Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 37
Filter
1.
Mol Carcinog ; 53(6): 498-504, 2014 Jun.
Article in English | MEDLINE | ID: mdl-23192887

ABSTRACT

The small molecule inhibitor of the MDM2/p53 interaction Nutlin-3 is a promising anti-cancer agent, which exhibits activity against a variety of cancers, including acute myeloid leukemia (AML). Previous studies have shown that Nutlin-3 variably induces apoptosis and cell cycle arrest in cancer cells while it shows low/absent cytotoxicity in normal cells. However, the reason for the selective pro-apoptotic activity in cancer cells with respect to normal counterparts is incompletely understood. In this study, we have compared the induction of several known target genes of p53 in two p53(wild-type) AML cell lines, OCI-AML3 and MOLM, in comparison with primary normal peripheral blood mononuclear cells (PBMC). Among several p53-target genes activated both in AML cell lines and normal PBMC (BBC3, BAX, MDM2, FAS, CDKN1A, GDF15, GADD45A, TNFRSF10B, TP53I3/PIG3), only TP53I3/PIG3 was selectively activated in MOLM and OCI-AML3, but not in PBMC. The important role of TP53I3/PIG3 in mediating the apoptotic activity of Nutlin-3 was underlined by knock-down experiments with siRNA specific for TP53I3/PIG3, which resulted in a significant decrease in the pro-apoptotic activity of Nutlin-3.


Subject(s)
Gene Expression Regulation, Leukemic/drug effects , Imidazoles/pharmacology , Intracellular Signaling Peptides and Proteins/genetics , Leukemia, Myeloid, Acute/genetics , Piperazines/pharmacology , Proto-Oncogene Proteins/genetics , Apoptosis/drug effects , Cell Cycle Checkpoints/drug effects , Cell Line, Tumor , Cell Survival/drug effects , Gene Knockdown Techniques , Humans , Imidazoles/toxicity , Leukocytes, Mononuclear/drug effects , Leukocytes, Mononuclear/metabolism , Piperazines/toxicity , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism
2.
Invest New Drugs ; 30(2): 803-7, 2012 Apr.
Article in English | MEDLINE | ID: mdl-20827501

ABSTRACT

A disseminated model of non-Hodgkin's lymphoma with prevalent liver metastasis was generated by intraperitoneal (i.p.) injection of EBV(+) B lymphoblastoid SKW6.4 in nude-SCID mice. The survival of SKW6.4 xenografts (median survival = 27 days) was significantly improved when hyaluronan scaffolds embedded with mesenchimal stem cells (MSC) were implanted in the abdominal area 4 days after SKW6.4 injection (median survival = 39.5 days). Mice implanted with MSC showed a significant improvement of hepatic functionality in lymphoma xenografts, as demonstrated by measurement of serum ALT/AST levels. Co-culture of MSC with lymphoma cells enhanced the release of hepatocyte growth factor (HGF) by MSC. These data suggest that hyaluronan-embedded MSC exert anti-lymphoma activity by ameliorating hepatic functionality.


Subject(s)
Liver Neoplasms/prevention & control , Liver/metabolism , Lymphoma, Non-Hodgkin/surgery , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells/metabolism , Alanine Transaminase/blood , Animals , Aspartate Aminotransferases/blood , Biomarkers/blood , Cell Communication , Cell Line, Tumor , Cell Survival , Coculture Techniques , Hepatocyte Growth Factor/metabolism , Humans , Hyaluronic Acid , Liver/pathology , Liver Neoplasms/metabolism , Liver Neoplasms/secondary , Lymphoma, Non-Hodgkin/metabolism , Lymphoma, Non-Hodgkin/pathology , Mice , Mice, Nude , Mice, SCID , Time Factors , Tissue Scaffolds , Xenograft Model Antitumor Assays
3.
Invest New Drugs ; 30(1): 417-22, 2012 Feb.
Article in English | MEDLINE | ID: mdl-20953816

ABSTRACT

The multi-kinase inhibitor dasatinib induced a variable but significant decrease of viability in both p53(wild-type) (EHEB, JVM-2, JVM-3) and p53(mutated) (MEC-1, MEC-2, BJAB) prolymphocytic B leukemic cells, due to a combination of cell cycle block in G1 and apoptosis. Antibody phospho-kinase array analysis revealed that dasatinib inhibited the phosphorylation of various kinases, including ERK1/2 and p38/MAPK as well as of STAT3 transcription factors, in both p53(wild-type) and p53(mutated) cells. Therefore, dasatinib might offer a novel therapeutic strategy not only for p53(wild-type), but also for p53(mutated) B malignancies that have the worst prognosis and urgently need innovative therapeutic approaches.


