Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
2.
Thorax ; 68(9): 803-11, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23739138

ABSTRACT

BACKGROUND: Vascular remodelling plays a central role in asthma and chronic obstructive pulmonary disease (COPD). Bradykinin (BK) is a vasoactive proinflammatory peptide mediating acute responses in asthma. We investigated the role of angiogenic factors in relation to BK receptors in asthma and COPD. METHODS: Bronchial biopsies from 33 patients with COPD, 24 old (≥50 years) patients with (≥50 years) asthma, 18 old control smokers, 11 old control non-smokers, 15 young (≤40yrs) patients with (≤40 years) asthma and 10 young control non-smokers were immunostained for CD31, vascular endothelial growth factor-A (VEGF-A), angiogenin and BK receptors (B2R and B1R). Fibroblast and endothelial co-localisation of relevant molecules were performed by immunofluorescence. BK-induced VEGF-A and angiogenin release was studied (ELISA) in bronchial fibroblasts from subjects with asthma and COPD. RESULTS: In bronchial lamina propria of old patients with asthma, CD31 and VEGF-A(+) cell numbers were higher than old control non-smokers (p<0.05). Angiogenin(+), B2R(+) and B1R(+) cell numbers in old patients with asthma were higher than in old control non-smokers, control smokers and patients with COPD (p<0.01). Angiogenin(+) cell numbers were higher in patients with COPD than both old control groups (p<0.05). In all patients with asthma the number of B2R(+) cells was positively related to the numbers of B1R(+) (rs=0.43), angiogenin(+) (rs=0.42) and CD31 cells (rs=0.46) (p<0.01). Angiogenin(+) cell numbers were negatively related to forced expiratory volume in 1 s (rs=-0.415, p=0.008). Double immunofluorescence revealed that CD31 cells of capillary vessels coexpressed B2R and that fibroblasts coexpressed B2R, VEGF-A and angiogenin. BK (10(-6)M) induced significant angiogenin release in fibroblasts from asthma and to a lesser extent in COPD. CONCLUSIONS: Unlike COPD, this study suggests the involvement of BK receptors in bronchial vascular remodelling in asthma.


Subject(s)
Asthma/metabolism , Bronchi/blood supply , Bronchi/metabolism , Capillaries/metabolism , Pulmonary Disease, Chronic Obstructive/metabolism , Receptor, Bradykinin B1/metabolism , Receptor, Bradykinin B2/metabolism , Adaptation, Physiological , Adult , Age Factors , Aged , Biomarkers/metabolism , Capillaries/physiopathology , Case-Control Studies , Endothelial Cells , Female , Fibroblasts , Humans , Male , Middle Aged , Platelet Endothelial Cell Adhesion Molecule-1/metabolism , Ribonuclease, Pancreatic/metabolism , Smoking/metabolism , Vascular Endothelial Growth Factor A/metabolism , Young Adult
3.
Eur J Pharmacol ; 694(1-3): 30-8, 2012 Nov 05.
Article in English | MEDLINE | ID: mdl-22935637

ABSTRACT

Bradykinin-induced interleukin (IL)-8 release should potentially activate neutrophils releasing myeloperoxidase (MPO) and subsequently generating "nitrosative stress". We studied bradykinin-induced expression of bradykinin B(2) receptor and bradykinin- and lipopolysaccharide (LPS)-induced IL-8 release, MPO (marker of neutrophil activation) and 3-nitrotyrosine (3-NT; marker of "nitrosative stress") production in human bronchial epithelial cells BEAS-2B alone or in co-culture with human neutrophils. We evaluated B(2) receptor protein expression in BEAS-2B cells by immunostainings and Western blot analysis, and measured respectively bradykinin- or LPS-induced IL-8 release in BEAS-2B cells and bradykinin- and/or LPS-induced MPO and 3-NT production in BEAS-2B cells co-cultured with human neutrophils by ELISA. In addition, we evaluated bradykinin- and/or LPS-induced 3-NT formation in BEAS-2B cells co-cultured with human neutrophils by immunocytochemistry. Bradykinin up-regulates B(2) receptor expression (P<0.05) and stimulate IL-8 release (P<0.001) in BEAS-2B cells. Either the selective bradykinin B(2) receptor antagonist HOE 140 or the selective bradykinin B(1) receptor antagonist Lys-(des-Arg(9), Leu(8))-bradykinin alone halved IL-8 release and the combination of both drugs suppressed this effect. In BEAS-2B cells co-cultured with human neutrophils bradykinin increased MPO release and 3-NT production compared to BEAS-2B cells with human neutrophils (P<0.001), and the addition of LPS in BEAS-2B cells with human neutrophils and bradykinin induced a further dramatically increase of MPO release and 3-NT formation (P<0.001). Bradykinin and LPS provoked "nitrosative stress", potentially mediated by IL-8, in bronchial epithelium co-cultured with neutrophils suggesting a role for bradykinin in the amplification of chronic airway inflammation via production of "nitrosative stress".


