(1): 2833, 2018 02 12.
Article in English | MEDLINE | ID: mdl-29434282

ABSTRACT

In adult mammals, hematopoietic stem cells (HSCs) reside in the bone marrow and are in part regulated by the bone marrow microenvironment, called the stem cell niche. We have previously identified the bone marrow morphogen osteopontin (OPN), which is abundantly present in the bone marrow extracellular matrix, as a negative regulator of the size of the HSC pool under physiological conditions. Here, we study the impact of OPN on HSC function during aging using an OPN-knockout mouse model. We show that during aging OPN deficiency is associated with an increase in lymphocytes and a decline in erythrocytes in peripheral blood. In a bone marrow transplantation setting, aged OPN-deficient stem cells show reduced reconstitution ability likely due to insufficient differentiation of HSCs into more mature cells. In serial bone marrow transplantation, aged OPN-/- bone marrow cells fail to adequately reconstitute red blood cells and platelets, resulting in severe anemia and thrombocytopenia as well as premature deaths of recipient mice. Thus, OPN has different effects on HSCs in aged and young animals and is particularly important to maintain stem cell function in aging mice.


Subject(s)
Aging/genetics , Anemia/genetics , Hematopoietic Stem Cells/cytology , Osteopontin/genetics , Thrombocytopenia/genetics , Aging/blood , Aging/metabolism , Anemia/blood , Anemia/metabolism , Animals , Cell Differentiation , Cells, Cultured , Erythrocytes/metabolism , Gene Knockout Techniques , Hematopoietic Stem Cell Transplantation , Hematopoietic Stem Cells/metabolism , Lymphocytes/metabolism , Mice , Mice, Inbred C57BL , Osteopontin/metabolism , Stem Cell Niche , Thrombocytopenia/blood , Thrombocytopenia/metabolism
12.
Clin Lymphoma Myeloma Leuk ; 18(4): 266-271, 2018 04.
Article in English | MEDLINE | ID: mdl-29510895

ABSTRACT

INTRODUCTION: Tyrosine kinase inhibitors (TKIs) can safely be discontinued in chronic myeloid leukemia (CML) patients with sustained deep molecular response. ABCG2 (breast cancer resistance protein), OCT1 (organic cation transporter 1), and ABCB1 (multidrug resistance protein 1) gene products are known to play a crucial role in acquired pharmacogenetic TKI resistance. Their influence on treatment-free remission (TFR) has not yet been investigated. MATERIALS AND METHODS: RNA was isolated on the last day of TKI intake from peripheral blood leukocytes of 132 chronic phase CML patients who discontinued TKI treatment within the European Stop Tyrosine Kinase Inhibitor Study trial. Plasmid standards were designed including subgenic inserts of OCT1, ABCG2, and ABCB1 together with GUSB as reference gene. For expression analyses, quantitative real-time polymerase chain reaction was used. Multiple Cox regression analysis was performed. In addition, gene expression cutoffs for patient risk stratification were investigated. RESULTS: The TFR rate of 132 patients, 12 months after TKI discontinuation, was 54% (95% confidence interval [CI], 46%-62%). ABCG2 expression (‰) was retained as the only significant variable (P = .02; hazard ratio, 1.04; 95% CI, 1.01-1.07) in multiple Cox regression analysis. Only for the ABCG2 efflux transporter, a significant cutoff was found (P = .04). Patients with an ABCG2/GUSB transcript level >4.5‰ (n = 93) showed a 12-month TFR rate of 47% (95% CI, 37%-57%), whereas patients with low ABCG2 expression (≤4.5‰; n = 39) had a 12-month TFR rate of 72% (95% CI, 55%-82%). CONCLUSION: In this study, we investigated the effect of pharmacogenetics in the context of a CML treatment discontinuation trial. The transcript levels of the efflux transporter ABCG2 predicted TFR after TKI discontinuation.