Subject(s)
Antineoplastic Agents/pharmacology , B-Lymphocytes/drug effects , Granulocyte Precursor Cells/drug effects , Leukemia, Prolymphocytic, B-Cell/metabolism , Mutation , Protein Kinase Inhibitors/pharmacology , Pyrimidines/pharmacology , Thiazoles/pharmacology , Tumor Suppressor Protein p53/genetics , Apoptosis/drug effects , B-Lymphocytes/metabolism , B-Lymphocytes/pathology , Cell Line, Tumor , Cell Survival/drug effects , Dasatinib , G1 Phase Cell Cycle Checkpoints/drug effects , Granulocyte Precursor Cells/metabolism , Granulocyte Precursor Cells/pathology , Humans , Leukemia, Prolymphocytic, B-Cell/genetics , Leukemia, Prolymphocytic, B-Cell/pathology , Mitogen-Activated Protein Kinase 1/antagonists & inhibitors , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/antagonists & inhibitors , Mitogen-Activated Protein Kinase 3/metabolism , Phosphorylation , STAT3 Transcription Factor/antagonists & inhibitors , STAT3 Transcription Factor/metabolism , Time Factors , p38 Mitogen-Activated Protein Kinases/antagonists & inhibitors , p38 Mitogen-Activated Protein Kinases/metabolism
4.
Invest New Drugs ; 30(1): 405-7, 2012 Feb.
Article in English | MEDLINE | ID: mdl-20714918

ABSTRACT

A new single-chain fragment variable (scFv) to TRAIL-R2 receptor produced as minibody (MB2.23) was characterized for anti-lymphoma activity in vivo. For this purpose, a disseminated lymphoma model was generated by intraperitoneal inoculation of BJAB cells in severe combined immunodeficiency mice. Two weekly injections with MB2.23 (10 mg/kg) were able to significantly increase the median survival time of lymphoma-bearing animals with respect to the vehicle-treated control mice, providing a rationale for further investigating the use of MB2.23 in anticancer therapy.


Subject(s)
Immunotherapy/methods , Lymphoma, B-Cell/therapy , Receptors, TNF-Related Apoptosis-Inducing Ligand/immunology , Single-Chain Antibodies/administration & dosage , Animals , Humans , Injections, Intraperitoneal , Lymphoma, B-Cell/immunology , Lymphoma, B-Cell/pathology , Mice , Mice, SCID , Time Factors , Xenograft Model Antitumor Assays
5.
J Cell Physiol ; 226(9): 2279-86, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21660951

ABSTRACT

The soluble member of the TNF-R superfamily osteoprotegerin (OPG) is abundantly released under basal conditions by both mesenchymal stem cells (MSC) and fibroblasts and by endothelial cells upon stimulation with inflammatory cytokines. Since MSC, fibroblasts and endothelial cells represent key elements of the normal and tumor microenvironment and express detectable levels of surface TRAIL receptors, we investigated the effect of TRAIL on OPG release. Unexpectedly, recombinant TRAIL decreased the spontaneous OPG release in all cell types examined. Moreover, TRAIL decreased OPG release also in stromal cells co-cultured with lymphoma cells and counteracted the OPG induction by TN-alpha in HUVEC and MSC. Such down-regulation was not due to a masking effect in the ELISA quantification of the OPG released in the culture supernatants due to binding of OPG to its ligands (TRAIL and RANKL), as demonstrated by competition experiments with recombinant TRAIL and by the lack of RANKL release/induction. In addition, OPG down-regulation was not due to induction of cytotoxic effects by TRAIL, since the degree of apoptosis in response to TRAIL was negligible in all primary cell types. With regards to the possible molecular mechanism accounting for the down-regulation of OPG release by TRAIL, we found that treatment of MSC with TRAIL significantly decreased the phosphorylation levels of p38/MAPK. There is a suggestion that this pathway is involved in the stabilization of OPG mRNA. In this respect, the ability of TRAIL to decrease the release of OPG, in the absence of cell cytotoxicity, was mimicked by the p38/MAPK inhibitor SB203580.


Subject(s)
Down-Regulation/drug effects , Osteoprotegerin/metabolism , TNF-Related Apoptosis-Inducing Ligand/pharmacology , Bone Marrow Cells/cytology , Bone Marrow Cells/drug effects , Bone Marrow Cells/enzymology , Cell Death/drug effects , Cells, Cultured , Coculture Techniques , Endothelial Cells/cytology , Endothelial Cells/drug effects , Endothelial Cells/enzymology , Enzyme Activation/drug effects , Enzyme-Linked Immunosorbent Assay , Fibroblasts/drug effects , Fibroblasts/metabolism , Humans , MAP Kinase Signaling System/drug effects , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/drug effects , Mesenchymal Stem Cells/enzymology , Osteoprotegerin/genetics , Protein Binding/drug effects , Recombinant Proteins/metabolism , Stromal Cells/drug effects , Stromal Cells/metabolism , Tumor Necrosis Factor-alpha/pharmacology , p38 Mitogen-Activated Protein Kinases/metabolism
6.
Mol Cell Endocrinol ; 331(1): 136-42, 2011 Jan 01.
Article in English | MEDLINE | ID: mdl-20832449