Subject(s)
Bradykinin/pharmacology , Epithelial Cells/drug effects , Interleukin-8/metabolism , Lipopolysaccharides/pharmacology , Neutrophils/metabolism , Reactive Nitrogen Species/metabolism , Receptor, Bradykinin B2/metabolism , Bronchi/cytology , Cell Line , Coculture Techniques , Epithelial Cells/cytology , Epithelial Cells/metabolism , Gene Expression Regulation/drug effects , Humans , Neutrophils/cytology , Neutrophils/drug effects , Peroxidase/metabolism , Tyrosine/analogs & derivatives , Tyrosine/biosynthesis , Tyrosine/metabolism
4.
Haematologica ; 95(8): 1308-16, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20534700

ABSTRACT

BACKGROUND: Usefulness of iron chelation therapy in myelodysplastic patients is still under debate but many authors suggest its possible role in improving survival of low-risk myelodysplastic patients. Several reports have described an unexpected effect of iron chelators, such as an improvement in hemoglobin levels, in patients affected by myelodysplastic syndromes. Furthermore, the novel chelator deferasirox induces a similar improvement more rapidly. Nuclear factor-kappaB is a key regulator of many cellular processes and its impaired activity has been described in different myeloid malignancies including myelodysplastic syndromes. DESIGN AND METHODS: We evaluated deferasirox activity on nuclear factor-kappaB in myelodysplastic syndromes as a possible mechanism involved in hemoglobin improvement during in vivo treatment. Forty peripheral blood samples collected from myelodysplastic syndrome patients were incubated with 50 muM deferasirox for 18h. RESULTS: Nuclear factor-kappaB activity dramatically decreased in samples showing high basal activity as well as in cell lines, whereas no similar behavior was observed with other iron chelators despite a similar reduction in reactive oxygen species levels. Additionally, ferric hydroxyquinoline incubation did not decrease deferasirox activity in K562 cells suggesting the mechanism of action of the drug is independent from cell iron deprivation by chelation. Finally, incubation with both etoposide and deferasirox induced an increase in K562 apoptotic rate. CONCLUSIONS: Nuclear factor-kappaB inhibition by deferasirox is not seen from other chelators and is iron and reactive oxygen species scavenging independent. This could explain the hemoglobin improvement after in vivo treatment, such that our hypothesis needs to be validated in further prospective studies.


Subject(s)
Benzoates/pharmacology , Iron/antagonists & inhibitors , NF-kappa B/antagonists & inhibitors , Reactive Oxygen Species/antagonists & inhibitors , Triazoles/pharmacology , Aged , Aged, 80 and over , Apoptosis/drug effects , Blotting, Western , Deferasirox , Electrophoretic Mobility Shift Assay , Female , Flow Cytometry , Fluorescent Antibody Technique , Humans , Iron/metabolism , Iron Chelating Agents/pharmacology , K562 Cells , Leukemia/metabolism , Leukemia/pathology , Male , Middle Aged , Myelodysplastic Syndromes/metabolism , Myelodysplastic Syndromes/pathology , NF-kappa B/metabolism , Protein Binding/drug effects , Reactive Oxygen Species/metabolism
5.
Blood ; 115(16): 3382-9, 2010 Apr 22.
Article in English | MEDLINE | ID: mdl-20179178