Subject(s)
Antineoplastic Agents/administration & dosage , Biomarkers, Tumor/genetics , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics , Pharmacogenomic Variants/genetics , ATP Binding Cassette Transporter, Subfamily B/genetics , ATP Binding Cassette Transporter, Subfamily G, Member 2/genetics , Adult , Aged , Aged, 80 and over , Disease-Free Survival , Female , Humans , Kaplan-Meier Estimate , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/mortality , Male , Middle Aged , Neoplasm Proteins/genetics , Octamer Transcription Factor-1/genetics , Protein Kinase Inhibitors/administration & dosage , Remission Induction , Transcriptome
14.
Z Orthop Unfall ; 155(6): 716-726, 2017 Dec.
Article in German | MEDLINE | ID: mdl-28934820

ABSTRACT

In Germany and other European countries, cancer is the second most common cause of death after cardiovascular disease. Although 5-year survival rates for several types of cancer have significantly improved over the last 30 years, metastasis to the bone almost always leads to incurable disease. Aside from the rare primary bone tumours, the treatment of bone metastases now accounts for a major part of tumour orthopaedic workload and requires close interdisciplinary coordination between specialists in oncology, radiology and the discipline of the primary tumour entity. Due to improvements in oncological treatment regimes, long survival times can be achieved. Therefore, the management of so-called "SRE" (skeletal-related events) has gained importance, even in palliative situations. On the basis of a selective literature review, the following article points out the underlying pathophysiological processes of bone metastases and outlines different diagnostic approaches and their relevance in the current clinical setting.


Subject(s)
Bone Neoplasms/secondary , Bone Neoplasms/therapy , Interdisciplinary Communication , Intersectoral Collaboration , Biopsy , Bone Neoplasms/diagnosis , Bone Neoplasms/physiopathology , Bone and Bones/pathology , Cell Communication/physiology , Diagnostic Imaging/methods , Fractures, Spontaneous/diagnosis , Fractures, Spontaneous/mortality , Fractures, Spontaneous/physiopathology , Fractures, Spontaneous/therapy , Humans , Neoplasms, Unknown Primary/diagnosis , Neoplasms, Unknown Primary/pathology , Neoplasms, Unknown Primary/physiopathology , Neoplasms, Unknown Primary/therapy , Neoplastic Cells, Circulating/pathology , Osteolysis/physiopathology , Survival Rate
15.
Sci Rep ; 7: 41427, 2017 01 27.
Article in English | MEDLINE | ID: mdl-28128288

ABSTRACT

Activating mutations leading to ligand-independent signaling of the stem cell factor receptor KIT are associated with several hematopoietic malignancies. One of the most common alterations is the D816V mutation. In this study, we characterized mice, which conditionally express the humanized KITD816V receptor in the adult hematopoietic system to determine the pathological consequences of unrestrained KIT signaling during blood cell development. We found that KITD816V mutant animals acquired a myeloproliferative neoplasm similar to polycythemia vera, marked by a massive increase in red blood cells and severe splenomegaly caused by excessive extramedullary erythropoiesis. Moreover, we found mobilization of stem cells from bone marrow to the spleen. Splenectomy prior to KITD816V induction prevented expansion of red blood cells, but rapidly lead to a state of aplastic anemia and bone marrow fibrosis, reminiscent of post polycythemic myeloid metaplasia, the spent phase of polycythemia vera. Our results show that the extramedullary hematopoietic niche microenvironment significantly influences disease outcome in KITD816V mutant mice, turning this model a valuable tool for studying the interplay between functionally abnormal hematopoietic cells and their microenvironment during development of polycythemia vera-like disease and myelofibrosis.


Subject(s)
Bone Marrow Neoplasms/genetics , Bone Marrow Neoplasms/pathology , Cell Transformation, Neoplastic/pathology , Proto-Oncogene Proteins c-kit/genetics , Spleen/pathology , Tumor Microenvironment , Animals , Bone Marrow Cells/pathology , Bone Marrow Neoplasms/blood , Cell Proliferation , Cell Transformation, Neoplastic/genetics , Disease Models, Animal , Erythrocytes/metabolism , Erythrocytes/pathology , Fibrosis , GATA2 Transcription Factor/metabolism , Gene Expression Regulation, Neoplastic , Hematopoiesis , Hematopoiesis, Extramedullary , Hematopoietic Stem Cells/metabolism , Mice, Inbred C57BL , Phenotype , Polycythemia Vera/genetics , Polycythemia Vera/pathology , Signal Transduction , Spleen/surgery , Splenomegaly/pathology
16.
Int J Oncol ; 27(5): 1273-82, 2005 Nov.
Article in English | MEDLINE | ID: mdl-16211222