ABSTRACT

Although serum osteoprotegerin (OPG) is significantly increased in diabetic subjects, its potential role in beta cell dysfunction has not been investigated. This study aimed to assess the effect of full-length OPG administered in vivo in mice on pancreatic islet structure and function and its interaction with the renin-angiotensin system (RAS). OPG-treated mice showed increased islet monocyte/macrophage infiltration, fibrosis and apoptosis with reduction of islet function. The remodeling of islet architecture was associated with increased pancreatic expression of components of the RAS, growth factor genes (transforming growth factor ß and connective tissue growth factor) and inflammatory molecules (monocyte chemotactic protein-1 and vascular adhesion molecule type 1). Prevention of these changes with improvement of insulin secretion was observed in ramipril treated animals. Our data suggest that OPG might play an important role in promoting beta cell dysfunction and that the upregulation of the local RAS represents one possible mechanism responsible for the OPG-induced beta cell dysfunction.


Subject(s)
Islets of Langerhans/metabolism , Islets of Langerhans/pathology , Osteoprotegerin/pharmacology , Animals , Apoptosis/drug effects , Blood Glucose/metabolism , Blood Pressure/drug effects , Body Weight/drug effects , Cell Lineage/drug effects , Cell Movement/drug effects , Chemokine CCL2/genetics , Chemokine CCL2/metabolism , Connective Tissue Growth Factor/genetics , Connective Tissue Growth Factor/metabolism , Fibrosis , Gene Expression Regulation/drug effects , Humans , Insulin/metabolism , Islets of Langerhans/drug effects , Macrophages/cytology , Macrophages/drug effects , Mice , Monocytes/cytology , Monocytes/drug effects , Organ Size/drug effects , Peptidyl-Dipeptidase A/genetics , Peptidyl-Dipeptidase A/metabolism , Receptor, Angiotensin, Type 1/genetics , Receptor, Angiotensin, Type 1/metabolism , Systole/drug effects , Transforming Growth Factor beta/genetics , Transforming Growth Factor beta/metabolism , Vascular Cell Adhesion Molecule-1/genetics , Vascular Cell Adhesion Molecule-1/metabolism
7.
Atherosclerosis ; 215(2): 452-8, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21122855

ABSTRACT

OBJECTIVE: Tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) exhibits biological activity on vascular cells in vitro. Rapid variation of circulating TRAIL levels occurs during acute coronary ischemia, suggesting that biological pathways involving TRAIL may be activated during ischemic heart disease. However, whether differential levels of soluble TRAIL in normal individuals are associated with adverse health outcomes has not been investigated. We tested the hypothesis that TRAIL levels predict mortality in a population based sample of community dwelling men and women. METHODS: Plasma TRAIL level was measured by ELISA at baseline in 1282 adults (mean age 68 years) enrolled in the InCHIANTI study. Vital status was ascertained over the six-year follow-up. RESULTS: In multivariable Cox regression analysis adjusted for potential confounders including prevalent cardiovascular diseases (CVD), ankle-brachial index, electrocardiogram abnormalities, and inflammatory markers, baseline TRAIL levels were inversely related to all-cause mortality (p=0.008). In stratified analyses, the prognostic effect of TRAIL level was strong and highly significant in participants with prevalent CVD (N=321), (lowest versus highest quartile: HR 3.1; 95% CI 1.5-6.5) while it was negligible in those free of CVD (p value for the interaction term between CVD status and TRAIL levels=0.038). Similar findings were obtained when CVD mortality was considered as the outcome of interest. CONCLUSIONS: In older patients with CVD, low levels of TRAIL were associated with increased risk of death over a period of 6 years. Lower concentration of circulating TRAIL may be related to the clinical evolution of older adults with CVD.