ABSTRACT

Transferrin receptor 2 (TFR2) is a transmembrane protein that is mutated in hemochromatosis type 3. The TFR2 gene is transcribed in 2 main isoforms: the full-length (alpha) and a shorter form (beta). alpha-Tfr2 is the sensor of diferric transferrin, implicated in the modulation of hepcidin, the main regulator of iron homeostasis. The function of the putative beta-Tfr2 protein is unknown. We have developed a new mouse model (KI) lacking beta-Tfr2 compared with Tfr2 knockout mice (KO). Adult Tfr2 KO mice show liver iron overload and inadequate hepcidin levels relative to body iron stores, even though they increase Bmp6 production. KI mice have normal transferrin saturation, liver iron concentration, hepcidin and Bmp6 levels but show a transient anemia at young age and severe spleen iron accumulation in adult animals. Fpn1 is strikingly decreased in the spleen of these animals. These findings and the expression of beta-Tfr2 in wild-type mice spleen suggest a role for beta-Tfr2 in Fpn1 transcriptional control. Selective inactivation of liver alpha-Tfr2 in KI mice (LCKO-KI) returned the phenotype to liver iron overload. Our results strengthen the function of hepatic alpha-Tfr2 in hepcidin activation, suggest a role for extrahepatic Tfr2 and indicate that beta-Tfr2 may specifically control spleen iron efflux.


Subject(s)
Iron/metabolism , Receptors, Transferrin/genetics , Receptors, Transferrin/metabolism , Animals , Antimicrobial Cationic Peptides/metabolism , Blotting, Western , Disease Models, Animal , Gene Expression Profiling , Hemochromatosis/genetics , Hemochromatosis/metabolism , Hepcidins , Liver/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Mutagenesis, Site-Directed , Protein Isoforms/genetics , Protein Isoforms/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Spleen/metabolism
7.
Haematologica ; 93(6): 921-4, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18443273

ABSTRACT

The Wilms' tumor gene WT1 is a reliable marker for minimal residual disease assessment in acute leukemia patients. The study was designed to demonstrate the potential use of WT1 to establish quality of remission in acute leukemia patients for early identification of patients at high risk of relapse. A prospective study based on a quantitative Real-Time PCR (TaqMan) assay in 562 peripheral blood samples collected from 82 acute leukemia patients at diagnosis and during follow-up was established. The evaluation of WT1 in peripheral blood samples after induction chemotherapy can distinguish the continuous complete remission patients from those who obtain only an "apparent" complete remission and who could relapse within a few months. WT1 helps identify patients at high risk of relapse soon after induction chemotherapy allowing post-induction therapy in high risk patients to be intensified.


Subject(s)
Antineoplastic Agents/therapeutic use , Gene Expression Regulation, Neoplastic , Genes, Wilms Tumor , Leukemia, Myeloid, Acute/blood , Leukemia, Myeloid, Acute/therapy , WT1 Proteins/blood , Adult , Gene Expression Profiling , Humans , Leukemia, Myeloid, Acute/diagnosis , Leukemia, Myeloid, Acute/genetics , Middle Aged , Neoplasm Proteins/metabolism , Polymerase Chain Reaction , Prognosis , Recurrence , Risk , Treatment Outcome , WT1 Proteins/physiology
8.
Cancer ; 106(5): 1188-96, 2006 Mar 01.
Article in English | MEDLINE | ID: mdl-16444746