ABSTRACT

Prostate cancer is among the most frequent tumours in industrialized nations and many questions remain open concerning the molecular events underlying its development and progression. In the present study we have combined cDNA array hybridization to laser-assisted microdissection (LAM) in order to investigate differences in gene expression between epithelial and stromal cells of prostate cancer and normal peripheral prostate tissue. Results have been verified for selected candidate genes by quantitative real-time RT-PCR. Using this approach and immunohistochemistry we could demonstrate a down-regulation of cellular retinoic acid binding protein 2 (CRABP2) mRNA and protein in carcinoma cells compared to normal glandular cells. CRABP2 is a main regulator of anti-carcinogenic activities of retinoic acid and may become a novel diagnostic marker and experimental therapeutic tool for prostate cancer. In addition, results of cDNA array hybridization suggest an up-regulation of 34 further genes and a down-regulation of 6 genes in cancer tissues compared to normal peripheral prostate tissues. Several of these genes have already been reported to be associated with carcinogenesis in organs such as the prostate.


Subject(s)
Prostatic Neoplasms/genetics , Receptors, Retinoic Acid/biosynthesis , Aged , Biomarkers, Tumor/analysis , Cell Transformation, Neoplastic , Down-Regulation , Epithelial Cells/physiology , Gene Expression Profiling , Humans , Immunohistochemistry , Male , Middle Aged , Oligonucleotide Array Sequence Analysis , Prostatic Neoplasms/pathology , Reverse Transcriptase Polymerase Chain Reaction , Stromal Cells/physiology
17.
Nat Commun ; 6: 5794, 2015 Jan 07.
Article in English | MEDLINE | ID: mdl-25565005

ABSTRACT

Early T-cell precursor leukaemia (ETP-ALL) is a high-risk subtype of human leukaemia that is poorly understood at the molecular level. Here we report translocations targeting the zinc finger E-box-binding transcription factor ZEB2 as a recurrent genetic lesion in immature/ETP-ALL. Using a conditional gain-of-function mouse model, we demonstrate that sustained Zeb2 expression initiates T-cell leukaemia. Moreover, Zeb2-driven mouse leukaemia exhibit some features of the human immature/ETP-ALL gene expression signature, as well as an enhanced leukaemia-initiation potential and activated Janus kinase (JAK)/signal transducers and activators of transcription (STAT) signalling through transcriptional activation of IL7R. This study reveals ZEB2 as an oncogene in the biology of immature/ETP-ALL and paves the way towards pre-clinical studies of novel compounds for the treatment of this aggressive subtype of human T-ALL using our Zeb2-driven mouse model.


Subject(s)
Gene Expression Regulation, Neoplastic/physiology , Homeodomain Proteins/genetics , Leukemia, T-Cell/physiopathology , Repressor Proteins/genetics , Signal Transduction/physiology , Animals , Blotting, Western , Chromatin Immunoprecipitation , Flow Cytometry , Gene Expression Profiling , Gene Expression Regulation, Neoplastic/genetics , Histological Techniques , Homeodomain Proteins/immunology , Humans , Immunohistochemistry , In Situ Hybridization, Fluorescence , Janus Kinases/metabolism , Kaplan-Meier Estimate , Karyotyping , Luciferases , Mice , Real-Time Polymerase Chain Reaction , Receptors, Interleukin-7/metabolism , Repressor Proteins/immunology , STAT Transcription Factors/metabolism , Signal Transduction/genetics , Zinc Finger E-box Binding Homeobox 2
18.
Int J Mol Med ; 11(4): 449-53, 2003 Apr.
Article in English | MEDLINE | ID: mdl-12632096

ABSTRACT

Laser-assisted microdissection (LAM) allows isolation of specific cell populations for molecular studies. The combination of LAM and of real-time quantitative reverse transcriptase-polymerase chain reaction (RT-PCR) enables generation of quantitative cell-specific gene expression data. Histochemical stains used to identify cells desired for LAM should provide acceptable morphology and not interfere with RNA or with subsequent molecular analysis techniques. To determine a reliable stain for analysing RNA, using the housekeeping gene, RPL13A, we performed quantitative gene expression analysis of laser microdissected cells from prostatic frozen tissues. The frozen sections were histochemically stained with hematoxylin, methyl green, toluidine blue O and May-Grunwald. After laser microdissection real-time quantitative RT-PCR was performed. Methyl green yielded more RT-PCR product than did the other dyes. The lowest yield of amplification was obtained after May-Grunwald staining. Therefore we recommend methyl green for general use in gene expression analysis, especially when handling small amounts of RNA.