Subject(s)
Cardiovascular Diseases/mortality , TNF-Related Apoptosis-Inducing Ligand/blood , Adult , Aged , Ankle Brachial Index , Female , Follow-Up Studies , Humans , Italy/epidemiology , Male , Middle Aged
8.
PLoS One ; 5(6): e11140, 2010 Jun 16.
Article in English | MEDLINE | ID: mdl-20585401

ABSTRACT

BACKGROUND: Although multimodality treatment can induce high rate of remission in many subtypes of non-Hodgkin's lymphoma (NHL), significant proportions of patients relapse with incurable disease. The effect of human bone marrow (BM) mesenchymal stem cells (MSC) on tumor cell growth is controversial, and no specific information is available on the effect of BM-MSC on NHL. METHODOLOGY/PRINCIPAL FINDINGS: The effect of BM-MSC was analyzed in two in vivo models of disseminated non-Hodgkin's lymphomas with an indolent (EBV(-) Burkitt-type BJAB, median survival = 46 days) and an aggressive (EBV(+) B lymphoblastoid SKW6.4, median survival = 27 days) behavior in nude-SCID mice. Intra-peritoneal (i.p.) injection of MSC (4 days after i.p. injection of lymphoma cells) significantly increased the overall survival at an optimal MSC:lymphoma ratio of 1:10 in both xenograft models (BJAB+MSC, median survival = 58.5 days; SKW6.4+MSC, median survival = 40 days). Upon MSC injection, i.p. tumor masses developed more slowly and, at the histopathological observation, exhibited a massive stromal infiltration coupled to extensive intra-tumor necrosis. In in vitro experiments, we found that: i) MSC/lymphoma co-cultures modestly affected lymphoma cell survival and were characterized by increased release of pro-angiogenic cytokines with respect to the MSC, or lymphoma, cultures; ii) MSC induce the migration of endothelial cells in transwell assays, but promoted endothelial cell apoptosis in direct MSC/endothelial cell co-cultures. CONCLUSIONS/SIGNIFICANCE: Our data demonstrate that BM-MSC exhibit anti-lymphoma activity in two distinct xenograft SCID mouse models of disseminated NHL.


Subject(s)
Hematopoietic Stem Cells/cytology , Lymphoma, Non-Hodgkin/pathology , Mesenchymal Stem Cells/cytology , Animals , Coculture Techniques , Humans , Mice , Mice, Nude , Mice, SCID , Transplantation, Heterologous
10.
J Rheumatol ; 37(7): 1461-6, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20472926

ABSTRACT

OBJECTIVE: To assess the relationship between serum concentrations of tumor necrosis factor (TNF)-related apoptosis-inducing ligand (TRAIL) and osteoprotegerin (OPG) and the therapeutic response to disease-modifying antirheumatic drugs (DMARD) in patients with early rheumatoid arthritis (RA). METHODS: Circulating levels of TRAIL and its soluble receptor OPG were measured by ELISA in paired serum samples obtained from 66 patients with early RA at their first visit (baseline) and after 1 year of therapy. Levels of TRAIL and OPG were analyzed in relation to the clinical response, defined by the 28-joint count Disease Activity Score (DAS28). RESULTS: Both serum TRAIL and OPG increased after DMARD therapy. Baseline levels of TRAIL, but not OPG, were significantly higher (p < 0.05) in the patients that achieved a clinical response by DAS28 after 1 year of therapy, versus patients without clinical response to DMARD. Baseline serum levels of TRAIL were higher (p < 0.01) in rheumatoid factor-negative patients. CONCLUSION: Our data suggest that the basal level of circulating TRAIL is an important determinant in the therapeutic response to DMARD in patients with early RA.


Subject(s)
Antirheumatic Agents/therapeutic use , Arthritis, Rheumatoid , TNF-Related Apoptosis-Inducing Ligand/blood , Aged , Arthritis, Rheumatoid/blood , Arthritis, Rheumatoid/drug therapy , Arthritis, Rheumatoid/immunology , Biomarkers/blood , Disease Progression , Female , Humans , Middle Aged , Osteoprotegerin/blood , Osteoprotegerin/immunology , Severity of Illness Index , TNF-Related Apoptosis-Inducing Ligand/immunology , Treatment Outcome
11.
Atherosclerosis ; 211(1): 333-6, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20233617

ABSTRACT

BACKGROUND: This study was designed to evaluate the potential role of metalloproteinase 2 (MMP2) in promoting the cleavage of TNF-related apoptosis inducing ligand (TRAIL), whose circulating levels are decreased after acute myocardial infarction (AMI). METHODS: The levels of MMP2 and of tissue inhibitor of metalloprotease 2 (TIMP2), as well as of TRAIL, were measured by ELISA in the serum of AMI patients and in the culture supernatant of endothelial cells. RESULTS: In AMI patients the serum levels of TRAIL showed a significant inverse correlation with the MMP2/TIMP2 ratio. In vitro MMP2 induced the cleavage of recombinant TRAIL and inactivated its ability of inducing apoptosis. Moreover, exposure of endothelial cells to TNF-alpha increased the MMP2/TIMP2 ratio in the culture supernatant. CONCLUSIONS: An impaired MMP2/TIMP2 ratio might be involved in the decreased levels of circulating TRAIL observed after AMI.