ABSTRACT

BACKGROUND: The objective of this study was to evaluate the ability of the clinically available histone deacetylase (HDAC) inhibitor valproate to enhance the cytotoxicity of the Bcr-Abl inhibitor imatinib in imatinib-resistant cell lines. METHODS: Interactions between imatinib, and valproate have been examined in imatinib-sensitive and -resistant chronic myeloid leukemia (CML)cell lines (K562, KCL-22, CML-T1) and in bone marrow mononuclear cells (MNCs) derived from imatinib-resistant CML patients. RESULTS: In imatinib-sensitive cell lines, cotreatment with imatinib 0.5 muM and valproate 5 microM for 48 hours potently enhanced imatinib-induced growth arrest and apoptosis. In resistant cell lines and in primary MNCs derived from imatinib-rsistant patients, valproate restored sensitivity to the cytotoxic effects of imatinib. Coexposure of cells to valproate and imatinib was associated with repression of several genes involved in Bcr-Abl transformation. In particular, the combination valproate-imatinib downregulated the expression of Bcr-Abl and the antiapoptotic protein Bcl-2, which is particularly overexpressed in imatinib-resistant clones. CONCLUSIONS: Data from this study suggested that administration of the clinically available HDAC inhibitor valproate may be a powerful strategy to enhance cytotoxic effects of imatinib in those patient resistant to imatinib or in which complete cytogenetic remission has been not reached.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Enzyme Inhibitors/pharmacology , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology , Piperazines/pharmacology , Pyrimidines/pharmacology , Valproic Acid/pharmacology , Benzamides , Drug Interactions , Drug Resistance, Neoplasm , Gene Expression Regulation, Neoplastic/drug effects , Genes, abl , Humans , Imatinib Mesylate , Tumor Cells, Cultured
9.
Ann N Y Acad Sci ; 1089: 411-23, 2006 Nov.
Article in English | MEDLINE | ID: mdl-17261784

ABSTRACT

Recent advances in molecular genetics have increased knowledge regarding the mechanisms leading to myelodysplastic syndrome (MDS), secondary acute myeloid leukemia (AML), and therapy-induced MDS. Many genetic defects underlying MDS and AML have been identified thereby allowing the development of new molecular-targeted therapies. Several new classes of drugs have shown promise in early clinical trials and may probably alter the standard of care of these patients in the near future. Among these new drugs are farnesyltransferase inhibitors and receptor tyrosine kinase inhibitors including FLT3 and VEGF inhibitors. These agents have been tested in patients with solid tumors and hematologic malignancies such as AML and MDS. Most of the studies in MDS are still in early stages of development. The DNA hypomethylating compounds azacytidine and decitabine may reduce hypermethylation and induce re-expression of key tumor suppressor genes in MDS. Biochemical compounds with histone deacetylase inhibitory activity, such as valproic acid (VPA), have been tested as antineoplastic agents. Finally, new vaccination strategies are developing in MDS patients based on the identification of MDS-associated antigens. Future therapies will attempt to resolve cytopenias in MDS, eliminate malignant clones, and allow differentiation by attacking specific mechanisms of the disease.


Subject(s)
Chromosome Aberrations , Enzyme Inhibitors/therapeutic use , Immunotherapy , Myelodysplastic Syndromes/genetics , Myelodysplastic Syndromes/therapy , Animals , DNA-Binding Proteins/antagonists & inhibitors , DNA-Binding Proteins/genetics , Epigenesis, Genetic , Farnesyltranstransferase/antagonists & inhibitors , Farnesyltranstransferase/genetics , Humans , MDS1 and EVI1 Complex Locus Protein , Myelodysplastic Syndromes/drug therapy , Proto-Oncogenes/genetics , Receptor Protein-Tyrosine Kinases/antagonists & inhibitors , Receptor Protein-Tyrosine Kinases/genetics , Transcription Factors/antagonists & inhibitors , Transcription Factors/genetics , WT1 Proteins/antagonists & inhibitors , WT1 Proteins/genetics
11.
Cancer ; 101(5): 979-88, 2004 Sep 01.
Article in English | MEDLINE | ID: mdl-15329907