Subject(s)
Gene Expression Profiling , Immunohistochemistry , Ribosomal Proteins/genetics , Ethidium/metabolism , Gene Expression/physiology , Humans , Male , Molecular Sequence Data , Prostate/metabolism , RNA/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Ribosomal Proteins/biosynthesis , Staining and Labeling
19.
PLoS One ; 9(10): e109266, 2014.
Article in English | MEDLINE | ID: mdl-25286245

ABSTRACT

Specialized blood cells are generated through the entire life of an organism by differentiation of a small number of hematopoietic stem cells (HSC). There are strictly regulated mechanisms assuring a constant and controlled production of mature blood cells. Although such mechanisms are not completely understood, some factors regulating cell cycle and differentiation have been identified. We have previously shown that Caspase-3 is an important regulator of HSC homeostasis and cytokine responsiveness. p21cip1/waf1 is a known cell cycle regulator, however its role in stem cell homeostasis seems to be limited. Several reports indicate interactions between p21cip1/waf1 and Caspase-3 in a cell type dependent manner. Here we studied the impact of simultaneous depletion of both factors on HSC homeostasis. Depletion of both Caspase-3 and p21cip1/waf1 resulted in an even more pronounced increase in the frequency of hematopoietic stem and progenitor cells. In addition, simultaneous deletion of both genes revealed a further increase of cell proliferation compared to single knock-outs and WT control mice, while apoptosis or self-renewal ability were not affected in any of the genotypes. Upon transplantation, p21cip1/waf1-/- bone marrow did not reveal significant alterations in engraftment of lethally irradiated mice, while Caspase-3 deficient HSPC displayed a significant reduction of blood cell production. However, when both p21cip1/waf1 and Caspase-3 were eliminated this differentiation defect caused by Caspase-3 deficiency was abrogated.


Subject(s)
Caspase 3/deficiency , Caspase 3/genetics , Cell Differentiation/genetics , Cyclin-Dependent Kinase Inhibitor p21/deficiency , Cyclin-Dependent Kinase Inhibitor p21/genetics , Gene Deletion , Hematopoietic Stem Cells/cytology , Animals , Apoptosis/genetics , Cell Proliferation/genetics , Cell Self Renewal/genetics , Female , Gene Knockout Techniques , Hematopoietic Stem Cell Transplantation , Mice , Signal Transduction/genetics
20.
PLoS One ; 9(2): e86910, 2014.
Article in English | MEDLINE | ID: mdl-24586258

ABSTRACT

Small cell lung carcinomas (SCLCs) represent highly aggressive tumors with an overall five-year survival rate in the range of 5 to 10%. Here, we show that four out of five SCLC cell lines reversibly develop a neuron-like phenotype on extracellular matrix constituents such as fibronectin, laminin or thrombospondin upon staurosporine treatment in an RGD/integrin-mediated manner. Neurite-like processes extend rapidly with an average speed of 10 µm per hour. Depending on the cell line, staurosporine treatment affects either cell cycle arrest in G2/M phase or induction of polyploidy. Neuron-like conversion, although not accompanied by alterations in the expression pattern of a panel of neuroendocrine genes, leads to changes in protein expression as determined by two-dimensional gel electrophoresis. It is likely that SCLC cells already harbour the complete molecular repertoire to convert into a neuron-like phenotype. More extensive studies are needed to evaluate whether the conversion potential of SCLC cells is suitable for therapeutic interventions.


Subject(s)
Extracellular Matrix Proteins/metabolism , Small Cell Lung Carcinoma/metabolism , Staurosporine/metabolism , Blotting, Western , Cell Adhesion/physiology , Cell Differentiation/physiology , Cell Line, Tumor , Cell Proliferation , Electrophoresis, Gel, Two-Dimensional , Flow Cytometry , Humans , Reverse Transcriptase Polymerase Chain Reaction
SELECTION OF CITATIONS
SEARCH DETAIL