Subject(s)
Anterior Wall Myocardial Infarction/blood , Matrix Metalloproteinase 2/metabolism , TNF-Related Apoptosis-Inducing Ligand/metabolism , Apoptosis/drug effects , Humans , Recombinant Proteins/metabolism , TNF-Related Apoptosis-Inducing Ligand/blood , Tissue Inhibitor of Metalloproteinase-2/blood , Tumor Necrosis Factor-alpha/pharmacology
12.
Rheumatology (Oxford) ; 49(6): 1173-6, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20299382

ABSTRACT

OBJECTIVE: To investigate the role of the TNF-related apoptosis-inducing ligand-osteoprotegerin (TRAIL-OPG) system in the pathogenesis of limited SSc (lSSc). METHODS: Circulating levels of TRAIL and of its soluble receptor OPG were measured by ELISA in serum samples obtained from 50 lSSc patients and 50 healthy controls. RESULTS: TRAIL serum levels in lSSc patients were similar to those of healthy controls, whereas the OPG serum levels were significantly increased (P < 0.0001). According to different subgroups of lSSc patients, TRAIL was not statistically different between each group and healthy controls; concerning OPG, the statistically different value was also maintained when comparing each single lSSc group with the whole control population. CONCLUSIONS: OPG serum levels, but not TRAIL, are elevated in lSSc patients. Since OPG binding to TRAIL inhibits TRAIL-TRAIL receptor interaction, the relative concentrations of these two molecules in the local micro-environment has to be considered. In this setting, OPG increase in lSSc patients may produce a detrimental effect by counteracting the vasoprotective activity of TRAIL. The TRAIL : OPG ratio and their relative levels of expression in lSSc patients should be taken into consideration as a possible novel marker of vascular damage.


Subject(s)
Endothelial Cells/metabolism , Osteoprotegerin/metabolism , RANK Ligand/metabolism , Scleroderma, Limited/metabolism , TNF-Related Apoptosis-Inducing Ligand/metabolism , Tumor Necrosis Factor-alpha/metabolism , Adult , Aged , Apoptosis , Apoptosis Inducing Factor , Biomarkers/metabolism , Case-Control Studies , Female , Humans , Male , Middle Aged , Statistics as Topic , Time Factors , White People
13.
Cell Mol Life Sci ; 67(8): 1307-14, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20063037

ABSTRACT

The number of circulating mesenchymal stem cells (MSC), analyzed after acute myocardial infarction (AMI), was lower in AMI patients who developed heart failure (HF) in the follow-up. Conversely, the circulating levels of tumor necrosis factor (TNF)-alpha, and osteoprotegerin (OPG) were higher in AMI patients who developed HF with respect to the patients who did not develop HF. In vitro exposure to TNF-alpha enhanced the migration of MSC in response to TNF-related apoptosis-inducing ligand (TRAIL) and significantly increased the release of OPG by endothelial cells. On the contrary, OPG dose-dependently neutralized the in vitro pro-migratory activity of TRAIL. Thus, TNF-alpha exhibits opposite effects on MSC migration driven by TRAIL: it is capable of potentiating MSC migration as well as of inhibiting MSC migration as an indirect consequence of OPG induction, which might result in a suboptimal recruitment of circulating MSC after AMI in those patients who develop HF in the follow-up.


Subject(s)
Cell Movement , Mesenchymal Stem Cells/metabolism , Myocardial Infarction/metabolism , TNF-Related Apoptosis-Inducing Ligand/pharmacology , Tumor Necrosis Factor-alpha/pharmacology , Acute Disease , Case-Control Studies , Enzyme-Linked Immunosorbent Assay , Female , Flow Cytometry , Follow-Up Studies , Humans , Male , Mesenchymal Stem Cells/cytology , Middle Aged , Myocardial Infarction/pathology , Osteoprotegerin/metabolism
14.
J Vasc Res ; 47(3): 252-61, 2010.
Article in English | MEDLINE | ID: mdl-19907187

ABSTRACT

BACKGROUND/AIMS: Since elevated plasma levels of osteoprotegerin (OPG) represent a risk factor for death and heart failure in patients affected by diabetes mellitus and coronary artery disease, this study aimed to elucidate potential roles of OPG in the pathogenesis of atherosclerosis. METHODS AND RESULTS: Recombinant human full-length OPG, used at concentrations comparable to the elevated levels found in the serum of diabetic patients, significantly increased the proliferation rate of rodent vascular smooth muscle cells (VSMC). To mimic the moderate chronic elevation of OPG observed in diabetic patients, low doses (1 microg/mouse) of full-length human OPG were injected intraperitoneally every 3 weeks in diabetic apolipoprotein E (apoE)-null mice. The group of animals treated for 12 weeks with recombinant OPG showed a small increase in the total aortic plaque area at necropsy in comparison to vehicle-treated animals. Importantly, while no differences in the amount of interstitial collagen or the degree of macrophage infiltration were observed between OPG-treated and vehicle-treated apoE-null diabetic animals, a significant increase in the number of alpha-actin-positive smooth muscle cells was observed in the plaques of OPG-treated mice. CONCLUSIONS: Our data suggest that OPG promotes VSMC proliferation and might be directly involved in pathogenetic aspects of atherosclerosis.