ABSTRACT

BACKGROUND: The objective of the current study was to verify the ability to predict response to imatinib therapy using in vitro assays to evaluate the inhibition of Wilms tumor gene (WT1) expression and colony growth after samples obtained from patients with chronic myelogenous leukemia (CML) before the start of treatment were subjected to short-term incubation with imatinib. METHODS: WT1 transcript levels and colony growth in bone marrow (BM) samples from 23 patients with CML that was later identified as being responsive to imatinib and from 13 patients with CML that was later identified as not being responsive to imatinib were evaluated after incubation of these samples with imatinib at a concentration of 1 microM for 18 hours. In addition, real-time quantitative polymerase chain reaction (RQ-PCR) analysis of WT1 expression was performed during follow-up, and the results were analyzed for associations with cytogenetic response and with BCR/ABL transcript levels as determined using RQ-PCR analysis. RESULTS: Before treatment, it was found that WT1 expression was elevated in BM samples obtained from all patients with CML. WT1 expression and colony growth were reduced significantly after an 18-hour incubation with imatinib in samples obtained from patients who were later identified as responders to treatment, but not in samples obtained from patients who did not experience responses to treatment. Inhibition of WT1 expression in vitro was associated with inhibition of imatinib-induced BCR-ABL tyrosine kinase activity, a finding that also has been made in studies involving certain Philadelphia chromosome (Ph)-positive and Ph-negative cell lines. CONCLUSIONS: Inhibition of WT1 transcript levels after a short period of in vitro exposure of pretherapy BM samples to imatinib was correlated with inhibition of colony growth and may represent the basis for an easy test that is capable of predicting the sensitivity of CML to treatment with imatinib for individual patients.


Subject(s)
Antineoplastic Agents/therapeutic use , Bone Marrow/pathology , Gene Expression Regulation, Neoplastic , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy , Piperazines/therapeutic use , Pyrimidines/therapeutic use , RNA, Messenger/analysis , WT1 Proteins/genetics , Animals , Apoptosis , Benzamides , COS Cells , Chlorocebus aethiops , Drug Resistance, Neoplasm , Follow-Up Studies , Fusion Proteins, bcr-abl/genetics , Humans , Imatinib Mesylate , In Vitro Techniques , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/metabolism , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology , Leukemia, Myeloid, Chronic, Atypical, BCR-ABL Negative/metabolism , Prognosis , Protein-Tyrosine Kinases/antagonists & inhibitors , RNA, Neoplasm/genetics , Reverse Transcriptase Polymerase Chain Reaction , Transfection , WT1 Proteins/antagonists & inhibitors , WT1 Proteins/metabolism
13.
Ann N Y Acad Sci ; 1028: 400-8, 2004 Dec.
Article in English | MEDLINE | ID: mdl-15650265

ABSTRACT

Efforts made during the last few years have helped unravel the complex pathogenesis of the myelodysplastic syndromes (MDS). A large number of studies, made possible by the introduction of newer technologies, have led to major progress in understanding the heterogeneous genetic and biological abnormalities contributing to the development and progression of myelodysplasia. Better insights into these pathogenetic processes will aid the development of newer and more successful therapies for MDS patients. The identification of specific genes involved in the emergence and progression of the myelodysplastic clone has extended biological findings into the clinic. Recently, several clinical trials have used selective compounds to target and inhibit the disrupted signal transduction pathway in myelodysplastic patients. The demonstration of genetic abnormalities present not only in MDS patients but also in acute myeloid leukemia (AML) patients or in chronic myeloproliferative disorders (CMPD) has prompted extension of a number of clinical trials from AML and CMPD to MDS patients. In spite of this, the more complex and heterogeneous pathogenesis underlying the myelodysplastic process is responsible for the often different and in same cases worse clinical results obtained in MDS patients. Finally, the identification of myelodysplasia-associated antigens that may be targeted by an immunotherapeutic approach represents a future perspective in tailored therapy for MDS patients.


Subject(s)
Myelodysplastic Syndromes/diagnosis , Myelodysplastic Syndromes/genetics , Animals , Chromosome Aberrations , Clinical Trials as Topic , Epitopes/chemistry , Humans , Immunotherapy/methods , Myelodysplastic Syndromes/therapy , Signal Transduction , Translocation, Genetic , WT1 Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...