Subject(s)
Atherosclerosis/etiology , Cell Proliferation , Diabetes Mellitus, Experimental/complications , Diabetic Angiopathies/etiology , Muscle, Smooth, Vascular/metabolism , Myocytes, Smooth Muscle/metabolism , Osteoprotegerin/metabolism , Angiopoietin-2/genetics , Animals , Apolipoproteins E/deficiency , Apolipoproteins E/genetics , Atherosclerosis/metabolism , Atherosclerosis/pathology , Cells, Cultured , Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Experimental/pathology , Diabetic Angiopathies/metabolism , Diabetic Angiopathies/pathology , Disease Models, Animal , Humans , Injections, Intraperitoneal , Mice , Mice, Knockout , Muscle, Smooth, Vascular/pathology , Myocytes, Smooth Muscle/pathology , Osteoprotegerin/administration & dosage , RNA, Messenger/metabolism , Rats , Recombinant Proteins/metabolism
15.
Atherosclerosis ; 210(1): 274-7, 2010 May.
Article in English | MEDLINE | ID: mdl-20015493

ABSTRACT

BACKGROUND: This study was designed to evaluate the biological significance of simultaneous changes in the circulating levels of osteoprotegerin (OPG) and TNF-related apoptosis inducing ligand (TRAIL) in patients with coronary artery disease (CAD), and, in particular, with acute myocardial infarction (AMI). METHODS: Total levels of OPG and TRAIL were measured by ELISA in patients with AMI (n=113), unstable angina (UA, n=21) and healthy controls (n=120). RESULTS: Since OPG was elevated during the acute phase (first 12-24-48h) after AMI and in patients with UA with respect to healthy controls, while TRAIL was decreased in acute AMI patients, CAD patients were characterized by an increased OPG/TRAIL ratio. Moreover, the OPG/TRAIL ratio was significantly (p<0.05) higher in the acute AMI patients who developed heart failure (HF) than in those who did not develop HF in the follow-up. CONCLUSIONS: An impaired OPG/TRAIL ratio after AMI is related to a higher risk of HF.


Subject(s)
Myocardial Infarction/blood , Osteoprotegerin/blood , TNF-Related Apoptosis-Inducing Ligand/blood , Angina, Unstable/blood , Coronary Disease/blood , Coronary Disease/complications , Enzyme-Linked Immunosorbent Assay , Female , Heart Failure/etiology , Humans , Male , Myocardial Infarction/complications
16.
PLoS One ; 4(2): e4442, 2009.
Article in English | MEDLINE | ID: mdl-19221598

ABSTRACT

BACKGROUND: Since soluble TRAIL exhibits anti-inflammatory and anti-atherosclerotic activities both in vitro and in animal models, this study was designed to assess the relationship between the serum levels of TRAIL and clinical outcomes in patients with acute myocardial infarction (AMI). METHODOLOGY/PRINCIPAL FINDINGS: Levels of TRAIL were measured by ELISA in serial serum samples obtained from 60 patients admitted for AMI, both during hospitalization and in a follow-up of 12 months, as well as in 60 healthy control subjects. Serum levels of TRAIL were significantly decreased in patients with AMI at baseline (within 24 hours from admission), compared with healthy controls, and showed a significant inverse correlation with a series of negative prognostic markers, such as CK, CK-MB and BNP. TRAIL serum levels progressively increased at discharge, but normalized only at 6-12 months after AMI. Of note, low TRAIL levels at the patient discharge were associated with increased incidence of cardiac death and heart failure in the 12-month follow-up, even after adjustment for demographic and clinical risk parameters (hazard ratio [HR] of 0.93 [95% CI, 0.89 to 0.97]; p = 0.001). CONCLUSIONS/SIGNIFICANCE: Although the number of patients studied was limited, our findings indicate for the first time that circulating TRAIL might represent an important predictor of cardiovascular events, independent of conventional risk markers.


Subject(s)
Myocardial Infarction , TNF-Related Apoptosis-Inducing Ligand/blood , Aged , Animals , Biomarkers/blood , Creatine Kinase, MB Form/blood , Humans , Male , Middle Aged , Myocardial Infarction/blood , Myocardial Infarction/diagnosis , Pilot Projects , Prognosis , Risk Factors
17.
Blood ; 113(18): 4300-8, 2009 Apr 30.
Article in English | MEDLINE | ID: mdl-19190243

ABSTRACT

The small molecule inhibitor of the MDM2/p53 interaction Nutlin-3 significantly up-regulated the steady-state mRNA and protein levels of Notch1 in TP53(wild-type) (OCI, SKW6.4) but not in TP53(deleted) (HL-60) or TP53(mutated) (BJAB) leukemic cell lines. A direct demonstration that NOTCH1 was a transcriptional target of p53 in leukemic cells was obtained in experiments carried out with siRNA for p53. Moreover, inhibition of Notch1 expression using Notch1-specific siRNA significantly increased cytotoxicity in TP53(wild-type) leukemic cells. Of note, Nutlin-3 up-regulated Notch1 expression also in primary TP53(wild-type) B-chronic lymphocytic leukemia (B-CLL) cells and the combined use of Nutlin-3 plus pharmacological gamma-secretase inhibitors of the Notch signaling showed a synergistic cytotoxicity in both TP53(wild-type) leukemic cell lines and primary B-CLL cells. A potential drawback of gamma-secretase inhibitors was their ability to enhance osteoclastic maturation of normal circulating preosteoclasts induced by RANKL + M-CSF. Notwithstanding, Nutlin-3 completely suppressed osteoclastogenesis irrespective of the presence of gamma-secretase inhibitors. Taken together, these data indicate that the p53-dependent up-regulation of Notch1 in response to Nutlin-3 represents an antiapoptotic feedback mechanism able to restrain the potential therapeutic efficacy of Nutlin-3 in hematologic malignancies. Therefore, therapeutic combinations of Nutlin-3 + gamma-secretase inhibitors might potentiate the cytotoxicity of Nutlin-3 in p53(wild-type) leukemic cells.


Subject(s)
Apoptosis , Feedback, Physiological , Imidazoles/pharmacology , Leukemia, Lymphocytic, Chronic, B-Cell/metabolism , Piperazines/pharmacology , Receptor, Notch1/metabolism , Adult , Aged , Aged, 80 and over , Amyloid Precursor Protein Secretases/antagonists & inhibitors , Amyloid Precursor Protein Secretases/metabolism , Blotting, Western , Carbamates/pharmacology , Cell Differentiation , Dipeptides/pharmacology , Drug Synergism , Drug Therapy, Combination , Female , HL-60 Cells , Humans , Leukemia, Lymphocytic, Chronic, B-Cell/genetics , Leukemia, Lymphocytic, Chronic, B-Cell/pathology , Male , Middle Aged , Osteoclasts/cytology , Osteoclasts/metabolism , RNA, Messenger/metabolism , Receptor, Notch1/genetics , Reverse Transcriptase Polymerase Chain Reaction , Stereoisomerism , Tumor Suppressor Protein p53/antagonists & inhibitors , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism , Up-Regulation
18.
Rheumatology (Oxford) ; 48(3): 293-8, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19168833

ABSTRACT

OBJECTIVE: Complement activation products contribute to a large number of inflammatory diseases, including RA. We have investigated whether osteoprotegerin (OPG) may concur with the soluble terminal complement complex (SC5b-9) to the inflammatory cascade characterizing RA. METHODS: Levels of SC5b-9 and OPG in the plasma and SF of patients with active RA were determined by ELISA. The presence of SC5b-9 and OPG in RA synovial lesions was analysed by immunohistochemistry. Cultured endothelial cells were used for in vitro leucocyte/endothelial cell adhesion assays. In addition, endothelial cells were exposed to SC5b-9 in order to evaluate the effects on the production of OPG protein, as well as the activation of the OPG promoter. RESULTS: Patients affected by active RA are characterized by elevated levels of both SC5b-9 and OPG in plasma and/or SF. Of note, we have observed a co-localization of SC5b-9 and OPG in endothelial cells of post-capillary venules of RA synovial lesions. Data on endothelial cell cultures showed that exposure to SC5b-9 induced the up-regulation of OPG expression/release, stimulating the transcriptional activity of the OPG promoter, and synergized with TNF-alpha in up-regulating OPG production. CONCLUSIONS: Our findings demonstrate that SC5b-9 induces OPG production by endothelial cells and we propose that the SC5b-9-mediated up-regulation of OPG may be an important mechanism whereby complement contributes in promoting and/or enhancing the inflammation in RA.


Subject(s)
Arthritis, Rheumatoid/metabolism , Complement Membrane Attack Complex/physiology , Endothelial Cells/metabolism , Osteoprotegerin/biosynthesis , Adult , Aged , Cell Adhesion/physiology , Cells, Cultured , Complement Membrane Attack Complex/pharmacology , Dose-Response Relationship, Drug , Endothelial Cells/drug effects , Endothelium, Vascular/metabolism , Female , Humans , Male , Middle Aged , Neutrophil Infiltration/physiology , Neutrophils/physiology , Synovial Membrane/metabolism , Up-Regulation/drug effects
19.
J Cell Physiol ; 218(1): 199-204, 2009 Jan.
Article in English | MEDLINE | ID: mdl-18780288

ABSTRACT

Little is known on the ability of different epithelia to release soluble TNF-related apoptosis-inducing ligand (TRAIL) and the relevance of TRAIL secretion by epithelial cells is still incompletely understood. On these bases, we have measured the concentration of soluble TRAIL by ELISA in the conjunctival sac fluid. It was the highest ever detected in a biological fluid (mean value of 26,800 pg/ml), being approximately 20-fold greater than that found in human saliva and >200-fold greater than that detected in human serum. On the other hand, osteoprotegerin, the soluble decoy receptor of TRAIL, was almost undetectable in the conjunctival sac fluid. Of note, the levels of soluble TRAIL measured in conjunctival sac fluid were in the range able to induce in vitro apoptosis of lymphoma cells. By in situ immunohistochemistry, TRAIL protein expression was predominantly detected in the corneal epithelium and, to a less extent, in the conjunctival epithelium. By flow cytometry analysis, membrane-associated TRAIL was documented in isolated corneal epithelial cells obtained from patients undergoing photorefractive keratectomy (PRK). The key contribution provided by corneal epithelium to the production of soluble TRAIL was underscored in time-course experiments, in which a marked decrease of the levels of soluble TRAIL in the conjunctival sac fluid was demonstrated 1 day after PRK followed by a progressive recovery at days 5-30 after PRK. Taken together, our findings strongly support a major role of soluble TRAIL in protecting cornea and conjunctiva from tumor formation and/or invasion.


Subject(s)
Conjunctiva/metabolism , TNF-Related Apoptosis-Inducing Ligand/metabolism , Apoptosis/drug effects , Apoptosis/physiology , Body Fluids/metabolism , Cell Line, Tumor , Cornea/cytology , Cornea/metabolism , Epithelial Cells/cytology , Epithelial Cells/metabolism , Eye Neoplasms/prevention & control , Humans , In Vitro Techniques , Osteoprotegerin/metabolism , Photorefractive Keratectomy , Recombinant Proteins/pharmacology , Solubility , TNF-Related Apoptosis-Inducing Ligand/pharmacology
20.
Stem Cells ; 26(11): 2955-63, 2008 Nov.
Article in English | MEDLINE | ID: mdl-18772312

ABSTRACT

Adult multipotent stromal cells (MSCs), also known as mesenchymal stem cells, represent an important source of cells for the repair of a number of damaged tissues. Both bone marrow (BM)-derived and amniotic MSCs expressed detectable surface levels of two (tumor necrosis factor-related apoptosis-inducing ligand receptor 2 [TRAIL-R2] and TRAIL-R4) of four transmembrane TRAIL receptors. Although the best-characterized activity of TRAIL-R2 is the transduction of apoptotic signals, neither recombinant TRAIL (rTRAIL) nor infection with an adenovirus-expressing TRAIL induced cytotoxic effects on MSCs. Moreover, whereas rTRAIL did not affect proliferation or differentiation of MSCs along the osteogenic and adipogenic lineages, it significantly promoted the migration of human MSCs in range of concentrations comparable to that of soluble TRAIL in human plasma (100 pg/ml). Since rTRAIL induced the rapid phosphorylation of extracellular signal-regulated kinase 1/2 (ERK1/2) in MSC cultures and pretreatment with pharmacological inhibitors of the ERK1/2 pathway efficiently counteracted the rTRAIL-induced human MSC migration, these data indicate that ERK1/2 is involved in mediating the ability of rTRAIL to stimulate MSC migration. Taking into consideration that the soluble factors able to induce MSC migration have not been extensively characterized, our current data indicate that the TRAIL/TRAIL-R system might play an important role in the biology of MSCs. Disclosure of potential conflicts of interest is found at the end of this article.


Subject(s)
Bone Marrow Cells/cytology , Cell Differentiation/physiology , Cell Movement/physiology , Mesenchymal Stem Cells/cytology , TNF-Related Apoptosis-Inducing Ligand/physiology , Bone Marrow Cells/physiology , Cell Proliferation , Cells, Cultured , Extracellular Signal-Regulated MAP Kinases/metabolism , Humans , Mesenchymal Stem Cells/physiology , Phosphorylation , Receptors, TNF-Related Apoptosis-Inducing Ligand/metabolism , Recombinant Proteins/pharmacology , Signal Transduction , TNF-Related Apoptosis-Inducing Ligand/pharmacology , Tumor Necrosis Factor Decoy Receptors/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...